1,884
Views
121
CrossRef citations to date
0
Altmetric
Review

Rethinking mercury: the role of selenium in the pathophysiology of mercury toxicity

ORCID Icon
Pages 313-326 | Received 17 Jul 2017, Accepted 31 Oct 2017, Published online: 10 Nov 2017

References

  • Clarkson TW, Magos L. The toxicology of mercury and its chemical compounds. Crit Rev Toxicol. 2006;36:609–662.
  • Sue YJ. Mercury. In: Nelson LS, Lewin NA, Howland MA, et al., editors. Goldfrank’s Toxicologic emergencies. New York: McGraw Hill Medical; 2011. p. 1299–1307.
  • Branco V, Godinho-Santos A, Goncalves J, et al. Mitochondrial thioredoxin reductase inhibition, selenium status, and Nrf-2 activation are determinant factors modulating the toxicity of mercury compounds. Free Radic Biol Med. 2014;73:95–105.
  • Carvalho CML, Chew EH, Hashemy SI, et al. Inhibition of the human thioredoxin system: a molecular mechanism of mercury toxicity. J Biol Chem. 2008;283:11913–11923.
  • Sugiura Y, Tamai Y, Tanaka H. Selenium protection against mercury toxicity: high binding affinity of methylmercury by selenium-containing ligands in comparison with sulfur-containing ligands. Bioinorg Chem. 1978;9:167–180.
  • Melnick JG, Yurkerwich K, Parkin G. On the chalcogenophilicity of mercury: evidence for a strong Hg − Se bond in [TmBut]HgSePh and its relevance to the toxicity of mercury. J Am Chem Soc. 2010;132:647–655.
  • Berry MJ, Ralston NVC. Mercury toxicity and the mitigating role of selenium. Ecohealth. 2008;5:456–459.
  • Dryssen D, Wedborg M. The surfur-mercury (II) system in natural waters. Water Air Soil Pollut. 1991;56:507–519.
  • Khan MAK, Wang F. Mercury-selenium compounds and their toxicological significance toward a molecular understanding of the mercury-selenium antagonism. Environ Toxicol Chem. 2009;28:1567–1577.
  • Farina M, Rocha JBT, Aschner M. Mechanisms of methylmercury-induced neurotoxicity: evidence from experimental studies. Life Sci. 2011;89:555–563.
  • Branco V, Godinho-Santos A, Gonçalves J, et al. Mitochondrial thioredoxin system as a primary target for mercury compounds. Toxicol Lett. 2014;229(Suppl):S57–S58.
  • Branco V, Lu J, Holmgren A, et al. Effect of mercury compounds over the redox state of thioredoxin 1 and 2 and its relation with glutathione levels and glutaredoxin activity in human neuroblastoma cells. Toxicol Lett. 2015;238(Suppl):S314.
  • Aschner M, Eberle NB, Goderie S, et al. Methylmercury uptake in rat primary astrocyte cultures: the role of the neutral amino acid transport system. Brain Res. 1990;521:221–228.
  • Kerper LE, Ballatori N, Clarkson TW. Methylmercury transport across the blood-brain barrier by an amino acid carrier. Am J Physiol. 1992;262:R761–R765.
  • Simone-Willis T, Koh AS, Clarkson TW, et al. Transport of the neurotoxicant by molecular mimicry: the methylmercury-l-cysteine complex is a substrate for human L-type large neutral amino acid transporter (LAT) 1 and LAT2. BioChem J. 2002;367:239–246.
  • Kageyama K, Onoyama Y, Kano E. Effects of methyl mercuric chloride and sulfhydryl inhibitors on phospholipid synthetic activity of lymphocytes. J Appl Toxicol. 1986;6:49–53.
  • Brown KM, Pickard K, Nicol F, et al. Effects of organic and inorganic selenium supplementation on selenoenzyme activity in blood lymphocytes, granulocytes, platelets and erythrocytes. Clin Sci. 2000;98:593–599.
  • Kung MP, Kostyniak P, Olsen J, et al. Studies of the in-vitro effect of methyl mercuric chloride on rat brain neurotransmitter enzymes. J Appl Toxicol. 1987;7:119–121.
  • Magour S. Studies on the inhibition of brain synaptosomal Na+/K + ATPase by mercuric chloride and methylmercury chloride. Arch Toxicol. 1986;9:393–396.
  • Cheek DB, Wu F. The effect of calomel on plasma epinephrine in the rat and the relationship to mechanisms in pink disease. Arch Dis Child. 1959;34:501–504.
  • Torres AD, Rai AN, Hardiek ML. Mercury intoxication and arterial hypertension: report of two patients and review of the literature. Pediatrics. 2000;105:e34.
  • Boadi WY, Urbach J, Barnea ER, et al. In vitro effect of mercury on aryl hydrocarbon hydroxylase, quinone reductase, catecholamine-O-methyltransferase and glucose-6-phosphate dehydrogenase activities in term human placenta. Pharmacol Toxicol. 1991;68:317–321.
  • Bottiglieri T. Ademetionine (S-adenosylmethionine) neuropharmacology: implications for drug therapies in psychiatric and neurological disorders. Expert Opin Investig Drugs. 1997;6:417–426.
  • Zalups RK, Bridges CC. Relationships between the renal handling of DMPS and DMSA and the renal handling of mercury. Chem Res Toxicol. 2012;25:1825–1838.
  • Becker A, Soliman KFA. The role of intracellular glutathione in inorganic mercury-induced toxicity in neuroblastoma cells. Neurochem Res. 2009;34:1677–1684.
  • Yasutake A, Hirayama K. Acute effects of methylmercury on hepatic and renal glutathione metabolisms in mice. Arch Toxicol. 1994;68:512–516.
  • Kaur P, Aschner M, Syversen T. Glutathione modulation influences methyl mercury induced neurotoxicity in primary cell cultures of neurons and astrocytes. Neurotoxicology. 2006;27:492–500.
  • Rush T, Liu X, Nowakowki AB, et al. Glutathione-mediated neuroprotection against methylmercury neurotoxicity in cortical culture is dependent on MRP1. Neurotoxicology. 2012;33:476–481.
  • Khan MAK, Wang F. Chemical demethylation of methylmercury by selenoamino acids. Chem Res Toxicol. 2010;23:1202–1206.
  • Dutczak WJ, Ballatori N. Transport of the glutathione-methylmercury complex across liver canalicular membranes on reduced glutathione carriers. J Biol Chem. 1994;269:9746–9751.
  • Smith JC, Farris FF. Methyl mercury pharmacokinetics in man: a reevaluation. Toxicol Appl Pharmacol. 1996;137:245–252.
  • Smith JC, Allen PV, Turner MD, et al. The kinetics of intravenously administered methyl mercury in man. Toxicol Appl Pharmacol. 1994;128:251–256.
  • Tanaka-Kagawa T, Naganuma A, Imura N. Tubular secretion and reabsorption of mercury compounds in mouse kidney. J Pharmacol Exp Ther. 1993;264:776–782.
  • Bridges CC, Joshee L, Zalops RK. Multidrug resistance proteins and the elimination of inorganic mercury mediated by 2,3-Dimercaptopropane-1-sulfonic acid and Meso-2,3-dimerpatosuccinic acid. J Pharmacol Exp Ther. 2008;324:383–390.
  • Li Y, Fan Y, Zhao J, et al. Elevated mercury bound to serum proteins in methylmercury poisoned rats after selenium treatment. Biometals. 2016;29:892–902.
  • Leyshon-Sorland K, Jasani B, Morgan AJ. The localization of mercury and metallothionein in the cerebellum of rats experimentally exposed to methylmercury. Histochem J. 1994;26:161–169.
  • Morcillo MA, Santamaria J. Mercury distribution and renal metallothionein induction after subchronic oral exposure in rats. Biometals. 1996;9:213–220.
  • Kehrig HA, Hauser-Davis RA, Seixas TG, et al. Mercury species, selenium, metalloproteins and glutathione in two dolphins from the southeastern Brazilian coast: mercury detoxification and physiological differences in diving capacity. Environ Pollut. 2016;213:7850792.
  • Culvin-Aralar LA, Furness RW. Mercury and selenium interaction: a review. Ecotoxicol Environ Saf. 1991;21:348–364.
  • Garcia-Sevillano MA, Rodriguez-Moro G, Garcia-Barrera T, et al. Biological interactions between mercury and selenium in distribution and detoxification process in mice under controlled exposure. Effects of selenoprotein. Chem Biol Interact. 2015;229:82–90.
  • Mengel H, Karlog O. Studies on the interaction and distribution of selenite, mercuric, methoxyethyl mercuric and methyl mercuric chloride in rats. II. Analysis of the soluble proteins and the precipitates of liver and kidney homogenates. Acta Pharmacol Toxicol (Copenh). 1980;46:25–31.
  • Carrier G, Bouchard M, Brunet RC, et al. A toxicokinetic model for predicting the tissue distribution and elimination of organic and inorganic mercury following exposure to methyl mercury in animals and humans. II. Application and validation of the model in humans. Toxicol Appl Pharmacol. 2001;171:50–60.
  • Ganther HE. Modification of methylmercury toxicity and metabolism by selenium and Vitamin E: possible mechanisms. Environ Health Perspect. 1978;25:71–76.
  • Sheikh TJ, Patel BJ, Joshi DV. Effects of mercuric chloride on oxidative stress and target organ pathology in the wistar rat. J App Pharm Sci. 2011;1:59–61.
  • Farina M, Aschner M, Rocha JBT. Oxidative stress in MeHg-induced neurotoxicity. Toxicol Appl Pharmacol. 2011;256:405–417.
  • Sugiura Y, Tamai Y, Tanaka H. Selenium protection against mercury toxicity: high binding affinity of methylmercury by selenium-containing ligands in comparison with sulfur-containing ligands. Bioorganic Chem. 1978;9:167–180.
  • Grumolato L, Ghzili H, Montero-Hadjadje M, et al. Selenoprotein T is a PACAP-regulated gene involved in intracellualr Ca+2 mobilization and neuroendocrine secretion. FASEB J. 2008;22:1756–1768.
  • Raymond LJ, Ralston NVC. Mercury: selenium interactions and health implications. Seychelles Med Den J. 2004;7:72–77.
  • Wang C, Li R, Huang Y, et al. Selenoprotein K modulate intracellular free Ca2+ by regulating expression of calcium homoeostasis endoplasmic reticulum protein. Biochem Biophys Res Commun. 2017;484:734–739.
  • Lu J, Holmgren A. The thioredoxin antioxidant system. Free Radic Biol Med. 2014;66:75–87.
  • Glaser V, Martins RP, Viera AJH, et al. Diphenyl diselenide administration enhances cortical mitochondrial number and activity by increasing hemeoxygenase type 1 content in a methylmercury-induced neurotoxicity mouse model. Mol Cell Biochem. 2014;390:1–9.
  • Mendelev N, Mehta SL, Idris H, et al. Selenite stimulates mitochondrial biogenesis signaling and enhances mitochondrial functional performance in murine hippocampal neuronal cells. PLoS One. 2012;7:e47910
  • Usaki F, Fujimura M. Decreased plasma thiol antioxidant barrier and selenoproteins as potential biomarkers for ongoing methylmercury intoxication and an individual protective capacity. Arch Toxicol. 2016;90:917–926.
  • Franco JL, Posser T, Dunkley PR, et al. Methylmercury neurotoxicity is associated with inhibition of the antioxidant enzyme glutathione peroxidase. Free Radic Biol Med. 2009;47:449–457.
  • Branco V, Canario J, Lu J, et al. Mercury and selenium interaction in vivo: effects on thioredoxin reductase and glutathione peroxidase. Free Radic Biol Med. 2012;52:781–793.
  • Burk RF, Jordan HE, Kiker KW. Some effects of selenium status on inorganic mercury metabolism in the rat. Toxicol App Pharmacol. 1977;40:71–82.
  • Ralston NCV, Ralston CR, Blackwell JL, et al. Dietary and tissue selenium in relation to methylmercury toxicity. Neurotoxicology. 2008;29:802–811.
  • Bulato C, Bosello V, Ursini F, et al. Effect of mercury on selenium utilization and selenoperoxidase activity in LNCaP cells. Free Radic Biol Med. 2007;42:118–123.
  • Carvalho CML, Lu J, Zhang X, et al. Effects of selenite and chelating agents on mammalian thioredoxin reductase inhibited by mercury: implications for treatment of mercury poisoning. FASEB J. 2011;25:370–381.
  • Ralston NVC, Blackwell JL, Raymond LJ. Importance of molar ratios in selenium dependent protection against methylmercury toxicity. Biol Trace Elem Res. 2007;119:255–268.
  • Glaser V, Moritz B, Schmitz A, et al. Protective effects of diphenyl diselenide in a mouse model of brain. Chem Biol Interact. 2013;206:18–26.
  • Glynn AW, Lind Y. Effect of long-term sodium selenite supplementation on levels and distribution of mercury in blood, brain and kidneys of methyl mercury-exposed female mice. Pharmacol Toxicol. 1995;77:41–47.
  • Meinerz DF, de Paula MT, Comparsi B, et al. Protective effect of organoselenium compounds against methylmercury-induced oxidative stress in mouse brain mitochondrial-enriched fractions. Braz J Med Biol Res. 2011;44:1156–1163.
  • Della Corte CL, Soares FAA, Aschner M, et al. Diphenyl Diselenide prevents methylmercury-induced mitochondrial dysfunction in rat liver slices. Tetrahedron 2012;68:10437–10443.
  • de Freitas AS, Funck VR, Rotta MDS, et al. Diphenyl diselenide a simple organoselenium compound decreases methylmercury-induced cerebral hepatic and renal oxidative stress and mercury deposition in adult mice. Brain Res Bull. 2009;79:77–84.
  • Aschner M, Yao CP, Allen JW, et al. Methylmercury alters glutamate transport in astrocytes. Neurochem Int. 2000;37:199–206.
  • Møller-Madsen B. Localization of mercury in CNS of the rat. II. Intraperitoneal injection of methylmercuric chloride (CH3HgCl) and mercuric chloride (HgCl2). Toxicol Appl Pharmacol. 1990;103:303–323.
  • Farina M, Frizzo MES, Soares FAA, et al. Ebselen protects against methylmercury-induced inhibition of glutamate uptake by cortical slices from adult mice. Toxicol Lett. 2003;144:351–357.
  • Porcinucula LO, Rocha JB, Tavares RG, et al. Methylmercury inhibits glutamate uptake by synaptic vesicles from rat brain. Neuroreport. 2003;14:577–580.
  • Allen JW, Mutkus LA, Aschner M. Mercuric chloride, but not methylmercury, inhibits glutamine synthetase activity in primary cultures of cortical astrocytes. Brain Res. 2001;891:148–157.
  • Avshalumov MV, Rice ME. Glutamate uptake inhibition by oxygen free radicals in rat cortical astrocytes. J Neurophysiol. 2002;87:2896–2903.
  • Volterra G, Trotti D, Tromba C, et al. Glutamate uptake inhibition by oxygen free radicals in rat cortical astrocytes. J Neurosci. 1994;14:2924–2932.
  • Tan X, Tang C, Castoldi AF, et al. Effects of inorganic and organic mercury on intracellular calcium levels in rat T-lymphocytes. J Toxicol Environ Health. 1993;38:159–170.
  • Limke TL, Otero-Montanez JKL, Atchison WD. Evidence for interaction between intracellular calcium stores during methylmercury-induced intracellular calcium dysregulation in the rat cerebellar granule neurons. J Pharmacol Exper Ther. 2003;304:949–958.
  • Fowler EA, Conner BA, Mechanisms of metal induced nephrotoxicity. In: Hook BJ. Goldman RS, editors. Toxicology of the kidney. New York: Raven Press; 1993. p. 437–457.
  • Gstraunthaler G, Pfaller W, Kotanko P. Glutathione depletion and in vitro lipid peroxidation in mercury or maleate induced acute renal failure. Biochem Pharmacol. 1983;32:2969–2972.
  • Nielsen JB, Andersen HR, Andersen O, et al. Mercuric chloride-induced kidney damage in mice: time course and effect of dose. J Toxicol Environ Health. 1991;34:469–483.
  • Komsta-Szumska E, Chmielnicka J. Binding of mercury and selenium in subcellular fractions of rat liver and kidneys following separate and joint administration. Arch Toxicol. 1977;38:217–228.
  • Uezono Y, Toyohira Y, Yanagihara N, et al. Inhibition by selenium compounds of catecholamine secretion due to inhibition of Ca2+ influx in cultured bovine adrenal chromaffin cells. J Pharmacol Sci. 2006;101:223–229.
  • Baoukhzar L, Tanguy Y, Abif H, et al. Selonoprotein T: from discovery to functional studies using conditional knockout mice. In: Hatfield DL, Schweizer U, Tsuji PA, Glasyshev VN, editors. Selenium its molecular biology and role in human health. New York: Springer; 2016. p. 275–286.
  • Weinberg F, Bickmeyer U, Wiegand H. Effects of inorganic mercury (Hg2+) on calcium channel currents and catecholamine release from bovine chromaffin cells. Arch Toxicol 1995;69:191–196.
  • Chanoine JP, Compagnone NA, Wong ACK, et al. Modulation of steroidogenesis by selenium in a novel adrenal cell line developed using targeted tumorigenisis. Biofactors. 2001;14:229–238.
  • Chanoine JP, Wong ACK, Lavoie JC. Selenium deficiency impairs corticosterone and leptin responses to adrenocorticotropin in the rat. Biofactors. 2004;20:109–118.
  • Saito Y, Takhashi K. Characterization of selenoprotein P as a selenium supply protein. Eur J Biochem. 2001;269:5746–5751.
  • Chen C, Yu H, Zhao J, et al. The roles of serum selenium and selenoproteins on mercury toxicity in environmental and occupational exposure. Environ Health Perspect. 2006;114:297–301.
  • Pallai R, Uyehara-Lock JH, Bellinger FP. Selenium and selenoprotein function in brain disorders. IUBMB Life. 2014;66:229–239.
  • Suzuki KT1, Sasakura C, Yoneda S. Binding sites for the (Hg-Se) complex on selenoprotein P. Biochim Biophys Acta. 1998;1429:102–112.
  • Yoneda S, Suzuki KT. Detoxification of mercury by selenium by binding of equimolar Hg-Se complex to a specific plasma protein. Toxicol Appl Pharmacol. 1997;143:274–280.
  • Komiya K, Kawauchi S. Properties of the mercury and selenium complex formed in rat plasma in vivo. J Pharmacobiodyn. 1981;4:545–551.
  • Parizek J, Ostadalova I. The protective effect of small amounts of selenite in sublimate intoxication. Experimentia. 1967;23:142–143.
  • Seppanen K, Laatikainen R, Salonen JT, et al. Mercury-binding capacity of organic and inorganic selenium in rat blood and liver. Biol Trace Elem Res. 1998;65:197–210.
  • Li YF, Dong Z, Chen C, et al. Organic selenium supplementation increases mercury excretion and decreases oxidative damage in long-term mercury-exposed residents from Wanshan China. Environ Sci Technol. 2012;46:11313–11318.
  • Li X, Yin D, Chen Q, et al. Dietary selenium protects against redox-mediated immune suppression induced by methylmercury exposure. Food Chem Toxicol. 2014;72:169–177.
  • Kosta L, Byrne AR, Zelenko V. Correlation between selenium and mercury in man following exposure to inorganic mercury. Nature. 1975;254:238–239.
  • Koeman JH, Peters W, Koudstaal-Hol C, et al. Mercury-selenium correlations in marine mammals. Nature. 1973;245:285–286.
  • Sakamoto M, Itai T, Nakamira M, et al. Detoxification of methylmercury by formation of mercury selenide in muscle of toothed-whale. Toxicol Lett. 2015;238S:S95.
  • Cappon CJ, Smith JC. Mercury and selenium content and chemical form in fish muscle. Arch Environ Contam Toxicol. 1981;10:305–319.
  • Ganther HE, Sunde ML. Factors in fish modifying methylmercury toxicity and metabolism. Biol Trace Elem Res. 2007;119:221–233.
  • Kosnett MJ. The role of chelation in the treatment of arsenic and mercury poisoning. J Med Toxicol. 2013;9:347–354.
  • Vahter ME, Mottet NK, Friberg LT, et al. Demethylation of methylmercury in different brain sites in Macaca fascicularis monkeys during long term subclinical methylmercury exposure. Toxicol App Pharmacol. 1995;143:273–284.
  • Hansen JC, Danscher G. Quantitative and qualitative distribution of mercury in organs from arctic sledgedogs: an atomic absorption spectrophotometric and histochemical study of tissue samples from natural long-termed high dietary organic mercury-exposed dogs from Thule, Greenland. Pharmacol Toxicol. 1995;77:189–195.
  • Rice DC. Brain and tissue levels of mercury after chronic methylmercury exposure in the monkey. J Toxicol Environ Health. 1989;27:189–198.
  • Cabanero AL, Madrid Y, Camara C. Selenium long-term administration and its effect on mercury toxicity. J Agric Food Chem. 2006;54:4461–4468.
  • Iwata H, Masukawa T, Kito H, et al. Degradation of methylmercury by selenium. Life Sci. 1982;31:859–866.
  • Glynn AW, Ilback NG, Brabencova D, et al. Influence of sodium selenite on 203Hg absorption, distribution, and elimination in male mice exposed to methyl203Hg. Biol Trace Elem Res. 1993;39:91–107.
  • Sakamoto M, Yasutake A, Kakita A, et al. Selenomethionine protects against neuronal degeneration by methylmercury in the developing rat cerebrum. Environ Sci Technol. 2013;47:2862–2868.
  • Glaser V, Nazari EM, Muller YMR, et al. Effects of inorganic selenium administration in methylmercury-induced neurotoxicity in mouse cerebral cortex. Int J Dev Neurosci. 2010;28:631–637.
  • Suda I, Hirayama K. Degradation of methyl and ethyl mercury into inorganic mercury by hydroxyl radical produced from rat liver microsomes. Arch Toxicol. 1992;66:398–402.
  • Yasutake A, Hirayama K. Evaluation of methylmercury biotransformation using rat liver slices. Arch Toxicol. 2001;75:400–406.
  • Della Corte CL, Ramos A, dos Santos CMM, et al. Selenium and mercury levels in rat liver slices co-treated with diphenyl diselenide and methylmercury. Biometals. 2016;29:543–550.
  • Della Corte CL, Wagner C, Sudati JH, et al. Effects of diphenyl diselenide on methylmercury toxicity in rats. Biomed Res Int. 2013;2013:983821
  • Hansen JC, Kristensen P, Westergaard I. The influence of selenium on mercury distribution in mice after exposure to low dose Hg vapours. J Appl Toxicol. 1981;1:141–153.
  • Newland MC, Reed MN, LeBlanc A, et al. Brain and blood mercury and selenium after chronic and developmental exposure to methylmercury. Neurotoxicology. 2006;27:710–720.
  • Joshi D, Mittal DK, Shukla S, et al. Methylmercury toxicity: amelioration by selenium and water-soluble chelators as N-acetyl Cysteine and dithiothreitol. Cell Biochem Funct. 2014;32:351–360.
  • Anderson O, Nielson JB. Effects of simultaneous low-level dietary supplementation with inorganic and organic selenium on whole-body, blood and organ levels of toxic metals in mice. Environ Health Perspect. 1994;102:321–324.
  • Li YF, Chen C, Li B, et al. Simultaneous speciation of selenium and mercury in human urine samples from long-term mercury-exposed populations with supplementation of selenium-enriched yeast by HPLC-ICP-MS. J Anal Spectrom. 2007;22:925–930.
  • Nakamura M, Hachiya N, Murata KY, Nakanshi I, et al. Methylmercury exposure and neurological outcomes in Taiji residents accustomed to consuming whale meat. Environ Int. 2014;68:25–32.
  • Mykkänen HM, Metsäniitty L. Selenium-mercury interaction during intestinal absorption of 75Se compounds in chicks. J Nutr. 1987;117:1453–1458.
  • Penglase S, Hamre K, Ellingsen S. Selenium prevents downregulation of antioxidant selenoprotein genes by methylmercury. Free Radic Biol Med. 2014;75:95–104.
  • Guerra I, Branco V, Rodrigues J, et al. Protective effects of selenium compounds in the toxicity of mercury over the thioredoxin system in neuroblastoma cells. Toxicol Lett. 2014;299S:S101–S102.
  • Grotto D, Barcelos GRM, Valentini J, et al. Low levels of methylmercury induce DNA damage in rats: protective effects of selenium. Arch Toxicol. 2009;83:249–254.
  • Cordero-Herrera I, Cuello S, Goya L, et al. Molecular mechanisms involved in the protective effect of selenocystine against methylmercury-induced cell death in human HepG2 cells. Food Chem Toxicol. 2013;59:554–563.
  • Heath JC, Banna KM, Reed MN, et al. Dietary selenium protects against selected signs of aging and methylmercury exposure. Neurotoxicology. 2010;31:169–179.
  • Lemire M, Fillion M, Frenette B, et al. Selenium from dietary sources and motor functions in the Brazilian Amazon. Neurotoxicology. 2011;32:944–953.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.