Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Metallomics ; 10(6): 818-827, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29770420

ABSTRACT

The essential micronutrient selenium (Se) is required for various systemic functions, but its beneficial range is narrow and overexposure may result in adverse health effects. Additionally, the chemical form of the ingested selenium contributes crucially to its health effects. While small Se species play a major role in Se metabolism, their toxicological effects, bioavailability and metabolic transformations following elevated uptake are poorly understood. Utilizing the tractable invertebrate Caenorhabditis elegans allowed for an alternative approach to study species-specific characteristics of organic and inorganic Se forms in vivo, revealing remarkable species-dependent differences in the toxicity and bioavailability of selenite, selenomethionine (SeMet) and Se-methylselenocysteine (MeSeCys). An inverse relationship was found between toxicity and bioavailability of the Se species, with the organic species displaying a higher bioavailability than the inorganic form, yet being less toxic. Quantitative Se speciation analysis with HPLC/mass spectrometry revealed a partial metabolism of SeMet and MeSeCys. In SeMet exposed worms, identified metabolites were Se-adenosylselenomethionine (AdoSeMet) and Se-adenosylselenohomocysteine (AdoSeHcy), while worms exposed to MeSeCys produced Se-methylselenoglutathione (MeSeGSH) and γ-glutamyl-MeSeCys (γ-Glu-MeSeCys). Moreover, the possible role of the sole selenoprotein in the nematode, thioredoxin reductase-1 (TrxR-1), was studied comparing wildtype and trxr-1 deletion mutants. Although a lower basal Se level was detected in trxr-1 mutants, Se toxicity and bioavailability following acute exposure was indistinguishable from wildtype worms. Altogether, the current study demonstrates the suitability of C. elegans as a model for Se species dependent toxicity and metabolism, while further research is needed to elucidate TrxR-1 function in the nematode.


Subject(s)
Caenorhabditis elegans/metabolism , Selenious Acid/metabolism , Selenocysteine/analogs & derivatives , Selenomethionine/analogs & derivatives , Animals , Biological Availability , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/growth & development , Selenious Acid/toxicity , Selenocysteine/metabolism , Selenocysteine/toxicity , Selenomethionine/metabolism , Selenomethionine/toxicity , Thioredoxin Reductase 1/metabolism
2.
Biomol Concepts ; 8(2): 93-104, 2017 May 24.
Article in English | MEDLINE | ID: mdl-28574376

ABSTRACT

Selenium is an essential trace element due to its incorporation into selenoproteins with important biological functions. However, at high doses it is toxic. Selenium toxicity is generally attributed to the induction of oxidative stress. However, it has become apparent that the mode of action of seleno-compounds varies, depending on its chemical form and speciation. Recent studies in various eukaryotic systems, in particular the model organism Saccharomyces cerevisiae, provide new insights on the cytotoxic mechanisms of selenomethionine and selenocysteine. This review first summarizes current knowledge on reactive oxygen species (ROS)-induced genotoxicity of inorganic selenium species. Then, we discuss recent advances on our understanding of the molecular mechanisms of selenocysteine and selenomethionine cytotoxicity. We present evidences indicating that both oxidative stress and ROS-independent mechanisms contribute to selenoamino acids cytotoxicity. These latter mechanisms include disruption of protein homeostasis by selenocysteine misincorporation in proteins and/or reaction of selenols with protein thiols.


Subject(s)
Organoselenium Compounds/toxicity , Saccharomyces cerevisiae/drug effects , Eukaryotic Cells/drug effects , Metabolic Networks and Pathways , Models, Biological , Selenocysteine/toxicity , Selenomethionine/toxicity
3.
Aquat Toxicol ; 189: 1-8, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28554051

ABSTRACT

Selenium (Se) is an intriguing element because it is metabolically required by a variety of organisms, but it may induce toxicity at high doses. Algae primarily absorb selenium in the form of selenate or selenite using mechanisms similar to those reported in plants. However, while Se is needed by several species of microalgae, the essentiality of this element for plants has not been established yet. The study of Se uptake and accumulation strategies in micro- and macro-algae is of pivotal importance, as they represent potential vectors for Se movement in aquatic environments and Se at high levels may affect their growth causing a reduction in primary production. Some microalgae exhibit the capacity of efficiently converting Se to less harmful volatile compounds as a strategy to cope with Se toxicity. Therefore, they play a crucial role in Se-cycling through the ecosystem. On the other side, micro- or macro-algae enriched in Se may be used in Se biofortification programs aimed to improve Se content in human diet via supplementation of valuable food. Indeed, some organic forms of selenium (selenomethionine and methylselenocysteine) are known to act as anticarcinogenic compounds and exert a broad spectrum of beneficial effects in humans and other mammals. Here, we want to give an overview of the developments in the current understanding of Se uptake, accumulation and metabolism in algae, discussing potential ecotoxicological implications and nutritional aspects.


Subject(s)
Chlorophyta/metabolism , Seaweed/metabolism , Selenium Compounds/metabolism , Selenium/metabolism , Water Pollutants, Chemical/metabolism , Animals , Chlorophyta/drug effects , Ecosystem , Environmental Monitoring , Humans , Seaweed/drug effects , Selenic Acid/metabolism , Selenic Acid/toxicity , Selenium/toxicity , Selenium Compounds/toxicity , Selenocysteine/analogs & derivatives , Selenocysteine/metabolism , Selenocysteine/toxicity , Selenomethionine/metabolism , Selenomethionine/toxicity , Water Pollutants, Chemical/toxicity
4.
Regul Toxicol Pharmacol ; 70(3): 720-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25444999

ABSTRACT

The significant toxicity of selenium emphasizes the need to assess the health risk of various selenocompounds as nutritional supplements. Se-methylselenocysteine (SeMC) was recently reported to be more bioactive but the toxicological effects have not been sufficiently characterized. This study aimed to evaluate the safety of SeMC and provide the Acceptable Daily Intake (ADI) for its use in human diet. Our results demonstrated that SeMC, with the Median Lethal Dose (LD50) of 12.6 and 9.26mg/kg BW in female and male mice, was of high potent of health hazard under acute oral exposure, but a battery of tests including Ames test, micronucleus assay and mouse sperm malformation assay suggested that SeMC was not genotoxic. The repeated dose study indicated little systemic toxicity of SeMC at supernutritional levels (0.5, 0.7, 0.9mg/kg BW/day) after 90-day oral exposure. Importantly, the 95% lower confidence value of Benchmark Dose (BMDL) was estimated as 0.34mg/kg BW/day according to the elevated relative liver weight. The ADI for human was established at 3.4µg/kg BW/day. The results suggested greater safety of SeMC as a nutritional selenium supplement, but health risk needs to be further evaluated when SeMC is applied beyond this level to achieve cancer chemoprevention.


Subject(s)
Dietary Supplements/toxicity , Selenocysteine/analogs & derivatives , Animals , Dose-Response Relationship, Drug , Female , Humans , Lethal Dose 50 , Male , Mice, Inbred BALB C , Models, Biological , Mutagenicity Tests , No-Observed-Adverse-Effect Level , Rats, Sprague-Dawley , Selenium , Selenocysteine/toxicity , Spermatozoa/drug effects , Spermatozoa/growth & development , Toxicity Tests, Acute , Toxicity Tests, Subchronic
5.
Environ Toxicol Chem ; 32(11): 2584-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24115124

ABSTRACT

Apis mellifera L. (Hymenoptera: Apidae) is an important agricultural pollinator in the United States and throughout the world. In areas of selenium (Se) contamination, honeybees may be at risk because of the biotransfer of Se from plant products such as nectar and pollen. Several forms of Se can occur in accumulating plants. In the present study, the toxicity of 4 compounds (selenate, selenite, methylselenocysteine, and selenocystine) to honeybee adult foragers and larvae was assessed using dose-response bioassays. Inorganic forms were more toxic than organic forms for both larvae (lethal concentration [LC50] selenate = 0.72 mg L(-1) , LC50 selenite = 1.0 mg L(-1) , LC50 methylselenocysteine = 4.7 mg L(-1) , LC50 selenocystine = 4.4 mg L(-1) ) and foragers (LC50 selenate = 58 mg L(-1) , LC50 selenite = 58 mg L(-1) , LC50 methylselenocysteine = 161 mg L(-1) , LC50 selenocystine = 148 mg L(-1) ). Inorganic forms of Se caused rapid mortality, and organic forms had sublethal effects on development. Larvae accumulated substantial amounts of Se only at the highest doses, whereas foragers accumulated large quantities at all doses. The present study documented very low larval LC50 values for Se; even modest transfer to brood will likely cause increased development times and mortality. The toxicities of the various forms of Se to honeybee larvae and foragers are discussed in comparison with other insect herbivores and detritivores.


Subject(s)
Bees/drug effects , Environmental Pollutants/toxicity , Organoselenium Compounds/toxicity , Selenium Compounds/toxicity , Animals , Bees/growth & development , Cystine/analogs & derivatives , Cystine/toxicity , Larva/drug effects , Larva/growth & development , Pollination , Selenic Acid/toxicity , Selenious Acid/toxicity , Selenocysteine/analogs & derivatives , Selenocysteine/toxicity
6.
Life Sci ; 75(4): 447-59, 2004 Jun 11.
Article in English | MEDLINE | ID: mdl-15147831

ABSTRACT

We have previously reported the synthesis and characterization of two new classes of selenazolidine-4(R)-carboxylic acids (2-oxo and 2-methyl-SCAs) (OSCA and MSCA, respectively), as well as the "parent" compound, selenazolidine-4(R)-carboxylic acid (SCA, selenaproline). These compounds were designed as prodrugs of L-selenocysteine with potential application in cancer chemoprevention or other clinical uses. We will be exploring the chemopreventive activity of the new compounds in the well-established A/J mouse model of tobacco-induced lung carcinogenesis. The objectives of the present study were to investigate several fundamental biochemical endpoints after selenazolidine administration compared with other selenium-containing agents. Groups of mice were fed either AIN-76A diet alone or the diet supplemented with the following selenium compounds (ppm Se): sodium selenite (5), L-selenomethionine (3.75), L-selenocystine (15), Se-methyl-L-selenocysteine (3), MSCA (5, 10, or 15), OSCA (5, 10, or 15), or SCA (5, 10, or 15). After 28 days of supplementation, toxicity of the selenazolidines was not evident, as measured by outward appearance and behavior, body and organ weight changes, and histological evaluation of liver and lung tissue. Select treatment groups showed significant increases in selenium levels in blood and tissues. Increased activity of selenium-dependent glutathione peroxidase (GPx) in blood and liver illustrated that the selenazolidines provided a source of biologically-available selenium.


Subject(s)
Glutathione Peroxidase/biosynthesis , Organoselenium Compounds/toxicity , Prodrugs/toxicity , Proline/analogs & derivatives , Proline/toxicity , Selenium/pharmacokinetics , Selenocysteine/toxicity , Animals , Body Weight/drug effects , Enzyme Induction , Female , Glutathione Peroxidase/blood , Liver/drug effects , Liver/enzymology , Liver/metabolism , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred Strains , Organ Size/drug effects , Organoselenium Compounds/pharmacokinetics , Organoselenium Compounds/pharmacology , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Proline/pharmacokinetics , Proline/pharmacology , Selenium/blood , Selenocysteine/pharmacokinetics , Selenocysteine/pharmacology , Tissue Distribution , Toxicity Tests
7.
J Med Chem ; 46(15): 3308-13, 2003 Jul 17.
Article in English | MEDLINE | ID: mdl-12852761

ABSTRACT

Novel selenazolidine prodrugs of selenocysteine are being developed as potential selenium delivery agents for cancer chemoprevention and other clinical uses. The 2-unsubstituted compound, selenazolidine-4(R)-carboxylic acid (L-SCA), and the 2-oxo- and 2-methyl analogues possessing D-stereochemistry (D-OSCA and D-MSCA, respectively) were synthesized and chemically characterized. L/D pairs, along with other organoselenium compounds and common inorganic forms, were studied in cultured V79 cells to understand their inherent toxicity and their ability to induce selenium-dependent glutathione peroxidase (GPx) activity, which indicates the provision of biologically available selenium. All of the selenazolidines were much less toxic to the cells than was sodium selenite (IC(50) approximately 17 microM) or the parent selenolamines, L- or D-selenocystine (IC(50) approximately 34 or 39 microM, respectively); OSCA was less toxic than MSCA. The stereoisomers of OSCA produced very different IC(50) values (L-OSCA, approximately 451 microM; D-OSCA, >3000 microM), while the IC(50) values derived for the stereoisomers of MSCA were of the same order of magnitude (L-MSCA, approximately 79 microM; D-MSCA, approximately 160 microM). Compounds possessing L-stereochemistry were at least as active with respect to GPx induction as was sodium selenite (2.2-fold increase at 15 microM). L-Selenocystine produced a 4.2-fold increase in GPx activity at 30 microM, while L-SCA produced a 5.9-fold increase, followed by L-OSCA (4.6-fold) and L-MSCA (2.1-fold), all at 100 microM. Compounds possessing D-stereochemistry showed minimal ability to induce GPx activity (D-selenocystine, 1.0-fold increase; D-OSCA, 1.4-fold increase; D-MSCA, 1.3-fold increase).


Subject(s)
Glutathione Peroxidase/biosynthesis , Organoselenium Compounds/toxicity , Prodrugs/toxicity , Proline/toxicity , Selenocysteine/analogs & derivatives , Selenocysteine/toxicity , Animals , Cricetinae , Cricetulus , Enzyme Induction , Organoselenium Compounds/metabolism , Prodrugs/chemical synthesis , Prodrugs/metabolism , Proline/analogs & derivatives , Proline/metabolism , Selenocysteine/metabolism , Stereoisomerism , Structure-Activity Relationship
8.
Chem Res Toxicol ; 15(12): 1610-8, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12482244

ABSTRACT

Tellurium compounds are effective antioxidants and chemoprotectors, even more active than their selenium and sulfur analogues. In addition to these properties, some selenium compounds, such as selenocysteine Se-conjugates, possess significant chemopreventive and antitumor activities, and selenol metabolites are considered as active species. In the present study, we have synthesized Te-phenyl-L-tellurocysteine and evaluated its bioactivation and cytotoxicity. The activities of this compound were compared with those of the corresponding selenium and sulfur analogues. Te-Phenyl-L-tellurocysteine is bioactivated into its corresponding tellurol, as detected by GC-MS, by cysteine conjugate beta-lyase and amino acid oxidase, analogously to what has been shown previously for Se-phenyl-L-selenocysteine. The rate of beta-elimination may reflect the bond strength of the corresponding C-S, C-Se, and C-Te bond. Bioactivation of Te-phenyl-L-tellurocysteine and its selenium and sulfur analogues by oxidative enzymes was evaluated by measuring NADPH-dependent activation of hepatic mGST and inhibition of EROD. Te-Phenyl-L-tellurocysteine and Se-phenyl-L-selenocysteine displayed strong and time-dependent mGST activation, while S-phenyl-L-cysteine resulted in no significant activation. Te-Phenyl-L-tellurocysteine was also a strong inhibitor of EROD activity. In addition to EROD inhibition, Te-phenyl-L-tellurocysteine was the strongest inhibitor of several human cytochrome P450 isoenzymes followed by Se-phenyl-L-selenocysteine, while S-phenyl-L-cysteine was the weakest inhibitor. Interestingly, Te-phenyl-L-tellurocysteine selectively inhibited cytochrome P450 1A1 directly, which is, for example, responsible for the activation of several procarcinogens. Preliminary cytotoxicity studies with Te-phenyl-L-tellurocysteine in freshly isolated rat hepatocytes showed a time-dependent depletion of GSH and LDH leakage comparable with the relatively nontoxic drug paracetamol, while the selenium and sulfur analogues were nontoxic toward rat hepatocytes. In conclusion, because the chemopreventive and antitumor activities of selenium compounds are thought to be mediated via their selenol metabolites and tellurium compounds might be even more active than selenium compounds, tellurocysteine Te-conjugates might be an interesting novel class of prodrugs for the formation of biologically active tellurols.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/pharmacokinetics , Cysteine/toxicity , Organometallic Compounds/toxicity , Selenocysteine/analogs & derivatives , Selenocysteine/pharmacokinetics , Selenocysteine/toxicity , Tellurium/toxicity , Animals , Biotransformation , Cytochrome P-450 CYP1A1/antagonists & inhibitors , D-Amino-Acid Oxidase/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glutathione Transferase/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kidney/enzymology , Kinetics , Lyases/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Organometallic Compounds/pharmacokinetics , Rats , Rats, Wistar , Tellurium/pharmacokinetics
9.
J Pharmacol Exp Ther ; 301(3): 884-92, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12023515

ABSTRACT

Cisplatin [cis-diamminedichloroplatinum(II)] is a widely used antitumor drug with dose-limiting nephrotoxic side effects due to selective toxicity to the proximal tubule. In the present study, the chemoprotective potential of three selenocysteine Se-conjugates, Se-methyl-L-selenocysteine, Se-(2-methoxyphenyl)-L-selenocysteine, and Se-(2-chlorobenzyl)-L-selenocysteine, belonging to three structural classes, against the nephrotoxic effects of cisplatin was investigated. Selenocysteine Se-conjugates have previously been proposed as kidney-selective prodrugs of pharmacologically active selenols because of their active uptake and bioactivation by cysteine conjugate beta-lyases in the kidney. To elucidate whether chemoprotection is beta-lyase-dependent wild-type LLC-PK(1) cells, possessing a very low beta-lyase activity, and LLC-PK(1) cells stably transfected with full-length cDNA coding for rat kidney cysteine conjugate beta-lyase/glutamine transaminase K (R1J) were used. The results indicate that all three selenocysteine Se-conjugates were able to attenuate the cisplatin-induced loss of viability in R1J cells but not in the parental LLC-PK(1) cells, as determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and neutral red uptake. In addition, cisplatin-induced reactive oxygen species (ROS) production was determined using 2',7'-dichlorodihydrofluorescein diacetate. The selenocysteine Se-conjugates were able to decrease ROS levels after cisplatin exposure in both cell types. However, this ROS-protective effect was more profound in R1J cells. Se-Methyl-L-selenocysteine provided the strongest protection. The protective activity against cisplatin-induced cytotoxicity and ROS generation was blocked by aminooxyacetic acid, a selective inhibitor of pyridoxal 5'-phosphate-dependent cysteine conjugate beta-lyases, further supporting the role of beta-lyase in the observed chemoprotection. The precise molecular mechanism by which selenols, generated by beta-lyase, provide protection against cisplatin-induced cytotoxicity, however, remains to be established.


Subject(s)
Cisplatin/toxicity , Kidney Tubules, Proximal/drug effects , Lyases/metabolism , Selenocysteine/metabolism , Animals , Antineoplastic Agents/toxicity , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/enzymology , LLC-PK1 Cells , Lyases/pharmacology , Lyases/toxicity , Rats , Reactive Oxygen Species/metabolism , Selenium/metabolism , Selenium/pharmacology , Selenium/toxicity , Selenocysteine/pharmacology , Selenocysteine/toxicity , Swine
10.
Toxicol Appl Pharmacol ; 141(1): 278-87, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8917701

ABSTRACT

Recently, Se-substituted selenocysteine conjugates were proposed as potential prodrugs to target biologically active selenol compounds to tissues containing high activities of cysteine conjugate beta-lyases, such as the kidneys. However, several selenium compounds are known to be relatively toxic compounds. In the present study, the cytotoxicity of 14 selenocysteine Se-conjugates was determined in freshly isolated rat renal proximal tubular cells (RPTC). The results of this study show that four selenocysteine Se-conjugates with alkyl substituents (methyl, ethyl, n-propyl, and n-butyl) did not cause significant cytotoxicity to RPTC up to concentrations of 500 microM after 90 min of incubation. Also, no effect was observed on mitochondrial functioning as indicated by the unaffected mitochondrial membrane potential (delta psi). Se-(i-Propyl)-selenocysteine, however, appeared to be a cytotoxic compound, causing time- and dose-dependent cytotoxicity, and caused a decrease of delta psi in remaining viable cells. Aminooxyacetic acid (AOAA) provided significant protection against cell death of Se-(i-propyl)-selenocysteine, pointing to involvement of cysteine conjugate beta-lyase. AOAA, however, did not prevent the decrease of delta psi. Differentially substituted Se-(phenyl)-L-selenocysteine and Se-(benzyl)-L-selenocysteine conjugates appeared to be cytotoxic to RPTC at a concentration of 200 microM, as indicated by increased cell death and a decreased delta psi in remaining viable cells. Within the Se-benzyl-series, Se-(4-methoxybenzyl)-L-selenocysteine was the most toxic conjugate, whereas Se-(4-chlorophenyl)-L-selenocysteine was the most toxic conjugate of the Se-phenyl compounds. The selenocysteine Se-conjugates with nonsubstituted phenyl and benzyl substituents were nontoxic at 200 microM, but caused significant cell death at a concentration of 500 microM. Preincubation with AOAA, an inhibitor of cysteine conjugate beta-lyase, provided only partial protection against the cytotoxicity of Se-(phenyl)-L-selenocysteine (500 microM) and Se-(4-methoxybenzyl)-L-selenocysteine (200 microM). AOAA did not protect against cytotoxicity of the other conjugates, suggesting direct effects of these compounds or involvement of alternative routes of bioactivation. This study demonstrates that cytotoxicity of selenocysteine Se-conjugates is strongly dependent on the nature of the Se-bound substituent. The nontoxic Se-(alkyl)-Se-conjugates may be promising candidates for further evaluation for chemopreventive activities.


Subject(s)
Kidney Tubules, Proximal/drug effects , Selenocysteine/analogs & derivatives , Selenocysteine/toxicity , Aminooxyacetic Acid/pharmacology , Animals , Cytosol/drug effects , Cytosol/metabolism , Enzyme Inhibitors/pharmacology , Kidney Tubules, Proximal/metabolism , Male , Membrane Potentials/drug effects , Mitochondria/drug effects , Rats , Rats, Wistar
11.
Arch Toxicol ; 71(1-2): 31-8, 1996.
Article in English | MEDLINE | ID: mdl-9010583

ABSTRACT

Mechanisms of selenium methylation and toxicity were investigated in the liver of ICR male mice treated with selenocystine. To elucidate the selenium methylation mechanism, animals received a single oral administration of selenocystine (Se-Cys; 5, 10, 20, 30, 40, or 50 mg/kg). In the liver, both accumulation of total selenium and production of trimethylselenonium (TMSe) as the end-product of methylation were increased by the dose of Se-Cys. A negative correlation was found between production of TMSe and level of S-adenosylmethionine (SAM) as methyl donor. The relationship between Se-Cys toxicity and selenium methylation was determined by giving mice repeated oral administration of Se-Cys (10 or 20 mg/kg) for 10 days. The animals exposed only to the high dose showed a significant rise of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activities in plasma. Urinary total selenium increased with Se-Cys dose. TMSe content in urine represented 85% of total selenium at the low dose and 25% at the high dose. The potential of Se-methylation and activity of methionine adenosyltransferase, the enzyme responsible for SAM synthesis, and the level of SAM in the liver were determined. The high dose resulted in inactivation of Se-methylation and decrease in SAM level due to the inhibition of methionine adenosyltransferase activity. To learn whether hepatic toxicity is induced by depressing selenium methylation ability, mice were injected intraperitoneally with periodate-oxidized adenosine (100 mumol/kg), a known potent inhibitor of the SAM-dependent methyltransferase, at 30 min before oral treatment of Se-Cys (10, 20, of 50 mg/kg). Liver toxicity induced by selenocystine was enhanced by inhibition of selenium methylation. These results suggest that TMSe was produced by SAM-dependent methyltransferases, which are identical with those involved in the methylation of inorganic selenium compounds such as selenite, in the liver of mice orally administered Se-Cys. Depression of selenium methylation ability resulting from inactivation of methionine adenosyltransferase and Se-methylation via enzymic reaction was also found in mice following repeated oral administration of a toxic dose of Se-Cys. The excess selenides accumulating during the depression of selenium methylation ability may be involved in the liver toxicity caused by Se-Cys.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver/drug effects , Selenium/metabolism , Selenocysteine/toxicity , Adenosine/analogs & derivatives , Animals , Liver/metabolism , Liver Function Tests , Male , Methionine Adenosyltransferase/metabolism , Methylation , Methyltransferases/metabolism , Mice , Mice, Inbred ICR , Selenium Compounds/metabolism , Selenocysteine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...