Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 299(8): 105009, 2023 08.
Article in English | MEDLINE | ID: mdl-37406814

ABSTRACT

Selenoprotein P (SeP, encoded by the SELENOP gene) is a plasma protein that contains selenium in the form of selenocysteine residues (Sec, a cysteine analog containing selenium instead of sulfur). SeP functions for the transport of selenium to specific tissues in a receptor-dependent manner. Apolipoprotein E receptor 2 (ApoER2) has been identified as a SeP receptor. However, diverse variants of ApoER2 have been reported, and the details of its tissue specificity and the molecular mechanism of its efficiency remain unclear. In the present study, we found that human T lymphoma Jurkat cells have a high ability to utilize selenium via SeP, while this ability was low in human rhabdomyosarcoma cells. We identified an ApoER2 variant with a high affinity for SeP in Jurkat cells. This variant had a dissociation constant value of 0.67 nM and a highly glycosylated O-linked sugar domain. Moreover, the acidification of intracellular vesicles was necessary for selenium transport via SeP in both cell types. In rhabdomyosarcoma cells, SeP underwent proteolytic degradation in lysosomes and transported selenium in a Sec lyase-dependent manner. However, in Jurkat cells, SeP transported selenium in Sec lyase-independent manner. These findings indicate a preferential selenium transport pathway involving SeP and high-affinity ApoER2 in a Sec lyase-independent manner. Herein, we provide a novel dynamic transport pathway for selenium via SeP.


Subject(s)
Lyases , Selenium , Humans , Lyases/metabolism , Selenium/metabolism , Selenocysteine/genetics , Selenocysteine/metabolism , Selenoprotein P/genetics , Selenoprotein P/metabolism , Selenoproteins , Jurkat Cells
2.
J Plant Physiol ; 271: 153665, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35279561

ABSTRACT

Selenium (Se) is a micronutrient essential for human and animal health. However, Se is toxic at high levels because the nonspecific substitution of cysteine by selenocysteine could lead to protein malfunction. In an attempt to prevent nonspecific selenocysteine incorporation into proteins, we simultaneously overexpressed the gene encoding selenocysteine lyase from Homo sapiens (HsSL), which specifically catalyzes the decomposition of selenocysteine into elemental Se0 and alanine, and the gene encoding selenocysteine methyltransferase from Astragalus bisulcatus (AbSMT), which methylates selenocysteine into methylselenocysteine in rice. The transgenic plants showed normal growth under standard conditions. Se treatment resulted in higher levels of alanine and methylselenocysteine in transgenic plants than in wild-type plants, which indicated that this approach might have successfully redirected Se flow in the plant. Overexpression of HsSL and AbSMT in rice also endows transgenic plants with hyposensitivity to Se stress at the seed germination stage. The transgenic plants showed enhanced selenate and selenite tolerance, which was simultaneously supported by fresh weight values. Moreover, our phytoremediation assay revealed that the transgenic plants exhibited greatly improved Se elimination capabilities and accumulated about 38.5% and 128.6% more Se than wild-type plants when treated with selenate and selenite, respectively. This study offers hope that genetically modified plants could play a role in the restoration of Se-contaminated environment.


Subject(s)
Oryza , Selenium , Animals , Biodegradation, Environmental , Oryza/genetics , Oryza/metabolism , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Selenic Acid/metabolism , Selenium/metabolism
3.
Front Nutr ; 8: 744825, 2021.
Article in English | MEDLINE | ID: mdl-34869521

ABSTRACT

Observational studies indicate that selenium may contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Transcriptomic exploration of the aetiology and progression of NAFLD may offer insight into the role selenium plays in this disease. This study compared gene expression levels of known selenoprotein pathways between individuals with a healthy liver to those with NAFLD. Publicly available gene expression databases were searched for studies that measured global gene expression in liver samples from patients with steatosis and non-alcoholic steatohepatitis (NASH) and healthy controls (with [HOC] or without [HC] obesity). A subset of five selenoprotein-related pathways (164 genes) were assessed in the four datasets included in this analysis. The gene TXNRD3 was less expressed in both disease groups when compared with HOC. SCLY and SELENOO were less expressed in NASH when compared with HC. SELENOM, DIO1, GPX2, and GPX3 were highly expressed in NASH when compared to HOC. Disease groups had lower expression of iron-associated transporters and higher expression of ferritin-encoding sub-units, consistent with dysregulation of iron metabolism often observed in NAFLD. Our bioinformatics analysis suggests that the NAFLD liver may have lower selenium levels than a disease-free liver, which may be associated with a disrupted iron metabolism. Our findings indicate that gene expression variation may be associated with the progressive risk of NAFLD.

4.
Front Nutr ; 8: 682700, 2021.
Article in English | MEDLINE | ID: mdl-34041261

ABSTRACT

The essential micronutrient selenium (Se) provides antioxidant defense and supports numerous biological functions. Obtained through dietary intake, Se is incorporated into selenoproteins via the amino acid, selenocysteine (Sec). Mice with genetic deletion of the Se carrier, selenoprotein P (SELENOP), and the Se recycling enzyme selenocysteine lyase (SCLY), suffer from sexually dimorphic neurological deficits and require Se supplementation for viability. These impairments are more pronounced in males and are exacerbated by dietary Se restriction. We report here that, by 10 weeks of age, female Selenop/Scly double knockout (DKO) mice supplemented with 1 mg/ml sodium selenite in drinking water develop signs of hyper-adiposity not seen in male DKO mice. Unexpectedly, this metabolic phenotype can be reversed by removing Se from the drinking water at post-natal day 22, just prior to puberty. Restricting access to Se at this age prevents excess body weight gain and restriction from either post-natal day 22 or 37 reduces gonadal fat deposits. These results provide new insight into the sex-dependent relationship between Se and metabolic homeostasis.

5.
Mol Cell Endocrinol ; 533: 111335, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34052303

ABSTRACT

People with obesity are often dyslipidemic and prescribed statins to prevent cardiovascular events. A common side effect of statin use is myopathy. This could potentially be caused by the reduction of selenoproteins that curb oxidative stress, in turn, affecting creatine metabolism. We determined if statins regulate hepatic and muscular selenoprotein expression, oxidative stress and creatine metabolism. Mice lacking selenocysteine lyase (Scly KO), a selenium-provider enzyme for selenoprotein synthesis, were fed a high-fat, Se-supplemented diet and treated with simvastatin. Statin improved creatine metabolism in females and oxidative responses in both sexes. Male Scly KO mice were heavier than females after statin treatment. Hepatic selenoproteins were unaffected by statin and genotype in females. Statin upregulated muscular Gpx1 in females but not males, while Scly loss downregulated muscular Gpx1 in males and Selenon in females. Osgin1 was reduced in statin-treated Scly KO males after AmpliSeq analysis. These results refine our understanding of the sex-dependent role of selenium in statin responses.


Subject(s)
Liver/metabolism , Lyases/genetics , Muscle, Skeletal/metabolism , Obesity/drug therapy , Selenoproteins/metabolism , Simvastatin/administration & dosage , Animals , Creatinine/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Glutathione Peroxidase/metabolism , Liver/drug effects , Male , Mice , Mice, Knockout , Mice, Obese , Muscle, Skeletal/drug effects , Obesity/chemically induced , Obesity/metabolism , Oxidative Stress/drug effects , Selenium , Sex Characteristics , Simvastatin/pharmacology , Glutathione Peroxidase GPX1
6.
Biochimie ; 182: 166-176, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33444662

ABSTRACT

Selenium is a vital micronutrient in many organisms. While traces are required for microbial utilization, excess amounts are toxic; thus, selenium can be regarded as a biological double-edged sword. Selenium is chemically similar to the essential element sulfur, but curiously, evolution has selected the former over the latter for a subset of oxidoreductases. Enzymes involved in sulfur metabolism are less discriminate in terms of preventing selenium incorporation; however, its specific incorporation into selenoproteins reveals a highly discriminate process that is not completely understood. We have identified SclA, a NifS-like protein in the nosocomial pathogen, Enterococcus faecalis, and characterized its enzymatic activity and specificity for l-selenocysteine over l-cysteine. It is known that Asp-146 is required for selenocysteine specificity in the human selenocysteine lyase. Thus, using computational biology, we compared the bacterial and mammalian enzymes and identified His-100, an Asp-146 ortholog in SclA, and generated site-directed mutants in order to study the residue's potential role in the l-selenocysteine discrimination mechanism. The proteins were overexpressed, purified, and characterized for their biochemical properties. All mutants exhibited varying Michaelis-Menten behavior towards l-selenocysteine, but His-100 was not found to be essential for this activity. Additionally, l-cysteine acted as a competitive inhibitor of all enzymes with higher affinity than l-selenocysteine. Finally, we discovered that SclA exhibited low activity with l-cysteine as a poor substrate regardless of mutations. We conclude that His-100 is not required for l-selenocysteine specificity, underscoring the inherent differences in discriminatory mechanisms between bacterial NifS-like proteins and mammalian selenocysteine lyases.


Subject(s)
Bacterial Proteins/chemistry , Enterococcus faecalis/enzymology , Lyases/chemistry , Selenium/chemistry , Sulfur/chemistry , Bacterial Proteins/metabolism , Lyases/metabolism , Selenium/metabolism , Substrate Specificity , Sulfur/metabolism
7.
J Trace Elem Med Biol ; 62: 126596, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32683228

ABSTRACT

BACKGROUND: The amino acid selenocysteine (Sec) is an integral part of selenoproteins, a class of proteins mostly involved in strong redox reactions. The enzyme Sec lyase (SCLY) decomposes Sec into selenide allowing for the recycling of the selenium (Se) atom via the selenoprotein synthesis machinery. We previously demonstrated that disruption of the Scly gene (Scly KO) in mice leads to the development of obesity and metabolic syndrome, with effects on glucose homeostasis, worsened by Se deficiency or a high-fat diet, and exacerbated in male mice. Our objective was to determine whether Se supplementation could ameliorate obesity and restore glucose homeostasis in the Scly KO mice. METHODS: Three-weeks old male and female Scly KO mice were fed in separate experiments a diet containing 45 % kcal fat and either sodium selenite or a mixture of sodium selenite and selenomethionine (selenite/SeMet) at moderate (0.25 ppm) or high (0.5-1 ppm) levels for 9 weeks, and assessed for metabolic parameters, oxidative stress and expression of selenoproteins. RESULTS: Se supplementation was unable to prevent obesity and elevated epididymal white adipose tissue weights in male Scly KO mice. Serum glutathione peroxidase activity in Scly KO mice was unchanged regardless of sex or dietary Se intake; however, supplementation with a mixture of selenite/SeMet improved oxidative stress biomarkers in the male Scly KO mice. CONCLUSION: These results unveil sex- and selenocompound-specific regulation of energy metabolism after the loss of Scly, pointing to a role of this enzyme in the control of whole-body energy metabolism regardless of Se levels.


Subject(s)
Lyases/metabolism , Obesity/metabolism , Selenium/therapeutic use , Animals , Biomarkers/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism/drug effects , Glutathione Peroxidase/metabolism , Lyases/genetics , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Mice , Mice, Knockout , Obesity/chemically induced , Oxidative Stress/drug effects , Selenious Acid/therapeutic use
8.
Int J Biol Macromol ; 156: 18-26, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32275991

ABSTRACT

The selenocysteine (Sec) incorporation is a co-translational event taking place at an in-frame UGA-codon and dependent on an organized molecular machinery. Selenium delivery requires mainly two enzymes, the selenocysteine lyase (CsdB) is essential for Sec recycling and conversion to selenide, further used by the selenophosphate synthetase (SelD), responsible for the conversion of selenide in selenophosphate. Therefore, understanding the catalytic mechanism involved in selenium compounds delivery, such as the interaction between SelD and CsdB (EcCsdB.EcSelD), is fundamental for the further comprehension of the selenocysteine synthesis pathway and its control. In Escherichia coli, EcCsdB.EcSelD interaction must occur to prevent cell death from the release of the toxic intermediate selenide. Here, we demonstrate and characterize the in vitro EcSelD.EcCsdB interaction by biophysical methods. The EcSelD.EcCsdB interaction occurs with a stoichiometry of 1:1 in presence of selenocysteine and at a low-nanomolar affinity (~1.8 nM). The data is in agreement with the small angle X-ray scattering model fitted using available structures. Moreover, yeast-2-hybrid assays supported the macromolecular interaction in the cellular environment. This is the first report that demonstrates the interaction between EcCsdB and EcSelD supporting the hypothesis that EcSelD.EcCsdB interaction is necessary to sequester the selenide during the selenocysteine incorporation pathway in Bacteria.


Subject(s)
Lyases/chemistry , Lyases/metabolism , Phosphotransferases/chemistry , Phosphotransferases/metabolism , Selenocysteine/biosynthesis , Calorimetry, Differential Scanning , Escherichia coli/genetics , Escherichia coli/metabolism , Models, Molecular , Protein Stability , Protein Unfolding , Scattering, Small Angle , Selenium/metabolism , Spectrometry, Fluorescence , Thermodynamics , Two-Hybrid System Techniques , Ultracentrifugation
9.
Antioxidants (Basel) ; 8(9)2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31480609

ABSTRACT

The enzyme selenocysteine ß-lyase (SCLY) was first isolated in 1982 from pig livers, followed by its identification in bacteria. SCLY works as a homodimer, utilizing pyridoxal 5'-phosphate as a cofactor, and catalyzing the specific decomposition of the amino acid selenocysteine into alanine and selenide. The enzyme is thought to deliver its selenide as a substrate for selenophosphate synthetases, which will ultimately be reutilized in selenoprotein synthesis. SCLY subcellular localization is unresolved, as it has been observed both in the cytosol and in the nucleus depending on the technical approach used. The highest SCLY expression and activity in mammals is found in the liver and kidneys. Disruption of the Scly gene in mice led to obesity, hyperinsulinemia, glucose intolerance, and hepatic steatosis, with SCLY being suggested as a participant in the regulation of energy metabolism in a sex-dependent manner. With the physiological role of SCLY still not fully understood, this review attempts to discuss the available literature regarding SCLY in animals and provides avenues for possible future investigation.

10.
Free Radic Biol Med ; 127: 182-189, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29567390

ABSTRACT

Selenoprotein P (SelenoP) functions as a plasma transporter of selenium (Se) from liver to other tissues via incorporation into multiple selenocysteine (Sec) residues. Selenocysteine lyase (Scly) is an intracellular enzyme that decomposes Sec into selenide, providing Se for the synthesis of new selenoproteins. Both SelenoP and Scly are mostly produced by the liver. Previous studies demonstrated that male mice lacking SelenoP (SelenoP KO) or Scly (Scly KO) had increased or decreased total hepatic Se, respectively. While SelenoP regulation by Se is well-studied, Scly regulation by Se has not been reported. We hypothesize that Scly is negatively regulated by Se levels, and that absence of SelenoP jeopardizes Scly-dependent Se recycling. Using in vitro and in vivo models, we unveiled a tissue-specific Se regulation of Scly gene expression. We also determined that SelenoP, a considered source of intracellular Se, affects Scly expression and activity in vitro but not in vivo, as in the absence of SelenoP, Scly levels and activity remain normal. We also showed that absence of SelenoP does not increase levels of transsulfuration pathway enzymes, which would result in available selenocompounds being decomposed by the actions of cystathionine γ-lyase (CGL or CTH) and cystathionine ß-synthase (CBS). Instead, it affects levels of thioredoxin reductase 1 (Txnrd1), an enzyme that can reduce selenite to selenide to be used in selenoprotein production. This study evaluates a potential interplay between SelenoP and Scly, providing further insights into the regulation of selenium metabolism.


Subject(s)
Lyases/metabolism , Selenoprotein P/metabolism , Animals , Humans , Liver/metabolism
11.
Annu Rev Nutr ; 35: 109-34, 2015.
Article in English | MEDLINE | ID: mdl-25974694

ABSTRACT

Selenium is regulated in the body to maintain vital selenoproteins and to avoid toxicity. When selenium is limiting, cells utilize it to synthesize the selenoproteins most important to them, creating a selenoprotein hierarchy in the cell. The liver is the central organ for selenium regulation and produces excretory selenium forms to regulate whole-body selenium. It responds to selenium deficiency by curtailing excretion and secreting selenoprotein P (Sepp1) into the plasma at the expense of its intracellular selenoproteins. Plasma Sepp1 is distributed to tissues in relation to their expression of the Sepp1 receptor apolipoprotein E receptor-2, creating a tissue selenium hierarchy. N-terminal Sepp1 forms are taken up in the renal proximal tubule by another receptor, megalin. Thus, the regulated whole-body pool of selenium is shifted to needy cells and then to vital selenoproteins in them to supply selenium where it is needed, creating a whole-body selenoprotein hierarchy.


Subject(s)
Homeostasis/physiology , Selenium/metabolism , Animals , Biological Availability , Biological Transport , Biomarkers , Diet , Dietary Supplements , Health Status , Humans , Kidney Tubules, Proximal/metabolism , LDL-Receptor Related Proteins/physiology , Liver/physiology , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Nutritional Requirements , Organ Specificity , Selenium/deficiency , Selenium/pharmacokinetics , Selenocysteine/metabolism , Selenomethionine/metabolism , Selenoprotein P/analysis , Selenoprotein P/blood , Selenoproteins/biosynthesis , Selenoproteins/metabolism
12.
J Biol Chem ; 289(14): 9662-74, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24519931

ABSTRACT

Selenoproteins are a unique family of proteins, characterized by the co-translational incorporation of selenium as selenocysteine, which play key roles in antioxidant defense. Among selenoproteins, selenoprotein P (Sepp1) is particularly distinctive due to the fact that it contains multiple selenocysteine residues and has been postulated to act in selenium transport. Within the brain, Sepp1 delivers selenium to neurons by binding to the ApoER2 receptor. Upon feeding a selenium-deficient diet, mice lacking ApoER2 or Sepp1 develop severe neurological dysfunction and exhibit widespread brainstem neurodegeneration, indicating an important role for ApoER2-mediated Sepp1 uptake in normal brain function. Selenocysteine lyase (Scly) is an enzyme that plays an important role in selenium homeostasis, in that it catalyzes the decomposition of selenocysteine and allows selenium to be recycled for additional selenoprotein synthesis. We previously reported that constitutive deletion of Scly results in neurological deficits only when mice are challenged with a low selenium diet. To gain insight into the relationship between Sepp1 and Scly in selenium metabolism, we created novel transgenic mice constitutively lacking both genes (Scly(-/-)Sepp1(-/-)) and characterized the neurobehavioral phenotype. We report that deletion of Scly in conjunction with Sepp1 further aggravates the phenotype of Sepp1(-/-) mice, as these mice needed supraphysiological selenium supplementation to survive, and surviving mice exhibited impaired motor coordination, audiogenic seizures, and brainstem neurodegeneration. These findings provide the first in vivo evidence that Scly and Sepp1 work cooperatively to maintain selenoprotein function in the mammalian brain.


Subject(s)
Behavior, Animal , Brain/metabolism , Lyases/metabolism , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Seizures/metabolism , Selenoprotein P/metabolism , Animals , Brain/pathology , Lyases/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Seizures/genetics , Seizures/pathology , Selenocysteine/genetics , Selenocysteine/metabolism , Selenoprotein P/genetics
13.
Free Radic Biol Med ; 65: 1548-1556, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23867154

ABSTRACT

Whereas selenium was found to act as an insulin mimic and to be antidiabetic in earlier studies, recent animal experiments and human trials have shown an unexpected risk of prolonged high Se intake in potentiating insulin resistance and type 2 diabetes. Elevating dietary Se intake (0.4 to 3.0mg/kg of diet) above the nutrient requirements, similar to overproduction of selenoproteins, led to insulin resistance and/or diabetes-like phenotypes in mice, rats, and pigs. Although its diabetogenic mechanism remains unclear, high Se intake elevated activity or production of selenoproteins including GPx1, MsrB1, SelS, and SelP. This upregulation diminished intracellular reactive oxygen species and then dysregulated key regulators of ß cells and insulin synthesis and secretion, leading to chronic hyperinsulinemia. Overscavenging intracellular H2O2 also attenuated oxidative inhibition of protein tyrosine phosphatases and suppressed insulin signaling. High Se intake might affect expression and/or function of key regulators of glycolysis, gluconeogenesis, and lipogenesis. Future research is needed to find out if certain forms of Se metabolites in addition to selenoproteins and if mechanisms other than intracellular redox control mediate the diabetogenic effects of high Se intake. Furthermore, a potential interactive role of high Se intake in the interphase of carcinogenesis and diabetogenesis should be explored to make optimal use of Se in human nutrition and health.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Hyperinsulinism , Insulin Resistance , Selenium/pharmacology , Animals , Gluconeogenesis/drug effects , Glycolysis/drug effects , Hydrogen Peroxide/metabolism , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Lipogenesis/drug effects , Mice , Oxidation-Reduction/drug effects , Rats , Selenoproteins/metabolism , Signal Transduction , Swine
14.
Free Radic Biol Med ; 65: 291-300, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23811004

ABSTRACT

Murine regenerating islet-derived 3ß (Reg3ß) represents a homologue of human hepatocarcinoma-intestine-pancreas/pancreatic-associated protein and enhances mouse susceptibility to acetaminophen (APAP)-induced hepatotoxicity. Our objective was to determine if and how knockout of Reg3ß (KO) affects APAP (300 mg/kg, ip)-mediated protein nitration in mouse liver. APAP injection produced greater levels of hepatic protein nitration in the KO than in the wild-type mice. Their elevated protein nitration was alleviated by a prior injection of recombinant mouse Reg3ß protein and was associated with an accelerated depletion of the peroxynitrite (ONOO(-)) scavenger glutathione by an upregulated hepatic glutathione peroxidase-1 (GPX1) activity. The enhanced GPX1 production in the KO mice was mediated by an 85% rise (p<0.05) in the activity of selenocysteine lyase (Scly), a key enzyme that mobilizes Se for selenoprotein biosynthesis. Knockout of Reg3ß enhanced AP-1 protein and its binding activity to the Scly gene promoter, upregulating its gene transcription. However, knockout of Reg3ß did not affect gene expression of other key factors for selenoprotein biosynthesis. In conclusion, our findings unveil a new metabolic role for Reg3ß in protein nitration and a new biosynthesis control of GPX1 by a completely "unrelated" regenerating protein, Reg3ß, via transcriptional activation of Scly in coping with hepatic protein nitration. Linking selenoproteins to tissue regeneration will have profound implications in understanding the mechanism of Se functions and physiological coordination of tissue regeneration with intracellular redox control.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Glutathione Peroxidase/biosynthesis , Lyases/metabolism , Proteins/metabolism , Animals , Blotting, Western , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins , Proteins/genetics , Real-Time Polymerase Chain Reaction , Selenoproteins/metabolism , Up-Regulation , Glutathione Peroxidase GPX1
SELECTION OF CITATIONS
SEARCH DETAIL