Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
BMC Gastroenterol ; 24(1): 57, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302914

ABSTRACT

BACKGROUND: Liver fibrosis is a major risk factor for hepatocellular carcinoma (HCC). We have previously reported that differentially methylated regions (DMRs) are correlated with the fibrosis stages of metabolic dysfunction-associated steatotic liver disease (MASLD). In this study, the methylation levels of those DMRs in liver fibrosis and subsequent HCC were examined. METHODS: The methylation levels of DMRs were investigated using alcoholic cirrhosis and HCC (GSE60753). The data of hepatitis C virus-infected cirrhosis and HCC (GSE60753), and two datasets (GSE56588 and GSE89852) were used for replication analyses. The transcriptional analyses were performed using GSE114564, GSE94660, and GSE142530. RESULTS: Hypomethylated DMR and increased transcriptional level of zinc finger and BTB domain containing 38 (ZBTB38) were observed in HCC. Hypermethylated DMRs, and increased transcriptional levels of forkhead box K1 (FOXK1) and zinc finger CCCH-type containing 3 (ZC3H3) were observed in HCC. The methylation levels of DMR of kazrin, periplakin interacting protein (KAZN) and its expression levels were gradually decreased as cirrhosis progressed to HCC. CONCLUSIONS: Changes in the methylation and transcriptional levels of ZBTB38, ZC3H3, FOXK1, and KAZN are important for the development of fibrosis and HCC; and are therefore potential therapeutic targets and diagnostic tools for cirrhosis and HCC.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis C , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , DNA Methylation , Liver Cirrhosis/complications , Hepatitis C/complications , Forkhead Transcription Factors
2.
J Clin Invest ; 133(13)2023 07 03.
Article in English | MEDLINE | ID: mdl-37166989

ABSTRACT

Although selenium deficiency correlates with colorectal cancer (CRC) risk, the roles of the selenium-rich antioxidant selenoprotein P (SELENOP) in CRC remain unclear. In this study, we defined SELENOP's contributions to sporadic CRC. In human single-cell cRNA-Seq (scRNA-Seq) data sets, we discovered that SELENOP expression rose as normal colon stem cells transformed into adenomas that progressed into carcinomas. We next examined the effects of Selenop KO in a mouse adenoma model that involved conditional, intestinal epithelium-specific deletion of the tumor suppressor adenomatous polyposis coli (Apc) and found that Selenop KO decreased colon tumor incidence and size. We mechanistically interrogated SELENOP-driven phenotypes in tumor organoids as well as in CRC and noncancer cell lines. Selenop-KO tumor organoids demonstrated defects in organoid formation and decreases in WNT target gene expression, which could be reversed by SELENOP restoration. Moreover, SELENOP increased canonical WNT signaling activity in noncancer and CRC cell lines. In defining the mechanism of action of SELENOP, we mapped protein-protein interactions between SELENOP and the WNT coreceptors low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6). Last, we confirmed that SELENOP-LRP5/6 interactions contributed to the effects of SELENOP on WNT activity. Overall, our results position SELENOP as a modulator of the WNT signaling pathway in sporadic CRC.


Subject(s)
Adenoma , Colorectal Neoplasms , Selenium , Mice , Animals , Humans , Wnt Signaling Pathway , Selenoprotein P/genetics , Selenoprotein P/metabolism , Colorectal Neoplasms/pathology , Selenium/metabolism , Carcinogenesis/genetics , Adenoma/metabolism , Gene Expression Regulation, Neoplastic , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism
3.
BMC Gastroenterol ; 22(1): 278, 2022 Jun 02.
Article in English | MEDLINE | ID: mdl-35655171

ABSTRACT

BACKGROUND: We previously reported that two differentially methylated region (DMR) networks identified by DMR and co-methylation analyses are strongly correlated with the fibrosis stages of nonalcoholic fatty liver disease (NAFLD). In the current study, we examined these DMR networks in viral hepatitis and hepatocellular carcinoma (HCC). METHODS: We performed co-methylation analysis of DMRs using a normal dataset (GSE48325), two NAFLD datasets (JGAS000059 and GSE31803), and two HCC datasets (GSE89852 and GSE56588). The dataset GSE60753 was used for validation. RESULTS: One DMR network was clearly observed in viral hepatitis and two HCC populations. Methylation levels of genes in this network were higher in viral hepatitis and cirrhosis, and lower in HCC. Fatty acid binding protein 1 (FABP1), serum/glucocorticoid regulated kinase 2 (SGK2), and hepatocyte nuclear factor 4 α (HNF4A) were potential hub genes in this network. Increased methylation levels of the FABP1 gene may be correlated with reduced protection of hepatocytes from oxidative metabolites in NAFLD and viral hepatitis. The decreased methylation levels of SGK2 may facilitate the growth and proliferation of HCC cells. Decreased methylation levels of HNF4A in HCC may be associated with tumorigenesis. The other DMR network was observed in NAFLD, but not in viral hepatitis or HCC. This second network included genes involved in transcriptional regulation, cytoskeleton organization, and cellular proliferation, which are specifically related to fibrosis and/or tumorigenesis in NAFLD. CONCLUSIONS: Our results suggest that one DMR network was associated with fibrosis and tumorigenesis in both NAFLD and viral hepatitis, while the other network was specifically associated with NAFLD progression. Furthermore, FABP1, SGK2, and HNF4A are potential candidate targets for the prevention and treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis, Viral, Human , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic , Hepatitis, Viral, Human/complications , Hepatitis, Viral, Human/genetics , Humans , Liver Cirrhosis/complications , Liver Neoplasms/pathology , Non-alcoholic Fatty Liver Disease/pathology
4.
Front Neurosci ; 15: 631825, 2021.
Article in English | MEDLINE | ID: mdl-33927588

ABSTRACT

Dopamine (DA) transmission plays a critical role in processing rewarding and pleasurable stimuli. Increased synaptic DA release in the nucleus accumbens (NAc) is a central component of the physiological effects of drugs of abuse. The essential trace element selenium mitigates methamphetamine-induced neurotoxicity. Selenium can also alter DA production and turnover. However, studies have not directly addressed the role of selenium in DA neurotransmission. Selenoprotein P (SELENOP1) requires selenium for synthesis and transports selenium to the brain, in addition to performing other functions. We investigated whether SELENOP1 directly impacts (1) DA signaling and (2) the dopaminergic response to methamphetamine. We used fast-scan cyclic voltammetry to investigate DA transmission and the response to methamphetamine in NAc slices from C57/BL6J SELENOP1 KO mice. Recordings from SELENOP1 KO mouse slices revealed reduced levels of evoked DA release and slower DA uptake rates. Methamphetamine caused a dramatic increase in vesicular DA release in SELENOP1 KO mice not observed in wild-type controls. This elevated response was attenuated by SELENOP1 application through a selenium-independent mechanism involving SELENOP1-apolipoprotein E receptor 2 (ApoER2) interaction to promote dopamine D2 receptor (D2R) function. In wild-type mice, increased vesicular DA release in response to methamphetamine was revealed by blocking D2R activation, indicating that the receptor suppresses the methamphetamine-induced vesicular increase. Our data provide evidence of a direct physiological role for SELENOP1 in the dopaminergic response to methamphetamine and suggest a signaling role for the protein in DA transmission.

5.
Biochem Biophys Res Commun ; 510(1): 59-64, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30665718

ABSTRACT

Neural stem/progenitor cells (NSPCs) express higher levels of poly(ADP-ribose) polymerase 1 (PARP1) than mouse embryonic fibroblasts (MEFs). Inhibition of PARP induces the expression of several genes in the p53 signaling pathway, including p21, which is critical for cell cycle control at the G1/S phase, triggers apoptosis, and suppresses cell cycle progression in NSPCs. However, upon the up-regulation of p21, the cell cycle does not arrest at any specific phase. In the present study, the expression of genes specific to the G1/S and G2/M phases of the cell cycle were analyzed following treatment with PJ34 (N-[6-oxo-5,6-dihydro-phenanthridin-2-yl]-N,N-dimethylacetamide), an inhibitor of PARP. PJ34 treatment dramatically down-regulated cyclin B1 expression in NSPCs, but not in MEFs, which was confirmed by a promoter assay. Down-regulation of FoxM1 and B-MYB revealed that the down-regulation of cyclin B occurs at the transcriptional level. GADD45 was also specifically up-regulated in NSPCs. Taken together, the activation of p53 by PJ34 treatment in NSPCs induced changes in the expression of genes involved in the cell cycle. Fluorescence-activated cell sorting analysis revealed that PJ34 treatment suppressed G2/M to G1 progression in NSPCs, but not in MEFs. These data indicate that PJ34 treatment inhibits cyclin expression at the mRNA level and suppresses cell cycle progression in NSPCs.


Subject(s)
Cell Cycle/drug effects , Neural Stem Cells/cytology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , Cyclin B/drug effects , Cyclin B/genetics , Fibroblasts/drug effects , Genes, cdc/drug effects , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , RNA, Messenger/drug effects
6.
BMC Neurosci ; 18(1): 14, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28095779

ABSTRACT

BACKGROUND: Poly(ADP-ribose) polymerase 1 (PARP-1), which catalyzes poly(ADP-ribosyl)ation of proteins by using NAD+ as a substrate, plays a key role in several nuclear events, including DNA repair, replication, and transcription. Recently, PARP-1 was reported to participate in the somatic cell reprogramming process. Previously, we revealed a role for PARP-1 in the induction of neural apoptosis in a cellular model of cerebral ischemia and suggested the possible use of PARP inhibitors as a new therapeutic intervention. In the present study, we examined the effects of PARP inhibitors on neural stem/progenitor cells (NSPCs) of the mouse brain. RESULTS: PARP-1 was more abundant and demonstrated higher activity in NSPCs than in mouse embryonic fibroblasts. Treatment with PARP inhibitors suppressed the formation of neurospheres by NSPCs through the suppression of cell cycle progression and the induction of apoptosis. In order to identify the genes responsible for these effects, we investigated gene expression profiles by microarray analyses and found that several genes in the p53 signaling pathway were upregulated, including Cdkn1a, which is critical for cell cycle control, and Fas, Pidd, Pmaip1, and Bbc3, which are principal factors in the apoptosis pathway. Inhibition of poly(ADP-ribosyl)ation increased the levels of p53 protein, but not p53 mRNA, and enhanced the phosphorylation of p53 at Ser18. Experiments with specific inhibitors and also shRNA demonstrated that PARP-1, but not PARP-2, has a role in the regulation of p53. The effects of PARP inhibitors on NSPCs were not observed in Trp53 -/- NSPCs, suggesting a key role for p53 in these events. CONCLUSIONS: On the basis of the finding that PARP inhibitors facilitated the p53 signaling pathway, we propose that poly(ADP-ribosyl)ation contributes to the proliferation and self-renewal of NSPCs through the suppression of p53 activation.


Subject(s)
Neural Stem Cells/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Profiling , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred ICR , Mice, Knockout , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics
7.
FASEB J ; 28(8): 3579-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24760755

ABSTRACT

Selenoprotein P (Sepp1) and its receptor, apolipoprotein E receptor 2 (apoER2), account for brain retaining selenium better than other tissues. The primary sources of Sepp1 in plasma and brain are hepatocytes and astrocytes, respectively. ApoER2 is expressed in varying amounts by tissues; within the brain it is expressed primarily by neurons. Knockout of Sepp1 or apoER2 lowers brain selenium from ∼120 to ∼50 ng/g and leads to severe neurodegeneration and death in mild selenium deficiency. Interactions of Sepp1 and apoER2 that protect against this injury have not been characterized. We studied Sepp1, apoER2, and brain selenium in knockout mice. Immunocytochemistry showed that apoER2 mediates Sepp1 uptake at the blood-brain barrier. When Sepp1(-/-) or apoER2(-/-) mice developed severe neurodegeneration caused by mild selenium deficiency, brain selenium was ∼35 ng/g. In extreme selenium deficiency, however, brain selenium of ∼12 ng/g was tolerated when both Sepp1 and apoER2 were intact in the brain. These findings indicate that tandem Sepp1-apoER2 interactions supply selenium for maintenance of brain neurons. One interaction is at the blood-brain barrier, and the other is within the brain. We postulate that Sepp1 inside the blood-brain barrier is taken up by neurons via apoER2, concentrating brain selenium in them.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , LDL-Receptor Related Proteins/physiology , Nerve Degeneration/prevention & control , Selenium/metabolism , Selenoprotein P/physiology , Animals , Animals, Congenic , Biological Transport , Brain/embryology , Brain/growth & development , Capillaries/metabolism , Choroid Plexus/embryology , Choroid Plexus/growth & development , Choroid Plexus/metabolism , Endocytosis , Endothelial Cells/metabolism , Female , LDL-Receptor Related Proteins/deficiency , Low Density Lipoprotein Receptor-Related Protein-2/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/etiology , Nerve Degeneration/metabolism , Neurons/metabolism , Pregnancy , Selenium/administration & dosage , Selenium/deficiency , Selenium/pharmacokinetics , Selenoprotein P/deficiency
8.
J Biol Chem ; 289(13): 9195-207, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24532792

ABSTRACT

Sepp1 supplies selenium to tissues via receptor-mediated endocytosis. Mice, rats, and humans have 10 selenocysteines in Sepp1, which are incorporated via recoding of the stop codon, UGA. Four isoforms of rat Sepp1 have been identified, including full-length Sepp1 and three others, which terminate at the second, third, and seventh UGA codons. Previous studies have shown that the longer Sepp1 isoforms bind to the low density lipoprotein receptor apoER2, but the mechanism remains unclear. To identify the essential residues for apoER2 binding, an in vitro Sepp1 binding assay was developed using different Sec to Cys substituted variants of Sepp1 produced in HEK293T cells. ApoER2 was found to bind the two longest isoforms. These results suggest that Sepp1 isoforms with six or more selenocysteines are taken up by apoER2. Furthermore, the C-terminal domain of Sepp1 alone can bind to apoER2. These results indicate that apoER2 binds to the Sepp1 C-terminal domain and does not require the heparin-binding site, which is located in the N-terminal domain. Site-directed mutagenesis identified three residues of Sepp1 that are necessary for apoER2 binding. Sequential deletion of extracellular domains of apoER2 surprisingly identified the YWTD ß-propeller domain as the Sepp1 binding site. Finally, we show that apoER2 missing the ligand-binding repeat region, which can result from cleavage at a furin cleavage site present in some apoER2 isoforms, can act as a receptor for Sepp1. Thus, longer isoforms of Sepp1 with high selenium content interact with a binding site distinct from the ligand-binding domain of apoER2 for selenium delivery.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/chemistry , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Selenium/metabolism , Selenoprotein P/metabolism , Amino Acid Sequence , Animals , Endocytosis , Female , HEK293 Cells , Humans , Ligands , Male , Mice , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Selenocysteine/metabolism , Selenoprotein P/chemistry , Sequence Alignment , Substrate Specificity
9.
Free Radic Biol Med ; 69: 67-76, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24434121

ABSTRACT

Mouse selenoprotein P (Sepp1) consists of an N-terminal domain (residues 1-239) that contains one selenocysteine (U) as residue 40 in a proposed redox-active motif (-UYLC-) and a C-terminal domain (residues 240-361) that contains nine selenocysteines. Sepp1 transports selenium from the liver to other tissues by receptor-mediated endocytosis. It also reduces oxidative stress in vivo by an unknown mechanism. A previously uncharacterized plasma form of Sepp1 is filtered in the glomerulus and taken up by renal proximal convoluted tubule (PCT) cells via megalin-mediated endocytosis. We purified Sepp1 forms from the urine of megalin(-/-) mice using a monoclonal antibody to the N-terminal domain. Mass spectrometry revealed that the purified urinary Sepp1 consisted of N-terminal fragments terminating at 11 sites between residues 183 and 208. They were therefore designated Sepp1(UF). Because the N-terminal domain of Sepp1 has a thioredoxin fold, Sepp1(UF) were compared with full-length Sepp1, Sepp1(Δ240-361), and Sepp1(U40S) as a substrate of thioredoxin reductase-1 (TrxR1). All forms of Sepp1 except Sepp1(U40S), which contains serine in place of the selenocysteine, were TrxR1 substrates, catalyzing NADPH oxidation when coupled with H2O2 or tert-butylhydroperoxide as the terminal electron acceptor. These results are compatible with proteolytic cleavage freeing Sepp1(UF) from full-length Sepp1, the form that has the role of selenium transport, allowing Sepp1(UF) to function by itself as a peroxidase. Ultimately, plasma Sepp1(UF) and small selenium-containing proteins are filtered by the glomerulus and taken up by PCT cells via megalin-mediated endocytosis, preventing loss of selenium in the urine and providing selenium for the synthesis of glutathione peroxidase-3.


Subject(s)
Glutathione Peroxidase/biosynthesis , Oxidation-Reduction , Selenoproteins/metabolism , Thioredoxin Reductase 1/metabolism , Animals , Biological Transport , Endocytosis , Hydrogen Peroxide/metabolism , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mice , Protein Structure, Tertiary , Selenocysteine/metabolism , Selenoproteins/chemistry
10.
FASEB J ; 27(8): 3249-56, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23651543

ABSTRACT

Selenoprotein P (Sepp1) is taken up by receptor-mediated endocytosis for its selenium. The other extracellular selenoprotein, glutathione peroxidase-3 (Gpx3), has not been shown to transport selenium. Mice with genetic alterations of Sepp1, the Sepp1 receptors apolipoprotein E receptor-2 (apoER2) and megalin, and Gpx3 were used to investigate maternal-fetal selenium transfer. Immunocytochemistry (ICC) showed receptor-independent uptake of Sepp1 and Gpx3 in the same vesicles of d-13 visceral yolk sac cells, suggesting uptake by pinocytosis. ICC also showed apoER2-mediated uptake of maternal Sepp1 in the d-18 placenta. Thus, two selenoprotein-dependent maternal-fetal selenium transfer mechanisms were identified. Selenium was quantified in d-18 fetuses with the mechanisms disrupted. Maternal Sepp1 deletion, which lowers maternal whole-body selenium, decreased fetal selenium under selenium-adequate conditions but deletion of fetal apoER2 did not. Fetal apoER2 deletion did decrease fetal selenium, by 51%, under selenium-deficient conditions, verifying function of the placental Sepp1-apoER2 mechanism. Maternal Gpx3 deletion decreased fetal selenium, by 13%, but only under selenium-deficient conditions. These findings indicate that the selenoprotein uptake mechanisms ensure selenium transfer to the fetus under selenium-deficient conditions. The failure of their disruptions (apoER2 deletion, Gpx3 deletion) to affect fetal selenium under selenium-adequate conditions indicates the existence of an additional maternal-fetal selenium transfer mechanism.


Subject(s)
Glutathione Peroxidase/metabolism , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Maternal-Fetal Exchange/physiology , Selenium/metabolism , Selenoprotein P/metabolism , Animals , Biological Transport , Female , Glutathione Peroxidase/genetics , Immunohistochemistry , LDL-Receptor Related Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-2/genetics , Male , Maternal-Fetal Exchange/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Placenta/embryology , Placenta/metabolism , Pregnancy , Selenoprotein P/genetics , Time Factors , Yolk Sac/embryology , Yolk Sac/metabolism
11.
Met Ions Life Sci ; 13: 499-534, 2013.
Article in English | MEDLINE | ID: mdl-24470102

ABSTRACT

Selenium is an essential micronutrient in mammals, but is also recognized as toxic in excess. It is a non-metal with properties that are intermediate between the chalcogen elements sulfur and tellurium. Selenium exerts its biological functions through selenoproteins. Selenoproteins contain selenium in the form of the 21st amino acid, selenocysteine (Sec), which is an analog of cysteine with the sulfur-containing side chain replaced by a Se-containing side chain. Sec is encoded by the codon UGA, which is one of three termination codons for mRNA translation in non-selenoprotein genes. Recognition of the UGA codon as a Sec insertion site instead of stop requires a Sec insertion sequence (SECIS) element in selenoprotein mRNAs and a unique selenocysteyl-tRNA, both of which are recognized by specialized protein factors. Unlike the 20 standard amino acids, Sec is biosynthesized from serine on its tRNA. Twenty-five selenoproteins are encoded in the human genome. Most of the selenoprotein genes were discovered by bioinformatics approaches, searching for SECIS elements downstream of in-frame UGA codons. Sec has been described as having stronger nucleophilic and electrophilic properties than cysteine, and Sec is present in the catalytic site of all selenoenzymes. Most selenoproteins, whose functions are known, are involved in redox systems and signaling pathways. However, several selenoproteins are not well characterized in terms of their function. The selenium field has grown dramatically in the last few decades, and research on selenium biology is providing extensive new information regarding its importance for human health.


Subject(s)
Genome, Human/physiology , Selenium/metabolism , Selenocysteine/metabolism , Selenoproteins/metabolism , Animals , Codon, Terminator/metabolism , Humans , RNA, Transfer, Amino Acyl/metabolism
12.
Mol Cell Biol ; 32(20): 4141-54, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22890841

ABSTRACT

Selenium (Se) is an essential trace element used for biosynthesis of selenoproteins and is acquired either through diet or cellular recycling mechanisms. Selenocysteine lyase (Scly) is the enzyme that supplies Se for selenoprotein biosynthesis via decomposition of the amino acid selenocysteine (Sec). Knockout (KO) of Scly in a mouse affected hepatic glucose and lipid homeostasis. Mice lacking Scly and raised on an Se-adequate diet exhibit hyperinsulinemia, hyperleptinemia, glucose intolerance, and hepatic steatosis, with increased hepatic oxidative stress, but maintain selenoprotein levels and circulating Se status. Insulin challenge of Scly KO mice results in attenuated Akt phosphorylation but does not decrease phosphorylation levels of AMP kinase alpha (AMPKα). Upon dietary Se restriction, Scly KO animals develop several characteristics of metabolic syndrome, such as obesity, fatty liver, and hypercholesterolemia, with aggravated hyperleptinemia, hyperinsulinemia, and glucose intolerance. Hepatic glutathione peroxidase 1 (GPx1) and selenoprotein S (SelS) production and circulating selenoprotein P (Sepp1) levels are significantly diminished. Scly disruption increases the levels of insulin-signaling inhibitor PTP1B. Our results suggest a dependence of glucose and lipid homeostasis on Scly activity. These findings connect Se and energy metabolism and demonstrate for the first time a unique physiological role of Scly in an animal model.


Subject(s)
Lyases/metabolism , Metabolic Syndrome/metabolism , Selenium/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose Intolerance , Glutathione Peroxidase/analysis , Hypercholesterolemia/blood , Hypercholesterolemia/metabolism , Hyperinsulinism/blood , Leptin/blood , Lyases/genetics , Male , Metabolic Syndrome/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Oxidative Stress , Protein Tyrosine Phosphatase, Non-Receptor Type 1/analysis , Proto-Oncogene Proteins c-akt/metabolism , Selenium/blood , Selenoproteins/analysis , Glutathione Peroxidase GPX1
13.
J Biol Chem ; 287(34): 28717-26, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22761431

ABSTRACT

In vivo studies have shown that selenium is supplied to testis and brain by apoER2-mediated endocytosis of Sepp1. Although cultured cell lines have been shown to utilize selenium from Sepp1 added to the medium, the mechanism of uptake and utilization has not been characterized. Rat L8 myoblast cells were studied. They took up mouse Sepp1 from the medium and used its selenium to increase their glutathione peroxidase (Gpx) activity. L8 cells did not utilize selenium from Gpx3, the other plasma selenoprotein. Neither did they utilize it from Sepp1(Δ240-361), the isoform of Sepp1 that lacks the selenium-rich C-terminal domain. To identify Sepp1 receptors, a solubilized membrane fraction was passed over a Sepp1 column. The receptors apoER2 and Lrp1 were identified in the eluate by mass spectrometry. siRNA experiments showed that knockdown of apoER2, but not of Lrp1, inhibited (75)Se uptake from (75)Se-labeled Sepp1. The addition of protamine to the medium or treatment of the cells with chlorate also inhibited (75)Se uptake. Blockage of lysosome acidification did not inhibit uptake of Sepp1 but did prevent its digestion and thereby utilization of its selenium. These results indicate that L8 cells take up Sepp1 by an apoER2-mediated mechanism requiring binding to heparin sulfate proteoglycans. The presence of at least part of the selenium-rich C-terminal domain of Sepp1 is required for uptake. RT-PCR showed that mouse tissues express apoER2 in varying amounts. It is postulated that apoER2-mediated uptake of long isoform Sepp1 is responsible for selenium distribution to tissues throughout the body.


Subject(s)
Endocytosis/physiology , LDL-Receptor Related Proteins/metabolism , Lysosomes/metabolism , Myoblasts/metabolism , Selenoproteins/metabolism , Animals , Cell Line , Chlorates/pharmacology , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , LDL-Receptor Related Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-1 , Lysosomes/genetics , Mice , Mice, Knockout , Myoblasts/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Receptors, LDL/genetics , Receptors, LDL/metabolism , Selenoproteins/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
14.
J Nutr Sci Vitaminol (Tokyo) ; 57(4): 298-305, 2011.
Article in English | MEDLINE | ID: mdl-22041913

ABSTRACT

Selenocysteine lyase (SCL) catalyzes the decomposition of L-selenocysteine to yield L-alanine and selenium by acting exclusively on l-selenocysteine. The X-ray structural analysis of rat SCL has demonstrated how SCL discriminates L-selenocysteine from L-cysteine on the molecular basis. SCL has been proposed to function in the recycling of the micronutrient selenium from degraded selenoproteins containing selenocysteine residues, but the role of SCL in selenium metabolism in vivo remains unclear. We here demonstrate that the (75)Se-labeling efficiency of selenoproteins with (75)Se-labeled selenoprotein P (Sepp1) as a selenium source was decreased in HeLa cells transfected with SCL siRNA as compared to the cells transfected with control siRNA. Immunocytochemical analyses showed high SCL expression in kidney and liver cells, where selenocysteine is recovered from selenoproteins. Mature testes of mice exhibited a specific staining pattern of SCL in spermatids that actively produce selenoproteins. However, SCL was weakly expressed in Sertoli cells, which receive Sepp1 and supply selenium to germ cells. These demonstrate that SCL occurs in the cells requiring selenoproteins, probably to recycle selenium derived from selenoproteins such as Sepp1.


Subject(s)
Kidney/metabolism , Liver/metabolism , Lyases/metabolism , Selenium/metabolism , Selenocysteine/metabolism , Selenoproteins/biosynthesis , Spermatids/metabolism , Acebutolol , Animals , HeLa Cells , Humans , Isotope Labeling , Male , Mice , RNA, Small Interfering/metabolism , Rats , Selenoprotein P/metabolism , Selenoproteins/metabolism , Sertoli Cells/metabolism , Substrate Specificity , Transfection
15.
J Biol Chem ; 285(16): 12133-9, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20164179

ABSTRACT

Selenocysteine lyase (SCL) catalyzes the pyridoxal 5'-phosphate-dependent removal of selenium from l-selenocysteine to yield l-alanine. The enzyme is proposed to function in the recycling of the micronutrient selenium from degraded selenoproteins containing selenocysteine residue as an essential component. The enzyme exhibits strict substrate specificity toward l-selenocysteine and no activity to its cognate l-cysteine. However, it remains unclear how the enzyme distinguishes between selenocysteine and cysteine. Here, we present mechanistic studies of selenocysteine lyase from rat. ESI-MS analysis of wild-type and C375A mutant SCL revealed that the catalytic reaction proceeds via the formation of an enzyme-bound selenopersulfide intermediate on the catalytically essential Cys-375 residue. UV-visible spectrum analysis and the crystal structure of SCL complexed with l-cysteine demonstrated that the enzyme reversibly forms a nonproductive adduct with l-cysteine. Cys-375 on the flexible loop directed l-selenocysteine, but not l-cysteine, to the correct position and orientation in the active site to initiate the catalytic reaction. These findings provide, for the first time, the basis for understanding how trace amounts of a selenium-containing substrate is distinguished from excessive amounts of its cognate sulfur-containing compound in a biological system.


Subject(s)
Lyases/chemistry , Lyases/metabolism , Selenium/metabolism , Sulfur/metabolism , Amino Acid Substitution , Animals , Base Sequence , Catalytic Domain/genetics , Conserved Sequence , Crystallography, X-Ray , Cysteine/chemistry , DNA Primers/genetics , In Vitro Techniques , Lyases/genetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Protein Multimerization , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity
16.
J Biochem ; 143(4): 467-73, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18182386

ABSTRACT

Escherichia coli growing under anaerobic conditions produce H(2) and CO(2) by the enzymatic cleavage of formate that is produced from pyruvate at the end of glycolysis. Selenium is an integral part of formate dehydrogenase H (FDH H), which catalyses the first step in the formate hydrogen lyase (FHL) system. The genes of FHL system are transcribed only under anaerobic conditions, in the presence of a sigma 54-dependent transcriptional activator FhlA that binds formate as an effector molecule. Although the formate addition to the nutrient media has been an established procedure for inducing high FDH H activity, we have identified a low-salt nutrient medium containing <0.1% NaCl enabled constitutive, high expression of FDH H even without formate and d-glucose added to the medium. The novel conditions allowed us to study the effects of disrupting genes like trxB (thioredoxin reductase) or gor (glutathione reductase) on the production of FDH H activity and also reductive assimilation of selenite ( SeO 3(2-)) into the selenoprotein. Despite the widely accepted hypothesis that selenite is reduced by glutathione reductase-dependent system, it was demonstrated that trxB gene was essential for FDH H production and for labelling the FDH H polypeptide with 75Se-selenite. Our present study reports for the first time the physiological involvement of thioredoxin reductase in the reductive assimilation of selenite in E. coli.


Subject(s)
Escherichia coli/enzymology , Formate Dehydrogenases/metabolism , Hydrogenase/metabolism , Multienzyme Complexes/metabolism , Sodium Selenite/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Culture Media , Escherichia coli/growth & development , Mutation , Thioredoxin-Disulfide Reductase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...