Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 49(12): 6893-6907, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34142161

RESUMO

Selenoprotein P (SELENOP) is a major plasma selenoprotein that contains 10 Sec residues, which is encoded by the UGA stop codon. The mRNA for SELENOP has the unique property of containing two Sec insertion sequence (SECIS) elements, which is located in the 3' untranslated region (3'UTR). Here, we coincidentally identified a novel gene, CCDC152, by sequence analysis. This gene was located in the antisense region of the SELENOP gene, including the 3'UTR region in the genome. We demonstrated that this novel gene functioned as a long non-coding RNA (lncRNA) that decreased SELENOP protein levels via translational rather than transcriptional, regulation. We found that the CCDC152 RNA interacted specifically and directly with the SELENOP mRNA and inhibited its binding to the SECIS-binding protein 2, resulting in the decrease of ribosome binding. We termed this novel gene product lncRNA inhibitor of SELENOP translation (L-IST). Finally, we found that epigallocatechin gallate upregulated L-IST in vitro and in vivo, to suppress SELENOP protein levels. Here, we provide a new regulatory mechanism of SELENOP translation by an endogenous long antisense ncRNA.


Assuntos
Regulação da Expressão Gênica , Biossíntese de Proteínas , RNA Longo não Codificante/metabolismo , Selenoproteína P/genética , Catequina/análogos & derivados , Catequina/farmacologia , Linhagem Celular Tumoral , Regulação para Baixo , Humanos , RNA Longo não Codificante/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Selenoproteína P/biossíntese
2.
J Biol Chem ; 292(26): 10791-10800, 2017 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-28465347

RESUMO

Selenoprotein P (encoded by SELENOP in humans, Selenop in rat), a liver-derived secretory protein, induces resistance to insulin and vascular endothelial growth factor (VEGF) in type 2 diabetes. Suppression of selenoprotein P may provide a novel therapeutic approach to treating type 2 diabetes; however, few drugs inhibiting SELENOP expression in hepatocytes have been identified. The present findings demonstrate that eicosapentaenoic acid (EPA) suppresses SELENOP expression by inactivating sterol regulatory element-binding protein-1c (SREBP-1c, encoded by Srebf1 in rat) in H4IIEC3 hepatocytes. Treatment with EPA caused concentration- and time-dependent reduction in SELENOP promoter activity. EPA activated AMP-activated protein kinase (AMPK); however, the inhibitory effect of EPA on SELENOP promoter activity was not canceled with an AMPK inhibitor compound C and dominant-negative AMPK transfection. Deletion mutant promoter assays and computational analysis of transcription factor-binding sites conserved among the species resulted in identification of a sterol regulatory element (SRE)-like site in the SELENOP promoter. A chromatin immunoprecipitation (ChIP) assay revealed that EPA decreases binding of SREBP-1c to the SELENOP promoter. Knockdown of Srebf1 resulted in a significant down-regulation of Selenop expression. Conversely, SREBP-1c overexpression inhibited the suppressive effect of EPA. These data provide a novel mechanism of action for EPA involving improvement of systemic insulin sensitivity through the regulation of selenoprotein P production independently of the AMPK pathway and suggest an additional approach to developing anti-diabetic drugs.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Ácido Eicosapentaenoico/farmacologia , Hepatócitos/metabolismo , Selenoproteína P/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Ratos , Selenoproteína P/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
3.
J Biol Chem ; 289(1): 335-45, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24257750

RESUMO

Selenoprotein P (SeP; encoded by SEPP1 in humans) is a liver-derived secretory protein that induces insulin resistance in type 2 diabetes. Suppression of SeP might provide a novel therapeutic approach to treating type 2 diabetes, but few drugs that inhibit SEPP1 expression in hepatocytes have been identified to date. The present findings demonstrate that metformin suppresses SEPP1 expression by activating AMP-activated kinase (AMPK) and subsequently inactivating FoxO3a in H4IIEC3 hepatocytes. Treatment with metformin reduced SEPP1 promoter activity in a concentration- and time-dependent manner; this effect was cancelled by co-administration of an AMPK inhibitor. Metformin also suppressed Sepp1 gene expression in the liver of mice. Computational analysis of transcription factor binding sites conserved among the species resulted in identification of the FoxO-binding site in the metformin-response element of the SEPP1 promoter. A luciferase reporter assay showed that metformin suppresses Forkhead-response element activity, and a ChIP assay revealed that metformin decreases binding of FoxO3a, a direct target of AMPK, to the SEPP1 promoter. Transfection with siRNAs for Foxo3a, but not for Foxo1, cancelled metformin-induced luciferase activity suppression of the metformin-response element of the SEPP1 promoter. The overexpression of FoxO3a stimulated SEPP1 promoter activity and rescued the suppressive effect of metformin. Metformin did not affect FoxO3a expression, but it increased its phosphorylation and decreased its nuclear localization. These data provide a novel mechanism of action for metformin involving improvement of systemic insulin sensitivity through the regulation of SeP production and suggest an additional approach to the development of anti-diabetic drugs.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Selenoproteína P/biossíntese , Proteínas Quinases Ativadas por AMP/genética , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/genética , Animais , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Ratos , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética , Selenoproteína P/genética
4.
Alcohol Clin Exp Res ; 39(5): 818-26, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25864381

RESUMO

BACKGROUND: Binge drinking (BD) is the most common ethanol (EtOH) intake consumption model among teenagers, but little is known about its effects on the liver. During its hepatic metabolism, acute alcohol exposure produces a great amount of reactive oxygen species which contributes to alcohol-induced liver injury. Selenium (Se) plays a key role in antioxidant defense as it forms part of selenoproteins, such as the antioxidant glutathione peroxidases (GPxs) or the selenoprotein P (SelP), synthesized mainly in liver. Chronic EtOH consumption decreases both Se deposits and this tissue's antioxidant activity. METHODS: Two BD administration routes (oral and intraperitoneal) were used in adolescent rats to analyze Se homeostasis; the main hepatic selenoproteins' expression: GPx1, GPx4, and SelP, and their biological roles related to oxidation. Their relationship with inflammatory processes was also determined by analyzing the expression of the transcriptional factor nuclear factor-kappa beta (NF-κB). RESULTS: It has been demonstrated for the first time that BD in adolescents alters Se homeostasis regardless of the administration route employed, despite the fact that the BD oral group ingested less Se in diet. This decrease of Se in serum and liver is directly related to a decrease in serum GPx3 and hepatic GPx1 activity, contributing to the oxidative imbalance found. The depletion of Se detected in liver affects GPx1 expression and, surprisingly, GPx4 expression. This could be related to the lower expression of the transcriptional factor NF-κB in the liver, a key player in the regulation of inflammatory processes. CONCLUSIONS: Due to the above, and to find whether a Se supplementation therapy improves these situations, it would be interesting to explore in more depth the relationship between Se, the high oxidation found, and the depressed immune response reported in BD adolescents.


Assuntos
Consumo Excessivo de Bebidas Alcoólicas/metabolismo , Etanol/administração & dosagem , Etanol/farmacologia , Homeostase/efeitos dos fármacos , Fígado/metabolismo , Selênio/metabolismo , Selenoproteína P/biossíntese , Administração Oral , Fatores Etários , Animais , Fezes/química , Glutationa Peroxidase/biossíntese , Glutationa Peroxidase/sangue , Infusões Parenterais , Masculino , NF-kappa B/biossíntese , Estresse Oxidativo/efeitos dos fármacos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Ratos , Selênio/análise , Selênio/sangue , Selênio/urina , Glutationa Peroxidase GPX1
5.
J Biol Chem ; 287(43): 36455-64, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22932905

RESUMO

Cellular methylation processes enable expression of gluconeogenic enzymes and metabolism of the nutrient selenium. Selenium status has been proposed to relate to type II diabetes risk, and plasma levels of selenoprotein P (SEPP1) have been positively correlated with insulin resistance. Increased expression of gluconeogenic enzymes glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxykinase 1 (PCK1) has negative consequences for blood glucose management in type II diabetics. Transcriptional regulation of SEPP1 is directed by the same transcription factors that control the expression of G6PC and PCK1, and these factors are activated by methylation of arginine residues. We sought to determine whether expression of SEPP1 and the aforementioned glucoconeogenic enzymes are regulated by protein methylation, the levels of which are reliant upon adequate S-adenosylmethionine (SAM) and inhibited by S-adenosylhomocysteine (SAH). We treated a human hepatocyte cell line, HepG2, with inhibitors of adenosylhomocysteine hydrolase (AHCY) known to increase concentration of SAH before analysis of G6PC, PCK1, and SEPP1 expression. Increasing SAH decreased 1) the SAM/SAH ratio, 2) protein-arginine methylation, and 3) expression of SEPP1, G6PC, and PCK1 transcripts. Furthermore, hormone-dependent induction of gluconeogenic enzymes was reduced by inhibition of protein methylation. When protein-arginine methyltransferase 1 expression was reduced by siRNA treatment, G6PC expression was inhibited. These findings demonstrate that hepatocellular SAM-dependent protein methylation is required for both SEPP1 and gluconeogenic enzyme expression and that inhibition of protein arginine methylation might provide a route to therapeutic interventions in type II diabetes.


Assuntos
Regulação da Expressão Gênica , Gluconeogênese , S-Adenosilmetionina/metabolismo , Selenoproteína P/biossíntese , Adenosil-Homocisteinase/biossíntese , Adenosil-Homocisteinase/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Glucose-6-Fosfato/genética , Glucose-6-Fosfato/metabolismo , Células Hep G2 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/genética , Metilação , Fosfoenolpiruvato Carboxiquinase (GTP)/biossíntese , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Proteína-Arginina N-Metiltransferases/biossíntese , Proteína-Arginina N-Metiltransferases/genética , S-Adenosilmetionina/genética , Selenoproteína P/genética
6.
Nihon Rinsho ; 70(1): 157-63, 2012 Jan.
Artigo em Japonês | MEDLINE | ID: mdl-22413512

RESUMO

The liver may maintain systemic homeostasis by releasing secretory proteins into the blood stream. In fact, comprehensive gene expression analysis revealed that the liver in humans expresses various kinds of genes encoding secretory proteins. Recently, function-unknown liver-derived secretory proteins have been termed as hepatokines. Using comprehensive gene expression analyses, we have identified selenoprotein P(SeP) as a novel hepatokine that elevates plasma glucose levels by inducing insulin resistance. RNA interference-mediated knockdown of SeP in the liver improved insulin resistance and hyperglycemia in type 2 diabetic mice, suggesting that SeP is a novel therapeutic target for type 2 diabetes. Further functional characterization of hepatokines might provide novel diagnostic and therapeutic targets for the other diseases except diabetes.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Selenoproteína P/biossíntese , Animais , Sistemas de Liberação de Medicamentos , Humanos , Fígado/metabolismo , Camundongos
7.
Biochem J ; 429(1): 43-51, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20370716

RESUMO

Selenium modifies inflammatory reactions in rodents and humans. The liver controls metabolism and transport of selenium via hepatically-derived SEPP (selenoprotein P). Intracellular SEPS (selenoprotein S) modifies endoplasmic-reticulum function and immune-cell activity. Polymorphisms in SEPS have been associated with cytokine levels and inflammatory diseases in a subset of clinical studies. In the present study, we hypothesized that sex and selenium represent decisive parameters controlling the immune response and regulation of SEPS expression in vivo. Male and female mice fed a selenium-poor diet were supplemented or not with selenite for 3 days and injected with saline or LPS (lipopolysaccharide) 24 h before analysis. Selenium supplementation mitigated the LPS-induced rise in circulating cytokines in male mice. Serum SepP and selenium concentrations decreased in response to LPS, whereas hepatic SepS was specifically up-regulated despite declining selenium concentrations in the liver. Hepatic SepS induction was mainly controlled by post-transcriptional mechanisms and attributed to hepatocytes by analysing transgenic mice. Notably, selenium supplementation was essential for an optimal SepS induction. We conclude that selenoprotein biosynthesis becomes redirected in hepatocytes during the acute-phase response at the expense of dispensable selenoproteins (e.g. SepP) and in favour of SepS expression, thereby causing declining serum selenium and improving liver function. The selenium status and sex control SepS expression and modify cytokine response patterns in serum, which might explain contradictory results on associations of SEPS genotype and inflammatory diseases in clinical studies.


Assuntos
Reação de Fase Aguda/metabolismo , Proteínas de Membrana/biossíntese , Selênio/administração & dosagem , Selenoproteína P/biossíntese , Selenoproteínas/biossíntese , Caracteres Sexuais , Reação de Fase Aguda/imunologia , Reação de Fase Aguda/fisiopatologia , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Feminino , Regulação da Expressão Gênica/imunologia , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Selênio/deficiência , Selenoproteína P/genética , Selenoproteínas/genética
8.
FASEB J ; 23(6): 1758-65, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19136613

RESUMO

The acute-phase response (APR) is characterized by an impaired metabolism of the essential trace element selenium (Se). Moreover, low-Se concentrations correlate to mortality risk in sepsis. Therefore, we analyzed the expression of the central Se transport and storage protein selenoprotein P (Sepp1) during an APR in mice. Serum Se and Sepp1 concentrations declined in parallel after injection of lipopolysaccharide to 50 and 39% of control-injected littermates, respectively. This negative APR proceeded largely independent from hepatic Sepp1 transcript concentrations. Instead, we identified a set of hepatic transcripts involved in Se metabolism, which declined coordinately during the APR, including the selenocysteine-specific elongation factor (EFsec), selenophosphate-synthetase 2 (Sephs2), selenocysteine-tRNA[Ser]Sec synthase (SecS), and phosphoseryl-tRNA[Ser]Sec kinase (Pstk). Pstk reacted most strongly and qualified as a new limiting factor for Sepp1 biosynthesis in siRNA-mediated knockdown experiments in hepatocytes in culture. Analogous experiments were performed with mice transgenic for hepatocyte-specific human Sepp1 cDNA to verify this hypothesis. Similar kinetics and effect sizes of Sepp1 expression were observed as before in wild-type mice. We conclude that hepatic translation of Sepp1 mRNA is specifically impaired during the APR. This deficit disrupts regular Se metabolism, transport, and supply to peripheral tissues and likely aggravates the pathological status.


Assuntos
Reação de Fase Aguda/metabolismo , Fígado/metabolismo , Selênio/metabolismo , Selenoproteína P/biossíntese , Animais , Regulação para Baixo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Selenoproteína P/sangue , Selenoproteína P/genética
9.
Biochem Biophys Res Commun ; 387(1): 158-63, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19576170

RESUMO

High serum selenium levels have been associated epidemiologically with increased incidence of type 2 diabetes. The major fraction of total selenium in serum is represented by liver-derived selenoprotein P (SeP). This study was undertaken to test for a hypothesized effect of hyperglycemia and the antihyperglycemic drug metformin on hepatic selenoprotein P biosynthesis. Cultivation of rat hepatocytes in the presence of high glucose concentrations (25 mmol/l) resulted in increased selenoprotein P mRNA expression and secretion. Treatment with metformin dose-dependently downregulated SeP mRNA expression and secretion, and suppressed glucocorticoid-stimulated production of SeP. Moreover, metformin strongly decreased mRNA levels of selenophosphate synthetase 2 (SPS-2), an enzyme essential for selenoprotein biosynthesis. Taken together, these results indicate an influence of metformin on selenium metabolism in hepatocytes. As selenoprotein P is the major transport form of selenium, metformin treatment may thereby diminish selenium supply to extrahepatic tissues.


Assuntos
Hiperglicemia/metabolismo , Hipoglicemiantes/farmacologia , Fígado/efeitos dos fármacos , Metformina/farmacologia , Selenoproteína P/antagonistas & inibidores , Animais , Células Cultivadas , Glucocorticoides/antagonistas & inibidores , Glucocorticoides/farmacologia , Glucose/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/biossíntese , Ratos , Selenoproteína P/biossíntese
10.
Mol Cell Biol ; 26(24): 9177-84, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17000762

RESUMO

Selenocysteine is incorporated into proteins via "recoding" of UGA from a stop codon to a sense codon, a process that requires specific secondary structures in the 3' untranslated region, termed selenocysteine incorporation sequence (SECIS) elements, and the protein factors that they recruit. Whereas most selenoprotein mRNAs contain a single UGA codon and a single SECIS element, selenoprotein P genes encode multiple UGAs and two SECIS elements. We have identified evolutionary adaptations in selenoprotein P genes that contribute to the efficiency of incorporating multiple selenocysteine residues in this protein. The first is a conserved, inefficiently decoded UGA codon in the N-terminal region, which appears to serve both as a checkpoint for the presence of factors required for selenocysteine incorporation and as a "bottleneck," slowing down the progress of elongating ribosomes. The second adaptation involves the presence of introns downstream of this inefficiently decoded UGA which confer the potential for nonsense-mediated decay when factors required for selenocysteine incorporation are limiting. Third, the two SECIS elements in selenoprotein P mRNA function with differing efficiencies, affecting both the rate and the efficiency of decoding different UGAs. The implications for how these factors contribute to the decoding of multiple selenocysteine residues are discussed.


Assuntos
Códon/genética , Biossíntese de Proteínas , Ribossomos/genética , Selenocisteína/metabolismo , Selenoproteína P/genética , Proteínas de Peixe-Zebra/genética , Animais , Linhagem Celular , Códon de Terminação/genética , Evolução Molecular , Humanos , Mutação , Biossíntese de Proteínas/genética , Precursores de RNA/biossíntese , Precursores de RNA/genética , Precursores de RNA/metabolismo , Selenocisteína/genética , Selenoproteína P/biossíntese , Selenoproteína P/metabolismo , Deleção de Sequência , Peixe-Zebra , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/metabolismo
11.
Cell Host Microbe ; 25(4): 588-601.e7, 2019 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-30974086

RESUMO

Patients infected with hepatitis C virus (HCV) have an increased risk of developing type 2 diabetes. HCV infection is linked to various liver abnormalities, potentially contributing to this association. We show that HCV infection increases the levels of hepatic selenoprotein P (SeP) mRNA (SEPP1 mRNA) and serum SeP, a hepatokine linked to insulin resistance. SEPP1 mRNA inhibits type I interferon responses by limiting the function of retinoic-acid-inducible gene I (RIG-I), a sensor of viral RNA. SEPP1 mRNA binds directly to RIG-I and inhibits its activity. SEPP1 mRNA knockdown in hepatocytes causes a robust induction of interferon-stimulated genes and decreases HCV replication. Clinically, high SeP serum levels are significantly associated with treatment failure of direct-acting antivirals in HCV-infected patients. Thus, SeP regulates insulin resistance and innate immunity, possibly inducing immune tolerance in the liver, and its upregulation may explain the increased risk of type 2 diabetes in HCV-infected patients.


Assuntos
Proteína DEAD-box 58/antagonistas & inibidores , Hepatite C/patologia , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , RNA Mensageiro/metabolismo , Selenoproteína P/biossíntese , Humanos , Receptores Imunológicos
12.
Free Radic Biol Med ; 118: 98-107, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29496617

RESUMO

Hepatic production and release of metabolites, nutrients and micronutrient transporters is tightly regulated at the level of gene expression. In this regard, transcription factor FOXO1 modulates the expression of genes such as G6PC and SELENOP, encoding the catalytic subunit of glucose 6-phosphatase and the plasma selenium transporter selenoprotein P, respectively. Here, we analyzed the role of cysteine residues in FOXO1 in controlling its activity with respect to regulation of G6PC and SELENOP in HepG2 human hepatoma cells. None of the seven FOXO1 cysteines affected FOXO1 binding to DNA or its basal subcellular distribution. Whereas overexpression of wildtype FOXO1 caused a strong induction of both G6PC and SELENOP promoter activities and mRNA levels, the induction was lowered by approx. 50% if cysteine-deficient FOXO1 was overexpressed instead. Only the most C-terminal of the seven FOXO1 cysteines, Cys612, was required and sufficient to ensure full FOXO1 transactivation activity. Coexpression of FOXO1 coregulators, CBP or PGC1α, had a strong synergistic effect in stimulating G6PC promoter activity and expression, fully relying on the presence of FOXO1 Cys612. Similarly, a synergistic effect of FOXO1 and CBP was observed for SELENOP. In contrast, stimulation of SELENOP by PGC1α was independent of FOXO1-Cys612, due to the close proximity of a hepatocyte nuclear factor-4α binding site to the FOXO1 binding site within the SELENOP promoter, as demonstrated using mutant SELENOP promoter constructs. In summary, full basal FOXO1 transactivation activity relies on Cys612, which mediates synergistic effects of coregulators, CBP or PGC1α, on FOXO1 transcriptional activity. The extent of Cys612 contribution depends on the promoter context of FOXO1 target genes.


Assuntos
Proteína Forkhead Box O1/química , Proteína Forkhead Box O1/metabolismo , Fragmentos de Peptídeos/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Sialoglicoproteínas/metabolismo , Ativação Transcricional/fisiologia , Cisteína/química , Glucose-6-Fosfatase/biossíntese , Células HEK293 , Células Hep G2 , Humanos , Selenoproteína P/biossíntese
13.
J Neural Transm (Vienna) ; 114(7): 877-84, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17245539

RESUMO

Selenoprotein P (SePP) is central to selenium (Se) metabolism in the mammalian organism. Human SePP contains 10 Se atoms that are covalent constituents of the polypeptide chain incorporated as the rare amino acid selenocysteine (Sec). Since hepatocytes secrete SePP into plasma, SePP is commonly regarded as a Se transport protein, although SePP mRNA is expressed in many organs. Gene targeting of SePP in mice leads to neurological dysfunction resulting from Se deficiency and associated reduction of selenoenzyme activities in the brain. However, more recent data revealed that isolated hepatic SePP deficiency does not alter brain Se levels, suggesting a role for SePP locally expressed in the brain. Some of the best characterized and most abundant selenoenzymes, glutathione peroxidases, thioredoxin reductases, and methionine sulfoxide reductase B, play major roles in the cellular defense against reactive oxygen species. Therefore, it was hypothesized that reduced brain Se bioavailability may be involved in the pathogenesis of neurodegenerative disease and normal ageing. We present evidence that human CSF contains SePP and that the human brain expresses SePP mRNA. Moreover, SePP-like immunoreactivity localizes to neurons and ependymal cells and thus appears strategically situated for maintenance and control of Se-dependent anti-oxidative defense systems.


Assuntos
Encéfalo/metabolismo , Epêndima/metabolismo , Regulação da Expressão Gênica , Neurônios/metabolismo , Proteoma/biossíntese , Selenoproteína P/biossíntese , Selenoproteína P/metabolismo , Adulto , Animais , Antioxidantes/metabolismo , Encéfalo/citologia , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Epêndima/química , Regulação da Expressão Gênica/fisiologia , Humanos , Soros Imunes/metabolismo , Imuno-Histoquímica , Recém-Nascido , Camundongos , Neurônios/química , Proteoma/química , Selênio/sangue , Selênio/fisiologia , Selenoproteína P/imunologia
14.
Sci Rep ; 7(1): 4391, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28663583

RESUMO

Selenoprotein biosynthesis relies on the co-translational insertion of selenocysteine in response to UGA codons. Aminoglycoside antibiotics interfere with ribosomal function and may cause codon misreading. We hypothesized that biosynthesis of the selenium (Se) transporter selenoprotein P (SELENOP) is particularly sensitive to antibiotics due to its ten in frame UGA codons. As liver regulates Se metabolism, we tested the aminoglycosides G418 and gentamicin in hepatoma cell lines (HepG2, Hep3B and Hepa1-6) and in experimental mice. In vitro, SELENOP levels increased strongly in response to G418, whereas expression of the glutathione peroxidases GPX1 and GPX2 was marginally affected. Se content of G418-induced SELENOP was dependent on Se availability, and was completely suppressed by G418 under Se-poor conditions. Selenocysteine residues were replaced mainly by cysteine, tryptophan and arginine in a codon-specific manner. Interestingly, in young healthy mice, antibiotic treatment failed to affect Selenop biosynthesis to a detectable degree. These findings suggest that the interfering activity of aminoglycosides on selenoprotein biosynthesis can be severe, but depend on the Se status, and other parameters likely including age and general health. Focused analyses with aminoglycoside-treated patients are needed next to evaluate a possible interference of selenoprotein biosynthesis by the antibiotics and elucidate potential side effects.


Assuntos
Aminoglicosídeos/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Selênio/deficiência , Selenoproteína P/biossíntese , Aminoácidos , Animais , Linhagem Celular Tumoral , Cromatografia Líquida , Códon de Terminação , Expressão Gênica , Gentamicinas/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Masculino , Camundongos , Selenoproteína P/genética , Espectrometria de Massas em Tandem
15.
J Mol Endocrinol ; 50(1): 31-42, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23125459

RESUMO

Progressive loss of pancreatic ß-cell mass is a crucial feature of type 2 diabetes mellitus. As ß-cells express very low amounts of the antioxidant enzymes catalase and glutathione peroxidase (GPx), they appear to be particularly vulnerable to oxidative damage in the pathogenesis of diabetes. Here, we investigated the pancreatic expression pattern and regulation of selenoprotein P (Sepp1), which may serve as an additional antioxidant enzyme inside and outside of cells. Sepp1 was detected in rodent pancreas by immunofluorescence and real-time RT-PCR. Regulation of Sepp1 biosynthesis in INS-1 rat insulinoma cells was investigated by real-time RT-PCR, luciferase gene reporter assay, and immunoblotting. Sepp1 and Gpx1 gene expressions in rat pancreas were 58 and 22% respectively of the liver values. Pancreatic Sepp1 expression was restricted to the endocrine tissue, with Sepp1 being present in the α- and ß-cells of mouse islets. In INS-1 insulinoma cells, Sepp1 expression was stimulated by the selenium compound sodium selenate and diminished in the presence of high glucose (16.7 vs 5  mM) concentrations. Sepp1 mRNA stability was also lowered at 16.7  mM glucose. Moreover, Sepp1 mRNA levels were decreased in isolated murine islets cultured in high-glucose (22  mM) medium compared with normal glucose (5.5  mM) medium. Pancreatic Sepp1 expression was elevated upon treatment of mice with the ß-cell toxin streptozotocin. This study shows that pancreatic islets express relatively high levels of Sepp1 that may fulfill a function in antioxidant protection of ß-cells. Downregulation of Sepp1 expression by high glucose might thus contribute to glucotoxicity in ß-cells.


Assuntos
Ilhotas Pancreáticas/metabolismo , Selenoproteína P/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Regulação para Baixo , Imunofluorescência , Genes Reporter , Glucose/administração & dosagem , Camundongos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Selenoproteína P/biossíntese , Selenoproteína P/genética , Estreptozocina/farmacologia
16.
Anim Reprod Sci ; 135(1-4): 31-6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23084568

RESUMO

In this experiment the effect of maternal dietary selenium on the expression of Sel P and apoER2 of goat offspring was studied. The experiment was conducted on 119 Taihang Black Goats randomly divided into 4 groups which were fed with a basal diet, supplemented with 0 (control), 0.5, 2 and 4 mg kg(-1) DM Se. Testis samples were collected from young male of each treatment group at the end of the study (30 d after weaning) for mRNA expression using real-time PCR and for protein expression by immunohistochemistry assay. A significant decrease was observed in mRNA expression of Sel P and apoER2 in the testis of the Se-deficient (Group 1) and the Se-excess (Group 4) compared with that in Groups 2 and 3. A similar trend of the protein expression of Sel P and apoER2 was also found. These data indicate that maternal and dietary selenium has an effect on the expression of Sel P and apoER2 in testis of their offspring. In addition, both groups were similar suggesting that the relationship between Sel P and apoER2, and apoER2 is a receptor of Sel P in the seminiferous epithelium to uptake the selenium.


Assuntos
Cabras/metabolismo , Proteínas Relacionadas a Receptor de LDL/biossíntese , Selênio/administração & dosagem , Selenoproteína P/biossíntese , Testículo/efeitos dos fármacos , Testículo/metabolismo , Animais , Suplementos Nutricionais , Feminino , Imuno-Histoquímica/veterinária , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas Relacionadas a Receptor de LDL/metabolismo , Masculino , Exposição Materna/normas , Gravidez , RNA/química , RNA/genética , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Selênio/metabolismo , Selenoproteína P/genética , Selenoproteína P/metabolismo
17.
J Mol Biol ; 399(3): 385-96, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20417644

RESUMO

Selenoproteins are a family of proteins that share the common feature of containing selenocysteine, the "twenty-first" amino acid. Selenocysteine incorporation occurs during translation of selenoprotein messages by redefinition of UGA codons, which normally specify termination of translation. Studies of the eukaryotic selenocysteine incorporation mechanism suggest that selenocysteine insertion is inefficient compared with termination. Nevertheless, selenoprotein P and several other selenoproteins are known to contain multiple selenocysteines. The production of full-length (FL) protein from these messages would seem to demand highly efficient selenocysteine incorporation due to the compounding effect of termination at each UGA codon. We present data demonstrating that efficient incorporation of multiple selenocysteines can be reconstituted in rabbit reticulocyte lysate translation reactions. Selenocysteine incorporation at the first UGA codon is inefficient but increases by approximately 10-fold at subsequent downstream UGA codons. We found that ribosomes in the "processive" phase of selenocysteine incorporation (i.e., after decoding the first UGA codon as selenocysteine) are fully competent to terminate translation at UAG and UAA codons, that ribosomes become less efficient at selenocysteine incorporation as the distance between UGA codons is increased, and that efficient selenocysteine incorporation is not dependent on cis-acting elements unique to selenoprotein P. Furthermore, we found that the percentage of ribosomes decoding a UGA codon as selenocysteine rather than termination can be increased by 3- to 5-fold by placing the murine leukemia virus UAG read-through element upstream of the first UGA codon or by providing a competing messenger RNA in trans. The mechanisms of selenocysteine incorporation and selenoprotein synthesis are discussed in light of these results.


Assuntos
Selenocisteína/metabolismo , Selenoproteínas/biossíntese , Animais , Códon , Códon de Terminação , Técnicas In Vitro , Coelhos , Reticulócitos/metabolismo , Selenocisteína/genética , Selenoproteína P/biossíntese , Selenoproteína P/genética , Selenoproteínas/genética
18.
Free Radic Biol Med ; 49(5): 777-85, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20542496

RESUMO

Selenoprotein P (SeP), serving as selenium transporter and extracellular antioxidant, is assumed to have a protective role in the gastrointestinal tract, which is particularly susceptible to oxidative damage. Decreased SeP mRNA levels have been found in colon cancer; however, information on the control of intestinal SeP biosynthesis is scarce. We analyzed SeP biosynthesis in human intestinal epithelial Caco-2 cells subject to differentiation from crypt- to villous-like enterocytes. In the course of Caco-2 cell differentiation, SeP mRNA expression and secretion increased concomitant with three regulators of SeP transcription: hepatocyte nuclear factor-4alpha, forkhead box class O1a, and peroxisomal proliferator-activated receptor-gamma coactivator 1alpha. Treatment of differentiated Caco-2 cells with the proinflammatory cytokines IL-1beta, TNF-alpha, and IFN-gamma caused a down-regulation of SeP biosynthesis, resulting from induction of nitric oxide synthase 2. These observations were corroborated by decreased SeP mRNA levels in the colon of dextran sodium sulfate-treated mice, an animal model of experimental colitis. We conclude that inflammation of the intestinal mucosa causes a decline in locally produced selenoprotein P in the colon that eventually may contribute to the emergence of inflammatory bowel disease-related colorectal cancer.


Assuntos
Citocinas/farmacologia , Mediadores da Inflamação/farmacologia , Intestinos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Selenoproteína P/biossíntese , Animais , Células CACO-2 , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Feminino , Humanos , Doenças Inflamatórias Intestinais/complicações , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Selenoproteína P/genética
19.
Lung Cancer ; 65(1): 34-40, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19058871

RESUMO

Aim of the study was to investigate the mRNA expression level of selenoprotein P (SEPP1), 15-kDa selenoprotein (SEP15) and glutathione peroxidase 1 (hGPX1) in paired malignant and non-malignant tissue. To achieve this goal, the quantitative real-time PCR technique was utilized in paired tissue samples from 33 non-small cell lung cancer (NSCLC) patients. Simultaneously, the activity of glutathione peroxidases (GPX) and the level of thiobarbituric acid-reactive species (TBARS) in paired tissue specimens and the blood plasma selenium level was measured. We found significant down-regulation of SEPP1 expression level in tumorous lung tissue (2.732-fold; p<0.001). The expression of hGPX1 and SEP15 in tumorous tissue remained unchanged compared to healthy tissue. The level of TBARS in malignant tissue was significantly increased (p<0.005) and negatively correlated with SEPP1 expression level (R(S)=-0.3238; p<0.05). The activity of GPX in malignant tissue was significantly increased compared to the non-malignant one (p<0.005) and negatively correlated with the expression level of SEPP1. It seems possible, that the down-regulation of SEPP1 expression may lead to an increased oxidative stress possibly resulting in lung carcinogenesis. Increased activity of GPX in tumorous lung tissue seems to be a feedback mechanism.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Glutationa Peroxidase/biossíntese , Neoplasias Pulmonares/metabolismo , Selenoproteína P/biossíntese , Selenoproteínas/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/sangue , Carcinoma Pulmonar de Células não Pequenas/genética , Estudos de Casos e Controles , Feminino , Glutationa Peroxidase/genética , Humanos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/genética , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Selênio/sangue , Selenoproteína P/genética , Selenoproteínas/genética , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo , Glutationa Peroxidase GPX1
20.
J Biol Chem ; 282(51): 36797-807, 2007 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-17954931

RESUMO

A selenocysteine insertion sequence (SECIS) element in the 3'-untranslated region and an in-frame UGA codon are the requisite cis-acting elements for the incorporation of selenocysteine into selenoproteins. Equally important are the trans-acting factors SBP2, Sec-tRNA[Ser]Sec, and eEFSec. Multiple in-frame UGAs and two SECIS elements make the mRNA encoding selenoprotein P (Sel P) unique. To study the role of codon context in determining the efficiency of UGA readthrough at each of the 10 rat Sel P Sec codons, we individually cloned 27-nucleotide-long fragments representing each UGA codon context into a luciferase reporter construct harboring both Sel P SECIS elements. Significant differences, spanning an 8-fold range of UGA readthrough efficiency, were observed, but these differences were dramatically reduced in the presence of excess SBP2. Mutational analysis of the "fourth base" of contexts 1 and 5 revealed that only the latter followed the established rules for hierarchy of translation termination. In addition, mutations in either or both of the Sel P SECIS elements resulted in differential effects on UGA readthrough. Interestingly, even when both SECIS elements harbored a mutation of the core region required for Sec incorporation, context 5 retained a significantly higher level of readthrough than context 1. We also show that SBP2-dependent Sec incorporation is able to repress G418-induced UGA readthrough as well as eRF1-induced stimulation of termination. We conclude that a large codon context forms a cis-element that works together with Sec incorporation factors to determine readthrough efficiency.


Assuntos
Regiões 3' não Traduzidas/metabolismo , Códon de Terminação/metabolismo , Terminação Traducional da Cadeia Peptídica/fisiologia , Aminoacil-RNA de Transferência/metabolismo , Proteínas de Ligação a RNA/metabolismo , Selenocisteína/metabolismo , Regiões 3' não Traduzidas/genética , Animais , Sistema Livre de Células/metabolismo , Coccidiostáticos/farmacologia , Códon de Terminação/genética , Gentamicinas/farmacologia , Luciferases/biossíntese , Luciferases/genética , Terminação Traducional da Cadeia Peptídica/efeitos dos fármacos , Fatores de Alongamento de Peptídeos/genética , Fatores de Alongamento de Peptídeos/metabolismo , Fatores de Terminação de Peptídeos/genética , Fatores de Terminação de Peptídeos/metabolismo , Aminoacil-RNA de Transferência/genética , Proteínas de Ligação a RNA/genética , Coelhos , Ratos , Selenocisteína/genética , Selenoproteína P/biossíntese , Selenoproteína P/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA