Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Redox Biol ; 61: 102641, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36842241

RESUMO

At physiological levels, the trace element selenium plays a key role in redox reactions through the incorporation of selenocysteine in antioxidant enzymes. Selenium has also been evaluated as a potential anti-cancer agent, where selenium nanoparticles have proven effective, and are well tolerated in vivo at doses that are toxic as soluble Se. The use of such nanoparticles, coated with either serum albumin or the naturally occurring alkaline polysaccharide chitosan, also serves to enhance biocompatibility and bioavailability. Here we demonstrate a novel role for selenium in regulating histone methylation in ovarian cancer cell models treated with inorganic selenium nanoparticles coated with serum albumin or chitosan. As well as inducing thioredoxin reductase expression, ROS activity and cancer cell cytotoxicity, coated nanoparticles caused significant increases in histone methylation. Specifically, selenium nanoparticles triggered an increase in the methylation of histone 3 at lysines K9 and K27, histone marks involved in both the activation and repression of gene expression, thus suggesting a fundamental role for selenium in these epigenetic processes. This direct function was confirmed using chemical inhibitors of the histone lysine methyltransferases EZH2 (H3K27) and G9a/EHMT2 (H3K9), both of which blocked the effect of selenium on histone methylation. This novel role for selenium supports a distinct function in histone methylation that occurs due to a decrease in S-adenosylhomocysteine, an endogenous inhibitor of lysine methyltransferases, the metabolic product of methyl-group transfer from S-adenosylmethionine in the one-carbon metabolism pathway. These observations provide important new insights into the action of selenium nanoparticles. It is now important to consider both the classic antioxidant and novel histone methylation effects of this key redox element in its development in cancer therapy and other applications.


Assuntos
Quitosana , Selênio , Histonas/metabolismo , Metilação , Selênio/metabolismo , Lisina/metabolismo , S-Adenosil-Homocisteína/metabolismo , Antioxidantes/metabolismo , Quitosana/metabolismo , Histona-Lisina N-Metiltransferase/genética
2.
J Ethnopharmacol ; 255: 112738, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32147479

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Qian Yang Yu Yin Granule (QYYY) is a Chinese herbal formulation. It is used to treat hypertensive nephropathy for decades in China, but it is unknown that the exact mechanism of QYYY on hypertensive nephropathy. AIMS OF STUDY: The present study was to elucidate its epigenetic mechanism of QYYY on hypertensive nephropathy. MATERIALS AND METHODS: In the current study, HEK293T cells' proliferation induced by Ang II was chosen to observe epigenetic mechanisms of QYYY on renal damage. The cell proliferation was examined by MTT assays and ethynyldeoxyuridine analysis. Cell cycle analysis was performed. After treatment with QYYY, expression of Nicotinamide N-methyltransferase (NNMT), sirtuin1(SIRT1), S-adenosylhomocysteine(SAH), histone H3K4 methylation, and cortactin acetylation(acetyl-cortactin,ac-cortactin) were further investigated by western-blotting and real time PCR. DNA methylation was detected by ELISA. The study also observed the changes of SIRT1, SAH, H3K4 methylation, acetyl-cortactin when NNMT over-expressed by lentivirus transfection. Angiotensin II(Ang II) induced renal damage in spontaneously hypertensive rats(SHR). After eight weeks treatment of QYYY, blood pressure, serum and urine creatinine, and urinary microalbumin(mAlb) were assessed. The concentration of N1 -methylnicotinamide were detected by liquid chromatography with tandem mass spectrometry. The protein of NNMT, ac-cortactin, H3K3me3 were also assessed in vivo. RESULTS: QYYY inhibited HEK293T cells' proliferation, down-regulated the expression of NNMT, SAH, acetyl-cortactin and DNA methylation, up-regulated the expression of SIRT1, histone H3K4 trimethylation(H3K4me3). Over-expression of NNMT increased the expression of SAH and acetyl-cortactin, and reduced the expression of SIRT1 and H3K4me3. The study also demonstrated that QYYY promoted urinary creatinine excretion and reduced serum creatinine and urinary mAlb in SHR. QYYY decreased the concentration of N1 -methylnicotinamide in Ang II group. QYYY decreased the protein of NNMT, ac-cortactin and increased H3K4me3 in vivo. CONCLUSION: The results showed that QYYY alleviated renal impairment of SHR and inhibited HEK293T cells' proliferation induced by Ang II through the pathway of epigenetic mechanism linked to Nicotinamide N-Methyltransferase (NNMT) expression, including histone methylation, DNA methylation and acetyl-cortactin. This study unveiled a novel molecular mechanism by which QYYY controlled the progression of hypertensive nephropathy.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Epigênese Genética/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Hipertensão/tratamento farmacológico , Nefropatias/prevenção & controle , Rim/efeitos dos fármacos , Nicotinamida N-Metiltransferase/metabolismo , Acetilação , Angiotensina II , Animais , Proliferação de Células/efeitos dos fármacos , Cortactina/metabolismo , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Células HEK293 , Histonas/metabolismo , Humanos , Hipertensão/complicações , Hipertensão/enzimologia , Hipertensão/genética , Rim/enzimologia , Rim/patologia , Nefropatias/induzido quimicamente , Nefropatias/enzimologia , Nefropatias/genética , Masculino , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , S-Adenosil-Homocisteína/metabolismo , Sirtuína 1/metabolismo
3.
Sci Rep ; 9(1): 8059, 2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31147608

RESUMO

Catechol O-methyltransferase (COMT) is widely distributed in nature and installs a methyl group onto one of the vicinal hydroxyl groups of a catechol derivative. Enzymes belonging to this family require two cofactors for methyl transfer: S-adenosyl-l-methionine as a methyl donor and a divalent metal cation for regiospecific binding and activation of a substrate. We have determined two high-resolution crystal structures of Rv0187, one of three COMT paralogs from Mycobacterium tuberculosis, in the presence and absence of cofactors. The cofactor-bound structure clearly locates strontium ions and S-adenosyl-l-homocysteine in the active site, and together with the complementary structure of the ligand-free form, it suggests conformational dynamics induced by the binding of cofactors. Examination of in vitro activities revealed promiscuous substrate specificity and relaxed regioselectivity against various catechol-like compounds. Unexpectedly, mutation of the proposed catalytic lysine residue did not abolish activity but altered the overall landscape of regiospecific methylation.


Assuntos
Proteínas de Bactérias/metabolismo , Catecol O-Metiltransferase/metabolismo , Mycobacterium tuberculosis/enzimologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/isolamento & purificação , Proteínas de Bactérias/ultraestrutura , Domínio Catalítico/genética , Catecol O-Metiltransferase/genética , Catecol O-Metiltransferase/isolamento & purificação , Catecol O-Metiltransferase/ultraestrutura , Coenzimas/metabolismo , Cristalografia por Raios X , Ensaios Enzimáticos , Lisina/genética , Lisina/metabolismo , Metilação , Modelos Moleculares , Mutação , Mycobacterium tuberculosis/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , S-Adenosil-Homocisteína/metabolismo , Estrôncio/metabolismo , Especificidade por Substrato/genética
4.
Nutrients ; 10(4)2018 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-29587354

RESUMO

Elevated plasma total homocysteine (tHcy) is associated with increased risk of cardiovascular disease, but the mechanisms underlying this association are not completely understood. Cellular hypomethylation has been suggested to be a key pathophysiologic mechanism, since S-adenosylhomocysteine (AdoHcy), the Hcy metabolic precursor and a potent inhibitor of methyltransferase activity, accumulates in the setting of hyperhomocysteinemia. In this study, the impact of folate and methionine on intracellular AdoHcy levels and protein arginine methylation status was studied. Human endothelial cells were incubated with increasing concentrations of folinic acid (FnA), a stable precursor of folate, with or without methionine restriction. The levels of intracellular AdoHcy and AdoMet, tHcy in the cell culture medium, and protein-incorporated methylarginines were evaluated by suitable liquid chromatography techniques. FnA supplementation, with or without methionine restriction, reduced the level of tHcy and did not affect intracellular AdoMet levels. Interestingly, FnA supplementation reduced intracellular AdoHcy levels only in cells grown under methionine restriction. Furthermore, these cells also displayed increased protein arginine methylation status. These observations suggest that folic acid supplementation may enhance cellular methylation capacity under a low methionine status. Our results lead us to hypothesize that the putative benefits of folic acid supplementation in restoring endothelial homeostasis, thus preventing atherothrombotic events, should be reevaluated in subjects under a methionine restriction diet.


Assuntos
Arginina/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Leucovorina/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Homocisteína/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Metionina/farmacologia , Metilação , S-Adenosil-Homocisteína/metabolismo
5.
J Biol Chem ; 293(15): 5544-5555, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29414770

RESUMO

S-Adenosyl-l-homocysteine hydrolase (AdoHcy hydrolase; Sah1 in yeast/AHCY in mammals) degrades AdoHcy, a by-product and strong product inhibitor of S-adenosyl-l-methionine (AdoMet)-dependent methylation reactions, to adenosine and homocysteine (Hcy). This reaction is reversible, so any elevation of Hcy levels, such as in hyperhomocysteinemia (HHcy), drives the formation of AdoHcy, with detrimental consequences for cellular methylation reactions. HHcy, a pathological condition linked to cardiovascular and neurological disorders, as well as fatty liver among others, is associated with a deregulation of lipid metabolism. Here, we developed a yeast model of HHcy to identify mechanisms that dysregulate lipid metabolism. Hcy supplementation to wildtype cells up-regulated cellular fatty acid and triacylglycerol content and induced a shift in fatty acid composition, similar to changes observed in mutants lacking Sah1. Expression of the irreversible bacterial pathway for AdoHcy degradation in yeast allowed us to dissect the impact of AdoHcy accumulation on lipid metabolism from the impact of elevated Hcy. Expression of this pathway fully suppressed the growth deficit of sah1 mutants as well as the deregulation of lipid metabolism in both the sah1 mutant and Hcy-exposed wildtype, showing that AdoHcy accumulation mediates the deregulation of lipid metabolism in response to elevated Hcy in yeast. Furthermore, Hcy supplementation in yeast led to increased resistance to cerulenin, an inhibitor of fatty acid synthase, as well as to a concomitant decline of condensing enzymes involved in very long-chain fatty acid synthesis, in line with the observed shift in fatty acid content and composition.


Assuntos
Adenosil-Homocisteinase/metabolismo , Ácidos Graxos/metabolismo , Metabolismo dos Lipídeos , S-Adenosil-Homocisteína/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Adenosil-Homocisteinase/genética , Ácidos Graxos/genética , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Modelos Biológicos , Mutação , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
6.
J Nutr Biochem ; 48: 112-119, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28810182

RESUMO

The average intake of the essential trace element selenium (Se) is below the recommendation in most European countries, possibly causing sub-optimal expression of selenoproteins. It is still unclear how a suboptimal Se status may affect health. To mimic this situation, mice were fed one of three physiologically relevant amounts of Se. We focused on the liver, the organ most sensitive to changes in the Se supply indicated by hepatic glutathione peroxidase activity. In addition, liver is the main organ for synthesis of methyl groups and glutathione via one-carbon metabolism. Accordingly, the impact of Se on global DNA methylation, methylation capacity, and gene expression was assessed. We observed higher global DNA methylation indicated by LINE1 methylation, and an increase of the methylation potential as indicated by higher S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH) ratio and by elevated mRNA expression of serine hydroxymethyltransferase in both or either of the Se groups. Furthermore, increasing the Se supply resulted in higher plasma concentrations of triglycerides. Hepatic expression of glycolytic and lipogenic genes revealed consistent Se-dependent up-regulation of glucokinase. The sterol regulatory element-binding transcription factor 1 (Srebf1) was also up-regulated by Se. Both effects were confirmed in primary hepatocytes. In contrast to the overall Se-dependent increase of methylation capacity, the up-regulation of Srebf1 expression was paralleled by reduced local methylation of a specific CpG site within the Srebf1 gene. Thus, we provided evidence that Se-dependent effects on lipogenesis involve epigenetic mechanisms.


Assuntos
Carbono/metabolismo , Metilação de DNA/efeitos dos fármacos , Fígado/efeitos dos fármacos , Selênio/farmacologia , Animais , Glicina Hidroximetiltransferase/genética , Glicólise/efeitos dos fármacos , Glicólise/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Camundongos Endogâmicos C57BL , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Triglicerídeos/sangue , Regulação para Cima/efeitos dos fármacos
7.
Int J Mol Sci ; 18(5)2017 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-28475147

RESUMO

The pathogenesis of atherosclerosis has been partly acknowledged to result from aberrant epigenetic mechanisms. Accordingly, low folate levels are considered to be a contributing factor to promoting vascular disease because of deregulation of DNA methylation. We hypothesized that increasing the levels of folic acid may act via an epigenetic gene silencing mechanism to ameliorate atherosclerosis. Here, we investigated the atheroprotective effects of folic acid and the resultant methylation status in high-fat diet-fed ApoE knockout mice and in oxidized low-density lipoprotein-treated human umbilical vein endothelial cells. We analyzed atherosclerotic lesion histology, folate concentration, homocysteine concentration, S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH), and DNA methyltransferase activity, as well as monocyte chemotactic protein-1 (MCP1) and vascular endothelial growth factor (VEGF) expression and promoter methylation. Folic acid reduced atherosclerotic lesion size in ApoE knockout mice. The underlying folic acid protective mechanism appears to operate through regulating the normal homocysteine state, upregulating the SAM: SAH ratio, elevating DNA methyltransferase activity and expression, altering MCP1 and VEGF promoter methylation, and inhibiting MCP1 and VEGF expression. We conclude that folic acid supplementation effectively prevented atherosclerosis by modifying DNA methylation through the methionine cycle, improving DNA methyltransferase activity and expression, and thus changing the expression of atherosclerosis-related genes.


Assuntos
Quimiocina CCL2/genética , Metilação de DNA , Ácido Fólico/uso terapêutico , Placa Aterosclerótica/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/genética , Complexo Vitamínico B/uso terapêutico , Animais , Apolipoproteínas E/genética , Suplementos Nutricionais , Ácido Fólico/administração & dosagem , Ácido Fólico/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/prevenção & controle , Regiões Promotoras Genéticas , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Complexo Vitamínico B/administração & dosagem , Complexo Vitamínico B/farmacologia
8.
J Inherit Metab Dis ; 40(1): 5-20, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27671891

RESUMO

Inherited methylation disorders are a group of rarely reported, probably largely underdiagnosed disorders affecting transmethylation processes in the metabolic pathway between methionine and homocysteine. These are methionine adenosyltransferase I/III, glycine N-methyltransferase, S-adenosylhomocysteine hydrolase and adenosine kinase deficiencies. This paper provides the first consensus recommendations for the diagnosis and management of methylation disorders. Following search of the literature and evaluation according to the SIGN-methodology of all reported patients with methylation defects, graded recommendations are provided in a structured way comprising diagnosis (clinical presentation, biochemical abnormalities, differential diagnosis, newborn screening, prenatal diagnosis), therapy and follow-up. Methylation disorders predominantly affect the liver, central nervous system and muscles, but clinical presentation can vary considerably between and within disorders. Although isolated hypermethioninemia is the biochemical hallmark of this group of disorders, it is not always present, especially in early infancy. Plasma S-adenosylmethionine and S-adenosylhomocysteine are key metabolites for the biochemical clarification of isolated hypermethioninemia. Mild hyperhomocysteinemia can be present in all methylation disorders. Methylation disorders do not qualify as primary targets of newborn screening. A low-methionine diet can be beneficial in patients with methionine adenosyltransferase I/III deficiency if plasma methionine concentrations exceed 800 µmol/L. There is some evidence that this diet may also be beneficial in patients with S-adenosylhomocysteine hydrolase and adenosine kinase deficiencies. S-adenosylmethionine supplementation may be useful in patients with methionine adenosyltransferase I/III deficiency. Recommendations given in this article are based on general principles and in practice should be adjusted individually according to patient's age, severity of the disease, clinical and laboratory findings.


Assuntos
Homocisteína/metabolismo , Erros Inatos do Metabolismo/diagnóstico , Metionina/metabolismo , Consenso , Humanos , Recém-Nascido , Erros Inatos do Metabolismo/metabolismo , Metionina Adenosiltransferase/deficiência , Metilação , Triagem Neonatal/métodos , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
9.
Am J Clin Nutr ; 104(5): 1459-1469, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27707701

RESUMO

BACKGROUND: Moderately high folic acid intake in pregnant women has led to concerns about deleterious effects on the mother and fetus. Common polymorphisms in folate genes, such as methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase-formyltetrahydrofolate synthetase (MTHFD1) R653Q, may modulate the effects of elevated folic acid intake. OBJECTIVES: We investigated the effects of moderate folic acid supplementation on reproductive outcomes and assessed the potential interaction of the supplemented diet with MTHFD1-synthetase (Mthfd1S) deficiency in mice, which is a model for the R653Q variant. DESIGN: Female Mthfd1S+/+ and Mthfd1S+/- mice were fed a folic acid-supplemented diet (FASD) (5-fold higher than recommended) or control diets before mating and during pregnancy. Embryos and placentas were assessed for developmental defects at embryonic day 10.5 (E10.5). Maternal folate and choline metabolites and gene expression in folate-related pathways were examined. RESULTS: The combination of FASD and maternal MTHFD1-synthetase deficiency led to a greater incidence of defects in E10.5 embryos (diet × maternal genotype, P = 0.0016; diet × embryonic genotype, P = 0.054). The methylenetetrahydrofolate reductase (MTHFR) protein and methylation potential [ratio of S-adenosylmethionine (major methyl donor):S-adenosylhomocysteine) were reduced in maternal liver. Although 5-methyltetrahydrofolate (methylTHF) was higher in maternal circulation, the methylation potential was lower in embryos. The presence of developmental delays and defects in Mthfd1S+/- embryos was associated with placental defects (P = 0.003). The labyrinth layer failed to form properly in the majority of abnormal placentas, which compromised the integration of the maternal and fetal circulation and presumably the transfer of methylTHF and other nutrients. CONCLUSIONS: Moderately higher folate intake and MTHFD1-synthetase deficiency in pregnant mice result in a lower methylation potential in maternal liver and embryos and a greater incidence of defects in embryos. Although maternal circulating methylTHF was higher, it may not have reached the embryos because of abnormal placental development; abnormal placentas were observed predominantly in abnormally developed embryos. These findings have implications for women with high folate intakes, particularly if they are polymorphic for MTHFD1 R653Q.


Assuntos
Aminoidrolases/deficiência , Aminoidrolases/genética , Ácido Fólico/farmacologia , Formiato-Tetra-Hidrofolato Ligase/deficiência , Formiato-Tetra-Hidrofolato Ligase/genética , Metilenotetra-Hidrofolato Desidrogenase (NADP)/deficiência , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Complexos Multienzimáticos/deficiência , Complexos Multienzimáticos/genética , Placenta/anormalidades , Placenta/enzimologia , Polimorfismo de Nucleotídeo Único , Aminoidrolases/metabolismo , Animais , Colina/farmacologia , Suplementos Nutricionais , Embrião de Mamíferos/enzimologia , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Formiato-Tetra-Hidrofolato Ligase/metabolismo , Modelos Logísticos , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Camundongos , Camundongos Transgênicos , Complexos Multienzimáticos/metabolismo , Gravidez , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
10.
Alcohol Clin Exp Res ; 40(11): 2312-2319, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27581622

RESUMO

BACKGROUND: Alcohol-induced reduction in the hepatocellular S-adenosylmethionine (SAM):S-adenosylhomocysteine (SAH) ratio impairs the activities of many SAM-dependent methyltransferases. These impairments ultimately lead to the generation of several hallmark features of alcoholic liver injury including steatosis. Guanidinoacetate methyltransferase (GAMT) is an important enzyme that catalyzes the final reaction in the creatine biosynthetic process. The liver is a major site for creatine synthesis which places a substantial methylation burden on this organ as GAMT-mediated reactions consume as much as 40% of all the SAM-derived methyl groups. We hypothesized that dietary creatine supplementation could potentially spare SAM, preserve the hepatocellular SAM:SAH ratio, and thereby prevent the development of alcoholic steatosis and other consequences of impaired methylation reactions. METHODS: For these studies, male Wistar rats were pair-fed the Lieber-DeCarli control or ethanol (EtOH) diet with or without 1% creatine supplementation. At the end of 4 to 5 weeks of feeding, relevant biochemical and histological analyses were performed. RESULTS: We observed that creatine supplementation neither prevented alcoholic steatosis nor attenuated the alcohol-induced impairments in proteasome activity. The lower hepatocellular SAM:SAH ratio seen in the EtOH-fed rats was also not normalized or SAM levels spared when these rats were fed the creatine-supplemented EtOH diet. However, a >10-fold increased level of creatine was observed in the liver, serum, and hearts of rats fed the creatine-supplemented diets. CONCLUSIONS: Overall, dietary creatine supplementation did not prevent alcoholic liver injury despite its known efficacy in preventing high-fat-diet-induced steatosis. Betaine, a promethylating agent that maintains the hepatocellular SAM:SAH, still remains our best option for treating alcoholic steatosis.


Assuntos
Creatina/uso terapêutico , Fígado Gorduroso Alcoólico/prevenção & controle , Amidinotransferases/metabolismo , Animais , Suplementos Nutricionais , Guanidinoacetato N-Metiltransferase/metabolismo , Rim/enzimologia , Fígado/enzimologia , Masculino , Miocárdio/metabolismo , Ratos Wistar , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
11.
Metab Syndr Relat Disord ; 14(4): 202-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26881897

RESUMO

BACKGROUND: Previous studies indicated that nonpurified and purified commercially available control murine diets have different metabolic effects with potential consequences on hepatic methionine metabolism and liver histology. METHODS: We compared the metabolic and histological effects of commercial nonpurified (13% calories from fat; 57% calories from carbohydrates with 38 grams/kg of sucrose) and purified control diets (12% calories from fat; 69% calories from carbohydrates with ∼500 grams/kg of sucrose) with or without choline supplementation administered to C3H mice with normal lipid and methionine metabolism. Diets were started 2 weeks before mating, continued through pregnancy and lactation, and continued in offspring until 24 weeks of age when we collected plasma and liver tissue to study methionine and lipid metabolism. RESULTS: Compared to mice fed nonpurified diets, the liver/body weight ratio was significantly higher in mice fed either purified diet, which was associated with hepatic steatosis and inflammation. Plasma alanine aminotransferase levels were higher in mice receiving the purified diets. The hepatic S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH) ratio was higher in female mice fed purified compared to nonpurified diet (4.6 ± 2 vs. 2.8 ± 1.9; P < 0.05). Choline supplementation was associated with improvement of some parameters of lipid and methionine metabolism in mice fed purified diets. CONCLUSIONS: Standard nonpurified and purified diets have significantly different effects on development of steatosis in control mice. These findings can help in development of animal models of fatty liver and in choosing appropriate laboratory control diets for control animals.


Assuntos
Colina/metabolismo , Fígado Gorduroso/metabolismo , Metabolismo dos Lipídeos , Metionina/metabolismo , Animais , Dieta , Gorduras na Dieta/administração & dosagem , Sacarose Alimentar/administração & dosagem , Suplementos Nutricionais , Comportamento Alimentar , Feminino , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C3H , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
12.
Int J Biochem Cell Biol ; 67: 158-66, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26117455

RESUMO

Transmethylation reactions utilize S-adenosylmethionine (SAM) as a methyl donor and are central to the regulation of many biological processes: more than fifty SAM-dependent methyltransferases methylate a broad spectrum of cellular compounds including DNA, histones, phospholipids and other small molecules. Common to all SAM-dependent transmethylation reactions is the release of the potent inhibitor S-adenosylhomocysteine (SAH) as a by-product. SAH is reversibly hydrolyzed to adenosine and homocysteine by SAH hydrolase. Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. However, a major unanswered question is if homocysteine is causally involved in disease pathogenesis or simply a passive and indirect indicator of a more complex mechanism. A chronic elevation in homocysteine levels results in a parallel increase in intracellular or plasma SAH, which is a more sensitive biomarker of cardiovascular disease than homocysteine and suggests that SAH is a critical pathological factor in homocysteine-associated disorders. Previous reports indicate that supplementation with folate and B vitamins efficiently lowers homocysteine levels but not plasma SAH levels, which possibly explains the failure of homocysteine-lowering vitamins to reduce vascular events in several recent clinical intervention studies. Furthermore, more studies are focusing on the role and mechanisms of SAH in different chronic diseases related to hyperhomocysteinemia, such as cardiovascular disease, kidney disease, diabetes, and obesity. This review summarizes the current role of SAH in cardiovascular disease and its effect on several related risk factors. It also explores possible the mechanisms, such as epigenetics and oxidative stress, of SAH. This article is part of a Directed Issue entitled: Epigenetic dynamics in development and disease.


Assuntos
Aterosclerose/metabolismo , Epigênese Genética , S-Adenosil-Homocisteína/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Estresse Oxidativo , S-Adenosilmetionina/metabolismo
13.
Nutrients ; 7(2): 1108-18, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25668155

RESUMO

Elevated circulating total homocysteine (tHcy) concentrations (hyperhomocysteinemia) have been regarded as an independent risk factor for cardiovascular disease (CVD). However, several large clinical trials to correct hyperhomocysteinemia using B-vitamin supplements (particularly folic acid) have largely failed to reduce the risk of CVD. There is no doubt that a large segment of patients with CVD have hyperhomocysteinemia; therefore, it is reasonable to postulate that circulating tHcy concentrations are in part a surrogate marker for another, yet-to-be-identified risk factor(s) for CVD. We found that iron catalyzes the formation of Hcy from methionine, S-adenosylhomocysteine and cystathionine. Based on these findings, we propose that an elevated amount of non-protein-bound iron (free Fe) increases circulating tHcy. Free Fe catalyzes the formation of oxygen free radicals, and oxidized low-density lipoprotein is a well-established risk factor for vascular damage. In this review, we discuss our findings on iron-catalyzed formation of Hcy from thioethers as well as recent findings by other investigators on this issue. Collectively, these support our hypothesis that circulating tHcy is in part a surrogate marker for free Fe, which is one of the independent risk factors for CVD.


Assuntos
Biomarcadores/sangue , Doenças Cardiovasculares/sangue , Homocisteína/metabolismo , Ferro/sangue , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Cistationina/metabolismo , Suplementos Nutricionais , Ácido Fólico/farmacologia , Radicais Livres/metabolismo , Humanos , Hiper-Homocisteinemia/dietoterapia , Hiper-Homocisteinemia/metabolismo , Ferro/metabolismo , Lipoproteínas LDL/metabolismo , Metionina/metabolismo , Fatores de Risco , S-Adenosil-Homocisteína/metabolismo , Complexo Vitamínico B/farmacologia
14.
Amino Acids ; 47(4): 839-46, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25577261

RESUMO

The purpose of this study was to examine the effects of betaine supplementation on the regulation of one-carbon metabolism and liver lipid accumulation induced by a high-fat diet in rats. Rats were fed one of three different liquid diets: control diet, high-fat diet and high-fat diet supplemented with betaine. The control and high-fat liquid diets contained, respectively, 35 and 71 % of energy derived from fat. Betaine supplementation involved the addition of 1 % (g/L) to the diet. After three weeks on the high-fat diet the rats had increased total liver fat concentration, liver triglycerides, liver TBARS and plasma TNF-α. The high-fat diet decreased the hepatic S-adenosylmethionine concentration and the S-adenosylmethionine/S-adenosylhomocysteine ratio compared to the control as well as altering the expression of genes involved in one-carbon metabolism. Betaine supplementation substantially increased the hepatic S-adenosylmethionine concentration (~fourfold) and prevented fatty liver and hepatic injury induced by the high-fat diet. It was accompanied by the normalization of the gene expression of BHMT, GNMT and MGAT, which code for key enzymes of one-carbon metabolism related to liver fat accumulation. In conclusion, the regulation of the expression of MGAT by betaine supplementation provides an additional and novel mechanism by which betaine supplementation regulates lipid metabolism and prevents accumulation of fat in the liver.


Assuntos
Betaína/administração & dosagem , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais/análise , Fígado Gorduroso/tratamento farmacológico , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Aciltransferases/genética , Aciltransferases/metabolismo , Animais , Carbono/metabolismo , Fígado Gorduroso/etiologia , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Glicina N-Metiltransferase/genética , Glicina N-Metiltransferase/metabolismo , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Triglicerídeos/metabolismo
15.
Eur Rev Med Pharmacol Sci ; 19(1): 154-60, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25635989

RESUMO

OBJECTIVE: The aim of this study was to evaluate plasma homocysteine (Hcy), malondialdehyde (MDA), glutathione (GSH) levels, glutathione peroxidase (GSH-Px) and glutathione-S-transferase (GST) activities and liver tissue S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) levels in control and vitamin B6-deficient rats. MATERIALS AND METHODS: Thirty-two male rats with a weight of 65-75 g were used for the experiment. The rats were divided into control (n=16) and vitamin B6-deficient groups. At the end of the experiment, the animals were anesthetized with ketamine-HCl (Ketalar, 20 mg/kg, i.p.), and the blood was collected by cardiac puncture after thoracotomy. Plasma Hcy, pyridoxal phosphate (PLP), liver SAM, SAH levels measured by an isocratic system with high performance liquid chromatography. Plasma GSH-Px, GSH activities and GSH, MDA levels were carried out using a spectrophotometer. RESULTS: Plasma Hcy, MDA, liver tissue SAH levels were significantly increased, whereas plasma GSH, PLP, liver tissue SAM levels, plasma GST, GSH-Px activities and SAM/SAH ratio were decreased compared to those of control group. CONCLUSIONS: Vitamin B6 deficiency causes an increase in plasma homocysteine levels. Thus, we think that vitamin B6 supplementation could be used for therapeutic purposes in hyperhomocysteinemia condition.


Assuntos
Homocisteína/sangue , Fígado/metabolismo , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Deficiência de Vitamina B 6/metabolismo , Animais , Glutationa Peroxidase/sangue , Glutationa Transferase/sangue , Masculino , Malondialdeído/sangue , Ratos , Ratos Sprague-Dawley , Deficiência de Vitamina B 6/sangue
16.
Sleep ; 38(2): 305-13, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25515097

RESUMO

STUDY OBJECTIVES: Gamma-hydroxybutyrate (GHB) was originally introduced as an anesthetic but was first abused by bodybuilders and then became a recreational or club drug.1 Sodium salt of GHB is currently used for the treatment of cataplexy in patients with narcolepsy. The mode of action and metabolism of GHB is not well understood. GHB stimulates growth hormone release in humans and induces weight loss in treated patients, suggesting an unexplored metabolic effect. In different experiments the effect of GHB administration on central (cerebral cortex) and peripheral (liver) biochemical processes involved in the metabolism of the drug, as well as the effects of the drug on metabolism, were evaluated in mice. DESIGN: C57BL/6J, gamma-aminobutyric acid B (GABAB) knockout and obese (ob/ob) mice were acutely or chronically treated with GHB at 300 mg/kg. MEASUREMENTS AND RESULTS: Respiratory ratio decreased under GHB treatment, independent of food intake, suggesting a shift in energy substrate from carbohydrates to lipids. GHB-treated C57BL/6J and GABAB null mice but not ob/ob mice gained less weight than matched controls. GHB dramatically increased the corticosterone level but did not affect growth hormone or prolactin. Metabolome profiling showed that an acute high dose of GHB did not increase the brain GABA level. In the brain and the liver, GHB was metabolized into succinic semialdehyde by hydroxyacid-oxoacid transhydrogenase. Chronic administration decreased glutamate, s-adenosylhomocysteine, and oxidized gluthathione, and increased omega-3 fatty acids. CONCLUSIONS: Our findings indicate large central and peripheral metabolic changes induced by GHB with important relevance to its therapeutic use.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Hidroxibutiratos/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Oxirredutases do Álcool/metabolismo , Animais , Composição Corporal/efeitos dos fármacos , Respiração Celular/efeitos dos fármacos , Corticosterona/metabolismo , Ácidos Graxos Ômega-3/metabolismo , Ácido Glutâmico/metabolismo , Dissulfeto de Glutationa/metabolismo , Hormônio do Crescimento/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Proteínas Mitocondriais/metabolismo , Oxirredução , Prolactina/metabolismo , S-Adenosil-Homocisteína/metabolismo , Aumento de Peso/efeitos dos fármacos , Ácido gama-Aminobutírico/análogos & derivados , Ácido gama-Aminobutírico/deficiência , Ácido gama-Aminobutírico/metabolismo
17.
Appl Physiol Nutr Metab ; 40(1): 46-50, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25485892

RESUMO

The purpose of this study was to examine the effects of whey protein supplementation on homocysteine (Hcy) metabolism and liver oxidative stress in rats. Twenty-four rats were divided into 3 groups (n = 8) to receive one of the following diets for 4 weeks: control diet (C), whey protein-composed diet (WP), and whey protein-supplemented diet (WPS). The C and WP diets consisted of AIN-93 with 20% casein and 20% whey protein as protein source, respectively. WPS was AIN-93 (20% casein) supplemented by the addition of 20% (w/w) whey protein. Four weeks of ingesting a WPS diet resulted in a significantly higher (P < 0.05) total protein and methionine intakes. Although a significant increase (P < 0.05) in the hepatic S-adenosylmethionine and S-adenosylhomocysteine levels occurred in WPS group compared with C and WP, no significant change was observed in plasma Hcy concentration between groups. Furthermore, the levels of lipid hydroperoxides and advanced oxidation protein products, known liver oxidative stress markers, were increased in the WPS group compared with the C group. In addition, no change in glutathione liver concentration was observed in any of the groups studied. In conclusion, whey protein supplementation increases methionine intake substantially; however, it does not change plasma Hcy concentrations. On the other hand, increased hepatic oxidative stress markers were observed in whey protein supplemented rats were probably due to high protein intake.


Assuntos
Suplementos Nutricionais/efeitos adversos , Hiper-Homocisteinemia/prevenção & controle , Fígado/metabolismo , Metionina/administração & dosagem , Estresse Oxidativo , Proteínas do Soro do Leite/efeitos adversos , Produtos da Oxidação Avançada de Proteínas/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Caseínas/efeitos adversos , Glutationa/metabolismo , Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/etiologia , Hiper-Homocisteinemia/metabolismo , Peroxidação de Lipídeos , Masculino , Metionina/efeitos adversos , Metionina/sangue , Metionina/metabolismo , Músculo Esquelético/metabolismo , Oxirredução , Distribuição Aleatória , Ratos Wistar , S-Adenosil-Homocisteína/agonistas , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/agonistas , S-Adenosilmetionina/metabolismo , Proteínas do Soro do Leite/administração & dosagem
18.
Exp Mol Pathol ; 97(1): 49-56, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24842317

RESUMO

We previously reported that chronic ethanol intake lowers hepatocellular S-adenosylmethionine to S-adenosylhomocysteine ratio and significantly impairs many liver methylation reactions. One such reaction, catalyzed by guanidinoacetate methyltransferase (GAMT), is a major consumer of methyl groups and utilizes as much as 40% of the SAM-derived groups to convert guanidinoacetate (GAA) to creatine. The exposure to methyl-group consuming compounds has substantially increased over the past decade that puts additional stresses on the cellular methylation potential. The purpose of our study was to investigate whether increased ingestion of a methyl-group consumer (GAA) either alone or combined with ethanol intake, plays a role in the pathogenesis of liver injury. Adult male Wistar rats were pair-fed the Lieber DeCarli control or ethanol diet in the presence or absence of GAA for 2weeks. At the end of the feeding regimen, biochemical and histological analyses were conducted. We observed that 2 weeks of GAA- or ethanol-alone treatment increases hepatic triglyceride accumulation by 4.5 and 7-fold, respectively as compared with the pair-fed controls. However, supplementing GAA in the ethanol diet produced panlobular macro- and micro-vesicular steatosis, a marked decrease in the methylation potential and a 28-fold increased triglyceride accumulation. These GAA-supplemented ethanol diet-fed rats displayed inflammatory changes and significantly increased liver toxicity compared to the other groups. In conclusion, increased methylation demand superimposed on chronic ethanol consumption causes more pronounced liver injury. Thus, alcoholic patients should be cautioned for increased dietary intake of methyl-group consuming compounds even for a short period of time.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Etanol/toxicidade , Glicina/análogos & derivados , Fígado/efeitos dos fármacos , Metilação/efeitos dos fármacos , Consumo de Bebidas Alcoólicas/metabolismo , Amidinotransferases/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/patologia , Dieta , Fígado Gorduroso Alcoólico/metabolismo , Glicina/farmacologia , Guanidinoacetato N-Metiltransferase/metabolismo , Homocisteína/sangue , Fígado/metabolismo , Fígado/patologia , Masculino , Ratos , Ratos Wistar , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Triglicerídeos/metabolismo
19.
Alcohol Clin Exp Res ; 38(6): 1540-9, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24730561

RESUMO

BACKGROUND: Alcoholic steatohepatitis (ASH) is caused in part by the effects of ethanol (EtOH) on hepatic methionine metabolism. METHODS: To investigate the phenotypic and epigenetic consequences of altered methionine metabolism in this disease, we studied the effects of 4-week intragastric EtOH feeding with and without the methyl donor betaine in cystathionine beta synthase (CßS) heterozygous C57BL/6J mice. RESULTS: The histopathology of early ASH was induced by EtOH feeding and prevented by betaine supplementation, while EtOH feeding reduced and betaine supplementation maintained the hepatic methylation ratio of the universal methyl donor S-adenosylmethionine (SAM) to the methyltransferase inhibitor S-adenosylhomocysteine (SAH). MethylC-seq genomic sequencing of heterozygous liver samples from each diet group found 2 to 4% reduced methylation in gene bodies, but not promoter regions of all autosomes of EtOH-fed mice, each of which were normalized in samples from mice fed the betaine-supplemented diet. The transcript levels of nitric oxide synthase (Nos2) and DNA methyltransferase 1 (Dnmt1) were increased, while those of peroxisome proliferator receptor-α (Pparα) were reduced in EtOH-fed mice, and each was normalized in mice fed the betaine-supplemented diet. DNA pyrosequencing of CßS heterozygous samples found reduced methylation in a gene body of Nos2 by EtOH feeding that was restored by betaine supplementation and was correlated inversely with its expression and positively with SAM/SAH ratios. CONCLUSIONS: The present study has demonstrated relationships among EtOH induction of ASH with aberrant methionine metabolism that was associated with gene body DNA hypomethylation in all autosomes and was prevented by betaine supplementation. The data imply that EtOH-induced changes in selected gene transcript levels and hypomethylation in gene bodies during the induction of ASH are a result of altered methionine metabolism that can be reversed through dietary supplementation of methyl donors.


Assuntos
Betaína/uso terapêutico , Metilação de DNA/efeitos dos fármacos , Etanol/farmacologia , Fígado Gorduroso Alcoólico/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Homocistinúria/metabolismo , Animais , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/análise , Suplementos Nutricionais , Fígado/química , Fígado/efeitos dos fármacos , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/análise , PPAR alfa/análise , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
20.
J Biol Chem ; 289(22): 15350-62, 2014 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-24719327

RESUMO

S-adenosylhomocysteine (SAH) is a negative regulator of most methyltransferases and the precursor for the cardiovascular risk factor homocysteine. We have previously identified a link between the homocysteine-induced suppression of the selenoprotein glutathione peroxidase 1 (GPx-1) and endothelial dysfunction. Here we demonstrate a specific mechanism by which hypomethylation, promoted by the accumulation of the homocysteine precursor SAH, suppresses GPx-1 expression and leads to inflammatory activation of endothelial cells. The expression of GPx-1 and a subset of other selenoproteins is dependent on the methylation of the tRNA(Sec) to the Um34 form. The formation of methylated tRNA(Sec) facilitates translational incorporation of selenocysteine at a UGA codon. Our findings demonstrate that SAH accumulation in endothelial cells suppresses the expression of GPx-1 to promote oxidative stress. Hypomethylation stress, caused by SAH accumulation, inhibits the formation of the methylated isoform of the tRNA(Sec) and reduces GPx-1 expression. In contrast, under these conditions, the expression and activity of thioredoxin reductase 1, another selenoprotein, is increased. Furthermore, SAH-induced oxidative stress creates a proinflammatory activation of endothelial cells characterized by up-regulation of adhesion molecules and an augmented capacity to bind leukocytes. Taken together, these data suggest that SAH accumulation in endothelial cells can induce tRNA(Sec) hypomethylation, which alters the expression of selenoproteins such as GPx-1 to contribute to a proatherogenic endothelial phenotype.


Assuntos
Células Endoteliais/enzimologia , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Metiltransferases/metabolismo , Aminoacil-RNA de Transferência/metabolismo , S-Adenosil-Homocisteína/metabolismo , Adesão Celular/fisiologia , Células Endoteliais/efeitos dos fármacos , Homocisteína/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Peróxido de Hidrogênio/metabolismo , Leucócitos/citologia , Metilação , Estresse Oxidativo/fisiologia , RNA de Transferência de Serina/metabolismo , S-Adenosilmetionina/metabolismo , Selênio/farmacologia , Selenoproteínas/metabolismo , Glutationa Peroxidase GPX1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA