Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Adicionar filtros








Intervalo de ano
1.
Biomolecules & Therapeutics ; : 167-174, 2018.
Artigo em Inglês | WPRIM | ID: wpr-713579

RESUMO

Alterations in sulfur amino acid metabolism are associated with an increased risk of a number of common late-life diseases, which raises the possibility that metabolism of sulfur amino acids may change with age. The present study was conducted to understand the age-related changes in hepatic metabolism of sulfur amino acids in 2-, 6-, 18- and 30-month-old male C57BL/6 mice. For this purpose, metabolite profiling of sulfur amino acids from methionine to taurine or glutathione (GSH) was performed. The levels of sulfur amino acids and their metabolites were not significantly different among 2-, 6- and 18-month-old mice, except for plasma GSH and hepatic homocysteine. Plasma total GSH and hepatic total homocysteine levels were significantly higher in 2-month-old mice than those in the other age groups. In contrast, 30-month-old mice exhibited increased hepatic methionine and cysteine, compared with all other groups, but decreased hepatic S-adenosylmethionine (SAM), S-adenosylhomocysteine and homocysteine, relative to 2-month-old mice. No differences in hepatic reduced GSH, GSH disulfide, or taurine were observed. The hepatic changes in homocysteine and cysteine may be attributed to upregulation of cystathionine β-synthase and down-regulation of γ-glutamylcysteine ligase in the aged mice. The elevation of hepatic cysteine levels may be involved in the maintenance of hepatic GSH levels. The opposite changes of methionine and SAM suggest that the regulatory role of SAM in hepatic sulfur amino acid metabolism may be impaired in 30-month-old mice.


Assuntos
Animais , Pré-Escolar , Humanos , Lactente , Masculino , Camundongos , Envelhecimento , Aminoácidos Sulfúricos , Cistationina , Cisteína , Regulação para Baixo , Glutationa , Homocisteína , Metabolismo , Metabolômica , Metionina , Plasma , S-Adenosil-Homocisteína , S-Adenosilmetionina , Enxofre , Taurina , Regulação para Cima
2.
The Korean Journal of Physiology and Pharmacology ; : 433-440, 2016.
Artigo em Inglês | WPRIM | ID: wpr-728688

RESUMO

Inositol-1,4,5-triphosphate [IP3] receptors binding protein released with IP3 (IRBIT) was previously reported as an activator of NBCe1-B. Recent studies have characterized IRBIT homologue S-Adenosylhomocysteine hydrolase-like 2 (AHCYL2). AHCYL2 is highly homologous to IRBIT (88%) and heteromerizes with IRBIT. The two important domains in the N-terminus of AHCYL2 are a PEST domain and a coiled-coil domain which are highly comparable to those in IRBIT. Therefore, in this study, we tried to identify the role of those domains in mouse AHCYL2 (Ahcyl2), and we succeeded in identifying PEST domain of Ahcyl2 as a regulation region for NBCe1-B activity. Site directed mutagenesis and coimmunoprecipitation assay showed that NBCe1-B binds to the N-terminal Ahcyl2-PEST domain, and its binding is determined by the phosphorylation of 4 critical serine residues (Ser151, Ser154, Ser157, and Ser160) in Ahcyl2 PEST domain. Also we revealed that 4 critical serine residues in Ahcyl2 PEST domain are indispensable for the activation of NBCe1-B using measurement of intracellular pH experiment. Thus, these results suggested that the NBCe1-B is interacted with 4 critical serine residues in Ahcyl2 PEST domain, which play an important role in intracellular pH regulation through NBCe1-B.


Assuntos
Animais , Camundongos , Proteínas de Transporte , Concentração de Íons de Hidrogênio , Mutagênese Sítio-Dirigida , Fosforilação , S-Adenosil-Homocisteína , Serina
3.
Nutrition Research and Practice ; : 144-149, 2015.
Artigo em Inglês | WPRIM | ID: wpr-143314

RESUMO

BACKGROUND/OBJECTIVE: The aim of this study was to examine the effect of high dietary methionine (Met) consumption on plasma and hepatic oxidative stress and dyslipidemia in chronic ethanol fed rats. MATERIALS/METHODS: Male Wistar rats were fed control or ethanol-containing liquid diets supplemented without (E group) or with DL-Met at 0.6% (EM1 group) or 0.8% (EM2 group) for five weeks. Plasma aminothiols, lipids, malondialdehyde (MDA), alanine aminotransferase (ALT), and aspartate aminotransferase were measured. Hepatic folate, S-adenosylmethionine (SAM), and S-adenosylhomocysteine (SAH) were measured. RESULTS: DL-Met supplementation was found to increase plasma levels of homocysteine (Hcy), triglyceride (TG), total cholesterol (TC), and MDA compared to rats fed ethanol alone and decrease plasma ALT. However, DL-Met supplementation did not significantly change plasma levels of HDL-cholesterol, cysteine, cysteinylglycine, and glutathione. In addition, DL-Met supplementation increased hepatic levels of folate, SAM, SAH, and SAM:SAH ratio. Our data showed that DL-Met supplementation can increase plasma oxidative stress and atherogenic effects by elevating plasma Hcy, TG, and TC in ethanol-fed rats. CONCLUSION: The present results demonstrate that Met supplementation increases plasma oxidative stress and atherogenic effects by inducing dyslipidemia and hyperhomocysteinemia in ethanol-fed rats.


Assuntos
Animais , Humanos , Masculino , Ratos , Alanina Transaminase , Aspartato Aminotransferases , Colesterol , Cisteína , Dieta , Dislipidemias , Etanol , Ácido Fólico , Glutationa , Homocisteína , Hiper-Homocisteinemia , Malondialdeído , Metionina , Estresse Oxidativo , Plasma , Ratos Wistar , S-Adenosil-Homocisteína , S-Adenosilmetionina , Triglicerídeos
4.
Nutrition Research and Practice ; : 144-149, 2015.
Artigo em Inglês | WPRIM | ID: wpr-143307

RESUMO

BACKGROUND/OBJECTIVE: The aim of this study was to examine the effect of high dietary methionine (Met) consumption on plasma and hepatic oxidative stress and dyslipidemia in chronic ethanol fed rats. MATERIALS/METHODS: Male Wistar rats were fed control or ethanol-containing liquid diets supplemented without (E group) or with DL-Met at 0.6% (EM1 group) or 0.8% (EM2 group) for five weeks. Plasma aminothiols, lipids, malondialdehyde (MDA), alanine aminotransferase (ALT), and aspartate aminotransferase were measured. Hepatic folate, S-adenosylmethionine (SAM), and S-adenosylhomocysteine (SAH) were measured. RESULTS: DL-Met supplementation was found to increase plasma levels of homocysteine (Hcy), triglyceride (TG), total cholesterol (TC), and MDA compared to rats fed ethanol alone and decrease plasma ALT. However, DL-Met supplementation did not significantly change plasma levels of HDL-cholesterol, cysteine, cysteinylglycine, and glutathione. In addition, DL-Met supplementation increased hepatic levels of folate, SAM, SAH, and SAM:SAH ratio. Our data showed that DL-Met supplementation can increase plasma oxidative stress and atherogenic effects by elevating plasma Hcy, TG, and TC in ethanol-fed rats. CONCLUSION: The present results demonstrate that Met supplementation increases plasma oxidative stress and atherogenic effects by inducing dyslipidemia and hyperhomocysteinemia in ethanol-fed rats.


Assuntos
Animais , Humanos , Masculino , Ratos , Alanina Transaminase , Aspartato Aminotransferases , Colesterol , Cisteína , Dieta , Dislipidemias , Etanol , Ácido Fólico , Glutationa , Homocisteína , Hiper-Homocisteinemia , Malondialdeído , Metionina , Estresse Oxidativo , Plasma , Ratos Wistar , S-Adenosil-Homocisteína , S-Adenosilmetionina , Triglicerídeos
5.
Chinese Journal of Stomatology ; (12): 210-213, 2011.
Artigo em Chinês | WPRIM | ID: wpr-339771

RESUMO

<p><b>OBJECTIVE</b>To investigate the function of luxS in sulfurmetabolism of Streptococcus mutans (Sm).</p><p><b>METHODS</b>The growth with absorbency (A) of the standards and mutant strains was measured and analyzed in the sulfur-limited defined medium at different periods. The laser scanning confocal microscopy (LSCM) was used to observe and compare the biofilm thickness of the two kinds of strains at different culture conditions.</p><p><b>RESULTS</b>The significant increases in the thickness of mutant strain biofilm and its growth were observed after the addition of cysteine, but did not reach the standards strain levels (P < 0.05). The growth and the biofilm thickness of the mutant strains were (1.301 ± 0.009) and (45.009 ± 0.429) µm. When methionine and S-adenosylhomocysteine of certain concentrations were respectively added, the biofilm thickness and the growth of mutant strain were raised but did not reach the level of the standards strain at 24 h (P < 0.05), but at 48 h they did. When the methionine was added in the mutant strains for 24 h, the biofilm thickness and the growth of mutant strain were (0.448 ± 0.028) and (37.068 ± 2.392) µm, as for the adding of S-adenosylhomocysteine were (0.460 ± 0.005) and (27.343 ± 1.107) µm. When adding the supernatant fluid of standard strains, the biofilm thickness and the growth levels of mutant strain were much higher than those of the standards strain. The biofilm thickness and growth of both kinds of strains decreased after the addition of S-adenosylmethionine.</p><p><b>CONCLUSIONS</b>luxS gene plays not only a role in quorum sensing but also a role in sulfurmetabolism.</p>


Assuntos
Proteínas de Bactérias , Genética , Metabolismo , Biofilmes , Liases de Carbono-Enxofre , Genética , Metabolismo , Meios de Cultura , Técnicas de Cultura , Cisteína , Metabolismo , Regulação Bacteriana da Expressão Gênica , Metionina , Metabolismo , Microscopia Confocal , Percepção de Quorum , S-Adenosil-Homocisteína , Metabolismo , S-Adenosilmetionina , Metabolismo , Streptococcus mutans , Genética , Metabolismo , Enxofre , Metabolismo
6.
The Korean Journal of Nutrition ; : 423-433, 2009.
Artigo em Coreano | WPRIM | ID: wpr-649786

RESUMO

Folate and vitamin B12 are essential cofactors for homocysteine (Hcy) metabolism. Homocysteinemia has been related with cardiovascular and neurodegenerative disease. We examined the effect of folate and/or vitamin B12 deficiency on biomarkers of one carbon metabolism in blood, liver and brain, and analyzed the correlation between vitamin biomarkers in mild and moderate homocysteinemia. In this study, Sprague-Dawley male rats (5 groups, n = 10) were fed folate-sufficient diet (FS), folate-deficient diet (FD) with 0 or 3 g homocystine (FSH and FDH), and folate-/vitamin B12-deficient diet with 3 g homocystine (FDHCD) for 8 weeks. The FDH diet induced mild homocysteinemia (plasma Hcy 17.41 +/- 1.94 nmol/mL) and the FDHCD diet induced moderate homocysteinemia (plasma Hcy 44.13 +/- 2.65 nmol/mL), respectively. Although liver and brain folate levels were significantly lower compared with those values of rats fed FS or FSH (p < 0.001, p < 0.01 respectively), there were no significant differences in folate levels in liver and brain among the rats fed FD, FDH and FDHCD diet. However, rats fed FDHCD showed higher plasma folate levels (126.5 +/- 9.6 nmol/L) compared with rats fed FD and FDH (21.1 +/- 1.4 nmol/L, 22.0 +/- 2.2 nmol/L)(p < 0.001), which is the feature of "ethyl-folate trap"by vitamin B12 deficiency. Plasma Hcy was correlated with hepatic folate (r = -0.641, p < 0.01) but not with plasma folate or brain folate in this experimental condition. However, as we eliminated FDHCD group during correlation test, plasma Hcy was correlated with plasma folate (r = -0.581, p < 0.01), hepatic folate (r = -0.684, p < 0.01) and brain folate (r = -0.321, p < 0.05). Hepatic S-adenosylmethionine (SAM) level was lower in rats fed FD, FDH and FDHCD than in rats fed FS and FSH (p < 0.001, p < 0.001 respectively) and hepatic S-adenosylhomocysteine (SAH) level was significantly higher in those groups. The SAH level in brain was also significantly increased in rats fed FDHCD (p < 0.05). However, brain SAM level was not affected by folate and/or vitamin B12 deficiency. This result suggests that dietary folate- and vitamin B12-deficiency may inhibit methylation in brain by increasing SAH rather than decreasing SAM level, which may be closely associated with impaired cognitive function in nutritional homocysteinemia.


Assuntos
Animais , Humanos , Masculino , Ratos , Biomarcadores , Encéfalo , Carbono , Dieta , Metilação de DNA , Ácido Fólico , Homocisteína , Homocistina , Hiper-Homocisteinemia , Fígado , Metilação , Doenças Neurodegenerativas , Plasma , S-Adenosil-Homocisteína , S-Adenosilmetionina , Vitamina B 12 , Deficiência de Vitamina B 12 , Vitaminas
7.
Nutrition Research and Practice ; : 122-127, 2009.
Artigo em Inglês | WPRIM | ID: wpr-80360

RESUMO

Folate is generally considered as a safe water-soluble vitamin for supplementation. However, we do not have enough information to confirm the potential effects and safety of folate supplementation and the interaction with vitamin B12 deficiency. It has been hypothesized that a greater methyl group supply could lead to compensation for vitamin B12 deficiency. On this basis, the present study was conducted to examine the effects of high-dose folic acid (FA) supplementation on biomarkers involved in the methionine cycle in vitamin B12-deficient rats. Sprague-Dawley rats were fed diets containing either 0 or 100 microg (daily dietary requirement) vitamin B12/kg diet with either 2 mg (daily dietary requirement) or 100 mg FA/kg diet for six weeks. Vitamin B12-deficiency resulted in increased plasma homocysteine (p<0.01), which was normalized by dietary supplementation of high-dose FA (p<0.01). However, FA supplementation and vitamin B12 deficiency did not alter hepatic and brain S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) concentrations and hepatic DNA methylation. These results indicated that supplementation of high-dose FA improved homocysteinemia in vitamin B12-deficiency but did not change SAM and SAH, the main biomarkers of methylating reaction.


Assuntos
Animais , Ratos , Biomarcadores , Encéfalo , Compensação e Reparação , Dieta , Suplementos Nutricionais , Metilação de DNA , Ácido Fólico , Homocisteína , Hiper-Homocisteinemia , Metionina , Plasma , Ratos Sprague-Dawley , S-Adenosil-Homocisteína , S-Adenosilmetionina , Deficiência de Vitamina B 12 , Vitaminas
8.
The Korean Journal of Nutrition ; : 14-23, 2007.
Artigo em Coreano | WPRIM | ID: wpr-653099

RESUMO

Elevated plasma homocysteine ( Hcy) is a risk factor for cognitive dysfunction and Alzheimer disease, although the mechanism is still unknown. Both folate and betaine, a choline metabolite, play essential roles in the remethylation of Hcy to methionine. Choline deficiency may be associated with low folate status and high plasma Hcy. Alterations in DNA methylation also have established critical roles for methylation in development of the nervous system. This study was un-dertaken to assess the effect of choline and folate deficiency on Hcy metabolism and genomic DNA methylation status of the liver and brain. Groups of adult male Sprague Dawley rats were fed on a control, choline-deficient ( CD) , folate-deficient ( FD) or choline/folate-deficient ( CFD) diets for 8 weeks. FD resulted in a significantly lower hepatic folate ( 23%)(p < 0.001) and brain folate ( 69%)(p < 0.05) compared to the control group. However, plasma and brain folate remained unaltered by CD and hepatic folate reduced to 85% of the control by CD ( p < 0.05) . Plasma Hcy was signi-ficantly increased by FD ( 18.34 +/- 1.62 micrometer) and CFD ( 19.35 +/-3.62 micrometer) compared to the control ( 6.29 +/-0.60 micrometer) ( p < 0.001) , but remained unaltered by CD. FD depressed S-adenosylmethionine ( SAM) by 59% ( p < 0.001) and ele-vated S-adenosylhomocysteine ( SAH) by 47% in liver compared to the control group ( p < 0.001) . In contrast, brain SAM levels remained unaltered in CD, FD and CFD rats. Genomic DNA methylation status was reduced by FD in liver ( p< 0.05) . Genomic DNA hypomethylation was also observed in brain by CD, FD and CFD although it was not signifi-cantly different from the control group. Genomic DNA methylation status was correlated with folate stores in liver ( r = - 0.397, p < 0.05) and brain ( r = - 0.390, p < 0.05) , respectively. In conclusion, our data demonstrated that genomic DNA methylation and SAM level were reduced by folate deficiency in liver, but not in brain, and correlated with folate concentration in the tissue. The fact that folate deficiency had differential effects on SAM, SAH and genomic DNA methylation in liver and brain suggests that the Hcy metabolism and DNA methylation are regulated in tissue-specific ways.


Assuntos
Adulto , Animais , Humanos , Masculino , Ratos , Doença de Alzheimer , Betaína , Encéfalo , Colina , Deficiência de Colina , Dieta , Metilação de DNA , DNA , Ácido Fólico , Homocisteína , Fígado , Metabolismo , Metionina , Metilação , Sistema Nervoso , Plasma , Ratos Sprague-Dawley , Fatores de Risco , S-Adenosil-Homocisteína , S-Adenosilmetionina
9.
The Korean Journal of Nutrition ; : 811-818, 2003.
Artigo em Coreano | WPRIM | ID: wpr-649455

RESUMO

We investigated the effects of dietary folate supplementation on plasma homocysteine, vitamin B12 and hepatic levels of S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) in diet-induced hyperhomocysteinemic rats. All animals were fed 0.3% homocysteine diet for 2 weeks, then they were placed either on a 0.3% homocystine or no homocystine with or without 8 mg/kg folate diet for 8 weeks. Homocystine diet induced hyperhomocysteinemia up to 3.5-fold at 10 weeks (28.0+/-4.8 micromol/l vs. 7.9+/-0.3 micromol/l). Dietary folate supplementation caused a significant decrease in plasma homocysteine levels which had been increased by a homocystine-diet. Also, dietary folate supplementation made them return to control levels at 4 wk when the diet was free of homocystine. Plasma folate levels were markedly decreased with homocystine diet with no folate supplementation. Plasma vitamin B12 did not differ between groups. Dietary homocystine increased hepatic levels of SAM in folate supplementation group at 10 weeks (p<0.05). Dietary folate supplementation increased hepatic levels of SAM/SAH ratios in homocystine group (p<0.05). In conclusion, dietary folate supplementation can effectively ameliorate the detrimental effects of hyperhomocysteinemia.mia.


Assuntos
Animais , Ratos , Dieta , Ácido Fólico , Homocisteína , Homocistina , Hiper-Homocisteinemia , Metabolismo , Plasma , S-Adenosil-Homocisteína , S-Adenosilmetionina , Vitamina B 12
10.
Journal of the Egyptian Medical Association [The]. 1988; 71 (1-4): 255-246
em Inglês | IMEMR | ID: emr-10873

RESUMO

Adenosyl homocysteinase was prepared and purified till homogenecidy gel chromatography. The molecular weight and amino acid composition of the native enzyme were determined before and after modification of cysteinyl residue by iodoacetamide. Also the native enzyme was labelled covalently by fluorescent isothiocyanate causing inhibition of its activity. The fluorescent labelled enzyme was digested by trypsin and the fluorescent labelled peptide was isolated and the amino acid composition of this peptide was identified. The experimental data indicate that fluorescent isothiocyanate can react with lysinyl residue; which is associated with cysteine in the catalytic function of the enzyme. The catalytic activity of adenosyl homocysteinase was inhibited after incubation of the enzyme with either iodoacetate or 2-deoxycoform either iodoacetate or 2-deoxycoformycin. 2-Deoxycoformycin interact with the enzyme bound NAD causing local conformational change by which the enzymatic activity was inhibited


Assuntos
S-Adenosil-Homocisteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA