Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Cell Physiol ; 236(11): 7376-7389, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33959973

RESUMO

Existing evidence suggests that adverse pregnancy outcomes are closely related to dietary factors. Folate plays an important role in neural tube formation and fetal growth, folate deficiency is a major risk factor of birth defects. Our early studies showed that folate deficiency could impair enddecidualization, however, the mechanism is still unclear. Dysfunctional autophagy is associated with many diseases. Here, we aimed to evaluate the adverse effect of folate deficiency on endometrial decidualization, with a particular focus on endometrial cell autophagy. Mice were fed with no folate diet in vivo and the mouse endometrial stromal cell was cultured in a folate-free medium in vitro. The decrease of the number of endometrial autophagosomes and the protein expressions of autophagy in the folate-deficient group indicated that autophagosome formation, autophagosome-lysosome fusion, and lysosomal degradation were inhibited. Autophagic flux examination using mCherry-GFP-LC3 transfection showed that the fusion of autophagosomes with lysosomes was inhibited by folate deficiency. Autophagy inducer rapamycin could reverse the impairment of folate deficiency on endometrial decidualization. Moreover, folate deficiency could reduce autophagy by disrupting AMPK/mTOR signaling, resulting in aberrant endometrial decidualization and adverse pregnancy outcomes. Further co-immunoprecipitation examination showed that decidual marker protein Hoxa10 could interact with autophagic marker protein Cathepsin L, and the interaction was notably reduced by folate deficiency. In conclusion, AMPK/mTOR downregulated autophagy was essential for aberrant endometrial decidualization in early pregnant mice, which could result in adverse pregnancy outcomes. This provided some new clues for understanding the causal mechanisms of birth defects induced by folate deficiency.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia , Decídua/enzimologia , Deficiência de Ácido Fólico/enzimologia , Ácido Fólico/metabolismo , Células Estromais/enzimologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagossomos/enzimologia , Autofagossomos/ultraestrutura , Células Cultivadas , Decídua/ultraestrutura , Modelos Animais de Doenças , Feminino , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/patologia , Lisossomos/enzimologia , Lisossomos/ultraestrutura , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Gravidez , Transdução de Sinais , Células Estromais/ultraestrutura
2.
Rinsho Shinkeigaku ; 56(2): 116-9, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-26797484

RESUMO

A 63-year-old man was admitted to our hospital because of convulsive seizures. Radiological examinations revealed cerebral venous sinus thrombosis in the anterior part of the superior sagittal sinus. He had marked hyperhomocysteinemia (93.5 nmol/ml) due to combined deficiencies of folate and vitamin B12. He was T/T homozygous for methylene tetrahydrofolate reductase C677T polymorphism. He received a supplement therapy of vitamins. First, he was administered folate orally. After 3 months, the serum level of homocysteine decreased to 22.6 nmol/ml (an 86% reduction), but was still above the normal level. Next, an additional supplement therapy of vitamin B12 lowered the homocysteine level to normal (12.3 nmol/ml) after 4 months. These results showed that the increase of homocysteine levels in this patient was mainly caused by the deficiency of folate. Additionally, acquired risk factors like vitamin deficiencies increased the level of serum homocysteine to almost 100 nmol/ml.


Assuntos
Deficiência de Ácido Fólico/complicações , Hiper-Homocisteinemia/etiologia , Trombose dos Seios Intracranianos/etiologia , Deficiência de Vitamina B 12/complicações , Ácido Fólico/administração & dosagem , Deficiência de Ácido Fólico/tratamento farmacológico , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/genética , Humanos , Masculino , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Pessoa de Meia-Idade , Mutação , Fatores de Risco , Resultado do Tratamento , Vitamina B 12/administração & dosagem , Deficiência de Vitamina B 12/tratamento farmacológico
3.
Am J Clin Nutr ; 102(5): 1279-88, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26354538

RESUMO

BACKGROUND: Folate status has been positively associated with cognitive function in many studies; however, some studies have observed associations of poor cognitive outcomes with high folate. In search of an explanation, we hypothesized that the association of folate with cognition would be modified by the interaction of high-folate status with a common 19-bp deletion polymorphism in the dihydrofolate reductase (DHFR) gene. To our knowledge, the cognitive effects of this gene have not been studied previously. OBJECTIVE: We examined the association between cognitive outcomes with the 19-bp deletion DHFR polymorphism, folate status, and their interaction with high or normal plasma folate. DESIGN: This was a pooled cross-sectional study of the following 2 Boston-based cohorts of community living adults: the Boston Puerto Rican Health Study and the Nutrition, Aging, and Memory in Elders study. Individuals were genotyped for the DHFR 19-bp deletion genotype, and plasma folate status was determined. Cognitive outcomes included the Mini-Mental State Examination, Center for Epidemiologic Studies Depression Scale, and factor scores for the domains of memory, executive function, and attention from a set of cognitive tests. RESULTS: The prevalence of the homozygous deletion (del/del) genotype was 23%. In a multivariable analysis, high folate status (>17.8 ng/mL) was associated with better memory scores than was normal-folate status (fourth-fifth quintiles compared with first-third quintiles: ß ± SE = -0.22 ± 0.06, P < 0.01). Carriers of the DHFR del/del genotype had worse memory scores (ß ± SE = -0.24 ± 0.10, P < 0.05) and worse executive scores (ß = -0.19, P < 0.05) than did those with the del/ins and ins/ins genotypes. Finally, we observed an interaction such that carriers of the del/del genotype with high folate had significantly worse memory scores than those of both noncarriers with high-folate and del/del carriers with normal-folate (ß-interaction = 0.26 ± 0.13, P < 0.05). CONCLUSIONS: This study identifies a putative gene-nutrient interaction that, if confirmed, would predict that a sizable minority carrying the del/del genotype might not benefit from high-folate status and could see a worsening of memory. An understanding of how genetic variation affects responses to high-folate exposure will help weigh risks and benefits of folate supplementation for individuals and public health.


Assuntos
Deficiência de Ácido Fólico/genética , Deleção de Genes , Transtornos da Memória/etiologia , Estado Nutricional , Polimorfismo Genético , Tetra-Hidrofolato Desidrogenase/genética , Negro ou Afro-Americano , Idoso , Idoso de 80 Anos ou mais , Boston/epidemiologia , Estudos de Coortes , Estudos Transversais , Feminino , Ácido Fólico/intoxicação , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/fisiopatologia , Estudos de Associação Genética , Hispânico ou Latino , Humanos , Masculino , Transtornos da Memória/epidemiologia , Pessoa de Meia-Idade , Nutrigenômica/métodos , Prevalência , Porto Rico/etnologia , Tetra-Hidrofolato Desidrogenase/metabolismo , População Branca
4.
J Nutr Health Aging ; 19(2): 123-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25651436

RESUMO

OBJECTIVES: Neurofibrillary tangles (NFTs), which are composed of intracellular filamentous aggregates of hyperphosphorylated tau protein, are one of the pathological hallmarks of Alzheimer's disease (AD). Because tau phosphorylation is regulated by phosphatases, abnormal metabolism of protein phosphatase 2A (PP2A) has been proposed to be a contributing factor to the disease process. RESULTS: To determine the function of folic acid on tau phosphorylation, an in vitro model of human neuroblastoma cells (SH-SY5Y) were exposed to folic acid (0-40 µmol/L) for 96 h, in the presence or absence of the phosphoesterase inhibitor okadaic acid (OA) (10 nmol/L) for 9 h. The data of western blot showed tau phosphorylation at the Ser396 site in OA-incubated SH-SY5Y cells was inhibited by folic acid in a concentration-dependent manner, with the folic acid concentration of 40 µmol/L providing maximal inhibition. Folic acid can downregulate tau protein phosphorylation by inhibiting the demethylation reactions of PP2A. High folic acid concentrations (20 and 40 µmol/L) increased SAM:SAH ratios and cell viability. CONCLUSION: Therefore, we can speculate that folate deficiency may be a cause of PP2A deregulation, which can in turn lead to expression of the abnormal hyperphosphorylated form of tau.


Assuntos
Ácido Fólico/farmacologia , Neuroblastoma/metabolismo , Proteína Fosfatase 2/química , Proteína Fosfatase 2/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/metabolismo , Humanos , Metilação/efeitos dos fármacos , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Ácido Okadáico/farmacologia , Fosforilação/efeitos dos fármacos
5.
J Biol Chem ; 289(43): 29642-50, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25213861

RESUMO

Folate-mediated one-carbon metabolism is a metabolic network of interconnected pathways that is required for the de novo synthesis of three of the four DNA bases and the remethylation of homocysteine to methionine. Previous studies have indicated that the thymidylate synthesis and homocysteine remethylation pathways compete for a limiting pool of methylenetetrahydrofolate cofactors and that thymidylate biosynthesis is preserved in folate deficiency at the expense of homocysteine remethylation, but the mechanisms are unknown. Recently, it was shown that thymidylate synthesis occurs in the nucleus, whereas homocysteine remethylation occurs in the cytosol. In this study we demonstrate that methylenetetrahydrofolate dehydrogenase 1 (MTHFD1), an enzyme that generates methylenetetrahydrofolate from formate, ATP, and NADPH, functions in the nucleus to support de novo thymidylate biosynthesis. MTHFD1 translocates to the nucleus in S-phase MCF-7 and HeLa cells. During folate deficiency mouse liver MTHFD1 levels are enriched in the nucleus >2-fold at the expense of levels in the cytosol. Furthermore, nuclear folate levels are resistant to folate depletion when total cellular folate levels are reduced by >50% in mouse liver. The enrichment of folate cofactors and MTHFD1 protein in the nucleus during folate deficiency in mouse liver and human cell lines accounts for previous metabolic studies that indicated 5,10-methylenetetrahydrofolate is preferentially directed toward de novo thymidylate biosynthesis at the expense of homocysteine remethylation during folate deficiency.


Assuntos
Núcleo Celular/metabolismo , Coenzimas/metabolismo , Deficiência de Ácido Fólico/enzimologia , Ácido Fólico/metabolismo , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Timidina Monofosfato/biossíntese , Animais , Pontos de Checagem do Ciclo Celular , Linhagem Celular , DNA/metabolismo , Dieta , Feminino , Deficiência de Ácido Fólico/patologia , Formiatos/sangue , Técnicas de Silenciamento de Genes , Glicina Hidroximetiltransferase/metabolismo , Humanos , Fígado/enzimologia , Masculino , Metionina/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , Purinas/biossíntese , Fase S , Uracila/metabolismo
6.
J Nutr ; 143(1): 41-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23190757

RESUMO

Impaired utilization of folate is caused by insufficient dietary intake and/or genetic variation and has been shown to prompt changes in related pathways, including choline and methionine metabolism. These pathways have been shown to be sensitive to variation within the Mthfd1 gene, which codes for a folate-metabolizing enzyme responsible for generating 1-carbon (1-C)-substituted folate derivatives. The Mthfd1(gt/+) mouse serves as a potential model of human Mthfd1 loss-of-function genetic variants that impair MTHFD1 function. This study investigated the effects of the Mthfd1(gt/+) genotype and folate intake on markers of choline, folate, methionine, and transsulfuration metabolism. Male Mthfd1(gt/+) and Mthfd1(+/+) mice were randomly assigned at weaning (3 wk of age) to either a control (2 mg/kg folic acid) or folate-deficient (0 mg/kg folic acid) diet for 5 wk. Mice were killed at 8 wk of age following 12 h of food deprivation; blood and liver samples were analyzed for choline, methionine, and transsulfuration biomarkers. Independent of folate intake, mice with the Mthfd1(gt/+) genotype had higher hepatic concentrations of choline (P = 0.005), betaine (P = 0.013), and dimethylglycine (P = 0.004) and lower hepatic concentrations of glycerophosphocholine (P = 0.002) relative to Mthfd1(+/+) mice. Mthfd1(gt/+) mice also had higher plasma concentrations of homocysteine (P = 0.0016) and cysteine (P < 0.001) as well as lower plasma concentrations of methionine (P = 0.0003) and cystathionine (P = 0.011). The metabolic alterations observed in Mthfd1(gt/+) mice indicate perturbed choline and folate-dependent 1-C metabolism and support the future use of Mthfd1(gt/+) mice as a tool to investigate the impact of impaired 1-C metabolism on disease outcomes.


Assuntos
Colina/metabolismo , Deficiência de Ácido Fólico/enzimologia , Fígado/metabolismo , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Colina/sangue , Cisteína/sangue , Cisteína/metabolismo , Modelos Animais de Doenças , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/metabolismo , Heterozigoto , Homocisteína/sangue , Homocisteína/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Fígado/enzimologia , Masculino , Metionina/sangue , Metionina/metabolismo , Metilação , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutagênese Insercional , Proteínas Mutantes/metabolismo , Distribuição Aleatória
7.
Cancer Causes Control ; 23(8): 1265-77, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22706675

RESUMO

PURPOSE: Fetal folate deficiency may increase the risk of subsequent childhood acute leukemia (AL), since folates are required for DNA methylation, synthesis, and repair, but the literature remains scarce. This study tested the hypothesis that maternal folic acid supplementation before or during pregnancy reduces AL risk, accounting for the SNPs rs1801133 (C677T) and rs1801131 (A1298C) in MTHFR and rs1801394 (A66G) and rs1532268 (C524T) in MTRR, assumed to modify folate metabolism. METHODS: The nationwide registry-based case-control study, ESCALE, carried out in 2003-2004, included 764 AL cases and 1,681 controls frequency matched with the cases on age and gender. Information on folic acid supplementation was obtained by standardized telephone interview. The genotypes were obtained using high-throughput platforms and imputation for untyped polymorphisms. Odds ratios (OR) were estimated using unconditional regression models adjusted for potential confounders. RESULTS: AL was significantly inversely associated with maternal folic acid supplementation before and during pregnancy (OR = 0.4; 95 % confidence interval: [0.3-0.6]). MTHFR and MTRR genetic polymorphisms were not associated with AL. However, AL was positively associated with homozygosity for any of the MTHFR polymorphisms and carriership of both MTRR variant alleles (OR = 1.6 [0.9-3.1]). No interaction was observed between MTHFR, MTRR, and maternal folate supplementation. CONCLUSION: The study findings support the hypothesis that maternal folic acid supplementation may reduce the risk of childhood AL. The findings also suggest that the genotype homozygous for any of the MTHFR variants and carrying both MTRR variants could be a risk factor for AL.


Assuntos
Ferredoxina-NADP Redutase/genética , Deficiência de Ácido Fólico/prevenção & controle , Ácido Fólico/administração & dosagem , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/prevenção & controle , Complicações na Gravidez/prevenção & controle , Estudos de Casos e Controles , Pré-Escolar , Suplementos Nutricionais , Feminino , Deficiência de Ácido Fólico/tratamento farmacológico , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/genética , Predisposição Genética para Doença , Humanos , Recém-Nascido , Masculino , Polimorfismo de Nucleotídeo Único , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimologia , Gravidez , Complicações na Gravidez/tratamento farmacológico , Complicações na Gravidez/enzimologia , Complicações na Gravidez/genética
8.
Reproduction ; 144(1): 91-100, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22580371

RESUMO

We have characterized the uterine expression of DNA methyltransferases (DNMTs) during early pregnancy in mice and determined whether a folate-deficient diet (FDD) can affect DNMTs in this context. Within endometrial cells, expressions of DNMT (cytosine-5) 1 (Dnmt1), Dnmt3a, and Dnmt3b were significantly elevated during the prereceptive phase of pregnancy but generally returned to baseline levels during receptive and postimplantation periods. As such, the transcription of DNMT genes is temporally regulated during early pregnancy. When comparisons were made between implantation sites (IS) and inter-IS on day 5 of pregnancy, lower levels of Dnmt3a were detected at IS. Comparisons between IS and inter-IS did not reveal significant expression differences for other DNMT genes. When tissue sections were examined, DNMT3A was specifically lower in the stroma of IS. Reduced DNMT1 and DNMT3B levels were also observed in the luminal and glandular epithelia of IS, whereas no obvious differences in the stroma were detected. In pseudo-pregnant mice subjected to a FDD, levels of Dnmt1 and Dnmt3a (but not Dnmt3b) were significantly upregulated in endometrial tissues, as compared with controls. When tissues from these folate-deficient mice were examined, DNMT1 levels were elevated in both the luminal and glandular epithelia, whereas DNMT3A was upregulated in the luminal epithelium and the stroma. A slight increase in DNMT3B levels was detected in the glandular epithelium. These results indicate that DNMTs may regulate the transcription of endometrial genes associated with embryo implantation and that levels of DNMTs are affected by dietary folate in mice.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Implantação do Embrião , Deficiência de Ácido Fólico/enzimologia , Útero/enzimologia , Animais , Western Blotting , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/análise , DNA Metiltransferase 3A , Dieta , Implantação do Embrião/genética , Implantação do Embrião/fisiologia , Feminino , Ácido Fólico/administração & dosagem , Expressão Gênica , Imuno-Histoquímica , Masculino , Camundongos , Gravidez , Pseudogravidez , RNA Mensageiro/análise , DNA Metiltransferase 3B
9.
J Nutr ; 140(10): 1736-41, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20724492

RESUMO

Disrupted choline metabolism may affect hepatic lipid metabolism and lead to steatosis. Because folate and the choline metabolite betaine independently serve as methyl donors for homocysteine (Hcy) remethylation to methionine, we assessed the impact of folate deficiency on steatosis, choline metabolism, and expression of 9 genes involved in folate-mediated one-carbon metabolism. Liver histology, choline metabolites, and mRNA and protein expression were examined in mice fed control (CD; 2 mg/kg folic acid) or folate-deficient diets (FD; 0.3 mg/kg folic acid) for 12 mo. Females fed CD were not steatotic (0/6), whereas males were mildly to moderately steatotic (5/6). Steatosis was observed in FD-fed males and females; it was more severe and more frequent in males (7/7) than in females (4/10) (P = 0.005). Hepatic betaine was lower in males (P = 0.014) and FD-fed mice (P < 0.001) and negatively correlated with steatosis severity in mice fed CD (r = -0.87; P = 0.001). Gender differences in the expression of 6 enzymes may contribute to increased steatosis susceptibility in males. Males relied more on betaine-dependent (folate-independent) Hcy remethylation [72% more betaine-Hcy methyltransferase (P < 0.001) and 28% less folate-dependent methionine synthase (MTR) (P < 0.001)]. FD-fed mice of both genders appeared to shift to betaine-dependent remethylation by reducing MTR expression 70% (P < 0.001) and increasing betaine demand; there was a correlation between MTR expression and betaine levels (r = 0.50; P = 0.031). Our work demonstrates that chronic folate insufficiency leads to steatosis in mice. Increased utilization of betaine for Hcy remethylation in males and in both genders during folate deficiency may lead to steatosis by disrupting choline metabolism.


Assuntos
Carbono/metabolismo , Fígado Gorduroso/enzimologia , Fígado Gorduroso/etiologia , Deficiência de Ácido Fólico/complicações , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Animais , Betaína/análise , Betaína-Homocisteína S-Metiltransferase/genética , Colina/metabolismo , Dieta , Feminino , Ferredoxina-NADP Redutase/genética , Ácido Fólico/administração & dosagem , Deficiência de Ácido Fólico/enzimologia , Expressão Gênica , Homocisteína/sangue , Homocisteína/metabolismo , Fígado/química , Fígado/enzimologia , Masculino , Metilação , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Fatores Sexuais
10.
J Gastroenterol Hepatol ; 25(6): 1157-61, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20594233

RESUMO

BACKGROUND AND AIMS: Methylenetetrahydrofolate reductase (MTHFR) encoding genes were associated with ulcerative colitis in Chinese in our previous study. We further studied association of a new polymorphism of MTHFR G1793A with ulcerative colitis and assessed relationship of this polymorphism with hyperhomocysteinemia (HHcy, > or = 15 mmol/L) and deficiency of folate (< or = 7 nmol/L) and vitamin B(12) (< or = 150 pmol/L) in a cohort of patients with ulcerative colitis in central China. METHODS: A total of 252 patients and 654 healthy controls were recruited. Polymorphism of MTHFR G1793A was examined using a polymerase chain reaction-restriction fragment length polymorphism method. Plasma levels of homocysteine (Hcy), folate and vitamin B(12) were determined by enzymatic cycling assay and corpuscle immune chemiluminescence assay, respectively. RESULTS: Frequencies of alleles and genotypes in MTHFR G1793A gene differed significantly between ulcerative colitis patients and the healthy controls (20.83% vs 10.47%, 95% confidence interval [CI]: 1.703-2.972, P = 0.0006; 40.48% vs 19.88%, 95% CI: 1.997-3.761, P = 0.0002, respectively). Plasma Hcy levels were higher and folate and vitamin B(12) concentrations were lower in the patients than in the healthy controls (21.72 +/- 6.59 vs 12.47 +/- 5.02, 95% CI: -10.93--7.58, P < 0.0001; 11.25 +/- 6.19 vs 15.28 +/- 7.72, 95% CI: 2.03-6.04; P < 0.001; 322.81 +/- 128.47 vs 442.59 +/- 129.36, 95% CI: 62.61-136.95, P < 0.0001, respectively). HHcy and folate deficiency were more prevalent in patients with ulcerative colitis (45.32% vs 26.17%, 95% CI: 1.285-4.378, P = 0.005; 30.68% vs 13.0%, 95% CI: 1.416-6.197, P = 0.003, respectively). CONCLUSIONS: MTHFR G1793A gene polymorphism, HHcy, folate deficiency and low vitamin B(12) concentration were associated with ulcerative colitis in central China. Our findings demonstrate that the Hcy-related gene and metabolites are involved in pathogenesis of ulcerative colitis.


Assuntos
Colite Ulcerativa/genética , DNA/genética , Deficiência de Ácido Fólico/etiologia , Ácido Fólico/metabolismo , Hiper-Homocisteinemia/etiologia , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Polimorfismo Genético , Adulto , Alelos , China/epidemiologia , Colite Ulcerativa/complicações , Colite Ulcerativa/enzimologia , Feminino , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/epidemiologia , Frequência do Gene , Predisposição Genética para Doença , Genótipo , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/enzimologia , Hiper-Homocisteinemia/epidemiologia , Técnicas Imunoenzimáticas , Masculino , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Pessoa de Meia-Idade , Prevalência , Vitamina B 12/sangue
11.
Eur J Pediatr ; 169(2): 241-3, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19434424

RESUMO

Betaine therapy was given for 2 years to a 2-year-old boy with 5,10-methylenetetrahydrofolate reductase deficiency. Used as a methyl donor to lower homocysteine levels through methylation of methionine, betaine has been reported to be effective in treating homocystinuria. Satisfactory biochemical and clinical responses were obtained with the following regimen: betaine started in the newborn period at increasing doses to reach 1 g given six times a day. It is suggested that frequent administration of a moderate dose may provide clinical and biochemical benefit.


Assuntos
5,10-Metilenotetra-Hidrofolato Redutase (FADH2)/deficiência , Betaína/administração & dosagem , Deficiência de Ácido Fólico/tratamento farmacológico , 5,10-Metilenotetra-Hidrofolato Redutase (FADH2)/sangue , Pré-Escolar , Relação Dose-Resposta a Droga , Esquema de Medicação , Deficiência de Ácido Fólico/enzimologia , Seguimentos , Homocisteína/sangue , Homocisteína/efeitos dos fármacos , Humanos , Lipotrópicos/administração & dosagem , Masculino , Fatores de Tempo
12.
Biochem Biophys Res Commun ; 379(3): 732-7, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19121630

RESUMO

Apoptosis (APO) and necrosis (NEC) are two different types of cell death occurring in response to cellular stress factors. Cells with DNA damage may undergo APO or NEC. Folate is an essential micronutrient associated with DNA synthesis, repair and methylation. Methylenetetrahydrofolate reductase (MTHFR) regulates intracellular folate metabolism. Folate deficiency and MTHFR C677T polymorphisms have been shown to be related to DNA damage. To verify the cytotoxic effects of folate deficiency on cells with different MTHFR C677T genotypes, 15 human peripheral lymphocyte cases with different MTHFR C677T genotypes were cultured in folic acid (FA)-deficient and -sufficient media for 9 days. Cytotoxicity was quantified using the frequencies of APO and NEC as endpoints, the nuclear division index (NDI), and the number of viable cells (NVC). These results showed that FA is an important factor in reducing cytotoxicity and increasing cell proliferation. Lymphocytes with the TT genotype proliferated easily under stress and exhibited different responses to FA deficiency than lymphocytes with the CC and CT genotypes. A TT individual may accumulate more cytotoxicity under cytotoxic stress, suggesting that the effects of FA deficiency on cytotoxicity are greater than the effects in individuals with the other MTHFR C677T variants.


Assuntos
Apoptose , Deficiência de Ácido Fólico/enzimologia , Ácido Fólico/metabolismo , Linfócitos/patologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Necrose/metabolismo , Adulto , Apoptose/genética , Dano ao DNA/genética , Feminino , Humanos , Linfócitos/metabolismo , Pessoa de Meia-Idade , Necrose/genética , Polimorfismo Genético
13.
Ann Nutr Metab ; 52(4): 315-21, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18714149

RESUMO

BACKGROUND: Folate and cobalamin are responsible for healthy growth. However, the B-vitamin and homocysteine status of adolescents is not well known. The aim was to assess the status of folate, cobalamin, and homocysteine in healthy Spanish adolescents. METHODS: Serum cobalamin, serum folate, homocysteine, methylenetetrahydrofolate reductase 677C>T variant, BMI, smoking habits, and Tanner stage were determined according to gender in 165 adolescents (84 females, 81 males; 13-18.5 years) using the Student's t test, Mann-Whitney U test and chi(2) test, respectively. Interactions between socioeconomic status, age group, methylenetetrahydrofolate reductase polymorphism, BMI, smoking habits, Tanner stage, and vitamin status, respectively, were examined by ANOVA or Kruskal-Wallis H test (p < 0.05). RESULTS: Boys had markedly higher homocysteine (males 8.92 (5.51-22.94) micromol/l; females 7.91 (5.09-13.86) micromol/l), whereas girls showed higher serum cobalamin concentrations (males 540.00 (268.00-946.47) pmol/l; females 594.82 (280.63-1,559.64) pmol/l). Data are shown as medians and the 2.5th to 97.5th percentiles in parentheses. Adolescents with the homozygous variant of methylenetetrahydrofolate reductase displayed significantly higher homocysteine and lower serum folate: normal 5.73 (3.09-10.73) ng/ml serum folate, 7.57 (4.94-12.94) micromol/l homocysteine; homozygous 4.10 (2.75-7.88) ng/ml serum folate, 10.83 (7.00-22.82) micromol/l homocysteine. CONCLUSION: The present study provides data on the folate, cobalamin, and homocysteine status of Spanish adolescents. To assure a better assessment, revision of references for adolescents is still needed.


Assuntos
Fenômenos Fisiológicos da Nutrição do Adolescente , Homocisteína/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Estado Nutricional , Polimorfismo Genético/genética , Complexo Vitamínico B/sangue , Adolescente , Análise de Variância , Índice de Massa Corporal , Feminino , Ácido Fólico/sangue , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/enzimologia , Humanos , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Avaliação Nutricional , Fatores Sexuais , Fumar , Espanha , Vitamina B 12/sangue
14.
Forum Nutr ; 60: 140-145, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17684410

RESUMO

Folate deficiency has been associated with certain types of human cancer. We therefore investigated the effects of genetic polymorphisms in folate-metabolizing enzymes on the risk of developing gastroesophageal cancers in a Chinese population where folate deficiency is common. We found that functional polymorphisms in methylenetetrahydrofolate reductase (MTHFR) and thymidylate synthase (TS), two key enzymes involved in folate and methyl group metabolism, were significantly associated with increased risk of esophageal squamous cell carcinoma, gastric cardia carcinoma, and pancreatic carcinoma. The polymorphisms modulate risk of these cancers associated with low folate status. Our results suggest that MTHFR and TS genotypes may be determinant of gastroesophageal cancers in this at-risk Chinese population.


Assuntos
Carbono-Nitrogênio Ligases/genética , Neoplasias Esofágicas/genética , Ácido Fólico/metabolismo , Neoplasias Gastrointestinais/genética , Polimorfismo Genético , Timidilato Sintase/genética , China , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/etnologia , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/metabolismo , Neoplasias Gastrointestinais/enzimologia , Neoplasias Gastrointestinais/etnologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Risco
15.
Alcohol Clin Exp Res ; 29(12): 2188-93, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16385189

RESUMO

BACKGROUND: We recently observed that ethanol feeding impairs 10-formyltetrahydrofolate (10-FTHF) dehydrogenase (EC 1.5.1.6.) and 10-FTHF hydrolase activity in rats. In the present study, we explored the effects of folate deficiency or sufficiency combined with alcoholic intake on 10-FTHF and possible mechanisms by which chronic ethanol ingestion produces folate deficiency. METHODS: Sprague-Dawley rats were fed either folate-sufficient (FS) or folate-deficient (FD) diets; with or without ethanol (E) for four weeks. Hepatic 10-FTHF dehydrogenase and hydrolase activity, plasma folate and homocysteine were measured at baseline and after feeding experimental diets. RESULTS: Liver weight increased slightly with either folate deficiency or ethanol consumption. In rats fed the folate-sufficient diet with ethanol (FSE), plasma folate was decreased slightly (p<0.05) and plasma homocysteine elevated compared to rats fed the FS diet without ethanol. Ethanol did not affect plasma folate and plasma homocysteine in FD rats. Red-blood cell (RBC) folate was increased similarly in rats by ethanol feeding (FSE and FDE>FS and FD). Feeding folate deficient or ethanol (FSE, FD and FDE) diets depressed hepatic activities of 10-FTHF dehydrogenase, which catalyzes the oxidative deformylation of 10-FTHF to tetrahydrofolate (THF) and carbon dioxide. Rats consuming the FDE diet had the lowest enzyme activities of the experimental groups, implying that folate deficiency and ethanol consumption each affect enzyme activity. CONCLUSIONS: We confirm that ethanol decreases hepatic 10-FTHF dehydrogenase activity and show that this decrease occurs irrespective of folate status. This shows that modulation of 10-FTHF is one possible mechanism by which ethanol intake decreases folate status and affects one-carbon metabolism.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Deficiência de Ácido Fólico/enzimologia , Fígado/enzimologia , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Amidoidrolases/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Ácido Fólico/sangue , Ácido Fólico/metabolismo , Ácido Fólico/urina , Homocisteína/sangue , Fígado/efeitos dos fármacos , Masculino , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Vitaminas/metabolismo , Vitaminas/urina , Aumento de Peso/efeitos dos fármacos
16.
J Nutr ; 135(12): 2780-5, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16317120

RESUMO

Glycine N-methyltransferase (GNMT) is a key regulatory protein in folate metabolism, methionine availability, and transmethylation reactions. Perturbations in GNMT may lead to aberrations in homocysteine metabolism, a marker of numerous pathologies. The primary objective of this study was to examine the influence of the GNMT 1289 C-->T alone, and in combination with the methylenetetrahydrofolate reductase (MTHFR) 677 C-->T variant, on plasma total homocysteine concentrations in healthy young women (n = 114). Plasma total homocysteine was measured at baseline (wk 0) and after 2 wk of controlled folate restriction (135 microg/d as dietary folate equivalents). Plasma homocysteine concentrations did not differ among the GNMT C1289T genotypes at baseline. However, after folate restriction, women with the GNMT 1289 TT genotype (n = 16) had higher (P = 0.019) homocysteine concentrations than women with the CT (n = 51) or CC (n = 47) genotype. The influence of the GNMT 1289 C-->T variant on homocysteine was dependent on the MTHFR C677T genotype. In subjects with the MTHFR 677 CC genotype, homocysteine was greater (P < or = 0.05) for GNMT 1289 TT subjects relative to 1289 CT or CC subjects. However, in subjects with the MTHFR 677 TT genotype, plasma homocysteine concentrations did not differ among the GNMT C1289T genotypes. Overall, these data suggest that the GNMT 1289 C-->T polymorphism influences plasma homocysteine and is responsive to folate intake.


Assuntos
Deficiência de Ácido Fólico/enzimologia , Glicina N-Metiltransferase/genética , Homocisteína/sangue , Polimorfismo de Nucleotídeo Único , Adulto , Feminino , Ácido Fólico/metabolismo , Humanos , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética
17.
J Nutr ; 135(5): 1045-50, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15867279

RESUMO

The effects of folate status and the methylenetetrahydrofolate reductase (MTHFR) 677C-->T polymorphism on the kinetics of homocysteine metabolism are unclear. We measured the effects of dietary folate restriction on the kinetics of homocysteine remethylation and synthesis in healthy women (20-30 y old) with the MTHFR 677 C/C or T/T genotypes (n = 9/genotype) using i.v. primed, constant infusions of [(13)C(5)]methionine, [3-(13)C]serine, and [(2)H(3)]leucine before and after 7 wk of dietary folate restriction (115 mug dietary folate equivalents/d). Dietary folate restriction significantly reduced folate status ( approximately 65% reduction in serum folate) in both genotypes. Total remethylation flux was not affected by dietary folate restriction, the MTHFR 677C-->T polymorphism, or their combination. However, the percentage of remethylation from serine was reduced approximately 15% (P = 0.031) by folate restriction in C/C subjects. Further, homocysteine synthesis rates of T/T subjects and folate-restricted C/C subjects were twice that of C/C subjects at baseline. In conclusion, elevated homocysteine synthesis is a cause of mild hyperhomocysteinemia in women with marginal folate status, particularly those with the MTHFR 677 T/T genotype.


Assuntos
Deficiência de Ácido Fólico/enzimologia , Ácido Fólico/sangue , Homocisteína/biossíntese , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo de Nucleotídeo Único , Adulto , Aminoácidos/sangue , Aminoácidos/química , Feminino , Cromatografia Gasosa-Espectrometria de Massas , Homocisteína/sangue , Humanos , Leucina/metabolismo , Metionina/metabolismo , Metilação , Serina/metabolismo
18.
Clin Chim Acta ; 340(1-2): 99-105, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14734201

RESUMO

BACKGROUND: A large body of evidence links plasma concentrations of homocysteine and cardiovascular disease. Several genetic and environmental variables may modulate such relationship. We investigated the influence of methylenetetrahydrofolate reductase (MTHFR) gene variants C677T, A1298C, and T1317C on homocysteine, folate, and cobalamin concentrations in a sample of individuals from a mild folate deficiency population to better clarify the complex interactions existing among these variables. METHODS: In the present study, 209 individuals belonging to an admixed urban population characterized by mild folate deficiency were investigated. MTHFR gene variants C677T, A1298C, and T1317C were genotyped and homocysteine-, folate-, and cobalamin-determined for each individual. RESULTS: Univariate analyses showed a significant association between the C677T variant with homocysteine (P<0.001) and cobalamin (P=0.005) as well as a significant relationship between the T allele and serum folate concentrations (P<0.05). The TT genotype of the C677T polymorphism remained significantly associated with log-transformed homocysteine even after adjustment for age, sex, smoking status, ethnicity, folate, and cobalamin concentrations (P<0.01). Both univariate and multivariate analysis have failed to show any effect of the A1298C and T1317C genetic variants in homocysteine concentrations in this population. Finally, a significant interaction between folate and C677T polymorphism in the determination of homocysteine was also disclosed (P<0.005). CONCLUSIONS: Taken together, these results demonstrate a significant interaction between serum folate and MTHFR genotype in predicting homocysteine concentrations. One may consider that a differential response of homocysteine to folic acid supplementation may depend on MTHFR genotype which may have important implications when attempting to lower homocysteine concentrations in populations with mild folate deficiency.


Assuntos
Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/enzimologia , Ácido Fólico/sangue , Homocisteína/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Polimorfismo Genético/genética , Adulto , Feminino , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Genótipo , Humanos , Masculino , Vitamina B 12/sangue
20.
Brain Dev ; 24(4): 223-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12015164

RESUMO

Hyperhomocysteinemia, a possible risk factor for vascular disease can result from folate deficiency due to anticonvulsant therapy. A reaction catalyzed by 5,10-methylenetetrahydrofolate reductase (MTHFR) supplies 5-methyltetrahydrofolate, needed to remethylate homocysteine to methionine. MTHFR gene mutation (C677T) also can lead to hyperhomocysteinemia. We examined interaction between anticonvulsant therapy, C677T homozygosity, serum folate concentration, and plasma total homocysteine (tHcy) concentration in 81 epileptic patients. Patients receiving monotherapy showed no difference in occurrence of hyperhomocysteinemia (tHcy>90th percentile for controls) between homozygotes for C677T and heterozygotes or patients with no mutant MTHFR. No monotherapy patient was folate deficient (<3 ng/ml). Among patients receiving multidrug therapy, hyperhomocysteinemia in homozygotes for C677T occured significantly more often than in heterozygotes or patients with no mutant enzyme (88.9 vs. 21.1%). The same was true for folate deficiency (44.4 vs. 0%). The C677T mutation is closely related to hyperhomocysteinemia and folate deficiency in epileptic patients taking multiple anticonvulsants.


Assuntos
Anticonvulsivantes/administração & dosagem , Epilepsia/genética , Hiper-Homocisteinemia/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Mutação Puntual , Adolescente , Adulto , Criança , Pré-Escolar , Epilepsia/tratamento farmacológico , Epilepsia/enzimologia , Ácido Fólico/sangue , Deficiência de Ácido Fólico/enzimologia , Deficiência de Ácido Fólico/genética , Genótipo , Humanos , Lactente , Metilenotetra-Hidrofolato Redutase (NADPH2)
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA