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.
Artículo en Inglés | MEDLINE | ID: mdl-33959973

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Decidua/enzimología , Deficiencia de Ácido Fólico/enzimología , Ácido Fólico/metabolismo , Células del Estroma/enzimología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagosomas/enzimología , Autofagosomas/ultraestructura , Células Cultivadas , Decidua/ultraestructura , Modelos Animales de Enfermedad , Femenino , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/patología , Lisosomas/enzimología , Lisosomas/ultraestructura , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Embarazo , Transducción de Señal , Células del Estroma/ultraestructura
2.
Rinsho Shinkeigaku ; 56(2): 116-9, 2016.
Artículo en Japonés | MEDLINE | ID: mdl-26797484

RESUMEN

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.


Asunto(s)
Deficiencia de Ácido Fólico/complicaciones , Hiperhomocisteinemia/etiología , Trombosis de los Senos Intracraneales/etiología , Deficiencia de Vitamina B 12/complicaciones , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/tratamiento farmacológico , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/genética , Humanos , Masculino , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Persona de Mediana Edad , Mutación , Factores de Riesgo , Resultado del Tratamiento , Vitamina B 12/administración & dosificación , Deficiencia de Vitamina B 12/tratamiento farmacológico
3.
Am J Clin Nutr ; 102(5): 1279-88, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26354538

RESUMEN

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.


Asunto(s)
Deficiencia de Ácido Fólico/genética , Eliminación de Gen , Trastornos de la Memoria/etiología , Estado Nutricional , Polimorfismo Genético , Tetrahidrofolato Deshidrogenasa/genética , Negro o Afroamericano , Anciano , Anciano de 80 o más Años , Boston/epidemiología , Estudios de Cohortes , Estudios Transversales , Femenino , Ácido Fólico/envenenamiento , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/fisiopatología , Estudios de Asociación Genética , Hispánicos o Latinos , Humanos , Masculino , Trastornos de la Memoria/epidemiología , Persona de Mediana Edad , Nutrigenómica/métodos , Prevalencia , Puerto Rico/etnología , Tetrahidrofolato Deshidrogenasa/metabolismo , Población Blanca
4.
J Nutr Health Aging ; 19(2): 123-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25651436

RESUMEN

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.


Asunto(s)
Ácido Fólico/farmacología , Neuroblastoma/metabolismo , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Enfermedad de Alzheimer/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/metabolismo , Humanos , Metilación/efectos de los fármacos , Neuroblastoma/enzimología , Neuroblastoma/patología , Ácido Ocadaico/farmacología , Fosforilación/efectos de los fármacos
5.
J Biol Chem ; 289(43): 29642-50, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25213861

RESUMEN

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.


Asunto(s)
Núcleo Celular/metabolismo , Coenzimas/metabolismo , Deficiencia de Ácido Fólico/enzimología , Ácido Fólico/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Timidina Monofosfato/biosíntesis , Animales , Puntos de Control del Ciclo Celular , Línea Celular , ADN/metabolismo , Dieta , Femenino , Deficiencia de Ácido Fólico/patología , Formiatos/sangre , Técnicas de Silenciamiento del Gen , Glicina Hidroximetiltransferasa/metabolismo , Humanos , Hígado/enzimología , Masculino , Metionina/biosíntesis , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas , Purinas/biosíntesis , Fase S , Uracilo/metabolismo
6.
J Nutr ; 143(1): 41-5, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23190757

RESUMEN

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.


Asunto(s)
Colina/metabolismo , Deficiencia de Ácido Fólico/enzimología , Hígado/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Colina/sangre , Cisteína/sangre , Cisteína/metabolismo , Modelos Animales de Enfermedad , Deficiencia de Ácido Fólico/sangre , Deficiencia de Ácido Fólico/metabolismo , Heterocigoto , Homocisteína/sangre , Homocisteína/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Hígado/enzimología , Masculino , Metionina/sangre , Metionina/metabolismo , Metilación , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis Insercional , Proteínas Mutantes/metabolismo , Distribución Aleatoria
7.
Cancer Causes Control ; 23(8): 1265-77, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22706675

RESUMEN

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.


Asunto(s)
Ferredoxina-NADP Reductasa/genética , Deficiencia de Ácido Fólico/prevención & control , Ácido Fólico/administración & dosificación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/prevención & control , Complicaciones del Embarazo/prevención & control , Estudios de Casos y Controles , Preescolar , Suplementos Dietéticos , Femenino , Deficiencia de Ácido Fólico/tratamiento farmacológico , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/genética , Predisposición Genética a la Enfermedad , Humanos , Recién Nacido , Masculino , Polimorfismo de Nucleótido Simple , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/enzimología , Complicaciones del Embarazo/genética
8.
Reproduction ; 144(1): 91-100, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22580371

RESUMEN

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.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Implantación del Embrión , Deficiencia de Ácido Fólico/enzimología , Útero/enzimología , Animales , Western Blotting , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/análisis , ADN Metiltransferasa 3A , Dieta , Implantación del Embrión/genética , Implantación del Embrión/fisiología , Femenino , Ácido Fólico/administración & dosificación , Expresión Génica , Inmunohistoquímica , Masculino , Ratones , Embarazo , Seudoembarazo , ARN Mensajero/análisis , ADN Metiltransferasa 3B
9.
J Nutr ; 140(10): 1736-41, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20724492

RESUMEN

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.


Asunto(s)
Carbono/metabolismo , Hígado Graso/enzimología , Hígado Graso/etiología , Deficiencia de Ácido Fólico/complicaciones , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Animales , Betaína/análisis , Betaína-Homocisteína S-Metiltransferasa/genética , Colina/metabolismo , Dieta , Femenino , Ferredoxina-NADP Reductasa/genética , Ácido Fólico/administración & dosificación , Deficiencia de Ácido Fólico/enzimología , Expresión Génica , Homocisteína/sangre , Homocisteína/metabolismo , Hígado/química , Hígado/enzimología , Masculino , Metilación , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/análisis , Factores Sexuales
10.
J Gastroenterol Hepatol ; 25(6): 1157-61, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20594233

RESUMEN

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.


Asunto(s)
Colitis Ulcerosa/genética , ADN/genética , Deficiencia de Ácido Fólico/etiología , Ácido Fólico/metabolismo , Hiperhomocisteinemia/etiología , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Polimorfismo Genético , Adulto , Alelos , China/epidemiología , Colitis Ulcerosa/complicaciones , Colitis Ulcerosa/enzimología , Femenino , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/epidemiología , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Homocisteína/sangre , Humanos , Hiperhomocisteinemia/enzimología , Hiperhomocisteinemia/epidemiología , Técnicas para Inmunoenzimas , Masculino , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Persona de Mediana Edad , Prevalencia , Vitamina B 12/sangre
11.
Eur J Pediatr ; 169(2): 241-3, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19434424

RESUMEN

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.


Asunto(s)
5,10-Metilenotetrahidrofolato Reductasa (FADH2)/deficiencia , Betaína/administración & dosificación , Deficiencia de Ácido Fólico/tratamiento farmacológico , 5,10-Metilenotetrahidrofolato Reductasa (FADH2)/sangre , Preescolar , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Deficiencia de Ácido Fólico/enzimología , Estudios de Seguimiento , Homocisteína/sangre , Homocisteína/efectos de los fármacos , Humanos , Lipotrópicos/administración & dosificación , Masculino , Factores de Tiempo
12.
Biochem Biophys Res Commun ; 379(3): 732-7, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19121630

RESUMEN

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.


Asunto(s)
Apoptosis , Deficiencia de Ácido Fólico/enzimología , Ácido Fólico/metabolismo , Linfocitos/patología , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Necrosis/metabolismo , Adulto , Apoptosis/genética , Daño del ADN/genética , Femenino , Humanos , Linfocitos/metabolismo , Persona de Mediana Edad , Necrosis/genética , Polimorfismo Genético
13.
Ann Nutr Metab ; 52(4): 315-21, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18714149

RESUMEN

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.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Adolescentes , Homocisteína/sangre , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Estado Nutricional , Polimorfismo Genético/genética , Complejo Vitamínico B/sangre , Adolescente , Análisis de Varianza , Índice de Masa Corporal , Femenino , Ácido Fólico/sangre , Deficiencia de Ácido Fólico/sangre , Deficiencia de Ácido Fólico/enzimología , Humanos , Masculino , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Evaluación Nutricional , Factores Sexuales , Fumar , España , Vitamina B 12/sangre
14.
Forum Nutr ; 60: 140-145, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17684410

RESUMEN

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.


Asunto(s)
Ligasas de Carbono-Nitrógeno/genética , Neoplasias Esofágicas/genética , Ácido Fólico/metabolismo , Neoplasias Gastrointestinales/genética , Polimorfismo Genético , Timidilato Sintasa/genética , China , Neoplasias Esofágicas/enzimología , Neoplasias Esofágicas/etnología , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/metabolismo , Neoplasias Gastrointestinales/enzimología , Neoplasias Gastrointestinales/etnología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Riesgo
15.
J Nutr ; 135(12): 2780-5, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16317120

RESUMEN

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.


Asunto(s)
Deficiencia de Ácido Fólico/enzimología , Glicina N-Metiltransferasa/genética , Homocisteína/sangre , Polimorfismo de Nucleótido Simple , Adulto , Femenino , Ácido Fólico/metabolismo , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética
16.
Alcohol Clin Exp Res ; 29(12): 2188-93, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16385189

RESUMEN

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.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Deficiencia de Ácido Fólico/enzimología , Hígado/enzimología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Amidohidrolasas/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Ácido Fólico/sangre , Ácido Fólico/metabolismo , Ácido Fólico/orina , Homocisteína/sangre , Hígado/efectos de los fármacos , Masculino , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Vitaminas/metabolismo , Vitaminas/orina , Aumento de Peso/efectos de los fármacos
17.
J Nutr ; 135(5): 1045-50, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15867279

RESUMEN

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.


Asunto(s)
Deficiencia de Ácido Fólico/enzimología , Ácido Fólico/sangre , Homocisteína/biosíntesis , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Polimorfismo de Nucleótido Simple , Adulto , Aminoácidos/sangre , Aminoácidos/química , Femenino , Cromatografía de Gases y Espectrometría de Masas , Homocisteína/sangre , Humanos , Leucina/metabolismo , Metionina/metabolismo , Metilación , Serina/metabolismo
18.
Clin Chim Acta ; 340(1-2): 99-105, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14734201

RESUMEN

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.


Asunto(s)
Deficiencia de Ácido Fólico/sangre , Deficiencia de Ácido Fólico/enzimología , Ácido Fólico/sangre , Homocisteína/sangre , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Polimorfismo Genético/genética , Adulto , Femenino , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Genotipo , Humanos , Masculino , Vitamina B 12/sangre
20.
Brain Dev ; 24(4): 223-6, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12015164

RESUMEN

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.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Epilepsia/genética , Hiperhomocisteinemia/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Mutación Puntual , Adolescente , Adulto , Niño , Preescolar , Epilepsia/tratamiento farmacológico , Epilepsia/enzimología , Ácido Fólico/sangre , Deficiencia de Ácido Fólico/enzimología , Deficiencia de Ácido Fólico/genética , Genotipo , Humanos , Lactante , Metilenotetrahidrofolato Reductasa (NADPH2)
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...