Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Am J Clin Nutr ; 112(5): 1390-1403, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33022704

RESUMO

Folate, an essential nutrient found naturally in foods in a reduced form, is present in dietary supplements and fortified foods in an oxidized synthetic form (folic acid). There is widespread agreement that maintaining adequate folate status is critical to prevent diseases due to folate inadequacy (e.g., anemia, birth defects, and cancer). However, there are concerns of potential adverse effects of excess folic acid intake and/or elevated folate status, with the original concern focused on exacerbation of clinical effects of vitamin B-12 deficiency and its role in neurocognitive health. More recently, animal and observational studies have suggested potential adverse effects on cancer risk, birth outcomes, and other diseases. Observations indicating adverse effects from excess folic acid intake, elevated folate status, and unmetabolized folic acid (UMFA) remain inconclusive; the data do not provide the evidence needed to affect public health recommendations. Moreover, strong biological and mechanistic premises connecting elevated folic acid intake, UMFA, and/or high folate status to adverse health outcomes are lacking. However, the body of evidence on potential adverse health outcomes indicates the need for comprehensive research to clarify these issues and bridge knowledge gaps. Three key research questions encompass the additional research needed to establish whether high folic acid or total folate intake contributes to disease risk. 1) Does UMFA affect biological pathways leading to adverse health effects? 2) Does elevated folate status resulting from any form of folate intake affect vitamin B-12 function and its roles in sustaining health? 3) Does elevated folate intake, regardless of form, affect biological pathways leading to adverse health effects other than those linked to vitamin B-12 function? This article summarizes the proceedings of an August 2019 NIH expert workshop focused on addressing these research areas.


Assuntos
Ácido Fólico/administração & dosagem , Adolescente , Adulto , Criança , Pré-Escolar , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Humanos , Pessoa de Meia-Idade , Estados Unidos
2.
J Nutr ; 150(Suppl 1): 2506S-2517S, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33000152

RESUMO

The metabolism of sulfur-containing amino acids (SAAs) requires an orchestrated interplay among several dozen enzymes and transporters, and an adequate dietary intake of methionine (Met), cysteine (Cys), and B vitamins. Known human genetic disorders are due to defects in Met demethylation, homocysteine (Hcy) remethylation, or cobalamin and folate metabolism, in Hcy transsulfuration, and Cys and hydrogen sulfide (H2S) catabolism. These disorders may manifest between the newborn period and late adulthood by a combination of neuropsychiatric abnormalities, thromboembolism, megaloblastic anemia, hepatopathy, myopathy, and bone and connective tissue abnormalities. Biochemical features include metabolite deficiencies (e.g. Met, S-adenosylmethionine (AdoMet), intermediates in 1-carbon metabolism, Cys, or glutathione) and/or their accumulation (e.g. S-adenosylhomocysteine, Hcy, H2S, or sulfite). Treatment should be started as early as possible and may include a low-protein/low-Met diet with Cys-enriched amino acid supplements, pharmacological doses of B vitamins, betaine to stimulate Hcy remethylation, the provision of N-acetylcysteine or AdoMet, or experimental approaches such as liver transplantation or enzyme replacement therapy. In several disorders, patients are exposed to long-term markedly elevated Met concentrations. Although these conditions may inform on Met toxicity, interpretation is difficult due to the presence of additional metabolic changes. Two disorders seem to exhibit Met-associated toxicity in the brain. An increased risk of demyelination in patients with Met adenosyltransferase I/III (MATI/III) deficiency due to biallelic mutations in the MATIA gene has been attributed to very high blood Met concentrations (typically >800 µmol/L) and possibly also to decreased liver AdoMet synthesis. An excessively high Met concentration in some patients with cystathionine ß-synthase deficiency has been associated with encephalopathy and brain edema, and direct toxicity of Met has been postulated. In summary, studies in patients with various disorders of SAA metabolism showed complex metabolic changes with distant cellular consequences, most of which are not attributable to direct Met toxicity.


Assuntos
Aminoácidos Sulfúricos/metabolismo , Cisteína/metabolismo , Homocisteína/metabolismo , Doenças Metabólicas/genética , Metionina/metabolismo , Compostos de Enxofre/metabolismo , Enxofre/metabolismo , Animais , Encefalopatias/etiologia , Encefalopatias/metabolismo , Glutationa/metabolismo , Homocistinúria/etiologia , Homocistinúria/metabolismo , Humanos , Sulfeto de Hidrogênio/metabolismo , Fígado/metabolismo , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Doenças Metabólicas/terapia , Erros Inatos do Metabolismo/patologia , Erros Inatos do Metabolismo/terapia , Metionina Adenosiltransferase/metabolismo , Metilação , S-Adenosilmetionina/metabolismo , Sulfitos/metabolismo
3.
Gut Microbes ; 10(6): 654-662, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31062653

RESUMO

Vitamin B12 is a critical nutrient for humans as well as microbes. Due to saturable uptake, high dose oral B12 supplements are largely unabsorbed and reach the distal gut where they are available to interact with the microbiota. The aim of this study was to determine if oral B12 supplementation in mice alters 1) the concentration of B12 and related corrinoids in the distal gut, 2) the fecal microbiome, 3) short chain fatty acids (SCFA), and 4) susceptibility to experimental colitis. C57BL/6 mice (up to 24 animals/group) were supplemented with oral 3.94 µg/ml cyanocobalamin (B12), a dose selected to approximate a single 5 mg supplement for a human. Active vitamin B12 (cobalamin), and four B12-analogues ([ADE]CN-Cba, [2Me-ADE]CN-Cba, [2MeS-ADE]CN-Cba, CN-Cbi) were analyzed in cecal and fecal contents using liquid chromatography/mass spectrometry (LC/MS), in parallel with evaluation of fecal microbiota, cecal SCFA, and susceptibility to dextran sodium sulfate (DSS) colitis. At baseline, active B12 was a minor constituent of overall cecal (0.86%) and fecal (0.44%) corrinoid. Oral B12 supplementation increased active B12 at distal sites by >130-fold (cecal B12 increased from 0.08 to 10.60 ng/mg, fecal B12 increased from 0.06 to 7.81 ng/ml) and reduced microbe-derived fecal corrinoid analogues ([ADE]CN-Cba, [2Me-ADE]CN-Cba, [2MeS-ADE]CN-Cba). Oral B12 had no effect on cecal SCFA. Microbial diversity was unaffected by this intervention, however a selective decrease in Bacteroides was observed with B12 treatment. Lastly, no difference in markers of DSS-induced colitis were detected with B12 treatment.


Assuntos
Bacteroides/efeitos dos fármacos , Corrinoides/análise , Suplementos Nutricionais/análise , Vitamina B 12/administração & dosagem , Complexo Vitamínico B/administração & dosagem , Administração Oral , Animais , Bacteroides/crescimento & desenvolvimento , Ceco/química , Colite/induzido quimicamente , Colite/dietoterapia , Sulfato de Dextrana/toxicidade , Ácidos Graxos Voláteis/análise , Fezes/química , Fezes/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Vitamina B 12/farmacologia , Complexo Vitamínico B/farmacologia
4.
J Inherit Metab Dis ; 42(3): 424-437, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30873612

RESUMO

STUDY OBJECTIVE: A phase 1/2 clinical trial was performed in individuals with cystathionine ß synthase (CBS) deficient homocystinuria with aims to: (a) assess pharmacokinetics and safety of taurine therapy, (b) evaluate oxidative stress, inflammation, and vascular function in CBS deficiency, and (c) evaluate the impact of short-term taurine treatment. METHODS: Individuals with pyridoxine-nonresponsive CBS deficiency with homocysteine >50 µM, without inflammatory disorder or on antioxidant therapy were enrolled. Biomarkers of oxidative stress and inflammation, endothelial function (brachial artery flow-mediated dilation [FMD]), and disease-related metabolites obtained at baseline were compared to normal values. While maintaining current treatment, patients were treated with 75 mg/kg taurine twice daily, and treatment response assessed after 4 hours and 4 days. RESULTS: Fourteen patients (8-35 years; 8 males, 6 females) were enrolled with baseline homocysteine levels 161 ± 67 µM. The study found high-dose taurine to be safe when excluding preexisting hypertriglyceridemia. Taurine pharmacokinetics showed a rapid peak level returning to near normal levels at 12 hours, but had slow accumulation and elevated predosing levels after 4 days of treatment. Only a single parameter of oxidative stress, 2,3-dinor-8-isoprostaglandin-F2α, was elevated at baseline, with no elevated inflammatory parameters, and no change in FMD values overall. Taurine had no effect on any of these parameters. However, the effect of taurine was strongly related to pretreatment FMD values; and taurine significantly improved FMD in the subset of individuals with pretreatment FMD values <10% and in individuals with homocysteine levels >125 µM, pertinent to endothelial function. CONCLUSION: Taurine improves endothelial function in CBS-deficient homocystinuria in patients with preexisting reduced function.


Assuntos
Biomarcadores/metabolismo , Cistationina beta-Sintase/metabolismo , Homocistinúria/tratamento farmacológico , Taurina/farmacocinética , Taurina/uso terapêutico , Adolescente , Adulto , Artéria Braquial/efeitos dos fármacos , Criança , Cistationina beta-Sintase/deficiência , Feminino , Homocisteína/metabolismo , Homocistinúria/genética , Humanos , Inflamação/tratamento farmacológico , Masculino , Estresse Oxidativo/efeitos dos fármacos , Estados Unidos , Adulto Jovem
5.
FASEB J ; 33(5): 6339-6353, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30768359

RESUMO

Classical cystathionine ß-synthase-deficient homocystinuria (HCU) is a life-threatening inborn error of sulfur metabolism. Treatment for pyridoxine-nonresponsive HCU involves lowering homocysteine (Hcy) with a methionine (Met)-restricted diet and betaine supplementation. Betaine treatment efficacy diminishes significantly over time due to impairment of betaine-Hcy S-methyltransferase (BHMT) function. Little is known regarding the regulation of BHMT in HCU. Using a betaine-responsive preclinical mouse model of HCU, we observed that this condition induces a 75% repression of BHMT mRNA, protein and enzyme activity, and significant depletion of hepatic betaine levels. BHMT repression was proportional to plasma Hcy levels but was not observed in mouse models of homocystinuria due to either methylenetetrahydrofolate reductase or Met synthase deficiency. Both Met supplementation and chemically induced glutathione depletion exacerbated hepatic BHMT repression in HCU mice but not wild-type (WT) controls. Conversely, cysteine treatment normalized hepatic BHMT expression in HCU mice but had no effect in WT control animals. Taurine treatment induced BHMT expression in HCU mice by 5-fold and restored maximal lowering of Hcy levels during long-term betaine treatment with a concomitant normalization of inflammatory cytokine expression and a significantly improved coagulative phenotype. Collectively, our findings indicate that adjuvantial taurine treatment has the potential to significantly improve clinical outcomes in HCU.-Maclean, K. N., Jiang, H, Phinney, W. N., Keating, A. K., Hurt, K. J., Stabler, S. P. Taurine alleviates repression of betaine-homocysteine S-methyltransferase and significantly improves the efficacy of long-term betaine treatment in a mouse model of cystathionine ß-synthase-deficient homocystinuria.


Assuntos
Betaína-Homocisteína S-Metiltransferase/metabolismo , Betaína/farmacologia , Homocistinúria , Fígado/enzimologia , Taurina/farmacologia , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Modelos Animais de Doenças , Homocistinúria/tratamento farmacológico , Homocistinúria/genética , Homocistinúria/metabolismo , Homocistinúria/patologia , Humanos , Fígado/patologia , Camundongos , Camundongos Knockout
6.
Nat Rev Dis Primers ; 3: 17040, 2017 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-28660890

RESUMO

Vitamin B12 (B12; also known as cobalamin) is a B vitamin that has an important role in cellular metabolism, especially in DNA synthesis, methylation and mitochondrial metabolism. Clinical B12 deficiency with classic haematological and neurological manifestations is relatively uncommon. However, subclinical deficiency affects between 2.5% and 26% of the general population depending on the definition used, although the clinical relevance is unclear. B12 deficiency can affect individuals at all ages, but most particularly elderly individuals. Infants, children, adolescents and women of reproductive age are also at high risk of deficiency in populations where dietary intake of B12-containing animal-derived foods is restricted. Deficiency is caused by either inadequate intake, inadequate bioavailability or malabsorption. Disruption of B12 transport in the blood, or impaired cellular uptake or metabolism causes an intracellular deficiency. Diagnostic biomarkers for B12 status include decreased levels of circulating total B12 and transcobalamin-bound B12, and abnormally increased levels of homocysteine and methylmalonic acid. However, the exact cut-offs to classify clinical and subclinical deficiency remain debated. Management depends on B12 supplementation, either via high-dose oral routes or via parenteral administration. This Primer describes the current knowledge surrounding B12 deficiency, and highlights improvements in diagnostic methods as well as shifting concepts about the prevalence, causes and manifestations of B12 deficiency.


Assuntos
Deficiência de Vitamina B 12/diagnóstico , Deficiência de Vitamina B 12/epidemiologia , Vitamina B 12/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Criança , Pré-Escolar , Feminino , Homocisteína/metabolismo , Humanos , Masculino , Ácido Metilmalônico/metabolismo , Pessoa de Meia-Idade , Prevalência , Fatores de Risco , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo , Deficiência de Vitamina B 12/complicações , Deficiência de Vitamina B 12/metabolismo , Adulto Jovem
7.
J Nutr Biochem ; 26(9): 903-11, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26025328

RESUMO

Demand for the vital nutrient choline is high during lactation; however, few studies have examined choline metabolism and requirements in this reproductive state. The present study sought to discern the effects of lactation and varied choline intake on maternal biomarkers of choline metabolism and breast milk choline content. Lactating (n=28) and control (n=21) women were randomized to 480 or 930 mg choline/day for 10-12 weeks as part of a controlled feeding study. During the last 4-6 weeks, 20% of the total choline intake was provided as an isotopically labeled choline tracer (methyl-d9-choline). Blood, urine and breast milk samples were collected for choline metabolite quantification, enrichment measurements, and gene expression analysis of choline metabolic genes. Lactating (vs. control) women exhibited higher (P < .001) plasma choline concentrations but lower (P ≤ .002) urinary excretion of choline metabolites, decreased use of choline as a methyl donor (e.g., lower enrichment of d6-dimethylglycine, P ≤ .08) and lower (P ≤ .02) leukocyte expression of most choline-metabolizing genes. A higher choline intake during lactation differentially influenced breast milk d9- vs. d3-choline metabolite enrichment. Increases (P ≤ .03) were detected among the d3-metabolites, which are generated endogenously via the hepatic phosphatidylethanolamine N-methyltransferase (PEMT), but not among the d9-metabolites generated from intact exogenous choline. These data suggest that lactation induces metabolic adaptations that increase the supply of intact choline to the mammary epithelium, and that extra maternal choline enhances breast milk choline content by increasing supply of PEMT-derived choline metabolites. This trial was registered at clinicaltrials.gov as NCT01127022.


Assuntos
Colina/administração & dosagem , Suplementos Nutricionais , Lactação/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Leite Humano/química , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Adulto , Biomarcadores/sangue , Biomarcadores/urina , Colina/análise , Colina/sangue , Colina/metabolismo , Estudos de Coortes , Deutério , Indução Enzimática , Feminino , Humanos , Lactação/sangue , Lactação/urina , Leucócitos/enzimologia , Leucócitos/metabolismo , Fígado/enzimologia , Fígado/metabolismo , Glândulas Mamárias Humanas/enzimologia , Glândulas Mamárias Humanas/metabolismo , Leite Humano/metabolismo , New York , Fosfatidiletanolamina N-Metiltransferase/química , Fosfatidiletanolamina N-Metiltransferase/genética , RNA Mensageiro/metabolismo , Recomendações Nutricionais , Adulto Jovem
8.
J Nutr ; 145(7): 1507-14, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25995278

RESUMO

BACKGROUND: Limited data are available from controlled studies on biomarkers of maternal vitamin B-12 status. OBJECTIVE: We sought to quantify the effects of pregnancy and lactation on the vitamin B-12 status response to a known and highly controlled vitamin B-12 intake. METHODS: As part of a 10-12 wk feeding trial, pregnant (26-29 wk gestation; n = 26), lactating (5 wk postpartum; n = 28), and control (nonpregnant, nonlactating; n = 21) women consumed vitamin B-12 amounts of ∼8.6 µg/d [mixed diet (∼6 µg/d) plus a prenatal multivitamin supplement (2.6 µg/d)]. Serum vitamin B-12, holotranscobalamin (bioactive form of vitamin B-12), methylmalonic acid (MMA), and homocysteine were measured at baseline and study-end. RESULTS: All participants achieved adequate vitamin B-12 status in response to the study dose. Compared with control women, pregnant women had lower serum vitamin B-12 (-21%; P = 0.02) at study-end, whereas lactating women had higher (P = 0.04) serum vitamin B-12 throughout the study (+26% at study-end). Consumption of the study vitamin B-12 dose increased serum holotranscobalamin in all reproductive groups (+16-42%; P ≤ 0.009). At study-end, pregnant (vs. control) women had a higher holotranscobalamin-to-vitamin B-12 ratio (P = 0.04) with ∼30% (vs. 20%) of total vitamin B-12 in the bioactive form. Serum MMA increased during pregnancy (+50%; P < 0.001) but did not differ by reproductive state at study-end. Serum homocysteine increased in pregnant women (+15%; P = 0.009) but decreased in control and lactating women (-16-17%; P < 0.001). Despite these changes, pregnant women had ∼20% lower serum homocysteine than the other 2 groups at study-end (P ≤ 0.02). CONCLUSION: Pregnancy and lactation alter vitamin B-12 status in a manner consistent with enhanced vitamin B-12 supply to the child. Consumption of the study vitamin B-12 dose (∼3 times the RDA) increased the bioactive form of vitamin B-12, suggesting that women in these reproductive states may benefit from vitamin B-12 intakes exceeding current recommendations. This trial was registered at clinicaltrials.gov as NCT01127022.


Assuntos
Ingestão de Energia , Micronutrientes/administração & dosagem , Vitamina B 12/sangue , Adulto , Biomarcadores/sangue , Aleitamento Materno , Colina/administração & dosagem , Colina/sangue , Suplementos Nutricionais , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Homocisteína/sangue , Homocisteína/urina , Humanos , Lactação/sangue , Ácido Metilmalônico/sangue , Período Pós-Parto , Gravidez , Recomendações Nutricionais , Vitamina B 12/administração & dosagem , Adulto Jovem
9.
FASEB J ; 28(9): 4044-54, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24891521

RESUMO

Cystathionine ß-synthase-deficient homocystinuria (HCU) is a serious life-threatening inborn error of sulfur metabolism with poorly understood pathogenic mechanisms. We investigated the effect of HCU on hepatic cysteine oxidation in a transgenic mouse model of the disease. Cysteine dioxygenase (CDO) protein levels were 90% repressed without any change in mRNA levels. Cysteinesulfinic acid decarboxylase (CSAD) was induced at both the mRNA (8-fold) and protein (15-fold) levels. Cysteine supplementation normalized CDO protein levels without reversing the induction of CSAD. Regulatory changes in CDO and CSAD expression were proportional to homocysteine elevation, indicating a possible threshold effect. Hepatic and blood taurine levels in HCU animals were decreased by 21 and 35%, respectively, and normalized by cysteine supplementation. Expression of the cytoplasmic (GOT1) and mitochondrial (GOT2) isoforms of glutamic-oxaloacetic transaminase were repressed in HCU animals by 86 and 30%, respectively. HCU induced regulatory changes in CSAD, CDO, and GOT1 expression were normalized by taurine supplementation, indicating that cysteine is not the only sulfur compound that regulates hepatic cysteine oxidation. Collectively, our results indicate that HCU induces significant alterations of sulfur metabolism with the potential to contribute to pathogenesis and that cysteine and taurine have the potential to serve as adjunctive treatments in this disease.


Assuntos
Cistationina beta-Sintase/fisiologia , Cisteína/metabolismo , Homocistinúria/fisiopatologia , Fígado/metabolismo , Enxofre/metabolismo , Taurina/farmacologia , Animais , Western Blotting , Carboxiliases/genética , Carboxiliases/metabolismo , Cisteína/química , Cisteína Dioxigenase/genética , Cisteína Dioxigenase/metabolismo , Suplementos Nutricionais , Feminino , Homocistinúria/tratamento farmacológico , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Oxirredução , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Am J Clin Nutr ; 98(6): 1459-67, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24132975

RESUMO

BACKGROUND: Although biomarkers of choline metabolism are altered by pregnancy, little is known about the influence of human pregnancy on the dynamics of choline-related metabolic processes. OBJECTIVE: This study used stable isotope methodology to examine the effects of pregnancy on choline partitioning and the metabolic activity of choline-related pathways. DESIGN: Healthy third-trimester pregnant (n = 26; initially week 27 of gestation) and nonpregnant (n = 21) women consumed 22% of their total choline intake (480 or 930 mg/d) as methyl-d9-choline for the final 6 wk of a 12-wk feeding study. RESULTS: Plasma d9-betaine:d9-phosphatidylcholine (PC) was lower (P ≤ 0.04) in pregnant than in nonpregnant women, suggesting greater partitioning of choline into the cytidine diphosphate-choline (CDP-choline) PC biosynthetic pathway relative to betaine synthesis during pregnancy. Pregnant women also used more choline-derived methyl groups for PC synthesis via phosphatidylethanolamine N-methyltransferase (PEMT) as indicated by comparable increases in PEMT-PC enrichment in pregnant and nonpregnant women despite unequal (pregnant > nonpregnant; P < 0.001) PC pool sizes. Pregnancy enhanced the hydrolysis of PEMT-PC to free choline as shown by greater (P < 0.001) plasma d3-choline:d3-PC. Notably, d3-PC enrichment increased (P ≤ 0.011) incrementally from maternal to placental to fetal compartments, signifying the selective transfer of PEMT-PC to the fetus. CONCLUSIONS: The enhanced use of choline for PC production via both the CDP-choline and PEMT pathways shows the substantial demand for choline during late pregnancy. Selective partitioning of PEMT-PC to the fetal compartment may imply a unique requirement of PEMT-PC by the developing fetus.


Assuntos
Colina/metabolismo , Dieta , Suplementos Nutricionais , Fenômenos Fisiológicos da Nutrição Materna , Troca Materno-Fetal , Gravidez/metabolismo , Adulto , Betaína/sangue , Colina/administração & dosagem , Colina/análogos & derivados , Colina/sangue , Deutério , Feminino , Sangue Fetal , Humanos , Hidrólise , Metilação , Fosfatidilcolinas/sangue , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Placenta/metabolismo , Gravidez/sangue , Terceiro Trimestre da Gravidez , Adulto Jovem
11.
J Nutr ; 143(7): 1028-35, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23700346

RESUMO

Impaired folate-mediated one-carbon metabolism (OCM) has emerged as a risk factor for several diseases associated with age-related cognitive decline, but the underlying mechanisms remain unknown and thus hinder the identification of subpopulations most vulnerable to OCM disruption. Here we investigated the role of serine hydroxymethyltransferase 1 (SHMT1), a folate-dependent enzyme regulating de novo thymidylate biosynthesis, in influencing neuronal and cognitive function in the adult mouse. We observed Shmt1 expression in the hippocampus, including the granule cell layer of the dentate gyrus (DG), and examined hippocampal neurogenesis and hippocampal-dependent fear conditioning in mice deficient for Shmt1. We used a 3 × 3 design in which adult male Shmt1(+/+), Shmt1(+/-), and Shmt1(-/-) mice were fed folic acid control (2 mg/kg), folic acid-deficient (0 mg/kg), or folic acid-supplemented (8 mg/kg) diets from weaning through the duration of the study. Proliferation within the DG was elevated by 70% in Shmt1(+/-) mice, yet the number of newborn mature neurons was reduced by 98% compared with that in Shmt1(+/+) mice. Concomitant with these alterations, Shmt1(+/-) mice showed a 45% reduction in mnemonic recall during trace fear conditioning. Dietary folate manipulations alone did not influence neural outcomes. Together, these data identify SHMT1 as one of the first enzymes within the OCM pathway to regulate neuronal and cognitive profiles and implicate impaired thymidylate biosynthesis in the etiology of folate-related neuropathogenesis.


Assuntos
Glicina Hidroximetiltransferase/genética , Hipocampo/patologia , Memória/fisiologia , Neurogênese/genética , Animais , Proliferação de Células , Giro Denteado/metabolismo , Medo/fisiologia , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Deficiência de Ácido Fólico/patologia , Regulação da Expressão Gênica , Glicina Hidroximetiltransferase/metabolismo , Hipocampo/metabolismo , Homocisteína/metabolismo , Hibridização In Situ , Masculino , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Fatores de Risco , Timidina Monofosfato/biossíntese
12.
PLoS One ; 8(2): e56253, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437105

RESUMO

Atherosclerosis is an inflammatory condition of the arterial wall mediated by cells of both innate and adaptive immunity. T lymphocytes play an important role in orchestrating the pathogenic immune response involved in the acceleration of atherosclerosis. Previously, we have shown that a prenatal methyl-donor supplementation diet (MS), when fed to dams during pregnancy and lactation, decreased the T cell-mediated pro-inflammatory cytokine and chemokine response in F1 mice. In the current study, we report feeding Apolipoprotein E (ApoE(-/-)) deficient dams with the MS diet during pregnancy reduces atherosclerotic plaques in F1 mice that were fed high fat diet (HFD) after weaning. F1 mice from dams on the MS diet exhibited increased global T cell DNA methylation. T-cell chemokines and their receptors (in particular CCR2, CCR5, and CXCR3) play important roles in the inflammatory cell recruitment to vascular lesions. MS diet significantly reduced Ccr2 mRNA and protein expression in CD3+ T cells but not in CD11b+ monocytes in MS F1 mice relative to controls. F1 litter size, HFD consumption, body weight, and body fat were similar between control and MS diet groups. Moreover, serum thiol metabolite levels were similar between the two groups. However, MS diet is associated with significantly higher serum HDL and lower LDL+VLDL levels in comparison to F1 mice from dams on the control diet. Inflammatory cytokines (IL-17, TNF-α, IL-6) were also lower in MS F1 mice serum and conditioned media from T-cell culture. Altogether, these data suggest that the MS diet ameliorates development of atherosclerosis by inhibiting the T-cell Ccr2 expression, reducing inflammatory cytokines production and increasing serum HDL:LDL ratio.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Cruzamentos Genéticos , Dieta , Suplementos Nutricionais , Animais , Aorta/patologia , Apolipoproteínas E/metabolismo , Aterosclerose/sangue , Aterosclerose/patologia , Composição Corporal , Colesterol/biossíntese , Dieta Hiperlipídica , Comportamento Alimentar , Feminino , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Lipoproteínas/sangue , Tamanho da Ninhada de Vivíparos , Fígado/metabolismo , Fígado/patologia , Ativação Linfocitária/imunologia , Masculino , Metilação , Camundongos , Monócitos/metabolismo , Receptores CCR2/metabolismo , Compostos de Sulfidrila/metabolismo , Linfócitos T/metabolismo , Aumento de Peso
13.
Mol Genet Metab ; 107(1-2): 55-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22633282

RESUMO

Classical homocystinuria (HCU) is caused by mutations in cystathionine beta-synthase (CBS) which, if untreated, typically results in cognitive impairment, thromboembolic complications and connective tissue disturbances. Paraoxonase-1 (PON1) and apolipoprotein apoA-I are both synthesized in the liver and contribute to much of the cardioprotective effects of high density lipoprotein. Additionally, apoA-I exerts significant neuro-protective effects that act to preserve cognition. Previous work in a Cbs null mouse model that incurs significant liver injury, reported that HCU dramatically decreases PON1 expression. Conflicting reports exist in the literature concerning the relative influence of homocysteine and cysteine upon apoA-I expression. We investigated expression of PON1 and apoA-I in the presence and absence of homocysteine lowering therapy, in both the HO mouse model of HCU and human subjects with this disorder. We observed no significant change in plasma PON1 paraoxonase activity in either mice or humans with HCU indicating that this enzyme is unlikely to contribute to the cardiovascular sequelae of HCU. Plasma levels of apoA-I were unchanged in mice with mildly elevated homocysteine due to CBS deficiency but were significantly diminished in both mice and humans with HCU. Subsequent experiments revealed that HCU acts to dramatically decrease apoA-I levels in the brain. Cysteine supplementation in HO mice had no discernible effect on plasma levels of apoA-I while treatment to lower homocysteine normalized plasma levels of this lipoprotein in both HO mice and humans with HCU. Our results indicate that plasma apoA-I levels in HCU are inversely related to homocysteine and are consistent with a plausible role for decreased expression of apoA-I as a contributory factor for both cardiovascular disease and cognitive impairment in HCU.


Assuntos
Apolipoproteína A-I/metabolismo , Apolipoproteínas A/metabolismo , Arildialquilfosfatase/metabolismo , Homocistinúria/metabolismo , Adolescente , Adulto , Animais , Apolipoproteína A-I/sangue , Apolipoproteínas A/sangue , Arildialquilfosfatase/sangue , Betaína/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Hidrolases de Éster Carboxílico/sangue , Criança , Pré-Escolar , Suplementos Nutricionais , Modelos Animais de Doenças , Homocisteína/sangue , Homocistinúria/dietoterapia , Homocistinúria/tratamento farmacológico , Humanos , Lipotrópicos/uso terapêutico , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Adulto Jovem
14.
Am J Clin Nutr ; 95(4): 882-91, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22378735

RESUMO

BACKGROUND: MTHFD1 encodes C1-tetrahydrofolate synthase, which is a folate-dependent enzyme that catalyzes the formation and interconversion of folate-activated one-carbon groups for nucleotide biosynthesis and cellular methylation. A polymorphism in MTHFD1 (1958G→A) impairs enzymatic activity and is associated with increased risk of adverse pregnancy outcomes, but the mechanisms are unknown. OBJECTIVE: The objective of this study was to determine whether disruption of the embryonic or maternal Mthfd1 gene or both interacts with impaired folate and choline status to affect neural tube closure, fetal growth, and fertility in mice and to investigate the underlying metabolic disruptions. DESIGN: Dams with a gene-trapped (gt) allele in Mthfd1 and wild-type dams were fed a control or folate- and choline-deficient AIN93G diet (Dyets Inc). Litters were examined for gross morphologic defects, crown-rump length, and resorptions. Folate status and amounts of folate-related metabolites were determined in pregnant dams. RESULTS: Reduced folate and choline status resulted in severe fetal growth restriction (FGR) and impaired fertility in litters harvested from Mthfd1(gt/+) dams, but embryonic Mthfd1(gt/+) genotype did not affect fetal growth. Gestational supplementation of Mthfd1(gt/+) dams with hypoxanthine increased FGR frequency and caused occasional neural tube defects (NTDs) in Mthfd1(gt/+) embryos. Mthfd1(gt/+) dams exhibited lower red blood cell folate and plasma methionine concentrations than did wild-type dams. CONCLUSIONS: Maternal Mthfd1(gt/+) genotype impairs fetal growth but does not cause NTDs when dams are maintained on a folate- and choline-deficient diet. Mthfd1(gt/+) mice exhibit a spectrum of adverse reproductive outcomes previously attributed to the human MTHFD1 1958G→A polymorphism. Mthfd1 heterozygosity impairs folate status in pregnant mice but does not significantly affect homocysteine metabolism.


Assuntos
Aminoidrolases/deficiência , Retardo do Crescimento Fetal/genética , Ácido Fólico/metabolismo , Formiato-Tetra-Hidrofolato Ligase/deficiência , Homocisteína/metabolismo , Metilenotetra-Hidrofolato Desidrogenase (NADP)/deficiência , Complexos Multienzimáticos/deficiência , Aminoidrolases/genética , Aminoidrolases/metabolismo , Animais , Colina/metabolismo , Deficiência de Colina/genética , Deficiência de Colina/metabolismo , Cruzamentos Genéticos , Modelos Animais de Doenças , Perda do Embrião/genética , Perda do Embrião/metabolismo , Feminino , Retardo do Crescimento Fetal/metabolismo , Ácido Fólico/sangue , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/metabolismo , Formiato-Tetra-Hidrofolato Ligase/genética , Formiato-Tetra-Hidrofolato Ligase/metabolismo , Genes Letais , Heterozigoto , Homocisteína/sangue , Hipoxantina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Metilenotetra-Hidrofolato Desidrogenase (NADP)/genética , Metilenotetra-Hidrofolato Desidrogenase (NADP)/metabolismo , Camundongos , Camundongos Mutantes , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutagênese Insercional , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Gravidez
15.
J Gerontol A Biol Sci Med Sci ; 67(1): 100-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21896502

RESUMO

BACKGROUND: Anemia has been associated with increased physical and financial costs and occurs more frequently in older individuals. Therefore, the primary objectives of this study were to examine the prevalence and possible predictors of anemia in the very old. METHODS: Hemoglobin was used to identify those with anemia in a group of centenarians and near centenarians (98+, n = 185) and octogenarians (n = 69), who were recruited as part of the population-based multidisciplinary Georgia Centenarian Study. Blood markers, including ferritin, vitamin B12, red blood cell folate, methylmalonic acid, creatinine, and C-reactive protein, demographic variables, and medication and/or supplement usage were used to determine possible predictors of anemia. RESULTS: The prevalence of anemia was 26.2% in octogenarians and 52.1% in centenarians. Low serum albumin (<3.6 g/dL) and decreased estimated glomerular filtration rate (<45 mL/min/m(2)) were predictors of anemia in centenarians. CONCLUSIONS: Anemia is a major health issue, particularly as people age. Because of the high prevalence of anemia in older individuals, awareness of the predictors associated with anemia becomes increasingly important so as to reduce the negative consequences associated with it and allow for the identification of steps that can be taken to correct anemia, including managing chronic disease.


Assuntos
Anemia/epidemiologia , Idoso de 80 Anos ou mais , Anemia/diagnóstico , Proteína C-Reativa/análise , Doença Crônica , Creatinina/sangue , Suplementos Nutricionais , Eritrócitos/química , Feminino , Ferritinas/sangue , Ácido Fólico/análise , Georgia/epidemiologia , Taxa de Filtração Glomerular , Hemoglobinas/análise , Humanos , Masculino , Ácido Metilmalônico/sangue , Prevalência , Albumina Sérica/análise , Vitamina B 12/sangue
16.
Mol Genet Metab ; 105(3): 395-403, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22192524

RESUMO

Classical homocystinuria (HCU) is caused by deficiency of cystathionine ß-synthase and is characterized by connective tissue disturbances, mental retardation and cardiovascular disease. Treatment for pyridoxine non-responsive HCU typically involves lowering homocysteine levels with a methionine-restricted diet and dietary supplementation with betaine. Compliance with the methionine-restricted diet is difficult and often poor. Investigating optimization of the efficacy of long-term betaine treatment in isolation from a methionine-restricted diet is precluded by ethical considerations regarding patient risk. The HO mouse model of HCU developed in our laboratory, exhibits constitutive expression of multiple pro-inflammatory cytokines and a hypercoagulative phenotype both of which respond to short-term betaine treatment. Investigation of the effects of long-term betaine treatment in the absence of methionine-restriction in HO HCU mice revealed that the ability of betaine treatment to lower homocysteine diminished significantly over time. Plasma metabolite analysis indicated that this effect was due at least in part, to reduced betaine-homocysteine S-methyltransferase (BHMT) mediated remethylation of homocysteine. Western blotting analysis revealed that BHMT protein levels are significantly repressed in untreated HCU mice but are significantly induced in the presence of betaine treatment. The observed increase in plasma homocysteine during prolonged betaine treatment was accompanied by a significant increase in the plasma levels of TNF-alpha and IL-1beta and reversion to a hypercoagulative phenotype. Our findings are consistent with a relatively sharp threshold effect between severely elevated plasma homocysteine and thrombotic risk in HCU and indicate that the HO mouse model can serve as a useful tool for both testing novel treatment strategies and examining the optimal timing and dosing of betaine treatment with a view toward optimizing clinical outcome.


Assuntos
Betaína-Homocisteína S-Metiltransferase/biossíntese , Betaína/uso terapêutico , Cistationina beta-Sintase/metabolismo , Homocisteína/sangue , Homocistinúria/tratamento farmacológico , Trombose/etiologia , Animais , Betaína/administração & dosagem , Betaína/farmacologia , Betaína-Homocisteína S-Metiltransferase/metabolismo , Coagulação Sanguínea , Cistationina beta-Sintase/deficiência , Dieta , Suplementos Nutricionais , Modelos Animais de Doenças , Homocisteína/metabolismo , Homocistinúria/sangue , Homocistinúria/genética , Interleucina-1beta/sangue , Masculino , Metionina , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Necrose Tumoral alfa/sangue
17.
Am J Clin Nutr ; 93(4): 789-98, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21346092

RESUMO

BACKGROUND: Folic acid supplementation prevents the occurrence and recurrence of neural tube defects (NTDs), but the causal metabolic pathways underlying folic acid-responsive NTDs have not been established. Serine hydroxymethyltransferase (SHMT1) partitions folate-derived one-carbon units to thymidylate biosynthesis at the expense of cellular methylation, and therefore SHMT1-deficient mice are a model to investigate the metabolic origin of folate-associated pathologies. OBJECTIVES: We examined whether genetic disruption of the Shmt1 gene in mice induces NTDs in response to maternal folate and choline deficiency and whether a corresponding disruption in de novo thymidylate biosynthesis underlies NTD pathogenesis. DESIGN: Shmt1 wild-type, Shmt1(+/-), and Shmt1(-/-) mice fed either folate- and choline-sufficient or folate- and choline-deficient diets were bred, and litters were examined for the presence of NTDs. Biomarkers of impaired folate metabolism were measured in the dams. In addition, the effect of Shmt1 disruption on NTD incidence was investigated in Pax3(Sp) mice, an established folate-responsive NTD mouse model. RESULTS: Shmt1(+/-) and Shmt1(-/-) embryos exhibited exencephaly in response to maternal folate and choline deficiency. Shmt1 disruption on the Pax3(Sp) background exacerbated NTD frequency and severity. Pax3 disruption impaired de novo thymidylate and purine biosynthesis and altered amounts of SHMT1 and thymidylate synthase protein. CONCLUSIONS: SHMT1 is the only folate-metabolizing enzyme that has been shown to affect neural tube closure in mice by directly inhibiting folate metabolism. These results provide evidence that disruption of Shmt1 expression causes NTDs by impairing thymidylate biosynthesis and shows that changes in the expression of genes that encode folate-dependent enzymes may be key determinates of NTD risk.


Assuntos
Deficiência de Ácido Fólico/complicações , Ácido Fólico/farmacologia , Glicina Hidroximetiltransferase/genética , Mutação , Defeitos do Tubo Neural/etiologia , Timidina Monofosfato/biossíntese , Complexo Vitamínico B/farmacologia , Animais , Biomarcadores/sangue , Colina/farmacologia , Deficiência de Colina/complicações , Modelos Animais de Doenças , Expressão Gênica , Glicina Hidroximetiltransferase/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Purinas/biossíntese , Índice de Gravidade de Doença , Timidilato Sintase/metabolismo
18.
Am J Clin Nutr ; 93(2): 348-55, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21123458

RESUMO

BACKGROUND: Homozygosity for the variant 677T allele in the methylenetetrahydrofolate reductase (MTHFR) gene increases the requirement for folate and may alter the metabolic use of choline. The choline adequate intake is 550 mg/d for men, although the metabolic consequences of consuming extra choline are unclear. OBJECTIVE: Deuterium-labeled choline (d9-choline) as tracer was used to determine the differential effects of the MTHFR C677T genotype and the effect of various choline intakes on the isotopic enrichment of choline derivatives in folate-compromised men. DESIGN: Mexican American men with the MTHFR 677CC or 677TT genotype consumed a diet providing 300 mg choline/d plus supplemental choline chloride for total choline intakes of 550 (n = 11; 4 with 677CC and 7 with 677TT) or 1100 (n = 12; 4 with 677CC and 8 with 677TT) mg/d for 12 wk. During the last 3 wk, 15% of the total choline intake was provided as d9-choline. RESULTS: Low but measurable enrichments of the choline metabolites were achieved, including that of d3-phosphatidylcholine (d3-PtdCho)--a metabolite produced in the de novo pathway via choline-derived methyl groups. Men with the MTHFR 677TT genotype had a higher urinary enrichment ratio of betaine to choline (P = 0.041), a higher urinary enrichment of sarcosine (P = 0.041), and a greater plasma enrichment ratio of d9-betaine to d9-PtdCho with the 1100 mg choline/d intake (P = 0.033). CONCLUSION: These data show for the first time in humans that choline itself is a source of methyl groups for de novo PtdCho biosynthesis and indicate that the MTHFR 677TT genotype favors the use of choline as a methyl donor.


Assuntos
Colina/metabolismo , Deficiência de Ácido Fólico/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Adolescente , Adulto , Betaína/sangue , Betaína/urina , Colina/administração & dosagem , Deutério/administração & dosagem , Deutério/metabolismo , Suplementos Nutricionais , Deficiência de Ácido Fólico/metabolismo , Genótipo , Humanos , Masculino , Metilação , Americanos Mexicanos , Pessoa de Meia-Idade , Fosfatidilcolinas/biossíntese , Sarcosina/urina , Coloração e Rotulagem , Adulto Jovem
19.
Free Radic Biol Med ; 47(8): 1147-53, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19616616

RESUMO

Lipoic acid is a disulfhydryl-containing compound used in clinical medicine and in experimental models as an antioxidant. We developed a stable isotope dilution capillary gas chromatography/mass spectrometry assay for lipoic acid. We assayed a panel of the metabolites of transmethylation and transsulfuration 30 min after injecting 100 mg/kg lipoic acid in a rat model. Lipoic acid values rose 1000-fold in serum and 10-fold in liver. A methylated metabolite of lipoic acid was also detected but not quantitated. Lipoic acid injection caused a massive increase in serum S-adenosylhomocysteine and marked depletion of liver S-adenosylmethionine. Serum total cysteine was depleted but liver cysteine and glutathione were maintained. Serum total homocysteine doubled, with increases also in cystathionine, N,N-dimethylglycine, and alpha-aminobutyric acid. In contrast, after injection of 2-mercaptoethane sulfonic acid, serum total cysteine and homocysteine were markedly depleted and there were no effects on serum S-adenosylmethionine or S-adenosylhomocysteine. We conclude that large doses of lipoic acid displace sulfhydryls from binding sites, resulting in depletion of serum cysteine, but also pose a methylation burden with severe depletion of liver S-adenosylmethionine and massive release of S-adenosylhomocysteine. These changes may have previously unrecognized deleterious effects that should be investigated in both human disease and experimental models.


Assuntos
Antioxidantes/farmacologia , Fígado/metabolismo , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo , Ácido Tióctico/farmacologia , Animais , Cisteína/sangue , Cisteína/deficiência , Cromatografia Gasosa-Espectrometria de Massas , Fígado/efeitos dos fármacos , Masculino , Metilação , Ratos , Ratos Sprague-Dawley , Ácido Tióctico/sangue
20.
Arch Anim Nutr ; 63(5): 379-88, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-26967796

RESUMO

Cobalamin analogues in samples of rumen fluid, duodenal and ileal digesta and faeces of lactating dairy cows collected during the course of two nutritional studies were identified using a recently developed liquid chromatography-mass spectrometry method in order to determine if changes in diet composition could alter their proportions. Only cobalamin and cobinamine were detected in feedstuffs; six supplementary analogues were detected in the gastrointestinal tract content. The proportion of analogues produced by the gastrointestinal microflora was higher than the vitamin itself. The mode of conservation of forage had no effect on the proportion of analogues in rumen fluid, duodenal digesta and faeces, whereas increasing metabolisable protein supply raised the proportion of analogues reaching the duodenum (p = 0.008). The proportion of analogues was higher in ileal than duodenal digesta, this observation probably indicates a preferential absorption of cobalamin in the small intestine. The present results showed that changes in diet composition could alter cobalamin synthesis in rumen.


Assuntos
Conteúdo Gastrointestinal/química , Vitamina B 12/análogos & derivados , Vitamina B 12/metabolismo , Ração Animal/análise , Animais , Bovinos , Cromatografia Líquida/veterinária , Dieta/veterinária , Duodeno/química , Fezes/química , Feminino , Íleo/química , Lactação , Espectrometria de Massas/veterinária , Rúmen/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA