Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 230
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Reproduction ; 161(4): 365-373, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33528379

RESUMO

Mechanistic target of rapamycin (MTOR) is essential for embryo development by acting as a nutrient sensor to regulate cell growth, proliferation and metabolism. Folate is required for normal embryonic development and it was recently reported that MTOR functions as a folate sensor. In this work, we tested the hypothesis that MTOR functions as a folate sensor in the embryo and its inhibition result in embryonic developmental delay affecting neural tube closure and that these effects can be rescued by folate supplementation. Administration of rapamycin (0.5 mg/kg) to rats during early organogenesis inhibited embryonic ribosomal protein S6, a downstream target of MTOR Complex1, markedly reduced embryonic folate incorporation (-84%, P < 0.01) and induced embryo developmental impairments, as shown by an increased resorption rate, reduced embryo somite number and delayed neural tube closure. These alterations were prevented by folic acid administered to the dams. Differently, although an increased rate of embryonic rotation defects was observed in the rapamycin-treated dams, this alteration was not prevented by maternal folic acid supplementation. In conclusion, MTOR inhibition during organogenesis in the rat resulted in decreased folate levels in the embryo, increased embryo resorption rate and impaired embryo development. These data suggest that MTOR signaling influences embryo folate availability, possibly by regulating the transfer of folate across the maternal-embryonic interface.


Assuntos
Embrião de Mamíferos/patologia , Desenvolvimento Embrionário , Deficiência de Ácido Fólico/fisiopatologia , Ácido Fólico/metabolismo , Organogênese , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Embrião de Mamíferos/metabolismo , Feminino , Deficiência de Ácido Fólico/metabolismo , Masculino , Gravidez , Ratos , Ratos Wistar , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
2.
FASEB J ; 33(4): 4688-4702, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30592646

RESUMO

Folate deficiency in early development leads to disturbance in multiple processes, including neurogenesis during which fibroblast growth factor (FGF) pathway is one of the crucial pathways. Whether folic acid (FA) directly affects FGF pathways to influence neurodevelopment and the possible mechanism remains unclear. In this study, we presented evidence that in human FA-insufficient encephalocele, the FGF pathway was interfered. Furthermore, in Brachyury knockout mice devoid of such T-box transcription factors regulating embryonic neuromesodermal bipotency and a key component of FGF pathway, change in expression of Brachyury downstream targets, activator Fgf8 and suppressor dual specificity phosphatase 6 was detected, along with the reduction in expression of other key FGF pathway genes. By using a FA-deficient cell model, we further demonstrated that decrease in Brachyury expression was through alteration in hypermethylation at the Brachyury promoter region under FA deficiency conditions, and suppression of Brachyury promoted the inactivation of the FGF pathway. Correspondingly, FA supplementation partially reverses the effects seen in FA-deficient embryoid bodies. Lastly, in mice with maternal folate-deficient diets, aberrant FGF pathway activity was found in fetal brain dysplasia. Taken together, our findings highlight the effect of FA on FGF pathways during neurogenesis, and the mechanism may be due to the low expression of Brachyury gene via hypermethylation under FA-insufficient conditions.-Chang, S., Lu, X., Wang, S., Wang, Z., Huo, J., Huang, J., Shangguan, S., Li, S., Zou, J., Bao, Y., Guo, J., Wang, F., Niu, B., Zhang, T., Qiu, Z., Wu, J., Wang, L. The effect of folic acid deficiency on FGF pathway via Brachyury regulation in neural tube defects.


Assuntos
Proteínas Fetais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/uso terapêutico , Defeitos do Tubo Neural/tratamento farmacológico , Defeitos do Tubo Neural/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Encefalocele/metabolismo , Feminino , Deficiência de Ácido Fólico/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Sulfitos/farmacologia
3.
Dev Biol ; 444 Suppl 1: S193-S201, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30098999

RESUMO

Folate deficiency has been known to contribute to neural tube and neural crest defects, but why these tissues are particularly affected, and which are the molecular mechanisms involved in those abnormalities are important human health questions that remain unanswered. Here we study the function of two of the main folate transporters, FolR1 and Rfc1, which are robustly expressed in these tissues. Folate is the precursor of S-adenosylmethionine, which is the main donor for DNA, protein and RNA methylation. Our results show that knockdown of FolR1 and/or Rfc1 reduced the abundance of histone H3 lysine and DNA methylation, two epigenetic modifications that play an important role during neural and neural crest development. Additionally, by knocking down folate transporter or pharmacologically inhibiting folate transport and metabolism, we observed ectopic Sox2 expression at the expense of neural crest markers in the dorsal neural tube. This is correlated with neural crest associated defects, with particular impact on orofacial formation. By using bisulfite sequencing, we show that this phenotype is consequence of reduced DNA methylation on the Sox2 locus at the dorsal neural tube, which can be rescued by the addition of folinic acid. Taken together, our in vivo results reveal the importance of folate as a source of the methyl groups necessary for the establishment of the correct epigenetic marks during neural and neural crest fate-restriction.


Assuntos
Deficiência de Ácido Fólico/fisiopatologia , Crista Neural/metabolismo , Fatores de Transcrição SOXB1/fisiologia , Animais , Embrião de Galinha , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/genética , Repressão Epigenética/genética , Repressão Epigenética/fisiologia , Epigenômica , Receptor 1 de Folato , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Histonas/metabolismo , Humanos , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/fisiopatologia
4.
Mov Disord ; 34(5): 625-636, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30913345

RESUMO

The term "cerebral palsy mimic" is used to describe a number of neurogenetic disorders that may present with motor symptoms in early childhood, resulting in a misdiagnosis of cerebral palsy. Cerebral palsy describes a heterogeneous group of neurodevelopmental disorders characterized by onset in infancy or early childhood of motor symptoms (including hypotonia, spasticity, dystonia, and chorea), often accompanied by developmental delay. The primary etiology of a cerebral palsy syndrome should always be identified if possible. This is particularly important in the case of genetic or metabolic disorders that have specific disease-modifying treatment. In this article, we discuss clinical features that should alert the clinician to the possibility of a cerebral palsy mimic, provide a practical framework for selecting and interpreting neuroimaging, biochemical, and genetic investigations, and highlight selected conditions that may present with predominant spasticity, dystonia/chorea, and ataxia. Making a precise diagnosis of a genetic disorder has important implications for treatment, and for advising the family regarding prognosis and genetic counseling. © 2019 International Parkinson and Movement Disorder Society.


Assuntos
Paralisia Cerebral/diagnóstico , Diagnóstico Diferencial , Transtornos dos Movimentos/diagnóstico , Adenilil Ciclases/genética , Ataxia/fisiopatologia , Ataxia Telangiectasia/diagnóstico , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/fisiopatologia , Ataxia Telangiectasia/terapia , Encéfalo/diagnóstico por imagem , Encefalopatias Metabólicas Congênitas/diagnóstico , Encefalopatias Metabólicas Congênitas/genética , Encefalopatias Metabólicas Congênitas/fisiopatologia , Encefalopatias Metabólicas Congênitas/terapia , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/fisiopatologia , Erros Inatos do Metabolismo dos Carboidratos/terapia , Paralisia Cerebral/fisiopatologia , Coreia/fisiopatologia , Creatina/deficiência , Creatina/genética , Discinesias/diagnóstico , Discinesias/genética , Discinesias/fisiopatologia , Discinesias/terapia , Distonia/fisiopatologia , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/genética , Deficiência de Ácido Fólico/fisiopatologia , Deficiência de Ácido Fólico/terapia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/genética , Humanos , Hiperargininemia/diagnóstico , Hiperargininemia/genética , Hiperargininemia/fisiopatologia , Hiperargininemia/terapia , Síndrome de Lesch-Nyhan/diagnóstico , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/fisiopatologia , Síndrome de Lesch-Nyhan/terapia , Imageamento por Ressonância Magnética , Deficiência Intelectual Ligada ao Cromossomo X/diagnóstico , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/fisiopatologia , Deficiência Intelectual Ligada ao Cromossomo X/terapia , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/genética , Transtornos dos Movimentos/genética , Transtornos dos Movimentos/fisiopatologia , Transtornos dos Movimentos/terapia , Deficiência Múltipla de Carboxilase/diagnóstico , Deficiência Múltipla de Carboxilase/genética
5.
Mov Disord ; 34(5): 598-613, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30557456

RESUMO

There are several hundred single-gene disorders that we classify as inborn errors of metabolism. Inborn errors of metabolism are often rare and highly heterogeneous multisystem diseases with non-neurological and neurological manifestations, commonly with onset during childhood. Movement disorders are among the most common neurological problems in inborn errors of metabolism, but, in many cases, remain poorly defined. Although movement disorders are usually not the only and often not the presenting symptom, their recognition can facilitate a diagnosis. Movement disorders contribute substantially to the morbidity in inborn errors of metabolism and can have a significant impact on quality of life. Common metabolic movement disorders include the monoamine neurotransmitter disorders, disorders of amino and organic acid metabolism, metal storage disorders, lysosomal storage disorders, congenital disorders of autophagy, disorders of creatine metabolism, vitamin-responsive disorders, and disorders of energy metabolism. Importantly, disease-modifying therapies exist for a number of inborn errors of metabolism, and early recognition and treatment can prevent irreversible CNS damage and reduce morbidity and mortality. A phenomenology-based approach, based on the predominant movement disorder, can facilitate a differential diagnosis and can guide biochemical, molecular, and imaging testing. The complexity of metabolic movement disorders demands an interdisciplinary approach and close collaboration of pediatric neurologists, neurologists, geneticists, and experts in metabolism. In this review, we develop a general framework for a phenomenology-based approach to movement disorders in inborn errors of metabolism and discuss an approach to identifying the "top ten" of treatable inborn errors of metabolism that present with movement disorders-diagnoses that should never be missed. © 2018 International Parkinson and Movement Disorder Society.


Assuntos
Erros Inatos do Metabolismo/fisiopatologia , Transtornos dos Movimentos/fisiopatologia , Erros Inatos do Metabolismo dos Aminoácidos/complicações , Erros Inatos do Metabolismo dos Aminoácidos/diagnóstico , Erros Inatos do Metabolismo dos Aminoácidos/fisiopatologia , Erros Inatos do Metabolismo dos Aminoácidos/terapia , Ataxia/complicações , Ataxia/diagnóstico , Ataxia/etiologia , Ataxia/fisiopatologia , Ataxia/terapia , Doenças dos Gânglios da Base/complicações , Doenças dos Gânglios da Base/diagnóstico , Doenças dos Gânglios da Base/fisiopatologia , Doenças dos Gânglios da Base/terapia , Encefalopatias Metabólicas/complicações , Encefalopatias Metabólicas/diagnóstico , Encefalopatias Metabólicas/fisiopatologia , Encefalopatias Metabólicas/terapia , Erros Inatos do Metabolismo dos Carboidratos/complicações , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/fisiopatologia , Erros Inatos do Metabolismo dos Carboidratos/terapia , Coreia/etiologia , Coreia/fisiopatologia , Distonia/etiologia , Distonia/fisiopatologia , Distúrbios Distônicos/complicações , Distúrbios Distônicos/diagnóstico , Distúrbios Distônicos/etiologia , Distúrbios Distônicos/fisiopatologia , Distúrbios Distônicos/terapia , Deficiência de Ácido Fólico/complicações , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/fisiopatologia , Deficiência de Ácido Fólico/terapia , Glutaril-CoA Desidrogenase/deficiência , Degeneração Hepatolenticular/complicações , Degeneração Hepatolenticular/diagnóstico , Degeneração Hepatolenticular/fisiopatologia , Degeneração Hepatolenticular/terapia , Humanos , Doenças Metabólicas/complicações , Doenças Metabólicas/diagnóstico , Doenças Metabólicas/fisiopatologia , Doenças Metabólicas/terapia , Erros Inatos do Metabolismo/complicações , Erros Inatos do Metabolismo/diagnóstico , Erros Inatos do Metabolismo/terapia , Proteínas de Transporte de Monossacarídeos/deficiência , Transtornos dos Movimentos/etiologia , Espasticidade Muscular/etiologia
6.
Mol Hum Reprod ; 23(7): 461-477, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28535307

RESUMO

STUDY QUESTION: Do paternal exposures to folic acid deficient (FD), and/or folic acid supplemented (FS) diets, throughout germ cell development adversely affect male germ cells and consequently offspring health outcomes? SUMMARY ANSWER: Male mice exposed over their lifetimes to both FD and FS diets showed decreased sperm counts and altered imprinted gene methylation with evidence of transmission of adverse effects to the offspring, including increased postnatal-preweaning mortality and variability in imprinted gene methylation. WHAT IS KNOWN ALREADY: There is increasing evidence that disruptions in male germ cell epigenetic reprogramming are associated with offspring abnormalities and intergenerational disease. The fetal period is the critical time of DNA methylation pattern acquisition for developing male germ cells and an adequate supply of methyl donors is required. In addition, DNA methylation patterns continue to be remodeled during postnatal spermatogenesis. Previous studies have shown that lifetime (prenatal and postnatal) folic acid deficiency can alter the sperm epigenome and increase the incidence of fetal morphological abnormalities. STUDY DESIGN, SIZE, DURATION: Female BALB/c mice (F0) were placed on one of four amino-acid defined diets for 4 weeks before pregnancy and throughout pregnancy and lactation: folic acid control (Ctrl; 2 mg/kg), 7-fold folic acid deficient (7FD; 0.3 mg/kg), 10-fold high FS (10FS, 20 mg/kg) or 20-fold high FS (20FS, 40 mg/kg) diets. F1 males were weaned to their respective prenatal diets to allow for diet exposure during all windows of germline epigenetic reprogramming: the erasure, re-establishment and maintenance phases. PARTICIPANTS/MATERIALS, SETTINGS, METHODS: F0 females were mated with chow-fed males to produce F1 litters whose germ cells were exposed to the diets throughout embryonic development. F1 males were subsequently mated with chow-fed female mice. Two F2 litters, unexposed to the experimental diets, were generated from each F1 male; one litter was collected at embryonic day (E)18.5 and one delivered and followed postnatally. DNA methylation at a global level and at the differentially methylated regions of imprinted genes (H19, Imprinted Maternally Expressed Transcript (Non-Protein Coding)-H19, Small Nuclear Ribonucleoprotein Polypeptide N-Snrpn, KCNQ1 Opposite Strand/Antisense Transcript 1 (Non-Protein Coding)-Kcnq1ot1, Paternally Expressed Gene 1-Peg1 and Paternally Expressed Gene 3-Peg3) was assessed by luminometric methylation analysis and bisulfite pyrosequencing, respectively, in F1 sperm, F2 E18.5 placenta and F2 E18.5 brain cortex. MAIN RESULTS AND THE ROLE OF CHANCE: F1 males exhibited lower sperm counts following lifetime exposure to both folic acid deficiency and the highest dose of folic acid supplementation (20FS), (both P < 0.05). Post-implantation losses were increased amongst F2 E18.5 day litters from 20FS exposed F1 males (P < 0.05). F2 litters derived from both 7FD and 20FS exposed F1 males had significantly higher postnatal-preweaning pup death (both P < 0.05). Sperm from 10FS exposed males had increased variance in methylation across imprinted gene H19, P < 0.05; increased variance at a few sites within H19 was also found for the 7FD and 20FS groups (P < 0.05). While the 20FS diet resulted in inter-individual alterations in methylation across the imprinted genes Snrpn and Peg3 in F2 E18.5 placenta, ≥50% of individual sites tested in Peg1 and/or Peg3 were affected in the 7FD and 10FS groups. Inter-individual alterations in Peg1 methylation were found in F2 E18.5 day 10FS group brain cortex (P < 0.05). LARGE SCALE DATA: Not applicable. LIMITATIONS REASONS FOR CAUTION: The cause of the increase in postnatal-preweaning mortality was not investigated post-mortem. Further studies are required to understand the mechanisms underlying the adverse effects of folic acid deficiency and supplementation on developing male germ cells. Genome-wide DNA and histone methylome studies as well as gene expression studies are required to better understand the links between folic acid exposures, an altered germ cell epigenome and offspring outcomes. WIDER IMPLICATIONS OF THE FINDINGS: The findings of this study provide further support for paternally transmitted environmental effects. The results indicate that both folic acid deficiency and high dose supplementation can be detrimental to germ cell development and reproductive fitness, in part by altering DNA methylation in sperm. STUDY FUNDING AND COMPETING INTERESTS: This study was supported by a grant to J.M.T. from the Canadian Institutes of Health Research (CIHR #89944). The authors declare they have no conflicts of interest.


Assuntos
Metilação de DNA/efeitos dos fármacos , Suplementos Nutricionais , Epigênese Genética , Deficiência de Ácido Fólico/genética , Ácido Fólico/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal/genética , Reprodução/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Feminino , Deficiência de Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/mortalidade , Deficiência de Ácido Fólico/fisiopatologia , Impressão Genômica , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/mortalidade , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Reprodução/genética , Espermatogênese/efeitos dos fármacos , Espermatogênese/genética , Espermatozoides/efeitos dos fármacos , Espermatozoides/crescimento & desenvolvimento , Espermatozoides/metabolismo , Análise de Sobrevida , Desmame , Proteínas Centrais de snRNP/genética , Proteínas Centrais de snRNP/metabolismo
7.
FASEB J ; 29(2): 418-32, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25384423

RESUMO

Nutritional imbalance is emerging as a causative factor of hearing loss. Epidemiologic studies have linked hearing loss to elevated plasma total homocysteine (tHcy) and folate deficiency, and have shown that folate supplementation lowers tHcy levels potentially ameliorating age-related hearing loss. The purpose of this study was to address the impact of folate deficiency on hearing loss and to examine the underlying mechanisms. For this purpose, 2-mo-old C57BL/6J mice (Animalia Chordata Mus musculus) were randomly divided into 2 groups (n = 65 each) that were fed folate-deficient (FD) or standard diets for 8 wk. HPLC analysis demonstrated a 7-fold decline in serum folate and a 3-fold increase in tHcy levels. FD mice exhibited severe hearing loss measured by auditory brainstem recordings and TUNEL-positive-apoptotic cochlear cells. RT-quantitative PCR and Western blotting showed reduced levels of enzymes catalyzing homocysteine (Hcy) production and recycling, together with a 30% increase in protein homocysteinylation. Redox stress was demonstrated by decreased expression of catalase, glutathione peroxidase 4, and glutathione synthetase genes, increased levels of manganese superoxide dismutase, and NADPH oxidase-complex adaptor cytochrome b-245, α-polypeptide (p22phox) proteins, and elevated concentrations of glutathione species. Altogether, our findings demonstrate, for the first time, that the relationship between hyperhomocysteinemia induced by folate deficiency and premature hearing loss involves impairment of cochlear Hcy metabolism and associated oxidative stress.


Assuntos
Cóclea/fisiopatologia , Deficiência de Ácido Fólico/fisiopatologia , Perda Auditiva/fisiopatologia , Homocisteína/metabolismo , Hiper-Homocisteinemia/fisiopatologia , Estresse Oxidativo , Animais , Apoptose , Betaína-Homocisteína S-Metiltransferase/genética , Catalase/metabolismo , Cromatografia Líquida de Alta Pressão , Feminino , Ácido Fólico/sangue , Deficiência de Ácido Fólico/complicações , Glutationa Peroxidase/metabolismo , Glutationa Sintase/metabolismo , Células Ciliadas Auditivas/citologia , Perda Auditiva/etiologia , Homocisteína/deficiência , Hiper-Homocisteinemia/complicações , Marcação In Situ das Extremidades Cortadas , Metionina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxirredução , Fosfolipídeo Hidroperóxido Glutationa Peroxidase
8.
Nutr Cancer ; 68(7): 1065-72, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27472187

RESUMO

Acute lymphoblastic leukemia (ALL) is the most common type of leukemia in children that can be affected by maternal diet. The aim of this study was to evaluate maternal dietary risk factors of ALL. We searched MEDLINE, Cochrane Library, Springer Link, Wiley Online, Science Direct, Mosby, ISI Web of Science, OVID, ProQuest, and Scopus from database inception until February 2, 2016. Two reviewers scanned titles, abstracts, and keywords of articles after excluding duplicates. We included case-control studies evaluating the relationship between maternal diet during pregnancy and childhood ALL. The search resulted in 2,940 papers, of which 11 full-text articles met the criteria for inclusion in the review and were analyzed. The finding of these studies suggest that maternal diet composed largely of vegetables, fruits, and protein sources before and during pregnancy can reduce the risk of ALL in offspring. Maternal alcohol intake had no effect. Nevertheless, inherent limitations of case-control studies like measurement error, random error, recall bias, and selection bias preclude conclusive evidence. Persuading pregnant women to follow a healthy diet rich in fruits, vegetables, and protein may reduce the risk of childhood ALL. Avoiding alcohol intake seems prudent.


Assuntos
Dieta Saudável , Medicina Baseada em Evidências , Fenômenos Fisiológicos da Nutrição Materna , Cooperação do Paciente , Leucemia-Linfoma Linfoblástico de Células Precursoras/prevenção & controle , Estudos de Casos e Controles , Dieta/efeitos adversos , Feminino , Deficiência de Ácido Fólico/etiologia , Deficiência de Ácido Fólico/fisiopatologia , Humanos , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiologia , Gravidez , Fatores de Risco , Deficiência de Vitamina A/etiologia , Deficiência de Vitamina A/fisiopatologia
9.
Nutr Cancer ; 68(7): 1097-107, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27340879

RESUMO

We investigated whether alcohol and dietary folate intakes were associated with promoter methylation in clear-cell renal cell carcinoma (ccRCC). The Netherlands Cohort Study with a case-cohort design included 120,852 subjects aged 55-69 yr in 1986. Diet was measured with a food-frequency questionnaire. After 20.3 yr of follow-up, paraffin-embedded tumor blocks were collected. Methylation-specific polymerase chain reaction (MSP) was used to analyze promoter methylation of 11 genes. ccRCC cases were classified into low (0-19% of the genes), intermediate (20-39%), and high (40%+) methylation. Multivariable Cox regression analyses were conducted, stratified according to methylation, including 3980 subcohort members and 297 ccRCC cases. Increasing alcohol intake was associated with decreased ccRCC risk, but was not statistically significant; multivariable adjusted hazard ratio (HR) for ≥30 g alcohol/day versus 0 g/day was 0.78 [95% confidence interval (CI): 0.48-1.24], and P-value for trend was 0.46. In strata according to methylation index, no significant heterogeneity was observed. Dietary folate intake was not associated with ccRCC risk. There was no significant heterogeneity between strata according to methylation index. There was no effect modification of alcohol and dietary folate intake on ccRCC risk, nor in strata according to methylation index. Our findings do not support the hypothesis that alcohol and dietary folate intakes are involved in ccRCC.


Assuntos
Carcinoma de Células Renais/prevenção & controle , Ilhas de CpG , Dieta Saudável , Ácido Fólico/uso terapêutico , Neoplasias Renais/prevenção & controle , Cooperação do Paciente , Regiões Promotoras Genéticas , Consumo de Bebidas Alcoólicas/efeitos adversos , Carcinoma de Células Renais/epidemiologia , Carcinoma de Células Renais/etiologia , Carcinoma de Células Renais/metabolismo , Estudos de Coortes , Metilação de DNA , Feminino , Deficiência de Ácido Fólico/fisiopatologia , Seguimentos , Humanos , Neoplasias Renais/epidemiologia , Neoplasias Renais/etiologia , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Estudos Prospectivos , Sistema de Registros , Reprodutibilidade dos Testes , Risco , Autorrelato
10.
Nutr Neurosci ; 19(7): 310-7, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25087906

RESUMO

Autism spectrum disorders (ASD) consist in a range of neurodevelopmental conditions that share common features with autism, such as impairments in communication and social interaction, repetitive behaviors, stereotypies, and a limited repertoire of interests and activities. Some studies have reported that folic acid supplementation could be associated with a higher incidence of autism, and therefore, we aimed to conduct a systematic review of studies involving relationships between this molecule and ASD. The MEDLINE database was searched for studies written in English which evaluated the relationship between autism and folate. The initial search yielded 60 potentially relevant articles, of which 11 met the inclusion criteria. The agreement between reviewers was κ = 0.808. The articles included in the present study addressed topics related to the prescription of vitamins, the association between folic acid intake/supplementation during pregnancy and the incidence of autism, food intake, and/or nutrient supplementation in children/adolescents with autism, the evaluation of serum nutrient levels, and nutritional interventions targeting ASD. Regarding our main issue, namely the effect of folic acid supplementation, especially in pregnancy, the few and contradictory studies present inconsistent conclusions. Epidemiological associations are not reproduced in most of the other types of studies. Although some studies have reported lower folate levels in patients with ASD, the effects of folate-enhancing interventions on the clinical symptoms have yet to be confirmed.


Assuntos
Transtorno do Espectro Autista/etiologia , Suplementos Nutricionais/efeitos adversos , Medicina Baseada em Evidências , Desenvolvimento Fetal , Ácido Fólico/efeitos adversos , Fenômenos Fisiológicos da Nutrição Materna , Adolescente , Fenômenos Fisiológicos da Nutrição do Adolescente , Transtorno do Espectro Autista/sangue , Transtorno do Espectro Autista/epidemiologia , Transtorno do Espectro Autista/prevenção & controle , Criança , Fenômenos Fisiológicos da Nutrição Infantil , Dieta/efeitos adversos , Feminino , Ácido Fólico/sangue , Ácido Fólico/uso terapêutico , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/dietoterapia , Deficiência de Ácido Fólico/fisiopatologia , Deficiência de Ácido Fólico/prevenção & controle , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/etiologia , Hiper-Homocisteinemia/fisiopatologia , Hiper-Homocisteinemia/prevenção & controle , Incidência , Masculino , Gravidez , Reprodutibilidade dos Testes , Fatores de Risco
11.
Andrologia ; 48(5): 532-5, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26302884

RESUMO

This study measured the serum folic acid (FA) level in patients with erectile dysfunction (ED) and evaluated the possible association between the serum FA level and erectile function. The study divided 120 patients with ED into 3 groups of 40 patients each: those with severe, moderate and mild ED. Forty healthy men served as controls. Fasting serum samples were obtained, and the total testosterone, cholesterol and FA levels were measured using chemiluminescent immunoassays. There were no significant differences in the mean age, mean body mass index or mean serum total testosterone and cholesterol levels among the three ED groups and controls (P > 0.05). The mean serum FA concentrations were 7.2 ± 3.7, 7.1 ± 3.2, 10.2 ± 4.6 and 10.7 ± 4.6 ng ml(-1) in the severe, moderate and mild ED and control groups respectively. The mean serum FA concentration was significantly higher in the control group than in the severe and moderate ED groups (both P < 0.001), but not the mild ED group (P = 0.95). Considering the significant differences in the serum FA levels between the control and ED groups, serum FA deficiency might reflect the severity of ED.


Assuntos
Disfunção Erétil/sangue , Ácido Fólico/sangue , Adulto , Idoso , Estudos de Casos e Controles , Colesterol/sangue , Disfunção Erétil/etiologia , Disfunção Erétil/fisiopatologia , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/complicações , Deficiência de Ácido Fólico/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Testosterona/sangue
12.
Matern Child Nutr ; 12(1): 39-50, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24934272

RESUMO

Hispanic women have higher rates of neural tube defects and report lower total folic acid intakes than non-Hispanic white (NHW) women. Total folic acid intake, which is associated with neural tube defect risk reduction, has been found to vary by acculturation factors (i.e. language preference, country of origin, or time spent in the United States) among Hispanic women. It is unknown whether this same association is present for blood folate status. The objective of this research was to assess the differences in serum and red blood cell (RBC) folate concentrations between NHW women and Mexican American (MA) women and among MA women by acculturation factors. Cross-sectional data from the 2001-2010 National Health and Nutrition Examination Survey (NHANES) were used to investigate how blood folate concentrations differ among NHW or MA women of childbearing age. The impact of folic acid supplement use on blood folate concentrations was also examined. MA women with lower acculturation factors had lower serum and RBC folate concentrations compared with NHW women and to their more acculturated MA counterparts. Consuming a folic acid supplement can minimize these disparities, but MA women, especially lower acculturated MA women, were less likely to report using supplements. Public health efforts to increase blood folate concentrations among MA women should consider acculturation factors when identifying appropriate interventions.


Assuntos
Aculturação , Deficiência de Ácido Fólico/prevenção & controle , Ácido Fólico/sangue , Defeitos do Tubo Neural/prevenção & controle , Adolescente , Adulto , Estudos Transversais , Assistência à Saúde Culturalmente Competente , Suplementos Nutricionais , Eritrócitos/química , Feminino , Ácido Fólico/uso terapêutico , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/etnologia , Deficiência de Ácido Fólico/fisiopatologia , Alimentos Fortificados , Humanos , Americanos Mexicanos , Defeitos do Tubo Neural/epidemiologia , Defeitos do Tubo Neural/etnologia , Defeitos do Tubo Neural/etiologia , Inquéritos Nutricionais , Cuidado Pré-Concepcional , Prevalência , Risco , Estados Unidos/epidemiologia , População Branca , Adulto Jovem
13.
Ann Nutr Metab ; 66(4): 202-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26067849

RESUMO

BACKGROUND/AIMS: Telomere biology plays a fundamental role in genomic integrity and cell physiology. The newborn setting of telomere length (TL) likely has important implications for telomere dynamics over the lifespan; however, its determinants are poorly understood. Folate is essential for DNA integrity. The maternal compartment is the only source of folate for the developing fetus. We, therefore, tested the hypothesis that variation in maternal folate during pregnancy is associated with newborn TL. METHODS: A prospective, longitudinal study was conducted in 119 mother-newborn dyads. Eligible mothers were enrolled at 9.5 (SD ±2.1) weeks gestation and followed through birth. Concentrations of maternal serum folate were measured in the first trimester of pregnancy. Newborn telomere length was measured in cord blood mononuclear cells (CBMC). RESULTS: After accounting for the effects of other established determinants of newborn TL, each 10 ng/ml increase in maternal total folate was associated with a 5.8% increase in median TL (p = 0.03). The median TL in newborns of mother in the lowest quartile of total folate levels was approximately 10% shorter than that of newborns of mothers in the highest folate quartile. CONCLUSIONS: Our findings suggest that fetal TL exhibits developmental plasticity, and provide evidence that maternal nutrition may exert a 'programming' effect on this system.


Assuntos
Doenças Assintomáticas , Desenvolvimento Fetal , Deficiência de Ácido Fólico/sangue , Ácido Fólico/sangue , Fenômenos Fisiológicos da Nutrição Materna , Complicações na Gravidez/sangue , Encurtamento do Telômero , Adulto , Estudos de Coortes , Feminino , Sangue Fetal , Deficiência de Ácido Fólico/fisiopatologia , Humanos , Recém-Nascido , Leucócitos Mononucleares , Estudos Longitudinais , Masculino , Philadelphia , Gravidez , Complicações na Gravidez/fisiopatologia , Primeiro Trimestre da Gravidez , Gravidez de Alto Risco/sangue , Estudos Prospectivos , Adulto Jovem
14.
Lik Sprava ; (1-2): 156-7, 2015.
Artigo em Russo | MEDLINE | ID: mdl-26118060

RESUMO

This study focuses on the problem of finding an effective diagnostic algorithm to identify unipolar depression by examining the C677T MTHFR gene polymorphism and features of folate metabolism in patients with unipolar depression. The study proves that C677T MTHFR gene polymorphism is significantly associated with unipolar depression, which can be used to improve the quality of depression identification, as well as forming new ways of diagnosis.


Assuntos
Transtorno Depressivo/genética , Deficiência de Ácido Fólico/genética , Ácido Fólico/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo de Nucleotídeo Único , Adulto , Transtorno Depressivo/sangue , Transtorno Depressivo/diagnóstico , Transtorno Depressivo/fisiopatologia , Feminino , Deficiência de Ácido Fólico/sangue , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/fisiopatologia , Expressão Gênica , Homozigoto , Humanos , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Taxa de Mutação , Testes Psicológicos
15.
J Nutr ; 144(4): 419-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24500934

RESUMO

Folate metabolism affects DNA synthesis, methylation, mutation rates, genomic stability, and gene expression, which are altered in colon cancer. Serine hydroxymethyltransferase 1 (SHMT1) regulates thymidylate (dTMP) biosynthesis and uracil accumulation in DNA, and as such affects genome stability. Previously, we showed that decreased SHMT1 expression in Shmt1 knockout mice (Shmt1(-/+)) or its impaired nuclear localization, as occurs in mice over-expressing an Shmt1 transgene (Shmt1(tg+)), results in elevated uracil incorporation into DNA, which could affect colon cancer risk. We used these 2 models to determine the effect of altered SHMT1 expression and localization, and its interaction with folate insufficiency, on azoxymethane (AOM)-induced colon cancer in mice. Shmt1(-/+) and Shmt1(tg+) mice were weaned to a control or folate-and-choline-deficient (FCD) diet and fed the diet for 28 or 32 wk, respectively. At 6 wk of age, mice were injected weekly for 6 wk with 10 mg/kg AOM (w/v in saline). Colon uracil concentrations in nuclear DNA were elevated 2-7 fold in Shmt1(-/+) and Shmt1(tg+) mice. However, colon tumor incidence and numbers were not dependent on SHMT1 expression in Shmt1(-/+) or Shmt1(-/-) mice. The FCD diet reduced tumor load independent of Shmt1 genotype. In contrast, Shmt1(tg+) mice exhibited a 30% reduction in tumor incidence, a 50% reduction in tumor number, and a 60% reduction in tumor load compared with wild-type mice independent of dietary folate intake. Our data indicate that uracil accumulation in DNA does not predict tumor number in AOM-mediated carcinogenesis. Furthermore, enrichment of SHMT1 in the cytoplasm, as observed in Shmt1(tg+) mice, protects against AOM-mediated carcinogenesis independent of its role in nuclear de novo dTMP biosynthesis.


Assuntos
Carcinogênese/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , DNA/metabolismo , Modelos Animais de Doenças , Ácido Fólico/metabolismo , Timidina Monofosfato/metabolismo , Animais , Azoximetano , Deficiência de Colina/fisiopatologia , Colo/enzimologia , Colo/patologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Cruzamentos Genéticos , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Ácido Fólico/efeitos adversos , Deficiência de Ácido Fólico/fisiopatologia , Glicina Hidroximetiltransferase/biossíntese , Glicina Hidroximetiltransferase/genética , Glicina Hidroximetiltransferase/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína-Lisina 6-Oxidase/genética , Proteína-Lisina 6-Oxidase/metabolismo , Distribuição Aleatória , Carga Tumoral , Uracila/metabolismo
16.
Br J Nutr ; 112(8): 1323-32, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25313575

RESUMO

Folate is essential for fetal development, and its deficiency during gestation causes behavioural deficits in the offspring. The present study investigated its influence during weaning on brain function in the pups of rats that were put on a folate-deficient (FD) diet on postnatal day (PND) 1. Systemic folate deficiency in pups on the FD diet (n 15) was evident from the dramatically lower hepatic folate concentrations (median 23·7, range 8·1-48·4 ng/mg protein) and higher homocysteine concentrations (median 27·7, range 14·7-45·5 pmol/mg protein), respectively, compared with those of pups on the normal diet (ND; n 9) (median 114·5, range 64·5-158·5 ng/mg protein and median 15·5, range 11·6-18·9 pmol/mg protein) on PND 23. Brain folate concentrations although low were similar in pups on the FD diet (median 10·5, range 5·5-24·5 ng/mg protein) and ND (median 11·1, range 7·1-24·2 ng/mg protein). There was a high accumulation of homocysteine in the brain of FD pups, mostly in the hippocampus (median 58·1, range 40·8-99·7 pmol/mg protein) and cerebellum (median 69·1, range 50·8-126·6 pmol/mg protein), indicating metabolic folate deficiency despite normal brain folate concentrations. Developmental deficits or autistic traits were more frequent in the FD group than in the ND group and repetitive self-grooming occurred, on average, three times (range 1-8) v. once (range 0-3) during 5 min, respectively. Long-term memory or spatial learning and set-shifting deficits affected 12 to 62% of rats in the FD group compared with none in the ND group. Post-weaning folic acid supplementation did not correct these deficits. These observations indicate that folate deficiency during weaning affects postnatal development even when gestational folate supply is normal.


Assuntos
Encéfalo/metabolismo , Dieta/efeitos adversos , Deficiência de Ácido Fólico/fisiopatologia , Ácido Fólico/metabolismo , Deficiências da Aprendizagem/etiologia , Transtornos da Memória/etiologia , Neurônios/metabolismo , Animais , Comportamento Animal , Encéfalo/patologia , Cerebelo/metabolismo , Cerebelo/patologia , Suscetibilidade a Doenças , Feminino , Ácido Fólico/uso terapêutico , Deficiência de Ácido Fólico/dietoterapia , Deficiência de Ácido Fólico/etiologia , Deficiência de Ácido Fólico/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Homocisteína/metabolismo , Lactação , Deficiências da Aprendizagem/prevenção & controle , Fígado/metabolismo , Fígado/patologia , Masculino , Fenômenos Fisiológicos da Nutrição Materna , Transtornos da Memória/prevenção & controle , Memória de Longo Prazo , Ratos Long-Evans , Aprendizagem Espacial , Desmame
17.
J Nutr ; 143(1): 27-33, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23190761

RESUMO

Low folate status is a risk factor for colon carcinogenesis; mechanisms proposed to account for this relationship include uracil misincorporation into DNA and global DNA hypomethylation. We investigated whether such biomarkers are related to folate status in isolated colonocytes from colonoscopy patients. In cases with adenomatous polyps (n = 40) or hyperplastic polyps (n = 16), colonocytes were isolated from biopsies from the polyp, from a site adjacent to the polyp, and from normal mucosa 10-15 cm distal to the polyp. In polyp-free controls (n = 53), biopsies were taken from ascending, transverse, and descending areas of colon. Within adenoma cases, there was a trend (P-trend < 0.001) of decreasing colonocyte folate (pg/105 cells, mean ± CI) from the site distal to the polyp (16.9 ± 2.4), to the site adjacent to the polyp (14.7 ± 2.3), to the polyp (12.8 ± 2.0). Correspondingly, there were increases in uracil misincorporation (P-trend < 0.001) and global DNA hypomethylation (P-trend = 0.012) across the 3 sites. Colonocyte folate concentrations were significantly correlated with RBC folate concentrations, but only in individuals with generally lower (≤484 µg/L) RBC folate status (r = 0.54; P = 0.006; n = 24), and were also significantly lower in normal mucosa of cases with adenomatous polyps than in controls matched for colonic segment. In conclusion, localized folate deficiency in specific areas of colon might create carcinogenic fields and affect the development of colorectal polyps through uracil misincorporation and DNA hypomethylation; alternatively, the polyp itself might deplete folate in the surrounding tissue. Folate supplementation trials aimed at colon cancer prevention should target individuals with suboptimal folate status.


Assuntos
Pareamento Incorreto de Bases , Colo/metabolismo , Pólipos do Colo/metabolismo , Metilação de DNA , Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/metabolismo , Mucosa Intestinal/metabolismo , Pólipos Adenomatosos/etiologia , Pólipos Adenomatosos/metabolismo , Pólipos Adenomatosos/patologia , Adulto , Idoso , Biomarcadores/metabolismo , Estudos de Casos e Controles , Colo/patologia , Pólipos do Colo/etiologia , Pólipos do Colo/patologia , DNA/biossíntese , Dano ao DNA , Feminino , Deficiência de Ácido Fólico/patologia , Deficiência de Ácido Fólico/fisiopatologia , Humanos , Hiperplasia , Mucosa Intestinal/patologia , Masculino , Pessoa de Meia-Idade , Especificidade de Órgãos , Reto/metabolismo , Reto/patologia , Uracila/metabolismo
18.
Eur J Nutr ; 52(3): 1157-67, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22832899

RESUMO

PURPOSE: Folic acid (FA) is an emerging nutritional factor in the pathogenesis of diverse neurodegenerative disorders by still unknown mechanisms. The hippocampus is altered during the loss of cognitive abilities in humans and selectively affected when homocysteine increases. The aim was to evaluate the potential protective role of folic acid in the maintenance of biochemical markers related to the methionine cycle, as well as the integrity of the hippocampus as part of the brain in aged rats. METHODS: Male Sprague-Dawley rats (18 months old) were assigned to four different folic acid groups (0 mg FA/kg diet, deficient; 2 mg FA/kg diet, control; 8 mg FA/kg diet, moderate supplementation; 40 mg FA/kg diet, extra supplementation) for 30 days. We evaluated several parameters related to the methionine cycle. In addition, hippocampus areas were immunostained for specific neuronal markers and astrocytes. RESULTS: Serum folate levels increased according to FA dietary level (p < 0.01). There was a significant increase in the serum homocysteine concentrations in the folic acid-deficient diet group (p < 0.01). However, brain S-adenosylmethionine and S-adenosylhomocysteine did not differ significantly between the folic acid groups. Consequently, the methylation ratio was also unchanged. The morphometric analysis did not show any differences in the number of neurons and astrocytes between groups, except when comparing the folic acid-deficient diet versus folic acid-supplemented diet in the striatum of the hippocampus. CONCLUSIONS: Clearly, the dietary FA deficiency negatively affects the methionine metabolism biomarkers, while excessive supplementation seems to be unnecessary for optimal maintenance of the methylation cycle and hippocampus integrity.


Assuntos
Envelhecimento , Disfunção Cognitiva/prevenção & controle , Dieta , Ácido Fólico/uso terapêutico , Hipocampo/metabolismo , Metionina/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Biomarcadores/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/patologia , Dieta/efeitos adversos , Suplementos Nutricionais , Ácido Fólico/administração & dosagem , Ácido Fólico/sangue , Ácido Fólico/metabolismo , Deficiência de Ácido Fólico/dietoterapia , Deficiência de Ácido Fólico/etiologia , Deficiência de Ácido Fólico/fisiopatologia , Hipocampo/patologia , Hiper-Homocisteinemia/etiologia , Hiper-Homocisteinemia/fisiopatologia , Hiper-Homocisteinemia/prevenção & controle , Masculino , Metilação , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
19.
Am J Physiol Endocrinol Metab ; 302(1): E61-7, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21934042

RESUMO

Formate can differentiate between hyperhomocysteinemia due to impaired remethylation and impaired transsulfuration. Am J Physiol Endocrinol Metab 301: E000-E000, 2011. First published September 20, 2011; 10.1152/ajpendo.00345.2011.-We carried out a (1)H-NMR metabolomic analysis of sera from vitamin B(12)-deficient rats. In addition to the expected increases in methylmalonate and homocysteine (Hcy), we observed an approximately sevenfold increase in formate levels, from 64 µM in control rats to 402 µM in vitamin B(12)-deficient rats. Urinary formate was also elevated. This elevation of formate could be attributed to impaired one-carbon metabolism since formate is assimilated into the one-carbon pool by incorporation into 10-formyl-THF via the enzyme 10-formyl-THF synthase. Both plasma and urinary formate were also increased in folate-deficient rats. Hcy was elevated in both the vitamin B(12)- and folate-deficient rats. Although plasma Hcy was also elevated, plasma formate was unaffected in vitamin B(6)-deficient rats (impaired transsulfuration pathway). These results were in accord with a mathematical model of folate metabolism, which predicted that reduction in methionine synthase activity would cause increased formate levels, whereas reduced cystathionine ß-synthase activity would not. Our data indicate that formate provides a novel window into cellular folate metabolism, that elevated formate can be a useful indicator of deranged one-carbon metabolism and can be used to discriminate between the hyperhomocysteinemia caused by defects in the remethylation and transsulfuration pathways.


Assuntos
Formiatos/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/etiologia , Animais , Biomarcadores/sangue , Biomarcadores/urina , Encéfalo/metabolismo , Deficiência de Ácido Fólico/diagnóstico , Deficiência de Ácido Fólico/fisiopatologia , Formiatos/urina , Homocisteína/sangue , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/urina , Fígado/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Metabolômica/métodos , Metionina/metabolismo , Metilação , Ácido Metilmalônico/sangue , Modelos Biológicos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Deficiência de Vitamina B 12/diagnóstico , Deficiência de Vitamina B 12/fisiopatologia , Deficiência de Vitamina B 6/sangue , Deficiência de Vitamina B 6/diagnóstico , Deficiência de Vitamina B 6/metabolismo , Deficiência de Vitamina B 6/urina
20.
Br J Nutr ; 107(5): 744-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21791146

RESUMO

Long-term supplementation with folic acid may improve cognitive performance in older individuals. The relationship between folate status and cognitive performance might be mediated by changes in methylation capacity, as methylation reactions are important for normal functioning of the brain. Although aberrant DNA methylation has been implicated in neurodevelopmental disorders, the relationship between DNA methylation status and non-pathological cognitive functioning in human subjects has not yet been investigated. The present study investigated the associations between global DNA methylation and key domains of cognitive functioning in healthy older adults. Global DNA methylation, defined as the percentage of methylated cytosine to total cytosine, was measured in leucocytes by liquid chromatography-MS/MS, in 215 men and women, aged 50-70 years, who participated in the Folic Acid and Carotid Intima-Media Thickness (FACIT) study (clinical trial registration number NCT00110604). Cognitive performance was assessed by means of the Visual Verbal Word Learning Task, the Stroop Colour-Word Interference Test, the Concept Shifting Test, the Letter-Digit Substitution Test and the Verbal Fluency Test. Using hierarchical linear regression analyses adjusted for age, sex, level of education, alcohol consumption, smoking status, physical activity, erythrocyte folate concentration and 5,10-methylenetetrahydrofolate reductase 677 C â†’ T genotype, we found that global DNA methylation was not related to cognitive performance on any of the domains measured. The present study results do not support the hypothesis that global DNA methylation, as measured in leucocytes, might be associated with cognitive functioning in healthy older individuals.


Assuntos
Envelhecimento/psicologia , Disfunção Cognitiva/metabolismo , Metilação de DNA , Leucócitos/metabolismo , 5,10-Metilenotetra-Hidrofolato Redutase (FADH2)/genética , Idoso , Disfunção Cognitiva/sangue , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/genética , Estudos Transversais , Citosina/metabolismo , Método Duplo-Cego , Eritrócitos/metabolismo , Feminino , Ácido Fólico/sangue , Deficiência de Ácido Fólico/fisiopatologia , Estudos de Associação Genética , Homocisteína/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Escalas de Graduação Psiquiátrica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA