Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Clin Nutr Metab Care ; 16(3): 339-45, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23493015

RESUMO

PURPOSE OF REVIEW: Humans eating diets low in choline develop fatty liver and liver damage. Rodents fed choline-methionine-deficient diets not only develop fatty liver, but also progress to develop fibrosis and hepatocarcinoma. This review focuses on the role of choline in liver function, with special emphasis on the epigenetic mechanisms of action. RECENT FINDINGS: Dietary intake of methyl donors like choline influences the methylation of DNA and histones, thereby altering the epigenetic regulation of gene expression. The liver is the major organ within which methylation reactions occur, and many of the hepatic genes involved in pathways for the development of fatty liver, hepatic fibrosis, and hepatocarcinomas are epigenetically regulated. SUMMARY: Dietary intake of choline varies over a three-fold range and many humans have genetic polymorphisms that increase their demand for choline. Choline is an important methyl donor needed for the generation of S-adenosylmethionine. Dietary choline intake is an important modifier of epigenetic marks on DNA and histones, and thereby modulates the gene expression in many of the pathways involved in liver function and dysfunction.


Assuntos
Colina/farmacologia , Epigênese Genética , Fígado/metabolismo , Animais , Metilação de DNA , Dieta , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Ácido Fólico/farmacologia , Expressão Gênica , Histonas/genética , Histonas/metabolismo , Humanos , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/metabolismo , Modelos Animais , Polimorfismo Genético , S-Adenosilmetionina/farmacologia
2.
J Biol Chem ; 286(42): 36258-67, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21878621

RESUMO

Betaine-homocysteine S-methyltransferase (BHMT) uses betaine to catalyze the conversion of homocysteine (Hcy) to methionine. There are common genetic polymorphisms in the BHMT gene in humans that can alter its enzymatic activity. We generated the first Bhmt(-/-) mouse to model the functional effects of mutations that result in reduced BHMT activity. Deletion of Bhmt resulted in a 6-fold increase (p < 0.01) in hepatic and an 8-fold increase (p < 0.01) in plasma total Hcy concentrations. Deletion of Bhmt resulted in a 43% reduction in hepatic S-adenosylmethionine (AdoMet) (p < 0.01) and a 3-fold increase in hepatic S-adenosylhomocysteine (AdoHcy) (p < 0.01) concentrations, resulting in a 75% reduction in methylation potential (AdoMet:AdoHcy) (p < 0.01). Bhmt(-/-) mice accumulated betaine in most tissues, including a 21-fold increase in the liver concentration compared with wild type (WT) (p < 0.01). These mice had lower concentrations of choline, phosphocholine, glycerophosphocholine, phosphatidylcholine, and sphingomyelin in several tissues. At 5 weeks of age, Bhmt(-/-) mice had 36% lower total hepatic phospholipid concentrations and a 6-fold increase in hepatic triacyglycerol concentrations compared with WT (p < 0.01), which was due to a decrease in the secretion of very low density lipoproteins. At 1 year of age, 64% of Bhmt(-/-) mice had visible hepatic tumors. Histopathological analysis revealed that Bhmt(-/-) mice developed hepatocellular carcinoma or carcinoma precursors. These results indicate that BHMT has an important role in Hcy, choline, and one-carbon homeostasis. A lack of Bhmt also affects susceptibility to fatty liver and hepatocellular carcinoma. We suggest that functional polymorphisms in BHMT that significantly reduce activity may have similar effects in humans.


Assuntos
Betaína-Homocisteína S-Metiltransferase/metabolismo , Carcinoma Hepatocelular/enzimologia , Colina/metabolismo , Fígado Gorduroso/enzimologia , Proteínas de Neoplasias/metabolismo , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Carbono/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Colina/genética , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Deleção de Genes , Humanos , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Fosfolipídeos/genética , Fosfolipídeos/metabolismo , S-Adenosilmetionina/genética , S-Adenosilmetionina/metabolismo , Triglicerídeos/genética , Triglicerídeos/metabolismo
3.
Proc Natl Acad Sci U S A ; 107(29): 12834-9, 2010 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-20624989

RESUMO

We examined whether maternal dietary choline modulates angiogenesis in fetal brain. Pregnant C57BL/6 mice were fed either a choline-deficient (CD), control (CT), or choline-supplemented diet (CS) from days 12 to 17 (E12-17) of pregnancy and then fetal brains were studied. In CD fetal hippocampus, proliferation of endothelial cells (EC) was decreased by 32% (p < 0.01 vs. CT or CS) while differentiated EC clusters (expressing factor VIII related antigen (RA)) increased by 25% (p < 0.01 vs. CT or CS). These changes were associated with > 25% decrease in the number of blood vessels in CD fetal hippocampus (p < 0.01 vs. CT and CS), with no change in total cross-sectional area of these blood vessels. Expression of genes for the angiogenic signals derived from both endothelial and neuronal progenitor cells (NPC) was increased in CD fetal hippocampus VEGF C (Vegfc), 2.0-fold, p < 0.01 vs. CT and angiopoietin 2 (Angpt2), 2.1-fold, (p < 0.01 vs. CT)). Similar increased expression was observed in NPC isolated from E14 fetal mouse brains and exposed to low (5 microM), CT (70 microM), or high choline (280 microM) media for 72 h (low choline caused a 9.7-fold increase in relative gene expression of Vegfc (p < 0.001 vs. CT and high) and a 3.4-fold increase in expression of Angpt2, (p < 0.05 vs. CT and high). ANGPT2 protein was increased 42.2% (p < 0.01). Cytosine-phosphate-guanine dinucleotide islands in the proximity of the promoter areas of Vegfc and Angpt2 were hypomethylated in low choline NPC compared to CT NPC (p < 0.01). We conclude that maternal dietary choline intake alters angiogenesis in the developing fetal hippocampus.


Assuntos
Deficiência de Colina/embriologia , Dieta , Feto/irrigação sanguínea , Feto/metabolismo , Hipocampo/irrigação sanguínea , Relações Materno-Fetais , Neovascularização Fisiológica , Indutores da Angiogênese/metabolismo , Animais , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/patologia , Contagem de Células , Proliferação de Células , Células Cultivadas , Deficiência de Colina/metabolismo , Metilação de DNA , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/genética , Neurônios/citologia , Fosforilcolina/metabolismo , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Sítio de Iniciação de Transcrição
4.
FASEB J ; 24(1): 184-95, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19752176

RESUMO

Maternal choline availability is essential for fetal neurogenesis. Choline deprivation (CD) causes hypomethylation of specific CpG islands in genes controlling cell cycling in fetal hippocampus. We now report that, in C57BL/6 mice, CD during gestational days 12-17 also altered methylation of the histone H3 in E17 fetal hippocampi. In the ventricular and subventricular zones, monomethyl-lysine 9 of H3 (H3K9me1) was decreased by 25% (P<0.01), and in the pyramidal layer, dimethyl-lysine 9 of H3 (H3K9me2) was decreased by 37% (P<0.05). These changes were region specific and were not observed in whole-brain preparations. Also, the same effects of CD on H3 methylation were observed in E14 neural progenitor cells (NPCs) in culture. Changes in G9a histone methyltransferase might mediate altered H3K9me2,1. Gene expression of G9a was decreased by 80% in CD NPCs (P<0.001). In CD, H3 was hypomethylated upstream of the RE1 binding site in the calbindin 1 promoter, and 1 CpG site within the calbindin1 promoter was hypermethylated. REST binding to RE1 (recruits G9a) was decreased by 45% (P<0.01) in CD. These changes resulted in increased expression of calbindin 1 in CD (260%; P<0.05). Thus, CD modulates histone methylation in NPCs, and this could underlie the observed changes in neurogenesis.


Assuntos
Deficiência de Colina/genética , Deficiência de Colina/metabolismo , Histonas/química , Histonas/metabolismo , Proteína G de Ligação ao Cálcio S100/genética , Animais , Apoptose , Sequência de Bases , Sítios de Ligação/genética , Calbindina 1 , Calbindinas , Células Cultivadas , Ilhas de CpG , DNA/genética , DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Epigênese Genética , Feminino , Hipocampo/embriologia , Hipocampo/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Modelos Biológicos , Gravidez , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Proteína G de Ligação ao Cálcio S100/metabolismo
5.
J Biol Chem ; 285(2): 1008-15, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-19889625

RESUMO

The development of fetal brain is influenced by nutrients such as docosahexaenoic acid (DHA, 22:6) and choline. Phosphatidylethanolamine-N-methyltransferase (PEMT) catalyzes the biosynthesis of phosphatidylcholine from phosphatidylethanolamine enriched in DHA and many humans have functional genetic polymorphisms in the PEMT gene. Previously, it was reported that Pemt(-/-) mice have altered hippocampal development. The present study explores whether abnormal phosphatidylcholine biosynthesis causes altered incorporation of DHA into membranes, thereby influencing brain development, and determines whether supplemental dietary DHA can reverse some of these changes. Pregnant C57BL/6 wild type (WT) and Pemt(-/-) mice were fed a control diet, or a diet supplemented with 3 g/kg of DHA, from gestational day 11 to 17. Brains from embryonic day 17 fetuses derived from Pemt(-/-) dams fed the control diet had 25-50% less phospholipid-DHA as compared with WT (p < 0.05). Also, they had 60% more neural progenitor cell proliferation (p < 0.05), 60% more neuronal apoptosis (p < 0.01), and 30% less calretinin expression (p < 0.05; a marker of neuronal differentiation) in the hippocampus compared with WT. The DHA-supplemented diet increased fetal brain Pemt(-/-) phospholipid-DHA to WT levels, and abrogated the neural progenitor cell proliferation and apoptosis differences. Although this diet did not change proliferation in the WT group, it halved the rate of apoptosis (p < 0.05). In both genotypes, the DHA-supplemented diet increased calretinin expression 2-fold (p < 0.05). These results suggest that the changes in hippocampal development in the Pemt(-/-) mouse could be mediated by altered DHA incorporation into membrane phospholipids, and that maternal dietary DHA can influence fetal brain development.


Assuntos
Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/farmacologia , Feto/embriologia , Hipocampo/embriologia , Fosfatidil-N-Metiletanolamina N-Metiltransferase , Animais , Química Encefálica/efeitos dos fármacos , Química Encefálica/genética , Proliferação de Células/efeitos dos fármacos , Feminino , Feto/citologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Fosfolipídeos/metabolismo , Gravidez , Células-Tronco/citologia , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...