Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Biochem Biophys Res Commun ; 651: 62-69, 2023 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-36791500

RESUMO

Obesity is a major risk factor for type 2 diabetes, coronary heart disease, and strok. These diseases are associated with profound alterations in gene expression in metabolic tissues. Epigenetic-mediated regulation of gene expression is one mechanism through which environmental factors, such as diet, modify gene expression and disease predisposition. However, epigenetic control of gene expression in obesity and insulin resistance is not fully characterized. We discovered that liver-specific stearoyl-CoA desaturase-1 (Scd1) knockout mice (LKO) fed a high-carbohydrate low-fat diet exhibit dramatic changes in hepatic gene expression and metabolites of the folate cycle and one-carbon metabolism respectively for the synthesis of S-adenosylmethionine (SAM). LKO mice show an increased ratio of S-adenosylmethionine to S-adenosylhomocysteine, a marker for increased cellular methylation capacity. Furthermore, expression of DNA and histone methyltransferase genes is up-regulated while the mRNA and protein levels of the non-DNA methyltransferases including phosphatidylethanolamine methyltransferase (PEMT), Betaine homocysteine methyltransferase (Bhmt), and the SAM-utilizing enzymes such as glycine-N-methyltransferase (Gnmt) and guanidinoacetate methyltransferase (Gamt) are generally down-regulated. Feeding LKO mice a high carbohydrate diet supplemented with triolein, but not tristearin, and increased endogenous hepatic synthesis of oleate but not palmitoleate in Scd1 global knockout mice normalized one carbon gene expression and metabolite levels. Additionally, changes in one carbon gene expression are independent of the PGC-1α-mediated ER stress response previously reported in the LKO mice. Together, these results highlight the important role of oleate in maintaining one-carbon cycle homeostasis and point to observed changes in one-carbon metabolism as a novel mediator of the Scd1 deficiency-induced liver phenotype.


Assuntos
Diabetes Mellitus Tipo 2 , Ácido Oleico , Camundongos , Animais , Ácido Oleico/metabolismo , S-Adenosilmetionina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Fígado/metabolismo , Carboidratos , Camundongos Knockout , Obesidade/metabolismo , Carbono/metabolismo , Fosfatidiletanolamina N-Metiltransferase/metabolismo
2.
J Cell Biochem ; 123(6): 1032-1052, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35416329

RESUMO

In Saccharomyces cerevisiae, the transcription factor GCR1 plays a vital role in carbohydrate metabolism and in the current study we tried to elucidate its role in lipid metabolism. In silico analysis revealed the upstream activation sequence (UAS) in the promoter region of OPI3 possessed six conserved recognition sequences for Gcr1p and the ChIP assay confirmed the binding of Gcr1p on the OPI3 promoter region. The real-time quantitative polymerase chain reaction and promoter-reporter activity revealed a substantial reduction in OPI3 expression and was supported with decreased phosphatidylcholine (PC) level that is rescued with exogenous choline supplementation in gcr1∆ cells. Simultaneously, there was an increase in triacylglycerol level, accompanied with increased number and size of lipid droplets in gcr1∆ cells. The expression of pT1, pT2 truncations in opi3∆ cells revealed the -1 to -500 bp in the promoter region is essential for the activation of OPI3 transcription. The mutation specifically at UASCT box (-265) in the OPI3 promoter region displayed a reduction in the PC level and the additional mutation at UASINO (-165) further reduced the PC level. Collectively, our data suggest that the GCR1 transcription factor also regulates the OPI3 expression and has an impact on lipid homeostasis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fosfatidiletanolamina N-Metiltransferase/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae , Fatores de Transcrição/metabolismo , Proteínas de Ligação a DNA/genética , Regulação Fúngica da Expressão Gênica , Ligação Proteica , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/genética
3.
Eur J Nutr ; 60(4): 1737-1759, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32797252

RESUMO

BACKGROUND: Cystic Fibrosis (CF) is an autosomal recessive disorder with life-threatening organ manifestations. 87% of CF patients develop exocrine pancreas insufficiency, frequently starting in utero and requiring lifelong pancreatic enzyme substitution. 99% develop progressive lung disease, and 20-60% CF-related liver disease, from mild steatosis to cirrhosis. Characteristically, pancreas, liver and lung are linked by choline metabolism, a critical nutrient in CF. Choline is a tightly regulated tissue component in the form of phosphatidylcholine (Ptd'Cho) and sphingomyelin (SPH) in all membranes and many secretions, particularly of liver (bile, lipoproteins) and lung (surfactant, lipoproteins). Via its downstream metabolites, betaine, dimethylglycine and sarcosine, choline is the major one-carbon donor for methionine regeneration from homocysteine. Methionine is primarily used for essential methylation processes via S-adenosyl-methionine. CLINICAL IMPACT: CF patients with exocrine pancreas insufficiency frequently develop choline deficiency, due to loss of bile Ptd'Cho via feces. ~ 50% (11-12 g) of hepatic Ptd'Cho is daily secreted into the duodenum. Its re-uptake requires cleavage to lyso-Ptd'Cho by pancreatic and small intestinal phospholipases requiring alkaline environment. Impaired CFTR-dependent bicarbonate secretion, however, results in low duodenal pH, impaired phospholipase activity, fecal Ptd'Cho loss and choline deficiency. Low plasma choline causes decreased availability for parenchymal Ptd'Cho metabolism, impacting on organ functions. Choline deficiency results in hepatic choline/Ptd'Cho accretion from lung tissue via high density lipoproteins, explaining the link between choline deficiency and lung function. Hepatic Ptd'Cho synthesis from phosphatidylethanolamine by phosphatidylethanolamine-N-methyltransferase (PEMT) partly compensates for choline deficiency, but frequent single nucleotide polymorphisms enhance choline requirement. Additionally, small intestinal bacterial overgrowth (SIBO) frequently causes intraluminal choline degradation in CF patients prior to its absorption. As adequate choline supplementation was clinically effective and adult as well as pediatric CF patients suffer from choline deficiency, choline supplementation in CF patients of all ages should be evaluated.


Assuntos
Fibrose Cística , Microbioma Gastrointestinal , Adulto , Criança , Colina , Humanos , Fígado , Pâncreas , Fosfatidiletanolamina N-Metiltransferase
4.
Nutr Diet ; 77(3): 368-372, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31044529

RESUMO

AIM: Physiological homocysteine (Hcy) concentrations depend on several factors, both dietary (including folate and choline intake) and biological (such as polymorphism of the genes involved in Hcy metabolism). This study aimed to thus test the associations between genes functionally linked with Hcy metabolism (MTHFR, BHMT and PEMT), folate and choline intakes, and total Hcy (tHcy) concentrations of healthy pregnant women. METHODS: One hundred and three healthy Polish women aged 18-44 years, in the third trimester of pregnancy, were enrolled. RESULTS: Mean blood tHcy and glutathione (GSH) concentrations were 8.08 ± 3.25 µM and 4.84 ± 1.21 µM, respectively. Concentrations of tHcy were found to be lower in the women who were taking folic acid supplements than in those who did not take these supplements (7.42 ± 1.78 µM vs 9.28 ± 4.42 µM, P < 0.05). There were no associations found between the examined parameters and BHMT (rs7356530), MTHFR (rs1801133) and PEMT (rs12325817) alone. However, blood tHcy concentrations differed in the PEMT genotype subgroups when choline and folate intakes were considered: respectively, 25% and 20% lower levels were observed in the C allele carriers who met their needs of choline or folate than in those who did not take enough these nutrients (P < 0.05 for both associations). CONCLUSIONS: This study suggests that choline and folate intakes might interact with MTHFR, BHMT and PEMT polymorphisms to determine tHcy and GSH blood concentrations in healthy pregnant women.


Assuntos
Betaína-Homocisteína S-Metiltransferase/genética , Colina/administração & dosagem , Ácido Fólico/administração & dosagem , Homocisteína/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Fosfatidiletanolamina N-Metiltransferase/genética , Adolescente , Adulto , Feminino , Genótipo , Glutationa/sangue , Humanos , Polônia , Polimorfismo Genético , Gravidez , Terceiro Trimestre da Gravidez , Adulto Jovem
5.
Nutrients ; 11(11)2019 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-31671528

RESUMO

Polyunsaturated fatty acids (PUFAs) play important roles in health and disease. PUFA levels are influenced by nutrition and genetic factors. The relationship between PUFA composition in red blood cells (RBCs) and genetic variations involved in PUFA metabolism has not been investigated in children with obesity. This study evaluated the association between several genetic variations and PUFA levels in RBCs in children with obesity. One hundred ninety-six children with obesity (101 females, 95 males) were evaluated using anthropometric measurements, dietary intakes, plasma and RBC PUFA quantification, blood biochemistry, and 55 single nucleotide polymorphisms within 14 genes. phosphatidylethanolamine N-methyltransferase (PEMT) rs1109859 and methylenetetrahydrofolate reductase gene (MTHFR) rs4846052 genotypes were associated with PUFA levels in RBCs. PUFA intake did not influence the RBC eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) levels. Higher RBC DHA and EPA levels were observed for PEMT rs1109859 GG and GA genotypes versus the AA genotype. Higher levels of RBC DHA, EPA, arachidonic acid (ARA), and linoleic acid (LA) and were observed for MTHFR rs4846052 TT genotype versus TC and CC genotypes. Genetic variations in PEMT rs1109859 and MTHFR rs4846052 were associated with different PUFA levels in RBC membranes and are estimators for PUFA species in RBCs. Further research is needed to establish whether these genotype-specific alterations are specific to overweight children.


Assuntos
Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-6/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Obesidade Infantil/genética , Fosfatidiletanolamina N-Metiltransferase/genética , Polimorfismo de Nucleotídeo Único , Adolescente , Membrana Celular/química , Membrana Celular/metabolismo , Criança , Eritrócitos/metabolismo , Ácidos Graxos Ômega-3/química , Ácidos Graxos Ômega-6/química , Feminino , Humanos , Masculino
6.
Br J Nutr ; 122(11): 1221-1229, 2019 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-31782377

RESUMO

The major facilitator superfamily domain 2a protein was identified recently as a lysophosphatidylcholine (LPC) symporter with high affinity for LPC species enriched with DHA (LPC-DHA). To test the hypothesis that reproductive state and choline intake influence plasma LPC-DHA, we performed a post hoc analysis of samples available through 10 weeks of a previously conducted feeding study, which provided two doses of choline (480 and 930 mg/d) to non-pregnant (n 21), third-trimester pregnant (n 26), and lactating (n 24) women; all participants consumed 200 mg of supplemental DHA and 22 % of their daily choline intake as 2H-labelled choline. The effects of reproductive state and choline intake on total LPC-DHA (expressed as a percentage of LPC) and plasma enrichments of labelled LPC and LPC-DHA were assessed using mixed and generalised linear models. Reproductive state interacted with time (P = 0·001) to influence total LPC-DHA, which significantly increased by week 10 in non-pregnant women, but not in pregnant or lactating women. Contrary to total LPC-DHA, patterns of labelled LPC-DHA enrichments were discordant between pregnant and lactating women (P < 0·05), suggestive of unique, reproductive state-specific mechanisms that result in reduced production and/or enhanced clearance of LPC-DHA during pregnancy and lactation. Regardless of the reproductive state, women consuming 930 v. 480 mg choline per d exhibited no change in total LPC-DHA but higher d3-LPC-DHA (P = 0·02), indicating that higher choline intakes favour the production of LPC-DHA from the phosphatidylethanolamine N-methyltransferase pathway of phosphatidylcholine biosynthesis. Our results warrant further investigation into the effect of reproductive state and dietary choline on LPC-DHA dynamics and its contribution to DHA status.


Assuntos
Colina/administração & dosagem , Ácidos Docosa-Hexaenoicos/sangue , Fosfatidilcolinas/sangue , Reprodução/fisiologia , Adulto , Deutério , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Feminino , Genótipo , Humanos , Lactação/sangue , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Gravidez , Terceiro Trimestre da Gravidez
7.
Biochim Biophys Acta Mol Basis Dis ; 1865(1): 14-25, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30300671

RESUMO

Phosphatidylethanolamine N-methyltransferase (PEMT) converts phosphatidylethanolamine (PE) to phosphatidylcholine (PC), mainly in the liver. Pemt-/- mice are protected from high-fat diet (HFD)-induced obesity and insulin resistance, but develop severe non-alcoholic fatty liver disease (NAFLD) when fed a HFD, mostly due to impaired VLDL secretion. Oxidative stress is thought to be an essential factor in the progression from simple steatosis to steatohepatitis. Vitamin E is an antioxidant that has been clinically used to improve NAFLD pathology. Our aim was to determine whether supplementation of the diet with vitamin E could attenuate HFD-induced hepatic steatosis and its progression to NASH in Pemt-/- mice. Treatment with vitamin E (0.5 g/kg) for 3 weeks improved VLDL-TG secretion and normalized cholesterol metabolism, but failed to reduce hepatic TG content. Moreover, vitamin E treatment was able to reduce hepatic oxidative stress, inflammation and fibrosis. We also observed abnormal ceramide metabolism in Pemt-/- mice fed a HFD, with elevation of ceramides and other sphingolipids and higher expression of mRNAs for acid ceramidase (Asah1) and ceramide kinase (Cerk). Interestingly, vitamin E supplementation restored Asah1 and Cerk mRNA and sphingolipid levels. Together this study shows that vitamin E treatment efficiently prevented the progression from simple steatosis to steatohepatitis in mice lacking PEMT.


Assuntos
Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Vitamina E/metabolismo , Vitamina E/farmacologia , Ceramidase Ácida , Animais , Antioxidantes/farmacologia , Colesterol/metabolismo , Dieta Hiperlipídica , Suplementos Nutricionais , Modelos Animais de Doenças , Progressão da Doença , Fígado Gorduroso/metabolismo , Fibrose/tratamento farmacológico , Inflamação/tratamento farmacológico , Resistência à Insulina , Metabolismo dos Lipídeos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfatidiletanolamina N-Metiltransferase/genética , Fosfotransferases (Aceptor do Grupo Álcool) , RNA Mensageiro , Vitamina E/administração & dosagem
8.
J Nutr ; 148(10): 1513-1520, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30281112

RESUMO

Background: Phosphatidylethanolamine N-methyltransferase (PEMT) converts phosphatidylethanolamine to phosphatidylcholine. Pemt-/-/low density lipoprotein receptor (Ldlr)-/- mice have significantly reduced plasma lipids and are protected against atherosclerosis. Recent studies have shown that choline can be metabolized by the gut flora into trimethylamine-N-oxide (TMAO), which is an emerging risk factor for atherosclerosis. Objective: The objective of this study was to determine whether ectopic hepatic PEMT expression or choline supplementation would promote atherosclerosis in Pemt-/-/Ldlr-/- mice. Methods: Male 8- to 10-wk-old Pemt+/+/Ldlr-/- (SKO) and Pemt-/-/Ldlr-/- (DKO) mice were injected with an adeno-associated virus (AAV) expressing green fluorescent protein (GFP) or human PEMT and fed a Western diet (40% of calories from fat, 0.5% cholesterol) for 8 wk. In a separate experiment, 8- to 10-wk-old SKO and half of the DKO male mice were fed a Western diet with normal (3 g/kg) choline for 12 wk. The remaining DKO mice [choline-supplemented (CS) DKO] were fed a CS Western diet (10 g choline/kg). Plasma lipid concentrations, choline metabolites, and aortic atherosclerosis were measured. Results: Plasma cholesterol, plasma TMAO, and aortic atherosclerosis were reduced by 60%, 40%, and 80%, respectively, in DKO mice compared with SKO mice. AAV-PEMT administration increased plasma cholesterol and TMAO by 30% and 40%, respectively, in DKO mice compared with AAV-GFP-treated DKO mice. Furthermore, AAV-PEMT-injected DKO mice developed atherosclerotic lesions similar to SKO mice. In the second study, there was no difference in atherosclerosis or plasma cholesterol between DKO and CS-DKO mice. However, plasma TMAO concentrations were increased 2.5-fold in CS-DKO mice compared with DKO mice. Conclusions: Reintroducing hepatic PEMT reversed the atheroprotective phenotype of DKO mice. Choline supplementation did not increase atherosclerosis or plasma cholesterol in DKO mice. Our data suggest that plasma TMAO does not induce atherosclerosis when plasma cholesterol is low. Furthermore, this is the first report to our knowledge that suggests that de novo choline synthesis alters TMAO status.


Assuntos
Aterosclerose/metabolismo , Colesterol/sangue , Colina/farmacologia , Fígado/metabolismo , Metilaminas/sangue , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Receptores de LDL/metabolismo , Animais , Aorta , Aterosclerose/etiologia , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Colesterol na Dieta/administração & dosagem , Colina/metabolismo , Dieta Ocidental , Suplementos Nutricionais , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidiletanolamina N-Metiltransferase/farmacologia , Fosfatidiletanolaminas/metabolismo
9.
J Lipid Res ; 56(9): 1701-10, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26113536

RESUMO

Mice that lack phosphatidylethanolamine N-methyltransferase (Pemt(-/-) mice) are protected from high-fat (HF) diet-induced obesity. HF-fed Pemt(-/-) mice show higher oxygen consumption and heat production, indicating that more energy might be utilized for thermogenesis and might account for the resistance to diet-induced weight gain. To test this hypothesis, HF-fed Pemt(-/-) and Pemt(+/+) mice were challenged with acute cold exposure at 4°C. Unexpectedly, HF-fed Pemt(-/-) mice developed hypothermia within 3 h of cold exposure. In contrast, chow-fed Pemt(-/-) mice, possessing similar body mass, maintained body temperature. Lack of PEMT did not impair the capacity for thermogenesis in skeletal muscle or brown adipose tissue. Plasma catecholamines were not altered by Pemt genotype, and stimulation of lipolysis was intact in brown and white adipose tissue of Pemt(-/-) mice. HF-fed Pemt(-/-) mice also developed higher systolic blood pressure, accompanied by reduced cardiac output. Choline supplementation reversed the cold-induced hypothermia in HF-fed Pemt(-/-) mice with no effect on blood pressure. Plasma glucose levels were ∼50% lower in HF-fed Pemt(-/-) mice compared with Pemt(+/+) mice. Choline supplementation normalized plasma hypoglycemia and the expression of proteins involved in gluconeogenesis. We propose that cold-induced hypothermia in HF-fed Pemt(-/-) mice is linked to plasma hypoglycemia due to compromised hepatic glucose production.


Assuntos
Metabolismo Energético/genética , Hipotermia/genética , Obesidade/metabolismo , Fosfatidiletanolamina N-Metiltransferase/genética , Animais , Temperatura Baixa , Dieta Hiperlipídica , Glucose/metabolismo , Humanos , Hipotermia/metabolismo , Hipotermia/patologia , Lipólise/genética , Fígado/metabolismo , Fígado/patologia , Camundongos , Obesidade/genética , Obesidade/patologia , Consumo de Oxigênio/genética
10.
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
11.
Am J Clin Nutr ; 100(4): 1069-74, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25240073

RESUMO

BACKGROUND: Low maternal choline intake and blood concentration may be risk factors for having a child with a neural tube defect (NTD); however, the data are inconsistent. This is an important question to resolve because choline, if taken periconceptionally, might add to the protective effect currently being achieved by folic acid. OBJECTIVE: We examined the relation between NTDs, choline status, and genetic polymorphisms reported to influence de novo choline synthesis to investigate claims that taking choline periconceptionally could reduce NTD rates. DESIGN: Two study groups of pregnant women were investigated: women who had a current NTD-affected pregnancy (AP; n = 71) and unaffected controls (n = 214) and women who had an NTD in another pregnancy but not in the current pregnancy [nonaffected pregnancy (NAP); n = 98] and unaffected controls (n = 386). Blood samples to measure betaine and total choline concentrations and single nucleotide polymorphisms related to choline metabolism were collected at their first prenatal visit. RESULTS: Mean (±SD) plasma total choline concentrations in the AP (2.8 ± 1.0 mmol/L) and control (2.9 ± 0.9 mmol/L) groups did not differ significantly. Betaine concentrations were not significantly different between the 2 groups. Total choline and betaine in the NAP group did not differ from controls. Cases were significantly more likely to have the G allele of phosphatidylethanolamine-N-methyltransferase (PEMT; V175M, +5465 G>A) rs7946 (P = 0.02). CONCLUSIONS: Our results indicate that maternal betaine and choline concentrations are not strongly associated with NTD risk. The association between PEMT rs7946 and NTDs requires confirmation. The addition of choline to folic acid supplements may not further reduce NTD risk.


Assuntos
Colina/sangue , Defeitos do Tubo Neural/genética , Polimorfismo de Nucleotídeo Único , Adulto , Betaína/sangue , Estudos de Casos e Controles , Suplementos Nutricionais , Feminino , Ácido Fólico/sangue , Genoma Humano , Genótipo , Humanos , Modelos Logísticos , Fosfatidiletanolamina N-Metiltransferase/sangue , Fosfatidiletanolamina N-Metiltransferase/genética , Gravidez , Fatores de Risco , Seleção Genética , Adulto Jovem
12.
FASEB J ; 28(10): 4312-23, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24963152

RESUMO

Maternal choline supplementation (MCS) induces lifelong cognitive benefits in the Ts65Dn mouse, a trisomic mouse model of Down syndrome and Alzheimer's disease. To gain insight into the mechanisms underlying these beneficial effects, we conducted a study to test the hypothesis that MCS alters choline metabolism in adult Ts65Dn offspring. Deuterium-labeled methyl-d9-choline was administered to adult Ts65Dn and disomic (2N) female littermates born to choline-unsupplemented or choline-supplemented Ts65Dn dams. Enrichment of d9-choline metabolites (derived from intact choline) and d3 + d6-choline metabolites [produced when choline-derived methyl groups are used by phosphatidylethanolamine N-methyltransferase (PEMT)] was measured in harvested tissues. Adult offspring (both Ts65Dn and 2N) of choline-supplemented (vs. choline-unsupplemented) dams exhibited 60% greater (P≤0.007) activity of hepatic PEMT, which functions in de novo choline synthesis and produces phosphatidylcholine (PC) enriched in docosahexaenoic acid. Higher (P<0.001) enrichment of PEMT-derived d3 and d6 metabolites was detected in liver, plasma, and brain in both genotypes but to a greater extent in the Ts65Dn adult offspring. MCS also yielded higher (P<0.05) d9 metabolite enrichments in liver, plasma, and brain. These data demonstrate that MCS exerts lasting effects on offspring choline metabolism, including up-regulation of the hepatic PEMT pathway and enhanced provision of choline and PEMT-PC to the brain.


Assuntos
Colina/farmacocinética , Síndrome de Down/metabolismo , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Animais , Colina/administração & dosagem , Colina/farmacologia , Colina/uso terapêutico , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/metabolismo , Síndrome de Down/tratamento farmacológico , Feminino , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilcolinas/metabolismo , Gravidez , Distribuição Tecidual
13.
J Nutr ; 144(3): 252-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24368431

RESUMO

Dietary choline is required for proper structure and dynamics of cell membranes, lipoprotein synthesis, and methyl-group metabolism. In mammals, choline is synthesized via phosphatidylethanolamine N-methyltransferase (Pemt), which converts phosphatidylethanolamine to phosphatidylcholine. Pemt(-/-) mice have impaired VLDL secretion and developed fatty liver when fed a high-fat (HF) diet. Because of the reduction in plasma lipids, Pemt(-/-)/low-density lipoprotein receptor knockout (Ldlr(-/-)) mice are protected from atherosclerosis. The goal of this study was to investigate the importance of dietary choline in the metabolic phenotype of Pemt(-/-)/Ldlr(-/-) male mice. At 10-12 wk of age, Pemt(+/+)/Ldlr(-/-) (HF(+/+)) and half of the Pemt(-/-)/Ldlr(-/-) (HF(-/-)) mice were fed an HF diet with normal (1.3 g/kg) choline. The remaining Pemt(-/-)/Ldlr(-/-) mice were fed an HF diet supplemented (5 g/kg) with choline (HFCS(-/-) mice). The HF diet contained 60% of calories from fat and 1% cholesterol, and the mice were fed for 16 d. HF(-/-) mice lost weight and developed hepatomegaly, steatohepatitis, and liver damage. Hepatic concentrations of free cholesterol, cholesterol-esters, and triglyceride (TG) were elevated by 30%, 1.1-fold and 3.1-fold, respectively, in HF(-/-) compared with HF(+/+) mice. Choline supplementation normalized hepatic cholesterol, but not TG, and dramatically improved liver function. The expression of genes involved in cholesterol transport and esterification increased by 50% to 5.6-fold in HF(-/-) mice when compared with HF(+/+) mice. Markers of macrophages, oxidative stress, and fibrosis were elevated in the HF(-/-) mice. Choline supplementation normalized the expression of these genes. In conclusion, HF(-/-) mice develop liver failure associated with altered cholesterol metabolism when fed an HF/normal choline diet. Choline supplementation normalized cholesterol metabolism, which was sufficient to prevent nonalcoholic steatohepatitis development and improve liver function. Our data suggest that choline can promote liver health by maintaining cholesterol homeostasis.


Assuntos
Colesterol/metabolismo , Colina/administração & dosagem , Dieta Hiperlipídica/efeitos adversos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Animais , Ésteres do Colesterol/metabolismo , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/etiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Fosfatidiletanolamina N-Metiltransferase/sangue , Receptores de LDL/sangue , Triglicerídeos/metabolismo
14.
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
15.
Am J Clin Nutr ; 97(4): 718-27, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23446897

RESUMO

BACKGROUND: Phosphatidylcholine (PC) produced via the S-adenosylmethionine-dependent phosphatidylethanolamine (PE) N-methyltransferase (PEMT) pathway is enriched with docosahexaenoic acid (DHA). DHA plays a critical role in fetal development and is linked to health endpoints in adulthood. It is unknown whether choline, which can serve as a source of S-adenosylmethionine methyl groups, influences PC-DHA or the PC:PE ratio in pregnant and nonpregnant women. OBJECTIVE: This study tested whether choline intake affects indicators of choline-related lipid metabolism, including erythrocyte and plasma PC-DHA and PC:PE ratios, in pregnant women in the third trimester and nonpregnant women. DESIGN: Pregnant (n = 26) and nonpregnant (n = 21) women consumed 480 or 930 mg choline/d and a daily DHA supplement for 12 wk. Blood was collected at baseline and at the midpoint and end of the study. PC-DHA was analyzed as the proportion of total PC fatty acids. RESULTS: Pregnant women had greater (P = 0.002) PC-DHA concentrations than did nonpregnant women at baseline. The proportion of erythrocyte and plasma PC-DHA increased (P ≤ 0.002) in pregnant and nonpregnant women regardless of choline intake. However, in nonpregnant women, consumption of 930 mg choline/d led to greater (P < 0.001) erythrocyte PC-DHA and a more rapid increase (P < 0.001) in plasma PC-DHA. Lower (P = 0.001-0.024) erythrocyte and plasma PC:PE in pregnant women was not modified by choline intake. CONCLUSIONS: A higher choline intake may increase PEMT activity, resulting in greater PC-DHA enrichment of the PC molecule in nonpregnant women. Increased production of PC-DHA during pregnancy indicates elevated PEMT activity and a higher demand for methyl donors. This trial was registered at clinicaltrials.gov as NCT01127022.


Assuntos
Colina/farmacologia , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/sangue , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipotrópicos/farmacologia , Fosfatidilcolinas/sangue , Gravidez/sangue , Eritrócitos/metabolismo , Feminino , Humanos , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Fosfatidiletanolaminas/sangue , Gravidez/metabolismo , Terceiro Trimestre da Gravidez
16.
Biochim Biophys Acta ; 1831(3): 626-32, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22877991

RESUMO

Phosphatidylethanolamine N-methyltransferase (PEMT) catalyzes the methylation of phosphatidylethanolamine to phosphatidylcholine (PC). This 22.3 kDa protein is localized to the endoplasmic reticulum and mitochondria associated membranes of liver. The supply of the substrates AdoMet and phosphatidylethanolamine, and the product AdoHcy, can regulate the activity of PEMT. Estrogen has been identified as a positive activator, and Sp1 as a negative regulator, of transcription of the PEMT gene. Targeted inactivation of the PEMT gene produced mice that had a mild phenotype when fed a chow diet. However, when Pemt(-/-) mice were fed a choline-deficient diet steatohepatitis and liver failure developed after 3 days. The steatohepatitis was due to a decreased ratio of PC to phosphatidylethanolamine that caused leakage from the plasma membrane of hepatocytes. Pemt(-/-) mice exhibited attenuated secretion of very low-density lipoproteins and homocysteine. Pemt(-/-) mice bred with mice that lacked the low-density lipoprotein receptor, or apolipoprotein E were protected from high fat/high cholesterol-induced atherosclerosis. Surprisingly, Pemt(-/-) mice were protected from high fat diet-induced obesity and insulin resistance compared to wildtype mice. If the diet were supplemented with additional choline, the protection against obesity/insulin resistance in Pemt(-/-) mice was eliminated. Humans with a Val-to-Met substitution in PEMT at residue 175 may have increased susceptibility to nonalcoholic liver disease. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metabolism.


Assuntos
Retículo Endoplasmático/metabolismo , Fígado Gorduroso/metabolismo , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , Fosfatidiletanolamina N-Metiltransferase/deficiência , Animais , Colina/metabolismo , Dieta Hiperlipídica/efeitos adversos , Retículo Endoplasmático/patologia , Estrogênios/metabolismo , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Humanos , Fígado/patologia , Camundongos , Camundongos Knockout , Mitocôndrias Hepáticas/patologia , Fosfatidilcolinas/metabolismo , Fosfatidiletanolamina N-Metiltransferase/genética , Fosfatidiletanolaminas/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
17.
J Biol Chem ; 288(2): 837-47, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23179947

RESUMO

Biosynthesis of hepatic choline via phosphatidylethanolamine N-methyltransferase (PEMT) plays an important role in the development of type 2 diabetes and obesity. We investigated the mechanism(s) by which choline modulates insulin sensitivity. PEMT wild-type (Pemt(+/+)) and knock-out (Pemt(-/-)) mice received either a high fat diet (HF; 60% kcal of fat) or a high fat, high choline diet (HFHC; 4 g of choline/kg of HF diet) for 1 week. Hepatic insulin signaling and glucose and lipid homeostasis were investigated. Glucose and insulin intolerance occurred in Pemt(-/-) mice fed the HFHC diet, but not in their Pemt(-/-) littermates fed the HF diet. Plasma glucagon was elevated in Pemt(-/-) mice fed the HFHC diet compared with Pemt(-/-) mice fed the HF diet, concomitant with increased hepatic expression of glucagon receptor, phosphorylated AMP-activated protein kinase (AMPK), and phosphorylated insulin receptor substrate 1 at serine 307 (IRS1-s307). Gluconeogenesis and mitochondrial oxidative stress were markedly enhanced, whereas glucose oxidation and triacylglycerol biosynthesis were diminished in Pemt(-/-) mice fed the HFHC diet. A glucagon receptor antagonist (2-aminobenzimidazole) attenuated choline-induced hyperglycemia and insulin intolerance and blunted up-regulation of phosphorylated AMPK and IRS1-s307. Choline induces glucose and insulin intolerance in Pemt(-/-) mice through modulating plasma glucagon and its action in liver.


Assuntos
Colina/administração & dosagem , Glucagon/fisiologia , Resistência à Insulina , Fígado/efeitos dos fármacos , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Animais , Sequência de Bases , Colina/farmacologia , Primers do DNA , Gluconeogênese/efeitos dos fármacos , Teste de Tolerância a Glucose , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidiletanolamina N-Metiltransferase/genética
18.
Ann Nutr Metab ; 61(3): 254-8, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23183298

RESUMO

In 1850, Theodore Gobley, working in Paris, described a substance, 'lecithine', which he named after the Greek 'lekithos' for egg yolk. Adolph Strecker noted in 1862 that when lecithin from bile was heated, it generated a new nitrogenous chemical that he named 'choline'. Three years later, Oscar Liebreich identified a new substance, 'neurine', in the brain. After a period of confusion, neurine and choline were found to be the same molecule, and the name choline was adapted. Lecithin was eventually characterized chemically as being phosphatidylcholine. In 1954, Eugene Kennedy described the cytidine 5-dihphosphocholine pathway by which choline is incorporated into phosphatidylcholine. A second route, the phosphatidylethanolamine-N-methyltransferase pathway, was identified by Jon Bremer and David Greenberg in 1960. The role of choline as part of the neurotransmitter acetylcholine was established by Otto Loewi and Henry Dale. Working in the 1930s at the University of Toronto, Charles Best showed that choline prevented fatty liver in dogs and rats. The importance of choline as an essential nutrient for human health was determined in the 1990s through controlled feeding studies in humans. Recently, an understanding of the role of genetic variation in setting the dietary requirement for choline in people is being unraveled.


Assuntos
Colina/química , Colina/história , Colina/farmacologia , Acetilcolina/metabolismo , Animais , Bile/metabolismo , Cães , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/prevenção & controle , História do Século XIX , História do Século XX , História do Século XXI , Humanos , Lecitinas/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neurotransmissores/metabolismo , Valor Nutritivo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Ratos
19.
Hepatology ; 55(4): 1094-102, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22095799

RESUMO

UNLABELLED: A major predictor of failed liver resection and transplantation is nonalcoholic fatty liver disease (NAFLD). NAFLD is linked to a wide spectrum of diseases including obesity and diabetes that are increasingly prevalent in Western populations. Thus, it is important to develop therapies aimed at improving posthepatectomy outcomes in patients with NAFLD, as well as to improve the evaluation of patients slated for hepatic surgery. Decreased hepatic phosphatidylcholine (PC) content and decreased ratio of hepatic PC to phosphatidylethanolamine (PE) have previously been linked to NAFLD. To determine if decreased hepatic PC/PE could predict survival after hepatectomy, we used mouse models lacking key enzymes in PC biosynthesis, namely, phosphatidylethanolamine N-methyltransferase and hepatic-specific CTP:phosphocholine cytidylyltransferase α. These mice were fed a high-fat diet to induce NAFLD. We then performed a 70% partial hepatectomy and monitored postoperative survival. We identified hepatic PC/PE to be inversely correlated with the development of steatosis and inflammation in the progression of NAFLD. Decreased hepatic PC/PE before surgery was also strongly associated with decreased rates of survival after partial hepatectomy. Choline supplementation to the diet increased hepatic PC/PE in Pemt(-/-) mice with NAFLD, decreased inflammation, and increased the survival rate after partial hepatectomy. CONCLUSION: Decreased hepatic PC/PE is a predictor of NAFLD and survival following partial hepatectomy. Choline supplementation may serve as a potential therapy to prevent the progression of NAFLD and to improve postoperative outcome after liver surgery.


Assuntos
Progressão da Doença , Fígado Gorduroso/mortalidade , Fígado Gorduroso/cirurgia , Hepatectomia , Fígado/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , Animais , Colina/administração & dosagem , Colina/uso terapêutico , Colina-Fosfato Citidililtransferase/deficiência , Colina-Fosfato Citidililtransferase/genética , Gorduras na Dieta/efeitos adversos , Suplementos Nutricionais , Modelos Animais de Doenças , Fígado Gorduroso/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Fosfatidiletanolamina N-Metiltransferase/deficiência , Fosfatidiletanolamina N-Metiltransferase/genética , Valor Preditivo dos Testes , Taxa de Sobrevida
20.
Nutr Res ; 31(7): 572-8, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21840474

RESUMO

The reactions of the methionine/homocysteine pathway are mediated by several enzymes, including phosphatidylethanolamine N-methyltransferase, cystathionine ß-synthase, and betaine-homocysteine methyltransferase. Homocysteine homeostasis is regulated by these enzymes. We hypothesized here that the protein and folic acid content in the maternal diet affects methionine/homocysteine metabolism in the progeny. To test this hypothesis, pregnant rats were fed a diet with normal protein and normal folic acid levels (a modified casein-based AIN-93G diet), a protein-restricted and normal folic acid diet, a protein-restricted and folic acid-supplemented diet, or a normal protein and folic acid-supplemented diet. The progeny were fed either the modified AIN-93G diet or a high-fat lard-based diet. Progeny were analyzed for expression of the phosphatidylethanolamine N-methyltransferase, cystathionine ß-synthase, and betaine-homocysteine methyltransferase genes in the liver and for serum homocysteine concentration. Interactions between prenatal and postnatal nutrition were also determined. The progeny of the dams fed the diets supplemented with folic acid showed decreased expression of all 3 genes (P < .001). An interaction effect between the protein and folic acid content in the maternal diet contributed to this down-regulation (P < .001), and the postweaning diet modified these effects. Serum homocysteine concentrations were approximately 15% higher in the male rats (P < .01), but neither prenatal nutrition nor the postweaning diet affected it significantly. We conclude that maternal diet during gestation has an important effect on the transcription level of these 3 genes, but changes in gene expression were not associated with significant changes in progeny homocysteine concentrations.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal , Cistationina beta-Sintase/metabolismo , Dieta , Ácido Fólico/farmacologia , Expressão Gênica/efeitos dos fármacos , Metiltransferases/metabolismo , Gravidez , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Betaína-Homocisteína S-Metiltransferase/metabolismo , Cistationina beta-Sintase/genética , Dieta Hiperlipídica , Dieta com Restrição de Proteínas , Gorduras na Dieta/farmacologia , Proteínas Alimentares/farmacologia , Suplementos Nutricionais , Regulação para Baixo , Feminino , Homeostase , Homocisteína/sangue , Fígado/metabolismo , Masculino , Metionina/metabolismo , Metiltransferases/genética , Fosfatidiletanolamina N-Metiltransferase/genética , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Fenômenos Fisiológicos da Nutrição Pré-Natal , Ratos Wistar , Complexo Vitamínico B/farmacologia , Desmame
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA