Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Biomed Pharmacother ; 127: 110121, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32407984

RESUMO

OBJECTIVE: Nicorandil exerts a protective effect against coronary microvascular dysfunction in acute myocardial infarction (AMI) patients. However, the mechanism and effect of nicorandil in hyperhomocysteinemia (HHcy) AMI patients remain unclear. METHODS: C57/BL6 mice with mild to moderate HHcy and human coronary artery endothelial cells (HCAECs) cotreated with HHcy (1 mmol/L) for 24 h and hypoxia for 6 h were selected as models. Small animal ultrasound detection was used to compare cardiac function. CD31 immunofluorescence staining and tomato lectin staining were used to assess the number of microcirculation changes in vivo. MTT, tube formation and western blotting assays were used to evaluate the effect of nicorandil on HCAECs and the PI3K/Akt/eNOS pathway. RESULTS: The results showed that nicorandil improved cell viability and p-PI3K/PI3K, p-Akt/Akt, and p-eNOS/eNOS expression in the vitro HHcy and hypoxia models. The beneficial effects of nicorandil on HCAECs could be inhibited by the phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 and the nitric oxide synthase (NOS) inhibitor L-NAME. In vivo, nicorandil improved the left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) in the post-HHcy + MI model, and the levels of CD31 and tomato lectin expression were higher in the nicorandil treatment group. The effectiveness of nicorandil was inhibited in the PI3K and L-NAME groups. CONCLUSION: The results suggest that nicorandil improves Hcy-induced coronary microvascular dysfunction through the PI3K/Akt/eNOS signalling pathway.


Assuntos
Hiper-Homocisteinemia/prevenção & controle , Microcirculação/fisiologia , Nicorandil/farmacologia , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Células Endoteliais/efeitos dos fármacos , Homocisteína , Humanos , Hiper-Homocisteinemia/induzido quimicamente , Hipóxia , Masculino , Camundongos , Morfolinas/farmacologia , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/prevenção & controle , NG-Nitroarginina Metil Éster/farmacologia , Nicorandil/antagonistas & inibidores , Lectinas de Plantas/biossíntese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Função Ventricular Esquerda/fisiologia
2.
Nutrients ; 12(5)2020 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-32349312

RESUMO

Cardiovascular morbidity and mortality are several-fold higher in patients with advanced chronic kidney disease (CKD) and end-stage renal disease (ESRD) than in the general population. Hyperhomocysteinemia has undoubtedly a central role in such a prominent cardiovascular burden. The levels of homocysteine are regulated by methyl donors (folate, methionine, choline, betaine), and cofactors (vitamin B6, vitamin B12,). Uremia-induced hyperhomocysteinemia has as its main targets DNA methyltransferases, and this leads to an altered epigenetic control of genes regulated through methylation. In renal patients, the epigenetic landscape is strictly correlated with the uremic phenotype and dependent on dietary intake of micronutrients, inflammation, gut microbiome, inflammatory status, oxidative stress, and lifestyle habits. All these factors are key contributors in methylome maintenance and in the modulation of gene transcription through DNA hypo- or hypermethylation in CKD. This is an overview of the epigenetic changes related to DNA methylation in patients with advanced CKD and ESRD. We explored the currently available data on the molecular dysregulations resulting from altered gene expression in uremia. Special attention was paid to the efficacy of B-vitamins supplementation and dietary intake of methyl donors on homocysteine lowering and cardiovascular protection.


Assuntos
Betaína/administração & dosagem , Colina/administração & dosagem , Metilação de DNA/genética , Suplementos Nutricionais , Ingestão de Alimentos/fisiologia , Epigênese Genética , Ácido Fólico/administração & dosagem , Metionina/administração & dosagem , Fenômenos Fisiológicos da Nutrição/genética , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Vitamina B 12/administração & dosagem , Vitamina B 6/administração & dosagem , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/prevenção & controle , Humanos , Hiper-Homocisteinemia/etiologia , Hiper-Homocisteinemia/prevenção & controle , Falência Renal Crônica , Fenômenos Fisiológicos da Nutrição/fisiologia , Insuficiência Renal Crônica/complicações , Uremia/complicações , Uremia/genética
3.
Ann Clin Transl Neurol ; 6(8): 1435-1444, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31353838

RESUMO

BACKGROUND: Postoperative neurocognitive disorder (PND) is a severe postoperative complication with no effective therapy that affects up to 19-52% of senior patients. Age and surgery type have been identified as risk factors. However, what caused the increased risk in the elderly is poorly understood. METHODS: We utilized a PND model in aged mice undergoing experimental laparotomy with general anesthesia to evaluate the causal relationship between hyperhomocysteinemia and increased PND susceptibility. PND was assessed by Novel Object Tasks, Fear Conditioning Tests, and Barnes Maze Tests. Serum homocysteine (Hcy) as well as vitamin B12 and folate acid levels were tested before, immediately after surgery and from day 1 to day 29 after surgery by ELISA. The effectiveness of preventative strategy including diet supplementation of vitamin B12 + folic acid (Vit B12 + FA) and S-adenosylmethionine (SAM) injection targeting hyperhomocysteinemia were also tested. RESULTS: PND in aged mice lasted for at least 2 weeks after experimental laparotomy, which was not observed in young adult mice. Serum Hcy results indicated a significant correlation between postoperative cognitive performance and perioperative Hcy level. Preoperative supplementation with VB12 and folic acid (FA) in the diet or S-adenosylmethionine (SAM) injection reduced perioperative serum Hcy level and inhibited the development of PND in aged mice. CONCLUSIONS: Serum homocysteine accumulation is a fundamental cause for increased susceptibility of PND in aged mice. Preoperative diet supplementation of VitB12 + FA can effectively reduce PND in aged mice, which may be a promising prophylaxis treatment in clinical settings.


Assuntos
Ácido Fólico/uso terapêutico , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/dietoterapia , Complicações Cognitivas Pós-Operatórias/prevenção & controle , Vitamina B 12/uso terapêutico , Animais , Cognição/efeitos dos fármacos , Suplementos Nutricionais , Ácido Fólico/sangue , Homocisteína/sangue , Hiper-Homocisteinemia/prevenção & controle , Camundongos , Período Pós-Operatório , Período Pré-Operatório , Fatores de Risco , S-Adenosilmetionina/sangue , Vitamina B 12/sangue
4.
J Nutr ; 149(8): 1369-1376, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31111947

RESUMO

BACKGROUND: Hyperhomocysteinemia is associated with increased cardiovascular disease risk. Whole eggs contain several nutrients known to affect homocysteine regulation, including sulfur amino acids, choline, and B vitamins. OBJECTIVE: The aim of this study was to determine the effect of whole eggs and egg components (i.e., egg protein and choline) with respect to 1) homocysteine balance and 2) the hepatic expression and activity of betaine-homocysteine S-methyltransferase (BHMT) and cystathionine ß-synthase (CBS) in a folate-restricted (FR) rat model of hyperhomocysteinemia. METHODS: Male Sprague Dawley rats (n = 48; 6 wk of age) were randomly assigned to a casein-based diet (C; n = 12), a casein-based diet supplemented with choline (C + Cho; 1.3%, wt:wt; n = 12), an egg protein-based diet (EP; n = 12), or a whole egg-based diet (WE; n = 12). At week 2, half of the rats in each of the 4 dietary groups were provided an FR (0 g folic acid/kg) diet and half continued on the folate-sufficient (FS; 0.2 g folic acid/kg) diet for an additional 6 wk. All diets contained 20% (wt:wt) total protein. Serum homocysteine was measured by HPLC and BHMT and CBS expression and activity were evaluated using real-time quantitative polymerase chain reaction, Western blot, and enzyme activity. A 2-factor ANOVA was used for statistical comparisons. RESULTS: Rats fed FR-C exhibited a 53% increase in circulating homocysteine concentrations compared with rats fed FS-C (P < 0.001). In contrast, serum homocysteine did not differ between rats fed FS-C and FR-EP (P = 0.078). Hepatic BHMT activity was increased by 45% and 40% by the EP (P < 0.001) and WE (P = 0.002) diets compared with the C diets, respectively. CONCLUSIONS: Dietary intervention with egg protein prevented elevated circulating homocysteine concentrations in a rat model of hyperhomocysteinemia, due in part to upregulation of hepatic BHMT. These data may support the inclusion of egg protein for dietary recommendations targeting hyperhomocysteinemia prevention.


Assuntos
Betaína-Homocisteína S-Metiltransferase/metabolismo , Proteínas Dietéticas do Ovo/administração & dosagem , Deficiência de Ácido Fólico/metabolismo , Hiper-Homocisteinemia/prevenção & controle , Fígado/enzimologia , Regulação para Cima , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Peso Corporal , Cisteína/sangue , Proteínas Dietéticas do Ovo/metabolismo , Masculino , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
5.
Nutr Res ; 46: 78-87, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29129471

RESUMO

Several B-vitamins act as co-factors in one-carbon metabolism, a pathway that plays a central role in several chronic diseases. However, there is a lack of knowledge of how diet affects markers in one-carbon metabolism. The aim of this study was to explore dietary patterns and components associated with one-carbon metabolites. We hypothesized that intake of whole-grains and fish would be associated with lower Hcy, and higher SAM:SAH ratio due to their nutrient content. We assessed dietary information using a four-day dietary record in 118 men and women with features of the metabolic syndrome. In addition we assessed whole-blood fatty acid composition and plasma alkylresorcinols. Plasma s-adenosylmethionine (SAM), s-adenosylhomocysteine (SAH), homocysteine (Hcy) and vitamin B12 was included as one-carbon metabolism markers. We used principal component analysis (PCA) to explore dietary patterns and multiple linear regression models to examine associations between dietary factors and one-carbon metabolites. PCA separated subjects based on prudent and unhealthy dietary patterns, but the dietary pattern score was not related to the one-carbon metabolites. Whole grain intake was found to be inversely associated to plasma Hcy (-4.7% (-9.3; 0.0), P=.05) and total grain intake tended to be positively associated with SAM and SAH (2.4% (-0.5; 5.5), P=.08; 5.8% (-0.2; 12.1), P=.06, respectively, per SD increase in cereal intake). Fish intake was inversely associated with plasma Hcy and SAH concentrations (-5.4% (-9.7; -0.8), P=.02 and -7.0% (-12.1; -1.5), P=.01, respectively) and positively associated with the SAM:SAH ratio (6.2% (1.6; 11.0), P=.008). In conclusion, intake and fish and whole-grain appear to be associated with a beneficial one-carbon metabolism profile. This indicates that dietary components could play a role in regulation of one-carbon metabolism with a potential impact on disease prevention.


Assuntos
Dieta Saudável , Peixes , Hiper-Homocisteinemia/prevenção & controle , Cooperação do Paciente , S-Adenosil-Homocisteína/sangue , Alimentos Marinhos , Grãos Integrais , Adulto , Animais , Biomarcadores/sangue , Estudos Transversais , Dinamarca/epidemiologia , Registros de Dieta , Ácidos Graxos Insaturados/administração & dosagem , Feminino , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/epidemiologia , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/epidemiologia , Síndrome Metabólica/prevenção & controle , Pessoa de Meia-Idade , Inquéritos Nutricionais , Análise de Componente Principal , Risco , Adulto Jovem
6.
Bull Exp Biol Med ; 162(6): 738-740, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28429210

RESUMO

Effects of prenatal hyperhomocysteinemia on hypothalamic regulation of estrous cycles were studied in female rats. In mature rats exposed to prenatal hyperhomocysteinemia, changes in the catecholamine content in hypothalamic areas responsible for the formation of the preovulatory surge of gonadotropin-releasing hormone were revealed: the level of norepinephrine in the medial preoptic area decreased and concentration of dopamine in the median eminence with arcuate nuclei increased. Administration of melatonin attenuated the observed changes, which can be related to neuroprotective effects of this hormone determined by its antioxidant properties.


Assuntos
Antioxidantes/farmacologia , Ciclo Estral/efeitos dos fármacos , Hiper-Homocisteinemia/prevenção & controle , Melatonina/farmacologia , Fármacos Neuroprotetores/farmacologia , Efeitos Tardios da Exposição Pré-Natal/prevenção & controle , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Núcleo Arqueado do Hipotálamo/fisiopatologia , Catecolaminas/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/fisiopatologia , Hormônio Luteinizante/metabolismo , Eminência Mediana/efeitos dos fármacos , Eminência Mediana/metabolismo , Eminência Mediana/fisiopatologia , Metionina/efeitos adversos , Metionina/metabolismo , Norepinefrina/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Área Pré-Óptica/fisiopatologia , Progesterona/metabolismo , Ratos , Ratos Wistar
7.
Nutrients ; 8(12)2016 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-27973419

RESUMO

Alzheimer's disease (AD) is the major cause of dementia and no preventive or effective treatment has been established to date. The etiology of AD is poorly understood, but genetic and environmental factors seem to play a role in its onset and progression. In particular, factors affecting the one-carbon metabolism (OCM) are thought to be important and elevated homocysteine (Hcy) levels, indicating impaired OCM, have been associated with AD. We aimed at evaluating the role of polymorphisms of key OCM enzymes in the etiology of AD, particularly when intakes of relevant B-vitamins are inadequate. Our review indicates that a range of compensatory mechanisms exist to maintain a metabolic balance. However, these become overwhelmed if the activity of more than one enzyme is reduced due to genetic factors or insufficient folate, riboflavin, vitamin B6 and/or vitamin B12 levels. Consequences include increased Hcy levels and reduced capacity to synthetize, methylate and repair DNA, and/or modulated neurotransmission. This seems to favor the development of hallmarks of AD particularly when combined with increased oxidative stress e.g., in apolipoprotein E (ApoE) ε4 carriers. However, as these effects can be compensated at least partially by adequate intakes of B-vitamins, achieving optimal B-vitamin status for the general population should be a public health priority.


Assuntos
Doença de Alzheimer/etiologia , Medicina Baseada em Evidências , Hiper-Homocisteinemia/fisiopatologia , Metionina/metabolismo , Modelos Biológicos , Polimorfismo Genético , Deficiência de Vitaminas do Complexo B/fisiopatologia , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Animais , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Ferredoxina-NADP Redutase/genética , Ferredoxina-NADP Redutase/metabolismo , Predisposição Genética para Doença , Glicina Hidroximetiltransferase/genética , Glicina Hidroximetiltransferase/metabolismo , Humanos , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/prevenção & controle , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Mutagênese Insercional , Nutrigenômica/métodos , Nutrigenômica/tendências , Estado Nutricional , Polimorfismo de Nucleotídeo Único , Sequências de Repetição em Tandem , Complexo Vitamínico B/metabolismo , Complexo Vitamínico B/uso terapêutico , Deficiência de Vitaminas do Complexo B/dietoterapia , Deficiência de Vitaminas do Complexo B/metabolismo , Deficiência de Vitaminas do Complexo B/prevenção & controle
8.
Amino Acids ; 48(8): 2015-24, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26781304

RESUMO

The purpose of this study was to investigate (1) the impact of tumor growth on homocysteine (Hcy) metabolism, liver oxidative stress and cancer cachexia and, (2) the potential benefits of creatine supplementation in Walker-256 tumor-bearing rats. Three experiments were conducted. First, rats were killed on days 5 (D5), 10 (D10) and 14 (D14) after tumor implantation. In experiment 2, rats were randomly assigned to three groups designated as control (C), tumor-bearing (T) and tumor-bearing supplemented with creatine (TCr). A life span experiment was conducted as the third experiment. Creatine was supplied in drinking water for 21 days (8 g/L) in all cases. Tumor implantation consisted of a suspension of Walker-256 cells (8.0 × 10(7) cells in 0.5 mL of PBS). The progressive increase (P < 0.05) in tumor mass coincided with a progressively lower body weight and higher hepatic oxidative stress; plasma Hcy concentration was 80 % higher (P < 0.05) by 10 days of tumor implantation. Impaired Hcy metabolism was evidenced by decreased hepatic betaine-homocysteine methyltransferase (Bhmt), glycine N-methyltransferase (Gnmt) and cystathionine beta synthase (CBS) gene expression. In contrast, creatine supplementation promoted a 28 % reduction of tumor weight (P < 0.05). Plasma Hcy (C 6.1 ± 0.6, T 10.3 ± 1.5, TCr 6.3 ± 0.9, µmol/L) and hepatic oxidative stress were lower in the TCr group compared to T. Creatine supplementation was unable to decrease Hcy concentration and to increase SAM/SAH ratio in tumor tissue. These data suggest that creatine effects on hepatic impaired Hcy metabolism promoted by tumor cell inoculation are responsible to decrease plasma Hcy in tumor-bearing rats. In conclusion, Walker-256 tumor growth is associated with progressive hyperhomocysteinemia, body weight loss and liver oxidative stress in rats. Creatine supplementation, however, prevented these tumor-associated perturbations.


Assuntos
Caquexia , Creatina/farmacologia , Hiper-Homocisteinemia , Neoplasias Experimentais , Estresse Oxidativo/efeitos dos fármacos , Animais , Caquexia/tratamento farmacológico , Caquexia/metabolismo , Caquexia/patologia , Creatina/farmacocinética , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Hiper-Homocisteinemia/prevenção & controle , Masculino , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Ratos , Ratos Wistar
9.
Nihon Arukoru Yakubutsu Igakkai Zasshi ; 51(5): 323-334, 2016 Oct.
Artigo em Inglês, Japonês | MEDLINE | ID: mdl-30462393

RESUMO

Chronic alcohol consumption is a major public health problem that frequently leads to the development of liver steatosis, fibrosis, and eventually cirrhosis and hepatocellular carcinoma. Hyperhomocysteinemia is a pathological consequence of alcoholic liver disease (ALD) and is attributed to hepatic endoplasmic reticulum (ER) stress and insulin resistance. However, the regulatory function of nuclear receptors in ALD associated with dysregulation of homocysteine metabolism remains largely unknown. Nuclear receptor small heterodimer partner (SHP, NROB2) is a pleiotropic transcriptional repressor involved in regulating various metabolic path-ways in the liver. This study investigated a critical role of SHP in alcohol-induced hyperhomocysteinemia. . The expression and enzymatic activities of betaine-homocysteine S-methyltransferase (BHMT) and cystathionine y -lyase (CTH) were significantly increased in the liver of SHP- knockout (SKO) mice as compared to the wild-type mice. The substrates of BHMT and CTH, such as betaine, choline and cystathionine, were decreased in SKO liver while their products including hydrogen sulfide and cysteine were increased. However, methionine and homocysteine were not altered by SHP- deficiency, suggesting that the methionine cycle is activated in SKO mice. Forkhead box A (FOXA)- binding site was identified in both the BHMT and CTH promoters. Luciferase assay demonstrated that FOXAI, but not FOXA2, activated both BHMT and CTH promoters through the FOXA-binding site. Overexpression of FOXA1 induced BHMT and CTH expression in Hepal-6 cells, which was inhibited by SHP coexpression. Consistently, alcohol-induced hyperhomocysteinemia, and homocysteine-induced hepatic ER stress and glucose intolerance were abrogated in SKO mice. These novel findings identified SHP and FOXA1 as important regulators of hepatic homocysteine metabolism. Because hyper-homocysteinemia is a risk factor for cardiovascular disease and insulin resistance, and is often associated with ALD and metabolic syndrome, SHP and FOXA1 could be used as potential targets for hyperhomocysteinemia and its related diseases. Taken together, these results shed light on the regulatory mechanism of homocysteine metabolism in the liver.


Assuntos
Etanol/toxicidade , Hiper-Homocisteinemia/prevenção & controle , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Etanol/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hiper-Homocisteinemia/induzido quimicamente , Camundongos , Multimerização Proteica , Receptores Citoplasmáticos e Nucleares/genética
10.
Toxicol Ind Health ; 32(9): 1607-18, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25731901

RESUMO

N-Acetylcysteine (NAC) could be included in protocols designed for the treatment of lead toxicity. Therefore, in this study, we decided to investigate the influence of NAC administration on homocysteine (Hcy) levels, oxidative damage to proteins, and the levels of iron (Fe), transferrin (TRF), and haptoglobin (HPG) in lead (Pb)-exposed workers. The examined population (n = 171) was composed of male employees who worked with Pb. They were randomized into four groups. Workers who were not administered any antioxidants, drugs, vitamins, or dietary supplements were classified as the reference group (n = 49). The remaining three groups consisted of workers who were treated orally with NAC at three different doses (1 × 200, 2 × 200, or 2 × 400 mg) for 12 weeks. After the treatment, blood Pb levels significantly decreased in the groups receiving NAC compared with the reference group. The protein concentration was not affected by NAC administration. In contrast, Hcy levels significantly decreased or showed a strong tendency toward lower values depending on the NAC dose. Levels of the protein carbonyl groups were significantly decreased in all of the groups receiving NAC. Conversely, glutamate dehydrogenase activity was significantly elevated in all of the groups receiving NAC, while the level of protein thiol groups was significantly elevated only in the group receiving 200 mg of NAC. Treatment with NAC did not significantly affect Fe and TRF levels, whereas HPG levels showed a tendency toward lower values. Treatment with NAC normalized the level of Hcy and decreased oxidative stress as measured by the protein carbonyl content; this effect occurred in a dose-dependent manner. Moreover, small doses of NAC elevated the levels of protein thiol groups. Therefore, NAC could be introduced as an alternative therapy for chronic Pb toxicity in humans.


Assuntos
Acetilcisteína/uso terapêutico , Anemia Ferropriva/prevenção & controle , Antioxidantes/uso terapêutico , Suplementos Nutricionais , Hiper-Homocisteinemia/prevenção & controle , Intoxicação por Chumbo/prevenção & controle , Doenças Profissionais/prevenção & controle , Acetilcisteína/administração & dosagem , Adulto , Poluentes Ocupacionais do Ar/toxicidade , Anemia Ferropriva/etiologia , Antioxidantes/administração & dosagem , Haptoglobinas/análise , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/etiologia , Exposição por Inalação/efeitos adversos , Ferro/sangue , Chumbo/sangue , Chumbo/toxicidade , Intoxicação por Chumbo/sangue , Intoxicação por Chumbo/fisiopatologia , Masculino , Pessoa de Meia-Idade , Doenças Profissionais/sangue , Doenças Profissionais/fisiopatologia , Exposição Ocupacional/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Polônia , Carbonilação Proteica , Protoporfirinas/sangue , Transferrina/análise
11.
Am J Clin Nutr ; 102(5): 1014-24, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26447155

RESUMO

BACKGROUND: Elevated total plasma homocysteine (tHcy) is considered to be an independent cardiovascular disease risk factor, although tHcy lowering by B-vitamins improves only certain clinical endpoints. N-acetylcysteine (NAC), a thiol-containing antioxidant, acutely lowers tHcy and possibly also blood pressure. However, to our knowledge, at present no conclusive long-term evaluation exists that controls for factors such as hyperlipidemia, smoking, medication, and disease stage, all of which affect the thiol redox state, including tHcy. OBJECTIVE: We reanalyzed 2 double-blind, placebo-controlled trials in unmedicated middle-aged men, one in a hyperlipidemic group (HYL group; n = 40) and one in a normolipidemic group (NOL group; n = 42), each stratified for smokers and nonsmokers. DESIGN: We evaluated the effect of 4 wk of oral NAC (1.8 g/d) on tHcy (primary endpoint), plasma thiol (cysteine), and intracellular glutathione concentrations as well as on blood pressure. The HYL group had total cholesterol >220 mg/dL or triglycerides >150 mg/dL. RESULTS: NAC treatment significantly (P = 0.001, multivariate analysis of variance for repeated measures) lowered postabsorptive plasma concentrations of tHcy by -11.7% ± 3.0% (placebo: 4.1% ± 3.6%) while increasing those of cysteine by 28.1% ± 5.7% (placebo: 4.0% ± 3.4%) with no significant impact of hyperlipidemia or smoking. Moreover, NAC significantly decreased systolic (P = 0.003) and diastolic (P = 0.017) blood pressure within all subjects with a significant reduction in diastolic pressure in the HYL group (P = 0.008) but not in the NOL group. An explorative stepwise multiple regression analysis identified 1) post-treatment cysteine as well as 2) pretreatment tHcy and 3) albumin plasma concentrations as being significant contributors to tHcy reduction. CONCLUSIONS: Four weeks of oral NAC treatment significantly decreased plasma tHcy concentrations, irrespective of lipid or smoking status, and lowered systolic blood pressure in both normolipidemic and hyperlipidemic men, with significant diastolic blood pressure reductions in the HYL group only. Increased oral intake of cysteine may therefore be considered for primary or secondary prevention of vascular events with regard to the 2 independent risk factors of hyperhomocysteinemia and arterial hypertension.


Assuntos
Acetilcisteína/uso terapêutico , Anti-Hipertensivos/uso terapêutico , Antioxidantes/uso terapêutico , Homocisteína/antagonistas & inibidores , Hiper-Homocisteinemia/prevenção & controle , Hipertensão/prevenção & controle , Acetilcisteína/administração & dosagem , Acetilcisteína/sangue , Acetilcisteína/farmacocinética , Administração Oral , Adulto , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/sangue , Anti-Hipertensivos/farmacocinética , Antioxidantes/administração & dosagem , Antioxidantes/análise , Antioxidantes/farmacocinética , Biotransformação , Colesterol/sangue , Cisteína/sangue , Método Duplo-Cego , Glutationa/sangue , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/metabolismo , Hiperlipidemias/complicações , Hipertensão/sangue , Hipertensão/complicações , Hipertensão/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Pessoa de Meia-Idade , Fumar/efeitos adversos , Triglicerídeos/sangue
12.
Clin Chim Acta ; 450: 316-21, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26343924

RESUMO

Homocysteine (Hcy) is a derived sulfur-containing and non-proteinogenic amino acid. The metabolism of Hcy occurs either through the remethylation to methionine or transsulfuration to cysteine. Studies have identified hyperhomocysteinemia (HHcy) as one of the possible risk factors for a multitude of diseases including vascular, neurodegenerative and ocular diseases. Association of HHcy with eye diseases such as retinopathy, pseudoexfoliative glaucoma maculopathy, cataract, optic atrophy and retinal vessel atherosclerosis is established. The molecular mechanism underlying these ocular diseases has been reported as impaired vascular endothelial function, apoptosis of retinal ganglion cells, extracellular matrix alterations, decreased lysyl oxidase activity and oxidative stress. The formed homocysteine-thiolactone in HHcy has stronger cytotoxicity and pro-inflammatory properties which can induce lens opacification and optic nerve damage. The metabolism of Hcy requires enzymes with vitamins such as folic acid, vitamins B12 and B6. Despite the mixed conclusion of various studies regarding the level of these vitamins in elder people, studies recommended the treatment with folate and B12 to reduce Hcy levels in subjects with or without any defect in the enzymes involved in its metabolism. The levels of Hcy, folate, B6 as well as B12 should be measured early in patients with visual impairment that would aid to screen patients for life-threatening disorders related with HHcy. Elder patients may supplement with these vitamins in order to attenuate the ocular damages. This article discusses the association of Hcy in ocular diseases and the possible mechanism in the pathogenesis.


Assuntos
Oftalmopatias , Homocisteína , Animais , Oftalmopatias/complicações , Oftalmopatias/metabolismo , Homocisteína/metabolismo , Humanos , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/prevenção & controle
13.
J Nutr ; 145(10): 2245-52, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26311810

RESUMO

BACKGROUND: Creatine synthesis from guanidinoacetate consumes ~50% of s-adenosylmethionine (SAM)-derived methyl groups, accounting for an equivalent proportion of s-adenosylhomocysteine (SAH) and total homocysteine (tHcys) synthesis. Dietary creatine inhibits the synthesis of guanidinoacetate, thereby lowering plasma tHcys in rats. OBJECTIVE: We tested the hypotheses that creatine supplementation lowers plasma guanidinoacetate, increases blood SAM, lowers blood SAH, and lowers plasma tHcys. METHODS: Bangladeshi adults were randomly assigned to receive 1 of 4 treatments for 12 wk: placebo (n = 101), 3 g/d creatine (Cr; n = 101), 400 µg/d folic acid (FA; n = 153), or 3 g/d creatine plus 400 µg/d folic acid (Cr+FA; n = 103). The outcomes of plasma guanidinoacetate and tHcys, as well as whole blood SAM and SAH, were analyzed at baseline and week 12 by HPLC. Treatment effects of creatine supplementation were examined with the use of the group comparisons of Cr vs. placebo and Cr+FA vs. FA. RESULTS: Plasma guanidinoacetate declined by 10.6% (95% CI: 4.9, 15.9) in the Cr group while increasing nonsignificantly in the placebo group (3.7%; 95% CI: -0.8, 8.5) (Pgroup difference = 0.0002). Similarly, plasma guanidinoacetate declined by 9.0% (95% CI: 3.4, 14.2) in the Cr+FA group while increasing in the FA group (7.0%; 95% CI: 2.0, 12.2) (Pgroup difference < 0.0001). Plasma tHcys declined by 23.4% (95% CI: 19.5, 27.1) and 21.0% (95% CI: 16.4, 25.2) in the FA and Cr+FA groups, respectively (Pgroup difference = 0.41), with no significant changes in the placebo or Cr groups (Pgroup difference = 0.35). A decrease in guanidinoacetate over time was associated with a decrease in tHcys over time in the Cr+FA group (ß = 0.30; 95% CI: 0.17, 0.43; P < 0.0001). CONCLUSIONS: Our findings indicate that whereas creatine supplementation downregulates endogenous creatine synthesis, this may not on average lower plasma tHcys in humans. However, tHcys did decrease in those participants who experienced a decline in plasma guanidinoacetate while receiving creatine plus folic acid supplementation. This trial was registered at clinicaltrials.gov as NCT01050556.


Assuntos
Creatina/uso terapêutico , Suplementos Nutricionais , Regulação para Baixo , Glicina/análogos & derivados , Homocisteína/sangue , Hiper-Homocisteinemia/prevenção & controle , Adulto , Bangladesh , Biomarcadores/sangue , Estudos de Coortes , Creatina/administração & dosagem , Creatina/efeitos adversos , Suplementos Nutricionais/efeitos adversos , Método Duplo-Cego , Feminino , Ácido Fólico/efeitos adversos , Ácido Fólico/uso terapêutico , Glicina/sangue , Humanos , Hiper-Homocisteinemia/sangue , Análise de Intenção de Tratamento , Masculino , Pessoa de Meia-Idade , Pacientes Desistentes do Tratamento , S-Adenosil-Homocisteína/sangue , S-Adenosilmetionina/sangue
14.
J Mol Cell Cardiol ; 81: 96-106, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25680906

RESUMO

Abdominal aortic aneurysm (AAA) is a serious vascular disease with high mortality. Our previous study suggested that hyperhomocysteinemia (HHcy) exaggerates the occurrence of AAA. Here, we investigated whether macrophage inflammasome is involved in HHcy-aggravated AAA formation. Two independent HHcy-aggravated AAA models, perivascular calcium phosphate-treated C57BL/6 mice and angiotensin II (Ang II)-infused apolipoprotein E-deficient (ApoE(-/-)) mice were used. NLPR3, caspase 1, and interleukin-1ß (IL-1ß) levels were higher in aneurysmal lesions of both HHcy models compared to controls, preferentially in macrophages. Similarly, macrophage inflammasome activation was observed in vitro. Folic acid administration reversed the HHcy-accelerated AAA, with ameliorated activation of inflammasome in the tunica adventitia. Lentiviral silencing of NLRP3 significantly ameliorated HHcy-aggravated AAA formation. We observed increased mitochondrial production of reactive oxygen species (ROS) and energy switch from oxidative phosphorylation to glycolysis with excess Hcy in macrophages. Blocking mitochondrial ROS production in macrophages abolished inflammasome activation. Our study highlights the potential importance of macrophage inflammasome in the pathogenesis and development of HHcy-aggravated AAA.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Proteínas de Transporte/metabolismo , Hiper-Homocisteinemia/metabolismo , Inflamassomos/metabolismo , Macrófagos/metabolismo , Túnica Adventícia/efeitos dos fármacos , Túnica Adventícia/metabolismo , Túnica Adventícia/patologia , Animais , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/complicações , Aneurisma da Aorta Abdominal/prevenção & controle , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Fosfatos de Cálcio/efeitos adversos , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Caspase 1/genética , Caspase 1/metabolismo , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Ácido Fólico/farmacologia , Expressão Gênica , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/prevenção & controle , Inflamassomos/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
15.
Gastroenterology ; 148(5): 1012-1023.e14, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25701738

RESUMO

BACKGROUND & AIMS: Hyperhomocysteinemia is often associated with liver and metabolic diseases. We studied nuclear receptors that mediate oscillatory control of homocysteine homeostasis in mice. METHODS: We studied mice with disruptions in Nr0b2 (called small heterodimer partner [SHP]-null mice), betaine-homocysteine S-methyltransferase (Bhmt), or both genes (BHMT-null/SHP-null mice), along with mice with wild-type copies of these genes (controls). Hyperhomocysteinemia was induced by feeding mice alcohol (National Institute on Alcohol Abuse and Alcoholism binge model) or chow diets along with water containing 0.18% DL-homocysteine. Some mice were placed on diets containing cholic acid (1%) or cholestyramine (2%) or high-fat diets (60%). Serum and livers were collected during a 24-hour light-dark cycle and analyzed by RNA-seq, metabolomic, and quantitative polymerase chain reaction, immunoblot, and chromatin immunoprecipitation assays. RESULTS: SHP-null mice had altered timing in expression of genes that regulate homocysteine metabolism compared with control mice. Oscillatory production of S-adenosylmethionine, betaine, choline, phosphocholine, glyceophosphocholine, cystathionine, cysteine, hydrogen sulfide, glutathione disulfide, and glutathione, differed between SHP-null mice and control mice. SHP inhibited transcriptional activation of Bhmt and cystathionine γ-lyase by FOXA1. Expression of Bhmt and cystathionine γ-lyase was decreased when mice were fed cholic acid but increased when they were placed on diets containing cholestyramine or high-fat content. Diets containing ethanol or homocysteine induced hyperhomocysteinemia and glucose intolerance in control, but not SHP-null, mice. In BHMT-null and BHMT-null/SHP-null mice fed a control liquid, lipid vacuoles were observed in livers. Ethanol feeding induced accumulation of macrovesicular lipid vacuoles to the greatest extent in BHMT-null and BHMT-null/SHP-null mice. CONCLUSIONS: Disruption of Shp in mice alters timing of expression of genes that regulate homocysteine metabolism and the liver responses to ethanol and homocysteine. SHP inhibits the transcriptional activation of Bhmt and cystathionine γ-lyase by FOXA1.


Assuntos
Ritmo Circadiano , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Homocisteína/metabolismo , Hiper-Homocisteinemia/metabolismo , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Betaína-Homocisteína S-Metiltransferase/genética , Betaína-Homocisteína S-Metiltransferase/metabolismo , Glicemia/metabolismo , Resina de Colestiramina , Ácido Cólico , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Etanol , Regulação Enzimológica da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/metabolismo , Homeostase , Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/prevenção & controle , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/deficiência , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Tempo , Ativação Transcricional
16.
Appl Physiol Nutr Metab ; 40(1): 46-50, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25485892

RESUMO

The purpose of this study was to examine the effects of whey protein supplementation on homocysteine (Hcy) metabolism and liver oxidative stress in rats. Twenty-four rats were divided into 3 groups (n = 8) to receive one of the following diets for 4 weeks: control diet (C), whey protein-composed diet (WP), and whey protein-supplemented diet (WPS). The C and WP diets consisted of AIN-93 with 20% casein and 20% whey protein as protein source, respectively. WPS was AIN-93 (20% casein) supplemented by the addition of 20% (w/w) whey protein. Four weeks of ingesting a WPS diet resulted in a significantly higher (P < 0.05) total protein and methionine intakes. Although a significant increase (P < 0.05) in the hepatic S-adenosylmethionine and S-adenosylhomocysteine levels occurred in WPS group compared with C and WP, no significant change was observed in plasma Hcy concentration between groups. Furthermore, the levels of lipid hydroperoxides and advanced oxidation protein products, known liver oxidative stress markers, were increased in the WPS group compared with the C group. In addition, no change in glutathione liver concentration was observed in any of the groups studied. In conclusion, whey protein supplementation increases methionine intake substantially; however, it does not change plasma Hcy concentrations. On the other hand, increased hepatic oxidative stress markers were observed in whey protein supplemented rats were probably due to high protein intake.


Assuntos
Suplementos Nutricionais/efeitos adversos , Hiper-Homocisteinemia/prevenção & controle , Fígado/metabolismo , Metionina/administração & dosagem , Estresse Oxidativo , Proteínas do Soro do Leite/efeitos adversos , Produtos da Oxidação Avançada de Proteínas/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Caseínas/efeitos adversos , Glutationa/metabolismo , Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Hiper-Homocisteinemia/etiologia , Hiper-Homocisteinemia/metabolismo , Peroxidação de Lipídeos , Masculino , Metionina/efeitos adversos , Metionina/sangue , Metionina/metabolismo , Músculo Esquelético/metabolismo , Oxirredução , Distribuição Aleatória , Ratos Wistar , S-Adenosil-Homocisteína/agonistas , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/agonistas , S-Adenosilmetionina/metabolismo , Proteínas do Soro do Leite/administração & dosagem
17.
ScientificWorldJournal ; 2014: 904501, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25250392

RESUMO

Betaine is an important natural component of rich food sources, especially spinach. Rats were fed diets with betaine or spinach powder at the same level of betaine for 10 days to investigate the dose-dependent effects of spinach powder supplementation on hyperhomocysteinemia induced by guanidinoacetic acid (GAA) addition and choline deprivation. The GAA-induced hyperhomocysteinemia in rats fed 25% casein diet (25 C) was significantly suppressed by supplementation with betaine or spinach, and it was completely suppressed by taking 11.0% spinach supplementation. The choline deprivation-induced enhancement of plasma homocysteine concentration in rats fed 25% soybean protein diet (25S) was markedly suppressed by 3.82% spinach. Supplementation with betaine or spinach partially prevented the effects of GAA on hepatic concentrations of methionine metabolites. The decrease in activity of betaine-homocysteine S-methyltransferase (BHMT) and cystathionine ß-synthase (CBS) in GAA-induced hyperhomocysteinemia was recovered by supplementation with betaine or spinach. Supplementation with betaine or spinach did not affect BHMT activity, whereas it partially restored CBS activity in choline-deprived 25S. The results indicated that betaine or spinach could completely suppress the hyperhomocysteinemia induced by choline deficiency resulting from stimulating the homocysteine removal by both remethylation and cystathionine formation.


Assuntos
Betaína/administração & dosagem , Deficiência de Colina/prevenção & controle , Suplementos Nutricionais , Glicina/análogos & derivados , Hiper-Homocisteinemia/prevenção & controle , Spinacia oleracea , Animais , Deficiência de Colina/complicações , Glicina/toxicidade , Hiper-Homocisteinemia/induzido quimicamente , Hiper-Homocisteinemia/etiologia , Masculino , Ratos , Ratos Wistar
18.
Przegl Lek ; 71(4): 189-92, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25141576

RESUMO

BACKGROUND: Hyperhomocysteinemia seems to be a common phenomenon in both patients with ulcerative colitis and Crohn's disease. Many factors including deficiencies of cobalamin, folate and pyridoxine, smoking habits, alcohol and coffee intake, some medications and age may predispose subjects to hyperhomocysteinemia. The study aimed to evaluate homocysteine levels in an inflammatory bowel disease cohort as dependent of life style and disease activity. METHODS: 85 consecutive patients with inflammatory bowel disease (38 with Crohn's disease and 47 with ulcerative colitis) and 65 control subjects were included in the prospective study. The following parameters were analyzed: disease activity, duration of the disease, location of pathological changes, presence of complications, current medications, past surgical procedures, smoking history, concomitant diseases, biochemical parameters and plasma homocysteine levels. RESULTS: Mild hyperhomocysteinemia was found in 16 patients with Crohn's disease (42%), 19 patients with ulcerative colitis (40%) and 19 patients in the control group (29%) (p = 0.59). There was not any significant correlation between homocysteine level and disease activity. Only folic acid supplementation and gender affected homocysteine level. Folic acid intake led to reduction of homocysteine levels in all groups of patients (11.8 micromol/l vs. 8.33 miccromol/l, p = 0.0065 in Crohn's disease patients and 10.94 micromol/l vs. 7.78 micromol/l, p = 0.0069 in ulcerative colitis patients). CONCLUSION: Homocysteine level in patients with inflammatory bowel disease is mostly normal or slightly elevated. Disease activity does not have an impact on homocysteine level. Folic acid is the most important factor having an influence on homocysteine level in patients with inflammatory bowel disease.


Assuntos
Ácido Fólico/uso terapêutico , Homocisteína/sangue , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/prevenção & controle , Doenças Inflamatórias Intestinais/sangue , Doenças Inflamatórias Intestinais/complicações , Adulto , Idoso , Colite Ulcerativa/sangue , Colite Ulcerativa/complicações , Doença de Crohn/complicações , Suplementos Nutricionais , Feminino , Homocisteína/efeitos dos fármacos , Humanos , Hiper-Homocisteinemia/sangue , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Adulto Jovem
19.
Int J Mol Sci ; 14(12): 24422-37, 2013 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-24351826

RESUMO

Maternal perinatal nutrition may program offspring metabolic features. Epigenetic regulation is one of the candidate mechanisms that may be affected by maternal dietary methyl donors intake as potential controllers of plasma homocysteine levels. Thirty-two Wistar pregnant rats were randomly assigned into four dietary groups during lactation: control, control supplemented with methyl donors, high-fat-sucrose and high-fat-sucrose supplemented with methyl donors. Physiological outcomes in the offspring were measured, including hepatic mRNA expression and global DNA methylation after weaning. The newborns whose mothers were fed the obesogenic diet were heavier longer and with a higher adiposity and intrahepatic fat content. Interestingly, increased levels of plasma homocysteine induced by the maternal high-fat-sucrose dietary intake were prevented in both sexes by maternal methyl donors supplementation. Total hepatic DNA methylation decreased in females due to maternal methyl donors administration, while Dnmt3a hepatic mRNA levels decreased accompanying the high-fat-sucrose consumption. Furthermore, a negative association between Dnmt3a liver mRNA levels and plasma homocysteine concentrations was found. Maternal high-fat-sucrose diet during lactation could program offspring obesity features, while methyl donors supplementation prevented the onset of high hyperhomocysteinemia. Maternal dietary intake also affected hepatic DNA methylation metabolism, which could be linked with the regulation of the methionine-homocysteine cycle.


Assuntos
Dieta Hiperlipídica , Sacarose Alimentar , Hiper-Homocisteinemia/prevenção & controle , Animais , Peso Corporal/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , DNA Metiltransferase 3A , Suplementos Nutricionais , Feminino , Homocisteína/sangue , Lactação , Masculino , Fenômenos Fisiológicos da Nutrição Materna/efeitos dos fármacos , Obesidade/prevenção & controle , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , S-Adenosilmetionina/farmacologia
20.
Nutr J ; 12: 136, 2013 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-24107225

RESUMO

BACKGROUND: Asymptomatic Indian lacto vegetarians, who make up more than half of the Indian population in different geographic regions, have distinctly low vitamin B-12 concentrations than non- vegetarians. Vegetarians consume milk but it seems that the amount is not enough to improve vitamin B-12 status or vitamin B-12 concentration in milk itself may be low. The aim of this study was to determine if daily milk consumption can improve vitamin B-12 status. METHODS: Fifteen male and 36 female, young healthy post-graduate volunteers participated. Blood from ten participants (4 males and 6 females) was collected (day-1). They continued their regular diet for next fourteen days and on day-15, blood of all 51 participants was collected, plasma vitamin B-12 concentration was measured and were divided into two groups; Normal (vitamin B-12 >148 pmol/L, n = 22) and Vitamin B-12 deficient (<148 pmol/L, n = 29), the remaining plasma was stored. All participants consumed 600 ml. of non-enriched buffalo milk (200 × 3) during the day along with their usual diet. Next day blood was collected for plasma holotranscobalamin II measurement. Subjects from deficient group continued to drink 400 ml of milk daily for next 14 days and blood was collected on day-30. Plasma holotranscoabalamin II (day-1, 15, 16, 30), vitamin B-12, folate, total homocysteine, creatinine and hematoloical parameters (day-1, 15, 30), and milk vitamin B-12 concentrations (day-15, 16, 30) were measured. RESULTS: Fifty seven per cent of the participants were vitamin B-12 deficient and 65% were hyperhomocysteinemic. No significant difference in biomarkers was observed when there was no intervention. Plasma holotranscobalamin II concentration increased from 19.6 to 22.27 pmol/L (p < 0.0001) 24 hrs after milk load in the whole group. Plasma vitamin B-12 increased from 92.5 to 122 pmol/L and tHcy concentrations decreased from 31.9 to 24.9 µ mol/L (p < 0.0001 for both) 14 days after regular milk intake in vitamin B-12 deficient subjects. CONCLUSIONS: Regular intake of milk improved vitamin B-12 status of vitamin B-12 deficient vegetarians indicating a potential dietary strategy to improve the vitamin status.


Assuntos
Dieta Vegetariana/efeitos adversos , Alimento Funcional , Leite , Estado Nutricional , Deficiência de Vitamina B 12/prevenção & controle , Vitamina B 12/administração & dosagem , Adulto , Animais , Búfalos , Dieta com Restrição de Gorduras/efeitos adversos , Dieta com Restrição de Gorduras/etnologia , Dieta Vegetariana/etnologia , Feminino , Alimento Funcional/análise , Promoção da Saúde , Homocisteína/sangue , Humanos , Hiper-Homocisteinemia/etnologia , Hiper-Homocisteinemia/etiologia , Hiper-Homocisteinemia/prevenção & controle , Índia , Masculino , Leite/química , Política Nutricional , Ciências da Nutrição/educação , Estado Nutricional/etnologia , Educação de Pacientes como Assunto , Transcobalaminas/análise , Vitamina B 12/análise , Vitamina B 12/sangue , Deficiência de Vitamina B 12/sangue , Deficiência de Vitamina B 12/etnologia , Deficiência de Vitamina B 12/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA