Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Nutrients ; 16(11)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38892484

RESUMO

Exploring the link between genetic polymorphisms in folate metabolism genes (MTHFR, MTR, and MTRR) and cardiovascular disease (CVD), this study evaluates the effect of B vitamin supplements (methylfolate, pyridoxal-5'-phosphate, and methylcobalamin) on homocysteine and lipid levels, potentially guiding personalized CVD risk management. In a randomized, double-blind, placebo-controlled trial, 54 patients aged 40-75 with elevated homocysteine and moderate LDL-C levels were divided based on MTHFR, MTR, and MTRR genetic polymorphisms. Over six months, they received either a combination of methylfolate, P5P, and methylcobalamin, or a placebo. At the 6 months follow-up, the treatment group demonstrated a significant reduction in homocysteine levels by 30.0% (95% CI: -39.7% to -20.3%) and LDL-C by 7.5% (95% CI: -10.3% to -4.7%), compared to the placebo (p < 0.01 for all). In the subgroup analysis, Homozygous Minor Allele Carriers showed a more significant reduction in homocysteine levels (48.3%, 95% CI: -62.3% to -34.3%, p < 0.01) compared to mixed allele carriers (18.6%, 95% CI: -25.6% to -11.6%, p < 0.01), with a notable intergroup difference (29.7%, 95% CI: -50.7% to -8.7%, p < 0.01). LDL-C levels decreased by 11.8% in homozygous carriers (95% CI: -15.8% to -7.8%, p < 0.01) and 4.8% in mixed allele carriers (95% CI: -6.8% to -2.8%, p < 0.01), with a significant between-group difference (7.0%, 95% CI: -13.0% to -1.0%, p < 0.01). Methylfolate, P5P, and methylcobalamin supplementation tailored to genetic profiles effectively reduced homocysteine and LDL-C levels in patients with specific MTHFR, MTR, and MTRR polymorphisms, particularly with homozygous minor allele polymorphisms.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase , LDL-Colesterol , Suplementos Nutricionais , Ferredoxina-NADP Redutase , Homocisteína , Metilenotetra-Hidrofolato Redutase (NADPH2) , Fosfato de Piridoxal , Tetra-Hidrofolatos , Vitamina B 12 , Humanos , Pessoa de Meia-Idade , Homocisteína/sangue , Feminino , Masculino , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Método Duplo-Cego , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , LDL-Colesterol/sangue , Idoso , Vitamina B 12/administração & dosagem , Vitamina B 12/análogos & derivados , Adulto , Ferredoxina-NADP Redutase/genética , Tetra-Hidrofolatos/administração & dosagem , Polimorfismo Genético , Complexo Vitamínico B/uso terapêutico , Complexo Vitamínico B/administração & dosagem , Complexo Vitamínico B/farmacologia
2.
Nutrients ; 16(13)2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38999770

RESUMO

Emerging evidence suggests that personalized dietary supplement regimens can significantly influence lipid metabolism and cardiovascular risk. The efficacy of AI-guided dietary supplement prescriptions, compared with standard physician-guided prescriptions, remains underexplored. In a randomized, parallel-group pilot study, 70 patients aged 40-75 years with LDL-C levels between 70 and 190 mg/dL were enrolled. Participants were randomized to receive either AI-guided dietary supplement prescriptions or standard physician-guided prescriptions for 90 days. The primary endpoint was the percent change in LDL-C levels. Secondary endpoints included changes in total cholesterol, HDL-C, triglycerides, and hsCRP. Supplement adherence and side effects were monitored. Sixty-seven participants completed the study. The AI-guided group experienced a 25.3% reduction in LDL-C levels (95% CI: -28.7% to -21.9%), significantly greater than the 15.2% reduction in the physician-guided group (95% CI: -18.5% to -11.9%; p < 0.01). Total cholesterol decreased by 15.4% (95% CI: -19.1% to -11.7%) in the AI-guided group compared with 8.1% (95% CI: -11.5% to -4.7%) in the physician-guided group (p < 0.05). Triglycerides were reduced by 22.1% (95% CI: -27.2% to -17.0%) in the AI-guided group versus 12.3% (95% CI: -16.7% to -7.9%) in the physician-guided group (p < 0.01). HDL-C and hsCRP changes were not significantly different between groups. The AI-guided group received a broader variety of supplements, including plant sterols, omega-3 fatty acids, red yeast rice, coenzyme Q10, niacin, and fiber supplements. Side effects were minimal and comparable between groups. AI-guided dietary supplement prescriptions significantly reduce LDL-C and triglycerides more effectively than standard physician-guided prescriptions, highlighting the potential for AI-driven personalization in managing hypercholesterolemia.


Assuntos
LDL-Colesterol , Suplementos Nutricionais , Humanos , Pessoa de Meia-Idade , Projetos Piloto , Masculino , Feminino , Idoso , LDL-Colesterol/sangue , Adulto , Hipercolesterolemia/sangue , Hipercolesterolemia/tratamento farmacológico , Triglicerídeos/sangue , Resultado do Tratamento , HDL-Colesterol/sangue
3.
Nutrients ; 16(1)2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38201926

RESUMO

Emerging evidence suggests that PPARG gene polymorphisms may influence lipid metabolism and cardiovascular risk, with omega-3 fatty acids proposed to modulate these effects. This study aims to assess the effects of fish oil supplementation on cardiovascular markers among adults with PPARG gene polymorphisms in a randomized, double-blind, placebo-controlled trial. A cohort of 102 patients with LDL-C 70-190 mg/dL was randomized to receive either 2000 mg of omega-3 fatty acids or a placebo daily for 90 days. In the omega-3 group with PPARG polymorphisms, LDL-C was reduced by 15.4% (95% CI: -19.8% to -11.0%), compared with a 2.6% decrease in the placebo group (95% CI: -4.1% to -1.1%; p < 0.01). In the omega-3 group without PPARG polymorphisms, LDL-C was reduced by 3.7% (95% CI: -6.9% to -0.6%), not significantly different from the placebo group's reduction of 2.9% (95% CI: -5.1% to -0.8%; p = 0.28). The reduction in LDL-C was notably 11.7% greater in those with PPARG polymorphisms than in those without (95% CI: -19.3% to -4.0%; p < 0.01). Triglycerides decreased by 21.3% in omega-3 recipients with PPARG polymorphisms (95% CI: -26.5% to -16.2%; p < 0.01), with no significant changes in HDL-C, total cholesterol, or hsCRP levels in any groups. Minor allele frequencies and baseline characteristics were comparable, ensuring a balanced genetic representation. Omega-3 fatty acids significantly reduce LDL-C and triglycerides in carriers of PPARG polymorphisms, underlining the potential for genetic-driven personalization of cardiovascular interventions.


Assuntos
Ácidos Graxos Ômega-3 , Adulto , Humanos , Ácidos Graxos Ômega-3/farmacologia , PPAR gama/genética , LDL-Colesterol , Polimorfismo Genético , Triglicerídeos , Suplementos Nutricionais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA