Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.400
Filtrer
1.
Mol Biol Rep ; 51(1): 883, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39088020

RÉSUMÉ

BACKGROUND: Cerebral venous sinus thrombosis (CVST) is a rare cause of stroke. Acquired and inherited prothrombotic conditions are the most common risk factors for CVST. Sometimes, an etiology is not found. Wide utilization of next generation sequencing technologies in clinical practice may lead to identification of risk factors other than those classically associated with CVST. METHOD AND RESULTS: This retrospective clinical-laboratory observational study has a reference patient who presented with CVST as an adolescent. Work up for prothrombotic conditions showed high homocysteine level secondary to homozygosity for a common polymorphism, c.677 C > T in the methylenetetrahydrofolate reductase (MTHFR) gene. His older unaffected brother has a similar MTHFR genotype and high homocysteine. The whole exome sequencing revealed a likely pathogenic variant in the sodium voltage gated channel, alpha subunit 1(SCN1A) gene. CONCLUSION: CVST is a multifactorial disease. Prothrombotic conditions are the most common risk factors for CVST. High homocysteine due to the common MTHFR polymorphisms was previously attributed to various thrombotic conditions including CVST. Although high homocysteine due to MTHFR polymorphism may be a contributing factor, additional risk factors such as blood flow abnormalities during SCN1A related seizures may be needed for thrombosis.


Sujet(s)
Methylenetetrahydrofolate reductase (NADPH2) , Canal sodique voltage-dépendant NAV1.1 , Thromboses des sinus intracrâniens , Humains , Thromboses des sinus intracrâniens/génétique , Mâle , Canal sodique voltage-dépendant NAV1.1/génétique , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Adolescent , Études rétrospectives , Prédisposition génétique à une maladie , Facteurs de risque , Homocystéine/sang , /méthodes , Polymorphisme de nucléotide simple/génétique
2.
BMC Womens Health ; 24(1): 407, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026333

RÉSUMÉ

BACKGROUND: Methylenetetrahydrofolate reductase (MTHFR) is essential for the metabolism of folic acid and homocysteine. The MTHFR C677T polymorphism is associated with several disorders. Our study aims to explore the geographical distributions of the MTHFR C677T polymorphism of women in China and how migration affected the polymorphism in Suzhou. METHODS: A total of 7188 women of reproductive age were recruited in Suzhou of the study. Subjects were classified according to their native places after data extraction. MTHFR C677T gene polymorphisms were detected by quantitative PCR with genomic DNA isolated from blood samples. RESULTS: The frequencies of the 677T allele and 677TT genotype were higher in northern China than that in southern China and decreased in geographical gradients from north to south. The frequencies were considerably higher in the migrant population than that in the indigenous population of Suzhou. The migrant population have gradually changed the prevalence in Suzhou. CONCLUSIONS: Our study suggested that the prevalence of MTHFR C677T polymorphisms among women varied across different geographical regions in Chinese Han populations. The 677T allele frequencies of the northern populations were significantly higher than those of the southern populations. The migrant population gradually changed the prevalence of the MTHFR C677T polymorphism in Suzhou.


Sujet(s)
Fréquence d'allèle , Methylenetetrahydrofolate reductase (NADPH2) , Adulte , Femelle , Humains , Adulte d'âge moyen , Jeune adulte , Allèles , Chine/épidémiologie , Peuples d'Asie de l'Est/génétique , Génotype , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Polymorphisme de nucléotide simple
3.
Nutrients ; 16(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38999753

RÉSUMÉ

This study aimed to explore the association of maternal diet, infant MTHFR gene polymorphisms, and their interactions with the risk of ventricular septal defects (VSDs). This case-control study recruited 448 mothers of VSD children and 620 mothers of healthy counterparts. Multivariable-adjusted logistic regression models were constructed to examine the association between maternal dietary habits during the first trimester of gestation, MTHFR gene polymorphisms, and VSD. Gene-environment interaction effects were analyzed through logistic regression models, with false discovery rate p-value (FDR_p) < 0.05. Maternal excessive intake of fermented bean curd (OR = 2.00, 95%CI: 1.59-2.52), corned foods (OR = 2.23, 1.76-2.84), fumatory foods (OR = 1.75, 1.37-2.23), grilled foods (OR = 1.34, 1.04-1.72), and fried foods (OR = 1.80, 1.42-2.27) was associated with an increased risk of VSD. Regular intake of fish and shrimp (OR = 0.42, 0.33-0.53), fresh eggs (OR = 0.58, 0.44-0.75), soy products (OR = 0.69, 0.56-0.85), and dairy products (OR = 0.71, 0.59-0.85) was found to reduce the occurrence of VSD. Moreover, MTHFR gene polymorphisms at rs2066470 (homozygous: OR = 4.28, 1.68-10.90), rs1801133 (homozygous: OR = 2.28, 1.39-3.74), and rs1801131 (heterozygous: OR = 1.75, 1.24-2.47; homozygous: OR = 3.45, 1.50-7.95) elevated offspring susceptibility to VSDs. Furthermore, significant interactions of MTHFR polymorphisms with maternal dietary habits were observed, encompassing corned foods, fermented bean curd, fried foods, and grilled foods. Maternal dietary habits; MTHFR polymorphisms at rs2066470, rs1801131, and rs1801133; and their interactions were significantly associated with the occurrence of VSDs in offspring.


Sujet(s)
Régime alimentaire , Comportement alimentaire , Communications interventriculaires , Phénomènes physiologiques nutritionnels maternels , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Femelle , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Études cas-témoins , Grossesse , Communications interventriculaires/génétique , Communications interventriculaires/épidémiologie , Adulte , Mâle , Interaction entre gènes et environnement , Polymorphisme de nucléotide simple , Nourrisson , Prédisposition génétique à une maladie , Facteurs de risque , Nouveau-né
4.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39000032

RÉSUMÉ

Methylation is a biochemical process involving the addition of a methyl group (-CH3) to various chemical compounds. It plays a crucial role in maintaining the homeostasis of the endothelium, which lines the interior surface of blood vessels, and has been linked, among other conditions, to coronary artery disease (CAD). Despite significant progress in CAD diagnosis and treatment, intensive research continues into genotypic and phenotypic CAD biomarkers. This review explores the significance of the methylation pathway and folate metabolism in CAD pathogenesis, with a focus on endothelial dysfunction resulting from deficiency in the active form of folate (5-MTHF). We discuss emerging areas of research into CAD biomarkers and factors influencing the methylation process. By highlighting genetically determined methylation disorders, particularly the MTHFR polymorphism, we propose the potential use of the active form of folate (5-MTHF) as a novel CAD biomarker and personalized pharmaceutical for selected patient groups. Our aim is to improve the identification of individuals at high risk of CAD and enhance their prognosis.


Sujet(s)
Maladie des artères coronaires , Acide folique , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Maladie des artères coronaires/métabolisme , Maladie des artères coronaires/génétique , Maladie des artères coronaires/étiologie , Acide folique/métabolisme , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Methylenetetrahydrofolate reductase (NADPH2)/métabolisme , Méthylation de l'ADN , Marqueurs biologiques , Méthylation , Animaux , Polymorphisme génétique
5.
Ann Afr Med ; 23(2): 234-236, 2024 Apr 01.
Article de Français, Anglais | MEDLINE | ID: mdl-39028172

RÉSUMÉ

Methylenetetrahydrofolate reductase (MTHFR) enzyme is one of the key enzymes involved in the metabolism of folate. Mutations in this enzyme can lead to a procoagulant state. We present a case of a 20-year-old male with no known comorbidities, who presented with fever and hemoptysis and was diagnosed as a case of pulmonary embolism. He was found to have a homozygous mutation in the MTHFR gene that was responsible for his disease state. He was started on unfractionated heparin infusion and underwent catheter-directed thrombolysis. He showed marked improvement in his condition and was discharged on oral anticoagulants with an advice to follow-up.


RésuméL'enzyme méthylènetétrahydrofolate réductase (MTHFR) est l'une des enzymes clés impliquées dans le métabolisme du folate. Les mutations de cette enzyme peuvent conduire à un état procoagulant. Nous présentons le cas d'un homme de 20 ans sans comorbidités connues, qui s'est présenté avec de la fièvre et une hémoptysie et a été diagnostiqué comme un cas d'embolie pulmonaire. Il s'est avéré qu'il présentait une mutation homozygote du gène MTHFR responsable de son état pathologique. Il a commencé une perfusion d'héparine non fractionnée et a subi une thrombolyse dirigée par cathéter. Il a montré une nette amélioration de son état et a été libéré sous anticoagulants oraux avec un conseil de suivi.


Sujet(s)
Anticoagulants , Methylenetetrahydrofolate reductase (NADPH2) , Mutation , Embolie pulmonaire , Humains , Mâle , Embolie pulmonaire/traitement médicamenteux , Embolie pulmonaire/génétique , Embolie pulmonaire/diagnostic , Embolie pulmonaire/imagerie diagnostique , Anticoagulants/usage thérapeutique , Jeune adulte , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Résultat thérapeutique , Héparine/usage thérapeutique , Traitement thrombolytique/méthodes , Homozygote
6.
Medicine (Baltimore) ; 103(28): e38648, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38996094

RÉSUMÉ

Gestational diabetes mellitus (GDM) is a common condition during pregnancy and is associated with an increased risk of pre-eclampsia. The methylenetetrahydrofolate reductase (MTHFR) gene plays a crucial role in folate metabolism and has been implicated in GDM. To investigate the relationship between the MTHFR C677T gene polymorphism and the conditions of GDM and gestational prediabetes in pregnant women. A case-control study was conducted in 114 pregnant women with GDM and 96 pregnant women without GDM, from the first trimester to the prenatal examination at Can Tho Obstetrics Hospital. The pregnant women underwent a 1-hour (G1) and 2-hour (G2) oral glucose tolerance test (OGTT) and genetic polymorphism analysis based on real-time PCR technique. In pregnant women with GDM, weight, concentrations of G0, G1, G2, and folic acid were higher than those in the non-GDM group, with P < .05. When analyzing the subgroup without gestational diabetes, we found that the rate of prediabetes was 16.6% (16/96 pregnant women). In this group, blood glucose levels at 1 hour and 2 hours during the OGTT were higher compared to the normal glucose group (P < .05). A 2-hour post-OGTT glucose level of 7.78 mmol/L had a sensitivity of 93.8%, a specificity of 100%, and an area under the curve of 0.987 for diagnosing gestational prediabetes (P < .001). However, there were no statistically significant differences in the CC, CT, and TT polymorphisms of the MTHFR C677T gene among pregnant women with or without pre-gestational and GDM. Both fasting blood glucose and 2-hour glucose concentrations during the OGTT, as well as folic acid concentrations, were higher in both the pre-gestational and GDM groups compared to the non-gestational diabetes cohort. However, the analysis of MTHFR C677T polymorphisms revealed no statistically significant differences among the groups, highlighting the necessity for more extensive investigations to gain deeper insights into this relationship.


Sujet(s)
Diabète gestationnel , Hyperglycémie provoquée , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Femelle , Grossesse , Diabète gestationnel/génétique , Diabète gestationnel/épidémiologie , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Adulte , Études cas-témoins , Glycémie/analyse , Glycémie/métabolisme , État prédiabétique/génétique , État prédiabétique/épidémiologie , Polymorphisme génétique , Polymorphisme de nucléotide simple
7.
Sci Rep ; 14(1): 15119, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956106

RÉSUMÉ

Analyze the relationship between genetic variations in the MTHFR gene at SNPs (rs1801131 and rs1801133) and the therapy outcomes for Iraqi patients with rheumatoid arthritis (RA). The study was conducted on a cohort of 95 RA Iraqi patients. Based on their treatment response, the cohort was divided into two groups: the responder (47 patients) and the nonresponder (48 patients), identified after at least three months of methotrexate (MTX) treatment. A polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique was employed to analyze the MTHFR variations, specifically at rs1801133 and rs1801131. Overall, rs1801131 followed both codominant and dominate models, in which in the codominant model, GG [OR (95% CI) 0.11 (0.022-0.553)] and TG [OR (95% CI) 0.106 (0.021-0.528)] predict responders compared to the TT genotype; meanwhile, for the dominate model, the presence of both GG and TG genotypes [OR (95% CI) 0.108 (0.023-0.507)] together predict responders compared to the TT genotype. The Ars1801133Grs1801131 haplotype was significantly associated with responders [OR (95% CI): 0.388 (0.208-0.723)], while the Grs1801133Trs1801131 haplotype was associated marginally with nonresponders [OR (95% CI) 1.980 (0.965-4.064)]. In the final multivariate analysis, GG/TGrs1801131 genotypes were independently related to responders after adjustment for patients, disease, and treatment characteristics, while TTrs1801131 genotypes were associated with nonresponders. The Iraqi RA patients showed genetic polymorphism in MTHFR gene rs1801131 with T carrier allele associated with nonresponders to MTX therapy. The rs1801131 followed both codominant and dominant models. The G-carried allele for rs1801131 showed an independent association with responder to MTX therapy after adjustment for patients, disease, and treatment characteristics.


Sujet(s)
Polyarthrite rhumatoïde , Méthotrexate , Methylenetetrahydrofolate reductase (NADPH2) , Polymorphisme de nucléotide simple , Humains , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/génétique , Méthotrexate/usage thérapeutique , Mâle , Femelle , Iraq , Adulte d'âge moyen , Adulte , Résultat thérapeutique , Antirhumatismaux/usage thérapeutique , Génotype
8.
Genes (Basel) ; 15(7)2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-39062651

RÉSUMÉ

Congenital heart disease is one of the most common congenital malformations and thus represents a considerable public health burden. Hence, the identification of individuals and families with an increased genetic predisposition to congenital heart disease (CHD) and its possible prevention is important. Even though CHD is associated with the lack of folate during early pregnancy, the genetic background of folate and methionine metabolism perturbations and their influence on CHD risk is not clear. While some genes, such as those coding for cytosolic enzymes of folate/methionine cycles, have been extensively studied, genetic studies of folate transporters (de)glutamation enzymes and mitochondrial enzymes of the folate cycle are lacking. Among genes coding for cytoplasmic enzymes of the folate cycle, MTHFR, MTHFD1, MTR, and MTRR have the strongest association with CHD, while among genes for enzymes of the methionine cycle BHMT and BHMT2 are the most prominent. Among mitochondrial folate cycle enzymes, MTHFD2 plays the most important role in CHD formation, while FPGS was identified as important in the group of (de)glutamation enzymes. Among transporters, the strongest association with CHD was demonstrated for SLC19A1.


Sujet(s)
Acide folique , Cardiopathies congénitales , Méthionine , Methylenetetrahydrofolate Dehydrogenase (NADP) , Humains , Acide folique/métabolisme , Cardiopathies congénitales/génétique , Méthionine/métabolisme , Méthionine/génétique , Methylenetetrahydrofolate Dehydrogenase (NADP)/génétique , Methylenetetrahydrofolate Dehydrogenase (NADP)/métabolisme , Antigènes mineurs d'histocompatibilité/génétique , Antigènes mineurs d'histocompatibilité/métabolisme , Ferredoxine-NADP reductase/génétique , Ferredoxine-NADP reductase/métabolisme , 5-Methyltetrahydrofolate-homocysteine s-methyltransferase/génétique , 5-Methyltetrahydrofolate-homocysteine s-methyltransferase/métabolisme , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Methylenetetrahydrofolate reductase (NADPH2)/métabolisme , Prédisposition génétique à une maladie , Betaine-homocysteine S-methyltransferase/génétique , Betaine-homocysteine S-methyltransferase/métabolisme , Aminohydrolases , Enzymes multifonctionnelles
9.
BMC Cardiovasc Disord ; 24(1): 345, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38977943

RÉSUMÉ

BACKGROUND AND AIMS: H-type hypertension is essential hypertension combined with high homocysteine, and both synergistically increase the risk of cardiovascular and cerebrovascular events. The aim of this study was to investigate the risk factors of H-type hypertension in Tibetan plateau population and correlation with MTHFR C677T gene. METHODS AND RESULTS: A multi-stage cluster random sampling method was used to select the research subjects in Tibet Autonomous Region from June 2020 to November 2021. Among Tibetans, the incidence of H-type hypertension accounted for 84.31% of hypertensive patients. The logistic regression analysis demonstrated that age, uric acid (UA), triglyceride (TG) and low-density lipoprotein cholesterol (LDL-C) were risk factors for the prevalence of H-type hypertension, the OR (95% CI) was 1.083(1.073-1.094), 1.002(1.001-1.004), 1.240(1.050-1.464) and 2.274(1.432-3.611), respectively. MTHFR C677T TT genotype patients with H-type hypertension OR (95% CI) was 1.629(1.004-2.643). Based on this, a nomogram model was established, and the reliability of the model was proved by area under ROC curve, Brier score and average absolute error. The model's results indicate that for every five years of age, the score increases by 6 points; for a 2mmol/L increase in TG, the score increases by 5.5 points; for a 1mmol/L increase in LDL-C, the score increases by 10 points; and individuals with the TT genotype receive 8 points. The higher the score, the greater the risk of disease. CONCLUSION: The MTHFR C677T TT genotype is a risk locus for Tibetan patients with H-type hypertension, with age, TG, and LDL-C were identified as risk factors for the disease.


Sujet(s)
Prédisposition génétique à une maladie , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Tibet/épidémiologie , Femelle , Mâle , Adulte d'âge moyen , Facteurs de risque , Appréciation des risques , Adulte , Prévalence , Phénotype , Hypertension essentielle/génétique , Hypertension essentielle/diagnostic , Hypertension essentielle/épidémiologie , Hypertension essentielle/physiopathologie , Pression sanguine/génétique , Sujet âgé , Incidence , Polymorphisme de nucléotide simple , Homocystéine/sang , Hyperhomocystéinémie/génétique , Hyperhomocystéinémie/diagnostic , Hyperhomocystéinémie/épidémiologie , Hyperhomocystéinémie/sang , Hypertension artérielle/génétique , Hypertension artérielle/diagnostic , Hypertension artérielle/épidémiologie , Hypertension artérielle/physiopathologie
11.
J Cell Mol Med ; 28(12): e18474, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38896027

RÉSUMÉ

Our previous study reckons that the impact of the rs1801133 variant of 5,10-methylenetetrahydrofolate reductase (MTHFR) on coronary artery disease (CAD) is possibly mediated by cardiometabolic disorder. This study is performed to verify this hypothesis. Four hundred and thirty CAD patients and 216 CAD-free individuals were enrolled in this case-control study. The rs1801133 variant was genotyped by PCR-RFLP. Severity of coronary lesions was evaluated by number of stenotic coronary vessels and extent of coronary stenosis. The rs1801133 T allele significantly increased homocysteine levels in patients with CAD and CAD-free individuals. Individuals with the T allele of rs1801133 had an increased risk of developing CAD. In contrast, individuals with the TT genotype of rs1801133 were at high risk of multiple vessel lesions. The carriers of CT genotype had higher levels of systolic blood pressure (SBP), low-density lipoprotein cholesterol (LDL-C), and high-sensitivity C-reactive protein (hs-CRP), and lower levels of apolipoprotein A1 (APOA1) than those with CC genotype in male patients with CAD. The receiver operating characteristic (ROC) curve and precision-recall (PR) curve indicated that hyperhomocysteinemia was sensitive to predict the severity of CAD. Multivariate logistic regression revealed that homocysteine, rs1801133, age, smoking, weight, body mass index (BMI), lipoprotein(a) [Lp(a)], and hs-CRP were independent risk factors for CAD. The increased risk of CAD and severity of coronary lesions associated with rs1801133 in the Chinese Han population were attributed, at least partly, to high homocysteine levels. Hyperhomocysteinemia had a high predictive value for severe CAD or multiple vessel lesions.


Sujet(s)
Maladie des artères coronaires , Homocystéine , Methylenetetrahydrofolate reductase (NADPH2) , Polymorphisme de nucléotide simple , Humains , Homocystéine/sang , Mâle , Maladie des artères coronaires/génétique , Maladie des artères coronaires/sang , Maladie des artères coronaires/anatomopathologie , Adulte d'âge moyen , Femelle , Études cas-témoins , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Indice de gravité de la maladie , Sujet âgé , Facteurs de risque , Prédisposition génétique à une maladie , Courbe ROC , Génotype , Protéine C-réactive/métabolisme , Protéine C-réactive/génétique , Allèles , Apolipoprotéine A-I/génétique , Apolipoprotéine A-I/sang
12.
Birth Defects Res ; 116(6): e2372, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38877667

RÉSUMÉ

OBJECTIVE: To determine the effect of maternal status in (plasma and red blood cell) folate, vitamin B12, homocysteine, and vitamin D, as well as their interaction with MTHFR (C677T and A1298C) and MTRR A66G polymorphisms, on maternal plasma docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (ARA) levels and the risk of neural tube defects (NTDs). METHODS: ARA, EPA, and DHA composition was assessed using capillary gas chromatography. RESULTS: ARA and DHA levels were higher in controls than in case mothers for low plasma folate status. For low red blood cell folate status, DHA levels were higher in controls than in case mothers. For high homocysteine levels, ARA and DHA levels were higher in controls than in case mothers. NTD mothers had lower EPA and DHA levels for low vitamin B12 levels. NTD mothers had lower DHA levels for low vitamin D levels. For low plasma folate status, DHA levels in the MTHFR C677T gene and ARA and EPA levels in MTHFR A1298C gene were different among the three genotypes in case mothers. DHA levels in the MTHFR C677T gene were different among the three genotypes in case mothers for both low and high homocysteine levels. For low vitamin B12 levels, ARA and DHA levels were different among the three genotypes of the MTHFR C677T gene in case mothers. In the MTHFR C677T gene, ARA and DHA levels were different among the three genotypes in case mothers for low vitamin D levels. CONCLUSIONS: More advanced research is required to verify a suitable biochemical parameter status in relation to the genotypes in pregnant women.


Sujet(s)
Acide arachidonique , Acide docosahexaénoïque , Acide eicosapentanoïque , Acide folique , Methylenetetrahydrofolate reductase (NADPH2) , Anomalies du tube neural , Humains , Acide eicosapentanoïque/sang , Acide docosahexaénoïque/sang , Femelle , Anomalies du tube neural/génétique , Acide arachidonique/sang , Acide arachidonique/métabolisme , Acide folique/sang , Adulte , Tunisie , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Homocystéine/sang , Homocystéine/génétique , Grossesse , Vitamine B12/sang , Études cas-témoins , Génotype , Vitamine D/sang , Vitamine D/génétique
13.
Epidemiology ; 35(4): 527-541, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38912713

RÉSUMÉ

BACKGROUND: Maternal folic acid intake has been associated with decreased risk for neurodevelopmental disorders including autism spectrum disorder (ASD). Genetic differences in folate metabolism could explain some inconsistencies. To our knowledge, newborn folate concentrations remain unexamined. METHODS: We measured folate in archived newborn dried blood spots of children from the CHARGE (Childhood Autism Risks from Genetics and the Environment) case-control study who were clinically confirmed at 24-60 months to have ASD (n = 380), developmental delay (n = 128), or typical development (n = 247). We quantified monthly folic acid intake from maternally-reported supplements and cereals consumed during pregnancy and 3 months prior. We assessed associations of newborn folate with maternal folic acid intake and with ASD or developmental delay using regression. We stratified estimates across maternal and child MTHFR genotypes. RESULTS: Among typically developing children, maternal folic acid intake in prepregnancy and each pregnancy month and prepregnancy prenatal vitamin intake were positively associated with newborn folate. Among children with ASD, prenatal vitamin intake in pregnancy months 2-9 was positively associated with newborn folate. Among children with developmental delay, maternal folic acid and prenatal vitamins during the first pregnancy month were positively associated with neonatal folate. Associations differed by MTHFR genotype. Overall, neonatal folate was not associated with ASD or developmental delay, though we observed associations with ASD in children with the MTHFR 677 TT genotype (odds ratio: 1.76, 95% CI = 1.19, 2.62; P for interaction = 0.08). CONCLUSION: Maternal prenatal folic acid intake was associated with neonatal folate at different times across neurodevelopmental groups. Neonatal folate was not associated with reduced ASD risk. MTHFR genotypes modulated these relationships.


Sujet(s)
Trouble du spectre autistique , Incapacités de développement , Acide folique , Methylenetetrahydrofolate reductase (NADPH2) , Autorapport , Humains , Acide folique/sang , Trouble du spectre autistique/épidémiologie , Trouble du spectre autistique/sang , Femelle , Études cas-témoins , Nouveau-né , Mâle , Grossesse , Incapacités de développement/épidémiologie , Incapacités de développement/sang , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Enfant d'âge préscolaire , Dépistage sur goutte de sang séché , Adulte , Compléments alimentaires , Génotype
14.
Nat Commun ; 15(1): 5167, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886362

RÉSUMÉ

Methylenetetrahydrofolate reductase (MTHFR) is a pivotal flavoprotein connecting the folate and methionine methyl cycles, catalyzing the conversion of methylenetetrahydrofolate to methyltetrahydrofolate. Human MTHFR (hMTHFR) undergoes elaborate allosteric regulation involving protein phosphorylation and S-adenosylmethionine (AdoMet)-dependent inhibition, though other factors such as subunit orientation and FAD status remain understudied due to the lack of a functional structural model. Here, we report crystal structures of Chaetomium thermophilum MTHFR (cMTHFR) in both active (R) and inhibited (T) states. We reveal FAD occlusion by Tyr361 in the T-state, which prevents substrate interaction. Remarkably, the inhibited form of cMTHFR accommodates two AdoMet molecules per subunit. In addition, we conducted a detailed investigation of the phosphorylation sites in hMTHFR, three of which were previously unidentified. Based on the structural framework provided by our cMTHFR model, we propose a possible mechanism to explain the allosteric structural transition of MTHFR, including the impact of phosphorylation on AdoMet-dependent inhibition.


Sujet(s)
Chaetomium , Methylenetetrahydrofolate reductase (NADPH2) , Adémétionine , Methylenetetrahydrofolate reductase (NADPH2)/métabolisme , Methylenetetrahydrofolate reductase (NADPH2)/composition chimique , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Adémétionine/métabolisme , Adémétionine/composition chimique , Régulation allostérique , Chaetomium/enzymologie , Chaetomium/métabolisme , Chaetomium/génétique , Phosphorylation , Humains , Cristallographie aux rayons X , Modèles moléculaires , Flavine adénine dinucléotide/métabolisme , Flavine adénine dinucléotide/composition chimique
15.
Article de Anglais | MEDLINE | ID: mdl-38908503

RÉSUMÉ

BACKGROUND: Low folate intake and methylenetetrahydrofolate reductase (MTHFR) C677T polymorphism have been suggested to increase the risk of Alzheimer's disease (AD). However, the synergistic effects and their impact on brain structure and perfusion remain unclear. METHODS: This study explored the effects of dietary and genetic deficiencies in folate metabolism on the volume of the hippocampal subregions, cerebral perfusion, and cognitive decline in 71 cognitively unimpaired (CU) individuals and 102 patients with mild cognitive impairment (MCI) due to AD or AD. All participants underwent magnetic resonance imaging, laboratory examinations, and neuropsychological assessments. The hippocampal subfields were segmented using Freesurfer, and arterial spin labeling was used to measure the cerebral blood flow. RESULTS: We found a significant group-by-MTHFR interaction effect on folate. Patients with AD and the 677 T allele showed hypoperfusion in the left precuneus compared to patients without this mutation, which mediated the relationship between low folate level and cognitive decline in patients carrying the 677 T allele. Moreover, a synergistic effect was observed for the combination of decreased folate concentrations and the presence of the MTHFR 677 T allele on the atrophy of specific hippocampal subregions in patients with AD. CONCLUSIONS: In addition to offering insights into the neuronal mechanism underlying gene-dependent folate-induced cognitive impairment in AD, these findings may have clinical significance for the allocation of auxiliary folate supplementation therapy in patients with AD with low folate levels and carrying the MTHFR 677 T allele and may eventually promote the selection of early individualized AD drug therapy.


Sujet(s)
Maladie d'Alzheimer , Circulation cérébrovasculaire , Dysfonctionnement cognitif , Acide folique , Hippocampe , Imagerie par résonance magnétique , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/métabolisme , Mâle , Hippocampe/imagerie diagnostique , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Femelle , Sujet âgé , Dysfonctionnement cognitif/génétique , Dysfonctionnement cognitif/métabolisme , Circulation cérébrovasculaire/physiologie , Circulation cérébrovasculaire/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Tests neuropsychologiques , Sujet âgé de 80 ans ou plus
16.
J Reprod Immunol ; 164: 104274, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38865894

RÉSUMÉ

Numerous recent studies have examined the impact epigenetics-including DNA methylation-has on spermatogenesis and male infertility. Differential methylation of several genes has been linked to compromised spermatogenesis and/or reproductive failure. Specifically, male infertility has been frequently associated with DNA methylation abnormalities of MEST and H19 inside imprinted genes and MTHFR within non-imprinted genes. Microbial infections mainly result in male infertility because of the immune response triggered by the bacteria' accumulation of immune cells, proinflammatory cytokines, and chemokines. Thus, bacterially produced epigenetic dysregulations may impact host cell function, supporting host defense or enabling pathogen persistence. So, it is possible to think of pathogenic bacteria as potential epimutagens that can alter the epigenome. It has been demonstrated that dysregulated levels of LncRNA correlate with motility and sperm count in ejaculated spermatozoa from infertile males. Therefore, a thorough understanding of the relationship between decreased reproductive capacity and sperm DNA methylation status should aid in creating new diagnostic instruments for this condition. To fully understand the mechanisms influencing sperm methylation and how they relate to male infertility, more research is required.


Sujet(s)
Méthylation de l'ADN , Épigenèse génétique , Infertilité masculine , Spermatogenèse , Spermatozoïdes , Mâle , Humains , Infertilité masculine/immunologie , Infertilité masculine/génétique , Infertilité masculine/microbiologie , Épigenèse génétique/immunologie , Méthylation de l'ADN/immunologie , Spermatozoïdes/immunologie , Spermatogenèse/génétique , Spermatogenèse/immunologie , Animaux , ARN long non codant/génétique , ARN long non codant/immunologie , Infections bactériennes/immunologie , Infections bactériennes/génétique , Methylenetetrahydrofolate reductase (NADPH2)/génétique
17.
Zhonghua Yi Xue Za Zhi ; 104(24): 2256-2259, 2024 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-38901983

RÉSUMÉ

The clinical manifestations, biochemical and metabolic data, genetic variations and treatment data of children with MTHFR gene variant induced hyperhomocysteinemia admitted to Hangzhou Children's Hospital and Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine from November 2015 to September 2021 were analysed retrospectively. A total of 15 pediatric patients were included, including 10 males and 5 females, with onset ages ranging from 6 days to 18 years old and confirmed ages ranging from 40 days to 18 years old. One confirmed case was detected through neonatal screening, and the remaining 14 cases were all diagnosed through genetic diagnosis after onset. The main clinical manifestations were feeding difficulties, hypotonia, epilepsy, developmental delay. All patients had elevated levels of blood homocysteine, with blood homocysteine levels before and after treatment being (151.46±57.44) µmol/L and (69.96±32.88) µmol/L, significantly decreased after treatment compared with before treatment, with a statistically significant difference (P<0.001). The blood methionine level before the treatment was 9.40 (6.20, 11.96) µmol/L, normal or slightly decreased compared to the reference range. The methionine level returned to normal after treatment. A total of 19 MTHFR gene variants were detected, with 6 being unreported variants and 13 being known variants. c.1316C>T (p.L439P) was the most common variant(16.6%,5/30). All the patients had varied neurological damages, with 7 patients improved after metabolic therapy by carnitine and folinic acid, 8 patients experiencing developmental delay, and 1 patient experiencing frequent epilepsy. The clinical manifestations of MTHFR gene variation-related hyperhomocysteinemia are complex and variable. Early-onset and homozygous variants often have a poor prognosis. Blood homocysteine, blood amino acid analysis, serum total homocysteine assay and gene testing are helpful for early diagnosis.


Sujet(s)
Homocystéine , Hyperhomocystéinémie , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Hyperhomocystéinémie/génétique , Mâle , Femelle , Enfant , Enfant d'âge préscolaire , Adolescent , Nourrisson , Études rétrospectives , Homocystéine/sang , Nouveau-né , Mutation , Méthionine
18.
Nutr Res ; 127: 63-74, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876040

RÉSUMÉ

Betaine supplementation is used by athletes, but its mechanism of action is still not fully understood. We hypothesized that betaine supplementation would increase betaine concentration and alter amino acid profiles in relation to MTHFR genotype and dose in physically active males. The study followed a randomized placebo-controlled cross-over design. Blood samples were collected before and after each supplementation period. Serum was analyzed for amino acid profile, homocysteine, betaine, choline, and trimethylamine N-oxide (TMAO) concentrations. For the washout analysis, only participants starting with betaine were included (n = 20). Statistical analysis revealed no differences in the amino acid profile after betaine supplementation. However, betaine concentration significantly increased after betaine supplementation (from 4.89 ± 1.59 µg/mL to 17.31 ± 9.21 µg/mL, P < .001), with a greater increase observed in MTHFR (C677T, rs180113) T-allele carriers compared to CC (P = .027). Betaine supplementation caused a decrease in homocysteine concentration (from 17.04 ± 4.13 µmol/L to 15.44 ± 3.48 µmol/L, P = .00005) and a non-significant increase in TMAO concentrations (from 0.27 ± 0.20 µg/ml to 0.44 ± 0.70 µg/ml, P = .053), but had no effect on choline concentrations. Serum betaine concentrations were not significantly different after the 21-day washout from the baseline values (baseline: 4.93 ± 1.87 µg/mL and after washout: 4.70 ± 1.70 µg/mL, P = 1.000). In conclusion, betaine supplementation increased betaine and decreased homocysteine concentrations, but did not affect the amino acid profile or choline concentrations in healthy active males. Betaine concentrations may be dependent on MTHFR genotype.


Sujet(s)
Acides aminés , Bétaïne , Choline , Études croisées , Compléments alimentaires , Génotype , Methylenetetrahydrofolate reductase (NADPH2) , Humains , Bétaïne/sang , Mâle , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Acides aminés/sang , Adulte , Jeune adulte , Choline/sang , Homocystéine/sang , Méthylamines/sang
19.
Nutrients ; 16(11)2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38892484

RÉSUMÉ

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.


Sujet(s)
5-Methyltetrahydrofolate-homocysteine s-methyltransferase , Cholestérol LDL , Compléments alimentaires , Ferredoxine-NADP reductase , Homocystéine , Methylenetetrahydrofolate reductase (NADPH2) , Phosphate de pyridoxal , Tétrahydrofolates , Vitamine B12 , Humains , Adulte d'âge moyen , Homocystéine/sang , Femelle , Mâle , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Méthode en double aveugle , 5-Methyltetrahydrofolate-homocysteine s-methyltransferase/génétique , Cholestérol LDL/sang , Sujet âgé , Vitamine B12/administration et posologie , Vitamine B12/analogues et dérivés , Adulte , Ferredoxine-NADP reductase/génétique , Tétrahydrofolates/administration et posologie , Polymorphisme génétique , Complexe vitaminique B/usage thérapeutique , Complexe vitaminique B/administration et posologie , Complexe vitaminique B/pharmacologie
20.
Nutrients ; 16(12)2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38931291

RÉSUMÉ

Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age. Many women with PCOS have been found to have an unbalanced diet and deficiencies in essential nutrients. This study aimed to assess the levels of folate and vitamin B12 (B12) and their relationship with metabolic factors in women with PCOS. Anthropometric, clinical, and genetic analyses were conducted to evaluate markers related to one-carbon metabolism in women with PCOS and in a control group. The PCOS group had a higher BMI and HOMA-IR (1.7 vs. 3.1; p < 0.0001). HDL cholesterol levels were 23% lower and triglyceride levels were 74% higher in women with PCOS. Although there were no significant differences in folate and B12 levels between the PCOS and control groups, over 60% of women with PCOS had low B12 levels (<300 pg/mL) and high homocysteine levels. In addition, the MTHFR A1298C and C677T polymorphisms were not associated with PCOS. Moreover, erythrocyte folate levels were positively correlated with fasting glucose, triglycerides, and free androgen index, and negatively correlated with SHBG and LH levels. These results suggest that B vitamins may be associated with the metabolic phenotype in PCOS. This study emphasizes the potential link between folate, vitamin B12, and metabolic and hormonal outcomes in women with PCOS.


Sujet(s)
Acide folique , Syndrome des ovaires polykystiques , Vitamine B12 , Humains , Femelle , Syndrome des ovaires polykystiques/sang , Syndrome des ovaires polykystiques/génétique , Vitamine B12/sang , Acide folique/sang , Adulte , Chili/épidémiologie , Jeune adulte , Triglycéride/sang , Homocystéine/sang , Indice de masse corporelle , Glycémie/métabolisme , Methylenetetrahydrofolate reductase (NADPH2)/génétique , Insulinorésistance , Cholestérol HDL/sang , Études cas-témoins , Marqueurs biologiques/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE