Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Biochemistry (Mosc) ; 89(3): 562-573, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648773

ABSTRACT

The contents of homocysteine (HCy), cyanocobalamin (vitamin B12), folic acid (vitamin B9), and pyridoxine (vitamin B6) were analyzed and the genotypes of the main gene polymorphisms associated with folate metabolism (C677T and A1298C of the MTHFR gene, A2756G of the MTR gene and A66G of the MTRR gene) were determined in children at the onset of multiple sclerosis (MS) (with disease duration of no more than six months), healthy children under 18 years (control group), healthy adults without neurological pathology, adult patients with MS at the onset of disease, and adult patients with long-term MS. A significant increase in the HCy levels was found in children at the MS onset compared to healthy children of the corresponding age. It was established that the content of HCy in children has a high predictive value. At the same time, an increase in the HCy levels was not accompanied by the deficiency of vitamins B6, B9, and B12 in the blood. The lack of correlation between the laboratory signs of vitamin deficiency and HCy levels may be due to the polymorphic variants of folate cycle genes. An increased HCy level should be considered as a marker of functional disorders of folate metabolism accompanying the development of pathological process in pediatric MS. Our finding can be used to develop new approaches to the prevention of demyelination in children and treatment of pediatric MS.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase , Folic Acid , Homocysteine , Methylenetetrahydrofolate Reductase (NADPH2) , Multiple Sclerosis , Humans , Homocysteine/blood , Homocysteine/metabolism , Multiple Sclerosis/blood , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Folic Acid/blood , Folic Acid/metabolism , Female , Male , Child , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Adult , Adolescent , Vitamin B Deficiency/complications , Vitamin B Deficiency/metabolism , Vitamin B Deficiency/blood , Ferredoxin-NADP Reductase/genetics , Ferredoxin-NADP Reductase/metabolism , Vitamin B 12/blood , Vitamin B 12/metabolism , Age of Onset
2.
Zhonghua Er Ke Za Zhi ; 62(4): 357-362, 2024 Mar 25.
Article in Chinese | MEDLINE | ID: mdl-38527507

ABSTRACT

Objective: To explore the diagnosis and treatment of adolescence-onset methylenetetrahydrofolate reductase (MTHFR) deficiency. Methods: This was a retrospective case study. Nine patients with adolescence-onset MTHFR deficiency were diagnosed at Peking University First Hospital from January 2016 to December 2022, and followed up for more than 1 year. Their general information, clinical manifestations, laboratory tests, cranial images, MTHFR gene variants, diagnosis, treatment, and outcome were analyzed retrospectively. Results: The 9 patients came from 8 families. They had symptoms at age of 8.0 years to 17.0 years and diagnosed at 9.0 years to 17.5 years. Eight were male and 1 was female. Two patients were brothers, the elder brother developed abnormal gait at 17.0 years; and the younger brother was then diagnosed at 15.0 years of age and treated at the asymptomatic stage, who was 18.0 years old with normal condition during this study. The main manifestations of the 8 symptomatic patients included progressive dyskinesia and spastic paralysis of the lower limbs, with or without intellectual decline, cognitive impairment and behavioral abnormalities. Totally, 15 variants of MTHFR gene were identified in the 9 patients, including 8 novel variants. Five patients had brain image abnormalities. Increased plasma total homocysteine level (65-221 µmol/L) was found in all patients, and decreased to 20-70 µmol/L after treatment with betaine and calcium folinate. Besides, the 8 symptomatic patients had their behavior and cognitive problems significantly improved, with a legacy of lower limb motor disorders. Conclusions: Late-onset MTHFR deficiency can occur in adolescence. The diagnosis is usually delayed because of non-specific clinical symptoms. The test of blood total homocysteine could be used as a selective screening test. Eight novel varients of MTHFR gene were identified. Timely treatment can improve clinical condition significantly, and pre-symptomatic treatment may prevent brain damage.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2) , Muscle Spasticity , Adolescent , Child , Female , Humans , Male , Homocysteine/therapeutic use , Homocystinuria , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/diagnosis , Muscle Spasticity/genetics , Muscle Spasticity/drug therapy , Psychotic Disorders , Retrospective Studies
3.
Cell Mol Life Sci ; 79(8): 410, 2022 Jul 11.
Article in English | MEDLINE | ID: mdl-35821533

ABSTRACT

Beyond well-assessed risk factors, cardiovascular events could be also associated with the presence of epigenetic and genetic alterations, such as the methylenetetrahydrofolate-reductase (MTHFR) C677T polymorphism. This gene variant is related to increased circulating levels of homocysteine (Hcy) and cardiovascular risk. However, heterozygous carriers have an augmented risk of cardiovascular accidents independently from normal Hcy levels, suggesting the presence of additional deregulated processes in MTHFR C677T carriers. Here, we hypothesize that targeting Sirtuin 1 (SIRT1) could be an alternative mechanism to control the cardiovascular risk associated to MTHFR deficiency condition. Flow Mediated Dilatation (FMD) and light transmission aggregometry assay were performed in subjects carrying MTHFR C677T allele after administration of resveratrol, the most powerful natural clinical usable compound that owns SIRT1 activating properties. MTHFR C677T carriers with normal Hcy levels revealed endothelial dysfunction and enhanced platelet aggregation associated with SIRT1 downregulation. SIRT1 activity stimulation by resveratrol intake was able to override these abnormalities without affecting Hcy levels. Impaired endothelial function, bleeding time, and wire-induced thrombus formation were rescued in a heterozygous Mthfr-deficient (Mthfr+/-) mouse model after resveratrol treatment. Using a cell-based high-throughput multiplexed screening (HTS) assay, a novel selective synthetic SIRT1 activator, namely ISIDE11, was identified. Ex vivo and in vivo treatment of Mthfr+/- mice with ISIDE11 rescues endothelial vasorelaxation and reduces wire-induced thrombus formation, effects that were abolished by SIRT1 inhibitor. Moreover, platelets from MTHFR C677T allele carriers treated with ISIDE11 showed normalization of their typical hyper-reactivity. These results candidate SIRT1 activation as a new therapeutic strategy to contain cardio and cerebrovascular events in MTHFR carriers.


Subject(s)
Homocystinuria , Methylenetetrahydrofolate Reductase (NADPH2) , Sirtuin 1 , Thrombosis , Animals , Genotype , Homocystinuria/drug therapy , Homocystinuria/metabolism , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Muscle Spasticity , Psychotic Disorders/metabolism , Resveratrol/pharmacology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Thrombosis/drug therapy , Thrombosis/genetics , Thrombosis/metabolism , Thrombosis/prevention & control
5.
J Inherit Metab Dis ; 45(4): 848-861, 2022 07.
Article in English | MEDLINE | ID: mdl-35460084

ABSTRACT

MTHFR deficiency is a severe inborn error of metabolism leading to impairment of the remethylation of homocysteine to methionine. Neonatal and early-onset patients mostly exhibit a life-threatening acute neurologic deterioration. Furthermore, data on early-onset patients' long-term outcomes are scarce. The aims of this study were (1) to study and describe the clinical and laboratory parameters of early-onset MTHFR-deficient patients (i.e., ≤3 months of age) and (2) to identify predictive factors for severe neurodevelopmental outcomes in a cohort with early and late onset MTHFR-deficient patients. To this end, we conducted a retrospective, multicentric, international cohort study on 72 patients with MTHFR deficiency from 32 international metabolic centres. Characteristics of the 32 patients with early-onset MTHFR deficiency were described at time of diagnosis and at the last follow-up visit. Logistic regression analysis was used to identify predictive factors of severe neurodevelopmental outcome in a broader set of patients with early and non-early-onset MTHFR deficiency. The majority of early-onset MTHFR-deficient patients (n = 32) exhibited neurologic symptoms (76%) and feeding difficulties (70%) at time of diagnosis. At the last follow-up visit (median follow-up time of 8.1 years), 76% of treated early-onset patients (n = 29) exhibited a severe neurodevelopmental outcome. Among the whole study population of 64 patients, pre-symptomatic diagnosis was independently associated with a significantly better neurodevelopmental outcome (adjusted OR 0.004, [0.002-0.232]; p = 0.003). This study provides evidence for benefits of pre-symptomatic diagnosis and appropriate therapeutic management, highlighting the need for systematic newborn screening for MTHFR deficiency and pre-symptomatic treatment that may improve outcome.


Subject(s)
Homocystinuria , Cohort Studies , Homocysteine , Homocystinuria/diagnosis , Homocystinuria/drug therapy , Humans , Infant, Newborn , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity/diagnosis , Psychotic Disorders , Retrospective Studies
6.
J Inherit Metab Dis ; 45(4): 719-733, 2022 07.
Article in English | MEDLINE | ID: mdl-35358327

ABSTRACT

Patient registries for rare diseases enable systematic data collection and can also be used to facilitate postauthorization safety studies (PASS) for orphan drugs. This study evaluates the PASS for betaine anhydrous (Cystadane), conducted as public private partnership (PPP) between the European network and registry for homocystinurias and methylation defects and the marketing authorization holder (MAH). Data were prospectively collected, 2013-2016, in a noninterventional, international, multicenter, registry study. Putative adverse and severe adverse events were reported to the MAH's pharmacovigilance. In total, 130 individuals with vitamin B6 nonresponsive (N = 54) and partially responsive (N = 7) cystathionine beta-synthase (CBS) deficiency, as well as 5,10-methylenetetrahydrofolate reductase (MTHFR; N = 21) deficiency and cobalamin C (N = 48) disease were included. Median (range) duration of treatment with betaine anhydrous was 6.8 (0-9.8) years. The prescribed betaine dose exceeded the recommended maximum (6 g/day) in 49% of individuals older than 10 years because of continued dose adaptation to weight; however, with disease-specific differences (minimum: 31% in B6 nonresponsive CBS deficiency, maximum: 67% in MTHFR deficiency). Despite dose escalation no new or potential risk was identified. Combined disease-specific treatment decreased mean ± SD total plasma homocysteine concentrations from 203 ± 116 to 81 ± 51 µmol/L (p < 0.0001), except in MTHFR deficiency. Recommendations for betaine anhydrous dosage were revised for individuals ≥ 10 years. PPPs between MAH and international scientific consortia can be considered a reliable model for implementing a PASS, reutilizing well-established structures and avoiding data duplication and fragmentation.


Subject(s)
Homocystinuria , Psychotic Disorders , Betaine/adverse effects , Cystathionine beta-Synthase , Homocysteine , Homocystinuria/drug therapy , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity
7.
BMJ Case Rep ; 15(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-34983810

ABSTRACT

Methylenetetrahydrofolate reductase (MTHFR) deficiency is a rare autosomal recessive inherited inborn error of metabolism, which presents with various severity depending on the level of residual enzyme activity. In neonates, it can present with recurrent hypoventilation episodes, persistent encephalopathy with or without microcephaly. MTHFR deficiency also results in hyperhomocysteinemia, homocystinuria and hypomethionemia. We report a male neonate with severe MTHFR deficiency presenting to us on third week of life with progressive encephalopathy, microcephaly, seizures, central hypoventilation. There was similar history in the previous sibling. The patient's blood lactate, ammonia, tandem mass spectrometry for amino acids and acyl carnitine were normal. He remained encephalopathic with progressive increase in need of respiratory support in spite of supportive treatment and metabolic cocktail consisting of riboflavin, pyridoxine, coenzyme Q and carnitine. This neonate had novel homozygous mutation, which results in MTHFR deficiency. In newborn with hypoventilation or recurrent apnoea with encephalopathy and microcephaly, MTHFR deficiency should be considered as a differential diagnosis. Mutation study helps in confirming diagnosis; however, extended newborn metabolic screening with homocysteine level could help in early diagnosis of these cases.


Subject(s)
Homocystinuria , Homocystinuria/complications , Homocystinuria/diagnosis , Homocystinuria/genetics , Humans , Hypoventilation , Infant, Newborn , Male , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity , Psychotic Disorders
9.
Clin Dysmorphol ; 31(2): 59-65, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-34845156

ABSTRACT

Inherited methylenetetrahydrofolate reductase (MTHFR) deficiency is associated with a wide spectrum of disorders including homocystinuria. This study aims to describe the neurological phenotypes and molecular profiles of patients with homocystinuria caused by biallelic variants in MTHFR. We report six subjects with MTHFR deficiency who presented with variable neurological phenotypes which could be viewed as a continuous spectrum. Fatal infantile encephalopathy was observed in one family, whereas another patient presented at 27 years with acute leukoencephalopathy and recovered within 3 months. Intermediate forms presenting as complicated hereditary spastic paraparesis of variable severity were observed in four subjects. Clinical and molecular information of the 207 cases reported in literature were also retrieved and analyzed. We categorized all subjects into three categories - severe, intermediate and mild forms according to the clinical presentation. In addition, a total of 286 disease-causing variations reported to date were analyzed. These included seven disease-causing variants reported in this study of which one is novel. Some genotype-phenotype correlation could be seen which corroborated with previous observations. However, inter- and intrafamilial variability was also noted. Treatment with betaine, B12 and folic acid was started in four subjects with variable outcomes.


Subject(s)
Homocystinuria , Betaine/therapeutic use , Homocystinuria/complications , Homocystinuria/diagnosis , Homocystinuria/genetics , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Muscle Spasticity/complications , Muscle Spasticity/diagnosis , Muscle Spasticity/genetics , Nervous System Diseases/genetics , Phenotype
10.
Nutr Neurosci ; 25(3): 558-566, 2022 Mar.
Article in English | MEDLINE | ID: mdl-32448097

ABSTRACT

Objective: Elevated homocysteine concentrations are a risk factor for stroke. A common genetic polymorphism in methylenetetrahydrofolate reductase (MTHFR 677 C→T) results in elevated levels of homocysteine. MTHFR plays a critical role in the synthesis of S-adenosylmethionine (SAM), a global methyl donor. Our previous work has demonstrated that Mthfr+/- mice, which model the MTHFR polymorphism in humans, are more vulnerable to ischemic damage. The aim of this study was to investigate the cellular mechanisms by which the MTHFR-deficiency changes the brain in the context of ischemic stroke injury.Methods: In the present study, three-month-old male Mthfr+/- and wild-type littermate mice were subjected to photothrombosis (PT) damage. Four weeks after PT damage, animals were tested on behavioral tasks, in vivo imaging was performed using T2-weighted MRI, and brain tissue was collected for histological analysis.Results: Mthfr+/- animals used their non-impaired forepaw more to explore the cylinder and had a larger damage volume compared to wild-type littermates. In brain tissue of Mthfr+/- mice methionine adenosyltransferase II alpha (MAT2A) protein levels were decreased within the damage hemisphere and increased levels in hypoxia-induced factor 1 alpha (HIF-1α) in non-damage hemisphere. There was an increased antioxidant response in the damage site as indicated by higher levels of nuclear factor erythroid 2-related factor 2 (Nrf2) in neurons and astrocytes and neuronal superoxide dismutase 2 (SOD2) levels.Conclusions: Our results suggest that Mthfr+/- mice are more vulnerable to PT-induced stroke damage through the regulation of the cellular response. The increased antioxidant response we observed may be compensatory to the damage amount.


Subject(s)
Homocystinuria , Ischemic Stroke , Methylenetetrahydrofolate Reductase (NADPH2) , Muscle Spasticity , Animals , Homocysteine , Homocystinuria/complications , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Male , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice , Psychotic Disorders
12.
Nutrients ; 13(6)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204335

ABSTRACT

BACKGROUND: DNA methylation is the best epigenetic mechanism for explaining the interactions between nutrients and genes involved in intrauterine growth and development programming. A possible contributor of methylation abnormalities to congenital heart disease is the folate methylation regulatory pathway; however, the mechanisms and methylation patterns of VSD-associated genes are not fully understood. OBJECTIVE: To determine if maternal dietary intake of folic acid (FA) is related to the methylation status (MS) of VSD-associated genes (AXIN1, MTHFR, TBX1, and TBX20). METHODS: Prospective case-control study; 48 mothers and their children were evaluated. The mothers' dietary variables were collected through a food frequency questionnaire focusing on FA and the consumption of supplements with FA. The MS of promoters of genes was determined in the children. RESULTS: The intake of FA supplements was significantly higher in the control mothers. In terms of maternal folic acid consumption, significant differences were found in the first trimester of pregnancy. Significant differences were observed in the MS of MTHFR and AXIN1 genes in VSD and control children. A correlation between maternal FA supplementation and MS of AXIN1 and TBX20 genes was found in control and VSD children, respectively. CONCLUSIONS: A lower MS of AXIN1 genes and a higher MS of TBX20 genes is associated with FA maternal supplementation.


Subject(s)
Folic Acid/metabolism , Genetic Association Studies , Genetic Predisposition to Disease/genetics , Heart Septal Defects, Ventricular/genetics , Case-Control Studies , Child , DNA Methylation , Diet , Dietary Supplements , Epigenesis, Genetic , Female , Heart Defects, Congenital , Homocystinuria , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity , Pregnancy , Prospective Studies , Psychotic Disorders
13.
Am J Hum Genet ; 108(7): 1283-1300, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34214447

ABSTRACT

Most rare clinical missense variants cannot currently be classified as pathogenic or benign. Deficiency in human 5,10-methylenetetrahydrofolate reductase (MTHFR), the most common inherited disorder of folate metabolism, is caused primarily by rare missense variants. Further complicating variant interpretation, variant impacts often depend on environment. An important example of this phenomenon is the MTHFR variant p.Ala222Val (c.665C>T), which is carried by half of all humans and has a phenotypic impact that depends on dietary folate. Here we describe the results of 98,336 variant functional-impact assays, covering nearly all possible MTHFR amino acid substitutions in four folinate environments, each in the presence and absence of p.Ala222Val. The resulting atlas of MTHFR variant effects reveals many complex dependencies on both folinate and p.Ala222Val. MTHFR atlas scores can distinguish pathogenic from benign variants and, among individuals with severe MTHFR deficiency, correlate with age of disease onset. Providing a powerful tool for understanding structure-function relationships, the atlas suggests a role for a disordered loop in retaining cofactor at the active site and identifies variants that enable escape of inhibition by S-adenosylmethionine. Thus, a model based on eight MTHFR variant effect maps illustrates how shifting landscapes of environment- and genetic-background-dependent missense variation can inform our clinical, structural, and functional understanding of MTHFR deficiency.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mutation, Missense , Amino Acid Substitution , DNA Mutational Analysis , Diploidy , Gene Library , Genotype , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/physiology , Saccharomyces cerevisiae/genetics
14.
Development ; 148(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34128976

ABSTRACT

5,10-Methylenetetrahydrofolate reductase (MTHFR) is a crucial enzyme in the folate metabolic pathway with a key role in generating methyl groups. As MTHFR deficiency impacts male fertility and sperm DNA methylation, there is the potential for epimutations to be passed to the next generation. Here, we assessed whether the impact of MTHFR deficiency on testis morphology and sperm DNA methylation is exacerbated across generations in mouse. Although MTHFR deficiency in F1 fathers has only minor effects on sperm counts and testis weights and histology, F2 generation sons show further deterioration in reproductive parameters. Extensive loss of DNA methylation is observed in both F1 and F2 sperm, with >80% of sites shared between generations, suggestive of regions consistently susceptible to MTHFR deficiency. These regions are generally methylated during late embryonic germ cell development and are enriched in young retrotransposons. As retrotransposons are resistant to reprogramming of DNA methylation in embryonic germ cells, their hypomethylated state in the sperm of F1 males could contribute to the worsening reproductive phenotype observed in F2 MTHFR-deficient males, compatible with the intergenerational passage of epimutations.


Subject(s)
DNA Methylation , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Reproduction/physiology , Retroelements/genetics , Animals , Epigenomics , Fathers , Female , Folic Acid/metabolism , Germ Cells , Homocystinuria , Male , Mice , Mice, Inbred C57BL , Muscle Spasticity , Psychotic Disorders , Spermatozoa/metabolism
15.
Mol Nutr Food Res ; 65(14): e2100197, 2021 07.
Article in English | MEDLINE | ID: mdl-34010503

ABSTRACT

SCOPE: Many pregnant women have higher folic acid (FA) intake due to food fortification and increased vitamin use. It is reported that diets containing five-fold higher FA than recommended for mice (5xFASD) during pregnancy resulted in methylenetetrahydrofolate reductase (MTHFR) deficiency and altered choline/methyl metabolism, with neurobehavioral abnormalities in newborns. The goal is to determine whether these changes have their origins in the placenta during embryonic development. METHODS AND RESULTS: Female mice are fed control diet or 5xFASD for a month before mating and maintained on these diets until embryonic day 17.5. 5xFASD led to pseudo-MTHFR deficiency in maternal liver and altered choline/methyl metabolites in maternal plasma (increased methyltetrahydrofolate and decreased betaine). Methylation potential (S-adenosylmethionine:S-adenosylhomocysteine ratio) and glycerophosphocholine are decreased in placenta and embryonic liver. Folic acid supplemented diet results in sex-specific transcriptome profiles in placenta, with validation of dietary expression changes of 29 genes involved in angiogenesis, receptor biology or neurodevelopment, and altered methylation of the serotonin receptor 2A gene. CONCLUSION: Moderate increases in folate intake during pregnancy result in placental metabolic and gene expression changes, particularly in angiogenesis, which may contribute to abnormal behavior in pups. These results are relevant for determining a safe upper limit for folate intake during pregnancy.


Subject(s)
Folic Acid/pharmacology , Homocystinuria/chemically induced , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/chemically induced , Placenta/metabolism , Sex Factors , Animals , DNA Methylation , Dietary Supplements , Female , Folic Acid/adverse effects , Gene Expression/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Phthalic Acids/blood , Pregnancy , Psychotic Disorders , S-Adenosylmethionine/blood , Transcriptome/drug effects
16.
Int J Mol Sci ; 23(1)2021 Dec 23.
Article in English | MEDLINE | ID: mdl-35008593

ABSTRACT

MTHFR deficiency still deserves an investigation to associate the phenotype to protein structure variations. To this aim, considering the MTHFR wild type protein structure, with a catalytic and a regulatory domain and taking advantage of state-of-the-art computational tools, we explore the properties of 72 missense variations known to be disease associated. By computing the thermodynamic ΔΔG change according to a consensus method that we recently introduced, we find that 61% of the disease-related variations destabilize the protein, are present both in the catalytic and regulatory domain and correspond to known biochemical deficiencies. The propensity of solvent accessible residues to be involved in protein-protein interaction sites indicates that most of the interacting residues are located in the regulatory domain, and that only three of them, located at the interface of the functional protein homodimer, are both disease-related and destabilizing. Finally, we compute the protein architecture with Hidden Markov Models, one from Pfam for the catalytic domain and the second computed in house for the regulatory domain. We show that patterns of disease-associated, physicochemical variation types, both in the catalytic and regulatory domains, are unique for the MTHFR deficiency when mapped into the protein architecture.


Subject(s)
Homocystinuria/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/genetics , Catalytic Domain/genetics , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Protein Interaction Maps/genetics , Psychotic Disorders/genetics
17.
J Inherit Metab Dis ; 44(3): 777-786, 2021 05.
Article in English | MEDLINE | ID: mdl-33089527

ABSTRACT

5,10-Methylenetetrahydrofolate reductase (MTHFR) deficiency usually presents as a severe neonatal disease. This study aimed to characterize natural history, biological and molecular data, and response to treatment of patients with late-onset MTHFR deficiency. The patients were identified through the European Network and Registry for Homocystinuria and Methylation Defects and the Adult group of the French Society for Inherited Metabolic Diseases; data were retrospectively colleted. To identify juvenile to adult-onset forms of the disease, we included patients with a diagnosis established after the age of 10 years. We included 14 patients (median age at diagnosis: 32 years; range: 11-54). At onset (median age: 20 years; range 9-38), they presented with walking difficulties (n = 8), cognitive decline (n = 3) and/or seizures (n = 3), sometimes associated with mild mental retardation (n = 6). During the disease course, symptoms were almost exclusively neurological with cognitive dysfunction (93%), gait disorders (86%), epilepsy (71%), psychiatric symptoms (57%), polyneuropathy (43%), and visual deficit (43%). Mean diagnostic delay was 14 years. Vascular events were observed in 28% and obesity in 36% of the patients. One patient remained asymptomatic at the age of 55 years. Upon treatment, median total homocysteine decreased (from 183 µmol/L, range 69-266, to 90 µmol/L, range 20-142) and symptoms improved (n = 9) or stabilized (n = 4). Missense pathogenic variants in the C-terminal regulatory domain of the protein were over-represented compared to early-onset cases. Residual MTHFR enzymatic activity in skin fibroblasts (n = 4) was rather high (17%-58%). This series of patients with late-onset MTHFR deficiency underlines the still unmet need of a prompt diagnosis of this treatable disease.


Subject(s)
Homocystinuria/diagnosis , Homocystinuria/pathology , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/diagnosis , Muscle Spasticity/pathology , Adolescent , Adult , Age of Onset , Child , Delayed Diagnosis , Epilepsy/diagnosis , Epilepsy/pathology , Female , Humans , Intellectual Disability/diagnosis , Intellectual Disability/pathology , Male , Middle Aged , Psychotic Disorders/diagnosis , Psychotic Disorders/pathology , Retrospective Studies , Seizures/diagnosis , Seizures/pathology , Young Adult
19.
J Nutr Biochem ; 88: 108554, 2021 02.
Article in English | MEDLINE | ID: mdl-33220403

ABSTRACT

Food fortification with folic acid and increased use of vitamin supplements have raised concerns about high folic acid intake. We previously showed that high folic acid intake was associated with hepatic degeneration, decreased levels of methylenetetrahydrofolate reductase (MTHFR), lower methylation potential, and perturbations of lipid metabolism. MTHFR synthesizes the folate derivative for methylation reactions. In this study, we assessed the possibility that high folic acid diets, fed to wild-type and Mthfr+/- mice, could alter DNA methylation and/or deregulate hepatic cholesterol homeostasis. Digital restriction enzyme analysis of methylation in liver revealed DNA hypomethylation of a CpG in the lipolysis-stimulated lipoprotein receptor (Lsr) gene, which is involved in hepatic uptake of cholesterol. Pyrosequencing confirmed this methylation change and identified hypomethylation of several neighboring CpG dinucleotides. Lsr expression was increased and correlated negatively with DNA methylation and plasma cholesterol. A putative binding site for E2F1 was identified. ChIP-qPCR confirmed reduced E2F1 binding when methylation at this site was altered, suggesting that it could be involved in increasing Lsr expression. Expression of genes in cholesterol synthesis, transport or turnover (Abcg5, Abcg8, Abcc2, Cyp46a1, and Hmgcs1) was perturbed by high folic acid intake. We also observed increased hepatic cholesterol and increased expression of genes such as Sirt1, which might be involved in a rescue response to restore cholesterol homeostasis. Our work suggests that high folic acid consumption disturbs cholesterol homeostasis in liver. This finding may have particular relevance for MTHFR-deficient individuals, who represent ~10% of many populations.


Subject(s)
Cholesterol/metabolism , DNA Methylation/drug effects , Folic Acid/pharmacology , Liver/metabolism , Receptors, Lipoprotein/metabolism , Animals , Choline/metabolism , Diet , Folic Acid/administration & dosage , Food, Fortified , Homeostasis/drug effects , Humans , Lipid Metabolism , Male , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Mice , Mice, Inbred BALB C , Multidrug Resistance-Associated Protein 2 , Receptors, Lipoprotein/genetics
20.
Arch Gynecol Obstet ; 303(4): 1089-1098, 2021 04.
Article in English | MEDLINE | ID: mdl-33128585

ABSTRACT

OBJECTIVE: To investigate whether polymorphism of MTHFR C677T or MTHFR A1298C is associated with recurrent implantation failure (RIF). STUDY DESIGN: This is a systematic review and meta-analysis. Pubmed, EMBASE, and CNKI (China national Knowledge Infrastructure) were searched for case-control studies that evaluated the associations between MTHFR polymorphisms (MTHFR C677T and MTHFR A1298C) and RIF. The pooled odds ratios (ORs) with 95% confidence intervals (CIs) were reported to evaluate the strength of association. Data were synthesized using the random-effect model. RESULTS: Nine case-control studies consisted of 1812 women were included in the quantitative meta-analyses (754 were RIF patients, 1058 were control participants). The synthesized results showed that polymorphism of MTHFR C677T (allele model: OR 1.23, 95% CI 0.99-1.53; dominant model: OR 1.24, 95% CI 0.99-1.54; recessive model: OR 1.31, 95% CI 0.78-2.12; homozygotic model: OR 1.39, 95% CI 0.84-2.28; heterozygotic model: OR 1.14, 95% CI 0.90-1.45) or MTHFR A1298C (allele model: OR 1.11, 95% CI 0.78-1.59; dominant model: OR 0.91, 95% CI 0.65-1.26; recessive model: OR 2.04, 95% CI 0.90-4.64; homozygotic model: OR 1.86, 95% CI 0.79-4.38; heterozygotic model: OR 0.77, 95% CI 0.59-0.99) was not significantly associated with RIF. CONCLUSIONS: Significant association of MTHFR polymorphisms (including MTHFR C677T and MTHFR A1298C) and RIF could not be confirmed.


Subject(s)
Embryo Implantation , Fertilization in Vitro , Genetic Predisposition to Disease , Homocystinuria/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Muscle Spasticity/genetics , Female , Humans , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Genetic , Pregnancy , Psychotic Disorders/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...