Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Eur J Clin Nutr ; 61(6): 769-78, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17151589

ABSTRACT

OBJECTIVE: To determine the effect of folic acid, vitamin B(6) and B(12) fortified spreads on the blood concentrations of these vitamins and homocysteine. DESIGN AND SETTING: A 6-week randomized, double-blinded, placebo-controlled, parallel trial carried out in a clinical research center. SUBJECTS: One hundred and fifty healthy volunteers (50% males). INTERVENTIONS: For 6 weeks, the subjects consumed the test spreads (20 g/day): containing per 20 g (1) 200 microg folic acid, 2 microg vitamin B(12) and 1 mg vitamin B(6), or (2) 400 microg folic acid, 2 microg vitamin B(12) and 1 mg vitamin B(6) or (3) no B-vitamins (control spread). RESULTS: The B-vitamin status increased on using the test spreads, with the largest effect on the serum folate concentration: 48% in men and 58% in women on spread 1 and 92 and 146%, respectively, on spread 2 (P-values all <0.05). The plasma homocysteine decreased in the groups treated with the fortified spreads as compared to the control group. Average decreases were for males: 0.7+/-1.5 micromol/l (6.8%) on spread 1 and 1.7+/-1.7 micromol/l (17.6%) on spread 2 and for females: 1.4+/-1.2 micromol/l (14.2%) and 2.4+/-2.0 micromol/l (23.3%), respectively (P-values all <0.05). CONCLUSIONS: Consumption of a spread fortified with folic acid, vitamin B(6) and vitamin B(12) for 6 weeks significantly increases the blood concentrations of these vitamins and significantly decreases the plasma concentration of homocysteine. Fortified staple foods like spreads can contribute to the lowering of homocysteine concentrations.


Subject(s)
Food, Fortified , Homocysteine/blood , Hyperhomocysteinemia/diet therapy , Vitamin B Complex/administration & dosage , Vitamin B Complex/blood , Adolescent , Adult , Aged , Dose-Response Relationship, Drug , Double-Blind Method , Female , Folic Acid/administration & dosage , Folic Acid/blood , Humans , Male , Middle Aged , Sex Factors , Vitamin B 12/administration & dosage , Vitamin B 12/blood , Vitamin B 6/administration & dosage , Vitamin B 6/blood
2.
Eur J Clin Nutr ; 59(4): 480-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15674310

ABSTRACT

BACKGROUND: Hyperhomocysteinemia (HHCY) is a risk factor for cardiovascular diseases (CVD). HHCY may interact with hypertension (HTEN) and an unfavorable cholesterol profile (UNFAVCHOL) to alter the risk of CVD. OBJECTIVES: To estimate the prevalences of HHCY (1) isolated and (2) in combination with UNFAVCHOL and/or HTEN in different age categories. To provide information that may improve the screening and treatment of subjects at risk of CVD. DESIGN: Cross-sectional data on 12,541 men and 12,948 women aged 20 + y were used from nine European studies. RESULTS: The prevalence of isolated HHCY was 8.5% in subjects aged 20-40 y, 4.7% in subjects aged 40-60 y and 5.9% in subjects aged over 60 y. When combining all age groups, 5.3% had isolated HHCY and an additional 5.6% had HHCY in combination with HTEN and/or UNFAVCHOL. The combinations of risk factors increased with age and, except for HHCY&UNFAVCHOL, were more prevalent than predicted by chance. Of the young subjects (20-40 y), 24% suffered from one or more of the investigated CVD risk factors. This figure was 75.1% in the old subjects (60+ years). CONCLUSIONS: A substantial number of subjects in selected European populations have HHCY (10.9%). In half of these cases, subjects suffer also from other CVD risk factors like UNFAVCHOL and HTEN. Older people in particular tend to have more than one risk factor. Healthcare professionals should be aware of this when screening and treating older people not only for the conventional CVD risk factors like UNFAVCHOL and HTEN but also HHCY, as this can easily be reduced through increased intake of folic acid via supplement or foods fortified with folic acid.


Subject(s)
Cardiovascular Diseases/blood , Hypercholesterolemia/epidemiology , Hyperhomocysteinemia/epidemiology , Hypertension/epidemiology , Adult , Age Factors , Blood Pressure/physiology , Cholesterol/blood , Cross-Sectional Studies , Europe/epidemiology , Female , Homocysteine/blood , Humans , Hypercholesterolemia/blood , Hyperhomocysteinemia/blood , Male , Middle Aged , Prevalence , Risk Factors , Sex Factors
3.
Birth Defects Res A Clin Mol Teratol ; 67(6): 421-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12962286

ABSTRACT

BACKGROUND: Periconceptional folic acid supplementation reduces the occurrence and recurrence risk of neural tube defects (NTD). Mothers of children with NTD have elevated plasma homocysteine levels. Administering homocysteine to chick embryos is reported to cause 27% NTD. Therefore, elevated plasma homocysteine levels per se or a disturbed homocysteine metabolism may be teratogenic to the embryo and may interfere with neural tube closure. Our aim was to obtain a chick embryo model to explore the interference of homocysteine in neural tube closure. METHODS: Homocysteine or saline was administered to chick embryos in ovo at 3 hr, 30 hr, and 60 hr of incubation and harvested at 74 hr. Homocysteine was then applied to chick embryos in vitro at a defined time window of four to six somites and followed for 6 hr. RESULTS: Homocysteine administration to chick embryos in ovo resulted in several malformations but not in an increased number of NTDs. Homocysteine administration to chick embryos in vitro resulted in a transient, dose-dependent widening of the anterior neuropore and closure delay of the rhombencephalic neuropore. After 16 hr of incubation the neural tube was closed. CONCLUSIONS: The in vitro chick embryo model appears a good model to explore the interference of a disturbed homocysteine metabolism in neurulation.


Subject(s)
Disease Models, Animal , Embryo, Nonmammalian/drug effects , Homocysteine/toxicity , Neural Tube Defects/chemically induced , Teratogens/toxicity , Abnormalities, Multiple/chemically induced , Abnormalities, Multiple/pathology , Animals , Chick Embryo , Dose-Response Relationship, Drug , Embryo, Nonmammalian/embryology , Neural Tube Defects/pathology , Organ Culture Techniques
4.
Eur J Clin Nutr ; 57(10): 1235-41, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14506483

ABSTRACT

OBJECTIVE: To explore the feasibility of low-fat spreads as vehicles for folic acid (FA) fortification by determining the acute absorption of FA from a fortified spread. DESIGN: Double blind, crossover study to test each of the following treatments administered at 1-weekly intervals: (A) 20 g low-fat (40%) spread fortified with 200 microg FA and a placebo tablet; (B) 20 g low-fat placebo spread and a 200 microg FA tablet; (C) 20 g low-fat placebo spread and a placebo tablet. SUBJECTS: A total of 13 male volunteers, aged 31.8+/-13.2 y. MAIN OUTCOME MEASURES: Plasma total folate concentrations, measured before and up to 10 h after each treatment (n=10 samples per treatment). RESULTS: Plasma folate concentrations were significantly increased compared with baseline values 1 h after administration of the FA tablet, and 1.5 h after the FA spread, and remained significantly higher than the baseline values for up to 7 h after both treatments. The maximum plasma folate response (R(max)), corrected for baseline values and 'placebo response', was established between 1 and 3 h postprandially in response to both FA spread and FA tablet, and no significant difference in R(max) was found between the two treatments (13.4 vs 14.4 nmol/l, P=0.9). The acute absorption of FA from fortified spread relative to that from the tablet, calculated on the basis of area under the plasma folate response curve, was 67% (P=0.04). CONCLUSION: The absorption of FA from fortified low-fat spread, although lower than from a tablet, is effective. These results suggest that low-fat spreads, typically associated with fat-soluble vitamin fortification, may also be considered feasible as vehicles for FA fortification.


Subject(s)
Dietary Supplements , Folic Acid/blood , Folic Acid/pharmacokinetics , Food, Fortified , Adult , Area Under Curve , Cross-Over Studies , Diet, Fat-Restricted , Double-Blind Method , Folic Acid/administration & dosage , Humans , Intestinal Absorption , Male , Tablets
5.
J Mol Med (Berl) ; 79(9): 522-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11692165

ABSTRACT

Molecular defects in genes encoding enzymes involved in homocysteine metabolism may account for mild hyperhomocysteinemia, an independent and graded risk factor for cardiovascular disease (CVD). We examined the relationship of two polymorphisms in the methylenetetrahydrofolate reductase (MTHFR) gene, the 677C-->T and 1298A-->C variants, to MTHFR activity, homocysteine concentrations, and risk of CVD in a population of 190 vascular disease patients and 601 apparently healthy controls. The mean specific and residual MTHFR activities were significantly lower in 677CT and 677TT individuals (both P<0.001). The 1298A-->C mutation alone showed no effect on MTHFR activities. However, when the 677C-->T genotype was taken into account, the 1298A-->C mutation also caused a significant decrease in MTHFR activities, which was observed in both the homozygous 1298CC (P<0.001) and the heterozygous 1298AC states (P=0.005). Both the 677TT as the 677CT genotypes were associated with significantly higher fasting and postload homocysteine levels than 677CC (P<0.001 and P=0.003, respectively). The 1298A-->C mutation had no effect on fasting or postload homocysteine levels. Since homocysteine itself is considered to be positively associated with the risk of CVD, these findings indicate that the 1298A-->C mutation cannot be considered a major risk factor for CVD.


Subject(s)
Cardiovascular Diseases/genetics , Genetic Predisposition to Disease , Homocysteine/metabolism , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Polymorphism, Genetic , Adult , Fasting , Female , Genotype , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Mutation , Risk Factors
6.
Nephron ; 89(1): 50-5, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11528232

ABSTRACT

Infantile nephropathic cystinosis, an inborn error of metabolism with an autosomal recessive inheritance pattern, is characterized by lysosomal storage of the amino acid cystine due to an impaired transport of cystine out of the lysosomes. Initial clinical features consist of the renal Fanconi syndrome and crystals in the cornea. Oral therapy with cysteamine lowers the intracellular cystine content. Recently, the gene coding for the integral membrane protein cystinosin, which is responsible for membrane transport of cystine (CTNS), was cloned. Mutation analysis of the CTNS gene of Caucasian patients revealed a common 57-kb deletion, and several other mutations spread throughout the entire gene. In the present study, we developed an improved screening method for the detection of the common 57-kb deletion. By use of this method we detected the 57-kb deletion in 59% of the examined Dutch alleles. The remaining alleles were screened for other mutations by genomic sequencing of the different exons, revealing three previously described mutations. Furthermore, we studied a possible genotype-phenotype relation of the homozygous deleted patients, which could not be demonstrated in our study population. Next to biochemical determination of cystine in leukocytes or fibroblasts, molecular genetic analysis enables prenatal diagnosis and facilitates identification of carriers.


Subject(s)
Cystinosis/genetics , Gene Deletion , Genetic Testing/methods , Glycoproteins , Membrane Proteins/genetics , Amino Acid Transport Systems, Neutral , Cystine/genetics , DNA Mutational Analysis , DNA Primers , Exons , Genotype , Humans , Introns , Membrane Transport Proteins , Netherlands , Phenotype
7.
Mol Genet Metab ; 73(2): 164-72, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11386852

ABSTRACT

Neural tube defects (NTD) arise in the first weeks of pregnancy due to a combination of environmental and genetic factors. In mothers of children with NTD elevated homocysteine (Hcy) levels and decreased plasma folate levels were observed, which suggests a defect in the folate-dependent Hcy metabolism. Therefore, mutations in genes coding for enzymes of this metabolism could be involved in NTD. Serine hydroxymethyltransferase (SHMT) catalyzes the reversible reaction of serine and tetrahydrofolate (THF) to glycine and 5,10-methylene THF. Two different isoforms of SHMT are known, one is present in the cytosol (cSHMT) and the other in the mitochondrion (mSHMT). Theoretically, mutated SHMT could lead to elevated Hcy levels and to an altered distribution of the different folate derivatives and might therefore become a risk factor for NTD. This study concerns the molecular genetic analysis of genes coding for both isoforms of the SHMT enzyme by single-stranded conformation polymorphism analysis. Several mutations as well as polymorphisms were found in both genes. The relevance of two variations, the 1420 C>T mutation of the cytosolic isoform and the 4-bp deletion of the mitochondrial isoform (delTCTT 1721-1724), to NTD risk was tested in a study group, which consisted of 109 NTD patients, 120 mothers of children with NTD, and 420 controls. Neither of the two polymorphisms led to an increased risk of NTD. In mothers with the 1420 CC genotype, significant increased Hcy levels are present. Also, significantly decreased red blood cell folate and plasma folate levels were present in individuals with the 1420 CC genotype. Probably, the 1420 C>T polymorphism causes a shift in distribution of the different folate derivatives. The 4-bp deletion of the mSHMT gene did not lead to altered Hcy or folate levels. So far, the results of this study provide no direct evidence for a role of defective SHMT functioning in NTD. Still, the influence of the 1420 C>T polymorphism of the cSHMT gene on the folate-related risk of NTD needs further investigation.


Subject(s)
Glycine Hydroxymethyltransferase/genetics , Neural Tube Defects/genetics , Adolescent , Adult , Alleles , Base Sequence , Child , Cytosol/enzymology , DNA Mutational Analysis , DNA, Complementary/chemistry , DNA, Complementary/genetics , Female , Folic Acid/blood , Genotype , Glycine Hydroxymethyltransferase/metabolism , Humans , Mitochondria/enzymology , Mutation , Neural Tube Defects/enzymology , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Sequence Deletion
8.
Exp Biol Med (Maywood) ; 226(4): 243-70, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11368417

ABSTRACT

Folate administration substantially reduces the risk on neural tube detects (NTD). The interest for studying a disturbed homocysteine (Hcy) metabolism in relation to NTD was raised by the observation of elevated blood Hcy levels in mothers of a NTD child. This observation resulted in the examination of enzymes involved in the folate-dependent Hcy metabolism. Thus far, this has led to the identification of the first and likely a second genetic risk factor for NTD. The C677T and A1298C mutations in the methylenetetrahydrofolate reductase (MTHFR) gene are associated with an increased risk of NTD and cause elevated Hcy concentrations. These levels can be normalized by additional folate intake. Thus, a dysfunctional MTHFR partly explains the observed elevated Hcy levels in women with NTD pregnancies and also, in part, the protective effect of folate on NTD. Although the MTHFR polymorphisms are only moderate risk factors, population-wide they may account for an important part of the observed NTD prevalence.


Subject(s)
Folic Acid/metabolism , Homocysteine/metabolism , Neural Tube Defects/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Animals , Female , Folic Acid/blood , Folic Acid/chemistry , Folic Acid/therapeutic use , Homocysteine/blood , Humans , Infant, Newborn , Intestinal Absorption , Methylenetetrahydrofolate Reductase (NADPH2) , Models, Chemical , Neural Tube Defects/embryology , Neural Tube Defects/prevention & control , Oxidoreductases Acting on CH-NH Group Donors/deficiency , Pregnancy , Prevalence
9.
QJM ; 94(3): 159-66, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11259691

ABSTRACT

Periconceptional folic acid supplementation reduces the risk of neural tube defects (NTD). Homocysteine levels are elevated in mothers of NTD children, which may be due to decreased cellular vitamin B12 levels, as vitamin B12 is a cofactor for the methylation of homocysteine. Transcobalamin II (TC II) transports vitamin B12 to the tissues. To examine whether altered plasma transcobalamin levels are a risk factor for NTD, we determined the apo and holo form of TC II and haptocorrin (TCI+TCIII), vitamin B12 and homocysteine concentrations in the plasma of 46 mothers with NTD children, and in 73 female controls. Holo-tc II levels and holo-tc II percentages (holo-tc II/total tc II) in the first quartile of the control distribution were related to a three-fold (OR 2.9, 95%CI 0.9-9.2) and five-fold (OR 5.0, 95%CI 1.3-19.3) risk, respectively, for having a child with NTD, when compared with the last quartile. Homocysteine levels were significantly higher among individuals with low holo-tc II, low total vitamin B12 concentrations and low holo-tc II percentages. These low holo-tc II percentages are probably caused by reduced affinity of TC II for vitamin B12, which may be explained by genetic variation in the TC II gene. Vitamin B12 supplementation might therefore be warranted, in addition to folate, in the prevention of NTD.


Subject(s)
Neural Tube Defects/etiology , Transcobalamins/metabolism , Vitamin B 12/metabolism , Adult , Case-Control Studies , Chromatography, High Pressure Liquid , Electrophoresis, Agar Gel , Female , Homocysteine/blood , Humans , Middle Aged , Neural Tube Defects/metabolism , Polymerase Chain Reaction , Polymorphism, Genetic , Pregnancy , Risk Factors , Statistics as Topic
10.
Clin Endocrinol (Oxf) ; 54(2): 197-204, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11207634

ABSTRACT

OBJECTIVE: Hyperhomocysteinaemia is a risk factor for premature atherosclerotic vascular disease and venous thrombosis. The aim of the present study was to assess plasma total homocysteine (tHCys) concentrations in hypo- as well as hyperthyroid patients before and after treatment, and to evaluate the role of potential determinants of plasma tHCys levels in these patients. DESIGN: Prospective follow up study. PATIENTS: Fifty hypothyroid and 46 hyperthyroid patients were studied in the untreated state and again after restoration of euthyroidism. MEASUREMENTS: Fasting plasma levels of tHCys and its putative determinants (plasma levels of free thyroxine (fT4), folate, vitamin B(12), renal function, sex, age, smoking status and the C677T polymorphism in the methylenetetrahydrofolate reductase (MTHFR) gene were measured before and after treatment. RESULTS: Restoration of the euthyroid state decreased both tHCys (17.6 +/- 10.2-13.0 +/- 4.7 micromol/l; P < 0.005) and creatinine (83.9 +/- 22.0-69.8 +/- 14.2 micromol/l; P < 0.005) in hypothyroid patients and increased both tHCys (10.7 +/- 2.5-13.4 +/- 3.3 micromol/l; P < 0.005) and creatinine (49.0 +/- 15.4-66.5 +/- 15.0 micromol/l; P < 0.005) in hyperthyroid patients (values as mean +/- SD). Folate levels were lower in the hypothyroid group compared to the hyperthyroid group (11.7 +/- 6.4 and 15.1 +/- 7.6 nmol/l; P < 0.05). Pretreatment tHCys levels correlated with log fT(4) (r = - 0.47), folate (r = - 0.21), plasma creatinine (r = 0.45) and age (r = 0.35) but not with C677T genotype. Multivariate analysis indicated that pretreatment log(fT(4)) levels and age accounted for 28% the variability of pre-treatment tHCys (tHCys = 14.2-5.50 log(fT(4)) + 0.14 age). After treatment the logarithm of the change (Delta) in fT(4) (expressed as the post-treatment fT(4)/pre-treatment fT(4) ratio) accounted for 45% of the variability in change of tHCys ( tHCys = - 0.07-4.94 log ( fT(4))); there was no independent contribution of changes in creatinine which was, however, strongly related to changes in tHCys (r = 0.61). CONCLUSIONS: Plasma tHCys concentrations increased in hypothyroidism and decreased in hyperthyroidism. Plasma fT(4) is an independent determinant of tHCys concentrations. Lower folate levels and a lower creatinine clearance in hypo-thyroidism, and a higher creatinine clearance in hyperthyroidism only partially explain the changes in tHCys.


Subject(s)
Cardiovascular Diseases/etiology , Homocysteine/blood , Hyperthyroidism/blood , Hypothyroidism/blood , Adult , Age Factors , Aged , Analysis of Variance , Biomarkers/blood , Creatinine/metabolism , Female , Folic Acid/blood , Follow-Up Studies , Humans , Hyperthyroidism/physiopathology , Hypothyroidism/physiopathology , Linear Models , Male , Metabolic Clearance Rate , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Middle Aged , Polymorphism, Genetic , Prospective Studies , Risk Factors , Sex Factors , Smoking , Thyroid Function Tests , Thyroxine/blood , Vitamin B 12/blood
11.
Eur J Obstet Gynecol Reprod Biol ; 92(1): 57-61, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10986435

ABSTRACT

Folate administration substantially reduces the risk on neural tube defects (NTD). The interest for a disturbed homocysteine (Hcy) metabolism in relation to NTD was raised by the observation of elevated blood Hcy levels in mothers of a NTD child. This observation resulted in the examination of enzymes involved in the folate dependent Hcy metabolism. This leads to the identification of the first and likely a second genetic risk factor for NTD. The C677T and A1298C mutations in the methylenetetrahydrofolate reductase (MTHFR) gene are associated with an increased risk of NTD and cause elevated Hcy concentrations. These levels can be normalized by an additional folate intake. Thus, a dysfunctional MTHFR partly explains the observed elevated Hcy levels in women with NTD pregnancies, and also in part the protective effect of folate on NTD. Although, the MTHFR polymorphisms are only moderate risk factors, population wide they may account for an important part of the observed NTD prevalence.


Subject(s)
Folic Acid/metabolism , Homocysteine/metabolism , Neural Tube Defects/etiology , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Cystathionine beta-Synthase/metabolism , Female , Folic Acid/pharmacology , Humans , Methylenetetrahydrofolate Reductase (NADPH2) , Neural Tube Defects/prevention & control , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Polymorphism, Genetic
12.
Neuropediatrics ; 31(6): 298-302, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11508548

ABSTRACT

Folic acid supplementation around conception decreases the risk of having offspring with a neural tube defect. However, the aetiology is often still unknown. This study investigated whether spina bifida patients have lower blood folate and higher fasting and post-methionine-load plasma total homocysteine (tHcy) concentrations than control patients. Moreover, the effects of supplementation with 500 microg folic acid/d on folate and tHcy concentrations were determined. Spina bifida patients (n = 12) and disabled control patients (n = 15) received 4 weeks of placebo treatment followed by 4 weeks of intervention with 500 microg folic acid/d. Blood was collected at the start and after 4 and 8 weeks. A methionine-loading test was performed at the start and the end of the study. At baseline, no significant differences occurred between spina bifida and control patients. Folic acid supplementation significantly increased plasma and red blood cell folate concentrations in both groups. Folic acid decreased fasting tHcy concentrations in control patients by 1.6+/-0.5 micromol/l (p<0.01) and in spina bifida patients by 2.2 +/- 1.3 micromol/l (p = 0.10). This study does not show a derangement in homocysteine metabolism in spina bifida compared to control patients. Moreover, folic acid supplementation seems at least as effective in spina bifida patients as in controls.


Subject(s)
Folic Acid/pharmacology , Hematinics/pharmacology , Homocysteine/metabolism , Spinal Dysraphism/drug therapy , Administration, Oral , Adolescent , Adult , Case-Control Studies , Female , Folic Acid/administration & dosage , Folic Acid/blood , Hematinics/administration & dosage , Hematinics/blood , Homocysteine/blood , Homocysteine/pharmacology , Humans , Male , Middle Aged , Spinal Dysraphism/pathology , Treatment Outcome
13.
Eur J Hum Genet ; 7(3): 393-6, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10234517

ABSTRACT

Neural tube defects (NTDs) are the most common congenital malformations and are considered to have a multifactorial origin, having both genetic and environmental components. Periconceptional folate administration reduces the recurrence and occurrence risk by 70-100%. Recently we discovered the first genetic risk factors for NTDs: the 677 C-->T and the 1298 A-->C mutations in the methylenetetrahydrofolate reductase gene explaining at the most 35-50% of the protective effect of folate. In this study we further explored the genetic component of NTDs by analysing the coding region, including the intron-exon boundaries and signal sequences of the folate receptor genes by SSCP analysis. Among 39 patients with spina bifida (SB), 47 mothers with a child with SB, and 10 controls, no polymorphism was present in the folate receptor alpha (FR-alpha) gene or in the folate receptor beta (FR-beta) gene.


Subject(s)
Carrier Proteins/genetics , Neural Tube Defects/genetics , Receptors, Cell Surface , Adult , Female , Folate Receptors, GPI-Anchored , Humans , Polymorphism, Single-Stranded Conformational
14.
Clin Genet ; 53(2): 119-25, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9611072

ABSTRACT

It is now well recognized that periconceptional folic acid or folic acid containing multivitamin supplementation reduces the risk of neural tube defects (NTDs). Recently we were able to show that homozygosity for a thermolabile variant of the enzyme methylenetetrahydrofolate reductase is associated with an increased risk for spina bifida in patients recruited from the Dutch population. However, this genetic risk factor could not account for all folic acid preventable NTDs. In an attempt to identify additional folate related enzymes that contribute to NTD etiology we now studied the methylenetetrahydrofolate dehydrogenase gene on chromosome 14q24 which encodes a single protein with three catalytic properties important in the folate metabolism. The cDNA sequence of 38 familial and 79 sporadic patients was screened for the presence of mutations by single strand conformation polymorphism (SSCP) analysis followed by sequencing. Two amino acid substitutions were identified. The first one (R293H) was detected in a patient with familial spina bifida and not in 300 control individuals. The mutation was inherited from the unaffected maternal grandmother and was also present in two younger brothers of the index patient, one of them displaying spina bifida occulta and the other being unaffected. The second change turned out to be an amino acid polymorphism (R653Q) that was present in both patients and controls with similar frequencies. Our results so far provide no evidence for a major role of the methylenetetrahydrofolate-dehydrogenase (MTHFD) gene in NTD etiology. However, the identification of a mutation in one family suggests that this gene can act as a risk factor for human NTD.


Subject(s)
Aminohydrolases/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Neural Tube Defects/enzymology , Neural Tube Defects/genetics , Base Sequence , DNA, Complementary , Female , Humans , Male , Methenyltetrahydrofolate Cyclohydrolase , Molecular Sequence Data , Pedigree , Polymorphism, Single-Stranded Conformational
15.
Am J Hum Genet ; 62(5): 1044-51, 1998 May.
Article in English | MEDLINE | ID: mdl-9545395

ABSTRACT

Recently, we showed that homozygosity for the common 677(C-->T) mutation in the methylenetetrahydrofolate reductase (MTHFR) gene, causing thermolability of the enzyme, is a risk factor for neural-tube defects (NTDs). We now report on another mutation in the same gene, the 1298(A-->C) mutation, which changes a glutamate into an alanine residue. This mutation destroys an MboII recognition site and has an allele frequency of .33. This 1298(A-->C) mutation results in decreased MTHFR activity (one-way analysis of variance [ANOVA] P < .0001), which is more pronounced in the homozygous than heterozygous state. Neither the homozygous nor the heterozygous state is associated with higher plasma homocysteine (Hcy) or a lower plasma folate concentration-phenomena that are evident with homozygosity for the 677(C-->T) mutation. However, there appears to be an interaction between these two common mutations. When compared with heterozygosity for either the 677(C-->T) or 1298(A-->C) mutations, the combined heterozygosity for the 1298(A-->C) and 677(C-->T) mutations was associated with reduced MTHFR specific activity (ANOVA P < .0001), higher Hcy, and decreased plasma folate levels (ANOVA P <.03). Thus, combined heterozygosity for both MTHFR mutations results in similar features as observed in homozygotes for the 677(C-->T) mutation. This combined heterozygosity was observed in 28% (n =86) of the NTD patients compared with 20% (n =403) among controls, resulting in an odds ratio of 2.04 (95% confidence interval: .9-4.7). These data suggest that the combined heterozygosity for the two MTHFR common mutations accounts for a proportion of folate-related NTDs, which is not explained by homozygosity for the 677(C-->T) mutation, and can be an additional genetic risk factor for NTDs.


Subject(s)
Adenine , Cytosine , Neural Tube Defects/enzymology , Neural Tube Defects/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Point Mutation , Adult , Female , Homocysteine/blood , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Prevalence , Pyridoxine/blood , Vitamin B 12/blood
16.
Arterioscler Thromb Vasc Biol ; 18(3): 356-61, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9514403

ABSTRACT

Hyperhomocysteinemia is a risk factor for atherosclerosis and thrombosis and is inversely related to plasma folate and vitamin B12 levels. We assessed the effects of vitamin supplementation on plasma homocysteine levels in 89 patients with a history of recurrent venous thrombosis and 227 healthy volunteers. Patients and hyperhomocysteinemic (homocysteine level >16 micromol/L) volunteers were randomized to placebo or high-dose multivitamin supplements containing 5 mg folic acid, 0.4 mg hydroxycobalamin, and 50 mg pyridoxine. A subgroup of volunteers without hyperhomocysteinemia was also randomized into three additional regimens of 5 mg folic acid, 0.5 mg folic acid, or 0.4 mg hydroxycobalamin. Before and after the intervention period, blood samples were taken for measurements of homocysteine, folate, cobalamin, and pyridoxal-5'-phosphate levels. Supplementation with high-dose multivitamin preparations normalized plasma homocysteine levels (< or = 16 micromol/L) in 26 of 30 individuals compared with 7 of 30 in the placebo group. Also in normohomocysteinemic subjects, multivitamin supplementation strongly reduced homocysteine levels (median reduction, 30%; range, -22% to 55%). In this subgroup the effect of folic acid alone was similar to that of multivitamin: median reduction, 26%; range, -2% to 52% for 5 mg folic acid and 25%; range, -54% to 40% for 0.5 mg folic acid. Cobalamin supplementation had only a slight effect on homocysteine lowering (median reduction, 10%; range, -21% to 41%). Our study shows that combined vitamin supplementation reduces homocysteine levels effectively in patients with venous thrombosis and in healthy volunteers, either with or without hyperhomocysteinemia. Even supplementation with 0.5 mg of folic acid led to a substantial reduction of blood homocysteine levels.


Subject(s)
Folic Acid/therapeutic use , Homocysteine/blood , Hydroxocobalamin/therapeutic use , Pyridoxine/therapeutic use , Thrombophlebitis/blood , Thrombophlebitis/drug therapy , Adult , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Female , Folic Acid/administration & dosage , Humans , Hydroxocobalamin/administration & dosage , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Mutation/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Pyridoxine/administration & dosage , Reference Values , Thrombophlebitis/genetics
17.
QJM ; 90(8): 505-10, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9327028

ABSTRACT

Folic acid intake reduces the risk of neural tube defects (NTDs). Although the 677C-->T mutation in the 5,10-methylenetetrahydrofolate reductase (MTHFR) gene is a risk factor for NTDs, it only partly explains the elevated homocysteine levels in mothers of children with NTDs. We measured vitamin B12, folate and homocysteine in patients with spina bifida (SB), their parents, and in controls, to investigate which other enzymes of homocysteine metabolism might be defective. Because homozygosity for the 677C-->T mutation causes decreased plasma folate and increased red-cell folate (RCF) and plasma homocysteine levels, we excluded individuals homozygous for that mutation. The remaining SB patients and their parents still had lowered plasma folate and elevated total homocysteine levels, and a small subset had decreased vitamin B12 levels. Red-cell folate was the same in all groups, suggesting that dietary folate intake and its uptake was normal. Risk of SB was increased at the 25th percentile of plasma folate and at the 75th percentile of homocysteine values in SB patients and their parents, and at the 5th and 25th percentiles of vitamin B12 in mothers with SB-affected offspring. This underlines the functional importance of homocysteine remethylation to methionine. There was no correlation between vitamin B12 and homocysteine or RCF. In combination with the lowered plasma folate (80-90% 5-methyltetrahydrofolate), our data do not support a major involvement of methionine synthase in the aetiology of SB. Our data rather favour the involvement of genetic variation at loci coding for the formation of 5-methyltetrahydrofolate, such as MTHFR, methylenetetrahydrofolate dehydrogenase or serine hydroxymethyltransferase.


Subject(s)
Folic Acid/metabolism , Spinal Dysraphism/metabolism , Vitamin B 12/metabolism , Adolescent , Adult , Erythrocytes/metabolism , Female , Homocysteine/metabolism , Humans , Male , Middle Aged , Risk , Spinal Dysraphism/blood , Spinal Dysraphism/genetics , Statistics, Nonparametric
18.
QJM ; 90(8): 511-7, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9327029

ABSTRACT

Elevated homocysteine (Hcy) levels are observed in two apparently unrelated diseases: neural-tube defects (NTD) and premature vascular disease. Defective human methionine synthase (MS) could result in elevated Hcy levels. We sequenced the coding region of MS in 8 hyperhomocysteinaemic patients (4 NTD patients and 4 patients with pregnancies complicated by spiral arterial disease, SAD). We identified only one mutation resulting in an amino acid substitution: an A-->G transition at bp 2756, converting an aspartic acid (D919) into a glycine (G). We screened genomic DNA for the presence of this mutation in 56 NTD patients, 69 mothers of children with NTD, 108 SAD patients and 364 controls. There was no increased prevalence of the GG and AG genotypes in NTD patients, their mothers or SAD patients. The D919G mutation does not seem to be a risk factor for NTD or vascular disease. We then examined the mean Hcy levels for each MS genotype. There was no correlation between GG- or AG-genotype and Hcy levels. The D919G mutation is thus a fairly prevalent, and probably benign polymorphism. This study, though limited, provides no evidence for a major involvement of MS in the aetiology of homocysteine-related diseases such as NTD or vascular disease.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , Arterial Occlusive Diseases/enzymology , Neural Tube Defects/enzymology , Pregnancy Complications, Cardiovascular/enzymology , Adolescent , Adult , Arterial Occlusive Diseases/blood , Female , Genotype , Homocysteine/blood , Humans , Male , Molecular Sequence Data , Mutation , Neural Tube Defects/blood , Odds Ratio , Polymerase Chain Reaction , Pregnancy , Pregnancy Complications, Cardiovascular/blood , Sequence Analysis, DNA
19.
QJM ; 90(2): 111-5, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9068801

ABSTRACT

The common 677C-->T mutation (+) in the 5,10-methylenetetrahydrofolate reductase gene, resulting in decreased activity of the enzyme, has been associated with spina bifida neural tube defects (NTD). We combined all known Dutch control groups, a total of 1273 individuals, and found a prevalence of the 677C-->T mutation of 8.4%. When compared with the frequencies in 55 SB patients and to mothers with a child with SB their parents, this gave an OR of 1.9 [95% CI 1.1-3.3] for mothers and an OR of 1.5 [95% CI 0.74-3.1] for patients. The frequency of this mutation and its associated risk for NTD may be population-dependent. However, the frequencies of the 677C-->T mutation in different national and international control groups are almost all in the same range. We therefore combined the observed frequencies of the 677C-->T mutation in all reported studies. The mutation was present in 9.2% of controls, resulting in ORs for all reported NTD patients and their parents of: 1.7 [95% CI: 1.1-2.6]; 1.8 [95% CI: 1.1-3.1] and 1.9 [95% CI: 1.3-2.8] for mothers (combined prevalence 14.5%), fathers (combined prevalence 15.5%) and NTD patients (combined prevalence 16.4%), respectively, vs. all international controls. This meta-analysis confirms that the 677C-->T mutation is a genetic risk factor for NTD.


Subject(s)
Neural Tube Defects/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Female , Genetic Markers , Homozygote , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Mutation , Netherlands , Odds Ratio , Prevalence
20.
J Mol Med (Berl) ; 74(11): 691-4, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8956155

ABSTRACT

Periconceptional folate intake reduces both the occurrence and recurrence risk of neural tube defects. Plasma homocysteine levels can be elevated in mothers of a child with a neural tube defect, suggesting a dysfunctional folate metabolism. Very recently we showed that a common 677C-->T mutation in the 5,10-methylene tetrahydrofolate reductase gene, causing thermolability of the enzyme, is a risk factor for spina bifida offspring. Restriction enzyme analysis of the genomic 5,10-methylene tetrahydrofolate reductase polymerase chain reaction fragment revealed a significantly higher prevalence of a +/+ genotype among spina bifida patients and their mothers. The risk for spina bifida offspring is the strongest if both the mother and her child have the mutation in the homozygous state. Enzymatic analysis showed that homozygosity for the 677C-->T mutation causes a decreased 5,10-methylene tetrahydrofolate reductase activity, resulting in elevated plasma homocysteine and red blood cell folate levels and lowered plasma folate and cysteine values. This extended study demonstrates that a nucleotide substitution in the coding region of 5,10-methylene tetrahydrofolate reductase, resulting in reduced activity and an impaired homocysteine and folate metabolism, is a genetic risk factor for spina bifida.


Subject(s)
Oxidoreductases/genetics , Point Mutation/genetics , Spinal Dysraphism/metabolism , 5,10-Methylenetetrahydrofolate Reductase (FADH2) , Adult , Aged , Child , Cysteine/blood , Cysteine/metabolism , Female , Folic Acid/blood , Genetic Linkage , Genotype , Homocysteine/blood , Homocysteine/metabolism , Homozygote , Humans , Lod Score , Lymphocytes/enzymology , Lymphocytes/metabolism , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Netherlands , Oxidoreductases/deficiency , Risk Factors , Spinal Dysraphism/epidemiology , Vitamin B 12/blood , Vitamin B 12/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...