Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
BMC Cancer ; 20(1): 940, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-32998716

ABSTRACT

BACKGROUND: After High-Dose Methotrexate (HD-MTX), folinic acid rescue therapy (Leucovorin) is administered to reduce side effects in pediatric acute lymphoblastic leukemia (ALL) patients. Leucovorin and MTX are structural analogues, possibly competing for cellular transport and intracellular metabolism. We hypothesize that Leucovorin accumulates during consecutive courses, which might result in a lower MTX uptake. METHODS: We prospectively measured red blood cell (RBC) folate and MTX levels during four HD-MTX and Leucovorin courses in 43 patients treated according the DCOG ALL-11 protocol with 2-weekly HD-MTX (5 g/m2/dose) and Leucovorin (15 mg/m2/dose) using LC-MS/MS. We estimated a linear mixed model to assess the relationship between these variables over time. RESULTS: Both RBC MTX-PG and folate levels increased significantly during protocol M. MTX-PG2-5 levels increased most substantially after the first two HD-MTX courses (until median 113.0 nmol/L, IQR 76.8-165.2) after which levels plateaued during the 3d and 4th course (until median 141.3 nmol/L, IQR 100.2-190.2). In parallel, folate levels increased most substantially after the first two HD-MTX courses (until median 401.6 nmol/L, IQR 163.3-594.2) after which levels plateaued during the 3d and 4th course (until median 411.5 nmol/L, IQR 240.3-665.6). The ratio folate/MTX-PG decreased significantly over time, which was mostly due to the relatively higher increase (delta) of MTX-PG. CONCLUSION: These results suggest that the increase in RBC folate levels does not seem to have a large effect on RBC MTX levels. Future studies, assessing competition of Leucovorin and MTX on other cellular mechanisms which might negatively affect treatment efficacy, are necessary.


Subject(s)
Folic Acid/blood , Methotrexate/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/blood , Child , Child, Preschool , Chromatography, Liquid , Erythrocytes/drug effects , Female , Humans , Infant , Leucovorin/administration & dosage , Leucovorin/blood , Male , Methotrexate/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tandem Mass Spectrometry , Treatment Outcome
3.
PLoS One ; 15(5): e0231588, 2020.
Article in English | MEDLINE | ID: mdl-32421698

ABSTRACT

We have recently established a protocol to grow wildtype human oral mucosa organoids. These three-dimensional structures can be maintained in culture long-term, do not require immortalization, and recapitulate the multilayered composition of the epithelial lining of the oral mucosa. Here, we validate the use of this model to study the effect of Leucovorin (LV) on Methotrexate (MTX)-induced toxicity. MTX is a chemotherapeutic agent used in the treatment of pediatric acute lymphoblastic leukemia. Although effective, the use of MTX often results in severe side-effects, including oral mucositis, which is characterized by epithelial cell death. Here, we show that organoids are sensitive to MTX, and that the addition of LV reduces MTX toxicity, in both a concentration- and timing-dependent manner. Additionally, we show that a 24 hour 'pretreatment' with LV reduces MTX-induced cell death, suggesting that such a pretreatment could decrease mucositis in patients. Taken together, we provide the first in vitro model to study the effect of MTX on wildtype oral mucosa cells. Our findings underscore the relevance of the clinically applied LV regimen and highlight the potential of this model to further optimize modifications in dosing and timing of Leucovorin on oral mucosa cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Apoptosis/drug effects , Mouth Mucosa/drug effects , Organoids/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Stomatitis/pathology , Adolescent , Child , Humans , In Vitro Techniques , Leucovorin/administration & dosage , Methotrexate/administration & dosage , Mouth Mucosa/pathology , Organ Culture Techniques , Organoids/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Stomatitis/chemically induced
4.
Crit Rev Oncol Hematol ; 142: 1-8, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31323533

ABSTRACT

INTRODUCTION: This study aimed to determine the efficacy of different Leucovorin regimens to reduce oral mucositis in children with acute lymphoblastic leukemia after high-dose Methotrexate (HD-MTX). METHODS: Twelve articles were included in a systematic literature review. Articles were categorized into low/medium/high risk of bias. RESULTS: As no randomized controlled trial assessing the effect of Leucovorin has been performed, the efficacy of Leucovorin to reduce oral mucositis remains unknown. Leucovorin was initiated at 24, 36 or 42 h after HD-MTX at a dose of 15 or 30 mg/m2. No meta-analysis could be performed as treatment regimens differed. When comparing studies with similar HD-MTX doses, we observed lower oral mucositis rates in regimens with higher cumulative doses of Leucovorin and early initiation of Leucovorin after MTX. CONCLUSION: Even though future studies are necessary, higher cumulative Leucovorin doses and early initiation of Leucovorin after start of MTX seem to reduce oral mucositis.


Subject(s)
Leucovorin/therapeutic use , Methotrexate/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Stomatitis/drug therapy , Adolescent , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , Humans , Infant , Leucovorin/administration & dosage , Methotrexate/therapeutic use , Stomatitis/chemically induced , Stomatitis/prevention & control , Treatment Outcome , Young Adult
5.
Support Care Cancer ; 27(1): 183-190, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29922939

ABSTRACT

PURPOSE: Children with acute lymphoblastic leukemia (ALL) are at increased risk of vitamin D deficiency, which might make them more susceptible to developing adverse events. Previous studies showed that low vitamin D levels were associated with an increased inflammatory mucosal state and impaired mucosal tissue barriers. We examined the prevalence of vitamin D deficiency and studied the association between vitamin D levels and methotrexate (MTX)-induced oral mucositis in pediatric ALL. METHODS: We assessed 25-hydroxyvitamin D (25(OH)D3) and 24,25-dihydroxyvitamin D (24,25(OH)2D3) levels in 99 children with ALL before the start of 4 × 5 g/m2 high-dose methotrexate (HD-MTX) (T0) and in 81/99 children after discontinuation of HD-MTX (T1). Two cutoff values for vitamin D deficiency exist: 25(OH)D3 levels < 30 and < 50 nmol/L. Oral mucositis was defined as grade ≥ 3 according to the National Cancer Institute Criteria. RESULTS: Vitamin D deficiency occurred in respectively 8% (< 30 nmol/L) and 33% (< 50 nmol/L) of the patients at T0, and more frequently in children > 4 years of age as compared to children between 1 and 4 years of age. A decrease in 25(OH)D3 levels during HD-MTX therapy was associated with developing severe oral mucositis (OR 1.6; 95% CI [1.1-2.4]). 25(OH)D3 and 24,25(OH)2D3 levels at T0 and the change in 24,25(OH)2D3 levels during therapy were not associated with the development of severe oral mucositis. CONCLUSIONS: This study showed that vitamin D deficiency occurs frequently in pediatric ALL patients above the age of 4 years. A decrease in 25(OH)D3 levels during MTX therapy was observed in children with ALL that developed severe oral mucositis.


Subject(s)
Methotrexate/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Stomatitis/chemically induced , Stomatitis/epidemiology , Vitamin D Deficiency/epidemiology , Vitamin D/blood , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Cohort Studies , Dose-Response Relationship, Drug , Female , Humans , Infant , Male , Methotrexate/administration & dosage , Netherlands/epidemiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Prevalence , Stomatitis/blood , Stomatitis/complications , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , Withholding Treatment , Young Adult
6.
Anal Bioanal Chem ; 405(5): 1673-81, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23239179

ABSTRACT

The folate antagonist methotrexate (MTX) is the anchor drug in the treatment of rheumatoid arthritis. The therapeutic effects of MTX are attributed to the intracellular levels of MTX, present in the cell as polyglutamates (MTXPGn). We developed a new liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI-MS/MS)-based assay to separately quantitate MTXPGn in red blood cells using stable-isotope-labelled internal standards. Samples were analyzed by LC-ESI-MS/MS using a Waters Acquity UPLC BEH C18 column with a 5-100% organic gradient of 10 mM ammonium bicarbonate (pH 10) and methanol. The analysis consisted of simple sample preparation and a 6-min run time. Detection was done using a Waters Acquity UPLC coupled to a Waters Quattro Premier XE with electrospray ionization operating in the positive ionization mode. Assay validation was performed following recent Food and Drug Administration guidelines. The method was linear from 1-1,000 nM for all MTXPGn (R(2) > 0.99). The coefficient of variation ranged from 1-4% for intraday precision and 6-15% for interday precision. Samples were stable for at least 1 month at -80 °C. Recovery ranged from 98-100%, and the relative matrix-effect varied from 95-99%. The lower limit of quantitation was 1 nM for each MTXPGn. Fifty patient samples from the tREACH study were analyzed. The MTXPGn concentration and distribution of these samples were comparable with values reported in literature. The developed LC-ESI-MS/MS method for the quantitative measurement of MTXPGn in red blood cells is both sensitive and precise within the clinically relevant range. The method can be easily applied in clinical laboratories due to the combination of simple pre-treatment with robust LC-ESI-MS/MS.


Subject(s)
Antirheumatic Agents/blood , Arthritis, Rheumatoid/drug therapy , Drug Monitoring/methods , Erythrocytes/chemistry , Methotrexate/blood , Polyglutamic Acid/blood , Spectrometry, Mass, Electrospray Ionization/methods , Antirheumatic Agents/analysis , Arthritis, Rheumatoid/blood , Chromatography, High Pressure Liquid/methods , Humans , Limit of Detection , Methotrexate/analysis , Polyglutamic Acid/analysis , Sensitivity and Specificity , Tandem Mass Spectrometry/methods
7.
Bone ; 50(6): 1401-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22465697

ABSTRACT

BACKGROUND: An elevated level of plasma homocysteine (Hcy) is a known risk factor for osteoporotic fractures. In addition, Hcy is related to DNA-methylation metabolism. To determine whether the association between Hcy and fractures is explained by an altered methylation capacity, we investigated the associations between levels of s-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) and fracture risk. METHODS: We studied 503 females aged 55 years and over from the Rotterdam Study (RS) in whom plasma Hcy, SAM and SAH levels were measured. Bone mineral density (BMD) at the hip was assessed using DXA. Incident fractures were recorded over a mean period of 7.0 years. Cox proportional hazards analysis and linear regression were used to assess relationships between plasma metabolite levels, incident osteoporotic fractures and BMD. RESULTS: Over a total of 3502 person-years of follow-up, 103 subjects sustained at least one osteoporotic fracture. Whereas incidence of osteoporotic fractures was associated with quartiles of Hcy (p=0.047), it was not associated with quartiles of SAM, SAH or SAM/SAH-ratio (all p for trend>0.6). Stepwise linear regression showed that SAM/SAH-ratio, but not Hcy, was independently associated with hip BMD (ß=0.073, p=0.025). CONCLUSION: Since SAM, SAH and SAM/SAH-ratio were not associated with osteoporotic fractures, alterations in methylation capacity most likely do not appear to be an important factor in the association between Hcy and fractures.


Subject(s)
Homocysteine/blood , Osteoporotic Fractures/blood , Osteoporotic Fractures/etiology , Aged , Bone Density , Female , Humans , Incidence , Linear Models , Methylation , Middle Aged , Netherlands/epidemiology , Osteoporotic Fractures/epidemiology , Proportional Hazards Models , Risk Factors , S-Adenosylhomocysteine/blood , S-Adenosylmethionine/blood
9.
J Thromb Haemost ; 2(5): 750-3, 2004 May.
Article in English | MEDLINE | ID: mdl-15099281

ABSTRACT

BACKGROUND: Venous thrombosis is a multicausal disease involving both genetic as well as acquired risk factors. Hyperhomocysteinemia is associated with a 2-fold increased risk of recurrent venous thrombosis (RVT). Recently, the 894 G > T variant of endothelial nitric oxide synthase (eNOS) was postulated to be associated with hyperhomocysteinemia. OBJECTIVES: We hypothesized an interrelation of hyperhomocysteinemia, the eNOS 894 G > T variant and RVT risk. METHODS: The eNOS 894 G > T variant was studied in 170 cases with a history of RVT and 433 controls from the general population. RESULTS: The eNOS 894 TT genotype may increase RVT risk [odds ratio (OR) 1.3 (0.7-2.6)], but no association of the eNOS 894 G > T variant with elevated homocysteine was found in controls. Interestingly, in RVT cases the coexistence of both the 894 TT genotype and elevated tHcy levels (> 90th percentile) was more frequently present than in controls, which led to a substantially increased risk of recurrent venous thrombosis [fasting tHcy OR 5.3 (1.1-24.1), postload tHcy OR 6.5 (1.6-29.5)]. CONCLUSION: The results of the present study demonstrate that the eNOS 894 G > T variation interacts with elevated tHcy levels, leading to an increased risk of recurrent thrombotic events. This interaction points in the direction of S-nitrosation as a mechanism by which homocysteine exerts its detrimental effects on the hemostatic system.


Subject(s)
Hyperhomocysteinemia/complications , Nitric Oxide Synthase/genetics , Polymorphism, Single Nucleotide , Venous Thrombosis/genetics , Case-Control Studies , Genotype , Homocysteine/metabolism , Humans , Hyperhomocysteinemia/epidemiology , Molecular Epidemiology , Nitric Oxide Synthase Type III , Nitrosation , Odds Ratio , Recurrence , Risk , Venous Thrombosis/epidemiology , Venous Thrombosis/etiology
10.
Cell Mol Biol (Noisy-le-grand) ; 50(8): 911-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15704255

ABSTRACT

Hyperhomocysteinemia (HHcy) is associated with impaired endothelial-dependent vasodilatation and increased risk of atherosclerosis and thrombosis. Here, we summarize some of our previous work on the effect of HHcy on pathways involved in endothelium-dependent vasodilatation, and present new data concerning the endothelium-derived hyperpolarizing factor (EDHF)-mediated vasodilatation. We showed that the 894 G>T single-nucleotide polymorphism in the human endothelial nitric oxide synthase gene (eNOS) increased the risk of recurrent venous thrombosis in individuals with elevated homocysteine levels, indicating that the pathophysiological mechanism in HHcy involves impaired NO-mediated vasodilatation. In addition, the EDHF-mediated vasodilatation of the renal artery was disturbed in diet-induced hyperhomocysteinemic rats. Interestingly, we demonstrated that pretreatment of rats with periodate-oxidized adenosine (Adox), which is an inhibitor of S-adenosylhomocysteine hydrolase, prevented the methionine-induced rise in plasma total Hcy (tHcy) levels but not the inhibition of the EDHF pathway. Furthermore, we demonstrated that S-adenosylhomocysteine (AdoHcy) and S-adenosylmethionine (AdoMet) levels were increased in the kidneys of diet-induced HHcy rats, resulting in a decreased AdoMet:AdoHcy ratio. In addition, we demonstrated that mRNA expression of Connexin 40, which is one of the structural subunits of gap-junctions, was down-regulated in endothelial cells of HHcy rats, and correlated with elevated AdoHcy levels in kidney of these rats. These finding suggest a key role for AdoHcy in relation to decreased Cx40 mRNA expression and impaired EDHF-mediated vasodilatation of HHcy rats.


Subject(s)
Biological Factors/metabolism , Hyperhomocysteinemia/metabolism , Nitric Oxide/metabolism , Animals , Connexins/metabolism , Endothelium, Vascular/metabolism , Gap Junctions , Humans , Kidney/metabolism , Models, Biological , Odds Ratio , Oxidative Stress , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylhomocysteine/metabolism , Time Factors , Vasodilation , Venous Thrombosis/metabolism , Gap Junction alpha-5 Protein
11.
Amino Acids ; 23(1-3): 331-6, 2002.
Article in English | MEDLINE | ID: mdl-12373555

ABSTRACT

UNLABELLED: Oxidative damage to DNA has been well documented in cardiac cells isolated from diabetic patients and rats with streptozotocin-induced diabetes mellitus (DM). This study evaluates possible molecular mechanisms for early events in the development of DM-induced cardiomyopathy. METHODS: To analyze the mechanism of overexpression of p21(WAF1/CIP1) and inhibition of cyclin D(1) expression in cardiomyocytes of diabetic rats we examined the methylation status of these genes by MS-PCR and assessed the possibility of epigenetic control of their expression. RESULTS: We found that the steady-state expression of both genes is influenced by their methylation status, as an epigenetic event, of their 5'-flanking regions upon development of diabetes. CONCLUSIONS: Oxidative damage contributes to the development of cardiomyopathy via p53-dependent activation of cardiac cell death. This pathway includes de novomethylation of the P53-inducible p21(WAF1/CIP1)-gene encoding a protein which binds to and inhibits a broad range of cyclin-cyclin-dependent kinase complexes.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cyclin D1/genetics , Cyclins/genetics , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Animals , CpG Islands , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , DNA Damage , DNA Methylation , Female , Myocytes, Cardiac/physiology , Promoter Regions, Genetic , Rats , Rats, Wistar
13.
Eur J Hum Genet ; 9(8): 583-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11528503

ABSTRACT

Molecular defects in genes encoding enzymes involved in homocysteine metabolism may account for mild hyperhomocysteinaemia, an independent and graded risk factor for cardiovascular disease (CVD). Although heterozygosity for cystathionine beta-synthase (CBS) deficiency has been excluded as a major genetic cause of mild hyperhomocysteinaemia in vascular disease, mutations in (non-)coding DNA sequences may lead to a mildly decreased CBS expression and, consequently, to elevated plasma homocysteine levels. We assessed the association between a 31 bp VNTR, that spans the exon 13-intron 13 boundary of the CBS gene, and fasting, post-methionine load and increase upon methionine load plasma homocysteine levels in 190 patients with arterial occlusive disease, and in 381 controls. The 31 bp VNTR consists of 16, 17, 18, 19 or 21 repeat units and shows a significant increase in plasma homocysteine concentrations with an increasing number of repeat elements, in particular after methionine loading. In 26 vascular disease patients the relationship between this 31 bp VNTR and CBS enzyme activity in cultured fibroblasts was studied. The CBS enzyme activity decreased with increasing number of repeat units of the 31 bp VNTR. RT-PCR experiments showed evidence of alternative splicing at the exon 13-intron 13 splice junction site. The 31 bp VNTR in the CBS gene is associated with post-methionine load hyperhomocysteinaemia that may predispose individuals to an increased risk of cardiovascular diseases.


Subject(s)
Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Homocysteine/blood , Homocysteine/genetics , Minisatellite Repeats/genetics , Alleles , Alternative Splicing/genetics , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/enzymology , Arterial Occlusive Diseases/genetics , Consensus Sequence/genetics , Enzyme Activation/genetics , Exons/genetics , Female , Gene Frequency/genetics , Genotype , Humans , Introns/genetics , Male , Middle Aged , Polymorphism, Genetic/genetics , Risk Factors
14.
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
15.
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
16.
Hypertens Pregnancy ; 19(3): 299-307, 2000.
Article in English | MEDLINE | ID: mdl-11118403

ABSTRACT

OBJECTIVE: To investigate the prevalence of the 677 (C --> T) and 1298 (A --> C) polymorphisms in the methylenetetrahydrofolate reductase (MTHFR) gene in our preeclamptic population. For a summary estimation of the risk of the 677 (C --> T) polymorphism for preeclampsia, we also performed a meta-analysis on four previously published case-control studies to which our results were added. METHODS: Genotypes were analyzed by polymerase chain reaction followed by restriction enzyme analysis. The results of 176 nonpregnant women, previously hospitalized for preeclampsia in a tertiary care center, were compared with 403 Dutch population-based controls. Results were statistically analyzed with a chi-square test. MEAN OUTCOME MEASURES: The incidence of the 677 (C --> T) and 1298 (A --> C) polymorphisms in the MTHFR gene. RESULTS: The incidence of both MTHFR missense polymorphisms was not significantly different between cases and controls. We found an odds ratio (OR) of 1.5 [95% confidence interval (CI) 0.8-2.6, p = 0.17] and an OR of 1.0 (95% CI 0.6-1.9, p = 0.23) for the 677 (C --> T) and the 1298 (A --> C) polymorphism, respectively, in cases comparing the prevalence of the homozygous genotype versus the other two genotypes. The meta-analysis resulted in a significant OR of 2.0 (95% CI 1.4-2.9). CONCLUSIONS: In contrast to four previous studies, we were neither able to confirm an increased risk for preeclampsia to the 677 (C --> T) polymorphism nor did we find an increased risk for preeclampsia to the 1298 (A --> C) polymorphism. From the meta-analysis, however, we conclude that it cannot be ruled out that the homozygous 677TT genotype is a modest but significant risk factor for preeclampsia.


Subject(s)
HELLP Syndrome/enzymology , HELLP Syndrome/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Polymorphism, Genetic/genetics , Pre-Eclampsia/enzymology , Pre-Eclampsia/genetics , Case-Control Studies , Chi-Square Distribution , Female , Genotype , HELLP Syndrome/epidemiology , Homozygote , Humans , Incidence , Methylenetetrahydrofolate Reductase (NADPH2) , Mutation, Missense/genetics , Netherlands/epidemiology , Odds Ratio , Polymerase Chain Reaction , Pre-Eclampsia/epidemiology , Pregnancy , Prevalence , Risk Factors
17.
Mol Genet Metab ; 71(3): 511-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11073719

ABSTRACT

Elevated homocysteine levels have been associated with arteriosclerosis and thrombosis. Hyperhomocysteinemia is caused by altered functioning of enzymes of its metabolism due to either inherited or acquired factors. Betaine-homocysteine methyltransferase (BHMT) serves, next to methionine synthase, as a facilitator of methyl group donation for remethylation of homocysteine into methionine, and reduced functioning of BHMT could theoretically result in elevated homocysteine levels. Recently, the genomic sequence of the BHMT gene was published. Mutation analysis may reveal mutations of the BHMT gene that could lead to hyperhomocysteinemia. In the present study we performed genomic sequencing of the BHMT gene of 16 vascular patients with hyperhomocysteinemia and detected three mutations in the coding region of this gene. The first was an amino acid substitution of glycine to serine (G199S), which was found only in the heterozygous state. The second mutation was a substitution of glutamine to arginine (Q239R), and the last mutation was an amino acid substitution of glutamine to histidine (Q406H). The latter was also found only in the heterozygous state. The relevance of these mutations was tested in a study group, which consists of 190 cases with vascular disease and 601 controls. The influence of these three mutations on homocysteine levels was investigated. None of the three mutations led to significantly changed homocysteine levels. In addition, no differences in genotype distribution between cases and controls were found. So far, our results provide no evidence for a role of defective BHMT functioning in hyperhomocysteinemia or subsequently in vascular disease.


Subject(s)
Hyperhomocysteinemia/genetics , Methyltransferases/genetics , Vascular Diseases/genetics , Amino Acid Substitution , Betaine-Homocysteine S-Methyltransferase , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Female , Gene Frequency , Genotype , Humans , Hyperhomocysteinemia/enzymology , Male , Methyltransferases/metabolism , Middle Aged , Mutation , Odds Ratio , Point Mutation , Sequence Analysis, DNA , Vascular Diseases/enzymology
18.
Biotechnol Appl Biochem ; 32(2): 121-5, 2000 10.
Article in English | MEDLINE | ID: mdl-11001872

ABSTRACT

Growing knowledge of the genetic basis of inheritable diseases has resulted in a rapidly increasing demand for DNA mutation analysis. Current methods are reliable and suitable for low-throughput mutation analyses, but are unable to cope with the increasing demand for genetic analyses, necessitating the development of new, fully automated and reliable methods. We developed a semi-automated method for DNA mutation analysis by integrating a thermocycler into a robotic pipetting workstation. DNA was extracted from 84 samples of 10 microl of EDTA-treated whole blood using magnetic beads within 2 h. Directly after isolation, the DNA was automatically transferred to an integrated thermocycler for amplification. Our semi-automated method proved to be reliable and robust, showing unambiguously interpretable PCR signals without occurrence of contamination. It is also faster than conventional manual methods. Only a brief manual intervention is required to remove and refit the seal of the PCR plate. This semi-automated assay is a step forward in the development of fully automated assays for DNA mutation analysis.


Subject(s)
DNA/blood , DNA/isolation & purification , Oxidoreductases Acting on CH-NH Group Donors/genetics , Polymerase Chain Reaction/instrumentation , Humans , Methylenetetrahydrofolate Reductase (NADPH2) , Oxidoreductases Acting on CH-NH Group Donors/blood , Point Mutation , Robotics
19.
Am J Obstet Gynecol ; 182(5): 1258-63, 2000 May.
Article in English | MEDLINE | ID: mdl-10819868

ABSTRACT

OBJECTIVE: This study was undertaken to investigate whether the cytosine-to-thymine substitution at nucleotide 677 (C677T) in the 5, 10-methylenetetrahydrofolate reductase gene is a risk factor for placental vasculopathy (abruptio placentae or placental infarction with fetal growth restriction). STUDY DESIGN: This case-control study enrolled 165 women with placental vasculopathy and 139 matched control women with normal pregnancy outcomes. Measurements included fasting total plasma homocysteine concentration, serum and red blood cell folate concentrations, serum vitamin B(12) concentration, whole-blood vitamin B(6) concentration, and analysis of the 5, 10-methylenetetrahydrofolate reductase gene C677T mutation. RESULTS: The median total plasma homocysteine concentration was significantly higher in the study group than in the control group (P <.01; odds ratio >97.5th percentile, 4.66; 95% confidence interval, 1.55-14.0). Homozygous genotype for the mutated 5,10-methylenetetrahydrofolate reductase gene was found in 12% of the study group and 5% of the control group (odds ratio, 2.45; 95% confidence interval, 1.00-6.02). CONCLUSIONS: Mild hyperhomocysteinemia was confirmed among women with placental vasculopathy, for which homozygosity for a mutated 5, 10-methylenetetrahydrofolate reductase gene was found to be a new risk factor. The risk of placental vasculopathy probably increases in conditions of low serum folate concentration.


Subject(s)
Mutation , Oxidoreductases/genetics , Placenta/blood supply , Vascular Diseases/genetics , 5,10-Methylenetetrahydrofolate Reductase (FADH2) , Adult , DNA Mutational Analysis , Female , Folic Acid/blood , Genotype , Homocysteine/blood , Homozygote , Humans , Methylenetetrahydrofolate Reductase (NADPH2) , Odds Ratio , Pregnancy , Vascular Diseases/enzymology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...