Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Intern Med ; 290(5): 1039-1047, 2021 11.
Article in English | MEDLINE | ID: mdl-33830582

ABSTRACT

BACKGROUND: Cerebrotendinous xanthomatosis (CTX) is an autosomal recessively inherited inborn error of metabolism. Neurological symptoms are considered to be a clinical hallmark of untreated adult patients. We describe a 'milder CTX phenotype', without neurological involvement. METHODS: We performed a retrospective patient file study in 79 genetically confirmed Dutch patients with CTX (55 patients aged ≥ 21 years) to study the clinical heterogeneity of CTX. We studied the frequency of adult patients with CTX without neurological involvement at diagnosis, in our Dutch cohort, and included a family from South Africa and patients from Italy, USA, Chile and Asia from the literature. RESULTS: In total, we describe 19 adult patients with CTX from 16 independent families, without neurological symptoms at diagnosis. A relatively small percentage (21%, n = 4) had a history of cataract. The majority, 84% (n = 16), presented with tendon xanthomas as the sole or predominant feature. The majority of patients showed increased plasma cholesterol levels. No correlation was found between this 'milder phenotype', the cholestanol levels and the CYP27A1 genotype. In addition, we describe three novel mutations in the CYP27A1 gene. CONCLUSIONS: This study shows the clinical heterogeneity of CTX, highlighting the existence of a 'milder phenotype', that is without neurological involvement at diagnosis. Adult patients with CTX may present with tendon xanthomas as the sole or predominant feature, mimicking familial hypercholesterolemia. It is important to realize that the absence of neurological symptoms does not rule out the development of future neurological symptoms. As CTX is a treatable disorder, early diagnosis and initiation of treatment when additional clinical signs occur is therefore essential.


Subject(s)
Xanthomatosis, Cerebrotendinous , Adult , Cholestanetriol 26-Monooxygenase/genetics , Humans , Retrospective Studies , Xanthomatosis, Cerebrotendinous/diagnosis , Xanthomatosis, Cerebrotendinous/genetics
2.
J Breath Res ; 10(4): 046014, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27902490

ABSTRACT

Staphylococcus aureus (S. aureus) is a common bacterium infecting children with cystic fibrosis (CF). Since current detection methods are difficult to perform in children, there is need for an alternative. This proof of concept study investigates whether breath profiles can discriminate between S. aureus infected and non-infected CF patients based on volatile organic compounds (VOCs). We collected exhaled breath of CF patients with and without S. aureus airways infections in which VOCs were identified using gas chromatography-mass spectrometry. We classified these VOC profiles with sparse partial least squares discriminant analysis. Multivariate breath VOC profiles discriminated infected from non-infected CF patients with high sensitivity (100%) and specificity (80%). We identified the nine compounds most important for this discrimination. We successfully detected S. aureus infection in CF patients, using breath VOC profiles. Nine highlighted compounds can be used as a focus point in further biomarker identification research. The results show considerable potential for non-invasive diagnosis of airway infections.


Subject(s)
Breath Tests/methods , Cystic Fibrosis/microbiology , Staphylococcus aureus/growth & development , Volatile Organic Compounds/adverse effects , Child , Female , Humans , Male , Volatile Organic Compounds/analysis
3.
J Breath Res ; 10(1): 016002, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26824272

ABSTRACT

Volatile organic compound (VOC) analysis in exhaled breath is proposed as a non-invasive method to detect respiratory infections in cystic fibrosis patients. Since polymicrobial infections are common, we assessed whether we could distinguish Pseudomonas aeruginosa and Aspergillus fumigatus mono- and co-cultures using the VOC emissions. We took headspace samples of P. aeruginosa, A. fumigatus and co-cultures at 16, 24 and 48 h after inoculation, in which VOCs were identified by thermal desorption combined with gas chromatography - mass spectrometry. Using multivariate analysis by Partial Least Squares Discriminant Analysis we found distinct VOC biomarker combinations for mono- and co-cultures at each sampling time point, showing that there is an interaction between the two pathogens, with P. aeruginosa dominating the co-culture at 48 h. Furthermore, time-independent VOC biomarker combinations were also obtained to predict correct identification of P. aeruginosa and A. fumigatus in mono-culture and in co-culture. This study shows that the VOC combinations in P. aeruginosa and A. fumigatus co-microbial environment are different from those released by these pathogens in mono-culture. Using advanced data analysis techniques such as PLS-DA, time-independent pathogen specific biomarker combinations can be generated that may help to detect mixed respiratory infections in exhaled breath of cystic fibrosis patients.


Subject(s)
Aspergillus fumigatus/metabolism , Pseudomonas aeruginosa/metabolism , Volatile Organic Compounds/analysis , Biomarkers/metabolism , Coculture Techniques , Exhalation , Gas Chromatography-Mass Spectrometry , Humans , Specimen Handling
5.
JIMD Rep ; 10: 39-40, 2013.
Article in English | MEDLINE | ID: mdl-23430798

ABSTRACT

Although the diagnosis of a primary carnitine deficiency is usually based on a very low level of free and total carnitine (free carnitine: 1-5 µM, normal 20-55 µM) (Longo et al. 2006), we detected a patient via newborn screening with a total carnitine level 67 % of the normal value. At the age of 1 year, after interruption of carnitine supplementation for a 4-week period the carnitine profile was assessed and the free carnitine level had dropped to 10.4 µmol/l (normal: 20-55 µM) and total carnitine level had dropped to 12.7 µmol/l (normal: 25-65 µM). Transient carnitine deficiency was not likely anymore and DNA mutation analysis of the OCTN2 (SLC22A5) gene showed a homozygous c.136C>T (p.P46S) mutation, confirming the diagnosis of primary carnitine deficiency. We would like to emphasize that neonates with a primary carnitine deficiency might present with relatively high levels of total carnitine due to placental carnitine transfer, and also draw the attention to the importance of regular follow-up and the significance of genetic diagnostics in patients with a nonclassical presentation.

6.
Clin Neuropathol ; 29(6): 361-4, 2010.
Article in English | MEDLINE | ID: mdl-21073839

ABSTRACT

Cerebrotendinous xanthomatosis (CTX) is a rare autosomal recessive lipid storage disorder due to a deficiency of the mitochondrial enzyme sterol 27-hydroxylase (CYP 27) with reduced or no chenodeoxycholic synthesis. This deficiency leads to an accumulation of cholestanol in different sites such as the eye lens, central nervous system or tendons. We report a 64-year-old female patient with a progressive gait disorder associated with cognitive decline since the age of 59. The patient had no mental retardation, cataract or chronic diarrhea. Her family reported increasing behavioral modifications 10 years previously. Clinical examination revealed a spastic paraplegia and bilateral xanthomas on the Achilles tendons. Cerebral magnetic resonance imaging (MRI) revealed diffuse hyperintense T2 abnormalities in the pyramidal tracts from the internal capsules to the cerebral peduncles also Technetium-99m-ECD brain SPECT showed a severe cerebellar hypoperfusion. Serum cholestanol analysis was 7 µmol/l (N). After 2 years, she was bedridden and died of aspiration pneumonia. The neuropathological study confirmed the CTX diagnosis and the sequencing analysis revealed that she was compound heterozygous for two mutations in the CYP27A1 gene: 1435 C > T (exon 7) on one allele and a new mutation, 1017 G > C (exon 5) on the other. The interest of the present case is to report neuropathology findings strongly correlated with the MRI and SPECT abnormalities.


Subject(s)
Xanthomatosis, Cerebrotendinous/diagnosis , Xanthomatosis, Cerebrotendinous/pathology , Cholestanetriol 26-Monooxygenase/genetics , Female , Humans , Magnetic Resonance Imaging , Middle Aged , Tomography, Emission-Computed, Single-Photon , Xanthomatosis, Cerebrotendinous/genetics
7.
Neurology ; 75(12): 1079-83, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20855850

ABSTRACT

OBJECTIVE: 3-Methylglutaconic aciduria type I is a rare inborn error of leucine catabolism. It is thought to present in childhood with nonspecific symptoms; it was even speculated to be a nondisease. The natural course of disease is unknown. METHODS: This is a study on 10 patients with 3-methylglutaconic aciduria type I. We present the clinical, neuroradiologic, biochemical, and genetic details on 2 new adult-onset patients and follow-up data on 2 patients from the literature. RESULTS: Two unrelated patients with the characteristic biochemical findings of 3- methylglutaconic aciduria type I presented in adulthood with progressive ataxia. One patient additionally had optic atrophy, the other spasticity and dementia. Three novel mutations were found in conserved regions of the AUH gene. In both patients, MRI revealed extensive white matter disease. Follow-up MRI in a 10-year-old boy, who presented earlier with isolated febrile seizures, showed mild abnormalities in deep white matter. CONCLUSION: We define 3-methylglutaconic aciduria type I as an inborn error of metabolism with slowly progressive leukoencephalopathy clinically presenting in adulthood. In contrast to the nonspecific findings in pediatric cases, the clinical and neuroradiologic pattern in adult patients is highly characteristic. White matter abnormalities may already develop in the first decades of life. The variable features found in affected children may be coincidental. Long-term follow-up in children is essential to learn more about the natural course of this presumably slowly progressive disease. Dietary treatment with leucine restriction may be considered.


Subject(s)
Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic, Inborn/pathology , Brain/pathology , Glutarates/metabolism , Leucine/metabolism , Leukoencephalopathies/pathology , Nerve Fibers, Myelinated/pathology , Adult , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Brain/metabolism , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/metabolism , Brain Mapping , Child , Disease Progression , Female , Humans , Image Processing, Computer-Assisted , Infant , Leukoencephalopathies/genetics , Leukoencephalopathies/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Fibers, Myelinated/metabolism
8.
QJM ; 101(11): 881-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18790817

ABSTRACT

BACKGROUND: Few studies have examined the effect of alcohol consumption on total homocysteine (tHcy) concentrations. AIM: To assess the effect of an 8-week intervention with vodka or red wine on plasma tHcy and B vitamin concentrations in healthy male volunteers. To assess the effect on tHcy according to methylenetetrahydrofolate reductase (MTHFR) 677C>T genotype. DESIGN AND METHODS: A randomized controlled crossover intervention study measuring tHcy and serum folate and vitamin B(12) concentrations was conducted in 78 male subjects (21-70 years). Following a 2-week washout period during which no alcohol was consumed, all subjects consumed 24 g alcohol (either 240 ml red wine or 80 ml vodka)/day for a 2-week period. Following a further 2-week washout, participants consumed the alternate intervention for 2 weeks. RESULTS: A significant increase in plasma tHcy was observed after the 2-week red wine intervention (5%, P = 0.03), and a non-significant increase in tHcy with vodka intervention (3%, P = 0.09). When the two interventions were compared, the change in tHcy did not differ between the vodka and red wine interventions (P = 0.57). There were significant decreases in serum vitamin B(12) and folate concentrations, and this decrease did not differ between interventions. The increase in tHcy observed in both interventions did not vary by MTHFR 677C>T genotype. CONCLUSION: A 2-week alcohol intervention resulted in a decrease in folate and vitamin B(12) status and an increase in plasma tHcy. The effect of alcohol intervention on tHcy, folate and vitamin B(12) concentrations did not differ between the red wine and vodka intervention groups.


Subject(s)
Alcohol Drinking/metabolism , Folic Acid/metabolism , Homocysteine/metabolism , Vitamin B 12/metabolism , Adult , Aged , Cross-Over Studies , Genotype , Humans , Male , Methylenetetrahydrofolate Reductase (NADPH2)/blood , Middle Aged , Young Adult
9.
Kidney Int ; 72(10): 1233-41, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17851469

ABSTRACT

The multidrug resistance gene 1 product, P-glycoprotein (P-gp), is expressed in several excretory organs, including the apical membrane of proximal tubules. After inducing acute renal failure, P-gp expression is upregulated and this might be a protective function by pumping out toxicants and harmful products of oxidative stress. We characterized renal function of P-gp knockout mice and studied its consequences in renal ischemic damage. Compared with wild-type mice, knockout mice have a lower glomerular filtration rate and renal plasma flow. An augmented urinary excretion of sodium, numerous amino acids, calcium, glucose, and low molecular weight proteins was observed along with an increased diuresis. A higher lithium plasma clearance in the knockout mice suggested proximal tubular dysfunction. Electron microscopy showed mitochondrial abnormalities in proximal tubular cells that could account for decreased adenosine triphosphate levels in the cortex. After inducing ischemia, wild-type mice showed a decrease in creatinine clearance and severe proximal tubular necrosis. In contrast, knockout mice had no signs of tubular damage. Our data indicate that P-gp knockout mice have impaired renal function but are protected against ischemic renal injury.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , Ischemia/prevention & control , Kidney Tubules, Proximal/physiopathology , Acute Kidney Injury/physiopathology , Amino Acids/urine , Animals , Calcium/urine , Diuresis , Fluorescent Antibody Technique , Glomerular Filtration Rate/genetics , Glycosuria , Immunohistochemistry , Ischemia/etiology , Kidney Tubules, Proximal/ultrastructure , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Mitochondria/pathology , Mitochondria/ultrastructure , Proteinuria , Renal Circulation/genetics , Sodium/urine
10.
Mol Genet Metab ; 88(1): 47-52, 2006 May.
Article in English | MEDLINE | ID: mdl-16527507

ABSTRACT

In this paper, we describe a distinct clinical subtype of 3-methylglutaconic aciduria. 3-Methylglutaconic aciduria is a group of different metabolic disorders biochemically characterized by increased urinary excretion of 3-methylglutaconic acid. We performed biochemical and genetic investigations, including urine organic acid analysis, NMR spectroscopy, measurement of 3-methylglutaconyl-CoA hydratase activity, cardiolipin levels, OPA3 gene analysis and measurement of the oxidative phosphorylation in four female patients with 3-methylglutaconic aciduria. 3-Methylglutaconic aciduria type I, Barth syndrome, and Costeff syndrome were excluded as the activity of 3-methylglutaconyl-CoA hydratase, the cardiolipin levels, and molecular analysis of the OPA3 gene, respectively, showed no abnormalities. The children presented with characteristic association of hearing loss and the neuro-radiological evidence of Leigh disease. They also had neonatal hypotonia, recurrent lactic acidemia, episodes with hypoglycemia and severe recurrent infections, feeding difficulties, failure to thrive, developmental delay, and progressive spasticity with extrapyramidal symptoms. Our patients were further biochemically characterized by a mitochondrial dysfunction and persistent urinary excretion of 3-methylglutaconic acid.


Subject(s)
Brain Diseases, Metabolic/physiopathology , Glutarates/urine , Hearing Loss, Sensorineural/physiopathology , Leigh Disease/physiopathology , Mitochondrial Diseases/physiopathology , Adolescent , Child , Child, Preschool , Consanguinity , Fatal Outcome , Female , Humans , Infant, Newborn , Male , Oxidative Phosphorylation , Syndrome , Valerates/urine
11.
Int J Obes (Lond) ; 30(6): 892-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16432543

ABSTRACT

OBJECTIVE: Familial combined hyperlipidemia (FCH) is characterized by elevated levels of total cholesterol (TC), triglycerides (TG) and apolipoprotein B (apo B) and is associated with premature cardiovascular disease (CVD). Other features of FCH are obesity and insulin resistance. Serum leptin levels have also been associated with obesity, insulin resistance and atherosclerosis. Leptin exerts its effect through the leptin receptor (LEPR). The aim of this study is to determine whether the Gln223Arg polymorphism in the LEPR gene contributes to FCH and its associated phenotypes. METHODS: The study population consists of 37 families, comprising 644 subjects, of whom 158 subjects were diagnosed as FCH. The FCH diagnosis was based on plasma TC and TG levels, adjusted for age and gender, and absolute apo B levels, according to our recently published nomogram. The Gln223Arg polymorphism was studied by restriction fragment length polymorphism-PCR. RESULTS: Carriers of one or two Arg alleles had an increased risk of FCH, compared to subjects homozygous for the Gln allele (OR=1.6 [95% CI 1.0-2.4]). A difference in high-density lipoprotein cholesterol (HDL-c) levels was present between carriers and non-carriers of an Arg allele, 1.21 vs 1.28 mmol/l, respectively (P=0.04), but no differences in obesity, insulin resistance and other lipid parameters were found. CONCLUSION: The Gln223Arg polymorphism in the LEPR gene is associated with FCH, which is supported by a significant association between HDL-c levels and the LEPR gene.


Subject(s)
Hyperlipidemia, Familial Combined/genetics , Polymorphism, Genetic , Receptors, Cell Surface/genetics , Adult , Aged , Anthropometry , Body Mass Index , Cholesterol/blood , Cholesterol, HDL/blood , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Hyperlipidemia, Familial Combined/blood , Male , Middle Aged , Phenotype , Polymorphism, Restriction Fragment Length , Receptors, Leptin , Triglycerides/blood
12.
Neuromuscul Disord ; 15(12): 844-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16288870

ABSTRACT

We present a patient, who experienced recurrent episodes of rhabdomyolysis. Her beneficial response to treatment with dantrolene sodium was previously reported. Adult onset Very Long Chain Acyl-CoA Dehydrogenase (VLCAD) deficiency has been diagnosed only recently. In adults, VLCAD deficiency results in recurrent fasting-, exercise-, or infection-induced muscle stiffness, muscle pain and myoglobinuria caused by rhabdomyolysis. This case illustrates for the first time the beneficial effect of dantrolene in VLCAD deficiency. We discuss the therapeutic mechanism of dantrolene sodium and its possible role as additional treatment modality for patients with VLCAD deficiency.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Dantrolene/therapeutic use , Muscle Relaxants, Central/therapeutic use , Rhabdomyolysis/drug therapy , Aged , Female , Humans , Rhabdomyolysis/metabolism
13.
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
14.
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
15.
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
16.
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
17.
J Clin Oncol ; 19(20): 4081-7, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11600611

ABSTRACT

PURPOSE: Irinotecan and raltitrexed display schedule-dependent synergy in vitro, which supports the clinical investigation of the combination. Functional polymorphisms of the methylenetetrahydrofolate reductase (MTHFR) gene result in intracellular redistribution of folate derivatives, which may affect raltitrexed-associated cytotoxicity. PATIENTS AND METHODS: Patients with a range of solid cancers and good performance status received irinotecan as a 90-minute infusion on day 1 and raltitrexed as a 15-minute infusion on day 2, repeated every 21 days. Samples were collected for MTHFR C677T genotyping and fasting plasma homocysteine during the first cycle. RESULTS: Thirty-nine assessable patients received 127 cycles of therapy. Irinotecan doses ranged from 100 to 350 mg/m(2), and raltitrexed, 1.0 to 4.0 mg/m(2). Raltitrexed doses of more than 3.0 mg/m(2) were not tolerated and were associated with dose-limiting asthenia, diarrhea, and AST/ALT elevation. Irinotecan/raltitrexed doses of 350/3.0 mg/m(2) were well-tolerated; principal toxicities included neutropenia, diarrhea, and fatigue. Two partial responses were observed in patients with pretreated gastroesophageal cancers. Homozygotes with the MTHFR 677 TT polymorphism incurred significantly less raltitrexed-associated toxicity than those with either wild-type or heterozygous genotypes (P = .05). No significant differences were noted in plasma homocysteine values between the genotypic subtypes, and plasma homocysteine levels did not predict the risk of toxicity. CONCLUSION: Irinotecan and raltitrexed doses of 350 and 3.0 mg/m(2) are recommended for further study on a day 1, 2 schedule every 21 days. Efficacy results suggest that trials in upper and lower gastrointestinal malignancies are warranted. MTHFR C677T genotypes may be predictive of clinical raltitrexed toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Neoplasms/drug therapy , Oxidoreductases Acting on CH-NH Group Donors/genetics , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Camptothecin/administration & dosage , Diarrhea/chemically induced , Dose-Response Relationship, Drug , Fatigue/chemically induced , Female , Genotype , Homocysteine/blood , Humans , Irinotecan , Male , Methylenetetrahydrofolate Reductase (NADPH2) , Middle Aged , Neoplasms/blood , Neoplasms/genetics , Neutropenia/chemically induced , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Quinazolines/administration & dosage , Thiophenes/administration & dosage
18.
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
19.
Atherosclerosis ; 157(2): 451-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11472746

ABSTRACT

Epidemiological evidence has revealed that an elevated plasma homocysteine level (hyperhomocysteinemia) confers an increased risk of cardiovascular disease and neural tube defects. Hyperhomocysteinemia is caused by both nutritional (e.g. folate, vitamins B(6) and B(12)) and genetic factors, including functional polymorphisms of key enzymes involved in homocysteine metabolism. One such enzyme, methionine synthase reductase (MTRR), maintains adequate levels of methylcob(III)alamin, the activated cofactor for methionine synthase, which catalyzes the remethylation of homocysteine to methionine. A common MTRR polymorphism, i.e. a 66 A-->G substitution specifying an isoleucine to methionine substitution (I22M), was recently identified. To assess the influence of this polymorphism on total plasma homocysteine (tHcy), we undertook a genotype/phenotype analysis in a study population of 601 Northern-Irish men, aged 30--49, for which biochemical and genetic data relevant to folate/homocysteine metabolism had already been acquired. The 66AA genotype has a frequency of 29% in this population. We established that there was a significant influence of MTRR genotype on tHcy ranking (P=0.004) and that the 66AA genotype contributes to a moderate increase in tHcy levels across the distribution [OR 1.59 (95% CI: 1.10--2.25) for the 66AA genotype to be in the upper half of the tHcy distribution, P=0.03]. The homocysteine-elevating effect of the 66AA genotype is independent of serum folate, vitamin B(12) and vitamin B(6) levels. Based on published estimates of the enhanced cardiovascular disease risk conferred by defined increments of plasma tHcy, we estimate that 66AA homozygotes have, on average, an approximately 4% increase in cardiovascular disease risk compared to 66GG homozygotes. This study provides the first evidence that the MTRR A66G polymorphism significantly influences the circulating tHcy concentration.


Subject(s)
Ferredoxin-NADP Reductase/genetics , Genetic Predisposition to Disease , Homocysteine/blood , Hyperhomocysteinemia/genetics , Polymorphism, Genetic/physiology , Adult , Gene Frequency , Genotype , Humans , Male , Middle Aged , Osmolar Concentration
20.
Eur Heart J ; 22(4): 294-9, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11161947

ABSTRACT

AIMS: Elevated plasma homocysteine is an independent risk factor for atherothrombotic disease. Individuals homozygous for the methylenetetrahydrofolate reductase (MTHFR) 677C allele exclusively accumulate 5methyltetrahydrofolate, the methyl donor for homocysteine remethylation, in their red blood cells; this contrasts with 677 TT homozygotes who also accumulate significant levels of non-methylated folate derivatives. Those with the MTHFR 677 TT, CT and CC genotypes may therefore differ qualitatively with respect to folate utilization and hence their capacity to remethylate homocysteine. This study was consequently designed to establish whether all three genotypes confer different levels of atherothrombotic risk. METHODS AND RESULTS: The risk of atherothrombotic disease conferred by the MTHFR 677 CT and 677 CC genotypes was assessed using a 'restricted' meta-analysis approach applied to subjects from the first ten studies reporting a significantly increased risk conferred by the 677 TT genotype. The defined risk of the TT genotype in each of these ten studies was judged by us to denote 'genetic vulnerability' in the populations from which subjects were drawn. After proportional adjustment for the greater number of case TT homozygotes, the CT and CC frequencies observed in cases were compared with expectations based on the frequencies of these genotypes in controls. The observed CT frequency among cases was higher than expected in eight of the ten studies. In the meta-analysis, which included 1857 cases and 2942 controls, 847 (45.6%) cases, instead of the 777 (41.8%) expected, had the MTHFR CT genotype (P=0.010). CONCLUSIONS: Our findings suggest that the three MTHFR C677T genotypes confer different levels of atherothrombotic risk in 'genetically vulnerable' populations: CT heterozygotes have an elevated risk over CC homozygotes. One explanation is that the CT genotype actively confers atherothrombotic risk. An alternative interpretation however, for which a biologically plausible mechanism is proposed, is that CC is a protective genotype.


Subject(s)
Coronary Artery Disease/genetics , Genetic Predisposition to Disease , Oxidoreductases Acting on CH-NH Group Donors/genetics , Folic Acid/metabolism , Genotype , Homocysteine/metabolism , Humans , Methylenetetrahydrofolate Reductase (NADPH2) , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL
...