Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Genet Metab ; 122(1-2): 100-106, 2017 09.
Article in English | MEDLINE | ID: mdl-28751108

ABSTRACT

BACKGROUND: The autosomal recessive, neurodegenerative disorder mucopolysaccharidosis type IIIB (MPSIIIB) is caused by a deficiency of the lysosomal enzyme N-acetyl-α-glucosaminidase (NAGLU), resulting in accumulation of heparan sulfate. The disease spectrum comprises a severe, rapidly progressing (RP) phenotype and a more attenuated, slowly progressing (SP) phenotype. Previous studies showed significantly higher NAGLU activity in skin fibroblasts of SP patients when cultured at 30°C which may be relevant for development of novel therapeutic strategies. Here we report on the processes involved in this phenomenon. METHODS: Fibroblasts from controls, one RP patient (homozygous for the p.R297* mutation) and three SP MPSIIIB patients (homozygous for the mutation p.S612G or p.R643C, or compound heterozygous for the mutations p.A72_G79dup8 and p.R565Q) were cultured at temperatures ranging from 37°C to 27°C and harvested at different time points to assess NAGLU activity, mRNA and protein levels, and NAGLU glycosylation. Intracellular localization of wild-type and mutant mCherry-tagged NAGLU was analyzed by immunofluorescence. RESULTS: In control fibroblasts NAGLU was present as a 85kDa precursor and a 82kDa mature form. In SP patients' fibroblasts cultured at 37°C, only the 85kDa form was detected. Culturing at lower temperatures resulted in higher NAGLU mRNA levels, increased levels of both precursor and mature NAGLU protein and improved processing. The formation of mature NAGLU corresponded with higher NAGLU activity levels. CONCLUSION: We show that the NAGLU protein consists of a precursor and a mature form and that in SP MPSIIIB patients' fibroblasts only the precursor protein is present at 37°C. Culturing at lower temperatures resulted in the formation of the mature, enzymatically active form, due to higher mRNA levels and improved processing.


Subject(s)
Acetylglucosaminidase/metabolism , Mucopolysaccharidosis III/genetics , Acetylglucosaminidase/genetics , Cell Culture Techniques , Cells, Cultured , Enzyme Precursors/metabolism , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Male , Mucopolysaccharidosis III/drug therapy , Mucopolysaccharidosis III/enzymology , Mutant Proteins/metabolism , Mutation , Real-Time Polymerase Chain Reaction , Skin/cytology , Temperature
2.
Anal Biochem ; 535: 47-55, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28757091

ABSTRACT

Current methodologies for the assessment of urea cycle (UC) enzymatic activity are insufficient to accurately evaluate this pathway in biological specimens where lower UC is expected. Liver cell lines, including HepaRG, have been described to have limited nitrogen fixation through the UC, limiting their applicability as biocomponents for Bioartificial Livers (BAL). This work aims to develop novel and sensitive analytical solutions using Mass Spectrometry-based methodology to measure the activity of four UC enzymes in human liver and HepaRG cells. Activity of carbamoyl-phosphate synthetase I (CPS I), ornithine transcarbamylase (OTC), argininosuccinate lyase (ASL) and arginase (ARG I and II) was determined on homogenates from normal human liver and HepaRG cells cultured in monolayer or in the AMC-BAL. Enzyme products were determined by stable-isotope dilution UPLC-MS/MS. Activity of CPS I, OTC and ARG I/II enzymes in HepaRG monolayer cultures was considerably lower than in human control livers albeit an increase was achieved in HepaRG-BAL cultures. Improved analytical assays developed for the study of UC enzyme activity, contributed to gain understanding of UC function in the HepaRG cell line. The decreased activity of CPS I suggests that it may be a potential rate-limiting factor underlying the low UC activity in this cell line.


Subject(s)
Arginase/metabolism , Argininosuccinate Lyase/metabolism , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Liver/enzymology , Liver/metabolism , Ornithine Carbamoyltransferase/metabolism , Urea/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Humans , Tandem Mass Spectrometry
3.
Biochim Biophys Acta ; 1842(12 Pt A): 2510-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-23643712

ABSTRACT

Hyperammonemia is a frequent finding in various organic acidemias. One possible mechanism involves the inhibition of the enzyme N-acetylglutamate synthase (NAGS), by short-chain acyl-CoAs which accumulate due to defective catabolism of amino acids and/or fatty acids in the cell. The aim of this study was to investigate the effect of various acyl-CoAs on the activity of NAGS in conjunction with the formation of glutamate esters. NAGS activity was measured in vitro using a sensitive enzyme assay with ultraperformance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) product analysis. Propionyl-CoA and butyryl-CoA proved to be the most powerful inhibitors of N-acetylglutamate (NAG) formation. Branched-chain amino acid related CoAs (isovaleryl-CoA, 3-methylcrotonyl-CoA, isobutyryl-CoA) showed less pronounced inhibition of NAGS whereas the dicarboxylic short-chain acyl-CoAs (methylmalonyl-CoA, succinyl-CoA, glutaryl-CoA) had the least inhibitory effect. Subsequent work showed that the most powerful inhibitors also proved to be the best substrates in the formation of N-acylglutamates. Furthermore, we identified N-isovalerylglutamate, N-3-methylcrotonylglutamate and N-isobutyrylglutamate (the latter two in trace amounts), in the urines of patients with different organic acidemias. Collectively, these findings explain one of the contributing factors to secondary hyperammonemia, which lead to the reduced in vivo flux through the urea cycle in organic acidemias and result in the inadequate elimination of ammonia.


Subject(s)
Acyl Coenzyme A/pharmacology , Amino-Acid N-Acetyltransferase/antagonists & inhibitors , Amino-Acid N-Acetyltransferase/metabolism , Glutamic Acid/metabolism , Acyl Coenzyme A/metabolism , Carboxylic Acids/metabolism , Chromatography, High Pressure Liquid/methods , Dicarboxylic Acids/metabolism , Dose-Response Relationship, Drug , Esters , Glutamic Acid/chemistry , Humans , Hyperammonemia/metabolism , Kinetics , Tandem Mass Spectrometry
4.
J Inherit Metab Dis ; 36(2): 271-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22968582

ABSTRACT

BACKGROUND: Sanfilippo disease (Mucopolysaccharidosis III) is a neurodegenerative lysosomal disorder characterized by accumulation of the glycosaminoglycan heparan sulfate (HS). MPS III has a large phenotypic variability and early assessment of disease severity is difficult. We investigated the correlation between disease severity and the plasma concentration of HS (pHS, defined by the sum of the heparan sulfate derived disaccharides obtained after enzymatic digestion) and urinary total GAGs level (uGAGs, measured by the dimethylene blue test) in a cross-sectional cohort of 44 MPS III patients. METHODS: Disease severity was established on the basis of the age of complete loss of independent walking and of full loss of speech in all patients. Hazard ratios (HR) were obtained with cox-regression analysis. In order to allow prediction of a severe phenotype based on a cut-off value for pHS, patients were divided in two groups (severely affected and less severely affected) based on predictive mutations or on the age of full loss of speech. Receiver operator characteristics (ROC) were obtained for pHS. RESULTS: pHS and uGAGs were independently and linearly associated with an increased risk of speech loss with a HR of 1.8 (95 % CI 1.3-2.7) per 500 ng/ml increase of HS in plasma (p = 0.002), and a HR of 2.7 (95 % CI 1.6-4.4) per 10 mg/mmol creatinine increase of uGAGs (p < 0.001). pHS and uGAGS were less strongly associated with loss of walking. The area under the ROC curve for pHS was 0.85, indicating good discrimination. CONCLUSION: pHS and uGAGs may be useful biomarkers for prediction of severity in MPS III.


Subject(s)
Disaccharides/blood , Glycosaminoglycans/urine , Heparitin Sulfate/blood , Mucopolysaccharidosis III/blood , Mucopolysaccharidosis III/urine , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Mucopolysaccharidosis III/pathology , Young Adult
5.
Nat Genet ; 17(2): 190-3, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326940

ABSTRACT

Refsum disease is an autosomal-recessively inherited disorder characterized clinically by a tetrad of abnormalities: retinitis pigmentosa, peripheral neuropathy, cerebellar ataxia and elevated protein levels in the cerebrospinal fluid (CSF) without an increase in the number of cells in the CSF. All patients exhibit accumulation of an unusual branched-chain fatty acid, phytanic acid (3,7,11,15-tetramethylhexadecanoic acid), in blood and tissues. Biochemically, the disease is caused by the deficiency of phytanoyl-CoA hydroxylase (PhyH), a peroxisomal protein catalyzing the first step in the alpha-oxidation of phytanic acid. We have purified PhyH from rat-liver peroxisomes and determined the N-terminal amino-acid sequence, as well as an additional internal amino-acid sequence obtained after Lys-C digestion of the purified protein. A search of the EST database with these partial amino-acid sequences led to the identification of the full-length human cDNA sequence encoding PhyH: the open reading frame encodes a 41.2-kD protein of 338 amino acids, which contains a cleavable peroxisomal targeting signal type 2 (PTS2). Sequence analysis of PHYH fibroblast cDNA from five patients with Refsum disease revealed distinct mutations, including a one-nucleotide deletion, a 111-nucleotide deletion and a point mutation. This analysis confirms our finding that Refsum disease is caused by a deficiency of PhyH.


Subject(s)
Mixed Function Oxygenases/genetics , Mutation , Refsum Disease/enzymology , Refsum Disease/genetics , Adult , Amino Acid Sequence , Animals , Base Sequence , Case-Control Studies , DNA Primers/genetics , DNA, Complementary/genetics , Female , Gene Expression , Humans , Infant , Liver/enzymology , Male , Microbodies/enzymology , Mixed Function Oxygenases/isolation & purification , Molecular Sequence Data , Point Mutation , Polymerase Chain Reaction , Rats , Sequence Deletion
6.
J Inherit Metab Dis ; 35(6): 1021-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22350545

ABSTRACT

Isovaleric acidemia (IVA) is one of the most common organic acidemias found in South Africa. Since 1983, a significant number of IVA cases have been identified in approximately 20,000 Caucasian patients screened for metabolic defects. IVA is caused by an autosomal recessive deficiency of isovaleryl-CoA dehydrogenase (IVD) resulting in the accumulation of isovaleryl-CoA and its metabolites. In total, 10 IVA patients and three carriers were available for phenotypic and genotypic investigation in this study. All patients were found to be homozygous for a single c.367 G > A (p.G123R) mutation. The amino acid substitution of a glycine to arginine resulted in a markedly reduced steady-state level of the IVD protein, which explains the nearly complete lack of IVD enzyme activity as assessed in fibroblast homogenates. Despite the genetic homogeneity of this South African IVA group, the clinical presentation varied widely, ranging from severe mental handicap and multiple episodes of metabolic derangement to an asymptomatic state. The variation may be due to poor dietary intervention, delayed diagnosis or even epigenetic and polygenetic factors of unknown origin.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Biomarkers/urine , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Female , Genotype , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Isovaleryl-CoA Dehydrogenase/deficiency , Isovaleryl-CoA Dehydrogenase/genetics , Isovaleryl-CoA Dehydrogenase/metabolism , Male , Molecular Sequence Data , Mutation, Missense , Phenotype , Sequence Homology, Amino Acid , South Africa , White People/genetics , Young Adult
7.
Neuropediatrics ; 42(1): 13-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21500142

ABSTRACT

Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is an autosomal recessive inborn error of metabolism, most frequently associated with developmental delay and/or epilepsy. Most SCADD patients carry common SCAD-encoding gene ( ACADS) variants or these variants in combination with a rare ACADS mutation, in the Netherlands predominantly the c.1058C>T. Epilepsy in childhood often remains unexplained and patients with epilepsy related to SCADD may remain undiagnosed because studies for SCADD are often not performed. To test this hypothesis and to further estimate the extent of the Dutch SCADD population, we performed a study on blood spot samples in 131 paediatric patients with epilepsy and 909 anonymous newborns and investigated the presence of the 2 common ACADS variants and the rare c.1058C>T mutation. Overall, the 2 common ACADS variants and the rare c.1058C>T mutation were detected in either homozygous or compound heterozygous forms in 9.2% of the epilepsy and 7.5% of the reference group. A birth prevalence of SCADD with a mutation/variant genotype in the Netherlands as high as >1:1,000 was calculated. This is in contrast with the low number of patients diagnosed clinically and supports the hypothesis that SCADD is clinically irrelevant. Furthermore our study does not support an association between SCADD and epilepsy.


Subject(s)
Epilepsy/epidemiology , Lipid Metabolism, Inborn Errors/epidemiology , Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/genetics , Adolescent , Butyryl-CoA Dehydrogenase/genetics , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Incidence , Infant , Infant, Newborn , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Male , Mutation/genetics , Netherlands/epidemiology , Pediatrics
8.
J Inherit Metab Dis ; 31(2): 205-16, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18392741

ABSTRACT

Valproic acid (VPA; 2-n-propylpentanoic acid) is widely used as a major drug in the treatment of epilepsy and in the control of several types of seizures. Being a simple fatty acid, VPA is a substrate for the fatty acid beta-oxidation (FAO) pathway, which takes place primarily in mitochondria. The toxicity of valproate has long been considered to be due primarily to its interference with mitochondrial beta-oxidation. The metabolism of the drug, its effects on enzymes of FAO and their cofactors such as CoA and/or carnitine will be reviewed. The cumulative consequences of VPA therapy in inborn errors of metabolism (IEMs) and the importance of recognizing an underlying IEM in cases of VPA-induced steatosis and acute liver toxicity are two different concepts that will be emphasized.


Subject(s)
Anticonvulsants/toxicity , Fatty Acids/metabolism , Mitochondria/drug effects , Valproic Acid/toxicity , Animals , Anticonvulsants/pharmacokinetics , Biotransformation , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Fatty Liver/chemically induced , Fatty Liver/metabolism , Humans , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/metabolism , Mitochondria/metabolism , Oxidation-Reduction , Risk Assessment , Risk Factors , Valproic Acid/pharmacokinetics
9.
Ned Tijdschr Geneeskd ; 152(30): 1678-85, 2008 Jul 26.
Article in Dutch | MEDLINE | ID: mdl-18714522

ABSTRACT

OBJECTIVE: To describe the clinical, genetic, and biochemical characteristics of short-chain acyl-CoA dehydrogenase deficiency (SCADD), a clinically heterogeneous metabolic disorder for which neonates are screened for in parts of the United States and Australia. To explore the genotype-phenotype relation and to discuss neonatal screening for SCADD. DESIGN: Retrospective study of 31 Dutch SCADD patients and 8 SCADD relatives. METHOD: Patients and relatives were included ifbiochemical SCADD characteristics (increased C4-carnitine and/or ethylmalonic acid) were present in combination with a mutation and/or the c.511C>T or c.625G>A variant on each SCAD-encoding (ACADS) allele. The patients were subdivided into 3 genotype groups: mutation/mutation, mutation/variant and variant/variant group. RESULTS: A birth prevalence for SCADD of at least 1:50,000 was calculated. Most patients presented before the age of 3 years, mainly with developmental delay, epilepsy, behavioural disturbances and/or hypoglycaemia. The ACADS genotype showed a statistically significant association with biochemical, but not with clinical characteristics. In total 7 out of 8 SCADD relatives were free of symptoms. In 5 of the 31 patients, of whom 2 had severe symptoms, a second diagnosis was made which might explain the symptoms. CONCLUSION: SCADD was far more common than had previously been assumed and clinical symptoms in SCADD were non-specific, often transient or absent and not correlated with specific ACADS genotypes. SCADD does not meet major neonatal screening criteria and is therefore not suited for inclusion in neonatal screening programmes.

10.
J Clin Invest ; 98(4): 1028-33, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8770876

ABSTRACT

Mitochondrial trifunctional protein (MTP) is a recently identified enzyme involved in mitochondrial beta-oxidation, harboring long-chain enoyl-CoA hydratase, long-chain 3-hydroxyacyl-CoA dehydrogenase (LCHAD) and long-chain 3-ketothiolase activity. A deficiency of this protein is associated with impaired oxidation of long-chain fatty acids which can lead to sudden infant death. Furthermore, it is clear that this inborn error of fatty acid oxidation is very frequent, second to medium chain acyl-CoA dehydrogenase deficiency. In most patients only the LCHAD activity of MTP is deficient with near normal activity of the two other enzyme activities of the complex. We recently described the occurrence of a frequent G1528C mutation in the cDNA coding for the a subunit of MTP. Using S. cerevisiae for expression of wild type and mutant protein we show that the G1528C mutation is directly responsible for the loss of LCHAD activity. Furthermore, we describe a newly developed method allowing identification of the G1528C mutation in genomic DNA. The finding of an 87% allele frequency of the G1528C mutation in 34 LCHAD deficient patients makes this a valuable test for prenatal diagnosis. Finally, we show that the gene encoding the alpha subunit of MTP is located on chromosome 2p24.1-23.3.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/genetics , Chromosomes, Human, Pair 2 , Multienzyme Complexes/genetics , Base Sequence , DNA Primers/chemistry , Genes , Humans , In Situ Hybridization, Fluorescence , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Trifunctional Protein , Molecular Sequence Data , Point Mutation , Polymorphism, Restriction Fragment Length , Recombinant Proteins , Saccharomyces cerevisiae
11.
J Clin Invest ; 102(3): 527-31, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9691089

ABSTRACT

Mitochondrial fatty acid beta-oxidation is important for energy production, which is stressed by the different defects found in this pathway. Most of the enzyme deficiencies causing these defects are well characterized at both the protein and genomic levels. One exception is carnitine palmitoyltransferase I (CPT I) deficiency, of which until now no mutations have been reported although the defect is enzymatically well characterized. CPT I is the key enzyme in the carnitine-dependent transport across the mitochondrial inner membrane and its deficiency results in a decreased rate of fatty acid beta-oxidation. Here we report the first delineation of the molecular basis of hepatic CPT I deficiency in a new case. cDNA analysis revealed that this patient was homozygous for a missense mutation (D454G). The effect of the identified mutation was investigated by heterologous expression in yeast. The expressed mutant CPT IA displayed only 2% of the activity of the expressed wild-type CPT IA, indicating that the D454G mutation is the disease-causing mutation. Furthermore, in patient's fibroblasts the CPT IA protein was markedly reduced on immunoblot, suggesting that the mutation renders the protein unstable.


Subject(s)
Carnitine O-Palmitoyltransferase/deficiency , Isoenzymes/deficiency , Lipid Metabolism, Inborn Errors/genetics , Amino Acid Sequence , Biological Transport , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Consanguinity , Consensus Sequence , Dicarboxylic Acids/urine , Fatty Acids/metabolism , Female , Humans , Hypoglycemia/etiology , Infant , Isoenzymes/genetics , Isoenzymes/physiology , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/urine , Liver/enzymology , Mitochondria/metabolism , Molecular Sequence Data , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity
12.
J Clin Invest ; 102(6): 1193-9, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9739053

ABSTRACT

Human mitochondrial trifunctional protein (TFP) is a heterooctamer of four alpha- and four beta-subunits that catalyzes three steps in the beta-oxidation spiral of long-chain fatty acids. TFP deficiency causes a Reye-like syndrome, cardiomyopathy, or sudden, unexpected death. We delineated the molecular basis for TFP deficiency in two patients with a unique phenotype characterized by chronic progressive polyneuropathy and myopathy without hepatic or cardiac involvement. Single-stranded conformation variance and nucleotide sequencing identified all patient mutations in exon 9 of the alpha-subunit. One patient is homozygous for the T845A mutation that substitutes aspartic acid for valine at residue 246. The second patient is a compound heterozygote for the T914A that substitutes asparagine for isoleucine at residue 269 and a C871T that creates a premature termination at residue 255. Allele-specific oligonucleotide hybridization studies revealed undetectable levels of the mRNA corresponding to the mutant allele carrying the termination codon. This study suggests a novel genotype-phenotype correlation in TFP deficiency; that is, mutations in exon 9 of the alpha-subunit, which encodes a linker domain between the NH2-terminal hydratase and the COOH-terminal 3-hydroxyacyl-CoA dehydrogenase, result in a unique neuromuscular phenotype.


Subject(s)
Hereditary Sensory and Motor Neuropathy/genetics , Mitochondrial Myopathies/genetics , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Mutation , Adolescent , Child , Chronic Disease , Exons , Fatty Acids/metabolism , Genotype , Heterozygote , Homozygote , Humans , Male , Mitochondrial Trifunctional Protein , Pedigree , Phenotype , Polymorphism, Single-Stranded Conformational , RNA, Messenger/genetics , Sequence Analysis, DNA
13.
Mol Cell Biol ; 21(13): 4321-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390660

ABSTRACT

We have characterized the role of YPR128cp, the orthologue of human PMP34, in fatty acid metabolism and peroxisomal proliferation in Saccharomyces cerevisiae. YPR128cp belongs to the mitochondrial carrier family (MCF) of solute transporters and is localized in the peroxisomal membrane. Disruption of the YPR128c gene results in impaired growth of the yeast with the medium-chain fatty acid (MCFA) laurate as a single carbon source, whereas normal growth was observed with the long-chain fatty acid (LCFA) oleate. MCFA but not LCFA beta-oxidation activity was markedly reduced in intact ypr128cDelta mutant cells compared to intact wild-type cells, but comparable activities were found in the corresponding lysates. These results imply that a transport step specific for MCFA beta-oxidation is impaired in ypr128cDelta cells. Since MCFA beta-oxidation in peroxisomes requires both ATP and CoASH for activation of the MCFAs into their corresponding coenzyme A esters, we studied whether YPR128cp is an ATP carrier. For this purpose we have used firefly luciferase targeted to peroxisomes to measure ATP consumption inside peroxisomes. We show that peroxisomal luciferase activity was strongly reduced in intact ypr128cDelta mutant cells compared to wild-type cells but comparable in lysates of both cell strains. We conclude that YPR128cp most likely mediates the transport of ATP across the peroxisomal membrane.


Subject(s)
Carrier Proteins/metabolism , Fatty Acids/metabolism , Fungal Proteins/metabolism , Nucleotide Transport Proteins , Peroxisomes/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/physiology , Adenosine Triphosphate/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Fractionation , Fungal Proteins/chemistry , Fungal Proteins/genetics , Genes, Reporter/genetics , Glucose/metabolism , Humans , Immunoblotting , Lauric Acids/metabolism , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/chemistry , Mitochondria/metabolism , Oleic Acid/metabolism , Oxidation-Reduction , Peroxisomes/chemistry , Peroxisomes/ultrastructure , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure
14.
Biochim Biophys Acta ; 1138(1): 80-4, 1992 Jan 16.
Article in English | MEDLINE | ID: mdl-1737072

ABSTRACT

In recent years an increasing number of inherited diseases in man have been identified in which there is an impairment in mitochondrial fatty acid oxidation. Diagnosis is usually done by gas-chromatographic analysis of urine, which may give difficulties, since urinary abnormalities may only be present intermittently. We therefore studied whether leukocytes could be used to study mitochondrial beta-oxidation directly. The results described herein show that leukocytes are able to beta-oxidize octanoate and palmitate. Furthermore, clear abnormalities in octanoate beta-oxidation were found in leukocytes from patients with an established deficiency of medium-chain acyl-CoA dehydrogenase, suggesting that measurement of octanoate and palmitate beta-oxidation in leukocytes may contribute to rapid diagnosis of medium-chain acyl-CoA dehydrogenase deficiency and presumably other mitochondrial beta-oxidation disorders.


Subject(s)
Acyl-CoA Dehydrogenases/deficiency , Fatty Acids/metabolism , Leukocytes/metabolism , Lipid Metabolism, Inborn Errors/enzymology , 3-Hydroxybutyric Acid , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenases/urine , Caprylates/metabolism , Humans , Hydroxybutyrates/metabolism , Leukocytes/enzymology , Lipid Metabolism, Inborn Errors/diagnosis , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Palmitates/metabolism , Time Factors
15.
Biochim Biophys Acta ; 1215(3): 347-50, 1994 Dec 08.
Article in English | MEDLINE | ID: mdl-7811722

ABSTRACT

Mitochondrial trifunctional protein is a newly identified enzyme involved in mitochondrial fatty acid beta-oxidation harbouring long-chain enoyl-CoA hydratase, long-chain 3-hydroxyacyl-CoA dehydrogenase and long-chain 3-ketothiolase activity. Over the last few years, we identified more than 26 patients with a deficiency in long-chain 3-hydroxyacyl-CoA dehydrogenase. In order to identify the molecular basis for the deficiency found in these patients, we sequenced the cDNAs encoding the alpha- and beta-subunits which revealed one G-->C mutation at nucleotide position 1528 in the 3-hydroxyacyl-CoA dehydrogenase encoding region of the alpha-subunit. The single base change results in the substitution of a glutamate for a glutamine at amino acid position 510. The base substitution creates a PstI restriction site. Using RFLP, we found that in 24 out of 26 unrelated patients only the C1528 was expressed. The other two patients were heterozygous for this mutation. This mutation was not found in 55 different control subjects. This indicates a high frequency for this mutation in long-chain 3-hydroxyacyl-CoA dehydrogenase deficient patients.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/deficiency , Mitochondria/enzymology , 3-Hydroxyacyl CoA Dehydrogenases/genetics , Amino Acid Sequence , Base Sequence , DNA, Complementary/chemistry , Fatty Acids/metabolism , Humans , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase , Molecular Sequence Data , Mutation , Polymorphism, Restriction Fragment Length , Sequence Alignment
16.
Biochim Biophys Acta ; 1393(1): 35-40, 1998 Jul 31.
Article in English | MEDLINE | ID: mdl-9714723

ABSTRACT

Oxidation of straight-chain fatty acids in mitochondria involves the complicated interaction between a large variety of different enzymes. So far four different mitochondrial straight-chain acyl-CoA dehydrogenases have been identified. The physiological function of three of the four acyl-CoA dehydrogenases has been resolved in recent years especially from studies on patients suffering from certain inborn errors of mitochondrial fatty acid beta-oxidation. The physiological role of long-chain acyl-CoA dehydrogenase (LCAD) has remained obscure, however. The results described in this paper provide strong evidence suggesting that LCAD plays a central role in branched-chain fatty acid metabolism since it turns out to be the major acyl-CoA dehydrogenase reacting with 2,6-dimethylheptanoyl-CoA, a metabolite of pristanic acid, which itself is the alpha-oxidation product of phytanic acid.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Coenzyme A/metabolism , Fatty Acids/metabolism , Oxidoreductases Acting on CH-CH Group Donors , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Cells, Cultured , Humans , Mitochondria/metabolism , Oxidation-Reduction , Oxidoreductases/metabolism , Substrate Specificity
17.
Biochim Biophys Acta ; 1180(1): 28-32, 1992 Oct 13.
Article in English | MEDLINE | ID: mdl-1390941

ABSTRACT

In this paper, we present a new method for measurement of long-chain acyl-CoA dehydrogenase (LCAD) activities in cultured skin fibroblasts. The method is based upon gas chromatographic/mass spectrometric determination of 3-OH-hexadecanoic acid formed during incubation of fibroblasts in a medium containing palmitoyl-CoA and crotonase, to convert the enoyl-CoA ester produced into the 3-hydroxyacyl-CoA ester. The validity of the method is demonstrated by the finding of a full deficiency of LCAD in fibroblasts from three patients with an established deficiency of LCAD.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/analysis , Gas Chromatography-Mass Spectrometry/methods , Skin/enzymology , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Cells, Cultured , Enoyl-CoA Hydratase/metabolism , Fibroblasts/enzymology , Humans , Isotopes , Palmitoyl Coenzyme A/metabolism , Skin/cytology
18.
Biochim Biophys Acta ; 1272(1): 14-20, 1995 Aug 15.
Article in English | MEDLINE | ID: mdl-7662716

ABSTRACT

Long-chain fatty acid oxidation deficient patients present early in life with more severe features than patients with a medium-chain fatty acid oxidation deficiency. This may be related to the more toxic effect of long-chain fatty acid derivatives. In this paper we have studied the effect of different acyl-CoA esters, and palmitoyl-CoA in particular, on succinate-driven oxidative phosphorylation, using digitonin permeabilized human fibroblasts. Palmitoyl-CoA was found to inhibit the succinate-driven oxidative phosphorylation in a concentration dependent manner. If the inhibition of the oxidative phosphorylation system is also expressed under in vivo conditions this might explain some of the abnormalities found in patients with defects in long-chain fatty acid beta-oxidation.


Subject(s)
Fatty Acids/metabolism , Fibroblasts/metabolism , Oxidative Phosphorylation/drug effects , Palmitoyl Coenzyme A/pharmacology , 2,4-Dinitrophenol , Acyl Coenzyme A/pharmacology , Adenosine Triphosphate/biosynthesis , Cell Membrane Permeability , Cells, Cultured , Citrate (si)-Synthase/metabolism , Digitonin , Dinitrophenols/pharmacology , Humans , Lipid Metabolism, Inborn Errors/metabolism , Octoxynol , Oxidation-Reduction , Succinates , Succinic Acid , Uncoupling Agents/pharmacology
19.
Cell Biochem Biophys ; 32 Spring: 1-8, 2000.
Article in English | MEDLINE | ID: mdl-11330035

ABSTRACT

The biogenesis of peroxisomes involves the synthesis of new proteins that after, completion of translation, are targeted to the organelle by virtue of peroxisomal targeting signals (PTS). Two types of PTSs have been well characterized for import of matrix proteins (PTS1 and PTS2). Induction of the genes encoding these matrix proteins takes place in oleate-containing medium and is mediated via an oleate response element (ORE) present in the region preceding these genes. The authors have searched the yeast genome for OREs preceding open reading frames (ORFs), and for ORFs that contain either a PTS1 or PTS2. Of the ORFs containing an ORE, as well as either a PTS1 or a PTS2, many were known to encode bona fide peroxisomal matrix proteins. In addition, candidate genes were identified as encoding putative new peroxisomal proteins. For one case, subcellular location studies validated the in silicio prediction. This gene encodes a new peroxisomal thioesterase.


Subject(s)
Genome, Fungal , Peroxisomes/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA
20.
J Neurol Sci ; 183(1): 61-7, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11166796

ABSTRACT

The Sjögren-Larsson Syndrome (SLS) is a neurocutaneous disorder, caused by deficient activity of the microsomal enzyme fatty aldehyde dehydrogenase (FALDH). FALDH catalyzes the oxidation of medium- and long-chain fatty aldehydes to their corresponding carboxylic acids. SLS is diagnosed by demonstrating the enzyme deficiency or by mutation analysis of the FALDH gene, while laboratory investigations of plasma, urine, and cerebrospinal fluid do not reveal any diagnostic abnormality. Leukotriene (LT) B4 is a pro-inflammatory mediator synthesized from arachidonic acid. LTB4 is inactivated by microsomal omega-oxidation, successively yielding 20-OH-LTB4, 20-CHO-LTB4 and 20-COOH-LTB4. Since FALDH is involved in LTB4 degradation, we have analyzed LTB4 and its metabolites in urine and cerebrospinal fluid as well as the degradation capacity for LTB4 in fresh polymorphonuclear leukocytes (PMN) of SLS patients. The urinary concentrations of LTB4, 20-OH-LTB4 and 20-COOH-LTB4 are below the detection limit in healthy controls. The urine of all SLS patients (n=13) exhibited highly elevated concentrations of LTB4 and 20-OH-LTB4, while 20-COOH-LTB4 was absent. Cerebrospinal fluid levels of LTB4, 20-OH-LTB4 and 20-COOH-LTB4 were found to be normal (n=7). PMN isolated from four patients were shown to be unable to convert 20-OH-LTB4 to 20-COOH-LTB4. Our findings provide unambiguous evidence for defective LTB4 degradation in SLS patients, and offer new and non-invasive diagnostic tools. Moreover, they open new pathophysiological considerations, with the prospect of rational treatment strategies.


Subject(s)
Alcohol Oxidoreductases/metabolism , Leukotriene B4/metabolism , Sjogren-Larsson Syndrome/enzymology , Adolescent , Arachidonic Acid/metabolism , Child , Child, Preschool , Humans , Hydroxyeicosatetraenoic Acids , Leukotriene B4/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL