Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Clin Genet ; 86(2): 142-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23889335

ABSTRACT

Peters plus syndrome (PPS) is a rare autosomal-recessive disorder characterized by Peters anomaly of the eye, short stature, brachydactyly, dysmorphic facial features, developmental delay, and variable other systemic abnormalities. In this report, we describe screening of 64 patients affected with PPS, isolated Peters anomaly and PPS-like phenotypes. Mutations in the coding region of B3GALTL were identified in nine patients; six had a documented phenotype of classic PPS and the remaining three had a clinical diagnosis of PPS with incomplete clinical documentation. A total of nine different pathogenic alleles were identified. Five alleles are novel including one frameshift, c.168dupA, p.(Gly57Argfs*11), one nonsense, c.1234C>T, p.(Arg412*), two missense, c.1045G>A, p.(Asp349Asn) and c.1181G>A, p.(Gly394Glu), and one splicing, c.347+5G>T, mutations. Consistent with previous reports, the c.660+1G>A mutation was the most common mutation identified, seen in eight of the nine patients and accounting for 55% of pathogenic alleles in this study and 69% of all reported pathogenic alleles; while two patients were homozygous for this mutation, the majority had a second rare pathogenic allele. We also report the absence of B3GALTL mutations in 55 cases of PPS-like phenotypes or isolated Peters anomaly, further establishing the strong association of B3GALTL mutations with classic PPS only.


Subject(s)
Cleft Lip/genetics , Cornea/abnormalities , Galactosyltransferases/genetics , Glucosyltransferases/genetics , Growth Disorders/genetics , Limb Deformities, Congenital/genetics , Mutation/genetics , Cohort Studies , Female , Gene Frequency/genetics , Genetic Association Studies , Humans , Male , Phenotype
2.
Hum Mol Genet ; 10(19): 2069-77, 2001 Sep 15.
Article in English | MEDLINE | ID: mdl-11590124

ABSTRACT

Although many patients have been found to have very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency, none have been documented with long-chain acyl-CoA dehydrogenase (LCAD) deficiency. In order to understand the metabolic pathogenesis of long-chain fatty acid oxidation disorders, we generated mice with VLCAD deficiency (VLCAD(-/-)) and compared their pathologic and biochemical phenotypes of mice with LCAD deficiency (LCAD(-/-)) and wild-type mice. VLCAD(-/-) mice had milder fatty change in liver and heart. Dehydrogenation of various acyl-CoA substrates by liver, heart and skeletal muscle mitochondria differed among the three genotypes. The results for liver were most informative as VLCAD(-/-) mice had a reduction in activity toward palmitoyl-CoA and oleoyl-CoA (58 and 64% of wild-type, respectively), whereas LCAD(-/-) mice showed a more profoundly reduced activity toward these substrates (35 and 32% of wild-type, respectively), with a significant reduction of activity toward the branched chain substrate 2,6-dimethylheptanoyl-CoA. C(16) and C(18) acylcarnitines were elevated in bile, blood and serum of fasted VLCAD(-/-) mice, whereas abnormally elevated C(12) and C(14) acylcarnitines were prominent in LCAD(-/-) mice. Progeny with the combined LCAD(+/+)//VLCAD(+/-) genotype were over-represented in offspring from sires and dams heterozygous for both LCAD and VLCAD mutations. In contrast, no live mice with a compound LCAD(-/-)//VLCAD(-/-) genotype were detected.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Carnitine/analogs & derivatives , Lipid Metabolism, Inborn Errors/enzymology , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Carnitine/metabolism , Fatty Acids/metabolism , Female , Humans , Lipid Metabolism, Inborn Errors/pathology , Litter Size , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/enzymology , Mitochondria, Liver/enzymology , Mitochondria, Muscle/enzymology
3.
Pediatr Res ; 49(1): 18-23, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134486

ABSTRACT

ABSTRACT Short-chain acyl-CoA dehydrogenase (SCAD) deficiency is considered a rare inherited mitochondrial fatty acid oxidation disorder. Less than 10 patients have been reported, diagnosed on the basis of ethylmalonic aciduria and low SCAD activity in cultured fibroblast. However, mild ethylmalonic aciduria, a biochemical marker of functional SCAD deficiency in vivo, is a common finding in patients suspected of having metabolic disorders. Based on previous observations, we have proposed that ethylmalonic aciduria in a small proportion of cases is caused by pathogenic SCAD gene mutations, and SCAD deficiency can be demonstrated in fibroblasts. Another - much more frequent - group of patients with mild ethylmalonic aciduria has functional SCAD deficiency due to the presence of susceptibility SCAD gene variations, i.e. 625G>A and 511C>T, in whom a variable or moderately reduced SCAD activity in fibroblasts may still be clinically relevant. To substantiate this notion we performed sequence analysis of the SCAD gene in 10 patients with ethylmalonic aciduria and diagnosed with SCAD deficiency in fibroblasts. Surprisingly, only one of the 10 patients carried pathogenic mutations in both alleles, while five were double heterozygotes for a pathogenic mutation in one allele and the 625G>A susceptibility variation in the other. The remaining four patients carried only either the 511C>T or the 625G>A variations in each allele. Our findings document that patients carrying these SCAD gene variations may develop clinically relevant SCAD deficiency, and that patients with even mild ethylmalonic aciduria should be tested for these variations.


Subject(s)
Acyl-CoA Dehydrogenases/genetics , Genetic Variation , Lipid Metabolism, Inborn Errors/genetics , Acyl-CoA Dehydrogenase , DNA, Complementary , Female , Haplotypes , Humans , Infant , Infant, Newborn , Male , Mutagenesis, Site-Directed , Mutation
5.
Am J Obstet Gynecol ; 180(4): 986-94, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10203668

ABSTRACT

OBJECTIVES: The aims of this article are to report on a review of cases of maternal phenylketonuria in the International Maternal Phenylketonuria Collaborative Study that were initially diagnosed during or after a pregnancy, to alert health care practitioners to the possible existence of women with undiagnosed phenylketonuria whose fetuses are at risk, and to emphasize that not all adults with untreated phenylketonuria are mentally retarded. STUDY DESIGN: The study was conducted through retrospective database review. RESULTS: Of 414 women with live-born infants, 17 fulfilled our criteria. Six had phenylketonuria diagnosed after they had produced >/=1 affected offspring, 2 had phenylketonuria diagnosed as a result of transient postnatal hyperphenylalaninemia in an offspring, and 9 had phenylketonuria diagnosed by prenatal screening. Undiagnosed maternal phenylketonuria in North America and Europe is currently estimated at 1 case/100,000 births; this rate could be higher elsewhere. CONCLUSIONS: Physicians and midwives should consider a protocol of selective prenatal screening or case finding to detect undiagnosed phenylketonuria among their patients.


Subject(s)
Fetal Diseases/diagnosis , Phenylketonuria, Maternal/diagnosis , Prenatal Diagnosis , Adolescent , Adult , Female , Humans , Phenylalanine/blood , Pregnancy , Pregnancy Outcome , Retrospective Studies
6.
Neurology ; 52(2): 366-72, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9932958

ABSTRACT

OBJECTIVE: To determine an underlying genetic defect within the differential diagnosis of congenital multicore myopathy. BACKGROUND: A 13.5-year-old girl presented with congenital-onset facial and neck weakness, slowly progressive severe limb girdle and axial myopathy, respiratory weakness, cardiomyopathy, progressive joint contractures, lumbar lordosis, progressive external ophthalmoplegia with ptosis, and cataracts. Muscle biopsy at 3 years revealed type I fiber predominance and hypotrophy, multicores with a focal decrease in mitochondria and oxidative enzymes, and internal nuclei. METHODS AND RESULTS: Serum carnitine was decreased (total, 18.2 micromol/L; free, 11.7 micromol/L). Urine organic acids intermittently revealed very large amounts of ethylmalonic and methylsuccinic acids intermittently, with elevated butyrylglycine, 2-methylbutyrylglycine, and tiglylglycine. Fibroblast acylcarnitine profiles revealed marked butyrylcarnitine elevation. Electron-transferring flavoprotein-linked reduction enzymatic assay of fibroblasts with butyryl-coenzyme A (CoA) as substrate, after immunoinactivation of medium-chain acyl-CoA dehydrogenase activity, revealed a complete absence of short-chain acyl-CoA dehydrogenase (SCAD) activity. No SCAD protein was detectable with Western blot analysis. CONCLUSIONS: This patient expands the clinical phenotype of SCAD deficiency and emphasizes the need for its consideration in the differential diagnosis of progressive external ophthalmoplegia and congenital multicore myopathy.


Subject(s)
Acyl-CoA Dehydrogenases/deficiency , Mitochondrial Myopathies/diagnosis , Ophthalmoplegia, Chronic Progressive External/diagnosis , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenases/genetics , Adolescent , Carnitine/analogs & derivatives , Carnitine/blood , Cells, Cultured , Diagnosis, Differential , Female , Fibroblasts/enzymology , Humans , Mitochondrial Myopathies/enzymology , Mitochondrial Myopathies/genetics , Ophthalmoplegia, Chronic Progressive External/enzymology , Ophthalmoplegia, Chronic Progressive External/genetics , Phenotype
7.
Adv Exp Med Biol ; 466: 395-402, 1999.
Article in English | MEDLINE | ID: mdl-10709668

ABSTRACT

The SCAD deficient mouse model has been useful to investigate mechanisms of deficient fatty acid oxidation disease in human patients. This mouse model has been thoroughly characterized and is readily available from the Jackson Laboratory. Using the new technologies of gene-knockout mouse modeling, we envisage developing additional members of the acyl-CoA dehydrogenase family of enzyme deficiencies in mice and furthering our understanding of fatty acid metabolism in health and disease.


Subject(s)
Acyl-CoA Dehydrogenases/deficiency , Acyl-CoA Dehydrogenases/genetics , Lipid Metabolism, Inborn Errors/enzymology , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenases/metabolism , Animals , Disease Models, Animal , Exons , Humans , Introns , Lipid Metabolism, Inborn Errors/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Sequence Deletion
8.
Proc Natl Acad Sci U S A ; 95(26): 15592-7, 1998 Dec 22.
Article in English | MEDLINE | ID: mdl-9861014

ABSTRACT

Abnormalities of fatty acid metabolism are recognized to play a significant role in human disease, but the mechanisms remain poorly understood. Long-chain acyl-CoA dehydrogenase (LCAD) catalyzes the initial step in mitochondrial fatty acid oxidation (FAO). We produced a mouse model of LCAD deficiency with severely impaired FAO. Matings between LCAD +/- mice yielded an abnormally low number of LCAD +/- and -/- offspring, indicating frequent gestational loss. LCAD -/- mice that reached birth appeared normal, but had severely reduced fasting tolerance with hepatic and cardiac lipidosis, hypoglycemia, elevated serum free fatty acids, and nonketotic dicarboxylic aciduria. Approximately 10% of adult LCAD -/- males developed cardiomyopathy, and sudden death was observed in 4 of 75 LCAD -/- mice. These results demonstrate the crucial roles of mitochondrial FAO and LCAD in vivo.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Humans , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Mitochondria, Liver/enzymology , Muscle, Skeletal/metabolism , Substrate Specificity
9.
J Pediatr ; 133(5): 697-700, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9821433

ABSTRACT

We report siblings with a variant of carbohydrate-deficient glycoprotein syndrome, type 1 (CDGS1), characterized by normal phosphomannomutase and phosphomannose isomerase activities, severe thrombocytopenia, and respiratory compromise. Each infant died after a course of intensive care, suggesting that infants with CDGS1 and normal phosphomannomutase and phosphomannose isomerase activities may have a more severe CDGS1 phenotype.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Infant, Premature, Diseases/diagnosis , Mannose-6-Phosphate Isomerase/blood , Phosphotransferases (Phosphomutases)/blood , Thrombocytopenia/diagnosis , Congenital Disorders of Glycosylation/enzymology , Congenital Disorders of Glycosylation/genetics , Fatal Outcome , Female , Humans , Infant, Newborn , Infant, Premature, Diseases/enzymology , Infant, Premature, Diseases/genetics , Male , Pregnancy , Prenatal Diagnosis , Thrombocytopenia/enzymology , Thrombocytopenia/genetics
10.
Pediatr Res ; 44(2): 210-4, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9702916

ABSTRACT

Acylcarnitines are important diagnostic markers for inborn errors of fatty acid oxidation, but their analysis in body fluids may not always be reliable. Recently, disease-specific acylcarnitine profiles generated by cultured skin fibroblasts were reported to facilitate the diagnosis by localizing a specific enzymatic defect in the mitochondrial beta-oxidation pathway. Using a novel methodologic approach, fibroblasts from 16 patients with inborn errors of fatty acid oxidation and 13 control subjects were preincubated with L-[3H]carnitine to label the intracellular carnitine pool. Cells were subsequently incubated with unlabeled palmitic acid and, after methanol extraction of cells and media, labeled free carnitine and acylcarnitines were analyzed by radio-HPLC. Quantitation was based on the integrated radioactivity of individual peaks relative to the total radioactivity recovered. In control cell lines, all saturated acylcarnitines were detected, and reference values were established. With the exception of one cell line deficient in electron transfer flavoprotein, all mutant cell lines showed abnormal and disease-specific relative concentrations of acylcarnitines. Advantages of the method include use of a small number of cells, no need for trypsinization and permeabilization of cells before incubation, simple extraction without purification of the specimen before HPLC, and relatively inexpensive equipment. The method allows a focused approach to the subsequent, more laborious confirmation of a particular disease by direct enzymatic and/or molecular analysis. It remains to be established whether the method can replace widely used global measurements of fatty acid oxidation rates in vitro that do not provide specific information about the enzyme deficiency involved.


Subject(s)
Carnitine/analysis , Chromatography, High Pressure Liquid/methods , Fatty Acids/metabolism , Mitochondria/metabolism , Oxidoreductases Acting on CH-CH Group Donors , Skin/chemistry , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenase, Long-Chain , Carnitine O-Palmitoyltransferase/metabolism , Cells, Cultured , Fatty Acid Desaturases/metabolism , Fibroblasts/chemistry , Humans , Lipid Metabolism, Inborn Errors/metabolism , Mitochondria/enzymology , Oxidation-Reduction , Oxidoreductases/metabolism , Palmitic Acid/metabolism , Skin/cytology
11.
Hum Mol Genet ; 6(9): 1451-5, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9285781

ABSTRACT

We report the therapeutic effects of liver-specific expression of a short-chain acyl-CoA dehydrogenase (SCAD) transgene in the SCAD-deficient mouse model. Transgenic mice were produced with a rat albumin promoter/enhancer driving a mouse SCAD minigene (ALB-SCAD) on both the SCAD normal genetic background and a SCAD-deficient background. In three transgenic lines produced on the SCAD-deficient background, recombinant SCAD activity and antigen in liver mitochondria were found up to 7-fold of normal control values. All three lines showed a markedly reduced organic aciduria and fatty liver, which are sensitive indicators of the metabolic abnormality seen in this disease found in children. We found no detrimental effects of high liver SCAD expression in transgenic mice on either background. These studies provide important basic and practical therapeutic information for the potential gene therapy of nuclear-encoded mitochondrial enzyme deficiencies, as well as insights into the mechanisms of the disease.


Subject(s)
Acyl-CoA Dehydrogenases/deficiency , Acyl-CoA Dehydrogenases/genetics , Genetic Therapy , Mitochondria, Liver/enzymology , Acyl-CoA Dehydrogenase , Animals , DNA, Mitochondrial/analysis , Gene Transfer Techniques , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism
12.
Nat Genet ; 14(3): 361-5, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8896573

ABSTRACT

5-Oxoprolinuria (pyroglutamic aciduria) resulting from glutathione synthetase (GSS) deficiency is an inherited autosomal recessive disorder characterized, in its severe form, by massive urinary excretion of 5-oxoproline, metabolic acidosis, haemolytic anaemia and central nervous system damage. The metabolic defect results in low GSH levels presumably with feedback over-stimulation of gamma-glutamylcysteine synthesis and its subsequent conversion to 5-oxoproline. In this study, we cloned and characterized the human GSS gene and examined three families with four cases of well-documented 5-oxoprolinuria. We identified seven mutations at the GSS locus on six alleles: one splice site mutation, two deletions and four missense mutations. Bacterial expression and yeast complementation assays of the cDNAs encoded by these alleles demonstrated their functional defects. We also characterized a fifth case, an homozygous missense mutation in the gene in an individual affected by a milder-form of the GSS deficiency, which is apparently restricted to erythrocytes and only associated with haemolytic anaemia. Our data provide the first molecular genetic analysis of 5-oxoprolinuria and demonstrate that GSS deficiency with oxoprolinuria and GSS deficiency without 5-oxoprolinuria are caused by mutations in the same gene.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Glutathione Synthase/genetics , Mutation , Pyrrolidonecarboxylic Acid/metabolism , Amino Acid Metabolism, Inborn Errors/complications , Anemia/complications , Anemia/genetics , Binding Sites , Erythrocytes/pathology , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Genetic Complementation Test , Glutathione Synthase/metabolism , Heterozygote , Humans , Male , Molecular Sequence Data , Pedigree , Polymerase Chain Reaction , RNA Splicing , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Sequence Analysis, DNA
13.
Am J Hum Genet ; 57(2): 273-83, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7668252

ABSTRACT

Two overlapping cDNA clones (1,991 bp and 736 bp, respectively) encoding the precursor of human mitochondrial very-long-chain acyl-coenzyme A dehydrogenase (VLCAD) were cloned and sequenced. The cDNA inserts of these clones together encompass a region of 2,177 bases, encoding the entire protein of 655 amino acids, including a 40-amino acid leader peptide and a 615-amino acid mature polypeptide. PCR-amplified VLCAD cDNAs were sequenced in cultured fibroblasts from two VLCAD-deficient patients. In both patients, a 105-bp deletion encompassing bases 1078-1182 in VLCAD cDNA was identified. The deletion seems to occur due to exon skipping during processing of VLCAD pre-mRNA. This is the first demonstration of a mutation causing VLCAD deficiency. Quantitative cDNA expression of normal human VLCAD was performed in the patients' fibroblasts, using vaccinia viral system, which demonstrated that the deficiency of the normal VLCAD protein causes impaired long-chain fatty acid beta-oxidation activity in the patients' fibroblasts. In patient fibroblasts, raising VLCAD activity to approximately 20% of normal control fibroblast activity raised palmitic acid beta-oxidation flux to the level found in control fibroblasts, which may offer important information for the rational design of future somatic gene therapy for VLCAD deficiency.


Subject(s)
Fatty Acid Desaturases/deficiency , Fatty Acid Desaturases/genetics , Lipid Metabolism, Inborn Errors/genetics , Acyl-CoA Dehydrogenase, Long-Chain , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Fibroblasts/metabolism , Humans , Molecular Sequence Data , Mutation , Polymerase Chain Reaction
14.
J Clin Invest ; 95(6): 2465-73, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7769092

ABSTRACT

Mitochondrial very-long-chain acyl-coenzyme A dehydrogenase (VLCAD) was purified from human liver. The molecular masses of the native enzyme and the subunit were estimated to be 154 and 70 kD, respectively. The enzyme was found to catalyze the major part of mitochondrial palmitoylcoenzyme A dehydrogenation in liver, heart, skeletal muscle, and skin fibroblasts (89-97, 86-99, 96-99, and 78-87%, respectively). Skin fibroblasts from 26 patients suspected of having a disorder of mitochondrial beta-oxidation were analyzed for VLCAD protein using immunoblotting, and 7 of them contained undetectable or trace levels of the enzyme. The seven deficient fibroblast lines were characterized by measuring acyl-coenzyme A dehydrogenation activities, overall palmitic acid oxidation, and VLCAD protein synthesis using pulse-chase, further confirming the diagnosis of VLCAD deficiency. These results suggested the heterogenous nature of the mutations causing the deficiency in the seven patients. Clinically, all patients with VLCAD deficiency exhibited cardiac disease. At least four of them presented with hypertrophic cardiomyopathy. This frequency (> 57%) was much higher than that observed in patients with other disorders of mitochondrial long-chain fatty acid oxidation that may be accompanied by cardiac disease in infants.


Subject(s)
Fatty Acid Desaturases/isolation & purification , Lipid Metabolism, Inborn Errors/enzymology , Mitochondria, Liver/enzymology , Acyl Coenzyme A/metabolism , Acyl-CoA Dehydrogenase, Long-Chain , Blotting, Western , Fatty Acid Desaturases/metabolism , Female , Fibroblasts , Humans , Male , Mitochondria, Heart/enzymology , Molecular Weight , Palmitates/metabolism
15.
Article in English | MEDLINE | ID: mdl-7540307

ABSTRACT

A new radiolabeled metabolite was released into the extracellular fluid by normal human skin fibroblasts that were labeled with [5,6,8,9,11,12,14,15-3H] arachidonic acid. This product continued to accumulate during a 24 h incubation, and its formation was not saturated at arachidonic acid concentrations up to 15 mumol/L. The compound, identified as hexadecatrienoic acid, was not produced by Zellweger fibroblasts which are deficient in peroxisomal fatty acid beta-oxidation. By contrast, radiolabeled hexadecatrienoic acid was produced by mutant fibroblasts having other peroxisomal defects, including X-linked adrenoleukodystrophy, adult Refsum's disease, and rhizomelic chondrodysplasia punctata. This radiolabeled metabolite also was produced by mutant fibroblasts that cannot oxidize long-chain fatty acids in the mitochondria. These results indicate that hexadecatrienoic acid is synthesized from arachidonic acid by peroxisomal beta-oxidation. The absence of this pathway may account for some of the biochemical and functional abnormalities that occur in Zellweger's syndrome.


Subject(s)
Arachidonic Acid/metabolism , Fatty Acids, Unsaturated/metabolism , Microbodies/metabolism , Adrenoleukodystrophy/metabolism , Cells, Cultured , Chondrodysplasia Punctata/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Humans , Mitochondria/metabolism , Oxidation-Reduction , Refsum Disease/metabolism , Time Factors , Zellweger Syndrome/metabolism
16.
J Biol Chem ; 270(2): 530-5, 1995 Jan 13.
Article in English | MEDLINE | ID: mdl-7822275

ABSTRACT

The accumulation of beta-oxidation intermediates was studied by incubating normal and beta-oxidation enzyme-deficient human fibroblasts with [2H4]linoleate and L-carnitine and analyzing the resultant acylcarnitines by tandem mass spectrometry. Labeled decenoyl-, octanoyl-, hexanoyl-, and butyrylcarnitines were the only intermediates observed with normal cells. Intermediates of longer chain length, corresponding to substrates for the beta-oxidation enzymes associated with the inner mitochondrial membrane, were not observed unless a cell line was deficient in one of these enzymes, such as very-long-chain acyl-CoA dehydrogenase, long-chain 3-hydroxyacyl-CoA dehydrogenase, or electron transfer flavoprotein dehydrogenase. Matrix enzyme deficiencies, such as medium- and short-chain acyl-CoA dehydrogenases, were characterized by elevated concentrations of intermediates corresponding to their respective substrates (octanoyl- and decenoylcarnitines in medium-chain acyl-CoA dehydrogenase deficiency and butyrylcarnitine in short-chain acyl-CoA dehydrogenase deficiency). These observations agree with the notion of intermediate channeling due to the organization of beta-oxidation enzymes in complexes. The only exception is the incomplete channeling from thiolase to acyl-CoA dehydrogenase in the matrix. This situation may be a consequence of only one 3-ketoacyl-CoA thiolase being unable to interact with the several acyl-CoA dehydrogenases in the matrix.


Subject(s)
Mitochondria/metabolism , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenase, Long-Chain , Cells, Cultured , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Linoleic Acid , Linoleic Acids/metabolism , Oxidation-Reduction , Palmitic Acid , Palmitic Acids/metabolism
17.
J Pediatr ; 123(3): 427-30, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8355121

ABSTRACT

A comatose male newborn infant with congenital lactic acidosis caused by pyruvate decarboxylase deficiency was treated with dichloroacetate (DCA), which stimulated an 88% drop in serum lactate concentration and reversed his coma. The response to DCA was temporary and the lactic acidosis worsened until his death, but DCA may confer more lasting benefit in less severely affected infants.


Subject(s)
Acidosis, Lactic/congenital , Acidosis, Lactic/drug therapy , Dichloroacetic Acid/therapeutic use , Pyruvate Decarboxylase/deficiency , Pyruvate Metabolism, Inborn Errors/drug therapy , Acidosis, Lactic/etiology , Humans , Infant, Newborn , Male , Pyruvate Metabolism, Inborn Errors/complications
18.
Biochem Biophys Res Commun ; 191(3): 1369-72, 1993 Mar 31.
Article in English | MEDLINE | ID: mdl-8466512

ABSTRACT

Palmitoyl-CoA dehydrogenase activity in skin fibroblasts from seven patients with unidentified defects of fatty acid oxidation was measured in the presence and absence of antibodies against medium-chain, long-chain, and very-long-chain acyl-CoA dehydrogenases (VLCAD). Two of the patients, 4-5 month old boys, were found to have a novel disease, VLCAD deficiency, as judged from the results of very low palmitoyl-CoA dehydrogenase activity and the lack of immunoreactivity toward antibody raised to purified VLCAD.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Lipid Metabolism, Inborn Errors/enzymology , Mitochondria/enzymology , Female , Humans , Male
19.
J Clin Invest ; 90(5): 1679-86, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1430199

ABSTRACT

We studied metabolic, polypeptide and genetic variation in eight glutaric acidemia type II (GA II) patients with electron transfer flavoprotein (ETF) deficiency. As measured by 3H-fatty acid oxidations in fibroblasts, beta-oxidation pathway flux correlated well with clinical phenotypes. In six patients with severe neonatal onset GA II, oxidation of [9,10(n)-3H]-palmitate ranged from 2% to 22% of control and of [9,10(n)-3H]myristate, from 2% to 26% of control. Of two patients with late onset GA II, one had intermediate residual activities with these substrates and the other normal activities. Radiolabeling and immunoprecipitation studies revealed that three of the six neonatal onset GA II patients had greatly diminished or absent alpha- and beta-ETF subunits, consistent with a failure to assemble a stable heterodimer. Another neonatal onset patient showed normal synthesis of beta-ETF but decreased synthesis of alpha-ETF. Two neonatal onset and two late onset GA II patients showed normal synthesis of both subunits. Analysis of the pre-alpha-ETF coding sequence revealed seven different mutations in the six patients with neonatal onset GA II. The most common mutation was a methionine for threonine substitution at codon 266 found in four unrelated patients, while all the other mutations were seen in single patients. No mutations were detected in the two patients with late onset GA II.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , DNA/genetics , Fatty Acids/metabolism , Flavoproteins/genetics , Glutarates/blood , Lipid Metabolism, Inborn Errors/metabolism , Peptide Biosynthesis , Amino Acid Metabolism, Inborn Errors/genetics , Base Sequence , Cells, Cultured , Electron-Transferring Flavoproteins , Humans , Lipid Metabolism, Inborn Errors/genetics , Molecular Sequence Data , Mutation , Oxidation-Reduction , Polymerase Chain Reaction
20.
Pediatr Res ; 31(6): 552-6, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1635815

ABSTRACT

BALB/cByJ (J) mice have short-chain acyl-CoA dehydrogenase (SCAD) deficiency and an organic aciduria similar to that of human SCAD deficiency. [9,10(n)-3H]- and [15,16(n)-3H]palmitate oxidations in J mouse fibroblasts were 96 and 35% of control, respectively, consistent with an isolated SCAD defect. Acyl-CoA dehydrogenase activities were assayed in muscle and fibroblast mitochondria from BALB/cBy controls (Y) and SCAD-deficient J mice. Medium-chain acyl-CoA dehydrogenase (MCAD) activities were comparable in both J and Y mice from all tissues. In the presence of MCAD antiserum, SCAD activities in J mice were undetectable in both tissues. Apparent Km and Vmax values in liver mitochondria suggested a somewhat increased affinity of MCAD for butyryl-CoA in J mice, as compared with MCAD from other species. Immunoblot studies using mitochondria revealed identical apparent SCAD molecular weight in liver, muscle, and fibroblasts from Y mice and no detectable SCAD antigen in J mice; MCAD antigen was detected in comparable amounts from both Y and J mice. Radiolabeling and immunoprecipitation studies in J mouse fibroblasts revealed no SCAD synthesis, but normal MCAD synthesis. These data argue against the existence of tissue-specific SCAD isoforms in the mouse and confirm that this mouse strain is a model for the human organic aciduria resulting from this beta-oxidation defect.


Subject(s)
Acyl-CoA Dehydrogenases/deficiency , Acyl-CoA Dehydrogenase , Acyl-CoA Dehydrogenases/genetics , Acyl-CoA Dehydrogenases/immunology , Acyl-CoA Dehydrogenases/metabolism , Animals , Antigens/analysis , Fibroblasts/enzymology , Immunochemistry , Kinetics , Liver/enzymology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Muscles/enzymology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...