Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Mol Genet Metab ; 122(3): 67-75, 2017 11.
Article in English | MEDLINE | ID: mdl-28801073

ABSTRACT

BACKGROUND: Carnitine palmitoyltransferase (CPT) II deficiency is one of the most common forms of mitochondrial fatty acid oxidation disorder (FAOD). However, newborn screening (NBS) for this potentially fatal disease has not been established partly because reliable indices are not available. METHODS: We diagnosed CPT II deficiency in a 7-month-old boy presenting with hypoglycemic encephalopathy, which apparently had been missed in the NBS using C16 and C18:1 concentrations as indices. By referring to his acylcarnitine profile from the NBS, we adopted the (C16+C18:1)/C2 ratio (cutoff 0.62) and C16 concentration (cutoff 3.0nmol/mL) as alternative indices for CPT II deficiency such that an analysis of a dried blood specimen collected at postnatal day five retroactively yielded the correct diagnosis. Thereafter, positive cases were assessed by measuring (1) the fatty acid oxidation ability of intact lymphocytes and/or (2) CPT II activity in the lysates of lymphocytes. The diagnoses were then further confirmed by genetic analysis. RESULTS: The disease was diagnosed in seven of 21 newborns suspected of having CPT II deficiency based on NBS. We also analyzed the false-negative patient and five symptomatic patients for comparison. Values for the NBS indices of the false-negative, symptomatic patient were lower than those of the seven affected newborns. Although it was difficult to differentiate the false-negative patient from heterozygous carriers and false-positive subjects, the fatty acid oxidation ability of the lymphocytes and CPT II activity clearly confirmed the diagnosis. Among several other indices proposed previously, C14/C3 completely differentiated the seven NBS-positive patients and the false-negative patient from the heterozygous carriers and the false-positive subjects. Genetic analysis revealed 16 kinds of variant alleles. The most prevalent, detected in ten alleles in nine patients from eight families, was c.1148T>A (p.F383Y), a finding in line with those of several previous reports on Japanese patients. CONCLUSIONS: These findings suggested that CPT II deficiency can be screened by using (C16+C18:1)/C2 and C16 as indices. An appropriate cutoff level is required to achieve adequate sensitivity albeit at the cost of a considerable increase in the false-positive rate, which might be reduced by using additional indices such as C14/C3.


Subject(s)
Carnitine O-Palmitoyltransferase/analysis , Carnitine O-Palmitoyltransferase/deficiency , Metabolism, Inborn Errors/diagnosis , Neonatal Screening , Palmitoylcarnitine/analysis , Alleles , Carnitine O-Palmitoyltransferase/genetics , Dried Blood Spot Testing/methods , False Negative Reactions , False Positive Reactions , Female , Humans , Hypoglycemia/complications , Infant , Infant, Newborn , Male , Metabolism, Inborn Errors/genetics , Sensitivity and Specificity , Tandem Mass Spectrometry
2.
Mol Genet Metab ; 119(4): 322-328, 2016 12.
Article in English | MEDLINE | ID: mdl-27856190

ABSTRACT

BACKGROUND: Medium-chain acyl-CoA dehydrogenase (MCAD) deficiency is a representative disorder of fatty acid oxidation and is one of the most prevalent inborn errors of metabolism among Caucasian populations. In Japan, however, it was as late as 2000 when the first patient was found, and enzymatic and genetic evaluation of MCAD deficiency began. METHODS: We measured octanoyl-CoA dehydrogenase activity in lymphocytes of symptomatic children and newborn screening (NBS)-positive subjects who showed elevated levels of C8-acylcarnitine in blood. The results were further confirmed by direct sequencing of the ACADM gene. RESULTS: The disease was diagnosed in 9 out of 18 symptomatic children. The affected patients showed residual activities from 0% to 3% of the normal average value, except for one patient with 10% activity. Concerning 50 NBS-positive subjects, 18 with enzymatic activities around 10% or lower and 14 with activities ranging from 13% to 30% were judged to be affected patients, and biallelic variants were detected in most of the cases tested. Newborns with higher enzymatic activities were estimated to be heterozygous carriers or healthy subjects, though biallelic variants were detected in 5 of them. Genetic analysis detected 22 kinds of variant alleles. The most prevalent was c.449_452delCTGA (p.T150Rfs), which was followed by c.50G>A (p.R17H), c.1085G>A (p.G362E), c.157C>T (p.R53C), and c.843A>T (p.R281S); these five variants accounted for approximately 60% of all the alleles examined. CONCLUSION: Our study has revealed the unique genetic backgrounds of MCAD deficiency among Japanese, based on the largest series of non-Caucasian cases. A continuous spectrum of severity was also observed in our series of NBS-positive cases, suggesting that it is essential for every nation and ethnic group to accumulate its own information on gene variants, together with their enzymatic evaluation, in order to establish an efficient NBS system for MCAD deficiency.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/genetics , Genetic Testing , Hypoglycemia/genetics , Lipid Metabolism, Inborn Errors/genetics , Neonatal Screening , Acyl-CoA Dehydrogenase/blood , Alleles , Child, Preschool , Female , Genotype , Heterozygote , Humans , Hypoglycemia/diagnosis , Hypoglycemia/epidemiology , Hypoglycemia/physiopathology , Infant , Infant, Newborn , Japan/epidemiology , Lipid Metabolism, Inborn Errors/blood , Lipid Metabolism, Inborn Errors/epidemiology , Lipid Metabolism, Inborn Errors/physiopathology , Male , Mutation , Polymorphism, Single Nucleotide/genetics
3.
Mol Genet Metab ; 118(1): 9-14, 2016 May.
Article in English | MEDLINE | ID: mdl-26947917

ABSTRACT

BACKGROUND: Since the first case was detected in 2000, there has been a remarkable increase in Japanese patients diagnosed with medium-chain acyl-CoA dehydrogenase (MCAD) deficiency. Genetic analysis has revealed a spectrum of mutations that is quite different from those observed in Caucasian populations. In 2014, Japan initiated nationwide newborn screening (NBS) for MCAD using tandem mass spectrometry (MS/MS). It is an urgent issue to assess the risk of acute metabolic decompensation from the respective novel mutations found thus far. METHODS: To evaluate the pathogenic effect of each mutation, we established a eukaryotic cell expression system and prepared 11 mutant proteins identified in five symptomatic patients and eight MS/MS-NBS-positive newborns, as well as two common Caucasian mutations, p.K329E (c.985G>A) and p.Y67H (c.157C>T) for comparison. RESULTS: The expression of four mutant proteins (p.Q45R, p.P92L, p.P128X and p.Y397N) were severely impaired, whereas the others expressed normally, as did p.K329E and p.Y67H. Based on their dehydrogenase activities toward n-octanoyl-CoA, we determined three mutations (p.R53C, p.R281S and p.G362E) to be disease-causing, two mutations having (p.R17H and p.M274V) to be of marginal risk, and two mutations (p.K271E and p.I416T) as benign. Their allele-specific activities were as a whole in accordance with those estimated from the results of measurement in peripheral blood mononuclear cells. CONCLUSION: As most of the mutations detected in the Japanese population are unique, prudent genetic and enzymatic analysis is essential to precisely evaluate the latent risk of clinical onset for screening-positive newborns.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Lipid Metabolism, Inborn Errors/diagnosis , Mutation , Neonatal Screening/methods , Tandem Mass Spectrometry/methods , Asian People/genetics , Female , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Japan , Lipid Metabolism, Inborn Errors/ethnology , Lipid Metabolism, Inborn Errors/genetics , Male , White People/genetics
4.
Mol Genet Metab ; 108(3): 172-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23375472

ABSTRACT

Mucopolysaccharidosis type II (MPS II) is a lysosomal storage disorder that is progressive and involves multiple organs and tissues. While enzyme replacement therapy (ERT) with idursulfase has been shown to improve many somatic features of the disease, some such as dysostosis multiplex and cardiac valve disease appear irreversible once established, and little is known about the preventative effects of ERT in pre-symptomatic patients. We report on two siblings with severe MPS II caused by an inversion mutation with recombination breakpoints located within the IDS gene and its adjacent pseudogene, IDS-2. The siblings initiated treatment with idursulfase at 3.0 years (older brother) and 4 months (younger brother) of age, and we compared their outcomes following 2 years of treatment. At the start of treatment, the older brother showed typical features of MPS II, including intellectual disability. After 34 months of ERT, his somatic disease was stable or improved, but he continued to decline cognitively. By comparison, after 32 months of ERT his younger brother remained free from most of the somatic features that had already appeared in his brother at the same age, manifesting only exudative otitis media. Skeletal X-rays revealed characteristic signs of dysostosis multiplex in the older brother at the initiation of treatment that were unchanged two years later, whereas the younger brother showed only slight findings of dysostosis multiplex throughout the treatment period. The younger brother's developmental quotient trended downward over time to just below the normal range. These findings suggest that pre-symptomatic initiation of ERT may prevent or attenuate progression of the somatic features of MPS II. Follow-up in a larger number of patients is required to confirm the additive long-term benefits of ERT in pre-symptomatic patients.


Subject(s)
Glycoproteins/genetics , Iduronate Sulfatase/therapeutic use , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis II/genetics , Mutation , Time-to-Treatment , Child, Preschool , Cognition/drug effects , Disease Progression , Enzyme Replacement Therapy , Glycoproteins/deficiency , Humans , Iduronate Sulfatase/pharmacology , Male , Mucopolysaccharidosis II/enzymology , Mucopolysaccharidosis II/physiopathology , Siblings
5.
Eur J Endocrinol ; 166(2): 235-40, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22048973

ABSTRACT

OBJECTIVE: Ghrelin requires a fatty acid modification for binding to the GH secretagogue receptor. Acylation of the Ser3 residue of ghrelin is essential for its biological activities. We hypothesized that acyl-CoA is the fatty acid substrate for ghrelin acylation. Because serum octanoyl-CoA levels are altered by fatty acid oxidation disorders, we examined circulating ghrelin levels in affected patients. MATERIALS AND METHODS: Blood levels of acyl (A) and des-acyl (D) forms of ghrelin and acylcarnitine of patients with medium-chain acyl-CoA dehydrogenase (MCAD) deficiency and glutaric aciduria type II (GA2) were measured. RESULTS: Plasma acyl ghrelin levels and A/D ratios increased in patients with MCAD deficiency or GA2 when compared with normal subjects. Reverse-phase HPLC confirmed that n-octanoylated ghrelin levels were elevated in these patients. CONCLUSION: Changing serum medium-chain acylcarnitine levels may affect circulating acyl ghrelin levels, suggesting that acyl-CoA is the substrate for ghrelin acylation.


Subject(s)
Ghrelin/blood , Lipid Metabolism, Inborn Errors/blood , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/blood , Acyl-CoA Dehydrogenase/blood , Acyl-CoA Dehydrogenase/deficiency , Acyl-CoA Dehydrogenase/metabolism , Adult , Blood Chemical Analysis/methods , Carnitine/analogs & derivatives , Carnitine/analysis , Carnitine/blood , Case-Control Studies , Child , Child, Preschool , Chromatography, High Pressure Liquid , Female , Ghrelin/analysis , Ghrelin/metabolism , Humans , Lipid Metabolism, Inborn Errors/metabolism , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/metabolism , Protein Processing, Post-Translational , Young Adult
6.
Pediatr Surg Int ; 26(10): 1025-30, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20661579

ABSTRACT

Patent ductus venosus (PDV) is a rare condition, which usually presents secondary to hepatic atrophy and hepatic failure. We have treated eight cases of PDV, all with hypergalactosemia and hyperbilirubinemia. Ultrasonography and three-dimensional computed tomography demonstrated communication between the portal vein and the inferior vena cava. Of the eight PDV cases, three from the older age group (ages 9, 11, and 14 years) had high-density lesions in their brain nucleus, and one case (age 19 years) had undergone prior Kasai portoenterostomy for biliary atresia. Six PDV patients underwent ligation of PDV and the remaining two cases underwent partial banding of PDV with intraoperative monitoring to maintain portal vein pressure (PVP) under 30 cm H(2)O. Improvement of the intrahepatic portal vein flow was achieved by ligation or banding of PDV. Postoperatively, serum galactose and bilirubin fell to normal ranges, but portal thrombus occurred postoperatively in the first case. We subsequently administered postoperative anticoagulation in the remaining cases and experienced no major complications. These results suggest that PDV ligation and banding are effective surgical approaches for patients with PDV. Close postoperative monitoring to avoid portal thrombus is imperative in these cases.


Subject(s)
Portal Vein/surgery , Vascular Malformations/surgery , Vascular Surgical Procedures/methods , Vena Cava, Inferior/surgery , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Infant, Newborn , Male , Phlebography , Portal Vein/abnormalities , Portal Vein/diagnostic imaging , Retrospective Studies , Tomography, X-Ray Computed , Treatment Outcome , Vascular Malformations/diagnosis , Vena Cava, Inferior/abnormalities , Vena Cava, Inferior/diagnostic imaging , Young Adult
7.
Hum Genet ; 127(6): 619-28, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20376488

ABSTRACT

Short-chain acyl-CoA dehydrogenase (SCAD) is a mitochondrial enzyme involved in the beta-oxidation of fatty acids. Genetic defect of SCAD was documented to cause clinical symptoms such as progressive psychomotor retardation, muscle hypotonia, and myopathy in early reports. However, clinical significance of SCAD deficiency (SCADD) has been getting ambiguous, for some variants in the ACADS gene, which encodes the SCAD protein, has turned out to be widely prevailed among general populations. Accordingly, the pathophysiology of SCADD has not been clarified thus far. The present report focuses on two suspected cases of SCADD detected through the screening of newborns by tandem mass spectrometry. In both subjects, compound heterozygous mutations in ACADS were detected. The mutated genes were expressed in a transient gene expression system, and the enzymatic activities of the obtained mutant SCAD proteins were measured. The activities of the mutant SCAD proteins were significantly lower than that of the wild-type enzyme, confirming the mechanism underlying the diagnosis of SCADD in both subjects. Moreover, the mutant SCAD proteins gave rise to mitochondrial fragmentation and autophagy, both of which were proportional to the decrease in SCAD activities. The association of autophagy with programmed cell death suggests that the mutant SCAD proteins are toxic to mitochondria and to the cells in which they are expressed. The expression of recombinant ACADS-encoded mutant proteins offers a technique to evaluate both the nature of the defective SCAD proteins and their toxicity. Moreover, our results provide insight into possible molecular pathophysiology of SCADD.


Subject(s)
Butyryl-CoA Dehydrogenase/deficiency , Butyryl-CoA Dehydrogenase/genetics , Genes , Lipid Metabolism Disorders/genetics , Mutation , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Base Sequence , Butyryl-CoA Dehydrogenase/metabolism , Genotype , Heterozygote , Humans , Infant, Newborn , Lipid Metabolism Disorders/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Molecular Sequence Data , Oxidation-Reduction , Protein Structure, Secondary/genetics , Recombinant Proteins/metabolism
8.
J Inherit Metab Dis ; 33(2): 141-50, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20162367

ABSTRACT

Mucopolysaccharidosis I (MPS I) is an autosomal recessive disorder caused by deficiency of alpha-L-iduronidase leading to accumulation of its catabolic substrates, dermatan sulfate (DS) and heparan sulfate (HS), in lysosomes. This results in progressive multiorgan dysfunction and death in early childhood. The recent success of enzyme replacement therapy (ERT) for MPS I highlights the need for biomarkers that reflect response to such therapy. To determine which biochemical markers are better, we determined serum and urine DS and HS levels by liquid chromatography tandem mass spectrometry in ERT-treated MPS I patients. The group included one Hurler, 11 Hurler/Scheie, and two Scheie patients. Seven patients were treated from week 1, whereas the other seven were treated from week 26. Serum and urine DS (DeltaDi-4S/6S) and HS (DeltaDiHS-0S, DeltaDiHS-NS) were measured at baseline, week 26, and week 72. Serum DeltaDi-4S/6S, DeltaDiHS-0S, and DeltaDiHS-NS levels decreased by 72%, 56%, and 56%, respectively, from baseline at week 72. Urinary glycosaminoglycan level decreased by 61.2%, whereas urine DeltaDi-4S/6S, DeltaDiHS-0S, and DeltaDiHS-NS decreased by 66.8%, 71.8%, and 71%, respectively. Regardless of age and clinical severity, all patients showed marked decrease of DS and HS in blood and urine samples. We also evaluated serum DS and HS from dried blood-spot samples of three MPS I newborn patients, showing marked elevation of DS and HS levels compared with those in control newborns. In conclusion, blood and urine levels of DS and HS provide an intrinsic monitoring and screening tool for MPS I patients.


Subject(s)
Dermatan Sulfate/blood , Dermatan Sulfate/urine , Heparitin Sulfate/blood , Heparitin Sulfate/urine , Mucopolysaccharidosis I , Adolescent , Adult , Biomarkers/blood , Biomarkers/urine , Child , Child, Preschool , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant, Newborn , Male , Mass Screening/methods , Mucopolysaccharidosis I/blood , Mucopolysaccharidosis I/diagnosis , Mucopolysaccharidosis I/urine , Neonatal Screening/methods , Tandem Mass Spectrometry , Young Adult
9.
Metabolism ; 59(1): 107-13, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19766269

ABSTRACT

Children with congenital portosystemic venous shunt (PSVS) are at risk for developing pulmonary hypertension, irrespective of the severity of portal hypertension or liver damage. Altered metabolisms of nitric oxide (NO) and endothelin-1 (ET-1), which are linked with oxidative stress and control vascular tone, might contribute to the vascular disturbance. This study examined 14 children (aged 1-5 years) with congenital PSVS lacking major liver damage and portal hypertension. Serum levels of nitrite/nitrate (NOx) as stable metabolites of NO, and of asymmetric dimethylarginine (ADMA) as an endogenous NO synthase inhibitor were determined, along with the plasma level of ET-1. Oxidative stress, which might affect the production of such mediators, was also examined using specific urinary and blood markers. The NOx levels were significantly lower in affected children than in the age-matched control group, although ET-1 levels were significantly higher than the control levels. In the affected children, the ADMA levels and ADMA/NOx ratios were higher, respectively, by 30% and 130% and showed significant positive correlations with the shunt ratios. Oxidative stress markers, including plasma thiobarbiturate reactive substances and urinary acrolein-lysine and 8-hydroxy-2'-deoxyguanosine, were significantly higher in affected children than in the control group, consistent with them being subjected to enhanced oxidative stress. These results suggest the presence of altered metabolisms of vascular mediators and enhanced oxidative stress in asymptomatic preschool children with congenital PSVS.


Subject(s)
Blood Vessels/physiopathology , Oxidative Stress , Portal System/abnormalities , Amino Acids/blood , Ammonia/blood , Arginine/analogs & derivatives , Arginine/blood , Child, Preschool , Endothelin-1/blood , Female , Humans , Infant , Male , Nitric Oxide/blood
10.
Pediatr Res ; 64(6): 667-72, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18670371

ABSTRACT

The introduction of tandem mass spectrometry (MS/MS) has made it possible to screen for very-long-chain acyl-CoA dehydrogenase (VLCAD) deficiency. To confirm the diagnosis in cases with an abnormal profile of blood acylcarnitines, we developed a new enzymatic assay method for determining dehydrogenase activity toward palmitoyl-CoA (C16:0) in lymphocytes. Using this method, the production of 2-hexadecenoyl-CoA (C16:1) by crude cell lysates can be directly quantified using high performance liquid chromatography (HPLC). We applied the assay to 7 myopathic patients, 7 hypoglycemic patients, and 2 presymptomatic newborns with elevated levels of tetradecenoylcarnitine (C14:1 AC) in blood, and found impaired VLCAD activity in all of the 7 myopathic patients and both of the 2 newborns. All of the 7 hypoglycemic patients had normal level of the enzyme activity. Results of the ACADVL gene analysis were in consistent with the enzymatic diagnosis. These results suggest that MS/MS-based screening for VLCAD deficiency using blood C14:1 AC as the indicator may show a considerably high false-positive rate in selective screening of symptomatic patients. Our practical enzymatic assay can be a useful test for the accurate diagnosis of VLCAD deficiency cases screened by MS/MS.


Subject(s)
Acyl Coenzyme A/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Lipid Metabolism Disorders/diagnosis , Neonatal Screening/methods , Tandem Mass Spectrometry/methods , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant, Newborn , Japan , Lipid Metabolism Disorders/genetics , Male , Palmitoyl Coenzyme A/metabolism , Young Adult
12.
Pediatr Neurol ; 34(3): 239-41, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16504797

ABSTRACT

This report presents a case of steroid sulfatase deficiency with bilateral periventricular nodular heterotopia. A 13-year-old male was diagnosed as having steroid sulfatase deficiency because steroid sulfatase activity was not detected in his leukocytes. In deoxyribonucleic acid studies, steroid sulfatase locus and adjacent loci were found to be deleted in his deoxyribonucleic acid. Cranial magnetic resonance imaging revealed periventricular nodular heterotopia, disclosing an irregular contour of the lateral walls of the lateral ventricles due to small nodular masses that were isointense as to the gray matter. In steroid sulfatase deficiency patients, bilateral periventricular nodular heterotopia must be considered.


Subject(s)
Brain , Cerebral Ventricles/pathology , Choristoma/genetics , Chromosome Deletion , Dominance, Cerebral/physiology , Ichthyosis, X-Linked , Ichthyosis, X-Linked/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Adolescent , Cerebral Ventriculography , Choristoma/diagnosis , Diagnosis, Differential , Humans , Ichthyosis, X-Linked/diagnosis , Lateral Ventricles/pathology , Magnetic Resonance Imaging , Male , Phenotype , Steryl-Sulfatase/genetics
14.
Article in English | MEDLINE | ID: mdl-16055049

ABSTRACT

We developed a simple and sensitive stable-isotope dilution method for the quantification of 3-hydroxyglutaric acid (3HGA) and glutaric acid (GA) in body fluids. In our method, tert-butyldimethylsilyl (tBDMS) derivatives of 3HGA and GA were measured with a conventional electron-impact ionization (EI) mode in gas chromatography-mass spectrometry (GC-MS). The control values for 3HGA in nmol/ml were 0.15+/-0.08 (serum; n=10) and 0.07+/-0.03 (CSF; n=10). In addition, glutarylcarnitine and free carnitine were quantified by electrospray tandem mass spectrometry. Using these methods, we monitored 3HGA, GA, and glutarylcarnitine in the body fluids of three patients with glutaric aciduria type 1 found during newborn screening. None of the patients had experienced neurological strokes, which are possibly caused by the accumulation of 3HGA, at 15-24 months of age under a disease-specific treatment, including carnitine supplementation. Our data showed that 3HGA levels were relatively high in some serum samples with lower glutarylcarnitine and carnitine levels, suggesting that carnitine supplementation may play a role in preventing the accumulation of 3HGA in patients with this disease.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Gas Chromatography-Mass Spectrometry/methods , Glutarates/analysis , Neonatal Screening/methods , Amino Acid Metabolism, Inborn Errors/metabolism , Deuterium/blood , Deuterium/cerebrospinal fluid , Deuterium/urine , Glutarates/metabolism , Humans , Infant, Newborn , Reproducibility of Results , Sensitivity and Specificity
15.
Article in English | MEDLINE | ID: mdl-16046200

ABSTRACT

Many of the previously described enzymatic assay methods for the diagnosis of medium-chain acyl-CoA dehydrogenase (MCAD) deficiency have been dependent upon the measurement of radioisotope-labeled co-products or reduction of electron acceptors. We have developed a direct assay method to detect 2-enoyl-CoA production using high-performance liquid chromatography (HPLC). Crude cell lysate prepared from lymphocytes were incubated with n-octanoyl-CoA and ferrocenium hexafluorophosphate. The detection of 2-octenoyl-CoA was significantly reproducible. We applied the assay to samples from four infants suspected to have MCAD deficiency by tandem mass spectrometry (MS/MS) newborn screening conducted in the Hiroshima area of Japan. Three of them were proved to have pathologically reduced residual enzyme activities, although they were associated with various clinical and biochemical phenotypes. In addition, another symptomatic Japanese patient and her presymptomatic sibling who were detected by MS/MS selective screening were successfully diagnosed by our enzymatic assay. These results indicate that the method can be a useful confirmatory test for MS/MS screening of MCAD deficiency.


Subject(s)
Acyl Coenzyme A/analysis , Acyl-CoA Dehydrogenase/deficiency , Chromatography, High Pressure Liquid/methods , Metabolism, Inborn Errors/diagnosis , Acyl Coenzyme A/metabolism , Acyl-CoA Dehydrogenase/genetics , Acyl-CoA Dehydrogenase/metabolism , Base Sequence , Child, Preschool , Female , Ferrous Compounds/metabolism , Humans , Infant , Infant, Newborn , Japan , Lymphocytes/enzymology , Lymphocytes/metabolism , Male , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Neonatal Screening/methods , Neonatal Screening/standards , Reproducibility of Results , Sequence Deletion , Sequence Homology, Nucleic Acid , Spectrometry, Mass, Electrospray Ionization/methods
16.
Clin Chim Acta ; 353(1-2): 193-9, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15698607

ABSTRACT

BACKGROUND: Isovaleric acidemia (IVA) is one of the various target disorders for tandem mass spectrometry (MS/MS) newborn screening. In the diagnosis of IVA, no enzymatic assay method for isovaleryl-CoA dehydrogenase (IVD) activity has been reported whereby the production of enoyl-CoA species was directly detected. We established a direct assay method to detect 3-methylcrotonyl-CoA (MC-CoA) production using high-performance liquid chromatography (HPLC). METHODS: Isovaleryl-CoA dehydrogenase crude enzyme was prepared by sonicating lymphocytes in peripheral blood. Aliquots were incubated with isovaleryl-CoA, flavin adenine dinucleotide, and phenazine methosulfate. 3-Methylcrotonyl-CoA produced in the samples was separated by HPLC and detected using an ultraviolet spectrophotometer. RESULTS: The detection of MC-CoA was reproducible depending upon the concentration of the substrates, the incubation time, and the number of cells contained in the crude enzyme solution. We applied this assay to three patients diagnosed with IVA and showed that neither of them had detectable residual activity. Only a few hours were required from the initial blood sampling to the end of the assay. CONCLUSIONS: These results demonstrate that this method for detecting MC-CoA production, using HPLC, is a practical assay for determining IVD activity. It can be a useful confirmatory test for IVA cases detected through MS/MS screening of newborns.


Subject(s)
Chromatography, High Pressure Liquid/methods , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Humans , Isovaleryl-CoA Dehydrogenase , Reproducibility of Results
17.
Mol Genet Metab ; 82(4): 329-33, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15308131

ABSTRACT

Methylmalonic acidemia (MMA) is caused by the deficient activity of l-methylmalonyl-CoA mutase, which is a vitamin B(12) (or cobalamin, Cbl)-dependent enzyme. MMA due to the effect of insufficient Cbl metabolism is classified into three forms (cblA, cblB, and cblH). Recently, the genes responsible for cblA and cblB were identified as MMAA and MMAB, respectively. The MMAA protein likely transports Cbl into the mitochondria for adenosylcobalamin synthesis, while the MMAB protein appears to be an adenosyltransferase. We performed a mutation analysis of 10 unrelated Japanese patients with vitamin B(12)-responsive MMA. Seven patients had mutations in MMAA, whereas the other three patients showed no disease-causing substitutions in either MMAA or MMAB. Five novel mutations were identified in MMAA (R22X, R145X, L217X, R359G, and 503delC). The 503delC mutation was observed in five of the seven MMAA patients, suggesting that the mutation is prevalent in Japanese patients. This finding may facilitate the DNA diagnosis of vitamin B(12)-responsive MMA within the Japanese population.


Subject(s)
Acidosis/genetics , Alkyl and Aryl Transferases/genetics , Membrane Transport Proteins/genetics , Methylmalonyl-CoA Mutase/deficiency , Mitochondrial Proteins/genetics , Mutation/genetics , Acidosis/ethnology , Amino Acid Sequence , DNA Mutational Analysis , Humans , Japan , Mitochondrial Membrane Transport Proteins , Molecular Sequence Data , Syndrome , Vitamin B 12/metabolism
18.
Article in English | MEDLINE | ID: mdl-15149612

ABSTRACT

Acrolein, the metabolite of cyclophosphamide and ifosphamide, irritates mucous membranes and is considered pathogenetically important in hemorrhagic cystitis. Increasing fluid intake or administering sodium 2-mercaptoethanesulfonate (mesna), a thiol compound, can reduce the risk of this complication. We measured urinary acrolein concentrations using headspace-solid-phase microextraction gas chromatography and mass spectrometry (headspace-SPME-GC-MS) in 19 patients receiving cyclophosphamide and ifosphamide (36 occasions). Peak acrolein concentrations occurred at 1-12h (mean +/- S.D., 5.0+/-2.7) after starting therapy, ranging from 0.3 to 406.8 nM (39.7+/-76.7), with varying patterns over time. Maintaining high urine volume was important for preventing increases in urinary acrolein concentration, as urinary acrolein concentration tended to rise as urine volume decreased. Urinalysis detected occult blood in three cases, but the patients had no clinical symptoms of hemorrhagic cystitis. In clinical trials involving cyclophosphamide and ifosphamide, monitoring of urinary acrolein concentration could indicate when to take heightened preventive measures against hemorrhagic cystitis.


Subject(s)
Acrolein/urine , Antineoplastic Agents, Alkylating/administration & dosage , Cyclophosphamide/administration & dosage , Ifosfamide/administration & dosage , Adolescent , Adult , Child , Child, Preschool , Female , Gas Chromatography-Mass Spectrometry , Humans , Male
19.
Pediatr Int ; 46(1): 5-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15043656

ABSTRACT

BACKGROUND: The most widely used method for newborn screening for homocystinuria (HCU) is a semi-quantitative bacterial inhibition assay for measuring methionine concentration in dried blood spots (DBS). Because this method has resulted in a number of missed cases due to many factors, we developed a high performance liquid chromatography (HPLC) method with fluorescence detection to measure total homocysteine (tHcy) in DBS which might be useful for newborn screening for HCU. METHODS: One disk of DBS 3 mm in diameter was sonicated in 10 min. The extract was reduced with dithioerythritol and was derivatized with 4-aminosulfonyl-7fluoro-2,1,3-benzoxadiazole before injection into HPLC. RESULTS: This method showed good linearity (r = 0.996), precision (coefficient of variation range 2.7-5%), and excellent correlation coefficient between DBS and serum tHcy, both in control (r = 0.932) and patient samples (r = 0.952). By this method, the mean tHcy concentration in DBS of preterm newborns, full-term newborns, and adults was 1.4 +/- 1.0, 2.5 +/- 1.6, and 4.9 +/- 1.5 micro mol/L, respectively. The mean tHcy DBS concentration in two cases of cystathionine-beta-synthase deficiency and one case of 5,10-methylentetrahydrofolate reductase deficiency was 22.7 +/- 2.88, 29.3 +/- 1.90, and 41.3 micro mol/L, respectively. CONCLUSIONS: The present method, which is rapid, user friendly and reliable, seems applicable to newborn screening of HCU in place of methionine measurement.


Subject(s)
Chromatography, High Pressure Liquid/methods , Homocysteine/blood , Homocystinuria/prevention & control , Neonatal Screening/methods , Analysis of Variance , Fluorescence , Homocystinuria/blood , Humans , Infant, Newborn , Linear Models , Reproducibility of Results
20.
No To Shinkei ; 56(1): 64-8, 2004 Jan.
Article in Japanese | MEDLINE | ID: mdl-15024832

ABSTRACT

We experienced a 24-year-old male patient with myalgia and myoglobuinuria followed by severe exercise from childhood. In 18 years old, he had severe myaglia after a long time-night trip by bus. He was diagnosed as acute renal failure induced by rhabdomyolysis and treated with hemodialysis. In 24 years old, he was admitted to our hospital because of repeated rhabdomyolysis. We performed muscle biopsy from right quadriceps femoris, however histological and immunohistochemistological studies were normal. Ischemic forearm exercise test showed the elevation of lactic acid in serum. Therefore, we performed the analysis of acylcarnitine in serum, and the measurement of enzyme in beta-oxidation in muscle and white blood cells. These showed the lack of very-long-chain-acyl coA dehydrogenase (VLCAD) activity. He was diagnosed as skeletal muscle type VLCAD deficiency. Under the guidance of high carbohydrate and low fat diet, creatine kinase was controlled around 400 IU/l. VLCAD deficiency is important to make a differential diagnosis of young cases with recurrent elevation of creatine kinase.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Rhabdomyolysis/etiology , Adult , Biopsy , Creatine Kinase/blood , Diagnosis, Differential , Humans , Male , Muscle, Skeletal/pathology , Recurrence , Rhabdomyolysis/diagnosis , Rhabdomyolysis/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...