Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Biology (Basel) ; 10(4)2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33916397

ABSTRACT

Visceral obesity is common in the United Arab Emirates and worldwide. Although laparoscopic sleeve gastrectomy (LSG) leads to effective and sustainable weight loss, its long-term beneficial impact on other risk factors, including blood lipid and fatty acid (FA) profiles, remains unknown. These two profiles were assessed in patients 3 years after undergoing LSG and in LSG candidates (controls). Lipid profiles were measured using the Cobas e411 modular analyzer, and 35 FAs were identified. The age and body mass index were 36.55 ± 8.65 years and 31.49 ± 6.43 kg/m2 in the LSG group and 35.44 ± 9.51 years and 32.29 ± 5.38 kg/m2 in the control group, respectively. The overall lipid profile was more favorable in the LSG group than in the control group. Total saturated, monounsaturated, and polyunsaturated FAs were similar between the groups, but total medium-chain FAs were more abundant in the LSG group. In endogenous FA synthesis, the estimated activity of C16Δ9 desaturase and Δ5 desaturase decreased, whereas that of elongase increased in the LSG group compared with that in the control group. The benefits of LSG on blood lipid and FA profiles in patients with 3-year LSG may be limited. Hence, lifestyle interventions combined with a long-term and strict regular follow-up regime may be warranted for patients undergoing LSG.

2.
Metabolites ; 10(10)2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33050140

ABSTRACT

This article reports a targeted metabolomic method for total plasma fatty acids (FAs) of clinical or nutritional relevance. Thirty-six saturated, unsaturated, or branched-chain FAs with a chain length of C8-C28 were quantified using reversed-phase liquid chromatography-tandem mass spectrometry. FAs in plasma (10 µL) were acid-hydrolyzed, extracted, and derivatized with DAABD-AE (4-[2-(N,N-Dimethylamino)ethylaminosulfonyl]-7-(2-aminoethylamino)-2,1,3-benzoxadiazole) at 60 °C for 1 h. Derivatization resulted in a staggering nine orders of magnitude higher sensitivity compared to underivatized analytes. FAs were measured by multiple-reaction monitoring using stable isotope internal standards. With physiological and pathological analyte levels in mind, linearity was established using spiked plasma. Intra-day (n = 15) and inter-day (n = 20) imprecisions expressed as variation coefficient were ≤10.2% with recovery ranging between 94.5-106.4%. Limits of detection and limit of quantitation ranged between 4.2-14.0 and 15.1-51.3 pmol per injection, respectively. Age-stratified reference intervals were established in four categories: <1 month, 1-12 month, 1-18 year, and >18 year. This method was assessed using samples from patients with disorders affecting FAs metabolism. For the first time, C28:0 and C28:0/C22:0 ratio were evaluated as novel disease biomarkers. This method can potentially be utilized in diagnosing patients with inborn errors of metabolism, chronic disease risk estimation, or nutritional applications.

3.
Medicina (Kaunas) ; 56(3)2020 Mar 20.
Article in English | MEDLINE | ID: mdl-32245061

ABSTRACT

Background and objectives: Although laparoscopic sleeve gastrectomy (LSG) is effective for obesity management, postoperative vitamin B12 (B12) deficiency is of major concern. In this cross-sectional study, we assessed the levels of B12 and its related functional biomarkers, namely, total homocysteine (tHcy), methylmalonic acid (MMA), folate, methylcitric acid (MCA), and hemoglobin (Hb), in one-year postoperative LSG patients and matched controls. Materials and Methods: Plasma B12, tHcy, MMA, folate, and MCA were measured in matched controls (n = 66) and patients (n = 71) using validated liquid chromatography-tandem mass spectrometry techniques and protocols in the United Arab Emirates (UAE). Results: The median B12 concentration in patients (177 pmol/L) was significantly lower (p < 0.001) than in the controls (334.7 pmol/L). The tHcy and MMA levels were significantly increased (p < 0.001 and p = 0.011, respectively) and folate levels were significantly decreased (p = 0.001) in the LSG patients compared to the controls. Interestingly, no significant difference in MCA levels were observed between the two groups. The levels of tHcy and MMA were concomitantly increased with the decreased folate levels in postoperative LSG patients when compared with the controls. The Hb levels were significantly lower in males and females in the patient group compared with those in the control group, respectively (p = 0.005 and p = 0.043). Conclusions: This is the first report of serum levels of B12 and its functional biomarkers in postoperative LSG patients among a local population from the UAE. Our findings revealed significant alterations of the B12 biomarkers, total B12, MMA, and tHcy in one-year postoperative LSG patients.


Subject(s)
Gastrectomy/adverse effects , Postoperative Complications/blood , Postoperative Complications/diagnosis , Vitamin B 12 Deficiency/blood , Vitamin B 12 Deficiency/diagnosis , Adult , Biomarkers/blood , Chromatography, Liquid , Citrates/blood , Cross-Sectional Studies , Female , Folic Acid/blood , Hemoglobins , Homocysteine/blood , Humans , Male , Mass Spectrometry , Methylmalonic Acid/blood , United Arab Emirates
4.
Hum Genet ; 139(5): 657-673, 2020 May.
Article in English | MEDLINE | ID: mdl-32219518

ABSTRACT

GM1-gangliosidosis, a lysosomal storage disorder, is associated with ~ 161 missense variants in the GLB1 gene. Affected patients present with ß-galactosidase (ß-Gal) deficiency in lysosomes. Loss of function in ER-retained misfolded enzymes with missense variants is often due to subcellular mislocalization. Deoxygalactonojirimycin (DGJ) and its derivatives are pharmaceutical chaperones that directly bind to mutated ß-Gal in the ER promoting its folding and trafficking to lysosomes and thus enhancing its activity. An Emirati child has been diagnosed with infantile GM1-gangliosidosis carrying the reported p.D151Y variant. We show that p.D151Y ß-Gal in patient's fibroblasts retained < 1% residual activity due to impaired processing and trafficking. The amino acid substitution significantly affected the enzyme conformation; however, p.D151Y ß-Gal was amenable for partial rescue in the presence of glycerol or at reduced temperature where activity was enhanced with ~ 2.3 and 7 folds, respectively. The butyl (NB-DGJ) and nonyl (NN-DGJ) derivatives of DGJ chaperoning function were evaluated by measuring their IC50s and ability to stabilize the wild-type ß-Gal against thermal degradation. Although NN-DGJ showed higher affinity to ß-Gal, it did not show a significant enhancement in p.D151Y ß-Gal activity. However, NB-DGJ promoted p.D151Y ß-Gal maturation and enhanced its activity up to ~ 4.5% of control activity within 24 h which was significantly increased to ~ 10% within 6 days. NB-DGJ enhancement effect was sustained over 3 days after washing it out from culture media. We therefore conclude that NB-DGJ might be a promising therapeutic chemical chaperone in infantile GM1 amenable variants and therefore warrants further analysis for its clinical applications.


Subject(s)
1-Deoxynojirimycin/pharmacology , Fibroblasts/metabolism , Gangliosidosis, GM1/metabolism , Mutant Proteins/metabolism , Mutation , Protein Processing, Post-Translational/drug effects , beta-Galactosidase/metabolism , 1-Deoxynojirimycin/chemistry , Child, Preschool , Endoplasmic Reticulum Stress/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Fibroblasts/pathology , Gangliosidosis, GM1/drug therapy , Gangliosidosis, GM1/pathology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Lysosomes/pathology , Male , Molecular Chaperones/pharmacology , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Conformation , Protein Transport , beta-Galactosidase/chemistry , beta-Galactosidase/genetics
5.
J Mol Neurosci ; 70(1): 45-55, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31468281

ABSTRACT

Schindler disease is a rare autosomal recessive lysosomal storage disorder caused by a deficiency in alpha-N-acetylgalactosaminidase (α-NAGA) activity due to defects in the NAGA gene. Accumulation of the enzyme's substrates results in clinically heterogeneous symptoms ranging from asymptomatic individuals to individuals with severe neurological manifestations. Here, a 5-year-old Emirati male born to consanguineous parents presented with congenital microcephaly and severe neurological manifestations. Whole genome sequencing revealed a homozygous missense variant (c.838C>A; p.L280I) in the NAGA gene. The allele is a reported SNP in the ExAC database with a 0.0007497 allele frequency. The proband's asymptomatic sister and cousin carry the same genotype in a homozygous state as revealed from the family screening. Due to the extreme intrafamilial heterogeneity of the disease as seen in previously reported cases, we performed further analyses to establish the pathogenicity of this variant. Both the proband and his sister showed abnormal urine oligosaccharide patterns, which is consistent with the diagnosis of Schindler disease. The α-NAGA activity was significantly reduced in the proband and his sister with 5.9% and 12.1% of the mean normal activity, respectively. Despite the activity loss, p.L280I α-NAGA processing and trafficking were not affected. However, protein molecular dynamic simulation analysis revealed that this amino acid substitution is likely to affect the enzyme's natural dynamics and hinders its ability to bind to the active site. Functional analysis confirmed the pathogenicity of the identified missense variant and the diagnosis of Schindler disease. Extreme intrafamilial clinical heterogeneity of the disease necessitates further studies for proper genetic counseling and management.


Subject(s)
Lysosomal Storage Diseases/genetics , Mutation, Missense , Neuroaxonal Dystrophies/genetics , Phenotype , alpha-N-Acetylgalactosaminidase/deficiency , Adult , Catalytic Domain , Cells, Cultured , Child , Female , Humans , Lysosomal Storage Diseases/pathology , Male , Neuroaxonal Dystrophies/pathology , Pedigree , Protein Binding , alpha-N-Acetylgalactosaminidase/chemistry , alpha-N-Acetylgalactosaminidase/genetics , alpha-N-Acetylgalactosaminidase/metabolism
6.
Sci Rep ; 9(1): 12366, 2019 08 26.
Article in English | MEDLINE | ID: mdl-31451751

ABSTRACT

Deficiency of propionyl-CoA carboxylase causes propionic acidemia and deficiencies of methylmalonyl-CoA mutase or its cofactor adenosylcobalamin cause methylmalonic acidemia. These inherited disorders lead to pathological accumulation of propionyl-CoA which is converted in Krebs cycle to methylcitrate (MCA) in a reaction catalyzed by citrate synthase. In healthy individuals where no propionyl-CoA accumulation occurs, this enzyme drives the condensation of acetyl-CoA with oxaloacetate to produce citric acid (CA), a normal Krebs cycle intermediate. The competitive synthesis of CA and MCA through the same enzymatic mechanism implies that increase in MCA production is accompanied by decrease in CA levels. In this study, we assessed MCA concentration and the ratio of MCA/CA as plausible markers for propionic and methylmalonic acidemias. We measured MCA and CA in dried blood spots using liquid chromatography tandem mass spectrometry. The reference ranges of MCA, CA and MCA/CA in 123 healthy individuals were ≤0.63 µmol/L, 36.6-126.4 µmol/L and 0.0019-0.0074, respectively. In patients with propionic and methylmalnic acidemias (n = 7), MCA concentration ranged between 1.0-12.0 µmol/L whereas MCA/CA was between 0.012-0.279. This is the first report to describe the potential role of MCA and MCA/CA in dried blood spots as diagnostic and monitoring biomarkers for inherited disorders of propionyl-CoA metabolism.


Subject(s)
Biomarkers/blood , Citrates/blood , Dried Blood Spot Testing , Metabolism, Inborn Errors/blood , Propionates/blood , Propionates/metabolism , Biological Assay , Case-Control Studies , Humans , Methylmalonic Acid/metabolism
7.
J Proteome Res ; 18(3): 841-854, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30507207

ABSTRACT

Cystic fibrosis (CF) is a complex multiorgan disorder that is among the most common fatal genetic diseases benefiting from therapeutic interventions early in life. Newborn screening (NBS) for presymptomatic detection of CF currently relies on a two-stage immunoreactive trypsinogen (IRT) and cystic fibrosis transmembrane conductance regulator (CFTR) mutation panel algorithm that is sensitive but not specific for identifying affected neonates with a low positive predictive value. For the first time, we report the discovery of a panel of CF-specific metabolites from a single 3.2 mm diameter dried blood spot (DBS) punch when using multisegment injection-capillary electrophoresis-mass spectrometry (MS) as a high-throughput platform for nontargeted metabolite profiling from volume-restricted/biobanked specimens with quality control. This retrospective case-control study design identified 32 metabolites, including a series of N-glycated amino acids, oxidized glutathione disulfide, and nicotinamide that were differentially expressed in normal birth weight CF neonates without meconium ileus ( n = 36) as compared to gestational age/sex-matched screen-negative controls ( n = 44) after a false discovery rate adjustment ( q < 0.05). Also, 16 metabolites from DBS extracts allowed for discrimination of true CF cases from presumptive screen-positive carriers with one identified CFTR mutation and transient neonatal hypertrypsinogenemic neonates ( n = 72), who were later confirmed as unaffected due to a low sweat chloride (<29 mM) test result. Importantly, six CF-specific biomarker candidates satisfying a Bonferroni adjustment ( p < 7.25 × 10-5) from three independent batches of DBS specimens included several amino acids depleted in circulation (Tyr, Ser, Thr, Pro, Gly) likely reflecting protein maldigestion/malabsorption. Additionally, CF neonates had lower ophthalmic acid as an indicator of oxidative stress due to impaired glutathione efflux from exocrine/epithelial tissue and elevation of an unknown trivalent peptide that was directly correlated with IRT (ρ = 0.332, p = 4.55 × 10-4). Structural elucidation of unknown metabolites was performed by high-resolution MS/MS, whereas biomarker validation was realized when comparing a subset of metabolites from matching neonatal DBS specimens independently analyzed by direct infusion-MS/MS at an accredited NBS facility. This work sheds new light into the metabolic phenotype of CF early in life, which is required for better functional understanding of CFTR mutations of unknown clinical consequence and the development of more accurate yet cost-effective strategies for CF screening.


Subject(s)
Cystic Fibrosis/diagnosis , Cystic Fibrosis/metabolism , Dried Blood Spot Testing/methods , Neonatal Screening/methods , Biomarkers/blood , Case-Control Studies , Electrophoresis, Capillary , Female , Humans , Infant, Newborn , Male , Retrospective Studies , Tandem Mass Spectrometry
8.
Brain Dev ; 41(1): 57-65, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30078639

ABSTRACT

BACKGROUND: Mutations in PEX16 cause peroxisome biogenesis disorder (PBD). Zellweger syndrome characterized by neurological dysfunction, dysmorphic features, liver disease and early death represents the severe end of this clinical spectrum. Here we discuss the diagnostic challenge of atypical PEX16 related PBD in 3 patients from highly inbred kindred and describe the role of specific metabolites analyses, fibroblasts studies, whole-exome sequencing (WES) and metabolomics profiling to establish the diagnosis. METHODS AND PATIENTS: The proband is a 12-year-old male born to consanguineous parents. Despite normal development in the first year, regression and progressive spastic diplegia, poor coordination and dysarthria occurred thereafter. Patient 2 (3-year old female) and Patient 3 (19-month old female) shared similar clinical course with the proband. Biochemical studies on plasma and fibroblasts, WES and global metabolomics analyses were performed. RESULTS: Very-long-chain fatty acids analysis showed subtle elevations in C26 and C26/C22. Global Metabolomics-Assisted Pathway profiling was not remarkable. Immunocytochemical investigations on fibroblasts revealed fewer catalase and PMP70-containing particles indicating aberrant peroxisomal assembly. Complementation studies were inconclusive. WES revealed a novel homozygous variant in PEX16 (c.859C>T). The biochemical profiles of Patient 2 and Patient 3 were similar to the proband and the same genotype was confirmed. CONCLUSION: This paper highlights the diagnostic challenge of PEX16 patients due to the widely variable clinical and biochemical phenotypes. It also emphasizes the important roles of combined biochemical assays with next generation sequencing techniques in reaching diagnosis in the context of atypical clinical presentations, subtle biomarker abnormalities and consanguinity.


Subject(s)
Membrane Proteins/genetics , Peroxisomal Disorders/diagnosis , Peroxisomal Disorders/metabolism , Cells, Cultured , Child , Child, Preschool , Diagnosis, Differential , Family , Female , Humans , Infant , Male , Metabolome , Peroxisomal Disorders/genetics , Phenotype , Zellweger Syndrome/diagnosis , Zellweger Syndrome/genetics , Zellweger Syndrome/metabolism
9.
Clin Chim Acta ; 487: 41-45, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30217751

ABSTRACT

Accumulation of methylcitrate is a biochemical hallmark of inborn errors of propionate metabolism, a group of disorders that include propionic acidemia, methylmalonic aciduria and cobalamin defects. In clinical laboratories, this analyte is measured without quantification by gas chromatography mass spectrometry as part of urine organic acids. Here we describe a simple, sensitive and specific method to quantify methylcitrate in dried urine spots by liquid chromatography tandem mass spectrometry. Methylcitrate is extracted and derivatized with 4-[2-(N,N-dimethylamino)ethylaminosulfonyl]-7-(2-aminoethylamino)-2,1,3-benzoxadiazole in a single step. A derivatization mixture was added to 3.2 mm disc of dried urine spots, incubated at 65 °C for 45 min and 4 µl of the reaction mixture were analyzed. Separation was achieved on C18 column with methylcitrate eluting at 3.8 min. Intraday and interday imprecision (n = 17) were ≤20.9%. The method was applied on dried urine spots from established patients and controls. In controls (n = 135), methylcitrate reference interval of 0.4-3.4 mmol/mol creatinine. In patients, methylcitrate ranged between 8.3 and 591 mmol/mol creatinine. Quantification of methylcitrate provides important diagnostic clues for propionic acidemia, methylmalonic aciduria and cobalamin disorders. The potential utilization of methylcitrate as monitoring biomarker of patients under treatment and whether it correlates with the clinical status has yet to be established.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Citrates/urine , Propionic Acidemia/diagnosis , Urinalysis , Amino Acid Metabolism, Inborn Errors/urine , Chromatography, Liquid , Humans , Propionic Acidemia/urine , Tandem Mass Spectrometry
10.
Clin Biochem ; 54: 131-138, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29402417

ABSTRACT

BACKGROUND: Several acylcarnitines used as primary markers on dried blood filter papers (DBS) for newborn screening lack specificity and contribute to a higher false positive rate. The analysis of urine acylglycines is useful in the diagnosis of inborn errors of metabolism (IEM) including medium chain acyl-CoA dehydrogenase deficiency (MCADD), isovaleric acidemia, and beta-ketothiolase deficiency (BKTD). Currently, no method for analyzing acylglycines from DBS has been published. METHODS: Acylglycines were extracted from two 3.2 mm DBS punches and butylated using Butanol-HCl. Ultra Performance Liquid Chromatography (UPLC-MS/MS) with run time of 10 min permits resolution and quantitation of 15 acylglycines; including several isobaric. Method development was completed. Reference intervals (n = 573) were established for four birth weight groups. Furthermore, samples from patients with a confirmed IEM (n = 11), and false positive screens (n = 78) were analyzed to validate the interpretation obtained from the newly established reference intervals. RESULTS: Calibration curves were linear from 0.005 to 25.0 µM. Ion suppression was evaluated as minimal (2 to 10%). Samples from known patients were used to validate the reference intervals. For C5OH-related disorders, tiglylglycine (TG), TG/acetylglycine (AG) ratio, 3methylcrotonylglycine (3MCG) and 3MCG/AG ratio increased specificity. Propionylglycine (PG) and PG/acetylglycine ratio were two discriminatory markers in the investigation of C3-related disorders. Hexanoylglycine (HG), octanoylglycine (OG), suberylglycine (SG), and the ratios HG/AG, OC/AG and SG/AG were excellent markers of MCADD deficiency. CONCLUSION: This method shows potential application as a second tier screen in order to reduce the false positive rate for a number of IEM targeted by newborn screening.


Subject(s)
Dried Blood Spot Testing , Glycine/blood , Metabolism, Inborn Errors/blood , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Dried Blood Spot Testing/instrumentation , Dried Blood Spot Testing/methods , Female , Humans , Infant, Newborn , Male , Metabolism, Inborn Errors/diagnosis , Tandem Mass Spectrometry/instrumentation , Tandem Mass Spectrometry/methods
11.
Anal Chem ; 90(1): 801-806, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29190072

ABSTRACT

Newborn screening programs have expanded to include molecular-based assays as first-tier tests and the success of these assays depends on the quality and yield of DNA extracted from neonatal dried blood spots (DBS). To meet high throughput and rapid turnaround time requirements, newborn screening laboratories adopted rapid DNA extraction methods that produce crude extracts. Quantification of DNA in neonatal DBS is not routinely performed due to technical challenges; however, this may enhance the performance of assays that are sensitive to amounts of input DNA. In this study, we developed a novel high throughput method to quantify total DNA in DBS. It is based on specific acid-catalyzed depurination of DNA followed by mass spectrometric quantification of adenine. The amount of adenine was used to calculate DNA quantity per 3.2 mm DBS. Reference intervals were established using archived, neonatal DBS (n = 501) and a median of 130.6 ng of DNA per DBS was obtained, which is in agreement with literature values. The intra- and interday variations were <15%. The limits of detection and quantification were 12.5 and 37.8 nmol/L adenine, respectively. We demonstrated that DNA from neonatal DBS can be successfully quantified in high throughput settings using instruments currently deployed in NBS laboratories.


Subject(s)
DNA/blood , Dried Blood Spot Testing/methods , DNA/chemistry , Humans , Infant, Newborn , Limit of Detection , Neonatal Screening/methods , Tandem Mass Spectrometry
12.
Mol Genet Metab ; 123(3): 309-316, 2018 03.
Article in English | MEDLINE | ID: mdl-29269105

ABSTRACT

Serine biosynthesis defects are autosomal recessive metabolic disorders resulting from the deficiency of any of the three enzymes involved in de novo serine biosynthesis, specifically phosphoglycerate dehydrogenase (PGDH), phosphoserine aminotransferase (PSAT), and phosphoserine phosphatase (PSP). In this study, we performed metabolomic profiling on 4 children with serine biosynthesis defects; 3 with PGDH deficiency and 1 with PSAT deficiency. The evaluations were performed at baseline and with serine and glycine supplementation. Metabolomic profiling performed at baseline showed low phospholipid species, including glycerophosphocholine, glycerophosphoethanolamine, and sphingomyelin. All children had low serine and glycine as expected. Low glycerophosphocholine compounds were found in 4 children, low glycerophosphoethanolamine compounds in 3 children, and low sphingomyelin species in 2 children. Metabolic profiling with serine and glycine supplementation showed normalization of most of the low phospholipid compounds in the 4 children. Phospholipids are the major component of plasma and intracellular membranes, and phosphatidylcholine is the most abundant phospholipid of all mammalian cell types and subcellular organelles. Phosphatidylcholine is of particular importance for the nervous system, where it is essential for neuronal differentiation. The observed low phosphatidylcholine species in children with serine biosynthesis defects that improved after serine supplementation, supports the role of serine as a significant precursor for phosphatidylcholine. The vital role that phosphatidylcholine has during neuronal differentiation and the pronounced neurological manifestations in serine biosynthesis defects suggest that phosphatidylcholine deficiency occurring secondary to serine deficiency may have a significant contribution to the development of the neurological manifestations in individuals with serine biosynthesis defects.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/metabolism , Dietary Supplements , Glycine/administration & dosage , Microcephaly/metabolism , Phosphatidylcholines/metabolism , Phosphoglycerate Dehydrogenase/deficiency , Psychomotor Disorders/metabolism , Seizures/metabolism , Serine/biosynthesis , Transaminases/deficiency , Carbohydrate Metabolism, Inborn Errors/blood , Carbohydrate Metabolism, Inborn Errors/diet therapy , Cell Differentiation , Child , Child, Preschool , Female , Glycine/blood , Humans , Infant , Male , Metabolomics/methods , Microcephaly/blood , Microcephaly/diet therapy , Neurons/metabolism , Phosphoglycerate Dehydrogenase/blood , Phosphoglycerate Dehydrogenase/metabolism , Psychomotor Disorders/blood , Psychomotor Disorders/diet therapy , Seizures/blood , Seizures/diet therapy , Serine/administration & dosage , Serine/blood , Transaminases/blood , Transaminases/metabolism
13.
JIMD Rep ; 39: 89-96, 2018.
Article in English | MEDLINE | ID: mdl-28766179

ABSTRACT

Glutaric aciduria type 3 (GA3) is associated with decreased conversion of free glutaric acid to glutaryl-coA, reflecting deficiency of succinate-hydroxymethylglutarate coA-transferase, caused by variants in the SUGCT (C7orf10) gene. GA3 remains less well known, characterised and understood than glutaric aciduria types 1 and 2. It is generally considered a likely "non-disease," but this is based on limited supporting information, with only nine individuals with GA3 described in the literature. Clinicians encountering a patient with GA3 therefore still face a dilemma of whether or not this should be dismissed as irrelevant.We have identified three unrelated Canadian patients with GA3. Two came to clinical attention because of symptoms, while the third was identified by a population urine-based newborn screening programme and has so far remained asymptomatic. We describe the clinical histories, biochemical characterisation and genotypes of these individuals. Examination of allele frequencies underlines the fact that GA3 is underdiagnosed. While one probable factor is that some GA3 patients remain asymptomatic, we highlight other plausible reasons whereby this diagnosis might be overlooked.Gastrointestinal disturbances were previously reported in some GA3 patients. In one of our patients, severe episodes of cyclic vomiting were the major problem. A trial of antibiotic treatment, to minimise bacterial GA production, was followed by significant clinical improvement.At present, there is insufficient evidence to define any specific clinical phenotype as attributable to GA3. However, we consider that it would be premature to assume that this condition is completely benign in all individuals at all times.

14.
Anal Chem ; 89(15): 8112-8121, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28648083

ABSTRACT

Mass spectrometry (MS)-based metabolomic initiatives that use conventional separation techniques are limited by low sample throughput and complicated data processing that contribute to false discoveries. Herein, we introduce a new strategy for unambiguous identification and accurate quantification of biomarkers for inborn errors of metabolism (IEM) from dried blood spots (DBS) with quality assurance. A multiplexed separation platform based on multisegment injection-capillary electrophoresis-mass spectrometry (MSI-CE-MS) was developed to provide comparable sample throughput to flow injection analysis-tandem MS (FIA-MS/MS) but with greater selectivity as required for confirmatory testing and discovery-based metabolite profiling of volume-restricted biospecimens. Mass spectral information is encoded temporally within a separation by serial injection of three or more sample pairs, each having a unique dilution pattern, alongside a quality control (QC) that serves as a reference in every run to facilitate between-sample comparisons and/or batch correction due to system drift. Optimization of whole blood extraction conditions on DBS filter paper cut-outs was first achieved to maximize recovery of a wide range of polar metabolites from DBS extracts. An interlaboratory comparison study was also conducted using a proficiency test and retrospective neonatal DBS that demonstrated good agreement between MSI-CE-MS and validated FIA-MS/MS methods within an accredited facility. Our work demonstrated accurate identification of various IEM based on reliable measurement of a panel of primary or secondary biomarkers above an upper cutoff concentration limit for presumptive screen-positive cases without stable isotope-labeled reagents. Additionally, nontargeted metabolite profiling by MSI-CE-MS with temporal signal pattern recognition revealed new biomarkers for early detection of galactosemia, such as N-galactated amino acids, that are a novel class of pathognomonic marker due to galactose stress in affected neonates.


Subject(s)
Biomarkers/analysis , Metabolism, Inborn Errors/diagnosis , Spectrometry, Mass, Electrospray Ionization/methods , Dried Blood Spot Testing , Electrophoresis, Capillary , Humans , Infant, Newborn , Metabolism, Inborn Errors/metabolism , Quality Control , Signal Processing, Computer-Assisted , Spectrometry, Mass, Electrospray Ionization/standards
15.
J Paediatr Child Health ; 53(6): 585-591, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28337809

ABSTRACT

AIM: To address the implementation of the National Newborn Screening Program (NBS) in Saudi Arabia and stratify the incidence of the screened disorders. METHODS: A retrospective study conducted between 1 August 2005 and 31 December 2012, total of 775 000 newborns were screened from 139 hospitals distributed among all regions of Saudi Arabia. The NBS Program screens for 16 disorders from a selective list of inborn errors of metabolism (IEM) and endocrine disorders. Heel prick dry blood spot samples were obtained from all newborns for biochemical and immunoassay testing. Recall screening testing was performed for Initial positive results and confirmed by specific biochemical assays. RESULTS: A total of 743 cases were identified giving an overall incidence of 1:1043. Frequently detected disorders nationwide were congenital hypothyroidism and congenital adrenal hyperplasia with an incidence of 1:7175 and 1:7908 correspondingly. The highest incidence among the IEM was propionic acidaemia with an incidence rate of 1:14 000. CONCLUSION: The article highlights the experience of the NBS Program in Saudi Arabia and providing data on specific regional incidences of all the screened disorders included in the programme; and showed that the incidence of these disorders is one of the highest reported so far world-wide.


Subject(s)
Endocrine System Diseases/diagnosis , Infant, Newborn, Diseases/diagnosis , Metabolism, Inborn Errors/diagnosis , Neonatal Screening/organization & administration , Databases, Factual , Developing Countries , Endocrine System Diseases/epidemiology , Female , Humans , Incidence , Infant, Newborn , Infant, Newborn, Diseases/epidemiology , Male , Metabolism, Inborn Errors/epidemiology , Program Evaluation , Retrospective Studies , Risk Assessment , Saudi Arabia , Severity of Illness Index
16.
J Med Screen ; 24(2): 58-61, 2017 06.
Article in English | MEDLINE | ID: mdl-27216769

ABSTRACT

Background Newborn screening for propionic acidemia and methylmalonic acidurias using the marker propionylcarnitine (C3) is neither sensitive nor specific. Using C3 to acetylcarnitine (C3/C2) ratio, together with conservative C3 cut-offs, can improve screening sensitivity, but the false positive rate remains high. Incorporating the marker 2-methylcitric acid has been suggested, to improve the positive predictive value for these disorders without compromising the sensitivity. Methods Between July 2011 and December 2012 at the Newborn Screening Ontario laboratory, all neonatal dried blood spot samples that were reported as screen positive for propionic acidemia or methylmalonic acidurias based on elevated C3 and C3/C2 ratio were analyzed for 2-methylcitric acid, using liquid chromatography tandem mass spectrometry. Results Of 222,420 samples screened, 103 were positive for methylmalonic acidurias or propionic acidemia using C3 and C3/C2 ratio as markers. There were nine true positives: propionic acidemia (n = 3), Cobalamin (Cbl) A (n=1), and Cbl C (n = 5). Among false positives there were 72 neonates not affected, 20 with maternal B12 deficiency, and two incidental finding (transcobalamin II and unclassified Cbl defect). 2-Methylcitric acid was analyzed in all 103 samples and ranged between 0.1 and 89.4 µmol/l (reference range 0.04-0.36). Only 14 samples exceeded the set 2-methylcitric acid cut-off of 1.0 µmol/l, including the samples from all nine true positives. Conclusion By including 2-methylcitric acid in the screening algorithm, the positive predictive value of our primary and secondary screening targets improved from 8.7 to 64.3%. This would have eliminated 89 unnecessary referrals while maintaining 100% sensitivity.


Subject(s)
Amino Acid Metabolism, Inborn Errors/blood , Citrates/blood , Dried Blood Spot Testing , Neonatal Screening/methods , Acetylcarnitine/blood , Algorithms , Amino Acid Metabolism, Inborn Errors/diagnosis , Biomarkers/blood , Carnitine/analogs & derivatives , Carnitine/blood , Chromatography, Liquid , False Positive Reactions , Humans , Infant, Newborn , Methylmalonic Acid/blood , Predictive Value of Tests , Reference Values , Referral and Consultation , Reproducibility of Results , Tandem Mass Spectrometry
17.
Mol Genet Metab Rep ; 9: 54-60, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27761413

ABSTRACT

Arginase-1 (Arg1) converts arginine to urea and ornithine in the distal step of the urea cycle in liver. We previously generated a tamoxifen-inducible Arg1 deficient mouse model (Arg1-Cre) that disrupts Arg1 expression throughout the whole body and leads to lethality ≈ 2 weeks after gene disruption. Here, we evaluate if liver-selective Arg1 loss is sufficient to recapitulate the phenotype observed in global Arg1 knockout mice, as well as to gauge the effectiveness of gene delivery or hepatocyte transplantation to rescue the phenotype. Liver-selective Arg1 deletion was induced by using an adeno-associated viral (AAV)-thyroxine binding globulin (TBG) promoter-Cre recombinase vector administered to Arg1 "floxed" mice; Arg1fl/fl ). An AAV vector expressing an Arg1-enhanced green fluorescent protein (Arg1-eGFP) transgene was used for gene delivery, while intrasplenic injection of wild-type (WT) C57BL/6 hepatocytes after partial hepatectomy was used for cell delivery to "rescue" tamoxifen-treated Arg1-Cre mice. The results indicate that liver-selective loss of Arg1 (> 90% deficient) leads to a phenotype resembling the whole body knockout of Arg1 with lethality ≈ 3 weeks after Cre-induced gene disruption. Delivery of Arg1-eGFP AAV rescues more than half of Arg1 global knockout male mice (survival > 4 months) but a significant proportion still succumb to the enzyme deficiency even though liver expression and enzyme activity of the fusion protein reach levels observed in WT animals. Significant Arg1 enzyme activity from engrafted WT hepatocytes into knockout livers can be achieved but not sufficient for rescuing the lethal phenotype. This raises a conundrum relating to liver-specific expression of Arg1. On the one hand, loss of expression in this organ appears to be both necessary and sufficient to explain the lethal phenotype of the genetic disorder in mice. On the other hand, gene and cell-directed therapies suggest that rescue of extra-hepatic Arg1 expression may also be necessary for disease correction. Further studies are needed in order to illuminate the detailed mechanisms for pathogenesis of Arg1-deficiency.

18.
Am J Physiol Cell Physiol ; 311(4): C559-C571, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27488667

ABSTRACT

The skeletal muscle ATP-sensitive K+ (KATP) channel is crucial in preventing fiber damage and contractile dysfunction, possibly by preventing damaging ATP depletion. The objective of this study was to investigate changes in energy metabolism during fatigue in wild-type and inwardly rectifying K+ channel (Kir6.2)-deficient (Kir6.2-/-) flexor digitorum brevis (FDB), a muscle that lacks functional KATP channels. Fatigue was elicited with one tetanic contraction every second. Decreases in ATP and total adenylate levels were significantly greater in wild-type than Kir6.2-/- FDB during the last 2 min of the fatigue period. Glycogen depletion was greater in Kir6.2-/- FDB for the first 60 s, but not by the end of the fatigue period, while there was no difference in glucose uptake. The total amount of glucosyl units entering glycolysis was the same in wild-type and Kir6.2-/- FDB. During the first 60 s, Kir6.2-/- FDB generated less lactate and more CO2; in the last 120 s, Kir6.2-/- FDB stopped generating CO2 and produced more lactate. The ATP generated during fatigue from phosphocreatine, glycolysis (lactate), and oxidative phosphorylation (CO2) was 3.3-fold greater in Kir6.2-/- than wild-type FDB. Because ATP and total adenylate were significantly less in Kir6.2-/- FDB, it is suggested that Kir6.2-/- FDB has a greater energy deficit, despite a greater ATP production, which is further supported by greater glucose uptake and lactate and CO2 production in Kir6.2-/- FDB during the recovery period. It is thus concluded that a lack of functional KATP channels results in an impairment of energy metabolism.


Subject(s)
Energy Metabolism/physiology , KATP Channels/deficiency , KATP Channels/metabolism , Muscle Fatigue/physiology , Muscle, Skeletal/metabolism , Adenosine Triphosphate/metabolism , Animals , Carbon Dioxide/metabolism , Glycolysis/physiology , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Oxidative Phosphorylation , Potassium Channels, Inwardly Rectifying/metabolism
19.
JIMD Rep ; 30: 73-79, 2016.
Article in English | MEDLINE | ID: mdl-27306203

ABSTRACT

Mutations of the mitochondrial citrate carrier (CIC) SLC25A1 cause combined D-2- and L-2-hydroxyglutaric aciduria (DL-2HGA; OMIM #615182), a neurometabolic disorder characterized by developmental delay, hypotonia, and seizures. Here, we describe the female child of consanguineous parents who presented neonatally with lactic acidosis, periventricular frontal lobe cysts, facial dysmorphism, recurrent apneic episodes, and deficient complex IV (cytochrome c oxidase) activity in skeletal muscle. Exome sequencing revealed a homozygous SLC25A1 missense mutation [NM_005984.4: c.593G>A; p.(Arg198His)] of a ubiquitously conserved arginine residue putatively situated within the substrate-binding site I of CIC. Retrospective review of the patient's organic acids confirmed the D- and L-2-hydroxyglutaric aciduria typical of DL-2HGA to be present, although this was not appreciated on initial presentation. Cultured patient skin fibroblasts showed reduced survival in culture, diminished mitochondrial spare respiratory capacity, increased glycolytic flux, and normal mitochondrial bulk, inner membrane potential, and network morphology. Neither cell survival nor cellular respiratory parameters were improved by citrate supplementation, although oral citrate supplementation did coincide with amelioration of lactic acidosis and apneic attacks in the patient. This is the fifth clinical report of CIC deficiency to date. The clinical features in our patient suggest that this disorder, which can potentially be recognized either by molecular means or based on its characteristic organic aciduria, should be considered in the differential diagnosis of pyruvate dehydrogenase deficiency and respiratory chain disorders. One-Sentence Summary A novel homozygous missense substitution in SLC25A1 was identified in a neonate presenting with lactic acidosis, intracerebral cysts, and an apparent mitochondrial complex IV defect in muscle.

20.
Mol Genet Metab ; 116(3): 146-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26361991

ABSTRACT

Untreated profound biotinidase deficiency results in a wide range of clinical features, including optic atrophy, cutaneous abnormalities, hearing loss and developmental delay. Ontario, Canada incorporated this treatable deficiency in newborn screening over the past 8years. This study elucidates the molecular, biochemical, and clinical findings from the pilot project. Information from initial screens, serum biotinidase activity level assays, molecular testing, and family history for 246 positive newborns screens were analyzed. A mutation spectrum was created for the province of Ontario, including common mutations such as D444H, D444H/A171T, Q456H, C33fs, and R157H. Individuals with partial deficiency were separated into 3 groups: D444H homozygotes (Group 1); compound heterozygotes for D444H with another profound allele (Group 2); compound heterozygotes with two non-D444H alleles (Group 3). Biochemical phenotype-genotype associations in partial deficiency showed a significant difference in serum biotinidase activity in between any given two groups. Three children with partial deficiency discontinued biotin for varied lengths of time. Two of whom became symptomatic with abnormal gait, alopecia, skin rashes and developmental delay. A need for more congruency in diagnostic, treatment and educational practices was highlighted across the province. Heterogeneity and variation in clinical presentations and management was observed in patients with the partial deficiency.


Subject(s)
Biotinidase Deficiency/enzymology , Biotinidase Deficiency/genetics , Neonatal Screening , Alleles , Amidohydrolases/genetics , Biotin/therapeutic use , Biotinidase/blood , Biotinidase/genetics , Biotinidase Deficiency/diagnosis , Biotinidase Deficiency/epidemiology , Child , Child, Preschool , Disease Management , Female , Genetic Association Studies , Hearing Loss/etiology , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Male , Mutation , Ontario/epidemiology , Pilot Projects
SELECTION OF CITATIONS
SEARCH DETAIL