Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Med Genet ; 52(4): 262-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25596309

ABSTRACT

BACKGROUND: Fabry disease (FD), a lysosomal storage disorder caused by α-galactosidase A (GLA) gene variants, has a heterogeneous phenotype. GLA variants can lead to classical FD, an attenuated non-classical phenotype, or no disease at all. This study investigates the value of plasma globotriaosylsphingosine (lysoGb3) to distinguish between these groups. This is of particular importance in the diagnosis of individuals with a GLA variant and an uncertain diagnosis of FD, lacking characteristic features of classical FD. METHODS: Subjects with GLA variants were grouped as classical, non-classical, uncertain or no FD, using strict phenotypical, biochemical and histological criteria. Plasma lysoGb3 was assessed by LC/MS/MS (normal ≤ 0.6 nmol/L). RESULTS: 154 subjects were grouped into classical (38 males (M), 66 females (F)), non-classical (13 M, 14 F), uncertain (5M, 9 F) or no FD (6M, 3F). All subjects with a classical phenotype had elevated lysoGb3 values (M: range 45-150, F: 1.5-41.5). LysoGb3 values in patients with a non-classical phenotype (M: 1.3-35.7, F: 0.5-2.0) were different from healthy controls (M: p<0.01, F: p<0.05), but females overlapped with controls. In the no-FD group, lysoGb3 was normal. CONCLUSIONS: LysoGb3 is a reliable diagnostic tool to discern classical FD from subjects without FD. This study suggests that the same applies to patients with a non-classical phenotype. LysoGb3 values of female patients overlap with controls. Consequently, in uncertain cases, increased lysoGb3 values are very suggestive for FD, but normal values cannot exclude FD. Confirmation in larger cohorts and data on the specificity of small lysoGb3 increases are necessary.


Subject(s)
Fabry Disease/blood , Glycolipids/blood , Sphingolipids/blood , Adolescent , Adult , Aged , Fabry Disease/genetics , Female , Humans , Male , Middle Aged , Phenotype , Trihexosylceramides/blood , Young Adult , alpha-Galactosidase/blood
2.
J Lipid Res ; 56(4): 936-43, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25632048

ABSTRACT

Sulfatides are found in brain as components of myelin, oligodendrocytes, and neurons but are also present in various visceral tissues. Metachromatic leukodystrophy (MLD) is an inherited lysosomal storage disorder caused by a deficiency of arylsulfatase A, leading to severe white matter disease due to the accumulation of sulfatides and lysosulfatides. To study the physiological role of sulfatides, accessible and sensitive quantitative methods are required. We developed a sensitive LC/MS/MS method to quantify total sulfatide and lysosulfatide content as well as individual molecular species in urine and plasma from MLD patients and plasma and tissues from an MLD mouse model. Our results demonstrate that the method can quantify a wide range of sulfatide concentrations and can be used to quantify total sulfatide content and levels of individual molecular species of sulfatides in tissues, cells, and body fluids. Even though plasma sulfatides and lysosulfatides would seem attractive candidate biomarkers that could possibly correlate with the severity of MLD and be of use to monitor the effects of therapeutic intervention, our results indicate that it is unlikely that the determination of these storage products in plasma will be useful in this respect.


Subject(s)
Blood Chemical Analysis/methods , Psychosine/analogs & derivatives , Sulfoglycosphingolipids/blood , Sulfoglycosphingolipids/urine , Urinalysis/methods , Adolescent , Adult , Aged , Animals , Child , Child, Preschool , Chromatography, Liquid , Female , Humans , Infant , Infant, Newborn , Leukodystrophy, Metachromatic/blood , Leukodystrophy, Metachromatic/pathology , Leukodystrophy, Metachromatic/urine , Male , Mice , Middle Aged , Psychosine/blood , Psychosine/urine , Tandem Mass Spectrometry , Young Adult
3.
Clin Chem ; 59(3): 547-56, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23237761

ABSTRACT

BACKGROUND: Biochemical markers that accurately reflect the severity and progression of disease in patients with Fabry disease and their response to treatment are urgently needed. Globotriaosylsphingosine, also called lysoglobotriaosylceramide (lysoGb3), is a promising candidate biomarker. METHODS: We synthesized lysoGb3 and isotope-labeled [5,6,7,8,9] (13)C5-lysoGb3 (internal standard). After addition of the internal standard to 25 µL plasma or 400 µL urine from patients with Fabry disease and healthy controls, samples were extracted with organic solvents and the lysoGb3 concentration was quantified by UPLC-ESI-MS/MS (ultraperformance liquid chromatography-electrospray ionization-tandem mass spectrometry). Calibration curves were constructed with control plasma and urine supplemented with lysoGb3. In addition to lysoGb3, lyso-ene-Gb3 was quantified. Quantification was achieved by multiple reaction monitoring of the transitions m/z 786.4 > 282.3 [M+H](+) for lysoGb3, m/z 791.4 > 287.3 [M+H](+) for [5,6,7,8,9] (13)C5-lysoGb3, and 784.4 > 280.3 [M+H](+) for lyso-ene-Gb3. RESULTS: The mean (SD) plasma lysoGb3 concentration from 10 classically affected Fabry hemizygotes was 94.4 (25.8) pmol/mL (range 52.7-136.8 pmol/mL), from 10 classically affected Fabry heterozygotes 9.6 (5.8) pmol/mL (range 4.1-23.5 pmol/mL), and from 20 healthy controls 0.4 (0.1) pmol/mL (range 0.3-0.5 pmol/mL). Lyso-ene-Gb3 concentrations were 10%-25% of total lysoGb3. The urine concentration of lysoGb3 was 40-480 times lower than in corresponding plasma samples. Lyso-ene-Gb3 concentrations in urine were comparable or even higher than the corresponding lysoGb3 concentrations. CONCLUSIONS: This assay for the quantification of lysoGb3 and lyso-ene-Gb3 in human plasma and urine samples will be an important tool in the diagnosis of Fabry disease and for monitoring the effect of enzyme replacement therapy in patients with Fabry disease.


Subject(s)
Chromatography, Liquid/methods , Fabry Disease/diagnosis , Glycolipids/analysis , Sphingolipids/analysis , Tandem Mass Spectrometry/methods , Adult , Calibration , Carbon Isotopes , Humans , Isotope Labeling , Middle Aged , Reproducibility of Results
4.
Anal Bioanal Chem ; 403(6): 1671-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22526647

ABSTRACT

Many lysosomal storage diseases are characterized by an increased urinary excretion of glycoconjugates and oligosaccharides that are characteristic for the underlying enzymatic defect. Here, we have used capillary high-performance anion-exchange chromatography (HPAEC) hyphenated to mass spectrometry to analyze free oligosaccharides from urine samples of patients suffering from the lysosomal storage disorders fucosidosis, α-mannosidosis, G(M1)-gangliosidosis, G(M2)-gangliosidosis, and sialidosis. Glycan fingerprints were registered, and the patterns of accumulated oligosaccharides were found to reflect the specific blockages of the catabolic pathway. Our analytical approach allowed structural analysis of the excreted oligosaccharides and revealed several previously unpublished oligosaccharides. In conclusion, using online coupling of HPAEC with mass spectrometric detection, our study provides characteristic urinary oligosaccharide fingerprints with diagnostic potential for lysosomal storage disorders.


Subject(s)
Chromatography, High Pressure Liquid/methods , Chromatography, Ion Exchange/methods , Lysosomal Storage Diseases/urine , Mass Spectrometry/methods , Oligosaccharides/urine , Anion Exchange Resins , Humans
5.
Hum Mutat ; 31(5): E1348-60, 2010 May.
Article in English | MEDLINE | ID: mdl-20232353

ABSTRACT

Mucopolysaccharidosis III D (Sanfilippo disease type D, MPS IIID) is a rare autosomal recessive lysosomal storage disorder previously described in only 20 patients. MPS IIID is caused by a deficiency of N-acetylglucosamine-6-sulphate sulphatase (GNS), one of the enzymes required for the degradation of heparan sulphate. So far only seven mutations in the GNS gene have been reported. The clinical phenotype of 12 new MPS IIID patients from 10 families was studied. Mutation analysis of GNS was performed in 16 patients (14 index cases). Clinical signs and symptoms of the MPS IIID patients appeared to be similar to previously described patients with MPS III. Early development was normal with onset of behavioral problems around the age of 4 years, followed by developmental stagnation, deterioration of verbal communication and subsequent deterioration of motor functions. Sequence analysis of the coding regions of the gene encoding GNS (GNS) resulted in the identification of 15 novel mutations: 3 missense mutations, 1 nonsense mutation, 4 splice site mutations, 3 frame shift mutations, 3 large deletions and 1 in-frame small deletion. They include the first missense mutations and a relatively high proportion of large rearrangements, which warrants the inclusion of quantitative techniques in routine mutation screening of the GNS gene.


Subject(s)
Mucopolysaccharidosis III/genetics , Sulfatases/deficiency , Sulfatases/genetics , Adolescent , Adult , Child , Child, Preschool , DNA Mutational Analysis , Female , Humans , Male , Mutation/genetics , Phenotype , Young Adult
6.
Eur J Pediatr ; 167(11): 1305-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18521627

ABSTRACT

Clara cell protein (CC16) is an anti-inflammatory protein and a biomarker of pulmonary epithelial cells and alveolocapillary membrane injury in adults. We investigated whether low cord blood concentrations of CC16 are associated with the development of respiratory distress syndrome (RDS) and bronchopulmonary dysplasia (BPD) in preterm infants and the relationship between CC16 and its pro-inflammatory counterpart, the secretory phospholipase A(2) (sPLA(2)) enzyme. CC16 concentration, sPLA(2) activity and IL-6 concentration were measured in cord blood plasma from 79 preterm infants (25 controls, 37 infants who developed RDS and 17 infants who developed BPD). After adjustment for gestational age and Apgar score at 5 min, the CC16 concentration was lower in BPD infants than in preterm controls (p<0.01). sPLA(2) activity was similar in all groups and the IL-6 concentrations were increased in both RDS and BPD infants (p<0.01 and p<0.05, respectively, vs. controls). We conclude that low cord blood CC16 concentrations in preterm infants independently predict the development of BPD. Low CC16 levels may reflect early lung injury, which contributes to the severity of RDS and progress towards BPD. Future studies are needed to assess whether the early administration of recombinant human CC16 in preterm infants with low cord blood CC16 prevents the development of BPD.


Subject(s)
Bronchopulmonary Dysplasia/genetics , Fetal Blood/metabolism , Uteroglobin/genetics , Uteroglobin/metabolism , Apgar Score , Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/epidemiology , Female , Fetal Blood/immunology , Gestational Age , Humans , Incidence , Infant, Newborn , Infant, Premature , Interleukin-6/immunology , Phospholipases A2, Secretory/metabolism , Predictive Value of Tests , Prospective Studies
7.
Free Radic Biol Med ; 36(6): 782-801, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14990357

ABSTRACT

Oxidative stress is an important factor in the pathogenesis of bronchopulmonary dysplasia (BPD), a chronic lung disease of premature infants characterized by arrested alveolar and vascular development of the immature lung. We investigated differential gene expression with DNA microarray analysis in premature rat lungs exposed to prolonged hyperoxia during the saccular stage of development, which closely resembles the development of the lungs of premature infants receiving neonatal intensive care. Expression profiles were largely confirmed by real-time RT-PCR (27 genes) and in line with histopathology and fibrin deposition studied by Western blotting. Oxidative stress affected a complex orchestra of genes involved in inflammation, coagulation, fibrinolysis, extracellular matrix turnover, cell cycle, signal transduction, and alveolar enlargement and explains, at least in part, the pathological alterations that occur in lungs developing BPD. Exciting findings were the magnitude of fibrin deposition; the upregulation of chemokine-induced neutrophilic chemoattractant-1 (CINC-1), monocyte chemoattractant protein-1 (MCP-1), amphiregulin, plasminogen activator inhibitor-1 (PAI-1), secretory leukocyte proteinase inhibitor (SLPI), matrix metalloproteinase-12 (MMP12), p21, metallothionein, and heme oxygenase (HO); and the downregulation of fibroblast growth factor receptor-4 (FGFR4) and vascular endothelial growth factor (VEGF) receptor-2 (Flk-1). These findings are not only of fundamental importance in the understanding of the pathophysiology of BPD, but also essential for the development of new therapeutic strategies.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Lung/pathology , Oxidative Stress , RNA, Messenger/metabolism , Animals , Animals, Newborn , Blood Coagulation Factors/genetics , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibrin/metabolism , Fibrinolysis/genetics , Gene Expression Profiling , Humans , Infant, Newborn , Inflammation/genetics , Inflammation/metabolism , Lung/metabolism , Rats , Receptor, Fibroblast Growth Factor, Type 4 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
J Appl Physiol (1985) ; 97(5): 2014-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15208286

ABSTRACT

Bronchopulmonary dysplasia is a leading cause of mortality and morbidity in preterm infants despite improved treatment modalities. Pentoxifylline, a phosphodiesterase inhibitor, inhibits multiple processes that lead to neonatal hyperoxic lung injury, including inflammation, coagulation, and edema. Using a preterm rat model, we investigated the effects of pentoxifylline on hyperoxia-induced lung injury and survival. Preterm rat pups were exposed to 100% oxygen and injected subcutaneously with 0.9% saline or 75 mg/kg pentoxifylline twice a day. On day 10, lung tissue was harvested for histology, fibrin deposition, and mRNA expression, and bronchoalveolar lavage fluid was collected for total protein concentration. Pentoxifylline treatment increased mean survival by 3 days (P = 0.0018) and reduced fibrin deposition by 66% (P < 0.001) in lung homogenates compared with untreated hyperoxia-exposed controls. Monocyte chemoattractant protein-1 expression in lung homogenates was decreased, but the expressions of TNF-alpha, IL-6, matrix metalloproteinase-12, tissue factor, and plasminogen activator inhibitor-1 were similar in both groups. Total protein concentration in bronchoalveolar lavage fluid was decreased by 33% (P = 0.029) in the pentoxifylline group. Pentoxifylline treatment attenuates alveolar fibrin deposition and prolongs survival in preterm rat pups with neonatal hyperoxic lung injury, probably by reducing capillary-alveolar protein leakage.


Subject(s)
Animals, Newborn , Fibrin/antagonists & inhibitors , Hyperoxia/pathology , Hyperoxia/physiopathology , Lung/pathology , Pentoxifylline/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CCL2/genetics , Hyperoxia/metabolism , Leukocyte Count , Lung/drug effects , Lung/metabolism , RNA, Messenger/antagonists & inhibitors , Rats , Rats, Wistar , Survival Analysis
9.
PLoS One ; 7(10): e47805, 2012.
Article in English | MEDLINE | ID: mdl-23094092

ABSTRACT

INTRODUCTION: Enzyme replacement therapy (ERT) with alpha-Galactosidase A (aGal A) may cause antibody (AB) formation against aGal A in males with Fabry disease (FD). Anti agalsidase ABs negatively influence globotriaosylceramide (Gb3) reduction. We investigated the impact of agalsidase AB on Gb3 and lysoGb3 and clinical outcome in Fabry patients on ERT. METHODS: Adult male and female patients on ERT for at least one year were included. Urinary Gb3 was measured by HPLC, plasma lysoGb3 by LC-ESI-MS/MS and AB with a neutralization assay. RESULTS: Of the 59 patients evaluable patients, 0/30 females and 17/29 males developed anti-agalsidase antibodies (AB+). Only 3/17 males had transient (low) titers (tolerized). All AB+ patients developed antibodies during the first year of treatment. Change of agalsidase preparation (or dose) did not induce antibody formation. AB+ males had significant less decline in plasma lysoGb3 compared to AB- males (p = 0.04). Urinary Gb3 levels decreased markedly in AB- but remained comparable to baseline in AB+ males (p<0.01). (Lyso)Gb3 reduction in plasma and urine on ERT was correlated with LVmass reduction in females and development white matter lesions and stroke. CONCLUSION: In male patients antibodies against aGal A remained present up to 10 years of ERT. The presence of these antibodies is associated with a less robust decrease in plasma lysoGb3 and a profound negative effect on urinary Gb3 reduction, which may reflect worse treatment outcome.


Subject(s)
Antibodies/blood , Fabry Disease/drug therapy , Fabry Disease/immunology , Globosides/urine , Glycolipids/blood , Sphingolipids/blood , Trihexosylceramides/urine , alpha-Galactosidase/therapeutic use , Adult , Chromatography, High Pressure Liquid , Chromatography, Liquid , Enzyme Replacement Therapy , Fabry Disease/blood , Fabry Disease/urine , Female , Humans , Male , Middle Aged , Neutralization Tests , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Time , Treatment Outcome , alpha-Galactosidase/pharmacology
10.
Orphanet J Rare Dis ; 6: 69, 2011 Oct 31.
Article in English | MEDLINE | ID: mdl-22041095

ABSTRACT

BACKGROUND: Enzyme replacement therapy is currently the only approved therapy for Fabry disease. From June 2009 on, viral contamination of Genzyme's production facility resulted in a worldwide shortage of agalsidase beta leading to involuntary dose reductions (approved dose 1 mg/kg/eow, reduced dose 0.5 mg/kg/m), or switch to agalsidase alpha (administered dose 0.2 mg/kg/eow). An assessment report from the European Medicines Agency (EMA) raised serious concerns about an increase in adverse events at lower dosages of agalsidase beta. We determined the influence of the shortage on clinical event incidence and the most sensitive biochemical marker (lysoGb3) in Dutch Fabry patients. METHODS: The incidence of clinical events per person per year was calculated from start of agalsidase beta treatment until the shortage, and was compared to the incidence of clinical events during the shortage period. In addition, plasma lysoGb3, eGFR, quality of life (SF-36) and brief pain inventory (BPI) questionnaires were analysed. RESULTS: All thirty-five Dutch Fabry patients using agalsidase beta (17 males) were included. Mean clinical event incidence was unchanged: 0.15 events per person per year before versus 0.15 during the shortage (p = 0.68). In total 28 clinical events occurred in 14 patients during 4.6 treatment years, compared to 7 events in 6 patients during the 1.3 year shortage period. eGFR and BPI scores were not significantly altered. Two SF-36 subscales were significantly but minimally reduced in females. In males, lysoGb3 increased with a median of 8.1 nM (range 2.5-29.2) after 1 year of shortage (p = 0.001). Increases in lysoGb3 were found in both patients switching to agalsidase alpha and on a reduced agalsidase beta dose. Antibody status, treatment duration or clinical event incidence showed no clear correlation to lysoGb3 increases. CONCLUSIONS: No increase in clinical event incidence was found in the adult Dutch Fabry cohort during the agalsidase beta shortage. Increases in lysoGb3, however, suggest recurrence of disease activity.


Subject(s)
Disease Progression , Fabry Disease/drug therapy , Fabry Disease/physiopathology , Glycolipids/blood , Isoenzymes/administration & dosage , Isoenzymes/supply & distribution , Sphingolipids/blood , alpha-Galactosidase/administration & dosage , alpha-Galactosidase/supply & distribution , alpha-Galactosidase/therapeutic use , Adolescent , Adult , Aged , Drug Administration Schedule , Enzyme Replacement Therapy/methods , Fabry Disease/epidemiology , Female , Humans , Incidence , Isoenzymes/adverse effects , Isoenzymes/therapeutic use , Male , Middle Aged , Netherlands/epidemiology , Treatment Outcome , Young Adult , alpha-Galactosidase/adverse effects
11.
FEBS J ; 277(14): 2970-86, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20546307

ABSTRACT

Urine, amniotic fluid and ascitic fluid samples of galactosialidosis patients were analyzed and structurally characterized for free oligosaccharides using capillary high-performance anion-exchange chromatography with pulsed amperometric detection and online mass spectrometry. In addition to the expected endo-beta-N-acetylglucosaminidase-cleaved products of complex-type sialylated N-glycans, O-sulfated oligosaccharide moieties were detected. Moreover, novel carbohydrate moieties with reducing-end hexose residues were detected. On the basis of structural features such as a hexose-N-acetylhexosamine-hexose-hexose consensus sequence and di-sialic acid units, these oligosaccharides are thought to represent, at least in part, glycan moieties of glycosphingolipids. In addition, C(1)-oxidized, aldohexonic acid-containing versions of most of these oligosaccharides were observed. These observations suggest an alternative catabolism of glycosphingolipids in galactosialidosis patients: oligosaccharide moieties from glycosphingolipids would be released by a hitherto unknown ceramide glycanase activity. The results show the potential and versatility of the analytical approach for structural characterization of oligosaccharides in various body fluids.


Subject(s)
Amniotic Fluid/chemistry , Ascitic Fluid/chemistry , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/urine , Neuraminidase/deficiency , Oligosaccharides/analysis , beta-Galactosidase/deficiency , Carbohydrate Sequence , Cathepsin A/genetics , Chromatography, Ion Exchange , Fetus/metabolism , Glycoside Hydrolases/metabolism , Glycosphingolipids/metabolism , Hexoses/analysis , Humans , Infant , Infant, Newborn/metabolism , Infant, Newborn/urine , Lysosomal Storage Diseases/genetics , Mass Spectrometry , Molecular Sequence Data , Oligosaccharides/chemistry , Oligosaccharides/urine , Oligosaccharides, Branched-Chain/analysis , Oligosaccharides, Branched-Chain/chemistry , Oligosaccharides, Branched-Chain/urine , Sugar Acids/analysis , Urine/chemistry
12.
Am J Physiol Lung Cell Mol Physiol ; 293(1): L35-44, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17384081

ABSTRACT

Administration of inhaled nitric oxide (iNO) is a potential therapeutic strategy to prevent bronchopulmonary dysplasia (BPD) in premature newborns with respiratory distress syndrome. We evaluated this approach in a rat model, in which premature pups were exposed to room air, hyperoxia, or a combination of hyperoxia and NO (8.5 and 17 ppm). We investigated the anti-inflammatory effects of prolonged iNO therapy by studying survival, histopathology, fibrin deposition, and differential mRNA expression (real-time RT-PCR) of key genes involved in the development of BPD. iNO therapy prolonged median survival 1.5 days (P = 0.0003), reduced fibrin deposition in a dosage-dependent way up to 4.3-fold (P < 0.001), improved alveolar development by reducing septal thickness, and reduced the influx of leukocytes. Analysis of mRNA expression revealed an iNO-induced downregulation of genes involved in inflammation (IL-6, cytokine-induced neutrophilic chemoattractant-1, and amphiregulin), coagulation, fibrinolysis (plasminogen activator inhibitor 1 and urokinase-type plasminogen activator receptor), cell cycle regulation (p21), and an upregulation of fibroblast growth factor receptor-4 (alveolar formation). We conclude that iNO therapy improves lung pathology and prolongs survival by reducing septum thickness, inhibiting inflammation, and reducing alveolar fibrin deposition in premature rat pups with neonatal hyperoxic lung injury.


Subject(s)
Fibrin/metabolism , Lung Diseases/pathology , Nitric Oxide/administration & dosage , Nitric Oxide/therapeutic use , Pneumonia/drug therapy , Administration, Inhalation , Animals , Animals, Newborn , Antioxidants/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Cell Cycle , Fibrinolysis , Gene Expression Regulation , Lung/metabolism , Lung/pathology , Nitric Oxide/pharmacology , Proteins/genetics , Proteins/metabolism , Pulmonary Alveoli/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction , Survival Analysis
13.
Clin Chem ; 53(4): 742-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17332150

ABSTRACT

BACKGROUND: Simple, reproducible assays are needed for the quantification of sphingolipids, ceramide (Cer), and sphingoid bases. We developed an HPLC method for simultaneous quantification of total plasma concentrations of Cer, glucosylceramide (GlcCer), and ceramide trihexoside (CTH). METHODS: After addition of sphinganine as internal calibrator, we extracted lipids from 50 microL plasma. We deacylated Cer and glycosphingolipids by use of microwave-assisted hydrolysis in methanolic NaOH, followed by derivatization of the liberated amino-group with o-phthaldialdehyde. We separated the derivatized sphingoid bases and lysoglycosphingolipids by HPLC on a C18 reversed-phase column with a methanol/water mobile phase (88:12, vol/vol) and quantified them by use of a fluorescence detector at lambda(ex) 340 nm and lambda(em) 435 nm. RESULTS: Optimal conditions in the Solids/Moisture System SAM-155 microwave oven (CEM Corp.) for the complete deacylation of Cer and neutral glycosphingolipids without decomposition were 60 min at 85% power, fan setting 7. Intra- and interassay CVs were <4% and <14%, respectively, and recovery rates were 87%-113%. The limit of quantification was 2 pmol (0.1 pmol on column), and the method was linear over the interval of 2-200 microL plasma. In samples from 40 healthy individuals, mean (SD) concentrations were 9.0 (2.3) micromol/L for Cer, 6.3 (1.9) micromol/L for GlcCer, and 1.7 (0.5) micromol/L for CTH. Plasma concentrations of GlcCer were higher in Gaucher disease patient samples and of CTH in Fabry disease patient samples. CONCLUSIONS: HPLC enables quantification of total Cer, GlcCer, and CTH in plasma and is useful for the follow-up of patients on therapy for Gaucher or Fabry disease.


Subject(s)
Ceramides/blood , Chromatography, High Pressure Liquid , Fabry Disease/diagnosis , Fabry Disease/therapy , Fluorometry , Gaucher Disease/diagnosis , Gaucher Disease/therapy , Glucosylceramides/blood , Humans , Plasma , Reference Values , Trihexosylceramides/blood
14.
Am J Hum Genet ; 79(5): 807-19, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17033958

ABSTRACT

Mucopolysaccharidosis IIIC (MPS IIIC, or Sanfilippo C syndrome) is a lysosomal storage disorder caused by the inherited deficiency of the lysosomal membrane enzyme acetyl-coenzyme A: alpha -glucosaminide N-acetyltransferase (N-acetyltransferase), which leads to impaired degradation of heparan sulfate. We report the narrowing of the candidate region to a 2.6-cM interval between D8S1051 and D8S1831 and the identification of the transmembrane protein 76 gene (TMEM76), which encodes a 73-kDa protein with predicted multiple transmembrane domains and glycosylation sites, as the gene that causes MPS IIIC when it is mutated. Four nonsense mutations, 3 frameshift mutations due to deletions or a duplication, 6 splice-site mutations, and 14 missense mutations were identified among 30 probands with MPS IIIC. Functional expression of human TMEM76 and the mouse ortholog demonstrates that it is the gene that encodes the lysosomal N-acetyltransferase and suggests that this enzyme belongs to a new structural class of proteins that transport the activated acetyl residues across the cell membrane.


Subject(s)
Acetyltransferases/genetics , Mucopolysaccharidosis III/enzymology , Mucopolysaccharidosis III/genetics , Mutation , Acetyltransferases/chemistry , Acetyltransferases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/genetics , Exons , Female , Gene Expression , Humans , Male , Mice , Molecular Sequence Data , Pedigree , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection
15.
Ann Neurol ; 58(4): 649-52, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16178026

ABSTRACT

Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) is caused by mutations in the gene encoding thymidine phosphorylase (TP). All MNGIE patients have had severe loss of TP function and prominent plasma accumulations of the TP substrates thymidine (dThd) and deoxyuridine (dUrd). Here, we report for the first time to our knowledge three MNGIE patients with later onset, milder phenotype, and less severe TP dysfunction, compared with typical MNGIE patients. This report demonstrates a direct relationship between the biochemical defects and clinical phenotypes in MNGIE and supports the notion that reduction of dThd and dUrd accumulation or TP replacement could be useful therapy for MNGIE.


Subject(s)
Mitochondrial Encephalomyopathies/enzymology , Thymidine Phosphorylase/metabolism , Arginine/metabolism , Chromatography, High Pressure Liquid/methods , Conserved Sequence , Female , Glycine/metabolism , Humans , Leucine/metabolism , Male , Middle Aged , Mitochondrial Encephalomyopathies/genetics , Polymorphism, Restriction Fragment Length , Thymidine Phosphorylase/genetics , Valine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL