Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
2.
Mol Neurobiol ; 56(1): 648-657, 2019 Jan.
Article En | MEDLINE | ID: mdl-29779173

Glutaric acidemia type I (GA-I) is a neurometabolic disease caused by deficient activity of glutaryl-CoA dehydrogenase (GCDH) that results in accumulation of metabolites derived from lysine (Lys), hydroxylysine, and tryptophan catabolism. GA-I patients typically develop encephalopatic crises with striatal degeneration and progressive white matter defects. However, late onset patients as well as Gcdh-/- mice only suffer diffuse myelinopathy, suggesting that neuronal death and white matter defects are different pathophysiological events. To test this hypothesis, striatal myelin was studied in Gcdh-/- mice fed from 30 days of age during up to 60 days with a diet containing normal or moderately increased amounts of Lys (2.8%), which ensure sustained elevated levels of GA-I metabolites. Gcdh-/- mice fed with 2.8% Lys diet showed a significant decrease in striatal-myelinated areas and progressive vacuolation of white matter tracts, as compared with animals fed with normal diet. Myelin pathology increased with the time of exposure to high Lys diet and was also detected in 90-day old Gcdh-/- mice fed with normal diet, suggesting that dietary Lys accelerated the undergoing white matter damage. Gcdh-/- mice fed with 2.8% Lys diet also showed increased GRP78/BiP immunoreactivity in oligodendrocytes and neurons, denoting ER stress. However, the striatal and cortical neuronal density was unchanged with respect to normal diet. Thus, myelin damage seen in Gcdh-/- mice fed with 2.8% Lys seems to be mediated by a long-term increased levels of GA-I metabolites having deleterious effects in myelinating oligodendrocytes over neurons.


Diet , Glutaryl-CoA Dehydrogenase/deficiency , Lysine/adverse effects , White Matter/enzymology , White Matter/injuries , Animals , Cell Count , Cell Death/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Endoplasmic Reticulum Chaperone BiP , Glutaryl-CoA Dehydrogenase/metabolism , Mice , Myelin Sheath/metabolism , Neurons/drug effects , Neurons/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , White Matter/pathology
3.
Mol Genet Metab ; 119(1-2): 50-6, 2016 09.
Article En | MEDLINE | ID: mdl-27397597

Glutaric aciduria type I (GA-I) is an autosomal recessive organic aciduria resulting from a functional deficiency of glutaryl-CoA dehydrogenase, encoded by GCDH. Two clinically indistinguishable diagnostic subgroups of GA-I are known; low and high excretors (LEs and HEs, respectively). Early medical and dietary interventions can result in significantly better outcomes and improved quality of life for patients with GA-I. We report on nine cases of GA-I LE patients all sharing the M405V allele with two cases missed by newborn screening (NBS) using tandem mass spectrometry (MS/MS). We describe a novel case with the known pathogenic M405V variant and a novel V133L variant, and present updated and previously unreported clinical, biochemical, functional and molecular data on eight other patients all sharing the M405V allele. Three of the nine patients are of African American ancestry, with two as siblings. GCDH activity was assayed in six of the nine patients and varied from 4 to 25% of the control mean. We support the use of urine glutarylcarnitine as a biochemical marker of GA-I by demonstrating that glutarylcarnitine is efficiently cleared by the kidney (50-90%) and that plasma and urine glutarylcarnitine follow a linear relationship. We report the allele frequencies for three known GA-I LE GCDH variants (M405V, V400M and R227P) and note that both the M405V and V400M variants are significantly more common in the population of African ancestry compared to the general population. This report highlights the M405V allele as another important molecular marker in patients with the GA-I LE phenotype. Therefore, the incorporation into newborn screening of molecular screening for the M405V and V400M variants in conjunction with MS/MS could help identify asymptomatic at-risk GA-I LE patients that could potentially be missed by current NBS programs.


Amino Acid Metabolism, Inborn Errors/genetics , Biomarkers , Brain Diseases, Metabolic/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Neonatal Screening , Black or African American/genetics , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/physiopathology , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/physiopathology , Female , Gene Frequency , Glutarates/metabolism , Humans , Infant, Newborn , Male , Mutation , Phenotype , Tandem Mass Spectrometry
4.
Hum Mol Genet ; 24(16): 4504-15, 2015 Aug 15.
Article En | MEDLINE | ID: mdl-25968119

Glutaric acidemia type I (GA-I) is an inherited neurometabolic childhood disorder caused by defective activity of glutaryl CoA dehydrogenase (GCDH) which disturb lysine (Lys) and tryptophan catabolism leading to neurotoxic accumulation of glutaric acid (GA) and related metabolites. However, it remains unknown whether GA toxicity is due to direct effects on vulnerable neurons or mediated by GA-intoxicated astrocytes that fail to support neuron function and survival. As damaged astrocytes can also contribute to sustain high GA levels, we explored the ability of Gcdh-/- mouse astrocytes to produce GA and induce neuronal death when challenged with Lys. Upon Lys treatment, Gcdh-/- astrocytes synthetized and released GA and 3-hydroxyglutaric acid (3HGA). Lys and GA treatments also increased oxidative stress and proliferation in Gcdh-/- astrocytes, both prevented by antioxidants. Pretreatment with Lys also caused Gcdh-/- astrocytes to induce extensive death of striatal and cortical neurons when compared with milder effect in WT astrocytes. Antioxidants abrogated the neuronal death induced by astrocytes exposed to Lys or GA. In contrast, Lys or GA direct exposure on Gcdh-/- or WT striatal neurons cultured in the absence of astrocytes was not toxic, indicating that neuronal death is mediated by astrocytes. In summary, GCDH-defective astrocytes actively contribute to produce and accumulate GA and 3HGA when Lys catabolism is stressed. In turn, astrocytic GA production induces a neurotoxic phenotype that kills striatal and cortical neurons by an oxidative stress-dependent mechanism. Targeting astrocytes in GA-I may prompt the development of new antioxidant-based therapeutical approaches.


Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Astrocytes/metabolism , Brain Diseases, Metabolic/genetics , Brain Diseases, Metabolic/metabolism , Corpus Striatum/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Neurons/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Astrocytes/pathology , Brain Diseases, Metabolic/pathology , Cell Survival/genetics , Corpus Striatum/pathology , Disease Models, Animal , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Mice , Mice, Knockout , Neurons/pathology
5.
PLoS One ; 9(3): e90477, 2014.
Article En | MEDLINE | ID: mdl-24594605

We determined mRNA expression of the ionotropic glutamate receptors NMDA (NR1, NR2A and NR2B subunits), AMPA (GluR2 subunit) and kainate (GluR6 subunit), as well as of the glutamate transporters GLAST and GLT1 in cerebral cortex and striatum of wild type (WT) and glutaryl-CoA dehydrogenase deficient (Gchh-/-) mice aged 7, 30 and 60 days. The protein expression levels of some of these membrane proteins were also measured. Overexpression of NR2A and NR2B in striatum and of GluR2 and GluR6 in cerebral cortex was observed in 7-day-old Gcdh-/-. There was also an increase of mRNA expression of all NMDA subunits in cerebral cortex and of NR2A and NR2B in striatum of 30-day-old Gcdh-/- mice. At 60 days of life, all ionotropic receptors were overexpressed in cerebral cortex and striatum of Gcdh-/- mice. Higher expression of GLAST and GLT1 transporters was also verified in cerebral cortex and striatum of Gcdh-/- mice aged 30 and 60 days, whereas at 7 days of life GLAST was overexpressed only in striatum from this mutant mice. Furthermore, high lysine intake induced mRNA overexpression of NR2A, NR2B and GLAST transcripts in striatum, as well as of GluR2 and GluR6 in both striatum and cerebral cortex of Gcdh-/- mice. Finally, we found that the protein expression of NR2A, NR2B, GLT1 and GLAST were significantly greater in cerebral cortex of Gcdh-/- mice, whereas NR2B and GLT1 was similarly enhanced in striatum, implying that these transcripts were translated into their products. These results provide evidence that glutamate receptor and transporter expression is higher in Gcdh-/- mice and that these alterations may be involved in the pathophysiology of GA I and possibly explain, at least in part, the vulnerability of striatum and cerebral cortex to injury in patients affected by GA I.


Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Transport System X-AG/metabolism , Brain Diseases, Metabolic/pathology , Cerebral Cortex/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Neostriatum/metabolism , Receptors, Glutamate/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Transport System X-AG/genetics , Animals , Brain Diseases, Metabolic/enzymology , Cerebral Cortex/pathology , Diet , Female , Gene Expression Regulation , Glutaryl-CoA Dehydrogenase/metabolism , Lysine/metabolism , Male , Mice , Neostriatum/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glutamate/genetics
6.
Mol Genet Metab ; 108(1): 30-9, 2013 Jan.
Article En | MEDLINE | ID: mdl-23218171

Deficiency of glutaryl-CoA dehydrogenase (GCDH) activity or glutaric aciduria type I (GA I) is an inherited neurometabolic disorder biochemically characterized by predominant accumulation of glutaric acid and 3-hydroxyglutaric acid in the brain and other tissues. Affected patients usually present acute striatum necrosis during encephalopathic crises triggered by metabolic stress situations, as well as chronic leukodystrophy and delayed myelination. Considering that the mechanisms underlying the brain injury in this disease are not yet fully established, in the present study we investigated important parameters of oxidative stress in the brain (cerebral cortex, striatum and hippocampus), liver and heart of 30-day-old GCDH deficient knockout (Gcdh(-/-)) and wild type (WT) mice submitted to a normal lysine (Lys) (0.9% Lys), or high Lys diets (2.8% or 4.7% Lys) for 60 h. It was observed that the dietary supplementation of 2.8% and 4.7% Lys elicited noticeable oxidative stress, as verified by an increase of malondialdehyde concentrations (lipid oxidative damage) and 2-7-dihydrodichlorofluorescein (DCFH) oxidation (free radical production), as well as a decrease of reduced glutathione levels and alteration of various antioxidant enzyme activities (antioxidant defenses) in the cerebral cortex and the striatum, but not in the hippocampus, the liver and the heart of Gcdh(-/-) mice, as compared to WT mice receiving the same diets. Furthermore, alterations of oxidative stress parameters in the cerebral cortex and striatum were more accentuated in symptomatic, as compared to asymptomatic Gcdh(-/-) mice exposed to 4.7% Lys overload. Histopathological studies performed in the cerebral cortex and striatum of these animals exposed to high dietary Lys revealed increased expression of oxidative stress markers despite the absence of significant structural damage. The results indicate that a disruption of redox homeostasis in the cerebral cortex and striatum of young Gcdh(-/-) mice exposed to increased Lys diet may possibly represent an important pathomechanism of brain injury in GA I patients under metabolic stress.


Brain/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Homeostasis , Lysine/administration & dosage , Animals , Dietary Supplements , Glutaryl-CoA Dehydrogenase/genetics , Mice , Mice, Knockout , Oxidation-Reduction , Oxidative Stress , Thiobarbituric Acid Reactive Substances/metabolism
7.
J Inherit Metab Dis ; 34(3): 677-94, 2011 Jun.
Article En | MEDLINE | ID: mdl-21431622

Glutaric aciduria type I (synonym, glutaric acidemia type I) is a rare organic aciduria. Untreated patients characteristically develop dystonia during infancy resulting in a high morbidity and mortality. The neuropathological correlate is striatal injury which results from encephalopathic crises precipitated by infectious diseases, immunizations and surgery during a finite period of brain development, or develops insidiously without clinically apparent crises. Glutaric aciduria type I is caused by inherited deficiency of glutaryl-CoA dehydrogenase which is involved in the catabolic pathways of L-lysine, L-hydroxylysine and L-tryptophan. This defect gives rise to elevated glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutarylcarnitine which can be detected by gas chromatography/mass spectrometry (organic acids) or tandem mass spectrometry (acylcarnitines). Glutaric aciduria type I is included in the panel of diseases that are identified by expanded newborn screening in some countries. It has been shown that in the majority of neonatally diagnosed patients striatal injury can be prevented by combined metabolic treatment. Metabolic treatment that includes a low lysine diet, carnitine supplementation and intensified emergency treatment during acute episodes of intercurrent illness should be introduced and monitored by an experienced interdisciplinary team. However, initiation of treatment after the onset of symptoms is generally not effective in preventing permanent damage. Secondary dystonia is often difficult to treat, and the efficacy of available drugs cannot be predicted precisely in individual patients. The major aim of this revision is to re-evaluate the previous diagnostic and therapeutic recommendations for patients with this disease and incorporate new research findings into the guideline.


Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/therapy , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/therapy , Practice Guidelines as Topic , Algorithms , Amino Acid Metabolism, Inborn Errors/complications , Brain Diseases, Metabolic/complications , Emergency Medical Services/methods , Glutaryl-CoA Dehydrogenase/deficiency , Humans , Infant, Newborn , Mass Screening/methods , Monitoring, Physiologic/methods , Neonatal Screening/methods , Nervous System Diseases/etiology , Nervous System Diseases/therapy
8.
J Bioenerg Biomembr ; 43(1): 31-8, 2011 Feb.
Article En | MEDLINE | ID: mdl-21249436

Organic acidurias or organic acidemias constitute a group of inherited disorders caused by deficient activity of specific enzymes of amino acids, carbohydrates or lipids catabolism, leading to large accumulation and excretion of one or more carboxylic (organic) acids. Affected patients usually present neurologic symptoms and abnormalities, sometimes accompanied by cardiac and skeletal muscle alterations, whose pathogenesis is poorly known. However, in recent years growing evidence has emerged indicating that mitochondrial dysfunction is directly or indirectly involved in the pathology of various organic acidemias. Mitochondrial impairment in some of these diseases are generally due to mutations in nuclear genes of the tricarboxylic acid cycle or oxidative phosphorylation, while in others it seems to result from toxic influences of the endogenous organic acids to the mitochondrion. In this minireview, we will briefly summarize the present knowledge obtained from human and animal studies showing that disruption of mitochondrial homeostasis may represent a relevant pathomechanism of tissue damage in selective organic acidemias. The discussion will focus on mitochondrial alterations found in patients affected by organic acidemias and by the deleterious effects of the accumulating organic acids on mitochondrial pathways that are crucial for ATP formation and transfer. The elucidation of the mechanisms of toxicity of these acidic compounds offers new perspectives for potential novel adjuvant therapeutic strategies in selected disorders of this group.


Adenosine Triphosphate/metabolism , Carboxylic Acids/metabolism , Homeostasis/physiology , Metabolism, Inborn Errors/physiopathology , Mitochondria/physiology , Mitochondrial Diseases/physiopathology , Acetyl-CoA C-Acyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Barth Syndrome/physiopathology , Brain Diseases, Metabolic/physiopathology , Brain Diseases, Metabolic, Inborn/physiopathology , Glutaryl-CoA Dehydrogenase/deficiency , Humans , Mitochondria/metabolism , Propionic Acidemia/physiopathology , Purpura/physiopathology
9.
J Inherit Metab Dis ; 34(1): 173-80, 2011 Feb.
Article En | MEDLINE | ID: mdl-20978942

Accumulation of glutaric acid (GA) and 3-hydroxyglutaric acid (3HGA) in body fluids is the biochemical hallmark of type 1 glutaric aciduria (GA1), a disorder characterized by acute striatal degeneration and a subsequent dystonia. To date, methods for quantification of 3HGA are mainly based on stable isotope dilution gas chromatography mass spectrometry (GC-MS) and require extensive sample preparation. Here we describe a simple liquid chromatography tandem MS (LC-MS/MS) method to quantify this important metabolite in dried urine spots (DUS). This method is based on derivatization with 4-[2-(N,N-dimethylamino)ethylaminosulfonyl]-7-(2-aminoethylamino)-2,1,3-benzoxadiazole (DAABD-AE). Derivatization was adopted to improve the chromatographic and mass spectrometric properties of the studied analytes. Derivatization was performed directly on a 3.2-mm disc of DUS as a sample without extraction. Sample mixture was heated at 60°C for 45 min, and 5 µl of the reaction solution was analyzed by LC-MS/MS. Reference ranges obtained were in excellent agreement with the literature. The method was applied retrospectively for the analysis of DUS samples from established low- and high-excreter GA1 patients as well as controls (n = 100). Comparison of results obtained versus those obtained by GC-MS was satisfactory (n = 14). In populations with a high risk of GA1, this approach will be useful as a primary screening method for high- or low-excreter variants. In these populations, however, DUS analysis should not be implemented before completing a parallel comparative study with the standard screening method (i.e., molecular testing). In addition, follow-up DUS GA and 3HGA testing of babies with elevated dried blood spot C5DC acylcarnitines will be useful as a first-tier diagnostic test, thus reducing the number of cases requiring enzymatic and molecular analyses to establish or refute the diagnosis of GA1.


Glutarates/urine , Tandem Mass Spectrometry/methods , Urinalysis/methods , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/urine , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/urine , Chromatography, Liquid/methods , Desiccation , Glutarates/analysis , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/urine , Humans , Infant, Newborn , Neonatal Screening/methods
10.
Biochim Biophys Acta ; 1782(6): 385-90, 2008 Jun.
Article En | MEDLINE | ID: mdl-18348873

Glutaric aciduria type 1 (GA1) is caused by the deficiency of glutaryl-CoA dehydrogenase (GCDH). Affected patients are prone to the development of encephalopathic crises during an early time window with destruction of striatal neurons and a subsequent irreversible movement disorder. 3-Hydroxyglutaric acid (3OHGA) accumulates in tissues and body fluids of GA1 patients and has been shown to mediate toxic effects on neuronal as well as endothelial cells. Injection of (3H)-labeled into 6 week-old Gcdh(-/-) mice, a model of GA1, revealed a low recovery in kidney, liver, or brain tissue that did not differ from control mice. Significant amounts of 3OHGA were found to be excreted via the intestinal tract. Exposure of Gcdh(-/-) mice to a high protein diet led to an encephalopathic crisis, vacuolization in the brain, and death after 4-5 days. Under these conditions, high amounts of injected 3H-3OHGA were found in kidneys of Gcdh(-/-) mice, whereas the radioactivity recovered in brain and blood was reduced. The data demonstrate that under conditions mimicking encephalopathic crises the blood-brain barrier appears to remain intact.


Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases/metabolism , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/complications , Animals , Biological Transport , Brain/metabolism , Brain/pathology , Brain Diseases/etiology , Dextrans/metabolism , Disease Models, Animal , Glutaryl-CoA Dehydrogenase/genetics , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Mol Genet Metab ; 93(4): 363-70, 2008 Apr.
Article En | MEDLINE | ID: mdl-18155630

3-MCC deficiency is among the most common inborn errors of metabolism identified on expanded newborn screening (1:36,000 births). However, evidence-based guidelines for diagnosis and management of this disorder are lacking. Using the traditional Delphi method, a panel of 15 experts in inborn errors of metabolism was convened to develop consensus-based clinical practice guidelines for the diagnosis and management of 3-MCC screen-positive infants and their mothers. The Oxford Centre for Evidence-based Medicine system was used to grade the literature review and create recommendations graded from A (evidence level of randomized clinical trials) to D (expert opinion). Panelists reviewed the initial evaluation of the screen-positive infant-mother dyad, diagnostic guidelines, and management of diagnosed patients. Grade D consensus recommendations were made in each of these three areas. The panel did not reach consensus on all issues. This consensus protocol is intended to assist clinicians in the diagnosis and management of screen-positive newborns for 3-MCC deficiency and to encourage the development of evidence-based guidelines.


Carbon-Carbon Ligases/deficiency , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/therapy , Carnitine/blood , Carnitine/therapeutic use , Delphi Technique , Energy Intake , Humans , Infant, Newborn , Leucine/administration & dosage , Mothers , Neonatal Screening
12.
J Clin Invest ; 117(11): 3258-70, 2007 Nov.
Article En | MEDLINE | ID: mdl-17932566

Glutaric acidemia type I (GA-I) is an inherited disorder of lysine and tryptophan metabolism presenting with striatal lesions anatomically and symptomatically similar to Huntington disease. Affected children commonly suffer acute brain injury in the context of a catabolic state associated with nonspecific illness. The mechanisms underlying injury and age-dependent susceptibility have been unknown, and lack of a diagnostic marker heralding brain injury has impeded intervention efforts. Using a mouse model of GA-I, we show that pathologic events began in the neuronal compartment while enhanced lysine accumulation in the immature brain allowed increased glutaric acid production resulting in age-dependent injury. Glutamate and GABA depletion correlated with brain glutaric acid accumulation and could be monitored in vivo by proton nuclear magnetic resonance (1H NMR) spectroscopy as a diagnostic marker. Blocking brain lysine uptake reduced glutaric acid levels and brain injury. These findings provide what we believe are new monitoring and treatment strategies that may translate for use in human GA-I.


Aging/physiology , Amino Acid Metabolism, Inborn Errors , Brain Diseases, Metabolic, Inborn , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Amino Acid Metabolism, Inborn Errors/diet therapy , Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain Diseases, Metabolic, Inborn/diet therapy , Brain Diseases, Metabolic, Inborn/pathology , Brain Diseases, Metabolic, Inborn/physiopathology , Child , Diet , Disease Models, Animal , Genetic Predisposition to Disease , Glucose/metabolism , Glucose/therapeutic use , Glutamic Acid/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Homoarginine/metabolism , Homoarginine/therapeutic use , Humans , Lysine/metabolism , Lysine/therapeutic use , Mice , Mice, Knockout , Mitochondria/metabolism , Neurons/metabolism , Neurons/pathology , Neurons/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Tryptophan/metabolism , gamma-Aminobutyric Acid/metabolism
13.
J Mol Med (Berl) ; 85(7): 763-70, 2007 Jul.
Article En | MEDLINE | ID: mdl-17356845

Patients with glutaryl-CoA dehydrogenase (GCDH) deficiency accumulate glutaric acid (GA) and 3-hydroxyglutaric acid (3OH-GA) in their blood and urine. To identify the transporter mediating the translocation of 3OH-GA through membranes, kidney tissue of Gcdh-/- mice have been investigated because of its central role in urinary excretion of this metabolite. Using microarray analyses of kidney-expressed genes in Gcdh-/- mice, several differentially expressed genes encoding transporter proteins were identified. Real-time polymerase chain reaction analysis confirmed the upregulation of the sodium-dependent dicarboxylate cotransporter 3 (NaDC3) and the organic cation transporter 2 (OCT2). Expression analysis of NaDC3 in Xenopus laevis oocytes by the two-electrode-voltage-clamp technique demonstrated the sodium-dependent translocation of 3OH-GA with a K (M) value of 0.95 mM. Furthermore, tracer flux measurements in Chinese hamster ovary cells overexpressing OCT2 showed that 3OH-GA inhibited significantly the uptake of methyl-4-phenylpyridinium, whereas 3OH-GA is not transported by OCT2. The data demonstrate for the first time the membrane translocation of 3OH-GA mediated by NaDC3 and the cis-inhibitory effect on OCT2-mediated transport of cations.


Dicarboxylic Acid Transporters/metabolism , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Kidney/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Cation Transport Proteins/metabolism , Symporters/metabolism , Animals , Biological Transport , Cricetinae , Cricetulus , Dicarboxylic Acid Transporters/genetics , Electrophysiology , Female , Gene Expression Profiling , Kidney/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Oocytes , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Cation Transporter 2 , Ovary , Symporters/genetics , Xenopus laevis
15.
Mol Genet Metab ; 89(1-2): 87-96, 2006.
Article En | MEDLINE | ID: mdl-16753325

Knockout mouse models have been created to study the consequences of deficiencies in arginase AI and AII, both individually and combined. The AI knockout animals die by 14 days of age from hyperammonemia, while the AII knockout has no obvious phenotype. The double knockout (AI(-/-)/AII(-/-)) exhibits the phenotype of the AI-deficient mice, with the additional absence of AII not exacerbating the observed phenotype of the AI knockout animals. Plasma amino acid measurements in the double knockout have shown arginine levels increased roughly 100-fold and ornithine decreased roughly 10-fold as compared to wildtype. Liver ornithine levels were reduced to 2% of normal in the double knockout with arginine very highly elevated. Arginine and ornithine were also altered in other tissues in the double knockout mice, such as kidney, brain, and small intestine. This is the first demonstration that the fatal hyperammonemia in the AI knockout mouse is almost certainly due to ornithine deficiency, the amino acid needed to drive the urea cycle. Others have shown that the expression of ornithine aminotransferase (OAT) rapidly decreases in the intestine at the same age when the AI-deficient animals die, indicating that this enzyme is critical to the maintenance of ornithine homeostasis, at least at this early stage of mouse development. Although most human AI-deficient patients have no symptomatic hyperammonemia at birth, it is possible that clinically significant ornithine deficiency is already present.


Arginase/genetics , Hyperammonemia/genetics , Ornithine/deficiency , Animals , Arginase/analysis , Arginine/analysis , Arginine/blood , Argininosuccinic Acid/analysis , Brain Chemistry , Down-Regulation , Intestine, Small/chemistry , Intestine, Small/enzymology , Kidney/chemistry , Liver/chemistry , Mice , Mice, Knockout , Ornithine/analysis , Ornithine/blood , Ornithine-Oxo-Acid Transaminase/analysis
16.
J Neurochem ; 97(3): 899-910, 2006 May.
Article En | MEDLINE | ID: mdl-16573641

Glutaric acid (GA) and 3-hydroxyglutaric acids (3-OH-GA) are key metabolites in glutaryl co-enzyme A dehydrogenase (GCDH) deficiency and are both considered to be potential neurotoxins. As cerebral concentrations of GA and 3-OH-GA have not yet been studied systematically, we investigated the tissue-specific distribution of these organic acids and glutarylcarnitine in brain, liver, skeletal and heart muscle of Gcdh-deficient mice as well as in hepatic Gcdh-/- mice and in C57Bl/6 mice following intraperitoneal loading. Furthermore, we determined the flux of GA and 3-OH-GA across the blood-brain barrier (BBB) using porcine brain microvessel endothelial cells. Concentrations of GA, 3-OH-GA and glutarylcarnitine were significantly elevated in all tissues of Gcdh-/- mice. Strikingly, cerebral concentrations of GA and 3-OH-GA were unexpectedly high, reaching similar concentrations as those found in liver. In contrast, cerebral concentrations of these organic acids remained low in hepatic Gcdh-/- mice and after intraperitoneal injection of GA and 3-OH-GA. These results suggest limited flux of GA and 3-OH-GA across the BBB, which was supported in cultured porcine brain capillary endothelial cells. In conclusion, we propose that an intracerebral de novo synthesis and subsequent trapping of GA and 3-OH-GA should be considered as a biochemical risk factor for neurodegeneration in GCDH deficiency.


Blood-Brain Barrier/physiopathology , Brain/metabolism , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Neurodegenerative Diseases/physiopathology , Amino Acids/metabolism , Animals , Biological Transport/physiology , Blotting, Western/methods , Brain/cytology , Carnitine/analogs & derivatives , Carnitine/metabolism , Cells, Cultured , Dicarboxylic Acids/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Glucose/metabolism , Heart/physiology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscles/metabolism , Neurodegenerative Diseases/genetics , Risk Factors , Statistics, Nonparametric , Swine , Time Factors , Tissue Distribution/physiology
17.
Mol Genet Metab ; 88(1): 16-21, 2006 May.
Article En | MEDLINE | ID: mdl-16448836

BACKGROUND: Tyrosinemia type I (TYR 1) is a severe disorder causing early death if left untreated. While tyrosine can be determined in dried blood spots (DBS), it is not a specific marker for TYR 1 and most often associated with benign transient tyrosinemia of the newborn. Succinylacetone (SUAC) is a specific marker for TYR 1 but not detectable by routine newborn screening. We developed a new assay that determines SUAC in DBS by liquid-chromatography tandem mass spectrometry (LC-MS/MS). METHODS: Whole blood is eluted from a 3/16-in. DBS by an aqueous solution containing deuterium labeled SUAC as internal standard (IS). SUAC and IS are oximated, then extracted, butylated, and analyzed by LC-MS/MS. Quantitation is from SUAC spiked calibrator DBS over the range 0-200 microM using selected reaction monitoring of transitions m/z 212 to 156 and m/z 214 to 140 for SUAC and IS, respectively. Analysis time is 5 min. To assess the effectiveness of a two-tier screening approach for TYR 1 we applied this assay to our newborn screening program over the last 15 months. RESULTS: The intra-assay precision was determined for three different levels of SUAC (5, 20, and 50 micromol/L) and the CV calculated to be 4.7, 2.6, and 3.1%, respectively (n=5). Inter-assay precision CVs were 12.7, 8.2, and 7.8%, respectively on the same samples. SUAC levels in DBS from 10 confirmed TYR 1 cases not treated with 2-(2-nitro-4-trifluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC) were clearly abnormal (16-150 micromol/L; mean: 61 micromol/L; controls: <5 micromol/L). Over a 15-month period, SUAC was determined in newborn screening samples with elevated tyrosine concentrations when applying different cut off values until it was settled at 150 micromol/L. No case of TYR 1 was detected in 124,780 newborns tested. CONCLUSION: We have developed a new LC-MS/MS based method for the determination of SUAC in DBS. This assay has the potential to significantly reduce the number of false positive results in newborn screening for TYR 1 and can also be used for the laboratory follow up of patients treated for TYR 1.


Heptanoates/blood , Neonatal Screening/methods , Tyrosinemias/diagnosis , Algorithms , Chromatography, Liquid/methods , Humans , Infant, Newborn , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Electrospray Ionization/methods , Tyrosinemias/epidemiology , United States/epidemiology
18.
Brain ; 129(Pt 4): 899-910, 2006 Apr.
Article En | MEDLINE | ID: mdl-16446282

In the autosomal recessive human disease, glutaric aciduria type I (GA-1), glutaryl-CoA dehydrogenase (GCDH) deficiency disrupts the mitochondrial catabolism of lysine and tryptophan. Affected individuals accumulate glutaric acid (GA) and 3-hydroxyglutaric acid (3-OHGA) in the serum and often suffer acute striatal injury in childhood. Prior attempts to produce selective striatal vulnerability in an animal model have been unsuccessful. We hypothesized that acute striatal injury may be induced in GCDH-deficient (Gcdh-/-) mice by elevated dietary protein and lysine. Here, we show that high protein diets are lethal to 4-week-old and 8-week-old Gcdh-/- mice within 2-3 days and 7-8 days, respectively. High lysine alone resulted in vasogenic oedema and blood-brain barrier breakdown within the striatum, associated with serum and tissue GA accumulation, neuronal loss, haemorrhage, paralysis, seizures and death in 75% of 4-week-old Gcdh-/- mice after 3-12 days. In contrast, most 8-week-old Gcdh-/- mice survived on high lysine, but developed white matter lesions, reactive astrocytes and neuronal loss after 6 weeks. Thus, the Gcdh-/- mouse exposed to high protein or lysine may be a useful model of human GA-1 including developmentally dependent striatal vulnerability.


Amino Acid Metabolism, Inborn Errors/etiology , Diet/adverse effects , Dietary Proteins/toxicity , Disease Models, Animal , Glutarates/urine , Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Blood-Brain Barrier , Capillary Permeability/drug effects , Corpus Striatum/blood supply , Dietary Proteins/administration & dosage , Female , Glutarates/pharmacology , Lysine/toxicity , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Survival Analysis , Tissue Culture Techniques
19.
Curr Protoc Hum Genet ; Chapter 17: Unit 17.2, 2006 Nov.
Article En | MEDLINE | ID: mdl-18428392

This unit describes methods for the preparation of samples for analysis of physiological amino acids and organic acids. Amino acids are analyzed by ion-exchange chromatography using an automated system. Organic acids are analyzed by gas-chromatography/mass spectrometry (GC-MS). Analysis of amino and organic acids is necessary to detect and monitor the treatment of many inborn errors of metabolism.


Amino Acids/analysis , Body Fluids/chemistry , Carboxylic Acids/analysis , Gas Chromatography-Mass Spectrometry/methods , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , Amino Acids/chemistry , Amino Acids/metabolism , Body Fluids/physiology , Carboxylic Acids/chemistry , Carboxylic Acids/metabolism , Chromatography, High Pressure Liquid/methods , Chromatography, Thin Layer/methods , Humans , Metabolism, Inborn Errors/metabolism
20.
Mol Genet Metab ; 86(3): 417-20, 2005 Nov.
Article En | MEDLINE | ID: mdl-16183314

Glutaryl-CoA dehydrogenase deficiency (GA-I) is associated with the onset of irreversible, disabling dystonia between 3 and 18 months of age. Presymptomatic identification and treatment can prevent the devastating disability associated with this disorder. We report the retrospective analysis of the newborn blood spot of an affected child with a low excretor phenotype. The level of glutarylcarnitine was below the newborn screening program cut-off. This suggests that some cases of GA-I may be missed by newborn screening by tandem mass spectrometry.


Glutarates/urine , Glutaryl-CoA Dehydrogenase/deficiency , Neonatal Screening , Carnitine/analogs & derivatives , Carnitine/blood , Cells, Cultured , Fibroblasts/metabolism , Glutaryl-CoA Dehydrogenase/blood , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Infant, Newborn , Mass Spectrometry , Retrospective Studies
...