Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 46(3): 482-519, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36221165

RESUMEN

Glutaric aciduria type 1 is a rare inherited neurometabolic disorder of lysine metabolism caused by pathogenic gene variations in GCDH (cytogenic location: 19p13.13), resulting in deficiency of mitochondrial glutaryl-CoA dehydrogenase (GCDH) and, consequently, accumulation of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid and glutarylcarnitine detectable by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Depending on residual GCDH activity, biochemical high and low excreting phenotypes have been defined. Most untreated individuals present with acute onset of striatal damage before age 3 (to 6) years, precipitated by infectious diseases, fever or surgery, resulting in irreversible, mostly dystonic movement disorder with limited life expectancy. In some patients, striatal damage develops insidiously. In recent years, the clinical phenotype has been extended by the finding of extrastriatal abnormalities and cognitive dysfunction, preferably in the high excreter group, as well as chronic kidney failure. Newborn screening is the prerequisite for pre-symptomatic start of metabolic treatment with low lysine diet, carnitine supplementation and intensified emergency treatment during catabolic episodes, which, in combination, have substantially improved neurologic outcome. In contrast, start of treatment after onset of symptoms cannot reverse existing motor dysfunction caused by striatal damage. Dietary treatment can be relaxed after the vulnerable period for striatal damage, that is, age 6 years. However, impact of dietary relaxation on long-term outcomes is still unclear. This third revision of evidence-based recommendations aims to re-evaluate previous recommendations (Boy et al., J Inherit Metab Dis, 2017;40(1):75-101; Kolker et al., J Inherit Metab Dis 2011;34(3):677-694; Kolker et al., J Inherit Metab Dis, 2007;30(1):5-22) and to implement new research findings on the evolving phenotypic diversity as well as the impact of non-interventional variables and treatment quality on clinical outcomes.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Encefalopatías Metabólicas , Humanos , Glutaril-CoA Deshidrogenasa , Lisina/metabolismo , Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/genética , Encefalopatías Metabólicas/terapia , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Glutaratos/metabolismo
2.
Arch Biochem Biophys ; 709: 108970, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34181873

RESUMEN

Glutaric acidemia type 1 (GA1) is caused by glutaryl-CoA dehydrogenase deficiency that leads to a blockage in the metabolic route of the amino acids lysine and tryptophan and subsequent accumulation of glutaric acid (GA), 3-hydroxyglutaric acids and glutarylcarnitine (C5DC). Patients predominantly manifest neurological symptoms, associated with acute striatal degeneration, as well as progressive cortical and striatum injury whose pathogenesis is not yet fully established. Current treatment includes protein/lysine restriction and l-carnitine supplementation of (L-car). The aim of this work was to evaluate behavior parameters and pro-inflammatory factors (cytokines IL-1ß, TNF-α and cathepsin-D levels), as well as the anti-inflammatory cytokine IL10 in striatum of knockout mice (Gcdh-/-) and wild type (WT) mice submitted to a normal or a high Lys diet. The potential protective effects of L-car treatment on these parameters were also evaluated. Gcdh-/- mice showed behavioral changes, including lower motor activity (decreased number of crossings) and exploratory activity (reduced number of rearings). Also, Gcdh-/- mice had significantly higher concentrations of glutarylcarnitine (C5DC) in blood and cathepsin-D (CATD), interleukin IL-1ß and tumor factor necrosis alpha (TNF-α) in striatum than WT mice. Noteworthy, L-car treatment prevented most behavioral alterations, normalized CATD levels and attenuated IL-1ß levels in striatum of Gcdh-/- mice. Finally, IL-1ß was positively correlated with CATD and C5DC levels and L-car was negatively correlated with CATD. Our results demonstrate behavioral changes and a pro-inflammatory status in striatum of the animal model of GA1 and, most importantly, L-car showed important protective effects on these alterations.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Encefalopatías Metabólicas/tratamiento farmacológico , Carnitina/uso terapéutico , Glutaril-CoA Deshidrogenasa/deficiencia , Inflamación/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Animales , Encefalopatías Metabólicas/genética , Carnitina/análogos & derivados , Carnitina/metabolismo , Catepsina D/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Glutaril-CoA Deshidrogenasa/genética , Aseo Animal/efectos de los fármacos , Inflamación/genética , Interleucina-1beta/metabolismo , Locomoción/efectos de los fármacos , Lisina/farmacología , Ratones Noqueados , Prueba de Campo Abierto/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
3.
Mol Genet Metab ; 131(3): 325-340, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33069577

RESUMEN

Glutaric acidemia type 1 (GA1) is a disorder of cerebral organic acid metabolism resulting from biallelic mutations of GCDH. Without treatment, GA1 causes striatal degeneration in >80% of affected children before two years of age. We analyzed clinical, biochemical, and developmental outcomes for 168 genotypically diverse GA1 patients managed at a single center over 31 years, here separated into three treatment cohorts: children in Cohort I (n = 60; DOB 2006-2019) were identified by newborn screening (NBS) and treated prospectively using a standardized protocol that included a lysine-free, arginine-enriched metabolic formula, enteral l-carnitine (100 mg/kg•day), and emergency intravenous (IV) infusions of dextrose, saline, and l-carnitine during illnesses; children in Cohort II (n = 57; DOB 1989-2018) were identified by NBS and treated with natural protein restriction (1.0-1.3 g/kg•day) and emergency IV infusions; children in Cohort III (n = 51; DOB 1973-2016) did not receive NBS or special diet. The incidence of striatal degeneration in Cohorts I, II, and III was 7%, 47%, and 90%, respectively (p < .0001). No neurologic injuries occurred after 19 months of age. Among uninjured children followed prospectively from birth (Cohort I), measures of growth, nutritional sufficiency, motor development, and cognitive function were normal. Adherence to metabolic formula and l-carnitine supplementation in Cohort I declined to 12% and 32%, respectively, by age 7 years. Cessation of strict dietary therapy altered plasma amino acid and carnitine concentrations but resulted in no serious adverse outcomes. In conclusion, neonatal diagnosis of GA1 coupled to management with lysine-free, arginine-enriched metabolic formula and emergency IV infusions during the first two years of life is safe and effective, preventing more than 90% of striatal injuries while supporting normal growth and psychomotor development. The need for dietary interventions and emergency IV therapies beyond early childhood is uncertain.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Encefalopatías Metabólicas/genética , Encéfalo/metabolismo , Cuerpo Estriado/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Errores Innatos del Metabolismo de los Aminoácidos/epidemiología , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encéfalo/patología , Encefalopatías Metabólicas/dietoterapia , Encefalopatías Metabólicas/epidemiología , Encefalopatías Metabólicas/metabolismo , Carnitina/metabolismo , Niño , Preescolar , Cuerpo Estriado/patología , Dieta , Femenino , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Lactante , Recién Nacido , Lisina/metabolismo , Masculino
4.
J Nutr ; 150(Suppl 1): 2556S-2560S, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33000154

RESUMEN

Lysine is an essential amino acid, and inherited diseases of its metabolism therefore represent defects of lysine catabolism. Although some of these enzyme defects are not well described yet, glutaric aciduria type I (GA1) and antiquitin (2-aminoadipic-6-semialdehyde dehydrogenase) deficiency represent the most well-characterized diseases. GA1 is an autosomal recessive disorder due to a deficiency of glutaryl-CoA dehydrogenase. Untreated patients exhibit early onset macrocephaly and may present a neurological deterioration with regression and movement disorder at the time of a presumably "benign" infection most often during the first year of life. This is associated with a characteristic neuroimaging pattern with frontotemporal atrophy and striatal injuries. Diagnosis relies on the identification of glutaric and 3-hydroxyglutaric acid in urine along with plasma glutarylcarnitine. Treatment consists of a low-lysine diet aiming at reducing the putatively neurotoxic glutaric and 3-hydroxyglutaric acids. Additional therapeutic measures include administration of l-carnitine associated with emergency measures at the time of intercurrent illnesses aiming at preventing brain injury. Early treated (ideally through newborn screening) patients exhibit a favorable long-term neurocognitive outcome, whereas late-treated or untreated patients may present severe neurocognitive irreversible disabilities. Antiquitin deficiency is the most common form of pyridoxine-dependent epilepsy. α-Aminoadipic acid semialdehyde (AASA) and Δ-1-piperideine-6-carboxylate (P6C) accumulate proximal to the enzymatic block. P6C forms a complex with pyridoxal phosphate (PLP), a key vitamer of pyridoxine, thereby reducing PLP bioavailability and subsequently causing epilepsy. Urinary AASA is a biomarker of antiquitin deficiency. Despite seizure control, only 25% of the pyridoxine-treated patients show normal neurodevelopment. Low-lysine diet and arginine supplementation are proposed in some patients with decrease of AASA, but the impact on neurodevelopment is unclear. In summary, GA1 and antiquitin deficiency are the 2 main human defects of lysine catabolism. Both include neurological impairment. Lysine dietary restriction is a key therapy for GA1, whereas its benefits in antiquitin deficiency appear less clear.


Asunto(s)
Aldehído Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas Innatas/metabolismo , Encefalopatías Metabólicas/metabolismo , Encéfalo/metabolismo , Epilepsia/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Lisina/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/metabolismo , Aldehído Deshidrogenasa/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Arginina/uso terapéutico , Encéfalo/patología , Encefalopatías Metabólicas/terapia , Encefalopatías Metabólicas Innatas/terapia , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina/uso terapéutico , Epilepsia/terapia , Glutaratos/metabolismo , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/terapia , Fosfato de Piridoxal/metabolismo , Piridoxina/metabolismo , Piridoxina/uso terapéutico
5.
Nutrients ; 12(10)2020 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-33081139

RESUMEN

Glutaric aciduria type 1 (GA-1) is a cerebral organic aciduria characterized by striatal injury and progressive movement disorder. Nutrition management shifted from a general restriction of intact protein to targeted restriction of lysine and tryptophan. Recent guidelines advocate for a low-lysine diet using lysine-free, tryptophan-reduced medical foods. GA-1 guideline recommendations for dietary management of patients over the age of six are unclear, ranging from avoiding excessive intake of intact protein to counting milligrams of lysine intake. A 22-question survey on the nutrition management of GA-1 was developed with the goal of understanding approaches to diet management for patients identified by newborn screening under age six years compared to management after diet liberalization, as well as to gain insight into how clinicians define diet liberalization. Seventy-six responses (25% of possible responses) to the survey were received. Nutrition management with GA-1 is divergent among surveyed clinicians. There was congruency among survey responses to the guidelines, but there is still uncertainty about how to counsel patients on diet optimization and when diet liberalization should occur. Ongoing clinical research and better understanding of the natural history of this disease will help establish stronger recommendations from which clinicians can best counsel families.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Encefalopatías Metabólicas/dietoterapia , Fenómenos Fisiológicos Nutricionales Infantiles/fisiología , Dietoterapia/métodos , Proteínas en la Dieta/administración & dosificación , Glutaril-CoA Deshidrogenasa/deficiencia , Fenómenos Fisiológicos Nutricionales del Lactante/fisiología , Lisina/efectos adversos , Triptófano/efectos adversos , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Guías de Práctica Clínica como Asunto , Ingesta Diaria Recomendada , Encuestas y Cuestionarios
6.
Int J Mol Sci ; 21(19)2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32992790

RESUMEN

Riboflavin is the biological precursor of two important flavin cofactors-flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN)-that are critical prosthetic groups in several redox enzymes. While dietary supplementation with riboflavin is a recognized support therapy in several inborn errors of metabolism, it has yet unproven benefits in several other pathologies affecting flavoproteins. This is the case for glutaric aciduria type I (GA-I), a rare neurometabolic disorder associated with mutations in the GCDH gene, which encodes for glutaryl-coenzyme A (CoA) dehydrogenase (GCDH). Although there are a few reported clinical cases that have responded to riboflavin intake, there is still not enough molecular evidence supporting therapeutic recommendation. Hence, it is necessary to elucidate the molecular basis in favor of riboflavin supplementation in GA-I patients. Here, using a combination of biochemical and biophysical methodologies, we investigate the clinical variant GCDH-p.Val400Met as a model for a phenotype associated with severe deflavinylation. Through a systematic analysis, we establish that recombinant human GCDH-p.Val400Met is expressed in a nonfunctional apo form, which is mainly monomeric rather than tetrameric. However, we show that exogenous FAD is a driver for structural reorganization of the mutant enzyme with concomitant functional recovery, improved thermolability, and resistance to trypsin digestion. Overall, these results establish proof of principle for the beneficial effects of riboflavin supplementation in GA-I patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Encefalopatías Metabólicas , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Riboflavina , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Glutaril-CoA Deshidrogenasa/química , Glutaril-CoA Deshidrogenasa/efectos de los fármacos , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Mutación , Pliegue de Proteína/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Proteínas Recombinantes , Riboflavina/farmacología
7.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2420-2427, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31181292

RESUMEN

The deficiency of the enzyme glutaryl-CoA dehydrogenase leads to predominant accumulation of glutaric acid (GA) in the organism and is known as glutaric acidemia type I (GA1). Despite the mechanisms of brain damage involved in GA1 are not fully understood, oxidative stress may be involved in this process. Treatment is based on protein/lysine (Lys) restriction and l-carnitine (L-car) supplementation. L-car was recently shown to have an important antioxidant role. A knockout mice model (Gcdh-/-) submitted to a dietary overload of Lys was developed to better understand the GA1 pathogenesis. In this study, we evaluated L-car and glutarylcarnitine levels, the lipid and protein damage, reactive oxygen species (ROS) production and antioxidant enzymes activities in striatum of Gcdh-/- and wild-type (WT) mice. We also determined the effect of the L-car treatment on these parameters. Thirty-day-old Gcdh-/- and WT mice were fed a normal chow (0.9% Lys) or submitted to a high Lys diet (4.7%) for 72 h. Additionally, these animals were administered with three intraperitoneal injections of saline or L-car in different times. Gcdh-/- mice were deficient in L-car and presented a higher glutarylcarnitine levels. They also presented lipid and protein damage, an increased ROS production and altered antioxidant enzymes compared to WT mice. Additionally, mice exposed to Lys overload presented higher alterations in these parameters than mice under normal diet, which were significantly decreased or normalized in those receiving L-car. Thus, we demonstrated a new beneficial effect of the L-car treatment attenuating or abolishing the oxidative stress process in Gcdh-/- mice.


Asunto(s)
Carnitina/farmacología , Cuerpo Estriado/metabolismo , Glutaril-CoA Deshidrogenasa/genética , Lisina/farmacología , Estrés Oxidativo/efectos de los fármacos , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/patología , Errores Innatos del Metabolismo de los Aminoácidos/veterinaria , Animales , Encefalopatías Metabólicas/metabolismo , Encefalopatías Metabólicas/patología , Encefalopatías Metabólicas/veterinaria , Carnitina/análogos & derivados , Carnitina/metabolismo , Dieta/veterinaria , Modelos Animales de Enfermedad , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/metabolismo , Glutatión Peroxidasa/metabolismo , Lisina/sangre , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
8.
J Cell Biochem ; 119(12): 10021-10032, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30129250

RESUMEN

The deficiency of the enzyme glutaryl-CoA dehydrogenase, known as glutaric acidemia type I (GA-I), leads to the accumulation of glutaric acid (GA) and glutarilcarnitine (C5DC) in the tissues and body fluids, unleashing important neurotoxic effects. l-carnitine (l-car) is recommended for the treatment of GA-I, aiming to induce the excretion of toxic metabolites. l-car has also demonstrated an important role as antioxidant and anti-inflammatory in some neurometabolic diseases. This study evaluated GA-I patients at diagnosis moment and treated the oxidative damage to lipids, proteins, and the inflammatory profile, as well as in vivo and in vitro DNA damage, reactive nitrogen species (RNS), and antioxidant capacity, verifying if the actual treatment with l-car (100 mg kg-1 day-1 ) is able to protect the organism against these processes. Significant increases of GA and C5DC were observed in GA-I patients. A deficiency of carnitine in patients before the supplementation was found. GA-I patients presented significantly increased levels of isoprostanes, di-tyrosine, urinary oxidized guanine species, and the RNS, as well as a reduced antioxidant capacity. The l-car supplementation induced beneficial effects reducing these biomarkers levels and increasing the antioxidant capacity. GA, in three different concentrations, significantly induced DNA damage in vitro, and the l-car was able to prevent this damage. Significant increases of pro-inflammatory cytokines IL-6, IL-8, GM-CSF, and TNF-α were shown in patients. Thus, the beneficial effects of l-car presented in the treatment of GA-I are due not only by increasing the excretion of accumulated toxic metabolites, but also by preventing oxidative damage.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Carnitina/farmacología , Daño del ADN , Glutaril-CoA Deshidrogenasa/deficiencia , Estrés Oxidativo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Carnitina/uso terapéutico , Niño , Preescolar , Femenino , Glutaril-CoA Deshidrogenasa/efectos de los fármacos , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Lactante , Masculino , Sustancias Protectoras/farmacología , Sustancias Protectoras/uso terapéutico , Especies de Nitrógeno Reactivo
9.
J Inherit Metab Dis ; 40(1): 75-101, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27853989

RESUMEN

Glutaric aciduria type I (GA-I; synonym, glutaric acidemia type I) is a rare inherited metabolic disease caused by deficiency of glutaryl-CoA dehydrogenase located in the catabolic pathways of L-lysine, L-hydroxylysine, and L-tryptophan. The enzymatic defect results in elevated concentrations of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutaryl carnitine in body tissues, which can be reliably detected by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Most untreated individuals with GA-I experience acute encephalopathic crises during the first 6 years of life that are triggered by infectious diseases, febrile reaction to vaccinations, and surgery. These crises result in striatal injury and consequent dystonic movement disorder; thus, significant mortality and morbidity results. In some patients, neurologic disease may also develop without clinically apparent crises at any age. Neonatal screening for GA-I us being used in a growing number of countries worldwide and is cost effective. Metabolic treatment, consisting of low lysine diet, carnitine supplementation, and intensified emergency treatment during catabolism, is effective treatment and improves neurologic outcome in those individuals diagnosed early; treatment after symptom onset, however, is less effective. Dietary treatment is relaxed after age 6 years and should be supervised by specialized metabolic centers. The major aim of this second revision of proposed recommendations is to re-evaluate the previous recommendations (Kölker et al. J Inherit Metab Dis 30:5-22, 2007b; J Inherit Metab Dis 34:677-694, 2011) and add new research findings, relevant clinical aspects, and the perspective of affected individuals.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/tratamiento farmacológico , Glutaril-CoA Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Suplementos Dietéticos , Glutaratos/metabolismo , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Lisina/metabolismo
10.
Neuroscience ; 308: 64-74, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26343296

RESUMEN

The brain of children affected by organic acidemias develop acute neurodegeneration linked to accumulation of endogenous toxic metabolites like glutaric (GA), 3-hydroxyglutaric (3-OHGA), methylmalonic (MMA) and propionic (PA) acids. Excitotoxic and oxidative events are involved in the toxic patterns elicited by these organic acids, although their single actions cannot explain the extent of brain damage observed in organic acidemias. The characterization of co-adjuvant factors involved in the magnification of early toxic processes evoked by these metabolites is essential to infer their actions in the human brain. Alterations in the kynurenine pathway (KP) - a metabolic route devoted to degrade tryptophan to form NAD(+) - produce increased levels of the excitotoxic metabolite quinolinic acid (QUIN), which has been involved in neurodegenerative disorders. Herein we investigated the effects of subtoxic concentrations of GA, 3-OHGA, MMA and PA, either alone or in combination with QUIN, on early toxic endpoints in rat brain synaptosomes. To establish specific mechanisms, we pre-incubated synaptosomes with different protective agents, including the endogenous N-methyl-d-aspartate (NMDA) receptor antagonist kynurenic acid (KA), the antioxidant S-allylcysteine (SAC) and the nitric oxide synthase (NOS) inhibitor nitro-l-arginine methyl ester (l-NAME). While the incubation of synaptosomes with toxic metabolites at subtoxic concentrations produced no effects, their co-incubation (QUIN+GA, +3-OHGA, +MMA or +PA) decreased the mitochondrial function and increased reactive oxygen species (ROS) formation and lipid peroxidation. For all cases, this effect was partially prevented by KA and l-NAME, and completely avoided by SAC. These findings suggest that early damaging events elicited by organic acids involved in metabolic acidemias can be magnified by toxic synergism with QUIN, and this process is mostly mediated by oxidative stress, and in a lesser extent by excitotoxicity and nitrosative stress. Therefore, QUIN can be hypothesized to contribute to the pathophysiology of brain degeneration in children with metabolic acidemias.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Encéfalo/metabolismo , Glutaratos/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Ácido Quinolínico/metabolismo , Sinaptosomas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Glutaratos/toxicidad , Glutaril-CoA Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ácido Metilmalónico/metabolismo , Ácido Metilmalónico/toxicidad , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Propionatos/metabolismo , Propionatos/toxicidad , Ácido Quinolínico/toxicidad , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Sinaptosomas/efectos de los fármacos
11.
PLoS One ; 9(10): e110181, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25333616

RESUMEN

Acute neurological crises involving striatal degeneration induced by a deficiency of glutaryl-CoA dehydrogenase (GCDH) and the accumulation of glutaric (GA) and 3-hydroxyglutaric acid (3-OHGA) are considered to be the most striking features of glutaric aciduria type I (GA1). In the present study, we investigated the mechanisms of apoptosis and energy metabolism impairment in our novel GA1 neuronal model. We also explored the effects of appropriate amounts of amino acids (2 mM arginine, 2 mM homoarginine, 0.45 g/L tyrosine and 10 mM leucine) and 2 g/L glucose on these cells. Our results revealed that the novel GA1 neuronal model effectively simulates the hypermetabolic state of GA1. We found that leucine, tyrosine, arginine, homoarginine or glucose treatment of the GA1 model cells reduced the gene expression of caspase-3, caspase-8, caspase-9, bax, fos, and jun and restored the intracellular NADH and ATP levels. Tyrosine, arginine or homoarginine treatment in particular showed anti-apoptotic effects; increased α-ketoglutarate dehydrogenase complex (OGDC), fumarase (FH), and citrate synthase (CS) expression; and relieved the observed impairment in energy metabolism. To the best of our knowledge, this study is the first to investigate the protective mechanisms of amino acids and glucose in GA1 at the cellular level from the point of view of apoptosis and energy metabolism. Our data support the results of previous studies, indicating that supplementation of arginine and homoarginine as a dietary control strategy can have a therapeutic effect on GA1. All of these findings facilitate the understanding of cell apoptosis and energy metabolism impairment in GA1 and reveal new therapeutic perspectives for this disease.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Técnicas de Cultivo de Célula , Glucosa/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Adenosina Trifosfato/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/genética , Animales , Animales Recién Nacidos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Encefalopatías Metabólicas/genética , Supervivencia Celular , Células Cultivadas , Ciclo del Ácido Cítrico/genética , Cuerpo Estriado/citología , Citometría de Flujo , Expresión Génica , Glutaril-CoA Deshidrogenasa/genética , Glutaril-CoA Deshidrogenasa/metabolismo , NAD/metabolismo , Neuronas/metabolismo , Ratas
12.
J Clin Anesth ; 25(2): 141-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23352788

RESUMEN

The management of cesarean delivery for a parturient with placenta previa at 36 weeks' gestation and glutaric aciduria type 1 is presented. The management goal was to prevent encephalopathic crisis by ensuring adequate caloric intake with dextrose infusion and to provide carnitine supplementation and adequate anesthesia.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Encefalopatías Metabólicas/terapia , Cesárea , Complicaciones del Embarazo/terapia , Atención Prenatal/métodos , Anestesia Obstétrica/métodos , Anestesia Raquidea/métodos , Carnitina/uso terapéutico , Femenino , Glucosa/administración & dosificación , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Infusiones Intravenosas , Atención Perioperativa/métodos , Placenta Previa/cirugía , Embarazo , Complejo Vitamínico B/uso terapéutico , Adulto Joven
13.
J Inherit Metab Dis ; 36(3): 525-33, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22971958

RESUMEN

BACKGROUND: Metabolic treatment in glutaric aciduria type I (GA-I) including a low lysine diet with lysine-free, tryptophan-reduced amino acid supplements (AAS), carnitine supplementation and early start of emergency treatment during putatively threatening episodes of intermittent febrile illness dramatically improves the outcome and thus has been recommended by an international guideline group (Kölker et al, J Inherit Metab Dis 30:5-22, 2007). However, possible affection of linear growth, weight gain and biochemical follow-up monitoring has not been studied systematically. METHODS: Thirty-three patients (n = 29 asymptomatic, n = 4 dystonic) with GA-I who have been identified by newborn screening in Germany from 1999 to 2009 were followed prospectively during the first six years of life. Dietary treatment protocols, anthropometrical and biochemical parameters were longitudinally evaluated. RESULTS: Mean daily intake as percentage of guideline recommendations was excellent for lysine (asymptomatic patients: 101 %; dystonic patients: 103 %), lysine-free, tryptophan-reduced AAS (108 %; 104 %), energy (106 %; 110 %), and carnitine (92 %; 102 %). Low lysine diet did not affect weight gain (mean SDS 0.05) but mildly impaired linear growth in asymptomatic patients (mean SDS -0.38), while dystonic patients showed significantly reduced weight gain (mean SDS -1.32) and a tendency towards linear growth retardation (mean SDS -1.03). Patients treated in accordance with recent recommendations did not show relevant abnormalities of routine biochemical follow-up parameters. INTERPRETATION: Low lysine diet promotes sufficient intake of essential nutrients and anthropometric development in asymptomatic children up to age 6 year, whereas individualized nutritional concepts are required for dystonic patients. Revised recommendations for biochemical monitoring might be required for asymptomatic patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Pesos y Medidas Corporales , Encefalopatías Metabólicas/dietoterapia , Alimentos Formulados , Glutaril-CoA Deshidrogenasa/deficiencia , Lisina/administración & dosificación , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Antropometría , Biomarcadores/análisis , Biomarcadores/sangre , Encefalopatías Metabólicas/sangre , Encefalopatías Metabólicas/metabolismo , Encefalopatías Metabólicas/fisiopatología , Carnitina/administración & dosificación , Niño , Preescolar , Suplementos Dietéticos , Ingestión de Alimentos/fisiología , Femenino , Estudios de Seguimiento , Glutaril-CoA Deshidrogenasa/sangre , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Lactante , Masculino , Monitoreo Fisiológico/métodos
14.
Mol Genet Metab ; 108(1): 30-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23218171

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Glutaril-CoA Deshidrogenasa/metabolismo , Homeostasis , Lisina/administración & dosificación , Animales , Suplementos Dietéticos , Glutaril-CoA Deshidrogenasa/genética , Ratones , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
15.
Mol Genet Metab ; 107(1-2): 72-80, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22520952

RESUMEN

The cerebral formation and entrapment of neurotoxic dicarboxylic metabolites (glutaryl-CoA, glutaric and 3-hydroxyglutaric acid) are considered to be important pathomechanisms of striatal injury in glutaric aciduria type I (GA-I). The quantitatively most important precursor of these metabolites is lysine. Recommended therapeutic interventions aim to reduce lysine oxidation (low lysine diet, emergency treatment to minimize catabolism) and to enhance physiologic detoxification of glutaryl-CoA via formation of glutarylcarnitine (carnitine supplementation). It has been recently shown in Gcdh(-/-) mice that cerebral lysine influx and oxidation can be modulated by arginine which competes with lysine for transport at the blood-brain barrier and the inner mitochondrial membrane [Sauer et al., Brain 134 (2011) 157-170]. Furthermore, short-term outcome of 12 children receiving arginine-fortified diet showed very promising results [Strauss et al., Mol. Genet. Metab. 104 (2011) 93-106]. Since lysine-free, arginine-fortified amino acid supplements (AAS) are commercially available and used in Germany for more than a decade, we evaluated the effect of arginine supplementation in a cohort of 34 neonatally diagnosed GA-I patients (median age, 7.43 years; cumulative follow-up period, 221.6 patient years) who received metabolic treatment according to a published guideline [Kölker et al., J. Inherit. Metab. Dis. 30 (2007) 5-22]. Patients used one of two AAS product lines during the first year of life, resulting in differences in arginine consumption [group 1 (Milupa Metabolics): mean=111 mg arginine/kg; group 2 (Nutricia): mean=145 mg arginine/kg; p<0.001]. However, in both groups the daily arginine intake was increased (mean, 137 mg/kg body weight) and the dietary lysine-to-arginine ratio was decreased (mean, 0.7) compared to infants receiving human milk and other natural foods only. All other dietary parameters were in the same range. Despite significantly different arginine intake, the plasma lysine-to-arginine ratio did not differ in both groups. Frequency of dystonia was low (group 1: 12.5%; group 2: 8%) compared with patients not being treated according to the guideline, and gross motor development was similar in both groups. In conclusion, the development of complementary dietary strategies exploiting transport competition between lysine and arginine for treatment of GA-I seems promising. More work is required to understand neuroprotective mechanisms of arginine, to develop dietary recommendations for arginine and to evaluate the usefulness of plasma monitoring for lysine and arginine levels as predictors of cerebral lysine influx.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Encefalopatías Metabólicas/dietoterapia , Suplementos Dietéticos , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Arginina/sangre , Arginina/metabolismo , Encéfalo/metabolismo , Encefalopatías Metabólicas/diagnóstico , Niño , Preescolar , Femenino , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Lactante , Lisina/sangre , Lisina/metabolismo , Masculino , Resultado del Tratamiento
16.
Med Sci Monit ; 17(7): PH55-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21709643

RESUMEN

BACKGROUND: Glutaric aciduria type I (GA I; MIM 231670) is a rare autosomal recessive disorder resulting from glutaryl-CoA dehydrogenase deficiency. This article reports our experience in the diagnosis, treatment and outcome of GA I patients in Zhejiang Province, China. MATERIAL/METHODS: A total of 129,415 newborns (accounting for approximately one-tenth of the annual births in Zhejiang Province) and 9640 high-risk infants were screened for inborn errors of metabolism in the Neonatal Screening Center of Zhejiang Province during a 3-year period. Tandem mass spectrometry and gas chromatography-mass spectrometry were used for diagnosis of the patients. Dietary modification, carnitine supplementation and aggressive treatment of intercurrent illnesses were adapted for GA I patients. RESULTS: Three infants were diagnosed with GA I by high-risk screening (detection rate: 1/3,213) and 2 were diagnosed by newborn screening (incidence: 1/64,708). Four patients (3 by high-risk screening and 1 by neonatal screening) undergoing MRI examination showed remarkable changes on T2-weighted image. Four patients accepted timely treatment, and in the patient diagnosed by neonatal screening, treatment was delayed until hypotonia appeared 3 months later. Neuropsychological assessment showed mental and motor retardation in 3 patients after treatment, including the patient diagnosed by neonatal screening. CONCLUSIONS: Individualized timely treatment and close monitoring of GA I patients needs to be optimized in China. Appropriate communication with parents may help to achieve successful management of GA I patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Errores Innatos del Metabolismo de los Aminoácidos/epidemiología , Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/tratamiento farmacológico , Encefalopatías Metabólicas/epidemiología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Encefalopatías Metabólicas/genética , Carnitina/uso terapéutico , China/epidemiología , Femenino , Cromatografía de Gases y Espectrometría de Masas , Genes Recesivos , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Humanos , Incidencia , Lactante , Recién Nacido , Masculino , Tamizaje Masivo , Pruebas Neuropsicológicas , Riboflavina/uso terapéutico , Espectrometría de Masas en Tándem , Resultado del Tratamiento
17.
Methods Enzymol ; 493: 91-114, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21371588

RESUMEN

Fragment screening using X-ray crystallography is a method that can provide direct three-dimensional readouts of the structures of protein-small molecule complexes for lead development and fragment-based drug discovery. With current technology, an amenable crystal form can be screened crystallographically against a library of 1000-2000 fragments in 1-2 weeks. We have performed over a dozen crystallographic screening campaigns using our own compound collection called Fragments of Life™ (FOL). While the majority of our fragment screening campaigns have generated multiple hits, some unexpectedly turned out to be nonproductive, either yielding no bound ligands, or only those thought to be inadequate for lead development. In this chapter, we have attempted to identify one or more parameters which could be used to predict whether a crystallized protein target would be a good candidate for fragment hit discovery. Here, we describe the parameters of crystals from 18 fragment screening campaigns, including six unsuccessful targets. From this analysis, we have concluded that there are no parameters that are absolutely predictive of fragment screening success. However, we do describe a parameter we have termed pocket factor which provides a statistically significant variance between nonproductive targets and productive targets shown to bind fragments. The pocket factor is calculated using a novel method of consensus scoring from three distinct pocket-finding algorithms, and the results may be used to prioritize targets for fragment screening campaigns based on an initial crystal structure.


Asunto(s)
Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos/métodos , Genómica/métodos , 3-Oxoacil-(Proteína Transportadora de Acil) Sintasa/química , Algoritmos , Burkholderia pseudomallei/enzimología , Enfermedades Transmisibles , Simulación por Computador , Cristalización , Descubrimiento de Drogas/métodos , Eritritol/análogos & derivados , Eritritol/biosíntesis , Glutaril-CoA Deshidrogenasa/química , Isoenzimas/química , Ligandos , Modelos Moleculares , Fosfoglicerato Mutasa/química , Liasas de Fósforo-Oxígeno/química , Unión Proteica , Bibliotecas de Moléculas Pequeñas , Solventes
18.
J Bioenerg Biomembr ; 43(1): 31-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21249436

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ácidos Carboxílicos/metabolismo , Homeostasis/fisiología , Errores Innatos del Metabolismo/fisiopatología , Mitocondrias/fisiología , Enfermedades Mitocondriales/fisiopatología , Acetil-CoA C-Aciltransferasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Animales , Síndrome de Barth/fisiopatología , Encefalopatías Metabólicas/fisiopatología , Encefalopatías Metabólicas Innatas/fisiopatología , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Mitocondrias/metabolismo , Acidemia Propiónica/fisiopatología , Púrpura/fisiopatología
19.
Brain ; 134(Pt 1): 157-70, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20923787

RESUMEN

Glutaric aciduria type I, an inherited deficiency of glutaryl-coenzyme A dehydrogenase localized in the final common catabolic pathway of L-lysine, L-hydroxylysine and L-tryptophan, leads to accumulation of neurotoxic glutaric and 3-hydroxyglutaric acid, as well as non-toxic glutarylcarnitine. Most untreated patients develop irreversible brain damage during infancy that can be prevented in the majority of cases if metabolic treatment with a low L-lysine diet and L-carnitine supplementation is started in the newborn period. The biochemical effect of this treatment remains uncertain, since cerebral concentrations of neurotoxic metabolites can only be determined by invasive techniques. Therefore, we studied the biochemical effect and mechanism of metabolic treatment in glutaryl-coenzyme A dehydrogenase-deficient mice, an animal model with complete loss of glutaryl-coenzyme A dehydrogenase activity, focusing on the tissue-specific changes of neurotoxic metabolites and key enzymes of L-lysine metabolism. Here, we demonstrate that low L-lysine diet, but not L-carnitine supplementation, lowered the concentration of glutaric acid in brain, liver, kidney and serum. L-carnitine supplementation restored the free L-carnitine pool and enhanced the formation of glutarylcarnitine. The effect of low L-lysine diet was amplified by add-on therapy with L-arginine, which we propose to result from competition with L-lysine at system y(+) of the blood-brain barrier and the mitochondrial L-ornithine carriers. L-lysine can be catabolized in the mitochondrial saccharopine or the peroxisomal pipecolate pathway. We detected high activity of mitochondrial 2-aminoadipate semialdehyde synthase, the rate-limiting enzyme of the saccharopine pathway, in the liver, whereas it was absent in the brain. Since we found activity of the subsequent enzymes of L-lysine oxidation, 2-aminoadipate semialdehyde dehydrogenase, 2-aminoadipate aminotransferase and 2-oxoglutarate dehydrogenase complex as well as peroxisomal pipecolic acid oxidase in brain tissue, we postulate that the pipecolate pathway is the major route of L-lysine degradation in the brain and the saccharopine pathway is the major route in the liver. Interestingly, treatment with clofibrate decreased cerebral and hepatic concentrations of glutaric acid in glutaryl-coenzyme A dehydrogenase-deficient mice. This finding opens new therapeutic perspectives such as pharmacological stimulation of alternative L-lysine oxidation in peroxisomes. In conclusion, this study gives insight into the discrepancies between cerebral and hepatic L-lysine metabolism, provides for the first time a biochemical proof of principle for metabolic treatment in glutaric aciduria type I and suggests that further optimization of treatment could be achieved by exploitation of competition between L-lysine and L-arginine at physiological barriers and enhancement of peroxisomal L-lysine oxidation and glutaric acid breakdown.


Asunto(s)
Encéfalo/metabolismo , Lisina/metabolismo , 2-Aminoadipato-Transaminasa/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Análisis de Varianza , Animales , Arginina/metabolismo , Arginina/uso terapéutico , Encefalopatías Metabólicas/dietoterapia , Encefalopatías Metabólicas/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina/uso terapéutico , Catalasa/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Ratones
20.
J Inherit Metab Dis ; 30(6): 979, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17957492

RESUMEN

Late-onset neurological disease has rarely been reported in patients with glutaryl-CoA dehydrogenase (GCDH) deficiency. We present two siblings with GCDH deficiency. One of them presented with the classic neurological disease (patient 1). Routine investigation of family members revealed that her apparently unharmed 13-year-old sister was also affected (patient 2). Patient 2 started to have academic difficulties in the months prior to our assessment. Her clinical examination was normal, with the exception of a cranial circumference of 57 cm (slightly over the 98 th centile). A severe leukoencephalopathy was demonstrated on MRI. Neuropsychological assessment showed an IQ within the normal-low range and a mild impairment of memory and executive function. Previous reports on late-onset neurological disease in GCDH deficiency have revealed that progressive leukoencephalopathy develops over time. Following the recently published guideline for the diagnosis and management of GCDH deficiency, both patients are receiving dietary treatment in combination with L-carnitine supplementation. We emphasize the need to search for chronic neurological changes of late-onset type in apparently unaffected GCDH deficiency cases diagnosed in routine family investigations.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/patología , Glutaril-CoA Deshidrogenasa/deficiencia , Adolescente , Carnitina/uso terapéutico , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Memoria , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/patología , Neurología/métodos , Pruebas Neuropsicológicas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA