Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Eur J Pediatr ; 183(9): 3665-3678, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38954008

RESUMEN

The purpose of this study is to investigate the diagnostic and prognostic role of cerebrospinal fluid (CSF) biomarkers in the diagnostic work-up of glucose transporter 1 (GLUT1) deficiency. Reported here is a systematic review according to PRISMA guidelines collecting clinical and biochemical data about all published patients who underwent CSF analysis. Clinical phenotypes were compared between groups defined by the levels of CSF glucose (≤ 2.2 mmol/L versus > 2.2 mmol/L), CSF/blood glucose ratio (≤ 0.45 versus > 0.45), and CSF lactate (≤ 1 mmol/L versus > 1 mmol/L). Five hundred sixty-two patients fulfilled the inclusion criteria with a mean age at the diagnosis of 8.6 ± 6.7 years. Patients with CSF glucose ≤ 2.2 mmol/L and CSF/blood glucose ratio ≤ 0.45 presented with an earlier onset of symptoms (16.4 ± 22.0 versus 54.4 ± 45.9 months, p < 0.01; 15.7 ± 23.8 versus 40.9 ± 38.0 months, p < 0.01) and received an earlier molecular genetic confirmation (92.1 ± 72.8 versus 157.1 ± 106.2 months, p < 0.01). CSF glucose ≤ 2.2 mmol/L was consistently associated with response to ketogenic diet (p = 0.018) and antiseizure medications (p = 0.025). CSF/blood glucose ratio ≤ 0.45 was significantly associated with absence seizures (p = 0.048), paroxysmal exercise-induced dyskinesia (p = 0.046), and intellectual disability (p = 0.016) while CSF lactate > 1 mmol/L was associated with a response to antiseizure medications (p = 0.026) but not to ketogenic diet.Conclusions:This systematic review supported the diagnostic usefulness of lumbar puncture for the early identification of patients with GLUT1 deficiency responsive to treatments especially if they present with co-occurring epilepsy, movement, and neurodevelopmental disorders. What is Known: • Phenotypes of GLUT1 deficiency syndrome range between early epileptic and developmental encephalopathy to paroxysmal movement disorders and developmental impairment What is New: • CSF blood/glucose ratio may predict better than CSF glucose the diagnosis in children presenting with early onset absences • CSF blood/glucose ratio may predict better than CSF glucose the diagnosis in children presenting with paroxysmal exercise induced dyskinesia and intellectual disability. • CSF glucose may predict better than CSF blood/glucose and lactate the response to ketogenic diet and antiseizure medications.


Asunto(s)
Biomarcadores , Errores Innatos del Metabolismo de los Carbohidratos , Humanos , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/sangre , Pronóstico , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Niño , Ácido Láctico/líquido cefalorraquídeo , Ácido Láctico/sangre , Glucemia/análisis , Dieta Cetogénica , Preescolar , Transportador de Glucosa de Tipo 1/genética , Glucosa/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/deficiencia
4.
Mol Genet Metab ; 123(3): 331-336, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29307761

RESUMEN

PURA is a DNA/RNA-binding protein known to have an important role as a transcriptional and translational regulator. Mutations in the PURA gene have been documented to cause mainly a neurologic phenotype including hypotonia, epilepsy, development delay and respiratory alterations. We report here a patient with a frame-shift deletion in the PURA gene that apart from the classical PURA deficiency phenotype had marked hypoglycorrhachia, overlapping the clinical findings with a GLUT1 deficiency syndrome. SLC2A1 (GLUT1) mutations were discarded, so we hypothesized that GLUT1 could be downregulated in this PURA deficient scenario. We confirmed reduced GLUT1 expression in the patient's peripheral blood cells compared to controls predicting that this could also be happening in the blood-brain barrier and in this way explain the hypoglycorrhachia. Based on PURA's known functions as a transcriptional and translational regulator, we propose GLUT1 as a new PURA target. Further in vitro and in vivo studies are needed to confirm this and to uncover the underlying molecular mechanisms.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/genética , Proteínas de Unión al ADN/genética , Transportador de Glucosa de Tipo 1/metabolismo , Glucosa/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/deficiencia , Factores de Transcripción/genética , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Errores Innatos del Metabolismo de los Carbohidratos/patología , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Femenino , Mutación del Sistema de Lectura , Humanos , Recién Nacido , Leucocitos/metabolismo , Proteínas de Transporte de Monosacáridos/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/genética , Factores de Transcripción/metabolismo , Secuenciación del Exoma
5.
Dev Med Child Neurol ; 58(12): 1295-1302, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27265003

RESUMEN

AIM: Loss-of-function mutations in SLC2A1, encoding glucose transporter-1 (GLUT-1), lead to dysfunction of glucose transport across the blood-brain barrier. Ten percent of cases with hypoglycorrhachia (fasting cerebrospinal fluid [CSF] glucose <2.2mmol/L) do not have mutations. We hypothesized that GLUT1 deficiency could be due to non-coding SLC2A1 variants. METHOD: We performed whole exome sequencing of one proband with a GLUT1 phenotype and hypoglycorrhachia negative for SLC2A1 sequencing and copy number variants. We studied a further 55 patients with different epilepsies and low CSF glucose who did not have exonic mutations or copy number variants. We sequenced non-coding promoter and intronic regions. We performed mRNA studies for the recurrent intronic variant. RESULTS: The proband had a de novo splice site mutation five base pairs from the intron-exon boundary. Three of 55 patients had deep intronic SLC2A1 variants, including a recurrent variant in two. The recurrent variant produced less SLC2A1 mRNA transcript. INTERPRETATION: Fasting CSF glucose levels show an age-dependent correlation, which makes the definition of hypoglycorrhachia challenging. Low CSF glucose levels may be associated with pathogenic SLC2A1 mutations including deep intronic SLC2A1 variants. Extending genetic screening to non-coding regions will enable diagnosis of more patients with GLUT1 deficiency, allowing implementation of the ketogenic diet to improve outcomes.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Errores Innatos del Metabolismo de los Carbohidratos/genética , Epilepsia/líquido cefalorraquídeo , Epilepsia/genética , Transportador de Glucosa de Tipo 1/genética , Glucosa/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/deficiencia , Adulto , Preescolar , Exoma , Femenino , Humanos , Lactante , Masculino , Proteínas de Transporte de Monosacáridos/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/genética , Linaje , Análisis de Secuencia
6.
JAMA Neurol ; 70(11): 1440-4, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23999624

RESUMEN

IMPORTANCE: GLUT1 deficiency syndrome is a treatable neurometabolic disorder, characterized by a low concentration of glucose in cerebrospinal fluid (CSF) and a decreased CSF to blood glucose ratio. Reports of patients with apparently normal CSF glucose levels, however, have raised the question whether CSF analysis is a reliable screening tool for GLUT1 deficiency syndrome. OBJECTIVE: To determine the value of CSF analysis in the workup of GLUT1 deficiency syndrome. EVIDENCE REVIEW: PubMed was searched until July 2012 by using the terms glucose transporter 1 (GLUT-1) deficiency syndrome, glucose transporter defect, and SLC2A1-gene. Relevant references mentioned in the articles were also included. The CSF results of all patients with genetically proven GLUT1 deficiency syndrome described in literature were reevaluated. FINDINGS: The levels of glucose in CSF, the CSF to blood glucose ratios, and the levels of lactate in CSF were reported for 147 (94%), 152 (97%), and 73 (46%) of 157 patients, respectively. The CSF glucose levels ranged from 16.2 to 50.5 mg/dL and were at or below the 10th percentile for all 147 patients. The CSF to blood glucose ratios ranged from 0.19 to 0.59 and were at or below the 10th percentile for 139 of 152 patients (91%), but they could be within the normal range as well. The CSF lactate levels ranged from 5.4 to 13.5 mg/dL and were at or below the 10th percentile for 59 of 73 patients (81%). A typical CSF profile for GLUT1 deficiency syndrome, which is defined as a CSF glucose level at or below the 10th percentile, a CSF to blood glucose ratio at or below the 25th percentile, and a CSF lactate level at or below the 10th percentile, was found in only 35 of 4099 CSF samples (0.9%) present in our CSF database of patients who received a diagnosis other than GLUT1 deficiency syndrome. CONCLUSIONS AND RELEVANCE: We conclude that if age-specific reference values are applied, CSF glucose and lactate levels are adequate biomarkers in the diagnostic workup of GLUT1 deficiency syndrome. Future availability of whole-exome sequencing in clinical practice will make the existence of a reliable biomarker for GLUT1 deficiency syndrome even more important, in order to interpret genetic results and, even more importantly, not to miss SLC2A1-negative patients with GLUT1 deficiency syndrome.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Glucosa/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/deficiencia , Factores de Edad , Glucemia , Errores Innatos del Metabolismo de los Carbohidratos/sangre , Humanos , Proteínas de Transporte de Monosacáridos/sangre , Proteínas de Transporte de Monosacáridos/líquido cefalorraquídeo , PubMed/estadística & datos numéricos
7.
J Child Neurol ; 27(6): 796-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22156785

RESUMEN

Mitochondrial disorders are varied in their clinical presentation and pathogenesis. Diagnosis is usually made clinically and genetic defects are often not identified. We present a 6-year-old female patient with a diagnosis of a mitochondrial disorder secondary to complex I deficiency with seizures and developmental delay from infancy. Glucose transporter deficiency was suspected after a lumbar puncture showed hypoglycorrhachia. Her disorder was confirmed genetically as a mutation in her solute carrier family 2, facilitated glucose transporter member 1 (SLCA2) gene. Delayed diagnosis led to delayed treatment, and neurologic sequelae may have been prevented by earlier recognition of this disorder.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/complicaciones , Errores Innatos del Metabolismo de los Carbohidratos/genética , Discapacidades del Desarrollo/genética , Transportador de Glucosa de Tipo 2/deficiencia , Enfermedades Mitocondriales/etiología , Enfermedades Mitocondriales/genética , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Niño , Discapacidades del Desarrollo/líquido cefalorraquídeo , Discapacidades del Desarrollo/complicaciones , Femenino , Glucosa/líquido cefalorraquídeo , Humanos , Enfermedades Mitocondriales/líquido cefalorraquídeo , Punción Espinal
8.
Dev Med Child Neurol ; 53(12): 1154-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21838819

RESUMEN

Glucose transporter type 1 deficiency syndrome (GLUT1-DS) is caused by a defect in glucose transport across the blood-brain barrier. The main symptoms are epilepsy, developmental delay, movement disorders, and deceleration of head circumference. A ketogenic diet has been shown to be effective in controlling epilepsy in GLUT1-DS. We report a female child (3 y 4 mo) who presented with delayed psychomotor development and frequent episodes of staggering, impaired vigilance, and vomiting that resolved promptly after food intake. Electroencephalography was normal. The cerebrospinal fluid-blood glucose ratio was 0.42 (normal ≥ 0.45). GLUT1-DS was confirmed by molecular genetic testing, which showed a novel de novo heterozygous mutation in the SLC2A1 gene (c.497_499delTCG, p.VAL166del). Before starting a ketogenic diet, the child's cognitive development was tested using the Snijders-Oomen Non-Verbal Intelligence Test, which revealed a heterogeneous intelligence profile with deficits in her visuomotor skills and spatial awareness. Her motor development was delayed. Three months after introducing a ketogenic diet, she showed marked improvement in speech and motor development, as tested by the Movement Assessment Battery for Children (manual dexterity 16th centile, ball skills 1st centile, static and dynamic balance 5th centile). This case demonstrates that GLUT1-DS should be investigated in individuals with unexplained developmental delay. Epilepsy is not a mandatory symptom. The ketogenic diet is also beneficial for non-epileptic symptoms in GLUT1-DS.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/fisiopatología , Dieta Cetogénica/estadística & datos numéricos , Epilepsia , Glucemia/genética , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Errores Innatos del Metabolismo de los Carbohidratos/terapia , Preescolar , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Epilepsia/genética , Femenino , Trastornos del Metabolismo de la Glucosa/genética , Trastornos del Metabolismo de la Glucosa/fisiopatología , Transportador de Glucosa de Tipo 1/genética , Heterocigoto , Humanos , Proteínas de Transporte de Monosacáridos/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/deficiencia , Trastornos del Movimiento/diagnóstico , Trastornos del Movimiento/genética , Mutación/genética , Pruebas Neuropsicológicas , Síndrome
9.
Dev Med Child Neurol ; 53(7): 664-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21649651

RESUMEN

Glucose transporter type 1 deficiency syndrome (GLUT1DS) is a treatable condition resulting from impaired glucose transport into the brain. The classical presentation is with infantile-onset epilepsy and severe developmental delay. Non-classical phenotypes with movement disorders and early-onset absence epilepsy are increasingly recognized and the clinical spectrum is expanding. The hallmark is hypoglycorrhachia (cerebrospinal fluid [CSF] glucose<2.2 mmol/l) in the presence of normoglycaemia with a CSF/blood glucose ratio of less than 0.4. GLUT1DS is due to a mutation in the solute carrier family 2, member 1 gene (SLC2A1). We present five individuals (four males, one female), all of whom had a mild phenotype, highlighting the importance of considering this diagnosis in unexplained neurological disorders associated with mild learning difficulties, subtle motor delay, early-onset absence epilepsy, fluctuating gait disorders, and/or dystonia. The mean age at diagnosis was 8 years 8 months. This paper also shows phenotypical parallels between GLUT1DS and paroxysmal exertion-induced dyskinesia.


Asunto(s)
Transportador de Glucosa de Tipo 1/genética , Glucosa/líquido cefalorraquídeo , Adolescente , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Errores Innatos del Metabolismo de los Carbohidratos/genética , Errores Innatos del Metabolismo de los Carbohidratos/fisiopatología , Niño , Preescolar , Diagnóstico Diferencial , Distonía/genética , Epilepsia Tipo Ausencia/genética , Femenino , Marcha , Humanos , Masculino , Proteínas de Transporte de Monosacáridos/líquido cefalorraquídeo , Proteínas de Transporte de Monosacáridos/deficiencia , Proteínas de Transporte de Monosacáridos/genética , Actividad Motora , Mutación , Fenotipo , Índice de Severidad de la Enfermedad
10.
Eur J Pediatr ; 161(6): 295-304, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12029447

RESUMEN

UNLABELLED: Facilitated glucose transporter protein type 1 (GLUT1) deficiency syndrome (MIM 138140) defines a prototype of a novel group of disorders resulting from impaired glucose transport across blood-tissue barriers. It is caused by a defect in glucose transport into brain, mediated by the facilitative glucose transporter GLUT1. Since 1991, more than 70 patients have been identified. The hallmark of the disease is a low glucose concentration in the CSF (hypoglycorrhachia) in the presence of normoglycaemia (CSF/blood glucose ratio <0.4). Clinical features are variable and include seizures, developmental delay, acquired microcephaly, hypotonia, and a complex motor disorder with elements of ataxia, dystonia, and spasticity. The GLUT1 defect can be confirmed in erythrocytes by glucose uptake studies and GLUT1 immunoreactivity, and by molecular analysis of the GLUT1 gene. Several heterozygous mutations resulting in GLUT1 haploinsufficiency have been identified. An effective treatment is available by means of a ketogenic diet as ketone bodies serve as an alternative fuel for the developing brain. CONCLUSION: this treatable condition should be suspected in children with unexplained neurological disorders associated with epilepsy and developmental delay and confirmed by a lumbar puncture.


Asunto(s)
Encéfalo/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/deficiencia , Barrera Hematoencefálica , Errores Innatos del Metabolismo de los Carbohidratos/sangre , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Preescolar , Glucosa/líquido cefalorraquídeo , Transportador de Glucosa de Tipo 1 , Humanos , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , Proteínas de Transporte de Monosacáridos/genética , Mutación , Síndrome
11.
NMR Biomed ; 14(3): 167-76, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11357181

RESUMEN

In vivo NMR spectroscopy was performed on the brain of a patient with a leukoencephalopathy, revealing unknown resonances between 3.5 and 4.0 ppm. In addition, urine and CSF of the patient were measured using high-resolution NMR spectroscopy. Also in these in vitro spectra, unknown resonances were observed in the 3.5-4.0 ppm region. Homonuclear (1)H two-dimensional J-resolved spectroscopy (JRES) and (1)H-(1)H correlation spectroscopy (COSY) were performed on the patient's urine for more accurate assignment of resonances. The NMR spectroscopic studies showed that the unknown resonances could be assigned to arabinitol and ribitol. This was confirmed using gas chromatography. The arabinitol was identified as D-arabinitol. The patient is likely to suffer from an as yet unknown inborn error of metabolism affecting D-arabinitol and ribitol metabolism. The primary molecular defect has not been found yet. Urine spectra of patients suffering from diabetes mellitus or galactosemia were recorded for comparison. Resonances outside the 3.2-4.0 ppm region, which are the most easy to recognize in body fluid spectra, allow easy recognition of various sugars and polyols. The paper shows that NMR spectroscopy in body fluids may help identifying unknown resonances observed in in vivo NMR spectra.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Espectroscopía de Resonancia Magnética/métodos , Ribitol/metabolismo , Alcoholes del Azúcar/metabolismo , Adolescente , Encefalopatías/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/líquido cefalorraquídeo , Errores Innatos del Metabolismo de los Carbohidratos/orina , Líquido Cefalorraquídeo/química , Cromatografía de Gases , Humanos , Masculino , Lóbulo Parietal/química , Ribitol/análisis , Ribitol/orina , Alcoholes del Azúcar/análisis , Alcoholes del Azúcar/orina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA