Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Neurogenetics ; 25(2): 69-78, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38190079

RESUMO

Glucose transporter type 1 deficiency syndrome (GLUT-1DS) is characterized by alterations in glucose translocation through the blood-brain barrier (BBB) due to mutation involving the GLUT-1 transporter. The fundamental therapy is ketogenic diet (KD) that provide an alternative energetic substrate - ketone bodies that across the BBB via MCT-1 - for the brain. Symptoms are various and include intractable seizure, acquired microcephalia, abnormal ocular movement, movement disorder, and neurodevelopment delay secondary to an energetic crisis for persistent neuroglycopenia. KD is extremely effective in controlling epileptic seizures and has a positive impact on movement disorders and cognitive impairment. Cases of KD resistance are rare, and only a few of them are reported in the literature, all regarding seizure. Our study describes a peculiar case of GLUT-1DS due to a new deletion involving the first codon of SLC2A1 gene determining a loss of function with a resistance to KD admitted to hospital due to intractable episodes of dystonia. This patient presented a worsening of symptomatology at higher ketonemia values but without hyperketosis and showed a complete resolution of symptomatology while maintaining low ketonemia values. Our study proposes an in-silico genomic and proteomic analysis aimed at explaining the atypical response to KD exhibited by our patient. In this way, we propose a new clinical and research approach based on precision medicine and molecular modelling to be applied to patients with GLUT-1DS resistant to first-line treatment with ketogenic diet by in silico study of genetic and altered protein product.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos , Dieta Cetogênica , Transportador de Glucose Tipo 1 , Proteínas de Transporte de Monossacarídeos/deficiência , Humanos , Transportador de Glucose Tipo 1/genética , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Masculino , Feminino , Simulação por Computador
2.
Epilepsia ; 65(8): 2213-2226, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38767952

RESUMO

In glucose transporter 1 deficiency syndrome (Glut1DS), glucose transport into brain is reduced due to impaired Glut1 function in endothelial cells at the blood-brain barrier. This can lead to shortages of glucose in brain and is thought to contribute to seizures. Ketogenic diets are the first-line treatment and, among many beneficial effects, provide auxiliary fuel in the form of ketone bodies that are largely metabolized by neurons. However, Glut1 is also the main glucose transporter in astrocytes. Here, we review data indicating that glucose shortage may also impact astrocytes in addition to neurons and discuss the expected negative biochemical consequences of compromised astrocytic glucose transport for neurons. Based on these effects, auxiliary fuels are needed for both cell types and adding medium chain triglycerides (MCTs) to ketogenic diets is a biochemically superior treatment for Glut1DS compared to classical ketogenic diets. MCTs provide medium chain fatty acids (MCFAs), which are largely metabolized by astrocytes and not neurons. MCFAs supply energy and contribute carbons for glutamine and γ-aminobutyric acid synthesis, and decanoic acid can also block α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors. MCTs do not compete with metabolism of ketone bodies mostly occurring in neurons. Triheptanoin, an anaplerotic but also gluconeogenic uneven MCT, may be another potential addition to ketogenic diets, although maintenance of "ketosis" can be difficult. Gene therapy has also targeted both endothelial cells and astrocytes. Other approaches to increase fuel delivery to the brain currently investigated include exchange of Glut1DS erythrocytes with healthy cells, infusion of lactate, and pharmacological improvement of glucose transport. In conclusion, although it remains difficult to assess impaired astrocytic energy metabolism in vivo, astrocytic energy needs are most likely not met by ketogenic diets in Glut1DS. Thus, we propose prospective studies including monitoring of blood MCFA levels to find optimal doses for add-on MCT to ketogenic diets and assessing of short- and long-term outcomes.


Assuntos
Astrócitos , Erros Inatos do Metabolismo dos Carboidratos , Dieta Cetogênica , Metabolismo Energético , Transportador de Glucose Tipo 1 , Astrócitos/metabolismo , Humanos , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Metabolismo Energético/fisiologia , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 1/genética , Glucose/metabolismo , Animais , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas de Transporte de Monossacarídeos/genética
3.
Neurol Sci ; 45(9): 4539-4547, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38589768

RESUMO

OBJECTIVE: The aim of the study is to analyze microstate patterns in GLUT1-DS, both before and after the ketogenic diet (KD). METHODS: We conducted microstate analysis of a patient with GLUT-1 DS and 27 healthy controls. A systematic literature review and meta-analysis was done. We compared the parameters of the patients with those of healthy controls and the incorporating findings in literature. RESULTS: The durations of the patient were notably shorter, and the occurrence rates were longer than those of healthy controls and incorporating findings from the review. After 10 months of KD, the patient's microstate durations exhibited an increase from 53.05 ms, 57.17 ms, 61.80 ms, and 49.49 ms to 60.53 ms, 63.27 ms, 71.11 ms, and 66.55 ms. The occurrence rates changed from 4.0774 Hz, 4.9462 Hz, 4.8006 Hz, and 4.0579 Hz to 3.3354 Hz, 3.7893 Hz, 3.5956 Hz, and 4.1672 Hz. In healthy controls, the durations of microstate class A, B, C, and D were 61.86 ms, 63.58 ms, 70.57 ms, and 72.00 ms, respectively. CONCLUSIONS: Our findings suggest EEG microstates may be a promising biomarker for monitoring the effect of KD. Administration of KD may normalize the dysfunctional patterns of temporal parameters.


Assuntos
Dieta Cetogênica , Eletroencefalografia , Humanos , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/fisiopatologia , Biomarcadores/sangue , Feminino , Proteínas de Transporte de Monossacarídeos/deficiência , Masculino , Estudos Prospectivos
4.
J Hum Nutr Diet ; 37(4): 827-846, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38838079

RESUMO

BACKGROUND: The ketogenic diet (KD) is a high fat, moderate protein and very low carbohydrate diet. It can be used as a medical treatment for drug-resistant epilepsy (DRE), glucose transporter 1 deficiency syndrome and pyruvate dehydrogenase deficiency. The aim of this scoping review was to map the KD literature, with a focus on epilepsy and associated metabolic conditions, to summarise the current evidence-base and identify any gaps. METHODS: This review was conducted using JBI scoping review methodological guidance and the PRISMA extension for scoping reviews reporting guidance. A comprehensive literature search was conducted in September 2021 and updated in February 2024 using MEDLINE, CINAHL, AMED, EmBASE, CAB Abstracts, Scopus and Food Science Source databases. RESULTS: The initial search yielded 2721 studies and ultimately, data were extracted from 320 studies that fulfilled inclusion criteria for the review. There were five qualitative studies, and the remainder were quantitative, including 23 randomised controlled trials (RCTs) and seven quasi-experimental studies. The USA published the highest number of KD studies followed by China, South Korea and the UK. Most studies focused on the classical KD and DRE. The studies key findings suggest that the KD is efficacious, safe and tolerable. CONCLUSIONS: There are opportunities available to expand the scope of future KD research, particularly to conduct high-quality RCTs and further qualitative research focused on the child's needs and family support to improve the effectiveness of KDs.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos , Dieta Cetogênica , Epilepsia Resistente a Medicamentos , Doença da Deficiência do Complexo de Piruvato Desidrogenase , Humanos , Dieta Cetogênica/métodos , Doença da Deficiência do Complexo de Piruvato Desidrogenase/dietoterapia , Criança , Epilepsia Resistente a Medicamentos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Proteínas de Transporte de Monossacarídeos/deficiência , Pré-Escolar , Masculino , Feminino , Adolescente
5.
J Inherit Metab Dis ; 43(2): 216-222, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31605543

RESUMO

Glucose transporter type 1 deficiency syndrome (GLUT1DS) is characterised by deficient glucose transport over the blood-brain barrier and reduced glucose availability in the brain. This causes epilepsy, movement disorders, and cognitive impairment. Treatment with ketogenic diet provides ketones as alternative energy source. However, not all GLUT1DS patients are on dietary treatment (worldwide registry: 77/181 [43%] of patients). The current 25-year experience allows evaluation of effects and tolerability of dietary treatment for GLUT1DS. To this end, literature was searched up to January 2019 for individual case reports and series reporting (side) effects of dietary treatment for GLUT1DS. Upon aggregation of data for analysis, we identified 270 GLUT1DS patients with dietary treatment with a mean follow-up of 53 months. Epilepsy improved for 83% of 230 patients and remained unchanged for 17%, movement disorders improved for 82% of 127 patients and remained unchanged for 17%, and cognition improved for 59% of 58 patients and remained stable for 40%. Effects on epilepsy were seen within days/weeks and were most pronounced in patients with early treatment initiation. Effects on movement disorders were noticed within months and were strongest in patients with higher cerebrospinal fluid-to-blood glucose ratio. Although side effects were minimal, 18% of 270 patients reported poor compliance. In individual patients, symptoms deteriorated upon low ketosis, poor compliance, or treatment discontinuation. Based on the good tolerability and strong favourable effect of dietary treatment on GLUT1DS symptoms, we advocate dietary treatment in all GLUT1DS patients and prompt diagnosis or screening to allow early treatment.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Disfunção Cognitiva/dietoterapia , Dieta Cetogênica , Epilepsia/dietoterapia , Proteínas de Transporte de Monossacarídeos/deficiência , Transtornos dos Movimentos/dietoterapia , Disfunção Cognitiva/etiologia , Epilepsia/etiologia , Humanos , Transtornos dos Movimentos/etiologia
6.
Epilepsy Behav ; 106: 107036, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32247176

RESUMO

OBJECTIVE: Glucose transporter type 1 deficiency (G1D) syndrome is generally a genetic disorder because of a mutation of the SLC2A1 gene. The clinical picture of G1D is heterogeneous. The aim of this paper was to present the case of G1D, recognized in a three-generation family, caused by missense mutation p.Arg92Trp in SLC2A1 gene, and showing high clinical heterogeneity and evolution of symptoms over time. METHODS: Three-generation family members, showing symptoms suggesting G1D, have been characterized in terms of the clinical picture, electroencephalogram (EEG) recordings, brain neuroimaging, and the psychological assessment data. All subjects were offered genetic testing of the SLC2A1 gene. RESULTS: We sequenced the SLC2A1 gene in the proband of the family and identified the c.274C > T variant (p.Arg92Trp). The presence of the same mutation was confirmed in all affected family members; however, significant variations in the clinical picture among them were observed. In addition to the typical symptoms for G1D (e.g., epilepsy, intellectual disability), patients presented movement disorders, stiffness, and dysarthria, as well as psychiatric symptoms. After using the ketogenic diet, epileptic seizures disappeared, but the rest of the symptoms were resistant to treatment. CONCLUSIONS: Despite the same underlying mutation, clinical symptoms may vary among members of one family. Different clinical symptoms are observed depending on the patient's age. Not all symptoms occur in all patients within one family despite the same genetic background. However, the importance of early therapy for the clinical course of the disease requires further study.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/diagnóstico por imagem , Erros Inatos do Metabolismo dos Carboidratos/genética , Epilepsia Resistente a Medicamentos/diagnóstico por imagem , Epilepsia Resistente a Medicamentos/genética , Características da Família , Transportador de Glucose Tipo 1/genética , Proteínas de Transporte de Monossacarídeos/deficiência , Adulto , Idoso , Encéfalo/diagnóstico por imagem , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Criança , Pré-Escolar , Dieta Cetogênica/métodos , Epilepsia Resistente a Medicamentos/dietoterapia , Feminino , Humanos , Lactente , Masculino , Proteínas de Transporte de Monossacarídeos/genética , Mutação/genética , Linhagem
7.
Epilepsy Behav ; 91: 90-93, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30076047

RESUMO

The glucose transporter type 1 (Glut1) is the most important energy carrier of the brain across the blood-brain barrier. In the early nineties, the first genetic defect of Glut1 was described and known as the Glut1 deficiency syndrome (Glut1-DS). It is characterized by early infantile seizures, developmental delay, microcephaly, and ataxia. Recently, milder variants have also been described. The clinical picture of Glut1 defects and the understanding of the pathophysiology of this disease have significantly grown. A special form of transient movement disorders, the paroxysmal exertion-induced dyskinesia (PED), absence epilepsies particularly with an early onset absence epilepsy (EOAE) and childhood absence epilepsy (CAE), myoclonic astatic epilepsy (MAE), episodic choreoathetosis and spasticity (CSE), and focal epilepsy can be based on a Glut1 defect. Despite the rarity of these diseases, the Glut1 syndromes are of high clinical interest since a very effective therapy, the ketogenic diet, can improve or reverse symptoms especially if it is started as early as possible. The present article summarizes the clinical features of Glut1 syndromes and discusses the underlying genetic mutations, including the available data on functional tests as well as the genotype-phenotype correlations. This article is part of the Special Issue "Individualized Epilepsy Management: Medicines, Surgery and Beyond".


Assuntos
Epilepsia/genética , Transportador de Glucose Tipo 1/genética , Transtornos dos Movimentos/genética , Mutação/genética , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/genética , Dieta Cetogênica/métodos , Distúrbios Distônicos/diagnóstico , Distúrbios Distônicos/dietoterapia , Distúrbios Distônicos/genética , Epilepsias Mioclônicas/diagnóstico , Epilepsias Mioclônicas/dietoterapia , Epilepsias Mioclônicas/genética , Epilepsias Parciais/diagnóstico , Epilepsias Parciais/dietoterapia , Epilepsias Parciais/genética , Epilepsia/diagnóstico , Epilepsia/dietoterapia , Epilepsia Tipo Ausência/diagnóstico , Epilepsia Tipo Ausência/dietoterapia , Epilepsia Tipo Ausência/genética , Humanos , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/genética , Transtornos dos Movimentos/diagnóstico , Transtornos dos Movimentos/dietoterapia
8.
Mol Genet Metab ; 123(3): 309-316, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29269105

RESUMO

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


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Suplementos Nutricionais , Glicina/administração & dosagem , Microcefalia/metabolismo , Fosfatidilcolinas/metabolismo , Fosfoglicerato Desidrogenase/deficiência , Transtornos Psicomotores/metabolismo , Convulsões/metabolismo , Serina/biossíntese , Transaminases/deficiência , Erros Inatos do Metabolismo dos Carboidratos/sangue , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Diferenciação Celular , Criança , Pré-Escolar , Feminino , Glicina/sangue , Humanos , Lactente , Masculino , Metabolômica/métodos , Microcefalia/sangue , Microcefalia/dietoterapia , Neurônios/metabolismo , Fosfoglicerato Desidrogenase/sangue , Fosfoglicerato Desidrogenase/metabolismo , Transtornos Psicomotores/sangue , Transtornos Psicomotores/dietoterapia , Convulsões/sangue , Convulsões/dietoterapia , Serina/administração & dosagem , Serina/sangue , Transaminases/sangue , Transaminases/metabolismo
9.
Can J Neurol Sci ; 45(1): 93-96, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29144225

RESUMO

Between July of 2012 and December of 2014, 39 patients were enrolled prospectively to investigate the prevalence of glucose transporter 1 (GLUT1) deficiency in a ketogenic diet clinic. None of them had GLUT1 deficiency. All patients seen in the same clinic within the same period were reviewed retrospectively. A total of 18 of these 85 patients had a genetic diagnosis, including GLUT1 deficiency, pathogenic copy number variants, congenital disorder of glycosylation, neuronal ceroid lipofuscinosis type II, mitochondrial disorders, tuberous sclerosis, lissencephaly, and SCN1A-, SCN8A-, and STXBP1-associated epileptic encephalopathies. The prevalence of genetic diagnoses was 21% and prevalence of GLUT1 deficiency was 2.4% in our retrospective cohort study.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/complicações , Erros Inatos do Metabolismo dos Carboidratos/genética , Dieta Cetogênica/métodos , Epilepsia/complicações , Proteínas de Transporte de Monossacarídeos/deficiência , Adolescente , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/epidemiologia , Criança , Pré-Escolar , Estudos de Coortes , Epilepsia/dietoterapia , Epilepsia/epidemiologia , Epilepsia/genética , Feminino , Humanos , Masculino , Proteínas de Transporte de Monossacarídeos/genética , Prevalência
10.
Epilepsy Behav ; 70(Pt A): 1-4, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28407523

RESUMO

INTRODUCTION: Glucose transporter type 1 deficiency syndrome (GLUT1-DS) is a rare metabolic encephalopathy with a wide variation of clinical phenotypes. Familial variants are often milder than de novo cases, and may therefore remain undiagnosed. The aim of this study was to characterize the clinical course of GLUT1-DS in a four-generation Norwegian family where the oldest generations had never received any treatment. METHOD: Through interviews and clinical investigations, we characterized a family of 26 members, where 11 members had symptoms strongly suggesting GLUT1-DS. All members were offered genetic testing of the SLC2A1 gene. Affected members were offered treatment with ketogenic diet, and the effect of the treatment was registered. RESULTS: We sequenced the SLC2A1 gene in 13 members, and found that 10, all with symptoms, had the c.823G>A (p.Ala275Thr) variant. All affected members had experienced early-onset epilepsy, paroxysmal exercise-induced dyskinesias, and most had mild learning disability. Moreover, some had symptoms and signs of a distal neuropathy in addition to reduced sense of orientation and excessive daytime sleep. Their load of symptoms had decreased over the years, although that they never had received any treatment. Nevertheless, those who started dietary treatment all experienced an improved quality of life. CONCLUSION: We report a four-generation family with GLUT1-DS where the disease has a mild course, even when untreated. In addition to classical GLUT1-DS features, we also describe symptoms which have never been reported in GLUT1-DS previously. As such, this family extends the phenotypic spectrum of GLUT1-DS and underlines the importance of diagnosing also relatively mildly affected patients, even in adult life, as they also seem to benefit from dietary treatment.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/genética , Proteínas de Transporte de Monossacarídeos/deficiência , Fenótipo , Adulto , Idoso , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Criança , Pré-Escolar , Coreia/dietoterapia , Coreia/genética , Dieta Cetogênica/métodos , Epilepsia/diagnóstico , Epilepsia/dietoterapia , Epilepsia/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Transporte de Monossacarídeos/genética , Mutação/genética , Noruega , Linhagem , Qualidade de Vida
11.
J Perinat Med ; 45(3): 375-382, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27564695

RESUMO

BACKGROUND: Published data on breast milk feeding in infants suffering from inherited metabolic disorders (IMDs) other than phenylketonuria (PKU) are limited and described outcome is variable. OBJECTIVE: We aimed to evaluate retrospectively whether breastfeeding and/or breast milk feeding are feasible in infants with IMDs including organic acidemias, fatty acid oxidation disorders, urea cycle disorders, aminoacidopathies or disorders of galactose metabolism. METHODS: Data on breastfeeding and breast milk feeding as well as monitoring and neurological outcome were collected retrospectively from our database of patients with the mentioned IMD, who were followed in our metabolic center within the last 10 years. RESULTS: Twenty patients were included in the study, who were either breast fed on demand or received expressed breast milk. All the infants were evaluated clinically and biochemically at 2-4-week intervals, with weight gain as the leading parameter to determine metabolic control. Good metabolic control and adequate neurological development were achieved in all patients but one, who experienced the only metabolic crisis observed within the study period. CONCLUSION: Breast milk feeding with close clinical and biochemical monitoring is feasible in most IMD and should be considered as it offers nutritional and immunological benefits.


Assuntos
Aleitamento Materno , Erros Inatos do Metabolismo/dietoterapia , Leite Humano , Erros Inatos do Metabolismo dos Aminoácidos/dietoterapia , Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Erros Inatos do Metabolismo Lipídico/dietoterapia , Erros Inatos do Metabolismo Lipídico/metabolismo , Masculino , Erros Inatos do Metabolismo/metabolismo , Estudos Retrospectivos , Distúrbios Congênitos do Ciclo da Ureia/dietoterapia , Distúrbios Congênitos do Ciclo da Ureia/metabolismo , Aumento de Peso
12.
Neuropediatrics ; 47(2): 132-6, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26902182

RESUMO

High-fat ketogenic diets are the only treatment available for Glut1 deficiency (Glut1D). Here, we describe an 8-year-old girl with classical Glut1D responsive to a 3:1 ketogenic diet and ethosuximide. After 3 years on the diet a gradual increase of blood lipids was followed by rapid, severe asymptomatic hypertriglyceridemia (1,910 mg/dL). Serum lipid apheresis was required to determine liver, renal, and pancreatic function. A combination of medium chain triglyceride-oil and a reduction of the ketogenic diet to 1:1 ratio normalized triglyceride levels within days but triggered severe myoclonic seizures requiring comedication with sultiam. Severe hypertriglyceridemia in children with Glut1D on ketogenic diets may be underdiagnosed and harmful. In contrast to congenital hypertriglyceridemias, children with Glut1D may be treated effectively by dietary adjustments alone.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/complicações , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Dieta Cetogênica/efeitos adversos , Hipertrigliceridemia/complicações , Hipertrigliceridemia/diagnóstico , Proteínas de Transporte de Monossacarídeos/deficiência , Criança , Feminino , Transportador de Glucose Tipo 1/deficiência , Humanos
13.
Dev Med Child Neurol ; 58(11): 1193-1199, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27273526

RESUMO

AIM: Glucose transporter type 1 deficiency syndrome (GLUT1-DS) results from impaired glucose transport into the brain, and is treated with a ketogenic diet. A few reports have suggested effectiveness of treatment using the modified Atkins diet (MAD). We aimed to assess the efficacy of MAD as a treatment for GLUT1-DS. METHOD: We evaluated the efficacy of MAD in 10 patients (four males, six females; mean age at diagnosis [SD] 6.2y [1.7], min-max: 4mo-12y) with GLUT1-DS. RESULTS: MAD was started at diagnosis in eight patients, including two infants. The mean duration (SD) under MAD was 2.5 [0.6] years (range 6mo-6y). Seven patients with epilepsy started MAD at GLUT1-DS diagnosis, and all experienced improvements in their epilepsy: five out of seven were seizure-free at M1, and three out of six at M3 and M6. The initiation of MAD allowed symptoms to be controlled in the three patients with movement disorders but without seizures. Two patients switched from the ketogenic diet to MAD. This switch was not responsible for the recurrence of any symptoms, and led to improvements in both physical abilities and growth parameters. INTERPRETATION: MAD, which is a less restrictive and more palatable diet than the ketogenic diet, seems to have comparable effectiveness. Moreover, a switch from the ketogenic diet to MAD appears to be beneficial for patients with GLUT1-DS.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Dieta com Restrição de Carboidratos/métodos , Epilepsia/dietoterapia , Proteínas de Transporte de Monossacarídeos/deficiência , Transtornos dos Movimentos/dietoterapia , Avaliação de Resultados em Cuidados de Saúde , Erros Inatos do Metabolismo dos Carboidratos/complicações , Criança , Pré-Escolar , Dieta Cetogênica/métodos , Epilepsia/etiologia , Feminino , França , Humanos , Lactente , Masculino , Transtornos dos Movimentos/etiologia , Estudos Retrospectivos
14.
Eur J Pediatr ; 175(10): 1267-76, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27586246

RESUMO

UNLABELLED: Ketogenic diet is an established and effective non-pharmacologic treatment for drug-resistant epilepsy. Ketogenic diet represents the treatment of choice for GLUT-1 deficiency syndrome and pyruvate dehydrogenase complex deficiency. Infantile spasms, Dravet syndrome and myoclonic-astatic epilepsy are epilepsy syndromes for which ketogenic diet should be considered early in the therapeutic pathway. Recently, clinical indications for ketogenic diet have been increasing, as there is emerging evidence regarding safety and effectiveness. Specifically, ketogenic diet response has been investigated in refractory status epilepticus and encephalopathy with status epilepticus during sleep. New targets in neuropharmacology, such as mitochondrial permeability transition, are being studied and might lead to using it effectively in other neurological diseases. But, inefficient connectivity and impaired ketogenic diet proposal limit ideal availability of this therapeutic option. Ketogenic diet in Italy is not yet considered as standard of care, not even as a therapeutic option for many child neurologists and epileptologists. CONCLUSIONS: The aim of this review is to revisit ketogenic diet effectiveness and safety in order to highlight its importance in drug-resistant epilepsy and other neurological disorders. WHAT IS KNOWN: • Ketogenic diet efficacy is now described in large case series, with adequate diet compliance and side effects control. • Ketogenic diet is far from being attempted as a first line therapy. Its availability varies worldwide. What is New: • New pharmacological targets such as mitochondrial permeability transition and new epileptic syndromes and etiologies responding to the diet such as refractory status epilepticus are being pointed out. • Ketogenic diet can function at its best when used as a tailor-made therapy. Fine tuning is crucial.


Assuntos
Dieta Cetogênica/métodos , Epilepsia Resistente a Medicamentos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Criança , Dieta Cetogênica/efeitos adversos , Dieta Cetogênica/economia , Humanos , Proteínas de Transporte de Monossacarídeos/deficiência , Neoplasias/dietoterapia , Doença da Deficiência do Complexo de Piruvato Desidrogenase/dietoterapia , Convulsões/dietoterapia
15.
BMC Gastroenterol ; 15: 90, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26215149

RESUMO

BACKGROUND: Congenital lactase deficiency is an extremely rare gastrointestinal disorder characterized by neonatal-onset watery diarrhoea and failure to thrive. We present the first genetically confirmed case of congenital lactase deficiency in Central Europe. CASE PRESENTATION: After an uneventful pregnancy and birth, a male newborn of consanguineous parents of Turkish origin presented with watery diarrhoea. On day 17, he was admitted to hospital with weight loss, hypertonic dehydration, and metabolic acidosis. Additionally, the patient showed an elevated calcium concentration in blood and urine as well as nephrocalcinosis. Diarrhoea stopped during intravenous rehydration and when feeding a glucose-, galactose-, and lactose-free formula. Therefore, glucose-galactose-malabsorption was assumed. However, genetic testing of the SGLT1 (SLC5A1) gene was negative and, indeed, feeding maltodextrine did not result in recurrence of diarrhoea. In contrast, lactose feeding immediately caused watery diarrhoea, suggesting congenital lactase deficiency. Genetic testing of the LCT gene revealed homozygosity for a 1-bp deletion in exon 8 (c.3448delT). Because of the nature of the mutation, causing a frame shift and a premature termination of translation, congenital lactase deficiency was confirmed and intestinal biopsies were unnecessary. The patient's general condition improved substantially on a lactose-free diet, including hypercalcaemia, hypercalciuria, and nephrocalcinosis which, however, only disappeared after months. CONCLUSION: This case demonstrates (a) that congenital lactase deficiency should be considered in cases of severe neonatal diarrhoea, (b) that intestinal biopsies can be avoided in typical cases that are confirmed by genetic testing, and


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/genética , Mutação da Fase de Leitura , Lactase/deficiência , Lactase/genética , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Diarreia/etiologia , Homozigoto , Humanos , Recém-Nascido , Masculino , Turquia
16.
Neuropediatrics ; 46(5): 313-20, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26267703

RESUMO

OBJECTIVES: The purpose of this study was to characterize patients who were diagnosed with glucose transporter protein 1 deficiency syndrome (Glut1D), and also to assess the efficacy of ketogenic diet (KD) therapy on seizure control, cognitive functions, and other neurological disorders. PATIENTS AND METHODS: We studied six unrelated patients with the classical phenotype of Glut1D, focusing on clinical and laboratory features, the KD therapy and outcome over the 25-month follow-up period. RESULTS: Five patients became seizure-free with the onset of ketosis, and anticonvulsants were discontinued. Other neurological features such as ataxia, spasticity, and dystonia showed a less striking improvement than seizure control. There was no significant change in the intelligence quotient (IQ) level or microcephaly. In all patients, alertness, concentration, motivation, and activity resulted in a moderate improvement of variable degree. The early-onset adverse effects of KD were observed in five patients. The KD regimen failed in one patient, therefore, his diet was changed with an alternative to KD. CONCLUSIONS: Treatment with KD resulted in a marked improvement in seizures and cognitive functions but its effect appeared to be less striking on the other neurological disorders of the patients. When the classic KD is not tolerated, an alternative to KD may be helpful.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/complicações , Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Cognição/fisiologia , Dieta Cetogênica , Proteínas de Transporte de Monossacarídeos/deficiência , Convulsões/dietoterapia , Adolescente , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Glicemia , Erros Inatos do Metabolismo dos Carboidratos/genética , Erros Inatos do Metabolismo dos Carboidratos/psicologia , Criança , Pré-Escolar , Feminino , Seguimentos , Glucose/líquido cefalorraquidiano , Transportador de Glucose Tipo 1/genética , Humanos , Ácido Láctico/líquido cefalorraquidiano , Masculino , Proteínas de Transporte de Monossacarídeos/genética , Mutação de Sentido Incorreto , Testes Neuropsicológicos , Fenótipo , Convulsões/etiologia , Resultado do Tratamento
17.
Dev Med Child Neurol ; 57(10): 969-76, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25914049

RESUMO

AIM: We aimed to determine whether response to ketogenic dietary therapies (KDT) was due to undiagnosed glucose transporter type 1 deficiency syndrome (GLUT1-DS). METHOD: Targeted resequencing of the SLC2A1 gene was completed in individuals without previously known GLUT1-DS who received KDT for their epilepsy. Hospital records were used to obtain demographic and clinical data. Response to KDT at various follow-up points was defined as seizure reduction of at least 50%. Seizure freedom achieved at any follow-up point was also documented. Fisher's exact and gene-burden association tests were conducted using the PLINK/SEQ open-source genetics library. RESULTS: Of the 246 participants, one was shown to have a novel variant in SLC2A1 that was predicted to be deleterious. This individual was seizure-free on KDT. Rates of seizure freedom in cases without GLUT1-DS were below 8% at each follow-up point. Two cases without SLC2A1 mutations were seizure-free at every follow-up point recorded. No significant results were obtained from Fisher's exact or gene-burden association tests. INTERPRETATION: A favourable response to KDT is not solely explained by mutations in SLC2A1. Other genetic factors should be sought to identify those who are most likely to benefit from dietary treatment for epilepsy, particularly those who may achieve seizure freedom.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Erros Inatos do Metabolismo dos Carboidratos/genética , Dieta Cetogênica , Epilepsia/dietoterapia , Epilepsia/genética , Transportador de Glucose Tipo 1/genética , Proteínas de Transporte de Monossacarídeos/deficiência , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Erros Inatos do Metabolismo dos Carboidratos/fisiopatologia , Criança , Pré-Escolar , Epilepsia/fisiopatologia , Feminino , Seguimentos , Técnicas de Genotipagem , Humanos , Masculino , Proteínas de Transporte de Monossacarídeos/genética , Convulsões/dietoterapia , Convulsões/genética , Convulsões/fisiopatologia , Resultado do Tratamento
18.
Epilepsia ; 55 Suppl 3: 37-40, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25209085

RESUMO

The transition of adolescents with refractory epilepsy to the care of adult neurologists can be challenging. For those patients with epilepsy due to mitochondrial disorders, Lafora disease, Unverricht-Lundborg disease, and GLUT1 deficiency syndrome, a successful transition can be even more problematic for both caregivers and neurologists. Many of these patients require dietary treatments (ketogenic and modified Atkins diets) for long-term management of their epilepsy. For these patients, coordinating transfer of their dietary management is necessary.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Dieta Cetogênica , Epilepsia/dietoterapia , Doenças Mitocondriais/dietoterapia , Proteínas de Transporte de Monossacarídeos/deficiência , Transição para Assistência do Adulto , Adolescente , Adulto , Erros Inatos do Metabolismo dos Carboidratos/diagnóstico , Criança , Epilepsia/diagnóstico , Humanos , Doenças Mitocondriais/diagnóstico , Doenças Mitocondriais/metabolismo , Resultado do Tratamento
19.
Neuropediatrics ; 45(2): 117-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23888468

RESUMO

This is a report on the successful treatment of a 6-year-old girl with genetically proven glucose transporter type 1 deficiency syndrome (GLUT1-DS) with modified Atkins diet (MAD). GLUT1-DS is an inborn disorder of glucose transport across the blood-brain barrier, which leads to energy deficiency of the brain with a broad spectrum of neurological symptoms including therapy-resistant epilepsy. Usually classical ketogenic diet (KD) is the standard treatment for patients with GLUT1-DS. Treatment with MAD, a variant of KD, for an observation period of 17 months resulted in improvement of seizures, alertness, cognitive abilities, and electroencephalography in this patient.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Dieta com Restrição de Carboidratos , Proteínas de Transporte de Monossacarídeos/deficiência , Criança , Dieta Cetogênica , Feminino , Humanos
20.
Epilepsy Behav ; 39: 111-5, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25240122

RESUMO

The aim of this study was to investigate the effects of ketogenic diet (KD) on cognitive function in patients with glucose transporter protein 1 deficiency syndrome (GLUT1-DS). Six patients with GLUT1-DS who were referred to the National Centre for Epilepsy in Norway during the period of November 2011-September 2013 were included. They were diagnosed with GLUT1-DS on the basis of early-onset seizures and developmental delay (with or without movement disorders or microcephaly) in addition to CSF-to-blood glucose ratio below 0.5. They were all treated with either classical KD or modified Atkins diet (MAD). The effect of the diet with >90% reduction in the seizure frequency was, in retrospect, considered as a support for the diagnosis. The patients underwent standardized neuropsychological assessment before the diet was initiated, and they were reassessed after a minimum of six months on the diet. The neuropsychological tests were individually selected for each patient in order to match their cognitive level. The main finding was a considerable improvement in several aspects of neuropsychological functioning after 6-17 months of dietary treatment in all the six patients. The greatest progress was seen in the youngest children. Our findings suggest that early diagnosis and dietary treatment are important in order to prevent developmental delay. However, also adults with GLUT1-DS may profit from dietary treatment by improving alertness, setting the stage for enhanced learning capacity, as well as physical endurance and quality of life.


Assuntos
Erros Inatos do Metabolismo dos Carboidratos/dietoterapia , Transtornos Cognitivos/dietoterapia , Dieta Cetogênica/métodos , Epilepsia/dietoterapia , Proteínas de Transporte de Monossacarídeos/deficiência , Adulto , Erros Inatos do Metabolismo dos Carboidratos/complicações , Criança , Pré-Escolar , Transtornos Cognitivos/etiologia , Epilepsia/etiologia , Feminino , Seguimentos , Humanos , Lactente , Masculino , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA