Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Cerebellum ; 2023 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-38123901

RESUMEN

Cells configure their metabolism in a synchronized and timely manner to meet their energy demands throughout development and adulthood. Transitions of developmental stages are coupled to metabolic shifts, such that glycolysis is highly active during cell proliferation, whereas oxidative phosphorylation prevails in postmitotic states. In the cerebellum, metabolic transitions are remarkable given its protracted developmental timelines. Such distinctive feature, along with its high neuronal density and metabolic demands, make the cerebellum highly vulnerable to metabolic insults. Despite the expansion of metabolomic approaches to uncover biological mechanisms, little is known about the role of metabolism on cerebellar development and maintenance. To illuminate the intricate connections between metabolism, physiology, and cerebellar disorders, we examined here the impact of metabolism on cerebellar growth, maturation, and adulthood through the lens of inborn errors of metabolism.

2.
Sci Transl Med ; 14(665): eabn2956, 2022 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-36197967

RESUMEN

Individuals with glucose transporter type I deficiency (G1D) habitually experience nutrient-responsive epilepsy associated with decreased brain glucose. However, the mechanistic association between blood glucose concentration and brain excitability in the context of G1D remains to be elucidated. Electroencephalography (EEG) in G1D individuals revealed nutrition time-dependent seizure oscillations often associated with preserved volition despite electrographic generalization and uniform average oscillation duration and periodicity, suggesting increased facilitation of an underlying neural loop circuit. Nonlinear EEG ictal source localization analysis and simultaneous EEG/functional magnetic resonance imaging converged on the thalamus-sensorimotor cortex as one potential circuit, and 18F-deoxyglucose positron emission tomography (18F-DG-PET) illustrated decreased glucose accumulation in this circuit. This pattern, reflected in a decreased thalamic to striatal 18F signal ratio, can aid with the PET imaging diagnosis of the disorder, whereas the absence of noticeable ictal behavioral changes challenges the postulated requirement for normal thalamocortical activity during consciousness. In G1D mice, 18F-DG-PET and mass spectrometry also revealed decreased brain glucose and glycogen, but preserved tricarboxylic acid cycle intermediates, indicating no overall energy metabolism failure. In brain slices from these animals, synaptic inhibition of cortical pyramidal neurons and thalamic relay neurons was decreased, and neuronal disinhibition was mitigated by metabolic sources of carbon; tonic-clonic seizures were also suppressed by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor inhibition. These results pose G1D as a thalamocortical synaptic disinhibition disease associated with increased glucose-dependent neuronal excitability, possibly in relation to reduced glycogen. Together with findings in other metabolic defects, inhibitory neuron dysfunction is emerging as a modulable mechanism of hyperexcitability.


Asunto(s)
Glucemia , Estado de Conciencia , Animales , Errores Innatos del Metabolismo de los Carbohidratos , Carbono/metabolismo , Desoxiglucosa , Electroencefalografía , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucógeno/metabolismo , Ratones , Proteínas de Transporte de Monosacáridos/deficiencia , Convulsiones , Tálamo/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico
3.
Sci Transl Med ; 11(480)2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787166

RESUMEN

Glucose is the ultimate substrate for most brain activities that use carbon, including synthesis of the neurotransmitters glutamate and γ-aminobutyric acid via mitochondrial tricarboxylic acid (TCA) cycle. Brain metabolism and neuronal excitability are thus interdependent. However, the principles that govern their relationship are not always intuitive because heritable defects of brain glucose metabolism are associated with the paradoxical coexistence, in the same individual, of episodic neuronal hyperexcitation (seizures) with reduced basal cerebral electrical activity. One such prototypic disorder is pyruvate dehydrogenase (PDH) deficiency (PDHD). PDH is central to metabolism because it steers most of the glucose-derived flux into the TCA cycle. To better understand the pathophysiology of PDHD, we generated mice with brain-specific reduced PDH activity that paralleled salient human disease features, including cerebral hypotrophy, decreased amplitude electroencephalogram (EEG), and epilepsy. The mice exhibited reductions in cerebral TCA cycle flux, glutamate content, spontaneous, and electrically evoked in vivo cortical field potentials and gamma EEG oscillation amplitude. Episodic decreases in gamma oscillations preceded most epileptiform discharges, facilitating their prediction. Fast-spiking neuron excitability was decreased in brain slices, contributing to in vivo action potential burst prolongation after whisker pad stimulation. These features were partially reversed after systemic administration of acetate, which augmented cerebral TCA cycle flux, glutamate-dependent synaptic transmission, inhibition and gamma oscillations, and reduced epileptiform discharge duration. Thus, our results suggest that dysfunctional excitability in PDHD is consequent to reduced oxidative flux, which leads to decreased neuronal activation and impaired inhibition, and can be mitigated by an alternative metabolic substrate.


Asunto(s)
Encéfalo/metabolismo , Neuronas/fisiología , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/metabolismo , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/fisiopatología , Acetatos/metabolismo , Algoritmos , Animales , Isótopos de Carbono , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Electroencefalografía , Potenciales Evocados , Ritmo Gamma , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Humanos , Aprendizaje Automático , Ratones , Inhibición Neural , Convulsiones/metabolismo , Convulsiones/fisiopatología , Vibrisas
4.
J Inherit Metab Dis ; 42(2): 220-236, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30734319

RESUMEN

Most of the energy produced in the brain is dedicated to supporting synaptic transmission. Glucose is the main fuel, providing energy and carbon skeletons to the cells that execute and support synaptic function: neurons and astrocytes, respectively. It is unclear, however, how glucose is provided to and used by these cells under different levels of synaptic activity. It is even more unclear how diseases that impair glucose uptake and oxidation in the brain alter metabolism in neurons and astrocytes, disrupt synaptic activity, and cause neurological dysfunction, of which seizures are one of the most common clinical manifestations. Poor mechanistic understanding of diseases involving synaptic energy metabolism has prevented the expansion of therapeutic options, which, in most cases, are limited to symptomatic treatments. To shed light on the intersections between metabolism, synaptic transmission, and neuronal excitability, we briefly review current knowledge of compartmentalized metabolism in neurons and astrocytes, the biochemical pathways that fuel synaptic transmission at resting and active states, and the mechanisms by which disorders of brain glucose metabolism disrupt neuronal excitability and synaptic function and cause neurological disease in the form of epilepsy.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético , Epilepsia/metabolismo , Neuronas/metabolismo , Transmisión Sináptica , Animales , Astrocitos/metabolismo , Glucosa/metabolismo , Humanos , Oxidación-Reducción , Convulsiones/metabolismo , Sinapsis/metabolismo
5.
J Med Genet ; 55(1): 48-54, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28626029

RESUMEN

BACKGROUND: Transport protein particle (TRAPP) is a multisubunit complex that regulates membrane trafficking through the Golgi apparatus. The clinical phenotype associated with mutations in various TRAPP subunits has allowed elucidation of their functions in specific tissues. The role of some subunits in human disease, however, has not been fully established, and their functions remain uncertain. OBJECTIVE: We aimed to expand the range of neurodevelopmental disorders associated with mutations in TRAPP subunits by exome sequencing of consanguineous families. METHODS: Linkage and homozygosity mapping and candidate gene analysis were used to identify homozygous mutations in families. Patient fibroblasts were used to study splicing defect and zebrafish to model the disease. RESULTS: We identified six individuals from three unrelated families with a founder homozygous splice mutation in TRAPPC6B, encoding a core subunit of the complex TRAPP I. Patients manifested a neurodevelopmental disorder characterised by microcephaly, epilepsy and autistic features, and showed splicing defect. Zebrafish trappc6b morphants replicated the human phenotype, displaying decreased head size and neuronal hyperexcitability, leading to a lower seizure threshold. CONCLUSION: This study provides clinical and functional evidence of the role of TRAPPC6B in brain development and function.


Asunto(s)
Trastorno Autístico/genética , Epilepsia/genética , Efecto Fundador , Estudios de Asociación Genética , Microcefalia/genética , Mutación/genética , Trastornos del Neurodesarrollo/genética , Proteínas de Transporte Vesicular/genética , Animales , Trastorno Autístico/complicaciones , Epilepsia/complicaciones , Homocigoto , Humanos , Microcefalia/complicaciones , Fenotipo , Pez Cebra
6.
FEBS Lett ; 591(21): 3548-3554, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28963851

RESUMEN

Malignant brain tumors are known to utilize acetate as an alternate carbon source in the citric acid cycle for their bioenergetics. 13 C NMR-based isotopomer analysis has been used to measure turnover of 13 C-acetate carbons into glutamate and glutamine pools in tumors. Plasma from the patients infused with [1,2-13 C]acetate further revealed the presence of 13 C isotopomers of glutamine, glucose, and lactate in the circulation that were generated due to metabolism of [1,2-13 C]acetate by peripheral organs. In the tumor cells, [4-13 C] and [3,4-13 C]glutamate and glutamine isotopomers were generated from blood-borne 13 C-labeled glucose and lactate which were formed due to [1,2-13 C[acetate metabolism of peripheral tissues. [4,5-13 C] and [3,4,5-13 C]glutamate and glutamine isotopomers were produced from [1,2-13 C]acetyl-CoA that was derived from direct oxidation of [1,2-13 C] acetate in the tumor. Major portion of C4 13 C fractional enrichment of glutamate (93.3 ± 0.02%) and glutamine (90.9 ± 0.03%) were derived from [1,2-13 C]acetate-derived acetyl-CoA.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Acetatos/administración & dosificación , Acetatos/farmacocinética , Neoplasias Encefálicas/diagnóstico por imagen , Isótopos de Carbono/farmacocinética , Femenino , Humanos , Masculino
7.
Am J Hum Genet ; 101(3): 441-450, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28823706

RESUMEN

Pontocerebellar hypoplasia (PCH) represents a group of recessive developmental disorders characterized by impaired growth of the pons and cerebellum, which frequently follows a degenerative course. Currently, there are 10 partially overlapping clinical subtypes and 13 genes known mutated in PCH. Here, we report biallelic TBC1D23 mutations in six individuals from four unrelated families manifesting a non-degenerative form of PCH. In addition to reduced volume of pons and cerebellum, affected individuals had microcephaly, psychomotor delay, and ataxia. In zebrafish, tbc1d23 morphants replicated the human phenotype showing hindbrain volume loss. TBC1D23 localized at the trans-Golgi and was regulated by the small GTPases Arl1 and Arl8, suggesting a role in trans-Golgi membrane trafficking. Altogether, this study provides a causative link between TBC1D23 mutations and PCH and suggests a less severe clinical course than other PCH subtypes.


Asunto(s)
Enfermedades Cerebelosas/genética , Proteínas Activadoras de GTPasa/genética , Homocigoto , Microcefalia/genética , Mutación , Adolescente , Animales , Enfermedades Cerebelosas/patología , Niño , Preescolar , Femenino , Células HeLa , Humanos , Masculino , Microcefalia/patología , Linaje , Fenotipo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
8.
J Med Genet ; 54(6): 399-403, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28280135

RESUMEN

BACKGROUND: Microcephaly with nephrotic syndrome is a rare co-occurrence, constituting the Galloway-Mowat syndrome (GAMOS), caused by mutations in WDR73 (OMIM: 616144). However, not all patients harbour demonstrable WDR73 deleterious variants, suggesting that there are other yet unidentified factors contributing to GAMOS aetiology. METHODS: Autozygosity mapping and candidate analysis was used to identify deleterious variants in consanguineous families. Analysis of patient fibroblasts was used to study splicing and alterations in cellular function. RESULTS: In two consanguineous families with five affected individuals from Turkey with a GAMOS-like presentation, we identified a shared homozygous variant leading to partial exon 4 skipping in nucleoporin, 107-KD (NUP107). The founder mutation was associated with concomitant reduction in NUP107 protein and in the obligate binding partner NUP133 protein, as well as density of nuclear pores in patient cells. CONCLUSION: Recently, NUP107 was suggested as a candidate in a family with nephrotic syndrome and developmental delay. Other NUP107-reported cases had isolated renal phenotypes. With the addition of these individuals, we implicate an allele-specific critical role for NUP107 in the regulation of brain growth and a GAMOS-like presentation.


Asunto(s)
Hernia Hiatal/genética , Microcefalia/genética , Mutación/genética , Nefrosis/genética , Síndrome Nefrótico/genética , Proteínas de Complejo Poro Nuclear/genética , Esteroides/metabolismo , Adolescente , Niño , Discapacidades del Desarrollo/genética , Femenino , Homocigoto , Humanos , Lactante , Riñón/metabolismo , Masculino , Linaje , Fenotipo , Proteínas/genética , Turquía
9.
Nat Genet ; 49(3): 457-464, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28092684

RESUMEN

Deadenylases are best known for degrading the poly(A) tail during mRNA decay. The deadenylase family has expanded throughout evolution and, in mammals, consists of 12 Mg2+-dependent 3'-end RNases with substrate specificity that is mostly unknown. Pontocerebellar hypoplasia type 7 (PCH7) is a unique recessive syndrome characterized by neurodegeneration and ambiguous genitalia. We studied 12 human families with PCH7, uncovering biallelic, loss-of-function mutations in TOE1, which encodes an unconventional deadenylase. toe1-morphant zebrafish displayed midbrain and hindbrain degeneration, modeling PCH-like structural defects in vivo. Surprisingly, we found that TOE1 associated with small nuclear RNAs (snRNAs) incompletely processed spliceosomal. These pre-snRNAs contained 3' genome-encoded tails often followed by post-transcriptionally added adenosines. Human cells with reduced levels of TOE1 accumulated 3'-end-extended pre-snRNAs, and the immunoisolated TOE1 complex was sufficient for 3'-end maturation of snRNAs. Our findings identify the cause of a neurodegenerative syndrome linked to snRNA maturation and uncover a key factor involved in the processing of snRNA 3' ends.


Asunto(s)
Enfermedades Cerebelosas/genética , Exonucleasas/genética , Mutación/genética , Proteínas Nucleares/genética , ARN Nuclear Pequeño/genética , Alelos , Animales , Femenino , Humanos , Masculino , Ratones , Enfermedades Neurodegenerativas/genética , ARN Mensajero/genética , Empalmosomas/genética , Pez Cebra
10.
Ann Neurol ; 80(1): 59-70, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27130255

RESUMEN

OBJECTIVE: A study was undertaken to characterize the clinical features of the newly described hypomyelinating leukodystrophy type 10 with microcephaly. This is an autosomal recessive disorder mapped to chromosome 1q42.12 due to mutations in the PYCR2 gene, encoding an enzyme involved in proline synthesis in mitochondria. METHODS: From several international clinics, 11 consanguineous families were identified with PYCR2 mutations by whole exome or targeted sequencing, with detailed clinical and radiological phenotyping. Selective mutations from patients were tested for effect on protein function. RESULTS: The characteristic clinical presentation of patients with PYCR2 mutations included failure to thrive, microcephaly, craniofacial dysmorphism, progressive psychomotor disability, hyperkinetic movements, and axial hypotonia with variable appendicular spasticity. Patients did not survive beyond the first decade of life. Brain magnetic resonance imaging showed global brain atrophy and white matter T2 hyperintensities. Routine serum metabolic profiles were unremarkable. Both nonsense and missense mutations were identified, which impaired protein multimerization. INTERPRETATION: PYCR2-related syndrome represents a clinically recognizable condition in which PYCR2 mutations lead to protein dysfunction, not detectable on routine biochemical assessments. Mutations predict a poor outcome, probably as a result of impaired mitochondrial function. Ann Neurol 2016;80:59-70.


Asunto(s)
Insuficiencia de Crecimiento/complicaciones , Insuficiencia de Crecimiento/genética , Microcefalia/complicaciones , Microcefalia/genética , Pirrolina Carboxilato Reductasas/genética , Adolescente , Niño , Preescolar , Codón sin Sentido , Exoma/genética , Femenino , Fibroblastos , Expresión Génica , Predisposición Genética a la Enfermedad/genética , Genotipo , Humanos , Lactante , Masculino , Microcefalia/diagnóstico , Mutación Missense , Fenotipo , Cultivo Primario de Células , Pirrolina Carboxilato Reductasas/biosíntesis , Síndrome , Transfección , Adulto Joven
11.
J Neurochem ; 132(1): 99-109, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25231025

RESUMEN

The (13) C-labeling patterns in glutamate and glutamine from brain tissue are quite different after infusion of a mixture of (13) C-enriched glucose and acetate. Two processes contribute to this observation, oxidation of acetate by astrocytes but not neurons, and preferential incorporation of α-ketoglutarate into glutamate in neurons, and incorporation of α-ketoglutarate into glutamine in astrocytes. The acetate:glucose ratio, introduced previously for analysis of a single (13) C NMR spectrum, provides a useful index of acetate and glucose oxidation in the brain tissue. However, quantitation of relative substrate oxidation at the cell compartment level has not been reported. A simple mathematical method is presented to quantify the ratio of acetate-to-glucose oxidation in astrocytes, based on the standard assumption that neurons do not oxidize acetate. Mice were infused with [1,2-(13) C]acetate and [1,6-(13) C]glucose, and proton decoupled (13) C NMR spectra of cortex extracts were acquired. A fit of those spectra to the model indicated that (13) C-labeled acetate and glucose contributed approximately equally to acetyl-CoA (0.96) in astrocytes. As this method relies on a single (13) C NMR spectrum, it can be readily applied to multiple physiologic and pathologic conditions. Differences in (13) C labeling of brain glutamate and glutamine have been attributed to metabolic compartmentation. The acetate:glucose ratio, introduced for description of a (13) C NMR (nuclear magnetic resonance) spectrum, is an index of glucose and acetate oxidation in brain tissue. A simple mathematical method is presented to quantify the ratio of acetate-to-glucose oxidation in astrocytes from a single NMR spectrum. As kinetic analysis is not required, the method is readily applicable to analysis of tissue extracts. α-KG = alpha-ketoglutarate; CAC = citric acid cycle; GLN = glutamine; GLU = glutamate.


Asunto(s)
Acetatos/metabolismo , Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Glucosa/metabolismo , Acetilcoenzima A/metabolismo , Animales , Astrocitos/química , Corteza Cerebral/química , Corteza Cerebral/citología , Femenino , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Ácidos Cetoglutáricos/metabolismo , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Neuronas/química , Neuronas/metabolismo , Oxidación-Reducción
12.
J Neurochem ; 132(3): 301-12, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25314677

RESUMEN

The mechanistic link of ketosis to neuroprotection under certain pathological conditions continues to be explored. We investigated whether chronic ketosis induced by ketogenic diet results in the partitioning of ketone bodies toward oxidative metabolism in brain. We hypothesized that diet-induced ketosis results in increased shunting of ketone bodies toward citric acid cycle and amino acids with decreased carbon shunting from glucose. Rats were fed standard (STD) or ketogenic (KG) diets for 3.5 weeks and then infused with [U-(13) C]glucose or [U-(13) C]acetoacetate tracers. Concentrations and (13) C-labeling pattern of citric acid cycle intermediates and amino acids were analyzed from brain homogenates using stable isotopomer mass spectrometry analysis. The contribution of [U-(13) C]glucose to acetyl-CoA and amino acids decreased by ~ 30% in the KG group versus STD, whereas [U-(13) C]acetoacetate contributions were more than two-fold higher. The concentration of GABA remained constant across groups; however, the (13) C labeling of GABA was markedly increased in the KG group infused with [U-(13) C]acetoacetate compared to STD. This study reveals that there is a significant contribution of ketone bodies to oxidative metabolism and GABA in diet-induced ketosis. We propose that this represents a fundamental mechanism of neuroprotection under pathological conditions.


Asunto(s)
Química Encefálica , Carbono/metabolismo , Dieta Cetogénica , Glucosa/metabolismo , Acidemia Propiónica/metabolismo , Acetoacetatos/metabolismo , Animales , Metabolismo Energético , Cuerpos Cetónicos/metabolismo , Masculino , Oxidación-Reducción , Ratas , Ratas Wistar , Ácido gamma-Aminobutírico/biosíntesis
13.
JAMA Neurol ; 71(10): 1255-65, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25110966

RESUMEN

IMPORTANCE: Disorders of brain metabolism are multiform in their mechanisms and manifestations, many of which remain insufficiently understood and are thus similarly treated. Glucose transporter type I deficiency (G1D) is commonly associated with seizures and with electrographic spike-waves. The G1D syndrome has long been attributed to energy (ie, adenosine triphosphate synthetic) failure such as that consequent to tricarboxylic acid (TCA) cycle intermediate depletion. Indeed, glucose and other substrates generate TCAs via anaplerosis. However, TCAs are preserved in murine G1D, rendering energy-failure inferences premature and suggesting a different hypothesis, also grounded on our work, that consumption of alternate TCA precursors is stimulated and may be detrimental. Second, common ketogenic diets lead to a therapeutically counterintuitive reduction in blood glucose available to the G1D brain and prove ineffective in one-third of patients. OBJECTIVE: To identify the most helpful outcomes for treatment evaluation and to uphold (rather than diminish) blood glucose concentration and stimulate the TCA cycle, including anaplerosis, in G1D using the medium-chain, food-grade triglyceride triheptanoin. DESIGN, SETTING, AND PARTICIPANTS: Unsponsored, open-label cases series conducted in an academic setting. Fourteen children and adults with G1D who were not receiving a ketogenic diet were selected on a first-come, first-enrolled basis. INTERVENTION: Supplementation of the regular diet with food-grade triheptanoin. MAIN OUTCOMES AND MEASURES: First, we show that, regardless of electroencephalographic spike-waves, most seizures are rarely visible, such that perceptions by patients or others are inadequate for treatment evaluation. Thus, we used quantitative electroencephalographic, neuropsychological, blood analytical, and magnetic resonance imaging cerebral metabolic rate measurements. RESULTS: One participant (7%) did not manifest spike-waves; however, spike-waves promptly decreased by 70% (P = .001) in the other participants after consumption of triheptanoin. In addition, the neuropsychological performance and cerebral metabolic rate increased in most patients. Eleven patients (78%) had no adverse effects after prolonged use of triheptanoin. Three patients (21%) experienced gastrointestinal symptoms, and 1 (7%) discontinued the use of triheptanoin. CONCLUSIONS AND RELEVANCE: Triheptanoin can favorably influence cardinal aspects of neural function in G1D. In addition, our outcome measures constitute an important framework for the evaluation of therapies for encephalopathies associated with impaired intermediary metabolism.


Asunto(s)
Glucemia/metabolismo , Encéfalo/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/tratamiento farmacológico , Ciclo del Ácido Cítrico , Suplementos Dietéticos , Proteínas de Transporte de Monosacáridos/deficiencia , Triglicéridos/uso terapéutico , Adolescente , Adulto , Encéfalo/fisiopatología , Errores Innatos del Metabolismo de los Carbohidratos/metabolismo , Niño , Preescolar , Estudios de Cohortes , Electroencefalografía , Femenino , Glucosa/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Proteínas de Transporte de Monosacáridos/metabolismo , Resultado del Tratamiento , Adulto Joven
14.
Pediatr Neurol ; 51(1): 100-3, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24938142

RESUMEN

INTRODUCTION: We present a developmentally appropriate adolescent boy who presented with upper and lower extremity glove-and-stocking paresthesias, distal weakness, vertigo, high-pitched voice, inattention, ataxia, and binocular diplopia after a voluntary 59-kg weight loss over 5 months. CLINICAL INVESTIGATIONS: Extensive investigations revealed serum thiamine levels <2 nmol/L. Brain magnetic resonance imaging revealed symmetric abnormal T2 prolongation of the mammillary bodies. Nerve conduction studies were consistent with axonal, length-dependent polyneuropathy. Together, these findings were diagnostic for peripheral polyneuropathy and Wernicke encephalopathy secondary to thiamine deficiency. CONCLUSION: This patient illustrates that eating disorders can be an uncommon cause of rapidly progressive paresthesias, weakness, and neurological decline due to thiamine deficiency.


Asunto(s)
Anorexia Nerviosa/complicaciones , Enfermedades del Sistema Nervioso Periférico/etiología , Deficiencia de Tiamina/complicaciones , Encefalopatía de Wernicke/etiología , Adolescente , Extremidades/fisiopatología , Humanos , Imagen por Resonancia Magnética , Masculino , Tubérculos Mamilares/patología , Tiamina/sangre
15.
Epilepsia ; 55(7): 970-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24861491

RESUMEN

Focal cortical dysplasias (FCDs) constitute a prevalent cause of intractable epilepsy in children, and is one of the leading conditions requiring epilepsy surgery. Despite recent advances in the cellular and molecular biology of these conditions, the pathogenetic mechanisms of FCDs remain largely unknown. The purpose if this work is to review the molecular underpinnings of FCDs and to highlight potential therapeutic targets. A systematic review of the literature regarding the histologic, molecular, and electrophysiologic aspects of FCDs was conducted. Disruption of the mammalian target of rapamycin (mTOR) signaling comprises a common pathway underlying the structural and electrical disturbances of some FCDs. Other mechanisms such as viral infections, prematurity, head trauma, and brain tumors are also posited. mTOR inhibitors (i.e., rapamycin) have shown positive results on seizure management in animal models and in a small cohort of patients with FCD. Encouraging progress has been achieved on the molecular and electrophysiologic basis of constitutive cells in the dysplastic tissue. Despite the promising results of mTOR inhibitors, large-scale randomized trials are in need to evaluate their efficacy and side effects, along with additional mechanistic studies for the development of novel, molecular-based diagnostic and therapeutic approaches.


Asunto(s)
Epilepsia/epidemiología , Epilepsia/genética , Malformaciones del Desarrollo Cortical/epidemiología , Malformaciones del Desarrollo Cortical/genética , Transducción de Señal/genética , Animales , Anticonvulsivantes/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Epilepsia/tratamiento farmacológico , Marcación de Gen/métodos , Humanos , Malformaciones del Desarrollo Cortical/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética
16.
Pediatr Neurol ; 49(1): 54-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23827428

RESUMEN

Attention-deficit/hyperactivity disorder is the most common neurobehavioral disorder in children and frequently associated with epilepsy. For patients with both conditions, methylphenidate remains a mainstay in the treatment of behavioral problems. Most studies demonstrate that methylphenidate is effective in treating children with well-controlled epilepsy, and that methylphenidate does not increase the risk of having seizures in patients with EEG abnormalities without epilepsy. However, in patients with active seizures, the results are somewhat contradictory. This article presents the case of a young girl with attention-deficit/hyperactivity disorder and behavioral problems on Depakote (valproic acid) who had an abnormal EEG with left centroparietal spikes but no history of electrographic seizures. She experienced a convulsion the day after her first dose of methylphenidate, and repeat EEG demonstrated continuous spike and slow wave during sleep. This case report suggests that children with continuous spike and slow wave during sleep may have a higher risk of developing seizures with methylphenidate treatment.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Estimulantes del Sistema Nervioso Central/efectos adversos , Metilfenidato/efectos adversos , Convulsiones/inducido químicamente , Convulsiones/fisiopatología , Sueño/fisiología , Trastorno por Déficit de Atención con Hiperactividad/diagnóstico , Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Niño , Femenino , Humanos , Convulsiones/diagnóstico , Sueño/efectos de los fármacos
17.
J Cereb Blood Flow Metab ; 33(8): 1160-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23652627

RESUMEN

Two variants of a widely used two-compartment model were prepared for fitting the time course of [1,6-(13)C2]glucose metabolism in rat brain. Features common to most models were included, but in one model the enrichment of the substrates entering the glia and neuronal citric acid cycles was allowed to differ. Furthermore, the models included the capacity to analyze multiplets arising from (13)C spin-spin coupling, known to improve parameter estimates in heart. Data analyzed were from a literature report providing time courses of [1,6-(13)C2]glucose metabolism. Four analyses were used, two comparing the effect of different pyruvate enrichment in glia and neurons, and two for determining the effect of multiplets present in the data. When fit independently, the enrichment in glial pyruvate was less than in neurons. In the absence of multiplets, fit quality and parameter values were typical of those in the literature, whereas the multiplet curves were not modeled well. This prompted the use of robust statistical analysis (the Kolmogorov-Smirnov test of goodness of fit) to determine whether individual curves were modeled appropriately. At least 50% of the curves in each experiment were considered poorly fit. It was concluded that the model does not include all metabolic features required to analyze the data.


Asunto(s)
Química Encefálica/fisiología , Ácido Pirúvico/metabolismo , Animales , Radioisótopos de Carbono , Ciclo del Ácido Cítrico/fisiología , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Espectroscopía de Resonancia Magnética , Modelos Estadísticos , Neuroglía/metabolismo , Neuronas/metabolismo , Ratas
18.
J Cereb Blood Flow Metab ; 33(2): 175-82, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23072752

RESUMEN

It has been postulated that triheptanoin can ameliorate seizures by supplying the tricarboxylic acid cycle with both acetyl-CoA for energy production and propionyl-CoA to replenish cycle intermediates. These potential effects may also be important in other disorders associated with impaired glucose metabolism because glucose supplies, in addition to acetyl-CoA, pyruvate, which fulfills biosynthetic demands via carboxylation. In patients with glucose transporter type I deficiency (G1D), ketogenic diet fat (a source only of acetyl-CoA) reduces seizures, but other symptoms persist, providing the motivation for studying heptanoate metabolism. In this work, metabolism of infused [5,6,7-(13)C(3)]heptanoate was examined in the normal mouse brain and in G1D by (13)C-nuclear magnetic resonance spectroscopy, gas chromatography-mass spectrometry (GC-MS), and liquid chromatography-mass spectrometry (LC-MS). In both groups, plasma glucose was enriched in (13)C, confirming gluconeogenesis from heptanoate. Acetyl-CoA and glutamine levels became significantly higher in the brain of G1D mice relative to normal mice. In addition, brain glutamine concentration and (13)C enrichment were also greater when compared with glutamate in both animal groups, suggesting that heptanoate and/or C5 ketones are primarily metabolized by glia. These results enlighten the mechanism of heptanoate metabolism in the normal and glucose-deficient brain and encourage further studies to elucidate its potential antiepileptic effects in disorders of energy metabolism.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético , Transportador de Glucosa de Tipo 1 , Glucosa/metabolismo , Glutamina/metabolismo , Heptanoatos/metabolismo , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Animales , Anticonvulsivantes/farmacología , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Glucosa/genética , Glutamina/genética , Heptanoatos/farmacología , Espectroscopía de Resonancia Magnética , Ratones , Ratones Transgénicos , Ácido Pirúvico/metabolismo , Convulsiones/tratamiento farmacológico , Convulsiones/genética , Convulsiones/metabolismo , Triglicéridos/farmacología
19.
Neurochem Int ; 61(7): 1036-43, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22884585

RESUMEN

The pyruvate dehydrogenase complex (PDC), required for complete glucose oxidation, is essential for brain development. Although PDC deficiency is associated with a severe clinical syndrome, little is known about its effects on either substrate oxidation or synthesis of key metabolites such as glutamate and glutamine. Computational simulations of brain metabolism indicated that a 25% reduction in flux through PDC and a corresponding increase in flux from an alternative source of acetyl-CoA would substantially alter the (13)C NMR spectrum obtained from brain tissue. Therefore, we evaluated metabolism of [1,6-(13)C(2)]glucose (oxidized by both neurons and glia) and [1,2-(13)C(2)]acetate (an energy source that bypasses PDC) in the cerebral cortex of adult mice mildly and selectively deficient in brain PDC activity, a viable model that recapitulates the human disorder. Intravenous infusions were performed in conscious mice and extracts of brain tissue were studied by (13)C NMR. We hypothesized that mice deficient in PDC must increase the proportion of energy derived from acetate metabolism in the brain. Unexpectedly, the distribution of (13)C in glutamate and glutamine, a measure of the relative flux of acetate and glucose into the citric acid cycle, was not altered. The (13)C labeling pattern in glutamate differed significantly from glutamine, indicating preferential oxidation of [1,2-(13)C]acetate relative to [1,6-(13)C]glucose by a readily discernible metabolic domain of the brain of both normal and mutant mice, presumably glia. These findings illustrate that metabolic compartmentation is preserved in the PDC-deficient cerebral cortex, probably reflecting intact neuron-glia metabolic interactions, and that a reduction in brain PDC activity sufficient to induce cerebral dysgenesis during development does not appreciably disrupt energy metabolism in the mature brain.


Asunto(s)
Corteza Cerebral/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/metabolismo , Animales , Isótopos de Carbono , Femenino , Masculino , Ratones , Ratones Transgénicos
20.
Cell Metab ; 15(6): 827-37, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22682223

RESUMEN

Dysregulated metabolism is a hallmark of cancer cell lines, but little is known about the fate of glucose and other nutrients in tumors growing in their native microenvironment. To study tumor metabolism in vivo, we used an orthotopic mouse model of primary human glioblastoma (GBM). We infused (13)C-labeled nutrients into mice bearing three independent GBM lines, each with a distinct set of mutations. All three lines displayed glycolysis, as expected for aggressive tumors. They also displayed unexpected metabolic complexity, oxidizing glucose via pyruvate dehydrogenase and the citric acid cycle, and using glucose to supply anaplerosis and other biosynthetic activities. Comparing the tumors to surrounding brain revealed obvious metabolic differences, notably the accumulation of a large glutamine pool within the tumors. Many of these same activities were conserved in cells cultured ex vivo from the tumors. Thus GBM cells utilize mitochondrial glucose oxidation during aggressive tumor growth in vivo.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glucosa/metabolismo , Mitocondrias/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioblastoma/genética , Glioblastoma/patología , Gluconeogénesis , Glutamato-Amoníaco Ligasa/metabolismo , Ácido Glutámico/metabolismo , Glutaminasa/metabolismo , Glutamina/metabolismo , Glucólisis , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Neostriado/metabolismo , Neostriado/patología , Oxidación-Reducción , Fenotipo , Piruvato Carboxilasa/metabolismo , Estadísticas no Paramétricas , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...