Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurosci ; 39(25): 4847-4863, 2019 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-30926746

RESUMEN

The glutamate transporter GLT-1 is highly expressed in astrocytes but also in neurons, primarily in axon terminals. We generated a conditional neuronal GLT-1 KO using synapsin 1-Cre (synGLT-1 KO) to elucidate the metabolic functions of GLT-1 expressed in neurons, here focusing on the cerebral cortex. Both synaptosomal uptake studies and electron microscopic immunocytochemistry demonstrated knockdown of GLT-1 in the cerebral cortex in the synGLT-1 KO mice. Aspartate content was significantly reduced in cerebral cortical extracts as well as synaptosomes from cerebral cortex of synGLT-1 KO compared with control littermates. 13C-Labeling of tricarboxylic acid cycle intermediates originating from metabolism of [U-13C]-glutamate was significantly reduced in synGLT-1 KO synaptosomes. The decreased aspartate content was due to diminished entry of glutamate into the tricarboxylic acid cycle. Pyruvate recycling, a pathway necessary for full glutamate oxidation, was also decreased. ATP production was significantly increased, despite unaltered oxygen consumption, in isolated mitochondria from the synGLT-1 KO. The density of mitochondria in axon terminals and perisynaptic astrocytes was increased in the synGLT-1 KO. Intramitochondrial cristae density of synGLT-1 KO mice was increased, suggesting increased mitochondrial efficiency, perhaps in compensation for reduced access to glutamate. SynGLT-1 KO synaptosomes exhibited an elevated oxygen consumption rate when stimulated with veratridine, despite a lower baseline oxygen consumption rate in the presence of glucose. GLT-1 expressed in neurons appears to be required to provide glutamate to synaptic mitochondria and is linked to neuronal energy metabolism and mitochondrial function.SIGNIFICANCE STATEMENT All synaptic transmitters need to be cleared from the extracellular space after release, and transporters are used to clear glutamate released from excitatory synapses. GLT-1 is the major glutamate transporter, and most GLT-1 is expressed in astrocytes. Only 5%-10% is expressed in neurons, primarily in axon terminals. The function of GLT-1 in axon terminals remains unknown. Here, we used a conditional KO approach to investigate the significance of the expression of GLT-1 in neurons. We found multiple abnormalities of mitochondrial function, suggesting impairment of glutamate utilization by synaptic mitochondria in the neuronal GLT-1 KO. These data suggest that GLT-1 expressed in axon terminals may be important in maintaining energy metabolism and biosynthetic activities mediated by presynaptic mitochondria.


Asunto(s)
Transportador 2 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Homeostasis/fisiología , Mitocondrias/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Animales , Ácido Aspártico/metabolismo , Corteza Cerebral/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Consumo de Oxígeno/fisiología , Terminales Presinápticos/metabolismo , Sinapsis/genética , Sinaptosomas/metabolismo
2.
J Neurosci Res ; 97(8): 890-896, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30801795

RESUMEN

The first two publications dealing with the question of the cellular localization of the enzyme pyruvate carboxylase (PC) which in the brain represents the most important metabolic pathway to allow anaplerosis of TCA cycle constituents were published in 1983 and 1985. Hence, 2018 marks the 35th anniversary of the notion based on the results of the publications provided above that PC-catalyzed anaplerosis in the brain is an astrocytic process. This review will provide the background for investigating this enzymatic pathway as well as a discussion of cataplerosis, the degradation of products from anaplerosis, and the current status of the functional significance of pyruvate carboxylation in brain metabolism.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Piruvato Carboxilasa/metabolismo , Ácido Pirúvico/metabolismo , Animales , Ciclo del Ácido Cítrico , Humanos , Neuronas/metabolismo
3.
Neurochem Res ; 42(3): 827-837, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27394419

RESUMEN

Oligodendroglial cells are known to de-acetylate the N-acetylaspartate (NAA) synthesized and released by neurons and use it for lipid synthesis. However, the role of NAA regarding their intermediary metabolism remains poorly understood. Two hypotheses were proposed regarding the fate of aspartate after being released by de-acetylation: (1) aspartate is metabolized in the mitochondria of oligodendrocyte lineage cells; (2) aspartate is released to the medium. We report here that aspartoacylase mRNA expression increases when primary rat oligodendrocyte progenitor cells (OPCs) differentiate into mature cells in culture. Moreover, characterising metabolic functions of acetyl coenzyme A and aspartate from NAA catabolism in mature oligodendrocyte cultures after 5 days using isotope-labelled glucose after 5-days of differentiation we found evidence of extensive NAA metabolism. Incubation with [1,6-13C]glucose followed by gas chromatography-mass spectrometry and high performance liquid chromatography analyses of cell extracts and media in the presence and absence of NAA established that the acetate moiety produced by hydrolysis of NAA does not enter mitochondrial metabolism in the form of acetyl coenzyme A. We also resolved the controversy concerning the possible release of aspartate to the medium: aspartate is not released to the medium by oligodendrocytes in amounts detectable by our methods. Therefore we propose that: aspartate released from NAA joins the cytosolic aspartate pool rapidly and takes part in the malate-aspartate shuttle, which transports reducing equivalents from glycolysis into the mitochondria for ATP production and enters the tricarboxylic acid cycle at a slow rate.


Asunto(s)
Ácido Aspártico/análogos & derivados , Oligodendroglía/metabolismo , Acetilcoenzima A/metabolismo , Acetilación , Animales , Animales Recién Nacidos , Ácido Aspártico/metabolismo , Células Cultivadas , Glucosa/metabolismo , Hidrólisis , Ácido Láctico/metabolismo , Mitocondrias/metabolismo , Cultivo Primario de Células , Prosencéfalo/citología , Prosencéfalo/metabolismo , Ratas Sprague-Dawley
4.
Neurochem Res ; 42(1): 115-132, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28019006

RESUMEN

Neonatal hypoxia-ischemia (HI) and the delayed injury cascade that follows involve excitotoxicity, oxidative stress and mitochondrial failure. The susceptibility to excitotoxicity of the neonatal brain may be related to the capacity of astrocytes for glutamate uptake. Furthermore, the neonatal brain is vulnerable to oxidative stress, and the pentose phosphate pathway (PPP) may be of particular importance for limiting this kind of injury. Also, in the neonatal brain, neurons depend upon de novo synthesis of neurotransmitters via pyruvate carboxylase in astrocytes to increase neurotransmitter pools during normal brain development. Several recent publications describing intermediary brain metabolism following neonatal HI have yielded interesting results: (1) Following HI there is a prolonged depression of mitochondrial metabolism in agreement with emerging evidence of mitochondria as vulnerable targets in the delayed injury cascade. (2) Astrocytes, like neurons, are metabolically impaired following HI, and the degree of astrocytic malfunction may be an indicator of the outcome following hypoxic and hypoxic-ischemic brain injury. (3) Glutamate transfer from neurons to astrocytes is not increased following neonatal HI, which may imply that astrocytes fail to upregulate glutamate uptake in response to the massive glutamate release during HI, thus contributing to excitotoxicity. (4) In the neonatal brain, the activity of the PPP is reduced following HI, which may add to the susceptibility of the neonatal brain to oxidative stress. The present review aims to discuss the metabolic temporal alterations observed in the neonatal brain following HI.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Glucosa/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Humanos , Ratas
5.
Neurochem Res ; 42(1): 244-253, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27068034

RESUMEN

Proliferation and differentiation of neural stem cells (NSCs) have a crucial role to ensure neurogenesis and gliogenesis in the mammalian brain throughout life. As there is growing evidence for the significance of metabolism in regulating cell fate, knowledge on the metabolic programs in NSCs and how they evolve during differentiation into somatic cells may provide novel therapeutic approaches to address brain diseases. In this work, we applied a quantitative analysis to assess how the central carbon metabolism evolves upon differentiation of NSCs into astrocytes. Murine embryonic stem cell (mESC)-derived NSCs and astrocytes were incubated with labelled [1-13C]glucose and the label incorporation into intracellular metabolites was followed by GC-MS. The obtained 13C labelling patterns, together with uptake/secretion rates determined from supernatant analysis, were integrated into an isotopic non-stationary metabolic flux analysis (13C-MFA) model to estimate intracellular flux maps. Significant metabolic differences between NSCs and astrocytes were identified, with a general downregulation of central carbon metabolism during astrocytic differentiation. While glucose uptake was 1.7-fold higher in NSCs (on a per cell basis), a high lactate-secreting phenotype was common to both cell types. Furthermore, NSCs consumed glutamine from the medium; the highly active reductive carboxylation of alpha-ketoglutarate indicates that this was converted to citrate and used for biosynthetic purposes. In astrocytes, pyruvate entered the TCA cycle mostly through pyruvate carboxylase (81%). This pathway supported glutamine and citrate secretion, recapitulating well described metabolic features of these cells in vivo. Overall, this fluxomics study allowed us to quantify the metabolic rewiring accompanying astrocytic lineage specification from NSCs.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/fisiología , Análisis de Flujos Metabólicos/métodos , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Línea Celular , Supervivencia Celular/fisiología , Células Cultivadas , Ratones
6.
Glia ; 64(1): 21-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26352325

RESUMEN

Although oligodendrocytes constitute a significant proportion of cells in the central nervous system (CNS), little is known about their intermediary metabolism. We have, therefore, characterized metabolic functions of primary oligodendrocyte precursor cell cultures at late stages of differentiation using isotope-labelled metabolites. We report that differentiated oligodendrocyte lineage cells avidly metabolize glucose in the cytosol and pyruvate derived from glucose in the mitochondria. The labelling patterns of metabolites obtained after incubation with [1,2-(13)C]glucose demonstrated that the pentose phosphate pathway (PPP) is highly active in oligodendrocytes (approximately 10% of glucose is metabolized via the PPP as indicated by labelling patterns in phosphoenolpyruvate). Mass spectrometry and magnetic resonance spectroscopy analyses of metabolites after incubation of cells with [1-(13)C]lactate or [1,2-(13)C]glucose, respectively, demonstrated that anaplerotic pyruvate carboxylation, which was thought to be exclusive to astrocytes, is also active in oligodendrocytes. Using [1,2-(13)C]acetate, we show that oligodendrocytes convert acetate into acetyl CoA which is metabolized in the tricarboxylic acid cycle. Analysis of labelling patterns of alanine after incubation of cells with [1,2-(13)C]acetate and [1,2-(13)C]glucose showed catabolic oxidation of malate or oxaloacetate. In conclusion, we report that oligodendrocyte lineage cells at late differentiation stages are metabolically highly active cells that are likely to contribute considerably to the metabolic activity of the CNS.


Asunto(s)
Glucosa/metabolismo , Oligodendroglía/metabolismo , Acetatos/metabolismo , Acetilcoenzima A/metabolismo , Animales , Radioisótopos de Carbono , Células Cultivadas , Ciclo del Ácido Cítrico/fisiología , Citosol/metabolismo , Ácido Láctico/metabolismo , Malatos/metabolismo , Mitocondrias/metabolismo , Células-Madre Neurales/metabolismo , Ácido Oxaloacético/metabolismo , Vía de Pentosa Fosfato/fisiología , Fosfoenolpiruvato/metabolismo , Ácido Pirúvico/metabolismo , Radiofármacos , Ratas Sprague-Dawley
7.
Glia ; 64(5): 695-715, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26689134

RESUMEN

Availability of homogeneous astrocyte populations would facilitate research concerning cell plasticity (metabolic and transcriptional adaptations; innate immune responses) and cell cycle reactivation. Current protocols to prepare astrocyte cultures differ in their final content of immature precursor cells, preactivated cells or entirely different cell types. A new method taking care of all these issues would improve research on astrocyte functions. We found here that the exposure of a defined population of pluripotent stem cell-derived neural stem cells (NSC) to BMP4 results in pure, nonproliferating astrocyte cultures within 24-48 h. These murine astrocytes generated from embryonic stem cells (mAGES) expressed the positive markers GFAP, aquaporin 4 and GLT-1, supported neuronal function, and acquired innate immune functions such as the response to tumor necrosis factor and interleukin 1. The protocol was applicable to several normal or disease-prone pluripotent cell lines, and the corresponding mAGES all exited the cell cycle and lost most of their nestin expression, in contrast to astrocytes generated by serum-addition or obtained as primary cultures. Comparative gene expression analysis of mAGES and NSC allowed quantification of differences between the two cell types and a definition of an improved marker set to define astrocytes. Inclusion of several published data sets in this transcriptome comparison revealed the similarity of mAGES with cortical astrocytes in vivo. Metabolic analysis of homogeneous NSC and astrocyte populations revealed distinct neurochemical features: both cell types synthesized glutamine and citrate, but only mature astrocytes released these metabolites. Thus, the homogeneous cultures allowed an improved definition of NSC and astrocyte features.


Asunto(s)
Astrocitos/fisiología , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Células-Madre Neurales/fisiología , Animales , Acuaporina 4/genética , Acuaporina 4/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Células Cultivadas , Citocinas/metabolismo , Citocinas/farmacología , Embrión de Mamíferos , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Antígeno Ki-67/metabolismo , Ácido Láctico/metabolismo , Ratones , Ratones Endogámicos BALB C , Nestina/metabolismo , Factores de Tiempo , Transcriptoma/fisiología
8.
J Neurochem ; 136(2): 329-38, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26442479

RESUMEN

Glutamine (Gln) is synthesized in astrocytes from glutamate (Glu) and ammonia, whereupon it can be released to be transferred to neurons. This study evaluated the as yet not definitely established role of the astrocytic Gln transporters SN1 and SN2 (Slc38a3 and Slc38a5 respectively) in Gln release and metabolic fluxes of glucose and acetate, the canonical precursors of Glu. Cultured neocortical astrocytes were grown in the absence or presence of ammonia (5 mM NH4 Cl, 24 h), which deregulates astrocytic metabolism in hyperammonemic encephalopathies. HPLC analyses of cell extracts of SN1/SN2 siRNA-treated (SN1/SN2-) astrocytes revealed a ~ 3.5-fold increase in Gln content and doubling of glutathione, aspartate, alanine and glutamate contents, as compared to SN1/SN2+ astrocytes. Uptake and efflux of preloaded [(3) H]Gln was likewise significantly decreased in SN1/SN2- astrocytes. The atom percent excess (13) C values (given as M + 1) for alanine, aspartate and glutamate were decreased when the SN1/SN2- cells were incubated with [1-(13) C] glucose, while Gln consumption was not changed. No difference was seen in M + 1 values in SN1/SN2- cells incubated with [2-(13) C] acetate, which were not treated with ammonia. In SN1/SN2- astrocytes, the increase in Gln content and the decrease in radiolabeled Gln release upon exposure to ammonia were found abrogated, and glutamate labeling from [2-(13) C]acetate was decreased as compared to SN1/SN2+ astrocytes. The results underscore a profound role of SN1 and/or SN2 in Gln release from astrocytes under physiological conditions, but less so in ammonia-overexposed astrocytes, and appear to manifest dependence of astrocytic glucose metabolism to Glu/Gln on unimpaired SN1/SN2- mediated Gln release from astrocytes. The astrocytic N system transporters SN1 and SN2 show preponderance to mediate glutamine (Gln) efflux. Under hyperammonemic conditions, accumulation of Gln, a direct product of ammonia detoxification, may deregulate astrocytic metabolism and seems to be responsible for astrocytic swelling. This study evaluated not definitely established role of SN1 and SN2 in Gln release and metabolic fluxes of radiolabeled glucose and acetate. Simultaneous silencing of SN1/SN2 transporters increase Gln, glutathione, aspartate, alanine and glutamate contents (Panel B; marked in red) as compare to non-silenced astrocytes (Panel A). The atom percent excess (13) C values (given as M + 1) for alanine, aspartate and glutamate were decreased when the cells with silenced transporters were incubated with [1-(13) C]glucose, whereas no difference was seen in M + 1 values when those cells were incubated with [2-(13) C]acetate. Ammonia abrogated the increase in Gln content and decrease in radiolabeled Gln release in astrocytes with silenced transporters, but caused a decrease in glutamate labeling from [2-(13) C]acetate.


Asunto(s)
Acetatos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Amoníaco/farmacología , Astrocitos/efectos de los fármacos , Corteza Cerebral/citología , Glucosa/metabolismo , Glutamina/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Isótopos de Carbono/metabolismo , Células Cultivadas , Cromatografía Líquida de Alta Presión , Cromatografía de Gases y Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Estadísticas no Paramétricas , Factores de Tiempo , Tritio/metabolismo
9.
J Neurochem ; 138(3): 423-35, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27128201

RESUMEN

The process of cell differentiation goes hand-in-hand with metabolic adaptations, which are needed to provide energy and new metabolites. Carbon monoxide (CO) is an endogenous cytoprotective molecule able to inhibit cell death and improve mitochondrial metabolism. Neuronal differentiation processes were studied using the NT2 cell line, which is derived from human testicular embryonic teratocarcinoma and differentiates into post-mitotic neurons upon retinoic acid treatment. CO-releasing molecule A1 (CORM-A1) was used do deliver CO into cell culture. CO treatment improved NT2 neuronal differentiation and yield, since there were more neurons and the total cell number increased following the differentiation process. CO supplementation enhanced the mitochondrial population in post-mitotic neurons derived from NT2 cells, as indicated by an increase in mitochondrial DNA. CO treatment during neuronal differentiation increased the extent of the classical metabolic change that occurs during neuronal differentiation, from glycolytic to more oxidative metabolism, by decreasing the ratio of lactate production and glucose consumption. The expression of pyruvate and lactate dehydrogenases was higher, indicating an augmented oxidative metabolism. Moreover, these findings were corroborated by an increased percentage of (13) C incorporation from [U-(13) C]glucose into the tricarboxylic acid cycle metabolites malate and citrate, and also glutamate and aspartate in CO-treated cells. Finally, under low levels of oxygen (5%), which enhances glycolytic metabolism, some of the enhancing effects of CO on mitochondria were not observed. In conclusion, our data show that CO improves neuronal and mitochondrial yield by stimulation of tricarboxylic acid cycle activity, and thus oxidative metabolism of NT2 cells during the process of neuronal differentiation. The process of cell differentiation is coupled with metabolic adaptations. Carbon monoxide (CO) is an endogenous cytoprotective gasotransmitter able to prevent cell death and improve mitochondrial metabolism. Herein CO supplementation improved neuronal differentiation yield, by enhancing mitochondrial population and promoting the classical metabolic change that occurs during neuronal differentiation, from glycolytic to oxidative metabolism.


Asunto(s)
Monóxido de Carbono/farmacología , Diferenciación Celular/efectos de los fármacos , ADN Mitocondrial/metabolismo , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Monóxido de Carbono/metabolismo , Línea Celular , Ciclo del Ácido Cítrico/efectos de los fármacos , Humanos , Mitocondrias/metabolismo , Neuronas/metabolismo , Tretinoina/farmacología
10.
J Neurochem ; 136(2): 339-50, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26526584

RESUMEN

Mitochondrial impairment is a key feature underlying neonatal hypoxic-ischemic (HI) brain injury and melatonin is potentially neuroprotective through its effects on mitochondria. In this study, we have used (1) H and (13) C NMR spectroscopy after injection of [1-(13) C]glucose and [1,2-(13) C]acetate to examine neuronal and astrocytic metabolism in the early reperfusion phase after unilateral HI brain injury in 7-day-old rat pups, exploring the effects of HI on mitochondrial function and the potential protective effects of melatonin on brain metabolism. One hour after hypoxia-ischemia, astrocytic metabolism was recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was clearly impaired. Pyruvate carboxylation was also lower in both hemispheres after HI. The transfer of glutamate from neurons to astrocytes was higher whereas the transfer of glutamine from astrocytes to neurons was lower 1 h after HI in the contralateral hemisphere. Neuronal metabolism was equally affected in pups treated with melatonin (10 mg/kg) immediately after HI as in vehicle treated pups indicating that the given dose of melatonin was not capable of protecting the neuronal mitochondria in this early phase after HI brain injury. However, any beneficial effects of melatonin might have been masked by modulatory effects of the solvent dimethyl sulfoxide on cerebral metabolism. Neuronal and astrocytic metabolism was examined by (13) C and (1) H NMR spectroscopy in the early reperfusion phase after unilateral hypoxic-ischemic brain injury and melatonin treatment in neonatal rats. One hour after hypoxia-ischemia astrocytic mitochondrial metabolism had recovered and glycolysis was normalized, whereas mitochondrial metabolism in neurons was impaired. Melatonin treatment did not show a protective effect on neuronal metabolism.


Asunto(s)
Antioxidantes/uso terapéutico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/terapia , Isquemia Encefálica/complicaciones , Melatonina/uso terapéutico , Reperfusión , Acetatos/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Lesiones Encefálicas/patología , Modelos Animales de Enfermedad , Femenino , Lateralidad Funcional/efectos de los fármacos , Glucosa/metabolismo , Isótopos/metabolismo , Espectroscopía de Resonancia Magnética , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuronas/metabolismo , Embarazo , Piruvato Carboxilasa/metabolismo , Ratas
11.
J Neurosci Res ; 94(12): 1561-1571, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27629247

RESUMEN

The cellular distribution of transporters and enzymes related to glutamate metabolism led to the concept of the glutamate-glutamine cycle. Glutamate is released as a neurotransmitter and taken up primarily by astrocytes ensheathing the synapses. The glutamate carbon skeleton is transferred back to the presynaptic neurons as the nonexcitatory amino acid glutamine. The cycle was initially thought to function with a 1:1 ratio between glutamate released and glutamine taken up by neurons. However, studies of glutamate metabolism in astrocytes have shown that a considerable proportion of glutamate undergoes oxidative degradation; thus, quantitative formation of glutamine from the glutamate taken up is not possible. Oxidation of glutamate is initiated by transamination catalyzed by an aminotransferase, or oxidative deamination catalyzed by glutamate dehydrogenase (GDH). We discuss methods available to elucidate the enzymes that mediate this conversion. Methods include pharmacological tools such as the transaminase inhibitor aminooxyacetic acid, studies using GDH knockout mice, and siRNA-mediated knockdown of GDH in astrocytes. Studies in brain slices incubated with [15 N]glutamate demonstrated activity of GDH in astrocytes in situ. These results, in conjunction with reports in the literature, support the conclusion that GDH is active in astrocytes both in culture and in vivo and that this enzyme plays a significant role in glutamate oxidation. Oxidative metabolism of glutamate, primarily mediated by GDH, but also by transamination by aspartate aminotransferase, provides considerably more energy than is required to maintain the activity of the high-affinity glutamate transporters needed for efficient removal of glutamate from the synaptic cleft. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Astrocitos/metabolismo , Glutamato Deshidrogenasa/metabolismo , Glutamatos/metabolismo , Transaminasas/metabolismo , Animales , Astrocitos/enzimología , Humanos , Oxidación-Reducción
12.
Neurochem Res ; 41(1-2): 86-95, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26433381

RESUMEN

Epilepsy is a severe neurological disorder characterized by altered electrical activity in the brain. Important pathophysiological mechanisms include disturbed metabolism and homeostasis of major excitatory and inhibitory neurotransmitters, glutamate and GABA. Current drug treatments are largely aimed at decreasing neuronal excitability and thereby preventing the occurrence of seizures. However, many patients are refractory to treatment and side effects are frequent. Temporal lobe epilepsy (TLE) is the most common type of drug-resistant epilepsy in adults. In rodents, the pilocarpine-status epilepticus model reflects the pathology and chronic spontaneous seizures of TLE and the pentylenetetrazole kindling model exhibits chronic induced limbic seizures. Accumulating evidence from studies on TLE points to alterations in astrocytes and neurons as key metabolic changes. The present review describes interventions which alleviate these disturbances in astrocyte-neuronal interactions by supporting mitochondrial metabolism. The compounds discussed are the endogenous transport molecule acetyl-L-carnitine and the triglyceride of heptanoate, triheptanoin. Both provide acetyl moieties for oxidation in the tricarboxylic acid cycle whereas heptanoate is also provides propionyl-CoA, which after carboxylation can produce succinyl-CoA, resulting in anaplerosis-the refilling of the tricarboxylic acid cycle.


Asunto(s)
Acetilcarnitina/uso terapéutico , Anticonvulsivantes/uso terapéutico , Astrocitos/metabolismo , Epilepsia/tratamiento farmacológico , Triglicéridos/uso terapéutico , Aminoácidos/metabolismo , Animales , Epilepsia/metabolismo , Humanos , Ratones , Neurotransmisores/metabolismo
13.
J Neurochem ; 132(5): 532-545, 2015 03.
Artículo en Inglés | MEDLINE | ID: mdl-25345404

RESUMEN

As reported previously, in the lithium-pilocarpine model of temporal lobe epilepsy (TLE), carisbamate (CRS) produces strong neuroprotection, leads to milder absence-like seizures, and prevents behavioral impairments in a subpopulation of rats. To understand the metabolic basis of these effects, here we injected 90 mg/kg CRS or vehicle twice daily for 7 days starting 1 h after status epilepticus (SE) induction in rats. Two months later, we injected [1-13 C]glucose and [1,2-13 C]acetate followed by head microwave fixation after 15 min. 13 C incorporation into metabolites was analyzed using 13 C magnetic resonance spectroscopy. We found that SE reduced neuronal mitochondrial metabolism in the absence but not in the presence of CRS. Reduction in glutamate level was prevented by CRS and aspartate levels were similar to controls only in rats displaying absence-like seizures after treatment [CRS-absence-like epilepsy (ALE)]. Glutamine levels in CRS-ALE rats were higher compared to controls in hippocampal formation and limbic structures while unchanged in rats displaying motor spontaneous recurrent seizures after treatment (CRS-TLE). Astrocytic mitochondrial metabolism was reduced in CRS-TLE, and either enhanced or unaffected in CRS-ALE rats, which did not affect the transfer of glutamine from astrocytes to neurons. In conclusion, CRS prevents reduction in neuronal mitochondrial metabolism but its effect on astrocytes is likely key in determining outcome of treatment in this model. To understand the metabolic basis of the strong neuroprotection and reduction in seizure severity caused by carisbamate (CRS) in the lithium-pilocarpine (Li-Pilo) model of temporal lobe epilepsy (TLE), we injected CRS for 7 days starting 1 h after status epilepticus and 2 months later [1-13 C]glucose and [1,2-13 C]acetate. 13 C Magnetic resonance spectroscopy analysis was performed on brain extracts and we found that CRS prevented reduction in neuronal mitochondrial metabolism but its effect on astrocytes was likely key in determining outcome of treatment in this model. ALE = absence like epilepsy; acetyl CoA = acetyl coenzyme A; GS = glutamine synthetase; PAG = phosphate activated glutaminase; PC = pyruvate carboxylase; OAA = oxaloacetate; TCA cycle = tricarboxylic acid cycle.

14.
J Neurosci Res ; 93(7): 1157-64, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25782696

RESUMEN

Pentylenetetrazol, kainic acid, or pilocarpine can be used to induce seizures in animal models of epilepsy. The present Review describes disturbances in astrocyte-neuron interactions in the acute, latent, and chronic phases analyzed by magnetic resonance spectroscopy of brain tissue extracts from rats injected with [1-(13)C]glucose and [1,2-(13)C]acetate. The most consistent change after onset of seizures was the decrease in (13)C labeling of glutamate (GLU) from [1-(13) C]glucose regardless of brain area, severity, or duration of the period with seizures and toxin used. In most cases this decrease was accompanied by a reduction in glutamine (GLN) labeling from [1-(13)C]glucose, presumably as a direct consequence of the reduction in labeling of GLU and the GLU-GLN cycle. Amounts of GLN were never changed. Reduction in the content of N-acetyl aspartate (NAA) was first detectable some time after status epilepticus but before the occurrence of spontaneous seizures. This decrease can be an indication of neuronal death and/or mitochondrial impairment and might indicate beginning gliosis. It is known that gliosis occurs in the chronic phase of temporal lobe epilepsy in hippocampus, but astrocyte metabolism appears normal in this phase, indicating that the gliotic astrocytes have a somewhat reduced metabolism per volume. A decrease in (13)C labeling of GLU from [1-(13)C]glucose is a very sensitive measure for the onset of epileptogenesis, whereas reduction of NAA is first detectable later. In the chronic phases of the hippocampal formation, astrocyte metabolism is upregulated given that the number of neurons is reduced.


Asunto(s)
Astrocitos/fisiología , Comunicación Celular/fisiología , Epilepsia/etiología , Neuronas/fisiología , Animales , Astrocitos/efectos de los fármacos , Convulsivantes/toxicidad , Modelos Animales de Enfermedad , Epilepsia/inducido químicamente , Humanos , Neuronas/efectos de los fármacos
15.
Neurochem Res ; 40(2): 402-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25380696

RESUMEN

The operation of a glutamine-glutamate/GABA cycle in the brain consisting of the transfer of glutamine from astrocytes to neurons and neurotransmitter glutamate or GABA from neurons to astrocytes is a well-known concept. In neurons, glutamine is not only used for energy production and protein synthesis, as in other cells, but is also an essential precursor for biosynthesis of amino acid neurotransmitters. An excellent tool for the study of glutamine transfer from astrocytes to neurons is [(14)C]acetate or [(13)C]acetate and the glial specific enzyme inhibitors, i.e. the glutamine synthetase inhibitor methionine sulfoximine and the tricarboxylic acid cycle (aconitase) inhibitors fluoro-acetate and -citrate. Acetate is metabolized exclusively by glial cells, and [(13)C]acetate is thus capable when used in combination with magnetic resonance spectroscopy or mass spectrometry, to provide information about glutamine transfer. The present review will give information about glutamine trafficking and the tools used to map it as exemplified by discussions of published work employing brain cell cultures as well as intact animals. It will be documented that considerably more glutamine is transferred from astrocytes to glutamatergic than to GABAergic neurons. However, glutamine does have an important role in GABAergic neurons despite their capability of re-utilizing their neurotransmitter by re-uptake.


Asunto(s)
Ácido Glutámico/metabolismo , Glutamina/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Ratones , Ácido gamma-Aminobutírico/biosíntesis
16.
Cephalalgia ; 35(11): 987-95, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25601916

RESUMEN

BACKGROUND: Preventive medication is indicated for many migraine patients, but is used in relatively few. The aim of the present study was to evaluate the efficacy of acetyl-l-carnitine as a prophylactic drug in migraine patients. METHODS: A single-center, randomized, triple-blind, placebo-controlled, crossover study was carried out. Men and women, age 18-65 years, with episodic migraine but otherwise healthy, were recruited mostly through advertisements. After a four-week run-in-phase, 72 participants were randomized to receive either placebo or 3 g acetyl-l-carnitine for 12 weeks. After a four-week washout, treatment was switched. The primary outcome was days with moderate or severe headache per four weeks. Secondary outcomes were days with headache, hours with headache, proportion of responders (>50% reduction in migraine days from baseline) and adverse events. RESULTS: In the complete case analyses, no statistically significant differences were found between acetyl-l-carnitine and placebo in severe or moderate headache days per month (3.0 versus 3.1, p = 0.80), headache days per month (5.1 versus 5.2, p = 0.73) or for the other secondary outcome measures. CONCLUSION: In this triple-blind crossover study no differences were found in headache outcomes between acetyl-l-carnitine and placebo. Our results do not provide evidence of benefit for efficacy of acetyl-l-carnitine as prophylactic treatment for migraine. TRIAL REGISTRATION: EUDRACT (2012-001624-36), ClinicalTrials.gov (NCT01695317).


Asunto(s)
Acetilcarnitina/uso terapéutico , Trastornos Migrañosos/prevención & control , Nootrópicos/uso terapéutico , Adolescente , Adulto , Anciano , Estudios Cruzados , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , Adulto Joven
17.
Stroke ; 45(9): 2777-85, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25052323

RESUMEN

BACKGROUND AND PURPOSE: Increased susceptibility to excitotoxicity of the neonatal brain after hypoxia-ischemia (HI) may be caused by limited capacity of astrocytes for glutamate uptake, and mitochondrial failure probably plays a key role in the delayed injury cascade. Male infants have poorer outcome than females after HI, possibly linked to differential intermediary metabolism. METHODS: [1-(13)C]glucose and [1,2-(13)C]acetate were injected at zero, 6, and 48 hours after unilateral HI in 7-day-old rats. Intermediary metabolism was analyzed with magnetic resonance spectroscopy. RESULTS: Mitochondrial metabolism was generally reduced in the ipsilateral hemisphere for ≤6 hours after HI, whereas contralaterally, it was reduced in neurons but not in astrocytes. Transfer of glutamate from neurons to astrocytes was increased in the contralateral, but not in the ipsilateral hemisphere at 0 hour, and reduced bilaterally at 6 hours after HI. The transfer of glutamine from astrocytes to glutamatergic neurons was unaltered in both hemispheres, whereas the transfer of glutamine to GABAergic neurons was increased ipsilaterally at 0 hour. Anaplerosis (astrocytes) was decreased, whereas partial pyruvate recycling (astrocytes) was increased directly after HI. Male pups had lower astrocytic mitochondrial metabolism than females immediately after HI, whereas that of females was reduced longer and encompassed both neurons and astrocytes. CONCLUSIONS: The prolonged depression in mitochondrial metabolism indicates that mitochondria are vulnerable targets in the delayed injury after neonatal HI. The degree of astrocytic malfunction may be a valid indicator of outcome after hypoxic/HI brain injury and may be linked to the differential outcome in males and females.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/crecimiento & desarrollo , Hipoxia-Isquemia Encefálica/patología , Neuronas/metabolismo , Acetatos/química , Animales , Encéfalo/metabolismo , Femenino , Neuronas GABAérgicas/metabolismo , Glucosa/química , Glucólisis , Espectroscopía de Resonancia Magnética , Masculino , Mitocondrias/metabolismo , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Factores de Tiempo , Resultado del Tratamiento
18.
J Neurochem ; 131(4): 399-406, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24989463

RESUMEN

The central process in energy production is the oxidation of acetyl-CoA to CO2 by the tricarboxylic acid (TCA, Krebs, citric acid) cycle. However, this cycle functions also as a biosynthetic pathway from which intermediates leave to be converted primarily to glutamate, GABA, glutamine and aspartate and to a smaller extent to glucose derivatives and fatty acids in the brain. When TCA cycle ketoacids are removed, they must be replaced to permit the continued function of this essential pathway, by a process termed anaplerosis. Since the TCA cycle cannot act as a carbon sink, anaplerosis must be coupled with cataplerosis; the exit of intermediates from the TCA cycle. The role of anaplerotic reactions for cellular metabolism in the brain has been studied extensively. However, the coupling of this process with cataplerosis and the roles that both pathways play in the regulation of amino acid, glucose, and fatty acid homeostasis have not been emphasized. The concept of a linkage between anaplerosis and cataplerosis should be underscored, because the balance between these two processes is essential. The hypothesis that cataplerosis in the brain is achieved by exporting the lactate generated from the TCA cycle intermediates into the blood and perivascular area is presented. This shifts the generally accepted paradigm of lactate generation as simply derived from glycolysis to that of oxidation and might present an alternative explanation for aerobic glycolysis. Intermediates leave the tricarboxylic acid cycle and must be replaced by a process termed anaplerosis that must be coupled to cataplerosis. We hypothesize that cataplerosis is achieved by exporting the lactate generated from the cycle into the blood and perivascular area. This shifts the paradigm of lactate generation as solely derived from glycolysis to that of oxidation and might present an alternative explanation for aerobic glycolysis.


Asunto(s)
Carbono/metabolismo , Ciclo del Ácido Cítrico/fisiología , Metabolismo Energético/fisiología , Glutamato Sintasa/metabolismo , Animales , Humanos , Oxidación-Reducción
19.
J Neurochem ; 128(5): 641-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24224925

RESUMEN

Glutamate is the major excitatory neurotransmitter, and is inactivated by cellular uptake catalyzed mostly by the glutamate transporter subtypes GLT-1 (EAAT2) and GLAST (EAAT1). Astrocytes express both GLT-1 and GLAST, while axon terminals in the neocortex only express GLT-1. To evaluate the role of GLT-1 in glutamate homeostasis, we injected GLT-1 knockout (KO) mice and wild-type littermates with [1-(13)C]glucose and [1,2-(13)C]acetate 15 min before euthanization. Metabolite levels were analyzed in extracts from neocortex and cerebellum and (13)C labeling in neocortex. Whereas the cerebellum in GLT-1-deficient mice had normal levels of glutamate, glutamine, and (13)C labeling of metabolites, glutamate level was decreased but labeling from [1-(13)C] glucose was unchanged in the neocortex. The contribution from pyruvate carboxylation toward labeling of these metabolites was unchanged. Labeling from [1,2-(13)C] acetate, originating in astrocytes, was decreased in glutamate and glutamine in the neocortex indicating reduced mitochondrial metabolism in astrocytes. The decreased amount of glutamate in the cortex indicates that glutamine transport into neurons is not sufficient to replenish glutamate lost because of neurotransmission and that GLT-1 plays a role in glutamate homeostasis in the cortex. Glutamate is the major excitatory neurotransmitter, and is inactivated by uptake via GLT-1 (EAAT2) and GLAST (EAAT1) transporters, while axon terminals in the neocortex only express GLT-1. To evaluate the role of GLT-1 in glutamate homeostasis, we used [1-(13)C]glucose and [1,2-(13)C]acetate injection and NMR spectroscopy. The results indicate that glutamine transport into neurons is not sufficient to replenish glutamate lost because of neurotransmission and that GLT-1 plays a role in glutamate homeostasis in the neocortex.


Asunto(s)
Transportador 2 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Homeostasis/fisiología , Neocórtex/fisiología , Aminoácidos/metabolismo , Animales , Cerebelo/citología , Cerebelo/metabolismo , Cromatografía Líquida de Alta Presión , Interpretación Estadística de Datos , Metabolismo Energético/fisiología , Transportador 2 de Aminoácidos Excitadores/genética , Femenino , Glucosa/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Ácido Pirúvico/metabolismo
20.
J Neurochem ; 129(1): 107-19, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24236946

RESUMEN

Triheptanoin, the triglyceride of heptanoate, is anticonvulsant in various epilepsy models. It is thought to improve energy metabolism in the epileptic brain by re-filling the tricarboxylic acid (TCA) cycle with C4-intermediates (anaplerosis). Here, we injected mice with [1,2-(13) C]glucose 3.5-4 weeks after pilocarpine-induced status epilepticus (SE) fed either a control or triheptanoin diet. Amounts of metabolites and incorporations of (13) C were determined in extracts of cerebral cortices and hippocampal formation and enzyme activity and mRNA expression were quantified. The percentage enrichment with two (13) C atoms in malate, citrate, succinate, and GABA was reduced in hippocampal formation of control-fed SE compared with control mice. Except for succinate, these reductions were not found in triheptanoin-fed SE mice, indicating that triheptanoin prevented a decrease of TCA cycle capacity. Compared to those on control diet, triheptanoin-fed SE mice showed few changes in most other metabolite levels and their (13) C labeling. Reduced pyruvate carboxylase mRNA and enzyme activity in forebrains and decreased [2,3-(13) C]aspartate amounts in cortex suggest a pyruvate carboxylation independent source of C-4 TCA cycle intermediates. Most likely anaplerosis was kept unchanged by carboxylation of propionyl-CoA derived from heptanoate. Further studies are proposed to fully understand triheptanoin's effects on neuroglial metabolism and interaction.


Asunto(s)
Ciclo del Ácido Cítrico/fisiología , Modelos Animales de Enfermedad , Epilepsia/metabolismo , Pilocarpina/toxicidad , Triglicéridos/administración & dosificación , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Ciclo del Ácido Cítrico/efectos de los fármacos , Epilepsia/inducido químicamente , Epilepsia/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Distribución Aleatoria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA