Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Biochem Biophys Res Commun ; 610: 140-146, 2022 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-35462095

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder leading to memory loss and impaired cognition. Despite several decades of research, AD therapeutic is not available. In this study, we have investigated the impact of a chronic intervention of riluzole on memory and neurometabolism in the AßPP-PS1 mouse model of AD. The 10-month-old AßPP-PS1 mice were administered 30 doses of riluzole (6 mg/kg, intragastrically) on an alternate day for two months. The memory was assessed using Morris Water Maze, while neurometabolism was evaluated by 1H-[13C]-NMR spectroscopy together with an intravenous infusion of [1,6-13C2]glucose. The normal saline-treated AßPP-PS1 mice exhibited a decrease in learning and memory that were restored to the control level following riluzole treatment. Most interestingly, the reduced 13C labeling of GluC4 and AspC3 from [1,6-13C]glucose in the AßPP-PS1 mice was restored to the control level following riluzole intervention. As a consequence, chronic riluzole treatment improved metabolic activity of glutamatergic neurons in AßPP-PS1 mice. Together these data suggest that riluzole may be useful for improving cognition in AD.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Glucosa/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/metabolismo , Riluzol/metabolismo , Riluzol/farmacología , Riluzol/uso terapéutico
2.
Indian J Med Res ; 156(4&5): 659-668, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36926783

RESUMEN

Background & objectives: COVID-19 has been a global pandemic since early 2020. It has diverse clinical manifestations, but consistent immunological and metabolic correlates of disease severity and protection are not clear. This study was undertaken to compare seropositivity rate, antibody levels against nucleocapsid and spike proteins, virus neutralization and metabolites between adult and child COVID-19 patients. Methods: Plasma samples from naïve control (n=14) and reverse transcription (RT)-PCR positive COVID-19 participants (n=132) were tested for reactivity with nucleocapsid and spike proteins by ELISA, neutralization of SARS-CoV-2 infectivity in Vero cells and metabolites by [1]H nuclear magnetic resonance (NMR) spectroscopy. Results: An ELISA platform was developed using nucleocapsid and spike proteins for COVID-19 serosurvey. The participants showed greater seropositivity for nucleocapsid (72%) than spike (55.3%), and males showed higher seropositivity than females for both the proteins. Antibody levels to both the proteins were higher in intensive care unit (ICU) than ward patients. Children showed lower seropositivity and antibody levels than adults. In contrast to ICU adults (81.3%), ICU children (33.3%) showed lower seropositivity for spike. Notably, the neutralization efficiency correlated with levels of anti-nucleocapsid antibodies. The levels of plasma metabolites were perturbed differentially in COVID-19 patients as compared with the naive controls. Interpretation & conclusions: Our results reflect the complexity of human immune response and metabolome to SARS-CoV-2 infection. While innate and cellular immune responses are likely to be a major determinant of disease severity and protection, antibodies to multiple viral proteins likely affect COVID-19 pathogenesis. In children, not adults, lower seropositivity rate for spike was associated with disease severity.


Asunto(s)
COVID-19 , SARS-CoV-2 , Masculino , Femenino , Animales , Chlorocebus aethiops , Humanos , Niño , Células Vero , Glicoproteína de la Espiga del Coronavirus , Formación de Anticuerpos , Anticuerpos Antivirales
3.
J Neurochem ; 146(6): 722-734, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29964293

RESUMEN

Depression is one of the most debilitating neuropsychiatric disorders. Most of the current antidepressants have long remission time and low recovery rate. This study explores the impact of ketamine on neuronal and astroglial metabolic activity in prefrontal cortex in a social defeat (SD) model of depression. C57BL/6 mice were subjected to a social defeat paradigm for 5 min a day for 10 consecutive days. Ketamine (10 mg/kg, intraperitoneal) was administered to mice for two consecutive days following the last defeat stress. Mice were infused with [1,6-13 C2 ]glucose or [2-13 C]acetate to assess neuronal and astroglial metabolic activity, respectively, together with proton-observed carbon-edited nuclear magnetic resonance spectroscopy in prefrontal cortex tissue extract. The 13 C labeling of amino acids from glucose and acetate was decreased in SD mice. Ketamine treatment in SD mice restored sucrose preference, social interaction and immobility time to control values. Acute subanesthetic ketamine restored the 13 C labeling of brain amino acids from glucose as well as acetate in SD mice to the respective control values, suggesting that rates of neuronal and astroglial tricarboxylic acid (TCA) cycle and neurotransmitter cycling were re-established to normal levels. The finding of improved energy metabolism in SD mice suggests that fast anti-depressant action of ketamine is linked with improved neurotransmitter cycling.


Asunto(s)
Analgésicos/uso terapéutico , Astrocitos/metabolismo , Trastorno Depresivo , Ketamina/uso terapéutico , Neuronas/metabolismo , Estrés Psicológico/complicaciones , Acetatos/farmacocinética , Animales , Astrocitos/efectos de los fármacos , Isótopos de Carbono/farmacocinética , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/etiología , Trastorno Depresivo/patología , Trastorno Depresivo/psicología , Modelos Animales de Enfermedad , Preferencias Alimentarias/efectos de los fármacos , Glucosa/farmacocinética , Jerarquia Social , Relaciones Interpersonales , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Natación/psicología
4.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 152-164, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27664837

RESUMEN

Cerebral ischemic stroke is one of the leading causes of death and disability worldwide. Therapeutic interventions to minimize ischemia-induced neural damage are limited due to poor understanding of molecular mechanisms mediating complex pathophysiology in stroke. Recently, epigenetic mechanisms mostly histone lysine (K) acetylation and deacetylation have been implicated in ischemic brain damage and have expanded the dimensions of potential therapeutic intervention to the systemic/local administration of histone deacetylase inhibitors. However, the role of other epigenetic mechanisms such as histone lysine methylation and demethylation in stroke-induced damage and subsequent recovery process is elusive. Here, we established an Internal Carotid Artery Occlusion (ICAO) model in CD1 mouse that resulted in mild to moderate level of ischemic damage to the striatum, as suggested by magnetic resonance imaging (MRI), TUNEL and histopathological staining along with an evaluation of neurological deficit score (NDS), grip strength and rotarod performance. The molecular investigations show dysregulation of a number of histone lysine methylases (KMTs) and few of histone lysine demethylases (KDMs) post-ICAO with significant global attenuation in the transcriptionally repressive epigenetic mark H3K9me2 in the striatum. Administration of Dimethyloxalylglycine (DMOG), an inhibitor of KDM4 or JMJD2 class of histone lysine demethylases, significantly ameliorated stroke-induced NDS by restoring perturbed H3K9me2 levels in the ischemia-affected striatum. Overall, these results highlight the novel role of epigenetic regulatory mechanisms controlling the epigenetic mark H3K9me2 in mediating the stroke-induced striatal damage and subsequent repair following mild to moderate cerebral ischemia.


Asunto(s)
Isquemia Encefálica/genética , Epigénesis Genética , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Lisina/genética , Aminoácidos Dicarboxílicos/farmacología , Aminoácidos Dicarboxílicos/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Muerte Celular/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Desmetilación/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Masculino , Metilación/efectos de los fármacos , Ratones
5.
Neurochem Res ; 42(1): 173-190, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28025798

RESUMEN

The 13C turnover of neurotransmitter amino acids (glutamate, GABA and aspartate) were determined from extracts of forebrain nerve terminals and brain homogenate, and fronto-parietal cortex from anesthetized rats undergoing timed infusions of [1,6-13C2]glucose or [2-13C]acetate. Nerve terminal 13C fractional labeling of glutamate and aspartate was lower than those in whole cortical tissue at all times measured (up to 120 min), suggesting either the presence of a constant dilution flux from an unlabeled substrate or an unlabeled (effectively non-communicating on the measurement timescale) glutamate pool in the nerve terminals. Half times of 13C labeling from [1,6-13C2]glucose, as estimated by least squares exponential fitting to the time course data, were longer for nerve terminals (GluC4, 21.8 min; GABAC2 21.0 min) compared to cortical tissue (GluC4, 12.4 min; GABAC2, 14.5 min), except for AspC3, which was similar (26.5 vs. 27.0 min). The slower turnover of glutamate in the nerve terminals (but not GABA) compared to the cortex may reflect selective effects of anesthesia on activity-dependent glucose use, which might be more pronounced in the terminals. The 13C labeling ratio for glutamate-C4 from [2-13C]acetate over that of 13C-glucose was twice as large in nerve terminals compared to cortex, suggesting that astroglial glutamine under the 13C glucose infusion was the likely source of much of the nerve terminal dilution. The net replenishment of most of the nerve terminal amino acid pools occurs directly via trafficking of astroglial glutamine.


Asunto(s)
Anestésicos por Inhalación/administración & dosificación , Encéfalo/metabolismo , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Terminales Presinápticos/metabolismo , Animales , Encéfalo/efectos de los fármacos , Isótopos de Carbono/metabolismo , Masculino , Terminales Presinápticos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar
6.
Proc Natl Acad Sci U S A ; 111(14): 5385-90, 2014 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-24706914

RESUMEN

Previous (13)C magnetic resonance spectroscopy experiments have shown that over a wide range of neuronal activity, approximately one molecule of glucose is oxidized for every molecule of glutamate released by neurons and recycled through astrocytic glutamine. The measured kinetics were shown to agree with the stoichiometry of a hypothetical astrocyte-to-neuron lactate shuttle model, which predicted negligible functional neuronal uptake of glucose. To test this model, we measured the uptake and phosphorylation of glucose in nerve terminals isolated from rats infused with the glucose analog, 2-fluoro-2-deoxy-D-glucose (FDG) in vivo. The concentrations of phosphorylated FDG (FDG6P), normalized with respect to known neuronal metabolites, were compared in nerve terminals, homogenate, and cortex of anesthetized rats with and without bicuculline-induced seizures. The increase in FDG6P in nerve terminals agreed well with the increase in cortical neuronal glucose oxidation measured previously under the same conditions in vivo, indicating that direct uptake and oxidation of glucose in nerve terminals is substantial under resting and activated conditions. These results suggest that neuronal glucose-derived pyruvate is the major oxidative fuel for activated neurons, not lactate-derived from astrocytes, contradicting predictions of the original astrocyte-to-neuron lactate shuttle model under the range of study conditions.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Ácido Láctico/metabolismo , Neuronas/metabolismo , Animales , Fosforilación , Ratas
7.
J Neurosci Res ; 93(7): 1101-8, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25663257

RESUMEN

γ-Aminobutyric acid (GABA) clearance from the extracellular space after release from neurons involves reuptake into terminals and astrocytes through GABA transporters (GATs). The relative flows through these two pathways for GABA released from neurons remains unclear. This study determines the effect of tiagabine, a selective inhibitor of neuronal GAT-1, on the rates of glutamate (Glu) and GABA metabolism and GABA resynthesis via the GABA-glutamine (Gln) cycle. Halothane-anesthetized rats were administered tiagabine (30 mg/kg, i.p.) and 45 min later received an intravenous infusion of either [1,6-(13)C2]glucose (in vivo) or [2-(13)C]acetate (ex vivo). Nontreated rats served as controls. Metabolites and (13)C enrichments were measured with (1)H-[(13)C]-nuclear magnetic resonance spectroscopy and referenced to their corresponding endpoint values measured in extracts from in situ frozen brain. Metabolic flux estimates of GABAergic and glutamatergic neurons were determined by fitting a metabolic model to the (13)C turnover data measured in vivo during [1,6-(13)C2]glucose infusion. Tiagabine-treated rats were indistinguishable (P > 0.05) from controls in tissue amino acid levels and in (13)C enrichments from [2-(13)C]acetate. Tiagabine reduced average rates of glucose oxidation and neurotransmitter cycling in both glutamatergic neurons (↓18%, CMR(glc(ox)Glu): control, 0.27 ± 0.05 vs. tiagabine, 0.22 ± 0.04 µmol/g/min; ↓11%, V(cyc(Glu-Gln)): control 0.23 ± 0.05 vs. tiagabine 0.21 ± 0.04 µmol/g/min and GABAergic neurons (↓18-25%, CMR(glc(ox)GABA): control 0.09 ± 0.02 vs. tiagabine 0.07 ± 0.03 µmol/g/min; V(cyc(GABA-Gln)): control 0.08 ± 0.02 vs. tiagabine 0.07 ± 0.03 µmol/g/min), but the changes in glutamatergic and GABAergic fluxes were not significant (P > 0.10). The results suggest that any reduction in GABA metabolism by tiagabine might be an indirect response to reduced glutamatergic drive rather than direct compensatory effects.


Asunto(s)
Anestesia , Encéfalo/efectos de los fármacos , Ácido Glutámico/metabolismo , Inhibidores de la Captación de Neurotransmisores/farmacología , Ácidos Nipecóticos/farmacología , Ácido gamma-Aminobutírico/metabolismo , Animales , Glucemia/metabolismo , Encéfalo/metabolismo , Isótopos de Carbono/administración & dosificación , Glutamina/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Wistar , Tiagabina
8.
J Biosci ; 492024.
Artículo en Inglés | MEDLINE | ID: mdl-38817158

RESUMEN

Adult muscle tissue largely comprised of differentiated myofibers also harbors quiescent muscle-resident stem cells (MuSCs) that are responsible for its maintenance, repair and regeneration. Emerging evidence suggests that quiescent MuSCs exhibit a specific metabolic state, which is regulated during physiological and pathological alterations. However, a detailed understanding of the metabolic state of quiescent MuSCs and its alteration during activation and repair is lacking. Direct profiling of MuSCs in vivo is challenging because the cells are rare and dispersed, while isolation and enrichment leads to their activation and loss of quiescence. In this study, we employed 1H-nuclear magnetic resonance (NMR) spectroscopy to profile metabolites in an established culture model of quiescent MuSC-derived myoblasts and compared with activated, proliferative and differentiated muscle cells to determine the state-specific metabolome. We report that the proliferating and differentiated cells are highly enriched in metabolites involved in energy generation, the quiescent state is enriched in metabolites related to phospholipid catabolism (glycerophosphocholine and choline) and depleted for phosphocholine which is enriched in proliferating cells. We propose that the ratio of these metabolites may be useful as a biomarker of MuSC quiescence.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Espectroscopía de Resonancia Magnética , Metabolómica , Metabolómica/métodos , Animales , Ratones , Espectroscopía de Resonancia Magnética/métodos , Mioblastos/metabolismo , Mioblastos/citología , Metaboloma , Músculo Esquelético/metabolismo , Músculo Esquelético/citología
9.
J Neurochem ; 127(3): 365-77, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23957451

RESUMEN

In this study, we have evaluated cerebral atrophy, neurometabolite homeostasis, and neural energetics in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP) model of Parkinson's disease. In addition, the efficacy of acute l-DOPA treatment for the reversal of altered metabolic functions was also evaluated. Cerebral atrophy and neurochemical profile were monitored in vivo using MRI and (1) H MR Spectroscopy. Cerebral energetics was studied by (1) H-[(13) C]-NMR spectroscopy in conjunction with infusion of (13) C labeled [1,6(-13) C2 ]glucose or [2-(13) C]acetate. MPTP treatment led to reduction in paw grip strength and increased level of GABA and myo-inositol in striatum and olfactory bulb. (13) C Labeling of glutamate-C4 (1.93 ± 0.24 vs. 1.48 ± 0.06 µmol/g), GABA-C2 (0.24 ± 0.04 vs. 0.18 ± 0.02 µmol/g) and glutamaine-C4 (0.26 ± 0.04 vs. 0.20 ± 0.04 µmol/g) from [1,6-(13) C2 ]glucose was found to be decreased with MPTP exposure in striatum as well as in other brain regions. However, glutamine-C4 labeling from [2-(13) C]acetate was found to be increased in the striatum of the MPTP-treated mice. Acute l-DOPA treatment failed to normalize the increased ventricular size and level of metabolites but recovered the paw grip strength and (13) C labeling of amino acids from [1,6-(13) C2 ]glucose and [2-(13) C]acetate in MPTP-treated mice. These data indicate that brain energy metabolism is impaired in Parkinson's disease and acute l-DOPA therapy could temporarily recover the cerebral metabolism. Cerebral atrophy, neurometabolite homeostasis, and neural energetics have been evaluated in an MPTP model of Parkinson's disease using MRI, in vivo (1) H MRS and (1) H-[(13) C]-NMR spectroscopy, respectively. MPTP treatment led to reduced paw grip strength and neuronal function. Acute Levodopa treatment was able to recover the diminished motor function and cerebral function. CMRGlc, Cerebral metabolic rate of glucose oxidation; MPTP, 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridin.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Química Encefálica/fisiología , Metabolismo Energético/fisiología , Levodopa/uso terapéutico , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/metabolismo , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Enfermedad de Parkinson Secundaria/metabolismo , Acetatos/metabolismo , Algoritmos , Animales , Glucemia/metabolismo , Química Encefálica/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Glutamatos/fisiología , Fuerza de la Mano/fisiología , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Neostriado/anatomía & histología , Neostriado/metabolismo , Oxidación-Reducción , Enfermedad de Parkinson Secundaria/inducido químicamente , Desempeño Psicomotor/fisiología , Radiofármacos/farmacocinética
10.
Neurotoxicology ; 94: 117-125, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36368548

RESUMEN

Lead poisoning has been identified as a problem in adults as well as in children. Chronic exposure to lead has been implicated in neurological disorders such as amyotrophic lateral sclerosis, Parkinson's disease, and Alzheimer's disease. In the present study, we evaluated the impact of chronic lead exposure on cerebral glutamatergic and GABAergic metabolic activity in mice. C57BL6 mice were provided lead acetate in drinking water for two months. The regional cerebral metabolic activity was measured using 1H-[13C]-NMR spectroscopy in conjunction with infusion of [1,6-13C2]glucose. The blood Pb2+ increased significantly in lead acetate treated mice. Concomitantly, there was a significant reduction in the forelimb strength. The level of myo-inositol was elevated in the cerebral cortex of mice chronically exposed to lead. The glutamatergic neurometabolic activity was found to be reduced following chronic lead exposure in the cerebral cortex, hippocampus, and striatum. In contrast, the GABAergic fluxes were impaired in the hippocampus and thalamus only. The metabolic fluxes in the cerebellum were unperturbed to Pb2+ toxicity. In conclusion, we report that chronic lead exposure in mice leads to an impairment in forelimb strength, and a perturbation in neurometabolism in brain regions involving cognition and movement.


Asunto(s)
Encéfalo , Plomo , Ratones , Animales , Plomo/toxicidad , Plomo/metabolismo , Ratones Endogámicos C57BL , Espectroscopía de Resonancia Magnética , Encéfalo/metabolismo , Acetatos
11.
J Cereb Blood Flow Metab ; 41(9): 2344-2355, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33657898

RESUMEN

Alzheimer's disease (AD) is a very common neurodegenerative disorder. Although a majority of the AD cases are sporadic, most of the studies are conducted using transgenic models. Intracerebroventricular (ICV) administered streptozotocin (STZ) animals have been used to explore mechanisms in sporadic AD. In this study, we have investigated memory and neurometabolism of ICV-STZ-administered C57BL6/J mice. The neuronal and astroglial metabolic activity was measured in 1H-[13C]-NMR spectrum of cortical and hippocampal tissue extracts of mice infused with [1,6-13C2]glucose and [2-13C]acetate, respectively. STZ-administered mice exhibited reduced (p = 0.00002) recognition index for memory. The levels of creatine, GABA, glutamate and NAA were reduced (p ≤ 0.04), while that of myo-inositol was increased (p < 0.05) in STZ-treated mice. There was a significant (p ≤ 0.014) reduction in aspartate-C3, glutamate-C4/C3, GABA-C2 and glutamine-C4 labeling from [1,6-13C2]glucose. This resulted in decreased rate of glucose oxidation in the cerebral cortex (0.64 ± 0.05 vs. 0.77 ± 0.05 µmol/g/min, p = 0.0008) and hippocampus (0.60 ± 0.04 vs. 0.73 ± 0.07 µmol/g/min, p = 0.001) of STZ-treated mice, due to similar reductions of glucose oxidation in glutamatergic and GABAergic neurons. Additionally, reduced glutamine-C4 labeling points towards compromised synaptic neurotransmission in STZ-treated mice. These data suggest that the ICV-STZ model exhibits neurometabolic deficits typically observed in AD, and its utility in understanding the mechanism of sporadic AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Encéfalo/metabolismo , Metabolismo Energético/genética , Espectroscopía de Resonancia Magnética/métodos , Estreptozocina/efectos adversos , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones
12.
Elife ; 92020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32955432

RESUMEN

Early adversity is a risk factor for the development of adult psychopathology. Common across multiple rodent models of early adversity is increased signaling via forebrain Gq-coupled neurotransmitter receptors. We addressed whether enhanced Gq-mediated signaling in forebrain excitatory neurons during postnatal life can evoke persistent mood-related behavioral changes. Excitatory hM3Dq DREADD-mediated chemogenetic activation of forebrain excitatory neurons during postnatal life (P2-14), but not in juvenile or adult windows, increased anxiety-, despair-, and schizophrenia-like behavior in adulthood. This was accompanied by an enhanced metabolic rate of cortical and hippocampal glutamatergic and GABAergic neurons. Furthermore, we observed reduced activity and plasticity-associated marker expression, and perturbed excitatory/inhibitory currents in the hippocampus. These results indicate that Gq-signaling-mediated activation of forebrain excitatory neurons during the critical postnatal window is sufficient to program altered mood-related behavior, as well as functional changes in forebrain glutamate and GABA systems, recapitulating aspects of the consequences of early adversity.


Stress and adversity in early childhood can have long-lasting effects, predisposing people to mental illness and mood disorders in adult life. The weeks immediately before and after birth are critical for establishing key networks of neurons in the brain. Therefore, any disruption to these neural circuits during this time can be detrimental to emotional development. However, it is still unclear which cellular mechanisms cause these lasting changes in behavior. Studies in animals suggest that these long-term effects could result from abnormalities in a few signaling pathways in the brain. For example, it has been proposed that overstimulating the cells that activate circuits in the forebrain ­ also known as excitatory neurons ­ may contribute to the behavioral changes that persist into adulthood. To test this theory, Pati et al. used genetic engineering to modulate a signaling pathway in male mice, which is known to stimulate excitatory neurons in the forebrain. The experiments showed that prolonged activation of excitatory neurons in the first two weeks after birth resulted in anxious and despair-like behaviors as the animals aged. The mice also displayed discrepancies in how they responded to certain external sensory information, which is a hallmark of schizophrenia-like behavior. However, engineering the same changes in adolescent and adult mice had no effect on their mood-related behaviors. This animal study reinforces just how critical the first few weeks of life are for optimal brain development. It provides an insight into a possible mechanism of how disruption during this time could alter emotional behavior. The findings are also relevant to psychiatrists interested in the underlying causes of mental illness after early childhood adversity.


Asunto(s)
Afecto/fisiología , Conducta Animal/fisiología , Neuronas/fisiología , Prosencéfalo/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/fisiología , Ansiedad/etiología , Femenino , Neuronas GABAérgicas/fisiología , Hipocampo/fisiología , Masculino , Ratones
13.
Brain Res ; 1229: 147-54, 2008 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-18621040

RESUMEN

Mechanisms underlying hypoxia-induced neuronal adaptation have not been fully elucidated. In the present study we investigated glucose metabolism and the activities of glycolytic and TCA cycle enzymes in cerebro-cortical neurons exposed to hypoxia (3 days in 1% of O2) or normoxia (room air). Hypoxia led to increased activities of LDH (194%), PK (90%), and HK (24%) and decreased activities of CS (15%) and GDH (34%). Neurons were incubated with [1-(13)C]glucose for 45 and 120 min under normoxic or hypoxic (120 min only) conditions and 13C enrichment determined in the medium and cell extract using 1H-{13C}-NMR. In hypoxia-treated neurons [3-(13)C]lactate release into the medium was 428% greater than in normoxia-treated controls (45-min normoxic incubation) and total flux through lactate was increased by 425%. In contrast glucose oxidation was reduced significantly in hypoxia-treated neurons, even when expressed relative to total cellular protein, which correlated with the reduced activities of the measured mitochondrial enzymes. The results suggest that surviving neurons adapt to prolonged hypoxia by up-regulation of glycolysis and down-regulation of oxidative energy metabolism, similar to certain other cell types. The factors leading to adaptation and survival for some neurons but not others remain to be determined.


Asunto(s)
Cerebelo/citología , Corteza Cerebral/citología , Metabolismo Energético/fisiología , Hipoxia/fisiopatología , Neuronas/fisiología , Animales , Isótopos de Carbono/metabolismo , Células Cultivadas , Citrato (si)-Sintasa/metabolismo , Embrión de Mamíferos , Femenino , Glucosa/metabolismo , Glutamato Deshidrogenasa/metabolismo , Hexoquinasa/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/metabolismo , Espectroscopía de Resonancia Magnética/métodos , Embarazo , Piruvato Quinasa/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
14.
J Cereb Blood Flow Metab ; 38(7): 1213-1226, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-28585882

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disease associated with progressive loss of cognitive function, personality, and behavior. The present study evaluates neuronal and astroglial metabolic activity, and neurotransmitter cycle fluxes in AßPP-PS1 mouse model of AD by using 1H-[13C]-nuclear magnetic resonance (NMR) spectroscopy together with an infusion of either [1,6-13C2]glucose or [2-13C]acetate. The levels of N-acetyl-aspartate (NAA) and glutamate were found to be decreased in the cerebral cortex and hippocampus in AßPP-PS1 mice, when compared with wild type controls. The cerebral metabolic rate of acetate oxidation was increased in the hippocampus and cerebral cortex of AßPP-PS1 mice suggesting enhanced astroglial activity in AD. AßPP-PS1 mice exhibit severe reduction in glutamatergic and gamma-amino butyric acid (GABA)ergic neuronal metabolic activity and neurotransmitter cycling fluxes in the hippocampus, cerebral cortex, and striatum as compared with controls. These data suggest that metabolic activity of excitatory and inhibitory neurons is compromised across brain in AßPP-PS1 mouse model of AD.


Asunto(s)
Astrocitos , Encéfalo , Imagen por Resonancia Magnética , Neuronas , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Presenilina-1/genética , Presenilina-1/metabolismo
15.
J Cereb Blood Flow Metab ; 27(12): 1895-907, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17440492

RESUMEN

The contribution of glutamatergic and gamma-aminobutyric acid (GABA)ergic neurons to oxidative energy metabolism and neurotransmission in the developing brain is not known. Glutamatergic and GABAergic fluxes were assessed in neocortex of postnatal day 10 (P10) and 30 (P30) urethane-anesthetized rats infused intravenously with [1,6-(13)C(2)]glucose for different time intervals (time course) or with [2-(13)C]acetate for 2 to 3 h (steady state). Amino acid levels and (13)C enrichments were determined in tissue extracts ex vivo using (1)H-[(13)C]-NMR spectroscopy. Metabolic fluxes were estimated from the best fits of a three-compartment metabolic model (glutamatergic neurons, GABAergic neurons, and astroglia) to the (13)C-enrichment time courses of amino acids from [1,6-(13)C(2)]glucose, constrained by the ratios of neurotransmitter cycling (V(cyc))-to-tricarboxylic acid (TCA) cycle flux (V(TCAn)) calculated from the steady-state [2-(13)C]acetate enrichment data. From P10 to P30 increases in total neuronal (glutamate plus GABA) TCA cycle flux (3 x ; 0.24+/-0.05 versus 0.71+/-0.07 micromol per g per min, P<0.0001) and total neurotransmitter cycling flux (3.1 to 5 x ; 0.07 to 0.11 (+/-0.03) versus 0.34+/-0.03 micromol per g per min, P<0.0001) were approximately proportional. Incremental changes in total cycling (DeltaV(cyc(tot))) and neuronal TCA cycle flux (DeltaV(TCAn(tot))) between P10 and P30 were 0.23 to 0.27 and 0.47 micromol per g per min, respectively, similar to the approximately 1:2 relationship previously reported for adult cortex. For the individual neurons, increases in V(TCAn) and V(cyc) were similar in magnitude (glutamatergic neurons, 2.7 x versus 2.8 to 4.6 x ; GABAergic neurons, approximately 5 x versus approximately 7 x), although GABAergic flux changes were larger. The findings show that glutamate and GABA neurons undergo large and approximately proportional increases in neurotransmitter cycling and oxidative energy metabolism during this major postnatal growth spurt.


Asunto(s)
Corteza Cerebral/metabolismo , Metabolismo Energético/fisiología , Ácido Glutámico/fisiología , Neurotransmisores/metabolismo , Ácido gamma-Aminobutírico/fisiología , Acetatos/farmacocinética , Algoritmos , Aminoácidos/metabolismo , Animales , Glucemia/metabolismo , Química Encefálica/fisiología , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Ciclo del Ácido Cítrico/fisiología , Interpretación Estadística de Datos , Glucosa/farmacocinética , Cinética , Espectroscopía de Resonancia Magnética , Masculino , Neuroglía/metabolismo , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
16.
J Biosci ; 42(3): 363-371, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29358550

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by progressive loss of memory and cognitive function. The cerebral metabolic rate of glucose oxidation has been shown to be reduced in AD. The present study evaluated efficacy of dietary Amalaki Rasayana (AR), an Ayurvedic formulation used in Indian traditional system, in AbPP-PS1 mouse model of AD in ameliorating memory and neurometabolism, and compared with donepezil, a standard FDA approved drug for AD. The memory of mice was measured using Morris Water Maze analysis. The cerebral metabolism was followed by 13C labelling of brain amino acids in tissue extracts ex vivo using 1H-[13C]-NMR spectroscopy together with a short time infusion of [1,6-13C2]glucose to mice. The intervention with Amalaki Rasayana showed improved learning and memory in AbPP-PS1 mice. The 13C labelings of GluC4, GABAC2 and GlnC4 were reduced in AbPP-PS1 mice when compared with wild-type controls. Intervention of AR increased the 13C labelling of amino acids suggesting a significant enhancement in glutamatergic and GABAergic metabolic activity in AbPP-PS1 mice similar to that observed with donepezil treatment. These data suggest that AR has potential to improve memory and cognitive function in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Cognición/efectos de los fármacos , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Isótopos de Carbono , Donepezilo , Expresión Génica , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Indanos/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Medicina Ayurvédica/métodos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Piperidinas/farmacología , Presenilina-1/genética , Presenilina-1/metabolismo , Ácido gamma-Aminobutírico/metabolismo
17.
Front Mol Neurosci ; 10: 323, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29089867

RESUMEN

Alzheimer's disease (AD) is an age-related neurodegenerative disorder, characterized by progressive loss of cognitive functions and memory. Excessive intake of aluminum chloride in drinking water is associated with amyloid plaques and neurofibrillary tangles in the brain, which are the hallmark of AD. We have evaluated brain energy metabolism in aluminum chloride (AlCl3) mouse model of AD. In addition, effectiveness of Rasa Sindoor (RS), a formulation used in Indian Ayurvedic medicine, for alleviation of symptoms of AD was evaluated. Mice were administered AlCl3 (40 mg/kg) intraperitoneally once a day for 60 days. The memory of mice was measured using Morris Water Maze test. The 13C labeling of brain amino acids was measured ex vivo in tissue extracts using 1H-[13C]-NMR spectroscopy with timed infusion of [1,6-13C2]glucose. The 13C turnover of brain amino acids was analyzed using a three-compartment metabolic model to derive the neurotransmitter cycling and TCA cycle rates associated with glutamatergic and GABAergic pathways. Exposure of AlCl3 led to reduction in memory of mice. The glutamatergic and GABAergic neurotransmitter cycling and glucose oxidation were found to be reduced in the cerebral cortex, hippocampus, and striatum following chronic AlCl3 treatment. The perturbation in metabolic rates was highest in the cerebral cortex. However, reduction in metabolic fluxes was higher in hippocampus and striatum following one month post AlCl3 treatment. Most interestingly, oral administration of RS (2 g/kg) restored memory as well as the energetics of neurotransmission in mice exposed to AlCl3. These data suggest therapeutic potential of RS to manage cognitive functions and memory in preclinical AD.

18.
Mater Sci Eng C Mater Biol Appl ; 72: 415-424, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28024604

RESUMEN

One-pot synthesis methods for development of hydrophilic imaging nanoprobes have advantages over multi-pot methods due to their simple procedures, less probability for degradation of efficiency, superior control over growth and morphology, cost effectiveness, improved scope for scale-up synthesis etc. Here, we present a novel one-pot facile synthesis of hydrophilic colloidal bimodal nanoprobe (FePt-CdS) prepared through a seed-mediated nucleation and growth technique. In this facile synthesis of complex nanostructure, glutathione (GSH) was used as the capping agent to render biocompatibility and dispersibility. The microstructure, surface, optical, magnetic, biocompatibility, relaxivity and imaging property of the developed nanoprobe have been studied. The microstructural characterizations reveal average size of the particle as ~9-11nm with bleb shaped morphology. Spectroscopic characterization depicts the development of GSH capped CdS QDs on FePt, surface functionalities and their stability. The magnetic measurements confirm the superparamagnetic property in the developed bimodal nanoprobe. In addition, the GSH capping imparts excellent biocompatibility, water dispersibility, and fluorescence property to the probe. In RAW 264.7 macrophage cells, the bimodal nanoprobes exhibit intense green and red fluorescence. The magnetic resonance imaging (MRI) and fluorescence imaging (FI) study depict high transverse relaxivity and visible range fluorescent property in the synthesized FePt-CdS nanoprobe. Hence, the developed bimodal nanoprobe can be used as a potential candidate in simultaneous FI and MR imaging.


Asunto(s)
Materiales Biocompatibles/química , Compuestos de Cadmio/química , Glutatión/química , Platino (Metal)/química , Puntos Cuánticos/química , Sulfuros/química , Animales , Materiales Biocompatibles/toxicidad , Supervivencia Celular/efectos de los fármacos , Interacciones Hidrofóbicas e Hidrofílicas , Imagen por Resonancia Magnética , Magnetismo , Ratones , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Puntos Cuánticos/toxicidad , Células RAW 264.7 , Espectrometría por Rayos X , Propiedades de Superficie , Difracción de Rayos X
19.
J Cereb Blood Flow Metab ; 26(7): 865-77, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16407855

RESUMEN

Prior 13C magnetic resonance spectroscopy (MRS) experiments, which simultaneously measured in vivo rates of total glutamate-glutamine cycling (V(cyc(tot))) and neuronal glucose oxidation (CMR(glc(ox), N)), revealed a linear relationship between these fluxes above isoelectricity, with a slope of approximately 1. In vitro glial culture studies examining glutamate uptake indicated that glutamate, which is cotransported with Na+, stimulated glial uptake of glucose and release of lactate. These in vivo and in vitro results were consolidated into a model: recycling of one molecule of neurotransmitter between glia and neurons was associated with oxidation of one glucose molecule in neurons; however, the glucose was taken up only by glia and all the lactate (pyruvate) generated by glial glycolysis was transferred to neurons for oxidation. The model was consistent with the 1:1 relationship between DeltaCMR(glc(ox), N) and DeltaV(cyc(tot)) measured by 13C MRS. However, the model could not specify the energetics of glia and gamma-amino butyric acid (GABA) neurons because quantitative values for these pathways were not available. Here, we review recent 13C and 14C tracer studies that enable us to include these fluxes in a more comprehensive model. The revised model shows that glia produce at least 8% of total oxidative ATP and GABAergic neurons generate approximately 18% of total oxidative ATP in neurons. Neurons produce at least 88% of total oxidative ATP, and take up approximately 26% of the total glucose oxidized. Glial lactate (pyruvate) still makes the major contribution to neuronal oxidation, but approximately 30% less than predicted by the prior model. The relationship observed between DeltaCMR(glc(ox), N) and DeltaV(cyc(tot)) is determined by glial glycolytic ATP as before. Quantitative aspects of the model, which can be tested by experimentation, are discussed.


Asunto(s)
Glucosa/metabolismo , Ácido Glutámico/fisiología , Neuroglía/metabolismo , Ácido gamma-Aminobutírico/fisiología , Animales , Glucosa/química , Humanos , Espectroscopía de Resonancia Magnética/métodos , Modelos Biológicos , Neuroglía/química , Oxidación-Reducción , Sensibilidad y Especificidad
20.
Neurochem Int ; 48(6-7): 508-14, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16517019

RESUMEN

Cellular GABA levels are determined by the dynamic balance between synthesis and catabolism and are regulated at the level of glutamate decarboxylase, precursor availability (e.g., glutamate and glutamine), and possibly GABA degradation. GABA levels rise and stabilize within hours in human cortex following orally administered vigabatrin, an irreversible inhibitor of GABA-T, suggesting potential product inhibition of GABA synthesis or enhanced GABA degradation through the non-inhibited GABA-T fraction. In this study time courses of the rise in cortical GABA were measured in anesthetized rats in vivo after vigabatrin treatment using localized (1)H magnetic resonance spectroscopy and the times to reach steady-state for a given dose were determined. Rates of GABA synthesis were estimated for the period of constant GABA level from the accumulation of [2-(13)C]GABA following a short intravenous infusion (20 min) of either [1,6-(13)C(2)]glucose or [2-(13)C]acetate. No evidence of product inhibition of glutamate decarboxylase by the increased GABA concentration or reduced synthesis from [1,6-(13)C(2)]glucose (control, 0.031+/-0.010; vigabatrin-treated, 0.037+/-0.004 micromol/g/min, P=0.30) or [2-(13)C]acetate (control, 0.078+/-0.010; vigabatrin-treated, 0.084+/-0.006 micromol/g/min, P=0.42) was found. Fractional changes in steady-state GABA levels and GABA-T activities 5-6 h after vigabatrin treatment were approximately equal. The lack of change in GABA synthesis (and GABA catabolic flux for constant GABA levels) suggests that GABA-T has a near-zero flux control coefficient in vivo-capable of greatly altering the steady-state GABA concentration but exerting little or no control on GABA synthesis or GABA/glutamine cycling flux. The findings are consistent with a Michaelis-Menten kinetic model whereby cellular GABA levels increase until flux through the remaining (uninhibited) transaminase equals the rate of GABA synthesis. The findings suggest that astroglia may be the site of continuing GABA catabolism after acute vigabatrin treatment.


Asunto(s)
4-Aminobutirato Transaminasa/antagonistas & inhibidores , Corteza Cerebral/efectos de los fármacos , Vigabatrin/farmacología , Ácido gamma-Aminobutírico/biosíntesis , Ácido Acético/metabolismo , Animales , Corteza Cerebral/metabolismo , Glucosa/metabolismo , Espectroscopía de Resonancia Magnética , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA