Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Brain Res ; 1771: 147660, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34529964

RESUMEN

Since we found that inhibition of cyclooxygenase-2 (COX-2) with concomitant application of a metabotropic glutamate receptor subtype 5 (mGluR5) antagonist (MTEP) down-regulates mGluR7 in the hippocampus (HC) and changes behavior of mice, our team decided to investigate the mechanism responsible for the observed changes. The amino acid glutamate (Glu) is a major excitatory neurotransmitter in the brain. Glu uptake is regulated by excitatory amino acid transporters (EAAT). There are five transporters with documented expression in neurons and glia in the central nervous system (CNS). EAATs, maintain the correct transmission of the Glu signal and prevent its toxic accumulation by removing Glu from the synapse. It has been documented that the toxic level of Glu is one of the main causes of mental and cognitive abnormalities. Given the above mechanisms involved in the functioning of the Glu synapse, we hypothesized modification of Glu uptake, involving EAATs as the cause of the observed changes. This study investigated the level of selected EAATs in the HC after chronic treatment with mGluR5 antagonist MTEP, NS398, and their combination using Western blot. Concomitant MTEP treatment with NS398 or a single administration of the above causes changes in LTP and modulation of EAAT levels in mouse HC. As EAATs are cellular markers of oxidative stress mechanisms, the E. coli lipopolysaccharide (LPS) challenge was performed. The modified Barnes maze test (MBM) revealed alterations in the mouse spatial learning abilities. This study reports an interaction between the mGluR5 and COX-2 in the HC, with EAAT1 and EAAT3 involvement.


Asunto(s)
Ciclooxigenasa 2/fisiología , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 3 de Aminoácidos Excitadores/biosíntesis , Hipocampo/metabolismo , Estrés Oxidativo , Receptores de Ácido Kaínico/fisiología , Animales , Inhibidores de la Ciclooxigenasa 2/farmacología , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/genética , Lipopolisacáridos/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Receptores de Ácido Kaínico/antagonistas & inhibidores , Aprendizaje Espacial/efectos de los fármacos
2.
Neurobiol Dis ; 154: 105340, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33753288

RESUMEN

Bergmann glia (BG) are highly specialized radial astrocytes of the cerebellar cortex, which play a key role in the uptake of synaptic glutamate via the excitatory amino acid transporter EAAT1. Multiple lines of evidence suggest that in cerebellar neurodegenerative diseases reactive BG has a negative impact on neuronal function and survival through compromised EAAT activity. A family of such diseases are those caused by expansion of CAG repeats in genes of the ataxin family, resulting in spinocerebellar ataxias (SCA). We investigated the contribution of BG to the pathogenesis of cerebellar neurodegeneration in a model of SCA1, which was induced by expression of a polyglutamine mutant of ataxin-1 (ATXN1[Q85]) in BG specifically. We compared the outcomes with a novel model where we triggered excitotoxicity by a chronic optogenetic activation of BG with channelrhodopsin-2 (ChR2). In both cases we detected evidence of reduced glutamate uptake manifested by prolongation of excitatory postsynaptic currents in Purkinje cells which is consistent with documented reduction of expression and/or function of EAAT1. In both models we detected astroglyosis and Purkinje cells atrophy. Finally, the same pattern was detected in a knock-in mouse which expresses a polyglutamine mutant ataxin-1 ATXN1[Q154] in a non-cell-selective manner. Our results suggest that ATXN1[Q85] and ChR2-induced insult targeted to BG closely mimics SCA1 pathology, where excessive glutamate signaling appears to be a common feature likely being an important contributor to cerebellar neurodegeneration.


Asunto(s)
Ataxina-1/biosíntesis , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Neuroglía/metabolismo , Optogenética/efectos adversos , Células de Purkinje/metabolismo , Animales , Ataxina-1/genética , Muerte Celular/fisiología , Transportador 1 de Aminoácidos Excitadores/genética , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroglía/patología , Estimulación Luminosa/efectos adversos , Células de Purkinje/patología
3.
Neurosci Lett ; 735: 135237, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32645399

RESUMEN

Glutamate-mediated excitatory synaptic signalling is primarily controlled by excitatory amino acid transporters (EAATs), such as EAAT1 and EAAT2, which are located mostly on astrocytes and, together, uptake more than 95 % of extracellular glutamate. Alterations in the functional expression levels of EAATs can lead to excessive extracellular glutamate accumulation, potentially triggering excitotoxicity and seizures, among other neurological disorders. Excitotoxicity induced in early developmental stages can lead to lasting changes in several neurotransmission systems, including the glutamatergic system, which could make the brain more susceptible to a second insult. In this study, the expression levels of EAAT1 (GLAST) and EAAT2 (GLT-1) proteins were assessed in the cerebral motor cortex (CMC), striatum, hippocampus and entorhinal cortex (EC) of male adult rats following the neonatal excitotoxic process triggered by monosodium glutamate (MSG)-treatment (4 g/kg of body weight at postnatal days 1,3,5 and 7, subcutaneously). Western blot analysis showed that neonatal MSG-treatment decreased EAAT1 expression levels in the CMC, striatum and hippocampus, while EAAT2 levels were increased in the striatum and EC and decreased in the CMC. Immunofluorescence staining confirmed the changes in EAAT1 and EAAT2 expression induced by neonatal MSG-treatment, which were accompanied by an increase in the glial fibrillary acidic protein (GFAP) immunofluorescence signalthat was particularly significant in the hippocampus. Our results show that a neonatal excitotoxic processes can induce lasting changes in the expression levels of EAAT1 and EAAT2 proteins and suggest that although astrogliosis occurs, glutamate uptake could be deficient, particularly in the CMC and hippocampus.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Glutamato de Sodio/toxicidad , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/efectos de los fármacos , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 2 de Aminoácidos Excitadores/genética , Expresión Génica , Ácido Glutámico/toxicidad , Masculino , Ratas , Ratas Wistar
4.
Neurochem Int ; 125: 111-116, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30817938

RESUMEN

We have analysed post-mortem samples of prefrontal cortex from control and alcoholic human brains by the technique of Western blotting to estimate and compare the expressions of glutamate transporter GLAST (Excitatory Amino Acid Transporter One; EAAT1). Furthermore, using the non-alcoholic prefrontal cortex and custom-made GLAST (EAAT1) antibody we determined GLAST (EAAT1) "interactome" i.e. the set of proteins selectively bound by GLAST (EAAT1). We found that GLAST (EAAT1) was significantly more abundant (about 1.6-fold) in the cortical tissue from alcoholic brains compared to that from non-alcoholic controls. The greatest increase in the level of GLAST (EAAT1) was found in plasma membrane fraction (2.2-fold). Additionally, using the prefrontal cortical tissue from control brains, we identified 38 proteins specifically interacting with GLAST (EAAT1). These can be classified as contributing to the cell structure (6 proteins; 16%), energy and general metabolism (18 proteins; 47%), neurotransmitter metabolism (three proteins; 8%), signalling (6 proteins: 16%), neurotransmitter storage/release at synapses (three proteins; 8%) and calcium buffering (two proteins; 5%). We discuss possible consequences of the increased expression of GLAST (EAAT1) in alcoholic brain tissue and whether or how this could disturb the function of the proteins potentially interacting with GLAST (EAAT1) in vivo. The data represent an extension of our previous proteomic and metabolomic studies of human alcoholism revealing another aspect of the complexity of changes imposed on brain by chronic long-term consumption of ethanol.


Asunto(s)
Alcoholismo/metabolismo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Metabolómica/métodos , Corteza Prefrontal/metabolismo , Proteómica/métodos , Adulto , Anciano , Alcohólicos , Alcoholismo/genética , Alcoholismo/patología , Transportador 1 de Aminoácidos Excitadores/genética , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Corteza Prefrontal/patología
5.
J Cereb Blood Flow Metab ; 39(1): 58-73, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29135354

RESUMEN

Depression after traumatic brain injury (TBI) is common but the mechanisms by which TBI causes depression are unknown. TBI decreases glutamate transporters GLT-1 and GLAST and allows extravasation of thrombin. We examined the effects of thrombin on transporter expression in primary hippocampal astrocytes. Application of a PAR-1 agonist caused down-regulation of GLT-1, which was prevented by inhibition of Rho kinase (ROCK). To confirm these mechanisms in vivo, we subjected mice to closed-skull TBI. Thrombin activity in the hippocampus increased one day following TBI. Seven days following TBI, expression of GLT-1 and GLAST was reduced in the hippocampus, and this was prevented by administration of the PAR-1 antagonist SCH79797. Inhibition of ROCK attenuated the decrease in GLT-1, but not GLAST, after TBI. We measured changes in glutamate levels in the hippocampus seven days after TBI using an implanted biosensor. Stress-induced glutamate levels were significantly increased following TBI and this was attenuated by treatment with the ROCK inhibitor fasudil. We quantified depressive behavior following TBI and found that inhibition of PAR-1 or ROCK decreased these behaviors. These results identify a novel mechanism by which TBI results in down-regulation of astrocyte glutamate transporters and implicate astrocyte and glutamate transporter dysfunction in depression following TBI.


Asunto(s)
Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/genética , Depresión/etiología , Depresión/genética , Hipocampo/metabolismo , Trombina , Proteínas de Transporte Vesicular de Glutamato/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Conducta Animal , Barrera Hematoencefálica/patología , Depresión/psicología , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/genética , Ácido Glutámico/metabolismo , Hipocampo/citología , Masculino , Ratones , Receptor PAR-1/genética , Proteínas de Transporte Vesicular de Glutamato/genética , Quinasas Asociadas a rho/antagonistas & inhibidores
6.
Neurotoxicology ; 67: 112-120, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29778792

RESUMEN

Exposure to elevated levels of manganese (Mn) causes manganism, a neurological disorder with similar characteristics to those of Parkinson's disease (PD). Valproic acid (VPA), an antiepileptic, is known to inhibit histone deacetylases and exert neuroprotective effects in many experimental models of neurological disorders. In the present study, we investigated if VPA attenuated Mn-induced dopaminergic neurotoxicity and the possible mechanisms involved in VPA's neuroprotection, focusing on modulation of astrocytic glutamate transporters (glutamate aspartate transporter, GLAST and glutamate transporter 1, GLT-1) and histone acetylation in H4 astrocyte culture and mouse models. The results showed that VPA increased promoter activity, mRNA/protein levels of GLAST/GLT-1 and glutamate uptake, and reversed Mn-reduced GLAST/GLT-1 in in vitro astrocyte cultures. VPA also attenuated Mn-induced reduction of GLAST and GLT-1 mRNA/protein levels in midbrain and striatal regions of the mouse brain when VPA (200 mg/kg, i.p., daily, 21 d) was administered 30 min prior to Mn exposure (30 mg/kg, intranasal instillation, daily, 21 d). Importantly, VPA attenuated Mn-induced dopaminergic neuronal damage by reversing Mn-induced decrease of tyrosine hydroxylase (TH) mRNA/protein levels in the nigrostriatal regions. VPA also reversed Mn-induced reduction of histone acetylation in astrocytes as well as mouse brain tissue. Taken together, VPA exerts attenuation against Mn-induced decrease of astrocytic glutamate transporters parallel with reversing Mn-induced dopaminergic neurotoxicity and Mn-reduced histone acetylation. Our findings suggest that VPA could serve as a potential neuroprotectant against Mn neurotoxicity as well as other neurodegenerative diseases associated with excitotoxicity and impaired astrocytic glutamate transporters.


Asunto(s)
Encéfalo/metabolismo , Dopamina/metabolismo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Manganeso/toxicidad , Ácido Valproico/farmacología , Animales , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Células Cultivadas , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 2 de Aminoácidos Excitadores/genética , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
EMBO J ; 37(9)2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29615452

RESUMEN

Tissues contain distinct stem cell niches, but whether cell turnover is coordinated between niches during growth is unknown. Here, we report that in mouse skin, hair growth is accompanied by sebaceous gland and interfollicular epidermis expansion. During hair growth, cells in the bulge and outer root sheath temporarily upregulate the glutamate transporter SLC1A3, and the number of SLC1A3+ basal cells in interfollicular epidermis and sebaceous gland increases. Fate mapping of SLC1A3+ cells in mice revealed transient expression in proliferating stem/progenitor cells in all three niches. Deletion of slc1a3 delays hair follicle anagen entry, uncouples interfollicular epidermis and sebaceous gland expansion from the hair cycle, and leads to reduced fur density in aged mice, indicating a role of SLC1A3 in stem/progenitor cell activation. Modulation of metabotropic glutamate receptor 5 activity mimics the effects of SLC1A3 deletion or inhibition. These data reveal that stem/progenitor cell activation is synchronized over distinct niches during growth and identify SLC1A3 as a general marker and effector of activated epithelial stem/progenitor cells throughout the skin.


Asunto(s)
Proliferación Celular/fisiología , Epidermis/crecimiento & desarrollo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Regulación de la Expresión Génica/fisiología , Glándulas Sebáceas/crecimiento & desarrollo , Células Madre/metabolismo , Animales , Transportador 1 de Aminoácidos Excitadores/genética , Ratones , Ratones Transgénicos , Glándulas Sebáceas/citología
8.
J Neuroinflammation ; 15(1): 5, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29304807

RESUMEN

BACKGROUND: Laquinimod is an immunomodulatory drug under clinical investigation for the treatment of the progressive form of multiple sclerosis (MS) with both anti-inflammatory and neuroprotective effects. Excitotoxicity, a prominent pathophysiological feature of MS and of its animal model, experimental autoimmune encephalomyelitis (EAE), involves glutamate transporter (GluT) dysfunction in glial cells. The aim of this study was to assess whether laquinimod might exert direct neuroprotective effects by interfering with the mechanisms of excitotoxicity linked to GluT function impairments in EAE. METHODS: Osmotic minipumps allowing continuous intracerebroventricular (icv) infusion of laquinimod for 4 weeks were implanted into C57BL/6 mice before EAE induction. EAE cerebella were taken to perform western blot and qPCR experiments. For ex vivo experiments, EAE cerebellar slices were incubated with laquinimod before performing electrophysiology, western blot, and qPCR. RESULTS: In vivo treatment with laquinimod attenuated EAE clinical score at the peak of the disease, without remarkable effects on inflammatory markers. In vitro application of laquinimod to EAE cerebellar slices prevented EAE-linked glutamatergic alterations without mitigating astrogliosis and inflammation. Moreover, such treatment induced an increase of Slcla3 mRNA coding for the glial glutamate-aspartate transporter (GLAST) without affecting the protein content. Concomitantly, laquinimod significantly increased the levels of the glial glutamate transporter 1 (GLT-1) protein and pharmacological blockade of GLT-1 function fully abolished laquinimod anti-excitotoxic effect. CONCLUSIONS: Overall, our results suggest that laquinimod protects against glutamate excitotoxicity of the cerebellum of EAE mice by bursting the expression of glial glutamate transporters, independently of its anti-inflammatory effects.


Asunto(s)
Antiinflamatorios/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Ácido Glutámico/metabolismo , Quinolonas/administración & dosificación , Animales , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Femenino , Infusiones Intraventriculares , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Distribución Aleatoria
10.
Invest Ophthalmol Vis Sci ; 57(6): 2721-8, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27196320

RESUMEN

PURPOSE: Müller cells support retinal neurons with essential functions. Here, we aim to examine the impact of starvation and oxidative stress on glutamate uptake and mitochondrial function in Müller cells. METHODS: Cultured human retinal Müller cells (MIO-M1) were exposed to H2O2 and additional starvation for 24 hours. Effects of starvation and H2O2 on glutamate uptake and mitochondrial function were assessed by kinetic glutamate uptake assays and Seahorse assays, respectively. Cell survival was evaluated by cell viability assays. mRNA and protein expressions were assessed by quantitative PCR and Western blot. RESULTS: Starvation of Müller cells increased the glutamate uptake capacity as well as the expression of the most abundant glutamate transporter, EAAT1. Mitochondrial and glycolytic activity were diminished in starved Müller cells despite unaffected cell viability. Simultaneous starvation and exposure to oxidative stress resulted in a reduced glutamate uptake and a collapsed mitochondrial function. In Müller cells with intact energy supply, the glutamate uptake and mitochondrial function were unaffected after exposure to oxidative stress. CONCLUSIONS: Here, we identify an increased susceptibility toward oxidative stress in starved Müller cells in spite of unaffected viability and an apparent decreased ability to transport glutamate. Solely exposure to oxidative stress did not affect Müller cell functions. Thus, our study suggests an increased susceptibility of Müller cells in case of more than one cellular stressor. Extrapolating these findings, age-related neurodegenerative retinal diseases may be the result of impaired Müller cell function.


Asunto(s)
Células Ependimogliales/metabolismo , Transportador 1 de Aminoácidos Excitadores/genética , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Estrés Oxidativo/genética , Retina/metabolismo , Inanición/metabolismo , Western Blotting , Supervivencia Celular , Células Cultivadas , Células Ependimogliales/patología , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Humanos , Mitocondrias/metabolismo , Reacción en Cadena de la Polimerasa , ARN/genética , Retina/patología , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Inanición/patología
11.
Development ; 143(7): 1170-81, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26893340

RESUMEN

Astrocytes are crucial in the formation, fine-tuning, function and plasticity of neural circuits in the central nervous system. However, important questions remain about the mechanisms instructing astrocyte cell fate. We have studied astrogenesis in the ventral nerve cord of Drosophila larvae, where astrocytes exhibit remarkable morphological and molecular similarities to those in mammals. We reveal the births of larval astrocytes from a multipotent glial lineage, their allocation to reproducible positions, and their deployment of ramified arbors to cover specific neuropil territories to form a stereotyped astroglial map. Finally, we unraveled a molecular pathway for astrocyte differentiation in which the Ets protein Pointed and the Notch signaling pathway are required for astrogenesis; however, only Notch is sufficient to direct non-astrocytic progenitors toward astrocytic fate. We found that Prospero is a key effector of Notch in this process. Our data identify an instructive astrogenic program that acts as a binary switch to distinguish astrocytes from other glial cells.


Asunto(s)
Astrocitos/citología , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Proteínas del Tejido Nervioso/genética , Neurópilo/citología , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas/genética , Receptores Notch/genética , Factores de Transcripción/genética , Animales , Astrocitos/metabolismo , Linaje de la Célula/fisiología , Sistema Nervioso Central/embriología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/biosíntesis , Proteínas de Drosophila/metabolismo , Transportador 1 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Neuroglía/citología , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores Notch/metabolismo , Factores de Transcripción/metabolismo
12.
J Biol Chem ; 290(39): 23725-37, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26269591

RESUMEN

Astrocytic glutamate transporter excitatory amino acid transporter (EAAT) 1, also known as glutamate aspartate transporter (GLAST) in rodents, is one of two glial glutamate transporters that are responsible for removing excess glutamate from synaptic clefts to prevent excitotoxic neuronal death. Despite its important role in neurophysiological functions, the molecular mechanisms of EAAT1 regulation at the transcriptional level remain to be established. Here, we report that NF-κB is a main positive transcription factor for EAAT1, supported by the following: 1) EAAT1 contains two consensus sites for NF-κB, 2) mutation of NF-κB binding sites decreased EAAT1 promoter activity, and 3) activation of NF-κB increased, whereas inhibition of NF-κB decreased EAAT1 promoter activity and mRNA/protein levels. EGF increased EAAT1 mRNA/protein levels and glutamate uptake via NF-κB. The transcription factor yin yang 1 (YY1) plays a role as a critical negative regulator of EAAT1, supported by the following: 1) the EAAT1 promoter contains multiple consensus sites for YY1, 2) overexpression of YY1 decreased EAAT1 promoter activity and mRNA/protein levels, and 3) knockdown of YY1 increased EAAT1 promoter activity and mRNA/protein levels. Manganese decreased EAAT1 expression via YY1. Epigenetic modifiers histone deacetylases (HDACs) served as co-repressors of YY1 to further decrease EAAT1 promoter activity, whereas inhibition of HDACs reversed manganese-induced decrease of EAAT1 expression. Taken together, our findings suggest that NF-κB is a critical positive regulator of EAAT1, mediating the stimulatory effects of EGF, whereas YY1 is a negative regulator of EAAT1 with HDACs as co-repressors, mediating the inhibitory effects of manganese on EAAT1 regulation.


Asunto(s)
Astrocitos/metabolismo , Epigénesis Genética/fisiología , Transportador 1 de Aminoácidos Excitadores/biosíntesis , FN-kappa B/metabolismo , Transcripción Genética/fisiología , Factor de Transcripción YY1/metabolismo , Animales , Astrocitos/citología , Células Cultivadas , Epigénesis Genética/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Manganeso/metabolismo , Manganeso/farmacología , Ratas , Ratas Sprague-Dawley , Elementos de Respuesta/fisiología , Transcripción Genética/efectos de los fármacos
13.
Cell Death Dis ; 6: e1693, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25789968

RESUMEN

Glaucoma is the second leading cause of blindness worldwide and is characterized by gradual visual impairment owing to progressive loss of retinal ganglion cells (RGCs) and their axons. Glutamate excitotoxicity has been implicated as a mechanism of RGC death in glaucoma. Consistent with this claim, we previously reported that glutamate/aspartate transporter (GLAST)-deficient mice show optic nerve degeneration that is similar to that observed in glaucoma. Therefore, drugs that upregulate GLAST may be useful for neuroprotection in glaucoma. Although many compounds are known to increase the expression of another glial glutamate transporter, EAAT2/GLT1, few compounds are shown to increase GLAST expression. Arundic acid is a glial modulating agent that ameliorates delayed ischemic brain damage by attenuating increases in extracellular glutamate. We hypothesized that arundic acid neuroprotection involves upregulation of GLAST. To test this hypothesis, we examined the effect of arundic acid on GLAST expression and glutamate uptake. We found that arundic acid induces GLAST expression in vitro and in vivo. In addition, arundic acid treatment prevented RGC death by upregulating GLAST in heterozygous (GLAST(+/-)) mice. Furthermore, arundic acid stimulates the human GLAST ortholog, EAAT1, expression in human neuroglioblastoma cells. Thus, discovering compounds that can enhance EAAT1 expression and activity may be a novel strategy for therapeutic treatment of glaucoma.


Asunto(s)
Caprilatos/administración & dosificación , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 1 de Aminoácidos Excitadores/genética , Glaucoma/genética , Degeneración Nerviosa/genética , Animales , Muerte Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glaucoma/tratamiento farmacológico , Glaucoma/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Ácido Glutámico/metabolismo , Humanos , Ratones , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/metabolismo , Nervio Óptico/patología , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/patología
14.
Neurochem Int ; 79: 33-43, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25454285

RESUMEN

The glutamate transporters GLAST and GLT-1 are mainly expressed in glial cells and regulate glutamate levels in the synapses. GLAST and GLT-1 are the targets of several signaling pathways. In this study we explore the possible functional interaction between these transporters and GSK3ß. This kinase is involved in multiple cellular processes including neuronal development and synaptic plasticity. To evaluate whether GLT-1 and GLAST were regulated by GSK3ß, we coexpressed these proteins in heterologous expression systems. In both COS-7 cells and Xenopus laevis oocytes, GSK3ß stimulated the activity of GLT-1 and reduced that of GLAST. These effects were associated with corresponding changes in the amounts of GLT-1 or GLAST in the plasma membrane. These effects were suppressed by inhibitors of GSK3ß or a catalytically inactive form of the kinase. GSK3ß also decreases the incorporation of (32)Pi into GLT-1 and increases GLAST phosphorylation. Pharmacological inhibition of endogenous GSK3ß in primary cultures of rat brain cortex also leads to a differential modulation of GLT-1 and GLAST. Our results suggest that constitutively active GSK3ß is important in controlling the expression of functional glutamate transporters on the plasma membrane. This regulation might be relevant in physiological and pathological conditions in which glutamate transporters and GSK3ß signaling are involved.


Asunto(s)
Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Glucógeno Sintasa Quinasa 3/metabolismo , Animales , Biotinilación , Células COS , Chlorocebus aethiops , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/genética , Regulación de la Expresión Génica/fisiología , Ácido Glutámico/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Oocitos/metabolismo , Técnicas de Placa-Clamp , Cultivo Primario de Células , Ratas , Xenopus
15.
Oncotarget ; 5(19): 8853-68, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25326682

RESUMEN

Signaling through glutamate receptors has been reported in human cancers, but the molecular mechanisms are not fully delineated. We report that in hepatocellular carcinoma and clear cell renal carcinoma cells, increased activity of hypoxia-inducible factors (HIFs) due to hypoxia or VHL loss-of-function, respectively, augmented release of glutamate, which was mediated by HIF-dependent expression of the SLC1A1 and SLC1A3 genes encoding glutamate transporters. In addition, HIFs coordinately regulated expression of the GRIA2 and GRIA3 genes, which encode glutamate receptors. Binding of glutamate to its receptors activated SRC family kinases and downstream pathways, which stimulated cancer cell proliferation, apoptosis resistance, migration and invasion in different cancer cell lines. Thus, coordinate regulation of glutamate transporters and receptors by HIFs was sufficient to activate key signal transduction pathways that promote cancer progression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Carcinoma Hepatocelular/genética , Carcinoma de Células Renales/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Hepáticas/genética , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular/genética , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 3 de Aminoácidos Excitadores/biosíntesis , Regulación Neoplásica de la Expresión Génica , Ácido Glutámico/metabolismo , Células HEK293 , Xenoinjertos , Humanos , Neoplasias Renales/genética , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-fyn/biosíntesis , Interferencia de ARN , ARN Interferente Pequeño , Receptores AMPA/biosíntesis , Transducción de Señal/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
16.
Br J Pharmacol ; 171(23): 5417-30, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25052197

RESUMEN

BACKGROUND AND PURPOSE: Astrocytic excitatory amino acid transporters (EAATs) regulate extracellular glutamate concentrations and play a role in preventing neuroexcitotoxicity. As the δ-opioid receptor (DOP receptor) is neuroprotective against excitotoxic injury, we determined whether DOP receptor activation up-regulates EAAT expression and function. EXPERIMENTAL APPROACH: We measured mRNA and protein expression of EAAT1, EAAT2 and EAAT3 in cultured mouse astrocytes exposed to a specific DOP receptor agonist (UFP-512) with or without a DOP receptor antagonist, DOP receptor siRNA or inhibitors of PKC, PKA, PI3K, p38, MAPK, MEK and ERK, and evaluated the function of EAATs by measuring glutamate uptake. KEY RESULTS: Astrocytic DOP receptor mRNA and protein were suppressed by DOP receptor siRNA knockdown. DOP receptor activation increased mRNA and protein expression of EAAT1 and EAAT2, but not EAAT3, thereby enhancing glutamate uptake of astrocytes. DOP receptor-induced EAAT1 and EAAT2 expression was largely reversed by DOP receptor antagonist naltrindole or by DOP receptor siRNA knockdown, and suppressed by inhibitors of MEK, ERK and p38. DOP receptor-accelerated glutamate uptake was inhibited by EAAT blockers, DOP receptor siRNA knockdown or inhibitors of MEK, ERK or p38. In contrast, inhibitors of PKA, PKC or PI3K had no significant effect on DOP receptor-induced EAAT expression. CONCLUSIONS AND IMPLICATIONS: DOP receptor activation up-regulates astrocytic EAATs via MEK-ERK-p38 signalling, suggesting a critical role for DOP receptors in the regulation of astrocytic EAATs and protection against neuroexcitotoxicity. As decreased EAAT expression contributes to pathophysiology in many neurological diseases, including amyotrophic lateral sclerosis, our findings present a new platform for potential treatments of these diseases.


Asunto(s)
Astrocitos/metabolismo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Receptores Opioides delta/metabolismo , Animales , Astrocitos/efectos de los fármacos , Bencimidazoles/farmacología , Células Cultivadas , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/genética , Transportador 3 de Aminoácidos Excitadores/metabolismo , Ratones Endogámicos C57BL , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Oligopéptidos/farmacología , ARN Interferente Pequeño/genética , Receptores Opioides delta/agonistas , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/genética , Regulación hacia Arriba
17.
Stem Cells Dev ; 23(23): 2851-61, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25036385

RESUMEN

MicroRNAs (miRNAs) are potential therapeutic targets in a variety of pathological conditions in the brain; however, their clinical application is hampered by lack of efficient delivery modes. Mesenchymal stromal stem cells (MSCs) migrate to sites of injury and inflammation and exert therapeutic effects in various neurological disorders. Here, we examined the ability of MSCs to deliver exogenous miRNA mimics and pre-miRNAs to human neural progenitor cells (NPCs) and astrocytes and characterized the functional impact of this delivery. We found that MSCs efficiently delivered fluorescent-labeled miR-124 and miR-145 mimics to cocultured NPCs and astrocytes. We further demonstrated the delivery of the miRNAs using novel reporter plasmids that contain a sequence complementary to miR-124 or miR-145 downstream of luciferase or mCherry. Binding of the specific miRNAs to these sequences results in decreased luciferase activity or mCherry fluorescence and therefore enable analysis of miRNA delivery in living cells. The delivered exogenous miR-124 significantly decreased the expression of the target gene Sox9 by targeting its 3'-UTR, and increased the neuronal differentiation of the NPCs. In addition, the delivered miR-124 increased the expression of the glutamate transporters, EAAT1 in NPCs and EAAT2 in both NPCs and astrocytes. Similar results were obtained with MSCs transfected with pre-miR-124. The miRNA delivery was mediated by MSC-derived exosomes and was cell contact independent. These results suggest that MSCs can functionally deliver exogenous miRNAs to neural cells and provide an efficient route of therapeutic miRNA delivery to the brain in pathological conditions with clinical implications for regenerative medicine.


Asunto(s)
Transportador 1 de Aminoácidos Excitadores/biosíntesis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/biosíntesis , Células Madre Mesenquimatosas/metabolismo , MicroARNs , Células-Madre Neurales/metabolismo , Regiones no Traducidas 3' , Diferenciación Celular , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores , Regulación de la Expresión Génica/genética , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Humanos , Células Madre Mesenquimatosas/citología , MicroARNs/genética , MicroARNs/metabolismo , Células-Madre Neurales/citología , Factor de Transcripción SOX9/biosíntesis
18.
Pharmacogenomics ; 15(7): 925-32, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24956246

RESUMEN

AIM: To evaluate the effect of functional polymorphisms (rs4354668 and rs2731880) of the excitatory amino acid transporters (EAAT1 and 2) on the cognitive dysfunction that characterizes schizophrenia. MATERIALS & METHODS: One hundred and ninety two subjects diagnosed with schizophrenia were assessed with Brief Assessment of Cognition in Schizophrenia, Wisconsin Card Sorting Test, Continuous Performance Test and N-back test and genotyped for rs4354668 and rs2731880. RESULTS: ANOVA showed a significant difference among both EAAT1 and EAAT2 genotype groups on different cognitive measures. Worse performances were observed among carriers of the genotypes associated with lower EAAT expression. CONCLUSION: RESULTS suggest that impaired activity and EAAT expression could influence cognitive performances in schizophrenia, thus representing a target of interest for development of pharmacological strategies aimed to improve cognition.


Asunto(s)
Trastornos del Conocimiento/genética , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/biosíntesis , Esquizofrenia/genética , Adulto , Trastornos del Conocimiento/complicaciones , Trastornos del Conocimiento/patología , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores , Femenino , Genotipo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Esquizofrenia/complicaciones , Esquizofrenia/patología
19.
J Neurochem ; 128(5): 617-27, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24164438

RESUMEN

In vitro and in vivo studies have suggested that reduced astrocytic uptake of neuronally released glutamate, alterations in expression of glial fibrillary acidic protein (GFAP) and aquaporin-4 (AQP-4) contribute to brain edema in acute liver failure (ALF). However, there is no evidence to date to suggest that these alterations occur in patients with ALF. We analyzed the mRNA expression of excitatory amino acid transporters (EAAT-1, EAAT-2), GFAP, and AQP-4 in the cerebral cortex obtained at autopsy from eight patients with ALF and from seven patients with no evidence of hepatic or neurological disorders by real-time PCR, and protein expression was assessed using immunoblotting and immunohistochemistry. We demonstrated a significant decrease in GFAP mRNA and protein levels in ALF patients compared to controls. While the loss of EAAT-2 protein in ALF samples was post-translational in nature, EAAT-1 protein remained within normal limits. Immunohistochemistry confirmed that, in all cases, the losses of EAAT-2 and GFAP were uniquely astrocytic in their localization. AQP-4 mRNA expression was significantly increased and its immunohistochemistry demonstrated increased AQP-4 immunoreactivity in the glial end-feet process surrounding the microvessels. These findings provide evidence of selective alterations in the expression of genes coding for key astrocytic proteins implicated in central nervous system (CNS) excitability and brain edema in human ALF. We investigated the gene expression of astrocytic proteins involved in astrocyte swelling causing brain edema in autopsied brain tissues of patients with acute liver failure. This study demonstrated loss of GFAP expression and up-regulation of AQP-4 protein expression leading to cerebral edema, and loss of EAAT-2 expression implicated in excitatory neurotransmission. These findings may provide new drug targets against CNS complications of acute liver failure.


Asunto(s)
Astrocitos/metabolismo , Edema Encefálico/genética , Expresión Génica/fisiología , Fallo Hepático Agudo/genética , Neuronas/fisiología , Adolescente , Adulto , Anciano , Acuaporina 4/metabolismo , Western Blotting , Edema Encefálico/metabolismo , Edema Encefálico/patología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , ADN Complementario/biosíntesis , ADN Complementario/genética , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Transporte de Glutamato en la Membrana Plasmática/biosíntesis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/genética , Humanos , Inmunohistoquímica , Fallo Hepático Agudo/metabolismo , Fallo Hepático Agudo/patología , Masculino , Persona de Mediana Edad , ARN/biosíntesis , ARN/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
20.
Neuroscience ; 244: 113-21, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23602887

RESUMEN

Glutamate plays a central role in brain physiology and pathology. The involvement of excitatory amino acid transporters (EAATs) in neurodegenerative disorders including acute stroke has been widely studied, but little is known about the role of glial glutamate transporters in white matter injury after chronic cerebral hypoperfusion. The present study evaluated the expression of glial (EAAT1 and EAAT2) and neuronal (EAAT3) glutamate transporters in subcortical white matter and cortex, before and 3-28 days after the ligation of bilateral common carotid arteries (LBCCA) in rat brain. K-B staining showed a gradual increase of demyelination in white matter after ischemia, while there was no cortical involvement. Between 3 and 7 days after LBCCA, a significant increase in EAAT2 protein levels was observed in the ischemic brain and the number of EAAT2-positive cells also significantly increased both in the cortical and white matter lesions. EAAT2 was detected in glial-fibrillary acidic protein (GFAP)-positive astrocytes in both the cortex and white matter, but not in neuronal and oligodendroglial cells. EAAT1 was slightly elevated after ischemia only in the white matter, but EAAT3 was at almost similar levels both in the cortex and white matter after ischemia. A significant increase in EAAT2 expression level was also noted in the deep white matter of chronic human ischemic brain tissue compared to the control group. Our findings suggest important roles for up-regulated EAAT2 in chronic brain ischemia especially in the regulation of high-affinity of extracellular glutamate and minimization of white matter damage.


Asunto(s)
Isquemia Encefálica/metabolismo , Cuerpo Calloso/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Proteínas de Transporte de Glutamato en la Membrana Plasmática/biosíntesis , Fibras Nerviosas Mielínicas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/metabolismo , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Estudios de Casos y Controles , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Enfermedad Crónica , Cuerpo Calloso/patología , Enfermedades Desmielinizantes/complicaciones , Enfermedades Desmielinizantes/metabolismo , Transportador 1 de Aminoácidos Excitadores/biosíntesis , Transportador 3 de Aminoácidos Excitadores/biosíntesis , Femenino , Humanos , Masculino , Fibras Nerviosas Mielínicas/patología , Neuronas/metabolismo , Oligodendroglía/metabolismo , Ratas , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA