Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
1.
Neuroscience ; 552: 89-99, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38909675

RESUMEN

Chronic ethanol consumption increased extracellular glutamate concentrations in several reward brain regions. Glutamate homeostasis is regulated in majority by astrocytic glutamate transporter 1 (GLT-1) as well as the interactive role of cystine/glutamate antiporter (xCT). In this study, we aimed to determine the attenuating effects of a novel beta-lactam MC-100093, lacking the antibacterial properties, on ethanol consumption and GLT-1 and xCT expression in the subregions of nucleus accumbens (NAc core and NAc shell) and medial prefrontal cortex (Infralimbic, mPFC-IL and Prelimbic, mPFC-PL) in male and female alcohol-preferring (P) rats. Female and male rats were exposed to free access to ethanol (15% v/v) and (30% v/v) and water for five weeks, and on Week 6, rats were administered 100 mg/kg (i.p) of MC-100093 or saline for five days. MC-100093 reduced ethanol consumption in both male and female P rats from Day 1-5. Additionally, MC-100093 upregulated GLT-1 and xCT expression in the mPFC and NAc subregions as compared to ethanol-saline groups in female and male rats. Chronic ethanol intake reduced GLT-1 and xCT expression in the IL and PL in female and male rats, except there was no reduction in GLT-1 expression in the mPFC-PL in female rats. Although, MC-100093 upregulated GLT-1 and xCT expression in the subregions of NAc, we did not observe any reduction in GLT-1 and xCT expression with chronic ethanol intake in female rats. These findings strongly suggest that MC-100093 treatment effectively reduced ethanol intake and upregulated GLT-1 and xCT expression in the mPFC and NAc subregions in male and female P rats.


Asunto(s)
Consumo de Bebidas Alcohólicas , Astrocitos , Etanol , Transportador 2 de Aminoácidos Excitadores , Núcleo Accumbens , Animales , Femenino , Masculino , Consumo de Bebidas Alcohólicas/metabolismo , Ratas , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Etanol/administración & dosificación , Etanol/farmacología , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efectos de los fármacos , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Depresores del Sistema Nervioso Central/administración & dosificación , Caracteres Sexuales
2.
J Neuroinflammation ; 21(1): 130, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38750510

RESUMEN

Epidemiological studies have unveiled a robust link between exposure to repetitive mild traumatic brain injury (r-mTBI) and elevated susceptibility to develop neurodegenerative disorders, notably chronic traumatic encephalopathy (CTE). The pathogenic lesion in CTE cases is characterized by the accumulation of hyperphosphorylated tau in neurons around small cerebral blood vessels which can be accompanied by astrocytes that contain phosphorylated tau, the latter termed tau astrogliopathy. However, the contribution of tau astrogliopathy to the pathobiology and functional consequences of r-mTBI/CTE or whether it is merely a consequence of aging remains unclear. We addressed these pivotal questions by utilizing a mouse model harboring tau-bearing astrocytes, GFAPP301L mice, subjected to our r-mTBI paradigm. Despite the fact that r-mTBI did not exacerbate tau astrogliopathy or general tauopathy, it increased phosphorylated tau in the area underneath the impact site. Additionally, gene ontology analysis of tau-bearing astrocytes following r-mTBI revealed profound alterations in key biological processes including immunological and mitochondrial bioenergetics. Moreover, gene array analysis of microdissected astrocytes accrued from stage IV CTE human brains revealed an immunosuppressed astroglial phenotype similar to tau-bearing astrocytes in the GFAPP301L model. Additionally, hippocampal reduction of proteins involved in water transport (AQP4) and glutamate homeostasis (GLT1) was found in the mouse model of tau astrogliopathy. Collectively, these findings reveal the importance of understanding tau astrogliopathy and its role in astroglial pathobiology under normal circumstances and following r-mTBI. The identified mechanisms using this GFAPP301L model may suggest targets for therapeutic interventions in r-mTBI pathogenesis in the context of CTE.


Asunto(s)
Acuaporina 4 , Astrocitos , Transportador 2 de Aminoácidos Excitadores , Ratones Transgénicos , Tauopatías , Proteínas tau , Astrocitos/metabolismo , Astrocitos/patología , Animales , Ratones , Proteínas tau/metabolismo , Proteínas tau/genética , Acuaporina 4/metabolismo , Acuaporina 4/genética , Tauopatías/metabolismo , Tauopatías/patología , Tauopatías/genética , Humanos , Transportador 2 de Aminoácidos Excitadores/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Conmoción Encefálica/metabolismo , Conmoción Encefálica/patología , Masculino , Fenotipo , Ratones Endogámicos C57BL
3.
BMC Neurosci ; 22(1): 54, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34521349

RESUMEN

BACKGROUND: Ceftriaxone is a ß-lactam antibiotic used to treat central nervous system infections. Whether the neuroprotective effects of ceftriaxone after TBI are mediated by attenuating neuroinflammation but not its antibacterial actions is not well established. METHODS: Anesthetized male Sprague-Dawley rats were divided into sham-operated, TBI + vehicle, and TBI + ceftriaxone groups. Ceftriaxone was intraperitoneally injected at 0, 24, and 48 h with 50 or 250 mg/kg/day after TBI. During the first 120 min after TBI, we continuously measured heart rate, arterial pressure, intracranial pressure (ICP), and cerebral perfusion pressure. The infarct volume was measured by TTC staining. Motor function was measured using the inclined plane. Glutamate transporter 1 (GLT-1), neuronal apoptosis and TNF-α expression in the perilesioned cortex were investigated using an immunofluorescence assay. Bacterial evaluation was performed by Brown and Brenn's Gram staining. These parameters above were measured at 72 h after TBI. RESULTS: Compared with the TBI + vehicle group, the TBI + ceftriaxone 250 mg/kg group showed significantly lower ICP, improved motor dysfunction, reduced body weight loss, decreased infarct volume and neuronal apoptosis, decreased TBI-induced microglial activation and TNF-α expression in microglia, and increased GLT-1 expression in neurons and microglia. However, the grades of histopathological changes of antibacterial effects are zero. CONCLUSIONS: The intraperitoneal injection of ceftriaxone with 250 mg/kg/day for three days may attenuate TBI by increasing GLT-1 expression and reducing neuroinflammation and neuronal apoptosis, thereby resulting in an improvement in functional outcomes, and this neuroprotective effect is not related to its antibacterial effects.


Asunto(s)
Antibacterianos , Antiinflamatorios/uso terapéutico , Lesiones Traumáticas del Encéfalo/metabolismo , Ceftriaxona/uso terapéutico , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Animales , Antiinflamatorios/farmacología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Ceftriaxona/farmacología , Relación Dosis-Respuesta a Droga , Transportador 2 de Aminoácidos Excitadores/agonistas , Masculino , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley
4.
Brain Res Bull ; 175: 224-233, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34343641

RESUMEN

Our previous finding suggests that p38 MAPK contributes to the GLT-1 upregulation during induction of brain ischemic tolerance by cerebral ischemic preconditioning (CIP), however, the underlying mechanism is still unclear. Here, we investigated the molecular mechanisms underlying the CIP-induced GLT-1 upregulation by using Western blotting, Co-immunoprecipitation (Co-IP), electrophoretic mobility shift assay (EMSA) and thionin staining in rat hippocampus CA1 subset. We found that application of BAY11-7082 (an inhibitor of NF-κB), or dihydrokainate (an inhibitor of GLT-1), or SB203580 (an inhibitor of p38 MAPK) could attenuate the CIP-induced neuronal protection in hippocampus CA1 region of rats. Moreover, CIP caused rapid activation of NF-κB, as evidenced by nuclear translocation of NF-κB p50 protein, which led to active p50/p65 dimer formation and increased DNA binding activity. GLT-1 was also increased after CIP. Pretreatment with BAY11-7082 blocked the CIP-induced GLT-1 upregulation. The above results suggest that NF-κB participates in GLT-1 up-regulation during the induction of brain ischemic tolerance by CIP. We also found that pretreatment with SB203580 caused significant reduction of NF-κB p50 protein in nucleus, NF-κB p50/p65 dimer nuclear translocation and DNA binding activity of NF-κB. Together, we conclude that p38 MAPK/NF-κB pathway participates in the mediation of GLT-1 up-regulation during the induction of brain ischemic tolerance induced by CIP.


Asunto(s)
Isquemia Encefálica/genética , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Precondicionamiento Isquémico , Sistema de Señalización de MAP Quinasas/genética , FN-kappa B/genética , Animales , Región CA1 Hipocampal/patología , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Imidazoles/farmacología , Ácido Kaínico/análogos & derivados , Ácido Kaínico/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , FN-kappa B/antagonistas & inhibidores , Subunidad p50 de NF-kappa B/metabolismo , Neuroprotección , Nitrilos/farmacología , Piridinas/farmacología , Ratas , Ratas Wistar , Sulfonas/farmacología , Factor de Transcripción ReIA/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
5.
Neurotoxicology ; 86: 94-103, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34310962

RESUMEN

Dysregulation of the astrocytic glutamate transporter excitatory amino acid transporter 2 (EAAT2) is associated with several neurological disorders, including Parkinson's disease, Alzheimer's disease, and manganism, the latter induced by chronic exposure to high levels of manganese (Mn). Mechanisms of Mn-induced neurotoxicity include impairment of EAAT2 function secondary to the activation of the transcription factor Yin Yang 1 (YY1) by nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). However, the upstream mechanisms by which Mn-induced NF-κB activates YY1 remain to be elucidated. In the present study, we used the H4 human astrocyte cell line to test if Mn activates YY1 through the canonical NF-κB signaling pathway, leading to EAAT2 repression. The results demonstrate that Mn exposure induced phosphorylation of the upstream kinase IκB kinase (IKK-ß), leading to NF-κB p65 translocation, increased YY1 promoter activity, mRNA/protein levels, and consequently repressed EAAT2. Results also demonstrated that Mn-induced oxidative stress and subsequent TNF-α production were upstream of IKK-ß activation, as antioxidants attenuated Mn-induced TNF-α production and IKK-ß activation. Moreover, TNF-α inhibition attenuated the Mn-induced activation of IKK-ß and YY1. Taken together, Mn-induced oxidative stress and TNF-α mediates activation of NF-κB signaling and YY1 upregulation, leading to repression of EAAT2. Thus, targeting reactive oxygen species (ROS), TNF-α and IKK-ß may attenuate Mn-induced YY1 activation and consequent EAAT2 repression.


Asunto(s)
Astrocitos/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Quinasa I-kappa B/metabolismo , Manganeso/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción YY1/biosíntesis , Astrocitos/efectos de los fármacos , Células Cultivadas , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Humanos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
6.
Mol Neurobiol ; 58(7): 3443-3456, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33723767

RESUMEN

Ketamine and MK-801 by blocking NMDA receptors may induce reinforcing effects as well as schizophrenia-like symptoms. Recent results showed that ketamine can also effectively reverse depressive signs in patients' refractory to standard therapies. This evidence clearly points to the need of characterization of effects of these NMDARs antagonists on relevant brain areas for mood disorders. The aim of the present study was to investigate the molecular changes occurring at glutamatergic synapses 24 h after ketamine or MK-801 treatment in the rat medial prefrontal cortex (mPFC) and hippocampus (Hipp). In particular, we analyzed the levels of the glutamate transporter-1 (GLT-1), NMDA receptors, AMPA receptors subunits, and related scaffolding proteins. In the homogenate, we found a general decrease of protein levels, whereas their changes in the post-synaptic density were more complex. In fact, ketamine in the mPFC decreased the level of GLT-1 and increased the level of GluN2B, GluA1, GluA2, and scaffolding proteins, likely indicating a pattern of enhanced excitability. On the other hand, MK-801 only induced sparse changes with apparently no correlation to functional modification. Differently from mPFC, in Hipp, both substances reduced or caused no changes of glutamate receptors and scaffolding proteins expression. Ketamine decreased NMDA receptors while increased AMPA receptors subunit ratios, an effect indicative of permissive metaplastic modulation; conversely, MK-801 only decreased the latter, possibly representing a blockade of further synaptic plasticity. Taken together, these findings indicate a fine tuning of glutamatergic synapses by ketamine compared to MK-801 both in the mPFC and Hipp.


Asunto(s)
Maleato de Dizocilpina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/efectos de los fármacos , Ketamina/farmacología , Corteza Prefrontal/efectos de los fármacos , Receptores de Glutamato/biosíntesis , Animales , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Expresión Génica , Hipocampo/metabolismo , Masculino , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley
7.
Cereb Cortex ; 31(4): 2026-2037, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33279960

RESUMEN

Visuospatial working memory (vsWM) requires information transfer among multiple cortical regions, from primary visual (V1) to prefrontal (PFC) cortices. This information is conveyed via layer 3 glutamatergic neurons whose activity is regulated by gamma-aminobutyric acid (GABA)ergic interneurons. In layer 3 of adult human neocortex, molecular markers of glutamate neurotransmission were lowest in V1 and highest in PFC, whereas GABA markers had the reverse pattern. Here, we asked if these opposite V1-visual association cortex (V2)-posterior parietal cortex (PPC)-PFC gradients across the vsWM network are present in layer 3 of monkey neocortex, when they are established during postnatal development, and if they are specific to this layer. We quantified transcript levels of glutamate and GABA markers in layers 3 and 6 of four vsWM cortical regions in a postnatal developmental series of 30 macaque monkeys. In adult monkeys, glutamate transcript levels in layer 3 increased across V1-V2-PPC-PFC regions, whereas GABA transcripts showed the opposite V1-V2-PPC-PFC gradient. Glutamate transcripts established adult-like expression patterns earlier during postnatal development than GABA transcripts. These V1-V2-PPC-PFC gradients and developmental patterns were less evident in layer 6. These findings demonstrate that expression of glutamate and GABA transcripts differs across cortical regions and layers during postnatal development, revealing potential molecular substrates for vsWM functional maturation.


Asunto(s)
Ácido Glutámico/biosíntesis , Lóbulo Parietal/metabolismo , Corteza Prefrontal/metabolismo , Transcripción Genética/fisiología , Corteza Visual/metabolismo , Ácido gamma-Aminobutírico/biosíntesis , Factores de Edad , Animales , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Femenino , Neuronas GABAérgicas/metabolismo , Expresión Génica , Ácido Glutámico/genética , Macaca mulatta , Lóbulo Parietal/crecimiento & desarrollo , Corteza Prefrontal/crecimiento & desarrollo , Receptores de GABA-A/biosíntesis , Receptores de GABA-A/genética , Corteza Visual/crecimiento & desarrollo , Ácido gamma-Aminobutírico/genética
8.
Eur Rev Med Pharmacol Sci ; 24(22): 11783-11792, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33275249

RESUMEN

OBJECTIVE: This study aimed to explore the correlation between serum EAAT2 and ADORA2A levels and Alzheimer's disease (AD). PATIENTS AND METHODS: A total of 68 patients with AD treated in our hospital from April 2017 to January 2019 were enrolled and assigned to group A, and 60 healthy individuals undergoing physical examinations in the same period were enrolled and assigned to group B. Enzyme-linked immunosorbent assay (ELISA) was used to measure the expression of serum EAAT2 and ADORA2A in the two groups, receiver operating characteristic (ROC) curve to assess the predictive value of diagnostic efficacy, Spearman correlation to perform correlation analysis, and multivariate logistic analysis to analyze risk factors of prognosis. RESULTS: Patients from group A showed significantly higher serum ADORA2A level and lower serum EAAT2 level than individuals from group B (all p<0.001). The severity of AD was negatively correlated with the relative expression of serum EAAT2 (r=-0.7286, p<0.001), positively correlated with the relative expression of serum ADORA2A (r=0.7381, p<0.001). The sensitivity, specificity, and area under the curve (AUC) of EAAT2 alone for the diagnosis of AD were 85.00%, 82.35%, and 0.8853, respectively, and those of ADORA2A alone for the diagnosis of AD were 71.67%, 79.41.00%, and 0.8369, respectively. Univariate and multivariate Logistic regression analysis showed that disease severity, EAAT2, and ADORA2A were independent risk factors of the prognosis of AD. CONCLUSIONS: Patients with AD have highly expressed ADORA2A and lowly expressed EAAT2 in the serum. EAAT2 and ADORA2A may play parts in the progression of AD, and they can act as potential serum biomarkers for the diagnosis and disease assessment of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Receptor de Adenosina A2A/biosíntesis , Anciano , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/diagnóstico , Ensayo de Inmunoadsorción Enzimática , Transportador 2 de Aminoácidos Excitadores/sangre , Femenino , Humanos , Masculino , Pronóstico , Curva ROC , Receptor de Adenosina A2A/sangre
9.
Neuropharmacology ; 181: 108339, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33010299

RESUMEN

Lead (Pb) is a neurotoxic heavy metal pollutant. Despite the efforts to reduce Pb environmental exposure and to prevent Pb poisoning, exposure in human populations persists. Studies of adults with history of childhood lead exposure have consistently demonstrated cognitive impairments that have been associated with sustained glutamate signaling. Additionally, some clinical studies have also found correlations between Pb exposure and increased proclivity to drug addiction. Thus, here we sought to investigate if developmental Pb exposure can increase propensity to alcohol consumption and relapse using an alcohol self-administration paradigm. Because Pb exposure is associated with increased glutamatergic tone, we also studied the effects on the expression of synaptic and non-synaptic glutamate transporters in brain regions associated with drug addiction such as the nucleus accumbens (NAc), dorsomedial striatum (DMS), dorsolateral striatum (DLS), and medial prefrontal cortex (mPFC). We found that while developmental Pb exposure did not increase risk for alcohol self-administration, it did play a role in relapsing to alcohol. The effects were associated with differential expression of the glutamate transporter 1 (GLT1) and the glutamate/cystine antiporter (xCT). In the NAc and DLS the expression of GLT1 was found to be significantly reduced, while no changes were found in DMS or mPFC. Contrastingly, xCT was found to be upregulated in NAc but downregulated in DLS, with no changes in DMS or mPFC. Our data suggest that lead exposure is involved in relapse to alcohol seeking, an effect that could be associated with downregulation of GLT1 and xCT in the DLS.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/biosíntesis , Depresores del Sistema Nervioso Central/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Etanol/farmacología , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Intoxicación por Plomo/psicología , Autoadministración , Sistema de Transporte de Aminoácidos y+/genética , Animales , Química Encefálica/efectos de los fármacos , Transportador 2 de Aminoácidos Excitadores/genética , Extinción Psicológica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Recurrencia
10.
J BUON ; 25(4): 2051-2058, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33099952

RESUMEN

PURPOSE: The purpose of our study was to investigate the mRNA expression profile of glutamate transporter 1 (GLT-1) in different types and grades of brain tumors, such as glioblastoma multiforme, astrocytomas (pilocytic, diffuse, anaplastic), oligodendrogliomas, ependydomas, medulloblastomas, and meningiomas using Real Time Quantitative PCR technique (qRT-PCR). METHODS: A total of 66 surgically removed primary brain tumors were collected retrospectively and the total RNA was isolated from each tumor sample. cDNA was generated and GLT-1 mRNA expression was evaluated with quantitative qRT-PCR. RESULTS: The mRNA expression of GLT-1 was significantly lower in primary brain tumors when compared to control brain tissues. GLT-1 expression was inversely correlated with the tumor grade, implicating its potential role in tumor progression. GLT-1 mRNA expression was lowest in grade 4 tumors, such as glioblastoma multiforme and medulloblastomas. The tumors with grade 3 and 4 combined displayed lower expression compared to tumors with grades 1 and 2. In grade 4 tumors, female patients displayed lower GLT-1 expression compared to male patients. In addition, glioblastoma multiforme patients older than 65 years of age showed lower GLT-1 expression when compared to the patients younger than 65. CONCLUSION: qRT-PCR was found to be a sensitive method in detecting GLT-1 expression in brain tumors. This study may lay the foundation for the future research about the excitotoxicity and brain tumors and GLT-1 might be a potential biomarker. Targeted therapies based on excitotoxic molecular pathways against gliomas should be designed to effectively combat these diseases.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Glioblastoma/metabolismo , Adulto , Factores de Edad , Anciano , Neoplasias Encefálicas/patología , Transportador 2 de Aminoácidos Excitadores/genética , Femenino , Glioblastoma/patología , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Estudios Retrospectivos , Factores Sexuales , Adulto Joven
11.
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
12.
Brain Res Bull ; 163: 49-56, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32621862

RESUMEN

A pivotal role of glutamatergic neurotransmission in the pathophysiology of major depressive disorder (MDD) has been supported in preclinical and clinical studies. Glutamate transporters are responsible for rapid uptake of glutamate to maintain glutamate homeostasis. Down-regulation of glutamate transporters has been reported in MDD patients and animal models. However, the mechanism for stress-induced modulation of glutamate transporter expression is poorly understood. Receptor for advanced glycosylation end products (RAGE), a member of immunoglobulin family, is found expressed widely in brain and play important roles in neuronal development, neurite growth, neurogenesis and neuroinflammation. Our study showed chronic unpredictable stress (CUS) induced depressive-like behaviors and reduced RAGE expression in hippocampus DG, CA1 and CA3 areas. The protein levels of GLT-1, p-CREB and p-p65 decreased in hippocampus DG as well. Knockdown of RAGE expression in hippocampus DG with RAGE shRNA lentivirus particles induced depressive-like behaviors. Meanwhile, the protein and mRNA levels of GLT-1 were significantly decreased as well as phosphorylation of CREB and p65. Neither CUS nor RAGE knockdown altered GLAST protein and mRNA levels. These findings suggested that RAGE/CREB-NF-κB signaling pathway in hippocampus DG involved in modulation of GLT-1 expression, which possibly contributed to the depressive-like behavior induced by CUS.


Asunto(s)
Giro Dentado/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Receptor para Productos Finales de Glicación Avanzada/deficiencia , Transducción de Señal/fisiología , Estrés Psicológico/metabolismo , Animales , Enfermedad Crónica , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Transportador 2 de Aminoácidos Excitadores/genética , Técnicas de Silenciamiento del Gen/métodos , Masculino , Ratas , Ratas Sprague-Dawley , Receptor para Productos Finales de Glicación Avanzada/genética , Estrés Psicológico/genética , Estrés Psicológico/psicología
13.
Neurochem Int ; 139: 104787, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32650029

RESUMEN

Astrocytes have diverse functions that are supported by their anatomic localization between neurons and blood vessels. One of these functions is the clearance of extracellular glutamate. Astrocytes clear glutamate using two Na+-dependent glutamate transporters, GLT-1 (also called EAAT2) and GLAST (also called EAAT1). GLT-1 expression increases during synaptogenesis and is a marker of astrocyte maturation. Over 20 years ago, several groups demonstrated that astrocytes in culture express little or no GLT-1 and that neurons induce expression. We recently demonstrated that co-culturing endothelia with mouse astrocytes also induced expression of GLT-1 and GLAST. These increases were blocked by an inhibitor of γ-secretase. This and other observations are consistent with the hypothesis that Notch signaling is required, but the ligands involved were not identified. In the present study, we used rat astrocyte cultures to further define the mechanisms by which endothelia induce expression of GLT-1 and GLAST. We found that co-cultures of astrocytes and endothelia express higher levels of GLT-1 and GLAST protein and mRNA. That endothelia activate Hes5, a transcription factor target of Notch, in astrocytes. Using recombinant Notch ligands, anti-Notch ligand neutralizing antibodies, and shRNAs, we provide evidence that both Dll1 and Dll4 contribute to endothelia-dependent regulation of GLT-1. We also provide evidence that astrocytes secrete a factor(s) that induces expression of Dll4 in endothelia and that this effect is required for Notch-dependent induction of GLT-1. Together these studies indicate that reciprocal communication between astrocytes and endothelia is required for appropriate astrocyte maturation and that endothelia likely deploy additional non-Notch signals to induce GLT-1.


Asunto(s)
Astrocitos/metabolismo , Comunicación Celular/fisiología , Células Endoteliales/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Animales , Células Cultivadas , Técnicas de Cocultivo , Transportador 2 de Aminoácidos Excitadores/genética , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratas , Ratas Sprague-Dawley
14.
Brain Res Bull ; 161: 136-146, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32433937

RESUMEN

Excitotoxicity is one of the main mechanisms related to hypoxia/reoxygenation (H/R) injury. Excitatory amino acid transporter (EAAT)2 mainly distributes on astrocytes and plays an important role on glutamate reuptake and glutamate homeostasis. Midazolam has a neuroprotective effect in some neuropathological conditions. The present study aimed to detect the role of EAAT2 in the neuroprotective effect of midazolam in neonatal rat brain subjected to H/R. Pretreatment with midazolam reversed H/R-induced apoptosis and downregulation of EAAT2 mRNA and protein expression in the hippocampus. Pretreatment with dihydrokainic acid (a selective inhibitor of EAAT2) exacerbated apoptosis, and thus inhibited the neuroprotective effect of midazolam against H/R injury. We demonstrated for the first time that dysregulation of EAAT2 expression may be related to the neural injury induced by H/R in rat pups, and pretreatment with midazolam attenuated apoptosis and improved learning and memory partly due to regulating EAAT2 expression.


Asunto(s)
Lesiones Encefálicas/metabolismo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Hipoxia Encefálica/metabolismo , Midazolam/administración & dosificación , Fármacos Neuroprotectores/administración & dosificación , Animales , Animales Recién Nacidos , Lesiones Encefálicas/prevención & control , Relación Dosis-Respuesta a Droga , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Hipnóticos y Sedantes/administración & dosificación , Hipoxia Encefálica/prevención & control , Ratas , Ratas Sprague-Dawley
15.
Glia ; 68(10): 2028-2039, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32170887

RESUMEN

Glial cells have a major role in protecting neurons against various forms of stress. Especially, astrocytes mediate the bulk of glutamate clearance in the brain via specific membrane transporters (GLAST and GLT1), thereby preventing the occurrence of excitotoxic events. Although glutamate-mediated mechanisms are thought to contribute to nigral dopaminergic neuron degeneration in Parkinson's disease, detailed information on the organization of glia in the substantia nigra is still lacking. The present study was performed to provide quantitative information on the organization of astroglia and on the relationships between astrocytes and excitatory synapses in the rat substantia nigra. Using immunolabeling of GLT1 and confocal imaging, we found that the substantia nigra was filled with a dense meshwork of immunoreactive astrocyte processes. Stereological analysis performed on electron microscope images revealed that the density of immunoreactive astrocyte plasma membranes was substantial, close to 1 µm2 /µm3 , in the substantia nigra neuropil, both in the pars compacta and the pars reticulata. Excitatory synapses had on average two thirds of their perimeters free from glia, a disposition that may favor transmitter spillover. The density of glutamatergic synapses, as quantified on confocal images by the simultaneous detection of bassoon and of vesicular glutamate transporter 1 or 2, was very low (0.01 and 0.025 per µm3 in the reticulata and compacta subdivisions, respectively). Thus the ratio of GLT1-expressing glial membrane surface to glutamatergic synapses was very high (40-100 µm2 ), suggesting an efficient regulation of extracellular glutamate concentrations.


Asunto(s)
Transportador 2 de Aminoácidos Excitadores/biosíntesis , Neuroglía/metabolismo , Sustancia Negra/metabolismo , Sinapsis/metabolismo , Animales , Transportador 2 de Aminoácidos Excitadores/ultraestructura , Masculino , Neuroglía/ultraestructura , Ratas , Ratas Wistar , Sustancia Negra/ultraestructura , Sinapsis/ultraestructura
16.
Mol Neurobiol ; 57(5): 2290-2300, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32008166

RESUMEN

The complement C1q plays a critical role in microglial phagocytosis of glutamatergic synapses and in the pathogenesis of neuroinflammation in Alzheimer's disease (AD). We recently reported that upregulation of metabotropic glutamate receptor signaling is associated with increased synaptic C1q production and subsequent microglial phagocytosis of synapses in the rodent models of AD. Here, we explored the role of astrocytic glutamate transporter in the synaptic C1q production and microglial phagocytosis of hippocampal glutamatergic synapses in a rat model of AD. Activation of astrocyte and reduction glutamate transporter 1 (GLT1) were noted after bilateral microinjection of amyloid-beta (Aß1-40) fibrils into the hippocampal CA1 area of rats. Ceftriaxone is a ß-lactam antibiotic that upregulates GLT1 expression. Bilateral microinjection of ceftriaxone recovered GLT1 expression, decreased synaptic C1q production, suppressed microglial phagocytosis of glutamatergic synapses in the hippocampal CA1, and attenuated synaptic and cognitive deficits in rats microinjected with Aß1-40. In contrast, artificial suppression of GLT1 activity by DL-threo-beta-benzyloxyaspartate (DL-TBOA) in naïve rats induced synaptic C1q expression and microglial phagocytosis of glutamatergic synapses in the hippocampal CA1 area, resulting in synaptic and cognitive dysfunction. These findings demonstrated that impairment of astrocytic glutamate transporter plays a role in the pathogenesis of AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/toxicidad , Astrocitos/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Trastornos del Conocimiento/inducido químicamente , Complemento C1q/fisiología , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Ácido Glutámico/fisiología , Microglía/fisiología , Neuronas/metabolismo , Fragmentos de Péptidos/toxicidad , Animales , Ácido Aspártico/farmacología , Astrocitos/metabolismo , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Ceftriaxona/farmacología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/patología , Complemento C1q/biosíntesis , Complemento C1q/genética , Modelos Animales de Enfermedad , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/fisiología , Masculino , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Prueba del Laberinto Acuático de Morris/fisiología , Técnicas de Placa-Clamp , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato/fisiología , Sinapsis/metabolismo , Regulación hacia Arriba/efectos de los fármacos
17.
J Neurochem ; 155(4): 370-389, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31872442

RESUMEN

Central sensitization is the potential pathogenesis of chronic migraine (CM) and is related to persistent neuronal hyperexcitability. Dysfunction of excitatory amino acid transporter 2 (EAAT2) leads to the accumulation of glutamate in the synaptic cleft, which may contribute to central sensitization by overactivating glutamate N-methyl-D-aspartate receptors and enhancing synaptic plasticity. However, the therapeutic potential of CM by targeting glutamate clearance remains largely unexplored. The purpose of this study was to investigate the role of EAAT2 in CM central sensitization and explore the effect of EAAT2 expression enhancer LDN-212320 in CM rats. The glutamate concentration was measured by high-performance liquid chromatography in a rat model of CM. Then, q-PCR and western blots were performed to detect EAAT2 expression, and the immunoreactivity of astrocytes was detected by immunofluorescence staining. To understand the effect of EAAT2 on central sensitization of CM, mechanical and thermal hyperalgesia and central sensitization-associated proteins were examined after administration of LDN-212320. In addition, the expression of synaptic-associated proteins was examined and Golgi-Cox staining was used to observe the dendritic spine density of trigeminal nucleus caudalis neurons. Also, the synaptic ultrastructure was observed by transmission electron microscope (TEM) to explore the changes of synaptic plasticity. In our study, elevated glutamate concentration and decreased EAAT2 expression were found in the trigeminal nucleus caudalis of CM rats, administration of LDN-212320 greatly up-regulated the protein expression of EAAT2, alleviated hyperalgesia, decreased the concentration of glutamate and the activation of astrocytes. Furthermore, reductions in calcitonin gene-related peptide, substance P(SP), and phosphorylated NR2B were examined after administration of LDN-212320. Moreover evaluation of the synaptic structure, synaptic plasticity-, and central sensitization-related proteins indicated that EAAT2 might participate in the CM central sensitization process by regulating synaptic plasticity. Taken together, up-regulation of EAAT2 expression has a protective effect in CM rats, and LDN-212320 may have clinical therapeutic potential. Cover Image for this issue: https://doi.org/10.1111/jnc.14769.


Asunto(s)
Astrocitos/metabolismo , Modelos Animales de Enfermedad , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Trastornos Migrañosos/metabolismo , Regulación hacia Arriba/fisiología , Animales , Enfermedad Crónica , Craneotomía/efectos adversos , Craneotomía/métodos , Mediadores de Inflamación/efectos adversos , Mediadores de Inflamación/metabolismo , Masculino , Trastornos Migrañosos/tratamiento farmacológico , Trastornos Migrañosos/etiología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Piridazinas/farmacología , Piridazinas/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
18.
J Clin Invest ; 129(8): 3103-3120, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31112137

RESUMEN

Mechanisms underlying motor neuron degeneration in amyotrophic lateral sclerosis (ALS) are yet unclear. Specific deletion of the ER-component membralin in astrocytes manifested postnatal motor defects and lethality in mice, causing the accumulation of extracellular glutamate through reducing the glutamate transporter EAAT2. Restoring EAAT2 levels in membralin KO astrocytes limited astrocyte-dependent excitotoxicity in motor neurons. Transcriptomic profiles from mouse astrocytic membralin KO motor cortex indicated significant perturbation in KEGG pathway components related to ALS, including downregulation of Eaat2 and upregulation of Tnfrsf1a. Changes in gene expression with membralin deletion also overlapped with mouse ALS models and reactive astrocytes. Our results shown that activation of TNF receptor (TNFR1)-NFκB pathway known to suppress Eaat2 transcription was upregulated with membralin deletion. Further, reduced membralin and EAAT2 levels correlated with disease progression in spinal cord from SOD1-mutant mouse models, and reductions in membralin/EAAT2 were observed in human ALS spinal cord. Importantly, overexpression of membralin in SOD1G93A astrocytes decreased TNFR1 levels and increased EAAT2 expression, and improved motor neuron survival. Importantly, upregulation of membralin in SOD1G93A mice significantly prolonged mouse survival. Together, our study provided a mechanism for ALS pathogenesis where membralin limited glutamatergic neurotoxicity, suggesting that modulating membralin had potentials in ALS therapy.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/metabolismo , Ácido Glutámico/metabolismo , Corteza Motora/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Astrocitos/patología , Regulación hacia Abajo , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Transportador 2 de Aminoácidos Excitadores/genética , Ácido Glutámico/genética , Humanos , Ratones , Ratones Noqueados , Corteza Motora/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Transcripción Genética , Regulación hacia Arriba
19.
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
20.
Neuropharmacology ; 135: 297-307, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29567092

RESUMEN

Cocaine use disorder is a chronically relapsing disease without FDA-approved treatments. Using a rodent model of cocaine relapse, we and others have previously demonstrated that the beta-lactam antibiotic ceftriaxone attenuates cue- and cocaine-primed reinstatement of cocaine-seeking. Ceftriaxone restores cocaine-induced deficits in both system xc- and GLT-1 expression and function in the nucleus accumbens core (NAc). We recently demonstrated that restoration of GLT-1 expression in the NAc is necessary for ceftriaxone to attenuate reinstatement of cocaine-seeking. Here we used an adeno-associated virus (AAV) to overexpress GLT-1a in the NAc to investigate whether such restoration is sufficient to attenuate cue- and cocaine-primed reinstatement. Rats self-administered cocaine for two weeks and received injections of either AAV-GFAP-GLT-1a or AAV-GFAP-eGFP in the NAc following the last day of self-administration. Rats then underwent three weeks of extinction training (during which time transduction and expression occurred) before undergoing a cue- or cocaine-primed reinstatement test. Microdialysis for the quantification of glutamate efflux in the NAc was conducted during the cocaine-primed test. Rats that received AAV-GFAP-GLT-1a reinstated cue-primed cocaine-seeking in a similar manner as rats that received the control AAV-GFAP-eGFP. Upregulation of GLT-1a attenuated glutamate efflux during a cocaine-primed reinstatement test, but was not sufficient to attenuate reinstatement. We confirmed that GLT-1a upregulation resulted in functional upregulation of glutamate transport and expression, without affecting sodium-independent glutamate uptake, indicating system xc-was not altered. These results indicate that upregulation of NAc GLT-1 transporters alone is not sufficient to prevent the reinstatement of cocaine-seeking and implicate additional mechanisms in regulating glutamate efflux.


Asunto(s)
Cocaína/farmacología , Transportador 2 de Aminoácidos Excitadores/biosíntesis , Ácido Glutámico/metabolismo , Núcleo Accumbens/metabolismo , Animales , Dependovirus , Extinción Psicológica/efectos de los fármacos , Vectores Genéticos , Masculino , Ratas , Recurrencia , Autoadministración , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA