Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Brain Res Bull ; 175: 224-233, 2021 10.
Article in English | MEDLINE | ID: mdl-34343641

ABSTRACT

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.


Subject(s)
Brain Ischemia/genetics , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Ischemic Preconditioning , MAP Kinase Signaling System/genetics , NF-kappa B/genetics , Animals , CA1 Region, Hippocampal/pathology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Imidazoles/pharmacology , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , MAP Kinase Signaling System/drug effects , Male , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Neuroprotection , Nitriles/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Sulfones/pharmacology , Transcription Factor RelA/metabolism , p38 Mitogen-Activated Protein Kinases
2.
Neuropharmacology ; 196: 108691, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34197892

ABSTRACT

Recent studies show that neuron-glial communication plays an important role in neurological diseases. Particularly, dysfunction of astroglial glutamate transporter GLT-1 has been involved in various neuropsychiatric disorders, including Parkinson's disease (PD) and depression. Our previous studies indicated hyperactivity of neurons in the lateral habenula (LHb) of hemiparkinsonian rats with depressive-like behaviors. Thus, we hypothesized that impaired expression or function of GLT-1 in the LHb might be a potential contributor to LHb hyperactivity, which consequently induces PD-related depression. In the study, unilateral lesions of the substantia nigra pars compacta (SNc) by 6-hydroxydopamine in rats induced depressive-like behaviors and resulted in neuronal hyperactivity as well as increased glutamate levels in the LHb compared to sham-lesioned rats. Intra-LHb injection of GLT-1 inhibitor WAY-213613 induced the depressive-like behaviors in both groups, but the dose producing behavioral effects in the lesioned rats was lower than that of sham-lesioned rats. In the two groups of rats, WAY-213613 increased the firing rate of LHb neurons and extracellular levels of glutamate, and these excitatory effects in the lesioned rats lasted longer than those in sham-lesioned rats. The functional changes of the GLT-1 which primarily expresses in astrocytes in the LHb may attribute to its downregulation after degeneration of the nigrostriatal pathway. Bioinformatics analysis showed that GLT-1 is correlated with various biomarkers of PD and depression risks. Collectively, our study suggests that astroglial GLT-1 in the LHb regulates the firing activity of the neurons, whereupon its downregulation and dysfunction are closely associated with PD-related depression.


Subject(s)
Astrocytes/metabolism , Depression/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Habenula/metabolism , Parkinsonian Disorders/metabolism , Pars Compacta/metabolism , Animals , Disease Models, Animal , Down-Regulation , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Oxidopamine/toxicity , Parkinsonian Disorders/pathology , Pars Compacta/pathology , Rats , Substantia Nigra/metabolism , Substantia Nigra/pathology , Thalamus/metabolism , Thalamus/pathology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/pathology
3.
Neurotoxicology ; 86: 94-103, 2021 09.
Article in English | MEDLINE | ID: mdl-34310962

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , I-kappa B Kinase/metabolism , Manganese/toxicity , Reactive Oxygen Species/metabolism , YY1 Transcription Factor/biosynthesis , Astrocytes/drug effects , Cells, Cultured , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Humans , Oxidative Stress/drug effects , Oxidative Stress/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
4.
Neurochem Int ; 149: 105142, 2021 10.
Article in English | MEDLINE | ID: mdl-34314789

ABSTRACT

Through the efficient clearance of extracellular glutamate, high affinity astrocytic glutamate transporters constantly shape excitatory neurotransmission in terms of duration and spreading. Even though the glutamate transporter GLT-1 (also known as EAAT2/SLC1A2) is amongst the most abundant proteins in the mammalian brain, its density and activity are tightly regulated. In order to study the influence of changes in the expression of GLT-1 on glutamate uptake capacity, we have developed a model in HEK cells where the density of the transporter can be manipulated thanks to a tetracycline-inducible promoter. Exposing the cells to doxycycline concentration-dependently increased GLT-1 expression and substrate uptake velocity. However, beyond a certain level of induction, increasing the density of transporters at the cell surface failed to increase the maximal uptake. This suggested the progressive generation of a pool of spare transporters, a hypothesis that was further validated using the selective GLT-1 blocker WAY-213613 of which potency was influenced by the density of the transporters. The curve showing inhibition of uptake by increasing concentrations of WAY-213613 was indeed progressively rightward shifted when tested in cells where the transporter density was robustly induced. As largely documented in the context of cell-surface receptors, the existence of 'spare' glutamate transporters in the nervous tissue and particularly in astrocytes could impact on the consequences of physiological or pathological regulation of these transporters.


Subject(s)
Brain/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/metabolism , Neurons/metabolism , Animals , Brain/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Doxycycline/pharmacology , Glutamine/metabolism , HEK293 Cells , Humans , Neurons/drug effects , Rats , Rats, Sprague-Dawley
5.
Neurosci Lett ; 755: 135847, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33774150

ABSTRACT

The development and maintenance of morphine tolerance showed association with neuroinflammation and dysfunction of central glutamatergic system (such as nitration of glutamate transporter). Recent evidence indicated that hydrogen could reduce the levels of neuroinflammation and oxidative stress, but its role in morphine tolerance has not been studied. The rats were intrathecally administered with morphine (10 µg/10 µL each time, twice/day for 5 days). Hydrogen enriched saline (HS) or saline was given intraperitoneally at 1, 3 and 10 mL/kg for 10 min before each dose of morphine administration. The tail-flick latency, mechanical threshold and thermal latency were assessed one day (baseline) before and daily for up to 5 days during morphine injection. The pro-inflammatory cytokine expressions [tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6)] (by western blotting), astrocyte activation (by immunofluorescence and western blotting), and nitration of glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) (by immunoprecipitation), membrane and total expression of N-methyl-d-aspartic acid (NMDA) receptor NR1 and NR2B subunits were carried out in the spinal dorsal horns. Chronic morphine administration induced antinociceptive tolerance, and together led to increased TNF-α, IL-1ß and IL-6 expression, astrocyte activation, GLT-1 and GS nitration, increased membrane and total NR1, NR2B expression. Injection of HS attenuated morphine tolerance in a dose-dependent manner, decreased proinflammatory cytokine expression, inhibited astrocyte activation, decreased GLT-1 and GS nitration, and inhibited membrane trafficking of NMDA receptor. Our result showed that hydrogen pretreatment prevented morphine tolerance by reducing neuroinflammation, GLT-1, GS nitration, NMDA receptor trafficking in the spinal dorsal horn. Pretreatment with hydrogen might be considered as a novel therapeutic strategy for the prevention of morphine tolerance.


Subject(s)
Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Glutamate-Ammonia Ligase/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Morphine/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Saline Solution/administration & dosage , Spinal Cord/metabolism , Analgesics, Opioid/administration & dosage , Animals , Dose-Response Relationship, Drug , Drug Tolerance/physiology , Excitatory Amino Acid Transporter 2/metabolism , Glutamate-Ammonia Ligase/metabolism , Hydrogen/administration & dosage , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Injections, Spinal , Male , Nitrates/antagonists & inhibitors , Nitrates/metabolism , Pain Measurement/drug effects , Pain Measurement/methods , Protein Transport/drug effects , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/drug effects
6.
Neuroreport ; 32(4): 312-320, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33470770

ABSTRACT

Ischemic stroke is the most frequent cause of long-term morbidity and mortality in the elderly worldwide. Mild hypothermia (32-35°C) has been found to have a neuroprotective effect against ischemic stroke. However, the protective mechanisms remain unclear. In the present study, we explore the neuroprotective effect of mild hypothermia in neuron-astrocyte cocultures by oxygen-glucose deprivation/reoxygenation (OGD/R) as well as the underlying mechanisms. Thionin staining was performed and cell viability, extracellular glutamate concentration and the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) pathway-related proteins were detected after OGD/R. The results indicated that mild hypothermia significantly alleviated damage to Nissl bodies and increased the viability of neurons, which alleviated OGD/R-triggered neuronal injury. Furthermore, mild hypothermia significantly enhanced the phosphorylation of Akt (pAkt) and glutamate transporter-1 (GLT-1) and reduced extracellular glutamate concentration after OGD/R. When the PI3K inhibitor LY294002 was added, neuronal viability and the expression of pAkt and GLT-1 decreased, and extracellular glutamate concentration increased. The protective effect of mild hypothermia was counteracted by LY294002. There was no significant change in neuronal viability or the expression of pAkt and GLT-1 in the group treated with dihydrokainate, an inhibitor of GLT-1-function, compared with the mild hypothermia + OGD/R (HOGD) group, but extracellular glutamate concentration was increased. Consequently, mild hypothermia promoted glutamate clearance by regulating GLT-1 expression via the PI3K/Akt pathway, providing a neuroprotective effect against OGD/R injury.


Subject(s)
Cerebral Cortex/cytology , Glutamic Acid/metabolism , Hypothermia, Induced , Ischemic Stroke/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reperfusion Injury/metabolism , Animals , Cell Survival , Chromones/pharmacology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/drug effects , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/drug effects , In Vitro Techniques , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Rats
7.
Neurosci Lett ; 735: 135237, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32645399

ABSTRACT

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.


Subject(s)
Brain/growth & development , Brain/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Sodium Glutamate/toxicity , Age Factors , Animals , Animals, Newborn , Brain/drug effects , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Expression , Glutamic Acid/toxicity , Male , Rats , Rats, Wistar
8.
Brain Res Bull ; 163: 49-56, 2020 10.
Article in English | MEDLINE | ID: mdl-32621862

ABSTRACT

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.


Subject(s)
Dentate Gyrus/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Receptor for Advanced Glycation End Products/deficiency , Signal Transduction/physiology , Stress, Psychological/metabolism , Animals , Chronic Disease , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Knockdown Techniques/methods , Male , Rats , Rats, Sprague-Dawley , Receptor for Advanced Glycation End Products/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology
9.
Brain Res Bull ; 161: 136-146, 2020 08.
Article in English | MEDLINE | ID: mdl-32433937

ABSTRACT

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.


Subject(s)
Brain Injuries/metabolism , Excitatory Amino Acid Transporter 2/biosynthesis , Hypoxia, Brain/metabolism , Midazolam/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Animals, Newborn , Brain Injuries/prevention & control , Dose-Response Relationship, Drug , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Hypnotics and Sedatives/administration & dosage , Hypoxia, Brain/prevention & control , Rats , Rats, Sprague-Dawley
10.
Pharmacol Biochem Behav ; 192: 172913, 2020 05.
Article in English | MEDLINE | ID: mdl-32201299

ABSTRACT

At sub-anaesthetic doses, ketamine, a non competitive N-methyl-d-aspartate (NMDA) receptor antagonist, has demonstrated remarkable and rapid antidepressant (AD) efficacy in patients with treatment-resistant depression (TRD). However, its mechanism of action of ketamine is not fully understood. Since comorbid depression and anxiety disorders often occur, GABAergic/inhibitory and glutamatergic/excitatory drug treatments may be co-administered in these patients. Information regarding this combination is critical to establish efficacy or treatment restrictions to maximize translation from animal models to TRD patients, effectiveness and safety. To assess the specific role of excitatory/inhibitory neurotransmission in the medial prefrontal cortex-raphe nuclei (mPFC-DRN) circuit in the sustained antidepressant-like activity (AD) of ketamine (at t24h post dose), AMPA-R antagonist (intra-DRN) and GABAA-R agonist (intra-mPFC) were co-administered with ketamine (intra-mPFC). Twenty-four hours later, responses in the forced swim test (FST) and neurochemical consequences on extracellular mPFC glutamate, GABA and 5-HT levels were measured in BALB/cJ mice. Intra-DRN NBQX prevented the sustained AD-like activity of ketamine evidenced by decreases in FST swimming duration and blunted cortical 5-HText and Gluext. Intra-mPFC muscimol blocked ketamine AD-like activity and its effects on cortical 5-HText. Moreover, a selective glutamate transporter GLT-1 inhibitor, dihydrokainic acid (DHK) locally perfused into the mPFC produced an AD-like activity at t24h associated with robust increases in mPFC 5-HText, Gluext and GABAext. Thus, the sustained AD-like activity of ketamine is triggered by AMPA-R activation in the DRN and 5-HT - glutamate release in the mPFC, but limited by GABAA-R activation - GABA release in the mPFC. The local blockade of GLT-1 in the mPFC also mimics the rapid responses of ketamine, thus highlighting the role of neuronal-glial adaptation in these effects. These results also suggests the need to test for the concomitant prescription of ketamine and BZD to see whether its sustained antidepressant activity is maintained in TRD patients.


Subject(s)
Antidepressive Agents/administration & dosage , Depression/drug therapy , Dorsal Raphe Nucleus/metabolism , Excitatory Amino Acid Transporter 2/metabolism , Ketamine/administration & dosage , Neuroglia/metabolism , Prefrontal Cortex/metabolism , Receptors, AMPA/metabolism , Receptors, GABA-A/metabolism , Animals , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Benzodiazepines/administration & dosage , Benzodiazepines/pharmacology , Dorsal Raphe Nucleus/drug effects , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacology , Kainic Acid/administration & dosage , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Ketamine/pharmacology , Male , Mice , Mice, Inbred BALB C , Microdialysis , Neuroglia/drug effects , Prefrontal Cortex/drug effects , Quinoxalines/administration & dosage , Quinoxalines/pharmacology , Receptors, AMPA/antagonists & inhibitors , Treatment Outcome
11.
J Biol Chem ; 295(13): 4359-4366, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32079674

ABSTRACT

Excitatory amino acid transporters (EAATs) represent a protein family that is an emerging drug target with great therapeutic potential for managing central nervous system disorders characterized by dysregulation of glutamatergic neurotransmission. As such, it is of significant interest to discover selective modulators of EAAT2 function. Here, we applied computational methods to identify specific EAAT2 inhibitors. Utilizing a homology model of human EAAT2, we identified a binding pocket at the interface of the transport and trimerization domain. We next conducted a high-throughput virtual screen against this site and identified a selective class of EAAT2 inhibitors that were tested in glutamate uptake and whole-cell electrophysiology assays. These compounds represent potentially useful pharmacological tools suitable for further exploration of the therapeutic potential of EAAT2 and may provide molecular insights into mechanisms of allosteric modulation for glutamate transporters.


Subject(s)
Amino Acid Transport System X-AG/antagonists & inhibitors , Binding Sites/drug effects , Central Nervous System Diseases/drug therapy , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Amino Acid Transport System X-AG/chemistry , Amino Acid Transport System X-AG/genetics , Animals , Binding Sites/genetics , Biological Transport/drug effects , Central Nervous System Diseases/genetics , Central Nervous System Diseases/pathology , Computational Biology , Excitatory Amino Acid Transporter 2/chemistry , Excitatory Amino Acid Transporter 2/genetics , Humans , Protein Binding/drug effects , Synaptic Transmission/drug effects , User-Computer Interface
12.
Mol Neurobiol ; 57(5): 2290-2300, 2020 May.
Article in English | MEDLINE | ID: mdl-32008166

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/toxicity , Astrocytes/drug effects , CA1 Region, Hippocampal/drug effects , Cognition Disorders/chemically induced , Complement C1q/physiology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Glutamic Acid/physiology , Microglia/physiology , Neurons/metabolism , Peptide Fragments/toxicity , Animals , Aspartic Acid/pharmacology , Astrocytes/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Ceftriaxone/pharmacology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Complement C1q/biosynthesis , Complement C1q/genetics , Disease Models, Animal , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/physiology , Male , Morris Water Maze Test/drug effects , Morris Water Maze Test/physiology , Patch-Clamp Techniques , Phagocytosis/drug effects , Phagocytosis/physiology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/physiology , Synapses/metabolism , Up-Regulation/drug effects
13.
Neuropharmacology ; 166: 107914, 2020 04.
Article in English | MEDLINE | ID: mdl-32045742

ABSTRACT

Major depressive disorder (MDD) is a leading cause of disability worldwide, with a poorly known pathophysiology and sub-optimal treatment, based on serotonin (5-hydroxytryptamine, 5-HT) reuptake inhibitors. We review existing theories on MDD, paying special attention to the role played by the ventral anterior cingulate cortex (vACC) or its rodent equivalent, infralimbic cortex (IL), which tightly control the activity of brainstem monoamine neurons (including raphe 5-HT neurons) via descending afferents. Further, astrocytes regulate excitatory synapse activity via glutamate reuptake through astrocytic transporters EAAT1 and EAAT2 (GLAST and GLT-1 in rodents), and alterations of astrocyte number/function have been reported in MDD patients and suicide victims. We recently assessed the impact of reducing GLAST/GLT-1 function in IL on emotional behavior and serotonergic function in rodents. The acute pharmacological blockade of GLT-1 with dihydrokainate (DHK) in rat IL evoked an antidepressant-like effect mediated by local AMPA-R activation and a subsequent enhancement of serotonergic function. No effects were produced by DHK microinfusion in prelimbic cortex (PrL). In the second model, a moderate small interfering RNAs (siRNA)-induced reduction of GLAST and GLT-1 expression in mouse IL markedly increased local glutamatergic neurotransmission and evoked a depressive-like phenotype (reversed by citalopram and ketamine), and reduced serotonergic function and BDNF expression in cortical/hippocampal areas. As for DHK, siRNA microinfusion in PrL did not evoke behavioral/neurochemical effects. Overall, both studies support a critical role of the astrocyte-neuron communication in the control of excitatory neurotransmission in IL, and subsequently, on emotional behavior, via the downstream associated changes on serotonergic function.


Subject(s)
Astrocytes/metabolism , Depressive Disorder, Major/metabolism , Emotions/physiology , Glutamic Acid/metabolism , Selective Serotonin Reuptake Inhibitors/metabolism , Serotonergic Neurons/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/psychology , Emotions/drug effects , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/metabolism , Humans , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Serotonergic Neurons/drug effects , Serotonergic Neurons/pathology , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Suicide/psychology
14.
ACS Sens ; 4(9): 2358-2366, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31393114

ABSTRACT

Glutamate is the main excitatory neurotransmitter in the mammalian central nervous system. Excitatory amino acid transporters (EAATs) are a family of transmembrane transporters responsible for glutamate uptake into cells, and their malfunction is related to a variety of diseases, including neurodegenerative diseases and stroke. Screening for and developing inhibitors of EAATs as well as related transporters is a significant field of study for biomedical and pharmaceutical applications. Rapid, high-throughput methods are critical for the study of glutamate transporters, and fluorescent methods are appealing for this purpose as compared to more traditional electrophysiological methods. In this study, we present a method for studying glutamate transporters and inhibitors by utilizing a mutated version of a yellow fluorescent protein (YFP) highly sensitive to quenching by anions (mClY). We applied this YFP variant to fluorescent imaging of anion flux in HEK293 cells caused by transiently expressed excitatory amino acid carrier 1 (EAAC1) and excitatory amino acid transporter 2 (EAAT2) and its inhibition by competitive blockers. This method enables rapid identification of inhibitors and, potentially, activators of EAAT function, which is critical for glutamate transport research.


Subject(s)
Glutamic Acid/metabolism , Halogens/metabolism , Optical Imaging/methods , Bacterial Proteins/genetics , Biological Transport , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acid Transporter 3/antagonists & inhibitors , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , HEK293 Cells , Humans , Luminescent Proteins/genetics , Time Factors
15.
CNS Neurosci Ther ; 25(4): 509-518, 2019 04.
Article in English | MEDLINE | ID: mdl-30311425

ABSTRACT

AIM: Deficient glutamate reuptake occurs in the cerebral cortex of Huntington's disease (HD) patients and murine models. Here, we examine the effects of partial or complete blockade of glutamate transporters on excitatory postsynaptic currents (EPSCs) of cortical pyramidal neurons (CPNs). METHODS: Whole-cell patch clamp recordings of CPNs in slices from symptomatic R6/2 mice and wild-type (WT) littermates were used to examine the effects of selective or concurrent inhibition of glutamate reuptake transporters. RESULTS: Selective inhibition of the glial glutamate transporter 1 (GLT-1) or the glutamate aspartate transporter (GLAST) produced slight decreases in decay time of evoked EPSCs in CPNs from WT and R6/2 mice with no significant differences between genotypes. In contrast, concurrent inhibition of both transporters with DL-TBOA induced a significant increase in area and decay time and this effect was significantly greater in R6/2 CPNs. Furthermore, full blockade also reduced spontaneous EPSC frequency and exacerbated epileptiform activity in CPNs from symptomatic R6/2 mice. CONCLUSIONS: R6/2 CPNs are more sensitive to glutamate accumulation during full inhibition of both glutamate transporters, and these neurons have homeostatic mechanisms to cope with inhibition of GLT-1 or GLAST by a mechanism that involves upregulation of either transporter when the other is deficient.


Subject(s)
Cerebral Cortex/physiopathology , Disease Models, Animal , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Huntington Disease/physiopathology , Animals , Aspartic Acid/pharmacology , Benzopyrans/pharmacology , Excitatory Amino Acid Transporter 1/physiology , Excitatory Amino Acid Transporter 2/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Huntington Disease/genetics , Male , Mice , Mice, Transgenic , Organ Culture Techniques
16.
Cell Cycle ; 18(1): 105-114, 2019 01.
Article in English | MEDLINE | ID: mdl-30558468

ABSTRACT

Little is known about the effect of astroglial GLT-1 of post-stroke depression (PSD) rat model on the function of neural stem cells (NSCs). This study aimed to investigate whether astroglial GLT-1 of PSD rats affect differentiation of NSCs from neonatal rat hippocampus and synaptic formation of NSC-derived neurons. Astrocytes were isolated from the left hippocampus of normal adult SD rats and PSD rats. A lentiviral vector was used to silence the expression of GLT-1 in astrocytes of PSD rats. NSCs were respectively co-cultured with normal (control), PSD, and GLT-1 silenced astrocytes for 7 days. GLT-1, GFAP, MAP2, Synaptophysin (SYN), glutamate (Glu) and glutamine (Gln) were respectively measured by qRT-PCR, western blot, immunofluorescence and efficient mass spectrometry (MS). PSD astrocytes increased the number of NSC-derived astrocytes, but inhibited the expression of GLT-1 of NSC-derived astrocytes and synapses of NSC-derived neurons. On the basis of the low expression of GLT-1 in PSD astrocytes, we further silenced GLT-1 in PSD astrocytes. Interestingly, GLT-1 silenced PSD astrocytes more obviously inhibited synapses of NSC-derived neurons, but increased the number of NSC-derived neurons and reversed the expression of GLT-1 in NSC-derived astrocytes. At the same time, concentration of glutamate in the medium elevated, and glutamine in the medium gradually reduced. In NSC-derived neurons and astrocytes, glutamate metabolism was also affected by changed GLT-1. Down-expressed GLT-1 in PSD astrocytes stimulated NSCs differentiating into astrocytes, but inhibiting the formation of functional synapses by influencing glutamate metabolism in vitro.


Subject(s)
Depression/genetics , Excitatory Amino Acid Transporter 2/genetics , Neurons/metabolism , Stroke/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Differentiation/genetics , Depression/etiology , Depression/pathology , Disease Models, Animal , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Gene Expression Regulation/genetics , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Neural Stem Cells/metabolism , Neurons/pathology , Rats , Stroke/complications , Stroke/pathology , Stroke Rehabilitation
17.
Physiol Rep ; 6(18): e13877, 2018 09.
Article in English | MEDLINE | ID: mdl-30230240

ABSTRACT

The astrocytic glutamate transporter (GLT1) plays an important role in the maintenance of extracellular glutamate concentration below neurotoxic levels in brain. However, the functional role of GLT1 within the nucleus of the solitary tract (NTS) in the regulation of cardiovascular function remains unclear. We examined the effect of inhibiting GLT1 in the subpostremal NTS on mean arterial pressure (MAP), renal sympathetic nerve activity (RSNA) and heart rate (HR) in anesthetized, artificially ventilated rats. It was found that dihydrokainate (DHK; inhibitor of GLT1, 5 mmol/L, 100 nL) injections into the NTS (n = 6) decreased MAP (50 ± 10 mmHg, mean ± SD), RSNA (89 ± 14%) and HR (37 ± 6 bpm). Pretreatment with kynurenate (KYN; glutamate receptor antagonist, 5 mmol/L, 30 µL) topically applied to the dorsal surface of the brainstem (n = 4) attenuated the responses to NTS injections of DHK (P < 0.01). The effect of DHK on arterial baroreflex function was examined using i.v. infusions of phenylephrine and nitroprusside. DHK reduced baroreflex response range (maximum-minimum) of RSNA by 91 ± 2% and HR by 83 ± 5% (n = 6, P < 0.001). These results indicate that inhibition of GLT1 within the NTS decreases MAP, RSNA, and HR by the activation of ionotropic glutamate receptors. As a result, baroreflex control of RSNA and HR was dramatically attenuated. The astrocytic glutamate transporter in the NTS plays an important role in the maintenance and regulation of cardiovascular function.


Subject(s)
Baroreflex/physiology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Heart Rate/physiology , Solitary Nucleus/metabolism , Sympathetic Fibers, Postganglionic/metabolism , Animals , Baroreflex/drug effects , Excitatory Amino Acid Transporter 2/metabolism , Heart Rate/drug effects , Kainic Acid/administration & dosage , Kainic Acid/analogs & derivatives , Male , Microinjections/methods , Rats , Rats, Sprague-Dawley , Solitary Nucleus/drug effects , Sympathetic Fibers, Postganglionic/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
18.
Neurotoxicology ; 67: 112-120, 2018 07.
Article in English | MEDLINE | ID: mdl-29778792

ABSTRACT

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.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Excitatory Amino Acid Transporter 1/biosynthesis , Excitatory Amino Acid Transporter 2/biosynthesis , Manganese/toxicity , Valproic Acid/pharmacology , Animals , Anticonvulsants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Cells, Cultured , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Int J Neuropsychopharmacol ; 20(12): 988-993, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29016806

ABSTRACT

Background: Glutamatergic neurotransmission has emerged as a novel target in antidepressant drug development, with a critical role of the ventral anterior cingulate cortex. We recently reported that blockade of the astrocytic glutamate transporter GLT-1 with dihydrokainic acid in infralimbic cortex (rodent equivalent of ventral anterior cingulate cortex), but not in the adjacent prelimbic cortex, evoked robust antidepressant-like effects through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor activation and increased serotonin release. Methods: 2-deoxy-2-[18F]-fluoro-D-glucose-positron emission tomography and computed tomography in 36 male Wistar rats microinfused bilaterally in prelimbic cortex or infralimbic cortex with dihydrokainic acid or vehicle. Results: Dihydrokainic acid microinfusion in infralimbic cortex and prelimbic cortex evoked dramatically different regional patterns of subcortical activity. In infralimbic cortex, dihydrokainic acid selectively affected midbrain areas, whereas in prelimbic cortex it affected the basal ganglia, the thalamus, and both superior and inferior colliculi. Conclusions: These results highlight the differential connectivity of infralimbic and prelimbic cortex with subcortical brain regions and support the involvement of infralimbic cortex-midbrain pathway in the antidepressant-like effects of dihydrokainic acid.


Subject(s)
Cerebral Cortex/drug effects , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Transporter 2/metabolism , Kainic Acid/analogs & derivatives , Animals , Brain Mapping , Cerebral Cortex/diagnostic imaging , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Fluorodeoxyglucose F18 , Kainic Acid/pharmacology , Male , Neural Pathways/drug effects , Neural Pathways/physiology , Positron-Emission Tomography , Rats , Rats, Wistar , Tomography Scanners, X-Ray Computed
20.
J Physiol ; 595(17): 6045-6063, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28677303

ABSTRACT

KEY POINTS: Excitatory amino acid transporter 2 (EAAT2) is present on astrocytes in the nucleus tractus solitarii (nTS), an important nucleus in cardiorespiratory control. Its specific role in influencing nTS neuronal activity and thereby basal and reflex cardiorespiratory function is unknown. The specific role of nTS EAAT2 was determined via whole animal and brainstem slice patch clamp experiments. Astrocytic EAAT2 buffers basal glutamate activation of AMPA-type glutamate receptors and therefore decreases baseline excitability of nTS neurons. EAAT2 modulates cardiorespiratory control and tempers excitatory cardiorespiratory responses to activation of the peripheral chemoreflex. This study supports the concept that nTS astrocyte transporters influence sympathetic nervous system activity and cardiorespiratory reflex function in health and disease. ABSTRACT: Glutamatergic signalling is critical in the nucleus tractus solitarii (nTS) for cardiorespiratory homeostasis and initiation of sensory reflexes, including the chemoreflex activated during hypoxia. Maintenance of nTS glutamate concentration occurs in part through astrocytic excitatory amino acid transporters (EAATs). We previously established the importance of EAATs in the nTS by demonstrating their inhibition produced neuronal excitation to alter basal cardiorespiratory function. Since EAAT2 is the most expressed EAAT in the nTS, this study specifically determined EAAT2's role in nTS astrocytes, its influence on neuronal and synaptic properties, and ultimately on basal and reflex cardiorespiratory function. The EAAT2-specific antagonist dihydrokainate (DHK) was microinjected into the anaesthetized rat nTS or applied to rat nTS slices. DHK produced depressor, bradycardic and sympathoinhibitory responses and reduced neural respiration in the intact rat, mimicking responses to glutamate excitation. DHK also enhanced responses to glutamate microinjection. DHK elevated extracellular nTS glutamate concentration, depolarized neurons and enhanced spontaneous EPSCs. EAAT2 block also augmented action potential discharge in chemosensitive nTS neurons. Glial recordings confirmed EAAT2 is functional on nTS astrocytes. Neuronal excitation and cardiorespiratory effects following EAAT2 inhibition were due to activation of putative extrasynaptic AMPA receptors as their antagonism blocked DHK responses in the intact rat nTS and the slice. The DHK-induced elevation of extracellular glutamate and neuronal excitation augmented chemoreflex-mediated pressor, sympathoexcitatory and minute neural ventilation responses in the rat. These data shed new light on the important role astrocytic EAAT2 plays on buffering nTS excitation and overall cardiorespiratory function.


Subject(s)
Excitatory Amino Acid Transporter 2/physiology , Neuroglia/physiology , Respiration , Solitary Nucleus/physiology , Action Potentials , Animals , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Kainic Acid/analogs & derivatives , Kainic Acid/pharmacology , Male , Neurons/physiology , Rats, Sprague-Dawley , Receptors, AMPA/physiology , Reflex/physiology , Solitary Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...