Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 353
1.
Addict Biol ; 27(1): e13085, 2022 01.
Article En | MEDLINE | ID: mdl-34390300

Disrupted brain gamma-aminobutyric acid (GABA)/glutamate homeostasis is a promising target for pharmacological intervention in co-occurring bipolar disorder (BD) and cannabis use disorder (CUD). Gabapentin is a safe and well-tolerated medication, FDA-approved to treat other neurological diseases, that restores GABA/glutamate homeostasis, with treatment studies supporting efficacy in treating CUD, as well as anxiety and sleep disorders that are common to both BD and CUD. The present manuscript represents the primary report of a randomized, double-blind, placebo-controlled, crossover (1-week/condition), multimodal-MRI (proton-MR spectroscopy, functional MRI) pilot study of gabapentin (1200 mg/day) in BD + CUD (n = 22). Primary analyses revealed that (1) gabapentin was well tolerated and adherence and retention were high, (2) gabapentin increased dorsal anterior cingulate cortex (dACC) and right basal ganglia (rBG) glutamate levels and (3) gabapentin increased activation to visual cannabis cues in the posterior midcingulate cortex (pMCC, a region involved in response inhibition to rewarding stimuli). Exploratory evaluation of clinical outcomes further found that in participants taking gabapentin versus placebo, (1) elevations of dACC GABA levels were associated with lower manic/mixed and depressive symptoms and (2) elevations of rBG glutamate levels and pMCC activation to cannabis cues were associated with lower cannabis use. Though promising, the findings from this study should be interpreted with caution due to observed randomization order effects on dACC glutamate levels and identification of statistical moderators that differed by randomization order (i.e. cigarette-smoking status on rBG glutamate levels and pMCC cue activation). Nonetheless, they provide the necessary foundation for a more robustly designed (urn-randomized, parallel-group) future study of adjuvant gabapentin for BD + CUD.


Bipolar Disorder/drug therapy , Gabapentin/therapeutic use , Glutamic Acid/drug effects , Marijuana Abuse/drug therapy , gamma-Aminobutyric Acid/drug effects , Adolescent , Adult , Bipolar Disorder/epidemiology , Cigarette Smoking/epidemiology , Double-Blind Method , Female , Gabapentin/administration & dosage , Gabapentin/adverse effects , Gyrus Cinguli/drug effects , Humans , Magnetic Resonance Imaging , Male , Marijuana Abuse/epidemiology , Middle Aged , Pilot Projects , Proton Magnetic Resonance Spectroscopy , Young Adult
2.
Addict Biol ; 27(1): e13068, 2022 01.
Article En | MEDLINE | ID: mdl-34128302

Methamphetamine (METH) elicits endogenous glutamate (Glu) in the brain, which could partially explain METH-induced memory deficits. Here, we investigated the therapeutic effects of electroacupuncture (EA) on spatial memory deficits in METH withdrawal mice and its potential synaptic mechanisms. We found that EA at acupoints 'Baihui' and 'Yintang' ameliorated the impaired spatial memory in METH withdrawal mice. In parallel, EA attenuated the Glu levels in vivo and suppressed the neuronal activities within dCA1 of METH withdrawal mice, as indicated by the decreasing c-Fos levels and the amplitude of mEPSP. In the dCA1, EA decreased A1-like astrocytes but increased astrocytic glutamatergic transporting molecules including glutamate transporter 1 and glutamine synthase. However, EA seemed to have no effects on presynaptic Glu transmission from the dCA3, as evidenced by the similiar levels of c-Fos in the dCA3 neurons, synaptic vesicular markers of dCA3 neural terminals and values of paired-pulse ratio in the dCA1 neurons between EA-treated and sham EA-treated METH withdrawal mice. These findings suggest that EA might normalize the dCA1 Glu levels at least in part through enhancing astrocyte-mediated Glu clearance. Taken together, astrocytes might be a novel target for developing therapeutic interventions against the impaired memory behaviours in METH users, and EA represents a promising non-invasive therapeutic strategy for the management of drug-caused memory deficits.


Amphetamine-Related Disorders/physiopathology , Astrocytes/drug effects , Electroacupuncture/methods , Glutamic Acid/drug effects , Memory Disorders/therapy , Spatial Memory/drug effects , Animals , Male , Methamphetamine/pharmacology , Mice , Neurons/drug effects
3.
Neuropharmacology ; 205: 108918, 2022 03 01.
Article En | MEDLINE | ID: mdl-34896402

The Central Amygdala (CeA) has been heavily implicated in many aspects of alcohol use disorder. Ethanol (EtOH) has been shown to modulate glutamatergic transmission in the lateral subdivision of the CeA, however, the exact mechanism of this modulation is still unclear. EtOH exposure is associated with increased pro-inflammatory cytokines in the CeA, and inhibition of neuroimmune cells (microglia and astrocytes) has previously been shown to reduce EtOH drinking in animal models. Since neuroimmune activation seems to be involved in many of the effects of EtOH, we hypothesized that acute EtOH exposure will increase excitatory glutamatergic transmission in the CeA via modulation of neuroimmune cells. Using ex vivo brain slice whole-cell patch clamp electrophysiology, it was found that a physiologically relevant concentration of EtOH (20 mM) significantly increased presynaptic glutamatergic transmission in the CeA. Pharmacologic and chemogenetic inhibition of astrocyte function significantly reduced the ability of EtOH to modulate CeA glutamatergic transmission with minimal impact of microglia inhibition. This finding prompted additional studies examining whether direct neuroimmune activation through lipopolysaccharide (LPS) might lead to an increase in the glutamatergic transmission in the CeA. It was found that LPS modulation of glutamatergic transmission was limited by microglia activation and required astrocyte signaling. Taken together these results support the hypothesis that acute EtOH enhances lateral CeA glutamatergic transmission through an astrocyte mediated mechanism.


Astrocytes/drug effects , Central Amygdaloid Nucleus/drug effects , Central Nervous System Depressants/pharmacology , Electrophysiological Phenomena/drug effects , Ethanol/pharmacology , Glutamic Acid/drug effects , Microglia/drug effects , Animals , Mice
4.
Int J Mol Sci ; 22(21)2021 Oct 23.
Article En | MEDLINE | ID: mdl-34768876

The glutamatergic neurotransmitter system has received substantial attention in research on the pathophysiology and treatment of neurological disorders. The study investigated the effect of the polyphenolic compound chlorogenic acid (CGA) on glutamate release in rat cerebrocortical nerve terminals (synaptosomes). CGA inhibited 4-aminopyridine (4-AP)-induced glutamate release from synaptosomes. This inhibition was prevented in the absence of extracellular Ca2+ and was associated with the inhibition of 4-AP-induced elevation of Ca2+ but was not attributed to changes in synaptosomal membrane potential. In line with evidence observed through molecular docking, CGA did not inhibit glutamate release in the presence of P/Q-type Ca2+ channel inhibitors; therefore, CGA-induced inhibition of glutamate release may be mediated by P/Q-type Ca2+ channels. CGA-induced inhibition of glutamate release was also diminished by the calmodulin and Ca2+/calmodilin-dependent kinase II (CaMKII) inhibitors, and CGA reduced the phosphorylation of CaMKII and its substrate, synapsin I. Furthermore, pretreatment with intraperitoneal CGA injection attenuated the glutamate increment and neuronal damage in the rat cortex that were induced by kainic acid administration. These results indicate that CGA inhibits glutamate release from cortical synaptosomes by suppressing P/Q-type Ca2+ channels and CaMKII/synapsin I pathways, thereby preventing excitotoxic damage to cortical neurons.


Calcium Channels/metabolism , Chlorogenic Acid/pharmacology , Glutamic Acid/metabolism , Animals , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Chlorogenic Acid/metabolism , Excitatory Amino Acid Agents , Glutamic Acid/drug effects , Kainic Acid/metabolism , Male , Membrane Potentials/drug effects , Molecular Docking Simulation , Neurons/drug effects , Neurons/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Synaptic Vesicles/metabolism , Synaptosomes/metabolism
5.
Nutrients ; 13(11)2021 Nov 20.
Article En | MEDLINE | ID: mdl-34836413

Anorexia nervosa (AN) is a highly complex disorder to treat, especially in severe and enduring cases. Whilst the precise aetiology of the disorder is uncertain, malnutrition and weight loss can contribute to reductions in grey and white matter of the brain, impairments in neuroplasticity and neurogenesis and difficulties with cognitive flexibility, memory and learning. Depression is highly comorbid in AN and may be a barrier to recovery. However, traditional antidepressants are often ineffective in alleviating depressive symptoms in underweight patients with AN. There is an urgent need for new treatment approaches for AN. This review gives a conceptual overview for the treatment of AN with ketamine. Ketamine has rapid antidepressant effects, which are hypothesised to occur via increases in glutamate, with sequelae including increased neuroplasticity, neurogenesis and synaptogenesis. This article provides an overview of the use of ketamine for common psychiatric comorbidities of AN and discusses particular safety concerns and side effects. Potential avenues for future research and specific methodological considerations are explored. Overall, there appears to be ample theoretical background, via several potential mechanisms, that warrant the exploration of ketamine as a treatment for adults with AN.


Anorexia Nervosa/drug therapy , Antidepressive Agents/therapeutic use , Depression/drug therapy , Ketamine/therapeutic use , Anorexia Nervosa/psychology , Brain/drug effects , Comorbidity , Depression/psychology , Glutamic Acid/drug effects , Humans , Neuronal Plasticity/drug effects
6.
Neurosci Lett ; 762: 136146, 2021 09 25.
Article En | MEDLINE | ID: mdl-34332028

INTRODUCTION: Current treatments for cocaine use disorder (CUD) are not very effective and better treatments are needed. This study investigates the effectiveness of a combined intervention that targets the assumed underlying glutamate pathology in cocaine users. To this end, the combined effects of N-acetylcysteine (NAC) and working memory (WM) training on glutamate concentrations in the dorsal and rostral ACC were investigated in a randomized, double-blind placebo-controlled design. METHODS: In this study, 38 regular cocaine-using men were randomized to either 25-days with 2400 mg/day NAC and WM-training or 25 days with placebo with WM-training. Cocaine use, impulsivity, and glutamate concentrations in the dACC and rACC using proton Magnetic Resonance Spectroscopy were assessed at baseline and after treatment. RESULTS: Twenty-four participants completed the study, of which 9 received NAC and 15 received placebo. There were no baseline correlations of glutamate concentrations in the dACC or rACC with cocaine use measures or impulsivity. Additionally, there were no effects of NAC, WM-training, or the combination thereof on (changes in) glutamate concentrations in the dACC or rACC. DISCUSSION: This randomized proof of concept study could not confirm our hypotheses. Possible explanations are insufficient power and the possible absence of deviant baseline glutamate concentrations in the included participants. Future studies should consider larger samples and a non-using control group to confirm baseline deviations in glutamate in cocaine users.


Acetylcysteine/therapeutic use , Cocaine-Related Disorders/therapy , Glutamic Acid/drug effects , Gyrus Cinguli/drug effects , Learning , Adult , Double-Blind Method , Glutamic Acid/metabolism , Gyrus Cinguli/metabolism , Humans , Male , Memory, Short-Term , Middle Aged , Proof of Concept Study
7.
Placenta ; 110: 46-55, 2021 07.
Article En | MEDLINE | ID: mdl-34120018

INTRODUCTION: Placental oxidative stress features in pregnancy pathologies but in clinical trials antioxidant supplementation has not improved outcomes. N-acetylcysteine (NAC) stimulates glutathione production and is proposed as a therapeutic agent in pregnancy. However, key elements of N-acetylcysteine biology, including its cellular uptake mechanism, remains unclear. This study explores how the cystine/glutamate transporter xCT may mediate N-acetylcysteine uptake and how N-acetylcysteine alters placental redox status. METHODS: The involvement of xCT in NAC uptake by the human placenta was studied in perfused placenta and Xenopus oocytes. The effect of short-term N-acetylcysteine exposure on the placental villous proteome was determined using LC-MS. The effect of N-acetylcysteine on Maxi-chloride channel activity was investigated in perfused placenta, villous fragments and cell culture. RESULTS: Maternoplacental N-acetylcysteine administration stimulated intracellular glutamate efflux suggesting a role of the exchange transporter xCT, which was localised to the microvillous membrane of the placental syncytiotrophoblast. Placental exposure to a bolus of N-acetylcysteine inhibited subsequent activation of the redox sensitive Maxi-chloride channel independently of glutathione synthesis. Stable isotope quantitative proteomics of placental villi treated with N-acetylcysteine demonstrated changes in pathways associated with oxidative stress, apoptosis and the acute phase response. DISCUSSION: This study suggests that xCT mediates N-acetylcysteine uptake into the placenta and that N-acetylcysteine treatment of placental tissue alters the placental proteome while regulating the redox sensitive Maxi-chloride channel. Interestingly N-acetylcysteine had antioxidant effects independent of the glutathione pathway. Effective placental antioxidant therapy in pregnancy may require maintaining the balance between normalising redox status without inhibiting physiological redox signalling.


Acetylcysteine/pharmacology , Amino Acid Transport System y+/genetics , Chloride Channels/antagonists & inhibitors , Placenta , Acetylcysteine/metabolism , Amino Acid Transport System y+/metabolism , Animals , Chloride Channels/metabolism , Chorionic Villi/drug effects , Chorionic Villi/metabolism , Female , Gene Expression/drug effects , Glutamic Acid/drug effects , Glutamic Acid/metabolism , HEK293 Cells , Humans , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/genetics , Placenta/drug effects , Placenta/metabolism , Pregnancy , Proteome/drug effects , Proteome/metabolism , Xenopus laevis
8.
Neurobiol Learn Mem ; 183: 107480, 2021 09.
Article En | MEDLINE | ID: mdl-34153453

Perturbations in the glutamate-glutamine cycle and glutamate release from presynaptic terminals have been involved in the development of cognitive deficits in Alzheimer's disease (AD) patients and mouse models. Glutamate transporter-1 (GLT-1) removes glutamate from the synaptic cleft and transports it into astrocytes, where it is used as substrate for the glutamate-glutamine cycle. Ceftriaxone has been reported to improve cognitive deficits in AD mice by increasing GLT-1 expression, glutamate transformation to glutamine, and glutamine efflux from astrocytes. However, the impact of ceftriaxone on glutamine metabolism in neurons is unknown. The present study aimed to investigate whether ceftriaxone regulated the production and vesicular assembly of glutamate in the presynaptic terminals of neurons and to determine GLT-1 involvement in this process. We used the amyloid precursor protein (APP)/presenilin-1 (PS1) AD mouse model and GLT-1 knockdown APP/PS1 (GLT-1+/-/APP/PS1) mice. The expression levels of sodium-coupled neutral amino-acid transporter 1 (SNAT1) and vesicular glutamate transporters 1 and 2 (VGLUT1/2) were analyzed by immunofluorescence and immunohistochemistry staining as well as by Western blotting. Glutaminase activity was assayed by fluorometry. Ceftriaxone treatment significantly increased SNAT1 expression and glutaminase activity in neurons in APP/PS1 mice. Similarly, VGLUT1/2 levels were increased in the presynaptic terminals of APP/PS1 mice treated with ceftriaxone. The deletion of one GLT-1 allele in APP/PS1 mice prevented the ceftriaxone-induced upregulation of SNAT1 and VGLUT1/2 expression, indicating that GLT-1 played an important role in ceftriaxone effect. Based on the role of SNAT1, glutaminase, and VGLUT1/2 in the glutamate-glutamine cycle in neurons, the present results suggested that ceftriaxone improved the production and vesicular assembly of glutamate as a neurotransmitter in presynaptic terminals by acting on GLT-1 in APP/PS1 mice.


Alzheimer Disease/metabolism , Anti-Bacterial Agents/pharmacology , Ceftriaxone/pharmacology , Excitatory Amino Acid Transporter 2/drug effects , Presynaptic Terminals/drug effects , Synaptic Vesicles/drug effects , Alzheimer Disease/genetics , Amino Acid Transport System A/drug effects , Amino Acid Transport System A/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Gene Knockdown Techniques , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Glutaminase/drug effects , Glutaminase/metabolism , Mice , Mice, Transgenic , Presenilin-1/genetics , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Protein 1/drug effects , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Glutamate Transport Protein 2/drug effects , Vesicular Glutamate Transport Protein 2/metabolism
9.
J Psychopharmacol ; 35(7): 814-822, 2021 Jul.
Article En | MEDLINE | ID: mdl-33860709

BACKGROUND: Emerging evidence supports the antipsychotic effect of cannabidiol, a non-intoxicating component of cannabis, in people with psychosis. Preclinical findings suggest that this antipsychotic effect may be related to cannabidiol modulating glutamatergic signalling in the brain. AIM: The purpose of this study was to investigate the effects of cannabidiol on the neurochemical mechanisms underlying psychosis. METHODS: We investigated the effects of a single oral dose of cannabidiol (600 mg) in patients with psychosis, using a double-blind, randomised, placebo-controlled, repeated-measures, within-subject cross-over design. After drug administration, 13 patients were scanned using proton magnetic resonance spectroscopy to measure left hippocampal glutamate levels. Symptom severity was rated using the Positive and Negative Syndrome Scale 60 min before drug administration (pre-scan), and 270 min after drug administration (post-scan). Effects of cannabidiol on hippocampal glutamate levels, symptom severity, and correlations between hippocampal glutamate and symptoms were investigated. RESULTS: Compared to placebo, there was a significant increase in hippocampal glutamate (p=0.035), and a significantly greater decrease in symptom severity (p=0.032) in the psychosis patients under cannabidiol treatment. There was also a significant negative relationship between post-treatment total Positive and Negative Syndrome Scale score and hippocampal glutamate (p=0.047), when baseline Positive and Negative Syndrome Scale score, treatment (cannabidiol vs placebo), and interaction between treatment and glutamate levels were controlled for. CONCLUSIONS: These findings may suggest a link between the increase in glutamate levels and concomitant decrease in symptom severity under cannabidiol treatment observed in psychosis patients. Furthermore, the findings provide novel insight into the potential neurochemical mechanisms underlying the antipsychotic effects of cannabidiol.


Antipsychotic Agents/pharmacology , Cannabidiol/pharmacology , Glutamic Acid/drug effects , Hippocampus/drug effects , Psychotic Disorders/drug therapy , Schizophrenia/drug therapy , Adult , Antipsychotic Agents/administration & dosage , Cannabidiol/administration & dosage , Female , Glutamic Acid/metabolism , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Humans , Male , Outcome Assessment, Health Care , Proton Magnetic Resonance Spectroscopy , Psychotic Disorders/diagnostic imaging , Psychotic Disorders/metabolism , Schizophrenia/diagnostic imaging , Schizophrenia/metabolism , Severity of Illness Index , Young Adult
10.
JAMA Psychiatry ; 78(6): 667-681, 2021 06 01.
Article En | MEDLINE | ID: mdl-33881460

Importance: Proton magnetic resonance spectroscopy (1H-MRS) studies indicate that altered brain glutamatergic function may be associated with the pathophysiology of schizophrenia and the response to antipsychotic treatment. However, the association of altered glutamatergic function with clinical and demographic factors is unclear. Objective: To assess the associations of age, symptom severity, level of functioning, and antipsychotic treatment with brain glutamatergic metabolites. Data Sources: The MEDLINE database was searched to identify journal articles published between January 1, 1980, and June 3, 2020, using the following search terms: MRS or magnetic resonance spectroscopy and (1) schizophrenia or (2) psychosis or (3) UHR or (4) ARMS or (5) ultra-high risk or (6) clinical high risk or (7) genetic high risk or (8) prodrome* or (9) schizoaffective. Authors of 114 1H-MRS studies measuring glutamate (Glu) levels in patients with schizophrenia were contacted between January 2014 and June 2020 and asked to provide individual participant data. Study Selection: In total, 45 1H-MRS studies contributed data. Data Extraction and Synthesis: Associations of Glu, Glu plus glutamine (Glx), or total creatine plus phosphocreatine levels with age, antipsychotic medication dose, symptom severity, and functioning were assessed using linear mixed models, with study as a random factor. Main Outcomes and Measures: Glu, Glx, and Cr values in the medial frontal cortex (MFC) and medial temporal lobe (MTL). Results: In total, 42 studies were included, with data for 1251 patients with schizophrenia (mean [SD] age, 30.3 [10.4] years) and 1197 healthy volunteers (mean [SD] age, 27.5 [8.8] years). The MFC Glu (F1,1211.9 = 4.311, P = .04) and Glx (F1,1079.2 = 5.287, P = .02) levels were lower in patients than in healthy volunteers, and although creatine levels appeared lower in patients, the difference was not significant (F1,1395.9 = 3.622, P = .06). In both patients and volunteers, the MFC Glu level was negatively associated with age (Glu to Cr ratio, F1,1522.4 = 47.533, P < .001; cerebrospinal fluid-corrected Glu, F1,1216.7 = 5.610, P = .02), showing a 0.2-unit reduction per decade. In patients, antipsychotic dose (in chlorpromazine equivalents) was negatively associated with MFC Glu (estimate, 0.10 reduction per 100 mg; SE, 0.03) and MFC Glx (estimate, -0.11; SE, 0.04) levels. The MFC Glu to Cr ratio was positively associated with total symptom severity (estimate, 0.01 per 10 points; SE, 0.005) and positive symptom severity (estimate, 0.04; SE, 0.02) and was negatively associated with level of global functioning (estimate, 0.04; SE, 0.01). In the MTL, the Glx to Cr ratio was positively associated with total symptom severity (estimate, 0.06; SE, 0.03), negative symptoms (estimate, 0.2; SE, 0.07), and worse Clinical Global Impression score (estimate, 0.2 per point; SE, 0.06). The MFC creatine level increased with age (estimate, 0.2; SE, 0.05) but was not associated with either symptom severity or antipsychotic medication dose. Conclusions and Relevance: Findings from this mega-analysis suggest that lower brain Glu levels in patients with schizophrenia may be associated with antipsychotic medication exposure rather than with greater age-related decline. Higher brain Glu levels may act as a biomarker of illness severity in schizophrenia.


Antipsychotic Agents/pharmacology , Brain/metabolism , Glutamic Acid/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Schizophrenia/physiopathology , Adult , Age Factors , Biomarkers/metabolism , Brain/diagnostic imaging , Brain/drug effects , Female , Glutamic Acid/drug effects , Glutamine/drug effects , Glutamine/metabolism , Humans , Male , Patient Acuity , Proton Magnetic Resonance Spectroscopy , Young Adult
11.
Neuropharmacology ; 187: 108492, 2021 04 01.
Article En | MEDLINE | ID: mdl-33582153

In this work, modulation by orexin-A of the release of glutamate and GABA from bipolar and amacrine cells respectively was studied by examining the effects of the neuropeptide on miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs) of rat retinal ganglion cells (GCs). Using RNAscope in situ hybridization in combination with immunohistochemistry, we showed positive signals for orexin receptor-1 (OX1R) mRNA in the bipolar cell terminals and those for orexin receptor-2 (OX2R) mRNA in the amacrine cell terminals. With whole-cell patch-clamp recordings in rat retinal slices, we demonstrated that application of orexin-A reduced the interevent interval of mEPSCs of GCs through OX1R. However, it increased the interevent interval of mIPSCs, mediated by GABAA receptors, through OX2R. Furthermore, orexin-A-induced reduction of mEPSC interevent interval was abolished by the application of PI-PLC inhibitors or PKC inhibitors. In contrast, orexin-A-induced increase of GABAergic mIPSC interevent interval was mimicked by 8-Br-cAMP or an adenylyl cyclase activator, but was eliminated by PKA antagonists. Finally, application of nimodipine, an L-type Ca2+ channel blocker, increased both mEPSC and mIPSC interevent interval, and co-application of orexin-A no longer changed the mEPSCs and mIPSCs. We conclude that orexin-A increases presynaptic glutamate release onto GCs by activating L-type Ca2+ channels in bipolar cells, a process that is mediated by an OX1R/PI-PLC/PKC signaling pathway. However, orexin-A decreases presynaptic GABA release onto GCs by inhibiting L-type Ca2+ channels in amacrine cells, a process that is mediated by an OX2R/cAMP-PKA signaling pathway.


Amacrine Cells/metabolism , Excitatory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/genetics , Orexin Receptors/genetics , Orexins/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Synaptic Transmission/genetics , Amacrine Cells/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Orexin Receptors/metabolism , Orexins/pharmacology , Patch-Clamp Techniques , Phosphoinositide Phospholipase C/antagonists & inhibitors , Phosphoinositide Phospholipase C/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Retinal Bipolar Cells/drug effects , Retinal Ganglion Cells/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/metabolism
12.
Schizophr Bull ; 47(3): 662-671, 2021 04 29.
Article En | MEDLINE | ID: mdl-33398325

It has been suggested that the antipsychotic clozapine may modulate brain glutamate, and that this effect could contribute to its efficacy in treatment-resistant schizophrenia (TRS). The aim of this study was to examine the effects of clozapine on brain glutamate in TRS longitudinally. This study examined individuals with TRS before and 12 weeks after switching from a non-clozapine antipsychotic to treatment with clozapine as part of their normal clinical care. Proton magnetic resonance spectroscopy (1H-MRS) measured concentrations, corrected for voxel tissue content, of glutamate (Glucorr), and glutamate plus glutamine (Glxcorr) in the anterior cingulate cortex (ACC) and right caudate nucleus. Symptoms were monitored using the Positive and Negative Syndrome Scale (PANSS). Of 37 recruited patients (27 men, 39.30 years old, 84% clozapine naïve), 25 completed 1H-MRS at both timepoints. 12 weeks of clozapine was associated with a longitudinal reduction in Glucorr in the caudate (n = 23, F = 7.61 P = .01) but not in the ACC (n = 24, F = 0.02, P = .59). Percentage reduction in caudate Glucorr was positively correlated with percentage improvement in symptoms (total PANSS score, n = 23, r = .42, P = .04). These findings indicate that reductions in glutamate in the caudate nucleus may contribute to symptomatic improvement during the first months of clozapine treatment.


Antipsychotic Agents/pharmacology , Caudate Nucleus , Clozapine/pharmacology , Glutamic Acid , Gyrus Cinguli , Outcome Assessment, Health Care , Schizophrenia , Adult , Antipsychotic Agents/administration & dosage , Caudate Nucleus/diagnostic imaging , Caudate Nucleus/metabolism , Clozapine/administration & dosage , Female , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Glutamine/drug effects , Glutamine/metabolism , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/metabolism , Humans , Longitudinal Studies , Male , Middle Aged , Proton Magnetic Resonance Spectroscopy , Schizophrenia/diagnostic imaging , Schizophrenia/drug therapy , Schizophrenia/metabolism , Schizophrenia/physiopathology
13.
Neuroreport ; 32(4): 312-320, 2021 03 03.
Article En | MEDLINE | ID: mdl-33470770

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.


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
14.
Neuroreport ; 32(2): 77-81, 2021 01 13.
Article En | MEDLINE | ID: mdl-33323835

Isoflurane is an inhaled anesthetic, though its actions at the cellular level remain controversial. By using acute spinal cord slices from adult rats and the whole-cell recording technique, we found that aqueous isoflurane at the minimum alveolar concentration decreased postsynaptic neural excitability and enhanced membrane conductance, while suppressing glutamate release from presynaptic afferent onto substantia gelatinosa (lamina II) neurons in the dorsal horn. The data demonstrate that isoflurane modulates synaptic transmission from peripheral to the spinal cord via both pre- and postsynaptic effects and these actions may underlie its spinal anesthesia.


Anesthetics, Inhalation/pharmacology , Isoflurane/pharmacology , Substantia Gelatinosa/drug effects , Animals , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Patch-Clamp Techniques , Posterior Horn Cells/drug effects , Posterior Horn Cells/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Rats , Spinal Cord Dorsal Horn/cytology , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Substantia Gelatinosa/metabolism , Synaptic Transmission/drug effects
15.
Addict Biol ; 26(2): e12896, 2021 03.
Article En | MEDLINE | ID: mdl-32187792

Vulnerability to drug addiction relies on substantial individual differences. We previously demonstrated that serotonin transporter knockout (SERT-/- ) rats show increased cocaine intake and develop signs of compulsivity. However, the underlying neural mechanisms are not fully understood. Given the pivotal role of glutamate and prefrontal cortex in cocaine-seeking behavior, we sought to investigate the expression of proteins implicated in glutamate neurotransmission in the prefrontal cortex of naïve and cocaine-exposed rats lacking SERT. We focused on the infralimbic (ILc) and prelimbic (PLc) cortices, which are theorized to exert opposing effects on the control over subcortical brain areas. SERT-/- rats, which compared to wild-type (SERT+/+ ) rats show increased ShA and LgA intake short-access (ShA) and long-access (LgA) cocaine intake, were sacrificed 24 h into withdrawal for ex vivo molecular analyses. In the ILc homogenate of SERT-/- rats, we observed a sharp increase in glial glutamate transporter 1 (GLT-1) after ShA, but not LgA, cocaine intake. This was paralleled by ShA-induced increases in GluN1, GluN2A, and GluN2B NMDA receptor subunits and their scaffolding protein SAP102 in the ILc homogenate, but not postsynaptic density, of these knockout animals. In the PLc, we found no major changes in the homogenate; conversely, the expression of GluN1 and GluN2A NMDA receptor subunits was increased in the postsynaptic density under ShA conditions and reduced under LgA conditions. These results point to SERT as a critical regulator of glutamate homeostasis in a way that differs between the subregions investigated, the duration of cocaine exposure as well as the cellular compartment analyzed.


Cocaine/pharmacology , Glutamic Acid/drug effects , Prefrontal Cortex/drug effects , Serotonin Plasma Membrane Transport Proteins/drug effects , Animals , Male , Rats , Synaptic Transmission/drug effects
16.
Addict Biol ; 26(2): e12900, 2021 03.
Article En | MEDLINE | ID: mdl-32212237

N-acetylcysteine (NAC) is a cystine prodrug shown to reduce cocaine- and cue-primed reinstatement of cocaine-seeking behavior in preclinical studies. In this inpatient study, the effects of NAC maintenance versus placebo on cocaine-seeking behavior were examined during cocaine-primed and unprimed self-administration sessions among non-treatment-seeking, cocaine-dependent individuals. Twelve participants completed this double-blind, placebo-controlled, within-subject crossover study. Each participant was maintained for 1 week (Sat-Fri) on NAC (1200-mg TID; 3600 mg/day total) and 1 week on placebo (0-mg TID); medication order was randomized. A subset of participants underwent proton magnetic resonance spectroscopy scans (n = 8) on the third day of medication (Mon) to assess neurochemistry in the rostral anterior cingulate (rACC; voxel = 4.5 cm3 ). In four randomized sessions (Tue-Fri) each week, each participant could earn unit amounts of cocaine (10 mg, fixed) versus money ($0.50 vs. $1.50) on a choice, progressive ratio schedule after insufflating active versus placebo cocaine-priming doses (110 mg vs. 4 mg). Relative to the placebo priming dose, the active cocaine priming dose (110 mg) increased cocaine-seeking behavior (p = .003). NAC reduced cocaine-primed cocaine-seeking behavior compared with placebo levels (p = .044) but did not alter placebo-primed cocaine-seeking behavior. The larger money alternative ($1.50) suppressed cocaine-seeking behavior relative to the smaller money alternative ($0.50; p = .011). Compared with placebo levels, NAC significantly decreased rACC glutamate + glutamine levels (p = .035) and numerically decreased rACC glutamate levels (p = .085). These preliminary findings indicate that NAC suppresses cocaine-seeking behavior in some, but not all, experimental scenarios. Further, our findings suggest NAC may exert its therapeutic effects by modulating excitatory tone in the rACC.


Acetylcysteine/therapeutic use , Cocaine-Related Disorders/drug therapy , Drug-Seeking Behavior/drug effects , Glutamic Acid/drug effects , Glutamine/drug effects , Gyrus Cinguli/drug effects , Acetylcysteine/pharmacology , Adult , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Proton Magnetic Resonance Spectroscopy , Reward
17.
Epilepsy Res ; 167: 106452, 2020 11.
Article En | MEDLINE | ID: mdl-32911258

OBJECTIVE: The purpose of the current analysis was to investigate the direct inhibitory effects of perampanel and other anti-seizure medications (ASMs) on the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartic acid (NMDA), and kainate glutamate receptor subtypes using electrophysiological assessments. METHODS: AMPA receptor subunit-expressing cell lines (hGluA1-4, including two kinds of Q/R RNA-editing variants of hGluA2), NMDA receptor-expressing cells (hNR1/hNR2B), and kainate receptor-expressing cells (hGluK2) were developed in house. The effects of perampanel, and other ASMs including topiramate, phenobarbital, lamotrigine, gabapentin, carbamazepine, valproate, levetiracetam, and lacosamide, on AMPA, NMDA, and kainate receptors were evaluated by automated patch-clamp technique. In the same way, 2,3-dihydroxy-6-nitro-7-sulfamoyl-benzo[f]quinoxaline (NBQX) and GYKI 52466 were evaluated as reference compounds of AMPA receptor antagonists. For the AMPA receptor functional assay, AMPA currents were elicited by AMPA in the presence of cyclothiazide. NMDA with glycine was used as a stimulant for the NMDA receptor assays, while glutamate was used for the kainate receptor assays. The mean 50 % inhibitory concentration (IC50) values were determined based on sigmoidal-curve fitting using GraphPad Prism software. RESULTS: Perampanel inhibited functions of hGluA1-4, but did not inhibit hNR1/hNR2B and hGluK2 up to 25 µM, the maximum soluble concentration. The IC50 values were 660 nM for hGluA1, 780 nM for hGluA2(R), 1200 nM for hGluA2(Q), 1200 nM for hGluA3, and 1800 nM for hGluA4. NBQX and GYKI 52466 also inhibited the function of all AMPA receptor subunits, but did not inhibit hNR1/hNR2B and hGluK2. The IC50 values for NBQX were 880 nM for hGluA1, 290 nM for hGluA2(R), 310 nM for hGluA2(Q), 330 nM for hGluA3, and 630 nM for hGluA4. For GYKI 52466, IC50 values were 25,000 nM for hGluA1, 30,000 nM for hGluA2(R), 42,000 nM for hGluA2(Q), 28,000 nM for hGluA3, and 53,000 nM for hGluA4. Phenobarbital inhibited hGluA2(R) at an IC50 value of 730,000 nM. The majority of other ASMs evaluated in this study did not show a direct inhibitory effect on almost any of the glutamate receptor functions examined up to 1 M. However, lamotrigine and carbamazepine inhibited hNR1/hNR2B function at IC50 values of 930,000 and 1,000,000 nM, respectively. SIGNIFICANCE: Only a few ASMs evaluated in this study showed direct interaction with ionotropic glutamate receptors. Perampanel is the only ASM that had a potent inhibitory effect on all AMPA receptor subtypes, but did not inhibit NMDA or kainate receptor subunits; while phenobarbital inhibited GluA2(R), and carbamazepine and lamotrigine inhibited the NMDA receptor at high concentration ranges.


Nitriles/pharmacology , Pyridones/pharmacology , Receptors, AMPA/drug effects , Receptors, Ionotropic Glutamate/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , Animals , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , N-Methylaspartate/pharmacology , Receptors, AMPA/metabolism , Receptors, Ionotropic Glutamate/metabolism , Receptors, Kainic Acid/antagonists & inhibitors , Receptors, Kainic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
18.
Neurobiol Dis ; 144: 105044, 2020 10.
Article En | MEDLINE | ID: mdl-32798726

Acetylcholine muscarinic receptors (mAChRs) contribute to both the facilitation and inhibition of levodopa-induced dyskinesia operated by striatal cholinergic interneurons, although the receptor subtypes involved remain elusive. Cholinergic afferents from the midbrain also innervate the substantia nigra reticulata, although the role of nigral mAChRs in levodopa-induced dyskinesia is unknown. Here, we investigate whether striatal and nigral M1 and/or M4 mAChRs modulate dyskinesia and the underlying striato-nigral GABAergic pathway activation in 6-hydroxydopamine hemilesioned rats. Reverse microdialysis allowed to deliver the mAChR antagonists telenzepine (M1 subtype preferring), PD-102807 and tropicamide (M4 subtype preferring), as well as the selective M4 mAChR positive allosteric modulator VU0152100 in striatum or substantia nigra, while levodopa was administered systemically. Dyskinetic movements were monitored along with nigral GABA (and glutamate) and striatal glutamate dialysate levels, taken as neurochemical correlates of striato-nigral pathway and cortico-basal ganglia-thalamo-cortical loop activation. We observed that intrastriatal telenzepine, PD-102807 and tropicamide alleviated dyskinesia and inhibited nigral GABA and striatal glutamate release. This was partially replicated by intrastriatal VU0152100. The M2 subtype preferring antagonist AFDX-116, used to elevate striatal acetylcholine levels, blocked the behavioral and neurochemical effects of PD-102807. Intranigral VU0152100 prevented levodopa-induced dyskinesia and its neurochemical correlates whereas PD-102807 was ineffective. These results suggest that striatal, likely postsynaptic, M1 mAChRs facilitate dyskinesia and striato-nigral pathway activation in vivo. Conversely, striatal M4 mAChRs can both facilitate and inhibit dyskinesia, possibly depending on their localization. Potentiation of striatal and nigral M4 mAChR transmission leads to powerful multilevel inhibition of striato-nigral pathway and attenuation of dyskinesia.


Dopamine Agents/adverse effects , Dyskinesia, Drug-Induced/metabolism , Levodopa/adverse effects , Neostriatum/metabolism , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M4/metabolism , Substantia Nigra/metabolism , Allosteric Regulation , Animals , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/physiopathology , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Microdialysis , Muscarinic Antagonists/pharmacology , Neostriatum/drug effects , Neural Pathways , Oxidopamine/toxicity , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/etiology , Parkinsonian Disorders/metabolism , Rats , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M4/antagonists & inhibitors , Substantia Nigra/drug effects , Sympatholytics/toxicity , gamma-Aminobutyric Acid/drug effects , gamma-Aminobutyric Acid/metabolism
19.
Headache ; 60(7): 1259-1272, 2020 07.
Article En | MEDLINE | ID: mdl-32602955

OBJECTIVE: To review the literature on the mechanism of action of onabotulinumtoxinA in chronic migraine. BACKGROUND: OnabotulinumtoxinA is a chronic migraine preventive treatment that significantly reduces headache frequency. The traditional mechanism described for onabotulinumtoxinA - reducing muscle contractions - is insufficient to explain its efficacy in migraine, which is primarily a sensory neurological disease. METHODS: A narrative literature review on the mechanism of action of onabotulinumtoxinA in chronic migraine. RESULTS: Following injection into tissues, onabotulinumtoxinA inhibits soluble N-ethylmaleimide-sensitive fusion attachment protein receptor (SNARE)-mediated vesicle trafficking by cleaving one of its essential proteins, soluble N-ethylmaleimide-sensitive fusion attachment protein (SNAP-25), which occurs in both motor and sensory nerves. OnabotulinumtoxinA inhibits regulated exocytosis of motor and sensory neurochemicals and proteins, as well as membrane insertion of peripheral receptors that convey pain from the periphery to the brain, because both processes are SNARE dependent. OnabotulinumtoxinA can decrease exocytosis of pro-inflammatory and excitatory neurotransmitters and neuropeptides such as substance P, calcitonin gene-related peptide, and glutamate from primary afferent fibers that transmit nociceptive pain and participate in the development of peripheral and central sensitization. OnabotulinumtoxinA also decreases the insertion of pain-sensitive ion channels such as transient receptor potential cation channel subfamily V member 1 (TRPV1) into the membranes of nociceptive neurons; this is likely enhanced in the sensitized neuron. For chronic migraine prevention, onabotulinumtoxinA is injected into 31-39 sites in 7 muscles of the head and neck. Sensory nerve endings of neurons whose cell bodies are located in trigeminal and cervical ganglia are distributed throughout the injected muscles, and are overactive in people with migraine. Through inhibition of these sensory nerve endings, onabotulinumtoxinA reduces the number of pain signals that reach the brain and consequently prevents activation and sensitization of central neurons postulated to be involved in migraine chronification. CONCLUSION: OnabotulinumtoxinA likely acts via sensory mechanisms to treat chronic migraine.


Botulinum Toxins, Type A/pharmacology , Glutamic Acid/drug effects , Migraine Disorders/prevention & control , Neuromuscular Agents/pharmacology , Neuropeptides/drug effects , Neurotransmitter Agents/pharmacology , SNARE Proteins/drug effects , Chronic Disease , Humans
20.
JCI Insight ; 5(13)2020 07 09.
Article En | MEDLINE | ID: mdl-32544091

Musculoskeletal disorders represent the third greatest burden in terms of death and disability in the developed world. Osteoarthritis is the single greatest cause of chronic pain, has no cure, and affects 8.5 and 27 million people in the UK and US, respectively. Osteoarthritis is most prevalent in older people, but as it commonly occurs after joint injury, young people with such injuries are also susceptible. Painful joints are often treated with steroid or hyaluronic acid (HA) injections, but treatments to prevent subsequent joint degeneration remain elusive. In animals, joint injury increases glutamate release into the joint, acting on nerves to cause pain, and joint tissues to cause inflammation and degeneration. This study investigated synovial fluid glutamate concentrations and glutamate receptor (GluR) expression in injured human joints and compared the efficacy of GluR antagonists with current treatments in a mouse model of injury-induced osteoarthritis (ACL rupture). GluRs were expressed in the ligaments and meniscus after knee injury, and synovial fluid glutamate concentrations ranged from 19 to 129 µM. Intra-articular injection of NBQX (GluR antagonist) at the time of injury substantially reduced swelling and degeneration in the mouse ACL rupture model. HA had no effect, and Depo-Medrone reduced swelling for 1 day but increased degeneration by 50%. Intra-articular administration of NBQX modified both symptoms and disease to a greater extent than current treatments. There is an opportunity for repurposing related drugs, developed for CNS disorders and with proven safety in humans, to prevent injury-induced osteoarthritis. This could quickly reduce the substantial burden associated with osteoarthritis.


Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/drug effects , Inflammation/drug therapy , Osteoarthritis/prevention & control , Adolescent , Aged , Aged, 80 and over , Animals , Female , Glutamic Acid/metabolism , Humans , Inflammation/metabolism , Inflammation/prevention & control , Kainic Acid/metabolism , Kainic Acid/pharmacology , Male , Mice, Inbred C57BL , Osteoarthritis/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
...