Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
PLoS One ; 14(12): e0226820, 2019.
Article in English | MEDLINE | ID: mdl-31881077

ABSTRACT

The metabotropic glutamate receptor subtype 1 (mGluR1) is a major subtype of group I mGluRs, which contributes to the development and plasticity of synapses in the brain. In the sensory thalamus, the thalamocortical neuron receives sensory afferents and massive feedback input from corticothalamic (CT) fibers. Notably, mGluR1 is more concentrated in CT synapses in the sensory thalamus. In the visual thalamus, mGluR1 maintains mature afferent synaptic connectivity. However, it is unknown whether mGluR1 contributes to strengthening of immature synapses or weakening of excess synapses during development and whether mGluR1 at CT synapses heterosynaptically regulates the development or refinement of afferent synapses. Here we investigated the effects of knocking out the gene encoding mGluR1 or pharmacologically blocking cortical activity on the development and maintenance of lemniscal synapses, i.e., the somatosensory afferent synapses, in the ventral posteromedial somatosensory thalamus. mGluR1-knockout (KO) mice exhibited delayed developmental strengthening as well as incomplete elimination and remodeling after maturation of lemniscal synapses. Similar to the phenotypes exhibited by mGluR1-KO mice, pharmacological blockade of somatosensory cortical activity from P12 or P21 for 1 week in wild-type mice perturbed elimination or maintenance of lemniscal synapses, respectively. The same manipulation in mGluR1-KO mice failed to induce additional abnormalities in lemniscal synaptic connectivity. These results suggest that activation of mGluR1, driven by CT input, regulates multiple stages of the development of lemniscal synapses, including strengthening, refinement, and maintenance in the somatosensory thalamus.


Subject(s)
Receptors, Metabotropic Glutamate/metabolism , Somatosensory Cortex/physiology , Synapses/physiology , Thalamus/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics
2.
Brain Dev ; 41(7): 567-576, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30954358

ABSTRACT

BACKGROUNDS: Metabotropic glutamate receptors, besides ionotropic receptors, mediate the complicated effect of glutamate on neurogenesis. Previous studies showed that metabotropic glutamate receptor 4 (mGluR4) regulated the proliferation and differentiation of neural stem/progenitor cells in vitro. However, little is known about the expression pattern of mGluR4 on prenatal central nervous system in vivo, especially the human being. METHODS: The normal brain tissues of human fetus were collected and divided into 4 groups according to the gestational age: 9-11 W, 14-16 W, 22-24 W and 32-36 W. Then the expression of mGluR4 was evaluated at mRNA and protein levels by means of PCR or immunohistochemistry method, respectively. The type of cell expressing mGluR4 was further investigated using double-labeling immunofluorescence. RESULTS: RT-PCR showed that the mRNA of mGluR4 could be detected in frontal lobe from 9 W to 32 W and real-time PCR quantificationally demonstrated the mRNA increased with development. Similarly, immnoreactivity was found in all layers of frontal lobe, VZ/SVZ. The intensity scores analysis showed that the staining became stronger and the range extended gradually with development. The double-labeling immunofluorescence showed that mGluR4 was present in neural stem/progenitor cells (nestin-positive cells after 9 W), young neurons (DCX-positive cells after 9 W), mature neurons (NeuN-positive cells in cortex after 32 W), as well as typical astrocytes (GFAP-positive cells in medulla after 32 W). CONCLUSION: These results supply an important evidence that mGluR4 is expressed in prenatal human cerebrum, and main kinds of cells related to neurogenesis are involved in its expression.


Subject(s)
Brain/embryology , Frontal Lobe/embryology , Receptors, Metabotropic Glutamate/metabolism , Brain/metabolism , Cell Differentiation/physiology , Central Nervous System/cytology , Central Nervous System/embryology , Central Nervous System/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Female , Fetal Development/genetics , Frontal Lobe/cytology , Frontal Lobe/metabolism , Glutamic Acid/metabolism , Humans , Immunohistochemistry , Male , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , Pregnancy , Receptors, Metabotropic Glutamate/genetics
3.
Int Immunopharmacol ; 46: 80-86, 2017 May.
Article in English | MEDLINE | ID: mdl-28273557

ABSTRACT

Th17 cells have been categorized as a new lineage of CD4+ T cells, and played a crucial role in the pathogenesis of numerous autoimmune disorders. Type 4 metabotropic glutamate receptor (mGluR4), a member of group III mGluRs, recently has been found to be expressed in many types of immune cells and mediate adaptive immunity. Curcumin has been shown to exhibit potent anti-inflammatory, antimutagenic and anticarcinogenic properties. For the past few years, it has gradually been regarded as an pluripotent immunomodulatory agent that can regulate the activation of immune cells. In the present study, we investigated the efficacy and mechanism of curcumin on Th17 cells. Treatment with curcumin significantly reduced IL-6 and IL-23 production by dendritic cells (DC). Additionally, it had a dramatic reduction in the proliferation of CD4+ T cells co-cultured with DC. Furthermore, expression of the Th17 cells related cytokine profiles (IL-17A and RORγt) was dramatically decreased in curcumin-treated groups. These findings indicated that curcumin inhibited the differentiation and development of Th17 cells. Besides, we found that mGluR4 was constitutively expressed in mouse bone marrow derived DC (BMDC) for the first time. In addition, mGluR4 siRNA-transfected BMDC tipped the balance of T cell differentiation in favor of the Th17 phenotype. We first reported that curcumin increased the mGluR4 expression in mouse BMDC activated with LPS, which likely contributed to the mechanism of inhibiting the Th17 cell differentiation. Our findings suggest that curcumin might be a potential candidate for Th17 related autoimmune disorders.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/drug therapy , Curcumin/therapeutic use , Dendritic Cells/drug effects , Receptors, Metabotropic Glutamate/metabolism , Th17 Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Small Interfering/genetics , Receptors, Metabotropic Glutamate/genetics , Th17 Cells/immunology
4.
CNS Neurosci Ther ; 23(1): 23-32, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27412260

ABSTRACT

AIMS: Glutamatergic transmission may play a critical role in the pathogenesis of Parkinson's disease (PD). Electroacupuncture (EA) has been demonstrated to effectively alleviate PD symptoms. In this study, a potential glutamate-dependent mechanism underlying the therapeutic action of EA was investigated. METHODS: The effects of EA stimulation on motor behaviors, dopamine contents, glutamate release, and group II metabotropic glutamate receptor (mGluR2/3) expression in unilateral 6-hydroxydopamine (6-OHDA)-lesioned rats were examined. RESULTS: Unilateral 6-OHDA lesions of the nigrostriatal system caused a marked increase in glutamate content in the ipsilateral cortex and striatum. mGluR2/3 protein expression and mGluR3 mRNA expression were reduced in the striatum. Noticeably, prolonged EA stimulation at 100 Hz significantly reversed these changes in the striatal glutamate system. Behaviorally, EA improved the motor deficits induced by 6-OHDA lesions. Intrastriatal infusion of an mGluR2/3 antagonist APICA blocked the improving effect of EA. CONCLUSIONS: These data collectively demonstrate that the group II mGluR-mediated glutamatergic transmission in the striatum is sensitive to dopamine depletion and may serve as a substrate of EA for mediating the therapeutic effect of EA in a rat model of PD.


Subject(s)
Corpus Striatum/metabolism , Electroacupuncture , Parkinsonian Disorders/pathology , Parkinsonian Disorders/therapy , Receptors, Metabotropic Glutamate/metabolism , Analysis of Variance , Animals , Chromatography, High Pressure Liquid , Disease Models, Animal , Excitatory Amino Acid Agents/pharmacology , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Male , Motor Activity/physiology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Sympatholytics/toxicity , Time Factors , Tyrosine 3-Monooxygenase/metabolism
5.
Cereb Cortex ; 27(5): 2841-2856, 2017 05 01.
Article in English | MEDLINE | ID: mdl-27178193

ABSTRACT

A unique population of cells, called "lot cells," circumscribes the path of the lateral olfactory tract (LOT) in the rodent brain and acts to restrict its position at the lateral margin of the telencephalon. Lot cells were believed to originate in the dorsal pallium (DP). We show that Lhx2 null mice that lack a DP show a significant increase in the number of mGluR1/lot cells in the piriform cortex, indicating a non-DP origin of these cells. Since lot cells present common developmental features with Cajal-Retzius (CR) cells, we analyzed Wnt3a- and Dbx1-reporter mouse lines and found that mGluR1/lot cells are not generated in the cortical hem, ventral pallium, or septum, the best characterized sources of CR cells. Finally, we identified a novel origin for the lot cells by combining in utero electroporation assays and histochemical characterization. We show that mGluR1/lot cells are specifically generated in the lateral thalamic eminence and that they express mitral cell markers, although a minority of them express ΔNp73 instead. We conclude that most mGluR1/lot cells are prospective mitral cells migrating to the accessory olfactory bulb (OB), whereas mGluR1+, ΔNp73+ cells are CR cells that migrate through the LOT to the piriform cortex and the OB.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Receptors, Metabotropic Glutamate/metabolism , Stem Cells/physiology , Thalamus/cytology , Thalamus/metabolism , Animals , Cell Movement , Cells, Cultured , Embryo, Mammalian , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Pregnancy , Receptors, Metabotropic Glutamate/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Protein p73/genetics , Tumor Protein p73/metabolism
6.
Neuron ; 91(5): 1097-1109, 2016 Sep 07.
Article in English | MEDLINE | ID: mdl-27545713

ABSTRACT

Neural circuits formed during postnatal development have to be maintained stably thereafter, but their mechanisms remain largely unknown. Here we report that the metabotropic glutamate receptor subtype 1 (mGluR1) is essential for the maintenance of mature synaptic connectivity in the dorsal lateral geniculate nucleus (dLGN). In mGluR1 knockout (mGluR1-KO) mice, strengthening and elimination at retinogeniculate synapses occurred normally until around postnatal day 20 (P20). However, during the subsequent visual-experience-dependent maintenance phase, weak retinogeniculate synapses were newly recruited. These changes were similar to those of wild-type (WT) mice that underwent visual deprivation or inactivation of mGluR1 in the dLGN from P21. Importantly, visual deprivation was ineffective in mGluR1-KO mice, and the changes induced by visual deprivation in WT mice were rescued by pharmacological activation of mGluR1 in the dLGN. These results demonstrate that mGluR1 is crucial for the visual-experience-dependent maintenance of mature synaptic connectivity in the dLGN.


Subject(s)
Geniculate Bodies/physiology , Receptors, Metabotropic Glutamate/physiology , Synapses/physiology , Thalamus/physiology , Visual Pathways/physiology , Animals , Carbamates/pharmacology , Geniculate Bodies/drug effects , Glycine/analogs & derivatives , Glycine/pharmacology , Mice , Mice, Knockout , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/genetics , Resorcinols/pharmacology , Retina/physiology , Sensory Deprivation/physiology , Xanthenes/pharmacology
7.
Zhen Ci Yan Jiu ; 41(2): 180-5, 2016 Apr.
Article in Chinese | MEDLINE | ID: mdl-27323449

ABSTRACT

Excitatory amino acids toxicity is an onset causation of cerebral ischemia injury cascade reaction, and eventually leading to brain cell necrosis and apoptosis. Acupuncture is reported to be effective for ischemic stroke in clinical practice and animal experiments, but its mechanism is still under exploring. In this paper the authors introduce the research status of antiexcitatory amino acids toxicity effect of acupuncture in ischemic stroke animals by summarizing its effects on subunits of ionotropic glutamate receptor (NMDA/AMPA) and metabotropic glutamate receptors (mGluRs), and on astrocyte activities. Results indicated that acupuncture intervention may down-regulate the expression levels of cerebral multi-types (NR 1, NR 2 B) of glutamate NMDA receptors, up-regulate expression of glutamate transporter-1, NR 2 A, cannabinoid receptor (CBR) type 1 and 2, and suppress activities of cerebral astrocytes, reduce the content of extracellular glutamate to lower its toxicity and to improve stroke at last. The present paper may provide a reference for acupuncture research on ischemic brain injury.


Subject(s)
Acupuncture Therapy , Excitatory Amino Acids/toxicity , Stroke/therapy , Animals , Brain/metabolism , Excitatory Amino Acids/metabolism , Humans , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Stroke/genetics , Stroke/metabolism
8.
Front Neural Circuits ; 10: 31, 2016.
Article in English | MEDLINE | ID: mdl-27199672

ABSTRACT

Mutation of the metabotropic glutamate receptor type 7 (mGlu7) induces absence-like epileptic seizures, but its precise role in the somatosensory thalamocortical network remains unknown. By combining electrophysiological recordings, optogenetics, and pharmacology, we dissected the contribution of the mGlu7 receptor at mouse thalamic synapses. We found that mGlu7 is functionally expressed at both glutamatergic and GABAergic synapses, where it can inhibit neurotransmission and regulate short-term plasticity. These effects depend on the PDZ-ligand of the receptor, as they are lost in mutant mice. Interestingly, the very low affinity of mGlu7 receptors for glutamate raises the question of how it can be activated, namely at GABAergic synapses and in basal conditions. Inactivation of the receptor activity with the mGlu7 negative allosteric modulator (NAM), ADX71743, enhances thalamic synaptic transmission. In vivo administration of the NAM induces a lethargic state with spindle and/or spike-and-wave discharges accompanied by a behavioral arrest typical of absence epileptic seizures. This provides evidence for mGlu7 receptor-mediated tonic modulation of a physiological function in vivo preventing synchronous and potentially pathological oscillations.


Subject(s)
Cerebral Cortex/cytology , Neural Pathways/physiology , Receptors, Metabotropic Glutamate/metabolism , Thalamus/physiology , Animals , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Cerebral Cortex/physiology , Channelrhodopsins , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Agents/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Transgenic , Mutation/genetics , Neurons/drug effects , Neurons/physiology , Post-Synaptic Density/drug effects , Post-Synaptic Density/genetics , Receptors, GABA-A/physiology , Receptors, Metabotropic Glutamate/genetics , Synaptic Potentials/drug effects , Synaptic Potentials/genetics
9.
J Tradit Chin Med ; 34(5): 566-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25417407

ABSTRACT

OBJECTIVE: To investigate the effect of catgut implantation at acupoints on the expressions of γ-amino butyric acid B receptor (GABA(B)) and metabotropic glutamate receptor 1 (mGluR1) in the brain stem of rats with spasticity after stroke. METHODS: In total, 60 male Sprague-Dawley rats were randomly divided into three groups: a sham group (n = 10), a model group (n = 25) and a treatment group (n = 25). The rats in both the model group and the treatment group were subjected to middle cerebral artery occlusion to establish a model of focal cerebral ischemia. Rats with limb-spasm met the inclusion criteria. Only the left carotid artery was isolated in sham group rats. Three days after modeling, the treatment group was subjected to catgut implantation at Dazhui (GV 14), Guanyuan (CV 4), and Zhongwan (CV 12). Neurological deficit symptoms were assessed with the Zea-Longa neurological deficit score. The Modified Ashworth Scale (MAS), and isolated muscle tone were used to evaluate spasticity before and after treatment. Immunohistochemistry was applied to determine the expression of GABA(B) and mGluR1 in the rat brain stem after treatment. RESULTS: After treatment, neural impairment symptoms had significantly improved in the treatment group when compared to the model group (P < 0.05). Both MAS and isolated muscle tone in the treatment group were significantly decreased when compared with the model group (P < 0.05), and were also lower than before treatment. GABA(B) expression was significantly higher and mGluR1 was lower in the treatment group when compared with the model group (P < 0.01 and P < 0.05, respectively). CONCLUSION: Catgut implantation at Dazhui (GV14), Guanyuan (CV 4), and Zhongwan (CV 12), can relieve limb spasticity by increasing the expression of GABA(B) and reducing the expression of mGluR1 in the brain stem of rats after stroke.


Subject(s)
Brain Stem/metabolism , Muscle Spasticity/genetics , Muscle Spasticity/therapy , Receptors, Metabotropic Glutamate/genetics , Stroke/complications , gamma-Aminobutyric Acid/metabolism , Acupuncture Points , Acupuncture Therapy , Animals , Catgut , Disease Models, Animal , Humans , Male , Muscle Spasticity/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , gamma-Aminobutyric Acid/genetics
10.
Neuropharmacology ; 85: 91-103, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24859611

ABSTRACT

Acute treatment with positive allosteric modulators (PAMs) of mGlu1 and mGlu5 metabotropic glutamate receptors (RO0711401 and VU0360172, respectively) reduces the incidence of spike-and wave discharges in the WAG/Rij rat model of absence epilepsy. However, from the therapeutic standpoint, it was important to establish whether tolerance developed to the action of these drugs. We administered either VU0360172 (3 mg/kg, s.c.) or RO0711401 (10 mg/kg, s.c.) to WAG/Rij rats twice daily for ten days. VU0360172 maintained its activity during the treatment, whereas rats developed tolerance to RO0711401 since the 3rd day of treatment and were still refractory to the drug two days after treatment withdrawal. In response to VU0360172, expression of mGlu5 receptors increased in the thalamus of WAG/Rij rats after 1 day of treatment, and remained elevated afterwards. VU0360172 also enhanced mGlu5 receptor expression in the cortex after 8 days of treatment without changing the expression of mGlu1a receptors. Treatment with RO0711401 enhanced the expression of both mGlu1a and mGlu5 receptors in the thalamus and cortex of WAG/Rij rats after 3-8 days of treatment. These data were different from those obtained in non-epileptic rats, in which repeated injections of RO0711401 and VU0360172 down-regulated the expression of mGlu1a and mGlu5 receptors. Levels of VU0360172 in the thalamus and cortex remained unaltered during the treatment, whereas levels of RO0711401 were reduced in the cortex at day 8 of treatment. These findings suggest that mGlu5 receptor PAMs are potential candidates for the treatment of absence epilepsy in humans.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy, Absence/drug therapy , Epilepsy, Absence/physiopathology , Excitatory Amino Acid Agents/pharmacology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Blotting, Western , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Disease Models, Animal , Drug Tolerance , Electrodes, Implanted , Electroencephalography , Male , Mice, Transgenic , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Rats , Rats, Inbred ACI , Rats, Wistar , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Metabotropic Glutamate/genetics , Thalamus/drug effects , Thalamus/physiopathology , Time Factors
11.
ACS Chem Neurosci ; 5(7): 597-610, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-24798819

ABSTRACT

Recent progress in the discovery of mGlu1 allosteric modulators has suggested the modulation of mGlu1 could offer possible treatment for a number of central nervous system disorders; however, the available chemotypes are inadequate to fully investigate the therapeutic potential of mGlu1 modulation. To address this issue, we used a fluorescence-based high-throughput screening assay to screen an allosteric modulator-biased library of compounds to generate structurally diverse mGlu1 negative allosteric modulator hits for chemical optimization. Herein, we describe the discovery and characterization of a novel mGlu1 chemotype. This series of succinimide negative allosteric modulators, exemplified by VU0410425, exhibited potent inhibitory activity at rat mGlu1 but was, surprisingly, inactive at human mGlu1. VU0410425 and a set of chemically diverse mGlu1 negative allosteric modulators previously reported in the literature were utilized to examine this species disconnect between rat and human mGlu1 activity. Mutation of the key transmembrane domain residue 757 and functional screening of VU0410425 and the literature compounds suggests that amino acid 757 plays a role in the activity of these compounds, but the contribution of the residue is scaffold specific, ranging from critical to minor. The operational model of allosterism was used to estimate the binding affinities of each compound to compare to functional data. This novel series of mGlu1 negative allosteric modulators provides valuable insight into the pharmacology underlying the disconnect between rat and human mGlu1 activity, an issue that must be understood to progress the therapeutic potential of allosteric modulators of mGlu1.


Subject(s)
Excitatory Amino Acid Agents/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Succinimides/pharmacology , Amino Acid Sequence , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cricetulus , Drug Evaluation, Preclinical , Excitatory Amino Acid Agents/chemistry , Fluorescence , HEK293 Cells , High-Throughput Screening Assays , Humans , Mutation , Rats , Receptors, Metabotropic Glutamate/genetics , Species Specificity , Succinimides/chemistry , Transfection
12.
Synapse ; 67(11): 794-800, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23766023

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is important for neuronal survival and plasticity. Incorporation of matured receptor proteins is an integral part of synapse formation. However, whether BDNF increases synthesis and integration of receptors in functional synapses directly is unclear. We are particularly interested in the regulation of the 5-hydroxytryptamine receptor 2A (5-HT(2A)R). This receptor form a functional complex with the metabotropic glutamate receptor 2 (mGluR2) and is recruited to the cell membrane by the corticotrophin-releasing factor receptor 1 (CRF-R1). The effect of BDNF on gene expression for all these receptors, as well as a number of immediate-early genes, was pharmacologically characterized in primary neurons from rat frontal cortex. BDNF increased CRF-R1 mRNA levels up to fivefold, whereas mGluR2 mRNA levels were proportionally downregulated. No effect on 5-HT(2A)R mRNA was seen. The effects were dose-dependent with half-maximal effective concentrations (EC(50)) around 1 ng/ml. After 24 h of incubation with BDNF, CRF-R1 mRNA levels had returned to baseline levels, whereas mGluR2 mRNA levels remained low. A significant reduction of all three receptor transcripts was observed after neuronal depolarization produced by high potassium. This study emphasizes the role of BDNF as an important regulator of receptor compositions in the synapse and provides further evidence that BDNF directly regulates important drug targets involved in cognition and mood.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Frontal Lobe/metabolism , Gene Expression Regulation , Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Genes, Immediate-Early , Membrane Potentials , Neurons/drug effects , Neurons/physiology , Potassium/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Metabotropic Glutamate/genetics , Synapses/metabolism , Transcription, Genetic
13.
Neuropharmacology ; 72: 66-73, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23603202

ABSTRACT

Our knowledge regarding the molecular pathophysiology underlying anxiety disorders remains incomplete. Increasing evidence points to a role of glutamate in anxiety. The group III metabotropic glutamate receptors (mGlu4, mGlu6, mGlu7 and mGlu8 receptors) remain the least investigated glutamate receptor subtypes partially due to a delay in the development of specific pharmacological tools. Early work using knockout animals and pharmacological tools aimed at investigating the role of mGlu7 receptor in the pathophysiology of anxiety disorders has yielded exciting yet not always consistent results. To further investigate the role this receptor plays in anxiety-like behaviour, we knocked down mGlu7 receptor mRNA levels in the adult mouse brain using siRNA delivered via an osmotic minipump. This reduced anxiety-like behaviour in the light-dark box coupled with an attenuation of stress-induced hyperthermia (SIH) and a reduction of the acoustic startle response (ASRs) in the fear-potentiated startle paradigm (FPS). These effects on anxiety-like behaviour were independent of any impairment of locomotor activity and surprisingly, no behavioural changes were observed in the forced swim test (FST), which is in contrast to mGlu7 receptor knockout animals. Furthermore, the previously reported epilepsy-prone phenotype seen in mGlu7 receptor knockout animals was not observed following siRNA-induced knockdown of the receptor. These data suggest targeting mGlu7 receptors with selective antagonist drugs may be an effective and safe strategy for the treatment of anxiety disorders.


Subject(s)
Anxiety/drug therapy , Anxiety/metabolism , RNA, Small Interfering/therapeutic use , Receptors, Metabotropic Glutamate/metabolism , Adaptation, Ocular/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Fear/drug effects , Hyperthermia, Induced/psychology , Male , Mice , Mice, Inbred BALB C , Motor Activity/drug effects , Pentylenetetrazole/toxicity , Receptors, Metabotropic Glutamate/genetics , Reflex, Startle/drug effects , Seizures/chemically induced , Seizures/drug therapy , Stress, Physiological/physiology , Swimming/psychology
14.
Mol Pharmacol ; 83(6): 1166-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508685

ABSTRACT

Serotonin 5-HT(2A) and metabotropic glutamate 2 (mGlu2) are G protein-coupled receptors suspected in the pathophysiology of psychiatric disorders, such as schizophrenia, depression, and suicide. Previous findings demonstrate that mGlu2 mRNA expression is down-regulated in brain cortical regions of 5-HT2A knockout (KO) mice. However, the molecular mechanism responsible for this alteration remains unknown. We show here repressive epigenetic changes at the promoter region of the mGlu2 gene in frontal cortex of 5-HT(2A)-KO mice. Disruption of 5-HT(2A) receptor-dependent signaling in mice was associated with decreased acetylation of histone H3 (H3ac) and H4 (H4ac) and increased tri-methylation of histone H3 at lysine 27 (H3K27me3) at the mGlu2 promoter, epigenetic changes that correlate with transcriptional repression. Neither methylation of histone H3 at lysine 4 (H3K4me1/2/3) nor tri-methylation of histone H3 at lysine 9 (H3K9me3) was affected. We found that Egr1, a transcription factor in which promoter activity was positively regulated by the 5-HT(2A) receptor agonist 4-bromo-3,6-dimethoxybenzocyclobuten-1-yl)methylamine hydrobromide, binds less to the mGlu2 promoter in frontal cortex of 5-HT(2A)-KO, compared with wild-type mice. Furthermore, expression of mGlu2 was increased by viral-mediated gene transfer of FLAG-tagged Egr1 in mouse frontal cortex. Together, these observations suggest that 5-HT(2A) receptor-dependent signaling epigenetically affects mGlu2 transcription in mouse frontal cortex.


Subject(s)
Epigenesis, Genetic , Frontal Lobe/metabolism , Promoter Regions, Genetic , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Metabotropic Glutamate/genetics , Animals , DNA Methylation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Histones/metabolism , Mice , Mice, Knockout , Protein Binding , Protein Processing, Post-Translational , Receptors, Metabotropic Glutamate/metabolism
15.
Cell Death Differ ; 20(3): 396-407, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23175182

ABSTRACT

Drug treatment of malignant gliomas is limited by the intrinsic resistance of glioma stem cells (GSCs) to chemotherapy. GSCs isolated from human glioblastoma multiforme (GBM) expressed metabotropic glutamate receptors (mGlu3 receptors). The DNA-alkylating agent, temozolomide, killed GSCs only if mGlu3 receptors were knocked down or pharmacologically inhibited. In contrast, mGlu3 receptor blockade did not affect the action of paclitaxel, etoposide, cis-platinum, and irinotecan. mGlu3 receptor blockade enabled temozolomide toxicity by inhibiting a phosphatidylinositol-3-kinase/nuclear factor-κB pathway that supports the expression of O(6)-methylguanine-DNA methyltransferase (MGMT), an enzyme that confers resistance against DNA-alkylating agents. In mice implanted with GSCs into the brain, temozolomide combined with mGlu3 receptor blockade substantially reduced tumor growth. Finally, 87 patients with GBM undergoing surgery followed by adjuvant chemotherapy with temozolomide survived for longer time if tumor cells expressed low levels of mGlu3 receptors. In addition, the methylation state of the MGMT gene promoter in tumor extracts influenced survival only in those patients with low expression of mGlu3 receptors in the tumor. These data encourage the use of mGlu3 receptor antagonists as add-on drugs in the treatment of GBM, and suggest that the transcript of mGlu3 receptors should be measured in tumor specimens for a correct prediction of patients' survival in response to temozolomide treatment.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acids/toxicity , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Chemotherapy, Adjuvant , Combined Modality Therapy , DNA Methylation/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Mice , NF-kappa B/metabolism , Neoplastic Stem Cells/cytology , O(6)-Methylguanine-DNA Methyltransferase/genetics , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/genetics , Signal Transduction , Survival Rate , Temozolomide , Transplantation, Heterologous , Tumor Cells, Cultured , Xanthenes/toxicity
16.
Mol Pain ; 8: 77, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23088864

ABSTRACT

BACKGROUND: Pharmacological activation of type-2 metabotropic glutamate receptors (mGlu2 receptors) causes analgesia in experimental models of inflammatory and neuropathic pain. Presynaptic mGlu2 receptors are activated by the glutamate released from astrocytes by means of the cystine/glutamate antiporter (System x(c)(-) or Sx(c)(-)). We examined the analgesic activity of the Sx(c)(-) activator, N-acetyl-cysteine (NAC), in mice developing inflammatory or neuropathic pain. RESULTS: A single injection of NAC (100 mg/kg, i.p.) reduced nocifensive behavior in the second phase of the formalin test. NAC-induced analgesia was abrogated by the Sxc- inhibitor, sulphasalazine (8 mg/kg, i.p.) or by the mGlu2/3 receptor antagonist, LY341495 (1 mg/kg, i.p.). NAC still caused analgesia in mGlu3(-/-) mice, but was inactive in mGlu2(-/-) mice. In wild-type mice, NAC retained the analgesic activity in the formalin test when injected daily for 7 days, indicating the lack of tolerance. Both single and repeated injections of NAC also caused analgesia in the complete Freund's adjuvant (CFA) model of chronic inflammatory pain, and, again, analgesia was abolished by LY341495. Data obtained in mice developing neuropathic pain in response to chronic constriction injury (CCI) of the sciatic nerve were divergent. In this model, a single injection of NAC caused analgesia that was reversed by LY341495, whereas repeated injections of NAC were ineffective. Thus, tolerance to NAC-induced analgesia developed in the CCI model, but not in models of inflammatory pain. The CFA and CCI models differed with respect to the expression levels of xCT (the catalytic subunit of Sx(c)(-)) and activator of G-protein signaling type-3 (AGS3) in the dorsal portion of the lumbar spinal cord. CFA-treated mice showed no change in either protein, whereas CCI mice showed an ipislateral reduction in xCT levels and a bilateral increase in AGS3 levels in the spinal cord. CONCLUSIONS: These data demonstrate that pharmacological activation of Sxc- causes analgesia by reinforcing the endogenous activation of mGlu2 receptors. NAC has an excellent profile of safety and tolerability when clinically used as a mucolytic agent or in the management of acetaminophen overdose. Thus, our data encourage the use of NAC for the experimental treatment of inflammatory pain in humans.


Subject(s)
Acetylcysteine/therapeutic use , Analgesics/therapeutic use , Pain/drug therapy , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics
17.
J Neurosci Res ; 90(10): 1913-23, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22714707

ABSTRACT

Group I metabotropic glutamate receptors (mGluRs) are linked to intracellular Ca(2+) signalling and play important roles related to synaptic plasticity and development. In neurons from the central nucleus of the inferior colliculus (CIC), the activation of these receptors evokes large [Ca(2+) ](i) responses. By using optical imaging of the fluorescent Ca(2+) -sensitive dye Fura-2, we have explored which [Ca(2+) ](i) routes are triggered by group I mGluR activation in young CIC neurons and whether mGluR-induced [Ca(2+) ](i) responses are regulated during postnatal development. In addition, real-time quantitative RT-PCR was used to study the developmental expression of both group I mGluR subtypes, mGluR1 and mGluR5. Application of DHPG, a specific agonist of group I mGluRs, was used on CIC slices from young rats to elicit [Ca(2+) ](i) responses. A majority of responses consisted of an initial thapsigargin-sensitive Ca(2+) peak, related to store depletion, followed by a plateau phase, sensitive to the store-operated Ca(2+) entry blocker 2-APB. During postnatal development, from P6 to P17, DHPG-induced [Ca(2+) ](i) responses changed. The largest Ca(2+) responses were reached at P6, whereas lower peak and plateau responses were found after hearing onset, at P13-P14 and P17. qRT-PCR analysis also revealed important differences in the expression of both mGluR1 and mGluR5 subtypes during development, with the highest levels of both subtypes at P7 and a developmental decrease of both transcripts. Our results suggest both intra- and extracellular routes for [Ca(2+) ](i) increases linked to group I mGluRs in CIC neurons and a regulation of group I mGluR activity and expression during auditory development.


Subject(s)
Auditory Cortex/physiology , Mesencephalon/physiology , Neurons/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Aging/physiology , Animals , Auditory Cortex/cytology , Auditory Cortex/drug effects , Calcium Channels/physiology , Calcium Signaling/physiology , Cell Membrane/metabolism , Cell Membrane/physiology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Down-Regulation/drug effects , In Vitro Techniques , Inferior Colliculi/physiology , Inositol 1,4,5-Trisphosphate/physiology , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/metabolism , Neurons/drug effects , Polymerase Chain Reaction , RNA/biosynthesis , RNA/genetics , RNA/isolation & purification , Rats , Rats, Wistar , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
18.
J Ethnopharmacol ; 140(2): 339-44, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22306289

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Buyang Huanwu Decoction, a traditional Chinese medicine, consists of different herbal medicines, and has been traditionally used for centuries to treat paralysis and stroke. However, its optimal therapeutic time window and the mechanism are still unclear. AIM OF THE STUDY: This study was designed to explore the therapeutic time window and mechanism of Buyang Huanwu Decoction on transient focal cerebral ischemia/reperfusion injury. MATERIALS AND METHODS: Middle cerebral artery occlusion was conducted in male Sprague-Dawley rats, and 40g/kg of Buyang Huanwu Decoction was intragastrically infused at different time points, and the same dose was infused every 24h for 3 days. The level of glutamate in cerebrospinal fluid and the expression of metabotropic glutamate receptor-1 RNA in striatum were detected before, during, and after ischemia/reperfusion. Neurological deficit scores and brain infarction volumes were measured at 72h after reperfusion. RESULT: Cerebral ischemia/reperfusion resulted in significant neurological deficit and extensive cerebral infarct volume, associated with a large amount of glutamate in cerebrospinal fluid and elevation of metabotropic glutamate receptor-1 RNA expression. Buyang Huanwu Decoction significantly suppressed the release of glutamate, and reduced the expression of metabotropic glutamate receptor-1 RNA. The neurological defect score and infarction volume were significantly improved by administration of Buyang Huanwu Decoction, when compared with the Ischemia group. CONCLUSIONS: Administration of Buyang Huanwu Decoction, within 4h of post-transient focal stroke, reduced significant cerebral ischemia/reperfusion damage. The neuroprotective mechanism of Buyang Huanwu Decoction is, in part, associated with the down-regulation of metabotropic glutamate receptor-1 RNA and inhibition of glutamate release resulting from cerebral ischemia.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Glutamic Acid/cerebrospinal fluid , Ischemic Attack, Transient/drug therapy , Magnoliopsida , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Stroke/drug therapy , Animals , Cerebral Infarction/prevention & control , Cerebrovascular Disorders , Drugs, Chinese Herbal/therapeutic use , Ischemic Attack, Transient/metabolism , Male , Nervous System Diseases/prevention & control , Neuroprotective Agents/therapeutic use , Oligochaeta , Phytotherapy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Reperfusion Injury/metabolism , Stroke/metabolism , Time Factors
19.
Neurosci Lett ; 513(1): 17-9, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22342304

ABSTRACT

In the mammalian CNS, the expression of neuronal gap junction protein, connexin 36 (Cx36), increases during the first 2 weeks of postnatal development and then decreases during the following 2 weeks. Recently we showed that the developmental increase in Cx36 expression is augmented by chronic (2 weeks) activation of group II metabotropic glutamate receptors (mGluR), prevented by chronic receptor inactivation, and the receptor-dependent increase in Cx36 expression is regulated via transcriptional control of the Cx36 gene activity. We demonstrate here that acute (60 min) activation of group II mGluRs in developing cortical neuronal cultures causes transient increase in Cx36 protein expression with decrease during the following 24h. However, there is no change in Cx36 mRNA expression. In addition, the data indicate that transient increase in Cx36 expression is due to new protein synthesis. The results suggest that, during development, acute activation of group II mGluRs causes up-regulation of Cx36 via post-transcriptional mechanisms. However, if the receptor activation is sustained, transcriptional activation of the Cx36 gene occurs.


Subject(s)
Aging/physiology , Connexins/biosynthesis , Neurons/metabolism , Receptors, Metabotropic Glutamate/biosynthesis , Amino Acids/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Hypothalamus/cytology , Hypothalamus/drug effects , Mice , Mice, Inbred C57BL , RNA Processing, Post-Transcriptional , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Somatosensory Cortex/cytology , Somatosensory Cortex/drug effects , Somatosensory Cortex/metabolism , Transcriptional Activation/physiology , Gap Junction delta-2 Protein
20.
PLoS One ; 6(11): e27119, 2011.
Article in English | MEDLINE | ID: mdl-22069494

ABSTRACT

A novel glutamate-binding protein was identified in Schistosoma mansoni. The protein (SmGBP) is related to metabotropic glutamate receptors from other species and has a predicted glutamate binding site located within a Venus Flytrap module but it lacks the heptahelical transmembrane segment that normally characterizes these receptors. The SmGBP cDNA was cloned, verified by 5' and 3' Rapid Amplification of cDNA Ends (RACE) and shown to be polyadenylated at the 3'end, suggesting the transcript is full-length. The cloned cDNA was subsequently expressed in bacteria and shown to encode a functional glutamate-binding protein. Other studies, using a specific peptide antibody, determined that SmGBP exists in two forms, a monomer of the expected size and a stable but non-covalent dimer. The monomer and dimer are both present in the membrane fraction of S. mansoni and are resistant to extraction with high-salt, alkaline pH and urea, suggesting SmGBP is either an integral membrane protein or a peripheral protein that is tightly associated with the membrane. Surface biotinylation experiments combined with western blot analyses and confocal immunolocalization revealed that SmGBP localized to the surface membranes of adult male schistosomes, especially the dorsal tubercles. In contrast, we detected little or no expression of SmGBP either in the females or larval stages. A comparative quantitative PCR analysis confirmed that the level of SmGBP expression is several-fold higher in male worms than cercariae, and it is barely detectable in adult females. Together, the results identify SmGBP as a new type of schistosome glutamate receptor that is both gender- and stage-specific. The high-level expression of this protein in the male tubercles suggests a possible role in host-parasite interaction.


Subject(s)
Peptide Fragments/immunology , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Schistosoma mansoni/metabolism , Snails/parasitology , Animals , Biotinylation , Blotting, Western , Cell Membrane/metabolism , Cloning, Molecular , DNA, Complementary/genetics , Female , Glutamic Acid/metabolism , Host-Parasite Interactions , Immunoglobulin G/immunology , Immunoprecipitation , Male , Phylogeny , Protein Conformation , RNA, Messenger/genetics , Rabbits , Real-Time Polymerase Chain Reaction , Receptors, Metabotropic Glutamate/immunology , Schistosoma mansoni/immunology
SELECTION OF CITATIONS
SEARCH DETAIL