Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Neurochem Int ; 150: 105193, 2021 11.
Article in English | MEDLINE | ID: mdl-34571049

ABSTRACT

Imipramine belongs to a group of tricyclic antidepressants (TCAs). It has been also documented that its antidepressant activity connects with the modulation of cytosolic phospholipase A2 (cPLA2) and arachidonic acid (AA) turnover. Through this mechanism, imipramine can indirectly modify glutamate (Glu) transmission. Additionally, it has been shown that chronic treatment with imipramine results in the upregulation of the metabotropic glutamate receptor subtype 5 (mGlu5 receptor) in the hippocampus of rats. Our previous study revealed that manipulation of the AA pathway via inhibition of cyclooxygenase-2 (COX-2) by selective COX-2 inhibitor (NS398) could effectively modulate the behavior of mice treated with imipramine. Here, we hypothesized that COX-2 inhibition could similarly to imipramine influence mGlu5 receptor, and thus NS398 can modulate the effect of imipramine on Glu. Moreover, such regulation changes should correspond with alterations in neurotransmission. Increased cPLA activity after imipramine administration may change the activity of the AA pathway and the endocannabinoid metabolism, e.g., 2-Arachidonyl-glycerol (2-AG). To verify the idea, mGlu5 receptor level was investigated in the hippocampus (HC) and prefrontal cortex (PFC) of mice treated for 7 or 14 days with imipramine and/or COX-2 inhibitor: NS398. Western blot and PCR analyses were conducted. Moreover, the excitatory (Glu) and inhibitory (gamma-aminobutyric acid; GABA) neurotransmitters were measured using HPLC and 2-AG using ELISA. A time-dependent change in mGlu5 receptor and COX-2 protein level, COX-2 expression, and 2-AG level in the PFC after imipramine administration was found. Up-regulation of mGlu5 receptor after NS398 was found in HC and PFC. A structure-dependent shift between excitatory vs. inhibitory transmission was detected when NS398 and imipramine were co-administered.


Subject(s)
Brain/metabolism , Cyclooxygenase 2/biosynthesis , Imipramine/pharmacology , Nitrobenzenes/pharmacology , Receptor, Metabotropic Glutamate 5/biosynthesis , Sulfonamides/pharmacology , Up-Regulation/physiology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Brain/drug effects , Cyclooxygenase Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Receptor, Metabotropic Glutamate 5/agonists , Up-Regulation/drug effects
2.
Neurochem Int ; 150: 105191, 2021 11.
Article in English | MEDLINE | ID: mdl-34547325

ABSTRACT

Spinal cord ischemia-reperfusion injury (SCIRI) can cause dramatic neuron loss and lead to paraplegia in patients. In this research, the role of mGluR5, a member of the metabotropic glutamate receptors (mGluRs) family, was investigated both in vitro and in vivo to explore a possible method to treat this complication. In vitro experiment, after activating mGluR5 via pretreating cells with (RS)-2-Chloro-5-hydroxyphenylglycine (CHPG) and 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB), excitotoxicity induced by glutamate (Glu) was attenuated in primary spinal cord neurons, evidenced by higher neuron viability, decreased lactate dehydrogenase (LDH) release and less detected TUNEL-positive cells. According to Western Blot (WB) results, Glu treatment resulted in a high level of large-conductance Ca2+- and voltage-activated K+ (BK) channels, with activation relying on the mGluR5-IP3R (inositol triphosphate) pathway. In vivo part, a rat model of SCIRI was built to further investigate the role of mGluR5. After pretreating them with CHPG and CDPPB, the rats showed markedly lower spinal water content, attenuated motor neuron injury in the spinal cord of L4 segments, and better neurological function. This effect could be partially reversed by paxilline, a blocker of BK channels. In addition, activating BK channels alone using specific openers: NS1619 or NS11021 can protect spinal cord neurons from injury induced by either SCIRI or Glu. In conclusion, in this research, we proved that mGluR5 exerts a protective role in SCIRI, and this effect partially works via IP3R-mediated activation of BK channels.


Subject(s)
Adenosylhomocysteinase/biosynthesis , Large-Conductance Calcium-Activated Potassium Channels/biosynthesis , Neuroprotection/physiology , Receptor, Metabotropic Glutamate 5/biosynthesis , Reperfusion Injury/metabolism , Spinal Cord Ischemia/metabolism , Animals , Benzamides/pharmacology , Cells, Cultured , Excitatory Amino Acid Agonists/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Neuroprotection/drug effects , Paxillin/pharmacology , Pyrazoles/pharmacology , Rats , Receptor, Metabotropic Glutamate 5/agonists , Reperfusion Injury/prevention & control , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Ischemia/prevention & control
3.
Neurotoxicology ; 84: 14-29, 2021 05.
Article in English | MEDLINE | ID: mdl-33571554

ABSTRACT

Due to limitations in early diagnosis and treatments of Parkinson's disease (PD), it is necessary to explore the neuropathological changes that occur early in PD progression and to design neuroprotective therapies to prevent or delay the ongoing degeneration process. Metabotropic glutamate receptor 5 (mGlu5) has shown both diagnostic and therapeutic potential in preclinical studies on PD. Clinical trials using mGlu5 negative allosteric modulators to treat PD have, however, raised limitations about the neuroprotective role of mGlu5. It is likely that mGlu5 has different regulatory roles in different stages of PD. Here, we investigated a protective role of cystic fibrosis transmembrane conductance regulator-associated ligand (CAL) in the progression of PD by differential regulation of mGlu5 expression and activity to protect against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity. Following treatment with 6-OHDA, mGlu5 and CAL expressions were elevated in the early stage and reduced in the late stage, both in vitro and in vivo. Activation of mGlu5 in the early stage by (RS)-2-chloro-5-hydroxyphenylglycine, or blocking mGlu5 in the late stage by 2-methyl-6-(phenylethynyl) pyridine, increased cell survival and inhibited apoptosis, but these effects were significantly weakened by knockdown of CAL. CAL alleviated 6-OHDA-induced neurotoxicity by regulating mGlu5-mediated signaling pathways, thereby maintaining the physiological function of mGlu5 in different disease stages. In PD rat model, CAL deficiency aggravated 6-OHDA toxicity on dopaminergic neurons and increased motor dysfunction because of lack of regulation of mGlu5 activity. These data reveal a potential mechanism by which CAL specifically regulates the opposite activity of mGlu5 in progression of PD to protect against neurotoxicity, suggesting that CAL is a favorable endogenous target for the treatment of PD.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Disease Progression , Dopaminergic Neurons/metabolism , Oxidopamine/toxicity , Parkinsonian Disorders/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Animals , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Excitatory Amino Acid Antagonists/pharmacology , Ligands , Male , Mice , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Parkinsonian Disorders/prevention & control , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
4.
Nat Neurosci ; 23(3): 386-397, 2020 03.
Article in English | MEDLINE | ID: mdl-32066985

ABSTRACT

Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.


Subject(s)
DNA Repeat Expansion/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Trinucleotide Repeats/genetics , Animals , Cell Line , Cell Survival/genetics , Female , Fragile X Mental Retardation Protein/biosynthesis , Induced Pluripotent Stem Cells , Male , Mice , Neurons/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics
5.
Article in English | MEDLINE | ID: mdl-31404590

ABSTRACT

Type-5 metabotropic glutamate receptors (mGlu5) have been implicated in the mechanism of resilience to stress. They form part of the postsynaptic density (PSD), a thickening of the glutamatergic synapse that acts as a multimodal hub for multiple cellular signaling. Perinatal stress in rats triggers alterations that make adult offspring less resilient to stress. In the present study, we examined the expression of gene encoding the mGlu5 (Grm5), as well as those encoding the short and long isoforms of Homer proteins in different brain regions of the offspring of dams exposed to repeated episodes of restraint stress during pregnancy ("perinatally stressed" or PRS offspring). To this end, we investigated unconditioned behavioral response using the light/dark box test, as well as the expression of PSD genes (Homer1a, Homer1b, and Grm5), in the medial prefrontal cortex, cortex, caudate-putamen, amygdala, and dorsal hippocampus. PRS rats spent significantly less time in the light area than the control group. In the amygdala, Homer1a mRNA levels were significantly increased in PRS rats, whereas Homer1b and Grm5 mRNA levels were reduced. In contrast, the transcript encoding for Homer1a was significantly reduced in the medial prefrontal cortex, caudate-putamen, and dorsal hippocampus of PRS rats. We also evaluated the relative ratio between Homer1a and Homer1b/Grm5 expression, finding a significant shift toward the expression of Homer1a in the amygdala and toward Homer1b/Grm5 in the other brain regions. These topographic patterns of Homer1a, Homer1b, and mGlu5 gene expression were significantly correlated with risk-taking behavior measured in the light/dark box test. Remarkably, in the amygdala and in other brain regions, Homer1b and Grm5 expression showed positive correlation with time spent in the light box, whereas Homer1a in the amygdala showed a negative correlation with risk-taking behavior, in contrast with all other brain regions analyzed, wherein these correlations were positive. These results suggest that perinatal stress programs the developmental expression of PSD molecules involved in mGlu5 signaling in discrete brain regions, with a predominant role for the amygdala.


Subject(s)
Brain/metabolism , Homer Scaffolding Proteins/biosynthesis , Post-Synaptic Density/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Stress, Psychological/metabolism , Stress, Psychological/psychology , Animals , Female , Gene Expression , Homer Scaffolding Proteins/genetics , Male , Post-Synaptic Density/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/genetics , Restraint, Physical
6.
Neurochem Int ; 129: 104515, 2019 10.
Article in English | MEDLINE | ID: mdl-31369778

ABSTRACT

Lysophosphatidic acid (LPA) is a glycerophospholipid that can be detected in serum, saliva and cerebrospinal fluid. However, the effect of LPA on neuronal death and survival has not been fully determined. In the present study, we investigated the potential neurotoxic effect of LPA in primary cultured cortical neurons. Treatment with LPA (0.5, 1 and 5 µM) markedly decreased neuronal viability, increased lactate dehydrogenase (LDH) release and promoted apoptosis in cortical neurons. The results of western blot showed that LPA increased the expression of endoplasmic reticulum (ER) stress associated factors, and the protein misfolding inhibitor 4-phenylbutyric acid (4-PBA) attenuated LPA-induced toxicity. In addition, treatment with LPA did not alter the expression and distribution of Homer1 in cortical neurons. The protein levels of metabotropic glutamate receptor 1 (mGluR1), but not metabotropic glutamate receptor 5 (mGluR5), were significantly increased by LPA at 12 and 24 h after treatment. Knockdown of Homer1 using specific siRNA partially prevented the LPA-induced neurotoxicity and ER stress. Furthermore, the results of Ca2+ imaging showed that treatment with LPA induced intracellular Ca2+ release, which could be partially prevented by 4-PBA and downregulation of Homer1. The LPA-induced intracellular Ca2+ release was associated with ER Ca2+ release through the Homer1-mGluR1 pathway. In summary, our results showed that LPA treatment induced ER stress and apoptosis in cortical neurons, and its neurotoxicity was partially mediated by Ca2+ release from the ER via the Homer1/mGluR1 pathway.


Subject(s)
Calcium Signaling/drug effects , Endoplasmic Reticulum Stress/drug effects , Homer Scaffolding Proteins/physiology , Lysophospholipids/toxicity , Nerve Tissue Proteins/physiology , Neurons/drug effects , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Down-Regulation , Female , Homer Scaffolding Proteins/antagonists & inhibitors , Homer Scaffolding Proteins/biosynthesis , Homer Scaffolding Proteins/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Neurons/metabolism , Phenylbutyrates/pharmacology , Primary Cell Culture , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics
7.
Neurosci Lett ; 705: 143-150, 2019 07 13.
Article in English | MEDLINE | ID: mdl-31029678

ABSTRACT

Hyperexcitability in the corticostriatal glutamatergic pathway may have a pivotal role in the pathogenesis of Parkinson's disease (PD). Metabotropic glutamate receptors (mGluRs) modulate glutamate transmission by both pre- and postsynaptic mechanisms, making them attractive targets for modifying pathological changes in the corticostriatal pathway. Exercise reportedly alleviates motor dysfunction and induced neuroplasticity in glutamatergic transmission. Here, the mGluR-mediated plasticity mechanism underlying behavioral improvement by exercise intervention was investigated. The experimental models were prepared by 6-hydroxydopamine injection into the right medial forebrain bundle. The models were evaluated with the apomorphine-induced rotation test. Starting 2 weeks postoperatively, exercise intervention was applied to the PD + Ex group for 4 weeks. The exercise-intervention effects on locomotor behavior, glutamate levels, and mGluR (mGluR2/3 and mGluR5) expression in hemiparkinsonian rats were investigated. The results showed that hemiparkinsonian rats have a significant increase in extracellular glutamate levels in the lesioned-lateral striatum. MGluR2/3 protein expression was reduced while mGluR5 protein expression was increased in the striatum. Notably, treadmill exercise markedly reversed these abnormal changes in the corticostriatal glutamate system and promoted motor performance in PD rats. These findings suggest that mGluR-mediated glutamatergic transmission in the corticostriatal pathway may serve as an attractive target for exercise-induced neuroplasticity in hemiparkinsonian rats.


Subject(s)
Corpus Striatum/physiopathology , Exercise Therapy , Locomotion/physiology , Parkinson Disease/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptors, Metabotropic Glutamate/biosynthesis , Animals , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Male , Medial Forebrain Bundle/drug effects , Oxidopamine , Rats
8.
Toxicol Lett ; 288: 71-81, 2018 May 15.
Article in English | MEDLINE | ID: mdl-29458170

ABSTRACT

Clinical epidemiological studies have shown that there is a link between Parkinson's disease (PD) and cancer, but how PD regulates cancer development remains unknown. In our study, the effect of metabotropic glutamate receptor 5 (mGlu5) on hepatoma was explored in a rotenone-induced PD model both in vitro and in vivo. We found that conditioned media derived from MN9D dopaminergic neuronal cells by rotenone-induced toxicity inhibited the growth, migration, invasion and promoted apoptosis of Hepa1-6 cells, which corresponded with decreased expression of mGlu5. Furthermore, treatment with 2-methyl-6-(phenylethynyl)pyridine (MPEP), a mGlu5 antagonist and knockdown of mGlu5, further reduced ATP levels and migration distance, and increased cleavage of caspase-3 in Hepa1-6 cells. Additionally, we found that conditioned media derived from rotenone-treated MN9D dopaminergic neuronal cells enhanced reactive oxygen species (ROS) generation and JNK phosphorylation, which could be further increased by MPEP treatment, and attenuated by mGlu5 agonist, (RS)-2-Chloro-5-hydroxyphenylglycine (CHPG) and ROS scavenger, N-acetyl-l-cysteine (NAC). The results indicated that down-regulation of mGlu5 promoted cell apoptosis through the intracellular ROS/JNK signaling pathway in a rotenone-induced cellular PD model. These findings were confirmed in vivo in a rotenone-induced rat model of PD combined with diethylnitrosamine (DEN)-induced hepatoma. Expression of Ki67 was decreased, and the levels of caspase-3 and p-JNK were increased in this model, which was accompanied by a decrease in protein expression of mGlu5. The study suggest that negative regulation of mGlu5 may inhibit hepatoma development in a rotenone-induced PD model, and as such may help with our further understanding of the correlation between PD and cancer.


Subject(s)
Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/prevention & control , Neurotoxicity Syndromes/pathology , Parkinson Disease, Secondary/pathology , Receptor, Metabotropic Glutamate 5/drug effects , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Gene Knockdown Techniques , Male , Mice , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/psychology , Pyridines/toxicity , Rats , Rats, Sprague-Dawley , Reactive Nitrogen Species/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics , Rotenone , Signal Transduction/drug effects , Uncoupling Agents
9.
Drug Alcohol Depend ; 184: 1-11, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29324247

ABSTRACT

Binge alcohol-drinking elicits symptoms of negative affect such as anxiety upon cessation, which is a source of negative reinforcement for perpetuating this pattern of alcohol abuse. Binge-induced anxiety during early (24 h) withdrawal is associated with increased expression of metabotropic glutamate receptor 5 (mGlu5) within the nucleus accumbens shell (AcbSh) of adult male mice, but was unchanged in anxiety-resilient adolescents. Herein, we determined the role of mGlu5 signaling in withdrawal-induced anxiety via pharmacological manipulation using the mGlu5 negative allosteric modulator MTEP and the positive allosteric modulator CDPPB. Adult (PND 56) and adolescent (PND 28) male C57BL/6J mice binge-drank for 14 days under 3-bottle-choice procedures for 2 h/day; control animals drank water only. Approximately 24 h following the final alcohol presentation, animals were treated with 30 mg/kg IP MTEP, CDPPB, or vehicle and then tested, thirty minutes later, for behavioral signs of anxiety. Vehicle-treated binge-drinking adults exhibited hyperanxiety in all paradigms, while vehicle-treated binge-drinking adolescents did not exhibit withdrawal-induced anxiety. In adults, 30 mg/kg MTEP decreased alcohol-induced anxiety across paradigms, while 3 mg/kg MTEP was anxiolytic in adult water controls. CDPPB was modestly anxiogenic in both alcohol- and water-drinking mice. Adolescent animals showed minimal response to either CDPPB or MTEP, suggesting that anxiety in adolescence may be mGlu5-independent. These results demonstrate a causal role for mGlu5 in withdrawal-induced anxiety in adults and suggest age-related differences in the behavioral pharmacology of the negative reinforcing properties of alcohol.


Subject(s)
Anxiety/metabolism , Binge Drinking/metabolism , Ethanol/toxicity , Receptor, Metabotropic Glutamate 5/biosynthesis , Substance Withdrawal Syndrome/metabolism , Age Factors , Animals , Anxiety/chemically induced , Anxiety/drug therapy , Benzamides/pharmacology , Benzamides/therapeutic use , Binge Drinking/drug therapy , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Substance Withdrawal Syndrome/drug therapy , Thiazoles/pharmacology , Thiazoles/therapeutic use
10.
Sci Rep ; 7(1): 9743, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851991

ABSTRACT

Patients with chronic pain easily accompany the negative mood symptoms such as depression and anxiety, and these disturbances in turn affect the aversive perception of pain. However, the underlying mechanisms are largely unknown. We hypothesized that the alteration of metabotropic glutamate receptor 5 (mGluR5) in the brain region underlies such a comorbidity of aversive states. We scanned the brain of chronic neuropathic pain model rats using positron emission tomography (PET) technique with an mGluR5-selective radiotracer [11C] ABP688 and found various brain regions with higher or lower level of mGluR5 compared to control rats. Among the brain areas, a prominent upregulation of mGluR5 was shown in the prelimbic region (PrL) of the medial prefrontal cortex (mPFC) of chronic neuropathic pain animals. A pharmacological blockade of upregulated mGluR5 in the PrL ameliorated the negative symptoms including tactile hypersensitivity and depressive-like behavior, which relieved the subjects from the unpleasant state of chronic neuropathic pain condition. Conversely, lentiviral overexpression of the mGluR5 in the PrL of naïve rats successfully induced comorbid pain and negative moods. Our data provide deeper insight into the shared mechanism of pain perception and negative emotions, identifying a therapeutic target for the treatment of chronic pain and mood disorders.


Subject(s)
Affect , Neuralgia/physiopathology , Pain Perception , Prefrontal Cortex/physiology , Receptor, Metabotropic Glutamate 5/biosynthesis , Up-Regulation , Animals , Disease Models, Animal , Positron-Emission Tomography , Prefrontal Cortex/diagnostic imaging , Rats , Spinal Nerves , Wounds and Injuries
11.
Sci Rep ; 7(1): 3546, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615682

ABSTRACT

Müller cells maintain retinal synaptic homeostasis by taking up glutamate from the synaptic cleft and transporting glutamine back to the neurons. To study the interaction between Müller cells and photoreceptors, we injected either DL-α-aminoadipate or L-methionine sulfoximine-both inhibitors of glutamine synthetase-subretinally in rats. Following injection, the a-wave of the electroretinogram (ERG) was attenuated, and metabotropic glutamate receptor 5 (mGluR5) was activated. Selective antagonism of mGluR5 by 2-methyl-6-(phenylethynyl)-pyridine increased the ERG a-wave amplitude and also increased rhodopsin expression. Conversely, activation of mGluR5 by the agonist, (R,S)-2-chloro-5-hydroxyphenylglycine, decreased both the a-wave amplitude and rhodopsin expression, but upregulated expression of Gq alpha subunit and phospholipase C ßIII. Overexpression of mGluR5 reduced the inward-rectifying potassium ion channel (Kir) current and decreased the expression of Kir4.1 and aquaporin-4 (AQP4). Further experiments indicated that mGluR5 formed a macromolecular complex with these two membrane channels. Lastly, increased expression of mGluR5 was found in Royal College of Surgeons rats-a model of retinitis pigmentosa (RP). Inhibition of mGluR5 in this model restored the amplitude of ERG features, and reduced the expression of glial fibrillary acidic protein. These results suggest that mGluR5 may be worth considering as a potential therapeutic target in RP.


Subject(s)
Ependymoglial Cells/physiology , Glutamic Acid/metabolism , Photoreceptor Cells/physiology , Receptor, Metabotropic Glutamate 5/biosynthesis , Retina/physiology , Action Potentials/drug effects , Animals , Cells , Down-Regulation , Electroretinography , Ependymoglial Cells/metabolism , Glutamate-Ammonia Ligase/antagonists & inhibitors , Photoreceptor Cells/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors
12.
Neuropharmacology ; 115: 10-19, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27211252

ABSTRACT

Trafficking of G protein-coupled receptors (GPCRs) to the plasma membrane is a pivotal process to fulfill their biological functions. Metabotropic glutamate receptors (mGluRs; mGluR1-8) are expressed throughout the CNS and are important for modulating synaptic transmission and plasticity. Group I mGluRs, including mGluR1 and mGluR5, have long intracellular C-terminal tails containing multiple protein binding domains and sites for phosphorylation and ER retention. We have now investigated some of the structural determinants for mGluR5 trafficking to the plasma membrane by studying a series of truncations and ligand binding mutants. We also take advantage of dimer formation between the extracellular domain (ECD) of mGluR5 and design an ECD based surface-binding assay to evaluate dimerization and surface expression of mGluR5 containing various truncations or point mutations. We found that the C terminus is not essential for mGluR5 surface expression. In contrast, the 7th transmembrane domain (TM7) plays a critical role in its surface expression in both heterologous cells and neurons. Furthermore, a ligand binding mutation within the ECD of mGluR5 (Y64A/T174A) that blocks ligand binding impairs both surface expression and dimerization of mGluR5 in neurons. The integrity of both the whole 7TM domain and the C- terminal tail of mGluR5 are also important for stabilizing dimerization with the ECD. Thus multiple domains regulate dimerization and trafficking of mGluR5. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.


Subject(s)
Cell Membrane/genetics , Cell Membrane/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptor, Metabotropic Glutamate 5/genetics , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Gene Expression , HEK293 Cells , Hippocampus/metabolism , Humans , Neurons/metabolism , Phosphorylation/physiology , Protein Multimerization/physiology , Protein Transport/physiology , Rats , Receptor, Metabotropic Glutamate 5/chemistry
13.
Sci Rep ; 6: 34391, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721389

ABSTRACT

Group 1 metabotropic glutamate receptors (mGluR1/mGluR5) play an integral role in neurodevelopment and are implicated in psychiatric disorders, such as schizophrenia. mGluR1 and mGluR5 are expressed as homodimers, which is important for their functionality and pharmacology. We examined the protein expression of dimeric and monomeric mGluR1α and mGluR5 in the prefrontal cortex (PFC) and hippocampus throughout development (juvenile/adolescence/adulthood) and in the perinatal phencyclidine (PCP) model of schizophrenia. Under control conditions, mGluR1α dimer expression increased between juvenile and adolescence (209-328%), while monomeric levels remained consistent. Dimeric mGluR5 was steadily expressed across all time points; monomeric mGluR5 was present in juveniles, dramatically declining at adolescence and adulthood (-97-99%). The mGluR regulators, Homer 1b/c and Norbin, significantly increased with age in the PFC and hippocampus. Perinatal PCP treatment significantly increased juvenile dimeric mGluR5 levels in the PFC and hippocampus (37-50%) but decreased hippocampal mGluR1α (-50-56%). Perinatal PCP treatment also reduced mGluR1α dimer levels in the PFC at adulthood (-31%). These results suggest that Group 1 mGluRs have distinct dimeric and monomeric neurodevelopmental patterns, which may impact their pharmacological profiles at specific ages. Perinatal PCP treatment disrupted the early expression of Group 1 mGluRs which may underlie neurodevelopmental alterations observed in this model.


Subject(s)
Gene Expression Regulation, Developmental , Hippocampus/embryology , Neurogenesis , Phencyclidine/adverse effects , Prefrontal Cortex/embryology , Protein Multimerization , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptors, Metabotropic Glutamate/biosynthesis , Schizophrenia , Animals , Female , Male , Phencyclidine/pharmacology , Rats , Rats, Sprague-Dawley , Schizophrenia/chemically induced , Schizophrenia/metabolism
14.
Pharmacol Biochem Behav ; 146-147: 50-9, 2016.
Article in English | MEDLINE | ID: mdl-27178815

ABSTRACT

PURPOSE: Ethosuximide (ETX) is the drug of choice for the treatment of patients with absence seizures - taking into account both its efficacy, tolerability and antiepileptogenic properties. However, 47% of subjects failed in ETX-therapy, and most antiepileptic drugs have cognitive side effects. VU0360172, a positive allosteric modulator (PAM) of mGluR5, has been proposed as a new anti-absence drug. Here it is investigated whether anti-epileptogenesis induced by ETX alters the sensitivity of VU0360172, and whether cognition is affected during and after chronic ETX treatment. METHOD: EEG's were recorded before and after a challenge with VU0360172 in chronic ETX and in control WAG/Rij rats during and after treatment. Rats were also exposed to a cue discrimination learning task in a Y-maze both during and after treatment. At the end of the experiment, mGlu5 receptors were quantified by Western Blot analysis. RESULTS: Antiepileptogenesis was successfully induced by ETX and VU0360172 showed a time and dose dependent anti-absence action in the control group. VU0360172 kept its anti-absence action in chronic ETX treated rats both during and after treatment, without time and dose dependency. This anti-absence effect of VU0360172 in both groups matched the lack of differences in mGluR5 expression. Chronic ETX enhanced the number of completed trials, the number of correct choices in the Y-maze and the number of consumed sucrose pallets. SIGNIFICANCE: VU0360172 maintains its anti-absence effects after chronic treatment; as such, VU0360172 can also be used as a adjunctive therapy in patients with absence epilepsy. The enhanced motivation and cognitive performance by ETX might be mediated by the antidepressant action of ETX as expressed by an increase in the rewarding properties of sucrose pallets.


Subject(s)
Anticonvulsants/pharmacology , Epilepsy, Absence/prevention & control , Ethosuximide/pharmacology , Niacinamide/analogs & derivatives , Receptor, Metabotropic Glutamate 5/biosynthesis , Animals , Cerebral Cortex/metabolism , Cognition/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Electroencephalography/drug effects , Food Preferences/drug effects , Male , Maze Learning/drug effects , Motor Activity/drug effects , Niacinamide/pharmacology , Rats , Thalamus/metabolism , Time Factors
15.
Hum Mol Genet ; 25(10): 1990-2004, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26936821

ABSTRACT

Rett syndrome (RS) is a neurodevelopmental disorder that shares many symptomatic and pathological commonalities with idiopathic autism. Alterations in protein synthesis-dependent synaptic plasticity (PSDSP) are a hallmark of a number of syndromic forms of autism; in the present work, we explore the consequences of disruption and rescue of PSDSP in a mouse model of RS. We report that expression of a key regulator of synaptic protein synthesis, the metabotropic glutamate receptor 5 (mGlu5) protein, is significantly reduced in both the brains of RS model mice and in the motor cortex of human RS autopsy samples. Furthermore, we demonstrate that reduced mGlu5 expression correlates with attenuated DHPG-induced long-term depression in the hippocampus of RS model mice, and that administration of a novel mGlu5 positive allosteric modulator (PAM), termed VU0462807, can rescue synaptic plasticity defects. Additionally, treatment of Mecp2-deficient mice with VU0462807 improves motor performance (open-field behavior and gait dynamics), corrects repetitive clasping behavior, as well as normalizes cued fear-conditioning defects. Importantly, due to the rationale drug discovery approach used in its development, our novel mGlu5 PAM improves RS phenotypes and synaptic plasticity defects without evoking the overt adverse effects commonly associated with potentiation of mGlu5 signaling (i.e. seizures), or affecting cardiorespiratory defects in RS model mice. These findings provide strong support for the continued development of mGlu5 PAMs as potential therapeutic agents for use in RS, and, more broadly, for utility in idiopathic autism.


Subject(s)
Autistic Disorder/genetics , Receptor, Metabotropic Glutamate 5/genetics , Rett Syndrome/genetics , Seizures/genetics , Adult , Allosteric Regulation/genetics , Animals , Autistic Disorder/drug therapy , Autistic Disorder/pathology , Autopsy , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Hippocampus/drug effects , Hippocampus/pathology , Humans , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Motor Cortex/drug effects , Motor Cortex/pathology , Neuronal Plasticity/drug effects , Pyrazoles/administration & dosage , Pyrimidinones/administration & dosage , Receptor, Metabotropic Glutamate 5/biosynthesis , Rett Syndrome/drug therapy , Rett Syndrome/pathology , Seizures/drug therapy , Seizures/pathology , Signal Transduction/drug effects , Young Adult
16.
Exp Neurol ; 279: 116-126, 2016 May.
Article in English | MEDLINE | ID: mdl-26896834

ABSTRACT

More efficient or translationally relevant approaches are needed to model acquired temporal lobe epilepsy (TLE) in genetically tractable mice. The high costs associated with breeding and maintaining transgenic, knock-in, or knock-out lines place a high value on the efficiency of induction and animal survivability. Herein, we describe our approaches to model acquired epilepsy in C57BL/6J mice using repeated, low-dose kainate (KA) administration paradigms. Four paradigms (i.p.) were tested for their ability to induce status epilepticus (SE), temporal lobe pathology, and the development of epilepsy. All four paradigms reliably induce behavioral and/or electrographic SE without mortality over a 7d period. Two of the four paradigms investigated produce features indicative of TLE pathology, including hippocampal cell death, widespread astrogliosis, and astrocyte expression of mGluR5, a feature commonly reported in TLE models. Three of the investigated paradigms were able to produce aberrant electrographic features, such as interictal spiking in cortex. However, only one paradigm, previously published by others, produces spontaneous recurrent seizures over an eight week period. Presentation of spontaneous seizures is rare (N=2/14), with epilepsy preferentially developing in animals having a high number of seizures during SE. Overall, repeated, low-dose KA administration improves the efficiency and pathological relevance of a systemic KA insult, but does not produce a robust epilepsy phenotype under the experimental paradigms described herein.


Subject(s)
Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/pathology , Excitatory Amino Acid Agonists/toxicity , Kainic Acid/toxicity , Animals , Astrocytes/pathology , Cell Death/drug effects , Disease Models, Animal , Electroencephalography , Glial Fibrillary Acidic Protein/metabolism , Gliosis/chemically induced , Gliosis/pathology , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Receptor, Metabotropic Glutamate 5/biosynthesis , Seizures/chemically induced , Status Epilepticus/chemically induced , Status Epilepticus/pathology
17.
J Neuroinflammation ; 12: 217, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26597638

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative motor neuron disorder. Genetic studies have linked mutation of the gene SOD1 to ALS pathology as well as several other pathological processes including modulation of glutamatergic function and inflammatory processes. Since therapeutic approaches for ALS are focused on glutamatergic function, we investigated modulation of glutamate transport based on its receptor function as well as excitotoxicity-induced inflammatory response. METHODS: In vivo positron emission tomography (PET) imaging studies of metabotropic glutamate receptor subtype 5 (mGluR5) using [(18)F]FPEB ([(18)F]3-fluoro-5-(2-pyridylethynyl)benzonitrile) and inflammatory response using [(11)C]PBR28 (peripheral benzodiazepine receptor ligand 28) were done in an early and a late phase of neurodegeneration in four ALS mice expressing SOD1-G93A gene and four control base mice (C57/BL6). Accumulation of [(18)F]FPEB and [(11)C]PBR28 were quantitated in several brain areas and spinal cord to determine degeneration-induced modulation. The studies were completed with immunohistochemical analyses of mGluR5 and inflammatory response. RESULTS: These studies showed enhanced binding potential of [(18)F]FPEB in several brain areas including striatum, hippocampus, and frontal cortex. In the whole brain, the binding potential increased 49 ± 9 % from base mice to ALS-type mice and further enhanced 23 ± 4 % during disease progression. Also, in the spinal cord 6-22 %, enhanced accumulation of [(18)F]FPEB was observed during progression of the disease. The accumulation of [(11)C]PBR28 increased by 110 ± 33 % in the whole brain during progression of the disease indicating significant inflammatory process. [(11)C]PBR28 accumulation enhanced 89-264 % in the spinal cord and 204 % in the lungs. The end point immunohistochemical analyses verified the enhanced mGluR5 expression and inflammation. CONCLUSIONS: These results confirm the role of glutamate and inflammation in ALS-type pathology. These data also support the hypothesis that excessive glutamate may contribute to inflammation in the chronic neurodegenerative processes in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnostic imaging , Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Positron-Emission Tomography , Receptor, Metabotropic Glutamate 5/biosynthesis , Superoxide Dismutase/biosynthesis , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Progression , Gene Expression Regulation , Humans , Inflammation/diagnostic imaging , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Positron-Emission Tomography/methods , Receptor, Metabotropic Glutamate 5/genetics , Superoxide Dismutase/genetics
18.
Neurochem Res ; 40(11): 2200-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26318863

ABSTRACT

The study assessed involvement of Ca(2+) signaling mediated by the metabotropic glutamate receptors mGluR1/5 in brain tolerance induced by hypoxic preconditioning. Acute slices of rat piriform cortex were tested 1 day after exposure of adult rats to mild hypobaric hypoxia for 2 h at a pressure of 480 hPa once a day for three consecutive days. We detected 44.1 ± 11.6 % suppression of in vitro anoxia-induced increases of intracellular Ca(2+) levels and a fivefold increase in Ca(2+) transients evoked by selective mGluR1/5 agonist, DHPG. Western blot analysis of cortical homogenates demonstrated a 11 ± 4 % decrease in mGluR1 immunoreactivity (IR), and in the nuclei-enriched fraction a 12 ± 3 % increase in IR of phospholipase Cß1 (PLCß1), which is a major mediator of mGluR1/5 signaling. Immunocytochemical analysis of the cortex revealed increase in the mGluR1/5 and PLCß1 IR in perikarya, and a decrease in IR of the neuronal inositol trisphosphate receptors (IP3Rs). We suggest that enhanced expression of mGluR5 and PLCß1 and potentiation of Ca(2+) signaling may represent pro-survival upregulation of Ca(2+)-dependent genomic processes, while decrease in mGluR1 and IP3R IR may be attributed to a feedback mechanism preventing excessive intracellular Ca(2+) release.


Subject(s)
Air Pressure , Cerebral Cortex/metabolism , Hypoxia/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptors, Metabotropic Glutamate/biosynthesis , Signal Transduction/genetics , Animals , Calcium Signaling/genetics , Inositol 1,4,5-Trisphosphate Receptors/biosynthesis , Inositol 1,4,5-Trisphosphate Receptors/genetics , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Phospholipase C beta/biosynthesis , Phospholipase C beta/genetics , Piriform Cortex/metabolism , Rats , Rats, Wistar , Receptor, Metabotropic Glutamate 5/agonists , Receptor, Metabotropic Glutamate 5/genetics , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/genetics , Up-Regulation
19.
Cell Rep ; 11(12): 1876-84, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26095359

ABSTRACT

Mutations affecting the levels of microRNA miR-137 are associated with intellectual disability and schizophrenia. However, the pathophysiological role of miR-137 remains poorly understood. Here, we describe a highly conserved miR-137-binding site within the mRNA encoding the GluA1 subunit of AMPA-type glutamate receptors (AMPARs) and confirm that GluA1 is a direct target of miR-137. Postsynaptic downregulation of miR-137 at the CA3-CA1 hippocampal synapse selectively enhances AMPAR-mediated synaptic transmission and converts silent synapses to active synapses. Conversely, miR-137 overexpression selectively reduces AMPAR-mediated synaptic transmission and silences active synapses. In addition, we find that miR-137 is transiently upregulated in response to metabotropic glutamate receptor 5 (mGluR5), but not mGluR1 activation. Consequently, acute interference with miR-137 function impedes mGluR-LTD expression. Our findings suggest that miR-137 is a key factor in the control of synaptic efficacy and mGluR-dependent synaptic plasticity, supporting the notion that glutamatergic dysfunction contributes to the pathogenesis of miR-137-linked cognitive impairments.


Subject(s)
MicroRNAs/biosynthesis , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptors, AMPA/genetics , Schizophrenia/genetics , Animals , Binding Sites , Gene Expression Regulation , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , MicroRNAs/genetics , Mutation , Neuronal Plasticity/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Metabotropic Glutamate 5/genetics , Receptors, AMPA/biosynthesis , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics , Schizophrenia/pathology , Synapses/genetics , Synapses/metabolism
20.
Neurochem Res ; 40(5): 1074-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25894678

ABSTRACT

Substantial evidence reveals that prenatal stress is closely linked with abnormal behavior in offspring, but the mechanism remains unclear. In this study, our aim was to observe the alterations of behaviors, metabotropic glutamate receptor-1/5 (mGluR1/5) and brain-derived neurotrophic factor (BDNF) in various brain regions of prenatally stressed offspring rats. The forced swimming test (FST) and the open field test (OFT) were carried on 1-month-old offspring rats. The expression levels of mGluR1, mGluR5, and BDNF mRNA were measured in various brain regions of the offspring rats. Our results showed that the immobile time in the FST was significantly increased in the late prenatal stress (LPS) group compared with that in the control group, especially in the female rats. Similarly, in the OFT, the rats in both the mid prenatal stress (MPS) and LPS groups demonstrated anxiety-like behavior, especially the male rats in the LPS group. The expression of mGluR1 protein was increased in the hippocampus and prefrontal cortex of rats from the LPS group, as well as in the prefrontal cortex of rats from the MPS group. Meanwhile, the expression of mGluR5 protein was facilitated in the hippocampus and prefrontal cortex of rats in the LPS group. The expression of mGluR5 protein was increased in the striatum of the female rats in both MPS and LPS groups, and only in the male rats from the LPS group. In addition, the reduced BDNF mRNA level was detected in the hippocampus and prefrontal cortex in the LPS rats, and in the striatum of the female rats in LPS group. These results indicate that alterations of the mGluR1, mGluR5 and BDNF mRNA may contribute to the depression-like and anxiety-like behaviors of prenatally stressed offspring rats.


Subject(s)
Brain-Derived Neurotrophic Factor/biosynthesis , Prenatal Exposure Delayed Effects/metabolism , Receptor, Metabotropic Glutamate 5/biosynthesis , Receptors, Metabotropic Glutamate/biosynthesis , Stress, Psychological/metabolism , Animals , Anxiety/metabolism , Anxiety/psychology , Brain/metabolism , Depression/metabolism , Depression/psychology , Female , Male , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Sprague-Dawley , Stress, Psychological/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...