Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Brain Res ; 1809: 148349, 2023 06 15.
Article in English | MEDLINE | ID: mdl-36972837

ABSTRACT

Overactivity of the corticostriatal glutamatergic pathway is documented in Parkinson's disease (PD) and stimulation of presynaptic metabotropic glutamate (mGlu) receptors 4 on these striatal afferents inhibits glutamate release normalizing neuronal activity in the basal ganglia. Moreover, mGlu4 receptors are also expressed in glial cells and are able to modulate glial function making this receptor a potential target for neuroprotection. Hence, we investigated whether foliglurax, a positive allosteric modulator of mGlu4 receptors with high brain exposure after oral administration, has neuroprotective effects in MPTP mice to model early PD. Male mice were treated daily from day 1 to 10 with 1, 3 or 10 mg/kg of foliglurax and administered MPTP on the 5th day then euthanized on the 11th day. Dopamine neuron integrity was assessed with measures of striatal dopamine and its metabolites levels, striatal and nigral dopamine transporter (DAT) binding and inflammation with markers of striatal astrocytes (GFAP) and microglia (Iba1). MPTP lesion produced a decrease in dopamine, its metabolites and striatal DAT specific binding that was prevented by treatment with 3 mg/kg of foliglurax, whereas 1 and 10 mg/kg had no beneficial effect. MPTP mice had increased levels of GFAP; foliglurax treatment (3 mg/kg) prevented this increase. Iba1 levels were unchanged in MPTP mice compared to control mice. There was a negative correlation between dopamine content and GFAP levels. Our results show that positive allosteric modulation of mGlu4 receptors with foliglurax provided neuroprotective effects in the MPTP mouse model of PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Antiparkinson Agents , Dopaminergic Neurons , Neuroprotective Agents , Receptors, Metabotropic Glutamate , Animals , Male , Mice , Allosteric Regulation/drug effects , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/pharmacology , Basal Ganglia/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Astrocytes/metabolism , Microglia/metabolism , Neostriatum/drug effects , Neostriatum/metabolism
2.
Neuropharmacology ; 218: 109205, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35940348

ABSTRACT

By decreasing glutamate transmission, mGlu4 receptor positive allosteric modulators (mGlu4-PAM), in combination with levodopa (l-DOPA) may restore the synergy between glutamatergic and dopaminergic transmissions, thus maximizing the improvement of motor function in Parkinson's disease (PD). This study aimed to clarify the effects of foliglurax, a selective mGlu4-PAM, on the loss of bidirectional synaptic plasticity associated with l-DOPA-induced dyskinesia (LID). Behavioral assessments compared dyskinesia intensity in 6-hydroxydopamine (6-OHDA)-lesioned rats treated with l-DOPA or l-DOPA plus foliglurax. In slices from the same rats, patch-clamp techniques were used to examine electrophysiological differences in glutamatergic synapses, evaluating the EPSCs mediated by NMDA and AMPA receptors in striatal spiny projection neurons. High-frequency stimulation of corticostriatal fibers was used as long-term potentiation (LTP)-inducing protocol. Conversely, 15 min of low-frequency stimulation was applied to depotentiate LTP. The density of dendritic spines was measured in striatal slices in the same experimental conditions. Our results show that, in corticostriatal slices, foliglurax decreased spontaneous glutamatergic transmission in both sham-operated and 6-OHDA lesioned rats. When co-administered with l-DOPA in 6-OHDA-lesioned rats, foliglurax fully restored dendritic spine density in a dose-dependent manner. Moreover, this co-treatment rescued striatal bidirectional plasticity and attenuated the intensity of l-DOPA-induced dyskinesia. This is the first demonstration in an animal model of PD and dyskinesia that a mGlu4 PAM can restore striatal synaptic plasticity.


Subject(s)
Dyskinesia, Drug-Induced , Parkinson Disease , Parkinsonian Disorders , Animals , Antiparkinson Agents/adverse effects , Corpus Striatum , Disease Models, Animal , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Rats
3.
Mov Disord ; 33(10): 1619-1631, 2018 10.
Article in English | MEDLINE | ID: mdl-30216534

ABSTRACT

BACKGROUND: Levodopa remains the gold-standard treatment for PD. However, it becomes less effective as the disease progresses and produces debilitating side effects, such as motor fluctuations and l-dopa-induced dyskinesia. Modulation of metabotropic glutamate receptor 4 represents a promising antiparkinsonian approach in combination with l-dopa, but it has not been demonstrated in primates. OBJECTIVE: We studied whether a novel positive allosteric modulator of the metabotropic glutamate receptor 4, PXT002331 (foliglurax), could reduce parkinsonism in primate models. METHODS: We assessed the therapeutic potential of PXT002331 in three models of MPTP-induced parkinsonism in macaques. These models represent three different stages of disease evolution: early stage and advanced stage with and without l-dopa-induced dyskinesia. RESULTS: As an adjunct to l-dopa, PXT002331 induced a robust and dose-dependent reversal of parkinsonian motor symptoms in macaques, including bradykinesia, tremor, posture, and mobility. Moreover, PXT002331 strongly decreased dyskinesia severity, thus having therapeutic efficacy on both parkinsonian motor impairment and l-dopa-induced dyskinesia. PXT002331 brain penetration was also assessed using PET imaging in macaques, and pharmacodynamic analyses support target engagement in the therapeutic effects of PXT002331. CONCLUSIONS: This work provides a demonstration that a positive allosteric modulator of metabotropic glutamate receptor 4 can alleviate the motor symptoms of PD and the motor complications induced by l-dopa in primates. PXT002331 is the first compound of its class to enter phase IIa clinical trials. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Parkinsonian Disorders/drug therapy , Receptors, Metabotropic Glutamate/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Antiparkinson Agents/chemistry , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/etiology , Excitatory Amino Acid Antagonists/chemistry , Levodopa/adverse effects , Macaca fascicularis , Parkinsonian Disorders/complications , Positron-Emission Tomography , Receptors, Metabotropic Glutamate/chemistry , Time Factors
4.
J Med Chem ; 60(20): 8515-8537, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28902994

ABSTRACT

The metabotropic glutamate receptor 4 (mGluR4) is an emerging target for the treatment of Parkinson's disease (PD). However, since the discovery of its therapeutic potential, no ligand has been successfully developed enough to be tested in the clinic. In the present paper, we report for the first time the medicinal chemistry efforts conducted around the pharmacological tool (-)-PHCCC. This work led to the identification of compound 40, a potent and selective mGluR4 positive allosteric modulator (PAM) with good water solubility and demonstrating consistent activity across validated preclinical rodent models of PD motor symptoms after intraperitoneal administration: haloperidol-induced catalepsy in mouse and the rat 6-hydroxydopamine (6-OHDA) lesion model. Moreover, we also describe the identification of compound 60 a close analogue of compound 40 with improved pharmacokinetic profile after oral administration. On the basis of its favorable and unique preclinical profile, compound 60 (PXT002331, now foliglurax) was nominated as a candidate for clinical development.


Subject(s)
Antiparkinson Agents/pharmacology , Brain/drug effects , Drug Discovery , Receptors, Metabotropic Glutamate/drug effects , Allosteric Regulation , Animals , Antiparkinson Agents/pharmacokinetics , Chromatography, Liquid , HEK293 Cells , Humans , Mass Spectrometry , Mice , Proton Magnetic Resonance Spectroscopy , Rats , Structure-Activity Relationship
5.
Dev Neurobiol ; 72(12): 1559-76, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22539416

ABSTRACT

The guidepost neurons for the lateral olfactory tract, which are called lot cells, are the earliest-generated neurons in the neocortex. They migrate tangentially and ventrally further down this tract, and provide scaffolding for the olfactory bulb axons projecting into this pathway. The molecular profiles of the lot cells are largely uncharacterized. We found that lot cells specifically express metabotropic glutamate receptor subtype-1 at a very early stage of development. This receptor is functionally competent and responds to a metabotropic glutamate receptor agonist with a transient increase in the intracellular calcium ion concentration. When the glutamatergic olfactory bulb axons were electrically stimulated, lot cells responded to the stimulation with a calcium increase mainly via ionotropic glutamate receptors, suggesting potential neurotransmission between the axons and lot cells during early development. Together with the finding that lot cells themselves are glutamatergic excitatory neurons, our results provide another notable example of precocious interactions between the projecting axons and their intermediate targets.


Subject(s)
Neurogenesis/physiology , Neurons/metabolism , Olfactory Bulb/metabolism , Olfactory Pathways/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Cell Communication/physiology , Cells, Cultured , Immunohistochemistry , Mice , Mice, Inbred ICR , Olfactory Bulb/growth & development , Olfactory Pathways/growth & development , Synaptic Transmission/physiology , Transfection
6.
J Neurosci ; 28(44): 11263-8, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18971468

ABSTRACT

ATP is a known mediator of inflammatory and neuropathic pain. However, the mechanisms by which specific purinergic receptors contribute to chronic pain states are still poorly characterized. Here, we demonstrate that in response to peripheral nerve injury, P2X(4) receptors (P2X(4)R) are expressed de novo by activated microglia in the spinal cord. Using in vitro and in vivo models, we provide direct evidence that P2X(4)R stimulation leads to the release of BDNF from activated microglia and, most likely phosphorylation of the NR1 subunit of NMDA receptors in dorsal horn neurons of the spinal cord. Consistent with these findings, P2X4-deficient mice lack mechanical hyperalgesia induced by peripheral nerve injury and display impaired BDNF signaling in the spinal cord. Furthermore, ATP stimulation is unable to stimulate BDNF release from P2X(4)-deficient mice microglia in primary cultures. These results indicate that P2X(4)R contribute to chronic pain through a central inflammatory pathway. P2X(4)R might thus represent a potential therapeutic target to limit microglia-mediated inflammatory responses associated with brain injury and neurodegenerative disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Microglia/metabolism , Neuralgia/metabolism , Peripheral Nerve Injuries , Receptors, Purinergic P2/physiology , Spinal Cord/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , COS Cells , Cells, Cultured , Chlorocebus aethiops , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/genetics , Pain/genetics , Pain/metabolism , Pain Measurement/methods , Peripheral Nerves/metabolism , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X4 , Up-Regulation/physiology
7.
Neuropharmacology ; 55(4): 454-8, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18606173

ABSTRACT

The requirements for the synaptic activation of metabotropic glutamate (mGlu) receptors and for the induction of metaplasticity in the hippocampus are not known. In the present study, we have investigated the synaptic activation of mGlu5 receptors and the setting of the molecular switch, a form of metaplasticity, at CA1 synapses in the mouse hippocampus. We find that as few as eight stimuli (delivered at 100Hz) are sufficient to set the molecular switch, since a subsequent tetanus delivered to the same input is able to induce long-term potentiation (LTP) in the presence of the mGlu receptor antagonist MCPG ((S)-alpha-methyl-4-carboxyphenylglycine). In addition, we find that the molecular switch can be activated over a wide frequency range. When 10 shocks were delivered the threshold frequency was 4Hz. The ability of 10 shocks (delivered at 100Hz) to set the molecular switch was lost in the mGlu5 knockout. These data show that mGlu5 receptors can be activated synaptically and metaplasticity can be induced by relatively few stimuli. Indeed, metaplasticity was induced by stimuli that were subthreshold for the induction of LTP per se. Thus, metaplasticity has a lower threshold than the synaptic plasticity that it regulates.


Subject(s)
Electric Stimulation , Neuronal Plasticity/physiology , Neuronal Plasticity/radiation effects , Neurons/physiology , Receptors, Metabotropic Glutamate/physiology , Synapses/physiology , Animals , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/cytology , In Vitro Techniques , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency , Synapses/drug effects , Synapses/radiation effects , Time Factors
8.
Neuropharmacology ; 49 Suppl 1: 13-25, 2005.
Article in English | MEDLINE | ID: mdl-16024054

ABSTRACT

The role of metabotropic glutamate (mGlu) receptors in long-term potentiation (LTP) in the hippocampus is controversial. In the present study, we have used mice in which the mGlu1, mGlu5 or mGlu7 receptor has been deleted, by homologous recombination, to study the role of these receptor subtypes in LTP at CA1 synapses. We investigated the effects of the knockouts on both LTP and the molecular switch, a form of metaplasticity that renders LTP insensitive to the actions of the mGlu receptor antagonist MCPG ((S)-alpha-methyl-4-carboxyphenylglycine). We find that LTP is readily induced in the three knockouts and in an mGlu1 and mGlu5 double knockout. In addition, the molecular switch operates normally in either the mGlu1 or mGlu7 knockout. In contrast, the molecular switch is completely non-functional in the mGlu5 knockout, such that MCPG invariably blocks the induction of additional LTP in an input where LTP has already been induced. The effect of the mGlu5 receptor knockout was replicated in wildtype mouse slices perfused with the specific mGlu5 receptor antagonist MPEP (2-methyl-6-(phenylethynyl)-pyridine). In addition, the mGlu5 selective agonist CHPG ((RS)-2-chloro-5-hydroxyphenylglycine) sets the molecular switch. These data demonstrate that the operation of the molecular switch requires activation of mGlu5 receptors.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials , In Vitro Techniques , Long-Term Potentiation/drug effects , Long-Term Potentiation/radiation effects , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/classification , Receptors, Metabotropic Glutamate/genetics , Time Factors
9.
J Neurophysiol ; 92(5): 3085-96, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15212418

ABSTRACT

In the rodent main olfactory bulb (MOB), mitral cells (MCs) express high levels of the group I metabotropic glutamate receptor (mGluR) subtype, mGluR1. The significance of this receptor in modulating MC excitability is unknown. We investigated the physiological role of mGluR1 in regulating MC activity in rat and mouse MOB slices. The selective group I agonist (RS)-3,5-dihydroxyphenylglycine (DHPG), but not group II or III agonists, induced potent, dose-dependent, and reversible depolarization and increased firing of MCs. These effects persisted in the presence of blockers of fast synaptic transmission, indicating that they are due to direct activation of mGluRs on MCs. Voltage-clamp recordings showed that DHPG elicited a voltage-dependent inward current consisting of multiple components sensitive to potassium and calcium channel blockade and intracellular calcium chelation. MC excitatory responses to DHPG were absent in mGluR1 knockout mice but persisted in mGluR5 knockout mice. Broad-spectrum LY341495, MCPG, as well as preferential mGluR1 LY367385 antagonists blocked the excitatory effects of DHPG and also potently modulated MC spontaneous and olfactory nerve-evoked excitability. mGluR antagonists altered spontaneous membrane potential bistability, increasing the duration of the up and down states. mGluR antagonists also substantially attenuated MC responses to sensory input, decreasing the probability and increasing the latency of olfactory nerve-evoked spikes. These findings suggest that endogenous glutamate tonically modulates MC excitability and responsiveness to olfactory nerve input, and hence the operation of the MOB circuitry, via activation of mGluR1.


Subject(s)
Methoxyhydroxyphenylglycol/analogs & derivatives , Neurons/physiology , Olfactory Bulb/physiology , Olfactory Nerve/physiology , Receptors, Metabotropic Glutamate/physiology , Animals , Base Sequence , DNA Primers , Electric Stimulation , Electrophysiology , Female , In Vitro Techniques , Male , Membrane Potentials/physiology , Methoxyhydroxyphenylglycol/pharmacology , Mice , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/drug effects , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
10.
J Physiol ; 555(Pt 1): 125-35, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14673190

ABSTRACT

Hippocampal CA1 inhibitory interneurones control the excitability and synchronization of pyramidal cells, and participate in hippocampal synaptic plasticity. Pairing theta-burst stimulation (TBS) with postsynaptic depolarization, we induced long-term potentiation (LTP) of putative single-fibre excitatory postsynaptic currents (EPSCs) in stratum oriens/alveus (O/A) interneurones of mouse hippocampal slices. LTP induction was absent in metabotropic glutamate receptor 1 (mGluR1) knockout mice, was correlated with the postsynaptic presence of mGluR1a, and required a postsynaptic Ca2+ rise. Changes in paired-pulse facilitation and coefficient of variation indicated that LTP expression involved presynaptic mechanisms. LTP was synapse specific, occurring selectively at synapses modulated by presynaptic group II, but not group III, mGluRs. Furthermore, the TBS protocol applied in O/A induced a long-term increase of polysynaptic inhibitory responses in CA1 pyramidal cells, that was absent in mGluR1 knockout mice. These results uncover the mechanisms of a novel form of interneurone synaptic plasticity that can adaptively regulate inhibition of hippocampal pyramidal cells.


Subject(s)
Egtazic Acid/analogs & derivatives , Hippocampus/physiology , Interneurons/physiology , Long-Term Potentiation/physiology , Receptors, Metabotropic Glutamate/physiology , Synapses/physiology , Animals , Egtazic Acid/pharmacology , Hippocampus/drug effects , Interneurons/drug effects , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Receptors, Metabotropic Glutamate/deficiency , Synapses/drug effects
11.
J Neurosci ; 23(13): 5634-44, 2003 Jul 02.
Article in English | MEDLINE | ID: mdl-12843266

ABSTRACT

The aim of this study was to describe the induction and expression mechanisms of a persistent bursting activity in a horizontal slice preparation of the rat limbic system that includes the ventral part of the hippocampus and the entorhinal cortex. Disinhibition of this preparation by bicuculline led to interictal-like bursts in the CA3 region that triggered synchronous activity in the entorhinal cortex. Washout of bicuculline after a 1 hr application resulted in a maintained production of hippocampal bursts that continued to spread to the entorhinal cortex. Separation of CA3 from the entorhinal cortex caused the activity in the latter to become asynchronous with CA3 activity in the presence of bicuculline and disappear after washout; however, in CA3, neither the induction of bursting nor its persistence were affected. Associated with the CA3 persistent bursting, a strengthening of recurrent collateral excitatory input to CA3 pyramidal cells and a decreased input to CA3 interneurons was found. Both the induction of the persistent bursting and the changes in synaptic strength were prevented by antagonists of metabotropic glutamate 5 (mGlu5) or NMDA receptors or protein synthesis inhibitors and did not occur in slices from mGlu5 receptor knock-out mice. The above findings suggest potential synaptic mechanisms by which the hippocampus switches to a persistent interictal bursting mode that may support a spread of interictal-like bursting to surrounding temporal lobe regions.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Entorhinal Cortex/cytology , Entorhinal Cortex/drug effects , Entorhinal Cortex/physiology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , GABA Antagonists/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Interneurons/drug effects , Interneurons/physiology , Long-Term Potentiation/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Protein Synthesis Inhibitors/pharmacology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
12.
J Physiol ; 541(Pt 1): 113-21, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12015424

ABSTRACT

The group I metabotropic glutamate receptor agonist DHPG has been shown to produce two major effects on CA3 pyramidal cells at rest: a reduction in the background conductance and an activation of a voltage-gated inward current (I(mGluR(V))). Both effects contribute to depolarising CA3 pyramidal cells and the latter has been implicated in eliciting prolonged epileptiform population bursts. We observed that DHPG-induced depolarisation was smaller in CA1 pyramidal cells than in CA3 cells. Voltage clamp studies revealed that while DHPG elicited I(mGluR(V)) in CA3 pyramidal cells, such a response was absent in CA1 pyramidal cells. Both mGluR1 and mGluR5 have been localised in CA3 pyramidal cells, whereas only mGluR5 has been detected in CA1 pyramidal cells. Using mGluR1 knockout mice, we evaluated whether the absence of an I(mGluR(V)) response can be correlated with the absence of mGluR1. In these experiments, DHPG failed to elicit I(mGluR(V)) in CA3 pyramidal cells. This suggests that the smaller depolarising effects of DHPG on wild-type CA1 pyramidal cells is caused, at least in part, by the absence of I(mGluR(V)) in these cells and that the difference in the responses of CA1 and CA3 cells may be attributable to the lack of mGluR1 in CA1 pyramidal cells.


Subject(s)
Hippocampus/physiology , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Electrophysiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , In Vitro Techniques , Membrane Potentials/physiology , Mice , Mice, Knockout , Neural Conduction/physiology , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/genetics , Resorcinols/pharmacology , Tetrodotoxin/pharmacology
13.
Synapse ; 44(2): 86-93, 2002 May.
Article in English | MEDLINE | ID: mdl-11891880

ABSTRACT

Metabotropic glutamate receptor 1 (mGluR1) is highly expressed in striatonigral projection neurons of rat striatum. To define the role of mGluR1 in the regulation of striatal gene expression, the responsiveness of the three neuropeptide gene expression to a single injection of the dopamine D(1) agonist SKF-82958 was compared between mGluR1 mutant and wild-type control mice. We found that acute injection of SKF-82958 increased preprodynorphin (PPD), substance P (SP), and preproenkephalin (PPE) mRNAs in the dorsal and ventral striatum of mutant and wild-type mice in a dose-dependent manner (0.125, 0.5, and 2 mg/kg, i.p.) as revealed by quantitative in situ hybridization. However, the induction of PPD mRNA in both the dorsal and ventral striatum of mGluR1 minus sign/minus sign mice was significantly less than that of wild-type +/+ mice in response to the two higher doses of SKF-82958. In contrast to PPD, SP and PPE in the dorsal and ventral striatum of mGluR1 mutant mice were elevated to a similar level as that of wild-type mice. There were no differences in basal levels and distribution patterns of all three mRNAs between the two genotypes of mice treated with saline. These results indicate that mGluR1 selectively participates in striatonigral PPD induction in response to D(1) receptor stimulation.


Subject(s)
Benzazepines/administration & dosage , Dynorphins/genetics , Enkephalins/genetics , Nucleus Accumbens/drug effects , Protein Precursors/genetics , Putamen/drug effects , RNA, Messenger/analysis , Receptors, Metabotropic Glutamate/drug effects , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Injections, Intraperitoneal , Male , Mice , Mice, Knockout , Nucleus Accumbens/metabolism , Nucleus Accumbens/ultrastructure , Putamen/metabolism , Putamen/ultrastructure , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/metabolism , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...