• Title/Summary/Keyword: GluR2

Search Result 88, Processing Time 0.019 seconds

Regulation of DREAM Expression by Group I mGluR

  • Lee, Jin-U;Kim, In-Sook;Oh, So-Ra;Ko, Suk-Jin;Lim, Mi-Kyung;Kim, Dong-Goo;Kim, Chul-Hoon
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.15 no.2
    • /
    • pp.95-100
    • /
    • 2011
  • DREAM (downstream regulatory element antagonistic modulator) is a calcium-binding protein that regulates dynorphin expression, promotes potassium channel surface expression, and enhances presenilin processing in an expression level-dependent manner. However, no molecular mechanism has yet explained how protein levels of DREAM are regulated. Here we identified group I mGluR (mGluR1/5) as a positive regulator of DREAM protein expression. Overexpression of mGluR1/5 increased the cellular level of DREAM. Up-regulation of DREAM resulted in increased DREAM protein in both the nucleus and cytoplasm, where the protein acts as a transcriptional repressor and a modulator of its interacting proteins, respectively. DHPG (3,5-dihydroxyphenylglycine), a group I mGluR agonist, also up-regulated DREAM expression in cortical neurons. These results suggest that group I mGluR is the first identified receptor that may regulate DREAM activity in neurons.

Roles of Metabotropic Glutamate Receptors 1 and 5 in Rat Medial Vestibular Nucleus Neurons

  • Lee, Hae-In;Lee, Sung-Hyo;Chun, Sang-Woo
    • International Journal of Oral Biology
    • /
    • v.36 no.2
    • /
    • pp.71-78
    • /
    • 2011
  • Using whole cell current- and voltage-clamp recording we investigated the characteristics and pharmacology of group I metabotropic glutamate receptor (mGluR)-mediated responses in rat medial vestibular nucleus (MVN) neurons. In current clamp conditions, activation of mGluR I by application of the group I mGluR agonist (R,S)-3,5-dihydroxyphenylglycine (DHPG) induced a direct excitation of MVN neurons that is characterized by depolarization and increased spontaneous firing frequency. To identify which of mGluR subtypes are responsible for the various actions of DHPG in MVN, we used two subtype-selective antagonists. (S)-(+)- alpha-amino-a-methylbenzeneacetic acid (LY367385) is a potent competitive antagonist that is selective for mGluR1, whereas 2-methyl-6-(phenylethynyl)-pyridine (MPEP) is a potent noncompetitive antagonist that is selective for mGluR5. In voltage clamp conditions, DHPG application increased the frequency of spontaneous and miniature inhibitory postsynaptic currents (IPSCs) but had no effect on amplitude distributions. Antagonism of the DHPG-induced increase of miniature IPSCs required the blockade of both mGluR1 and mGluR5. DHPG application induced an inward current, which can be enhanced under depolarized conditions. DHPG-induced current was blocked by LY367385, but not by MPEP. Both LY367385 and MPEP antagonized the DHPG-induced suppression of the calcium activated potassium current ($I_{AHP}$). These data suggest that mGluR1 and mGluR5 have similar roles in the regulation of the excitability of MVN neurons, and show a little distinct. Furthermore, mGluR I, via pre- and postsynaptic actions, have the potential to modulate the functions of the MVN.

Effect of Electroacupuncture on AMPA Receptor GluR2 Subunit in Complete Freund's Adjuvant-induced Inflammatory Pain Model

  • Kim, Chul-Yun;Choi, Hye-Young;Yang, Yeun-Jin;Choi, Byung-Tae
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.25 no.5
    • /
    • pp.881-886
    • /
    • 2011
  • AMPA receptor (AMPAR)s are heterotetrameric structures made up from 4 units (GluR1-4) and are thought to underlie perception of persistent inflammatory pain. Complete Freund's adjuvant (CFA)-evoked inflammation induces synaptic GluR2 internalization, which is initiated by GluR2 phosphorylation, in dorsal horn neurons during the maintenance of CFA-induced hypersensitivity. The present study investigated whether electroacupuncture (EA) stimulation has any effect on GluR2 trafficking by using immunoblot and immunohistochemistry. We examined that CFA-induced dorsal horn GluR2 internalization was attenuated by EA treatment. EA treatment could also decrease the level of pGluR2 regardless of whether CFA injection was administrated or not. In addition, previous studies suggest that microglial cells are increased without morphological change in CFA injected animal. In our study, increases in microglial cells in CFA group were observed, whereas EA with or without CFA-injected group showed similar aspects with normal group. In conclusion, our results indicate that EA might blunt CFA-evoked inflammation by coordinating mechanisms at the upstream step of neuron activation and GluR2 phosphorylation.

The Effect of Topiramate on Hippocampal Neuronal Death and Expression of Glutamate Receptor in Kainate-induced Status Epilepticus Model (Kainate 유발 간질중첩증 모델에서 topiramate가 해마 신경세포사와 glutamate 수용체 발현에 미치는 영향)

  • Park Min-Jeong;Ha Se-Un;Bae Hae-Rahn;Kim Sang-Ho
    • Journal of Life Science
    • /
    • v.15 no.3 s.70
    • /
    • pp.505-512
    • /
    • 2005
  • Excitotoxicity and epileptogenesis have often been associated with glutamate receptor activation. Accumulating evidences indicates that topiramate (TPM), an antiepileptic drug with multiple mechanisms of action has neuroprotective activity. We explored the neuroprotective effect of TPM on the status epilepticus (SE)-induced hippocampal neuronal death. After development of SE by kainite injection (15 mg/Kg), rats were treated with TPM (10mg/kg) for 1 week. The neuronal death was detected by Apop tag in situ detection kit, and the expression levels of glutamate receptors were semi-quantitatively analyzed by immunoblot. Kainate-induced SE caused a significant neuronal death and cell loss in CAI and CA3 regions of hippocampus at 1 week. However, treatment of TPM for 1 week after SE markedly reduced hippocampal neuronal death. The expression of N-methyl-D-aspartate (NMDA) receptor subunit 1, was increased by SE, but was not affected by 1 week treatment of TPM. The expressions of NMDA receptor subunit 2a and 2b were not changed by either SE or TPM. As for ${\alpha}-amino-3-hydroxy-5-methyl-4-isoxazole-propionate$ (AMPA) glutamate receptors (GluR), kainate-induced SE markedly up-regulated GluR1 expression but down-regulated GluR2 expression, leading to increased formation of $Ca^{2+}$ permeable GluR2- lacking AMPA receptors. TPM administration for 1 week attenuated SE-induced expression of both the up-regulation of GluR1 and down-regulation of GluR2, reversing the ratio of GluR1/GluR2 to the control value. In conclusion, TPM protects neuronal cell death against glutamate induced excitotoxicity in kainate-induced SE model, supporting the potential of TPM as a neuroprotective agent.

Retrograde Tracer Studies of Tecto-Reticulospinal Pathway and Dorsal Lateral Geniculate Nucleus on GluR1- and GluR4-Immunoreactive Neurons in the Hamster Superior Colliculus (Tecto-reticulospinal pathway (TRS)와 dorsal lateral geniculate nucleus (dLGN)에서 역행성이동추적물질 이용 햄스터 상구에서 GluR1-, GluR4- 면역반응 신경세포 연구)

  • Choi, Jae-Sik;Lee, Jea-Young;Jang, Yu-Jin;Lee, Eun-Shil;Jeon, Chang-Jin
    • Journal of Life Science
    • /
    • v.20 no.1
    • /
    • pp.1-8
    • /
    • 2010
  • We recently reported the distributions of AMPA ($\alpha$-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate) receptor subtypes glutamate receptors (GluR) 1 and GluR4 in the superior colliculi (SC) of hamsters with antibody immunocytochemistry and the effect of enucleation on these distributions. We also compared these labelings to those of calcium-binding proteins calbindin D28K, calretinin, parvalbumin, and GABA. In the present study, we investigated whether the GluR1- and GluR4-immunoreactive (IR) neurons are interneurons or projection neurons by injection of the retrograde tracer horseradish peroxidase (HRP) into one of each major ascending and descending pathways of the SC. HRP injections were made into a tecto-reticulospinal pathway (TRS) and dorsal lateral geniculate nucleus (dLGN). Animals were then allowed to recover and to survive for 48 hr before perfusion. Sections containing retrograde-labeled neurons were then treated for GluR-immunoreactivity. HRP injections proved that only a small population of the GluR1-IR cells project into TRS (1.4%) and dLGN (2.6%). However, a large subpopulation of GluR4-IR cells project into TRS (32.7%). The differential compositions of inter/projection neurons, along with our previous studies on the separate distribution of the GluR subunits, its differential co-localization with calcium-binding proteins and GABA, and differential reactions to enucleations, strongly imply the functional variety of the receptor subunits in visual behavior responses.

Mechanism of Glutamate-induced $[Ca^{2+}]i$ Increase in Substantia Gelatinosa Neurons of Juvenile Rats

  • Jung, Sung-Jun;Choi, Jeong-Sook;Kwak, Ji-Yeon;Kim, Jun;Kim, Jong-Whan;Kim, Sang-Jeong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.7 no.2
    • /
    • pp.53-57
    • /
    • 2003
  • The glutamate receptors (GluRs) are key receptors for modulatory synaptic events in the central nervous system. It has been reported that glutamate increases the intracellular $Ca^{2+}$ concentration ($[Ca^{2+}]_i$) and induces cytotoxicity. In the present study, we investigated whether the glutamate-induced $[Ca^{2+}]_i$ increase was associated with the activation of ionotropic (iGluR) and metabotropic GluRs (mGluR) in substantia gelatinosa neurons, using spinal cord slice of juvenile rats (10${\sim}21 day). $[Ca^{2+}]_i$ was measured using conventional imaging techniques, which was combined with whole-cell patch clamp recording by incorporating fura-2 in the patch pipette. At physiological concentration of extracellular $Ca^{2+}$, the inward current and $[Ca^{2+}]_i$ increase were induced by membrane depolarization and application of glutamate. Dose-response relationship with glutamate was observed in both $Ca^{2+}$ signal and inward current. The glutamate-induced $[Ca^{2+}]_i$ increase at holding potential of -70 mV was blocked by CNQX, an AMPA receptor blocker, but not by AP-5, a NMDA receptor blocker. The glutamate-induced $[Ca^{2+}]_i$ increase in $Ca^{2+}$ free condition was not affected by iGluR blockers. A selective mGluR (group I) agonist, RS-3,5-dihydroxyphenylglycine (DHPG), induced $[Ca^{2+}]_i$ increase at holding potential of -70 mV in SG neurons. These findings suggest that the glutamate-induced $[Ca^{2+}]_i$ increase is associated with AMPA-sensitive iGluR and group I mGluR in SG neurons of rats.

TRPC-Mediated Current Is Not Involved in Endocannabinoid-Induced Short-Term Depression in Cerebellum

  • Chang, Won-Seok;Kim, Jun;Kim, Sang-Jeong
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.16 no.2
    • /
    • pp.139-144
    • /
    • 2012
  • It has been reported that activation of metabotropic glutamate receptor 1 (mGluR1) can mediate endocannabinoid-induced short-term depression of synaptic transmission in cerebellar parallel fiber (PF)-Purkinje cell (PC) synapse. mGluR1 has signaling pathways involved in intracellular calcium increase which may contribute to endocannabinoid release. Two major mGluR1-evoked calcium signaling pathways are known: (1) slow-kinetic inward current carried by transient receptor potential canonical (TRPC) channel which is permeable to $Ca^{2+}$; (2) $IP_3$-induced calcium release from intracellular calcium store. However, it is unclear how much each calcium source contributes to endocannabinoid signaling. Here, we investigated whether calcium influx through mGluR1-evoked TRPC channel contributes to endocannabinoid signaling in cerebellar Purkinje cells. At first, we applied SKF96365 to inhibit TRPC, which blocked endocannabinoid-induced short-term depression completely. However, an alternative TRP channel inhibitor, BTP2 did not affect endocannabinoid-induced short-term depression although it blocked mGluR1-evoked TRPC currents. Endocannabinoid signaling occurred normally even though the TRPC current was mostly blocked by BTP2. Our data imply that TRPC current does not play an important role in endocannabinoid signaling. We also suggest precaution in applying SKF96365 to inhibit TRP channels and propose BTP2 as an alternative TRPC inhibitor.

Type II and III Taste Bud Cells Preferentially Expressed Kainate Glutamate Receptors in Rats

  • Lee, Sang-Bok;Lee, Cil-Han;Kim, Se-Nyun;Chung, Ki-Myung;Cho, Young-Kyung;Kim, Kyung-Nyun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.13 no.6
    • /
    • pp.455-460
    • /
    • 2009
  • Glutamate-induced cobalt uptake reveals that non-NMDA glutamate receptors (GluRs) are present in rat taste bud cells. Previous studies involving glutamate induced cobalt staining suggest this uptake mainly occurs via kainate type GluRs. It is not known which of the 4 types of taste bud cells express subunits of kainate GluR. Circumvallate and foliate papillae of Sprague-Dawley rats (45~60 days old) were used to search for the mRNAs of subunits of non-NMDA GluRs using RT-PCR with specific primers for GluR1-7, KA1 and KA2. We also performed RT-PCR for GluR5, KA1, $PLC\beta2$, and NCAM/SNAP 25 in isolated single cells from taste buds. Taste epithelium, including circumvallate or foliate papilla, express mRNAs of GluR5 and KA1. However, non-taste tongue epithelium expresses no subunits of non-NMDA GluRs. Isolated single cell RT-PCR reveals that the mRNAs of GluR5 and KA1 are preferentially expressed in Type II and Type III cells over Type I cells.

Antinociceptive Effects of Intrathecal Metabotropic Glutamate Receptor Compounds and Morphine in Rats

  • Choi, Jeong II;Lee, Hyung Kon;Chung, Sung Tae;Kim, Chang Mo;Bae, Hong Beom;Kim, Seok Jai;Yoon, Myung Ha;Chung, Sung Su;Jeong, Chang Young
    • The Korean Journal of Pain
    • /
    • v.18 no.1
    • /
    • pp.1-9
    • /
    • 2005
  • Background: Spinal metabotropic glutamate receptors (mGluRs) and opioid receptors are involved in the modulation of nociception. Although opioid receptors agonists are active for pain, the effects of the compounds for the mGluRs have not been definitely investigated at the spinal level. We examined the effects of the intrathecal mGluR compounds and morphine in the nociceptive test, and then we further clarified the role of the spinal mGluRs. In addition, the nature of the pharmacological interaction after the coadministration of mGluRs compounds with morphine was determined. Methods: Catheters were inserted into the intrathecal space of male SD rats. For the induction of pain, $50{\mu}l$ of 5% formalin solution or a thermal stimulus was applied to the hindpaw. An isobolographic analysis was used for the evaluation of the drug interaction. Results: Neither group I mGluR compounds nor group III mGluR compounds produced any antinociceptive effect in the formalin test. The group II mGluR agonist (APDC) had little effect on the formalin-induced nociception. The group II mGluR antagonist (LY 341495) caused a dose-dependent suppression of the phase 2 flinching response on the formalin test, but it did not reduce the phase 1 response of the formalin test nor did it increase the withdrawal latency of the thermal stimulus. Isobolographic analysis revealed a synergistic interaction after the intrathecal delivery of a LY 341495-morphine mixture. Conclusions: These results suggest that group II mGluRs are involved in the facilitated processing at the spinal level, and the combination of LY 341495 with morphine may be useful to manage the facilitated pain state.

Overexpression in Arabidopsis of a Plasma Membrane-targeting Glutamate Receptor from Small Radish Increases Glutamate-mediated Ca2+ Influx and Delays Fungal Infection

  • Kang, Seock;Kim, Ho Bang;Lee, Hyoungseok;Choi, Jin Young;Heu, Sunggi;Oh, Chang Jae;Kwon, Soon Il;An, Chung Sun
    • Molecules and Cells
    • /
    • v.21 no.3
    • /
    • pp.418-427
    • /
    • 2006
  • Ionotropic glutamate receptors (iGluRs) are ligand-gated nonselective cation channels that mediate fast excitatory neurotransmission. Although homologues of the iGluRs have been identified in higher plants, their roles are largely unknown. In this work we isolated a full-length cDNA clone (RsGluR) encoding a putative glutamate receptor from small radish. An RsGluR:mGFP fusion protein was localized to the plasma membrane. In Arabidopsis thaliana overexpressing the fulllength cDNA, glutamate treatment triggered greater $Ca^{2+}$ influx in the root cells of transgenic seedlings than in those of the wild type. Transgenic plants exhibited multiple morphological changes such as necrosis at their tips and the margins of developing leaves, dwarf stature with multiple secondary inflorescences, and retarded growth, as previously observed in transgenic Arabidopsis overexpressing AtGluR3.2 [Kim et al. (2001)]. Microarray analysis showed that jasmonic acid (JA)-responsive genes including defensins and JA-biosynthetic genes were up-regulated. RsGluR overexpression also inhibited growth of a necrotic fungal pathogen Botrytis cinerea possibly due to up-regulation of the defensins. Based on these results, we suggest that RsGluR is a glutamate-gated $Ca^{2+}$ channel located in the plasma membrane of higher plants and plays a direct or indirect role in defense against pathogen infection by triggering JA biosynthesis.