1
|
Gaidin SG, Kosenkov AM. mRNA editing of kainate receptor subunits: what do we know so far? Rev Neurosci 2022; 33:641-655. [DOI: 10.1515/revneuro-2021-0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/18/2022] [Indexed: 11/15/2022]
Abstract
Abstract
Kainate receptors (KARs) are considered one of the key modulators of synaptic activity in the mammalian central nervous system. These receptors were discovered more than 30 years ago, but their role in brain functioning remains unclear due to some peculiarities. One such feature of these receptors is the editing of pre-mRNAs encoding GluK1 and GluK2 subunits. Despite the long history of studying this phenomenon, numerous questions remain unanswered. This review summarizes the current data about the mechanism and role of pre-mRNA editing of KAR subunits in the mammalian brain and proposes a perspective of future investigations.
Collapse
Affiliation(s)
- Sergei G. Gaidin
- Institute of Cell Biophysics of the Russian Academy of Sciences , 142290 , Pushchino , Russia
| | - Artem M. Kosenkov
- Institute of Cell Biophysics of the Russian Academy of Sciences , 142290 , Pushchino , Russia
| |
Collapse
|
2
|
de Melo IS, Dos Santos YMO, Pacheco ALD, Costa MA, de Oliveira Silva V, Freitas-Santos J, de Melo Bastos Cavalcante C, Silva-Filho RC, Leite ACR, Gitaí DGL, Duzzioni M, Sabino-Silva R, Borbely AU, de Castro OW. Role of Modulation of Hippocampal Glucose Following Pilocarpine-Induced Status Epilepticus. Mol Neurobiol 2021; 58:1217-1236. [PMID: 33123979 DOI: 10.1007/s12035-020-02173-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/14/2020] [Indexed: 02/08/2023]
Abstract
Status epilepticus (SE) is defined as continuous and self-sustaining seizures, which trigger hippocampal neurodegeneration, mitochondrial dysfunction, oxidative stress, and energy failure. During SE, the neurons become overexcited, increasing energy consumption. Glucose uptake is increased via the sodium glucose cotransporter 1 (SGLT1) in the hippocampus under epileptic conditions. In addition, modulation of glucose can prevent neuronal damage caused by SE. Here, we evaluated the effect of increased glucose availability in behavior of limbic seizures, memory dysfunction, neurodegeneration process, neuronal activity, and SGLT1 expression. Vehicle (VEH, saline 0.9%, 1 μL) or glucose (GLU; 1, 2 or 3 mM, 1 μL) were administered into hippocampus of male Wistar rats (Rattus norvegicus) before or after pilocarpine to induce SE. Behavioral analysis of seizures was performed for 90 min during SE. The memory and learning processes were analyzed by the inhibitory avoidance test. After 24 h of SE, neurodegeneration process, neuronal activity, and SGLT1 expression were evaluated in hippocampal and extrahippocampal regions. Modulation of hippocampal glucose did not protect memory dysfunction followed by SE. Our results showed that the administration of glucose after pilocarpine reduced the severity of seizures, as well as the number of limbic seizures. Similarly, glucose after SE reduced cell death and neuronal activity in hippocampus, subiculum, thalamus, amygdala, and cortical areas. Finally, glucose infusion elevated the SGLT1 expression in hippocampus. Taken together our data suggest that possibly the administration of intrahippocampal glucose protects brain in the earlier stage of epileptogenic processes via an important support of SGLT1.
Collapse
Affiliation(s)
- Igor Santana de Melo
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | | | - Amanda Larissa Dias Pacheco
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Maisa Araújo Costa
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Vanessa de Oliveira Silva
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Jucilene Freitas-Santos
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | | | - Reginaldo Correia Silva-Filho
- Bioenergetics Laboratory, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Ana Catarina Rezende Leite
- Bioenergetics Laboratory, Institute of Chemistry and Biotechnology, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Daniel Góes Leite Gitaí
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Marcelo Duzzioni
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia (UFU), Uberlândia, MG, Brazil
| | - Alexandre Urban Borbely
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil
| | - Olagide Wagner de Castro
- Department of Physiology, Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceió, AL, Brazil.
| |
Collapse
|
3
|
Activation of Phosphatidylinositol-Linked Dopamine Receptors Induces a Facilitation of Glutamate-Mediated Synaptic Transmission in the Lateral Entorhinal Cortex. PLoS One 2015; 10:e0131948. [PMID: 26133167 PMCID: PMC4489908 DOI: 10.1371/journal.pone.0131948] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/08/2015] [Indexed: 11/19/2022] Open
Abstract
The lateral entorhinal cortex receives strong inputs from midbrain dopamine neurons that can modulate its sensory and mnemonic function. We have previously demonstrated that 1 µM dopamine facilitates synaptic transmission in layer II entorhinal cortex cells via activation of D1-like receptors, increased cAMP-PKA activity, and a resulting enhancement of AMPA-receptor mediated currents. The present study assessed the contribution of phosphatidylinositol (PI)-linked D1 receptors to the dopaminergic facilitation of transmission in layer II of the rat entorhinal cortex, and the involvement of phospholipase C activity and release of calcium from internal stores. Whole-cell patch-clamp recordings of glutamate-mediated evoked excitatory postsynaptic currents were obtained from pyramidal and fan cells. Activation of D1-like receptors using SKF38393, SKF83959, or 1 µM dopamine induced a reversible facilitation of EPSCs which was abolished by loading cells with either the phospholipase C inhibitor U-73122 or the Ca2+ chelator BAPTA. Neither the L-type voltage-gated Ca2+ channel blocker nifedipine, nor the L/N-type channel blocker cilnidipine, blocked the facilitation of synaptic currents. However, the facilitation was blocked by blocking Ca2+ release from internal stores via inositol 1,4,5-trisphosphate (InsP3) receptors or ryanodine receptors. Follow-up studies demonstrated that inhibiting CaMKII activity with KN-93 failed to block the facilitation, but that application of the protein kinase C inhibitor PKC(19-36) completely blocked the dopamine-induced facilitation. Overall, in addition to our previous report indicating a role for the cAMP-PKA pathway in dopamine-induced facilitation of synaptic transmission, we demonstrate here that the dopaminergic facilitation of synaptic responses in layer II entorhinal neurons also relies on a signaling cascade dependent on PI-linked D1 receptors, PLC, release of Ca2+ from internal stores, and PKC activation which is likely dependent upon both DAG and enhanced intracellular Ca2+. These signaling pathways may collaborate to enhance sensory and mnemonic function in the entorhinal cortex during tonic release of dopamine.
Collapse
|
4
|
Zheng C, Deng QQ, Liu LL, Wang MY, Zhang G, Sheng WL, Weng SJ, Yang XL, Zhong YM. Orexin-A differentially modulates AMPA-preferring responses of ganglion cells and amacrine cells in rat retina. Neuropharmacology 2015; 93:80-93. [PMID: 25656479 DOI: 10.1016/j.neuropharm.2015.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 12/22/2014] [Accepted: 01/20/2015] [Indexed: 01/18/2023]
Abstract
By activating their receptors (OX1R and OX2R) orexin-A/B regulate wake/sleeping states, feeding behaviors, but the function of these peptides in the retina remains unknown. Using patch-clamp recordings and calcium imaging in rat isolated retinal cells, we demonstrated that orexin-A suppressed α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA)-preferring receptor-mediated currents (AMPA-preferring currents) in ganglion cells (GCs) through OX1R, but potentiated those in amacrine cells (ACs) through OX2R. Consistently, in rat retinal slices orexin-A suppressed light-evoked AMPA-preferring receptor-mediated excitatory postsynaptic currents in GCs, but potentiated those in ACs. Intracellular dialysis of GDP-β-S or preincubation with the Gi/o inhibitor pertussis toxin (PTX) abolished both the effects. Either cAMP/the protein kinase A (PKA) inhibitor Rp-cAMP or cGMP/the PKG blocker KT5823 failed to alter the orexin-A effects. Whilst both of them involved activation of protein kinase C (PKC), the effects on GCs and ACs were respectively eliminated by the phosphatidylinositol (PI)-phospholipase C (PLC) inhibitor and phosphatidylcholine (PC)-PLC inhibitor. Moreover, in GCs orexin-A increased [Ca(2+)]i and the orexin-A effect was blocked by intracellular Ca(2+)-free solution and by inositol 1,4,5-trisphosphate (IP3) receptor antagonists. In contrast, orexin-A did not change [Ca(2+)]i in ACs and the orexin-A effect remained in intracellular or extracellular Ca(2+)-free solution. We conclude that a distinct Gi/o/PI-PLC/IP3/Ca(2+)-dependent PKC signaling pathway, following the activation of OX1R, is likely responsible for the orexin-A effect on GCs, whereas a Gi/o/PC-PLC/Ca(2+)-independent PKC signaling pathway, following the activation of OX2R, mediates the orexin-A effect on ACs. These two actions of orexin-A, while working in concert, provide a characteristic way for modulating information processing in the inner retina.
Collapse
Affiliation(s)
- Chao Zheng
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China; Cell Electrophysiology Laboratory, Wannan Medical College, 22 West Wenchang Road, Wuhu, Anhui 241002, China
| | - Qin-Qin Deng
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Lei-Lei Liu
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Meng-Ya Wang
- Cell Electrophysiology Laboratory, Wannan Medical College, 22 West Wenchang Road, Wuhu, Anhui 241002, China
| | - Gong Zhang
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Wen-Long Sheng
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Shi-Jun Weng
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Xiong-Li Yang
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| | - Yong-Mei Zhong
- Institute of Neurobiology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| |
Collapse
|
5
|
Adzovic L, Domenici L. Insulin induces phosphorylation of the AMPA receptor subunit GluR1, reversed by ZIP, and over-expression of Protein Kinase M zeta, reversed by amyloid beta. J Neurochem 2014; 131:582-7. [PMID: 25230927 DOI: 10.1111/jnc.12947] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 08/22/2014] [Accepted: 09/10/2014] [Indexed: 11/30/2022]
Abstract
Insulin receptor (IR) in the brain plays a role in synaptic plasticity and cognitive functions. Phosphorylation of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors GluR1 subunit at Serine 831 is regulated by calcium-calmodulin-dependent protein kinase II and protein kinase C that underlie long-term potentiation and learning/memory. Recent studies have shown that the novel Protein Kinase M zeta (PKMζ) underlies synaptic plasticity and may regulate AMPAr. In this study, we show that insulin induces phosphorylation of Serine 831 GluR1 subunit of AMPAr and induces over-expression of PKMζ; pre-treatment with either the IR inhibitor 3-Bromo-5-t-butyl-4-hydroxy-benzylidenemalonitrile (AG1024) or PKMζ inhibitor protein kinase C zeta pseudo-substrate inhibitor returned the phosphorylation value of GluR1 to control level. Amyloid beta (Aβ) peptide in the form of oligomers interferes with IR signaling. Pre-treating neuronal cultures with Aβ following incubation with insulin, we found a reduction of insulin-dependent PKMζ over-expression and MAPK/Erk (1/2) phosphorylation, i.e., signaling pathways involved in synaptic plasticity and learning/memory. These results indicate a new intracellular insulin signaling pathway, and, additionally, that insulin resistance in Alzheimer's disease is a response to the production and accumulation of Aβ.
Collapse
Affiliation(s)
- Linda Adzovic
- Neuroscience Institute, CNR, Pisa, Italy; Department of Psychology, The Ohio State University, Columbus, Ohio, USA
| | | |
Collapse
|
6
|
Zhu QJ, Kong FS, Xu H, Wang Y, Du CP, Sun CC, Liu Y, Li T, Hou XY. Tyrosine phosphorylation of GluK2 up-regulates kainate receptor-mediated responses and downstream signaling after brain ischemia. Proc Natl Acad Sci U S A 2014; 111:13990-5. [PMID: 25201974 PMCID: PMC4183319 DOI: 10.1073/pnas.1403493111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although kainate receptors play important roles in ischemic stroke, the molecular mechanisms underlying postischemic regulation of kainate receptors remain unclear. In this study we demonstrate that Src family kinases contribute to the potentiation of kainate receptor function. Brain ischemia and reperfusion induce rapid and sustained phosphorylation of the kainate receptor subunit GluK2 by Src in the rat hippocampus, implicating a critical role for Src-mediated GluK2 phosphorylation in ischemic brain injury. The NMDA and kainate receptors are involved in the tyrosine phosphorylation of GluK2. GluK2 binds to Src, and the tyrosine residue at position 590 (Y590) on GluK2 is a major site of phosphorylation by Src kinases. GluK2 phosphorylation at Y590 is responsible for increases in whole-cell currents and calcium influx in response to transient kainate stimulation. In addition, GluK2 phosphorylation at Y590 facilitates the endocytosis of GluK2 subunits, and the activation of JNK3 and its substrate c-Jun after long-term kainate treatment. Thus, Src phosphorylation of GluK2 plays an important role in the opening of kainate receptor channels and downstream proapoptosis signaling after brain ischemia. The present study reveals an additional mechanism for the regulation of GluK2-containing kainate receptors by Src family kinases, which may be of pathological significance in ischemic stroke.
Collapse
Affiliation(s)
- Qiu-Ju Zhu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Fan-Shu Kong
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Hao Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Yi Wang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Cai-Ping Du
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Chang-Cheng Sun
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Yong Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Ting Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| | - Xiao-Yu Hou
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical College, Jiangsu 221004, China
| |
Collapse
|
7
|
Schauwecker PE. The effects of glycemic control on seizures and seizure-induced excitotoxic cell death. BMC Neurosci 2012; 13:94. [PMID: 22867059 PMCID: PMC3465215 DOI: 10.1186/1471-2202-13-94] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Accepted: 07/24/2012] [Indexed: 12/20/2022] Open
Abstract
Background Epilepsy is the most common neurological disorder after stroke, affecting more than 50 million persons worldwide. Metabolic disturbances are often associated with epileptic seizures, but the pathogenesis of this relationship is poorly understood. It is known that seizures result in altered glucose metabolism, the reduction of intracellular energy metabolites such as ATP, ADP and phosphocreatine and the accumulation of metabolic intermediates, such as lactate and adenosine. In particular, it has been suggested that the duration and extent of glucose dysregulation may be a predictor of the pathological outcome of status. However, little is known about neither the effects of glycemic control on brain metabolism nor the effects of managing systemic glucose concentrations in epilepsy. Results In this study, we examined glycemic modulation of kainate-induced seizure sensitivity and its neuropathological consequences. To investigate the relationship between glycemic modulation, seizure susceptibility and its neuropathological consequences, C57BL/6 mice (excitotoxin cell death resistant) were subjected to hypoglycemia or hyperglycemia, followed by systemic administration of kainic acid to induce seizures. Glycemic modulation resulted in minimal consequences with regard to seizure severity but increased hippocampal pathology, irrespective of whether mice were hypoglycemic or hyperglycemic prior to kainate administration. Moreover, we found that exogenous administration of glucose following kainic acid seizures significantly reduced the extent of hippocampal pathology in FVB/N mice (excitotoxin cell death susceptible) following systemic administration of kainic acid. Conclusion These findings demonstrate that modulation of the glycemic index can modify the outcome of brain injury in the kainate model of seizure induction. Moreover, modulation of the glycemic index through glucose rescue greatly diminishes the extent of seizure-induced cell death following kainate administration. Our data support the hypothesis that deficient insulin signaling may represent a critical contributing factor in the susceptibility to seizure-induced cell death and this may be an important therapeutic target.
Collapse
Affiliation(s)
- Paula Elyse Schauwecker
- Department of Cell and Neurobiology, USC Keck School of Medicine, 1333 San Pablo Street, BMT 403, Los Angeles, CA 90089-9112, USA.
| |
Collapse
|
8
|
Cooke SF, Bear MF. Stimulus-selective response plasticity in the visual cortex: an assay for the assessment of pathophysiology and treatment of cognitive impairment associated with psychiatric disorders. Biol Psychiatry 2012; 71:487-95. [PMID: 22019003 DOI: 10.1016/j.biopsych.2011.09.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 08/10/2011] [Accepted: 09/01/2011] [Indexed: 10/16/2022]
Abstract
Long-term potentiation (LTP) is a form of experimentally induced enhancement of chemical synaptic transmission that has long been proposed as a model of the endogenous processes of synaptic plasticity that mediate memory. There is a large body of evidence that the molecular mechanisms underlying experimentally induced LTP also subserve various forms of naturally occurring, experience-dependent synaptic plasticity in animals and humans. Here we describe a phenomenon called stimulus-specific response potentiation (SRP), which occurs in the primary visual cortex of mice as a result of repeated exposure to visual stimuli and is believed to reveal the mechanisms that underlie perceptual learning. We first describe evidence that SRP represents naturally occurring LTP of thalamo-cortical synaptic transmission. We then discuss the potential value of SRP as a preclinical assay for the assessment of putative drug treatments on synaptic plasticity. Stimulus-specific response potentiation is not only easy to assay and robust but captures features of feed-forward glutamatergic function and visual learning that are deficient in human psychiatric disorders, notably including schizophrenia. We suggest that phenomena analogous to SRP in humans are likely to be useful biomarkers of altered cortical LTP and of treatment response in diseases associated with impaired cognition.
Collapse
Affiliation(s)
- Sam F Cooke
- Howard Hughes Medical Institute, The Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
9
|
Synaptic targeting of AMPA receptors is regulated by a CaMKII site in the first intracellular loop of GluA1. Proc Natl Acad Sci U S A 2010; 107:22266-71. [PMID: 21135237 DOI: 10.1073/pnas.1016289107] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The accumulation of AMPA receptors (AMPARs) at synapses is essential for excitatory synaptic transmission. However, the mechanisms underlying synaptic targeting of AMPARs remain elusive. We have now used a molecular replacement approach on an AMPAR-null background to investigate the targeting mechanisms necessary for regulating AMPAR trafficking in the hippocampus. Although there is an extensive literature on the role of the GluA1 C-tail in AMPAR trafficking, there is no effect of overexpressing the C-tail on basal transmission. Instead, we found that the first intracellular loop domain (Loop1) of GluA1, a previously overlooked region within AMPARs, is critical for receptor targeting to synapses, but not for delivery of receptors to the plasma membrane. We also identified a CaMKII phosphorylation site (S567) in the GluA1 Loop1, which is phosphorylated in vitro and in vivo. Furthermore, we show that S567 is a key residue that regulates Loop1-mediated AMPAR trafficking. Thus, our study reveals a unique mechanism for targeting AMPARs to synapses to mediate synaptic transmission.
Collapse
|
10
|
Beazely MA, Lim A, Li H, Trepanier C, Chen X, Sidhu B, Macdonald JF. Platelet-derived growth factor selectively inhibits NR2B-containing N-methyl-D-aspartate receptors in CA1 hippocampal neurons. J Biol Chem 2008; 284:8054-63. [PMID: 19106110 DOI: 10.1074/jbc.m805384200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Platelet-derived growth factor (PDGF) beta receptor activation inhibits N-methyl-d-aspartate (NMDA)-evoked currents in hippocampal and cortical neurons via the activation of phospholipase Cgamma, PKC, the release of intracellular calcium, and a rearrangement of the actin cytoskeleton. In the hippocampus, the majority of NMDA receptors are heteromeric; most are composed of 2 NR1 subunits and 2 NR2A or 2 NR2B subunits. Using NR2B- and NR2A-specific antagonists, we demonstrate that PDGF-BB treatment preferentially inhibits NR2B-containing NMDA receptor currents in CA1 hippocampal neurons and enhances long-term depression in an NR2B subunit-dependent manner. Furthermore, treatment of hippocampal slices or cultures with PDGF-BB decreases the surface localization of NR2B but not of NR2A subunits. PDGFbeta receptors colocalize to a higher degree with NR2B subunits than with NR2A subunits. After neuronal injury, PDGFbeta receptors and PDGF-BB are up-regulated and PDGFbeta receptor activation is neuroprotective against glutamate-induced neuronal damage in cultured neurons. We demonstrate that the neuroprotective effects of PDGF-BB are occluded by the NR2B antagonist, Ro25-6981, and that PDGF-BB promotes NMDA signaling to CREB and ERK1/2. We conclude that PDGFbetaR signaling, by preferentially targeting NR2B receptors, provides an important mechanism for neuroprotection by growth factors in the central nervous system.
Collapse
Affiliation(s)
- Michael A Beazely
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | | | | | |
Collapse
|
11
|
Tavalin SJ. AKAP79 selectively enhances protein kinase C regulation of GluR1 at a Ca2+-calmodulin-dependent protein kinase II/protein kinase C site. J Biol Chem 2008; 283:11445-52. [PMID: 18305116 PMCID: PMC2431053 DOI: 10.1074/jbc.m709253200] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 02/22/2008] [Indexed: 01/12/2023] Open
Abstract
Enhancement of AMPA receptor activity in response to synaptic plasticity inducing stimuli may arise, in part, through phosphorylation of the GluR1 AMPA receptor subunit at Ser-831. This site is a substrate for both Ca(2+)-calmodulin-dependent protein kinase II (CaMKII) and protein kinase C (PKC). However, neuronal protein levels of CaMKII may exceed those of PKC by an order of magnitude. Thus, it is unclear how PKC could effectively regulate this common target site. The multivalent neuronal scaffold A-kinase-anchoring protein 79 (AKAP79) is known to bind PKC and is linked to GluR1 by synapse-associated protein 97 (SAP97). Here, biochemical studies demonstrate that AKAP79 localizes PKC activity near the receptor, thus accelerating Ser-831 phosphorylation. Complementary electrophysiological studies indicate that AKAP79 selectively shifts the dose-dependence for PKC modulation of GluR1 receptor currents approximately 20-fold, such that low concentrations of PKC are as effective as much higher CaMKII concentrations. By boosting PKC activity near a target substrate, AKAP79 provides a mechanism to overcome limitations in kinase abundance thereby ensuring faithful signal propagation and efficient modification of AMPA receptor-mediated responses.
Collapse
Affiliation(s)
- Steven J Tavalin
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA.
| |
Collapse
|
12
|
Kornreich BG, Niu L, Roberson MS, Oswald RE. Identification of C-terminal domain residues involved in protein kinase A-mediated potentiation of kainate receptor subtype 6. Neuroscience 2007; 146:1158-68. [PMID: 17379418 PMCID: PMC2700767 DOI: 10.1016/j.neuroscience.2007.02.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 02/07/2007] [Accepted: 02/07/2007] [Indexed: 11/18/2022]
Abstract
Glutamate receptors are the major excitatory receptors in the vertebrate CNS and have been implicated in a number of physiological and pathological processes. Previous work has shown that glutamate receptor function may be modulated by protein kinase A (PKA)-mediated phosphorylation, although the molecular mechanism of this potentiation has remained unclear. We have investigated the phosphorylation of specific amino acid residues in the C-terminal cytoplasmic domain of the rat kainate receptor subtype 6 (GluR6) as a possible mechanism for regulation of receptor function. The C-terminal tail of rat GluR6 can be phosphorylated by PKA on serine residues as demonstrated using [gamma-32P]ATP kinase assays. Whole cell recordings of transiently transfected human embryonic kidney (HEK) 293 cells showed that phosphorylation by PKA potentiates whole cell currents in wildtype GluR6 and that removal of the cytoplasmic C-terminal domain abolishes this potentiation. This suggested that the C-terminal domain may contain residue(s) involved in the PKA-mediated potentiation. Single mutations of each serine residue in the C-terminal domain (S815A, S825A, S828A, and S837A) and a truncation after position 855, which removes all threonines (T856, T864, and T875) from the domain, do not abolish PKA potentiation. However, the S825A/S837A mutation, but no other double mutation, abolishes potentiation. These results demonstrate that phosphorylation of the C-terminal tail of GluR6 by PKA leads to potentiation of whole cell response, and the combination of S825 and S837 in the C-terminal domain is a vital component of the mechanism of GluR6 potentiation by PKA.
Collapse
Affiliation(s)
| | - Li Niu
- Dept of Molecular Medicine, Cornell University, Ithaca, NY 14853
- Dept of Chemistry, Center of Biochemistry & Biophysics, SUNY Albany, Albany, NY 12222
| | - Mark S. Roberson
- Dept of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Robert E. Oswald
- Dept of Molecular Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
13
|
Gao ZB, Chen XQ, Hu GY. Inhibition of excitatory synaptic transmission by trans-resveratrol in rat hippocampus. Brain Res 2006; 1111:41-7. [PMID: 16876771 DOI: 10.1016/j.brainres.2006.06.096] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2006] [Revised: 06/20/2006] [Accepted: 06/27/2006] [Indexed: 02/05/2023]
Abstract
The red wine polyphenol trans-resveratrol has been found to exert potent protective actions in a variety of cerebral ischemia models. The neuroprotection by trans-resveratrol thus far is mainly attributed to its intrinsic antioxidant properties. In the present study, the effects of the red wine polyphenol on excitatory synaptic transmission were investigated in the CA1 region of rat hippocampal slices. Perfusion with trans-resveratrol (10-100 microM) caused a concentration-dependent inhibition on the filed excitatory postsynaptic potentials (the field EPSPs) without detectable effect on the presynaptic volleys. The inhibition had a slow onset and was reversible. Trans-resveratrol (30 microM) did not change the ratios of paired-pulse facilitation of the field EPSPs tested at intervals of 20, 40 and 80 ms, nor did it alter the membrane properties of postsynaptic CA1 pyramidal neurons. However, trans-resveratrol (30 microM) significantly suppressed glutamate-induced currents in postsynaptic CA1 pyramidal neurons. In dissociated hippocampal neurons, the IC(50) value of trans-resveratrol in inhibition of glutamate-induced currents was 53.3+/-9.4 microM. Kainite and NMDA receptors were more sensitive to the red wine polyphenol than AMPA receptors. The present study for the first time demonstrates that trans-resveratrol inhibits the postsynaptic glutamate receptors, which probably works in concert with its antioxidant action for ameliorating the brain ischemic injury. The findings also support the future use of trans-resveratrol in the treatment of various neurodegenerative disorders.
Collapse
Affiliation(s)
- Zhao-Bing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 555 Zu-Chong-Zhi Road, Shanghai 201203, PR China
| | | | | |
Collapse
|
14
|
Grabauskas G, Chapman H, Wheal HV. Role of protein kinase C in modulation of excitability of CA1 pyramidal neurons in the rat. Neuroscience 2006; 139:1301-13. [PMID: 16533575 DOI: 10.1016/j.neuroscience.2006.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 01/06/2006] [Accepted: 01/21/2006] [Indexed: 11/18/2022]
Abstract
Biochemical and in situ hybridization studies demonstrated that the levels of protein kinase C variants were significantly increased in the hippocampus of the experimental models of epilepsy in rats. In addition it has been demonstrated that protein kinase C plays an important role in modulating synaptic transmission in the hippocampus. We examined the effects of activating of protein kinase C on the excitability of CA1 pyramidal neurons and synaptic transmission, using whole-cell current-clamp and extracellular field potential recording techniques. Indolactam V (1 microM) a novel protein kinase C activator, increased the excitability of CA1 neurons acting at both pre- and post-synaptic sites. Indolactam V, acting postsynaptically, significantly reduced the threshold for initiation of action potential from -42+/-3.8 mV to -51+/-3.1 mV and selectively inhibited the slow afterhyperpolarizing potential. Indolactam V also altered the neuronal firing properties in response to prolonged depolarizing pulse by eliminating the spike frequency accommodation. Our data indicate that indolactam V potentiated both amplitudes of Shaffer-collateral stimulation evoked excitatory postsynaptic currents and disynaptically evoked inhibitory evoked postsynaptic currents. However, the potentiation of inhibitory evoked postsynaptic currents amplitudes was not observed after blockade of NMDA and AMPA/kainate currents suggesting it was due to excitatory activity driving inhibitory neurons. The results indicate that the potentiation of pharmacologically isolated excitatory postsynaptic currents (215% of control) and amplitudes of population spikes (290% of control) was due to action of indolactam V presynaptically since the agonist reduced the paired-pulse ratio and the potentiating effect was not blocked by dialyzing the postsynaptic neuron through the recording electrode with a specific protein kinase C inactivator calphostin C. These findings suggest that protein kinase C increases the amplitude of epileptiform activity by causing potentiation of excitatory synaptic transmission, increasing the excitability of postsynaptic neurons and reducing negative feed back provided by slow afterhyperpolarizing potential.
Collapse
Affiliation(s)
- G Grabauskas
- Neuroscience Research Group, School of Biological Sciences, University of Southampton, Bassett Crescent East, UK
| | | | | |
Collapse
|
15
|
Hori K, Yasuda H, Konno D, Maruoka H, Tsumoto T, Sobue K. NMDA receptor-dependent synaptic translocation of insulin receptor substrate p53 via protein kinase C signaling. J Neurosci 2006; 25:2670-81. [PMID: 15758177 PMCID: PMC6725157 DOI: 10.1523/jneurosci.3638-04.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The activity-dependent remodeling of postsynaptic structure is a fundamental process underlying learning and memory. Insulin receptor substrate p53 (IRSp53), a key player in cytoskeletal dynamics, is enriched in the postsynaptic density (PSD) fraction, but its significance in synaptic functions remains unclear. We report here that IRSp53 is accumulated rapidly at the postsynaptic sites of cultured hippocampal neurons after glutamate or NMDA stimulation in an actin cytoskeleton-dependent manner. Pharmacological profiles showed that a PKC inhibitor, but not other kinase inhibitors, specifically suppressed the synaptic translocation of IRSp53 in response to NMDA, and the selective activation of PKC with phorbol ester markedly induced the synaptic translocation. Reverse transcriptase-PCR and Western blotting showed that IRSp53-S is the major isoform expressed in cultured hippocampal neurons. The synaptic targeting of IRSp53-S was found to be mediated through N-terminal coiled-coil domain and the PDZ (PSD-95/Discs large/zona occludens-1)-binding sequence at its C-terminal end and regulated by the PKC phosphorylation of its N terminus. In electrophysiological experiments, overexpression of IRSp53-S wild type and IRSp53-S mutant that is spontaneously accumulated at the postsynaptic sites enhanced the postsynaptic function as detected by an increased miniature EPSC amplitude. These data suggest that IRSp53 is involved in NMDA receptor-linked synaptic plasticity via PKC signaling.
Collapse
Affiliation(s)
- Kei Hori
- Department of Neuroscience (D13) and Division of Neurophysiology, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | | | | | | | | |
Collapse
|
16
|
Abstract
Glutamate receptor antagonists, although effective in preventing in vitro excitotoxic death, also block the glutamatergic signalling that is essential for normal excitatory neurotransmission and neuronal survival. This has contributed to the failure of clinical trials employing glutamate receptor antagonists as stroke therapeutics. However, recent years have seen an increased understanding of the molecular organisation of glutamate receptors in the neuronal postsynaptic density. This and a dissection of their associated intracellular signalling cascades has allowed the identification of distinct pathways responsible for excitotoxicity. It has become possible to uncouple toxic signalling cascades from glutamate receptors by targeting the interactions of membrane receptors with downstream proteins. Toxic signalling can be effectively uncoupled from glutamate receptors using targeted, cell-permeable peptides to disrupt specific protein-protein interactions. This approach does not block essential excitatory neurotransmission, but attenuates neurotoxic signals specifically and reduces stroke damage. This novel approach to blocking excitotoxic signalling in cerebral ischaemia may constitute a practical approach to stroke therapy.
Collapse
Affiliation(s)
- Michelle M Aarts
- Toronto Western Hospital, Suite 4W-435, 399 Bathurst Street, Toronto, Ontario, M5T 2S8, Canada
| | | |
Collapse
|
17
|
Townsend M, Liu Y, Constantine-Paton M. Retina-driven dephosphorylation of the NR2A subunit correlates with faster NMDA receptor kinetics at developing retinocollicular synapses. J Neurosci 2004; 24:11098-107. [PMID: 15590926 PMCID: PMC6730286 DOI: 10.1523/jneurosci.1207-04.2004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2004] [Revised: 10/25/2004] [Accepted: 10/30/2004] [Indexed: 11/21/2022] Open
Abstract
We describe a homeostatic mechanism that limits NMDA receptor currents in response to early light activation of a developing visual pathway. During the second postnatal week of rodent retinocollicular development, the Ca2+-activated phosphatase calcineurin (CaN) mediates a rapid, activity-induced shortening in the decay time of NMDA receptor (NMDAR) currents. We show that protein kinase A acts in opposition to CaN to maintain NMDAR currents with long decay times. The CaN-mediated change is coincident with the initial expression of the NMDAR subunit NR2A. Using NR2A knock-out mice and dialyzing neurons with a constitutively active CaN, we demonstrate that NR2A subunits are necessary for the effect of CaN on NMDAR current kinetics. In wild-type mice, Ser900 of NR2A, previously implicated in CaN-mediated glycine-independent desensitization, becomes chronically dephosphorylated by postnatal day 11 as NMDAR current decay times become faster. Pharmacologically disrupting early photoreceptor-driven activity in the retina eliminates the dephosphorylation of NR2A and prevents the shortening in NMDAR current decay time. These data suggest that the developmental onset of retinal activity increases CaN-mediated dephosphorylation of NR2A subunits newly incorporated into synaptic NMDARs of the superior colliculus, thereby providing a mechanism for the early and rapid reduction of NMDAR current decay time in visual neurons.
Collapse
Affiliation(s)
- Matthew Townsend
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
18
|
Guan Y, Guo W, Robbins MT, Dubner R, Ren K. Changes in AMPA receptor phosphorylation in the rostral ventromedial medulla after inflammatory hyperalgesia in rats. Neurosci Lett 2004; 366:201-5. [PMID: 15276247 DOI: 10.1016/j.neulet.2004.05.051] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Revised: 05/13/2004] [Accepted: 05/15/2004] [Indexed: 10/26/2022]
Abstract
To study the glutamatergic mechanisms underlying changes in excitability in the brain stem pain modulatory circuitry after injury, we examined GluR1 serine 831 phosphorylation in the rostral ventromedial medulla (RVM) after complete Freund's adjuvant-induced hindpaw inflammation. Western blots indicated a rapid and prolonged (30 min and 7 days post-inflammation) increase in phosphoserine 831 GluR1 protein levels in the RVM. The upregulated GluR1 phosphorylation was blocked by pretreatment, but not by post-treatment, with the local anesthetic, lidocaine, at the site of inflammation. The upregulation of phosphoserine 831 GluR1 was attenuated by pretreatment with chelerythrine, a selective PKC inhibitor, KN-93, a selective CaMKII inhibitor, and two NMDA receptor antagonists, MK-801 and APV. These findings provide new evidence linking in vivo AMPA receptor phosphorylation in the RVM pain modulatory circuitry to the enhanced descending pain modulation after inflammation.
Collapse
Affiliation(s)
- Yun Guan
- Department of Biomedical Sciences, Dental School and Program in Neuroscience University of Maryland, Rm. 5A12, 666 W. Baltimore Street, Baltimore, MD 21201, USA
| | | | | | | | | |
Collapse
|
19
|
Goforth PB, Ellis EF, Satin LS. Mechanical Injury Modulates AMPA Receptor Kinetics via an NMDA Receptor–Dependent Pathway. J Neurotrauma 2004; 21:719-32. [PMID: 15253800 DOI: 10.1089/0897715041269704] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Alterations in glutamatergic transmission are thought to contribute to secondary neuronal damage following traumatic brain injury. Using an in vitro cell injury model, we previously demonstrated an apparent reduction in AMPA receptor desensitization and resultant potentiation of AMPA-evoked currents after stretch injury of cultured neonatal rat cortical neurons. In the present study, we sought to further characterize injury-induced enhancement of AMPA current and elucidate the mechanisms responsible for this pathological process. Using the patch-clamp technique, agonist-activated currents were recorded from control and injured neurons. Potentiation of AMPA-mediated currents occurred quickly, within 15-30 min following injury, and persisted for at least 24 h. Stretch-injury slowed the activation and desensitization of AMPA mediated currents recorded from excised outside-out patches. The co-application of 100 microM AMPA and 20 microM thiocyanate enhanced AMPA receptor desensitization in control neurons and restored desensitization in injured neurons. The potentiation of AMPA-elicited current was prevented by the NMDA receptor antagonist D-APV (20 microM) or the CaMKII inhibitor KN93 (10 microM). These results suggest that mechanical injury initiates a biochemical cascade that involves NMDA receptor and CaMKII activation and produces a long-lasting reduction of AMPA receptor desensitization, which may contribute to the pathophysiology of traumatic brain injury.
Collapse
Affiliation(s)
- Paulette B Goforth
- Department of Pharmacology and Toxicology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | | | | |
Collapse
|
20
|
Bauman AL, Goehring AS, Scott JD. Orchestration of synaptic plasticity through AKAP signaling complexes. Neuropharmacology 2004; 46:299-310. [PMID: 14975685 DOI: 10.1016/j.neuropharm.2003.09.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2003] [Accepted: 09/16/2003] [Indexed: 11/20/2022]
Abstract
Significant progress has been made toward understanding the mechanisms by which organisms learn from experiences and how those experiences are translated into memories. Advances in molecular, electrophysiological and genetic technologies have permitted great strides in identifying biochemical and structural changes that occur at synapses during processes that are thought to underlie learning and memory. Cellular events that generate the second messenger cyclic AMP (cAMP) and activate protein kinase A (PKA) have been linked to synaptic plasticity and long-term memory. In this review we will focus on the role of PKA in synaptic plasticity and discuss how the compartmentalization of PKA through its association with A-Kinase Anchoring Proteins (AKAPs) affect PKA function in this process.
Collapse
Affiliation(s)
- Andrea L Bauman
- Howard Hughes Medical Institute, Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
21
|
Harlan RE, Kailas SR, Tagoe CEF, Garcia MM. Morphine actions in the rat forebrain: role of protein kinase C. Brain Res Bull 2004; 62:285-95. [PMID: 14709343 DOI: 10.1016/j.brainresbull.2003.09.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Acute administration of morphine induces expression of the immediate-early gene (IEG) c-Fos in dorsomedial striatum, portions of cerebral cortex, and in several midline-intralaminar thalamic nuclei, partly via a trans-synaptic mechanism that involves activation of glutamate receptors. Because activation of protein kinase C (PKC) may occur following the activation of glutamate receptors, we determined whether pharmacological inhibition of PKC would attenuate morphine-induced c-Fos expression, and whether acute administration of morphine would induce translocation of PKC. The selective PKC antagonist NPC 15437 given 30 min prior to morphine significantly decreased morphine-induced c-Fos expression in striatum and cingulate cortex, but not in centrolateral thalamus. In another experiment, rats were given an acute dose of morphine, and immunocytochemical analysis was performed for the betaI and betaII isoforms of PKC. Morphine induced a rapid and transient translocation of PKC betaII, but not betaI, from perinuclear spots to plasma membrane in numerous cortical and striatal neurons. Prior administration of naloxone blocked this response. Ultrastructural studies confirmed translocation from Golgi apparatus to plasma membrane 15 min after morphine injection. Double immunocytochemistry at the light microscopic level demonstrated co-localization of translocated PKC betaII and c-Fos in some cortical neurons 90 min after morphine injection. These results support a role for PKC, especially PKC betaII, in the rapid effects of morphine on the brain.
Collapse
Affiliation(s)
- Richard E Harlan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, 1430 Tulane Avenue, SL-49, New Orleans, LA 70112, USA.
| | | | | | | |
Collapse
|
22
|
Abstract
Excitatory synapses in the CNS release glutamate, which acts primarily on two sides of ionotropic receptors: AMPA receptors and NMDA receptors. AMPA receptors mediate the postsynaptic depolarization that initiates neuronal firing, whereas NMDA receptors initiate synaptic plasticity. Recent studies have emphasized that distinct mechanisms control synaptic expression of these two receptor classes. Whereas NMDA receptor proteins are relatively fixed, AMPA receptors cycle synaptic membranes on and off. A large family of interacting proteins regulates AMPA receptor turnover at synapses and thereby influences synaptic strength. Furthermore, neuronal activity controls synaptic AMPA receptor trafficking, and this dynamic process plays a key role in the synaptic plasticity that is thought to underlie aspects of learning and memory.
Collapse
Affiliation(s)
- David S Bredt
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | |
Collapse
|
23
|
Gomes AR, Correia SS, Carvalho AL, Duarte CB. Regulation of AMPA receptor activity, synaptic targeting and recycling: role in synaptic plasticity. Neurochem Res 2003; 28:1459-73. [PMID: 14570391 DOI: 10.1023/a:1025610122776] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors for the neurotransmitter glutamate are oligomeric structures responsible for most fast excitatory responses in the central nervous system. The activity of AMPA receptors can be directly regulated by protein phosphorylation, which may also affect the interaction with intracellular proteins and, consequently, their recycling and localization to defined postsynaptic sites. This review focuses on recent advances in understanding the dynamic regulation of AMPA receptors, on a short- and long-term basis, and its implications in synaptic plasticity.
Collapse
Affiliation(s)
- André R Gomes
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | | | | | | |
Collapse
|
24
|
Aarts MM, Tymianski M. Novel treatment of excitotoxicity: targeted disruption of intracellular signalling from glutamate receptors. Biochem Pharmacol 2003; 66:877-86. [PMID: 12963474 DOI: 10.1016/s0006-2952(03)00297-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glutamate signalling plays key physiological roles in excitatory neurotransmission and CNS plasticity, but also mediates excitotoxicity, the process responsible for triggering neurodegeneration through glutamate receptor overactivation. Excitotoxicity is thought to be a key neurotoxic mechanism in neurological disorders, including brain ischemia, CNS trauma and epilepsy. However, treating excitotoxicity using glutamate receptor antagonists has not proven clinically viable, necessitating more sophisticated approaches. Increasing knowledge of the composition of the postsynaptic density at glutamatergic synapses has allowed us to extend our understanding of the molecular mechanisms of excitotoxicity and to dissect out the distinct signalling pathways responsible for excitotoxic damage. Key molecules in these pathways are physically linked to the cytoplasmic face of glutamate receptors by scaffolding proteins that exhibit binding specificity for some receptors over others. This imparts specificity to physiological and pathological glutamatergic signalling. Recently, we have capitalized on this knowledge and, using targeted peptides to selectively disrupt intracellular interactions linked to glutamate receptors, have blocked excitotoxic signalling in neurones. This therapeutic approach circumvents the negative consequences of blocking glutamate receptors, and may be a practical strategy for treating neurological disorders that involve excitotoxicity.
Collapse
Affiliation(s)
- Michelle M Aarts
- Toronto Western Research Institute, McPav 11-416, 399 Bathurst Street, Toronto, Ont., Canada M5T 2S8.
| | | |
Collapse
|
25
|
Li HF, Kendig JJ. Ethanol withdrawal hyper-responsiveness mediated by NMDA receptors in spinal cord motor neurons. Br J Pharmacol 2003; 139:73-80. [PMID: 12746225 PMCID: PMC1573814 DOI: 10.1038/sj.bjp.0705198] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2002] [Accepted: 01/20/2003] [Indexed: 11/09/2022] Open
Abstract
1. Following ethanol (EtOH) exposure, population excitatory postsynaptic potentials (pEPSPs) in isolated spinal cord increase to a level above control (withdrawal hyper-responsiveness). The present studies were designed to characterize this phenomenon and in particular to test the hypothesis that protein kinases mediate withdrawal. 2. Patch-clamp studies were carried out in motor neurons in rat spinal cord slices. Currents were evoked by brief pulses of glutamate, alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) or N-methyl-D-aspartic acid (NMDA). 3. Of 15 EtOH-sensitive neurons in which currents were evoked by glutamate, four (27%) displayed withdrawal hyper-responsiveness in the washout period. Mean current area after washout was 129.6+/-5% of control. 4. When currents were evoked by AMPA, two of 10 neurons (20%) displayed withdrawal hyper-responsiveness, with a mean current area 122+/-8% of control on washout. 5. Of a group of 11 neurons in which currents were evoked by NMDA, nine (82%) displayed withdrawal hyper-responsiveness. Mean increase in current area at the end of the washout period was to 133+/-6% of control (n=9, P<0.001). When NMDA applications were stopped during the period of EtOH exposure, mean area of NMDA-evoked responses on washout was only 98.0+/-5% of control (n=6, P>0.05). 6. The tyrosine kinase inhibitor genistein (10-20 microM) blocked withdrawal hyper-responsiveness. Of six EtOH-sensitive neurons, the mean NMDA-evoked current area after washout was 89+/-6% of control, P>0.05. 7 The protein kinase A (PKA) inhibitor Rp-cAMP (20-500 microM) did not block withdrawal hyper-responsiveness. On washout, the mean NMDA-evoked current area was 124+/-6% of control (n=5, P<0.05). 8 Two broad-spectrum specific protein kinase C (PKC) inhibitors, GF-109203X (0.3 microM) and chelerythrine chloride (0.5-2 nM), blocked withdrawal hyper-responsiveness. Responses on washout were 108+/-7%, n=5 and 88+/-4%, n=4 of control, respectively, P>0.05. 9 NMDA activation during EtOH exposure is necessary for withdrawal hyper-responsiveness. Both tyrosine kinase and PKC, but not PKA, appear to be essential for EtOH withdrawal hyper-responsiveness mediated by postsynaptic NMDA receptors in spinal cord motor neurons.
Collapse
Affiliation(s)
- Hui-Fang Li
- Department of Anesthesia, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305, U.S.A.
| | | |
Collapse
|
26
|
Chen HX, Roper SN. PKA and PKC enhance excitatory synaptic transmission in human dentate gyrus. J Neurophysiol 2003; 89:2482-8. [PMID: 12611980 DOI: 10.1152/jn.01031.2002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
cAMP-dependent protein kinase (PKA) and protein kinase C (PKC) are two major modulators of synaptic transmission in the CNS but little is known about how they affect synaptic transmission in the human CNS. In this study, we used forskolin, a PKA activator, and phorbol ester, a PKC activator, to examine the effects of these kinases on synaptic transmission in granule cells of the dentate gyrus in human hippocampal slices using whole-cell recording methods. We found that both forskolin and phorbol ester increased the frequency of spontaneous and miniature excitatory postsynaptic currents (sEPSCs and mEPSCs) but left the amplitude unaffected. Inactive forskolin and phorbol ester had no effect on sEPSCs in human dentate granule cells. Prior application of forskolin occluded the effects of phorbol ester on mEPSC frequency. Tetanic stimulation applied to the perforant path induced short-term depression in dentate gyrus granule cells. Both forskolin and phorbol ester significantly enhanced this short-term depression. Taken together, these results demonstrate that PKA and PKC are involved in up-regulation of excitatory synaptic transmission in human dentate granule cells, primarily by presynaptic mechanisms. In addition, the occlusion experiments suggest that the two kinases may share a common signal pathway.
Collapse
Affiliation(s)
- Huan-Xin Chen
- Department of Neurological Surgery and Evelyn F. and William L. McKnight Brain Institute, Gainesville, Florida 32610, USA
| | | |
Collapse
|
27
|
Abstract
Ionotropic neurotransmitter receptors mediate rapid synaptic transmission in the CNS and PNS. Owing to this central role in trans-synaptic signal transduction, modulation of these receptors could play a crucial role in the expression of synaptic plasticity in the brain. AMPA receptors mediate the majority of rapid excitatory synaptic transmission in the CNS. Recent studies have indicated that the activity and synaptic distribution of these receptors is dynamically regulated and could be crucial for the short- and long-term modification of synaptic efficacy. Here we review recent data on the molecular mechanisms that underlie the modulation of AMPA receptors and the role of AMPA-receptor regulation in mediating synaptic plasticity.
Collapse
Affiliation(s)
- Insuk Song
- Dept of Neuroscience, Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, 904A PCTB, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | | |
Collapse
|
28
|
Zeron MM, Hansson O, Chen N, Wellington CL, Leavitt BR, Brundin P, Hayden MR, Raymond LA. Increased sensitivity to N-methyl-D-aspartate receptor-mediated excitotoxicity in a mouse model of Huntington's disease. Neuron 2002; 33:849-60. [PMID: 11906693 DOI: 10.1016/s0896-6273(02)00615-3] [Citation(s) in RCA: 437] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Previous work suggests N-methyl-D-aspartate receptor (NMDAR) activation may be involved in degeneration of medium-sized spiny striatal neurons in Huntington's disease (HD). Here we show that these neurons are more vulnerable to NMDAR-mediated death in a YAC transgenic FVB/N mouse model of HD expressing full-length mutant huntingtin, compared with wild-type FVB/N mice. Excitotoxic death of these neurons was increased after intrastriatal injection of quinolinate in vivo, and after NMDA but not AMPA exposure in culture. NMDA-induced cell death was abolished by an NR2B subtype-specific antagonist. In contrast, NMDAR-mediated death of cerebellar granule neurons was not enhanced, consistent with cell-type and NMDAR subtype specificity. Moreover, increased NMDA-evoked current amplitude and caspase-3 activity were observed in transgenic striatal neurons. Our data support a role for NR2B-subtype NMDAR activation as a trigger for selective neuronal degeneration in HD.
Collapse
Affiliation(s)
- Melinda M Zeron
- Kinsmen Laboratory of Neurological Research, Department of Psychiatry, 221 84, Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Dineley KT, Weeber EJ, Atkins C, Adams JP, Anderson AE, Sweatt JD. Leitmotifs in the biochemistry of LTP induction: amplification, integration and coordination. J Neurochem 2001; 77:961-71. [PMID: 11359861 DOI: 10.1046/j.1471-4159.2001.00321.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hippocampal long-term potentiation (LTP) is a robust and long-lasting form of synaptic plasticity that is the leading candidate for a cellular mechanism contributing to mammalian learning and memory. Investigations over the past decade have revealed that the biochemistry of LTP induction involves mechanisms of great subtlety and complexity. This review highlights themes that have emerged as a result of our increased knowledge of the signal transduction pathways involved in the induction of NMDA receptor-dependent LTP in area CA1 of the hippocampus. Among these themes are signal amplification, signal integration and signal coordination. Here we use these themes as an organizing context for reviewing the profusion of signaling mechanisms involved in the induction of LTP.
Collapse
Affiliation(s)
- K T Dineley
- Division of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Glutamate is the major excitatory neurotransmitter in the brain. It acts at ligand-gated cationic channels (NMDA, AMPA and kainate receptors) and at G protein-coupled metabotropic glutamate receptors as well. The glutamatergic transmission is suggested to be involved in development, learning and memory. Its dysfunction can be detected in epilepsy, stroke, neurodegenerative disorders and drug abuse. This paper summarizes the present knowledge on the modulation of glutamate-gated ion channels in the central nervous system by phosphorylation. An inhibitory interaction between adenosine A2A receptors and NMDA receptors in the neostriatum is described as an example. mediated by the phospholipase C/inositol trisphosphate/calmodulin and calmodulin kinase II pathway.
Collapse
Affiliation(s)
- L Köles
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, Germany
| | | | | |
Collapse
|
31
|
Phosphorylation of the AMPA receptor subunit GluR2 differentially regulates its interaction with PDZ domain-containing proteins. J Neurosci 2001. [PMID: 11007883 DOI: 10.1523/jneurosci.20-19-07258.2000] [Citation(s) in RCA: 392] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PSD-95, DLG, ZO-1 (PDZ) domain-mediated protein interactions have been shown to play important roles in the regulation of glutamate receptor function at excitatory synapses. Recent studies demonstrating the rapid regulation of AMPA receptor function during synaptic plasticity have suggested that AMPA receptor interaction with PDZ domain-containing proteins may be dynamically modulated. Here we show that PKC phosphorylation of the AMPA receptor GluR2 subunit differentially modulates its interaction with the PDZ domain-containing proteins GRIP1 and PICK1. The serine residue [serine-880 (Ser880)] in the GluR2 C-terminal sequence (IESVKI) critical for PDZ domain binding is a substrate of PKC and is phosphorylated in vivo. In vitro binding and coimmunoprecipitation studies show that phosphorylation of serine-880 within the GluR2 PDZ ligand significantly decreases GluR2 binding to GRIP1 but not to PICK1. Immunostaining of cultured hippocampal neurons demonstrates that the Ser880-phosphorylated GluR2 subunits are enriched and colocalized with PICK1 in the dendrites, with very little staining observed at excitatory synapses. Interestingly, PKC activation in neurons increases the Ser880 phosphorylation of GluR2 subunits and recruits PICK1 to excitatory synapses. Moreover, PKC stimulation in neurons results in rapid internalization of surface GluR2 subunits. These results suggest that GluR2 phosphorylation of serine-880 may be important in the regulation of the AMPA receptor internalization during synaptic plasticity.
Collapse
|
32
|
Hicks JH, Dani JA, Lester RA. Regulation of the sensitivity of acetylcholine receptors to nicotine in rat habenula neurons. J Physiol 2000; 529 Pt 3:579-97. [PMID: 11118491 PMCID: PMC2270233 DOI: 10.1111/j.1469-7793.2000.00579.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Time-dependent changes in nicotinic acetylcholine receptor (nAChR) function were studied in acutely isolated medial habenula neurons during whole-cell perfusion. The peak amplitude of inward currents induced by 1 s pulses of nicotinic agonists, applied at 30 s intervals, gradually increased over the first several minutes of whole-cell recording. The ratio of response amplitudes at 1 and 15 min (t15/t1) was 1.9. Run-up of responses occurred independently of channel activation and was specific to nAChRs. The channel blocker chlorisondamine (30 microM), co-applied with nicotine, was used to irreversibly block the majority (91 %) of the nAChRs that opened in the first 2 min of recording. Run-up in the remaining 9 % unblocked channels assessed at 15 min (t15/t2 = 3.4) was similar to that in control cells not exposed to nicotine and chlorisondamine simultaneously, implying that run-up is not due to the incorporation of new receptors. A marked alteration in the sensitivity of nAChRs to extracellular Ca2+ was also observed during whole-cell perfusion. The ratio of current amplitudes obtained in 0.2 and 4.0 mM Ca2+ changed from 0.54 (t = 5 min) to 0.82 (t = 30 min). Inward rectification of nicotine-induced responses was reduced during internal dialysis. Voltages for half-maximal conductance were -23.0 and -13.8 mV at 2 and 15 min, respectively. Inclusion of either free Mg2+ ( approximately 2 mM) or spermine (100 microM) in the internal solution counteracted the change in rectification, but did not prevent run-up. The period of run-up was followed by a use-dependent run-down phase. Little run-down in peak current amplitude was induced provided that agonist was applied infrequently (5 min intervals), whereas applications at 30 s intervals produced a loss of channel function after approximately 15 min whole-cell perfusion. The time at which run-down began ( approximately 5-30 min) was correlated with the initial rate of nAChR desensitization ( approximately 200-4000 ms); slowly desensitizing nicotinic currents demonstrated delayed run-down. We suggest that run-up of nAChR-mediated responses does not require receptor activation and may result from a change in channel open probability. We also hypothesize that channel run-down reflects accumulation of nAChRs in long-lived desensitized/inactivated states.
Collapse
Affiliation(s)
- J H Hicks
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
33
|
Daw MI, Chittajallu R, Bortolotto ZA, Dev KK, Duprat F, Henley JM, Collingridge GL, Isaac JT. PDZ proteins interacting with C-terminal GluR2/3 are involved in a PKC-dependent regulation of AMPA receptors at hippocampal synapses. Neuron 2000; 28:873-86. [PMID: 11163273 DOI: 10.1016/s0896-6273(00)00160-4] [Citation(s) in RCA: 274] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We investigated the role of PDZ proteins (GRIP, ABP, and PICK1) interacting with the C-terminal GluR2 by infusing a ct-GluR2 peptide ("pep2-SVKI") into CA1 pyramidal neurons in hippocampal slices using whole-cell recordings. Pep2-SVKI, but not a control or PICK1 selective peptide, caused AMPAR-mediated EPSC amplitude to increase in approximately one-third of control neurons and in most neurons following the prior induction of LTD. Pep2-SVKI also blocked LTD; however, this occurred in all neurons. A PKC inhibitor prevented these effects of pep2-SVKI on synaptic transmission and LTD. We propose a model in which the maintenance of LTD involves the binding of AMPARs to PDZ proteins to prevent their reinsertion. We also present evidence that PKC regulates AMPAR reinsertion during dedepression.
Collapse
Affiliation(s)
- M I Daw
- MRC Centre for Synaptic Plasticity, Department of Anatomy, University of Bristol, University Walk, BS8 1TD, Bristol, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Xia J, Chung HJ, Wihler C, Huganir RL, Linden DJ. Cerebellar long-term depression requires PKC-regulated interactions between GluR2/3 and PDZ domain-containing proteins. Neuron 2000; 28:499-510. [PMID: 11144359 DOI: 10.1016/s0896-6273(00)00128-8] [Citation(s) in RCA: 303] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cerebellar LTD requires activation of PKC and is expressed, at least in part, as postsynaptic AMPA receptor internalization. Recently, it was shown that AMPA receptor internalization requires clathrin-mediated endocytosis and depends upon the carboxy-terminal region of GluR2/3. Phosphorylation of Ser-880 in this region by PKC differentially regulates the binding of the PDZ domain-containing proteins GRIP/ABP and PICK1. Peptides, corresponding to the phosphorylated and dephosphorylated GluR2 carboxy-terminal PDZ binding motif, were perfused in cerebellar Purkinje cells grown in culture. Both the dephospho form (which blocks binding of GRIP/ABP and PICK1) and the phospho form (which selectively blocks PICK1) attenuated LTD induction by glutamate/depolarization pairing, as did antibodies directed against the PDZ domain of PICK1. These findings indicate that expression of cerebellar LTD requires PKC-regulated interactions between the carboxy-terminal of GluR2/3 and PDZ domain-containing proteins.
Collapse
Affiliation(s)
- J Xia
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
35
|
Abstract
The AMPA receptors for glutamate are oligomeric structures that mediate fast excitatory responses in the central nervous system. Phosphorylation of AMPA receptors is an important mechanism for short-term modulation of their function, and is thought to play an important role in synaptic plasticity in different brain regions. Recent studies have shown that phosphorylation of AMPA receptors by cAMP-dependent protein kinase (PKA) and Ca2+- and calmodulin-dependent protein kinase II (CaMKII) potentiates their activity, but phosphorylation of the receptor subunits may also affect their interaction with intracellular proteins, and their expression at the plasma membrane. Phosphorylation of AMPA receptor subunits has also been investigated in relation to processes of synaptic plasticity. This review focuses on recent advances in understanding the molecular mechanisms of regulation of AMPA receptors, and their implications in synaptic plasticity.
Collapse
Affiliation(s)
- A L Carvalho
- Center for Neuroscience of Coimbra, Department of Zoology, University of Coimbra, Portugal.
| | | | | |
Collapse
|
36
|
Chung HJ, Xia J, Scannevin RH, Zhang X, Huganir RL. Phosphorylation of the AMPA receptor subunit GluR2 differentially regulates its interaction with PDZ domain-containing proteins. J Neurosci 2000; 20:7258-67. [PMID: 11007883 PMCID: PMC6772789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
PSD-95, DLG, ZO-1 (PDZ) domain-mediated protein interactions have been shown to play important roles in the regulation of glutamate receptor function at excitatory synapses. Recent studies demonstrating the rapid regulation of AMPA receptor function during synaptic plasticity have suggested that AMPA receptor interaction with PDZ domain-containing proteins may be dynamically modulated. Here we show that PKC phosphorylation of the AMPA receptor GluR2 subunit differentially modulates its interaction with the PDZ domain-containing proteins GRIP1 and PICK1. The serine residue [serine-880 (Ser880)] in the GluR2 C-terminal sequence (IESVKI) critical for PDZ domain binding is a substrate of PKC and is phosphorylated in vivo. In vitro binding and coimmunoprecipitation studies show that phosphorylation of serine-880 within the GluR2 PDZ ligand significantly decreases GluR2 binding to GRIP1 but not to PICK1. Immunostaining of cultured hippocampal neurons demonstrates that the Ser880-phosphorylated GluR2 subunits are enriched and colocalized with PICK1 in the dendrites, with very little staining observed at excitatory synapses. Interestingly, PKC activation in neurons increases the Ser880 phosphorylation of GluR2 subunits and recruits PICK1 to excitatory synapses. Moreover, PKC stimulation in neurons results in rapid internalization of surface GluR2 subunits. These results suggest that GluR2 phosphorylation of serine-880 may be important in the regulation of the AMPA receptor internalization during synaptic plasticity.
Collapse
Affiliation(s)
- H J Chung
- Department of Neuroscience, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
37
|
Abstract
alpha-Amino-3-hydroxy-5-methylisoxazolepropionate (AMPA) receptors mediate most fast excitatory synaptic transmission in the mammalian CNS. They play a central role in synapse stabilisation and plasticity and their prolonged activation is potently neurotoxic. Developmental and activity-dependent changes in the functional synaptic expression of these receptors are subject to tight cellular regulation. The molecular and cellular mechanisms which control the postsynaptic insertion and arrangement of individual AMPA receptor variants are therefore the subject of intense investigation and in the last two years there has been significant progress towards elucidating some of the processes involved. Much of the new information has come from the application of the yeast two-hybrid assay, which has led to the discovery of a hitherto unexpected complexity of proteins which selectively interact with individual AMPA receptor subunits. These proteins have been implicated in the regulation of AMPA receptor post-translational modification, targeting and trafficking, surface expression and anchoring. The aim of this article is to present an overview of the major interacting proteins described so far and to place these in the context of how they may participate in the well ordered series of events controlling the cell biology of AMPA receptors.
Collapse
Affiliation(s)
- S P Braithwaite
- MRC Centre for Synaptic Plasticity, Department of Anatomy, School of Medical Science, University of Bristol, University Walk, Bristol, UK
| | | | | |
Collapse
|
38
|
Abstract
Movement, the fundamental component of behavior and the principal extrinsic action of the brain, is produced when skeletal muscles contract and relax in response to patterns of action potentials generated by motoneurons. The processes that determine the firing behavior of motoneurons are therefore important in understanding the transformation of neural activity to motor behavior. Here, we review recent studies on the control of motoneuronal excitability, focusing on synaptic and cellular properties. We first present a background description of motoneurons: their development, anatomical organization, and membrane properties, both passive and active. We then describe the general anatomical organization of synaptic input to motoneurons, followed by a description of the major transmitter systems that affect motoneuronal excitability, including ligands, receptor distribution, pre- and postsynaptic actions, signal transduction, and functional role. Glutamate is the main excitatory, and GABA and glycine are the main inhibitory transmitters acting through ionotropic receptors. These amino acids signal the principal motor commands from peripheral, spinal, and supraspinal structures. Amines, such as serotonin and norepinephrine, and neuropeptides, as well as the glutamate and GABA acting at metabotropic receptors, modulate motoneuronal excitability through pre- and postsynaptic actions. Acting principally via second messenger systems, their actions converge on common effectors, e.g., leak K(+) current, cationic inward current, hyperpolarization-activated inward current, Ca(2+) channels, or presynaptic release processes. Together, these numerous inputs mediate and modify incoming motor commands, ultimately generating the coordinated firing patterns that underlie muscle contractions during motor behavior.
Collapse
Affiliation(s)
- J C Rekling
- Department of Neurobiology, University of California, Los Angeles, California 90095-1763, USA
| | | | | | | | | |
Collapse
|
39
|
Tapia R, Peña F, Arias C. Neurotoxic and synaptic effects of okadaic acid, an inhibitor of protein phosphatases. Neurochem Res 1999; 24:1423-30. [PMID: 10555783 DOI: 10.1023/a:1022588808260] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Protein phosphorylation and dephosphorylation reactions, catalyzed by kinases and phosphatases, are involved in the regulation of a wide variety of physiological processes. In the nervous system, such reactions seem to modulate the function of several proteins crucial in synaptic transmission, including voltage-gated and ligand-gated channels, neurotransmitter release, and neurotransmitter transporters. On the other hand, hyperphosphorylation of certain cytoskeletal proteins or receptors may lead to neuronal death. In the present work we review the neurotoxic effect of okadaic acid (OKA), a potent and specific inhibitor of the serine/threonine protein phosphatases 1 and 2A, as well as its action on synaptic function. We analyze recent findings demonstrating that the microinjection of OKA in rat hippocampus induces neuronal stress, hyperexcitation and neurodegeneration, and discuss their possible relationships to alterations of protein phosphorylation-dephosphorylation observed in Alzheimer's disease brain. These results suggest that protein hyperphosphorylation due to inhibition of phosphatases in vivo induces neuronal stress and subsequent neurodegeneration.
Collapse
Affiliation(s)
- R Tapia
- Departamento de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, DF, México.
| | | | | |
Collapse
|
40
|
Faux MC, Rollins EN, Edwards AS, Langeberg LK, Newton AC, Scott JD. Mechanism of A-kinase-anchoring protein 79 (AKAP79) and protein kinase C interaction. Biochem J 1999; 343 Pt 2:443-52. [PMID: 10510312 PMCID: PMC1220573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
The A-kinase-anchoring protein AKAP79 co-ordinates the location of cAMP-dependent protein kinase, phosphatase 2B (PP2B/calcineurin) and protein kinase C (PKC) at postsynaptic sites in neurons. In this report we focus on the mechanism of interaction between AKAP79 and PKC. We show that neither lipid activators nor kinase activation are required for association with AKAP79. The anchoring protein binds and inhibits the conserved catalytic core of PKCbetaII. AKAP79 also associates with conventional, novel and atypical isoforms of PKC in vitro and in vivo, and immunofluorescence staining of rat hippocampal neurons demonstrates that the murine anchoring-protein homologue AKAP150 is co-distributed with PKCalpha/beta, PKCepsilon or PKCiota. Binding of the AKAP79(31-52) peptide, which inhibits kinase activity, exposes the pseudosubstrate domain of PKCbetaII, allowing endoproteinase Arg-C proteolysis in the absence of kinase activators. Reciprocal experiments have identified two arginine residues at positions 39 and 40 that are essential for AKAP79(31-52) peptide inhibition of PKCbetaII. Likewise, the same mutations in the full-length anchoring protein reduced inhibition of PKCbetaII. Thus AKAP79 associates with multiple PKC isoforms through a mechanism involving protein-protein interactions at the catalytic core where binding of the anchoring protein inhibits kinase activity through displacement of the pseudosubstrate.
Collapse
Affiliation(s)
- M C Faux
- Howard Hughes Medical Institute, Vollum Institute, L474, Oregon Health Sciences University, 3181 SW Sam Jackson Park Road, Portland, OR 97201, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Overactivation of ionotropic glutamate receptors has been implicated in the pathophysiology of traumatic brain injury. Using an in vitro cell injury model, we examined the effects of stretch-induced traumatic injury on the AMPA subtype of ionotropic glutamate receptors in cultured neonatal cortical neurons. Recordings made using the whole-cell patch-clamp technique revealed that a subpopulation of injured neurons exhibited an increased current in response to AMPA. The current-voltage relationship of these injured neurons showed an increased slope conductance but no change in reversal potential compared with uninjured neurons. Additionally, the EC(50) values of uninjured and injured neurons were nearly identical. Thus, current potentiation was not caused by changes in the voltage-dependence, ion selectivity, or apparent agonist affinity of the AMPA channel. AMPA-elicited current could also be fully inhibited by the application of selective AMPA receptor antagonists, thereby excluding the possibility that current potentiation in injured neurons was caused by the activation of other, nondesensitizing receptors. The difference in current densities between control and injured neurons was abolished when AMPA receptor desensitization was inhibited by the coapplication of AMPA and cyclothiazide or by the use of kainate as an agonist, suggesting that mechanical injury alters AMPA receptor desensitization. Reduction of AMPA receptor desensitization after brain injury would be expected to further exacerbate the effects of increased postinjury extracellular glutamate and contribute to trauma-related cell loss and dysfunctional synaptic information processing.
Collapse
|
42
|
Goforth PB, Ellis EF, Satin LS. Enhancement of AMPA-mediated current after traumatic injury in cortical neurons. J Neurosci 1999; 19:7367-74. [PMID: 10460243 PMCID: PMC6782494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/1999] [Revised: 06/21/1999] [Accepted: 06/23/1999] [Indexed: 02/13/2023] Open
Abstract
Overactivation of ionotropic glutamate receptors has been implicated in the pathophysiology of traumatic brain injury. Using an in vitro cell injury model, we examined the effects of stretch-induced traumatic injury on the AMPA subtype of ionotropic glutamate receptors in cultured neonatal cortical neurons. Recordings made using the whole-cell patch-clamp technique revealed that a subpopulation of injured neurons exhibited an increased current in response to AMPA. The current-voltage relationship of these injured neurons showed an increased slope conductance but no change in reversal potential compared with uninjured neurons. Additionally, the EC(50) values of uninjured and injured neurons were nearly identical. Thus, current potentiation was not caused by changes in the voltage-dependence, ion selectivity, or apparent agonist affinity of the AMPA channel. AMPA-elicited current could also be fully inhibited by the application of selective AMPA receptor antagonists, thereby excluding the possibility that current potentiation in injured neurons was caused by the activation of other, nondesensitizing receptors. The difference in current densities between control and injured neurons was abolished when AMPA receptor desensitization was inhibited by the coapplication of AMPA and cyclothiazide or by the use of kainate as an agonist, suggesting that mechanical injury alters AMPA receptor desensitization. Reduction of AMPA receptor desensitization after brain injury would be expected to further exacerbate the effects of increased postinjury extracellular glutamate and contribute to trauma-related cell loss and dysfunctional synaptic information processing.
Collapse
Affiliation(s)
- P B Goforth
- Departments of Pharmacology/Toxicology and Physiology, Medical College of Virginia, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | |
Collapse
|
43
|
Swope SL, Moss SJ, Raymond LA, Huganir RL. Regulation of ligand-gated ion channels by protein phosphorylation. ADVANCES IN SECOND MESSENGER AND PHOSPHOPROTEIN RESEARCH 1999; 33:49-78. [PMID: 10218114 DOI: 10.1016/s1040-7952(99)80005-6] [Citation(s) in RCA: 254] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The studies discussed in this review demonstrate that phosphorylation is an important mechanism for the regulation of ligand-gated ion channels. Structurally, ligand-gated ion channels are heteromeric proteins comprised of homologous subunits. For both the AChR and the GABA(A) receptor, each subunit has a large extracellular N-terminal domain, four transmembrane domains, a large intracellular loop between transmembrane domains M3 and M4, and an extracellular C-terminal domain (Fig. 1B). All the phosphorylation sites on these receptors have been mapped to the major intracellular loop between M3 and M4 (Table 1). In contrast, glutamate receptors appear to have a very large extracellular N-terminal domain, one membrane hairpin loop, three transmembrane domains, a large extracellular loop between transmembrane domains M3 and M4, and an intracellular C-terminal domain (Fig. 1C). Most phosphorylation sites on glutamate receptors have been shown to be on the intracellular C-terminal domain, although some have been suggested to be on the putative extracellular loop between M3 and M4 (Table 1). A variety of extracellular factors and intracellular signal transduction cascades are involved in regulating phosphorylation of these ligand-gated ion channels (Fig. 2). Once again, the AChR at the neuromuscular junction is the most fully understood system. Phosphorylation of the AChR by PKA is stimulated synaptically by the neuropeptide CGRP and in an autocrine fashion by adenosine released from the muscle in response to acetylcholine. In addition, acetylcholine, via calcium influx through the AChR, appears to activate calcium-dependent kinases including PKC to stimulate serine phosphorylation of the receptor. Presently, agrin is the only extracellular factor known to stimulate phosphorylation of the AChR on tyrosine residues. For glutamate receptors, non-NMDA receptor phosphorylation by PKA is stimulated by dopamine, while NMDA receptor phosphorylation by PKA and PKC can be induced via the activation of beta-adrenergic receptors, and metabotropic glutamate or opioid receptors, respectively. In addition, Ca2+ influx through the NMDA receptor has been shown to activate PKC. CaMKII, and calcineurin, resulting in phosphorylation of AMPA receptors (by CaMKII) and inactivation of NMDA receptors (at least in part through calcineurin). In contrast to the AChR and glutamate receptors, no information is presently available regarding the identities of the extracellular factors and intracellular signal transduction cascades that regulate phosphorylation of the GABA(A) receptor. Surely, future studies will be aimed at further clarifying the molecular mechanisms by which the central receptors are regulated. The presently understood functional effects of ligand-gated ion channel phosphorylation are diverse. At the neuromuscular junction, a regulation of the AChR desensitization rate by both serine and tyrosine phosphorylation has been demonstrated. In addition, tyrosine phosphorylation of the AChR or other synaptic components appears to play a role in AChR clustering during synaptogenesis. For the GABA(A) receptor, the data are complex. Both activation and inhibition of GABA(A) receptor currents as a result of PKA and PKC phosphorylation have been reported, while phosphorylation by PTK enhances function. The predominant effect of glutamate receptor phosphorylation by a variety of kinases is a potentiation of the peak current response. However, PKC also modulates clustering of NMDA receptors. This complexity in the regulation of ligand-gated ion channels by phosphorylation provides diverse mechanisms for mediating synaptic plasticity. In fact, accumulating evidence supports the involvement of protein phosphorylation and dephosphorylation of AMPA receptors in LTP and LTD respectively. There has been a dramatic increase in our understanding of the nature by which phosphorylation regulates ligand-gated ion channels. However, many questions remain unanswered. (AB
Collapse
Affiliation(s)
- S L Swope
- Department of Neurology, Georgetown Institute for Cognitive and Computational Neuroscience, Georgetown University Medical Center, Washington, DC 20007, USA
| | | | | | | |
Collapse
|
44
|
Abstract
Several protein kinases are known to phosphorylate Ser/Thr residues of certain GABAA receptor subunits. Yet, the effect of phosphorylation on GABAA receptor function in neurons remains controversial, and the functional consequences of phosphorylating synaptic GABAA receptors of adult CNS neurons are poorly understood. We used whole-cell patch-clamp recordings of GABAA receptor-mediated miniature IPSCs (mIPSCs) in CA1 pyramidal neurons and dentate gyrus granule cells (GCs) of adult rat hippocampal slices to determine the effects of cAMP-dependent protein kinase (PKA) and Ca2+/phospholipid-dependent protein kinase (PKC) activation on the function of synaptic GABAA receptors. The mIPSCs recorded in CA1 pyramidal cells and in GCs were differentially affected by PKA and PKC. In pyramidal cells, PKA reduced mIPSC amplitudes and enhanced the fraction of events decaying with a double exponential, whereas PKC was without effect. In contrast, in GCs PKA was ineffective, but PKC increased the peak amplitude of mIPSCs and also favored double exponential decays. Intracellular perfusion of the phosphatase inhibitor microcystin revealed that synaptic GABAA receptors of pyramidal cells, but not those of GCs, are continually phosphorylated by PKA and conversely, dephosphorylated, most likely by phosphatase 1 or 2A. This differential, brain region-specific phosphorylation of GABAA receptors may produce a wide dynamic range of inhibitory synaptic strength in these two regions of the hippocampal formation.
Collapse
|
45
|
MacDonald JF, Xiong XG, Lu WY, Raouf R, Orser BA. Modulation of NMDA receptors. PROGRESS IN BRAIN RESEARCH 1999; 116:191-208. [PMID: 9932378 DOI: 10.1016/s0079-6123(08)60438-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Affiliation(s)
- J F MacDonald
- Department of Physiology, University of Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
46
|
Capogna M, Fankhauser C, Gagliardini V, Gähwiler BH, Thompson SM. Excitatory synaptic transmission and its modulation by PKC is unchanged in the hippocampus of GAP-43-deficient mice. Eur J Neurosci 1999; 11:433-40. [PMID: 10051744 DOI: 10.1046/j.1460-9568.1999.00450.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We compared excitatory synaptic transmission between hippocampal pyramidal cells in dissociated hippocampal cell cultures and in area CA3 of hippocampal slice cultures derived from wild-type mice and mice with a genetic deletion of the presynaptic growth associated protein GAP-43. The basal frequency and amplitude of action potential-dependent and -independent spontaneous excitatory postsynaptic currents were similar in both groups. The probability that any two CA3 pyramidal cells in wild-type or GAP-43 knockout (-/-) slice cultures were synaptically connected was assessed with paired recordings and was not different. Furthermore, unitary synaptic responses were similar in the two genotypes. Bath application of phorbol 12,13-diacetate (0.6-3 microM) elicited a comparable increase in the frequency of miniature excitatory synaptic currents in wild-type and GAP-43 (-/-) cultures. This effect was blocked by the protein kinase C inhibitor, bisindolylmaleimide I (1.2 microM). Finally, 3 microM phorbol 12,13-diacetate potentiated the amplitude of unitary synaptic currents to a comparable extent in wild-type and GAP-43 (-/-) slice cultures. We conclude that GAP-43 is not required for normal excitatory synaptic transmission or the potentiation of presynaptic glutamate release mediated by activation of protein kinase C in the hippocampus.
Collapse
Affiliation(s)
- M Capogna
- Brain Research Institute, University of Zurich, Switzerland
| | | | | | | | | |
Collapse
|
47
|
Poisbeau P, Cheney MC, Browning MD, Mody I. Modulation of synaptic GABAA receptor function by PKA and PKC in adult hippocampal neurons. J Neurosci 1999; 19:674-83. [PMID: 9880588 PMCID: PMC6782188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Several protein kinases are known to phosphorylate Ser/Thr residues of certain GABAA receptor subunits. Yet, the effect of phosphorylation on GABAA receptor function in neurons remains controversial, and the functional consequences of phosphorylating synaptic GABAA receptors of adult CNS neurons are poorly understood. We used whole-cell patch-clamp recordings of GABAA receptor-mediated miniature IPSCs (mIPSCs) in CA1 pyramidal neurons and dentate gyrus granule cells (GCs) of adult rat hippocampal slices to determine the effects of cAMP-dependent protein kinase (PKA) and Ca2+/phospholipid-dependent protein kinase (PKC) activation on the function of synaptic GABAA receptors. The mIPSCs recorded in CA1 pyramidal cells and in GCs were differentially affected by PKA and PKC. In pyramidal cells, PKA reduced mIPSC amplitudes and enhanced the fraction of events decaying with a double exponential, whereas PKC was without effect. In contrast, in GCs PKA was ineffective, but PKC increased the peak amplitude of mIPSCs and also favored double exponential decays. Intracellular perfusion of the phosphatase inhibitor microcystin revealed that synaptic GABAA receptors of pyramidal cells, but not those of GCs, are continually phosphorylated by PKA and conversely, dephosphorylated, most likely by phosphatase 1 or 2A. This differential, brain region-specific phosphorylation of GABAA receptors may produce a wide dynamic range of inhibitory synaptic strength in these two regions of the hippocampal formation.
Collapse
Affiliation(s)
- P Poisbeau
- Laboratoire de Neurophysiologie Cellulaire et Intégrée, Centre National de la Recherche Scientifique UMR 7519, Université Louis Pasteur, 67084 Strasbourg, France
| | | | | | | |
Collapse
|
48
|
D'Angelo E, Rossi P, Armano S, Taglietti V. Evidence for NMDA and mGlu receptor-dependent long-term potentiation of mossy fiber-granule cell transmission in rat cerebellum. J Neurophysiol 1999; 81:277-87. [PMID: 9914288 DOI: 10.1152/jn.1999.81.1.277] [Citation(s) in RCA: 184] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Long-term potentiation (LTP) is a form of synaptic plasticity that can be revealed at numerous hippocampal and neocortical synapses following high-frequency activation of N-methyl--aspartate (NMDA) receptors. However, it was not known whether LTP could be induced at the mossy fiber-granule cell relay of cerebellum. This is a particularly interesting issue because theories of the cerebellum do not consider or even explicitly negate the existence of mossy fiber-granule cell synaptic plasticity. Here we show that high-frequency mossy fiber stimulation paired with granule cell membrane depolarization (-40 mV) leads to LTP of granule cell excitatory postsynaptic currents (EPSCs). Pairing with a relatively hyperpolarized potential (-60 mV) or in the presence of NMDA receptor blockers [5-amino--phosphonovaleric acid (APV) and 7-chloro-kynurenic acid (7-Cl-Kyn)] prevented LTP, suggesting that the induction process involves a voltage-dependent NMDA receptor activation. Metabotropic glutamate receptors were also involved because blocking them with (+)-alpha-methyl-4-carboxyphenyl-glycine (MCPG) prevented potentiation. At the cytoplasmic level, EPSC potentiation required a Ca2+ increase and protein kinase C (PKC) activation. Potentiation was expressed through an increase in both the NMDA and non-NMDA receptor-mediated current and by an NMDA current slowdown, suggesting that complex mechanisms control synaptic efficacy during LTP. LTP at the mossy fiber-granule cell synapse provides the cerebellar network with a large reservoir for memory storage, which may be needed to optimize pattern recognition and, ultimately, cerebellar learning and computation.
Collapse
Affiliation(s)
- E D'Angelo
- Institute of General Physiology and Istituto Nazionale Fisica della Materia, Pavia Unit, I-27100 Pavia, Italy
| | | | | | | |
Collapse
|
49
|
Xiong ZG, Raouf R, Lu WY, Wang LY, Orser BA, Dudek EM, Browning MD, MacDonald JF. Regulation of N-Methyl-d-Aspartate Receptor Function by Constitutively Active Protein Kinase C. Mol Pharmacol 1998. [DOI: 10.1124/mol.54.6.1055] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
50
|
Oehrlein SA, Maelicke A, Herget T. Expression of protein kinase C gene family members is temporally and spatially regulated during neural development in vitro. Eur J Cell Biol 1998; 77:323-37. [PMID: 9930657 DOI: 10.1016/s0171-9335(98)80091-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We used primary cultures of rat hippocampal neurons and PCC7-Mz1 cells to correlate the expression of the protein kinase C (PKC) gene family with specific events during neural differentiation. Multipotent PCC7-Mz1 embryonic carcinoma stem cells develop into a tissue-like pattern of neuronal, fibroblast-like and astroglial cells by all-trans retinoic acid (RA) treatment. Western blot analyses demonstrate that PKCalpha, betaI, gamma, theta, mu, lambda, and zeta were constitutively expressed but the expression of PKCbetaII, delta, epsilon, and eta was up-regulated three days after addition of RA when cells mature morphologically. While the protein levels of the PKC isoforms betaII, delta and eta decreased after d6, when the major phenotypical alterations of the developing neurons were completed, PKCepsilon expression remained at a high level. Immunofluorescence studies demonstrated that PKCalpha, lambda and zeta were constantly expressed in stem cells and the arising cell types. PKCdelta was detected in all differentiated cell types, whereby PKCbetaII, gamma, epsilon, and zeta were solely found in the neuronal derivatives with PKCgamma predominantly located in the nuclei. PKCeta was weakly expressed at the Golgi complex of stem cells but expanded throughout the entire somata of all developing neurons. In contrast, PKCbetaII was abundant only in the somata of a minor fraction of all neurons (approximately 2.5%). Also, PKCepsilon was exclusively synthesized by a subpopulation of neurons (40+/-5%), where it was localized in the somata and in the axons. PKCzeta was persistently expressed in two forms, the full-length PKCzeta and the constitutively active, proteolytic product PKMzeta, reasoning that permanent PKCzeta activity is important for PCC7-Mz1 physiology. Fractionation of extracts from undifferentiated and differentiating PCC7-Mz1 cells revealed that the conventional cPKCalpha was partly and the cPKCbetaI and the novel nPKCs delta and epsilon were mainly membrane bound, implying that they were also in an active state. However, when using the PKC substrate MARCKS (myristoylated alanine-rich C kinase substrate) to monitor cellular PKC activity, we observed that activation of PKC by phorbol ester was required for complete MARCKS phosphorylation and its translocation from the membrane to the cytoplasm. Our data show that the cell type-specific expression, subcellular localization and activation of PKCs are regulated in an isoform-specific manner during neurogenesis suggesting that they are involved in the control of neural development and in particular in neuronal differentiation.
Collapse
Affiliation(s)
- S A Oehrlein
- Johannes Gutenberg-University, Laboratory of Molecular Neurobiology, Institute of Physiological Chemistry and Pathobiochemistry, Mainz, Germany
| | | | | |
Collapse
|