Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS One ; 9(3): e91288, 2014.
Article in English | MEDLINE | ID: mdl-24608178

ABSTRACT

Glaucoma is a progressive neurodegenerative disease of retinal ganglion cells (RGCs) associated with characteristic axon degeneration in the optic nerve. Excitotoxic damage due to increased Ca(2+) influx, possibly through NMDA-type glutamate receptors, has been proposed to be a cause of RGC dysfunction and death in glaucoma. Recent work has found that expression of another potentially critical receptor, the Ca(2+)-permeable AMPA receptor (CP-AMPAR), is elevated during various pathological conditions (including ALS and ischemia), resulting in increased neuronal death. Here we test the hypothesis that CP-AMPARs contribute to RGC death due to elevated Ca(2+) influx in glaucoma. AMPA receptors are impermeable to Ca(2+) if the tetrameric receptor contains a GluA2 subunit that has undergone Q/R RNA editing at a site in the pore region. The activity of ADAR2, the enzyme responsible for this RNA editing, generally ensures that the vast majority of GluA2 proteins are edited. Here, we demonstrate that ADAR2 levels decrease in a mouse model of glaucoma in which IOP is chronically elevated. Furthermore, using an in vitro model of RGCs, we find that knockdown of ADAR2 using siRNA increased the accumulation of Co(2+) in response to glutamate, and decreased the rectification index of AMPA currents detected electrophysiologically, indicating an increased Ca(2+) permeability through AMPARs. The RGCs in primary culture also exhibited increased excitotoxic cell death following knock down of ADAR2. Furthermore, cell death was reversed by NASPM, a specific blocker for CP-AMPARs. Together, our data suggest that chronically elevated IOP in adult mice reduces expression of the ADAR2 enzyme, and the loss of ADAR2 editing and subsequent disruption of GluA2 RNA editing might potentially play a role in promoting RGC neuronal death as observed in glaucoma.


Subject(s)
Adenosine Deaminase/metabolism , Down-Regulation , Glaucoma/enzymology , Glaucoma/pathology , RNA Editing/genetics , RNA-Binding Proteins/metabolism , Retinal Ganglion Cells/enzymology , Retinal Ganglion Cells/pathology , Animals , Cell Death , Cells, Cultured , Cobalt/metabolism , Disease Models, Animal , Female , Gene Knockdown Techniques , Glaucoma/physiopathology , In Situ Nick-End Labeling , Intraocular Pressure , Mice, Inbred C57BL , RNA, Small Interfering/metabolism , Receptors, AMPA/metabolism , Staining and Labeling
2.
J Neurosci ; 34(2): 656-61, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24403163

ABSTRACT

Retinal ganglion cells (RGCs) receive glutamatergic input from bipolar cells through NMDA- and AMPA-type glutamate receptors. Both GluA2-containing, Ca(2+)-impermeable AMPA receptors (CI-AMPARs) and GluA2-lacking, Ca(2+)-permeable AMPA receptors (CP-AMPARs) contribute to light-evoked responses in ON RGCs; however, specific roles for each subtype are not well understood. Here, we present evidence that light intensity determines the subtype of AMPAR that is activated during the synaptic response in ON RGCs. Using current voltage analysis of the EPSC we show that light intensities near RGC threshold, intensities that travel through the well described primary rod pathway, evoke synaptic currents that are preferentially mediated by CP-AMPARs. Synaptic responses evoked by spontaneous release of transmitter from bipolar cell terminals also preferentially activate CP-AMPARs. Conversely, higher light intensities, most likely carried by secondary rod pathways, activate CI-AMPARs. The same pattern of CP-AMPAR and CI-AMPAR activation was observed in mice containing only functional rods, suggesting that the recruitment of CI-AMPARs at higher light intensity does not require cone stimulation. When glutamate spillover was induced by blocking transporters with TBOA, both the near threshold and spontaneous EPSCs contained a significant CI-AMPAR component. We propose that CI-AMPARs are activated by "spillover" of synaptic glutamate only during bright illumination, or when glutamate uptake is blocked. Glutamate may spill over to more distant sites at the same synapse, or perhaps as far as neighboring synapses. Together, our data suggest that the spatial organization of AMPARs at ON RGCs synapses allows for selective, intensity-dependent activation of AMPARs with distinct subunit composition.


Subject(s)
Receptors, AMPA/metabolism , Retinal Ganglion Cells/metabolism , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Photic Stimulation
3.
Mol Cell Neurosci ; 56: 384-92, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23911793

ABSTRACT

On retinal ganglion cells (RGCs) transmit light encoded information to the brain and receive excitatory input from On cone bipolar cells (CBPs). The synaptic CBP input onto On RGCs is mediated by AMPA-type glutamate receptors (AMPARs) that include both those lacking a GluA2 subunit, and are therefore permeable to Ca(2+), and those that possess at least one GluA2 subunit and are Ca(2+)-impermeable. We have previously demonstrated in electrophysiological studies that periods of low synaptic activity, brought about by housing animals in darkness, enhance the proportion of GluA2-lacking AMPARs at the On CBP-On RGC synapse by mobilizing surface GluA2 containing receptors into a receptor pool that rapidly cycles in and out of the membrane. AMPAR cycling induction by reduced synaptic activity takes several hours. This delay suggests that changes in expression of proteins which regulate AMPAR trafficking may mediate the altered mobility of GluA2 AMPARs in RGCs. In this study, we test the hypothesis that AMPAR trafficking proteins couple synaptic activity to AMPAR cycling in RGCs. Immunocytochemical and biochemical analyses confirmed that darkness decreases surface GluA2 in RGCs and changed the expression levels of three proteins associated with GluA2 trafficking. GRIP was decreased, while PICK1 and Arc were increased. Knockdown of GRIP with siRNA elevated constitutive AMPAR cycling, mimicking effects of reduced synaptic activity, while knockdown of PICK1 and Arc blocked increases in constitutive GluA2 trafficking. Our results support a role for correlated, activity-driven changes in multiple AMPAR trafficking proteins that modulate GluA2 cycling which can in turn affect synaptic AMPAR composition in RGCs.


Subject(s)
Receptors, AMPA/metabolism , Retinal Ganglion Cells/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, AMPA/genetics , Retinal Ganglion Cells/physiology , Synaptic Membranes/metabolism , Synaptic Transmission
4.
Neuron ; 75(3): 467-78, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22884330

ABSTRACT

Light-evoked responses of all three major classes of retinal ganglion cells (RGCs) are mediated by NMDA receptors (NMDARs) and AMPA receptors (AMPARs). Although synaptic activity at RGC synapses is highly dynamic, synaptic plasticity has not been observed in adult RGCs. Here, using patch-clamp recordings in dark-adapted mouse retina, we report a retina-specific form of AMPAR plasticity. Both chemical and light activation of NMDARs caused the selective endocytosis of GluA2-containing, Ca(2+)-impermeable AMPARs on RGCs and replacement with GluA2-lacking, Ca(2+)-permeable AMPARs. The plasticity was expressed in ON but not OFF RGCs and was restricted solely to the ON responses in ON-OFF RGCs. Finally, the plasticity resulted in a shift in the light responsiveness of ON RGCs. Thus, physiologically relevant light stimuli can induce a change in synaptic receptor composition of ON RGCs, providing a mechanism by which the sensitivity of RGC responses may be modified under scotopic conditions.


Subject(s)
Light , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Retinal Ganglion Cells/metabolism , Synapses/metabolism , Animals , Excitatory Postsynaptic Potentials , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Synaptic Transmission/physiology
5.
Proc Natl Acad Sci U S A ; 109(22): 8752-7, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22586107

ABSTRACT

ON bipolar cells are critical for the function of the ON pathway in the visual system. They express a metabotropic glutamate receptor (mGluR6) that, when activated, couples to the G(o) class of G protein. The channel that is primarily responsible for the synaptic response has been recently identified as the transient receptor potential cation channel subfamily M member 1 (TRPM1); TRPM1 is negatively coupled to the mGluR6/Go cascade such that activation of the cascade results in closure of the channel. Light indirectly opens TRPM1 by reducing transmitter release from presynaptic photoreceptors, resulting in a decrease in mGluR6 activation. Conversely, in the dark, binding of synaptic glutamate to mGluR6 inhibits TRPM1 current. Closure of TRPM1 by G-protein activation in the dark is a critical step in the process of ON bipolar cell signal transduction, but the precise pathway linking these two events is not understood. To address this question, we measured TRPM1 activity in retinal bipolar cells, in human ependymal melanocytes (HEMs) that endogenously express TRPM1, and in HEK293 cells transfected with TRPM1. Dialysis of the Gßγ subunit dimer, but not Gα(o), closed TRPM1 channels in every cell type that we tested. In addition, activation of an endogenous G-protein-coupled receptor pathway in HEK293 cells that releases Gßγ without activating Go protein also closed TRPM1 channels. These results suggest a model in which the Gßγ dimer that is released as a result of the dissociation from Gα(o) upon activation of mGluR6 closes the TRPM1 channel, perhaps via a direct interaction.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , TRPM Cation Channels/physiology , Amino Acids/pharmacology , Animals , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein gamma Subunits/chemistry , Glutamic Acid/metabolism , HEK293 Cells , Humans , Light , Membrane Potentials/drug effects , Membrane Potentials/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/drug effects , Protein Binding/radiation effects , Protein Multimerization , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/physiology , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , Xanthenes/pharmacology
6.
Mol Cell Neurosci ; 48(2): 161-70, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807099

ABSTRACT

Activation of metabotropic- (mGluRs) or NMDA-type glutamate receptors (NMDARs) each can induce long-term depression (LTD) of synaptic transmission in CA1 hippocampal neurons. These two forms of LTD are triggered by diverse signaling pathways yet both are expressed by the internalization of AMPA-type glutamate receptors (AMPARs). An unanswered question remains as to whether the convergence of the mGluR and NMDAR signaling pathways on AMPAR endocytosis renders these two forms of plasticity functionally equivalent, with both pathways inducing endocytosis of the same population of synaptic AMPARs. We now report evidence that these pathways couple to the endocytosis of distinct populations of AMPARs defined by their mobility in the membrane surface. NMDAR activation enhances removal of surface AMPARs that rapidly cycle into and out of the membrane surface, while activation of mGluRs with DHPG results in the internalization of a non-mobile population of AMPARs. Glutamate Receptor Interacting Proteins 1 and 2 (GRIP1/2) play a key role in defining the non-cycling receptor population. GRIP1/2 knockdown with siRNA increases the proportion of rapidly cycling surface AMPARs and inhibits mGluR- but not NMDAR-mediated AMPAR internalization. Additionally, we find that mGluR activation dissociates surface AMPARs from GRIP1/2 while stimulation of NMDARs elicits the loss of membrane receptors not bound to GRIP1/2. We propose that these two receptor pathways can drive the endocytosis of distinct populations of AMPARs: NMDARs activation induces the endocytosis of rapidly cycling surface AMPARs not directly associated with GRIP1/2 while mGluR activation induces the endocytosis of non-cycling GRIP-bound surface AMPARs.


Subject(s)
Endocytosis/physiology , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Hippocampus/cytology , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Long-Term Synaptic Depression/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology
7.
Curr Opin Neurobiol ; 21(2): 328-38, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21334194

ABSTRACT

Experience-dependent modifications of neural circuits and function are believed to heavily depend on changes in synaptic efficacy such as LTP/LTD. Hence, much effort has been devoted to elucidating the mechanisms underlying these forms of synaptic plasticity. Although most of this work has focused on excitatory synapses, it is now clear that diverse mechanisms of long-term inhibitory plasticity have evolved to provide additional flexibility to neural circuits. By changing the excitatory/inhibitory balance, GABAergic plasticity can regulate excitability, neural circuit function and ultimately, contribute to learning and memory, and neural circuit refinement. Here we discuss recent advancements in our understanding of the mechanisms and functional relevance of GABAergic inhibitory synaptic plasticity.


Subject(s)
Neuronal Plasticity/physiology , Synapses/physiology , Animals , Humans
8.
Proc Natl Acad Sci U S A ; 107(47): 20559-64, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059908

ABSTRACT

Ca(2+)/calmodulin protein kinase IIα (CaMKIIα) has a central role in regulating neuronal excitability. It is well established that CaMKIIα translocates to excitatory synapses following strong glutamatergic stimuli that induce NMDA-receptor (NMDAR)-dependent long-term potentiation in CA1 hippocampal neurons. We now show that CaMKIIα translocates to inhibitory but not excitatory synapses in response to more moderate NMDAR-activating stimuli that trigger GABA(A)-receptor (GABA(A)R) insertion and enhance inhibitory transmission. Such moderate NMDAR activation causes Thr286 autophosphorylation of CaMKIIα, which our results demonstrate is necessary and sufficient, under basal conditions, to localize CaMKIIα at inhibitory synapses and enhance surface GABA(A)R expression. Although stronger glutamatergic stimulation coupled to AMPA receptor insertion also elicits Thr286 autophosphorylation, accumulation of CaMKIIα at inhibitory synapses is prevented under these conditions by the phosphatase calcineurin. This preferential targeting of CaMKIIα to glutamatergic or GABAergic synapses provides neurons with a mechanism whereby activity can selectively potentiate excitation or inhibition through a single kinase mediator.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Long-Term Potentiation/physiology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Calcineurin/metabolism , Hippocampus/cytology , Image Processing, Computer-Assisted , Immunohistochemistry , Microscopy, Fluorescence , Rats , Receptors, GABA-A/metabolism
9.
Neuron ; 59(1): 1-3, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18614022

ABSTRACT

Neuroscientists have been looking for good examples linking neuronal activity to gene expression/regulation involved in synaptic plasticity and the formation of long-term memories. New findings from Park et al. and Waung et al. in this issue of Neuron show that fast dendritic translation of the immediate-early gene Arc/Arg3.1 is involved in hippocampal mGluR-LTD, a protein synthesis-dependent form of plasticity.


Subject(s)
Neuronal Plasticity/physiology , Neurons/metabolism , Protein Biosynthesis/physiology , Animals , Gene Expression Regulation/physiology , Hippocampus/cytology
10.
Neuron ; 58(1): 104-17, 2008 Apr 10.
Article in English | MEDLINE | ID: mdl-18400167

ABSTRACT

While most studies have focused on the role of long-term potentiation in behavior, far less is known about the role of long-term depression (LTD). To examine the potential involvement of LTD in learning and memory, we generated transgenic mice that express a fragment of the SV40 small t antigen known to inhibit protein phosphatase 2A (PP2A). Small t antigen expression blocked both stimulus-induced and chemically induced NMDAR-dependent LTD at Schaffer collateral synapses but did not affect potentiation, depotentiation, or mGluR-dependent LTD. This physiological phenotype was associated with deficits in behavioral flexibility in both the Morris water maze and a delayed nonmatch to place T-maze task, suggesting that NMDAR-dependent LTD is required for behavioral flexibility and may act by weakening previously encoded memory traces when new information is learned.


Subject(s)
Long-Term Synaptic Depression/genetics , Maze Learning/physiology , Receptors, N-Methyl-D-Aspartate/deficiency , Receptors, N-Methyl-D-Aspartate/genetics , Adaptation, Psychological/physiology , Animals , Behavior, Animal/physiology , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Receptors, N-Methyl-D-Aspartate/physiology
11.
J Neurosci ; 27(52): 14326-37, 2007 Dec 26.
Article in English | MEDLINE | ID: mdl-18160640

ABSTRACT

The trafficking of postsynaptic AMPA receptors (AMPARs) is a powerful mechanism for regulating the strength of excitatory synapses. It has become clear that the surface levels of inhibitory GABA(A) receptors (GABA(A)Rs) are also subject to regulation and that GABA(A)R trafficking may contribute to inhibitory plasticity, although the underlying mechanisms are not fully understood. Here, we report that NMDA receptor activation, which has been shown to drive excitatory long-term depression through AMPAR endocytosis, simultaneously increases expression of GABA(A)Rs at the dendritic surface of hippocampal neurons. This NMDA stimulus increases miniature IPSC amplitudes and requires the activity of Ca2+ calmodulin-dependent kinase II and the trafficking proteins N-ethylmaleimide-sensitive factor, GABA receptor-associated protein (GABARAP), and glutamate receptor interacting protein (GRIP). These data demonstrate for the first time that endogenous GABARAP and GRIP contribute to the regulated trafficking of GABA(A)Rs. In addition, they reveal that the bidirectional trafficking of AMPA and GABA(A) receptors can be driven by a single glutamatergic stimulus, providing a potent postsynaptic mechanism for modulating neuronal excitability.


Subject(s)
Exocytosis/physiology , Glutamate Decarboxylase/metabolism , Inhibitory Postsynaptic Potentials/physiology , Microtubule-Associated Proteins/metabolism , Neurons/physiology , Receptors, GABA/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Animals, Newborn , Biotinylation/methods , Cells, Cultured , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Exocytosis/drug effects , Hippocampus/cytology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/radiation effects , N-Methylaspartate/pharmacology , Neurons/drug effects , Patch-Clamp Techniques/methods , Protein Transport/drug effects , Protein Transport/physiology , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
12.
J Neurosci ; 27(48): 13273-8, 2007 Nov 28.
Article in English | MEDLINE | ID: mdl-18045921

ABSTRACT

The mGluR-dependent endocytosis of AMPA receptors (AMPARs) in the CA1 region is protein synthesis dependent. However, why this form of trafficking, and not that mediated by NMDA receptor activation, is dependent on protein translation is unclear. Here we have studied the contribution of the cytoskeletal microtubule-associated protein 1B (MAP1B) to the pathway-specific internalization of AMPARs. Treatments of cultured neurons with 3,4-dihydroxyphenylglycol (DHPG) or NMDA, both of which drive AMPAR endocytosis, caused a translation-dependent increase in the dendritic levels of MAP1B protein. Although interfering with protein synthesis using short interfering RNA (siRNA) to eEF2 kinase (eukaryotic elongation factor 2 kinase) blocked the dendritic MAP1B increase by both pathways, it selectively blocked the DHPG- and not the NMDA-induced AMPAR endocytosis. In support of MAP1B synthesis contributing to metabotropic glutamate receptor (mGluR)-mediated AMPAR endocytosis, siRNA against MAP1B in CA1 cultured neurons specifically blocked the DHPG-induced AMPAR internalization. Previous studies suggest a direct interaction between MAP1B and the AMPAR-binding protein GRIP1. Biochemical studies establish that MAP1B associates with GRIP1 and forms a complex with GluR2 in vivo in rat hippocampus. Furthermore, the interaction between MAP1B and GRIP1 increased significantly in acute slices after treatment with DHPG and not NMDA. Together, these findings suggest that MAP1B plays a selective role in the DHPG-induced endocytosis of AMPARs, perhaps through its interaction with GRIP1.


Subject(s)
Endocytosis/physiology , Hippocampus/physiology , Microtubule-Associated Proteins/metabolism , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Animals, Newborn , Biotinylation/methods , Cells, Cultured , Drug Interactions , Elongation Factor 2 Kinase/genetics , Endocytosis/drug effects , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation/drug effects , Hippocampus/cytology , Immunoprecipitation/methods , In Vitro Techniques , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , N-Methylaspartate/pharmacology , Neurons/drug effects , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley
13.
J Neurosci ; 27(45): 12221-9, 2007 Nov 07.
Article in English | MEDLINE | ID: mdl-17989288

ABSTRACT

At many excitatory synapses, AMPA-type receptors (AMPARs) are not statically situated in the membrane, but undergo continuous rounds of endocytosis and exocytosis, referred to as rapid cycling. AMPAR cycling is believed to play a role in certain forms of synaptic plasticity, but the link between cycling and synaptic function is not well understood. We have previously demonstrated that AMPARs cycle in neurons of the inner retina, including amacrine and ganglion cells, and that cycling is inhibited by synaptic activity. Recording from cultured neurons and ON ganglion cells in the flat-mount retina, we now show that rapid cycling is primarily, perhaps exclusively, restricted to AMPARs that contain the GluR2 subunit, and that cycle is confined to extrasynaptic receptors. We also demonstrate a form of plasticity at the ON bipolar cell-ON ganglion cell synapse, whereby synaptic quiescence drives a change in the composition of AMPARs from predominantly GluR2-containing to GluR2-lacking. Finally, we provide evidence linking synaptic receptor composition and cycling, showing that disruption of cycling leads increases the number of GluR2-containing receptors in the ON bipolar-ON ganglion cell synapse. We propose that cycling lowers the number of GluR2-containing receptors at the surface and, consequently, within the synapse. After increased levels of synaptic activity, cycling ceases, and all GluR2-containing receptors are free to go to the surface, where they can be delivered to synapses. Our results suggest that by regulating the cycling of AMPARs, ambient light can modulate the composition of synaptic receptors in ON ganglion cells.


Subject(s)
Neuronal Plasticity/physiology , Retinal Ganglion Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Exocytosis/physiology , Mice , Mice, Inbred C57BL , Rats , Receptors, AMPA/physiology
14.
Neuropharmacology ; 53(1): 92-100, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17555774

ABSTRACT

The activation of NMDA receptors (NMDARs) triggers long-term changes in AMPA receptor-mediated synaptic transmission in the CNS. These long-lasting changes occur via the addition or removal of AMPA receptors (AMPARs) at the synaptic membrane and are mediated by a number of regulatory proteins including the GluR2 AMPAR-interacting proteins n-ethylmaleimide sensitive factor (NSF) and Protein Interacting with C Kinase (PICK1). We have shown that the potent activation of NMDARs drives unclustering of PICK1 and PICK1-GluR2 dissociation in dendrites resulting in increased surface delivery of AMPARs. Here we show that the dispersal of PICK1 is mediated by the actions of NSF. We find that elevated NMDAR signaling leads to the S-nitrosylation of NSF and increased NSF-GluR2 association. Both NMDAR-dependent unclustering of PICK1 and the delivery of surface AMPARs are dependent on release of nitric oxide (NO). Our data suggest that NMDAR activation can drive the surface delivery of AMPARs from a pool of intracellular AMPARs retained by PICK1 through the NO-dependent modification of NSF.


Subject(s)
Carrier Proteins/metabolism , Exocytosis/physiology , Nitric Oxide/physiology , Nuclear Proteins/metabolism , Receptors, AMPA/metabolism , Animals , Animals, Newborn , Carrier Proteins/genetics , Cells, Cultured , Cyclic N-Oxides/pharmacology , Cytoskeletal Proteins , Drug Interactions , Exocytosis/drug effects , Free Radical Scavengers/pharmacology , Hippocampus/cytology , Imidazoles/pharmacology , N-Ethylmaleimide-Sensitive Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Nitric Oxide Donors/pharmacology , Nuclear Proteins/genetics , Patch-Clamp Techniques/methods , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , RNA, Small Interfering/pharmacology , Rats
15.
J Neurosci ; 26(32): 8339-51, 2006 Aug 09.
Article in English | MEDLINE | ID: mdl-16899729

ABSTRACT

Activity-dependent regulation of synaptic AMPA receptor (AMPAR) number is critical to NMDA receptor (NMDAR)-dependent synaptic plasticity. Using quantitative high-resolution in situ hybridization, we show that mRNAs encoding the AMPA-type glutamate receptor subunits (GluRs) 1 and 2 are localized to dendrites of hippocampal neurons and are regulated by paradigms that alter synaptic efficacy. A substantial fraction of synaptic sites contain AMPAR mRNA, consistent with strategic positioning and availability for "on-site" protein synthesis. NMDAR activation depletes dendritic levels of AMPAR mRNAs. The decrease in mRNA occurs via rise in intracellular Ca2+, activation of extracellular signal-regulated kinase/mitogen-activated protein kinase signaling, and transcriptional arrest at the level of the nucleus. The decrease in mRNA is accompanied by a long-lasting reduction in synaptic AMPAR number, consistent with reduced synaptic efficacy. In contrast, group I metabotropic GluR signaling promotes microtubule-based trafficking of existing AMPAR mRNAs from the soma to dendrites. Bidirectional regulation of dendritic mRNA abundance represents a potentially powerful means to effect long-lasting changes in synaptic strength.


Subject(s)
Dendrites/metabolism , Hippocampus/physiology , Neuronal Plasticity/physiology , Neurons/physiology , RNA, Messenger/metabolism , Receptors, AMPA/metabolism , Synaptic Transmission/physiology , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley , Tissue Distribution
16.
J Neurosci ; 26(19): 5028-36, 2006 May 10.
Article in English | MEDLINE | ID: mdl-16687494

ABSTRACT

The rapid cycling of AMPA receptors (AMPARs) at the membrane maintains synaptic transmission at a number of CNS synapses and may play a role in several forms of synaptic plasticity. It is unclear, however, how prevalent the trafficking of AMPARs is in the CNS, particularly at synapses not known to exhibit activity-dependent plasticity. Because trafficking is regulated by basal synaptic activity, a question also remains as to how receptor trafficking is modulated at synapses subject to different patterns of synaptic activation. We have investigated whether trafficking of AMPARs occurs in retinal neurons, which are subject to tonic glutamate release. We find two distinct states of AMPAR trafficking in ON ganglion cells. Light adaptation serves to stabilize AMPARs in a noncycling mode. However, dark adaptation for as little as 8 h triggers a switch to a second state of trafficking characterized by rapid cycling. We provide evidence that the activation of AMPARs is critical for switching between cycling and noncycling states. The induction of cycling further appears to be modulated by changes in the function of glutamate receptor 2/3-interacting proteins. Our results suggest that there is a strong link between synaptic activity and AMPAR trafficking in retinal neurons. These results further suggest the existence of a previously unknown form of activity-dependent plasticity in the retina that may be regulated in the course of a normal light/dark cycle.


Subject(s)
Protein Transport/physiology , Receptors, AMPA/metabolism , Retinal Ganglion Cells/physiology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Light , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Retina/physiology , Retina/radiation effects , Retinal Ganglion Cells/radiation effects , Synaptic Transmission/radiation effects
17.
Mol Cell Neurosci ; 32(1-2): 37-48, 2006.
Article in English | MEDLINE | ID: mdl-16631377

ABSTRACT

Genetic deficiency of the mRNA binding protein FMRP results in the most common inherited form of mental retardation, Fragile X syndrome. We investigated the localization and function of FMRP during development of hippocampal neurons in culture. FMRP was distributed within granules that extended into developing axons and growth cones, detectable at distances over 300 microm from the cell body. In mature cultures, FMRP granules were present in both axons and dendrites, with pockets of higher concentrations appearing intermittently, along distal axon segments and near synapses. MAP1b mRNA, a known FMRP target, was also localized to axon growth cones. Morphometric analysis of growth cones from the FMR1 KO revealed both excess filopodia and reduced motility. At later stages during synapse formation, FMR1 KO neurons exhibited excessive filopodia and long spines along dendrites, yet there was a marked decrease in the density of spine-like protrusions juxtaposed to presynaptic terminals. In contrast, there was no difference in the density of shaft synapses between FMR1 KO and WT. Brief depolarization of WT neurons resulted in increased numbers of filopodia and spine synapses, whereas no additional morphologic changes were observable in dendrites of FMR1 KO neurons that already had increased density of filopodia-spines. These findings suggest that alterations in the regulation of axonal growth and innervation in FMR1 KO neurons may contribute to the dendritic and spine pathology in Fragile X syndrome. This work has broader implications for understanding the role of mRNA binding proteins in developmental and protein-synthesis-dependent plasticity.


Subject(s)
Cell Movement , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/metabolism , Growth Cones/metabolism , Pseudopodia/metabolism , Synapses/metabolism , Animals , Animals, Newborn , Cell Differentiation/physiology , Cell Movement/physiology , Cells, Cultured , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Dendritic Spines/metabolism , Dendritic Spines/pathology , Dendritic Spines/ultrastructure , Female , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Growth Cones/pathology , Growth Cones/ultrastructure , Hippocampus/abnormalities , Hippocampus/growth & development , Hippocampus/pathology , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Pseudopodia/pathology , Pseudopodia/ultrastructure , RNA, Messenger/metabolism , Synapses/pathology , Synapses/ultrastructure , Synaptic Transmission/genetics
18.
J Neurosci ; 24(11): 2648-55, 2004 Mar 17.
Article in English | MEDLINE | ID: mdl-15028757

ABSTRACT

Fragile X syndrome is caused by the absence of the mRNA-binding protein Fragile X mental retardation protein (FMRP), which may play a role in activity-regulated localization and translation of mRNA in dendrites and at synapses. We investigated whether neuronal activity and glutamatergic signals regulate trafficking of FMRP and its encoding Fmr1 mRNA into dendrites or at synapses. Using high-resolution fluorescence and digital imaging microscopy in cultured hippocampal neurons, FMRP and Fmr1 mRNA were localized in granules throughout dendrites and within spines. KCl depolarization rapidly increased FMRP and Fmr1 mRNA levels in dendrites. Metabotropic glutamate receptor (mGluR) activation, in particular mGluR5 activation, was necessary for localization of FMRP into dendrites. Blockade of either PKC or internal calcium prevented mGluR-dependent localization of both FMRP and Fmr1 mRNA in dendrites. The activity-dependent localization of FMRP was not dependent on protein synthesis. Fluorescence recovery after photobleaching analysis of live neurons transfected with enhanced green fluorescent protein-FMRP revealed increased granule trafficking in response to KCl depolarization. In contrast to its dendritic localization, mGluR activation diminished FMRP, but not Fmr1 mRNA, localization at synapses. These results demonstrate regulation of FMRP and Fmr1 mRNA trafficking in dendrites and synapses in response to specific glutamatergic signals.


Subject(s)
Dendrites/metabolism , Fragile X Syndrome/metabolism , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism , Animals , Biological Transport , Calcium/metabolism , Cell Division , Cells, Cultured , Fluorescent Antibody Technique , Fragile X Mental Retardation Protein , Gene Expression Regulation, Developmental , Hippocampus/cytology , Hippocampus/embryology , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Nerve Tissue Proteins/genetics , Rats , Receptor, Metabotropic Glutamate 5
19.
Trends Neurosci ; 25(11): 571-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12392932

ABSTRACT

Dynamic regulation of synaptic efficacy is thought to play a crucial role in formation of neuronal connections and in experience-dependent modification of neural circuitry. The molecular and cellular mechanisms by which synaptic changes are triggered and expressed are the focus of intense interest. This articles reviews recent evidence that NMDA receptors undergo dynamically regulated targeting and trafficking, and that the physical transport of NMDA receptors in and out of the synaptic membrane contributes to several forms of long-lasting synaptic plasticity. The identification of targeting and internalization sequences in NMDA-receptor subunits has begun the unraveling of some mechanisms that underlie activity-dependent redistribution of NMDA receptors. Given that NMDA receptors are widely expressed throughout the CNS, regulation of NMDA-receptor trafficking provides a potentially important way to modulate efficacy of synaptic transmission.


Subject(s)
Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synaptic Transmission/physiology , Animals , Humans , Protein Transport/physiology , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...