Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
J Cell Sci ; 137(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38668720

ABSTRACT

Amyloid ß (Aß) is a central contributor to neuronal damage and cognitive impairment in Alzheimer's disease (AD). Aß disrupts AMPA receptor-mediated synaptic plasticity, a key factor in early AD progression. Numerous studies propose that Aß oligomers hinder synaptic plasticity, particularly long-term potentiation (LTP), by disrupting GluA1 (encoded by GRIA1) function, although the precise mechanism remains unclear. In this study, we demonstrate that Aß mediates the accumulation of GM1 ganglioside in lipid raft domains of cultured cells, and GluA1 exhibits preferential localization in lipid rafts via direct binding to GM1. Aß enhances the raft localization of GluA1 by increasing GM1 in these areas. Additionally, chemical LTP stimulation induces lipid raft-dependent GluA1 internalization in Aß-treated neurons, resulting in reduced cell surface and postsynaptic expression of GluA1. Consistent with this, disrupting lipid rafts and GluA1 localization in rafts rescues Aß-mediated suppression of hippocampal LTP. These findings unveil a novel functional deficit in GluA1 trafficking induced by Aß, providing new insights into the mechanism underlying AD-associated cognitive dysfunction.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Hippocampus , Long-Term Potentiation , Membrane Microdomains , Receptors, AMPA , Amyloid beta-Peptides/metabolism , Receptors, AMPA/metabolism , Membrane Microdomains/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Hippocampus/metabolism , G(M1) Ganglioside/metabolism , Humans , Neurons/metabolism , Rats , Mice , Protein Transport
2.
Neurosci Res ; 198: 21-29, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37429464

ABSTRACT

In the present study, we attempted to temporally and quantitatively analyze the functional contributions of Ca2+-permeable (CP) α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) during long-term potentiation (LTP) expression using electrophysiological and pharmacological approaches. In hippocampal CA1 neurons, using 1-naphthyl acetyl spermine (NASPM), a CP-AMPAR antagonist, we began by demonstrating that NASPM-sensitive components, probably including the GluA1 homomer, functionally contributed to about 15% of AMPAR-mediated EPSC amplitude in basal conditions. Then, when NASPM was treated at different time points (3-30 min) after LTP induction, it was found that LTP was almost completely impaired at 3 or 10 min but maintained at 20 or 30 min, although its potentiation was reduced. Further temporal and quantitative analysis revealed that the functional expression of CP-AMPARs began increasing approximately 20 min after LTP induction and reached more than twice the basal level at 30 min. These results suggest that CP-AMPARs in the first 3-10 min of LTP might play an important role in LTP maintenance. Moreover, their decay time was also significantly increased at 30 min, suggesting that CP-AMPARs changed not only quantitatively in LTP but also qualitatively.


Subject(s)
Long-Term Potentiation , Receptors, AMPA , Long-Term Potentiation/physiology , Receptors, AMPA/metabolism , Hippocampus/metabolism , Spermine/pharmacology , Calcium/metabolism , Synapses/metabolism
3.
FEBS Open Bio ; 13(9): 1615-1624, 2023 09.
Article in English | MEDLINE | ID: mdl-36999634

ABSTRACT

Genetic disruption of glycosyltransferases has provided clear information on the roles of their reaction products in the body. Our group has studied the function of glycosphingolipids by genetic engineering of glycosyltransferases in cell culture and in mice, which has demonstrated both expected and unexpected results. Among these findings, aspermatogenesis in ganglioside GM2/GD2 synthase knockout mice was one of the most surprising and intriguing results. There were no sperms in testis, and multinuclear giant cells were detected instead of spermatids. Although serum levels of testosterone in the male mice were extremely low, testosterone accumulated in the interstitial tissues, including Leydig cells, and seemed not to be transferred into the seminiferous tubules or vascular cavity from Leydig cells. This was considered to be the cause of aspermatogenesis and low serum levels of testosterone. Patients with a mutant GM2/GD2 synthase gene (SPG26) showed similar clinical signs, not only in terms of the neurological aspects, but also in the male reproductive system. The mechanisms for testosterone transport by gangliosides are discussed here based on our own results and reports from other laboratories.


Subject(s)
Gangliosides , N-Acetylgalactosaminyltransferases , Animals , Male , Mice , G(M2) Ganglioside , Gangliosides/genetics , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , Testosterone
4.
Cereb Cortex ; 31(4): 2205-2219, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33251537

ABSTRACT

Changes in the shape and size of the dendritic spines are critical for synaptic transmission. These morphological changes depend on dynamic assembly of the actin cytoskeleton and occur differently in various types of neurons. However, how the actin dynamics are regulated in a neuronal cell type-specific manner remains largely unknown. We show that Fhod3, a member of the formin family proteins that mediate F-actin assembly, controls the dendritic spine morphogenesis of specific subpopulations of cerebrocortical pyramidal neurons. Fhod3 is expressed specifically in excitatory pyramidal neurons within layers II/III and V of restricted areas of the mouse cerebral cortex. Immunohistochemical and biochemical analyses revealed the accumulation of Fhod3 in postsynaptic spines. Although targeted deletion of Fhod3 in the brain did not lead to any defects in the gross or histological appearance of the brain, the dendritic spines in pyramidal neurons within presumptive Fhod3-positive areas were morphologically abnormal. In primary cultures prepared from the Fhod3-depleted cortex, defects in spine morphology were only detected in Fhod3 promoter-active cells, a small population of pyramidal neurons, and not in Fhod3 promoter-negative pyramidal neurons. Thus, Fhod3 plays a crucial role in dendritic spine morphogenesis only in a specific population of pyramidal neurons in a cell type-specific manner.


Subject(s)
Cerebral Cortex/metabolism , Dendritic Spines/metabolism , Formins/biosynthesis , Pyramidal Cells/metabolism , Animals , Cells, Cultured , Cerebral Cortex/ultrastructure , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Formins/genetics , HEK293 Cells , Humans , Mice , Mice, Transgenic , Pyramidal Cells/ultrastructure
5.
Int J Mol Sci ; 21(14)2020 Jul 19.
Article in English | MEDLINE | ID: mdl-32707739

ABSTRACT

The AMPA-type glutamate receptor (AMPAR) is a homotetrameric or heterotetrameric ion channel composed of various combinations of four subunits (GluA1-4), and its abundance in the synapse determines the strength of synaptic activity. The formation of oligomers in the endoplasmatic reticulum (ER) is crucial for AMPAR subunits' ER-exit and translocation to the cell membrane. Although N-glycosylation on different AMPAR subunits has been shown to regulate the ER-exit of hetero-oligomers, its role in the ER-exit of homo-oligomers remains unclear. In this study, we investigated the role of N-glycans at GluA1N63/N363 and GluA2N370 in ER-exit under the homo-oligomeric expression conditions, whose mutants are known to show low cell surface expressions. In contrast to the N-glycosylation site mutant GluA1N63Q, the cell surface expression levels of GluA1N363Q and GluA2N370Q increased in a time-dependent manner. Unlike wild-type (WT) GluA1, GluA2WT rescued surface GluA2N370Q expression. Additionally, the expression of GluA1N63Q reduced the cell surface expression level of GluA1WT. In conclusion, our findings suggest that these N-glycans have distinct roles in the ER-exit of GluA1 and GluA2 homo-oligomers; N-glycan at GluA1N63 is a prerequisite for GluA1 ER-exit, whereas N-glycans at GluA1N363 and GluA2N370 control the ER-exit rate.


Subject(s)
Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Amino Acid Substitution , Binding Sites/genetics , Cell Membrane/metabolism , Gene Expression , Glycosylation , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mutagenesis, Site-Directed , Mutation , Protein Structure, Quaternary , Receptors, Glutamate/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
J Neurochem ; 153(5): 567-585, 2020 06.
Article in English | MEDLINE | ID: mdl-31958346

ABSTRACT

In the mammalian nervous system, protein N-glycosylation plays an important role in neuronal physiology. In this study, we performed a comprehensive N-glycosylation analysis of mouse GluA1, one of the major subunits of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate type glutamate receptor, which possesses six potential N-glycosylation sites in the N-terminal domain. By mass spectrometry-based analysis, we identified the N-glycoforms and semiquantitatively determined the site-specific N-glycosylation occupancy of GluA1. In addition, only the N401-glycosylation site demonstrated incomplete N-glycosylation occupancy. Therefore, we generated a peptide antibody that specifically detects the N401-glycan-free form to precisely quantify N401-glycosylation occupancy. Using this antibody, we clarified that N401 occupancy varies between cell types and increases in an age-dependent manner in mouse forebrains. To address the regulatory mechanism of N401-glycosylation, binding proteins of GluA1 around the N401 site were screened. HSP70 family proteins, including Bip, were identified as candidates. Bip has been known as a molecular chaperone that plays a key role in protein folding in the ER (endoplasmic reticulum). To examine the involvement of Bip in N401-glycosylation, the effect of Bip over-expression on N401 occupancy was evaluated in HEK293T cells, and the results demonstrated Bip increases the N401 glycan-free form by mediating selective prolongation of its protein half-life. Taken together, we propose that the N401-glycosite of GluA1 receives a unique control of modification, and we also propose a novel N-glycosylation occupancy regulatory mechanism by Bip that might be associated with α-amino-3-hydroxy-5-methyl-4-isoxazole-propionate receptors function in the brain.


Subject(s)
Antibodies/genetics , Antibodies/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Animals , Binding Sites/physiology , Female , Glycosylation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Pregnancy
7.
Nat Commun ; 10(1): 5245, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31748519

ABSTRACT

The number and subunit compositions of AMPA receptors (AMPARs), hetero- or homotetramers composed of four subunits GluA1-4, in the synapse is carefully tuned to sustain basic synaptic activity. This enables stimulation-induced synaptic plasticity, which is central to learning and memory. The AMPAR tetramers have been widely believed to be stable from their formation in the endoplasmic reticulum until their proteolytic decomposition. However, by observing GluA1 and GluA2 at the level of single molecules, we find that the homo- and heterotetramers are metastable, instantaneously falling apart into monomers, dimers, or trimers (in 100 and 200 ms, respectively), which readily form tetramers again. In the dendritic plasma membrane, GluA1 and GluA2 monomers and dimers are far more mobile than tetramers and enter and exit from the synaptic regions. We conclude that AMPAR turnover by lateral diffusion, essential for sustaining synaptic function, is largely done by monomers of AMPAR subunits, rather than preformed tetramers.


Subject(s)
Neuronal Plasticity , Neurons/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , CHO Cells , Cell Membrane/metabolism , Cricetulus , Dendrites/metabolism , Diffusion , HEK293 Cells , Humans , Mice , Microscopy, Fluorescence , Patch-Clamp Techniques , Single Molecule Imaging
8.
J Neurochem ; 147(6): 730-747, 2018 12.
Article in English | MEDLINE | ID: mdl-30092607

ABSTRACT

The AMPA-type glutamate receptor (AMPA-R) plays a primary role in principal excitatory synaptic transmission and many neuronal functions including synaptic plasticity that underlie learning and memory. N-glycosylation is one of the major post-translational modifications of membrane proteins, but its specific roles in neurons remain largely unknown. AMPA-R subunits are N-glycosylated at their extracellular domains during their biosynthesis in the lumen of the endoplasmic reticulum and Golgi system. Six N-glycosylation sites are presumed to exist in the extracellular domain of GluA1, which is a member of the AMPA-R subunits. We observed that the intracellular trafficking and cell surface expression were strongly suppressed in the GluA1 mutants lacking N-glycans at N63/N363 in HEK293T cells. Multimer analysis using Blue Native-PAGE displayed the impaired tetramer formation in the glycosylation mutants (N63S and N363S), indicating that the mis-transport was caused by impaired tetramer formation. N63S and N363S mutants were primarily degraded via the lysosomal pathway. Flag-tagged N363S GluA1, but not N63S GluA1, expressed in primary cortical neuron cultures prepared from GluA1 knockout mice was observed to localize at the cell surface. Co-expression of GluA2 partially rescued tetramer formation and the cell surface expression of N363S GluA1 but not N63S GluA1, in HEK293T cells. Electrophysiological analysis also demonstrated functional heteromers of N363S GluA1 with GluA2. These data suggest that site-specific N-glycans on GluA1 subunit regulates tetramer formation, intracellular trafficking, and cell surface expression of AMPA-R. OPEN SCIENCE BADGES: This article has received a badge for *Open Materials* because it provided all relevant information to reproduce the study in the manuscript. The complete Open Science Disclosure form for this article can be found at the end of the article. More information about the Open Practices badges can be found at https://cos.io/our-services/open-science-badges/.


Subject(s)
Glycosylation , Ion Channels/physiology , Membrane Proteins/biosynthesis , Receptors, AMPA/physiology , Animals , Electrophysiological Phenomena/genetics , HEK293 Cells , Humans , Lysosomes/metabolism , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Primary Cell Culture , Receptors, AMPA/biosynthesis , Receptors, AMPA/genetics , Receptors, AMPA/metabolism
9.
Neuron ; 93(6): 1405-1419.e8, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28285821

ABSTRACT

Learning depends on experience-dependent modification of synaptic efficacy and neuronal connectivity in the brain. We provide direct evidence for physiological roles of the recycling endosome protein GRASP1 in glutamatergic synapse function and animal behavior. Mice lacking GRASP1 showed abnormal excitatory synapse number, synaptic plasticity, and hippocampal-dependent learning and memory due to a failure in learning-induced synaptic AMPAR incorporation. We identified two GRASP1 point mutations from intellectual disability (ID) patients that showed convergent disruptive effects on AMPAR recycling and glutamate uncaging-induced structural and functional plasticity. Wild-type GRASP1, but not ID mutants, rescued spine loss in hippocampal CA1 neurons in Grasp1 knockout mice. Together, these results demonstrate a requirement for normal recycling endosome function in AMPAR-dependent synaptic function and neuronal connectivity in vivo, and suggest a potential role for GRASP1 in the pathophysiology of human cognitive disorders.


Subject(s)
Carrier Proteins/metabolism , Endosomes/metabolism , Learning/physiology , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Animals , Avoidance Learning/physiology , CA1 Region, Hippocampal/physiology , Carrier Proteins/genetics , Case-Control Studies , Cells, Cultured , Glutamic Acid/physiology , Humans , Intellectual Disability/genetics , Male , Maze Learning/physiology , Mice , Mice, Knockout , Mutation , Receptors, AMPA/physiology , Synapses/physiology
10.
PLoS One ; 8(12): e83129, 2013.
Article in English | MEDLINE | ID: mdl-24358255

ABSTRACT

Methods of cell biology and electrophysiology using dissociated primary cultured neurons allow in vitro study of molecular functions; however, analysis of intact neuronal circuitry is often preferable. To investigate exogenous genes, viral vectors are most commonly injected using a pipette that is inserted from the top of the cortex. Although there are few reports that describe the success rate of injection in detail, it is sometimes difficult to locate the pipette tip accurately within the CA1 pyramidal cell layer because the pyramidal layer is only 0.1 mm thick. In the present study, we have developed a system to inject viral vectors accurately into the mouse hippocampal CA1 pyramidal cell layer using a stereotaxic injection system with simultaneous electrophysiological monitoring of theta oscillation. The pipette tip was positioned reliably based on integrated values of the theta oscillation in the hippocampal CA1 pyramidal cell layer. This approach allows accurate injection of solutions and provides an efficient method of gene transfer using viral vectors into the hippocampus, which can be a useful tool for studies involving the molecular mechanisms of neuronal functions.


Subject(s)
CA1 Region, Hippocampal , Gene Transfer Techniques , Microinjections/instrumentation , Microinjections/methods , Theta Rhythm , Animals , CA1 Region, Hippocampal/metabolism , Coloring Agents/administration & dosage , Coloring Agents/pharmacology , Electrophysiology , Gene Transfer Techniques/instrumentation , HEK293 Cells , Humans , Injections, Intraventricular/instrumentation , Injections, Intraventricular/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microspheres , Receptors, AMPA/genetics , Theta Rhythm/genetics
11.
Arterioscler Thromb Vasc Biol ; 33(1): 76-86, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23117660

ABSTRACT

OBJECTIVE: Notch3 is critically important for the structure and myogenic response of distal arteries, particularly of cerebral arteries. However, signaling pathways acting downstream of Notch3 remain largely unknown. METHODS AND RESULTS: Transcriptome analysis using tail arteries of Notch3-null mice identified a core set of 17 novel Notch3-regulated genes confirmed in tail or brain arteries. Postnatal deletion of RBP-Jκ in smooth muscle cells recapitulated the structural, functional, and molecular defects of brain arteries induced by Notch3 deficiency. Transient in vivo blockade of the Notch pathway with γ-secretase inhibitors uncovered, in addition to Notch3, 6 immediate responders, including the voltage-gated potassium channel Kv1.5, which opposes to myogenic constriction of brain arteries, and the glutamate receptor-interacting protein 2 (Grip2) with no previously established role in the cerebrovasculature. We identified a vascular smooth muscle cell isoform of Grip2. We showed that Notch3-RBP-Jκ specifically regulates this isoform. Finally, we found that cerebral arteries of Grip2 mutant mice, which express an N-terminally truncated Grip2 protein, exhibited selective attenuation of pressure-induced contraction. CONCLUSIONS: Our data provide insight into how Notch3 signals in the brain arteries, establishing the postnatal requirement of smooth muscle RBP-Jκ in this context. Notch3-regulated transcriptome provides potential for modulating myogenic response in the cerebrovasculature.


Subject(s)
Carrier Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Notch/metabolism , Vasoconstriction , Alanine/analogs & derivatives , Alanine/pharmacology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Azepines/pharmacology , Carrier Proteins/genetics , Cerebral Arteries/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Expression Regulation , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Intracellular Signaling Peptides and Proteins , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/growth & development , Myocytes, Smooth Muscle/drug effects , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptor, Notch3 , Receptors, Notch/deficiency , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation , Vasodilator Agents/pharmacology
12.
PLoS One ; 7(11): e48967, 2012.
Article in English | MEDLINE | ID: mdl-23145041

ABSTRACT

From a genetic screen for Drosophila melanogaster mutants with altered ethanol tolerance, we identified intolerant (intol), a novel allele of discs large 1 (dlg1). Dlg1 encodes Discs Large 1, a MAGUK (Membrane Associated Guanylate Kinase) family member that is the highly conserved homolog of mammalian PSD-95 and SAP97. The intol mutation disrupted specifically the expression of DlgS97, a SAP97 homolog, and one of two major protein isoforms encoded by dlg1 via alternative splicing. Expression of the major isoform, DlgA, a PSD-95 homolog, appeared unaffected. Ethanol tolerance in the intol mutant could be partially restored by transgenic expression of DlgS97, but not DlgA, in specific neurons of the fly's brain. Based on co-immunoprecipitation, DlgS97 forms a complex with N-methyl-D-aspartate (NMDA) receptors, a known target of ethanol. Consistent with these observations, flies expressing reduced levels of the essential NMDA receptor subunit dNR1 also showed reduced ethanol tolerance, as did mutants in the gene calcium/calmodulin-dependent protein kinase (caki), encoding the fly homolog of mammalian CASK, a known binding partner of DlgS97. Lastly, mice in which SAP97, the mammalian homolog of DlgS97, was conditionally deleted in adults failed to develop rapid tolerance to ethanol's sedative/hypnotic effects. We propose that DlgS97/SAP97 plays an important and conserved role in the development of tolerance to ethanol via NMDA receptor-mediated synaptic plasticity.


Subject(s)
Ethanol/toxicity , Guanylate Kinases/genetics , Membrane Proteins/genetics , Neurons/metabolism , Alleles , Alternative Splicing , Animals , Discs Large Homolog 1 Protein , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Guanylate Kinases/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Protein Isoforms , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
13.
PLoS One ; 7(9): e45276, 2012.
Article in English | MEDLINE | ID: mdl-23028902

ABSTRACT

BACKGROUND: The polarized reorganization of the T cell membrane and intracellular signaling molecules in response to T cell receptor (TCR) engagement has been implicated in the modulation of T cell development and effector responses. In siRNA-based studies Dlg1, a MAGUK scaffold protein and member of the Scribble polarity complex, has been shown to play a role in T cell polarity and TCR signal specificity, however the role of Dlg1 in T cell development and function in vivo remains unclear. METHODOLOGY/PRINCIPAL FINDINGS: Here we present the combined data from three independently-derived dlg1-knockout mouse models; two germline deficient knockouts and one conditional knockout. While defects were not observed in T cell development, TCR-induced early phospho-signaling, actin-mediated events, or proliferation in any of the models, the acute knockdown of Dlg1 in Jurkat T cells diminished accumulation of actin at the IS. Further, while Th1-type cytokine production appeared unaffected in T cells derived from mice with a dlg1 germline-deficiency, altered production of TCR-dependent Th1 and Th2-type cytokines was observed in T cells derived from mice with a conditional loss of dlg1 expression and T cells with acute Dlg1 suppression, suggesting a differential requirement for Dlg1 activity in signaling events leading to Th1 versus Th2 cytokine induction. The observed inconsistencies between these and other knockout models and siRNA strategies suggest that 1) compensatory upregulation of alternate gene(s) may be masking a role for dlg1 in controlling TCR-mediated events in dlg1 deficient mice and 2) the developmental stage during which dlg1 ablation begins may control the degree to which compensatory events occur. CONCLUSIONS/SIGNIFICANCE: These findings provide a potential explanation for the discrepancies observed in various studies using different dlg1-deficient T cell models and underscore the importance of acute dlg1 ablation to avoid the upregulation of compensatory mechanisms for future functional studies of the Dlg1 protein.


Subject(s)
Nerve Tissue Proteins/genetics , Receptors, Antigen, T-Cell/genetics , Th1 Cells/metabolism , Th1-Th2 Balance , Th2 Cells/metabolism , Actins/genetics , Actins/immunology , Animals , Cell Communication/genetics , Cell Differentiation , Cytokines/biosynthesis , Cytokines/immunology , Discs Large Homolog 1 Protein , Gene Expression Regulation, Developmental , Genetic Engineering , Germ-Line Mutation , Lymphocyte Activation , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/immunology , Receptors, Antigen, T-Cell/immunology , SAP90-PSD95 Associated Proteins , Signal Transduction , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
14.
PLoS One ; 7(8): e44055, 2012.
Article in English | MEDLINE | ID: mdl-22937151

ABSTRACT

To evaluate the involvement of trafficking of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) GluR2 and GluR3 subunits in an acute inflammatory orofacial pain, we analyzed nocifensive behavior, phosphorylated extracellular signal-regulated kinase (pERK) and Fos expression in Vi/Vc, Vc and C1/C2 in GluR2 delta7 knock-in (KI), GluR3 delta7 KI mice and wild-type mice. We also studied Vc neuronal activity to address the hypothesis that trafficking of GluR2 and GluR3 subunits plays an important role in Vi/Vc, Vc and C1/C2 neuronal activity associated with orofacial inflammation in these mice. Late nocifensive behavior was significantly depressed in GluR2 delta7 KI and GluR3 delta7 KI mice. In addition, the number of pERK-immunoreactive (IR) cells was significantly decreased bilaterally in the Vi/Vc, Vc and C1/C2 in GluR2 delta7 KI and GluR3 delta7 KI mice compared to wild-type mice at 40 min after formalin injection, and was also significantly smaller in GluR3 delta7 KI compared to GluR2 delta7 KI mice. The number of Fos protein-IR cells in the ipsilateral Vi/Vc, Vc and C1/C2 was also significantly smaller in GluR2 delta7 KI and GluR3 delta7 KI mice compared to wild-type mice 40 min after formalin injection. Nociceptive neurons functionally identified as wide dynamic range neurons in the Vc, where pERK- and Fos protein-IR cell expression was prominent, showed significantly lower spontaneous activity in GluR2 delta7 KI and GluR3 delta7 KI mice than wild-type mice following formalin injection. These findings suggest that GluR2 and GluR3 trafficking is involved in the enhancement of Vi/Vc, Vc and C1/C2 nociceptive neuronal excitabilities at 16-60 min following formalin injection, resulting in orofacial inflammatory pain.


Subject(s)
Acute Pain/metabolism , Facial Pain/metabolism , Nociceptors/metabolism , Receptors, AMPA/metabolism , Trigeminal Caudal Nucleus/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Acute Pain/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Facial Pain/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Mice , Mice, Transgenic , Nociceptors/drug effects , Pain Measurement , Phosphorylation/drug effects , Trigeminal Caudal Nucleus/drug effects , Trigeminal Caudal Nucleus/physiopathology
15.
Behav Brain Res ; 229(1): 265-72, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22285418

ABSTRACT

Glutamate signaling has been implicated in the regulation of social behavior. AMPA-glutamate receptors are assembled from four subunits (GluA1-4) of mainly GluA1/2 and GluA2/3 tetramers that form ion channels of distinct functional properties. Mice lacking GluA1 showed a reduced anxiety and male aggression. To understand the role of GluA3 in modulating social behavior, we investigated GluA3-deficient mice (Gria3-/Y) on C57BL/6J background. Compared to wild type (WT) littermates (n=14), Gria3-/Y mice (n=13) showed an increase in isolation-induced male aggression (p=0.011) in home cage resident-intruder test; an increase in sociability (p=0.01), and increase in male-male social interactions in neutral arena (p=0.005); an increase in peripheral activities in open field test (p=0.037) with normal anxiety levels in elevated plus maze and light-dark box; and minor deficits in motor and balance function in accelerating rotarod test (p=0.016) with normal grip strength. Gria3-/Y mice showed no significant deficit in spatial memory function in Morris-water maze and Y-maze tests, and normal levels of testosterone. Increased dopamine concentrations in stratum (p=0.034) and reduced serotonin turnover in olfactory bulb (p=0.002) were documented in Gria3-/Y mice. These results support a role of GluA3 in the modulation of social behavior through brain dopamine and/or serotonin signaling and different AMPA receptor subunits affect social behavior through distinct mechanisms.


Subject(s)
Aggression/physiology , Corpus Striatum/metabolism , Dopamine/metabolism , Receptors, AMPA/deficiency , Social Behavior , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Anxiety/genetics , Anxiety/physiopathology , Dark Adaptation/genetics , Exploratory Behavior/physiology , Hand Strength/physiology , Homovanillic Acid/metabolism , Male , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reaction Time/genetics , Smell/genetics , Space Perception/physiology , Testosterone/blood
16.
Neuron ; 71(6): 1022-9, 2011 Sep 22.
Article in English | MEDLINE | ID: mdl-21943600

ABSTRACT

KIBRA has recently been identified as a gene associated with human memory performance. Despite the elucidation of the role of KIBRA in several diverse processes in nonneuronal cells, the molecular function of KIBRA in neurons is unknown. We found that KIBRA directly binds to the protein interacting with C-kinase 1 (PICK1) and forms a complex with α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptors (AMPARs), the major excitatory neurotransmitter receptors in the brain. KIBRA knockdown accelerates the rate of AMPAR recycling following N-methyl-D-aspartate receptor-induced internalization. Genetic deletion of KIBRA in mice impairs both long-term depression and long-term potentiation at hippocampal Schaffer collateral-CA1 synapses. Moreover, KIBRA knockout mice have severe deficits in contextual fear learning and memory. These results indicate that KIBRA regulates higher brain function by regulating AMPAR trafficking and synaptic plasticity.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Memory/physiology , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Receptors, AMPA/metabolism , Animals , Behavior, Animal/physiology , Carrier Proteins/genetics , Cells, Cultured , Conditioning, Classical/physiology , Electrophysiology , Fear , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Learning/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/physiology , Nuclear Proteins/genetics , Phosphoproteins/metabolism , Receptors, AMPA/genetics
17.
J Physiol ; 589(17): 4209-27, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21690196

ABSTRACT

Fast excitatory synaptic transmission in central synapses is mediated primarily by AMPA receptors (AMPARs), which are heteromeric assemblies of four subunits, GluA1-4. Among these subunits, rapidly gating GluA3/4 appears to be the most abundantly expressed to enable neurotransmission with submillisecond precision at fast rates in subsets of central synapses. However, neither definitive identification of the molecular substrate for native AMPARs in these neurons, nor their hypothesized functional roles in vivo has been unequivocally demonstrated, largely due to lack of specific antagonists. Using GluA3 or GluA4 knockout (KO) mice, we investigated these issues at the calyx of Held synapse, which is known as a high-fidelity synapse involved in sound localization. Patch-clamp recordings from postsynaptic neurons showed that deletion of GluA4 significantly slowed the time course of both evoked and miniature AMPAR-mediated excitatory postsynaptic currents (AMPAR-EPSCs), reduced their amplitude, and exacerbated AMPAR desensitization and short-term depression (STD). Surprisingly, presynaptic release probability was also elevated, contributing to severe STD at GluA4-KO synapses. In contrast, only marginal changes in AMPAR-EPSCs were found in GluA3-KO mice. Furthermore, independent of changes in intrinsic excitability of postsynaptic neurons, deletion of GluA4 markedly reduced synaptic drive and increased action potential failures during high-frequency activity, leading to profound deficits in specific components of the auditory brainstem responses associated with synchronized spiking in the calyx of Held synapse and other related neurons in vivo. These observations identify GluA4 as the main determinant for fast synaptic response, indispensable for driving high-fidelity neurotransmission and conveying precise temporal information.


Subject(s)
Patch-Clamp Techniques , Synapses , Animals , Excitatory Postsynaptic Potentials , Receptors, AMPA/metabolism , Synaptic Transmission
18.
Proc Natl Acad Sci U S A ; 108(20): 8450-5, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21536866

ABSTRACT

Phosphorylation of the GluA1 subunit of AMPA receptors has been proposed to regulate receptor trafficking and synaptic transmission and plasticity. However, it remains unclear whether GluA1 phosphorylation is permissive or sufficient for enacting these functional changes. Here we investigate the role of GluA1 phosphorylation at S831 and S845 residues in the hippocampus through the analyses of GluA1 S831D/S845D phosphomimetic knock-in mice. S831D/S845D mice showed normal total and surface expression and subcellular localization of GluA1 as well as intact basal synaptic transmission. In addition, theta-burst stimulation, a protocol that was sufficient to induce robust long-term potentiation (LTP) in WT mice, resulted in LTP of similar magnitude in S831D/S845D mice. However, S831D/S845D mice showed LTP induced with 10-Hz stimulation, a protocol that is weaker than theta-burst stimulation and was not sufficient to induce LTP in WT mice. Moreover, S831D/S845D mice exhibited LTP induced with spike-timing-dependent plasticity (STDP) protocol at a long pre-post interval that was subthreshold for WT mice, although a suprathreshold STDP protocol at a short pre-post interval resulted in similarly robust LTP for WT and S831D/S845D mice. These results indicate that phosphorylation of GluA1 at S831 and S845 is sufficient to lower the threshold for LTP induction, increasing the probability of synaptic plasticity.


Subject(s)
Long-Term Potentiation , Mutation , Neuronal Plasticity , Receptors, AMPA/genetics , Animals , Hippocampus , Mice , Phosphorylation , Receptors, AMPA/metabolism , Synaptic Transmission
19.
Neuron ; 70(1): 43-50, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21482355

ABSTRACT

Long-term depression at parallel fiber-Purkinje cell synapses (PF-PC LTD) has been proposed to be required for cerebellar motor learning. To date, tests of this hypothesis have sought to interfere with receptors (mGluR1) and enzymes (PKC, PKG, or αCamKII) necessary for induction of PF-PC LTD and thereby determine if cerebellar motor learning is impaired. Here, we tested three mutant mice that target the expression of PF-PC LTD by blocking internalization of AMPA receptors. Using three different cerebellar coordination tasks (adaptation of the vestibulo-ocular reflex, eyeblink conditioning, and locomotion learning on the Erasmus Ladder), we show that there is no motor learning impairment in these mutant mice that lack PF-PC LTD. These findings demonstrate that PF-PC LTD is not essential for cerebellar motor learning.


Subject(s)
Blinking/physiology , Cerebellum/physiology , Learning/physiology , Long-Term Synaptic Depression/physiology , Motor Activity/physiology , Reflex, Vestibulo-Ocular/physiology , Animals , Gene Knock-In Techniques , Mice , Mice, Knockout , Nerve Net/physiology , Neuronal Plasticity/physiology
20.
PLoS One ; 6(3): e18264, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21483826

ABSTRACT

Sensory experience, and the lack thereof, can alter the function of excitatory synapses in the primary sensory cortices. Recent evidence suggests that changes in sensory experience can regulate the synaptic level of Ca(2+)-permeable AMPA receptors (CP-AMPARs). However, the molecular mechanisms underlying such a process have not been determined. We found that binocular visual deprivation, which is a well-established in vivo model to produce multiplicative synaptic scaling in visual cortex of juvenile rodents, is accompanied by an increase in the phosphorylation of AMPAR GluR1 (or GluA1) subunit at the serine 845 (S845) site and the appearance of CP-AMPARs at synapses. To address the role of GluR1-S845 in visual deprivation-induced homeostatic synaptic plasticity, we used mice lacking key phosphorylation sites on the GluR1 subunit. We found that mice specifically lacking the GluR1-S845 site (GluR1-S845A mutants), which is a substrate of cAMP-dependent kinase (PKA), show abnormal basal excitatory synaptic transmission and lack visual deprivation-induced homeostatic synaptic plasticity. We also found evidence that increasing GluR1-S845 phosphorylation alone is not sufficient to produce normal multiplicative synaptic scaling. Our study provides concrete evidence that a GluR1 dependent mechanism, especially S845 phosphorylation, is a necessary pre-requisite step for in vivo homeostatic synaptic plasticity.


Subject(s)
Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Sensory Deprivation/physiology , Synapses/metabolism , Animals , Electrophoresis, Polyacrylamide Gel , Immunoblotting , In Vitro Techniques , Mice , Neuronal Plasticity/genetics , Phosphorylation , Receptors, AMPA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...