Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
1.
iScience ; 24(2): 102037, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33532714

ABSTRACT

Gephyrin is critical for the structure, function, and plasticity of inhibitory synapses. Gephyrin mutations have been linked to various neurological disorders; however, systematic analyses of the functional consequences of these mutations are lacking. Here, we performed molecular dynamics simulations of gephyrin to predict how six reported point mutations might change the structural stability and/or function of gephyrin. Additional in silico analyses revealed that the A91T and G375D mutations reduce the binding free energy of gephyrin oligomer formation. Gephyrin A91T and G375D displayed altered clustering patterns in COS-7 cells and nullified the inhibitory synapse-promoting effect of gephyrin in cultured neurons. However, only the G375D mutation reduced gephyrin interaction with GABAA receptors and neuroligin-2 in mouse brain; it also failed to normalize deficits in GABAergic synapse maintenance and neuronal hyperactivity observed in hippocampal dentate gyrus-specific gephyrin-deficient mice. Our results provide insights into biochemical, cell-biological, and network-activity effects of the pathogenic G375D mutation.

2.
Neuron ; 96(4): 730-735, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29144972

ABSTRACT

Science is ideally suited to connect people from different cultures and thereby foster mutual understanding. To promote international life science collaboration, we have launched "The Science Bridge" initiative. Our current project focuses on partnership between Western and Middle Eastern neuroscience communities.


Subject(s)
International Cooperation , Neurosciences/history , Europe , History, 15th Century , History, 21st Century , History, Ancient , History, Medieval , Humans , Middle East
3.
Proc Natl Acad Sci U S A ; 114(34): E7179-E7186, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28784756

ABSTRACT

Neuronal inhibition can occur via synaptic mechanisms or through tonic activation of extrasynaptic receptors. In spinal cord, glycine mediates synaptic inhibition through the activation of heteromeric glycine receptors (GlyRs) composed primarily of α1 and ß subunits. Inhibitory GlyRs are also found throughout the brain, where GlyR α2 and α3 subunit expression exceeds that of α1, particularly in forebrain structures, and coassembly of these α subunits with the ß subunit appears to occur to a lesser extent than in spinal cord. Here, we analyzed GlyR currents in several regions of the adolescent mouse forebrain (striatum, prefrontal cortex, hippocampus, amygdala, and bed nucleus of the stria terminalis). Our results show ubiquitous expression of GlyRs that mediate large-amplitude currents in response to exogenously applied glycine in these forebrain structures. Additionally, tonic inward currents were also detected, but only in the striatum, hippocampus, and prefrontal cortex (PFC). These tonic currents were sensitive to both strychnine and picrotoxin, indicating that they are mediated by extrasynaptic homomeric GlyRs. Recordings from mice deficient in the GlyR α3 subunit (Glra3-/-) revealed a lack of tonic GlyR currents in the striatum and the PFC. In Glra2-/Y animals, GlyR tonic currents were preserved; however, the amplitudes of current responses to exogenous glycine were significantly reduced. We conclude that functional α2 and α3 GlyRs are present in various regions of the forebrain and that α3 GlyRs specifically participate in tonic inhibition in the striatum and PFC. Our findings suggest roles for glycine in regulating neuronal excitability in the forebrain.


Subject(s)
Glycine Agents/pharmacology , Glycine/metabolism , Prosencephalon/physiology , Receptors, Glycine/metabolism , Strychnine/pharmacology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Picrotoxin/pharmacology , Prosencephalon/drug effects , Receptors, Glycine/genetics
4.
J Biol Chem ; 292(4): 1160-1177, 2017 01 27.
Article in English | MEDLINE | ID: mdl-27941024

ABSTRACT

The formation of neuronal synapses and the dynamic regulation of their efficacy depend on the proper assembly of the postsynaptic neurotransmitter receptor apparatus. Receptor recruitment to inhibitory GABAergic postsynapses requires the scaffold protein gephyrin and the guanine nucleotide exchange factor collybistin (Cb). In vitro, the pleckstrin homology domain of Cb binds phosphoinositides, specifically phosphatidylinositol 3-phosphate (PI3P). However, whether PI3P is required for inhibitory postsynapse formation is currently unknown. Here, we investigated the role of PI3P at developing GABAergic postsynapses by using a membrane-permeant PI3P derivative, time-lapse confocal imaging, electrophysiology, as well as knockdown and overexpression of PI3P-metabolizing enzymes. Our results provide the first in cellula evidence that PI3P located at early/sorting endosomes regulates the postsynaptic clustering of gephyrin and GABAA receptors and the strength of inhibitory, but not excitatory, postsynapses in cultured hippocampal neurons. In human embryonic kidney 293 cells, stimulation of gephyrin cluster formation by PI3P depends on Cb. We therefore conclude that the endosomal pool of PI3P, generated by the class III phosphatidylinositol 3-kinase, is important for the Cb-mediated recruitment of gephyrin and GABAA receptors to developing inhibitory postsynapses and thus the formation of postsynaptic membrane specializations.


Subject(s)
Carrier Proteins/metabolism , Endosomes/metabolism , GABAergic Neurons/metabolism , Membrane Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Synaptic Membranes/metabolism , Synaptic Potentials/physiology , Animals , GABAergic Neurons/cytology , Humans , Phosphatidylinositol 3-Kinases/metabolism , Rats , Receptors, GABA-A/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism
5.
Front Mol Neurosci ; 9: 75, 2016.
Article in English | MEDLINE | ID: mdl-27625595

ABSTRACT

The causal interrogation of neuronal networks involved in specific behaviors requires the spatially and temporally controlled modulation of neuronal activity. For long-term manipulation of neuronal activity, chemogenetic tools provide a reasonable alternative to short-term optogenetic approaches. Here we show that virus mediated gene transfer of the ivermectin (IVM) activated glycine receptor mutant GlyRα1 (AG) can be used for the selective and reversible silencing of specific neuronal networks in mice. In the striatum, dorsal hippocampus, and olfactory bulb, GlyRα1 (AG) promoted IVM dependent effects in representative behavioral assays. Moreover, GlyRα1 (AG) mediated silencing had a strong and reversible impact on neuronal ensemble activity and c-Fos activation in the olfactory bulb. Together our results demonstrate that long-term, reversible and re-inducible neuronal silencing via GlyRα1 (AG) is a promising tool for the interrogation of network mechanisms underlying the control of behavior and memory formation.

6.
Hear Res ; 337: 25-34, 2016 07.
Article in English | MEDLINE | ID: mdl-27208792

ABSTRACT

Inhibitory glycine receptors containing the α3 subunit (GlyRα3) regulate sensory information processing in the CNS and retina. In previous work, we demonstrated the presence of postsynaptic GlyRα3 immunoreactivity at efferent synapses of the medial and lateral olivocochlear bundle in the organ of Corti; however, the role of these α3-GlyRs in auditory signalling has remained elusive. The present study analyzes distortion-product otoacoustic emissions (DPOAEs) and auditory brainstem responses (ABRs) of knockout mice with a targeted inactivation of the Glra3 gene (Glra3(-/-)) and their wildtype littermates (Glra3(+/+)) before and seven days after acoustic trauma (AT; 4-16 kHz, 120 dB SPL, 1 h). Before AT, DPOAE thresholds were slightly, but significantly lower, and DPOAE amplitudes were slightly larger in Glra3(-/-) as compared to Glra3(+/+) mice. While click- and f-ABR thresholds were similar in both genotypes before AT, threshold-normalized click-ABR wave I amplitudes were smaller in Glra3(-/-) mice as compared to their wildtype littermates. Following AT, both the decrement of ABR wave I amplitudes and the delay of wave I latencies were more pronounced in Glra3(-/-) than Glra3(+/+) mice. Accordingly, correlation between early click-evoked ABR signals (0-2.5 ms from stimulus onset) before and after AT was significantly reduced for Glra3(-/-) as compared to Glra3(+/+) mice. In summary, these results show that loss of α3-GlyRs compromises suprathreshold auditory nerve activity, but not outer hair cell function.


Subject(s)
Cochlear Nerve/physiopathology , Hair Cells, Vestibular/pathology , Hearing Loss, Noise-Induced/physiopathology , Otoacoustic Emissions, Spontaneous , Receptors, Glycine/genetics , Acoustic Stimulation , Animals , Auditory Threshold/physiology , Cochlea/physiology , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory, Outer/metabolism , Hearing , Mice , Mice, Knockout , Receptors, Glycine/physiology , Synapses/pathology
7.
Mol Cell Neurosci ; 72: 101-13, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26829712

ABSTRACT

Mutations that result in the defective trafficking of γ2 subunit containing GABAA receptors (γ2-GABAARs) are known to reduce synaptic inhibition. Whether perturbed clustering of non-mutated GABAARs similarly reduces synaptic inhibition in vivo is less clear. In this study we provide evidence that the loss of postsynaptic γ2-GABAARs upon postnatal ablation of gephyrin, the major scaffolding protein of inhibitory postsynapses, from mature principal neurons within the forebrain results in reduced induction of long-term potentiation (LTP) and impaired network excitability within the hippocampal dentate gyrus. The preferential reduction in not only synaptic γ2-GABAAR cluster number at dendritic sites but also the decrease in γ2-GABAAR density within individual clusters at dendritic inhibitory synapses suggests that distal synapses are more sensitive to the loss of gephyrin expression than proximal synapses. The fact that these mice display behavioural features of anxiety and epilepsy emphasises the importance of postsynaptic γ2-GABAAR clustering for synaptic inhibition.


Subject(s)
Carrier Proteins/genetics , Long-Term Potentiation , Membrane Proteins/genetics , Prosencephalon/metabolism , Receptors, GABA-A/metabolism , Synaptic Potentials , Animals , Carrier Proteins/metabolism , Cell Line , Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Dentate Gyrus/physiology , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Prosencephalon/cytology , Prosencephalon/physiology , Receptors, GABA-A/genetics , Synapses/metabolism , Synapses/physiology
8.
Neuropharmacology ; 105: 133-141, 2016 06.
Article in English | MEDLINE | ID: mdl-26777280

ABSTRACT

N-methyl-d-aspartate (NMDA) receptors composed of glycine-binding GluN1 and GluN3 subunits function as excitatory glycine receptors that respond to agonist application only with a very low efficacy. Binding of glycine to the high-affinity GluN3 subunits triggers channel opening, whereas glycine binding to the low-affinity GluN1 subunits causes an auto-inhibition of the maximal glycine-inducible receptor current (Imax). Hence, competitive antagonists of the GluN1 subunit strongly potentiate glycine responses of wild type (wt) GluN1/GluN3 receptors. Here, we show that co-expression of N-terminal domain (NTD) deleted GluN1 (GluN1(ΔNTD)) and GluN3 (GluN3(ΔNTD)) subunits in Xenopus oocytes generates GluN1/GluN3 receptors with a large increase in the glycine-inducible Imax accompanied by a strongly impaired GluN1 antagonist-mediated potentiation. Affinity purification after metabolic or surface labeling revealed no differences in subunit stoichiometry and surface expression between wt GluN1/GluN3A and mutant GluN1(ΔNTD)/GluN3A(ΔNTD) receptors, indicating a specific effect of NTD deletions on the efficacy of receptor opening. Notably, GluN1/GluN3A(ΔNTD) receptors showed a similar increase in Imax and a greatly reduced GluN1 antagonist-mediated current potentiation as GluN1(ΔNTD)/GluN3A(ΔNTD) receptors, whereas the glycine-induced currents of GluN1(ΔNTD)/GluN3A receptors resembled those of wt GluN1/GluN3A receptors. Furthermore, oxidative crosslinking of the homophilic GluN3A NTD intersubunit interface in mutant GluN1/GluN3A(R319C) receptors caused both a decrease in the glycine-induced Imax concomitantly with a marked increase in GluN1 antagonist-mediated current potentiation, whilst mutations within the intrasubunit region linking the GluN3A NTD to the ligand binding domain had opposite effects. Together these results show that the GluN3A NTD constitutes a crucial regulatory determinant of GluN1/GluN3A receptor function.


Subject(s)
Glycine/physiology , Receptors, Glycine/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Binding Sites , Glycine/pharmacology , Protein Structure, Tertiary , Receptors, Glycine/agonists , Receptors, Glycine/chemistry , Xenopus laevis
9.
J Biol Chem ; 289(16): 11396-11409, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24509844

ABSTRACT

Glycine receptors (GlyRs) mediate inhibitory neurotransmission in spinal cord and brainstem. They are clustered at inhibitory postsynapses via a tight interaction of their ß subunits (GlyRß) with the scaffolding protein gephyrin. In an attempt to isolate additional proteins interacting with GlyRß, we performed pulldown experiments with rat brain extracts using a glutathione S-transferase fusion protein encompassing amino acids 378-455 of the large intracellular loop of GlyRß as bait. This identified syndapin I (SdpI) as a novel interaction partner of GlyRß that coimmunoprecipitates with native GlyRs from brainstem extracts. Both SdpI and SdpII bound efficiently to the intracellular loop of GlyRß in vitro and colocalized with GlyRß upon coexpression in COS-7 cells. The SdpI-binding site was mapped to a proline-rich sequence of 22 amino acids within the intracellular loop of GlyRß. Deletion and point mutation analysis disclosed that SdpI binding to GlyRß is Src homology 3 domain-dependent. In cultured rat spinal cord neurons, SdpI immunoreactivity was found to partially colocalize with marker proteins of inhibitory and excitatory synapses. When SdpI was acutely knocked down in cultured spinal cord neurons by viral miRNA expression, postsynaptic GlyR clusters were significantly reduced in both size and number. Similar changes in GlyR cluster properties were found in spinal cultures from SdpI-deficient mice. Our results are consistent with a role of SdpI in the trafficking and/or cytoskeletal anchoring of synaptic GlyRs.


Subject(s)
Brain/metabolism , Carrier Proteins/metabolism , Neuropeptides/metabolism , Phosphoproteins/metabolism , Receptors, Glycine/metabolism , Spinal Cord/metabolism , Synapses/metabolism , Adaptor Proteins, Signal Transducing , Animals , Brain/cytology , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , Cytoskeletal Proteins , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Mutant Strains , Neurons/cytology , Neurons/metabolism , Neuropeptides/genetics , Phosphoproteins/genetics , Point Mutation , Protein Binding , Protein Structure, Secondary , Protein Transport/physiology , Proteomics , Rats , Rats, Wistar , Receptors, Glycine/genetics , Spinal Cord/cytology , Synapses/genetics
10.
Proc Natl Acad Sci U S A ; 110(51): 20795-800, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297911

ABSTRACT

In many brain regions, gephyrin and GABAA receptor clustering at developing inhibitory synapses depends on the guanine nucleotide exchange factor collybistin (Cb). The vast majority of Cb splice variants contain an autoinhibitory src homology 3 domain, and several synaptic proteins are known to bind to this SH3 domain and to thereby activate gephyrin clustering. However, many functional GABAergic synapses form independently of the known Cb-activating proteins, indicating that additional Cb activators must exist. Here we show that the small Rho-like GTPase TC10 stimulates Cb-dependent gephyrin clustering by binding in its active, GTP-bound state to the pleckstrin homology domain of Cb. Overexpression of a constitutively active TC10 variant in neurons causes an increase in the density of synaptic gephyrin clusters and mean miniature inhibitory postsynaptic current amplitudes, whereas a dominant negative TC10 variant has opposite effects. The enhancement of Cb-induced gephyrin clustering by GTP-TC10 does not depend on the guanine nucleotide exchange activity of Cb but involves an interaction that resembles reported interactions of other small GTPases with their effectors. Our data indicate that GTP-TC10 activates the major src homology 3 domain-containing Cb variants by relieving autoinhibition and thus define an alternative GTPase-driven signaling pathway in the genesis of inhibitory synapses.


Subject(s)
Carrier Proteins/metabolism , GABAergic Neurons/metabolism , Guanosine Triphosphate/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Post-Synaptic Density/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Synaptic Potentials/physiology , rho GTP-Binding Proteins/metabolism , Animals , COS Cells , Carrier Proteins/genetics , Chlorocebus aethiops , GABAergic Neurons/cytology , Guanosine Triphosphate/genetics , Hippocampus/cytology , Humans , Membrane Proteins/genetics , Post-Synaptic Density/genetics , Protein Structure, Tertiary , Rats , Rho Guanine Nucleotide Exchange Factors/genetics , rho GTP-Binding Proteins/genetics
11.
Cereb Cortex ; 23(11): 2700-11, 2013 Nov.
Article in English | MEDLINE | ID: mdl-22918984

ABSTRACT

Gephyrin is a scaffolding protein important for the postsynaptic clustering of inhibitory neurotransmitter receptors. Here, we investigated the properties of gephyrin scaffolds at γ-aminobutyric acid- (GABA-)ergic synapses in organotypic entorhino-hippocampal cultures prepared from a transgenic mouse line, which expresses green fluorescent protein-tagged gephyrin under the control of the Thy1.2 promoter. Fluorescence recovery after photobleaching revealed a developmental stabilization of postsynaptic gephyrin clusters concomitant with an increase in cluster size and synaptic strength between 1 and 4 weeks in vitro. Prolonged treatment of the slice cultures with diazepam or a GABAA receptor antagonist disclosed a homeostatic regulation of both inhibitory synaptic strength and gephyrin cluster size and stability in 4-weeks-old cultures, whereas at 1 week in vitro, the same drug treatments modulated GABAergic postsynapse and gephyrin cluster properties following a Hebbian mode of synaptic plasticity. Our data are consistent with a model in which the postnatal maturation of the hippocampal network endows CA1 pyramidal neurons with the ability to homeostatically adjust the strength of their inhibitory postsynapses to afferent GABAergic drive by regulating gephyrin scaffold properties.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/physiology , Membrane Proteins/metabolism , Miniature Postsynaptic Potentials , Receptors, GABA/metabolism , Synapses/physiology , Animals , Cells, Cultured , Homeostasis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity , Synapses/drug effects , gamma-Aminobutyric Acid/pharmacology
12.
J Pharmacol Exp Ther ; 344(2): 489-500, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230213

ABSTRACT

Ethanol is a widely used drug, yet an understanding of its sites and mechanisms of action remains incomplete. Among the protein targets of ethanol are glycine receptors (GlyRs), which are potentiated by millimolar concentrations of ethanol. In addition, zinc ions also modulate GlyR function, and recent evidence suggests that physiologic concentrations of zinc enhance ethanol potentiation of GlyRs. Here, we first built a homology model of a zinc-bound GlyR using the D80 position as a coordination site for a zinc ion. Next, we investigated in vitro the effects of zinc on ethanol action at recombinant wild-type (WT) and mutant α1 GlyRs containing the D80A substitution, which eliminates zinc potentiation. At D80A GlyRs, the effects of 50 and 200 mM ethanol were reduced as compared with WT receptors. Also, in contrast to what was seen with WT GlyRs, neither adding nor chelating zinc changed the magnitude of ethanol enhancement of mutant D80A receptors. Next, we evaluated the in vivo effects of the D80A substitution by using heterozygous Glra1(D80A) knock-in (KI) mice. The KI mice showed decreased ethanol consumption and preference, and they displayed increased startle responses compared with their WT littermates. Other behavioral tests, including ethanol-induced motor incoordination and strychnine-induced convulsions, revealed no differences between the KI and WT mice. Together, our findings indicate that zinc is critical in determining the effects of ethanol at GlyRs and suggest that zinc binding at the D80 position may be important for mediating some of the behavioral effects of ethanol action at GlyRs.


Subject(s)
Alcohol Drinking , Behavior, Animal/drug effects , Ethanol/pharmacology , Point Mutation , Receptors, Glycine/genetics , Zinc/metabolism , Acoustic Stimulation , Alcohol Drinking/genetics , Amino Acid Substitution , Animals , Binding Sites , Electrophysiological Phenomena , Ethanol/administration & dosage , Female , Gene Knock-In Techniques , Homozygote , Male , Mice , Mice, Mutant Strains , Models, Molecular , Mutagenesis, Site-Directed , Oocytes/metabolism , Reflex, Righting/drug effects , Reflex, Righting/genetics , Reflex, Startle/drug effects , Reflex, Startle/genetics , Rotarod Performance Test , Strychnine/pharmacology , Transfection , Xenopus laevis , Zinc/pharmacology
13.
J Biol Chem ; 287(48): 40216-23, 2012 Nov 23.
Article in English | MEDLINE | ID: mdl-23038260

ABSTRACT

Strychnine-sensitive glycine receptors (GlyRs) mediate synaptic inhibition in the spinal cord, brainstem, and other regions of the mammalian central nervous system. In this minireview, we summarize our current view of the structure, ligand-binding sites, and chloride channel of these receptors and discuss recently emerging functions of distinct GlyR isoforms. GlyRs not only regulate the excitability of motor and afferent sensory neurons, including pain fibers, but also are involved in the processing of visual and auditory signals. Hence, GlyRs constitute promising targets for the development of therapeutically useful compounds.


Subject(s)
Receptors, Glycine/antagonists & inhibitors , Receptors, Glycine/metabolism , Strychnine/pharmacology , Animals , Binding Sites , Humans , Receptors, Glycine/chemistry , Receptors, Glycine/genetics , Synapses/chemistry , Synapses/genetics , Synapses/metabolism
14.
Biochem Biophys Res Commun ; 423(4): 661-6, 2012 Jul 13.
Article in English | MEDLINE | ID: mdl-22695116

ABSTRACT

Glycine serves as a neurotransmitter in spinal cord and brain stem, where it activates inhibitory glycine receptors. In addition, it serves as an essential co-agonist of excitatory N-methyl-d-aspartate receptors. In the central nervous system, extracellular glycine concentrations are regulated by two specific glycine transporters (GlyTs), GlyT1 and GlyT2. Here, we determined the relative transport activities and protein levels of GlyT1 and GlyT2 in membrane preparations from mouse brain stem and spinal cord at different developmental stages. We report that early postnatally (up to postnatal day P5) GlyT1 is the predominant transporter isoform responsible for a major fraction of the GlyT-mediated [(3)H]glycine uptake. At later stages (≥ P10), however, the transport activity and expression of GlyT2 increases, and in membrane fractions from adult mice both GlyTs contribute about equally to glycine uptake. These alterations in the activities and expression profiles of the GlyTs suggest that the contributions of GlyT1 and GlyT2 to the regulation of extracellular glycine concentrations at glycinergic synapses changes during development.


Subject(s)
Brain Stem/growth & development , Glycine Plasma Membrane Transport Proteins/biosynthesis , Glycine/metabolism , Spinal Cord/growth & development , Animals , Biological Transport , Brain Stem/metabolism , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Glycine Plasma Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Oocytes , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Sarcosine/analogs & derivatives , Sarcosine/pharmacology , Spinal Cord/metabolism , Xenopus laevis
15.
J Comp Neurol ; 520(17): 3962-81, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22592841

ABSTRACT

Inhibitory glycine receptors (GlyRs) are composed of homologous α- (α1-4) and ß-subunits. The ß-subunits (GlyRß) interact via their large cytosolic loops with the postsynaptic scaffolding protein gephyrin and are therefore considered essential for synaptic localization. In situ hybridization studies indicate a widespread distribution of GlyRß transcripts throughout the mammalian central nervous system (CNS), whereas GlyRα mRNAs and proteins display more restricted expression patterns. Here we report the generation of a monoclonal antibody that specifically recognizes rodent GlyRß (mAb-GlyRß) and does not exhibit crossreactivity with any of the GlyRα1-4 subunits. Immunostaining with this antibody revealed high densities of punctate GlyRß immunoreactivity at inhibitory synapses in mouse spinal cord, brainstem, midbrain, and olfactory bulb but not in the neocortex, cerebellum, or hippocampus. This contrasts the abundance of GlyRß transcripts in all major regions of the rodent brain and suggests that GlyRß protein levels are regulated posttranscriptionally. When mAb-GlyRß was used in double-labeling experiments with GlyRα1-, α2-, α3-, or α4-specific antibodies to examine the colocalization of GlyRß with these GlyR subunits in the mouse retina, >90% of the GlyRα1-3 clusters detected were found to be GlyRß-immunoreactive. A subset (about 50%) of the GlyRα4 puncta in the inner plexiform layer, however, was found to lack GlyRß and gephyrin immunostaining. These GlyRα4-only clusters were apposed to bassoon immunoreactivity and hence synaptically localized. Their existence points to a gephyrin-independent synaptic localization mechanism for a minor subset of GlyRs.


Subject(s)
Antibodies, Monoclonal , Brain Chemistry , Brain/metabolism , Receptors, Glycine/analysis , Spinal Cord/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Rats , Receptors, Glycine/biosynthesis , Spinal Cord/chemistry , Synapses/chemistry , Synapses/metabolism
16.
J Neurochem ; 122(1): 38-47, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22486198

ABSTRACT

Synaptic glycine receptors (GlyRs) are hetero-pentameric chloride channels composed of α and ß subunits, which are activated by agonist binding at subunit interfaces. To examine the pharmacological properties of each potential agonist-binding site, we substituted residues of the GlyR α(1) subunit by the corresponding residues of the ß subunit, as deduced from sequence alignment and homology modeling based on the recently published crystal structure of the glutamate-gated chloride channel GluCl. These exchange substitutions allowed us to reproduce the ßα, αß and ßß subunit interfaces present in synaptic heteromeric GlyRs by generating recombinant homomeric receptors. When the engineered α(1) GlyR mutants were expressed in Xenopus oocytes, all subunit interface combinations were found to form functional agonist-binding sites as revealed by voltage clamp recording. The ßß-binding site displayed the most distinct pharmacological profile towards a range of agonists and modulators tested, indicating that it might be selectively targeted to modulate the activity of synaptic GlyRs. The mutational approach described here should be generally applicable to heteromeric ligand-gated ion channels composed of homologous subunits and facilitate screening efforts aimed at targeting inter-subunit specific binding sites.


Subject(s)
Protein Subunits/metabolism , Receptors, Glycine/agonists , Receptors, Glycine/genetics , Amino Acid Sequence , Animals , Binding Sites/drug effects , Computer Simulation , Copper , Ethanol/pharmacology , Glycine Agents/pharmacology , Inhibitory Concentration 50 , Ivermectin/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Microinjections , Models, Molecular , Mutagenesis , Mutation/genetics , Nortropanes/pharmacology , Oocytes , Patch-Clamp Techniques , Protein Subunits/chemistry , Protein Subunits/genetics , Receptors, Glycine/chemistry , Receptors, Glycine/metabolism , Xenopus laevis , Zinc/pharmacology
17.
J Neurosci ; 32(10): 3321-32, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22399754

ABSTRACT

In the retina, the receptive fields (RFs) of almost all ganglion cells (GCs) are comprised of an excitatory center and a suppressive surround. The RF center arises from local excitatory bipolar cell (BC) inputs and the surround from lateral inhibitory inputs. Selective antagonists have been used to define the roles of GABA(A) and GABA(C) receptor-mediated input in RF organization. In contrast, the role of glycine receptor (GlyR) subunit-specific inhibition is less clear because the only antagonist, strychnine, blocks all GlyR subunit combinations. We used mice lacking the GlyRα2 (Glra2(-/-)) and GlyRα3 (Glra3(-/-)) subunits, or both (Glra2/3(-/-)), to explore their roles in GC RF organization. By comparing spontaneous and visually evoked responses of WT with Glra2(-/-), Glra3(-/-) and Glra2/3(-/-) ON- and OFF-center GCs, we found that both GlyRα2 and GlyRα3 modulate local RF interactions. In the On pathway, both receptors enhance the excitatory center response; however, the underlying inhibitory mechanisms differ. GlyRα2 participates in crossover inhibition, whereas GlyRα3 mediates serial inhibition. In the Off pathway, GlyRα2 plays a similar role, again using crossover inhibition and enhancing excitatory responses within the RF center. Comparisons of single and double KOs indicate that GlyRα2 and GlyRα3 inhibition are independent and additive, consistent with the finding that they use different inhibitory circuitry. These findings are the first to define GlyR subunit-specific control of visual function and GlyRα2 subunit-specific control of crossover inhibition in the retina.


Subject(s)
Evoked Potentials, Visual/genetics , Neural Inhibition/genetics , Receptors, Glycine/physiology , Retina/physiology , Visual Pathways/physiology , Animals , Evoked Potentials, Visual/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glycine/deficiency , Receptors, Glycine/genetics , Retina/metabolism , Visual Pathways/metabolism
18.
Behav Brain Res ; 226(1): 106-11, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21924294

ABSTRACT

Glycinergic synapses play a major role in shaping the activity of spinal cord neurons under normal conditions and during persistent pain. However, the role of different glycine receptor (GlyR) subtypes in pain processing has only begun to be unraveled. Here, we analysed whether the GlyR alpha2 subunit might be involved in the processing of acute or persistent pain. Real-time RT-PCR and in situ hybridization analyses revealed that GlyR alpha2 mRNA is enriched in the dorsal horn of the mouse spinal cord. Mice lacking GlyR alpha2 (Glra2(-/-) mice) demonstrated a normal nociceptive behavior in models of acute pain and after peripheral nerve injury. However, mechanical hyperalgesia induced by peripheral injection of zymosan was significantly prolonged in Glra2(-/-) mice as compared to wild-type littermates. We conclude that spinal GlyRs containing the alpha2 subunit exert a previously unrecognized role in the resolution of inflammatory pain.


Subject(s)
Hyperalgesia/genetics , Receptors, Glycine/genetics , Spinal Cord/metabolism , Zymosan/pharmacology , Animals , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Receptors, Glycine/metabolism , Synapses/metabolism
19.
Biochem Biophys Res Commun ; 412(3): 435-40, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21821005

ABSTRACT

Inhibitory glycine receptors (GlyRs) are densely packed in the postsynaptic membrane due to a high-affinity interaction of their ß-subunits with the scaffolding protein gephyrin. Here, we used an affinity-based proteomic approach to identify the trafficking proteins Vacuolar Protein Sorting 35 (Vps35) and Neurobeachin (Nbea) as novel GlyR ß-subunit (GlyRß) interacting proteins in rat brain. Recombinant Vps35 and a central fragment of Nbea bound to the large intracellular loop of GlyRß in glutathione-S-transferase pull-downs; in addition, Vps35 displayed binding to gephyrin. Immunocytochemical staining of spinal cord sections revealed Nbea immunoreactivity apposed to and colocalizing with marker proteins of inhibitory synapses. Our data are consistent with roles of Vps35 and Nbea in the retrieval and post-Golgi trafficking of synaptic GlyRs and possibly other neurotransmitter receptors.


Subject(s)
Brain/metabolism , Receptors, Glycine/metabolism , Vesicular Transport Proteins/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Golgi Apparatus/metabolism , Humans , Membrane Proteins/metabolism , Protein Transport , Proteomics/methods , Rats , Receptors, Glycine/genetics , Spinal Cord , Synapses/metabolism , Vesicular Transport Proteins/genetics
20.
Biochem Biophys Res Commun ; 410(4): 737-43, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21683688

ABSTRACT

Channelrhodopsin 2 (ChR2) is a microbial-type rhodopsin with a putative heptahelical structure that binds all-trans-retinal. Blue light illumination of ChR2 activates an intrinsic leak channel conductive for cations. Sequence comparison of ChR2 with the related ChR1 protein revealed a cluster of charged amino acids within the predicted transmembrane domain 2 (TM2), which includes glutamates E90, E97 and E101. Charge inversion substitutions of these residues significantly altered ChR2 function as revealed by two-electrode voltage-clamp recordings of light-induced currents from Xenopus laevis oocytes expressing the respective mutant proteins. Specifically, replacement of E90 by lysine or alanine resulted in differential effects on H(+)- and Na(+)-mediated currents. Our results are consistent with this glutamate side chain within the proposed TM2 contributing to ion flux through and the cation selectivity of ChR2.


Subject(s)
Rhodopsin/metabolism , Amino Acid Sequence , Animals , Cations/metabolism , Chlamydomonas reinhardtii/genetics , Chlamydomonas reinhardtii/metabolism , Glutamic Acid/chemistry , Glutamic Acid/genetics , Hydrogen-Ion Concentration , Ion Transport , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary/genetics , Rhodopsin/chemistry , Rhodopsin/genetics , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...