Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 171(4): 985-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24206220

ABSTRACT

BACKGROUND AND PURPOSE: Although the stoichiometry of the major synaptic αßγ subunit-containing GABAA receptors has consensus support for 2α:2ß:1γ, a clear view of the stoichiometry of extrasynaptic receptors containing δ subunits has remained elusive. Here we examine the subunit stoichiometry of recombinant α4ß3δ receptors using a reporter mutation and a functional electrophysiological approach. EXPERIMENTAL APPROACH: Using site-directed mutagenesis, we inserted a highly characterized 9' serine to leucine mutation into the second transmembrane (M2) region of α4, ß3 and δ subunits that increases receptor sensitivity to GABA. Whole-cell, GABA-activated currents were recorded from HEK-293 cells co-expressing different combinations of wild-type (WT) and/or mutant α4(L297S), ß3(L284S) and δ(L288S) subunits. KEY RESULTS: Recombinant receptors containing one or more mutant subunits showed increased GABA sensitivity relative to WT receptors by approximately fourfold, independent of the subunit class (α, ß or δ) carrying the mutation. GABA dose-response curves of cells co-expressing WT subunits with their respective L9'S mutants exhibited multiple components, with the number of discernible components enabling a subunit stoichiometry of 2α, 2ß and 1δ to be deduced for α4ß3δ receptors. Varying the cDNA transfection ratio by 10-fold had no significant effect on the number of incorporated δ subunits. CONCLUSIONS AND IMPLICATIONS: Subunit stoichiometry is an important determinant of GABAA receptor function and pharmacology, and δ subunit-containing receptors are important mediators of tonic inhibition in several brain regions. Here we demonstrate a preferred subunit stoichiometry for α4ß3δ receptors of 2α, 2ß and 1δ.


Subject(s)
Protein Subunits/physiology , Receptors, GABA-A/physiology , Amino Acid Sequence , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Protein Subunits/chemistry , Receptors, GABA-A/chemistry , Sequence Alignment , gamma-Aminobutyric Acid/pharmacology
2.
Schmerz ; 25(3): 272-81, 2011 Jun.
Article in German | MEDLINE | ID: mdl-21499860

ABSTRACT

To control the breathing rhythm the medullary respiratory network generates periodic salvo activities for inspiration, post-inspiration and expiration. These are under permanent modulatory control by serotonergic neurons of the raphe which governs the degree of phosphorylation of the inhibitory glycine receptor α3. The specific activation of serotonin receptor type 1A (5-HTR(1A)), which is strongly expressed in the respiratory neurons, functions via inhibition of adenylate cyclase and the resulting reduction of the intracellular cAMP level and a gradual dephosphorylation of the glycine receptor type α3 (GlyRα3). This 5-HTR(1A)-GlyRα3 signal pathway is independent of the µ-opioidergic transduction pathway and via a synaptic inhibition caused by an increase in GlyRα3 stimulates a disinhibition of some target neurons not only from excitatory but also from inhibitory neurons. Our physiological investigations show that this 5-HTR(1A)-GlyRα3 modulation allows treatment of respiratory depression due to opioids without affecting the desired analgesic effects of opioids. The molecular mechanism presented here opens new pharmacological possibilities to treat opioid-induced respiratory depression and respiratory disorders due to disturbed inhibitory synaptic transmission, such as hyperekplexia.


Subject(s)
Analgesics, Opioid/toxicity , Exhalation/physiology , Fentanyl/toxicity , Inhalation/physiology , Medulla Oblongata/physiopathology , Pain Threshold/drug effects , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT1A/physiology , Receptors, Glycine/physiology , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/physiopathology , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/physiology , Analgesics, Opioid/administration & dosage , Animals , Buspirone/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Fentanyl/administration & dosage , In Vitro Techniques , Interneurons/drug effects , Interneurons/physiology , Male , Medulla Oblongata/drug effects , Mice , Mice, Inbred C57BL , Nerve Net/drug effects , Nerve Net/physiopathology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Neurons/physiology , Nociceptors/drug effects , Nociceptors/physiology , Pain Threshold/physiology , Pentobarbital/administration & dosage , Pentobarbital/toxicity , Phosphorylation/physiology , Premedication , Raphe Nuclei/drug effects , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin Receptor Agonists/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
3.
Eur J Neurosci ; 24(9): 2504-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100839

ABSTRACT

The gamma-aminobutyric acid type A (GABA(A)) receptor is a pentameric ligand-gated ion channel responsible for fast synaptic inhibition in the brain. Phosphorylation of the GABA(A) receptor by serine/threonine protein kinases, at residues located in the intracellular loop between the third and fourth transmembrane domains of each subunit, can dynamically modulate receptor trafficking and function. In this study, we have assessed the effect that Ca(2+)-calmodulin-dependent protein kinase-II (CaMK-II) has on GABA(A) receptors. The intracellular application of preactivated CaMK-II failed to modulate the function of alphabeta and alphabetagamma subunit GABA(A) receptors heterologously expressed in human embryonic kidney (HEK)293 cells. However, application of similarly preactivated alpha-CaMK-II significantly potentiated the amplitudes of whole-cell GABA currents recorded from rat cultured cerebellar granule neurons and from recombinant GABA(A) receptors expressed in neuroblastoma, NG108-15, cells. The modulation by alpha-CaMK-II of current amplitude depended upon the subunit composition of GABA(A) receptors. alpha-CaMK-II potentiated GABA currents recorded from alpha1beta3 and alpha1beta3gamma2 GABA(A) receptors, but was unable to functionally modulate beta2 subunit-containing receptors. Similar results were obtained from beta2 -/- mouse cerebellar granule cell cultures and from rat granule cell cultures overexpressing recombinant alpha1beta2 or alpha1beta3 GABA(A) receptors. alpha-CaMK-II had a greater effect on the modulation of GABA responses mediated by alpha1beta3gamma2 compared with alpha1beta3 receptors, indicating a possible role for the gamma2 subunit in CaMK-II-mediated phosphorylation. In conclusion, CaMK-II can upregulate the function of GABA(A) receptors expressed in neurons or a neuronal cell line that is dependent on the beta subunit co-assembled into the receptor complex.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cerebellum/metabolism , Neurons/metabolism , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Cells, Cultured , Humans , Immunohistochemistry , Membrane Potentials/physiology , Mice , Mice, Knockout , Patch-Clamp Techniques , Phosphorylation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
4.
Br J Pharmacol ; 147 Suppl 1: S109-19, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16402094

ABSTRACT

gamma-Aminobutyric acid (GABA) emerged as a potentially important brain chemical just over 50 years ago, but its significance as a neurotransmitter was not fully realized until over 16 years later. We now know that at least 40% of inhibitory synaptic processing in the mammalian brain uses GABA. Establishing its role as a transmitter was a lengthy process and it seems hard to believe with our current knowledge that there was ever any dispute about its role in the mammalian brain. The detailed information that we now have about the receptors for GABA together with the wealth of agents which facilitate or reduce GABA receptor mechanisms make the prospects for further research very exciting. The emergence of glycine as a transmitter seems relatively painless by comparison to GABA. Perhaps this is appropriate for the simplest of transmitter structures! Its discovery within the spinal cord and brainstem approximately 40 years ago was followed only 2 years later by the proposal that it be conferred with 'neurotransmitter' status. It was another 16 years before the receptor was biochemically isolated. Now it is readily accepted as a vital spinal and supraspinal inhibitory transmitter and we know many details regarding its molecular structure and trafficking around neurones. The pharmacology of these receptors has lagged behind that of GABA. There is not the rich variety of allosteric modulators that we have come to readily associate with GABA receptors and which has provided us with a virtual treasure trove of important drugs used in anxiety, insomnia, epilepsy, anaesthesia, and spasticity, all stemming from the actions of the simple neutral amino acid GABA. Nevertheless, the realization that glycine receptors are involved in motor reflexes and nociceptive pathways together with the more recent advent of drugs that exhibit some subtype selectivity make the goal of designing selective therapeutic ligands for the glycine receptor that much closer.


Subject(s)
Glycine/history , Neurotransmitter Agents/history , gamma-Aminobutyric Acid/history , Animals , Glycine/physiology , History, 20th Century , History, 21st Century , Humans , Ligands , Neurotransmitter Agents/physiology , Receptors, GABA/chemistry , Receptors, GABA/drug effects , Receptors, GABA/history , Receptors, Glycine/chemistry , Receptors, Glycine/drug effects , Receptors, Glycine/history , gamma-Aminobutyric Acid/physiology
5.
Neuropharmacology ; 43(6): 938-44, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12423663

ABSTRACT

Redox reagents are thought to modulate gamma-Aminobutyric acid type A (GABA(A)) receptors by regulating the redox state of the N-terminal disulphide bridge. Examining the redox sensitivity of recombinant GABA(A) receptors in human embryonic kidney cells, using whole-cell patch clamp techniques, revealed that alpha1beta2(H267A) and alpha1beta2gamma2 receptors, which are both less sensitive to Zn(2+) and H(+) modulation, ablated the potentiating effect of the reducing agent, dithiothreitol (DTT) seen for alpha1beta2 receptors. This effect could result from disruption to the redox signal transduction pathway or be due to DTT chelating Zn(2+) from its H267 inhibitory binding site, consequently potentiating GABA-activated currents in alpha1beta2 but not alpha1beta2(H267A) or alpha1beta2gamma2 receptors. A Zn(2+) chelating agent, tricine, potentiated GABA currents for the alphabeta constructs and vertically displaced GABA dose-response curves, suggesting that these receptors are subject to some inhibition by basal Zn(2+). Tricine, did not affect the GABA currents of either alpha1beta2(H267A) or alpha1beta2gamma2 receptors but did prevent the potentiation by 2 mM DTT and reduced the potentiation caused by 10 mM DTT on alpha1beta2 receptors. Thus, at low concentrations of DTT, a substantial component of the potentiation probably occurs via Zn(2+) chelation from H267 in the ion channel. In contrast, at higher DTT concentrations, it is more likely to be acting as a redox agent, which modulates both alphabeta and alphabetagamma subunit receptors.


Subject(s)
Dithiothreitol/pharmacology , Glycine/analogs & derivatives , Receptors, GABA-A/metabolism , Zinc/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Allosteric Regulation/physiology , Animals , Cell Culture Techniques , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Glycine/pharmacology , Humans , Membrane Potentials , Mice , Mutagenesis, Site-Directed , Oxidation-Reduction/drug effects , Patch-Clamp Techniques , Receptors, GABA-A/classification , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
6.
Nat Neurosci ; 5(5): 415-24, 2002 May.
Article in English | MEDLINE | ID: mdl-11976702

ABSTRACT

GABA (gamma-aminobutyric acid)(B) receptors are heterodimeric G protein-coupled receptors that mediate slow synaptic inhibition in the central nervous system. Here we show that the functional coupling of GABA(B)R1/GABA(B)R2 receptors to inwardly rectifying K(+) channels rapidly desensitizes. This effect is alleviated after direct phosphorylation of a single serine residue (Ser892) in the cytoplasmic tail of GABA(B)R2 by cyclic AMP (cAMP)-dependent protein kinase (PKA). Basal phosphorylation of this residue is evident in rat brain membranes and in cultured neurons. Phosphorylation of Ser892 is modulated positively by pathways that elevate cAMP concentration, such as those involving forskolin and beta-adrenergic receptors. GABA(B) receptor agonists reduce receptor phosphorylation, which is consistent with PKA functioning in the control of GABA(B)-activated currents. Mechanistically, phosphorylation of Ser892 specifically enhances the membrane stability of GABA(B) receptors. We conclude that signaling pathways that activate PKA may have profound effects on GABA(B) receptor-mediated synaptic inhibition. These results also challenge the accepted view that phosphorylation is a universal negative modulator of G protein-coupled receptors.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Receptors, GABA-B/metabolism , Animals , Brain/metabolism , Brain Chemistry , CHO Cells , COS Cells , Cell Membrane/chemistry , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , GABA Agonists/pharmacology , GABA-B Receptor Agonists , Humans , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Phosphoproteins/agonists , Phosphoproteins/analysis , Phosphoproteins/metabolism , Phosphorylation , Potassium Channels/metabolism , Protein Isoforms/agonists , Protein Isoforms/analysis , Protein Isoforms/metabolism , Rats , Receptors, GABA-B/analysis , Recombinant Proteins/metabolism , Signal Transduction/physiology
7.
Neuropharmacology ; 41(6): 745-52, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11640929

ABSTRACT

GABA(A) receptors are the major sites of fast synaptic inhibition in the brain, where they are predominantly composed of alpha, beta and gamma2 subunits. A role for direct tyrosine phosphorylation of residues 365 and 367 (Y365/367) within the intracellular domain of the gamma2 subunit has been suggested to be important in modulating GABA(A) receptor function, based on the study of recombinant receptors. To address the relevance of these observations for neuronal GABA(A) receptors we have studied the phosphorylation of the gamma2 subunit in the brain. In adult rat brain the gamma2 subunit is phosphorylated on tyrosine residues, including Y365/367 as defined using a phosphospecific antisera. In cultured cortical neurones, phosphorylation of Y365/367 is highly regulated and was only evident upon inhibition of tyrosine phosphatases. We also establish that the tyrosine kinase Src is capable of specifically interacting with the intracellular domains of receptor beta and gamma2 subunits. This may specifically localise tyrosine kinase activity to GABA(A) receptors, facilitating rapid receptor tyrosine phosphorylation upon kinase activation. Together our results suggests that tyrosine phosphorylation of the gamma2 subunit, possibly by closely associated Src, may be a dynamic mechanism for regulating GABA(A) receptor function in the brain.


Subject(s)
Brain/metabolism , Receptors, GABA-A/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Antibody Specificity , Cell Line , Cells, Cultured , Cerebral Cortex/metabolism , Embryo, Mammalian , Guinea Pigs , Humans , Immune Sera/biosynthesis , Intracellular Fluid/metabolism , Mice , Molecular Sequence Data , Neurons/metabolism , Phosphoproteins/immunology , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/immunology , Tyrosine/immunology , src-Family Kinases/metabolism
8.
Nat Neurosci ; 4(9): 908-16, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11528422

ABSTRACT

Controlling the number of functional gamma-aminobutyric acid A (GABA(A)) receptors in neuronal membranes is a crucial factor for the efficacy of inhibitory neurotransmission. Here we describe the direct interaction of GABA(A) receptors with the ubiquitin-like protein Plic-1. Furthermore, Plic-1 is enriched at inhibitory synapses and is associated with subsynaptic membranes. Functionally, Plic-1 facilitates GABA(A) receptor cell surface expression without affecting the rate of receptor internalization. Plic-1 also enhances the stability of intracellular GABA(A) receptor subunits, increasing the number of receptors available for insertion into the plasma membrane. Our study identifies a previously unknown role for Plic-1, a modulation of GABA(A) receptor cell surface number, which suggests that Plic-1 facilitates accumulation of these receptors in dendritic membranes.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , Receptors, GABA-A/metabolism , Ubiquitins/physiology , Adaptor Proteins, Signal Transducing , Animals , Autophagy-Related Proteins , Cell Membrane/metabolism , Drug Stability , Protein Isoforms/metabolism , Rats , Subcellular Fractions/metabolism , Tissue Distribution , Ubiquitins/metabolism
9.
Nat Rev Neurosci ; 2(4): 240-50, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283747

ABSTRACT

Control of nerve-cell excitability is crucial for normal brain function. Two main groups of inhibitory neurotransmitter receptors--GABA(A) and glycine receptors--fulfil a significant part of this role. To mediate fast synaptic inhibition effectively, these receptors need to be localized and affixed opposite nerve terminals that release the appropriate neurotransmitter at multiple sites on postsynaptic neurons. But for this to occur, neurons require intracellular anchoring molecules, as well as mechanisms that ensure the efficient turnover and transport of mature, functional inhibitory synaptic receptor proteins. This review describes the dynamic regulation of synaptic GABA(A) and glycine receptors and discusses recent advances in this rapidly evolving field.


Subject(s)
Receptors, GABA-A/metabolism , Receptors, Glycine/metabolism , Synapses/metabolism , Animals , Carrier Proteins/metabolism , Humans , Ion Channel Gating , Membrane Proteins/metabolism , Protein Binding , Protein Transport , Synapses/chemistry
10.
J Physiol ; 530(Pt 2): 219-33, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11208970

ABSTRACT

The effect of GABAA receptor development in culture on the modulation of GABA-induced currents by external H+ was examined in cerebellar granule cells using whole-cell and single-channel recording. Equilibrium concentration-response curves revealed a lower potency for GABA between 11 and 12 days in vitro (DIV) resulting in a shift of the EC50 from 10.7 to 2.4 uM. For granule cells before 11 DIV, the peak GABA-activated current was inhibited at low external pH and enhanced at high pH with a pKa of 6.6. For the steady-state response, low pH was inhibitory with a pKa of 5.56. After 11 DIV, the peak GABA-activated current was largely pH insensitive; however, the steady-state current was potentiated at low pH with a pKa of 6.84. Single GABA-activated ion channels were recorded from outside-out patches of granule cell bodies. At pH 5.4-9.4, single GABA channels exhibited multiple conductance states occurring at 22-26, 16-17 and 12-14 pS. The conductance levels were not significantly altered over the time period of study, nor by changing the external H+ concentration. Two exponential functions were required to fit the open-time frequency histograms at both early (< 11 DIV) and late (> 11 DIV) development times at each H+ concentration. The short and long open time constants were unaffected either by the extracellular H+ concentration or by neuronal development. The distribution of all shut times was fitted by the sum of three exponentials designated as short, intermediate and long. At acidic pH, the long shut time constant decreased with development as did the relative contribution of these components to the overall distribution. This was concurrent with an increase in the mean probability of channel opening. In conclusion, this study demonstrates in cerebellar granule cells that external pH can either reduce, have no effect on, or enhance GABA-activated responses depending on the stage of development, possibly related to the subunit composition of the GABAA receptors. The mode of interaction of H+ at the single-channel level and implications of such interactions at cerebellar granule cell GABAA receptors are discussed.


Subject(s)
Cerebellum/metabolism , Neurons/physiology , Receptors, GABA-A/physiology , Animals , Cerebellum/cytology , Electric Conductivity , Electrophysiology , Ion Channels/metabolism , Protons , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/drug effects , Time Factors , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology
11.
Mol Cell Neurosci ; 16(4): 440-52, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11085880

ABSTRACT

Type A gamma-aminobutyric acid receptors (GABAA), the major sites of fast synaptic inhibition in the brain, are believed to be predominantly composed of alpha, beta, and gamma subunits. To examine the membrane trafficking of GABAA receptors we have produced gamma 2L subunit chimeras with green fluorescent protein (GFP). Addition of GFP to the N-terminus of the gamma 2 subunit (gamma 2L-GFPN) was functionally silent for alpha 1 beta 2 gamma 2L-GFPN receptors expressed in A293 cells. Furthermore, this chimera allowed the visualization of receptor membrane targeting and endocytosis in live cells. In contrast, incorporation of GFP at the C-terminus reduced subunit stability, impairing assembly with receptor alpha and beta subunits. Using gamma 2L-GFPN we were able to demonstrate that targeting of the gamma 2 subunit to GABAergic synapses in hippocampal neurons was dependent upon coassembly with receptor alpha and beta subunits. Together our results demonstrate that the assembly and membrane targeting of GABAA receptors composed of alpha 1 beta 2 gamma 2L-GFPN subunits follow similar itineraries in heterologous systems and neurons.


Subject(s)
Hippocampus/cytology , Neurons/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Animals , Cell Line , Cell Membrane/enzymology , Endocytosis/physiology , Gene Expression/physiology , Genes, Reporter , Green Fluorescent Proteins , Humans , Indicators and Reagents/metabolism , Ion Channel Gating/physiology , Kidney/cytology , Ligands , Luminescent Proteins/genetics , Mammals , Mice , Neurons/cytology , Protein Kinase C/metabolism , Protein Structure, Tertiary , Receptors, GABA-A/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synapses/metabolism
12.
J Neurosci ; 20(21): 7972-7, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11050117

ABSTRACT

Type A GABA receptors (GABA(A)) mediate the majority of fast synaptic inhibition in the brain and are believed to be predominantly composed of alpha, beta, and gamma subunits. Although changes in cell surface GABA(A) receptor number have been postulated to be of importance in modulating inhibitory synaptic transmission, little is currently known on the mechanism used by neurons to modify surface receptor levels at inhibitory synapses. To address this issue, we have studied the cell surface expression and maintenance of GABA(A) receptors. Here we show that constitutive internalization of GABA(A) receptors in hippocampal neurons and recombinant receptors expressed in A293 cells is mediated by clathrin-dependent endocytosis. Furthermore, we identify an interaction between the GABA(A) receptor beta and gamma subunits with the adaptin complex AP2, which is critical for the recruitment of integral membrane proteins into clathrin-coated pits. GABA(A) receptors also colocalize with AP2 in cultured hippocampal neurons. Finally, blocking clathrin-dependant endocytosis with a peptide that disrupts the association between amphiphysin and dynamin causes a large sustained increase in the amplitude of miniature IPSCs in cultured hippocampal neurons. These results suggest that GABA(A) receptors cycle between the synaptic membrane and intracellular sites, and their association with AP2 followed by recruitment into clathrin-coated pits represents an important mechanism in the postsynaptic modulation of inhibitory synaptic transmission.


Subject(s)
Endocytosis/physiology , Membrane Proteins/metabolism , Neurons/metabolism , Receptors, GABA-A/metabolism , Synaptic Transmission/physiology , Adaptor Protein Complex 2 , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Animals , Cells, Cultured , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Dynamins , Endocytosis/drug effects , Fluorescent Antibody Technique , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/metabolism , Glutathione Transferase/genetics , Hippocampus/cytology , Hippocampus/metabolism , Mice , Neural Inhibition/drug effects , Neurons/cytology , Patch-Clamp Techniques , Peptides/pharmacology , Precipitin Tests , Protein Kinase C/metabolism , Receptors, GABA-A/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
13.
J Biol Chem ; 275(49): 38856-62, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-10978327

ABSTRACT

GABA(A) receptors are critical mediators of fast synaptic inhibition in the brain, and the predominant receptor subtype in the central nervous system is believed to be a pentamer composed of alpha, beta, and gamma subunits. Previous studies on recombinant receptors have shown that protein kinase C (PKC) and PKA directly phosphorylate intracellular serine residues within the receptor beta subunit and modulate receptor function. However, the relevance of this regulation for neuronal receptors remains poorly characterized. To address this critical issue, we have studied phosphorylation and functional modulation of GABA(A) receptors in cultured cortical neurons. Here we show that the neuronal beta3 subunit is basally phosphorylated on serine residues by a PKC-dependent pathway. PKC inhibitors abolish basal phosphorylation, increasing receptor activity, whereas activators of PKC enhance beta3 phosphorylation with a concomitant decrease in receptor activity. PKA activators were shown to increase the phosphorylation of the beta3 subunit only in the presence of PKC inhibitors. We also show that the main sites of phosphorylation within the neuronal beta3 subunit are likely to include Ser-408 and Ser-409, residues that are important for the functional modulation of beta3-containing recombinant receptors. Furthermore, PKC activation did not change the total number of GABA(A) receptors in the plasma membrane, suggesting that the effects of PKC activation are on the gating or conductance of the channel. Together, these results illustrate that cell-signaling pathways that activate PKC may have profound effects on the efficacy of synaptic inhibition by directly modulating GABA(A) receptor function.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Protein Kinases/metabolism , Receptors, GABA-A/physiology , Signal Transduction/physiology , Animals , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Embryo, Mammalian , Humans , Kinetics , Naphthalenes/pharmacology , Neurons/cytology , Neurons/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Protein Subunits , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Receptors, GABA-A/metabolism , Recombinant Proteins/metabolism , Serine , Transfection
14.
J Neurosci ; 20(4): 1297-306, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10662819

ABSTRACT

GABA(A) receptors can be constructed from a range of differing subunit isoforms: alpha, beta, gamma, delta, and epsilon. Expression studies have revealed that production of GABA-gated channels is achieved after coexpression of alpha and beta subunits. The expression of a gamma subunit isoform is essential to confer benzodiazepine sensitivity on the expressed receptor. However, how the specificity of subunit interactions is controlled during receptor assembly remains unknown. Here we demonstrate that residues 58-67 within alpha subunit isoforms are important in the assembly of receptors comprised of alphabeta and alphabetagamma subunits. Deletion of these residues from the alpha1 or alpha6 subunits results in retention of either alpha subunit isoform in the endoplasmic reticulum on coexpression with the beta3, or beta3 and gamma2 subunits. Immunoprecipitation revealed that residues 58-67 mediated oligomerization of the alpha1 and beta3 subunits, but were without affect on the production of alpha/gamma complexes. Within this domain, glutamine 67 was of central importance in mediating the production of functional alpha1beta3 receptors. Mutation of this residue resulted in a drastic decrease in the cell surface expression of alpha1beta3 receptors and the resulting expression of beta3 homomers. Sucrose density gradient centrifugation revealed that this residue was important for the production of a 9S alpha1beta3 complex representing functional GABA(A) receptors. Therefore, our studies detail residues that specify GABA(A) receptor alphabeta subunit interactions. This domain, which is conserved in all alpha subunit isoforms, will therefore play a critical role in the assembly of GABA(A) receptors composed of alphabeta and alphabetagamma subunits.


Subject(s)
Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Amino Acid Sequence , Amino Acid Substitution , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Kidney , Macromolecular Substances , Mutagenesis, Site-Directed , Pentobarbital/pharmacology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, GABA-A/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Transfection , Tumor Cells, Cultured , gamma-Aminobutyric Acid/pharmacology
15.
J Biol Chem ; 274(51): 36565-72, 1999 Dec 17.
Article in English | MEDLINE | ID: mdl-10593956

ABSTRACT

Type A gamma-aminobutyric acid receptors (GABA(A)), the major sites of fast synaptic inhibition in the brain, are believed to be composed predominantly of alpha, beta, and gamma subunits. Although cell surface expression is essential for GABA(A) receptor function, little is known regarding its regulation. To address this issue, the membrane stability of recombinant alpha(1)beta(2) or alpha(1)beta(2)gamma(2) receptors was analyzed in human embryonic kidney cells. Alpha(1)beta(2)gamma(2) but not alpha(1)beta(2) receptors were found to recycle constitutively between the cell surface and a microtubule-dependent, perinuclear endosomal compartment. Similar GABA(A) receptor endocytosis was also seen in cultured hippocampal and cortical neurons. GABA(A) receptor surface levels were reduced upon protein kinase C (PKC) activation. Like basal endocytosis, this response required the gamma(2) subunit but not receptor phosphorylation. Although inhibiting PKC activity did not block alpha(1)beta(2)gamma(2) receptor endocytosis, it did prevent receptor down-regulation, suggesting that PKC activity may block alpha(1)beta(2)gamma(2) receptor recycling to the cell surface. In agreement with this observation, blocking recycling from endosomes with wortmannin selectively reduced surface levels of gamma(2)-containing receptors. Together, our results demonstrate that the surface stability of GABA(A) receptors can be dynamically and specifically regulated, enabling neurons to modulate cell surface receptor number upon the appropriate cues.


Subject(s)
Protein Kinase C/metabolism , Receptors, GABA-A/metabolism , Signal Transduction , Cell Line , Humans , Receptors, GABA-A/chemistry , Synaptic Transmission
16.
J Physiol ; 520 Pt 1: 53-64, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10517800

ABSTRACT

1. Whole-cell glycine-activated currents were recorded from human embryonic kidney (HEK) cells expressing wild-type and mutant recombinant homomeric glycine receptors (GlyRs) to locate the inhibitory binding site for Zn2+ ions on the human alpha1 subunit. 2. Glycine-activated currents were potentiated by low concentrations of Zn2+ (<10 microM) and inhibited by higher concentrations (>100 microM) on wild-type alpha1 subunit GlyRs. 3. Lowering the external pH from 7.4 to 5.4 inhibited the glycine responses in a competitive manner. The inhibition caused by Zn2+ was abolished leaving an overt potentiating effect at 10 microM Zn2+ that was exacerbated at 100 microM Zn2+. 4. The identification of residues involved in the formation of the inhibitory binding site was also assessed using diethylpyrocarbonate (DEPC), which modifies histidines. DEPC (1 mM) abolished Zn2+-induced inhibition and also the potentiation of glycine-activated currents by Zn2+. 5. The reduction in glycine-induced whole-cell currents in the presence of high (100 microM) concentrations of Zn2+ did not increase the rate of glycine receptor desensitisation. 6. Systematic mutation of extracellular histidine residues in the GlyR alpha1 subunit revealed that mutations H107A or H109A completely abolished inhibition of glycine-gated currents by Zn2+. However, mutation of other external histidines, H210, H215 and H419, failed to prevent inhibition by Zn2+ of glycine-gated currents. Thus, H107 and H109 in the extracellular domain of the human GlyR alpha1 subunit are major determinants of the inhibitory Zn2+ binding site. 7. An examination of Zn2+ co-ordination in metalloenzymes revealed that the histidine- hydrophobic residue-histidine motif found to be responsible for binding Zn2+ in the human GlyR alpha1 subunit is also shared by some of these enzymes. Further comparison of the structure and location of this motif with a generic model of the GlyR alpha1 subunit suggests that H107 and H109 participate in the formation of the inhibitory Zn2+ binding site at the apex of a beta sheet in the N-terminal extracellular domain.


Subject(s)
Receptors, Glycine/metabolism , Zinc/metabolism , Amino Acid Sequence , Binding Sites/genetics , Cell Line , Diethyl Pyrocarbonate/pharmacology , Electrophysiology , Extracellular Matrix/metabolism , Genetic Vectors , Glycine/physiology , Histidine/physiology , Humans , Hydrogen-Ion Concentration , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed/genetics , Mutagenesis, Site-Directed/physiology , Patch-Clamp Techniques , Receptors, Glycine/genetics
17.
Eur J Neurosci ; 11(8): 2897-905, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10457186

ABSTRACT

Mefenamic acid (MFA) has anti-convulsant and pro-convulsant effects in vivo, and has been shown to potentiate and inhibit GABAA (gamma-aminobutyric acid) receptors in vitro. In this study, whole-cell currents were recorded from Xenopus oocytes and human embryonic kidney (HEK) cells expressing human recombinant GABAA receptors to resolve the molecular mechanisms by which MFA modulates GABAA receptor function. We demonstrate that MFA potentiated GABA-activated currents for alpha1beta2 gamma2S (EC50 = 3.2 +/- 0.5 microM), but not for alpha1beta1 gamma2S receptors. MFA also enhanced GABA-activated responses and directly activated alpha1beta2/beta3 GABAA receptors, but inhibited responses to GABA on alpha1beta1 constructs (IC50 = 40 +/- 7.2 microM). A comparison of beta1, beta2 and beta3 subunits suggested that the positive modulatory action of MFA involved asparagine (N) 290 in the second transmembrane domain (TM2) of the beta2 and beta3 subunits. Mutation of N290 to serine (S) markedly reduced modulation by MFA in alpha1beta2(N290S)gamma2S receptors, whereas alpha1beta1(S290N)gamma2S constructs revealed potentiated responses to GABA (EC50 = 7.8 +/- 1.7 microM) and direct activation by MFA. The potentiation by MFA displayed voltage sensitivity. The direct activation, potentiation and inhibitory aspects of MFA action were predominantly conferred by the beta subunits as the spontaneously active homomeric beta1 and beta3 receptors were susceptible to modulation by MFA. Molecular comparisons of MFA, loreclezole and etomidate, agents which exhibit similar selectivity for GABAA receptors, revealed their ability to adopt similar structural conformations. This study indicates that N290 in TM2 of beta2 and beta3 subunits is important for the regulation of GABAA receptor function by MFA. Our data provide a potential molecular mechanism for the complex central effects of MFA in vivo.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Mefenamic Acid/pharmacology , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Amino Acid Sequence/genetics , Amino Acid Sequence/physiology , Animals , Cell Line/physiology , Electric Conductivity , Electrophysiology , Etomidate/chemistry , Humans , Mefenamic Acid/chemistry , Models, Molecular , Molecular Conformation , Oocytes/metabolism , Oocytes/physiology , Protein Isoforms/drug effects , Protein Isoforms/physiology , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Triazoles/chemistry , Triazoles/pharmacology , Xenopus laevis
18.
J Neurosci ; 19(15): 6360-71, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10414965

ABSTRACT

GABA(A) receptors are believed to be heteropentamers that can be constructed from six subunit classes: alpha(1-6), beta(1-4), gamma(1-3), delta, epsilon, and pi. Given that individual neurons often express multiple receptor subunits, it is important to understand how these receptors assemble. To determine which domains of receptor subunits control assembly, we have exploited the differing capabilities of the beta2 and beta3 subunits to form functional cell surface homomeric receptors. Using a chimeric approach, we have identified four amino acids in the N-terminal domain of the beta3 subunit that mediate functional cell surface expression of this subunit compared with beta2, which is retained within the endoplasmic reticulum. Substitution of these four amino acids-glycine 171, lysine 173, glutamate 179, and arginine 180-into the beta2 subunit was sufficient to enable the beta2 subunit to homo-oligomerize. The effect of this putative "assembly signal" on the production of heteromeric receptors composed of alphabeta and betagamma subunits was also analyzed. This signal was not critical for the formation of receptors composed of either alpha1beta2 or alpha1beta3 subunits, suggesting that mutation of these residues did not disrupt subunit folding. However, this signal was important in the formation of betagamma2 receptors. These residues did not seem to affect the initial association of beta2 and gamma2 subunits but appeared to be important for the subsequent production of functional receptors. Our studies identify, for the first time, key residues within the N-terminal domains of receptor beta subunits that mediate the selective assembly of GABA(A) receptors.


Subject(s)
Amino Acids/analysis , Amino Acids/physiology , Receptors, GABA-A/chemistry , Receptors, GABA-A/physiology , Amino Acid Sequence/genetics , Animals , Cell Line , Cell Membrane/metabolism , Centrifugation, Density Gradient , Chimera/genetics , Chimera/physiology , Chlorocebus aethiops , Humans , Ion Channels/metabolism , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism
19.
J Physiol ; 517 ( Pt 1): 35-50, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10226147

ABSTRACT

1. The functional role played by the postulated disulphide bridge in gamma-aminobutyric acid type A (GABAA) receptors and its susceptibility to oxidation and reduction were studied using recombinant (murine receptor subunits expressed in human embryonic kidney cells) and rat neuronal GABAA receptors in conjunction with whole-cell and single channel patch-clamp techniques. 2. The reducing agent dithiothreitol (DTT) reversibly potentiated GABA-activated responses (IGABA) of alpha1beta1 or alpha1beta2 receptors while the oxidizing reagent 5, 5'-dithio-bis-(2-nitrobenzoic acid) (DTNB) caused inhibition. Redox modulation of IGABA was independent of GABA concentration, membrane potential and the receptor agonist and did not affect the GABA EC50 or Hill coefficient. The endogenous antioxidant reduced glutathione (GSH) also potentiated IGABA in alpha1beta2 receptors, while both the oxidized form of DTT and glutathione (GSSG) caused small inhibitory effects. 3. Recombinant receptors composed of alpha1beta1gamma2S or alpha1beta2gamma2S were considerably less sensitive to DTT and DTNB. 4. For neuronal GABAA receptors, IGABA was enhanced by flurazepam and relatively unaffected by redox reagents. However, in cultured sympathetic neurones, nicotinic acetylcholine-activated responses were inhibited by DTT whilst in cerebellar granule neurones, NMDA-activated currents were potentiated by DTT and inhibited by DTNB. 5. Single GABA-activated ion channel currents exhibited a conductance of 16 pS for alpha1beta1 constructs. DTT did not affect the conductance or individual open time constants determined from dwell time histograms, but increased the mean open time by affecting the channel open probability without increasing the number of cell surface receptors. 6. A kinetic model of the effects of DTT and DTNB suggested that the receptor existed in equilibrium between oxidized and reduced forms. DTT increased the rate of entry into reduced receptor forms and also into desensitized states. DTNB reversed these kinetic effects. 7. Our results indicate that GABAA receptors formed by alpha and beta subunits are susceptible to regulation by redox agents. Inclusion of the gamma2 subunit in the receptor, or recording from some neuronal GABAA receptors, resulted in reduced sensitivity to DTT and DTNB. Given the suggested existence of alphabeta subunit complexes in some areas of the central nervous system together with the generation and release of endogenous redox compounds, native GABAA receptors may be subject to regulation by redox mechanisms.


Subject(s)
Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Sulfhydryl Reagents/pharmacology , Animals , Cell Line , Cells, Cultured , Dithionitrobenzoic Acid/pharmacology , Dithiothreitol/pharmacology , Humans , Kinetics , Membrane Potentials , Mice , Models, Biological , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Patch-Clamp Techniques , Protein Conformation , Rats , Receptors, GABA-A/genetics , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
20.
Mol Cell Neurosci ; 13(4): 259-71, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10328885

ABSTRACT

The expression of alpha and beta gamma-aminobutyric acid type A receptor subunits produces GABA-gated channels which require the incorporation of either the gamma2 or gamma3 subunit for benzodiazepine modulation. Here we examine the role of the gamma2 subunit splice variants, gamma2S and gamma2L which differ by eight amino acids in the major intracellular domain, in mediating cell surface expression. Using immunocytochemistry we have demonstrated that when expressed alone, the gamma2S subunit can access the cell surface and internalize constitutively. In contrast, alpha1, beta2 and gamma2L are retained predominantly in the endoplasmic reticulum (ER) when expressed alone. Replacing the insert which differentiates gamma2L from gamma2S (LLRMFSFK) with eight alanines produces a phenotype identical to gamma2S. Both gamma2 subunits fail to produce high molecular weight oligomers observed for alpha1beta2 and alpha1beta2gamma2 heterooligomers and do not form functional ion channels. Surface expression of gamma2S is repressed upon the coexpression of alpha1 or beta2 subunits, resulting in ER-retained heterooligomers, suggesting that homomeric gamma2S is unlikely to occur in vivo. However, its independent maturation to surface competence and preferential assembly with alpha and beta subunits may ensure the production of functional benzodiazepine-sensitive receptors. Furthermore, the presence of the gamma2 subunit appears to confer an endocytotic capacity to these heterooligomeric receptors.


Subject(s)
DNA, Recombinant , Endocytosis/physiology , Genetic Variation , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Subcellular Fractions/metabolism , Animals , Cell Line/metabolism , Cell Line/ultrastructure , Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Endosomes/metabolism , Humans , Immunohistochemistry , Isomerism , Mice , Microscopy, Electron , Receptors, GABA-A/metabolism , Recombinant Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...