Elsevier

Brain Research

Volume 1463, 29 June 2012, Pages 1-20
Brain Research

Research Report
Regulation of the surface expression of α4β2δ GABAA receptors by high efficacy states

https://doi.org/10.1016/j.brainres.2012.04.047Get rights and content

Abstract

α4βδ GABAA receptors (GABARs) have low CNS expression, but their expression is increased by 48 h exposure to the neurosteroid THP (3α-OH-5α[β]-pregnan-20-one). THP also increases the efficacy of δ-containing GABARs acutely, where GABA is a partial agonist. Thus, we examined effects of THP (100 nM) and full GABA agonists at α4β2δ (gaboxadol, 10 μM, and β-alanine, 10 μM–1 mM), on surface expression of α4β2δ. To this end, we used an α4 construct tagged with a 3XFLAG (F) epitope or measured expression of native α4 and δ. HEK-293 cells or cultured hippocampal neurons were transfected with α4Fβ2δ and treated 24 h later with GABA agonists, THP, GABA plus THP or vehicle (0.01% DMSO) for 0.5 h–48 h. Immunocytochemistry was performed under both non-permeabilized and permeabilized conditions to detect surface and intracellular labeling, respectively, using confocal microscopy. The high efficacy agonists and GABA (1 or 10 μM) plus THP increased α4β2δ surface expression up to 3-fold after 48 h, an effect first seen by 0.5 h. This effect was not dependent upon the polarity of GABAergic current, although expression was increased by KCC2. Intracellular labeling was decreased while functional expression was confirmed by whole cell patch clamp recordings of responses to GABA agonists. GABA plus THP treatment did not alter the rate of receptor removal from the surface membrane, suggesting that THP-induced α4β2δ expression is likely via receptor insertion. Surface expression of α4β2δ was decreased by rottlerin (10 μM), suggesting a role for PKC-δ. These results suggest that trafficking of α4β2δ GABARs is regulated by high efficacy states.

Highlights

► Surface expression of α4FLAGβ2δ is increased by high efficacy states. ► Internalized α4FLAGβ2δ is decreased by high efficacy states. ► α4FLAGβ2δ expression is not dependent on the polarity of GABAergic current. ► α4FLAGβ2δ expression is dependent upon PKC-δ. ► THP increases α4FLAGβ2δ via increased receptor insertion rather than endocytosis.

Introduction

The ligand-gated GABAA receptor (GABAR) is responsible for most inhibition in the CNS (Olsen and Sieghart, 2009). This receptor is a pentameric membrane protein which gates a Clconductance, and is generally of the forms 2α, 2β and 1γ (Chang et al., 1990) although many subtypes exist from a pool of 6α, 3β, 3γ, δ, ε, π, ρ and θ. The remarkable diversity of the properties associated with the various receptor subtypes provides a basis for the selective expression of particular subtypes in a region or function-specific manner. In particular, receptors containing the δ subunit have relatively low expression in the CNS (Pirker et al., 2000, Wisden et al., 1992) but display a high degree of plasticity (Shen and Smith, 2009). These receptors express at extrasynaptic locations (Wei et al., 2003) and generate a tonic current (Stell and Mody, 2002) in response to ambient levels of GABA (~ 1 μM) (Wu et al., 2001). The tonic inhibitory current has been shown to generate more current (i.e., total charge transfer) than the phasic inhibitory synaptic current (Bai et al., 2000), suggesting that regulating the tonic inhibitory current may be an efficient mechanism to reduce neuronal excitability.

One trigger for altered expression of α4βδ GABAR is exposure to the neurosteroid THP (3α-OH-5[α]β-pregnan-20-one or [allo]pregnanolone), a metabolite of the ovarian steroid progesterone (Compagnone and Mellon, 2000) which can also be formed directly in hippocampal pyramidal cells from cholesterol (Agis-Balboa et al., 2006). Levels of this steroid fluctuate across the ovarian cycle, pregnancy and at the onset of puberty (Compagnone and Mellon, 2000, Shen et al., 2007) and are also increased by sustained stress (Girdler et al., 2001, Higashi et al., 2005, Purdy et al., 1991). In vivo administration of this steroid to female rats increases hippocampal expression of α4 and δ subunits by 2 to 3-fold above control levels after 48 h (Shen et al., 2005), accompanied by increases in the tonic inhibitory current. The naturally occurring fluctuations in this steroid also alter expression of α4βδ GABAR in a number of CNS sites, including CA1 hippocampus, dentate gyrus and the midbrain central grey (Lovick et al., 2005, Maguire and Mody, 2009, Maguire et al., 2005, Sanna et al., 2009, Shen et al., 2007, Shen et al., 2010). Alterations in expression of this receptor by fluctuating steroid levels, either endogenous or exogenously administered (Smith et al., 2006), can be associated with alterations in anxiety behavior, panic responses and seizure susceptibility, suggesting that this receptor may play a role in certain neuropsychological pathologies. Despite this in vivo evidence of neurosteroid regulation of α4βδ GABAR expression, however, little is known of the cellular mechanisms which underlie these changes in expression. Although recent studies have shown that brain-derived neurotrophic factor (BDNF) (Joshi and Kapur, 2009) and protein kinase C (PKC)-induced phosphorylation (Abramian et al., 2010) increase δ and α4 expression, respectively, the mechanism by which THP alters surface expression of these receptors is not known.

Recent studies have shown that GABA can increase trafficking of α1β2γ2 GABARs to the cell membrane (Eshaq et al., 2010). α4βδ GABARs have a unique pharmacological profile, however, different from α1β2γ2 GABARs. Although they have a high sensitivity to GABA (EC50 = 0.5 μM) (Brown et al., 2002), GABA is a partial agonist at these receptors (Bianchi and Macdonald, 2003, Zheleznova et al., 2008), unlike its effect at α1β2γ2 where it acts as a full agonist. However, δ-containing GABARs are the most sensitive target for THP (Belelli et al., 2002) and the related steroid THDOC (3α,21-dihydroxy-5α-pregnan-20-one) (Brown et al., 2002, Wohlfarth et al., 2002), which are positive modulators at physiological concentrations. These steroids increase receptor efficacy (Bianchi and Macdonald, 2003, Zheleznova et al., 2008), producing current greater than the maximal GABA-gated current by increasing long duration receptor channel openings. A number of high efficacy agonists for α4βδ GABARs have been reported, which include both synthetic (THIP or gaboxadol) (Brown et al., 2002) and endogenous (β-alanine (Bianchi and Macdonald, 2003) and taurine (Jia et al., 2008)) compounds. Thus, we initially tested the effect of THP in combination with GABA on cell surface expression of a FLAG-tagged α4 construct transfected with β2 and δ in HEK-293 cells and cultured hippocampal neurons. We assessed receptor trafficking by employing a high expression CMV promoter and assessed surface receptor expression under non-permeabilized conditions following expression of intracellular protein (Eshaq et al., 2010). This 3XFLAG tag on the C-terminus of α4 produces a highly visible signal when targeted with monoclonal anti-FLAG antibodies and a fluorescent secondary antibody (Hernan et al., 2000). Functional receptor expression was assessed with whole cell patch clamp recordings from transfected cells. These findings were compared with those obtained with high efficacy agonists and GABA itself in their effect on trafficking of α4βδ GABARs to the cell surface in order to determine whether steroid effects on expression of this receptor are due to increases in receptor efficacy.

Regulation of cell surface expression of α4βδ GABAR protein may either be due to an increase in receptor insertion or a reduction in receptor internalization and degradation. Recent studies have suggested that δ-containing GABARs have a greater stability in the membrane than γ2-containing GABARs, with a τ1/2 for internalization of hours versus minutes, respectively (Joshi and Kapur, 2009). Thus, regulation of receptor insertion rate may be a more likely mechanism for increasing cell surface expression. Our findings suggest that conditions which increase receptor efficacy increase expression of α4βδ GABARs, regardless of whether the steroid was present. These increases in receptor expression appear to be due to increased receptor insertion.

Section snippets

The α4(3XFLAG)β2δ GABAR displays functional expression in HEK-293 cells

We initially characterized our novel 3XFLAG-tagged α4 (α4F) subunit via electrophysiological techniques comparing the GABA responses of α4F to untagged α4 co-transfected with β2 and δ cDNAs (Fig. 1A). To this end, whole cell voltage clamp recordings were used to determine responses to GABA (0.01–100 μM) of the two constructs expressed in HEK-293 cells in the presence of 1 μM ZnCl which inhibits current from binary receptors (Meera et al., 2011). In fact, concentration–response curves for α4β2δ

Discussion

This study shows that cell surface expression of α4Fβδ GABARs is increased by GABAR modulators and agonists which increase the efficacy of this receptor. 48 h exposure of HEK-293 cells or neurons to the neurosteroid THP along with GABA or to the GABA agonists gaboxadol and β-alanine produced significant increases in expression of α4Fβ2δ GABARs, which were tightly correlated with increases in current gated by GABA agonists. Expression of native α4 and δ GABAR subunits was also increased by

HEK-293 cells

Human embryonic kidney (HEK) 293 cells (ATCC, Manassas, VA) were grown in Dulbecco's Modified Eagle's Medium (DMEM/F-12, Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum (FBS, Sigma, St. Louis, MO), penicillin (100 IU/ml) and streptomycin (100 μg/ml) (Invitrogen, Carlsbad, CA) on MatTek glass bottom dishes (MatTek Corp, Ashland, MA) at 37 °C in a humidified incubation chamber (5% CO2, 95% O2).

Neurons

Embryonic day 18 (E18) dissociated rat hippocampal cells were removed from timed

Acknowledgments

The authors thank NL Leidenheimer for the immunocytochemistry protocol, DH Smith for helpful discussions as well as Q H Gong and Y Deng (Microscopy Core of NYU Langone Medical Center, NY, NY) for helpful technical assistance. This work was supported by NIH grants DA09618 and AA12958 to SSS.

References (82)

  • S.S. Girdler et al.

    Allopregnanolone levels and reactivity to mental stress in premenstrual dysphoric disorder

    Biol. Psychiatry

    (2001)
  • M. Gschwendt et al.

    Rottlerin, a novel protein kinase inhibitor

    Biochem. Biophys. Res. Commun.

    (1994)
  • M. Gulinello et al.

    Short-term exposure to a neuroactive steroid increases α4 GABAA receptor subunit levels in association with increased anxiety

    Brain Res.

    (2001)
  • T. Higashi et al.

    Studies on neurosteroids XVII. Analysis of stress-induced changes in neurosteroid levels in rat brains using liquid chromatography-electron capture atmosperic pressure chemical ionization-mass spectrometry

    Steroids

    (2005)
  • S. Joshi et al.

    Slow intracellular accumulation of GABA(A) receptor delta subunit is modulated by brain-derived neurotrophic factor

    Neuroscience

    (2009)
  • J.T. Kittler et al.

    Analysis of GABAA receptor assembly in mammalian cell lines and hippocampal neurons using gamma 2 subunit green fluorescent protein chimeras

    Mol. Cell. Neurosci.

    (2000)
  • T.A. Lovick et al.

    Changes in GABA(A) receptor subunit expression in the midbrain during the oestrous cycle in Wistar rats

    Neuroscience

    (2005)
  • L. Ma et al.

    Transcriptional regulation of the mouse gene encoding the alpha[4] subunit of the GABA-A receptor

    J. Biol. Chem.

    (2004)
  • M.J. Mason et al.

    The interpretation of current-clamp recordings in the cell-attached patch-clamp configuration

    Biophys. J.

    (2005)
  • D.A. Mathers et al.

    Effects of the beta-amino acid antagonist TAG on thalamocortical inhibition

    Neuropharmacology

    (2009)
  • Z. Mtchedlishvili et al.

    Increase of GABA-A receptor-mediated tonic inhibition in dentate granule cells after traumatic brain injury

    Neurobiol. Dis.

    (2010)
  • R.W. Olsen et al.

    GABA A receptors: subtypes provide diversity of function and pharmacology

    Neuropharmacology

    (2009)
  • K.L. Perkins

    Cell-attached voltage-clamp and current-clamp recording and stimulation techniques in brain slices

    J. Neurosci. Methods

    (2006)
  • S. Pirker et al.

    GABA(A) receptors: immunocytochemical distribution of 13 subunits in the adult rat brain

    Neuroscience

    (2000)
  • S.A. Pless et al.

    Magnitude of a conformational change in the glycine receptor â1-â2 loop is correlated with agonist efficacy

    J. Biol. Chem.

    (2009)
  • D.S. Roberts et al.

    Brain-derived neurotrophic factor (BDNF)-induced synthesis of early growth response factor 3 (Egr3) controls the levels of type A GABA receptor alpha4 subunits in hippocampal neurons

    J. Biol. Chem.

    (2006)
  • F. Sancar et al.

    Allosteric modulators induce distinct movements at the GABA-binding site interface of the GABA-A receptor

    Neuropharmacology

    (2011)
  • V. Santhakumar et al.

    Developmental regulation and neuroprotective effects of striatal tonic GABA-A currents

    Neuroscience

    (2010)
  • H. Shen et al.

    Short-term steroid treatment increases delta GABA-A receptor subunit expression in rat CA1 hippocampus: pharmacological and behavioral effects

    Neuropharmacology

    (2005)
  • S.P. Soltoff

    Rottlerin: an inappropriate and ineffective inhibitor of PKCdelta

    Trends Pharmacol. Sci.

    (2007)
  • K.E. Tiedje et al.

    Beta-alanine as a small molecule neurotransmitter

    Neurochem. Int.

    (2010)
  • X. Zhou et al.

    Steroid requirements for regulation of the alpha-4 subunit of the GABA-A receptor in an in vitro model

    Neurosci. Lett.

    (2007)
  • X. Zhou et al.

    Expression levels of the alpha4 subunit of the GABA(A) receptor in differentiated neuroblastoma cells are correlated with GABA-gated current

    Neuropharmacology

    (2009)
  • A.M. Abramian et al.

    Protein kinase C phosphorylation regulates membrane insertion of GABAA receptor subtypes that mediate tonic inhibition

    J. Biol. Chem.

    (2010)
  • R.C. Agis-Balboa et al.

    Characterization of brain neurons that express enzymes mediating neurosteroid biosynthesis

    Proc. Natl. Acad. Sci.

    (2006)
  • D. Bai et al.

    Distinct functional and pharmacological properties of tonic and quantal inhibitory postsynaptic currents mediated by gamma-aminobutyric acid(A) receptors in hippocampal neurons

    Mol. Pharmacol.

    (2000)
  • M.T. Bianchi et al.

    Neurosteroids shift partial agonist activation of GABA(A) receptor channels from low- to high-efficacy gating patterns

    J. Neurosci.

    (2003)
  • N. Brown et al.

    Pharmacological characterization of a novel cell line expressing human α4β3δ GABA(A) receptors

    Br. J. Pharmacol.

    (2002)
  • Y. Chang et al.

    Stoichiometry of a recombinant GABA-A receptor

    J. Neurosci.

    (1990)
  • X. Chen et al.

    Inhibition of a background potassium channel by Gq protein alpha-subunits

    Proc. Natl. Acad. Sci.

    (2006)
  • D.S. Choi

    Protein kinase C delta regulates ethanol intoxication and enhancement of GABA-stimulated tonic current

    J. Neurosci.

    (2008)
  • Cited by (25)

    • Role of α4-containing GABA<inf>A</inf> receptors in limiting synaptic plasticity and spatial learning of female mice during the pubertal period

      2017, Brain Research
      Citation Excerpt :

      However, these receptors exhibit a high degree of plasticity. Their expression is influenced by ovarian and stress steroids (Kuver et al., 2012; Sabaliauskas et al., 2014; Shen et al., 2005), and are increased by up to 8-fold at the onset of puberty in the female mouse (Shen et al., 2010). At puberty, α4βδ GABARs express on the dendritic shaft and spines, for a period of about 10 days before declining to low levels in adulthood (Aoki et al., 2012; Shen et al., 2010).

    • Flumazenil decreases surface expression of α4β2δ GABA<inf>A</inf> receptors by increasing the rate of receptor internalization

      2016, Brain Research Bulletin
      Citation Excerpt :

      Dynasore acts immediately to block the dynamin protein (Macia et al., 2006), so an exposure time of 30 min has been used in internalization studies of membrane receptors (Oh et al., 2012). Cells were treated with dynasore during the final 30 min of a 48 h exposure of transfected HEK-293 cells to GABA plus THP with or without flumazenil, because GABA plus THP can increase α4β2δ surface expression within 20 min (Kuver et al., 2012). In fact, 30 min of dynasore treatment prevented the decrease in cell surface expression of α4Fβ2δ produced by flumazenil on GABA plus THP-treated cells (Fig. 5).

    View all citing articles on Scopus
    View full text