9
Cellular/Molecular The Short Splice Variant of the 2 Subunit Acts as an External Modulator of GABA A Receptor Function Andrew J. Boileau, 1,2 Robert A. Pearce, 3 and Cynthia Czajkowski 2 1 Department of Physiology, Ponce School of Medicine, Ponce, Puerto Rico 00732, and Departments of 2 Physiology and 3 Anesthesiology, University of Wisconsin-Madison, Madison, Wisconsin 53711 GABA A receptors (GABA A Rs) regulate the majority of fast inhibition in the mammalian brain and are the target for multiple drug types, including sleep aids, anti-anxiety medication, anesthetics, alcohol, and neurosteroids. A variety of subunits, including the highly distrib- uted 2, allow for pharmacologic and kinetic differences in particular brain regions. The two common splice variants 2S (short) and 2L (long) show different patterns of regional distribution both in adult brain and during the course of development, but show few notable differences when incorporated into pentameric receptors. However, results presented here show that the 2S variant can strongly affect both GABA A R pharmacology and kinetics by acting as an external modulator of fully formed receptors. Mutation of one serine residue can confer 2S-like properties to 2L subunits, and addition of a modified 2 N-terminal polypeptide to the cell surface recapitulates the pharmacological effect. Thus, rather than incorporation of a separate accessory protein as with voltage-gated channels, this is an example of an ion channel using a common subunit for dual purposes. The modified receptor properties conferred by accessory 2S have implications for understanding GABA A R pharmacology, receptor kinetics, stoichiometry, GABAergic signaling in the brain during development, and altered function in disease states such as epilepsy. Introduction Exquisite and precise signaling control in a healthy brain is achieved via complex network circuitry composed of excitatory and inhibitory neuronal connections. The majority of inhibitory signals are mediated by a Cl conductance through ionotropic GABA A receptors (GABA A Rs). Despite myriad possible types of subunits and combinations, the most common GABA A R hetero- pentamers in the mammalian brain are composed of two , two , and one subunit (Akabas, 2004). GABA A Rs are also of particular interest as the targets of several classes of drugs and modulators, including benzodiazepines (BDZs), anesthetics, neurosteroids, and Zn 2 ions (Hevers and Luddens, 1998). However, different GABA A R subtypes respond to modulators with their own speci- ficity (Mohler et al., 1995), and, to control the placement of dif- ferent subtypes to their appropriate loci in the brain, elaborate (but not fully elaborated) mechanisms are in place to regulate assembly and trafficking. One element of this fine control is the use of splice variants. The 2 subunit has two forms, designated “short” (2S) and “long” (2L) that differ only in eight amino acids inserted into the large intracellular loop of the subunit (Whiting et al., 1990). The 2S variant is more prevalent in early development (Wang and Burt, 1991) and is found at higher density in the cerebral cortex, olfactory bulb, and hippocampal formation (Gutie ´rrez et al., 1994; Miralles et al., 1994). In old age and in schizophrenia, 2S levels fall (Gutie ´rrez et al., 1996; Huntsman et al., 1998). However, transgenic mice with only 2S or 2L appear thus far to have few if any clear phenotypic differences (Wick et al., 2000). In previous experiments, BDZ potentiation was found to sat- urate as the ratio of GABA A R 2 subunit was increased with fixed amounts of 1 and 2 subunits (Boileau et al., 2002) or by using subunit concatamers (Baumann et al., 2002; Boileau et al., 2005; Baur et al., 2006; Ericksen and Boileau, 2007). However, in trans- fections with lower 2S ratios that displayed minimal BDZ po- tentiation and thus a large ratio of zinc-sensitive 12 receptors, the blockade by Zn 2 ions was unexpectedly low. In the present study, a striking difference between the two splice variants 2S and 2L was found in zinc sensitivity. In mixtures of and 2L GABA A Rs, the diazepam (DZ) potentiation of submaxi- mal GABA-induced currents correlated well with zinc blockade of the mixture, whereas in mixtures of and 2S receptors, the zinc block was much less than predicted. Colocalization showed that 2S subunits express on the cell surface alone, even in the presence of and subunits. Both the anomalous zinc sensitivity and surface expression of 2S could be mimicked in 2L by a single point mutation. Finally, external addition of a 2 N-terminal polypeptide protected GABA A Rs from zinc block. The “accessory” 2S subunit also altered the kinetics of the ex- pressed receptors. Together, the data demonstrate that the 2S splice variant serves dual roles: as an integral subunit of the GABA A R and as an accessory protein acting as an external mod- ulator of GABA A R function. Received Oct. 9, 2009; revised Jan. 21, 2010; accepted Jan. 31, 2010. This work was funded by National Institutes of Health Grants MH66406 (A.J.B., C.C.) and NS34727 (C.C.). We thank the W. M. Keck Laboratory for Biological Imaging for help with confocal images, Drs. Mathew V. Jones, Eugenia M. C. Jones (University of Wisconsin-Madison, Madison, WI), Xue Han (Massachusetts Institute of Technology/ Harvard University, Cambridge, MA), and Robert L. Macdonald (Vanderbilt University, Nashville, TN) for helpful discussions, Dr. Srinivasan Venkatachalan for editing assistance, and Dr. Chenlei Leng (National University of Singa- pore, Statistics Department, Singapore) for help with statistical analysis. Correspondence should be addressed to Andrew J. Boileau, Department of Physiology and Pharmacology, Ponce School of Medicine, P.O. Box 7004, Ponce, Puerto Rico 00732. E-mail: [email protected]. DOI:10.1523/JNEUROSCI.5039-09.2010 Copyright © 2010 the authors 0270-6474/10/304895-09$15.00/0 The Journal of Neuroscience, April 7, 2010 30(14):4895– 4903 • 4895

Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

Cellular/Molecular

The Short Splice Variant of the �2 Subunit Acts as anExternal Modulator of GABAA Receptor Function

Andrew J. Boileau,1,2 Robert A. Pearce,3 and Cynthia Czajkowski2

1Department of Physiology, Ponce School of Medicine, Ponce, Puerto Rico 00732, and Departments of 2Physiology and 3Anesthesiology, University ofWisconsin-Madison, Madison, Wisconsin 53711

GABAA receptors (GABAARs) regulate the majority of fast inhibition in the mammalian brain and are the target for multiple drug types,including sleep aids, anti-anxiety medication, anesthetics, alcohol, and neurosteroids. A variety of subunits, including the highly distrib-uted �2, allow for pharmacologic and kinetic differences in particular brain regions. The two common splice variants �2S (short) and �2L(long) show different patterns of regional distribution both in adult brain and during the course of development, but show few notabledifferences when incorporated into pentameric receptors. However, results presented here show that the �2S variant can strongly affectboth GABAAR pharmacology and kinetics by acting as an external modulator of fully formed receptors. Mutation of one serine residue canconfer �2S-like properties to �2L subunits, and addition of a modified �2 N-terminal polypeptide to the cell surface recapitulates thepharmacological effect. Thus, rather than incorporation of a separate accessory protein as with voltage-gated channels, this is an exampleof an ion channel using a common subunit for dual purposes. The modified receptor properties conferred by accessory �2S haveimplications for understanding GABAAR pharmacology, receptor kinetics, stoichiometry, GABAergic signaling in the brain duringdevelopment, and altered function in disease states such as epilepsy.

IntroductionExquisite and precise signaling control in a healthy brain isachieved via complex network circuitry composed of excitatoryand inhibitory neuronal connections. The majority of inhibitorysignals are mediated by a Cl� conductance through ionotropicGABA

Areceptors (GABAARs). Despite myriad possible types of

subunits and combinations, the most common GABAAR hetero-pentamers in the mammalian brain are composed of two �, two �,and one � subunit (Akabas, 2004). GABAARs are also of particularinterest as the targets of several classes of drugs and modulators,including benzodiazepines (BDZs), anesthetics, neurosteroids,and Zn 2� ions (Hevers and Luddens, 1998). However, differentGABAAR subtypes respond to modulators with their own speci-ficity (Mohler et al., 1995), and, to control the placement of dif-ferent subtypes to their appropriate loci in the brain, elaborate(but not fully elaborated) mechanisms are in place to regulateassembly and trafficking.

One element of this fine control is the use of splice variants.The �2 subunit has two forms, designated “short” (�2S) and“long” (�2L) that differ only in eight amino acids inserted intothe large intracellular loop of the subunit (Whiting et al., 1990).

The �2S variant is more prevalent in early development (Wangand Burt, 1991) and is found at higher density in the cerebralcortex, olfactory bulb, and hippocampal formation (Gutierrez etal., 1994; Miralles et al., 1994). In old age and in schizophrenia,�2S levels fall (Gutierrez et al., 1996; Huntsman et al., 1998).However, transgenic mice with only �2S or �2L appear thus far tohave few if any clear phenotypic differences (Wick et al., 2000).

In previous experiments, BDZ potentiation was found to sat-urate as the ratio of GABAAR �2 subunit was increased with fixedamounts of �1 and �2 subunits (Boileau et al., 2002) or by usingsubunit concatamers (Baumann et al., 2002; Boileau et al., 2005;Baur et al., 2006; Ericksen and Boileau, 2007). However, in trans-fections with lower �2S ratios that displayed minimal BDZ po-tentiation and thus a large ratio of zinc-sensitive �1�2 receptors,the blockade by Zn 2� ions was unexpectedly low. In the presentstudy, a striking difference between the two splice variants �2Sand �2L was found in zinc sensitivity. In mixtures of �� and���2L GABAARs, the diazepam (DZ) potentiation of submaxi-mal GABA-induced currents correlated well with zinc blockadeof the mixture, whereas in mixtures of �� and ���2S receptors,the zinc block was much less than predicted. Colocalizationshowed that �2S subunits express on the cell surface alone, evenin the presence of � and � subunits. Both the anomalous zincsensitivity and surface expression of �2S could be mimicked in�2L by a single point mutation. Finally, external addition of a �2N-terminal polypeptide protected �� GABAARs from zinc block.The “accessory” �2S subunit also altered the kinetics of the ex-pressed receptors. Together, the data demonstrate that the �2Ssplice variant serves dual roles: as an integral subunit of theGABAAR and as an accessory protein acting as an external mod-ulator of GABAAR function.

Received Oct. 9, 2009; revised Jan. 21, 2010; accepted Jan. 31, 2010.This work was funded by National Institutes of Health Grants MH66406 (A.J.B., C.C.) and NS34727 (C.C.). We

thank the W. M. Keck Laboratory for Biological Imaging for help with confocal images, Drs. Mathew V. Jones, EugeniaM. C. Jones (University of Wisconsin-Madison, Madison, WI), Xue Han (Massachusetts Institute of Technology/Harvard University, Cambridge, MA), and Robert L. Macdonald (Vanderbilt University, Nashville, TN) for helpfuldiscussions, Dr. Srinivasan Venkatachalan for editing assistance, and Dr. Chenlei Leng (National University of Singa-pore, Statistics Department, Singapore) for help with statistical analysis.

Correspondence should be addressed to Andrew J. Boileau, Department of Physiology and Pharmacology, PonceSchool of Medicine, P.O. Box 7004, Ponce, Puerto Rico 00732. E-mail: [email protected].

DOI:10.1523/JNEUROSCI.5039-09.2010Copyright © 2010 the authors 0270-6474/10/304895-09$15.00/0

The Journal of Neuroscience, April 7, 2010 • 30(14):4895– 4903 • 4895

Page 2: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

Materials and MethodsCell culture and transfection. HEK293 cells (CRL 1573; American TypeCulture Collection) were grown on 100 or 35 mm tissue culture dishes inMinimum Essential Medium with Earle’s salts (Invitrogen) containing10% fetal bovine serum (Hyclone Laboratories) in a 37°C incubatorunder a 5% CO2 atmosphere. Transfections with cDNAs for rat GABAAR�1, �2, �2S, and �2L subunits, tandem subunits, mutant subunits, andenhanced green fluorescent protein (EGFP) in pCEP4 (Invitrogen) wereperformed as described previously (Boileau et al., 2005). For example,transfection with free subunits used 200 ng of pCEP4 –�1 plus 200 ng ofpCEP4 –�2 plus a varied weight ratio of pCEP4 –�2S, pCEP4 –�2L, orpCEP4 –�2L mutant; thus, a 1:1:10 ratio had 2000 ng of pCEP4 –�2, and�2S or �2L were used in the same amounts and yielded a similar range ofcurrent amplitudes (1–10 nA of peak current at �40 mV). Cells werecotransfected at 70 –90% confluence using Lipofectamine 2000 (Invitro-gen), washed, and replated to coverslips after 4 – 8 h.

Drug application and recording. Solutions were applied to excisedoutside-out patches using a four-barrel square glass application pipetteor theta pipette connected to a piezoelectric stacked translator (PhysikInstrumente) as described previously (Boileau et al., 2005). The voltageinput to the high-voltage amplifier (Physik Instrumente) driving thestacked translator was filtered at 90 Hz using an eight-pole Bessel filter(Frequency Devices) to reduce oscillations arising from rapid accelera-tion of the pipette. The open-tip solution exchange time (typically 100 –300 �s) was estimated using a solution of lower ionic strength in the drugbarrel after experiments were completed.

The recording chamber was perfused continuously with HEPES-buffered saline containing the following (in mM): 135 NaCl, 5.4 KCl, 1MgCl2, 1.8 CaCl2, and 5 HEPES, pH 7.2. Recording pipettes were filledwith the following (in mM): 129 KCl, 9 NaCl, 5 EGTA, 10 HEPES, and 4MgCl2, pH 7.2. The Cl � equilibrium potential across the patch was �0mV. GABA solutions were prepared daily from powder and diluted todesired concentrations in the same control/bath solution. Recordingswere performed at room temperature (22–25°C).

Recording electrodes were fabricated from KG-33 glass (Garner Glass)using a multistage puller (Flaming-Brown model P-97; Sutter Instru-ments) and coated with Sylgard to reduce electrode capacitance. The tipswere fire polished. Open-tip electrode resistance was typically 2– 4 M�when filling with standard recording solution. Outside-out patches wereheld at �40 mV using a low noise patch amplifier (Axopatch 200A;Molecular Devices). Data were low-pass filtered at 2–5 kHz using ampli-fier circuitry, sampled at 5–10 kHz, and stored on the computer harddrive.

Axograph, pClamp (Molecular Devices), Excel (Microsoft), and Prism(GraphPad Software) were used for data acquisition and analysis. Forcomparisons of multiple datasets, one-way ANOVA was used, with Dun-nett’s or Bonferroni’s multiple comparison post hoc tests for significanceof difference (GraphPad Software), and confirmed with a false discoveryrate test at http://www.unt.edu/benchmarks/archives/2002/april02/rss.htm(Benjamini and Hochberg, 1995).

Zn 2� blockade tests were performed as follows: patches were testedwith 200 ms pulses of 1 mM GABA, and peak current was measured forthree or more trials to assess rundown. Patches exhibiting �10% run-down were discarded. Patches were then preincubated for �30 s in con-trol solution containing 30 �M ZnCl2 and then exposed to 1 mM GABAplus 30 �M ZnCl2, and peak current was measured. Cells were thenwashed back into control solutions to recover from Zn 2� block andretested. If the post hoc test peaks were decreased by �10% from theoriginal control peaks, the test was repeated for that patch or discarded.Percentage block was calculated as [1 � (peak current in Zn 2�)/(peakcurrent in control)] � 100%.

Potentiation of IGABA by DZ was performed as follows: patches wereexposed repeatedly to a low concentration of GABA (3 �M) for 500 msuntil a stable current level was achieved. Patches were next preincubated�30 s with control solution plus 1 �M DZ and then pulsed 500 ms with 3�M GABA plus 1 �M DZ until a stable current level was achieved. Poten-tiation was calculated from peak currents before and during DZ exposureusing the equation (IGABA � DZ/IGABA) � 1.

Immunostaining. Transfected cells were replated on coverslips, washedwith PBS, fixed for 15 min in 4% paraformaldehyde in PBS, blocked with1% BSA in PBS, and incubated with M2 monoclonal 1° antibody (Sigma-Aldrich) against FLAG-tagged �2 subunits, chimeras, or truncationmutants. After multiple washes, coverslips were incubated with 2° goatanti-mouse antibodies labeled with 655 nm peak emission quantum dots(Invitrogen). Images were acquired using a Bio-Rad MRC 1024 confocallaser-scanning microscope with 488 nm laser excitation at 15–30%power and emission filters at 522 nm (bandpass, 35 nm) for EGFP and680 nm (bandpass, 32 nm) for red quantum dots.

Colocalization measurements. Transfected cells were replated on cov-erslips, washed with PBS, fixed for 15 min in 4% paraformaldehyde inPBS, blocked with 1% BSA in PBS, and incubated simultaneously withpolyclonal 1° antibody against �1 subunits and monoclonal 1° antibodyagainst FLAG-tagged �2 subunits. After multiple washes, coverslips wereincubated with 2° antibodies labeled with quantum dots at 565 nm(green, goat anti-rabbit) for �1 subunits and 655 nm (red, goat anti-mouse) for �2 subunits. Images were acquired using a Bio-Rad MRC1024 confocal laser scanning microscope with 488 nm laser excitation at15–30% power and emission filters at 522 nm (bandpass, 35 nm) and 680nm (bandpass, 32 nm), respectively. Colocalization between green andred signals was measured using ColocalizerPro software on selected re-gions of interest (whole-cell edges or nonoverlapping membrane seg-ments) after background correction. Values reported correspond to M1(see Fig. 2 B) and M2 (see Fig. 2C), where M1 is the �Rcoloc/�Rtotal andM2 is �Gcoloc/�Gtotal (R represents red pixels, and G represents greenpixels).

Truncation and point mutant construction, purification, and quantita-tion. Point mutants in �2L (S343D, S343V) were made in pCEP4 usingrecombinant PCR overlap extension with diagnostic restriction sites en-gineered into the mutating oligonucleotides. N-terminal truncated sub-unit constructs were made by PCR with an upstream oligonucleotide anda downstream oligonucleotide including coding for six lysines (or argi-nines) and a stop codon. For �2N–K6 (supplemental Fig. S3K, availableat www.jneurosci.org as supplemental material), the native sequenceends at Met 233 (in the mature rat protein sequence) with six lysinesadded before the new stop codon, whereas �2N–R6 includes Arg 231 andArg 232 plus four more arginines. �1N–K6 ends at I222 plus six lysines,and �2N–K6 ends at I218 plus six lysines using FLAG-tagged �1 (Horen-stein et al., 2001) or �2 subunit cDNA as template. A FLAG-tagged �2subunit was constructed for this study and used as template for �2N–K6

with FLAG inserted between the third and fourth amino acid residues ofthe mature rat �2 subunit. Coexpression of �2–FLAG with �1 gave cur-rents indistinguishable from nontagged �2. Constructs for �2N�M1ends at I257 with K6 or R6 tails (supplemental Fig. S3J, available at www.jneurosci.org as supplemental material). All constructs were subclonedinto pCEP4 and were confirmed with double-stranded DNA sequencing.

For protein production, HEK293 cells were transfected withN-terminal truncated subunit plasmid constructs at 500 –1000 ngcDNA/35 mm plate. Forty-eight to 72 h after transfection, intact cellswere washed with ice-cold PBS (in mM: 2.7 KCl, 1.5 KH2PO4, 0.5 MgCl2,137 NaCl, and 14 Na2HPO4, pH 7.1). Sulfhydryl groups were blocked byincubating cells with 10 mM N-ethylmaleimide (NEM) in PBS (15 min,room temperature). Cells were solubilized (overnight, 4°C) in lysis buffer(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA,pH7.5) supplemented with protease inhibitors (Complete, mini; RocheDiagnostics) and 10 mM NEM. Lysates were cleared by centrifugation(16,000 � g, 10 min, 4°C). The FLAG-tagged N-terminal truncationswere immunopurified from the cell lysates by incubating (4 – 6 h, 4°C,rotating) with 40 �l of EZview Red ANTI-FLAG M2 affinity gel (Sigma-Aldrich), washed six times with 1 ml of wash buffer (150 mM NaCl and 50mM Tris-Cl, pH 7.5). The FLAG-tagged subunits were eluted from thebeads with 100 �l of 400 �g/ml FLAG peptide in recording buffer (1 h,4°C rotating). After the incubation, the samples were centrifuged(8200 � g, 10 min, 4°C), and the eluate was collected and stored at�20°C.

For protein concentration determinations, samples were denaturedwith 2� Laemli’s sample buffer (3% SDS, 0.6 M sucrose, 0.325 M Tris-HCl, pH 6.8, and 10 mM DTT) and then separated by SDS-PAGE on

4896 • J. Neurosci., April 7, 2010 • 30(14):4895– 4903 Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR

Page 3: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

4 –15% gradient Tris-HCl Ready Gels (Bio-Rad). Samples were runalongside a twofold dilution series of Met-FLAG-BAP (Sigma) from 400to 6.25 ng. After transfer to nitrocellulose, blots were processed using theOne-Step Western Blot kit for mouse (Genscript), according to the in-structions of the manufacturer and using M2 anti-FLAG antibody

(Sigma) at 1:400 dilution. Blots were thentreated with ECL Western Blotting DetectionReagents (RPN2106V1; GE Healthcare) andseveral exposures on Kodak X-Omat AR,XAR-5 film (Eastman Kodak) were taken forcomparison of signal linearity. It was notedthat Pierce SuperSignal West Pico Chemilumi-nescent Substrate was distinctly nonlinear andwas therefore not used for these determina-tions. Exposed films were scanned at 1200 dpiand analyzed using LabWorks software version4.6 (UVP Inc.) and confirmed using NIH Im-ageJ software (http://rsb.info.nih. gov/ij/).

ResultsPotentiation of IGABA by the classical BDZDZ was measured (Fig. 1, green traces) in�1�2 receptors (Fig. 1A, without �2 sub-units), �1�2�2 receptors with stoichiom-etry fixed by using a 1:1:10 ratio, and intransfections with lower �2 ratios (Fig.1B). Potentiation values for receptorswith fixed stoichiometry by using high �2ratios or by using subunit concatamersranged from 2.0 to 2.5 (IGABA � DZ/IGABA � 1), whereas �1�2 receptorsshowed no potentiation, as expected (Fig.1A). Receptors formed from transfectionsof �1�2�2 in a 1:1:1 ratio showed less po-tentiation with greater variability (1.26 �0.55 for �2S and 1.20 � 0.23 for �2L,mean � SD), as predicted for a mixture of�1�2 and �1�2�2 receptors. Moreover,as the �2S or �2L ratio was decreased, themean potentiation dropped further, asmore �1�2 receptors were formed. Thevariability of BDZ potentiation at low ra-tios is illustrated using �1�2�2L transfec-tions at 1:1:0.1 (Fig. 1, right).

The same patches were also tested forZn 2� block (Fig. 1, red traces). For trans-fections with the �2S splice variant (Fig.1B, left), the vast majority of patches fromdifferent transfection ratios, ranging from�1�2�2 1:1:10 down to 1:1:0.1, demon-strated minimal Zn 2� block. At lower �2Sratios such as �1�2�2 1:1:0.03 and 1:1:0.01, the majority of patches showed nopotentiation by DZ and maximal Zn 2�

block, as if they contained mainly �� re-ceptors. What is striking is that there werealmost no intermediate values for Zn 2�

block with �2S. When the paired DZ andZn 2� data were plotted together (Fig. 1C),it became clear that, in the majority ofpatches, even when small DZ potentiationwas detected (indicative of a preponder-ance of �1�2 receptors), the same patchwas relatively insensitive to Zn 2� block.Most of the Zn 2�-insensitive patches

from �1�2�2 1:1:�1 transfections fell outside the 95% confi-dence intervals of a model prediction for a mixture of �1�2and �1�2�2 receptors, derived from measurements for EC50,open probability, and single-channel conductances (Boileau

Figure 1. Diazepam potentiation and Zn 2� block for varying �2S or �2L transfection ratios. A, HEK293 cells expressing �1 plus�2 subunits. Left, Current traces from 500 ms, 3 �M GABA pulses (black) are superimposed with currents from the same outside-out patch exposed to a coapplication of 3 �M GABA plus 1 �M DZ (green) after equilibration in control solution plus 1 �M DZ.Potentiation (Pot) is reported as (IGABA � DZ/IGABA) � 1. Right, In the same patch, near-maximal current is elicited by 200 ms, 1 mM

GABA pulses (black) and blocked by coapplication of 30 �M ZnCl2 (red). Note that �1�2 receptors display significant Zn 2� blockbut no appreciable DZ potentiation. B, Traces for cells expressing �1 � �2 � �2S (left) or �2L (right) at indicated ratios. Notethat three �2L patches are shown from 1:1:0.1 ratio transfections to exhibit variability. Also included are traces for the �1�2�2Lreceptors constrained by use of �1–�2 tandem receptors cotransfected with �2L at a 2:1 ratio. Note that these receptors showsimilar high potentiation by DZ, minimal blockade by Zn 2�, and minimal desensitization in 200 ms pulses (right) at 1 mM GABA,similar to �1�2�2S receptors constrained by transfection at a 1:1:10 ratio. C, Paired Zn 2� block plotted against DZ potentiationfor transfection ratios ranging from 1:1:0.01 to 1:1:10 for �1�2�2, as well as tandem-constrained ��tan�� (Boileau et al.,2005) and �1�2. Note that �� receptors (open squares) show high zinc sensitivity and no potentiation, as do some of the patchesfrom transfections with very low �2 ratios, indicative of no �2 subunits incorporated in that patch. The solid curve represents aninterpolation between �� and ��� DZ potentiation and Zn 2� block values, modeled using EC50, single-channel conductance,and open probability differences between the two receptor types (Boileau et al., 2005). The open circle lies on the model curve andhas error bars corresponding to the 95% confidence intervals (CIs) for DZ potentiation and Zn 2� block (dashed lines). Note thatmany of the Zn 2� block values for transfections with �2S ratios below 1:1:10 fall beneath the 95% confidence interval (C, left),whereas transfections varying �2L ratios do not (C, right).

Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR J. Neurosci., April 7, 2010 • 30(14):4895– 4903 • 4897

Page 4: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

et al., 2005). However, DZ potentiationcompared with deactivation and concen-tration–response ratios fit within mod-eled confidence estimates (supplementalFig. S1, available at www.jneurosci.org assupplemental material).

In stark contrast, the �2L subunitshowed DZ potentiation and Zn 2� blockthat fit well with the same model (Fig. 1C,right). When DZ potentiation was high,Zn 2� block was low and vice versa, andintermediate values mimicked those ex-pected for a mixture of �1�2 and �1�2�2receptors in a continuum of ratios. Theseresults suggest that �2S, but not �2L, sub-units somehow confer anomalous Zn 2�

pharmacology to the �1�2 receptors inthe mixture that should be blocked byZn 2� but are not. There was no correla-tion between current size (i.e., expressionlevels) and zinc blockade. For example,current amplitudes ranged from 23 to 503pA for �1�2�2L 1:1:0.1 transfections (co-efficient of variation of 0.93) (Fig. 1B),but correlation with zinc block was notsignificant (R 2 0.126; data not shown).Similar data were obtained in Xenopus oo-cyte voltage-clamp experiments (data notshown), lending confidence to the gener-alizable nature of the phenomenon. Theresults also demonstrate that reliance onZn 2� block for estimations of �2 incorpo-ration into receptors is risky at best for either splice variant (Fig.1C). Mixtures of �1�2 and �1�2�2L receptors may be difficult todistinguish from pure �1�2�2 receptors using zinc alone (e.g.,70% �1�2�2L receptors would be indistinguishable from 100%).Evidently, currents with even miniscule �2S can appear to beentirely Zn 2� insensitive, giving a false impression of full �incorporation.

One documented difference between the two �2 splice vari-ants is the ability of �2S to express on the surface of the cell in theabsence of � and � subunits, whereas �2L is retained in intracel-lular compartments unless coexpressed with � and � subunits(supplemental Fig. S2, available at www.jneurosci.org as supple-mental material), presumably in the endoplasmic reticulum(Connolly et al., 1999). Could �2S subunits express on the surfacein the presence of �1 and �2 subunits but not incorporate into�1�2�2 receptors? To test this, colocalization of �1 subunits toeither �2 splice variant was measured, in �1�2�2 transfectionratios 1:1:0.1, 1:1:1, and 1:1:10 (Fig. 2). Even in the lowest trans-fection ratio, �2S subunits showed significantly less colocaliza-tion to �1 subunits than did �2L subunits (Fig. 2B). In the highestratio, the difference was overwhelming and shows that, contraryto a previous suggestion (Connolly et al., 1999), �2S subunits canexpress on the surface as “rogues” even in the presence of coex-pressed �1 and �2 subunits. The converse control, colocalizationof �1 to �2 subunits, showed no differences between splice vari-ants (Fig. 2C). Whether the rogue �2S subunits are present on thesurface as monomers or multimers is presently unknown.

To test further the apparent correlation between surface ex-pression and anomalous Zn 2� block, mutations were introducedinto the eight amino acid region that differentiates the two splicevariants. Serine at position 343 in the �2L subunit has been

shown to be a target for kinases (Whiting et al., 1990; Moss et al.,1992), and �2S does not contain this residue. S343 was mutatedto nonphosphorylatable valine or to an aspartate as an attempt toadd a phosphomimetic side chain (Lin et al., 1998). Both muta-tions resulted in surface expression of the �2L subunit when ex-pressed alone, like �2S (Fig. 3A). Moreover, in �1�2�2L(mut)transfections at 1:1:0.1 ratios, patches excised from cells express-ing either mutant showed low Zn 2� blockade, inconsistent withthe paired DZ potentiation measurements from the same patches(Fig. 3B). In other words, the surface expression of �2L(mut)correlated with the anomalous Zn 2� block and now mimicked�2S. Because the two types of mutations showed no apparentdifferences, the aspartate substitution either conferred no phos-phomimetic effect and/or the ER retention regulome of theHEK293 cell recognizes a serine or phosphoserine in particular aspart of the retention signal.

Several unsuccessful chimeric and truncated subunits wereconstructed to narrow the regions of the �2S subunit responsiblefor conferring protection from Zn 2� block to �1�2 receptors(supplemental Fig. S3, available at www.jneurosci.org as supple-mental material). For example, coexpression of the �2S M3–M4cytoplasmic loop with �1 and �2 subunits was not sufficient toconfer protection (data not shown), nor did a chimeric �1/�2/�1chimera with the �2S M3–M4 loop (supplemental Fig. S3E, avail-able at www.jneurosci.org as supplemental material) show Zn 2�

insensitivity when expressed with �1 and �2 subunits or with �2subunits alone. Receptors formed from coexpression of �1, �2,and a previously described chimeric �2/�1 subunit with �2 se-quence through to the M3 transmembrane region (supplementalFig. S3D, available at www.jneurosci.org as supplemental mate-rial) (Boileau and Czajkowski, 1999) at 1:1:0.5 ratio were variable

Figure 2. �2S subunits can also express independently, even in the presence of �1 and �2 subunits. A, Selected images fromcolocalization experiments performed on HEK293 cells transfected with �1�2�2 cDNAs at 1:1:0.1, 1:1:1, and 1:1:10 ratios. Cellswere labeled with 565 nm quantum dot-labeled antibodies (green) for �1 subunits and 655 nm (far red, pseudocolored red) for �2subunits (see Materials and Methods). Colocalized signal ranges from yellow– green to orange–red. Scale bar, 20 �m. B, Mea-surements of colocalization of �2L or �2S with �1 signal. At each transfection ratio, �2S subunits were significantly different from�2L subunits in the amount of red signal that was not colocalized with green (�1) signal. Data are mean � SEM for n � 50 cellsper condition. *p 0.01, **p 0.001. C, Control measurements of �1 colocalization with �2L or �2S. At no transfection ratiodoes �2S differ significantly from �2L for this measurement.

4898 • J. Neurosci., April 7, 2010 • 30(14):4895– 4903 Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR

Page 5: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

in Zn 2� block (i.e., not protected), possibly because this chimeradid not express as a rogue subunit (data not shown). No trunca-tion mutants (supplemental Fig. S3F–K, available at www.jneurosci.org as supplemental material) expressed on the cellsurface (supplemental Fig. S2 shows two examples, available atwww.jneurosci.org as supplemental material).

One truncation mutant, consisting of the extracellularN-terminal domain of the �2 subunit (�2N), was outfitted withsix additional positively charged lysines (supplemental Fig. S3K,available at www.jneurosci.org as supplemental material) tomimic a cell-penetrating peptide (Han et al., 2001). Studies usingpeptide carriers with fluorescent tags have shown that a substan-tial portion of the signal remains affixed to the cell surface unlessit is cleaved away with proteases (Richard et al., 2003). For thepurpose of probing for a direct interaction between the Nterminus of the �2S subunit with �1�2 receptors, sticking tothe surface was an exploitable artifact. Confocal images of immu-nopurified �2N–K6 applied externally to �1�2-transfected cellsfor 1 h show that indeed the peptide adheres to the cell surface(Fig. 4A).

When �1�2 receptors treated with modified �2N were testedfor Zn 2� blockade, protection was prominent (Fig. 4B). Controlsticky proteins constructed from the N termini of the �1 subunit(�1N–K6), �2 subunit (�2N–K6), and quantum dot-linkedwheat germ agglutinin (WGA) were also able to adhere to the cellsurfaces (Fig. 4A) but did not protect �1�2 receptors from Zn 2�

block (Fig. 4B). Summary quantitation of these effects are seen inFigure 4C, where Zn 2� block for �1�2 and �1�2�2 receptors iscompared with �1�2 receptors treated with �2N–K6, �1N–K6,�2N–K6, or WGA. Only �2N–K6 application protected �1�2receptors significantly and yielded Zn 2� blockade not signifi-cantly different from that for �1�2�2 receptors. A hexaarginineversion of the �2N was also constructed and tested but did notyield protection as effectively as the hexalysine �2N, nor didhexaarginine and hexalysine versions of �2N plus the M1 trans-membrane region (supplemental Fig. S3J, available at www.jneurosci.org as supplemental material), for reasons that are notunderstood (data not shown). Because the �2N–K6 proteins lackany transmembrane regions to anchor in the membrane or formpart of the channel pore (formed by transmembrane region M2),these results strongly suggest that the N terminus of rogue �2Srenders �1�2 receptors insensitive to Zn 2� block, without beingincorporated into receptor pentamers themselves. This may oc-cur by either occluding external Zn 2� binding site(s) (Hosie et

al., 2003) or causing conformationalchanges to render Zn 2� ineffective (Fig.4Diii) (supplemental Fig. S4, available atwww.jneurosci.org as supplemental ma-terial). The rogue �2S subunit may act as alone subunit or multimer (Fig. 4Diii) ormay induce clustering of �1�2 recep-tors, leading to alteration or occlusionof the extracellular Zn 2� binding sites(Fig. 4Div). Regardless of the exact mech-anism, it is clear that the �2S subunit canhave an extra-receptor, modulatory role.

As is often observed in the literature atlow to intermediate �2S ratios (e.g., 1:1:1and 1:1:0.5), many patches showed a veryfast desensitization component (Fig. 5A–C), concomitant with a trend towardfaster rise times (Fig. 5D,E). The fastestcomponent of desensitization in �1�2 re-

ceptors is 21 � 4 ms (mean � SD), whereas in pure populationsof �1�2�2 receptors (�1�2�2 at 1:1:10, ��tan��2, �-�-���-�concatamers), the fastest time constant is on the order of 1 s(Boileau et al., 2005; Ericksen and Boileau, 2007). However, inmixed �1�2/�1�2�2 populations, the fastest time constant com-ponent is �7 ms (Fig. 5C), representing a new “ultrafast” com-ponent more rapid than either �1�2 or �1�2�2 receptors.Comparable fast time constants could be seen with �1�2�2Lwhen transfected at low ratios that yield a mixed �1�2/�1�2�2Lpopulation, suggesting that this effect is not attributable to rogue�2S subunits. These data suggest rather that mixtures or clustersof �1�2�2 with �1�2 receptors change the characteristics of bothreceptor subtypes compared with a pure population of either type(Fig. 5F). A fast time constant component can also be observed inacutely dissociated neurons (Kapur et al., 1999), suggesting thepossibility of mixed receptor types in vivo. The 10 –90% rise timesfor �1�2�2L transfections are fairly uniform in mixed �1�2/�1�2�2L populations (�1 ms) (Fig. 5D), suggesting that cluster-ing of �1�2 with �1�2�2 receptors affects receptor kinetics,coincident with the appearance of the ultrafast desensitizationcomponent. An example of two �� receptors clustered with two��� receptors is depicted in Figure 5Fiii.

A second kinetic phenomenon that does not appear to dependon clustering, but rather on �2S expression, was also observed. Inthe case of increased transfection ratio of �2S in particular (1:1:1,1:1:10), the rise time becomes slower (Fig. 5E). Parsimony sug-gests that rogue �2S binding to �1�2�2S receptors mightlengthen their rise times (compare 1:1:1 and 1:1:10 �1�2�2L risetimes in Fig. 5D with �1�2�2S in Fig. 5E), such as by sterichindrance of the GABA binding site(s) or allosteric changes, sim-ilar to the possible mechanism of protection from Zn 2� blockade(Fig. 5Fi). Alternative explanations for this difference in risetimes include the following possibilities: (1) �2S-containing re-ceptors normally have slower rise times than �2L-containing re-ceptors without the need to invoke rogue subunits, and the fasterrise times at lower transfection ratios are a common function of�1�2/�1�2�2 clustering independent of splice variant (Fig.5Fiii); (2) the additional �2S subunits that are expressed but notincorporated may bind to intracellular proteins that affect�1�2�2 binding, gating, or desensitization mechanisms, or (3) acombination thereof. However, rise times for �1�2 receptorswere unaffected by �2 N-terminal peptides (data not shown),suggesting that more of the �2 subunit than the extracellularportion would be needed to slow rise times for �� receptors, if

Figure 3. Mutations of serine 343 in �2L subunits allow for rogue surface expression and anomalous Zn 2� blockade. A,Confocal images of cells cotransfected with EGFP and �2L–S343D (serine to aspartate) or �2L–S343V (serine to valine) are shownon the left. Scale bar, 20 �m. B, In separate experiments, excised patches from cells transfected with �1�2�2L–S343D or�1�2�2L–S343V (both at the transfection ratio 1:1:0.1) were tested for DZ potentiation and Zn 2� blockade, as in Figure 1. Notethat both mutations cause �2L to resemble �2S.

Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR J. Neurosci., April 7, 2010 • 30(14):4895– 4903 • 4899

Page 6: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

indeed it can. Possible effects of rogue �on �� receptors in native tissue awaits ad-ditional study, because the existence ofthis receptor combination in vivo is still afairly recent discovery (Mortensen andSmart, 2006). Effects of �2S rogue sub-units on mixed clusters (as in Fig. 5Fiii)that display the fast desensitization com-ponent and fast rise times (perhaps repre-senting the synaptic configuration)(Boileau et al., 2005) could be studied us-ing concatamer pentamers coexpressedwith rogue �2S.

DiscussionIn the present study, it has been demon-strated that the ability of the short �2 vari-ant to express independently on the cellsurface correlates with changes in coex-pressed GABAARs in both pharmacology(zinc blockade) and fast receptor kinetics.However, what might be the physiologicalrole of the modulatory effect of the rogue�2S subunit? At this juncture, there aresome speculations that can be broached.In early development, brain slice record-ings show GABAARs to be less BDZ sensi-tive, coincident with lower expression of�2 subunits (Brooks-Kayal et al., 1998). In�2S-enriched dentate gyrus, Kapur andMacdonald (1999) observed that, at ratpostnatal day 45 (P45) to P52, dentategranule cells had normal (high) diazepampotentiation and normal (low) zinc inhi-bition. However, at P7–P14, althoughzinc inhibition was still low, BDZ poten-tiation was vastly reduced. This could re-sult from fewer fully incorporated ���receptors, with a few �2S rogues confer-ring protection from zinc to �� receptors.

Developmental differences in zincinhibition could be related to receptorclustering. Clustering of �2-containingsynaptic receptors become evident by P20in rats (Scotti and Reuter, 2001). How-ever, in hippocampal neurons, Zn 2� sen-sitivity is relatively low at P7–P14. Wecan speculate that, because the �2S variantdominates in that region, it might protect nascent neurons fromdiffuse, nonvesicularized Zn 2� ions (Valente et al., 2002). It isinteresting that very young rats (P10) cannot be made epilepticwith pilocarpine (Zhang et al., 2004), perhaps because the higherexpression level of the �2S subunit protects young neurons fromdeleterious overstimulation. Pathogenic conditions such as neo-natal stress keep GABAARs in an immature declustered state withfewer �2 subunits in the mixture (Hsu et al., 2003), and micro-tubule depolymerizing drug treatment in hippocampal neuronsshowed declustering of GABAARs concomitant with a loss of �2subunit-containing receptors as assessed by the BDZ sensitivity(Petrini et al., 2004). This declustering also led to faster rise timesand faster desensitization of miniature IPSCs, reminiscent of thechanges in �1�2�2S kinetics with decreased �2S ratio (Fig.5A,D).

Several papers have suggested the existence of �� receptors invivo under normal (Brickley et al., 1999; Yeung et al., 2003) or �2knock-out (Gunther et al., 1995) conditions, and a recent studyhas described �� receptors extrasynaptically (Mortensen andSmart, 2006). In the latter study, Zn 2� block of �� receptors wasdetectable, but, in a brain region in which �2S subunits predom-inate or in the synapses in which �2 subunits tend to cluster(Alldred et al., 2005; Christie et al., 2006), the present studywould predict that �� receptors would be difficult if not impos-sible to detect using Zn 2� pharmacology alone (Fig. 1). Even inon-cell patch recordings, an �1�2/�1�2�2 mixture might be easyto miss because �1�2 receptors desensitize to a much greaterextent, tend to stand silent (desensitized) under constant agonistexposure, and have a main conductance (�15 pS) close to the mainsubconductance state of �1�2�2 receptors (�21 pS) (Boileau et al.,

Figure 4. External application of truncated extracellular �2 subunits reduces Zn 2� block in �� receptors. A, Confocal imagesof cells cotransfected with �1 and �2 subunits and treated with immunopurified sticky proteins �2N–K6, �1N–K6, �2N–K6, orWGA linked to quantum dots. Cells were incubated in PBS for 60 min with immunopurified protein, washed several times, fixed,and treated with anti-FLAG antibody with 655 nm quantum dot-linked 2° antibody for the GABAAR extracellular polypeptides. Onthe left are transmitted light images, and on the right are the pseudocolored far red images. Scale bar, 20 �m. B, Top, Traces fromexcised patches showing reduced Zn 2� blockade of �1�2 receptors incubated with the �2 N-terminal protein �2N–K6. Traceswith or without coapplied Zn 2� are the average of three traces from the same patch. Below are shown traces from �1�2 receptorsincubated with control �1N–K6, �2N–K6, or quantum dot-linked WGA, none of which protected the receptors from Zn 2� block.C, Summary of Zn 2� block for �1�2 receptors (white bar), �1�2�2 receptors (black; pooled �1�2�2S 1:1:10, �1�2�2L1:1:10, ��tan��2S and ��tan��2L), and �1�2 receptors treated with �2N–K6 (red bar, average of 200 ng of protein per 12mm coverslip in a 1 ml well) or controls �1N–K6, �2N–K6, or WGA (gray bars, average of 500 ng of protein per coverslip). *p 0.001 significance of difference from �1�2 receptors; n.s., �1�2�2 and �1�2 � �2N-K6 peptide not significantly differentfrom one another. Inset, Depiction of the sticky �2N–K6 protein. D, Schematic of possible mechanisms for Zn 2� protective effectof �2S on �1�2 receptors. i, The �1�2�2 receptor has one putative external Zn 2� binding site (black hexagon) and one “weak”luminal binding site (gray hexagon). ii, The �1�2 receptor has two identical external Zn 2� binding sites and one internal site thatmay have higher affinity than in the case of �1�2�2 receptors. i and ii patterned after Hosie et al. (2003). iii, Rogue �2S subunits(as lone subunits or multimers of unknown number) may bind to �1�2 receptors and hinder binding of Zn 2� ions. iv, �2Ssubunits may cause �1�2 receptors to cluster, thus reducing Zn 2� block of several receptors.

4900 • J. Neurosci., April 7, 2010 • 30(14):4895– 4903 Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR

Page 7: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

2005). Therefore, BDZ potentiation, which is thoroughly � sub-unit dependent, appears to be a more reliable test for receptormixtures. For example, in �2S-rich posterior pituitary nerve ter-minals, Zhang and Jackson (1993) observed only small potentia-tion by the full agonist BDZ chlordiazepoxide on subsaturatingGABA currents in membrane patches, indicating that not everyreceptor incorporates �2 in this region. Meanwhile, the minimalblockade by 100 �M Zn 2� (24 � 6%) in the same cells (Zhangand Jackson, 1995) would suggest that most or all of the receptorshave �2, if that were the only criterion used. The lower BDZpotentiation is likely not attributable to the presence of native �4or �6 subunits, because these have been shown to exhibit highersensitivity to Zn 2� block (Knoflach et al., 1996).

One may also speculate about the involvement of �2 slicevariants in the formation or prevention of epileptiform activity.

Certain epileptiform mutations in the�2 gene (Macdonald et al., 2004; Hirose,2006) can cause a reduction in �2 expres-sion, sometimes from assembly impair-ments (Sancar and Czajkowski, 2004;Hales et al., 2005; Kang et al., 2006;Eugene et al., 2007). Animal models ofepileptogenesis and adult human epilep-tic brain have hinted at reduced �2 ex-pression along with increased expressionof BDZ-insensitive �4 and � subunits(Brooks-Kayal et al., 1998; Coulter, 2001),both of which form receptors more zincsensitive than �1�2�2 (Saxena andMacdonald, 1994; Knoflach et al., 1996).The effects on BDZ and zinc modulationof GABAARs during induction of statusepilepticus were tested previously in rat(Kapur and Macdonald, 1997). The au-thors observed a rapid reduction in BZDsensitivity (on the order of minutes), and,although there was a shift in IC50 for Zn 2�

blockade, the relative sensitivity in the 30�M range remained rather low. Again, apossible involvement of �2S would beworth exploring with what we know now.Whether rogue �2S subunits can also pro-tect these receptor subtypes is an intrigu-ing possibility. It will also be interesting totest for differences in Zn 2� protection us-ing disease state assays (e.g., kindling orother epileptogenic treatment) in trans-genic mice with �2S or �2L knocked intothe �2 knock-out background (Wick etal., 2000), to determine whether inhibi-tion or behavior differ for the splice vari-ants under stressed conditions.

Another possible protecting proteinis the recently described GABAAR � sub-unit. Data from one study with limiting �coexpressed with �1 and �1 might be re-interpreted as the � subunit conferringZn 2� protection in a manner similar to�2S (Thompson et al., 2002). The obviousfollow-up question— do � subunits ex-press on the surface by themselves?— hasvery recently been answered in the affir-mative (Jones and Henderson, 2007) in a

study that also sheds some light on the functional variability seenin heterologously expressed � subunits.

Trafficking and assembly of GABAARs regulates the subtypesof inhibitory GABAergic signaling in the brain. An intriguing lineof inquiry arising from this study would be to determine thedifferences in the trafficking of rogue �2S subunits versus those�2S subunits that incorporate into normal synaptic and extrasyn-aptic GABAARs. Recent studies of GABAAR trafficking proteinsshow that, in mice knock-outs of PRIP (phospholipase C-relatedcatalytically inactivated proteins that bind to GABARAP), diaze-pam function was reduced, possibly as a result of disassembly of��� receptors (Kanematsu et al., 2007). PRIP associates not onlywith the � subunit but also with GABARAP (GABAA receptor-associated protein), an accessory protein that binds to and en-hances �2 subunit incorporation and/or stability (Boileau et al.,

Figure 5. Mixtures of �1�2 and �1�2�2 receptors exhibit faster kinetics than either pure population but are altered by rogue�2S subunits. A, Overlaid current traces from four patches each, from �1�2�2S 1:1:10, 1:1:1, 1:1:0.5, 1:1:0.1, 1:1:0.03, and 1:1:0(�1�2) transfections, in response to 200 ms pulses of 1 mM GABA. B, Expanded trace comparing fast desensitization in �1�2receptors with �1�2�2S 1:1:0.5. Note that the �1�2�2 1:1:0.5 current is not only faster to desensitize but also faster to rise topeak. C, Plot of fast desensitization time constants (fast) transfected with �1 and �2 subunits, with varying ratios of �2S or �2Lsubunits, wherein a fast component could be detected. Data are mean � SEM for n 17, 6, 13, 8, 3, 6, 7, and 10 patches (left toright). *p 0.01 compared with �1�2 patches. D, Scatter plot of 10 –90% rise times for the cells transfected with �1 and �2subunits, with varying ratios of �2L subunits. Note that ratios over a 100-fold range (1:1:0.1 to 1:1:10) give similar rise times for�2L. E, Scatter plot of 10 –90% rise times for ���2S transfections in varying ratios. Note that, as �2S ratio is increased above 0.5,rise times are slowed. F, Models depicting fast desensitization differences between receptors and mixtures. i, �1�2�2 receptors,possibly clustered, with trace showing no fast desensitization. In the case of �2S only, rogue �2S subunits may bind at the �/�interface to cause a slowing of the rise time (compare D with E at 1:1:1 and 1:1:10). ii, �1�2 receptor with fast desensitization. iii,Mixtures of �� and ��� receptors may cluster and change kinetic profiles to allow for the ultrafast (7 ms) desensitization timeconstant. Traces were culled from Figure 1.

Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR J. Neurosci., April 7, 2010 • 30(14):4895– 4903 • 4901

Page 8: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

2005). One current model of interaction is that �2 subunit bind-ing and incorporation is facilitated by the quaternary interactionsof � subunit to PRIP, PRIP to GABARAP, and in turn to the �2subunit (Kanematsu et al., 2007). Does this scheme differ for thetwo splice variants? Do GABARAP or PRIP-bound GABARAPbind to rogue �2S subunits, and, if so, are they then held in closeproximity to fully formed receptors by indirect association with �subunits? Or do rogues disregard all affiliations on their route tothe surface and bind to fully formed receptors via translocation,followed by extracellular protein–protein interactions? A recentpaper combining molecular modeling with experimental data onthe high-affinity Zn 2� binding site in the pore of �1�2 receptors(partially reconstituted in �1�2�2S receptors by mutation of the�2S subunit) suggests a possible role for membrane-spanningresidues in rogue protection as well (Trudell et al., 2008). Traf-ficking and binding mechanisms notwithstanding, whether otherCys-loop receptor subunits can go rogue and alter their familialreceptors in vivo remains to be determined. Rogue subunits addanother level of intricacy to the exquisite control of neuronalsignaling by nature.

ReferencesAkabas MH (2004) GABAA receptor structure-function studies: a reexam-

ination in light of new acetylcholine receptor structures. Int Rev Neuro-biol 62:1– 43.

Alldred MJ, Mulder-Rosi J, Lingenfelter SE, Chen G, Luscher B (2005) Dis-tinct �2 subunit domains mediate clustering and synaptic function ofpostsynaptic GABAA receptors and gephyrin. J Neurosci 25:594 – 603.

Baumann SW, Baur R, Sigel E (2002) Forced subunit assembly in �1�2�2GABAA receptors. Insight into the absolute arrangement. J Biol Chem277:46020 – 46025.

Baur R, Minier F, Sigel E (2006) A GABA(A) receptor of defined subunitcomposition and positioning: concatenation of five subunits. FEBS Lett580:1616 –1620.

Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: apractical and powerful approach to multiple testing. J R Stat Soc Series BStat Methodol 57:289 –300.

Boileau AJ, Czajkowski C (1999) Identification of transduction elements forbenzodiazepine modulation of the GABAA receptor: three residues arerequired for allosteric coupling. J Neurosci 19:10213–10220.

Boileau AJ, Baur R, Sharkey LM, Sigel E, Czajkowski C (2002) The relativeamount of cRNA coding for gamma2 subunits affects stimulation bybenzodiazepines in GABAA receptors expressed in Xenopus oocytes. Neu-ropharmacology 43:695–700.

Boileau AJ, Pearce RA, Czajkowski C (2005) Tandem subunits effectivelyconstrain GABAA receptor stoichiometry and recapitulate receptor kinet-ics but are insensitive to GABAA receptor-associated protein. J Neurosci25:11219 –11230.

Brickley SG, Cull-Candy SG, Farrant M (1999) Single-channel properties ofsynaptic and extrasynaptic GABAA receptors suggest differential targetingof receptor subtypes. J Neurosci 19:2960 –2973.

Brooks-Kayal AR, Jin H, Price M, Dichter MA (1998) Developmental ex-pression of GABAA receptor subunit mRNAs in individual hippocampalneurons in vitro and in vivo. J Neurochem 70:1017–1028.

Christie SB, Li RW, Miralles CP, Yang BY, De Blas AL (2006) Clustered andnon-clustered GABAA receptors in cultured hippocampal neurons. MolCell Neurosci 31:1–14.

Connolly CN, Uren JM, Thomas P, Gorrie GH, Gibson A, Smart TG, Moss SJ(1999) Subcellular localization and endocytosis of homomeric gamma2subunit splice variants of gamma-aminobutyric acid type A receptors.Mol Cell Neurosci 13:259 –271.

Coulter DA (2001) Epilepsy-associated plasticity in gamma-aminobutyricacid receptor expression, function, and inhibitory synaptic properties. IntRev Neurobiol 45:237–252.

Ericksen SS, Boileau AJ (2007) Tandem couture: cys-loop receptor con-catamer insights and caveats. Mol Neurobiol 35:113–128.

Eugene E, Depienne C, Baulac S, Baulac M, Fritschy JM, Le Guern E, Miles R,Poncer JC (2007) GABAA receptor �2 subunit mutations linked to hu-man epileptic syndromes differentially affect phasic and tonic inhibition.J Neurosci 27:14108 –14116.

Gunther U, Benson J, Benke D, Fritschy JM, Reyes G, Knoflach F, Crestani F,Aguzzi A, Arigoni M, Lang Y (1995) Benzodiazepine-insensitivemice generated by targeted disruption of the gamma 2 subunit gene ofgamma-aminobutyric acid type A receptors. Proc Natl Acad Sci U S A92:7749 –7753.

Gutierrez A, Khan ZU, De Blas AL (1994) Immunocytochemical localiza-tion of �2 short and �2 long subunits of the GABAA receptor in the ratbrain. J Neurosci 14:7168 –7179.

Gutierrez A, Khan ZU, Miralles CP, De Blas AL (1996) Altered expression ofgamma 2L and gamma 2S GABAA receptor subunits in the aging ratbrain. Brain Res Mol Brain Res 35:91–102.

Hales TG, Tang H, Bollan KA, Johnson SJ, King DP, McDonald NA, Cheng A,Connolly CN (2005) The epilepsy mutation, gamma2(R43Q) disrupts ahighly conserved inter-subunit contact site, perturbing the biogenesis ofGABAA receptors. Mol Cell Neurosci 29:120 –127.

Han K, Jeon MJ, Kim SH, Ki D, Bahn JH, Lee KS, Park J, Choi SY (2001)Efficient intracellular delivery of an exogenous protein GFP with geneti-cally fused basic oligopeptides. Mol Cells 12:267–271.

Hevers W, Luddens H (1998) The diversity of GABAA receptors. Pharma-cological and electrophysiological properties of GABAA channel sub-types. Mol Neurobiol 18:35– 86.

Hirose S (2006) A new paradigm of channelopathy in epilepsy syndromes:intracellular trafficking abnormality of channel molecules. Epilepsy Res70 [Suppl 1]:S206 –S217.

Horenstein J, Wagner DA, Czajkowski C, Akabas MH (2001) Protein mo-bility and GABA-induced conformational changes in GABAA receptorpore-lining M2 segment. Nat Neurosci 4:477– 485.

Hosie AM, Dunne EL, Harvey RJ, Smart TG (2003) Zinc-mediated inhibi-tion of GABAA receptors: discrete binding sites underlie subtype specific-ity. Nat Neurosci 6:362–369.

Hsu FC, Zhang GJ, Raol YS, Valentino RJ, Coulter DA, Brooks-Kayal AR(2003) Repeated neonatal handling with maternal separation perma-nently alters hippocampal GABAA receptors and behavioral stress re-sponses. Proc Natl Acad Sci U S A 100:12213–12218.

Huntsman MM, Tran BV, Potkin SG, Bunney WE Jr, Jones EG (1998) Al-tered ratios of alternatively spliced long and short gamma2 subunit mR-NAs of the gamma-amino butyrate type A receptor in prefrontal cortex ofschizophrenics. Proc Natl Acad Sci U S A 95:15066 –15071.

Jones BL, Henderson LP (2007) Trafficking and potential assembly patternsof epsilon-containing GABAA receptors. J Neurochem 103:1258 –1271.

Kanematsu T, Mizokami A, Watanabe K, Hirata M (2007) Regulation ofGABAA-receptor surface expression with special reference to the involve-ment of GABARAP (GABAA receptor-associated protein) and PRIP(phospholipase C-related, but catalytically inactive protein). J PharmacolSci 104:285–292.

Kang JQ, Shen W, Macdonald RL (2006) Why does fever trigger febrileseizures? GABAA receptor �2 subunit mutations associated with idio-pathic generalized epilepsies have temperature-dependent trafficking de-ficiencies. J Neurosci 26:2590 –2597.

Kapur J, Macdonald RL (1997) Rapid seizure-induced reduction of benzo-diazepine and Zn 2� sensitivity of hippocampal dentate granule cellGABAA receptors. J Neurosci 17:7532–7540.

Kapur J, Macdonald RL (1999) Postnatal development of hippocampaldentate granule cell gamma-aminobutyric acidA receptor pharmacolog-ical properties. Mol Pharmacol 55:444 – 452.

Kapur J, Haas KF, Macdonald RL (1999) Physiological properties ofGABAA receptors from acutely dissociated rat dentate granule cells.J Neurophysiol 81:2464 –2471.

Knoflach F, Benke D, Wang Y, Scheurer L, Luddens H, Hamilton BJ, CarterDB, Mohler H, Benson JA (1996) Pharmacological modulation of thediazepam-insensitive recombinant gamma-aminobutyric acidA recep-tors alpha 4 beta 2 gamma 2 and alpha 6 beta 2 gamma 2. Mol Pharmacol50:1253–1261.

Lin R, Heylbroeck C, Pitha PM, Hiscott J (1998) Virus-dependent phos-phorylation of the IRF-3 transcription factor regulates nuclear transloca-tion, transactivation potential, and proteasome-mediated degradation.Mol Cell Biol 18:2986 –2996.

Macdonald RL, Gallagher MJ, Feng HJ, Kang J (2004) GABAA receptorepilepsy mutations. Biochem Pharmacol 68:1497–1506.

Miralles CP, Gutierrez A, Khan ZU, Vitorica J, De Blas AL (1994) Differen-tial expression of the short and long forms of the gamma 2 subunit of theGABAA/benzodiazepine receptors. Brain Res Mol Brain Res 24:129 –139.

4902 • J. Neurosci., April 7, 2010 • 30(14):4895– 4903 Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR

Page 9: Cellular/Molecular TheShortSpliceVariantofthe ...(2% Triton X-100, 50 mM Tris-HCl, 150 mM NaCl, and 5 mM EDTA, pH7.5) supplemented with protease inhibitors (Complete, mini; Roche Diagnostics)

Mohler H, Knoflach F, Paysan J, Motejlek K, Benke D, Luscher B, Fritschy JM(1995) Heterogeneity of GABAA-receptors: cell-specific expression,pharmacology, and regulation. Neurochem Res 20:631– 636.

Mortensen M, Smart TG (2006) Extrasynaptic �� subunit GABAA recep-tors on rat hippocampal pyramidal neurons. J Physiol 577:841– 856.

Moss SJ, Doherty CA, Huganir RL (1992) Identification of the cAMP-dependent protein kinase and protein kinase C phosphorylation siteswithin the major intracellular domains of the beta 1, gamma 2S, andgamma 2L subunits of the gamma-aminobutyric acid type A receptor.J Biol Chem 267:14470 –14476.

Petrini EM, Marchionni I, Zacchi P, Sieghart W, Cherubini E (2004) Clus-tering of extrasynaptic GABAA receptors modulates tonic inhibition incultured hippocampal neurons. J Biol Chem 279:45833– 45843.

Richard JP, Melikov K, Vives E, Ramos C, Verbeure B, Gait MJ, ChernomordikLV, Lebleu B (2003) Cell-penetrating peptides. A reevaluation of the mech-anism of cellular uptake. J Biol Chem 278:585–590.

Sancar F, Czajkowski C (2004) A GABAA receptor mutation linked to hu-man epilepsy (�2R43Q) impairs cell surface expression of ��� receptors.J Biol Chem 279:47034 – 47039.

Saxena NC, Macdonald RL (1994) Assembly of GABAA receptor subunits:role of the delta subunit. J Neurosci 14:7077–7086.

Scotti AL, Reuter H (2001) Synaptic and extrasynaptic gamma-aminobutyricacid type A receptor clusters in rat hippocampal cultures during develop-ment. Proc Natl Acad Sci U S A 98:3489–3494.

Thompson SA, Bonnert TP, Cagetti E, Whiting PJ, Wafford KA (2002)Overexpression of the GABAA receptor epsilon subunit results in insen-sitivity to anaesthetics. Neuropharmacology 43:662– 668.

Trudell JR, Yue ME, Bertaccini EJ, Jenkins A, Harrison NL (2008) Molecu-lar modeling and mutagenesis reveals a tetradentate binding site for Zn 2�

in GABAA �� receptors and provides a structural basis for the modulatingeffect of the gamma subunit. J Chem Inf Model 48:344 –349.

Valente T, Auladell C, Perez-Clausell J (2002) Postnatal development ofzinc-rich terminal fields in the brain of the rat. Exp Neurol 174:215–229.

Wang JB, Burt DR (1991) Differential expression of two forms of GABAAreceptor gamma 2-subunit in mice. Brain Res Bull 27:731–735.

Whiting P, McKernan RM, Iversen LL (1990) Another mechanism for cre-ating diversity in gamma-aminobutyrate type A receptors: RNA splicingdirects expression of two forms of gamma 2 phosphorylation site. ProcNatl Acad Sci U S A 87:9966 –9970.

Wick MJ, Radcliffe RA, Bowers BJ, Mascia MP, Luscher B, Harris RA, WehnerJM (2000) Behavioural changes produced by transgenic overexpressionof gamma2L and gamma2S subunits of the GABAA receptor. Eur J Neu-rosci 12:2634 –2638.

Yeung JY, Canning KJ, Zhu G, Pennefather P, MacDonald JF, Orser BA(2003) Tonically activated GABAA receptors in hippocampal neuronsare high-affinity, low-conductance sensors for extracellular GABA. MolPharmacol 63:2– 8.

Zhang G, Raol YH, Hsu FC, Coulter DA, Brooks-Kayal AR (2004) Effects ofstatus epilepticus on hippocampal GABAA receptors are age-dependent.Neuroscience 125:299 –303.

Zhang SJ, Jackson MB (1993) GABA-activated chloride channels in secre-tory nerve endings. Science 259:531–534.

Zhang SJ, Jackson MB (1995) Properties of the GABAA receptor of rat pos-terior pituitary nerve terminals. J Neurophysiol 73:1135–1144.

Boileau et al. • Accessory Actions of the �2S Subunit of the GABAAR J. Neurosci., April 7, 2010 • 30(14):4895– 4903 • 4903