neuronally restricted rna splicing regulates the ... · neuronally restricted rna splicing...

11
Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional Properties Paul J. Whiting, 1 George McAllister, 1 Demetrios Vassilatis, 2 Timothy P. Bonnert, 1 Robert P. Heavens, 1 David W. Smith, 1 Louise Hewson, 1 Ruth O’Donnell, 1 Michael R. Rigby, 1 Dalip J. S. Sirinathsinghji, 1 George Marshall, 1 Sally A. Thompson, 1 and Keith A. Wafford 1 1 Neuroscience Research Centre, Merck Sharp & Dohme Research Laboratories, Harlow, Essex CM20 2QR, United Kingdom, and 2 Merck Research Laboratories, Rahway, New Jersey 07065 We report the isolation and characterization of a cDNA encod- ing a novel member of the GABA receptor gene family, e. This polypeptide is 506 amino acids in length and exhibits its great- est amino acid sequence identity with the GABA A receptor g3 subunit (47%), although this degree of homology is not suffi- cient for it to be classified as a fourth g subunit. The e subunit coassembles with GABA A receptor a and b subunits in Xeno- pus laevis oocytes and transfected mammalian cells to form functional GABA-gated channels. a1b1e GABA A receptors, like a1b1g2s receptors, are modulated by pentobarbital and the steroid 5a-pregnan-3a-ol-20-one but, unlike a1b1g2s recep- tors, are insensitive to flunitrazepam. Additionally, a1b1e recep- tors exhibit rapid desensitization kinetics, as compared with a1b1 or a1b1g2s. Northern analysis demonstrates widespread expression of a large e subunit transcript in a variety of non- neuronal tissues and expression of a smaller transcript in brain and spinal cord. Sequence analysis demonstrated that the large transcript contained an unspliced intron, whereas the small transcript represents the mature mRNA, suggesting reg- ulation of expression of the e subunit via neuronally restricted RNA splicing. In situ hybridization and immunocytochemistry reveal a pattern of expression in the brain restricted primarily to the hypothalamus, suggesting a role in neuroendocrine regula- tion, and also to subfields of the hippocampus, suggesting a role in the modulation of long term potentiation and memory. Key words: GABA A receptor; e subunit; function; RNA splic- ing; hypothalamus: hippocampus GABA is the major inhibitory neurotransmitter of the vertebrate CNS. It modulates inhibitory tone throughout the CNS by acti- vating two classes of receptors, GABA A and GABA B . The latter are G-protein-coupled receptors, and their molecular structure recently has been elucidated (Kaupmann et al., 1997). GABA A receptors are ligand-gated ion channels and are part of the same gene super family as nicotinic receptors (Schofield et al., 1989). The binding of the agonist GABA to the receptor complex results in the rapid opening of the intrinsic anion channel through which anions, primarily chloride, flow into the cell. This leads to hyper- polarization of the cell membrane and an increase in the inhibi- tory tone at that synapse. This receptor is the target for a number of classes of drugs, including the benzodiazepines, barbiturates, general anesthetics, neurosteroids, and alcohols (for review, see Macdonald and Angelotti, 1993; Sieghart, 1995; Whiting et al., 1995). Molecular biological approaches have revealed that the GABA A receptor exists as a gene family of polypeptides. These are divided into subcategories on the basis of their relative se- quence identities: a1–a6, b1–b4, g1–g4, d, and r1–r2 (see Darli- son and Albrecht, 1995; Sieghart 1995; Whiting et al., 1995). b4 and g4 have not been identified in mammals. The unique phar- macology of receptors formed by r1 or r2 has led to suggestions that they should be classified as GABA C receptors (Johnston, 1996). Each GABA A receptor subunit has a unique pattern of expression in the mammalian brain, although the g2 subunit is the most ubiquitous (Laurie et al., 1992; Wisden et al., 1992). Sub- units are thought to coassemble as pentamers to form a family of receptor subtypes that are expressed differentially throughout the mammalian brain. Experiments in a number of laboratories that use subunit-specific antibodies to characterize native GABA A receptors indicate that the minimum subunit composition of na- tive receptors is thought to be abg or abd (see Darlison and Albrecht, 1995; McKernan and Whiting, 1996). Studies using recombinant receptors have demonstrated that the subunit com- position determines the pharmacology of the receptor and the affinity for GABA (see Ebert et al., 1994; Whiting et al., 1995) and perhaps the targeting of the receptor to different subcellular domains (Connolly et al., 1996). However, relatively little is known about the biophysical properties of the various GABA A receptor subtypes. Here we report the identification of a novel GABA receptor subunit, e, which coassembles with a and b GABA A receptor subunits to form a functional receptor with unique properties. The expression of the mature form of e mRNA transcript seems to be regulated via a neuronally restricted RNA splicing mechanism. The limited expression of this subunit in hypothalamic and hip- pocampal regions suggests a specialized role in inhibitory neurotransmission. MATERIALS AND METHODS Isolation and sequencing of a cDNA encoding the GABA A receptor e subunit. The Merck.EST (Expressed Sequence Tag) database was Received Feb. 19, 1997; revised April 7, 1997; accepted April 10, 1997. Correspondence should be addressed to Dr. Paul J. Whiting, Neuroscience Re- search Centre, Merck Sharp & Dohme Research Laboratories, Eastwick Road, Harlow, Essex CM20 2QR, United Kingdom. Copyright © 1997 Society for Neuroscience 0270-6474/97/175027-11$05.00/0 The Journal of Neuroscience, July 1, 1997, 17(13):5027–5037

Upload: trandan

Post on 14-May-2018

221 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

Neuronally Restricted RNA Splicing Regulates the Expressionof a Novel GABAA Receptor Subunit Conferring AtypicalFunctional Properties

Paul J. Whiting,1 George McAllister,1 Demetrios Vassilatis,2 Timothy P. Bonnert,1 Robert P. Heavens,1David W. Smith,1 Louise Hewson,1 Ruth O’Donnell,1 Michael R. Rigby,1 Dalip J. S. Sirinathsinghji,1George Marshall,1 Sally A. Thompson,1 and Keith A. Wafford1

1Neuroscience Research Centre, Merck Sharp & Dohme Research Laboratories, Harlow, Essex CM20 2QR, UnitedKingdom, and 2Merck Research Laboratories, Rahway, New Jersey 07065

We report the isolation and characterization of a cDNA encod-ing a novel member of the GABA receptor gene family, e. Thispolypeptide is 506 amino acids in length and exhibits its great-est amino acid sequence identity with the GABAA receptor g3subunit (47%), although this degree of homology is not suffi-cient for it to be classified as a fourth g subunit. The e subunitcoassembles with GABAA receptor a and b subunits in Xeno-pus laevis oocytes and transfected mammalian cells to formfunctional GABA-gated channels. a1b1e GABAA receptors, likea1b1g2s receptors, are modulated by pentobarbital and thesteroid 5a-pregnan-3a-ol-20-one but, unlike a1b1g2s recep-tors, are insensitive to flunitrazepam. Additionally, a1b1e recep-tors exhibit rapid desensitization kinetics, as compared witha1b1 or a1b1g2s. Northern analysis demonstrates widespread

expression of a large e subunit transcript in a variety of non-neuronal tissues and expression of a smaller transcript in brainand spinal cord. Sequence analysis demonstrated that thelarge transcript contained an unspliced intron, whereas thesmall transcript represents the mature mRNA, suggesting reg-ulation of expression of the e subunit via neuronally restrictedRNA splicing. In situ hybridization and immunocytochemistryreveal a pattern of expression in the brain restricted primarily tothe hypothalamus, suggesting a role in neuroendocrine regula-tion, and also to subfields of the hippocampus, suggesting arole in the modulation of long term potentiation and memory.

Key words: GABAA receptor; e subunit; function; RNA splic-ing; hypothalamus: hippocampus

GABA is the major inhibitory neurotransmitter of the vertebrateCNS. It modulates inhibitory tone throughout the CNS by acti-vating two classes of receptors, GABAA and GABAB. The latterare G-protein-coupled receptors, and their molecular structurerecently has been elucidated (Kaupmann et al., 1997). GABAA

receptors are ligand-gated ion channels and are part of the samegene super family as nicotinic receptors (Schofield et al., 1989).The binding of the agonist GABA to the receptor complex resultsin the rapid opening of the intrinsic anion channel through whichanions, primarily chloride, flow into the cell. This leads to hyper-polarization of the cell membrane and an increase in the inhibi-tory tone at that synapse. This receptor is the target for a numberof classes of drugs, including the benzodiazepines, barbiturates,general anesthetics, neurosteroids, and alcohols (for review, seeMacdonald and Angelotti, 1993; Sieghart, 1995; Whiting et al.,1995).

Molecular biological approaches have revealed that theGABAA receptor exists as a gene family of polypeptides. Theseare divided into subcategories on the basis of their relative se-quence identities: a1–a6, b1–b4, g1–g4, d, and r1–r2 (see Darli-son and Albrecht, 1995; Sieghart 1995; Whiting et al., 1995). b4and g4 have not been identified in mammals. The unique phar-macology of receptors formed by r1 or r2 has led to suggestionsthat they should be classified as GABAC receptors (Johnston,

1996). Each GABAA receptor subunit has a unique pattern ofexpression in the mammalian brain, although the g2 subunit is themost ubiquitous (Laurie et al., 1992; Wisden et al., 1992). Sub-units are thought to coassemble as pentamers to form a family ofreceptor subtypes that are expressed differentially throughout themammalian brain. Experiments in a number of laboratories thatuse subunit-specific antibodies to characterize native GABAA

receptors indicate that the minimum subunit composition of na-tive receptors is thought to be abg or abd (see Darlison andAlbrecht, 1995; McKernan and Whiting, 1996). Studies usingrecombinant receptors have demonstrated that the subunit com-position determines the pharmacology of the receptor and theaffinity for GABA (see Ebert et al., 1994; Whiting et al., 1995) andperhaps the targeting of the receptor to different subcellulardomains (Connolly et al., 1996). However, relatively little isknown about the biophysical properties of the various GABAA

receptor subtypes.Here we report the identification of a novel GABA receptor

subunit, e, which coassembles with a and b GABAA receptorsubunits to form a functional receptor with unique properties. Theexpression of the mature form of e mRNA transcript seems to beregulated via a neuronally restricted RNA splicing mechanism.The limited expression of this subunit in hypothalamic and hip-pocampal regions suggests a specialized role in inhibitoryneurotransmission.

MATERIALS AND METHODSIsolation and sequencing of a cDNA encoding the GABAA receptor esubunit. The Merck.EST (Expressed Sequence Tag) database was

Received Feb. 19, 1997; revised April 7, 1997; accepted April 10, 1997.Correspondence should be addressed to Dr. Paul J. Whiting, Neuroscience Re-

search Centre, Merck Sharp & Dohme Research Laboratories, Eastwick Road,Harlow, Essex CM20 2QR, United Kingdom.Copyright © 1997 Society for Neuroscience 0270-6474/97/175027-11$05.00/0

The Journal of Neuroscience, July 1, 1997, 17(13):5027–5037

Page 2: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

searched with the human GABAA receptor b1 subunit-deduced (Gen-Bank accession number X14767) primary amino acid sequence, using theBLAST search tool (Altschul et al., 1990), and a number of EST se-quences were identified. Two of these, R07883 and R49718, were inves-tigated in more detail. PCR was performed to determine whether the twoESTs encoded the same gene product. For PCR, a sense primer wasgenerated from the R07883 sequence (59 ctgttggagtttggtgtgctcaac 39), andan antisense primer was generated from the R49718 sequence (59 ac-cagctggtacctacaagttaag 39). PCR was performed under standard condi-tions (Whiting et al., 1990), using human subthalamic nucleus cDNA(Clontech, Cambridge, UK) as a template.

cDNA sequences 59 of the R07883 sequence were obtained by 59 rapidamplification of cDNA ends (RACE), using the human brain MarathoncDNA cloning kit (Clontech) according to the manufacturer’s protocols.The nested antisense primers that were used were derived from theR07883 sequence (AS1, 59 catcgtggtcacggaagaagggac 39; AS2, 59 gccaaac-cgcctgctcacattgaa 39). PCR products were subcloned into pMOS vector(Amersham, Braunschweig, Germany), using standard techniques, andsequenced by using an Applied Biosystems (Foster City, CA) 373 DNAsequencer and dye terminator chemistry. One of the PCR products wasfound to extend far enough to contain a sequence encoding a putativeinitiating methionine and a 59 untranslated region (UT).

A full-length cDNA was generated by PCR with primers derived fromsequences in the 59 UT of the RACE PCR product and the 39 UTsequences in R49718 (59 caggtggtgcggccgctctccgcggaaatgttgt 39 and 59ccacagggcggccgctggtacctacaagttaag 39, both incorporating a NotI site forsubcloning). PCR products (1550 bp) were subcloned into pCDNAI/Ampand sequenced completely on both strands by primer walking. Sequenceanalysis was performed with Inherit (Applied Biosystems) and GeneticsComputer Group (University of Wisconsin) computer programs.

Northern blot analysis. Northern blots containing poly(A 1) RNA fromvarious human tissues were obtained from Clontech. The R07883 intronprobe was generated as a 283 bp EcoRI–BspHI fragment by digestion ofR07883 cDNA, and the R07883 exon probe was generated as a 365 bpBspHI–PacI fragment. 32P-labeled probes were generated by randompriming (Prime-It kit, Stratagene, La Jolla, CA), and blots were probedunder high stringency in 53 SSPE (13 SSPE is 0.18 M NaCl, 10 mM Naphosphate, pH 7.4, and 1 mM EDTA) containing 50% formamide at 42°C.Filters were washed in 0.33 SSPE at 65°C and exposed to Kodak XARfilm for 2 d at 270°C, using Cronex QIII intensifying screens.

Characterization of placental e subunit transcript. Placental cDNA (Clon-tech) was used as a template for PCR reactions. To generate PCR products59 and 39 of the intron, we used the following oligonucleotide primers: for the59 PCR, the sense primer was 59 gcggccgctctccgcggaaatgttgt 39 (bp 1–24) andthe antisense primer was 59 gggttgtgaattatttcagtt 39 (bp 776–795); for the 39PCR, the sense primer was 59 aactatgtcccttcttccgtg 39 (bp 867–887) and theantisense primer was 59 gcggccgctggtacctacaagttaag 39 (bp 1532–1552). PCRwas performed under standard conditions (Whiting et al., 1990), and prod-ucts were resolved by electrophoresis through 1% agarose gels stained withethidium bromide.

To generate PCR products containing the intron sequence, we used thefollowing oligonucleotide primers: sense, 59 agaactcctggaagctcttccagt 39(bp 727–750) and antisense, 59 ccaaaccgcctgctcacattgaa 39 (bp 828–850).PCR was performed with Expand Long Template PCR (BoehringerMannheim, Mannheim, Germany). PCR was performed by using buffer 1,supplied by manufacturers, with a final concentration of 350 mM de-oxynucleotide triphosphates, ;10 mM of each primer, and 0.75 U ofenzyme. PCR conditions were as follows: 2 min initial denaturation,followed by 35 cycles of 94°C for 20 sec, 60°C for 1 min, and 68°C for 3min. PCR products were resolved by electrophoresis through 0.8% aga-rose gels stained with ethidium bromide. PCR products were subclonedinto pMOS vector (Amersham), using standard methodologies and par-tial sequencing performed by primer walking, as above.

Cloning from human genomic DNA of e subunit sequences containing theunspliced intron. PCR was performed with rTth DNA polymerase (XLPCR, Perkin-Elmer, Oak Brook, IL) and 500 ng of human genomic DNA(Clontech) as a template. Buffer was as supplied by the manufacturer; thefinal concentration of other constituents was 200 mM deoxynucleotidetriphosphates, 1.1 mM magnesium acetate, ;10 mM of each primer, and 2U of enzyme. Conditions for PCR were as follows: 1 min denaturation at94°C, followed by 35 cycles each of 30 sec at 94°C and 3 min at 68°C. PCRproducts were resolved by electrophoresis through 0.8% agarose gelsstained with ethidium bromide. For subcloning, PCR products wereblunt-ended, using T4 DNA polymerase, purified by electrophoresis, and

ligated into EcoRV cut pBluescript (Stratagene). The PCR product wassequenced completely by primer walking, as above.

Localization of the e subunit in monkey brain by in situ hybridization.Antisense oligonucleotide probes to the human e subunit sequence weregenerated on an Applied Biosystems Model 394 DNA synthesizer andpurified by preparative polyacrylamide electrophoresis: probe 1, 59 ggt-gacaatcaggcacacaaaagcttcctgatgttggcggg caca 39 (antisense to bp 1206–1250); probe 2, 59 cctgctgccaggtactgccctcacaatcggg gaccatgcagaagt 39 (an-tisense to bp 1384–1428). Each oligonucleotide was 39 end-labeled with[ 35S]deoxyadenosine 59-(thiotriphosphate) in a 30:1 molar ratio of 35S-isotope/oligonucleotide, using terminal deoxynucleotidyl transferase for15 min at 37°C in the reaction buffer supplied (Boehringer Mannheim).Radiolabeled oligonucleotide was separated from unincorporated nucle-otides, using Sephadex G50 spin columns. The specific activities of thelabeled probes in several labeling reactions varied from 1.2–2.3 3 10 9

cpm/mg. Monkey brains were removed and fresh-frozen in 1 cm blocks.Sections of 12 mm were taken and fixed for in situ hybridization. Hybrid-ization of the sections was performed according to the method of Siri-nathsinghji and Dunnett (1993). Briefly, sections were removed fromalcohol and air-dried; 5 3 10 5 cpm of each 35S-labeled probe in 100 ml ofhybridization buffer was applied to each slide. Labeled “antisense” probealso was used in the presence of an excess (1003) concentration ofunlabeled antisense probe to define nonspecific hybridization. Parafilmcoverslips were placed over the sections that were incubated overnight(;16 hr) at 37°C. After hybridization the sections were washed for 1 hrat 57°C in 13 SSC and then rinsed briefly in 0.13 SSC, dehydrated in aseries of alcohols, air-dried, and exposed to Amersham Hyperfilm bmaxx-ray film. Autoradiographs were analyzed by a microcomputer imagingdevice (MCID) computerized image analysis system (Image Research,Ontario, Canada).

Localization of the e subunit in monkey brain and in transfected HEK 293cells by immunohistochemistry. A rabbit antiserum was generated to thesynthetic peptide I-V-T-T-E-G-S-D-G-E-E-R-P-S-C-S-A-Q-Q that wascoupled to keyhole limpet hemocyanin (Affiniti Research Products, Ex-eter, UK). This sequence is residues 409–427 of the e subunit, located inthe putative cytoplasmic loop between TM3 and TM4. For immunohis-tochemistry, antibodies were affinity-purified by using the synthetic pep-tide coupled to Sepharose.

A squirrel monkey was deeply anesthetized with ketamine and sodiumpentobarbitone and transcardially perfused with saline, followed by 10%formalin in 0.1 M PBS. The brain was removed, post-fixed in 10%formalin in PBS for 24 hr, and sliced into coronal blocks, which then wereembedded in paraffin wax. Coronal sections (10 mm) were cut on a rotarymicrotome, deparaffinized, rinsed in PBS, and treated with 0.3% H2O2for 30 min (to block endogenous peroxidase activity). Background stain-ing was blocked by incubating the sections in 3% normal horse serum for1 hr. Sections were incubated overnight at 4°C with the anti-e subunitrabbit polyclonal antibody (1:500 dilution). Immunostaining was visual-ized by using the ABC elite system (Vector Laboratories, Peterborough,UK), followed by development in diaminobenzidine (DAB). Finally,sections were counterstained in Gill’s hematoxylin, dehydrated, andmounted for microscopical examination. For immunofluorescent labelingof transiently transfected HEK 293 cells, the procedure was, in general,similar to that used for brain sections. Cells were transfected with cDNAs

Figure 1. Schematic representation of a GABAA receptor subunit indi-cating the relative positions of the ESTs. SP, Signal peptide. 1–4, Putativetransmembrane regions 1–4. The hatched area of the box representingEST R07883 is an intron sequence. The boxes open at one end represent-ing ESTs R49718, R19732, T27015, and T78142 indicate that the 39 end ofthese sequences has not been determined.

5028 J. Neurosci., July 1, 1997, 17(13):5027–5037 Whiting et al. • GABA Receptor e Subunit Cloning and Characterization

Page 3: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

Figure 2. Comparison of the deduced amino acid sequence of the esubunit with sequences of other GABAA receptor subunits. A, Alignmentof the deduced amino acid sequences of the human GABAA receptor a1(Schofield et al., 1989), b1 (Schofield et al., 1989), g1 (Ymer et al., 1990),d (P. Whiting, unpublished data), and e subunits. Positions in which aminoacid residues are conserved in four or more sequences are boxed. Num-bering of amino acids is given by assigning the initiating methionine as 1.Putative transmembrane regions TM1–TM4 are indicated by a solid line,and the two cysteine residues conserved in the ligand-gated ion channelfamily are indicated by filled circles joined by a dotted line. B, Dendrogramof the deduced amino acid sequences of the GABAA receptor family,including the e subunit. The analysis was performed by using PileUp(Genetics Computer Group, University of Wisconsin). The distance alongthe horizontal is proportional to the differences between the sequences.

Whiting et al. • GABA Receptor e Subunit Cloning and Characterization J. Neurosci., July 1, 1997, 17(13):5027–5037 5029

Page 4: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

as described below and 48 hr later were processed for immunocytochem-istry. The cells were fixed in 4% paraformaldehyde, rinsed in PBS, andincubated with the anti-e subunit antibody at a dilution of 1:1000. Goatanti-rabbit fluorescein isothiocyanate (FITC; 1:100 dilution) was used asthe detection system.

Expression in Xenopus oocytes. Adult female Xenopus laevis wereanesthetized by immersion in a 0.4% solution of 3-aminobenzoic acidethylester for 30–45 min (or until unresponsive). Ovary tissue was re-moved via a small abdominal incision, and Stage V and Stage VI oocyteswere isolated with fine forceps. After mild collagenase treatment toremove follicle cells (Type IA, 0.5 mg/ml, for 8 min), the oocyte nucleiwere injected directly with 10–20 nl of injection buffer (88 mM NaCl, 1mM KCl, and 15 mM HEPES, at pH 7, filtered through nitrocellulose) orsterile water containing different combinations of human GABAA sub-unit cDNAs (20 ng/ml) engineered into the expression vector pCDM8 orpcDNAI/Amp. After incubation for 24–72 hr, oocytes were placed in a 50ml bath and perfused at 4–6 ml/min with modified Barth’s medium(MBS) consisting of (in mM): 88 NaCl, 1 KCl, 10 HEPES, 0.82 MgSO4 ,0.33 Ca(NO3 )2 , 0.91 CaCl2 , and 2.4 NaHCO3 , at pH 7.5. Cells wereimpaled with two 1–3 MV electrodes containing 2 M KCl and voltage-clamped between 240 and 270 mV.

In all experiments, drugs were applied in the perfusate until the peakof the response was observed. Noncumulative concentration–responsecurves to agonists were constructed, allowing at least 3 min between eachagonist application to prevent desensitization. Curves were fit by using anonlinear square-fitting program to the equation f(x) 5 BMAX /[11(EC50 /x)n], where x is the drug concentration, EC50 is the concentration of drugeliciting a half-maximal response, and n is the Hill coefficient. The effectsof GABAA receptor modulators were examined on control GABA EC20responses with a preapplication time of 30 sec.

Whole-cell patch clamp of HEK 293 cells transiently transfected withhuman GABAA receptors. Experiments were performed on HEK 293 cellstransiently transfected with human cDNA combinations a1b1, a1b1e,and a1b1g2s (6 mg of cDNA total per coverslip), using calcium phosphateprecipitation (Chen and Okayama, 1988) as previously described (Had-ingham et al., 1993). Glass coverslips containing the cells in a monolayerculture were transferred to a chamber on the stage of a Nikon Diaphotinverted microscope. Cells were perfused continuously with a solutioncontaining (in mM): 124 NaCl, 2 KCl, 2 CaCl2 , 1 MgCl2 , 1.25 KH2PO4 ,25 NaHCO3 , and 11 D-glucose, pH 7.2, and observed with phase-contrastoptics. Patch pipettes were pulled with an approximate tip diameter of 2mm and a resistance of 4 MV with borosilicate glass and were filled with(in mM): 130 CsCl, 10 HEPES, 10 EGTA, and 3 Mg 1-ATP, pH-adjustedto 7.3 with CsOH. Cells were patch-clamped in whole-cell mode with aList LM-EPC 7 patch-clamp amplifier (List Biologic, Campbell, CA).Drug solutions were applied by a double-barreled pipette assembly, whichwas controlled by a stepping motor attached to a Prior manipulator,enabling rapid equilibration around the cell. Increasing GABA concen-trations were applied for 10 sec pulses with a 30 sec interval betweenapplications.

RESULTSIdentification of a novel member of the GABAreceptor familySo that new GABA receptor subunits genes could be identified,the Expressed Sequence Tag (EST) database was searched, usingthe primary amino acid sequence of the human GABAA receptorb1 subunit (Schofield et al., 1989) as a query. Two ESTs, R07883(from a human fetal liver/spleen cDNA library) and R49718 (froma human infant brain cDNA library), were identified that exhib-ited significant deduced amino acid sequence identity with the b1subunit but that were not any known GABA receptor subunit.Further searching of the EST database using R49718 as a queryrevealed three more ESTs that overlapped with R49718 (Fig. 1).PCR experiments that used primers derived from R07883 andR49718 demonstrated that they were derived from the same gene(data not shown). EST R07883 is 650 bp in length, and furtheranalysis suggested that the 59 270 bp of this cDNA was either acloning artifact or, more likely, an intron sequence. Indeed, theputative intron–exon boundary (Fig. 1) is at exactly the sameposition as the 59 splice site of exon 7 of other GABAA receptor

genes such as b1 (Kirkness et al., 1991) and d (Sommer et al.,1990), and subsequent cloning of genomic DNA confirmed this270 bp to be an intron sequence (see below). 59 RACE fromhuman brain cDNA was used to obtain cDNA sequences of the 59end of the R07883 gene product. Then a full-length cDNA wasobtained by PCR with oligonucleotide primers derived from the 59untranslated region sequence of 59 RACE-generated cDNAs andthe 39 untranslated region nucleotide sequence from R49718. TwocDNAs were sequenced in full on both strands, as well as numer-ous RACE cDNA products.

Primary structure of the e subunitThe deduced primary amino acid sequence of the R07883 geneproduct, which we have termed e, is shown in Figure 2A, alignedwith other members of the GABAA receptor gene family. The esubunit has an open reading frame of 506 amino acids containingthe motifs expected of a member of the ligand-gated ion channelsuper family exemplified by the nicotinic receptor: an 18 residuesignal peptide [using the prediction of von Heijne (1986)], twocysteine residues separated by 13 amino acids, and four hydro-phobic regions, which are putative transmembrane-spanning do-mains (TM1–TM4). The putative extracellular domain containsthree potential N-glycosylation sites (Asn residues 134, 252, and272). The most conserved regions are the putative transmembranedomains, whereas the putative cytoplasmic loop between TM3and TM4 is completely divergent. The e subunit has its highestamino acid sequence identity with the g subunits (42–47% iden-tity with g1–g3) (Pritchett et al., 1989; Ymer et al., 1990; Hading-ham et al., 1995). It exhibits 49% sequence identity with chickeng4 (Harvey et al., 1993); the human homolog of this avian subunithas yet to be identified. However, because the amino acid se-quence conservation of other GABAA receptor subunits betweenspecies is .90%, e.g., between chicken g2 (Glencorse et al., 1990)and human g2 (Pritchett et al., 1989) it is 92%, the e subunit is notthe human homolog of g4. The analysis shown in Figure 2Bindicates that e represents a novel subfamily of GABA receptorsubunits.

Characterization of e subunit transcriptsNorthern blot analysis (Fig. 3) that used a probe derived from thecoding region (i.e., exon sequences) of the e subunit revealedexpression of a large transcript (;5.5 kb) in numerous peripheraltissues (placenta, pancreas, spleen, prostate, testis, ovary, andintestine) as well as a smaller transcript (;3.5 kb) in various brainregions, although it was not detectable in whole-brain mRNA.Spinal cord and also the heart appear to express both transcripts.When Northern blots were probed with the intron sequenceidentified in the R07883 EST (Fig. 1), only the large transcript wasidentified, suggesting that the large mRNA species representsincompletely spliced transcripts.

The widespread expression of the incompletely spliced e RNAspecies and the more restricted expression of the smaller RNAspecies suggested that the expression of the mature e transcript iscontrolled, at least in part, via a tissue-specific RNA splicingmechanism. To investigate this further, we characterized thegenomic sequence around the putative incompletely spliced in-tron sequence, as well as the large transcript expressed in pla-centa. PCR from genomic DNA with primers derived from theexon sequence on either side of the putative intron yielded aproduct of ;4000 bp (Fig. 4A). Complete sequencing of the PCRproduct (Fig. 4B) indicated that the intron is 3991 bp and that the39 splice site is identical to the sequence identified in the EST

5030 J. Neurosci., July 1, 1997, 17(13):5027–5037 Whiting et al. • GABA Receptor e Subunit Cloning and Characterization

Page 5: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

R07883. PCR from placenta cDNA with the same primer pairyielded two products of ;3300–3500 bp (Fig. 4A). In both ofthese cDNAs the intronic sequence starts at a presumed crypticsplice site 692 bp from the 59 end of the genomic intron sequence.The smaller PCR product also contains an additional deletion of129 bp, as compared with the genomic intron sequence (Fig. 4B).These data suggest that in peripheral tissues such as placenta the59 splice donor of this intron is used, whereas the 39 spliceacceptor is not. As may be expected, the intron sequence retainedin the placental transcripts contained numerous stop codons,indicating that it would not lead to translation of a GABAA

receptor subunit open reading frame. Additional PCR experi-ments were performed with primer pairs flanking the intron–exonsplice site and in the 59 or 39 untranslated region to determine thestructure of the 59 and 39 end of the placental e subunit RNA. A59 PCR product of ;800 bp was generated, which is the predictedsize (798 bp) if this region were spliced appropriately so as tocontain only exon sequences (data not shown). Similarly, a 39 PCRproduct of ;700 bp was generated, in agreement with the 686 bpsize predicted if this region were spliced appropriately so as tocontain only exon sequences (data not shown). These data suggestthat in placental RNA only the intron between exons tentativelyassigned as 6 and 7 has not been spliced out.

The regional expression of e subunit transcripts inthe brainIn the absence of human brain material, the expression of etranscripts was determined in the squirrel monkey brain by in situhybridization with 35S-labeled oligonucleotide probes. PCR andsequencing experiments demonstrated that nucleotide sequenceschosen for synthesis of oligonucleotide probes were conserved

between human and monkey (data not shown). The e subunittranscript was highly localized, primarily in the hypothalamus,with high densities of expression principally within the arcuate–ventromedial area (ARC–VMH); much weaker expression of thetranscript was detected in the dorsomedial hypothalamus (Fig.5A). Dense mRNA expression also was found in the hilus of thedentate gyrus of the hippocampus (Fig. 5B). It is of interest tonote that no expression was detected in other hippocampal fieldsnor in the amygdala or subthalamic nucleus, two regions that,according to the Northern blot analysis, contained transcripts.This discrepancy may reflect the difficulties in accurately dissect-ing human brain regions for RNA purification. Immunohisto-chemical labeling of squirrel monkey brain sections was per-formed with a polyclonal antiserum generated to a syntheticpeptide derived from amino acids 409–427 of the e subunitsequence. Immunohistochemical staining with the e subunit anti-body showed a distribution of immunoreactivity correspondingwith the mRNA localization (Fig. 5C,D). Figure 5C shows labelingof magnocellular cells and numerous smaller cells within thedorsomedial hypothalamus, and Figure 5D shows labeling of cellswithin the hilus and CA3 region of the hippocampus. Othersubfields of the hippocampus showed minimal specific labeling.The immunohistochemical staining patterns obtained with the esubunit antibody were specific because preabsorption experimentson adjacent brain sections with the peptide gave no labeling (datanot shown). Similarly, immunofluorescent labeling with the esubunit antibody of HEK 293 cells transiently transfected with a1,b1, and e subunits showed positively labeled cells (Fig. 5E), thepattern of staining indicating membrane association of the sub-unit. Nontransfected cells gave no labeling (Fig. 5F).

Figure 3. Expression of e subunit transcripts in human tissues by Northern blot analysis. Each lane contains 2 mg of poly(A 1) RNA purified from varioushuman tissues and resolved through 1.2% agarose in formaldehyde (Clontech). Blots were hybridized with 32P-labeled intron or exon cDNA probes, asdetailed in Materials and Methods. RNA size marker bands are indicated on the left of the figure.

Whiting et al. • GABA Receptor e Subunit Cloning and Characterization J. Neurosci., July 1, 1997, 17(13):5027–5037 5031

Page 6: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

Functional expression of e subunit containingGABAA receptor

To investigate whether the e subunit could form functional GABAgated channels, we expressed it in Xenopus oocytes, either aloneor in combination with other members of the GABAA receptorgene family. When expressed alone, e failed to show any responseto applied GABA, indicating that this subunit is unable to formfunctional homomeric channels. Similarly, no functional channelswere formed when e was coexpressed in oocytes with an a1subunit or a b1 subunit. Coexpression with both an a1 and a b1subunit resulted in the formation of functional GABA-gatedchannels. However, because a1 and b1 are able to coassemble toform functional receptors, further pharmacological analysis wasrequired. Maximum current sizes for all three subunit combina-tions were similar (Table 1). GABA concentration–effect curveswere generated to a1b1, a1b1g2s, and a1b1e (Fig. 6a). The EC50

for GABA was similar for both a1b1 and a1b1e subunit combi-nations [5.9 6 1.1 mM (n 5 7) and 4.0 6 1.2 mM (n 5 8),respectively]. The Hill slope, however, was significantly lower forreceptors containing an e subunit (0.85 6 0.08 compared with1.32 6 0.06 for a1b1; Table 1). This was found for receptorsexpressed in oocytes or in HEK 293 cells and is in contrast to thatobserved for native GABAA receptors (Sakmann et al., 1983) andother recombinant GABAA receptor subtypes so far examined

(Horne et al., 1993; Verdoorn, 1994; Hadingham et al., 1996;Saxena and Macdonald, 1996), for which a Hill slope of .1 isobserved. This suggested that e was coassembling with a1 and b1.Definitive evidence for coassembly of the e subunit came fromexamining the inhibition of various subunit combinations by zinc.Zn 21 ions are 175-fold more potent at ab than at abe combina-tions (Fig. 6b, Table 1), demonstrating that e was coassemblingwith a1 and b1 to form a GABAA receptor with low affinity forzinc. Additionally, the zinc inhibition curve could be fit to a singlesite, suggesting that a homogeneous population of abe receptors(rather than a mixed population of ab and abe) was beingformed.

The ability of different GABAA receptor modulators to act ata1b1e was investigated (Fig. 7). Unlike receptors containing a g2ssubunit, the GABA currents of a1b1e receptors were not poten-tiated by the benzodiazepine flunitrazepam (1 mM). The barbitu-rate pentobarbital (100 mM) potentiated a1b1e to the same extentas a1b1g2s (200–300% of GABA EC20), but this was significantlylower than that observed with a1b1 receptors (Fig. 7a). Fullconcentration–response curves for the steroid 5a-pregnan-3a-ol-20-one on all three receptor subunit combinations revealed asimilar maximum potentiation of ;250% on all, but a significantlyhigher affinity of 78 nM ( p , 0.05) on a1b1, as compared with 194nM on a1b1e and 114 nM on a1b1g2s (Fig. 7b, Table 1).

Figure 4. Cloning of e subunit genomic sequences containing the intron between exons 6 and 7 and characterization of placental e subunit transcripts.A, Generation of products containing unspliced intron by PCR, using placenta cDNA or genomic DNA as a template. PCR primers (indicated by arrows)were derived from exon sequences flanking the intron. DNA size markers are indicated to the right of the figure. The single line represents 59 and 39 UT,the hatched box represents the coding region, and the open box with dashed lines represents the intron. B, Nucleotide sequence of the intron between exons6 and 7 cloned from genomic DNA and from two cDNAs cloned from placental mRNA. The deduced amino acid sequence of the surrounding exon isshown also, with the amino acid number indicated above it.

5032 J. Neurosci., July 1, 1997, 17(13):5027–5037 Whiting et al. • GABA Receptor e Subunit Cloning and Characterization

Page 7: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

The current–voltage relationship for a1b1e receptors was notsignificantly different from receptors consisting of a1b1 (data notshown). Maximum GABA responses in oocytes expressing a1b1eappeared to show a greater rate of desensitization than thoseexpressing a1b1 or a1b1g2s. To investigate this further, we trans-

fected these subunits into HEK 293 cells and studied them byusing whole-cell patch-clamp techniques in which rapid solutionchanges allowed a more accurate measure of desensitization ki-netics. With this method it was clear that e was conferring rapiddesensitization to the receptor, as compared with a1b1 and

Figure 5. In situ hybridization autoradiograms and immunohistochemical staining of squirrel monkey brain sections and transfected HEK 293 cellsshowing expression of the expression of e subunit. A, Dense mRNA expression in the arcuate–ventromedial area ( ARC–VMH ) and weaker expressionin the dorsomedial hypothalamus (DMH ). The signal intensity is indicated in the scale bar in B, with white representing the strongest signal. B, DensemRNA expression in the hilus ( H ) of the dentate gyrus of the hippocampus. C, Immunoreactive cells in the DMH. Arrowhead indicates a labeledmagnocellular neuron. Brown is the immunostaining reaction product, and purple is the hematoxylin counterstain. D, Immunoreactivity in the hilus of thedentate gyrus and in the CA3 region of the hippocampus. E, e subunit immunofluorescent labeling of HEK 293 cells transiently transfected with a1, b1,and e subunits. F, Lack of labeling in untransfected HEK 293 cells. Scale bars: A, B, 0.5 cm; C, 10 mm; D, 100 mm. E, Magnification, 1003.

Whiting et al. • GABA Receptor e Subunit Cloning and Characterization J. Neurosci., July 1, 1997, 17(13):5027–5037 5033

Page 8: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

a1b1g2s (Fig. 8). At a concentration of 30 mM, which was maxi-mum for all combinations, the peak/steady-state (after 10 sec)ratios were 4.22 6 0.5 (n 5 8) for a1b1e, 2.17 6 0.2 (n 5 9) fora1b1, and 2.18 6 0.3 (n 5 6) for a1b1g2s, the a1b1e beingsignificantly higher than the other two combinations ( p , 0.005).Fitting single exponentials to the desensitization phase revealedrate constants of 1.79 6 0.33, 4.23 6 0.83, and 4.55 6 1.2 sec fora1b1e, a1b1, and a1b1g2s, respectively. Rapid desensitizationmay affect the GABA concentration–response curve in oocytes,and the slightly higher affinities determined by using patch clampsuggest that this may be the case; however, the GABA EC50

values and Hill coefficients showed a similar relationship amongthe different subunit combinations to that observed in oocytes(data not shown).

DISCUSSIONe coassembles with other GABAA receptor subunits toform a functional channelBased on deduced amino acid sequence comparisons, the e subunithas 32–49% sequence identity with other GABAA receptor polypep-tides and thus is a member of the GABAA receptor gene family. edoes not form functional channels when expressed alone. Similarly,when coexpressed with other GABAA receptor subunits, it is unableto substitute for an a or b subunit. However, when it is coexpressedwith both an a and a b subunit, a channel with a unique pharmacol-ogy is formed, demonstrating coassembly of the three subunits into areceptor complex. Most GABAA receptors studied to date arethought to consist of a, b, and g or d subunits (see Darlison andAlbrecht, 1995; McKernan and Whiting, 1996). Thus e seems to beable to substitute for a g or d subunit. Detailed immunoprecipitationexperiments will be required to determine the subunit composition ofnative e-containing receptors.

Zinc ions are known to be negative modulators of GABAA

receptor function (Smart and Constanti, 1990), and indeed theyhave been shown to be more potent at receptors consisting of aand b subunits than those constituted by a, b, and g subunits(Draguhn et al., 1990; Smart et al., 1991) (Fig. 6b). Similarly, thesubunit combination a1b1e had 175-fold lower affinity for zincthan receptors composed of ab. A lowering of the potency of zincis observed also when the d subunit is coexpressed with a and b;zinc has a 20-fold lower affinity at a6b3d, as compared with a6b3receptors (Thompson et al., 1997). Thus, inclusion of a thirdsubunit into a binary ab complex appears to have the generalproperty of lowering the potency for zinc.

a1b1e receptors are not modulated by the benzodiazepineflunitrazepam. This is perhaps not surprising because it is thoughtthat the benzodiazepine binding site is made up of determinantsfrom both the a and g subunits (Pritchett et al., 1989; Stephensonet al., 1990). Similarly, GABAA receptors made up of a, b, and d

Figure 6. Functional expression of receptors containing the e subunit. a,Concentration–response curves to GABA of a1b1, a1b1g2s, and a1b1eGABAA receptors expressed in Xenopus oocytes. b, Concentration–re-sponse curves for inhibition of an EC50 concentration of GABA byincreasing concentrations of zinc. Data represent mean curves from thenumber of cells indicated for each subtype. Curves were fit as described inMaterials and Methods, with the exception of a1b1g2s, which was best fitwith two components of inhibition.

Table 1. Comparison of the properties of a1b1, a1b1e, and a1b1g2s GABAA receptors expressed in Xenopus oocytes

a1b1 a1b1e a1b1g2s

GABA EC50 5.9 6 1.1 mM (n 5 7) 4.0 6 1.2 mM (n 5 8) 19.9 6 4.6 mM (n 5 7)Hill slope 1.32 6 0.06 0.85 6 0.08 1.36 6 0.18Mean maximum current (nA) 1307 6 224 (n 5 13) 973 6 78 (n 5 15) 1519 6 134 (n 5 10)Zinc IC50 0.24 6 0.12 mM (n 5 4) 41.9 6 3.6 mM (n 5 4) 712 6 296 mM (n 5 6)5a-Pregnan-3a-ol-20-one EC50 78 6 10.9 nMb 194 6 12 nM 114 6 21 nM

5a-Pregnan-3a-ol-20-one potentiationa 265 6 38% (n 5 6) 260 6 45 (n 5 4) 214 6 35 (n 5 4)

aMaximum potentiation of an EC20 concentration of GABA.bSignificantly different from a1b1g2s and a1b1e, p , 0.05.

5034 J. Neurosci., July 1, 1997, 17(13):5027–5037 Whiting et al. • GABA Receptor e Subunit Cloning and Characterization

Page 9: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

subunits are reported not to be modulated by benzodiazepines(Saxena and Macdonald, 1994).

The barbiturate pentobarbital interacts with the GABAA re-ceptor at both a modulatory site and a directly activating site, andthe affinity and efficacy of pentobarbital for direct activation showsubunit selectivity, being higher at a6-containing (a6b2g2s) re-ceptors, as compared with receptors containing other a subunits(Thompson et al., 1996). Here we show that the potentiation by100 mM pentobarbital was not significantly different betweena1b1e and a1b1g2s receptors. However, the potentiation at a1b1receptors by the same concentration of pentobarbital was signif-icantly higher than at either of the trimeric receptors, suggestingthat the inclusion of a third subunit lowers the efficacy. Pentobar-bital (100 mM) elicited a small response in the absence of GABAat a1b1e, but not at a1b1 or a1b1g2s receptors (data not shown).

Concentration–response curves for the steroid 5a-pregnan-3a-ol-20-one demonstrate an equivalent maximum level of potentia-tion on all three subunit combinations; however, the ab assembly

shows a small but significantly higher affinity for the steroid overa1b1e or a1b1g2s. The location of the binding sites for bothbarbiturates (Thompson et al., 1996) and steroids (Lambert et al.,1995) currently is unknown, but it is clearly not critically depen-dent on the presence of g, d, or e subunits. The cloning of the esubunit recently has been reported by Davies et al. (1997), whoalso reported insensitivity of e-containing receptors to pentobar-bital and pregnenolone; however, the clear potentiation observedhere with both of these agents suggests that e does not conferinsensitivity to anesthetics. The reason for this discrepancy iscurrently unclear, because it is unlikely that the different a and bsubunits used in the study could account for this and it is unknownwhich subunits coassemble with e in vivo. The deduced amino acidsequence of the subunit reported by Davies et al. (1997) differs atposition 102 (an alanine in the sequence of Davies et al. (1997), aserine in the sequence described here), but this difference doesnot account for the pharmacological inconsistencies (data notshown).

A novel observation was the rapid rate of desensitization ofa1b1e receptors, as compared with a1b1 or a1b1g2s (Fig. 8). Thee subunit clearly confers rapid desensitization kinetics to theGABAA receptor. In contrast, the presence of a d subunit (ina1b1d receptors) appears to decrease the rate of GABA-induceddesensitization (Saxena and Macdonald, 1994).

Expression of the e transcript in brain ishighly restrictedOnly the r subunits, the expression of which is primarily retinal (Enzet al., 1995), and a6, the expression of which is limited to cerebellargranule cells (Luddens et al., 1990; Hadingham et al., 1996), have amore restricted pattern of expression than the e subunit. The mostabundant expression of the mRNA was found within the hypotha-lamic region, especially within the arcuate–ventromedial area. OtherGABAA receptor subunits are known to be expressed in hypotha-lamic nuclei, primarily, but not exclusively, a2, b3, and g2 (Wisden etal., 1992; Fenelon and Herbison, 1995; Fenelon et al., 1995).GABAergic transmission is known to play a key role in the hypo-thalamus, modulating the synthesis or release of vasopressin andoxytocin (Bisset et al., 1990), somatostatin (Gillies and Davidson,1992), luteinizing hormone-releasing hormone (Mitsushima et al.,1994), and pro-opiomelanocortin (Blazquez et al., 1994). Colocaliza-tion studies that use anti-e subunit and antipeptide hormone antiserawill be required to define in detail a role for e-containing receptors inthe modulation of hormonal systems. Interestingly, the other site ofhigh e subunit mRNA expression was found in the hilus (polymor-phic layer) of the dentate gyrus of the hippocampus. There was alsolow expression in the granule cell layer, but no other hippocampalfield showed expression. Immunohistochemical labeling of monkeybrain sections showed slight immunoreactivity in cells of the granulelayer but dense immunoreactivity in the polymorphic layer and in theCA3 region. This is consistent with labeling of the mossy fibers in thepathway from dentate to CA3 and indicates that the e subunit mayplay a specific role in the modulation of this excitatory pathway, withimportance to long-term potentiation and memory.

e subunit expression is regulated in part by tissue-specific RNA splicingNorthern analysis revealed that the e subunit was expressed as twomajor polyadenylated transcripts. The larger transcript was ex-pressed in a variety of peripheral tissues, but at barely detectablelevels, in the brain. The smaller transcript was expressed withinthe brain but, with the exception of the heart and spinal cord, was

Figure 7. Effects of 100 mM pentobarbital, 1 mM flunitrazepam, and5a-pregnan-3a-ol-20-one on a1b1, a1b1g2s, and a1b1e GABAA receptorsubunit combinations in Xenopus oocytes. a, Potentiation by 100 mMpentobarbital and 1 mM flunitrazepam. Data represent the percentage ofmodulation of an EC20 GABA response determined for each individualoocyte and are the mean 6 SE of at least four determinations. b, Poten-tiation of the response to a GABA EC20 by increasing concentrations of5a-pregnan-3a-ol-20-one on oocytes expressing a1b1 (F), a1b1g2s (M),and a1b1e (E) GABAA receptors. Data represent mean 6 SE of at leastfour individual concentration–response curves, and values calculated fromthis are shown in Table 1.

Whiting et al. • GABA Receptor e Subunit Cloning and Characterization J. Neurosci., July 1, 1997, 17(13):5027–5037 5035

Page 10: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

not detectable in peripheral tissues. cDNA cloning revealed thatthe smaller transcript was appropriately spliced mature mRNA,whereas the larger transcript contained an unspliced intron be-tween putative exons 6 and 7. These data suggest that the molec-ular mechanism for appropriate splicing of this particular intron ispresent within neurons but largely absent in peripheral tissues.Furthermore, because there is abundant expression of the un-spliced transcript in a number of peripheral tissues, the neuronal-specific expression of mature e subunit transcript is being regu-lated via a neuronally restricted RNA splicing mechanism. Othermechanisms, such as alternative splicing (for review, see Lewin,1994) and RNA editing (Sommer et al., 1991), are known bywhich expression of the gene product can be regulated at the levelof the RNA. Indeed, tissue-specific alternative RNA splicing hasbeen described for the synaptic terminal protein synaptojanin(Ramjaun and McPherson, 1996) and the Pem homeobox gene(Maiti et al., 1996). The alternative splicing of neural cell adhesionmolecule (NCAM) mRNA, which leads to the inclusion of exon18 in differentiated N2a neuroblastoma cells and the omission inundifferentiated cells, has been characterized in detail and shownto depend on the 59 splice site of the following intron and iscontrolled presumably by trans-acting factors (Tacke and Goridis,1991).

Splicing is obviously not the only mode of regulation of the esubunit transcript expression; the discrete regional expression ofthe e subunit transcript within the brain presumably is beingcontrolled at the level of the promoter. Two tissues, heart andspinal cord, expressed both the unspliced and the mature mRNAspecies. The precise cellular localization of the two mRNAs withinthese tissues is not known, but it is possible that the appropriatelyspliced mature mRNA is restricted to neuronal cells. GABAergicinhibition via GABAA receptors is known to be important in thespinal cord, and indeed several GABAA receptor subunits, par-

ticularly a2, b3, and g2, are known to be expressed in this tissue(Persohn et al., 1991).

REFERENCESAltschul SF, Gish W, Miller W, Myers EW, Lipman D (1990) Basic local

alignment search tool. J Mol Biol 215:403–410.Blazquez C, Jegou S, Feuillololey M, Rosier A, Vandesande F, Vaudry H

(1994) Visualization of g aminobutyric acid-A receptors on pro-opiomelanocortin-producing neurons in the rat hypothalamus. Endo-crinology 135:2759–2764.

Bisset GW, Chowdrey HS, Fairhall KM, Gunn LK (1990) Central inhi-bition by gamma-aminobutyric acid and muscimol of the release ofvasopressin and oxytocin by an osmotic stimulus in the rat. Br J Phar-macol 99:529–535.

Chen CA, Okayama H (1988) Calcium phosphate-mediated gene trans-fer: a highly efficient transfection system for stably transforming cellswith plasmid DNA. Biotechniques 6:632–638.

Connolly CN, Wooltorton JRA, Smart TG, Moss SJ (1996) Subcellularlocalization of g-aminobutyric acid type A receptors is determined byreceptor b subunits. Proc Natl Acad Sci USA 93:9899–9904.

Darlison MG, Albrecht BE (1995) GABAA receptors; which, where, andwhy? Semin Neurosci 7:115–126.

Davies PA, Hanna MC, Hales TG, Kirkness EF (1997) Insensitivity toanaesthetic agents conferred by a class of GABAA receptor subunit.Nature 385:820–823.

Draguhn A, Verdoorn TA, Ewert M, Seeburg PH, Sakmann B (1990)Function and molecular distinction between recombinant rat GABAAreceptor subtypes by Zn 21. Neuron 5:781–788.

Ebert B, Wafford KA, Whiting PJ, Krogsgaard-Larsen P, Kemp JA (1994)Molecular pharmacology of g-aminobutyric acid type A receptor ago-nists and partial agonists in oocytes injected with different a, b, and greceptor subunit combinations. Mol Pharmacol 46:957–963.

Enz R, Brandstatter JH, Hartveit E, Wassle H, Bormann J (1995)Expression of GABA receptor r1 and r2 subunits in the retina and brainof the rat. Eur J Neurosci 7:1495–1501.

Fenelon VS, Herbison AE (1995) Characterisation of GABAA receptorgamma subunit expression by magnocellular neurones in the rat hypo-thalamus. Mol Brain Res 34:45–56.

Fenelon VS, Sieghart W, Herbison AE (1995) Cellular localisation and

Figure 8. GABA-gated current responses of HEK 293 cells transiently transfected with human a1b1, a1b1e, and a1b1g2s GABAA receptor subunitcDNAs. Recordings were made under whole-cell patch-clamp conditions. The concentration of GABA is indicated above each application. So that peakto steady-state ratios could be determined, applications lasted 10 sec, with at least 30 sec between applications.

5036 J. Neurosci., July 1, 1997, 17(13):5027–5037 Whiting et al. • GABA Receptor e Subunit Cloning and Characterization

Page 11: Neuronally Restricted RNA Splicing Regulates the ... · Neuronally Restricted RNA Splicing Regulates the Expression of a Novel GABA A Receptor Subunit Conferring Atypical Functional

differential distribution of GABAA receptor subunit proteins and mes-senger RNAs within hypothalamic magnocellular neurons. Neuro-science 64:1129–1143.

Gillies G, Davidson K (1992) GABAergic influences on somatostatinsecretion from hypothalamic neurons cultured in defined medium.Neuroendocrinology 55:248–256.

Glencorse TA, Bateson A, Darlison MG (1990) Sequence of the chickenGABAA receptor gamma 2-subunit cDNA. Nucleic Acids Res18:7155–7157.

Hadingham KL, Wingrove P, Le Bourdelles B, Palmer KJ, Ragan CI,Whiting PJ (1993) Cloning of cDNA sequences encoding human alpha2 and alpha 3 gamma-aminobutyric acid-A receptor subunits and char-acterization of the benzodiazepine pharmacology of recombinant alpha1-, alpha 2-, alpha 3-, and alpha 5-containing human gamma-aminobutyric acid-A receptors. Mol Pharmacol 43:970–975.

Hadingham KL, Wafford KA, Palmer K, Whiting PJ (1995) Cloning ofcDNAs encoding the human g-aminobutyric acid type A receptor andcharacterisation of the pharmacology of g3-containing receptors. EurJ Pharmacol Mol Pharmacol Sect 291:301–309.

Hadingham KL, Garrett EM, Wafford KA, Bain C, Heavens RP, Siri-nathsinghji DJ, Whiting PJ (1996) Cloning of cDNAs encoding thehuman g-aminobutyric acid type A receptor a6 subunit and characteri-sation of the pharmacology of a6-containing receptors. Mol Pharmacol49:253–259.

Harvey RJ, Kim HC, Darlison MG (1993) Molecular cloning reveals theexistence of a fourth gamma subunit of the vertebrate brain GABAAreceptor. FEBS Lett 331:211–216.

Horne AL, Macaulay AJ, Harkness PC, Hadingham K, Whiting P, KempJA (1993) On the effect of the g2L subunit on modulation of responsesto GABAA receptor activation. Br J Pharmacol 108:711–716.

Johnston GAR (1996) GABAB receptors: relatively simple transmitter-gated ion channels? Trends Pharmacol Sci 17:319–323.

Kaupmann K, Huggel K, Heid J, Flor PJ, Bischoff S, Mickel SJ, McMasterG, Angst C, Bittiger H, Froesti W, Bettler B (1997) Expression cloningof GABAB receptors uncovers similarity to metabotropic glutamatereceptors. Nature 386:239–246.

Kirkness EF, Kusiak JW, Fleming JT, Menninger J, Gocayne JD, WardDC, Venter JC (1991) Isolation, characterization, and localization ofhuman genomic DNA encoding the b1 subunit of the GABAA receptor(GABRB1). Genomics 10:985–995.

Lambert JJ, Belilli D, Hill-Venning C, Peters JA (1995) Neurosteroidsand GABAA receptor function. Trends Pharmacol Sci 16:295–303.

Laurie DJ, Seeburg PH, Wisden W (1992) The distribution of 13GABAA receptor subunit mRNAs in the rat brain. II. Olfactory bulband cerebellum. J Neurosci 12:1063–1076.

Lewin B (1994) Eukaryotic transcription and RNA processing. In:Genes, Vol V, pp 845–964. Oxford: Oxford UP.

Luddens H, Keinanen K, Monyer H, Sprengel R, Seeburg PH (1990)Cerebellar GABAA receptor selective for a behavioral alcohol antago-nist. Nature 346:648–651.

Macdonald RL, Angelotti TP (1993) Native and recombinant GABAAreceptor channels. Cell Physiol Biochem 3:352–373.

Maiti S, Doskow J, Li-S, Nhim RP, Lindsey JS, Wilkinson MF (1996) ThePem homeobox gene: androgen-dependent and independent promotersand tissue-specific alternative RNA splicing. J Biol Chem271:17536–17546.

McKernan RM, Whiting PJ (1996) Which GABAA receptor subtypesreally occur in the brain? Trends Neurosci 19:139–143.

Mitsushima D, Hei DL, Terasawa E (1994) g-Aminobutyric acid is aninhibitory neurotransmitter restricting the release of luteinizinghormone-releasing hormone before the onset of puberty. Proc NatlAcad Sci USA 91:395–399.

Persohn E, Malherbe P, Richards JG (1991) In situ hybridisation histo-chemistry reveals a diversity of GABAA receptor mRNAs in neurons ofthe rat spinal cord and dorsal root ganglia. Neuroscience 42:497–507.

Pritchett DB, Sontheimer H, Shivers BH, Ymer S, Kettenmann H,

Schofield PH, Seeburg PH (1989) Importance of a novel GABAAreceptor subunit for benzodiazepine pharmacology. Nature338:582–585.

Ramjaun AR, McPherson PS (1996) Tissue-specific alternative splicinggenerates two synaptojanin isoforms with differential membrane-binding properties. J Biol Chem 271:24856–24861.

Sakmann B, Hamill OP, Bormann J (1983) Patch-clamp measurementsof elementary chloride currents activated by the putative inhibitorytransmitters GABA and glycine in mammalian spinal neurons. J NeuralTransm [Suppl] 18:83–95.

Saxena NC, Macdonald RL (1994) Assembly of GABAA receptor sub-units: role of the d subunit. J Neurosci 14:7077–7086.

Saxena NC, Macdonald RL (1996) Properties of putative cerebellarg-aminobutyric acid-A receptor isoforms. Mol Pharmacol 49:567–579.

Schofield PR, Pritchett DB, Sontheimer H, Kettenmann H, Seeburg PH(1989) Sequence and expression of human GABAA receptor a1 and b1subunits. FEBS Lett 244:361–364.

Sieghart W (1995) Structure and pharmacology of g-aminobutyric acid-Areceptor subtypes. Pharmacol Rev 47:181–234.

Sirinathsinghji DJS, Dunnett S (1993) Imaging gene expression in neuralgraft. In: Molecular imaging in neuroscience: a practical approach(Sharif NA, ed), pp 43–70. Oxford: Oxford UP.

Smart TG, Constanti A (1990) Studies on the mechanism of action ofpicrotoxinin and other convulsants at the crustacean muscle GABAreceptor. Br J Pharmacol 99:643–654.

Smart TG, Moss SJ, Xie X, Huganir RL (1991) GABAA receptors aredifferentially sensitive to zinc dependence on subunit composition. Br JPharmacol 103:1837–1839.

Sommer B, Poustka A, Spurr NK, Seeburg PH (1990) The murineGABAA receptor d-subunit gene: structure and assignment to humanchromosome 1. DNA Cell Biol 9:561–568.

Sommer B, Kohler M, Sprengel R, Seeburg PH (1991) RNA editing inbrain controls a determinant of ion flow in glutamate-gated channels.Cell 67:11–19.

Stephenson FA, Duggan MJ, Pollard S (1990) The g2 subunit is anintegral component of the g-aminobutyric acid-A receptor, but the a1polypeptide is the principal site of the agonist benzodiazepinephotoaffinity-labeling reaction. J Biol Chem 265:21160–21165.

Tacke R, Goridis C (1991) Alternative splicing in the neural cell adhe-sion molecule pre mRNA: regulation of exon 18 skipping depends uponthe 59 splice site. Genes Dev 5:1416–1429.

Thompson SA, Whiting PJ, Wafford KA (1996) Alpha subunits influencethe action of pentobarbital on recombinant GABAA receptors. Br JPharmacol 117:521–527.

Thompson SA, Thomas D, Whiting PJ, Wafford KA (1997) Expressionand pharmacology of the human GABAA receptor d subunit. Br JPharmacol 120:283P.

Verdoorn T (1994) Formation of heteromeric g-aminobutyric acid type Areceptors containing two different a subunits. Mol Pharmacol 45:475–480.

von Heijne G (1986) A new method for predicting signal sequence cleav-age sites. Nucleic Acids Res 14:4683–4690.

Whiting P, McKernan RM, Iversen LL (1990) Another mechanism forcreating diversity in g-aminobutyrate type A receptors: RNA splicingdirects expression of two forms of the g2 subunit, one of which containsa protein kinase C phosphorylation site. Proc Natl Acad Sci USA87:9966–9970.

Whiting PJ, McKernan RM, Wafford KA (1995) Structure and pharma-cology of vertebrate GABAA receptor subtypes. Int Rev Neurobiol38:95–138.

Wisden W, Laurie DJ, Monyer HM, Seeburg PH (1992) The distributionof 13 GABAA receptor subunit mRNAs in rat brain. I. Telencephalon,diencephalon, mesencephalon. J Neurosci 12:1040–1062.

Ymer S, Draguhn A, Wisden W, Werner P, Keinanen K, Schofield PR,Sprengel R, Pritchett DB, Seeburg PH (1990) Structural and func-tional characterisation of the g1 subunit of GABA benzodiazepinereceptors. EMBO J 9:3261–3267.

Whiting et al. • GABA Receptor e Subunit Cloning and Characterization J. Neurosci., July 1, 1997, 17(13):5027–5037 5037