Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 279, No. 53, Issue of December 31, pp.

55728 –55736, 2004


© 2004 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

Oligomerization of the ␥-Aminobutyric Acid Transporter-1 Is Driven


by an Interplay of Polar and Hydrophobic Interactions in
Transmembrane Helix II*
Received for publication, August 17, 2004, and in revised form, October 18, 2004
Published, JBC Papers in Press, October 20, 2004, DOI 10.1074/jbc.M409449200

Vladimir M. Korkhov‡, Hesso Farhan, Michael Freissmuth§, and Harald H. Sitte


From the Institute of Pharmacology, Medical University of Vienna, Waehringer Strasse 13a, Vienna A-1090, Austria

The available evidence indicates that members of the GAT2, and GAT3) belong to the neurotransmitter:sodium sym-
neurotransmitter:sodium symporter family form consti- porter (NSS) (1) family, which also includes carriers for dopam-
tutive oligomers. Their second transmembrane helix ine, serotonin, glycine, etc. (2). All members of the family share
(TM2) contains a leucine heptad repeat proposed to be sequence similarity, membrane topology, and common func-
involved in oligomerization. In artificial transmem- tional features (e.g. ion channel-like properties, sodium, and
brane segments, interhelical interactions are stabilized chloride) as transported cosubstrates and bidirectional trans-
by polar residues. We searched for these hydrogen bond port of substrates. The interest in transporters of this family is
donors in TM2 by mutating the five polar residues in motivated in part by their clinical relevance; they are targets
TM2 of the ␥-aminobutyric acid transporter-1 (GAT1). for drugs of abuse (e.g. cocaine, ecstasy, and metamphetamine)
We tested the ability of the resulting mutants to oli-
and for therapeutic agents including antidepressants (amitryp-
gomerize by fluorescence microscopy, Foerster reso-
tiline, desipramine, citalopram, and paroxetine) and anticon-
nance energy transfer, and ␤-lactamase fragment
complementation. Of all generated mutants, only Y86A- vulsants (tiagabine), etc. (reviewed in Ref. 3).
(but not Y86F-), E101A-, E101Q-, and E101D-GAT1 were With the notable exception of the glycine transporter-1 (4),
judged by these criteria to be deficient in oligomeriza- all members of the neurotransmitter transporter family that
tion and were retained intracellularly. The observations have so far been examined form oligomers (reviewed in Ref. 5).
are consistent with a model where the leucine heptad This is also true for GAT1, for which oligomers have been
repeat in TM2 drives a homophilic association that is visualized in membranes of living cells by fluorescence reso-
stabilized by Tyr86 and Glu101; Tyr86 participates in hy- nance energy transfer (FRET) microscopy (6). The TM2 of
drophobic stacking. Glu101 is in the a-position of the GAT1 contains a leucine heptad repeat that seems to supports
leucine heptad repeat (where positions 1–7 are denoted oligomer formation (7). The leucine heptad repeat in TM2 is a
a– g, and each leucine is in the central d-position). Thus, highly conserved feature throughout the NSS family. Consist-
Glu101 is in the position predicted for the hydrogen bond ent with this finding, TM2-mediated dimer formation was also
donor (i.e. sandwiched between Leu97 and Leu104, which suggested for dopamine transporter (8).
are one helical turn above and below Glu101). These key By analogy with leucine zippers in soluble proteins, the
residues, namely Tyr86 and Glu101, are conserved in re- stretch of leucine residues in the TM2 of GAT1 lines one side of
lated transporters from archaeae to humans; they are a hypothetical ␣-helix. If this leucine heptad repeat is to serve
therefore likely to support oligomeric assembly in trans-
as an interface between two transporter molecules, there must
porter orthologs and possibly other proteins with mul-
be additional forces that stabilize this arrangement. In aqueous
tiple transmembrane segments.
solution, the hydrophobic leucine (or isoleucine) side chains are
tightly packed to minimize the solvent exposed surface. It is,
␥-Aminobutyric acid (GABA)1-mediated inhibitory synaptic however, difficult to conceive how the hydrophobic environ-
transmission is terminated by clearance of GABA from the ment in the membrane provides the force to drive the associa-
synaptic cleft by high affinity uptake proteins. The four known tion of leucine residues. In fact, synthetic polyleucine segments
GABA transporters (GAT1, BGT1 (betaine/GABA transporter), show little propensity for self-association in the membrane (9,
10). Nevertheless, in phospholamban and in the erythropoietin
receptor (11, 12), leucine and isoleucine-based packing interac-
* This work was supported by Austrian Science Fund Grants P15034
(to M. F.) and P17076 (to H. H. S.), by Austrian National Bank Grant tions do occur between transmembrane domains. Similarly,
10507 (to H. F.), and by 5th Framework Programme of the European artificial leucine-rich transmembrane segments can be forced
Union Grant QLG3-CT-2001-00929, “Epileptosome.” The costs of pub- to self-associate, provided that the interaction is stabilized by a
lication of this article were defrayed in part by the payment of page polar hydrogen bond-donating residue (i.e. Ser, Thr, Gln, Glu,
charges. This article must therefore be hereby marked “advertisement”
in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. and Asp in the a-position sandwiched between two leucine
‡ Recipient of a Bertha von Suttner-scholarship from the Austrian residues in the d-positions of the heptad repeats) (9, 13). Based
Academic Exchange Service. on these findings, we surmised that a polar residue must be
§ To whom correspondence should be addressed: Institute of Pharma- essential to stabilize the homophilic interaction of TM2 seg-
cology, Vienna Medical University, Waehringer Strasse 13a, A-1090,
Vienna, Austria. Tel.: 43-1-4277-64171; Fax: 43-1-4277-9641; E-mail:
ments in the GAT1 oligomer if the interaction is supported by
michael.freissmuth@meduniwien.ac.at. packing of the leucine heptad repeat. We therefore focused on
1
The abbreviations used are: GABA, ␥-aminobutyric acid; GAT1, -2, the polar or ionizable residues in TM2 of GAT1: Tyr86, Thr89,
and -3, GABA transporter-1, -2, and -3, respectively; NSS, neurotrans- Glu101, Cys102, and Ser103. Our experiments identified the two
mitter:sodium symporter; TM2, transmembrane domain II; FRET, flu-
orescence resonance energy transfer; DRAP, donor recovery after ac-
residues Tyr86 and Glu101, both invariably present in the cor-
ceptor photobleaching; CFP, cyan fluorescent protein; YFP, yellow responding positions in all neurotransmitter:sodium symport-
fluorescent protein; ANOVA, analysis of variance. ers, as key players in TM2-mediated intramembrane interac-

55728 This paper is available on line at http://www.jbc.org


TM2 Controls GABA Transporter-1 Oligomerization 55729
TABLE I Fluorescence Microscopy—Fluorescence microscopy was performed
Primers employed for the generation of mutated versions of GAT1 using a Zeiss Axiovert 200M inverted epifluorescence microscope
Primer Sequence
equipped with a CoolSNAP fx cooled CCD camera (Photometrics, Roper
Scientific, Tucson, AZ). The fluorescence filter sets were purchased from
Y86A-fw 5⬘-cttcctaattccagctttcctg-3⬘ Chroma (Chroma Technology Corp., Brattleboro, VT; CFP filter set:
Y86A-rv 5⬘-gagcgtcaggaaagctggaat-3⬘ excitation 436 nm, dichroic 455 nm, emission 480 nm; YFP filter set:
Y86F-fw 5⬘-cttcctaattccattcttcctg-3⬘ excitation 500 nm, dichroic 515 nm, emission 535 nm; FRET filter
Y86F-rv 5⬘-gagcgtcaggaagaatggaat-3⬘ set: excitation 436 nm, dichroic 455 nm, emission 535 nm). Coverslips
T89A-fw 5⬘-catatttcctggcgctcatc-3⬘ with attached cells were mounted in the microscope chamber and put on
T89A-rv 5⬘-gcaaagatgagcgccaggaa-3⬘ the microscope stage. Images of cells with CFP- or YFP-tagged trans-
T89F-fw 5⬘-tccatatttcctgttcctcatc-3⬘ porters were acquired through corresponding filter channels. For donor
T89F-rv 5⬘-ccgcaaagatgaggaacaggaa-3⬘ photobleaching, FRET cells expressing either CFP-tagged or CFP- and
E101A-fw 5⬘-tctcttccttttggcgtgct-3⬘
YFP-tagged transporters were continuously illuminated with a 100-
E101A-rv 5⬘-ctagggagcacgccaaaagg-3⬘
watt mercury lamp and the CFP filter set for 1 min, a time interval
E101D-fw 5⬘-tctcttccttttggactgctc-3⬘
E101D-rv 5⬘-ctagggagcagtccaaaagg-3⬘ sufficient to bleach the donor to an extent of less than 20% (with
E101Q-fw 5⬘-tctcttccttttgcagtgctc-3⬘ acquisition of one image every 3 s). Regions of interest were selected,
E101Q-RV 5⬘-ctagggagcactgcaaaagg-3⬘ and fluorescence emission intensities were quantified. The resulting
C102A-fw 5⬘-ccttttggaggcctccctagg-3⬘ fluorescence decay curves were fitted to the equation for a single expo-
C102A-rv 5⬘-ctggcctagggaggcctcca-3⬘ nential decay approaching a constant value: fluorescence intensity ⫽
S103A-fw 5⬘-cttttggagtgcgccctag-3⬘ A0e⫺Kt ⫹ offset, where A0 denotes the starting value, offset is the final
S103A-rv 5⬘-actggcctagggcgcactc-3⬘ fluorescence signal, and K is the decay constant. The photobleaching
lifetime constant ␶ is defined as 1/K. To measure donor recovery after
acceptor photobleaching (DRAP), we acquired a donor (CFP) image
tion. However, in Tyr86 the hydrogen bond-donating hydroxyl
before (Ib) and after (Ia) photobleaching using the YFP setting for 90 s
group is dispensable, and it is the aromatic ring (and hence a (excitation 500 nm, dichroic mirror 525 nm, and emission 535 nm).
hydrophobic interaction) that is (appears) essential for stabi- DRAP was quantified by FRET efficiency (E) according to the following
lizing the oligomer. In contrast, both, the size of the side chain equation: E ⫽ (Ia ⫺ Ib)/Ia (16) and corrected for cross-bleaching of CFP
and the hydroxyl group of Glu101 seem to be essential for through the YFP filter set (⬃9 –10%). Fluorescence images were ac-
supporting oligomer formation. Glu101 is in the a-position of the quired and analyzed using the MetaMorph and MetaFluor of MetaSer-
ies software package (release 4.6; Universal Imaging Corp., Downing-
heptad repeat, sandwiched between Leu97 and Leu104, which,
town, PA). For statistical comparisons of time constants of fluorescence
in a helical arrangement, are one turn above and below Glu101. efficiencies, ANOVA followed by Dunnett’s or Tukey’s test was used.
Thus, Glu101 fulfills the criteria of the hydrogen bond donor ␤-Lactamase Protein Fragment Complementation Assay—For the
capable of stabilizing the interhelical contact. ␤-lactamase protein fragment complementation assay, we used plas-
mids pcDNA3.1-Zeo/F[1]m182T and pcDNA3.1-Zeo/F[2] to express of
EXPERIMENTAL PROCEDURES
proteins fused to ␤-lactamase fragments F[1] and F[2] (17). Constructs
cDNA Constructs and Mutagenesis—Plasmid DNA constructs YFP- of wild type and mutant GAT1 were fused by PCR cloning. Cells were
GAT1, CFP-GAT1, YFP-GAT1, YFP-GAT⌬37, CFP-GAT⌬37, and CFP- transfected with the indicated combinations of plasmids using the cal-
GAT1-L97A were described elsewhere (7, 14). Mutated versions of cium phosphate precipitation method. After 24 h, cells were detached
GAT1, carrying single amino acid substitutions (Y86A, Y86F, T89A, mechanically, lysed by a freeze-thaw cycle, and homogenized in phos-
T89F, G94A, E101A, E101D, E101Q, C102A, and S103A) were created phate-buffered saline. Membranes were sedimented by centrifugation
by PCR. For amplification of DNA fragments encompassing nucleotides at 50,000 ⫻ g for 20 min, and pellets were resuspended in phosphate-
1–1032 of mutant GAT1 DNA sequences, we used the pairs of primers buffered saline. The membrane suspension (20-␮l aliquot containing
listed in Table I. 50 –100 ␮g of membrane protein) was incubated in the presence of 100
The HindIII/AccI fragments excised from PCR products were cloned ␮M nitrocefin in phosphate-buffered saline in a total volume of 200 ␮l;
into a HindIII/AccI-digested CFP-GAT1-L97A construct. At this step, the incubation times were as indicated for the indicated periods at
the L97A mutation in the former was eliminated, and the desired ones 37 °C. Absorption at 492 nm was measured using an automated plate
were introduced. The derived transporters or truncated versions (amino reader. The comparisons were performed between the activities meas-
acids 1–110) were cloned in frame with F[1] and F[2] ␤-lactamase ured within linear range, before onset of substrate (nitrocefin) deple-
fragment via AscI/NotI sites introduced by PCR. Sequences of all con- tion. The results were analyzed by one-way ANOVA followed by Dun-
structs were verified. nett’s test.
Cell Culture, Heterologous Expression, and Uptake Assays—Human Molecular Graphics—The Swiss-PDBViewer was used to generate
embryonic kidney 293 (HEK-293) cells were grown in Dulbecco’s mod- the molecular model of TM2 dimer. The sequence of GAT1 TM2
ified Eagle’s medium containing L-alanyl-L-glutamine, 10% fetal bovine (AFLIPYFLTLIFAGVPLFLLECSL) was fitted onto the ␣-carbon back-
serum, and 50 mg/liter gentamicin on 10-cm diameter cell culture bone of the two GCN-4 (2ZTA) leucine zipper helices. The improbable
dishes at 37 °C in an atmosphere of 5% CO2, 95% air. One day before side chain conformations were excluded by selection of allowed conform-
transfection, cells were replated to obtain subconfluent cultures either ers following energy minimization of the whole structure by Swiss-
on glass coverslips (22 mm in diameter and placed into 6-well plates, PDBViewer implementation of GROMOS96. The derived model was
3 ⫻ 105 cells/well plate) or on 10-cm plates (1 ⫻ 106 cells/well plate) for rendered by POV-Ray 3.0.
uptake experiments. Transient transfections were done using the
CaPO4 precipitation method (15) and whole-cell uptake assays as de- RESULTS
scribed previously (6). In brief, 48 h after transfection with either
CFP-GAT1 or its mutated versions, HEK-293 cells were incubated for 3 Site-directed Mutagenesis—The hydrophobic environment of
min at room temperature in 0.1 ml of Krebs-Ringer-Hepes buffer (10 the membrane does not provide any driving force for the as-
mM Hepes, 120 mM NaCl, 3 mM KCl, 2 mM CaCl2, 2 mM MgCl2, 20 mM sembly or stabilization of a leucine zipper (9, 10). Thus, the
glucose, final pH 7.3) containing 15–20 nM [3H]GABA in the presence of amino acid residues of TM2 were examined for their ability to
increasing concentrations of unlabeled GABA, with total GABA concen-
form hydrogen bonds and selected by sequence comparison
tration ranging from 1 to 60 ␮M (resulting in a specific activity ranging
from 1000 to 10 cpm/pmol). Nonspecific uptake was determined in the between the members of the NSS family. For lack of structural
presence of 10 ␮M tiagabine, with 3 min of preincubation. The uptake information, we considered the TM2 region as the sequence
buffer was aspirated, and the cells were washed with 1 ml of ice-cold between Ala81 and Leu104 (Fig. 1A); the leucine heptad repeat
Krebs-Ringer-Hepes buffer and lysed with 0.5 ml of 1% SDS. The comprises residues Leu83–Leu104. We identified residues tyro-
amount of accumulated radioactivity was determined by liquid scintil- sine 86 (Tyr86), threonine 89 (Thr89), glutamate 101 (Glu101),
lation counting. For vesicular uptake assays, membranes were pre-
cysteine 102 (Cys102), and serine 103 (Ser103) as the candidates
pared from cells that expressed intracellularly retained GAT1 mutants
in isotonic sucrose; NaCl-driven uptake of [3H]GABA into these mem- for site-directed mutagenesis (marked by the arrows in Fig.
brane vesicles (50 –100 ␮g/assay) was measured by employing a filtra- 1A). Side chains of each of these residues may participate in
tion assay as described earlier (7). inter- or intrahelical interactions between transmembrane do-
55730 TM2 Controls GABA Transporter-1 Oligomerization

FIG. 1. Sequence conservation of TM2 within NSS family, [3H]GABA uptake, and fluorescence microscopy of the corresponding
mutated GAT1 proteins in living cells. A, amino acid residues 81–104 conform to the putative TM2 of GAT1. The homologues from archaeal,
bacterial, and mammalian sources share high sequence similarity with rat GAT1 TM2. The arrows indicate the residues mutated herein. B, cells
(1–2 ⫻ 105/well) expressing wild type GAT1 and the indicated GAT1 mutants were incubated with 5 ␮M [3H]GABA (specific activity ⬃160
cpm/pmol) for 3 min, and uptake was determined as outlined under “Experimental Procedures.” Data are means from two independent experiments
that were carried out in triplicate and done in parallel on cells expressing the wild type transporter and each indicated mutant. The uptake of the
wild type was 50 – 80 pmol䡠min⫺1䡠10⫺6 cells and was set to 100%. C, HEK-293 cells were transfected with equal amounts of vectors encoding for
GAT1 (WT) or its mutated versions (substitution indicated in each image). The next day, images (⫻ 63, oil immersion) were taken using the
acceptor (WT) or donor filter set. The images are representative of 4 – 6 independent transfections.

mains of polytopic proteins. Tyr86 and Glu101 are conserved in centration of [3H]GABA, the transport velocities of the mutants
all members of the family including bacterial and archaeal Y86F, C102A, and S103A were comparable with that of the
transporters (three examples shown in Fig. 1A). This suggests fluorescently tagged wild type transporter (Fig. 1B). Accord-
similar or related functional roles in different transporters. ingly, in saturation experiments, these mutants had Km values
Ser103 is present in close relatives of GAT1, namely glycine and that were indistinguishable from those of wild type GAT1 (Ta-
proline transporters. The equivalent positions in TM2 of more ble II). In contrast, substitutions of Thr89 by alanine and by
distantly related transporters are predominantly occupied by phenylalanine progressively reduced the affinity for substrate
alanine. Thr89 and Cys102 are present exclusively in GAT1. (see Fig. 2A and Table II). The Km reflects the overall process of
Thus, we searched for the hydrogen donor that may support substrate transport (i.e. binding of substrate and the cosub-
oligomer formation by generating the following point-mutated strates Na⫹ and Cl⫺, their translocation, and intracellular
versions of GAT1 (see also Fig. 1A); the Tyr86 residue was release). Because there is no suitable radioligand for GAT1
mutated to alanine (the smallest side chain, incapable of form- binding assays, it is not possible to determine substrate bind-
ing hydrogen bonds) and phenylalanine (lacking only the hy- ing affinity directly. We probed the binding pocket of GAT1
droxyl group when compared with tyrosine). Thr89 was substi- indirectly by determining the IC50 of tiagabine for GAT1 at a
tuted by alanine and phenylalanine (present in analogous very low substrate concentration. Under these conditions, dif-
positions of dopamine transporter, norepinephrin transporter, ferences in substrate affinity are irrelevant, and the IC50 ap-
GAT2, GAT3, etc.). Glu101 was changed to alanine and aspar- proaches the Ki value of the inhibitor. As can be seen from Fig.
tate (a charged residue one -CH2- group shorter than gluta- 2B, mutation of Thr89 did not impair binding of the inhibitor
mate) and glutamine (devoid of charge but same size as gluta- regardless of whether threonine was substituted by alanine or
mate). Cys102 and Ser103 were substituted by alanine residues. by the bulky phenylalanine (see also Table II). These findings
Transport of GABA by GAT1 Mutants—Cellular uptake as- argue against a role of Thr89 in initial inhibitor recognition and
says showed that the mutated transporters were capable of binding but imply a role in translocation of the substrate.
transporting GABA with a high rate and that transport was Y86A-GAT1 transported substrate with a significantly lower
blocked by the competitive inhibitor tiagabine. By definition, rate when compared with wild type GAT1 (second bar from left
cellular uptake demonstrates that the transporters are ex- in Fig. 1B). In experiments performed with E101A, E101D, and
pressed on the cell surface. When assayed at a low fixed con- E101Q-GAT1, the transport of [3H]GABA could barely be de-
TM2 Controls GABA Transporter-1 Oligomerization 55731
TABLE II Endoplasmic Reticulum Export of the Wild Type Transporter
Km values for uptake of 关3H兴GABA uptake by wild type (WT) and in the Presence of GAT1 Mutants—Previous work by us and
mutated versions of GAT1 and IC50 values for tiagabine-induced
inhibition thereof others has led to the hypothesis that the export of the NSS
members GAT1 (7, 14), serotonin transporter (18), and dopa-
Km Km IC50 for
TM2 mutation (whole cell)a (vesicular uptake)b mine transporter (8, 19) is contingent on oligomerization. This
tiagabine
model also explains the dominant negative phenotype (i.e. fa-
␮M ␮M nM
milial postural hypotension) that results from a point mutation
WT GAT1 4.2 ⫾ 1.0 4.1 ⫾ 0.6c 408 ⫾ 26
Y86A 9.2 ⫾ 3.0 in one allele of the norepinephrine transporter (20). A compar-
Y86F 2.0 ⫾ 0.4 ison of Fig. 4, A and B, illustrates this phenomenon; cells were
T89A 17.6 ⫾ 2.3 233 ⫾ 63 transfected with CFP-tagged GAT1-⌬37 (visualized in Fig. 4A)
T89F 57.8 ⫾ 15.0 244 ⫾ 91 and YFP-tagged wild type GAT1 (visualized in Fig. 4B). In the
E101A NMd
E101D 1.6 ⫾ 0.5 cell that expressed GAT1-⌬37 (visualized in the left corner of
E101Q NM Fig. 4A), the wild type transporter was trapped in the endo-
C102A 3.0 ⫾ 0.5 plasmic reticulum (Fig. 4B). In contrast, in the cell that only
S103A 4.7 ⫾ 0.9
expressed YFP-tagged GAT1, the transporter was found at the
a
Km values were calculated from whole cell uptake experiments plasma membrane (top right corner of Fig. 4B). As mentioned
(performed as shown in Fig. 1A); the differences between wild type earlier, GAT1-⌬37 is trapped in the endoplasmic reticulum,
GAT1, T89A-GAT1, and T89F are statistically significant judged by
one-way ANOVA followed by Tukey’s post hoc test (n ⫽ 4 – 6). because it lacks the docking site for the coatomer protein II
b
Km values were calculated from uptake assays in vesicular mem- component Sec24 (14). Thus, this approach can serve as a
brane preparations (n ⫽ 4). means to detect the capacity of mutant transporters to associ-
c
Km was determined for intracellularly retained GAT1-⌬37. ate with wild type GAT1. It is evident that all Glu101-substi-
d
NM, not measurable.
tuted GAT1 variants (Fig. 4, E, G, and I) did not impair cell
surface targeting of the wild type transporter to any apprecia-
tected irrespective of the transfection method employed (cal- ble extent (Fig. 4, F, H, and J). In cells that expressed high
cium phosphate precipitation in Fig. 1B). This observation levels of Y86A-GAT1 (cell on the left in Fig. 4C), there was some
indicated that the mutated transporters were either not prop- intracellular retention of wild type GAT1 (Fig. 4D); wild type
erly folded or failed to get inserted into the plasma membrane. GAT1 was, however, visualized predominantly at the cell sur-
We verified that all mutants were expressed by visualizing the face in cells that expressed Y86A-GAT1 at moderate levels
transporters in living cells by means of fluorescence micros- (exemplified by the cell on the right in Fig. 4, C and D). We
copy. Consistent with [3H]GABA uptake measurements, CFP- interpret these observations as indicative of a reduced associ-
tagged GAT containing Y86F, T89A, T89F, C102A, and S103A ation of the Y86A mutant and the Glu101-substituted versions
substitutions were expressed at the surface of heterologously with the wild type transporter.
transfected HEK-293 cells (Fig. 1C). By contrast with wild type Impaired Oligomerization of Intracellular GAT1 Mutants—
CFP-GAT1 (and the other mutants), there was little, if any, The fact that the Y86A and Glu101 mutants were retained within
fluorescence recorded over the plasma membrane with Y86A- the cell and failed to retain the wild type transporter in the
GAT1, E101A-GAT1, E101D-GAT1, and E101Q-GAT1; these endoplasmic reticulum provided circumstantial evidence for a
mutants were trapped within the cell. defect in their ability to form oligomers. To directly visualize the
Transport of GABA by Intracellularly Retained GAT1 Mu- oligomeric status of individual transporters in living cells, we
tants—The mutations Y86A, E101A, and E101D caused ineffi- performed FRET microscopy. FRET microscopy relies on the
cient targeting of GAT1 to the plasma membrane of HEK-293 energy transfer that occurs if donor and acceptor fluorophore are
cells. This may have resulted from misfolding of these mutated in close vicinity; in the pair CFP (donor) and YFP (acceptor), this
transporters. To assess the conformational integrity of the mu- Foerster distance is in the range of 50 Å (Fig. 5A) (21) and thus
tants, we prepared membrane vesicles from appropriately requires the formation of an oligomer. Here we used approaches
transfected cells and measured vesicular [3H]GABA uptake. As that rely either on the protection of the donor against bleaching
an internal control, we used GAT1-⌬37, which lacks the last 37 in the presence of acceptor (donor photobleaching FRET) or on
amino acids. This truncated version is retained within the cell, the increase in donor emission after acceptor bleaching (donor
because it lacks the docking sites for the coatomer protein II recovery after acceptor photobleaching). Donor photobleaching
components, but it has a wild type transmembrane core and FRET did not reveal any difference in the lifetime (␶) of the
hence transports at normal rates (14). The transporters were fluorescent decay upon bleaching the mutant transporters ex-
tagged with a fluorescent protein; this allowed us to correct for pressed in the plasma membrane (Fig. 5, B and C). Thus, the
the variability in expression inherent in transient transfections mutations of residues Thr89, Cys102, and Ser103 as well as sub-
by normalizing the transport velocity for recorded fluorescence. stitution of Tyr86 to phenylalanine did not impede the interaction
It is evident from Fig. 3A that Y86A-GAT1 transported with wild type GAT1. In other words, the hydrophilic side chains
[3H]GABA with a Km and a Vmax value that were reasonably of these residues are unlikely to participate in forming the oligo-
similar to that of the wild type transporter. Likewise, E101D- meric interface.
GAT1 translocated [3H]GABA with an apparent affinity that We have also verified this conclusion by using DRAP-FRET
was within the range seen with GAT1-⌬37 (see Table II). In microsocopy, an independent approach that relies on bleaching
contrast, we failed to detect any measurable [3H]GABA influx of the acceptor rather than the donor. The data set depicted in
with vesicles containing the other two Glu101 mutants (i.e. Fig. 5D shows a representative experiment for GAT1-⌬37 (top
E101A-GAT1 and E101Q-GAT1). Thus, while we cannot rule row) and for GAT1–Y86A (bottom row), where the fluorescence
out the possibility that the latter two mutants are misfolded, it intensity is shown in pseudocolors. We used GAT1-⌬37 to
is clear that Y86A-GAT1 and E101D-GAT1 adopt a conforma- gauge the oligomeric status of those mutants that are retained
tion that binds and translocates substrate with an affinity that within the cell; because GAT1-⌬37 is trapped in the endoplas-
is only modestly different from that of the wild type trans- mic reticulum, it serves as a good control and obviates a com-
porter. In other words, retention of Y86A-GAT1 and of E101D- parison over different membrane compartments. The increase
GAT1 is unlikely to be due to misfolding. in fluorescence that can be seen for GAT1-⌬37 (cf. CFP images
55732 TM2 Controls GABA Transporter-1 Oligomerization

FIG. 2. Uptake of [3H]GABA by cells expressing wild type and mutant GAT1. A, cells (1–2 ⫻ 105/well) expressing wild type GAT1 (f),
T89A (Œ), or T89F () were incubated with the indicated total concentrations of GABA for 3 min, and uptake was determined as outlined under
“Experimental Procedures.” B, cells (as in A) were incubated with 1 ␮M [3H]GABA (specific activity 800 cpm/pmol) and increasing concentrations
of tiagabine; data represent means ⫾ S.E. in at least three independent experiments performed in duplicate. Transport velocity in the absence of
inhibitor amounted to ⬃20 pmol/min/106 cells (⬃8000 –10,000 cpm) and was set to 1.0 to normalize for interassay variation. The solid lines were
drawn by subjecting the data to a nonlinear, least-squares curve fit using the equation for a rectangular hyperbola.

FIG. 3. Uptake of [3H]GABA in vesicles incorporating retained GAT1 mutants. A, the vesicles (20 –50 ␮g of protein/assay) were prepared
from cells expressing CFP-GAT1 (f) and Y86A-GAT1 (Œ) and were incubated with the indicated concentrations of [3H]GABA at 20 °C for 3 min
as described under “Experimental Procedures.” The solid lines were drawn by subjecting the data to a nonlinear, least-squares curve fit using the
equation for a rectangular hyperbola. B, membrane vesicles were prepared from HEK-293 cells, expressing wild type CFP-GAT1, Y86A-GAT1,
E101A-GAT1, and E101D-GAT1. The vesicle preparations (40 ␮g of protein/assay) were incubated with a single concentration of [3H]GABA (⬃500
nM) in a final volume of 50 ␮l. Nonspecific uptake/binding determined in the presence of 100 ␮M tiagabine was subtracted.

FIG. 4. Intracellular retention of in-


tact GAT1 by intracellular mutants.
Images were captured from HEK-293
cells co-expressing YFP-GAT1 and the in-
dicated CFP-tagged mutants by fluores-
cence microscopy as described in the leg-
end to Fig. 1C. Images were captured
through the CFP filter set for cells co-
expressing CFP-tagged GAT1-⌬37, Y86A,
E101A, E101D, and E101Q mutants, re-
spectively (A, C, E, G, and I), and then
through the YFP filter set to visualize the
YFP-tagged wild type GAT1 (B, D, F, H,
and J).

before and after bleaching in the top row) is not readily detect- results indicate that the phenyl ring of Tyr86 and the side chain
able for the Y86A mutant (bottom row). Fig. 5E summarizes the of Glu101 are crucial for formation of the GAT1 oligomer. Two
results from DRAP-FRET microscopy (Fig. 5, D and E), which different FRET-based methods that monitor protein-protein
allows for quantitative comparisons of energy transfer efficien- interaction lead to the same conclusions. Furthermore, FRET
cies; it is evident that the FRET efficiency is comparable for microscopy provides evidence for a homophilic TM2-driven oli-
wild type GAT1 and Y86F (white columns) and that they are gomerization. The mutated transporters fused to CFP and YFP
similar in magnitude to the C-terminally truncated GAT1-⌬37. comprise the FRET pairs with FRET efficiencies roughly equal
In contrast, the FRET efficiency is reduced by about 50% for to those of pairs with the acceptor molecule being GAT1-⌬37.
the mutants Y86A-GAT1 and E101A/D/Q-GAT1 regardless of Mutation of a single residue (Tyr86 or Glu101) in a TM2 of one
whether they are allowed to interact with GAT1-⌬37 or with of the interacting molecules was sufficient to impair this inter-
themselves (Fig. 5E). Consistent with these DRAP measure- action. Mutation of the respective residue in both interacting
ments, donor photobleaching experiments showed reduced pho- subunits did not increase the difference in FRET efficiency,
tobleaching lifetimes with the representative pairs of the in- compared with the GAT1-⌬37 control. This suggests that the
tracellularly retained mutants: Y86A versus GAT1 and E101D mutated residues form contacts in a mutually dependent fash-
versus GAT1 (black columns in Fig. 5C). Taken together, these ion, constituting a homophilic (TM2-TM2) interaction domain.
TM2 Controls GABA Transporter-1 Oligomerization 55733

FIG. 5. Donor photobleaching and DRAP FRET microscopy of intracellular GAT1 mutants. The indicated pairs of CFP- or YFP-labeled
GAT1 mutant constructs were coexpressed in HEK-293 cells at a 1:1 ratio of plasmids encoding donor and acceptor. Donor photobleaching (B, C)
and DRAP (D, E) FRET microscopy experiments were performed 24 –28 h after transfection. A, scheme illustrating the principle of FRET. B,
photobleaching of the donor (CFP) was achieved by illuminating the cell at the center of the visual field with a mercury arc lamp (Zeiss HBO;
100-watt intensity) for 60 s. Images were captured every 3 s, digitized, and stored. Decay of intensity (in the regions of interest) was quantified
in successive images to generate the decay curves and calculate photobleaching lifetimes, ␶. C, a summary of ␶-values determined as shown in B
for the indicated mutants. The ␶ values for all mutants that were expressed at the cell surface (white bars) were not significantly different from
those of the wild type GAT1; for mutants that were retained within the cell (represented by black bars), there was a significant difference between
the ability of self-association of the control protein GAT1-⌬37 and its association with the Y86A and the E101 mutant (*, p ⬍ 0.05; **, p ⬍ 0.01;
significance judged by one-way ANOVA followed by Dunnett’s test; values represent mean ⫾ S.E. from 15–22 individual curves). D, to measure
DRAP, a donor (CFP) image was acquired before (left image) and after photobleaching (middle image) using the YFP setting for 90 s (excitation
500 nm, dichroic mirror 525 nm, and emission 535 nm); shown are pseudocolor images for the control protein (GAT1-⌬37) and a representative,
intracellularly retained mutant, Y86A-GAT1; the YFP image prior to bleaching is also shown to document the presence of acceptor within the same
cell. E, summary of FRET efficiencies obtained from DRAP experiments carried out as in C. The FRET efficiency (mean ⫾ S.E.; n ⫽ 11–29,
calculated as described under “Experimental Procedures”) determined for GAT1-⌬37 versus GAT1-⌬37 was significantly different from that
determined for all other intracellularly retained mutants (represented by black bars; p ⬍ 0.05; one-way ANOVA followed by Dunnett’s test) but
comparable with the ransporters expressed at the cell surface (indicated by white bars) (i.e. self-association of wild type GAT1 (abbreviated as CrG
versus YrG) and Y86F versus wild type GAT1 (abbreviated as Y86F versus YrG).

A decrease in FRET efficiencies may arise from an increase branes were prepared from HEK-293 cells that co-expressed
in the distance between the fluorophores, a change in the these constructs, and the reconstitution of enzymatic activity
relative orientation of the fluorophore dipoles, or a decline in was assessed colorimetrically. As predicted, the rates of nitro-
the steady state number of oligomeric complexes. The data in cefin cleavage in membranes containing GAT1-F[1] and GAT1-
Fig. 4 also suggest that the mutations reduced the level of F[2] were comparable with those of membranes from cells
oligomeric complexes. We obtained independent evidence for expressing GAT1-F[1] and Y86F-GAT1-F[2] (squares and
this interpretation by employing a third approach, namely a downward pointing triangles in Fig. 6B). However, Y86A-
␤-lactamase fragment complementation assay (17). This GAT1-F[2] and the corresponding fusion protein containing the
method of proving interaction between two proteins relies on mutations E101A, E101D, and E101Q reconstituted ␤-lacta-
reconstitution of enzyme activity (cleavage of nitrocefin) upon mase activity with GAT1-F[1] to a lower extent (Fig. 6, B and
interaction of two proteins that are tagged by two split parts of C). This finding is consistent with the results of FRET micros-
the enzyme ␤-lactamase (Fig. 6A). We fused GAT1 and its copy (Fig. 5) and of the retention assay (Fig. 4); taken together,
mutated versions in frame with two complementary fragments the observations show that the mutations Y86A and E101A/
(fragment F[1] and fragment F[2]) of TEM ␤-lactamase. Mem- D/Q impair the ability of the transporter to form an oligomer.
55734 TM2 Controls GABA Transporter-1 Oligomerization
␤-lactamase complementation (Fig. 6B). These observations
suggest that there are additional contact sites in the oligomeric
interface (e.g. TM4 (22) and TM11/12 (18)). To study the ho-
mophilic interaction between TM2 segments, we generated
constructs that comprised TM1, TM2, and either the ␤-lacta-
mase fragment F[1] or F[2] (schematic rendering in Fig. 6E).
TM1, which functions as a (noncleavable) signal sequence, was
included to ascertain the correct orientation of the polypeptide.
These short GAT1-derived constructs carrying mutations
Y86A/F and a representative of the glutamate 101 (E101D)
were co-expressed as F[1]- and as F[2]-tagged versions. The
activities of these pairs of fusion constructs were compared
with those of the wild type pair: Y86F was again indistinguish-
able from wild type, whereas Y86A and E101D had lower
activities (Fig. 6D). A comparison of Fig. 6, C and D, shows that
the differences in association of Y86A and E101D mutants was
more pronounced when presented in the context of the short
TM1-TM2 construct.
DISCUSSION
For many membrane proteins, a correct quaternary assem-
bly is a prerequisite for efficient exit from the endoplasmic
reticulum and subsequent delivery to the plasma membrane.
Prominent examples are GABAB receptors and ATP-sensitive
K⫹ channels (23, 24). The mechanisms that drive the oligo-
meric assembly of proteins with multiple transmembrane seg-
ments are, in many instances, poorly understood. Neurotrans-
mitter transporters are of particular interest, because they
contain multiple motifs known to support the association of
transmembrane segments: a glycophorin-like motif in TM6
(19), a leucine heptad repeat in TM2 (7), and additional inter-
action motifs in TM4 (22) and TM11–12 (18).
In the present work, we focused on TM2 of GAT1 and
searched specifically for the polar residue that may be required
to drive the association of leucine heptad repeats in the mem-
brane (9). Our observations exclude Tyr86, Thr89, Cys102, and
Ser103 as hydrogen bond donors required for stabilization of the
oligomer. In contrast, Glu101 meets several essential criteria. (i)
Glu101 occupies the a-position in the leucine heptad repeat (i.e.
it is sandwiched between Leu97 and Leu104, which are one
helical turn above and below Glu101 and thus occupy the d-
position in their respective heptad repeat (where positions 1–7
are denoted a– g) (9); (ii) replacement of Glu101 by mutation
greatly reduced the ability of the resulting mutants to support
oligomerization: cell surface delivery, resonance energy trans-
FIG. 6. ␤-Lactamase fragment complementation. A, the scheme fer, and ␤-lactamase complementation; and (iii) the loss of
depicts the principle of the ␤-lactamase protein complementation assay; oligomerization cannot be attributed to a major misfolding.
the two fragments of ␤-lactamase (labeled F[1] and F[2]) refold upon Although E101A and E101Q failed to translocate substrate, the
interaction of transporter molecules and cleave substrate-nitrocefin (tri- activity of E101D-GAT1 was readily measurable and its affinity
angle labeled with N). B, representative experiment of ␤-lactamase
complementation by full-length wild type GAT1-F[1] co-expressed with for substrate was within the range observed with the wild type
full-length wild type or mutant GAT1-F[2]; HEK-293 cells were co- transporter. This unequivocally shows that E101D-GAT1 adopts
transfected with plasmids encoding the indicated combinations of con- an active conformation. We therefore rule out the possibility that
structs fused to either fragment of ␤-lactamase (F[1] or F[2]). The enzy- the E101D mutant is retained in the endoplasmic reticulum due
matic activity was measured in duplicate as described under
“Experimental Procedures.” The inset shows the initial 6 min that were
to misfolding at the level of tertiary structure. Loss of function in
used to calculate the rate of enzymatic activity from the linear portion. C, E101A and E101Q mutants may not be directly addressed. How-
summary of three independent experiments, performed in duplicate as in ever, in norepinephrin transporter (25), these mutations result in
B. The activity determined with wild type GAT1-containing constructs a transporter with at least partial activity. Taken together, these
was the reference (equal to 100%). D, the ␤-lactamase activities of coex-
observations suggest that a major folding problem is unlikely to
pressed TM1-TM2-F[1]/TM1-TM2-F[2] pairs were determined in a man-
ner analogous to that described for B. Data are means ⫾ S.E. from three be present in GAT1-E101D.
independent experiments carried out in duplicate. The asterisks (p ⬍ 0.05) Substitution of Tyr86 also affected oligomerization of GAT1.
and double asterisks (p ⬍ 0.01) indicate statistical significance of differ- The bulk of the Y86A mutant was trapped in the intracellular
ence from control (one-way ANOVA followed by Dunnett’s test). compartment and showed reduced interaction with itself or the
wild type transporter. Because of its hydroxyl group, tyrosine is
␤-Lactamase Fragment Complementation by Isolated Trans- a polar residue; nevertheless, it is the hydrophobic moment of
membrane Domains—It is evident that the point mutations in Tyr86 that matters for TM2-mediated transporter association.
TM2 (at positions Tyr86 and of Glu101) neither fully eliminated Rather than donating a hydroxyl for a hydrogen bond, Tyr86 is
resonance energy transfer (Fig. 5E) nor completely abolished likely to engage in a hydrophobic interaction via the aromatic
TM2 Controls GABA Transporter-1 Oligomerization 55735
The side chain of Glu101 is a likely hydrogen bonding entity
that may provide the driving force for TM2 dimerization in the
hydrophobic environment of the membrane. In accordance with
our hypothesis, the protonated E101 may donate its hydrogen
for a hydrogen bond with a heteroatom of the Glu101 of a
juxtaposed helix. The acidic residues in TM spans have been
shown to cause strong hydrogen bond-based association within
the lipid bilayer of ErbB2 receptor and cystic fibrosis trans-
membrane conductance regulator Cl⫺ channel (30, 31). Glu101
is flanked along its helical side by two leucine residues, Leu97
and Leu104, which may protect Glu101 from the aqueous cyto-
solic milieu (Fig. 7B). Mutation of Leu97 to alanine destabilizes
the dimer, whereas mutation of both Leu97 and Leu104 com-
pletely abolishes TM2-TM2 dimerization (7). This may be ex-
plained by a loss of hydrophobic contacts between leucine res-
idues, which allows for access of water and thus results in
deprotonation of the ␦-carboxyl group of Glu101 (Fig. 7C). Our
interpretation is that the hydrogen bonding of Glu101 occurs in
the context of hydrophobic side chains of its neighbors and is
strictly directional; mutations to Asp or Gln are detrimental to
hydrogen bond formation. The E101Q mutant may not form the
equivalent hydrogen bond. Likewise, the transporter with
FIG. 7. Molecular modeling of the dimeric interface formed by E101D substitution is unable to form the hydrogen bond and
the TM2 of GAT1. A, proposed model of the TM2 dimer of GAT1, as
viewed in the plane of the membrane. The stretch Ala81 to Leu104 of may be more easily accessible to water from the cytosol, leading
GAT1 were fitted onto the ␣-carbon backbone of the two GCN-4 (2ZTA) to deprotonation and subsequent mutual helix repulsion. In
leucine zipper helices as outlined under “Experimental Procedures.” serotonin transporter, the substitution of the homologous
The features responsible for TM2 dimer formation are shown in color: Glu136 by cysteine renders this residue sensitive to modifica-
leucine heptad repeat (green), Tyr86 (yellow), Glu101 (red). Residues
Thr89 and Ser103 (both in yellow) are shown to emphasize their irrele- tion by a hydrophilic sulfhydryl-reactive compound (32). How-
vance for oligomerization. B and C, the enlarged Glu101-containing ever, this observation may be compatible with our model if
portion of TM2 dimer. If shielded from access of water from the cytosol replacement by cysteine creates a water-accessible cavity; as
(indicated by the space-filling representation in B), Glu101 on one helix mentioned earlier, in the hydrophobic environment of the lipid
forms a strong hydrogen bond with Glu101 on the other helix. Substitu-
tion of Leu104 to alanine abolishes hydrogen bonding by making Glu101 bilayer, the stable association of leucine heptad repeats re-
water-accessible and ionized (indicated by the gap in C). quires a hydrogen bond donor in the a-position, which is sand-
wiched between two leucines that are one helical turn below
and above in the d-position of the repeat (9, 13). If the side
ring (e.g. ␲-␲ stacking), which supports transporter oligomer-
chain of cysteine is too short to stabilize the leucine zipper, the
ization and subsequent plasma membrane targeting. The phe-
nol ring of tyrosine and the phenyl-side chain of phenylalanine driving force that keeps the leucine side chains aligned may be
have a propensity to interact with the planar hydrophobic reduced, and the cysteine at the bottom of TM2 is likely to
moieties in a stacked or T-shaped conformation (26). These become accessible to water.
interactions between phenyls were shown to drive self-associ- These conjectures are well supported by a statistical assess-
ation of a transmembrane segment of IsK ion channel (27). ment of Adamian and Liang (33); their survey scored the prob-
Thus, Tyr86-mediated stacking is a plausible mechanism. In abilities for residues in TM segments to interact with each
case of parallel stacking of aromatic rings, the hydroxyl-group other within the membrane. In agreement with our results,
obviously must be satisfied by intramolecular hydrogen bond- Adamian and Liang reported that a Glu-Glu pair is frequently
ing (i.e. an interaction within the same transporter moiety) or found in contact regions within the membrane, whereas Asp-
by an interaction with a lipid head group. Asp and Gln-Gln pairs are very unlikely to occur (33). Thus, the
In order to visualize how the transmembrane dimer interface interaction between two Glu residues are expected to support a
may look, we threaded the sequences of two GAT1 TM2 do- strong intramembrane association of two transporter mole-
mains on the ␣-carbon backbone of the GCN4 leucine zipper cules. Recent work suggests that the sequence context strongly
(28). The geometry of the obtained structure was optimized to modulates transmembrane hydrogen bonding interactions (34).
exclude improbable side chain conformations. Due to the pres- Accordingly, the specificity of hydrogen bonding between GAT1
ence of two prolines and one glycine residue, which may distort TM2 helices could be provided by intrinsic properties of the
the ␣-helical structure of the transmembrane segment (29), the Glu101 side chain and by the surrounding residues, which
derived molecular model is but a rough estimation of the TM2 would form a hydrophobic cage.
dimer structure. According to our model, the association of two Our model is strongly supported by the evidence obtained
TM2 helices may be supported in part via a net of extensive van from the ␤-lactamase complementation experiments performed
der Waals contacts between the corresponding hydrophobic with full-length transporters and fragments thereof. The asso-
side chains. In this case, the dimer interface would involve the ciation patterns are similar, and, in fact, the effects of the
leucines of the heptad repeat and Tyr86 (shown in green and mutations are more pronounced when assessed in the isolated
yellow, respectively, in Fig. 7A). These residues would stabilize TM1-TM2 segments. This approach demonstrates that the pu-
the packing of the TM segments against each other. However, tative transmembrane domain interface involving Tyr86 and
we cannot rule out that Tyr86, in particular, may mediate Glu101 is present in full-length GAT1 as well as in its severed
packing against residues on other helices of the full-length TM1-TM2 segment. Thus, the available evidence supports a
transporter. Most importantly, however, the dimer formation model where the interface depicted in Fig. 7A may play a role
appears to be dependent on the glutamates on adjacent TM2 in helical packing; this is apparently true even when the con-
domains of two transporters (shown in red in Fig. 7, A and B). straints are relieved that are projected on TM1-TM2 in the con-
55736 TM2 Controls GABA Transporter-1 Oligomerization
text of an intact transporter and despite the concomitant increase 4, 13–25
3. Zahniser, N. R., and Doolen, S. (2001) Pharmacol. Ther. 92, 21–55
in the conformational flexibility of isolated TM segments. 4. Horiuchi, M., Nicke, A., Gomeza, J., Aschrafi, A., Schmalzing, G., and Betz, H.
It is worth pointing out that a cysteine scanning mutagenesis (2001) Proc. Natl. Acad. Sci. U. S. A. 98, 1448 –1453
5. Sitte, H. H., and Freissmuth, M. (2003) Eur. J. Pharmacol. 479, 229 –236
of TM2 also identified the homologous residues (i.e. Tyr121 and 6. Schmid, J. A., Scholze, P., Kudlacek, O., Freissmuth, M., Singer, E. A., and
Glu136) in serotonin transporter as essential for cell surface Sitte, H. H. (2001) J. Biol. Chem. 276, 3805–3810
expression and for transporter activity (32). Previously, the 7. Scholze, P., Freissmuth, M., and Sitte, H. H. (2002) J. Biol. Chem. 277,
43682– 43690
residue Glu101 of GAT1 was proposed to act as the counterion 8. Torres, G. E., Carneiro, A., Seamans, K., Fiorentini, C., Sweeney, A., Yao,
for binding of the cosubstrate Na⫹ (35). This interpretation W. D., and Caron, M. G. (2003) J. Biol. Chem. 278, 2731–2739
9. Zhou, F. X., Merianos, H. J., Brunger, A. T., and Engelman, D. M. (2001) Proc.
fails to account for the reduction in cellular [3H]GABA uptake Natl. Acad. Sci. U. S. A. 98, 2250 –2255
that Keshet et al. (35) observed with the mutant E101D-GAT1 10. Gurezka, R., and Langosch, D. (2001) J. Biol. Chem. 276, 45580 – 45587
if we assume that aspartate can still provide a counterion for 11. Simmerman, H. K., Kobayashi, Y. M., Autry, J. M., and Jones, L. R. (1996)
J. Biol. Chem. 271, 5941–5946
Na⫹ binding. However, the reduction in cellular uptake and in 12. Seubert, N., Royer, Y., Staerk, J., Kubatzky, K. F., Moucadel, V., Krishnaku-
transporter-associated currents is readily explained by the im- mar, S., Smith, S. O., and Constantinescu, S. N. (2003) Mol. Cell. 12,
1239 –1250
paired cell surface expression of the Glu101 mutants that we 13. Dawson, J. P., Weinger, J. S., and Engelman, D. M. (2002) J. Mol. Biol. 316,
documented in the present study. Accordingly, we consider our 799 – 805
model more plausible, where the carboxyl group of Glu101 is 14. Farhan, H., Korkhov, V. M., Paulitschke, V., Dorostkar, M. M., Scholze, P.,
Kudlacek, O., Freissmuth, M., and Sitte, H. H. (2004) J. Biol. Chem. 279,
shielded from the aqueous phase and is thus unavailable for 28553–28563
binding of Na⫹. Our results rather emphasize the central role 15. Okayama, H., and Berg, P. (1985) Mol. Cell. Biol. 5, 1136 –1142
16. Miyawaki, A., and Tsien, R. Y. (2000) Methods Enzymol. 327, 472–500
of this polar residue Glu101 in driving GAT1 oligomerization by 17. Galarneau, A., Primeau, M., Trudeau, L. E., and Michnick, S. W. (2002) Nat.
intramembrane contacts via adjacent TM2 helices. In our opin- Biotechnol. 20, 619 – 622
ion, this association is supported by a meshwork of hydrophobic 18. Just, H., Sitte, H. H., Schmid, J. A., Freissmuth, M., and Kudlacek, O. (2004)
J. Biol. Chem. 279, 6650 – 6657
residues (leucine heptad repeat and Tyr86) dispersed along the 19. Hastrup, H., Karlin, A., Javitch, J. A. (2001) Proc. Natl. Acad. Sci. U. S. A. 98,
helical interface. It is worth pointing out that the residues 10055–10060
20. Hahn, M. K., Robertson, D., and Blakely, R. D. (2003) J. Neurosci. 23,
involved are conserved among the homologues of GAT1 from 4470 – 4478
archaeae to humans (Fig. 1A). In addition, the leucine zipper- 21. Schmid, J. A., Sitte, H. H. (2003) Curr. Opin. Oncol. 15, 55– 64
like motifs are present in other transport proteins (e.g. GLUT4, 22. Hastrup, H., Sen, N., Javitch, J. A. (2003) J. Biol. Chem. 278, 45045– 45048
23. Margeta-Mitrovic, M., Jan, Y. N., and Jan, L. Y. (2000) Neuron 27, 97–106
Sglt1, etc.). Thus, the mode of transmembrane association that 24. Zerangue, N., Schwappach, B., Jan, Y. N., and Jan, L. Y. (1999) Neuron 22,
we propose for GABA transporter molecules may also govern 537–548
25. Sucic, S., Paczkowski, F. A., Runkel, F., Bonisch, H., Bryan-Lluka, L. J. (2002)
the oligomerization of other members of the transporter family. J. Neurochem. 81, 344 –354
Finally, our work proves that the concepts developed for arti- 26. Chelli, R., Gervasio, F. L., Procacci, P., Schettino, V. (2002) J. Am. Chem. Soc.
ficial single transmembrane segments (9, 10, 34) are applicable 124, 6133– 6143
27. Aggeli, A., Bannister, M. L., Bell, M., Boden, N., Findlay, J. B., Hunter, M.,
to proteins with a more complex architecture (i.e. several trans- Knowles, P. F., and Yang, J. C. (1998) Biochemistry 37, 8121– 8131
membrane ␣-helices). Related combinations of hydrophobic and 28. O’Shea, E. K., Klemm, J. D., Kim, P. S., and Alber, T. (1991) Science 254,
539 –544
polar interactions are likely to support oligomer formation in 29. Cordes, F. S., Bright, J. N., and Sansom, M. S. (2002) J. Mol. Biol. 323,
other proteins with multiple transmembrane spans. 951–960
30. Smith, S. O., Smith, C. S., and Bormann, B. J. (1996) Nat. Struct. Biol. 3,
Acknowledgments—We are grateful to Stephen Michnick for the 252–258
plasmids encoding ␤-lactamase fragments. We thank Frank Eisenhaber 31. Therien, A. G., Grant, F. E., and Deber, C. M. (2001) Nat. Struct. Biol. 8,
597– 601
for helpful suggestions regarding molecular modeling and Christoforos
32. Sato, Y., Zhang, Y. W., Androutsellis-Theotokis, A., and Rudnick, G. (2004)
Charalambous for critical reading of the manuscript. J. Biol. Chem. 279, 22926 –22933
33. Adamian, L., and Liang J. (2001) J. Mol. Biol. 311, 891–907
REFERENCES 34. Dawson, J. P., Melnyk, R. A., Deber, C. M., and Engelman, D. M. (2003) J. Mol.
1. Busch, W., and Saier, M. H., Jr. (2002) Crit. Rev. Biochem. Mol. Biol. 37, Biol. 331, 255–262
287–337 35. Keshet, G. I., Bendahan, A., Su, H., Mager, S., Lester, H. A., Kanner, B. I.
2. Torres, G. E., Gainetdinov, R. R., and Caron, M. G. (2003) Nat. Rev. Neurosci. (1995) FEBS Lett. 371, 39 – 42

You might also like