Вы находитесь на странице: 1из 10

Research

Purification and Identification of G Protein-


coupled Receptor Protein Complexes under
Native Conditions*□ S

Avais M. Daulat‡§¶储**, Pascal Maurice‡§¶储, Carine Froment‡‡,


Jean-Luc Guillaume‡§¶储, Cédric Broussard‡§¶储, Bernard Monsarrat‡‡§§,
Philippe Delagrange¶¶, and Ralf Jockers‡§¶储 储储

G protein-coupled receptors (GPCRs) constitute the larg- half of the drugs prescribed for human diseases (3). GPCRs
est family of membrane receptors and are of major thera- are key controllers of physiological processes such as neuro-
peutic importance. The identification of GPCR-associated transmission, cellular metabolism, secretion, cell differentia-
proteins is an important step toward a better understanding tion, and growth. The prototypic topology of GPCRs consists
of these receptors. However, current methods are not sat- of an extracellular amino-terminal segment, a hydrophobic
isfying as only isolated receptor domains (intracellular core of seven transmembrane (7TM) ␣-helices that interact
loops or carboxyl-terminal tails) can be used as “bait.” We
together to form a three-dimensional barrel within the plasma
report here a method based on tandem affinity purification
membrane, and a cytosolic carboxyl-terminal tail (C-tail).
coupled to mass spectrometry that overcomes these limi-
tations as the entire receptor is used to identify protein Whereas amino acids within the extracellular and/or hydro-
complexes formed in living mammalian cells. The human phobic 7TM core of the receptor are involved in ligand bind-
MT1 and MT2 melatonin receptors were chosen as model ing, the intracellular domain of the receptor composed of
GPCRs. Both receptors were tagged with the tandem affin- three loops and the C-tail is important for signal transduction
ity purification tag at their carboxyl-terminal tails and ex- (4).
pressed in human embryonic kidney 293 cells. Receptor Several strategies have been used to identify GPCR-asso-
solubilization and purification conditions were optimized. ciated complexes. Early work used affinity columns with bio-
The method was validated by the co-purification of Gi pro- tinylated ligands to purify somatostatin receptor-G protein
teins, which are well known GPCR interaction partners but complexes from tissues (5). Subsequently based on the pio-
which are difficult to identify with current protein-protein
neering work of Husi et al. (6), more systematic proteomics
interaction assays. Several new and functionally relevant
analysis of GPCR-associated protein complexes was con-
MT1- and MT2-associated proteins were identified; some of
them were common to both receptors, and others were ducted using receptor-specific antibodies (7). However, the
specific for each subtype. Taken together, our protocol general application of these approaches was limited by the
allowed for the first time the purification of GPCR-associ- availability of adequate tools (labeled ligands, antibodies, etc.)
ated proteins under native conditions in quantities suitable for each GPCR. Later on, isolated intracellular domains were
for mass spectrometry analysis. Molecular & Cellular widely used to identify GPCR-associated proteins either as
Proteomics 6:835– 844, 2007. bait in yeast two-hybrid screens or to generate affinity matri-
ces for the purification of interacting proteins from cell ex-
tracts (8 –11). Using the entire C-tail of the 5HT2c receptor
With more than 800 members, GPCRs1 constitute the larg- expressed as GST fusion protein, more than 15 proteins have
est family of membrane receptors (1, 2). They respond to a been identified (12). Furthermore isolated protein-protein in-
wide variety of extracellular stimuli and are targeted by about teraction motifs such as PDZ domain recognition motifs of
GPCRs have been successfully used to identify interacting
partners of the PDZ domain recognition motifs of the 5HT2a,
From the ‡Department of Cell Biology, Institut Cochin, §INSERM
U567, ¶CNRS UMR 8104, and 储Faculté de Médecine René Descartes,
5HT2c, and 5HT4 receptors (13, 14).
UMR-S 8104, Université Paris Descartes, Paris F-75014, France, Although several GPCR-interacting proteins could be iden-
‡‡Institut de Biologie Structurale et de Pharmacologie, CNRS UMR tified, these methods obviously have important limitations as
5089, Toulouse 31076, France, and ¶¶Institut de Recherches SER- isolated receptor subdomains 1) do not mimic the GPCR
VIER, Suresnes 92150, France topology (7TM domain, arrangement of intracellular loops and
Received, August 7, 2006, and in revised form, November 24, 2006
Published, MCP Papers in Press, January 9, 2007, DOI 10.1074/
mcp.M600298-MCP200 dem affinity purification; TEV, tobacco etch virus; HEK, human em-
1
The abbreviations used are: GPCR, G protein-coupled receptor; bryonic kidney; [125I]MLT, 2-[125I]iodomelatonin; NT, non-transfected;
7TM, seven transmembrane; C-tail, carboxyl-terminal tail; ERK, ex- MAPK, mitogen-activated protein kinase; IRS, insulin receptor sub-
tracellular signal-regulated kinase; MT, melatonin receptor; TAP, tan- strate; PP2, protein phosphatase 2; ER, endoplasmic reticulum.

© 2007 by The American Society for Biochemistry and Molecular Biology, Inc. Molecular & Cellular Proteomics 6.5 835
This paper is available on line at http://www.mcponline.org
GPCR Protein Complexes

the C-tail, and receptor oligomerization), 2) do not provide an solubilized in solubilization buffer supplemented with increasing con-
adequate membrane environment, and 3) do not allow the centrations of CHAPS, Brij96V, dodecylmaltoside, or digitonin. The
soluble fraction was separated from the insoluble fraction by centrif-
recruitment of protein complexes upon agonist activation. Not
ugation at 40000 ⫻ g. The insoluble fraction (pellet) was resuspended
surprisingly, well known interaction partners of GPCRs such in the same buffer, and luciferase activity was measured in the soluble
as heterotrimeric G proteins are difficult to identify using these and resuspended insoluble fraction by adding coelenterazine h (In-
techniques. terchim, Montluçon, France) at a final concentration of 5 ␮M. Read-
Recently a tandem affinity purification (TAP) method has ings were performed with a luminometer at 488 nm (FusionTM, Pack-
ard Instrument Co.). Solubilization yields were defined as the
been described (15). Importantly this method overcomes the
percentage of luciferase activity in the supernatant over total lucifer-
aforementioned limitations and can be potentially applied to ase activity (pellet ⫹ supernatant). Use of Nonidet P-40 was not
any protein. The full-length protein of interest can be ex- compatible with the luciferase activity assay.
pressed in mammalian cells where subcellular localization and Immunofluorescence Microscopy—HEK 293 cells were grown on
post-translational modifications are conserved. In addition, sterile coverslips and fixed and permeabilized for 20 min in ethanol at
the recruitment of protein complexes may be induced by ⫺20 °C. After blocking in PBS, 3% BSA for 20 min, cells were incu-
bated with polyclonal anti-MT1 or anti-MT2 antibodies for 1 h at room
treatment of the cells with different hormonal or pharmaco-
temperature. Coverslips were washed three times with PBS and
logical compounds. To perform a two-step affinity chroma- incubated with a FITC-coupled secondary antibody at 1:1000 dilution
tography purification of complexes formed in intact cells, the in PBS, 3% BSA (Jackson ImmunoResearch Laboratories) for 30 min
TAP method relies on the presence of a TAP tag composed of at room temperature. Coverslips were mounted and analyzed by
two IgG binding domains, a tobacco etch virus (TEV) protease confocal laser microscopy (Leica TCS SP2 AOBS).
SDS-PAGE and Western Blotting—Whole cells (ERK activation) or
cleavage site, and a calmodulin binding domain (16). The TAP
crude membranes (receptor detection) were denatured overnight at
method has been successfully used for high throughput iden- room temperature in SDS-PAGE loading buffer (62.5 mM Tris/HCl, pH
tification of soluble proteins engaged in interacting complexes 6.8, 5% SDS, 10% glycerol, 0.5% bromphenol blue). Proteins were
in yeast and mammalian cells (17, 18). However, purification separated by 10% SDS-PAGE and transferred to nitrocellulose. Im-
of membrane protein complexes in general and of GPCR- munoblot analysis was carried out with polyclonal anti-MT1, anti-MT2
(22), anti-phospho-ERK, or anti-ERK2 antibodies (Santa Cruz Bio-
associated complexes in particular has been unsuccessful.
technology). Immunoreactivity was revealed using secondary anti-
In the present study, we developed a modified TAP method bodies coupled to horseradish peroxidase and the ECL reagent
suitable for the purification of GPCR-associated complexes. (Perbio).
The human MT1 and MT2 melatonin receptors were chosen as Optimized TAP Tag Procedure—All purification steps were con-
model GPCRs. Both receptors were tagged with the TAP tag ducted at 4 °C in the presence of a protease inhibitor mixture (Roche
Applied Science), 1 mM orthovanadate, and 2 mM NaF. For large scale
at their C-tails, and corresponding stable clones were estab-
experiments, crude membranes were prepared from ⬃1 ⫻ 109 HEK
lished in HEK 293 cells. Receptor solubilization and purifica- 293 cells and solubilized overnight in solubilization buffer with 0.5%
tion was optimized in small scale experiments, and receptor- digitonin or 0.25% Brij96V at a concentration of 2 mg of protein/ml.
associated complexes were purified from large scale The supernatant was recovered after centrifugation at 40,000 ⫻ g for
experiments and subsequently identified by nano-LC-nano- 30 min and incubated for 4 h with 400 ␮l of rabbit IgG-Agarose
ESI MS/MS. (Sigma). The resin was washed three times with 1 ml of solubilization
buffer, resuspended in 500 ␮l of the same buffer, and incubated
overnight with 100 units of TEV protease (Invitrogen). The supernatant
EXPERIMENTAL PROCEDURES was collected, mixed with 500 ␮l of calmodulin buffer (75 mM Tris, 5
Receptor Constructs—MT1-Rluc and MT2-Rluc constructs have mM MgCl2, 50 mM CaCl2, and 0.5% digitonin or 0.25% Brij96V, pH
been described elsewhere (19). To obtain the MT1-TAP and MT2-TAP 8.0) and incubated for 2 h with 100 ␮l of calmodulin beads (Strat-
constructs, the TAP tag cassette from the pcDNA3-CMV-TAP plas- agene, La Jolla, CA). Beads were washed five times with 1 ml of
mid (a gift from Nicolas Goardon, Institut Cochin, Paris, France) was calmodulin buffer, and retained proteins were eluted with SDS-PAGE
fused in frame to the 3⬘-end of the MT1 and MT2 coding region. loading buffer.
Cell Culture and Transfection—HEK 293 cells were grown and Mass Spectrometry and Protein Identification—Coomassie Blue-
transfected as described elsewhere (19). Stable cell lines were se- stained or silver-stained (23) bands were excised and subjected to
lected with G418 (Invitrogen). in-gel tryptic digestion using modified porcine trypsin (Promega,
Crude Membrane Preparation, Radioligand Binding Assay, and Sol- Lyon, France) as described previously (24). The tryptic digest was
ubilization—Crude membranes were prepared as described previ- analyzed by on-line capillary HPLC (Dionex/LC Packings) coupled to
ously (20, 21) from non-transfected, MT1-TAP-, or MT2-TAP-express- a nanospray Qq-TOF mass spectrometer (QSTAR Pulsar XL, Applied
ing HEK 293 cells. Membranes were labeled with a saturating Biosystems, Foster City, CA). Peptides were separated on a 75-␮m
concentration (500 pM) of 2-[125I]iodomelatonin ([125I]MLT), and inner diameter ⫻ 15-cm C18 PepMapTM column after loading onto a
[125I]MLT binding sites were determined on crude membranes, solu- 300-␮m inner diameter ⫻ 5-mm PepMap C18 precolumn (Dionex/LC
bilized extracts, or at different steps of the TAP procedure. 0.5% Packings). The flow rate was set at 200 nl/min. Peptides were eluted
CHAPS, 0.25% Brij姞96V, 0.5% digitonin, 0.5% Nonidet P-40 (all from using a 0 –50% linear gradient of solvent B in 50 min (solvent A was
Sigma) and 0.5% dodecylmaltoside (Roche Applied Science) were 0.2% formic acid in 5% acetonitrile, and solvent B was 0.2% formic
used for overnight solubilization at 4 °C in solubilization buffer (75 mM acid in 90% acetonitrile). The mass spectrometer was operated in
Tris, 2 mM EDTA, 5 mM MgCl2, pH 8.0). positive ion mode at a 2.1- kV needle voltage. MS and MS/MS data
Luminescence Measurements—Crude membranes were prepared were continuously acquired in an information-dependent acquisition
from HEK 293 cell lines stably expressing MT1-Rluc or MT2-Rluc and mode consisting of a 10-s cycle time. Within each cycle, a MS

836 Molecular & Cellular Proteomics 6.5


GPCR Protein Complexes

a b

AP
P
1 -T A

2 -T

2
MT

NT
MT

MT
kDa

NT

NT
kDa kDa
97 97
97 MT2-TAP
MT1 66
66 NS 66
56 56
56
42 MT2 42
42
Anti-MT1 Anti-MT2 Anti-MT2

FIG. 1. Functional expression of


MT1-TAP and MT2-TAP in stably
transfected HEK 293 cells. a and b, MT1 MT1-TAP
crude membranes were prepared from
HEK 293 cells stably transfected with the
indicated receptors. Denatured samples c d
were separated by SDS-PAGE and ana-
lyzed by Western blot using anti-MT1 (a)
Anti-MT1
or anti-MT2 (b) antibodies. Subcellular
localization of MT1 (c), MT1-TAP (d), MT2
(e), or MT2-TAP (f) in stably transfected
HEK 293 cells was monitored by immu-
nofluorescence using anti-MT1 (c and d)
Anti-MT2
or anti-MT2 (e and f) antibodies. g, kinet-
ics of melatonin-stimulated (100 nM) ERK
activation in stable clones expressing
e f
MT1, MT1-TAP, MT2, or MT2-TAP. No
activation of the ERK pathway by mela-
MT2 MT2-TAP
tonin was observed in non-transfected
HEK 293 cells (not shown). NS, nonspe-
cific; P-ERK, phospho-ERK.

MT1-TAP MT2-TAP

MT1 MT2

spectrum was accumulated for 1 s over the range m/z 300 –2000 ation of the peak lists, the default charge state was set to 2⫹, 3⫹, and
followed by three MS/MS acquisitions of 3 s each on the three most 4⫹; MS/MS data were centroided and deisotoped with a threshold at
abundant ions in the MS spectrum. A dynamic exclusion duration was 0% of the base peak; MS/MS spectra with less than five peaks were
used to prevent repetitive selection of the same ions within 30 s. rejected; the precursor mass tolerance for grouping was set to 0.1;
MS/MS data were acquired using a 3 m/z unit ion isolation window. and the maximum number of cycles between groups and the mini-
Collision energies were automatically adjusted according to the mum cycles per group were set to 60 and 1, respectively. MS/MS
charge state and mass value of the precursor ions. Peak lists of data were searched against a human sequence in-house database, a
MS/MS spectra were created using Mascot.dll script (version 1.6b21) compilation of UniProt Swiss-Prot and UniProt TrEMBL databases
in Analyst QS software (version 1.1, Applied Biosystems). For gener- (72,049 entries, versions 49.1 and 32.1, respectively) using the Mas-

Molecular & Cellular Proteomics 6.5 837


GPCR Protein Complexes

100 100

Solubilization
(% of total)
FIG. 2. Detergent selection and opti-
mization of solubilization conditions 75 75
of melatonin receptors. a and b, mem-
50 50
brane preparations of HEK 293 cells that
stably express MT1-Rluc or MT2-Rluc 25 25
were incubated either for 15 h with in- MT1 MT2
creasing detergent concentrations at 0 0
4 °C (a) or for the indicated times in the 0 0.25 0.50 0.75 1.00 0 0.25 0.50 0.75 1.00
Detergent conc. (%) Detergent conc. (%)
presence of 0.5% CHAPS (●), digitonin
(E), or dodecylmaltoside (䡺) or 0.25% 100 100
Brij96V (f) (b). Luciferase activity was

Solubilization
75 75

(% of total)
measured in the soluble and insoluble
fraction to determine the percentage of
50 50
solubilization. The use of Nonidet P-40
was not compatible with the luciferase 25 25
activity assay. c, membrane prepara- MT1 MT2
tions of HEK 293 cells that stably ex- 0 0
press MT1-TAP or MT2-TAP were la- 0 4 8 12 16 0 4 8 12 16
beled with [125I]MLT, incubated Time (h) Time (h)
overnight at 4 °C in the presence of a
0.5% concentration of the indicated de-
tergents with the exception of Brij96V, MT1 MT2
which was used at 0.25%. The amount
of radioactivity in the soluble fraction
was determined (black bars), [125I]MLT-
labeled MT1-TAP and MT2-TAP were re-
covered on IgG-coated beads, and the
amount of recovered radioactivity was
determined (white bars). The amount of
[125I]MLT binding to non-transfected
HEK 293 cells was negligible.

cot姞 search engine (version 2.1.04). Up to two trypsin missed cleav- of human origin, are extremely well characterized in terms of
ages were allowed, and the mass tolerance for peptide and MS/MS
GPCR signal transduction, can be produced in quantities that
fragment ions was 0.5 Da. Cysteine carbamidomethylation and pro-
pionamide and methionine oxidation were set as variable modifica- are compatible with proteomics analysis, and do not express
tions. Identification was considered positive if the protein was iden- significant amounts of endogenous melatonin receptors.
tified with at least one peptide with an ion score greater than the Western blot analysis with receptor-specific antibodies re-
Mascot significance threshold of 36 (p ⬍ 0.05). For the protein with a vealed immunoreactive bands at the expected molecular size
score close to the threshold value, the identification was confirmed by
manual interpretation of corresponding MS/MS data. To evaluate the
of 85 kDa in an HEK-MT1-TAP clone expressing 1.0 ⫾ 0.2
false-positive rate in these large scale experiments, we repeated the pmol of MT1-TAP/mg of protein (n ⫽ 3) and in an HEK-MT2-
searches using identical search parameters and validation criteria TAP clone expressing 340 ⫾ 41 fmol of MT2-TAP/mg of
against a random database made of the same compilation in which protein (n ⫽ 3) (Fig. 1, a and b). The affinity constants (Kd) of
the sequences have been reversed. These statistical analyses pro-
the melatonin receptor agonist [125I]MLT were 85 ⫾ 53 (n ⫽
vided respectively 2.1, 4.1, and 5.8% false-positive rate for MT1-TAP,
MT2-TAP, and control experiments. See Supplemental Tables I and II 3) and 267 ⫾ 78 pM (n ⫽ 3) for MT1-TAP and MT2-TAP,
for a detailed list of identified peptides. respectively, which are in agreement with previously re-
ported values for wild-type receptors (25). Expression of
RESULTS AND DISCUSSION receptors at the cell surface was assessed by fluorescence
Functional Expression of MT1-TAP and MT2-TAP Fusion microscopy using receptor-specific antibodies (Fig. 1, c–f).
Proteins in HEK 293 Cells TAP-tagged and wild-type receptors were all expressed at
The human MT1 and MT2 melatonin receptors were tagged the plasma membrane of stably transfected HEK 293
with the TAP tag at their C-tails, and corresponding stable clones. Incubation of HEK-MT1-TAP and HEK-MT2-TAP
clones were established in HEK 293 cells. HEK 293 cells are clones with melatonin led to the expected transient increase

838 Molecular & Cellular Proteomics 6.5


GPCR Protein Complexes

100

Purification yield (% of total)


a MT1-TAP
80
60
40
20

0
FIG. 3. Purification of MT1-TAP and Total Sol IgG TEV CAM
MT2-TAP and associated Gi proteins.
100
b

Purification yield (% of total)


a and b, monitoring of the purification
of the digitonin-solubilized (white bars) MT2-TAP
or Brij96V-solubilized (black bars) 80
[125I]MLT-labeled MT1-TAP (a) and MT2-
TAP (b) at each step of the TAP protocol.
The purification yield was expressed as
60
percentage of total membrane [125I]MLT
binding sites. CAM, eluate from calmod- 40
ulin column; IgG, bound to IgG-coated
column; Sol, solubilized fraction; TEV, 20
elution with TEV protease. c, monitoring
of the co-purification of receptor-associ- 0
ated Gi␣ proteins by Western blot from
melatonin-stimulated cells (15 min, 1
Total Sol IgG TEV CAM
␮M). mb, crude membrane fraction.
c Solubilization IgG column CAM column
Cells mb Supernatant Pellet TEV Eluate Unretained Eluate
MT1 - + - + - + - + - + - +
Giα
CAM column
Eluate
MT2 - +
Giα

of ERK1/2 phosphorylation, which was comparable to that 50% (CHAPS), 65% (Brij96V), 70% (digitonin), or 80% (dode-
obtained for wild-type receptors (Fig. 1g). Altogether these cylmaltoside). Data obtained with Nonidet P-40 could not be
data demonstrate that the addition of the TAP tag does not analyzed in this assay as Nonidet P-40 inhibits the luciferase
affect the subcellular localization and the functionality of the activity. Comparable results were obtained for MT1-Rluc and
melatonin receptor. MT2-Rluc. Minimal detergent concentrations, which gave max-
Optimization of Receptor Solubilization and Purification— imal receptor solubilization, were used to study the solubiliza-
The crucial step for successful purification of GPCRs and tion kinetics (Fig. 2b). Both receptors solubilized progressively
associated proteins consists of mild but efficient solubilization with time reaching a plateau after 15 h. For further experiments,
of cells to extract maximal amounts of intact membrane- receptors were solubilized with 0.5% CHAPS, digitonin, dode-
bound complexes. To quantify the amount of solubilized MT1 cylmaltoside, or Nonidet P-40 or with 0.25% for Brij96V for 15 h.
and MT2, we used cells expressing Rluc (Renilla luciferase)- To evaluate the topological integrity of solubilized MT1-TAP
tagged MT1 and MT2 receptors (19). These cells were incu- and MT2-TAP, their ability to bind [125I]MLT was used. Mem-
bated overnight with varying concentrations of detergents, branes prepared from HEK-MT1-TAP and HEK-MT2-TAP cells
and the solubilization yield was determined by measuring were labeled with [125I]MLT. The receptors were solubilized
luciferase activity in the soluble and non-soluble fractions (Fig. and immobilized on IgG-coated beads via the IgG binding
2a). The amount of solubilized receptor increased as a func- modules of the TAP tag (Fig. 2c). Digitonin and Brij96V were
tion of the detergent concentration and reached maxima at chosen for further experiments as 40 –50% of [125I]MLT bind-

Molecular & Cellular Proteomics 6.5 839


GPCR Protein Complexes

visible in non-transfected (NT) HEK 293 cells, several specific


protein bands were reproducibly present in MT1-TAP- and
MT2-TAP-expressing cells (n ⫽ 4). Lanes were systematically
excised and digested with trypsin, and the resulting peptides
were analyzed by nano-LC-nano-ESI MS/MS and identified
with Mascot software in Swiss-Prot and TrEMBL databases.
Several abundantly expressed proteins, mostly of mitochon-
drial or ribosomal origin, were detected in all three lanes
(NT, MT1-TAP, and MT2-TAP) and were classified as non-
specific proteins. The proteins repeatedly present in the
MT1-TAP or MT2-TAP lane but absent from the NT lane were
considered to be specifically associated to MT1 or MT2,
respectively (Tables I and II). Consistently both bait proteins
(MT1 and MT2) were identified in the corresponding lanes.
Each receptor was identified in two regions, at 45 and 90
FIG. 4. Large scale purification of the MT1-TAP (a) and MT2-
kDa, corresponding to the well documented monomeric and
TAP-associated (b) complex. Approximately 1 ⫻ 109 HEK-MT1- SDS-resistant dimeric form of the receptors, respectively
TAP, HEK-MT2-TAP, and non-transfected HEK 293 cells were sub- (26). This indicates that both receptors reached their fully
mitted to the TAP protocol. Eluates were separated by gel functional quaternary structure.
electrophoresis, and proteins were detected by Coomassie Blue Importantly the presence of heterotrimeric G proteins in
staining (a) or silver staining (b).
MT1- and MT2-associated complexes was confirmed by mass
spectrometry. Consistent with the known coupling of melato-
ing sites were routinely retained on IgG beads under these nin receptors to Gi proteins, all three Gi␣ isoforms (specific
conditions. Using these optimized solubilization conditions, peptides for Gi␣1–3) and two different G␤ isoforms (specific
the entire TAP procedure was carried out. The purification of peptides for G␤1,4) were identified. Co-purification of G pro-
functional receptors was monitored at each step with the teins and receptors validates our method and provides a
[125I]MLT binding assay. The overall yield of [125I]MLT-labeled major advantage compared with other currently available pro-
receptors varied from 27 ⫾ 3 (digitonin) to 15 ⫾ 2% (Brij96V) tein-protein interaction assays where the G protein/receptor
and from 33 ⫾ 2 (digitonin) to 25 ⫾ 5% (Brij96V) (n ⬎ 5) for interaction is generally lost. These results clearly demonstrate
MT1 and MT2, respectively (Fig. 3, a and b). The co-purifica- that our procedure can identify functionally relevant GPCR-
tion of associated proteins upon melatonin stimulation was interacting proteins that associate only with intact receptors
evaluated by the presence of the ␣ subunit of the heterotri- expressed in the natural membrane environment.
meric Gi protein (Gi␣) (20, 21). Whereas Gi␣ was readily de- Several previously unknown melatonin receptor-associated
tected throughout the purification of digitonin-solubilized proteins were identified. Interestingly these proteins localized
MT1-TAP and MT2-TAP (Fig. 3c), Gi␣ was rarely co-purified to different subcellular compartments (cytosol, plasma mem-
when using Brij96V (not shown) because this detergent ap- brane, and different intracellular membrane compartments
parently destabilized the receptor/G protein interaction. The such as the endoplasmic reticulum). Melatonin receptor-as-
integrity of the heterotrimeric G protein was further confirmed sociated proteins could be divided into three functionally dis-
by the presence of the G␤ subunit at the final purification step tinct groups: proteins likely to be involved in receptor biosyn-
in the presence of digitonin (not shown), which was used for thesis, intracellular trafficking, and signaling/regulation of
further experiments. GPCRs (Tables I and II). The function of remaining proteins,
Purification and Identification of MT1- and MT2-associated classified as “others,” are unknown or appear not to relate
Proteins directly to known GPCR function. This is the case for the
The ultimate aim of the TAP tag procedure is the purification MT1-specific heterogeneous nuclear ribonucleoprotein A0
of sufficient amounts of receptor to identify associated pro- that is suspected to participate in mRNA maturation (27) and
teins by mass spectrometry analysis. To reach this goal, is a major substrate for MAPK-activated protein kinase 2,
crude membranes of ⬃1 ⫻ 109 HEK-MT1-TAP, HEK-MT2- which is itself activated upon GPCR-promoted p38 MAPK
TAP, and non-transfected HEK 293 cells were prepared, and stimulation (28).
the digitonin-solubilized fraction was submitted to the TAP Filamin A and insulin receptor substrate 4 (IRS4) were iden-
procedure. Eluates were separated by one-dimensional gel tified as common members of MT1- and MT2-associated
electrophoresis, and depending on the receptor expression complexes. Consistent with these findings, filamin A has been
levels, proteins were detected either by Coomassie Blue shown previously to interact with several other members of
(MT1-TAP, ⬃1 pmol/mg) or by silver staining (MT2-TAP, ⬃0.3 the GPCR family, including dopamine D2/D3 (29, 30), calci-
pmol/mg) (Fig. 4, a and b). Whereas only a few bands were um-sensing (31, 32), and ␮-opioid receptors (33). The role of

840 Molecular & Cellular Proteomics 6.5


GPCR Protein Complexes

TABLE I
MT1-associated proteins
Trypsin-digested protein bands were analyzed by nano-LC-nano-ESI MS/MS, and proteins were identified with Mascot software in
Swiss-Prot and TrEMBL databases. Comparison of proteins identified in eluates from MT1-TAP-expressing cells with those of non-transfected
HEK 293 cells led to the identification of MT1-specific proteins. Data represent the common results of four experiments.
Number Theoretical Also
Swiss-Prot/TrEMBL Subcellular
Protein identity Score Coverage of unique molecular identified
accession no. localization
peptides mass in MT2
% Da

Bait protein
P48039 Melatonin receptor type 1A (MT1) 353 24 9 39,349 ⫹ Plasma membrane
Signal transduction
P62873 Guanine nucleotide-binding protein ␤ 405 29 10 37,222 ⫹ Plasma membrane
subunit 1
Q9HAV0 Guanine nucleotide-binding protein ␤ 318 22 8 37,412 ⫹ Plasma membrane
subunit 4
P08754 Guanine nucleotide-binding protein Gi␣3 639 46 16 40,375 ⫹ Plasma membrane
P63096 Guanine nucleotide-binding protein Gi␣1 566 46 14 40,204 ⫹ Plasma membrane
P04899 Guanine nucleotide-binding protein Gi␣2 409 34 11 40,294 ⫹ Plasma membrane
P09543 2⬘,3⬘-cyclic-nucleotide 3⬘- 97 7 3 47,549 ⫺ Plasma membrane
phosphodiesterase
P62834 Ras-related protein RAP-1A 93 11 2 20,974 ⫺ Plasma membrane
P63000 p21-Rac1 82 8 2 21,436 ⫺ Membrane
P26641 Elongation factor 1-␥ 197 11 5 49,956 ⫺ Cytoplasm
O14654 Insulin receptor substrate 4 146 4 4 133,685 ⫹ Cytoplasm
Q60FE5 Filamin A 94 1 3 278,053 ⫹ Cytoplasm
Biosynthesis
P27824 Calnexin precursor 837 30 20 67,526 ⫹ ER membrane
P11021 78-kDa glucose-regulated protein 511 25 16 72,288 ⫹ ER lumen
P27797 Calreticulin 189 12 7 48,112 ⫺ ER lumen
Q15084 Protein-disulfide isomerase A6 155 7 3 48,091 ⫹ ER lumen
Traffic
O95292 Vesicle-associated membrane protein- 83 15 4 27,080 ⫺ Membrane
associated protein B/C
P61026 Ras-related protein Rab-10 121 14 3 22,527 ⫺ Endosome
Others
O00264 Membrane-associated progesterone 191 24 6 21,527 ⫺ Plasma membrane
receptor component 1
Q96AG4 Leucine-rich repeat-containing protein 59 312 36 9 34,909 ⫺ Intracellular
membrane
P57088 Transmembrane protein 33 102 12 3 27,960 ⫺ Membrane
Q13151 Heterogeneous nuclear ribonucleoprotein 377 28 7 30,822 ⫺ Cytoplasm
A0

IRS4 is less well documented. The involvement of IRS4 in interaction with the receptor (38, 39).
fibroblast growth factor receptor signaling (34) and interaction Catenin ␦1 (p120) and the protein phosphatase 2C␥
with the protein phosphatase 4 have been described (35). (PP2C␥) have been identified as MT2-specific signaling pro-
We were also able to identify several MT1-specific signaling teins. Whereas p120 is known to affect intracellular signaling
proteins such as Rac1, RAP-1A, and the 2⬘,3⬘-cyclic-nucleo- by NF␬B activation through regulation of Rho GTPases, its
tide 3⬘-phosphodiesterase and the protein elongation factor specific role in GPCR signaling is currently unknown (40).
1-␥ (eEF-1B␥). Interestingly the small GTPases Rac1 and Several serine/threonine phosphatases participate in the de-
RAP-1 have been shown to function downstream of 5HT4 phosphorylation of activated GPCRs (41). Phosphatases of
receptors and the cAMP guanine nucleotide exchange factor the PP2A and PP2B subfamilies have been reported to target
Epac1 (36). The 2⬘,3⬘-cyclic-nucleotide 3⬘-phosphodiester- GPCRs, whereas PP2C subfamily members have been shown
ase, belonging to the PDE3A family, is involved in the degra- to dephosphorylate the metabotropic glutamate receptor 3
dation of second messengers such as cAMP and cGMP (37). (42). It will be interesting to determine whether PP2C␥ partic-
Activation of melatonin receptors is known to modulate both ipates in MT2 dephosphorylation.
second messengers (25). eEF-1B␥ and other elongation fac- Most of the identified proteins that are involved in receptor
tors have been reported to modulate GPCR function by direct biosynthesis are present in both receptor-associated com-

Molecular & Cellular Proteomics 6.5 841


GPCR Protein Complexes

TABLE II
MT2-associated proteins
Trypsin-digested protein bands were analyzed by nano-LC-nano-ESI MS/MS, and proteins were identified with Mascot software in
Swiss-Prot and TrEMBL databases. Comparison of proteins identified in eluates from MT2-TAP-expressing cells with those of non-transfected
HEK 293 cells led to the identification of MT2-specific proteins. Data represent the common results of four experiments.
Number Theoretical Also
Swiss-Prot/TrEMBL
Protein identity Score Coverage of unique molecular identified Subcellular localization
accession no.
peptides mass in MT1
% Da

Bait protein
P49286 Melatonin receptor type 1B (MT2) 191 10 4 40,162 Plasma membrane
Signal transduction
P62879 Guanine nucleotide-binding protein ␤ 929 52 15 37,222 ⫹ Plasma membrane
subunit 1
Q9HAV0 Guanine nucleotide-binding protein ␤ 511 29 10 37,412 ⫹ Plasma membrane
subunit 4
P63096 Guanine nucleotide-binding protein Gi␣1 905 60 20 40,204 ⫹ Plasma membrane
P08754 Guanine nucleotide-binding protein Gi␣3 899 59 19 40,375 ⫹ Plasma membrane
P04899 Guanine nucleotide-binding protein Gi␣2 775 64 20 40,294 ⫹ Plasma membrane
O14654 Insulin receptor substrate 4 546 12 16 133,685 ⫹ Cytoplasm
Q60FE5 Filamin A 369 6 14 278,053 ⫹ Cytoplasm
O60716 Catenin ␦1 126 6 4 108,103 ⫺ Cytoplasm
O15355 Protein phosphatase 2C isoform ␥ 119 5 3 59,235 ⫺ Cytoplasm
Biosynthesis
P27824 Calnexin 747 28 18 67,526 ⫹ ER membrane
P11021 78-kDa glucose-regulated protein 641 28 16 72,288 ⫹ ER lumen
Q15084 Protein-disulfide isomerase A6 235 16 4 48,091 ⫹ ER lumen
P27348 14-3-3 protein ␪ 96 8 2 27,747 ⫺ Cytoplasm
Traffic
Q5T201 Coatomer protein complex, ␣ subunit 547 15 20 138,258 ⫺ ER/Golgi
Q96QK1 Vacuolar sorting protein 35 377 15 11 91,649 ⫺ Membrane-associated
protein
P42356 Phosphatidylinositol 4-kinase ␣ 134 3 4 231,143 ⫺ Intracellular membrane
Others
Q9NQX7 Integral membrane protein 2C 80 12 2 30,224 ⫺ Membrane

plexes. This is expected because all GPCRs are suspected to cate that melatonin receptors (43), like most other GPCRs,
follow the same biosynthetic pathway. Interestingly identified interact with ␤-arrestins, a family of proteins involved in
proteins interact with different distinct receptor domains in- receptor desensitization, internalization, and signaling (44).
cluding the cytoplasmic and the endoplasmic reticulum (ER) This interaction is known to be agonist-stimulated and tran-
luminal receptor interface. In contrast, proteins involved in sient. The absence of ␤-arrestin in our data set suggests
trafficking differ clearly between the two receptor subtypes that this transient interaction may need to be stabilized for
indicating different trafficking behavior. instance by chemically cross-linking the partners prior to
Apart from heterotrimeric Gi proteins, not much is known purification.
about the repertoire of melatonin receptor interaction part- Purification of protein complexes from intact mammalian
ners. This makes it difficult to estimate the proportion of cells is one of the advantages of the TAP technique. This also
known interaction partners that are covered by our data set. implies that the components of the interacting complexes
However, a rough estimation can be made with the assump- depend on the chosen cellular context. As we selected HEK
tion that many interaction partners are likely to interact at least 293 fibroblasts for our study we expected to identify rather
with several other GPCRs. For MT1 and MT2 35 and 45%, ubiquitously expressed interaction partners than cell type-
respectively, of the identified proteins have been shown to specific (i.e. neuron-specific) partners. The repertoire of pro-
interact with other GPCRs. An additional 25 and 20% of the teins identified for both melatonin receptors confirmed this
MT1 and MT2 interactors, respectively, have been associated prediction. The next step will be to identify cell type-specific
with GPCR function (signaling, trafficking, etc.). Thus the interaction partners by expressing TAP-tagged melatonin re-
overall rate of interaction partners that “make sense” repre- ceptors in neurons or endocrine cells that express endoge-
sents ⬃60% for both receptors. nous receptors with well defined functions.
Did we miss any known/suspected interaction partners? Taken together, our TAP protocol allowed for the first time
Although not directly shown, functional studies clearly indi- the purification of GPCR-associated proteins under native

842 Molecular & Cellular Proteomics 6.5


GPCR Protein Complexes

conditions in quantities suitable for mass spectrometry anal- protein-coupled receptors. Trends Pharmacol. Sci. 22, 513–518
9. El Far, O., and Betz, H. (2002) G-protein-coupled receptors for neurotrans-
ysis. Interaction partners from different cellular compartments mitter amino acids: C-terminal tails, crowded signalosomes. Biochem. J.
recognizing extra- and intracellular receptor domains of mo- 2, 329 –336
nomeric and/or dimeric receptor species were identified. This 10. Bockaert, J., Marin, P., Dumuis, A., and Fagni, L. (2003) The ‘magic tail’ of
G protein-coupled receptors: an anchorage for functional protein net-
presents a major methodological advance in the identification
works. FEBS Lett. 546, 65–72
of GPCR-associated protein complexes. In addition, this 11. Bockaert, J., Roussignol, G., Becamel, C., Gavarini, S., Joubert, L., Dumuis,
method is relatively fast, generates a low number of nonspe- A., Fagni, L., and Marin, P. (2004) GPCR-interacting proteins (GIPs):
nature and functions. Biochem. Soc. Trans. 32, 851– 855
cific proteins, and needs no confirmation of the interaction by
12. Becamel, C., Alonso, G., Geleotti, N., Demey, E., Jouin, P., Ullmer, C.,
co-immunoprecipitation experiments. Similar results were ob- Dumuis, A., Bockaert, J., and Marin, P. (2002) Synaptic multiprotein
tained between MT1 and MT2 in terms of solubilization effi- complexes associated with 5-HT2C receptors: a proteomic approach.
ciency, complex stability, and purification yields with this pro- EMBO J. 21, 2332–2342
13. Becamel, C., Gavarini, S., Chanrion, B., Alonso, G., Galeotti, N., Dumuis, A.,
tocol, suggesting a more general application. The increasing Bockaert, J., and Marin, P. (2004) The serotonin 5-HT2A and 5-HT2C
number of TAP tag variants, including the split TAP tag (16), receptors interact with specific sets of PDZ proteins. J. Biol. Chem. 279,
demonstrates the flexibility of this method and allows the 20257–20266
14. Joubert, L., Hanson, B., Barthet, G., Sebben, M., Claeysen, S., Hong, W.,
possibility of rapid optimization for any GPCR homo- and Marin, P., Dumuis, A., and Bockaert, J. (2004) New sorting nexin (SNX27)
heterodimer. and NHERF specifically interact with the 5-HT4a receptor splice variant:
roles in receptor targeting. J. Cell Sci. 117, 5367–5379
Acknowledgments—We are grateful to N. Goardon (Institut Cochin, 15. Rigaut, G., Shevchenko, A., Rutz, B., Wilm, M., Mann, M., and Seraphin, B.
Paris, France) and to Drs. G. Fraschini (Milan, Italy) and D. Angeloni (1999) A generic protein purification method for protein complex char-
(Pisa, Italy) for kindly providing the TAP tag expression cassette and acterization and proteome exploration. Nat. Biotechnol. 17, 1030 –1032
anti-melatonin receptor antibodies, respectively. We thank Patty 16. Dziembowski, A., and Seraphin, B. (2004) Recent developments in the
Chen for comments on the manuscript, Dr. Luc Camoin for expert analysis of protein complexes. FEBS Lett. 556, 1– 6
17. Gavin, A. C., Bosche, M., Krause, R., Grandi, P., Marzioch, M., Bauer, A.,
advice, and Viola Baradari for help in the initial phase of the project.
Schultz, J., Rick, J. M., Michon, A. M., Cruciat, C. M., Remor, M., Hofert,
C., Schelder, M., Brajenovic, M., Ruffner, H., Merino, A., Klein, K., Hudak,
* This work was supported in part by grants from SERVIER, IN- M., Dickson, D., Rudi, T., Gnau, V., Bauch, A., Bastuck, S., Huhse, B.,
SERM, and CNRS. The costs of publication of this article were de- Leutwein, C., Heurtier, M. A., Copley, R. R., Edelmann, A., Querfurth, E.,
frayed in part by the payment of page charges. This article must Rybin, V., Drewes, G., Raida, M., Bouwmeester, T., Bork, P., Seraphin,
therefore be hereby marked “advertisement” in accordance with 18 B., Kuster, B., Neubauer, G., and Superti-Furga, G. (2002) Functional
U.S.C. Section 1734 solely to indicate this fact. organization of the yeast proteome by systematic analysis of protein
□S The on-line version of this article (available at http://www. complexes. Nature 415, 141–147
mcponline.org) contains supplemental material. 18. Bouwmeester, T., Bauch, A., Ruffner, H., Angrand, P. O., Bergamini, G.,
** Holds an EGIDE fellowship. Croughton, K., Cruciat, C., Eberhard, D., Gagneur, J., Ghidelli, S., Hopf,
C., Huhse, B., Mangano, R., Michon, A. M., Schirle, M., Schlegl, J.,
§§ Supported by grants from Génopole Toulouse Midi-Pyrénées
Schwab, M., Stein, M. A., Bauer, A., Casari, G., Drewes, G., Gavin, A. C.,
and Région Midi Pyrénées.
Jackson, D. B., Joberty, G., Neubauer, G., Rick, J., Kuster, B., and
储储 To whom correspondence should be addressed. Tel.: 331-40- Superti-Furga, G. (2004) A physical and functional map of the human
51-64-34; Fax: 331-40-51-64-30; E-mail: jockers@cochin.inserm.fr. TNF-␣/NF-␬B signal transduction pathway. Nat. Cell Biol. 6, 97–105
19. Ayoub, M. A., Couturier, C., Lucas-Meunier, E., Angers, S., Fossier, P.,
REFERENCES Bouvier, M., and Jockers, R. (2002) Monitoring of ligand-independent
1. Fredriksson, R., Lagerstrom, M. C., Lundin, L. G., and Schioth, H. B. (2003) dimerization and ligand-induced conformational changes of melatonin
The G-protein-coupled receptors in the human genome form five main receptors in living cells by bioluminescence resonance energy transfer.
families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol. J. Biol. Chem. 277, 21522–21528
Pharmacol. 63, 1256 –1272 20. Barrett, P., MacLean, A., and Morgan, P. J. (1994) Evidence for multiple
2. Vassilatis, D. K., Hohmann, J. G., Zeng, H., Li, F., Ranchalis, J. E., Mortrud, forms of melatonin receptor-G-protein complexes by solubilization and
M. T., Brown, A., Rodriguez, S. S., Weller, J. R., Wright, A. C., Bergmann, gel electrophoresis. J. Neuroendocrinol. 6, 509 –515
J. E., and Gaitanaris, G. A. (2003) The G protein-coupled receptor rep- 21. Brydon, L., Roka, F., Petit, L., deCoppet, P., Tissot, M., Barrett, P., Morgan,
ertoires of human and mouse. Proc. Natl. Acad. Sci. U. S. A. 100, P. J., Nanoff, C., Strosberg, A. D., and Jockers, R. (1999) Dual signaling
4903– 4908 of human Mel1a melatonin receptors via Gi2, Gi3, and Gq/11 proteins. Mol.
3. Nature Reviews Drug Discovery GPCR Questionnaire Participants (2004) Endocrinol. 13, 2025–2038
The state of GPCR research in 2004. Nat. Rev. Drug Discov. 3, 575, 22. Angeloni, D., Longhi, R., and Fraschini, E. (2000) Production and charac-
577– 626 terization of antibodies directed against the human melatonin receptors
4. Gainetdinov, R. R., Premont, R. T., Bohn, L. M., Lefkowitz, R. J., and Caron, Mel-1a (Mt1) and Mel-1b (MT2). Eur. J. Histochem. 44, 199 –204
M. G. (2004) Desensitization of G protein-coupled receptors and neuro- 23. Rabilloud, T. (1999) Silver staining of 2-D electrophoresis gels. Methods
nal functions. Annu. Rev. Neurosci. 27, 107–144 Mol. Biol. 112, 297–305
5. Brown, P. J., and Schonbrunn, A. (1993) Affinity purification of a soma- 24. Shevchenko, A., Wilm, M., Vorm, O., and Mann, M. (1996) Mass spectro-
tostatin receptor-G-protein complex demonstrates specificity in recep- metric sequencing of proteins silver-stained polyacrylamide gels. Anal.
tor-G-protein coupling. J. Biol. Chem. 268, 6668 – 6676 Chem. 68, 850 – 858
6. Husi, H., Ward, M. A., Choudhary, J. S., Blackstock, W. P., and Grant, 25. Petit, L., Lacroix, I., deCoppet, P., Strosberg, A. D., and Jockers, R. (1999)
S. G. N. (2000) Proteomic analysis of NMDA receptor-adhesion protein Differential signaling of human Mel1a and Mel1b melatonin receptors
signaling complexes. Nat. Neurosci.3, 661– 669 through the cyclic guanosine 3⬘-5⬘-monophosphate pathway. Biochem.
7. Farr, C. D., Gafken, P. R., Norbeck, A. D., Doneanu, C. E., Stapels, M. D., Pharmacol. 58, 633– 639
Barofsky, D. F., Minami, M., and Saugstad, J. A. (2004) Proteomic 26. Ayoub, M. A., Levoye, A., Delagrange, P., and Jockers, R. (2004) Prefer-
analysis of native metabotropic glutamate receptor 5 protein complexes ential formation of MT1/MT2 melatonin receptor heterodimers with dis-
reveals novel molecular constituents. J. Neurochem. 91, 438 – 450 tinct ligand interaction properties compared with MT2 homodimers. Mol.
8. Milligan, G., and White, J. H. (2001) Protein-protein interactions at G- Pharmacol. 66, 312–321

Molecular & Cellular Proteomics 6.5 843


GPCR Protein Complexes

27. Krecic, A. M., and Swanson, M. S. (1999) hnRNP complexes: composition, glio, J., Zugaza, J. L., Fischmeister, R., and Lezoualc’h, F. (2003)
structure, and function. Curr. Opin. Cell Biol. 11, 363–371 Crosstalk between Rap1 and Rac regulates secretion of sAPP␣. Nat. Cell
28. Rousseau, S., Morrice, N., Peggie, M., Campbell, D. G., Gaestel, M., and Biol. 5, 633– 639
Cohen, P. (2002) Inhibition of SAPK2a/p38 prevents hnRNP A0 phos- 37. Lugnier, C. (2006) Cyclic nucleotide phosphodiesterase (PDE) superfamily:
phorylation by MAPKAP-K2 and its interaction with cytokine mRNAs. a new target for the development of specific therapeutic agents. Phar-
EMBO J. 21, 6505– 6514 macol. Ther. 109, 366 –398
29. Li, M., Bermak, J. C., Wang, Z. W., and Zhou, Q. Y. (2000) Modulation of 38. McClatchy, D. B., Knudsen, C. R., Clark, B. F., Kahn, R. A., Hall, R. A., and
dopamine D2 receptor signaling by actin-binding protein (ABP-280). Mol. Levey, A. I. (2002) Novel interaction between the M4 muscarinic acetyl-
Pharmacol. 57, 446 – 452 choline receptor and elongation factor 1A2. J. Biol. Chem. 277,
30. Lin, R., Karpa, K., Kabbani, N., Goldman-Rakic, P., and Levenson, R. (2001) 29268 –29274
Dopamine D2 and D3 receptors are linked to the actin cytoskeleton via 39. Cho, D. I., Oak, M. H., Yang, H. J., Choi, H. K., Janssen, G. M., and Kim,
interaction with filamin A. Proc. Natl. Acad. Sci. U. S. A. 98, 5258 –5263 K. M. (2003) Direct and biochemical interaction between dopamine D3
31. Awata, H., Huang, C., Handlogten, M. E., and Miller, R. T. (2001) Interaction
receptor and elongation factor-1B␤␥. Life Sci. 73, 2991–3004
of the calcium-sensing receptor and filamin, a potential scaffolding pro-
40. Perez-Moreno, M., Davis, M. A., Wong, E., Pasolli, H. A., Reynolds, A. B.,
tein. J. Biol. Chem. 276, 34871–34879
and Fuchs, E. (2006) p120-catenin mediates inflammatory responses in
32. Hjalm, G., MacLeod, R. J., Kifor, O., Chattopadhyay, N., and Brown, E. M.
the skin. Cell 124, 631– 644
(2001) Filamin-A binds to the carboxyl-terminal tail of the calcium-sens-
41. Shih, M. L., Lin, F. B., Scott, J. D., Wang, H. Y., and Malbon, C. C. (1999)
ing receptor, an interaction that participates in CaR-mediated activation
Dynamic complexes of ␤2-adrenergic receptors with protein kinases and
of mitogen-activated protein kinase. J. Biol. Chem. 276, 34880 –34887
33. Onoprishvili, I., Andria, M. L., Kramer, H. K., Ancevska-Taneva, N., Hiller, phosphatases and the role of gravin. J. Biol. Chem. 274, 1588 –1595
J. M., and Simon, E. J. (2003) Interaction between the ␮ opioid receptor 42. Flajolet, M., Rakhilin, S., Wang, H., Starkova, N., Nuangchamnong, N.,
and filamin A is involved in receptor regulation and trafficking. Mol. Nairn, A. C., and Greengard, P. (2003) Protein phosphatase 2C binds
Pharmacol. 64, 1092–1100 selectively to and dephosphorylates metabotropic glutamate receptor 3.
34. Hinsby, A. M., Olsen, J. V., and Mann, M. (2004) Tyrosine phosphopro- Proc. Natl. Acad. Sci. U. S. A. 100, 16006 –16011
teomics of fibroblast growth factor signaling: a role for insulin receptor 43. Levoye, A., Dam, J., Ayoub, M. A., Guillaume, J. L., Couturier, C., Dela-
substrate-4. J. Biol. Chem. 279, 46438 – 46447 grange, P., and Jockers, R. (2006) The orphan GPR50 receptor specifi-
35. Mihindukulasuriya, K. A., Zhou, G., Qin, J., and Tan, T. H. (2004) Protein cally inhibits MT1 melatonin receptor function through heterodimeriza-
phosphatase 4 interacts with and down-regulates insulin receptor sub- tion. EMBO J. 25, 3012–3023
strate 4 following tumor necrosis factor-alpha stimulation. J. Biol. Chem. 44. Reiter, E., and Lefkowitz, R. J. (2006) GRKs and ␤-arrestins: roles in
279, 46588 – 46594 receptor silencing, trafficking and signaling. Trends Endocrinol. Metab.
36. Maillet, M., Robert, S. J., Cacquevel, M., Gastineau, M., Vivien, D., Berto- 17, 159 –165

844 Molecular & Cellular Proteomics 6.5

Вам также может понравиться