You are on page 1of 10

Journal of Immunological Methods 370 (2011) 14–23

Contents lists available at ScienceDirect

Journal of Immunological Methods


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / j i m

Research paper

Novel antigen design for the generation of antibodies to


G-protein-coupled receptors
K. Larsson a, C. Hofström b, C. Lindskog c, M. Hansson d, P. Angelidou d, T. Hökfelt e, M. Uhlén a,
H. Wernérus d, T. Gräslund b, S. Hober a,⁎
a
Division of Proteomics, School of Biotechnology, KTH/AlbaNova University Center, Stockholm, Sweden
b
Division of Molecular Biotechnology, School of Biotechnology, KTH/AlbaNova University Center, Stockholm, Sweden
c
Department of Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
d
Atlas Antibodies, AlbaNova University Center, Stockholm, Sweden
e
Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden

a r t i c l e i n f o a b s t r a c t

Article history: Antibodies are important tools for the study of G-protein-coupled receptors, key proteins in
Received 27 December 2010 cellular signaling. Due to their large hydrophobic membrane spanning regions and often very
Received in revised form 5 May 2011 short loops exposed on the surface of the cells, generation of antibodies able to recognize the
Accepted 5 May 2011
receptors in the endogenous environment has been difficult. Here, we describe an antigen-
Available online 12 May 2011
design method where the extracellular loops and N-terminus are combined to a single antigen for
generation of antibodies specific to three selected GPCRs: NPY5R, B2ARN and GLP1R. The design
Keywords: strategy enabled straightforward antigen production and antibody generation. Binding of the
GPCR
antibodies to intact receptors was analyzed using flow cytometry and immunofluorescence based
Antibody
confocal microscopy on A-431 cells overexpressing the respective GPCR. The antibody–antigen
Antigen design
interactions were characterized using epitope mapping, and the antibodies were applied in
immunohistochemical staining of human tissues. Most of the antibodies showed specific binding
to their respective overexpressing cell line but not to the non-transfected cells, thus indicating
binding to their respective target receptor. The epitope mapping showed that sub-populations
within the purified antibody pool recognized different regions of the antigen. Hence, the genetic
combination of several different epitopes enables efficient generation of specific antibodies with
potential use in several applications for the study of endogenous receptors.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction with a diverse selection of ligands, such as neurotransmitters,


hormones, light, odors and taste (Lundström, 2005).
G-protein-coupled receptors (GPCRs) belong to a protein Due to the regulating activities, GPCRs are very interesting
family characterized by seven transmembrane helices con- as drug targets and currently 60–70% of the drug development
nected by loops, an extracellular N-terminal and an intracellular is focused on GPCRs (Lundström, 2005). However, because of
C-terminal tail (Fig. 1A). This family of cellular receptor proteins difficulties in production and purification of these membrane
is the largest protein family with more than 1000 members proteins (Lundström, 2005; Sarkar et al., 2008), only a handful
(Wess, 1997), regulating cellular function through interactions of solved structures exists in the Protein Data Bank, and thus
information valuable for the drug development process is often
lacking (Berman et al., 2000). In order to further understand
Abbreviations: GPCR, G-protein-coupled receptor; B2AR, beta 2-adrenergic and study GPCRs, genetic fusion to tags has commonly been
receptor; GLP1R, glucagon-like peptide 1 receptor; NPY5R, neuropeptide Y
receptor 5; ABP, albumin binding protein; HPA, Human Protein Atlas.
used. To this end, small tags like His6-, HA- or Flag have been
⁎ Corresponding author. Tel.: + 46 8 5537 8330; fax: +46 8 5537 8481. expressed together with the target receptor (Jongsma et al.,
E-mail address: sophia.hober@biotech.kth.se (S. Hober). 2007). Hence, monoclonal antibodies recognizing the tag can

0022-1759/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.jim.2011.05.001
K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23 15

A bodies were purified using the antigen as affinity ligand and


successfully validated for target specificity in a number of
different applications such as flow cytometry, immunofluores-
cence-based confocal microscopy and immunohistochemistry
using human cell lines and tissues.

2. Materials and methods

2.1. Materials

The oligonucleotides for assembly of the GPCR constructs


were synthesized by Thermo Electron Corporation and Operon.
All primers were purchased from Thermo Fisher Scientific. Full-
length GPCR cDNA clones were acquired from the Mammalian
B Gene Collection via geneservice (glucagon peptide 1 receptor,
His6-ABP
GLP1R, AccNo: BC113493 and beta2-adrenogenic receptor,
B2AR, AccNo: BC063486) and imaGenes (neuropeptide Y
Linker (GSSSG)
receptor, NPY5R, AccNo: BC042416). Commercial antibodies
used were: rabbit anti-B2AR (Abcam, ab13300); mouse anti-
Fig. 1. Schematic representation of an intact G-protein-coupled receptor and GLP1R (R&D systems, MAB28141); rabbit anti-GLP1R (Strategic
outline of the antigen used for generation of GPCR-specific antibodies. (A) A
Diagnostics, GA1940) and rabbit anti-GLP1R (Chemicon,
seven transmembrane domain core is characteristic for all GPCRs. The N-
terminus together with the extracellular loops is responsible for ligand binding
AB9433). For NPY5R, two additional rabbit antibodies produced
and the intracellular loops and C-terminus for intracellular signaling. (B) The by the Human Protein Atlas (HPA) project were used:
extracellular loops and N-terminus were assembled to a single antigen, separated HPA013790 and HPA014056.
by a polar linker (GSSSG) and N-terminally fused to a purification/immunization
tag, His6-ABP.
2.2. Plasmid construction

The extracellular parts of the GPCRs were cloned in series,


separated by a polar five amino acid linker—GSSSG. The GPCR
be utilized to elucidate localization and function of the constructs were generated using solid-phase gene assembly
receptors. Another approach is to express the receptor fused (Nguyen et al., 1994; Ståhl et al., 1993), where a longer DNA
to a fluorescent group, such as green fluorescent protein (GFP) sequence was built on a solid support from shorter oligonucle-
(Castro et al., 2005). In both these methods a genetic alteration otides by parallel ligations. The oligonucleotides (Thermo
is made by adding a protein tag, a procedure that may affect the Electron Corporation and Operon) were 50 bases long with
behavior of the receptor regarding localization, quantity and 10 bases overlap. In brief, 300 μg of streptavidin-coated
activity. Therefore, specific antibodies recognizing the native DynaBeads (DynaBeads M-280 streptavidin (10 mg/ml),
forms of GPCRs are invaluable tools for the study of these cell Dynal Biotech) was used to couple 80 pmol of the first
surface receptors and their function (Michel et al., 2009). biotinylated segment. After incubation for 1 h at RT and
Antibodies are normally produced by immunization of an subsequent washing, 40 pmol each of the remaining segments
animal with the pure protein of interest. Due to the inherent was mixed for 30 min at 65 °C. T4 DNA ligase (Invitrogen) was
problems in the production and purification of GPCRs, the use then added, and the ligation reaction proceeded for 4 h at RT.
of the intact GPCR-proteins as antigen has been difficult. The assembled constructs were amplified using nested PCR
Hence, most antibodies recognizing GPCRs have so far been with AmpliTaq Gold DNA polymerase (Applied Biosystems).
generated using synthetic peptide fragments of the receptor Due to the long N-terminal region of GLP1R, it was PCR
protein as antigens (Gupta and Devi, 2006; Mackrill, 2004). amplified separately and ligated to the rest of the construct
Zhang et al. (2004) further developed this strategy by using a naturally occurring PstI site. The assembled GPCR
synthetically producing cyclic peptides, thought to mimic constructs were NotI/AscI digested and ligated into the
the extracellular loops of the CCR5 receptor, for the selection expression vector pAff8c (Larsson et al., 2000) in frame with a
of single-chain Fv (scFv) fragments. These approaches for hexahistidine (His6) purification tag, as well as an Albumin
production of antigens have given rise to functional anti- binding protein (ABP) derived from streptococcal protein G
bodies and scFv-fragments. used for immunization purposes.
To facilitate the production of antigens and allow the
exposure of optional loop regions, we present an alternative 2.3. Protein production and immunization
method for antigen design of GPCRs. This antigen design
strategy has the potential to be used as a general method for BL21(DE3) cells, containing the expression vector encod-
the generation of GPCR specific antibodies. As a proof of concept, ing the antigen, were grown, harvested and lysated essen-
three different receptors were chosen, and the N-terminal tail tially as formerly described (Agaton et al., 2003), and the
together with the three extracellular loops were genetically lysed cell cultures were IMAC-purified under denaturing
linked together to form a continuous antigen, where the conditions using an automated set-up for affinity chroma-
different segments were separated by polar and flexible tography (Steen et al., 2006). Moreover, the fusion proteins
glycine/serine (GSSSG) containing linkers. The acquired anti- were analyzed with SDS-PAGE (BioRad), and the
16 K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23

concentrations were determined using a BCA™ Protein Assay Phusion High-Fidelity DNA polymerase (Finnzymes) and
Kit (Pierce). The dual affinity handle protein, His6ABP, was furthermore cloned into the lentivector 2K7bsd (Suter et al.,
also expressed and purified from the pAff8c vector as 2006) using Gateway® cloning (Invitrogen). The 2K7bsd-GPCR
previously described (Agaton et al., 2004). The three GPCR vectors were thereafter transfected to HEK293-T cells together
antigens were immunized in two New Zealand white rabbits with packaging and envelope plasmids using a Calcium chloride
each (Young In Frontier, South Korea) in accordance with the transfection procedure, as previously described (Pear et al.,
national guidelines. The primary immunization was per- 1993; Vernet et al., 2009). The produced viral particles were in
formed using 200 μg of antigen in Freund's complete turn used to infect A-431 cells in the presence of Polybrene
adjuvant, followed by three booster injections of 100 μg (8 μg/ml).
each in Freund's incomplete adjuvant with four week in-
tervals. The rabbit serum was collected two weeks after the
2.6. Flow cytometry analysis
final booster injection.
All generated antibodies as well as several commercial
2.4. Antibody purification and characterization
antibodies were analyzed for binding to the full-length
protein using the overexpressing cell lines and non-trans-
Antisera from immunized animals were purified by a two-
fected controls on a flow cytometer. Briefly, the cells were
step immunoaffinity protocol (Agaton et al., 2004). Initially,
trypsinated, washed with PBS and resuspended in PBS
12 ml serum was filtrated and buffered with PBS. Subse-
containing 1% bovine serum albumin (PBSB). 200,000 cells
quently, antibodies specific for the tag protein were depleted
were used for staining with each antibody. Primary anti-
from the sample using vector protein, His6ABP, coupled to
bodies were diluted with the PBS containing BSA to a final
NHS-activated Sepharose™ 4 Fast Flow matrix (Amersham
concentration of 5 μg/ml and secondary antibodies to
Biosciences). The depleted sera samples were additionally
10 μg/ml. The cells were incubated with 70 μl of primary
affinity purified on prepacked NHS-activated one ml HiTrap
antibody for 1 h at RT followed by washing with PBSB. After
columns (GE Healthcare), coupled with the respective antigen,
incubation with 70 μl of secondary antibody (Alexa Fluor 488
using the ÄKTAxpress chromatography system (GE Healthcare).
goat anti-rabbit or goat anti-mouse, Molecular probes) and
After subsequent washing of the columns, antibodies were
subsequent wash with PBSB the cells were analyzed on a FACS
eluted with a low pH buffer (0.2 M glycine, 1 mM EGTA, pH 2.5),
Vantage SE (BD Biosciences). Analyzed samples included: no
followed by a buffer exchange to 1× PBS. Finally, glycerol and
antibodies on normal A-431 cells, secondary antibodies on
NaN3 were added to a final concentration of 50% and 0.02%,
normal A-431 control cells, primary and secondary antibodies
respectively. Binding to respective antigen and lack of cross-
on normal A-431 cells and primary and secondary antibodies
reactive binding to tag sequence and linker region was confirmed
on GPCR overexpressing A-431 cells.
by Western blot. Briefly, the respective antigen and one of the
other non-related antigens were separated on SDS-PAGE
gradient gels (10–20% Criterion, Bio-Rad Laboratories) and 2.7. Immunofluorescence and immunohistochemistry analysis
transferred to PVDF membranes (Criterion Gel™ Blotting
Sandwiches, Bio-Rad Laboratories). Subsequent to blocking of To confirm that the antibodies bind a protein on the cell
the membrane in blocking buffer (5% dry milk, 0.5% Tween20, 1× surface, confocal fluorescence microscopy was utilized, essen-
TBS; 0.1 M Tris–HCl, 0.5 M NaCl), the anti-GPCR antibodies, tially as described previously (Barbe et al., 2008). Briefly, glass
diluted 1/300–1/600, were added and the membrane was bottom plates (Whatman) were coated with fibronectin
incubated for 1 h. Secondary HRP conjugated goat-anti rabbit (Sigma) and seeded with 10,000 cells per well, grown for 5 h
antibody (DakoCytomation), diluted 1/3000, was added and and fixed with ice-cold 4% paraformaldehyde (Sigma). After
following incubation for 1 h, the membranes were soaked in permeabilization with 0.1% Triton X-100 (Sigma) the cells were
substrate solution (Immobilon Western Chemiluminescent HRP incubated with the primary antibody as well as a mouse anti-
Substrate, Millipore Corporation), and binding events were tubulin antibody (Abcam) overnight at 4 °C. Moreover, the cells
visualized in a Chemidoc CCD camera system (Bio-Rad were incubated for 1.5 h with secondary antibodies, goat anti-
Laboratories). rabbit IgG conjugated with Alexa Fluor 488 and rabbit anti-
mouse IgG Alexa Fluor 555 (Molecular Probes) and counter-
2.5. Generation of GPCR-expressing cell lines stained with DAPI, a nuclear probe. Image acquisition was
performed with a LSM 510 Meta confocal laser-scanning
The HEK 293T cell line was obtained from LGC Promochem microscope (Carl Zeiss GmbH). For the immunofluorescence
and the A431 cell line from DSMZ. Cells were cultivated in 5% staining of human brain tissue, unfixed cryostat sections were
CO2 at 37 °C in Dulbecco's modified Eagle's media (DMEM, incubated with antibodies NPY5R:1 and NPY5R:2 against the
Invitrogen) supplemented with 10% fetal bovine serum (FBS) NPY5R (dilution 1/1000) and processed according to the TSA+
and 1% Antibiotic–Antimycotic solution. For selection purposes, method (Adams, 1992) using a commercial kit and with green
the medium of infected cell lines contained blasticidin fluorescein isothiocyanate (FITC) as marker, as described in
(20 μg/ml media). B2AR, GLP1R and NPY5R were overexpressed detail elsewhere (Mulder et al., 2009). Immunohistochemical
in the human epidermoid carcinoma cell line, A-431, using a staining of human pancreatic islets with the GLP1R-specific
lentiviral gene delivery system (Suter et al., 2006). Full-length antibodies (GLP1R:1, GLP1R:2, MAB28141, GA1940 and
cDNA clones of B2AR, GLP1R and NPY5R were purchased from AB9433) was performed using formalin fixed, paraffin embed-
the Mammalian Gene Collection (Genservice and imaGenes, ded tissue samples. The staining was performed according to
GmbH). The gene fragments were amplified with PCR using previously described methods (Nilsson et al., 2005).
K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23 17

2.8. RT-PCR subsequent purification of the polyclonal antibodies resulted


in antibodies able to specifically recognize the desired target
The expression of NPY5R, B2AR and GLP1R in the transduced protein.
cell lines was investigated on mRNA level. Overexpressing cells
and normal A-431 cells were trypsinated, washed with PBS and
frozen as pellets in −80 °C. Messenger RNA was extracted from 3.1. Antigen production and antibody purification
cells using RNeasy Mini Kit (Qiagen) with on-column digestion
of chromosomal DNA. Prior to amplification, the mRNA was To enable simple and efficient protein purification, the
converted to cDNA using Super Script III (Invitrogen). PCR antigen-constructs were cloned in fusion to a hexahistidine
amplification was conducted using Phusion High-Fidelity DNA (His6) tag. Moreover, due to its immunopotentiating effects,
polymerase (Finnzymes) with specific primers. Each GPCR- an albumin binding protein (ABP) was inserted between the
specific primer pair was used for amplification of both normal His6-tag and the antigen (Sjölander et al., 1997). The
A-431 cells and the respective overexpressing cell line. generated constructs were expressed in Escherichia coli and
Amplification of all cDNA fragments was also conducted purified by IMAC utilizing the His6-tag. Purified proteins were
utilizing β-actin-specific primers, generating a PCR product of used for immunization of two rabbits, giving two polyclonal
70 bp, for normalization. antisera for each receptor. To purify antibodies targeting the
three receptors included in this study and thereby achieving
2.9. Epitope mapping of GPCR antigens specific antibodies, a stringent two-step affinity purification
method was applied, where the antigen was used as affinity
Peptides covering the extracellular loops and N-terminus ligand (Nilsson et al., 2005). Antibody yield varied from 0.2 mg
were designed for all GPCR antigens. The total 25 biotinylated to 1.0 mg with an average of 0.5 mg of pure antibody from
peptides (PEPscreen library, Sigma-Aldrich) were between 7 12 ml of antiserum. Binding to the antigen and lack of cross-
and 20 amino acids long depending on loop/N-terminal size. reactive binding to the tag sequence and linker regions was
For longer loops overlapping peptides were designed, except confirmed by Western blot analysis (data not shown). The
for the N-terminus of GLP1R, which was kept intact and antibodies will further on be referred to as NPY5R:1 and
expressed as a 118 amino acid long peptide. All peptides were NPY5R:2 for the antibodies generated toward the neuropeptide
resolved in 80% DMSO and diluted in BRE (Blocking reagent Y receptor 5; B2AR:1 and B2AR:2 for the beta 2 adrenergic
for ELISA, Roche) to a final concentration of 20 μM. Coupling receptor antibodies and GLP1R:1 and GLP1R:2 for the glucagon-
of the Luminex beads was essentially done as previously like peptide 1 receptor antibodies. All antibodies used, both
described (Larsson et al., 2009). Neutravidin (Pierce) diluted those produced in this study and acquired commercially, are
to 0.1 μg/μl in MES was immobilized on 106 carboxylated beads listed in Table 1.
(COOH Microspheres, Luminex-Corp.) per color-coded ID,
according to the manufacturer's protocol. To couple the
biotinylated peptides about 5 × 10 5 neutravidin-coupled 3.2. Cell line generation and flow cytometric analysis
beads per ID were used. In total 100 μl of 20 μM diluted peptide
was utilized in the coupling reaction and following incubation In order to analyze if the GPCR-targeting antibodies were
for 1 h and subsequent washing steps, the Luminex beads were able to recognize the full-length receptors as expressed on the
stored in azide containing BRE buffer. Prior to analysis of the surface, three cell lines overexpressing NPY5R, B2AR and
antibodies, a bead stock was made containing 10 μl of each GLP1R, respectively, were constructed. RT-PCR was performed
peptide-coupled bead ID. All antibodies were diluted to 50 ng/ on the respective cell lines and all three cell lines showed a high
ml of which 45 μl was added to 5 μl of the bead stock, incubated level of receptor specific transcripts (Fig. 2A). Subsequently,
for 1 h and washed in PBST. Following incubation with a several antibodies were used for analysis of the surface exposed
secondary R-Phycoerythrine labeled anti-rabbit IgG (0.5 μg/ml, GPCRs with flow cytometry to assess differences between
Jackson ImmunoResearch) and additional washing, binding differently produced antibodies. The resulting histograms show
events were measured using a Luminex LX200 instrument with that all NPY5R antibodies (NPY5R:1, NPY5R:2, HPA013790 and
xPONENT software. For each experiment 50 events per bead ID HPA014056) bind to the A-431 cells overexpressing NPY5R
were counted and the median fluorescence intensity (MFI) was (A-431NPY5R) but not to non-transfected A-431 cells (A-431)
used. Each experiment was performed in triplicates. (Fig. 2B). For B2AR only one antibody, B2AR:1, generated a shift
in fluorescence, hence indicating binding to the receptor.
3. Results B2AR:2 and ab13300 showed no elevated fluorescence signals
when analyzing the overexpressing A-431 cells (A-431B2AR)
In this study, we have developed a novel strategy for (Fig. 2C). All antibodies generated toward GLP1R (GLP1R:1 and
antigen design to generate specific antibodies against GPCRs. GLP1R:2) as well as the commercial antibodies resulted in
Three GPCRs were selected for this study: the neuropeptide Y binding to the cells overexpressing GLP1R (A-431GLP1R). There
receptor 5 (NPY5R), the beta 2 adrenergic receptor (B2AR) was no binding of the antibodies to A-431 cells except for the
and the glucagon-like peptide 1 receptor (GLP1R). By mouse antibody MAB28141 that displayed a slight shift in
combining the N-terminal tail with the three extracellular fluorescence (Fig. 2D). In general, the flow cytometry analyses
loops to a single antigen (Fig. 1, Supplementary) an easily gave rise to broad peaks, indicating different levels of receptor
produced and coherent antigen only displaying the surface expression. Depending on genomic position of the integration
accessible parts of the GPCR receptor was created. Immuni- of the gene encoding the GPCR, expression of the transgene
zation of two rabbits with each of the acquired antigens and may vary due to chromatin packing.
18 K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23

Table 1 Moreover, when staining human pancreatic tissues, known to


Antibodies used in this study are listed together with their corresponding express GLP1R (Thorens, 1992) using GLP1R:1, distinct
receptors.
staining of the plasma membrane was observed in pancreatic
GPCR Antibody islets. Antibody GLP1R:2 also showed a membranous staining
pattern, although accompanied with an additional cytoplas-
NPY5R NPY5R:1
NPY5R:2 mic signal of the surrounding exocrine glandular cells. The
HPA013790 other antibodies used (GA1940 and AB9433) both gave rise to
HPA014056 a more unspecific staining pattern (Fig. 4B).
B2AR B2AR:1
B2AR:2
ab13300 3.4. Epitope mapping
GLP1R GLP1R:1
GLP1R:2 In order to characterize the binding of the different
AB9433 antibodies to the GPCRs, peptides covering the primary sequence
GA1940
of the antigens were synthesized. The peptides corresponded to
MAB28141
the N-terminus and extracellular loops of the antigens respec-
tively (see Supplementary data for sequences). The biotinylated
peptides differed in length between 7 and 20 amino acids,
depending on the length of the loops and N-terminus. When
3.3. Immunohistochemical and immunofluorescence analyses loops exceeded 20 amino acids, the sequences were covered
using overlapping peptides, except for the N-terminus of GLP1R,
To further validate binding to the full-length protein and which was expressed as a continuous 118 amino acid long
assure specificity, i.e. no unspecific staining in other cell peptide. The peptides were immobilized on streptavidin-coated
compartments besides the membrane or membrane-associated beads via an N-terminal biotin and the analyses were performed
compartments (e.g. Golgi), immunofluorescence analysis with with a Luminex instrument. All antibodies showed different
confocal microscopy was performed. Included in the study were patterns of recognition, regarding which part of the antigen they
the GPCR overexpressing A-431 cell lines (A-431NPY5R, A-431B2AR interacted with. Even though a majority of the response was
and A-431GLP1R) as well as non-transfected A-431 cells. directed to the N-terminus, possibly due to the longer amino
All antibodies directed toward NPY5R (NPY5R:1, NPY5R:2, acid sequences, this was not always the case.
HPA013790 and HPA014056) showed staining of the plasma Antibody NPY5R:1 bound to all segments of the antigen but
membrane, in general more pronounced in the cell junctions. with very low signal intensities, while NPY5R:2 generated a peak
Additional Golgi-like staining was seen as exemplified in Fig. 3A with high intensity for the first peptide of the N-terminus
(NPY5R:2). In the control samples (A-431 cells), only very faint indicating an epitope (Fig. 5A). B2AR:1 recognized the N-
staining could be observed (Fig. 3B), except for antibody terminus and the second extracellular loop, while B2AR:2
NPY5R:1 which generated a nuclear staining pattern, not bound both to the second and third extracellular loop as well
detectable in A-431NPY5R cells (data not shown). The B2AR:1 as to the N-terminus (Fig. 5B). The third extracellular loop of
antibody exhibited binding to the plasma membrane (Fig. 3C). B2AR is short, only 7 amino acids, therefore an extra peptide was
Additional staining, likely in the Golgi apparatus, was detected inserted where the 7 amino acids were placed two times in a
but less pronounced than for NPY5R antibodies. No significant row, head-to-tail, in order to extend the distance of the possible
staining of B2AR:1 was detected in A-431 cells (Fig. 3D). epitope from the bead surface. The antibody B2AR:2 showed
Staining with B2AR:2 resulted in a fluorescent signal in the binding to this ‘double’ peptide but no binding to the original
membrane but compared to A-431 cells, no elevated levels one. Binding of GLP1R:1 was only detected toward the N-
were detected (data not shown). The commercial ab13300 was terminus, while GLP1R:2 showed binding to the extracellular
completely negative both in A-431B2AR cells and A-431 cells loop 3, as well as the N-terminus (Fig. 5C).
(data not shown). The antibodies expected to bind to GLP1R
were also used for confocal microscopy. In these cases no 4. Discussion
certain membrane staining could be established (Fig. 3E). On
the other hand MAB28141 stained the plasma membrane. Here we describe a novel method for design of GPCR antigens
However, none of these antibodies gave staining in non- and the subsequent generation of antigen-purified antibodies
transfected A-431 cells (Fig. 3F), which indicates binding to the with successful outcome. This was accomplished by merging the
overexpressed receptor. Antibody GA1940 gave staining in the N-terminus and extracellular loops into a single antigen, thus
plasma membrane, but also displayed staining in the nucleus of facilitating the production compared to full-length GPCR
A-431 cells, a pattern that was not detected in the A-431GLP1R production, often requiring time-consuming optimization of
cell line (data not shown). No staining could be observed for expression parameters (Sarkar et al., 2008). By using this
antibody AB9433, neither in A-431GLP1R cells nor in A-431 cells approach it can be decided beforehand whether an antibody
(data not shown). should bind to the extracellular or intracellular regions of the
To further assess the specificity of the produced anti- receptor. Moreover, the immunization with a longer antigen
bodies, tissue sections from parts of the human brain possibly including several potential epitopes increases the probability of
expressing NPY5R were analyzed by immunofluorescence success in a broader repertoire of applications compared to
microscopy. No detectable staining was obtained with the peptide immunizations previously used for the generation of
NPY5R:1 antibody, but as can be seen in Fig. 4A, the NPY5R:2 GPCR specific antibodies. In fact, data from the Human Protein
antibody gave rise to a large number of fluorescent processes. Atlas program (www.proteinatlas.org), where antibodies are
K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23 19

A
B2AR 1002 bp

NPY5R 781 bp

GLP1R 848 bp
+ -
β-actin 70 bp

31
AR

1R
5R

A4
B2

LP
PY

G
N

B NPY5R:1 NPY5R:2 HPA013790 HPA014056

100

0
1 2 3 4 1 2 3 4 1 2 3 4 1 2 3 4
10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10

C B2AR:1 B2AR:2 ab13300

120
Cell count

0
1 2 3 4 1 2 3 4 1 2 3 4
10 10 10 10 10 10 10 10 10 10 10 10

D GLP1R:1 GLP1R:2 AB9433

100

0
1 2 3 4 1 2 3 4 1 2 3 4
10 10 10 10 10 10 10 10 10 10 10 10
GA1940 MAB28141

100
Secondary antibody + A-431 cells
Primary/secondary antibody + A-431 cells
Primary/secondary antibody + over expressing A-431 cells

0
1 2 3 4 1 2 3 4
10 10 10 10 10 10 10 10

Fluorescence intensity

Fig. 2. (A) Detection of GPCR mRNA in the cell lines was performed by RT-PCR of extracted mRNA from overexpressing (+) as well as non-transfected A-431 cells
(−) using specific primers from NPY5R, B2AR and GLP1R respectively. Primers specific for the housekeeping gene β-actin were used to verify equal amount of
sample loading. (B) Flow cytometric analysis of antibodies specific for NPY5R, (C) B2AR and (D) GLP1R. Each histogram shows data obtained from three samples:
non-transfected A-431 cells incubated solely with secondary antibody (thin lines), non-transfected A-431 cells incubated with both primary and secondary
antibodies (dashed lines) and over expressing cells immunostained with the corresponding primary antibody and secondary antibody (thick lines).
20 K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23

A B

C D

E F

Fig. 3. Representative immunofluorescent staining of GPCR-overexpressing as well as non-transfected A-431 cells. Antibody NPY5R:2 staining of (A) NPY5R
overexpressing A-431 cells and (B) A-431 cells, B2AR:1 staining of (C) B2AR overexpressing cells and (D) A-431 cells and finally GLP1R:2 staining of (E) GLP1R
overexpressing cells and (F) A-431 cells. The cells were counterstained with the nuclear probe DAPI (blue). Antibody binding is depicted in green. Bar indicates
10 μm.

produced toward the non-redundant set of human proteins, Validation of antibodies in general, and GPCR-specific
suggest that immunization with antigens shorter than 50 amino antibodies in particular, is of great importance and seldom an
acids generate significantly lower antibody titers as well as easy task. The specificity of antibodies targeting GPCRs is a
lower success rate in subsequent applications (unpublished well-known issue and has recently been discussed by Michel
data). We chose to assemble the antigen using the extracellular et al.(2009) Since most GPCRs are endogenously expressed at
parts of the GPCRs in order to detect the receptor protein on very low levels (Sarkar et al., 2008) validation of them is
intact cells, thus allowing research on endogenous receptor in its difficult, and therefore we generated three different cell lines,
native membrane bound form on living cells, but the same design overexpressing NPY5R, B2AR and GLP1R respectively. Over-
strategy could also be envisioned for the intracellular parts. expression of the specific G-protein-coupled receptors was
K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23 21

Fig. 4. Immunofluorescence staining of human brain tissue and immunohistochemical staining of human pancreas. (A) Immunofluorescence micrograph taken from the
dorsal part of the periaqueductal gray in the human mesencephalon after incubation of an unfixed cryostat section with NPY5R:2 antibody. A large number of
fluorescent processes are seen, but no cell bodies. Bar indicates 1 μm. (B) Immunohisotochemical staining of human formalin fixed paraffin embedded pancreatic tissues
using GLP1R antibodies. GLP1R:1 gives a clear staining of plasma membranes (brown) in the pancreatic islets with no staining observed in exocrine glandular cells. No
specific immunohistochemical staining pattern could be detected after incubation with GA1940. The nuclei of the cells are stained with hematoxylin (blue). Bar indicates
100 μm.

successfully examined by RT-PCR, showing high expression in cell line (data not shown), indicating unspecific binding in the
the transfected cells and no detectable expression in the non- absence of the intended target protein. The antibodies
transfected cells. This permitted validation of the obtained generated against NPY5R and GLP1R were also used for
antibodies with flow cytometry and immunofluorescence- immunohistochemical and immunofluorescent staining of
based confocal microscopy. According to the flow cytometric human tissues expected to express the target proteins. In
analysis, most of the antibodies showed specific binding, i.e. both cases the antibodies showed staining that also supports
increased fluorescence intensity, to their respective GPCR- the earlier acquired validation data, indicating correct and
overexpressing cell line but not to non-transfected control selective binding of the antibodies.
cells. However, when using B2AR:2 and ab13300 no increased The epitope mapping showed that sub-populations of
fluorescence signals could be detected. This could possibly be antibodies within the purified antibody pools recognized
explained by low affinity of the antibodies to their target different regions of the respective antigen. Hence, there is a
receptor or that epitopes are hidden or conformational large potential in this novel strategy. Through the genetic fusion
different on the native receptor compared to the antigen. of many different epitopes, and by avoiding family specific
Immunofluorescence staining and analysis by confocal mi- regions, specific antibodies can potentially be raised with high
croscopy of overexpressing cells showed that many of the success rate. For large-scale production of antibodies toward
used antibodies gave staining of the plasma membrane, hence GPCRs using synthetic peptides, B-cell epitope prediction would
indicating binding to the membrane-bound target protein. be a requirement in order to reduce the number of experiments
Moreover, no or very weak staining could be detected in non- needed to achieve a good antibody. However, prediction of B-cell
transfected A-431 cells. The antibodies NPY5R:1 and GA1940 epitopes has recently been demonstrated to be very difficult
showed nuclear staining in non-transfected A-431 cells that (Blythe and Flower, 2005). The results from the epitope mapping
could not be observed in the corresponding overexpressing showed that a majority of the response was directed to the N-
22 K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23

Fig. 5. Epitope mapping of GPCR antigens using synthetic peptides and Luminex technology. Intensity plots showing binding of (A) NPY5R:1 and NPY5R:2, (B)
B2AR:1 and B2AR:2 and (C) GLP1R:1 and GLP1R:2 to the N-terminus and extracellular loops of the respective antigen. Intensity is measured as median fluorescent
intensity (MFI). For sequences longer than 20 amino acids, overlapping peptides were used. The N-terminal of the GLP1R was represented as a 118 amino acids
long peptide.

terminus, probably due to the longer amino acid sequences, technical assistance. C. Stadler is acknowledged for assistance
especially for GLP1R. However, the B2AR:2 antibody almost with the confocal microscopy imaging, Dr. David Suter for
exclusively recognized the second and third extracellular loop. providing the 2K7-plasmid and Dr. Erwan LeMaitre for taking
Therefore our method to include all or selected extracellular parts immunofluorescence micrographs. The Knut and Alice Wal-
of the receptor is a promising approach for GPCR-specific lenberg Foundation financially supported this work. The
antibody generation. Since there are some inherited limitations support of the Swedish Research Council (04X-2888) is
with polyclonal antibodies our strategy to use these combined gratefully acknowledged.
antigens could also be envisioned for production of monoclonal
antibodies. The antigen could then be used for immunization, in
References
order to have a broader range of B-cells to choose from when
producing the hybridoma cells. Adams, J.C., 1992. Biotin amplification of biotin and horseradish peroxidase signals
Supplementary materials related to this article can be in histochemical stains. J. Histochem. Cytochem. 40 (10), 1457.
found online at doi:10.1016/j.jim.2011.05.001 Agaton, C., et al., 2003. Affinity proteomics for systematic protein profiling of
chromosome 21 gene products in human tissues. Mol. Cell. Proteomics 2
(6), 405.
Acknowledgements Agaton, C., et al., 2004. Selective enrichment of monospecific polyclonal antibodies
for antibody-based proteomics efforts. J. Chromatogr. A 1043 (1), 33.
Barbe, L., et al., 2008. Toward a confocal subcellular atlas of the human
The authors would like to thank Dr. J. Galli, Dr. J. Feldwisch proteome. Mol. Cell. Proteomics 7 (3), 499.
and K. Johansson (Affibody AB) for fruitful discussions and Berman, H.M., et al., 2000. The Protein Data Bank. Nucleic Acids Res. 28 (1), 235.
K. Larsson et al. / Journal of Immunological Methods 370 (2011) 14–23 23

Blythe, M.J., Flower, D.R., 2005. Benchmarking B cell epitope prediction: Pear, W.S., et al., 1993. Production of high-titer helper-free retroviruses by
underperformance of existing methods. Protein Sci. 14 (1), 246. transient transfection. Proc. Natl. Acad. Sci. U. S. A. 90 (18), 8392.
Castro, M., et al., 2005. Turn-on switch in parathyroid hormone receptor by a Sarkar, C.A., et al., 2008. Directed evolution of a G protein-coupled receptor
two-step parathyroid hormone binding mechanism. Proc. Natl. Acad. Sci. for expression, stability, and binding selectivity. Proc. Natl. Acad. Sci. U. S. A.
U. S. A. 102 (44), 16084. 105 (39), 14808.
Gupta, A., Devi, L.A., 2006. The use of receptor-specific antibodies to study G- Sjölander, A., et al., 1997. The serum albumin-binding region of streptococcal
protein-coupled receptors. Mt. Sinai J. Med. 73 (4), 673. protein G: a bacterial fusion partner with carrier-related properties.
Jongsma, M., et al., 2007. Validation of a rapid, non-radioactive method to J. Immunol. Methods 201 (1), 115.
quantify internalisation of G-protein coupled receptors. Naunyn- Ståhl, S., et al., 1993. Solid-phase gene assembly of constructs derived from
Schmiedebergs Arch. Pharmacol. 375 (5), 329. the Plasmodium falciparum malaria blood-stage antigen Ag332. Bio-
Larsson, M., et al., 2000. High-throughput protein expression of cDNA techniques 14 (3), 424.
products as a tool in functional genomics. J. Biotechnol. 80 (2), 143. Steen, J., et al., 2006. High-throughput protein purification using an
Larsson, K., et al., 2009. Characterization of PrEST-based antibodies towards automated set-up for high-yield affinity chromatography. Protein Expr.
human Cytokeratin-17. J. Immunol. Methods 342 (1–2), 20. Purif. 46 (2), 173.
Lundström, K., 2005. Structural genomics of GPCRs. Trends Biotechnol. 23 (2), 103. Suter, D.M., et al., 2006. Rapid generation of stable transgenic embryonic
Mackrill, J.J., 2004. Generation, use, and validation of receptor-selective stem cell lines using modular lentivectors. Stem Cells 24 (3), 615.
antibodies. Methods Mol. Biol. 259, 47. Thorens, B., 1992. Expression cloning of the pancreatic beta cell receptor for
Michel, M.C., Wieland, T., Tsujimoto, G., 2009. How reliable are G-protein- the gluco-incretin hormone glucagon-like peptide 1. Proc. Natl. Acad. Sci.
coupled receptor antibodies? Naunyn-Schmiedebergs Arch. Pharmacol. U. S. A. 89 (18), 8641.
379 (4), 385. Vernet, E., et al., 2009. Affibody-mediated retention of the epidermal growth
Mulder, J., et al., 2009. Tissue profiling of the mammalian central nervous system factor receptor in the secretory compartments leads to inhibition of
using human antibody-based proteomics. Mol. Cell. Proteomics 8 (7), 1612. phosphorylation in the kinase domain. N. Biotechnol.
Nguyen, T.N., Uhlén, M., Ståhl, S., 1994. De novo gene assembly using a Wess, J., 1997. G-protein-coupled receptors: molecular mechanisms involved
paramagnetic solid support. In: Uhlén, M., Hornes, E., Olsvik, Ö. (Eds.), in receptor activation and selectivity of G-protein recognition. FASEB J.
Advances in Biomagnetic Separation. Eaton Publishing Co., Natick, p. 73. 11 (5), 346.
Nilsson, P., et al., 2005. Towards a human proteome atlas: high-throughput Zhang, Y., et al., 2004. Selection of active ScFv to G-protein-coupled receptor
generation of mono-specific antibodies for tissue profiling. Proteomics 5 CCR5 using surface antigen-mimicking peptides. Biochemistry 43 (39),
(17), 4327. 12575.

You might also like