Identification of Binding Domains of the Growth Hormone-Releasing Hormone Receptor by Analysis of Mutant and Chimeric Receptor Proteins
Venita I. DeAlmeida and
Kelly E. Mayo
Department of Biochemistry, Molecular Biology and Cell Biology
Northwestern University Evanston, Illinois 60208
 |
ABSTRACT
|
---|
The hypothalamic peptide GH-releasing hormone
(GHRH) stimulates the release of GH from the pituitary through binding
and activation of the GHRH receptor, which belongs to the family of G
protein-coupled receptors. The objective of this study was to identify
regions of the receptor critical for interaction with the ligand by
expressing and analyzing truncated and chimeric epitope-tagged GHRH
receptors. Two truncated receptors, GHRH
N, in which part of the
N-terminal domain between the putative signal sequence and the first
transmembrane domain was deleted, and GHRH
C, which was truncated
downstream of the first intracellular loop, were generated. Both the
receptors were deficient in ligand binding, indicating that neither the
N-terminal extracellular domain (N terminus) nor the membrane-spanning
domains with the associated extracellular loops (C terminus) are alone
sufficient for interaction with GHRH. In subsequent studies, chimeric
proteins between the receptors for GHRH and vasoactive intestinal
peptide (VIP) or secretin were generated, using the predicted start of
the first transmembrane domain as the junction for the exchange of the
N terminus between receptors. The chimeras having the N terminus of the
GHRH receptor and the C terminus of either the VIP or secretin receptor
(GNVC and
GNSC) did not bind GHRH
or activate adenylate cyclase after GHRH treatment. The reciprocal
chimeras having the N terminus of either the VIP or secretin receptors
and the C terminus of the GHRH receptor
(VNGC and
SNGC) bound GHRH and
stimulated cAMP accumulation after GHRH treatment. These results
suggest that although the N-terminal extracellular domain is essential
for ligand binding, the transmembrane domains and associated
extracellular loop regions of the GHRH receptor provide critical
information necessary for specific interaction with GHRH.
(Molecular Endocrinology 12: 750765, 1998)
 |
INTRODUCTION
|
---|
GH-releasing hormone (GHRH) is a peptide hormone secreted from the
hypothalamus that stimulates the proliferation of pituitary
somatotrophs and induces the synthesis and secretion of GH by these
cells. High-affinity binding of GHRH to its receptors on pituitary
somatotrophs results in G protein coupling, adenylate cyclase
activation and cAMP production, Ca2+ influx, increased
expression of the GH gene, and enhanced GH secretion (1, 2). The GHRH
receptors of several species have been cloned (3, 4, 5, 6), and inactivating
mutations in the receptor have been found in heritable GH deficiency
diseases in both mice (7, 8) and humans (9, 10). Additionally,
transcripts for alternatively spliced forms of the GHRH receptor
truncated in the third intracellular loop have been found in patients
with GH-producing pituitary tumors, although a role for these mutant
receptors in tumorigenesis has not been established (11, 12).
The GHRH receptor is a G protein-coupled receptor (GPCR) with seven
potential membrane-spanning domains and belongs to family B, group III
(B-III) of the GPCR superfamily (13). It is highly homologous to the
receptors for secretin (SEC) (14), vasoactive intestinal peptide (VIP)
(15), pituitary adenylate cyclase-activating peptide (PACAP) (16),
glucagon (17), glucagon-like peptide-1 (GLP-1) (18), and gastric
inhibitory peptide (GIP) (19), which are also grouped in family B-III
(13). These receptors are related to the receptors for PTH (20),
PTH-related peptide (21), CRH (22), and calcitonin (23), which have a
longer N-terminal extracellular domain but share many common amino
acids and a similar overall structure to the receptors of family
B-III (24).
The receptors of family B-III have several conserved residues in the
N-terminal extracellular domain, including six cysteines, an aspartate,
a tryptophan, and a glycine at positions corresponding to amino acids
60, 65, and 100 in the GHRH receptor (24). Mutation of the conserved
cysteines in the VIP receptor (25), the aspartate in the GHRH (7, 8, 26), VIP (27), and glucagon (28) receptors, and the tryptophan or
glycine in the VIP receptor (27) results in a loss of hormone binding
and hormone-stimulated cAMP accumulation in cells expressing these
mutant receptors. The N-terminal domains of the receptors of this
family are predicted to have a similar structure, presumably maintained
by disulfide bonds between the conserved cysteines, and are thought to
be involved in ligand binding (29). An
-helical region within this
N-terminal domain is hypothesized to be involved in a coiled-coil
interaction with an
-helical region in the respective hormone (30),
again suggesting a role for this domain in ligand binding.
The superfamily of GPCRs bind to, and are activated by, a diverse group
of ligands ranging from ions and small peptides to large glycoproteins,
and different receptors exhibit considerable variation in the
structural determinants of ligand recognition (31, 32, 33, 34, 35, 36). Ligand binding
by several receptors in family B has been studied using mutagenesis and
chimeric receptor approaches. These studies reveal that the large
N-terminal extracellular domains of the GLP-1 (37), VIP (38, 39), and
PACAP (40) receptors are sufficient for low levels of specific ligand
binding. However, other studies indicate that the presence of one or
more extracellular loop regions and residues within the transmembrane
domains is required for high-affinity ligand binding by the receptors
for secretin, VIP, glucagon, calcitonin, PTH, GIP, and CRH (38, 39, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52), indicating that family B receptors are likely to exhibit
multiple determinants of ligand interaction.
Other than the known importance of the conserved aspartate at position
60, which is mutated in the little mouse, there is no
information about the role of specific domains or residues of the GHRH
receptor in ligand binding and signaling. As an initial step toward
understanding the molecular mechanism of agonist-dependent activation
of the GHRH receptor, we designed experiments to identify domains of
the GHRH receptor required for high-affinity GHRH binding. We generated
two truncation mutants of the human GHRH receptor and four chimeras
between the human GHRH receptor and the receptors for VIP or secretin
and studied their ligand-binding and signaling properties.
 |
RESULTS
|
---|
Generation and Analysis of Epitope-Tagged GHRH, VIP, and Secretin
Receptors
The analysis of mutant and chimeric receptors requires
determination of the levels of receptor protein expressed and
confirmation that the receptor is appropriately localized to the cell
membrane. The amino acids encoding an influenza virus hemagglutinin
(HA) epitope (YPYDVPDYA) were therefore introduced at the C-terminal
ends of the wild-type GHRH, VIP, and secretin receptors, allowing
detection of receptor protein using a monoclonal antibody against the
epitope. In addition, the amino acids encoding the FLAG epitope
(DYKDADDDK) were introduced in the N terminus of the wild-type GHRH
receptor, downstream of the predicted signal peptide. A schematic
representation of the GHRH receptor depicting the location of these
epitope tags, as well as the boundaries of truncations and chimera
junctions to be discussed subsequently, is shown in Fig. 1
.

View larger version (43K):
[in this window]
[in a new window]
|
Figure 1. Structure of the GHRH Receptor Showing the Location
of Epitope Tags and Endpoints of the Truncated and Chimeric Mutant
Receptors
The seven membrane-spanning domains are shown as
cylinders crossing the lipid bilayer. The gray
shaded circles represent the putative signal sequence, and the
black shaded circles represent amino acids that are
conserved in the related receptors for the hormones secretin, glucagon,
GLP-1, GIP, VIP, and PACAP. The ( ) represents the putative
signal-peptide cleavage site, the ( ) represents the consensus site
for N-linked glycosylation, the ( ) represents
aspartate residue 60, which is mutated in the little
mouse, and the ( ) represents the cysteines that are conserved within
family B-III receptors. The location of the AflII and
KpnI sites used to make truncations and fusions are
indicated by arrows. The location and sequences of the
FLAG and HA epitope-tags are also shown.
|
|
To ascertain whether the epitope tags affected receptor function,
expression constructs for the wild-type and epitope-tagged receptors
were transiently transfected into HeLa T4 cells, and the binding and
signaling properties of the receptors were compared. Binding of GHRH to
membranes of cells expressing the wild-type (GHRHR.wt),
HA-tagged (GHRHR.HA), and FLAG-tagged (GHRHR.F) GHRH receptors was
measured in competition assays (Fig. 2A
),
and it was observed that the membranes of cells expressing the
wild-type and HA-tagged GHRH receptors showed a similar dose-dependent
competition of GHRH binding, with ED50 values for
competition of 5.2 and 3.2 nM, respectively. In contrast,
membranes of cells expressing the FLAG-tagged GHRH receptor construct
did not bind GHRH. From Scatchard analysis of saturation-binding data,
the apparent dissociation constants (KD) for GHRH binding
by the wild-type and HA-tagged GHRH receptors were determined to be 36
and 31.6 pM, respectively, with maximal binding
(BMAX) values of 50.9 and 34.2 pmol/mg protein (data not
shown). In response to GHRH stimulation, cells expressing the wild-type
and HA-tagged forms of the GHRH receptor showed a similar
dose-dependent accumulation of cAMP (Fig. 2B
) with ED50
values of 5.7 and 4.4 nM, respectively. Cells expressing
the FLAG-tagged GHRH receptor did not accumulate cAMP, consistent with
the inability of this receptor to bind GHRH. Receptor constructs with
the FLAG epitope tag were therefore used only for comparing the
cellular localization of various receptor proteins in intact cells. In
studies not shown, the presence of the C-terminal HA epitope tag was
found not to affect the properties of the epitope-tagged VIP (VIPR.HA)
and secretin (SECR.HA) receptors compared with their wild-type
counterparts.

View larger version (26K):
[in this window]
[in a new window]
|
Figure 2. Binding and Signaling Properties of Epitope-Tagged
GHRH Receptors
A, Dose-dependent competition of GHRH binding to the wild-type and
epitope-tagged GHRH receptors expressed in HeLa T4 cells. The relative
amount of input radioligand bound in the presence of increasing
concentrations of competitor is shown for each of the receptor
constructs. The amount of GHRH bound in the absence of competitor
corresponds to 8.2 pmol/tube. B, Dose-dependent cAMP accumulation after
GHRH treatment of HeLa T4 cells transfected with wild-type and
epitope-tagged GHRH receptors. The relative amount of intracellular
cAMP produced in response to increasing concentrations of hormone is
shown for each of the receptor constructs. The amount of intracellular
cAMP accumulated in cells treated with 1 µM GHRH
corresponds to 52 pmol/well. Data points represent the mean of
duplicate samples with the range of values indicated by the
error bars; each panel is representative of at least two
independent experiments.
|
|
Expression of Truncated GHRH Receptors
For several GPCRs, it has been possible to broadly localize
hormone-binding domains by expression and analysis of deletion
constructs generated by the removal of large regions of the receptor
protein. To determine whether either the N terminus or the C terminus
of the GHRH receptor could bind ligand in the absence of the other
domain, two truncation mutants were generated and transiently expressed
in HeLa T4 cells. The amino acids between the signal peptide and the
first transmembrane domain were deleted in the receptor GHRH
N by
removing sequences between the two AflII sites, and the
amino acids downstream of the first intracellular loop were deleted in
the receptor GHRH
C by removing sequences between the two
KpnI sites (Fig. 1
). Immunoprecipitation of the
metabolically labeled HA-tagged receptor proteins (Fig. 3A
) shows that the two truncated
receptors, GHRH
N and GHRH
C, were expressed at or above levels of
the full-length receptor. The apparent molecular masses were
approximately 40 kDa and 20 kDa, respectively; however, the
immunoprecipitated protein from cells expressing GHRH
N showed the
presence of two products, differing in size by 4 kDa. The cellular
localization of the full-length and truncated epitope-tagged receptors
was determined by immunofluorescence analysis using confocal microscopy
with intact (for the FLAG-tagged receptors) or permeabilized (for the
FLAG- and HA-tagged receptors) cells (Fig. 3B
). The full-length (GHRHR)
and C-terminally truncated (GHRH
C) receptors were localized
similarly; receptor protein was distributed both on the cell surface
and intracellularly, with the latter presumably representing receptor
protein being transported to, or internalized from, the cell surface.
For these two receptors, similar localization patterns were observed
using antibodies against either the N-terminal FLAG tag or the
C-terminal HA tag in permeabilized cells. In contrast, cells expressing
the N-terminally truncated receptor (GHRH
N) appeared to accumulate
protein preferentially in intracellular membranes, with a smaller
proportion of the total receptor expressed on the cell surface. A
perinuclear localization of this receptor protein was observed using
both the FLAG and HA antibodies in permeabilized cells (Fig. 3B
).

View larger version (88K):
[in this window]
[in a new window]
|
Figure 3. Expression of Truncated GHRH Receptors
A, Immunoprecipitation of the HA-tagged wild-type and truncated human
GHRH receptors. Equivalent amounts of metabolically labeled protein
from HeLa T4 cells transfected with the indicated receptor constructs
were immunoprecipitated using the monoclonal antibody 12CA5 against the
HA-epitope and separated by SDS-PAGE on a 12% gel. The sizes of the
mol wt standards included on the gel are shown on the
left. B, Immunofluorescence localization of
epitope-tagged wild-type and truncated human GHRH receptors. Indirect
immunofluorescence of HeLa T4 cells transfected with pcDNA-3 or the
indicated receptor constructs was performed using the anti-M2
monoclonal antibody against the FLAG-epitope (top and middle
rows) or the 12CA5 antibody against the HA-epitope
(bottom row). The panels in the top row
are images of intact cells while those in the middle and
bottom rows are images of cells permeabilized with 0.1%
saponin. All slides were scanned using a confocal microscope under the
same magnification and contrast settings for equivalent times, and the
panels shown are representative of at least 20 fields observed in two
independent experiments.
|
|
Analysis of Truncated GHRH Receptors
Binding of GHRH to membranes of HeLa T4 cells expressing the
HA-tagged full-length or truncated forms of the GHRH receptor was
measured in competition assays (Fig. 4A
).
The truncated receptors GHRH
N and GHRH
C did not bind GHRH when
expressed by themselves or when coexpressed. In addition, neither
truncated receptor decreased the ligand-binding ability of the
full-length GHRH receptor when coexpressed with it. GHRH treatment did
not stimulate cAMP accumulation in cells expressing the truncated
receptor GHRH
N, expressed either alone or together with the receptor
GHRH
C (Fig. 4B
), consistent with the results of the binding studies.
Coexpression of either GHRH
N or GHRH
C with the full-length GHRH
receptor did not diminish activation of adenylate cyclase by the
full-length receptor (Fig. 4B
).

View larger version (28K):
[in this window]
[in a new window]
|
Figure 4. Functional Analysis of Truncated GHRH Receptors
A, Binding of GHRH to membranes of cells expressing the wild-type and
truncated human GHRH receptors. The relative amount of input
radioligand bound in the absence and presence of two doses of unlabeled
competitor is shown for each construct tested. The amount of GHRH bound
in the absence of competitor corresponds to 6.7 pmol/tube. B,
Stimulation of intracellular cAMP levels by GHRH in HeLa T4 cells
expressing the wild-type and truncated GHRH receptors. The relative
amount of intracellular cAMP accumulated in response to two doses of
GHRH is shown for each construct tested. The amount of intracellular
cAMP accumulated in cells treated with 1 µM GHRH
corresponds to 80.2 pmol/well. Data points represent the mean of
duplicate samples with the range of values indicated by the
error bars; each panel is representative of at least
three independent experiments.
|
|
The properties of the truncated GHRH receptors indicate that both the N
terminus as well as the C terminus of the receptor are required for
ligand binding and signaling. We therefore pursued an alternative
approach to define the ligand-binding determinants of the GHRH
receptor. Chimeras between related receptors have been used with great
success in defining the ligand-binding domains of several receptors,
such as those for the biogenic amines, glycoprotein hormones, and small
peptides, and of several receptors of family B. We generated chimeras
between the VIP or secretin receptors and the GHRH receptor and studied
their ligand-binding and signaling properties to determine whether the
N- or C-terminal regions of the receptor were most critical for
specific interaction with GHRH.
Expression of Chimeric GHRH and VIP or Secretin Receptors
As described in Table 1
, the
chimeras between the GHRH and VIP or secretin receptors were generated
by the exchange of the N termini of the HA-tagged receptors, with the
fusions made at the AflII site immediately before the first
transmembrane domain. The GHRH-VIP receptor chimeras were
GNVC, which consists of the N-terminal 127
residues of the GHRH receptor and residues 144459 of the VIP
receptor, and VNGC, generated by replacement of
the 127 residues constituting the N terminus of the GHRH receptor with
the equivalent 143 residues forming the N terminus of the VIP receptor.
The GHRH-secretin receptor chimeras were GNSC,
consisting of residues 1127 of the GHRH receptor and residues
143449 of the secretin receptor, and SNGC,
consisting of residues 1142 of the secretin receptor and residues
128423 of the GHRH receptor. The expression of the wild-type and
chimeric receptor proteins was assessed by immunoprecipitation and, as
shown in Fig. 5A
, the proteins produced
had the expected molecular sizes, and the chimeric receptors were
expressed at levels comparable to, or above, those of the wild-type
receptors. The chimeric receptors were glycosylated as determined by
treatment with peptide-N-glycosidase F (data not shown), which reduced
the observed size of the receptor proteins in accord with the number of
potential glycosylation sites in the N-terminal extracellular domain of
each (one in GNVC, three in
VNGC, one in GNSC, and
four in SNGC).

View larger version (96K):
[in this window]
[in a new window]
|
Figure 5. Expression and Cellular Localization of Wild-Type
and Chimeric GHRH Receptors
A, Immunoprecipitation of wild-type and chimeric HA-tagged GHRH, VIP,
and secretin receptors. Equivalent amounts of protein from
metabolically labeled HeLa T4 cells transfected with the various
receptor constructs were immunoprecipitated and separated by SDS-PAGE
on a 10% gel. The sizes of the mol wt standards included on the gel
are shown on the left. B, Immunofluorescence
localization of HA-tagged wild-type and chimeric receptors in cells
permeabilized with 0.1% saponin was performed, and the cells were
observed by fluorescence microscopy. All slides were photographed using
the same exposure times, and the panels shown are representative of at
least 20 fields observed in two independent experiments. The monoclonal
antibody 12CA5 against the HA epitope-tag was used for both
immunoprecipitation and immunofluorescence analyses.
|
|
To confirm that the exchange of domains did not affect the cellular
localization of the chimeric receptors, immunofluorescence analysis of
permeabilized cells expressing the wild-type and chimeric receptors was
performed, and the cells were observed using fluorescence microscopy.
As seen in Fig. 5B
, the wild-type and chimeric receptors are localized
in a similar manner within the cell. All receptor proteins are
distributed both on the cell surface and intracellularly, with the
latter presumably representing receptor protein that is being
transported to, or internalized from, the cell surface.
Analysis of Chimeric GHRH and VIP Receptors
The ligand-binding properties of the chimeras between the GHRH and
VIP receptors were analyzed by measurement of binding to both ligands
using competition assays (Fig. 6
). The
chimera GNVC bound GHRH and VIP at extremely
low levels, while the chimera VNGC bound GHRH
at levels comparable to membranes of cells expressing the wild-type
GHRH receptor (Fig. 6A
) but bound VIP at much lower levels than
membranes of cells expressing the wild-type VIP receptor (Fig. 6B
).
Scatchard analysis of saturation-binding data for binding of GHRH and
VIP to the chimera VNGC was performed, and the
KD values were determined to be 46 pM and 156
pM, respectively. Dose-dependent accumulation of cAMP in
cells transfected with the wild-type and chimeric receptors in response
to treatment with GHRH and VIP is shown in Fig. 7
. Cells expressing the chimera
GNVC show an increase in intracellular cAMP in
response to GHRH and VIP only at high hormone concentrations
(>10-7 M). However, cells expressing the
chimera VNGC showed a robust accumulation of
cAMP in response to treatment with both GHRH and VIP, with
ED50 values of 1.75 and 7.95 nM, respectively,
consistent with the ability of this receptor to bind GHRH, and more
weakly, VIP. Interestingly, the chimera VNGC
mediates a much stronger cAMP response to GHRH than does the wild-type
GHRH receptor, although the two receptors bound GHRH at comparable
levels. The binding and signaling properties of the wild-type and
chimeric GHRH and VIP receptors are summarized in Table 2
.

View larger version (24K):
[in this window]
[in a new window]
|
Figure 6. Binding of GHRH and VIP to Wild-Type and Chimeric
GHRH and VIP Receptors
Dose-dependent competition of GHRH (A) and VIP (B) binding to membranes
of cells expressing the wild-type and chimeric GHRH and VIP receptors.
The relative amount of input radioligand bound in the presence of
increasing concentrations of the respective unlabeled competitor is
shown for each construct tested. The amount of GHRH and VIP bound in
the absence of competitor corresponds to 8.3 and 7.4 pmol/tube,
respectively. Data points represent the mean of duplicate samples with
the range of values indicated by the error bars; each
panel is representative of at least three independent experiments.
|
|

View larger version (29K):
[in this window]
[in a new window]
|
Figure 7. Stimulation of cAMP Production by GHRH and VIP in
Cells Expressing Wild-Type and Chimeric GHRH and VIP Receptors
Dose-dependent cAMP accumulation in HeLa T4 cells expressing wild-type
and chimeric GHRH and VIP receptors after treatment with GHRH (A) and
VIP (B). The relative amount of intracellular cAMP produced in response
to increasing concentrations of hormone is shown for each construct
tested. The amount of intracellular cAMP accumulated in cells treated
with 1 µM GHRH and VIP corresponds to 280.4 and 229.8
pmol/well, respectively. Data points represent the mean of duplicate
samples with the range of values indicated by the error
bars; each panel is representative of at least three
independent experiments.
|
|
Analysis of Chimeric GHRH and Secretin Receptors
The GHRH and VIP receptors are among the most homologous in family
B-III. Although advantageous from the point of minimally disrupting
structure, chimeras between these receptors might not fully reveal GHRH
binding determinants if these determinants are present in the analogous
region of the VIP receptor. We therefore generated additional chimeras
using the secretin receptor, which is less homologous than the VIP
receptor to the GHRH receptor (35% vs. 40% identical) and
which does not detectably bind GHRH. The GHRH-binding ability of the
two chimeras between the GHRH and secretin receptors was determined
using binding competition assays (Fig. 8
). The chimera
GNSC did not show any significant binding of
GHRH while the reciprocal chimera SNGC bound
GHRH at lower levels than the wild-type GHRH receptor, although it was
expressed at higher levels than the wild-type receptor. The
ED50 for competition of GHRH binding by the chimera
SNGC was 1.15 nM, and the
KD value determined by Scatchard analysis of
saturation-binding data was 259.3 pM. We were unable to
iodinate secretin to high specific activity and therefore could not
directly examine binding of secretin to these chimeras, but we studied
the interaction of these chimeras with secretin by quantifying the
intracellular cAMP levels in response to secretin stimulation. After
treatment with GHRH, adenylate cyclase was not activated in cells
expressing the chimera GNSC. In contrast, cells
expressing the reciprocal chimera SNGC
accumulated cAMP in a dose-dependent manner, although there was a small
rightward shift in the response curve (Fig. 9A
). Cells expressing either of the two
chimeras GNSC or SNGC
did not accumulate cAMP after treatment with secretin (Fig. 9B
). The
binding and signaling properties of the wild-type and chimeric GHRH and
secretin receptors are summarized in Table 3
.

View larger version (21K):
[in this window]
[in a new window]
|
Figure 8. GHRH Binding to Wild-Type and Chimeric GHRH and
Secretin Receptors
Dose-dependent competition of GHRH binding to membranes of cells
expressing the wild-type and chimeric GHRH and secretin receptors. The
relative amount of input radioligand bound in the presence of
increasing doses of unlabeled GHRH is shown for each construct tested.
The amount of GHRH bound in the absence of competitor corresponds to
8.4 pmol/tube. Data points represent the mean of duplicate samples with
the range of values indicated by the error bars; each
panel is representative of at least three independent experiments.
|
|

View larger version (27K):
[in this window]
[in a new window]
|
Figure 9. Stimulation of cAMP Production by GHRH and Secretin
in Cells Expressing Wild-Type and Chimeric Human GHRH and Secretin
Receptors
Dose-dependent cAMP accumulation in HeLa T4 cells expressing the
wild-type and chimeric GHRH and secretin receptors after treatment with
GHRH (A) and secretin (B). The relative amount of intracellular cAMP
produced in response to increasing amounts of the hormones is shown for
each construct tested. The amount of intracellular cAMP accumulated in
cells treated with 1 µM GHRH and secretin corresponds to
348.3 and 187 pmol/well, respectively. Data points represent the mean
of duplicate samples with the range of values indicated by the
error bars; each panel is representative of at least
three independent experiments.
|
|
 |
DISCUSSION
|
---|
GHRH and its receptor play an important role in the
proliferation of pituitary somatotroph cells and the regulation of GH
secretion (1, 2, 53), and high-affinity binding of GHRH by its receptor
is a critical prerequisite for normal function of the GH axis (7, 8, 9, 10).
The molecular characterization of the association between GHRH and its
receptor provides a framework for understanding their role in the
regulation of GH secretion and growth, as well as their involvement in
disorders of GH secretion. The approach we have taken toward this
objective was to construct and analyze truncated and chimeric
receptors, to begin to identify the extracellular domains of the GHRH
receptor that are involved in its interaction with GHRH. Our studies of
truncated receptors led us to conclude that neither the N-terminal
extracellular domain nor the C-terminal domain, consisting of the
transmembrane domains and the associated extracellular loops, can bind
ligand in the absence of the other. The analysis of chimeras between
the GHRH receptor and the receptors for VIP and secretin has revealed
that although the N terminus is a critical domain for GHRH binding, the
N terminus of a related receptor can be substituted, suggesting that
the transmembrane domains and associated extracellular loops of the
GHRH receptor form the key determinants for the specific binding of
GHRH.
Studies on the GHRH receptor of the little mouse, in which
mutation of the conserved aspartate at position 60 results in a loss of
the ability of the receptor to bind ligand (26) and activate adenylate
cyclase (7, 8), provided the first indication that the integrity of the
N terminus of the GHRH receptor is essential for ligand binding. In
this study, we found that the insertion of a FLAG epitope tag at
position 37 in the N terminus of the receptor resulted in inactivation
of the receptor. Although this effect could be due in part to altered
expressed or localization of the FLAG-tagged receptor, our data are
consistent with this insertion directly affecting the ligand-binding
properties of the receptor. From computational structural studies on
the receptors of family B, it has been proposed that an
-helical
region in the N terminus of these receptors interacts with an
-helical region in the C terminus of the bioactive region of the
corresponding peptide hormone to form a coiled-coil motif (30). For the
GHRH receptor, this
-helical region is predicted to extend from
residues 2640 and to interact with an
-helix formed by residues
1323 of the hormone. Inactivation of the receptor by the introduction
of the FLAG epitope tag at position 37 might therefore result from the
interruption of this
-helical domain.
Deletion of the N terminus affected transport of the receptor GHRH
N
to the cell surface and resulted in accumulation of the receptor
protein in intracellular membranes, despite inclusion of the signal
peptide in this construct. This abnormal localization of GHRH
N could
be a consequence of the absence of the site for N-linked glycosylation
or of defective transport due to the proximity of the signal peptide
and the first transmembrane domain. The larger of the two bands seen in
immunoprecipitation analysis of extracts from cells expressing GHRH
N
is likely to be receptor protein in which cleavage of the signal
peptide has not occurred, potentially due to structural constraints
resulting from the adjacency of the first transmembrane domain. The
receptor GHRH
N did not bind GHRH in a competition assay using total
cell membranes, nor did it activate adenylate cyclase upon hormone
treatment, implying that the N terminus is essential for GHRH binding.
The deletion of the N termini of the receptors for VIP and glucagon
also resulted in proteins that did not bind to their respective ligands
(27, 48), suggesting that the N terminus plays an important role in the
formation of the binding site of receptors of family B-III.
The receptor truncated in the first intracellular loop, GHRH
C, was
localized within cells in a manner similar to the wild-type GHRH
receptor but did not bind GHRH, indicating that although essential for
ligand binding, the N terminus alone is not sufficient for interaction
with GHRH. Our results with GHRH
C are similar to those obtained with
a C-terminally truncated glucagon receptor (48) but are distinct from
those obtained with a similarly truncated PACAP receptor that
specifically bound PACAP, although with significantly lower affinity
than the wild-type PACAP receptor (40).
Although coexpression of N- and C-terminal domains of receptors can
result in partially functional receptors (32, 54, 55), suggesting
reconstitution of receptors by domain-domain interaction, coexpression
of the two truncated forms of the GHRH receptor, GHRH
N and GHRH
C,
did not reconstitute ligand-binding activity. This could be due to poor
colocalization of the receptor fragments or due to a lack of
interaction of the two truncated proteins resulting from disruption of
the complex by the presence of the first transmembrane domain in both
receptor fragments. Additionally, we coexpressed the truncated and
wild-type GHRH receptors and found that there was no decrease in
hormone binding or signaling by the wild-type receptor, implying that
the truncated receptors GHRH
N and GHRH
C do not act as functional
dominant negative mutants.
The generation of truncation constructs of GPCRs has been useful for
the identification of structural domains involved in ligand binding in
several receptors (40, 54, 55, 56, 57, 58). However, this approach has
disadvantages, in that deletion of domains or truncations can result in
structural alterations and mislocalization of the receptor proteins
(48, 58). The generation of chimeras between two similar but distinct
receptors can be used to correlate gain or loss of function with the
exchanged domain, often with minimal effects on the structure of the
receptor. Chimeric receptors generated by the exchange of homologous
domains have proven useful in localizing the ligand-binding domains of
receptors for biogenic amines (31, 32), glycoprotein hormones (33), and
small peptides (34, 35, 36), and also of several receptors in family B
including the VIP, secretin, glucagon, GIP, GLP-1, CRH, and PTH
receptors (38, 39, 44, 46, 47, 49, 51, 52). We therefore applied this
approach to characterize the ligand-binding domains of the GHRH
receptor.
The wild-type GHRH, VIP, and secretin receptors show a high degree of
specificity in their interaction with their respective ligands,
although some ability of GHRH to activate the VIP receptor and of VIP
to activate the GHRH receptor at high ligand concentrations has been
observed (3, 59). Both the VIP and secretin receptors require the
N-terminal extracellular domain as well as the first extracellular loop
for high-affinity ligand recognition (29, 38, 39, 41, 42, 43, 44). Exchange of
the N-terminus of the VIP or secretin receptors with the corresponding
region of the GHRH receptor resulted in chimeras
GNVC, which showed a very weak response to both
GHRH and VIP, and GNSC, which was unresponsive
to both GHRH and secretin, consistent with studies of chimeras between
the VIP and secretin receptors (38, 39). However, the chimeric
receptors VNGC and
SNGC, having the N terminus of the VIP or
secretin receptors and the C terminus of the GHRH receptor, bound GHRH
and stimulated adenylate cyclase in a dose-dependent manner in response
to GHRH. The chimera SNGC bound GHRH at lower
levels but had a similar cAMP response compared with the wild-type
receptor, while the chimera VNGC showed similar
binding but a higher signaling response to GHRH than the wild-type
receptor. Our observation that the chimera VNGC
bound VIP and activated adenylate cyclase in response to VIP can be
explained by the fact that the aspartate residue at position 196 in the
first extracellular loop of the VIP receptor, which is predicted to be
important in interaction with VIP (43), is also present in the
corresponding region of the GHRH receptor. The VIP receptor and the
chimeric receptor GNVC mediate cAMP
accumulation in response to very high concentrations of GHRH,
consistent with a low-affinity interaction of the VIP receptor with
GHRH. The differences between the binding and signaling responses in
the chimeric receptors SNGC and
VNGC suggest that some of the determinants
required for GHRH binding are present in the N terminus of the VIP
receptor but not the secretin receptor. Alignment of the N-terminal
regions of the three receptors (3, 24) indicates that among the
residues predicted to form the
-helix suggested to be involved in
the coiled-coil interaction with the ligand (30), there is a higher
homology between the GHRH and VIP receptors than between the GHRH and
secretin receptors. Additionally, comparison of the amino acids at
positions 1323 in the hormones GHRH, VIP, and secretin, which are
predicted to form the
-helices that interact with the receptors
(30), shows that this region of GHRH is more homologous to VIP than to
secretin. These differences between VIP and secretin, and between their
receptors, are likely to account for the differences in the
GHRH-binding and signaling responses of the chimeras
VNGC and SNGC.
Our data from the truncated receptors demonstrate that neither the N
terminus nor the C terminus of the GHRH receptor can bind ligand in the
absence of the other domain. Our studies of the chimeric receptors
suggest that one or more of the extracellular loops, in conjunction
with residues in the transmembrane domains, function as the
determinants of binding specificity and activation for the GHRH
receptor. Although the N terminus is essential for ligand binding,
substitution of this domain with the corresponding domain of a related
family B-III receptor reconstitutes functional GHRH binding. It has
been hypothesized that all the receptors in this family have a similar
structure in the N-terminal domain as a result of the conserved pattern
of cysteine residues forming conserved disulfide linkages (29), and an
-helix in this region is predicted to be involved in the primary
interaction with the ligand (30). From our results and those obtained
from studies on other receptors in this family (38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 49, 50, 51, 52, 60), it
is conceivable that the N terminus of these receptors plays a primary
structural role in the initial interaction with the ligand, while
residues in the extracellular loops and the transmembrane domains are
involved in secondary interactions with the ligand that determine the
specificity of ligand binding. Our results provide a basis for future
studies to identify specific receptor domains and residues involved in
recognition and binding of GHRH. Chimeras in which individual
extracellular loops of the receptor are substituted will enable the
localization of binding determinants in a more precise manner, allowing
site-directed mutagenesis to be used to define the key residues
involved in specific ligand interaction. Identification of domains that
are structurally important for the interaction of GHRH with its
receptor will facilitate the design of specific receptor agonists and
antagonists that might have therapeutic use in the diagnosis or
treatment of diseases of the GH axis.
 |
MATERIALS AND METHODS
|
---|
Plasmids, Receptor Constructs, and Hormones
All receptor constructs were made in the vector pcDNA-3
(Invitrogen, Carlsbad, CA) downstream of the T7 polymerase promoter and
were flanked by HindIII and XbaI sites at the 5'-
and 3'-ends, respectively. Their sequences were confirmed using the
dideoxy nucleotide chain termination method (Amersham Life Sciences,
Arlington Heights, IL). In the numbering system used for all clones in
subsequent sections, the first nucleotide of the initiation codon is
defined as position 1. The cDNA clones for the rat VIP (15) and
secretin receptors (14) were provided by Dr. S. Nagata (Osaka
Bioscience Institute, Osaka, Japan), and the cDNA for the human GHRH
receptor was previously cloned in the laboratory (3). Peptide hormones
were obtained from Peptides International (Louisville, KY) or Peninsula
Laboratories (Belmont, CA).
Generation of Epitope-Tagged Receptor Constructs
Oligonucleotides having 2022 nucleotides complementary to the
C-terminal end of the GHRH, VIP, or secretin receptors and encoding the
influenza virus HA epitope (61) were synthesized. Recognition sites for
the enzymes KpnI and XbaI were engineered on
either side of the HA epitope with the stop codon within the
XbaI site. These primers, together with upstream primers
within each of the receptor cDNAs, were used to amplify the C-terminal
fragment of each receptor. The PCR products were cloned in context with
the respective receptor into the expression vector pcDNA-3 to
generate full-length clones for the epitope-tagged receptors (GHRHR.HA,
VIPR.HA, and SECR.HA).
The amino acids for the FLAG epitope tag (62) were inserted at the
N-terminal domain of the GHRH receptor downstream of the putative
signal peptide at residue 37 using gene splicing by overlap extension
(63). An oligonucleotide primer encoding the FLAG epitope tag with a
site for the enzyme EcoRV, 3' of the tag and having 22
nucleotides complementary to the GHRH receptor was synthesized. This
primer and a downstream primer were used to amplify a fragment
corresponding to amino acids 130580 of the receptor. A primer
complementary to the sense primer for the FLAG epitope and the T7
promoter primer were used to amplify a fragment corresponding to amino
acids 1140 of the receptor. The two fragments were annealed and
amplified using the two external primers, and the product was cloned in
context with the C-terminal region of the receptor to generate the
FLAG-tagged GHRH receptor GHRHR.F.
Generation of Truncation and Chimeric Constructs of the GHRH
Receptor
Using site-directed mutagenesis (64), the recognition site for
the enzyme AflII (CTTAAG) was engineered in the tagged GHRH
receptors as shown in Fig. 1
. The site was introduced at nucleotides
100106 and 385390 (corresponding to amino acids 3435 and
129130), respectively, in the HA-tagged receptor and at nucleotides
385390 in the FLAG-tagged receptor. This resulted in changes in amino
acids 35 and 129 (Arg to Lys and Val to Leu, respectively).
Removal of the 285-bp AflII-AflII fragment
resulted in the deletion of the extracellular domain between amino
acids 35 and 129 to give the N-terminally truncated HA-tagged construct
(GHRH
N.HA). Deletion of the 278-bp EcoRV-AflII
fragment of the FLAG-tagged GHRH receptor, followed by filling-in with
Klenow polymerase and ligation, generated the N-terminally truncated
FLAG-tagged construct (GHRH
N.F).
An oligonucleotide complementary to the first intracellular loop of the
GHRH receptor and with nucleotides 467472 altered to form a
recognition site for the enzyme KpnI, together with the T7
promoter primer, was used to amplify a 480-bp N-terminal fragment of
the GHRH receptor. The PCR product was digested with HindIII
and KpnI and ligated with the large fragment obtained by
digestion of the full-length HA-tagged GHRH receptor with the same
enzymes, to generate the construct GHRH
C.HA, truncated within the
first intracellular loop. The same primers were used to amplify a
516-bp fragment using the FLAG-tagged GHRH receptor as a template, and
the product was ligated into pcDNA-3 to generate the clone
GHRH
C.F.
The chimeras were constructed by the exchange of N-terminal
extracellular domains between the HA-tagged receptors with the
predicted start of the first transmembrane domain of the receptors as
the junction (Table 1
). The recognition site for AflII was
engineered at nucleotide positions 385390, 427432, and 424439
(amino acids 129130, 143144, and 142143) in the cDNAs for the
GHRH, VIP, and secretin receptors, respectively, and the wild-type
receptors having the epitope tag and AflII site were used
for all subsequent studies. The N-terminal fragments that were
exchanged between the receptors were generated by digestion of the
cDNAs for the wild-type receptors with HindIII and
AflII.
Expression of Receptor Constructs in HeLa T4 cells Using the
Vaccinia-T7 RNA Polymerase Hybrid Expression System
All experiments were performed using cells transfected with
various receptor constructs using the Vaccinia Virus-T7 polymerase
hybrid expression system (65). Subconfluent monolayers of HeLa T4 cells
cultured in DMEM (Sigma Chemical Co., St. Louis, MO) supplemented with
5% FBS (GIBCO BRL, Grand Island, NY) were infected with Vaccinia virus
vTF7.3 expressing the bacteriophage T7 RNA polymerase (obtained under
license from Dr. Bernard Moss, NIH, Bethesda, MD), at a multiplicity of
infection of 10, for 30 min in PBS/0.1% BSA. The various plasmid DNAs
were incubated with liposomes (66) at the ratio of 45 µg of lipid
per µg DNA, in Opti-MEM I medium (GIBCO BRL), for 2030 min at room
temperature. Virus was aspirated from the cells, and the DNA/liposome
mixture was added to the cells and incubated at 37 C in 5%
CO2 for 1516 h. The amount of DNA used for transfection
varied with the size of the plates.
Metabolic Labeling of Transfected Cells and Immunoprecipitation
of Epitope-Tagged Receptors
Cells grown in 35-mm dishes and transfected with 5 µg DNA per
plate were starved in cysteine/methionine-deficient DMEM (ICN
Biomedical Inc., Irvine, CA) for 30 min and labeled with 50 µCi/plate
ProMix (Amersham) in the same medium for 2 h at 37 C in 5%
CO2. The cells were washed with PBS, harvested, and
subjected to one cycle of freeze-thaw. The cell pellets were
resuspended in 400 µl RIPA buffer [150 mM NaCl, 50
mM Tris-HCl (pH 7.5), 1% Nonidet P-40, 0.5% deoxycholic
acid, and 0.1% SDS] containing 0.1 mM
phenylmethylsulfonyl fluoride and 1 µg/ml leupeptin, and the
membranes were solubilized by incubation on ice for 1 h with
vortexing. The lysate was clarified by centrifugation and incubated
with 0.5 µg of the HA-specific 12CA5 ascites fluid (a gift from Dr.
Robert A. Lamb, Northwestern University) for 34 h at 4 C on a
hematology mixer. At the end of this period, 30 µl of a 50%
suspension of protein A-Sepharose beads (Pharmacia, Piscataway, NJ) in
PBS were added to the tubes, and the incubation was continued for 30
min. The beads were washed four times with 500 µl cold RIPA buffer
and once with cold wash buffer (50 mM Tris-HCl/pH 7.5, 150
mM NaCl, 5 mM EDTA) and resuspended in 30 µl
of 2x SDS-PAGE sample buffer (50 mM Tris-HCl/pH 6.8, 2%
SDS, 10% glycerol, 5% ß-mercaptoethanol, 0.1% bromophenol blue).
The samples were boiled for 5 min before separation by SDS-PAGE using a
Tris-glycine buffer with See-Blue Pre-Stained Standard (NOVEX, San
Diego, CA) as size markers. The gels were fixed in 20% methanol/7%
acetic acid, saturated with glacial acetic acid, impregnated with 22%
wt/vol of 2,5-diphenyl-oxazole in acetic acid, dried, and exposed to
Kodak X-OMAT AR film (Eastman Kodak Company, Rochester, NY). For
glycosylation analysis, immunoprecipitated proteins were digested for
4 h at 37 C with 0.2 U of peptide-N-glycosidase F
(Boehringer Mannheim Corp., Indianapolis, IN) in a buffer containing 20
mM Na2HPO4 (pH 8.0), 20
mM EDTA, 1% NP40, 1 µg/ml leupeptin, 0.1 µg/ml
pepstatin A, and 1 µg/ml aprotinin.
Immunofluorescence Localization of Epitope-Tagged Receptors
HeLa T4 cells cultured on glass coverslips in 35-mm plates were
transfected as described. For permeabilized cells expressing the
FLAG-tagged constructs, the coverslips were washed twice in PBS and
incubated for 2 h at 4 C with 3 µg/ml of the anti-FLAG M2
monoclonal antibody (Kodak IBI, New Haven, CT) in PBS containing 0.1%
saponin. After extensive washing, the coverslips were incubated at 4 C
for 30 min with 2 µg/ml of fluorescein isothiocyanate
(FITC)-conjugated goat anti-mouse secondary antibody (Jackson
ImmunoResearch Laboratories, West Grove, PA) in PBS containing 0.1%
saponin and 0.2% whole goat serum, and after extensive washing with
PBS, the coverslips were mounted in FITC-Guard (Testog Inc., Chicago,
IL). For nonpermeabilized cells, incubation was carried out under
similar conditions using primary and secondary antibodies diluted in
PBS (without saponin). For cells expressing the HA-tagged receptor
constructs, the coverslips were washed twice in PBS, fixed with 1%
(wt/vol) paraformaldehyde in PBS for 10 min, and immunofluorescense
analysis was performed as described, using 1 µg/ml of the HA-specific
12CA5 ascites fluid. The images of cells expressing the FLAG-tagged and
HA-tagged receptors in Fig. 3B
were optical sections obtained using
confocal laser scanning microscopy with a Bio-Rad MRC 600 (Bio-Rad
Laboratories, Richmond, CA) connected to a Nikon microscope using a
40x objective. All samples were scanned under the same contrast
settings for equivalent times, and optical sections were taken through
the central plane of the cell. Photomicroscopy of cells expressing the
wild-type and chimeric receptors in Fig. 5B
was performed using a 63x
objective on a Zeiss Axiophot microscope (Carl Zeiss Inc., Oberkochen,
Germany) with equivalent exposure times.
Measurement of Ligand Binding
For binding assays, cells grown in 10-cm dishes were
transfected with 10 µg DNA per plate. Binding assays were performed
using approximately 50 µg of membrane protein per reaction, prepared
from transfected cells as described (3). The reactions were carried out
at 25 C for 1 h in a 300-µl reaction volume and were terminated
by centrifugation at 4 C for 10 min. The membrane pellets were washed
once with binding buffer, and the amount of bound radioligand was
measured using a
-counter. The assays were performed with duplicate
samples, and the means were used for all further calculations. For
competition studies, the membrane proteins were incubated with
either
(3-[125I]iodotyrosyl10)GHRH(144)-amide or
(3-[125I] iodotyrosyl10)VIP present at a
concentration of 70 pM (Amersham), in the absence or
presence of increasing concentrations of the unlabeled hormone. The
nonspecific binding, determined as the percent of input counts bound in
the presence of 1 µM unlabeled hormone, was approximately
13% and 7% for GHRH and VIP, respectively, and was subtracted from
all raw data to give the specific bound counts. The percentage of the
maximum specific bound counts for each data point was calculated using
the specific counts bound by the wild-type receptor in the absence of
competitor as 100%. Saturation-binding assays are representative of at
least two independent experiments. The concentration of the radioligand
ranging from 1.37 pM to 700 pM and the
corresponding unlabeled hormone was present at 1 µM. The
binding-competition data were fit to a one-site competition equation to
determine ED50, and Scatchard analysis of
saturation-binding data was used to determine KD and
BMAX values using the program GraphPad Prism (GraphPad
Software Inc., San Diego, CA).
Measurement of Intracellular cAMP Levels
For the measurement of cAMP responses, cells were transfected in
12-well plates using 2.5 µg plasmid DNA per well. The cells were
treated with hormones as described (3) for 20 min at 37 C, lysed in 150
µl of cold 0.1 M HCl, and harvested, and the lysates were
neutralized with an equal volume of 150 mM Tris-HCl (pH 8)
containing 4 mM EDTA. The protein was removed by
centrifugation for 10 min at 4 C, and the supernatants were used to
assay cAMP by a competitive protein-binding assay (67) using 4.5
nM [8-3H]cAMP (Amersham), as a tracer. The
protein-bound [8-3H]cAMP in the supernatant was measured
by liquid scintillation counting using CytoScint (ICN), and a linear
standard curve was performed in each assay. The assays were performed
with duplicate samples, and the means were used for all further
calculations. The percentage of the maximum cAMP accumulated for each
data point was calculated using the cAMP accumulated in cells
expressing the wild-type receptor, and treated with 1 µM
hormone, as 100%. The dose-response curves were fit to a sigmoidal
dose-response equation, and ED50 values were determined
using the program GraphPad Prism (GraphPad Software Inc.).
 |
ACKNOWLEDGMENTS
|
---|
We thank Dr. Shigekazu Nagata (Osaka Bioscience Institute) for
the cDNA clones for the VIP and secretin receptors, Dr. Bernard Moss
(NIH, Bethesda, MD) for the use of the Vaccinia-T7 polymerase system,
Dr. Robert Lamb (Northwestern University, Evanston, IL) for the
monoclonal antibody against the HA epitope, Ken Wu for technical
assistance, Katherine Lee for participating in the generation of the
HA-tagged truncation constructs, and Drs. Daniel Linzer and Teresa
Miller for comments on the manuscript.
 |
FOOTNOTES
|
---|
Address requests for reprints to: Kelly E. Mayo, Department of Biochemistry, Molecular Biology and Cell Biology, Northwestern University, Evanston, Illinois 60208. E-mail: k-mayo{at}nwu.edu
This work was supported by NIH Grant DK-48071 (to K.E.M.).
Received for publication October 13, 1997.
Revision received January 15, 1998.
Accepted for publication January 15, 1998.
 |
REFERENCES
|
---|
-
Frohman LA, Jansson JO 1986 Growth hormone-releasing
hormone. Endocr Rev 7:223253[Abstract]
-
Gelato MC, Merriam GR 1986 Growth hormone releasing hormone.
Annu Rev Physiol 48:569591[CrossRef][Medline]
-
Mayo KE 1992 Molecular cloning and expression of a
pituitary-specific receptor for growth hormone-releasing hormone. Mol
Endocrinol 6:17341744[Abstract]
-
Lin C, Lin SC, Chang CP, Rosenfeld MG 1992 Pit-1-dependent
expression of the receptor for growth hormone releasing factor mediates
pituitary cell growth. Nature 360:765768[CrossRef][Medline]
-
Gaylinn BD, Harrison JK, Zysk JR, Lyons CE, Lynch KR, Thorner
MO 1993 Molecular cloning and expression of a human anterior pituitary
receptor for growth hormone-releasing hormone. Mol Endocrinol 7:7784[Abstract]
-
Hsiung HM, Smith DP, Zhang XY, Bennett T, Rosteck PJ, Lai MH 1993 Structure and functional expression of a complementary DNA for
porcine growth hormone-releasing hormone receptor. Neuropeptides 25:110[Medline]
-
Godfrey P, Rahal JO, Beamer WG, Copeland NG, Jenkins NA, Mayo
KE 1993 GHRH receptor of little mice contains a missense
mutation in the extracellular domain that disrupts receptor function.
Nat Genet 4:227232[Medline]
-
Lin SC, Lin CR, Gukovsky I, Lusis AJ, Sawchenko PE, Rosenfeld
MG 1993 Molecular basis of the little mouse phenotype
and implications for cell type-specific growth. Nature 364:208213[CrossRef][Medline]
-
Wajnrajch MP, Gertner JM, Harbison MD, Chua Jr SC, Leibel RL 1996 Nonsense mutation in the human growth hormone-releasing hormone
receptor causes growth failure analogous to the little
(lit) mouse. Nat Genet 12:8890[Medline]
-
Maheshwari I, Silverman BL, Dupuis J, Baumann G, Dwarfism of
Sindh: a novel form of familial isolated GH deficiency linked to the
locus for the GH releasing hormone receptor. Program of the 10th
International Congress of Endocrinology, San Francisco, CA, 1996, p 709
(Abstract)
-
Tang J, Lagace G, Castagne J, Collu R 1995 Identification of
human growth hormone-releasing hormone receptor splicing variants.
J Clin Endocrinol Metab 80:23812387[Abstract]
-
Hashimoto K, Koga M, Motomura T, Kasayama S, Kouhara H,
Ohnishi T, Arita N, Hayakawa T, Sato B, Kishimoto T 1995 Identification
of alternatively spliced messenger ribonucleic acid encoding truncated
growth hormone-releasing hormone receptor in human pituitary adenomas.
J Clin Endocrinol Metab 80:29332939[Abstract]
-
Kolakowski LF, Zhuang J 1997 G Protein-Coupled Receptor Family
B. In: The G protein-Coupled Receptor DataBase, World-Wide-Web Site.
URL: http://www.gcrdb.uthscsa.edu/FBintro.html
-
Ishihara T, Nakamura S, Kaziro Y, Takahashi T, Takahashi K,
Nagata S 1991 Molecular cloning and expression of a cDNA encoding the
secretin receptor. EMBO J 10:16351641[Abstract]
-
Ishihara T, Shigemoto R, Mori K, Takahashi K, Nagata S 1992 Functional expression and tissue distribution of a novel receptor for
vasoactive intestinal polypeptide. Neuron 8:811819[Medline]
-
Pisegna JR, Wank SA 1993 Molecular cloning and functional
expression of the pituitary adenylate cyclase-activating polypeptide
type I receptor. Proc Natl Acad Sci USA 90:63456349[Abstract]
-
Jelinek LJ, Lok S, Rosenberg GB, Smith RA, Grant FJ, Biggs S,
Bensch PA, Kuijper JL, Sheppard PO, Sprecher CA, OHara PJ, Foster J,
Walker KM, Chen LHJ, McKernan PA, Kindsvogel W 1993 Expression
cloning and signaling properties of the rat glucagon receptor. Science 259:16141616[Medline]
-
Thorens B 1992 Expression cloning of the pancreatic beta cell
receptor for the gluco-incretin hormone glucagon-like peptide 1. Proc
Natl Acad Sci USA 89:86418645[Abstract]
-
Usdin TB, Mezey E, Button DC, Brownstein MJ, Bonner TI 1993 Gastric inhibitory polypeptide receptor, a member of the
secretin-vasoactive intestinal peptide receptor family, is widely
distributed in peripheral organs and the brain. Endocrinology 133:28612870[Abstract]
-
Juppner H, Abou-Samra A, Freeman M, Kong XF, Schipani E,
Richards J, Kolakowski Jr LF, Hock J, Potts Jr JT, Kronenberg HM, Segre
GV 1991 A G protein-linked receptor for parathyroid hormone and
parathyroid hormone-related peptide. Science 254:10241026[Medline]
-
Abou-Samra A, Juppner H, Force T, Freeman MW, Kong XF,
Schipani E, Urena P, Richards J, Bonventre JV, Potts Jr JT, Kronenberg
HM, Segre GV 1992 Expression cloning of a common receptor for
parathyroid hormone and parathyroid hormone-related peptide from rat
osteoblast-like cells: a single receptor stimulates intracellular
accumulation of both cAMP and inositol trisphosphates and increases
intracellular free calcium. Proc Natl Acad Sci USA 89:27322736[Abstract]
-
Chen R, Lewis KA, Perrin MH, Vale WW 1993 Expression cloning
of a human corticotropin-releasing-factor receptor. Proc Natl Acad Sci
USA 90:89678971[Abstract]
-
Lin HY, Harris TL, Flannery MS, Aruffo A, Kaji EH, Gorn A,
Kolakowski LJ, Lodish HF, Goldring SR 1991 Expression cloning of an
adenylate cyclase-coupled calcitonin receptor. Science 254:10221024[Medline]
-
Segre GV, Goldring SR 1993 Receptors for secretin, calcitonin,
parathyroid hormone (PTH)/PTH-related peptide, vasoactive intestinal
peptide, glucagon-like peptide 1, growth hormone-releasing hormone and
glucagon belong to a newly discovered G-protein-linked receptor family.
Trends Endocrinol Metab 4:309314
-
Gaudin P, Couvineau A, Maoret JJ, Rouyer FC, Laburthe M 1995 Mutational analysis of cysteine residues within the extracellular
domains of the human vasoactive intestinal peptide (VIP) 1 receptor
identifies seven mutants that are defective in VIP binding. Biochem
Biophys Res Commun 211:901908[CrossRef][Medline]
-
Gaylinn BD, Lyons CEJ, Mayo KE, Thorner MO, The
little mouse GHRH receptor mutation (D60->G) prevents
GHRH binding. Program of the 76th Annual Meeting of the Endocrine
Society, Anaheim, CA, 1994, p 367 (Abstract)
-
Couvineau A, Gaudin P, Maoret JJ, Rouyer FC, Nicole P,
Laburthe M 1995 Highly conserved aspartate 68, tryptophane 73 and
glycine 109 in the N-terminal extracellular domain of the human VIP
receptor are essential for its ability to bind VIP. Biochem Biophys Res
Commun 206:246252[CrossRef][Medline]
-
Carruthers CJ, Unson CG, Kim HN, Sakmar TP 1994 Synthesis and
expression of a gene for the rat glucagon receptor. Replacement of an
aspartic acid in the extracellular domain prevents glucagon binding.
J Biol Chem 269:2932129328[Abstract/Free Full Text]
-
Laburthe M, Couvineau A, Gaudin P, Maoret JJ, Rouyer-Fessard
C, Nicole P 1996 Receptors for VIP, PACAP, secretin, GRF, glucagon,
GLP-1, and other members of their new family of G protein-linked
receptors: structure-function relationship with special reference to
the human VIP-1 receptor. Ann NY Acad Sci 805:94109[Medline]
-
Momany FA, Bowers CY 1996 Speculations on the mechanism of
hormone-receptor interactions of the secretin/glucagon family of
polypeptide hormones derived from computational structural studies. Ann
NY Acad Sci 805:172181[Medline]
-
Wess J 1993 Mutational analysis of muscarinic acetylcholine
receptors: structural basis of ligand/receptor/G protein interactions.
Life Sci 53:14471463[CrossRef][Medline]
-
Kobilka BK, Kobilka TS, Daniel K, Regan JW, Caron MG,
Lefkowitz RJ 1988 Chimeric alpha 2-, beta 2-adrenergic receptors:
delineation of domains involved in effector coupling and ligand binding
specificity. Science 240:13101316[Medline]
-
Combarnous Y 1992 Molecular basis of the specificity of
binding of glycoprotein hormones to their receptors. Endocr Rev 13:670691[Medline]
-
Fong TM, Huang RR, Yu H, Strader CD 1993 Mapping the ligand
binding site of the NK-1 receptor. Regul Pept 46:4348[CrossRef][Medline]
-
Fukuda K, Terasako K, Kato S, Mori K 1995 Identification of
the amino acid residues involved in selective agonist binding in the
first extracellular loop of the delta- and mu-opioid receptors. FEBS
Lett 373:177181[CrossRef][Medline]
-
Hunyady L, Balla T, Catt KJ 1996 The ligand binding site of
the angiotensin AT1 receptor. Trends Pharmacol Sci 17:135140[CrossRef][Medline]
-
Wilmen A, Goke B, Goke R 1996 The isolated N-terminal
extracellular domain of the glucagon-like peptide-1 (GLP)-1 receptor
has intrinsic binding activity. FEBS Lett 398:4347[CrossRef][Medline]
-
Holtmann MH, Hadac EM, Miller LJ 1995 Critical contributions
of amino-terminal extracellular domains in agonist binding and
activation of secretin and vasoactive intestinal polypeptide receptors.
Studies of chimeric receptors. J Biol Chem 270:1439414398[Abstract/Free Full Text]
-
Vilardaga JP, De NP, Di PE, Bollen A, Waelbroeck M, Robberecht
P 1995 Properties of chimeric secretin and VIP receptor proteins
indicate the importance of the N-terminal domain for ligand
discrimination. Biochem Biophys Res Commun 211:885891[CrossRef][Medline]
-
Cao YJ, Gimpl G, Fahrenholz F 1995 The amino-terminal fragment
of the adenylate cyclase activating polypeptide (PACAP) receptor
functions as a high affinity PACAP binding domain. Biochem Biophys Res
Commun 212:673680[CrossRef][Medline]
-
Vilardaga JP, di Paolo E, de Neef P, Waelbroeck M, Bollen A,
Robberecht P 1996 Lysine 173 residue within the first exoloop of rat
secretin receptor is involved in carboxylate moiety recognition of Asp
3 in secretin. Biochemistry 218:842846
-
Holtmann MH, Ganguli S, Hadac EM, Dolu V, Miller LJ 1996 Multiple extracellular loop domains contribute critical determinants
for agonist binding and activation of the secretin receptor. J
Biol Chem 271:1494414949[Abstract/Free Full Text]
-
Du K, Nicole P, Couvineau A, Laburthe M 1997 Aspartate 196 in
the first extracellular loop of the human VIP1 receptor is essential
for VIP binding and VIP-stimulated cAMP production. Biochem Biophys Res
Commun 230:289292[CrossRef][Medline]
-
Hashimoto H, Ogawa N, Hagihara N, Yamamoto K, Imanishi K, Nogi
H, Nishino A, Fujita T, Matsuda T, Nagata S, Baba A 1997 Vasoactive
intestinal polypeptide and pituitary adenylate cyclase-activating
polypeptide receptor chimeras reveal domains that determine specificity
of vasoactive intestinal polypeptide binding and activation. Mol
Pharmacol 52:128135[Abstract/Free Full Text]
-
Turner PR, Bambino T, Nissenson RA 1996 A putative selectivity
filter in the G-protein-coupled receptors for parathyroid hormone and
secretin. J Biol Chem 271:92059208[Abstract/Free Full Text]
-
Buggy JJ, Livingston JN, Rabin DU, Yoo-Warren H 1995 Glucagon.glucagon-like peptide I receptor chimeras reveal domains that
determine specificity of glucagon binding. J Biol Chem 270:74747478[Abstract/Free Full Text]
-
Stroop SD, Kuestner RE, Serwold TF, Chen L, Moore EE 1995 Chimeric human calcitonin and glucagon receptors reveal two dissociable
calcitonin interaction sites. Biochemistry 34:10501057[Medline]
-
Unson CG, Cypess AM, Kim HN, Goldsmith PK, Carruthers CJ,
Merrifield RB, Sakmar TP 1995 Characterization of deletion and
truncation mutants of the rat glucagon receptor. Seven transmembrane
segments are necessary for receptor transport to the plasma membrane
and glucagon binding. J Biol Chem 270:2772027727[Abstract/Free Full Text]
-
Juppner H, Schipani E, Bringhurst FR, McClure I, Keutmann HT,
Potts Jr JT, Kronenberg HM, Abou-Samra A, Segre GV, Gardella TJ 1994 The extracellular amino-terminal region of the parathyroid hormone
(PTH)/PTH-related peptide receptor determines the binding affinity for
carboxyl-terminal fragments of PTH-(134). Endocrinology 134:879884[Abstract]
-
Lee C, Luck MD, Juppner H, Potts Jr JT, Kronenberg HM,
Gardella TJ 1995 Homolog-scanning mutagenesis of the parathyroid
hormone (PTH) receptor reveals PTH-(134) binding determinants in the
third extracellular loop. Mol Endocrinol 9:12691278[Abstract]
-
Gelling RW, Wheeler MB, Xue J, Gyomorey S, Nian C, Pederson
RA, McIntosh CHS 1997 Localization of the domains involved in ligand
binding and activaton of the glucose-dependent insulinotropic
polypeptide receptor. Endocrinology 138:26402643[Abstract/Free Full Text]
-
Liaw CW, Grigoriadis DE, Lovenberg TW, De Souza EB, Maki RA 1997 Localization of ligand-binding domains of human
corticotropin-releasing factor receptor: a chimeric receptor approach.
Mol Endocrinol 11:980987[Abstract/Free Full Text]
-
Mayo KE, Godfrey PA, Suhr ST, Kulik DJ, Rahal JO 1995 Growth
hormone-releasing hormone: synthesis and signaling. Recent Prog Horm
Res 50:3573[Medline]
-
Maggio R, Vogel Z, Wess J 1993 Reconstitution of functional
muscarinic receptors by co-expression of amino- and carboxyl-terminal
receptor fragments. FEBS Lett 319:195200[CrossRef][Medline]
-
Remy JJ, Bozon V, Couture L, Goxe B, Salesse R, Garnier J 1993 Reconstitution of a high-affinity functional lutropin receptor by
coexpression of its extracellular and membrane domains. Biochem Biophys
Res Commun 193:10231030[CrossRef][Medline]
-
Xie YB, Wang H, Segaloff DL 1990 Extracellular domain of
lutropin/choriogonadotropin receptor expressed in transfected cells
binds choriogonadotropin with high affinity. J Biol Chem 265:2141121414[Abstract/Free Full Text]
-
Ji IH, Ji TH 1991 Human choriogonadotropin binds to a
lutropin receptor with essentially no N-terminal extension and
stimulates cAMP synthesis. J Biol Chem 266:1307613079[Abstract/Free Full Text]
-
Moyle WR, Bernard MP, Myers RV, Marko OM, Strader CD 1991 Leutropin/beta-adrenergic receptor chimeras bind choriogonadotropin and
adrenergic ligands but are not expressed at the cell surface. J
Biol Chem 266:1080710812[Abstract/Free Full Text]
-
Usdin TB, Bonner TI, Mezey E 1994 Two receptors for vasoactive
intestinal polypeptide with similar specificity and complementary
distributions. Endocrinology 135:26622680[Abstract]
-
Unson CG, Cypess AM, Wu CR, Goldsmith PK, Merrifield RB,
Sakmar TP 1996 Antibodies against specific extracellular epitopes of
the glucagon receptor block glucagon binding. Proc Natl Acad Sci USA 93:310315[Abstract/Free Full Text]
-
Kolodziej PA, Young RA 1991 Epitope tagging and protein
surveillance. Methods Enzymol 194:508519[Medline]
-
Hopp TP, Prickett KS, Price V, Libby RT, March CJ, Ceretti P,
Urdal DL, Conlon PJ 1988 A short polypeptide marker sequence useful for
recombinant protein identification and purification. Biotechnology 6:12051210
-
Horton RM, Hunt HD, Ho SN, Pullen JK, Pease LR 1989 Engineering hybrid genes without the use of restriction enzymes: gene
splicing by overlap extension. Gene 77:6168[CrossRef][Medline]
-
Deng WP, Nickoloff JA 1992 Site-directed mutagenesis of
virtually any plasmid by eliminating a unique site. Anal Biochem 200:8188[Medline]
-
Fuerst TR, Earl PL, Moss B 1987 Use of a hybrid vaccinia
virus-T7 RNA polymerase system for expression of target genes. Mol Cell
Biol 7:25382544[Medline]
-
Campbell MJ 1995 Lipofection reagents prepared by a simple
ethanol injection technique. Biotechniques 18:10271032[Medline]
-
Tovey KC, Oldham KG, Whelan JAM 1974 A simple direct assay for
cyclic AMP in plasma and other biological samples using an improved
competitive protein binding technique. Clin Chim Acta 56:221234[CrossRef][Medline]