You are on page 1of 18

Cecilia G.

Unson
The Rockefeller University,
1230 York Avenue, Molecular Determinants of
Box 294,
New York, NY 10021
Glucagon Receptor Signaling

Abstract: A 29-amino acid polypeptide hormone, glucagon has been one of the most prolific
models in the study of hormone action. The key biologic function of glucagon is to counterbalance
the actions of insulin and maintain a normal level of serum glucose. Diabetes mellitus can thus be
considered a bihormonal disorder with an excess of glucagon contributing to the hyperglycemic
state. The effects of glucagon are mediated by the glucagon receptor, which is itself a prototypical
member of a distinct category called family B receptors within the G protein-coupled superfamily
of seven-helical transmembrane receptors (GPCRs). At the structural level, the peptide ligands of
family B receptors are highly homologous, in particular in the N-terminal region of the molecules.
The mechanism by which highly homologous peptide ligands selectively recognize their receptors
involves distinct molecular interactions that are gradually being elucidated. This review focuses on
structural determinants of the glucagon receptor that are important for its activity with respect to
interaction with its ligand and G proteins. Information about the glucagon receptor is presented
within the context of what is known about other members of the family B GPCRs. © 2002 Wiley
Periodicals, Inc. Biopolymers (Pept Sci) 66: 218 –235, 2002

Keywords: G protein-coupled receptors; glucagon receptor; peptide ligand binding; G protein;


family B GPCR; structure–activity; receptor chimeras; receptor binding model

INTRODUCTION ogous hormones: glucagon, glucagon-like peptide 1


(GLP-1), and glucagon-like peptide 2 (GLP-2).2– 4
The principal role of glucagon is to maintain normal Glucagon is the major product of proglucagon pro-
serum glucose concentration. Glucagon is a 29-amino cessing by prohormone convertase 2 (PC2) in the islet
acid peptide hormone that is produced and secreted in A cells and GLP-1 and GLP-2 are produced in the
the A cells of the islets of Langerhans when glucose intestinal L-cells.5 In addition to a major role in glu-
levels fall below the physiological range. Glucagon cose homeostasis, glucagon is known to have a wide
exerts its effects mostly in the liver, where it coun- range of effects in tissues other than the liver. Gluca-
terbalances the action of insulin by stimulating gly- gon receptor mRNA expression was also found to be
cogenolysis, gluconeogenesis, and ketogenesis, a se- relatively abundant in adipose tissue, kidney, heart,
ries of events that ultimately results in a rise in hepatic spleen, pancreatic islets, ovary, and thymus.6 In the
glucose output.1 Biosynthetic studies have shown that pancreas, glucagon potentiates glucose-induced insu-
glucagon is synthesized as the 180-amino acid pre- lin release from B cells.7,8 Glucagon also has a role in
cursor molecule proglucagon, which is proteolytically free fatty acid metabolism and induces lipolytic re-
processed in specific tissues to produce three homol- sponse in adipocytes.9 Glucagon has been shown to

Correspondence to: C.G. Unson; email: unsonc@mail.


rockefeller.edu
Biopolymers (Peptide Science), Vol. 66, 218 –235 (2002)
© 2002 Wiley Periodicals, Inc.

218
Molecular Determinants of Glucagon Receptor Signaling 219

exert a positive inotropic effect on the heart and which constitutes approximately 1–2% of the total
regulate renal function.10,11 Lower expression levels number of genes in the human genome. A cDNA
of receptor mRNA were detectable in skeletal muscle, encoding the rat glucagon receptor was isolated from
adrenal glands, stomach, small intestine, and brain, rat liver mRNA by an expression cloning strategy.16,17
signifying that novel activities of glucagon have yet to The human glucagon receptor (hGR) cDNA obtained
be determined. from a liver library by a combination of polymerase
The driving force for continued efforts to study chain reaction (PCR) and colony hybridization en-
glucagon lies primarily in the still unresolved problem codes a 477-residue protein that is 80% identical to rat
of diabetes mellitus, which ranks among the most GR in its primary structure.18 Fluorescence in situ
widespread diseases of our time, afflicting 1–2% of hybridization to metaphase chromosome preparations
the population of the Western world. The careful mapped the gene encoding the human glucagon re-
regulation of plasma glucagon and insulin levels is ceptor to chromosome band 17q25.19,20 Analysis of
seriously impaired in patients with either insulin-de- the genomic sequence shows that the coding region
pendent (IDDM) or non-insulin-dependent (NIDDM) spans over 5.5 kb and is interrupted by 12 introns,
diabetes mellitus. A two-hormone abnormality hy- allowing for alternative splicing and receptor hetero-
pothesis has been enlisted to account for the observa- geneity.21 However, because no alternate products
tion that the major metabolic complications of diabe- were detected by RT-PCR analysis, the glucagon re-
tes, impaired glucose tolerance and ketoacidosis, are ceptor expressed in all tissues appears to exist as one
often accompanied by an abnormal increase in the form and derives from the same gene.22,23
level of plasma glucagon relative to insulin.12,13 Thus, The biologic effects of glucagon are initiated by
glucose underutilization due to insulin deficiency and high-affinity binding to its membrane-bound receptor.
overproduction of glucose by elevated physiological Upon activation of a GPCR, a heterotrimeric G pro-
concentrations of glucagon both contribute to hyper- tein transduces the signal from the interior of the cell
glycemia and implicate a role for glucagon in the membrane to activate cellular signaling pathways.
pathogenesis of diabetes. The activated receptor induces a conformation change
Consistent with these observations, a structural in the associated G protein, and, following GTP ex-
analog of glucagon that inhibits glucagon action by change for GDP, the GTP-bound ␣-subunit of the G
competing effectively for receptor binding sites is a protein, G␣GTP, dissociates from the receptor and
reasonable target for drug design as an alternative or from the ␤␥ subunits. Both G␣GTP and the released
adjunct to insulin therapy in the management of dia- ␤␥ dimer can modulate several downstream effector
betic complications. In addition, genetic evidence has pathways. Glucagon receptors activate adenylyl cy-
led to the proposal that a defect in the glucagon clase via the heterotrimeric G protein Gs to produce
receptor that mediates the actions of glucagon might cAMP, which mediates most of glucagon’s cellular
constitute a genetic basis for the disease. A single effects. However, it is now known that glucagon
heterozygous missense mutation in exon 2 of the activates alternative signaling pathways, leading to
glucagon receptor gene that changes a glycine to a intracellular accumulation of additional second mes-
serine (Gly40Ser) was reported to be associated with sengers, inositol triphosphates (IP3) and free Ca⫹2.24,25
late-onset NIDDM in some populations.14,15 Subse- It has been suggested but not proven that the glucagon
quent expression of a cDNA containing a Gly40Ser receptor is coupled to the phosphoinositide signaling
single-point mutation in the extracellular region of pathway via the G protein Gq or G11. Subsequent
work has shown that glucagon-induced increase in
GR produced a mutant receptor with decreased ligand
intracellular [Ca⫹2] results from activation of PKA
binding affinity and attenuated signaling response to
and thus is mediated by an increase in cyclic AMP
glucagon. Thus, a full understanding of the interaction
concentration.26 The generation of these second mes-
between glucagon and its receptor would confirm
sengers triggers other physiological effects via a com-
their involvement in the pathogenesis of diabetes and
plex synergism among effector pathways that remain
might lead to effective therapeutics.
to be sorted out. Emerging evidence show that GPCR
signaling previously thought to be sequential but uni-
directional involves cross-activation and concurrent
regulation between different GPCRs and other path-
GLUCAGON RECEPTOR
ways.
Recently, a stable cell line expressing glucagon
The glucagon receptor (Figure 1) is a member of the receptors activated the mitogen-activated protein ki-
superfamily of G protein-coupled receptors (GPCRs), nase (MAPK) pathway upon glucagon treatment,
220 Unson

FIGURE 1 Schematic representation of the rat glucagon receptor primary and secondary struc-
tures. Amino acid residues are depicted in single-letter code. The extracellular domain consisting of
the amino terminal tail and extracellular loops e1, e2, and e3 is at the top, and the cytoplasmic
domain consisting of the carboxy terminal tail and intracellular loops i1, i2, and i3 is toward the
bottom. The red arrow indicates a putative signal sequence proteolytic site. The four sites of
potential N-linked glycosylation on the N terminus are labeled with asterisks. Six of the cysteines
in the N terminus are conserved in family B GPCRs and are involved in a pattern of three disulfide
bonds shown in orange to link Cys44 and Cys68, Cys59 and Cys101, and Cys82 and Cys122. An
essential disulfide bridge between extracellular loops e1 and e2 is a conserved feature in all GPCRs,
and is depicted here between Cys225 and Cys295. The seven TM ␣-helical segments I to VII are
shown in blue cylinders.

causing a dose-dependent phosphorylation of extra- to be Gs-mediated and involved a concerted action of


cellular signal-regulated protein kinases (ERK1/2).27 glucagon-induced intracellular [Ca2⫹] elevation and
While G protein-coupled receptors lack intrinsic ty- cAMP-dependent protein kinase A. The specific bio-
rosine kinase activity, G proteins can couple GPCRs chemical routes that switch a glucagon-dependent sig-
to MAP kinases, which are key components of a nal from a G protein-dependent pathway to a signal-
signaling pathway that controls cell proliferation.28,29 ing cascade used by receptor tyrosine kinases have not
The transactivation of MAPK by glucagon appeared been determined.27
Molecular Determinants of Glucagon Receptor Signaling 221

GLUCAGON RECEPTOR STRUCTURE that are presumed to form a complex network of three
disulfide bridges. The human glucagon receptor
GPCRs do not share overall sequence homology, but cDNA encodes a 477-aa protein with a putative 27-
significant sequence homology is conserved within amino acid signal peptide that is presumably cleaved
each of the five GPCR subfamilies.30,31 The isolation between A27 and Q28 to yield a mature 450-aa recep-
of the cDNA of rat and human glucagon receptors tor, consisting of a 121-aa extracellular amino-termi-
confirmed that it is a prototype of family B, a unique nal domain, a 404-aa seven-helical core, and a 73-aa
class of receptors within the GPCR superfam- intracellular carboxyl-terminal domain. The conflu-
ily16,18,32,33 (Figure 1). Members of family B GPCRs ence of the extended N-terminus and the three extra-
include receptors for structurally related peptides glu- cellular loops is believed to embody the hormone
cagon-like peptide 134; glucagon-like peptide 235; se- binding site.
cretin36; vasoactive intestinal peptide37,38; gastric inhib-
itory peptide39; growth hormone releasing hormone40;
pituitary adenylyl cyclase-activating peptide41; as well
as receptors for peptides unrelated to glucagon such as RECEPTOR–G PROTEIN INTERACTION
calcitonin42; calcitonin gene-related peptide43; PTH/
PTH-related peptide44,45; and corticotropin-releasing As for all GPCRs, the binding of glucagon to GR is
factor.46 The molecular basis by which these receptors thought to trigger conformational changes in the re-
recognize their peptide ligands and convey the signal ceptor that constitute the common activation mecha-
across the membrane to G protein-coupled effectors nism. These changes might first involve reorganiza-
inside the cell is still largely unknown. tion of the extracellular loops to accommodate the
GPCRs have in common a central core consisting ligand, followed by movement of the TM domains at
of seven membrane-spanning helices connected by the central core of the receptor. It is the modification
alternating extracellular and intracellular loops, along of interhelical constraints comprised of electrostatic
with an amino-terminal extracellular domain and an and hydrophobic interactions that subsequently affect
intracellular carboxyl terminal extension. The extra- the cytoplasmic side of the receptor and enable spe-
cellular domains typically contain consensus sites for cific regions of the intracellular loops to interact with
asparagine-linked glycosylation. The transmembrane G proteins. Among family A GPCRs, one major in-
(TM) domains form a seven-helical bundle oriented tramolecular constraint was found to be interactions
roughly orthogonal to the plane of the membrane in a surrounding the highly conserved E(D)RY(W) motif.
counterclockwise configuration. A characteristic di- In the crystal structure of rhodopsin, E134R135Y136 are
sulfide bridge that connects extracellular loops 1 and involved in hydrogen bonds and a salt bridge and a
2 is found in all GPCRs and is probably important for reorientation of interactions in this region occurs dur-
packing and stabilization of the central core. This ing activation.47 A closer look at family B GPCRs
basic arrangement was validated in the 2.8-Å resolu- reveals that the DRY counterpart could be furnished
tion structure of rhodopsin, the first 3-D structure of a by a conserved R (R174 in GR) in the intracellular end
GPCR.47,48 The most conserved residues among of TM II predicted to be at the same level in the
GPCRs are located within the TM domains and rep- membrane as the R in DRY.49 The E(D) function is
resent essential structural determinants of receptor predicted to align with an ExxY (E246 in GR) motif in
structure and function. For all GPCRs, receptor acti- the intracellular end of TM III.49 The conserved res-
vation is associated with a change in conformation idues Y239 and L240 of human VPAC1 receptor are
within this core domain. Other domains are not as predicted to be at the same location as the asparagine
conserved and sequence variations in the extracellular and arginine in the “DRY” motif in the rhodopsin
and intracelullar domains bestow specific properties family of GPCRs. The VPAC1 receptor mutants
to each receptor to regulate molecular events leading Y239A had a moderate and L240A a pronounced
to this change. impaired ability to produce cAMP, suggesting that the
The most conserved family B receptors do not effect of these mutants is a perturbation of the G
contain any of the hallmarks that characterize family protein interaction.50 Thus, the different chemical
A receptors. Notably the Asp-Arg-Tyr (DRY) motif is properties of the hydrophobic YL motif and charged
absent and the prolines conserved among family A DR(Y) motif could be a crucial difference between
receptors are distinct from those conserved in family the family B GPCRs and the rhodopsin family with
B receptors. The most prominent feature of family B respect to receptor activation and G protein coupling.
GPCRs is a long N-terminal domain of 100 –200 The general topography of GPCRs necessitates
amino acids, which contains six conserved cysteines that receptor G protein-coupling domains lie within
222 Unson

sequences of the cytoplasmic domains, specifically way, or, more likely, that glucagon receptors were
regions close to the plasma membrane at the ends of activating two signaling pathways via different G
the TM helices. It is well known that the second (i2) protein species.24 Subsequent mutagenesis of the cy-
and third (i3) intracellular loops of GPCRs are closely toplasmic domains of GR demonstrated that the sec-
involved in coupling to G proteins and in determining ond and third intracellular loops, i2 and i3, synergis-
specificity toward a particular G protein class. The tically contribute determinants required for activation
change in conformation of the helical bundle in gen- of Gs.56 Deletion or replacement of residues from each
eral affects the disposition of the i2 and i3 loops, both of either the i2 or i3 loops resulted in a marked
of which are directly connected to TMIII and TMVI. attenuation of the ability of glucagon to activate ad-
It has been proposed that the C-terminus of the G enylyl cyclase.56,57 Replacement of both the i2 and i3
protein ␣-subunit binds in a pocket formed by these loops of GR with a sequence from the i1 loop of the
intracellular loops. The residues in the first intracel- unrelated D4 dopamine receptor in the receptor mu-
lular loop of family B receptors are highly conserved, tant G23D1R resulted in the abrogation of all gluca-
which may indicate an essential role for these residues gon-dependent activities.27,56 Consequently, GR ap-
in protein folding and in surface expression of the pears to regulate its primary signal transduction path-
receptors. In GR, the long C-terminal tail does not ways by faithfully coupling only to Gs.
contribute significantly to ligand binding or G protein GR is known to exhibit both high and low binding
coupling. Truncation of variable portions of the affinities for glucagons, which is thought to be due to
COOH terminal of GR had no effect on binding and a heterogeneous population of receptors that are mod-
glucagon-stimulated cAMP accumulation, or Ca2⫹ ulated by association with Gs, even prior to ligand
mobilization.51,52 Indeed, the absence of the tail binding.58 – 60 Variations in the relative densities of
slightly increased receptor affinity for glucagon, per- receptors and G proteins in turn influence effector
haps by making the intracellular loops more accessi- coupling and the efficacy of activation.21 Two pre-
ble for coupling to G proteins.51 However, the C- dominant splice variants of G␣s, a short and a long
terminal tail appears to be involved in mechanisms form (G␣s-S and G␣s-L) resulting from differential
that regulate the functional level and activity of GR. splicing of a single precursor RNA, exist in most
Glucagon-induced phosphorylation of serine residues cells, but their relative proportions vary.61– 63 To as-
in the cytoplasmic C-terminal tail is crucial for recep- sess functional differences in the interactions of the
tor internalization.53 GR with the two predominant splice variants of G␣s,
All members of the glucagon family of receptors GR was covalently linked to the short and the long
couple to Gs to activate adenylyl cyclase and elevate forms, G␣s-S and G␣s-L, to produce the fusion pro-
intracellular cAMP concentration. At high nonphysi- teins GR-G␣s-S and GR-G␣s-L.64 GR-G␣s-S bound
ological concentration (⬎10⫺5), glucagon inhibits the glucagon with an affinity similar to that of GR, while
accumulation of cAMP induced by isoproterenol, an GR-G␣s-L showed a 10-fold higher affinity for glu-
effect that is attenuated by pertussis toxin, suggesting cagon. Both GR-G␣s-L and GR-G␣s-S were consti-
involvement of Gi.54 Glucagon-stimulated GR also tutively active, causing elevated basal levels of cAMP
results in mobilization of intracellular Ca⫹2 stores and even in the absence of glucagon. The mutant GR that
the breakdown of inositol lipids to produce inositol failed to activate Gs (G23D1R) was fused to G␣s-L.
phosphates and diacylglycerol. Recently, it was G23D1R-G␣s-L bound glucagon with high affinity
shown that glucagon activates the MAP kinase path- but failed to elevate cAMP levels, suggesting that the
way via a cAMP-dependent pathway in a cell line mechanisms of GR-mediated G␣s-L activation and
stably expressing GR.27 In most GPCRs, activation of G␣s-L-induced high-affinity glucagon binding are in-
phospholipse C and the activation of the MAP kinases dependent.64 Both GR-G␣s-S and GR-G␣s-L bound
are mediated by Gq or G11. While it is well established the glucagon antagonist desHis1[Nle9Ala11Ala16]-
that GR couples to Gs to generate cAMP, the involve- glucagon amide with similar affinities as GR.64 The
ment of Gq with activated GR is less clear. In earlier antagonist displayed partial agonist activity with GR-
studies, a glucagon antagonist, (1-N-alpha- trinitro- G␣s-L, but not with GR-G␣s-S, proving that the par-
phenylhistidine,12-homoarginine)glucagon (TH-glu- tial agonist activity of the antagonist observed in
cagon), which does not activate adenylyl cyclase, was intact cells appears to be due to GRs coupled to
shown to stimulate the production of inositol phos- G␣s-L. That G␣s-S and G␣s-L isoforms appear to
phates.24,55 These observations initially suggested ei- interact differently with GR has physiological impli-
ther that hepatocytes possessed two distinct receptors cations and emphasizes that the efficacy and potency
for glucagon, one coupled to adenylyl cyclase activa- of ligands are regulated not only by the receptor but
tion and another coupled to the phospholipase C path- also by the type and relative amounts of G proteins
Molecular Determinants of Glucagon Receptor Signaling 223

FIGURE 2 Comparison of sequences of peptide ligands of the glucagon receptor family. The
most invariant residues are located in the N-terminal part of the peptides. Based on homology
between both receptors and ligands, the conformation of receptor-bound ligand may be similar.

accessible to the receptor molecules. Receptors asso- influence either binding affinity or activation and af-
ciating with different isoforms of G␣s or other G forded some insight into the molecular requirements
proteins may vary in their ability to isomerize to the of the receptor for optimum recognition and interac-
activated state and thus differ in their affinity for tion. The negatively charged side-chain of the aspartic
agonists and antagonists and provoke a graded range acid residue at position 9 is absolutely required for
of potencies. Some endogenous GPCRs exhibit de- receptor activation and less for binding, so that re-
tectable activity even in the absence of agonist, an placement with any other amino acid resulted in a
occurrence frequently associated with pathogenic glucagon antagonist.68,69 The N-terminal histidine,
states. This can be accounted for by mutations in the which is strictly conserved within the glucagon fam-
cytoplasmic domains that facilitate isomerization of ily, furnishes determinants of both binding and activ-
the receptor to the active conformation. There are no ity of the hormone.70 –72 Aspartic acids at positions 15
known constitutively active GR mutants except for and 21 also regulate the binding or activity function of
one report in which His178 in the i1 loop, which is the hormone.69,73 The positively charged groups at
strictly conserved in family B receptors, was replaced positions 12, 17, and 18 stabilize the binding interac-
with an arginine.65 The corresponding mutation in the tion to ensure maximum potency.74 –76 In addition, the
PTH-PTHrP and VIP1 receptors exhibited constitu- crystal structure of the peptide superagonist [Lys17,18,
tive activation of Gs.66,67 In the PTH-PTHrP receptor, Glu21]glucagon amide and cyclic analogs of glucagon
this mutation was identified in a patient with Jansen- suggest that an intramolecular salt bridge between
type metaphyseal chondrodysplasia characterized by side-chains of aspartic acid and lysine may be a fea-
the severe ligand-independent hypercalcemia and hy- ture of the active conformation of the hormone.75,77
pophosphatemia and a rare form of dwarfism.66 Con- These studies confirmed that even the flexible middle
stitutively active GPCRs allow the study of ligands portion of the peptide ligand between positions 12 to
that elicit inverse agonism or lowering of the basal 21 is important for recognition and transduction. In
activity. Thus, the constitutive activity of GR-G␣s-L addition to electrostatic interactions, a topographical
will be useful in assessing novel glucagon analogs for requirement for the proper stacking of hydrophobic
inverse agonist activity. residues Phe6 and Tyr10,13 in glucagon has also been
proposed.78 – 80 Collectively, structure–function stud-
ies of glucagon confirmed that virtually the entire
LIGAND–RECEPTOR INTERACTION hormone is necessary for the expression of full hor-
monal activity.
The peptide ligands of family B receptors are highly
homologous to each other, yet each will activate a
specific receptor to elicit the appropriate physiological HORMONE BINDING SITE
response (Figure 2). Despite the initial suggestion that
the C-terminal part of the peptide is primarily in- While extensive structure–activity studies of glucagon
volved in receptor recognition and binding while the have identified specific residues in the peptide ligand
N-terminal half is important for transduction, the gen- that play a role in binding and transduction, the com-
eral picture that has emerged is that the active phar- plementary recognition site that is the “switch” on the
macophore is dispersed throughout the glucagon mol- receptor has yet to be deduced. Members of the glu-
ecule. Numerous structure–function studies of gluca- cagon family of peptide hormones are 50% identical
gon have singled out strategic positions throughout and more than 70% homologous (Figure 2). The
the peptide ligand in which the amino acid residues structural basis by which the receptors discriminate
224 Unson

among similar ligand structures most likely lies in its ability to bind GLP-1. In particular, the replace-
structural subtleties within the receptors themselves. ment of W39 in GLP-1R led to loss of receptor func-
Structure–function studies of related family B recep- tion.85,99 More recently, a nonpeptide ligand was
tors indicate that the peptide ligand binding site is shown to bind to the human GLP-1R and inhibit
discontinuous and consists of contact points from the GLP-1-induced cAMP production by targetting
extended N-terminus and the three extracellular loops W.33,100 Most of these tryptophan residues are con-
connecting the TM helices.81– 85 This observation was served in both rat and human GR and undoubtedly
further corroborated by cross-linking experiments that play a similar role in the binding of glucagon to GR.
have mapped the peptide ligand binding site to the The N-terminal domains of the human parathyroid
extracellular domain of the receptors.86 –90 hormone, PACAP, GLP-1, and CRF type 1 receptors
The putative 121-residue N-terminal domain of have been expressed, correctly refolded, and puri-
GR contains six cysteine residues, which are strictly fied.84,91–93,101 The soluble proteins corresponding to
conserved within family B members and are pre- the receptor N-terminus manifested significant ligand
sumed to form three specific disulfide bonds. The binding activity. In particular, the CRF-R1 N-terminal
precise disulfide connectivity was determined for the fragment bound a CRF agonist and antagonist just as
N-terminal fragment of human parathyroid hormone avidly as intact wild-type CRF-R1, indicating that the
receptor (PTHR), for the corticotropin releasing factor major determinants for high-affinity binding of CRF
(CRF) type 1 receptor, and, most recently, for GLP- are found in this domain.92,102 Nonetheless, the bind-
1R,91–93 suggesting a conserved structural pattern in ing affinities of the isolated N-terminal fragments of
the N-terminal domain of family B receptors. Treat- PTHR, PACAPR, and GLP-1R for their ligands were
ment with cell non-permeabilizing reducing reagents sufficiently weak to infer that the N-terminal domain
or mutagenesis of the cysteine residues to alanine had alone cannot account for the binding affinity of an
adverse effects on receptor binding, validating the intact receptor.84,91,93,101 In addition, when the entire
critical importance of the complex, highly folded, N-terminal domains of the glucagon and the GLP-1
disulfide arrangement to family B receptor func- receptors were exchanged, glucagon or GLP-1 bound
tion.94,95 Based on the known disulfide-linking pattern to neither the GR-GLP-1R nor the GLP-1R-GR chi-
of these family B receptors, the analogous disulfide mera.103 The accepted view that has emerged from
bonds in rat GR can be predicted to occur between these studies is that the N-terminus is an important
cysteines 44 and 68, 59 and 101, and 82 and 122 part of the peptide binding pocket but is not adequate,
(Figure 1). requiring supplementary contributions from residues
Mutagenesis studies of family B GPCRs have located in the extracellular loops and in the upper
shown direct involvement of the N-terminal domain portion of the TM helices close to the intradiscal
in peptide ligand binding. A GR mutant with a 96- membrane bilayer interface.
residue deletion from the N-terminal tail, including 5 The partial boundaries of the glucagon binding site
of the 6 conserved cysteine residues, was expressed on the glucagon receptor were demarcated using anti-
on the cell surface but failed to bind glucagon, em- GR antibodies (Figure 3). Antibodies DK-12 and
phasizing that the integrity of the tertiary structure KD-14 generated against synthetic peptides with
was crucial for high-affinity glucagon binding.51 Sin- amino acid sequences corresponding to residues 126 –
gle point mutations throughout the N-terminal tail of 137 of the N-terminal tail and 206 –219 of the first
family B receptors have resulted in either complete extracellular loop, respectively, competed with gluca-
loss of binding or an attenuation of the activity func- gon for receptor binding binding sites.104 In contrast,
tion. Photoaffinity cross-linking and mutational data ST-18 antibody raised against an 18-residue peptide
showed that residues at the extreme amino-terminus from the C-terminal end of GR located in the cyto-
of the PTH receptor participate in ligand binding.96 In plasmic domains had no effect on the binding of
GR, single amino acid replacements of D64 resulted in glucagon. DK-12 and KD-14 also behaved like glu-
GR mutants that failed to bind glucagon.97 This site in cagon antagonists, preventing glucagon-dependent
the related growth hormone releasing factor receptor activation of adenylyl cyclase.104 Although the results
was shown to be responsible for the little mouse (lit) indicate that the antibodies bind to the glucagon bind-
genetic defect that results in mice of small size with ing site in GR, the exact nature of the competition
hypoplastic pituitary glands.98 A G40S GR mutant cannot be specified. It is possible that antibody bind-
exhibited attenuated adenylyl cyclase activity and has ing causes an effect from a distance and sterically
been linked to late-onset NIDDM in some popula- displaces bound glucagon. The peptide epitope may
tions.14,15 Five out of six tryptophan residues in the actually be part of the binding interface or may be a
N-terminal domain of GLP-1 receptor are essential for sequence spatially close to the binding site. Nonethe-
Molecular Determinants of Glucagon Receptor Signaling 225

FIGURE 3 Schematic representation of the GR showing the locations of the peptide segments that
were used to generate polyclonal antibodies to GR. PR-15 and DK-12 were raised against amino
acid residues 103–117 and 126 –137 of the N terminus, respectively. KD-14 was raised against
residues 206 –219 of the e1 loop. ST-18 was raised against C-terminal residues 468 – 485. Antibod-
ies DK-12 and KD-14 competed with glucagon for GR binding and inhibited glucagon-dependent
receptor activation. A series of chimeric receptors in which the corresponding peptide epitopes were
each replaced with their counterparts from GLP-1R and SR indicated that the sequences played a
role in GR binding and activation.

less, the antibody studies were consistent with the dues 103–117 and 126 –137 of the N-terminus and
idea that the glucagon binding domain involves part 206 –219 and 220 –231 of the first extracellular loop
of the N-terminal tail close to the junction at which were each replaced with the corresponding sequences
the first TM domain begins and includes the first from the GLP-1 receptor or secretin receptor (SR).106
extracellular loop. Evidence that polyclonal antibodies against peptides
Chimeric constructs of structurally related recep- representing sequences 126 –137 and 206 –219 effec-
tors have been used to correlate a specific structural tively blocked glucagon binding strongly indicated
domain with function and identify determinants of that these domains were involved in ligand bind-
ligand binding affinity and selectivity. A chimeric ing.104 Receptors for GLP-1 and secretin are the clos-
receptor in which residues 29 –32 of the N-terminus of est relatives of GR within family B GPCRs, sharing
the GLP-1 receptor were replaced with the corre- 50% overall identity in their primary structures. In-
sponding amino acids from GR showed a 10-fold terestingly, the substituted sequences represented
decrease in affinity for GLP-1 and an increase in nonconserved areas in these receptors and implied
affinity for glucagon, representing a 50-fold decrease that these domains may also embody determinants of
in the selectivity of this receptor for its natural li- ligand selectivity. The chimeric receptor study
gand.105 A composite of findings utilizing glucagon/ showed that these segments indeed played a role in
GLP-1 receptor chimeras demonstrated that the glu- glucagon binding.106 Segment substitutions within the
cagon binding pocket is a discontinuous domain that first extracellular loop led to a greater detrimental
encompasses the membrane-proximal region of the effect on both binding and activity than either replace-
amino-terminal extension and the first extracellular ment of residues 103–117 or 126 –137 in the mem-
loop, further corroborating the GR antibody studies.81 brane proximal region of the N-terminal extension,
In addition, the chimeric receptor studies revealed that supporting the notion that the N-terminal domain of
glucagon binding was sensitive to replacement of TM GR is not sufficient for glucagon binding. Replace-
helices III, IV, and VI with the corresponding se- ment with e1 sequence from SR had a more detrimen-
quences from GLP-1R, indicating that amino acid tal effect than a sequence from GLP-1R, which sug-
residues within these TM domains, in particular those gested that glucagon recognized structural patterns in
located close to the lipid and aqueous interphase, are the sequence from the e1 loop of GLP-1R that were
in contact with ligand during the binding event.81 not contained in the analogous SR sequence.
A receptor chimeric study was conducted to exam- A cross-chimeric analysis was employed to ad-
ine the role of these domains in greater detail. Resi- dress the possibility that the homologous peptide hor-
226 Unson

mones GLP-1 and glucagon may interact with the to ligand binding affinity but may instead provide
same regions in their respective receptors. A peptide molecular contacts that trigger activation.106
hybrid comprising the N-terminal 1–14 residues of Single substitution mutagenesis of the e1 loop fur-
glucagon linked to the C-terminal 15–31 residues of ther revealed a critical requirement for a positive
GLP-1 was shown to possess the chemical features charge at the extracellular border of TM helix II at the
necessary for selective recognition by both recep- junction with the e1 loop. Replacement of R202 with a
tors.107 The e1 loop chimera bearing a sequence from negatively charged aspartic acid in R202D resulted in
GLP-1R exhibited greater potency toward the GLP- a complete loss of receptor function.106 Although a
1/glucagon chimera relative to glucagons, suggesting positively charged group is required to define the
both GLP-1 and glucagon associate with amino acid topology of a TM helix within the bilayer, it is also
residues in the e1 loop of their respective receptors possible that by replacing R202 an important associa-
during binding and activation.106 This observation is tion with specific residues of the ligand was lost. The
consistent with the finding that residues located at the importance of amino acid residues H1, S8, D9, D15,
N-terminal half of glucagon predominantly determine S16, and D21 of glucagon for either binding or activa-
selective recognition of GR. Conversely, the selective tion has been established.69,70,72,112,113 Any of these
binding epitope of GLP-1 is primarily located near the amino acids residues could contribute to glucagon
C-terminus. binding energy through ionic or hydrogen bond for-
Residues 206 –219 of the first extracellular loop are mation with R202. In vasoactive intestinal peptide
most likely situated in the central portion of the e1 (VIP), D3 was shown to contact an arginine and a
loop. Single substitution mutagenesis of the charged lysine residue at analogous positions at the extracel-
residues within the 206 –219 stretch failed to mimic lular end of TM helix II of the VIP receptor.114,115
the phenotype of the entire 206 –219 amino acid sub- Alternatively, arginines can interact with aromatic
side-chains of phenylalanine, tyrosine, or tryptophan
stitution, suggesting that the effect may be conforma-
through cation ␲-orbital interaction. This potent non-
tional or hydrophobic in nature.106 In the human
covalent binding force occurs at or near the surface of
PTH1 receptor (hPTHR), a leucine (L261) located in
proteins and contributes to ligand–receptor interac-
the central helical portion of the e1 loop has been
tion.116 –118
identified as the contact site for K27 of PTH(1-
34).108,109 Replacement of the adjacent 220 –231 res-
idues that extend to the border of TM helix III of GR
BINDING MODEL FOR FAMILY B
did not seriously impair high-affinity binding to glu-
GPCRS
cagon. The central portion of the e1 loop is important
in glucagon binding but residues 220 –231 near helix The ultimate goal of the different approaches to study
III are not. A conserved cysteine in this segment the molecular mechanism of ligand-specific recogni-
(C225) is likely to be involved in a disulfide bridge tion, binding, and receptor activation is to determine
with C295 of the second extracellular loop (e2 loop). A the structure and conformation of the peptide ligand
disulfide bond joining the e1 and e2 loops is a struc- while bound within a ligand–receptor complex and
tural feature that is strictly conserved in both family A generate a template for rational drug design. Difficul-
and family B GPCRs.30 In the crystal structure of ties in the expression and solubilization of family B
rhodopsin, a disulfide bridge between C110 and C187 receptors have not provided practical amounts of pu-
draws the e2 loop away from the surface and extends rified receptors required for structural studies. Re-
it over a crevice in the seven-helix bundle.47,110,111 cently, the conformation of an analog pituitary adeny-
The analogous disulfide bridge in GR that occurs lyl cyclase activating polypeptide PACAP(1-21),
between C225 of the e1 loop and C295 of the e2 loop bound to a PACAP–specific receptor, was determined
may restrain the 220 –231 segment of the e2 loop and by nuclear magnetic resonance (NMR) spectroscopy.119
pull it toward the closely packed region at the top of This is the first structure determination of a ligand
the helices, an area not easily accessible to a peptide peptide bound to a GPCR, which provided a unique
ligand the size of glucagon. Indeed, a receptor chi- insight on the structural basis for how a receptor
mera in which residues 220 –231 were replaced re- recognizes its ligand. The PACAP receptor was ex-
tained glucagon binding affinities similar to that of pressed in Sf9 insect cells, solubilized in digitonin,
wild-type GR but exhibited a disproportionate loss in and purified by affinity chromatography.120 The struc-
the ability to transduce the signal. A disproportionate ture revealed a unique ␤-coil structure formed by
change in agonist potency vs. binding affinity indi- residues 3–7 next to an N-terminal extended tail. The
cates that the 220 –231 region contributes less directly ␤-coil creates a hydrophobic cluster made up of the
Molecular Determinants of Glucagon Receptor Signaling 227

FIGURE 4 Working model for the binding and activation of family B GPCRs. The mechanism
is proposed to involve initial association of the peptide with the membrane, followed by recognition
and capture of the C terminus of the ligand by the N-terminus of the receptor, which reins in the
N-terminus of the peptide toward the extracellular loops and the helical bundle. Both ligand and
receptor undergo conformational changes that subsequently lead to activation.

side-chains of I5, F6, and Y10, together with an R14. A tide by the extracellular N-terminus of the receptor
G4 is necessary for the formation of a type II’ ␤-turn, (Figure 4). Results from structure–function studies of
which is important in maintaining the relative orien- C-terminally truncated secretin and VIP molecules
tation of the hydrophobic patch and the extreme N- suggest that the C-terminal end of secretin and VIP
terminus. In contrast, the C-terminal region of recep- interacts with the N-terminal domains of their recep-
tor-bound PCAP21 forms an ␣-helix similar to its tors.124 Thus, the receptor N-terminus may initially
structure in micelles.119 At the structural level, pep- associate with the C-terminus of the peptides. Mem-
tide ligands of family B GPCRs are highly homolo- bers of the glucagon family are most divergent at the
gous, in particular in the N-terminal domain of the C-terminus. In addition, while the three disulfide
molecules where the characteristic ␤-coil structure is bonds establish a common overall tertiary fold for all
induced upon receptor binding. Based on the homol- family B GPCRs, many of the intervening residues
ogy between both receptors and ligands, the structural are not conserved and may function as the first line of
features that were found in receptor-bound PACAP21 selectivity filters that restrict access of inappropriate
may parallel those found in ligands belonging to the ligands to an activation site. The ligand now brought
glucagon branch of family B GPCRs. Indeed, the within the receptor molecule interacts more avidly
NMR structure of glucagon in a micelle–water inter- with additional residues in the central core of the
face showed that residues 17–29 of glucagon form an receptor, leading to formation of a high-affinity com-
amphiphilic helix.121 Synthetic analogs containing plex and ultimately to activation (Figure 4).103 The
amino acid replacements that increased the ␣-helical working model is bolstered further by the NMR struc-
character of the C-terminus displayed improved bind- ture of PACAP, which advocates that binding of the
ing affinities.122,123 Structure–activity studies have peptide ligand to its receptor is kinetically favored
proposed a role for a hydrophobic patch in glucagon when it occurs in two steps. The ligand first associates
and emphasized that proper stacking of F6, Y10, and nonspecifically with the membrane surface, establish-
Y13 was important for binding and transduction.78 – 80 ing an ␣-helical structure at the peptide C-terminus,
More importantly, the fact that H1, G4, F6, T7, and Y10 then diffuses two-dimensionally until it finds its re-
are invariant in the N-terminal part of both PACAP ceptor. The characteristic N-terminal ␤-coil is in-
and glucagon strongly implies that glucagon may duced upon binding, which enables interaction with
behave similarly in the presence of its receptor. residues that drives the receptor activation process.
Based on a composite of structure–function anal- Extensive studies of the PTH receptor system have
yses of single replacement and chimeric receptor mu- also corroborated the two-site hypothesis of ligand–
tants as well as analogs of the peptide ligands, tenta- receptor interaction.125–127 Moreover, the combined
tive models have been proposed for the binding of functional and photoaffinity labeling data have estab-
family B receptors to their peptide ligands.103,108 One lished the relative spatial orientation of PTH(1-34)
model proposes a sequential process that involves when bound to the PTH receptor. The N-terminal
low-affinity association and initial capture of the pep- residue in PTH(1-34) cross-links to M425 located in
228 Unson

FIGURE 5 Schematic of a model for the interaction of glucagon with GR. Glucagon forms an
␣-helix at the C-terminal region upon association with the membrane bilayer, and initially engages
sequences involving residues 103–117 and 126 –137 of the N-terminal domain of GR. The N-
terminus of the peptide subsequently interacts with a predominantly ionic binding pocket involving
acidic residues from 206 –219 of the e1 loop and is positioned for additional interaction with sites
that lead to receptor activation.

TMVI of PTH receptor.128 K13 contacts a site close to quired several thousand-fold more bovine PTH or
R186 at the junction of the N-terminal extracellular human PTH(1-34) than glucagon. However, the PTH
domain and TMI of PTHR, and K27 cross-links to peptides were more effective than secretin, which up
L261 in the e1 loop of PTHR.90,108,129 These findings to a concentration of 10⫺5M displayed no ability to
further support a model in which the C-terminal por- displace [125I]iodoglucagon, despite the fact that se-
tion of PTH(1-34) initially interacts with the large cretin is homologous with glucagon while PTH is not.
extracellular N-terminal domain close to the junction Circular dichroism studies showed that in the pres-
with TMI. This facilitates a second interaction be- ence of 3 mM SDS glucagon and hPTH(1-34) have
tween the N-terminal region of the ligand with the similar secondary structure, which indicated that re-
juxtamembrane domain at the top of the seven-helix ceptors can recognize gross conformational features
bundle forming an interface with the e1 loop and of a peptide hormone as well as a specific amino acid
making contact with the e3 loop. Computer modeling sequence.131
of this topology shows the active conformation to be Unlike in the PTH system, specific contact sites
a closed U-shaped hairpin structure with the N-termi- between glucagon and GR have not been identified. A
nus of the ligand facing the extracellular loops.126,130 recent report of a mutational analysis of GR suggested
The PTH receptor belongs to family B GPCRs but is that Q3 of glucagon binds close to a region at the top
not a close relative of GR. Interestingly, receptor of TM II, consistent with the findings on the R202D
binding assays demonstrated that bovine parathyroid GR mutant.106,132 Nevertheless, structure–activity
hormone (PTH) and human PTH(1-34) can displace studies of glucagon analogs and GR mutants are also
[125I]iodoglucagon from GR in rat liver plasma mem- consistent with the two-site models for the binding of
branes.131 The displacement of [125I]iodoglucagon re- peptide ligands like PTH(1-34) to family B receptors.
Molecular Determinants of Glucagon Receptor Signaling 229

FIGURE 6 The organization of the genes that have been sequenced from vertebrates are shown
for the glucagon superfamily members. The structural organization of the genes is similar and
ecodes a signal peptide (SP), one to three bioactive peptides, and a C-terminal peptide. Exons are
depicted as boxes and the introns as lines. Glucagon, GRF, GIP, and secretin genes evolved from
the same line of evolution because all lack an exon between the exon coding for the signal peptide
and bioactive peptide. The glucagon gene is unique in that it encodes three peptides— glucagon,
GLP-1, and GLP-2—suggesting that there were exon duplications (adapted from Ref. 135).

It is in general accepted that the N-terminus of glu- conserved members are PACAP and glucagon, which
cagon, which contains both H1 and D9 residues, is the may be the ancestral molecules.135 A gene encoding
principal region responsible for receptor activa- PACAP was isolated from a protochordate, suggest-
tion.69,70 Indeed, a receptor mutant in which the N- ing that the origin of the ancestral family goes as far
terminal domain of GR is replaced by the first 15 back as the invertebrates.136 The organization of the
residues of glucagon displayed a significant level of known genes for the glucagon family is similar (Fig-
constitutive activity (Unson, unpublished results). ure 6). Each gene encodes a signal peptide, an N-
Constitutive activation by a similarly tethered ligand– terminal peptide, bioactive peptides, and a C-terminal
receptor chimera has recently been demonstrated for peptide. Glucagon, GRF, GIP, and secretin genes lack
the PTH and corticotropin-releasing factor recep- an exon between the exons encoding the signal pep-
tors.133,134 This observation further supports a model tide and the bioactive peptide. Evidence to date indi-
in which the C-terminus of the ligand initially engages
cates that the glucagon gene family arose from exon
the N-terminus of the receptor and subsequently
duplication of a single ancestral gene that continued to
draws the N-terminus of the peptide toward the e1
diverge. These gene duplications are likely to have
loop and the body of the receptor, where additional
occurred around 400 –500 million years ago prior to
favorable interactions provide the stabilization energy
the evolution of vertebrates.137 The receptors for these
that leads to activation (Figure 5).106 Whether the bio-
active conformation of receptor-bound glucagon is ex- ligands are structurally related and similarly origi-
tended (Figure 5) or folded into a U-hairpin (Figure 4) as nated by gene duplication events. It is reasonable to
has been postulated for PTH ultimately will have to be speculate that the glucagon family of receptors may
determined by NMR analysis or X-ray crystallography. also have evolved from a common ancestral gene in
invertebrates, although no receptors have been iso-
lated. One theory surmises that the genes for PACAP
MOLECULAR EVOLUTION OF THE and glucagon receptors are derived from protochor-
GLUCAGON RECEPTOR date (invertebrate) ancestral genes that led to the
vertebrate forms of the receptors. Early in chordate
The glucagon subfamily includes nine homologous evolution, two gene duplications might have led to
polypeptide ligands. The most ancient and tightly four copies of each gene that encoded a family B
230 Unson

receptor, followed by structural alterations that might linotropic function in mammals.144,146 Recent isola-
have resulted in eight receptor types.135 tion and structure function studies of a GLP-1 recep-
The mammalian proglucagon gene is unique in that tor from goldfish Carassius auratus demonstrated that
it encodes three biologically active peptides: gluca- it responded to goldfish glucagon, human glucagon,
gon, glucagon-like pepide 1 (GLP-1), and glucagon- and human GLP-1, which are structurally different
like pepide 2 (GLP-2). Each of these functionally peptides (S. Mojsov, personal communication). This
distinct peptides exerts its actions through a specific was evidence that the goldfish receptor was more
and unique but structurally related receptor.16,34,35 accommodating than its mammalian counterpart. Ear-
Glucagon is functionally and structurally conserved lier characterization of frog GRs showed varying se-
across vertebrate species, although some of its activ- lectivities toward mammalian and fish gluca-
ities have diversified during evolution. The tight con- gons.138,144 Deviations in glucagon sequences are
servation of the primary structure of glucagon found in teleost fishes, suggesting that GRs in species
throughout most vertebrates suggests a strong evolu- that existed before frogs would have different ligand
tionary pressure to preserve the molecule, consistent recognition specificities (Figure 7). The observations
with its pivotal role in the regulation of metabo- led to the speculation that evolutionary pressures
lism.138 Two proglucagon cDNAs were characterized manifested at the time of the emergence of the frogs
from the intestine of the sea lamprey Petromyzon may have necessitated mutations in the sequences of
marinus.139 As in other vertebrates, sea lamprey pro- the ligands and the recognition domains of the recep-
glucagon genes encode three related sequences— glu- tors.138 These mutations directed more stringent rec-
cagon and GLP-1 and 2—which is an indication that ognition requirements that were maintained through-
all three glucagon-like sequences existed prior to the out the evolution of the mammalian species and re-
divergence of jawed and jawless vertebrates. Esti- sulted in highly specific receptors. Functional
mates of the rates of evolution for the glucagon-like characterization of GRs from other species is needed
sequences suggest that glucagon had the most ancient to fully understand the molecular evolution of verte-
origin and diverged nearly 1 billion years ago, while brate glucagons and their receptors and the structural
GLP-1 and GLP-2 diverged from each other by an determinants that dictate specificity within the gluca-
exon duplication event about 700 million years ago. gon receptor family.
Whether the origin of the receptors for these pep-
tides parallel their ligands is not known. While the
amino acid sequence of many mammalian glucagons, CONCLUSION AND PERSPECTIVES
bird, several species of frogs, bony fish, cartilaginous
fish, and jawless fish are known, to date, only a few The glucagon signaling system has been one of the
nonmammalian glucagon receptors have been charac- most prolific models in the study of peptide hormone
terized.3,138,140 –144 However, based on the sequences action. About 30 years ago, Rodbell first characterized
known, a phylogenetic analysis using both parsimony receptors for glucagon in hepatocytes and demon-
and distance methods reveals that the evolutionary strated that glucagon-dependent stimulation of adeny-
history of the receptors for proglucagon-derived pep- lyl cylcase had a GTP requirement, an observation
tides differs from that of the ligands. These observa- that gave rise to the field of signal transduction me-
tions suggest that the diversification of the peptides diated by guanine nucleotide binding proteins (G pro-
encoded by the proglucagon gene and the receptors teins) and the classification of a superfamily of recep-
for these peptides occurred independently and that the tors coupled to G proteins. Considerable structure and
hormones or their receptors have been recruited for activity studies of GPCRs have since been carried out
new functions.144 The difference in the phylogenies that have provided a general picture of GPCR struc-
for receptors and ligands may indicate that the spec- ture and improved the understanding of receptor func-
ificity of the receptor–ligand pair evolved much later tion. To date, only the photoreceptor rhodopsin, a
than the peptides or receptors themselves. The pep- prototype of family A GPCRs, has its structure deter-
tides and receptors originally may have had a gluca- mined to atomic resolution. Because there is consid-
gon-like function and the different physiological func- erable divergence in the way GPCR ligands interact
tions evolved only much later. For example, while the with their receptors, the rhodopsin crystal structure
physiological function of glucagon remained un- may not be an adequate homology template on which
changed throughout vertebrate evolution, the function to base predictions of how peptide ligands of family B
of GLP-1 has changed.145 In fish, glucagon and interact with their receptors. Nevertheless, it is a good
GLP-1 have overlapping glucagon-like activity that vantage point from which information from structure–
counteracts insulin action, which deviated to an insu- function studies of family B GPCRs can be reexam-
Molecular Determinants of Glucagon Receptor Signaling 231

FIGURE 7 Comparison of amino acid sequences of glucagon in vertebrate species. Shaded areas
represent invariant residues (adapted from Ref. 138).

ined and molecular models refined and validated. family where ligands themselves are highly homolo-
Structural and functional relationships among mem- gous. Thus, ligand structure–activity studies and re-
bers of the glucagon subfamily of GPCRs have gen- ceptor structure–function relationships will continue
erated a model for receptor binding and activation. to provide important information. The amino acid
The experimental data has been supported by molec- residues that serve as selectivity filters that screen for
ular modeling, computational simulations, and, in the appropriate ligands and permit entry into the receptor
case of the receptor for PACAP, an NMR-determined binding pocket have to be delineated. No doubt, as
structure of receptor-bound PACAP. It is now gener- GR family receptor sequences from other species
ally accepted that the large amino terminus of family become available, differences in receptor structure
B GPCRs plays a key role in the binding of peptide might provide additional insight into how highly spe-
ligands with additional interactions from the extracel- cific receptors evolved in vertebrates.
lular loops. At present, there is no clear evidence that The nature of the conformational changes that link
any of the peptides penetrate deep into a TM binding agonist binding to receptor activation and subsequent
crevice. The situation will most likely be different in transmission of the initial signal to G proteins remains
the case of nonpeptide ligands of family B GPCRs to be addressed. The counterpart for the D(E)-R-Y
and will require a modification of the existing models. sequence that is a key player in the activation of all
Small-molecule nonpeptide compounds would diffuse family A receptors has not been identified in GR and
more easily into the TM bundle and occupy a binding members of family B GPCRs. Biophysical studies
pocket distinct from the natural peptide ligands. similar to those that have been carried out with rho-
The mechanism by which different ligands activate dopsin and ␤2-adrenergic receptors will allow direct
receptors believed to share similar overall structure is structural observation of movements of TM domains
still poorly understood, especially within the GR sub- and other conformational changes accompanying re-
232 Unson

ceptor activation. Methods of expression and isolation 18. MacNeil, D. J.; Occi, J. L.; Hey, P. J.; Strader, C. D.;
of detergent-stabilized receptor protein will have to be Graziano, M. P. Biochem Biophys Res Commun
developed for these studies. This is currently an oner- 1994, 198, 328 –334.
ous undertaking for the GR subfamily of GPCRs. 19. Lok, S.; Kuijper, J. L.; Jelinek, L. J.; Kramer, J. M.;
Adequate amounts of receptor protein will enable Whitmore, T. E.; Sprecher, C. A.; Mathewes, S.;
Grant, F. J.; Biggs, S. H.; Rosenberg, G. B.; et al.
determination of high-resolution structure of receptor-
Gene 1994, 140, 203–209.
bound glucagon by NMR and eventually atomic res-
20. Menzel, S.; Stoffel, M.; Espinosa, R., 3rd; Fernald,
olution structure by X-ray analysis. Understanding A. A.; Le Beau, M. M.; Bell, G. I. Genomics 1994, 20,
GR function at a molecular level will advance the 327–328.
development of new molecules for the management of 21. Christophe, J. Biochim Biophys Acta 1995, 1241,
diabetes mellitus. 45–57.
22. Svoboda, M.; Tastenoy, M.; Vertongen, P.; Rob-
berecht, P. Mol Cell Endocrinol 1994, 105, 131–137.
REFERENCES 23. Burcelin, R.; Li, J.; Charron, M. J. Gene 1995, 164,
305–310.
1. Granner, D.; Pilkis, S. J Biol Chem 1990, 265, 10173– 24. Wakelam, M. J.; Murphy, G. J.; Hruby, V. J.; Houslay,
10176. M. D. Nature 1986, 323, 68 –71.
2. Mojsov, S.; Heinrich, G.; Wilson, I. B.; Ravazzola, 25. Hansen, L. H.; Gromada, J.; Bouchelouche, P.; Whit-
M.; Orci, L.; Habener, J. F. J Biol Chem 1986, 261, more, T.; Jelinek, L.; Kindsvogel, W.; Nishimura, E.
11880 –11889.
Am J Physiol 1998, 274, C1552–C1562.
3. Bell, G. I.; Santerre, R. F.; Mullenbach, G. T. Nature
26. Staddon, J. M.; Hansford, R. G. Eur J Biochem 1989,
1983, 302, 716 –718.
179, 47–52.
4. Heinrich, G.; Gros, P.; Habener, J. F. J Biol Chem
27. Jiang, Y.; Cypess, A. M.; Muse, E. D.; Wu, C. R.;
1984, 259, 14082–14087.
Unson, C. G.; Merrifield, R. B.; Sakmar, T. P. Proc
5. Furuta, M.; Zhou, A.; Webb, G.; Carroll, R.; Ravaz-
Natl Acad Sci USA 2001, 98, 10102–10107.
zola, M.; Orci, L.; Steiner, D. F. J Biol Chem 2001,
28. Post, G. R.; Brown, J. H. Faseb J 1996, 10, 741–749.
276, 27197–27202.
29. Lopez-Ilasaca, M.; Crespo, P.; Pellici, P. G.; Gutkind,
6. Christophe, J. Ann NY Acad Sci 1996, 805, 31– 42;
J. S.; Wetzker, R. Science 1997, 275, 394 –397.
discussion 42– 43.
30. Gether, U. Endocrinol Rev 2000, 21, 90 –113.
7. Van Schravendijk, C. F.; Foriers, A.; Hooghe-Peters,
31. Horn, F.; Weare, J.; Beukers, M. W.; Horsch, S.;
E. L.; Rogiers, V.; De Meyts, P.; Sodoyez, J. C.;
Pipeleers, D. G. Endocrinology 1985, 117, 841– 848. Bairoch, A.; Chen, W.; Edvardsen, O.; Campagne, F.;
8. Kofod, H.; Unson, C. G.; Merrifield, R. B. Int J Vriend, G. Nucleic Acids Res 1998, 26, 275–279.
Peptide Protein Res 1988, 32, 436 – 440. 32. Ulrich, C. D. Jr.; Holtmann, M.; Miller, L. J. Gastro-
9. Jensen, M. D.; Heiling, V. J.; Miles, J. M. J Clin enterology 1998, 114, 382–397.
Endocrinol Metab 1991, 72, 308 –315. 33. Brown, E. M.; Segre, G. V.; Goldring, S. R. Baillieres
10. MacLeod, K. M.; Rodgers, R. L.; McNeill, J. H. Clin Endocrinol Metab 1996, 10, 123–161.
J Pharmacol Exp Ther 1981, 217, 798 – 804. 34. Thorens, B. Proc Natl Acad Sci USA 1992, 89, 8641–
11. Delahousse, M.; Mercier, O.; Bichara, M.; Paillard, 8645.
M.; Leviel, F.; Assan, R. Am J Physiol 1988, 254, 35. Munroe, D. G.; Gupta, A. K.; Kooshesh, F.; Vyas,
F762–F769. T. B.; Rizkalla, G.; Wang, H.; Demchyshyn, L.; Yang,
12. Dobbs, R.; Sakurai, H.; Sasaki, H.; Faloona, G.; Z. J.; Kamboj, R. K.; Chen, H.; McCallum, K.; Sum-
Valverde, I.; Baetens, D.; Orci, L.; Unger, R. Science ner-Smith, M.; Drucker, D. J.; Crivici, A. Proc Natl
1975, 187, 544 –547. Acad Sci USA 1999, 96, 1569 –1573.
13. Unger, R. H. Diabetologia 1985, 28, 574 –578. 36. Ishihara, T.; Nakamura, S.; Kaziro, Y.; Takahashi, T.;
14. Hansen, L. H.; Abrahamsen, N.; Hager, J.; Jelinek, L.; Takahashi, K.; Nagata, S. Embo J 1991, 10, 1635–
Kindsvogel, W.; Froguel, P.; Nishimura, E. Diabetes 1641.
1996, 45, 725–730. 37. Ishihara, T.; Shigemoto, R.; Mori, K.; Takahashi, K.;
15. Hager, J.; Hansen, L.; Vaisse, C.; Vionnet, N.; Phil- Nagata, S. Neuron 1992, 8, 811– 819.
ippi, A.; Poller, W.; Velho, G.; Carcassi, C.; Contu, L.; 38. Lutz, E. M.; Sheward, W. J.; West, K. M.; Morrow,
Julier, C.; et al. Nat Genet 1995, 9, 299 –304. J. A.; Fink, G.; Harmar, A. J. FEBS Lett 1993, 334,
16. Jelinek, L. J.; Lok, S.; Rosenberg, G. B.; Smith, R. A.; 3– 8.
Grant, F. J.; Biggs, S.; Bensch, P. A.; Kuijper, J. L.; 39. Usdin, T. B.; Mezey, E.; Button, D. C.; Brownstein,
Sheppard, P. O.; Sprecher, C. A.; et al. Science 1993, M. J.; Bonner, T. I. Endocrinology 1993, 133, 2861–
259, 1614 –1616. 2870.
17. Svoboda, M.; Ciccarelli, E.; Tastenoy, M.; Rob- 40. Mayo, K. E. Mol Endocrinol 1992, 6, 1734 –1744.
berecht, P.; Christophe, J. Biochem Biophys Res 41. Pisegna, J. R.; Wank, S. A. Proc Natl Acad Sci USA
Commun 1993, 192, 135–142. 1993, 90, 6345– 6349.
Molecular Determinants of Glucagon Receptor Signaling 233

42. Lin, H. Y.; Harris, T. L.; Flannery, M. S.; Aruffo, A.; 64. Unson, C. G.; Wu, C. R.; Sakmar, T. P.; Merrifield,
Kaji, E. H.; Gorn, A.; Kolakowski, L. F. Jr.; Yamin, R. B. J Biol Chem 2000, 275, 21631–21638.
M.; Lodish, H. F.; Goldring, S. R. Trans Assoc Am 65. Hjorth, S. A.; Orskov, C.; Schwartz, T. W. Mol En-
Physicians 1991, 104, 265–272. docrinol 1998, 12, 78 – 86.
43. Aiyar, N.; Rand, K.; Elshourbagy, N. A.; Zeng, Z.; 66. Schipani, E.; Kruse, K.; Juppner, H. Science 1995,
Adamou, J. E.; Bergsma, D. J.; Li, Y. J Biol Chem 268, 98 –100.
1996, 271, 11325–11329. 67. Gaudin, P.; Maoret, J. J.; Couvineau, A.; Rouyer-
44. Juppner, H.; Abou-Samra, A. B.; Freeman, M.; Kong, Fessard, C.; Laburthe, M. J Biol Chem 1998, 273,
X. F.; Schipani, E.; Richards, J.; Kolakowski, L. F. Jr.; 4990 – 4996.
Hock, J.; Potts, J. T. Jr.; Kronenberg, H. M.; et al. 68. Unson, C. G.; Andreu, D.; Gurzenda, E. M.; Merri-
Science 1991, 254, 1024 –1026. field, R. B. Proc Natl Acad Sci USA 1987, 84, 4083–
45. Abou-Samra, A. B.; Juppner, H.; Force, T.; Freeman, 4087.
M. W.; Kong, X. F.; Schipani, E.; Urena, P.; Richards, 69. Unson, C. G.; Macdonald, D.; Ray, K.; Durrah, T. L.;
J.; Bonventre, J. V.; Potts, J. T. Jr.; et al. Proc Natl Merrifield, R. B. J Biol Chem 1991, 266, 2763–2766.
Acad Sci USA 1992, 89, 2732–2736. 70. Lin, M. C., Wright, D. E., Hruby, V. J.; Rodbell, M.
46. Chen, R.; Lewis, K. A.; Perrin, M. H.; Vale, W. W. Biochemistry 1975, 14, 1559 –1563.
Proc Natl Acad Sci USA 1993, 90, 8967– 8971. 71. Bregman, M. D.; Hruby, V. J. FEBS Lett 1979, 101,
47. Palczewski, K.; Kumasaka, T.; Hori, T.; Behnke, 191–194.
C. A.; Motoshima, H.; Fox, B. A.; Le Trong, I.; Teller, 72. Unson, C. G.; Macdonald, D.; Merrifield, R. B. Arch
D. C.; Okada, T.; Stenkamp, R. E.; Yamamoto, M.; Biochem Biophys 1993, 300, 747–750.
Miyano, M. Science 2000, 289, 739 –745. 73. Unson, C. G.; Wu, C. R.; Merrifield, R. B. Biochem-
48. Teller, D. C.; Okada, T.; Behnke, C. A.; Palczewski, istry 1994, 33, 6884 – 6887.
K.; Stenkamp, R. E. Biochemistry 2001, 40, 7761– 74. Unson, C. G.; Wu, C. R.; Cheung, C. P.; Merrifield,
7772. R. B. J Biol Chem 1998, 273, 10308 –10312.
49. Frimurer, T. M.; Bywater, R. P. Proteins 1999, 35,
75. Sturm, N. S.; Lin, Y.; Burley, S. K.; Krstenansky,
375–386.
J. L.; Ahn, J. M.; Azizeh, B. Y.; Trivedi, D.; Hruby,
50. Tams, J. W.; Knudsen, S. M.; Fahrenkrug, J. J Mol
V. J. J Med Chem 1998, 41, 2693–2700.
Neurosci 2001, 17, 325–330.
76. Trivedi, D.; Lin, Y.; Ahn, J. M.; Siegel, M.; Mollova,
51. Unson, C. G.; Cypess, A. M.; Kim, H. N.; Goldsmith,
N. N.; Schram, K. H.; Hruby, V. J. J Med Chem 2000,
P. K.; Carruthers, C. J.; Merrifield, R. B.; Sakmar,
43, 1714 –1722.
T. P. J Biol Chem 1995, 270, 27720 –27727.
77. Sapse, A. M.; Rothchild, R.; Jain, D. R.; Unson, C. G.
52. Buggy, J. J.; Heurich, R. O.; MacDougall, M.; Kelley,
Mol. Med. 2002, 7, in press.
K. A.; Livingston, J. N.; Yoo-Warren, H.; Rosso-
78. Krstenansky, J. L.; Trivedi, D.; Hruby, V. J. Biochem-
mando, A. J. Diabetes 1997, 46, 1400 –1405.
istry 1986, 25, 3833–3839.
53. Heurich, R. O.; Buggy, J. J.; Vandenberg, M. T.;
Rossomando, A. J. Biochem Biophys Res Commun 79. Azizeh, B. Y.; Ahn, J. M.; Caspari, R.; Shenderovich,
1996, 220, 905–910. M. D.; Trivedi, D.; Hruby, V. J. J Med Chem 1997,
54. Grady, T.; Fickova, M.; Tager, H. S.; Trivedi, D.; 40, 2555–2562.
Hruby, V. J. J Biol Chem 1987, 262, 15514 –15520. 80. Azizeh, B. Y.; Shenderovich, M. D.; Trivedi, D.; Li,
55. Murphy, G. J.; Hruby, V. J.; Trivedi, D.; Wakelam, G.; Sturm, N. S.; Hruby, V. J. J Med Chem 1996, 39,
M. J.; Houslay, M. D. Biochem J 1987, 243, 39 – 46. 2449 –2455.
56. Cypess, A. M.; Unson, C. G.; Wu, C. R.; Sakmar, T. P. 81. Buggy, J. J.; Livingston, J. N.; Rabin, D. U.; Yoo-
J Biol Chem 1999, 274, 19455–19464. Warren, H. J Biol Chem 1995, 270, 7474 –7478.
57. Chicchi, G. G.; Graziano, M. P.; Koch, G.; Hey, P.; 82. Holtmann, M. H.; Hadac, E. M.; Miller, L. J. J Biol
Sullivan, K.; Vicario, P. P.; Cascieri, M. A. J Biol Chem 1995, 270, 14394 –14398.
Chem 1997, 272, 7765–7769. 83. Holtmann, M. H.; Ganguli, S.; Hadac, E. M.; Dolu, V.;
58. Lad, P. M.; Welton, A. F.; Rodbell, M. J Biol Chem Miller, L. J. J Biol Chem 1996, 271, 14944 –14949.
1977, 252, 5942–5946. 84. Xiao, Q.; Jeng, W.; Wheeler, M. B. J Mol Endocrinol
59. Sonne, O.; Berg, T.; Christoffersen, T. J Biol Chem 2000, 25, 321–335.
1978, 253, 3203–3210. 85. Wilmen, A.; Van Eyll, B.; Goke, B.; Goke, R. Pep-
60. Horwitz, E. M.; Wyborski, R. J.; Gurd, R. S. J Biol tides 1997, 18, 301–305.
Chem 1986, 261, 13670 –13676. 86. Dong, M.; Asmann, Y. W.; Zang, M.; Pinon, D. I.;
61. Robishaw, J. D.; Smigel, M. D.; Gilman, A. G. J Biol Miller, L. J. J Biol Chem 2000, 275, 26032–26039.
Chem 1986, 261, 9587–9590. 87. Dong, M.; Wang, Y.; Hadac, E. M.; Pinon, D. I.;
62. Kozasa, T.; Itoh, H.; Tsukamoto, T.; Kaziro, Y. Proc Holicky, E.; Miller, L. J. J Biol Chem 1999, 274,
Natl Acad Sci USA 1988, 85, 2081–2085. 19161–19167.
63. Mumby, S. M.; Kahn, R. A.; Manning, D. R.; Gilman, 88. Dong, M.; Wang, Y.; Miller, L. J. Ann NY Acad Sci
A. G. Proc Natl Acad Sci USA 1986, 83, 265–269. 2000, 921, 381–386.
234 Unson

89. Dong, M.; Wang, Y.; Pinon, D. I.; Hadac, E. M.; 111. Okada, T.; Ernst, O. P.; Palczewski, K.; Hofmann,
Miller, L. J. J Biol Chem 1999, 274, 903–909. K. P. Trends Biochem Sci 2001, 26, 318 –324.
90. Zhou, A. T.; Bessalle, R.; Bisello, A.; Nakamoto, C.; 112. Unson, C. G.; Merrifield, R. B. Proc Natl Acad Sci
Rosenblatt, M.; Suva, L. J.; Chorev, M. Proc Natl USA 1994, 91, 454 – 458.
Acad Sci USA 1997, 94, 3644 –3649. 113. Unson, C. G.; Wu, C. R.; Fitzpatrick, K. J.; Merrifield,
91. Grauschopf, U.; Lilie, H.; Honold, K.; Wozny, M.; R. B. J Biol Chem 1994, 269, 12548 –12551.
Reusch, D.; Esswein, A.; Schafer, W.; Rucknagel, 114. Vertongen, P.; Solano, R. M.; Perret, J.; Langer, I.;
K. P.; Rudolph, R. Biochemistry 2000, 39, 8878 – Robberecht, P.; Waelbroeck, M. Br J Pharmacol 2001,
8887. 133, 1249 –1254.
92. Perrin, M. H.; Fischer, W. H.; Kunitake, K. S.; Craig, 115. Solano, R. M.; Langer, I.; Perret, J.; Vertongen, P.;
A. G.; Koerber, S. C.; Cervini, L. A.; Rivier, J. E.; Juarranz, M. G.; Robberecht, P.; Waelbroeck, M.
Groppe, J. C.; Greenwald, J.; Moller Nielsen, S.; Vale, J Biol Chem 2001, 276, 1084 –1088.
W. W. J Biol Chem 2001, 276, 31528 –31534. 116. Gallivan, J. P.; Dougherty, D. Proc. Natl. Acad. Sci
93. Bazarsuren, A.; Grauschopf, U.; Wozny, M.; Reusch, USA 1999, 96, 9459 –9464.
D.; Hoffmann, E.; Schaefer, W.; Panzner, S.; Rudolph, 117. Dougherty, D. A. Science 1996, 271, 163–168.
R. Biophys Chem 2002, 96, 305–318. 118. Ma, J. C.; Dougherty, D. A. Chem Rev 1997, 97,
94. Vilardaga, J. P.; Di Paolo, E.; Bialek, C.; De Neef, P.; 1303–1324.
Waelbroeck, M.; Bollen, A.; Robberecht, P. Eur J Bio- 119. Inooka, H.; Endo, S.; Kitada, C.; Mizuta, E.; Fujino,
chem 1997, 246, 173–180. M. Int J Peptide Protein Res 1992, 40, 456 – 464.
95. Asmann, Y. W.; Dong, M.; Ganguli, S.; Hadac, E. M.; 120. Ohtaki, T.; Ogi, K.; Masuda, Y.; Mitsuoka, K.; Fu-
Miller, L. J. Mol Pharmacol 2000, 58, 911–919. jiyoshi, Y.; Kitada, C.; Sawada, H.; Onda, H.; Fujino,
96. Mannstadt, M.; Luck, M. D.; Gardella, T. J.; Juppner, M. J Biol Chem 1998, 273, 15464 –15473.
H. J Biol Chem 1998, 273, 16890 –16896. 121. Braun, W.; Wider, G.; Lee, K. H.; Wuthrich, K. J Mol
97. Carruthers, C. J.; Unson, C. G.; Kim, H. N.; Sakmar, Biol 1983, 169, 921–948.
122. Krstenansky, J. L.; Zechel, C.; Trivedi, D.; Hruby,
T. P. J Biol Chem 1994, 269, 29321–29328.
V. J. Int J Peptide Protein Res 1988, 32, 468 – 475.
98. Lin, S. C.; Lin, C. R.; Gukovsky, I.; Lusis, A. J.;
123. Unson, C. G.; Gurzenda, E. M.; Iwasa, K.; Merrifield,
Sawchenko, P. E.; Rosenfeld, M. G. Nature 1993, 364,
R. B. J Biol Chem 1989, 264, 789 –794.
208 –213.
124. Gourlet, P.; Vilardaga, J. P.; De Neef, P.; Waelbroeck,
99. Van Eyll, B.; Goke, B.; Wilmen, A.; Goke, R. Pep-
M.; Vandermeers, A.; Robberecht, P. Peptides 1996,
tides 1996, 17, 565–570.
17, 825– 829.
100. Tibaduiza, E. C.; Chen, C.; Beinborn, M. J Biol Chem
125. Hoare, S. R.; Usdin, T. B. Curr Pharm Des 2001, 7,
2001, 276, 37787–37793.
689 –713.
101. Cao, Y. J.; Gimpl, G.; Fahrenholz, F. Biochem Bio-
126. Gardella, T. J.; Juppner, H. Trends Endocrinol Metab
phys Res Commun 1995, 212, 673– 680.
2001, 12, 210 –217.
102. Perrin, M. H.; Sutton, S.; Bain, D. L.; Berggren, 127. Mannstadt, M.; Juppner, H.; Gardella, T. J. Am J
W. T.; Vale, W. W. Endocrinology 1998, 139, 566 – Physiol 1999, 277, F665– 675.
570. 128. Lee, C.; Luck, M. D.; Juppner, H.; Potts, J. T. Jr.;
103. Hjorth, S. A.; Schwartz, T. W. Acta Physiol Scand Kronenberg, H. M.; Gardella, T. J. Mol Endocrinol
1996, 157, 343–345. 1995, 9, 1269 –1278.
104. Unson, C. G.; Cypess, A. M.; Wu, C. R.; Goldsmith, 129. Adams, A. E.; Bisello, A.; Chorev, M.; Rosenblatt,
P. K.; Merrifield, R. B.; Sakmar, T. P. Proc Natl Acad M.; Suva, L. J. Mol Endocrinol 1998, 12, 1673–1683.
Sci USA 1996, 93, 310 –315. 130. Carter, P. H.; Juppner, H.; Gardella, T. J. Endocrinol-
105. Graziano, M. P.; Hey, P. J.; Strader, C. D. Receptors ogy 1999, 140, 4972– 4981.
Channels 1996, 4, 9 –17. 131. Shah, G. V.; Epand, R. M.; Orlowski, R. C. Mol Cell
106. Unson, C. G.; Wu, C. R.; Jiang, Y.; Yoo, B.; Cheung, Endocrinol 1987, 49, 203–210.
C.; Sakmar, T. P.; Merrifield, R. B. Biochemistry 132. Perret, J.; Van Craenenbroeck, M.; Langer, I.; Verton-
2002, 41, in press. gen, P.; Gregoire, F.; Robberecht, P.; Waelbroeck, M.
107. Hjorth, S. A.; Adelhorst, K.; Pedersen, B. B.; Kirk, O.; Biochem J 2002, 362, 389 –394.
Schwartz, T. W. J Biol Chem 1994, 269, 30121– 133. Nielsen, S. M.; Nielsen, L. Z.; Hjorth, S. A.; Perrin,
30124. M. H.; Vale, W. W. Proc Natl Acad Sci USA 2000,
108. Greenberg, Z.; Bisello, A.; Mierke, D. F.; Rosenb- 97, 10277–10281.
latt, M.; Chorev, M. Biochemistry 2000, 39, 8142– 134. Shimizu, M.; Carter, P. H.; Gardella, T. J. J Biol Chem
8152. 2000, 275, 19456 –19460.
109. Piserchio, A.; Bisello, A.; Rosenblatt, M.; Chorev, M.; 135. Sherwood, N. M.; Krueckl, S. L.; McRory, J. E. En-
Mierke, D. F. Biochemistry 2000, 39, 8153– 8160. docrinol Rev 2000, 21, 619 – 670.
110. Menon, S. T.; Han, M.; Sakmar, T. P. Physiol Rev 136. McRory, J.; Sherwood, N. M. Endocrinology 1997,
2001, 81, 1659 –1688. 138, 2380 –2390.
Molecular Determinants of Glucagon Receptor Signaling 235

137. Campbell, R. M.; Scanes, C. G. Growth Regul 1992, 142. Pollock, H. G.; Kimmel, J. R.; Ebner, K. E.; Hamilton,
2, 175–191. J. W.; Rouse, J. B.; Lance, V.; Rawitch, A. B. Gen
138. Ngan, E. S.; Chow, L. S.; Tse, D. L.; Du, X.; Wei, Y.; Comp Endocrinol 1988, 69, 133–140.
Mojsov, S.; Chow, B. K. FEBS Lett 1999, 457, 499 – 143. Conlon, J. M.; Abdel-Wahab, Y. H.; O’Harte, F. P.;
504. Nielsen, P. F.; Whittaker, J. Endocrinology 1998, 139,
139. Irwin, D. M.; Huner, O.; Youson, J. H. Mol Biol Evol 3442–3448.
1999, 16, 1548 –1557. 144. Sivarajah, P.; Wheeler, M. B.; Irwin, D. M. Comp
140. Irwin, D. M.; Wong, J. Mol Endocrinol 1995, 9, Biochem Physiol B Biochem Mol Biol 2001, 128,
267–277. 517–527.
141. Irwin, D. M.; Satkunarajah, M.; Wen, Y.; Brubaker, 145. Lefebvre, P. J. Diabetes Care 1995, 18, 715–730.
P. L.; Pederson, R. A.; Wheeler, M. B. Proc Natl Acad 146. Plisetskaya, E. M.; Mommsen, T. P. Int Rev Cytol
Sci USA 1997, 94, 7915–7920. 1996, 168, 187–257.

You might also like