throbber
Receptors and Channels, 8:179–188, 2002
`Copyright c(cid:176) 2002 Taylor & Francis
`1060-6823/02 $12.00 + .00
`DOI: 10.1080/10606820290005155
`
`Structure-Function of the Glucagon Receptor Family
`of G Protein–Coupled Receptors: The Glucagon, GIP,
`GLP-1, and GLP-2 Receptors
`
`P. L. Brubaker
`Departments of Physiology and Medicine, University of Toronto, Toronto, Ontario, Canada
`D. J. Drucker
`Department of Medicine, Banting and Best Diabetes Centre, Toronto General Hospital,
`Toronto, Ontario, Canada
`
`The glucagon-like peptides include glucagon, GLP-1, and
`GLP-2, and exert diverse actions on nutrient intake, gastrointesti-
`nal motility, islet hormone secretion, cell proliferation and apopto-
`sis, nutrient absorption, and nutrient assimilation. GIP, a related
`member of the glucagon peptide superfamily, also regulates nutri-
`ent disposal via stimulation of insulin secretion. The actions of these
`peptides are mediated by distinct members of the glucagon recep-
`tor superfamily of G protein–coupled receptors. These receptors
`exhibit unique patterns of tissue-specific expression, exhibit consid-
`erable amino acid sequence identity, and share similar structural
`and functional properties with respect to ligand binding and sig-
`nal transduction. This article provides an overview of the biology
`of these receptors with an emphasis on understanding the unique
`actions of glucagon-related peptides through studies of the biology
`of their cognate receptors.
`
`Keywords Diabetes, GIP, GLP-1, GLP-2, Glucagon, Glucose,
`Intestine, Regulatory Peptides
`
`GENERAL INTRODUCTION
`The gastroenteropancreatic-brain axis expresses a diverse
`number of peptide hormones, including several that are struc-
`turally related to the pancreatic hormone, glucagon (Table 1).
`Within the mammalian glucagon superfamily of peptides,
`glucagon and the glucagon-like peptides, GLP-1 and GLP-2,
`are all encoded by a single proglucagon gene (Bell et al. 1983;
`Irwin 2001; White and Saunders 1986). Tissue-specific
`posttranslational processing of proglucagon by prohormone
`
`Address correspondence to Daniel J. Drucker, M.D., Banting and
`Best Diabetes Centre, Toronto General Hospital, 200 Elizabeth Street,
`Toronto, Ontario, M5G 2C4 Canada. E-mail: d.drucker@utoronto.ca
`
`convertases results in the liberation of glucagon in the pancreatic
`A cell, and GLP-1 and GLP-2 in the intestinal L cell and brain
`(Mojsov et al. 1986; Orskov et al. 1987). As discussed below,
`all three proglucagon-derived peptides (PGDPs) play impor-
`tant roles in the physiologic regulation of nutrient homeosta-
`sis, through effects on energy intake and satiety, nutrient fluxes
`through and across the gastrointestinal tract, and energy as-
`similation. Several of these biological activities are shared by
`a fourth glucagon-related peptide hormone, glucose-dependent
`insulinotropic peptide (GIP) (Table 1). The diverse biological
`activities of these glucagon-related peptides are exerted through
`highly specific G protein–coupled receptors (GPCR), members
`of the glucagon receptor superfamily. For the purposes of this
`article, the biological activities and mechanisms of action of
`each of these peptides will be discussed individually.
`
`Biological Actions of the Glucagon-Related Peptides
`Glucagon
`Glucagon is a 29 amino acid peptide hormone liberated from
`islet A cells in the endocrine pancreas. The principal actions
`of glucagon involve regulation of metabolic pathways involved
`in glucose homeostasis. Glucagon secretion from the A cell is
`coupled to the ambient levels of circulating glucose, with hypo-
`glycemia and hyperglycemia leading to stimulation and inhibi-
`tion of glucagon release, respectively (Unger 1985). Glucagon
`action in the liver impinges on numerous enzymes important
`for control of gluconeogenesis, glycogenolysis, and fatty acid
`metabolism, leading to restoration of circulating glucose con-
`centrations in the setting of energy depletion. The actions of
`glucagon on hepatic glucose production are exerted at multiple
`levels, including regulation of gene transcription and modula-
`tion of enzyme activity (Pilkis and Granner 1992). Defective
`
`179
`
`Page 1
`
`NPS EX. 2038
`CFAD v. NPS
`IPR2015-00990
`
`

`
`180
`
`P. L. BRUBAKER AND D. J. DRUCKER
`
`TABLE 1
`The amino acid sequences of human glucagon, GLP-1, GLP-2, and GIP
`
`Glucagon
`GLP-1
`GLP-2
`GIP
`
`HSQGTFTSDYSKYLDSRRAQDFVQWLMNT
`HAEGTFTSDVSSYLEGQAAKEFIAWLVKGRG
`HADGSFSDEMNTILDNLAARDFINWLIQTKITD
`YAEGTFISDYSIAMDKIRQQDFVNWLLAQ
`
`glucagon secretion contributes to the development of hypo-
`glycemia in insulin-treated patients with diabetes (Gerich 1988;
`Taborsky, Jr. et al. 1998).
`Glucagon receptors are widely expressed in multiple tissues
`including the liver, brain, pancreas, heart, kidney, and smooth
`muscle cell in the gastrointestinal tract and peripheral vascula-
`ture. In the heart, glucagon stimulates inotropic and chronotropic
`activity (Parmley et al. 1968), leading to the selective use of
`glucagon in medical emergencies characterized by refractory
`bradycardia due to beta blocker toxicity or cardiogenic shock
`(White 1999). Supraphysiological levels of glucagon also exert
`vasodilator effects by reducing vascular resistance in specific
`vascular beds (Farah 1983).
`Glucagon infusion activates nephrogenic adenylyl cyclase
`(leading to increased urinary cAMP excretion), increases the
`glomerular filtration rate, and regulates ion transport and elec-
`trolyte excretion in the kidney (Ahloulay et al. 1992; Broadus
`et al. 1970; Schwartz Sorensen et al. 1993). Although hypo-
`glycemia is associated with increased hepatic and renal glu-
`cose production, a role for glucagon in the regulation of renal
`gluconeogenesis is unclear (Cersosimo et al. 1999; Stumvoll
`et al. 1997, 1998). Whether glucagon actions on the kidney
`are essential for regulation of renal physiology remains to be
`determined.
`Within the brain, PGDPs, including glucagon, are synthe-
`sized principally in the brainstem and, to a lesser extent, in
`the hypothalamus (Drucker and Asa 1988); however, glucagon-
`immunoreactive nerve fibers are widely distributed through the
`mammalian central nervous system (Dorn et al. 1983). Glucagon
`administration elicits a broad spectrum of actions, including
`stimulation of anterior pituitary hormone secretion (Spathis et al.
`1974), hypothalamic somatostatin release (Shimatsu et al. 1983),
`and ketone utilization (Kirsch and D’Alecy 1984). Intracere-
`broventricular administration of glucagon potently suppresses
`food intake in the rat (Inokuchi et al. 1984) and enhances sym-
`pathetic nerve activity (Krzeski et al. 1989; Shimizu et al. 1993),
`whereas glucagon suppresses the activity of hypothalamic
`glucose-sensitive neurons but has no effect on cortical neurons
`in the rat (Inokuchi et al. 1986).
`Glucagon stimulates lipolysis in isolated adipocyte prepara-
`tions from young animals (Heckemeyer et al. 1983; Lefebvre and
`Luyckx 1969); however, these actions are significantly attenu-
`ated with aging or weight gain (Bertrand et al. 1980). The poten-
`tial importance of glucagon for stimulation of human adipocyte
`lipolysis is less clear (Richter et al. 1989; Richter and Schwandt
`1985). Glucagon withdrawal or physiological hyperglucagone-
`
`mia did not produce significant changes in palmitate flux in nor-
`mal or diabetic human subjects (Jensen et al. 1991). Similarly,
`subcutaneous infusion of glucagon into the gastrocnemius mus-
`cle or abdominal tissue had no effects on lipolysis rates or blood
`flow in normal human subjects (Bertin et al. 2001; Gravholt
`et al. 2001). Glucagon modulates electrical activity in gastroin-
`testinal smooth muscle (Patel et al. 1979; Taylor et al. 1975),
`leading to its clinical use as spasmolytic agent for modulation
`of motility during endoscopic and radiologic examination of the
`gastrointestinal tract.
`
`Glucagon-Like Peptide-1 and Glucose-Dependent
`Insulinotropic Peptide
`The incretin concept was first developed in 1930 to describe
`the observation that administration of glucose by the oral route
`was associated with a greater increment in insulin secretion than
`was a euglycemic load given intravenously (Elrick et al. 1964;
`LaBarre and Still 1930). This concept was further refined by both
`Unger (Unger and Eisentraut 1969) and Creutzfeldt (Creutzfeldt
`1979) to define an incretin as a gut hormone released in response
`to nutrient ingestion that stimulates glucose-dependent insulin
`secretion. The first incretin to be identified was the duodenal
`hormone, GIP. Although initially reported to possess gastric in-
`hibitory actions (Brown 1982; Brown and Dryburgh 1971), GIP
`was subsequently discovered to be a potent stimulator of insulin
`secretion (Dupre et al. 1973). Subsequent studies using GIP re-
`ceptor antagonists have clearly established that GIP fulfills all
`of the requirements for an incretin (Baggio et al. 2000; Lewis
`et al. 2000; Tseng, Kieffer et al. 1996). However, reports that
`immunoneutralization of GIP does not abolish the incretin effect
`of ingested nutrients (Ebert and Creutzfeldt 1982; Ebert et al.
`1983), led to an ongoing search for additional insulinotropic
`gut hormones. It was not until 1987 that the second major in-
`cretin was discovered to be another member of the glucagon-
`family of peptides, GLP-1 (Kreymann et al. 1987; Mojsov et al.
`1987; Orskov and Nielsen 1988). Numerous studies on the in-
`sulinotropic actions of GIP and GLP-1 have now established
`that, together, these peptides constitute the majority of the in-
`cretin effect following ingestion of a meal, and that the two hor-
`mones contribute equivalently to this effect (Kieffer and Habener
`1999). The importance of GIP and GLP-1 in glucose homeosta-
`sis is suggested by the strong degree of sequence conservation
`that exists for each of these hormones (Irwin 2001). Consistent
`with this hypothesis, receptor antagonist studies and the analysis
`of mice with null mutations in the receptors for GIP (GIPR) or
`
`Page 2
`
`

`
`STRUCTURE-FUNCTION OF G PROTEIN–COUPLED RECEPTORS
`
`181
`
`GLP-1 (GLP-1R) have demonstrated that loss of GIP or GLP-
`1 action results in an impaired insulin response to oral glucose
`(Miyawaki et al. 1999; Scrocchi et al. 1996). Whether these pep-
`tides are the sole hormones mediating the incretin effect awaits
`the development of mice with combined defects in both GIP
`and GLP-1 receptor signaling. Finally, very recent studies have
`indicated an exciting role for GLP-1 as a fl cell tropic factor.
`Administration of GLP-1, or exendin-4, a long-acting GLP-1R
`agonist, to mice or rats with diminished fl cell reserve leads to
`enhancements in fl cell mass and an increased capacity for in-
`sulin secretion (Stoffers et al. 2000; Xu et al. 1999). Consistent
`with these observations, mice with a null mutation in the GLP-
`1R exhibit alterations in fl cell topography and reduced insulin
`secretion (Ling et al. 2001). Whether GIP also regulates fl cell
`growth remains to be established.
`Because of their insulinotropic and glucose-lowering actions,
`both GIP and GLP-1 are candidate peptides for the treatment
`of type 2 diabetes mellitus. However, only GLP-1 is currently
`in clinical trials for such therapeutic use, for two main rea-
`sons. First, although both GIP and GLP-1 inhibit gastric emp-
`tying (Nauck et al. 1996), an effect that delays postprandial
`rises in glycemia, only GLP-1 also induces satiety (Scrocchi
`et al. 1996; Turton et al. 1996). In addition, these peptides
`have opposing effects on glucagon release, with GLP-1 inhibit-
`ing, but GIP stimulating, glucagon secretion (Elahi et al. 1979;
`Komatsu et al. 1989). More importantly, although GLP-1 low-
`ers glycemia in patients with type 2 diabetes (Gutniak et al.
`1992; Nauck et al. 1996), the insulinotropic actions of GIP ap-
`pear to be markedly abrogated in such individuals (Elahi et al.
`1994; Nauck et al. 1993). These findings are consistent with
`previous observations that the incretin effect is diminished in
`patients with type 2 diabetes (Nauck et al. 1986), but, in addi-
`tion, suggest that this defect occurs consequent to abnormalities
`at the level of the GIPR and/or in its postreceptor signaling cas-
`cade. Indeed, diabetic Zucker fatty rats, a rodent model of type 2
`diabetes, exhibit decreased islet expression of the GIPR in as-
`sociation with diminished fl cell responsiveness to GIP (Lynn
`et al. 2001).
`
`Glucagon-Like Peptide-2
`GLP-2, located carboxyterminal to GLP-1 in the proglucagon
`sequence, is cosecreted with GLP-1, oxyntomodulin, and gli-
`centin from gut L cells, primarily in response to nutrient in-
`gestion (Brubaker, Crivici et al. 1997; Xiao et al. 1999; Xiao,
`Boushey et al. 2000). Following clinical and experimental ob-
`servations linking intestinal growth, injury, and gut adaptation
`to increased levels of circulating PGDPs (Drucker 1999, 2001),
`GLP-2 was subsequently identified as the PGDP exhibiting sig-
`nificant intestinotrophic properties in vivo (Drucker et al. 1996).
`Administration of GLP-2 to normal mice and rats results in
`significant increases in mucosal thickness of the small and large
`intestine, with more prominent trophic effects consistently ob-
`served in the small bowel, especially in the jejunum (Drucker et al.
`1996; Drucker, Deforest et al. 1997; Drucker, Shi et al. 1997;
`
`Tsai, Hill, Asa et al. 1997; Tsai, Hill, and Drucker 1997). The
`intestinotrophic effects of GLP-2 are mediated by stimulation
`of crypt cell proliferation and inhibition of apoptosis in both
`the crypt and villus compartments of the small bowel (Drucker
`et al. 1996; Tsai, Hill, Asa et al. 1997). GLP-2 administration
`also results in inhibition of gastric emptying, reduced gastric
`acid secretion, stimulation of intestinal hexose transport and
`nutrient absorption, and reduction in mucosal epithelial perme-
`ability in the small bowel (Benjamin et al. 2000; Brubaker, Izzo
`et al. 1997; Cheeseman and Tsang 1996; Wojdemann et al. 1998,
`1999).
`The reparative and antiapoptotic actions of GLP-2 have also
`been examined in the setting of experimental intestinal injury.
`GLP-2 reduces mucosal damage and decreases morbidity and
`mortality in a broad spectrum of rodent models of small and
`large bowel injury, including enteral nutrient deprivation, intesti-
`nal inflammation, ischemia, and chemotherapy-induced enteritis
`(Alavi et al. 2000; Boushey et al. 1999, 2001; Chance et al. 1997,
`2001; Drucker et al. 1999). GLP-2 also enhances nutrient ab-
`sorption and intestinal adaptation in rats with experimental small
`bowel resection (Scott et al. 1998) and in human subjects with
`short bowel syndrome (Jeppesen et al. 2001).
`
`RECEPTOR CLONING: SPECIFICITY
`AND DISTRIBUTION
`The GLP-1R was the first member of this group of receptors to
`be cloned (Thorens 1992). A 463 amino acid 7 transmembrane-
`spanning protein, the GLP-1R exhibits 27% to 40% sequence
`homology to the receptors for secretin, calcitonin, and parathy-
`roid hormone. As these receptors shared higher identity with
`each other than with other members of the G protein–coupled
`receptor superfamily, they were classified together into a new
`family of receptors (Thorens 1992) now known as the type II
`receptor family. However, despite the strong sequence homol-
`ogy between GLP-1 and the other members of the glucagon-
`related family of peptides, the GLP-1R recognizes GLP-1 specif-
`ically, with no demonstrable binding by a number of related
`peptides, including secretin and vasoactive intestinal peptide
`(VIP) (Fehmann et al. 1994). Although binding of GIP and
`glucagon to the GLP-1R has been observed in some studies, this
`appears to occur only at micromolar concentrations (Thorens
`1992). Furthermore, only GLP-1 appears to activate the GLP-
`1R, stimulating production of cAMP with an ED50 of 3 nM in
`COS cells transfected with the human receptor (Dillon et al.
`1993). Consistent with the very high degree of sequence conser-
`vation found for GLP-1 in most mammalian species, the GLP-
`1R also exhibits conservation between species, with 90% ho-
`mology between the rat and human receptors (Sivarajah et al.
`2001). The distribution of the mammalian GLP-1 receptor also
`appears to be conserved, with RT-PCR and Northern blot analy-
`ses demonstrating expression in tissues known to be responsive
`to GLP-1, including the pancreatic fl cells, the stomach, and
`the brain (Bullock et al. 1996; Campos et al. 1994; Thorens
`1992).
`
`Page 3
`
`

`
`182
`
`P. L. BRUBAKER AND D. J. DRUCKER
`
`The rat glucagon receptor (GR) cDNA was cloned in 1993
`and encodes a 485 amino acid protein of »54,962 daltons
`(Jelinek et al. 1993). Glucagon stimulates an increase in cAMP
`and intracellular calcium in transfected cells expressing the
`cloned receptor, with much weaker receptor activation noted
`only with pharmacological concentrations of the related pep-
`tides secretin, calcitonin, and parathyroid hormone (Jelinek et al.
`1993). The human glucagon receptor cDNA encodes a protein
`of 477 amino acids that exhibits 82% identity to the rat receptor
`(Lok et al. 1994; MacNeil et al. 1994). Glucagon inhibits binding
`of [125-I]-glucagon to the cloned human receptor with an IC50
`of 5 nM, with tenfold lower tracer displacement exhibited by
`oxyntomodulin and GLP-1, whereas GIP, GLP-2, and secretin
`fail to displace binding of [125-I]-glucagon binding at concen-
`trations up to 3 uM (MacNeil et al. 1994). Mammalian glucagon
`receptor RNA transcripts are widely expressed in peripheral tis-
`sues, including liver, kidney, heart, adipose tissue, pancreatic
`islets, the gastrointestinal tract, spleen, thymus, adrenal gland,
`ovary, testes, and the central nervous system (Campos et al.
`1994; Dunphy et al. 1998; Hansen et al. 1995; Svoboda et al.
`1994).
`Cloning of the GIPR in 1993 demonstrated that this 455
`amino acid protein also belongs to the 7 transmembrane-
`spanning, glucagon receptor-related superfamily of receptors,
`sharing 44% sequence identity with the glucagon receptor (Usdin
`et al. 1993). The GIPR exhibits highly specific GIP binding,
`although some binding by exendin-4, a GLP-1R agonist, was
`noted at very high concentrations of 1–10 „M (Gremlich et al.
`1995). However, there was no demonstrable response of the
`GIPR to other members of the glucagon-related family of hor-
`mones (Gremlich et al. 1995; Usdin et al. 1993). A high degree
`of sequence conservation (>80%) between the rat, hamster, and
`human GIPR’s has been reported (Gremlich et al. 1995; Yamada
`et al. 1995). Consistent with its known sites of action, the GIP
`receptor is expressed in tissues such as the pancreas and upper
`gastrointestinal tract (Usdin et al. 1993). Interestingly, the GIPR
`is also expressed at low levels in the adrenal gland, where over-
`expression appears to contribute to the development of “Food-
`Dependent Cushing’s Syndrome” (N’Diaye et al. 1998). The
`receptor is also found in adipose tissue, where it may play a role
`in lipolysis (Yip and Wolfe 2000), and in the brain, where GIP
`function remains to be ascertained. GIP also exerts anabolic ac-
`tions in bone cells (Bollag et al. 2000), and GIP administration
`prevents ovariectomy-associated bone loss in vivo (Bollag et al.
`2001).
`The human and rat GLP-2 receptor cDNAs were isolated from
`intestinal and hypothalamic cDNA libraries in 1999 (Munroe
`et al. 1999). The rat GLP-2R cDNA comprises 2357 nucleotides
`and encodes a 550 amino acid receptor precursor protein. Two
`putative functionally identical translation start sites are present at
`0
`end of the rat receptor, whereas only a single upstream ATG
`the 5
`is present in the human sequence; the mouse GLP-2R sequence
`contains only the downstream ATG codon (Lovshin et al. 2001).
`The GLP-2R specifically recognizes GLP-2 with an EC50 for
`
`cAMP stimulation of 0.58 nM, with no significant stimulation of
`cAMP generation detected with related glucagon-like peptides
`and peptide members of the glucagon-secretin superfamily at
`concentrations of 10 nM (DaCambra et al. 2000; Munroe et al.
`1999). Unlike the more widely expressed GLP-1 and glucagon
`receptors, GLP-2R expression is more restricted, predominantly
`to endocrine cells and enteric neurons in the gastrointestinal
`tract and selected brain neurons (Bjerknes and Cheng 2001;
`Lovshin et al. 2001; Munroe et al. 1999; Yusta, Huang et al.
`2000). GLP-2R expression has been localized to distinct subsets
`of enteroendocrine cells in the human stomach and both small
`and large intestine, implying that GLP-2R receptor signaling in
`gut endocrine cells stimulates the activation and/or repression
`of endocrine-derived downstream mediators of GLP-2 action
`(Yusta, Huang et al. 2000).
`
`GENOMIC ORGANIZATION
`The human, mouse, and rat glucagon receptor genes contain
`13 exons, and the human gene has been mapped to chromo-
`some 17q24 (Burcelin et al. 1995; Lok et al. 1994; Maget et al.
`1994), with the relative location of introns conserved among
`members of the receptor superfamily. Although RT-PCR stud-
`ies have identified incompletely processed glucagon receptor
`transcripts, with the potential to give rise to variant glucagon re-
`ceptor proteins (Maget et al. 1994), variant receptor transcripts
`have not been detected in subsequent studies (Dunphy et al.
`1998; Hansen et al. 1995), hence the significance of potential
`glucagon receptor splice variants requires further clarification.
`The human GIPR gene contains 14 exons (Yamada et al.
`1995), whereas the rat gene contains 16 exons (Boylan et al.
`1999). The human GIPR gene is located on chromosome 19q13.3
`(Gremlich et al. 1995). As is the case with the glucagon, GLP-1,
`0
`and GLP-2 receptor genes (Geiger et al. 2000), the GIPR 5
`-
`flanking region contains several Sp1 binding sites, but no func-
`tional TATA or CAAT boxes (Boylan et al. 1999; Geiger et al.
`2000; Lovshin et al. 2001; Wildhage et al. 1999). The human
`GLP-1R gene is located on band 21 of chromosome 6 (Stoffel
`et al. 1993), whereas the GLP-2R gene has been localized to hu-
`man chromosome 17p13.3 (Munroe et al. 1999). The structural
`organization of the GLP-1 and GLP-2 receptor genes remains
`incompletely characterized.
`
`ALTERNATIVE RNA SPLICING
`RNA splicing resulting in functionally distinct receptor iso-
`forms has not been conclusively proven for any of the glucagon-
`receptor related receptors with the exception of the GIPR. Ge-
`nomic analysis of the GIPR identified one exon, designated
`#8a, that appears to be alternatively spliced, resulting in a vari-
`ant mRNA transcript that encodes a truncated receptor (Boylan
`et al. 1999). Although the functional properties of the variant
`receptor are unknown, an internally deleted GIPR, missing nu-
`cleotides 793-924, that does not bind GIP in transient expression
`assays, has been isolated (Gremlich et al. 1995). A second mutant
`
`Page 4
`
`

`
`STRUCTURE-FUNCTION OF G PROTEIN–COUPLED RECEPTORS
`
`183
`
`receptor cDNA, isolated from human insulinoma cells, lacks an
`internal 62 bp exon, resulting in a premature stop codon that
`encodes a truncated receptor RNA (Volz et al. 1995). Similar
`variant GIP receptor transcripts were detected in RNA from en-
`dothelial cell lines (Zhong et al. 2000). In contrast, a second
`human GIPR with a 27 amino acid insertion in the C-terminal
`tail appears to be fully functional (Gremlich et al. 1995); how-
`ever, the tissue distribution and functional significance of this
`receptor has not been examined in human tissues in vivo.
`
`SIGNALING/G PROTEIN INTERACTIONS
`Glucagon activates multiple G protein–mediated signal trans-
`duction pathways in liver cells, leading to stimulation of ade-
`nylyl cyclase (Pohl et al. 1969) and phosphoinositol turnover
`(Wakelam et al. 1986). The importance of dual signaling sys-
`tems is exemplified by studies with glucagon analogues that fail
`to stimulate cAMP formation, yet activate glycogenolysis and
`gluconeogenesis, via stimulation of glucagon receptor signal-
`ing, coupled to inositol phosphate production (Wakelam et al.
`1986). Although both glucagon and fl-adrenergic agonists stim-
`ulate adenylyl cyclase in liver cells, the glucagon receptor is
`coupled to both Gsfi-s and Gsfi-L whereas hepatic fl-adrenergic
`receptors are primarily coupled to Gsfi-L (Yagami 1995). Gener-
`ation of the (H178R) mutation in the glucagon receptor results in
`increased levels of basal cAMP and increased glucagon binding
`affinity (Hjorth et al. 1998). However, glucagon-mediated stimu-
`lation of cAMP or intracellular calcium appears to be dependent
`on amino acid sequences residing within the second and third
`intracellular loops (Cypess et al. 1999). Furthermore, deletional
`studies have demonstrated that the majority of carboxytermi-
`nal amino acid sequences distal to TM7 are not required for
`glucagon binding or adenylyl cyclase activation (Unson et al.
`1995).
`Differential interaction of the glucagon receptor with specific
`Gs isoforms has been detected in studies using GR-Gfis fusion
`proteins. The affinity of the wild-type GR for glucagon (»IC50
`of »46 nM) was comparable to the affinity observed with the
`GR-Gfis-S hybrid receptor, whereas the GR-Gfis-L receptor ex-
`hibited a lower IC50 for glucagon binding of »3.2 nM and a
`fourfold higher IC50 for adenylyl cyclase activation compared
`to GR-Gfis-L (Unson et al. 2000). The higher affinity reversed
`in the presence of GTP(cid:176) S, suggesting that specific coupling of
`the GR to distinct Gs isoforms may account for the dual class of
`high and low affinity glucagon receptors described in previous
`studies (Horwitz et al. 1985, 1986).
`Although coupling of the GLP-1R to the adenylyl cyclase
`pathway has been well established (Drucker et al. 1987), GLP-1
`also activates phospholipase C and MAPK in some cell types
`(Montrose-Rafizadeh et al. 1999; Wheeler et al. 1993), and stim-
`ulates calcium influx leading to increases in cytosolic free cal-
`cium [Ca2C
`]i. Consistent with this observation, the GLP-1R cou-
`ples to multiple G proteins, including Gfis, Gfiq=11 and Gfii1;2
`(Montrose-Rafizadeh et al. 1999) and a GLP-1R-G protein in-
`teraction domain includes amino acid sequences within the third
`
`intracellular loop (Hallbrink et al. 2001). Consistent with these
`findings, structural comparison of the GLP-1R, with related
`members of the GPCR superfamily, implicated the sequence
`K334-L335-K336 in the 3rd intracellular loop as a potential site
`for G protein coupling, and deletion of these amino acids abol-
`ishes both coupling to adenylyl cyclase and insulin secretion
`(Salapatek et al. 1999; Takhar et al. 1996). Alanine scanning
`mutagenesis further identified amino acids V327, I328, V331,
`and K334 as essential for the stimulation of adenylyl cyclase by
`GLP-1 (Mathi et al. 1997). Interestingly, 3-dimensional model-
`ing of the 3rd intracellular loop demonstrates that these amino
`acids form one face of an fi-helix, similar to that reported for the
`G protein coupling region of the m5 muscarinic receptor (Hill-
`Eubanks et al. 1996). The importance of amino acids within
`the third intracellular loop for G protein coupling is further
`emphasized by experiments using synthetic peptide sequences
`that demonstrate activation of both pertussis- and cholera toxin-
`sensitive G proteins as assessed by measurement of GTPase
`activity (Hallbrink et al. 2001).
`GIP also stimulates adenylyl cyclase activation in islet and
`intestinal cells and in heterologous cells expressing a transfected
`receptor (Amiranoff et al. 1984; Emami et al. 1986). GIP in-
`creases intracellular calcium in endothelial and some (Wheeler
`et al. 1995) but not all (Gremlich et al. 1995; Volz et al. 1995)
`cell lines transfected with the GIPR cDNA. A T340P mutation
`in the rat GIP receptor expressed in human embryonal kidney
`cells results in constitutive activation of adenylyl cyclase yet re-
`tention of GIP responsivity (Tseng and Lin 1997). Intriguingly,
`GIP also stimulates arachidonic acid production and release in
`Chinese hamster ovary cells expressing the rat GIPR, and in islet
`flTC-3 cells (Ehses et al. 2001).
`GLP-2 receptor activation is also coupled to activation of
`cAMP-dependent pathways in heterologous cell lines transfected
`with the rat or human GLP-2R (Munroe et al. 1999; Yusta
`et al. 1999). GLP-2 activates AP-1-dependent signaling path-
`ways, immediate early gene expression, and p70 S6 kinase ac-
`tivity, but does not stimulate intracellular calcium accumulation
`in BHK cells expressing the rat GLP-2R (Yusta et al. 1999). The
`GLP-2 effects on AP-1-dependent pathways are likely indirect,
`as GLP-2-stimulated induction of AP-1 luciferase activity was
`markedly attenuated in the presence of PKA inhibition (Yusta
`et al. 1999). GLP-2R signaling in BHK-GLP-2R cells is also
`coupled to activation of an antiapoptotic signaling program in
`a PKA-, phosphatidyl inositol 3-kinase-, and mitogen-activated
`protein kinase-independent manner (Boushey et al. 2001; Yusta,
`Boushey et al. 2000).
`
`RECEPTOR DESENSITIZATION
`The glucagon receptor undergoes both homologous and het-
`erologous desensitization in hepatocytes in vitro, and protein
`kinase C–selective inhibitors abrogate the heterologous desen-
`sitization process (Savage et al. 1995). Agonist occupancy of
`related hormone receptors, coupled to adenylyl cyclase in hep-
`atocytes, results in heterologous desensitization with kinetics
`
`Page 5
`
`

`
`184
`
`P. L. BRUBAKER AND D. J. DRUCKER
`
`than those observed for homologous
`slower
`somewhat
`desensitization (Premont and Iyengar 1988). Both homologous
`and heterologous glucagon receptor desensitization are indepen-
`dent of increases in intracellular calcium (Savage et al. 1995);
`receptor desensitization is also independent of cAMP activa-
`tion and is induced by activators of inositol phospholipid
`metabolism such as protein kinase C, independent of the in-
`hibitory guanine nucleotide regulatory protein Gi. (Murphy et al.
`1987, 1989).
`Prolonged activation of the GLP-1R by GLP-1 agonists
`results in a diminished response to subsequence ligand bind-
`ing in vitro (Fehmann and Habener 1991). Mutagenesis studies
`have identified three pairs of serine residues in the C-terminal
`tail of the receptor (S441/S442, S444/S445, and S451/S452)
`important for ligand-induced desensitization of the GLP-1R
`(Widmann et al. 1996a, 1996b, 1997). These amino acids are
`also necessary for internalization of the receptor, while phos-
`phorylation of an additional serine pair (S431/S432) by pro-
`tein kinase C appears to be involved in heterologous desensiti-
`zation of the GLP-1R (Widmann et al. 1996a, 1996b, 1997). It
`has been suggested that this heterologous desensitization may
`occur in the fl cell in response to ligand-mediated activation
`of phospholipase C, such as occurs in response to physiologi-
`cal regulators, including acetylcholine (Widmann et al. 1996b).
`Nevertheless, diminution of GLP-1 responsivity has not yet
`been clearly described in vivo, suggesting that other compen-
`satory mechanisms may serve to protect the receptor from
`downregulation.
`Homologous and/or heterologous desensitization of the GIPR
`may account for the reduced response to GIP that has been ob-
`served in patients with type 2 diabetes (Elahi et al. 1994; Nauck
`et al. 1993). The GIPR undergoes homologous desensitization
`(Fehmann and Habener 1991; Tseng, Boylan et al. 1996; Tseng
`and Zhang 2000), while incubation of islets with high glucose
`also reduces the response to GIP in a manner that is independent
`of protein kinases A and C (Hinke et al. 2000). The rat GIPR
`possesses only a single pair of serine residues in its C-terminal
`tail (S426/S427), and a combination of site directed mutagenesis
`and carboxyterminal deletions have implicated a role for these
`residues in receptor internalization, whereas S406 and C411
`contribute to receptor desensitization (Tseng and Zhang 1998;
`Wheeler et al. 1999). Cotransfection of a cDNA encoding fl-
`arrestin-1, or the G protein–coupled receptor kinase 2 (GRK2),
`but not GRK5 or GRK6, attenuated GIP-stimulated cAMP ac-
`cumulation in L293-GIPR and flTC3 cells (Tseng and Zhang
`2000). However, none of these proteins appear to be involved in
`GIPR internalization.
`In contrast to the information available for the glucagon,
`GLP-1, and GIP receptors, GLP-2 receptor desensitization has
`not yet been examined in cells expressing a GLP-2R in vitro.
`Although a diminished response to supraphysiological levels of
`GLP-2 is apparent in heterologous cells expressing the GLP-2R
`(Lovshin et al. 2001), mice treated with daily injections of GLP-
`2 for 12 weeks continued to exhibit enhanced intestinal mass,
`
`consistent with a lack of GLP-2R desensitization in vivo (Tsai,
`Hill, Asa et al. 1997).
`
`MOLECULAR BASIS OF LIGAND
`RECEPTOR-INTERACTION
`A series of N-terminal truncation and internal deletion mu-
`tants has been used to demonstrate that glycosylation is unlikely
`to be important for glucagon receptor intracellular trafficking
`and plasma membrane insertion (Unson et al. 1995). Remark-
`ably, deletion of the intracellular carboxyterminal tail of the rat
`receptor does not abrogate glucagon binding or adenylyl cy-
`clase generation in transiently transfected COS-1 or CHO cells
`(Bu

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket