throbber
Proc. Natl. Acad. Sci. USA
`Vol. 96, pp. 1569–1573, February 1999
`Medical Sciences
`
`Prototypic G protein-coupled receptor for the intestinotrophic
`factor glucagon-like peptide 2
`DONALD G. MUNROE*†, ASHWANI K. GUPTA*, FATEMEH KOOSHESH*, TEJAL B. VYAS*, GEIHAN RIZKALLA*,
`HONG WANG*, LIDIA DEMCHYSHYN*, ZHI-JIE YANG*, RAJENDER K. KAMBOJ*, HONGYUN CHEN*,
`KIRK MCCALLUM*, MARTIN SUMNER-SMITH*‡, DANIEL J. DRUCKER§, AND ANNA CRIVICI*¶
`*Allelix Biopharmaceuticals Inc., 6850 Goreway Drive, Mississauga, Ontario L4V 1V7, Canada; and §Department of Medicine, The Toronto Hospital, Banting and
`Best Diabetes Centre, University of Toronto, Toronto, Ontario M5G 2C4, Canada
`
`Edited by Donald F. Steiner, The University of Chicago, Chicago, IL, and approved November 18, 1998 (received for review August 18, 1998)
`
`Glucagon-like peptide 2 (GLP-2) is a 33-aa
`ABSTRACT
`proglucagon-derived peptide produced by intestinal enteroen-
`docrine cells. GLP-2 stimulates intestinal growth and up-
`regulates villus height in the small intestine, concomitant with
`increased crypt cell proliferation and decreased enterocyte
`apoptosis. Moreover, GLP-2 prevents intestinal hypoplasia
`resulting from total parenteral nutrition. However, the mech-
`anism underlying these actions has remained unclear. Here we
`report the cloning and characterization of cDNAs encoding rat
`and human GLP-2 receptors (GLP-2R), a G protein-coupled
`receptor superfamily member expressed in the gut and closely
`related to the glucagon and GLP-1 receptors. The human
`GLP-2R gene maps to chromosome 17p13.3. Cells expressing
`the GLP-2R responded to GLP-2, but not GLP-1 or related
`peptides, with increased cAMP production (EC50 5 0.58 nM)
`and displayed saturable high-affinity radioligand binding (Kd
`5 0.57 nM), which could be displaced by synthetic rat GLP-2
`(Ki 5 0.06 nM). GLP-2 analogs that activated GLP-2R signal
`transduction in vitro displayed intestinotrophic activity in vivo.
`These results strongly suggest that GLP-2, like glucagon and
`GLP-1, exerts its actions through a distinct and specific novel
`receptor expressed in its principal target tissue, the gastro-
`intestinal tract.
`
`Glucagon-like peptides (GLPs) encoded by the proglucagon
`gene play key roles in glucose homeostasis, gastric emptying,
`insulin secretion, and appetite regulation (1). Glucagon and
`GLP-1 exert their effects through distinct G protein-coupled
`receptors (GPCRs). In contrast, unique receptors for GLP-2,
`glicentin, and oxyntomodulin have not yet been identified,
`despite considerable attempts at receptor isolation via classical
`molecular biology approaches (2). Recent studies have shown
`that GLP-2 is a potent intestinal growth factor that stimulates
`crypt cell proliferation and inhibits epithelial apoptosis (3).
`GLP-2 promotes epithelial proliferation in both small and
`large intestine; however, the mechanisms utilized by GLP-2 for
`promotion of intestinal growth remain unclear.
`To understand the mechanisms underlying GLP-2 action, we
`have carried out studies directed at the identification and
`cloning of the putative GLP-2 receptor. We now have isolated
`rat and human cDNAs encoding GLP-2-responsive GPCRs,
`which show highest similarity to receptors for glucagon and
`GLP-1. The GLP-2R is coupled to activation of adenylate
`cyclase, and the receptor is expressed selectively in rat hypo-
`thalamus and the gastrointestinal tract, known targets of
`GLP-2 action. These findings establish GLP-2 as a novel
`hormone that, like glucagon and GLP-1, exerts its actions
`through a distinct receptor expressed in a highly tissue-
`restricted manner. The GLP-2R should provide an important
`
`The publication costs of this article were defrayed in part by page charge
`payment. This article must therefore be hereby marked ‘‘advertisement’’ in
`accordance with 18 U.S.C. §1734 solely to indicate this fact.
`PNAS is available online at www.pnas.org.
`
`target for isolation of small molecules mimicking GLP-2 action
`and for future studies delineating specific mechanisms under-
`lying GLP-2 action in the gut and central nervous system.
`
`Methods and Materials
`Primers, cDNA Libraries, and Cloning Strategy. Initial
`attempts at low-stringency hybridization of intestine and brain
`cDNA libraries using GLP-1RyGlucagonR cDNA sequences
`were not successful. Two million cDNA clones from rat
`hypothalamus and rat duodenumyjejunum cDNA libraries
`subsequently were screened with degenerate oligonucleotides
`derived from conserved transmembrane II and VII GPCR
`coding sequences: C4–4 (59-AACTACATCCACMKGM
`AYCTGTTYVYGTCBTTCATSCT-39) (IUB nomenclature)
`and C9 –2R (59-TCYRNCTGSACCTCMYYRTTGAS-
`RAARCAGTA-39) (for nomenclature, see ref. 4). First-round
`cDNA plugs (1,057) were isolated in this screen. In a comple-
`mentary strategy, PCR was conducted on intestinal cDNA
`templates by using sets of degenerate PCR primers, based on
`conserved transmembrane amino acid motifs from family B
`GPCRs or from motifs conserved mainly within the glucagony
`glucose-dependent insulinotropic polypeptide (GIP)yGLP-1
`receptor subfamily. PCR products were Southern-blotted and
`probed with 32P-end-labeled C4–4 oligonucleotide. PCRs,
`amplified from rat neonatal
`intestine cDNA (Stratagene;
`catalog no. 936508) were chosen for cloning. These products
`had been amplified with the degenerate primers M2F (59-
`TTTTTCTAGAASRTSATSTACACNGT SGGCTAC-39 )
`(based on conserved transmembrane domain I sequences) and
`M7R (59-TTTTCTCGAGCCARCARCCASSWRTART-
`TGGC-39) (based on conserved transmembrane III sequenc-
`es). PCR products were cloned into pBluescript, screened by
`filter hybridization with the nested C4-4 oligonucleotide, and
`sequenced, leading to the identification of a sequence frag-
`ment from a novel GPCR family B member, designated WBR,
`that ultimately proved to be the GLP-2R. Two new GLP-2R-
`specific PCR primers, P23-F1 (59-TCTGACAGATATGA-
`CATCCATCCAC-39) and P23-R1 (59-TCATCTCCCTCT-
`TCTTGGCTCTTAC-39), were used to screen the 1,057 cDNA
`plugs obtained by hybridization screening,
`leading to the
`
`This paper was submitted directly (Track II) to the Proceedings office.
`Abbreviations: EBNA, Epstein–Barr nuclear antigen; GPCR, G pro-
`tein-coupled receptor; GLP-1 and -2, glucagon-like peptide 1 and 2,
`respectively; GLP-2R, GLP-2 receptor; GIP, glucose-dependent insu-
`linotropic polypeptide.
`Data deposition: The sequences reported in this paper have been
`deposited in the GenBank database (accession nos. AF105367 and
`AF105368).
`†To whom reprint requests should be addressed. e-mail: dmunroe@
`allelix.com.
`‡Present address: Base4 Bioinformatics, 6299 Airport Road, Missis-
`sauga, Ontario L4V 1N3, Canada.
`¶Present address: Ligard Pharmaceuticals, Inc. 10275 Science Center
`Drive, San Diego, CA 92121.
`
`1569
`
`CFAD Exhibit 1022
`
`1
`
`

`
`1570
`
`Medical Sciences: Munroe et al.
`
`Proc. Natl. Acad. Sci. USA 96 (1999)
`
`FIG. 1. Multiple alignment of the human (GL2RoHUMAN) and rat (GL2RoRAT) GLP-2R amino acid sequences with human GLP-1, GIP,
`and glucagon receptor sequences. Known receptor sequences are designated by Swiss-Prot identifiers. Alignment was performed with CLUSTAL W
`1.60 and rendered with GENEDOC 2.2. Identities of rat and human GLP-2R sequences is shown in gray, and identities across all five receptor members
`are indicated by black shading. A predicted signal-peptide cleavage site in human and rat GLP-2R is indicated by an inverted triangle. Six conserved
`cysteine residues are indicated by arrows. Seven predicted transmembrane domains are shown as solid, black boxes labeled with Roman numerals,
`and asterisks are shown for spacing every 20 aa. The GenBank accession numbers for the human and rat GLP-2R sequences are AF105367 and
`AF105368, respectively.
`
`identification of three independent clones, two from the
`duodenumyjejunum library and a third from hypothalamus,
`which, together, contained a 2,537-bp cDNA insert encoding
`full-length rat GLP-2R.
`To clone the human GLP-2R, the coding region of the rat
`GLP-2R was used to screen a human hypothalamus cDNA
`library (CLONTECH; catalog no. 1172a), from which a single
`positive clone (HHT13) was isolated and sequenced. A full-
`length insert was ligated into pcDNA3 for expression studies.
`Intestinotrophic Activity, cAMP Determination, and Radi-
`oligand-Binding Studies. For cAMP assays, an episomal stable
`
`cell line was prepared by lipofection of 293-EBNA (Epstein–
`Barr nuclear antigen) cells (Invitrogen) with a pREP7-based
`(Invitrogen) construct containing the Met-42 3 Ile-550 ORF
`of rGLP-2R. Parental 293-EBNA cells, as well as the stable cell
`line rG2R, expressed receptors for vasoactive intestinal
`polypeptide and pituitary adenylate cyclase-activating
`polypeptide. Therefore, the ligand specificity of GLP-2R was
`tested further in transiently transfected COS cells, which
`expressed no functional receptors for any of the ligands tested.
`For cAMP assays, cells were treated at 80% confluency with
`GLP-2 peptide analogs at concentrations ranging from 10212
`
`2
`
`

`
`Medical Sciences: Munroe et al.
`
`Proc. Natl. Acad. Sci. USA 96 (1999)
`
`1571
`
`to 1025 M for 30 min in medium containing 3-isobutylmeth-
`ylxanthine. The reaction was terminated with the addition of
`95% ethanol and 5 mM EDTA. Aliquots of the ethanol extract
`were used to determine cAMP levels using an enzyme immu-
`noassay kit (Amersham) as described by the manufacturer.
`Results were analyzed with GRAPHPAD PRISM software and
`expressed as pmol cAMP per well. For radioligand-binding
`assays, cells expressing GLP-2R were harvested and homog-
`enized in 25 mM Hepes (pH 7.4) buffer containing 140 mM
`NaCl, 0.9 mM MgCl2, 5 mM KCl, 1.8 mM CaCl2, 17 mg/ml
`Diprotin A, and 100 mM phenanthroline. Homogenates were
`centrifuged for 10 min at 1,000 3 g at 4°C to remove cellular
`debris. For saturation experiments, membranes containing 25
`mg protein were incubated with increasing concentrations of
`125I-[Tyr-34]GLP-2 (5–2,000 pM final concentration) in a
`volume of 0.5 ml for 2 hr at 4°C. Nonspecific binding was
`determined by the addition of 10 mM of native rat GLP-2 and
`subtracted from total binding to estimate specific binding to
`GLP-2R. Parallel experiments confirmed the lack of specific
`binding when the GLP-2R expression construct was not used.
`For competition-binding experiments, assays were initiated by
`the addition of 200 pM (final concentration) of 125I-
`[Tyr34]GLP-2 with increasing concentrations of competing
`peptide analogs (10211 to 1025 M) for 2 hr as described above.
`Reactions were terminated by centrifugation at 13,000 3 g for
`15 min at 4°C. The pellets were washed three times with cold
`50 mM Tris buffer, and radioactivity was quantitated in a
`gamma counter. Results were analyzed by GRAPHPAD PRISM
`software.
`Intestinotrophic activities of various peptide analogs were
`determined by assessment of small bowel weight as described
`(5), after 14-day treatments with 2.5 mg of test peptide or PBS
`(vehicle-treated control) administered twice daily. Activity
`was defined as follows: active, small bowel wet weight 40–70%
`greater than in vehicle-treated control animals; partially active,
`20–40% greater than controls; inactive, less than 20% greater
`than controls.
`RNAse Protection Assay. A fragment of GLP-2R cDNA was
`subcloned into pBluescript (Stratagene) for in vitro transcrip-
`tion with T3 or T7 RNA polymerases. The probe, called F1,
`spanned nucleotides encoding amino acids Met-1 3 Arg-210.
`RNase protection assay was carried out essentially as described
`(6), using 50 mg of total RNA from adult rat tissues or 50 mg
`of yeast tRNA (negative control) or tRNA spiked with a
`known copy number of sense-strand cRNA (for standard curve
`construction). Each sample was hybridized with 100,000 cpm
`of [32P]CTP-labeled antisense cRNA and then digested with
`RNases T1 (140 unitsyml) and A (8 mgyml) at 30 C for 1 hr.
`The deproteinized, ethanol-precipitated probe was run on a
`5% sequencing gel and analyzed after PhosphorImaging (Mo-
`lecular Dynamics) with IMAGEQUANT software. RNA copy
`number was calculated by interpolation relative to the standard
`curve after taking the lengths and specific activities of undi-
`gested and digested probes into account. A second RNase
`probe from a different region of the GLP-2R cDNA was used
`to confirm the quantitative results (data not shown).
`
`RESULTS AND DISCUSSION
`GLP-2 and the peptide hormones GLP-1, glucagon, and GIP
`have closely related amino acid sequences (7). Similarly, the
`sequences of cloned receptors for the latter three peptides
`form a cluster within the parathyroid hormone receptor-like
`GPCRs family B (8–11), suggesting that the GLP-2 receptor
`might also be found within this subfamily. Initial attempts at
`GLP-2 receptor cloning by conventional screening of cDNA
`libraries at low stringency with a combination of GLP-1 and
`glucagon receptor cDNA probes were not successful. Accord-
`ingly, we next used a combined approach of reverse transcrip-
`tion–PCR and hybridization screening followed by expression
`
`FIG. 2. Ligand-selective and concentration-dependent cAMP re-
`sponse to rat GLP-2 in transiently lipofected COS cells. (A) Ligand
`specificity of cAMP response to GLP-2. cAMP response to peptide
`analogs of family 2 GPCR ligands was determined in COS cells
`transiently lipofected with the GLP-2R expression vector p587-70 or
`the parental pcDNA3 expression vector. GLP-2 concentration was 1
`nM; all other peptides were used at 10 nM. A similar profile of peptide
`specificity was observed with the human GLP-2R (data not shown).
`(B) Concentration–response curve for cAMP accumulation in re-
`sponse to synthetic rat GLP-2 in rG2R cells stably expressing GLP-2R.
`
`analysis of candidate cDNAs. As described in Methods and
`Materials, this strategy resulted in the isolation of a 2,537-bp rat
`GLP-2R cDNA insert encoding a 550-aa putative family B
`GPCR (Fig. 1). Hydropathy analysis of the GLP-2R amino acid
`sequence revealed a typical 7-transmembrane topology plus a
`hydrophobic amino-terminal signal peptide (data not shown).
`The GLP-2R gene product belongs to the GLP-1yglucagony
`GIP receptor gene subfamily. Conserved features include a
`possible signal-peptide cleavage site between Val-64 and Thr-
`65, potential N-glycosylation sites within the amino-terminal
`putative extracellular domain, and six cysteine residues con-
`served in the mature GLP-1, GIP, and glucagon receptor
`amino-terminal domains (Fig. 1). Two putative alternative
`translation initiation codons, Met-1 and Met-42, were found
`amino-terminal to the first transmembrane domain in the rat
`GLP-2R. Functional analysis of Met-1 and Met-42 site-
`directed mutants showed they were functionally identical
`(unpublished results), consistent with signal-peptide removal
`predicted to yield an identical 486-aa mature polypeptide.
`Overlapping regions of the hypothalamus and duodenumy
`jejunum cDNA clones encoded homologous polypeptide frag-
`ments. Moreover, no evidence was obtained for differential
`splicing of intestinal GLP-2R RNA from an RNase protection
`assay employing two nonoverlapping probes derived from
`GLP-2R cDNA, which together spanned 387 of the 550
`GLP-2R codons (data not shown). Additionally, sequencing of
`
`3
`
`

`
`1572
`
`Medical Sciences: Munroe et al.
`
`Proc. Natl. Acad. Sci. USA 96 (1999)
`
`GLP-2-binding sites on these cells was shown by using a radio-
`iodinated, C-terminally extended GLP-2 analog, 125I-[Tyr-
`34]GLP-2 (Fig. 3A). From Scatchard analysis of the saturation
`isotherms, a Bmax value of 1,839 fmolymg of protein and a Kd of
`0.57 nM were obtained for the radioligand. Mock-transfected
`cells showed no specific binding to the radioligand (data not
`shown). Competition-binding studies with rat GLP-2 revealed a
`high-affinity site (Ki 5 0.06 nM) and a low-affinity site (Ki 5 259
`nM) (Fig. 3B). In contrast, no high-affinity GLP-1 sites were
`observed in the transfected GLP-2RyrG2R clone. Ki values
`determined for GLP-1, glucagon, and GIP peptides were 928,
`500, and 765 nM, respectively. Although the binding and Scat-
`chard data may reflect, in part, a degree of receptor overexpres-
`sion, the functional studies and binding data provide firm evi-
`dence for a cDNA that encodes a functional high-affinity, ligand-
`selective GLP-2 receptor.
`The human GLP-2R polypeptide showed 81.6% similarity to
`rat GLP-2R (Fig. 1). Functional expression of the cloned human
`GLP-2R conferred a functional response to GLP-2 and the
`appearance of high-affinity ligand-binding sites on 293-EBNA
`cells, similar to data obtained with the rat GLP-2R (data not
`shown). Furthermore, the cloned human GLP-2 receptor exhib-
`ited the same profile of peptide-binding specificity (Fig. 2A and
`unpublished data) as the rat receptor. The gene encoding human
`GLP-2R was identified by screening an arrayed BAC library of
`human genomic DNA (Genome Systems, St. Louis), confirmed
`by sequencing, and mapped to chromosome 17p13.3 by fluores-
`cence in situ hybridization analysis (data not shown).
`A quantitative ribonuclease protection assay method was
`used to determine the tissue distribution of rat GLP-2R RNA
`because no signals were detected on multitissue Northern
`blots. GLP-2R RNA levels were highest in jejunum, followed
`by duodenum, ileum, colon, and stomach, whereas expression
`was undetectable in seven other tissues (Table 1). This expres-
`sion pattern is clearly concordant with previously reported
`functional responses to GLP-2 in duodenum (12, 13), jejunum,
`ileum (5, 12, 14, 15), and colon (12, 16, 17); in contrast, no
`proliferative or histological changes were seen after GLP-2
`treatment in spleen, heart, kidney, lung, or brain (18). Thus,
`GLP-2R expression is detected in known GLP-2 target tissues.
`This observation, together with the functional data from
`
`Table 1. Quantitative GLP-2R RNA distribution in various rat
`tissues determined by RNase protection
`b-Actin
`F1 quantitation,
`quantitation,
`GLP-2Ry-actin,
`copies per mg
`copies per mg
`ratio
`total RNA
`total RNA
`Tissue
`76.8 3 1025
`15,500,000
`11,900
`Jejunum
`10.7 3 1025
`85,700,000
`9,150
`Duodenum
`14.6 3 1025
`51,400,000
`7,490
`Ileum
`21.0 3 1025
`19,800,000
`4,150
`Colon
`6.48 3 1025
`23,600,000
`1,530
`Stomach
`,1.48 3 1025
`,600
`40,600,000
`Brain
`,9.09 3 1025
`,600
`6,600,000
`Heart
`,4.03 3 1025
`,600
`14,900,000
`Kidney
`,3.59 3 1025
`,600
`16,700,000
`Liver
`,1.56 3 1025
`,600
`38,500,000
`Lung
`,13.0 3 1025
`,600
`4,600,000
`Muscle
`,1.34 3 1025
`,600
`44,800,000
`Spleen
`Total RNA (50 mg) from rat tissues or sense-strand cRNA standards
`was hybridized to radiolabeled antisense cRNA probes prepared in
`vitro from GLP-2R cDNA (F1) or actin cDNA. After RNase digestion
`as described in Methods and Materials, protected probe was precipi-
`tated, electrophoresed, and quantitated by PhosphorImage analysis
`relative to the standard curve obtained from sense-strand cRNA. RNA
`quantitation is expressed as copy number per mg of total RNA.
`GLP-2R RNA copy number was detectable to a lower limit of 30,000
`copies per 50 mg sample, setting the limit of detection shown above for
`nongastrointestinal tissues.
`
`FIG. 3. Binding of 125I -[Tyr-34]GLP-2 to cell membranes prepared
`from rG2R cells stably transfected with GLP-2R cDNA. (A) Satura-
`tion isotherms of the specific binding of 125I -[Tyr-34]GLP-2 to
`membranes. Results shown are representative of six independent
`experiments, each conducted in triplicate. From Scatchard analysis
`(Inset), maximal-binding Bmax was estimated at 1,839 fmolymg protein,
`and a Kd value of 0.57 nM was obtained. (B) Competition binding of
`125I-[Tyr-34]GLP-2 binding to cell membranes in the presence of
`unlabeled peptides. Data are shown for the concentration-dependent
`inhibition of 125I-[Tyr-34]GLP-2 binding (200 pmol) to GLP-2R by
`various peptide analogs from at least two independent experiments
`conducted in triplicate. Inhibitory constants (Ki) were estimated by
`using GRAPHPAD PRISM.
`
`the full-length human GLP-2R cDNAs confirmed the identity
`of hypothalamic and gastrointestinal GLP-2Rs (Fig. 1).
`To assess whether the predicted GLP-2R sequence encodes
`a functional GLP-2 receptor, a GLP-2R expression construct,
`p587-70, was transiently transfected into COS cells. Because
`related family B GPCRs show functional coupling to cAMP
`production mediated by the heterotrimeric G protein Gs,
`cAMP accumulation was measured after incubation with
`GLP-2. Treatment of GLP-2R-transfected COS cells with 1
`nM GLP-2 resulted in a 4-fold rise in cAMP levels relative to
`untreated cells, approximately equal to the response seen with
`10 mM forskolin (Fig. 2A). Treatment with 10 nM GLP-1,
`glucagon, GIP, exendin-3, and seven other family B GPCR
`ligands did not
`induce cAMP production in p587-70-
`transfected cells. Control cells transfected with vector DNA
`alone failed to respond to any of the peptides tested, including
`GLP-2, though forskolin did induce cAMP production.
`With a stable GLP-2R episomal expression cell line, rG2R,
`greater than 20-fold cAMP induction routinely was achieved with
`1 or 10 nM GLP-2 (data not shown), confirming the potent
`induction of cAMP accumulation by GLP-2. In contrast, the
`nontransfected 293-EBNA cells showed no response to GLP-2.
`The EC50 of the cAMP response to GLP-2 in rG2R cells was 0.58
`nM (Fig. 2B). The presence of saturable, specific, ligand-selective
`
`4
`
`

`
`Medical Sciences: Munroe et al.
`
`Proc. Natl. Acad. Sci. USA 96 (1999)
`
`1573
`
`Table 2.
`
`In vitro and in vivo activity profiles of selected peptide analogs of GLP-2
`Ki, nMa*
`
`In vivo
`activity§
`Active
`Partially active
`Inactive
`Active
`Active
`Inactive
`Inactive
`Partially active
`Inactive
`Active
`Active
`Inactive
`Inactive
`Inactive
`
`Low-afinity
`High-affinity
`Peptide
`259 6 46
`0.06 6 0.00
`rGLP-2(1-33)¶
`140 6 2
`—
`N-Ac-rGLP-2(1-33)
`[Arg-1]rGLP-2(21-33)
`n*
`NA
`n§
`ND
`[Arg-34]rGLP-2(1-34)
`255 6 7
`0.56 6 0.3
`[Tyr-34]hGLP-2
`876 6 147
`—
`rGLP-2(2-33)
`251 6 8
`—
`rGLP-2(3-33)
`584 6 19
`0.30 6 0.00
`rGLP-2(1-29)
`977 6 470
`—
`[Thr-7 insertion]
`126 6 5
`—
`[Gly-2]GLP-2(1-33)
`596 6 9
`1.7 6 0.4
`hGLP-2(1-33)¶
`500 6 332
`—
`Glucagon
`928 6 1
`—
`GLP-1(7-36)amide
`765 (n 5 1)
`—
`GIP
`NA, not active—no detectable binding; ND, not determined.
`*n 5 2, except where indicated.
`†n 5 3.
`‡Relative to 100 nM rGLP-2(1-33), n 5 3.
`§Relative to vehicle-treated control animals, n 5 4 or greater. In vivo activity is based on changes in small bowel wet weight after 14-day treatment
`as described in Methods and Materials.
`¶rGLP-2(1-33) is native rat GLP-2 peptide; hGLP-2(1-33) is native human GLP-2 peptide.
`
`EC50, nM†
`1.00 6 0.2
`20.8 6 0.1
`901 6 41
`3.1 6 0.3
`1.4 6 0.1
`210 6 22
`10.7 6 0.8
`3.60 6 0.4
`1,100 6 30
`2.0 6 0.2
`1.3 6 0.20
`NA
`NA
`NA
`
`Emac, %†‡
`100 6 0
`80.1 6 7
`69.0 6 2
`105 6 10
`113 6 5
`109 6 8
`115 6 5
`102 6 8
`76 6 4.0
`103 6 6
`99 6 10.6
`NA
`NA
`NA
`
`experiments with cloned GLP-2R cDNA, suggests that this
`receptor mediates the intestinotrophic actions of GLP-2.
`Pharmacological support for this hypothesis was obtained from
`parallel in vivoyin vitro studies of GLP-2 analogs containing
`simple changes in sequence and length (Table 2). Carboxyl-
`terminal extension analogs bound and activated GLP-2R and
`retained in vivo activity whereas those with amino-terminal
`extensions lost both activities. Analogs with blocked amino- or
`carboxyl-terminal residues displayed diminished in vivo activity
`and GLP-2R activation. Insertion of a Thr residue between
`GLP-2 residues 6 and 7 resulted in loss of activity in vivo and in
`vitro. Truncation of the carboxyl-terminus to a 29-residue peptide
`(analogous to glucagon) reduced but did not eliminate in vivo or
`in vitro activities. Interestingly, truncation of one or two amino-
`terminal residues abolished in vivo activity but did not completely
`eliminate binding or the GLP-2R cAMP response. Taken to-
`gether, a clear correspondence was revealed between the struc-
`tural requirements for GLP-2R binding and activation and the in
`vivo intestinotrophic activity of GLP-2, providing additional
`evidence that the GLP-2R isolated here and the intestinotrophic
`GLP-2 receptor mediating GLP-2 action in vivo are synonymous.
`Enteroglucagon synthesis long has been associated with a
`humoral adaptive response to massive small bowel resection, in
`which hyperplasia and elongation of jejunal villi are seen (19–22).
`Proglucagon-derived GLP-2 is detectable in plasma from fasted
`rats and humans and rises 1.5- to 3.6-fold after feeding (23).
`Moreover, the intestinotrophic efficacy of GLP-2 has been shown
`after administration by i.p., i.m., or s.c. routes (14), as well as by
`coinfusion in parenterally fed rats (12). Thus, it is likely that
`circulating GLP-2 mediates adaptive changes in the villus-
`absorptive area in the small intestine. The cloning and charac-
`terization of a GLP-2 receptor expressed in the gastrointestinal
`tract qualifies GLP-2, like GLP-1, glucagon, and GIP, as a bona
`fide endocrine hormone and should facilitate the discovery of
`novel pharmacologic agents with similar functional activity. The
`expression of GLP-2R in hypothalamus also raises the possibility
`of as yet undescribed role(s) for this intestinotrophic hormone in
`the central nervous system.
`
`We thank P. Khanna, Y.-D. Huang, R. Mathieson, Y.-P. Zhang, and
`E. Fan for technical assistance; and J. W. Dietrich, R. Zastawny, and
`D. Lee for advice and critical reading of this manuscript. D.J.D. is a
`consultant to Allelix Biopharmaceuticals Inc.
`
`10.
`
`1. Drucker, D. J. (1998) Diabetes 47, 159–169.
`2. McGregor, G. P., Goke, R. & Goke, B. (1998) Exp. Clin.
`Endocrinol. Diabetes 106, 25–28.
`3. Tsai, C.-H., Hill, M., Asa, S. L., Brubaker, P. L. & Drucker, D.
`J. (1997) Am. J. Physiol. 273, E77–E84.
`4. Nomenclature Committee of the International Union of Bio-
`chemistry (NC-IUB) (1986) J. Biol. Chem. 261, 13–17.
`5. Drucker, D. J., Ehrlich, P., Asa, S. L. & Brubaker, P. L. (1996)
`Proc. Natl. Acad. Sci. USA 93, 7911–7916.
`6. Ausubel, F. M., Brent, R., Kingston, R. E., Moore, D. D.,
`Seidman, J. G., Smith, J. A. & Struhl, K. (1998) Current Protocols
`in Molecular Biology (Wiley, New York).
`7. Fehmann, H.-C., Goke, R. & Goke, B. (1995) Endocrine Rev. 16,
`390–410.
`8. Thorens, B. (1992) Proc. Natl. Acad. Sci. USA 89, 8641–8645.
`9. MacNeil, D. J., Occi, J. L., Hey, P. J., Strader, C. D. & Graziano,
`M. P. (1994) Biochem. Biophys. Res. Commun. 198, 328–334.
`Jelinek, L. J., Lok, S., Rosenberg, G. B., Smith, R. A., Grant, F. J.,
`Biggs, S., Bensch, P. A., Kuijper, J. L., Sheppard, P. O., Sprecher,
`C. A., et al. (1993) Science 259, 1614–1616.
`11. Yasuda, K., Inagaki, N., Yamada, Y., Kubota, A., Seino, S. & Seino,
`Y. (1994) Biochem. Biophys. Res. Commun. 205, 1556–1562.
`12. Chance, W. T., Foley-Nelson, T., Thomas, I. & Balasubrama-
`niam, A. (1997) Am. J. Physiol. 273, G559–G563.
`13. Brubaker, P. L., Izzo, A., Hill, M. & Drucker, D. J. (1997) Am. J.
`Physiol. 272, E1050–E1058.
`14. Tsai, C.-H., Hill, M. & Drucker, D. J. (1997) Am. J. Physiol. 272,
`G662–G668.
`15. Cheeseman, C. I. (1997) Am. J. Physiol. 273, R1965–R1971.
`16. Drucker, D. J., Deforest, L. & Brubaker, P. L. (1997) Am. J.
`Physiol. 273, G1252–G1262.
`17. Litvak, D. A., Hellmich, M. R., Evers, B. M., Banker, N. A. &
`Townsend, C. M., Jr. (1998) J. Gastrointest. Surg. 2, 146–150.
`18. Tsai, C.-H., Hill, M., Asa, S. L., Brubaker, P. L. & Drucker, D. J.
`(1997) Am. J. Physiol. 273, E77–E84.
`19. Buchan, A. M. J., Griffiths, C. J., Morris, J. F. & Polak, J. M.
`(1985) Gastroenterology 88, 8–12.
`20. Rountree, D. B., Ulshen, M. H., Selub, S., Fuller, C. R., Bloom,
`S. R., Ghatei, M. A. & Lund, P. K. (1992) Gastroenterology 103,
`462–468.
`21. Gornacz, G. E., Ghatei, M. A. & Al-Mukthar, M. Y. T. (1984)
`Dig. Dis. Sci. 29, 1041–1049.
`22. Taylor, R. G. & Fuller, P. J. (1994) Baillieres Clin. Endocrinol.
`Metab. 8, 165–183.
`23. Brubaker, P. L., Crivici, A., Izzo, A., Ehrlich, P., Tsai, C.-H. &
`Drucker, D. J. (1997) Endocrinology 138, 4837–4843.
`
`5

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