throbber
THE JOURNAL OF BIOLOGICAL CHEMISTRY
`© 1998 by The American Society for Biochemistry and Molecular Biology, Inc.
`
`Vol. 273, No. 17, Issue of April 24, pp. 10308-10312, 1998
`Printed in U.S.A.
`
`Positively Charged Residuesat Positions 12, 17, and 18 of
`Glucagon Ensure Maximum Biological Potency*
`
`(Received for publication, November 7, 1997, and in revised form, February 13, 1998)
`
`Cecilia G. Unson{, Cui-Rong Wu, Connie P. Cheung, and R.B. Merrifield
`From the Rockefeller University, New York, New York 10021
`
`Glucagon is a peptide hormonethat plays a central
`role in the maintenance of normal circulating glucose
`levels. Structure-activity studies have previously dem-
`onstrated the importanceof histidine at position 1 and
`the absolute requirement for aspartic acid at position 9
`for transduction of the hormonal signal. Site-directed
`mutagenesis of the receptor protein identified Asp™ on
`the extracellular N-terminal tail to be crucial for the
`recognition function of the receptor. In addition, anti-
`bodies generated against aspartic acid-rich epitopes
`from the extracellular region competed effectively with
`glucagon for receptor sites, which suggested that nega-
`tive charges may line the putative glucagon binding
`pocket in the receptor. These observations led to the
`idea that positively charged residues on the hormone
`may act as counterions to these sites. Based on these
`initial findings, we synthesized glucagon analogs in
`which basic residues at positions 12, 17, and 18 were
`replaced with neutral or acidic residues to examine the
`effect of altering the positive charge on those sites on
`binding and adenylyl cyclase activity.
`Theresults indicate that unlike N-terminal histidine,
`Lys!”, Arg!’, and Arg?® of glucagon havevery largeef-
`fects on receptor binding and transduction of the hor-
`monal signal, although they are not absolutely critical.
`They contribute strongly to the stabilization of the bind-
`ing interaction with the glucagon receptor that leads to
`maximum biological potency.
`
`Glucagonis a polypeptide hormonethatconsists of 29 amino
`acid residues and is a memberof a highly homologous family of
`biologically active peptides. Secreted by pancreatic A cells, its
`primary target organis the liver where, together with insulin,
`it plays a central role in the maintenance of normalcirculating
`glucose levels critical to the survival of the organism. The
`initial event in glucagonaction is bindingto its receptor on the
`surface of liver cells. The binding message constitutes the sig-
`nal that is transmitted across the membraneto guaninenucle-
`otide binding protein-linkedintracellular effectors that are ul-
`timately responsible for glucose production.
`The glucagon receptor is a memberof a unique branchof the
`G protein-coupled receptor superfamily that has highly homol-
`ogous sequences but shares very few of the conserved struc-
`tural features found within the rest of the G protein-coupled
`
`
`* This work wassupported by U. S. Public Health Service Grant DK
`24039. A preliminary report of part of this work was presented at the
`15th American Peptide Symposium, June 14-19, 1997, Nashville, TN.
`The costs of publication of this article were defrayed in part by the
`paymentof page charges.This article must therefore be hereby marked
`“advertisement” in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`+ To whom correspondence should be addressed: The Rockefeller Uni-
`versity, 1230 York Ave., New York, New York 10021. Tel.: 212-327-
`8239; Fax: 212-327-8245.
`
`receptor family (1, 2). Membersof this receptor group include
`receptors for the glucagon family of hormones, glucagon-like
`peptide 1 (GLP-1)(3), secretin (4), gastrin inhibitory peptide
`(5), and vasoactive intestinal peptide (6). The receptor has a
`relatively large extracellular N terminus thought to be in-
`volved in hormone-receptor interaction, followed by hydropho-
`bic helical segments postulated to span the membrane seven
`times and a cytoplasmic C-terminal domain (Fig. 1). Signal
`transduction is thought to proceed upon bindingof the hormone
`with the extracellular region of the receptor. The mechanism
`by which thesignal is conveyed from thecell surface across the
`transmembranehelical network to activate G protein-coupled
`effectors on the surface of cytoplasm is not understood.
`Extensive structure-function studies of glucagon have af-
`forded someinsight into the understandingof its mechanism of
`action. The general picture that has emergedis that the active
`pharmacophoreis dispersed throughout the glucagon molecule
`andthat the intact hormoneis necessary for the expression of
`full hormonal activity. Nevertheless, specific active site resi-
`dues responsible for either high affinity binding or activation
`have beensingled out. Electrostatic interactions of the nega-
`tively charged side chain of aspartic acid 9, 15, and 21 were
`shownto be essential in glucagon function (7, 8). Activity was
`lost when Asp? was deleted or replaced by any other amino
`acid.
`An important early finding from pioneering structure-activ-
`ity studies established that the N-terminal histidine which is
`strictly conserved within the glucagon peptide family was es-
`sential for receptor activation and less so for binding and im-
`plied that the deletion of histidine would produce a glucagon
`antagonist (9). Indeed, the first partial antagonists that were
`developed were des-His' derivatives or glucagon analogs that
`had modified histidines at the N terminus (10-11). Further
`studies demonstrated that the imidazole ring of histidine at
`position one of the hormone furnishes determinants for both
`receptor binding affinity and activity (12).
`Serine residues at positions 2, 8, and 16 were also shown to
`play prominent roles in glucagon action (13). The apparent
`connection of His', Asp®, and Ser?residues led to the hypoth-
`esis that a catalytic triad resembling that of a serine prote-
`ase might be involved in the mechanism of glucagon signal
`transduction (13).
`The glucagon binding cavity on the receptor is mostlikely a
`discontinuous domain that involves contributions from the long
`N-terminal extension as well as from the three extracellular
`loops that connect the seven transmembranehelices (14). In-
`formation about the complementary peptide andprotein inter-
`actions that dictate the binding phenomenonis central to the
`design of antagonists of the hormone that might beclinically
`relevant. To investigate the molecular mechanism of hormone-
`receptor interaction and of receptor activation by site-directed
`
`
`1 The abbreviation used is: GLP-1, glucagon-like peptide 1.
`
`10308
`
`This paperis available online at http://www.jbc.org
`
`This is an Open Accessarticle under the CC BYlicense.
`
`MSN Exhibit 1015 - Page 1 of 5
`MSNv. Bausch - IPR2023-00016
`
`

`

`Glucagon Structure and Function
`
`10309
`
`a M
`
`LLTQLHCPYLYy
`t
`*
`pero T NHHCOOSYLKWKEFLE DMVQAS PAKPLCSLVVL™
`re
`*
`*
`PRTELVCNRT
`FOKYSCWPOTPPNTTANI SCPWYL PWYHKv
`KQVEIEDE
`Qsa
`6K
`OVETEDOIDMQCOSADRWSOGRPGRV WOGD
`Q
`PeckKEVURH
`e
`Vv;
`MG
`vac
`2% GAv
`tre
`Nn w NoOF

`ots
`69
`—E
`T.o%9 w
`Ye \a\NW Jt
`c
`tLe
`F
`Ir
`VW,
`R
`140
`%g
`'g 380
`be
`'
`y
`WY
`V
`&
`7
`
`9 “Sy, 4AKn WALK
`Extracellular
`"4
`FL
`wy
`FP Voy
`pt
`vic Kye L yg
`pw’
`vil
`Dp.
`OfeL
`sy.
`oO ly
`yA Lye
`! a L
`viaF Fs
`ot Sa as
`cYLW Sy
`Nero
`Vevy afs
`uty
`ALK
`tiv
`16,c¢ Foy
`HG
`av
`E
`y L
`! R
`ut p
`Vey
`A
`Vv sf
`var
`ips
`H!
`ult
`tec
`vet
`GAP
`
`_tLS_gN _y. gh. utav st5 oo.
`a
`H

`&
`are K
`eR &
`Intracellular
`rok
`t
`s
`L
`Vv
`"
`N
`Sir
`60
`«OM HY ADY KER
`Q
`cyR
`M
`AgLLR
`0
`45)
`Cc PDGHCT GAPAMHS GHSSAMREEQLAK®
`go SL aL KE
`SsaTGcEPSAKTSLASSL PRLAOSPT485
`
`Y
`
`TF
`
`wp
`w
`
`Fic. 1. Schematic representation of
`the rat glucagon receptor primary
`Pp
`nq
`.
`and secondary structure. Seven puta-
`tive transmembranehelices based on pre-
`vious models of G protein-coupled recep-
`.
`tors are shown. The N terminus and
`extracellular surface is toward the top,
`1
`M
`and the C terminus and cytoplasmic sur-
`face is toward the bottom of the figure.
`Asp™, which was studied by site-specific
`mutagenesis,
`is numbered and labeled
`:
`2
`.
`with an arrow (15). The locations of se-
`quences from the extracellular domain
`that were used as the peptides for anti-
`receptor anti-peptide antibody production
`are boxed.
`
`mutagenesis, we have synthesized a genefor the rat glucagon
`receptor (15, 16).
`Theearliest information to come from site-directed mutagen-
`esis of the glucagon receptor protein identified Asp®™in the
`extracellular N-terminaltail to be absolutely required for the
`recognition function of the receptor (15). Recent studies have
`also implicated areas in the membraneproximalportion of the
`N terminus and thefirst extracellular loop to be part of the
`hormonebinding site (17). More importantly, antibodies raised
`against peptides representing sequences from these regions
`were inhibitors of glucagon bindingto the receptor (18). These
`peptide epitopes containedclusters of negatively charged resi-
`dues, which suggested that an electrostatic association with a
`complementary positively charged residue on the ligand might
`occur at these receptorsites.
`To test this possibility, we assessed the contribution of the
`positively charged groups at positions 12, 17, and 18, to gluca-
`gon receptor recognition, which to date has not been clearly
`established. We synthesized glucagon analogs containing sub-
`stitutions ofLys!?, Arg’, and Arg’® ofglucagon with neutralor
`negatively chargedresiduesto examinetheroles ofthe positive
`charge on those sites on binding and adenylyl cyclase activity.
`It was reported in an earlier study of the C-terminal region of
`the hormone that the analog [Lys'’,Lys'®,Glu?"]glucagon ex-
`hibited enhancedreceptor binding and was a superagonist(19).
`This behavior was attributed to an increased a-helical content
`and the possible formation of an intramolecular salt bridge
`between charged side chains at positions 18 and 21 (20, 21).
`Our results demonstrate that glucagon binding andactivity are
`not dictated solely by electrostatic interactions but include the
`interactions of hydrophobic side chains with the receptor.
`EXPERIMENTAL PROCEDURES
`
`Peptide Synthesis and Purification—Thirty three analogs of glucagon
`with replacements at positions 12, 17, and 18 were assembled by the
`solid-phase method (22, 23), on an Applied Biosystems 430A peptide
`synthesizer, using procedures previously described for the synthesis of
`glucagon analogs (24). Briefly, the peptide analogs with C-terminal
`amides were prepared on p-methylbenzhydrylamine-resin (Peptides In-
`ternational, 0.66 mmol/g) using N*-Boc [tert-butyloxycarbonyl] protec-
`tion chemistry. N%-Boc-protected amino acids were purchased from
`Peptide Institute. Side chain protection was Arg(Tos), Asp(OcHx), Glu-
`(OcHx), His(Tos), Lys[Z(CD], Ser(Bzl), Thr(Bzl), Trp(For), and Tyr-
`[Z(Br)] (where Tosis tosyl; cHx is cyclohexyl; Z(Cl) is 2-chlorobenzyl-
`oxycarbonyl; Bzl is benzyl; and For is formyl). Standard protocol for
`double couplings with preformed symmetric anhydrides in dimethylfor-
`mamide were used routinely, except for arginine, asparagine, and glu-
`
`tamine which were coupled as N1-hydroxybenzotriazole esters (25). The
`N*-formyl group on tryptophan was removed with 50% piperidine in
`dimethylformamide, prior to HF treatment. After cleavage by anhy-
`drous HF, the crude peptides were purified by preparative low pressure
`reverse-phase liquid chromatography on octadecyl-silica (Vydac C18,
`Separations Group). The peptides were eluted by applying a linear
`gradient of 25-40% acetonitrile in 0.05% trifluoroacetic acid. Purity of
`the lyophilized product was evaluated by analytical high pressure liq-
`uid chromatography (Vydac 218TP54) in at least two different solvent
`systems and massspectral analysis by the electrospray methodidenti-
`fied the expected (M + H)* peaks within +0.3 Da. Aminoacid analysis
`yielded amino acid compositions consistent with theory.
`Receptor Binding Assay—Liver plasma membranes were prepared
`from rat liver (Sprague-Dawley, 100-150 g, Charles River) following
`the method of Neville with modifications described by Pohl (26). Mem-
`brane aliquots were stored in liquid nitrogen and used within 4-6
`months. The receptor binding assay was done according to Wright and
`Rodbell (27), in which competition for glucagon receptors in 10 pg of
`liver membraneprotein, between '”°I-labeled glucagon (NEN Life Sci-
`ence Products) (1.6 nM) and the synthetic analogs in concentrations
`ranging from 10711 to 107° M, was measured. Assay suspensions were
`filtered on Durapore membranefilters (0.45 wm) using a vacuum fil-
`tration manifold (Millipore). Binding affinity (percent) is calculated as
`the ratio of the concentration of glucagon that inhibits 50% of the
`binding of tracer (IC5,) to that of peptide analog x 100. Duplicate
`determinations were made for each concentration point, and each assay
`wasrun at least twice. Nonspecific binding, determinedin the presence
`of 10° M unlabeled glucagon, wastypically 10% of total binding.
`Adenylyl Cyclase Assay—Adenylyl cyclase activity was measured
`according to the procedure described by Salomonet al. (28). cAMP
`released was determined with a commercially available assay kit, from
`Amersham Pharmacia Biotech, in which unlabeled cAMP produced
`competes with [8-H]cAMP for a cAMP-binding protein. Data for stim-
`ulation of adenylyl cyclase are expressed as picomoles of cAMP pro-
`duced per mg of membrane protein per min and plotted against the
`logarithm of peptide analog concentration. Maximum activity (percent)
`of an analog is the percentage of maximum stimulation of cAMP pro-
`duction abovebasal by glucagon. Relative potency (percent) is the ratio
`of the concentration of natural glucagon that elicits 50% maximum
`production of cAMP (EC,,) to that concentration of peptide analog x
`100. Inhibition of CAMP production was determined in a similar assay
`in which a constant amount of glucagon is allowed to compete with
`increasing concentrations of analog. The inhibition index (I/A);, is de-
`fined as the ratio of the concentrationsof inhibitor to agonist when the
`response is reduced to 50% of the response to agonist in the absence of
`inhibitor. Analogs were tested for inhibitory properties if they had
`relative potencies of =1% and a binding affinity of =1%. The pA, value,
`calculated by the method of Arunlakshana and Schild (29), is the
`negative logarithm of the concentration of inhibitor that reduces the
`response to 1 unit of agonist to the response produced by 0.5 unit of
`agonist. Duplicate determinations were made for each concentration
`point, and each experiment wascarried outat least twice.
`
`MSN Exhibit 1015 - Page 2 of 5
`MSNv. Bausch - IPR2023-00016
`
`

`

`10310
`
`Glucagon Structure and Function
`TABLE I
`Glucagon analogs with neutral residue replacements at positions 12, 17, and 18
`Adenylyl cyclase activity
`
`
`
`
`
`Bindin,
`Analog of glucagon amide®
`
` ofa od Relate Wags pad
`
`%
`%
`%.
`15
`100
`100
`Glucagon amide
`15.8
`17.3 + 0.2
`59.7
`1. Ala?”
`0.15
`0.91
`12.5
`2. des-His',Ala’™”
`13.8
`11.4
`85.1
`3. Gly?
`0.13
`0.58
`19.4
`4, des-His',Gly'”
`31.6
`47 +1
`90.5
`5. N*-acetyl-Lys’”
`0.013
`38
`29
`6. Ala?”
`0.28
`2.3
`28
`7. des-His*,Ala’”
`37.1 + 1.7
`30+1.8
`88+ 2
`8. Leu?”
`2.13
`9.3
`23
`9. des-His',Leu'”
`70.8
`13
`94.4
`10. Ala'®
`0.14
`3.1
`14
`11. des-His*,Ala?®
`45.7+ 1.6
`56 + 1.5
`95 +3
`12. Leu’®
`1.5
`3.6
`22.5
`13. des-His?,Leu?®
`27.5
`8
`97
`14, Alat”,Ala?®
`0.19
`0.32
`10
`15. des-His',Ala’’,Alal®
`52.5 + 1.2
`7+0.1
`85.4 + 0.8
`16. Leu?’,Leu'®
`7.1
`43.7
`1.15
`1
`17
`17. des-His',Leu!’,Leu'®
`
`18. Ala!?,Ala’”,Ala!® 13 0.08 62.8
`
`
`* Analogs of glucagon amide were assayed using native glucagon as the standard, in both membrane binding and adenylyl cyclase activity.
`[des-His']Glucagon amide hada bindingaffinity of 63% and a relative potency of 0.16% in the adenylyl cyclase assay.
`> Bindingaffinity (%) is the ratio of agonist concentration to analog concentration at 50% receptor occupancy (IC5,) x 100.
`©“ Maximumactivity (%) is the percentage of maximum glucagon stimulation of cAMP production above basal.
`@ Relative potency (%) is the ratio of glucagon concentration to analog concentration at 50% response (EC,,) x 100.
`© The inhibition index (I/A);, is the ratio of peptide inhibitor concentration to glucagon concentration whenthe responseis reduced to 50% of the
`response of agonist in the absence of inhibitor.
`f The pA, valueis the negative logarithm of the concentration of inhibitor that reduces the responseto 1 unit of agonist to the response obtained
`from 0.5 unit of agonist.
`
`43.7
`
`34.7
`
`43.7
`
`85.1
`
`7.0
`
`7.5
`
`7.3
`
`7.0
`
`RESULTS
`
`Thirty three glucagon analogs have been synthesized to as-
`sess the roles of the positively charged basic residues at posi-
`tions 12, 17, and 18 of glucagon, in receptor binding affinity as
`well as in adenylyl cyclase activation. Ourinitial approach was
`to examine the effect of neutralizing the positive charge by
`substituting uncharged aminoacidsat positions 12, 17, and 18
`(Table I). Replacing Lys’? with neutral residues in the analogs
`Ala! and Gly?” glucagon amides (analogs 1 and3) resulted in
`an 80-90% reduction in bindingaffinity relative to glucagon for
`the glucagon receptor in rat liver membranes. However, both
`analogs werestill capable of a full agonist response, with re-
`duced potency. Similarly, acetylation of the «amino group of
`Lys” provided [N‘-acetyl-Lys?]glucagon amide (analog 5),
`which bound with 47% affinity and elicited 90% maximum
`adenylyl cyclase stimulation. These results were consistent
`with an earlier observation that N‘-acylated derivatives of
`glucagon werefull agonists (30) and further implicated a pref-
`erence for a hydrophobic functional group at position 12. An
`exchange of Arg?” for alanine or leucine in analogs 6 and 8
`(Table I) effected a loss of 60-70% bindingaffinity. Ala!’ was a
`weak agonist, whereas Leu?” stimulated adenylyl cyclase with
`an 88% maximum activity. Replacing Arg’® with alanine (an-
`alog 10) led to an 87% loss in binding, whereas a more hydro-
`phobic leucine substitution (analog 12) suffered a smaller loss
`of 44% affinity. Both analogs were capable of a full agonist
`response. Substitution of both sequential arginines with a neutral
`aminoacid in Ala‘”,Ala’® (analog 14) and Leu?’,Leu’® (analog 16)
`resulted in a 90% loss of binding, which in the case of Leu?’,Leu’®
`appeared to be additive. The concurrent loss of both positive
`charges did not influence the ability to activate adenylyl cyclase
`since the doubly substituted analogs were full agonists but with
`lowered potency. In contrast, the exchangeof all three basic resi-
`dues with alanines in [Ala!’,Ala’’,Ala']glucagon amide induced
`almost complete loss ofbinding and resulted in a very weak partial
`agonist. Deletion of Hist from someof the analogs that retained
`
`good bindingaffinities (analogs 6, 8, 10, 12, and 16), produced the
`corresponding des-His? derivatives (analogs 7, 9, 11, 13, and 17)
`that exhibited lowered potencies and measurable antagonist prop-
`erties, which is consistent with the establishedrole of histidine in
`glucagon (9-12). Because these des-His? analogs retained the ca-
`pacity to induce low levels ofcAMP,they were only partial glucagon
`antagonists (31).
`Aside from neutral amino acids, positive residues at posi-
`tions 12, 17, and 18 were each replaced with an aspartic or
`glutamic acid to examine theeffect of a reversal of charge.
`Asp’? and Glu’? (analogs 19 and 20, Table II) displayed poor
`bindingaffinities of 0.6 and 1%, respectively. Likewise, substi-
`tution by aspartic acid at positions 17 or 18 as in [Asp?”]- and
`[Asp?®]glucagon amides (analogs 22 and 26)led to 99% loss of
`binding. Interestingly, glutamic acid was better tolerated at
`these positions, with Glu!” and Glu’® glucagon amides (analogs
`24 and 28) exhibiting a retention of 21 and 6% bindingaffinity,
`respectively, and full stimulation of adenylyl cyclase. Despite
`reduced bindingaffinities a reversal of charge at all positions
`produced glucagon analogs that elicited substantial agonist
`responsesalthough with reduced potencies. Unlike Alat’,Ala?®,
`however, a double replacement with acidic residues in
`[Asp!”,Asp?°]- and in [Glu?”,Glu?®]glucagon amides(analogs 30
`and 32) rendered the peptides incapable of receptor recogni-
`tion. Since it is acknowledged that an intact N-terminalhisti-
`dine provides determinants for both the binding andactivation
`function of glucagon, the des-histidine derivative of every po-
`sition —12, —17, and —18 replacement analog predictably lost
`additional receptor binding affinity and potency of adenylyl
`cyclase activation.
`
`DISCUSSION
`
`There is renewed interest in the peptide glucagon because of
`its role in diabetes mellitus. Despite considerable positive evi-
`dence, the participation of glucagon is still somewhat contro-
`versial and further evidencefor its role is needed. It has been
`
`MSNExhibit 1015 - Page 3 of 5
`MSNv. Bausch - IPR2023-00016
`
`

`

`10311
`
`57.5
`
`6.4
`
`Glucagon Structure and Function
`TABLE II
`
`Glucagon analogs with acidic residue replacements at positions 12, 17, and 18
`
`.
`Adenylyl cyclase activity
`Analog ofglucagon amide®
`Binding
`Maximum
`Relative
`WA)ac?
`Af
`
`activity®
`poten
`50
`PAs
`%
`%
`%
`100
`100
`15
`Glucagon amide
`0.6
`78.4
`10
`19. Asp?
`1
`80.4
`50.1
`20. Glu’?
`0.11
`28
`0.22
`21. des-His',Glu”
`1.4
`82.4
`44
`22. Asp?”
`0.1
`11.5
`0.08
`23. des-His',Asp*”
`21.3 + 0.5
`94.8 + 0.2
`40.7+3
`24. Glu!’
`1.7
`21.5
`1.0
`25. des-His',Glu’”
`0.22
`69.2
`0.24
`26. Asp?®
`<0.038
`27. des-His',Asp1®
`6.2 + 0.2
`28. Glu!
`0.44
`29. des-His',Glu’®
`<0.032
`30. Asp?’,Asp?®
`<0.032
`31. des-His*,Asp?”,Asp?®
`1.2
`100
`0.036
`32. Glu?”,Glu’®
`33. des-His’,Glu’’,Glu'® <0.050
`
`* Analogs of glucagon amide were assayed using native glucagon as the standard, in both membrane binding and adenylyl cyclase activity.
`[des-His']Glucagon amide hada binding affinity of 63% and a relative potency of 0.16% in the adenylyl cyclase assay.
`> Bindingaffinity (%) is the ratio of agonist concentration to analog concentration at 50% receptor occupancy (IC;,) x 100.
`° Maximum activity (%) is the percentage of maximum glucagon stimulation of cAMP production above basal.
`4 Relative potency (%) is the ratio of glucagon concentration to analog concentration at 50% response (EC,,) < 100.
`© The inhibition index (I/A);, is the ratio of peptide inhibitor concentration to glucagon concentration when the responseis reduced to 50% of the
`response of agonist in the absenceof inhibitor.
`f The pA,valueis the negative logarithm of the concentration of inhibitor that reduces the responseto 1 unit of agonist to the response obtained
`from 0.5 unit of agonist.
`
`100 + 2
`18
`
`3.3 + 0.3
`0.24
`
`observed that overproduction of glucose by elevated circulating
`levels of glucagon may be a contributing factor to hyperglyce-
`mia and ketoacidosis that is characteristic of the disease (32,
`33). It was reasonable to assume that antagonists of the hor-
`mone that are able to inhibit the actions of this endogenous
`glucagon by competing for the same binding cavity in the
`glucagon receptor could haveclinical potential in the manage-
`ment of diabetic complications (9, 32, 33). Indeed, several pep-
`tide analogs have been developed that have been shown to
`effectively inhibit the effects of glucagon both in vitro and in
`vivo (24, 31, 34-38). Continuedefforts in the study of glucagon
`are spurred by the idea that the ability to single out specific
`contact points between the peptide ligand and its receptor
`protein would serve as a basisfor the rational design of analogs
`that bind yet do not activate adenylyl cyclase.
`This study of the electrostatic interaction of the basic resi-
`dues Lys”, Arg”, and Arg?® of glucagon with acidic residues of
`the glucagon receptor is based on the supposition that any one
`or all of these groups may provide a counterion to a specific
`aspartic acid residue on the extracellular domainof the recep-
`tor that has been shown in recent mutagenesis studies to be
`critical for ligand recognition (15, 18). The importanceofhisti-
`dine at position 1 to both receptor binding andactivation has
`been firmly established (9-12). Removal of the histidine group
`afforded an analog that retained affinity for the receptor but,
`more importantly, appeared to partially inhibit glucagon-stim-
`ulated adenylyl cyclase (9). Although the N-terminalhistidine
`is strictly conserved within the glucagon family of peptide
`hormones, Lys!”, Arg!’, and Arg!® are relatively unique at
`these positions in glucagon and mightalso serve as determi-
`nants of receptor specificity.
`The results of our study reveal that while neutral residue
`scanning of positions 12, 17, or 18 of glucagon strongly atten-
`uated receptor binding, most of the resulting analogs were
`weak but full agonists, suggesting that a positive charge at
`these particular positions was not absolutely critical for activ-
`ity. That no positively charged aminoacids of glucagon, with
`the exception of histidine 1, are critical for the activation func-
`tion is indicated by the observation that none of the replace-
`
`glucagon |H S|Q|G TFT SD/Y/S KIY LIDJS R AJAQ DIF VQIWIL MIN T -
`
`
`
`|H[AJEG TF TSDIV S[SY L GQ AJA|K E/F I[AWILV|IKGR
`gip!
`
`secretin TSO GET SEL SAG SARILQ RIL LQIG/L VI- - -
`vip [HS DJA VLE TIDN Y TIRJLR KQM ALVJK KLY_ LNJSLI LIN - -
`
`Consensus HSDGTFTSD-SR--D---A--FLQ-LV---
`
`
`
`Fic. 2. Sequence alignmentof peptides of the glucagon family
`that have close homology. Residues that are conserved in either
`charge or hydrophobic character are boxed.
`
`ment analogs in Table I behaved as antagonists. However,
`someof the corresponding des-His! derivatives of positions 17
`and 18 replacement analogs displayed partial antagonist prop-
`erties, which was therefore associated with the deletion of
`position 1 histidine (31). These were in the (I/A);, range of
`34-85, whereas the most potent glucagon antagonist reported
`to date was 0.85 (36).
`A negatively charged acidic amino acid was, however, less
`tolerated at these positions and impaired receptor binding by a
`hundred-fold or more. Strongly reduced bindingaffinities were
`coupled with adenylyl cyclase responses with much weakened
`potencies. Thus, a positive charge at these positions is neces-
`sary for optimal hormonal function.
`Our previous findings established the roles of the aspartic
`acid residues at positions 9, 15, and 21 of glucagon. Asp? is
`critical for transduction but not for binding (7), whereas the
`negative charge at Asp?®is absolutely essential for binding(8).
`In contrast, the positively charged residues in the central re-
`gion of the hormonehave a specific role in achieving optimal, or
`even significant, binding and maximalbiological potency. An
`alteration in the charge distribution along the molecule clearly
`results in decreased binding of the derivatives. Unlike posi-
`tions 9 and 15 wheretheprecise location of an aspartic acid
`residue is critical to hormone-receptor interaction, it appears
`that glucagon binding affinity is not regulated by the topo-
`graphic location of a specific positive charge but by a net pos-
`itive charge. An overall positively charged molecule definitely
`enhancestheaffinity for its membrane-boundreceptorprotein.
`A single replacement in glucagon amide with an uncharged
`residue did not adequately alter the overall charge to reduce
`MSNExhibit 1015 - Page 4 of 5
`MSNv. Bausch - IPR2023-00016
`
`

`

`10312
`
`Glucagon Structure and Function
`
`binding andactivity, but one substitution with a negatively
`charged residue resulted in a neutral molecule.
`Secretin, GLP-1, and vasoactive intestinal peptide, gluca-
`gon’s closest relatives within the family of peptide hormones,
`share 50% sequence homology mostly at the N-terminal half of
`the molecule (Fig. 2). With the exception of His", the positions
`of basic residues scattered primarily along the C-terminal part
`of these sequences are not well conserved. In an earlier report,
`a glucagon-GLP-1 chimeric peptide, in which thefirst 14 resi-
`dues of glucagon were combined with the last 16 residues of
`GLP-1, bound to both glucagon and GLP-1 receptors (39). The
`normal peptides only bind weakly to each other’s receptor.
`Interestingly, what appears to be preserved is an overall posi-
`tive charge, suggesting that a positively charged ligand may be
`one requisite feature common to membersof this G protein-
`coupled receptor sub-group andtherefore not a strict determi-
`nant of specificity.
`The study also reveals a hydrophobicity component of the
`bindinginteraction. The basic amino acids arginine andlysine
`can contribute not only a charged group but also an aliphatic
`component to the polar and non-polar interface of the ligand
`binding pocket. The proposed ligand binding site should lie
`within a hydrophobic core of the receptor where nonspecific
`hydrophobic interactions between the hormoneandits receptor
`embedded in the membranebilayer augment bindingaffinity.
`This explains whysubstitution with a neutral yet hydrophobic
`molecule like alanine or leucine was well tolerated despite the
`loss of a positive charge and could sustain 30-60% of the
`affinity for the receptor. A reversal of charge on the other hand
`adversely altered the polar character of the peptide and led to
`a greater loss of receptor recognition (Table II). Nevertheless,
`an increased hydrophobic contribution from a glutamic acid
`side chain probably compensatedfor the reversal of charge and
`accounted for the retention of substantial binding affinity and
`potency of the Glu?” and Glu’® replacement analogs (analogs
`24 and 28, Table II). In addition, des-His',Glu’” (analog 25,
`Table II), which retained 2% binding and a weakened potency
`due to deletion of histidine, wasstill a partial antagonist. The
`analog [Lys?’,Lys?®,Glu”"]glucagon has been reported to be a
`superagonist which bound 5-fold better than the natural hor-
`mone and had a higherpotency(19, 20). A recent x-ray crystal
`structure for [Lys!’,Lys?®,Glu”|glucagon suggested that the
`formation of a salt bridge between the e-amino group of Lys'®
`and the carboxyl of Glu?! maystabilize the turn of a putative
`a-helix at residues 18—21 and contributes to its superagonist
`activity (21). However, bindingaffinity remains relatively high
`even when hydrophobic residues are substituted for either
`Arg?” or Arg!®. Presumably, enhanced activity may also be
`attributed to the increased hydrophobicity of the longer ali-
`phatic side chains of lysine and glutamic acid relative to those
`of the normal arginine and aspartic acid residues.
`Thus, the positively charged amino acids Lys??, Arg!”, and
`Arg?® of glucagon have large effects but are not absolutely
`critical for the binding and activation function of the hormone.
`However,the functional groups of these basic residues bolster
`both the polar and non-polar aspects of the peptide and protein
`interactions that occur within the receptor binding site and
`ensure maximumbiological activity. The aliphatic backbone of
`arginine andlysine residues optimize ligand “fitting” within a
`hydrophobic pocket in the receptor. Our data demonstrate that
`at these positions in glucagon, nonspecific hydrophobic inter-
`
`actions are as importanta contributing factor to bindingaffin-
`ity as the electrostatic effects. Mutagenesis studies on the
`receptor have outlined the perimeter of a putative bindingsite
`bordered by negatively charged residues. Thus, an overall pos-
`itively charged glucagon molecule contributes to the stabiliza-
`tion of the hormone-receptor complex and secures the binding
`conformation that subsequently leads to activation.
`
`REFERENCES
`1. Jelinek, L. J., Lok, S., Rosenberg, G. B., Smith, R. A., Grant, F. J., Biggs, S.,
`Bensch,P. A., Kuifper, J. L., Sheppard, P. O., Sprecher, C. A., O’Hara,P. J.,
`Foster, D., Walker, K. M., Chen, L. H. J., Mckernan, P. A. & Kindsvogel, W.
`(1993) Science 259, 1614-1616
`2. Segre, G. V. & Goldring, S. R. (1993) Trends Endocrinol. Metab. 4, 309-314
`3. Thorens, B. M. (1992) Proc. Natl. Acad. Sci. U. S. A. 89, 8641-8646
`4. Ishihara, T., Nakamura, S., Kaziro, Y., Takahashi, T. Takahashi, K. & Nagata,
`S. (1991) EMBOJ. 10, 1635-1641
`5. Usdin, T. B., Mesey, E., Button, D. C., Brownstein, M. J. & Bonner, T. J. (1993)
`Endocrinology 133, 2861-2870
`6. Ishihara, T., Shigemoto, R., Mori, K. & Nagata, S. (1992) Neuron 8, 811-819
`7. Unson, C. G., Macdonald, D., Ray, K., Durrah, T. L. & Merrifield, R. B. (1991)
`J. Biol. Chem. 266, 2763-2766
`8. Unson, C. G., Wu, C.-R. & Merrifield, R. B. (1994) Biochemistry 33, 6884-6887
`9. Lin, M. C., Wright, D. E., Hruby, V. J. & Rodbell, M. R. (1975) Biochemistry 14,
`1559-1563
`10. Hruby, V. J. (1982) Mol. Cell. Biochem. 44, 49-64
`11. Hruby, V. J., Krstenansky,J. L., McKee, R. & Pelton, J. T. (1986) in Hormonal
`Control of Gluconeogenesis (Kraus-Friedman, N., ed) Vol. 2, pp. 3-20, CRC
`Press, Inc., Boca Raton, FL
`12. Unson, C. G., Macdonald, D. & Merrifield, R. B. (1993) Arch. Biochem. Bio-
`phys. 300, 747-750
`13. Unson, C. G. & Merrifield, R. B. (1994) Proc. Natl. Acad. Sci. U. S.A. 91,
`454-458
`14. Unson, C. G., Cypess, A. M., Kim,

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