throbber
Proc. Natl. Acad. Sci. USA
`Proc. Natl. Acad. Sci. USA
`Vol. 89, pp. 947-951, February 1992
`Vol. 89, pp. 947-951, February 1992
`Pharmacology
`Pharmacology
`
`Guanylin: An endogenous activator of intestinal guanylate cyclase
`Guanylin: An endogenous activator of intestinal guanylate cyclase
`(intestine/cyclic GMP/heat-stable enterotoxin/diarrhea/peptide)
`(intestine/cyclic GMP/heat-stable enterotoxin/diarrhea/peptide)
`MARK G. CURRIE*t, KAM F. FOKt, JowI KATO*, ROSALYN J. MOORE*, FRANKLIN K. HAMRA*,
`MARK G. CURRIE*t, KAM F. FOO, JOHJI KATO*, ROSALYN J. MOORE*, FRANKLIN K. HAMRA*,
`KEVIN L. DUFFIN§, AND CHRISTINE E. SMITHS
`KEVIN L. DUFFIN§, AND CHRISTINE E. SMITH¶
`Departments of *Molecular Pharmacology, tBiological Chemistry, §Physical Sciences, and lProtein Biochemistry, Monsanto Corporate Research, Monsanto
`Departments of *Molecular Pharmacology, *Biological Chemistry, *Physical Sciences, and 1Protein Biochemistry, Monsanto Corporate Research, Monsanto
`Company, St. Louis, MO 63167
`Company, St. Louis, MO 63167
`
`Communicated by Philip Needleman, October 11, 1991
`Communicated by Philip Needleman, October 11, 1991
`
`Intestinal guanylate cyclase mediates the ac-
`ABSTRACT
`Intestinal guanylate cyclase mediates the ac-
`ABSTRACT
`tion of the heat-stable enterotoxin to cause a decrease in
`tion of the heat-stable enterotoxin to cause a decrease in
`intestinal fluid absorption and to increase chloride secretion,
`intestinal fluid absorption and to increase chloride secretion,
`ultimately causing diarrhea. An endogenous ligand that acts on
`ultimately causing diarrhea. An endogenous ligand that acts on
`this guanylate cyclase has not previously been found. To search
`this guanylate cyclase has not previously been found. To search
`for a potential endogenous ligand, we utilized T84 cells, a
`for a potential endogenous ligand, we utilized T84 cells, a
`human colon carcinoma-derived cell line, in culture as a
`human colon carcinoma-derived cell line, in culture as a
`bioassay. This cell line selectively responds to the toxin in a very
`bioassay. This cell line selectively responds to the toxin in a very
`sensitive manner with an increase in intracellular cyclic GMP.
`sensitive manner with an increase in intracellular cyclic GMP.
`In the present study, we describe the purification and structure
`In the present study, we describe the purification and structure
`of a peptide from rat jejunum that activates this enzyme. This
`of a peptide from rat jejunum that activates this enzyme. This
`peptide, which we have termed guanylin, is composed of 15
`peptide, which we have termed guanylin, is composed of 15
`amino acids and has the following amino acid sequence,
`amino acids and has the following amino acid sequence,
`PNTCEICAYAACTGC, as determined by automated Edman
`PNTCEICAYAACTGC, as determined by automated Edman
`degradation sequence analysis and electrospray mass spec-
`degradation sequence analysis and electrospray mass spec-
`trometry. Analysis of the amino acid sequence of this peptide
`trometry. Analysis of the amino acid sequence of this peptide
`reveals a high degree of homology with heat-stable enterotox-
`reveals a high degree of homology with heat-stable enterotox-
`ins. Solid-phase synthesis of this peptide confirmed that it
`ins. Solid-phase synthesis of this peptide confirmed that it
`stimulates increases in T84 cyclic GMP levels. Guanylin re-
`stimulates increases in T84 cyclic GMP levels. Guanylin re-
`quired oxidation for expression of bioactivity and subsequent
`quired oxidation for expression of bioactivity and subsequent
`reduction of the oxidized peptide eliminated the effect on cyclic
`reduction of the oxidized peptide eliminated the effect on cyclic
`GMP, indicating a requirement for cysteine disulfide bond
`GMP, indicating a requirement for cysteine disulfide bond
`formation. Synthetic guanylin also displaces heat-stable enter-
`formation. Synthetic guanylin also displaces heat-stable enter-
`otoxin binding to cultured T84 cells. Based on these data, we
`otoxin binding to cultured T84 cells. Based on these data, we
`propose that guanylin is an activator of intestinal guanylate
`propose that guanylin is an activator of intestinal guanylate
`cyclase and that it stimulates this enzyme through the same
`cyclase and that it stimulates this enzyme through the same
`receptor binding region as the heat-stable enterotoxins.
`receptor binding region as the heat-stable enterotoxins.
`
`Guanylate cyclase is comprised of a group of proteins that
`Guanylate cyclase is comprised of a group of proteins that
`share a common enzymatic function ofproducing cyclic GMP
`share a common enzymatic function of producing cyclic GMP
`but differ quite remarkably in their selectivity toward acti-
`but differ quite remarkably in their selectivity toward acti-
`vation by ligands. The three.major types of guanylate cyclase
`vation by ligands. The three major types of guanylate cyclase
`are the soluble, particulate, and intestinal [cytoskeletal-
`are the soluble, particulate, and intestinal [cytoskeletal-
`associated particulate or heat-stable enterotoxin (STa)-
`associated particulate or heat-stable enterotoxin (STa)-
`sensitive] forms and each is regulated by different ligands (1,
`sensitive] forms and each is regulated by different ligands (1,
`2). Activation of the soluble guanylate cyclase occurs in
`2). Activation of the soluble guanylate cyclase occurs in
`response to nitric oxide, whereas activation of the particulate
`response to nitric oxide, whereas activation of the particulate
`enzyme occurs in response to the natriuretic peptides (atrial
`enzyme occurs in response to the natriuretic peptides (atrial
`natriuretic peptide, brain natriuretic peptide, and C-type
`natriuretic peptide, brain natriuretic peptide, and C-type
`natriuretic peptide) (1, 2). An endogenous activator of the
`natriuretic peptide) (1, 2). An endogenous activator of the
`intestinal guanylate cyclase has not previously been identi-
`intestinal guanylate cyclase has not previously been identi-
`fied. However, the STa from Escherichia coli is known to
`fied. However, the STa from Escherichia coli is known to
`selectively activate this form of the enzyme (3, 4). The
`selectively activate this form of the enzyme (3, 4). The
`intestinal form is predominantly found in the intestinal epi-
`intestinal form is predominantly found in the intestinal epi-
`thelial cells with the largest number of receptors oriented
`thelial cells with the largest number of receptors oriented
`toward the lumen (1, 2). Recently, it has been cloned and
`toward the lumen (1, 2). Recently, it has been cloned and
`expressed from rat small intestinal mucosa (5). This enzyme
`expressed from rat small intestinal mucosa (5). This enzyme
`is characterized by an extracellular receptor binding region,
`is characterized by an extracellular receptor binding region,
`a transmembrane region, an intracellular protein kinase-like
`a transmembrane region, an intracellular protein kinase-like
`region, and a cyclase catalytic domain (5).
`region, and a cyclase catalytic domain (5).
`
`The publication costs of this article were defrayed in part by page charge
`The publication costs of this article were defrayed in part by page charge
`payment. This article must therefore be hereby marked "advertisement"
`payment. This article must therefore be hereby marked "advertisement"
`in accordance with 18 U.S.C. §1734 solely to indicate this fact.
`in accordance with 18 U.S.C. §1734 solely to indicate this fact.
`
`Pathogenic strains of E. coli and other bacteria produce a
`Pathogenic strains of E. coli and other bacteria produce a
`family of heat-stable enterotoxins (STs) that activate intes-
`family of heat-stable enterotoxins (STs) that activate intes-
`tinal guanylate cyclase. STs are acidic peptides that contain
`tinal guanylate cyclase. STs are acidic peptides that contain
`18 or 19 amino acids with six cysteines and three disulfide
`18 or 19 amino acids with six cysteines and three disulfide
`bridges that are required for full expression of bioactivity (6).
`bridges that are required for full expression of bioactivity (6).
`The increase of intestinal epithelial cyclic GMP elicited by
`The increase of intestinal epithelial cyclic GMP elicited by
`STs is thought to cause a decrease in water and sodium
`STs is thought to cause a decrease in water and sodium
`absorption and an increase in chloride secretion (7, 8). These
`absorption and an increase in chloride secretion (7, 8). These
`changes in intestinal fluid and electrolyte transport then act
`changes in intestinal fluid and electrolyte transport then act
`to cause secretory diarrhea. In developing countries, the
`to cause secretory diarrhea. In developing countries, the
`diarrhea resulting from STs causes many deaths, particularly
`diarrhea resulting from STs causes many deaths, particularly
`in the infant population (9). STs are also considered a major
`in the infant population (9). STs are also considered a major
`cause of traveler's diarrhea in developed countries (10). They
`cause of traveler's diarrhea in developed countries (10). They
`have also been reported to be a leading cause of morbidity
`have also been reported to be a leading cause of morbidity
`and death in domestic animals (11).
`and death in domestic animals (11).
`In the present study, we designed a bioassay to search for
`In the present study, we designed a bioassay to search for
`a potential endogenous ligand that activates the intestinal
`a potential endogenous ligand that activates the intestinal
`guanylate cyclase. This bioassay is based on the demonstra-
`guanylate cyclase. This bioassay is based on the demonstra-
`tion that T84 cells in culture respond to ST in a selective and
`tion that T84 cells in culture respond to ST in a selective and
`sensitive manner with graded increases of intracellular cyclic
`sensitive manner with graded increases of intracellular cyclic
`GMP. This bioassay revealed that the intestine as well as the
`GMP. This bioassay revealed that the intestine as well as the
`kidney possessed an active material. Purification of this
`kidney possessed an active material. Purification of this
`material from the rat intestine was accomplished and the
`material from the rat intestine was accomplished and the
`structure was determined to be a 15-amino acid peptide with
`structure was determined to be a 15-amino acid peptide with
`4 cysteines that must be disulfide-linked for bioactivity. The
`4 cysteines that must be disulfide-linked for bioactivity. The
`peptide, termed guanylin, also possesses a high degree of
`peptide, termed guanylin, also possesses a high degree of
`homology with STs.
`homology with STs.
`
`MATERIALS AND METHODS
`MATERIALS AND METHODS
`Cell Culture. A cultured human colon carcinoma cell line
`Cell Culture. A cultured human colon carcinoma cell line
`(T84) was obtained from the American Type Culture Collec-
`(T84) was obtained from the American Type Culture Collec-
`tion at passage 52. Cells were grown to confluency in 24-well
`tion at passage 52. Cells were grown to confluency in 24-well
`culture plates with a 1:1 mixture of Ham's F12 medium and
`culture plates with a 1:1 mixture of Ham's F12 medium and
`Dulbecco's modified Eagle's medium (DMEM) supple-
`Dulbecco's modified Eagle's medium (DMEM) supple-
`mented with 10% fetal calf serum, 100 international units of
`mented with 10% fetal calf serum, 100 international units of
`penicillin per ml, and 100 ,g of streptomycin per ml. Cells
`penicillin per ml, and 100 µg of streptomycin per ml. Cells
`were used at passages 54-60.
`were used at passages 54-60.
`Cyclic GMP Determination. Monolayers of T84 cells in
`Cyclic GMP Determination. Monolayers of T84 cells in
`24-well plates were washed twice with 1 ml of DMEM per ml
`24-well plates were washed twice with 1 ml of DMEM per ml
`and then incubated at 37°C for 10 min with 0.5 ml of DMEM
`and then incubated at 37°C for 10 min with 0.5 ml of DMEM
`containing 1 mM isobutylmethylxanthine (IBMX), a cyclic
`containing 1 mM isobutylmethylxanthine (IBMX), a cyclic
`nucleotide phosphodiesterase inhibitor. Agents and fractions
`nucleotide phosphodiesterase inhibitor. Agents and fractions
`were then added for the indicated time as described in
`were then added for the indicated time as described in
`Results. The media was then aspirated and the reaction was
`Results. The media was then aspirated and the reaction was
`terminated by the addition of ice-cold 0.5 ml of 0.1 M HCL.
`terminated by the addition of ice-cold 0.5 ml of 0.1 M HCI.
`Aliquots were then evaporated to dryness under nitrogen and
`Aliquots were then evaporated to dryness under nitrogen and
`resuspended in 5 mM sodium acetate buffer (pH 6.4). The
`resuspended in 5 mM sodium acetate buffer (pH 6.4). The
`
`Abbreviations: IBMX, isobutylmethylxanthine; TFA, trifluoroacetic
`Abbreviations: IBMX, isobutylmethylxanthine; TFA, trifluoroacetic
`acid; C18, octadecasilyl; STa, heat-stable enterotoxin; ST, heat-
`acid; C18, octadecasilyl; STa, heat-stable enterotoxin; ST, heat-
`stable enterotoxin; DPI, dithiothreitol; PTH, phenylthiohydantoin.
`stable enterotoxin; DTT, dithiothreitol; PTH, phenylthiohydantoin.
`tTo whom reprint requests should be addressed at: Monsanto
`tTo whom reprint requests should be addressed at: Monsanto
`Company, Mail Zone T3P, 800 North Lindbergh Boulevard, St.
`Company, Mail Zone T3P, 800 North Lindbergh Boulevard, St.
`Louis, MO 63167.
`Louis, MO 63167.
`
`947
`947
`
`MYLAN EXHIBIT - 1031
`Mylan Pharmaceuticals, Inc. v. Bausch Health Ireland, Ltd. - IPR2022-00722
`
`

`

`948
`
`Pharmacology: Currie et al.
`
`Proc. Natl. Acad. Sci. USA 89 (1992)
`
`samples were subsequently measured for cyclic GMP by RIA
`as described by Steiner et al. (12).
`Purification of Guanylin. Rat jejunums flushed of luminal
`contents with 50 ml of saline and immediately placed on dry
`ice were obtained from Bioproducts for Science (Indianap-
`olis). The jejunums were thawed, minced, and boiled for 10
`min in 1 M acetic acid. The extract was centrifuged at 20,000
`x g for 20 min at 4°C. The resulting supernatant was filtered
`and applied to an octadecasilyl (C18) Sep-Pak (Waters). The
`column was washed with 10% acetonitrile/0.1% trifluoroace-
`tic acid (TFA)/H2O and eluted with 60% acetonitrile/0.1%
`TFA/H2O. The eluted peptide fraction was lyophilized and
`resuspended in 50 ml of distilled H2O containing 0.8% am-
`pholytes, pH range 3-10, and applied to a preparative iso-
`electric focusing cell (Rotofor, Bio-Rad). The sample was
`focused for 150 min at 12 W constant power. The fractions
`were harvested, pH determined, and bioassayed. The active
`fractions, which focused around pH 3.8, were then refocused
`under similar conditions and the resulting active fractions
`were lyophilized. The sample was then resuspended in 1 ml
`of 10% acetonitrile/0.1% TFA/H2O, applied to a C18 semi-
`preparative HPLC column (Vydac, Hesperia, CA), and
`eluted at a flow rate of 3 ml/min. The following gradient was
`used to fractionate the sample: 10% acetonitrile, 0.1% TFA
`to 30% acetonitrile, 0.1% TFA in 180 min. The active fraction
`was then determined by bioassay and lyophilized. This
`sample was resuspended in 1 ml of 10% acetonitrile/0.1%
`TFA/H2O and applied to a phenyl analytical HPLC column
`(Vydac, Hesperia, CA). The conditions for elution were
`similar to that described above for the semipreparative col-
`umn except the flow was 1 ml/min. The active fraction was
`lyophilized and then resuspended in 1 ml of 10% acetonitrile/
`0.1% TFA/H2O. The sample was then applied to a C18
`analytical HPLC column (Vydac) and eluted according to the
`above description for the phenyl column. The active fraction
`was identified by bioassay and lyophilized. The sample was
`reconstituted in 1 ml of 10% acetonitrile/0.1% TFA/H2O,
`reapplied to the analytical C18 column, and eluted by a
`gradient of 10% acetonitrile/10 mM ammonium acetate/H2O,
`pH 6.2, to 30% acetonitrile/10 mM ammonium acetate/H2O,
`pH 6.2, in 180 min. The active fraction was lyophilized and
`reconstituted in 0.05 ml of 0.1% TFA/H2O. The sample was
`then applied to a C8 microbore column and eluted by an
`increasing gradient of 0.33%/min of acetonitrile/0.1% TFA/
`H2O. Two separate batches of purified peptide were then
`subjected to sequence analysis.
`N-Terminal Protein Sequence Analysis. Automated Edman
`degradation chemistry was used to determine the NH2-
`terminal protein sequence. An Applied Biosystems model
`470A gas-phase sequencer was employed for the degrada-
`tions (13) using the standard sequencer cycle 03RPTH. The
`respective phenylthiohydantoin (PTH) amino acid deriva-
`tives were identified by reverse-phase HPLC analysis in an
`on-line fashion employing an Applied Biosystems model
`120A PTH analyzer fitted with a Brownlee 2.1-mm i.d. PTH
`C18 column. On-sequencer pyridylethylation was performed
`as outlined by Kruft et al. (14). The PTH derivative of
`pyridylethylcysteine was identified by HPLC as eluting
`slightly prior to the PTH derivative of methionine.
`Electrospray Mass Spectrometry. Individual samples of
`native and synthetic guanylin were purified by microbore C8
`reverse-phase HPLC (Brownlee Aquapore RP-300 7-µm col-
`umn, P. J. Cobert, St. Louis, MO) and eluting fractions of the
`pmol/µ1 for
`peptides were collected and concentrated to
`mass analysis. Sample solutions were introduced to the mass
`spectrometer via injection into a stream of acetonitrile/H2O/
`TFA, 1000:1000:1, vol/vol/vol, which continuously flowed
`to the mass spectrometer at a flow of 10 µ1/min. Three
`microliters of each of the concentrated guanylin samples was
`injected to obtain the results that are presented in this paper.
`
`A Sciex API III triple-quadrupole mass spectrometer
`(Thornhill, Ontario, Canada) equipped with an atmospheric
`pressure ion source was used to sample positive ions pro-
`duced from an electrospray interface (15). Mass analysis of
`sample ions was accomplished by scanning the first quadru-
`pole in 1 atomic mass unit increments from 1000 to 2400
`atomic mass units in
`s and passing mass-selected ions
`through the second and third quadrupoles operated in the
`rf-only mode to the multiplier. For maximum sensitivity, the
`mass resolution of the quadrupole mass analyzer was set so
`that ion signals were "---2 atomic mass units wide at half peak
`height, but the centroid of the ion signal still represented the
`correct mass of the ion. Comparison of the oxidized and
`reduced guanylin molecular ion region was made by scanning
`the quadrupole mass analyzer in 0.1 atomic mass unit steps
`from 1510 to 1525 atomic mass units in 2 s. Mass spectra of
`the guanylin samples were averaged over all of the scans that
`were acquired during elution of the 3-µI sample solution.
`Binding Assay. 125I-labeled STa (125I-STa) was prepared by
`the Iodo-Gen method (16). T84 cell monolayers were washed
`twice with 1 ml of DMEM and then incubated for 30 min at
`37°C in 0.5 ml of DMEM with 125I-STa (amino acids 5-18)
`(100,000 cpm per well) and either guanylin or 100 nM STa.
`The cells were then washed four times with 1 ml of DMEM
`and solubilized with 0.5 ml of 1 M NaOH per well. This
`volume was transferred to tubes and assayed for radioactivity
`by a y counter. Results are expressed as the percentage
`specifically bound.
`Chemical Synthesis of Guanylin. Guanylin was synthesized
`by the solid-phase method (17) with an Applied Biosystems
`430A peptide synthesizer on Cys(4-CH3Bzl)-OCH2-phenyl-
`acetamidomethyl resin using double coupling cycles to en-
`sure complete coupling at each step. Coupling was effected
`with preformed symmetrical anhydride of tert-butoxycar-
`bonyl amino acids (Applied Biosystems), and peptides were
`cleaved from the solid support in hydrogen fluoride/
`dimethylsulfide/anisole/p-thiocresol, 8:1:1:0.5, vol/vol/
`vol/wt, at 0°C for 60 min. Peptides were cyclized using
`dimethylsulfoxide as described by Tam et al. (18). Peptides
`were purified by successive reverse-phase chromatography
`on a 45 x 300 mm Vydac C18 column and on a 19 x 150 mm
`µBondapak C18 column, using a gradient of 10-30% aceto-
`nitrile in 0.5% TFA. The structures and purity of the syn-
`thetic peptides were verified by fast atom bombardment mass
`spectrometry, amino acid analysis, and gas-phase sequence
`analysis.
`
`RESULTS
`Initial characterization of the T84 cell response indicated that
`these cells were very sensitive to STa with a concentration of
`10-1° M eliciting a 4-fold increase in cyclic GMP. The cells
`also displayed a remarkable range, with a maximal response
`of STa (10-7 M) eliciting a >1000-fold increase in cyclic
`GMP. Furthermore, we failed to detect an effect of either
`sodium nitroprusside (10-3 M) or atrial natriuretic peptide
`(10-6 M) on cyclic GMP levels, suggesting that the T84 serves
`as a selective bioassay for agents that activate the intestinal
`guanylate cyclase. Various rat tissues were surveyed as
`sources for T84 cell cyclic GMP agonist activity, and jejunum
`and kidney were found to possess activity while liver, brain,
`pancreas, spleen, lung, and testes lacked detectable activity
`(Fig. 1). We also observed that rat embryonic intestine
`possessed a similar activity. Treatment of the T84 cells with
`10% of embryonic intestinal extract increased the cyclic GMP
`from a basal level of 120 ± 10 fmol per well to 270 ± 10 fmol
`per well (mean ± SE).
`Purification of the adult rat jejunal bioactivity was accom-
`plished by the processing scheme described in Materials and
`Methods. Briefly, following acid boiling and extraction by a
`
`

`

`Pharmacology: Currie et al.
`
`Proc. Natl. Acad. Sci. USA 89 (1992)
`
`949
`
`3000
`
`a)
`
`2000
`
`0
`E
`2a_ 1000
`
`Gds
`c)./
`
`•d
`/?
`
`<5,-
`i ro
`o
`Pd S
`
`47 4'
`
`"12
`
`FIG. 1. Effect of extracts from various tissues on TM cyclic GMP
`levels. Acid extracts were prepared from 1 g of tissue and 10% of each
`extract was applied to IBMX-treated cells. Values are means ± SE
`(n = 3).
`
`C18 reverse-phase matrix, the material was fractionated on a
`preparative isoelectric focusing cell, which resulted in a
`200-fold purification and indicated that the isoelectric point
`was about 3.8. Refocusing of the active fraction resulted in a
`further 5- to 10-fold purification. The active fraction was then
`purified to homogeneity by a series of reverse-phase HPLC
`steps, including a semipreparative C18 column, a phenyl
`column, two separations on a C18 column utilizing different
`ion-pairing reagents, and final purification on a microbore C8
`column (Fig. 2).
`Preliminary experiments suggested that the material was a
`low molecular weight peptide; therefore the material was
`subjected to N-terminal protein sequence analysis and to
`electrospray mass spectrometric analysis. The combination
`of the data derived from these two techniques yielded the
`complete sequence for guanylin: Pro-Asn-Thr-Cys-Glu-Ile-
`Cys-Ala-Tyr-Ala-Ala-Cys-Thr-Gly-Cys. The N-terminal se-
`quence through 14 places was determined by two indepen-
`dent gas-phase sequencing experiments. The C-terminal
`amino acid was deduced from data obtained by electrospray
`mass spectroscopy. The initial results yielded a sequence in
`which no PTH amino acid derivative was observed at posi-
`tions 4, 7, and 12. Since cysteine residues cannot be posi-
`tively identified during gas-phase sequencing without reduc-
`tion and alkylation, the lack of a PTH amino acid derivative
`at these positions suggested the presence of cysteine resi-
`dues. For complete verification, the putative cysteine resi-
`dues of guanylin were pyridylethylated and the peptide was
`resequenced. The subsequent N-terminal gas-phase se-
`quence analysis verified cysteine residues at positions 4, 7,
`and 12. Further primary structure information was obtained
`by electrospray mass spectrometry. The electrospray mass
`spectrum of native guanylin (Fig. 3A) contains an ion signal
`at m/z 1516 that corresponds to the protonated peptide. This
`assignment is 103 atomic mass units higher than the mass
`expected for the peptide whose sequence was obtained by
`gas-phase sequence analysis. This difference of 103 atomic
`
`FIG. 2. Final purification of guany-
`lin by C8 reverse-phase microbore
`HPLC. Chromatographic peaks (A214,
`0.1 absorbance unit full scale) were
`collected and measured for activity.
`The active peak (55% of a full-scale
`response) is indicated by shading, with
`1% of the fraction giving a 10-fold in-
`crease in cyclic GMP.
`
`I
`10
`
`I
`20
`
`I
`30
`
`Time (min)
`
`mass units is consistent with an additional disulfide-linked
`cysteine or with a threonine at the C terminus of the peptide.
`Reduction of the disulfide bonds of guanylin with dithiothrei-
`tol (DTT) resulted in a 4 atomic mass unit increase in
`molecular weight of the peptide (Fig. 3 B and C), indicating
`that it contains two disulfide bonds. Therefore, since there
`are only three cysteines in the original 14 N-terminal amino
`acids, the 103 atomic mass unit difference must result from an
`additional C-terminal cysteine that is disulfide-linked to one
`of the other three cysteines in the guanylin sequence.
`The resulting full amino acid sequence of the peptide was
`compared with all other proteins in the GenBank, National
`Biomedical Research Foundation, and SwissProt databases
`by a computer-based search. This search revealed that gua-
`nylin has homology with the STs, with the greatest homology
`identified in the cysteine-rich regions of the molecules (19,
`20). The distinctive difference between guanylin and the STs
`is that guanylin possesses four cysteines with two disulfide-
`linked bridges while all of the known STs have six cysteines
`with three disulfide-linked bridges (Fig. 4).
`Chemical synthesis of guanylin based on the experimentally
`derived sequence resulted in three different HPLC fractions
`following oxidation in air. Each of these fractions contained a
`peptide with the same molecular weight as native guanylin
`(1516 atomic mass units) as determined by mass spectrometric
`analysis. However, only one of these fractions exhibited
`potent bioactivity in the T84 cell bioassay consistent with
`guanylin. This fraction also exhibited a similar HPLC reten-
`tion time to that of native guanylin. Since guanylin has four
`cysteine residues, the three fractions of synthetic guanylin
`probably represented the three possible different disulfide
`bridge alignments. Bioactive synthetic guanylin stimulated
`increases in cyclic GMP levels of T84 cells that were time and
`concentration dependent. Guanylin (10-8 M) caused a marked
`elevation of cyclic GMP after 1 min, which progressively
`increased through 30 min (Fig. 5A). Examination of the
`concentration—response curve shows that guanylin elicited an
`increase in cyclic GMP at 10-10 M and this response increased
`through the range of concentrations tested (Fig. 5B). To
`characterize the effect of treatment of reducing agents on the
`bioactivity of guanylin, we pretreated the peptide for 30 min
`with 1 mM DTT. The basal level of cyclic GMP for this
`experiment was 160 ± 50 fmol per well, which increased to
`2820 ± 500 fmol per well after a 30-min treatment with guanylin
`(10' M). However, following the pretreatment of the peptide
`with DTT, the effect of the 30-min treatment with the peptide
`on cyclic GMP was almost completely abolished (250 ± 50
`fmol per well). The action of DTT does not appear to be a
`direct effect of DTT on guanylate cyclase since treatment of
`the cells with 10 µM DTT (final concentration of DTT that the
`cells were exposed to in the experiment) failed to affect their
`responsiveness to STa treatment (data not shown). Finally, we
`examined in preliminary experiments the ability of guanylin to
`displace specifically bound 125I-STa from T84 cells. In this
`experiment, guanylin caused a concentration-dependent dis-
`placement of labeled STa from the T84 cells (Fig. 6) with an
`IC50 of 5 x 10-8 M.
`
`DISCUSSION
`In the present study, we describe the purification and se-
`quence of a rat intestinal peptide that possesses the proper-
`ties consistent with an endogenous ligand for the intestinal
`guanylate cyclase. We have termed this peptide guanylin
`because of its ability to stimulate intestinal guanylate cyclase.
`Synthetic guanylin was found to increase cyclic GMP levels
`in T84 cells in a time- and concentration-dependent manner.
`Guanylin was also found to displace the specific binding of
`' 25I-STa from T84 cells. Therefore, these data support our
`proposal that guanylin is an endogenous activator of the
`
`

`

`950
`
`Pharmacology: Currie et al.
`
`Proc. Natl. Acad. Sci. USA 89 (1992)
`
`100
`
`A
`
`1 5 1 6
`
`1000
`
`1250
`
`1500
`
`1750
`
`2000
`
`2250
`
`100
`
`1515.6
`
`m/z
`
`C
`
`100
`
`cc
`
`1519.6
`
`1510
`
`1515
`
`1520
`
`1525
`
`1510
`
`1515
`
`1520
`
`1525
`
`m/z
`
`m/z
`
`FIG. 3. Electrospray mass spectra of native guanylin. (A) Mass spectral analysis of native guanylin in a range of 1000-2400 atomic mass units
`shows the molecular weight to be 1516 atomic mass units. (B and C) Comparison of the mass spectra of oxidized (B) and reduced (C) native
`guanylin in the range 1510-1525 atomic mass units. R.A., relative abundance.
`
`peaks, each of which contained a peptide of the same
`molecular weight as native guanylin. Since guanylin has four
`cysteine residues, the three fractions of synthetic guanylin
`probably represent the three possible alignments of the
`disulfide bridges. It is likely that the two inactive fractions
`represent improperly folded peptide. The alignment of the
`
`3000
`
`a
`
`2000 -
`
`A
`
`1000 -
`
`20
`10
`Time (min)
`
`30
`
`10 5
`
`1'
`
`
`
`4
`
`10
`
`1'
`
`
`
`3
`
`10
`
`2
`
`10
`
`10
`
`-7 -6
`-8
`-10 -9
`0 10 10 10 10 10
`Guanylin (M)
`
`cGMP (fmol/well)
`
`FIG. 5. Time course (A) and concentration—response (B) effect of
`synthetic guanylin on cyclic GMP levels in T84 cells. In the time
`course experiment, TM cells were treated with 10-8 M guanylin for
`the indicated times. For the concentration—response, the cells were
`incubated with various concentrations of guanylin for 30 min. Cells
`for both experiments were treated with 1 mM IBMX. Values
`represent means ± SE (n = 4).
`
`intestinal guanylate cyclase and suggest that this peptide may
`influence intestinal fluid and electrolyte transport.
`Purification of guanylin was accomplished by capitalizing
`on the stable nature of this peptide, its acidic isoelectric
`point, and its characteristic elution on reverse-phase HPLC.
`Initially, we were concerned that the activity may result from
`bacterial contamination. To limit this possibility the purifi-
`cation was limited to jejunum, which in normal animals is
`considered unlikely to contain considerable bacterial con-
`tamination. Furthermore, we found in preliminary experi-
`ments that every individual rat intestine that we extracted
`possessed bioactivity. We also found that embryonic intes-
`tine, which is considered free of bacteria, exhibits similar
`activity, strongly suggesting that the intestine indeed pos-
`sesses a unique ligand. The structure of guanylin further
`strengthens this proposal. However, definitive proof of the
`intestinal source of guanylin must await a thorough analysis
`of the tissue by immunological and molecular methods.
`The unique structure of the 15-amino acid peptide guanylin
`is characterized by an N-terminal proline, a C-terminal
`cysteine, a glutamic acid, a total of four cysteines, and the
`absence of basic amino acids. The conditions under which
`guanylin was isolated may have given rise to a truncated form
`that possesses the properties required for bioactivity but is
`derived from a larger precursor. The synthetic peptide re-
`quired cyclization for expression of bioactivity and this
`activity was abolished by treatment with the reducing agent
`DTT. Interestingly, during the purification of oxidized syn-
`thetic guanylin, we observed three major chromatographic
`
`GUANYLIN
`
`PNTCE ICAYAACTGC
`
`STa
`
`NT F Y C C ELCCN PA C AGCY
`I
`I
`I
`
`
`FIG. 4. Comparison of the structures of guanylin and STa.
`Identical amino acids are indicated by the dotted lines. The reported
`disulfide alignment for STa (20) is represented by the solid lines.
`
`

`

`Pharmacology: Currie et al.
`
`Proc. Natl. Acad. Sci. USA 89 (1992)
`
`951
`
`-10 -9 -8 -7 -6 -5
`0 10 10 10 10 10 10
`Guanylin (M)
`
`FIG. 6. Displacement of 125I-STa specific binding from T84 cells
`by guanylin. Cells were incubated for 30 min at 37°C with labeled STa
`and various concentrations of guanylin. Specific binding (%) was
`determined by dividing the specifically bound 125I-STa at each
`guanylin concentration by the specifically bound 125I-STa in the
`absence of guanylin. Each point represents the mean of triplicates.
`
`disulfide bridges at this time remains undetermined; how-
`ever, a comparison of the conserved cysteines in guanylin
`with the

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