throbber
PEPTIDES
`
` seees
`ELSEVIER
`Regulatory Peptides 68 (1997) 45-56
`
`
`
`REGULATORY
`
`Purification, cDNA sequence, andtissue distribution of rat uroguanylin
`
`Zhiping Li*, Ashley G. Perkins, Matthew F. Peters, Michael J. Campa, Michael F. Goy
`
`Department of Physiology and Center for Gastrointestinal Biology and Disease at the University of North Carolina, Chapel Hill NC 27599, USA
`
`Received 29 June 1996; revised 17 October 1996; accepted 31 October 1996
`
`Abstract
`
`Guanylin, a peptide purified from rat jejunum,is thought to regulate water and electrolyte balance in the intestine. We show here, using
`a combination of Northern blots, Western blots, and functional assays, that guanylin and its receptor (GCC)are not distributed in parallel
`within the rat intestine. To investigate the possibility that there might be a secondintestinal peptide that serves as a ligand for GCC, we
`assayed tissue extracts for the ability to stimulate cyclic GMP synthesis in a GCC-expressingcell line. Duodenal extracts display a peak
`of biologicalactivity that is not present in colon and that does not comigrate with guanylin or proguanylin. The activity co-purifies with a
`novel peptide (TIATDECELCINVACTGC) that has high homology with uroguanylin, a peptide initially purified from human and
`opossum urine. A rat uroguanylin cDNA clone was found to encode a propeptide whose C-terminus corresponds to our purified peptide.
`Northern blots with probes generated from this clone reveal that prouroguanylin mRNAis strongly expressed in proximal small intestine,
`but virtually absent from colon, corroborating our biochemical measurements. Taken together, these studies demonstrate an intestinal
`origin for uroguanylin, and show that within the intestine its distribution is complementary to that of guanylin. © 1997 Elsevier Science
`BV.
`
`Keywords: GCC; STa receptor; CFTR; Guanylin; Uroguanylin
`
`1. Introduction
`
`A considerable body of evidence supports a role for the
`cyclic GMP pathway in the control of ion transport in the
`gastrointestinal tract. Elevation of intracellular cyclic GMP
`levels in intestinal epithelial cells enhances secretion of
`chloride into the intestinal
`lumen [1], and diminishes
`absorption of sodium and chloride [2]. The combination of
`increased secretion and decreased absorption elevates the
`osmolarity of the lumen, and drives the luminal accumula-
`tion of water. This mechanism was initially identified
`because it can be induced by heat-stable enterotoxin
`(STa)', an 18 amino acid peptide secreted by pathogenic
`
`*Corresponding author. Tel.: +1 919 9666993;
`9666927; e-mail: zpli@med.unc.edu
`enterotoxin; GCA,
`"The
`abbreviations used are: STa, heat-stable
`guanylate cyclase type A; GCB, guanylate cyclase type B; GCC,
`guanylate cyclase type C; HBSS, Hanks’ buffered salt solution; IBMX,
`3-isobutyl-1-methylxanthine; TFA,trifluoroacetic acid; TCA, trichloro-
`acetic acid; BSA, bovine serum albumin; RIA, radioimmunoassay.
`
`fax: +1 919
`
`0167-0115/97/$17.00 © 1997 Elsevier Science BY. All rights reserved
`PIT 80167-0115(96)02103-9
`
`strains of Escherichia coli [3-5]. Exposure to high levels
`of toxin, as occurs during acute bacterial
`infections,
`triggers non-physiological movementof electrolytes, and
`produces a watery diarrhea that can lead to dehydration
`and death.
`When the STa receptor was cloned from a small
`intestinal cDNA library [6], it was found to belong to a
`family of receptors that contain endogenous guanylate
`cyclase (GC) activity. Two other members of this family
`are the natriuretic peptide receptors, GCA and GCB [7].
`Because the STa receptor was the third such receptor
`cloned, it was named GCC. All members of this family
`contain:
`(a) an intracellular catalytic domain responsible
`for the conversion of GTP to cyclic GMP, (b) an intracellu-
`lar regulatory domain that controls the activity of the
`catalytic domain, (c) a single transmembrane domain, and
`(d) an extracellular receptor domain that provides an
`agonist binding site [7].
`These findings led to the hypothesis that GCC serves as
`a receptor for one or more endogenousligands in the GI
`
`MSN Exhibit 1006 - Page 1 of 12
`MSNv. Bausch - IPR2023-00016
`
`

`

`46
`
`Z. Li et al. / Regulatory Peptides 68 (1997) 45-56
`
`opossum uroguanylin) than to guanylin (50% amino acid
`identity with rat guanylin). (c) Cloning of a rat uroguanylin
`cDNA confirms that this second peptide represents the rat
`isoform of uroguanylin. (d) Northern blots show relatively
`selective expression of uroguanylin mRNA in proximal
`small intestine.
`
`tract, by analogy to the way that the other membrane-
`associated guanylate cyclases (GCA and GCB) serve as
`receptors for peptide ligands produced bytissues like the
`heart and brain [7]. Recently, a candidate intestinal pep-
`tide, called guanylin, was purified from jejunal extracts [8].
`Guanylin is believed to be a natural
`ligand for GCC
`because it (a) elevates intracellular cyclic GMP levels in
`GCC-expressing cells
`[8],
`(b) competes with STa for
`binding to GCC [8-10], and (c) stimulates the secretion of
`chloride by intestinal epithelial cells [9,11—15]. One key
`feature shared by guanylin and STa is a set of four
`conserved cysteines connected by specific disulfide bonds;
`Tissues were removed from Sprague—Dawleyrats (250—
`this provides the secondary structure required for bio-
`275 g) under urethane anesthesia (1.6 g urethane/kg
`logical activity [8,16].
`administered via i.p.
`injection). For RNA isolation,
`the
`The mRNAencoding GCCis strongly expressed in the
`whole intestinal tract was removed and put in ice cold
`intestine [6,17], exclusively in epithelial cells [18,19]. No
`Ringer’s-glucose (130 mM Na’, 120 mM Cl, 25 mM
`other tissue tested displays significant
`levels. However,
`HCO,, 1.2 mM Mg”", 1.2 mM Ca”, 2.4 mM K,HPO,,
`binding studies indicate that receptors for guanylin are
`present
`in the kidney [20,21]. Furthermore, STa and
`0.4 mM KH,PO,, 10 mM glucose). Tissues were then
`isolated as rapidly as possible, frozen on dry ice, and
`guanylin can stimulate sodium and potassium excretion by
`stored at — 80°C until used for RNA purification.
`the isolated, perfused kidney [22,23] and can elevate cyclic
`For the preparation of peptide-containing extracts for
`GMP in organ cultured kidney slices
`[21,24]. These
`Western blots,
`the mucosal
`layer of each tissue was
`observations suggest
`that guanylin, or a guanylin-like
`stripped free of the muscle layers, and homogenized in a
`peptide, mayplay a role in regulating kidney function. One
`buffer containing protease inhibitors (25 mM HEPES
`possibility is that guanylin is produced locally in the
`pH=7.4,
`1 mM phenylmethylsulfonylfluoride, 10 w~M
`kidney. Indeed, low levels of guanylin probe hybridization
`trans-epoxysuccinyl-L-leucylamido(4-guanidino)butane, 10
`have been reported on Northern blots of mRNA isolated
`
`
`from rat kidney [25]. However, this observation was not 1 mM_ben-pg/ml aprotinin, 10 pg/ml leupeptin,
`
`
`
`confirmed in comparable studies with mouse [26] or
`zamidine). After homogenization, the extracts were cen-
`human [10] kidney, and, furthermore, only small amounts
`trifuged at 10 000 X g for 20 min and insoluble material
`of guanylin-like bioactivity are present
`in rat kidney
`was discarded. The protein concentration was determined
`extracts [8,27].
`by the bicinchoninic acid method (BCA kit obtained from
`Pierce).
`A secondpossibility is that guanylin or a related peptide
`is delivered to the kidney from some other source. In an
`For the preparation of peptide-containing extracts for
`effort to identify such a peptide, Hamraet al. [28] and Kita
`HPLCfractionation, rat intestines were frozen on dry ice,
`and kept at — 80°C until used. After thawing, the intes-
`et al.
`[29] analyzed opossum and human urine for the
`presence of guanylin-like peptides. They found small
`tines were divided into regions corresponding to duodenum
`and colon. Duodenal tissue was taken as the 4 to 5 cm
`amounts of guanylin, and larger amounts of a second,
`structurally-similar
`peptide,
`which
`they
`named
`uroguanylin. Uroguanylin can bind to the STa/guanylin
`receptor:
`its EC, for activating cyclic GMP synthesis in
`GCC-expressing cells is
`intermediate between that of
`guanylin and STa [28,29], and it can competitively dis-
`place '**I-STa binding [28,29]. Uroguanylin is similar in
`amino acid sequence to guanylin and STa, and it retains
`their characteristic disulfide bond structure. Thus, guanylin
`and uroguanylin define a family of naturally occurring
`peptides that are structurally and functionally related.
`These studies leave open the question of what tissue
`serves as the biological source of uroguanylin found in the
`urine. In our current study, we show that small intestine is
`potentially one such source:
`(a) Rat duodenal extracts
`contain a peptide that displays guanylin-like bioactivity,
`but is chromatographically distinct from guanylin. (b) This
`second
`peptide
`is much more
`closely related
`to
`uroguanylin (80% amino acid identity with human and
`
`to the stomach.
`segment of intestine immediately distal
`Colon was taken to include both proximal and distal
`segments of the large bowel (posterior to the caecum and
`anterior to the sigmoid colon). Tissue pieces were split
`lengthwise and rinsed with normalsaline. The tissue was
`minced in 10 volumes | M acetic acid, placed in a boiling
`water bath for 5 min, and then homogenized. Boiled
`extracts were centrifuged at 4°C for 20 min at 230 000 X g.
`The resulting supernatant fractions were filtered through
`Whatman No.2 paper and applied to Waters C,, Sep-Pak
`cartridges. Unbound and weakly bound material was
`washed through with a solution of 10% acetonitrile—0.1%
`trifluoroacetic acid (TFA) in water. Tightly bound material
`was eluted with 60% acetonitrile—0.1% TFA. The eluted
`material was dried under vacuum and reconstituted either
`
`2. Experimental procedures
`
`2.1. Tissue and Extract Preparation
`
`in bioassay medium (Hanks’ buffered salt solution con-
`taining 1 mM IBMX)for bioassay on T84 cells, or in 10%
`acetonitrile/0.1% TFA for HPLC analysis (see below).
`MSNExhibit 1006 - Page 2 of 12
`MSNv. Bausch - IPR2023-00016
`
`

`

`Z. Li et al. / Regulatory Peptides 68 (1997) 45-56
`
`47
`
`For measuring region-specific responses to STa, seg-
`ments of tissue approximately 4 cm in length were placed
`in a dissecting pan filled with 37°C Ringer’s-glucose
`bubbled continuously with 95% O,-5% CO, Tubular
`sections were cut longitudinally, exposing a flat luminal
`surface, and luminal contents were discarded. Individual
`pieces of tissue (0.5 cm X 0.5 cm) were excised, placed in
`shell vials containing standard Ringer’s-glucose solution at
`37°C, and exposed to test solutions (see below).
`
`2.2. Northern blots
`
`Selected regions of the uroguanylin, guanylin, and GCC
`genes were amplified by PCR and subcloned into plasmid
`vectors (pBS, Stratagene), as described below and in a
`previous publication [18]. The cDNAinserts were isolated
`and used as templates for the synthesis of randomly-
`primed [°’P]-labeled cDNA probes (DECAprime II kit,
`Ambion).
`Total RNA was isolated by standard techniques [30],
`fractionated on a 1% agarose formaldehyde gel (2.2 M
`formaldehyde), and transferred to a nylon membrane
`(ICN). Membranes were treated with 10 ml prehybridiza-
`tion solution (50% formamide, 5X SSPE, 5X Denhardt’s,
`0.25mg/ml sperm DNA, 0.5% SDS) for 3 h and then
`hybridized in 10 ml Northern blot hybridization solution
`(50% formamide, 5X SSPE, 1X Denhardt’s, 0.1 mg/ml
`sperm DNA, 0.1% SDS, 10% dextran sulfate with 10°
`cpm/ml of each [*’P]-labeled probe) at 42°C for 24 h
`(SSPE = 150 mM NaCl, 11.5 mM NaH,PO,,
`1 mM
`EDTA, pH 7.4). Membranes were then washed twice (15
`min each) at room temperature in 2X SSC with 0.1% SDS
`(SSC = 150 mM NaCl, 15 mM Nacitrate, pH 7), followed
`by two 30 min washes at 55°C in 0.1X SSC with 0.1%
`SDS.
`
`2.3. Western blots
`
`Samples of tissue extracts or HPLC fractions were dried
`under vacuum,boiled in electrophoresis sample buffer, and
`fractionated on 15% SDS-—polyacrylamide gels made
`according to standard procedures
`[31] except
`for
`the
`composition of the electrode buffers (upper = 100 mM
`Tris-OH, 100mM tricine, 0.1% SDS, pH 8.3;
`lower =
`200mM Tris-Cl, pH 8.9). After electrophoresis, samples
`were transferred to 0.1 wm nitrocellulose membranes
`(Schleicher and Schuell) using a TE 22 transphor ap-
`paratus (Hoefer Scientific). The membranes were blocked
`with 3% BSA, washed, and incubated for 1 h at room
`temperature with a 1:500 dilution of antiserum 2538, a
`polyclonal antiserum that was raised against a 14 amino
`acid synthetic peptide whose sequence appears near the
`amino terminus of the rat guanylin prohormone [32]. The
`membranes were then washed and incubated with a
`
`secondary antibody (horseradish peroxidase-conjugated
`sheep-antirabbit IgG diluted 1:10 000, Boehringer-Mann-
`
`heim) for 60 min at room temperature. After an additional
`wash,
`the membranes were treated with chemilumines-
`cence reagent as specified by the manufacturer (Boeh-
`ringer-Mannheim), and exposed to Kodak XAR-5 film.
`
`2.4. Bioassay
`
`The studies described below have made use of two
`
`different bioassay procedures. Thefirst procedure was used
`to evaluate the relative ability of colon and duodenum to
`respond to STa (Fig. 1b). Shell vials containing 1 ml
`standard Ringer’s-glucose solution with 0.5 mM IBMX
`were placed in a 37°C water bath. Vials were continuously
`bubbled with 95% O,-5% CO,
`throughout the experi-
`ment. During the period of temperature and gas equilibra-
`tion, 100 units/ml STa (Sigma) was added to the appro-
`priate vials. Pieces of tissue from each region of the gut
`were then placed in the vials for 30 min. Reactions were
`stopped by removing the tissues from the vials and quick
`freezing them on a metalplate resting on a bed ofdry ice.
`The frozen tissue was then homogenized in 6% trichloro-
`acetic acid (TCA) and centrifuged to separate TCA-insolu-
`ble protein from TCA-soluble cyclic GMP. The protein
`pellet was dissolved by heating (37°C) in 1 M NaOH.
`Cyclic GMP was quantitated by RIA [33,34] and protein
`was determined using a dye-binding assay (Bio-Rad
`Bradford Assay) with BSA as
`standard. Results are
`reported as pmol cyclic GMP/mgprotein.
`The second procedure was used to evaluate HPLC
`fractions for guanylin-like activity (Figs. 2-5). This bioas-
`say, based on the method of Currie et al. [8], employs a
`reporter cell line (T84 cells) to detect the presence of GCC
`ligands, as manifested by an increase in intracellular cyclic
`GMP levels. T84 cells are derived from a human colon
`
`carcinoma, and retain many properties of crypt epithelium,
`including expression of high levels of GCC [35].
`In
`addition, because T84 cells express very low levels of the
`other known membrane cyclases [8,36], they provide a
`relatively specific bioassay system for ligands that target
`GCC. The cells were grown to confluency in 12- or
`24-well plastic trays. Cells were then washed two times
`with HBSS and incubated for 10 min at 37°C in HBSS
`
`containing 1 mM IBMX. HPLCfractions were reconsti-
`tuted in bioassay medium, then applied to the cells for an
`additional 30 min. The reaction was stopped by removing
`the incubation solution and replacing it with 0.5 ml ice
`cold 6% TCA.Cells were scraped andtransferred to plastic
`microcentrifuge tubes and the wells were rinsed with an
`additional 0.5 ml ice cold 6% TCA. Protein and cyclic
`GMP content were determined as above. Because the
`
`is quite uniform for T84 cell
`protein content per well
`cultures within a single plating, experimental results are
`reported as pmol cyclic GMP/well rather than pmol cyclic
`GMP/mgprotein.
`Activation of HPLC fractions: In our initial studies, the
`biological activity of our HPLC fractions was quite low.
`MSNExhibit 1006 - Page 3 of 12
`MSNv. Bausch - IPR2023-00016
`
`

`

`48
`
`Z. Li et al. / Regulatory Peptides 68 (1997) 45-56
`
`2.4
`
`1.35
`
`15
`
`—i °
`
`uo
`
`8
`Se
`SE
`gE
`@27

`
`b
`
`B_
`as
`SE
`BE
`Qe
`ge22
`
` a
`blot
`
`1.0
`0.8
`
`SS
`0.6 g
`0.4
`02
`,
`
`Western
`oe 2 blot
`Sanaa
`5
`10
`15
`20
`25
`30
`0
`fraction number
`
`1.0
`0.8
`0.6 0
`g
`0.4 <
`0.2
`,
`
`Western
`

`30
`
`0
`
`5
`
`20
`15
`10
`fraction number
`
`25
`
`0.78
`
`
`response(pmolcGMP/mgprotein)
`bioassay
`
`0SSaeO)S)aaaII1IIInho==nN8onoF&© oon
`
`
`
`signaldensity(arbitraryunits)
`nmnNaOoua
`
`Cc
`
`kDa
`
`35
`30
`
`15
`
`10
`
`Cc
`D
`
`Fig. 1. Distribution of guanylin and GCC along the rostrocaudal axis of
`the GItract. (a) Northern blot of poly A” RNA (10 pg/lane) hybridized
`with radiolabeled guanylin and GCC probes. RNA wasisolated from rat
`stomach (S), duodenum (D), jejunum (J), ileum (1), or colon (C). All
`lanes are from a single membrane hybridized simultaneously with probes
`for GCC (upper), and guanylin (lower). Because the guanylin transcriptis
`much more abundant than the GCC transcript, the lowerhalf of the blot is
`shownafter a 14 h exposure, and the upper half after a 4 day exposure.
`Size standards, in kb, are indicated on the right. Tissue was pooled from
`three animals to obtain the RNA for this blot. Comparable results (not
`shown) have been obtained with RNA isolated from individual animals.
`(b) Cyclic GMP levels in duodenum (D) and colon (C) after incubation
`with (+) or without (—) STa at 100 units/ml. Each bar indicates the
`mean and standard error of nine determinations. (c) Western blot of tissue
`extracts performed with an antibody raised against an amino terminal
`domain of the guanylin prohormone. The autoradiograms (left) are of
`representative duodenal (D) and colonic (C) samples obtained from a
`single animal, with the positions of molecular weight standards as
`indicated. The bar graph (right) presents the densitometrically-determined
`mean (+SEM)ofdata obtained from four separate animals.
`
`(a) Two
`Fig. 2. HPLC comparison of colonic and duodenal extracts.
`extracts (generated from 15 and 30 colons, respectively) were indepen-
`dently analyzed by reverse phase HPLC,as described in Section 2. Upper
`trace: representative UV absorbance profile from one of the extracts.
`Middle trace (dashed line): the gradient of acetonitrile used to elute the
`column. Lower trace: bioassay responses to individual column fractions
`(mean+range from the two column runs, normalized as a percentage of
`the maximum response to correct for differences in the potencies of the
`extracts). Fractions were preincubated at 37°C before the bioassay was
`performed,
`in order to enhance their activity (see Section 2). At the
`bottom of the panel are Western blot data for HPLC fractions from one
`column run (pooled in pairs, except for fractions 19-26, which were
`analyzed individually) using an antibody that recognizes the N-terminus
`of the guanylin prohormone. An abbreviated region of the gel containing
`the immunoreactive proguanylin band is shown. The black and white
`arrows indicate the retention times of synthetic rat guanylin and opossum
`uroguanylin standards, respectively. (b) Three extracts (each generated
`from 15-30 duodena) were independently chromatographed under con-
`ditions identical to those employed for colonic extracts. UV absorbance,
`elution profile, and bioassay responses are plotted as in panel (a).
`
`Therefore, in order to obtain duplicate bioassay measure-
`ments, we dried each HPLCfraction, reconstituted it in
`bioassay buffer, applied the entire fraction to one well of
`T84 cells for 30 min, and then transferred it to a second
`well for 30 min. We noticed that the response of the cells
`in the second well was alwayssignificantly greater than the
`response of the cells in the first well, suggesting that
`something was happening during the first incubation to
`activate the sample. Further experiments showed that
`samples could be activated simply by incubating them at
`37°C for 30 min. As described in Section 3, we believe
`that this is due to proteolytic cleavage by a contaminating
`protease. We have used this activation procedure to evalu-
`ate samples in all of the HPLC analyses presented below;
`in some cases the activation was achieved by incubating
`the sample on a well of T84 cells at 37°C, in others by
`MSNExhibit 1006 - Page 4 of 12
`MSNv. Bausch - IPR2023-00016
`
`

`

`Z. Li et al. / Regulatory Peptides 68 (1997) 45-56
`
`49
`
`0.01 0.00
`
`0.04
`0.03
`
`0.02 A280
`
`oOo oa
`
`°°
`
`bioassayresponse(pmol
`
`cGMP/well)
`
`0
`
`5
`
`20
`15
`10
`fraction number
`
`25
`
`30
`
`Fig. 4. Preincubation at 37°C leadsto a shift in the retention time of the
`duodenal peptide. An extract of 30 duodena was fractionated as in Fig.
`2b. Fractions 23 and 24 were pooled, dried under vacuum (to remove
`HPLCsolvents), resuspended in bioassay medium, incubated for 60 min
`at 37°C, and rechromatographed on the same column using the same
`elution profile. The upper trace shows UV absorbance; the dashed line
`showsthe elution profile; the lower trace shows bioassay responses to
`individual fractions. Biological activity now elutes in fraction 16, while
`most of the UV absorbing material continues to elute in fractions 23 and
`24. The large peak marked with the asterisk is due to IBMX, which was
`present in the bioassay medium.
`
`=2
`
`control
`(n =3)
`
`figure 2a
`fraction 21
`(n =2)
`
`figure 2b
`fraction 23/24
`(n= 6)
`
`guanylin
`(n=3)
`
`uroguanylin
`(n=2)
`
`STa
`(n= 2)
`
`figure 4
`fraction 16
`(n=1)
`
`0
`
`0.5
`
`1
`
`20
`
`40
`
`60
`
`80
`
`cGMP pmol/well
`
`0.02 0.01
`
`$=
`Fig. 3. Preincubation at 37°C enhances the activity of HPLC column
`5g
`fractions, but not of synthetic guanylin or uroguanylin. The bars show the
`£2
`
`mean level of cyclic GMP (+SEM orrange) in T84 cells after exposure
`
`
`zoaS
`to the indicated stimuli. Each stimulus was either held at 4°C (C4) or
`incubated at 37°C (Ml)prior to applyingit to the cells. The numbers used
`gs
`ge o°
`to identify the HPLC fractions correspond to (i) the figure in which the
`0 TTTTTTT
`pertinent HPLC run is shown and(ii) the appropriate fraction number(s)
`0
`20
`40
`60
`80
`100
`120
`from that column run.
`fraction number
`
`0.00
`
`1.0
`
`incubating it without cells at 37°C. We have the impression
`that the activation process occurs more efficiently if T84
`cells are present, but we have not compared the two
`procedures in enough detail to be certain ofthis.
`
`2.5. HPLC purification
`
`2.5.1, Step 1
`Reconstituted Sep-Pak fractions were fractionated on a
`PepRPC HRS5S/5 (C,,) column (Pharmacia), pre-equili-
`brated with 10% buffer B (buffer A= 0.1% TFA; buffer
`B= 99.9% acetonitrile + 0.1% TFA). After sample appli-
`cation, the column waseluted isocratically for 5 min with
`10% buffer B, followed bya linear gradient to 50% buffer
`B over 43 min, and a final 12 min elution at 100% buffer
`B. One ml fractions were collected at a flow rate of 0.5
`ml/min, while absorbance was monitored at 214 and 280
`nm. After chromatography, a portion of each fraction was
`lyophilized (to eliminate the acetonitrile and TFA), re-
`suspended in bioassay medium, incubated at 37°C for 30
`
`Fig. 5. Final step in purification of the duodenal peptide. Fraction 16 from
`the column run illustrated in Figure 4 was dried under vacuum,
`resuspended in 10% acetonitrile + 0.1% TFA, and rechromatographedas
`described in Section 2. The upper trace shows UV absorbance; the dashed
`line showsthe elution profile; the lower trace shows bioassay responses to
`individual fractions.
`
`min to activate the peptide (see above), and tested for
`activity on T84 cells. Typical examples of such chromato-
`grams are shown in Fig. 2.
`
`2.5.2. Step 2
`fractions with activity were combined for
`Duodenal
`further purification. The pooled fractions were incubated at
`37°C for 60 min, then reapplied to the PepRPC HR5/5
`column and eluted without modification of the protocol
`described in step 1. The duodenal peptide now elutes
`earlier than in step 1, whereas most contaminants retain
`their original
`retention time. The fractions containing
`activity were identified by bioassay, as shownin Fig. 4.
`MSNExhibit 1006 - Page 5 of 12
`MSNv. Bausch - IPR2023-00016
`
`

`

`50
`
`Z. Li et al. / Regulatory Peptides 68 (1997) 45-56
`
`2.5.3. Step 3
`Active fractions from step 2 were pooled and applied to
`a Cy, analytical column (Vydac), pre-equilibrated with
`10% buffer B (buffer A, 10 mM ammonium acetate pH
`6.2; buffer B, 99% acetonitrile +10 mM ammonium
`acetate pH 6.2). The column waseluted isocratically for 10
`min with 10% buffer B, followed by a linear gradient to
`40% buffer B over 160 min, and a final 10 min elution at
`100% buffer B. Fractions (1.5 ml) were collected at a flow
`rate of 1 ml/min, and analyzed by bioassay, as shown in
`Fig. 5.
`Synthetic 15 amino acid rat guanylin and opossum
`uroguanylin (used as HPLC standards) were generously
`provided by Drs. Ding Chang (Peninsula Labs) and
`Leonard Forte (University of Missouri), respectively.
`
`2.6. Peptide characterization
`
`Sequencing was performed on a sample of peptide
`adsorbed to a 0.22 micron PVDF membrane, using a
`Perkin Elmer/ABI model 491 sequencer with on-line PTH
`amino acid detection.
`
`2.7. CDNA analysis
`
`Rat duodenal RNA wasprepared as described previous-
`ly [18], and reverse transcribed with SuperScript II using
`an oligo dT primer (Gibco BRL). The resulting cDNA was
`then subjected to PCR, using degenerate primers designed
`from areas of high homology in published sequences of
`human and opossum uroguanylin [37,38]. The sense
`primer, TACATCCAGTA(CT)(GC)A(AG)GCCTTCC,and
`antisense primer, GCAGCC(GT)GTACA(GC)GC(AC)-
`ACGTT,correspond, respectively, to base pairs 104—124
`and 334-354 of the human transcript
`[38]. PCR was
`performed for 40 cycles (denaturation for 1 min at 94°C,
`annealing for 1 min at 55°C, and extension at 72°C for 2
`min) followed by a final extension for 10 min at 72° C
`using Taq polymerase (Boehringer Mannheim). A 250 bp
`product was amplified, as expected. This PCR product was
`subcloned into the pBluescript II SK vector using the
`T/A cloning method [39]. DNA was sequenced at
`the
`UNC-CH Automated DNA Sequencing Facility on a
`Model 373A DNA Sequencer (Applied Biosystems) using
`the Taq DyeDeoxy™ Terminator Cycle Sequencing Kit
`(Applied Biosystems). Sequence analysis revealed a high
`degree of homology at the nucleotide level to the human
`and opossum forms of uroguanylin (72 and 60%, respec-
`tively), confirming that we had amplified the appropriate
`target sequence.
`To determine the full-length uroguanylin cDNA se-
`quence, we used the PCR product to produce a random-
`primed probe for screening a rat duodenal cDNAlibrary
`constructed in AGT-11 [40]
`(a gift
`from Dr. Andrew
`Leiter). Phage plaques were adsorbed onto nitrocellulose
`filters, hybridized with the probe in 50% formamide, 0.8 M
`
`NaCl, 20 mM PIPES pH 6.5, 0.5% SDS and 100 pg/ml
`salmon sperm DNA at 42°C, and washed in 0.1X SSC/
`0.1% SDS at 55°C. Positive plaques were purified by
`sequential low-density plating and bacteriophage DNA was
`isolated with a Qiagen Lambda kit (Qiagen). The cDNA
`insert was excised with EcoRI and subcloned into the
`
`pBluescript II SK vector for sequencing, as described
`above.
`
`3. Results
`
`3.1. Guanylin and its receptor are not distributed in
`parallel in the GI tract
`
`Previous studies have shown that guanylin mRNA is
`expressed in a rostrocaudal gradient, ranging from quite
`low in duodenum to quite high in colon [9,32,41,42]. In
`contrast, as shown in Fig.
`la, the mRNA encoding the
`guanylin receptor
`(GCC)
`is expressed at high levels
`throughout the GI tract. These data reveal a nonparallel
`distribution of guanylin and its receptor, with the mismatch
`particularly evident in duodenum.
`in
`To verify that
`the high level of GCC transcript
`duodenum corresponds to a high level of functional
`receptor, we applied a sub-saturating dose of a GCC-
`specific ligand (STa) to excised pieces of tissue in organ
`culture, and measured intracellular levels of cyclic GMP
`after 30 min of exposure (a time at which the responseis
`still proceeding linearly). This experiment was performed
`in the presence of a phosphodiesterase inhibitor, isobutyl
`methylxanthine (IBMX), to minimize the effects of phos-
`phodiesterase enzymes on cyclic GMP metabolism. The
`agonist-dependent
`increase in cyclic GMP levels (the
`amount of cyclic GMP in STa-stimulated tissue minus the
`amountin unstimulated control tissue) is slightly greater in
`duodenum than it
`is in colon (Fig. 1b),
`indicating that
`duodenum does express substantial
`levels of functional
`receptor.
`To verify that the low level of guanylin transcript in
`duodenum corresponds to a low level of guanylin prop-
`eptide, we performed Western blots on extracts of
`duodenum and colon, using an antibody that recognizes the
`N-terminus of the propeptide [32]. Extracts were prepared
`from four separate animals;
`in each case the level of
`proguanylin in duodenum, as measured densitometrically,
`was less than 11% of the level
`in colon (mean = 8.7%
`+£1.0% (SEM)) (Fig. Ic).
`
`3.2. HPLC analysis of guanylin-like peptides in
`intestinal extracts
`
`The ligand/receptor mismatch described above suggests
`the possibility that duodenum might produce a ligand that
`resembles guanylin in its
`receptor specificity, but
`is
`biochemically distinct. In order to compare guanylin-like
`MSNExhibit 1006 - Page 6 of 12
`MSNv. Bausch- IPR2023-00016
`
`

`

`Z. Li et al. / Regulatory Peptides 68 (1997) 45-56
`
`51
`
`peptides expressed in duodenum to those expressed in
`colon, we prepared aqueous extracts of each tissue, frac-
`tionated the extracts by HPLC, and assayed an aliquot of
`each fraction for
`its ability to stimulate cyclic GMP
`synthesis in cultured T84 cells, as described in the Section
`2. In parallel with these bioassays, we performed Western
`blots on a sample of each HPLC fraction, using the
`antibody described in Fig. 1c. For convenience, only the
`region of the blot containing immunoreactive proguanylin
`is shown in the figures below.
`Colonic extracts contain both guanylin and proguanylin.
`Fig. 2a illustrates the HPLC analysis of a colonic extract.
`The Western blot inset at the bottom of the figure shows
`that proguanylin elutes in fraction 21. When the fractions
`are bioassayed, a peak of cyclic GMP-promotingactivity is
`also seen in fraction 21. In the course of characterizing this
`peak, we noticed that its biological activity increases with
`timeif it is incubated at 37°C (Fig. 3). Such an increase is
`not
`observed when
`synthetic
`guanylin,
`synthetic
`uroguanylin, or commercially-purified STa are incubated
`under similar conditions (Fig. 3). We do not yet know the
`mechanism by which this time-dependent enhancement of
`activity occurs; however, as discussed below, we consider
`it likely that proguanylin, which is biologically relatively
`inactive
`[10,25,42,43], has co-eluted with a protease
`capable of converting it to a smaller, more active peptide.
`Hamra et al. have previously demonstrated that specific
`proteases enhancethe activity of proguanylin [44].
`Duodenal extracts contain biologically-active material
`that is distinct from both guanylin and proguanylin. When
`duodenal extracts are analyzed by HPLC, proguanylin-like
`immunoreactivity is again observed in fraction 21 (West-
`ern blot inset, Fig. 2b), though, as expected, the amountis
`muchless than can be seen in comparable colonic extracts
`(Western blot inset, Fig. 2a). The T84 cell assay confirms
`that this duodenal proguanylin is associated with a small
`peak of biological activity, which, as above, can be
`enhanced by preincubation at 37°C. However,
`the most
`conspicuousaspect of the chromatogram is the presence of
`a second, much larger peak of activity in fractions 23 and
`24. This material differs from proguanylin in two ways: it
`is retained more tightly by the column,andit fails to react
`with our proguanylin-specific antibody. In addition,
`the
`retention time of the material is significantly different from
`that of synthetic rat guanylin (fraction 13) or synthetic
`opossum uroguanylin (fraction 15). The novel duodenal
`peak does, however, share one property with proguanylin:
`its activity is also enhanced by preincubation at 37°C (Fig.
`3). If an intestinal protease is indeed responsible for this
`activation phenomenon,then the data suggest that such a
`protease is likely to come off the HPLC column in a broad
`peak that overlaps both proguanylin (fraction 21) and the
`material in fractions 23-24.
`In order to test whether the duodenal material
`
`is a
`
`peptidase, and S. aureus V8 protease). After a 2 h
`incubation,its biological activity was completely destroyed
`(data not shown). This, in conjunction with the purification
`and sequencing studies described below, confirms the
`peptide nature of the active material in fractions 23-24.
`
`3.3. Preincubation alters the chromatographic properties
`of the duodenal peptide
`
`If a proteolytic mechanism is responsible for the activa-
`tion of the duodenal peptide, then the size of the molecule
`should be altered once it has been activated, and this
`should be reflected by a change in its chromatographic
`properties. To test this prediction, we partially purified the
`duodenal peptide on our standard reverse phase HPLC
`column (as in Fig. 2b), allowed it to preincubate at 37°C,
`rechromatographed it on the same HPLC column, and
`bioassayed the resulting fractions

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