throbber
ELSEVIER
`
`REGULATORY
`
`PEPTIDES
`
`2 75 99,
`
`

`

`46
`
`Z Li et al
`
`/ Regulatory Peptides
`
`–
`68 (1997) 45 56
`
`tract, by analogy to the way that the other membrane-
`associated guanylate cyclases (GCA and GCB) serve as
`receptors for peptide ligands produced by tissues 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;
`this provides the secondary structure required for bio-
`logical activity [8,16].
`The mRNA encoding GCC is strongly expressed in the
`intestine [6,17], exclusively in epithelial cells [18,19]. No
`other tissue tested displays significant
`levels. However,
`binding studies indicate that receptors for guanylin are
`present
`in the kidney [20,21]. Furthermore, STa and
`guanylin can stimulate sodium and potassium excretion by
`the isolated, perfused kidney [22,23] and can elevate cyclic
`GMP in organ cultured kidney slices [21,24]. These
`observations suggest
`that guanylin, or a guanylin-like
`peptide, may play a role in regulating kidney function. One
`possibility is that guanylin is produced locally in the
`kidney. Indeed, low levels of guanylin probe hybridization
`have been reported on Northern blots of mRNA isolated
`from rat kidney [25]. However, this observation was not
`confirmed in comparable studies with mouse [26] or
`human [10] kidney, and, furthermore, only small amounts
`of guanylin-like bioactivity are present
`in rat kidney
`extracts [8,27].
`A second possibility is that guanylin or a related peptide
`is delivered to the kidney from some other source. In an
`effort to identify such a peptide, Hamra et al. [28] and Kita
`et al. [29] analyzed opossum and human urine for the
`presence of guanylin-like peptides. They found small
`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
`50
`GCC-expressing cells is intermediate between that of
`guanylin and STa [28,29], and it can competitively dis-
`125
`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
`
`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.
`
`2. Experimental procedures
`
`2.1.
`
`Tissue and Extract Preparation
`
`"
`
`!
`
`Tissues were removed from Sprague–Dawley rats (250–
`275 g) under urethane anesthesia (1.6 g urethane/kg
`administered via i.p.
`injection). For RNA isolation,
`the
`whole intestinal tract was removed and put in ice cold
`Ringer’s-glucose (130 mM Na , 120 mM Cl
`, 25 mM
`2!
`2!
`HCO , 1.2 mM Mg , 1.2 mM Ca
`, 2.4 mM K HPO ,
`"
`3
`2
`4
`0.4 mM KH PO , 10 mM glucose). Tissues were then
`2
`4
`isolated as rapidly as possible, frozen on dry ice, and
`stored at " 80&C until used for RNA purification.
`For the preparation of peptide-containing extracts for
`Western blots,
`the mucosal
`layer of each tissue was
`stripped free of the muscle layers, and homogenized in a
`buffer containing protease inhibitors (25 mM HEPES
`pH $ 7.4, 1 mM phenylmethylsulfonylfluoride, 10 )M
`trans-epoxysuccinyl-L-leucylamido(4-guanidino)butane, 10
`)g/ml aprotinin, 10 )g/ml
`leupeptin, 1 mM ben-
`zamidine). After homogenization, the extracts were cen-
`trifuged at 10 000 # g for 20 min and insoluble material
`was discarded. The protein concentration was determined
`by the bicinchoninic acid method (BCA kit obtained from
`Pierce).
`For the preparation of peptide-containing extracts for
`HPLC fractionation, rat intestines were frozen on dry ice,
`and kept at " 80&C until used. After thawing, the intes-
`tines were divided into regions corresponding to duodenum
`and colon. Duodenal tissue was taken as the 4 to 5 cm
`segment of intestine immediately distal to the stomach.
`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 normal saline. The tissue was
`minced in 10 volumes 1 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 # g.
`The resulting supernatant fractions were filtered through
`Whatman No. 2 paper and applied to Waters C Sep-Pak
`18
`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
`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).
`
`

`

`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
`2
`2.
`sections were cut longitudinally, exposing a flat luminal
`surface, and luminal contents were discarded. Individual
`pieces of tissue (0.5 cm # 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 cDNA inserts were isolated
`and used as templates for the synthesis of randomly-
`32
`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
`6
`sperm DNA, 0.1% SDS, 10% dextran sulfate with 10
`32
`cpm/ml of each [ P]-labeled probe) at 42&C for 24 h
`(SSPE $ 150 mM NaCl, 11.5 mM NaH PO , 1 mM
`2
`4
`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 Na citrate, 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 )m 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. The first 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-
`2
`2
`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 metal plate resting on a bed of dry 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/mg protein.
`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. HPLC fractions 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 and transferred 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
`protein content per well
`is quite uniform for T84 cell
`cultures within a single plating, experimental results are
`reported as pmol cyclic GMP/well rather than pmol cyclic
`GMP/mg protein.
`Activation of HPLC fractions: In our initial studies, the
`biological activity of our HPLC fractions was quite low.
`
`

`

`68 (1997) 45 56
`
`a
`
`100
`-8-- 75
`N2 2 50
`
`mo
`
`25
`
`0
`
`b
`
`_J
`
`15
`10
`20
`fraction number
`
`25
`
`0I
`
`1.0
`
`0.8
`
`0.6 g
`
`0.4 <
`
`0.2
`
`0.0
`
`4 iVe blot Tr
`30
`
`1 . 0
`0.8
`0.6 00
`
`0.4 a
`
`0.2
`
`0.0
`
`0
`
`100
`
`co
`
`0
`
`C
`
`a
`
`100
`
` 75
`z
`E
`.5 50
`
`E
`8u
`0
`
`25
`0
`
`
`6-
`
` 6
`
`1'5'
`10
`20
`fraction number
`
`-11 1Vebtr
`30
`
`25
`
`a S D J
`
`I C
`
`
`IP
`
`kb
`9.5
`7.5
`—
`_ 4.4
`
`-
`
`2.4
`
`eed- 1.35
`
`—
`
`0.78
`
`15
`
`C
`
`0
`STa:
`
`-
`
`kDa
`
`- 29
`
`.4 - 18
`
`- 14.3
`
`35-
`
`30-
`25-
`20-
`15-
`10-
`
`5-
`
`ignal density (arbitrary units)
`
`- 6.2
`- 2.3 s
`
`100
`

`

`control
`(n = 3)
`
`figure 2a
`fraction 21
`(n = 2)
`
`figure 2b
`fraction 23/24
`(n = 6)
`
`guanylin
`(n = 3) =IN
`
`uroguanylin
`(n = 2)
`
`STa
`(n = 2)
`
`figure 4
`fraction 16
`(n = 1)
`
`0
`
`0.5
`
`1
`
`20 40 60 80
`
`cGMP pmol/well
`
`2.5.
`
`2.5.1.
`
`1
`
`68 (1997) 45 56
`
`100
`
`1.0
`
`as
`>,O 0.5-
`‘i 2
`
`8 k 0.0
`
`0.04
`
`0.03
`
`0.02 co
`
`0.01
`0.00
`
`20
`15
`10
`fraction number
`
`25
`
`30
`
`0.02
`
`0.01
`
`2--
`
`0.00
`
`100
`
`120
`
`•
`
`.
`
`.
`
`.
`
`.
`
`.
`
`80
`60
`40
`fraction number
`
`100
`
`ao
`
`0
`
`2.0
`
`3
`g d 1.5-
`05
`22 1.0-
`
`E 0.5
`
`0
`
`r
`
`.
`
`.
`
`.
`
`.
`
`20
`
`0
`
`2.5.2.
`
`2
`
`

`

`50
`
`Z Li et al
`
`/ Regulatory Peptides
`
`–
`68 (1997) 45 56
`
`Step
`3
`2.5.3.
`Active fractions from step 2 were pooled and applied to
`a C
`analytical column (Vydac), pre-equilibrated with
`18
`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 was eluted 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 was prepared 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
`TM
`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 cDNA library
`constructed in (GT-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 )g/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
`
`Guanylin and its receptor are not distributed in
`3.1.
`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. 1a, 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 response is
`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
`amount in 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. 1c).
`
`HPLC analysis of guanylin-like peptides in
`3.2.
`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
`
`

`

`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-promoting activity is
`also seen in fraction 21. In the course of characterizing this
`peak, we noticed that its biological activity increases with
`time if 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 enhance the 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 amount is
`much less 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
`conspicuous aspect 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, and it 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.
`is a
`In order to test whether the duodenal material
`peptide, we incubated it with a mixture of exogenously-
`added proteases (trypsin, chymotrypsin, elastase, amino-
`
`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.
`
`Preincubation alters the chromatographic properties
`3.3.
`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. Although biological
`activity is still quite evident (Fig. 4), it now elutes from the
`column much earlier (fraction 16), consistent with cleav-
`age to a smaller (less hydrophobic) peptide. Furthermore,
`the material in fraction 16 has become fully active: no
`enhancement of the T84 cell response can be induced by
`additional preincubation at 37&C (Fig. 3).
`
`3.4.
`
`Purification of the duodenal peptide
`
`In order to achieve further purification, the material in
`fraction 16 (Fig. 4) was again applied to a C-18 column,
`but now subjected to a new set of elution parameters using
`a different ion pairing reagent and a different pH (see
`Section 2). Activity was recovered as a single, sharp peak
`that aligns precisely with a major peak of UV absorbance,
`and appears well separated from other contaminants (Fig.
`5).
`This material was submitted for amino acid sequencing,
`revealing the presence of approximately 15–18 pmol of the
`peptide shown at the top of Fig. 6a. During the sequencing
`reaction, no amino acid could be identified in cycles 7, 10,
`15, and 18, indicating that the amino acid at each of those
`positions is most likely cysteine. This is consistent with the
`observation that the blank cycles align exactly with a set of
`four cysteines that are absolutely required for biological
`activity in all known guanylin-like peptides (guanylin,
`uroguanylin, and STa-see Fig. 6a). Also,
`in the first
`sequencing cycle, both threonine and glutamate were
`found in approximately equal abundance and thus the
`residue at this position is ambiguous. The sequence of the
`purified peptide suggests that it may be the rat homolog of
`uroguanylin.
`
`Sequence and tissue distribution of the rat
`3.5.
`uroguanylin transcript
`
`In order to confirm and extend our peptide sequence
`analysis, we used PCR to generate a 250 bp nucleotide
`
`

`

`68 (1997) 45 56
`
`a
`
`C
`T/E I ATCI F
`C
`O E L,'
`C
`N
`D
`PN T
`C
`SH T
`C
`P GT
`C
`P N T
`C
`NTFYC
`C
`
`E
`E
`E
`E
`E
`E
`E
`
`L C
`L C
`L C
`I C
`I C
`C
`C
`L C
`
`V
`I NVACT
`I NVAC T
`v N V A C T
`AYAACT
`AFAAC t.
`AYAACT
`A Y A A C T
`C
`P litie A
`
`GC
`GC
`G C•L
`G C
`G C,
`GC
`GC
`,GCY
`
`rat uroguanyl in
`opossum uroguanyl in
`human uroguanyl in
`rat guanyl in
`opossum guanyl in
`human guanyl in
`mouse guanyl in
`E Col i STa
`
`b
`
`I
`
`cgttgtcgactgtcoggcagoaacccataggt2tgagctgggaagccggg -1
`ATGTCAGGAAGCCAACTGIGGGCTGCTGTACTCCIGCTGCTGGIGCTGCAGAGTGCCCAG
`60
`M
`S
`G
`S
`O
`L
`W
`A
`A
`V
`L
`L
`L
`L
`V
`L
`O S
`A
`O
`GGTGTCTACATCAAGTACCATGGCTTCCAAGTCCAGCTAGAATCGGTGAAGAAGCTGAAT 120
`21 G
`V
`Y
`I
`K
`Y
`H
`G
`F
`O
`L
`N
`V
`O
`L
`E S
`V
`K
`K
`GAGTTGGAAGAGAAGCAGAIGTCCGATCCCCAGCAGCAGAAAAGTGGCCICCICCCCGAT 180
`41 E
`L E E K 0 M S D P G
`O O
`K S G L L
`P D
`GIGTGCTACAACCCCGCCITGCCCCIGGACCTCCAGCCTGTTIGTGCATCCCAGGAAGCT
`V
`C
`Y
`N
`P
`A
`L
`P
`L
`D
`L
`O
`P
`V
`C
`A
`S O
`E
`A
`GCCAGCACCTICAAGGCCTIGAGGACCATTGCCACTGATGAATGTGAGCTGTGTATAAAT
`A
`S
`T
`F
`K
`A
`L
`R
`T
`I
`A
`T
`D
`E
`C
`E
`L
`C
`I
`NI
`GITGCCIGTACGGGCTgCtgatgaciatgactccagacaccttacccccacagcctaccct
`V
`A
`C
`T
`G
`CI stop
`gcccatacttaggtaccattgacataattaccaccctcccagcacaaatggatccatagc 420
`ciagacaatatggatgcagagccgccatatttggtccccaggcagctgcaccggaataaaa 480
`atctgacagtcgacao
`496
`
`ei
`
`RI
`
`loI
`
`240
`
`300
`
`360
`
`C
`
`d D J I pCdC H K U Lu S T
`
`L ESVKKL
`
`L ESVKOL
`LESVKKL
`LESVKKL
`
`rat uroguanyl in
`opossum uroguanyl in
`human uroguanyl in
`rat guanyl in
`human guanyl in
`mouse guanyl in
`
`

`

`Z Li et al
`
`/ Regulatory Peptides
`
`–
`68 (1997) 45 56
`
`53
`
`opossum
`and
`human
`for
`established
`previously
`prouroguanylin (70% and 73% identity at the nucleotide
`level, and 67% and 66% identity at the amino acid level,
`respectively). In addition, as has been noted in these other
`species [37,38], there are two discrete regions of homology
`between rat prouroguanylin and rat proguanylin. The first
`region (boxed in Fig. 6b, and compared across all species
`for which sequences are available in Fig. 6a) comprises the
`biologically active C-terminus of each propeptide. The
`second region (underlined in Fig. 6b, and compared across
`all species for which sequences are available in Fig. 6c) is
`found closer to the N-terminus, separated from the con-
`served C-terminal domain by about 50 residues. The
`significance of this second conserved motif is not clear, but
`its retention across multiple species suggests that it has
`some as-yet unknown biological function.
`The tissue distribution of the uroguanylin transcript is
`shown in Fig. 6d. The size of the uroguanylin transcript is
`approximately 600 bp, just slightly smaller than the size of
`the guanylin transcript. It
`is clear that,
`like guanylin,
`uroguanylin mRNA is expressed primarily in the intestine.
`However, within the intestine,
`its distribution is quite
`distinct from that of the guanylin transcript (compare Fig.
`6d to Fig. 1a): uroguanylin is most prominent in proximal
`small intestine, while guanylin is most prominent in distal
`large intestine. This confirms the distribution of guanylin
`and uroguanylin peptides determined in our biochemical
`studies (Fig. 2).
`In addition, there is a limited amount of uroguanylin
`expression outside the intestine,
`in kidney,
`testis, and
`possibly spleen (the exposure time shown in Fig. 6d for
`non-intestinal tissues is 20 times longer than the exposure
`time for intestinal tissues).
`
`4. Discussion
`
`Guanylin was initially discovered in a search for endog-
`enous agonists for
`the STa receptor. Guanylin fulfils
`several of the criteria expected of such an agonist: it is
`present in the intestine, it competes with STa for binding to
`the receptor, and it activates cyclic GMP synthesis when
`applied to cells that express the receptor. Therefore, in a
`simplest-case scenario, guanylin expression might be ex-
`pected to be high in tissues where receptor expression is
`high, and low in tissues where receptor expression is low.
`However, within the intestine the tissue distributions of
`guanylin and its receptor are surprisingly non-parallel. We
`were particularly struck by the fact that duodenum expres-
`ses high levels of receptor but low levels of peptide. In the
`experiments described above, we have confirmed this
`mismatch at both the mRNA and polypeptide levels.
`These observations led us to consider the possibility that
`duodenum might produce its own endogenous ligand that
`resembles guanylin in its target specificity. Our HPLC
`
`analysis of duodenal extracts has confirmed this idea, and
`allowed us to purify a second peptide with guanylin-like
`biological activity. Interestingly, guanylin and the new
`peptide have complementary distributions: duodenum has
`low levels of guanylin and high levels of the new peptide,
`while colon has high levels of guanylin and little or none
`of the new peptide. Alignment of the sequence of the
`duodenal peptide with the appropriate regions of
`rat
`guanylin and uroguanylin (Fig. 6a) reveals that the new
`peptide is more closely related to uroguanylin (80%
`identity when compared across species, with nearly all
`differences representing conservative amino acid substitu-
`tions) than it is to guanylin (47–53% identity, with few of
`the differences representing conservative amino acid sub-
`stitutions). These observations suggest that the duodenal
`peptide represents the rat isoform of uroguanylin, whose
`sequence has not yet been determined.
`In a previous study, Hamra et al. [28] analyzed extracts
`of opossum intestinal mucosa by preparative isoelectric
`focusing, and found two distinct peaks of guanylin-like
`activity. Purification and sequencing of one of the peaks
`(pI $ 5.2) showed that
`it was the opossum form of
`guanylin. The other peak was not purified, but its isoelec-
`tric point (pI $ 3.0) was consistent with the idea that it
`could be uroguanylin. In a subsequent publication, Hamra
`et al. [44] showed that opossum intestine contains both
`proguanylin and a second inactive propeptide whose
`biological activity could be enhanced by proteolysis with
`chymotrypsin. Purification and C-terminal sequencing of
`this second peptide revealed that
`it was distinct from
`proguanylin, and that it had properties more closely related
`to those of uroguanylin than to those of guanylin. Our
`present study provides a direct biochemical demonstration
`that the rat intestine produces a peptide with an N-terminal
`sequence corresponding to uroguanylin. These results,
`together with the analysis of
`rat uroguanylin mRNA
`expression presented in Fig. 6d, provide convincing evi-
`dence that uroguanylin is produced by the rat intestine. In
`addition, recent cloning studies have identified human [38]
`and opossum [37] cDNAs encoding a uroguanylin prop-
`eptide, and have demonstrated expression of a transcript
`encoding this propeptide in the human and opossum
`intestine.
`In the course of identifying and purifying uroguanylin,
`we learned that
`the biological activity of the partially
`purified material could be enhanced by incubating it at
`37&C. This enhancement was accompanied by a shift in its
`HPLC retention time. We have observed a similar phenom-
`enon with partially purified proguanylin extracted from the
`colon. As it is unlikely that this is a spontaneous process,
`we believe it is most likely that a co-eluting protease is
`converting inactive or weakly active precursors into active
`products. As of yet, the peptide processing sites respon-
`sible for generating biologically active guanylin or
`uroguanylin in vivo have not been identified. Thus, the
`N-terminally extended form of uroguanylin that we have
`
`

`

`54
`
`Z Li et al
`
`/ Regulatory Peptides
`
`–
`68 (1997) 45 56
`
`identified may actually be the form that is active in the
`intestine.
`Interestingly, an N-terminally extended form of guanylin
`(containing an extra aspartate residue) has recently been
`detected in extracts of rat ileum and colon, using an RIA
`directed against
`the proguanylin C-terminus [45]. This
`N-terminally extended peptide accounts for about 50% of
`the total guanylin-like immunoreactivity present
`in the
`colon. We have not detected this peptide in our present
`studies, nor should we have for two reasons: (a) it
`is
`inactive when assayed on T84 cells, and thus would fail to
`show up in our bioassays, and (b) it lacks the proguanylin
`N-terminus, and thus would fail to show up on our Western
`blots.
`The affinity of GCC for uroguanylin (opossum or
`human)
`is about 10-fold higher
`than its affinity for
`guanylin (rat or human) [28,29]. Thus, features that are
`found in uroguanylin, but not
`in guanylin, offer infor-
`mation about structural elements that specify the strength
`of the ligand/receptor interaction. Of particular interest are
`two residues that are basic or uncharged in guanylin but
`acidic in uroguanylin (stippled arrowheads), and one
`residue that contains an aromatic ring in guanylin but an
`acid amide in uroguanylin (solid arrowhead). At all three
`positions, our duodenal peptide follows the consensus
`sequence of uroguanylin rather than that of guanylin, and
`thus we would expect its affinity to be comparable to that
`of opossum or human uroguanylin. Dos

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