throbber
Structure and activity of uroguanylin and guanylin
`from the intestine and urine of rats
`
`XIAOHUI FAN,1,2 F. KENT HAMRA,1,2 ROSLYN M. LONDON,1,2
`SAMMY L. EBER,1,2 WILLIAM J. KRAUSE,2 RONALD H. FREEMAN,2
`CHRISTINE E. SMITH,3 MARK G. CURRIE,3 AND LEONARD R. FORTE1,2
`1
`2
`
`3
`
`Fan, Xiaohui, F. Kent Hamra, Roslyn M. London,
`Sammy L. Eber, William J. Krause, Ronald H. Freeman,
`Christine E. Smith, Mark G. Currie, and Leonard R.
`Forte. Structure and activity of uroguanylin and guanylin
`from the intestine and urine of rats.
`273
`(
`36): E957²E964, 1997.ÑUroguanylin
`and guanylin are related peptides that activate common
`guanylate cyclase signaling molecules in the intestine and
`kidney. Uroguanylin was isolated from urine and duodenum
`but was not detected in extracts from the colon of rats.
`Guanylin was identified in extracts from small and large
`intestine but was not detected in urine. Uroguanylin and
`guanylin have distinct biochemical and chromatographic
`properties that facilitated the separation, purification, and
`identification of these peptides. Northern assays revealed
`that mRNA transcripts for uroguanylin were more abundant
`in small intestine compared with large intestine, whereas
`guanylin mRNA levels were greater in large intestine relative
`to small intestine. Synthetic rat uroguanylin and guanylin
`had similar potencies in the activation of receptors in T84
`intestinal cells. Production of uroguanylin and guanylin in
`the mucosa of duodenum is consistent with the postulate that
`both peptides influence the activity of an intracellular guano-
`sine 3!,5!-cyclic monophosphate signaling pathway that regu-
`lates the transepithelial secretion of chloride and bicarbonate
`in the intestinal epithelium.
`guanylate cyclase; guanosine 3!,5!-cyclic monophosphate; kid-
`ney; heat-stable enterotoxin; human T84 intestinal cells
`
`

`

`E958
`
`STRUCTURE AND ACTIVITY OF UROGUANYLIN AND GUANYLIN
`
`of 10%, followed by 30% and then 60% acetonitrile solutions
`containing 0.1% TFA. The bioactive peptides were eluted with
`the 30% acetonitrile-0.1% TFA solution, and this fraction was
`dried, resuspended in 10% acetonitrile and 0.1% TFA, and
`applied to a C18 semipreparative high-performance liquid
`chromatography (HPLC) column (Waters semipreparative
`€Bondapak, 7.8 mm " 30 cm). The peptides were eluted with
`a gradient of 10% acetonitrile-0.1% TFA to 30% acetonitrile-
`0.1% TFA over a period of 180 min. The peaks of bioactive
`peptides were pooled, dried, resuspended in H2O with 0.8%
`ampholytes [pH range 3²10 (Bio-Rad)], and subjected to
`preparative isoelectric focusing (Rotorfor, Bio-Rad). The frac-
`tions containing bioactivity were combined, applied to a C18
`HPLC column (Waters analytic €Bondapak, 3.9 mm " 30 cm),
`and eluted with a gradient of 5% acetonitrile-10 mM ammo-
`nium acetate (pH 6.2) to 25% acetonitrile-10 mM ammonium
`acetate (pH 6.2) over 180 min. The peak of bioactive peptides
`was subjected to a second purification procedure with the
`same C18 analytic HPLC column, but with the acetonitrile
`gradient containing 0.1% TFA instead of ammonium acetate.
`The bioactive peptides were then applied to a C8 microbore
`column and eluted with a gradient of 0.33% of acetonitrile
`and 0.1% TFA per minute as previously described (4, 14, 15).
`The purified peptides were subjected to automated Edman
`NH2-terminal sequencing, as previously described (4, 14, 15).
`
`The mucosa (100 g wet weight) was scraped from
`colons by use of a glass microscope slide and then boiled in 10
`volumes of 1 M acetic acid for 10 min, homogenized, and
`centrifuged at 10,000
`for 20 min. The supernatant was
`extracted with C18 Sep-Pak cartridges followed by Sephadex
`G-25 column fractionation, as described above. The bioactive
`peptide fractions from the gel column were combined and
`fractionated a second time with C18 Sep-Pak cartridges. The
`peptides were eluted using 5, 10, 15, 20, 25, 40, and 60%
`acetonitrile solutions containing 0.1% TFA. The bioactive
`peptide fractions (i.e., 25% acetonitrile) were pooled and
`subjected to isoelectric focusing as described above. The final
`purification of the active peptides was accomplished by HPLC
`by use of a series of C18 columns as we have described.
`Fifty-five grams wet weight of mucosa were scraped from
`the duodenum (proximal one-third of the small intestine),
`and the bioactive peptides were purified using the same
`methods as described above for the peptides isolated from
`colonic mucosa, except that the isoelectric focusing step was
`not used.
`
`Total
`RNA was prepared (RNeasy kit, Qiagen) from the mucosa of
`individual intestinal segments, and 20 €g of each RNA
`preparation were subjected to electrophoresis in formalde-
`hyde-agarose gels and then transferred to nylon membranes
`(Bio-Rad). The blots were hybridized with rat uroguanylin
`and #-actin cDNAs or rat guanylin plus #-actin cDNAs (27).
`Prehybridization was for 1 h with QuickHyb (Stratagene, La
`Jolla, CA) at 68ÊC, which was followed by hybridization for 2
`h at 68ÊC with each cDNA probe labeled by random priming
`(Boehringer Mannheim). The blots were washed twice with
`2" standard sodium citrate (SSC) and 0.1% sodium dodecyl
`sulfate (SDS) for 15 min at room temperature and once with
`0.2" SSC and 0.1% SDS for 15 min at 60ÊC. The exposure to
`film was for 24 h at !80ÊC with intensifying screens. Rat
`uroguanylin cDNA (nucleotides 117²292) was produced by
`polymerase chain reaction (PCR) amplification from intesti-
`nal mRNA-cDNA (1, 27). This cDNA was isolated and se-
`quenced to confirm that it matched the uroguanylin ex-
`pressed sequence tag (EST) of rat uroguanylin with 100%
`identity. A rat guanylin cDNA (nucleotides 1²531) was gen-
`
`erously provided by Dr. Roger Weigand (Monsanto, St.
`Louis, MO).
`
`T84 cells were obtained from Dr. Jim McRob-
`erts (Harbor-University of California, Los Angeles, CA) at
`passage 21. Cells were cultured in Dulbecco's modified Eagle's
`medium (DMEM) and Ham's F-12 medium (1:1), supple-
`mented with 5% fetal bovine serum, 60 mg penicillin/ml, and
`100 mg streptomycin/ml.
`
`T84 cells were cultured in
`24-well plastic dishes, and cellular cGMP levels were mea-
`sured in control and agonist-stimulated cells by radioimmuno-
`assay (12²15). Briefly, column fractions of the synthetic
`peptides, uroguanylin and guanylin, were suspended in 200
`€l of DMEM containing 20 mM -(2-hydroxyethyl)piperazine-
`!-(2-ethanesulfonic acid) (HEPES), pH 7.4 or 5.5 buffer,
`consisting of DMEM, 20 mM 2-(
`-morpholino)-ethanesul-
`fonic acid (MES, pH 5.5), and 1 mM isobutylmethylxanthine
`(IBMX). The solutions containing bioactive peptides were
`then added to cultured cells and incubated at 37ÊC for 40 min.
`After incubation, the reaction medium was aspirated and 200
`€l of 3.3% perchloric acid were added per well to stop the
`reaction and extract cGMP. The extract was adjusted to pH
`7.0 with KOH and centrifuged, and 50 €l of the supernatant
`were used to measure cGMP.
`
`RESULTS
`
`Uroguanylin was purified from rat urine using C18
`Sep-Pak and gel filtration chromatography, prepara-
`tive isoelectric focusing, and a series of reverse phase
`(RP)-HPLC steps (4, 13²15).
`After the isolation of bioactive peptides with C18
`cartridges, a second chromatographic step with a Sepha-
`dex G-25 column yielded a single peak of peptides that
`stimulated cGMP accumulation in T84 cells (data not
`shown). This peak of bioactive peptides eluted at a
`position identical to that previously found for opossum
`uroguanylin (14, 15). Preparative isoelectric focusing
`separates the more highly acidic uroguanylin from
`guanylin (14, 15). The bioactive peptides eluting from
`Sephadex G-25 columns were subjected to preparative
`isoelectric focusing, and the active peptides migrated to
`the most acidic region, eluting at pH values of 2.4²3.7
`in fractions 1²3 (Fig. 1). This peptide fraction from
`urine stimulated cGMP accumulation in the T84 cells
`to a greater magnitude when the medium pH was 5.5
`compared with the stimulation at pH 7.4. The profile of
`pH dependence for agonist activity in T84 cells is
`consistent with this urine peptide being uroguanylin
`(12). This peptide fraction was then combined and
`subjected to RP-HPLC by use of C18 columns and a
`gradient of 5²25% acetonitrile containing 10 mM ammo-
`nium acetate, pH 6.2 (13²15). Under these RP-HPLC
`conditions, guanylin elutes at 16²18% acetonitrile,
`whereas uroguanylin elutes at 10²11% acetonitrile.
`Fractions 1²3 from the isoelectric focusing purification
`step (Fig. 1) were combined for RP-HPLC under these
`conditions. A single peak of bioactive peptides eluted at
`10% acetonitrile and 10 mM ammonium acetate, an
`elution pattern consistent with this peptide being uro-
`guanylin (Fig. 2). This peak of bioactive peptides was
`purified further using the same C18 column by RP-
`HPLC with an acetonitrile gradient containing 0.1%
`
`Downloaded from journals.physiology.org/journal/ajpendo (165.001.202.250) on January 18, 2022.
`
`

`

`STRUCTURE AND ACTIVITY OF UROGUANYLIN AND GUANYLIN
`
`E959
`
`Fig. 1. Isolation of uroguanylin from rat urine by isoelectric focusing.
`Rat urine was first chromatographed with C18 Sep-Pak cartridges
`followed by gel filtration chromatography with Sephadex G-25, as
`described in MATERIALS AND METHODS. Active fractions from the G-25
`column were pooled, lyophilized, and subjected to isoelectric focusing.
`Fractions were assayed using the T84 cell guanosine 3!,5!-cyclic
`monophosphate (cGMP) accumulation bioassay under conditions of
`MES, DMEM at pH 5.5 (open bars), or HEPES and DMEM at pH 7.4
`(solid bars).
`
`TFA (20, 21). The bioactive peptides were eluted at 21%
`acetonitrile and were combined for microbore RP-
`HPLC (data not shown). After further purification with
`RP-HPLC with a C8 microbore column (Fig. 3), the
`bioactive peptides in the shaded portion of the ultravio-
`let absorbance tracing were combined and subjected to
`NH2-terminal sequence analysis (5, 20). A partial se-
`quence of E/DXXELXINVAXTGX (X is unknown) was
`obtained because of the low quantity of peptides remain-
`ing at this stage of purification. The partial amino acid
`sequence obtained for the rat urine peptide is similar to
`the corresponding residues reported for opossum and
`human forms of bioactive uroguanylin isolated from
`urine (14, 18) and identical to the deduced sequence
`from a uroguanylin EST cDNA isolated from rat intes-
`tine (1). An acidic residue of either glutamate or
`aspartate was observed at the position where gluta-
`mate is found in opossum uroguanylin and where
`aspartate occurs in human uroguanylin (14, 18). The
`amino acids identified by sequence analysis consisting
`of ELXINVAXTGX are identical to the corresponding
`
`Fig. 3. Purification of uroguanylin by RP-HPLC from urine. Ultravio-
`let (UV) absorbance of the last RP-HPLC step using a C8 microbore
`column. Arbitrary units for UV absorbance are used.
`(shaded
`area) contains the bioactive peptides eluted; this fraction was sub-
`jected to sequence analysis. A residue of either glutamate or aspar-
`tate was observed at the first position, and the second position was
`not determined. The other 4 positions marked as X correspond to the
`conserved cysteine residues within this family of peptides. The
`partial amino acid sequence that was obtained is shown at top.
`
`residues found in opossum uroguanylin. Taken to-
`gether, these findings suggest that uroguanylin is the
`major bioactive peptide appearing in the urine of rats.
`
`Isolation of uroguanylin and prouroguanylin from
`colon and small
`intestine and uroguanylin mRNA
`expression in the intestinal mucosa of other species
`suggests that the intestine of rats may be a source of
`uroguanylin in urine (5, 13, 15, 16). To investigate this
`possibility, we isolated bioactive peptides from the
`mucosa of colon and duodenum from rats. Extracts of
`colonic mucosa were prepared and purified by C18
`chromatography, followed by Sephadex G-25 chromatog-
`raphy as described above. A single peak of bioactive
`peptides was observed eluting from the Sephadex G-25
`column (data not shown). The active peptide peak was
`combined and subjected to preparative isoelectric focus-
`ing. The bioactive peptides eluted in fractions 1²3 with
`pH values of 2.6²3.5 (Fig. 4). At this stage of purifica-
`tion, the peptide components from rat colon exhibited a
`property similar to that of guanylin, because the colon
`peptides stimulated cGMP accumulation in T84 cells to
`a greater level at the medium pH of 7.4 compared with
`the cGMP responses at pH 5.5 (12). When the active
`
`Fig. 2. Purification of uroguanylin from rat urine by reverse-phase
`high-performance liquid chromatography (RP-HPLC). Active frac-
`tions from the isoelectric focusing step were combined and subjected
`to RP-HPLC using a C18 analytic column. Peptides were eluted with a
`gradient from 5% acetonitrile containing 10 mM ammonium acetate
`to 25% acetonitrile containing 10 mM ammonium acetate over a
`period of 180 min. Bioassay was conducted with T84 cells in HEPES
`and DMEM at pH 7.4. Bioactive peptides eluted from this column at
`10% acetonitrile.
`
`Fig. 4. Isolation of guanylin from colonic mucosa by isoelectric
`focusing. Extracts of colonic mucosa were chromatographed with C18
`Sep-Pak cartridges and then fractionated on a Sephadex G-25
`column before bioactive peptides were subjected to isoelectric focus-
`ing. Each fraction was assayed with T84 cells in MES, DMEM at pH
`5.5 (open bars), or HEPES and DMEM at pH 7.4 (solid bars).
`
`Downloaded from journals.physiology.org/journal/ajpendo (165.001.202.250) on January 18, 2022.
`
`

`

`E960
`
`STRUCTURE AND ACTIVITY OF UROGUANYLIN AND GUANYLIN
`
`fractions were combined and subjected to RP-HPLC
`with a C18 analytic column, the bioactive peptides
`eluted at 15.5% acetonitrile (Fig. 5). This characteristic
`elution profile for rat guanylin (4) indicates that the
`active peptides isolated from the colonic mucosa of rats
`are predominantly guanylin. This peak of guanylin-like
`peptides was purified further by use of C18 RP-HPLC
`with an acetonitrile gradient containing 0.1% TFA and
`finally by microbore RP-HPLC with a C8 column as
`described above. This peptide fraction was then sub-
`jected to NH2-terminal sequence analysis, and the
`15-residue peptide PNTCEICAYAACTGC was ob-
`tained. This is the same amino acid sequence as that
`obtained when guanylin was originally isolated from
`the jejunum of rats (4).
`The duodenum may produce uroguanylin, because
`the content of guanylin mRNA in duodenum of rats is
`considerably lower than the mRNA levels of colon, and
`the duodenum has substantial cGMP responses to
`these peptides (20, 21, 31). Bioactive peptides were
`isolated from the mucosa of rat duodenum, and two
`separate peaks of peptide bioactivity eluted at different
`positions within the internal volume of Sephadex G-25
`columns (Fig. 6). When these fractions were bioassayed
`using T84 cells, we found that
`stimulated cGMP
`accumulation greater at pH 5.0 than at pH 8.0 (urogua-
`nylin-like) and that
`stimulated cGMP accumula-
`tion greater at pH 8.0 than at pH 5.0 (guanylin-like).
`The very low stimulation of cGMP accumulation ob-
`served for the
`aliquot at pH 8.0 and the
`correspondingly low stimulation for the
`aliquot
`at pH 5.0 may be explained by the relatively low
`concentrations of these peptides in the aliquots from
`the columns that were bioassayed.
`(urogua-
`nylin) was pooled and further purified by C18 RP-HPLC
`by use of a 5²25% acetonitrile gradient containing
`ammonium acetate. The bioactive peptides eluted at
`11% acetonitrile, which is consistent with this peptide
`being uroguanylin (Fig. 7). Moreover, this peptide
`stimulated cGMP accumulation greater at the medium
`pH of 5.0 than at pH 8.0, which is also a property found
`in the uroguanylin peptides. To summarize, the chro-
`
`Fig. 6. Separation of uroguanylin-like and guanylin-like peptides
`from duodenum by gel filtration chromatography. Mucosa from
`duodenum was heated at 100ÊC in 1 M acetic acid; then extracts were
`fractionated with C18 Sep-Pak before application to a Sephadex G-25
`column. Fractions were assayed using the T84 cell cGMP accumula-
`tion bioassay in MES and DMEM at pH 5.0 (dashed line) and in
`HEPES and DMEM adjusted to pH 8.0 with 50 mM sodium bicarbon-
`ate (solid line).
`
`matographic elution profile using RP-HPLC and the
`pH dependency for activation of receptor guanylate
`cyclases (GCs) of this peptide from the duodenum
`mucosa are characteristic properties of uroguanylin.
`An insufficient quantity of the purified uroguanylin-
`like peptide was available for NH2-terminal sequence
`analysis; thus confirmation of these findings by elucida-
`tion of the peptide's sequence was not possible.
`A partial cDNA EST encoding the COOH-terminal
`portion of prouroguanylin was isolated from the duode-
`num of zinc-deficient rats (1). This information facili-
`tated the production of a uroguanylin cDNA by use of
`reverse transcription of RNA from rat duodenum and
`the PCR to amplify this form of uroguanylin cDNA. The
`uroguanylin cDNA was cloned and sequenced to con-
`firm its identity and then used as a cDNA probe in
`Northern assays to assess the relative abundance of
`uroguanylin mRNA compared with guanylin mRNA in
`the intestine. Uroguanylin transcripts of !0.75 kilo-
`
`Fig. 5. Purification of guanylin from colonic mucosa by RP-HPLC.
`Bioactive fractions from the isoelectric focusing step were applied to a
`C18 RP-HPLC analytic column and eluted with acetonitrile/ammo-
`nium acetate, as described in Fig. 2. Fractions were bioassayed using
`T84 cells in HEPES and DMEM adjusted to pH 8.0 with 50 mM
`sodium bicarbonate. Peak of bioactive peptides eluted from this
`column at 15.5% acetonitrile is similar to that of authentic guanylin.
`
`Fig. 7. Isolation of uroguanylin from duodenum by RP-HPLC.
`from the Sephadex G-25 gel filtration column step was combined and
`subjected to C18 semipreparative RP-HPLC and fractionated with a
`gradient of acetonitrile containing 10 mM ammonium acetate, as
`described in Fig. 2. Eluted fractions were assayed by T84 cell cGMP
`stimulation bioassay in MES and DMEM at pH 5.0 (dashed line) and
`in HEPES and DMEM adjusted to pH 8.0 with 50 mM sodium
`bicarbonate (solid line). Major peak of bioactive peptides eluted from
`this column at 11% acetonitrile, consistent with the chromatographic
`properties of uroguanylin.
`
`Downloaded from journals.physiology.org/journal/ajpendo (165.001.202.250) on January 18, 2022.
`
`

`

`STRUCTURE AND ACTIVITY OF UROGUANYLIN AND GUANYLIN
`
`E961
`
`base (kb) were detected throughout the intestinal tract,
`but the highest levels were found in the duodenum and
`jejunum of small intestine (Fig. 8). Lower levels of
`uroguanylin mRNA were observed in ileum and the
`cecum and colon compared with duodenum and jeju-
`num. Guanylin mRNA transcripts of !0.6 kb were
`detected throughout the intestinal tract, with the high-
`est mRNA levels observed in cecum and colon compared
`with the levels in small intestine. The lowest guanylin
`mRNA levels were found in the duodenum relative to
`other segments of intestine. Progressively greater lev-
`els of guanylin mRNA were found along the longitudi-
`nal axis of the small intestine from duodenum to ileum,
`with the greatest mRNA levels observed in the cecum
`and colon.
`Analysis of the EST for uroguanylin derived from rat
`intestine (1) confirmed that the partial amino acid
`sequence obtained for the urinary peptide was consis-
`tent with the sequence predicted by the uroguanylin
`EST. Thus a synthetic peptide was prepared on the
`basis of the amino acid sequence TDECELCINVAC-
`TGC, and the potency of this peptide was compared
`with the potencies of rat guanylin and a truncated form
`of uroguanylin, CELCINVACTGC, by use of the T84
`cell bioassay (4, 14). Uroguanylin and guanylin had
`similar potencies in the activation of receptor GCs in
`T84 cells, but the truncated form of uroguanylin was
`
`substantially less potent (Fig. 9). These data indicate
`that the NH2-terminal residues found in the bioactive
`uroguanylin peptide consisting of TDE increase the
`potencies of this peptide agonist for activation of recep-
`tor GCs on T84 cells compared with the potency of the
`truncated 12-residue form of uroguanylin. However,
`the 12 amino acids in the truncated uroguanylin analog
`containing the peptide domain between the first and
`last cysteine residues with two intramolecular disulfide
`bonds represent a core structure that is required for
`biological activity in this assay.
`
`DISCUSSION
`
`Uroguanylin was isolated from both urine and duode-
`num mucosa of rats and identified by its unique bio-
`chemical and pharmacological properties (12²15). Uro-
`guanylin is present in the urine of rats, as it is in the
`urine of the opossum and human species (14, 18).
`Guanylin was isolated from mucosa of both the duode-
`num and large intestine, but active guanylin peptides
`were not detected in urine. Sequence analysis of urogua-
`nylin from rat urine revealed that the eight residues
`obtained were identical to those found in opossum
`uroguanylin. One of the two NH2-terminal acidic amino
`acids unique to uroguanylin was not clearly defined
`(Glu or Asp), and the other acidic amino acid was not
`
`Fig. 8. Distribution of uroguanylin and gua-
`nylin mRNA in the intestine. Total RNA of 20 €g
`from mucosa of rat proximal small intestine
`(Prox. SI), middle small intestine (Mid. SI),
`distal small intestine (Dist. SI), cecum, and
`colon were loaded on each lane.
`: arrows mark
`single transcripts for #-actin mRNA of 1.9 kilo-
`base (kb) and uroguanylin mRNA of 0.75 kb;
`:
`arrows indicate single transcripts for #-actin
`mRNA of 1.9 kb and guanylin mRNA of 0.6 kb.
`
`Downloaded from journals.physiology.org/journal/ajpendo (165.001.202.250) on January 18, 2022.
`
`

`

`E962
`
`STRUCTURE AND ACTIVITY OF UROGUANYLIN AND GUANYLIN
`
`sodium diets downregulate the guanylin receptor-GC
`signaling pathway in the rat colon suggests that the
`guanylin signaling pathway may participate in the
`maintenance of salt and water homeostasis (24).
`In rats, guanylin mRNA levels appear to be most
`abundant in colon and ileum, with intermediate mRNA
`levels in jejunum and the lowest mRNA levels in
`duodenum (25, 27, 31). In the present experiments,
`bioactive guanylin was isolated from colonic mucosa
`but bioactive uroguanylin was not detected. This find-
`ing is consistent with the high levels of guanylin mRNA
`that were detected using Northern assays with total
`RNA from colon and cecum in this study compared with
`the lower uroguanylin mRNA levels of large intestine.
`The lower abundance of uroguanylin mRNAs in the
`colon and cecum provides one explanation for our
`inability to detect bioactive uroguanylin in extracts of
`large intestine. Isolation of uroguanylin and guanylin
`from duodenum in this study suggests that both pep-
`tides are present and may regulate the activity of
`receptor-GC signaling molecules in this segment.
`Whereas Northern assays suggest that uroguanylin
`mRNA expression is greater than guanylin mRNA
`expression in the duodenum, both peptides are present
`in the mucosa of the duodenum in concentrations
`sufficient for purification and identification of this
`bioactive peptide (25, 27, 29). Other studies have also
`found a similar pattern of expression of guanylin and
`uroguanylin mRNA levels along the longitudinal axis of
`the intestinal tract of rats (25, 27, 29).
`Uroguanylin and guanylin markedly stimulate the
`transepithelial secretion of both Cl! and HCO3! anions
`
`in the duodenum (11, 17). Exposure of the apical
`surface of duodenum to these peptides elicits a stimula-
`tion of short-circuit current consisting of both Cl! and
`HCO3! transport components. Both peptides may be
`
`released from enterocytes into the luminal microdo-
`main at the surface of this epithelium where binding of
`the peptides to receptor-GCs occurs, thus activating
`these signaling molecules and regulating anion secre-
`tion via intracellular cGMP. Because the potency of
`uroguanylin is markedly enhanced when the intralumi-
`nal pH is acidic and an acidic pH markedly decreases
`the potency of guanylin, it may be postulated that the
`secretion of uroguanylin is increased when acidic chyme
`is delivered from the stomach to the duodenum (12, 13,
`15). The relative potencies of guanylin and uroguanylin
`for activation of intestinal receptor GCs and the stimu-
`lation of transepithelial Cl! secretion are markedly
`influenced by mucosal acidity (12). In the present study,
`uroguanylin isolated from rat duodenum (or urine)
`stimulates cGMP accumulation in T84 cells to a greater
`magnitude at a medium pH of 5.5 than at pH 7.5.
`Guanylin isolated from the intestine increased cGMP
`to a greater level in T84 cells at pH 7.5 than at pH 5.5.
`Thus uroguanylin and guanylin isolated from rat intes-
`tine exhibit properties similar to those previously de-
`fined for the homologous peptides derived from human
`subjects and opossums (12, 13, 15). Evolution of the
`unique primary structures for uroguanylin and gua-
`nylin may have occurred to allow different peptide
`
`Fig. 9. Bioactivity of synthetic uroguanylin and guanylin in T84
`cells. Values are representative of 3 experiments conducted with
`cultured T84 cells and are means of duplicate assays at each peptide
`concentration. !, Rat guanylin (PNTCEICAYAACTGC); ", rat uro-
`guanylin (TDECELCINVACTGC); #, 12-residue portion of urogua-
`nylin (CELCINVACTGC). Disulfide bonds in these synthetic peptides
`occur between 1st to 3rd and 2nd to 4th cysteine residues. Medium is
`DMEM at pH 7.4 for this assay.
`
`determined. The amino acid sequence of rat urogua-
`nylin has been recently elucidated by the isolation of
`cDNA clones encoding preprouroguanylin and by purifi-
`cation of uroguanylin from duodenum and NH2-
`terminal sequence analysis (1, 26, 29). These studies
`revealed that the sequence of the 15 COOH-terminal
`residues for rat uroguanylin is TDECELCINVACTGC,
`which agrees with the partial sequence that we ob-
`tained in the present study. A synthetic peptide pre-
`pared according to this sequence activated the T84 cell
`receptor GC with potency and efficacy similar to the
`activation elicited by synthetic rat guanylin.
`The reason bioactive guanylin is not found in rat or
`human urine is unclear, but it may be the susceptibility
`of guanylin in the tubular filtrate to cleavage and
`inactivation by proteases within renal tubules. Gua-
`nylin is inactivated by chymotrypsin, which cleaves the
`peptide bond COOH terminal to the aromatic residues
`of guanylin peptides (13, 15). In contrast, uroguanylin
`and
`ST are resistant to chymotrypsin
`because these peptides have asparagine residues in-
`stead of tyrosine or phenylalanine (13²15, 18). Plasma
`uroguanylin and guanylin may enter the tubular fil-
`trate by glomerular filtration (5, 19). Also, recent
`evidence suggests that the kidney produces urogua-
`nylin, because mRNAs encoding this peptide are ex-
`pressed in the kidney (29). The presence of bioactive
`uroguanylin in urine is significant because renal recep-
`tors for uroguanylin are localized on the apical mem-
`branes of proximal tubular cells (9, 10, 21). Circulating
`uroguanylin enters the tubules by glomerular filtration
`to gain access to these receptors. Activation of the
`receptor GCs in the perfused rat kidney elicits a
`natriuresis, kaliuresis, and diuresis (unpublished obser-
`vations). Thus uroguanylin may serve in an endocrine
`pathway to regulate renal function in vivo (unpub-
`lished observations; 8²10). Recent evidence that low-
`
`Downloaded from journals.physiology.org/journal/ajpendo (165.001.202.250) on January 18, 2022.
`
`

`

`STRUCTURE AND ACTIVITY OF UROGUANYLIN AND GUANYLIN
`
`E963
`
`hormones that function cooperatively to regulate intes-
`tinal Cl! and HCO3! secretion during digestion. The
`
`lumen of the intestine and the mucosal (microclimate)
`surface is acidified when chyme containing HCl enters
`the duodenum. Under this condition, uroguanylin may
`be a more effective agonist for regulating receptor-GC
`activity than is guanylin. When the mucosal surface of
`the duodenum becomes alkalinized through enhanced
`HCO3! secretion, the affinity of guanylin for binding to
`
`receptor GCs would be increased, thus facilitating the
`binding of guanylin to receptors and activation of these
`signaling molecules (12, 13, 15). Thus uroguanylin and
`guanylin may participate in a cGMP signaling pathway
`controlling intestinal Cl! and HCO3! secretion (4, 6, 11,
`
`14, 17).
`In summary, uroguanylin was isolated from urine
`and the duodenum of rats. Bioactive guanylin was not
`detected in urine, but this peptide was isolated from
`colon and duodenum. Uroguanylin mRNA levels were
`greater in small than in large intestine, whereas the
`levels of guanylin mRNA transcripts were greater in
`large than in small intestine. Synthetic rat urogua-
`nylin was approximately equipotent to rat guanylin in
`the activation of cGMP production in T84 intestinal
`cells when assessed at the medium pH of 7.4. Urogua-
`nylin and guanylin may both participate in the regula-
`tion of Cl! and HCO3! secretion via the intracellular
`
`second messenger cGMP.
`Address for reprint requests: L. R. Forte, Dept. of Pharmacology,
`M-515 Medical Sciences Bldg., School of Medicine, Univ. of Missouri,
`Columbia, MO 65212.
`Received 14 March 1997; accepted in final form 30 July 1997.
`
`REFERENCES
`1. Blanchard, R. K., and R. J. Cousins. Differential display of
`intestinal mRNAs regulated by dietary zinc.
`93: 6863²6868, 1996.
`2. Cetin, Y., M. Kuhn, H. Kulaksiz, K. Adermann, G. Bargsten,
`D. Grube, and W.-G. Forssmann. Enterochromaffin cells of the
`digestive system: cellular source of guanylin, a guanylate cyclase-
`activating peptide.
`91: 2935²2939,
`1994.
`3. Cohen, M. B., D. P. Witte, J. A. Hawkins, and M. G. Currie.
`Immunohistochemical localization of guanylin in the rat small
`intestine and colon.
`209:
`803²808, 1995.
`4. Currie, M. G., K. F. Fok, J. Kato, R. J. Moore, F. K. Hamra,
`K. L. Duffin, and C. E. Smith. Guanylin: an endogenous
`activator of intestinal guanylate cyclase.
`89: 947²951, 1992.
`5. Fan, X., F. K. Hamra, R. H. Freeman, S. L. Eber, W. J.
`Krause, R. W. Lim, V. M. Pace, M. G. Currie, and L. R. Forte.
`Uroguanylin: cloning of preprouroguanylin cDNA, mRNA expres-
`sion in the intestine and heart and isolation of uroguanylin and
`prouroguanylin from plasma.
`219: 457²462, 1996.
`6. Field, M., L. H. Graf, W. J. Laird, and P. L. Smith. Heat-stable
`enterotoxin of
`: in vivo effects on guanylate
`cyclase activity, cyclic GMP concentration, and ion transport in
`small intestine.
`75: 2800²2804, 1978.
`8. Forte, L. R., and F. K. Hamra. Guanylin and uroguanylin:
`intestinal peptide hormones that regulate epithelial transport.
`11: 17²24, 1996.
`9. Forte, L. R., W. J. Krause, and R. H. Freeman. Receptors and
`cGMP signalling mechanism for
`enterotoxin in opossum
`kidney.
`255 (
`):
`F1040²F1046, 1988.
`
`10. Forte, L. R., W. J. Krause, and R. H. Freeman.
`enterotoxin receptors: localization in opossum kidney, intes-
`tine and testis.
`257 (
`26): F874²F881, 1989.
`11. Guba, M., M. Kuhn, W.-G. Forssmann, M. Classen, M.
`Gregor, and U. Seidler. Guanylin strongly stimulates rat
`duodenal HCO3! secretion: proposed mechanism and comparison
`
`with other secretagogues.
`111: 1558²1568, 1996.
`12. Hamra, F. K., S. L. Eber, D. T. Chin, M. G. Currie, and L. R.
`Forte. Regulation of intestinal uroguanylin/guanylin receptor-
`mediated responses by mucosal acidity.
`94: 2705²2710, 1997.
`13. Hamra, F. K., X. Fan, W. J. Krause, R. H. Freeman, D. T.
`Chin, C. E. Smith, M. G. Currie, and L. R. Forte. Prourogua-
`nylin and proguanylin: purification from colon, structure, and
`modulation of bioactivity by proteases.
`137: 257²
`265, 1996.
`14. Hamra, F. K., L. R. Forte, S. L. Eber, N. V. Pidhorodeckyj,
`W. J. Krause, R. H. Freeman, D. T. Chin, J. A. Tompkins,
`K. F. Fok, C. E. Smith, K. L. Duffin, N. R. Siegel, and M. G.
`Currie. Uroguanylin: structure and activity of a second endog-
`enous peptide that stimulates intestinal guanylate cyclase.
`90: 10464²10468, 1993.
`15. Hamra, F. K., W. J. Krause, S. L. Eber, R. H. Freeman, C. E.
`Smith, M. G. Currie, and L. R. Forte. Opossum colonic mucosa
`contains uroguanylin and guanylin peptides.
`270
`(
`33): G708²G716, 1996.
`16. Hill, O., Y. Cetin, A. Cieslak, H.-J. Magert, and W.-G.
`Forssmann. A new human guanylate cyclase-activating peptide
`(uroguanylin): precursor cDNA and colonic expression.
`1253: 146²149, 1995.
`17. Joo, N. S., H. D. Kim, L. R. Forte, and L. L. Clarke.
`Uroguanylin regulation of anion secretion in mouse intestine
`(Abstract).
`11: A297, 1997.
`18. Kita, T., C. E. Smith, K. F. Fok, K. L. Duffin, W. M. Moore,
`P. J. Karabatsos, J. F. Kachur, F. K. Hamra, N. V. Pidhoro-
`deckyj, L. R. Forte, and M. G. Currie. Characterization of
`human uroguanylin: a member of the guanylin peptide family.
`266 (
`35): F342²
`
`F348, 1994.
`19. Kuhn, M., M. Raida, K. Adermann, P. Schulz-Knappe, R.
`Gerzer, J.-M. Heim, and W.-G. Forssmann. The circulating
`bioactive form of hu

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