throbber
rapid communication
`
`Characterization of human uroguanylin: a member
`of the guanylin peptide family
`
`TOSHIHIRO KITA, CHRISTINE E. SMITH, KAM F. FOK, KEVIN L. DUFFIN,
`WILLIAM M. MOORE, PETER J. KARABATSOS, JAMES F. KACHUR, F. KENT HAMRA,
`NYKOLAI V. PIDHORODECKYJ, LEONARD R. FORTE, AND MARK G. CURRIE
`Monsanto Corporate Research, Monsanto, St. Louis 63167; Department of Pharmacology,
`Missouri University, and Truman Veterans Affairs Hospital, Columbia, Missouri 65212;
`and Department of Inflammatory Disease Research, G. D. Searle, Skokie, Illinois 60077
`
`Kita, Toshihiro, Christine E. Smith, Kam F. Fok,
`Kevin L. Duffin, William M. Moore, Peter J. Karabatsos,
`James F. Kachur, F. Kent Hamra, Nykolai V. Pidhoro(cid:173)
`deckyj, Leonard R. Forte, and Mark G. Currie. Character(cid:173)
`ization of human uroguanylin: a member of the guanylin
`peptide family. Am. J. Physiol. 266 (Renal Fluid Electrolyte
`Physiol. 35): F342-F348, 1994.-Guanylin, a peptide homo(cid:173)
`logue of the bacterial heat-stable enterotoxins (ST), is an
`endogenous activator of guanylate cyclase C (GC-C). We have
`initiated a search for other members of the guanylin peptide
`family and in the current study describe a "guanylin-like
`peptide" from human urine. Bioactivity was monitored by
`determining the effect of urine extracts on T84 cell guanosine
`3' ,5' -cyclic monophosphate (cGMP) levels. Purification yielded
`two bioactive peaks of peptides that, when sequenced by
`NHrterminal analysis, possessed 15 and 16 amino acids. The
`sequence of the smaller peptide represented an NH2-terminal
`truncation of the larger peptide. We have termed the larger
`peptide human uroguanylin; it has the following amino acid
`sequence: NDDCELCVNVACTGCL. Human uroguanylin
`shares amino acid sequence homology with guanylin and ST.
`Synthetic uroguanylin increased cGMP levels in T84 cells,
`competed with 1251-labeled ST for receptors, and stimulated
`Cl- secretion as reflected by an increased short-circuit current.
`Thus we report the isolation from human urine of a unique
`peptide, uroguanylin, that behaves in a manner similar to
`guanylin and appears to be a new member of this peptide
`family.
`guanosine 3' ,5' -cyclic monophosphate; guanylate cyclase; chlo(cid:173)
`ride secretion; T84 cell; rat colon
`
`THE MEMBRANE-BOUND FORMS of guanylate cyclase have
`been found to serve as receptors for specific peptide
`ligands and are thought to participate in the regulation
`of blood pressure as well as water and electrolyte
`homeostasis (8, 22). These receptors share several struc(cid:173)
`tural motifs, including an extracellular ligand-binding
`site, a transmembrane region, an intracellular ATP
`regulatory domain, and an intracellular catalytic site for
`the generation ofguanosine 3',5'-cyclic monophosphate
`(cGMP) (3, 8). The natriuretic peptide family of recep(cid:173)
`tors with guanylate cyclase activity has been particu(cid:173)
`larly well characterized. These receptors include guanyl(cid:173)
`ate cyclases A and B (GC-A and GC-B, respectively), also
`
`known as natriuretic peptide receptors A and B (8, 22).
`The two subtypes of natriuretic peptide receptor show
`different tissue distributions and exhibit selective affini(cid:173)
`ties for the three different members of the natriuretic
`peptide family (18). GC-A has been shown to be quite
`sensitive to stimulation by atrial natriuretic peptide
`(ANP) and brain natriuretic peptide (BNP) but is very
`insensitive to stimulation by C-type natriuretic peptide
`(CNP) (8, 13, 18). On the other hand, GC-B is potently
`stimulated by CNP and only slightly responsive to either
`ANP or BNP (13).
`An additional form of membrane guanylate cyclase is
`GC-C (intestinal guanylate cyclase), which is an appar(cid:173)
`ent receptor for guanylin and bacterial heat-stable en(cid:173)
`terotoxins (ST) (2, 3, 17). This receptor is abundant in
`intestinal epithelial cells, where it serves as a target for
`ST to cause secretory diarrhea (16, 17). Recently, gua(cid:173)
`nylin, a peptide composed of 15 amino acids, has been
`identified as an endogenous ligand for GC-C (2, 23, 24).
`Guanylin was initially purified from rat jejunum and
`proposed as a modulator of intestinal fluid and electro(cid:173)
`lyte homeostasis (2). Subsequent cloning of a guanylin
`cDNA from rat (16, 23) and human (4, 24) intestinal
`cDNA libraries has determined that this peptide is
`synthesized as part of an 115-amino acid precursor. The
`COOR-terminal region is highly conserved between
`human, rat, and mouse species, and it is this portion of
`the prohormone that contains the guanylin peptide
`sequence (12). In these studies, it was found that
`proguanylin was relatively inactive and required the
`cleavage of the COOR-terminal bioactive region (4, 16).
`Guanylin mRNA, as has been previously described for
`GC-C mRNA, is most abundant in the intestinal tract,
`with the ileum and colon possessing the greatest level of
`expression (12, 23, 24). However, guanylin expression
`has also been detected in extra-intestinal sites, including
`the kidney, uterus, and adrenal gland (16).
`Because of the similarities of the natriuretic peptide
`guanylate cyclase system with the guanylin GC-C sys(cid:173)
`tem (8), we have initiated a search for guanylin and
`potential members of this peptide family in sites other
`than the intestine. Previous evidence suggested that the
`kidney may be a fruitful site for such a search. Rat
`kidney has a modest amount of guanylin-like bioactivity
`
`F342
`
`0363-6127 /94 $3.00 Copyright
`
`1994 the American Physiological Society
`
`Bausch Health Ireland Exhibit 2065, Page 1 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`BIOLOGICAL AND STRUCTURAL CHARACTERIZATION OF HUMAN UROGUANYLIN
`
`F343
`
`on T84 cell cGMP levels (2) and detectable levels of
`guanylin mRNA (16). ST binding sites, which have also
`been reported to be localized in the proximal tubules of
`opossum kidney and ST, caused large increases in cGMP
`levels in opossum renal tissue (6). Thus we hypothesized
`that endogenous ligand(s) for GC-C may be produced by
`the kidney and excreted in the urine. Efforts were
`initiated to isolate endogenous peptide activators of
`GC-C from both human and opossum urine. The find(cid:173)
`ings with the opossum are the subject of a separate
`manuscript (10). In the current study, we report the
`isolation and characterization of a novel 16-amino acid
`peptide from human urine with biological activity and
`structural similarities to guanylin. However, it is dis(cid:173)
`tinct from guanylin, and we contend that uroguanylin
`represents a new member of the guanylin family. We
`have designated this peptide as uroguanylin because
`urine was the source and because of its sequence
`similarity with guanylin, including the four conserved
`cysteine residues.
`
`MATERIALS AND METHODS
`
`Cell culture. T84 cells were obtained from the American
`Type Culture Collection at passage 52. Cells were grown to
`confluence in 24-well culture plates with a 1:1 mixture of
`Ham's F-12 medium and Dulbecco's modified Eagle's medium
`(DMEM) supplemented with 10% fetal bovine serum, 100 U
`penicillin/ml, and 100 µg streptomycin/ml. Cells were used at
`passages 56-58.
`cGMP determination. Confluent monolayers of T84 cells in
`24-well plates were washed twice with 250 µl of DMEM
`containing 50 mM N-2-hydroxyethylpiperazine-N' -2-ethanesul(cid:173)
`fonic acid (HEPES) (pH 7.4), preincubated at 37°C for 10 min
`with 250 µl of DMEM containing 50 mM HEPES and 1 mM
`isobutylmethylxanthine, and incubated with agents or frac(cid:173)
`tions for 30 min. The medium was aspirated, and the reaction
`was terminated by the addition of 0.5 ml of ice-cold 0.1 M HCl.
`Aliquots (150 µl total) were evaporated under a hot-air dryer
`and resuspended in 5 mM sodium acetate buffer, pH 6.4. The
`samples were subsequently measured for cGMP by radioimmu(cid:173)
`noassay as described previously (19).
`Purification of uroguanylin. Five separate batches of adult
`male human urine samples, 5 1 each, were collected and
`immediately placed on ice. The urine samples were applied to
`C18 Sep-Pak columns (Waters). The columns were washed
`with 10% acetonitrile/0.1 % trifluoracetic acid (TFA)/H20 and
`eluted with 40% acetonitrile/0.1 % TFA/H20. The eluted
`peptide fraction was lyophilized, resuspended in 7 ml of
`distilled H20, and centrifuged at 20,000 g for 20 min at 4°C.
`The resulting supernatant was separated by gel filtration
`chromatography (Sephadex G-25, superfine, 2.6 x 94 cm). The
`fractions were bioassayed, and the active fraction was lyophi(cid:173)
`lized. The sample was resuspended in 1 ml of 10% acetonitrile/
`0.1% TFA/H20 and applied to a C18 semipreparative high(cid:173)
`performance liquid chromatography (HPLC) column (Vydac,
`Hesperia, CA). The column was developed with the following
`linear gradient: 10% acetonitrile/0.1 % TFA/H20 to 40% aceto(cid:173)
`nitrile/0.1 % TFA/1120 in 120 min at a flow rate of 3 ml/min.
`The active fraction was lyophilized and resuspended in 1 ml of
`10% acetonitrile/0.1 % TFA/H20. The sample was applied to a
`C18 analytical HPLC column (Vydac), and active peptides were
`eluted using the above gradient over 180 min at a flow rate of 1
`ml/min. Two active fractions were separately lyophilized and
`reconstituted in 0.05 ml of 0.1 % TFA 1120. The samples were
`then separately applied to a C8 microbore column (Applied
`
`Biosystems, Foster City, CA) eluted with an increasing gradi(cid:173)
`ent of 0.33% acetonitrile/min in 0.1% TFA/H20. Two sepa(cid:173)
`rately purified peptides of each batch were then subjected to
`sequence analyses.
`NHTterminal protein sequence analysis. Automated Edman
`degradation chemistry was used to determine the NHr
`terminal protein sequence. An applied Biosystems model 4 70A
`gas-phase sequencer was employed for the degradations (11)
`using the standard sequencer cycle 03RPTH. The respective
`phenylthiohydantoin (PTH)-amino acid derivatives were iden(cid:173)
`tified by reverse-phase HPLC analysis in an on-line fashion
`employing an Applied Biosystems model 120A PTH analyzer
`fitted with a Brownlee PTH-C18 column. Reduction and pyri(cid:173)
`dylethylation for cysteine residue identification were per(cid:173)
`formed directly on the filter.
`Electrospray mass spectrometry. The molecular weights of
`uroguanylin samples were determined by a previously de(cid:173)
`scribed technique (2). Briefly, a triple quadrupole mass spec(cid:173)
`trometer equipped with an atmospheric pressure ion source
`was used to sample positive ions produced from an electro(cid:173)
`spray interface. Mass analysis of sample ions was accom(cid:173)
`plished by scanning the first quadrupole in increments of 1
`atomic mass unit from 1,000 to 2,400 atomic mass units in "'3
`s and passing mass-selected ions through the second and third
`quadrupoles operated in the radio frequency-only mode to the
`multiplier. For maximum sensitivity, the mass resolution of
`the quadrupole mass analyzer was set so that ion signals were
`z 2 atomic mass units wide at one-half peak height but the
`centroid of the ion signal still represented the correct mass of
`the ion.
`Radioligand binding assay. 1251-labclcd ST-(5-18) was
`prepared by the lodo-Gen method as previously described (7).
`T84 cell monolayers were washed two times with 1.0 ml/well
`of ice-cold binding assay buffer (Earle's medium containing 25
`mM 2-N-morpholino ethanesulfonic acid, pH 5.5). The cells
`were incubated for 30 min at 37°C in 0.5 ml/well of binding
`assay buffer with 1251-ST [100,000 counts/min (cpm) per wellJ
`and unlabeled peptides. Then the cells were washed four times
`with 1 ml of ice-cold binding assay buffer and solubilized with
`0.5 ml of 1 M NaOH per well. This volume was transferred to
`tubes and assayed for radioactivity by a multigamma counter.
`Results are expressed as the percentage of cells specifically
`bound.
`Measurement of Isc in T84 monolayers. Analysis of the effect
`of human uroguanylin on Isc in T84 cells was performed as
`previously described (5). Briefly, T84 cells raised on permeable
`filters were mounted in a custom-made Ussing chamber for
`measurement of Cl secretion. The buffer was a Krebs-Ringer
`bicarbonate solution, pH 7.4, containing 10 mM glucose. Both
`reservoir buffer solutions were mixed and oxygenated by
`bubbling 95% Or5% CO2 through the medium. Isc was mea(cid:173)
`sured continuously, and the potential difference across the
`epithelium was measured intermittently.
`Measurement of Isc in rat colon. Rat proximal colon tissue,
`consisting of only mucosa and submucosa, was mounted
`between two U ssing half-chambers and bathed on both sides in
`a manner similar to that previously reported (21). Electrical
`measurements were monitored with an automatic voltage
`clamp, and direct-connecting voltage- and current-passing
`electrodes were used to measure transepithelial potential
`difference and Isc· Tissues were equilibrated under short(cid:173)
`circuit conditions until J50 had stabilized.
`Chemical synthesis of uroguanylin. Uroguanylin was synthe(cid:173)
`sized by the solid-phase method (20) on an Applied Biosystems
`model 430A peptide synthesizer and purified by reverse-phase
`C18 chromatography. The purity and the structure were
`
`Bausch Health Ireland Exhibit 2065, Page 2 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`F344
`
`BIOLOGICAL AND STRUCTURAL CHARACTERIZATION OF HUMAN UROGUANYLIN
`
`Fig. 1. Purification ofuroguanylin from human urine by
`gel filtration chromatography. Extract of 5 I of human
`urine was applied to a 2.6 x 94-cm Sephadex G-25
`(superfine) gel filtration column. Isocratic 50 mM ammo(cid:173)
`nium acetate was used to elute peptides at a rate of 0.5
`ml/min, and 5 ml of fractions were collected after 100 ml
`of initial elution. Molecular weight standards were sepa(cid:173)
`rately assessed lvoid volume (V0 ): blue dextran 200,
`insulin (mo! wt 5,750), atriopeptin III (APIII; mo! wt
`2,550), rat guanylin (mo! wt 1,516)]. All fractions were
`assessed in T84 cell cGMP accumulation bioassay.
`
`1000 ~
`
`Q)
`
`~
`0
`
`E -._..
`
`800 ~
`
`600 ~
`
`400 •
`
`200 •
`
`0
`
`insulin
`
`APIII
`
`guanylin
`
`Vo
`
`.J.
`
`,1,11.,■ulld1JI
`
`I
`
`20
`
`I 11,1 l,1,l,■1■,■1■ 1,l 1,1 ■,I I ■1111,11
`.
`.
`
`I
`
`30
`
`40
`
`50
`
`60
`
`Fraction number
`
`verified by analytical HPLC, amino acid composition analysis,
`mass spectroscopy, and sequence analysis.
`
`RESULTS
`
`In initial experiments, the peptide fraction of human
`urine samples resulting from C18 Sep-Pak extraction
`was assayed for activity to increase cGMP levels in T84
`cells. These preliminary experiments strongly suggested
`the presence of GC-C stimulatory activity. The urine
`extract was subjected to fractionation by gel filtration
`and a series of reverse-phase HPLC steps to produce a
`sufficiently pure preparation for the purpose of struc(cid:173)
`tural determination. Fractionation by G-25 gel filtration
`chromatography yielded a single major bioactive frac(cid:173)
`tion that migrated on the column with an apparent size
`of 5,000 Da (Fig. 1). Subsequently, this active fraction
`was further purified by reverse-phase HPLC using a
`semipreparative C18 column, and the bioactivity was
`determined to reside in only one fraction eluting at
`27.8% acetonitrile/0.1% TFA/H20 (data not shown).
`Further purification by reverse-phase HPLC using a C18
`analytical column yielded two active fractions that ap(cid:173)
`peared to elute with peaks of substances that absorbed
`at 220 nm (Fig. 2). These two fractions were separately
`subjected to further characterization by microbore HPLC
`(C8 column), and each fraction exhibited a single bioac(cid:173)
`tive peak that absorbed at 220 nm (data not shown). The
`amino acid sequences of the two peaks were indepen(cid:173)
`dently determined by the Edman degradation proce(cid:173)
`dure. The sequences are NDDCELCVNV ACTGCL and
`DDCELCVNVACTGCL for peaks 1 and 2, respectively.
`These two peptides are identical except that the peptide
`contained in peak 1 possesses an additional amino acid
`(asparagine) at the NHrterminus. It is likely that peak 2
`is a degradation product of peak 1, probably a result of
`aminopeptidase action. Electrospray mass spectromet(cid:173)
`ric analysis of the two fractions yielded observed molecu(cid:173)
`lar masses of 1,666.6 and 1,552.6 atomic mass units,
`
`respectively, for the peptides contained in peaks 1 and 2.
`These molecular weights correspond to the theoretical
`molecular weights derived from the sequences if two
`disulfide bonds link the four cysteines, and therefore
`indicate that the full sequences of these peptides were
`determined by NHrterminal protein sequence analysis.
`Comparison of the sequence of peak 1 with other
`proteins in the GenBank, National Biomedical Research
`Foundation, and SwissProt databases by computer(cid:173)
`based search indicates that this sequence is a unique
`sequence. This search did reveal that human urogua(cid:173)
`nylin shares homology with guanylin and ST. The
`comparison between human uroguanylin, opossum uro(cid:173)
`guanylin, Escherichia coli ST, and human guanylin is
`shown in Fig. 3. The comparisons indicate that the four
`cysteine residues and the ACTGC COOR-terminal amino
`acid region of all four peptides are conserved. A unique
`feature of uroguanylin is the more acidic nature of the
`NHrterminal region, because the human peptide pos(cid:173)
`sesses two adjacent aspartic acid residues and the
`opossum peptide has a glutamate and an aspartate
`residue at these positions. Thus human uroguanylin
`appears to be a member of the guanylin/ST family of
`peptides.
`Chemical synthesis of bioactive human uroguanylin
`(the 16-amino acid-containing peptide) was accom(cid:173)
`plished by directed folding of the peptide. The synthetic
`bioactive peptide possesses disulfide-linked bridges be(cid:173)
`tween the 4-12 and 7-15 amino acid positions as
`previously suggested for the disulfide links of guanylin
`(2, 24). Analysis of the biological activity of human
`uroguanylin was assessed by determining its effect on
`T84 cGMP levels, competition-binding studies with
`1251-ST as the radioligand in T84 cells, and stimulation
`of Cl- secretion as reflected by increases in lsc using T84
`cells and rat colon.
`Synthetic human uroguanylin caused a concentration(cid:173)
`dependent increase in T84 cell cGMP (Fig. 4A). Human
`
`Bausch Health Ireland Exhibit 2065, Page 3 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`BIOLOGICAL AND STRUCTURAL CHARACTERIZATION OF HUMAN UROGUANYLIN
`
`F345
`
`--------------
`
`---------
`
`-------------------------
`
`--------
`
`~ ' '
`30 ..!.-
`z
`25 U
`M :c
`20 u
`~ 0
`
`Fig. 2. Purification ofuroguanylin from human urine by
`reverse-phase high-performance liquid chromatography
`(HPLC). Five liters of human urine extract was purified
`through the semipreparative reverse-phase HPLC, and
`active fraction was fractionated on an analytical C1s
`column (Vydac). A linear gradient of 10-40% acetoni(cid:173)
`trile, 0.1% TFA was developed at 1.0 ml/min over 3 h.
`One-minute fractions were collected and assayed for
`activity in T84 cell cGMP bioassay. This figure shows the
`biologically active region, with two peaks associated with
`changes in ultraviolet absorbance.
`
`0.04
`
`0.03
`
`0.02
`
`0.01
`
`0
`
`E
`s:::
`
`0
`N
`N
`i;
`
`Q)
`(J
`s:::
`111 .c
`...
`0
`Ul .c
`<
`
`Q)
`
`15000
`~
`0
`10000
`
`E --
`
`C.
`:E
`(!)
`u
`
`5000
`
`0
`
`90
`
`100
`
`110
`
`120
`
`130
`
`Time
`
`(min)
`
`uroguanylin appeared to be more potent than human
`guanylin but less potent than ST for activation of GC-C
`in T84 cells. A similar profile of relative affinity was
`obtained using the competitive binding assay with 1251-
`ST-(5-18) as the radioligand (Fig. 4B), but it should be
`noted that all of the peptides acted in a more potent
`manner in causing 1251-ST-(5-18) displacement than in
`increasing cGMP levels. This difference in potencies for
`receptor binding and guanylate cyclase activation has
`been noted for the other members of the particulate
`guanylate cyclase family (8) and has recently been
`observed for cells transfected with human GC-C (9). The
`data indicate that these peptides all possess the ability to
`
`Human Uroguanylin
`
`Human Guanylin
`
`PGTCEICAYAACTGC
`
`stimulate GC-C and share similar binding sites with
`varying degrees of relative affinities for the receptors in
`T84 cells.
`To assess the effect of human uroguanylin on well(cid:173)
`characterized ST- and guanylin-sensitive transport func(cid:173)
`tions, we assessed the effects of the peptide on Isc of T84
`cells and proximal rat colon. In these experiments, the
`measurement of I sc is used as an indicator of transepithe(cid:173)
`lial chloride secretion. Previous studies in these prepara(cid:173)
`tions have indicated that the change in /sc elicited by ST
`and guanylin is mostly accounted for by an increase in
`chloride secretion (5). Human uroguanylin added to the
`apical reservoir of T84 cells mounted in Ussing cham(cid:173)
`bers caused a marked and rapid increase in lsc (Fig. 5A).
`Similarly, human uroguanylin added to the mucosal
`reservoir of rat colon mounted in an U ssing chamber
`also caused a sustained rise in lsc (Fig. 5B).
`
`Opossum Uroguanylin
`
`QEDCE C INVACTGC
`
`DISCUSSION
`
`E.coli ST
`
`Fig. 3. Comparison of the structures of human uroguanylin, human
`guanylin, opossum uroguanylin, and E. coli heat-stable enterotoxin
`(ST). Identical amino acids are boxed. Human uroguanylin (position
`1-15) is 53%, 80%, and 73% identical to human guanylin, opossum
`uroguanylin, and ST, respectively.
`
`The present study describes the purification and
`structural identification of a new peptide, isolated from
`human urine, that activates GC-C. The finding of a
`bioactive peptide in human urine extends our prior
`efforts to isolate and identify peptides in the opossum
`(10). We have termed this peptide human uroguanylin
`
`Bausch Health Ireland Exhibit 2065, Page 4 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`F346
`
`BIOLOGICAL AND STRUCTURAL CHARACTERIZATION OF HUMAN UROGUANYLIN
`
`10 6 A
`---o-- STa
`-
`Human Uroguanylin
`---o-- Human Guany/in
`
`:::-
`'ii 105
`~
`
`(I)
`Q)
`0
`
`E 10 4 -~
`
`D.
`:l:
`C,
`()
`
`10 3
`
`10 2
`
`18
`
`110
`
`0
`
`-1 0
`
`- 9
`
`-8
`
`- 7
`
`- 6
`
`0
`Ill
`0 90
`~ 0
`,:i 70
`C
`:::l
`0
`
`Ill .,
`I-
`~ 30
`.,
`« - 10
`
`50
`
`-10
`
`0
`
`-1 0
`
`-9
`
`-8
`
`-7
`
`-6
`
`- 5
`
`[Peptide], log (M)
`
`Fig. 4. A: concentration-response effect of synthetic human urogua(cid:173)
`nylin, human guanylin, and E.coli ST-(5~18) (STa) on cGMP levelR in
`T84 cells. Cells were incubated with various concentrations of ligands
`for 30 min. Values are means ± SE (n = 4). B: displacement of
`1251-STa-specific binding from T84 cells by human uroguanylin,
`human guanylin, and STa. Cells were incubated for 30 min at 37°C
`with labeled STa and indicated concentrations of ligands. Specific
`binding(%) was determined by dividing specifically bound 1251-STa at
`each ligand concentration by specifically bound 1251-STa in absence of
`ligands. Each determination represents the mean of 4 wells examined.
`
`because of its source, human urine, and because of its
`strong structural similarity to guanylin and opossum
`uroguanylin. Uroguanylin shares many biological prop(cid:173)
`erties with guanylin and ST. Uroguanylin stimulated
`cGMP accumulation and competed with 125I-ST-(5-18)
`for binding sites on T84 cells. The more potent effect of
`uroguanylin on binding displacement than on the activa(cid:173)
`tion of GC-C is an unexplained observation, but it is
`characteristic of particulate guanylate cyclases, includ(cid:173)
`ing GC-C (8, 9). In the T84 cell, this discrepancy may in
`part be explained by the presence of multiple receptors
`with different binding affinities. Indeed, our recent
`detailed characterization of ST and guanylin binding in
`T84 cells was consistent with a model predicting two
`receptors with different affinities for the peptide ligands.
`Uroguanylin also stimulated an incr~ase in lsc when
`added to the apical surface of both T84 cell monolayers
`and isolated proximal rat colon. These properties make
`uroguanylin an ideal candidate to serve as a paracrine/
`
`endocrine regulator of fluid and electrolyte homeostasis
`in tissues that express GC-C.
`The observation that human urine contains urogua(cid:173)
`nylin but little or perhaps no guanylin is different from
`our results with opossum urine. We isolated both urogua(cid:173)
`nylin and a second peptide that is a putative opossum
`homologue of guanylin (10) from opossum urine. Spe(cid:173)
`cies differences between human and opossum may ac(cid:173)
`count for this difference. It is also possible that guanylin
`was secreted in an inactive form. Recently, it was
`reported that the plasma form of guanylin from human
`renal patients is a much larger precursor that may have
`less intrinsic activity (14). We could find no evidence for
`the presence of a guanylin precursor. Attempts were
`made to activate the crude extract and larger-molecular(cid:173)
`weight fractions from the G-25 column by acetic acid
`treatment, with no success (data not shown). This
`treatment has been shown to activate the guanylin
`precursor because of cleavage at an acid-labile bond (4,
`16). These data strongly indicate that the major and
`perhaps the sole source of GC-C stimulatory activity in
`human urine is uroguanylin.
`The finding of another member of the guanylin pep(cid:173)
`tide family has parallels with other peptides and is
`particularly relevant to the natriuretic peptides. Both
`the guanylin peptides and the natriuretic peptides acti(cid:173)
`vate specific forms of particulate guanylate cyclase. ANP
`and BNP both potently activate GC-A, whereas CNP
`activates GC-B (13). These peptides appear to play an
`important role in the regulation of blood pressure and
`
`14 A
`12
`
`"'
`!310
`' < ... 8
`u
`Ill
`1-t 6
`
`4
`
`20
`
`40
`
`B
`
`60
`
`80
`TIME MIN
`
`100
`
`120
`
`140
`
`t
`
`Uroguanylln [0.2 µM]
`
`S minutes
`
`Fig. 5. Effect of synthetic human uroguanylin on short-circuit current
`Uscl of T84 cells or rat colon. A: stimulation of transepithelial Cl(cid:173)
`secretion in T84 cells by human uroguanylin. T84 cells cultured on
`collagen-coated filters were mounted in a modified Ussing chamber at
`time 0. After a steady baseline was achieved, 1 µM human uroguanylin
`was added to the apical reservoir. B: effect of human uroguanylin (0.2
`µM) on l,c across rat proximal colon after a mucosa! addition.
`Response in both A and B is characteristic of results from 3 other
`experiments.
`
`Bausch Health Ireland Exhibit 2065, Page 5 of 7
`Mylan v. Bausch Health Ireland - IPR2022-00722
`
`

`

`BIOLOGICAL AND STRUCTURAL CHARACTERIZATION OF HUMAN UROGUANYLIN
`
`F347
`
`fluid and electrolyte homeostasis through cGMP(cid:173)
`mediated processes (8, 22). Similarly, guanylin and
`uroguanylin activate GC-C and possess the characteris(cid:173)
`tics of paracrine regulators of electrolyte transport
`through cGMP-mediated processes. Guanylin, urogua(cid:173)
`nylin, and other potential members of this peptide
`family may play a critical role in the regulation of
`epithelial cell function by acting on GC-C, proteins
`homologous to GC-C, and perhaps non-guanylate cy(cid:173)
`clase receptors. Future exploration of this area may
`reveal an intricate paracrine/endocrine network for the
`regulation of epithelial cell function through the manipu(cid:173)
`lation of multiple guanylin-like peptides and their recep(cid:173)
`tors.
`The role of uroguanylin in the control of kidney
`function is unknown at present. However, ST has been
`shown to cause a marked increase of sodium excretion in
`the perfused rat kidney (15). It has been proposed that
`guanylin is secreted into the lumen of the intestine and
`subsequently activates the apically located GC-C (2, 23,
`24). Activation of GC-C results in an increase in intracel(cid:173)
`lular cGMP levels, a decrease in sodium and water
`absorption, and an increase in chloride secretion (2, 5).
`Uroguanylin could act in a similar manner in the
`kidney. It may be secreted or filtered into the lumen of
`the nephron to act on local or distal sites in the nephron
`to regulate the action of apical receptors and to influence
`fluid and electrolyte homeostasis. Because the rat kid(cid:173)
`ney responds to ST with an increase of sodium excre(cid:173)
`tion, a receptor for guanylin/uroguanylin is likely pre(cid:173)
`sent in renal tissue. ST binding has been demonstrated
`on the apical surface of the opossum proximal tubule,
`and ST increased cGMP levels in opossum renal tissue
`(6). However, attempts to demonstrate the presence of
`GC-C mRNA in rat renal tissue have been unfruitful
`(16, 17). A potential molecular target for cGMP / cGMP(cid:173)
`dependent protein kinase resulting from uroguanylin
`stimulation is the cystic fibrosis transmembrane conduc(cid:173)
`tance regulator (CFTR). CFTR has been localized to the
`apical surface of proximal and distal human renal
`tubules (1). Further studies examining the role of
`guanylin/uroguanylin and GC-C in the regulation of
`CFTR appear to be warranted.
`Determination of the tissue and cellular source of
`uroguanylin will provide useful information regarding
`the potential physiological role of this peptide. The
`kidney is a likely source of uroguanylin because the
`urine levels appear to be relatively high, but it is possible
`that uroguanylin is made by another organ and is
`filtered by the kidney into the urine. To rule out the
`possibility that the source of uroguanylin in males was
`the prostate gland or other male accessory sex glands,
`we purified the source of bioactivity from urine obtained
`from female subjects. The bioactivity found in female
`urine was similar in quantity to that found in male urine
`and eluted with similar retention times using gel filtra(cid:173)
`tion and reverse-phase HPLC (data not shown). Thus
`future studies targeted towarcl determining the exact
`tissue and cellular sources of this peptide are critical to
`our understanding of the physiological actions of urogua(cid:173)
`nylin.
`
`In summary, the present study describes the discov(cid:173)
`ery of a novel bioactive peptide, uroguanylin, from
`human urine with GC-C stimulatory activity. Urogua(cid:173)
`nylin appears to belong to the guanylin/ST peptide
`family. Human urine contains a relatively large amount
`of uroguanylin but an undetectable level of guanylin.
`This observation indicates that at least two independent
`regulatory systems for GC-C may exist. It is possible
`that renal electrolyte balance is under the control of
`uroguanylin and that guanylin acts predominantly in
`the intestine. Thus the findings of this study should
`provide an impetus to elucidate the role of uroguanylin
`in the control of renal function and to further examine
`the molecular features of the guanylin/ guanylate cy(cid:173)
`clase system.
`
`Address for reprint requests: M. G. Currie, Mail Zone T3P,
`Monsanto Corporate Research, 800 N. Lindbergh Blvd., St. Louis, MO
`63167.
`Received 29 September 1993; accepted in final form 12 November
`1993.
`
`REFERENCES
`1. Crawford, I., P. C. Maloney, P. L. Zeitlin, W. B. Guggino,
`S. C. Hyde, H. Turley, K. C. Gatter, A. Harris, and C. F.
`Higgins. Immunocytochemical localization of the cystic fibrosis
`gene product CFTR. Proc. Natl. Acad. Sci. USA 88: 9262-9266,
`1991.
`2. 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. Proc. Natl. Acad. Sci.
`USA 89: 94 7-951, 1992.
`3. De Sauvage, F. J., T. R. Camerato, and D. V. Goeddel.
`Primary structure and functional expression of the human recep(cid:173)
`tor for Escherichia coli heat-stable enterotoxin. J. Biol. Chem.
`266: 17912-17918, 1991.
`4. De Sauvage, F. J., S. Keshav, W.-J. Kuang, N. Gillett,
`W. Henzel, and D. V. Goedde!. Precursor structure, expression,
`and tissue distribution of human guanylin. Proc. Natl. Acad. Sci.
`USA 89: 9089-9093, 1992.
`5. Forte, L. R., S. L. Eber, J. T. Turner, R.H. Freeman, K. F.
`Fok, and M. G. Currie. Guanylin stimulation of Cl- secretion in
`human intestinal Ts4 cells via cyclic guanosine monophosphate.
`J. Clin. Invest. 91: 2423-2428, 1993.
`6. Forte, L. R., W. J. Krause, and R.H. Freeman. Escherichia
`coli enterotoxin receptors: localization in opossum kidney, intes(cid:173)
`tine, and testis. Am. J. Physiol. 257 (Renal Fluid Electrolyte
`Physiol. 26): F874-F881, 1989.
`7. Fraker, P. J., and J.C. Speck, Jr. Protein and cell membrane
`iodinations with a sparingly soluble chloroamide, 1,3,4,6-tetra(cid:173)
`chloro-3a,6a-diphenylglycoluril. Biochem. Biophys. Res. Com(cid:173)
`mun. 80: 849-857, 1978.
`8. Garbers, D. L. The gua

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