throbber
J Gastroenterol 2001; 36:219-225
`
`Review
`
`Journal o
`Gastroenterology
`© Springer-Verlag 2001
`
`Guanylin family: new intestinal peptides regulating electrolyte and
`water homeostasis
`
`Masamitsu NAKAZATO
`
`Third Departmentof Internal Medicine, Miyazaki Medical College, Miyazaki 889-1692, Japan
`
`The regulation of intestinal salt and water transportis
`critical to the maintenanceof fluid volume. Control of
`this life-sustaining activity is mediated by the concerted
`actions of hormones, neurotransmitters, and locally
`acting factors. Guanylin and uroguanylin are novel
`peptides that were first isolated from rat jejunum and
`opossum urine, respectively. They bind to and activate
`guanylyl cyclase-C (GC-C) receptors to regulate intesti-
`nal and renalfluid and electrolyte transport through the
`second messenger, cyclic guanosine 3’,5'-monophos-
`phate (GMP). Heat-stable enterotoxins produced by
`pathogenic bacteria have close structural similarities to
`guanylin and uroguanylin, and they use this mimicry to
`act on GC-C, causing life-threatening secretory diar-
`rhea. Guanylin primarily is restricted to the intestine,
`whereas uroguanylin is present in the stomach,kidney,
`lung, and pancreas, in addition to intestine. Guanylin
`and uroguanylin are secreted into the intestinal lumen
`and blood in response to sodium chloride administra-
`tion. These peptides function in salt and water transport
`in the intestine and kidney by luminocrine and endo-
`crine actions. The guanylin family is involved in the
`pathophysiology of some gastrointestinal, renal, and
`heart diseases.
`
`guanylin,
`enterotoxin,
`heat-stable
`Key words:
`uroguanylin, guanylyl cyclase, salt homeostasis, intesti-
`nal natriuretic factor
`
`
`Introduction
`
`Diarrheal diseases are a leading cause of morbidity
`and mortality in humans, causing up to 50% of infant
`
`Received: August 21, 2000 / Accepted: November 17, 2000
`Reprint requests to: M. Nakazato
`
`deaths in developing countries.! Heat-stable enterotox-
`ins (STs) elaborated by various pathogenic bacteria,
`including enterotoxigenic Escherichia coli and Yersinia
`enterocolitica, bring about acute diarrhea, and also
`cause‘travelers diarrhea’ or‘turista’. STs are 15- to 30-
`amino acid peptides. In 1990, STs were shownto bind to
`intestinal receptor-guanylyl cyclase (GC-C) in the brush
`border membrane,” which subsequently leads to the
`activation of guanylyl cyclase.* Human GC-C,a 1050-
`amino acid protein and rat GC-C, a 1053-amino acid
`protein, have an extracellular domain, a transmem-
`brane domain, and intracellular protein kinase-like and
`guanylyl cyclase catalytic domains. Includedin this type
`of plasma membraneform of guanylyl cyclase are GC-
`A and GC-B, which are receptors of the natriuretic
`peptide family that functions in the regulation of body
`fluid balance. The paradox of a bacterial toxin acting
`on a mammalian receptor remained unclear until the
`following recent discoveries were made.
`
`Discoveries of guanylin and uroguanylin
`
`Just as opiates derived from the poppy predicted
`the existence of endogenous opium-like regulatory
`peptides, the isolation of bacterial ST peptides and
`elucidation of their cyclic guanosine 3’,5'-monophos-
`phate (CGMP)-regulating activity foreshadowedthe dis-
`covery of endogenous ligands for GC-C in mammals.
`STs act on GC-C, produce a second messenger, cGMP,
`and elicit the stimulation of Cl- secretion and the inhi-
`bition of Na* and H,O absorption, thereby causing
`secretory diarrhea.‘ Cells transfected with GC-C cDNA
`were proposedas a meansto search for the endogenous
`ligand by monitoring cGMPproduction, butcells that
`had a high sensitivity and a maximum response to STs
`were unavailable. Currie et al. found that T,, cells, a
`humancolon cancercell line, had the desired ability for
`the monitoring of cGMP production by STs, and an
`
`MSNExhibit 1020 - Page 1 of 7
`MSNv. Bausch - IPR2023-00016
`
`

`

`220
`
`M.Nakazato: Guanylins regulate electrolyte and water homeostasis
`
`human guanyl in
`
`rat guanylin
`
`1
`
`15
`
`Pro-Gly-Thr-Cys-Glu-Ile-Cys-Ala-Tyr-Ala-Ala-Cys-Thr-Gly-Cys
`
`--- “Al arn rn rrr rrr rrr rrr rrr rrr rcs rrr errs cscs
`
`human uroguanyl in
`
`Asn-Asp-Asp--------- Leu----- Val-Asn-Val------------rrrrr rrr Leu
`
`rat uroguanylin
`
`Thr-Asp-Glu--------- Leu----- lfe-Asn-Val--~------- 55555 r rr rne
`
`115
`
`115
`
`112
`
`106
`
`E.coli
`
`enterotoxin Asn-Ser-Ser-Asn-Tyr-Cys--------- Leu----- Cys-Asn-Pro--------- Thr--------- Tyr
`Lo
`
`Fig. 1. Primary structures of guanylin and uroguanylin. Dotted lines indicate the same amino acids as those in humanguanlyin,
`and brackets indicate disulfide bond linkage. Guanylin and rat uroguanylin are 15-aminoacid peptides, and human uroguanylin
`is a 16-aminoacid peptide. The numbers of amino acid residues of precursor proteins are shown on the right
`
`unidentified endogenous ligand. Indeed, T,, cells pos-
`sessed the sensitivity to detect 10pM ST, a maximum
`30 000-fold increase in cGMPto ST,and specificity for
`ST-like agonists. Using this assay combined with multi-
`step chromatography, Currie’s group isolated a cognate
`endogenousligand for GC-C from rat jejunum.’ This
`peptide wasdesignated guanylinafter its binding to GC-
`C. One year after the discovery of guanylin, another
`guanylin-like peptide, called uroguanylin, was isolated
`from the urine and intestinal mucosa of an opossum
`(Didelphis virniana, an American marsupial), using the
`same methodology.® Uroguanylin then wasidentified in
`the intestines and urine of humansandrats.** Guanylin
`and uroguanylin are 15- to 16-amino acid peptides(Fig.
`1). These two are novel peptides that have no sequence
`identities to other known peptides. Human and rat
`uroguanylin has two additional acidic residues that are
`not found in guanylin.
`As expected, the primary structures of STs are quite
`similar to those of the guanylin family (Fig. 1). Human
`guanylin shares 7 of 15 amino acids with Escherichia
`coli ST, and human uroguanylin shares 9 of 16 residues
`with it (47% and 56% similarity, respectively). STs are
`thought to use this mimicry to act on GC-C. A major
`structural difference between the guanlyin family pep-
`tides and STs is the numberof cysteines and disulfide
`bonds: guanylin and uroguanylin have four cysteines
`and two disulfide bridges compared with STs, with six
`cysteines and three disulfides. These disulfides are indis-
`pensable for optimal peptide potencies in the stimula-
`tion of cGMPproduction in vitro. The potency of the
`stimulation of guanylyl cyclase activity, in descending
`order,
`is STs, uroguanylin, and guanylin.*’? An addi-
`tional pair of cysteine residues in STs may contribute
`to their apparently higher potencies.+? Guanylin and
`uroguanylin are small molecules and have two intra-
`molecular disulfide bonds. This structural characteristic
`produces two topological isomers, a right-handed spiral
`
`form and a left-handed spiral form.'*!! One form is
`bioactive to stimulate cGMP production, but the other
`is not. These isomersare interconvertible; however, the
`biological significance and mechanism of the inter-
`conversion are still unclear.
`
`Gene sequenceandtissue distribution
`
`Based on the primary structure of guanylin and
`uroguanylin, cDNAs encoding their precursors were
`isolated.’**° Guanylin and uroguanylin mRNAsin hu-
`mans and rats have approximately 600 bases. Mature
`guanylin and uroguanylin peptides are located at the
`carboxy terminal ends of their precursor proteins.
`The sequence identity between human guanylin and
`uroguanylin is 20%,
`that between human and rat
`guanylin is 64%, and that between human andrat
`uroguanylin is 63%. Ten-kilodalton proguanylin is a
`major guanylin molecule in the intestinal mucosa and
`plasma of humans andrats, but it has no biological
`activity until cleaved by proteolytic enzymesto release
`the biologically active 15-amino acid peptide.?!* Ten-
`kilodalton prouroguanylin is also a major molecular
`form in the intestine of humans and rats, whereas
`mature 16-amino acid uroguanyin is a major form in the
`urine of both species.”
`Uroguanylin and guanylin genes are arranged in a
`tail-to-tail array on the short arm of human chromo-
`some 1p33-p34 and on mouse chromosome 4.207425
`The respective intergenic distances between the two
`genes in the human and mouse genomesare approxi-
`mately 6.5 and 8kb. The intergenic region between the
`human uroguanylin and guanylin genes has an Alu se-
`quence and a (CA/TG),, microsatellite sequence,
`suggesting that these two genes are produced by gene
`duplication. The genes of both human guanylin and
`uroguanylin consist of three exons and two introns
`
`MSNExhibit 1020 - Page 2 of 7
`MSNv. Bausch - IPR2023-00016
`
`

`

`M. Nakazato: Guanylins regulate electrolyte and water homeostasis
`
`221
`
`within an overall length of 2.5kb. The 5’ flanking region
`has TATA and CAAT boxes. The genes also have
`multiple binding sites for promoter-specific transcrip-
`tion factor, activator protein-1, activator protein-2,
`and a cyclic adenosine 3’,5'-monophosphate (cAMP)-
`regulated enhancer element.
`Northern blot and reverse transcription-polymerase
`chain reaction (RT-PCR) analyses have revealed that
`guanylin mRNAis most abundant in the intestine, and
`there are small amounts in the kidney, adrenal gland,
`uterus, and oviduct.’The distribution of guanylin
`mRNAintheintestine is characterized by relatively low
`levels in the duodenum and jejunum and much higher
`levels in the ileum and colon. Two radioimmunoassays
`(RIAs) for guanylin and uroguanylin, respectively, were
`developed.’Guanylin peptide is distributed widely
`from the duodenum to colon in both rats and humans,
`the highest contents being in the ileum and proximal
`colon. The plasma concentration of immunoreactive
`guanylin was 31.2 + 3.0fmol/ml (mean + SE)in normal
`individuals and was markedly higher in patients with
`renal
`insufficiency, which may be due to impaired
`catabolism of guanylin. Patients with carcinoid tumors
`that cause secretory diarrhea had elevated levels of cir-
`culating guanylin.2”? The tumors contained guanylin, as
`detected by immunohistochemical study. Although se-
`rotonin is considered to be a major cause of diarrhea in
`patients with carcinoid tumors, guanylin released from
`the tumors may also be responsible for the stimulation
`of intestinal fluid secretion. Northern blot analysis and
`RIA showedthat uroguanylin mRNAandpeptide were
`expressed in the stomach, small and large intestines,
`kidney, heart, and lung of humans andrats.*!*° The
`highest values were found in the upper small intestine.
`The plasma concentration of bioactive uroguanylin was
`5.0 + 0.3fmol/ml in normal individuals, and it was
`higher in patients with renal
`insufficiency.’ Plasma
`uroguanylin concentration in patients with heart failure
`was also significantly higher than that in normal con-
`trols, and increased with the severity of heart failure.
`Plasma uroguanylin levels in the coronary sinus and
`anterior interventricular vein were higher than that in
`the aorta, indicating that uroguanylin is secreted from
`the heart in heart failure. Uroguanylin has natriuretic
`activity, as described below. The possible function of
`uroguanylin in the regulation of body fluid balance in
`heart failure needs further investigation.
`
`Cellular localization
`
`The specific cellular sites of guanylin mRNA produc-
`tion in the intestine are a source of considerable debate,
`with conflicting reports.5?*° A recent immunohisto-
`chemical finding indicates that the enterochromaffin
`
`(EC) cells of guinea pig stomach and small intestine are
`a potential source of guanylin.° On the other hand,
`uroguanylin-producing cells have been well defined.
`Uroguanylin-immunoreactive cells in the rat intestine
`are round, basket-shapedortall flask-shaped cells with
`a dense accumulation of uroguanylin in the luminalcy-
`toplasm and a long, thin basal process immunoreactive
`for uroguanylin.”?! Uroguanylin-positive cells in the
`rat intestine were identified as EC cells because they
`reacted with serotonin antibody. The ECcell, the most
`abundant type of enteroendocrinecell, is widely distri-
`buted in the intestine. When stimulated, the EC cell
`releases serotonin and substance P both apically (into
`the lumen) and basolaterally (into the circulation).*
`Uroguanylin also is
`released from the intestine
`bidirectionally and is
`thought
`to function in a
`luminocrine (luminal secretion), endocrine, and/or
`paracrine fashion. The ECcell appears to have a fea-
`sible mechanism for delivering uroguanylin to luminally
`oriented GC-C,as well as to remote tissues such as the
`kidneyvia the circulation.
`Uroguanylin-producingcells in the rat stomach were
`identified as enterochromaffin-like (ECL) by im-
`munocytochemical methods and in situ hybridization
`cytochemistry using gastric mucosal cells isolated by
`counterflow elutriation.** ECL cells release histamine,
`leading to the stimulation of gastric acid secretion from
`parietal cells. Seven distinct endocrine cells: EC, ECL,
`D, D1, P, G, and X cells, have been identified ultrastruc-
`turally and immunohistochemically in rat and human
`gastric mucosa.***° Very recently, X cells have been
`clarified to produce ghrelin, a novel peptide that stimu-
`lates the release of growth hormone from the pitu-
`itary.*”*8 X cells thus can be abbreviated as Grcells.
`ECLcells are small cells (8- to 10-nm in diameter) that
`contain cytoplasmic vesicles with eccentric electron-
`dense cores. ECL cells are scattered in the oxyntic
`glands, often in direct contact with parietal cells.
`Patients with Zollinger-Ellison syndrome, presenting
`with gastrinoma, peptic ulcer, and ECL hyperplasia,
`had markedly high plasma concentrations of uro-
`guanylin. Further investigation of the cGMP-mediated
`gastric ion transport mechanism is a fascinating topic
`that could lead to better understanding of the regulation
`of gastric acid secretion.
`Uroguanylin and GC-C mRNAare present in B
`cells of the pancreatic islets, but the involvement of
`uroguanylin in the regulation of glucose metabolism has
`not been determined. The peptides produced in the
`“gastro-entero-pancreatic (GEP) endocrine system”
`function to control or modulate all the processes linked
`to the digestion and absorption of nutrients and water.”
`Uroguanylin may be a constituent of the GEP endo-
`crine system becauseofits cellular source and biological
`activity.
`
`MSNExhibit 1020 - Page 3 of 7
`MSNv. Bausch - IPR2023-00016
`
`

`

`222
`
`M.Nakazato: Guanylins regulate electrolyte and water homeostasis
`
`Escherichia coli
`Yersinia enterocolitica
`
`!
`
`enterotoxin
`
`guanylin or
`uroguanylin
`
`Guanylin or
`
`Nat’, H2O
`CFTR
`
`inhibition
`
`
`
`
`
`
`Uroguanylin-
` GTP
`
`
`cGMP
`Producing Cell
`
`
`enterocyte
`
`vessel
`Cl”
`guanylin or uroguanylin
`
`Cl”
`
`the guanylyl
`for
`Fig. 2. A model
`cyclase-C (GC-C) signaling pathway
`in intestinal Cl--secreting cells. Lumi-
`nally secreted guanylin and urogu-
`anylin activate GC-C in nearbycells.
`These peptides are also secreted into
`the bloodstream to function in an
`endocrine fashion. Cl” enters across
`the basolateral (serosal) membrane.
`Cyclic
`guanosine
`3’,5’-monophos-
`phate
`(cGMP)
`activates
`cGMP-
`dependent protein kinase II (PKG
`II). A key substrate for phosphoryla-
`tion by PKG II is the apical mem-
`brane-localized CFTR protein. CFTR
`serves as a channel for Cl- and HCO,”
`secretion across the apical (mucosal)
`membrane of epithelial cells. G7P,
`Guanosine 5’-triphosphate
`
`stimulation
`
`Kinase
`(PKGII)
`
`Molecular mechanism of guanylin and
`uroguanylin action
`
`A model for the GC-Csignaling pathway in intestinal
`Cl--secreting cells is shown in Fig. 2. GC-C is more
`abundantin the small intestine than in the large intes-
`tine. Moreover, a gradient of receptor levels was ob-
`served in small intestine, with the highest amounts in
`the upper portions of crypts and in the lowerparts of
`villi adjacent to the crypts.“ A truncated, GC-C-like
`mRNAthat has a 159-nucleotide deletion was found in
`the mucosa of rat stomach and intestine.*! The physi-
`ological implications of this truncated GC-C remain to
`be elucidated. Intestinal cells produce preproguanylin
`or preprouroguanylin. Luminal secretion of active
`peptides can activate GC-C in nearbycells of this epi-
`thelium in a luminocrine fashion. Proguanylin and
`prouroguanylin are secreted into the bloodstream to
`function in an endocrine fashion. cGMP activates
`cGMP-dependent protein kinase II (PKG II), which
`phosphorylates the apical membrane-localized CFTR
`protein. CFTRis present immunohistochemically in the
`epithelia of the intestine, stomach, airway, small pancre-
`atic ducts, renal tubules, and the sweat duct. CFTR is
`one member of a large family of ABC proteins that
`transport small molecules across the cell membrane in
`an ATP-dependentfashion. Guanylin and uroguanylin
`were verified to stimulate Cl- secretion in T,, cells, as
`measured byincreasesin the shortcircuit current, using
`an Ussing chamber.°* Cl-
`is secreted through the
`CFTR channel pathway across the apical (mucosal)
`membrane.
`
`transduction system and physiological
`The signal
`implications of guanylin and uroguanylin were also
`investigated by using transgenic and knockout mice.
`The mutation of CFTR genes, resulting in either loss of
`the protein or modification of its activity, underlies the
`genetic disease of cystic fibrosis. CFTR knockout mice
`had marked reductions of intestinal Cl- and HCO,-
`secretion responses to guanylin and uroguanylin.*4
`Mice deficient in PKG II wereresistant to E. coli.* PKG
`II knockout mice also developed dwarfism that was
`caused by a severe defect in endochondral ossification
`at
`the growth plates. GC-C-null mice had no STa-
`stimulable guanylyl cyclase activity.“ Gavage with STa
`resulted in marked fluid accumulation within the intes-
`tine of wild-type and heterozygous suckling mice,
`but GC-C-null animals were resistant. Infection with
`enterotoxigenic bacteria that produce STaled to diar-
`rhea and death in wild-type and heterozygous mice,
`whereas the null mice were protected. Diets including
`high carbohydrate, fat, or protein, or drinking water
`including high K* or Na* did not severely affect the
`GC-C-null animals.
`A wide range of mucosal acidity occurs within the
`intestinal lumen during digestion. At an acidic mucosal
`pH of 5.0, uroguanylin was 100-fold more potent than
`guanylin, but at an alkaline pH of 8.0, guanylin was
`more potent
`than uroguanylin in stimulating intra-
`cellular cGMP production and transepithelial chloride
`secretion.*”7 Uroguanylin and guanylin appear
`to
`cooperatively regulate the guanylyl cyclase activity of a
`common set of mucosal receptors in a pH-dependent
`fashion.
`
`MSNExhibit 1020 - Page 4 of 7
`MSNv. Bausch - IPR2023-00016
`
`

`

`M. Nakazato: Guanylins regulate electrolyte and water homeostasis
`
`223
`
`Uroguanylin as a candidate for intestinal
`natriuretic factor
`
`When dietary sodium chloride is low, the regulation
`of salt homeostasis is maintained by mineralcorticoids.
`However, contemporary diets commonly have excess
`sodium;
`thus, mechanisms are required to achieve
`salt homeostasis during sodium surfeit. An intestinal
`natriuretic factor has been sought, because the oral
`ingestion of sodium chloride causes a dramatic increase
`in urine salt excretion, whereas the same amount ad-
`ministered intravenously haslittle effect on renal salt
`excretion.
`The intravenous administration of uroguanylin to
`mice induced diuresis, natriuresis, and kaliuresis.*
`Guanylin was less potent than uroguanylin and STa.
`When uroguanylin was administered into the renal
`artery, it was filtered through the glomerulus and then
`activated the GC-C/CFTR system localized in the
`tubules, thereby causing natriuresis and an increase
`in cGMPexcretion.*° Urinary uroguanylin excretion in
`persons who took a high-salt diet was significantly
`higher than that in persons with a low-salt diet.*! The
`magnitude of uroguanylin excretion was proportional to
`increases in urinary Na* and cGMPexcretion in sub-
`jects receiving high-salt diets. The oral administration of
`salt to rats augmented uroguanylin mRNAlevels in the
`intestine and kidney. Uroguanylin secretion from the
`intestine in response to salt was studied in vitro. In
`isolated vascularly and luminally perfused rat intestine,
`uroguanylin produced in the intestine was secreted
`mainly in the lumen, but in part in the blood, in re-
`sponse to high-concentration sodium chloride adminis-
`tration. These findings taken together indicate that
`uroguanylin is a prime candidate for a substance that
`could link the intestine and kidney in an endocrine
`pathway that regulates renal salt metabolism. Uro-
`guanylin secretion from the gastrointestinal tract or
`other organs such as the heart could be increased
`secondary to renal Na* retention as a compensatory
`mechanism involving enhanced circulating levels of
`uroguanylin functioning as a natriuretic and diuretic
`hormone. Thus, plasma uroguanylin may be elevated
`to help maintain body salt and water balance in both
`normal physiological circumstances and in diseases such
`as congestive heart failure and nephrosis,*° in which
`Na* and water retention cause edema. In contrast to
`uroguanylin, guanylin gene expression wasnotaffected
`by a high-salt diet? A low-salt diet reduced the ex-
`pression of guanylin to 30%-40% of the level found
`in control animals. The guanylin pathway is thought
`to be down-regulated as an adaptive response to salt
`restriction.
`In conclusion, membrane guanylyl cyclase-C, GC-C,
`is an intestinal receptor for guanylin and uroguanylin
`
`that is responsible for the stimulation of Cl- and HCO,~
`secretion into the intestinal
`lumen. Guanylin and
`uroguanylin are produced mainly in the gastrointestinal
`epithelial cells to serve in a luminocrine mechanism for
`the regulation of gastrointestinal fluid and electrolyte
`secretion. Uroguanylin also serves in an endocrine axis
`linking the intestine and kidney, where its natriuretic
`and diuretic actions contribute to the maintenance of
`Na? balance after the oral ingestion of NaCl. Enteric
`bacteria secrete peptide toxin that mimics guanylin pep-
`tides, activating GC-C, to produce secretory diarrhea.
`Guanylin peptides are involved in the pathophysiology
`of such gastrointestinal diseases as diarrhea and peptic
`ulcer, and in chronic renal diseases or congestive heart
`failure,
`in which guanylin and/or uroguanylin levels
`in the circulation and/or urine are elevated. Guanylin
`peptides function in the regulation of salt and water
`homeostasis in the gastrointestinal tract and kidney.
`
`References
`
`1. Field M, Rao MC, Chang EB. Intestinal electrolyte transport and
`diarrheal disease. N Engl J Med 1989;321:879-83.
`2. Schulz S, Green CK, Yuen PS, Garbers DL. Guanylyl cyclase is a
`heat-stable enterotoxin receptor. Cell 1990;63:941-8.
`3. Garbers DL, Lowe DG. Guanylyl cyclase receptors. J Biol Chem
`1994;269:30741-4.
`4. Giannella RA, Luttrell M, Thompson M.Binding of Escherichia
`coli heat-stable enterotoxin to receptors on rat intestinal cells.
`Am J Physiol 1983;245:G492-8.
`5. Currie MG, Fok KF, Kato J, Moore RJ, Hamra FK, Duffin KL,
`et al. Guanylin: an endogenousactivator of intestinal guanylate
`cyclase. Proc Natl Acad Sci USA 1992;89:947-51.
`6. Hamra FK, Forte LR, Eber
`SL, Pidhorodeckyj NV,
`Krause WJ, Freeman RH, et al. Uroguanylin: structure and
`activity of a second endogenous peptide that stimulates in-
`testinal guanylate cyclase. Proc Natl Acad Sci USA 1993;90:
`10464-8.
`7. Kita T, Smith CE, Fok KF, Duffin KL, Moore WM,KarabatsosP,
`et al. Characterization of human uroguanylin: a member of the
`guanylin peptide family. Am J Physiol 1994;266:F342-8.
`8. Nakazato M, Yamaguchi H, Kinoshita H, Kangawa K, Matsuo H,
`Chino N,et al. Identification of biologically active and inactive
`human uroguanylins in plasma and urine and their increases
`in renalinsufficiency. Biochem Biophys Res Commun 1996;220:
`586-93.
`9. Fan X, Hamra FK, London RM, Eber SL, Krause WJ,
`Freeman RH,et al. Structure and activity of uroguanylin and
`guanylin from the intestine and urine of rats. Am J Physiol 1997;
`273:E957-64.
`10. Chino N, Kubo S, Miyazato M, Nakazato M, Kangawa K,
`Sakakibara S. Generation of two isomers with the same disulfide
`connectivity during
`disulfide
`bond formation of human
`uroguanylin. Lett Pept Sci 1996;3:45-52.
`11. Chino N, Kubo §S, Kitani T, Yoshida T, Tanabe R, Kobayashi Y,
`et al. Topological isomers of human uroguanylin: interconversion
`between biologically active and inactive isomers. FEBS Lett
`1998;421:27-31.
`12. Wiegand RC, Kato J, Currie MG. Rat guanylin cDNA:character-
`ization of the precursor of an endogenousactivator of intestinal
`guanylate cyclase. Biochem Biophys Res Commun 1992;185:
`812-7.
`
`MSNExhibit 1020 - Page 5 of 7
`MSNv.Bausch - IPR2023-00016
`
`

`

`224
`
`13.
`
`14.
`
`15.
`
`16.
`
`17.
`
`18.
`
`19.
`
`20.
`
`21.
`
`22.
`
`23.
`
`24.
`
`25.
`
`26.
`
`27.
`
`28.
`
`29.
`
`30.
`
`M.Nakazato: Guanylins regulate electrolyte and water homeostasis
`
`Wiegand RC, Kato J, Huang MD, Fok KF, Kachur JF, Currie
`MG. Human guanylin: cDNAisolation, structure, and activity.
`FEBSLett 1992;311:150-4.
`Schulz S, Chrisman TD, Garbers DL. Cloning and expression of
`guanylin. Its existence in various mammaliantissues. J Biol Chem
`1992;267:16 019-21.
`de Sauvage FJ, Keshav S, Kuang WJ, Gillett N, Henzel W,
`Goeddel DV. Precursor
`structure,
`expression,
`and tissue
`distribution of human guanylin. Proc Natl Acad Sci USA 1992;89:
`9089-93.
`Hill O, Cetin Y, Cieslak A, Magert HJ, Forssmann WG. A
`new human guanylate cyclase-activating peptide (GCAP-I,
`uroguanylin): precursor cDNA andcolonic expression. Biochim
`Biophys Acta 1995;1253:146-9.
`Miyazato M, Nakazato M, Matsukura S, Kangawa K, Matsuo H.
`Uroguanylin gene expression in the alimentary tract and extra-
`gastrointestinal tissues. FEBS Lett 1996;398:170-4.
`Fan X, Hamra FK, Freeman RH,Eber SL, Krause WJ, Lim RW,
`et al. Uroguanylin: cloning of preprouroguanylin cDNA, mRNA
`expressionin the intestine and heartandisolation of uroguanylin
`and prouroguanylin from plasma. Biochem Biophys Res
`Commun 1996;219:457-62.
`Li Z, Perkins AG, Peters MF, Campa MJ, Goy MF.Purification,
`cDNAsequence,andtissue distribution of rat uroguanylin. Regul
`Pept 1997;68:45-56.
`Magert HJ, Reinecke M, David I, Raab HR, Adermann K, Zucht
`HD, et al. Uroguanylin: gene structure, expression, processing
`as a peptide hormone, and co-storage with somatostatin in
`gastrointestinal D-cells. Regul Pept 1998;73:165-76.
`Nakazato M, Yamaguchi H, Shiomi K, Date Y, Fujimoto S,
`Kangawa K,et al. Identification of 10-kDa proguanylin as a major
`guanylin molecule in humanintestine and plasmaandits increase
`in renal insufficiency. Biochem Biophys Res Commun 1994;
`205:1966-75.
`Hamra FK, Fan X, Krause WJ, Freeman RH, Chin DT, Smith CE,
`et al. Prouroguanylin and proguanylin: purification from colon,
`structure, and modulation of bioactivity by proteases. Endocrinol-
`ogy 1996;137:257-65.
`Yamaguchi H, Nakazato M, Miyazato M, Kangawa K,
`Matsuo H, Matsukura S. Two novel rat guanylin molecules,
`guanylin-94 and guanylin-16, do notincrease cyclic GMP produc-
`tion in T84 cells. Biochem Biophys Res Commun 1995;214:
`1204-10.
`
`Miyazato M, Nakazato M, Matsukura S, Kangawa K, Matsuo H.
`Genomic structure and chromosomal
`localization of human
`uroguanylin gene. Genomics 1997;43:359-65.
`Whitaker TL, Steinbrecher KA, Copeland NG, Gilbert DJ,
`Jenkins NA, Cohen MB. The uroguanylin gene (Gucalb) is
`linked to guanylin (Guca2) on mouse chromosome 4. Genomics
`1997;45:348-54.
`Nakazato M, Yamaguchi H, Date Y, Miyazato M, Kangawa K,
`Goy MF,etal. Tissue distribution, cellular source, and structural
`analysis of rat immunoreactive uroguanylin. Endocrinology 1998;
`139:5247-54.
`Lewis LG, Witte DP, Laney DW, Currie MG, Cohen MB.
`Guanylin mRNAis expressed in villous enterocytes of the rat
`small intestine and superficial epithelia of the rat colon. Biochem
`Biophys Res Commun 1993;196:553-60.
`Li Z, Goy MF.Peptide-regulated guanylate cyclase pathways in
`rat colon:in situ localization of GCA, GCC, and guanylin mRNA.
`Am J Physiol 1993;265:G394—402.
`Kuhn M, Kulaksiz H, Cetin Y, Frank M, Nold R, Arnold R,
`et al. Circulating and tissue guanylin immunoreactivity in
`intestinal
`secretory diarrhoea. Eur J Clin Invest 1995;25:
`899-905.
`Cetin Y, Kuhn M,Kulaksiz H, Adermann K, Bargsten G, Grube
`D,et al. Enterochromaffin cells of the digestive system: cellular
`source of guanylin, a guanylate cyclase-activating peptide. Proc
`Natl Acad Sci USA 1994;91:2935-9.
`
`Bis
`
`32,
`
`33.
`
`34.
`
`35.
`
`36.
`
`37.
`
`38.
`
`39.
`
`40.
`
`41.
`
`42.
`
`43.
`
`44.
`
`45.
`
`46.
`
`47.
`
`48.
`
`49.
`
`Perkins A, Goy MF, Li Z. Uroguanylin is expressed by entero-
`chromaffin cells in the rat gastrointestinal tract. Gastroenterology
`1997;113:1007-14.
`Grgnstad KO, DeMagistris L, Dahlstrém A, Nilsson O, Price B,
`Zinner MJ, et al. The effects of vagal nerve stimulation on
`endoluminal release of serotonin and substance P into the feline
`small intestine. Scand J Gastroenterol 1985;20:163-9.
`Date Y, Nakazato M, Yamaguchi H, Kangawa K, Kinoshita Y,
`Chiba T, et al. Enterochromaffin-like cell, a cellular source of
`uroguanylin in rat stomach. Endocrinology 1999;140:2398-404.
`Hakanson R, Ekelund M, Sundler F. Activation and proliferation
`of gastric endocrinecells. In: Falkmer S, Hakanson R, Sundler F,
`editors. Evolution and tumor pathology of the neuroendocrine
`system. Amsterdam:Elsevier; 1984. p. 371-98.
`Solcia E, Capella C, Buffa R, Usellini L, Fiocca R, Sessa F. Endo-
`crinecells of the digestive system. In: Johnson LR,editor. Physi-
`ology of the gastrointestinal tract. New York: Raven; 1987. vol 1,
`p. 111-30.
`Sachs G, Zeng N,Prinz C. Physiology ofisolated gastric endocrine
`cells. Annu Rev Physiol 1997;59:234—56.
`Date Y, Kojima M, Hosoda H, Sawaguchi A, Mondal MS,
`SuganumaT,et al. Ghrelin, a novel growth-hormone-releasing
`acylated peptide, is synthesized in a distinct endocrinecell type in
`the gastrointestinal tracts of rats and humans. Endocrinology
`2000;141:4255-61.
`Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa
`K. Ghrelin is a growth-hormone-releasing acylated peptide from
`stomach. Nature 1999;402:656-60.
`Grube D. The endocrine cells of the digestive system: amines,
`peptides, and modes of action. Anat Embryol 1986;175:
`151-62.
`
`Krause WJ, Cullingford GL, Freeman RH,Eber SL, Richardson
`KC, Fok KF, et al. Distribution of heat-stable enterotoxin/
`guanylin receptorsin the intestinal tract of man and other mam-
`mals. J Anat 1994;184:407-17.
`London RM, Krause WJ, Fan X, Eber SL, Forte LR. Signal
`transduction pathways via guanylin and uroguanylin in stomach
`and intestine. Am J Physiol 1997;273:G93-105.
`Forte LR, Eber SL, Turner JT, Freeman RH, Fok KF, Currie
`MG.Guanylin stimulation of Cl- secretion in human intestinal
`T84 cells via cyclic guanosine monophosphate. J Clin Invest
`1993;91:2423-8.
`Cuthbert AW, Hickman ME, MacVinish LJ, Evans MJ, Colledge
`WH,Ratcliff R, et al. Chloride secretion in response to guanylin
`in colonic epithelial from normal and transgenic cystic fibrosis
`mice. Br J Pharmacol 1994;112:31-6.
`Joo NS, London RM, Kim HD,Fort LR, Clarke LL. Regulation
`of intestinal Cl- and HCO;~ secretion by uroguanylin. Am J
`Physiol 1998;274:G633-44.
`Pfeifer A, Aszodi A, Seidler U, Ruth P, Hofmann F,Fassler R.
`Intestinal secretory defects and dwarfism in mice lacking cGMP-
`dependentprotein kinase II. Science 1996;274:2082-6.
`Schulz S, Lopez MJ, Kuhn M, Garbers DL. Disruption of the
`guanylyl cyclase-C gene leads to a paradoxical phenotype of
`viable but heat-stable enterotoxin-resistant mice. J Clin Invest
`1997;100:1590-5.
`Hamra FK, Eber SL, Chin DT, Currie MG, Forte LR. Regu-
`lation of
`intestinal uroguanylin/guanylin receptor-mediated
`responses by mucosalacidity. Proc Natl Acad Sci USA 1997;94:
`2705-10.
`
`Lennane RJ, Peart WS, Carey RM, Shaw J. A comparison on
`natriuresis after oral and intravenous sodium loading in sodium-
`depleted rabbits: evidence for a gastrointestinal or portal monitor
`of sodium intake. Clin Sci Mol Med 1975;49:433-6.
`Greenberg RN,Hill M, Crytzer J, Krause WJ, Eber SL, Hamra
`FK, et al. Comparison of effects of uroguanylin, guanylin, and
`Escherichia coli heat-stable enterotoxin STa in mouse intestine
`and kidney: evidence that uroguanylin is an intestinal natriuretic
`hormone. J Invest Med 1997;45:276-83.
`
`MSNExhibit 1020 - Page 6 of 7
`MSNv. Bausch - IPR2023-00016
`
`

`

`M. Nakazato: Guanylins regulate electrolyte and water homeostasis
`
`225
`
`50. Fonteles MC, Greenberg RN, Monteiro HS, Currie MG,
`Forte LR. Natriuretic and kaliuretic activities of guanylin and
`uroguanylin in the isolated perfused rat kidney. Am J Physiol
`1998;275:F191-7.
`51. Kinoshita H, Fujimoto S, Nakazato M, Yokota N, Date Y,
`Yamaguchi H, et al. Plasma and urine levels of uroguanylin
`
`and its molecular forms in renal diseases. Kidney Int 1997;52:
`1028-34.
`52. Li Z, Knowles JW, Goyeau D, Prabhakar S, Short DB,
`Perkins AG,et al. Low salt intake down-regulates the guanylin
`signaling pathway in rat distal colon. Gastroenterology 1996;
`111:1714-21.
`
`MSNExhibit 1020 - Page 7 of 7
`MSNv. Bausch - IPR2023-00016
`
`

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