JASN Express. Published on December 28, 2005 as doi: 10.1681/ASN.2005080818 Review

Cellular Effects of Guanylin and Uroguanylin

Aleksandra Sinic´and Eberhard Schlatter Universita¨tsklinikum Mu¨nster, Medizinische Klinik und Poliklinik D, Experimentelle Nephrologie, Mu¨nster, Germany

Ingestion of a salty meal induces secretion of guanylin (GN) and uroguanylin (UGN) into the intestinal lumen, where they ؉ ؊ ؊ inhibit Na absorption and induce Cl , HCO3 , and water secretion. Simultaneously, these hormones stimulate renal electrolyte excretion by inducing natriuresis, kaliuresis, and diuresis. GN and UGN therefore participate in the prevention of hypernatremia and hypervolemia after salty meals. The signaling pathway of GN and UGN in the intestine is well known. They activate enterocytes via guanylate cyclase C (GC-C), which leads to cGMP-dependent inhibition of Na؉/H؉ exchange and activation of the cystic fibrosis transmembrane regulator. In GC-C–deficient mice, GN and UGN still produce renal natriuresis, kaliuresis, and diuresis, suggesting different signaling pathways in the compared with the intestine. Signaling pathways for GN and UGN in the kidney differ along the various nephron segments. In proximal tubule cells, a cGMP- and GC-C–dependent signaling was demonstrated for both . In addition, UGN activates a pertussis toxin– sensitive G-protein–coupled receptor. A similar dual signaling pathway is also known for atrial natriuretic . Recently, a cGMP-independent signaling pathway for GN and UGN was also shown in principal cells of the human and mouse cortical collecting duct. Because GN and UGN activate different signaling pathways in specific organs and even within the kidney, this review focuses on more recent findings on cellular effects and signaling mechanisms of these peptides and their pathophysiologic implications in the intestine and the kidney. J Am Soc Nephrol ●●: ●●●–●●●, ●●●●. doi: 10.1681/ASN.2005080818

uanylin peptides, guanylin (GN) and uroguanylin endogenous peptides GN and UGN, STa contains three di- (UGN), are small, heat-stable peptides with 15 to 19 sulfide bonds. It is presumed that this is the reason for its G amino acids. Human GN consists of 15 amino acids stronger and uncontrolled activation of GC-C that leads to and possesses two disulfide bonds between the cysteins in marked intestinal secretion of electrolytes and water and is positions 4 to 12 and 7 to 15 (1). Human UGN consists of 16 manifested as secretory diarrhea (9). The identification of a amino acids and also has two disulfide bonds at the same receptor for the exogenous enterotoxin in the intestine and in positions (2). These disulfide bonds are essential for the activity other tissues (kidney, reproductive system, and lung) led to of the peptides. The for guanylin peptides are located on the search for endogenous ligands for GC-C and their phys- the human (p33 to p36) and the mouse chromo- iologic function. In the early 1990s, GN was isolated from rat some 4 (3). Human GN and UGN are coded by different but intestine (5) and UGN from opossum urine (6). Other re- similar genes that consist of three exons and two introns. Both cently discovered new members of this peptide family are peptides are synthesized as prepropeptide. Guanylin peptides renoguanylin (10) and lymphoguanylin (11). Like GN and are produced in the intestine after ingestion of a salty meal and UGN, renoguanylin has two disulfide bonds, whereas lym- secreted into the intestinal lumen (4). In the intestine, GN and phoguanylin possesses only one. Lymphoguanylin is less UGN activate enterocytes via guanylate cyclase C (GC-C), with potent than GN and UGN in the intestine (11). As guanylin cGMP as second messenger (5,6). Activation of this pathway Ϫ Ϫ peptides regulate electrolyte excretion by the intestine and leads to the secretion of Cl and HCO3 and to the inhibition ϩ of Na absorption, which drives water secretion. In addition, the kidney, they complement other well-known hormonal these hormones induce an increased salt and water excretion in systems that are involved in regulation of water and electro- the kidney (7). The decrease of salt absorption in the intestine lyte homeostasis, such as the renin-angiotensin-aldosterone together with the increase of renal salt excretion prevents the system, adiuretin (vasopressin) or atrial natriuretic peptide development of hypernatremia after ingestion of high amounts (ANP), brain natriuretic peptide (BNP), and C type natri- of salt. uretic peptide (CNP). GC-C was first described as a receptor for the exogenous A number of excellent reviews on various aspects of guanylin heat-stable enterotoxin of Escherichia coli (STa) (8). Unlike the peptides exist (12,13); however, recent growing understanding of cellular signaling mechanisms also in humans is not covered in these reviews. Therefore, the aim of this review is to sum- Published online ahead of print. Publication date available at www.jasn.org. marize the current knowledge on cellular effects of guanylin Address correspondence to: Dr. Eberhard Schlatter, Universita¨tsklinikum Mu¨n- peptides and their signaling pathways and possible pathophys- ster, Medizinische Klinik und Poliklinik D, Experimentelle Nephrologie, iologic implications in different organs, mainly the intestine Domagkstrasse 3a, Mu¨nster 48149, Germany. Phone: ϩ49-251-83-56991; Fax: ϩ49- 251-83-56973; E-mail: [email protected] and the kidney of different species including man.

Copyright © ●●●● by the American Society of Nephrology ISSN: 1046-6673/●●●●-0001 2 Journal of the American Society of Nephrology J Am Soc Nephrol ●●: ●●●–●●●, ●●●●

Guanylate Cyclases The big family of guanylate cyclases consists of one cytoplas- mic (soluble) guanylate cyclase, which is the receptor for nitric oxide and carbonic monoxide, and six isoforms of membrane guanylate cyclases. GC-A, GC-B, and GC-C are receptors for natriuretic peptides that regulate electrolyte and water ho- meostasis and control BP (14). GC-A (or NPR-A) is the receptor for ANP and BNP; GC-B (or NPR-B) is the receptor for CNP. A third receptor that binds these three peptides, the so-called clearance receptor (or NPR-C), has no guanylate cyclase activ- ity, and hormone binding to this receptor leads to endocytosis of the hormone-receptor complex and activation of a pertussis toxin–sensitive G-protein. The latter mediates an inhibition of adenylate cyclase (15,16). Guanylate cyclases D, E, and F (GC-D, GC-E, and GC-F) are expressed in sensory organs, and, until now, their agonists and functions are not known (17–20). Figure 1. Guanylate cyclase C (GC-C) forming a dimer. The GC-G is also an orphan receptor without known agonist, and it binding of guanylin (GN), uroguanylin (UGN), or heat-stable is present in skeletal muscles, lungs, and intestine. Hirsch et al. enterotoxin of Escherichia coli (STa) to the extracellular domain (21) showed for the first time the presence of GC-G also in the of GC-C leads to activation of the catalytic domain with the kidney and identified this guanylate cyclase as the only mem- production of cGMP and the dephosphorylation of the kinase brane-bound isoform present in principal cells of rat cortical homology domain with receptor desensitization. For details, collecting duct (CCD). Table 1 summarizes the tissue distribu- see text. L, ligand. tion and known agonists for the eight guanylate cyclases iden- tified (22,23). terminal tail increased STa-mediated cGMP generation by 70% GC-C when compared with the nonphosphorylated receptor (31,32). GC-C is the main receptor for guanylin peptides in the intes- The existence of additional receptors for guanylin peptides tine (Figure 1). GC-C forms dimers, trimers, or tetramers when became evident when, for example, intestinal and renal effects inserted into the plasma membrane (24). mRNA for GC-C is of these peptides were examined in GC-C–deficient mice. present also in adrenal glands, brain, the embryonic or regen- These mice are resistant to intestinal secretion produced by STa erating but not adult liver, placenta, testis, airways, spleen, (33–35), but approximately 10% of the intestinal binding sites thymus, and lymphatic nodes (25–27). Human and rat GC-C for these peptides are still present when compared with wild- have 71% homology in the extracellular domain and 91% in the type mice. In the kidney, guanylin peptides still induce salure- intracellular domain. The extracellular domain must be glyco- sis and diuresis in these GC-C–deficient mice (36), strongly sylated for complete receptor activity (28,29), and dephosphor- suggesting an additional receptor and possibly signaling cas- ylation caused by binding of an agonist to the extracellular cade for guanylin peptides at least in the kidney and to some domain leads to loss of receptor sensitivity (30). extent also in the intestine. That renal effects of guanylin pep- The most studied regulator of GC-C activity is protein kinase tides are maintained when GC-C is absent also indicates that C (PKC). GC-C phosphorylation by PKC leads to an increase in GC-C plays no or only a minor role for these peptides in the cGMP accumulation. PKC-induced phosphorylation at the C- kidney.

Table 1. Expression of cytoplasmic and membrane-bound guanylate cyclasesa

Cyclases Tissue Distribution Agonists

Cytoplasmic Skeletal muscle, platelets, lung, liver, kidney, heart, CNS NO, CO GC-A Smooth muscle, kidney, adrenal gland, heart, CNS ANP, BNP GC-B Fibroblasts, heart, CNS CNP GC-C Intestine, adrenal gland, CNS, lung, reproductive glands, kidney STa, GN, UGN GC-D Olfactory cells Unknown GC-E Retina Unknown GC-F Retina Unknown GC-G Skeletal muscle, lung, intestine, kidney Unknown

aANP, atrial natriuretic peptide; BNP, brain natriuretic peptide; CNP, C-type natriuretic peptide; CNS, central nervous system; CO, carbon monoxide; NO, nitric oxide; STa, heat-stable enterotoxin of Escherichia coli; GN, guanylin; UGN, uroguanylin. J Am Soc Nephrol ●●: ●●●–●●●, ●●●● Cellular Effects of Guanylin and Uroguanylin 3

Signaling Pathways of Guanylin Peptides in the Intestine Guanylin peptides secreted into the intestinal lumen in re- sponse to high oral salt intake bind to GC-C localized in the luminal membrane of enterocytes and induce electrolyte and water excretion by a complex signaling cascade (Figure 2): 1. Increase of the intracellular concentration of cGMP (5,6,8) ϩ ϩ 2. Inhibition of Na /H exchange, which results in decreased ϩ Na reabsorption (37) 3. Activation of PKG II (38,39) 4. Activation of PKA directly (40,41) or indirectly via inhibition of phosphodiesterase III (PDE III), which leads to an increase of cellular cAMP and activation of PKA (39) 5. PKG II and PKA activate cystic fibrosis transmembrane reg- Ϫ ulator (CFTR) in the luminal membrane (42), leading to Cl secretion into the intestinal lumen Ϫ Ϫ 6. CFTR activates the Cl /HCO3 exchanger, which leads to bicarbonate secretion into the intestinal lumen (43) Figure 3. Relative expression of GN (f) and UGN (Ⅺ) mRNA along the intestinal tract. Adapted from reference (46). GN and UGN are expressed along the intestinal tract (Figure 3) together with GC-C (44–46), which is the major intestinal receptor for GN and UGN. However, the expression pattern of tinct from GC-C are also located on the basolateral membrane ϩ these two hormones is differential, i.e., highest expression of of colonocytes (47). STa also activates the Ca2 signaling path- UGN in the jejunum and highest expression of GN in ileum to way with activation of PKC (48,49), which could be indepen- proximal colon. The distal colon expresses only small amounts dent of GC-C activation. of both hormones. The effects of GN and UGN on cellular cGMP via GC-C As indicated above, a few studies show the existence of a depend on extracellular pH. UGN is more potent at pH 5 signaling pathway at least for STa distinct of GC-C (33–35). compared with pH 8. Conversely, GN is more potent at pH 8 Furthermore, binding sites for guanylin peptides that are dis- compared with pH 5 (Figure 4). As this pH dependence affects the ligand/receptor interaction, it was suggested that the N- terminal ends of the UGN and GN molecules are responsible for this pH dependence (50). The duodenum secretes bicarbon- ate to neutralize the acidic pH of the stomach contents that enter the duodenum. This acid-stimulated duodenal mucosal bicarbonate secretion is cGMP dependent. UGN regulates the luminal pH in the duodenum by activation of bicarbonate ϩ secretion and inhibition of H secretion because unlike GN,

Figure 2. Signaling pathway of guanylin peptides in the intes- tine. GN, UGN, and STa activate membrane-bound GC-C, which increases the intracellular concentration of cGMP. cGMP ϩ ϩ inhibits the Na /H exchanger (NHE) and activates protein kinase G type II (PKG II) and protein kinase A (PKA). Both Ϫ Ϫ protein kinases activate Cl and HCO3 secretion via activation of cystic fibrosis transmembrane regulator (CFTR) followed by Ϫ Ϫ an activation of the Cl /HCO3 exchanger. Depicted is a sche- matic drawing of an enterocyte including the major compo- Figure 4. GN and UGN increase the cellular concentration of nents necessary for electrolyte transport. PDE III, phosphodi- cGMP pH-dependently. GN is more potent at pH 8.0, and UGN esterase type III (inhibited by cGMP). is more potent at pH 5.0. Adapted from reference (50). 4 Journal of the American Society of Nephrology J Am Soc Nephrol ●●: ●●●–●●●, ●●●●

UGN is localized predominantly in the proximal part of intes- tinal tract (Figure 3). This effect is enhanced as UGN has higher Ϫ activity at acidic pH and leads to stronger HCO3 secretion into duodenal lumen.

Signaling Mechanisms of Guanylin Peptides in the Kidney ϩ ϩ Guanylin peptides increase the secretion of Na ,K , and water in the kidney without changes in GFR or renal blood flow (7,36). UGN is proposed to be an intestinal natriuretic peptide, as natriuresis produced by oral salt load is decreased in UGN- deficient mice (51). Thus, the action of UGN in the kidney can be endocrine (produced in the intestine with actions in the kidney), paracrine (produced by the kidney), or both. Kinoshita et al. (52) showed higher UGN concentration in blood and urine of individuals who were on a high-salt diet compared with those who were on a low-salt diet, which speaks in favor of an endocrine function of UGN. Recently, Fukae et al. (53) showed that rats that are fed a high-salt diet have higher UGN and cGMP concentrations in the urine but show no changes in the Figure 5. Relative mRNA expression of GN (A) and UGN (B) in concentration of UGN in the plasma compared with control various mouse nephron segments. GL, glomerulus; PT, proxi- animals. UGN is expressed in the kidney, and its expression is mal tubule; TAL, thick ascending limb of Henle’s loop; CD, higher in animals that are on a high-salt diet (53,54). It is still collecting duct; GAPDH, glyceraldehyde-3-phosphate dehy- not clear whether the increase in the urinary concentrations of drogenase. Mean values Ϯ SEM, n ϭ 4. Adapted from reference UGN and cGMP (52,53) are due to local production and secre- (54). tion of UGN within the kidney or to filtration in the glomeru- lus. Both GN and UGN circulate in the plasma and can be filtered (54) localized GC-C in the mouse kidney only in glomeruli and in the glomerulus into the primary urine. UGN is excreted via proximal tubules. In the human kidney, we detected GC-C the final urine, whereas hardly any GN can be found in the again only in the kidney cortex and specifically in proximal urine (6,55). A possible reason that GN is not present in the final tubules (Figure 6) (60,61). In contrary to these findings in mouse urine is its sensitivity to chymotrypsin, which quickly degrades and human, in rat kidney, Carrithers et al. (58) found mRNA for GN after glomerular filtration or secretion of GN into the GC-C in all nephron segments, suggesting differences in tissue tubular lumen (9,56). These differences in the degradation and distribution of GC-C in different species. A few years ago, it expression pattern of GN and UGN suggest that GN probably became evident that proximal tubules are targets of guanylin acts strictly autocrine, whereas UGN has paracrine activity in peptides. Guanylin peptides increase the intracellular cGMP the kidney. The concentration of UGN in the urine correlates concentration in a proximal tubule cell line of opossum kidney with that of urinary cGMP, which again supports the hypoth- (OK cells) via the opossum kidney GC-C (OK-GC), which is the esis that cGMP and the GC-C signaling pathways are activated analog to the human GC-C (62). Recently, we demonstrated an by UGN at least in the cells of the proximal tubule. However, increase in the cellular cGMP concentration induced by guany- another possible source of cGMP in the urine could be the lin peptides also in a human proximal tubule cell line (IHKE1) activation of other members of the guanylate cyclase family, (61). In this cell line, GN and UGN also activated GC-C. such as the orphan receptor GC-G, which is present in the The intestinal pH dependence for the binding of GN and luminal membrane of principal cells of rat and mouse CCD UGN to GC-C (see above) probably also is relevant in the (21,57). The source of guanylin peptides in the tubular lumen kidney. In the human proximal tubule cell line, UGN activated besides glomerular filtration from the circulation could also be tubular cells because mRNA for GN and UGN is also expressed in kidney epithelial cells (54,58,59). Similar to the difference in the axial expression along the intestine, GN and UGN are not expressed equally along the different nephron segments (Fig- ure 5). UGN is present mostly in the proximal tubule, again comparable to the proximal intestine, and GN is more pro- nounced in the collecting duct, in analogy to GN expression in the colon. Figure 6. Expression of mRNA for GC-C in human nephron Markedly different from the expression along the entire in- segments. G, glomeruli; CCD, cortical collecting ducts; M, testinal tract, GC-C, the first receptor for guanylin peptides marker; (Ϫ), negative control (no cDNA); (ϩ), positive control. identified, is not present in all nephron segments. Potthast et al. Adapted from reference (60). J Am Soc Nephrol ●●: ●●●–●●●, ●●●● Cellular Effects of Guanylin and Uroguanylin 5

GC-C at pH 5 more pronounced than at pH 8 (Figure 4). At alkaline pH, when UGN effects on GC-C in these proximal tubular cells were lower, an additional signaling pathway via pertussis toxin–sensitive G-proteins was revealed (61). There- fore, tubular pH can play an important role in activating GN versus UGN signaling pathways and activating different signal- ing pathways for UGN (Figure 7). This might be even more relevant along the medullary collecting duct, where tubular pH can be significantly acidic. Data on effects of guanylin peptides along this nephron segment, however, are still completely miss- ing. Data from various proximal tubule cells in culture suggest ϩ that guanylin peptides inhibit Na reabsorption in this ϩ nephron segment. UGN decreases the expression of the Na / ϩ K -ATPase, which would lower the concentration gradient for ϩ ϩ Na and therefore reduce Na -coupled electrolyte and sub- strate reabsorption from the tubular lumen (63). According to our own observations in the human proximal tubule cell line, ϩ GN and UGN decrease the electrical driving force for Na ϩ reabsorption by inhibiting K channels via cGMP and depolar- izing cells (61). Finally, the increase in cellular cGMP by GN and UGN should, like in the intestine (37), inhibit luminal ϩ ϩ ϩ Na /H exchange and thereby reduce Na and volume reab- sorption. Indeed, in the human proximal tubule cell line, UGN ϩ ϩ inhibited Na /H exchange activity (Schlatter E., unpublished results). As mentioned above, guanylin peptides still cause Figure 8. Signaling pathways of guanylin peptides in renal natriuresis, kaliuresis, and diuresis in GC-C–deficient mice, proximal tubule cells. Guanylin peptides activate two signaling suggesting a second receptor in the kidney for these peptides pathways. One is GC-C and cGMP dependent. cGMP decreases ϩ (36). In addition to this GC-C– and cGMP-dependent signaling Na reabsorption by inhibiting different proteins in the mem- ϩ ϩ ϩ ϩ ϩ pathway, UGN can activate a pertussis toxin–sensitive G-pro- brane: K channels, Na /H exchanger, and Na /K -AT- ϩ tein that leads to activation of a K conductance (61). Which Pase. The other signaling pathway involves activation of a signaling pathway will be activated by UGN could depend on pertussis toxin–sensitive G-protein. AQP1, aquaporin 1; NBC, ϩ Ϫ ϩ ϩ luminal pH (Figure 7). Figure 8 shows a summary of actions of Na /HCO3 co-transporter; NHE3, Na /H exchanger; OK- guanylin peptides on proximal tubular cells on ion and water GC, guanylate cyclase from opossum kidney. transport. The molecular identity of this second receptor, the physiologic role of this cGMP-independent signaling pathway, and the relative contributions of these two signaling pathways remains to be elucidated. The presence of two receptors for one peptide even at the same cell is known for other hormones that also act in the kidney. ANP activates two different types of receptors (64); one is GC-A, and, like GC-C, its activation leads to the production of cGMP (65). The other receptor is the natriuretic peptide receptor type C, or clearance receptor (NPR-C), which is a pertussis toxin–sensitive G-protein–coupled receptor (15,16). It remains to be clarified whether the G-protein–coupled recep- tors for UGN and ANP are the same or similar. Fine regulation of renal electrolyte and water excretion takes place in the collecting duct. Principal cells of the CCD are ϩ ϩ ϩ responsible for K secretion (via K channels [ROMK]), Na ϩ reabsorption (via epithelial Na channels [ENaC]), and water Figure 7. pH dependence of effects of guanylin peptides on reabsorption (via aquaporins 2, 3, and 4). In line with the membrane voltages of human proximal tubule cells. At pH 5.5, UGN activated GC-C and depolarized cells; at pH 8.0, it acti- observation that GC-C–deficient mice still exert natriuresis, vated a G-protein–coupled receptor and hyperpolarized the kaliuresis, and diuresis upon infusion of guanylin peptides (36) cells. GN and STa depolarize cells only via activation of the is the absence of mRNA for GC-C in CCD of mouse and man GC-C–dependent signaling pathway. Mean values Ϯ SEM, n (54,60). However, guanylin peptides induce changes in mem- given in brackets. Adapted from reference (61). brane voltages of principal cells of mouse and human CCD 6 Journal of the American Society of Nephrology J Am Soc Nephrol ●●: ●●●–●●●, ●●●● both in wild-type and in GC-C–deficient mice, suggesting mod- ification of electrogenic ion transport also in this nephron seg- ment (57,60). Similar to the situation in the human proximal tubule cell line, in principal cells of isolated mouse and human CCD segments, guanylin peptides activate a signaling pathway distinct from GC-C and independent of cGMP. Guanylin pep- tides depolarize these cells, whereas the membrane-permeable analog of cGMP, 8Br-cGMP, hyperpolarizes the same cells, excluding cGMP as a possible second messenger in this signal- ing pathway for guanylin peptides in these cells (60). This pathway of GN and UGN in the CCD involves again a G- protein–coupled receptor, like in the proximal tubule cell line. Activation of this receptor leads to stimulation of phospho- lipase A2 (PLA2) and liberation of arachidonic acid. It has been reported before that arachidonic acid inhibits the secretory ROMK channels located at the luminal membrane of principal ϩ cells (66). Inhibition of the secretory K channels by guanylin ϩ peptides therefore decreases K secretion and lowers the elec- ϩ trical driving force for Na reabsorption that results in natri- uresis. In mouse principal cells of the CCD, apparently in addition to this cGMP-independent signaling pathway, another pathway for guanylin peptides exists. This pathway is cGMP dependent but GC-C independent (57). The receptor that is activated by guanylin peptides in the mouse CCD and leads to Figure 9. Signaling pathways of GN and UGN in a principal cell cGMP and PKG activation is still not identified. A possible of the CCD. Guanylin peptides activate phospholipase A2 candidate for this receptor is the orphan receptor guanylate (PLA2), which leads to an increase in the intracellular concen- tration of arachidonic acid and an inhibition of luminal ROMK cyclase G (21,57). Activation of this pathway results in activa- ϩ ϩ K channels. ENaC, epithelial sodium channels; AQP 2, 3, 4, tion of cGMP-dependent K channels in the basolateral mem- ϩ ϩ aquaporins 2, 3, and 4; NKA, Na /K ATPase. brane of principal cells and cell hyperpolarization. Figure 9 shows a model of a principal cell of the CCD indicating all mechanisms of actions of guanylin peptides identified so far. It tides are also involved in the regulation of electrolyte and water is evident from this scheme that the guanylin peptides are the first secretion in those organs via CFTR. Guanylin peptides and peptides identified that act in this nephron segment primarily on parts of their signaling pathway are present in the pancreas. ϩ ϩ K channels without directly affecting Na channels. In the hu- John et al. (69) showed that GN and STa increase intracellular man CCD, the depolarization of principal cells via inhibition of the cGMP via GC-C in a human pancreatic cell line. Kulaksiz et al. ϩ secretory K channels results in natriuresis as the predominant (70,71) detected mRNA for GC-C and UGN in cells of the ϩ effect observed in vivo with these peptides (7,36,67). Reduced Na exocrine part of the pancreas, and guanylin peptides increase reabsorption also decreases water reabsorption in this nephron cGMP, which leads to activation of CFTR in these cells. segment and thus causes diuresis. GN was shown to decrease the Guanylin peptides and parts of the GC-C–dependent signal- cell volume and increase the luminal space in a concentration- and ing pathway are also found in airways (26,27), sweat glands, time-dependent manner, which suggests secretion of water and the adrenal medulla, and the male reproductive system (72–74). consequently diuresis (68). However, the kaliuresis also caused by According to this limited information on effects of guanylin guanylin peptides cannot be explained from the data obtained in peptides in various organs, the GC-C–dependent signaling the CCD and may be due to effects of these peptides in other parts pathway seems to be the predominant signaling pathway in of the nephron, e.g., the thick ascending limb or the medullary most organs and only in the kidney does the signaling involve collecting duct. Again, the physiologic role of the second cGMP- different receptors and signaling mechanisms. dependent signaling pathway and the relative contributions of these two signaling pathways in the CCD of the mouse remains to Guanylin Peptides in Pathophysiology be elucidated. This second pathway involves an activation of Exiting observations are the involvement of guanylin pep- ϩ ϩ basolateral K channels and thereby should increase Na reab- tides in the development and possible treatment of intestinal ϩ sorption without an increase in K secretion. tumors. GN and UGN are less expressed or they are not present at all in colon adenocarcinoma, adenoma, and intestine polyps Cellular Effects of Guanylin Peptides in in human and mouse (75–77). The for GN is located at 1 Other Organs p34–35, which is a tumor-modifying region. One can presume Patients with cystic fibrosis undergo changes in water and that loss of GN activity leads to or is the result of tumor electrolyte secretion in the liver, the pancreas, the lung, and the development. Pitari et al. (78) hypothesized that the reason for intestine. Thus, it might be speculated whether guanylin pep- the lower incidence of intestinal tumors in third-world coun- J Am Soc Nephrol ●●: ●●●–●●●, ●●●● Cellular Effects of Guanylin and Uroguanylin 7 tries is the higher incidence of intestinal infections that stimu- ular identity of these renal receptors for guanylin peptides late intestinal GC-C and lead to an increase in cGMP. Guanylin remains to be determined. First indications of an involvement peptides and also STa in addition regulate proliferation and of these peptides in various diseases indicate also a pathophys- differentiation, prolonging the cell cycle, and induce apoptosis iologic role and possibly a therapeutic value of these peptides. of T84, CaCo2 cells, and mouse intestinal cells (78–81). Shailubahai et al. (77) showed that oral application of UGN Acknowledgments leads to an decrease in number and size of polyps in mice that The data of the authors laboratory cited and presented in this review develop intestinal polyposis, which allows us to speculate on were funded by a grant of the Medical Faculty of the University of the possible usage of UGN in the treatment of intestinal tumors. Mu¨nster (IMF, KU 21 98 09) and by grants of the Deutsche Forschungs- The involvement of guanylin peptides was suggested also in gemeinschaft (Schl 277/11-1 to 11-3). different kidney diseases. More than 11 yr ago, Nakazato et al. (82) showed that human GN increases in patients who have chronic renal failure and undergo hemodialysis. The UGN References concentration in plasma and/or in urine is increased in patients 1. de Sauvage FJ, Keshav S, Kuang WJ, Gillett N, Henzel W, with chronic renal failure and glomerulonephritis and in pa- Goeddel DV: Precursor, structure, expression, and tissue tients on hemodialysis (52,83–85). In patients with nephrotic distribution of human guanylin. Proc Natl Acad Sci U S A syndrome, UGN plasma concentration was higher and urinary 89: 9089–9093, 1992 concentration was lower compared with values in healthy vol- 2. Kita T, Smith CE, Fok KF, Duffin KL, Moore WM, Kara- batsos PJ, Kachur JF, Hamra FK, Pidhorodeckyj NV, Forte unteers (84). Possible explanations for these observations are LR, Currie MG: Characterization of human uroguanylin: A kidney damages and reduced capability to metabolize and member of the guanylin peptide family. Am J Physiol Renal excrete guanylin peptides. Recently, Kikuchi et al. (86) sug- Physiol 266: F342–F348, 1994 gested that UGN plays a role as a natriuretic factor in nephrotic 3. Schulz S: Targeted gene disruption in the development of syndrome. In experimental nephrotic syndrome, induced by mouse models to elucidate the role of receptor guanylyl intraperitoneal injection of puromycin aminonucleoside (PAN), cyclase signaling pathways in physiological function. changes in UGN concentrations in urine and plasma corre- Methods 19: 551–558, 1999 ϩ sponded to changes in Na excretion. In the same animals, the 4. Kita T, Kitamura K, Sakata J, Eto T: Marked increase of expression of mRNA for UGN changed in the kidney but not in guanylin secretion in response to salt loading in the rat the intestine, and it was not seen in control animals. . Am J Physiol 277: G960–G966, 1999 In contrast to other natriuretic peptides, like ANP, the con- 5. Currie MG, Fok KF, Kato J, Morre RJ, Hamra FK, Duffin KL, Smith CE: Guanylin: An endogenous activator of in- nection between guanylin peptides and hypertension is not testinal guanylate cyclase. Proc Natl Acad Sci U S A 89: well understood. UGN-deficient mice have increased mean 947–951, 1992 arterial pressure, which is salt insensitive (51). However, GC- 6. Hamra FK, Forte LR, Eber SL, Pidhorodeckyj NV, Krause C–deficient mice show no difference in BP compared with WJ, Freeman RH, Chin DT, Tompkins JA, Fok KF, Smith wild-type mice (33,34), which rules out a significant involve- CE, Duffin KL, Siegel NR, Currie MG: Uroguanylin: Struc- ment of GC-C in the regulation of BP. As shown in this review, ture and activity of a second endogenous peptide that guanylin peptides activate at least one cGMP-, GC-C–indepen- stimulates intestinal guanylate cyclase. Proc Natl Acad Sci dent signaling pathway. Liberation of arachidonic acid in the USA90: 10464–10468, 1993 renal collecting duct by guanylin peptides inhibits the secretory 7. Fonteles MC, Greenberg RN, Monteiro HSA, Currie MG, ROMK channels, which lowers the electrical driving force for Forte LR: Natriuretic and kaliuretic activities of guanylin ϩ Na reabsorption, resulting in natriuresis. In UGN-deficient and uroguanylin in the isolated perfused rat kidney. Am J Physiol Renal Physiol 275: F191–F197, 1998 mice, UGN cannot activate this signaling pathway, which will ϩ 8. Schulz S, Green KC, Yuen PST, Garbers DL: Guanylyl lead to higher reabsorption of Na and possibly induce hyper- cyclase is a heat-stable enterotoxin receptor. Cell 63: 941– tension (51). 948, 1990 9. Carpick BW, Gariepy J: The Escherichia coli heat-stable en- Conclusions terotoxin is a long-lived superagonist of guanylin. Infect GN and UGN regulate electrolyte and water transport in the Immun 61: 4710–4715, 1993 intestine and cause kaliuresis, natriuresis, and diuresis in the 10. Yuge S, Inoue K, Hyodo S, Takei Y: A novel guanylin kidney. The membrane-bound GC-C is the receptor for these family (guanylin, uroguanylin, and renoguanylin) in eels: peptides in the intestine. In the kidney, these peptides activate Possible osmoregulatory hormones in intestine and kid- different signaling pathways along the nephron. Although ney. J Biol Chem 278: 22726–22733, 2003 11. Forte LR, Eber SL, Fan X, London RM, Wang Y, Roland L, GC-C is the predominant receptor mediating the effects of Chin DT, Freeman RH, Krause WJ: Lymphoguanylin: guanylin peptides in the intestine, it plays only a minor role in Cloning and characterization of a unique member of the the kidney, probably restricted to the proximal tubule. Whereas guanylin peptide family. Endocrinology 140: 1800–1806, in the intestine the signaling pathway of guanylin peptides is 1990 singular, in the kidney, at least three different receptors with 12. Forte LR: Uroguanylin and guanylin peptides: Pharmacol- different second messenger systems are activated and mediate ogy and experimental therapeutics. Pharmacol Ther 104: complex effects on electrolyte and water excretion. The molec- 137–162, 2004 8 Journal of the American Society of Nephrology J Am Soc Nephrol ●●: ●●●–●●●, ●●●●

13. Vaandrager AB: Structure and function of the heat-stable 29. Hasegawa M, Hidika Y, Wada A, Hirayama T, Shimonishi enterotoxin receptor/guanylyl cyclase C. Mol Cell Biochem Y: The relevance of N-linked glycosylation to the binding 230: 73–83, 2002 of a ligand to guanylate cyclase C. Eur J Biochem 263: 14. Kuhn M: Molecular physiology of natriuretic peptide sig- 338–345, 1999 naling. Basic Res Cardiol 99: 76–82, 2004 30. Potter LR, Garbers DL: Dephosphorylation of the guanylyl 15. Anand-Srivastava MB, Sehl PD, Lowe DG: Cytoplasmatic cyclase-A receptor causes desensitization. J Biol Chem 267: domain of natriuretic peptide receptor-C inhibits adenylyl 14531–14534, 1992 cyclase: Involvement of a pertussis toxin-sensitive G pro- 31. Wada A, Hasegawa M, Matsumoto K, Niidome T, Kawano tein. J Biol Chem 271: 19324–19329, 1996 Y, Hidaka Y, Padilla PI, Kurazono H, Shimonishi Y, 16. Murthy KS, Teng BQ, Zhou H, Jin JG, Grider JR, Makhlouf Hirayama T: The significance of Ser1029 of the heat-stable GM: Gi-1/Gi-2-dependent signaling by single-transmem- enterotoxin receptor (STaR): Relation of STa-mediated gua- brane natriuretic peptide clearance receptor. Am J Physiol nylyl cyclase activation and signaling by phorbol myristate 278: G974–G980, 2000 acetate. FEBS Lett 384: 75–77, 1996 17. Seebacher T, Beitz E, Kumagami H, Wild K, Ruppersberg 32. Crane JK, Wehner MS, Bolen EJ, Sando JJ, Linden J, Guer- JP, Schultz JE: Expression of membrane-bound and cyto- rant RL, Sears CL: Regulation of intestinal guanylate cy- solic guanylyl cyclase in the rat inner ear. Hearing Res 127: clase by the heat-stable enterotoxin of Escherichia coli (STa) 95–102, 1999 and protein kinase C. Infect Immun 60: 5004–5012, 1992 18. Juilfs DM, Fulle HJ, Zhao AZ, Houslay MD, Garbers DL, 33. Mann EA, Jump ML, Wu J, Yee E, Giannella RA: Mice Beavo JA: The subset of olfactory neurons that selectively lacking the guanylyl cyclase C receptor are resistant to express cGMP-stimulated phosphodiesterase (PDE2) and STa-induced intestinal secretion. Biochem Biophys Res Com- guanylyl cyclase D define a unique olfactory signal trans- mun 239: 463–466, 1997 duction pathway. Proc Natl Acad Sci U S A 94: 3388–3395, 34. Schulz S, Lopez MJ, Kuhn M, Garbers DL: Disruption of 1997 the guanylyl cyclase-C gene leads to a paradoxical pheno- 19. Fu¨lle HJ, Vassar R, Foster DC, Yang RB, Axel R, Garbers type of viable but heat-stable enterotoxin-resistant mice. DL: A receptor guanylyl cyclase expressed specifically in J Clin Invest 100: 1590–1595, 1997 olfactory sensory neurons. Proc Natl Acad Sci U S A 92: 35. Charney AN, Egnor RW, Alexander-Chacko JT, Zaharia V, 3571–3575, 1995 Mann EA, Giannella RA: Effect of E. coli heat-stable entero- 20. Yang RB, Foster DC, Garbers DL, Fulle H-J: Two mem- toxin on colonic transport in guanylyl cyclase C receptor- brane forms of guanylyl cyclase found in the eye. Proc Natl deficient mice. Am J Physiol 280: G216–G221, 2001 Acad Sci U S A 92: 602–606, 1995 36. Carrithers SL, Ott CE, Hill MJ, Johnson BR, Cai W, Chang 21. Hirsch JR, Kruhoffer M, Herter P, Adermann K, Heitland JJ, Shah RG, Sun C, Mann EA, Fonteles MC, Forte LR, A, Meyer M, Forssmann WG, Plenz G, Schlatter E: Cellular Jackson BA, Giannella RA, Greenberg RN: Guanylin and localization, membrane distribution and possible function uroguanylin induce natriuresis in mice lacking guanylyl of guanylyl cyclase A and 1 in collecting duct of rat. cyclase-C receptor. Kidney Int 65: 40–53, 2004 Cardiovasc Res 51: 553–561, 2001 37. Fawcus K, Gorrton VJ, Lucas ML, McEwan GTA: Stimula- 22. Garbers DL: The guanylyl cyclase receptors. Methods 19: tion of three distinct guanylate cyclases induces mucosal 477–484, 1999 surface alkalinisation in rat small intestine in vitro. Comp 23. Lucas KA, Pitari GM, Kazerounian S, Ruiz-Stewart I, Park Biochem Physiol A Physiol 118: 291–295, 1997 J, Schulz S, Chepenik KP, Waldman SA: Guanylyl cyclases Ј Ј and signaling by cyclic GMP. Pharmacol Rev 52: 375–414, 38. Vaandrager AB, Bot AGM, de Jonge HR: Guanosine 3 ,5 - 2000 cyclic monophosphate-dependent protein kinase II medi- 24. Vijayachandra K, Guruprasad M, Bhandari R, Manjunath ates heat-stable enterotoxin-provoked chloride secretion in UH, Somesh BP, Srinivasan N, Suguna K, Visweswariah rat intestine. Gastroenterology 112: 437–443, 1997 SS: Biochemical characterization of the intracellular do- 39. Vaandrager AB, Bot AGM, Ruth P, Pfeifer A, Hofmann F, main of the human guanylyl cyclase C receptor provides de Jonge HR: Differential role of cyclic GMP-dependent evidence for a catalytically active homotrimer. protein kinase II in ion transport in murine small intestine 39: 16075–16083, 2000 and colon. Gastroenterology 118: 108–114, 2000 25. Fan X, Wang Y, London RM, Eber SL, Krause WJ, Freeman 40. Forte LR, Thorne PK, Eber SL, Krause WJ, Freeman MJ, Ϫ RH, Forte LR: Signaling pathways for guanylin and Francis SH, Corbin JD: Stimulation of intestinal Cl trans- uroguanylin in the digestive, renal, central nervous, repro- port by heat-stable enterotoxin: Activation of cAMP-de- ductive, and lymphoid systems. Endocrinology 138: 4636– pendent protein kinase by cGMP. Am J Physiol 263: C607– 4648, 1997 C615, 1992 26. Range SP, Holland ED, Basten GP, Knox AJ: Regulation of 41. Chao AC, de Sauvage FJ, Dong YJ, Wagner JA, Goeddel guanosine 3Ј,5Ј-cyclic monophosphate in ovine tracheal DV, Gardner P: Activation of intestinal CFTR ClϪ channel epithelial cells. Br J Pharmacol 120: 1249–1254, 1997 by heat-stable enterotoxin and guanylin via cAMP-depen- 27. Schulz S, Chrisman TD, Garbers DL: Cloning and expres- dent protein kinase. EMBO J 13: 1065–1072, 1994 sion of guanylin its existence in various mammalian tis- 42. Crawford I, Maloney PC, Zeitlin PL, Guggino WB, Hyde sues. J Biol Chem 267: 16019–16021, 1992 SC, Turley H, Gatter KC, Harris A, Higgins CF: Immuno- 28. Vaandrager AB, Schulz S, de Jonge HR, Garbers DL: Gua- cytochemical localization of the cystic fibrosis gene prod- nylyl cyclase C is an N-linked glycoprotein receptor that uct CFTR. Proc Natl Acad Sci U S A 88: 9262–9266, 1991 accounts for multiple heat-stable enterotoxin-binding pro- 43. Ko SB, Zeng W, Dorwart MR, Luo X, Kim KH, Millen L, teins in the intestine. J Biol Chem 268: 2174–2179, 1993 Goto H, Naruse S, Soyombo A, Thomas PJ, Muallem S: J Am Soc Nephrol ●●: ●●●–●●●, ●●●● Cellular Effects of Guanylin and Uroguanylin 9

Gating of CFTR by the STAS domain of SLC26 transport- mRNA distribution along the rat nephron. Regul Pept 95: ers. Nat Cell Biol 6: 343–350, 2004 65–74, 2000 44. Nandi A, Bhandari R, Visweswariah SS: Epitope conserva- 59. Fujimoto S, Kinoshita H, Hara S, Nakazato M, Hisanaga S, tion and immunohistochemical localization of the guany- Eto T: Immunohistochemical localization of uroguanylin in lin/stable toxin peptide receptor, guanylyl cyclase C. J Cell the human kidney. Nephron 84: 88–89, 2000 Biochem 66: 500–511, 1997 60. Sindic A, Hirsch JR, Velic A, Piechota H, Schlatter E: Gua- 45. Qian X, Prabhakar S, Nandi A, Visweswariah SS, Goy MF: nylin and uroguanylin regulate electrolyte transport in Expression of GC-C, a receptor-guanylate cyclase, and its isolated human cortical collecting ducts. Kidney Int 67: endogenous ligands uroguanylin and guanylin along the 1420–1427, 2005 rostrocaudal axis of the intestine. Endocrinology 141: 3210– 61. Sindic A, Basoglu C, Cerci A, Hirsch JR, Potthast R, Kuhn 3224, 2000 M, Ghanekar Y, Visweswariah SS, Schlatter E: Guanylin, 46. Whitaker TL, Witte DP, Scott MC, Cohen MB: Uroguanylin uroguanylin and STa activate guanylate cyclase C and/or and guanylin: Distinct but overlapping patterns of messen- a PT-sensitive G protein in human proximal tubule cells. ger RNA expression in mouse intestine. Gastroenterology J Biol Chem 277: 17758–17764, 2002 113: 1000–1006, 1997 62. London RM, Eber SL, Visweswariah SS, Krause WJ, Forte 47. Albano F, Brasitus T, Mann EA, Guarino A, Giannella RA: LR: Structure and activity of OK-GC: A kidney receptor Colonocyte basolateral membranes contain E. coli heat- guanylate cyclase activated by guanylin peptides. Am J stable enterotoxin receptors. Bichem Biophys Res Commun Physiol 276: F882–F891, 1999 284: 331–334, 2001 63. Carrithers SL, Hill MJ, Johnson BR, O’Hara SM, Jackson 48. Ganguly U, Chaudhury AG, Basu A, Sen PC: STa-induced BA, Ott CE, Lorenz J, Mann EA, Giannella RA, Forte LR, translocation of protein kinase C from cytosol to mem- Greenberg RN: Renal effects of uroguanylin and guanylin brane in rat enterocytes. FEMS Microbiol Lett 204: 65–69, in vivo. Braz J Med Biol Res 32: 1337–1344, 1999 2001 64. Maack T, Suzuki M, Almeida FA, Nussenzveig D, Scarbor- 49. Weikel CS, Spann CL, Chambers CP, Crane JK, Linden J, ough RM, McEnroe GA, Lewicki JA: Physiological role of Hewlett EL: Phorbol esters enhance the cyclic GMP re- silent receptors of atrial natriuretic factor. Science 238: 675– sponse of T84 cells to the heat-stable enterotoxin of Esche- 678, 1987 richia coli (STa). Infect Immun 58: 1402–1407, 1990 65. Waldman SA, Rapoport RM, Murad F: Atrial natriuretic 50. Hamra FK, Eber SL, Chin DT, Currie MG, Forte LR: Reg- factor selectively activates particulate guanylate cyclase ulation of intestinal uroguanylin/guanylin receptor-medi- and elevates cyclic GMP in rat tissues. J Biol Chem 259: ated responses by mucosal acidity. Proc Natl Acad Sci U S A 14332–14334, 1984 94: 2705–2710, 1997 66. Macica CM, Yang Y, Hebert SC, Wang WH: Arachidonic 51. Lorenz JN, Nieman M, Sabo J, Sanford P, Hawkins JA, acid inhibits activity of cloned renal Kϩ channel, ROMK1. Elitsur N, Gawenis LR, Clarke LL, Cohen MB: Uroguanylin Am J Physiol Renal Physiol 271: F588–F594, 1996 knockout mice have increased blood pressure and im- 67. Greenberg RN, Hill MJ, Crytzer J, Krause WJ, Eber SL, paired natriuretic response to enteral NaCl load. J Clin Hamra FK, Forte LR: Comparison of effects of uroguany- Invest 122: 1244–1254, 2003 lin, guanylin and Escherichia coli heat-stable enterotoxin 52. Kinoshita H, Fujimoto S, Nakazato M, Yokota N, Date Y, STa in mouse intestine and kidney: Evidence that urogua- Yamaguchi H, Hisanaga S, Eto T: Urine and plasma levels nylin is an intestinal natriuretic hormone. J Invest Med 45: of uroguanylin and its molecular form in renal diseases. 276–283, 1997 Kidney Int 52: 1028–1034, 1997 68. Furuya S, Naruse S, Ando E, Nikohara K, Hayakawa T: 53. Fukae H, Kinoshita H, Fujimoto S, Kita T, Nakazato M, Eto Effect and distribution of intravenously injected 125I-gua- T: Changes in urinary levels and renal expression of nylin an rat kidney examined by high-resolution light mi- uroguanylin on low or high salt diets in rats. Nephron 92: croscopic radioautography. Anat Embryol 196: 185–193, 373–378, 2002 1997 54. Potthast R, Ehler E, Scheving LA, Sindic A, Schlatter E, 69. John M, Wiedenmann B, Hruhoffer M, Adermann K, Kuhn M: High salt intake increases uroguanylin expres- Ankorina-Stark I, Schlatter E, Ahnert-Hilger G, Forssmann sion in mouse kidney. Endocrinology 142: 3087–3097, 2001 WG, Kuhn M: Guanylin stimulates regulated secretion 55. Fan X, Hamra FK, London RM, Eber SL, Krause WJ, Free- from human neuroendocrine pancreatic cells. Gastroenter- man RH, Smith CE, Currie MG, Forte LR: Structure and ology 114: 791–797, 1998 activity of uroguanylin and guanylin from the intestine 70. Kulaksiz H, Schmid A, Honscheid M, Eissele R, Klemp- and urine of rats. Am J Physiol 273: E957–E964, 1997 nauer J, Cetin Y: Guanylin in the human pancreas: A novel 56. Hamra FK, Fan X, Krause WJ, Freeman RH, Chin DT, luminocrine regulatory pathway of electrolyte secretion Smith CE, Currie MG, Forte LR: Prouroguanylin and via cGMP and CFTR in the ductal system. Histochem Cell proguanylin: Purification from colon, structure, and mod- Biol 115: 131–145, 2001 ulation of bioactivity by proteases. Endocrinology 137: 257– 71. Kulaksiz H, Cetin Y: Uroguanylin and guanylate cyclase C 265, 1996 in the human pancreas: Expression and mutuality of li- 57. Sindic A, Velic A, Basoglu C, Hirsch JR, Edemir B, Kuhn M, gand/receptor localization as indicators of intracellular Schlatter E: Uroguanylin and guanylin regulate transport paracrine signaling pathways. J Endocrinol 170: 267–275, of mouse cortical collecting duct independent of guanylate 2001 cyclase C. Kidney Int 68: 1008–1017, 2005 72. Spreca A, Simonetti S, Rambotti MG: Atrial natriuretic 58. Carrithers SL, Taylor B, Cai WY, Johnson BR, Ott CE, peptide and guanylin-activated guanylate cyclase isoforms Greenberg RN, Jackson BA: Guanylyl cyclase-C receptor in human sweat glands. Histochem J 32: 725–731, 2000 10 Journal of the American Society of Nephrology J Am Soc Nephrol ●●: ●●●–●●●, ●●●●

73. Reinecke M, David I, Loffing-Cueni D, Ablinger P, Cetin Y, 80. Suhasini M, Li H, Lohmann SM, Boss GR, Pilz RB: Cyclic- Kuhn M, Forssmann WG: Localization, expression and GMP-dependent protein kinase inhibits the ras/mitogen- characterization of guanylin in the rat adrenal medulla. activated protein kinase pathway. Mol Cell Biol 18: 6983– Histochem Cell Biol 106: 367–374, 1996 6994, 1998 74. Jaleel M, London RM, Eber SL, Forte LR, Visweswariah SS: 81. Steinbrecher KA, Wowk SA, Rudolph JA, Witte DP, Cohen Expression of the receptor guanylyl cyclase C and its li- MB: Targeted inactivation of the mouse guanylin gene gands in reproductive tissues of the rat: A potential role for results in altered dynamics of colonic epithelial prolifera- a novel signaling pathway in the epididymis. Biol Reprod tion. Am J Pathol 161: 2169–2178, 2002 67: 1975–1980, 2002 82. Nakazato M, Yamaguchi H, Shiomi K, Date Y, Fujimoto S, 75. Steinbrecher KA, Tuohy TMF, Goss KH, Scott MC, Witte Kangawa K, Matsuo H, Matsukura S: Identification of 10- DP, Groden J, Cohen MB: Expression of guanylin is down kDa proguanylin as a major guanylin molecule in human regulated in mouse and human intestinal adenomas. intestine and plasma and its increase in renal insufficiency. Bichem Biophys Res Commun 273: 225–230, 2000 Biochem Biophys Res Commun 205: 1966–1975, 1994 76. Cohen MB, Hawkins JA, Witte DP: Guanylin mRNA ex- 83. Nakazato M, Yamaguchi H, Kinoshita H, Kangawa K, pression in human intestine and colorectal adenocarci- Matsuo H, Chino N, Matsukura S: Identification of biolog- noma. Lab Invest 78: 101–108, 1998 ically active and inactive human uroguanylins in plasma 77. Shailubhai K, Yu HH, Karunanandaa K, Wang JY, Eber SL, and urine and their increases in renal insufficiency. Bio- Wang Y, Joo NS, Kim HD, Miedema BW, Abbas SZ, chem Biophys Res Commun 220: 586–593, 1996 Boddupalli SS, Currie MG, Forte LR: Uroguanylin treat- ment suppresses polyp formation in the Apc(Min/ϩ) 84. Kinoshita H, Fujimoto S, Fukae H, Yokota N, Hisanaga S, mouse and induces apoptosis in human colon adenocarci- Nakazato M, Eto T: Plasma and urine levels of uroguanylin noma cells via cyclic GMP. Cancer Res 60: 5151–5157, 2000 a new peptide in nephrotic syndrome. Nephron 81: 160– 78. Pitari GM, Di Guglielmo MD, Park J, Schulz S, Waldman 164, 1999 SA: Guanylyl cyclase C agonists regulate progression 85. Fukae H, Kinoshita H, Fujimoto S, Nakazato M, Eto T: through the cell cycle of human colon carcinoma cells. Proc Plasma concentration of uroguanylin in patients on main- Natl Acad Sci U S A 98: 7846–7851, 2001 tenance dialysis therapy. Nephron 84: 206–210, 2000 79. Liu L, Li H, Underwood T, Lloyd M, David M, Sperl G, 86. Kikuchi M, Fujimoto S, Fukae H, Kinoshita H, Kita T, Pamukcu R, Thompson WJ: Cyclic GMP-dependent protein Nakazato M, Eto T: Role of uroguanylin, a peptide with kinase activation and induction by exisulind and CP461 in natriuretic activity, in rats with experimental nephrotic colon tumor cells. J Pharmacol Exp Ther 299: 583–592, 2001 syndrome. J Am Soc Nephrol 16: 392–397, 2005