当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2005年第12期 > 正文
编号:11411125
Minireview: Aquaporin 2 Trafficking
http://www.100md.com 《内分泌学杂志》
     Department of General and Environmental Physiology (G.V., G.P., G.T., M.C., M.S.) and Centro di Eccellenza in Genomica Comparata (G.V., M.S.), University of Bari, 70126 Bari, Italy

    Abstract

    In the kidney aquaporin-2 (AQP2) provides a target for hormonal regulation of water transport by vasopressin. Short-term control of water permeability occurs via vesicular trafficking of AQP2 and long-term control through changes in the abundance of AQP2 and AQP3 water channels. Defective AQP2 trafficking causes nephrogenic diabetes insipidus, a condition characterized by the kidney inability to produce concentrated urine because of the insensitivity of the distal nephron to vasopressin. AQP2 is redistributed to the apical membrane of collecting duct cells through activation of a cAMP signaling cascade initiated by the binding of vasopressin to its V2-receptor. Protein kinase A-mediated phosphorylation of AQP2 has been proposed to be essential in regulating AQP2-containing vesicle exocytosis. Cessation of the stimulus is followed by endocytosis of the AQP2 proteins exposed on the plasma membrane and their recycling to the original stores, in which they are retained. Soluble N-ethylmaleimide sensitive fusion factor attachment protein receptors (SNARE) and actin cytoskeleton organization regulated by small GTPase of the Rho family were also proved to be essential for AQP2 trafficking. Data for functional involvement of the SNARE vesicle-associated membrane protein 2 in AQP2 targeting has recently been provided. Changes in AQP2 expression/trafficking are of particular importance in pathological conditions characterized by both dilutional and concentrating defects. One of these conditions, hypercalciuria, has shown to be associated with alteration of AQP2 urinary excretion. More precisely, recent data support the hypothesis that, in vivo external calcium, through activation of calcium-sensing receptors, modulates the expression/trafficking of AQP2. Together these findings underscore the importance of AQP2 in kidney pathophysiology.

    Introduction

    A MAJOR BREAKTHROUGH in understanding how the kidney regulates water transport was the discovery that vasopressin stimulates the translocation of a specific water channel protein, aquaporin-2 (AQP2), from intracellular vesicles to the cell surface. Elucidating how this process is regulated has remained a challenge because it represents a convergence of two different and complex fields of research, namely vesicle transport and signal transduction. AQP2 protein possesses the typical aquaporin fold observed in the aquaporin water channel family members, as recently confirmed by crystallization and structural characterization by atomic force microscopy (1).

    The trafficking of the vasopressin-sensitive water channel AQP2, reviewed in several excellent publications (2, 3, 4, 5, 6, 7, 8), is the paradigm of how cells control the movement of membrane proteins through complex pathways in response to external stimuli and how, by doing so, they regulate their function. In response to an increase in vasopressin levels, AQP2, intracellularly localized in resting collecting duct principal cells, becomes exposed on the cell surface on which it facilitates water reabsorption and consequently urine concentration. Cessation of the stimulus is followed by endocytosis of the AQP2 proteins by a clathrin-mediated mechanism (3, 9, 10) and their recycling to the original stores, in which they are retained. These complex mechanisms include the spatiotemporal modulation of several regulatory proteins. The role of some accessory proteins such as soluble N-ethylmaleimide sensitive fusion factor attachment protein receptors (SNARE) proteins and small GTPases in AQP2 shuttle has been recently clarified. Moreover, actin cytoskeleton remodeling regulated by small GTPase of the Rho family were also proved to be essential for AQP2 trafficking.

    In addition to a vasopressin-regulated pathway, it has been shown that AQP2 recycles constitutively in epithelial cells, and it can be inserted in different membrane domains. In epithelial cell lines, AQP2 has been shown to be targeted to the apical or basolateral surface, although the polarity signals are still to be clarified (3). Osmolality environment can influence the final membrane destination. Madin-Darby canine kidney cell growth in a hypertonic medium showed a basolateral localization of AQP2 on hormonal stimulation (11). Moreover, hypertonicity has been proved to modulate AQP2 expression in immortalized mouse collecting duct principal cells (12).

    On the other hand, maintaining AQP2 membrane compartment identity and fusion specificity is of critical importance because mislocalization or promiscuous fusion of AQP2 vesicles could compromise vasopressin responsivity.

    Defective AQP2 trafficking can lead to severe diseases like congestive heart failure, liver cirrhosis, and nephrogenic diabetes insipidus (NDI), a condition characterized by the kidney’s inability to produce concentrated urine due to the insensitivity of the distal nephron to vasopressin (2, 13, 14, 15, 16). NDI can be acquired or congenital. Congenital NDI can be due to either mutation of the V2 receptor gene or mutations in AQP2 gene. To date, 30 mutations in the AQP2 gene have been reported (17, 18, 19).

    The acquired forms of NDI may occur at any time in life and are associated with urinary concentrating defect due to decreased expression of AQP2 or impaired targeting of these channels to the apical plasma membrane. Many of these forms are reversible and secondary to drug treatment and metabolic disturbances. For example, studies in rats have shown the reversible down-regulation of AQP2 after treatment with lithium and hypokalemia (20, 21). However, patients treated with lithium with an average exposure of 8 yr recovered only in a very small percentage, whereas most failed to subside after lithium cessation (16).

    Whereas many aspects of AQP2 shuttle have been clarified in more than 10 yr of investigation, several features are still unclear.

    This brief review discusses recent advances in some selected aspects of this research field.

    Role of Phosphorylation in Regulated AQP2 Exocytosis

    Binding of vasopressin to V2 receptors on the basolateral surface of principal cells stimulates the G protein Gs, which activates adenyl cyclase. The resulting increase in intracellular cAMP leads to activation of protein kinase A (PKA) that phosphorylates many substrates including the C-terminal Ser-256 of AQP2.

    It has to be underlined that the AQP2 primary structure exhibits several potential consensus sequences for at least five kinases, namely PKA, protein kinase C (PKC), protein kinase G, casein kinase II and Golgi casein kinase. To date, however, the only site that has been demonstrated to be involved in AQP2 trafficking is Ser-256. This was first investigated by Kuwahara et al. (22) in 1995 using site-directed mutagenesis. In this study, the author demonstrated that oocytes expressing wild-type, but not Ser-256-mutated, AQP2 responded to cAMP injection by increasing membrane water permeability (Pf). In vitro phosphorylation studies also showed that mutating Ser-256 prevented AQP2 phosphorylation, indirectly implicating this site for PKA-mediated phosphorylation.

    Whether AQP2 phosphorylation can regulate channel osmotic Pf was addressed by Lande et al. (23), who showed that Pf of endosomes containing phosphorylated or dephosphorylated AQP2 was not significantly different, suggesting that phosphorylation regulate AQP2 accumulation to the plasma membrane rather than gating the channel. Stopped-flow light-scattering in LLC-PK1 cells transfected with wild-type or S256A AQP2 demonstrated that Ser-256 substitution impaired the surface expression of AQP2 in response to forskolin (24). Moreover, phosphorylation experiments in intact cells demonstrated significant phosphorylation of wild-type AQP2 and only minimal phosphorylation of S256A AQP2 (24).

    In a separate study, Katsura et al. (25) showed that vasopressin-induced membrane insertion of AQP2 was completely inhibited by pretreatment of AQP2-expressing LLC-PK1 cells with N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide dihydrochloride (H89).

    Studies using intact rat renal tissue confirmed data obtained using cultured renal cells. Physiological serum levels of vasopressin were able to induce a time- and dose-dependent increase in AQP2 phosphorylation, and the same effect was obtained using the vasopressin V2 receptor agonist 1-desamino-8-D-arginine vasopressin or forskolin (26). Moreover, phosphorylation state-specific antiserum revealed an increase in AQP2 phosphorylation at Ser-256 after incubation of renal tissue with arginine vasopressin (AVP). Using the same antibody as a tool to determine the subcellular localization of phosphorylated AQP2 (p-AQP2), it was shown that a consistent amount of the water channel, localized in both the plasma membrane and intracellular vesicles, is constitutively phosphorylated, even in low circulating vasopressin states (27). Because AQP2 is expressed at the plasma membrane as a tetramer, coinjection in oocytes of different ratios of AQP2-S256D and AQP2-S256A mimicking p-AQP2 and non-p-AQP2, respectively, demonstrated that at least three monomers in an AQP2 tetramer should be phosphorylated for steady-state plasma membrane localization (28).

    The possible role of the other putative kinase sites in the trafficking and hormonal regulation of AQP2 has been investigated by van Balkom et al. (29). Three putative casein kinase II (Ser-148, Ser-229, Thr-244), one protein kinase C [PKC (Ser-231)], and one PKA (Ser-256) site were mutated to mimic a constitutively AQP2 nonphosphorylated/phosphorylated state and expressed in Madin-Darby canine kidney cells (29). The subcellular localization and the forskolin responsiveness of all the proteins, except for the Ser-256 mutants, were no different from wild-type AQP2. Not surprisingly, AQP2-S256A was retained in intracellular vesicles, and AQP2/S256D was localized to the plasma membrane. Interestingly though, PKC activation after forskolin treatment, increased the endocytosis of all the mutants (including AQP2/S256D), without affecting phosphorylation at Ser-256. The authors concluded that phosphorylation of Ser-256 is necessary and sufficient for expression of AQP2 in the apical membrane, and PKC-mediated endocytosis of AQP2 is independent of the AQP2 phosphorylation state (29).

    In an apparent contradiction to these findings, Valenti et al. (30) demonstrated that AQP2 fusion with the plasma membrane might occur even in the absence of phosphorylation by PKA. Using the phosphatase inhibitor okadaic acid in an AQP2-transfected renal cell line, the author demonstrated that by increasing the phosphorylation state of intracellular proteins, it is possible to induce translocation of AQP2 and increase Pf independent of AQP2 phosphorylation state, even in the presence of the PKA inhibitor H89 (30). AQP2 phosphorylation, therefore, may not be responsible solely for water channel translocation in renal cells, and other pathways may exist to account for AQP2 membrane translocation. In agreement with this finding, it has been shown that AQP2 recycles constitutively and rapidly between intracellular storage compartments and the cell surface in renal cells using a phosphorylation-independent process (31).

    Another signaling pathway that leads to AQP2 membrane insertion was demonstrated by Bouley et al. (32) in both transfected epithelial cells and kidney collecting duct. In this pathway, elevation of intracellular cGMP levels induced AQP2 redistribution, probably through a protein kinase G-dependent AQP2 phosphorylation at Ser-256 (32). This cGMP-mediated process does not involve V2 receptor signaling and intracellular cAMP elevation. More recently the same group showed that the cGMP phosphodiesterase type 5 inhibitor, sildenafil citrate (Viagra), caused plasma membrane accumulation of AQP2 mimicking vasopressin effect in LLC-PK1 cells (33). The results of this study provide the backbone for a promising approach in the treatment of several forms of NDI characterized by V2 receptor dysfunction. In a recent work (34), AQP2 phosphorylation dynamics during maturation from the endoplasmic reticulum to the vasopressin regulated compartment have been reported. Interestingly, AQP2 transition in the Golgi apparatus was associated with a PKA-independent increase in AQP2 phosphorylation at Ser-256, possibly accounted for by the activity of a Golgi-localized serine kinase, the Golgi casein kinase (34). According to this hypothesis, the phosphorylation-defective E258K AQP2 mutant causing a dominant form of NDI would be routed from Golgi to lysosomes for degradation (34).

    Despite this plethora of experimental evidence indicating phosphorylation as a key event regulating different steps in AQP2 intracellular trafficking, nobody has so far demonstrated a direct interaction of p-AQP2 with any motor or regulatory proteins. That would be the challenge of investigators in the field in the years to come.

    PKA Anchoring Proteins (AKAPs)

    Compartmentalization of signaling proteins through association with anchoring proteins ensures specificity in signal transduction. In this respect, the phosphorylation by PKA and also the tethering of PKA by AKAPs to subcellular compartments are prerequisites for AQP2 translocation. In IMCD cells, AQP2 trafficking required anchoring of PKA to AKAPs. Preincubation of cells with the synthetic peptide, S-Ht31, which prevents the binding between AKAPs and the regulatory subunit of PKA, significantly impaired hormonal-dependent AQP-2 trafficking (5, 35, 36). Klussman et al. (36) cloned and sequenced the full length of the rat ortholog Ht31, showing that it not only contains the anchoring domain but also directly interacts with RhoA in vivo, thus integrating PKA and RhoA, both of which are involved in the vasopressin-dependent water reabsorption.

    A new splice variant of AKAP18, AKAP18, has been found to be colocalized with AQP2 in IMCD cells, and increases in cAMP produced the dissociation of AKAP18 and PKA, suggesting that AKAP18 plays a role during AQP2 trafficking (37).

    Cytoskeletal Dynamics and Rho Signaling

    Cytoskeleton remodeling at the cell cortex participates in several cellular processes including endocytosis and exocytosis. In amphibian bladder vasopressin-induced osmotic water flow decreases on colchicines and nocodazole treatment, indicating that microtubule might play a role in the regulation of water permeability (38, 39, 40). Moreover, it has been reported that the driving force to promote intracellular trafficking along microtubules is provided from several motor proteins like dynactin and dynein, which have been found in immunoisolated AQP2-bearing vesicles (41).

    Actin cytoskeletal dynamics have also been shown to be essential in the trafficking of AQP2-bearing vesicles. It was already known from previous studies that in mammalian collecting duct and in the amphibian bladder vasopressin increased the apical sorting of water channels was associated with depolymerization of the actin cytoskeleton (42, 43, 44). Consistently, in toad bladder epithelial cells, cytochalasin D, in the presence of vasopressin, significantly increased the fusion rate of water channel-bearing vesicles, compared with vasopressin alone, likely indicating that actin filaments might retard fusion processes (45, 46). On the other hand, in renal collecting duct CD8 cells (47), okadaic acid, a specific phosphatase inhibitor, caused actin depolymerization at the cell cortex and increase in the osmotic water transport (30). Recent studies highlighted the importance of the small GTPase Rho as a key player regulating AQP2 trafficking through regulation of the actin network (48, 49). RhoA inhibition occurring in response to forskolin stimulation represents a physiological step in the signal transduction cascade activated by hormonal stimulation leading to a partial depolymerization of the actin cytoskeleton, which facilitates the apical targeting of AQP2 (50). Consistent with these observations, Rho-dependent actin polymerization observed either on prostaglandin EP3 receptor activation (51) or by selective activation of the sensing calcium receptor (CaR) significantly impaired vasopressin-stimulated AQP2 trafficking (52). These data indicate that RhoA exerts a bidirectional control in the regulation of AQP2 trafficking though the regulation of actin network organization.

    Ezrin-Radixin-Moesin (ERM) Protein Involvement in AQP2 Trafficking

    ERM proteins cross-link actin filaments with plasma membrane proteins and are important integrators at the cell cortex, where they participate in the reorganization of F-actin-containing cytoskeletal structures.

    Rho activity is controlled by several regulators, and ERM proteins function both upstream and downstream of Rho GTPases, implying that there could be a feedback loop for Rho pathway autoregulation (36, 53, 54, 55). Recent data have provided the first evidence that AQP2 translocation to the plasma membrane requires the functional involvement of the ERM protein moesin (56). ERM may represent scaffolding proteins mediating AQP2 interaction with actin. Interestingly, immunoaffinity chromatography experiments in rat kidney papilla revealed that AQP2 interacts with not only several actin binding proteins but also - and -isoforms of actin (57, 58). Proteomic analysis of immunoisolated AQP2-bearing vesicles from renal inner medullary collecting duct suggested the existence of a dynamic complex including myosin IC, nonmuscle myosin IIA and IIB, myosin VI, and myosin IXB involved in the generation of force necessary to promote AQP2 trafficking to the plasma membrane (59). More recently Chou et al. (60) showed that Pf is partially due to the activation of calcium/calmodulin-dependent myosin light chain kinase. Moreover, consistent with these observations, AQP2-containing exosomes isolated from human urine also expressed several cytoskeletal proteins like ERM, many isoforms of actin, cofilin, dynein, and tubulin (61).

    Role of Calcium in Vasopressin-Stimulated AQP2 Translocation

    Studies of a wide variety of vesicular-trafficking processes have pointed to a key role for local increases in intracellular calcium concentration ([Ca2+]i) in triggering the fusion of vesicles to their target membranes (62, 63, 64). Whether vasopressin stimulated exocytosis of AQP2-containing vesicles in collecting duct cells requires calcium as an intracellular mediator is still unclear. Vasopressin, acting via V2 receptors, causes a transient increase in [Ca2+]i and calcium oscillations in IMCD cells (65, 66, 67, 68, 69, 70). Buffering of intracellular calcium with bis-(o-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid(BAPTA) completely blocked the Pf response to vasopressin in IMCD (65, 68), suggesting that intracellular calcium mobilization is required for exocytotic insertion of AQP2. Removal of extracellular Ca2+ in perfused IMCD did not prevent the initial rise [Ca2+]i induced by vasopressin but inhibited the sustained oscillations (65). These observations indicate that vasopressin induces calcium release from intracellular stores, but extracellular calcium is required to sustain the [Ca2+]i oscillations likely needed to prolong the exocytotic activity induced by vasopressin. Little is known about the mechanism of the vasopressin-induced increase in intracellular Ca2+, although previous studies established that it occurs in the absence of activation of the phosphoinositide signaling pathway (71). In primary cultures of IMCD cells, ryanodine, a ryanodine receptor antagonist, inhibits vasopressin-dependent redistribution of AQP2 to the plasma membrane, suggesting a role for ryanodine-sensitive stores in the calcium-dependent AQP2 trafficking (65, 68).

    However, reports from other authors do not support a regulatory function for calcium in AQP2 insertion into the plasma membrane (72). By analysis of endocytic capacitance in primary cultured epithelial cells from renal inner medulla, these authors showed that cAMP is sufficient for triggering the exocytic recruitment of AQP2, which is not evoked by vasopressin-induced intracellular calcium increases.

    SNAREs for Directing Vesicular Trafficking of AQP2

    Fusion of AQP2 vesicles with the plasma membrane is a key terminal step in vasopressin-regulated water transport. This fusion event is possibly mediated by SNARE proteins through a process regulated by accessory proteins whose roles are still unclear. SNARE proteins have been detected in the collecting duct principal cells and colocalize with AQP2-bearing vesicles (73, 74). In particular, syntaxin 4 is present in the apical plasma membrane of collecting duct principal cells (75, 76), and soluble N-ethylmaleimide-associated protein-23 (SNAP23) has also been found in collecting duct principal cells both in the apical plasma membrane and AQP2-bearing vesicles (77). SNAP23 has been demonstrated to interact with syntaxin-4 and vesicle-associated membrane protein 2 (VAMP2) (78). However, another study, using different antibodies, reported an opposite polarity for sintaxin-4 in principal cells (79).

    The functional involvement of VAMP2 in AQP2 targeting came from the demonstration that tetanus neurotoxin was efficient in cleaving the synaptobrevin-like protein in intact collecting duct renal cells, and cAMP-stimulated AQP2 targeting to the plasma membrane was completely abolished in treated cells (80). This represents the first evidence for the functional requirement of VAMP2 in cAMP-induced AQP2 exocytosis in renal cells. The identification and functional involvement of the counterpart VAMP2 proteins is still lacking.

    Other Accessory Proteins Involved in the AQP2 Shuttle

    Heterotrimeric GTP binding proteins of the Gi family has also been proposed to be essential for AQP2 targeting (81). Treatment of cultured rabbit cortical collecting duct cells with pertussis toxin or incubation of permeabilized cells with synthetic peptides reproducing the COOH terminus of the Gi-3 subunit abolished cAMP-induced AQP2 trafficking to the cell apical plasma membrane. This leads to the conclusion that Gi-3 plays a facilitatory role in AQP2 targeting. On the other hand, it has also been proposed that proteins of the Gi family impair AQP2 translocation to the plasma membrane via activation of the calcium-sensing receptor (82). The precise functional role of this G protein in AQP2 trafficking remains to be clarified.

    Very recently Noda et al. (58, 83) identified two AQP2 binding proteins, actin and signal-induced proliferation-associated protein 1 (SPA-1). SPA-1 is a specific GTPase-activating protein for Rap1, whose activity was shown to be required for AQP2 trafficking to the apical membrane.

    Attenuation of Vasopressin 2 Receptor Signaling

    Luminal calcium

    Recent data support the hypothesis that high calcium levels (>2 mM) may act as a negative regulator of V2 receptor signaling in the kidney. Calcium plays a role in AQP2-bearing vesicles trafficking not only at second messenger level but also as a first messenger acting through a specific G protein-coupled receptor. Purified AQP2 endosomes contain a CaR (82) similar to that identified in both the parathyroid gland and renal thick ascending limb (TAL) (84, 85). CaRs have been shown to sense the extracellular concentration of divalent cations, including Ca2+ or Mg2+ (84, 86, 87) and activate various signal transduction pathways in multiple cell types (88, 89). Although little significant transepithelial transport of Ca2+ or Mg2+ occurs in IMCD, the luminal concentrations of these divalent cations undergo significant alterations as a result of vasopressin elicited water reabsorption. Studies in isolated perfused rat IMCD show that apical membrane exposure to CaR agonists rapidly reduces AVP-elicited Pf without an alteration in P urea (82). In addition, during sustained hypercalcemia in chronically hypercalcemic rats, the Pf in inner medullary collecting duct did not increase significantly after vasopressin and was accompanied by an 87% reduction in AQP2 protein (90), which may contribute to the lack of vasopressin responsiveness. Moreover it has been observed that extracellular calcium acting through the endogenous CaR, antagonizes forskolin-induced AQP2 translocation to the apical plasma membrane in renal AQP2 expressing CD8 cells (52).

    The signal transduction pathways linking urinary calcium levels to AQP2 expression/trafficking in the collecting duct is poorly understood. A Gi-coupled receptor activation signaling may mediate the extracellular calcium effects trough CaR (91, 92, 93). In some cells expressing CaR, CaR agonists can inhibit cAMP accumulation through a mechanism involving a CaR-induced increase in [Ca2+]i, which then inhibits a calcium-sensitive form of adenylate cyclase (94). Alternatively, a Ca2+-dependent increase in [Ca2+]i can impair cAMP accumulation by an increase in cAMP hydrolysis through a Ca2+-mediated process (95, 96). Another intriguing possibility is that CaR-mediated Rho activation (97) induces stabilization of cortical F-actin cytoskeleton that impairs AQP2 targeting, as previously suggested (49, 50). Thus, the apical membrane CaR may provide a mechanism for the modulation of vasopressin-elicited water transport in IMCD by altering AQP2 vesicle apical membrane retrieval. The presence of this apical CaR permits the IMCD cell to continuously integrate AVP water transport with alterations in luminal Ca2+ preventing further calcium concentration and thus protecting against a potential risk of calcium oxalate- or calcium phosphate-containing kidney stone formation.

    Prostaglandin

    In the kidney, prostaglandin E2 (PGE2) is involved in the regulation of epithelial solute and water reabsorption (98). PGE2 and its synthetic analog sulprostone are known to antagonize vasopressin-mediated water reabsorption. It has been reported that PGE2 stimulates internalization of AQP2 from the plasma membrane of principal cells when added after vasopressin treatment without affecting its phosphorylated state (99). In IMCD cells, preincubation of the cells with sulprostone, a synthetic analog of PGE2, inhibited the effect of vasopressin on AQP2 translocation independently of inositol-1,4,5-trisphosphate or [Ca2+]i (51) (Fig. 1). AQP2 down-regulation is also present in long-standing urinary obstruction and persists even after the obstruction is resolved. Quite interestingly in humans, AQP2 urinary excretion in postobstructed kidney was associated with a local increase in PGE2 synthesis, which may be, in part, responsible for selective down-regulation of AQP2 trafficking/expression (100).

    Bradykinin (BK)

    The nonapeptide BK is a known stimulator of prostaglandin production in the kidney through activation of phospholipase A2, which stimulates AA release. In a rabbit collecting duct cell line expressing endogenous BK2R receptors, BK inhibited deoxy-1-desamino-8-D-arginine vasopressin-dependent cAMP production. This event was mimicked by PGE2 and suppressed with indomethacin, suggesting that the signal transduction initiated by BK included PGE2 synthesis (101). However, in the same cells stably transfected with AQP2, recent data indicate that BK causes an increase of Rho activity associated with stabilization of cortical F-actin network, thus impairing AQP2 targeting (102). These effects counteract the physiological vasopressin stimulation. This indicates that BK may directly contribute to a decrease in the kidney concentrating ability impairing cell surface expression of the vasopressin sensitive AQP2 water channel (Fig. 1).

    Body Fluid Regulation in Microgravity, Involvement of AQP2

    Continuous sodium retention not being followed by water has been observed in astronauts (103). This is in agreement with the persistent plasma volume deficit occurring in microgravity despite the continuously activated counterregulatory endocrine and autonomous nervous systems. As discussed above, extracellular CaRs might sense alterations of calcium concentration in the collecting duct lumen and in turn activate a signal transduction cascade influencing AQP2 trafficking/expression. Because astronauts, in addition to their water-handling disorder, experience hypercalciuria during their presence aboard space vehicles, it has been hypothesized that the impaired water handling occurring in astronauts during space flight, is due to an attenuated AQP2 response to vasopressin as a consequence of persistent hypercalciuria (104, 105). The link between hypovolemia-induced activation of fluid retaining systems and reduction in bone mineral density, promoting hypercalciuria during space flight, with its postulated modulating effect on renal water and sodium handling, shall stimulate space physiology research during real space flight and on the ground.

    Concluding Remarks

    After the discovery of the vasopressin-sensitive AQP2 water channel, significant progress has been made in understanding the molecular basis of AQP2 trafficking between intracellular vesicles and the cell surface. These events are regulated by hormone-induced protein phosphorylation and involve the interaction and/or functional involvement of several regulatory proteins as GTP binding proteins, cytoskeletal elements, ERM, and SNAREs. Additional features of AQP2 shuttle have emerged recently related to intracellular pathways activated by physiological molecules such as prostaglandins, bradykinin, and divalent cations like calcium that counteract vasopressin response (Fig. 1). These analyses provide insight into the physiology and pathophysiology of water balance leading to possible therapeutic intervention for water balance disorders including NDI, heart failure, and hypertension.

    Acknowledgments

    We thank all colleagues who contributed to these studies.

    Footnotes

    This work was supported by the "Ministero della Ricerca Scientifica e Tecnologica," and the CEGBA (Centro di Eccellenza di Genomica in Campo Biomedico ed Agrario), and Laboratorio Analisi del Gene.

    First Published Online September 8, 2005

    Abbreviations: AKAP, PKA anchoring protein; AQP2, aquaporin-2; AVP, arginine vasopressin; BK, bradykinin; [Ca2+]i, intracellular calcium concentration; CaR, calcium receptor; ERM, ezrin-radixin-moesin; NDI, nephrogenic diabetes insipidus; p-AQP2, phosphorylated AQP2; Pf, osmotic water permeability coefficient; PGE2, prostaglandin E2; PKA, protein kinase A; PKC, protein kinase C; SNARE, soluble N-ethylmaleimide sensitive fusion factor attachment protein receptor; VAMP2, vesicle-associated membrane protein.

    Accepted for publication August 5, 2005.

    References

    Schenk AD, Werten PJ, Scheuring S, de Groot BL, Muller SA, Stahlberg H, Philippsen A, Engel A 2005 The 4.5A structure of human AQP2. J Mol Biol 350:278–289

    Noda Y, Sasaki S 2004 Molecular mechanisms and drug development in aquaporin water channel diseases: molecular mechanism of water channel aquaporin-2 trafficking. J Pharmacol Sci 96:249–254

    Brown D 2003 The ins and outs of aquaporin-2 trafficking. Am J Physiol Renal Physiol 284:F893–F901

    Nielsen S, Frokiaer J, Marples D, Kwon TH, Agre P, Knepper MA 2002 Aquaporins in the kidney: from molecules to medicine. Physiol Rev 82:205–244

    Klussmann E, Maric K, Rosenthal W 2000 The mechanisms of aquaporin control in the renal collecting duct. Rev Physiol Biochem Pharmacol 141:33–95

    Agre P, Nielsen S 1996 The aquaporin family of water channels in kidney. Nephrologie 17:409–415

    Knepper MA 1997 Molecular physiology of urinary concentrating mechanism: regulation of aquaporin water channels by vasopressin. Am J Physiol 272:F3–F12

    Tajika Y, Matsuzaki T, Suzuki T, Ablimit A, Aoki T, Hagiwara H, Kuwahara M, Sasaki S, Takata K 2005 Differential regulation of AQP2 trafficking in endosomes by microtubules and actin filaments. Histochem Cell Biol:1–12

    Sun TX, Van Hoek A, Huang Y, Bouley R, McLaughlin M, Brown D 2002 Aquaporin-2 localization in clathrin-coated pits: inhibition of endocytosis by dominant-negative dynamin. Am J Physiol Renal Physiol 282:F998–F1011

    Katsura T, Ausiello DA, Brown D 1996 Direct demonstration of aquaporin-2 water channel recycling in stably transfected LLC-PK1 epithelial cells. Am J Physiol 270:F548–F553

    van Balkom BW, van Raak M, Breton S, Pastor-Soler N, Bouley R, van der Sluijs P, Brown D, Deen PM 2003 Hypertonicity is involved in redirecting the aquaporin-2 water channel into the basolateral, instead of the apical, plasma membrane of renal epithelial cells. J Biol Chem 278:1101–1107

    Hasler U, Vinciguerra M, Vandewalle A, Martin PY, Feraille E 2005 Dual effects of hypertonicity on aquaporin-2 expression in cultured renal collecting duct principal cells. J Am Soc Nephrol 16:1571–1582

    Knoers NV, Deen PM 2001 Molecular and cellular defects in nephrogenic diabetes insipidus. Pediatr Nephrol 16:1146–1152

    Oksche A, Rosenthal W 1998 The molecular basis of nephrogenic diabetes insipidus. J Mol Med 76:326–337

    Morello JP, Bichet DG 2001 Nephrogenic diabetes insipidus. Annu Rev Physiol 63:607–630

    Garofeanu CG, Weir M, Rosas-Arellano MP, Henson G, Garg AX, Clark WF 2005 Causes of reversible nephrogenic diabetes insipidus: a systematic review. Am J Kidney Dis 45:626–637

    Bichet DG 1998 Nephrogenic diabetes insipidus. Am J Med 105:431–442

    Knoers N, van den Ouweland A, Dreesen J, Verdijk M, Monnens LA, van Oost BA 1993 Nephrogenic diabetes insipidus: identification of the genetic defect. Pediatr Nephrol 7:685–688

    Ishikawa S 1996 Nephrogenic diabetes insipidus. Nippon Rinsho 54:819–824

    Marples D, Frokiaer J, Dorup J, Knepper MA, Nielsen S 1996 Hypokalemia-induced downregulation of aquaporin-2 water channel expression in rat kidney medulla and cortex. J Clin Invest 97:1960–1968

    Rojek A, Nielsen J, Brooks HL, Gong H, Kim YH, Kwon TH, Frokaer J, Nielsen S 2005 Altered expression of selected genes in kidney of rats with lithium-induced NDI. Am J Physiol Renal Physiol 288:F1276–F1289

    Kuwahara M, Fushimi K, Terada Y, Bai L, Marumo F, Sasaki S 1995 cAMP-dependent phosphorylation stimulates water permeability of aquaporin-collecting duct water channel protein expressed in Xenopus oocytes. J Biol Chem 270:10384–10387

    Lande MB, Jo I, Zeidel ML, Somers M, Harris Jr HW 1996 Phosphorylation of aquaporin-2 does not alter the membrane water permeability of rat papillary water channel-containing vesicles. J Biol Chem 271:5552–5557

    Fushimi K, Sasaki S, Marumo F 1997 Phosphorylation of serine 256 is required for cAMP-dependent regulatory exocytosis of the aquaporin-2 water channel. J Biol Chem 272:14800–14804

    Katsura T, Gustafson CE, Ausiello DA, Brown D 1997 Protein kinase A phosphorylation is involved in regulated exocytosis of aquaporin-2 in transfected LLC-PK1 cells. Am J Physiol 272:F817–F822

    Nishimoto G, Zelenina M, Li D, Yasui M, Aperia A, Nielsen S, Nairn AC 1999 Arginine vasopressin stimulates phosphorylation of aquaporin-2 in rat renal tissue. Am J Physiol 276:F254–F259

    Christensen BM, Zelenina M, Aperia A, Nielsen S 2000 Localization and regulation of PKA-phosphorylated AQP2 in response to V(2)-receptor agonist/antagonist treatment. Am J Physiol Renal Physiol 278:F29–F42

    Kamsteeg EJ, Heijnen I, van Os CH, Deen PM 2000 The subcellular localization of an aquaporin-2 tetramer depends on the stoichiometry of phosphorylated and nonphosphorylated monomers. J Cell Biol 151:919–930

    van Balkom BW, Savelkoul PJ, Markovich D, Hofman E, Nielsen S, van der Sluijs P, Deen PM 2002 The role of putative phosphorylation sites in the targeting and shuttling of the aquaporin-2 water channel. J Biol Chem 277:41473–41479

    Valenti G, Procino G, Carmosino M, Frigeri A, Mannucci R, Nicoletti I, Svelto M 2000 The phosphatase inhibitor okadaic acid induces AQP2 translocation independently from AQP2 phosphorylation in renal collecting duct cells. J Cell Sci 113(Pt 11):1985–1992

    Lu H, Sun TX, Bouley R, Blackburn K, McLaughlin M, Brown D 2004 Inhibition of endocytosis causes phosphorylation (S256)-independent plasma membrane accumulation of AQP2. Am J Physiol Renal Physiol 286:F233–F243

    Bouley R, Breton S, Sun T, McLaughlin M, Nsumu NN, Lin HY, Ausiello DA, Brown D 2000 Nitric oxide and atrial natriuretic factor stimulate cGMP-dependent membrane insertion of aquaporin 2 in renal epithelial cells. J Clin Invest 106:1115–1126

    Bouley R, Pastor-Soler N, Cohen O, McLaughlin M, Breton S, Brown D 2005 Stimulation of AQP2 membrane insertion in renal epithelial cells in vitro and in vivo by the cGMP phosphodiesterase inhibitor sildenafil citrate (Viagra). Am J Physiol Renal Physiol 288:F1103–F1112

    Procino G, Carmosino M, Marin O, Brunati AM, Contri A, Pinna LA, Mannucci R, Nielsen S, Kwon TH, Svelto M, Valenti G 2003 Ser-256 phosphorylation dynamics of aquaporin 2 during maturation from the ER to the vesicular compartment in renal cells. FASEB J 17:1886–1888

    Klussmann E, Maric K, Wiesner B, Beyermann M, Rosenthal W 1999 Protein kinase A anchoring proteins are required for vasopressin-mediated translocation of aquaporin-2 into cell membranes of renal principal cells. J Biol Chem 274:4934–4938

    Klussmann E, Edemir B, Pepperle B, Tamma G, Henn V, Klauschenz E, Hundsrucker C, Maric K, Rosenthal W 2001 Ht31: the first protein kinase A anchoring protein to integrate protein kinase A and Rho signaling. FEBS Lett 507:264–268

    Henn V, Edemir B, Stefan E, Wiesner B, Lorenz D, Theilig F, Schmitt R, Vossebein L, Tamma G, Beyermann M, Krause E, Herberg FW, Valenti G, Bachmann S, Rosenthal W, Klussmann E 2004 Identification of a novel A-kinase anchoring protein 18 isoform and evidence for its role in the vasopressin-induced aquaporin-2 shuttle in renal principal cells. J Biol Chem 279:26654–26665

    Phillips ME, Taylor A 1989 Effect of nocodazole on the water permeability response to vasopressin in rabbit collecting tubules perfused in vitro. J Physiol 411:529–544

    Valenti G, Hugon JS, Bourguet J 1988 To what extent is microtubular network involved in antidiuretic response Am J Physiol 255:F1098–F1106

    Bourguet J, Hugon JS, Valenti G, Svelto M 1988 ADH-induced water permeability: what role for the microtubular network Comp Biochem Physiol A 90:669–672

    Marples D, Schroer TA, Ahrens N, Taylor A, Knepper MA, Nielsen S 1998 Dynein and dynactin colocalize with AQP2 water channels in intracellular vesicles from kidney collecting duct. Am J Physiol 274:F384–F394

    Ding GH, Franki N, Condeelis J, Hays RM 1991 Vasopressin depolymerizes F-actin in toad bladder epithelial cells. Am J Physiol 260:C9–C16

    Hays RM, Condeelis J, Gao Y, Simon H, Ding G, Franki N 1993 The effect of vasopressin on the cytoskeleton of the epithelial cell. Pediatr Nephrol 7:672–679

    Simon H, Gao Y, Franki N, Hays RM 1993 Vasopressin depolymerizes apical F-actin in rat inner medullary collecting duct. Am J Physiol 265:C757–C762

    Dibas A, Mia A, Yorio T 2000 Microfilament network is needed for the endocytosis of water channels and not for apical membrane insertion upon vasopressin action. Proc Soc Exp Biol Med 223:203–209

    Franki N, Ding G, Gao Y, Hays RM 1992 Effect of cytochalasin D on the actin cytoskeleton of the toad bladder epithelial cell. Am J Physiol 263:C995–C1000

    Valenti G, Frigeri A, Ronco PM, D’Ettorre C, Svelto M 1996 Expression and functional analysis of water channels in a stably AQP2-transfected human collecting duct cell line. J Biol Chem 271:24365–24370

    Klussmann E, Tamma G, Lorenz D, Wiesner B, Maric K, Hofmann F, Aktories K, Valenti G, Rosenthal W 2001 An inhibitory role of Rho in the vasopressin-mediated translocation of aquaporin-2 into cell membranes of renal principal cells. J Biol Chem 276:20451–20457

    Tamma G, Klussmann E, Maric K, Aktories K, Svelto M, Rosenthal W, Valenti G 2001 Rho inhibits cAMP-induced translocation of aquaporin-2 into the apical membrane of renal cells. Am J Physiol Renal Physiol 281:F1092–F1101

    Tamma G, Klussmann E, Procino G, Svelto M, Rosenthal W, Valenti G 2003 cAMP-induced AQP2 translocation is associated with RhoA inhibition through RhoA phosphorylation and interaction with RhoGDI. J Cell Sci 116:1519–1525

    Tamma G, Wiesner B, Furkert J, Hahm D, Oksche A, Schaefer M, Valenti G, Rosenthal W, Klussmann E 2003 The prostaglandin E2 analogue sulprostone antagonizes vasopressin-induced antidiuresis through activation of Rho. J Cell Sci 116:3285–3294

    Procino G, Carmosino M, Tamma G, Gouraud S, Laera A, Riccardi D, Svelto M, Valenti G 2004 Extracellular calcium antagonizes forskolin-induced aquaporin 2 trafficking in collecting duct cells. Kidney Int 66:2245–2255

    Hirao M, Sato N, Kondo T, Yonemura S, Monden M, Sasaki T, Takai Y, Tsukita S, Tsukita S 1996 Regulation mechanism of ERM (ezrin/radixin/moesin) protein/plasma membrane association: possible involvement of phosphatidylinositol turnover and Rho-dependent signaling pathway. J Cell Biol 135:37–51

    Takahashi K, Sasaki T, Mammoto A, Takaishi K, Kameyama T, Tsukita S, Takai Y 1997 Direct interaction of the Rho GDP dissociation inhibitor with ezrin/radixin/moesin initiates the activation of the Rho small G protein. J Biol Chem 272:23371–23375

    Mammoto A, Takahashi K, Sasaki T, Takai Y 2000 Stimulation of Rho GDI release by ERM proteins. Methods Enzymol 325:91–101

    Tamma G, Klussmann E, Oehlke J, Krause E, Rosenthal W, Svelto M, Valenti G2005 Actin remodeling requires ERM function to facilitate AQP2 apical targeting. J Cell Sci 118(Pt 16):3623–3630

    Noda Y, Horikawa S, Katayama Y, Sasaki S 2005 Identification of a multiprotein "motor" complex binding to water channel aquaporin-2. Biochem Biophys Res Commun 330:1041–1047

    Noda Y, Horikawa S, Katayama Y, Sasaki S 2004 Water channel aquaporin-2 directly binds to actin. Biochem Biophys Res Commun 322:740–745

    Barile M, Pisitkun T, Yu MJ, Chou CL, Verbalis MJ, Shen RF, Knepper MA 2005 Large-scale protein identification in intracellular aquaporin-2 vesicles from renal inner medullary collecting duct. Mol Cell Proteomics 4:1095–1106

    Chou CL, Christensen BM, Frische S, Vorum H, Desai RA, Hoffert JD, de Lanerolle P, Nielsen S, Knepper MA 2004 Non-muscle myosin II and myosin light chain kinase are downstream targets for vasopressin signaling in the renal collecting duct. J Biol Chem 279:49026–49035

    Pisitkun T, Shen RF, Knepper MA 2004 Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci USA 101:13368–13373

    Burgoyne RD, Morgan A 1995 Ca2+ and secretory-vesicle dynamics. Trends Neurosci 18:191–196

    Bajjalieh SM, Scheller RH 1995 The biochemistry of neurotransmitter secretion. J Biol Chem 270:1971–1974

    Bokvist K, Eliasson L, Ammala C, Renstrom E, Rorsman P 1995 Co-localization of L-type Ca2+ channels and insulin-containing secretory granules and its significance for the initiation of exocytosis in mouse pancreatic B-cells. EMBO J 14:50–57

    Yip KP 2002 Coupling of vasopressin-induced intracellular Ca2+ mobilization and apical exocytosis in perfused rat kidney collecting duct. J Physiol 538:891–899

    Star RA, Nonoguchi H, Balaban R, Knepper MA 1988 Calcium and cyclic adenosine monophosphate as second messengers for vasopressin in the rat inner medullary collecting duct. J Clin Invest 81:1879–1888

    Maeda Y, Han JS, Gibson CC, Knepper MA 1993 Vasopressin and oxytocin receptors coupled to Ca2+ mobilization in rat inner medullary collecting duct. Am J Physiol 265:F15–F25

    Chou CL, Yip KP, Michea L, Kador K, Ferraris JD, Wade JB, Knepper MA 2000 Regulation of aquaporin-2 trafficking by vasopressin in the renal collecting duct. Roles of ryanodine-sensitive Ca2+ stores and calmodulin. J Biol Chem 275:36839–36846

    Champigneulle A, Siga E, Vassent G, Imbert-Teboul M 1993 V2-like vasopressin receptor mobilizes intracellular Ca2+ in rat medullary collecting tubules. Am J Physiol 265:F35–F45

    Ecelbarger CA, Chou CL, Lolait SJ, Knepper MA, DiGiovanni SR 1996 Evidence for dual signaling pathways for V2 vasopressin receptor in rat inner medullary collecting duct. Am J Physiol 270:F623–F633

    Chou CL, Rapko SI, Knepper MA 1998 Phosphoinositide signaling in rat inner medullary collecting duct. Am J Physiol 274:F564–F572

    Lorenz D, Krylov A, Hahm D, Hagen V, Rosenthal W, Pohl P, Maric K 2003 Cyclic AMP is sufficient for triggering the exocytic recruitment of aquaporin-2 in renal epithelial cells. EMBO Rep 4:88–93

    Nielsen S, Marples D, Birn H, Mohtashami M, Dalby NO, Trimble M, Knepper M 1995 Expression of VAMP-2-like protein in kidney collecting duct intracellular vesicles. Colocalization with aquaporin-2 water channels. J Clin Invest 96:1834–1844

    Franki N, Macaluso F, Gao Y, Hays RM 1995 Vesicle fusion proteins in rat inner medullary collecting duct and amphibian bladder. Am J Physiol 268:C792–C797

    Mandon B, Nielsen S, Kishore BK, Knepper MA 1997 Expression of syntaxins in rat kidney. Am J Physiol 273:F718–F730

    Mandon B, Chou CL, Nielsen S, Knepper MA 1996 Syntaxin-4 is localized to the apical plasma membrane of rat renal collecting duct cells: possible role in aquaporin-2 trafficking. J Clin Invest 98:906–913

    Inoue T, Nielsen S, Mandon B, Terris J, Kishore BK, Knepper MA 1998 SNAP-23 in rat kidney: colocalization with aquaporin-2 in collecting duct vesicles. Am J Physiol 275:F752–F760

    Ravichandran V, Chawla A, Roche PA 1996 Identification of a novel syntaxin- and synaptobrevin/VAMP-binding protein, SNAP-23, expressed in non-neuronal tissues. J Biol Chem 271:13300–13303

    Li X, Low SH, Miura M, Weimbs T 2002 SNARE expression and localization in renal epithelial cells suggest mechanism for variability of trafficking phenotypes. Am J Physiol Renal Physiol 283:F1111–F1122

    Gouraud S, Laera A, Calamita G, Carmosino M, Procino G, Rossetto O, Mannucci R, Rosenthal W, Svelto M, Valenti G 2002 Functional involvement of VAMP/synaptobrevin-2 in cAMP-stimulated aquaporin 2 translocation in renal collecting duct cells. J Cell Sci 115:3667–3674

    Valenti G, Procino G, Liebenhoff U, Frigeri A, Benedetti PA, Ahnert-Hilger G, Nurnberg B, Svelto M, Rosenthal W 1998 A heterotrimeric G protein of the Gi family is required for cAMP-triggered trafficking of aquaporin 2 in kidney epithelial cells. J Biol Chem 273:22627–22634

    Sands JM, Naruse M, Baum M, Jo I, Hebert SC, Brown EM, Harris HW 1997 Apical extracellular calcium/polyvalent cation-sensing receptor regulates vasopressin-elicited water permeability in rat kidney inner medullary collecting duct. J Clin Invest 99:1399–1405

    Noda Y, Horikawa S, Furukawa T, Hirai K, Katayama Y, Asai T, Kuwahara M, Katagiri K, Kinashi T, Hattori M, Minato N, Sasaki S 2004 Aquaporin-2 trafficking is regulated by PDZ-domain containing protein SPA-1. FEBS Lett 568:139–145

    Brown EM, Pollak M, Hebert SC 1995 Sensing of extracellular Ca2+ by parathyroid and kidney cells: cloning and characterization of an extracellular Ca(2+)-sensing receptor. Am J Kidney Dis 25:506–513

    Riccardi D, Hall AE, Chattopadhyay N, Xu JZ, Brown EM, Hebert SC 1998 Localization of the extracellular Ca2+/polyvalent cation-sensing protein in rat kidney. Am J Physiol 274:F611–F22

    Hebert SC, Brown EM, Harris HW 1997 Role of the Ca(2+)-sensing receptor in divalent mineral ion homeostasis. J Exp Biol 200:295–302

    Brown EM, MacLeod RJ 2001 Extracellular calcium sensing and extracellular calcium signaling. Physiol Rev 81:239–297

    Hofer AM, Brown EM 2003 Extracellular calcium sensing and signalling. Nat Rev Mol Cell Biol 4:530–538

    Ward DT, Riccardi D 2002 Renal physiology of the extracellular calcium-sensing receptor. Pflugers Arch 445:169–176

    Sands JM, Flores FX, Kato A, Baum MA, Brown EM, Ward DT, Hebert SC, Harris HW 1998 Vasopressin-elicited water and urea permeabilities are altered in IMCD in hypercalcemic rats. Am J Physiol 274:F978–F985

    Arthur JM, Collinsworth GP, Gettys TW, Quarles LD, Raymond JR 1997 Specific coupling of a cation-sensing receptor to G protein -subunits in MDCK cells. Am J Physiol 273:F129–F135

    Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, Sun A, Hediger MA, Lytton J, Hebert SC 1993 Cloning and characterization of an extracellular Ca(2+)-sensing receptor from bovine parathyroid. Nature 366:575–580

    Takaichi K, Kurokawa K 1988 AVP-sensitive cAMP production is dependent on calmodulin in both MTAL and MCT. Am J Physiol 255:F834–F840

    De Jesus Ferreira MC, Bailly C 1998 Extracellular Ca2+ decreases chloride reabsorption in rat CTAL by inhibiting cAMP pathway. Am J Physiol 275:F198–F203

    Aarab L, Siaume-Perez S, Chabardes D 1993 The activation of protein kinase C prevents PGE2-induced inhibition of AVP-dependent cAMP accumulation in the rat outer medullary collecting tubule. Pflugers Arch 425:417–425

    Aarab L, Montegut M, Siaume-Perez S, Imbert-Teboul M, Chabardes D 1993 PGE2-induced inhibition of AVP-dependent cAMP accumulation in the OMCD of the rat kidney is cumulative with respect to the effects of 2-adrenergic and 1-adenosine agonists, insensitive to pertussis toxin and dependent on extracellular calcium. Pflugers Arch 423:397–405

    Pi M, Spurney RF, Tu Q, Hinson T, Quarles LD 2002 Calcium-sensing receptor activation of rho involves filamin and -guanine nucleotide exchange factor. Endocrinology 143:3830–3838

    Breyer MD, Breyer RM 2000 Prostaglandin E receptors and the kidney. Am J Physiol Renal Physiol 279:F12–F23

    Zelenina M, Christensen BM, Palmer J, Nairn AC, Nielsen S, Aperia A 2000 Prostaglandin E(2) interaction with AVP: effects on AQP2 phosphorylation and distribution. Am J Physiol Renal Physiol 278:F388–F394

    Murer L, Addabbo F, Carmosino M, Procino G, Tamma G, Montini G, Rigamonti W, Zucchetta P, Della Vella M, Venturini A, Zacchello G, Svelto M, Valenti G 2004 Selective decrease in urinary aquaporin 2 and increase in prostaglandin E2 excretion is associated with postobstructive polyuria in human congenital hydronephrosis. J Am Soc Nephrol 15:2705–2712

    Prie D, Dussaule JC, Lelongt B, Geniteau-Legendre M, Chatelet F, Cassingena R, Vandewalle A, Ronco PM 1994 Principal cell-specific antigen and hormonal regulatory network in RC.SVtsA58 cell line. Am J Physiol 266:C1628–C1638

    Tamma G, Carmosino M, Svelto M, Valenti G, Bradykinin signaling counteracts cAMP elicited AQP2 translocation in renal cells. J Am Soc Nephrol, in press

    Drummer C, Norsk P, Heer M 2001 Water and sodium balance in space. Am J Kidney Dis 38:684–690

    Gaspare De Santo N, Cirillo M, Valenti G, Perna A, Anastasio P, Drummer C 2005 Renal function in space: the link between osteoporosis, hypercalciuria, and aquaporins. J Ren Nutr 15:183–188

    Drummer C, Valenti G, Cirillo M, Perna A, Bellini L, Nenov V, De Santo NG 2002 Vasopressin, hypercalciuria and aquaporin—the key elements for impaired renal water handling in astronauts Nephron 92:503–514(Giovanna Valenti, Giuseppe Procino, Graz)