Combination of secretin and fluvastatin ameliorates the polyuria associated with X-linked nephrogenic diabetes insipidus in mice

Giuseppe Procino, Serena Milano, Monica Carmosino, Claudia Barbieri, Maria C Nicoletti, Jian H Li, Jürgen Wess, Maria Svelto, Giuseppe Procino, Serena Milano, Monica Carmosino, Claudia Barbieri, Maria C Nicoletti, Jian H Li, Jürgen Wess, Maria Svelto

Abstract

X-linked nephrogenic diabetes insipidus (X-NDI) is a disease caused by inactivating mutations of the vasopressin (AVP) type 2 receptor (V2R) gene. Loss of V2R function prevents plasma membrane expression of the AQP2 water channel in the kidney collecting duct cells and impairs the kidney concentration ability. In an attempt to develop strategies to bypass V2R signaling in X-NDI, we evaluated the effects of secretin and fluvastatin, either alone or in combination, on kidney function in a mouse model of X-NDI. The secretin receptor was found to be functionally expressed in the kidney collecting duct cells. Based on this, X-NDI mice were infused with secretin for 14 days but urinary parameters were not altered by the infusion. Interestingly, secretin significantly increased AQP2 levels in the collecting duct but the protein primarily accumulated in the cytosol. Since we previously reported that fluvastatin treatment increased AQP2 plasma membrane expression in wild-type mice, secretin-infused X-NDI mice received a single injection of fluvastatin. Interestingly, urine production by X-NDI mice treated with secretin plus fluvastatin was reduced by nearly 90% and the urine osmolality was doubled. Immunostaining showed that secretin increased intracellular stores of AQP2 and the addition of fluvastatin promoted AQP2 trafficking to the plasma membrane. Taken together, these findings open new perspectives for the pharmacological treatment of X-NDI.

Figures

Figure 1
Figure 1
Analysis of the expression of secretin receptor (SCTR) in the mouse kidney by reverse transcriptase-PCR (RT-PCR) and western blotting. (a) Total RNAs extracted from kidney inner medulla (IM), outer medulla (OM), cortex (CTX), and pancreas were probed for the presence of mRNA encoding SCTR. Specific bands were obtained in all kidney fractions. Control RT-PCR was performed using primers amplifying mouse beta-actin mRNA. Three independent experiments were carried out. (b) Total protein extract from mouse kidney IM, OM, and CTX, were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotted for the presence of SCTR. Mouse brain, cerebellum, liver, heart, and pancreas were used as additional controls. The specificity of the immunodetected band was demonstrated using anti-SCTR antibodies pre-adsorbed on the immunizing peptide. Three independent experiments were carried out.
Figure 2
Figure 2
Immunolocalization of secretin receptor (SCTR) in human kidney sections. Immunofluorescence detection of SCTR in human kidney. (a) SCTR was stained with Alexa Fluor-555 (red), aquaporin 2 (AQP2) was stained with Alexa Fluor-488 (green), and Na+/K+-ATPase with Alexa Fluor-633 (blue). AQP2 was seen at the apical pole of the collecting duct (CD) principal cells, whereas the SCTR antibody decorated the basolateral membrane of the same cells and colocalized with the basolateral marker Na+/K+-ATPase (pink color in the × 3 magnified signal). (b) SCTR was stained with Alexa Fluor-488 (green), AQP3 was stained with Alexa Fluor-555 (red), tissue counterstained with TO-PRO (blue). SCTR largely colocalized with AQP3 at the basolateral membrane of CD cells (yellow color in the × 3 magnified signal). (c) Co-immunostaining in serial sections using AQP2 (green) as marker of the CD and Tamm–Horsfall protein (THP, red) as marker of the thick ascending limb (TAL). SCTR-positive tubules (asterisks) were also positive for THP. Magnification × 60.
Figure 3
Figure 3
Effect of treatment with secretin (SCT) and dDAVP on cyclic adenosine monophosphate (cAMP) concentrations and AQP2 plasma membrane localization on kidney collecting ducts. (a) SCT and dDAVP-induced cAMP production in mouse inner medullary collecting duct (IMCD) tubule suspensions. Freshly isolated IMCD tubule suspensions from C57BL/6 WT or tamoxifen (TMX)-induced X-linked nephrogenic diabetes insipidus (X-NDI) mice (12-week-old males; n=8 per individual experiment) were pooled and equally distributed into 24-well plates. Samples were treated with the indicated concentrations of dDAVP or SCT for 60 min at 37 °C. Total cAMP generated in each well was normalized to the amount of protein present in each well. Three independent experiments were carried out. Data are provided as mean±s.e.m. (b) Indirect immunofluorescence images of kidney tissue slices showing AQP2 in principal cells of collecting ducts (CDs). One C57BL/6 WT mouse kidney was cut into thin slices and incubated in vitro with buffer alone (control) or with dDAVP 10−7 mol/l or SCT 10−7 mol/l for 1 h before fixation by immersion, sectioning, and immunostaining to detect AQP2. Under control conditions, AQP2 was mainly localized throughout the cytoplasm and showed little apical membrane staining in CDs. In the presence of dDAVP or SCT, AQP2 staining of the apical membrane region was increased in principal cells of the cortical CD. The images are representative of three independent experiments. (c) Quantification of the number of cells displaying plasma membrane (PM) localization was determined by counting the fraction of cells showing apical PM localization. The extent of translocation is expressed as the percentage (means±s.e.) of cells showing a continuous apical PM localization in at least 100 randomly chosen CD principal cells from three independent determinations. Quantitation of cells displaying AQP2 apical membrane expression expressed as a percentage of total cells observed. ***P<0.0005 with Student's t-test. dDAVP, 1-deamino-8-D-arginine-vasopressin.
Figure 4
Figure 4
Effect of prolonged infusion of X-linked nephrogenic diabetes insipidus (X-NDI) mice with secretin (SCT) on urine output, urine osmolality, aquaporin 2 (AQP2) abundance, and membrane expression. (a) Five months after the end of the tamoxifen (TMX)-injection period, X-NDI mice were subcutaneously implanted with osmotic minipumps and continuously infused with saline alone or SCT at a dose of 1 nmol/kg/day over a 14-day period. Urine output, normalized for body weight, and osmolality were measured daily. No significant changes in urinary parameters were seen in SCT-infused animals. (b) At the end of the experiment, quantitative reverse transcription polymerase chain reaction was performed on kidneys from X-NDI infused with saline (−SCT) or with secretin (+SCT). Control animals that did not receive TMX were included (CTR). Relative quantification of gene expression (RQ) was performed setting the amount of AQP2 mRNA in vehicle-infused animals (−SCT) as 1. SCT-infused animals exhibited an approximately twofold increase in AQP2 gene transcription compared with saline-infused mice. (*P<0.05, **P<0.005, ***P<0.0005 with Student's t-test, n=6 per group.) (c) Western blotting with anti-AQP2 antibodies was carried out using homogenates prepared from whole left kidneys. Kidneys from control animals of the same age that had not received TMX were used as control. Protein extract from mouse inner medulla (IM) was loaded as positive control for AQP2 immunodetection. 29 kDa AQP2 band (AQP2) and glycosylated AQP2 band (gly) are indicated. Densitometric analysis of AQP2 bands, normalized for the actin abundance in each fraction, is reported on the right indicating that SCT infusion promoted a nearly 90% increase of AQP2 abundance. (d) The right kidneys were subjected to immunofluorescence localization of AQP2 in the three kidney regions. The number of AQP2-positive CD cells and the intensity of AQP2 fluorescence were increased in SCT-infused animals. Observation of single tubules at higher magnification (right panels) unambiguously showed that AQP2 was localized intracellularly. The experiment was repeated three times and comparable results were obtained. *P<0.05; ***P<0.0001; N=6 per group.
Figure 5
Figure 5
Combination of prolonged secretin (SCT) infusion and a single fluvastatin (FLU) injection markedly ameliorates nephrogenic diabetes insipidus (NDI) symptoms in X-linked NDI (X-NDI) mice. Five months after the end of tamoxifen (TMX)-injection period, X-NDI mice were subcutaneously implanted with osmotic minipumps and continuously infused with either saline alone or SCT at a dose of 1 nmol/kg/day over a 14-day period. On the last day of SCT infusion, half of the control and SCT-infused mice were given an intraperitoneal (i.p.) injection of FLU (50 mg/kg) and urine output and osmolality were monitored for the following 6 h. Compared with untreated animals (CTR), FLU alone had a slight effect on urine output and urine osmolality. As already shown in Figure 4, SCT alone had no significant effect on urinary parameters. Interestingly, a single FLU injection in SCT-infused mice (SCT+FLU) led to a marked reduction of urine output, accompanied by a significant increase of urine osmolality. Data are presented as mean±s.e.m. **P<0.05; ***P<0.001. N=5 per group.
Figure 6
Figure 6
Combination of prolonged secretin (SCT) infusion and a single fluvastatin injection upregulates aquaporin 2 (AQP2) expression and promotes AQP2 plasma membrane localization in X-linked nephrogenic diabetes insipidus (X-NDI) mice. At the end of the treatment described in Figure 5, mice were killed and their kidneys subjected to AQP2 immunolocalization followed by confocal examination. V2Rfl/yEsr1-Cre mice of the same age that had not received tamoxifen (−TMX) were analyzed in parallel. Random pictures were taken in the kidney cortex, outer medulla, and inner medulla. A high magnification of a detail of each image is shown in the upper left corner of each panel. Compared with non-induced animals (−TMX, CTR), mice with V2R deletion (+TMX, CTR) showed a marked reduction of AQP2 immunoreactivity in each kidney portion. FLU injection triggered the accumulation of the remaining amount of AQP2 on the luminal membrane. SCT infusion greatly upregulated AQP2 immunoreactivity in the kidney collecting ducts (CDs). At higher magnification, AQP2 was seen mainly in the cytoplasm with negligible plasma membrane expression. In mice chronically infused with SCT and injected with fluvastatin (SCT+FLU), AQP2 was upregulated and mainly expressed at the luminal membrane of CD tubules. Representative images are shown (N=5). G, glomerulus.

References

    1. Bichet DG. V2R mutations and nephrogenic diabetes insipidus. Prog Mol Biol Transl Sci. 2009;89:15–29.
    1. Robben JH, Knoers NV, Deen PM. Cell biological aspects of the vasopressin type-2 receptor and aquaporin 2 water channel in nephrogenic diabetes insipidus. Am J Physiol Renal Physiol. 2006;291:F257–F270.
    1. Wesche D, Deen PM, Knoers NV. Congenital nephrogenic diabetes insipidus: the current state of affairs. Pediatr Nephrol. 2012;27:2183–2204.
    1. Boone M, Deen PM. Physiology and pathophysiology of the vasopressin-regulated renal water reabsorption. Pflugers Arch. 2008;456:1005–1024.
    1. Hasler U, Leroy V, Martin PY, et al. Aquaporin-2 abundance in the renal collecting duct: new insights from cultured cell models. Am J Physiol Renal Physiol. 2009;297:F10–F18.
    1. Knepper MA, Inoue T. Regulation of aquaporin-2 water channel trafficking by vasopressin. Curr Opin Cell Biol. 1997;9:560–564.
    1. Moeller HB, Fenton RA. Cell biology of vasopressin-regulated aquaporin-2 trafficking. Pflugers Arch. 2012;464:133–144.
    1. Nedvetsky PI, Tamma G, Beulshausen S, et al. Regulation of aquaporin-2 trafficking. Handb Exp Pharmacol. 2009. pp. 133–157.
    1. Tamma G, Procino G, Svelto M, et al. Cell culture models and animal models for studying the patho-physiological role of renal aquaporins. Cell Mol Life Sci. 2012;69:1931–1946.
    1. Tamma G, Procino G, Strafino A, et al. Hypotonicity induces aquaporin-2 internalization and cytosol-to-membrane translocation of ICln in renal cells. Endocrinology. 2007;148:1118–1130.
    1. Bouley R, Hawthorn G, Russo LM, et al. Aquaporin 2 (AQP2) and vasopressin type 2 receptor (V2R) endocytosis in kidney epithelial cells: AQP2 is located in 'endocytosis-resistant' membrane domains after vasopressin treatment. Biol Cell. 2006;98:215–232.
    1. Yasui M, Zelenin SM, Celsi G, et al. Adenylate cyclase-coupled vasopressin receptor activates AQP2 promoter via a dual effect on CRE and AP1 elements. Am J Physiol. 1997;272:F443–F450.
    1. Deen PM, van Aubel RA, van Lieburg AF, et al. Urinary content of aquaporin 1 and 2 in nephrogenic diabetes insipidus. J Am Soc Nephrol. 1996;7:836–841.
    1. Kanno K, Sasaki S, Hirata Y, et al. Urinary excretion of aquaporin-2 in patients with diabetes insipidus. N Engl J Med. 1995;332:1540–1545.
    1. Li JH, Chou CL, Li B, et al. A selective EP4 PGE2 receptor agonist alleviates disease in a new mouse model of X-linked nephrogenic diabetes insipidus. J Clin Invest. 2009;119:3115–3126.
    1. Promeneur D, Kwon TH, Frokiaer J, et al. Vasopressin V(2)-receptor-dependent regulation of AQP2 expression in Brattleboro rats. Am J Physiol Renal Physiol. 2000;279:F370–F382.
    1. Knoers N, Monnens LA. Nephrogenic diabetes insipidus: clinical symptoms, pathogenesis, genetics and treatment. Pediatr Nephrol. 1992;6:476–482.
    1. Knoers NV, Deen PM. Molecular and cellular defects in nephrogenic diabetes insipidus. Pediatr Nephrol. 2001;16:1146–1152.
    1. Bouley R, Breton S, Sun T, et al. Nitric oxide and atrial natriuretic factor stimulate cGMP-dependent membrane insertion of aquaporin 2 in renal epithelial cells. J Clin Invest. 2000;106:1115–1126.
    1. Bouley R, Hasler U, Lu HA, et al. Bypassing vasopressin receptor signaling pathways in nephrogenic diabetes insipidus. Semin Nephrol. 2008;28:266–278.
    1. Bouley R, Lu HA, Nunes P, et al. Calcitonin has a vasopressin-like effect on aquaporin-2 trafficking and urinary concentration. J Am Soc Nephrol. 2011;22:59–72.
    1. Li W, Zhang Y, Bouley R, et al. Simvastatin enhances aquaporin-2 surface expression and urinary concentration in vasopressin-deficient Brattleboro rats through modulation of Rho GTPase. Am J Physiol Renal Physiol. 2011;301:F309–F318.
    1. Olesen ET, Rutzler MR, Moeller HB, et al. Vasopressin-independent targeting of aquaporin-2 by selective E-prostanoid receptor agonists alleviates nephrogenic diabetes insipidus. Proc Natl Acad Sci USA. 2011;108:12949–12954.
    1. Procino G, Barbieri C, Carmosino M, et al. Fluvastatin modulates renal water reabsorption in vivo through increased AQP2 availability at the apical plasma membrane of collecting duct cells. Pflugers Arch. 2011;462:753–766.
    1. Russo LM, McKee M, Brown D. Methyl-beta-cyclodextrin induces vasopressin-independent apical accumulation of aquaporin-2 in the isolated, perfused rat kidney. Am J Physiol Renal Physiol. 2006;291:F246–F253.
    1. Los EL, Deen PM, Robben JH. Potential of nonpeptide (ant)agonists to rescue vasopressin V2 receptor mutants for the treatment of X-linked nephrogenic diabetes insipidus. J Neuroendocrinol. 2010;22:393–399.
    1. Jean-Alphonse F, Perkovska S, Frantz MC, et al. Biased agonist pharmacochaperones of the AVP V2 receptor may treat congenital nephrogenic diabetes insipidus. J Am Soc Nephrol. 2009;20:2190–2203.
    1. Robben JH, Kortenoeven ML, Sze M, et al. Intracellular activation of vasopressin V2 receptor mutants in nephrogenic diabetes insipidus by nonpeptide agonists. Proc Natl Acad Sci USA. 2009;106:12195–12200.
    1. Chu JY, Yung WH, Chow BK. Secretin: a pleiotrophic hormone. Ann N Y Acad Sci. 2006;1070:27–50.
    1. Trimble ER, Bruzzone R, Biden TJ, et al. Secretin stimulates cyclic AMP and inositol trisphosphate production in rat pancreatic acinar tissue by two fully independent mechanisms. Proc Natl Acad Sci USA. 1987;84:3146–3150.
    1. Chu JY, Chung SC, Lam AK, et al. Phenotypes developed in secretin receptor-null mice indicated a role for secretin in regulating renal water reabsorption. Mol Cell Biol. 2007;27:2499–2511.
    1. Charlton CG, Quirion R, Handelmann GE, et al. Secretin receptors in the rat kidney: adenylate cyclase activation and renal effects. Peptides. 1986;7:865–871.
    1. Velmurugan S, Brunton PJ, Leng G, et al. Circulating secretin activates supraoptic nucleus oxytocin and vasopressin neurons via noradrenergic pathways in the rat. Endocrinology. 2010;151:2681–2688.
    1. Glaser S, Lam IP, Franchitto A, et al. Knockout of secretin receptor reduces large cholangiocyte hyperplasia in mice with extrahepatic cholestasis induced by bile duct ligation. Hepatology. 2010;52:204–214.
    1. Wang X, Ye H, Ward CJ, et al. Insignificant effect of secretin in rodent models of polycystic kidney and liver disease. Am J Physiol Renal Physiol. 2012;303:F1089–F1098.
    1. Holtmann MH, Roettger BF, Pinon DI, et al. Role of receptor phosphorylation in desensitization and internalization of the secretin receptor. J Biol Chem. 1996;271:23566–23571.
    1. Ozcelebi F, Holtmann MH, Rentsch RU, et al. Agonist-stimulated phosphorylation of the carboxyl-terminal tail of the secretin receptor. Mol Pharmacol. 1995;48:818–824.
    1. Shetzline MA, Premont RT, Walker JK, et al. A role for receptor kinases in the regulation of class II G protein-coupled receptors. Phosphorylation and desensitization of the secretin receptor. J Biol Chem. 1998;273:6756–6762.
    1. Kortenoeven ML, Trimpert C, van den Brand M, et al. In mpkCCD cells, long-term regulation of aquaporin-2 by vasopressin occurs independent of protein kinase A and CREB but may involve Epac. Am J Physiol Renal Physiol. 2012;302:F1395–F1401.
    1. Ares GR, Caceres PS, Ortiz PA. Molecular regulation of NKCC2 in the thick ascending limb. Am J Physiol Renal Physiol. 2011;301:F1143–F1159.
    1. Hardcastle IR, Rowlands MG, Barber AM, et al. Inhibition of protein prenylation by metabolites of limonene. Biochem Pharmacol. 1999;57:801–809.
    1. Oppermann M, Mizel D, Kim SM, et al. Renal function in mice with targeted disruption of the A isoform of the Na-K-2Cl co-transporter. J Am Soc Nephrol. 2007;18:440–448.

Source: PubMed

3
구독하다