A novel therapeutic effect of statins on nephrogenic diabetes insipidus

Leonilde Bonfrate, Giuseppe Procino, David Q-H Wang, Maria Svelto, Piero Portincasa, Leonilde Bonfrate, Giuseppe Procino, David Q-H Wang, Maria Svelto, Piero Portincasa

Abstract

Statins competitively inhibit hepatic 3-hydroxy-3-methylglutaryl-coenzyme A reductase, resulting in reduced plasma total and low-density lipoprotein cholesterol levels. Recently, it has been shown that statins exert additional 'pleiotropic' effects by increasing expression levels of the membrane water channels aquaporin 2 (AQP2). AQP2 is localized mainly in the kidney and plays a critical role in determining cellular water content. This additional effect is independent of cholesterol homoeostasis, and depends on depletion of mevalonate-derived intermediates of sterol synthetic pathways, i.e. farnesylpyrophosphate and geranylgeranylpyrophosphate. By up-regulating the expression levels of AQP2, statins increase water reabsorption by the kidney, thus opening up a new avenue in treating patients with nephrogenic diabetes insipidus (NDI), a hereditary disease that yet lacks high-powered and limited side effects therapy. Aspects related to water balance determined by AQP2 in the kidney, as well as standard and novel therapeutic strategies of NDI are discussed.

Keywords: HMG-CoA; apical membrane; aquaporin; cholesterol-lowering drugs; hypercholesterolaemia; kidney; nephrogenic diabetes insipidus; vasopressin; water channels.

© 2015 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.

Figures

Fig 1
Fig 1
Anatomic structure of the nephron and collecting duct system, and localization of different aquaporins (AQPs) in the kidneys with vasopressin (AVP) effect. Sites of reabsorption of water and sodium chloride (NaCl) are shown. AQP6 is localized in the intracellular vesicle membranes of type-A intercalated cells of the collecting duct.
Fig 2
Fig 2
The topology of AQP2 with the COOH-terminal phosphorylation sites. AQP2 is a tetramer consisting of four identical protein subunits placed in the plasma membrane. Six transmembrane α-helices are arranged in a right-handed bundle and are represented by cylinders, with the amino (NH2-) and the carboxyl (COOH-) termini located on the cytoplasmic surface of the membrane. Five interhelical loop regions (A–E) form the extracellular and cytoplasmic vestibules. Loops B and E are hydrophobic loops that contain the highly, although not completely conserved, asparagine–proline–alanine (NPA) motifs. Such motifs appear to dip and overlap into the membrane, to construct the water pore ,. Serine residues at potential phosphorylation sites are labelled with their amino acid numbers at the carboxyl-terminal tail. AVP mediated increased (+) phosphorylation at S256, S264 and S269, and decreased (−) phosphorylation at S261. Both S269 and S256 phosphorylation are involved in AQP2 accumulation in the plasma membrane ,,.
Fig 3
Fig 3
Molecular pathways involved in AQP2-mediated water transport in the kidney. (A) Signalling cascades and molecular pathways involved in AQP2-mediated water transport in relation to vasopressin (AVP) and vasopressin receptor (AVPR2) in the principal cells of the collecting ducts ,,,. The increased influx of water by AQP2 tetramer at the apical site requires a complex cascade of intracellular processes in concert with efflux of water by AQP3 and AQP4 tetramers at the basolateral membrane. The AVPR2 is composed of 7 membrane-spanning helices. Upon binding of AVP within the transmembrane helices II–IV, allosteric structural changes occur ,, the G-alpha-s heterodimeric protein is stimulated, and activates the adenylyl cyclase. This step results in increased intracellular levels of cyclic adenosine monophosphate (cAMP), activation of protein kinase A (PKA), phosphorylation of AQP2 in intracellular vesicles at serine 256 and other residues in the AQP2 OOH terminal , (see also Fig.2), trafficking of endocytic vesicles to the apical plasma membrane, and fusion of AQP2-containing vesicles with the apical membrane. As stated in the text, PKA is also responsible for phosphorylation of the membrane-associated RhoA, association with GDI to form the inactive complex RhoA-GDI, a step facilitating AQP2 insertion into the plasma membrane during VP/PKA/cAMP-induced AQP2 translocation . The docking system for vesicles might include specific receptors in the collecting duct cells which are associated with certain membrane domains housing AQP2 (e.g. syntaxin-4). Abbreviation: PDEs, phosphodiesterases. See also ,,,. (B) Proposed model of transcytotic trafficking of AQP2 from basolateral to apical membrane in principal cell of the collecting ducts. At least eight steps are involved: (1) Synthesis in the endoplasmic reticulum and transport to the trans-Golgi network; (2) rapid insertion of AQP2 into the basolateral membrane; (3) rapid internalization by clathrin-dependent endocytosis which is responsible for limited expression of basolateral AQP2. This step is blockable by low temperature (4°C); (5) AQP2 transcytosis to the perinuclear recycling compartment and the apical recycling endosomes via the microtubule-dependent mechanism. This step is inhibitable by colchicine; (7) exocytosis of AQP2 at the apical membrane; (8) recycling of AQP2 towards the apical recycling endosomes via the clathrin-dependent endocytosis. Thin dotted arrows show alternative pathways (?) of AQP2. Asterisks indicate where vasopressin (AVP) stimulus is inducing increased exocytosis and recycling of AQP2 with effect on transepithelial water flux (apical side) and cell migration, tubulogenesis, and likely transepithelial water flux (basolateral side). See also ,.

References

    1. Shepherd J, Cobbe SM, Ford I, et al. Prevention of coronary heart disease with pravastatin in men with hypercholesterolemia. West of Scotland Coronary Prevention Study Group. N Engl J Med. 1995;333:1301–7.
    1. Jukema JW, Bruschke AV, van Boven AJ, et al. Effects of lipid lowering by pravastatin on progression and regression of coronary artery disease in symptomatic men with normal to moderately elevated serum cholesterol levels. The Regression Growth Evaluation Statin Study (REGRESS) Circulation. 1995;91:2528–40.
    1. Davignon J. The cardioprotective effects of statins. Curr Atheroscler Rep. 2004;6:27–35.
    1. The Long-Term Intervention with Pravastatin in Ischaemic Disease (LIPID) Study Group. Prevention of cardiovascular events and death with pravastatin in patients with coronary heart disease and a broad range of initial cholesterol levels. N Engl J Med. 1998;339:1349–57.
    1. Jones PH, Davidson MH, Stein EA, et al. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial) Am J Cardiol. 2003;92:152–60.
    1. Jones P, Kafonek S, Laurora I, et al. Comparative dose efficacy study of atorvastatin versus simvastatin, pravastatin, lovastatin, and fluvastatin in patients with hypercholesterolemia (the CURVES study) Am J Cardiol. 1998;81:582–7.
    1. Goff DC, Lloyd-Jones DM, Bennett G, et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63:2935–59.
    1. Stone NJ, Robinson J, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults. J Am Coll Cardiol. 2014;63:2889–934.
    1. Baigent C, Keech A, Kearney PM, et al. Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet. 2005;366:1267–78.
    1. Josan K, Majumdar SR, McAlister FA. The efficacy and safety of intensive statin therapy: a meta-analysis of randomized trials. CMAJ. 2008;178:576–84.
    1. Istvan ES, Deisenhofer J. Structural mechanism for statin inhibition of HMG-CoA reductase. Science. 2001;292:1160–4.
    1. Kovanen PT, Bilheimer DW, Goldstein JL, et al. Regulatory role for hepatic low density lipoprotein receptors in vivo in the dog. Proc Natl Acad Sci USA. 1981;78:1194–8.
    1. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature. 1990;343:425–30.
    1. Kallien G, Lange K, Stange EF, et al. The pravastatin-induced decrease of biliary cholesterol secretion is not directly related to an inhibition of cholesterol synthesis in humans. Hepatology. 1999;30:14–20.
    1. Duane WC, Hunninghake DB, Freeman ML, et al. Simvastatin, a competitive inhibitor of HMG-CoA reductase, lowers cholesterol saturation index of gallbladder bile. Hepatology. 1988;8:1147–50.
    1. Loria P, Bertolotti M, Cassinadri MT, et al. Short-term effects of simvastatin on bile acid synthesis and bile lipid secretion in human subjects. Hepatology. 1994;19:882–8.
    1. Logan GM, Duane WC. Lovastatin added to ursodeoxycholic acid further reduces biliary cholesterol saturation. Gastroenterology. 1990;98:1572–6.
    1. Takemoto M, Liao JK. Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors. Arterioscler Thromb Vasc Biol. 2001;21:1712–9.
    1. McFarlane SI, Muniyappa R, Francisco R, et al. Clinical review 145: pleiotropic effects of statins: lipid reduction and beyond. J Clin Endocrinol Metab. 2002;87:1451–8.
    1. McKenney JM. Potential nontraditional applications of statins. Ann Pharmacother. 2003;37:1063–71.
    1. Moeller HB, Rittig S, Fenton RA. Nephrogenic diabetes insipidus: essential insights into the molecular background and potential therapies for treatment. Endocr Rev. 2013;34:278–301.
    1. Agre P, King LS, Yasui M, et al. Aquaporin water channels–from atomic structure to clinical medicine. J Physiol. 2002;542:3–16.
    1. Portincasa P, Palasciano G, Svelto M, et al. Aquaporins in the hepatobiliary tract. Which, where and what they do in health and disease. Eur J Clin Invest. 2008;38:1–10.
    1. Calamita G, Ferri D, Gena P, et al. Water transport into bile and role in bile formation. Curr Drug Targets Immune Endocr Metabol Disord. 2005;5:137–42.
    1. Portincasa P, Calamita G. Water channel proteins in bile formation and flow in health and disease: when immiscible becomes miscible. Mol Aspects Med. 2012;33:651–64.
    1. Valenti G, Procino G, Liebenhoff U, et al. A heterotrimeric G protein of the Gi family is required for cAMP-triggered trafficking of aquaporin 2 in kidney epithelial cells. J Biol Chem. 1998;273:22627–34.
    1. Marples D. Water channels: who needs them anyway? The Lancet. 2000;355:1571–2.
    1. Nichols R. Polyuria and polydipsia. Diagnostic approach and problems associated with patient evaluation. Vet Clin North Am Small Anim Pract. 2001;31:833–44.
    1. Wilson JL, Miranda CA, Knepper MA. Vasopressin and the regulation of aquaporin-2. Clin Exp Nephrol. 2013;17:751–64.
    1. Takata K, Matsuzaki T, Tajika Y. Aquaporins: water channel proteins of the cell membrane. Prog Histochem Cytochem. 2004;39:1–83.
    1. Nagase H, Agren J, Saito A, et al. Molecular cloning and characterization of mouse aquaporin 6. Biochem Biophys Res Commun. 2007;352:12–6.
    1. Procino G, Mastrofrancesco L, Sallustio F, et al. AQP5 is expressed in type-B intercalated cells in the collecting duct system of the rat, mouse and human kidney. Cell Physiol Biochem. 2011;28:683–92.
    1. Nielsen S, Frokiaer J, Marples D, et al. Aquaporins in the kidney: from molecules to medicine. Physiol Rev. 2002;82:205–44.
    1. Nielsen S, Kwon TH, Christensen BM, et al. Physiology and pathophysiology of renal aquaporins. J Am Soc Nephrol. 1999;10:647–63.
    1. Hadchouel J, Busst C, Procino G, et al. Regulation of extracellular fluid volume and blood pressure by pendrin. Cell Physiol Biochem. 2011;28:505–12.
    1. Fushimi K, Uchida S, Hara Y, et al. Cloning and expression of apical membrane water channel of rat kidney collecting tubule. Nature. 1993;361:549–52.
    1. Nielsen S, Kwon TH, Frokiaer J, et al. Regulation and dysregulation of aquaporins in water balance disorders. J Intern Med. 2007;261:53–64.
    1. Ecelbarger CA, Terris J, Frindt G, et al. Aquaporin-3 water channel localization and regulation in rat kidney. Am J Physiol. 1995;269:F663–72.
    1. Terris J, Ecelbarger CA, Marples D, et al. Distribution of aquaporin-4 water channel expression within rat kidney. Am J Physiol. 1995;269:F775–85.
    1. Fenton RA, Pedersen CN, Moeller HB. New insights into regulated aquaporin-2 function. Curr Opin Nephrol Hypertens. 2013;22:551–8.
    1. Deen PM, Verdijk MA, Knoers NV, et al. Requirement of human renal water channel aquaporin-2 for vasopressin-dependent concentration of urine. Science. 1994;264:92–5.
    1. Deen PM, Croes H, van Aubel RA, et al. Water channels encoded by mutant aquaporin-2 genes in nephrogenic diabetes insipidus are impaired in their cellular routing. J Clin Invest. 1995;95:2291–6.
    1. Nielsen S, DiGiovanni SR, Christensen EI, et al. Cellular and subcellular immunolocalization of vasopressin-regulated water channel in rat kidney. Proc Natl Acad Sci USA. 1993;90:11663–7.
    1. Harris HW, Jr, Strange K, Zeidel ML. Current understanding of the cellular biology and molecular structure of the antidiuretic hormone-stimulated water transport pathway. J Clin Invest. 1991;88:1–8.
    1. Brown D. Membrane recycling and epithelial cell function. Am J Physiol. 1989;256:F1–12.
    1. Zimmerman EA, Nilaver G, Hou-Yu A, et al. Vasopressinergic and oxytocinergic pathways in the central nervous system. Fed Proc. 1984;43:91–6.
    1. Oakley RH, Laporte SA, Holt JA, et al. Association of beta-arrestin with G protein-coupled receptors during clathrin-mediated endocytosis dictates the profile of receptor resensitization. J Biol Chem. 1999;274:32248–57.
    1. Brown D. The ins and outs of aquaporin-2 trafficking. Am J Physiol Renal Physiol. 2003;284:F893–901.
    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. AJP Renal Physiol. 2011;301:F309–18.
    1. Hoffert JD, Pisitkun T, Wang G, et al. Quantitative phosphoproteomics of vasopressin-sensitive renal cells: regulation of aquaporin-2 phosphorylation at two sites. Proc Natl Acad Sci USA. 2006;103:7159–64.
    1. Nielsen S, Chou CL, Marples D, et al. Vasopressin increases water permeability of kidney collecting duct by inducing translocation of aquaporin-CD water channels to plasma membrane. Proc Natl Acad Sci USA. 1995;92:1013–7.
    1. Klussmann E, Maric K, Rosenthal W. The mechanisms of aquaporin control in the renal collecting duct. Rev Physiol Biochem Pharmacol. 2000;141:33–95.
    1. Knepper MA, Inoue T. Regulation of aquaporin-2 water channel trafficking by vasopressin. Curr Opin Cell Biol. 1997;9:560–4.
    1. Hayashi M, Sasaki S, Tsuganezawa H, et al. Expression and distribution of aquaporin of collecting duct are regulated by vasopressin V2 receptor in rat kidney. J Clin Invest. 1994;94:1778–83.
    1. Sasaki S, Fushimi K, Saito H, et al. Cloning, characterization, and chromosomal mapping of human aquaporin of collecting duct. J Clin Invest. 1994;93:1250–6.
    1. Liebenhoff U, Rosenthal W. Identification of Rab3-, Rab5a- and synaptobrevin II-like proteins in a preparation of rat kidney vesicles containing the vasopressin-regulated water channel. FEBS Lett. 1995;365:209–13.
    1. Ren XD, Kiosses WB, Schwartz MA. Regulation of the small GTP-binding protein Rho by cell adhesion and the cytoskeleton. EMBO J. 1999;18:578–85.
    1. Nobes CD, Hall A. Rho, rac, and cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia. Cell. 1995;81:53–62.
    1. Dong JM, Leung T, Manser E, et al. cAMP-induced morphological changes are counteracted by the activated RhoA small GTPase and the Rho kinase ROKalpha. J Biol Chem. 1998;273:22554–62.
    1. Sasaki T, Takai Y. The Rho small G protein family-Rho GDI system as a temporal and spatial determinant for cytoskeletal control. Biochem Biophys Res Commun. 1998;245:641–5.
    1. Van Aelst L, D'Souza-Schorey C. Rho GTPases and signaling networks. Genes Dev. 1997;11:2295–322.
    1. Tamma G, Klussmann E, Procino G, et al. cAMP-induced AQP2 translocation is associated with RhoA inhibition through RhoA phosphorylation and interaction with RhoGDI. J Cell Sci. 2003;116:1519–25.
    1. Forget MA, Desrosiers RR, Gingras D, et al. Phosphorylation states of Cdc42 and RhoA regulate their interactions with Rho GDP dissociation inhibitor and their extraction from biological membranes. Biochem J. 2002;361:243–54.
    1. DiGiovanni SR, Nielsen S, Christensen EI, et al. Regulation of collecting duct water channel expression by vasopressin in Brattleboro rat. Proc Natl Acad Sci USA. 1994;91:8984–8.
    1. Gustafson CE, Katsura T, McKee M, et al. Recycling of AQP2 occurs through a temperature- and bafilomycin-sensitive trans-Golgi-associated compartment. Am J Physiol Renal Physiol. 2000;278:F317–26.
    1. Lu H, Sun TX, Bouley R, et al. Inhibition of endocytosis causes phosphorylation (S256)-independent plasma membrane accumulation of AQP2. Am J Physiol Renal Physiol. 2004;286:F233–43.
    1. Sun TX, Van Hoek A, Huang Y, et al. Aquaporin-2 localization in clathrin-coated pits: inhibition of endocytosis by dominant-negative dynamin. Am J Physiol Renal Physiol. 2002;282:F998–1011.
    1. Chen Y, Rice W, Gu Z, et al. Aquaporin 2 promotes cell migration and epithelial morphogenesis. J Am Soc Nephrol. 2012;23:1506–17.
    1. Yui N, Lu HAJ, Chen Y, et al. Basolateral targeting and microtubule-dependent transcytosis of the aquaporin-2 water channel. AJP Cell Physiol. 2012;304:C38–48.
    1. Okamoto CT. Caring about the other 47% of the water channels. Focus on “Basolateral targeting and microtubule-dependent transcytosis of the aquaporin-2 water channel”. AJP Cell Physiol. 2012;304:C33–5.
    1. van Balkom BW, van Raak M, Breton S, et al. 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. 2003;278:1101–7.
    1. Coleman RA, Wu DC, Liu J, et al. Expression of aquaporins in the renal connecting tubule. Am J Physiol Renal Physiol. 2000;279:F874–83.
    1. de Seigneux S, Nielsen J, Olesen ET, et al. Long-term aldosterone treatment induces decreased apical but increased basolateral expression of AQP2 in CCD of rat kidney. Am J Physiol Renal Physiol. 2007;293:F87–99.
    1. Babey M, Kopp P, Robertson GL. Familial forms of diabetes insipidus: clinical and molecular characteristics. Nat Rev Endocrinol. 2011;7:701–14.
    1. Bichet DG. Hereditary polyuric disorders: new concepts and differential diagnosis. Semin Nephrol. 2006;26:224–33.
    1. Devonald MA, Karet FE. Renal epithelial traffic jams and one-way streets. J Am Soc Nephrol. 2004;15:1370–81.
    1. Deen PM, Marr N, Kamsteeg EJ, et al. Nephrogenic diabetes insipidus. Curr Opin Nephrol Hypertens. 2000;9:591–5.
    1. Slusarz MJ, Gieldon A, Slusarz R, et al. Analysis of interactions responsible for vasopressin binding to human neurohypophyseal hormone receptors-molecular dynamics study of the activated receptor-vasopressin-G(alpha) systems. J Pept Sci. 2006;12:180–9.
    1. Slusarz MJ, Slusarz R, Ciarkowski J. Molecular dynamics simulation of human neurohypophyseal hormone receptors complexed with oxytocin-modeling of an activated state. J Pept Sci. 2006;12:171–9.
    1. Bichet DG, Razi M, Lonergan M, et al. Hemodynamic and coagulation responses to 1-desamino[8-D-arginine] vasopressin in patients with congenital nephrogenic diabetes insipidus. N Engl J Med. 1988;318:881–7.
    1. Knops NB, Bos KK, Kerstjens M, et al. Nephrogenic diabetes insipidus in a patient with L1 syndrome: a new report of a contiguous gene deletion syndrome including L1CAM and AVPR2. Am J Med Genet A. 2008;146A:1853–8.
    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:F257–70.
    1. Yun J, Schoneberg T, Liu J, et al. Generation and phenotype of mice harboring a nonsense mutation in the V2 vasopressin receptor gene. J Clin Invest. 2000;106:1361–71.
    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;119:3115–26.
    1. Schliebe N, Strotmann R, Busse K, et al. V2 vasopressin receptor deficiency causes changes in expression and function of renal and hypothalamic components involved in electrolyte and water homeostasis. Am J Physiol Renal Physiol. 2008;295:F1177–90.
    1. Knoers NV, Deen PM. Molecular and cellular defects in nephrogenic diabetes insipidus. Pediatr Nephrol. 2001;16:1146–52.
    1. Wesche D, Deen PM, Knoers NV. Congenital nephrogenic diabetes insipidus: the current state of affairs. Pediatr Nephrol. 2012;27:2183–204.
    1. Bichet DG. Nephrogenic diabetes insipidus. Am J Med. 1998;105:431–42.
    1. Knoers N, Monnens LA. Nephrogenic diabetes insipidus: clinical symptoms, pathogenesis, genetics and treatment. Pediatr Nephrol. 1992;6:476–82.
    1. Heinke F, Labudde D. Membrane protein stability analyses by means of protein energy profiles in case of nephrogenic diabetes insipidus. Comput Math Methods Med. 2012;2012:790281.
    1. Sasaki S. Nephrogenic diabetes insipidus: update of genetic and clinical aspects. Nephrol Dial Transplant. 2004;19:1351–3.
    1. Hochberg Z, Van Lieburg A, Even L, et al. Autosomal recessive nephrogenic diabetes insipidus caused by an aquaporin-2 mutation. J Clin Endocrinol Metab. 1997;82:686–9.
    1. Kamsteeg EJ, Bichet DG, Konings IB, et al. Reversed polarized delivery of an aquaporin-2 mutant causes dominant nephrogenic diabetes insipidus. J Cell Biol. 2003;163:1099–109.
    1. Boone M, Deen PM. Congenital nephrogenic diabetes insipidus: what can we learn from mouse models? Exp Physiol. 2009;94:186–90.
    1. Frokiaer J, Marples D, Knepper MA, et al. Bilateral ureteral obstruction downregulates expression of vasopressin-sensitive AQP-2 water channel in rat kidney. Am J Physiol. 1996;270:F657–68.
    1. Frokiaer J, Christensen BM, Marples D, et al. Downregulation of aquaporin-2 parallels changes in renal water excretion in unilateral ureteral obstruction. Am J Physiol. 1997;273:F213–23.
    1. Li C, Wang W, Knepper MA, et al. Downregulation of renal aquaporins in response to unilateral ureteral obstruction. Am J Physiol Renal Physiol. 2003;284:F1066–79.
    1. Kim SW, Cho SH, Oh BS, et al. Diminished renal expression of aquaporin water channels in rats with experimental bilateral ureteral obstruction. J Am Soc Nephrol. 2001;12:2019–28.
    1. Jensen AM, Li C, Praetorius HA, et al. Angiotensin II mediates downregulation of aquaporin water channels and key renal sodium transporters in response to urinary tract obstruction. Am J Physiol Renal Physiol. 2006;291:F1021–32.
    1. Norregaard R, Jensen BL, Li C, et al. COX-2 inhibition prevents downregulation of key renal water and sodium transport proteins in response to bilateral ureteral obstruction. Am J Physiol Renal Physiol. 2005;289:F322–33.
    1. Norregaard R, Jensen BL, Topcu SO, et al. COX-2 activity transiently contributes to increased water and NaCl excretion in the polyuric phase after release of ureteral obstruction. Am J Physiol Renal Physiol. 2007;292:F1322–33.
    1. Stone KA. Lithium-induced nephrogenic diabetes insipidus. J Am Board Fam Pract. 1999;12:43–7.
    1. Garofeanu CG, Weir M, Rosas-Arellano MP, et al. Causes of reversible nephrogenic diabetes insipidus: a systematic review. Am J Kidney Dis. 2005;45:626–37.
    1. Grunfeld JP, Rossier BC. Lithium nephrotoxicity revisited. Nat Rev Nephrol. 2009;5:270–6.
    1. Marples D, Christensen S, Christensen EI, et al. Lithium-induced downregulation of aquaporin-2 water channel expression in rat kidney medulla. J Clin Invest. 1995;95:1838–45.
    1. Laursen UH, Pihakaski-Maunsbach K, Kwon TH, et al. Changes of rat kidney AQP2 and Na, K-ATPase mRNA expression in lithium-induced nephrogenic diabetes insipidus. Nephron Exp Nephrol. 2004;97:e1–16.
    1. Sands JM, Naruse M, Baum M, et al. Apical extracellular calcium/polyvalent cation-sensing receptor regulates vasopressin-elicited water permeability in rat kidney inner medullary collecting duct. J Clin Invest. 1997;99:1399–405.
    1. Bustamante M, Hasler U, Leroy V, et al. Calcium-sensing receptor attenuates AVP-induced aquaporin-2 expression via a calmodulin-dependent mechanism. J Am Soc Nephrol. 2008;19:109–16.
    1. Rao R, Patel S, Hao C, et al. GSK3beta mediates renal response to vasopressin by modulating adenylate cyclase activity. J Am Soc Nephrol. 2010;21:428–37.
    1. Nielsen J, Hoffert JD, Knepper MA, et al. Proteomic analysis of lithium-induced nephrogenic diabetes insipidus: mechanisms for aquaporin 2 down-regulation and cellular proliferation. Proc Natl Acad Sci USA. 2008;105:3634–9.
    1. Roth H, Becker KL, Shalhoub RJ, et al. Nephrotoxicity of demethylchlortetracycline hydrochloride. A prospective study. Arch Intern Med. 1967;120:433–5.
    1. Navarro JF, Quereda C, Quereda C, et al. Nephrogenic diabetes insipidus and renal tubular acidosis secondary to foscarnet therapy. Am J Kidney Dis. 1996;27:431–4.
    1. Metzger NL, Varney Gill KL. Nephrogenic diabetes insipidus induced by two amphotericin B liposomal formulations. Pharmacotherapy. 2009;29:613–20.
    1. Skinner R. Chronic ifosfamide nephrotoxicity in children. Med Pediatr Oncol. 2003;41:190–7.
    1. Radin MJ, Yu MJ, Stoedkilde L, et al. Aquaporin-2 regulation in health and disease. Vet Clin Pathol. 2012;41:455–70.
    1. Rutecki GW, Cox JW, Robertson GW, et al. Urinary concentrating ability and antidiuretic hormone responsiveness in the potassium-depleted dog. J Lab Clin Med. 1982;100:53–60.
    1. Berl T, Linas SL, Aisenbrey GA, et al. On the mechanism of polyuria in potassium depletion. The role of polydipsia. J Clin Invest. 1977;60:620–5.
    1. Marples D, Frokiaer J, Dorup J, et al. Hypokalemia-induced downregulation of aquaporin-2 water channel expression in rat kidney medulla and cortex. J Clin Invest. 1996;97:1960–8.
    1. Amlal H, Krane CM, Chen Q, et al. Early polyuria and urinary concentrating defect in potassium deprivation. Am J Physiol Renal Physiol. 2000;279:F655–63.
    1. Procino G, Carmosino M, Tamma G, et al. Extracellular calcium antagonizes forskolin-induced aquaporin 2 trafficking in collecting duct cells. Kidney Int. 2004;66:2245–55.
    1. Procino G, Mastrofrancesco L, Tamma G, et al. Calcium-sensing receptor and aquaporin 2 interplay in hypercalciuria-associated renal concentrating defect in humans. An in vivo and in vitro study. PLoS ONE. 2012;7:e33145.
    1. Pretzer SD. Clinical presentation of canine pyometra and mucometra: a review. Theriogenology. 2008;70:359–63.
    1. Kenney KJ, Matthiesen DT, Brown NO, et al. Pyometra in cats: 183 cases (1979-1984) J Am Vet Med Assoc. 1987;191:1130–2.
    1. Grinevich V, Knepper MA, Verbalis J, et al. Acute endotoxemia in rats induces down-regulation of V2 vasopressin receptors and aquaporin-2 content in the kidney medulla. Kidney Int. 2004;65:54–62.
    1. Hasler U, Leroy V, Jeon US, et al. NF-kappaB modulates aquaporin-2 transcription in renal collecting duct principal cells. J Biol Chem. 2008;283:28095–105.
    1. Hocherl K, Schmidt C, Kurt B, et al. Inhibition of NF-kappaB ameliorates sepsis-induced downregulation of aquaporin-2/V2 receptor expression and acute renal failure in vivo. Am J Physiol Renal Physiol. 2010;298:F196–204.
    1. Stone EA, Littman MP, Robertson JL, et al. Renal dysfunction in dogs with pyometra. J Am Vet Med Assoc. 1988;193:457–64.
    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;106:1115–26.
    1. Morishita T, Tsutsui M, Shimokawa H, et al. Nephrogenic diabetes insipidus in mice lacking all nitric oxide synthase isoforms. Proc Natl Acad Sci. 2005;102:10616–21.
    1. Bouley R, Pastor-Soler N, Cohen O, et al. 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. 2005;288:F1103–12.
    1. Sanches TR, Volpini RA, Massola Shimizu MH, et al. Sildenafil reduces polyuria in rats with lithium-induced NDI. AJP Renal Physiol. 2011;302:F216–25.
    1. Sugimoto Y, Narumiya S. Prostaglandin E receptors. J Biol Chem. 2006;282:11613–7.
    1. Breyer MD, Breyer RM. G Protein–coupled prostanoid receptors and the kidney. Annu Rev Physiol. 2001;63:579–605.
    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;108:12949–54.
    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. Robinson AG. DDAVP in the treatment of central diabetes insipidus. N Engl J Med. 1976;294:507–11.
    1. Muller D, Marr N, Ankermann T, et al. Desmopressin for nocturnal enuresis in nephrogenic diabetes insipidus. Lancet. 2002;359:495–7.
    1. Yoshida M, Iwasaki Y, Asai M, et al. Gene therapy for central diabetes insipidus: effective antidiuresis by muscle-targeted gene transfer. Endocrinology. 2004;145:261–8.
    1. Soylu A, Kasap B, Ogun N, et al. Efficacy of COX-2 inhibitors in a case of congenital nephrogenic diabetes insipidus. Pediatr Nephrol. 2005;20:1814–7.
    1. Pattaragarn A, Alon US. Treatment of congenital nephrogenic diabetes insipidus by hydrochlorothiazide and cyclooxygenase-2 inhibitor. Pediatr Nephrol. 2003;18:1073–6.
    1. Hochberg Z, Even L, Danon A. Amelioration of polyuria in nephrogenic diabetes insipidus due to aquaporin-2 deficiency. Clin Endocrinol. 1998;49:39–44.
    1. Earley LE, Orloff J. The mechanism of antidiuresis associated with the administration of hydrochlorothiazide to patients with vasopressin-resistant diabetes insipidus. J Clin Invest. 1962;41:1988–97.
    1. Knoers N, Monnens LA. Amiloride-hydrochlorothiazide versus indomethacin-hydrochlorothiazide in the treatment of nephrogenic diabetes insipidus. J Pediatr. 1990;117:499–502.
    1. Monnens L, Jonkman A, Thomas C. Response to indomethacin and hydrochlorothiazide in nephrogenic diabetes insipidus. Clin Sci. 1984;66:709–15.
    1. Libber S, Harrison H, Spector D. Treatment of nephrogenic diabetes insipidus with prostaglandin synthesis inhibitors. J Pediatr. 1986;108:305–11.
    1. Wade JB. Statins affect AQP2 traffic. AJP Renal Physiol. 2011;301:F308.
    1. Procino G, Barbieri C, Carmosino M, et al. Lovastatin-induced cholesterol depletion affects both apical sorting and endocytosis of aquaporin-2 in renal cells. Am J Physiol Renal Physiol. 2010;298:F266–78.
    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;462:753–66.
    1. Procino G, Milano S, Carmosino M, et al. Combination of secretin and fluvastatin ameliorates the polyuria associated with X-linked nephrogenic diabetes insipidus in mice. Kidney Int. 2014;86:127–38.
    1. Takai Y, Sasaki T, Matozaki T. Small GTP-binding proteins. Physiol Rev. 2001;81:153–208.
    1. Sidaway JE, Davidson RG, McTaggart F, et al. Inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase reduce receptor-mediated endocytosis in opossum kidney cells. J Am Soc Nephrol. 2004;15:2258–65.
    1. Maher BM, Dhonnchu TN, Burke JP, et al. Statins alter neutrophil migration by modulating cellular Rho activity-a potential mechanism for statins-mediated pleotropic effects? J Leukoc Biol. 2009;85:186–93.
    1. Rikitake Y, Liao JK. Rho GTPases, statins, and nitric oxide. Circ Res. 2005;97:1232–5.
    1. Ridley AJ. Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. Trends Cell Biol. 2006;16:522–9.
    1. Zelenina M, Christensen BM, Palmer J, et al. Prostaglandin E(2) interaction with AVP: effects on AQP2 phosphorylation and distribution. Am J Physiol Renal Physiol. 2000;278:F388–94.
    1. Laycock JF, Lee J, Lewis AF. The effect of chlorpropamide on water balance in pitressin-treated Brattleboro rats. Br J Pharmacol. 1974;52:253–63.
    1. Hall A. Rho GTPases and the control of cell behaviour. Biochem Soc Trans. 2005;33:891–5.
    1. Brown LM, Rogers SJ, Cello JP, et al. Cost-effective treatment of patients with symptomatic cholelithiasis and possible common bile duct stones. J Am Coll Surg. 2011;212:1049–60. e1–7.
    1. Pediconi MF, Gallegos CE, De Los Santos EB, et al. Metabolic cholesterol depletion hinders cell-surface trafficking of the nicotinic acetylcholine receptor. Neuroscience. 2004;128:239–49.
    1. Hansen GH, Niels-Christiansen LL, Thorsen E, et al. Cholesterol depletion of enterocytes. Effect on the Golgi complex and apical membrane trafficking. J Biol Chem. 2000;275:5136–42.
    1. Sparrow CP, Burton CA, Hernandez M, et al. Simvastatin has anti-inflammatory and antiatherosclerotic activities independent of plasma cholesterol lowering. Arterioscler Thromb Vasc Biol. 2001;21:115–21.
    1. Ni W, Egashira K, Kataoka C, et al. Antiinflammatory and antiarteriosclerotic actions of HMG-CoA reductase inhibitors in a rat model of chronic inhibition of nitric oxide synthesis. Circ Res. 2001;89:415–21.
    1. Dimmeler S, Aicher A, Vasa M, et al. HMG-CoA reductase inhibitors (statins) increase endothelial progenitor cells via the PI 3-kinase/Akt pathway. J Clin Invest. 2001;108:391–7.
    1. Kita T, Brown MS, Goldstein JL. Feedback regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase in livers of mice treated with mevinolin, a competitive inhibitor of the reductase. J Clin Invest. 1980;66:1094–100.
    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:F246–53.
    1. Wang Y, Thiele C, Huttner WB. Cholesterol is required for the formation of regulated and constitutive secretory vesicles from the trans-Golgi network. Traffic. 2000;1:952–62.
    1. Heiniger HJ, Kandutsch AA, Chen HW. Depletion of L-cell sterol depresses endocytosis. Nature. 1976;263:515–7.
    1. Krukoff TL, Calaresu FR. Exogenous vasopressin reverses hyperactivity in the hypothalamus of Brattleboro rats. Am J Physiol. 1984;247:R932–5.
    1. Tiwari S, Blasi ER, Heyen JR, et al. Time course of AQP-2 and ENaC regulation in the kidney in response to PPAR agonists associated with marked edema in rats. Pharmacol Res. 2008;57:383–92.
    1. Song J, Knepper MA, Hu X, et al. Rosiglitazone activates renal sodium- and water-reabsorptive pathways and lowers blood pressure in normal rats. J Pharmacol Exp Ther. 2004;308:426–33.
    1. McKenney JM. Pharmacologic characteristics of statins. Clin Cardiol. 2003;26:III32–8.
    1. Paulsen L, Holm C, Bech JN, et al. Effects of statins on renal sodium and water handling. Nephrol Dial Transplant. 2008;23:1556–61.
    1. Alsheikh-Ali AA, Ambrose MS, Kuvin JT, et al. The safety of rosuvastatin as used in common clinical practice: a postmarketing analysis. Circulation. 2005;111:3051–7.
    1. Vidt DG, Cressman MD, Harris S, et al. Rosuvastatin-induced arrest in progression of renal disease. Cardiology. 2004;102:52–60.
    1. Brewer HB., Jr Benefit-risk assessment of Rosuvastatin 10 to 40 milligrams. Am J Cardiol. 2003;92:23K–9K.
    1. Kostapanos MS, Milionis HJ, Saougos VG, et al. Dose-dependent effect of rosuvastatin treatment on urinary protein excretion. J Cardiovasc Pharmacol Ther. 2007;12:292–7.
    1. Wang L, Ellis MJ, Gomez JA, et al. Mechanisms of the proteinuria induced by Rho GTPases. Kidney Int. 2012;81:1075–85.
    1. Ellis S, Mellor H. Regulation of endocytic traffic by rho family GTPases. Trends Cell Biol. 2000;10:85–8.
    1. Pizon V, Desjardins M, Bucci C, et al. Association of Rap1a and Rap1b proteins with late endocytic/phagocytic compartments and Rap2a with the Golgi complex. J Cell Sci. 1994;107(Pt 6):1661–70.
    1. Verhulst A, D'Haese PC, De Broe ME. Inhibitors of HMG-CoA reductase reduce receptor-mediated endocytosis in human kidney proximal tubular cells. J Am Soc Nephrol. 2004;15:2249–57.
    1. Grundy SM. The issue of statin safety: where do we stand? Circulation. 2005;111:3016–9.
    1. Garcia-Rodriguez LA, Masso-Gonzalez EL, Wallander MA, et al. The safety of rosuvastatin in comparison with other statins in over 100,000 statin users in UK primary care. Pharmacoepidemiol Drug Saf. 2008;17:943–52.
    1. Garcia-Rodriguez LA, Gonzalez-Perez A, Stang MR, et al. The safety of rosuvastatin in comparison with other statins in over 25,000 statin users in the Saskatchewan Health Databases. Pharmacoepidemiol Drug Saf. 2008;17:953–61.
    1. Guthrie RM, Martin DR. The safety of rosuvastatin: effects on renal and hepatic function. Expert Opin Drug Saf. 2007;6:573–81.
    1. McAfee AT, Ming EE, Seeger JD, et al. The comparative safety of rosuvastatin: a retrospective matched cohort study in over 48,000 initiators of statin therapy. Pharmacoepidemiol Drug Saf. 2006;15:444–53.
    1. Kassimatis TI, Konstantinopoulos PA. The role of statins in chronic kidney disease (CKD): friend or foe? Pharmacol Ther. 2009;122:312–23.
    1. McKenney JM, Davidson MH, Jacobson TA, et al. Final conclusions and recommendations of the National Lipid Association Statin Safety Assessment Task Force. Am J Cardiol. 2006;97:89C–94C.
    1. Shepherd J, Vidt DG, Miller E, et al. Safety of rosuvastatin: update on 16,876 rosuvastatin-treated patients in a multinational clinical trial program. Cardiology. 2007;107:433–43.
    1. Sandhu S, Wiebe N, Fried LF, et al. Statins for improving renal outcomes: a meta-analysis. J Am Soc Nephrol. 2006;17:2006–16.
    1. Tonelli M, Moye L, Sacks FM, et al. Pravastatin for secondary prevention of cardiovascular events in persons with mild chronic renal insufficiency. Ann Intern Med. 2003;138:98–104.
    1. Verhulst A, Geryl H, D'Haese P. No evidence for statin-induced proteinuria in healthy volunteers as assessed by proteomic analysis. J Biomed Biotechnol. 2011;2011:456076.
    1. Wilson SH, Chade AR, Feldstein A, et al. Lipid-lowering-independent effects of simvastatin on the kidney in experimental hypercholesterolaemia. Nephrol Dial Transplant. 2003;18:703–9.
    1. Kido M, Ando K, Oba S, et al. Renoprotective effect of pravastatin in salt-loaded Dahl salt-sensitive rats. Hypertens Res. 2005;28:1009–15.
    1. van Dijk MA, Kamper AM, van Veen S, et al. Effect of simvastatin on renal function in autosomal dominant polycystic kidney disease. Nephrol Dial Transplant. 2001;16:2152–7.
    1. Athyros VG, Mikhailidis DP, Liberopoulos EN, et al. Effect of statin treatment on renal function and serum uric acid levels and their relation to vascular events in patients with coronary heart disease and metabolic syndrome: a subgroup analysis of the GREek Atorvastatin and Coronary heart disease Evaluation (GREACE) Study. Nephrol Dial Transplant. 2007;22:118–27.
    1. Atthobari J, Brantsma AH, Gansevoort RT, et al. The effect of statins on urinary albumin excretion and glomerular filtration rate: results from both a randomized clinical trial and an observational cohort study. Nephrol Dial Transplant. 2006;21:3106–14.
    1. Bruckert E, Hayem G, Dejager S, et al. Mild to moderate muscular symptoms with high-dosage statin therapy in hyperlipidemic patients–the PRIMO study. Cardiovasc Drugs Ther. 2005;19:403–14.
    1. Bjornsson E, Jacobsen EI, Kalaitzakis E. Hepatotoxicity associated with statins: reports of idiosyncratic liver injury post-marketing. J Hepatol. 2012;56:374–80.
    1. Cohen DE, Anania FA, Chalasani N National Lipid Association Statin Safety Task Force Liver Expert P. An assessment of statin safety by hepatologists. Am J Cardiol. 2006;97:77C–81C.
    1. Scandinavian Simvastatin Survival Study Group. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S) Lancet. 1994;344:1383–9.
    1. Pfeffer MA, Keech A, Sacks FM, et al. Safety and tolerability of pravastatin in long-term clinical trials: prospective Pravastatin Pooling (PPP) Project. Circulation. 2002;105:2341–6.
    1. Smith CC, Bernstein LI, Davis RB, et al. Screening for statin-related toxicity: the yield of transaminase and creatine kinase measurements in a primary care setting. Arch Intern Med. 2003;163:688–92.
    1. Wagstaff LR, Mitton MW, Arvik BM, et al. Statin-associated memory loss: analysis of 60 case reports and review of the literature. Pharmacotherapy. 2003;23:871–80.
    1. Preiss D, Seshasai SR, Welsh P, et al. Risk of incident diabetes with intensive-dose compared with moderate-dose statin therapy: a meta-analysis. JAMA. 2011;305:2556–64.
    1. Ness GC, Zhao Z, Lopez D. Inhibitors of cholesterol biosynthesis increase hepatic low-density lipoprotein receptor protein degradation. Arch Biochem Biophys. 1996;325:242–8.
    1. Conde K, Vergara-Jimenez M, Krause BR, et al. Hypocholesterolemic actions of atorvastatin are associated with alterations on hepatic cholesterol metabolism and lipoprotein composition in the guinea pig. J Lipid Res. 1996;37:2372–82.
    1. Arad Y, Ramakrishnan R, Ginsberg HN. Lovastatin therapy reduces low density lipoprotein apoB levels in subjects with combined hyperlipidemia by reducing the production of apoB-containing lipoproteins: implications for the pathophysiology of apoB production. J Lipid Res. 1990;31:567–82.
    1. Collins R, Armitage J, Parish S, et al. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet. 2003;361:2005–16.
    1. Marchesi S, Lupattelli G, Siepi D, et al. Short-term atorvastatin treatment improves endothelial function in hypercholesterolemic women. J Cardiovasc Pharmacol. 2000;36:617–21.
    1. Cannon CP, Braunwald E, McCabe CH, et al. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med. 2004;350:1495–504.
    1. Zwaka TP, Hombach V, Torzewski J. C-reactive protein-mediated low density lipoprotein uptake by macrophages: implications for atherosclerosis. Circulation. 2001;103:1194–7.
    1. Kleemann R, Verschuren L, de Rooij BJ, et al. Evidence for anti-inflammatory activity of statins and PPARalpha activators in human C-reactive protein transgenic mice in vivo and in cultured human hepatocytes in vitro. Blood. 2004;103:4188–94.
    1. Kaski JC. Infection, endothelial dysfunction, and atherogenesis. Circulation. 2003;108:E171–2.
    1. Callahan AS., 3rd Vascular pleiotropy of statins: clinical evidence and biochemical mechanisms. Curr Atheroscler Rep. 2003;5:33–7.
    1. Levy Y. Beyond cholesterol lowering: effect of statins on markers of cardiovascular disease. Isr Med Assoc J. 2004;6:490–1.
    1. Fassbender K, Simons M, Bergmann C, et al. Simvastatin strongly reduces levels of Alzheimer's disease beta -amyloid peptides Abeta 42 and Abeta 40 in vitro and in vivo. Proc Natl Acad Sci USA. 2001;98:5856–61.
    1. Snowdon DA, Greiner LH, Mortimer JA, et al. Brain infarction and the clinical expression of Alzheimer disease. The Nun Study. JAMA. 1997;277:813–7.
    1. Tonelli M, Moye L, Sacks FM, et al. Effect of pravastatin on loss of renal function in people with moderate chronic renal insufficiency and cardiovascular disease. J Am Soc Nephrol. 2003;14:1605–13.
    1. Ruan F, Zheng Q, Wang J. Mechanisms of bone anabolism regulated by statins. Biosci Rep. 2012;32:511–9.
    1. Chan KA, Andrade SE, Boles M, et al. Inhibitors of hydroxymethylglutaryl-coenzyme A reductase and risk of fracture among older women. Lancet. 2000;355:2185–8.
    1. Wang PS, Solomon DH, Mogun H, et al. HMG-CoA reductase inhibitors and the risk of hip fractures in elderly patients. JAMA. 2000;283:3211–6.
    1. Lawes CM, Thornley S, Young R, et al. Statin use in COPD patients is associated with a reduction in mortality: a national cohort study. Prim Care Respir J. 2012;21:35–40.
    1. Young RP, Hopkins R, Eaton TE. Pharmacological actions of statins: potential utility in COPD. Eur Respir Rev. 2009;18:222–32.
    1. Gokce MI, Gulpinar O, Ozturk E, et al. Effect of atorvastatin on erectile functions in comparison with regular tadalafil use. A prospective single-blind study. Int Urol Nephrol. 2012;44:683–7.
    1. La Vignera S, Condorelli RA, Vicari E, et al. Statins and erectile dysfunction: a critical summary of current evidence. J Androl. 2012;33:552–8.
    1. Di Ciaula A, Wang DQ, Wang HH, et al. Targets for current pharmacologic therapy in cholesterol gallstone disease. Gastroenterol Clin North Am. 2010;39:245–64. viii-ix.
    1. Bodmer M, Brauchli YB, Krahenbuhl S, et al. Statin use and risk of gallstone disease followed by cholecystectomy. JAMA. 2009;302:2001–7.
    1. Bouley R, Hasler U, Lu HA, et al. Bypassing vasopressin receptor signaling pathways in nephrogenic diabetes insipidus. Semin Nephrol. 2008;28:266–78.
    1. Kim SW, Kim JW, Choi KC, et al. Indomethacin enhances shuttling of aquaporin-2 despite decreased abundance in rat kidney. J Am Soc Nephrol. 2004;15:2998–3005.
    1. Shaw S, Marples D. N-ethylmaleimide causes aquaporin-2 trafficking in the renal inner medullary collecting duct by direct activation of protein kinase A. Am J Physiol Renal Physiol. 2005;288:F832–9.
    1. Schrier RW, Ohara M, Rogachev B, et al. Aquaporin-2 water channels and vasopressin antagonists in edematous disorders. Mol Genet Metab. 1998;65:255–63.
    1. Batlle DC, von Riotte AB, Gaviria M, et al. Amelioration of polyuria by amiloride in patients receiving long-term lithium therapy. N Engl J Med. 1985;312:408–14.
    1. Kirchlechner V, Koller DY, Seidl R, et al. Treatment of nephrogenic diabetes insipidus with hydrochlorothiazide and amiloride. Arch Dis Child. 1999;80:548–52.
    1. Kim GH, Lee JW, Oh YK, et al. Antidiuretic effect of hydrochlorothiazide in lithium-induced nephrogenic diabetes insipidus is associated with upregulation of aquaporin-2, Na-Cl co-transporter, and epithelial sodium channel. J Am Soc Nephrol. 2004;15:2836–43.
    1. Stokes JB. Integrated actions of renal medullary prostaglandins in the control of water excretion. Am J Physiol. 1981;240:F471–80.
    1. Berl T, Raz A, Wald H, et al. Prostaglandin synthesis inhibition and the action of vasopressin: studies in man and rat. Am J Physiol. 1977;232:F529–37.
    1. Bernier V, Morello JP, Zarruk A, et al. Pharmacologic chaperones as a potential treatment for X-linked nephrogenic diabetes insipidus. J Am Soc Nephrol. 2006;17:232–43.
    1. Morello JP, Salahpour A, Laperriere A, et al. Pharmacological chaperones rescue cell-surface expression and function of misfolded V2 vasopressin receptor mutants. J Clin Invest. 2000;105:887–95.
    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;20:2190–203.
    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;106:12195–200.
    1. Robben JH, Sze M, Knoers NV, et al. Functional rescue of vasopressin V2 receptor mutants in MDCK cells by pharmacochaperones: relevance to therapy of nephrogenic diabetes insipidus. Am J Physiol Renal Physiol. 2007;292:F253–60.
    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;22:393–9.
    1. Yang B, Zhao D, Verkman AS. Hsp90 inhibitor partially corrects nephrogenic diabetes insipidus in a conditional knock-in mouse model of aquaporin-2 mutation. FASEB J. 2009;23:503–12.
    1. Sohara E, Rai T, Yang SS, et al. Pathogenesis and treatment of autosomal-dominant nephrogenic diabetes insipidus caused by an aquaporin 2 mutation. Proc Natl Acad Sci USA. 2006;103:14217–22.
    1. de Rouffignac C, Elalouf JM. Effects of calcitonin on the renal concentrating mechanism. Am J Physiol. 1983;245:F506–11.
    1. Hoffert JD, Fenton RA, Moeller HB, et al. Vasopressin-stimulated increase in phosphorylation at Ser269 potentiates plasma membrane retention of aquaporin-2. J Biol Chem. 2008;283:24617–27.
    1. Bichet DG. Nephrogenic and central diabetes insipidus. In: Coffman TM, Falk RJ, Molitoris BA, Neilson EG, editors. Schrier's diseases of the kidney. Philadelphia: Lippincott Williams & Wilkins; 2012. pp. 2055–81. 9th ed.
    1. Eckardt KU, Coresh J, Devuyst O, et al. Evolving importance of kidney disease: from subspecialty to global health burden. Lancet. 2013;382:158–69.

Source: PubMed

3
订阅