NDT Advance Access published October 3, 2014 Nephrol Dial Transplant (2014) 0: 1–10 doi: 10.1093/ndt/gfu307

Full Review The multiple roles of pendrin in the kidney Manoocher Soleimani1,2,3 1

Center on Genetics of Transport and Epithelial Biology, University of Cincinnati, Cincinnati, OH, USA, 2Research Services, Veterans Affairs

Correspondence and offprint requests to: Manoocher Soleimani; E-mail: [email protected]; soleimm@ ucmail.uc.edu

A B S T R AC T The Cl =HCO3 exchanger pendrin (SLC26A4, PDS) is located on the apical membrane of B-intercalated cells in the kidney cortical collecting duct and the connecting tubules and mediates the secretion of bicarbonate and the reabsorption of chloride. Given its dual function of bicarbonate secretion and chloride reabsorption in the distal tubules, it was thought that pendrin plays important roles in systemic acid–base balance and electrolyte and vascular volume homeostasis under basal conditions. Mice with the genetic deletion of pendrin or humans with inactivating mutations in PDS gene, however, do not display excessive salt and fluid wasting or altered blood pressure under baseline conditions. Very recent reports have unmasked the basis of incongruity between the mild phenotype in mutant mice and the role of pendrin as an important player in salt reabsorption in the distal tubule. These studies demonstrate that pendrin and the Na–Cl cotransporter (NCC; SLC12A3) cross compensate for the loss of each other, therefore masking the role that each transporter plays in salt reabsorption under baseline conditions. In addition, pendrin regulates calcium reabsorption in the distal tubules. Furthermore, combined deletion of pendrin and NCC not only causes severe volume depletion but also results in profound calcium wasting and luminal calcification in medullary collecting ducts. Based on studies in pathophysiological states and the examination of genetically engineered mouse models, the evolving picture points to important roles for pendrin (SLC26A4) in kidney physiology and in disease states. This review summarizes recent advances in the characterization of pendrin and the multiple roles it plays in the kidney, with emphasis on its essential roles in several diverse physiological processes, including chloride homeostasis, vascular volume and blood pressure regulation, calcium excretion and kidney stone formation. © The Author 2014. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved.

Keywords: hypertension, oxalate stone, renal tubular acidosis, salt excretion, volume depletion

INTRODUCTION The reabsorption of sodium and chloride represents a major function of kidney tubules and is essential for the vascular volume homeostasis and blood pressure regulation. The bulk of filtered sodium and chloride is reabsorbed in the proximal tubule and the thick ascending limb of Henle (TALH), with the remaining load being reabsorbed in the more distal tubule segments, including the collecting duct [1–9]. Both trans- and para-cellular pathways participate in the reabsorption of salt with varying proportions in different kidney segments [1–9]. The trans-cellular reabsorption of sodium is predominantly mediated via the Na+/H+ exchanger NHE3 in the proximal tubule, by the apical Na+–K+–2Cl− cotransporter (NKCC2) and NHE3 in the TALH, through the Na+–Cl− cotransporter (NCC; SLC12A3) in the distal convoluted tubule (DCT) and by the epithelial sodium channel (ENaC) in the connecting tubule (CNT) and collecting duct [1–4]. Chloride reabsorption involves both para- and trans-cellular pathways, with the trans-cellular pathway mediated via apical chloride/base exchangers in the proximal tubule and the collecting duct, by NKCC2 (SLC12A2) in TALH and through NCC (SLC12A3) in the DCT [1–9]. The para-cellular reabsorption of chloride is predominantly secondary to the gradients generated by trans-cellular absorption of sodium and occurs through tight junctions [9–11]. The apical chloride/ base exchangers in the proximal tubule work in tandem with NHE3 [1, 5, 7, 8], whereas in the collecting duct they primarily function in collaboration with the ENaC and in part with the 1

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

Medical Center, Cincinnati, OH, USA and 3Department of Medicine, University of Cincinnati, Cincinnati, OH, USA

SLC26 family of anion transporters The main apical chloride/base exchangers in epithelial tissues, including the kidney and gastrointestinal tract, belong to the family of solute transporters SLC26A [21–30]. This family is genetically distinct from the SLC4 family of anion transporters, which encompasses AE1, AE2, AE3 and AE4, and can function as Cl =HCO3 exchangers [31]. The SCL26 family comprises 11 distinct members (SLC26A1–11) [21–30]. Modes of transport mediated by SLC26 members include the exchange of chloride for bicarbonate, hydroxyl, sulfate, formate, iodide or oxalate with variable specificity [5, 7, 32–36]. Several SLC26 family members can specifically function as Cl =HCO3 exchangers. These include SLC26A3 (DRA), SLC26A4 ( pendrin), SLC26A6 (PAT1 or CFEX), SLC26A7, SLC26A9 and SLC26A11 [32–42]. In addition to mediating chloride/base exchange, SLC26A7, SLC26A9 and SLC26A11 can also function as chloride channels [41–47]. Five well-known SLC26 isoforms that show distinct tubule segment distributions are SLC26A4, SLC26A6, SLC26A7, SLC26A9 and SLC26A11 [7, 46–50]. SLC26A4 is expressed on the apical membrane of a subset of intercalated cells (B-IC) and mediates chloride/bicarbonate exchange [6, 7, 18–20, 48–50]. SLC26A6 is expressed on the apical membrane of kidney proximal tubule cells and mediates chloride/oxalate and chloride/bicarbonate exchange [51–54]. SLC26A7 is

2

FULL REVIEW

expressed on the basolateral membrane of A-intercalated cells in the outer medullary collecting duct and can function as a chloride channel as well as a chloride/bicarbonate exchanger [45, 55–57]. SLC26A9 is expressed on the apical membrane of principal cells in the outer medullary and initial inner medullary collecting duct and can function as a chloride channel, an electrogenic chloride/cation cotransport and an electrogenic chloride/bicarbonate exchange [40, 41, 43, 44, 46, 58]. SLC26A11 is expressed on the apical membrane of A-intercalated cells and the basolateral membrane of B-intercalated cells in the collecting duct and can function as a chloride channel, an electrogenic chloride transporter and a chloride/bicarbonate exchanger [42, 47]. In addition to the above three isoforms, SLC26A3 (DRA) can also function as a chloride/bicarbonate exchanger but is primarily expressed in the intestine and is absent in the kidney [32, 38]. Figure 1 compares the expression pattern of the main SLC26 members between kidney and intestine and their predominant functional modes in native tissues. SLC26A4 ( pendrin): a multifaceted transporter Cloning and localization. The SLC26A4 gene was first identified by linkage analysis and positional cloning studies in patients with Pendred syndrome [24], a genetically inherited, autosomal recessive disorder characterized by deafness and thyroid enlargement or goiter [59, 60]. The pendrin gene SLC26A4 transcribes an mRNA of ∼5 kb which encodes a protein of ∼95–100 kDa [24]. Pendrin is abundantly expressed in the thyroid and inner ear [24], with very low levels in the kidney [24]. The gene encoding pendrin is located next to the gene encoding DRA (SLC26A3) on chromosome 7 [24]. They show 45% homology, suggesting ancient gene duplication. Pendrin, as well as other SLC26 isoforms, contains a C-terminal cytoplasmic fragment organized around a central Sulfate Transporter and Anti-Sigma factor antagonist (STAS) domain [7, 8]. Published reports indicate that STAS domain plays an important role in surface trafficking and functional expression for SLC26 isoforms [7, 8]. Expression of SLC26A4 in cultured kidney cells showed that it can function in Cl =HCO3 exchange, Cl−/OH− exchange and Cl−/formate exchange modes [20]. Northern hybridization and nephron segment distribution experiments showed that pendrin mRNA expression was exclusively detected in the cortex of the kidney, with high levels in the CCD and lower levels in the proximal tubule [20]. Immunofluorescence studies localized pendrin to the apical membrane of B-intercalated cells as well as non-A, non-B- intercalated cells in the CNT and CCDs, with no protein expression in the proximal tubule [18, 19, 49, 50]. Figure 2A is a schematic diagram depicting the nephron segment distribution of pendrin. Figure 2B demonstrates the cellular and subcellular distribution of pendrin in the CCD. Mutations and structure–function relationship. The SLC26A4 gene encodes a transmembrane protein of 780 amino acids, with either 11 or 12 membrane-spanning domains [24]. More than 60 mutations have been reported in the SLC26A4 gene, predominantly distributed throughout the

M. Soleimani

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

sodium-dependent chloride/bicarbonate exchanger (NDCBE; SLC4A8) to mediate the reabsorption of NaCl [5–9, 12]. In addition to salt reabsorption, the kidney plays an essential role in systemic acid–base homeostasis by eliminating excess acid and reabsorbing the filtered bicarbonate through the synchronized action of specific acid–base transporters that are expressed on the apical and basolateral membranes of various tubular segments [1, 2, 4–7, 9, 12–15]. The net effect of the action of these transporters is the maintenance of the systemic pH within a narrow physiological range, which is vital for the normal functioning of cells and tissues. The cortical collecting duct (CCD) plays a major role in acid–base regulation through acid or bicarbonate secretion in specialized intercalated cells. The acid is secreted into the lumen in A-intercalated cells predominantly via apical H+ATPase, and bicarbonate is transported to the blood via basolateral AE1 (SLC4A1) [16, 17]. Bicarbonate secretion into the lumen occurs in B-intercalated cells and is mediated via pendrin [18, 19], which exchanges luminal chloride for cellular bicarbonate [20]. Na+ reabsorption in these nephron segments and in the CNT is trans-cellular and is mediated by ENaC located on the apical membrane of the principal cell. Recent studies have demonstrated an important role for pendrin in acid–base regulation and/or systemic electrolyte homeostasis and blood pressure regulation in pathophysiologic states. The focus of this review is on pendrin, a member of a large, conserved family of anion transporters called SLC26, and the multiple roles it plays in the kidney, with emphasis on important contributions to several diverse physiological processes, including chloride homeostasis, vascular volume and blood pressure regulation, acid–base regulation, calcium excretion and kidney stone formation.

tissues. SLC26A3 (DRA), SLC26A4 ( pendrin), SLC26A6 (PAT1; CFEX), SLC26A7, SLC26A9 and SLC26A11 (KBAT) are expressed in the kidney and/or gastrointestinal tract.

conditions is the fact that pendrin KO mice have an acidic urine pH versus WT littermates [19, 63]. This latter observation is important, as it suggests that pendrin may be active under baseline conditions, and its inactivation or inhibition causes acidic urine. Alternative possibilities such as enhanced ammonium excretion in pendrin KO mice should be considered as possible explanation for reduced urine pH in these mutant animals [19, 63].

Pendrin-deficient mice. The generation of mice with a genetic deletion of pendrin has significantly advanced our understanding of the role of this exchanger in normal physiology and pathophysiological states.

Pendrin and salt-sensitive hypertension. Published reports indicate that pendrin plays an important role in aldosteronemediated hypertension. In detailed studies, Verlander et al. demonstrated that under enhanced salt intake, the aldosterone analog, deoxycorticosterone acetate, causes translocation of pendrin to the apical membrane in mouse kidney CCD and increases systemic blood pressure [65]. The same maneuver in pendrin KO mice failed to increase systemic blood pressure [65]. These results convincingly show that pendrin enhances salt absorption in the kidney collecting duct in the presence of increased aldosterone and contributes to enhanced systemic blood pressure [6, 65]. A recent report examined the role of pendrin overexpression in the pathogenesis of hypertension. Using a mouse transgenic model, the authors showed that pendrin overexpression

Pendrin and bicarbonate loading. Pendrin knockout (KO) mice showed impaired ability to excrete bicarbonate in response to bicarbonate loading, indicating an important role for this transporter in bicarbonate secretion in the kidney [18]. Studies in microperfused kidney collecting ducts showed significant down-regulation of apical Cl =HCO3 exchanger activity in B-intercalated cells in pendrin KO mice, directly verifying that pendrin is the dominant apical Cl =HCO3 exchanger in the kidney collecting duct [19]. Consistent with an important role for pendrin in bicarbonate secretion under basal condition in mice kept under standard laboratory

Pendrin and the kidney

Pendrin and salt depletion. Kidney functions, including salt excretion, glomerular filtration and systemic blood pressure, are all within the normal range in pendrin KO mice under basal conditions [18, 19, 63]. Pendrin KO mice, however, develop volume depletion, metabolic alkalosis and hypotension when subjected to salt restriction [63]. These results are congruent with a recent report indicating the development of metabolic alkalosis in a patient with Pendred syndrome during vascular volume depletion [64] and clearly support an important role for pendrin in salt absorption and bicarbonate secretion in volume-depleted states.

3

FULL REVIEW

coding sequence. The mutations are primarily missense, with a smaller number of deletions, frame-shift or insertions also reported [60, 61]. Functional analysis of wild-type and several mutations of pendrin have been carried out in various expression systems. The majority of these mutations show loss-of-function phenotypes due to misfolding and lack of membrane targeting, with some others displaying the loss of function, but with normal membrane insertion. A considerable number of patients with Pendred syndrome lack mutations in the SLC26A4 coding region in one or both alleles. Mutations in both FOXI1, a transcriptional activator of SLC26A4, and the SLC26A4 promoter that binds FOXI1, have been shown to interfere with FOXI1binding and FOXI1-mediated transcriptional activation of the SLC26A4 gene, resulting in reduced activity of pendrin [62]. No detailed studies have been performed on kidney functions, including salt excretion, divalent ion (calcium or magnesium) excretion or urine concentrating ability, in patients with Pendred syndrome.

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

F I G U R E 1 : Expression of major SLC26 transporters in the kidney and gastrointestinal tract and their predominant functional modes in native

FULL REVIEW distribution (A) and subcellular localization of pendrin in CCD cells (B). Pendrin is localized to the apical membrane of B- and non-A, non-B intercalated cells in the CCD.

along the length of the collecting duct increases systemic blood pressure when animals are subjected to increased dietary salt intake [66]. Whether gain-of-function mutations in human pendrin can result in systemic hypertension remains speculative. There are no published linkage analysis studies identifying pendrin as a candidate gene product in salt-sensitive hypertension. Regulation of pendrin in acid–base and electrolyte disorders Pendrin and acid–base disorders. Pendrin is up-regulated in bicarbonate loading and down-regulated in acidosis [67–73]. These results are consistent with an adaptive role for pendrin in eliminating excess bicarbonate in alkalosis and conserving bicarbonate in acidosis. An alternative view suggests that the up-regulation of pendrin in alkalosis could be secondary to decreased chloride intake (secondary to the consumption of a chloride-free diet and HCO3 -rich drinking water). Conversely, the down-regulation of pendrin in acidosis

4

Pendrin-dependent, sodium absorption pathways in the collecting duct: electrogenic versus electroneutral pathway. The accepted belief, based on published studies, was that pendrin-mediated Cl =HCO3 exchange in B-intercalated cells is primarily coupled to sodium absorption via ENaC in principal cells [6, 65]. However, that view has been revisited. A recent publication indicated that pendrin could work in tandem with the NDCBE to absorb sodium [12, and reviewed in 80], although the exact localization of NDCBE in CCD cells (B-intercalated cells or principal cells) remains unresolved. Such a coordinated mechanism would be electroneutral, could provide an ENaC-independent salt reabsorption pathway in

M. Soleimani

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

F I G U R E 2 : A schematic diagram depicting the nephron segment

is attributed to increased chloride intake in the form of NH4Cl added to the drinking water [74, 75]. A possible direct effect of systemic pH (acidosis or alkalosis) on pendrin expression needs to be investigated in carefully controlled experiments. Pendrin and potassium depletion. Pendrin expression is down-regulated in rats on a potassium restricted diet [67, 72]. Whether this is an adaptive or a maladaptive process remains speculative. Given the role of pendrin as a bicarbonate-secreting transporter in the collecting duct, we speculate that its downregulation may contribute to the maintenance of metabolic alkalosis in conditions such as hypokalemia. It should be noted that the induction of potassium depletion in rodents has been generally achieved by dietary potassium restriction [76], which results in reduced aldosterone secretion. However, some of the well-known pathophysiological states associated with potassium depletion (hypokalemia) are due to the treatment with diuretics, such as furosemide, which cause hypovolemia and result in enhanced aldosterone release [77], which can subsequently activate pendrin [65]. It is noteworthy to mention that potassium homeostasis has a unique effect on NCC expression and regulation. Published reports indicate that both dietary K+ depletion and dietary K+ loading provoke renal Na+ retention and increase blood pressure in Na+ replete mice, but these occur through different kinase signaling pathways and Na+ transporting molecules [78, 79]. While K+ loading decreases NCC expression and activity [78], K+ depletion activates NCC to promote sodium retention [79]. This latter view was recently emphasized at the American Society of Nephrology (Atlanta, November 2013), where well-designed studies were presented which showed that in conditions of total body potassium depletion NCC is up-regulated (Dr. David Ellison, Robert W. Schrier Endowed lecture, ASN 2013 and Andrew Terker, Chao-Ling Yang, David H. Ellison. A Western diet activates NCC to promote sodium retention. ASN annual 2013, Atlanta). The differential regulation of NCC and pendrin by potassium depletion (above and Refs. 67, 72 and 79) suggests the decoupling of these two transporters in sodium absorption in the distal tubule. It is plausible that the activation of NCC in potassium depletion is meant to maximize sodium absorption in the DCT and minimize potassium secretion in the CCD. Hypothetically, it is also plausible that NCC up-regulation in potassium depletion is secondary to the down-regulation of NKCC2 in the thick ascending limb in the same condition [76], and is an attempt to prevent massive salt wasting.

Pendrin and the kidney

Pendrin and signaling transduction pathways. The effect of protein kinase A and C on pendrin expression and/or function has been examined. In oocytes injected with pendrin cRNA, phorbol ester only mildly inhibited pendrin (SLC26A4) activity whereas it had a significant inhibitory effect on PAT-1 (SLC26A6) activity [91]. In cultured CCDs pendrin protein abundance increased in the presence of adenylyl cyclase agonist forskolin, a cAMP analog [92]. In cultured kidney cells stably transfected with pendrin cDNA and in isolated microperfused CCDs, stimulation of the cAMP– PKA pathway by isoproterenol increased the apical abundance of pendrin at the cell surface along with its transport activity [93]. These effects were mediated via increased trafficking of pendrin and were associated with its phosphorylation. These

5

FULL REVIEW

Essential role of pendrin in salt reabsorption in the setting of Na–Cl cotransporter inactivation. Based on published studies on KO mice, investigators concluded that pendrin contribution to salt absorption is minimal under basal conditions but it becomes active during salt depletion and/or in response to aldosterone [7, 65]. However, a recent study from our group indicated that pendrin and NCC cross-compensate for salt absorption under baseline conditions. Our results showed that pendrin/NCC dKO mice displayed severe salt wasting under basal conditions, which resulted in profound volume depletion, hypotension, renal failure and metabolic alkalosis [83]. Salt replacement rectified the abnormalities [83]. Pendrin- or NCC-single KO mice did not display any salt wasting or volume depletion under baseline state [83]. It was concluded that pendrin could be a novel target for a new diuretic, which in conjunction with thiazide, a specific inhibitor of NCC, could be an effective regimen for patients with fluid overload, such as those with congestive heart failure, diuretic resistance or advanced chronic kidney disease [83, 85]. It should be

noted that while the inhibition of pendrin could interfere with the thyroid function, it is unlikely that it would affect hearing acuity. It is well known that the deafness in patients with Pendred syndrome is caused by structural abnormalities of the inner ear during embroyonic development [86]. Pendrin can also transport iodide in the kidney [87]. A recent study from our laboratory extended the above observations by using a pharmacological model of salt wasting. Based on the known effect of the carbonic anhydrase inhibitor acetazolamide on B-intercalated cells in rat kidney [88], we were able to elicit a profound diuretic response in rats that were treated with a combination of hydrochlothiazide and acetazolamide, two mild diuretics that have never been used in combination as an effective diuretic regimen [89]. The results demonstrated that the treatment of rats with acetazolamide for 1 week down-regulated the expression of pendrin, leaving NCC as the major salt absorbing transporter in the distal nephron in the setting of increased delivery of salt from the proximal tubule [89]. Under these circumstances, the inhibition of NCC with the use of hydrochlorothiazide caused profound salt wasting and resulted in volume depletion and renal failure [89]. Treatment with hydrochlorothiazide alone did not cause significant diuresis [89]. These results confirm the essential role of pendrin in salt absorption in the distal nephron in the context of NCC inactivation or inhibition [83, 89]. The above studies in rats and mice [82, 83, 85] raise the possibility that the down-regulation or inactivation of pendrin can cause profound sensitivity to the diuretic effect of hydrochlorothiazide in humans. Indeed, a recent study showed that hydrochlorothiazide treatment resulted in severe volume depletion, profound metabolic alkalosis and pre-renal kidney failure in a patient with Pendred syndrome [90]. An extremely unusual presentation for thiazides, the presenting manifestations in this patient closely mimics the clinical picture in double pendrin/NCC KO mice and supports the conclusion that a functional pendrin blunts the diuretic effect of NCC inhibition subsequent to treatment with thiazides by compensatory absorption of salt in the distal nephron. The schematic diagram in Figure 4A and B depicts the synergistic effects of pendrin and NCC inhibition on salt and water excretion. Whether thiazide in combination with pendrin inhibitors is more advantageous than thiazide in combination with amiloride remains to be determined.

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

the collecting duct and is not associated with net potassium secretion [12, 80], whereas the pendrin-dependent ENaC salt absorption is associated with a net K+ secretion. The schematic diagram in Figure 3A and B depicts the two modes of salt absorption via pendrin. It is plausible that the ENaC-dependent, pendrin-mediated functional complex plays a dominant role in salt absorption in the collecting duct, specifically in the setting of NCC inactivation or inhibition. Whether the ENaCindependent (NDCBE-dependent), pendrin-mediated salt absorption could function as a substitute for the former (i.e. in the absence of ENaC) or is differentially regulated in pathophysiologic states remains to be determined. Published studies indicate that in NCC KO mice, the expression levels of both pendrin and ENaC in the distal nephron are increased [74, 81–83]. A recent study from our laboratory shows that treatment with amiloride caused significant diuresis in NCC KO but not in wild-type mice, strongly supporting an important role for ENaC in compensatory salt absorption in the setting of NCC inactivation (M. PatelChamberlin, K. Zahedi, S. Barone and M. Soleimani. The role of ENaC and the apical Cl =HCO3 exchanger pendrin in compensatory salt absorption in the distal nephron in NCC KO mice, manuscript under review; abstract presented at the ASN 2013 in Atlanta). Coupled with the massive diuresis found in pendrin/NCC double KO (dKO) mice, but not in NCC or pendrin only KO mice (see below and 83), it was proposed that pendrin-dependent, ENaC-mediated salt absorption is the dominant salt absorbing pathway in the distal nephron in the setting of NCC deletion or inactivation. These results do not conflict with published studies that show ENaC deletion in the collecting duct does not cause excess salt wasting in wild-type mice [84]. Whether pendrin-dependent, NDCBE-mediated salt absorption is differentially regulated and plays an important role in salt absorption in certain pathophysiological states remains to be determined. It is worth mentioning that for pendrin to operate in tandem with NDCBE, it should couple 2Cl− with 2HCO3 (stoichiometry of 2Cl−/2HCO3 ) to absorb chloride [80].

FULL REVIEW

effect of this synergy is the absorption of sodium and chloride and the secretion of bicarbonate. Right (B) Pendrin functioning in collaboration with NDCBE (Na-dependent chloride/bicarbonate exchanger). The net effect of this interaction is the absorption of sodium and chloride, with the recycling of bicarbonate via NDCBE and pendrin.

F I G U R E 4 : A schematic diagram highlighting the synergistic effects of pendrin and NCC inhibition on salt and water excretion. (A) Normal

condition: salt reabsorption by NCC and pendrin (working in tandem with ENaC) minimizes the amount of salt that is excreted. (B) Combined inactivation or inhibition of pendrin and NCC causes severe salt wasting.

studies strongly suggest that pendrin activation by the cAMP– PKA pathway could contribute to enhanced NaCl reabsorption in the kidney collecting duct by β-adrenergic agonists [93]. Pendrin and cystic fibrosis. Cystic fibrosis (CF) is the most common lethal genetic disease in the USA. It is caused by mutations in CFTR (cystic fibrosis transmembrane regulator), a cAMP-activated chloride channel, which is widely expressed in epithelial tissues. CFTR plays an important role in HCO3 secretion in the duodenum and pancreatic duct by activating the apical Cl =HCO3 exchangers SLC26A3 (DRA), and SLC26A6 (PAT-1) [94]. Published reports indicate that CFTR and SLC26 bicarbonate transporters construct a bicarbonatesecreting complex via binding at their PDZ-binding ligands [94]. While there are conflicting reports on nephron segment distribution of CFTR in the kidney [95–100], several studies in primary cultures, patches from kidney collecting duct and

6

cultured cells show that CFTR is abundantly expressed in various nephron segments, including the CCD [95, 97–100]. However, the exact role of CFTR in kidney physiology remains speculative. Patients with cystic fibrosis on many occasions present with metabolic alkalosis, specifically at a young age [101, 102]. While the generation of metabolic alkalosis has been attributed to volume depletion subsequent to the loss of electrolytes through the skin, it is highly plausible that pendrin, which plays a major role in chloride reabsorption and bicarbonate secretion in the distal nephron in volume-depleted states, remains inactive in patients with cystic fibrosis, resulting in the inability of the kidney to secret HCO3 . In support of this hypothesis, recent studies demonstrated that CFTR and pendrin mutually activate each other [103, 104]. These studies showed that a functional, but not a mutant CFTR activates pendrin-mediated Cl =HCO3 exchange [103, 104]. While the interaction of CFTR and pendrin was

M. Soleimani

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

F I G U R E 3 : Pendrin-dependent salt absorption in the collecting duct. Left (A) Pendrin functioning in conjunction with the ENaC. The net

F I G U R E 5 : A hypothetical diagram depicting the interaction of CFTR and pendrin in the kidney intercalated cells. (A) Control: the CFTR and

Pendrin-deficient mice and hypercalciuria. Calcium reabsorption in the DCT and CNT is mediated via apical ECaC (TRPV5), cytosolic Calbindin 28 and basolateral Na/Ca exchange and Ca2+ ATPases [105, 106]. A recent study examined the impact of pendrin deletion on calcium excretion and the expression of calcium-absorbing proteins in the distal nephron [107]. These results demonstrated that the expression of calcium-absorbing molecules (TRPV5, Calbindin 28 and the Na/Ca exchanger) were all decreased in the kidneys of pendrin KO mice [107]. These changes were associated with significant hypercalciuria [107]. Bicarbonate loading for 7 days increased the expression of calcium-absorbing proteins and improved the hypercalciuria in pendrin KO mice [107]. It is plausible that the acidic urine in pendrin KO mice causes hypercalciuria subsequent to the down-regulation of calciumabsorbing molecules in the distal nephron. This is congruent with published reports indicating the down-regulation of ECaC (TRPV5) by acidic pH [81]. The authors [107] proposed that humans with unexplained acidic urine and hypercalciuria [108, 109] could have decreased pendrin activity. It is worth mentioning that in a rat model of hypercalcemia and hypercalciuria, triggered by PTH infusion, urinary excretion of acid increased subsequent to enhanced activity of H+-ATPase, likely to prevent stone formation [110].

Pendrin and the kidney

Based on published studies, the working model elucidating chloride reabsorption pathways in the collecting duct indicates the following major mechanisms: first, an active trans-cellular reabsorption by pendrin in B-intercalated cells, which is coupled to the basolateral Cl channel ClC-K (Figure 2) [9]. Second, a passive diffusion of chloride down its electrochemical gradients via the para-cellular pathway [9–11]. This is secondary to a lumen-negative, trans-epithelial electrical potential difference (Vte) generated via Na absorption across the apical membrane through the ENaC (Figure 2) and likely involves claudins [9–11]. Third, the recycling of Cl− across the basolateral membrane of acid-secreting A-intercalated cells. This is accomplished via the electroneutral Cl =HCO3 exchanger AE1 (SLC4A1) working in tandem with the basolateral Cl− channel CLC-K (Figure 2) [16, 17, 31]. Taken together, these studies demonstrate that Cl− transport is required for both the acidification and alkalinization of the urine. The simultaneous operation of these pathways allows the fine-tuning of acid–base excretion. In addition, recent published studies indicate the presence of an apical sodiumdependent chloride/bicarbonate exchange that can function in tandem with pendrin [12]. Studies examining the role of NDCBE in salt absorption in the collecting duct in salt deplete/salt replete states or in the setting of NCC inactivation in NDCBE KO mice are wanting. In conclusion, pendrin is a multifaceted transporter that plays important roles in various functions of the kidney, including chloride homeostasis, vascular volume and blood pressure regulation, acid–base balance and calcium excretion. Furthermore, recent studies demonstrate that pendrin plays an essential role in distal tubule salt reabsorption in the setting of NCC inactivation. We propose that pendrin could provide a novel target for a new class of diuretics that, in conjunction with thiazide derivatives, can be an effective regimen for patients with fluid overload, such as those with congestive heart failure.

7

FULL REVIEW

specifically demonstrated in cultured tracheal and parotid epithelial cells [103, 104] such a synergy could be at work in CCD cells, where abundant levels of CFTR and pendrin are expressed. It is interesting to note that the highest expression levels of CFTR in the collecting duct are observed in B-intercalated cells, which are the exclusive site of pendrin expression in the kidney [95]. The schematic diagram in Figure 5A and B depicts a hypothetical interaction between CFTR and pendrin in the CCD to facilitate HCO3 secretion. Chloride is reabsorbed across the apical membrane via pendrin and transported to the blood via the basolateral chloride channel CLC-K.

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

pendrin are both expressed in apical membrane of B-intercalated cells and work in tandem with each other to enhance the secretion of bicarbonate into the lumen of CCD. (B) Cystic fibrosis: the mutant CFTR (specifically the delta 508 mutation) is retained in the cytoplasm which in turn impairs the trafficking of pendrin and HCO3 secretion into the lumen.

FUNDING These studies in the author’s laboratory were supported by Merit Review awards from the Department of Veterans Affairs, NIH grants RO1 DK62809 and R56DK62809 and by grants from US Renal Care (to M.S.).

1. Aronson PS, Giebisch G. Mechanisms of chloride transport in the proximal tubule. Am J Physiol 1997; 273(2 Pt 2): F179–F192 2. Karet FE, Lifton RP. Mutations contributing to human blood pressure variation. Recent Prog Horm Res 1997; 52: 263–276 discussion 276–7 3. Gattineni J, Baum M. Developmental changes in renal tubular transportan overview. Pediatr Nephrol 2013 Nov 20. [Epub ahead of print] 4. Ellison DH. Through a glass darkly: salt transport by the distal tubule. Kidney Int 2011; 79: 5–8 5. Sindić A, Chang MH, Mount DB et al. Renal physiology of SLC26 anion exchangers. Curr Opin Nephrol Hypertens 2007; 16: 484–490 6. Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hypertension. Cell 2001; 104: 545–556 7. Soleimani M. SLC26 Cl-/HCO3- exchangers in the kidney: roles in health and disease. Kidney Int 2013; 84: 657–666 8. Alper SL, Sharma AK. The SLC26 gene family of anion transporters and channels. Mol Aspects Med 2013; 34: 494–515 9. Wall SM, Weinstein AM. Cortical distal nephron Cl(−) transport in volume homeostasis and blood pressure regulation. Am J Physiol Renal Physiol 2013; 305: F427–F438 10. Hou J, Rajagopal M, Yu AS. Claudins and the kidney. Annu Rev Physiol 2013; 75: 479–501 2012 11. Muto S, Furuse M, Kusano E. Claudins and renal salt transport. Clin Exp Nephrol 2012; 16: 61–67 12. Leviel F, Hübner CA, Houillier P et al. The Na+-dependent chloridebicarbonate exchanger SLC4A8 mediates an electroneutral Na+ reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 2010; 120: 1627–1635 13. Parker MD, Boron WF. The divergence, actions, roles, and relatives of sodium-coupled bicarbonate transporters. Physiol Rev 2013; 93: 803–959 14. Soleimani M, Burnham CE. Physiologic and molecular aspects of the Na +:HCO− 3 cotransporter in health and disease processes. Kidney Int 2000; 57: 371–384 15. Kurtz I, Zhu Q. Structure, function, and regulation of the SLC4 NBCe1 transporter and its role in causing proximal renal tubular acidosis. Curr Opin Nephrol Hypertens 2013; 22: 572–583 16. Christov M, Alper SL. Tubular transport: core curriculum 2010. Am J Kidney Dis 2010; 56: 1202–1217 17. Schwartz GJ, Al-Awqati Q. Role of hensin in mediating the adaptation of the cortical collecting duct to metabolic acidosis. Curr Opin Nephrol Hypertens 2005; 14: 383–388 18. Royaux IE, Wall SM, Karniski LP et al. Pendrin, encoded by the Pendred syndrome gene, resides in the apical region of renal intercalated cells and mediates bicarbonate secretion. Proc Natl Acad Sci USA 2001; 98: 4221–4226 19. Amlal H, Petrovic S, Xu J et al. Deletion of the anion exchanger Slc26a4 ( pendrin) decreases apical Cl(−)/HCO3(−) exchanger activity and impairs bicarbonate secretion in kidney collecting duct. Am J Physiol Cell Physiol 2010; 299: C33–C41 20. Soleimani M, Greeley T, Petrovic S et al. Pendrin: an apical Cl-/OH-/ HCO− 3 exchanger in the kidney cortex. Am J Physiol Renal Physiol 2001; 280: F356–F364 21. Bissig M, Hagenbuch B, Stieger B et al. Functional expression cloning of the canalicular sulfate transport system of rat hepatocytes. J Biol Chem 1994; 269: 3017–3021 22. Hastbacka J, de la Chapelle A, Mahtani MM et al. The diastrophic dysplasia gene encodes a novel sulfate transporter: positional cloning by fine-structure linkage disequilibrium mapping. Cell 1994; 78: 1073–1087

8

M. Soleimani

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

FULL REVIEW

REFERENCES

23. Hoglund P, Haila S, Socha J et al. Mutations of the down-regulated in adenoma (DRA) gene cause congenital chloride diarrhea. Nature Genet 1996; 14: 316–319 24. Everett LA, Glaser B, Beck JC et al. Pendred syndrome is caused by mutations in a putative sulphate transporter gene (PDS). Nat Gen 1997; 17: 411–422 25. Zheng J, Shen W, He DZ et al. Prestin is the motor protein of cochlear outer hair cells. Nature 2000; 405: 149–155 26. Lohi H, Kujala M, Kerkela E et al. Mapping of five new putative anion transporter genes in human and characterization of SLC26A6, a candidate gene for pancreatic anion exchanger. Genomics 2000; 70: 102–112 27. Lohi H, Kujala M, Makela S et al. Functional characterization of three novel tissue-specific anion exchangers SLC26A7, -A8, and -A9. J Biol Chem 2002; 277: 14246–14254 28. Vincourt JB, Jullien D, Amalric F et al. Molecular and functional characterization of SLC26A11, a sodium-independent sulfate transporter from high endothelial venules. FASEB J 2003; 17: 890–892 29. Romero MF, Chang MH, Plata C et al. Physiology of electrogenic SLC26 paralogues. Novartis Found Symp 2006; 273: 126–138 30. Soleimani M, Xu J. SLC26 chloride/base exchangers in the kidney in health and disease. Semin Nephrol 2006; 26: 375–385 31. Alper SL. Molecular physiology and genetics of Na+-independent SLC4 anion exchangers. J Exp Biol. 2009; 212 (Pt 11): 1672–1683 32. Melvin JE, Park K, Richardson L et al. Mouse down-regulated in adenoma (DRA) is an intestinal Cl(−)/HCO(3)(−) exchanger and is upregulated in colon of mice lacking the NHE-3 Na(+)/H(+) exchanger. J Biol Chem 1999; 274: 22855–22861 33. Jiang Z, Grichtchenko II, Boron WF et al. Specificity of anion exchange mediated by mouse Slc26a6. J Biol Chem 2002; 277: 33963–33967 34. Xie Q, Welch R, Mercado A et al. Molecular characterization of the murine Slc26a6 anion exchanger: functional comparison with Slc26a1. Am J Physiol Renal Physiol 2002; 283: F826–F838 35. Chernova MN, Jiang L, Shmukler BE et al. Acute regulation of the SLC26A3 congenital chloride diarrhoea anion exchanger (DRA) expressed in Xenopus oocytes. J Physiol 2003; 549 (Pt 1): 3–19 36. Chernova MN, Jiang L, Friedman DJ et al. Functional comparison of mouse slc26a6 anion exchanger with human SLC26A6 polypeptide variants: differences in anion selectivity, regulation, and electrogenicity. J Biol Chem 2005; 280: 8564–8580 37. Wang Z, Petrovic S, Mann E et al. Identification of an apical Cl-/HCO3exchanger in the small intestine. Am J Physiol Gastrointest Liver Physiol 2002; 282: G573–G579 38. Schweinfest CW, Spyropoulos DD, Henderson KW et al. Slc26a3 (dra)deficient mice display chloride-losing diarrhea, enhanced colonic proliferation, and distinct up-regulation of ion transporters in the colon. J Biol Chem 2006; 281: 37962–37971 39. Petrovic S, Ju X, Barone S et al. Identification of a basolateral Cl-/HCO3exchanger specific to gastric parietal cells. Am J Physiol Gastrointest Liver Physiol 2003; 284: G1093–G1103 40. Xu J, Henriksnäs J, Barone S et al. SLC26A9 is expressed in gastric surface epithelial cells, mediates Cl-/HCO3- exchange, and is inhibited by NH4+. Am J Physiol Cell Physiol 2005; 289: C493–C505 41. Xu J, Song P, Miller ML et al. Deletion of the chloride transporter Slc26a9 causes loss of tubulovesicles in parietal cells and impairs acid secretion in the stomach. Proc Natl Acad Sci USA 2008; 105: 17955–17960 42. Xu J, Barone S, Li H et al. Slc26a11, a chloride transporter, localizes with the vacuolar H(+)-ATPase of A-intercalated cells of the kidney. Kidney Int 2011; 80: 926–937 43. Chen AP, Chang MH, Romero MF. Functional analysis of nonsynonymous single nucleotide polymorphisms in human SLC26A9. Hum Mutat 2012; 33: 1275–1284 44. Bertrand CA, Zhang R, Pilewski JM et al. SLC26A9 is a constitutively active, CFTR-regulated anion conductance in human bronchial epithelia. J Gen Physiol 2009; 133: 421–438 45. Kim KH, Shcheynikov N, Wang Y et al. SLC26A7 is a Cl− channel regulated by intracellular pH. J Biol Chem 2005; 280: 6463–6470 46. Dorwart MR, Shcheynikov N, Wang Y et al. SLC26A9 is a Cl− channel regulated by the WNK kinases. J Physiol 2007; 584: 333–345

9

FULL REVIEW

Pendrin and the kidney

72. Mohebbi N, Perna A, van der Wijst J et al. Regulation of two renal chloride transporters, AE1 and pendrin, by electrolytes and aldosterone. PLoS ONE 2013; 8: e55286 73. Purkerson JM, Heintz EV, Nakamori A et al. Insights into acidosisinduced regulation of SLC26A4 ( pendrin) and SLC4A9 (AE4) transporters using 3-dimensional morphometric analysis of β-intercalated cells. Am J Physiol Renal Physiol 2014; 307: F601–F611 74. Quentin F, Chambrey R, Trinh-Trang-Tan MM et al. The Cl−/HCO− 3 exchanger pendrin in the rat kidney is regulated in response to chronic alterations in chloride balance. Am J Physiol Renal Physiol 2004; 287: F1179–F1188 75. Vallet M, Picard N, Loffing-Cueni D et al. Pendrin regulation in mouse kidney primarily is chloride-dependent. J Am Soc Nephrol 2006; 17: 2153–2163 76. Amlal H, Wang Z, Soleimani M. Potassium depletion downregulates chloride-absorbing transporters in rat kidney. J Clin Invest 1998; 101: 1045–1054 77. Khanna A, Kurtzman NA Metabolic alkalosis. J Nephrol 2006; 19: (Suppl 9): S86–S96 78. van der Lubbe N, Moes AD, Rosenbaek LL et al. K+-induced natriuresis is preserved during Na+ depletion and accompanied by inhibition of the Na +-Cl- cotransporter. Am J Physiol Renal Physiol 2013; 305: F1177–F1188 79. Vitzthum H, Seniuk A, Schulte LH et al. Functional coupling of renal K+ and Na+ handling causes high blood pressure in Na+ replete mice. J Physiol 2014; 592 (Pt 5): 1139–1157 80. Eladari D, Hübner CA. Novel mechanisms for NaCl reabsorption in the collecting duct. Curr Opin Nephrol Hypertens 2011; 20: 506–511 81. Loffing J, Vallon V, Loffing-Cueni D et al. Altered renal distal tubule structure and renal Na+ and Ca2+ handling in a mouse model for Gitelman’s syndrome. J Am Soc Nephrol 2004; 15: 2276–2288 82. Xu J, Barone B, Brooks M et al. Double knockout of carbonic anhydrase II (CAII) and Na+-Cl− cotransporter (NCC) causes salt wasting and volume depletion. Cell Physiol and Biochem 2013; 32: 173–183 83. Soleimani M, Barone S, Xu J et al. Double knockout of pendrin and NaCl cotransporter (NCC) causes severe salt wasting, volume depletion, and renal failure. Proc Natl Acad Sci USA 2012; 109: 13368–13373 84. Rubera I, Loffing J, Palmer LG et al. Collecting duct-specific gene inactivation of alphaENaC in the mouse kidney does not impair sodium and potassium balance. J Clin Invest 2003; 112: 554–565 85. Amlal H, Soleimani M. Pendrin as a novel target for diuretic therapy. Cell Physiol Biochem 2011; 28: 521–526 86. Wangemann P. The role of pendrin in the development of the murine inner ear. Cell Physiol Biochem 2011; 28: 527–534 87. Kim YH, Pham TD, Zheng W et al. Role of pendrin in iodide balance: going with the flow. Am J Physiol Renal Physiol 2009; 297: F1069–F1079 88. Bagnis C, Marshansky V, Breton S et al. Remodeling the cellular profile of collecting ducts by chronic carbonic anhydrase inhibition. Am J Physiol Renal Physiol 2001; 280: F437–F448 89. Zahedi K, Barone S, Xu J et al. Potentiation of the effect of thiazide derivatives by carbonic anhydrase inhibitors: molecular mechanisms and potential clinical implications. PLoS ONE 2013; 8: e79327 90. Pela I, Bigozzi M, Bianchi B. Profound hypokalemia and hypochloremic metabolic alkalosis during thiazide therapy in a child with Pendred syndrome. Clin Nephrol 2008; 69: 450–453 91. Reimold FR, Heneghan JF, Stewart AK et al. Pendrin function and regulation in Xenopus oocytes. Cell Physiol Biochem 2011; 28: 435–450 92. Thumova M, Pech V, Froehlich O et al. Pendrin protein abundance in the kidney is regulated by nitric oxide and cAMP. Am J Physiol Renal Physiol 2012; 303: F812–F820 93. Azroyan A, Morla L, Crambert G et al. Regulation of pendrin by cAMP: possible involvement in β-adrenergic-dependent NaCl retention. Am J Physiol Renal Physiol 2012; 302: F1180–F1187 94. Ko SB, Zeng W, Dorwart MR et al. Gating of CFTR by the STAS domain of SLC26 transporters. Nat Cell Biol 2004; 6: 343–350 95. Todd-Turla KM, Rusvai E, Náray-Fejes-Tóth A et al. CFTR expression in cortical collecting duct cells. Am J Physiol 1996; 270 (1 Pt 2): F237–F244 96. Jouret F, Bernard A, Hermans C et al. Cystic fibrosis is associated with a defect in apical receptor-mediated endocytosis in mouse and human kidney. J Am Soc Nephrol 2007; 18: 707–718

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

47. Rahmati N, Kunzelmann K, Xu J et al. Slc26a11 is prominently expressed in the brain and functions as a chloride channel: expression in Purkinje cells and stimulation of V H(+)-ATPase. Pflugers Arch 2013; 465: 1583–1597 48. Karniski LP, Wang T, Everett LA et al. Formate-stimulated NaCl absorption in the proximal tubule is independent of the pendrin protein. Am J Physiol Renal Physiol 2002; 283: F952–F956 49. Kim YH, Kwon TH, Frische S et al. Immunocytochemical localization of pendrin in intercalated cell subtypes in rat and mouse kidney. Am J Physiol Renal Physiol 2002; 283: F744–F754 50. Wall SM, Hassell KA, Royaux IE et al. Localization of pendrin in mouse kidney. Am J Physiol Renal Physiol 2003; 284: F229–F241 51. Bonar PT, Casey JR. Plasma membrane Cl–/HCO₃– exchangers: structure, mechanism and physiology. Channels (Austin) 2008; 2: 337–345 52. Knauf F, Yang CL, Thomson RB et al. Identification of a chlorideformate exchanger expressed on the brush border membrane of renal proximal tubule cells. Proc Natl Acad Sci USA 2001; 98: 9425–9430 53. Petrovic S, Ma L, Wang Z et al. Identification of an apical Cl-/HCO3exchanger in rat kidney proximal tubule. Am J Physiol Cell Physiol 2003; 285: C608–C617 54. Wang Z, Wang T, Petrovic S et al. Renal and intestinal transport defects in Slc26a6-null mice. Am J Physiol Cell Physiol 2005; 288: C957–C965 55. Petrovic S, Barone S, Xu J et al. SLC26A7: a basolateral Cl-/HCO3- exchanger specific to intercalated cells of the outer medullary collecting duct. Am J Physiol Renal Physiol 2004; 286: F161–F169 56. Xu J, Worrell RT, Li HC et al. Chloride/bicarbonate exchanger SLC26A7 is localized in endosomes in medullary collecting duct cells and is targeted to the basolateral membrane in hypertonicity and potassium depletion. J Am Soc Nephrol 2006; 17: 956–967 57. Xu J, Song P, Nakamura S et al. Deletion of the chloride transporter Slc26a7 causes distal renal tubular acidosis and impairs gastric acid secretion. J Biol Chem 2009; 284: 29470–29479 58. Amlal H, Xu J, Barone S et al. The chloride channel/transporter Slc26a9 regulates the systemic arterial pressure and renal chloride excretion. J Mol Med (Berl) 2013; 91: 561–572 59. Pendred V. Deaf-mutism and goitre. Lancet 2005; 2: 532–356 60. Bizhanova A, Kopp P. Genetics and phenomics of Pendred syndrome. Mol Cell Endocrinol 2010; 322: 83–90 61. Bogazzi F, Russo D, Raggi F et al. Mutations in the SLC26A4 ( pendrin) gene in patients with sensorineural deafness and enlarged vestibular aqueduct. J Endocrinol Invest 2004; 27: 430–435 62. Hulander M, Kiernan AE, Blomqvist SR et al. Lack of pendrin expression leads to deafness and expansion of the endolymphatic compartment in inner ears of Foxi1 null mutant mice. Development 2003; 130: 2013–2025 63. Wall SM, Kim YH, Stanley L et al. NaCl restriction upregulates renal Slc26a4 through subcellular redistribution: role in Cl- conservation. Hypertension 2004; 44: 982–987 64. Kandasamy N, Fugazzola L, Evans M et al. Life-threatening metabolic alkalosis in Pendred syndrome. Eur J Endocrinol 2011; 165: 167–170 65. Verlander JW, Hassell KA, Royaux IE et al. Deoxycorticosterone upregulates PDS (Slc26a4) in mouse kidney: role of pendrin in mineralocorticoid-induced hypertension. Hypertension 2003; 179: 356–362 66. Jacques T, Picard N, Miller RL et al. Overexpression of pendrin in intercalated cells produces chloride-sensitive hypertension. J Am Soc Nephrol 2013; 24: 1104–1113 67. Wagner CA, Finberg KE, Stehberger PA et al. Regulation of the expression of the Cl-/anion exchanger pendrin in mouse kidney by acid-base status. Kidney Int 2002; 62: 2109–2117 68. Petrovic S, Wang Z, Ma L et al. Regulation of the apical Cl-/HCO3- exchanger pendrin in rat cortical collecting duct in metabolic acidosis. Am J Physiol Renal Physiol 2003; 284: F103–F112 69. Carraro-Lacroix LR, Malnic G. Acid base transport by the renal distal nephron. J Nephrol 2010; 23(Suppl 16): S19–S27 70. Welsh-Bacic D, Nowik M, Kaissling B et al. Proliferation of acid-secretory cells in the kidney during adaptive remodelling of the collecting duct. PLoS ONE 2011; 6: e25240 71. Alesutan I, Daryadel A, Mohebbi N et al. Impact of bicarbonate, ammonium chloride, and acetazolamide on hepatic and renal SLC26A4 expression. Cell Physiol Biochem 2011; 28: 553–558

104. Garnett JP, Hickman E, Burrows R et al. Novel role for pendrin in orchestrating bicarbonate secretion in cystic fibrosis transmembrane conductance regulator (CFTR)-expressing airway serous cells. J Biol Chem 2011; 286: 41069–41082 105. Hoenderop JG, Hartog A, Stuiver M et al. Localization of the epithelial Ca(2+) channel in rabbit kidney and intestine. J Am Soc Nephrol 2000; 11: 1171–1178 106. Hoenderop JG, Nilius B, Bindels RJ. Molecular mechanism of active Ca2+ reabsorption in the distal nephron. Annu Rev Physiol 2002; 64: 529–549 107. Barone S, Amlal H, Xu J et al. Deletion of the Cl-/HCO3- exchanger pendrin downregulates calcium-absorbing proteins in the kidney and causes calcium wasting. Nephrol Dial Transplant 2012; 27: 1368–1379 108. Bonny O, Rubin A, Huang CL et al. Mechanism of urinary calcium regulation by urinary magnesium and pH. J Am Soc Nephrol 2008; 19: 1530–1537 109. Sakhaee K. Recent advances in the pathophysiology of nephrolithiasis. Kidney Int 2009; 75: 585–595 110. Wang W, Praetorius J, Li C et al. Vacuolar H+-ATPase expression is increased in acid-secreting intercalated cells in kidneys of rats with hypercalcaemia-induced alkalosis. Acta Physiol (Oxf ) 2007; 189: 359–368

FULL REVIEW

Received for publication: 25.6.2014; Accepted in revised form: 25.8.2014

10

M. Soleimani

Downloaded from http://ndt.oxfordjournals.org/ at University of Oklahoma on October 26, 2014

97. Lu M, Dong K, Egan ME et al. Mouse cystic fibrosis transmembrane conductance regulator forms cAMP-PKA-regulated apical chloride channels in cortical collecting duct. Proc Natl Acad Sci USA 2010; 107: 6082–6087 98. Raksaseri P, Chatsudthipong V, Muanprasat C et al. Activation of liver X receptors reduces CFTR-mediated Cl(−) transport in kidney collecting duct cells. Am J Physiol Renal Physiol 2013; 305: F583–F591 99. Chang CT, Bens M, Hummler E et al. Vasopressin-stimulated CFTR Clcurrents are increased in the renal collecting duct cells of a mouse model of Liddle’s syndrome. J Physiol 2005; 562 (Pt 1): 271–284 100. Bens M, Duong Van Huyen JP, Cluzeaud F et al. CFTR disruption impairs cAMP-dependent Cl(−) secretion in primary cultures of mouse cortical collecting ducts. Am J Physiol Renal Physiol 2001; 281: F434–F442 101. Bates CM, Baum M, Quigley R. Cystic fibrosis presenting with hypokalemia and metabolic alkalosis in a previously healthy adolescent. J Am Soc Nephrol 1997; 8: 352–355 102. Baird JS, Walker P, Urban A et al. Metabolic alkalosis and cystic fibrosis. Chest 2002; 122: 755–756 103. Shcheynikov N, Yang D, Wang Y et al. The Slc26a4 transporter functions as an electroneutral Cl-/I-/HCO3- exchanger: role of Slc26a4 and Slc26a6 in I- and HCO3- secretion and in regulation of CFTR in the parotid duct. J Physiol 2008; 586: 3813–3824

The multiple roles of pendrin in the kidney.

The [Formula: see text] exchanger pendrin (SLC26A4, PDS) is located on the apical membrane of B-intercalated cells in the kidney cortical collecting d...
750KB Sizes 4 Downloads 6 Views