8
Perspectives on edema in childhood nephrotic syndrome Chia Wei Teoh, Lisa A. Robinson, and Damien Noone Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada Submitted 29 May 2015; accepted in final form 11 August 2015 Teoh CW, Robinson LA, Noone D. Perspectives on edema in childhood nephrotic syndrome. Am J Physiol Renal Physiol 309: F575–F582, 2015. First published August 19, 2015; doi:10.1152/ajprenal.00229.2015.—There have been two major theories surrounding the development of edema in nephrotic syndrome (NS), namely, the under- and overfill hypoth- eses. Edema is one of the cardinal features of NS and remains one of the principal reasons for admission of children to the hospital. Re- cently, the discovery that proteases in the glomerular filtrate of patients with NS are activating the epithelial sodium channel (ENaC), resulting in intrarenal salt retention and thereby contributing to edema, might suggest that targeting ENaC with amiloride might be a suitable strategy to manage the edema of NS. Other potential agents, partic- ularly urearetics and aquaretics, might also prove useful in NS. Recent evidence also suggests that there may be other areas involved in salt storage, especially the skin, and it will be intriguing to study the implications of this in NS. aquaretic; edema; ENaC; nephrotic syndrome; urearetic NEPHROTIC SYNDROME (NS) is one of the most common child- hood kidney diseases with a cumulative prevalence of 16 in 100,000 children (26). The diagnosis of NS is clinical, char- acterized by heavy proteinuria, hypoalbuminemia, and edema. NS can be classified into two main groups: the genetically determined forms, involving podocyte and slit diaphragm pro- teins, which usually manifest in the first year of life (39), and the idiopathic form. The main causes of idiopathic childhood NS are minimal-change disease (MCD) (77.1%) and focal segmental glomerulosclerosis (FSGS) (7.9%) (16). The pathophysiological mechanisms of the syndrome re- main poorly defined despite decades of study. It is a multifac- torial disease, where the immune system targets structural components of the glomerular filtration barrier in a genetically susceptible patient. Many causative factors, including circulat- ing factors, genetic polymorphisms implicated in lymphocyte maturation and differentiation, and DNA epigenetic modifica- tions, have been proposed and tested; none fully explains the pathogenesis of the disease (43). The familial type of NS is primarily considered a “podocy- topathy” as it involves inherited abnormalities in slit dia- phragm or podocyte proteins, and the translational opportuni- ties and challenges associated with this form of NS have recently been reviewed in this journal (37). However, the term podocytopathy fails to take into account the fact that mutations in proteins that make up the glomerular basement membrane, such as the collagen chains, 3, 4, and 5 (62) or the laminins, may also lead to a proteinuric state, with laminin 2 (LAMB2) being rarely associated with infantile or the congen- ital form of NS (17). The search also continues for the elusive T lymphocyte-derived soluble factor proposed by Shalhoub in 1974 (78). The reader is also directed to recent comprehensive reviews on the subject of permeability factors in NS, detailing the soluble urokinase plasminogen activator receptor story and proposing putative factors and translational strategies (18, 59). More recently, it has been suggested that even in MCD, both a permeability factor of immune origin and podocyte-derived factors may contribute to the development of NS (reviewed in Ref. 43). Edema is one of the classic clinical features of childhood NS and one of the principal reasons for admission to the hospital. The underlying mechanisms of edema formation remain a subject of ongoing investigation. This review aims to focus on the current understanding of edema formation in NS and to discuss recent developments in this area, potential targets for therapy, and novel areas that need further scientific explora- tion. Pathogenesis of Edema in NS: Under- vs. Overfill Hypothesis Two major opposing theories on the pathophysiological mechanisms underlying the development of edema in NS have been proposed: 1) the underfill hypothesis and 2) the overfill hypothesis (7, 13). The underfill hypothesis. The underfill hypothesis was gen- erated almost a century ago (27). It postulates that high-grade proteinuria results in hypoalbuminemia, leading to a reduction in plasma oncotic pressure with consequent leakage of plasma water into the interstitium, generating edema. The resultant diminished intravascular volume manifests with tachycardia, hypotension, and oliguria as well as peripheral vasoconstric- tion and water and sodium retention. This is affected by activation of the renin-angiotensin-aldosterone system (RAAS), coupled with an increase in plasma norepinephrine and argi- nine vasopressin (AVP) concentrations (65, 76, 95). Together, this neurohormonal activation results in a highly concentrated urine with very low sodium content (44). In a study of 16 pediatric and adult patients with NS with normal renal func- tion, Usberti et al. (92) found decreased plasma sodium con- centration, increased plasma AVP concentration, and an ele- vation in plasma renin activity (PRA) and urinary norepineph- rine levels, coupled with impaired excretion of an acute water load, compared with controls. They demonstrated a highly significant inverse correlation between plasma AVP concen- tration and blood volume in these nephrotic patients (92). Therefore, in this scenario, the renal sodium retention was thought to occur as a secondary physiological response to underfilling (a reduced effective intravascular volume) (8). The therapeutic implications of this hypothesis are clear: expansion of intravascular volume and restoration of plasma oncotic pressure by administration of a colloid such as albumin would prove beneficial. Indeed, albumin infusion-induced volume expansion in children with NS has been shown to reduce PRA, Address for reprint requests and other correspondence: D. Noone, Div. of Nephrology, The Hospital for Sick Children, 555 Univ. Ave., Toronto, ON M5G 1X8, Canada (e-mail: [email protected]). Am J Physiol Renal Physiol 309: F575–F582, 2015. First published August 19, 2015; doi:10.1152/ajprenal.00229.2015. Perspective 1931-857X/15 Copyright © 2015 the American Physiological Society http://www.ajprenal.org F575

Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

  • Upload
    others

  • View
    11

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

Perspectives on edema in childhood nephrotic syndrome

Chia Wei Teoh, Lisa A. Robinson, and Damien NooneDivision of Nephrology, The Hospital for Sick Children, Toronto, Ontario, Canada

Submitted 29 May 2015; accepted in final form 11 August 2015

Teoh CW, Robinson LA, Noone D. Perspectives on edema in childhoodnephrotic syndrome. Am J Physiol Renal Physiol 309: F575–F582, 2015.First published August 19, 2015; doi:10.1152/ajprenal.00229.2015.—Therehave been two major theories surrounding the development of edemain nephrotic syndrome (NS), namely, the under- and overfill hypoth-eses. Edema is one of the cardinal features of NS and remains one ofthe principal reasons for admission of children to the hospital. Re-cently, the discovery that proteases in the glomerular filtrate ofpatients with NS are activating the epithelial sodium channel (ENaC),resulting in intrarenal salt retention and thereby contributing to edema,might suggest that targeting ENaC with amiloride might be a suitablestrategy to manage the edema of NS. Other potential agents, partic-ularly urearetics and aquaretics, might also prove useful in NS. Recentevidence also suggests that there may be other areas involved in saltstorage, especially the skin, and it will be intriguing to study theimplications of this in NS.

aquaretic; edema; ENaC; nephrotic syndrome; urearetic

NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of �16 in100,000 children (26). The diagnosis of NS is clinical, char-acterized by heavy proteinuria, hypoalbuminemia, and edema.NS can be classified into two main groups: the geneticallydetermined forms, involving podocyte and slit diaphragm pro-teins, which usually manifest in the first year of life (39), andthe idiopathic form. The main causes of idiopathic childhoodNS are minimal-change disease (MCD) (77.1%) and focalsegmental glomerulosclerosis (FSGS) (7.9%) (16).

The pathophysiological mechanisms of the syndrome re-main poorly defined despite decades of study. It is a multifac-torial disease, where the immune system targets structuralcomponents of the glomerular filtration barrier in a geneticallysusceptible patient. Many causative factors, including circulat-ing factors, genetic polymorphisms implicated in lymphocytematuration and differentiation, and DNA epigenetic modifica-tions, have been proposed and tested; none fully explains thepathogenesis of the disease (43).

The familial type of NS is primarily considered a “podocy-topathy” as it involves inherited abnormalities in slit dia-phragm or podocyte proteins, and the translational opportuni-ties and challenges associated with this form of NS haverecently been reviewed in this journal (37). However, the termpodocytopathy fails to take into account the fact that mutationsin proteins that make up the glomerular basement membrane,such as the collagen chains, �3, �4, and �5 (62) or thelaminins, may also lead to a proteinuric state, with laminin �2(LAMB2) being rarely associated with infantile or the congen-ital form of NS (17). The search also continues for the elusiveT lymphocyte-derived soluble factor proposed by Shalhoub in

1974 (78). The reader is also directed to recent comprehensivereviews on the subject of permeability factors in NS, detailingthe soluble urokinase plasminogen activator receptor story andproposing putative factors and translational strategies (18, 59).More recently, it has been suggested that even in MCD, both apermeability factor of immune origin and podocyte-derivedfactors may contribute to the development of NS (reviewed inRef. 43).

Edema is one of the classic clinical features of childhood NSand one of the principal reasons for admission to the hospital.The underlying mechanisms of edema formation remain asubject of ongoing investigation. This review aims to focus onthe current understanding of edema formation in NS and todiscuss recent developments in this area, potential targets fortherapy, and novel areas that need further scientific explora-tion.

Pathogenesis of Edema in NS: Under- vs. OverfillHypothesis

Two major opposing theories on the pathophysiologicalmechanisms underlying the development of edema in NS havebeen proposed: 1) the underfill hypothesis and 2) the overfillhypothesis (7, 13).

The underfill hypothesis. The underfill hypothesis was gen-erated almost a century ago (27). It postulates that high-gradeproteinuria results in hypoalbuminemia, leading to a reductionin plasma oncotic pressure with consequent leakage of plasmawater into the interstitium, generating edema. The resultantdiminished intravascular volume manifests with tachycardia,hypotension, and oliguria as well as peripheral vasoconstric-tion and water and sodium retention. This is affected byactivation of the renin-angiotensin-aldosterone system (RAAS),coupled with an increase in plasma norepinephrine and argi-nine vasopressin (AVP) concentrations (65, 76, 95). Together,this neurohormonal activation results in a highly concentratedurine with very low sodium content (44). In a study of 16pediatric and adult patients with NS with normal renal func-tion, Usberti et al. (92) found decreased plasma sodium con-centration, increased plasma AVP concentration, and an ele-vation in plasma renin activity (PRA) and urinary norepineph-rine levels, coupled with impaired excretion of an acute waterload, compared with controls. They demonstrated a highlysignificant inverse correlation between plasma AVP concen-tration and blood volume in these nephrotic patients (92).Therefore, in this scenario, the renal sodium retention wasthought to occur as a secondary physiological response tounderfilling (a reduced effective intravascular volume) (8). Thetherapeutic implications of this hypothesis are clear: expansionof intravascular volume and restoration of plasma oncoticpressure by administration of a colloid such as albumin wouldprove beneficial. Indeed, albumin infusion-induced volumeexpansion in children with NS has been shown to reduce PRA,

Address for reprint requests and other correspondence: D. Noone, Div. ofNephrology, The Hospital for Sick Children, 555 Univ. Ave., Toronto, ONM5G 1X8, Canada (e-mail: [email protected]).

Am J Physiol Renal Physiol 309: F575–F582, 2015.First published August 19, 2015; doi:10.1152/ajprenal.00229.2015. Perspective

1931-857X/15 Copyright © 2015 the American Physiological Societyhttp://www.ajprenal.org F575

Page 2: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

aldosterone, and AVP and to improve glomerular filtration rate(GFR), urine output, and sodium excretion (70, 90).

However, a decrease in measured plasma volume in ne-phrotic patients has not been a universal finding, and in somenephrotic states plasma volume may actually be increased (24,34, 66). Furthermore, elevated PRA or aldosterone, expected inthe setting of decreased blood volume, has only been con-firmed in about half of studied nephrotic subjects (35, 66).However, these variations in findings may be accounted for bythe fact that these parameters are dependent on the hemody-namic status of the patients at the time of the study.

Some clinical situations also do not fit well with the underfillhypothesis. The treatment of edema in NS with albumin alonereduces PRA, but it is often insufficient to induce diuresiswithout the addition of a diuretic (11, 94). In contrast, somepatients respond to diuretics alone without the need for albu-min (44). Moreover, the reduction of the renin-aldosterone axisby mineralocorticoid receptor antagonists or angiotensin-con-verting enzyme inhibitors does not result in increased sodiumexcretion in most patients with NS (10, 94). In fact, neurohor-monal markers such as renin and aldosterone levels do notshow a consistent pattern, suggesting volume depletion insome, and normal or excess volume in other patients (9, 11, 52,61, 93, 94, 96). In some patients, the administration of albuminis associated with volume overload and causes hypertensionand pulmonary edema (71). To the observant physician (andparent), the earliest sign of the patient entering remission is alarge diuresis, subsequently confirmed by the improvement inproteinuria. This occurs well before plasma protein levels (andthus plasma oncotic pressure) have normalized (64). Finally,there are patients with other causes of analbuminemia who donot typically suffer from edema (12, 54). Taken together, thesefindings suggest that the generation of edema in NS is not justsecondary to a reduction in plasma oncotic pressure as a resultof hypoalbuminemia caused by high-grade proteinuria.

Ichikawa et al. (42) provided strong experimental evidenceagainst the underfill hypothesis in a rat model of nephrosis, inwhich they performed nephron puncture after infusing puro-mycin aminoglycoside (PAN) into one kidney to independentlyassess the effect of proteinuria in both the nephrotic and thenormal kidney. They noted that only the nephrotic kidneyavidly conserved sodium, despite maintaining euvolemia bycontrolling plasma protein levels within the normal range byinfusion of homologous rat plasma. This suggested an intrare-nal mechanism of salt retention as opposed to a circulatingneurohumoral factor. In a separate study, Chandra et al. (15)found that the control kidney actually increased sodium excre-tion, as if sensing intravascular volume excess. They showedthat sodium delivery to the collecting duct was comparable inboth kidneys, whereas the final sodium excretion was higher inthe normal kidney, therefore suggesting that the collecting ductwas the main site of avid sodium reabsorption in the nephrotickidney.

The overfill hypothesis. Due to the inconsistent findingsdescribed above, the overfill hypothesis was proposed as analternative explanation for the development of edema in NS:proteinuria causes primary sodium retention with consequentvolume expansion and leakage of excess fluid into the inter-stitium (25, 61). In support of this idea, Dorhout Mees et al.(25) observed that blood pressure and plasma volume fell inpatients with NS after prednisone-induced remission (25). In

88 patients with NS, Gur et al. (36, 77) reported higher plasmaand blood volume corrected for estimated lean body masscompared with controls.

The Epithelial Sodium Channel and Sodium Retention in NS

What mediates the primary sodium retention? Ichikawa et al.(42) identified the collecting duct as the main segment involvedin sodium retention in NS (42). Initial focus was on thebasolateral Na-K-ATPase, as there is evidence from studies inrodent models of nephrosis to suggest an increase in levels andactivity of Na-K-ATPase in the collecting ducts of PAN-induced nephrotic rats (20, 97). Recent evidence suggests theprincipal means of sodium reabsorption in NS occurs in thedistal segment of the nephron via the epithelial sodium channel(ENaC) (Fig. 1) (50). Using both the PAN and HgCl2 rodentmodels of NS, Kim et al. (46, 47) demonstrated an increase inthe expression and targeting of ENaC to the apical cell surface,which is aldosterone dependent (19, 57). Lourdel et al. (57)showed a linear correlation between plasma aldosterone andENaC abundance. Although the prevention of hyperaldosteron-emia by adrenalectomy prevented the increase in apical ENaCtargeting, it did not prevent sodium retention and the develop-ment of NS in the PAN-treated rats (19, 57). Although theadrenalectomized rats did not show an increase in ENaCexpression, they had a similarly low urinary sodium excretionas adrenal-intact nephrotic rats with increased ENaC expres-sion (57). Treatment with amiloride returned sodium excretionto normal despite a lack of increase in ENaC expression (57).This aldosterone-independent sodium retention in NS by theENaC was further illustrated by Deschenes et al. (22), whoshowed that sodium retention can be prevented in the PAN-induced rat by amiloride and not aldosterone inhibition.

ENaC activity depends on channel density at the apicalmembrane, which is aldosterone and vasopressin dependent,and on channel activity, which is regulated by proteolyticprocessing and by anionic phospholipids on the inner cellmembrane (Fig. 1) (38, 58). The ENaC is composed of the �-,�-, and �-subunits (74), of which the �- and �-chains have aregulatory role. An ENaC channel with uncleaved �- and�-chains has low open channel probability and conducts littlesodium (40). Sequential proteolytic cleavage of the �- and�-subunits results in increasing open channel probability (40).Cleavage of an inhibitory tract from the �-subunit by serineproteases such as plasmin results in near full activation of theENaC (14). More recently, plasmin has been directly shown toactivate ENaC and promote sodium and water retention innephrotic rodents and humans (Fig. 2) (83). The plasminprecursor, plasminogen, is filtered by the nephrotic kidney andis activated to plasmin by the tubular urokinase-type plasmin-ogen activator present in the rat and human kidney. Use of thepotassium-sparing diuretic, amiloride, resulted in increasedurine sodium excretion and reduced ascites volume in the PANnephrosis rat. These effects were attributed both to inhibitionof ENaC and inhibition of urokinase-type plasminogen activa-tor by amiloride, thus reducing the amount of active plasminpresent in the urine (Fig. 2).

The dominant role of the ENaC in sodium retention and thedevelopment of nephrotic edema are underscored by the rever-sal of sodium retention by amiloride in animal models. Itprovides a rationale for specific blockade of ENaC with

Perspective

F576

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org

Page 3: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

amiloride for the treatment of sodium retention and edemaformation in NS. While the administration of amiloride beforethe onset of sodium retention fully prevented sodium retentionand edema formation in PAN-induced nephrotic rats (22),clinicians usually encounter patients who are well beyond thisearly stage, many of whom have anasarca. In this situation,treatment aims both to limit further sodium retention and topromote diuresis and natriuresis. Under normal conditions,sodium reabsorption along the collecting duct is quantitativelylow, thus reducing the ability of amiloride to promote massivesodium excretion. Loop diuretics decrease sodium reabsorptionalong the thick ascending limb of the loop of Henle, increasingdistal sodium delivery, which, in turn, overloads the sodiumreabsorptive capacity of the distal nephron, resulting in natri-uresis. However, nephrotic patients display increased sodiumreabsorptive capacity along the cortical collecting ducts,thereby blunting the natriuretic effect. Combined use of loopdiuretics and amiloride, which inhibits distal sodium reabsorp-tion, could overcome the resistance to therapy with a loopdiuretic alone. Accordingly, Deschenes et al. (21) showed thatcombined therapy with amiloride and furosemide resulted ingreater natriuresis and weight loss than monotherapy witheither agent alone in pediatric patients (21). There has been nostudy interrogating the effectiveness of amiloride monotherapyin childhood NS. It has to be emphasized that most of thefindings in support of the overfill hypothesis are derived fromanimal studies with few studies in humans.

In NS, ENaC activation occurs following proteolytic pro-cessing of the protein. Therefore, inhibition of serine proteaseactivity could represent another potential therapeutic target(82). Accordingly, camostat mesilate (CM), a synthetic inhib-

itor of serine proteases such as plasmin and prostasin, wasshown to reduce aldosterone-induced renal ENaC proteolysisin rats (91). In a small case series, treatment with CM causeda significant reduction in proteinuria in patients with NS ofvarious etiologies. Although these reports suggest that sys-temic administration of serine protease inhibitors to humanpatients is well tolerated, improves proteinuria, and attenuatesedema formation through inhibition of ENaC-mediated sodiumretention, the potential for systemic effects related to un-specific protease inhibition suggests a note of caution (82).Using inhibitors that are freely filtered across the glomerularbarrier or secreted into the tubules could potentially reducethe side effects associated with systemic inhibition of pro-tease activity (82).

The Role of Corin

Corin is an endogenous type II transmembrane serine pro-tease that cleaves pro atrial natriuretic peptide (ANP) and probrain natriuretic peptide (BNP) to their biologically activeforms and is detected in multiple organs, including the heart,brain, and kidney as well as in the serum and urine (23, 31, 68,102, 103). Its importance in blood pressure was demonstratedby the demonstration of salt-sensitive hypertension in corin-deficient mice (102). ANP has three important roles in sodiumand water excretion in the kidney, namely, 1) inducing afferentarteriolar vasodilation and efferent vasoconstriction, therebyincreasing GFR; 2) causing natriuresis through inhibition of theN�/H� exchanger in the proximal tubule, the distal Na�-Cl�

cotransporter, and ENaC; and 3) causing diuresis throughinhibition of aquaporin-2 insertion in the apical membrane of

Fig. 1. Model of epithelial sodium channel(ENaC) regulation by aldosterone and vaso-pressin. Upon binding of aldosterone to itsreceptor (MR), the ligand-receptor translo-cates into the nucleus, leading to the induc-tion of a specific set of aldosterone-inducedtranscripts (AIT) such as ENaC, the Na/K-ATPase, or SGK1. Vasopressin binds to itsreceptor (V2R), leading to the activation of adownstream protein kinase A (PKA). BothSGK1 and PKA inactivate Nedd4 uponphosphorylation, leading to retention of ac-tive ENaC at the cell surface. Adapted fromRef. 34 with permission.

Perspective

F577

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org

Page 4: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

the collecting duct, normally mediated via arginine vasopressin(reviewed in Ref. 2). Polzin et al. (69) also suggested the rolefor corin in the salt retention in NS. They demonstrated areduction in kidney corin expression in the PAN nephrosis ratcoupled with a concomitant increase in pro-ANP and decreasedANP levels. They then proceeded to demonstrate increasedamounts of renal �-ENaC and its activators (phosphodiesterase5 and protein kinase GII) in corin knockout mice comparedwith wild-type mice (69). Further exploration of the linkbetween corin and ENaC may further enhance our understand-ing of the salt retention in NS (48).

The Role of Aquaretics

In NS, vasopressin levels are reported to be elevated, leadingto the production of concentrated urine and impaired excretionof a water load (44, 65, 95). In keeping with this observation,nephrotic patients are often hyponatremic, consistent with a

relative water excess (92). Type 2 vasopressin receptors(AVPR2) mediate urinary concentration in the collecting duct,and AVPR2 inhibitors have proven effective in the treatment ofedema (1). Konomoto et al. (51) highlighted a potential role ofvasopressin in edema formation in a child with relapsingsteroid-sensitive NS who developed cranial diabetes insipidus.Relapses of NS before the development of cranial diabetesinsipidus and after treatment with DDAVP were associatedwith edema. However, when a relapse occurred in the presenceof untreated cranial diabetes insipidus, the patient was edemafree despite high levels of proteinuria and low plasma albumin.In another report, a child with steroid-resistant NS and edema,pleural and pericardial effusions resistant to treatment withalbumin/furosemide responded significantly to tolvaptan (aselective, competitive vasopressin receptor 2 antagonist), de-spite persistent nephrotic range proteinuria and unchangedplasma albumin levels (79).

Fig. 2. Plasmin stimulates ENaC-mediatedsodium reabsorption in proteinuria. Duringproteinuria, plasminogen that is present inthe tubular lumen is activated to plasmin byurokinase-type plasminogen activator (uPA),which is bound to its receptor (uPAR). Plas-min can subsequently activate ENaC eitherthrough interaction with prostasin or throughdirect cleavage of the �-ENaC subunits.Adapted from Ref. 84 with permission.

Perspective

F578

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org

Page 5: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

While these case reports offer a novel approach to treatingedema in nephrotic patients, injudicious use of aquaretics maybe fraught with potential risks of serious hypovolemia, hyper-natremic dehydration, shock, and thrombosis (6), especially inthe consideration of the thrombophilic tendencies in NS (80).Aquaretics are, thus, best avoided in NS.

“Urearetics”: Blocking Urea Transporter Proteins

Since urea transport plays an important role in urinaryconcentration, urea-blocking agents may represent anothertherapeutic tool to treat NS-related edema (67, 72). Ureatransporter (UT) proteins facilitate the passive transport ofurea, driven by a concentration gradient, across cell mem-

branes, and are essential in the urinary concentrating mecha-nism (28). Different isoforms of the transporters UT-A andUT-B are expressed in the descending limb and the collectingduct of the medullary nephrons (28, 45, 81, 85, 98). UTreceptor accumulation in the plasma membrane is regulatedmainly by vasopressin (via V2 receptors) (4, 5, 41, 49, 106)and hypertonicity (3, 99, 100). UT expressed in the innermedullary collecting duct is responsible for urea reabsorption,which maintains a hypertonic medullary interstitium. Thehighly urea-permeable fenestrated ascending vasa recta facili-tate transfer of urea from the inner to the outer medulla. Theinterstitial urea from the ascending vasa recta is partiallyreabsorbed by the nonfenestrated descending vasa recta and the

Fig. 3. Urea transporter blockade by “urearetics”, salt-sparing diuretics. Aquaporin and urea transporters in the descending thin limb of the loop of Henle andinner medullary collecting duct facilitate the passive transport of urea, driven by a concentration gradient, across cell membranes, and are essential in the urinaryconcentrating mechanism. Urea transporter inhibition and its subsequent effect on the countercurrent mechanism may be promising therapeutic targets as it hasthe potential to be effective in refractory edema, producing a relatively salt-sparing diuresis compared with conventional diuretics. Adapted from Ref. 33.

Perspective

F579

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org

Page 6: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

thin descending limb of the loop of Henle by a countercurrentexchange mechanism in which urea recycling conserves renalurea (67, 72, 73). UT-deficient mice demonstrate a reducedcapacity to concentrate urea in the urine, manifesting as areduction in maximum urinary osmolality and up to a twofoldincrease in daily urine output (32, 104) (Fig. 3).

Various small-molecule inhibitors of UT isoforms have beendeveloped and tested successfully as diuretics in differentrodent models (29, 30, 55, 105). Selective UT-A isoforminhibition has an added advantage for potential diuretic therapyin that it is primarily expressed in the kidneys, reducing thepotential of extrarenal effects compared with UT-B, which isrelatively ubiquitous (28). These studies collectively highlighturea transporter inhibition as potential targets for therapeuticdrug development. Their effect on the countercurrent mecha-nism may be promising as it has the potential to be effective inrefractory edema, producing a relatively salt-sparing diuresiscompared with conventional diuretics. Its use (with or withoutconventional diuretics) may be effective in hyponatremia dueto the syndrome of inappropriate antidiuretic hormone secre-tion and in fluid-overload states like congestive cardiac failure,chronic liver failure, chronic kidney disease, and NS. However,further testing of these inhibitors in animal models, and ulti-mately in clinical trials, is needed to define their safety andefficacy before mainstream therapeutic use.

Alternative View on Extrarenal Sodium Balance

Conventional understanding of sodium balance maintainsthat extracellular body fluids readily equilibrate across com-partments, that electrolyte concentrations in various compart-ments are constant, and that the kidney is the main player incontrolling the body’s sodium content. This paradigm has beenquestioned by recent studies that showed “kidney-like” lym-phatic and blood vessel countercurrent systems in the skin,intestines, bone, and elsewhere in the body (53, 56, 86–88).The notion that the immune system plays a role as homeostaticregulator of interstitial electrolyte homeostasis and that saltinduces proinflammatory immune cell polarization has ex-tended the role of the immune system to physiological adap-tation, interstitial fluid matrix regulation, blood pressure con-trol, and cardiovascular disease (60, 101). The protein-rich andnegatively charged glycosaminoglycan-rich endothelial sur-face layer may act as a buffer to excess intravascular sodium,by binding and osmotically inactivating it (89), and a barrier towater entering the interstitium, which make this an intriguingarea of future study for sodium hemostasis in both health anddisease (reviewed in Ref. 63). These findings may have impli-cations for our understanding of the sodium and fluid retentionin NS and drive the development of entirely novel therapeuticagents.

Conclusion

Understanding the mechanism of edema formation in NS hasfascinated clinicians and scientists for decades, and the studyof renal physiology in the nephrotic state has shed light onnormal physiology. The observation that proteases in the glo-merular filtrate activate the sodium-retaining ENaC, thus pro-moting intrarenal salt retention, has yet to be translated into anew therapeutic approach, despite the availability of amiloride.Is this because of concerns about hyperkalemia and the poten-

tial ineffectiveness of ENaC inhibition alone, or is there moreto the story? Can aquaretics and urearetics be used to manageedema in NS? These key questions need to be answered.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

Author contributions: C.W.T., L.A.R., and D.G.N. provided conception anddesign of research; C.W.T., L.A.R., and D.G.N. prepared figures; C.W.T.,L.A.R., and D.G.N. drafted manuscript; C.W.T., L.A.R., and D.G.N. editedand revised manuscript; C.W.T., L.A.R., and D.G.N. approved final version ofmanuscript.

REFERENCES

1. Ali F, Guglin M, Vaitkevicius P, Ghali JK. Therapeutic potential ofvasopressin receptor antagonists. Drugs 67: 847–858, 2007.

2. Armaly Z, Assady S, Abassi Z. Corin: a new player in the regulation ofsalt-water balance and blood pressure. Curr Opin Nephrol Hypertens 22:713–722, 2013.

3. Blessing NW, Blount MA, Sands JM, Martin CF, Klein JD. Ureatransporters UT-A1 and UT-A3 accumulate in the plasma membrane inresponse to increased hypertonicity. Am J Physiol Renal Physiol 295:F1336–F1341, 2008.

4. Blount MA, Klein JD, Martin CF, Tchapyjnikov D, Sands JM.Forskolin stimulates phosphorylation and membrane accumulation ofUT-A3. Am J Physiol Renal Physiol 293: F1308–F1313, 2007.

5. Blount MA, Mistry AC, Fröhlich O, Price SR, Chen G, Sands JM,Klein JD. Phosphorylation of UT-A1 urea transporter at serines 486 and499 is important for vasopressin-regulated activity and membrane accu-mulation. Am J Physiol Renal Physiol 295: F295–F299, 2008.

6. Bockenhauer D. Draining the edema: a new role for aquaretics? PediatrNephrol 29: 767–769, 2014.

7. Bockenhauer D. Over- or underfill: not all nephrotic states are createdequal. Pediatr Nephrol 28: 1153–1156, 2013.

8. Bockenhauer D, Aitkenhead H. The kidney speaks: interpreting urinarysodium and osmolality. Arch Dis Child Educ Pract Ed 96: 223–227,2011.

9. Brown EA, Markandu ND, Roulston JE, Jones BE, Squires M,MacGregor GA. Is the renin-angiotensin-aldosterone system involved inthe sodium retention in the nephrotic syndrome? Nephron 32: 102–107,1982.

10. Brown EA, Markandu ND, Sagnella GA, Jones BE, MacGregor GA.Lack of effect of captopril on the sodium retention of the nephroticsyndrome. Nephron 37: 43–48, 1984.

11. Brown EA, Markandu ND, Sagnella GA, Squires M, Jones BE,Macgregor GA. Evidence that some mechanism other than the reninsystem causes sodium retention in nephrotic syndrome. Lancet 1237–1240, 1982.

12. Buehler B. Hereditary disorders of albumin synthesis. Ann Clin Lab Sci8: 283–286, 1978.

13. Cadnapaphornchai MA, Tkachenko O, Shchekochikhin D, SchrierRW. The nephrotic syndrome: pathogenesis and treatment of edemaformation and secondary complications. Pediatr Nephrol 29: 1159–1167, 2014.

14. Carattino MD, Hughey RP, Kleyman TR. Proteolytic processing of theepithelial sodium channel gamma subunit has a dominant role in channelactivation. J Biol Chem 283: 25290–25295, 2008.

15. Chandra M, Hoyer JR, Lewy JE. Renal function in rats with unilateralproteinuria produced by renal perfusion with aminonucleoside. PediatrRes 15: 340–344, 1981.

16. Churg J, Habib R, White RHR. Pathology of the nephrotic syndromein children: a report for the International Study of Kidney Disease inChildren. Lancet 760: 1299–1302, 1970.

17. Cil O, Besbas N, Duzova A, Topaloglu R, Peco-Antic A, Korkmaz E,Ozaltin F. Genetic abnormalities and prognosis in patients with congen-ital and infantile nephrotic syndrome. Pediatr Nephrol 30: 1279–1287,2015.

18. Davin JC. The glomerular permeability factors in idiopathic nephroticsyndrome. Pediatr Nephrol. doi:10.1007/s00467-015-3082-x. 2015.

19. de Seigneux S, Kim SW, Hemmingsen SC, Frøkiaer J, Nielsen S.Increased expression but not targeting of ENaC in adrenalectomized rats

Perspective

F580

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org

Page 7: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

with PAN-induced nephrotic syndrome. Am J Physiol Renal Physiol 291:F208–F217, 2006.

20. Deschenes G, Doucet A. Collecting duct (Na�/K�)-ATPase activity iscorrelated with urinary sodium excretion in rat nephrotic syndromes. JAm Soc Nephrol 11: 604–615, 2000.

21. Deschenes G, Guigonis V, Doucet A. [Molecular mechanism of edemaformation in nephrotic syndrome]. Arch Pediatr 11: 1084–1094, 2004.

22. Deschenes G, Wittner M, Di Stefano A, Jounier S, Doucet A. Col-lecting duct is a site of sodium retention in PAN nephrosis: a rationale foramiloride therapy. J Am Soc Nephrol 12: 598–601, 2001.

23. Dong N, Chen S, Wang W, Zhou Y, Wu Q. Corin in clinical laboratorydiagnostics. Clin Chim Acta 413: 378–383, 2012.

24. Dorhout EJ, Roos JC, Boer P, Yoe OH, Simatupang TA. Observa-tions on edema formation in the nephrotic syndrome in adults withminimal lesions. Am J Med 67: 378–384, 1979.

25. Dorhout Mees EJ, Koomans HA. Understanding the nephrotic syn-drome: what’s new in a decade? Nephron 70: 1–10, 1995.

26. Eddy AA, Symons JM. Nephrotic syndrome in childhood. Lancet 362:629–639, 2003.

27. Epstein AA. Concerning the causation of edema in chronic parenchy-matous nephritis: method for its alleviation. Am J Med 13: 638–647,1917.

28. Esteva-Font C, Anderson MO, Verkman AS. Urea transporter proteinsas targets for small-molecule diuretics. Nat Rev Nephrol 11: 113–123,2014.

29. Esteva-Font C, Cil O, Phuan PW, Su T, Lee S, Anderson MO,Verkman AS. Diuresis and reduced urinary osmolality in rats producedby small-molecule UT-A-selective urea transport inhibitors. FASEB J 28:3878–3890, 2014.

30. Esteva-Font C, Phuan PW, Anderson MO, Verkman AS. A smallmolecule screen identifies selective inhibitors of urea transporter UT-A.Chem Biol 20: 1235–1244, 2013.

31. Fang C, Shen L, Dong L, Liu M, Shi S, Dong N, Wu Q. Reducedurinary corin levels in patients with chronic kidney disease. Clin Sci(Lond) 124: 709–717, 2013.

32. Fenton RA, Chou CL, Stewart GS, Smith CP, Knepper MA. Urinaryconcentrating defect in mice with selective deletion of phloretin-sensitiveurea transporters in the renal collecting duct. Proc Natl Acad Sci USA101: 7469–7474, 2004.

33. Fenton RA, Knepper MA. Mouse models and the urinary concentratingmechanism in the new millennium. Physiol Rev 87: 1083–1112, 2007.

34. Garnett ES, Webber CE. Changes in blood-volume produced bytreatment in the nephrotic syndrome. Lancet 2: 798–799, 1967.

35. Geers AB, Koomans HA, Roos JC, Boer P, Dorhout Mees EJ.Functional relationships in the nephrotic syndrome. Kidney Int 26:324–330, 1984.

36. Gur A, Adefuin PY, Siegel NJ, Hayslett JP. A study of the renalhandling of water in lipoid necrosis. Pediatr Res 10: 197–201, 1976.

37. Hall G, Gbadegesin RA. Translating genetic findings in hereditarynephrotic syndrome: the missing loops. Am J Physiol Renal Physiol 309:F24–F28, 2015.

38. Hamm LL, Feng Z, Hering-Smith KS. Regulation of sodium transportby ENaC in the kidney. Curr Opin Nephrol Hypertens 19: 98–105, 2010.

39. Hinkes BG, Mucha B, Vlangos CN, Gbadegesin R, Liu J, Has-selbacher K, Hangan D, Ozaltin F, Zenker M, Hildebrandt F.Nephrotic syndrome in the first year of life: two thirds of cases are causedby mutations in 4 genes (NPHS1, NPHS2, WT1, and LAMB2). Pediat-rics 119: e907–e919, 2007.

40. Hughey RP, Carattino MD, Kleyman TR. Role of proteolysis in theactivation of epithelial sodium channels. Curr Opin Nephrol Hypertens16: 444–450, 2007.

41. Hwang S, Gunaratne R, Rinschen MM, Yu MJ, Pisitkun T, HoffertJD, Fenton RA Knepper MA, Chou CL. Vasopressin increases phos-phorylation of Ser84 and Ser486 in Slc14a2 collecting duct urea trans-porters. Am J Physiol Renal Physiol 299: F559–F567, 2010.

42. Ichikawa I, Rennke HG, Hoyer JR, Badr KF, Schor N, Troy JL,Lechene CP, Brenner BM. Role for intrarenal mechanisms in theimpaired salt excretion of experimental nephrotic syndrome. J Clin Invest71: 91–103, 1983.

43. Kaneko K, Tsuji S, Kimata T, Kitao T, Yamanouchi S, Kato S.Pathogenesis of childhood idiopathic nephrotic syndrome: a paradigmshift from T-cells to podocytes. World J Pediatr 11: 21–28, 2015.

44. Kapur G, Valentini RP, Imam AA, Mattoo TK. Treatment of severeedema in children with nephrotic syndrome with diuretics alone—aprospective study. Clin J Am Soc Nephrol 4: 907–913, 2009.

45. Karakashian A, Timmer RT, Klein JD, Gunn RB, Sands JM, Bag-nasco SM. Cloning and characterization of two new isoforms of the ratkidney urea transporter: UT-A3 and UT-A4. J Am Soc Nephrol 10:230–237, 1999.

46. Kim SW, de Seigneux S, Sassen MC, Lee J, Kim J, Knepper MA,Frøkiær J, Nielsen S. Increased apical targeting of renal ENaC subunitsand decreased expression of 11�HSD2 in HgCl2-induced nephroticsyndrome in rats. Am J Physiol Renal Physiol 290: F674–F687, 2006.

47. Kim SW, Wang W, Nielsen J, Praetorius J, Kwon TH, Knepper MA,Frøkiær J, Nielsen S. Increased expression and apical targeting of renalENaC subunits in puromycin aminonucleoside-induced nephrotic syn-drome in rats. Am J Physiol Renal Physiol 286: F922–F935, 2004.

48. Klein JD. Corin: an ANP protease that may regulate sodium reabsorptionin nephrotic syndrome. Kidney Int 78: 635–637, 2010.

49. Klein JD, Fröhlich O, Blount MA, Martin CF, Smith TD, Sands JM.Vasopressin increases plasma membrane accumulation of urea trans-porter UT-A1 in rat inner medullary collecting ducts. J Am Soc Nephrol17: 2680–2686, 2006.

50. Kleta R, Bockenhauer D. Bartter syndromes and other salt-losingtubulopathies. Nephron Physiol 104: 73–80, 2006.

51. Konomoto T, Tanaka E, Imamura H, Orita M, Sawada H, Nunoi H.Nephrotic syndrome complicated by idiopathic central diabetes insipidus.Pediatr Nephrol 29: 927–930, 2014.

52. Koomans HA, Braam B, Geers AB, Roos JC, Dorhout Mees EJ. Theimportance of plasma protein for blood volume and blood pressurehomeostasis. Kidney Int 30: 730–735, 1986.

53. Kopp C, Linz P, Dahlmann A, Hammon M, Jantsch J, Müller DN,Schmieder RE, Cavallaro A, Eckardt KU, Uder M, Luft FC, Titze J.23Na magnetic resonance imaging-determined tissue sodium in healthysubjects and hypertensive patients. Hypertension 61: 635–640, 2013.

54. Lecomte J, Juchmes J. So-called absence of edema in analbuminemia.Rev Med Liege 33: 766–770, 1978.

55. Levin MH, de la Fuente R, Verkman AS. Urearetics: a small moleculescreen yields nanomolar potency inhibitors of urea transporter UT-B.FASEB J 21: 551–563, 2007.

56. Linz P, Santoro D, Renz W, Rieger J, Ruehle A, Ruff J, Deimling M,Rakova N, Muller DN, Luft FC, Titze J, Niendorf T. Skin sodiummeasured with 23 Na MRI at 7.0 T. NMR Biomed 28: 54–62, 2015.

57. Lourdel S, Loffing J, Favre G, Paulais M, Nissant A, Fakitsas P,Creminon C, Feraille E, Verrey F, Teulon J, Doucet A, Deschenes G.Hyperaldosteronemia and activation of the epithelial sodium channel arenot required for sodium retention in puromycin-induced nephrosis. J AmSoc Nephrol 16: 3642–3650, 2005.

58. Ma HP, Eaton DC. Acute regulation of epithelial sodium channel byanionic phospholipids. J Am Soc Nephrol 16: 3182–3187, 2005.

59. Maas RJ, Deegens JK, Wetzels JF. Permeability factors in idiopathicnephrotic syndrome: historical perspectives and lessons for the future.Nephrol Dial Transplant 29: 2207–2216, 2014.

60. Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T,Machura K, Park JK, Beck FX, Müller DN, Derer W, Goss J,Ziomber A, Dietsch P, Wagner H, van Rooijen N, Kurtz A, HilgersKF, Alitalo K, Eckardt KU, Luft FC, Kerjaschki D, Titze J. Macro-phages regulate salt-dependent volume and blood pressure by a vascularendothelial growth factor-C-dependent buffering mechanism. Nat Med15: 545–552, 2009.

61. Meltzer JI, Keim HJ, Laragh JH, Sealey JE, Jan KM, Chien S.Nephrotic syndrome: vasoconstriction and hypervolemic types indicatedby renin-sodium profiling. Ann Intern Med 91: 688–696, 1979.

62. Noone D, Licht C. An update on the pathomechanisms and futuretherapies of Alport syndrome. Pediatr Nephrol 28: 1025–1036, 2013.

63. Olde Engberink RH, Rorije NM, Homan van der Heide JJ, van denBorn BJ, Vogt L. Role of the vascular wall in sodium homeostasis andsalt sensitivity. J Am Soc Nephrol 26: 777–783, 2015.

64. Oliver WJ. Physiologic responses associate with steroid-induced diure-sis in the nephrotic syndrome. J Lab Clin Med 62: 449–464, 1963.

65. Oliver WJ, Owings CL. Sodium excretion in the nephrotic syndrome.Relation to serum albumin concentration, glomerular filtration rate, andaldosterone excretion rate. Am J Dis Child 113: 352–362, 1967.

66. Palmer BF. Nephrotic edema—pathogenesis and treatment. Am J MedSci 306: 53–67, 1993.

Perspective

F581

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org

Page 8: Perspectives on edema in childhood nephrotic syndrome€¦ · NEPHROTIC SYNDROME (NS) is one of the most common child-hood kidney diseases with a cumulative prevalence of 16 in 100,000

67. Pannabecker TL. Comparative physiology and architecture associatedwith the mammalian urine concentrating mechanism: role of innermedullary water and urea transport pathways in the rodent medulla. AmJ Physiol Regul Integr Comp Physiol 304: R488–R503, 2013.

68. Peleg A, Jaffe AS, Hasin Y. Enzyme-linked immunoabsorbent assay fordetection of human serine protease corin in blood. Clin Chim Acta 409:85–89, 2009.

69. Polzin D, Kaminski HJ, Kastner C, Wang W, Krämer S, GambaryanS, Russwurm M, Peters H, Wu Q, Vandewalle A, Bachmann S,Theilig F. Decreased renal corin expression contributes to sodiumretention in proteinuric kidney diseases. Kidney Int 78: 650–659, 2010.

70. Rascher W, Tulassay T. Hormonal regulation of water metabolism inchildren with nephrotic syndrome. Kidney Int Suppl 21: S83–S89, 1987.

71. Reid CJ, Marsh MJ, Murdoch IM, Clark G. Nephrotic syndrome inchildhood complicated by life threatening pulmonary oedema. BMJ 312:36–38, 1996.

72. Sands JM. Critical role of urea in the urine-concentrating mechanism. JAm Soc Nephrol 18: 670–671, 2007.

73. Sands JM, Layton HE. The physiology of urinary concentration: anupdate. Semin Nephrol 29: 178–195, 2009.

74. Schild L. The epithelial sodium channel and the control of sodiumbalance. Biochim Biophys Acta 1802: 1159–1165, 2010.

76. Schrier RW. Pathogenesis of sodium and water retention in high-outputand low-output cardiac failure, nephrotic syndrome, cirrhosis, and preg-nancy. N Eng J Med 319: 1065–1072, 1988.

77. Schrier RW, Fassett RG. A critique of the overfill hypothesis of sodiumand water retention in the nephrotic syndrome. Kidney Int 53: 1111–1117, 1998.

78. Shalhoub RJ. Pathogenesis of lipoid nephrosis: a disorder of T-cellfunction. Lancet 2: 556–560, 1974.

79. Shimizu M, Ishikawa S, Yachi Y, Muraoka M, Tasaki Y, Iwasaki H,Kuroda M, Ohta K, Yachie A. Tolvaptan therapy for massive edema ina patient with nephrotic syndrome. Pediatr Nephrol 29: 915–917, 2014.

80. Singhal R, Brimble KS. Thromboembolic complications in the ne-phrotic syndrome: pathophysiology and clinical management. ThrombRes 118: 397–407, 2006.

81. Stewart GS, Fenton RA, Wang W, Kwon TH, White SJ, Collins VM,Cooper G, Nielsen S, Smith CP. The basolateral expression of mUT-A3in the mouse kidney. Am J Physiol Renal Physiol 286: F979–F987, 2004.

82. Svenningsen P, Andersen H, Nielsen LH, Jensen BL. Urinary serineproteases and activation of ENaC in kidney-implications for physiolog-ical renal salt handling and hypertensive disorders with albuminuria.Pflügers Arch 467: 531–542, 2015.

83. Svenningsen P, Bistrup C, Friis UG, Bertog M, Haerteis S, KruegerB, Stubbe J, Jensen ON, Thiesson HC, Uhrenholt TR, Jespersen B,Jensen BL, Korbmacher C, Skøtt O. Plasmin in nephrotic urineactivates the epithelial sodium channel. J Am Soc Nephrol 20: 299–310,2009.

84. Svenningsen P, Friis UG, Versland JB, Buhl KB, Møller FrederiksenB, Andersen H, Zachar RM, Bistrup C, Skøtt O, Jørgensen JS,Andersen RF, Jensen BL. Mechanisms of renal NaCl retention inproteinuric disease. Acta Physiol (Oxf) 207: 536–545, 2013.

85. Terris JM, Knepper MA, Wade JB. UT-A3: localization and charac-terization of an additional urea transporter isoform in the IMCD. Am JPhysiol Renal Physiol 280: F325–F332, 2001.

86. Titze J. Sodium balance is not just a renal affair. Curr Opin NephrolHypertens 23: 101–105, 2014.

87. Titze J, Dahlmann A, Lerchl K, Kopp C, Rakova N, Schröder A,Luft FC. Spooky sodium balance. Kidney Int 85: 759–767, 2014.

88. Titze J, Müller DN, Luft FC. Taking another “look” at sodium. Can JCardiol 30: 473–475, 2014.

89. Titze J, Shakibaei M, Schafflhuber M, Schulze-Tanzil G, Porst M,Schwind KH, Dietsch P, Hilgers KF. Glycosaminoglycan polymeriza-tion may enable osmotically inactive Na� storage in the skin. Am JPhysiol Heart Circ Physiol 287: H203–H208, 2004.

90. Tulassay T, Rascher W, Lang RE, Seyberth HW, Scharer K. Atrialnatriuretic peptide and other vasoactive hormones in nephrotic syndrome.Kidney Int 31: 1391–1395, 1987.

91. Uchimura K, Kakizoe Y, Onoue T, Hayata M, Morinaga J, YamazoeR, Ueda M, Mizumoto T, Adachi M, Miyoshi T, Shiraishi N, Sakai Y,Tomita K, Kitamura K. In vivo contribution of serine proteases to theproteolytic activation of �ENaC in aldosterone-infused rats. Am J PhysiolRenal Physiol 303: F939–F943, 2012.

92. Usberti M, Federico S, Meccariello S, Cianciaruso B, Balletta M,Pecoraro C, Sacca L, Ungaro B, Pisanti N, Andreucci VE. Role ofplasma vasopressin in the impairment of water excretion in nephroticsyndrome. Kidney Int 25: 422–429, 1984.

93. Usberti M, Gazzotti RM. Hyporeninemic hypoaldosteronism in patientswith nephrotic syndrome. Am J Nephrol 18: 251–255, 1998.

94. Usberti M, Gazzotti RM, Poiesi C, D’Avanzo L, Ghielmi S. Consid-erations on the sodium retention in nephrotic syndrome. Am J Nephrol15: 38–47, 1995.

95. Vande Walle J, Donckerwolcke R, Wimersma Greidanus TB, JolesJA, Koomans HA. Renal sodium handling in children with nephroticrelapse: relation to hypovolaemic symptoms. Nephrol Dial Transplant11: 2202–2208, 1996.

96. Vande Walle JG, Donckerwolcke RA, van Isselt JW, Joles JA,Koomans HA, Derkx FH. Volume regulation in children with earlyrelapse of minimal-change nephrosis with or without hypovolaemicsymptoms. Lancet 346: 148–152, 1995.

97. Vogt B, Favre H. Na�, K�-ATPase activity and hormones in singlenephron segments from nephrotic rats. Clin Sci (Lond) 80: 599–604,1991.

98. Wade JB, Lee AJ, Liu J, Ecelbarger CA, Mitchell C, Bradford AD,Terris J, Kim GH, Knepper MA. UT-A2: a 55-kDa urea transporter inthin descending limb whose abundance is regulated by vasopressin. AmJ Physiol Renal Physiol 278: F52–F62, 2000.

99. Wang Y, Klein JD, Froehlich O, Sands JM. Role of protein kinase C-�in hypertonicity-stimulated urea permeability in mouse inner medullarycollecting ducts. Am J Physiol Renal Physiol 304: F233–F238, 2013.

100. Wang Y, Klein JD, Liedtke CM, Sands JM. Protein kinase C regulatesurea permeability in the rat inner medullary collecting duct. Am J PhysiolRenal Physiol 299: F1401–F1406, 2010.

101. Wiig H, Schröder A, Neuhofer W, Jantsch J, Kopp C, Karlsen TV,Boschmann M, Goss J, Bry M, Rakova N, Dahlmann A, Brenner S,Tenstad O, Nurmi H, Mervaala E, Wagner H, Beck FX, Müller DN,Kerjaschki D, Luft FC, Harrison DG, Alitalo K, Titze J. Immune cellscontrol skin lymphatic electrolyte homeostasis and blood pressure. J ClinInvest 123: 2803–2815, 2013.

102. Wu Q. The serine protease corin in cardiovascular biology and disease.Front Biosci 12: 4179–4190, 2007.

103. Yan W, Sheng N, Seto M, Morser J, Wu Q. Corin, a mosaic trans-membrane serine protease encoded by a novel cDNA from human heart.J Biol Chem 274: 14926–14935, 1999.

104. Yang B, Bankir L, Gillespie A, Epstein CJ, Verkman AS. Urea-selective concentrating defect in transgenic mice lacking urea transporterUT-B. J Biol Chem 277: 10633–10637, 2002.

105. Yao C, Anderson MO, Zhang J, Yang B, Phuan PW, Verkman AS.Triazolothienopyrimidine inhibitors of urea transporter UT-B reduceurine concentration. J Am Soc Nephrol 23: 1210–1220, 2012.

106. Zhang C, Sands JM, Klein JD. Vasopressin rapidly increases phos-phorylation of UT-A1 urea transporter in rat IMCDs through PKA. Am JPhysiol Renal Physiol 282: F85–F90, 2002.

Perspective

F582

AJP-Renal Physiol • doi:10.1152/ajprenal.00229.2015 • www.ajprenal.org