Skip to main content
Physiological Genomics logoLink to Physiological Genomics
. 2017 Apr 7;49(5):261–276. doi: 10.1152/physiolgenomics.00017.2017

Role of renal transporters and novel regulatory interactions in the TAL that control blood pressure

Lesley A Graham 1,, Anna F Dominiczak 1, Nicholas R Ferreri 2
PMCID: PMC5451551  PMID: 28389525

Abstract

Hypertension (HTN), a major public health issue is currently the leading factor in the global burden of disease, where associated complications account for 9.4 million deaths worldwide every year. Excessive dietary salt intake is among the environmental factors that contribute to HTN, known as salt sensitivity. The heterogeneity of salt sensitivity and the multiple mechanisms that link high salt intake to increases in blood pressure are of upmost importance for therapeutic application. A continual increase in the kidney’s reabsorption of sodium (Na+) relies on sequential actions at various segments along the nephron. When the distal segments of the nephron fail to regulate Na+, the effects on Na+ homeostasis are unfavorable. We propose that the specific nephron region where increased active uptake occurs as a result of variations in Na+ reabsorption is at the thick ascending limb of the loop of Henle (TAL). The purpose of this review is to urge the consideration of the TAL as contributing to the pathophysiology of salt-sensitive HTN. Further research in this area will enable development of a therapeutic application for targeted treatment.

Keywords: thick ascending limb of the loop of Henle; hypertension; salt sensitivity; sodium, kidney


essential hypertension (HTN) is a highly hereditable trait of complex etiology, where multiple genetic and environmental factors contribute to blood pressure (BP) variation. The study of genetic architecture has proven useful to detect a small number of genes, loci, and single nucleotide polymorphisms that have appreciable effects on BP (40, 94, 123). Among the environmental factors that contribute to HTN, excessive dietary salt intake is one of the common and important risk factor (31). The link between dietary salt intake and HTN is well established, and a reduction in salt intake is known to lower BP (29, 137). Although individuals respond differently to dietary salt; the increase in BP with an increase in salt intake occurs in some individuals, whereas others show no significant change in BP (179). This phenomenon is known as salt sensitivity, which is estimated to be present in 51% of the hypertensive and 26% of the normotensive populations (67, 68). Although salt sensitivity is a well-established phenomenon in experimental and human HTN, the pathophysiological mechanisms are not fully elucidated (7). The heterogeneity of salt sensitivity and the multiple mechanisms that link high salt intake to increases in BP are important for clinical application. To this end many investigators over the decades have focused on elucidating the pathophysiological mechanisms of salt sensitivity and the associated risk of developing HTN using epidemiological studies (70, 148), clinical trials (1, 2, 53, 106, 180, 181), in vivo studies (11, 33, 36), and more recently systems approaches by means of metabolomics (110, 167), proteomics (58, 115), and transcriptomics [reviewed in (39, 125)]. Even so, historical research on salt handling and BP control has been vigorously perused, demonstrating long-term regulation of BP relies on the synchronized regulation of Na+ (sodium), K+ (potassium), and Cl (chloride) movement in the kidney to maintain blood volume and water balance (60, 70) (97).

The ability of the kidneys to absorb large quantities of Na+ relies on sequential actions at various segments along the nephron, each with highly specialized transport capacities (outlined in Fig. 1). Na+ transport along the nephron is flow dependent, and a basic form of communication between parts of the nephron occurs as a result of changes in Na+ delivery by altered glomerular filtration rate (GFR). These alterations affect Na+ reabsorption in the upstream segments of the nephron, provoking a change in delivery/absorption of Na+ to the downstream nephron (133) and suggesting the regulation of Na+ reabsorption is highly controlled at the distal nephron, and failure of these downstream segments has more adverse effects on Na+ homeostasis such that alterations that occur in the proximal tubule. We propose that sustained increases of Na+ reabsorption at the distal segments of the nephron is generated by a higher natriuretic pressure, and the specific nephron region where increased active uptake occurs as a result of variations in Na+ reabsorption is at the thick ascending limb of the loop of Henle (TAL). Accordingly, this review will discuss the evidence implicating variations of Na+ reabsorption at the TAL that are detrimental to sodium homeostasis and BP control. This review will direct future studies to consider the TAL when investigating the pathophysiological mechanisms of salt sensitivity and HTN, which will aid the search for precise therapies for essential HTN.

Fig. 1.

Fig. 1.

Schematic of a nephron segment. The nephron consists of a renal corpuscle, a proximal tubule, a loop of Henle, a distal tubule, and a collecting duct system. The renal corpuscle consists of glomerular capillaries and Bowman’s capsule. The proximal tubule initially forms several coils followed by a straight segment that descends into the medulla. The next segment is the loop of Henle, which consists of a straight section of the proximal tubule, the descending thin limb (which ends in a hair pin turn), the thin ascending limb, and the thick ascending limb. Near the end of the thick ascending limb, the nephron passes between its afferent and efferent arterioles. The short segment of the thick ascending limb is called the macula densa. The distal tubule begins a short distance beyond the macula densa and extends to the point in the cortex where 2 or more nephrons join to form the cortical collecting duct. The collecting ducts enter the medulla and become the outer medulla collecting ducts, and then the inner medullary collecting ducts. In terms of ion transport the proximal tubule is responsible for 60–70% of the filtered Na+, whereas 15–25% is absorbed by the loop of Henle. The distal tubule reabsorbs 5–10% and the collecting ducts only 1−2% (149).

Ion Transport along the TAL

Anatomy and morphology of the loop of Henle.

The loop of Henle encompasses the thin descending limb, the thin ascending limb, and the TAL (Fig. 2). Within the inner stripe of the outer medulla there are short looped nephrons that derive from superficial and midcortical nephrons. These segments have a short descending limb, and, close to the hairpin turn of the loop, these tubules merge into the TAL. In contrast, the long-looped nephrons (originating from juxtamedullary glomeruli) have a long ascending thin limb where the TAL begins at the boundary between the inner and outer medulla. The TAL proceeds immediately after the thin ascending limb of long-looped nephrons. Aquaporin 1 (AQP1) is expressed abundantly in the proximal tubule; however, expression of AQP1 is also used as a marker to determine the junction between thin limbs of the loops of Henle and the TAL (124). The TAL begins after an AQP1-negative segment of short-limbed nephrons, immediately following the AQP1-positive thin descending limb (119, 124). Near the end of the TAL, the nephron passes between its afferent and efferent arterioles and meets its parent glomerulus; the macula densa (MD) constitutes the tightly packed epithelial cells at the junction of the TAL and the distal convoluted tubule (DCT) (119).

Fig. 2.

Fig. 2.

Organization of the loop of Henle. Structures are as follows: 1, thin descending limb of the loop of Henle; 2, thin ascending limb of the loop of Henle; and 3, thick ascending limb of the loop of Henle. See main text for details relevant to the labeled structures.

Characteristics of the thin descending limb include high water permeability and low permeability for Na+ and urea, with exceedingly low expression of Na+-K+-ATPase. Therefore, this segment is unlikely to contribute markedly to renal Na+ reabsorption. Thin ascending limbs reabsorb some Na+, but this absorption is likely passive as Na+-K+-ATPase expression is low (150). The TAL is the major Na+ reabsorbing segment of the nephron accounting for ~20–25% of filtered sodium chloride (NaCl). Features of the TAL include maintenance of the extracellular fluid (ECF), regulation of arterial hemodynamics, and regulation of NaCl homeostasis contributing to the regulation of the urinary concentrating mechanisms. It is essential for the kidney to osmotically concentrate the urine above isotonicity; this is dependent on Na+ reabsorption in the absence of measurable water permeability (124). Specifically, active NaCl absorption by the TAL dilutes luminal fluid to drive the countercurrent gradient that generates the axial osmolality gradient in the outer medulla region (119). The MD cells sense luminal NaCl concentrations and share most transport properties with TAL cells even though they are distinct in aspects of tubuloglomerular feedback (TGF). An increase of luminal NaCl at the MD generates a signaling cascade that induces vasoconstriction of the afferent arterioles, which reduces GFR. The augmented uptake of NaCl at the MD via the Na+-K+-Cl transporter (NKCC2) mediates TGF (71, 133). These signals cause the release of ATP into the juxtaglomerular interstitium and the subsequent generation of the nucleoside adenosine that constricts afferent arterioles (93). Figure 3 outlines the main ion transporters found at each nephron segment.

Fig. 3.

Fig. 3.

Schematic of a nephron segment indicating ion transporter positions. PCT, proximal convoluted tubule; TAL, thick ascending limb of the loop of Henle; DCT, distal convoluted tubule.

Thick ascending limb – principal ion transporters.

At least 80% of NaCl uptake across the apical membrane of TAL cells is mediated by a cotransport process in which the influx of Na+ drives the uptake of Cl and K+. To achieve net salt absorption, apical electroneutral cotransport of Na+, K+, and Cl via NKCC2 is complemented by recycling of K+ via renal outer medullary potassium channel (ROMK), efflux of Cl via basolateral Cl channels (CLC-Kb/Barttin), coupled transport of Na+ and H+ via NHE3 transporters, basolateral extrusion of Na+ by Na+-K+-ATPase, and the final adjustment to urine concentration, K+ homeostasis, acid-base balance with Na+ reabsorption is accomplished by ENaC (8).

The Na+/H+ exchangers (NHEs) are typical Na+-coupled transporters that mediate the countertransport of one extracellular Na+ for one cytosolic proton (H+) at the luminal apical membrane in the TAL. NHE3 is confined mainly to the apical membrane of renal cells (6) and functions by direct reabsorption of filtered Na+ along with indirect reabsorption of bicarbonate (HCO3) and chloride and secretion of ammonium. The NHE3 antiporters are positioned at the luminal apical membrane in the proximal convoluted tubule and the TAL and are responsible for reabsorbing up to 70% of filtered Na+. NHE3 transports Na+ into the cell in exchange for H+ into the tubular lumen by the electrochemical gradient produced by Na+-K+-ATPase. The secreted H+ combines with HCO3 to form H2O and CO2, and the latter diffuses into cells. Carbonic anhydrase allows rapid conversion of H2CO3 into H2O and CO2. Secreted H+ generates OH inside the cell, which is then converted into HCO3 by combining with CO2. The leaves the cell via the basolateral Na+−3HCO3 cotransporter [adapted from (149)] (Fig. 4A).

Fig. 4.

Fig. 4.

Schematic of ion transport in the TAL by actions of NHE3 (A) and NKCC2 (B). ATP, adenosine triphosphate; CA, carbonic anhydrase; Ca+, calcium; CO2, carbon dioxide; Cl, chloride; H+, hydrogen; H2O, water; H2CO3, carbonic acid; HCO3, bicarbonate; K+, potassium; Mg+, magnesium; Na+, sodium; NKCC2, Na+-K+-Cl transporter; ROMK, renal outer medullary potassium channel.

NKCC2 is localized at the apical membrane of the TAL. The function of this cotransporter is to reabsorb Na+ without reabsorbing water. Na+, K+, and Cl are transported into the cell across the apical membrane at a stoichiometry of 1:1:2 via the electrochemical gradient of the sodium pump. Na+ and Cl are transported into the peritubular fluid, while K+ recycles back into the tubular lumen via ROMK (8). NKCC2 mediates ~90–100% of Cl reabsorption, and 30–50% of all Na+ transport; furthermore, its function is linked with paracellular Na+, Ca+, and Mg+ flux (73). For each Na+ ion transported into the cell by NKCC2, one Na+ ion is absorbed via paracellular pathways. Inhibition of NKCC2 results in decreased K+ exit through ROMK, halting NaCl reabsorption and paracellular transport of Ca+ and Mg+ (72, 76). ROMK channels supply a sufficient amount of K+ to the NKCC2 cotransporter to maintain reabsorptive transport of NaCl. As Na+ transport of NKCC2 is rate limited by the availability of luminal K+ in the TAL, changes in activity of ROMK have regulatory effects of NaCl reabsorption (46) (Fig. 4B). Previous immunolocalization experiments revealed that NKCC2 is expressed at the apical cell surface of the TAL and MD but also in subapical vesicles, where intracellular trafficking regulates NKCC2 membrane expression. Vasopressin has been shown to shuttle the subapical vesicles of NKCC2 to the apical surface, leading to enhanced activity and expression levels (54). The transporter undergoes constitutive exocytosis at the TAL (27), suggesting NKCC2 trafficking is a dynamic continuous event rather than a triggered action. It was reported by Ares et al. (8) that endocytosis and recycling of NKCC2 in the absence of stimuli result in the retrieval of the protein from the plasma membrane at a rate that is closely matched to that of exocytosis insertion. These reports suggest that under basal conditions and in the absence of stimuli dynamic trafficking of NKCC2 occurs. Three NKCC2 isoforms (A, B, and F) are derived by differential splicing of exon 4 of the SLC12A1 gene. These isoforms differ in their distribution along the nephron and account for 20, 10, and 70%, for A, B, and F, respectively, of the total NKCC2 transcript abundance in mice (28). NKCC2A is located in the medullary and cortical TAL and exhibits immediate affinity for Cl, whereas NKCC2B shows highest affinity for Cl and is primarily located in the MD cells and cortical region. NKCC2F has the lowest affinity for ion transport, however, and is mainly located in the medullary TAL. The relative distribution of these isoforms and affinity for ion transport match the luminal concentration of NaCl along the TAL. Mice lacking NKCC2A revealed lower urine osmolality and decreased Cl reabsorption with enhanced expression of NKCC2B indicating compensation (135).

The basolateral Na+-K+-ATPase is the primary exit pathway for Na+ at the basolateral membrane of TAL cells. This transporter is composed of α-, β-, and γ-subunits; studies have demonstrated the α-subunit at the TAL participates in the natriuretic response to salt load (101). Suppression of Na+-K+-ATPase activity markedly increases intracellular sodium (127). Studies have shown that rodents fed high-salt diets display reduced levels of Na+-K+-ATPase in the renal tissues (105) and pigs administered deoxycorticosterone plus 1% NaCl in the drinking water develop reduced natriuresis, increasing Na+-K+-ATPase inactivity by 30-fold and resulting in augmented arterial pressure (63). The activity of Na+-K+-ATPase generates an Na+ gradient that enables apical entry of Na+, K+, and Cl via NKCC2. Ouabain can inhibit the activity of Na+-K+-ATPase, leading to the collapse of the lumen positive potential. This in turn abolishes the transepithelial Na+-Cl transport at the TAL (75). Cl channels primarily mediate basolateral exit of Cl from TAL cells; there are two chloride channels coexpressed at the TAL, CLC-K1 and CLC-K2 (denoted CLC-NKA and CLC-NKB in humans), with the dominant Cl channel in the TAL being encoded by CLC-NKB (178). The characterization of “Barttin,” which is coexpressed with CLC-K2 in several nephron segments, including the TAL, has aided the physiological understanding of the TAL. The human CLC-NKA and CLC-NKB paralogs do not function correctly in the absence of Barttin coexpression. CLC-NKB coexpressed with Barttin is highly selective for Cl (44).

Paracellular transport.

Crossing the tubular epithelium can be performed in a single step (paracellular) or a two-step processes (transcellular). The paracellular route of ionic flow occurs when the substance goes through the matrix of the tight junctions that link each epithelial cell to its neighboring cell; an example of paracellular movement is the reabsorption of calcium and magnesium. The TAL reabsorbs ~50–60% of filtered magnesium and ~20% of filtered calcium, exclusively via the paracellular pathway (119). A lumen-positive transepithelial voltage (Vte) drives the paracellular reabsorption of Mg2+ and Ca2+. The generation of Vte can be caused by active transport due to apical K+ secretion through ROMK and basolateral Cl exit through ClC-Kb/barttin channels (78). Therefore, the paracellular channels function by being charge and size selective. In particular, the charge and size selectivity of the tight junctions of the epithelia is predominantly by the claudins, a large gene family of tetraspan transmembrane proteins. Claudins are the key components of the paracellular pathway. Defects in claudin-16 and -19 result in a range of renal diseases, including hypomagnesemia, hypercalciuria, and nephrolithiasis. The claudin-16 channel provides cation permeability to the tight junction, and the claudin-19 channel increases the cation selectivity of the tight junction alongside the diffusional Vte by selectively blocking anion permeation. Additionally, claudin-19 interacts with claudin-16 to increase the overall cation selectivity of the tight junction. Removal of either of these claudins results in the tight junctions losing cation selectivity, creating renal defects in Mg2+ and Ca2+ reabsorption (78). Genetic studies in humans suffering from familial hypomagnesemia with hypercalciuria and nephrocalcinosis (FHHNC) have demonstrated that claudin-16 and claudin-19 play an associated role in the cation selectivity of TAL tight junctions. In vivo the knockdown of claudin-16 increases Na+ absorption in the collecting duct, accompanied by increases of ECF volume after treatment with amiloride (77, 79). Mice lacking Claudin-19 exhibit increased natriuresis with marked elevations in serum aldosterone, both strains replicating the phenotypes of human FHHNC (79). Collectively, these studies illustrate the vital role of claudin-16 and claudin-19 at the tight junctions of the TAL, representing contributing significantly to the selectivity of transepithelial absorption of Na+, Ca2+, and Mg2+.

Regulation of Ion Transport at the Thick Ascending Limb by Altered Luminal Flow

Tubular luminal flow in the nephron constantly changes, where the rate of luminal flow varies acutely in response to changes of, e.g., glomerular hemodynamics and/or changes in upstream fluid reabsorption. Increases in luminal flow are associated with enhanced Na+ reabsorption at major sites along the nephron (2426, 128), and chronic increases in luminal flow are observed in HTN. Thus it is important to understand the cellular mechanisms in which each nephron segment senses and responds to changes in flow by appropriately altering transport rates. In vivo microperfusion studies have provided much of the evidence for flow-dependent NaCl reabsorption along the TAL. The mechanisms of signaling events and interactions between soluble mediators and mechanical forces at the TAL have yet to be defined. Nevertheless, the actions of furosemide on NaCl reabsorption enforces the notion that flow dependence of loop Na+ transport is mediated mainly by the TAL (153, 185). Wright and Schnermann (185) report a fourfold increase in flow rate leads to a doubling of NaCl reabsorption, demonstrating that fractional reabsorption of NaCl falls with increasing flow rates. Flow dependence of TAL NaCl transport relies on the activity of NKCC2 (57). At times of low luminal flow, NaCl concentrations decrease along the TAL. If flow rate increases, the drop in NaCl concentration is reduced, leading to higher luminal concentrations of NaCl, which augment NaCl transport. In these conditions, minimum NaCl concentrations are not reached, resulting in flow-dependent increases in NaCl concentration at the TAL (133). The regulation of function along the TAL and NaCl transport capacities at this nephron segment are regulated by numerous biological factors, such as autacoids, hormones, and eicosanoids. Increased intracellular cAMP stimulates transepithelial NaCl transport in the TAL, specifically vasopressin (AVP), parathyroid hormone, glucagon, calcitonin, and β-adrenergic receptor signaling (49). This activation can subsequently be modulated by a number of negative influences, notably prostaglandin E2, extracellular Ca2+, hormones, and autocoids working via cGMP-dependent signaling, including nitric oxide, all having powerful negative effects on NaCl transport within the TAL (8).

Mechanisms that activate NKCC2.

The most extensively studied activating modulator of NaCl transport in the TAL is AVP, mediated via the V2 receptor (122). Vasopressin activates NKCC2 in perfused mouse TAL segments, exerting longer-term effects on NKCC2 expression and function at the apical surface. Previous immunohistochemical localization experiments revealed that NKCC2 is expressed not only at the apical cell surface but also in subapical vesicles, where intracellular trafficking regulates NKCC2 membrane expression. AVP has been shown to facilitate the shuttling of NKCC2 in subapical vesicles to the apical membrane, leading to enhanced activity and expression levels (54). In vivo studies have demonstrated that the phosphorylation of NKCC2 is associated with a cluster of NH2-terminal threonines residues (54). Rats treated with desmopressin showed induced phosphorylation of these residues, which are substrates for the homologous STE20/SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1) kinases (54). These kinases mediate hypotonicity and low chloride-induced activation of NKCC2 and sodium reabsorption. Studies have shown that NKCC2 phosphorylation is mainly driven by OSR1 abundance, whereas SPAK abundance is responsible for NCC phosphorylation (99, 116, 190). SPAK and OSR1 are activated by upstream WNK [with no lysine (K)] kinases, and the NH2 terminus of NKCC2 contains binding sites for SPAK and OSR1 (35) (51), where OSR1 kinase has been reported as responsible for NH2-terminal phosphorylation of NKCC2 and is critical for activity of the transporter. Reduced NH2-terminal NKCC2 phosphoprotein in mice with targeted TAL-specific deletion of OSR1 results in the loss of function of the TAL. Lin et al. (99) generated global and tubule-specific ORS1 knockout mice and found global knockout was embryonically lethal, whereas tubule-specific OSR1+/− mice displayed lower BP-associated and reduced phosphorylated NKCC2 and NCC. The tubule-specific knockout mice had reduced Na+ reabsorption at the TAL and a blunted response to furosemide with significantly reduced pNKCC2. The study also demonstrated that total expression of SPAK and pSPAK was increased in parallel to NCC despite unchanged NKCC2 expression, suggesting that ORS1 is necessary for BP and renal sodium reabsorption via activation of NKCC2. The only upstream stimulators of SPAK/OSR1 are the family of WNKs: WNK1, WNK2, WNK3, and WNK4. Phenotypic studies of WNK “knock-in” mice have demonstrated the role of the upstream WNK kinases in which mutant SPAK or OSR1 cannot be activated by upstream WNK kinases; these mice have reduced phosphorylation of NKCC2 and NCC, with associated salt-sensitive hypotension (119, 141). These studies report that WNK kinases appear to regulate SPAK/OSR1 and NKCC2 in chloride-dependent fashion, phosphorylating and activating SPAK/OSR1 and the transporter in response to changes in intracellular Cl activity as a result of altered luminal flow and NaCl uptake at the TAL (136). The effect of hypotonicity/low Cl on NKCC2 is enhanced/activated by WNK3, i.e., decreased intracellular Cl induces phosphorylation of NKCC2 to enhance its activity. The loss of WNK3 expression is compensated by WNK1 (117). One group used wild-type (WT) and WNK3−/− mice and studied GFR, renin levels, urine output, and urine osmolarity during a normal diet and a salt-restricted diet. WNK1 was markedly increased in WNK3−/− mice compared with WT during a normal diet. During a salt-restricted diet, levels of pSPAK/OSR1, pNKCC2, and pNCC were upregulated in WNK3−/− mice. These mice also displayed increased diuresis in response to hydrochlorothiazide. Therefore, WNK3 may function as a Cl sensor in TAL cells (130).

WNKs are thought to regulate ROMK by interacting with intersectin and influencing clathrin-mediated endocytosis. He et al. (69) have demonstrated that ROMK is inhibited by WNK1 and WNK4 due to increased endocytosis of the channel. WNK4 phosphorylates tight junction proteins claudins 1–4 to regulate the paracellular chloride permeability (86, 187). This paracellular chloride permeability in cells expressing mutant WNK4 is much greater than that of cells expressing WT WNK4 proteins, demonstrating that HTN in patients with WNK4 mutations is caused by increased NaCl reabsorption (18, 87, 188). The endocytosis occurs via clathrin-coated vesicles, among these, intersectin, which contains Src-homology 3 domains that can interact with PXXP proline-rich motifs (88). The interaction between WNK1 and WNK4 was specific to intersectin via the WNK proline-rich motifs but not their kinase activity. When intersectin was knocked out the endocytosis of ROMK was prevented, providing a molecular mechanism for stimulation of ROMK endocytosis and reduced expression by WNK kinases.

Tumor necrosis factor as a negative regulator of the TAL.

Tumor necrosis factor (TNF) is produced by and affects the functions of several renal cell types including proximal tubule, TAL, and collecting duct (CD), as well as podocytes and mesangial cells (34, 52, 83, 91, 107, 109, 113, 147). While TNF is best known for its proinflammatory actions, this cytokine also exhibits immunosuppressive, immunomodulatory, and regulatory effects in diverse tissues including kidney, lung, and colon, where active Na+ transport is important for fluid clearance and other transport mechanisms (5, 21, 30, 42, 43, 114, 146). Data from TNF knockout mice and the clinical use of anti-TNF drugs have exposed the intrinsic effects of this cytokine that preclude the neutralization of TNF in heart failure and systemic lupus erythematosus, for example (42, 82, 89, 112, 134, 169, 184, 193). Context-dependent effects of TNF also are observed in BP regulation, where studies have documented the prohypertensive effects of TNF in experimental models involving inflammation; however, TNF does not increase BP in all models of HTN and does not elevate BP per se when given to normal animals (4, 41, 45, 61, 92, 111, 120, 144, 164, 173, 175). Collectively, these studies infer the importance of determining the mechanisms that subserve both the beneficial and deleterious effects of TNF in individual cell types as an approach that may facilitate the development of cell-targeted therapies.

Moreover, it is important to determine if the autocrine inhibitory effects of TNF on TAL cells involving NKCC2 in vitro, which are consistent with the TNF-dependent polyuria and natriuresis observed in various studies, are operational in vivo (14, 43, 45, 64, 154, 155, 171). Defining the role of TNF in vivo for individual cell types is noteworthy since there are conflicting accounts, for instance, of TNF effects on amiloride-sensitive Na+ transport, where it inhibits transport in vivo (108) while stimulating it in vitro (176). Similarly, the sensitizing effect of TNF to increase ENaC activity in distal tubules from diabetic rats contrasts with its effects in control rats or mice where TNF inhibits ENaC activity (12, 38, 108). The heterogeneity of tubular epithelial cells may necessitate distinct regulatory mechanisms for each cell type by molecules such as TNF that affect the expression and activity of assorted transporters including NHE8, SGLT2, NKCC2, ENaC, and Na+-K+-ATPase (12, 14, 38, 109, 176, 186). Renal transporters must be tightly regulated and coordinated to maintain Na+ and BP homeostasis, and new models are being generated to help advance the field by uncovering the in vivo effects of TNF derived from an individual nephron segment, the TAL, where variations in Na+ reabsorption have been linked to HTN (47, 85). Understanding how autocrine cascades in the TAL operate in normal and diseased states may provide insight toward the development of new “loop diuretic” therapies that could offer fine-tuning of Na+ reabsorption via discrete targeting of regulatory molecules.

Uromodulin

Protein structure.

Uromodulin (UMOD) is a kidney protein exclusively synthesized at the TAL and is encoded by the UMOD gene. Based on the cDNA sequence, the UMOD precursor is composed of 640 amino acid residues, and motifs include signal sequence residues 1–24, one epidermal growth factor-like and two calcium-binding epidermal growth factor-like domains (residues 31–64, 65–107, and 108–149), one zona pellucid domain at residues 334–585 (this is essential for polymerization), eight potential N-glycosylation sites, and one stretch of hydrophobic amino acids similar that acquire glycosylphosphatidylinositol (GPI) attachment site (residue 614). There are 48 cysteine residues involved in disulfide bond formation (62). It was proposed an additional epidermal growth factor-like domain spanning residues 281–336 was present in the region between the second calcium-binding epidermal growth factor domain and zona pellucida domain. However, a new domain, D8C (residues 199–287), common to families of proteins including liver-specific zona pellucida, glycoprotein 2, UMOD, and several other uncharacterized proteins, was described by Yang et al. (189).

UMOD is GPI anchored to the apical plasma membrane; thus its biosynthesis and intracellular trafficking proceed through a secretory pathway. During biosynthesis, the UMOD precursor is translocated to the ER (endoplasmic reticulum). The signal peptide is then cleaved, and the protein glycosylated on seven of the eight potential N-glycosylation sites. Disulfide bridges are formed and glypiation on its COOH terminus occurs; the Golgi apparatus further modifies the N-glycan moieties. The mature glycan moieties and the GPI modifications act to route the protein to the apical membrane of epithelial cells in the TAL; this is when UMOD is finally GPI anchored and facing the tubular lumen; here it is said to form supramolecular structures to ensure its proposed physiological properties are performed (81). The protein is released for the lumen side of the membrane by specific but currently unidentified protease(s). Proteolytic cleavage was originally thought to occur after residue F548 (50), but later thought F587 (151). Proteolytic cleavage of the GPI anchor still remains to be determined; however, once cleaved from the apical membrane UMOD forms polymers in the urine and becomes the most abundant protein in mammalian urine.

Physiological and pathological roles of UMOD.

Due to UMOD's structure it has one peculiar feature: it has a tendency to aggregate with gel-like properties in solution when NaCl concentrations are close to 100 mmol/l or CaCl (calcium chloride) is 1 mmol/l (15, 23), which may be responsible for the binding properties of the protein. This, along with the GPI anchoring and multidomain structure on the luminal side of the apical membrane, in conjunction with rich and highly variable posttranslational protein modification, and a large presence of polymerized protein in the urine, suggests that UMOD may play multiple roles with site-specific physiological functions. However, over 60 yr of research has not elucidated the biological role of UMOD. Sorting of the apical membrane proteins requires GPI anchors, N-glycosylation, and polymerization; with the large turnover of UMOD and its half-life of 16 h it is assumed that the biosynthesis plays either a direct or indirect role in the formation of the apical membrane-targeted cargo vesicles and vesicle trafficking. GPI-anchored proteins associate with lipid raft domains that play a role in organizing the apical membrane and signaling transduction pathways (142). When the apical membrane is highly ordered it allows for close packing of GPI-anchored proteins on the surface of cell membranes; in the case of UMOD this may promote formation of the complex gel-like structures providing a water barrier at the luminal membrane of the TAL cells. This physical barrier to water permeability may play a role in ion transport to maintain countercurrent gradients in the interstitium (80).

As the TAL are nephron segments characterized by high electrolyte and water impermeability it was proposed that UMOD plays a role in salt transports and acts as a water barrier at this level, a process crucial for urine concentration. Bleyer et al. (19) reported low urine osmolality was consistent in subjects with UMOD mutations. A transgenic mouse model for uromodulin-associated kidney diseases (UAKD) confirmed urinary concentrating deficit (16). Water reabsorption by the tubule is regulated by transmembrane systems of aquaporin and ion channels (3), Bachmann and colleagues (10) studied the renal effects of UMOD deficiency in UMOD knockout mice and reported the inability of these mice to concentrate urine possibly due to a decrease in cyclooxygenase-2 (COX-2) expression. COX-2 inhibition prevents regulation of key renal water and sodium transport proteins including aquaporin 2, NHE3, and NKCC2 (126), confirming the role of UMOD in urine osmolality. More commonly in the clinic, UMOD is associated with renal cystic diseases. These are a major group of inherited renal conditions representing the leading cause of end-stage renal disease. Cystic kidney disease (CKD), in both the dominant and recessive variants, accounts for the clinical conditions. Hart et al. (66) and Rampoldi et al. (143) discovered autosomal dominant mutations in UMOD lead to medullary cystic kidney disease type 2 (MCKD2), familial juvenile hyperuricemic nephropathy (FJHN), and glomerular cystic kidney disease (GCKD). These conditions are characterized by urinary concentration deficits, urinary salt wasting, hyperuricemia, gout, medullary cysts, interstitial nephritis, glomerular cysts, hypertension, and end-stage renal failure. MCKD2 is an autosomal dominant disorder mainly characterized by HTN and end-stage renal failure, in the fourth decade of life, with renal complications including tubular membrane disintegration, tubular atrophy, with cyst development at the corticomedullary border and interstitial cell infiltration associated with fibrosis. MCKD2 has clinical and morphological overlap with the autosomal dominant FJHN. GCKD is characterized by a cystic dilatation of Bowman’s capsule and collapse of the glomerulus (191). All three disorders have significant clinical overlap and arise from UMOD mutations and are often referred to as UAKD and are said to cause the so called “uromodulin storage disease” (177, 183).

To date there have been more than 58 UMOD mutations reported, which mainly localize to exon 3 and 4 of the UMOD gene, with the majority being missense mutations or small in-frame deletions (95). Early genome-wide linkage mapping in Italian, Czech, and Belgian families revealed loci for MCKD and FJHN on chromosome 16 in the regions of 16p11.2 and 16p12 (32, 166) in close proximity to the UMOD gene. These conditions are associated with mutations that lead to amino acid changes at cysteine sites causing defective protein folding and immature UMOD being retained at the ER and not released at the apical membrane and remains intracellular (16, 143). Accumulation of misfolded UMOD in the ER causes ER stress and degradation by increased synthesis of chaperones and foldases that in turn activate the misfolded protein (90). This unfolded protein response may trigger apoptosis and autophagy or alternatively lead to cell activation via MAP kinases and NF-κB leading to the eventual progressive renal failure seen in MCKD2, FJHN, and GCKD (96). It is known that the transcription factor hepatic nuclear factor 1-β (HNFI-β) positively regulates UMOD expression and binds to the promoter elements of the gene. Inactivation of HNFI-β in vivo is associated with decreased UMOD transcription (100). Interestingly, mutations of HNFI-β are associated with features of MCKD2, FJHN, and GCKD (160). This transcription factor is also known to regulate nephrocystins, which prompted work by Zaucke et al. in 2010 (191) to investigate if UMOD is linked to ciliary cystogenesis. They reported seven novel UMOD mutations (missense or deletion mutations) between exons 4 and 5 and that UMOD is expressed in primary cilia of renal tubules and the number of UMOD positive tubules declines in UAKD. The mutations caused localization of UMOD in the mitotic spindle poles colocalized with nephrocystin-1, suggesting a novel cause of CKD pathologies of MCKD2, FJHN, and GCKD.

In the absence of UMOD there is susceptibility to calcium oxalate stones due to the lack of gel-like properties of the protein; thus altered salt concentration at the apical membrane is apparent (165). Mo et al. (118) showed that UMOD knockout mice spontaneously formed intrarenal crystals predominantly in the interstitial space and in the CD of the deep medulla and papilla. They reported that the stones consisted primarily of calcium phosphate in the form of hydroxyapatite and strongly resemble the stones found in humans caused by idiopathic calcium oxalate stones. The inhibitory effect of UMOD in stone aggregation had been described in cases of calcium oxalate (13, 37). There is consensus that inhibition of stone formation in normal urine is caused mainly by urinary macromolecules rather than low-molecular-weight components, and this property has been associated with polyanionic structures (48). UMOD is said to be a polyanionic macromolecule due to the large extent of sialylation and presence of sulfate groups bound to the N-linked glycans, thus playing a crucial role in regulating stone formation (172). Urinary tract infections (UTIs) are mainly caused by Escherichia coli and critically depend on filamentous appendages on the bacterial surface called fimbriae (13). Colonization is mediated by binding of lectin-like adhesins present on E. coli fimbriae to carbohydrate structures carried by glycoproteins exposed at the cell surface. E. coli fimbriae are classified according to their sugar specificity: type 1, type P, and type S. Pak et al. (132) illustrated that UMOD binds with type 1 E. coli fimbriae in vitro, and Raffi et al. (140) described UMOD as a general host defense mechanism against UTIs in vivo with a UMOD knockout mouse model, supporting the concept that urinary UMOD represents a protective mechanism against UTIs. In terms of urinary excretion, in adults, there appears to be considerable variation in daytime excretion values that does, however, correlate with urinary volumes brought about with diuresis in those drinking in response to thirst (103). A positive correlation between urinary UMOD and dietary salt intake revealed that in subjects with high salt sensitivity, i.e., with exaggerated BP response to high salt intake, there is a greater excretion of UMOD in the urine compared with low salt intake (168). UMOD excretion seems to increase gradually from birth to adulthood, where it remains stable until a decline after the sixth decade of life (129, 161), with urinary UMOD/creatinine ratio also remaining stable from the age of 4 yr until the age of 70 yr. Reduced urinary excretion of UMOD has also been correlated with GFR and corresponds to declining kidney function in virtually all chronic kidney diseases (138). In an attempt to elucidate why urinary UMOD excretion is altered in disease, Ma et al. (104) studied potential mechanisms using human UMOD mutations in polarized Madin Darby canine kidney cells. They report that cysteine mutations inside and out of the domain are able to specifically bind and trap UMOD, preventing it from exiting the ER and translocating to the cell surface. However, they found a specifically cysteine-altering mutation in the cysteine-rich domain had more severe deficits in ER exit and surface translocation, initiating increased apoptosis explaining partly in some diseases the marked reductions in urinary UMOD.

UMOD, BP, and ion transport.

Genome-wide association studies (GWAS) recently linked more common genetic variants in the UMOD promoter with the risk of HTN (131). Our previous GWAS identified a locus upstream of the UMOD gene transcriptional start site, which was associated with altered BP (131). It was reported that the minor G allele of rs13333226 when adjusted for estimated GFR was associated with a 7% lower risk of developing HTN. This GWAS discovery followed by functional validation studies have now resulted in a refocusing of interest in UMOD and its role in BP regulation (56, 170). Results from our recent studies in UMOD−/− mice suggest a biological link between UMOD and ion transport in the TAL. In the absence of UMOD there is augmented sodium excretion in the UMOD−/− mice, thought to be a consequence of reduced expression of NKCC2. This modulated Na+ reabsorption by reduced NKCC2 leads to exaggerated natriuresis and lower arterial pressure in the UMOD−/− mice, consistent with findings in humans where salt-wasting phenotypes and hypotension are characteristics of Bartter syndrome (139, 157, 174). In a complementary set of experiments, Trudu et al. (170) demonstrated that UMOD-transgenic mice overexpressing UMOD manifested salt-sensitive HTN, because of activation of the SPAK kinase and NH2-terminal phosphorylation of NKCC2. Biochemical and histological analysis of UMOD has been used to assess changes in biosynthesis of proteins related to NaCl transport along the nephron in UMOD−/− mice. Bachmann et al. (10, 121) revealed upregulation of major distal transporters (Na+-K+-ATPase, NKCC2, NHE3, ROMK, and ENaC) and downregulation of juxtaglomerular apparatus components. They reported that the augmented total NKCC2 expression was in fact a result of increased intracellular expression of NKCC2, where it remained unphosphorylated and inactive in UMOD−/− mice (121). This group concluded that in the absence of UMOD the reduced NKCC2 activity results in impaired NaCl reabsorption at the TAL, implying a permissive role of UMOD in the modulation of Na+ transport. ROMK resides in the apical membrane in the TAL and processes NaCl reabsorption via the recycling of K+ ions into the tubular lumen. It forms a functional unit with NKCC2, the rate-limiting transporter. Disrupted ROMK function at the TAL limits NaCl reabsorption, resulting in the salt-wasting phenotypes observed in Bartter syndrome. Renigunta et al. (145) identified UMOD as an ROMK-interacting protein, regulating ROMK function by increasing its expression at the cell surface of the TAL apical membrane. Furthermore, this group observed a decrease in ROMK immunoreactivity in the plasma membrane-enriched fractions of UMOD−/− mice kidney compared with UMOD+/+ counterparts. They demonstrated that UMOD ablation results in large accumulations of ROMK in the subapical vesicles, resulting in delayed or decreased surface expression, leading to salt wasting. This accumulation of ROMK is a result of the fluid entering the TAL having high concentrations of Na+ and Cl with low concentrations of K+. Therefore, efficient reabsorption of Na+ and Cl would be prevented as the stoichiometric flux of Na+ and K+ into the cell (via NKCC2) would quickly deplete K+ levels in the luminal fluid. This stimulation is normally avoided by the recycling of K+ entering the cell back into the lumen. Thus, the absence of K+ recycling results in markedly impaired NaCl reabsorption, which may be occurring in the UMOD−/− mice. A physiological regulator of NKCC2 transport rate is the Cl ion; in times of low luminal Cl the activity of NKCC2 is attenuated (22, 57). Mutig et al. (121) have shown that activation of NKCC2 is facilitated by UMOD in a Cl-sensitive manner. These findings suggest inactivation of NKCC2 and ROMK explains the phenotypes similar to Bartter we reported previously in these mice (56). To date the differential expression analysis of the major Na+-handling transporters in the kidney of UMOD+/+ and UMOD−/− mice before and after salt loading has not been explored.

Pathophysiology of the TAL

Given its role in renal physiology, it is not surprising that the TAL plays an important part in the pathophysiological role of disease. An extensive understanding of the TAL in renal ion transport systems will encourage precision medicine to provide stratified treatments for individuals. Single gene mutations present in Mendelian forms of HTN all increase Na+ reabsorption at the distal segments of the nephron, but not at the TAL (97, 182). Monogenic BP disorders where TAL ion gene expression is altered result in hypotension, suggesting a role for variants in the TAL to lower rather than raise BP, reducing the risk for HTN. Resequencing of three salt-handling genes [SLC12A3 (NCC), SLC12A1 (NKCC2), and KCNJ1 (ROMK)] in a cohort of over 5,000 subjects of the Framingham Heart study showed the large influence rare variants have on BP (84). The authors documented 30 mutations in these three genes that were inferred to have functional consequences. Carriers of any of the rare variants in the three salt-handling genes (with minor allele frequency ≤ 1%) had mean reductions of 6.3 mmHg in systolic BP (SBP) and 3.4 mmHg in diastolic BP compared with the entire cohort. Mutation carriers had mean SBP values 6.6 mmHg less than their noncarrier siblings. These are large effects when compared with those of common variants, for which the effect size is usually 1 mmHg or less (84). The authors conclusively demonstrated that these variants were associated with lower BP (~6–10 mmHg) and were protective against increased risk for HTN, emphasizing the contribution of sodium handling in BP regulation in the general population.

Studies so far suggest that variations in Na+ uptake by the TAL influence an individual’s chronic level of BP. Loss-of-function mutations in TAL Na+-Cl transport are associated with Bartter syndrome. Simon and colleagues (156158) reported that Bartter syndrome results from mutations in NKCC2 or its regulators and is referred to as type 1. Cl carried into the cell by NKCC2 exits through the Cl channels CLCNKA and CLCNKB with facilitation by BSND (barttin gene). Mutations in CLCNKB and BSND result in, respectively, types 3 and 4 Bartter syndrome. Bartter syndrome type 3 is caused by mutations of the Cl channel CLCNKB (156). NaCl entering the cell via NKCC2 at the apical membrane must exit the cell via the Na+-K+/ATPase at the basolateral membrane; however, this mutation prevents normal Cl exit because of inhibition of membrane localization. Thus, the functional activity of NKCC2 and BP control is determined by clearing excess Cl from TAL cells. Additionally, these patients may have an increase in urinary Ca2+ excretion and ~20% are hypomagnesemic (59). Upon binding of the Ca2+-sensing receptor on the basolateral surface in TAL leads to inhibition of NKCC2 (74), resulting in a more negatively charged lumen and less recycling of K+. These actions facilitate excretion of Ca2+, serving as a means for correcting and/or preventing a state of hypercalcemia. Loss-of-function mutations in ROMK also result in Bartter syndrome (type 2). The mutations results in salt wasting as fluid entering the TAL has increased concentrations of Na+ and Cl with low K+, thus the stoichiometric flux of Na+ and K+ via NKCC2 rapidly depletes K+ in the luminal fluid, which prevents efficient reabsorption of Na+ and Cl (157). This chain of events can be avoided by K+ entering back into the luminal fluid, implicating ROMK as the necessary channel for the recycling of K+ to maintain Na+ and Cl reabsorption, demonstrating the importance of synchronized actions of these channels in BP regulation.

Gitelman syndrome presents as a form of hypotension and renal sodium wasting caused by monogenic mutations. This condition is genetically homogeneous and is caused by loss-of-function mutations of the NCC transporter [Na-Cl cotransporter in the distal convoluted tubule (DCT)] (159). Patients usually present in adulthood with lower BP than the general population and display neuromuscular symptoms. These individuals have low serum Mg+ and low urinary Ca+ (17). The sodium wasting at the DCT activates the renin-angiotensin-aldosterone system (RAAS); however, in this condition there is augmented ENaC activity, protecting sodium homeostasis at the expense of increased H+ and K+ secretion. In order for Na+ reabsorption in TAL to reach a level sufficient to raise BP it requires help from another nephron site. For example, if Na+ reabsorption by NKCC2 were increased, the distal nephron should in response to the RAAS reduce Na+ downstream. Should these compensatory mechanisms not occur, a higher BP could develop. When investigating the role of the TAL to increase BP, one should consider the distal nephron or proximal tubule. Simultaneously, greater Na+ reabsorption elsewhere could require a more active TAL before manifesting in an increase in BP.

Role of the TAL in Salt-Sensitive Hypertension

A role for TAL in regulating BP in humans is difficult to establish in the clinical setting because of limitations essential for medical assessments. If TAL function increased, the increments in Na+ reabsorption necessary to raise BP would be relatively small and only enough over time to exceed any adjustments either proximally or distally. For example, ENaC in the CD reabsorbs only a small fraction of filtered Na+ (2%); thus Na+ reabsorption in TAL would need only to exceed this small fractional range to affect BP. A more active TAL could explain greater proximal tubular reabsorption of Na+. This would result in reduced TGF followed by increased glomerular filtration, thereby increasing delivery of Na+ to the proximal tubule that then increases absolute amounts of Na+ reabsorption (20). Indeed, Aviv et al. (9) extensively reviewed clinical studies demonstrating the TAL as a pivotal part of the nephron contributing to the heightened susceptibility salt-sensitive hypertension. A plausible mechanism responsible was proposed as a consequence of increased NKCC2 activity leading to an expansion of extracellular volume, decreased urinary K+ excretion, and increased Na+ reabsorption with greater water conservation. An increase in Na+ uptake by NKCC2 would enhance TGF (less Na+ reaching the MD), leading to glomerular hyperfiltration, delivering more Na+ to the proximal tubule, resulting in an increase in the absolute amount of Na+ reabsorbed in the proximal tubule and downstream in TAL (85). In 2004, Sonalker et al. (163) showed that spontaneously hypertensive rats (SHR) had fourfold higher expression levels of total NKCC2. This group later measured membrane-bound and total NKCC2 in the outer medulla in these rats by subcellular fractionation and found no difference in the surface to intracellular ratio of NKCC2 (162). Yet progression from normotension to hypertension (5–8 wk of age) in SHR was accompanied by a twofold increase in the surface to intracellular ratio of NKCC2 (54). Additionally, in Dahl salt-sensitive rats, NKCC2-dependent TAL transport, phosphorylation, and cell surface NKCC2 were enhanced upon salt loading with increased BP (65), indicating that salt stimuli modulate NKCC2 trafficking beyond essential requirements, altering Na+ uptake, ECF volume, and increasing arterial pressure. Zhou et al. (192) studied the effects of ROMKi B (a selective ROMK inhibitor) on systemic hemodynamics, renal function and structure, and vascular function in Dahl salt-sensitive rats on a high-salt diet. They reported a potential utility of ROMKi B as a novel antihypertensive agent, particularly for the treatment of the salt-sensitive HTN patient population as this inhibitor promoted increased natriuresis without any adverse effects on electrolytes and acid-base balance, kidney function, and structure.

Other TAL ion transporters may also contribute to salt-sensitive HTN; a high sodium intake increases the capacity of the TAL to absorb HCO3. Good et al. (55) examined the role of the apical NHE3 and basolateral NHE1 Na+/H+ exchangers in TAL tubules from salt-sensitive rats drinking normal tap water or 0.28 M NaCl for 5–7 days. They reported that high sodium intake increased HCO3 absorption rate by 60%, and this was mediated by an increase in apical NHE3 activity. Inhibiting basolateral NHE1 eliminated 60% of the HCO3 absorption, demonstrating that during a high-salt diet, these rats have increases in NHE3 activity dependent on NHE1. Studies in NHE3 knockout mice report chronic volume depletion and hypotension (152). Furthermore, NHE3−/− mice display blunted proximal fluid reabsorption without altered distal delivery of fluid (102). This compensation is largely attributable to decreased GFR due to activation of TGF, suggesting normalization of fluid delivery to the distal tubule is achieved through alterations in filtration rate and/or downstream transport processes. Reabsorption of filtered Na+ along the proximal convoluted tubule (PCT) in proportion to GFR suggests that rates of Na+ and fluid not being reabsorbed will change in proportion to the rate of filtration, denoting that notion that despite glomerulotubular balance, the delivery rate of Na+ to the tubular segments beyond the PCT will increase whenever GFR increases. Thus, Na+ homeostasis is regulated by TGF, a counterregulatory mechanism that senses an increase in NaCl delivery to the late nephron segments. These studies support a role of the TAL enabling the kidneys to regulate acid-base balance during changes in sodium and volume balance.

Future Direction

A considerable body of evidence links salt-sensitive HTN with the role of the TAL. Further research in this area will enable development of a therapeutic application for targeted treatment. This is crucial because despite major advances in cardiovascular health, hypertension remains the risk factor contributing most to the overall burden of disease globally, and there is a paucity of novel antihypertensive drugs in clinical trials or the pharmaceutical development pipeline.

DISCLOSURES

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

AUTHOR CONTRIBUTIONS

L.A.G. prepared figures; L.A.G. drafted manuscript; L.A.G., A.D., and N.R.F. edited and revised manuscript; N.R.F. approved final version of manuscript.

REFERENCES

  • 1.The effects of nonpharmacologic interventions on blood pressure of persons with high normal levels. Results of the Trials of Hypertension Prevention, Phase I. JAMA 267: 1213–1220, 1992. doi: 10.1001/jama.1992.03480090061028. [DOI] [PubMed] [Google Scholar]
  • 2.Sodium, potassium, body mass, alcohol and blood pressure: the INTERSALT Study. J Hypertens Suppl 6: S584–S586, 1988. doi: 10.1097/00004872-198812040-00183. [DOI] [PubMed] [Google Scholar]
  • 3.Agre P, Kozono D. Aquaporin water channels: molecular mechanisms for human diseases. FEBS Lett 555: 72–78, 2003. doi: 10.1016/S0014-5793(03)01083-4. [DOI] [PubMed] [Google Scholar]
  • 4.Alexander BT, Cockrell KL, Massey MB, Bennett WA, Granger JP. Tumor necrosis factor-alpha-induced hypertension in pregnant rats results in decreased renal neuronal nitric oxide synthase expression. Am J Hypertens 15: 170–175, 2002. doi: 10.1016/S0895-7061(01)02255-5. [DOI] [PubMed] [Google Scholar]
  • 5.Amasheh S, Barmeyer C, Koch CS, Tavalali S, Mankertz J, Epple H-J, Gehring MM, Florian P, Kroesen A-J, Zeitz M, Fromm M, Schulzke JD. Cytokine-dependent transcriptional down-regulation of epithelial sodium channel in ulcerative colitis. Gastroenterology 126: 1711–1720, 2004. doi: 10.1053/j.gastro.2004.03.010. [DOI] [PubMed] [Google Scholar]
  • 6.Amemiya M, Loffing J, Lötscher M, Kaissling B, Alpern RJ, Moe OW. Expression of NHE-3 in the apical membrane of rat renal proximal tubule and thick ascending limb. Kidney Int 48: 1206–1215, 1995. doi: 10.1038/ki.1995.404. [DOI] [PubMed] [Google Scholar]
  • 7.Ando K, Fujita T. Pathophysiology of salt sensitivity hypertension. Ann Med 44, Suppl 1: S119–S126, 2012. doi: 10.3109/07853890.2012.671538. [DOI] [PubMed] [Google Scholar]
  • 8.Ares GR, Caceres PS, Ortiz PA. Molecular regulation of NKCC2 in the thick ascending limb. Am J Physiol Renal Physiol 301: F1143–F1159, 2011. doi: 10.1152/ajprenal.00396.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Aviv A, Hollenberg NK, Weder A. Urinary potassium excretion and sodium sensitivity in blacks. Hypertension 43: 707–713, 2004. doi: 10.1161/01.HYP.0000120155.48024.6f. [DOI] [PubMed] [Google Scholar]
  • 10.Bachmann S, Mutig K, Bates J, Welker P, Geist B, Gross V, Luft FC, Alenina N, Bader M, Thiele BJ, Prasadan K, Raffi HS, Kumar S. Renal effects of Tamm-Horsfall protein (uromodulin) deficiency in mice. Am J Physiol Renal Physiol 288: F559–F567, 2005. doi: 10.1152/ajprenal.00143.2004. [DOI] [PubMed] [Google Scholar]
  • 11.Ball CO, Meneely GR. Observations on dietary sodium chloride. J Am Diet Assoc 33: 366–370, 1957. [PubMed] [Google Scholar]
  • 12.Bao HF, Zhang ZR, Liang YY, Ma JJ, Eaton DC, Ma HP. Ceramide mediates inhibition of the renal epithelial sodium channel by tumor necrosis factor-alpha through protein kinase C. Am J Physiol Renal Physiol 293: F1178–F1186, 2007. doi: 10.1152/ajprenal.00153.2007. [DOI] [PubMed] [Google Scholar]
  • 13.Bates JM Jr, Raffi HM, Prasadan K, Mascarenhas R, Laszik Z, Maeda N, Hultgren SJ, Kumar S. Tamm-Horsfall protein knockout mice are more prone to urinary tract infection: rapid communication. Kidney Int 65: 791–797, 2004. doi: 10.1111/j.1523-1755.2004.00452.x. [DOI] [PubMed] [Google Scholar]
  • 14.Battula S, Hao S, Pedraza PL, Stier CT, Ferreri NR. Tumor necrosis factor-alpha is an endogenous inhibitor of Na+-K+-2Cl- cotransporter (NKCC2) isoform A in the thick ascending limb. Am J Physiol Renal Physiol 301: F94–F100, 2011. doi: 10.1152/ajprenal.00650.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Benting JH, Rietveld AG, Simons K. N-Glycans mediate the apical sorting of a GPI-anchored, raft-associated protein in Madin-Darby canine kidney cells. J Cell Biol 146: 313–320, 1999. doi: 10.1083/jcb.146.2.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bernascone I, Vavassori S, Di Pentima A, Santambrogio S, Lamorte G, Amoroso A, Scolari F, Ghiggeri GM, Casari G, Polishchuk R, Rampoldi L. Defective intracellular trafficking of uromodulin mutant isoforms. Traffic 7: 1567–1579, 2006. doi: 10.1111/j.1600-0854.2006.00481.x. [DOI] [PubMed] [Google Scholar]
  • 17.Bettinelli A, Bianchetti MG, Girardin E, Caringella A, Cecconi M, Appiani AC, Pavanello L, Gastaldi R, Isimbaldi C, Lama G, Marchesoni C, Matteucci C, Patriarca P, Natale BD, Setzu C, Vitucci P. Use of calcium excretion values to distinguish two forms of primary renal tubular hypokalemic alkalosis: Bartter and Gitelman syndromes. J Pediatr 120: 38–43, 1992. doi: 10.1016/S0022-3476(05)80594-3. [DOI] [PubMed] [Google Scholar]
  • 18.Bindels RJ. A molecular switch controlling renal sodium and potassium excretion. Nat Genet 35: 302–303, 2003. doi: 10.1038/ng1203-302. [DOI] [PubMed] [Google Scholar]
  • 19.Bleyer AJ, Woodard AS, Shihabi Z, Sandhu J, Zhu H, Satko SG, Weller N, Deterding E, McBride D, Gorry MC, Xu L, Ganier D, Hart TC. Clinical characterization of a family with a mutation in the uromodulin (Tamm-Horsfall glycoprotein) gene. Kidney Int 64: 36–42, 2003. doi: 10.1046/j.1523-1755.2003.00081.x. [DOI] [PubMed] [Google Scholar]
  • 20.Bochud M, Staessen JA, Maillard M, Mazeko MJ, Kuznetsova T, Woodiwiss A, Richart T, Norton G, Thijs L, Elston R, Burnier M. Ethnic differences in proximal and distal tubular sodium reabsorption are heritable in black and white populations. J Hypertens 27: 606–612, 2009. doi: 10.1097/HJH.0b013e32832104b1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Braun C, Hamacher J, Morel DR, Wendel A, Lucas R. Dichotomal role of TNF in experimental pulmonary edema reabsorption. J Immunol 175: 3402–3408, 2005. doi: 10.4049/jimmunol.175.5.3402. [DOI] [PubMed] [Google Scholar]
  • 22.Briggs JP, Schnermann JB. Whys and wherefores of juxtaglomerular apparatus function. Kidney Int 49: 1724–1726, 1996. doi: 10.1038/ki.1996.255. [DOI] [PubMed] [Google Scholar]
  • 23.Brown DA, Rose JK. Sorting of GPI-anchored proteins to glycolipid-enriched membrane subdomains during transport to the apical cell surface. Cell 68: 533–544, 1992. doi: 10.1016/0092-8674(92)90189-J. [DOI] [PubMed] [Google Scholar]
  • 24.Cabral PD, Capurro C, Garvin JL. TRPV4 mediates flow-induced increases in intracellular Ca in medullary thick ascending limbs. Acta Physiol (Oxf) 214: 319–328, 2015. doi: 10.1111/apha.12528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cabral PD, Hong NJ, Garvin JL. ATP mediates flow-induced NO production in thick ascending limbs. Am J Physiol Renal Physiol 303: F194–F200, 2012. doi: 10.1152/ajprenal.00504.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cabral PD, Hong NJ, Garvin JL. Shear stress increases nitric oxide production in thick ascending limbs. Am J Physiol Renal Physiol 299: F1185–F1192, 2010. doi: 10.1152/ajprenal.00112.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Caceres PS, Ares GR, Ortiz PA. cAMP stimulates apical exocytosis of the renal Na(+)-K(+)-2Cl(-) cotransporter NKCC2 in the thick ascending limb: role of protein kinase A. J Biol Chem 284: 24965–24971, 2009. doi: 10.1074/jbc.M109.037135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Castrop H, Schnermann J. Isoforms of renal Na-K-2Cl cotransporter NKCC2: expression and functional significance. Am J Physiol Renal Physiol 295: F859–F866, 2008. doi: 10.1152/ajprenal.00106.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen J. Sodium sensitivity of blood pressure in Chinese populations. Curr Hypertens Rep 12: 127–134, 2010. doi: 10.1007/s11906-009-0088-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chen X, Oppenheim JJ. The phenotypic and functional consequences of tumour necrosis factor receptor type 2 expression on CD4(+) FoxP3(+) regulatory T cells. Immunology 133: 426–433, 2011. doi: 10.1111/j.1365-2567.2011.03460.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Choi HY, Park HC, Ha SK. Salt sensitivity and hypertension: a paradigm shift from kidney malfunction to vascular endothelial dysfunction. Electrolyte Blood Press 13: 7–16, 2015. doi: 10.5049/EBP.2015.13.1.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dahan K, Fuchshuber A, Adamis S, Smaers M, Kroiss S, Loute G, Cosyns JP, Hildebrandt F, Verellen-Dumoulin C, Pirson Y. Familial juvenile hyperuricemic nephropathy and autosomal dominant medullary cystic kidney disease type 2: two facets of the same disease? J Am Soc Nephrol 12: 2348–2357, 2001. [DOI] [PubMed] [Google Scholar]
  • 33.Dahl LK, Heine M, Tassinari L. Role of genetic factors in susceptibility to experimental hypertension due to chronic excess salt ingestion. Nature 194: 480–482, 1962. doi: 10.1038/194480b0. [DOI] [PubMed] [Google Scholar]
  • 34.David S, Biancone L, Caserta C, Bussolati B, Cambi V, Camussi G. Alternative pathway complement activation induces proinflammatory activity in human proximal tubular epithelial cells. Nephrol Dial Transplant 12: 51–56, 1997. doi: 10.1093/ndt/12.1.51. [DOI] [PubMed] [Google Scholar]
  • 35.Delpire E, Gagnon KB. Genome-wide analysis of SPAK/OSR1 binding motifs. Physiol Genomics 28: 223–231, 2007. doi: 10.1152/physiolgenomics.00173.2006. [DOI] [PubMed] [Google Scholar]
  • 36.Denton D, Weisinger R, Mundy NI, Wickings EJ, Dixson A, Moisson P, Pingard AM, Shade R, Carey D, Ardaillou R, Paillard F, Chapman J, Thillet J, Baptiste Michel J. The effect of increased salt intake on blood pressure of chimpanzees. Nat Med 1: 1009–1016, 1995. doi: 10.1038/nm1095-1009. [DOI] [PubMed] [Google Scholar]
  • 37.Diamond HS, Paolino JS. Evidence for a postsecretory reabsorptive site for uric acid in man. J Clin Invest 52: 1491–1499, 1973. doi: 10.1172/JCI107323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.DiPetrillo K, Coutermarsh B, Soucy N, Hwa J, Gesek F. Tumor necrosis factor induces sodium retention in diabetic rats through sequential effects on distal tubule cells. Kidney Int 65: 1676–1683, 2004. doi: 10.1111/j.1523-1755.2004.00606.x. [DOI] [PubMed] [Google Scholar]
  • 39.Dominiczak A. Sy 03-1 Genetic Basis of Blood Pressure and Hypertension. J Hypertens 34, Suppl 1 - ISH 2016 Abstract Book: e14, 2016. doi: 10.1097/01.hjh.0000499903.43288.23. 27753828 [DOI] [Google Scholar]
  • 40.Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang SJ, Pihur V, Vollenweider P, O’Reilly PF, Amin N, Bragg-Gresham JL, Teumer A, Glazer NL, Launer L, Zhao JH, Aulchenko Y, Heath S, Sõber S, Parsa A, Luan J, Arora P, Dehghan A, Zhang F, Lucas G, Hicks AA, Jackson AU, Peden JF, Tanaka T, Wild SH, Rudan I, Igl W, Milaneschi Y, Parker AN, Fava C, Chambers JC, Fox ER, Kumari M, Go MJ, van der Harst P, Kao WH, Sjögren M, Vinay DG, Alexander M, Tabara Y, Shaw-Hawkins S, Whincup PH, Liu Y, Shi G, Kuusisto J, Tayo B, Seielstad M, Sim X, Nguyen KD, Lehtimäki T, Matullo G, Wu Y, Gaunt TR, Onland-Moret NC, Cooper MN, Platou CG, Org E, Hardy R, Dahgam S, Palmen J, Vitart V, Braund PS, Kuznetsova T, Uiterwaal CS, Adeyemo A, Palmas W, Campbell H, Ludwig B, Tomaszewski M, Tzoulaki I, Palmer ND, Aspelund T, Garcia M, Chang YP, O’Connell JR, Steinle NI, Grobbee DE, Arking DE, Kardia SL, Morrison AC, Hernandez D, Najjar S, McArdle WL, Hadley D, Brown MJ, Connell JM, Hingorani AD, Day IN, Lawlor DA, Beilby JP, Lawrence RW, Clarke R, Hopewell JC, Ongen H, Dreisbach AW, Li Y, Young JH, Bis JC, Kähönen M, Viikari J, Adair LS, Lee NR, Chen MH, Olden M, Pattaro C, Bolton JA, Köttgen A, Bergmann S, Mooser V, Chaturvedi N, Frayling TM, Islam M, Jafar TH, Erdmann J, Kulkarni SR, Bornstein SR, Grässler J, Groop L, Voight BF, Kettunen J, Howard P, Taylor A, Guarrera S, Ricceri F, Emilsson V, Plump A, Barroso I, Khaw KT, Weder AB, Hunt SC, Sun YV, Bergman RN, Collins FS, Bonnycastle LL, Scott LJ, Stringham HM, Peltonen L, Perola M, Vartiainen E, Brand SM, Staessen JA, Wang TJ, Burton PR, Soler Artigas M, Dong Y, Snieder H, Wang X, Zhu H, Lohman KK, Rudock ME, Heckbert SR, Smith NL, Wiggins KL, Doumatey A, Shriner D, Veldre G, Viigimaa M, Kinra S, Prabhakaran D, Tripathy V, Langefeld CD, Rosengren A, Thelle DS, Corsi AM, Singleton A, Forrester T, Hilton G, McKenzie CA, Salako T, Iwai N, Kita Y, Ogihara T, Ohkubo T, Okamura T, Ueshima H, Umemura S, Eyheramendy S, Meitinger T, Wichmann HE, Cho YS, Kim HL, Lee JY, Scott J, Sehmi JS, Zhang W, Hedblad B, Nilsson P, Smith GD, Wong A, Narisu N, Stančáková A, Raffel LJ, Yao J, Kathiresan S, O’Donnell CJ, Schwartz SM, Ikram MA, Longstreth WT Jr, Mosley TH, Seshadri S, Shrine NR, Wain LV, Morken MA, Swift AJ, Laitinen J, Prokopenko I, Zitting P, Cooper JA, Humphries SE, Danesh J, Rasheed A, Goel A, Hamsten A, Watkins H, Bakker SJ, van Gilst WH, Janipalli CS, Mani KR, Yajnik CS, Hofman A, Mattace-Raso FU, Oostra BA, Demirkan A, Isaacs A, Rivadeneira F, Lakatta EG, Orru M, Scuteri A, Ala-Korpela M, Kangas AJ, Lyytikäinen LP, Soininen P, Tukiainen T, Würtz P, Ong RT, Dörr M, Kroemer HK, Völker U, Völzke H, Galan P, Hercberg S, Lathrop M, Zelenika D, Deloukas P, Mangino M, Spector TD, Zhai G, Meschia JF, Nalls MA, Sharma P, Terzic J, Kumar MV, Denniff M, Zukowska-Szczechowska E, Wagenknecht LE, Fowkes FG, Charchar FJ, Schwarz PE, Hayward C, Guo X, Rotimi C, Bots ML, Brand E, Samani NJ, Polasek O, Talmud PJ, Nyberg F, Kuh D, Laan M, Hveem K, Palmer LJ, van der Schouw YT, Casas JP, Mohlke KL, Vineis P, Raitakari O, Ganesh SK, Wong TY, Tai ES, Cooper RS, Laakso M, Rao DC, Harris TB, Morris RW, Dominiczak AF, Kivimaki M, Marmot MG, Miki T, Saleheen D, Chandak GR, Coresh J, Navis G, Salomaa V, Han BG, Zhu X, Kooner JS, Melander O, Ridker PM, Bandinelli S, Gyllensten UB, Wright AF, Wilson JF, Ferrucci L, Farrall M, Tuomilehto J, Pramstaller PP, Elosua R, Soranzo N, Sijbrands EJ, Altshuler D, Loos RJ, Shuldiner AR, Gieger C, Meneton P, Uitterlinden AG, Wareham NJ, Gudnason V, Rotter JI, Rettig R, Uda M, Strachan DP, Witteman JC, Hartikainen AL, Beckmann JS, Boerwinkle E, Vasan RS, Boehnke M, Larson MG, Järvelin MR, Psaty BM, Abecasis GR, Chakravarti A, Elliott P, van Duijn CM, Newton-Cheh C, Levy D, Caulfield MJ, Johnson T, International Consortium for Blood Pressure Genome-Wide Association Studies; CARDIoGRAM Consortium; CKDGen Consortium; KidneyGen Consortium; EchoGen Consortium; CHARGE-HF Consortium . Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature 478: 103–109, 2011. doi: 10.1038/nature10405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Elmarakby AA, Quigley JE, Imig JD, Pollock JS, Pollock DM. TNF-alpha inhibition reduces renal injury in DOCA-salt hypertensive rats. Am J Physiol Regul Integr Comp Physiol 294: R76–R83, 2008. doi: 10.1152/ajpregu.00466.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ernandez T, Mayadas TN. Immunoregulatory role of TNFalpha in inflammatory kidney diseases. Kidney Int 76: 262–276, 2009. doi: 10.1038/ki.2009.142. [DOI] [PubMed] [Google Scholar]
  • 43.Escalante BA, Ferreri NR, Dunn CE, McGiff JC. Cytokines affect ion transport in primary cultured thick ascending limb of Henle’s loop cells. Am J Physiol Cell Physiol 266: C1568–C1576, 1994. [DOI] [PubMed] [Google Scholar]
  • 44.Estévez R, Boettger T, Stein V, Birkenhäger R, Otto E, Hildebrandt F, Jentsch TJ. Barttin is a Cl- channel beta-subunit crucial for renal Cl- reabsorption and inner ear K+ secretion. Nature 414: 558–561, 2001. doi: 10.1038/35107099. [DOI] [PubMed] [Google Scholar]
  • 45.Evans DA, Jacobs DO, Revhaug A, Wilmore DW. The effects of tumor necrosis factor and their selective inhibition by ibuprofen. Ann Surg 209: 312–321, 1989. doi: 10.1097/00000658-198903000-00011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fang L, Li D, Welling PA. Hypertension resistance polymorphisms in ROMK (Kir1.1) alter channel function by different mechanisms. Am J Physiol Renal Physiol 299: F1359–F1364, 2010. doi: 10.1152/ajprenal.00257.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Felder RA, Jose PA. Mechanisms of disease: the role of GRK4 in the etiology of essential hypertension and salt sensitivity. Nat Clin Pract Nephrol 2: 637–650, 2006. doi: 10.1038/ncpneph0301. [DOI] [PubMed] [Google Scholar]
  • 48.Fellström B, Danielson BG, Ljunghall S, Wikström B. Crystal inhibition: the effects of polyanions on calcium oxalate crystal growth. Clin Chim Acta 158: 229–235, 1986. doi: 10.1016/0009-8981(86)90286-X. [DOI] [PubMed] [Google Scholar]
  • 49.Féraille E, Doucet A. Sodium-potassium-adenosinetriphosphatase-dependent sodium transport in the kidney: hormonal control. Physiol Rev 81: 345–418, 2001. [DOI] [PubMed] [Google Scholar]
  • 50.Fukuoka S, Kobayashi K. Analysis of the C-terminal structure of urinary Tamm-Horsfall protein reveals that the release of the glycosyl phosphatidylinositol-anchored counterpart from the kidney occurs by phenylalanine-specific proteolysis. Biochem Biophys Res Commun 289: 1044–1048, 2001. doi: 10.1006/bbrc.2001.6112. [DOI] [PubMed] [Google Scholar]
  • 51.Gagnon KB, England R, Delpire E. A single binding motif is required for SPAK activation of the Na-K-2Cl cotransporter. Cell Physiol Biochem 20: 131–142, 2007. doi: 10.1159/000104161. [DOI] [PubMed] [Google Scholar]
  • 52.Gao HX, Campbell SR, Burkly LC, Jakubowski A, Jarchum I, Banas B, Saleem MA, Mathieson PW, Berman JW, Michaelson JS, Putterman C. TNF-like weak inducer of apoptosis (TWEAK) induces inflammatory and proliferative effects in human kidney cells. Cytokine 46: 24–35, 2009. doi: 10.1016/j.cyto.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 53.GenSalt Collaborative Research Group GenSalt: rationale, design, methods and baseline characteristics of study participants. J Hum Hypertens 21: 639–646, 2007. doi: 10.1038/sj.jhh.1002207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Giménez I, Forbush B. Short-term stimulation of the renal Na-K-Cl cotransporter (NKCC2) by vasopressin involves phosphorylation and membrane translocation of the protein. J Biol Chem 278: 26946–26951, 2003. doi: 10.1074/jbc.M303435200. [DOI] [PubMed] [Google Scholar]
  • 55.Good DW, George T, Watts BA 3rd. High sodium intake increases HCO3 absorption in medullary thick ascending limb through adaptations in basolateral and apical Na+/H+ exchangers. Am J Physiol Renal Physiol 301: F334–F343, 2011. doi: 10.1152/ajprenal.00106.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Graham LA, Padmanabhan S, Fraser NJ, Kumar S, Bates JM, Raffi HS, Welsh P, Beattie W, Hao S, Leh S, Hultstrom M, Ferreri NR, Dominiczak AF, Graham D, McBride MW. Validation of uromodulin as a candidate gene for human essential hypertension. Hypertension 63: 551–558, 2014. doi: 10.1161/HYPERTENSIONAHA.113.01423. [DOI] [PubMed] [Google Scholar]
  • 57.Greger R. Ion transport mechanisms in thick ascending limb of Henle’s loop of mammalian nephron. Physiol Rev 65: 760–797, 1985. [DOI] [PubMed] [Google Scholar]
  • 58.Grussenmeyer T, Meili-Butz S, Roth V, Dieterle T, Brink M, Winkler B, Matt P, Carrel TP, Eckstein FS, Lefkovits I, Grapow MT. Proteome analysis in cardiovascular pathophysiology using Dahl rat model. J Proteomics 74: 672–682, 2011. doi: 10.1016/j.jprot.2011.02.015. [DOI] [PubMed] [Google Scholar]
  • 59.Guay-Woodford LM. Bartter syndrome: unraveling the pathophysiologic enigma. Am J Med 105: 151–161, 1998. doi: 10.1016/S0002-9343(98)00196-X. [DOI] [PubMed] [Google Scholar]
  • 60.Guyton AC, Young DB, DeClue JW, Trippodo N, Hall JE. Fluid balance, renal function, and blood pressure. Clin Nephrol 4: 122–126, 1975. [PubMed] [Google Scholar]
  • 61.Guzik TJ, Hoch NE, Brown KA, McCann LA, Rahman A, Dikalov S, Goronzy J, Weyand C, Harrison DG. Role of the T cell in the genesis of angiotensin II induced hypertension and vascular dysfunction. J Exp Med 204: 2449–2460, 2007. doi: 10.1084/jem.20070657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hamlin LM, Fish WW. Physical properties of Tamm-Horsfall glycoprotein and its glycopolypeptide. Int J Pept Protein Res 10: 270–276, 1977. doi: 10.1111/j.1399-3011.1977.tb02797.x. [DOI] [PubMed] [Google Scholar]
  • 63.Hamlyn JM. Increased levels of a humoral digitalis-like factor in deoxycorticosterone acetate-induced hypertension in the pig. J Endocrinol 122: 409–420, 1989. doi: 10.1677/joe.0.1220409. [DOI] [PubMed] [Google Scholar]
  • 64.Hao S, Zhao H, Darzynkiewicz Z, Battula S, Ferreri NR. Expression and function of NFAT5 in medullary thick ascending limb (mTAL) cells. Am J Physiol Renal Physiol 296: F1494–F1503, 2009. doi: 10.1152/ajprenal.90436.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Haque MZ, Ares GR, Caceres PS, Ortiz PA. High salt differentially regulates surface NKCC2 expression in thick ascending limbs of Dahl salt-sensitive and salt-resistant rats. Am J Physiol Renal Physiol 300: F1096–F1104, 2011. doi: 10.1152/ajprenal.00600.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hart TC, Gorry MC, Hart PS, Woodard AS, Shihabi Z, Sandhu J, Shirts B, Xu L, Zhu H, Barmada MM, Bleyer AJ. Mutations of the UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet 39: 882–892, 2002. doi: 10.1136/jmg.39.12.882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.He FJ, MacGregor GA. Effect of modest salt reduction on blood pressure: a meta-analysis of randomized trials. Implications for public health. J Hum Hypertens 16: 761–770, 2002. doi: 10.1038/sj.jhh.1001459. [DOI] [PubMed] [Google Scholar]
  • 68.He FJ, MacGregor GA. Reducing population salt intake worldwide: from evidence to implementation. Prog Cardiovasc Dis 52: 363–382, 2010. doi: 10.1016/j.pcad.2009.12.006. [DOI] [PubMed] [Google Scholar]
  • 69.He G, Wang HR, Huang SK, Huang CL. Intersectin links WNK kinases to endocytosis of ROMK1. J Clin Invest 117: 1078–1087, 2007. doi: 10.1172/JCI30087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.He J, Ogden LG, Vupputuri S, Bazzano LA, Loria C, Whelton PK. Dietary sodium intake and subsequent risk of cardiovascular disease in overweight adults. JAMA 282: 2027–2034, 1999. doi: 10.1001/jama.282.21.2027. [DOI] [PubMed] [Google Scholar]
  • 71.He XR, Greenberg SG, Briggs JP, Schnermann J. Effects of furosemide and verapamil on the NaCl dependency of macula densa-mediated renin secretion. Hypertension 26: 137–142, 1995. doi: 10.1161/01.HYP.26.1.137. [DOI] [PubMed] [Google Scholar]
  • 72.Hebert SC, Andreoli TE. Control of NaCl transport in the thick ascending limb. Am J Physiol Renal Physiol 246: F745–F756, 1984. [DOI] [PubMed] [Google Scholar]
  • 73.Hebert SC, Andreoli TE. Ionic conductance pathways in the mouse medullary thick ascending limb of Henle. The paracellular pathway and electrogenic Cl- absorption. J Gen Physiol 87: 567–590, 1986. doi: 10.1085/jgp.87.4.567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hebert SC, Brown EM, Harris HW. Role of the Ca(2+)-sensing receptor in divalent mineral ion homeostasis. J Exp Biol 200: 295–302, 1997. [DOI] [PubMed] [Google Scholar]
  • 75.Hebert SC, Culpepper RM, Andreoli TE. NaCl transport in mouse medullary thick ascending limbs. II. ADH enhancement of transcellular NaCl cotransport; origin of transepithelial voltage. Am J Physiol Renal Physiol 241: F432–F442, 1981. [DOI] [PubMed] [Google Scholar]
  • 76.Herrera M, Hong NJ, Ortiz PA, Garvin JL. Endothelin-1 inhibits thick ascending limb transport via Akt-stimulated nitric oxide production. J Biol Chem 284: 1454–1460, 2009. doi: 10.1074/jbc.M804322200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Himmerkus N, Shan Q, Goerke B, Hou J, Goodenough DA, Bleich M. Salt and acid-base metabolism in claudin-16 knockdown mice: impact for the pathophysiology of FHHNC patients. Am J Physiol Renal Physiol 295: F1641–F1647, 2008. doi: 10.1152/ajprenal.90388.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hou J. Lecture: New light on the role of claudins in the kidney. Organogenesis 8: 1–9, 2012. doi: 10.4161/org.19808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hou J, Renigunta A, Gomes AS, Hou M, Paul DL, Waldegger S, Goodenough DA. Claudin-16 and claudin-19 interaction is required for their assembly into tight junctions and for renal reabsorption of magnesium. Proc Natl Acad Sci USA 106: 15350–15355, 2009. doi: 10.1073/pnas.0907724106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hoyer JR, Seiler MW. Pathophysiology of Tamm-Horsfall protein. Kidney Int 16: 279–289, 1979. doi: 10.1038/ki.1979.130. [DOI] [PubMed] [Google Scholar]
  • 81.Hoyer JR, Sisson SP, Vernier RL. Tamm-Horsfall glycoprotein: ultrastructural immunoperoxidase localization in rat kidney. Lab Invest 41: 168–173, 1979. [PubMed] [Google Scholar]
  • 82.Jacob N, Stohl W. Cytokine disturbances in systemic lupus erythematosus. Arthritis Res Ther 13: 228, 2011. doi: 10.1186/ar3349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Jevnikar AM, Brennan DC, Singer GG, Heng JE, Maslinski W, Wuthrich RP, Glimcher LH, Kelley VER. Stimulated kidney tubular epithelial cells express membrane associated and secreted TNF alpha. Kidney Int 40: 203–211, 1991. doi: 10.1038/ki.1991.201. [DOI] [PubMed] [Google Scholar]
  • 84.Ji W, Foo JN, O’Roak BJ, Zhao H, Larson MG, Simon DB, Newton-Cheh C, State MW, Levy D, Lifton RP. Rare independent mutations in renal salt handling genes contribute to blood pressure variation. Nat Genet 40: 592–599, 2008. doi: 10.1038/ng.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Jung J, Basile DP, Pratt JH. Sodium reabsorption in the thick ascending limb in relation to blood pressure: a clinical perspective. Hypertension 57: 873–879, 2011. doi: 10.1161/HYPERTENSIONAHA.108.120246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kahle KT, Macgregor GG, Wilson FH, Van Hoek AN, Brown D, Ardito T, Kashgarian M, Giebisch G, Hebert SC, Boulpaep EL, Lifton RP. Paracellular Cl- permeability is regulated by WNK4 kinase: insight into normal physiology and hypertension. Proc Natl Acad Sci USA 101: 14877–14882, 2004. doi: 10.1073/pnas.0406172101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kahle KT, Wilson FH, Leng Q, Lalioti MD, O’Connell AD, Dong K, Rapson AK, MacGregor GG, Giebisch G, Hebert SC, Lifton RP. WNK4 regulates the balance between renal NaCl reabsorption and K+ secretion. Nat Genet 35: 372–376, 2003. doi: 10.1038/ng1271. [DOI] [PubMed] [Google Scholar]
  • 88.Kay BK, Williamson MP, Sudol M. The importance of being proline: the interaction of proline-rich motifs in signaling proteins with their cognate domains. FASEB J 14: 231–241, 2000. [PubMed] [Google Scholar]
  • 89.Khanna D, McMahon M, Furst DE. Anti-tumor necrosis factor alpha therapy and heart failure: what have we learned and where do we go from here? Arthritis Rheum 50: 1040–1050, 2004. doi: 10.1002/art.20164. [DOI] [PubMed] [Google Scholar]
  • 90.Kitamura M. Endoplasmic reticulum stress and unfolded protein response in renal pathophysiology: Janus faces. Am J Physiol Renal Physiol 295: F323–F334, 2008. doi: 10.1152/ajprenal.00050.2008. [DOI] [PubMed] [Google Scholar]
  • 91.Lai KN, Leung JC, Chan LY, Saleem MA, Mathieson PW, Lai FM, Tang SC. Activation of podocytes by mesangial-derived TNF-α: glomerulo-podocytic communication in IgA nephropathy. Am J Physiol Renal Physiol 294: F945–F955, 2008. doi: 10.1152/ajprenal.00423.2007. [DOI] [PubMed] [Google Scholar]
  • 92.LaMarca BB, Cockrell K, Sullivan E, Bennett W, Granger JP. Role of endothelin in mediating tumor necrosis factor-induced hypertension in pregnant rats. Hypertension 46: 82–86, 2005. doi: 10.1161/01.HYP.0000169152.59854.36. [DOI] [PubMed] [Google Scholar]
  • 93.Lapointe JY, Laamarti A, Bell PD. Ionic transport in macula densa cells. Kidney Int Suppl 67: S58–S64, 1998. doi: 10.1046/j.1523-1755.1998.06712.x. [DOI] [PubMed] [Google Scholar]
  • 94.Levy D, Ehret GB, Rice K, Verwoert GC, Launer LJ, Dehghan A, Glazer NL, Morrison AC, Johnson AD, Aspelund T, Aulchenko Y, Lumley T, Köttgen A, Vasan RS, Rivadeneira F, Eiriksdottir G, Guo X, Arking DE, Mitchell GF, Mattace-Raso FU, Smith AV, Taylor K, Scharpf RB, Hwang SJ, Sijbrands EJ, Bis J, Harris TB, Ganesh SK, O’Donnell CJ, Hofman A, Rotter JI, Coresh J, Benjamin EJ, Uitterlinden AG, Heiss G, Fox CS, Witteman JC, Boerwinkle E, Wang TJ, Gudnason V, Larson MG, Chakravarti A, Psaty BM, van Duijn CM. Genome-wide association study of blood pressure and hypertension. Nat Genet 41: 677–687, 2009. doi: 10.1038/ng.384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Lhotta K. Uromodulin and chronic kidney disease. Kidney Blood Press Res 33: 393–398, 2010. doi: 10.1159/000320681. [DOI] [PubMed] [Google Scholar]
  • 96.Lhotta K, Gruber J, Sgonc R, Fend F, König P. Apoptosis of tubular epithelial cells in familial juvenile gouty nephropathy. Nephron 79: 340–344, 1998. doi: 10.1159/000045060. [DOI] [PubMed] [Google Scholar]
  • 97.Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hypertension. Cell 104: 545–556, 2001. doi: 10.1016/S0092-8674(01)00241-0. [DOI] [PubMed] [Google Scholar]
  • 98.Lim SS, Vos T, Flaxman AD, Danaei G, Shibuya K, Adair-Rohani H, Amann M, Anderson HR, Andrews KG, Aryee M, Atkinson C, Bacchus LJ, Bahalim AN, Balakrishnan K, Balmes J, Barker-Collo S, Baxter A, Bell ML, Blore JD, Blyth F, Bonner C, Borges G, Bourne R, Boussinesq M, Brauer M, Brooks P, Bruce NG, Brunekreef B, Bryan-Hancock C, Bucello C, Buchbinder R, Bull F, Burnett RT, Byers TE, Calabria B, Carapetis J, Carnahan E, Chafe Z, Charlson F, Chen H, Chen JS, Cheng AT, Child JC, Cohen A, Colson KE, Cowie BC, Darby S, Darling S, Davis A, Degenhardt L, Dentener F, Des Jarlais DC, Devries K, Dherani M, Ding EL, Dorsey ER, Driscoll T, Edmond K, Ali SE, Engell RE, Erwin PJ, Fahimi S, Falder G, Farzadfar F, Ferrari A, Finucane MM, Flaxman S, Fowkes FG, Freedman G, Freeman MK, Gakidou E, Ghosh S, Giovannucci E, Gmel G, Graham K, Grainger R, Grant B, Gunnell D, Gutierrez HR, Hall W, Hoek HW, Hogan A, Hosgood HD 3rd, Hoy D, Hu H, Hubbell BJ, Hutchings SJ, Ibeanusi SE, Jacklyn GL, Jasrasaria R, Jonas JB, Kan H, Kanis JA, Kassebaum N, Kawakami N, Khang YH, Khatibzadeh S, Khoo JP, Kok C, Laden F, Lalloo R, Lan Q, Lathlean T, Leasher JL, Leigh J, Li Y, Lin JK, Lipshultz SE, London S, Lozano R, Lu Y, Mak J, Malekzadeh R, Mallinger L, Marcenes W, March L, Marks R, Martin R, McGale P, McGrath J, Mehta S, Mensah GA, Merriman TR, Micha R, Michaud C, Mishra V, Mohd Hanafiah K, Mokdad AA, Morawska L, Mozaffarian D, Murphy T, Naghavi M, Neal B, Nelson PK, Nolla JM, Norman R, Olives C, Omer SB, Orchard J, Osborne R, Ostro B, Page A, Pandey KD, Parry CD, Passmore E, Patra J, Pearce N, Pelizzari PM, Petzold M, Phillips MR, Pope D, Pope CA 3rd, Powles J, Rao M, Razavi H, Rehfuess EA, Rehm JT, Ritz B, Rivara FP, Roberts T, Robinson C, Rodriguez-Portales JA, Romieu I, Room R, Rosenfeld LC, Roy A, Rushton L, Salomon JA, Sampson U, Sanchez-Riera L, Sanman E, Sapkota A, Seedat S, Shi P, Shield K, Shivakoti R, Singh GM, Sleet DA, Smith E, Smith KR, Stapelberg NJ, Steenland K, Stöckl H, Stovner LJ, Straif K, Straney L, Thurston GD, Tran JH, Van Dingenen R, van Donkelaar A, Veerman JL, Vijayakumar L, Weintraub R, Weissman MM, White RA, Whiteford H, Wiersma ST, Wilkinson JD, Williams HC, Williams W, Wilson N, Woolf AD, Yip P, Zielinski JM, Lopez AD, Murray CJ, Ezzati M, AlMazroa MA, Memish ZA. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380: 2224–2260, 2012. doi: 10.1016/S0140-6736(12)61766-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lin SH, Yu IS, Jiang ST, Lin SW, Chu P, Chen A, Sytwu HK, Sohara E, Uchida S, Sasaki S, Yang SS. Impaired phosphorylation of Na(+)-K(+)-2Cl(-) cotransporter by oxidative stress-responsive kinase-1 deficiency manifests hypotension and Bartter-like syndrome. Proc Natl Acad Sci USA 108: 17538–17543, 2011. doi: 10.1073/pnas.1107452108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Liu Y, El-Achkar TM, Wu XR. Tamm-Horsfall protein regulates circulating and renal cytokines by affecting glomerular filtration rate and acting as a urinary cytokine trap. J Biol Chem 287: 16365–16378, 2012. doi: 10.1074/jbc.M112.348243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Loreaux EL, Kaul B, Lorenz JN, Lingrel JB. Ouabain-Sensitive alpha1 Na,K-ATPase enhances natriuretic response to saline load. J Am Soc Nephrol 19: 1947–1954, 2008. doi: 10.1681/ASN.2008020174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lorenz JN, Schultheis PJ, Traynor T, Shull GE, Schnermann J. Micropuncture analysis of single-nephron function in NHE3-deficient mice. Am J Physiol Renal Physiol 277: F447–F453, 1999. [DOI] [PubMed] [Google Scholar]
  • 103.Lynn KL, Shenkin A, Marshall RD. Factors affecting excretion of human urinary Tamm-Horsfall glycoprotein. Clin Sci (Lond) 62: 21–26, 1982. doi: 10.1042/cs0620021. [DOI] [PubMed] [Google Scholar]
  • 104.Ma L, Liu Y, El-Achkar TM, Wu XR. Molecular and cellular effects of Tamm-Horsfall protein mutations and their rescue by chemical chaperones. J Biol Chem 287: 1290–1305, 2012. doi: 10.1074/jbc.M111.283036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.MacGregor GA, Fenton S, Alaghband-Zadeh J, Markandu ND, Roulston JE, de Wardener HE. An increase in a circulating inhibitor of Na+,K+-dependent ATPase: a possible link between salt intake and the development of essential hypertension. Clin Sci (Lond) 61, Suppl 7: 17s–20s, 1981. doi: 10.1042/cs061017s. [DOI] [PubMed] [Google Scholar]
  • 106.MacGregor GA, Markandu ND, Best FE, Elder DM, Cam JM, Sagnella GA, Squires M. Double-blind randomised crossover trial of moderate sodium restriction in essential hypertension. Lancet 1: 351–355, 1982. doi: 10.1016/S0140-6736(82)91389-7. [DOI] [PubMed] [Google Scholar]
  • 107.Macica CM, Escalante BA, Conners MS, Ferreri NR. TNF production by the medullary thick ascending limb of Henle’s loop. Kidney Int 46: 113–121, 1994. doi: 10.1038/ki.1994.250. [DOI] [PubMed] [Google Scholar]
  • 108.Majid DS. Tumor necrosis factor-α and kidney function: experimental findings in mice. Adv Exp Med Biol 691: 471–480, 2011. doi: 10.1007/978-1-4419-6612-4_48. [DOI] [PubMed] [Google Scholar]
  • 109.Maldonado-Cervantes MI, Galicia OG, Moreno-Jaime B, Zapata-Morales JR, Montoya-Contreras A, Bautista-Perez R, Martinez-Morales F. Autocrine modulation of glucose transporter SGLT2 by IL-6 and TNF-α in LLC-PK(1) cells. J Physiol Biochem 68: 411–420, 2012. doi: 10.1007/s13105-012-0153-3. [DOI] [PubMed] [Google Scholar]
  • 110.Mao G, Seebeck T, Schrenker D, Yu O. CYP709B3, a cytochrome P450 monooxygenase gene involved in salt tolerance in Arabidopsis thaliana. BMC Plant Biol 13: 169, 2013. doi: 10.1186/1471-2229-13-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mariappan N, Soorappan RN, Haque M, Sriramula S, Francis J. TNF-α-induced mitochondrial oxidative stress and cardiac dysfunction: restoration by superoxide dismutase mimetic Tempol. Am J Physiol Heart Circ Physiol 293: H2726–H2737, 2007. doi: 10.1152/ajpheart.00376.2007. [DOI] [PubMed] [Google Scholar]
  • 112.Marino MW, Dunn A, Grail D, Inglese M, Noguchi Y, Richards E, Jungbluth A, Wada H, Moore M, Williamson B, Basu S, Old LJ. Characterization of tumor necrosis factor-deficient mice. Proc Natl Acad Sci USA 94: 8093–8098, 1997. doi: 10.1073/pnas.94.15.8093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Markewitz BA, Michael JR, Kohan DE. Cytokine-induced expression of a nitric oxide synthase in rat renal tubule cells. J Clin Invest 91: 2138–2143, 1993. doi: 10.1172/JCI116439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Markossian S, Kreydiyyeh SI. TNF-alpha down-regulates the Na+-K+ ATPase and the Na+-K+-2Cl-cotransporter in the rat colon via PGE2. Cytokine 30: 319–327, 2005. doi: 10.1016/j.cyto.2004.11.009. [DOI] [PubMed] [Google Scholar]
  • 115.Matafora V, Zagato L, Ferrandi M, Molinari I, Zerbini G, Casamassima N, Lanzani C, Delli Carpini S, Trepiccione F, Manunta P, Bachi A, Capasso G. Quantitative proteomics reveals novel therapeutic and diagnostic markers in hypertension. BBA Clin 2: 79–87, 2014. doi: 10.1016/j.bbacli.2014.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.McCormick JA, Mutig K, Nelson JH, Saritas T, Hoorn EJ, Yang CL, Rogers S, Curry J, Delpire E, Bachmann S, Ellison DH. A SPAK isoform switch modulates renal salt transport and blood pressure. Cell Metab 14: 352–364, 2011. doi: 10.1016/j.cmet.2011.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Mederle K, Mutig K, Paliege A, Carota I, Bachmann S, Castrop H, Oppermann M. Loss of WNK3 is compensated for by the WNK1/SPAK axis in the kidney of the mouse. Am J Physiol Renal Physiol 304: F1198–F1209, 2013. doi: 10.1152/ajprenal.00288.2012. [DOI] [PubMed] [Google Scholar]
  • 118.Mo L, Huang HY, Zhu XH, Shapiro E, Hasty DL, Wu XR. Tamm-Horsfall protein is a critical renal defense factor protecting against calcium oxalate crystal formation. Kidney Int 66: 1159–1166, 2004. doi: 10.1111/j.1523-1755.2004.00867.x. [DOI] [PubMed] [Google Scholar]
  • 119.Mount DB. Thick ascending limb of the loop of Henle. Clin J Am Soc Nephrol 9: 1974–1986, 2014. doi: 10.2215/CJN.04480413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Muller DN, Shagdarsuren E, Park JK, Dechend R, Mervaala E, Hampich F, Fiebeler A, Ju X, Finckenberg P, Theuer J, Viedt C, Kreuzer J, Heidecke H, Haller H, Zenke M, Luft FC. Immunosuppressive treatment protects against angiotensin II-induced renal damage. Am J Pathol 161: 1679–1693, 2002. doi: 10.1016/S0002-9440(10)64445-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Mutig K, Kahl T, Saritas T, Godes M, Persson P, Bates J, Raffi H, Rampoldi L, Uchida S, Hille C, Dosche C, Kumar S, Castañeda-Bueno M, Gamba G, Bachmann S. Activation of the bumetanide-sensitive Na+,K+,2Cl- cotransporter (NKCC2) is facilitated by Tamm-Horsfall protein in a chloride-sensitive manner. J Biol Chem 286: 30200–30210, 2011. doi: 10.1074/jbc.M111.222968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Mutig K, Paliege A, Kahl T, Jöns T, Müller-Esterl W, Bachmann S. Vasopressin V2 receptor expression along rat, mouse, and human renal epithelia with focus on TAL. Am J Physiol Renal Physiol 293: F1166–F1177, 2007. doi: 10.1152/ajprenal.00196.2007. [DOI] [PubMed] [Google Scholar]
  • 123.Newton-Cheh C, Johnson T, Gateva V, Tobin MD, Bochud M, Coin L, Najjar SS, Zhao JH, Heath SC, Eyheramendy S, Papadakis K, Voight BF, Scott LJ, Zhang F, Farrall M, Tanaka T, Wallace C, Chambers JC, Khaw KT, Nilsson P, van der Harst P, Polidoro S, Grobbee DE, Onland-Moret NC, Bots ML, Wain LV, Elliott KS, Teumer A, Luan J, Lucas G, Kuusisto J, Burton PR, Hadley D, McArdle WL, Brown M, Dominiczak A, Newhouse SJ, Samani NJ, Webster J, Zeggini E, Beckmann JS, Bergmann S, Lim N, Song K, Vollenweider P, Waeber G, Waterworth DM, Yuan X, Groop L, Orho-Melander M, Allione A, Di Gregorio A, Guarrera S, Panico S, Ricceri F, Romanazzi V, Sacerdote C, Vineis P, Barroso I, Sandhu MS, Luben RN, Crawford GJ, Jousilahti P, Perola M, Boehnke M, Bonnycastle LL, Collins FS, Jackson AU, Mohlke KL, Stringham HM, Valle TT, Willer CJ, Bergman RN, Morken MA, Döring A, Gieger C, Illig T, Meitinger T, Org E, Pfeufer A, Wichmann HE, Kathiresan S, Marrugat J, O’Donnell CJ, Schwartz SM, Siscovick DS, Subirana I, Freimer NB, Hartikainen AL, McCarthy MI, O’Reilly PF, Peltonen L, Pouta A, de Jong PE, Snieder H, van Gilst WH, Clarke R, Goel A, Hamsten A, Peden JF, Seedorf U, Syvänen AC, Tognoni G, Lakatta EG, Sanna S, Scheet P, Schlessinger D, Scuteri A, Dörr M, Ernst F, Felix SB, Homuth G, Lorbeer R, Reffelmann T, Rettig R, Völker U, Galan P, Gut IG, Hercberg S, Lathrop GM, Zelenika D, Deloukas P, Soranzo N, Williams FM, Zhai G, Salomaa V, Laakso M, Elosua R, Forouhi NG, Völzke H, Uiterwaal CS, van der Schouw YT, Numans ME, Matullo G, Navis G, Berglund G, Bingham SA, Kooner JS, Connell JM, Bandinelli S, Ferrucci L, Watkins H, Spector TD, Tuomilehto J, Altshuler D, Strachan DP, Laan M, Meneton P, Wareham NJ, Uda M, Jarvelin MR, Mooser V, Melander O, Loos RJ, Elliott P, Abecasis GR, Caulfield M, Munroe PB; Wellcome Trust Case Control Consortium . Genome-wide association study identifies eight loci associated with blood pressure. Nat Genet 41: 666–676, 2009. doi: 10.1038/ng.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Nielsen S, Pallone T, Smith BL, Christensen EI, Agre P, Maunsbach AB. Aquaporin-1 water channels in short and long loop descending thin limbs and in descending vasa recta in rat kidney. Am J Physiol Renal Physiol 268: F1023–F1037, 1995. [DOI] [PubMed] [Google Scholar]
  • 125.Niiranen TJ, Vasan RS. Epidemiology of cardiovascular disease: recent novel outlooks on risk factors and clinical approaches. Expert Rev Cardiovasc Ther 14: 855–869, 2016. doi: 10.1080/14779072.2016.1176528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Nørregaard R, Jensen BL, Li C, Wang W, Knepper MA, Nielsen S, Frøkiaer J. COX-2 inhibition prevents downregulation of key renal water and sodium transport proteins in response to bilateral ureteral obstruction. Am J Physiol Renal Physiol 289: F322–F333, 2005. doi: 10.1152/ajprenal.00061.2005. [DOI] [PubMed] [Google Scholar]
  • 127.O’Brien WJ, Lingrel JB, Wallick ET. Ouabain binding kinetics of the rat alpha two and alpha three isoforms of the sodium-potassium adenosine triphosphate. Arch Biochem Biophys 310: 32–39, 1994. doi: 10.1006/abbi.1994.1136. [DOI] [PubMed] [Google Scholar]
  • 128.O’Connor PM. A radical approach to balancing the tides of tubular flow. Am J Physiol Renal Physiol 307: F917–F918, 2014. doi: 10.1152/ajprenal.00436.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ollier-Hartmann MP, Pouget-Abadie C, Bouillie J, Hartmann L. Variations of urinary Tamm-Horsfall protein in humans during the first thirty years of life. Nephron 38: 163–166, 1984. doi: 10.1159/000183300. [DOI] [PubMed] [Google Scholar]
  • 130.Pacheco-Alvarez D, Gamba G. WNK3 is a putative chloride-sensing kinase. Cell Physiol Biochem 28: 1123–1134, 2011. doi: 10.1159/000335848. [DOI] [PubMed] [Google Scholar]
  • 131.Padmanabhan S, Melander O, Johnson T, Di Blasio AM, Lee WK, Gentilini D, Hastie CE, Menni C, Monti MC, Delles C, Laing S, Corso B, Navis G, Kwakernaak AJ, van der Harst P, Bochud M, Maillard M, Burnier M, Hedner T, Kjeldsen S, Wahlstrand B, Sjögren M, Fava C, Montagnana M, Danese E, Torffvit O, Hedblad B, Snieder H, Connell JM, Brown M, Samani NJ, Farrall M, Cesana G, Mancia G, Signorini S, Grassi G, Eyheramendy S, Wichmann HE, Laan M, Strachan DP, Sever P, Shields DC, Stanton A, Vollenweider P, Teumer A, Völzke H, Rettig R, Newton-Cheh C, Arora P, Zhang F, Soranzo N, Spector TD, Lucas G, Kathiresan S, Siscovick DS, Luan J, Loos RJ, Wareham NJ, Penninx BW, Nolte IM, McBride M, Miller WH, Nicklin SA, Baker AH, Graham D, McDonald RA, Pell JP, Sattar N, Welsh P, Munroe P, Caulfield MJ, Zanchetti A, Dominiczak AF; Global BPgen Consortium . Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet 6: e1001177, 2010. doi: 10.1371/journal.pgen.1001177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Pak J, Pu Y, Zhang ZT, Hasty DL, Wu XR. Tamm-Horsfall protein binds to type 1 fimbriated Escherichia coli and prevents E. coli from binding to uroplakin Ia and Ib receptors. J Biol Chem 276: 9924–9930, 2001. doi: 10.1074/jbc.M008610200. [DOI] [PubMed] [Google Scholar]
  • 133.Palmer LG, Schnermann J. Integrated control of Na transport along the nephron. Clin J Am Soc Nephrol 10: 676–687, 2015. doi: 10.2215/CJN.12391213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Papathanasiou S, Rickelt S, Soriano ME, Schips TG, Maier HJ, Davos CH, Varela A, Kaklamanis L, Mann DL, Capetanaki Y. Tumor necrosis factor-α confers cardioprotection through ectopic expression of keratins K8 and K18. Nat Med 21: 1076–1084, 2015. doi: 10.1038/nm.3925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Payne JA, Xu JC, Haas M, Lytle CY, Ward D, Forbush B 3rd. Primary structure, functional expression, and chromosomal localization of the bumetanide-sensitive Na-K-Cl cotransporter in human colon. J Biol Chem 270: 17977–17985, 1995. doi: 10.1074/jbc.270.30.17977. [DOI] [PubMed] [Google Scholar]
  • 136.Ponce-Coria J, San-Cristobal P, Kahle KT, Vazquez N, Pacheco-Alvarez D, de Los Heros P, Juárez P, Muñoz E, Michel G, Bobadilla NA, Gimenez I, Lifton RP, Hebert SC, Gamba G. Regulation of NKCC2 by a chloride-sensing mechanism involving the WNK3 and SPAK kinases. Proc Natl Acad Sci USA 105: 8458–8463, 2008. doi: 10.1073/pnas.0802966105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Poulter NR, Khaw KT, Hopwood BE, Mugambi M, Peart WS, Rose G, Sever PS. The Kenyan Luo migration study: observations on the initiation of a rise in blood pressure. BMJ 300: 967–972, 1990. doi: 10.1136/bmj.300.6730.967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Prajczer S, Heidenreich U, Pfaller W, Kotanko P, Lhotta K, Jennings P. Evidence for a role of uromodulin in chronic kidney disease progression. Nephrol Dial Transplant 25: 1896–1903, 2010. doi: 10.1093/ndt/gfp748. [DOI] [PubMed] [Google Scholar]
  • 139.Pressler CA, Heinzinger J, Jeck N, Waldegger P, Pechmann U, Reinalter S, Konrad M, Beetz R, Seyberth HW, Waldegger S. Late-onset manifestation of antenatal Bartter syndrome as a result of residual function of the mutated renal Na+-K+-2Cl- co-transporter. J Am Soc Nephrol 17: 2136–2142, 2006. doi: 10.1681/ASN.2005101071. [DOI] [PubMed] [Google Scholar]
  • 140.Raffi HS, Bates JM Jr, Laszik Z, Kumar S. Tamm-Horsfall protein acts as a general host-defense factor against bacterial cystitis. Am J Nephrol 25: 570–578, 2005. doi: 10.1159/000088990. [DOI] [PubMed] [Google Scholar]
  • 141.Rafiqi FH, Zuber AM, Glover M, Richardson C, Fleming S, Jovanović S, Jovanović A, O’Shaughnessy KM, Alessi DR. Role of the WNK-activated SPAK kinase in regulating blood pressure. EMBO Mol Med 2: 63–75, 2010. doi: 10.1002/emmm.200900058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Rajendran L, Simons K. Lipid rafts and membrane dynamics. J Cell Sci 118: 1099–1102, 2005. doi: 10.1242/jcs.01681. [DOI] [PubMed] [Google Scholar]
  • 143.Rampoldi L, Caridi G, Santon D, Boaretto F, Bernascone I, Lamorte G, Tardanico R, Dagnino M, Colussi G, Scolari F, Ghiggeri GM, Amoroso A, Casari G. Allelism of MCKD, FJHN and GCKD caused by impairment of uromodulin export dynamics. Hum Mol Genet 12: 3369–3384, 2003. doi: 10.1093/hmg/ddg353. [DOI] [PubMed] [Google Scholar]
  • 144.Ramseyer VD, Garvin JL. Tumor necrosis factor-α: regulation of renal function and blood pressure. Am J Physiol Renal Physiol 304: F1231–F1242, 2013. doi: 10.1152/ajprenal.00557.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Renigunta A, Renigunta V, Saritas T, Decher N, Mutig K, Waldegger S. Tamm-Horsfall glycoprotein interacts with renal outer medullary potassium channel ROMK2 and regulates its function. J Biol Chem 286: 2224–2235, 2011. doi: 10.1074/jbc.M110.149880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Rezaiguia S, Garat C, Delclaux C, Meignan M, Fleury J, Legrand P, Matthay MA, Jayr C. Acute bacterial pneumonia in rats increases alveolar epithelial fluid clearance by a tumor necrosis factor-alpha-dependent mechanism. J Clin Invest 99: 325–335, 1997. doi: 10.1172/JCI119161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Rosa AC, Rattazzi L, Miglio G, Collino M, Fantozzi R. Angiotensin II induces tumor necrosis factor-α expression and release from cultured human podocytes. Inflamm Res 61: 311–317, 2012. doi: 10.1007/s00011-011-0412-8. [DOI] [PubMed] [Google Scholar]
  • 148.Sacks FM, Svetkey LP, Vollmer WM, Appel LJ, Bray GA, Harsha D, Obarzanek E, Conlin PR, Miller ER 3rd, Simons-Morton DG, Karanja N, Lin PH; DASH-Sodium Collaborative Research Group . Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. DASH-Sodium Collaborative Research Group. N Engl J Med 344: 3–10, 2001. doi: 10.1056/NEJM200101043440101. [DOI] [PubMed] [Google Scholar]
  • 149.Sahay M, Narayen G, Anuradha. Sodium transporters in kidney role in health and disease. J Assoc Physicians India 55: 135–139, 2007. [PubMed] [Google Scholar]
  • 150.Sands JM, Layton HE. Advances in understanding the urine-concentrating mechanism. Annu Rev Physiol 76: 387–409, 2014. doi: 10.1146/annurev-physiol-021113-170350. [DOI] [PubMed] [Google Scholar]
  • 151.Santambrogio S, Cattaneo A, Bernascone I, Schwend T, Jovine L, Bachi A, Rampoldi L. Urinary uromodulin carries an intact ZP domain generated by a conserved C-terminal proteolytic cleavage. Biochem Biophys Res Commun 370: 410–413, 2008. doi: 10.1016/j.bbrc.2008.03.099. [DOI] [PubMed] [Google Scholar]
  • 152.Schultheis PJ, Clarke LL, Meneton P, Miller ML, Soleimani M, Gawenis LR, Riddle TM, Duffy JJ, Doetschman T, Wang T, Giebisch G, Aronson PS, Lorenz JN, Shull GE. Renal and intestinal absorptive defects in mice lacking the NHE3 Na+/H+ exchanger. Nat Genet 19: 282–285, 1998. doi: 10.1038/969. [DOI] [PubMed] [Google Scholar]
  • 153.Seney FD Jr, Persson EG, Wright FS. Modification of tubuloglomerular feedback signal by dietary protein. Am J Physiol 252: F83–F90, 1987. [DOI] [PubMed] [Google Scholar]
  • 154.Shahid M, Francis J, Majid DS. Tumor necrosis factor-alpha induces renal vasoconstriction as well as natriuresis in mice. Am J Physiol Renal Physiol 295: F1836–F1844, 2008. doi: 10.1152/ajprenal.90297.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Shahid M, Francis J, Matrougui K, Majid DS. Involvement of tumor necrosis factor-α in natriuretic response to systemic infusion of nitric oxide synthase inhibitor in anesthetized mice. Am J Physiol Renal Physiol 299: F217–F224, 2010. doi: 10.1152/ajprenal.00611.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Simon DB, Bindra RS, Mansfield TA, Nelson-Williams C, Mendonca E, Stone R, Schurman S, Nayir A, Alpay H, Bakkaloglu A, Rodriguez-Soriano J, Morales JM, Sanjad SA, Taylor CM, Pilz D, Brem A, Trachtman H, Griswold W, Richard GA, John E, Lifton RP. Mutations in the chloride channel gene, CLCNKB, cause Bartter’s syndrome type III. Nat Genet 17: 171–178, 1997. doi: 10.1038/ng1097-171. [DOI] [PubMed] [Google Scholar]
  • 157.Simon DB, Karet FE, Hamdan JM, DiPietro A, Sanjad SA, Lifton RP. Bartter’s syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotransporter NKCC2. Nat Genet 13: 183–188, 1996. doi: 10.1038/ng0696-183. [DOI] [PubMed] [Google Scholar]
  • 158.Simon DB, Karet FE, Rodriguez-Soriano J, Hamdan JH, DiPietro A, Trachtman H, Sanjad SA, Lifton RP. Genetic heterogeneity of Bartter’s syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet 14: 152–156, 1996. doi: 10.1038/ng1096-152. [DOI] [PubMed] [Google Scholar]
  • 159.Simon DB, Nelson-Williams C, Bia MJ, Ellison D, Karet FE, Molina AM, Vaara I, Iwata F, Cushner HM, Koolen M, Gainza FJ, Gitleman HJ, Lifton RP. Gitelman’s variant of Bartter’s syndrome, inherited hypokalaemic alkalosis, is caused by mutations in the thiazide-sensitive Na-Cl cotransporter. Nat Genet 12: 24–30, 1996. doi: 10.1038/ng0196-24. [DOI] [PubMed] [Google Scholar]
  • 160.Smagula RM, Van Halbeek H, Decker JM, Muchmore AV, Moody CE, Sherblom AP. Pregnancy-associated changes in oligomannose oligosaccharides of human and bovine uromodulin (Tamm-Horsfall glycoprotein). Glycoconj J 7: 609–624, 1990. doi: 10.1007/BF01189081. [DOI] [PubMed] [Google Scholar]
  • 161.Sobel JD, Kaye D. Reduced uromucoid excretion in the elderly. J Infect Dis 152: 653, 1985. doi: 10.1093/infdis/152.3.653. [DOI] [PubMed] [Google Scholar]
  • 162.Sonalker PA, Tofovic SP, Jackson EK. Cellular distribution of the renal bumetanide-sensitive Na-K-2Cl cotransporter BSC-1 in the inner stripe of the outer medulla during the development of hypertension in the spontaneously hypertensive rat. Clin Exp Pharmacol Physiol 34: 1307–1312, 2007. doi: 10.1111/j.1440-1681.2007.04747.x. [DOI] [PubMed] [Google Scholar]
  • 163.Sonalker PA, Tofovic SP, Jackson EK. Increased expression of the sodium transporter BSC-1 in spontaneously hypertensive rats. J Pharmacol Exp Ther 311: 1052–1061, 2004. doi: 10.1124/jpet.104.071209. [DOI] [PubMed] [Google Scholar]
  • 164.Sriramula S, Haque M, Majid DS, Francis J. Involvement of tumor necrosis factor-alpha in angiotensin II-mediated effects on salt appetite, hypertension, and cardiac hypertrophy. Hypertension 51: 1345–1351, 2008. doi: 10.1161/HYPERTENSIONAHA.107.102152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Stevenson FK, Cleave AJ, Kent PW. The effect of ions on the viscometric and ultracentrifugal behaviour of Tamm-Horsfall glycoprotein. Biochim Biophys Acta 236: 59–66, 1971. doi: 10.1016/0005-2795(71)90149-8. [DOI] [PubMed] [Google Scholar]
  • 166.Stibůrková B, Majewski J, Sebesta I, Zhang W, Ott J, Kmoch S. Familial juvenile hyperuricemic nephropathy: localization of the gene on chromosome 16p11.2-and evidence for genetic heterogeneity. Am J Hum Genet 66: 1989–1994, 2000. doi: 10.1086/302936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Thouvenot L, Deleu C, Berardocco S, Haury J, Thiébaut G. Characterization of the salt stress vulnerability of three invasive freshwater plant species using a metabolic profiling approach. J Plant Physiol 175: 113–121, 2015. doi: 10.1016/j.jplph.2014.11.007. [DOI] [PubMed] [Google Scholar]
  • 168.Torffvit O, Melander O, Hultén UL. Urinary excretion rate of Tamm-Horsfall protein is related to salt intake in humans. Nephron Physiol 97: 31–36, 2004. doi: 10.1159/000077600. [DOI] [PubMed] [Google Scholar]
  • 169.Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther 117: 244–279, 2008. doi: 10.1016/j.pharmthera.2007.10.001. [DOI] [PubMed] [Google Scholar]
  • 170.Trudu M, Janas S, Lanzani C, Debaix H, Schaeffer C, Ikehata M, Citterio L, Demaretz S, Trevisani F, Ristagno G, Glaudemans B, Laghmani K, Dell’Antonio G; Swiss Kidney Project on Genes in Hypertension (SKIPOGH) team, Loffing J, Rastaldi MP, Manunta P, Devuyst O, Rampoldi L. Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Med 19: 1655–1660, 2013. doi: 10.1038/nm.3384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.van Lanschot JJ, Mealy K, Jacobs DO, Evans DA, Wilmore DW. Splenectomy attenuates the inappropriate diuresis associated with tumor necrosis factor administration. Surg Gynecol Obstet 172: 293–297, 1991. [PubMed] [Google Scholar]
  • 172.van Rooijen JJ, Kamerling JP, Vliegenthart JF. Sulfated di-, tri- and tetraantennary N-glycans in human Tamm-Horsfall glycoprotein. Eur J Biochem 256: 471–487, 1998. doi: 10.1046/j.1432-1327.1998.2560471.x. [DOI] [PubMed] [Google Scholar]
  • 173.Van Zee KJ, Stackpole SA, Montegut WJ, Rogy MA, Calvano SE, Hsu KC, Chao M, Meschter CL, Loetscher H, Stüber D, Ettlin U, Wipf B, Lesslauer W, Lowry SF, Moldawer LL. A human tumor necrosis factor (TNF) alpha mutant that binds exclusively to the p55 TNF receptor produces toxicity in the baboon. J Exp Med 179: 1185–1191, 1994. doi: 10.1084/jem.179.4.1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Vargas-Poussou R, Feldmann D, Vollmer M, Konrad M, Kelly L, van den Heuvel LP, Tebourbi L, Brandis M, Karolyi L, Hebert SC, Lemmink HH, Deschênes G, Hildebrandt F, Seyberth HW, Guay-Woodford LM, Knoers NV, Antignac C. Novel molecular variants of the Na-K-2Cl cotransporter gene are responsible for antenatal Bartter syndrome. Am J Hum Genet 62: 1332–1340, 1998. doi: 10.1086/301872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Venegas-Pont M, Manigrasso MB, Grifoni SC, LaMarca BB, Maric C, Racusen LC, Glover PH, Jones AV, Drummond HA, Ryan MJ. Tumor necrosis factor-alpha antagonist etanercept decreases blood pressure and protects the kidney in a mouse model of systemic lupus erythematosus. Hypertension 56: 643–649, 2010. doi: 10.1161/HYPERTENSIONAHA.110.157685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Vinciguerra M, Hasler U, Mordasini D, Roussel M, Capovilla M, Ogier-Denis E, Vandewalle A, Martin PY, Feraille E. Cytokines and sodium induce protein kinase A-dependent cell-surface Na,K-ATPase recruitment via dissociation of NF-kappaB/IkappaB/protein kinase A catalytic subunit complex in collecting duct principal cells. J Am Soc Nephrol 16: 2576–2585, 2005. doi: 10.1681/ASN.2005040448. [DOI] [PubMed] [Google Scholar]
  • 177.Vylet’al P, Kublová M, Kalbácová M, Hodanová K, Baresová V, Stibůrková B, Sikora J, Hůlková H, Zivný J, Majewski J, Simmonds A, Fryns JP, Venkat-Raman G, Elleder M, Kmoch S. Alterations of uromodulin biology: a common denominator of the genetically heterogeneous FJHN/MCKD syndrome. Kidney Int 70: 1155–1169, 2006. doi: 10.1038/sj.ki.5001728. [DOI] [PubMed] [Google Scholar]
  • 178.Waldegger S, Jeck N, Barth P, Peters M, Vitzthum H, Wolf K, Kurtz A, Konrad M, Seyberth HW. Barttin increases surface expression and changes current properties of ClC-K channels. Pflugers Arch 444: 411–418, 2002. doi: 10.1007/s00424-002-0819-8. [DOI] [PubMed] [Google Scholar]
  • 179.Weinberger MH, Miller JZ, Luft FC, Grim CE, Fineberg NS. Definitions and characteristics of sodium sensitivity and blood pressure resistance. Hypertension 8: II127–II134, 1986. doi: 10.1161/01.HYP.8.6_Pt_2.II127. [DOI] [PubMed] [Google Scholar]
  • 180.Whelton PK, Appel LJ, Espeland MA, Applegate WB, Ettinger WH Jr, Kostis JB, Kumanyika S, Lacy CR, Johnson KC, Folmar S, Cutler JA. Sodium reduction and weight loss in the treatment of hypertension in older persons: a randomized controlled trial of nonpharmacologic interventions in the elderly (TONE). TONE Collaborative Research Group. JAMA 279: 839–846, 1998. doi: 10.1001/jama.279.11.839. [DOI] [PubMed] [Google Scholar]
  • 181.Whelton PK, Hebert PR, Cutler J, Applegate WB, Eberlein KA, Klag MJ, Keough ME, Hamill S, Borhani NO, Hollis J, Oberman A. Baseline characteristics of participants in phase I of the Trials of Hypertension Prevention. Ann Epidemiol 2: 295–310, 1992. doi: 10.1016/1047-2797(92)90062-U. [DOI] [PubMed] [Google Scholar]
  • 182.Wilson FH, Disse-Nicodème S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, Lifton RP. Human hypertension caused by mutations in WNK kinases. Science 293: 1107–1112, 2001. doi: 10.1126/science.1062844. [DOI] [PubMed] [Google Scholar]
  • 183.Wolf MT, Beck BB, Zaucke F, Kunze A, Misselwitz J, Ruley J, Ronda T, Fischer A, Eifinger F, Licht C, Otto E, Hoppe B, Hildebrandt F. The Uromodulin C744G mutation causes MCKD2 and FJHN in children and adults and may be due to a possible founder effect. Kidney Int 71: 574–581, 2007. doi: 10.1038/sj.ki.5002089. [DOI] [PubMed] [Google Scholar]
  • 184.Wong M, Ziring D, Korin Y, Desai S, Kim S, Lin J, Gjertson D, Braun J, Reed E, Singh RR. TNFalpha blockade in human diseases: mechanisms and future directions. Clin Immunol 126: 121–136, 2008. doi: 10.1016/j.clim.2007.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Wright FS, Schnermann J. Interference with feedback control of glomerular filtration rate by furosemide, triflocin, and cyanide. J Clin Invest 53: 1695–1708, 1974. doi: 10.1172/JCI107721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Xu H, Chen H, Dong J, Li J, Chen R, Uno JK, Ghishan FK. Tumor necrosis factor-α downregulates intestinal NHE8 expression by reducing basal promoter activity. Am J Physiol Cell Physiol 296: C489–C497, 2009. doi: 10.1152/ajpcell.00482.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Yamauchi K, Rai T, Kobayashi K, Sohara E, Suzuki T, Itoh T, Suda S, Hayama A, Sasaki S, Uchida S. Disease-causing mutant WNK4 increases paracellular chloride permeability and phosphorylates claudins. Proc Natl Acad Sci USA 101: 4690–4694, 2004. doi: 10.1073/pnas.0306924101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Yang CL, Angell J, Mitchell R, Ellison DH. WNK kinases regulate thiazide-sensitive Na-Cl cotransport. J Clin Invest 111: 1039–1045, 2003. doi: 10.1172/JCI17443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Yang H, Wu C, Zhao S, Guo J. Identification and characterization of D8C, a novel domain present in liver-specific LZP, uromodulin and glycoprotein 2, mutated in familial juvenile hyperuricaemic nephropathy. FEBS Lett 578: 236–238, 2004. doi: 10.1016/j.febslet.2004.10.092. [DOI] [PubMed] [Google Scholar]
  • 190.Yang SS, Lo YF, Wu CC, Lin SW, Yeh CJ, Chu P, Sytwu HK, Uchida S, Sasaki S, Lin SH. SPAK-knockout mice manifest Gitelman syndrome and impaired vasoconstriction. J Am Soc Nephrol 21: 1868–1877, 2010. doi: 10.1681/ASN.2009121295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Zaucke F, Boehnlein JM, Steffens S, Polishchuk RS, Rampoldi L, Fischer A, Pasch A, Boehm CW, Baasner A, Attanasio M, Hoppe B, Hopfer H, Beck BB, Sayer JA, Hildebrandt F, Wolf MT. Uromodulin is expressed in renal primary cilia and UMOD mutations result in decreased ciliary uromodulin expression. Hum Mol Genet 19: 1985–1997, 2010. doi: 10.1093/hmg/ddq077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Zhou X, Forrest MJ, Sharif-Rodriguez W, Forrest G, Szeto D, Urosevic-Price O, Zhu Y, Stevenson AS, Zhou Y, Stribling S, Dajee M, Walsh SP, Pasternak A, Sullivan KA. Chronic inhibition of renal outer medullary potassium channel not only prevented but also reversed development of hypertension and end-organ damage in Dahl salt-sensitive rats. Hypertension 6: 332-338, 2017. doi: 10.1161/HYPERTENSIONAHA.116.08358. [DOI] [PubMed] [Google Scholar]
  • 193.Zhu LJ, Yang X, Yu XQ. Anti-TNF-alpha therapies in systemic lupus erythematosus. J Biomed Biotechnol 2010: 465898, 2010. doi: 10.1155/2010/465898. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Physiological Genomics are provided here courtesy of American Physiological Society

RESOURCES