The sodium chloride cotransporter (NCC) has been identified as a key molecule regulating potassium balance. The mechanisms of NCC regulation during low extracellular potassium concentrations have been studied in vitro. These studies have shown that hyperpolarization increased chloride efflux, leading to the activation of chloride-sensitive with-no-lysine kinase (WNK) kinases and their downstream molecules, including STE20/SPS1-related proline/alanine-rich kinase (SPAK) and NCC. However, this mechanism was not studied in vivo. Previously, we developed the barttin hypomorphic mouse (Bsndneo/neo mice), expressing very low levels of barttin and ClC-K channels, because barttin is an essential β-subunit of ClC-K. In contrast with Bsnd−/− mice, Bsndneo/neo mice survived to adulthood. In Bsndneo/neo mice, SPAK and NCC activation after consuming a low-potassium diet was clearly impaired compared with that in wild-type (WT) mice. In ex vivo kidney slice experiment, the increase in pNCC and SPAK in low-potassium medium was also impaired in Bsndneo/neo mice. Furthermore, increased blood pressure was observed in WT mice fed a high-salt and low-potassium diet, which was not evident in Bsndneo/neo mice. Thus, our study provides in vivo evidence that, in response to a low-potassium diet, ClC-K and barttin play important roles in the activation of the WNK4-SPAK-NCC cascade and blood pressure regulation.

Hypertension is a major worldwide public health problem associated with a variety of complications including stroke, heart failure, and kidney failure. Diets play a strong contributory role in blood pressure. Dietary potassium (K+) intake is highly related to blood pressure and mortality, and recent studies have shown that K+ intake was inversely related to blood pressure [13]. The sodium (Na+) chloride (Cl) cotransporter (NCC) expressed in the distal convoluted tubules (DCT) in the kidney plays an important role in the regulation of urinary K+ excretion, as well as in blood pressure regulation by NaCl reabsorption. In previous animal studies, a low-K+ diet increased the total amount and the phosphorylation of NCC [49] and elevated blood pressure [4,10]. This elevation of blood pressure with a low-K+ diet was dependent upon NCC, because NCC−/− mice did not show an elevation of blood pressure when under a low-K+ diet [4]. Although NCC itself does not directly transport K+, the amount of NaCl reabsorption in the DCT affects the delivery of Na+ to the downstream cortical collecting ducts where K+ is excreted, based upon Na+ reabsorption via epithelial Na+ channels. The notion that the NCC is important for regulating K+ excretion is also supported by the facts that two genetic diseases, Gitelman syndrome (caused by the loss-of-function of NCC) and pseudohypoaldosteronism type II (caused by the gain-of-function of NCC) show hypokalemia and hyperkalemia, respectively [11,12].

With-no-lysine kinase (WNK) kinases phosphorylate the STE20/SPS1-44 related proline-alanine-rich protein kinase (SPAK) and the related oxidative stress-related kinase 1 (OSR1), which directly activate NCC [13,14]. It has been proposed that the phosphorylation of NCC with a low-K+ diet is dependent upon WNK4 and OSR1/SPAK kinases. Low K+ activates not only NCC but also WNKs and SPAK [4,7,8,10]. Furthermore, WNK4−/− and SPAK−/− mice showed either no increase or only a mild increase, in pNCC in response to a low-K+ diet, respectively [4,79,15]. Disrupting both SPAK and OSR1 almost completely ablated the response of pNCC to dietary K+ restriction [4,9]. The mechanism by which low K+ activates WNKs was previously investigated in cultured cells and postulated as follows: a decrease in extracellular K+ concentration ([K+]ex) affects the membrane potential of cells, thereby decreasing intracellular Cl concentration ([Cl]i) with Cl efflux via a Cl channel [4]. Since WNK kinases are regulated by [Cl]i [15,16], this decrease in [Cl]i by low [K+]ex activates WNK signaling. As for the molecular identity of the channel responsible for the Cl efflux in DCT, ClC-Kb (a human homolog of ClC-K2) chloride channel has been postulated due to its localization in the DCT [17,18]. Additional genetic evidence showed that Barter syndrome type III caused by the loss-of-function mutations in the CLCKB gene (which codes ClC-Kb) had similar phenotypes to Gitelman syndrome [19,20]. Further, a recent study using patch clamp analysis of tubules isolated from the ClC-K2 knockout mouse reported that ClC-K2 is the predominant Cl channel on the basolateral membrane of DCT cells [21]. Indeed, Terker et al. [4] prepared HEK293 cells overexpressing wild-type (WT) or loss-of-function mutant ClC-Kb, and they found that low-K+-induced phosphorylation of NCC was attenuated by the mutation suggesting the potential role of ClC-Kb in regulating [Cl]i in the DCT cells. However, these data were obtained only from in vitro cultured cell studies. To clarify the contribution of ClC-K2 in the mechanism of NCC phosphorylation in response to K+ restriction in vivo, we performed animal studies. Previously, we generated Bsndneo(R8L)/neo(R8L) (Bsndneo/neo) mice, which are hypomorphic of a disease-causing mutant barttin (R8L barttin) [22]. Barttin (coded by the Bsnd gene) is an essential β-subunit for both ClC-Ka/1 and ClC-Kb/2 channels [23,24]. Since barttin is crucial for ClC-K membrane localization and stability, the genetic ablation of barttin resulted in a ClC-K knockout condition [25]. We used Bsndneo/neo mice to investigate the role of ClC-K and barttin in NCC activation by low-K+ diet.

Animal experiments

All experiments were performed in accordance with the guidelines for animal research of Tokyo Medical and Dental University, and the protocol was approved by The Animal Care and Use Committee of Tokyo Medical and Dental University. Studies were performed on Bsndneo/neo mice (C57BL6 background) as a loss-of-function model for ClC-K [22]. Littermates WT mice or C57BL/6 mice (Japan SLC, Inc., Hamamatsu, Japan) were used for WT control mice. We used mixed sex, 20–30 g body weight, 10–16 weeks old mice. These parameters were matched in each group. A high-salt and normal-K+ diet (6% NaCl, 1% K+, a high-salt and normal-K (HSNK) diet) and a high-salt and low-K+ diet (6% NaCl, 0.01% K+, a high-salt and low-K (HSLK) diet) were prepared by adding NaCl and KCl (or sucrose for adjustment of volume) to a K+-deficient diet which was modified from AIN-76 diet (Oriental Yeast Co., Tokyo, Japan). Blood was collected from the retro-orbital venous plexus under anesthesia. Blood data were analyzed by iSTAT EC8+ (Abbott, Inc., Abbott Park, IL). Serum aldosterone levels were measured by the SRL clinical laboratory service (Tokyo, Japan). Noninvasive systolic blood pressures were measured by a programmable tail-cuff sphygmomanometer (MK-2000A, Muromachi Kikai Co., Tokyo, Japan) with the investigator blinded for the treatment groups. Systolic blood pressure of mice consuming an HSNK diet was measured after a 2-day period of acclimation to the instrument. Blood pressure was recorded again 2 days after switching diets from an HSNK diet to an HSLK diet. The changes in blood pressure after the switching diets were compared between WT mice and Bsndneo/neo mice with unpaired t test. Each blood pressure data point was calculated as the mean of ~20 sequential measurements.

Western blotting and immunofluorescence

Immunoblotting and immunofluorescence were performed as previously described [7]. The detail of the method is described in Supplementary information. Briefly, kidneys were homogenized and then the homogenates were centrifuged to separate entire kidney samples without the nuclear fraction, as either whole kidney lysates (600 g, supernatant) and crude membrane fraction (17000 g, pellet). The membrane fractions were used for the analysis of barttin, ClC-K, pNCC and total NCC (tNCC). The whole kidney lysates were used for the WNK4, pSPAK and tSPAK detection. The relative intensities of immunoblot bands were analyzed and quantitated using ImageJ software (National Institutes of Health, Bethesda, MD). For the experiment of correlation between pNCC and plasma K+ concentration, each value of pNCC/tNCC from HSLK groups were compared with their internal control group on an HSNK diet. pNCC/tNCC average of the internal control group was set to 1.

For immunofluorescence, kidneys were fixed by perfusion through the left ventricle with periodate lysine (0.2 M) and paraformaldehyde (2%) in PBS. Tissue samples were soaked for several hours in 20% sucrose in PBS, embedded in Tissue Tek O.C.T. Compound (Sakura Finetechnical Co., Ltd, Tokyo, Japan), and frozen in liquid nitrogen.

Antibodies are listed in Supplementary Table S1. The primary antibodies used in the present study were as follows: rabbit anti-WNK4 [7], rabbit anti-pSPAK (Ser383, kindly gifted by Dr S.S. Yang, National Defense Medical Center) [26], rabbit anti-total SPAK (Cell Signaling Technology, Inc., Danvers, MA, #2281), rabbit anti-pNCC (Ser71) [27], rabbit anti-tNCC [7], guinea pig anti-tNCC [7,22], rabbit anti-ClC-K (kindly gifted by Dr T.J. Jentsch, Max-Delbrck-Centrüm für Molekulare Medizin) [25], and rabbit anti-actin antibody (Cytoskeleton, Inc. Denver, CO. AAN01, Lot 121). We raised a new anti-pSPAK antibody in rabbit against the synthetic peptide, which recognizes the same phosphorylation site as the previous one [26]. The band of WNK4, pSPAK and total SPAK had been confirmed by using the knockout mice (Supplementary Figure S1) [7]. We used two different pSPAK antibodies because we had used up the previous one. As for secondary antibodies, alkaline phosphatase-conjugated anti-IgG antibodies (Promega Corporation, Fitchburg, WI) and Alexa 488 or 546 dye-labeled antibodies (Molecular Probes, Inc., Eugene, OR) were used for Western blotting and immunofluorescence, respectively. Western Blue® (Promega Corporation, Fitchburg, WI) was used to detect the signals and immunofluorescence images were acquired using the LSM510 Meta confocal microscope (Carl Zeiss, Oberkochen, Germany). The linearity of protein detection for each antibody was confirmed in Supplementary Figures S2 and S3.

Ex vivo kidney slice experiment

Kidney slices were prepared as described previously [28]. Kidney slices of less than 0.5 mm were cut using a microslicer (Natume Seisakusho Co., Ltd, Tokyo, Japan) on ice-cold Hank’s buffer medium (110 mM NaCl, 3 mM KCl, 1.2 mM MgSO4, 1.8 mM CaCl2, 4 mM Na acetate, 1 mM Na citrate, 6 mM d-glucose, 6 mM l-alanine, 1 mM NaH2PO4, 3 mM Na2HPO4, 25 mM NaHCO3). All the sliced kidneys were incubated in the Hank’s buffer medium at room temperature for 20 min. After the recovery in the Hank’s buffer medium for 20 min, the slices from the same kidney were separated in normal-K+ (4 mEq/l) or low-K+ (2 mEq/l) medium, then incubated for 30 min at 28°C. Different K+ concentration medium was prepared by using KCl or choline chloride to maintain the same Cl concentration in each medium. During the experiments, all solutions were continuously bubbled with 95% O2 and 5% CO2. After the incubation, slices were snap-frozen in liquid nitrogen and processed for immunoblotting as same as for whole kidney samples described above and Supplementary information. After Western blotting, the fold change of the band density in a low-K+ medium to the one in a normal medium were analyzed.

Statistical analysis

Data were presented as the means ± S.E.M. Two-way ANOVA and Bonferroni’s post test was used to compare the multiple groups while the ttest was used to compare two groups. For all analyses, a P-value <0.05 was considered to be statistically significant. For correlation analyzes, Pearson’s test was performed.

Bsndneo/neo mice showed very low amount of barttin and ClC-K and no change in NCC phosphorylation

Previously, we generated Bsndneo/neo mice which contained a Neo-cassette and expressed a disease-causing R8L mutant of barttin [22]. Without deleting the Neo-cassette, the transcription of the Bsnd gene was significantly reduced. Unlike Bsnd−/− mice [25], Bsndneo/neo mice could thrive to adulthood. In the present study, we confirmed that Bsndneo/neo mice expressed only minimal amounts of barttin and ClC-K in their kidneys (Figure 1A). We performed immunofluorescence to investigate the amount and localization of barttin in DCT in Bsndneo/neo mice. In Bsndneo/neomice, the staining of barttin was reduced in the DCT cells. Furthermore, basolateral staining of the R8L barttin mutant was clearly impaired (Figure 1B) as we previously reported [22]. These results suggested that ClC-K function in DCT cells was significantly reduced in Bsndneo/neo mice.

Baseline characteristics of Bsndneo/neo mice

Figure 1
Baseline characteristics of Bsndneo/neo mice

(A) Representative immunoblots and densitometric analysis of barttin and ClC-K in the mice consuming a normal diet. The expression of barttin and ClC-K was quite low in the kidneys of Bsndneo/neo mice; n=4. **P<0.005 by unpaired ttest. (B) Representative immunofluorescences in the DCTs of WT mice and Bsndneo/neo mice. In Bsndneo/neo mice, barttin staining (red) was low and the localization upon the basolateral membrane was impaired. NCC (green) was stained as a marker of DCTs. Scale bars indicate 50 µm. (C) Representative immunoblots and densitometric analysis of NCC; n=4. Abbreviation: n.s., not significant.

Figure 1
Baseline characteristics of Bsndneo/neo mice

(A) Representative immunoblots and densitometric analysis of barttin and ClC-K in the mice consuming a normal diet. The expression of barttin and ClC-K was quite low in the kidneys of Bsndneo/neo mice; n=4. **P<0.005 by unpaired ttest. (B) Representative immunofluorescences in the DCTs of WT mice and Bsndneo/neo mice. In Bsndneo/neo mice, barttin staining (red) was low and the localization upon the basolateral membrane was impaired. NCC (green) was stained as a marker of DCTs. Scale bars indicate 50 µm. (C) Representative immunoblots and densitometric analysis of NCC; n=4. Abbreviation: n.s., not significant.

Close modal

We performed immunoblotting to determine whether Bsndneo/neo mice on a normal diet have alterations in the NCC expression. No significant differences were observed in pNCC and tNCC between Bsndneo/neo mice and WT mice (Figure 1C).

The increase in NCC and SPAK phosphorylation in response to a low-K+ diet was significantly impaired in Bsndneo/neo mice

To investigate the change of pNCC to K+ restriction, both WT mice and the Bsndneo/neo mice were fed an HSNK diet or an HSLK diet same as a previous study [4]. Although general appearance of the mice consuming an HSLK diet were normal, Bsndneo/neo mice started to lose weight after 3 days on an HSLK diet, which is probably due to potassium deficiency. Thus, we fed an HSLK diet or an HSNK diet, to the mice for only 2 days to avoid non-specific effects due to intolerance to HSLK diet. Bsndneo/neo mice showed hypokalemia, metabolic alkalosis with low Cl and higher aldosterone levels when fed an HSNK diet (Table 1). In both WT mice and Bsndneo/neo mice, plasma K+ levels were significantly reduced with an HSLK diet. There was no significant difference in the aldosterone level between Bsndneo/neo and WT mice when compared with mice fed upon an HSLK diet.

Table 1
Blood data from mice receiving an HSNK diet or an HSLK diet for 2 days
WTBsndneo/neo
HSNKnHSLKnHSNKnHSLKn
Na 148 ± 1 147 ± 2 147 ± 2 146 ± 3 
4.7 ± 0.2 3.7 ± 0.2* 3.4 ± 0.1 2.7 ± 0.2* 
Cl 118 ± 1 114 ± 1 108 ± 2 107 ± 2 
tCO2 20 ± 1 20 ± 1 29 ± 1 31 ± 2 
BUN 21 ± 2 17 ± 1 25 ± 2 25 ± 1 
Ht 42 ± 1 43 ± 2 48 ± 2 46 ± 2 
Aldosterone 163 ± 39 98 ± 48 418 ± 81 112 ± 66* 
WTBsndneo/neo
HSNKnHSLKnHSNKnHSLKn
Na 148 ± 1 147 ± 2 147 ± 2 146 ± 3 
4.7 ± 0.2 3.7 ± 0.2* 3.4 ± 0.1 2.7 ± 0.2* 
Cl 118 ± 1 114 ± 1 108 ± 2 107 ± 2 
tCO2 20 ± 1 20 ± 1 29 ± 1 31 ± 2 
BUN 21 ± 2 17 ± 1 25 ± 2 25 ± 1 
Ht 42 ± 1 43 ± 2 48 ± 2 46 ± 2 
Aldosterone 163 ± 39 98 ± 48 418 ± 81 112 ± 66* 

Values represent means ± S.E.M. Two-way ANOVA and Bonferroni’s test were performed to analyze statistical differences. *P<0.05, HSNK compared with HSLK in the same genotype. P<0.05, WT compared withBsndneo/neo mice on the same diet. Abbreviations: BUN, blood urea nitrogen (mg/dl); Cl, chloride (mmol/l); Ht, hematocrit (%), aldosterone (pg/ml); K, potassium (mmol/l); Na, sodium (mmol/l); tCO2, total CO2 (mmol/l).

We performed immunoblotting to evaluate the relative levels of WNK4-SPAK-NCC cascade. First, we analyzed the expression levels of barttin and ClC-K in the WT mice consuming an HSNK or HSLK diet, and we found that there was no significant difference in barttin and ClC-K between the diets (Figure 2A). Next, we found that both pNCC and tNCC were more abundant in the kidneys of WT mice consuming HSLK diet than those maintained upon HSNK diet (Figure 2B). In Bsndneo/neo mice, the increase in pNCC and tNCC with HSLK diet was not enough to show a statistical difference (Figure 2B). The abundance of pSPAK and WNK4 in WT mice consuming HSLK diet showed significant increase and increasing tendency, respectively. However, the increase was not evident in Bsndneo/neo mice (Figure 2C). The abundance of pNCC is known to correlate with plasma K+ concentration [6]. We found that pNCC/tNCC was well correlated with plasma K+ level in both WT mice (r = −0.76, P=0.0038) and Bsndneo/neo mice (r = −0.68, P=0.0071) (Figure 2D). However, the slope of the regression lines was significantly greater in WT mice than in Bsndneo/neo mice (P=0.020). These results indicate that the activation of WNK4-SPAK-NCC cascade in response to a low-K+ diet is dependent upon ClC-K function.

Relationship between plasma potassium and the WNK4-SPAK-NCC cascade

Figure 2
Relationship between plasma potassium and the WNK4-SPAK-NCC cascade

(A) Representative immunoblots of barttin and ClC-K, and densitometric analysis in WT mice (n=6). (B) Representative immunoblots of NCC and densitometric analysis. WT-HSNK, n=8; WT-HSLK, n=9; Bsndneo/neo-HSNK, n=7; Bsndneo/neo-HSLK, n=7. (C) Representative immunoblots of WNK4 and SPAK, and densitometric analysis. Arrows on the left side of WNK4, pSPAK and tSPAK blots indicate WNK4, pSPAK and tSPAK bands (anti-pSPAK antibody from Dr Yang). Number of animals was same as (B). (D) Regression lines between the intensities of pNCC/tNCC and plasma K+ levels. Gray squares, open squares, gray circles, and open circles indicate data from WT-HSLK, WT-HSNK, Bsndneo/neo-HSLK, and Bsndneo/neo-HSNK groups, respectively. The slopes of the regression lines were significantly different (P<0.05). Response of pNCC and pSPAK to an HSLK diet was impaired in Bsndneo/neo mice. *P<0.05 and **P<0.005 by Bonferroni’s test after two-way ANOVA. The linearity of protein detection for each antibody was confirmed in Supplementary Figures S2. Abbreviations: K, potassium; n.s., not significant.

Figure 2
Relationship between plasma potassium and the WNK4-SPAK-NCC cascade

(A) Representative immunoblots of barttin and ClC-K, and densitometric analysis in WT mice (n=6). (B) Representative immunoblots of NCC and densitometric analysis. WT-HSNK, n=8; WT-HSLK, n=9; Bsndneo/neo-HSNK, n=7; Bsndneo/neo-HSLK, n=7. (C) Representative immunoblots of WNK4 and SPAK, and densitometric analysis. Arrows on the left side of WNK4, pSPAK and tSPAK blots indicate WNK4, pSPAK and tSPAK bands (anti-pSPAK antibody from Dr Yang). Number of animals was same as (B). (D) Regression lines between the intensities of pNCC/tNCC and plasma K+ levels. Gray squares, open squares, gray circles, and open circles indicate data from WT-HSLK, WT-HSNK, Bsndneo/neo-HSLK, and Bsndneo/neo-HSNK groups, respectively. The slopes of the regression lines were significantly different (P<0.05). Response of pNCC and pSPAK to an HSLK diet was impaired in Bsndneo/neo mice. *P<0.05 and **P<0.005 by Bonferroni’s test after two-way ANOVA. The linearity of protein detection for each antibody was confirmed in Supplementary Figures S2. Abbreviations: K, potassium; n.s., not significant.

Close modal

In ex vivo kidney slice experiment, the increase in NCC phosphorylation in a low-K+ medium was not evident in Bsndneo/neo mice

To exclude non-specific and indirect effects on pNCC and pSPAK with HSLK diet in Bsndneo/neo mice, we performed ex vivo kidney slice experiments, as previously performed [29]. In the kidney slices from WT mice, pNCC and pSPAK were significantly increased in a low-K+ medium, however, which was not observed in the kidney slices from Bsndneo/neo mice (Figure 3). These data strongly supported our in vivo evidence that ClC-K is involved in SPAK and NCC activation in response to K+ restriction.

Ex vivo kidney slices experiment

Figure 3
Ex vivo kidney slices experiment

(A) Representative immunoblots of kidney slices incubated in a normal or low-K+ medium. (B) Densitometric analysis of pNCC and SPAK in kidney slices (new anti-pSPAK antibody). The slices from the same mouse kidney were incubated in a normal medium or a low-K+ medium. The fold change to a normal medium was analyzed by unpaired ttest; *P<0.05, n=6. Open columns indicate normal medium groups, gray columns indicate low-K medium groups. The linearity of protein detection for each antibody was confirmed in Supplementary Figure S3. Abbreviations: LK, low potassium medium (K+ 2 mEq/l); NK, normal potassium medium (K+ 4 mEq/l); n.s., not significant.

Figure 3
Ex vivo kidney slices experiment

(A) Representative immunoblots of kidney slices incubated in a normal or low-K+ medium. (B) Densitometric analysis of pNCC and SPAK in kidney slices (new anti-pSPAK antibody). The slices from the same mouse kidney were incubated in a normal medium or a low-K+ medium. The fold change to a normal medium was analyzed by unpaired ttest; *P<0.05, n=6. Open columns indicate normal medium groups, gray columns indicate low-K medium groups. The linearity of protein detection for each antibody was confirmed in Supplementary Figure S3. Abbreviations: LK, low potassium medium (K+ 2 mEq/l); NK, normal potassium medium (K+ 4 mEq/l); n.s., not significant.

Close modal

Bsndneo/neo mice did not show an elevation in blood pressure when fed an HSLK diet

To determine the contribution of ClC-K in the rise in blood pressure induced by a low-K+ diet, we compared the effects of diet to blood pressure between Bsndneo/neo mice and WT mice. In WT mice, blood pressure showed increasing tendency (n=14, P=0.10), when fed an HSLK diet, but did not show an increase in Bsndneo/neo mice (n=11, P=0.10) (Figure 4A). The change in blood pressure between the diets was significantly greater in the WT mice than in the Bsndneo/neo mice (P=0.009) (Figure 4B). These data indicate that ClC-K plays a role in hypertension induced by a low-K+ diet, probably via the activation of WNK4, SPAK, and NCC.

Systolic blood pressure measurement by tail cuffs

Figure 4
Systolic blood pressure measurement by tail cuffs

At first, the mice were maintained on an HSNK diet, then the food was switched to an HSLK diet. BP were compared between the HSNK diet and the HSLK diet. (A) The average of systolic BP. BP from HSLK diet was significantly greater in WT than in Bsndneo/neo. (B) A box-and-whisker plot of the change in BP between the diets. Boxes demonstrate median and 25–75% range; whiskers extend to the most extreme data point. The averages were shown as ‘x’s in the boxes. The change in BP was greater in WT than in Bsndneo/neo. *P<0.05 according to the unpaired t test. n=14 (WT), n=11 (Bsndneo/neo). Abbreviations: BP, blood pressure; HSLK, high-salt and low-potassium diet; ΔBP, change in BP.

Figure 4
Systolic blood pressure measurement by tail cuffs

At first, the mice were maintained on an HSNK diet, then the food was switched to an HSLK diet. BP were compared between the HSNK diet and the HSLK diet. (A) The average of systolic BP. BP from HSLK diet was significantly greater in WT than in Bsndneo/neo. (B) A box-and-whisker plot of the change in BP between the diets. Boxes demonstrate median and 25–75% range; whiskers extend to the most extreme data point. The averages were shown as ‘x’s in the boxes. The change in BP was greater in WT than in Bsndneo/neo. *P<0.05 according to the unpaired t test. n=14 (WT), n=11 (Bsndneo/neo). Abbreviations: BP, blood pressure; HSLK, high-salt and low-potassium diet; ΔBP, change in BP.

Close modal

Using Bsndneo/neo mice, we demonstrated that ClC-K and barttin play an important role in the activation of WNK4-SAPK-NCC cascade in low-K+ conditions. When WT mice were fed an HSLK diet, the WNK4-SPAK-NCC cascade was activated, however, such activation did not occur in Bsndneo/neo mice. In previous in vitro studies using HEK293 cells, loss-of-function mutant ClC-K2 transfection and a Cl channel inhibitor (DIDS) treatment showed a lower increase in pNCC in the response to a low-K+ condition [4]. Consistent with these findings in vitro, our in vivo findings strongly support the contribution of ClC-K2 in the mechanism underlying the low-K+-induced phosphorylation of NCC.

The potential importance of the Cl channel in the regulation of WNK kinases is based on the idea that WNKs behave as Cl-sensitive kinases [6,30]. WNKs have direct Cl-binding sites in their catalytic sites, and these residues are conserved amongst WNKs [15,16]. The Cl ion binding to these sites inhibits autophosphorylation (= activity) of WNK kinases. It is thought that the negative basolateral membrane potential (hyperpolarization) is the main driving force for Cl to exit the cell [31]. In low-K+ condition, the Kir4.1/Kir5.1 complex makes the driving force because it is thought to be the predominant K+ channels in the basolateral membrane of DCT cells [32,33]. Indeed, the recent report about the doxycycline inducible kidney-specific Kcnj10 knockout mouse showed that the lack of Kir4.1 (coded by Kcnj10) decreased the K+ reversal potential and basolateral Cl conductance in DCT cells [34]. In humans, loss-of-function mutations in the gene encoding Kir4.1 cause SeSAME/EAST syndrome, characterized by an electrolyte imbalance reminiscent of Gitelman syndrome, including salt wasting, hypocalciuria, hypomagnesemia, and hypokalemic metabolic alkalosis [35]. Because ClC-K2/b is the main Cl channel in the basolateral membrane of DCT cell [21], it is expected that the lack of ClC-K2/b disrupts the Cl exit in response to the change of [K+]ex. Indeed, although we could not analyze a change in [Cl]i in DCT cells, our finding that the Bsndneo/neo mice did not show an increase in pSPAK after consuming a low-K+ diet strongly supports this hypothesis. We designed the scheme in Figure 5.

A scheme illustrating the mechanism

Figure 5
A scheme illustrating the mechanism

Basolateral low extracellular potassium regulates the phosphorylation of the apical NCC in DCT cells of WT mice (left panel). In Bsndneo/neo mice (right panel), NCC was not activated by low extracellular potassium.

Figure 5
A scheme illustrating the mechanism

Basolateral low extracellular potassium regulates the phosphorylation of the apical NCC in DCT cells of WT mice (left panel). In Bsndneo/neo mice (right panel), NCC was not activated by low extracellular potassium.

Close modal

Furthermore, we found that a low-K+ diet induced an increase in blood pressure in WT mice, which was not observed in the Bsndneo/neo mice. This indicates that ClC-K2/b contributes to the low-K+-induced hypertension via the WNK4-SPAK-NCC cascade. Because the elevation of blood pressure by the low-K+ diets was related to cardiovascular events and mortality [3], our findings suggested that CLC-K2/b could also be a drug target to decrease cardiovascular events as well as blood pressure.

A previous study clearly showed the decrease in NCC in ClC-K2 KO mice on the normal diet [21]. This result indicates the importance of ClC-K2 in NCC expression. However, the Bsndneo/neo mice on the normal diet did not show a decrease in NCC (Figure 1C). One possible explanation for the discrepancy is that a minimal expression of ClC-K2 in Bsndneo/neo mice might be enough for NCC expression. A previous experiment using isolated distal tubules showed that the Cl channel inhibitor NPPB had no effect on basolateral resting membrane potential [36]. Thus, it seems that the Cl efflux via ClC-K2 mainly occurs when a negative basolateral potential is generated by K+ channels, and the contribution of ClC-K2 for NCC expression on normal diet might be minimal.

In summary, Bsndneo/neo mice consuming a low-K+ diet showed a blunted activation of the WNK4-SPAK-NCC cascade and a lesser increase in blood pressure. ClC-K2/b and barttin play important roles in low-K+-induced phosphorylation of NCC and regulation of blood pressure via the WNK4-SPAK cascade.

We thank Enago (www.enago.jp) for the English language review.

The authors declare that there are no competing interests associated with the manuscript.

This work was supported by the Grants-in-Aid for Scientific Research (KAKENHI) from Japan Society of the Promotion of Science (JSPS) [grant numbers JP125221306, JP15K15327, JP16K09642, JP16H05314, JP16K15467, JP16K19478, 15H06183, 15H06184]; and the Salt Science Research Foundation [grant number 1629].

N.N. conceived and designed studies, conducted the experiments with acquisition and analyses of data, and wrote the manuscript. W.S. and Y.W. conducted part of the experiments with acquisition of data. S.M. and T.F. confirmed the specificity of antibodies. D.T., M.Z., E.S., and T.R. participated in discussions and interpretation of the data. S.U. supervised the project and revised the manuscript.

DCT

distal convoluted tubule

DIDS

4,4′-diisothiocyanato-2,2′-stilbenedisulfonic acid disodium salt

HSLK

high-salt and low-K

HSNK

high-salt and normal-K

KO

knockout

NCC

sodium chloride cotransporter

NPPB

5-Nitro-2-(3-phenylpropylamino)benzoic acid

OSR1

oxidative stress-related kinase 1

SPAK

STE20/SPS1-44 related proline/alanine-rich kinase

tNCC

total NCC

tSPAK

total SPAK

WNK

with-no-lysine kinase

WT

wilsd-type

1
Mente
A.
,
O’Donnell
M.J.
,
Rangarajan
S.
,
McQueen
M.J.
,
Poirier
P.
,
Wielgosz
A.
et al
(
2014
)
Association of urinary sodium and potassium excretion with blood pressure
.
N. Engl. J. Med.
371
,
601
611
[PubMed]
2
Riphagen
I.J.
,
Gijsbers
L.
,
van Gastel
M.D.A.
,
Kema
I.P.
,
Gansevoort
R.T.
,
Navis
G.
et al
(
2016
)
Effects of potassium supplementation on markers of osmoregulation and volume regulation
.
J. Hypertens.
34
,
215
220
[PubMed]
3
O’Donnell
M.
,
Mente
A.
,
Rangarajan
S.
,
McQueen
M.J.
,
Wang
X.
,
Liu
L.
et al
(
2014
)
Urinary sodium and potassium excretion, mortality, and cardiovascular events
.
N. Engl. J. Med.
371
,
612
623
[PubMed]
4
Terker
A.S.
,
Zhang
C.
,
McCormick
J.A
,
Lazelle
R.A
,
Zhang
C.
,
Meermeier
N.P.
et al
(
2015
)
Potassium modulates electrolyte balance and blood pressure through effects on distal cell voltage and chloride
.
Cell Metab.
21
,
39
50
[PubMed]
5
Castañeda-Bueno
M.
,
Cervantes-Perez
L.G.
,
Rojas-Vega
L.
,
Arroyo-Garza
I.
,
Vázquez
N.
,
Moreno
E.
et al
(
2014
)
Modulation of NCC activity by low and high K(+) intake: insights into the signaling pathways involved
.
Am. J. Physiol. Renal Physiol.
306
,
F1507
F1519
[PubMed]
6
Terker
A.S.
,
Zhang
C.
,
Erspamer
K.J.
,
Gamba
G.
,
Yang
C.
and
Ellison
D.H.
(
2016
)
Unique chloride-sensing properties of WNK4 permit the distal nephron to modulate potassium homeostasis
.
Kidney Int.
89
,
127
134
7
Takahashi
D.
,
Mori
T.
,
Nomura
N.
,
Khan
M.Z.H.
,
Araki
Y.
,
Zeniya
M.
et al
(
2014
)
WNK4 is the major WNK positively regulating NCC in the mouse kidney
.
Biosci. Rep.
34
,
[PubMed]
8
Wade
J.B.
,
Liu
J.
,
Coleman
R.
,
Grimm
P.R.
,
Delpire
E.
and
Welling
P.A.
(
2015
)
SPAK-mediated NCC regulation in response to low-K+ diet
.
Am. J. Physiol. Renal Physiol.
308
,
F923
F931
[PubMed]
9
Ferdaus
M.Z.
,
Barber
K.W.
,
López-Cayuqueo
K.I.
,
Terker
A.S.
,
Argaiz
E.R.
,
Gassaway
B.M.
et al
(
2016
)
SPAK and OSR1 play essential roles in potassium homeostasis through actions on the distal convoluted tubule
.
J. Physiol.
594
,
4945
4966
[PubMed]
10
Vitzthum
H.
,
Seniuk
A.
,
Schulte
L.H.
,
Müller
M.L.
,
Hetz
H.
and
Ehmke
H.
(
2014
)
Functional coupling of renal K+ and Na+ handling causes high blood pressure in Na+ replete mice
.
J. Physiol.
592
,
1139
1157
[PubMed]
11
Wilson
F.H.
,
Disse-Nicodeme
S.
,
Choate
K.A.
,
Ishikawa
K.
,
Nelson-Willams
C.
,
Desitter
I.
et al
(
2001
)
Human hypertension caused by mutations in WNK kinases
.
Science
293
,
1107
1112
12
Simon
D.B.
,
Nelson-Williams
C.
,
Bia
M.J.
,
Ellison
D.
,
Karet
F.E.
,
Molina
A.M.
et al
(
1996
)
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
[PubMed]
13
Moriguchi
T.
,
Urushiyama
S.
,
Hisamoto
N.
,
Iemura
S.I.
,
Uchida
S.
,
Natsume
T.
et al
(
2005
)
WNK1 regulates phosphorylation of cation-chloride-coupled cotransporters via the STE20-related kinases, SPAK and OSR1
.
J. Biol. Chem.
280
,
42685
42693
[PubMed]
14
Vitari
A.C.
,
Deak
M.
,
Morrice
N.A.
and
Alessi
D.R.
(
2005
)
The WNK1 and WNK4 protein kinases that are mutated in Gordon’s hypertension syndrome phosphorylate and activate SPAK and OSR1 protein kinases
.
Biochem. J.
391
,
17
[PubMed]
15
Bazúa-Valenti
S.
,
Chávez-Canales
M.
,
Rojas-Vega
L.
,
González-Rodríguez
X.
,
Vázquez
N.
,
Rodríguez-Gama
A.
et al
(
2015
)
The effect of WNK4 on the Na+-Cl- cotransporter is modulated by intracellular chloride
.
J. Am. Soc. Nephrol.
26
,
1781
1786
[PubMed]
16
Piala
A.T.
,
Moon
T.M.
,
Akella
R.
,
He
H.
,
Cobb
M.H.
and
Goldsmith
E.J.
(
2014
)
Chloride sensing by WNK1 involves inhibition of autophosphorylation
.
Sci. Signal.
7
,
ra41
[PubMed]
17
Kobayashi
K.
,
Uchida
S.
,
Mizutani
S.
,
Sasaki
S.
and
Marumo
F.
(
2001
)
Intrarenal and cellular localization of CLC-K2 protein in the mouse kidney
.
J. Am. Soc. Nephrol.
12
,
1327
1334
[PubMed]
18
Yoshikawa
M.
,
Uchida
S.
,
Yamauchi
A
,
Miyai
A
,
Tanaka
Y.
,
Sasaki
S.
et al
(
1999
)
Localization of rat CLC-K2 chloride channel mRNA in the kidney
.
Am. J. Physiol.
276
,
F552
F558
[PubMed]
19
Nozu
K.
,
Iijima
K.
,
Kanda
K.
,
Nakanishi
K.
,
Yoshikawa
N.
,
Satomura
K.
et al
(
2010
)
The pharmacological characteristics of molecular-based inherited salt-losing tubulopathies
.
J. Clin. Endocrinol. Metab.
95
,
E511
E518
[PubMed]
20
Fukuyama
S.
,
Okudaira
S.
,
Yamazato
S.
,
Yamazato
M.
and
Ohta
T.
(
2003
)
Analysis of renal tubular electrolyte transporter genes in seven patients with hypokalemic metabolic alkalosis
.
Kidney Int.
64
,
808
816
[PubMed]
21
Hennings
J.C.
,
Andrini
O.
,
Picard
N.
,
Paulais
M.
,
Huebner
A.K.
,
Cayuqueo
I.K.L.
et al
(
2017
)
The ClC-K2 chloride channel is critical for salt handling in the distal nephron
.
J. Am. Soc. Nephrol.
28
,
209
217
[PubMed]
22
Nomura
N.
,
Tajima
M.
,
Sugawara
N.
,
Morimoto
T.
,
Kondo
Y.
,
Ohno
M.
et al
(
2011
)
Generation and analyses of R8L barttin knockin mouse
.
Am. J. Physiol. Renal Physiol.
301
,
F297
F307
[PubMed]
23
Birkenhäger
R.
,
Otto
E.
,
Schürmann
M.J.
,
Vollmer
M.
,
Ruf
E.M.
,
Maier-Lutz
I.
et al
(
2001
)
Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure
.
Nat. Genet.
29
,
310
314
[PubMed]
24
Estévez
R.
,
Boettger
T.
,
Stein
V.
,
Birkenhäger
R.
,
Otto
E.
,
Hildebrandt
F.
et al
(
2001
)
Barttin is a Cl- channel β-subunit crucial for renal Cl- reabsorption and inner ear K+ secretion
.
Nature
414
,
558
561
[PubMed]
25
Rickheit
G.
,
Maier
H.
,
Strenzke
N.
,
Andreescu
C.E.
,
De Zeeuw
C.I.
,
Muenscher
A.
et al
(
2008
)
Endocochlear potential depends on Cl- channels: mechanism underlying deafness in Bartter syndrome IV
.
EMBO J.
27
,
2907
2917
[PubMed]
26
Sohara
E.
,
Rai
T.
,
Yang
S.-S.
,
Ohta
A.
,
Naito
S.
,
Chiga
M.
et al
(
2011
)
Acute insulin stimulation induces phosphorylation of the Na-Cl cotransporter in cultured distal mpkDCT cells and mouse kidney
.
PLoS ONE
6
,
e24277
[PubMed]
27
Yang
S.
,
Morimoto
T.
,
Rai
T.
,
Chiga
M.
,
Sohara
E.
,
Ohno
M.
et al
(
2007
)
Molecular pathogenesis of pseudohypoaldosteronism type II: generation and analysis of a Wnk4(D561A/+) knockin mouse model
.
Cell Metab.
5
,
331
344
[PubMed]
28
Nomura
N.
,
Nunes
P.
,
Bouley
R.
,
Nair
A.V
,
Shaw
S.
,
Ueda
E.
et al
(
2014
)
High-throughput chemical screening identifies AG-490 as a stimulator of aquaporin 2 membrane expression and urine concentration
.
Am. J. Physiol. Cell Physiol.
307
,
C597
C605
[PubMed]
29
Penton
D.
,
Czogalla
J.
,
Wengi
A.
,
Himmerkus
N.
,
Loffing-Cueni
D.
,
Carrel
M.
et al
(
2016
)
Extracellular K + rapidly controls NCC phosphorylation in native DCT by Cl-dependent and -independent mechanisms
.
J. Physiol.
594
,
6319
6331
30
Naito
S.
,
Ohta
A.
,
Sohara
E.
,
Ohta
E.
,
Rai
T.
,
Sasaki
S.
et al
(
2011
)
Regulation of WNK1 kinase by extracellular potassium
.
Clin. Exp. Nephrol.
15
,
195
202
[PubMed]
31
Zaika
O.
,
Tomilin
V.
,
Mamenko
M.
,
Bhalla
V.
and
Pochynyuk
O.
(
2016
)
New perspective of ClC-Kb/2 Cl − channel physiology in the distal renal tubule
.
Am. J. Physiol. Renal Physiol.
310
,
F923
F930
32
Zhang
C.
,
Wang
L.
,
Zhang
J.
,
Su
X.
,
Lin
D.
,
Scholl
U.I.
et al
(
2014
)
KCNJ10 determines the expression of the apical Na-Cl cotransporter (NCC) in the early distal convoluted tubule (DCT1)
.
Proc. Natl. Acad. Sci. U.S.A.
111
,
6
11
33
Lourdel
S.
,
Paulais
M.
,
Cluzeaud
F.
,
Bens
M.
,
Tanemoto
M.
,
Kurachi
Y.
et al
(
2002
)
An inward rectifier K(+) channel at the basolateral membrane of the mouse distal convoluted tubule: similarities with Kir4-Kir5.1 heteromeric channels
.
J. Physiol.
538
,
391
404
[PubMed]
34
Cuevas
C.A.
,
Su
X.-T.
,
Wang
M.-X.
,
Terker
A.S.
,
Lin
D.-H.
,
McCormick
J.A.
et al
(
2017
)
Potassium sensing by renal distal tubules requires Kir4.1
.
J. Am. Soc. Nephrol.
28
,
1814
1825
[PubMed]
35
Bockenhauer
D.
,
Feather
S.
,
Stanescu
H.C.
,
Bandulik
S.
,
Zdebik
A.A.
,
Reichold
M.
et al
(
2009
)
Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations
.
N. Engl. J. Med.
360
,
1960
1970
[PubMed]
36
Zaika
O.
,
Mamenko
M.
,
Boukelmoune
N.
and
Pochynyuk
O.
(
2015
)
IGF-1 and insulin exert opposite actions on ClC-K2 activity in the cortical collecting ducts
.
Am. J. Physiol. Renal Physiol
308
,
F39
F48
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).

Supplementary data