Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2022 May 30;323(2):F141–F155. doi: 10.1152/ajprenal.00013.2022

NHE3 in the thick ascending limb is required for sustained but not acute furosemide-induced urinary acidification

Jianxiang Xue 1, Linto Thomas 1, Jessica A Dominguez Rieg 1,2, Robert A Fenton 3, Timo Rieg 1,2,4,
PMCID: PMC9306792  PMID: 35635321

graphic file with name f-00013-2022r01.jpg

Keywords: acid-base, furosemide, kidney, Na+/H+ exchanger isoform 3, thick ascending limb

Abstract

Na+/H+ exchanger isoform 3 (NHE3) facilitates Na+ reabsorption and H+ secretion by the kidneys. Despite stronger NHE3 abundance in the thick ascending limb (TAL) compared with the S1 and S2 segments of the proximal tubule, the role of NHE3 in the TAL is poorly understood. To investigate the role of NHE3 in the TAL, we generated and phenotyped TAL-specific NHE3 knockout (NHE3TAL-KO) mice. Compared with control mice, NHE3TAL-KO mice did not show significant differences in body weight, blood pH, or plasma Na+, K+, or Cl levels. Fluid intake trended to be higher and urine osmolality was significantly lower in NHE3TAL-KO mice. Despite a similar glomerular filtration rate, NHE3TAL-KO mice had a greater urinary K+-to-creatinine ratio. One proposed role of NHE3 relates to furosemide-induced urinary acidification. Acute bolus treatment with furosemide under anesthesia did not result in differences in the dose dependence of urinary flow rate, Cl excretion, or maximal urinary acidification between genotypes; however, in contrast with control mice, urinary pH returned immediately toward baseline levels in NHE3TAL-KO mice. Chronic furosemide treatment reduced urine osmolality similarly in both genotypes but metabolic alkalosis, hypokalemia, and calciuresis were absent in NHE3TAL-KO mice. Compared with vehicle, chronic furosemide treatment resulted in greater Na+-K+-2Cl abundance regardless of genotype. Na+-phosphate cotransporter 2a abundance was also greater in furosemide-treated control mice compared with vehicle treatment, an effect that was absent in NHE3TAL-KO mice. In summary, NHE3 in the TAL plays a role in the sustained acidification effect of furosemide. Consistent with this, long-term treatment with furosemide did not result in metabolic alkalosis in NHE3TAL-KO mice.

NEW & NOTEWORTHY Na+/H+ exchanger isoform 3 (NHE3) is very abundant in the thick ascending limb (TAL) compared with the proximal tubule. Much has been learned about the role of NHE3 in the proximal tubule; however, the function of NHE3 in the TAL remains elusive. A novel mouse model that lacks NHE3 selectively in the TAL not only shows a phenotype under baseline conditions but also identifies that NHE3 is required for sustained but not acute furosemide-induced urinary acidification.

INTRODUCTION

Na+/H+ exchanger isoform 3 (NHE3) is important for Na+ absorption in the intestine and kidney. In the intestine, apical NHE3 is critical for Na+ and fluid absorption (1), and knockout of NHE3 selectively in the small intestine and colon results in persistent diarrhea, increased mortality rate, metabolic acidosis, lower blood HCO3 levels, hyponatremia, and hyperkalemia associated with drastically elevated plasma aldosterone levels and changes to intestinal structural integrity (2).

In the kidney, NHE3 is expressed in the apical membrane of the S1 and S2 segments of the proximal tubule (3, 4) and thick ascending limb (TAL). In the proximal tubule, NHE3 facilitates the majority of renal Na+ reabsorption (60%–75%) and H+ secretion (1, 3, 5). Ex vivo perfusion studies have shown that proximal tubule Na+ and HCO3 reabsorption were reduced by ∼60% in conventional NHE3-knockout mice compared with wild-type mice. In rat micropuncture experiments, in vivo perfusion with an NHE3 inhibitor (S3226) demonstrated that the maximum reduction of Na+ reabsorption was only ∼30% in superficial nephrons accessible by micropuncture (6). Renal NHE3 contributes to the NaCl sensitivity of blood pressure, and studies have identified that dietary NaCl alterations directly impact total body NaCl levels by disturbing glomerulotubular balance (7). Of note, knockout of NHE3 along the nephron did not affect blood pH or HCO3 (7).

Immunostaining of NHE3 in the TAL was qualitatively stronger compared with the proximal tubule (3, 4), suggesting that the abundance of NHE3 in this segment is greater. Supporting this, RNA-sequencing analysis from microdissected kidney tubule segments demonstrated that NHE3 mRNA is quantitatively greater in the TAL relative to the proximal tubule of rats and mice (810); however, protein copy numbers per cell showed the opposite (10, 11). Functionally, the role of NHE3 in the TAL is less clearly defined. Hormonal regulation of NHE3 in the TAL by arginine-vasopressin (AVP) has been described, with quantitative phosphoproteomics demonstrating AVP-dependent phosphorylation of NHE3 at serine residue 552 (12), which is an inhibitory phosphorylation site (13, 14). Furthermore, administration of angiotensin II to rats caused a significant increase in NHE3 abundance in the inner stripe of the outer medulla, a region where the medullary TAL, but not the proximal tubule, is found (15). NHE3 in the TAL contributes to acid-base regulation (16), in particular HCO3 reabsorption (17). More recently, a new mechanism for the role of NHE3 in the TAL during furosemide-induced urinary acidification was proposed (18). Here, furosemide inhibition of Na+-K+-2Cl cotransporter (NKCC2) in the TAL increases the driving force for Na+ reabsorption by NHE3, which subsequently results in increased H+ secretion and a more acidic tubular fluid and urine.

To increase our understanding of the role of NHE3 in the TAL, we generated a TAL-specific NHE3-knockout (NHE3TAL-KO) mouse model by crossing mice with tamoxifen-inducible Cre recombinase expression under the regulation of the uromodulin gene (encoding Tamm–Horsfall protein) with floxed NHE3 mice (19). We subsequently used this model to study the isolated role of NHE3 in the TAL of adult mice for electrolyte and acid-base homeostasis. Second, we used the model to investigate in vivo whether NHE3 in the TAL is required for urinary acidification in response to furosemide. Our results demonstrate that NHE3 in the TAL is important for a sustained but not acute acidification effect of furosemide.

MATERIALS AND METHODS

Animals

All animal experimentation was conducted in accordance with the National Institutes of Health Guide for Care and Use of Laboratory Animals (National Institute of Health, Bethesda, MD) and was approved by the University of South Florida Institutional Animal Care and Use Committee (3338 R). Floxed NHE3 (NHE3loxlox) female mice (19) were crossed with hemizygous male C57BL/6N-Umodem1(cre/ERT2)Amc/J mice (Stock No. 030601, Jackson Laboratory, Bar Harbor, ME) that express tamoxifen-inducible Cre recombinase under the control of the uromodulin gene, which is expressed exclusively in the TAL and early distal tubule (20). From the F1 progeny, male mice heterozygous for the floxed NHE3 allele (NHE3lox/wild-type) and Umodem1(cre/ERT2)Amc/J were bred to female NHE3lox/lox mice to generate final breeder pairs in the F2 progeny. To generate male experimental NHE3TAL-KO and NHE3lox/lox (control) mice, female NHE3lox/lox mice were bred to male NHE3TAL-KO mice. Mice were genotyped by PCR from genomic DNA isolated from ear punch. Genotyping for the floxed allele was as previously described (4); TAL-Cre/ERT2 genotyping was performed according to Protocol No. 29530 (Jackson Laboratory). NHE3 deletion was induced by tamoxifen (200 mg·kg−1 body wt), which was initially dissolved in 5% (vol/vol) of ethanol followed by adding 95% (vol/vol) of corn oil and given by oral gavage (volume: 1% of body wt) for three consecutive days. An equal dose of tamoxifen was given to control and NHE3TAL-KO mice. Littermate age-matched, 3- to 6-mo-old mice were used for experiments. Male mice were used except where indicated in Fig. 3. All acute physiological experiments were performed between 0900 and 1200.

Figure 3.

Figure 3.

Baseline physiological analysis in control and thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice. NHE3TAL-KO mice showed a trend for higher fluid intake (A) associated with a significantly lower urine osmolality (B) compared with control mice. No differences were found in plasma osmolality (C), food intake (D), hematocrit (E), and glomerular filtration rate (GFR; F) between genotypes. Data were analyzed by an unpaired Student’s t test. In addition to single data, means ± SE values are shown. n = 5 or 6 male and 5 female mice/genotype for A and D, n = 10 male and 10 female mice/genotype for B, n = 9 male mice/genotype for C, n = 13–15 male and 6 female mice/genotype for E, and n = 7 or 8 male and 8 female mice/genotype for F. *P < 0.05 vs. control. bw, body weight.

Physiological Analysis

Body weight and fluid and food intake were determined for 10 days starting after tamoxifen administration (21). Spontaneous-voided urine was collected daily by reflex urination/gentle bladder massage and holding mice over a clean Petri dish (22). Values from multiple days were averaged. Ten days after tamoxifen administration, blood was collected under brief isoflurane anesthesia from the retroorbital plexus for analysis.

Acute Responses to Furosemide

Acute responses to furosemide were assessed in anesthetized mice as previously described (23). Mice were anesthetized with thiobutabarbital (100 mg·kg−1 body wt ip, 2 μL·g−1 body wt, Sigma-Aldrich, St. Louis, MO) and ketamine (100 mg·kg−1 body wt im, 2 μL·g−1 body wt) (24). Mice were placed on an operating table with a servo-controlled heating plate (RT, Effenberger, Munich, Germany) to maintain body temperature at 37.5°C. The trachea was cannulated, and 100% oxygen was blown toward the tracheal tube throughout the experiment. The right jugular vein was cannulated for continuous maintenance infusion of 2.25% BSA in 0.85% NaCl at a rate of 500 µL·h−1·30 g body wt−1. A catheter was placed into the bladder for urine collection. The aforementioned preparation took ∼15 min, after which mice were allowed another 45 min to stabilize before baseline urine was collected for three consecutive 5-min periods. Mice were treated with increasing doses of furosemide (0.1, 0.3, 1, 3, and 10 mg·kg−1 body wt) administered via bolus application (0.5 µL·g−1 body wt, infusion rate: 25 µL·min−1). After each bolus, allowing 2 min for furosemide distribution, urine was collected for two consecutive 5-min periods before the next bolus was applied. The urinary flow rate was calculated based on urine weight, which was determined gravimetrically. Urine was analyzed as described in the Analysis of Urine and Plasma Samples.

Chronic Responses to Furosemide

Chronic responses to furosemide (40 mg·kg−1 ip given twice a day at 08:00 and 20:00) were studied in awake mice. Urine was collected at baseline and after 8 days of furosemide administration. On day 8, mice were anesthetized with isoflurane and a blood collection was performed. Subsequently, the left kidney was removed and immediately snap-frozen. Tissue was processed for immunoblot analysis (22).

Eighteen-Hour Water Deprivation

Water deprivation was conducted overnight (18 h) (25, 26). Blood was taken from the retroorbital plexus for the determination of hematocrit and osmolality, after brief isoflurane anesthesia, before and after water deprivation. At the same time, spontaneous-voided urine was collected for the determination of urine osmolality. After reintroduction of the water bottle, fluid intake was determined after 3, 6, and 24 h.

Analysis of Urine and Plasma Samples

Blood chemistry was determined by an OPTI CCA-TS2 blood gas analyzer using an E-Cl type cassette (OPTIMedical, Roswell, GA). Plasma and urine osmolality were measured using an Osmomat 3000 (Gonotec, Berlin, Germany). Urine clinical chemistry was performed using commercially available assays that were modified to work with small volumes (7, 25, 27). Urinary Cl was analyzed by the thiocyanide method (Stanbio Laboratory, Boerne, TX), and urinary creatinine was determined using Infinity Creatinine Liquid Stable Reagent (Thermo Fisher Scientific, Middletown, VA). Urinary pH was determined using a pH electrode immediately after collection (No. 9810BN, Thermo Fisher Scientific, Pittsburgh, PA).

Measurement of Glomerular Filtration Rate in Conscious Mice

Glomerular filtration rate (GFR) measurements were performed by determining the plasma kinetics of FITC-sinistrin (Fresenius-Kabi, Linz, Austria) following an intravenous bolus injection (28). FITC-sinistrin (2% in isotonic saline) was injected into the retroorbital plexus of mice (2 µL·g−1 body wt) during brief isoflurane anesthesia. At 3, 5, 7, 10, 15, 35, 56, and 75 min after injection, blood was collected from the tip of the tail into Na+-heparinized 10-µL microcaps (Hirschmann Laborgeräte, Eberstadt, Germany). After centrifugation, plasma was diluted at 1:10 in 0.5 mol·L−1 HEPES (pH 7.4), and fluorescence was determined with a fluorospectrometer (Cytation 3, Biotek, Winooski, VT). GFR was calculated using a two-compartment model of two-phase exponential decay (GraphPad Prism, San Diego, CA).

Quantitative RT-PCR

In a different cohort of control and NHE3TAL-KO mice, the left kidney was removed for immunoblot analysis and the right kidney was separated into the cortex and renal medulla and processed for mRNA analysis. Tissue was homogenized using a Tissue Tearor (Bartlesville, OK) and QIAshredder (Qiagen, Valencia, CA) according to the manufacturer’s instructions. Total RNA was purified using the RNeasy Plus Mini Kit (Qiagen). cDNA was made by reverse transcribing total RNA using a QuantiTect reverse transcription kit (Qiagen) according to the manufacturer’s instructions. Semiquantitative RT-PCR was performed using TaqMan Universal PCR Master Mix (Fisher Scientific) in a QuantStudio 3 (Applied Biosystems, Foster City, CA). The template concentration was 12.5 ng cDNA per 25-µL reaction (performed in triplicate) and was used in conjunction with primer pairs specific for NHE3 (Mm01352478_g1, efficiency: 98%) and Sglt2 (Mm00453831_m1, efficiency: 98%) with GAPDH (Mm99999915_g1) as a reference gene. Data analysis used the ΔΔCt method (where Ct is threshold cycle) and normalized to GAPDH expression and compared with control (2).

Immunofluorescent Staining

Tissue preparation, kidney sectioning, and immunolabeling were performed as previously described (2, 29) using antibodies against Lotus tetragonolobus lectin (LTL; 1:1,000, B-1325-2, Vector Laboratories, Burlingame, CA), NHE3 (1:500, SPC-400, StressMarq Biosciences, Victoria, BC, Canada), and NKCC2 (1:100, sc-293222, Santa Cruz Biotechnology, Dallas, TX). DyLight 649 streptavidin (1:1,000, SA-5649-1, Vector Laboratories), Texas red 595 goat anti-rabbit (1:1,500, TI-1000, Vector Laboratories), and Alexa Fluor 488 donkey anti-mouse (1:1,500, A-21202, Life Technologies, Carlsbad, CA) were used for the visualization of LTL, NHE3, and NKCC2, respectively. A mouse-on-mouse kit (BMK-2202, Vector Laboratories) was used to reduce endogenous mouse IgG staining. Sections were mounted using HardSet Antifade Mounting Medium with DAPI (H-1500, Vector Laboratories). An Olympus (Center Valley, PA) FV1000 MPE Multiphoton Laser Scanning Microscope with a ×40 UPLFL objective lens (numerical aperture: 0.75) was used for imaging of labeled sections.

Immunoblot Analysis

Kidney tissue was homogenized in dissection buffer (250 mmol·L−1 sucrose and 10 mmol·L−1 triethanolamine buffer) containing protease inhibitor cocktail (Roche Applied Science, Penzberg, Germany) and Halt phosphatase inhibitor cocktail (Thermo Fisher Scientific). Homogenates were centrifuged at 4,000 g before protein quantity was determined using a standard BCA assay (Thermo Fisher Scientific). Gel samples of equal concentration were made by the addition of Laemmli sample buffer (final concentration of 0.1 mol·L−1 SDS and 15 mg·L−1 DTT). Samples were heated at 65°C for 15 min before Western blot analysis. Proteins were transferred to polyvinylidene difluoride membranes and immunoblotted with rabbit polyclonal antibodies against NHE3 (AB3085, Millipore, Billerica, MA; characterized in Ref. 30), type II Na+-phosphate cotransporter (Npt2a) (27), NKCC2 (31), phospho-Thr58 Na+-Cl cotransporter (NCC) (32), H+-ATPase (33), anion exchanger 2 (AE2) (34), pendrin (35), electrogenic Na+-bicarbonate cotransporter 1 (NBCe1) (36), H+-K+-ATPase M79 (37), Na+-K+-ATPase [No. 06-520, Merck Millipore, Darmstadt, Germany (7)], and mouse monoclonal antibody against NCC (38). For detailed antibody information, see Table 1. Detection was performed with ECL Plus (Amersham, Piscataway, NJ). Secondary antibodies were from Dako (Jena, Germany), and sites of antibody/antigen interaction were visualized using the Enhanced Chemiluminescence System (GE Healthcare, Buckinghamshire, UK) and an Image-Quant LAS 4000 imager (GE Healthcare). Coomassie-stained gels were used to adjust for equal protein loading for immunoblot analysis. Densitometric analyses were performed using Image Studio Lite (Qiagen).

Table 1.

Antibodies used in the present study

Target Source Use Concentration/Dilution Validation
NHE1 sc-136239, Santa Cruz Biotechnology, Dallas, TX WB 1 μg/mL Specific IHC labeling of the basolateral membrane of distal tubule/collecting duct segments, single protein band of ∼90 (∼115 glycosylated) kDa on WB, signal absent in kidney-specific NHE1 knockout mice (unpublished observations)
NHE3 AB3085, Millipore, Billerica, MA WB 2 μg/mL Specific IHC labeling of the apical brush border of proximal tubules, single protein band of ∼75 kDa on WB, signal absent in NHE3 knockout mice (2,3)
Npt2a Researcher generated (39) WB 1 μg/mL Specific IHC labeling of the apical brush border of proximal tubules, single protein band of ∼80 kDa on WB, regulation of band intensities by parathyroid hormone (40)
NKCC2 Researcher generated (31) WB 0.2 μg/mL Specific IHC labeling of the apical membrane of cells in the TAL, single protein band of ∼160 kDa on WB of renal outer medulla homogenates that is absent in knockout mice (41)
NCC Researcher generated (38) WB 0.1 μg/mL Single protein band of ∼140 kDa on WB of renal cortex homogenates that is absent in NCC knockout mice or mock NCC-transfected cells (38)
pT58 NCC Researcher generated (32) WB 0.01 μg/mL Specific IHC labeling of the distal convoluted tubule, single protein band of ∼140 kDa on WB of renal cortex homogenates (32) or NCC-transfected cells that is absent in cells expressing mutant (T58A) form (42)
H+-ATPase B1 subunit Researcher generated (43) WB 0.1 μg/mL Single protein band of ∼62 kDa on WB of rat kidney membrane preparations that disappears on peptide preabsorption, specific labeling of intercalated cells in the kidney collecting duct by IHC that is absent in knockout mice (44)
AE2 Researcher generated (45) WB 1 μg/mL Specific IHC labeling of cells throughout the renal tubule (46), single protein band on WB of lysates from AE2-transfected cells absent in mock-transfected cells (34)
Pendrin Researcher generated (35) WB 0.1 μg/mL Specific IHC labeling of type B and non-A, non-B intercalated cells (35), single protein band of ∼85 kDa on WB of kidney lysates
NBCe1 Researcher generated (36) WB 1 μg/mL Single protein band of ∼120 kDa on WB of lysates from the kidney cortex that is absent from lysates from the outer and inner medulla (36), specific IHC labeling of the basolateral membrane of proximal tubule cells that is absent in knockout mice (47)
H+-K+-ATPase Researcher generated (37) WB 1 μg/mL Nonglycosylated and core-glycosylated forms of protein only observed in H+-K+-ATPase-expressing cells and not mock-transfected cells (37)
Na+-K+-ATPase α1-subunit No. 06-520, Merck Millipore, Darmstadt, Germany WB 0.01 μg/mL Single protein band of ∼100 kDa on WB of lysates from the kidney that is increased by high aldosterone (48)

AE2, anion exchanger 2; IHC, immunohistochemistry; NBCe1, electrogenic Na+-bicarbonate cotransporter 1; NCC, Na+-Cl cotransporter; NHE, Na+-H+ exchanger; NKCC2, Na+-K+-2Cl cotransporter; Npt2a, type II Na+-phosphate cotransporter; pT58 NCC, phospho-Thr58 Na+-Cl cotransporter; WB, Western blot.

Statistical Analyses

Data are expressed as means ± SE. An unpaired Student’s t test was performed to analyze statistical differences between the two groups. One- and two-way ANOVA or two-way mixed-effects ANOVA followed by the two-stage linear step-up procedure of Benjamini, Krieger, and Yekutieli or Tukey multiple comparison tests, as indicated in the figures, were used to test for significant differences between genotype or experimental conditions. All data were analyzed via GraphPad Prism, v. 8.3 or SigmaPlot, v. 12.5. Significance was considered at P < 0.05.

RESULTS

Confirmation of NHE3 Knockout in the TAL

NHE3TAL-KO mice were born at the predicted Mendelian frequencies, appeared grossly indistinguishable from control mice, and developed normally. Mice were treated with tamoxifen, and qualitative immunofluorescence was used to assess NHE3 distribution in the kidney of control and NHE3TAL-KO mice (Fig. 1). LTL was used to guide the identification of proximal tubules, and NKCC2 was used as a marker for the TAL. Staining intensity and NHE3 localization in the proximal tubule were not different between control and NHE3TAL-KO mice. In contrast, NHE3 labeling in the TAL was completely absent in NHE3TAL-KO mice (Fig. 1). Kidneys were dissected into the cortex and medulla and processed for mRNA analysis. Expression of SGLT2, expressed in the S1 and S2 segments of the proximal tubule, was ∼93% lower in the medulla versus the cortex, independent of genotype (Fig. 2A), confirming the purity of the kidney dissection. In contrast, NHE3 mRNA was significantly lower in the cortex (by ∼32%) and medulla (by ∼90%) of NHE3TAL-KO mice compared with control mice (Fig. 2B). Similar results were found at the protein level, with NHE3 abundance in the cortex and medulla of NHE3TAL-KO mice ∼55% and ∼90% lower, respectively, compared with control mice (Fig. 2C). The specificity of the antibody was confirmed using homogenates from an NHE3loxloxPax8Cre mouse (7), which lacks renal NHE3 expression.

Figure 1.

Figure 1.

Thick ascending limb (TAL)-specific Na+/H+ exchanger isoform 3 (NHE3) knockout (NHE3TAL-KO) mice lack immunofluorescent labeling of NHE3 in the TAL of the kidney. Representative confocal images of NHE3 localization in control and NHE3TAL-KO mice are shown. In control mice, NHE3 labeling (red) overlapped in the S1 and S2 segments of the proximal tubule with Lotus tetragonolobus lectin (LTL; proximal tubule marker). Na+-K+-2Cl cotransporter (NKCC2; green signal) is a marker of the TAL and identifies overlapping expression of NHE3 in the TAL (arrowheads, yellow signal in the merged image). In contrast, no NHE3 labeling (arrows) was found in the TAL of NHE3TAL-KO mice (lack of yellow labeling); however, NHE3 staining in the proximal tubule was preserved (magenta signal in the merged image), indicating selective deletion of NHE3 in the TAL.

Figure 2.

Figure 2.

Na+/H+ exchanger isoform 3 (NHE3) mRNA and protein expression are significantly reduced in thick ascending limb-specific NHE3 knockout (NHE3TAL-KO) mice. A: as a marker of the purity of the preparation, mRNA expression of Na+-glucose transporter 2 (SGLT2; expressed in the S1 and S2 segments of the proximal tubule) was used. Independent of genotype, SGLT2 mRNA expression was almost completely absent in the medulla vs. cortex preparation. B: NHE3 mRNA expression in NHE3TAL-KO mice was ∼30% lower in the cortex and ∼94% lower in the medulla compared with control mice. C: compared with control mice, NHE3 protein expression (molecular weight: ∼93 kDa) in NHE3TAL-KO mice was ∼55% and ∼90% lower in the cortex and medulla, respectively. Data were analyzed by an unpaired Student’s t test. In addition to single data, means ± SE values of n = 4–8 male mice/genotype are shown. *P < 0.05 vs. control. WT, wild type.

Baseline Physiological Analysis in Control and NHE3TAL-KO Mice

Body weight was not significantly different between genotypes 10 days after tamoxifen administration (control: 32 ± 1 g and NHE3TAL-KO: 32 ± 1 g, n = 18 and 19, respectively). Measurement of fluid and food intake when mice were housed in their home cages showed that NHE3TAL-KO mice had a tendency for higher fluid intake (Fig. 3A), but this did not reach statistical significance (P = 0.08). Urine osmolality (Fig. 3B) was significantly lower (∼400 mmol·kg−1, P < 0.05) in NHE3TAL-KO mice compared with control mice. No significant differences between genotypes were observed in plasma osmolality, food intake, hematocrit, or GFR (Fig. 3, C–F).

Blood analysis showed no significant differences between genotypes in the concentrations of Na+, K+, Cl, or pH (Fig. 4, A–D). Urinary Na+- and Cl-to-creatinine ratios as well as urinary pH were not significantly different between genotypes (Fig. 4, E–H). In contrast, urinary K+/creatinine (Fig. 4F) was slightly but significantly elevated in NHE3TAL-KO mice compared with control mice.

Figure 4.

Figure 4.

Thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice show no electrolyte or acid-base abnormalities. No differences were observed between genotypes in plasma Na+ (A), K+ (B), Cl (C), blood pH (D), urinary Na+-to-creatinine (E), Cl-to-creatinine ratios (G), or urinary pH (H). F: NHE3TAL-KO mice had significantly higher urinary K+-to-creatinine ratios compared with control mice. Data were analyzed by an unpaired Student’s t test. In addition to single data, means ± SE values of n = 23–25 male mice/genotype are shown. *P < 0.05 vs. control.

Acute Furosemide Responses in Control and NHE3TAL-KO Mice

To test the role of NHE3 in the TAL for furosemide-induced urinary acidification, experiments were performed in anesthetized mice. Furosemide inhibits NKCC2, which, in addition to transporting Na+, K+, and Cl, can transport NH4+, which could acidify the urine (49). Bolus intravenous administration of furosemide resulted in comparable diuretic (ED50: 1.8 ± 0.3 vs. 1.8 ± 0.4 mg·kg−1; Fig. 5A) and chloruretic (ED50: 1.6 ± 0.3 vs. 2.1 ± 0.5 mg·kg−1; Fig. 5B) dose-response curves in control and NHE3TAL-KO mice. Similar results were obtained when urinary Cl excretion was corrected by urinary creatinine (Fig. 5C). Relative to baseline, urinary pH significantly decreased at a dose of 0.3 mg·kg−1 in control mice; however, higher doses did not further decrease urinary pH in the timeframe examined (Fig. 6, Aand B). The maximum decrease was ∼0.45 ± 0.1 pH units. In contrast, urinary pH did not significantly differ from baseline in NHE3TAL-KO mice at a dose of 0.3 mg·kg−1 (Fig. 6, Aand B). At a dose of 1 mg·kg−1, urinary pH significantly decreased versus baseline (−0.6 ± 0.1 pH units), reaching a maximum response that was comparable to control mice. In contrast to control mice, this response was immediately followed by a return toward baseline levels (Fig. 6, Aand B). No additional effects were observed at 3 and 10 mg·kg−1 compared with baseline or previous dose.

Figure 5.

Figure 5.

Acute diuretic and chloruretic responses to furosemide are similar in control and thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice. After administration of increasing doses of furosemide under anesthesia, the diuretic (A), chloruretic (B), and urinary Cl-to-creatinine ratio (C) dose-response curves showed no differences between genotypes. Data were fit to a four-parameter logistic curve, and ED50 values were calculated with SigmaPlot. Data are expressed as means ± SE; n = 10–12 male mice/genotype.

Figure 6.

Figure 6.

Thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice lack a sustained urinary acidification in response to furosemide. A: in control mice, furosemide at doses of 0.3 and 1 mg·kg−1 resulted in a dose-dependent decrease in urinary pH; however, higher doses of furosemide (3 and 10 mg·kg−1) did not further acidify the urine. In NHE3TAL-KO mice, furosemide significantly decreased urinary pH only at a dose of 1 mg·kg−1, followed by an acute increase toward baseline levels. B: single data are shown to better illustrate the lack of change at 1 and 3 mg·kg−1 in control mice, which contrasted with NHE3TAL-KO mice, where urinary pH recovered and remained at a constant higher level. In A, data are shown as means ± SE; in B, single data and means ±SE values of n = 10–12 male mice/genotype are shown. #P < 0.05 vs. vehicle for control. §P < 0.05 vs. vehicle in NHE3TAL-KO mice. †P < 0.05 vs. previous period NHE3TAL-KO mice. Data were analyzed by two-way mixed-effects ANOVA followed by the two-stage linear step-up procedure of Benjamini, Krieger, and Yekutieli.

Chronic Effects of Furosemide Treatment in Control and NHE3TAL-KO Mice

To study the chronic effects of NHE3 in furosemide-induced acidification, mice were treated for 8 days with furosemide. Vehicle treatment (Fig. 7A) did not significantly change urine osmolality in control and NHE3TAL-KO mice (−180 ± 140 vs. −8 ± 214 mmol·kg−1, not significant). In contrast, furosemide treatment significantly reduced urine osmolality in control mice (−1,230 ± 238 mmol·kg−1) and NHE3TAL-KO mice (−1,510 ± 77 mmol·kg−1), with no significantly different effects between genotypes. In contrast to acute experiments under anesthesia (Fig. 6), no difference in urinary pH was observed in response to furosemide (Fig. 7B). Consistent with furosemide-induced hypokalemia and metabolic alkalosis, blood K+ (Fig. 7C) and blood pH (Fig. 7D) were significantly lower and higher, respectively, in furosemide-treated control mice compared with vehicle-treated mice. Neither plasma K+ (Fig. 7C) nor blood pH (Fig. 7D) was significantly different between vehicle- and furosemide-treated NHE3TAL-KO mice. Furosemide also increases urinary Ca2+ excretion (50). Consistent with this, in control mice, the urinary Ca2+-to-creatinine ratio increased approximately threefold compared with baseline (Fig. 7E). In contrast, in furosemide-treated NHE3TAL-KO mice, the urinary Ca2+-to-creatinine ratio increase was ∼1.5-fold compared with baseline, which was not statistically significant, and remained significantly lower compared with furosemide-treated control mice. Vehicle treatment did not affect urinary Ca2+-to-creatinine ratios in control or NHE3TAL-KO mice.

Figure 7.

Figure 7.

Thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice lack hypokalemia, metabolic alkalosis, and calciuresis in response to chronic furosemide treatment. A: chronic (8 day) furosemide treatment significantly reduced urine osmolality in control and NHE3TAL-KO mice; vehicle treatment was without effect in either genotype. B: urinary pH was not affected by genotype or treatment. In control mice, furosemide treatment resulted in hypokalemia (C), metabolic alkalosis (D), and calciuresis (E). In NHE3TAL-KO mice, these effects were absent. Data were analyzed by repeated-measures two-way ANOVA (A, B, and E) and two-way ANOVA (C and D), with both followed by a Tukey’s multiple comparison test. In addition to single data, means ± SE values of n = 6 or 7 male mice/genotype are shown. *P < 0.05 vs. control. #P < 0.05 vs. baseline same genotype.

Response to Water Deprivation

To test for the role of NHE3 in the TAL in urinary concentrating ability, mice were challenged by an 18-h water deprivation. Urine osmolality increased significantly in control mice (change: 1,255 ± 121 mmol·kg−1) and NHE3TAL-KO mice (change: 1,489 ± 132 mmol·kg−1), which was not significantly different between genotypes (Supplemental Fig. S1A: https://doi.org/10.6084/m9.figshare.19705429.v1). In response to water deprivation, both genotypes showed a significant increase in plasma osmolality (Supplemental Fig. S1B) and hematocrit (Supplemental Fig. S1C) as well as a decrease in body weight (Supplemental Fig. 1D) that were not significantly different between control and NHE3TAL-KO mice. Fluid intake, up to 24 h after water deprivation, was also not significantly different between control mice (7.6 ± 0.5 mL) and NHE3TAL-KO mice (8.5 ± 0.4 mL; Supplemental Fig. S1E).

Profiling of Transporters, Pumps, and Channels Along the Nephron in Response to Chronic Furosemide Treatment in Control and NHE3TAL-KO Mice

Expression levels of NKCC2 were greater in furosemide-treated mice compared with vehicle-treated mice, regardless of genotype (Fig. 8A). In the distal convoluted tubule, the expression (Fig. 8B) and phosphorylation (Fig. 8C) of NCC at Thr58 were greater in both genotypes in response to furosemide treatment compared with vehicle-treated mice. Levels of the Na+-K+-ATPase α-subunit (Fig. 8D) were not affected by treatment or genotype. Full-length Npt2a and NBCe1 are expressed in the luminal and basolateral membrane of the proximal tubule, respectively. In response to chronic furosemide treatment in control mice, full-length Npt2a (Fig. 9A) expression was significantly lower (∼45%, P < 0.05) compared with vehicle-treated control mice. Levels were not significantly affected by treatment in NHE3TAL-KO mice. Levels of NBCe1 (Fig. 9B) were not affected by treatment or genotype. Full-length AE2 is localized to the basolateral membrane in the TAL. No differences in expression levels were observed between genotypes or treatments (Fig. 9C). Expression levels of nongastric H+-K+-ATPase (Fig. 9D), found from the TAL to the inner medullary collecting duct, were not affected by treatment or genotype. Levels of the H+-ATPase β1-subunit (Fig. 9E), located in type A, type B, and non-A, non-B intercalated cells, were not affected by treatment or genotype. Levels of the Cl/HCO3 exchanger pendrin (Fig. 9F), expressed in type B and non-A, non-B intercalated cells, were not significantly affected by treatment or genotype. No compensatory changes were observed in Na+/H+ exchanger isoform 1 (NHE1) expression between genotypes, and NHE1 was not affected by furosemide treatment (Supplemental Fig. S2).

Figure 8.

Figure 8.

Furosemide (Furo)-induced greater abundance of Na+-K+-Cl cotransporter (NKCC2) in control (Con) mice is absent in thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice. A: chronic furosemide treatment (8 days) resulted in significantly higher NKCC2 expression (molecular weight: ∼121 kDa) in control and NHE3TAL-KO mice compared with vehicle (Veh) treatment. B and C: Na+-Cl cotransporter (NCC) expression (B; molecular weight: ∼160 kDa) and phosphorylation at Thr58 (pT58 NCC; C; molecular weight: ∼160 kDa) were significantly higher in response to furosemide treatment independent of genotype. D: expression of the Na+-K+-ATPase α-subunit (molecular weight: ∼112 kDa) was not affected by treatment or genotype. Data were analyzed by two-way ANOVA followed by a Tukey’s multiple comparison test. In addition to single data, means ± SE values of n = 6 male mice/genotype are shown. §P < 0.05 vs. vehicle same genotype.

Figure 9.

Figure 9.

Lower type II Na+-phosphate cotransporter (Npt2a) expression in response to chronic furosemide (Furo) treatment in control (Con) mice is absent in thick ascending limb-specific Na+/H+ exchanger isoform 3 knockout (NHE3TAL-KO) mice. A: chronic furosemide treatment resulted in significantly lower Npt2a (molecular weight: ∼75 kDa) expression in control mice compared with vehicle (Veh), an effect absent in NHE3TAL-KO mice. B–F: no differences were observed in the levels of electrogenic Na+-bicarbonate cotransporter 1 (NBCe1; B; molecular weight: ∼116 kDa), anion exchanger 2 (AE2; C; molecular weight: ∼180 kDa), nongastric H+-K+-ATPase (D; molecular weight: ∼100 kDa), H+-ATPase β1-subunit (E; molecular weight: ∼57 kDa), or the Cl/HCO3 exchanger pendrin (F; molecular weight: ∼95 kDa) between treatment or genotype. Data were analyzed by two-way ANOVA followed by a Tukey’s multiple comparison test. In addition to single data, means ± SE values of n = 6 male mice/genotype are shown. §P < 0.05 vehicle same genotype.

DISCUSSION

Studies to define the general role of NHE3 in the TAL were hampered by the lack of a suitable animal model. To navigate this problem, we generated NHE3TAL-KO mice and used this model to determine the contribution of NHE3 in the TAL to renal acid-base/electrolyte/water homeostasis as well as its contribution to furosemide-induced urinary acidification. In this model, NHE3 mRNA and protein were greatly reduced in the medulla, with immunohistochemistry confirming that this is due to an almost complete absence of NHE3 in the TAL, whereas proximal tubule NHE3 expression is unaffected. Despite the Cre-driver mouse line we used being tamoxifen-inducible, it was previously reported that a small number of tdTomato-positive cells exist within a subpopulation of uromodulin-positive cells (51), suggesting tamoxifen-independent Cre-mediated recombination. Consequently, we have not studied the phenotype of NHE3 knockout before and after tamoxifen administration.

Under standard (baseline) conditions, there were no significant differences between control and NHE3TAL-KO mice in body weight, food intake, and plasma concentrations of Na+, K+, and Cl. Fluid intake tended to be higher, and when combined with the observed lower urine osmolality in NHE3TAL-KO mice without any differences in kidney function, plasma osmolality, or hematocrit, it suggests that NHE3TAL-KO mice have a mild defect in the renal handling of water but remain in fluid balance. Mice with knockout of NHE3 along the entire nephron also have a mild urinary concentrating defect (7); however, when NHE3TAL-KO mice were challenged by water deprivation, no phenotype was detected, implying that other mechanisms are fully capable to compensate for the lack of NHE3 in the TAL under these conditions. Furthermore, under baseline conditions, blood and urine pH in NHE3TAL-KO mice were not different from controls, suggesting that lack of NHE3 in the TAL does not cause an acid-base phenotype. This is in contrast to mice deficient in NHE3 along the entire nephron, which have a higher urinary pH (7). This suggests that NHE3 activity in the proximal tubule is more important for renal HCO3 reabsorption than NHE3 activity in the TAL.

In addition to being a diuretic, furosemide causes urinary acidification (52). The mechanism for this was originally proposed to be dependent on epithelial Na+ channel-induced activation of vacuolar H+-ATPase (B1 subunit) in the connecting tubule (53, 54). In rats, 7 days of furosemide treatment increased H+-ATPase B1 subunit abundance in the cortex and outer medulla (∼1.8-fold and ∼2.4-fold, respectively) without affecting mRNA expression (55). However, we did not observe changes in the protein abundance of the H+-ATPase B1 subunit or other transporters/pumps involved in acid-base regulation after 8 days of furosemide treatment. Previous studies have identified that in adult NHE3−/− mice Na+/H+ exchanger isoform 8 expression is increased (56). Our data expand this knowledge and show that the ubiquitously expressed NHE1 isoform is not increased in the absence of NHE3 in the TAL and not regulated by furosemide. Whether other Na+/H+ exchanger isoforms compensate in NHE3TAL-KO mice remains to be determined.

More recently, the effects of furosemide have been suggested to be mediated by NHE3 activity in the TAL, with urinary acidification markedly reduced after pharmacological NHE3 inhibition because of a decrease in intracellular Na+ concentration, increasing the driving force for NHE3 (18). Our novel mouse line provided us the ideal opportunity to test this mechanism for furosemide-induced urinary acidification and has provided significant new insights. In acute experiments under anesthesia, furosemide induced a rapid and sustained decrease of urinary pH in control mice. NHE3IEC-KO mice showed a response of similar magnitude. However, this acidification was followed by an immediate recovery to levels not significantly different from baseline, indicating that NHE3 activity is required for sustained furosemide-induced urinary acidification. In control mice, doses of >1 mg·kg−1 did not result in an additional effect on urinary acidification despite having a significant effect on urinary flow rate and urinary Cl excretion. In contrast to the acute experiments, chronic furosemide administration had no effect on urinary acidification. The reason for this remains elusive, but we do not see furosemide-induced urinary acidification during short-term (3-h) studies when mice are housed in metabolic cages (unpublished observations). A similar lack of urinary acidification in response to furosemide has been observed by others. For example, acute studies in humans, 15-min collection after furosemide administration, showed an increase in urinary pH in conjunction with an increase in HCO3 excretion (57, 58). This effect was suggested to be caused by furosemide-inhibiting carbonic anhydrase (50, 58). Other possibilities include changes in titratable acid excretion and/or ammonium trapping (59). Taken together, furosemide-induced urinary acidification might depend on species and/or other experimental conditions.

Despite a lack of furosemide-induced urinary acidification from spontaneous urine collections in our long-term experiments, control mice developed metabolic alkalosis. However, an effect of furosemide on acid-base status was absent in NHE3TAL-KO mice, highlighting the importance of this segment for long-term acid-base regulation in response to furosemide. Metabolic alkalosis is usually associated with hypokalemia, which can be compensated for by muscle and kidneys (60). Furosemide causes flow-induced K+ secretion, which requires functional epithelial Na+ channels as a prerequisite (61), which might further worsen hypokalemia. Consistent with this, in control mice, plasma K+ was significantly lower in response to furosemide compared with vehicle treatment. In contrast, plasma K+ remained unaffected in NHE3TAL-KO mice, possibly because of a lack of furosemide-induced changes in blood pH. We hypothesize that the exchange of H+ versus K+ in muscle cells is the dominating factor for hypokalemia because similar urinary flow rates and urine osmolalities in response to furosemide were observed between genotypes. However, our study cannot completely exclude that changes in the expression levels/activity of the renal outer medullary K+ channel (62) and/or maxi K+ channel exist (63) between the genotypes that affect the kaliuretic response. The observed higher urinary K+-to-creatinine ratio in NHE3TAL-KO mice under baseline conditions could also relate to flow-induced K+ secretion considering the lower urine osmolality and trend for greater fluid intake, both indicators of a higher urinary flow rate.

Furosemide treatment is associated with an increase in urinary Ca2+ excretion (50, 64, 65). Mechanistically, the furosemide-induced increase in Ca2+ excretion is proposed to be due to a reduction in the lumen-positive transepithelial voltage and consequently a reduction in paracellular Ca2+ (and Mg2+) transport in the TAL (66). In addition, increased Na+ and Cl reabsorption in the distal convoluted tubule is associated with decreased Ca2+ reabsorption. Regarding the latter, it was shown that the Ca2+-sensing receptor can increase NCC activity via with-no-lysine kinases acting upon the Ste20-related proline alanine-rich kinase pathway resulting in inhibition of Ca2+ reabsorption (67). The furosemide-induced calciuresis is associated with an increased risk of hyperparathyroidism in patients with normal kidney function (68) and with chronic kidney disease (69). A reduction in plasma Ca2+ increases parathyroid hormone, which can reduce Npt2a abundance in response to furosemide treatment; however, if this is the primary effect or if off-target effects on carbonic anhydrase in the proximal tubule (70) play a role remains to be determined. In humans, furosemide increased urinary Pi excretion in the first hour after administration, which was followed by a reduction of Pi excretion in the subsequent hour if volume contraction was not corrected (71). Clearance experiments in dogs showed that furosemide increased fractional Pi excretion, an effect that was dependent on parathyroid hormone (72). Our data expand this previous knowledge by showing that Npt2a abundance is reduced by ∼50% in control mice chronically treated with furosemide. In contrast, NHE3TAL-KO mice do not have significantly lower Npt2a abundance or calciuresis after furosemide. The reason(s) for this is unknown, but it could imply that NHE3 in the TAL plays a role in Ca2+ reabsorption. The fact that NHE3TAL-KO mice lack the sustained urinary acidification in response to furosemide and that the Ca2+-sensing receptor is altered by extracellular pH (73) could have secondary consequences on claudin 14 and 16, both regulators of paracellular cation permeability in the TAL (74).

Furosemide treatment induces a compensatory increase in NKCC2 expression (75), a mechanism speculated to contribute to diuretic resistance (76). Our results confirm this phenomenon, with chronic furosemide treatment increasing the abundance of NKCC2 (75) and NCC (77, 78) as well as phosphorylation of NCC (20, 67, 78, 79). In addition, the growth of the distal tubule has been described as a response to increased NaCl delivery into the distal tubule (80, 81). Despite the possible increase in distal convoluted tubule mass, we did not find greater Na+-K+-ATPase expression in whole kidneys in response to furosemide treatment, which is consistent with unaffected mRNA expression in response to furosemide treatment in Wistar rats (82). However, another study found that Na+-K+-ATPase activity was increased in the distal convoluted tubule and collecting duct of furosemide-treated rats (83).

In summary, we have generated a novel TAL-specific NHE3 knockout model. Using this tool, we were able to demonstrate that under normal conditions, the contribution of NHE3 in the TAL for acid-base regulation is minor or can be compensated for in distal nephron segments. We also provide new evidence for a role of NHE3 in the TAL for furosemide-induced acidification, with acute urinary acidification unaffected in NHE3TAL-KO mice but an immediate recovery to baseline levels, suggesting a role for NHE3 in sustained urinary acidification. Consistent with this, NHE3TAL-KO mice lack metabolic alkalosis in response to long-term treatment with furosemide. Of note, it is possible that under certain conditions (low HCO3 or metabolic acidosis), NHE3 in the TAL may play a permissive role for NaCl and acid-base homeostasis that is not observed under baseline conditions or when NKCC2 is maximally inhibited by furosemide.

SUPPLEMENTAL DATA

GRANTS

This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Grant 1R01DK110621 (to T.R.), Veterans Affairs Merit Review Award IBX004968A (to T.R.), and American Heart Association Transformational Research Award 19TPA34850116 (to T.R.). Financial support for this work was also provided by NIDDK Diabetic Complications Consortium (RRID: SCR_001415, www.diacomp.org) Grants DK076169 and DK115255 (to T.R.). L.T. was supported by American Heart Association Postdoctoral Fellowship 828731. Further funding to R.A.F. was provided by the Novo Nordisk Foundation (NNF21OC0067647, NNF17OC0029724, and NNF19OC0058439), the Leducq Foundation (17CVD05), and the Independent Research Fund Denmark (0134-00018B).

DISCLOSURES

T.R. and R.A.F. are Associate Editors of the American Journal of Physiology-Renal Physiology but are not involved and did not have access to information regarding the peer-review process or final disposition of this article. An alternate editor oversaw the peer-review and decision-making process for this article. None of the other authors has any conflicts of interest, financial or otherwise, to disclose.

AUTHOR CONTRIBUTIONS

J.X. and T.R. conceived and designed research; J.X., L.T., J.A.D.R., R.A.F., and T.R. performed experiments; J.X., L.T., J.A.D.R., R.A.F., and T.R. analyzed data; J.X., L.T., J.A.D.R., R.A.F., and T.R. interpreted results of experiments; J.X., R.A.F., and T.R. prepared figures; J.A.D.R. and T.R. drafted manuscript; J.X., L.T., J.A.D.R., R.A.F., and T.R. edited and revised manuscript; J.X., L.T., J.A.D.R., R.A.F., and T.R. approved final version of manuscript.

ACKNOWLEDGMENTS

The authors thank Debra Fong for technical assistance.

REFERENCES

  • 1.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]
  • 2.Xue J, Thomas L, Tahmasbi M, Valdez A, Dominguez Rieg JA, Fenton RA, Rieg T. An inducible intestinal epithelial cell-specific NHE3 knockout mouse model mimicking congenital sodium diarrhea. Clin Sci (Lond) 134: 941–953, 2020. doi: 10.1042/CS20200065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Amemiya M, Loffing J, Lotscher 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]
  • 4.Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Busslinger M, Dominguez Rieg JA, Rieg T. Caffeine-induced diuresis and natriuresis is independent of renal tubular NHE3. Am J Physiol Renal Physiol 308: F1409–F1420, 2015. doi: 10.1152/ajprenal.00129.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Biemesderfer D, Pizzonia J, Abu-Alfa A, Exner M, Reilly R, Igarashi P, Aronson PS. NHE3: a Na+/H+ exchanger isoform of renal brush border. Am J Physiol Renal Physiol 265: F736–F742, 1993. doi: 10.1152/ajprenal.1993.265.5.F736. [DOI] [PubMed] [Google Scholar]
  • 6.Vallon V, Schwark JR, Richter K, Hropot M. Role of Na+/H+ exchanger NHE3 in nephron function: micropuncture studies with S3226, an inhibitor of NHE3. Am J Physiol Renal Physiol 278: F375–379, 2000. doi: 10.1152/ajprenal.2000.278.3.F375. [DOI] [PubMed] [Google Scholar]
  • 7.Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Dominguez Rieg JA, Rieg T. Renal tubular NHE3 is required in the maintenance of water and sodium chloride homeostasis. Kidney Int 92: 397–414, 2017. doi: 10.1016/j.kint.2017.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lee JW, Chou CL, Knepper MA. Deep sequencing in microdissected renal tubules identifies nephron segment-specific transcriptomes. J Am Soc Nephrol 26: 2669–2677, 2015. doi: 10.1681/ASN.2014111067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen L, Chou CL, Knepper MA. A comprehensive map of mRNAs and their isoforms across all 14 renal tubule segments of mouse. J Am Soc Nephrol 32: 897–912, 2021. doi: 10.1681/ASN.2020101406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lewis S, Chen L, Raghuram V, Khundmiri SJ, Chou CL, Yang CR, Knepper MA. “SLC-omics” of the kidney: solute transporters along the nephron. Am J Physiol Cell Physiol 321: C507–C518, 2021. doi: 10.1152/ajpcell.00197.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Limbutara K, Chou CL, Knepper MA. Quantitative proteomics of all 14 renal tubule segments in rat. J Am Soc Nephrol 31: 1255–1266, 2020. doi: 10.1681/ASN.2020010071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gunaratne R, Braucht DW, Rinschen MM, Chou CL, Hoffert JD, Pisitkun T, Knepper MA. Quantitative phosphoproteomic analysis reveals cAMP/vasopressin-dependent signaling pathways in native renal thick ascending limb cells. Proc Natl Acad Sci USA 107: 15653–15658, 2010. doi: 10.1073/pnas.1007424107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kurashima K, Yu FH, Cabado AG, Szabo EZ, Grinstein S, Orlowski J. Identification of sites required for down-regulation of Na+/H+ exchanger NHE3 activity by cAMP-dependent protein kinase. phosphorylation-dependent and -independent mechanisms. J Biol Chem 272: 28672–28679, 1997. doi: 10.1074/jbc.272.45.28672. [DOI] [PubMed] [Google Scholar]
  • 14.Zhao H, Wiederkehr MR, Fan L, Collazo RL, Crowder LA, Moe OW. Acute inhibition of Na/H exchanger NHE-3 by cAMP. Role of protein kinase a and NHE-3 phosphoserines 552 and 605. J Biol Chem 274: 3978–3987, 1999. doi: 10.1074/jbc.274.7.3978. [DOI] [PubMed] [Google Scholar]
  • 15.Kwon T-H, Nielsen J, Kim Y-H, Knepper MA, Frøkiær J, Nielsen S. Regulation of sodium transporters in the thick ascending limb of rat kidney: response to angiotensin II. Am J Physiol Renal Physiol 285: F152–F165, 2003. doi: 10.1152/ajprenal.00307.2002. [DOI] [PubMed] [Google Scholar]
  • 16.Laghmani K, Chambrey R, Froissart M, Bichara M, Paillard M, Borensztein P. Adaptation of NHE-3 in the rat thick ascending limb: effects of high sodium intake and metabolic alkalosis. Am J Physiol 276: F18–F26, 1999. doi: 10.1152/ajprenal.1999.276.1.F18. [DOI] [PubMed] [Google Scholar]
  • 17.Wang T, Hropot M, Aronson PS, Giebisch G. Role of NHE isoforms in mediating bicarbonate reabsorption along the nephron. Am J Physiol Renal Physiol 281: F1117–F1122, 2001. doi: 10.1152/ajprenal.2001.281.6.F1117. [DOI] [PubMed] [Google Scholar]
  • 18.de Bruijn PI, Larsen CK, Frische S, Himmerkus N, Praetorius HA, Bleich M, Leipziger J. Furosemide-induced urinary acidification is caused by pronounced H+ secretion in the thick ascending limb. Am J Physiol Renal Physiol 309: F146–F153, 2015. doi: 10.1152/ajprenal.00154.2015. [DOI] [PubMed] [Google Scholar]
  • 19.Li HC, Du Z, Barone S, Rubera I, McDonough AA, Tauc M, Zahedi K, Wang T, Soleimani M. Proximal tubule specific knockout of the Na+/H+ exchanger NHE3: effects on bicarbonate absorption and ammonium excretion. J Mol Med (Berl) 91: 951–963, 2013. doi: 10.1007/s00109-013-1015-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tokonami N, Takata T, Beyeler J, Ehrbar I, Yoshifuji A, Christensen EI, Loffing J, Devuyst O, Olinger EG. Uromodulin is expressed in the distal convoluted tubule, where it is critical for regulation of the sodium chloride cotransporter NCC. Kidney Int 94: 701–715, 2018. doi: 10.1016/j.kint.2018.04.021. [DOI] [PubMed] [Google Scholar]
  • 21.Poulsen SB, Kristensen TB, Brooks HL, Kohan DE, Rieg T, Fenton RA. Role of adenylyl cyclase 6 in the development of lithium-induced nephrogenic diabetes insipidus. JCI Insight 2: e91042, 2017. doi: 10.1172/jci.insight.91042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Thomas L, Xue J, Murali SK, Fenton RA, Dominguez Rieg JA, Rieg T. Pharmacological Npt2a inhibition causes phosphaturia and reduces plasma phosphate in mice with normal and reduced kidney function. J Am Soc Nephrol 30: 2128–2139, 2019. doi: 10.1681/ASN.2018121250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Vallon V, Rieg T, Ahn SY, Wu W, Eraly SA, Nigam SK. Overlapping in vitro and in vivo specificities of the organic anion transporters OAT1 and OAT3 for loop and thiazide diuretics. Am J Physiol Renal Physiol 294: F867–F873, 2008. doi: 10.1152/ajprenal.00528.2007. [DOI] [PubMed] [Google Scholar]
  • 24.Dominguez Rieg JA, Burt JM, Ruth P, Rieg T. P2Y2 receptor activation decreases blood pressure via intermediate conductance potassium channels and connexin 37. Acta Physiol (Oxf) 213: 628–641, 2015. doi: 10.1111/apha.12446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rieg T, Tang T, Murray F, Schroth J, Insel PA, Fenton RA, Hammond HK, Vallon V. Adenylate cyclase 6 determines cAMP formation and aquaporin-2 phosphorylation and trafficking in inner medulla. J Am Soc Nephrol 21: 2059–2068, 2010. doi: 10.1681/ASN.2010040409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Xue J, Thomas L, Dominguez Rieg JA, Fenton RA, Rieg T. Genetic deletion of connexin 37 causes polyuria and polydipsia. PLoS One 15: e0244251, 2020. doi: 10.1371/journal.pone.0244251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fenton RA, Murray F, Dominguez Rieg JA, Tang T, Levi M, Rieg T. Renal phosphate wasting in the absence of adenylyl cyclase 6. J Am Soc Nephrol 25: 2822–2834, 2014. doi: 10.1681/ASN.2013101102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rieg T. A high-throughput method for measurement of glomerular filtration rate in conscious mice. J Vis Exp 75: e50330, 2013. doi: 10.3791/50330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Fenton RA, Murali SK, Kaji I, Akiba Y, Kaunitz JD, Kristensen TB, Poulsen SB, Dominguez Rieg JA, Rieg T. Adenylyl cyclase 6 expression is essential for cholera toxin-induced diarrhea. J Infect Dis 220: 1719–1728, 2019. doi: 10.1093/infdis/jiz013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kim GH, Ecelbarger C, Knepper MA, Packer RK. Regulation of thick ascending limb ion transporter abundance in response to altered acid/base intake. J Am Soc Nephrol 10: 935–942, 1999. doi: 10.1681/ASN.V105935. [DOI] [PubMed] [Google Scholar]
  • 31.Ecelbarger CA, Terris J, Hoyer JR, Nielsen S, Wade JB, Knepper MA. Localization and regulation of the rat renal Na+-K+-2Cl cotransporter, BSC-1. Am J Physiol Renal Physiol 271: F619–F628, 1996. doi: 10.1152/ajprenal.1996.271.3.F619. [DOI] [PubMed] [Google Scholar]
  • 32.Pedersen NB, Hofmeister MV, Rosenbaek LL, Nielsen J, Fenton RA. Vasopressin induces phosphorylation of the thiazide-sensitive sodium chloride cotransporter in the distal convoluted tubule. Kidney Int 78: 160–169, 2010. doi: 10.1038/ki.2010.130. [DOI] [PubMed] [Google Scholar]
  • 33.Poulsen SB, Marin De Evsikova C, Murali SK, Praetorius J, Chern Y, Fenton RA, Rieg T. Adenylyl cyclase 6 is required for maintaining acid-base homeostasis. Clin Sci (Lond) 132: 1779–1796, 2018. doi: 10.1042/CS20180060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Frische S, Zolotarev AS, Kim YH, Praetorius J, Alper S, Nielsen S, Wall SM. AE2 isoforms in rat kidney: immunohistochemical localization and regulation in response to chronic NH4Cl loading. Am J Physiol Renal Physiol 286: F1163–1170, 2004. doi: 10.1152/ajprenal.00409.2003. [DOI] [PubMed] [Google Scholar]
  • 35.Kim YH, Kwon TH, Frische S, Kim J, Tisher CC, Madsen KM, Nielsen S. Immunocytochemical localization of pendrin in intercalated cell subtypes in rat and mouse kidney. Am J Physiol Renal Physiol 283: F744–F754, 2002. doi: 10.1152/ajprenal.00037.2002. [DOI] [PubMed] [Google Scholar]
  • 36.Maunsbach AB, Vorum H, Kwon T-H, Nielsen S, Simonsen B, Choi I, Schmitt BM, Boron WF, AALKJæR C. Immunoelectron microscopic localization of the electrogenic Na/HCO3 cotransporter in rat and ambystoma kidney. J Am Soc Nephrol 11: 2179–2189, 2000. doi: 10.1681/ASN.V11122179. [DOI] [PubMed] [Google Scholar]
  • 37.Swarts HG, Koenderink JB, Willems PH, De Pont JJ. The non-gastric H, K-ATPase is oligomycin-sensitive and can function as an H+, NH4+ ATPase. J Biol Chem 280: 33115–33122, 2005. doi: 10.1074/jbc.M504535200. [DOI] [PubMed] [Google Scholar]
  • 38.Kortenoeven MLA, Esteva-Font C, Dimke H, Poulsen SB, Murali SK, Fenton RA. High dietary potassium causes ubiquitin-dependent degradation of the kidney sodium-chloride cotransporter. J Biol Chem 297: 100915, 2021. doi: 10.1016/j.jbc.2021.100915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Breusegem SY, Takahashi H, Giral-Arnal H, Wang X, Jiang T, Verlander JW, Wilson P, Miyazaki-Anzai S, Sutherland E, Caldas Y, Blaine JT, Segawa H, Miyamoto K, Barry NP, Levi M. Differential regulation of the renal sodium-phosphate cotransporters NaPi-IIa, NaPi-IIc, and PiT-2 in dietary potassium deficiency. Am J Physiol Renal Physiol 297: F350–F361, 2009. doi: 10.1152/ajprenal.90765.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lanzano L, Lei T, Okamura K, Giral H, Caldas Y, Masihzadeh O, Gratton E, Levi M, Blaine J. Differential modulation of the molecular dynamics of the type IIa and IIc sodium phosphate cotransporters by parathyroid hormone. Am J Physiol Cell Physiol 301: C850–C861, 2011. doi: 10.1152/ajpcell.00412.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Takahashi N, Chernavvsky DR, Gomez RA, Igarashi P, Gitelman HJ, Smithies O. Uncompensated polyuria in a mouse model of Bartter's syndrome. Proc Natl Acad Sci USA 97: 5434–5439, 2000. doi: 10.1073/pnas.090091297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rosenbaek LL, Kortenoeven ML, Aroankins TS, Fenton RA. Phosphorylation decreases ubiquitylation of the thiazide-sensitive cotransporter NCC and subsequent clathrin-mediated endocytosis. J Biol Chem 289: 13347–13361, 2014. doi: 10.1074/jbc.M113.543710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Christensen BM, Kim YH, Kwon TH, Nielsen S. Lithium treatment induces a marked proliferation of primarily principal cells in rat kidney inner medullary collecting duct. Am J Physiol Renal Physiol 291: F39–F48, 2006. doi: 10.1152/ajprenal.00383.2005. [DOI] [PubMed] [Google Scholar]
  • 44.Frische S, Chambrey R, Trepiccione F, Zamani R, Marcussen N, Alexander RT, Skjodt K, Svenningsen P, Dimke H. H+-ATPase B1 subunit localizes to thick ascending limb and distal convoluted tubule of rodent and human kidney. Am J Physiol Renal Physiol 315: F429–F444, 2018. doi: 10.1152/ajprenal.00539.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Stuart-Tilley A, Sardet C, Pouyssegur J, Schwartz MA, Brown D, Alper SL. Immunolocalization of anion exchanger AE2 and cation exchanger NHE-1 in distinct adjacent cells of gastric mucosa. Am J Physiol 266: C559–C568, 1994. doi: 10.1152/ajpcell.1994.266.2.C559. [DOI] [PubMed] [Google Scholar]
  • 46.Alper SL, Stuart-Tilley AK, Biemesderfer D, Shmukler BE, Brown D. Immunolocalization of AE2 anion exchanger in rat kidney. Am J Physiol 273: F601–F614, 1997[Erratum inAm J Physiol274: 7433, 1998]. doi: 10.1152/ajprenal.1997.273.4.F601. [DOI] [PubMed] [Google Scholar]
  • 47.Lee HW, Verlander JW, Shull GE, Harris AN, Weiner ID. Acid-base effects of combined renal deletion of NBCe1-A and NBCe1-B. Am J Physiol Renal Physiol 322: F208–F224, 2022. doi: 10.1152/ajprenal.00358.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wu Q, Poulsen SB, Murali SK, Grimm PR, Su XT, Delpire E, Welling PA, Ellison DH, Fenton RA. Large-scale proteomic assessment of urinary extracellular vesicles highlights their reliability in reflecting protein changes in the kidney. J Am Soc Nephrol 32: 2195–2209, 2021. doi: 10.1681/ASN.2020071035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Weiner ID, Verlander JW. Role of NH3 and NH4+ transporters in renal acid-base transport. Am J Physiol Renal Physiol 300: F11–F23, 2011. doi: 10.1152/ajprenal.00554.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lee CT, Chen HC, Lai LW, Yong KC, Lien YH. Effects of furosemide on renal calcium handling. Am J Physiol Renal Physiol 293: F1231–F1237, 2007. doi: 10.1152/ajprenal.00038.2007. [DOI] [PubMed] [Google Scholar]
  • 51.Guo J, Jill McMahon JL, McMahon AP. Umod IRES CRE-ERT2 Allele Characterization. https://www.gudmap.org/Docs/Mouse_Strains/UmodIRESCE_strain_report_051017.FNa_APM.J.pdf. [Accessed 9 June 2022].
  • 52.Hropot M, Fowler N, Karlmark B, Giebisch G. Tubular action of diuretics: distal effects on electrolyte transport and acidification. Kidney Int 28: 477–489, 1985. doi: 10.1038/ki.1985.154. [DOI] [PubMed] [Google Scholar]
  • 53.Kovacikova J, Winter C, Loffing-Cueni D, Loffing J, Finberg KE, Lifton RP, Hummler E, Rossier B, Wagner CA. The connecting tubule is the main site of the furosemide-induced urinary acidification by the vacuolar H+-ATPase. Kidney Int 70: 1706–1716, 2006. doi: 10.1038/sj.ki.5001851. [DOI] [PubMed] [Google Scholar]
  • 54.Finberg KE, Wagner CA, Bailey MA, Paunescu TG, Breton S, Brown D, Giebisch G, Geibel JP, Lifton RP. The B1-subunit of the H+ ATPase is required for maximal urinary acidification. Proc Natl Acad Sci USA 102: 13616–13621, 2005. doi: 10.1073/pnas.0506769102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Na KY, Kim GH, Joo KW, Lee JW, Jang HR, Oh YK, Jeon US, Chae SW, Knepper MA, Han JS. Chronic furosemide or hydrochlorothiazide administration increases H+-ATPase B1 subunit abundance in rat kidney. Am J Physiol Renal Physiol 292: F1701–F1709, 2007. doi: 10.1152/ajprenal.00270.2006. [DOI] [PubMed] [Google Scholar]
  • 56.Baum M, Twombley K, Gattineni J, Joseph C, Wang L, Zhang Q, Dwarakanath V, Moe OW. Proximal tubule Na+/H+ exchanger activity in adult NHE8−/−, NHE3−/−, and NHE3−/−/NHE8−/− mice. Am J Physiol Renal Physiol 303: F1495–F1502, 2012. doi: 10.1152/ajprenal.00415.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Stein JH, Wilson CB, Kirkendall WM. Differences in the acute effects of furosemide and ethacrynic acid in man. J Lab Clin Med 71: 654–665, 1968. [PubMed] [Google Scholar]
  • 58.Puschett JB, Goldberg M. The acute effects of furosemide on acid and electrolyte excretion in man. J Lab Clin Med 71: 666–677, 1968. [PubMed] [Google Scholar]
  • 59.Weiner ID, Verlander JW. Renal ammonia metabolism and transport. Compr Physiol 3: 201–220, 2013. doi: 10.1002/cphy.c120010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Aronson PS, Giebisch G. Effects of pH on potassium: new explanations for old observations. J Am Soc Nephrol 22: 1981–1989, 2011. doi: 10.1681/ASN.2011040414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ayasse N, Berg P, Leipziger J, Sorensen MV. ENaC expression correlates with the acute furosemide-induced K+ excretion. Physiol Rep 9: e14668, 2021. doi: 10.14814/phy2.14668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Jensen IS, Larsen CK, Leipziger J, Sorensen MV. Na+ dependence of K+-induced natriuresis, kaliuresis and Na+/Cl cotransporter dephosphorylation. Acta Physiol (Oxf) 218: 49–61, 2016. doi: 10.1111/apha.12707. [DOI] [PubMed] [Google Scholar]
  • 63.Rieg T, Vallon V, Sausbier M, Sausbier U, Kaissling B, Ruth P, Osswald H. The role of the BK channel in potassium homeostasis and flow-induced renal potassium excretion. Kidney Int 72: 566–573, 2007. doi: 10.1038/sj.ki.5002369. [DOI] [PubMed] [Google Scholar]
  • 64.Bushinsky DA, Favus MJ, Langman CB, Coe FL. Mechanism of chronic hypercalciuria with furosemide: increased calcium absorption. Am J Physiol Renal Physiol 251: F17–F24, 1986. doi: 10.1152/ajprenal.1986.251.1.F17. [DOI] [PubMed] [Google Scholar]
  • 65.Hanson RC, White JB, Gomoll AW. Acute and chronic diuretic-induced alterations in urinary sodium and calcium excretion in the conscious rat. Miner Electrolyte Metab 8: 314–324, 1982. [PubMed] [Google Scholar]
  • 66.Wittner M, Mandon B, Roinel N, de Rouffignac C, Di Stefano A. Hormonal stimulation of Ca2+ and Mg2+ transport in the cortical thick ascending limb of Henle's loop of the mouse: evidence for a change in the paracellular pathway permeability. Pflugers Arch 423: 387–396, 1993. doi: 10.1007/BF00374932. [DOI] [PubMed] [Google Scholar]
  • 67.Bazua-Valenti S, Rojas-Vega L, Castaneda-Bueno M, Barrera-Chimal J, Bautista R, Cervantes-Perez LG, Vazquez N, Plata C, Murillo-de-Ozores AR, Gonzalez-Mariscal L, Ellison DH, Riccardi D, Bobadilla NA, Gamba G. The calcium-sensing receptor increases activity of the renal NCC through the WNK4-SPAK pathway. J Am Soc Nephrol 29: 1838–1848, 2018. doi: 10.1681/ASN.2017111155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Corapi KM, McMahon GM, Wenger JB, Seifter JL, Bhan I. Association of loop diuretic use with higher parathyroid hormone levels in patients with normal renal function. JAMA Intern Med 175: 137–138, 2015. doi: 10.1001/jamainternmed.2014.5857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Vasco RF, Moyses RM, Zatz R, Elias RM. Furosemide increases the risk of hyperparathyroidism in chronic kidney disease. Am J Nephrol 43: 421–430, 2016. doi: 10.1159/000446449. [DOI] [PubMed] [Google Scholar]
  • 70.Carta F, Supuran CT. Diuretics with carbonic anhydrase inhibitory action: a patent and literature review (2005–2013). Expert Opin Ther Pat 23: 681–691, 2013. doi: 10.1517/13543776.2013.780598. [DOI] [PubMed] [Google Scholar]
  • 71.Steele TH. Dual effect of potent diuretics on renal handling of phosphate in man. Metabolism 20: 749–761, 1971. doi: 10.1016/s0026-0495(71)80005-7. [DOI] [PubMed] [Google Scholar]
  • 72.Haas JA, Larson MV, Marchand GR, Lang FC, Greger RF, Knox FG. Phosphaturic effect of furosemide: role of PTH and carbonic anhydrase. Am J Physiol Renal Physiol 232: F105–F110, 1977. doi: 10.1152/ajprenal.1977.232.2.F105. [DOI] [PubMed] [Google Scholar]
  • 73.Quinn SJ, Bai M, Brown EM. pH sensing by the calcium-sensing receptor. J Biol Chem 279: 37241–37249, 2004. doi: 10.1074/jbc.M404520200. [DOI] [PubMed] [Google Scholar]
  • 74.Toka HR, Al-Romaih K, Koshy JM, DiBartolo S, 3rd, Kos CH, Quinn SJ, Curhan GC, Mount DB, Brown EM, Pollak MR. Deficiency of the calcium-sensing receptor in the kidney causes parathyroid hormone-independent hypocalciuria. J Am Soc Nephrol 23: 1879–1890, 2012. doi: 10.1681/ASN.2012030323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Na KY, Oh YK, Han JS, Joo KW, Lee JS, Earm JH, Knepper MA, Kim GH. Upregulation of Na+ transporter abundances in response to chronic thiazide or loop diuretic treatment in rats. Am J Physiol Renal Physiol 284: F133–F143, 2003. doi: 10.1152/ajprenal.00227.2002. [DOI] [PubMed] [Google Scholar]
  • 76.Huang X, Dorhout Mees E, Vos P, Hamza S, Braam B. Everything we always wanted to know about furosemide but were afraid to ask. Am J Physiol Renal Physiol 310: F958–F971, 2016. doi: 10.1152/ajprenal.00476.2015. [DOI] [PubMed] [Google Scholar]
  • 77.van Angelen AA, van der Kemp AW, Hoenderop JG, Bindels RJ. Increased expression of renal TRPM6 compensates for Mg2+ wasting during furosemide treatment. Clin Kidney J 5: 535–544, 2012. doi: 10.1093/ckj/sfs140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Abdallah JG, Schrier RW, Edelstein C, Jennings SD, Wyse B, Ellison DH. Loop diuretic infusion increases thiazide-sensitive Na+/Cl-cotransporter abundance: role of aldosterone. J Am Soc Nephrol 12: 1335–1341, 2001. doi: 10.1681/ASN.V1271335. [DOI] [PubMed] [Google Scholar]
  • 79.Hunter RW, Ivy JR, Flatman PW, Kenyon CJ, Craigie E, Mullins LJ, Bailey MA, Mullins JJ. Hypertrophy in the distal convoluted tubule of an 11beta-hydroxysteroid dehydrogenase type 2 knockout model. J Am Soc Nephrol 26: 1537–1548, 2015. doi: 10.1681/ASN.2013060634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kaissling B, Bachmann S, Kriz W. Structural adaptation of the distal convoluted tubule to prolonged furosemide treatment. Am J Physiol Renal Physiol 248: F374–F381, 1985. doi: 10.1152/ajprenal.1985.248.3.F374. [DOI] [PubMed] [Google Scholar]
  • 81.Ellison DH, Velazquez H, Wright FS. Adaptation of the distal convoluted tubule of the rat. Structural and functional effects of dietary salt intake and chronic diuretic infusion. J Clin Invest 83: 113–126, 1989. doi: 10.1172/JCI113847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Merino A, Moreno G, Mercado A, Bobadilla NA, Gamba G. Na+:K+:ATPase mRNA expression in the kidney during adaptation to sodium intake and furosemide treatment. Arch Med Res 31: 486–492, 2000. doi: 10.1016/s0188-4409(00)00102-8. [DOI] [PubMed] [Google Scholar]
  • 83.Mernissi GE, Doucet A. Stimulation of Na-K-ATPase in the rat collecting tubule by two diuretics: furosemide and amiloride. Am J Physiol Renal Physiol 247: F485–F490, 1984. doi: 10.1152/ajprenal.1984.247.3.F485. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials


Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES