Abstract
The aldosterone-sensitive distal nephron (ASDN) exhibits axial heterogeneity in structure and function from the distal convoluted tubule to the medullary collecting duct. Ion and water transport is primarily divided between the cortex and medulla of the ASDN, respectively. Transcellular transport in this segment is highly regulated in health and disease and is integrated across different cell types. We currently lack an inexpensive, high-yield, and tractable technique to harvest and culture cells for the study of gene expression and physiological properties of mouse cortical ASDN. To address this need, we harvested tubules bound to Dolichos biflorus agglutinin lectin-coated magnetic beads from the kidney cortex and characterized these cell preparations. We determined that these cells are enriched for markers of distal convoluted tubule, connecting tubule, and cortical collecting duct, including principal and intercalated cells. In primary culture, these cells develop polarized monolayers with high resistance (1,000-1,500 Ω * cm2) and maintain expression and activity of key channels. These cells demonstrate an amiloride-sensitive short-circuit current that can be enhanced with aldosterone and maintain measurable potassium and anion secretion. Our method can be easily adopted to study the biology of the ASDN and to investigate phenotypic differences between wild-type and transgenic mouse models.
Keywords: distal nephron, distal convoluted tubule, collecting duct, principal cells, intercalated cells
the aldosterone-sensitive distal nephron (ASDN) is defined as the distal convoluted tubule through the connecting tubule to the cortical and medullary collecting duct and is responsible for several important ion transport pathways. The distal convoluted tubule transports sodium (Na+), potassium (K+), magnesium, and calcium and is divided both functionally and anatomically into the distal convoluted tubule, part 1 and part 2 (DCT1 and DCT2)(64). The connecting tubule (CNT) and cortical collecting duct (CCD) are composed mainly of two interdigitating cell types: 1) principal cells (PCs) and 2) intercalated cells (ICs). Principal cells are responsible for regulating Na+, K+, and water homeostasis (50), and the main carrier of Na+ in PCs is the epithelial sodium channel (ENaC). ENaC is stimulated by several hormones including aldosterone, is regulated in health and disease, and its importance in determining sodium and potassium balance is dependent on the activity of the Na-Cl cotransporter (NCC) in neighboring DCT cells (5). Principal cells also secrete potassium through renal outer medullary potassium (ROMK) and big potassium (BK) channels (50, 70, 71). Intercalated cells have traditionally been shown to mediate acid-base and potassium transport (23), but more recent studies demonstrate an additional role for these cells in mediating direct sodium reabsorption and in regulating sodium transport across PCs through ENaC (26, 58, 69). Thus the ASDN represents a tightly regulated, multifaceted, interdependent nephron segment.
Isolation and primary culture of nephron segments of the ASDN have primarily been restricted to the medullary collecting duct, and thus current methodologies limit better characterization of the roles of the cortical ASDN in physiological and pathophysiological states or from wild-type and transgenic mice (e.g., for gene expression, biochemical, cytological, and electrophysiological studies). Existing methodologies that isolate DCT or CCD include laser capture microdissection (12, 51) and transgenic mice expressing green fluorescent protein with Complex Object Parametric Analyzer and Sorter (COPAS) technology (36, 39). These techniques are not easily accessible to most laboratories and require technical expertise and/or specialized mouse strains. Laser capture microdissection also generates a low yield of cells that cannot be typically used for biochemistry. Moreover, existing technologies do not capture cells that represent the heterogeneity that is present in the ASDN. Thus more accessible methods that concomitantly produce a higher yield of primary cells from the ASDN are needed.
Different lectins bind specifically to different segments of the mammalian nephron (21, 30, 31) and thereby may be useful for isolation of cells from the ASDN. The Dolichos biflorus agglutinin (DBA) lectin has been used to localize and isolate collecting duct cells from the rodent medulla (17, 44, 62, 76, 77), but this reagent has not been previously validated for isolation of cortical cells. Herein, we describe the development and validation of a relatively inexpensive and simple method to harvest and culture epithelial cells from murine ASDN. We have performed biochemical, cytological, and electrophysiological assays to validate this method, and these preparations provide a model for quantitative, segment-specific studies.
MATERIALS AND METHODS
Animals.
Six- to eight-week-old C57BL/6 mice (Jackson Laboratories, Bar Harbor, ME) were provided food and water intake ad libitum and maintained through a 12:12-h light-dark cycle in a climate-controlled environment. The Institutional Animal Care and Use Committee at Stanford University approved the experiments, and mice were euthanized in accordance with the National Institutes of Health Guide for the Care and Use of Laboratory Animals.
Antibodies.
We employed rabbit polyclonal antibodies for detection of endogenous aquaporin-2 (gift of Robert Fenton, University of Aarhus); Tamm-Horsfall protein (Santa Cruz Biotechnology, Santa Cruz, CA); NCC (gift of David Ellison and James McCormick, Oregon Health Sciences University); zona occludin 1 (ZO-1; Invitrogen, Camarillo, CA); acetylated tubulin (Sigma-Aldrich, St. Louis, MO); TRPV5 (Everest Biotech, Ramona, CA); SGK1 (Sigma-Aldrich); and β-actin (EMD Millipore, Billerica, MA). We also generated and validated a mouse monoclonal antibody to detect mouse α-ENaC (Fig. 1).
Fig. 1.
Characterization of α-epithelial Na channel (ENaC) antibody. A: epitope derived from N-terminal intracellular sequence, L(74)IEFHRSYRE(83), (arrow) predicting migration of ∼30- and ∼90-kDa bands for cleaved and uncleaved α-ENaC, respectively. Cleavage occurs on extrafacial loop (scissors). B: detection of transfected α-ENaC in HEK293T cells (mock transfection, left; cotransfection with Nedd4-2, middle; and alone, right). C: detection (arrow) of endogenous uncleaved α-ENaC in immortalized mpkCCDc14 cells treated as indicated with vehicle or aldosterone (Aldo; 6 h) and vehicle, LY294002 (LY), or insulin for the final 2 h. Cleaved and uncleaved α-ENaC represented by arrows (black and gray, respectively). WCL, whole cell lysate.
Generation of α-ENaC antibody.
An immunogen was generated to the peptide sequence corresponding to amino acids 74–83, which is conserved from human to rat to mouse and doesn't share homology with β- or γ-ENaC. Peptide synthesis, purification, verification of peptide sequence (by mass spectrometry), conjugation to carrier (keyhole limpet hemocyanin), injection of mice (female BALB/c mice), formation of hybridoma cell lines, collection of ascites, and confirmation of specificity by ELISA were all carried out by a commercial vendor (Abmart, Beijing, China).
Immortalized cell culture.
Human embryonic kidney (HEK293T) cells and mouse polarized kidney cortical collecting duct (mpkCCDc14) cells were maintained and cultured as described previously (8). MpkCCDc14 cells were pretreated for 6 h with vehicle (100% ethanol) or 1 μM aldosterone (Sigma-Aldrich) with 2 final hours of vehicle (DMSO, Sigma-Aldrich), 50 μM LY294002 (EMD Millipore), or 100 nM insulin (Sigma-Aldrich).
Transient transfection of Nedd4-2 and α-ENaC.
HEK293T cells were transiently transfected as indicated with FLAG-tagged mouse Nedd4-2 with α (HA-tagged)-, β-, and γ-subunits of mouse ENaC as described previously (8). Twenty-four hours after transfection, cells were harvested in 500 μl lysis buffer containing protease and phosphatase inhibitors as described previously (18). Samples were separated by SDS-PAGE and immunoblotted with anti-α-ENaC antibody.
Western blotting: SDS-PAGE.
For immunoblots, samples lysed in lysis buffer {20 mM Tris·HCl, pH 8.5, 1% Nonidet P-40, 1% deoxycholate acid, 62.5% EDTA containing protease inhibitors [1 mM PMSF, 1 mM, benzamidine, and 1× Complete Protease Inhibitor Cocktail (Roche, Indianapolis, IN)] and phosphatase inhibitors [25 mM sodium fluoride, 2 μM microcystin-LR, and 1× Phosphatase-Inhibitor Cocktail Sets I and II (EMD Millipore)]} were separated by SDS-PAGE and transferred to polyvinylidene difluoride membranes and incubated with indicated antibodies and ECL (EMD Millipore). As indicated, we blotted endogenous 14-3-3 proteins as a loading control with anti-pan-14-3-3β (K-19) antibody (Santa Cruz Biotechnology).
Immunofluorescence microscopy.
Mice were anesthetized using inhaled isoflurane and perfused with chilled PBS (pH 7.4). Kidneys were removed, decapsulated, bisected sagittally, and incubated in 4% paraformaldehyde for 16 h overnight at 4°C. The tissue was then incubated in 30% sucrose in PBS for 24 h, embedded in Tissue-Tek optimal cutting temperature (OCT) compound (10.24% polyvinyl alcohol, 4.26% polyethylene glycol), frozen on liquid nitrogen-cooled N-methyl butane, sliced into 5-μm sections, mounted on superfrosted glass slides, and saved overnight at −80°C. We permeabilized slides in CSK buffer [50 mM NaCl, 300 mM sucrose, 10 mM PIPES (pH 6.8), 3 mM MgCl2, 0.5% (vol/vol) Triton X-100] for 15 s then washed twice in PBS for 5 min and incubated in blocking solution (PBS containing 50 mM NH4Cl, 25 mM l-lysine, 25 mM glycine, 0.2% BSA, 20% donkey serum) for 2 h at room temperature. Sections were then incubated with biotin-conjugated DBA (Vector Laboratories, Burlingame, CA) and/or primary antibody diluted in DAKO antibody diluent (Dako North America, Carpinteria, CA) overnight at 4°C. Sections were rinsed three times with PBS- 0.1% BSA and incubated with the appropriate secondary reagents [Alexa 488-conjugated donkey anti-rabbit antibody or Alexa 568-conjugated streptavidin (Life Technologies, Grand Island, NY)] for 2 h at room temperature. Slides were rinsed in PBS and mounted with Vectashield hardset media with 4,6-diamidino-2-phenylindole (DAPI; Vector Laboratories), and images were obtained using a confocal microscope (Leica SP2).
Isolation and culture of primary cell preparations.
We perfused mice with cold 1× PBS for 3–4 min, decapsulated the kidneys, coarsely dissected cortices, minced in modified Ringer's buffer (mRB; 118 mM NaCl, 16 mM H+-HEPES, 17 mM Na+-HEPES, 14 mM glucose, 3.2 mM KCl, 2.5 mM CaCl2, 1.8 mM Mg2SO4, and 1.8 mM KH2PO4), and pelleted at 175 g for 8 min in a swinging-bucket centrifuge with an A-4-62 rotor (Eppendorf 5810R, Hauppauge, NY) at room temperature. Pellets were resuspended in digestion buffer [0.2% collagenase (Worthington Biochemical, Lakewood, NJ); 0.2% hyaluronidase in mRB (Sigma-Aldrich)] and incubated at 200 rpm for 45 min in a shaker (Thermo Scientific MAKQ420HP, Waltham, MA) at 37°C. To mechanically disrupt tubular fragments, we passed the pellet 10–15 times through flame-tapered 9-in. pasteur pipettes and then reincubated it with DNAse I (Life Technologies) for 25 min at 37°C. Glomeruli and undigested tubules were captured on a 40-μm cell strainer (BD Biosciences, San Jose, CA) and washed. The sieved cells and tubules were then centrifuged three times at 28 g for 3 min, resuspended in mRB, and rocked with biotin-conjugated DBA and streptavidin-linked beads (Dynabeads M-280 Streptavidin, Life Technologies) for 15 min at room temperature (ATR Rotamix, Laurel, MD). Magnetically bound cells/tubules were washed three times and eluted twice with 150 mM α-N-acetyl galactosamine (NAG; Sigma-Aldrich) in mRB for 45 min at room temperature. We separated eluted cells and tubules from magnetic beads and recovered cells by brief centrifugation. We then washed and resuspended eluted cells and tubules in mRB and plated them in DMEM with 10% FBS and penicillin/streptomycin (100 IU/ml/100 μg/ml, Mediatech, Manassas, VA). After 48 h, cells were trypsinized, and we seeded 150,000 cells/cm2 onto collagen-coated filter cups in defined media [Renal Epithelial Cell Growth Basal Medium (REBM), Lonza, Allendale, NJ] with supplements, growth factors, and antibiotics (Bullet Kit: hydrocortisone, FBS, gentamicin, hInsulin, hEGF, transferrin, triiodothyronine, epinephrine, Lonza). After 7 days (5 days after replating), cell sheets were incubated for 24 h in basal REBM to deplete them of growth factors and supplements.
RNA extraction and semiquantitative PCR.
Total RNA from kidney, liver, DBA-bound, and unbound cell preparations was isolated using an RNeasy Mini kit per the manufacturer's instructions (Qiagen, Redwood City, CA). Reverse transcription was performed according to the manufacturer's instructions (SuperScript First-Strand Synthesis System for RT-PCR, Life Technologies) from 1 μg of total RNA using random hexamers. To study gene expression, we synthesized primers for nephron segment-specific markers using custom-designed (PrimerBlast tool, NCBI) or published sequences (Table 1). PCR reactions were performed with cDNA, 300 nM for each primer, and 2× Fast SYBR Green PCR master mix (Life Technologies). Fivefold serial dilution standard curves for all primers were made to test the efficiency of each primer pair. We performed reactions, including no-template controls, in triplicate in the StepOnePlus Real-Time PCR System (Life Technologies). To confirm specificity of amplicon products, a dissociation curve was performed for each PCR reaction. Complementary DNA from liver was used as a negative control for nephron segment-specific markers (data not shown). The ΔΔCt method was used to measure relative quantification (48) of each marker to β-actin.
Table 1.
Primers for semiquantitative PCR
| Transcript | Forward Primer | Reverse Primer |
|---|---|---|
| NPHS1 | TGCTGCCTTACCAAGTCCAG | GCTTCTGGGCCGGGTATTTT |
| SGLT2 | TGGCGGTGTCCGTGGCTTGG | CGGACACTGGAGGTGCCAGATAGC |
| GLUT2 (65) | ATCGCCCTCTGCTTCCAGTAC | GAACACGTAAGGCCCAAGGA |
| UTA2 | GCAATCTCGGGTTGCTTGGGCA | ACACGGCCATCAGGAGCCCCA |
| ROMK (4) | CCGTGTTCATCACAGCCTTCTT | CCGTAACCTATGGTCACTTGGG |
| NKCC2 (4) | CCGTGGCCTACATAGGTGTT | GGCTCGTGTTGACATCTTGA |
| NCC (73) | CAGTGCCTGGTGCTTACAGGGC | CATCATGCAGGACACCAATG |
| TRPM6 (36) | AAAGCCATGCGAGTTATCAGC | CTTCACAATGAAAACCTGCCC |
| α-ENaC (59) | TGCTCCTGTCACTTCAGCAC | CCCCTTGCTTAGCCTGTTC |
| Pendrin (68) | GCTGGCCTCATCTCAGCTG | GCAAGGGTTCCAGAAGCCT |
| AE1 (6) | AGGACCTGGTGTTGCCAGAG | CGGTTATGCGCCATGGA |
| AQP2 (4) | TCACTGGGTCTTCTGGATCG | CGTTCCTCCCAGTCAGTGT |
| β-Actin (59) | GGTCAGAAGGACTCCTATGTGG | TGTCGTCCCAGTTGGTAACA |
See the text for definitions.
Immunocytochemistry.
All solutions were made in PBS-CM [PBS (pH 7.4) containing 1 mM CaCl2 and 0.1 mM MgCl2]. Cells were fixed in filter cups in 4% paraformaldehyde for 15 or 30 min at room temperature or 4°C, respectively, permeabilized in 0.2% Triton X-100 for 10 min, and blocked in 0.2% gelatin or 3% BSA with 0.1% Tween and 5% normal donkey serum in PBS-CM. Antibody dilutions were made in blocking solution and incubated for 30–60 min at 37°C in a humidified chamber for both surfaces. Filters were cut off from the cups and mounted in DAPI-mounting media. We imaged Z-slices using a Leica-SP2 confocal microscope.
Quantification of cell types in polarized monolayers.
Primary cells from three separate mice were cultured on permeable supports for 7 days as described above and then incubated for 24 h in basal REBM followed by 1 μM aldosterone for 6 h to enhance the sensitivity of detection of α-ENaC. Immunocytochemistry was performed as described above, with primary antibodies against NCC, TRPV5, and α-ENaC. Cells were counted from four representative images at ×400 total magnification per filter (mean cells counted per filter, 1,726 ± 85). Cells were classified as DCT if NCC(+); CNT principal cells if TRPV5(+), α-ENaC(+); CCD principal cells if NCC(−), TRPV5(−), and α-ENaC(+)(32); or other if not positively stained for any of the three markers.
Electrophysiology.
We measured potential difference and transepithelial resistance (Rte) using an Evometer “chopstick voltmeter” (World Precision Instruments, Sarasota, FL). Cell sheets were deemed viable once Rte exceeded 1,000 Ω·cm2, which occurred 5–6 days after harvesting. One day before the experiments, cell sheets were incubated in basal REBM for 24 h, then treated with 1 μM aldosterone or an equivalent volume of vehicle (100% ethanol) 6 h before short-circuit current measurement (Isc). Cell sheets were studied in Ussing chambers (Physiological Instruments, San Diego, CA), as described previously (53). Briefly, cell sheets were bathed (exposed area 1.2 cm2) in Krebs-Henseleit solution (in mM: 118 NaCl, 25 NaHCO3, 1.2 KH2PO4, 4.7 KCl, 10 glucose, 1.2 CaCl2, and 1.2 MgCl2) and gassed with 95% oxygen and 5% CO2 to produce a pH of 7.4 maintained at 37°C. Transepithelial voltage (Vte) was clamped to 0 mV, and the resulting Isc was displayed continuously on a chart recorder. A set voltage pulse (0.5–2.0 mV) was applied across the cell sheet for 200 milliseconds every 1–20 s, and Rte was determined from the ratio of applied voltage to additional clamp current required. Isc and Rte stabilized within the first 10 min of mounting of the tissue, at which point we added a series of pharmacological agents (Sigma-Aldrich) to the luminal side. Cell sheets were treated with 10−5 M amiloride to inhibit ENaC-mediated Na+ absorption, 10−3 M barium chloride to inhibit potassium channels, and/or 10−5 M ATP to stimulate calcium-dependent anion secretion. All agents were added as 1:1,000 dilutions of stocks in water. Vehicle controls had no effect on Isc.
Statistical analysis.
Data analysis was performed using a paired or unpaired t-test using the Analysis Toolpack of Microsoft Excel. Results are provided as means ± SE; n = number of samples; N = number of mice; and we defined statistical significance at a P value <0.05.
RESULTS
DBA-linked biotin selectively binds the connecting tubule and collecting duct from the mouse kidney.
DBA, a lectin that binds terminal NAG, has previously been shown to bind the rat proximal tubule and rat/mouse collecting duct (21, 22, 30, 31), and DBA-biotin can be linked to streptavidin-coated magnetic beads for isolation of lectin-bound cells. To determine the utility of DBA-biotin to harvest cortical tubule cells, we first tested the specificity of DBA-biotin in an adult mouse kidney (Fig. 2). On longitudinal sections through a single tubule, DBA staining primarily colocalized with cortical tubules stained for aquaporin-2, a marker of principal cells in CNT and the cortical and medullary collecting duct (27, 45) (Fig. 2A). Similar to aquaporin-2 immunostaining, DBA staining was more pronounced in medullary than in cortical tubules. DBA-stained cortical tubules shared minimal overlap with markers for the thick ascending limb of the loop of Henle (Tamm-Horsfall protein) (29, 74) or for DCT (NCC) (7, 32) (Fig. 2, B and C, respectively). This expression profile predicts that cell preparations harvested from adult murine kidney cortex using DBA-conjugated beads would be enriched for CNT and CCD, but not thick ascending limb or DCT.
Fig. 2.
Dolichos biflorus agglutinin (DBA) lectin colocalizes with markers of the murine connecting tubule and collecting duct. Immunofluorescence microscopy is shown of representative adult mouse kidney sections with staining for α-N-acetyl galactosamine (NAG) using DBA-biotin and markers for various subtypes of tubular epithelial cells: aquaporin-2 (AQP2; A); principal cells: Tamm-Horsfall protein (THP; thick ascending limb; B); and Na-Cl cotransporter (NCC; distal convoluted tubule; C). Cortical and medullary sections are labeled as indicated. The red signal indicates DBA-bound NAG, and the green signal indicates colocalizing markers. The blue signal indicates nuclear staining (4,6-diamidino-2-phenylindole; DAPI). G, glomeruli in cortical sections. The scale bars are indicated in micrometers.
Gene expression of primary cell preparations.
As shown in Fig. 3A, we used DBA-conjugated magnetic beads to isolate cells/tubules from perfused adult mouse kidney cortex. We grossly separated cortical from medullary tissue and then added collagenase/hyalurondiase to minced cortical tissue. The resultant preparation contained a heterogeneous mixture of single cells, short tubules, longer undigested tubules, and glomeruli. We removed glomeruli and longer undigested tubules by sieving through a 40-μm strainer. Finally, we used DBA-conjugated magnetic beads to select DBA-expressing cortical cells. The protocol from death to elution requires 4–5 h.
Fig. 3.
Gene expression profile of stage-specific isolation of cell preparations using DBA-lectin. A: flow chart of isolation protocol from digestion of tissue to isolation and elution of DBA-bound cells using magnetic beads. B: real-time PCR relative quantification of segment-specific genes from whole kidney (light gray bars); DBA-bound cells (black bars); and unbound cells (white bars). Segment-specific genes are assigned as indicated. G, glomerulus; PT, proximal tubule; tDL, thin descending limb of loop of Henle; TAL, thick ascending limb; DCT, distal convoluted tubule; CNT, connecting tubule; CCD, cortical collecting duct; MCD, medullary collecting duct. *P < 0.05 between selected populations vs. whole kidney; N = 3 mice.
We compared relative gene expression profiles from this preparation to that from whole kidney (Fig. 3, B and C). The volume of cells bound to DBA beads was approximately one-tenth of the volume of unbound cells. Expression of nephrin (NPHS1), a marker for glomeruli (52), was low in DBA-bound and unbound preparations compared with whole kidney. Expression of markers for the medulla, including UTA2 (thin descending limb of the loop of Henle) (13) and aquaporin-2 (medullary >> CCD) (27), was also low in both DBA-bound and unbound preparations. In contrast, expression of markers for the proximal tubule, including SGLT2 and GLUT2 (65), was significantly enriched in unbound preparations. Surprisingly, expression of markers for the distal cortical tubule, including NCC (DCT1 and DCT2), TRPM6 (DCT1 > DCT2) (72), ENaC (DCT2 to principal cells of the CNT and CCD) (43), and pendrin (intercalated cells from CCD) (25, 40), were enriched in DBA-bound preparations. This was unexpected because DBA staining by immunofluorescence microscopy was much stronger in only CNT and CCD (Fig. 2). Markers that are not confined to the distal cortical tubule from DCT to CCD (e.g., ROMK and AE1) (34, 40) were present but not enriched in DBA-bound preparations. Notably, our DBA-bound preparations contained markers for DCT1 and DCT2 (NCC, TRPM6) and intercalated cells (pendrin, AE1), supporting our observation that we were digesting tubular fragments from the distal nephron rather than individual principal cells.
Primary culture of DBA-bound preparations demonstrates a tight, differentiated epithelial monolayer composed predominantly of principal cells from the CCD.
To characterize the electrophysiological properties of DBA-bound cell preparations, we examined whether these cells differentiate and polarize in cell culture. As depicted in Fig. 4A, we incubated DBA-bound preplated cells with trypsin and plated single cells on permeable supports to create a monolayer. As shown in Fig. 4B, transepithelial resistance rose significantly each day in these preparations. At day 7, we fixed and stained cells for markers of a differentiated, tight epithelium. As shown in Fig. 4, C and D, we stained cell monolayers with ZO-1 and α-tubulin to mark tight junctions and primary cilia, respectively (49, 61). Z-stack images show a polarized monolayer with a single cilium at the apical surface. We next evaluated the proportion of these cells that belong to each segment of the ASDN.
Fig. 4.
Development of a high-resistance, polarized monolayer of distal nephron cells in culture. A: flow chart of plating and culture of DBA-bound preparations. B: transepithelial resistance leading up to day of treatment; n = 34 wells; N = 4 mice. *P < 0.05 vs. the resistance on the prior day. Also shown is confocal microscopy of immunocytochemical stain of slides from a representative culture of cells from day 0 for zonula occludin (ZO)-1 (green; C), a marker for tight junctions, and acetylated tubulin (green; D), a marker for primary cilia. The red signal indicates DBA-bound NAG, and the blue signal indicates nuclear staining (DAPI). Images are shown in the xy-plane, and in D the xz-stack is shown below. The scale bars are indicated in micrometers. *P < 0.05 vs. the resistance on the prior day. E: individual (circles) and mean (rectangles) values for cells identified as DCT (light gray), CNT principal cells (dark gray), CCD principal cells (black; PC), and other (white); n = 4 images/mouse; N = 3 mice. *P < 0.05 vs. CCD principal cells.
As shown in Fig. 4E, we estimate that 61.4 ± 1.85% of cultured cells are principal cells from the CCD, and the remainder are DCT (4.8 ± 0.43%), principal cells from CNT (7.4 ± 0.57%), and other cell types (26.4 ± 1.43%). We next evaluated whether these cultured cells retain the function of channels from these segments of the nephron.
Primary culture of DBA-bound preparations retain expression and electrophysiological properties of the aldosterone-sensitive distal nephron.
At day 7, we incubated cells in basal media for 24 h and treated with either vehicle or 1 μM aldosterone. We then harvested these cells and probed for the aldosterone-stimulated genes SGK1 and α-ENaC (67). As shown in Fig. 5A, protein expression of both gene products markedly increased with aldosterone compared with vehicle treatment at 6 and 24 h. In Fig. 5B, we show the staining against α-ENaC in primary cell preparations treated with vehicle or aldosterone for 24 h, as indicated. In Ussing chambers, we detected a significantly increased amiloride-sensitive apical-to-basal sodium current (Fig. 5C) with 1 nM and 1 μM aldosterone- vs. vehicle-treated cells (1 nM, 32.8 ± 2.6; 1 μM, 45.7 ± 3.5 vs. vehicle, 28.2 ± 1.3 μA/cm2; P < 0.05) at 6 h (Fig. 5D).
Fig. 5.
Aldosterone responsiveness of primary distal nephron cultures. A: representative immunoblots from lysates of cells plated for 7 days, then incubated in depleted media for 24 h, followed by treatment with vehicle (V) or 1 μM aldosterone (A) as indicated. Arrows indicate expected bands for SGK1 and α-ENaC. B: confocal microscopy of representative immunocytochemical stain of primary cells treated with vehicle (top) or aldosterone (Aldo; bottom) for 24 h. The green signal indicates ZO-1; the red signal indicates α-ENaC; and the blue signal indicates nuclear staining (DAPI). The scale bars are indicated in micrometers. C: representative tracings of amiloride-sensitive short-circuit current from cells plated for 7 days, depleted for 24 h, treated with vehicle (light gray trace), 1 nM aldosterone (dark gray trace), or 1 μM aldosterone (black trace) for 6 h, and mounted in Ussing chambers. Cells were treated with amiloride as indicated by the black bar. The scale bar is indicated in seconds (sec). D: individual (circles) and mean (rectangles) values for amiloride-sensitive current from vehicle (light gray)-, 1 nM aldosterone (dark gray)-, or 1 μM aldosterone-treated (black) cells; n = 6 filters; N = 6 mice. *P < 0.05 vs. vehicle-treated cells. #P < 0.05 vs. 1 nM aldosterone-treated cells.
Interestingly, the electrical response of DBA cell preparations to aldosterone was dependent on tissue culture conditions. The aldosterone-sensitive induction of SGK1 and α-ENaC was observed in cells cultured in either REBM or mpkCCDc14 media (3), a defined medium commonly used for immortalized principal cells. However, aldosterone-induced, amiloride-sensitive current was present only with incubation of cells in REBM (Fig. 6).
Fig. 6.
Comparison of aldosterone response using different defined media. DBA-isolated primary cells from mouse cortical collecting tubule were plated on semipermeable, collagen-coated filters, cultured in mpkCCDc14 vs. Renal Epithelial Cell Growth Basal Medium (REBM) for 7 days, and corresponding depleted media for 24 h before treatment with vehicle (ethanol) vs. 1 μM aldosterone for 6 h. Shown are transepithelial resistance (A) and potential difference (B) for days before incubation in depleted media (day 0); n = 26–36 wells; N = 6 mice/group. C: change in equivalent current from 0 to 6 h with vehicle vs. aldosterone treatment for cells grown in mpkCCDc14 (light gray bars) vs. REBM (black bars); n = 6–13 wells; N = 6 mice/group. D: representative immunoblots from lysates of cells plated for 7 days, then depleted for 24 h, followed by treatment with vehicle (V) or 1 μM aldosterone (A) as indicated. Arrows indicate expected bands for SGK1, α-ENaC, and 14-3-3. See materials and methods for details. #P < 0.05 vs. the measurement on the prior day for cells incubated in mpkCCDc14 or REBM, respectively. *P < 0.05 between vehicle- and aldosterone-treated cells.
Potassium and anion currents of primary cell preparations.
To detect the presence of other ion currents from our primary cell preparations, as shown in Fig. 7, A and B, we also measured apical barium-sensitive currents that are likely mediated by ROMK channels on the apical surface (19, 70). Under short-circuit conditions, we detected no significant difference in aldosterone- vs. vehicle-treated cells in the presence of amiloride (1.4 ± 0.3 vs. 2.0 ± 0.5 μA/cm2, P = 0.45). We also detected ATP-sensitive anion currents (Fig. 7, C and D), presumably due to chloride secretion (54), and these currents were not influenced by aldosterone under short-circuit conditions (54) (aldosterone, 0.9 ± 0.1; vehicle, 0.5 ± 0.5 μA/cm2, P = 0.28). Potassium and anion currents were of comparable magnitude to those measured in other CCD cell lines (14, 54).
Fig. 7.
Potassium and anion currents of primary distal nephron cultures. A: representative tracings of barium-sensitive current from cells plated for 7 days, depleted for 24 h, treated with vehicle (light gray trace) or aldosterone (black trace) for 6 h, and mounted in Ussing chambers. Cells were treated with barium as indicated by the black bar. Tracings are normalized to baseline short-circuit current. B: individual (circles) and mean (rectangles) values for barium-sensitive current from vehicle (light gray)- or aldosterone-treated (black) cells. C: representative tracings of ATP-sensitive transient and sustained current from cells plated for 5 days, depleted for 24 h, treated with vehicle (gray trace) or aldosterone (black trace) for 6 h, and mounted in Ussing chambers. Cells were treated with ATP as indicated by the black bar. B: individual (circles) and mean (rectangles) values for ATP-sensitive sustained current from vehicle (light gray)- or aldosterone-treated (black) cells. The scale bars are indicated in seconds (sec); n = 3–4 chambers; N = 3 mice.
DISCUSSION
The ideal preparation of murine ASDN would be a primary culture of a heterogeneous mixture of cells with sufficient yield for gene expression, biochemical, cytological, and electrophysiological studies. The method for isolating and growing these cells would be efficient, tractable, and available for different types of transgenic lines or a treated or diseased mouse model. The protocol would be standardized and require no specialized technique or expensive equipment. The cells would need to retain their differentiated state as polarized renal tubular epithelia and be electrically active.
The DBA lectin binds NAG and has been described as a marker of the collecting duct in the developing and adult kidneys of several mammalian species, including the mouse, rat, and rabbit (21, 30, 31, 38, 57), and other investigators have used DBA lectin binding to isolate medullary collecting duct cells (17, 42, 62, 76, 77). We have advanced this technology and developed the use of DBA-biotin linked to streptavidin-coated beads for isolation and primary culture of adult mouse cortical distal tubule. To isolate cortical preparations, we crudely separated cortex from medulla, and we also took advantage of the relative resistance of medullary fragments to digestion, which has been attributed to the presence of desmosomes (1, 62, 75). Aquaporin-2 is more abundantly expressed in medullary than in cortical collecting duct (27); hence, when we isolated cortical tissue, we lost yield of the aquaporin-2-expressing cells. However, we retained DBA-bound cortical cells, which developed high-resistance epithelia and retained sensitivity to aldosterone.
Primary culture of these DBA-bound cortical cells recapitulates a polarized tubular epithelial monolayer ex vivo. First, primary cultures of cortical preparations express both ZO-1 and primary cilia, indicating that these cells do not dedifferentiate in cell culture. Moreover, the presence of primary cilia in our primary cells indicates that our preparations exhibit characteristics of renal cortex because inner medullary collecting duct cells have been reported to lack primary cilia (9). Second, these preparations exhibit high transepithelial resistance, comparable with those found in M-1, mpkCCD14, or mCCD11 cells (3, 16, 63). Last, these high-resistance monolayers demonstrate functional segment-specific sodium, potassium, and chloride channels. Taken together, primary cultures of these preparations represent viable, terminally differentiated distal cortical tubular epithelia.
These primary cultures were a heterogeneous collection of cell types, comprised mostly of principal cells of the CCD, but the proportion of each segment represented is consistent across preparations (Fig. 4E). Approximately one-quarter of the cells did not stain positive for markers of DCT, CNT, or CCD, and we presume that the majority of these unlabeled cells are intercalated cells. This would closely approximate the ratio of principal cells to intercalated cells in mouse CNT and CCD (24). We treated cells with aldosterone for 6 h to maximize detection of α-ENaC, but we cannot exclude that changes in the culture media could enhance expression, and therefore alter identification, of other cell types. For example, we did not treat cells with angiotensin II or hypotonic low-chloride buffer to stimulate NCC (36) or calciotropic hormones to stimulate TRPV5 expression (66).
The cortical ASDN is responsive to aldosterone in vivo (46), and an ideal preparation should retain this property ex vivo. Aldosterone treatment of our cell preparation induced protein expression of SGK1 and α-ENaC, comparable to what has been demonstrated in vivo (33). Aldosterone also increased amiloride-sensitive sodium current when measured either by short-circuit current in Ussing chambers (Fig. 5) or by equivalent current using chopstick voltmeters (Fig. 6). When measured in Ussing chambers, increased amiloride-sensitive current was observed with physiological concentrations of aldosterone (1 nM), similar to mCCD11 cells (16). Interestingly, to retain electrical sensitivity to aldosterone, the cells require a defined medium that is slightly different from the common media used for growth of immortalized principal cells, mpkCCDc14 or mCCD11 (Fig. 6) (3, 16). We speculate that the higher baseline potential difference and hence, equivalent current, and the insensitivity to aldosterone observed with primary cells in mpkCCDc14 media may have favored a DCT2/CNT phenotype. In contrast, cells incubated with REBM, which exhibited a lower baseline but higher aldosterone-stimulated sodium current, resembled a CCD phenotype (43). REBM has three known differences from mpkCCDc14 media: 1) a lack of supplemental selenium; 2) substitution of dexamethasone with hydrocortisone; and 3) the presence of epinephrine. Hydrocortisone is less potent and less stable than dexamethasone (37), and norepinephrine has been shown to variably promote apical chloride secretion or sodium reabsorption in CCD cells (10, 35). Perhaps these differences may prime the cells for aldosterone-stimulated sodium uptake. The mechanism by which REBM promotes electrical sensitivity of our cell preparations to aldosterone is not known, but once discovered, may provide insight into the determinants that dictate axial heterogeneity between the DCT2/CNT and CCD.
Potassium secretion, a vital component of ion transport along the ASDN, has been reported in immortalized mCCDc11 cells (14) and patch-clamp studies of CCD (15, 47), but not in primary cultures isolated with DBA beads or COPAS. Under short-circuit conditions, we obtained a barium-sensitive potassium current that was not directly modified by aldosterone, similar to that reported for immortalized principal cell preparations obtained from microdissected CCD (14). Further studies may determine the magnitude of potassium flux attributed directly or indirectly to aldosterone. The response to apical barium likely reflects secretion through ROMK, as opposed to iberitoxin-sensitive, barium-insensitive BK channels on the apical membrane. Since both apical and basal potassium channels in this CCD are mutated in human disease (14), our preparations may provide a novel culture system that can be used to study the regulation of potassium transport in the ASDN.
More specialized techniques such as microdissection (2, 12, 51, 56) yield a higher purity of specific nephron segments but compromise on time, yield, and feasibility for most investigators. The COPAS technique provides high yield and purity for DCT or CD (20, 36, 39) but requires the time and expense associated with breeding to fluorescent reporter mice (that may not be of similar background to one's target mouse line), specialized equipment, and technical personnel. The hypotonic lysis method for isolation of the medullary collecting duct is fairly inexpensive, but this method cannot be used for sensitive measurements (e.g., oxygen consumption) (62) and does not capture cortical segments of the ASDN. Patch-clamp studies of split-open murine CCD provide for precise electrophysiology but require trained personnel and cannot be used for gene expression, biochemical, or cytological studies (60). Tubular microperfusion is also a superior technique for segment-specific electrophysiology and flux studies (e.g., flow-mediated potassium secretion), but requires young mice, a trained perfusionist, has lower yield, and cannot capture the CNT or medullary CD due to the requirement for a single linear tubule (55).
Our method has several limitations. First, there is inherent variability with partial digestion of cortical tubules; however, we contend that sufficient digestion was achieved as our final preparation demonstrated depletion of medullary cell markers, (e.g., aquaporin-2), and enrichment of cortical markers, (e.g., α-ENaC and pendrin). Second, our technique is limited in cell specificity compared with more precise isolation methods such as microdissection or cell-sorting by COPAS. The presence of DCT cells in our preparation was unexpected based on the observation that DBA does not stain for nephron segments that express NCC. We infer that DCT cells were harvested by DBA-conjugated beads because bound tubules may contain DCT, or NAG that is present in DCT may bind DBA more efficiently under harvest vs. immunohistochemical conditions (41). With our culture conditions, we preserved only a minority of DCT cells compared with more specific techniques such as COPAS, and thus these cells are not a good model for studying NCC-mediated transport.
Despite these limitations, our technique can be used to characterize distal nephron function in wild-type or transgenic mice, and the yield of cells from this technique is sufficient for gene expression, biochemical, cytological, and electrophysiological studies. Moreover, the lack of specificity allows one to capture the entirety of the ASDN, from DCT to CCD, including intercalated cells; thus our method can be utilized to study the relationship between cell types in this highly regulated and interdependent stretch of the nephron. Our primary culture method may be superior to immortalized epithelia such as M-1 (63), mpkCCDc14 (3), or mCCDc11 (16) cells because it does not depend on the immortalization process, which may alter cell cycle or differentiation pathways. Immortalized cell lines also provide a high-yield continuous source of cells but are limited to one cell type and may not be useful to study the integrated ASDN from treated or transgenic mice. The addition of markers for DCT, including NCC and TRPM6, provide for an alternative to COPAS (36), mpkDCT (11), or mDCT15 (28) cells for biochemical or immunocytochemical characterization of this segment. Previously available techniques have both strengths and limitations, but our preparation from the mouse kidney cortex using DBA-coated beads provides for a pragmatic approach.
In summary, we have optimized the protocol of Grupp et al. (17) using DBA-conjugated beads for an inexpensive, efficient, and simple technique that provides sufficient yield and purity for phenotypic characterization of ASDN from mice.
GRANTS
M. Labarca received support from the Becas Chile scholarship. J. M. Nizar is supported by the Tashia and John Morgridge Endowed Postdoctoral Fellowship and the Child Health Research Institute at Stanford University. E. M. Walczak is supported by the NIH (T32 DK007357). V. Bhalla has received support from the NIH (5R03 DK083613), and the American Society of Nephrology (Carl W. Gottschalk Research Grant), and currently receives support from the NIH (1R01 DK091565).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the authors.
AUTHOR CONTRIBUTIONS
Author contributions: M.L., J.M.N., E.M.W., and W.D. performed experiments; M.L., J.M.N., E.M.W., W.D., and V.B. analyzed data; M.L., J.M.N., E.M.W., A.C.P., and V.B. interpreted results of experiments; M.L., J.M.N., E.M.W., and V.B. prepared figures; M.L., J.M.N., E.M.W., A.C.P., and V.B. edited and revised manuscript; M.L., J.M.N., E.M.W., W.D., A.C.P., and V.B. approved final version of manuscript; V.B. provided conception and design of research; V.B. drafted manuscript.
ACKNOWLEDGMENTS
We thank Drs. Tianxin Yang and Glenn Chertow for helpful suggestions for troubleshooting the isolation technique and for a critical review of the manuscript, respectively. We also are indebted to the late Dr. John Stokes (University of Iowa) for his helpful scientific discussions. We thank the Stanford University Cell Sciences Imaging Facility for assistance with confocal microscopy and the Protein and Nucleic Acid Facility for assistance with semiquantitative PCR.
The project described was supported in part by Grant S10RR017959 from the National Center for Research Resources (NCRR). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of the NCRR or the National Institutes of Health (NIH).
V. Bhalla is the guarantor of this manuscript and takes full responsibility for the content herein.
REFERENCES
- 1.Bachmann S, Kriz W, Kuhn C, Franke WW. Differentiation of cell types in the mammalian kidney by immunofluorescence microscopy using antibodies to intermediate filament proteins and desmoplakins. Histochemistry 77: 365–394, 1983. [DOI] [PubMed] [Google Scholar]
- 2.Bens M, Duong Van Huyen JP, Cluzeaud F, Teulon J, Vandewalle A. CFTR disruption impairs cAMP-dependent Cl− secretion in primary cultures of mouse cortical collecting ducts. Am J Physiol Renal Physiol 281: F434–F442, 2001. [DOI] [PubMed] [Google Scholar]
- 3.Bens M, Vallet V, Cluzeaud F, Pascual-Letallec L, Kahn A, Rafestin-Oblin ME, Rossier BC, Vandewalle A. Corticosteroid-dependent sodium transport in a novel immortalized mouse collecting duct principal cell line. J Am Soc Nephrol 10: 923–934, 1999. [DOI] [PubMed] [Google Scholar]
- 4.Bernascone I, Janas S, Ikehata M, Trudu M, Corbelli A, Schaeffer C, Rastaldi MP, Devuyst O, Rampoldi L. A transgenic mouse model for uromodulin-associated kidney diseases shows specific tubulo-interstitial damage, urinary concentrating defect and renal failure. Hum Mol Genet 19: 2998–3010, 2010. [DOI] [PubMed] [Google Scholar]
- 5.Bhalla V, Hallows KR. Mechanisms of ENaC regulation and clinical implications. J Am Soc Nephrol 19: 1845–1854, 2008. [DOI] [PubMed] [Google Scholar]
- 6.Bonnici B, Wagner CA. Postnatal expression of transport proteins involved in acid-base transport in mouse kidney. Pflügers Arch 448: 16–28, 2004. [DOI] [PubMed] [Google Scholar]
- 7.Campean V, Kricke J, Ellison D, Luft FC, Bachmann S. Localization of thiazide-sensitive Na+-Cl− cotransport and associated gene products in mouse DCT. Am J Physiol Renal Physiol 281: F1028–F1035, 2001. [DOI] [PubMed] [Google Scholar]
- 8.Chandran S, Li H, Dong W, Krasinska K, Adams C, Alexandrova L, Chien A, Hallows KR, Bhalla V. Neural precursor cell-expressed developmentally down-regulated protein 4-2 (Nedd4-2) regulation by 14-3-3 protein binding at canonical serum and glucocorticoid kinase 1 (SGK1) phosphorylation sites. J Biol Chem 286: 37830–37840, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Clapp WL, Madsen KM, Verlander JW, Tisher CC. Morphologic heterogeneity along the rat inner medullary collecting duct. Lab Invest 60: 219–230, 1989. [PubMed] [Google Scholar]
- 10.Cuffe JE, Howard DP, Bertog M, Korbmacher C. Basolateral adrenoceptor activation mediates noradrenaline-induced Cl− secretion in M-1 mouse cortical collecting duct cells. Pflügers Arch 445: 381–389, 2002. [DOI] [PubMed] [Google Scholar]
- 11.Duong Van Huyen JP, Bens M, Teulon J, Vandewalle A. Vasopressin-stimulated chloride transport in transimmortalized mouse cell lines derived from the distal convoluted tubule and cortical and inner medullary collecting ducts. Nephrol Dial Transplant 16: 238–245, 2001. [DOI] [PubMed] [Google Scholar]
- 12.Fakitsas P, Adam G, Daidie D, van Bemmelen MX, Fouladkou F, Patrignani A, Wagner U, Warth R, Camargo SM, Staub O, Verrey F. Early aldosterone-induced gene product regulates the epithelial sodium channel by deubiquitylation. J Am Soc Nephrol 18: 1084–1092, 2007. [DOI] [PubMed] [Google Scholar]
- 13.Fenton RA, Knepper MA. Urea and renal function in the 21st century: insights from knockout mice. J Am Soc Nephrol 18: 679–688, 2007. [DOI] [PubMed] [Google Scholar]
- 14.Fodstad H, Gonzalez-Rodriguez E, Bron S, Gaeggeler H, Guisan B, Rossier BC, Horisberger JD. Effects of mineralocorticoid and K+ concentration on K+ secretion and ROMK channel expression in a mouse cortical collecting duct cell line. Am J Physiol Renal Physiol 296: F966–F975, 2009. [DOI] [PubMed] [Google Scholar]
- 15.Frindt G, Palmer LG. Apical potassium channels in the rat connecting tubule. Am J Physiol Renal Physiol 287: F1030–F1037, 2004. [DOI] [PubMed] [Google Scholar]
- 16.Gaeggeler HP, Gonzalez-Rodriguez E, Jaeger NF, Loffing-Cueni D, Norregaard R, Loffing J, Horisberger JD, Rossier BC. Mineralocorticoid versus glucocorticoid receptor occupancy mediating aldosterone-stimulated sodium transport in a novel renal cell line. J Am Soc Nephrol 16: 878–891, 2005. [DOI] [PubMed] [Google Scholar]
- 17.Grupp C, Troche I, Steffgen J, Langhans S, Cohen DI, Brandl L, Muller GA. Highly specific separation of heterogeneous cell populations by lectin-coated beads: application for the isolation of inner medullary collecting duct cells. Exp Nephrol 6: 542–550, 1998. [DOI] [PubMed] [Google Scholar]
- 18.Hallows KR, Bhalla V, Oyster NM, Wijngaarden MA, Lee JK, Li H, Chandran S, Xia X, Huang Z, Chalkley RJ, Burlingame AL, Pearce D. Phosphopeptide screen uncovers novel phosphorylation sites of Nedd4-2 that potentiate its inhibition of the epithelial Na+ channel. J Biol Chem 285: 21671–21678, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hamilton KL, Devor DC. Basolateral membrane K+ channels in renal epithelial cells. Am J Physiol Renal Physiol 302: F1069–F1081, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Helbert MJ, Dauwe SE, De Broe ME. Flow cytometric immunodissection of the human distal tubule and cortical collecting duct system. Kidney Int 59: 554–564, 2001. [DOI] [PubMed] [Google Scholar]
- 21.Holthofer H. Lectin binding sites in kidney. A comparative study of 14 animal species. J Histochem Cytochem 31: 531–537, 1983. [DOI] [PubMed] [Google Scholar]
- 22.Holthofer H, Schulte BA, Spicer SS. Heterogeneity of apical glycoconjugates in kidney collecting ducts: further studies using simultaneous detection of lectin binding sites and immunocytochemical detection of key transport enzymes. Histochem J 20: 471–477, 1988. [DOI] [PubMed] [Google Scholar]
- 23.Holtzclaw JD, Grimm PR, Sansom SC. Intercalated cell BK-alpha/beta4 channels modulate sodium and potassium handling during potassium adaptation. J Am Soc Nephrol 21: 634–645, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kim J, Kim YH, Cha JH, Tisher CC, Madsen KM. Intercalated cell subtypes in connecting tubule and cortical collecting duct of rat and mouse. J Am Soc Nephrol 10: 1–12, 1999. [DOI] [PubMed] [Google Scholar]
- 25.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] [PubMed] [Google Scholar]
- 26.Kim YH, Pech V, Spencer KB, Beierwaltes WH, Everett LA, Green ED, Shin W, Verlander JW, Sutliff RL, Wall SM. Reduced ENaC protein abundance contributes to the lower blood pressure observed in pendrin-null mice. Am J Physiol Renal Physiol 293: F1314–F1324, 2007. [DOI] [PubMed] [Google Scholar]
- 27.Kishore BK, Terris JM, Knepper MA. Quantitation of aquaporin-2 abundance in microdissected collecting ducts: axial distribution and control by AVP. Am J Physiol Renal Fluid Electrolyte Physiol 271: F62–F70, 1996. [DOI] [PubMed] [Google Scholar]
- 28.Ko B, Mistry AC, Hanson L, Mallick R, Cooke LL, Hack BK, Cunningham P, Hoover RS. A new model of the distal convoluted tubule. Am J Physiol Renal Physiol 303: F700–F710, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kohda Y, Ding W, Phan E, Housini I, Wang J, Star RA, Huang CL. Localization of the ROMK potassium channel to the apical membrane of distal nephron in rat kidney. Kidney Int 54: 1214–1223, 1998. [DOI] [PubMed] [Google Scholar]
- 30.Laitinen L, Virtanen I, Saxen L. Changes in the glycosylation pattern during embryonic development of mouse kidney as revealed with lectin conjugates. J Histochem Cytochem 35: 55–65, 1987. [DOI] [PubMed] [Google Scholar]
- 31.Le Hir M, Dubach UC. The cellular specificity of lectin binding in the kidney. I. A light microscopical study in the rat. Histochemistry 74: 521–530, 1982. [DOI] [PubMed] [Google Scholar]
- 32.Loffing J, Loffing-Cueni D, Valderrabano V, Klausli L, Hebert SC, Rossier BC, Hoenderop JG, Bindels RJ, Kaissling B. Distribution of transcellular calcium and sodium transport pathways along mouse distal nephron. Am J Physiol Renal Physiol 281: F1021–F1027, 2001. [DOI] [PubMed] [Google Scholar]
- 33.Loffing J, Zecevic M, Feraille E, Kaissling B, Asher C, Rossier BC, Firestone GL, Pearce D, Verrey F. Aldosterone induces rapid apical translocation of ENaC in early portion of renal collecting system: possible role of SGK. Am J Physiol Renal Physiol 280: F675–F682, 2001. [DOI] [PubMed] [Google Scholar]
- 34.Lu M, Wang T, Yan Q, Yang X, Dong K, Knepper MA, Wang W, Giebisch G, Shull GE, Hebert SC. Absence of small conductance K+ channel (SK) activity in apical membranes of thick ascending limb and cortical collecting duct in ROMK (Bartter's) knockout mice. J Biol Chem 277: 37881–37887, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mansley MK, Neuhuber W, Korbmacher C, Bertog M. Norepinephrine stimulates the epithelial Na+ channel in cortical collecting duct cells via α2-adrenoceptors. Am J Physiol Renal Physiol 308: F450–F458, 2015. [DOI] [PubMed] [Google Scholar]
- 36.Markadieu N, San-Cristobal P, Nair AV, Verkaart S, Lenssen E, Tudpor K, van Zeeland F, Loffing J, Bindels RJ, Hoenderop JG. A primary culture of distal convoluted tubules expressing functional thiazide-sensitive NaCl transport. Am J Physiol Renal Physiol 303: F886–F892, 2012. [DOI] [PubMed] [Google Scholar]
- 37.Meikle AW, Tyler FH. Potency and duration of action of glucocorticoids. Effects of hydrocortisone, prednisone and dexamethasone on human pituitary-adrenal function. Am J Med 63: 200–207, 1977. [DOI] [PubMed] [Google Scholar]
- 38.Michael L, Sweeney DE, Davies JA. The lectin Dolichos biflorus agglutinin is a sensitive indicator of branching morphogenetic activity in the developing mouse metanephric collecting duct system. J Anat 210: 89–97, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Miller RL, Zhang P, Chen T, Rohrwasser A, Nelson RD. Automated method for the isolation of collecting ducts. Am J Physiol Renal Physiol 291: F236–F245, 2006. [DOI] [PubMed] [Google Scholar]
- 40.Mohebbi N, Pena A, van der West J, Becker HM, Caspase G, Wagner CA. regulation of two renal chloride transporters, AE1 and pendrin, by electrolytes and aldosterone. Plops One 8: e55286, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Murata F, Tsuyama S, Suzuki S, Hamada H, Ozawa M, Muramatsu T. Distribution of glycoconjugates in the kidney studied by use of labeled lectins. J Histochem Cytochem 31: 139–144, 1983. [DOI] [PubMed] [Google Scholar]
- 42.Nauli SM, Alenghat FJ, Luo Y, Williams E, Vassilev P, Li X, Elia AE, Lu W, Brown EM, Quinn SJ, Ingber DE, Zhou J. Polycystins 1 and 2 mediate mechanosensation in the primary cilium of kidney cells. Nat Genet 33: 129–137, 2003. [DOI] [PubMed] [Google Scholar]
- 43.Nesterov V, Dahlmann A, Krueger B, Bertog M, Loffing J, Korbmacher C. Aldosterone-dependent and -independent regulation of the epithelial sodium channel (ENaC) in mouse distal nephron. Am J Physiol Renal Physiol 303: F1289–F1299, 2012. [DOI] [PubMed] [Google Scholar]
- 44.Newby LJ, Streets AJ, Zhao Y, Harris PC, Ward CJ, Ong AC. Identification, characterization, and localization of a novel kidney polycystin-1-polycystin-2 complex. J Biol Chem 277: 20763–20773, 2002. [DOI] [PubMed] [Google Scholar]
- 45.Nielsen S, DiGiovanni SR, Christensen EI, Knepper MA, Harris HW. Cellular and subcellular immunolocalization of vasopressin-regulated water channel in rat kidney. Proc Natl Acad Sci USA 90: 11663–11667, 1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pacha J, Frindt G, Antonian L, Silver RB, Palmer LG. Regulation of Na channels of the rat cortical collecting tubule by aldosterone. J Gen Physiol 102: 25–42, 1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Palmer LG, Frindt G. Aldosterone and potassium secretion by the cortical collecting duct. Kidney Int 57: 1324–1328, 2000. [DOI] [PubMed] [Google Scholar]
- 48.Pao AC, McCormick JA, Li H, Siu J, Govaerts C, Bhalla V, Soundararajan R, Pearce D. NH2 terminus of serum and glucocorticoid-regulated kinase 1 binds to phosphoinositides and is essential for isoform-specific physiological functions. Am J Physiol Renal Physiol 292: F1741–F1750, 2007. [DOI] [PubMed] [Google Scholar]
- 49.Pazour GJ, San Agustin JT, Follit JA, Rosenbaum JL, Witman GB. Polycystin-2 localizes to kidney cilia and the ciliary level is elevated in orpk mice with polycystic kidney disease. Curr Biol 12: R378–R380, 2002. [DOI] [PubMed] [Google Scholar]
- 50.Pearce D, Soundararajan R, Trimpert C, Kashlan OB, Deen PM, Kohan DE. Collecting duct principal cell transport processes and their regulation. Clin J Am Soc Nephrol 10: 135–146, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Pradervand S, Vandewalle A, Bens M, Gautschi I, Loffing J, Hummler E, Schild L, Rossier BC. Dysfunction of the epithelial sodium channel expressed in the kidney of a mouse model for Liddle syndrome. J Am Soc Nephrol 14: 2219–2228, 2003. [DOI] [PubMed] [Google Scholar]
- 52.Putaala H, Soininen R, Kilpelainen P, Wartiovaara J, Tryggvason K. The murine nephrin gene is specifically expressed in kidney, brain and pancreas: inactivation of the gene leads to massive proteinuria and neonatal death. Hum Mol Genet 10: 1–8, 2001. [DOI] [PubMed] [Google Scholar]
- 53.Rajagopal M, Kathpalia PP, Thomas SV, Pao AC. Activation of P2Y1 and P2Y2 receptors induces chloride secretion via calcium-activated chloride channels in kidney inner medullary collecting duct cells. Am J Physiol Renal Physiol 301: F544–F553, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rajagopal M, Kathpalia PP, Widdicombe JH, Pao AC. Differential effects of extracellular ATP on chloride transport in cortical collecting duct cells. Am J Physiol Renal Physiol 303: F483–F491, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Satlin LM, Sheng S, Woda CB, Kleyman TR. Epithelial Na+ channels are regulated by flow. Am J Physiol Renal Physiol 280: F1010–F1018, 2001. [DOI] [PubMed] [Google Scholar]
- 56.Schlatter E, Frobe U, Greger R. Ion conductances of isolated cortical collecting duct cells. Pflügers Arch 421: 381–387, 1992. [DOI] [PubMed] [Google Scholar]
- 57.Schumacher K, Strehl R, Minuth WW. Detection of glycosylated sites in embryonic rabbit kidney by lectin chemistry. Histochem Cell Biol 118: 79–87, 2002. [DOI] [PubMed] [Google Scholar]
- 58.Shibata S, Rinehart J, Zhang J, Moeckel G, Castaneda-Bueno M, Stiegler AL, Boggon TJ, Gamba G, Lifton RP. Mineralocorticoid receptor phosphorylation regulates ligand binding and renal response to volume depletion and hyperkalemia. Cell Metab 18: 660–671, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Shigemura N, Ohkuri T, Sadamitsu C, Yasumatsu K, Yoshida R, Beauchamp GK, Bachmanov AA, Ninomiya Y. Amiloride-sensitive NaCl taste responses are associated with genetic variation of ENaC α-subunit in mice. Am J Physiol Regul Integr Comp Physiol 294: R66–R75, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Staruschenko A, Pochynyuk O, Vandewalle A, Bugaj V, Stockand JD. Acute regulation of the epithelial Na+ channel by phosphatidylinositide 3-OH kinase signaling in native collecting duct principal cells. J Am Soc Nephrol 18: 1652–1661, 2007. [DOI] [PubMed] [Google Scholar]
- 61.Stevenson BR, Siliciano JD, Mooseker MS, Goodenough DA. Identification of ZO-1: a high molecular weight polypeptide associated with the tight junction (zonula occludens) in a variety of epithelia. J Cell Biol 103: 755–766, 1986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Stokes JB, Grupp C, Kinne RK. Purification of rat papillary collecting duct cells: functional and metabolic assessment. Am J Physiol Renal Fluid Electrolyte Physiol 253: F251–F262, 1987. [DOI] [PubMed] [Google Scholar]
- 63.Stoos BA, Naray-Fejes-Toth A., Carretero OA, Ito S, and Fejes-Toth G. Characterization of a mouse cortical collecting duct cell line. Kidney Int 39: 1168–1175, 1991. [DOI] [PubMed] [Google Scholar]
- 64.Subramanya AR, Ellison DH. Distal convoluted tubule. Clin J Am Soc Nephrol 9: 2147–2163, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Vallon V, Platt KA, Cunard R, Schroth J, Whaley J, Thomson SC, Koepsell H, Rieg T. SGLT2 mediates glucose reabsorption in the early proximal tubule. J Am Soc Nephrol 22: 104–112, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.van Abel M, Hoenderop JG, van der Kemp AW, Friedlaender MM, van Leeuwen JP, Bindels RJ. Coordinated control of renal Ca2+ transport proteins by parathyroid hormone. Kidney Int 68: 1708–1721, 2005. [DOI] [PubMed] [Google Scholar]
- 67.Verrey F. Early aldosterone action: toward filling the gap between transcription and transport. Am J Physiol Renal Physiol 277: F319–F327, 1999. [DOI] [PubMed] [Google Scholar]
- 68.Wall SM, Hassell KA, Royaux IE, Green ED, Chang JY, Shipley GL, Verlander JW. Localization of pendrin in mouse kidney. Am J Physiol Renal Physiol 284: F229–F241, 2003. [DOI] [PubMed] [Google Scholar]
- 69.Wall SM, Pech V. Pendrin and sodium channels: relevance to hypertension. J Nephrol 23, Suppl 16: S118–S123, 2010. [PubMed] [Google Scholar]
- 70.Wang WH, Yue P, Sun P, Lin DH. Regulation and function of potassium channels in aldosterone-sensitive distal nephron. Curr Opin Nephrol Hypertens 19: 463–470, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Woda CB, Bragin A, Kleyman TR, Satlin LM. Flow-dependent K+ secretion in the cortical collecting duct is mediated by a maxi-K channel. Am J Physiol Renal Physiol 280: F786–F793, 2001. [DOI] [PubMed] [Google Scholar]
- 72.Xi Q, Hoenderop JG, Bindels RJ. Regulation of magnesium reabsorption in DCT. Pflügers Arch 458: 89–98, 2009. [DOI] [PubMed] [Google Scholar]
- 73.Yang SS, Lo YF, Yu IS, Lin SW, Chang TH, Hsu YJ, Chao TK, Sytwu HK, Uchida S, Sasaki S, Lin SH. Generation and analysis of the thiazide-sensitive Na+-Cl− cotransporter (Ncc/Slc12a3) Ser707X knockin mouse as a model of Gitelman syndrome. Hum Mutat 31: 1304–1315, 2010. [DOI] [PubMed] [Google Scholar]
- 74.Yang T, Huang YG, Singh I, Schnermann J, Briggs JP. Localization of bumetanide- and thiazide-sensitive Na-K-Cl cotransporters along the rat nephron. Am J Physiol Renal Fluid Electrolyte Physiol 271: F931–F939, 1996. [DOI] [PubMed] [Google Scholar]
- 75.Yu ASL, Hanner F, Peti-Peterdi J. Intercellular junctions. In: Seldin and Giebisch's The Kidney. New York: Academic, 2013, p. 347–368. [Google Scholar]
- 76.Zhang H, Zhang A, Kohan DE, Nelson RD, Gonzalez FJ, Yang T. Collecting duct-specific deletion of peroxisome proliferator-activated receptor gamma blocks thiazolidinedione-induced fluid retention. Proc Natl Acad Sci USA 102: 9406–9411, 2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ziehr J, Sheibani N, Sorenson CM. Alterations in cell-adhesive and migratory properties of proximal tubule and collecting duct cells from bcl-2 −/− mice. Am J Physiol Renal Physiol 287: F1154–F1163, 2004. [DOI] [PubMed] [Google Scholar]







