Comprehensive Physiology Wiley Online Library

In Vivo and Ex Vivo Analysis of Tubule Function

Full Article on Wiley Online Library



Abstract

Analysis of tubule function with in vivo and ex vivo approaches has been instrumental in revealing renal physiology. This work allows assignment of functional significance to known gene products expressed along the nephron, primary of which are proteins involved in electrolyte transport and regulation of these transporters. Not only we have learned much about the key roles played by these transport proteins and their proper regulation in normal physiology but also the combination of contemporary molecular biology and molecular genetics with in vivo and ex vivo analysis opened a new era of discovery informative about the root causes of many renal diseases. The power of in vivo and ex vivo analysis of tubule function is that it preserves the native setting and control of the tubule and proteins within tubule cells enabling them to be investigated in a “real‐life” environment with a high degree of precision. In vivo and ex vivo analysis of tubule function continues to provide a powerful experimental outlet for testing, evaluating, and understanding physiology in the context of the novel information provided by sequencing of the human genome and contemporary genetic screening. These tools will continue to be a mainstay in renal laboratories as this discovery process continues and as we continue to identify new gene products functionally compromised in renal disease. © 2012 American Physiological Society. Compr Physiol 2:2495‐2525, 2012.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Preparing the kidney for micropuncture. (A) After a flank incision, the left kidney is carefully freed from perirenal fat and the adrenal gland, overturned, and placed in a cup such that the dorsal kidney surface faces up (B). (C) In the rat, the ureter is catheterized for collection of urine; this is less useful in mice because of a tendency of catheters to obstruct. To immobilize the kidney, the cup is lined with moleskin (D) and cotton soaked in saline is carefully placed around the kidney. 2% to 3% agar is dripped onto the cotton, which, after cooling, prevents leakage of the superfused mineral oil or saline (37°C) thereby establishing an oil or saline layer, respectively, over the kidney surface to keep the kidney surface warm and protected. Adapted, with permission, from reference 17.

Figure 2. Figure 2.

Identifying tubular segments accessible to micropuncture on the kidney surface. The Bowman's space of a Munich‐Wistar‐Fromter rat is injected with small amounts of artificial tubular fluid containing a blue dye. The nephron is mapped by following the dye moving along proximal and distal segments (1‐4). Adapted, with permission, from reference 331.

Figure 3. Figure 3.

Reabsorption of glucose along the proximal tubule. (A) Free‐flow collections of tubular fluid were performed along accessible proximal tubules at the kidney surface to establish a profile for fractional reabsorption of glucose versus fluid. (B) Glucose is rapidly and nearly completely reabsorbed within the early proximal tubule in wild‐type mice (WT). Reabsorption of glucose is absent in the early proximal tubule of mice lacking the sodium glucose cotransporter, SGLT2 (Sglt2−/−). (C) Mean fractional reabsorption of glucose for early (<40%) and late (≥40%) proximal tubular collections and up to the urine. *P<0.001 versus WT. Adapted, with permission, from reference 337.

Figure 4. Figure 4.

Reabsorption of Ca2+ along the nephron. (A) Free‐flow collections of tubular fluid were performed in the last surface loop of the proximal tubule (LPT), as well as, along distal segments (DS) using luminal K+ concentration as a reference for puncturing sites. (B) Ca2+ reabsorption was unaffected in Trpv5−/− mice up to the the proximal tubule surface loop (LPT); in contrast, mean Ca2+ delivery to puncturing sites in the distal segments (DS) accessible to micropuncture as well as urine (U) was significantly enhanced in Trpv5−/− mice. *P<0.001 versus wild type (WT). (C) To directly assess Ca2+ reabsorption along the distal segments, luminal K+ concentration was used as a reference with low and high concentrations indicating early and late aspects of the DS, respectively. Whereas, the Ca2+ delivery profile indicated net reabsorption along the distal segments in WT mice, fractional Ca2+ delivery increased in Trpv5−/− mice, indicating Ca2+ back leak along the distal segments in the absence of Trpv5. Finally, comparing the fractional Ca2+ delivery in the late distal segment with the values in urine indicated significant compensation in the collecting duct of Trpv5−/− (consistent with expression of TRPV6 in that segment). Adapted, with permission, from reference 141.

Figure 5. Figure 5.

Role of Na+/H+ exchanger (NHE3) in proximal tubular absorption. (A) Quantitative collection of tubular fluid was performed during perfusion of superficial proximal tubular segments downstream of an obstructing wax block with control and vehicle solution with and without the addition of the unselective NHE inhibitor ethylisopropylamiloride (EIPA) or the selective NHE3 inihitor S3226. (B) Proximal tubule absorption of HCO3 (JHCO3) and fluid (Jv) were greater in wild‐type (WT) compared to NHE3 null mice (Nhe3−/−). Moreover, EIPA (Black bars) inhibits absorption in WT but not in Nhe3−/− mice. (C) S3226 induces a dose‐dependent reduction in proximal tubular absorption of fluid and sodium in normal rats. *P<0.05 versus control/vehicle. #P<0.05 versus WT. Adapted, with permission, from references 362 and 342.

Figure 6. Figure 6.

Role of maxi‐K (BK) channels in K+ secretion in distal nephron. (A) Stationary microperfusion has been used to study distal K+ secretion. A double‐barreled perfusion pipette containing perfusion solution (P; to identify distal surface segments) and castor oil (CO; to fill the downstream segments) was inserted in the late proximal tubule. A double‐barreled microelectrode with K+‐sensitive ion‐exchange resin (IE) and reference solution (ref) is inserted into a distal segment. Another pipette (S) is placed into the distal tubule to split the oil column and add a low K+‐control solution. The increase in luminal K+ activity is followed and analyzed as a measure of secretory K+ flux, JK. In paired experiments, the BK channel blocker IBTX is added to the tubular fluid. (B) Mice lacking the ROMK channel continue to secrete K+, albeit at a reduced rate, by an IBTX‐sensitive mechanism consistent with BK channels. (C) A high K+ diet increases distal K+ secretion primarily by an IBTX‐sensitive K+ flux. * P<0.05 versus Control. #P<0.05 versus WT or 1.2% K+ diet, respectively. Adapted, with permission, from reference 9.

Figure 7. Figure 7.

Isolated split‐open tubule – a segment of the cortical collecting duct isolated from rat is shown here. The middle of this segment has been split open to allow patch clamp access to the apical membranes of the lining epithelial cells. Apical membranes are shown at higher magnification in the inset.

Figure 8. Figure 8.

Patch clamp configurations used in the isolated split‐open tubule preparation. This illustration depicts the five different seal configurations available for probing channel activity with the voltage clamp approach in isolated split‐open tubules: three amenable to single channel recording, cell‐attached, excised inside‐out, and excised outside‐out; and two for macroscopic current recording, perforated and whole cell. In this figure, the cell is represented as a circle and the recording pipette as a V. Gray shading defines the continuity between the solution in the recording pipette and intracellular solution. Arrows represent transition steps during seal formation taken to establish the distinct types of seals. Undershown are representative current data from configurations with single channel (left) and macroscopic (right) resolutions. Closed (C) and open (O) states are noted in the single channel trace. Data adapted, with permission, from J.D. Stockand or published previously in reference 256.

Figure 9. Figure 9.

SK channel in TAL epithelial cells. Shown is a typical single channel current trace of an SK channel in an inside‐out patch held at hyperpolarizing test potentials made from the apical membrane of an epithelial cell lining a split‐open murine TAL. Patch solutions contained 140/5 mmol/L K+ in the pipette/bath. Inward K+ current is downward. Test voltages and closed state are noted in the figure. Figure originally presented in reference 204.

Figure 10. Figure 10.

IK channel in TAL epithelial cells. Shown is a typical single channel current trace of an IK channel in a cell attached patch held on the apical membrane of an epithelial cell lining a split‐open rat TAL. For this experiment, pipette potential was 0 mV and the recording pipette contained 140 mmol/L KCl. Closed state noted with C. Data provided, with permission, by W‐H. Wang.

Figure 11. Figure 11.

Depiction of ion channel and transport proteins in a TAL epithelial cell.

Figure 12. Figure 12.

Basolateral Cl channels in TAL cells. Shown are typical single channel current traces of the small (∼10 pS; A) and the double‐barreled (B) Cl channels in cell attached patches formed on the basolateral membranes of epithelial cells in isolated, split‐open TAL. For the current trace containing only the small channel (A), the bath and pipette contained symmetrical NaCl and the patch was exposed to hyperpolarizing test pulses. The patch in B contains both the larger double‐barreled channel, as well as, the smaller Cl channel (openings for the smaller channel noted with arrows). This patch was held at 50 and −80 mV test potentials also with symmetrical bath and pipette solutions. Closed state noted with C. Data in A and B originally published in references 125 and 127, respectively.

Figure 13. Figure 13.

Apical BK channel in cortical collecting duct (CCD). Shown here is a representative single channel current trace for a BK channel in the absence and presence of exogenous calcium in an excised, inside‐out patch from a principal cell in an isolated split‐open collecting duct. For this experiment, the patch was held at 0 mV, and the pipette and bath solutions contained 140 and 5 mmol/L KCl, respectively. Closed state noted with C. Data adapted, with permission, from W‐H Wang.

Figure 14. Figure 14.

Depiction of ion channel and transport proteins in a CD principal cell.

Figure 15. Figure 15.

Apical ENaC in CCD. (A) Shown here is a representative single channel current trace for ENaC in a cell‐attached patch from a principal cell in an isolated split‐open collecting duct before and after addition of vasopressin. For this experiment, the patch was held at 0 mV, and the pipette and bath solutions contained physiological NaCl. Closed state noted with C and areas below 1 (before) and 2 (after addition of vasopressin) shown at an expanded time scale below. (B) Summary results for change in ENaC Po in response to vasopressin. Data published previously in reference 31.

Figure 16. Figure 16.

Regulation of ENaC by local autocrine/paracrine signaling systems intrinsic to the CCD. Shown here is a representative single channel current trace (A) and summary data (B) for ENaC in cell attached patches from principal cells in an isolated split‐open collecting ducts before and after sequential addition of ATP and the broad‐spectrum P2 antagonist suramin. For these paired experiment, patches were held at 0 mV, and the pipette and bath solutions contained physiological NaCl. Closed state noted with C and areas below 1, 2, and 3 shown at expanded time scale below. Data published previously in reference 255.



Figure 1.

Preparing the kidney for micropuncture. (A) After a flank incision, the left kidney is carefully freed from perirenal fat and the adrenal gland, overturned, and placed in a cup such that the dorsal kidney surface faces up (B). (C) In the rat, the ureter is catheterized for collection of urine; this is less useful in mice because of a tendency of catheters to obstruct. To immobilize the kidney, the cup is lined with moleskin (D) and cotton soaked in saline is carefully placed around the kidney. 2% to 3% agar is dripped onto the cotton, which, after cooling, prevents leakage of the superfused mineral oil or saline (37°C) thereby establishing an oil or saline layer, respectively, over the kidney surface to keep the kidney surface warm and protected. Adapted, with permission, from reference 17.



Figure 2.

Identifying tubular segments accessible to micropuncture on the kidney surface. The Bowman's space of a Munich‐Wistar‐Fromter rat is injected with small amounts of artificial tubular fluid containing a blue dye. The nephron is mapped by following the dye moving along proximal and distal segments (1‐4). Adapted, with permission, from reference 331.



Figure 3.

Reabsorption of glucose along the proximal tubule. (A) Free‐flow collections of tubular fluid were performed along accessible proximal tubules at the kidney surface to establish a profile for fractional reabsorption of glucose versus fluid. (B) Glucose is rapidly and nearly completely reabsorbed within the early proximal tubule in wild‐type mice (WT). Reabsorption of glucose is absent in the early proximal tubule of mice lacking the sodium glucose cotransporter, SGLT2 (Sglt2−/−). (C) Mean fractional reabsorption of glucose for early (<40%) and late (≥40%) proximal tubular collections and up to the urine. *P<0.001 versus WT. Adapted, with permission, from reference 337.



Figure 4.

Reabsorption of Ca2+ along the nephron. (A) Free‐flow collections of tubular fluid were performed in the last surface loop of the proximal tubule (LPT), as well as, along distal segments (DS) using luminal K+ concentration as a reference for puncturing sites. (B) Ca2+ reabsorption was unaffected in Trpv5−/− mice up to the the proximal tubule surface loop (LPT); in contrast, mean Ca2+ delivery to puncturing sites in the distal segments (DS) accessible to micropuncture as well as urine (U) was significantly enhanced in Trpv5−/− mice. *P<0.001 versus wild type (WT). (C) To directly assess Ca2+ reabsorption along the distal segments, luminal K+ concentration was used as a reference with low and high concentrations indicating early and late aspects of the DS, respectively. Whereas, the Ca2+ delivery profile indicated net reabsorption along the distal segments in WT mice, fractional Ca2+ delivery increased in Trpv5−/− mice, indicating Ca2+ back leak along the distal segments in the absence of Trpv5. Finally, comparing the fractional Ca2+ delivery in the late distal segment with the values in urine indicated significant compensation in the collecting duct of Trpv5−/− (consistent with expression of TRPV6 in that segment). Adapted, with permission, from reference 141.



Figure 5.

Role of Na+/H+ exchanger (NHE3) in proximal tubular absorption. (A) Quantitative collection of tubular fluid was performed during perfusion of superficial proximal tubular segments downstream of an obstructing wax block with control and vehicle solution with and without the addition of the unselective NHE inhibitor ethylisopropylamiloride (EIPA) or the selective NHE3 inihitor S3226. (B) Proximal tubule absorption of HCO3 (JHCO3) and fluid (Jv) were greater in wild‐type (WT) compared to NHE3 null mice (Nhe3−/−). Moreover, EIPA (Black bars) inhibits absorption in WT but not in Nhe3−/− mice. (C) S3226 induces a dose‐dependent reduction in proximal tubular absorption of fluid and sodium in normal rats. *P<0.05 versus control/vehicle. #P<0.05 versus WT. Adapted, with permission, from references 362 and 342.



Figure 6.

Role of maxi‐K (BK) channels in K+ secretion in distal nephron. (A) Stationary microperfusion has been used to study distal K+ secretion. A double‐barreled perfusion pipette containing perfusion solution (P; to identify distal surface segments) and castor oil (CO; to fill the downstream segments) was inserted in the late proximal tubule. A double‐barreled microelectrode with K+‐sensitive ion‐exchange resin (IE) and reference solution (ref) is inserted into a distal segment. Another pipette (S) is placed into the distal tubule to split the oil column and add a low K+‐control solution. The increase in luminal K+ activity is followed and analyzed as a measure of secretory K+ flux, JK. In paired experiments, the BK channel blocker IBTX is added to the tubular fluid. (B) Mice lacking the ROMK channel continue to secrete K+, albeit at a reduced rate, by an IBTX‐sensitive mechanism consistent with BK channels. (C) A high K+ diet increases distal K+ secretion primarily by an IBTX‐sensitive K+ flux. * P<0.05 versus Control. #P<0.05 versus WT or 1.2% K+ diet, respectively. Adapted, with permission, from reference 9.



Figure 7.

Isolated split‐open tubule – a segment of the cortical collecting duct isolated from rat is shown here. The middle of this segment has been split open to allow patch clamp access to the apical membranes of the lining epithelial cells. Apical membranes are shown at higher magnification in the inset.



Figure 8.

Patch clamp configurations used in the isolated split‐open tubule preparation. This illustration depicts the five different seal configurations available for probing channel activity with the voltage clamp approach in isolated split‐open tubules: three amenable to single channel recording, cell‐attached, excised inside‐out, and excised outside‐out; and two for macroscopic current recording, perforated and whole cell. In this figure, the cell is represented as a circle and the recording pipette as a V. Gray shading defines the continuity between the solution in the recording pipette and intracellular solution. Arrows represent transition steps during seal formation taken to establish the distinct types of seals. Undershown are representative current data from configurations with single channel (left) and macroscopic (right) resolutions. Closed (C) and open (O) states are noted in the single channel trace. Data adapted, with permission, from J.D. Stockand or published previously in reference 256.



Figure 9.

SK channel in TAL epithelial cells. Shown is a typical single channel current trace of an SK channel in an inside‐out patch held at hyperpolarizing test potentials made from the apical membrane of an epithelial cell lining a split‐open murine TAL. Patch solutions contained 140/5 mmol/L K+ in the pipette/bath. Inward K+ current is downward. Test voltages and closed state are noted in the figure. Figure originally presented in reference 204.



Figure 10.

IK channel in TAL epithelial cells. Shown is a typical single channel current trace of an IK channel in a cell attached patch held on the apical membrane of an epithelial cell lining a split‐open rat TAL. For this experiment, pipette potential was 0 mV and the recording pipette contained 140 mmol/L KCl. Closed state noted with C. Data provided, with permission, by W‐H. Wang.



Figure 11.

Depiction of ion channel and transport proteins in a TAL epithelial cell.



Figure 12.

Basolateral Cl channels in TAL cells. Shown are typical single channel current traces of the small (∼10 pS; A) and the double‐barreled (B) Cl channels in cell attached patches formed on the basolateral membranes of epithelial cells in isolated, split‐open TAL. For the current trace containing only the small channel (A), the bath and pipette contained symmetrical NaCl and the patch was exposed to hyperpolarizing test pulses. The patch in B contains both the larger double‐barreled channel, as well as, the smaller Cl channel (openings for the smaller channel noted with arrows). This patch was held at 50 and −80 mV test potentials also with symmetrical bath and pipette solutions. Closed state noted with C. Data in A and B originally published in references 125 and 127, respectively.



Figure 13.

Apical BK channel in cortical collecting duct (CCD). Shown here is a representative single channel current trace for a BK channel in the absence and presence of exogenous calcium in an excised, inside‐out patch from a principal cell in an isolated split‐open collecting duct. For this experiment, the patch was held at 0 mV, and the pipette and bath solutions contained 140 and 5 mmol/L KCl, respectively. Closed state noted with C. Data adapted, with permission, from W‐H Wang.



Figure 14.

Depiction of ion channel and transport proteins in a CD principal cell.



Figure 15.

Apical ENaC in CCD. (A) Shown here is a representative single channel current trace for ENaC in a cell‐attached patch from a principal cell in an isolated split‐open collecting duct before and after addition of vasopressin. For this experiment, the patch was held at 0 mV, and the pipette and bath solutions contained physiological NaCl. Closed state noted with C and areas below 1 (before) and 2 (after addition of vasopressin) shown at an expanded time scale below. (B) Summary results for change in ENaC Po in response to vasopressin. Data published previously in reference 31.



Figure 16.

Regulation of ENaC by local autocrine/paracrine signaling systems intrinsic to the CCD. Shown here is a representative single channel current trace (A) and summary data (B) for ENaC in cell attached patches from principal cells in an isolated split‐open collecting ducts before and after sequential addition of ATP and the broad‐spectrum P2 antagonist suramin. For these paired experiment, patches were held at 0 mV, and the pipette and bath solutions contained physiological NaCl. Closed state noted with C and areas below 1, 2, and 3 shown at expanded time scale below. Data published previously in reference 255.

References
 1. Adrian RH. The effect of internal and external potassium concentration on the membrane potential of frog muscle. J Physiol 133: 631‐658, 1956.
 2. Ahn D, Ge Y, Stricklett PK, Gill P, Taylor D, Hughes AK, Yanagisawa M, Miller L, Nelson RD, Kohan DE. Collecting duct‐specific knockout of endothelin‐1 causes hypertension and sodium retention. J Clin Invest 114: 504‐511, 2004.
 3. Amorim JB, Bailey MA, Musa‐Aziz R, Giebisch G, Malnic G. Role of luminal anion and pH in distal tubule potassium secretion. Am J Physiol Renal Physiol 284: F381‐F388, 2003.
 4. Amorim JB, Malnic G. V(1) receptors in luminal action of vasopressin on distal K(+) secretion. Am J Physiol Renal Physiol 278: F809‐F816, 2000.
 5. Andreucci VE, Herrera‐Acosta J, Rector FC, Jr. and Seldin DW. Measurement of single‐nephron glomerular filtration rate by micropuncture: Analysis of error. Am J Physiol 221: 1551‐1559, 1971.
 6. Ares GR, Caceres P, varez‐Leefmans FJ, Ortiz PA. cGMP decreases surface NKCC2 levels in the thick ascending limb: Role of phosphodiesterase 2 (PDE2). Am J Physiol Renal Physiol 295: F877‐F887, 2008.
 7. Babilonia E, Li D, Wang Z, Sun P, Lin DH, Jin Y, Wang WH. Mitogen‐activated protein kinases inhibit the ROMK (Kir 1.1)‐like small conductance K channels in the cortical collecting duct. J Am Soc Nephrol 17: 2687‐2696, 2006.
 8. Babilonia E, Wei Y, Sterling H, Kaminski P, Wolin M, Wang WH. Superoxide anions are involved in mediating the effect of low K intake on c‐Src expression and renal K secretion in the cortical collecting duct. J Biol Chem 280: 10790‐10796, 2005.
 9. Bailey MA, Cantone A, Yan Q, Macgregor GG, Leng Q, Amorim JB, Wang T, Hebert SC, Giebisch G, Malnic G. Maxi‐K channels contribute to urinary potassium excretion in the ROMK‐deficient mouse model of Type II Bartter's syndrome and in adaptation to a high‐K diet. Kidney Int 70: 51‐59, 2006.
 10. Bailey MA, Giebisch G, Abbiati T, Aronson PS, Gawenis LR, Shull GE, Wang T. NHE2‐mediated bicarbonate reabsorption in the distal tubule of NHE3 null mice. J Physiol 561: 765‐775, 2004.
 11. Bailey MA, Unwin RJ, Shirley DG. In vivo inhibition of renal 11beta‐hydroxysteroid dehydrogenase in the rat stimulates collecting duct sodium reabsorption. Clin Sci (Lond) 101: 195‐198, 2001.
 12. Balaban RS, Dennis VW, Mandel LJ. Microfluorometric monitoring of NAD redox state in isolated perfused renal tubules. Am J Physiol 240: F337‐F342, 1981.
 13. Barratt LJ, Rector FC, Jr., Kokko JP, Seldin DW. Factors governing the transepithelial potential difference across the proximal tubule of the rat kidney. J Clin Invest 53: 454‐464, 1974.
 14. Beck JS, Hurst AM, Lapointe J‐Y, Laprade R. Regulation of basolateral K channels in proximal tubule studied during continuous microperfusion. Am J Physiol 264: F496‐F501, 1993.
 15. Bell PD, Peti‐Peterdi J. Angiotensin II stimulates macula densa basolateral sodium/hydrogen exchange via type 1 angiotensin II receptors. J Am Soc Nephrol 10(Suppl 11): S225‐S229, 1999.
 16. Biagi B, Kubota T, Sohtell M, Giebisch G. Intracellular potentials in rabbit proximal tubules perfused in vitro. Am J Physiol 240: F200‐F210, 1981.
 17. Blantz RC, Tucker BJ. Measurements of glomerular dynamics. In: Martinez‐Maldonado, editor. Methods in Pharmacology. Plenum Publishing Corporation, 1978, pp. 141‐163.
 18. Bleich M, Kottgen M, Schlatter E, Greger R. Effect of NH4+/NH3 on cytosolic pH and the K+ channels of freshly isolated cells from the thick ascending limb of Henle's loop. Pflugers Arch 429: 345‐354, 1995.
 19. Bleich M, Schlatter E, Greger R. The luminal K+ channel of the thick ascending limb of Henle's loop. Pflugers Arch 415: 449‐460, 1990.
 20. Bomsztyk K, Calalb MB. A new microelectrode method for simultaneous measurement of pH and PCO2. Am J Physiol 251: F933‐F937, 1986.
 21. Bomsztyk K, George JP, Wright FS. Effects of luminal fluid anions on calcium transport by proximal tubule. Am J Physiol 246: F600‐F608, 1984.
 22. Bonny O, Hummler E. Dysfunction of epithelial sodium transport: From human to mouse. Kidney Int 57: 1313‐1318, 2000.
 23. Booth RE, Johnson JP, Stockand JD. Aldosterone. Adv Physiol Educ 26: 8‐20, 2002.
 24. Boron WF, Boulpaep EL. Intracellular pH regulation in the renal proximal tubule of the Salamander. Na‐H exchange. J Gen Physiol 81: 29‐52, 1983a.
 25. Boron WF, Boulpaep EL. Intracellular pH regulation in the renal proximal tubule of the Salamander.Basolateral HCO3‐ transport. J Gen Physiol 81: 53‐94, 1983b.
 26. Boulpaep EL. Electrophysiological techniques in kidney micropuncture. Yale J Biol Med 45: 397‐413, 1972.
 27. Boulpaep EL, Seely JF. Electrophysiology of proximal and distal tubules in the autoperfused dog kidney. Am J Physiol 221: 1084‐1096, 1971.
 28. Bourdeau JE, Carone FA, Ganote CE. Serum albumin uptake in isolated perfused renal tubules. Quantitative and electron microscope radioautographic studies in three anatomical segments of the rabbit nephron. J Cell Biol 54: 382‐398, 1972.
 29. Braam B, Mitchell KD, Fox J, Navar LG. Proximal tubular secretion of angiotensin II in rats. Am J Physiol 264: F891‐F898, 1993.
 30. Bugaj V, Pochynyuk O, Mironova E, Vandewalle A, Medina JL, Stockand JD. Regulation of the epithelial Na+ channel by endothelin‐1 in rat collecting duct. Am J Physiol Renal Physiol 295: F1063‐F1070, 2008.
 31. Bugaj V, Pochynyuk O, Stockand JD. Activation of the epithelial Na+ channel in the collecting duct by vasopressin contributes to water reabsorption. Am J Physiol Renal Physiol 297: F1411‐F1418, 2009.
 32. Burg M, Grantham J, Abramow M, Orloff J. Preparation and study of fragments of single rabbit nephrons. Am J Physiol 210: 1293‐1298, 1966.
 33. Burnstock G. Purinergic signalling and disorders of the central nervous system. Nat Rev Drug Discov 7: 575‐590, 2008.
 34. Busjahn A, Aydin A, Uhlmann R, Krasko C, Bahring S, Szelestei T, Feng Y, Dahm S, Sharma AM, Luft FC, Lang F. Serum‐ and glucocorticoid‐regulated kinase (SGK1) gene and blood pressure. Hypertension 40: 256‐260, 2002.
 35. Caceres PS, Ares GR, Ortiz PA. cAMP stimulates apical exocytosis of the renal Na(+)‐K(+)‐2Cl[‐] cotransporter NKCC2 in the thick ascending limb: Role of protein kinase A. J Biol Chem 284: 24965‐24971, 2009.
 36. Canessa CM, Horisberger JD, Rossier BC. Epithelial sodium channel related to proteins involved in neurodegeneration. Nature 361: 467‐470, 1993.
 37. Canessa CM, Schild L, Buell G, Thorens B, Gautschi I, Horisberger JD, Rossier BC. Amiloride‐sensitive epithelial Na channel is made of three homologous subunits. Nature 367: 463‐467, 1994.
 38. Cantone A, Yang X, Yan Q, Giebisch G, Hebert SC, Wang T. Mouse model of type II Bartter's syndrome. I. Upregulation of thiazide‐sensitive Na‐Cl cotransport activity. Am J Physiol Renal Physiol 294: F1366‐F1372, 2008.
 39. Carey RM, Douglas JG, Schweikert JR, Liddle GW. The syndrome of essential hypertension and suppressed plasma renin activity. Normalization of blood pressure with spironolactone. Arch Intern Med 130: 849‐854, 1972.
 40. Carone FA, Pullman TN, Oparil S, Nakamura S. Micropuncture evidence of rapid hydrolysis of bradykinin by rat proximal tubule. Am J Physiol 230: 1420‐1424, 1976.
 41. Cassola AC, Giebisch G, Wang W. Vasopressin increases density of apical low‐conductance K+ channels in rat CCD. Am J Physiol 264: F502‐F509, 1993.
 42. Chaillet JR, Boron WF. Intracellular calibration of a pH‐sensitive dye in isolated, perfused salamander proximal tubules. J Gen Physiol 86: 765‐794, 1985.
 43. Chang SS, Grunder S, Hanukoglu A, Rosler A, Mathew PM, Hanukoglu I, Schild L, Lu Y, Shimkets RA, Nelson‐Williams C, Rossier BC, Lifton RP. Mutations in subunits of the epithelial sodium channel causes salt wasting with hyperkalaemic acidosis, pseudohypoaldosteronism type 1. Nature Genet 12: 248‐253, 1996.
 44. Cheema‐Dhadli S, Hamat R, Sonnenberg H, Halperin M. A micromethod to measure ammonia. Kidney Int 19: 80‐82, 1981.
 45. Chen L, Williams SK, Schafer JA. Differences in synergistic actions of vasopressin and deoxycorticosterone in rat and rabbit CCD. Am J Physiol 259: F147‐F156, 1990.
 46. Chen S, Agarwal A, Glushakova OY, Jorgensen MS, Salgar SK, Poirier A, Flotte TR, Croker BP, Madsen KM, Atkinson MA, Hauswirth WW, Berns KI, Tisher CC. Gene delivery in renal tubular epithelial cells using recombinant adeno‐associated viral vectors. J Am Soc Nephrol 14: 947‐958, 2003.
 47. Chen S, Bhargava A, Meijer O, Rozansky D, Spindler B, Verrey F, Pearce D. Identification and characterization of novel mineralocorticoid‐regulated genes in tight epithelia. J Am Soc Nephrol 9(A2030): 398A, 1998.
 48. Chen S, Bhargava S, Mastroberardino L, Meijer OC, Wang J, Firestone P, Verrey F, Pearce D. Epithelial sodium channel regualted by aldosterone‐induced protein sgk. Proc Nat Acad Sci U S A 96: 2514‐2519, 1999.
 49. Cheng QL, Chen XM, Li F, Lin HL, Ye YZ, Fu B. Effects of ICAM‐1 antisense oligonucleotide on the tubulointerstitium in mice with unilateral ureteral obstruction. Kidney Int 57: 183‐190, 2000.
 50. Colindres RE, Kramp RA, Allison ME, Gottschalk CW. Hydrodynamic alterations during distal tubular fluid collections in the rat kidney. Am J Physiol 232: F497‐F506, 1977.
 51. Cortell S. Silicone rubber for renal tubular injection. J Appl Physiol 26: 158‐159, 1969.
 52. Costanzo LS. Comparison of calcium and sodium transport in early and late rat distal tubules: Effect of amiloride. Am J Physiol 246: F937‐F945, 1984.
 53. Darling IM, Morris ME. Evaluation of “true” creatinine clearance in rats reveals extensive renal secretion. Pharm Res 8: 1318‐1322, 1991.
 54. Davidman M, Lalone RC, Alexander EA, Levinsky NG. Some micropuncture techniques in the rat. Am J Physiol 221: 1110‐1114, 1971.
 55. Deen WM, Robertson CR, Brenner BM. A model of glomerular ultrafiltration in the rat. Am J Physiol 223: 1178‐1183, 1972.
 56. Dhaun N, Goddard J, Kohan DE, Pollock DM, Schiffrin EL, Webb DJ. Role of endothelin‐1 in clinical hypertension: 20 years on. Hypertension 52: 452‐459, 2008.
 57. Dunn SR, Qi Z, Bottinger EP, Breyer MD, Sharma K. Utility of endogenous creatinine clearance as a measure of renal function in mice. Kidney Int 65: 1959‐1967, 2004.
 58. Eisner C, Faulhaber‐Walter R, Wang Y, Leelahavanichkul A, Yuen PS, Mizel D, Star RA, Briggs JP, Levine M, Schnermann J. Major contribution of tubular secretion to creatinine clearance in mice. Kidney Int 77: 519‐526, 2010.
 59. Estevez R, Boettger T, Stein V, Birkenhager R, Otto E, Hildebrandt F, Jentsch TJ. Barttin is a Cl‐ channel beta‐subunit crucial for renal Cl‐ reabsorption and inner ear K+ secretion. Nature 414: 558‐561, 2001.
 60. Faria NJ, Dobbie H, Slater JM, Shirley DG, Stocking CJ, Unwin RJ. Simultaneous determination of anions in nanoliter volumes. Kidney Int 67: 357‐363, 2005.
 61. Fejes‐Toth G, Frindt G, Naray‐Fejes‐Toth A, Palmer LG. Epithelial Na+ channel activation and processing in mice lacking SGK1. Am J Physiol Renal Physiol 294: F1298‐F1305, 2008.
 62. Fenton RA, Chou CL, Ageloff S, Brandt W, Stokes JB, Knepper MA. Increased collecting duct urea transporter expression in Dahl salt‐sensitive rats. Am J Physiol Renal Physiol 285: F143‐F151, 2003.
 63. Fenton RA, Knepper MA. Mouse models and the urinary concentrating mechanism in the new millennium. Physiol Rev 87: 1083‐1112, 2007.
 64. Flores SY, Loffing‐Cueni D, Kamynina E, Daidie D, Gerbex C, Chabanel S, Dudler J, Loffing J, Staub O. Aldosterone‐induced serum and glucocorticoid‐induced kinase 1 expression is accompanied by Nedd4‐2 phosphorylation and increased Na+ transport in cortical collecting duct cells. J Am Soc Nephrol 16: 2279‐2287, 2005.
 65. Fouladkou F, ikhani‐Koopaei R, Vogt B, Flores SY, Malbert‐Colas L, Lecomte MC, Loffing J, Frey FJ, Frey BM, Staub O. A naturally occurring human Nedd4‐2 variant displays impaired ENaC regulation in Xenopus laevis oocytes. Am J Physiol Renal Physiol 287: F550‐F561, 2004.
 66. Frindt G, Burg MB. Effect of Vasopressin on sodium transport in renal cortical collecting tubules. Kidney International 1: 224‐231, 1972.
 67. Frindt G, Ergonul Z, Palmer LG. Na channel expression and activity in the medullary collecting duct of rat kidney. Am J Physiol Renal Physiol 292: F1190‐F1196, 2007.
 68. Frindt G, Masilamani S, Knepper MA, Palmer LG. Activation of epithelial Na channels during short‐term Na deprivation. Am J Physiol Renal Physiol 280: F112‐F118, 2001.
 69. Frindt G, McNair T, Dahlmann A, Jacobs‐Palmer E, Palmer LG. Epithelial Na channels and short‐term renal response to salt deprivation. Am J Physiol Renal Physiol 283: F717‐F726, 2002.
 70. Frindt G, Palmer LG. Ca‐activated K channels in apical membrane of mammalian CCT, and their role in K secretion. Am J Physiol 252: 458‐467, 1987.
 71. Frindt G, Palmer LG. Low‐conductance K channels in apical membrane of rat cortical collecting tubule. Am J Physiol 256: F143‐F151, 1989.
 72. Frindt G, Palmer LG. Regulation of Na channels in the rat cortical collecting tubule: Effects of cAMP and methyl donors. Am J Physiol 271: F1086‐F1092, 1996.
 73. Frindt G, Palmer LG. Apical potassium channels in the rat connecting tubule. Am J Physiol Renal Physiol 287: F1030‐F1037, 2004.
 74. Frindt G, Palmer LG, Windhager EE. Feedback regulation of Na channels in rat CCT. IV. Mediation by activation of protein kinase C. Am J Physiol 270: F371‐F376, 1996.
 75. Frindt G, Sackin H, Palmer LG. Whole‐cell currents in rat cortical collecting tubule: Low‐Na diet increases amiloride‐sensitive conductance. Am J Physiol Renal,Fluid Electrolyte Physiol 258: F562‐F567, 1990.
 76. Frindt G, Shah A, Edvinsson J, Palmer LG. Dietary K regulates ROMK channels in connecting tubule and cortical collecting duct of rat kidney. Am J Physiol Renal Physiol 296: F347‐F354, 2009.
 77. Frindt G, Silver RB, Windhager EE, Palmer LG. Feedback regulation of Na channels in rat CCT. II. Effects of inhibition of Na entry. Am J Physiol 264: F565‐F574, 1993.
 78. Frindt G, Zhou H, Sackin H, Palmer LG. Dissociation of K channel density and ROMK mRNA in rat cortical collecting tubule during K adaptation. Am J Physiol 274: F525‐F531, 1998.
 79. Fritzsch G, Haase W, Rumrich G, Fasold H, Ullrich KJ. A stopped flow capillary perfusion method to evaluate contraluminal transport parameters of methylsuccinate from interstitium into renal proximal tubular cells. Pflugers Arch 400: 250‐256, 1984.
 80. Fromter E. Electrophysiological analysis of rat renal sugar and amino acid transport. I. Basic phenomena. Pflugers Arch 393: 179‐189, 1982.
 81. Fromter E. The electrophysiological analysis of tubular transport. Kidney Int 30: 216‐228, 1986.
 82. Fromter E, Gessner K. Active transport potentials, membrane diffusion potentials and streaming potentials across rat kidney proximal tubule. Pflugers Arch 351: 85‐98, 1974.
 83. Fromter E, Gessner K. Free‐flow potential profile along rat kidney proximal tubule. J Am Soc Nephrol 12: 2197‐2206, 2001.
 84. Fromter E, Muller CW, Wick T. Permeability properties of proximal tubular epithelium of the rat kidney studied with electrophysiological methods. In: Giebisch G, editor. Electrophysiology of Epithelial Cells. Stuttgart: Schartauer, 1971, pp. 119‐146.
 85. Ganote CE, Grantham JJ, Moses HL, Burg MB, Orloff J. Ultrastructural studies of vasopressin effect on isolated perfused renal collecting tubules of the rabbit. J Cell Biol 36: 355‐367, 1968.
 86. Garcia NH, Plato CF, Garvin JL. Fluorescent determination of chloride in nanoliter samples. Kidney Int 55: 321‐325, 1999.
 87. Garcia NH, Plato CF, Stoos BA, Garvin JL. Nitric oxide‐induced inhibition of transport by thick ascending limbs from Dahl salt‐sensitive rats. Hypertension 34: 508‐513, 1999.
 88. Garty H, Palmer LG. Epithelial sodium channels: function, structure, and regulation. [Review] [451 refs]. Physiological Reviews 77: 359‐396, 1997.
 89. Garvin JL. A simple method to determine millimolar concentrations of sodium in nanoliter samples. Kidney Int 44: 875‐880, 1993.
 90. Garvin JL. Glucose absorption by isolated perfused rat proximal straight tubules. Am J Physiol 259: F580‐F586, 1990.
 91. Garvin JL, Burg MB, Knepper MA. Ammonium replaces potassium in supporting sodium transport by the Na‐K‐ATPase of renal proximal straight tubules. Am J Physiol 249: F785‐F788, 1985.
 92. Garvin JL, Knepper MA. Bicarbonate and ammonia transport in isolated perfused rat proximal straight tubules. Am J Physiol 253: F277‐F281, 1987.
 93. Ge Y, Ahn D, Stricklett PK, Hughes AK, Yanagisawa M, Verbalis JG, Kohan DE. Collecting duct‐specific knockout of endothelin‐1 alters vasopressin regulation of urine osmolality. Am J Physiol Renal Physiol 288: F912‐F920, 2005.
 94. Ge Y, Bagnall A, Stricklett PK, Strait K, Webb DJ, Kotelevtsev Y, Kohan DE. Collecting duct‐specific knockout of the endothelin B receptor causes hypertension and sodium retention. Am J Physiol Renal Physiol 291: F1274‐F1280, 2006.
 95. Ge Y, Bagnall A, Stricklett PK, Webb D, Kotelevtsev Y, Kohan DE. Combined knockout of collecting duct endothelin A and B receptors causes hypertension and sodium retention. Am J Physiol Renal Physiol 295: F1635‐F1640, 2008.
 96. Geibel J, Volkl H, Lang F. A microelectrode for continuous recording of volume fluxes in isolated perfused tubule segments. Pflugers Arch 400: 388‐392, 1984.
 97. Geibel J, Zweifach A, White S, Wang WH, Giebisch G. K+ channels of the mammalian collecting duct. Ren Physiol Biochem 13: 59‐69, 1990.
 98. Geller DS, Farhi A, Pinkerton N, Fradley M, Moritz M, Spitzer A, Meinke G, Tsai FTF, Sigler PB, and Lifton RP. Activating mineralocorticoid receptor mutation in hypertension exacerbated by pregnancy. Science 289, 119‐123, 2000.
 99. Geller DS, Rodriguez‐Soriano J, Vallo BA, Schifter S, Bayer M, Chang SS, Lifton RP. Mutations in the mineralocorticoid receptor gene cause autosomal dominant pseudohypoaldosteronism type I. Nat Genet 19: 279‐281, 1998.
 100. Gertz KH. Transtubulare Natriumchloridfluesse und Permeabilitaet fuer Nichtelektrolyte im proximalen und distalen Konvolut der Rattenniere. Pflugers Arch 276: 336‐356, 1963.
 101. Gertz KH, Ullrich KJ. Methode zur Analyse des Stofftransportes am einzelnen Tubulus der intakten Rattenniere. Pflugers Arch 274: 61‐62, 1961.
 102. Geyti CS, Odgaard E, Overgaard MT, Jensen ME, Leipziger J, Praetorius HA. Slow spontaneous [Ca2+] i oscillations reflect nucleotide release from renal epithelia 2. Pflugers Arch 455: 1105‐1117, 2008.
 103. Giebisch G, Hunter M, Kawahara K. Apical potassium channels in Amphiuma diluting segment: Effect of barium. J Physiol (Lond) 420: 313‐323, 1990.
 104. Gogelein H. Chloride channels in epithelia. Biochim Biophys Acta 947: 521‐547, 1988.
 105. Gogelein H. Ion channels in mammalian proximal renal tubules. Ren Physiol Biochem 13: 8‐25, 1990.
 106. Gogelein H, Greger R. Single channel recordings from basolateral and apical membranes of renal proximal tubules. Pflugers Arch 401: 424‐426, 1984.
 107. Gogelein H, Greger R. Na+ selective channels in the apical membrane of rabbit late proximal tubules (pars recta). Pflugers Arch 406: 198‐203, 1986.
 108. Gogelein H, Greger R. Properties of single channels in the basolateral membrane of rabbit proximal straight tubules. Pflugers Arch 410: 288‐295, 1987.
 109. Good DW, Vurek GG. Picomole quantitation of ammonia by flow‐through fluorometry. Anal Biochem 130: 199‐202, 1983.
 110. Good DW, Wright FS. Luminal influences on potassium secretion: Transepithelial voltage. Am J Physiol 239: F289‐F298, 1980.
 111. Gottschalk CW. A history of renal physiology to 1950. In: Seldin DW, Giebisch G, editors. The Kidney. Physiology and Pathophysiology. New York: Raven, 1992, pp. 1‐29.
 112. Gottschalk CW, Morel F, Mylle M. Tracer microinjection studies of renal tubular permeability. Am J Physiol 209: 173‐178, 1965.
 113. Gottschalk CW, Mylle M. Micropuncture study of the mammalian urinary concentrating mechanism: Evidence for the countercurrent hypothesis. Am J Physiol 196: 927‐936, 1959.
 114. Grantham JJ. Mode of water transport in mammalian renal collecting tubules. Fed Proc 30: 14‐21, 1971.
 115. Grantham JJ, Burg MB. Effect of vasopressin and cyclic AMP on permeability of isolated collecting tubules 6. Am J Physiol 211: 255‐259, 1966.
 116. Grantham JJ, Ganote CE, Burg MB, Orloff J. Paths of transtubular water flow in isolated renal collecting tubules. J Cell Biol 41: 562‐576, 1969.
 117. Grantham JJ, Kurg MB, Obloff J. The nature of transtubular Na and K transport in isolated rabbit renal collecting tubules. J Clin Invest 49: 1815‐1826, 1970.
 118. Gray DA, Frindt G, Palmer LG. Quantification of K+ secretion through apical low‐conductance K channels in the CCD. Am J Physiol Renal Physiol 289: F117‐F126, 2005.
 119. Gray DA, Frindt G, Zhang YY, Palmer LG. Basolateral K+ conductance in principal cells of rat CCD. Am J Physiol Renal Physiol 288: F493‐F504, 2005.
 120. Greger R, Bleich M, Schlatter E. Ion channels in the thick ascending limb of Henle's loop. Ren Physiol Biochem 13: 37‐50, 1990.
 121. Greger R, Schlatter E. Presence of luminal K+, a prerequisite for active NaCl transport in the cortical thick ascending limb of Henle's loop of rabbit kidney. Pflugers Arch 392: 92‐94, 1981.
 122. Gross V, Luft FC. Adapting renal and cardiovascular physiology to the genetically hypertensive mouse. Semin Nephrol 22: 172‐179, 2002.
 123. Grunder S, Firsov D, Chang SS, Jaeger NF, Gautschi I, Schild L, Lifton RP, Rossier BC. A mutation causing pseudohypoaldosteronism type 1 identifies a conserved glycine that is involved in the gating of the epithelial sodium channel. EMBO J 16: 899‐907, 1997.
 124. Gu RM, Yang L, Zhang Y, Wang L, Kong S, Zhang C, Zhai Y, Wang M, Wu P, Liu L, Gu F, Zhang J, Wang WH. CYP‐omega‐hydroxylation‐dependent metabolites of arachidonic acid inhibit the basolateral 10 pS chloride channel in the rat thick ascending limb. Kidney Int 76: 849‐856, 2009.
 125. Guggino WB, London R, Boulpaep EL, Giebisch G. Chloride transport across the basolateral cell membrane of the Necturus proximal tubule: Dependence on bicarbonate and sodium. J Membr Biol 71: 227‐240, 1983.
 126. Guinamard R, Chraibi A, Teulon J. A small‐conductance Cl‐ channel in the mouse thick ascending limb that is activated by ATP and protein kinase A. J Physiol 485(Pt 1): 97‐112, 1995.
 127. Guzman RJ, Lemarchand P, Crystal RG, Epstein SE, Finkel T. Efficient gene transfer into myocardium by direct injection of adenovirus vectors. Circ Res 73: 1202‐1207, 1993.
 128. Gyory AZ. Reexamination of the split oil droplet methodas applied to kidney tubules. Pflugers Arch 324: 328‐343, 1975.
 129. Hamill OP, Marty A, Neher E, Sakmann B, Sigworth FJ. Improved patch‐clamp techniques for high‐resolution current recording from cells and cell‐free membrane patches. Pflugers Arch 391: 85‐100, 1981.
 130. Hansson JH, Nelson‐Williams C, Suzuki H, Schild L, Shimkets R, Lu Y, Canessa C, Iwasaki T, Rossier B, Lifton RP. Hypertension caused by a truncated epithelial sodium channel gamma subunit: Genetic heterogeneity of Liddle syndrome. Nat Genet 11: 76‐82, 1995.
 131. Hawk CT, Li L, Schafer JA. AVP and aldosterone at physiological concentrations have synergistic effects on Na+ transport in rat CCD. Kidney Int Suppl 57: S35‐S41, 1996.
 132. Helman SI, Grantham JJ, Burg MB. Effect of vasopressin on electrical resistance of renal cortical collecting tubules. Am J Physiol 220: 1825‐1832, 1971.
 133. Helman SI, Koeppen BM, Beyenbach KW, Baxendale LM. Patch clamp studies of apical membranes of renal cortical collecting ducts. Pflugers Arch 405(Suppl 1): S71‐S76, 1985.
 134. Herget S, Lohse MJ, Nikolaev VO. Real‐time monitoring of phosphodiesterase inhibition in intact cells. Cell Signal 20: 1423‐1431, 2008.
 135. Herrera M, Hong NJ, Ortiz PA, Garvin JL. Endothelin‐1 inhibits thick ascending limb transport via Akt‐stimulated nitric oxide production. J Biol Chem 284: 1454‐1460, 2009.
 136. Hirsch J, Leipziger J, Frobe U, Schlatter E. Regulation and possible physiological role of the Ca(2+)‐dependent K+ channel of cortical collecting ducts of the rat. Pflugers Arch 422: 492‐498, 1993.
 137. Hirsch J, Schlatter E. K+ channels in the basolateral membrane of rat cortical collecting duct. Pflugers Arch 424: 470‐477, 1993.
 138. Hirsch J, Schlatter E. K+ channels in the basolateral membrane of rat cortical collecting duct are regulated by a cGMP‐dependent protein kinase. Pflugers Arch 429: 338‐344, 1995.
 139. Ho K, Nichols CG, Lederer WJ, Lytton J, Vassilev PM, Kanazirska MV, Hebert SC. Cloning and expression of an inwardly rectifying ATP‐regulated potassium channel. Nature 362: 31‐38, 1993.
 140. Hoenderop JG, van Leeuwen JP, van der Eerden BC, Kersten FF, van der Kemp AW, Merillat AM, Waarsing JH, Rossier BC, Vallon V, Hummler E, Bindels RJ. Renal Ca2+ wasting, hyperabsorption, and reduced bone thickness in mice lacking TRPV5. J Clin Invest 112: 1906‐1914, 2003.
 141. Hong NJ, Garvin JL. Flow increases superoxide production by NADPH oxidase via activation of Na‐K‐2Cl cotransport and mechanical stress in thick ascending limbs. Am J Physiol Renal Physiol 292: F993‐F998, 2007.
 142. Hong NJ, Silva GB, Garvin JL. PKC‐alpha mediates flow‐stimulated superoxide production in thick ascending limbs. Am J Physiol Renal Physiol 298: F885‐F891, 2010.
 143. Hou J, Renigunta A, Gomes AS, Hou M, Paul DL, Waldegger S, Goodenough DA. Claudin‐16 and claudin‐19 interaction is required for their assembly into tight junctions and for renal reabsorption of magnesium. Proc Natl Acad Sci U S A 106: 15350‐15355, 2009.
 144. Hou J, Shan Q, Wang T, Gomes AS, Yan Q, Paul DL, Bleich M, Goodenough DA. Transgenic RNAi depletion of claudin‐16 and the renal handling of magnesium. J Biol Chem 282: 17114‐17122, 2007.
 145. Huang DY, Osswald H, Vallon V. Intratubular application of sodium azide inhibits loop of Henle reabsorption and tubuloglomerular feedback response in anesthetized rats. Naunyn Schmiedebergs Arch Pharmacol 358: 367‐373, 1998.
 146. Huang DY, Osswald H, Vallon V. Eukaliuric diuresis and natriuresis in response to the KATP channel blocker U37883A: Micropuncture studies on the tubular site of action. Br J Pharmacol 127: 1811‐1818, 1999.
 147. Huang DY, Osswald H, Vallon V. Sodium reabsorption in thick ascending limb of Henle's loop: Effect of potassium channel blockade in vivo. Br J Pharmacol 130: 1255‐1262, 2000.
 148. Huang DY, Wulff P, Volkl H, Loffing J, Richter K, Kuhl D, Lang F, Vallon V. Impaired regulation of renal K+ elimination in the sgk1‐knockout mouse. J Am Soc Nephrol 15: 885‐891, 2004.
 149. Hummler E. Implication of ENaC in salt‐sensitive hypertension. J Steroid Biochem Mol Biol 69: 385‐390, 1999.
 150. Hummler E, Horisberger JD. Genetic disorders of membrane transport. V. The epithelial sodium channel and its implication in human diseases. Am J Physiol 276: G567‐G571, 1999.
 151. Hunter M, Kawahara K, Giebisch G. Potassium channels along the nephron. Fed Proc 45: 2723‐2726, 1986.
 152. Hunter M, Kawahara K, Giebisch G. Calcium‐activated epithelial potassium channels. Miner Electrolyte Metab 14: 48‐57, 1988.
 153. Hunter M, Lopes AG, Boulpaep EL, Cohen B, Giebisch G. Single channel recordings of calcium‐activated Potassium channels in the apical membrane of rabbit cortical collecting tubules. Proc Natl Acad Sci U S A 81: 4237‐4239, 1984.
 154. Hunter M, Lopes AG, Boulpaep EL, Giebisch GH. Regulation of single potassium ion channels from apical membrane of rabbit collecting tubule. Am J Physiol 251: 725‐733, 1986.
 155. Hurst AM, Beck JS, Laprade R, Lapointe J‐Y. Na+ pump inhibition downregulates an ATP‐sensitive K+ channel in rabbit proximal convoluted tubule. Am J Physiol 264: F760‐F764, 1993.
 156. Hurst AM, Duplain M, Lapointe J‐Y. Basolateral membrane potassium channels in rabbit cortical thick ascending limb. Am J Physiol 263: F262‐F267, 1992.
 157. Hurst AM, Lapointe J‐Y, Laamarti A, Bell PD. Basic properties and potential regulators of the apical K+ channel in macula densa cells. J Gen Physiol 103: 1055‐1070, 1994.
 158. Imai M, Kokko JP. Sodium chloride, urea, and water transport in the thin ascending limb of Henle. Generation of osmotic gradients by passive diffusion of solutes. J Clin Invest 53: 393‐402, 1974.
 159. Ito K, Chen J, Khodadadian JJ, Vaughan ED Jr., Lipkowitz M, Poppas DP, Felsen D. Adeno‐associated viral vector transduction of green fluorescent protein in kidney: Effect of unilateral ureteric obstruction. BJU Int 101: 376‐381, 2008.
 160. Jamison RL. Micropuncture study of superficial and juxtamedullary nephrons in the rat. Am J Physiol 218: 46‐55, 1970.
 161. Jensen ME, Odgaard E, Christensen MH, Praetorius HA, Leipziger J. Flow‐induced [Ca2+]i increase depends on nucleotide release and subsequent purinergic signaling in the intact nephron. J Am Soc Nephrol 18: 2062‐2070, 2007.
 162. Karlmark B, Jaeger P, Fein H, Giebisch G. Coulometric acid‐base titration in nanoliter samples with glass and antimony electrodes. Am J Physiol 242: F95‐F99, 1982.
 163. Kawahara K, Hunter M, Giebisch G. Potassium channels in necturus proximal tubule. Am J Physiol 253: F488‐F494, 1987.
 164. Kellenberger S, Schild L. Epithelial sodium channel/degenerin family of ion channels: A variety of functions for a shared structure. Physiol Rev 82: 735‐767, 2002.
 165. Kibble JD, Audsley N, Day JP, Green R. A new protocol for the measurement of picomole quantities of magnesium in rat renal tubular fluid. Exp Physiol 83: 11‐22, 1998.
 166. Koeppen BM. Electrophysiology of ion transport in renal tubule epithelia. Seminars in Nephrology 7: 37‐47, 1987.
 167. Koeppen BM, Beyenbach KW, Helman SI. Single‐channel current in renal tubules. Am J Physiol 247: F380‐F384, 1984.
 168. Kohan DE. Endothelins in the normal and diseased kidney. Am J Kidney Dis 29: 2‐26, 1997.
 169. Kohan DE. Biology of endothelin receptors in the collecting duct. Kidney Int 76: 481‐486, 2009.
 170. Kohan DE. Endothelin, hypertension and chronic kidney disease: New insights. Curr Opin Nephrol Hypertens 19: 134‐139, 2010.
 171. Kokko JP. Proximal tubule potential difference. Dependence on glucose on glucose, HCO 3, and amino acids. J Clin Invest 52: 1362‐1367, 1973.
 172. Kokko JP, Rector FC. Flow dependence of transtubular potential difference in isolated perfused segments of rabbit proximal convoluted tubule. J Clin Invest 50: 2745‐2750, 1971.
 173. Kone BC, Wenzhang Z, Zhiyuan Y. New mechanisms for transcriptional repression of ENaC And iNOS. Trans Am Clin Climatol Assoc 118: 45‐56, 2007.
 174. Kovacs G, Peti‐Peterdi J, Rosivall L, Bell PD. Angiotensin II directly stimulates macula densa Na‐2Cl‐K cotransport via apical AT(1) receptors. Am J Physiol Renal Physiol 282: F301‐F306, 2002.
 175. Krapf R, Berry CA, Verkman AS. Estimation of intracellular chloride activity in isolated perfused rabbit proximal convoluted tubules using a fluorescent indicator. Biophys J 53: 955‐962, 1988.
 176. Kunkel MT, Ni Q, Tsien RY, Zhang J, Newton AC. Spatio‐temporal dynamics of protein kinase B/Akt signaling revealed by a genetically encoded fluorescent reporter. J Biol Chem 280: 5581‐5587, 2005.
 177. Kuntziger H, Antonetti A, Couette S, Coureau C, Amiel C. Ultramicro (nanoliter range) determination of calcium concentration (10‐3 M) by atomic absorption. Anal Biochem 60: 449‐454, 1974.
 178. Lachheb S, Cluzeaud F, Bens M, Genete M, Hibino H, Lourdel S, Kurachi Y, Vandewalle A, Teulon J, Paulais M. Kir4.1/Kir5.1 channel forms the major K+ channel in the basolateral membrane of mouse renal collecting duct principal cells. Am J Physiol Renal Physiol 294: F1398‐F1407, 2008.
 179. Lameire NH, Lifschitz MD, Stein JH. Heterogeneity of nephron function. Annu Rev Physiol 39: 159‐184, 1977.
 180. Lechene C, Morel F. Microinjections of tagged sodium and insulin into the renal capillaries of the hamster. I. Permeability of cortical tubular segments to sodium. Nephron 2: 207‐218, 1965.
 181. Leviel F, Hubner CA, Houillier P, Morla L, El MS, Brideau G, Hatim H, Parker MD, Kurth I, Kougioumtzes A, Sinning A, Pech V, Riemondy KA, Miller RL, Hummler E, Shull GE, Aronson PS, Doucet A, Wall SM, Chambrey R, Eladari D. The Na+‐dependent chloride‐bicarbonate exchanger SLC4A8 mediates an electroneutral Na+ reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 120: 1627‐1635, 2010.
 182. Leyssac PP, Baumbach L. An oscillating intratubular pressure response to alterations in Henle loop flow in the rat kidney. Acta Physiol Scand 117: 415‐419, 1983.
 183. Li D, Wang Z, Sun P, Jin Y, Lin DH, Hebert SC, Giebisch G, Wang WH. Inhibition of MAPK stimulates the Ca2+‐dependent big‐conductance K channels in cortical collecting duct. Proc Natl Acad Sci U S A 103: 19569‐19574, 2006.
 184. Lifton RP. Genetic determinants of human hypertension. [Review] [56 refs]. Proc Natl Acad Sci U S A 92: 8545‐8551, 1995.
 185. Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hypertension. Cell 104: 545‐556, 2001.
 186. Lin DH, Sterling H, Wang WH. The protein tyrosine kinase‐dependent pathway mediates the effect of K intake on renal K secretion. Physiology (Bethesda) 20: 140‐146, 2005.
 187. Lingueglia E, Voilley N, Waldmann R, Lazdunski M, Barbry P. Expression cloning of an epithelial amiloride‐sensitive Na+ channel. A new channel type with homologies to Caenorhabditis elegans degenerins. FEBS Lett 318: 95‐99, 1993.
 188. Lissandron V, Terrin A, Collini M, D'alfonso L, Chirico G, Pantano S, Zaccolo M. Improvement of a FRET‐based indicator for cAMP by linker design and stabilization of donor‐acceptor interaction. J Mol Biol 354: 546‐555, 2005.
 189. Liu R, Carretero OA, Ren Y, Garvin JL. Increased intracellular pH at the macula densa activates nNOS during tubuloglomerular feedback. Kidney Int 67: 1837‐1843, 2005.
 190. Liu W, Morimoto T, Woda C, Kleyman TR, Satlin LM. Ca2+ dependence of flow‐stimulated K secretion in the mammalian cortical collecting duct. Am J Physiol Renal Physiol 293: F227‐F235, 2007.
 191. Liu W, Wei Y, Sun P, Wang WH, Kleyman TR, Satlin LM. Mechanoregulation of BK channel activity in the mammalian cortical collecting duct: Role of protein kinases A and C. Am J Physiol Renal Physiol 297: F904‐F915, 2009.
 192. Loffing J, Vallon V, Loffing‐Cueni D, Aregger F, Richter K, Pietri L, Bloch‐Faure M, Hoenderop JG, Shull GE, Meneton P, Kaissling B. Altered renal distal tubule structure and renal Na(+) and Ca(2+) handling in a mouse model for Gitelman's syndrome. J Am Soc Nephrol 15: 2276‐2288, 2004.
 193. Lohse MJ, Bunemann M, Hoffmann C, Vilardaga JP, Nikolaev VO. Monitoring receptor signaling by intramolecular FRET. Curr Opin Pharmacol 7: 547‐553, 2007.
 194. Lorenz JN. Considerations for the evaluation of renal function in genetically engineered mice. Curr Opin Nephrol Hypertens 10: 65‐69, 2001.
 195. Lorenz JN. A practical guide to evaluating cardiovascular, renal, and pulmonary function in mice. Am J Physiol Regul Integr Comp Physiol 282: R1565‐R1582, 2002.
 196. Lorenz JN, Baird NR, Judd LM, Noonan WT, Andringa A, Doetschman T, Manning PA, Liu LH, Miller ML, Shull GE. Impaired renal NaCl absorption in mice lacking the ROMK potassium channel, a model for type II Bartter's syndrome. J Biol Chem 277: 37871‐37880, 2002.
 197. Lorenz JN, Gruenstein E. A simple, nonradioactive method for evaluating single‐nephron filtration rate using FITC‐inulin. Am J Physiol 276: F172‐F177, 1999.
 198. Lourdel S, Paulais M, Marvao P, Nissant A, Teulon J. A chloride channel at the basolateral membrane of the distal‐convoluted tubule: A candidate ClC‐K channel. J Gen Physiol 121: 287‐300, 2003.
 199. Lowitz KH, Stumpe KO, Ochwadt B. Micropuncturestudy of the action of angiotensin II on tubular sodium and water reabsorption in the rat. Nephron 6: 173‐187, 1969.
 200. Lu M, Macgregor GG, Wang W, Giebisch G. Extracellular ATP inhibits the small‐conductance K channel on the apical membrane of the cortical collecting duct from mouse kidney. J Gen Physiol 116: 299‐310, 2000.
 201. Lu M, Wang T, Yan Q, Wang W, Giebisch G, Hebert SC. ROMK is required for expression of the 70‐pS K channel in the thick ascending limb. Am J Physiol Renal Physiol 286: F490‐F495, 2004.
 202. 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.
 203. Lu M, Wang W. Two types of K(+) channels are present in the apical membrane of the thick ascending limb of the mouse kidney. Kidney Blood Press Res 23: 75‐82, 2000.
 204. Ludwig C. De viribus physicis secretionem urinae adjuvantibus. Thesis, Marburg: N.G. Elwert, 1842.
 205. Ludwig CFW. Beitraege zur Lehre vom Mechanismus der Harnsekretion. Marburg: N.G. Elwert, 1843.
 206. Malnic G. Combined in vivo and in vitro approaches to analysis of renal tubule function. Exp Nephrol 6: 454‐461, 1998.
 207. Malnic G, Vieira FL. The antimony microelectrode in kidney micropuncture. Yale J Biol Med 45: 356‐367, 1972.
 208. Marsh D, Frasier C, Decter J. Measurement of urea concentrations in nanoliter specimens of renal tubular fluid and capillary blood. Anal Biochem 11: 73‐80, 1965.
 209. Meneton P, Ichikawa I, Inagami T, Schnermann J. Renal physiology of the mouse. Am J Physiol Renal Physiol 278: F339‐F351, 2000.
 210. Mick VE, Itani OA, Loftus RW, Husted RF, Schmidt TJ, Thomas CP. The alpha‐subunit of the epithelial sodium channel is an aldosterone‐induced transcript in mammalian collecting ducts, and this transcriptional response is mediated via distinct cis‐elements in the 5’‐flanking region of the gene. Mol Endocrinol 15: 575‐588, 2001.
 211. Misler S, Gillis K, Tabcharani J. Modulation of gating of a metabolically regulated, ATP‐dependent K+ channel by intracellular pH in B cells of the pancreatic islet. J Membr Biol 109: 135‐143, 1989.
 212. Molitoris BA, Dagher PC, Sandoval RM, Campos SB, Ashush H, Fridman E, Brafman A, Faerman A, Atkinson SJ, Thompson JD, Kalinski H, Skaliter R, Erlich S, Feinstein E. siRNA targeted to p53 attenuates ischemic and cisplatin‐induced acute kidney injury. J Am Soc Nephrol 20: 1754‐1764, 2009.
 213. Morel F, Mylle M, Gottschalk CW. Tracer microinjection studies of effect of adh on renal tubular diffusion of water. Am J Physiol 209: 179‐187, 1965.
 214. Morel F, Roinel N, Le GC. Electron probe analysis of tubular fluid composition. Nephron 6: 350‐364, 1969.
 215. Moullier P, Friedlander G, Calise D, Ronco P, Perricaudet M, Ferry N. Adenoviral‐mediated gene transfer to renal tubular cells in vivo. Kidney Int 45: 1220‐1225, 1994.
 216. Muto S. Potassium transport in the mammalian collecting duct. Physiol Rev 81: 85‐116, 2001.
 217. Muto S, Asano Y, Wang W, Seldin D, Giebisch G. Activity of the basolateral K+ channels is coupled to the Na+‐K+‐ATPase in the cortical collecting duct. Am J Physiol Renal Physiol 285: F945‐F954, 2003.
 218. Nagami GT, Kurokawa K. Regulation of ammonia production by mouse proximal tubules perfused in vitro. Effect of luminal perfusion. J Clin Invest 75: 844‐849, 1985.
 219. Nakajima K, Clapp JR, Robinson RR. Limitationsof the shrinking‐drop micropuncture technique. Am J Physiol 219: 345‐357, 1970.
 220. Nakayama I, Kawahara Y, Tsuda T, Koide M, Yokoyama M. Cyclic AMP elevating agents synergize with inflammatory cytokines to induce an inducible type of nitric oxide synthase in cultured vascular smooth muscle cells. Ann NY Acad Sci U S A 748: 586‐589, 1995.
 221. Naray‐Fejes‐Toth A, Canessa C, Cleaveland ES, Aldrich G, Fejes‐Toth G. sgk is an aldosterone‐induced kinase in the renal collecting duct. J Biol Chem 274: 16973‐16978, 1999.
 222. Neher E, Sakmann B. Single‐channel currents recorded from membrane of denervated frog muscle fibers. Nature 260(5554): 799‐802, 1976.
 223. Nijenhuis T, Vallon V, van der Kemp AW, Loffing J, Hoenderop JG, Bindels RJ. Enhanced passive Ca2+ reabsorption and reduced Mg2+ channel abundance explains thiazide‐induced hypocalciuria and hypomagnesemia. J Clin Invest 115: 1651‐1658, 2005.
 224. Nikolaev VO, Gambaryan S, Lohse MJ. Fluorescent sensors for rapid monitoring of intracellular cGMP. Nat Methods 3: 23‐25, 2006.
 225. Nissant A, Paulais M, Lachheb S, Lourdel S, Teulon J. Similar chloride channels in the connecting tubule and cortical collecting duct of the mouse kidney. Am J Physiol Renal Physiol 290: F1421‐F1429, 2006.
 226. Noonan WT, Lorenz JN. Clearance studies in genetically altered mice. Methods Mol Med 86: 315‐327, 2003.
 227. O'Neil RG, Boulpaep EL. Ionic conductives properties and electrophysiology of the rabbit cortical collecting tubule. Am J Physiol 243: 81‐95, 1982.
 228. O'Neil RG, Hayhurst AR. Functional differentiation of cell types of cortical collecting duct. Am J Physiol 248: 449‐453, 1985.
 229. Ortiz PA, Garvin JL. Interaction of O(2)[‐] and NO in the thick ascending limb. Hypertension 39: 591‐596, 2002.
 230. Ortiz PA, Hong NJ, Garvin JL. NO decreases thick ascending limb chloride absorption by reducing Na(+)‐K(+)‐2Cl[‐] cotransporter activity. Am J Physiol Renal Physiol 281: F819‐F825, 2001.
 231. Ortiz PA, Hong NJ, Garvin JL. Luminal flow induces eNOS activation and translocation in the rat thick ascending limb. Am J Physiol Renal Physiol 287: F274‐F280, 2004a.
 232. Ortiz PA, Hong NJ, Garvin JL. Luminal flow induces eNOS activation and translocation in the rat thick ascending limb. II. Role of PI3‐kinase and Hsp90. Am J Physiol Renal Physiol 287: F281‐F288, 2004b.
 233. Ortiz PA, Hong NJ, Plato CF, Varela M, Garvin JL. An in vivo method for adenovirus‐mediated transduction of thick ascending limbs. Kidney Int 63: 1141‐1149, 2003.
 234. Ortiz PA, Hong NJ, Wang D, Garvin JL. Gene transfer of eNOS to the thick ascending limb of eNOS‐KO mice restores the effects of L‐arginine on NaCl absorption. Hypertension 42: 674‐679, 2003.
 235. Pácha 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.
 236. Pácha J, Frindt G, Sackin H, Palmer LG. Apical maxi K channels in intercalated cells of CCT. Am J Physiol 261: F696‐F705, 1991.
 237. Palmer LG, Choe H, Frindt G. Is the secretory K channel in the rat CCT ROMK? Am J Physiol 273: F404‐F410, 1997.
 238. Palmer LG, Frindt G. Amiloride‐sensitive Na channels from the apical membrane of the rat cortical collecting tubule. Proc Natl Acad Sci U S A 83: 2767‐2770, 1986.
 239. Palmer LG, Frindt G. Effects of cell Ca and pH on Na channels from rat cortical collecting tubule. Am J Physiol 253: 333‐339, 1987.
 240. Palmer LG, Frindt G. Gating of Na channels in the rat cortical collecting tubule: Effects of voltage and membrane stretch. J Gen Physiol 107: 35‐45, 1996.
 241. Palmer LG, Frindt G. Regulation of apical K channels in rat cortical collecting tubule during changes in dietary K intake. Am J Physiol 277: F805‐F812, 1999.
 242. Palmer LG, Frindt G. Aldosterone and potassium secretion by the cortical collecting duct. Kidney Int 57: 1324‐1328, 2000.
 243. Palmer LG, Frindt G. Cl‐ channels of the distal nephron. Am J Physiol Renal Physiol 291: F1157‐F1168, 2006.
 244. Palmer LG, Frindt G. High‐conductance K channels in intercalated cells of the rat distal nephron. Am J Physiol Renal Physiol 292: F966‐F973, 2007.
 245. Panico C, Luo Z, Damiano S, Artigiano F, Gill P, Welch WJ. Renal proximal tubular reabsorption is reduced in adult spontaneously hypertensive rats: Roles of superoxide and Na+/H +exchanger 3. Hypertension 54: 1291‐1297, 2009.
 246. Parent L, Cardinal J, Sauve R. Single‐channel analysis of a K channel at basolateral membrane of rabbit proximal convoluted tubule. Am J Physiol 254: F105‐F113, 1988.
 247. Paulais M, Teulon J. cAMP‐activated chloride channel in the basolateral membrane of the thick ascending limb of the mouse kidney. J Membr Biol 113: 253‐260, 1990.
 248. Pech V, Zheng W, Pham TD, Verlander JW, Wall SM. Angiotensin II activates H+‐ATPase in type A intercalated cells. J Am Soc Nephrol 19: 84‐91, 2008.
 249. Peti‐Peterdi J. Multiphoton imaging of renal tissues in vitro. Am J Physiol Renal Physiol 288: F1079‐F1083, 2005.
 250. Peti‐Peterdi J, Bebok Z, Lapointe JY, Bell PD. Novel regulation of cell [Na(+)] in macula densa cells: Apical Na(+) recycling by H‐K‐ATPase. Am J Physiol Renal Physiol 282: F324‐F329, 2002.
 251. Peti‐Peterdi J, Morishima S, Bell PD, Okada Y. Two‐photon excitation fluorescence imaging of the living juxtaglomerular apparatus. Am J Physiol Renal Physiol 283: F197‐F201, 2002.
 252. Pluznick J, Wei P, Carmines P, Sansom SC. Renal fluid and electrolyte handling in BKCa‐beta1 ‐/‐ mice. Am J Physiol 284: F1274‐F1279, 2003.
 253. Pochynyuk O, Bugaj V, Rieg T, Insel PA, Mironova E, Vallon V, Stockand JD. Paracrine regulation of the epithelial Na+ channel in the mammalian collecting duct by purinergic P2Y2 receptor tone. J Biol Chem 283: 36599‐36607, 2008.
 254. Pochynyuk O, Kucher V, Boiko N, Mironova E, Staruschenko A, Karpushev AV, Tong Q, Hendron E, Stockand J. Intrinsic voltage dependence of the epithelial Na+ channel is masked by a conserved transmembrane domain tryptophan. J Biol Chem 284: 25512‐25521, 2009.
 255. Pochynyuk O, Rieg T, Bugaj V, Schroth J, Fridman A, Boss GR, Insel PA, Stockand JD, Vallon V. Dietary Na+ inhibits the open probability of the epithelial sodium channel in the kidney by enhancing apical P2Y2‐receptor tone. FASEB J 24: 2056‐2065, 2010.
 256. Preisig PA, Ives HE, Cragoe EJ Jr., Alpern RJ, Rector FC Jr. Role of the Na+/H+ antiporter in rat proximal tubule bicarbonate absorption. J Clin Invest 80: 970‐978, 1987.
 257. Qi Z, Whitt I, Mehta A, Jin J, Zhao M, Harris RC, Fogo AB, Breyer MD. Serial determination of glomerular filtration rate in conscious mice using FITC‐inulin clearance. Am J Physiol Renal Physiol 286: F590‐F596, 2004.
 258. Ramsay MA, Brown RHJ. Simplified apparatus and procedure for freezing point determinations upon small volumes of fluid. J Scient Instruments 32: 372, 1955.
 259. Ramsey J, Brown R, Croghan P. Electrometric titration of chloride in small volume. J Exp Biol 32: 822‐829, 1955.
 260. Rao S, Verkman AS. Analysis of organ physiology in transgenic mice. Am J Physiol Cell Physiol 279: C1‐C18, 2000.
 261. Reif MC, Troutman SL, Schafer JA. Sustained response to vasopressin in isolated rat cortical collecting tubule. Kidney Int 26: 725‐732, 1984.
 262. Reif MC, Troutman SL, Schafer JA. Sodium transport by rat cortical collecting tubule. J Clin Invest 77: 1291‐1298, 1986.
 263. Richards AN, Walker AM. Methods of collecting fluid from known regions of the renal tubules of Amphibia and of perfusing the lumen of a single tubule. Am J Physiol 118: 111‐120, 1936.
 264. Rieg T, Bundey RA, Chen Y, Deschenes G, Junger W, Insel PA, Vallon V. Mice lacking P2Y2 receptors have salt‐resistant hypertension and facilitated renal Na+ and water reabsorption. FASEB J 21: 3717‐3726, 2007.
 265. Rieg T, Tang T, Murray F, Schroth J, Insel PA, Fenton RA, Hammond HK, Vallon V. Adenylyl cyclase 6 determines cAMP formation and aquaporin 2 phosphorylation and trafficking in renal inner medullary collecting duct. J Am Soc Nephrol 21: 2059‐2062, 2010.
 266. Rieg T, Vallon V. ATP and adenosine in the local regulation of water transport and homeostasis by the kidney. Am J Physiol Regul Integr Comp Physiol 296: R419‐R427, 2009.
 267. 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.
 268. Roch‐Ramel F. An enzymic and fluorophotometric method for estimating urea concentrations in nanoliter specimens. Anal Biochem 21: 372‐381, 1967.
 269. Roman RJ, Bonventre JV, Lechene CP. Fluorometric assay for urea in urine, plasma, and tubular fluid. Anal Biochem 98: 136‐141, 1979.
 270. Rossier BC, Pradervand S, Schild L, Hummler E. Epithelial sodium channel and the control of sodium balance: Interaction between genetic and environmental factors. Annu Rev Physiol 64: 877‐897, 2002.
 271. Rubera I, Loffing J, Palmer LG, Frindt G, Fowler‐Jaeger N, Sauter D, Carroll T, McMahon A, Hummler E, Rossier BC. Collecting duct‐specific gene inactivation of alphaENaC in the mouse kidney does not impair sodium and potassium balance. J Clin Invest 112: 554‐565, 2003.
 272. Sackin H, Boulpaep EL. Isolated perfused salamander proximal tubule: Methods, electrophysiology, and transport. Am J Physiol 241: F39‐F52, 1981.
 273. Sackin H, Boulpaep EL. Rheogenic transport in the renal proximal tubule. J Gen Physiol 82: 819‐851, 1983.
 274. Sansom SC, La B‐Q, Carosi SL. Double‐barreled chloride channels of collecting duct basolateral membrane. Am J Physiol 259: F46‐F52, 1990.
 275. Satlin LM, Palmer LG. Apical Na+ conductance in maturing rabbit principal cell. Am J Physiol 270: F391‐F397, 1996.
 276. Schafer JA, Andreoli TE. Rheogenic and passive Na+ absorption by the proximal nephron. Annu Rev Physiol 41: 211‐227, 1979.
 277. Schafer JA, Patlak CS, Troutman SL, Andreoli TE. Volume absorption in the pars recta. II. Hydraulic conductivity coefficient. Am J Physiol 234: F340‐F348, 1978.
 278. Schafer JA, Troutman SL. cAMP mediates the increase in apical membrane Na+ conductance produced in rat CCD by vasopressin. Am J Physiol 259: F823‐F831, 1990.
 279. Schafer JA, Troutman SL, Andreoli TE. Volume reabsorption, transepithelial potential differences, and ionic permeability properties in mammalian superficial proximal straight tubules. J Gen Physiol 64: 582‐607, 1974.
 280. Schafer JA, Troutman SL, Watkins ML, Andreoli TE. Volume absorption in the pars recta. I. “Simple” active Na+ transport. Am J Physiol 234: F332‐F339, 1978.
 281. Schild L, Canessa CM, Shimkets RA, Gautschi I, Lifton RP, Rossier BC. A mutation in the epithelial sodium channel causing Liddle disease increases channel activity in the Xenopus laevis oocyte expression system. Proc Natl Acad Sci U S A 92: 5699‐5703, 1995.
 282. Schild L, Lu Y, Gautschi I, Schneeberger E, Lifton RP, Rossier BC. Identification of a PY motif in the epithelial Na channel subunits as a target sequence for mutations causing channel activation found in Liddle syndrome. EMBO J 15: 2381‐2387, 1996.
 283. Schlanger LE, Kleyman TR, Ling BN. K+‐sparing diuretic actions of trimethoprim: Inhibition of Na+ channels in A6 distal nephron cells. Kidney Int 45: 1070‐1076, 1994.
 284. Schlatter E. Regulation of ion channels in the cortical collecting duct. Renal Physiol Biochem 16: 21‐36, 1993.
 285. Schlatter E, Bleich M, Hirsch J, Markstahler U, Frobe U, Greger R. Cation specificity and pharmacological properties of the Ca(2+)‐dependent K+ channel of rat cortical collecting ducts. Pflugers Arch 422: 481‐491, 1993.
 286. Schlatter E, Fröbe U, Greger R. Ion conductances of isolated cortical collecting duct cells. Pflugers Arch 421: 381‐387, 1992.
 287. Schlatter E, Haxelmans S, Hirsch J, Leipziger J. pH dependence of K+ conductances of rat cortical collecting duct principal cells. Pflugers Arch 428: 631‐640, 1994.
 288. Schlatter E, Schafer JA. Electrophysiological studies in principal cells of rat cortical collecting tubules. ADH increases the apical membrane Na+‐conductance. Pflugers Arch 409: 81‐92, 1987.
 289. Schneider MP, Ge Y, Pollock DM, Pollock JS, Kohan DE. Collecting duct‐derived endothelin regulates arterial pressure and Na excretion via nitric oxide. Hypertension 51: 1605‐1610, 2008.
 290. 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.
 291. Shalmi M, Kibble JD, Day JP, Christensen P, Atherton JC. Improved analysis of picomole quantities of lithium, sodium, and potassium in biological fluids. Am J Physiol 267: F695‐F701, 1994.
 292. Sheridan E, Rumrich G, Ullrich KJ. Reabsorption of dicarboxylic acids from the proximal convolution of rat kidney. Pflugers Arch 399: 18‐28, 1983.
 293. Shi LB, Fushimi K, Verkman AS. Solvent drag measurement of transcellular and basolateral membrane NaCl reflection coefficient in kidney proximal tubule. J Gen Physiol 98: 379‐398, 1991.
 294. Shi PP, Cao XR, Sweezer EM, Kinney TS, Williams NR, Husted RF, Nair R, Weiss RM, Williamson RA, Sigmund CD, Snyder PM, Staub O, Stokes JB, Yang B. Salt‐sensitive hypertension and cardiac hypertrophy in mice deficient in the ubiquitin ligase Nedd4‐2. Am J Physiol Renal Physiol 295: F462‐F470, 2008.
 295. Shipp J, Hanenson I, Windhager EE, Schatzmann H, Whittembury G, Yoshimura H, Solomon A. Single proximal tubules of the Necturus kidney; methods for micropuncture and microperfusion. Am J Physiol 195: 563‐569, 1958.
 296. Shirley DG, Bailey MA, Unwin RJ. In vivo stimulation of apical P2 receptors in collecting ducts: Evidence for inhibition of sodium reabsorption. Am J Physiol Renal Physiol 288: F1243‐F1248, 2005.
 297. Sigworth FJ, Neher E. Single Na+ channel currents observed in cultured rat muscle cells. Nature 287: 447‐449, 1980.
 298. Silbernagl S, Ganapathy V, Leibach FH. H+ gradient‐driven dipeptide reabsorption in proximal tubule of rat kidney. Studies in vivo and in vitro. Am J Physiol 253: F448‐F457, 1987.
 299. Silva GB, Garvin JL. TRPV4 mediates hypotonicity‐induced ATP release by the thick ascending limb. Am J Physiol Renal Physiol 295: F1090‐F1095, 2008.
 300. Silver RB, Frindt G, Windhager EE, Palmer LG. Feedback regulation of Na channels in rat CCT. I. Effects of inhibition of Na pump. Am J Physiol 264: F557‐F564, 1993.
 301. Simon DB, Karet FE, Hamdan JM, DiPietro A, Sanjad SA, Lifton RP. Bartter's syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na‐K‐2Cl cotransporter NKCC2. Nat Genet 13: 183‐188, 1996.
 302. Simon DB, Karet FE, Rodriguez‐Soriano J, Hamdan JH, DiPietro A, Trachtman H, Sanjad SA, Lifton RP. Genetic heterogeneity of Bartter's syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet 14: 152‐156, 1996.
 303. Sipos A, Vargas SL, Toma I, Hanner F, Willecke K, Peti‐Peterdi J. Connexin 30 deficiency impairs renal tubular ATP release and pressure natriuresis. J Am Soc Nephrol 20: 1724‐1732, 2009.
 304. Snyder PM. Regulation of epithelial Na+ channel trafficking. Endocrinology 146: 5079‐5085, 2005.
 305. Snyder PM. Down‐regulating destruction: Phosphorylation regulates the E3 ubiquitin ligase Nedd4‐2. Sci Signal 2: e41, 2009.
 306. Snyder PM, Olson DR, Kabra R, Zhou R, Steines JC. cAMP and serum and glucocorticoid‐inducible kinase (SGK) regulate the epithelial Na(+) channel through convergent phosphorylation of Nedd4‐2. J Biol Chem 279: 45753‐45758, 2004.
 307. Snyder PM, Price MP, McDonald FJ, Adams CM, Volk KA, Zeiher BG, Stokes JB, Welsh MJ. Mechanism by which Liddle's syndrome mutations increase activity of a human epithelial Na +channel. Cell 83: 969‐978, 1995.
 308. Star RA. Quantitation of total carbon dioxide in nanoliter samples by flow‐through fluorometry. Am J Physiol 258: F429‐F432, 1990.
 309. Star RA, Burg MB, Knepper MA. Bicarbonate secretion and chloride absorption by the rabbit cortical collecting ducts. J Clin Invest 76: 1123‐1130, 1985.
 310. 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.
 311. Sterling H, Lin DH, Chen YJ, Wei Y, Wang ZJ, Lai J, Wang WH. PKC expression is regulated by dietary K intake and mediates internalization of SK channels in the CCD. Am J Physiol Renal Physiol 286: F1072‐F1078, 2004.
 312. Stockand JD. New ideas about aldosterone signaling in epithelia. Am J Physiol Renal Physiol 282: F559‐F576, 2002.
 313. Stockand JD. Vasopressin regulation of renal sodium excretion. Kidney Int 78: 849‐856, 2010.
 314. Stocking CJ, Slater JM, Unwin R, Walter S, Folkerd E. An automated technique for the simultaneous determination of cations in nanoliter volumes. Kidney Int 56: 338‐343, 1999.
 315. Stokes JB, Ingram MJ, Williams AD, Ingram D. Heterogeneity of the rabbit collecting tubule: Localization of mineralocorticoid hormone action to the cortical portion. Kidney Int 20: 340‐347, 1981.
 316. Stoner LC, Morley GE. Effect of basolateral or apical hyposmolarity on apical maxi K channels of everted rat collecting tubule. Am J Physiol 268: F569‐F580, 1995.
 317. Strait KA, Stricklett PK, Kohan JL, Miller MB, Kohan DE. Calcium regulation of endothelin‐1 synthesis in rat inner medullary collecting duct. Am J Physiol Renal Physiol 293: F601‐F606, 2007.
 318. Strieter J, Stephenson JL, Giebisch G, Weinstein AM. A mathematical model of the rabbit cortical collecting tubule. Am J Physiol 263: F1063‐F1075, 1992.
 319. Sun Z, Bello‐Roufai M, Wang X. RNAi inhibition of mineralocorticoid receptors prevents the development of cold‐induced hypertension. Am J Physiol Heart Circ Physiol 294: H1880‐H1887, 2008.
 320. Tanner GA, Sandoval RM, Molitoris BA, Bamburg JR, Ashworth SL. Micropuncture gene delivery and intravital two‐photon visualization of protein expression in rat kidney. Am J Physiol Renal Physiol 289: F638‐F643, 2005.
 321. Terada Y, Knepper MA. Continuous‐flow quantitation of Na+ and K+ in nanoliter samples using chromogenic macrocyclic ionophores. Am J Physiol 257: F893‐F898, 1989.
 322. Thiemann A, Grunder S, Pusch M, Jentsch TJ. A chloride channel widely expressed in epithelial and non‐epithelial cells. Nature 346: 57‐60, 1992.
 323. Thomas RC. Construction and properties of recessed‐tip microelectrodes for sodium and chloride ions and pH. In: Kessler R, Clark LC, Lubbers DW, Silver IA, Simon W, editors. Ion and Enzyme Electrodes in Biology and Medicine. Baltimore: University Park Press, 1976, pp. 141‐148.
 324. Thomson SC, Blantz RC. Biophysical basis of glomerular filtration. In: Alpern RJ, Hebert SC, editors. Seldin and Giebisch's The Kidney. Waltham, Massachusetts: Academic Press, 2008, pp. 565‐587.
 325. Thomson SC, Deng A, Bao D, Satriano J, Blantz RC, Vallon V. Ornithine decarboxylase, kidney size, and the tubular hypothesis of glomerular hyperfiltration in experimental diabetes. J Clin Invest 107: 217‐224, 2001.
 326. Thomson SC, Deng A, Wead L, Richter K, Blantz RC, Vallon V. An unexpected role for angiotensin II in the link between dietary salt and proximal reabsorption. J Clin Invest 116: 1110‐1116, 2006.
 327. Tong Q, Gamper N, Medina JL, Shapiro MS, Stockand JD. Direct activation of the epithelial Na(+) channel by phosphatidylinositol 3,4,5‐trisphosphate and phosphatidylinositol 3,4‐bisphosphate produced by phosphoinositide 3‐OH kinase. J Biol Chem 279: 22654‐22663, 2004.
 328. Vallon V. In vivo studies of the genetically modified mouse kidney. Nephron Physiol 94: 1‐5, 2003.
 329. Vallon V. Micropuncturing the nephron. Pflugers Arch 458: 189‐201, 2009.
 330. Vallon V, Grahammer F, Richter K, Bleich M, Lang F, Barhanin J, Volkl H, Warth R. Role of KCNE1‐dependent K+ fluxes in mouse proximal tubule. J Am Soc Nephrol 12: 2003‐2011, 2001.
 331. Vallon V, Grahammer F, Volkl H, Sandu CD, Richter K, Rexhepaj R, Gerlach U, Rong Q, Pfeifer K, Lang F. KCNQ1‐dependent transport in renal and gastrointestinal epithelia. Proc Natl Acad Sci U S A 102: 17864‐17869, 2005.
 332. Vallon V, Huang DY, Deng A, Richter K, Blantz RC, Thomson S. Salt‐sensitivity of proximal reabsorption alters macula densa salt and explains the paradoxical effect of dietary salt on glomerular filtration rate in diabetes mellitus. J Am Soc Nephrol 13: 1865‐1871, 2002.
 333. Vallon V, Hummler E, Rieg T, Pochynyuk O, Bugaj V, Schroth J, Dechenes G, Rossier B, Cunard R, Stockand J. Thiazolidinedione‐induced fluid retention is independent of collecting duct alphaENaC activity. J Am Soc Nephrol 20: 721‐729, 2009.
 334. Vallon V, Osswald H, Blantz RC, Thomson S. Potential role of luminal potassium in tubuloglomerular feedback. J Am Soc Nephrol 8: 1831‐1837, 1997.
 335. 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.
 336. Vallon V, Richter K, Blantz RC, Thomson S, Osswald H. Glomerular hyperfiltration in experimental diabetes mellitus: Potential role of tubular reabsorption. J Am Soc Nephrol 10: 2569‐2576, 1999.
 337. Vallon V, Richter K, Heyne N, Osswald H. Effect of intratubular application of angiotensin 1‐7 on nephron function. Kidney Blood Press Res 20: 233‐239, 1997.
 338. Vallon V, Richter K, Huang DY, Rieg T, Schnermann J. Functional consequences at the single‐nephron level of the lack of adenosine A1 receptors and tubuloglomerular feedback in mice. Pflugers Arch 448: 214‐221, 2004.
 339. Vallon V, Schnermann J. Tubuloglomerular feedback. Methods Mol Med 86: 429‐441, 2003.
 340. 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‐F379, 2000.
 341. Vallon V, Traynor T, Barajas L, Huang YG, Briggs JP, Schnermann J. Feedback control of glomerular vascular tone in neuronal nitric oxide synthase knockout mice. J Am Soc Nephrol 12: 1599‐1606, 2001.
 342. Vallon V, Verkman AS, Schnermann J. Luminal hypotonicity in proximal tubules of aquaporin‐1‐knockout mice. Am J Physiol Renal Physiol 278: F1030‐F1033, 2000.
 343. van de Water FM, Boerman OC, Wouterse AC, Peters JG, Russel FG, Masereeuw R. Intravenously administered short interfering RNA accumulates in the kidney and selectively suppresses gene function in renal proximal tubules. Drug Metab Dispos 34: 1393‐1397, 2006.
 344. Vargas‐Poussou R, Feldmann D, Vollmer M, Konrad M, Kelly L, van den Heuvel LP, Tebourbi L, Brandis M, Karolyi L, Hebert SC, Lemmink HH, Deschenes G, Hildebrandt F, Seyberth HW, Guay‐Woodford LM, Knoers NV, Antignac C. Novel molecular variants of the Na‐K‐2Cl cotransporter gene are responsible for antenatal Bartter syndrome. Am J Hum Genet 62: 1332‐1340, 1998.
 345. Vekaria RM, Unwin RJ, Shirley DG. Intraluminal ATP concentrations in rat renal tubules. J Am Soc Nephrol 17: 1841‐1847, 2006.
 346. Velazquez H, Wright FS. Renal micropuncture techniques. Comp Physiol 249‐269, 2011.
 347. Verrey F. Early aldosterone effects. Exp Nephrol 6: 294‐301, 1998.
 348. Verrey F. Early aldosterone action: Toward filling the gap between transcription and transport. Am J Physiol 277: F319‐F327, 1999.
 349. Violin JD, Zhang J, Tsien RY, Newton AC. A genetically encoded fluorescent reporter reveals oscillatory phosphorylation by protein kinase C. J Cell Biol 161: 899‐909, 2003.
 350. Vurek GG. Flow‐through nanocolorimeter for measurement of picomole amounts of magnesium and phosphate. Anal Lett 14: 261‐269, 1981a.
 351. Vurek GG. Calcium measurement: Picomole quantitation by continuous‐flow colorimetry. Anal Biochem 114: 288‐293, 1981b.
 352. Vurek GG, Bowman RL. Helium‐glow photometer for picomole analysis of alkali metals. Science 149: 448‐450, 1965.
 353. Vurek GG, Knepper MA. A colorimeter for measurement of picomole quantities of urea. Kidney Int 21: 656‐658, 1982.
 354. Vurek GG, Warnock DG, Corsey R. Measurement of picomole amounts of carbon dioxide by calorimetry. Anal Chem 47: 765‐767, 1975.
 355. Waldegger S, Jentsch TJ. Functional and structural analysis of ClC‐K chloride channels involved in renal disease. J Biol Chem 275: 24527‐24533, 2000.
 356. Walker AM, Bott PA, Oliver J, MacDowel MC. The collection and analysis of fluid from single nephrons of the mammalian kidney. Am J Physiol 134: 580‐595, 1941.
 357. Walker AM, Hudson CL. The reabsorption of glucose from the renal tubule in Amphibia and the action of phlorhizin upon it. Am J Physiol 118: 130‐143, 1936.
 358. Walker AM, Oliver J. Methods for the collection of fluid from single glomeruli and tubules of the mammalian kidney. Am J Physiol 134: 562‐579, 1941.
 359. Walker JL. Ion specific liquid ion exchanger microelectrodes. Anal Chem 43: 89A‐93A, 1971.
 360. Wang T, Yang CL, Abbiati T, Schultheis PJ, Shull GE, Giebisch G, Aronson PS. Mechanism of proximal tubule bicarbonate absorption in NHE3 null mice. Am J Physiol 277: F298‐F302, 1999.
 361. Wang T, Yang CL, Abbiati T, Shull GE, Giebisch G, Aronson PS. Essential role of NHE3 in facilitating formate‐dependent NaCl absorption in the proximal tubule. Am J Physiol Renal Physiol 281: F288‐F292, 2001.
 362. Wang W, Hebert SC, Giebisch G. Renal K+ channels: Structure and function. Annu Rev Physiol 59: 413‐436, 1997.
 363. Wang W, Henderson RM, Geibel J, White S, Giebisch G. Mechanism of aldosterone‐induced increase of K+ conductance in early distal renal tubule cells of the frog. J Membr Biol 111: 277‐289, 1989.
 364. Wang W, Schwab A, Giebisch G. Regulation of small‐conductance K+ channel in apical membrane of rat cortical collecting tubule. Am J Physiol 259: F494‐F502, 1990.
 365. Wang W, White S, Geibel J, Giebisch G. A potassium channel in the apical membrane of rabbit thick ascending limb of Henle's loop. Am J Physiol 258: F244‐F253, 1990.
 366. Wang WH, Geibel J, Giebisch G. Mechanism of apical K+ channel modulation in principal renal tubule cells. Effect of inhibition of basolateral Na(+)‐K(+)‐ATPase. J Gen Physiol 101: 673‐694, 1993.
 367. Wang WH, Lu M, Hebert SC. Cytochrome P‐450 metabolites mediate extracellular Ca(2+)‐induced inhibition of apical K+ channels in the TAL. Am J Physiol 271: C103‐C111, 1996.
 368. Wang W‐H, McNicholas CM, Segal AS, Giebisch G. A novel approach allows identification of K channels in the lateral membrane of rat CCD. Am J Physiol 266: F813‐F822, 1994.
 369. Wang X, Skelley L, Cade R, Sun Z. AAV delivery of mineralocorticoid receptor shRNA prevents progression of cold‐induced hypertension and attenuates renal damage. Gene Ther 13: 1097‐1103, 2006.
 370. Wang Y, Sun Z. Klotho gene delivery prevents the progression of spontaneous hypertension and renal damage. Hypertension 54: 810‐817, 2009.
 371. Warnock DG, Burg MB. Urinary acidification: CO2 transport by the rabbit proximal straight tubule. Am J Physiol 232: F20‐F25, 1977.
 372. Wearn JT, Richards AN. Observations on the composition of glomerular urine, with particular reference to the problem of reabsorption in the renal tubules. Am J Physiol 71: 209‐227, 1924.
 373. Wei Y, Babilonia E, Sterling H, Jin Y, Wang WH. Mineralocorticoids decrease the activity of the apical small‐conductance K channel in the cortical collecting duct. Am J Physiol Renal Physiol 289: F1065‐F1071, 2005.
 374. Wei Y, Bloom P, Lin D, Gu R, Wang WH. Effect of dietary K intake on apical small‐conductance K channel in CCD: Role of protein tyrosine kinase. Am J Physiol Renal Physiol 281: F206‐F212, 2001.
 375. Wei Y, Wang WH. Role of the cytoskeleton in mediating effect of vasopressin and herbimycin A on secretory K channels in CCD. Am J Physiol Renal Physiol 282: F680‐F686, 2002.
 376. Wei Y, Zavilowitz B, Satlin LM, Wang WH. Angiotensin II inhibits the ROMK‐like small conductance K channel in renal cortical collecting duct during dietary potassium restriction. J Biol Chem 282: 6455‐6462, 2007.
 377. Weinmann EJ, Hardy RJ, Kashgarian M, Hayslett JP. Examination of the Gertz technique as applied to the proximal tubule of the rat kidney. Yale BioI Med 45: 289‐298, 1972.
 378. Wildman SS, Marks J, Turner CM, Yew‐Booth L, Peppiatt‐Wildman CM, King BF, Shirley DG, Wang W, Unwin RJ. Sodium‐dependent regulation of renal amiloride‐sensitive currents by apical P2 receptors. J Am Soc Nephrol 19: 731‐742, 2008.
 379. Windhager EE. Micropuncture and microperfusion, Chapter IV, in Renal Physiology. In: Gottschalk CW, Berliner RW, Giebisch GH, editors. People and Ideas. Am Physiol Soc. Bethesda: Maryland, pp. 101‐129, 1987.
 380. Windhager EE, Giebisch G. Micropuncture study of renal tubular transfer of sodium chloride in the rat. Am J Physiol 200: 581‐590, 1961.
 381. Wirz H. Der osmotische Druck des Blutes in der Nierenpapille. Helv Physiol Pharmacol Acta 11: 20‐29, 1953.
 382. Wirz H. Der osmotische Druck in den corticalen Tubuli der Rattenniere. Helv Physiol Pharmacol Acta 14: 353‐362, 1956.
 383. Wirz H, Hargitay B, Kuhn W. Lokalisation des Konzentrierungsprozesses in der Niere durch direkte Kryoskopie. Helv Physiol Pharmacol Acta 9: 196‐207, 1951.
 384. Woda C, Mulroney SE, Halaihel N, Sun L, Wilson PV, Levi M, Haramati A. Renal tubular sites of increased phosphate transport and NaPi‐2 expression in the juvenile rat. Am J Physiol Regul Integr Comp Physiol 280: R1524‐R1533, 2001.
 385. 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.
 386. Wright FS. Increasing magnitude of electrical potential along the renal distal tubule. Am J Physiol 220: 624‐638, 1971.
 387. Wright FS, Giebisch G. Glomerular filtration in single nephrons. Kidney Int 1: 201‐209, 1972.
 388. Wulff P, Vallon V, Huang DY, Volkl H, Yu F, Richter K, Jansen M, Schlunz M, Klingel K, Loffing J, Kauselmann G, Bosl MR, Lang F, Kuhl D. Impaired renal Na(+) retention in the sgk1‐knockout mouse. J Clin Invest 110: 1263‐1268, 2002.
 389. Wyckoff JA, Seely EW, Hurwitz S, Anderson BF, Lifton RP, Dluhy RG. Glucocorticoid‐remediable aldosteronism and pregnancy. Hypertension 35: 668‐672, 2000.
 390. Yang B, Zhao D, Qian L, Verkman AS. Mouse model of inducible nephrogenic diabetes insipidus produced by floxed aquaporin‐2 gene deletion. Am J Physiol Renal Physiol 291: F465‐F472, 2006.
 391. Yip KP. Coupling of vasopressin‐induced intracellular Ca2+ mobilization and apical exocytosis in perfused rat kidney collecting duct. J Physiol 538: 891‐899, 2002.
 392. Zhelyaskov VR, Liu S, Broderick MP. Analysis of nanoliter samples of electrolytes using a flow‐through microfluorometer. Kidney Int 57: 1764‐1769, 2000.
 393. Zhu G, Nicolson AG, Cowley BD, Rosen S, Sukhatme VP. In vivo adenovirus‐mediated gene transfer into normal and cystic rat kidneys. Gene Ther 3: 298‐304, 1996.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

James D. Stockand, Volker Vallon, Pablo Ortiz. In Vivo and Ex Vivo Analysis of Tubule Function. Compr Physiol 2012, 2: 2495-2525. doi: 10.1002/cphy.c100051