Comprehensive Physiology Wiley Online Library

Na+‐K+‐2Cl− Cotransporter (NKCC) Physiological Function in Nonpolarized Cells and Transporting Epithelia

Full Article on Wiley Online Library



ABSTRACT

Two genes encode the Na+‐K+‐2Cl cotransporters, NKCC1 and NKCC2, that mediate the tightly coupled movement of 1Na+, 1K+, and 2Cl across the plasma membrane of cells. Na+‐K+‐2Cl cotransport is driven by the chemical gradient of the three ionic species across the membrane, two of them maintained by the action of the Na+/K+ pump. In many cells, NKCC1 accumulates Cl above its electrochemical potential equilibrium, thereby facilitating Cl channel‐mediated membrane depolarization. In smooth muscle cells, this depolarization facilitates the opening of voltage‐sensitive Ca2+ channels, leading to Ca2+ influx, and cell contraction. In immature neurons, the depolarization due to a GABA‐mediated Cl conductance produces an excitatory rather than inhibitory response. In many cell types that have lost water, NKCC is activated to help the cells recover their volume. This is specially the case if the cells have also lost Cl. In combination with the Na+/K+ pump, the NKCC's move ions across various specialized epithelia. NKCC1 is involved in Cl‐driven fluid secretion in many exocrine glands, such as sweat, lacrimal, salivary, stomach, pancreas, and intestine. NKCC1 is also involved in K+‐driven fluid secretion in inner ear, and possibly in Na+‐driven fluid secretion in choroid plexus. In the thick ascending limb of Henle, NKCC2 activity in combination with the Na+/K+ pump participates in reabsorbing 30% of the glomerular‐filtered Na+. Overall, many critical physiological functions are maintained by the activity of the two Na+‐K+‐2Cl cotransporters. In this overview article, we focus on the functional roles of the cotransporters in nonpolarized cells and in epithelia. © 2018 American Physiological Society. Compr Physiol 8:871‐901, 2018.

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. Visual representation of the relationship between the nine human SLC12A cotransporters. The cluster dendrogram highlights two distinct subfamilies: the Na+‐dependent branch on the upper left from the Na+‐independent branch of the bottom right. There are also two orphan members, CCC8 and CCC9 that as yet do not have specific functions or substrates assigned to them. The two Na+‐K+‐2Cl cotransporters are inhibited by the loop diuretic bumetanide, whereas, the Na+‐Cl cotransporter is inhibited by thiazides diuretics, such as metolazone and hydrochlorothiazide. The four K+‐Cl cotransporters are inhibited by furosemide, another loop diuretic, and ML077 a novel more potent inhibitor. The length of dendrogram branches represents the number of amino acid substitutions per site (bar = 0.1 amino acid substitutions per site).
Figure 2. Figure 2. Model of transmembrane core of NKCC1. Model of NKCC1 core obtained by I‐TASSER based on the crystal structure of several related transporters such as ApcT K158A transporter (PDB: 3gi8C), L‐arginine/agmatine antiporter (PDB: 5j4iA), glutamate‐GABA antiporter GadC (PDB: 4djiA), LeuT (PDB: 2g6HA). (A). The long alpha‐helices representing the transmembrane (TM) domains are displayed as solid straight tubes. Highlighted are TM2 and TM7, which run parallel to each other. (B). Note that the valine (Val) and serine (Ser) residues on TM2 that affect the Na+ affinity are close to the extracellular domain, whereas the two methionine (Met) residues that affect the K+ affinity are located deeper in the topology of the cotransporter. Alanine (Ala), isoleucine (Ile), and leucine (Leu) residues with similar properties are identified on TM7.
Figure 3. Figure 3. Exon structure of human SLC12A2 (NKCC1) and SLC12A1 (NKCC2). The 27 exons of both the NKCC1 and NKCC2 genes have a high degree of conservation that diverged at the beginning of vertebrate evolution. Exon 1 in the NKCC1 gene (orange) is split into two exons in the NKCC2 gene (blue). As a result, exons 2 thru 20 in NKCC1 align with exons 3 thru 21 in NKCC2. Highlighted in light blue is exon 21 in NKCC1 that is missing in NKCC2, resulting in a realigning of exons 22 thru 27 for both genes. Also highlighted in light blue is exon 4 in the NKCC2 gene that is alternatively spliced giving three mutually exclusive variants (A, B, and F) which provides distinct affinities for Cl. We have also highlighted the shorter isoform of NKCC2 found in mouse kidney formed by extension of exon 17 (C4 3′ end). In NKCC1, a small 16 amino acid residue exon (exon 21) is alternatively spliced. This exon is absent in the NKCC2 gene.
Figure 4. Figure 4. Role of NKCC1 in colonic epithelium. The figure separates the surface cells from the crypt cells. (A) In the surface cells, apical Na+ channels and basolateral Na+/K+‐ATPase (pump) participate in Na+ reabsorption. (B) In cells from the crypts, K+ and Cl channels are expressed on both the apical and basolateral membrane and cells can mediate preferentially Cl secretion or K+ secretion, depending on stimuli. Many basolateral signals (e.g., VIP, ATP, AngII, and Epinephrine) lead to activation of adenylate cyclase (AC) and production of cAMP, which stimulates CFTR Cl channels on the apical membrane through protein kinase A (PKA). Cl is replenished at the serosal (basolateral) side by NKCC1. Part of the Cl that is secreted can be exchanged for HCO3, which enters the basolateral membrane through the Na+/HCO3 cotransporter (NBC1). In this Cl secretory mode, a basolateral K+ conductance is needed to recycle the K+ that enters through the Na+/K+‐ATPase (Pump) and NKCC1. Note that the crypt cells are also able to secrete K+ when Cl secretion is “silent.” In this case, a Cl conductance is needed on the basolateral membrane to recycle Cl.
Figure 5. Figure 5. NKCC1 in choroid plexus. (A) Intense apical staining of NKCC1 in mouse choroid plexus is evidenced by expression at the surface of finger‐like structures. The rabbit polyclonal antibody was raised against a 74 amino acid portion of the COOH‐terminal tail of the cotransporter (142). Bar = 50 μm. (B) Two possible models of NKCC1 function in choroid plexus (CP) epithelial cells are illustrated. When carbonic anhydrase (CA) activity is high, exchange of Cl for HCO3 accumulates Cl into the CP epithelial cell leading to outward NKCC1 transport. In this situation, NKCC1 participates in Na+ secretion and CSF production. Intracerebral application of bumetanide slows down the rate of CSF production. In contrast, if CA activity is low, intracellular Cl might be lower and the gradients now favor typical inward NKCC1‐mediated transport. The cotransporter in this case might participate in the reabsorption of K+.
Figure 6. Figure 6. Two cavities in the inner ear (cochlear and endolymphatic sac) with very different ionic compositions. The two compartments have distinct K+ concentrations: 140 mmol/L in the cochlear endolymph and 13 mmol/L in the endolymphatic sac. The stria vascularis produces the K+‐rich endolymphatic fluid. It is a multilayered epithelium composed of basal fibrocytes (Fibrocytes), basal cells (BC), intermediate cells (IC), and marginal cells (MC). The epithelium expresses NKCC1 in abundance as well as Na+/K+‐ATPase (pump) and K+ channels (Kir4.1 and Kv7.1). Gap junctions connect fibrocytes to BC and IC. The driving force for K+ secretion, like in many other epithelia, is the Na+/K+ pump. The cochlea connects to the endolymphatic sac via a narrow canal.
Figure 7. Figure 7. K+ leak generates varying membrane potentials in different cell types and the endolymphatic potential of the inner ear. (A) The leak of K+ ions from cytosol to the extracellular environment creates a membrane potential (Vm) that varies from ‐8 mV in chondrocytes to ‐70 mV in neurons to ‐95 mV in skeletal muscle. The minus sign indicates that the inside of the cell is negative with respect to the extracellular environment. (B) Similarly, the leak of K+ from the K+‐rich endolymphatic cavity to the interstitium creates the endolymphatic potential (EP). Note the reversed sign (+80‐90 mV), which indicates that the interstitial environment is positive with respect to the endolymphatic cavity. (C) Anatomy of a turn of the cochlea from the inner ear of a wild‐type mouse: SM: scala media, SV: Scala Vestibuli, St.v. stria vascularis, sl: spiral ligament, tm: tectorial membrane, oc: outer hair cells, Rm: Reissner's membrane. (D) The scala media and Reissner's membrane are intact in the inner ear of heterozygous NKCC1 knockout mice. (E) However, in homozygous NKCC1 knockout mice, Reissner's membrane is collapsed and there is no sign of the scala media. Bar = 50 μm.
Figure 8. Figure 8. Evidence of apoptosis in the seminiferous tubule (gonads) of NKCC1 knockout mice. Panels A and B show thionin‐stained sections treated with DIG‐dUTP, followed by a peroxidase‐conjugated anti‐DIG antibody. Note the presence of only one cell undergoing apoptosis at the base of a seminiferous tubule in a wild‐type control mouse (box in A), whereas multiple apoptotic cells are present in the seminiferous tubule from the homozygous NKCC1 knockout mouse (black arrows in B). Spermatids (sp) are numerous in wild‐type (A), but absent in the knockout mouse (B). Black arrowheads point to spermatogonia. Panels C and D show fluorescein‐based in situ cell death‐detection method. Sections were fixed with paraformaldehyde and exposed to fluorescein‐dUTP. Note the background fluorescence given by spermatids in the seminiferous tubules from control mice (white arrowheads), but absence of TUNEL staining at the base of the seminiferous tubules (C). In the knockout (D), many apoptotic cells are labeled (white arrows). Scale bars = 50 μmol/L.
Figure 9. Figure 9. Plot of testis versus body weights in heterozygous versus homozygous NKCC1 knockout mice. Adult wild‐type (not shown) and heterozygous NKCC1 knockout mice weighing around 30 g had testis weighing around 200 mg. Severe deficits in both body (60%) and testis (25%) weights were noted in homozygous NKCC1 knockout mice.
Figure 10. Figure 10. Model of ion transport in Thick Ascending Limb of Henle (TAL). (A) Multiple transport mechanisms act in concert to mediate Na+ reabsorption in the TAL. Na+ enters the apical membrane through NKCC2 and leaves the basolateral membrane through the Na+/K+‐ATPase (pump). Because K+ is limiting in the urine, NKCC2 works in concert with an apical K+ channel: ROMK, which provides K+ for K+‐coupled Na+ transport via NKCC2. The Cl that enters the cells via NKCC2 on the apical side is extruded by CLCNK Cl channels. The combined effects of K+ leak through ROMK on the apical membrane and Cl leak through CLCNKA/B on the basolateral membrane creates an electropositive lumen that facilitates the movement of divalent cations (Ca2+ and Mg2+) through paracellular pathways. NKCC2 is inhibited by the “loop” diuretic furosemide (LASIX). (B) Absence of Na+ reabsorption (or Na+ wasting) occurs in conditions of loss‐of‐function mutations in these transport pathways. Mutations in NKCC2 lead to Bartter's syndrome type I (BARTS1), mutations in ROMK cause Bartter's syndrome type II (BARTS2), and mutations in CLCNK channels or in Barttin (an associated protein) lead to Bartter's syndrome types III and IV (BARTS3 and 4).
Figure 11. Figure 11. Depiction of cAMP as a “node” in NKCC2 regulation. Activation of adenylate cyclase by G‐coupled protein αs (stimulated by vasopressin receptors) and inhibition by Gαi (activated by prostaglandin receptor) regulates the synthesis of cAMP from ATP. cAMP is degraded into AMP by phosphodiesterases, one of which PDE2, is under the control of the PI3K‐Akt‐eNOS, nitric oxide, cGMP signaling pathway initiated by the endothelin receptor. cAMP has genomic effects, binding to CRE element in the NKCC2 promoter, leading to increased transcription. cAMP also stimulates PKA which plays a role in the forward trafficking of NKCC2 to the plasma membrane. Synaptic protein, VAMP2, is involved in the PKA‐mediated trafficking process.


Figure 1. Visual representation of the relationship between the nine human SLC12A cotransporters. The cluster dendrogram highlights two distinct subfamilies: the Na+‐dependent branch on the upper left from the Na+‐independent branch of the bottom right. There are also two orphan members, CCC8 and CCC9 that as yet do not have specific functions or substrates assigned to them. The two Na+‐K+‐2Cl cotransporters are inhibited by the loop diuretic bumetanide, whereas, the Na+‐Cl cotransporter is inhibited by thiazides diuretics, such as metolazone and hydrochlorothiazide. The four K+‐Cl cotransporters are inhibited by furosemide, another loop diuretic, and ML077 a novel more potent inhibitor. The length of dendrogram branches represents the number of amino acid substitutions per site (bar = 0.1 amino acid substitutions per site).


Figure 2. Model of transmembrane core of NKCC1. Model of NKCC1 core obtained by I‐TASSER based on the crystal structure of several related transporters such as ApcT K158A transporter (PDB: 3gi8C), L‐arginine/agmatine antiporter (PDB: 5j4iA), glutamate‐GABA antiporter GadC (PDB: 4djiA), LeuT (PDB: 2g6HA). (A). The long alpha‐helices representing the transmembrane (TM) domains are displayed as solid straight tubes. Highlighted are TM2 and TM7, which run parallel to each other. (B). Note that the valine (Val) and serine (Ser) residues on TM2 that affect the Na+ affinity are close to the extracellular domain, whereas the two methionine (Met) residues that affect the K+ affinity are located deeper in the topology of the cotransporter. Alanine (Ala), isoleucine (Ile), and leucine (Leu) residues with similar properties are identified on TM7.


Figure 3. Exon structure of human SLC12A2 (NKCC1) and SLC12A1 (NKCC2). The 27 exons of both the NKCC1 and NKCC2 genes have a high degree of conservation that diverged at the beginning of vertebrate evolution. Exon 1 in the NKCC1 gene (orange) is split into two exons in the NKCC2 gene (blue). As a result, exons 2 thru 20 in NKCC1 align with exons 3 thru 21 in NKCC2. Highlighted in light blue is exon 21 in NKCC1 that is missing in NKCC2, resulting in a realigning of exons 22 thru 27 for both genes. Also highlighted in light blue is exon 4 in the NKCC2 gene that is alternatively spliced giving three mutually exclusive variants (A, B, and F) which provides distinct affinities for Cl. We have also highlighted the shorter isoform of NKCC2 found in mouse kidney formed by extension of exon 17 (C4 3′ end). In NKCC1, a small 16 amino acid residue exon (exon 21) is alternatively spliced. This exon is absent in the NKCC2 gene.


Figure 4. Role of NKCC1 in colonic epithelium. The figure separates the surface cells from the crypt cells. (A) In the surface cells, apical Na+ channels and basolateral Na+/K+‐ATPase (pump) participate in Na+ reabsorption. (B) In cells from the crypts, K+ and Cl channels are expressed on both the apical and basolateral membrane and cells can mediate preferentially Cl secretion or K+ secretion, depending on stimuli. Many basolateral signals (e.g., VIP, ATP, AngII, and Epinephrine) lead to activation of adenylate cyclase (AC) and production of cAMP, which stimulates CFTR Cl channels on the apical membrane through protein kinase A (PKA). Cl is replenished at the serosal (basolateral) side by NKCC1. Part of the Cl that is secreted can be exchanged for HCO3, which enters the basolateral membrane through the Na+/HCO3 cotransporter (NBC1). In this Cl secretory mode, a basolateral K+ conductance is needed to recycle the K+ that enters through the Na+/K+‐ATPase (Pump) and NKCC1. Note that the crypt cells are also able to secrete K+ when Cl secretion is “silent.” In this case, a Cl conductance is needed on the basolateral membrane to recycle Cl.


Figure 5. NKCC1 in choroid plexus. (A) Intense apical staining of NKCC1 in mouse choroid plexus is evidenced by expression at the surface of finger‐like structures. The rabbit polyclonal antibody was raised against a 74 amino acid portion of the COOH‐terminal tail of the cotransporter (142). Bar = 50 μm. (B) Two possible models of NKCC1 function in choroid plexus (CP) epithelial cells are illustrated. When carbonic anhydrase (CA) activity is high, exchange of Cl for HCO3 accumulates Cl into the CP epithelial cell leading to outward NKCC1 transport. In this situation, NKCC1 participates in Na+ secretion and CSF production. Intracerebral application of bumetanide slows down the rate of CSF production. In contrast, if CA activity is low, intracellular Cl might be lower and the gradients now favor typical inward NKCC1‐mediated transport. The cotransporter in this case might participate in the reabsorption of K+.


Figure 6. Two cavities in the inner ear (cochlear and endolymphatic sac) with very different ionic compositions. The two compartments have distinct K+ concentrations: 140 mmol/L in the cochlear endolymph and 13 mmol/L in the endolymphatic sac. The stria vascularis produces the K+‐rich endolymphatic fluid. It is a multilayered epithelium composed of basal fibrocytes (Fibrocytes), basal cells (BC), intermediate cells (IC), and marginal cells (MC). The epithelium expresses NKCC1 in abundance as well as Na+/K+‐ATPase (pump) and K+ channels (Kir4.1 and Kv7.1). Gap junctions connect fibrocytes to BC and IC. The driving force for K+ secretion, like in many other epithelia, is the Na+/K+ pump. The cochlea connects to the endolymphatic sac via a narrow canal.


Figure 7. K+ leak generates varying membrane potentials in different cell types and the endolymphatic potential of the inner ear. (A) The leak of K+ ions from cytosol to the extracellular environment creates a membrane potential (Vm) that varies from ‐8 mV in chondrocytes to ‐70 mV in neurons to ‐95 mV in skeletal muscle. The minus sign indicates that the inside of the cell is negative with respect to the extracellular environment. (B) Similarly, the leak of K+ from the K+‐rich endolymphatic cavity to the interstitium creates the endolymphatic potential (EP). Note the reversed sign (+80‐90 mV), which indicates that the interstitial environment is positive with respect to the endolymphatic cavity. (C) Anatomy of a turn of the cochlea from the inner ear of a wild‐type mouse: SM: scala media, SV: Scala Vestibuli, St.v. stria vascularis, sl: spiral ligament, tm: tectorial membrane, oc: outer hair cells, Rm: Reissner's membrane. (D) The scala media and Reissner's membrane are intact in the inner ear of heterozygous NKCC1 knockout mice. (E) However, in homozygous NKCC1 knockout mice, Reissner's membrane is collapsed and there is no sign of the scala media. Bar = 50 μm.


Figure 8. Evidence of apoptosis in the seminiferous tubule (gonads) of NKCC1 knockout mice. Panels A and B show thionin‐stained sections treated with DIG‐dUTP, followed by a peroxidase‐conjugated anti‐DIG antibody. Note the presence of only one cell undergoing apoptosis at the base of a seminiferous tubule in a wild‐type control mouse (box in A), whereas multiple apoptotic cells are present in the seminiferous tubule from the homozygous NKCC1 knockout mouse (black arrows in B). Spermatids (sp) are numerous in wild‐type (A), but absent in the knockout mouse (B). Black arrowheads point to spermatogonia. Panels C and D show fluorescein‐based in situ cell death‐detection method. Sections were fixed with paraformaldehyde and exposed to fluorescein‐dUTP. Note the background fluorescence given by spermatids in the seminiferous tubules from control mice (white arrowheads), but absence of TUNEL staining at the base of the seminiferous tubules (C). In the knockout (D), many apoptotic cells are labeled (white arrows). Scale bars = 50 μmol/L.


Figure 9. Plot of testis versus body weights in heterozygous versus homozygous NKCC1 knockout mice. Adult wild‐type (not shown) and heterozygous NKCC1 knockout mice weighing around 30 g had testis weighing around 200 mg. Severe deficits in both body (60%) and testis (25%) weights were noted in homozygous NKCC1 knockout mice.


Figure 10. Model of ion transport in Thick Ascending Limb of Henle (TAL). (A) Multiple transport mechanisms act in concert to mediate Na+ reabsorption in the TAL. Na+ enters the apical membrane through NKCC2 and leaves the basolateral membrane through the Na+/K+‐ATPase (pump). Because K+ is limiting in the urine, NKCC2 works in concert with an apical K+ channel: ROMK, which provides K+ for K+‐coupled Na+ transport via NKCC2. The Cl that enters the cells via NKCC2 on the apical side is extruded by CLCNK Cl channels. The combined effects of K+ leak through ROMK on the apical membrane and Cl leak through CLCNKA/B on the basolateral membrane creates an electropositive lumen that facilitates the movement of divalent cations (Ca2+ and Mg2+) through paracellular pathways. NKCC2 is inhibited by the “loop” diuretic furosemide (LASIX). (B) Absence of Na+ reabsorption (or Na+ wasting) occurs in conditions of loss‐of‐function mutations in these transport pathways. Mutations in NKCC2 lead to Bartter's syndrome type I (BARTS1), mutations in ROMK cause Bartter's syndrome type II (BARTS2), and mutations in CLCNK channels or in Barttin (an associated protein) lead to Bartter's syndrome types III and IV (BARTS3 and 4).


Figure 11. Depiction of cAMP as a “node” in NKCC2 regulation. Activation of adenylate cyclase by G‐coupled protein αs (stimulated by vasopressin receptors) and inhibition by Gαi (activated by prostaglandin receptor) regulates the synthesis of cAMP from ATP. cAMP is degraded into AMP by phosphodiesterases, one of which PDE2, is under the control of the PI3K‐Akt‐eNOS, nitric oxide, cGMP signaling pathway initiated by the endothelin receptor. cAMP has genomic effects, binding to CRE element in the NKCC2 promoter, leading to increased transcription. cAMP also stimulates PKA which plays a role in the forward trafficking of NKCC2 to the plasma membrane. Synaptic protein, VAMP2, is involved in the PKA‐mediated trafficking process.
References
 1.Károlyi L, Konrad M, Köckerling A, Ziegler A, Zimmermann DK, Roth B, Wieg C, Grzeschik K‐H, Koch MC, Seyberth HW, Vargas R, Forestier L, Jean G, Deschaux M, Rizzoni GF, Niaudet P, Antignac C, Feldmann D, Lorridon F, Cougoureux E, Laroze F, Alessandri J‐L, David L, Saunier P, Deschenes G, Hildebrandt F, Vollmer M, Proesmans W, Brandis M, van den Heuvel LPWJ, Lemmink HH, Nillesen W, Monnens LAH, Knoers NVAM, GuayWoodford LM, Wright CJ, Madrigal G, Hebert SC. Mutations in the gene encoding the inwardly‐rectifying renal potassium channel, ROMK, cause the antenatal variant of Bartter syndrome: Evidence for genetic heterogeneity. Hum Mol Genet 6: 17‐26, 1997.
 2.Adorante JS, Miller SS. Potassium‐dependent volume regulation in retinal pigment epithelium is mediated by Na,K,Cl cotransport. J Gen Physiol 96: 1153‐1176, 1990.
 3.Aickin CC, Betz WJ, Harris GL. Intracellular chloride and the mechanism for its accumulation in rat lumbrical muscle. J Physiol 411: 437‐455, 1989.
 4.Akar F, Jiang G, Paul RJ, O'Neill WC. Contractile regulation of the Na(+)‐K(+)‐2Cl(−) cotransporter in vascular smooth muscle. Am J Physiol Cell Physiol 281: C579‐C584, 2001.
 5.Akar F, Skinner E, Klein JD, Jena M, Paul RJ, O'Neill WC. Vasoconstrictors and nitrovasodilators reciprocally regulate the Na+‐K+‐2Cl− cotransporter in rat aorta. Am J Physiol 276: C1383‐C1390, 1999.
 6.Alshahrani S, Almutairi MM, Kursan S, Dias‐Junior E, Almiahuob MM, Aguilar‐Bryan L, Di Fulvio M. Increased Slc12a1 expression in β‐cells and improved glucose disposal in Slc12a2 heterozygous mice. J Endocrinol 227: 153‐165, 2015.
 7.Altamirano AA, Breitwieser GE, Russel JM. Vanadate and fluoride effects on Na‐K‐Cl cotransport in squid giant axon. Am J Physiol 254: C582‐C586, 1988.
 8.Alvarez‐Leefmans FJ, Delpire E. Physiology and Pathology of Chloride Transporters and Channels in the Nervous System: From Molecules to Diseases. Academic Press, 2009.
 9.Alvarez‐Leefmans FJ, Gamiño SM, Giraldez F, Nogueron I. Intracellular chloride regulation in amphibian dorsal root ganglion neurons studied with ion‐selective microelectrodes. J Physiol (Lond) 406: 225‐246, 1988.
 10.Alvarez‐Leefmans FJ, León‐Olea M, Mendoza‐Sotelo J, Alvarez FJ, Antón B, Garduño R. Immunolocalization of the Na(+)‐K(+)‐2Cl(−) cotransporter in peripheral nervous tissue of vertebrates. Neuroscience 104: 569‐582, 2001.
 11.Ando Z, Sato S, Ikeda K, Kawakami K. Slc12a2 is a direct target of two closely related homeobox proteins, Six1 and Six4. FEBS Lett 272: 3026‐3041, 2005.
 12.Andrade L, Rodrigues ACJ, Sanches TR, Souza RB, Seguro AC. Leptospirosis leads to dysregulation of sodium transporters in the kidney and lung. Am J Physiol Renal Physiol 292: F586‐F592, 2007.
 13.Ares GR, Caceres P, Alvarez‐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.
 14.Ares GR, Caceres PS, Ortiz PA. Molecular regulation of NKCC2 in the thick ascending limb. Am J Physiol Renal Physiol 301: F1143‐F1159, 2011.
 15.Argenzio RA, Armstrong M. ANP inhibits NaCl absorption and elicits Cl secretion in porcine colon: Evidence for cGMP and Ca mediation. Am J Physiol 265: R57‐R65, 1993.
 16.Ayalon A, Corcia A, Klemperer G, Caplan SR. Suppression of gastric acid secretion by furosemide in isolated gastric mucosa of guinea pig. Am J Physiol 239: G532‐G535, 1980.
 17.Bailly C. Effect of luminal atrial natriuretic peptide on chloride reabsorption in mouse cortical thick ascending limb: Inhibition by endothelin. J Am Soc Nephrol 11: 1791‐1797, 2000.
 18.Bairamian D, Johanson CE, Parmelee JT, Epstein MH. Potassium cotransport with sodium and chloride in the choroid plexus. J Neurochem 56: 1623‐1629, 1991.
 19.Bakker‐Grunwald T. Effect of anions of potassium self‐exchange in ascites tumor cells. Biochim Biophys Acta 513: 292‐295, 1978.
 20.Barber HS. The mercurial diuretics. Postgrad Med J 14: 288‐294, 1938.
 21.Baron A, Frieden M, Chabaud F, Bény JL. Ca(2+)‐dependent non‐selective cation and potassium channels activated by bradykinin in pig coronary artery endothelial cells. J Physiol 493: 691‐706, 1996.
 22.Bartke A. Increased sensitivity of seminal vesicles to testosterone in a mouse strain with low plasma testosterone levels. J Endocrinol 60: 145‐148, 1974.
 23.Begum G, Yuan H, Kahle KT, Li L, Wang S, Shi Y, Shmukler BE, Yang SS, Lin SH, Alper SL, Sun D. Inhibition of WNK3 kinase signaling reduces brain damage and accelerates neurological recovery after stroke. Stroke 46: 1956‐1965, 2015.
 24.Ben‐Ari Y. Excitatory actions of gaba during development: The nature of the nurture. Nat Rev Neurosci 3: 728‐739, 2002.
 25.Ben‐Ari Y, Cherubini E, Corradetti R, Gaiarsa JL. Giant synaptic potentials in immature rat CA3 hippocampal neurones. J Physiol (London) 416: 303‐325, 1989.
 26.Bergaya S, Faure S, Baudrie V, Rio M, Escoubet B, Bonnin P, Henrion D, Loirand G, Achard JM, Jeunemaitre X, Hadchouel J. WNK1 regulates vasoconstriction and blood pressure response to a 1‐adrenergic stimulation in mice. Hypertension 58: 439‐445, 2011.
 27.Birkenhager R, Otto E, Schurmann MJ, Vollmer M, Ruf E‐M, Maier‐Lutz I, Beekmann F, Fekete A, Omran H, Feldmann D, Milford DV, Jeck N, Konrad M, Landau D, Knoers NVAM, Antignac C, Sudbrak R, Kispert A, Hildebrandt F. Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nature Genet 29: 310‐314, 2001.
 28.Bouyer PG, Tang X, Weber CR, Shen L, Turner JR, Matthews JB. Capsaicin induces NKCC1 internalization and inhibits chloride secretion in colonic epithelial cells independently of TRPV1. Am J Physiol Gastrointest Liver Physiol 304: G142‐G156, 2013.
 29.Brock TA, Brugnara C, Canessa M, Gimbrone MAJ. Bradykinin and vasopressin stimulate Na+‐K+‐Cl− cotransport in cultured endothelial cells. Am J Physiol 250: C888‐C895, 1986.
 30.Bulley S, Jaggar JH. Cl− channels in smooth muscle cells. Pflügers Arch Eur J Physiol 466: 5861‐5872, 2014.
 31.Burg M, Stoner L, Cardinal J, Green N. Furosemide effect on isolated perfused tubules. Am J Physiol 225: 119‐124, 1973.
 32.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.
 33.Caceres PS, Mendez M, Ortiz PA. Vesicle‐associated membrane protein 2 (VAMP2) but Not VAMP3 mediates cAMP‐stimulated trafficking of the renal Na+‐K+‐2Cl− co‐transporter NKCC2 in thick ascending limbs. J Biol Chem 289: 23951‐24962, 2014.
 34.Cala PM. Volume regulation by Amphiuma red blood cells: The membrane potential and its implications regarding the nature of the ion‐flux pathways. J Gen Physiol 76: 683‐708, 1980.
 35.Callaway E. The revolution will not be crystallized: A new method sweeps through structural biology. Nature 525: 172‐174, 2015.
 36.Carmosino M, Giménez I, Caplan M, Forbush B. Exon loss accounts for differential sorting of Na‐K‐Cl cotransporters in polarized epithelial cells. Mol Biol Cell 19: 4341‐4351, 2008.
 37.Casimiro MC, Knollmann BC, Ebert SN, Vary JC, Jr., Greene AE, Franz MR, Grinberg A, Huang SP, Pfeifer K. Targeted disruption of the Kcnq1 gene produces a mouse model of Jervell and Lange‐Nielsen syndrome. Proc Natl Acad Sci U S A 98: 2526‐2531, 2001.
 38.Castrop H, Lorenz JN, Hansen PB, Friis U, Mizel D, Oppermann M, Jensen BL, Briggs J, Skøtt O, Schnermann J. Contribution of the basolateral isoform of the Na‐K‐2Cl‐ cotransporter (NKCC1/BSC2) to renin secretion. Am J Physiol Renal Physiol 289: F1185‐F1192, 2005.
 39.Castrop H, Schnermann J. Isoforms of renal Na‐K‐2Cl cotransporter NKCC2: Expression and functional significance. Am J Physiol Renal Physiol 295: F859‐F866, 2008.
 40.Chen H, Luo J, Kintner DB, Shull GE, Sun D. Na(+)‐dependent chloride transporter (NKCC1)‐null mice exhibit less gray and white matter damage after focal cerebral ischemia. J Cereb Blood Flow Metab 25: 54‐66, 2005.
 41.Chen X, Kintner DB, Luo J, Baba A, Matsuda T, Sun D. Endoplasmic reticulum Ca2+ dysregulation and endoplasmic reticulum stress following in vitro neuronal ischemia: Role of Na+‐K+‐Cl− cotransporter. J Neurochem 106: 1563‐1576, 2008.
 42.Cho HM, Lee DY, Kim HY, Lee HA, Seok YM, Kim IK. Upregulation of the Na(+)‐K(+)‐2Cl(−) cotransporter 1 via histone modification in the aortas of angiotensin II‐induced hypertensive rats. Hypertens Res 35: 819‐824, 2012.
 43.Cho HM, Lee HA, Kim HY, Lee DY, Kim IK. Recruitment of specificity protein 1 by CpG hypomethylation upregulates Na+‐K+‐2Cl− cotransporter 1 in hypertensive rats. J Hypertens 31: 1406‐1413, 2013.
 44.Chou CL, Yu MJ, Kassai EM, Morris RG, Hoffert JD, Wall SM, Knepper MA. Roles of basolateral solute uptake via NKCC1 and of myosin II in vasopressin‐induced cell swelling in inner medullary collecting duct. Am J Physiol Renal Physiol 295: F192‐F201, 2008.
 45.Ciuman RR. Stria vascularis and vestibular dark cells: Characterisation of main structures responsible for inner‐ear homeostasis, and their pathophysiological relations. J Laryngol Otol 123: 151‐162, 2009.
 46.Crouch JJ, Sakaguchi N, Lytle C, Schulte BA. Immunohistochemical localization of the Na‐K‐Cl co‐transporter (NKCC1) in the gerbil inner ear. J Histochem 45: 773‐778, 1997.
 47.Culp DJ, Quivey RQ, Bowen WH, Fallon MA, Pearson SK, Faustoferri R. A mouse caries model and evaluation of aqp5‐/‐ knockout mice. Caries Res 39: 448‐454, 2005.
 48.Cuthbert AW, Hickman ME. Indirect effects of adenosine triphosphate on chloride secretion in mammalian colon. J Membrane Biol 86: 157‐166, 1985.
 49.Cuthbert AW, MacVinish LJ, Hickman ME, Ratcliff R, Colledge WH, Evans MJ. Ion‐transporting activity in the murine colonic epithelium of normal animals and animals with cystic fibrosis. Pflugers Arch 428: 508‐515, 1994.
 50.Dagher G, Brugnara C, Canessa M. Effect of metabolic depletion on the furosemide‐sensitive Na and K fluxes in human red cells. J Membrane Biol 86: 145‐155, 1985.
 51.Daigle ND, Carpentier GA, Frenette‐Cotton R, Simard MG, Lefoll MH, Noël M, Caron L, Noël J, Isenring P. Molecular characterization of a human cation‐Cl− cotransporter (SLC12A8A, CCC9A) that promotes polyamine and amino acid transport. J Cell Physiol 220: 680‐689, 2009.
 52.Damkier HH, Brown PD, Praetorius J. Cerebrospinal fluid secretion by the choroid plexus. Physiol Rev 93: 1847‐1892, 2013.
 53.Dan I, Watanabe NM, Kusumi A. The Ste20 group kinases as regulators of MAP kinase cascades. Trends Cell Biol 11: 220‐230, 2001.
 54.Darman RB, Forbush B. A regulatory locus of phosphorylation in the N terminus of the Na‐K‐Cl cotransporter, NKCC1. J Biol Chem 277: 37542‐37550, 2002.
 55.Dartsch PC, Ritter M, Häussinger D, Lang F. Cytoskeletal reorganization in NIH 3T3 fibroblasts expressing the ras oncogene. Eur J Cell Biol 63: 316‐325, 1994.
 56.Dartt DA, Møller M, Poulsen JH. Lacrimal gland electrolyte and water secretion in the rabbit: Localization and role of (Na+ + K+)‐activated ATPase. J Physiol 321: 557‐569, 1981.
 57.Dayioglu E, Buharalioglu CK, Saracoglu F, Akar F. The effects of bumetanide on human umbilical artery contractions. Reprod Sci 14: 246‐252, 2007.
 58.DeFazio RA, Heger S, Ojeda SR, Moenter SM. Activation of A‐type gamma‐aminobutyric acid receptors excites gonadotropin‐releasing hormone neurons. Mol Endocrinol 16: 2872‐2891, 2002.
 59.del Castillo JR, Arévalo JC, Burguillos L, Súlbaran‐Carrasco MC. beta‐adrenergic agonists stimulate Na+‐K+‐Cl− cotransport by inducing intracellular Ca2+ liberation in crypt cells. Am J Physiol 277: G563‐G571, 1999.
 60.Delpire E. The Mammalian family of Sterile20p‐like protein kinases. Pflügers Arch 458: 953‐967, 2009.
 61.Delpire E, Gagnon KB. Genome‐wide analysis of SPAK/OSR1 binding motifs. Physiol Genomics 28: 223‐231, 2007.
 62.Delpire E, Gagnon KB. Kinetics of hyperosmotically‐stimulated Na‐K‐2Cl cotransporter in Xenopus laevis oocytes. Am J Physiol Cell Physiol 301: C1074‐C1085, 2011.
 63.Delpire E, Lu J, England R, Dull C, Thorne T. Deafness and imbalance associated with inactivation of the secretory Na‐K‐2Cl co‐transporter. Nat Genet 22: 192‐195, 1999.
 64.Delpire E, Rauchman MI, Beier DR, Hebert SC, Gullans SR. Molecular cloning and chromosome localization of a putative basolateral Na‐K‐2Cl cotransporter from mouse inner medullary collecting duct (mIMCD‐3) cells. J Biol Chem 269: 25677‐25683, 1994.
 65.Delpire E, Wolfe L, Flores B, Koumangoye R, Schornak CC, Omer S, Pusey B, Lau C, Markello T, Adams DR. A patient with multisystem dysfunction carries a truncation mutation in human SLC12A2, the gene encoding the Na‐K‐2Cl cotransporter, NKCC1. Cold Spring Harb Mol Case Studies 2: a001289, 2016.
 66.Delporte C, O'Connell BC, He X, Lancaster HE, O'Connell AC, Agre P, Baum BJ. Increased fluid secretion after adenoviral‐mediated transfer of the aquaporin‐1 cDNA to irradiated rat salivary glands. Proc Natl Acad Sci U S A 94: 3268‐3273, 1997.
 67.Dharmsathaphorn K, Mandel KG, Masui H, McRoberts JA. Vasoactive intestinal polypeptide‐induced chloride secretion by a colonic epithelial cell line. Direct participation of a basolaterally localized Na+,K+,Cl‐cotransport system. J Clin Invest 75: 462‐471, 1985.
 68.Diaz RC, Vazquez AE, Dou H, Wei D, Cardell EL, Lingrel J, Shull GE, Doyle KJ, Yamoah EN. Conservation of hearing by simultaneous mutation of Na,K‐ATPase and NKCC1. J Assoc Res Otolaryngol 8: 422‐434, 2007.
 69.Dixon MJ, Gazzard J, Chaudhry SS, Sampson N, Schulte BA, Steel KP. Mutation of the Na‐K‐Cl co‐transporter gene Slc12a2 results in deafness in mice. Hum Mol Genet 8: 1579‐1584, 1999.
 70.Dolganov GM, Woodruff PG, Novikov AA, Zhang Y, Ferrando RE, Szubin R, Fahy J. A novel method of gene transcript profiling in airway biopsy homogenates reveals increased expression of a Na+‐K+‐Cl− cotransporter (NKCC1) in asthmatic subjects. Genome Res 11: 1473‐1483, 2001.
 71.Douglas IJ, Brown PD. Regulatory volume increase in rat lacrimal gland acinar cells. J Membr Biol 150: 209‐217, 1996.
 72.Dowd BF, Forbush B. PASK (proline‐alanine‐rich STE20‐related kinase), a regulatory kinase of the Na‐K‐Cl cotransporter (NKCC1). J Biol Chem 278: 27347‐27353, 2003.
 73.Dzhala VI, Brumback AC, Staley K. Bumetanide enhances phenobarbital efficacy in a neonatal seizure model. Ann Neurol 63: 222‐235, 2008.
 74.Dzhala VI, Kuchibhotla KV, Glykys JC, Kahle KT, Swiercz WB, Feng G, Kuner T, Augustine GJ, Bacskai BJ, Staley KJ. Progressive NKCC1‐dependent neuronal chloride accumulation during neonatal seizures. J Neurosci 30: 11745‐11761, 2010.
 75.Dzhala VI, Talos DM, Sdrulla DA, Brumback AC, Mathews GC, Benke TA, Delpire E, Jensen FE, Staley KJ. NKCC1 transporter facilitates seizures in the developing brain. Nat Med 11: 1205‐1213, 2005.
 76.Evans RL, Turner RJ. Upregulation of Na(+)‐K(+)‐2Cl− cotransporter activity in rat parotid acinar cells by muscarinic stimulation. J Physiol 499: 351‐359, 1997.
 77.Fernandez‐Llama P, Ecelbarger CA, Ware JA, Andrews P, Lee AJ, Turner R, Nielsen S, Knepper MA. Cyclooxygenase inhibitors increase Na‐K‐2Cl cotransporter abundance in thick ascending limb of Henle's loop. Am J Physiol (Renal Physiol) 277: F219‐F226, 1999.
 78.Filippi BM, de los Heros P, Mehellou Y, Navratilova I, Gourlay R, Deak M, Plater L, Toth R, Zeqiraj E, Alessi DR. MO25 is a master regulator of SPAK/OSR1 and MST3/MST4/YSK1 protein kinases. Embo J 30: 1730‐1741, 2011.
 79.Flagella M, Clarke LL, Miller ML, Erway LC, Giannella RA, Andringa A, Gawenis LR, Kramer J, Duffy JJ, Doetschman T, Lorenz JN, Yamoah EN, Cardell EL, Shull GE. Mice lacking the basolateral Na‐K‐2Cl cotransporter have impaired epithelial chloride secretion and are profoundly deaf. J Biol Chem 274: 26946‐26955, 1999.
 80.Flatman PW. The effects of magnesium on potassium transport in ferret red cells. J Physiol (Lond) 397: 471‐487, 1988.
 81.Flemmer AW, Forbush BI. Changes in amount and phosphorylation state of the Na‐K‐Cl cotransporter (NKCC) in teleost gill during salt adaptation. FASEB J 13: A399, 1999.
 82.Foster ES, Dudeja PK, Brasitus TA. Contribution of Cl(−)‐OH‐ exchange to electroneutral NaCl absorption in rat distal colon. Am J Physiol 258: G261‐G267, 1990.
 83.Frank J. Advances in the field of single‐particle cryo‐electron microscopy over the last decade. Nature Protocols 12: 209‐212, 2017.
 84.Gabriel SE, Brigman KN, Koller BH, Boucher RC, Stutts MJ. Cystic fibrosis heterozygote resistance to cholera toxin in the cystic fibrosis mouse model. Science 266: 107‐109, 1994.
 85.Gagnon E, Forbush B, Caron L, Isenring P. Functional comparison of renal Na‐K‐Cl cotransporters between distant species. Am J Physiol Cell Physiol 284: C365‐C370, 2003.
 86.Gagnon KB, Delpire E. Molecular determinants of hyperosmotically activated NKCC1‐mediated K+/K+ exchange. J Physiol (Lond) 588: 3385‐3396, 2010.
 87.Gagnon KB, Delpire E. Molecular physiology of SPAK and OSR1: Two Ste20‐related protein kinases regulating ion transport. Physiol Rev 92: 1577‐1617, 2012.
 88.Gagnon KB, Delpire E. Physiology of SLC12 transporters: Lessons from inherited human genetic mutations and genetically‐engineered mouse knockouts. Am J Physiol Cell Physiol 304: C693‐C714, 2013.
 89.Gagnon KB, England R, Delpire E. Characterization of SPAK and OSR1, regulatory kinases of the Na‐K‐2Cl cotransporter. Mol Cell Biol 26: 689‐698, 2006.
 90.Gagnon KB, England R, Delpire E. Volume sensitivity of cation‐chloride cotransporters is modulated by the interaction of two kinases: SPAK and WNK4. Am J Physiol Cell Physiol 290: C134‐C142, 2006.
 91.Gagnon KB, England R, Diehl L, Delpire E. Apoptosis associated tyrosine kinase scaffolding of protein phosphatase 1 and SPAK reveals a novel pathway for Na‐K‐2Cl cotransporter regulation. Am J Physiol (Cell Physiol) 292: C1809‐C1815, 2007.
 92.Gamba G, Miyanoshita A, Lombardi M, Lytton J, Lee W‐S, Hediger M, Hebert SC. Molecular cloning, primary structure, and characterization of two members of the mammalian electroneutral sodium‐(potassium)‐chloride cotransporter family expressed in kidney. J Biol Chem 269: 17713‐17722, 1994.
 93.Gamba G, Saltzberg SN, Lombardi M, Miyanoshita A, Lytton J, Hediger MA, Brenner BM, Hebert SC. Primary structure and functional expression of a cDNA encoding the thiazide‐sensitive, electroneutral sodium‐chloride cotransporter. Proc Natl Acad Sci U S A 90: 2749‐2753, 1993.
 94.Garg P, Martin CF, Elms SC, Gordon FJ, Wall SM, Garland CJ, Sutliff RL, O'Neill WC. Effect of the Na‐K‐2Cl cotransporter NKCC1 on systemic blood pressure and smooth muscle tone. Am J Physiol Heart Circ Physiol 292: H2100‐H2105, 2007.
 95.Geck P, Pietrzyk C, Burckhardt B‐C, Pfeiffer B, Heinz E. Electrically silent cotransport of Na+, K+ and Cl− in Ehrlich cells. Biochim Biophys Acta 600: 432‐447, 1980.
 96.Geng Y, Hoke A, Delpire E. The Ste20 kinases SPAK and OSR1 regulate NKCC1 function in sensory neurons. J Biol Chem 284: 14020‐14028, 2009.
 97.Gillen CM, Brill S, Payne JA, Forbush BI. Molecular cloning and functional expression of the K‐Cl cotransporter from rabbit, rat, and human. A new member of the cation‐chloride cotransporter family. J Biol Chem 271: 16237‐16244, 1996.
 98.Gillie DJ, Pace AJ, Coakley RJ, Koller BH, Barker PM. Liquid and ion transport by fetal airway and lung epithelia of mice deficient in sodium‐potassium‐2‐chloride transporter. Am J Respir Cell Mol Biol 25: 14‐20, 2001.
 99.Giménez I, Forbush B. Short‐term stimulation of the renal Na‐K‐Cl cotransporter (NKCC2) by vasopressin involves phosphorylation and membrane translocation of the protein. J Biol Chem 278: 26946‐26951, 2003.
 100.Giménez I, Isenring P, Forbush B. Spatially distributed alternative splice variants of the renal Na‐K‐Cl cotransporter exhibit dramatically different affinities for the transported ions. J Biol Chem 277: 8767‐8770, 2002.
 101.Ginns SM, Knepper MA, Ecelbarger CA, Terris J, He X, Coleman RA, Wade JB. Immunolocalization of the secretory isoform of Na‐K‐Cl cotransporter in rat renal intercalated cells. J Am Soc Nephrol 7: 2533‐2542, 1996.
 102.Glanville M, Kingscote S, Thwaites DT, Simmons NL. Expression and role of sodium, potassium, chloride cotransport (NKCC1) in mouse inner medullary collecting duct (mIMCD‐K2) epithelial cells. Pflugers Arch 443: 123‐131, 2001.
 103.Glykys J, Dzhala V, Egawa K, Kahle KT, Delpire E, Staley K. Chloride, seizures, and edema: A relationship with therapeutic potential. Trends Neurosci 40: 176‐194, 2017.
 104.Gosmanov AR, Wong JA, Thomason DB. Duality of G protein‐coupled mechanisms for beta‐adrenergic activation of NKCC activity in skeletal muscle. Am J Physiol (Cell Physiol) 283: C1025‐C1032, 2002.
 105.Granados‐Soto V, Arguelles CF, Alvarez‐Leefmans FJ. Peripheral and central antinociceptive action of Na–K–2Cl cotransporter blockers on formalin‐induced nociception in rats. Pain 114: 231‐238, 2005.
 106.Grimm PR, Taneja TK, Liu J, Coleman R, Chen YY, Delpire E, Wade JB, Welling PA. SPAK isoforms and OSR1 regulate sodium‐chloride co‐transporters in a nephron‐specific manner. J Biol Chem 287: 37673‐37690, 2012.
 107.Grinstein S, Clarke CA, Rothstein A. Activation of Na+/H+ exchange in lymphocytes by osmotically induced volume changes and by cytoplasmic acidification. J Gen Physiol 82: 619‐638, 1983.
 108.Grubb BR, Lee E, Pace AJ, Koller BH, Boucher RC. Intestinal ion transport in NKCC1‐deficient mice. Am J Physiol Gastrointest Liver Physiol 279: G707‐G718, 2000.
 109.Grubb BR, Pace AJ, Lee E, Koller BH, Boucher RC. Alterations in airway ion transport in NKCC1‐deficient mice. Am J Physiol Cell Physiol 281: C615‐C623, 2001.
 110.Guggino WB. Gates of Janus: Cystic fibrosis and diarrhea. Trends in Microbiol 2: 91‐94, 1994.
 111.Hamann S, Herrera‐Perez JJ, Zeuthen T, Alvarez‐Leefmans FJ. Cotransport of water by the Na+‐K+‐2Cl(−) cotransporter NKCC1 in mammalian epithelial cells. J Physiol 588: 4089‐4101, 2010.
 112.Han J, Chen Y, Liu Y, Liang Y, Wang X, Liu L, Wang F, Zhang L, Zhang H, Wang H. Common variants of the UMOD promoter associated with blood pressure in a community‐based Chinese cohort. Hypertens Res 35: 769‐774, 2012.
 113.Hansson GC. Cystic fibrosis and chloride secreting diahrreoa. Nature 333, 1988.
 114.He Q, Halm ST, Zhang J, Halm DR. Activation of the basolateral membrane Cl− conductance essential for electrogenic K+ secretion suppresses electrogenic Cl− secretion. Exp Physiol 96: 305‐316, 2011.
 115.He X, Tse CM, Donowitz M, Alper SL, Gabriel SE, Baum BJ. Polarized distribution of key membrane transport proteins in the rat submandibular gland. Pflugers Arch 433: 260‐268, 1997.
 116.Hebert SC, Culpepper RM, Andreoli TE. NaCl transport in mouse medullary thick ascending limbs. II. ADH enhancement of transcellular NaCl cotransport: Origin of transepithelial voltage. Am J Physiol Renal Physiol 241: F432‐F442, 1981.
 117.Heintze K, Stewart CP, Frizzell RA. Sodium‐dependent chloride secretion across rabbit descending colon. Am J Physiol 244: G357‐G375, 1983.
 118.Hendil KB, Hoffmann EK. Cell volume regulation in Ehrlich ascites tumor cells. J Cell Physiol 84: 115‐125, 1974.
 119.Hendy GN, D'Souza‐Li L, Yang B, Canaff L, Cole DE. Mutations of the calcium‐sensing receptor (CASR) in familial hypocalciuric hypercalcemia, neonatal severe hyperparathyroidism, and autosomal dominant hypocalcemia. Hum mut 16: 281‐296, 2000.
 120.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.
 121.Hladky SB, Barrand MA. Fluid and ion transfer across the blood‐brain and blood‐cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers 13: 19, 2016.
 122.Hoffmann EK, Lambert IH, Pedersen SF. Physiology of cell volume regulation in vertebrates. Physiol Rev 89: 193‐277, 2009.
 123.Igarashi P, Vanden Heuvel GB, Quaggin SE, Payne JA, Forbush BI. Cloning, embryonic expression, and chromosomal localization of murine renal Na‐K‐Cl cotransporter (NKCC2). J Am Soc Nephrol 5: 288, 1994.
 124.Igarashi P, Whyte DA, Li K, Nagami GT. Cloning and kidney cell‐specific activity of the promoter of the murine renal Na‐K‐Cl cotransporter gene. J Biol Chem 271: 9666‐9674, 1996.
 125.Ikebe M, Nonoguchi H, Nakayama Y, Tashima Y, Tomita K. Upregulation of the secretory‐type Na(+)/K(+)/2Cl(−)‐cotransporter in the kidney by metabolic acidosis and dehydration in rats. J Am Soc Nephrol 12: 423‐430, 2001.
 126.Isenring P, Forbush BI. Ion and bumetanide binding by the Na‐K‐2Cl cotransporter: Importance of transmembrane domains. J Biol Chem 272: 24556‐24562, 1997.
 127.Isenring P, Jacoby SC, Chang J, Forbush BI. Mutagenic mapping of the Na‐K‐Cl cotransporter for domains involved in ion transport and bumetanide binding. J Gen Physiol 112: 549‐558, 1998.
 128.Isenring P, Jacoby SC, Forbush BI. The role of transmembrane domain 2 in cation transport by the Na‐K‐Cl cotransporter. Proc Natl Acad Sci U S A 95: 7179‐7184, 1998.
 129.Javaheri S. Role of NaCl cotransport in cerebrospinal fluid production: Effects of loop diuretics. J Appl Physiol 71: 795‐800, 1991.
 130.Jennings ML, al‐Rohil N. Kinetics of activation and inactivation of swelling‐stimulated K+/Cl− transport. The volume‐sensitive parameter is the rate constant for inactivation. J Gen Physiol 95: 1021‐1040, 1990.
 131.Jiang C, Kawabe H, Rotin D. The ubiquitin ligase Nedd4L regulates the Na/K/2Cl co‐transporter NKCC1/SLC12A2 in the colon. J Biol Chem 292: 3137‐3145, 2017.
 132.Johanson CE, Palm DE, Dyas ML, Knuckey NW. Microdialysis analysis of effects of loop diuretics and acetazolamide on chloride transport from blood to CSF. Brain Res 641: 121‐126, 1994.
 133.Johanson CE, Sweeney SM, Parmelee JT, Epstein MH. Cotransport of sodium and chloride by the adult mammalian choroid plexus. Am J Physiol (Cell Physiol) 258: C211‐C216, 1990.
 134.Joo NS, Cho HJ, Khansaheb M, Wine JJ. Hyposecretion of fluid from tracheal submucosal glands of CFTR‐deficient pigs. J Clin Invest 120: 3161‐3166, 2010.
 135.Joo NS, Irokawa T, Wu JV, Robbins RC, Whyte RI, Wine JJ. Absent secretion to vasoactive intestinal peptide in cystic fibrosis airway glands. J Biol Chem 277: 50710‐50715, 2002.
 136.Joo NS, Saenz Y, Krouse ME, Wine JJ. Mucus secretion from single submucosal glands of pig. Stimulation by carbachol and vasoactive intestinal peptide. J Biol Chem 277: 28167‐28175, 2004.
 137.Joo NS, Wine JJ, Cuthbert AW. Lubiprostone stimulates secretion from tracheal submucosal glands of sheep, pigs, and humans. Am J Physiol Lung Cell Mol Physiol 296: L811‐L824, 2009.
 138.K. O‐B, Linde A, Verreynne E, Melgarejo T. Salivary gland transcriptome analysis of the Wild living and Captive Spotted Hyena. FASEB J 26: 1120‐1123, 2012.
 139.Kahle KT, Delpire E. Kinase‐KCC2 coupling: Cl− rheostasis, disease susceptibility, therapeutic target. J Neurophysiology 115: 8‐18, 2016.
 140.Kahle KT, Khanna AR, Duan J, Staley KJ, Delpire E, Poduri A. The KCC2 cotransporter and human epilepsy: Getting excited about inhibition. Neuroscientist 22: 555‐562, 2016.
 141.Kakigi A, Nishimura M, Takeda T, Taguchi D, Nishioka R. Expression of aquaporin1, 3, and 4, NKCC1, and NKCC2 in the human endolymphatic sac. Auris Nasus Larynx 36: 135‐139, 2009.
 142.Kanai Y, Ohnuma N, Matsuo H. Rat atrial natriuretic polypeptide increases net water, sodium and chloride absorption across rat small intestine in vivo. Jpn J Physiol 45: 7‐13, 1987.
 143.Kaplan MR, Plotkin MD, Brown D, Hebert SC, Delpire E. Expression of the mouse Na‐K‐2Cl cotransporter, mBSC2, in the terminal IMCD, the glomerular and extraglomerular mesangium and the glomerular afferent arteriole. J Clin Invest 98: 723‐730, 1996.
 144.Kaplan MR, Plotkin MD, Lee W‐S, Xu Z‐C, Lytton J, Hebert SC. Apical localization of the Na‐K‐2Cl cotransporter, rBSC1, on rat thick ascending limbs. Kidney Int 49: 40‐47, 1996.
 145.Karimy JK, Zhang J, Carusillo Theriault B, Duran D, Gavankar C, Diluna ML, Delpire E, Medzhitov R, Gerzanich V, Simard J‐M, Kahle KT. Kinase‐dependent cerebrospinal fluid hypersecretion triggered by choroid plexus inflammation in hemorrhagic hydrocephalus. Nature Med 23: 997‐1003, 2017.
 146.Keep RF, Xiang J, Betz AL. Potassium cotransport at the rat choroid plexus. Am J Physiol (Cell Physiol) 267: C1616‐C1622, 1994.
 147.Kim DH, Ahn MD. Expression of the Na+‐K+‐2Cl(−)‐cotransporter 2 in the normal and pressure‐induced ischemic rat retina. Korean J Ophthalmol 26: 203‐211, 2012.
 148.Kim GH, Ecelbarger CA, Mitchell C, Packer RK, Wade JB, Knepper MA. Vasopressin increases Na‐K‐2Cl cotransporter expression in thick ascending limb of Henle's loop. Am J Physiol 276: F96‐F103, 1999.
 149.Kim SM, Eisner C, Faulhaber‐Walter R, Mizel D, Wall SM, Briggs JP, Schnermann J. Salt sensitivity of blood pressure in NKCC1‐deficient mice. Am J Physiol Renal Physiol 295: F1280‐F1238, 2008.
 150.Kintner DB, Look A, Shull GE, Sun D. Stimulation of astrocyte Na+/H+ exchange activity in response to in vitro ischemia depends in part on activation of ERK1/2. Am J Physiol Cell Physiol 289: C934‐C945, 2005.
 151.Kintner DB, Luo J, Gerdts J, Ballard AJ, Shull GE, Sun D. Role of Na+‐K+‐Cl− cotransport and Na+/Ca2+ exchange in mitochondrial dysfunction in astrocytes following in vitro ischemia. Am J Physiol Cell Physiol 292: C1113‐C1122, 2007.
 152.Klein JD, O'Neill WC. Effect of bradykinin on Na‐K‐2Cl cotransport and bumetanide binding in aortic endothelial cells. J Biol Chem 265: 22238‐22242, 1990.
 153.Knight DA, Holgate ST. The airway epithelium: Structural and functional properties in health and disease. Respirology 8: 432‐446, 2003.
 154.Kobayashi K, Yanagihara K, Ishiguro K, Fukuoka S. GP2/THP gene family of self‐binding, GPI‐anchored proteins forms a cluster at chromosome 7F1 region in mouse genome. Biochem Biophys Res Commun 322: 659‐664, 2004.
 155.Koltsova SV, Kotelevtsev SV, Tremblay J, Hamet P, Orlov SN. Excitation‐contraction coupling in resistance mesenteric arteries: Evidence for NKCC1‐mediated pathway. Biochem Biophys Res Commun 379: 1080‐1183, 2009.
 156.Kregenow FM. The response of duck erythrocytes to hypertonic media. Further evidence for a volume‐controlling mechanism. J Gen Physiol 58: 396‐411, 1971.
 157.Kristensen M, Juel C. Potassium‐transporting proteins in skeletal muscle: Cellular location and fibre‐type differences. Acta Physiol (Oxford) 198: 105‐123, 2010.
 158.Kusakari J, Kambayashi J, Ise I, Kawamoto K. Reduction of the endocochlear potential by the new “loop” diuretic, bumetanide. Acta Otolaryngol 86: 336‐341, 1978.
 159.Laird JM, Garcia‐Nicas E, Delpire EJ, Cervero F. Presynaptic inhibition and spinal pain processing in mice: A possible role of the NKCC1 cation‐chloride co‐transporter in hyperalgesia. Neurosci Lett 361: 200‐203, 2004.
 160.Lauf PK, McManus TJ, Haas M, Forbush B, 3rd, Duhm J, Flatman PW, Saier MH, Jr., Russell JM. Physiology and biophysics of chloride and cation cotransport across cell membranes. Fed Proc 46: 2377‐2394, 1987.
 161.Lebeau C, Hanaoka K, Moore‐Hoon ML, Guggino WB, Beauwens R, Devuyst O. Basolateral chloride transporters in autosomal dominant polycystic kidney disease. Pflugers Arch 444: 722‐731, 2002.
 162.Lee HA, Baek I, Seok YM, Yang E, Cho HM, Lee DY, Hong SH, Kim IK. Promoter hypomethylation upregulates Na+‐K+‐2Cl− cotransporter 1 in spontaneously hypertensive rats. Biochem Biophys Res Commun 396: 252‐257, 2010.
 163.Lee HA, Hong SH, Kim JW, Jang IS. Possible involvement of DNA methylation in NKCC1 gene expression during postnatal development and in response to ischemia. J Neurochem 114: 520‐529, 2010.
 164.Lee SJ, Cobb MH, Goldsmith EJ. Crystal structure of domain‐swapped STE20 OSR1 kinase domain. Protein Sci 18: 304‐313, 2009.
 165.Li J, Nielsen S, Dai Y, Lazowski KW, Christensen EI, Tabak LA, Baum BJ. Examination of rat salivary glands for the presence of the aquaporin CHIP. Pflugers Arch 428: 455‐460, 1994.
 166.Li Y, Hu J, Vita R, Sun B, Tabata H, Altman A. SPAK kinase is a substrate and target of PKCtheta in T‐cell receptor‐induced AP‐1 activation pathway. Embo J 23: 1112‐1122, 2004.
 167.Liedtke CM. Adrenergic regulation of Na‐Cl cotransport human airway epithelium. Am J Physiol (Lung Cell Moll Physiol) 257: L125‐L129, 1989.
 168.Liedtke CM. Bumetanide‐sensitive NaCl uptake in rabbit tracheal epithelial cells is stimulated by neurohormones and hypertonicity. Am J Physiol 262: L621‐L627, 1992.
 169.Lin SH, Yu IS, Jiang ST, Lin SW, Chu P, Chen A, Sytwu HK, Sohara E, Uchida S, Sasaki S, Yang SS. Impaired phosphorylation of Na+‐K+‐2Cl− cotransporter by oxidative stress‐responsive kinase‐1 deficiency manifests hypotension and Bartter‐like syndrome. Proc Natl Acad Sci U S A 108: 17538‐17543, 2011.
 170.Lindsey AE, Schneider K, Simmons DM, Baron R, Lee BS, Kopito RR. Functional expression and subcellular localization of an anion exchanger cloned from choroid plexus. Proc Natl Acad Sci USA 87: 5278‐5282, 1990.
 171.Liu W, Schreck C, Coleman RA, Wade JB, Hernandez Y, Zavilowitz B, Warth R, Kleyman TR, Satlin LM. Role of NKCC in BK channel‐mediated net K+ secretion in the CCD. Am J Physiol Renal Physiol 301: 88‐F1097, 2011.
 172.Liu Y, Chu H, Chen J, Zhou L, Chen Q, Yu Y, Wu Z, Wang S, Lai Y, Pan C, Cui Y. Age‐related change in the expression of NKCC1 in the cochlear lateral wall of C57BL/6J mice. Acta Otolaryngol 134: 1047‐1047, 2014.
 173.Lohrmann E, Greger R. Isolated perfused rabbit colon crypts: Stimulation of Cl− secretion by forskolin. Pflugers Arch 425: 373‐380, 1993.
 174.Luo J, Chen H, Kintner DB, Shull GE, Sun D. Decreased neuronal death in Na+/H+ exchanger isoform 1‐null mice after in vitro and in vivo ischemia. J Neurosci 25: 11256‐11268, 2005.
 175.Luo J, Wang Y, Chen H, Kintner DB, Cramer SW, Gerdts JK, Chen X, Shull GE, Philipson KD, Sun D. A concerted role of Na+‐K+‐Cl− cotransporter and Na+/Ca2+ exchanger in ischemic damage. J Cereb Blood Flow Metab 28: 736‐746, 2008.
 176.Lytle C. Activation of the avian erythrocyte Na‐K‐Cl cotransport protein by cell shrinkage, cAMP, fluoride, and calyculin‐A involves phosphorylation at common sites. J Biol Chem 272: 15069‐15077, 1997.
 177.Lytle C, Forbush BI. Na‐K‐Cl cotransport in the shark rectal gland. II. Regulation in isolated tubules. Am J Physiol Cell Physiol 262: C1009‐C10117, 1992.
 178.Lytle C, Forbush BI. The Na‐K‐Cl cotransport protein of shark rectal gland. I. Development of monoclonal antibodies, immunoaffinity purification, and partial biochemical characterization. J Biol Chem 267: 25428‐25437, 1992.
 179.Lytle C, Forbush BI. Regulatory phosphorylation of the secretory Na‐K‐Cl cotransporter: Modulation by cytoplasmic Cl. Am J Physiol (Cell Physiol) 270: C437‐C448, 1996.
 180.Lytle C, McManus TJ, Haas M. A model of Na‐K‐2Cl cotransport based on ordered ion binding and glide symmetry. Am J Physiol 274: C299‐C309, 1998.
 181.MacLeod RJ, Hamilton JR. Regulatory volume increase in mammalian jejunal villus cells is due to bumetanide‐sensitive NaKCl2 cotransport. Am J Physiol Gastrointest Liver Physiol 285: G665‐G674, 1990.
 182.MacLeod RJ HJ. Regulatory volume increase in mammalian jejunal villus cells is due to bumetanide‐sensitive NaKCl2 cotransport. Am J Physiol 258: G665‐G674, 1990.
 183.Markadieu N, Rios K, Spiller BW, McDonald WH, Welling PA, Delpire E. Short forms of Ste20‐related Proline/Alanine‐rich Kinase (SPAK) in the kidney are created by aspartyl aminopeptidase (Dnpep)‐mediated proteolytic cleavage. J Biol Chem 289: 29273‐29284, 2014.
 184.Martinez JR, Cassity N. Effect of transport inhibitors on secretion by perfused rat submandibular gland. Am J Physiol 245: G711‐G716, 1983.
 185.Mathog RH, Matz GJ. Ototoxic effects of ethacrynic acid. Ann Otol Rhinol Laryngol 81: 871‐875, 1972.
 186.Matthews JB, Awtrey CS, Madara JL. Microfilament‐dependent activation of Na+/K+/2Cl− cotransport by cAMP in intestinal epithelial monolayers. J Clin Invest 90: 1608‐1613, 1992.
 187.Matthews JB, Awtrey CS, Thompson R, Hung T, Tally KJ, Madara JL. Na+‐K+‐2Cl− cotransport and Cl− secretion evoked by heat‐stable enterotoxin is microfilament dependent in T84 cells. Am J Physiol 265: G370‐G378, 1993.
 188.Matthews JB, Smith JA, Hrnjez BJ. Effects of F‐actin stabilization or disassembly on epithelial Cl− secretion and Na‐K‐2Cl cotransport. Am J Physiol 272: C254‐C262, 1997.
 189.Mazzone SB, McGovern AE. Na+‐K+‐2Cl− cotransporters and Cl− channels regulate citric acid cough in guinea pigs. J Appl Physiol 101: 635‐643, 2006.
 190.McCormick JA, Ellison DH. The WNKs: Atypical protein kinases with pleiotropic actions. Physiol Rev 91: 177‐219, 2011.
 191.McCormick JA, Mutig K, Nelson JH, Saritas T, Hoorn EJ, Yang C‐L, Rogers S, Curry J, Delpire E, Bachmann S, Ellison DH. A SPAK isoform switch modulates renal salt transport and blood pressure. Cell Metabolism 14: 352‐364, 2011.
 192.McDaniel N, Lytle C. Parietal cells express high levels of Na‐K‐2Cl cotransporter on migrating into the gastric gland neck. Am J Physiol 276: G1273‐G1278, 1999.
 193.McNicholas CM, Brownm CD, Turnberg LA. Na‐K‐Cl cotransport in villus and crypt cells from rat duodenum. Am J Phsyiol 267: G1004‐G1011, 1994.
 194.Merner ND, Mercado A, Khanna AR, Hodgkinson A, Bruat V, Awadalla P, Gamba G, Rouleau GA, Kahle KT. Gain‐of‐function missense variant in SLC12A2, encoding the bumetanide‐sensitive NKCC1 cotransporter, identified in human schizophrenia. J Psychiatr Res 77: 22‐26, 2016.
 195.Meyer JW, Flagella M, Sutliff RL, Lorenz JN, Nieman ML, Weber CS, Paul RJ, Shull GE. Decreased blood pressure and vascular smooth muscle tone in mice lacking basolateral Na(+)‐K(+)‐2Cl(−) cotransporter. Am J Physiol Heart Circ Physiol 283: H1846‐H1855, 2002.
 196.Miller TB, Wilkinson HA, Rosenfield SA, Furata T. Intracranial hypertension and cerebrospinal fluid production in dogs: effects of furosemide. Exp Neurol 94: 66‐80, 1986.
 197.Moenter SM, DeFazio RA. Endogenous gamma‐aminobutyric acid can excite gonadotropin‐releasing hormone neurons. Endocrinology 146: 5374‐5379, 2005.
 198.Montesano R, Ghzili H, Carrozzino F, Rossier BC, Féraille E. cAMP‐dependent chloride secretion mediates tubule enlargement and cyst formation by cultured mammalian collecting duct cells. Am J Physiol Renal Physiol 296: F446‐F457, 2009.
 199.Montrose MH, Knoblauch C, Murer H. Separate control of regulatory volume increase and Na+‐H+ exchange by cultured renal cells. Am J Physiol 255: C76‐C85, 1988.
 200.Moore‐Hoon ML, Turner RJ. The structural unit of the secretory Na+‐K+‐2Cl− cotransporter (NKCC1) is a homodimer. Biochemistry 39: 3718‐3724, 2000.
 201.Moriguchi T, Urushiyama S, Hisamoto N, Iemura SI, Uchida S, Natsume T, Matsumoto K, Shibuya H. WNK1 regulates phosphorylation of cation‐chloride‐coupled cotransporters via the STE20‐related kinases, SPAK and OSR1. J Biol Chem 280: 42685‐42693, 2005.
 202.Mount DB, Baekgaard A, Hall AE, Plata C, Xu JZ, Beier DR, Gamba G, Hebert SC. Isoforms of the apical Na‐K‐2Cl transporter in murine thick ascending limb. I: Molecular characterization and intra‐renal localization. Am J Physiol (Renal Physiol) 276: F347‐F358, 1999.
 203.Mount DB, Mercado A, Song L, Xu J, George JAL, Delpire E, Gamba G. Cloning and characterization of KCC3 and KCC4, new members of the cation‐chloride cotransporter gene family. J Biol Chem 274: 16355‐16362, 1999.
 204.Moyer JH, Ford RV, Spurr CL. Pharmacodynamics of chlorothiazide (Diuril), an orally effective non‐mercurial diuretic agent. 95(3): 529‐531, 1957.
 205.Mutig K, Borowski T, Boldt C, Borschewski A, Paliege A, Popova E, Bader M, Bachmann S. Demonstration of the functional impact of vasopressin signaling in the thick ascending limb by a targeted transgenic rat approach. Am J Physiol Renal Physiol 311: F411‐F423, 2016.
 206.Mutig K, Paliege A, Kahl T, Jöns T, Müller‐Esterl W, Bachmann S. Vasopressin V2 receptor expression along rat, mouse, and human renal epithelia with focus on TAL. Am J Physiol Renal Physiol 293: F1166‐F1177, 2007.
 207.Mykoniatis A, Shen L, Fedor‐Chaiken M, Tang J, Tang X, Worrell RT, Delpire E, Turner JR, Matlin KS, Bouyer P, Matthews JB. Phorbol 12‐myristate 13‐acetate‐induced endocytosis of the Na‐K‐2Cl cotransporter in MDCK cells is associated with a clathrin‐dependent pathway. Am J Physiol Cell Physiol 298: C85‐C97, 2010.
 208.Nielsen S, Smith BL, Christensen EI, Agre P. Distribution of the aquaporin CHIP in secretory and resorptive epithelia and capillary endothelia. Proc Natl Acad Sci U S A 90: 7275‐7279, 1993.
 209.Nishimura M, Kakigi A, Takeda T, Takeda S, Doi K. Expression of aquaporins, vasopressin type 2 receptor, and Na+‐K+‐Cl− cotransporters in the rat endolymphatic sac. Acta Otolaryngol 129: 812‐818, 2009.
 210.Nozu K, Inagaki T, Fu XJ, Nozu Y, Kaito H, Kanda K, Sekine T, Igarashi T, Nakanishi K, Yoshikawa N, Iijima K, Matsuo M. Molecular analysis of digenic inheritance in Bartter syndrome with sensorineural deafness. J Med Genet 45: 182‐186, 2008.
 211.O'Donnell ME. Regulation of Na‐K‐2Cl cotransport in endothelial cells by atrial natriuretic factor. Am J Physiol Cell Physiol 257: C36‐C44, 1989.
 212.O'Donnell ME. Endothelial cell sodium‐potassium‐chloride cotransport. Evidence of regulation by Ca2+ and protein kinase C. J Biol Chem 266: 11559‐11566, 1991.
 213.O'Donnell ME. Role of Na‐K‐Cl cotransport in vascular endothelial cell volume regulation. Am J Physiol Cell Physiol 264: C1316‐C1326, 1993.
 214.O'Donnell ME. Blood‐brain barrier Na transporters in ischemic stroke. Adv Pharmacol 71: 113‐146, 2014.
 215.O'Donnell ME, Martinez A, Sun D. Endothelial Na‐K‐Cl cotransport regulation by tonicity and hormones: Phosphorylation of cotransport protein. Am J Physiol (Cell Physiol) 269: C1513‐C1523, 1995.
 216.O'Shaughnessy PJ, Sheffield JW. Effect of testosterone on testicular steroidogenesis in the hypogonadal (hpg) mouse. J Steroid Biochem 35: 729‐734, 1990.
 217.Oppermann M, Hansen PB, Castrop H, Schnermann J. Vasodilatation of afferent arterioles and paradoxical increase of renal vascular resistance by furosemide in mice. Am J Physiol Renal Physiol 293: F279‐F287, 2007.
 218.Oppermann M, Mizel D, Huang G, Li C, Deng C, Theilig F, Bachmann S, Briggs J, Schnermann J, Castrop H. Macula densa control of renin secretion and preglomerular resistance in mice with selective deletion of the B isoform of the Na,K,2Cl co‐transporter. J Am Soc Nephrol 17: 2143‐2152, 2006.
 219.Oppermann M, Mizel D, Kim SM, Chen L, Faulhaber‐Walter R, Huang Y, Li C, Deng C, Briggs J, Schnermann J, Castrop H. Renal function in mice with targeted disruption of the A isoform of the Na‐K‐2Cl co‐transporter. J Am Soc Nephrol 18: 440‐448, 2007.
 220.Orlov SN. Decreased Na+,K+,2Cl− cotransport and salt retention in Blacks: A provocative hypothesis. J Hypertens 23: 1929‐1930, 2005.
 221.Orlov SN, Tremblay J, Hamet P. Altered beta‐adrenergic regulation of Na‐K‐Cl cotransport in cultured smooth muscle cells from the aorta of spontaneously hypertensive rats. Role of the cytoskeleton network. Am J Hypertens 8: 739‐747, 1995.
 222.Ortiz PA. cAMP increases surface expression of NKCC2 in rat thick ascending limbs: Role of VAMP. Am J Physiol Renal Physiol 290: F608‐F616, 2006.
 223.Owens DF, Boyce LH, Davis MBE, Kriegstein AR. Excitatory GABA responses in embryonic and neonatal cortical slices demonstrated by gramicidin perforated‐patch recordings and calcium imaging. J Neurosci 16: 6414‐6423, 1996.
 224.Ozawa T, Saito Y, Nishiyama A. Mechanism of uphill chloride transport of the mouse lacrimal acinar cells: Studies with Cl−‐sensitive microelectrode. Pflugers Arch 412: 509‐515, 1988.
 225.Pace AJ, Lee E, Athirakul K, Coffman TM, O'Brien DA, Koller BH. Failure of spermatogenesis in mouse lines deficient in the Na+‐K+‐2Cl− cotransporter. J Clin Invest 105: 441‐450, 2000.
 226.Pace AJ, Madden VJ, Henson OWJ, Koller BH, Henson MM. Ultrastructure of the inner ear of NKCC1‐deficient mice. Hear Res 156: 17‐30, 2001.
 227.Painter MJ, Scher MS, Stein AD, Armatti S, Wang Z, Gardiner JC, Paneth N, Minnigh B, Alvin J. Phenobarbital compared with phenytoin for the treatment of neonatal seizures. N Engl J Med 341: 485‐489, 1999.
 228.Palfrey HC, Pewitt EB. The ATP and Mg2+ dependence of Na(+)‐K(+)‐2Cl− cotransport reflects a requirement for protein phosphorylation: studies using calyculin A. Pflugers Archiv 425: 321‐328, 1993.
 229.Palfrey HC, Rao MC. Na/K/Cl co‐transport and its regulation. J Exp Biol 106: 43‐54, 1983.
 230.Park HJ, Curry JN, McCormick JA. Regulation of NKCC2 activity by inhibitory SPAK isoforms: KS‐SPAK is a more potent inhibitor than SPAK2. Am J Physiol Renal Physiol 305: F1687‐F1696, 2013.
 231.Payne JA, Forbush BI. Alternatively spliced isoforms of the putative renal Na‐K‐Cl cotransporter are differentially distributed within the rabbit kidney. Proc Natl Acad Sci U S A 91: 4544‐4548, 1994.
 232.Payne JA, Stevenson TJ, Donaldson LF. Molecular characterization of a putative K‐Cl cotransporter in rat brain. A neuronal‐specific isoform. J Biol Chem 271: 16245‐16252, 1996.
 233.Payne JA, Xu J‐C, Haas M, Lytle CY, Ward D, Forbush BI. Primary structure, functional expression, and chromosome localization of the bumetanide sensitive Na‐K‐Cl cotransporter in human colon. J Biol Chem 270: 17977‐17985, 1995.
 234.Pech V, Thumova M, Kim YH, Agazatian D, Hummler E, Rossier BC, Weinstein AM, Nanami M, Wall SM. ENaC inhibition stimulates Cl‐ secretion in the mouse cortical collecting duct through an NKCC1‐dependent mechanism. Am J Physiol Renal Physiol 303: F45‐F55, 2012.
 235.Petersen OH. Calcium‐activated potassium channels and fluid secretion by exocrine glands. Am J Physiol 251: G1‐G13, 1986.
 236.Pewitt EB, Hegde RS, Haas M, Palfrey HC. The regulation of Na/K/2Cl cotransport and bumetanide binding in avian erythrocytes by protein phosphorylation and dephosphorylation. Effects of kinase inhibitors and okadaic acid. J Biol Chem 265: 20747‐20756, 1990.
 237.Piala AT, Moon TM, Akella R, He H, Cobb MH, Goldsmith EJ. Chloride sensing by WNK1 involves inhibition of autophosphorylation. Sci Signal 7: ra41, 2014.
 238.Piechotta K, Garbarini NJ, England R, Delpire E. Characterization of the interaction of the stress kinase SPAK with the Na+‐K+‐2Cl− cotransporter in the nervous system: Evidence for a scaffolding role of the kinase. J Biol Chem 278: 52848‐52856, 2003.
 239.Piechotta K, Lu J, Delpire E. Cation‐chloride cotransporters interact with the stress‐related kinases SPAK and OSR1. J Biol Chem 277: 50812‐50819, 2002.
 240.Pieraut S, Matha V, Sar C, Hubert T, Méchaly I, Hilaire C, Mersel M, Delpire E, Valmier J, Scamps F. NKCC1 phosphorylation stimulates neurite growth of injured adult sensory neurons. J Neurosci 27: 6751‐6759, 2007.
 241.Plata C, Meade P, Vazquez N, Hebert SC, Gamba G. Functional properties of the apical Na+‐K+‐2Cl− cotransporter isoforms. J Biol Chem 277: 11004‐11012, 2002.
 242.Plato CF, Stoos BA, Wang D, Garvin JL. Endogenous nitric oxide inhibits chloride transport in the thick ascending limb. Am J Physiol Renal Physiol 276: F159‐F163, 1999.
 243.Plotkin MD, Kaplan MR, Peterson LN, Gullans SR, Hebert SC, Delpire E. Expression of the Na+‐K+‐2Cl− cotransporter BSC2 in the nervous system. Am J Physiol Cell Physiol 272: C173‐C183, 1997.
 244.Plotkin MD, Kaplan MR, Verlander JW, Lee W‐S, Brown D, Poch E, Gullans SR, Hebert SC. Localization of the thiazide sensitive Na‐Cl cotransporter, rTSC, in the rat kidney. Kidney Int 50: 174‐183, 1996.
 245.Plotkin MD, Snyder EY, Hebert SC, Delpire E. Expression of the Na‐K‐2Cl cotransporter is developmentally regulated in postnatal rat brains: A possible mechanism underlying GABA's excitatory role in immature brain. J Neurobiol 33: 781‐795, 1997.
 246.Ponce‐Coria J, Gagnon KB, Delpire E. Calcium‐binding protein 39 facilitates molecular interaction between Ste20p proline alanine‐rich kinase and oxidative stress response 1 monomers. Am J Physiol Cell Physiol 303: C1198‐C1205, 2012.
 247.Pondugula SR, Kampalli SB, Wu T, De Lisle RC, Raveendran NN, Harbidge DG, Marcus DC. cAMP‐stimulated Cl‐ secretion is increased by glucocorticoids and inhibited by bumetanide in semicircular canal duct epithelium. BMC Physiol 13: 6, 2013.
 248.Preston GM, Agre P. Isolation of the cDNA for erythrocyte integral membrane protein of 28 kilodaltons: Member of an ancient channel family. Proc Natl Acad Sci U S A 88: 11110‐11114, 1991.
 249.Quick CA, Hoppe W. Permanent deafness associated with furosemide administration. Ann Otol Rhinol Laryngol 84: 94‐101, 1975.
 250.Quinton PM. What is good about cystic fibrosis? Curr Biol 4: 742‐743, 1994.
 251.Qusous A, Geewan CS, Greenwell P, Kerrigan MJ. siRNA‐mediated inhibition of Na(+)‐K(+)‐2Cl‐ cotransporter (NKCC1) and regulatory volume increase in the chondrocyte cell line C‐20/A4. J Membrane Biol 243: 25‐34, 2011.
 252.Raat NJH, Delpire E, van Os CH, Bindels RJM. Culturing induced expression of basolateral Na+‐K+‐2Cl− cotransporter BSC2 in proximal tubule, aortic endothelium, and vascular smooth muscle. Pflugers Arch 431: 458‐460, 1996.
 253.Raina S, Preston GM, Guggino WB, Agre P. Molecular cloning and characterization of an aquaporin cDNA from salivary, lacrimal, and respiratory tissues. J Biol Chem 270: 1908‐1912, 1995.
 254.Randall J, Thorne T, Delpire E. Partial cloning and characterization of Slc12a2: the gene encoding the secretory Na+‐K+‐2Cl− cotransporter. Am J Physiol (Cell Physiol) 273: C1267‐C1277, 1997.
 255.Rees H, Sluka KA, Lu Y, Westlund KN, Willis WD. Dorsal root reflexes in articular afferents occur bilaterally in a chronic model of arthritis in rats. J Neurophysiol 76: 4190‐4193, 1996.
 256.Reinalter SC, Jeck N, Brochhausen C, Watzer B, Nüsing RM, Seyberth HW, Kömhoff M. Role of cyclooxygenase‐2 in hyperprostaglandin E syndrome/antenatal Bartter syndrome. Kidney Int 62: 253‐260, 2002.
 257.Reynolds A, Parris A, Evans LA, Lindqvist S, Sharp P, Lewis M, Tighe R, Williams MR. Dynamic and differential regulation of NKCC1 by calcium and cAMP in the native human colonic epithelium. J Physiol 582: 507‐524, 2007.
 258.Robertson M, Foskett JK. Membrane crosstalk in secretory epithelial cells mediated by intracellular chloride concentration. Jpn J Physiol 44: S309‐S315, 1994.
 259.Robertson MA, Foskett JK. Na+ transport pathways in secretory acinar cells: Membrane cross talk mediated by [Cl−]i. Am J Physiol (Cell Physiol) 267: C146,C156, 1994.
 260.Rozengurt N, Lopez I, Chiu CS, Kofuji P, Lester HA, Neusch C. Time course of inner ear degeneration and deafness in mice lacking the Kir4.1 potassium channel subunit. Hear Res 177: 71‐80, 2003.
 261.Russell JM. Sodium‐potassium‐chloride cotransport. Physiol Rev 80: 211‐276, 2000.
 262.Saga K, Sato K. Electron probe X‐ray microanalysis of cellular ions in the eccrine secretory coil cells during methacholine stimulation. J Membrane Biol 107: 13‐24, 1989.
 263.Sahi J, Goldstein JL, Layden TJ, Rao MC. Cyclic AMP‐ and phorbol ester‐regulated Cl− permeabilities in primary cultures of human and rabbit colonocytes. Am J Phsyiol 266: G846‐G855, 1994.
 264.Sang Y, Teresa Ortega M, Rune K, Xiau W, Zhang G, Soulages JL, Lushington GH, Fang J, Williams TD, Blecha F, Melgarejo T. Canine cathelicidin (K9CATH): Gene cloning, expression, and biochemical activity of a novel pro‐myeloid antimicrobial peptide. Dev Comp Immunol 31: 1278‐1296, 2007.
 265.Schulze‐Bahr E, Wang Q, Wedekind H, Haverkamp W, Chen Q, Sun Y, Rubie C, Hordt M, Towbin JA, Borggrefe M, Assmann G, Qu X, Somberg JC, Breithardt G, Oberti C, Funke H. KCNE1 mutations causes Jervell and Lange‐Nielsen syndrome. Nature Genet 17: 267‐268, 1997.
 266.Seidler U, Rottinghaus I, Hillesheim J, Chen M, Riederer B, Krabbenhöft A, Engelhardt R, Wiemann M, Wang Z, Barone S, Manns MP, Soleimani M. Sodium and chloride absorptive defects in the small intestine in Slc26a6 null mice. Pflugers Arch 455: 757‐766, 2008.
 267.Shumaker H, Soleimani M. CFTR upregulates the expression of the basolateral Na(+)‐K(+)‐2Cl(−) cotransporter in cultured pancreatic duct cells. Am J Physiol 277: C1100‐C1110, 1999.
 268.Silen W, Harper HA, Mawdsley DL, Weirich WL. Effect of antibacterial agents on ammonia production within the intestine. Proc Soc Exp Biol Med 88: 138‐140, 1955.
 269.Simard CF, Brunet GM, Daigle ND, Montminy V, Caron L, Isenring P. Self‐interacting domains in the C terminus of a cation‐Cl− cotransporter described for the first time. J Biol Chem 279: 40769‐40777, 2004.
 270.Simon DB, Bindra RS, Mansfield TA, Nelson‐Williams C, Mendonca E, Stone R, Schurman S, Nayir A, Alpay H, Bakkaloglu A, Rodriguez‐Soriano J, Morales JM, Sanjad SA, Taylor CM, Pilz D, Brem A, Trachtman H, Griswold W, Richard GA, John E, Lifton RP. Mutations in the chloride channel gene, CLCNKB, cause Bartter's syndrome type III. Nat Genet 17: 171‐178, 1997.
 271.Simon DB, Karet FE, Hamdan JM, Di Pietro A, Sanjad SA, Lifton RP. Bartter's syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na‐K‐2Cl cotransporter NKCC2. Nature Gen 13: 183‐188, 1996.
 272.Simon DB, Karet FE, Rodriquez‐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.
 273.Sluka KA, Rees H, Westlund KN, Willis WD. Fiber types contributing to dorsal root reflexes induced by joint inflammation in cats and monkeys. J Neurophysiol 74: 981‐989, 1995.
 274.Smith L, Smallwood N, Altman A, Liedtke CM. PKCdelta acts upstream of SPAK in the activation of NKCC1 by hyperosmotic stress in human airway epithelial cells. J Biol Chem 283: 22147‐22156, 2008.
 275.Smith QR, Woodbury DM, Johanson CE. Uptake of 36Cl and 22Na by the choroid plexus‐cerebrospinal fluid system: Evidence for active chloride transport by the choroidal epithelium. J Neurochem 37: 107‐116, 1980.
 276.Sollini M, Frieden M, Bény J‐L. Charybdotoxin‐sensitive small conductance KCa channel activated by bradykinin and substance P in endothelial cells. Br J Pharmacol 136: 1201‐1209, 2002.
 277.Solymosi EA, Kaestle‐Gembardt SM, Vadász I, Wang L, Neye N, Chupin CJ, Rozowsky S, Ruehl R, Tabuchi A, Schulz H, Kapus A, Morty RE, Kuebler WM. Chloride transport‐driven alveolar fluid secretion is a major contributor to cardiogenic lung edema. Proc Natl Acad Sci U S A 110: E2308‐E2316, 2013.
 278.Soybel DI, Davis MB, Cheung LY. Characteristics of basolateral Cl‐ transport by gastric surface epithelium in Necturus antral mucosa. Am J Physiol 264: G910‐G920, 1993.
 279.Soybel DI, Gullans SR, Maxwell F, Delpire E. Role of basolateral Na‐K‐Cl cotransport in HCl secretion by Amphibian gastric mucosa. Am J Physiol Cell Physiol 269: C242‐C249, 1995.
 280.Stec DE, Drummond HA, Gousette MU, Storm MV, Abraham NG, Csongradi E. Expression of heme oxygenase‐1 in thick ascending loop of henle attenuates angiotensin II‐dependent hypertension. J Am Soc Nephrol 23: 834‐841, 2012.
 281.Stokes JB, Lee I, D'Amico M. Sodium chloride absorption by the urinary bladder of the winter flounder: A thiazide‐sensitive, electrically neutral transport system. J Clin Invest 74: 7‐16, 1984.
 282.Stokes W, Nunn LCA. A new effective diuretic—LASIX. Br Med J 2: 910‐914, 1964.
 283.Su G, Kintner DB, Flagella M, Shull GE, Sun D. Astrocytes from Na(+)‐K(+)‐Cl(−) cotransporter‐null mice exhibit absence of swelling and decrease in EAA release. Am J Physiol Cell Physiol 282: C1147‐C1160, 2002.
 284.Su G, Kintner DB, Sun D. Contribution of Na(+)‐K(+)‐Cl(−) cotransporter to high‐[K(+)](o)‐induced swelling and EAA release in astrocytes. Am J Physiol Cell Physiol 282: C1136‐C1146, 2002.
 285.Sugita M, Hirono C, Shiba Y. Gramicidin‐perforated patch recording revealed the oscillatory nature of secretory Cl− movements in salivary acinar cells. J Gen Physiol 124: 59‐69, 2004.
 286.Sun X, Sui H, Fisher JT, Yan Z, Liu X, Cho HJ, Joo NS, Zhang Y, Zhou W, Yi Y, Kinyon JM, Lei‐Butters DC, Griffin MA, Naumann P, Luo M, Ascher J, Wang K, Frana T, Wine JJ, Meyerholz DK, Engelhardt JF. Disease phenotype of a ferret CFTR‐knockout model of cystic fibrosis. J Clin Invest 120: 3149‐3160, 2010.
 287.Sung K‐W, Kirby M, McDonald MP, Lovinger DM, Delpire E. Abnormal GABAA‐receptor mediated currents in dorsal root ganglion neurons isolated from Na‐K‐2Cl cotransporter null mice. J Neurosci 20: 7531‐7538, 2000.
 288.Susa K, Kita S, Iwamoto T, Yang SS, Lin SH, Ohta A, Sohara E, Rai T, Sasaki S, Alessi DR, Uchida S. Effect of heterozygous deletion of WNK1 on the WNK‐OSR1/SPAK‐NCC/NKCC1/NKCC2 signal cascade in the kidney and blood vessels. Clin Exp Nephrol 16: 530‐538, 2012.
 289.Takahashi N, Chernavvsky DR, Gomez RA, Igarashi P, Gitelman HJ, Smithies O. Uncompensated polyuria in a mouse model of Bartter's syndrome. Proc Natl Acad Sci U S A 97: 5434‐5439, 2000.
 290.Takiguchi Y, Sun GW, Ogawa K, Matsunaga T. Long‐lasting changes in the cochlear K+ recycling structures after acute energy failure. Neurosci Res 77: 33‐41, 2013.
 291.Thomson RB, Mentone S, Kim R, Earle K, Delpire E, Somlo S, Aronson PS. Histopathological analysis of renal cystic epithelia in the Pkd2WS25/‐ mouse model of ADPKD. Am J Physiol Renal Physiol 285: F870‐F880, 2003.
 292.Toka HR, Al‐Romaih K, Koshy JM, DiBartolo Sr, Kos CH, Quinn SJ, Curhan GC MD, Brown EM, Pollak MR. Deficiency of the calcium‐sensing receptor in the kidney causes parathyroid hormone‐independent hypocalciuria. J Am Soc Nephrol 23: 1879‐1890, 2012.
 293.Topper JN, Wasserman SM, Anderson KR, Cai J, Falb D, Gimbrone MAJ. Expression of the bumetanide‐sensitive Na‐K‐Cl cotransporter BSC2 is differentially regulated by fluid mechanical and inflammatory cytokine stimuli in vascular endothelium. J Clin Invest 99: 2941‐2949, 1997.
 294.Torchia J, Lytle C, Pon DJ, Forbush BI, Sen AK. The Na‐K‐Cl cotransporter of avian salt gland. Phosphorylation in response to cAMP‐dependent and calcium‐dependent secretogogues. J Biol Chem 267: 25444‐25450, 1992.
 295.Trudu M, Janas S, Lanzani C, Debaix H, Schaeffer C, Ikehata M, Citterio L, Demaretz S, Trevisani F, Ristagno G, Glaudemans B, Laghmani K, Dell'Antonio G, team SKPoGiHS, Loffing J, Rastaldi MP, Manunta P, Devuyst O, Rampoldi L. Common noncoding UMOD gene variants induce salt‐sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Medicine 19: 1655‐1660, 2013.
 296.Turner RJ, George JN. Cl−‐HCO3− exchange is present with Na+‐K+‐Cl− cotransport in rabbit parotid acinar basolateral membranes. Am J Physiol Cell Physiol 254: C391‐C396, 1988.
 297.Tyzio R, Nardou R, Ferrari DC, Tsintsadze T, Shahrokhi A, Eftekhari S, Khalilov I, Tsintsadze V, Brouchoud C, Chazal G, Lemonnier E, Lozovaya N, Burnashev N, Ben‐Ari Y. Oxytocin‐mediated GABA inhibition during delivery attenuates autism pathogenesis in rodent offspring. Science 343: 675‐679, 2014.
 298.Ueberschär S, Bakker‐Grunwald T. Effects of ATP and cAMP on the (Na++K++2Cl−)‐cotransport system in turkey erythrocytes. Biochim Biophys Acta 818: 260‐266, 1985.
 299.van Emst MG, Klarenbeek S, Schot A, Plomp JJ, Doornenbal A, Everts ME. Reducing chloride conductance prevents hyperkalaemia‐induced loss of twitch force in rat slow‐twitch muscle. J Physiol 561: 169‐181, 2004.
 300.Vigne P, Lopez Farre A, Frelin C. Na(+)‐K(+)‐Cl− cotransporter of brain capillary endothelial cells. Properties and regulation by endothelins, hyperosmolar solutions, calyculin A, and interleukin‐1. J Biol Chem 269: 19925‐19930, 1994.
 301.Villa F, Goebel J, Rafiqi FH, Deak M, Thastrup J, Alessi DR, van Aalten DMF. Structural insights into the recognition of substrates and activators by the OSR1 kinase. EMBO Rep 8: 839‐845, 2007.
 302.Vitari AC, Thastrup J, Rafiqi FH, Deak M, Morrice NA, Karlsson HK, Alessi DR. Functional interactions of the SPAK/OSR1 kinases with their upstream activator WNK1 and downstream substrate NKCC1. Biochem J 397: 223‐231, 2006.
 303.Vogh BP, Doyle AS. The effect of carbonic anhydrase inhibitors and other drugs on sodium entry to cerebrospinal fluid. J Pharmacol Exp Ther 217: 51‐56, 1981.
 304.Vogh BP, Langham MRJ. The effect of furosemide and bumetanide on cerebrospinal fluid formation. Brain Res 221: 171‐183, 1981.
 305.Wakamatsu S, Nonoguchi H, Ikebe M, Machida K, Izumi Y, Memetimin H, Nakayama Y, Nakanishi T, Kohda Y, Tomita K. Vasopressin and hyperosmolality regulate NKCC1 expression in rat OMCD. Hypertens Res 32: 481‐487, 2009.
 306.Walker NM, Flagella M, Gawenis LR, Shull GE, Clarke LL. An alternate pathway of cAMP‐stimulated Cl secretion across the NKCC1‐null murine duodenum. Gastroenterology 123: 531‐541, 2002.
 307.Wall SM, Fischer MP. Contribution of the Na(+)‐K(+)‐2Cl(−) cotransporter (NKCC1) to transepithelial transport of H(+), NH(4)(+), K(+), and Na(+) in rat outer medullary collecting duct. J Am Soc Nephrol 13: 827‐835, 2002.
 308.Wall SM, Fischer MP, Mehta P, Hassell KA, Park SJ. Contribution of the Na+‐K+‐2Cl− cotransporter NKCC1 to Cl− secretion in rat OMCD. Am J Physiol Renal Physiol 280: F913‐F921, 2001.
 309.Wall SM, Knepper MA, Hassell KA, Fischer MP, Shodeinde A, Shin W, Pham TD, Meyer JW, Lorenz JN, Beierwaltes WH, Dietz JR, Shull GE, Kim YH. Hypotension in NKCC1 null mice: Role of the kidneys. Am J Physiol Renal Physiol 290: F409‐F416, 2006.
 310.Wallace BK, Foroutan S, O'Donnell ME. Ischemia‐induced stimulation of Na‐K‐Cl cotransport in cerebral microvascular endothelial cells involves AMP kinase. Am J Physiol Cell Physiol 301: C316‐C326, 2011.
 311.Wallace DP, Christensen M, Reif G, Belibi F, Thrasher B, Herrell D, Grantham JJ. Electrolyte and fluid secretion by cultured human inner medullary collecting duct cells. Am J Physiol Renal Physiol 383: F1337‐F1350, 2002.
 312.Wallace DP, Grantham JJ, Sullivan LP. Chloride and fluid secretion by cultured human polycystic kidney cells. Kidney Int 50: 1327‐1336, 1996.
 313.Walz W, Hertz L. Intense furosemide‐sensitive potassium accumulation in astrocytes in the presence of pathologically high extracellular potassium levels. J Cereb Blood Flow Metab 4: 301‐304, 1984.
 314.Walz W, Hinks EC. Carrier‐mediated KCl accumulation accompanied by water movements is involved in the control of physiological K+ levels by astrocytes. Brain Res 343: 44‐51, 1985.
 315.Wang X, Breaks J, Loutzenhiser K, Loutzenhiser R. Effects of inhibition of the Na+/K+/2Cl− cotransporter on myogenic and angiotensin II responses of the rat afferent arteriole. Am J Physiol Renal Physiol 292: F999‐F1006, 2007.
 316.Warmuth S, Zimmermann I, Dutzler R. X‐ray structure of the C‐terminal domain of a prokaryotic cation‐chloride cotransporter. Structure 17: 538‐546, 2009.
 317.Whisenant N, Zhang B‐X, Khademazad M, Loessberg P, Muallem S. Regulation of Na‐K‐2Cl cotransport in osteoblasts. Am J Physiol 261: C433‐C440, 1991.
 318.Wiemuth D, Ke Y, Rohlfs M, McDonald FJ. Epithelial sodium channel (ENaC) is multi‐ubiquitinated at the cell surface. Biochem J 405: 147‐155, 2007.
 319.Willis EF, Clough GF, Church MK. Investigation into the mechanisms by which nedocromil sodium, frusemide and bumetanide inhibit the histamine‐induced itch and flare response in human skin in vivo. Clin Exp Allergy 34: 450‐455, 2004.
 320.Willis WD. John Eccles' studies of spinal cord presynaptic inhibition. Prog Neurobiol 78: 189‐214, 2006.
 321.Wilms V, Köppl C, Söffgen C, Hartmann AM, Nothwang HG. Molecular bases of K+ secretory cells in the inner ear: Shared and distinct features between birds and mammals. Sci Rep 6: 34203, 2016.
 322.Wong FH, Chen JS, Reddy V, Day JL, Shlykov MA, Wakabayashi ST, Saier MHJ. The amino acid‐polyamine‐organocation superfamily. J Mol Microbiol Biotechnol 22: 105‐113, 2012.
 323.Worrell RT, Merk L, Matthews JB. Ammonium transport in the colonic crypt cell line, T84: Role for Rhesus glycoproteins and NKCC1. Am J Physiol Gastrointest Liver Physiol 294: G429‐G440, 2008.
 324.Wouters M, De Laet A, Ver Donck L, Delpire E, van Bogaert PP, Timmermans JP, de Kerchove d'Exaerde A, Smans K, Vanderwinden JM. Subtractive hybridization unravels a role for the ion co‐transporter NKCC1 in the murine intestinal pacemaker. Am J Physiol Gastrointest Liver Physiol 290: G1219‐G1227, 2006.
 325.Wright EM. Mechanisms of ion transport across choroid plexus. J Physiol (Lond) 226: 545‐571, 1972.
 326.Wright EM. Transport processes in the formation of the cerebrospinal fluid. Rev Physiol Biochem Pharmacol 83: 1‐34, 1978.
 327.Wu Q, Delpire E, Hebert SC, Strange K. Functional demonstration of Na‐K‐2Cl cotransporter activity in isolated, polarized choroid plexus cells. Am J Physiol Cell Physiol 275: C1565‐C1572, 1998.
 328.Xu J‐C, Lytle C, Zhu TT, Payne JA, Benz EJ, Forbush BI. Molecular cloning and functional expression of the bumetanide‐sensitive Na‐K‐2Cl cotransporter. Proc Natl Acad Sci USA 91: 2201‐2205, 1994.
 329.Xue H, Tang X. Effect of vasopressin on Na(+)‐K(+)‐2Cl(−) cotransporter (NKCC) and the signaling mechanisms on the murine late distal colon. Eur J Pharmacol 771: 241‐246, 2016.
 330.Yamashita A, Singh SK, Kawate T, Jin Y, Gouaux E. Crystal structure of a bacterial homologue of Na+/Cl−‐dependent neurotransmitter transporters. Nature 437: 215‐223, 2005.
 331.Yan Y, Dempsey RY, Flemmer A, Forbush B, Sun D. Inhibition of Na(+)‐K(+)‐Cl(−) cotransporter during focal cerebral ischemia decreases edema and neuronal damage. Brain Res 961: 22‐31, 2003.
 332.Yang SS, Lo YF, Wu CC, Lin SW, Yeh CJ, Chu P, Sytwu HK, Uchida S, Sasaki S, Lin SH. SPAK‐knockout mice manifest Gitelman syndrome and impaired vasoconstriction. J Am Soc Nephrol 21: 1868‐1877, 2010.
 333.Yang T, Huang YG, Singh I, Schnermann J, Briggs JP. Localization of bumetanide‐ and thiazide‐sensitive Na‐K‐Cl cotransporters along the rat nephron. Am J Physiol (Renal Physiol) 271: F931‐F939, 1996.
 334.Yang T, Park JM, Arend L, Huang Y, Topaloglu R, Pasumarthy A, Praetorius H, Spring K, Briggs JP, Schnermann J. Low chloride stimulation of prostaglandin E2 release and cyclooxygenase‐2 expression in a mouse macula densa cell line. J Biol Chem 275: 37922‐37929, 2000.
 335.Zanou N, Mondin L, Fuster C, Seghers F, Dufour I, de Clippele M, Schakman O, Tajeddine N, Iwata Y, Wakabayashi S, Voets T, Allard B, Gailly P. Osmosensation in TRPV2 dominant negative expressing skeletal muscle fibres. J Physiol 593: 3849‐3863, 2015.
 336.Zeniya M, Sohara E, Kita S, Iwamoto T, Susa K, Mori T, Oi K, Chiga M, Takahashi D, Yang SS, Lin SH, Rai T, Sasaki S, Uchida S. Dietary salt intake regulates WNK3‐SPAK‐NKCC1 phosphorylation cascade in mouse aorta through angiotensin II. Hypertension 62: 872‐878, 2013.
 337.Zhao H, Nepomuceno R, Gao X, Foley LM, Wang S, Begum G, Zhu W, Pigott VM, Falgoust LM, Kahle KT, Yang SS, Lin SH, Alper SL, Hitchens TK, Hu S, Zhang Z, Sun D. Deletion of the WNK3‐SPAK kinase complex in mice improves radiographic and clinical outcomes in malignant cerebral edema after ischemic stroke. J Cereb Blood Flow Metab 37: 550‐563, 2017.
 338.Zhu MH, Sung TS, Kurahashi M, O'Kane LE, O'Driscoll K, Koh SD, Sanders KM. Na+‐K+‐Cl− cotransporter (NKCC) maintains the chloride gradient to sustain pacemaker activity in interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 311: G1037‐G1046, 2016.

Further Reading

Russell JM. Sodium-Potassium-Chloride Cotransport. Physiol Rev 80: 211-276, 2000. (261) This is the most comprehensive review ever written on Na+-K+-2Cl- cotransport. It contains a trove of information on the biophysical properties of the cotransporter.

Gagnon KB, Delpire E. Molecular physiology of SPAK and OSR1: Two Ste20-related protein kinases regulating ion transport. Physiol Rev 92: 1577-1617, 2012. (87)

McCormick JA, Ellison DH. The WNKs: atypical protein kinases with pleiotropic actions. Physiol Rev 91: 177-219, 2011. (190) These two reviews summarize most of what is known on the regulatory kinases that act upstream of the Na+-K+-2Cl- cotransporter.

Hoffmann EK, Lambert IH, Pedersen SF. Physiology of cell volume regulation in vertebrates. Physiol Rev 89: 193-277, 2009. (122) This review article is written by the most proficient scientist in the field of cell volume regulation.

Ares GR, Caceres PS, Ortiz PA. Molecular regulation of NKCC2 in the thick ascending limb. Am J Physiol Renal Physiol 301: F1143-1159, 2011 (14) This is a recent review article summarizing most of what is known about the regulation of NKCC2.

Alvarez-Leefmans FJ, Delpire E. Physiology and Pathology of Chloride Transporters and Channels in the Nervous System: From Molecules to Diseases: Academic Press, 2009. (8) This book was contributed by many leaders in the field of Cl- transport and homeostasis. While covering on all mechanisms transporting Cl- and mostly focusing on the nervous system, the book contains several chapters covering the function of the Na+-K+-2Cl- cotransporter, including its function in many epithelial cells.

Teaching Material

E. Delpire, K. B. Gagnon. Na+-K+-2Cl Cotransporter (NKCC) Physiological Function in Nonpolarized Cells and Transporting Epithelia. Compr Physiol. 8: 2018, 871-901.

Didactic Synopsis

Major Teaching Points:

  • Na+-K+-2Cl cotransporters are secondary active transport mechanisms.
  • NKCC1 and NKCC2 are phosphoproteins, active when phosphorylated and inactive when dephosphorylated.
  • Transport through NKCC is most active when intracellular Cl is low.
  • NKCC1 is a mechanism of cell volume regulation that facilitates regulatory volume increase after cell shrinkage.
  • NKCC1 is a basolateral cotransporter in most epithelial cells. It participates in Cl based fluid secretion in airway, gastric and intestinal epithelium, as well as sweat, lacrimal, and salivary glands.
  • NKCC1 also participates in the formation of the K+-rich endolymph in inner ear.
  • NKCC1 is highly expressed on the apical membrane of choroid plexus epithelial cells. Its direction of transport might be reversed due to high intracellular Cl concentration maintained by carbonic anhydrase and a Cl/HCO3 exchanger.
  • In nonepithelial cells, the cotransporter participates in Cl homeostasis impacting for instance smooth muscle cell contraction and synaptic transmission.
  • NKCC2 is an apical cotransporter. It lacks a COOH-terminal motif that sends NKCC1 to the basolateral membrane of epithelial cells.
  • NKCC2 participates in NaCl reabsorption in the thick ascending limb of Henle and fluid and Cl sensing in the macula densa.

Didactic Legends

The figures—in a freely downloadable PowerPoint format—can be found on the Images tab along with the formal legends published in the article. The following legends to the same figures are written to be useful for teaching.

Figure 1. Teaching points: The SCL12A family of transporters is relatively small as it only comprises of nine genes. Two genes (CCC8 and CCC9) encode orphan proteins or proteins to which function has yet to be described. Based on the length of the lines separating the members of a subfamily, there is less genetic diversity in the four K+-Cl cotransporters than the three Na+-(K+)-Cl cotransporters. Based on the chemical gradients of transporter ions, the Na+-dependent cotransporters carry ions into the cell (out to in), whereas the Na+-independent cotransporters carrion ions out of the cell. Note that transport direction reflects net transport only, as transporters are able to move ions in both directions.

Figure 2. Teaching points:The Figure is a simulated representation of the transmembrane domains of NKCC1. It is modeled based on known crystal structure of distantly related transporters, as there is no crystal structure of NKCC1 available. Hydrophobic alpha-helices are represented as tubes whereas hydrophilic linkers are represented as threads. Note the compactness of the structure and the parallel orientation of several pairs of transmembrane domains. Panel B adds the volume occupied by specific amino acids involved in ion affinity. Notice that the amino acid residues turn around the axis of the transmembrane domain—which reflects the structure of the alpha helix with a 3.6 residues per turn.

Figure 3. Teaching points:The two genes (SLC12A1 and SLC12A2) have a very similar exon/intron structure, which is expected as the SLC12A1 gene arose from gene duplication. Overtime during evolution some changes occurred, but they are minimal compared to the overall gene structure. Most striking differences are the size of the introns, the sizes of the first and last exons, and the presence of some gene-specific exons (light blue). Alternatively spliced exons are the three exons 4 in SLC12A1—labeled A, B, and F, and exon 21 in SLC12A2.

Figure 4. Teaching points: A variety of transporters are expressed on the apical and basolateral membranes of epithelial cells. The figure presents three models. In (A), we represent an intestinal surface cell, which function is to reabsorb Na+. The transcellular pathway for Na+ movement consists of apical Na+ channels and the basolateral Na+/K+ pump. Note the direction of the arrows, pointing to the movement of Na+ from the luminal side to the blood side (i.e., reabsorption). In (B), we represent a crypt cell, or a cell that lives deeper in an intestinal crypt, in Cl secreting mode (larger cell on left) or K+ secreting mode (narrower cell on the right). Note that NKCC1 and the Na+/K+ pump on the basolateral side are active in the two models. Difference between the two models include activation of apical Cl channels and basolateral K+ conductance under Clsecretion mode; while activation of apical K+ channels and basolateral Cl conductance under K+ secretion mode. Bicarbonate can be secreted along with Cl through a basolateral cotransport mechanism and an apical exchanger mechanism. Also note that several receptors are located on the basolateral membrane (in red). Through activation of adenylcyclase (AC) and the production of cAMP, they stimulate apical Cl channels.

Figure 5. Teaching points:The choroid plexus constitutes a unique epithelium as both the Na+/K+ pump and NKCC1 are located on the apical (CSF facing) membrane. This raises an interesting conundrum as to the function of NKCC1 and the direction of transport. In (A), we propose that the epithelium can switch from a Na+ secretion mode (top model) to K+ reabsorption mode (bottom model). To make this work, one assumes high carbonic anhydrase (CA) activity leading to production of large amounts of bicarbonate. As HCO3 is exchanged with Cl at the basolateral membrane, cytosolic Cl raises enough to reverse the direction of NKCC1 transport. In this model, both NKCC1 and the pump participate to CSF Na+ secretion, while K+ can cycle at the apical membrane. In (B), we propose an alternative model where the direction of NKCC1 transport remains inward. K+ can be reabsorbed through the combined action of NKCC1 and the pump. If the pump was alone in mediating Na+ secretion and K+ reabsorption, these two processes would always be linked. NKCC1 on the same membrane then the pump unlinks K+ reabsorption from Na+ secretion.

Figure 6. Teaching points: The stria vascularis is a stratified epithelium in the inner ear. As seen in the Figure it is composed of multiple cell layers. Cells from the three basal layers are connected by gap junctions or large structures that connect the cytoplasm of cells and allow the movement of small substances. Basolateral NKCC1 together with apical K+ channels in both fibrocytes and marginal cells (MC) provide a pathway for K+ secretion from blood to the endolymphatic cavity. Note the unusually high concentration of K+in the endolymphatic cavity. The endolymph is a K+ rich medium, unlike all other environments external to cells. In this figure, we also drew the epithelium of the endolymphatic sac, which is thought to be reabsorptive. Studies have shown that NKCC2 might be expressed on the apical membrane providing a pathway for Na+ and fluid reabsorption. Note that in contrast to our model, many models still consider the endolymphatic sac to contain a K+-rich endolymph. Notice the small passage that connects the endolymphatic sac from the main cavity. It is meant to represent the long and narrow tube that connects the sac to the saccule. It does not provide an accurate representation of the anatomy.

Figure 7. Teaching points: K+ is the major ion involved in creating the electrical potential across the plasma membrane of a cell (A) or across the wall of the inner ear (B). Because the reference electrode is outside and the interior of the cell is negative, the membrane potential has a negative value (from -8 mV in chondrocytes to -95 mV in skeletal muscle). Similarly, because the reference electrode is outside and the endolymphatic cavity is positive, due to the marginal cells leak of K+, the endolymphatic potential has a positive value. In C to E, the structure of the inner ear is shown. The focus is on the Reissner's membrane (Rm) which separates the scala media (SM) from the scala vestibule (SV). The K+-rich endolymph that is secreted by the stria vascularis (St.v.) epithelium fills the scala media. In the absence of fluid secretion, the scala media is empty and the Reissner's membrane sits on top of the stria vascularis. Thus, striking morphological or anatomical changes signal functional changes.

Figure 8. Teaching points:Size of the testes in NKCC1 knockout mice can be explained by absence of spermatid maturation. Notice the mass of spermatozoa filling the lumen of the seminiferous tubule in heterozygous mice (A) versus complete absence in homozygous knockouts (B). The Figure also reports Terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL) for detection of apoptotic cell death. As cells undergo apoptosis, the cell's DNA is broken, exposing blunt-ends that can be labeled with HRP-conjugated (A and B) or FITC-conjugated dUTPs (C and D). As shown in the panels, an increased number of positive (red for A and B, and fluorescent green for C and D) cells is detected in the tubules from homozygote mice.

Figure 9. Teaching points: Homozygote NKCC1 mice are smaller and have smaller testis sizes than their heterozygote counterparts. While this information could be visualized with bar graphs, by plotting the size of the testes on one axis against/versus the overall body weight on the other axis, once can see (i) a correlation between testis weight and body weight (almost parallel red and blue lines), and (ii) a clear drop in testis weights in the homozygote mice, indicating a testis-specific phenotype.

Figure 10. Teaching points: The absorptive function of the Thick Ascending Limb is explained in (A). Transporters on apical and basolateral sides work in concert to move Na+ and Cl from lumen to blood. As in most epithelia, the transport is driven via ATP consumption by the Na+/K+ pump. Na+ and Cl enter the apical membrane through NKCC2, while Cl is released at the serosal side by CLC-Ka/b Cl channels. It is believed that the K+ concentration in the urine is limiting to the movement of Na+ through NKCC2, which is mitigated by (ROMK) K+ channels, which recycle the K+ that enters the cell through NKCC2. The leak of K+ at the apical membrane in combination with the leak of Cl at the basolateral membrane creates an electropositive lumen that helps the movement of divalent cations through paracellular pathways. The system is so well designed that mutations in either component of the Na+ absorptive pathway gives rise to variants of Bartter syndrome (BARTS 1-4).

Figure 11. Teaching points: NKCC2 function is regulated at many levels: transcription, trafficking, endocytosis. One pathway not displayed here is phosphorylation by WNK/SPAK-OSR1 kinases, which likely affects transport cycle turnover of transporters already existing at the plasma membrane. The Figure focuses on a major signaling molecule, cAMP, which leads to NKCC2 activation. Vasopressin activates, whereas prostaglandin inhibits, adenylate cyclase, the enzyme that produces cAMP. Endothelin, through production of nitrous oxide, activate guanylyl cyclase, which produces cGMP, which in turn activates the enzyme that degrades cAMP. Thus, activation of endothelin receptor inhibits NKCC2 function. cAMP not only affects NKCC2 function at the genomic level (through a cAMP-responsive element, CRE) in the promoter of NKCC2, but also facilitates the forward trafficking of the transporter and its insertion in the plasma membrane through PKA and VAMP2.


Related Articles:

Auditory Perception
Epilepsy: Insights into Higher Brain Functions in Humans
Pain and Nociception
Comparative Physiology of Colonic Electrolyte Transport
Fluid and Electrolyte Secretion by Salivary Glands
Interstitial cells of Cajal
Ion Transport Across Mammalian Small Intestine
Ion Transport Across the Large Intestine
Principles of Membrane Transport
Calcium Homeostasis in Health and in Kidney Disease
Chloride Transport
Chloride Transport
Classical Renin‐Angiotensin System in Kidney Physiology
Hypertension: Physiology and Pathophysiology
Ion and Water Transport in Toad Urinary Epithelia in Vitro
Mechanisms of Fluid Transport Across Renal Tubules
Tubuloglomerular Feedback Control of Renal Vascular Resistance
Osmolytes and Cell‐Volume Regulation: Physiological and Evolutionary Principles
Physiology of Electrolyte Transport in the Gut: Implications for Disease

Contact Editor

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

* Required Field

How to Cite

Eric Delpire, Kenneth B. Gagnon. Na+‐K+‐2Cl− Cotransporter (NKCC) Physiological Function in Nonpolarized Cells and Transporting Epithelia. Compr Physiol 2018, 8: 871-901. doi: 10.1002/cphy.c170018