Skip to main content
Top
Published in: Clinical and Experimental Nephrology 1/2012

01-02-2012 | Review Article

Serum- and glucocorticoid-inducible kinase 1 in the regulation of renal and extrarenal potassium transport

Authors: Florian Lang, Volker Vallon

Published in: Clinical and Experimental Nephrology | Issue 1/2012

Login to get access

Abstract

Serum- and glucocorticoid inducible-kinase 1 (SGK1) is an early gene transcriptionally upregulated by cell stress such as cell shrinkage and hypoxia and several hormones including gluco- and mineralocorticoids. It is activated by insulin and growth factors. SGK1 is a powerful regulator of a wide variety of channels and transporters. The present review describes the role of SGK1 in the regulation of potassium (K+) channels, K+ transporters and K+ homeostasis. SGK1-regulated K+ channels include renal outer medullary K+ channel, Kv1.3, Kv1.5, KCNE1/KCNQ1, KCNQ4 and, via regulation of calcium (Ca2+) entry, Ca2+-sensitive K+ channels. SGK1-sensitive transporters include sodium–potassium-chloride cotransporter 2 and sodium/potassium-adenosine triphosphatase. SGK1-dependent regulation of K+ channels and K+ transport contributes to the stimulation of renal K+ excretion following high K+ intake, to insulin-induced cellular K+ uptake and hypokalemia, to inhibition of insulin release by glucocorticoids, to stimulation of mast cell degranulation and gastric acid secretion, and to cardiac repolarization. Thus, SGK1 has a profound effect on K+ homeostasis and on a multitude of K+-sensitive cellular functions.
Literature
1.
go back to reference Lang F. SGK1 in the regulation of renal tubular transport and blood pressure control. Kitasato Med J. 2009;39:1–12. Lang F. SGK1 in the regulation of renal tubular transport and blood pressure control. Kitasato Med J. 2009;39:1–12.
2.
go back to reference Lang F, Bohmer C, Palmada M, Seebohm G, Strutz-Seebohm N, Vallon V. (Patho)physiological significance of the serum- and glucocorticoid-inducible kinase isoforms. Physiol Rev. 2006;86:1151–78.PubMedCrossRef Lang F, Bohmer C, Palmada M, Seebohm G, Strutz-Seebohm N, Vallon V. (Patho)physiological significance of the serum- and glucocorticoid-inducible kinase isoforms. Physiol Rev. 2006;86:1151–78.PubMedCrossRef
3.
go back to reference Vallon V, Wulff P, Huang DY, Loffing J, Volkl H, Kuhl D, et al. Role of Sgk1 in salt and potassium homeostasis. Am J Physiol Regul Integr Comp Physiol. 2005;288:R4–10.PubMedCrossRef Vallon V, Wulff P, Huang DY, Loffing J, Volkl H, Kuhl D, et al. Role of Sgk1 in salt and potassium homeostasis. Am J Physiol Regul Integr Comp Physiol. 2005;288:R4–10.PubMedCrossRef
4.
go back to reference Firestone GL, Giampaolo JR, O’Keeffe BA. Stimulus-dependent regulation of the serum and glucocorticoid inducible protein kinase (Sgk) transcription, subcellular localization and enzymatic activity. Cell Physiol Biochem. 2003;13:1–12.PubMedCrossRef Firestone GL, Giampaolo JR, O’Keeffe BA. Stimulus-dependent regulation of the serum and glucocorticoid inducible protein kinase (Sgk) transcription, subcellular localization and enzymatic activity. Cell Physiol Biochem. 2003;13:1–12.PubMedCrossRef
5.
go back to reference Webster MK, Goya L, Firestone GL. Immediate-early transcriptional regulation and rapid MRNA turnover of a putative serine/threonine protein kinase. J Biol Chem. 1993;268:11482–5.PubMed Webster MK, Goya L, Firestone GL. Immediate-early transcriptional regulation and rapid MRNA turnover of a putative serine/threonine protein kinase. J Biol Chem. 1993;268:11482–5.PubMed
6.
go back to reference Webster MK, Goya L, Ge Y, Maiyar AC, Firestone GL. Characterization of Sgk, a novel member of the serine/threonine protein kinase gene family which is transcriptionally induced by glucocorticoids and serum. Mol Cell Biol. 1993;13:2031–40.PubMed Webster MK, Goya L, Ge Y, Maiyar AC, Firestone GL. Characterization of Sgk, a novel member of the serine/threonine protein kinase gene family which is transcriptionally induced by glucocorticoids and serum. Mol Cell Biol. 1993;13:2031–40.PubMed
7.
go back to reference Waldegger S, Barth P, Raber G, Lang F. Cloning and characterization of a putative human serine/threonine protein kinase transcriptionally modified during anisotonic and isotonic alterations of cell volume. Proc Natl Acad Sci USA. 1997;94:4440–5.PubMedCrossRef Waldegger S, Barth P, Raber G, Lang F. Cloning and characterization of a putative human serine/threonine protein kinase transcriptionally modified during anisotonic and isotonic alterations of cell volume. Proc Natl Acad Sci USA. 1997;94:4440–5.PubMedCrossRef
8.
go back to reference Kobayashi T, Deak M, Morrice N, Cohen P. Characterization of the structure and regulation of two novel isoforms of serum- and glucocorticoid-induced protein kinase. Biochem J. 1999;344:189–97.PubMedCrossRef Kobayashi T, Deak M, Morrice N, Cohen P. Characterization of the structure and regulation of two novel isoforms of serum- and glucocorticoid-induced protein kinase. Biochem J. 1999;344:189–97.PubMedCrossRef
9.
go back to reference Biondi RM, Kieloch A, Currie RA, Deak M, Alessi DR. The PIF-binding pocket in PDK1 is essential for activation of S6K and SGK, but not PKB. EMBO J. 2001;20:4380–90.PubMedCrossRef Biondi RM, Kieloch A, Currie RA, Deak M, Alessi DR. The PIF-binding pocket in PDK1 is essential for activation of S6K and SGK, but not PKB. EMBO J. 2001;20:4380–90.PubMedCrossRef
10.
go back to reference Collins BJ, Deak M, Arthur JS, Armit LJ, Alessi DR. In vivo role of the PIF-binding docking site of PDK1 defined by knock-in mutation. EMBO J. 2003;22:4202–11.PubMedCrossRef Collins BJ, Deak M, Arthur JS, Armit LJ, Alessi DR. In vivo role of the PIF-binding docking site of PDK1 defined by knock-in mutation. EMBO J. 2003;22:4202–11.PubMedCrossRef
11.
go back to reference Frodin M, Antal TL, Dummler BA, Jensen CJ, Deak M, Gammeltoft S, et al. A phosphoserine/threonine-binding pocket in AGC kinases and PDK1 mediates activation by hydrophobic motif phosphorylation. EMBO J. 2002;21:5396–407.PubMedCrossRef Frodin M, Antal TL, Dummler BA, Jensen CJ, Deak M, Gammeltoft S, et al. A phosphoserine/threonine-binding pocket in AGC kinases and PDK1 mediates activation by hydrophobic motif phosphorylation. EMBO J. 2002;21:5396–407.PubMedCrossRef
12.
go back to reference Kobayashi T, Cohen P. Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. Biochem J. 1999;339:319–28.PubMedCrossRef Kobayashi T, Cohen P. Activation of serum- and glucocorticoid-regulated protein kinase by agonists that activate phosphatidylinositide 3-kinase is mediated by 3-phosphoinositide-dependent protein kinase-1 (PDK1) and PDK2. Biochem J. 1999;339:319–28.PubMedCrossRef
13.
go back to reference Mora A, Komander D, van Aalten DM, Alessi DR. PDK1, the master regulator of AGC kinase signal transduction. Semin Cell Dev Biol. 2004;15:161–70.PubMedCrossRef Mora A, Komander D, van Aalten DM, Alessi DR. PDK1, the master regulator of AGC kinase signal transduction. Semin Cell Dev Biol. 2004;15:161–70.PubMedCrossRef
14.
go back to reference Nilsen T, Slagsvold T, Skjerpen CS, Brech A, Stenmark H, Olsnes S. Peroxisomal targeting as a tool for assaying potein-protein interactions in the living cell: cytokine-independent survival kinase (CISK) binds PDK-1 in vivo in a phosphorylation-dependent manner. J Biol Chem. 2004;279:4794–801.PubMedCrossRef Nilsen T, Slagsvold T, Skjerpen CS, Brech A, Stenmark H, Olsnes S. Peroxisomal targeting as a tool for assaying potein-protein interactions in the living cell: cytokine-independent survival kinase (CISK) binds PDK-1 in vivo in a phosphorylation-dependent manner. J Biol Chem. 2004;279:4794–801.PubMedCrossRef
15.
go back to reference Park J, Leong ML, Buse P, Maiyar AC, Firestone GL, Hemmings BA. Serum and glucocorticoid-inducible kinase (SGK) is a target of the PI 3-kinase-stimulated signaling pathway. EMBO J. 1999;18:3024–33.PubMedCrossRef Park J, Leong ML, Buse P, Maiyar AC, Firestone GL, Hemmings BA. Serum and glucocorticoid-inducible kinase (SGK) is a target of the PI 3-kinase-stimulated signaling pathway. EMBO J. 1999;18:3024–33.PubMedCrossRef
16.
go back to reference Palmada M, Embark HM, Yun C, Bohmer C, Lang F. Molecular requirements for the regulation of the renal outer medullary K(+) channel ROMK1 by the serum- and glucocorticoid-inducible kinase SGK1. Biochem Biophys Res Commun. 2003;311:629–34.PubMedCrossRef Palmada M, Embark HM, Yun C, Bohmer C, Lang F. Molecular requirements for the regulation of the renal outer medullary K(+) channel ROMK1 by the serum- and glucocorticoid-inducible kinase SGK1. Biochem Biophys Res Commun. 2003;311:629–34.PubMedCrossRef
17.
go back to reference Yun CC, Palmada M, Embark HM, Fedorenko O, Feng Y, Henke G, et al. The serum and glucocorticoid-inducible kinase SGK1 and the Na(+)/H(+) exchange regulating factor NHERF2 synergize to stimulate the renal outer medullary K(+) channel ROMK1. J Am Soc Nephrol. 2002;13:2823–30.PubMedCrossRef Yun CC, Palmada M, Embark HM, Fedorenko O, Feng Y, Henke G, et al. The serum and glucocorticoid-inducible kinase SGK1 and the Na(+)/H(+) exchange regulating factor NHERF2 synergize to stimulate the renal outer medullary K(+) channel ROMK1. J Am Soc Nephrol. 2002;13:2823–30.PubMedCrossRef
18.
go back to reference Yoo D, Kim BY, Campo C, Nance L, King A, Maouyo D, et al. Cell surface expression of the ROMK (Kir 1.1) channel is regulated by the aldosterone-induced kinase, SGK-1, and protein kinase A. J Biol Chem. 2003;278:23066–75.PubMedCrossRef Yoo D, Kim BY, Campo C, Nance L, King A, Maouyo D, et al. Cell surface expression of the ROMK (Kir 1.1) channel is regulated by the aldosterone-induced kinase, SGK-1, and protein kinase A. J Biol Chem. 2003;278:23066–75.PubMedCrossRef
19.
go back to reference Shenolikar S, Weinman EJ. NHERF: targeting and trafficking membrane proteins. Am J Physiol Renal Physiol. 2001;280:F389–95.PubMed Shenolikar S, Weinman EJ. NHERF: targeting and trafficking membrane proteins. Am J Physiol Renal Physiol. 2001;280:F389–95.PubMed
20.
go back to reference Yun CC. Concerted roles of SGK1 and the Na+/H+ exchanger regulatory factor 2 (NHERF2) in regulation of NHE3. Cell Physiol Biochem. 2003;13:29–40.CrossRef Yun CC. Concerted roles of SGK1 and the Na+/H+ exchanger regulatory factor 2 (NHERF2) in regulation of NHE3. Cell Physiol Biochem. 2003;13:29–40.CrossRef
21.
go back to reference Palmada M, Embark HM, Wyatt AW, Bohmer C, Lang F. Negative charge at the consensus sequence for the serum- and glucocorticoid-inducible kinase, SGK1, determines PH sensitivity of the renal outer medullary K+ channel, ROMK1. Biochem Biophys Res Commun. 2003;307:967–72.PubMedCrossRef Palmada M, Embark HM, Wyatt AW, Bohmer C, Lang F. Negative charge at the consensus sequence for the serum- and glucocorticoid-inducible kinase, SGK1, determines PH sensitivity of the renal outer medullary K+ channel, ROMK1. Biochem Biophys Res Commun. 2003;307:967–72.PubMedCrossRef
22.
go back to reference Embark HM, Bohmer C, Vallon V, Luft F, Lang F. Regulation of KCNE1-dependent K(+) current by the serum and glucocorticoid-inducible kinase (SGK) isoforms. Pflugers Arch Eur J Physiol. 2003;445:601–6. Embark HM, Bohmer C, Vallon V, Luft F, Lang F. Regulation of KCNE1-dependent K(+) current by the serum and glucocorticoid-inducible kinase (SGK) isoforms. Pflugers Arch Eur J Physiol. 2003;445:601–6.
23.
go back to reference Takumi T, Ohkubo H, Nakanishi S. Cloning of a membrane protein that induces a slow voltage-gated potassium current. Science. 1988;242:1042–5.PubMedCrossRef Takumi T, Ohkubo H, Nakanishi S. Cloning of a membrane protein that induces a slow voltage-gated potassium current. Science. 1988;242:1042–5.PubMedCrossRef
24.
go back to reference Debonneville C, Flores SY, Kamynina E, Plant PJ, Tauxe C, Thomas MA, et al. Phosphorylation of Nedd4-2 by Sgk1 regulates epithelial Na(+) channel cell surface expression. EMBO J. 2001;20:7052–9.PubMedCrossRef Debonneville C, Flores SY, Kamynina E, Plant PJ, Tauxe C, Thomas MA, et al. Phosphorylation of Nedd4-2 by Sgk1 regulates epithelial Na(+) channel cell surface expression. EMBO J. 2001;20:7052–9.PubMedCrossRef
25.
go back to reference Ichimura T, Yamamura H, Sasamoto K, Tominaga Y, Taoka M, Kakiuchi K, et al. 14-3-3 proteins modulate the expression of epithelial Na+ channels by phosphorylation-dependent interaction with Nedd4-2 ubiquitin ligase. J Biol Chem. 2005;280:13187–94.PubMedCrossRef Ichimura T, Yamamura H, Sasamoto K, Tominaga Y, Taoka M, Kakiuchi K, et al. 14-3-3 proteins modulate the expression of epithelial Na+ channels by phosphorylation-dependent interaction with Nedd4-2 ubiquitin ligase. J Biol Chem. 2005;280:13187–94.PubMedCrossRef
26.
go back to reference Liang X, Peters KW, Butterworth MB, Frizzell RA. 14-3-3 isoforms are induced by aldosterone and participate in its regulation of epithelial sodium channels. J Biol Chem. 2006;281:16323–32.PubMedCrossRef Liang X, Peters KW, Butterworth MB, Frizzell RA. 14-3-3 isoforms are induced by aldosterone and participate in its regulation of epithelial sodium channels. J Biol Chem. 2006;281:16323–32.PubMedCrossRef
27.
go back to reference Nagaki K, Yamamura H, Shimada S, Saito T, Hisanaga S, Taoka M, et al. 14-3-3 mediates phosphorylation-dependent inhibition of the interaction between the ubiquitin E3 ligase Nedd4-2 and epithelial Na+ channels. Biochemistry. 2006;45:6733–40.PubMedCrossRef Nagaki K, Yamamura H, Shimada S, Saito T, Hisanaga S, Taoka M, et al. 14-3-3 mediates phosphorylation-dependent inhibition of the interaction between the ubiquitin E3 ligase Nedd4-2 and epithelial Na+ channels. Biochemistry. 2006;45:6733–40.PubMedCrossRef
28.
go back to reference Seebohm G, Strutz-Seebohm N, Baltaev R, Korniychuk G, Knirsch M, Engel J, et al. Regulation of KCNQ4 potassium channel prepulse dependence and current amplitude by SGK1 in Xenopus oocytes. Cell Physiol Biochem. 2005;16:255–62.PubMedCrossRef Seebohm G, Strutz-Seebohm N, Baltaev R, Korniychuk G, Knirsch M, Engel J, et al. Regulation of KCNQ4 potassium channel prepulse dependence and current amplitude by SGK1 in Xenopus oocytes. Cell Physiol Biochem. 2005;16:255–62.PubMedCrossRef
29.
go back to reference Hougaard C, Klaerke DA, Hoffmann EK, Olesen SP, Jorgensen NK. Modulation of KCNQ4 channel activity by changes in cell volume. Biochim Biophys Acta. 2004;1660:1–6.PubMedCrossRef Hougaard C, Klaerke DA, Hoffmann EK, Olesen SP, Jorgensen NK. Modulation of KCNQ4 channel activity by changes in cell volume. Biochim Biophys Acta. 2004;1660:1–6.PubMedCrossRef
30.
go back to reference Kubisch C, Schroeder BC, Friedrich T, Lutjohann B, El Amraoui A, Marlin S, et al. KCNQ4, a novel potassium channel expressed in sensory outer hair cells, is mutated in dominant deafness. Cell. 1999;96:437–46.PubMedCrossRef Kubisch C, Schroeder BC, Friedrich T, Lutjohann B, El Amraoui A, Marlin S, et al. KCNQ4, a novel potassium channel expressed in sensory outer hair cells, is mutated in dominant deafness. Cell. 1999;96:437–46.PubMedCrossRef
31.
go back to reference Marcotti W, Kros CJ. Developmental expression of the potassium current IK, n contributes to maturation of mouse outer hair cells. J Physiol. 1999;520(Pt 3):653–60.PubMedCrossRef Marcotti W, Kros CJ. Developmental expression of the potassium current IK, n contributes to maturation of mouse outer hair cells. J Physiol. 1999;520(Pt 3):653–60.PubMedCrossRef
32.
go back to reference Aronzon A, Ruckenstein MJ, Bigelow DC. The efficacy of corticosteroids in restoring hearing in patients undergoing conservative management of acoustic neuromas. Otol Neurotol. 2003;24:465–8.PubMedCrossRef Aronzon A, Ruckenstein MJ, Bigelow DC. The efficacy of corticosteroids in restoring hearing in patients undergoing conservative management of acoustic neuromas. Otol Neurotol. 2003;24:465–8.PubMedCrossRef
33.
go back to reference Hillman TM, Arriaga MA, Chen DA. Intratympanic steroids: do they acutely improve hearing in cases of cochlear hydrops? Laryngoscope. 2003;113:1903–7.PubMedCrossRef Hillman TM, Arriaga MA, Chen DA. Intratympanic steroids: do they acutely improve hearing in cases of cochlear hydrops? Laryngoscope. 2003;113:1903–7.PubMedCrossRef
34.
go back to reference Shulman A, Goldstein B. Intratympanic drug therapy with steroids for tinnitus control: a preliminary report. Int Tinnitus J. 2000;6:10–20.PubMed Shulman A, Goldstein B. Intratympanic drug therapy with steroids for tinnitus control: a preliminary report. Int Tinnitus J. 2000;6:10–20.PubMed
35.
go back to reference Gamper N, Fillon S, Huber SM, Feng Y, Kobayashi T, Cohen P, et al. IGF-1 up-regulates K+ channels via PI3-kinase, PDK1 and SGK1. Pflugers Arch Eur J Physiol. 2002;443:625–34.CrossRef Gamper N, Fillon S, Huber SM, Feng Y, Kobayashi T, Cohen P, et al. IGF-1 up-regulates K+ channels via PI3-kinase, PDK1 and SGK1. Pflugers Arch Eur J Physiol. 2002;443:625–34.CrossRef
36.
go back to reference Henke G, Maier G, Wallisch S, Boehmer C, Lang F. Regulation of the voltage gated K+ channel Kv1.3 by the ubiquitin ligase Nedd4-2 and the serum and glucocorticoid inducible kinase SGK1. J Cell Physiol. 2004;199:194–9.PubMedCrossRef Henke G, Maier G, Wallisch S, Boehmer C, Lang F. Regulation of the voltage gated K+ channel Kv1.3 by the ubiquitin ligase Nedd4-2 and the serum and glucocorticoid inducible kinase SGK1. J Cell Physiol. 2004;199:194–9.PubMedCrossRef
37.
go back to reference Wärntges S, Friedrich B, Henke G, Duranton C, Lang PA, Waldegger S, et al. Cerebral localization and regulation of the cell volume-sensitive serum- and glucocorticoid-dependent kinase SGK1. Pflugers Arch Eur J Physiol. 2002;443:617–24.CrossRef Wärntges S, Friedrich B, Henke G, Duranton C, Lang PA, Waldegger S, et al. Cerebral localization and regulation of the cell volume-sensitive serum- and glucocorticoid-dependent kinase SGK1. Pflugers Arch Eur J Physiol. 2002;443:617–24.CrossRef
38.
go back to reference Boehmer C, Laufer J, Jeyaraj S, Klaus F, Lindner R, Lang F, et al. Modulation of the voltage-gated potassium channel Kv1.5 by the SGK1 protein kinase involves inhibition of channel ubiquitination. Cell Physiol Biochem. 2008;22:591–600.PubMedCrossRef Boehmer C, Laufer J, Jeyaraj S, Klaus F, Lindner R, Lang F, et al. Modulation of the voltage-gated potassium channel Kv1.5 by the SGK1 protein kinase involves inhibition of channel ubiquitination. Cell Physiol Biochem. 2008;22:591–600.PubMedCrossRef
39.
go back to reference Laufer J, Boehmer C, Jeyaraj S, Knuwer M, Klaus F, Lindner R, et al. The C-terminal PDZ-binding motif in the Kv1.5 potassium channel governs its modulation by the Na+/H+ exchanger regulatory factor 2. Cell Physiol Biochem. 2009;23:25–36.PubMedCrossRef Laufer J, Boehmer C, Jeyaraj S, Knuwer M, Klaus F, Lindner R, et al. The C-terminal PDZ-binding motif in the Kv1.5 potassium channel governs its modulation by the Na+/H+ exchanger regulatory factor 2. Cell Physiol Biochem. 2009;23:25–36.PubMedCrossRef
40.
go back to reference Ullrich S, Berchtold S, Ranta F, Seebohm G, Henke G, Lupescu A, et al. Serum- and glucocorticoid-inducible kinase 1 (SGK1) mediates glucocorticoid-induced inhibition of insulin secretion. Diabetes. 2005;54:1090–9.PubMedCrossRef Ullrich S, Berchtold S, Ranta F, Seebohm G, Henke G, Lupescu A, et al. Serum- and glucocorticoid-inducible kinase 1 (SGK1) mediates glucocorticoid-induced inhibition of insulin secretion. Diabetes. 2005;54:1090–9.PubMedCrossRef
41.
go back to reference Baltaev R, Strutz-Seebohm N, Korniychuk G, Myssina S, Lang F, Seebohm G. Regulation of cardiac shal-related potassium channel Kv 4.3 by serum- and glucocorticoid-inducible kinase isoforms in Xenopus oocytes. Pflugers Arch. 2005;450:26–33.PubMedCrossRef Baltaev R, Strutz-Seebohm N, Korniychuk G, Myssina S, Lang F, Seebohm G. Regulation of cardiac shal-related potassium channel Kv 4.3 by serum- and glucocorticoid-inducible kinase isoforms in Xenopus oocytes. Pflugers Arch. 2005;450:26–33.PubMedCrossRef
42.
go back to reference Gamper N, Fillon S, Feng Y, Friedrich B, Lang PA, Henke G, et al. K(+) channel activation by all three isoforms of serum- and glucocorticoid-dependent protein kinase SGK. Pflugers Arch Eur J Physiol. 2002;445:60–6.CrossRef Gamper N, Fillon S, Feng Y, Friedrich B, Lang PA, Henke G, et al. K(+) channel activation by all three isoforms of serum- and glucocorticoid-dependent protein kinase SGK. Pflugers Arch Eur J Physiol. 2002;445:60–6.CrossRef
43.
go back to reference Shumilina E, Lampert A, Lupescu A, Myssina S, Strutz-Seebohm N, Henke G, et al. Deranged Kv channel regulation in fibroblasts from mice lacking the serum and glucocorticoid inducible kinase SGK1. J Cell Physiol. 2005;204:87–98.PubMedCrossRef Shumilina E, Lampert A, Lupescu A, Myssina S, Strutz-Seebohm N, Henke G, et al. Deranged Kv channel regulation in fibroblasts from mice lacking the serum and glucocorticoid inducible kinase SGK1. J Cell Physiol. 2005;204:87–98.PubMedCrossRef
44.
go back to reference Alvarez dlR, Coric T, Todorovic N, Shao D, Wang T, Canessa CM. Distribution and regulation of expression of serum- and glucocorticoid-induced kinase-1 in the rat kidney. J Physiol. 2003;551:455–66.CrossRef Alvarez dlR, Coric T, Todorovic N, Shao D, Wang T, Canessa CM. Distribution and regulation of expression of serum- and glucocorticoid-induced kinase-1 in the rat kidney. J Physiol. 2003;551:455–66.CrossRef
45.
go back to reference Setiawan I, Henke G, Feng Y, Bohmer C, Vasilets LA, Schwarz W, et al. Stimulation of Xenopus Oocyte Na(+), K(+)ATPase by the serum and glucocorticoid-dependent kinase Sgk1. Pflugers Arch Eur J Physiol. 2002;444:426–31.CrossRef Setiawan I, Henke G, Feng Y, Bohmer C, Vasilets LA, Schwarz W, et al. Stimulation of Xenopus Oocyte Na(+), K(+)ATPase by the serum and glucocorticoid-dependent kinase Sgk1. Pflugers Arch Eur J Physiol. 2002;444:426–31.CrossRef
46.
go back to reference Verrey F, Summa V, Heitzmann D, Mordasini D, Vandewalle A, Feraille E, et al. Short-term aldosterone action on Na, K-ATPase surface expression: role of aldosterone-induced SGK1? Ann N Y Acad Sci. 2003;986:554–61.PubMedCrossRef Verrey F, Summa V, Heitzmann D, Mordasini D, Vandewalle A, Feraille E, et al. Short-term aldosterone action on Na, K-ATPase surface expression: role of aldosterone-induced SGK1? Ann N Y Acad Sci. 2003;986:554–61.PubMedCrossRef
47.
go back to reference Zecevic M, Heitzmann D, Camargo SM, Verrey F. SGK1 increases Na, K-ATP cell-surface expression and function in Xenopus laevis oocytes. Pflugers Arch. 2004;448:29–35.PubMedCrossRef Zecevic M, Heitzmann D, Camargo SM, Verrey F. SGK1 increases Na, K-ATP cell-surface expression and function in Xenopus laevis oocytes. Pflugers Arch. 2004;448:29–35.PubMedCrossRef
48.
go back to reference Alvarez dlR, Gimenez I, Forbush B, Canessa CM. SGK1 activates Na+-K+-ATPase in amphibian renal epithelial cells. Am J Physiol Cell Physiol. 2006;290:C492–8.CrossRef Alvarez dlR, Gimenez I, Forbush B, Canessa CM. SGK1 activates Na+-K+-ATPase in amphibian renal epithelial cells. Am J Physiol Cell Physiol. 2006;290:C492–8.CrossRef
49.
go back to reference Ullrich S, Zhang Y, Avram D, Ranta F, Kuhl D, Haring HU, et al. Dexamethasone increases Na+/K+ ATPase activity in insulin secreting cells through SGK1. Biochem Biophys Res Commun. 2007;352:662–7.PubMedCrossRef Ullrich S, Zhang Y, Avram D, Ranta F, Kuhl D, Haring HU, et al. Dexamethasone increases Na+/K+ ATPase activity in insulin secreting cells through SGK1. Biochem Biophys Res Commun. 2007;352:662–7.PubMedCrossRef
50.
go back to reference Henke G, Setiawan I, Bohmer C, Lang F. Activation of Na+/K+-ATPase by the serum and glucocorticoid-dependent kinase isoforms. Kidney Blood Press Res. 2002;25:370–4.PubMedCrossRef Henke G, Setiawan I, Bohmer C, Lang F. Activation of Na+/K+-ATPase by the serum and glucocorticoid-dependent kinase isoforms. Kidney Blood Press Res. 2002;25:370–4.PubMedCrossRef
51.
go back to reference Ali S, Wei Y, Lerea KM, Becker L, Rubin CS, Wang W. PKA-induced stimulation of ROMK1 channel activity is governed by both tethering and non-tethering domains of an A kinase anchor protein. Cell Physiol Biochem. 2001;11:135–42.PubMedCrossRef Ali S, Wei Y, Lerea KM, Becker L, Rubin CS, Wang W. PKA-induced stimulation of ROMK1 channel activity is governed by both tethering and non-tethering domains of an A kinase anchor protein. Cell Physiol Biochem. 2001;11:135–42.PubMedCrossRef
52.
go back to reference Giebisch G. Renal potassium transport: mechanisms and regulation. Am J Physiol. 1998;274:F817–33.PubMed Giebisch G. Renal potassium transport: mechanisms and regulation. Am J Physiol. 1998;274:F817–33.PubMed
53.
go back to reference Wang W. Regulation of the ROMK channel: interaction of the ROMK with associate proteins. Am J Physiol. 1999;277:F826–31.PubMed Wang W. Regulation of the ROMK channel: interaction of the ROMK with associate proteins. Am J Physiol. 1999;277:F826–31.PubMed
54.
go back to reference Wald H, Garty H, Palmer LG, Popovtzer MM. Differential regulation of ROMK expression in kidney cortex and medulla by aldosterone and potassium. Am J Physiol. 1998;275:F239–45.PubMed Wald H, Garty H, Palmer LG, Popovtzer MM. Differential regulation of ROMK expression in kidney cortex and medulla by aldosterone and potassium. Am J Physiol. 1998;275:F239–45.PubMed
55.
go back to reference Vallon V, Schroth J, Lang F, Kuhl D, Uchida S. Expression and phosphorylation of the Na+-Cl− cotransporter NCC in vivo is regulated by dietary salt, potassium, and SGK1. Am J Physiol Renal Physiol. 2009;297:F704–12.PubMedCrossRef Vallon V, Schroth J, Lang F, Kuhl D, Uchida S. Expression and phosphorylation of the Na+-Cl cotransporter NCC in vivo is regulated by dietary salt, potassium, and SGK1. Am J Physiol Renal Physiol. 2009;297:F704–12.PubMedCrossRef
56.
go back to reference Huang DY, Wulff P, Volkl H, Loffing J, Richter K, Kuhl D, et al. Impaired regulation of renal K+ elimination in the Sgk1-knockout mouse. J Am Soc Nephrol. 2004;15:885–91.PubMedCrossRef Huang DY, Wulff P, Volkl H, Loffing J, Richter K, Kuhl D, et al. Impaired regulation of renal K+ elimination in the Sgk1-knockout mouse. J Am Soc Nephrol. 2004;15:885–91.PubMedCrossRef
57.
go back to reference Fejes-Toth G, Frindt G, Naray-Fejes-Toth A, Palmer LG. Epithelial Na+ channel activation and processing in mice lacking SGK1. Am J Physiol Renal Physiol. 2008;294:F1298–305.PubMedCrossRef Fejes-Toth G, Frindt G, Naray-Fejes-Toth A, Palmer LG. Epithelial Na+ channel activation and processing in mice lacking SGK1. Am J Physiol Renal Physiol. 2008;294:F1298–305.PubMedCrossRef
58.
go back to reference Wulff P, Vallon V, Huang DY, Volkl H, Yu F, Richter K, et al. Impaired renal Na(+) retention in the Sgk1-knockout mouse. J Clin Invest. 2002;110:1263–8.PubMed Wulff P, Vallon V, Huang DY, Volkl H, Yu F, Richter K, et al. Impaired renal Na(+) retention in the Sgk1-knockout mouse. J Clin Invest. 2002;110:1263–8.PubMed
59.
go back to reference Sandulache D, Grahammer F, Artunc F, Henke G, Hussain A, Nasir O, et al. Renal Ca2+ handling in Sgk1 knockout mice. Pflugers Arch. 2006;452:444–52.PubMedCrossRef Sandulache D, Grahammer F, Artunc F, Henke G, Hussain A, Nasir O, et al. Renal Ca2+ handling in Sgk1 knockout mice. Pflugers Arch. 2006;452:444–52.PubMedCrossRef
60.
go back to reference Yao X, Chang AY, Boulpaep EL, Segal AS, Desir GV. Molecular cloning of a glibenclamide-sensitive, voltage-gated potassium channel expressed in rabbit kidney. J Clin Invest. 1996;97:2525–33.PubMedCrossRef Yao X, Chang AY, Boulpaep EL, Segal AS, Desir GV. Molecular cloning of a glibenclamide-sensitive, voltage-gated potassium channel expressed in rabbit kidney. J Clin Invest. 1996;97:2525–33.PubMedCrossRef
61.
go back to reference Huang DY, Boini KM, Friedrich B, Metzger M, Just L, Osswald H, et al. Blunted hypertensive effect of combined fructose and high-salt diet in gene-targeted mice lacking functional serum- and glucocorticoid-inducible kinase SGK1. Am J Physiol Regul Integr Comp Physiol. 2006;290:R935–44.PubMedCrossRef Huang DY, Boini KM, Friedrich B, Metzger M, Just L, Osswald H, et al. Blunted hypertensive effect of combined fructose and high-salt diet in gene-targeted mice lacking functional serum- and glucocorticoid-inducible kinase SGK1. Am J Physiol Regul Integr Comp Physiol. 2006;290:R935–44.PubMedCrossRef
62.
go back to reference Huang DY, Boini KM, Osswald H, Friedrich B, Artunc F, Ullrich S, et al. Resistance of mice lacking the serum- and glucocorticoid-inducible kinase SGK1 against salt-sensitive hypertension induced by a high-fat diet. Am J Physiol Renal Physiol. 2006;291:F1264–73.PubMedCrossRef Huang DY, Boini KM, Osswald H, Friedrich B, Artunc F, Ullrich S, et al. Resistance of mice lacking the serum- and glucocorticoid-inducible kinase SGK1 against salt-sensitive hypertension induced by a high-fat diet. Am J Physiol Renal Physiol. 2006;291:F1264–73.PubMedCrossRef
63.
go back to reference Lang F, Klingel K, Wagner CA, Stegen C, Warntges S, Friedrich B, et al. Deranged transcriptional regulation of cell-volume-sensitive kinase HSGK in diabetic nephropathy. Proc Natl Acad Sci USA. 2000;97:8157–62.PubMedCrossRef Lang F, Klingel K, Wagner CA, Stegen C, Warntges S, Friedrich B, et al. Deranged transcriptional regulation of cell-volume-sensitive kinase HSGK in diabetic nephropathy. Proc Natl Acad Sci USA. 2000;97:8157–62.PubMedCrossRef
64.
go back to reference Ackermann TF, Boini KM, Volkl H, Bhandaru M, Bareiss PM, Just L, et al. SGK1-sensitive renal tubular glucose reabsorption in diabetes. Am J Physiol Renal Physiol. 2009;296:F859–66.PubMedCrossRef Ackermann TF, Boini KM, Volkl H, Bhandaru M, Bareiss PM, Just L, et al. SGK1-sensitive renal tubular glucose reabsorption in diabetes. Am J Physiol Renal Physiol. 2009;296:F859–66.PubMedCrossRef
65.
go back to reference Dieter M, Palmada M, Rajamanickam J, Aydin A, Busjahn A, Boehmer C, et al. Regulation of glucose transporter SGLT1 by ubiquitin ligase Nedd4-2 and kinases SGK1, SGK3, and PKB. Obes Res. 2004;12:862–70.PubMedCrossRef Dieter M, Palmada M, Rajamanickam J, Aydin A, Busjahn A, Boehmer C, et al. Regulation of glucose transporter SGLT1 by ubiquitin ligase Nedd4-2 and kinases SGK1, SGK3, and PKB. Obes Res. 2004;12:862–70.PubMedCrossRef
66.
go back to reference Vallon V, Grahammer F, Richter K, Bleich M, Lang F, Barhanin J, et al. Role of KCNE1-dependent K+ fluxes in mouse proximal tubule. J Am Soc Nephrol. 2001;12:2003–11.PubMed Vallon V, Grahammer F, Richter K, Bleich M, Lang F, Barhanin J, et al. Role of KCNE1-dependent K+ fluxes in mouse proximal tubule. J Am Soc Nephrol. 2001;12:2003–11.PubMed
67.
go back to reference Vallon V, Grahammer F, Volkl H, Sandu CD, Richter K, Rexhepaj R, et al. KCNQ1-dependent transport in renal and gastrointestinal epithelia. Proc Natl Acad Sci USA. 2005;102:17864–9.PubMedCrossRef Vallon V, Grahammer F, Volkl H, Sandu CD, Richter K, Rexhepaj R, et al. KCNQ1-dependent transport in renal and gastrointestinal epithelia. Proc Natl Acad Sci USA. 2005;102:17864–9.PubMedCrossRef
68.
go back to reference Shibata S, Nagase M, Yoshida S, Kawachi H, Fujita T. Podocyte as the target for aldosterone: roles of oxidative stress and Sgk1. Hypertension. 2007;49:355–64.PubMedCrossRef Shibata S, Nagase M, Yoshida S, Kawachi H, Fujita T. Podocyte as the target for aldosterone: roles of oxidative stress and Sgk1. Hypertension. 2007;49:355–64.PubMedCrossRef
69.
go back to reference Nagase M, Yoshida S, Shibata S, Nagase T, Gotoda T, Ando K, et al. Enhanced aldosterone signaling in the early nephropathy of rats with metabolic syndrome: possible contribution of fat-derived factors. J Am Soc Nephrol. 2006;17:3438–46.PubMedCrossRef Nagase M, Yoshida S, Shibata S, Nagase T, Gotoda T, Ando K, et al. Enhanced aldosterone signaling in the early nephropathy of rats with metabolic syndrome: possible contribution of fat-derived factors. J Am Soc Nephrol. 2006;17:3438–46.PubMedCrossRef
70.
go back to reference Artunc F, Amann K, Nasir O, Friedrich B, Sandulache D, Jahovic N, et al. Blunted DOCA/high salt induced albuminuria and renal tubulointerstitial damage in gene-targeted mice lacking SGK1. J Mol Med. 2006;84:737–46.PubMedCrossRef Artunc F, Amann K, Nasir O, Friedrich B, Sandulache D, Jahovic N, et al. Blunted DOCA/high salt induced albuminuria and renal tubulointerstitial damage in gene-targeted mice lacking SGK1. J Mol Med. 2006;84:737–46.PubMedCrossRef
71.
go back to reference Coric T, Hernandez N, Alvarez dlR, Shao D, Wang T, Canessa CM. Expression of ENaC and serum- and glucocorticoid-induced kinase 1 in the rat intestinal epithelium. Am J Physiol Gastrointest Liver Physiol. 2004;286:G663–70.PubMedCrossRef Coric T, Hernandez N, Alvarez dlR, Shao D, Wang T, Canessa CM. Expression of ENaC and serum- and glucocorticoid-induced kinase 1 in the rat intestinal epithelium. Am J Physiol Gastrointest Liver Physiol. 2004;286:G663–70.PubMedCrossRef
72.
go back to reference Waldegger S, Klingel K, Barth P, Sauter M, Rfer ML, Kandolf R, et al. H-Sgk serine-threonine protein kinase gene as transcriptional target of transforming growth factor beta in human intestine. Gastroenterology. 1999;116:1081–8.PubMedCrossRef Waldegger S, Klingel K, Barth P, Sauter M, Rfer ML, Kandolf R, et al. H-Sgk serine-threonine protein kinase gene as transcriptional target of transforming growth factor beta in human intestine. Gastroenterology. 1999;116:1081–8.PubMedCrossRef
73.
go back to reference Palmada M, Dieter M, Speil A, Bohmer C, Mack AF, Wagner HJ, et al. Regulation of intestinal phosphate cotransporter NaPi IIb by ubiquitin ligase Nedd4-2 and by serum- and glucocorticoid-dependent kinase 1. Am J Physiol Gastrointest Liver Physiol. 2004;287:G143–50.PubMedCrossRef Palmada M, Dieter M, Speil A, Bohmer C, Mack AF, Wagner HJ, et al. Regulation of intestinal phosphate cotransporter NaPi IIb by ubiquitin ligase Nedd4-2 and by serum- and glucocorticoid-dependent kinase 1. Am J Physiol Gastrointest Liver Physiol. 2004;287:G143–50.PubMedCrossRef
74.
go back to reference Rotte A, Bhandaru M, Foller M, Biswas R, Mack AF, Friedrich B, et al. APC sensitive gastric acid secretion. Cell Physiol Biochem. 2009;23:133–42.PubMedCrossRef Rotte A, Bhandaru M, Foller M, Biswas R, Mack AF, Friedrich B, et al. APC sensitive gastric acid secretion. Cell Physiol Biochem. 2009;23:133–42.PubMedCrossRef
75.
go back to reference Rotte A, Mack AF, Bhandaru M, Kempe DS, Beier N, Scholz W, et al. Pioglitazone induced gastric acid secretion. Cell Physiol Biochem. 2009;24:193–200.PubMedCrossRef Rotte A, Mack AF, Bhandaru M, Kempe DS, Beier N, Scholz W, et al. Pioglitazone induced gastric acid secretion. Cell Physiol Biochem. 2009;24:193–200.PubMedCrossRef
76.
go back to reference Sandu C, Artunc F, Grahammer F, Rotte A, Boini KM, Friedrich B, et al. Role of the serum and glucocorticoid inducible kinase SGK1 in glucocorticoid stimulation of gastric acid secretion. Pflugers Arch. 2007;455:493–503.PubMedCrossRef Sandu C, Artunc F, Grahammer F, Rotte A, Boini KM, Friedrich B, et al. Role of the serum and glucocorticoid inducible kinase SGK1 in glucocorticoid stimulation of gastric acid secretion. Pflugers Arch. 2007;455:493–503.PubMedCrossRef
77.
go back to reference Rotte A, Bhandaru M, Ackermann TF, Boini KM, Lang F. Role of PDK1 in regulation of gastric acid secretion. Cell Physiol Biochem. 2008;22:725–34.PubMedCrossRef Rotte A, Bhandaru M, Ackermann TF, Boini KM, Lang F. Role of PDK1 in regulation of gastric acid secretion. Cell Physiol Biochem. 2008;22:725–34.PubMedCrossRef
78.
go back to reference Grahammer F, Artunc F, Sandulache D, Rexhepaj R, Friedrich B, Risler T, et al. Renal function of gene-targeted mice lacking both SGK1 and SGK3. Am J Physiol Regul Integr Comp Physiol. 2006;290:R945–50.PubMedCrossRef Grahammer F, Artunc F, Sandulache D, Rexhepaj R, Friedrich B, Risler T, et al. Renal function of gene-targeted mice lacking both SGK1 and SGK3. Am J Physiol Regul Integr Comp Physiol. 2006;290:R945–50.PubMedCrossRef
79.
go back to reference Rexhepaj R, Rotte A, Kempe DS, Sopjani M, Foller M, Gehring EM, et al. Stimulation of electrogenic intestinal dipeptide transport by the glucocorticoid dexamethasone. Pflugers Arch. 2009;459:191–202.PubMedCrossRef Rexhepaj R, Rotte A, Kempe DS, Sopjani M, Foller M, Gehring EM, et al. Stimulation of electrogenic intestinal dipeptide transport by the glucocorticoid dexamethasone. Pflugers Arch. 2009;459:191–202.PubMedCrossRef
80.
go back to reference Boehmer C, Palmada M, Klaus F, Jeyaraj S, Lindner R, Laufer J, et al. The peptide transporter PEPT2 is targeted by the protein kinase SGK1 and the scaffold protein NHERF2. Cell Physiol Biochem. 2008;22:705–14.PubMedCrossRef Boehmer C, Palmada M, Klaus F, Jeyaraj S, Lindner R, Laufer J, et al. The peptide transporter PEPT2 is targeted by the protein kinase SGK1 and the scaffold protein NHERF2. Cell Physiol Biochem. 2008;22:705–14.PubMedCrossRef
81.
go back to reference Yun CC, Chen Y, Lang F. Glucocorticoid activation of Na(+)/H(+) exchanger isoform 3 revisited. The roles of SGK1 and NHERF2. J Biol Chem. 2002;277:7676–83.PubMedCrossRef Yun CC, Chen Y, Lang F. Glucocorticoid activation of Na(+)/H(+) exchanger isoform 3 revisited. The roles of SGK1 and NHERF2. J Biol Chem. 2002;277:7676–83.PubMedCrossRef
82.
go back to reference Wang D, Sun H, Lang F, Yun CC. Activation of NHE3 by dexamethasone requires phosphorylation of NHE3 at Ser663 by SGK1. Am J Physiol Cell Physiol. 2005;289:C802–10.PubMedCrossRef Wang D, Sun H, Lang F, Yun CC. Activation of NHE3 by dexamethasone requires phosphorylation of NHE3 at Ser663 by SGK1. Am J Physiol Cell Physiol. 2005;289:C802–10.PubMedCrossRef
83.
go back to reference Rauz S, Walker EA, Murray PI, Stewart PM. Expression and distribution of the serum and glucocorticoid regulated kinase and the epithelial sodium channel subunits in the human cornea. Exp Eye Res. 2003;77:101–8.PubMedCrossRef Rauz S, Walker EA, Murray PI, Stewart PM. Expression and distribution of the serum and glucocorticoid regulated kinase and the epithelial sodium channel subunits in the human cornea. Exp Eye Res. 2003;77:101–8.PubMedCrossRef
84.
go back to reference Rauz S, Walker EA, Hughes SV, Coca-Prados M, Hewison M, Murray PI, et al. Serum- and glucocorticoid-regulated kinase isoform-1 and epithelial sodium channel subunits in human ocular ciliary epithelium. Invest Ophthalmol Vis Sci. 2003;44:1643–51.PubMedCrossRef Rauz S, Walker EA, Hughes SV, Coca-Prados M, Hewison M, Murray PI, et al. Serum- and glucocorticoid-regulated kinase isoform-1 and epithelial sodium channel subunits in human ocular ciliary epithelium. Invest Ophthalmol Vis Sci. 2003;44:1643–51.PubMedCrossRef
85.
go back to reference Pondugula SR, Raveendran NN, Ergonul Z, Deng Y, Chen J, Sanneman JD, et al. Glucocorticoid regulation of genes in the amiloride-sensitive sodium transport pathway by semicircular canal duct epithelium of neonatal rat. Physiol Genomics. 2006;24:114–23.PubMed Pondugula SR, Raveendran NN, Ergonul Z, Deng Y, Chen J, Sanneman JD, et al. Glucocorticoid regulation of genes in the amiloride-sensitive sodium transport pathway by semicircular canal duct epithelium of neonatal rat. Physiol Genomics. 2006;24:114–23.PubMed
86.
go back to reference Schlingmann KP, Konrad M, Jeck N, Waldegger P, Reinalter SC, Holder M, et al. Salt wasting and deafness resulting from mutations in two chloride channels. N Engl J Med. 2004;350:1314–9.PubMedCrossRef Schlingmann KP, Konrad M, Jeck N, Waldegger P, Reinalter SC, Holder M, et al. Salt wasting and deafness resulting from mutations in two chloride channels. N Engl J Med. 2004;350:1314–9.PubMedCrossRef
87.
go back to reference Wangemann P. K(+) cycling and its regulation in the cochlea and the vestibular labyrinth. Audiol Neurootol. 2002;7:199–205.PubMedCrossRef Wangemann P. K(+) cycling and its regulation in the cochlea and the vestibular labyrinth. Audiol Neurootol. 2002;7:199–205.PubMedCrossRef
88.
go back to reference Klip A, Ramlal T, Cragoe EJ Jr. Insulin-induced cytoplasmic alkalinization and glucose transport in muscle cells. Am J Physiol. 1986;250:C720–8.PubMed Klip A, Ramlal T, Cragoe EJ Jr. Insulin-induced cytoplasmic alkalinization and glucose transport in muscle cells. Am J Physiol. 1986;250:C720–8.PubMed
89.
go back to reference Zhao H, Hyde R, Hundal HS. Signalling mechanisms underlying the rapid and additive stimulation of NKCC activity by insulin and hypertonicity in rat L6 skeletal muscle cells. J Physiol. 2004;560:123–36.PubMedCrossRef Zhao H, Hyde R, Hundal HS. Signalling mechanisms underlying the rapid and additive stimulation of NKCC activity by insulin and hypertonicity in rat L6 skeletal muscle cells. J Physiol. 2004;560:123–36.PubMedCrossRef
90.
go back to reference Cleland SJ, Petrie JR, Ueda S, Elliott HL, Connell JM. Insulin as a vascular hormone: implications for the pathophysiology of cardiovascular disease. Clin Exp Pharmacol Physiol. 1998;25:175–84.PubMedCrossRef Cleland SJ, Petrie JR, Ueda S, Elliott HL, Connell JM. Insulin as a vascular hormone: implications for the pathophysiology of cardiovascular disease. Clin Exp Pharmacol Physiol. 1998;25:175–84.PubMedCrossRef
91.
92.
go back to reference Bobulescu IA, Dubree M, Zhang J, McLeroy P, Moe OW. Effect of renal lipid accumulation on proximal tubule Na+/H+ exchange and ammonium secretion. Am J Physiol Renal Physiol. 2008;294:F1315–22.PubMedCrossRef Bobulescu IA, Dubree M, Zhang J, McLeroy P, Moe OW. Effect of renal lipid accumulation on proximal tubule Na+/H+ exchange and ammonium secretion. Am J Physiol Renal Physiol. 2008;294:F1315–22.PubMedCrossRef
93.
go back to reference Sjaastad MD, Zettl KS, Parry G, Firestone GL, Machen TE. Hormonal regulation of the polarized function and distribution of Na/H exchange and Na/HCO3 cotransport in cultured mammary epithelial cells. J Cell Biol. 1993;122:589–600.PubMedCrossRef Sjaastad MD, Zettl KS, Parry G, Firestone GL, Machen TE. Hormonal regulation of the polarized function and distribution of Na/H exchange and Na/HCO3 cotransport in cultured mammary epithelial cells. J Cell Biol. 1993;122:589–600.PubMedCrossRef
94.
go back to reference Ueda-Nishimura T, Niisato N, Miyazaki H, Naito Y, Yoshida N, Yoshikawa T, et al. Synergic action of insulin and genistein on Na+/K+/2Cl− cotransporter in renal epithelium. Biochem Biophys Res Commun. 2005;332:1042–52.PubMedCrossRef Ueda-Nishimura T, Niisato N, Miyazaki H, Naito Y, Yoshida N, Yoshikawa T, et al. Synergic action of insulin and genistein on Na+/K+/2Cl cotransporter in renal epithelium. Biochem Biophys Res Commun. 2005;332:1042–52.PubMedCrossRef
95.
go back to reference Longo N. Insulin stimulates the Na+, K(+)-ATPase and the Na+/K+/Cl− cotransporter of human fibroblasts. Biochim Biophys Acta. 1996;1281:38–44.PubMedCrossRef Longo N. Insulin stimulates the Na+, K(+)-ATPase and the Na+/K+/Cl cotransporter of human fibroblasts. Biochim Biophys Acta. 1996;1281:38–44.PubMedCrossRef
96.
go back to reference Pouyssegur J, Chambard JC, Franchi A, Paris S, Obberghen-Schilling E. Growth factor activation of an amiloride-sensitive Na+/H+ exchange system in quiescent fibroblasts: coupling to ribosomal protein S6 phosphorylation. Proc Natl Acad Sci USA. 1982;79:3935–9.PubMedCrossRef Pouyssegur J, Chambard JC, Franchi A, Paris S, Obberghen-Schilling E. Growth factor activation of an amiloride-sensitive Na+/H+ exchange system in quiescent fibroblasts: coupling to ribosomal protein S6 phosphorylation. Proc Natl Acad Sci USA. 1982;79:3935–9.PubMedCrossRef
97.
go back to reference Tupper JT, Smith JW. Growth factor regulation of membrane transport in human fibroblasts and its relationship to stimulation of DNA synthesis. J Cell Physiol. 1985;125:443–8.PubMedCrossRef Tupper JT, Smith JW. Growth factor regulation of membrane transport in human fibroblasts and its relationship to stimulation of DNA synthesis. J Cell Physiol. 1985;125:443–8.PubMedCrossRef
98.
go back to reference Sargeant RJ, Liu Z, Klip A. Action of insulin on Na(+)-K(+)-ATPase and the Na(+)-K(+)-2Cl− cotransporter in 3T3–L1 adipocytes. Am J Physiol. 1995;269:C217–25.PubMed Sargeant RJ, Liu Z, Klip A. Action of insulin on Na(+)-K(+)-ATPase and the Na(+)-K(+)-2Cl cotransporter in 3T3–L1 adipocytes. Am J Physiol. 1995;269:C217–25.PubMed
99.
go back to reference Ceolotto G, Conlin P, Clari G, Semplicini A, Canessa M. Protein kinase C and insulin regulation of red blood cell Na+/H+ exchange. Am J Physiol. 1997;272:C818–26.PubMed Ceolotto G, Conlin P, Clari G, Semplicini A, Canessa M. Protein kinase C and insulin regulation of red blood cell Na+/H+ exchange. Am J Physiol. 1997;272:C818–26.PubMed
100.
go back to reference Kaloyianni M, Bourikas D, Koliakos G. The effect of insulin on Na+-H+ antiport activity of obese and normal subjects erythrocytes. Cell Physiol Biochem. 2001;11:253–8.PubMedCrossRef Kaloyianni M, Bourikas D, Koliakos G. The effect of insulin on Na+-H+ antiport activity of obese and normal subjects erythrocytes. Cell Physiol Biochem. 2001;11:253–8.PubMedCrossRef
101.
go back to reference Romero JR, Rivera A, Conlin PR. Red blood cell Na+/H+ exchange activity is insulin resistant in hypertensive patients. Clin Exp Hypertens. 2002;24:277–87.PubMedCrossRef Romero JR, Rivera A, Conlin PR. Red blood cell Na+/H+ exchange activity is insulin resistant in hypertensive patients. Clin Exp Hypertens. 2002;24:277–87.PubMedCrossRef
102.
go back to reference Sauvage M, Maziere P, Fathallah H, Giraud F. Insulin stimulates NHE1 activity by sequential activation of phosphatidylinositol 3-kinase and protein kinase C zeta in human erythrocytes. Eur J Biochem. 2000;267:955–62.PubMedCrossRef Sauvage M, Maziere P, Fathallah H, Giraud F. Insulin stimulates NHE1 activity by sequential activation of phosphatidylinositol 3-kinase and protein kinase C zeta in human erythrocytes. Eur J Biochem. 2000;267:955–62.PubMedCrossRef
103.
go back to reference Haussinger D, Hallbrucker C, vom DS, Lang F, Gerok W. Cell swelling inhibits proteolysis in perfused rat liver. Biochem J. 1990;272:239–42.PubMed Haussinger D, Hallbrucker C, vom DS, Lang F, Gerok W. Cell swelling inhibits proteolysis in perfused rat liver. Biochem J. 1990;272:239–42.PubMed
104.
go back to reference Benziane B, Chibalin AV. Frontiers: skeletal muscle sodium pump regulation: a translocation paradigm. Am J Physiol Endocrinol Metab. 2008;295:E553–8.PubMedCrossRef Benziane B, Chibalin AV. Frontiers: skeletal muscle sodium pump regulation: a translocation paradigm. Am J Physiol Endocrinol Metab. 2008;295:E553–8.PubMedCrossRef
105.
go back to reference Ewart HS, Klip A. Hormonal regulation of the Na(+)-K(+)-ATPase: mechanisms underlying rapid and sustained changes in pump activity. Am J Physiol. 1995;269:C295–311.PubMed Ewart HS, Klip A. Hormonal regulation of the Na(+)-K(+)-ATPase: mechanisms underlying rapid and sustained changes in pump activity. Am J Physiol. 1995;269:C295–311.PubMed
106.
go back to reference Graf J, Haussinger D. Ion transport in hepatocytes: mechanisms and correlations to cell volume, hormone actions and metabolism. J Hepatol. 1996;24(Suppl 1):53–77.PubMed Graf J, Haussinger D. Ion transport in hepatocytes: mechanisms and correlations to cell volume, hormone actions and metabolism. J Hepatol. 1996;24(Suppl 1):53–77.PubMed
107.
go back to reference McDonough AA, Youn JH. Role of muscle in regulating extracellular [K+]. Semin Nephrol. 2005;25:335–42.PubMedCrossRef McDonough AA, Youn JH. Role of muscle in regulating extracellular [K+]. Semin Nephrol. 2005;25:335–42.PubMedCrossRef
108.
go back to reference Sweeney G, Klip A. Regulation of the Na+/K+-ATPase by insulin: why and how? Mol Cell Biochem. 1998;182:121–33.PubMedCrossRef Sweeney G, Klip A. Regulation of the Na+/K+-ATPase by insulin: why and how? Mol Cell Biochem. 1998;182:121–33.PubMedCrossRef
109.
go back to reference Sweeney G, Klip A. Mechanisms and consequences of Na+, K+-pump regulation by insulin and leptin. Cell Mol Biol (Noisy -le-grand). 2001;47:363–72. Sweeney G, Klip A. Mechanisms and consequences of Na+, K+-pump regulation by insulin and leptin. Cell Mol Biol (Noisy -le-grand). 2001;47:363–72.
110.
go back to reference Rosenbloom AL, Hanas R. Diabetic ketoacidosis (DKA): treatment guidelines. Clin Pediatr (Phila). 1996;35:261–6.CrossRef Rosenbloom AL, Hanas R. Diabetic ketoacidosis (DKA): treatment guidelines. Clin Pediatr (Phila). 1996;35:261–6.CrossRef
111.
go back to reference Bia MJ, Tyler K, DeFronzo R. The effect of dexamethasone on renal potassium excretion and acute potassium tolerance. Endocrinology. 1983;113:1690–6.PubMedCrossRef Bia MJ, Tyler K, DeFronzo R. The effect of dexamethasone on renal potassium excretion and acute potassium tolerance. Endocrinology. 1983;113:1690–6.PubMedCrossRef
112.
go back to reference Boini KM, Graf D, Kuhl D, Haussinger D, Lang F. SGK1 dependence of insulin induced hypokalemia. Pflugers Arch. 2009;457:955–61.PubMedCrossRef Boini KM, Graf D, Kuhl D, Haussinger D, Lang F. SGK1 dependence of insulin induced hypokalemia. Pflugers Arch. 2009;457:955–61.PubMedCrossRef
113.
go back to reference Klingel K, Warntges S, Bock J, Wagner CA, Sauter M, Waldegger S, et al. Expression of cell volume-regulated kinase H-Sgk in pancreatic tissue. Am J Physiol Gastrointest Liver Physiol. 2000;279:G998–1002.PubMed Klingel K, Warntges S, Bock J, Wagner CA, Sauter M, Waldegger S, et al. Expression of cell volume-regulated kinase H-Sgk in pancreatic tissue. Am J Physiol Gastrointest Liver Physiol. 2000;279:G998–1002.PubMed
114.
go back to reference Kawakami T, Galli SJ. Regulation of mast-cell and basophil function and survival by IgE. Nat Rev Immunol. 2002;2:773–86.PubMedCrossRef Kawakami T, Galli SJ. Regulation of mast-cell and basophil function and survival by IgE. Nat Rev Immunol. 2002;2:773–86.PubMedCrossRef
115.
go back to reference Kawakami T, Kitaura J. Mast cell survival and activation by IgE in the absence of antigen: a consideration of the biologic mechanisms and relevance. J Immunol. 2005;175:4167–73.PubMed Kawakami T, Kitaura J. Mast cell survival and activation by IgE in the absence of antigen: a consideration of the biologic mechanisms and relevance. J Immunol. 2005;175:4167–73.PubMed
116.
go back to reference Bradding P, Okayama Y, Kambe N, Saito H. Ion channel gene expression in human lung, skin, and cord blood-derived mast cells. J Leukoc Biol. 2003;73:614–20.PubMedCrossRef Bradding P, Okayama Y, Kambe N, Saito H. Ion channel gene expression in human lung, skin, and cord blood-derived mast cells. J Leukoc Biol. 2003;73:614–20.PubMedCrossRef
118.
go back to reference Duffy SM, Cruse G, Lawley WJ, Bradding P. Beta2-adrenoceptor regulation of the K+ channel ikca1 in human mast cells. FASEB J. 2005;19:1006–8.PubMed Duffy SM, Cruse G, Lawley WJ, Bradding P. Beta2-adrenoceptor regulation of the K+ channel ikca1 in human mast cells. FASEB J. 2005;19:1006–8.PubMed
119.
go back to reference Mark DS, Berger P, Cruse G, Yang W, Bolton SJ, Bradding P. The K+ channel IKCA1 potentiates Ca2+ influx and degranulation in human lung mast cells. J Allergy Clin Immunol. 2004;114:66–72.CrossRef Mark DS, Berger P, Cruse G, Yang W, Bolton SJ, Bradding P. The K+ channel IKCA1 potentiates Ca2+ influx and degranulation in human lung mast cells. J Allergy Clin Immunol. 2004;114:66–72.CrossRef
120.
go back to reference Shumilina E, Lam RS, Wolbing F, Matzner N, Zemtsova IM, Sobiesiak M, et al. Blunted IgE-mediated activation of mast cells in mice lacking the Ca2+-activated K+ channel KCa3.1. J Immunol. 2008;180:8040–7.PubMed Shumilina E, Lam RS, Wolbing F, Matzner N, Zemtsova IM, Sobiesiak M, et al. Blunted IgE-mediated activation of mast cells in mice lacking the Ca2+-activated K+ channel KCa3.1. J Immunol. 2008;180:8040–7.PubMed
121.
go back to reference Lam RS, Shumilina E, Matzner N, Zemtsova IM, Sobiesiak M, Lang C, et al. Phosphatidylinositol-3-kinase regulates mast cell ion channel activity. Cell Physiol Biochem. 2008;22:169–76.PubMedCrossRef Lam RS, Shumilina E, Matzner N, Zemtsova IM, Sobiesiak M, Lang C, et al. Phosphatidylinositol-3-kinase regulates mast cell ion channel activity. Cell Physiol Biochem. 2008;22:169–76.PubMedCrossRef
122.
go back to reference Sobiesiak M, Shumilina E, Lam RS, Wolbing F, Matzner N, Kaesler S, et al. Impaired mast cell activation in gene-targeted mice lacking the serum- and glucocorticoid-inducible kinase SGK1. J Immunol. 2009;183:4395–402.PubMedCrossRef Sobiesiak M, Shumilina E, Lam RS, Wolbing F, Matzner N, Kaesler S, et al. Impaired mast cell activation in gene-targeted mice lacking the serum- and glucocorticoid-inducible kinase SGK1. J Immunol. 2009;183:4395–402.PubMedCrossRef
123.
go back to reference Vincent GM. The molecular genetics of the long QT syndrome: genes causing fainting and sudden death. Annu Rev Med. 1998;49:263–74.PubMedCrossRef Vincent GM. The molecular genetics of the long QT syndrome: genes causing fainting and sudden death. Annu Rev Med. 1998;49:263–74.PubMedCrossRef
124.
go back to reference Busjahn A, Seebohm G, Maier G, Toliat MR, Nurnberg P, Aydin A, et al. Association of the serum and glucocorticoid regulated kinase (Sgk1) gene with QT interval. Cell Physiol Biochem. 2004;14:135–42.PubMedCrossRef Busjahn A, Seebohm G, Maier G, Toliat MR, Nurnberg P, Aydin A, et al. Association of the serum and glucocorticoid regulated kinase (Sgk1) gene with QT interval. Cell Physiol Biochem. 2004;14:135–42.PubMedCrossRef
125.
go back to reference Seebohm G, Strutz-Seebohm N, Birkin R, Dell G, Bucci C, Spinosa MR, et al. Regulation of endocytic recycling of KCNQ1/KCNE1 potassium channels. Circ Res. 2007;100:686–92.PubMedCrossRef Seebohm G, Strutz-Seebohm N, Birkin R, Dell G, Bucci C, Spinosa MR, et al. Regulation of endocytic recycling of KCNQ1/KCNE1 potassium channels. Circ Res. 2007;100:686–92.PubMedCrossRef
126.
go back to reference Busjahn A, Aydin A, Uhlmann R, Krasko C, Bahring S, Szelestei T, et al. Serum- and glucocorticoid-regulated kinase (SGK1) gene and blood pressure. Hypertension. 2002;40:256–60.PubMedCrossRef Busjahn A, Aydin A, Uhlmann R, Krasko C, Bahring S, Szelestei T, et al. Serum- and glucocorticoid-regulated kinase (SGK1) gene and blood pressure. Hypertension. 2002;40:256–60.PubMedCrossRef
127.
go back to reference Busjahn A, Luft FC. Twin studies in the analysis of minor physiological differences between individuals. Cell Physiol Biochem. 2003;13:51–8.PubMedCrossRef Busjahn A, Luft FC. Twin studies in the analysis of minor physiological differences between individuals. Cell Physiol Biochem. 2003;13:51–8.PubMedCrossRef
128.
go back to reference Schwab M, Lupescu A, Mota M, Mota E, Frey A, Simon P, et al. Association of SGK1 gene polymorphisms with type 2 diabetes. Cell Physiol Biochem. 2008;21:151–60.PubMedCrossRef Schwab M, Lupescu A, Mota M, Mota E, Frey A, Simon P, et al. Association of SGK1 gene polymorphisms with type 2 diabetes. Cell Physiol Biochem. 2008;21:151–60.PubMedCrossRef
129.
go back to reference Maier G, Palmada M, Rajamanickam J, Shumilina E, Bohmer C, Lang F. Upregulation of HERG channels by the serum and glucocorticoid inducible kinase isoform SGK3. Cell Physiol Biochem. 2006;18:177–86.PubMedCrossRef Maier G, Palmada M, Rajamanickam J, Shumilina E, Bohmer C, Lang F. Upregulation of HERG channels by the serum and glucocorticoid inducible kinase isoform SGK3. Cell Physiol Biochem. 2006;18:177–86.PubMedCrossRef
130.
go back to reference Charlier C, Singh NA, Ryan SG, Lewis TB, Reus BE, Leach RJ, et al. A pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family. Nat Genet. 1998;18:53–5.PubMedCrossRef Charlier C, Singh NA, Ryan SG, Lewis TB, Reus BE, Leach RJ, et al. A pore mutation in a novel KQT-like potassium channel gene in an idiopathic epilepsy family. Nat Genet. 1998;18:53–5.PubMedCrossRef
131.
go back to reference Eunson LH, Rea R, Zuberi SM, Youroukos S, Panayiotopoulos CP, Liguori R, et al. Clinical, genetic, and expression studies of mutations in the potassium channel gene KCNA1 reveal new phenotypic variability. Ann Neurol. 2000;48:647–56.PubMedCrossRef Eunson LH, Rea R, Zuberi SM, Youroukos S, Panayiotopoulos CP, Liguori R, et al. Clinical, genetic, and expression studies of mutations in the potassium channel gene KCNA1 reveal new phenotypic variability. Ann Neurol. 2000;48:647–56.PubMedCrossRef
132.
go back to reference Lau D, Vega-Saenz de Miera EC, Contreras D, Ozaita A, Harvey M, Chow A, et al. Impaired fast-spiking, suppressed cortical inhibition, and increased susceptibility to seizures in mice lacking Kv3.2 K+ channel proteins. J Neurosci. 2000;20:9071–85. Lau D, Vega-Saenz de Miera EC, Contreras D, Ozaita A, Harvey M, Chow A, et al. Impaired fast-spiking, suppressed cortical inhibition, and increased susceptibility to seizures in mice lacking Kv3.2 K+ channel proteins. J Neurosci. 2000;20:9071–85.
133.
134.
go back to reference Pongs O. Voltage-gated potassium channels: from hyperexcitability to excitement. FEBS Lett. 1999;452:31–5.PubMedCrossRef Pongs O. Voltage-gated potassium channels: from hyperexcitability to excitement. FEBS Lett. 1999;452:31–5.PubMedCrossRef
135.
go back to reference Singh NA, Charlier C, Stauffer D, DuPont BR, Leach RJ, Melis R, et al. A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns. Nat Genet. 1998;18:25–9.PubMedCrossRef Singh NA, Charlier C, Stauffer D, DuPont BR, Leach RJ, Melis R, et al. A novel potassium channel gene, KCNQ2, is mutated in an inherited epilepsy of newborns. Nat Genet. 1998;18:25–9.PubMedCrossRef
136.
go back to reference Smart SL, Lopantsev V, Zhang CL, Robbins CA, Wang H, Chiu SY, et al. Deletion of the K(V)1.1 potassium channel causes epilepsy in mice. Neuron. 1998;20:809–19.PubMedCrossRef Smart SL, Lopantsev V, Zhang CL, Robbins CA, Wang H, Chiu SY, et al. Deletion of the K(V)1.1 potassium channel causes epilepsy in mice. Neuron. 1998;20:809–19.PubMedCrossRef
137.
go back to reference Strutz-Seebohm N, Seebohm G, Shumilina E, Mack AF, Wagner HJ, Lampert A, et al. Glucocorticoid adrenal steroids and glucocorticoid-inducible kinase isoforms in the regulation of GluR6 expression. J Physiol. 2005;565:391–401.PubMedCrossRef Strutz-Seebohm N, Seebohm G, Shumilina E, Mack AF, Wagner HJ, Lampert A, et al. Glucocorticoid adrenal steroids and glucocorticoid-inducible kinase isoforms in the regulation of GluR6 expression. J Physiol. 2005;565:391–401.PubMedCrossRef
138.
go back to reference Contractor A, Swanson GT, Sailer A, O’Gorman S, Heinemann SF. Identification of the kainate receptor subunits underlying modulation of excitatory synaptic transmission in the CA3 region of the hippocampus. J Neurosci. 2000;20:8269–78.PubMed Contractor A, Swanson GT, Sailer A, O’Gorman S, Heinemann SF. Identification of the kainate receptor subunits underlying modulation of excitatory synaptic transmission in the CA3 region of the hippocampus. J Neurosci. 2000;20:8269–78.PubMed
139.
go back to reference Contractor A, Swanson G, Heinemann SF. Kainate receptors are involved in short- and long-term plasticity at mossy fiber synapses in the hippocampus. Neuron. 2001;29:209–16.PubMedCrossRef Contractor A, Swanson G, Heinemann SF. Kainate receptors are involved in short- and long-term plasticity at mossy fiber synapses in the hippocampus. Neuron. 2001;29:209–16.PubMedCrossRef
140.
141.
go back to reference Mulle C, Sailer A, Perez-Otano I, Dickinson-Anson H, Castillo PE, Bureau I, et al. Altered synaptic physiology and reduced susceptibility to kainate-induced seizures in GluR6-deficient mice. Nature. 1998;392:601–5.PubMedCrossRef Mulle C, Sailer A, Perez-Otano I, Dickinson-Anson H, Castillo PE, Bureau I, et al. Altered synaptic physiology and reduced susceptibility to kainate-induced seizures in GluR6-deficient mice. Nature. 1998;392:601–5.PubMedCrossRef
142.
go back to reference Kaufer D, Ogle WO, Pincus ZS, Clark KL, Nicholas AC, Dinkel KM, et al. Restructuring the neuronal stress response with anti-glucocorticoid gene delivery. Nat Neurosci. 2004;7:947–53.PubMedCrossRef Kaufer D, Ogle WO, Pincus ZS, Clark KL, Nicholas AC, Dinkel KM, et al. Restructuring the neuronal stress response with anti-glucocorticoid gene delivery. Nat Neurosci. 2004;7:947–53.PubMedCrossRef
143.
go back to reference McEwen BS, Albeck D, Cameron H, Chao HM, Gould E, Hastings N, et al. Stress and the brain: a paradoxical role for adrenal steroids. Vitam Horm. 1995;51:371–402.PubMedCrossRef McEwen BS, Albeck D, Cameron H, Chao HM, Gould E, Hastings N, et al. Stress and the brain: a paradoxical role for adrenal steroids. Vitam Horm. 1995;51:371–402.PubMedCrossRef
144.
go back to reference Reagan LP, McEwen BS. Controversies surrounding glucocorticoid-mediated cell death in the hippocampus. J Chem Neuroanat. 1997;13:149–67.PubMedCrossRef Reagan LP, McEwen BS. Controversies surrounding glucocorticoid-mediated cell death in the hippocampus. J Chem Neuroanat. 1997;13:149–67.PubMedCrossRef
145.
go back to reference Lee CT, Ma YL, Lee EH. Serum- and glucocorticoid-inducible kinase1 enhances contextual fear memory formation through down-regulation of the expression of Hes5. J Neurochem. 2007;100:1531–42.PubMed Lee CT, Ma YL, Lee EH. Serum- and glucocorticoid-inducible kinase1 enhances contextual fear memory formation through down-regulation of the expression of Hes5. J Neurochem. 2007;100:1531–42.PubMed
146.
go back to reference von Hertzen LS, Giese KP. Memory reconsolidation engages only a subset of immediate-early genes induced during consolidation. J Neurosci. 2005;25:1935–42.CrossRef von Hertzen LS, Giese KP. Memory reconsolidation engages only a subset of immediate-early genes induced during consolidation. J Neurosci. 2005;25:1935–42.CrossRef
147.
go back to reference Koya E, Spijker S, Homberg JR, Voorn P, Schoffelmeer AN, De Vries TJ, et al. Molecular reactivity of mesocorticolimbic brain areas of high and low grooming rats after elevated plus maze exposure. Brain Res Mol Brain Res. 2005;137:184–92.PubMedCrossRef Koya E, Spijker S, Homberg JR, Voorn P, Schoffelmeer AN, De Vries TJ, et al. Molecular reactivity of mesocorticolimbic brain areas of high and low grooming rats after elevated plus maze exposure. Brain Res Mol Brain Res. 2005;137:184–92.PubMedCrossRef
148.
go back to reference Lee EH, Hsu WL, Ma YL, Lee PJ, Chao CC. Enrichment enhances the expression of Sgk, a glucocorticoid-induced gene, and facilitates spatial learning through glutamate AMPA receptor mediation. Eur J Neurosci. 2003;18:2842–52.PubMedCrossRef Lee EH, Hsu WL, Ma YL, Lee PJ, Chao CC. Enrichment enhances the expression of Sgk, a glucocorticoid-induced gene, and facilitates spatial learning through glutamate AMPA receptor mediation. Eur J Neurosci. 2003;18:2842–52.PubMedCrossRef
149.
go back to reference Chao CC, Ma YL, Lee EH. Protein kinase CK2 impairs spatial memory formation through differential cross talk with PI-3 kinase signaling: activation of Akt and inactivation of SGK1. J Neurosci. 2007;27:6243–8.PubMedCrossRef Chao CC, Ma YL, Lee EH. Protein kinase CK2 impairs spatial memory formation through differential cross talk with PI-3 kinase signaling: activation of Akt and inactivation of SGK1. J Neurosci. 2007;27:6243–8.PubMedCrossRef
150.
go back to reference David S, Stegenga SL, Hu P, Xiong G, Kerr E, Becker KB, et al. Expression of serum- and glucocorticoid-inducible kinase is regulated in an experience-dependent manner and can cause dendrite growth. J Neurosci. 2005;25:7048–53.PubMedCrossRef David S, Stegenga SL, Hu P, Xiong G, Kerr E, Becker KB, et al. Expression of serum- and glucocorticoid-inducible kinase is regulated in an experience-dependent manner and can cause dendrite growth. J Neurosci. 2005;25:7048–53.PubMedCrossRef
151.
go back to reference Yang YC, Lin CH, Lee EH. Serum- and glucocorticoid-inducible kinase 1 (SGK1) increases neurite formation through microtubule depolymerization by SGK1 and by SGK1 phosphorylation of tau. Mol Cell Biol. 2006;26:8357–70.PubMedCrossRef Yang YC, Lin CH, Lee EH. Serum- and glucocorticoid-inducible kinase 1 (SGK1) increases neurite formation through microtubule depolymerization by SGK1 and by SGK1 phosphorylation of tau. Mol Cell Biol. 2006;26:8357–70.PubMedCrossRef
152.
go back to reference Tsai KJ, Chen SK, Ma YL, Hsu WL, Lee EH. Sgk, a primary glucocorticoid-induced gene, facilitates memory consolidation of spatial learning in rats. Proc Natl Acad Sci USA. 2002;99:3990–5.PubMedCrossRef Tsai KJ, Chen SK, Ma YL, Hsu WL, Lee EH. Sgk, a primary glucocorticoid-induced gene, facilitates memory consolidation of spatial learning in rats. Proc Natl Acad Sci USA. 2002;99:3990–5.PubMedCrossRef
153.
go back to reference Buse P, Tran SH, Luther E, Phu PT, Aponte GW, Firestone GL. Cell cycle and hormonal control of nuclear-cytoplasmic localization of the serum- and glucocorticoid-inducible protein kinase, Sgk, in mammary tumor cells. A novel convergence point of anti-proliferative and proliferative cell signaling pathways. J Biol Chem. 1999;274:7253–63.PubMedCrossRef Buse P, Tran SH, Luther E, Phu PT, Aponte GW, Firestone GL. Cell cycle and hormonal control of nuclear-cytoplasmic localization of the serum- and glucocorticoid-inducible protein kinase, Sgk, in mammary tumor cells. A novel convergence point of anti-proliferative and proliferative cell signaling pathways. J Biol Chem. 1999;274:7253–63.PubMedCrossRef
154.
go back to reference Amato R, Menniti M, Agosti V, Boito R, Costa N, Bond HM, et al. IL-2 signals through sgk1 and inhibits proliferation and apoptosis in kidney cancer cells. J Mol Med. 2007;85:707–21.PubMedCrossRef Amato R, Menniti M, Agosti V, Boito R, Costa N, Bond HM, et al. IL-2 signals through sgk1 and inhibits proliferation and apoptosis in kidney cancer cells. J Mol Med. 2007;85:707–21.PubMedCrossRef
155.
go back to reference Brunet A, Park J, Tran H, Hu LS, Hemmings BA, Greenberg ME. Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol. 2001;21:952–65.PubMedCrossRef Brunet A, Park J, Tran H, Hu LS, Hemmings BA, Greenberg ME. Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a). Mol Cell Biol. 2001;21:952–65.PubMedCrossRef
156.
go back to reference Shelly C, Herrera R. Activation of SGK1 by HGF, Rac1 and integrin-mediated cell adhesion in MDCK cells: PI-3K-dependent and -independent pathways. J Cell Sci. 2002;115:1985–93.PubMed Shelly C, Herrera R. Activation of SGK1 by HGF, Rac1 and integrin-mediated cell adhesion in MDCK cells: PI-3K-dependent and -independent pathways. J Cell Sci. 2002;115:1985–93.PubMed
157.
go back to reference Zhang L, Cui R, Cheng X, Du J. Antiapoptotic effect of serum and glucocorticoid-inducible protein kinase is mediated by novel mechanism activating I{Kappa}B kinase. Cancer Res. 2005;65:457–64.PubMed Zhang L, Cui R, Cheng X, Du J. Antiapoptotic effect of serum and glucocorticoid-inducible protein kinase is mediated by novel mechanism activating I{Kappa}B kinase. Cancer Res. 2005;65:457–64.PubMed
158.
go back to reference Lang F, Busch GL, Ritter M, Völkl H, Waldegger S, Gulbins E, et al. Functional significance of cell volume regulatory mechanisms. Physiol Rev. 1998;78:247–306.PubMed Lang F, Busch GL, Ritter M, Völkl H, Waldegger S, Gulbins E, et al. Functional significance of cell volume regulatory mechanisms. Physiol Rev. 1998;78:247–306.PubMed
159.
go back to reference Lewis RS, Cahalan MD. Subset-specific expression of potassium channels in developing murine T lymphocytes. Science. 1988;239:771–5.PubMedCrossRef Lewis RS, Cahalan MD. Subset-specific expression of potassium channels in developing murine T lymphocytes. Science. 1988;239:771–5.PubMedCrossRef
160.
go back to reference Ouadid-Ahidouch H, Chaussade F, Roudbaraki M, Slomianny C, Dewailly E, Delcourt P, et al. KV1.1 K(+) channels identification in human breast carcinoma cells: involvement in cell proliferation. Biochem Biophys Res Commun. 2000;278:272–7.PubMedCrossRef Ouadid-Ahidouch H, Chaussade F, Roudbaraki M, Slomianny C, Dewailly E, Delcourt P, et al. KV1.1 K(+) channels identification in human breast carcinoma cells: involvement in cell proliferation. Biochem Biophys Res Commun. 2000;278:272–7.PubMedCrossRef
161.
go back to reference Ritter M, Wöll E. Modification of cellular ion transport by the Ha-Ras oncogene: steps towards malignant transformation. Cell Physiol Biochem. 1996;6:245–70.CrossRef Ritter M, Wöll E. Modification of cellular ion transport by the Ha-Ras oncogene: steps towards malignant transformation. Cell Physiol Biochem. 1996;6:245–70.CrossRef
162.
go back to reference Sobko A, Peretz A, Shirihai O, Etkin S, Cherepanova V, Dagan D, et al. Heteromultimeric delayed-rectifier K+ channels in schwann cells: developmental expression and role in cell proliferation. J Neurosci. 1998;18:10398–408.PubMed Sobko A, Peretz A, Shirihai O, Etkin S, Cherepanova V, Dagan D, et al. Heteromultimeric delayed-rectifier K+ channels in schwann cells: developmental expression and role in cell proliferation. J Neurosci. 1998;18:10398–408.PubMed
Metadata
Title
Serum- and glucocorticoid-inducible kinase 1 in the regulation of renal and extrarenal potassium transport
Authors
Florian Lang
Volker Vallon
Publication date
01-02-2012
Publisher
Springer Japan
Published in
Clinical and Experimental Nephrology / Issue 1/2012
Print ISSN: 1342-1751
Electronic ISSN: 1437-7799
DOI
https://doi.org/10.1007/s10157-011-0488-z

Other articles of this Issue 1/2012

Clinical and Experimental Nephrology 1/2012 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine