Skip to main content
Top
Published in: Pediatric Nephrology 7/2017

01-07-2017 | Review

Potassium: friend or foe?

Author: Aylin R. Rodan

Published in: Pediatric Nephrology | Issue 7/2017

Login to get access

Abstract

The kidney plays an essential role in maintaining homeostasis of ion concentrations in the blood. Because the concentration gradient of potassium across the cell membrane is a key determinant of the membrane potential of cells, even small deviations in serum potassium level from the normal setpoint can lead to severe muscle dysfunction, resulting in respiratory failure and cardiac arrest. Less severe hypo- and hyperkalemia are also associated with morbidity and mortality across various patient populations. In addition, deficiencies in potassium intake have been associated with hypertension and adverse cardiovascular and renal outcomes, likely due in part to the interrelated handling of sodium and potassium by the kidney. Here, data on the beneficial effects of potassium on blood pressure and cardiovascular and renal outcomes will be reviewed, along with the physiological basis for these effects. In some patient populations, however, potassium excess is deleterious. Risk factors for the development of hyperkalemia will be reviewed, as well as the risks and benefits of existing and emerging therapies for hyperkalemia.
Literature
1.
2.
go back to reference Keith NM, Binger MW (1935) Diuretic action of potassium. J Am Med Assoc 105:1584–1591CrossRef Keith NM, Binger MW (1935) Diuretic action of potassium. J Am Med Assoc 105:1584–1591CrossRef
3.
go back to reference Barker M (1932) Edema as influenced by a low ratio of sodium to potassium intake: clinical observations. J Am Med Assoc 98:2193–2197CrossRef Barker M (1932) Edema as influenced by a low ratio of sodium to potassium intake: clinical observations. J Am Med Assoc 98:2193–2197CrossRef
4.
go back to reference Aburto NJ, Hanson S, Gutierrez H, Hooper L, Elliott P, Cappuccio FP (2013) Effect of increased potassium intake on cardiovascular risk factors and disease: systematic review and meta-analyses. BMJ 346:f1378PubMedCrossRefPubMedCentral Aburto NJ, Hanson S, Gutierrez H, Hooper L, Elliott P, Cappuccio FP (2013) Effect of increased potassium intake on cardiovascular risk factors and disease: systematic review and meta-analyses. BMJ 346:f1378PubMedCrossRefPubMedCentral
5.
go back to reference National Academy of Sciences, Institute of Medicine, Food and Nutrition Board (2005) Dietary reference intakes for water, potassium, sodium, chloride, and sulfate. The National Academies Press, Washington, DC National Academy of Sciences, Institute of Medicine, Food and Nutrition Board (2005) Dietary reference intakes for water, potassium, sodium, chloride, and sulfate. The National Academies Press, Washington, DC
6.
go back to reference Mente A, O'Donnell MJ, Rangarajan S, McQueen MJ, Poirier P, Wielgosz A, Morrison H, Li W, Wang X, Di C, Mony P, Devanath A, Rosengren A, Oguz A, Zatonska K, Yusufali AH, Lopez-Jaramillo P, Avezum A, Ismail N, Lanas F, Puoane T, Diaz R, Kelishadi R, Iqbal R, Yusuf R, Chifamba J, Khatib R, Teo K, Yusuf S, PURE Investigators (2014) Association of urinary sodium and potassium excretion with blood pressure. N Engl J Med 371:601–611PubMedCrossRef Mente A, O'Donnell MJ, Rangarajan S, McQueen MJ, Poirier P, Wielgosz A, Morrison H, Li W, Wang X, Di C, Mony P, Devanath A, Rosengren A, Oguz A, Zatonska K, Yusufali AH, Lopez-Jaramillo P, Avezum A, Ismail N, Lanas F, Puoane T, Diaz R, Kelishadi R, Iqbal R, Yusuf R, Chifamba J, Khatib R, Teo K, Yusuf S, PURE Investigators (2014) Association of urinary sodium and potassium excretion with blood pressure. N Engl J Med 371:601–611PubMedCrossRef
7.
go back to reference Cogswell ME, Zhang Z, Carriquiry AL, Gunn JP, Kuklina EV, Saydah SH, Yang Q, Moshfegh AJ (2012) Sodium and potassium intakes among US adults: NHANES 2003–2008. Am J Clin Nutr 96:647–657PubMedCrossRefPubMedCentral Cogswell ME, Zhang Z, Carriquiry AL, Gunn JP, Kuklina EV, Saydah SH, Yang Q, Moshfegh AJ (2012) Sodium and potassium intakes among US adults: NHANES 2003–2008. Am J Clin Nutr 96:647–657PubMedCrossRefPubMedCentral
8.
go back to reference O'Donnell M, Mente A, Rangarajan S, McQueen MJ, Wang X, Liu L, Yan H, Lee SF, Mony P, Devanath A, Rosengren A, Lopez-Jaramillo P, Diaz R, Avezum A, Lanas F, Yusoff K, Iqbal R, Ilow R, Mohammadifard N, Gulec S, Yusufali AH, Kruger L, Yusuf R, Chifamba J, Kabali C, Dagenais G, Lear SA, Teo K, Yusuf S, PURE Investigators (2014) Urinary sodium and potassium excretion, mortality, and cardiovascular events. N Engl J Med 371:612–623PubMedCrossRef O'Donnell M, Mente A, Rangarajan S, McQueen MJ, Wang X, Liu L, Yan H, Lee SF, Mony P, Devanath A, Rosengren A, Lopez-Jaramillo P, Diaz R, Avezum A, Lanas F, Yusoff K, Iqbal R, Ilow R, Mohammadifard N, Gulec S, Yusufali AH, Kruger L, Yusuf R, Chifamba J, Kabali C, Dagenais G, Lear SA, Teo K, Yusuf S, PURE Investigators (2014) Urinary sodium and potassium excretion, mortality, and cardiovascular events. N Engl J Med 371:612–623PubMedCrossRef
9.
go back to reference Smyth A, Dunkler D, Gao P, Teo KK, Yusuf S, O'Donnell MJ, Mann JF, Clase CM, ONTARGET and TRANSCEND Investigators (2014) The relationship between estimated sodium and potassium excretion and subsequent renal outcomes. Kidney Int 86:1205–1212PubMedCrossRef Smyth A, Dunkler D, Gao P, Teo KK, Yusuf S, O'Donnell MJ, Mann JF, Clase CM, ONTARGET and TRANSCEND Investigators (2014) The relationship between estimated sodium and potassium excretion and subsequent renal outcomes. Kidney Int 86:1205–1212PubMedCrossRef
10.
go back to reference Araki S, Haneda M, Koya D, Kondo K, Tanaka S, Arima H, Kume S, Nakazawa J, Chin-Kanasaki M, Ugi S, Kawai H, Araki H, Uzu T, Maegawa H (2015) Urinary potassium excretion and renal and cardiovascular complications in patients with type 2 diabetes and normal renal function. Clin J Am Soc Nephrol 10:2152–2158PubMedCrossRefPubMedCentral Araki S, Haneda M, Koya D, Kondo K, Tanaka S, Arima H, Kume S, Nakazawa J, Chin-Kanasaki M, Ugi S, Kawai H, Araki H, Uzu T, Maegawa H (2015) Urinary potassium excretion and renal and cardiovascular complications in patients with type 2 diabetes and normal renal function. Clin J Am Soc Nephrol 10:2152–2158PubMedCrossRefPubMedCentral
11.
go back to reference He J, Mills KT, Appel LJ, Yang W, Chen J, Lee BT, Rosas SE, Porter A, Makos G, Weir MR, Hamm LL, Kusek JW, Chronic Renal Insufficiency Cohort Study Investigators (2016) Urinary sodium and potassium excretion and CKD progression. J Am Soc Nephrol 27:1202–1212PubMedCrossRef He J, Mills KT, Appel LJ, Yang W, Chen J, Lee BT, Rosas SE, Porter A, Makos G, Weir MR, Hamm LL, Kusek JW, Chronic Renal Insufficiency Cohort Study Investigators (2016) Urinary sodium and potassium excretion and CKD progression. J Am Soc Nephrol 27:1202–1212PubMedCrossRef
13.
go back to reference Buendia JR, Bradlee ML, Daniels SR, Singer MR, Moore LL (2015) Longitudinal effects of dietary sodium and potassium on blood pressure in adolescent girls. JAMA Pediatr 169:560–568PubMedCrossRef Buendia JR, Bradlee ML, Daniels SR, Singer MR, Moore LL (2015) Longitudinal effects of dietary sodium and potassium on blood pressure in adolescent girls. JAMA Pediatr 169:560–568PubMedCrossRef
15.
go back to reference Krishna GG, Miller E, Kapoor S (1989) Increased blood-pressure during potassium-depletion in normotensive men. N Engl J Med 320:1177–1182PubMedCrossRef Krishna GG, Miller E, Kapoor S (1989) Increased blood-pressure during potassium-depletion in normotensive men. N Engl J Med 320:1177–1182PubMedCrossRef
16.
go back to reference Malnic G, Giebisch G, Muto S, Wang W, Bailey MA, Satlin LM (2013) Chapter 49 - regulation of K+ excretion. In: Caplan RJAWM (ed) Seldin and Giebisch’s the kidney, 5th edn. Academic Press, Cambridge, pp 1659–1715CrossRef Malnic G, Giebisch G, Muto S, Wang W, Bailey MA, Satlin LM (2013) Chapter 49 - regulation of K+ excretion. In: Caplan RJAWM (ed) Seldin and Giebisch’s the kidney, 5th edn. Academic Press, Cambridge, pp 1659–1715CrossRef
17.
go back to reference Welling PA (2013) Regulation of renal potassium secretion: molecular mechanisms. Semin Nephrol 33:215–228PubMedCrossRef Welling PA (2013) Regulation of renal potassium secretion: molecular mechanisms. Semin Nephrol 33:215–228PubMedCrossRef
18.
go back to reference Satlin LM (2004) Developmental regulation of expression of renal potassium secretory channels. Curr Opin Nephrol Hypertens 13:445–450PubMedCrossRef Satlin LM (2004) Developmental regulation of expression of renal potassium secretory channels. Curr Opin Nephrol Hypertens 13:445–450PubMedCrossRef
19.
go back to reference Brandis M, Keyes J, Windhager EE (1972) Potassium-induced inhibition of proximal tubular fluid reabsorption in rats. Am J Physiol 222:421–427PubMed Brandis M, Keyes J, Windhager EE (1972) Potassium-induced inhibition of proximal tubular fluid reabsorption in rats. Am J Physiol 222:421–427PubMed
20.
go back to reference Stokes JB (1982) Consequences of potassium recycling in the renal medulla. Effects of ion transport by the medullary thick ascending limb of Henle’s loop. J Clin Invest 70:219–229PubMedCrossRefPubMedCentral Stokes JB (1982) Consequences of potassium recycling in the renal medulla. Effects of ion transport by the medullary thick ascending limb of Henle’s loop. J Clin Invest 70:219–229PubMedCrossRefPubMedCentral
21.
go back to reference Battilana CA, Dobyan DC, Lacy FB, Bhattacharya J, Johnston PA, Jamison RL (1978) Effect of chronic potassium loading on potassium secretion by the pars recta or descending limb of the juxtamedullary nephron in the rat. J Clin Invest 62:1093–1103PubMedCrossRefPubMedCentral Battilana CA, Dobyan DC, Lacy FB, Bhattacharya J, Johnston PA, Jamison RL (1978) Effect of chronic potassium loading on potassium secretion by the pars recta or descending limb of the juxtamedullary nephron in the rat. J Clin Invest 62:1093–1103PubMedCrossRefPubMedCentral
22.
go back to reference Higashihara E, Kokko JP (1985) Effects of aldosterone on potassium recycling in the kidney of adrenalectomized rats. Am J Physiol 248:F219–F227PubMed Higashihara E, Kokko JP (1985) Effects of aldosterone on potassium recycling in the kidney of adrenalectomized rats. Am J Physiol 248:F219–F227PubMed
23.
go back to reference Cheng CJ, Truong T, Baum M, Huang CL (2012) Kidney-specific WNK1 inhibits sodium reabsorption in the cortical thick ascending limb. Am J Physiol Renal Physiol 303:F667–F673PubMedCrossRefPubMedCentral Cheng CJ, Truong T, Baum M, Huang CL (2012) Kidney-specific WNK1 inhibits sodium reabsorption in the cortical thick ascending limb. Am J Physiol Renal Physiol 303:F667–F673PubMedCrossRefPubMedCentral
24.
go back to reference Pacheco-Alvarez D, Cristobal PS, Meade P, Moreno E, Vazquez N, Munoz E, Diaz A, Juarez ME, Gimenez I, Gamba G (2006) The Na+:Cl- cotransporter is activated and phosphorylated at the amino-terminal domain upon intracellular chloride depletion. J Biol Chem 281:28755–28763 Pacheco-Alvarez D, Cristobal PS, Meade P, Moreno E, Vazquez N, Munoz E, Diaz A, Juarez ME, Gimenez I, Gamba G (2006) The Na+:Cl- cotransporter is activated and phosphorylated at the amino-terminal domain upon intracellular chloride depletion. J Biol Chem 281:28755–28763
25.
go back to reference Richardson C, Rafiqi FH, Karlsson HK, Moleleki N, Vandewalle A, Campbell DG, Morrice NA, Alessi DR (2008) Activation of the thiazide-sensitive Na+−Cl- cotransporter by the WNK-regulated kinases SPAK and OSR1. J Cell Sci 121:675–684 Richardson C, Rafiqi FH, Karlsson HK, Moleleki N, Vandewalle A, Campbell DG, Morrice NA, Alessi DR (2008) Activation of the thiazide-sensitive Na+−Cl- cotransporter by the WNK-regulated kinases SPAK and OSR1. J Cell Sci 121:675–684
26.
go back to reference Wade JB, Fang L, Coleman RA, Liu J, Grimm PR, Wang T, Welling PA (2011) Differential regulation of ROMK (Kir1.1) in distal nephron segments by dietary potassium. Am J Physiol Renal Physiol 300:F1385–F1393PubMedCrossRefPubMedCentral Wade JB, Fang L, Coleman RA, Liu J, Grimm PR, Wang T, Welling PA (2011) Differential regulation of ROMK (Kir1.1) in distal nephron segments by dietary potassium. Am J Physiol Renal Physiol 300:F1385–F1393PubMedCrossRefPubMedCentral
27.
go back to reference van der Lubbe N, Moes AD, Rosenbaek LL, Schoep S, Meima ME, Danser AH, Fenton RA, Zietse R, Hoorn EJ (2013) K + −induced natriuresis is preserved during Na+ depletion and accompanied by inhibition of the Na+−Cl-cotransporter. Am J Physiol Renal Physiol 305:F1177–F1188 van der Lubbe N, Moes AD, Rosenbaek LL, Schoep S, Meima ME, Danser AH, Fenton RA, Zietse R, Hoorn EJ (2013) K + −induced natriuresis is preserved during Na+ depletion and accompanied by inhibition of the Na+−Cl-cotransporter. Am J Physiol Renal Physiol 305:F1177–F1188
28.
go back to reference Rengarajan S, Lee DH, Oh YT, Delpire E, Youn JH, McDonough AA (2014) Increasing plasma [K+] by intravenous potassium infusion reduces NCC phosphorylation and drives kaliuresis and natriuresis. Am J Physiol Renal Physiol 306:F1059–F1068PubMedCrossRefPubMedCentral Rengarajan S, Lee DH, Oh YT, Delpire E, Youn JH, McDonough AA (2014) Increasing plasma [K+] by intravenous potassium infusion reduces NCC phosphorylation and drives kaliuresis and natriuresis. Am J Physiol Renal Physiol 306:F1059–F1068PubMedCrossRefPubMedCentral
29.
go back to reference Castaneda-Bueno M, Cervantes-Perez LG, Rojas-Vega L, Arroyo-Garza I, Vazquez N, Moreno E, Gamba G (2014) Modulation of NCC activity by low and high K(+) intake: insights into the signaling pathways involved. Am J Physiol Renal Physiol 306:F1507–F1519PubMedCrossRefPubMedCentral Castaneda-Bueno M, Cervantes-Perez LG, Rojas-Vega L, Arroyo-Garza I, Vazquez N, Moreno E, Gamba G (2014) Modulation of NCC activity by low and high K(+) intake: insights into the signaling pathways involved. Am J Physiol Renal Physiol 306:F1507–F1519PubMedCrossRefPubMedCentral
30.
go back to reference Vallon V, Schroth J, Lang F, Kuhl D, Uchida S (2009) Expression and phosphorylation of the Na+−Cl-cotransporter NCC in vivo is regulated by dietary salt, potassium, and SGK1. Am J Physiol Renal Physiol 297:F704–F712 Vallon V, Schroth J, Lang F, Kuhl D, Uchida S (2009) Expression and phosphorylation of the Na+−Cl-cotransporter NCC in vivo is regulated by dietary salt, potassium, and SGK1. Am J Physiol Renal Physiol 297:F704–F712
31.
go back to reference Frindt G, Houde V, Palmer LG (2011) Conservation of Na+ vs. K+ by the rat cortical collecting duct. Am J Physiol Renal Physiol 301:F14–F20 Frindt G, Houde V, Palmer LG (2011) Conservation of Na+ vs. K+ by the rat cortical collecting duct. Am J Physiol Renal Physiol 301:F14–F20
32.
go back to reference Wade JB, Liu J, Coleman R, Grimm PR, Delpire E, Welling PA (2015) SPAK-mediated NCC regulation in response to low-K+ diet. Am J Physiol Renal Physiol 308:F923–F931 Wade JB, Liu J, Coleman R, Grimm PR, Delpire E, Welling PA (2015) SPAK-mediated NCC regulation in response to low-K+ diet. Am J Physiol Renal Physiol 308:F923–F931
33.
go back to reference Sorensen MV, Grossmann S, Roesinger M, Gresko N, Todkar AP, Barmettler G, Ziegler U, Odermatt A, Loffing-Cueni D, Loffing J (2013) Rapid dephosphorylation of the renal sodium chloride cotransporter in response to oral potassium intake in mice. Kidney Int 83:811–824PubMedCrossRef Sorensen MV, Grossmann S, Roesinger M, Gresko N, Todkar AP, Barmettler G, Ziegler U, Odermatt A, Loffing-Cueni D, Loffing J (2013) Rapid dephosphorylation of the renal sodium chloride cotransporter in response to oral potassium intake in mice. Kidney Int 83:811–824PubMedCrossRef
34.
go back to reference Chiga M, Rai T, Yang SS, Ohta A, Takizawa T, Sasaki S, Uchida S (2008) Dietary salt regulates the phosphorylation of OSR1/SPAK kinases and the sodium chloride cotransporter through aldosterone. Kidney Int 74:1403–1409PubMedCrossRef Chiga M, Rai T, Yang SS, Ohta A, Takizawa T, Sasaki S, Uchida S (2008) Dietary salt regulates the phosphorylation of OSR1/SPAK kinases and the sodium chloride cotransporter through aldosterone. Kidney Int 74:1403–1409PubMedCrossRef
35.
go back to reference Turban S, Thompson CB, Parekh RS, Appel LJ (2013) Effects of sodium intake and diet on racial differences in urinary potassium excretion: results from the dietary approaches to stop hypertension (DASH)-sodium trial. Am J Kidney Dis 61:88–95PubMedCrossRef Turban S, Thompson CB, Parekh RS, Appel LJ (2013) Effects of sodium intake and diet on racial differences in urinary potassium excretion: results from the dietary approaches to stop hypertension (DASH)-sodium trial. Am J Kidney Dis 61:88–95PubMedCrossRef
36.
go back to reference Vitzthum H, Seniuk A, Schulte LH, Muller ML, Hetz H, Ehmke H (2014) Functional coupling of renal K+ and Na+ handling causes high blood pressure in Na + replete mice. J Physiol 592:1139–1157PubMedCrossRefPubMedCentral Vitzthum H, Seniuk A, Schulte LH, Muller ML, Hetz H, Ehmke H (2014) Functional coupling of renal K+ and Na+ handling causes high blood pressure in Na + replete mice. J Physiol 592:1139–1157PubMedCrossRefPubMedCentral
37.
go back to reference Terker AS, Zhang C, McCormick JA, Lazelle RA, Zhang C, Meermeier NP, Siler DA, Park HJ, Fu Y, Cohen DM, Weinstein AM, Wang WH, Yang CL, Ellison DH (2015) Potassium modulates electrolyte balance and blood pressure through effects on distal cell voltage and chloride. Cell Metab 21:39–50PubMedCrossRefPubMedCentral Terker AS, Zhang C, McCormick JA, Lazelle RA, Zhang C, Meermeier NP, Siler DA, Park HJ, Fu Y, Cohen DM, Weinstein AM, Wang WH, Yang CL, Ellison DH (2015) Potassium modulates electrolyte balance and blood pressure through effects on distal cell voltage and chloride. Cell Metab 21:39–50PubMedCrossRefPubMedCentral
38.
go back to reference Hou J, Renigunta A, Yang J, Waldegger S (2010) Claudin-4 forms paracellular chloride channel in the kidney and requires claudin-8 for tight junction localization. Proc Natl Acad Sci USA 107:18010–18015PubMedCrossRefPubMedCentral Hou J, Renigunta A, Yang J, Waldegger S (2010) Claudin-4 forms paracellular chloride channel in the kidney and requires claudin-8 for tight junction localization. Proc Natl Acad Sci USA 107:18010–18015PubMedCrossRefPubMedCentral
39.
go back to reference Gong Y, Wang J, Yang J, Gonzales E, Perez R, Hou J (2015) KLHL3 regulates paracellular chloride transport in the kidney by ubiquitination of claudin-8. Proc Natl Acad Sci USA 112:4340–4345PubMedCrossRefPubMedCentral Gong Y, Wang J, Yang J, Gonzales E, Perez R, Hou J (2015) KLHL3 regulates paracellular chloride transport in the kidney by ubiquitination of claudin-8. Proc Natl Acad Sci USA 112:4340–4345PubMedCrossRefPubMedCentral
40.
go back to reference Gong Y, Yu M, Yang J, Gonzales E, Perez R, Hou M, Tripathi P, Hering-Smith KS, Hamm LL, Hou J (2014) The Cap1-claudin-4 regulatory pathway is important for renal chloride reabsorption and blood pressure regulation. Proc Natl Acad Sci USA 111:E3766–E3774PubMedCrossRefPubMedCentral Gong Y, Yu M, Yang J, Gonzales E, Perez R, Hou M, Tripathi P, Hering-Smith KS, Hamm LL, Hou J (2014) The Cap1-claudin-4 regulatory pathway is important for renal chloride reabsorption and blood pressure regulation. Proc Natl Acad Sci USA 111:E3766–E3774PubMedCrossRefPubMedCentral
41.
go back to reference Terada Y, Knepper MA (1990) Thiazide-sensitive NaCl absorption in rat cortical collecting duct. Am J Physiol 259:F519–F528PubMed Terada Y, Knepper MA (1990) Thiazide-sensitive NaCl absorption in rat cortical collecting duct. Am J Physiol 259:F519–F528PubMed
42.
go back to reference Gueutin V, Vallet M, Jayat M, Peti-Peterdi J, Corniere N, Leviel F, Sohet F, Wagner CA, Eladari D, Chambrey R (2013) Renal beta-intercalated cells maintain body fluid and electrolyte balance. J Clin Invest 123:4219–4231PubMedCrossRefPubMedCentral Gueutin V, Vallet M, Jayat M, Peti-Peterdi J, Corniere N, Leviel F, Sohet F, Wagner CA, Eladari D, Chambrey R (2013) Renal beta-intercalated cells maintain body fluid and electrolyte balance. J Clin Invest 123:4219–4231PubMedCrossRefPubMedCentral
43.
go back to reference Chambrey R, Kurth I, Peti-Peterdi J, Houillier P, Purkerson JM, Leviel F, Hentschke M, Zdebik AA, Schwartz GJ, Hubner CA, Eladari D (2013) Renal intercalated cells are rather energized by a proton than a sodium pump. Proc Natl Acad Sci USA 110:7928–7933PubMedCrossRefPubMedCentral Chambrey R, Kurth I, Peti-Peterdi J, Houillier P, Purkerson JM, Leviel F, Hentschke M, Zdebik AA, Schwartz GJ, Hubner CA, Eladari D (2013) Renal intercalated cells are rather energized by a proton than a sodium pump. Proc Natl Acad Sci USA 110:7928–7933PubMedCrossRefPubMedCentral
44.
go back to reference Leviel F, Hubner CA, Houillier P, Morla L, El Moghrabi S, Brideau G, Hassan H, Parker MD, Kurth I, Kougioumtzes A, Sinning A, Pech V, Riemondy KA, Miller RL, Hummler E, Shull GE, Aronson PS, Doucet A, Wall SM, Chambrey R, Eladari D (2010) The Na+-dependent chloride-bicarbonate exchanger SLC4A8 mediates an electroneutral Na + reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 120:1627–1635PubMedCrossRefPubMedCentral Leviel F, Hubner CA, Houillier P, Morla L, El Moghrabi S, Brideau G, Hassan H, Parker MD, Kurth I, Kougioumtzes A, Sinning A, Pech V, Riemondy KA, Miller RL, Hummler E, Shull GE, Aronson PS, Doucet A, Wall SM, Chambrey R, Eladari D (2010) The Na+-dependent chloride-bicarbonate exchanger SLC4A8 mediates an electroneutral Na + reabsorption process in the renal cortical collecting ducts of mice. J Clin Invest 120:1627–1635PubMedCrossRefPubMedCentral
45.
go back to reference Ellison DH, Velazquez H, Wright FS (1989) Adaptation of the distal convoluted tubule of the rat. Structural and functional effects of dietary salt intake and chronic diuretic infusion. J Clin Invest 83:113–126PubMedCrossRefPubMedCentral Ellison DH, Velazquez H, Wright FS (1989) Adaptation of the distal convoluted tubule of the rat. Structural and functional effects of dietary salt intake and chronic diuretic infusion. J Clin Invest 83:113–126PubMedCrossRefPubMedCentral
46.
go back to reference Nesterov V, Dahlmann A, Krueger B, Bertog M, Loffing J, Korbmacher C (2012) Aldosterone-dependent and -independent regulation of the epithelial sodium channel (ENaC) in mouse distal nephron. Am J Physiol Renal Physiol 303:F1289–F1299PubMedCrossRef Nesterov V, Dahlmann A, Krueger B, Bertog M, Loffing J, Korbmacher C (2012) Aldosterone-dependent and -independent regulation of the epithelial sodium channel (ENaC) in mouse distal nephron. Am J Physiol Renal Physiol 303:F1289–F1299PubMedCrossRef
47.
go back to reference Frindt G, Sackin H, Palmer LG (1990) Whole-cell currents in rat cortical collecting tubule: low-Na diet increases amiloride-sensitive conductance. Am J Physiol 258:F562–F567PubMed Frindt G, Sackin H, Palmer LG (1990) Whole-cell currents in rat cortical collecting tubule: low-Na diet increases amiloride-sensitive conductance. Am J Physiol 258:F562–F567PubMed
48.
go back to reference Shibata S, Rinehart J, Zhang J, Moeckel G, Castaneda-Bueno M, Stiegler AL, Boggon TJ, Gamba G, Lifton RP (2013) Mineralocorticoid receptor phosphorylation regulates ligand binding and renal response to volume depletion and hyperkalemia. Cell Metab 18:660–671PubMedCrossRefPubMedCentral Shibata S, Rinehart J, Zhang J, Moeckel G, Castaneda-Bueno M, Stiegler AL, Boggon TJ, Gamba G, Lifton RP (2013) Mineralocorticoid receptor phosphorylation regulates ligand binding and renal response to volume depletion and hyperkalemia. Cell Metab 18:660–671PubMedCrossRefPubMedCentral
49.
go back to reference Grimm PR, Lazo-Fernandez Y, Delpire E, Wall SM, Dorsey SG, Weinman EJ, Coleman R, Wade JB, Welling PA (2015) Integrated compensatory network is activated in the absence of NCC phosphorylation. J Clin Invest 125:2136–2150PubMedCrossRefPubMedCentral Grimm PR, Lazo-Fernandez Y, Delpire E, Wall SM, Dorsey SG, Weinman EJ, Coleman R, Wade JB, Welling PA (2015) Integrated compensatory network is activated in the absence of NCC phosphorylation. J Clin Invest 125:2136–2150PubMedCrossRefPubMedCentral
50.
go back to reference Yang SS, Lo YF, Wu CC, Lin SW, Yeh CJ, Chu P, Sytwu HK, Uchida S, Sasaki S, Lin SH (2010) SPAK-knockout mice manifest Gitelman syndrome and impaired vasoconstriction. J Am Soc Nephrol 21:1868–1877PubMedCrossRefPubMedCentral Yang SS, Lo YF, Wu CC, Lin SW, Yeh CJ, Chu P, Sytwu HK, Uchida S, Sasaki S, Lin SH (2010) SPAK-knockout mice manifest Gitelman syndrome and impaired vasoconstriction. J Am Soc Nephrol 21:1868–1877PubMedCrossRefPubMedCentral
51.
go back to reference Grimm PR, Taneja TK, Liu J, Coleman R, Chen YY, Delpire E, Wade JB, Welling PA (2012) SPAK isoforms and OSR1 regulate sodium-chloride co-transporters in a nephron-specific manner. J Biol Chem 287:37673–37690PubMedCrossRefPubMedCentral Grimm PR, Taneja TK, Liu J, Coleman R, Chen YY, Delpire E, Wade JB, Welling PA (2012) SPAK isoforms and OSR1 regulate sodium-chloride co-transporters in a nephron-specific manner. J Biol Chem 287:37673–37690PubMedCrossRefPubMedCentral
52.
go back to reference Tokonami N, Morla L, Centeno G, Mordasini D, Ramakrishnan SK, Nikolaeva S, Wagner CA, Bonny O, Houillier P, Doucet A, Firsov D (2013) alpha-Ketoglutarate regulates acid–base balance through an intrarenal paracrine mechanism. J Clin Invest 123:3166–3171PubMedCrossRefPubMedCentral Tokonami N, Morla L, Centeno G, Mordasini D, Ramakrishnan SK, Nikolaeva S, Wagner CA, Bonny O, Houillier P, Doucet A, Firsov D (2013) alpha-Ketoglutarate regulates acid–base balance through an intrarenal paracrine mechanism. J Clin Invest 123:3166–3171PubMedCrossRefPubMedCentral
53.
go back to reference Soleimani M, Barone S, Xu J, Shull GE, Siddiqui F, Zahedi K, Amlal H (2012) Double knockout of pendrin and Na-Cl cotransporter (NCC) causes severe salt wasting, volume depletion, and renal failure. Proc Natl Acad Sci USA 109:13368–13373PubMedCrossRefPubMedCentral Soleimani M, Barone S, Xu J, Shull GE, Siddiqui F, Zahedi K, Amlal H (2012) Double knockout of pendrin and Na-Cl cotransporter (NCC) causes severe salt wasting, volume depletion, and renal failure. Proc Natl Acad Sci USA 109:13368–13373PubMedCrossRefPubMedCentral
54.
go back to reference Pela I, Bigozzi M, Bianchi B (2008) Profound hypokalemia and hypochloremic metabolic alkalosis during thiazide therapy in a child with Pendred syndrome. Clin Nephrol 69:450–453PubMedCrossRef Pela I, Bigozzi M, Bianchi B (2008) Profound hypokalemia and hypochloremic metabolic alkalosis during thiazide therapy in a child with Pendred syndrome. Clin Nephrol 69:450–453PubMedCrossRef
55.
go back to reference Xu B, English JM, Wilsbacher JL, Stippec S, Goldsmith EJ, Cobb MH (2000) WNK1, a novel mammalian serine/threonine protein kinase lacking the catalytic lysine in subdomain II. J Biol Chem 275:16795–16801PubMedCrossRef Xu B, English JM, Wilsbacher JL, Stippec S, Goldsmith EJ, Cobb MH (2000) WNK1, a novel mammalian serine/threonine protein kinase lacking the catalytic lysine in subdomain II. J Biol Chem 275:16795–16801PubMedCrossRef
56.
go back to reference Wilson FH, Disse-Nicodeme S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, Lifton RP (2001) Human hypertension caused by mutations in WNK kinases. Science 293:1107–1112PubMedCrossRef Wilson FH, Disse-Nicodeme S, Choate KA, Ishikawa K, Nelson-Williams C, Desitter I, Gunel M, Milford DV, Lipkin GW, Achard JM, Feely MP, Dussol B, Berland Y, Unwin RJ, Mayan H, Simon DB, Farfel Z, Jeunemaitre X, Lifton RP (2001) Human hypertension caused by mutations in WNK kinases. Science 293:1107–1112PubMedCrossRef
57.
go back to reference Hadchouel J, Ellison DH, Gamba G (2016) Regulation of renal electrolyte transport by WNK and SPAK-OSR1 kinases. Annu Rev Physiol 78:367–389PubMedCrossRef Hadchouel J, Ellison DH, Gamba G (2016) Regulation of renal electrolyte transport by WNK and SPAK-OSR1 kinases. Annu Rev Physiol 78:367–389PubMedCrossRef
58.
59.
go back to reference Wu Y, Schellinger JN, Huang CL, Rodan AR (2014) Hypotonicity stimulates potassium flux through the WNK-SPAK/OSR1 kinase cascade and the Ncc69 sodium-potassium-2-chloride cotransporter in the drosophila renal tubule. J Biol Chem 289:26131–26142PubMedCrossRefPubMedCentral Wu Y, Schellinger JN, Huang CL, Rodan AR (2014) Hypotonicity stimulates potassium flux through the WNK-SPAK/OSR1 kinase cascade and the Ncc69 sodium-potassium-2-chloride cotransporter in the drosophila renal tubule. J Biol Chem 289:26131–26142PubMedCrossRefPubMedCentral
61.
62.
go back to reference Terker AS, Zhang C, Erspamer KJ, Gamba G, Yang CL, Ellison DH (2015) Unique chloride-sensing properties of WNK4 permit the distal nephron to modulate potassium homeostasis. Kidney Int. doi:10.1038/ki.2015.289 Terker AS, Zhang C, Erspamer KJ, Gamba G, Yang CL, Ellison DH (2015) Unique chloride-sensing properties of WNK4 permit the distal nephron to modulate potassium homeostasis. Kidney Int. doi:10.​1038/​ki.​2015.​289
63.
go back to reference Petrov DB (2012) Images in clinical medicine. An electrocardiographic sine wave in hyperkalemia. N Engl J Med 366:1824PubMedCrossRef Petrov DB (2012) Images in clinical medicine. An electrocardiographic sine wave in hyperkalemia. N Engl J Med 366:1824PubMedCrossRef
64.
65.
go back to reference Palmer BF (2010) A physiologic-based approach to the evaluation of a patient with hyperkalemia. Am J Kidney Dis 56:387–393PubMedCrossRef Palmer BF (2010) A physiologic-based approach to the evaluation of a patient with hyperkalemia. Am J Kidney Dis 56:387–393PubMedCrossRef
66.
go back to reference Palmer BF (2004) Managing hyperkalemia caused by inhibitors of the renin-angiotensin-aldosterone system. N Engl J Med 351:585–592PubMedCrossRef Palmer BF (2004) Managing hyperkalemia caused by inhibitors of the renin-angiotensin-aldosterone system. N Engl J Med 351:585–592PubMedCrossRef
67.
go back to reference Einhorn LM, Zhan M, Hsu VD, Walker LD, Moen MF, Seliger SL, Weir MR, Fink JC (2009) The frequency of hyperkalemia and its significance in chronic kidney disease. Arch Intern Med 169:1156–1162PubMedCrossRefPubMedCentral Einhorn LM, Zhan M, Hsu VD, Walker LD, Moen MF, Seliger SL, Weir MR, Fink JC (2009) The frequency of hyperkalemia and its significance in chronic kidney disease. Arch Intern Med 169:1156–1162PubMedCrossRefPubMedCentral
68.
go back to reference Jain N, Kotla S, Little BB, Weideman RA, Brilakis ES, Reilly RF, Banerjee S (2012) Predictors of hyperkalemia and death in patients with cardiac and renal disease. Am J Cardiol 109:1510–1513PubMedCrossRef Jain N, Kotla S, Little BB, Weideman RA, Brilakis ES, Reilly RF, Banerjee S (2012) Predictors of hyperkalemia and death in patients with cardiac and renal disease. Am J Cardiol 109:1510–1513PubMedCrossRef
69.
go back to reference Michel A, Martin-Perez M, Ruigomez A, Garcia Rodriguez LA (2015) Risk factors for hyperkalaemia in a cohort of patients with newly diagnosed heart failure: a nested case–control study in UK general practice. Eur J Heart Fail 17:205–213PubMedCrossRef Michel A, Martin-Perez M, Ruigomez A, Garcia Rodriguez LA (2015) Risk factors for hyperkalaemia in a cohort of patients with newly diagnosed heart failure: a nested case–control study in UK general practice. Eur J Heart Fail 17:205–213PubMedCrossRef
70.
go back to reference Korgaonkar S, Tilea A, Gillespie BW, Kiser M, Eisele G, Finkelstein F, Kotanko P, Pitt B, Saran R (2010) Serum potassium and outcomes in CKD: insights from the RRI-CKD cohort study. Clin J Am Soc Nephrol 5:762–769PubMedCrossRefPubMedCentral Korgaonkar S, Tilea A, Gillespie BW, Kiser M, Eisele G, Finkelstein F, Kotanko P, Pitt B, Saran R (2010) Serum potassium and outcomes in CKD: insights from the RRI-CKD cohort study. Clin J Am Soc Nephrol 5:762–769PubMedCrossRefPubMedCentral
71.
go back to reference Gwoo S, Kim YN, Shin HS, Jung YS, Rim H (2014) Predictors of hyperkalemia risk after hypertension control with aldosterone blockade according to the presence or absence of chronic kidney disease. Nephron Clin Pract 128:381–386PubMedCrossRef Gwoo S, Kim YN, Shin HS, Jung YS, Rim H (2014) Predictors of hyperkalemia risk after hypertension control with aldosterone blockade according to the presence or absence of chronic kidney disease. Nephron Clin Pract 128:381–386PubMedCrossRef
72.
go back to reference Weinberg JM, Appel LJ, Bakris G, Gassman JJ, Greene T, Kendrick CA, Wang X, Lash J, Lewis JA, Pogue V, Thornley-Brown D, Phillips RA, African American Study of Hypertension and Kidney Disease Collaborative Research Group (2009) Risk of hyperkalemia in nondiabetic patients with chronic kidney disease receiving antihypertensive therapy. Arch Intern Med 169:1587–1594PubMedCrossRef Weinberg JM, Appel LJ, Bakris G, Gassman JJ, Greene T, Kendrick CA, Wang X, Lash J, Lewis JA, Pogue V, Thornley-Brown D, Phillips RA, African American Study of Hypertension and Kidney Disease Collaborative Research Group (2009) Risk of hyperkalemia in nondiabetic patients with chronic kidney disease receiving antihypertensive therapy. Arch Intern Med 169:1587–1594PubMedCrossRef
73.
go back to reference Alappan R, Buller GK, Perazella MA (1999) Trimethoprim-sulfamethoxazole therapy in outpatients: is hyperkalemia a significant problem? Am J Nephrol 19:389–394PubMedCrossRef Alappan R, Buller GK, Perazella MA (1999) Trimethoprim-sulfamethoxazole therapy in outpatients: is hyperkalemia a significant problem? Am J Nephrol 19:389–394PubMedCrossRef
74.
go back to reference Antoniou T, Gomes T, Juurlink DN, Loutfy MR, Glazier RH, Mamdani MM (2010) Trimethoprim-sulfamethoxazole-induced hyperkalemia in patients receiving inhibitors of the renin-angiotensin system: a population-based study. Arch Intern Med 170:1045–1049PubMedCrossRef Antoniou T, Gomes T, Juurlink DN, Loutfy MR, Glazier RH, Mamdani MM (2010) Trimethoprim-sulfamethoxazole-induced hyperkalemia in patients receiving inhibitors of the renin-angiotensin system: a population-based study. Arch Intern Med 170:1045–1049PubMedCrossRef
75.
go back to reference Antoniou T, Gomes T, Mamdani MM, Yao Z, Hellings C, Garg AX, Weir MA, Juurlink DN (2011) Trimethoprim-sulfamethoxazole induced hyperkalaemia in elderly patients receiving spironolactone: nested case–control study. BMJ 343:d5228PubMedCrossRefPubMedCentral Antoniou T, Gomes T, Mamdani MM, Yao Z, Hellings C, Garg AX, Weir MA, Juurlink DN (2011) Trimethoprim-sulfamethoxazole induced hyperkalaemia in elderly patients receiving spironolactone: nested case–control study. BMJ 343:d5228PubMedCrossRefPubMedCentral
76.
go back to reference Antoniou T, Hollands S, MacDonald EM, Gomes T, Mamdani MM, Juurlink DN, Canadian Drug Safety and Effectiveness Research Network (2015) Trimethoprim-sulfamethoxazole and risk of sudden death among patients taking spironolactone. CMAJ 187:E138–E143PubMedCrossRefPubMedCentral Antoniou T, Hollands S, MacDonald EM, Gomes T, Mamdani MM, Juurlink DN, Canadian Drug Safety and Effectiveness Research Network (2015) Trimethoprim-sulfamethoxazole and risk of sudden death among patients taking spironolactone. CMAJ 187:E138–E143PubMedCrossRefPubMedCentral
77.
go back to reference Fralick M, MacDonald EM, Gomes T, Antoniou T, Hollands S, Mamdani MM, Juurlink DN, Canadian Drug Safety and Effectiveness ResearchNetwork (2014) Co-trimoxazole and sudden death in patients receiving inhibitors of renin-angiotensin system: population based study. BMJ 349:g6196PubMedCrossRefPubMedCentral Fralick M, MacDonald EM, Gomes T, Antoniou T, Hollands S, Mamdani MM, Juurlink DN, Canadian Drug Safety and Effectiveness ResearchNetwork (2014) Co-trimoxazole and sudden death in patients receiving inhibitors of renin-angiotensin system: population based study. BMJ 349:g6196PubMedCrossRefPubMedCentral
78.
go back to reference Muschart X, Boulouffe C, Jamart J, Nougon G, Gerard V, de Canniere L, Vanpee D (2014) A determination of the current causes of hyperkalaemia and whether they have changed over the past 25 years. Acta Clin Belg 69:280–284PubMedCrossRef Muschart X, Boulouffe C, Jamart J, Nougon G, Gerard V, de Canniere L, Vanpee D (2014) A determination of the current causes of hyperkalaemia and whether they have changed over the past 25 years. Acta Clin Belg 69:280–284PubMedCrossRef
79.
go back to reference Weir MR, Rolfe M (2010) Potassium homeostasis and renin-angiotensin-aldosterone system inhibitors. Clin J Am Soc Nephrol 5:531–548PubMedCrossRef Weir MR, Rolfe M (2010) Potassium homeostasis and renin-angiotensin-aldosterone system inhibitors. Clin J Am Soc Nephrol 5:531–548PubMedCrossRef
80.
go back to reference Juurlink DN, Mamdani MM, Lee DS, Kopp A, Austin PC, Laupacis A, Redelmeier DA (2004) Rates of hyperkalemia after publication of the randomized aldactone evaluation study. N Engl J Med 351:543–551PubMedCrossRef Juurlink DN, Mamdani MM, Lee DS, Kopp A, Austin PC, Laupacis A, Redelmeier DA (2004) Rates of hyperkalemia after publication of the randomized aldactone evaluation study. N Engl J Med 351:543–551PubMedCrossRef
81.
go back to reference Moore C, Lin J, McGinn T, Halm E (2007) Factors associated with time to follow-up of severe hyperkalemia in the ambulatory setting. Am J Med Qual 22:428–437PubMedCrossRef Moore C, Lin J, McGinn T, Halm E (2007) Factors associated with time to follow-up of severe hyperkalemia in the ambulatory setting. Am J Med Qual 22:428–437PubMedCrossRef
82.
go back to reference Moore CR, Lin JJ, O'Connor N, Halm EA (2006) Follow-up of markedly elevated serum potassium results in the ambulatory setting: implications for patient safety. Am J Med Qual 21:115–124PubMedCrossRef Moore CR, Lin JJ, O'Connor N, Halm EA (2006) Follow-up of markedly elevated serum potassium results in the ambulatory setting: implications for patient safety. Am J Med Qual 21:115–124PubMedCrossRef
83.
go back to reference Field MJ, Stanton BA, Giebisch GH (1984) Differential acute effects of aldosterone, dexamethasone, and hyperkalemia on distal tubular potassium secretion in the rat kidney. J Clin Invest 74:1792–1802PubMedCrossRefPubMedCentral Field MJ, Stanton BA, Giebisch GH (1984) Differential acute effects of aldosterone, dexamethasone, and hyperkalemia on distal tubular potassium secretion in the rat kidney. J Clin Invest 74:1792–1802PubMedCrossRefPubMedCentral
84.
go back to reference Hirsch D, Kashgarian M, Boulpaep EL, Hayslett JP (1984) Role of aldosterone in the mechanism of potassium adaptation in the initial collecting tubule. Kidney Int 26:798–807PubMedCrossRef Hirsch D, Kashgarian M, Boulpaep EL, Hayslett JP (1984) Role of aldosterone in the mechanism of potassium adaptation in the initial collecting tubule. Kidney Int 26:798–807PubMedCrossRef
85.
go back to reference Stanton B, Pan L, Deetjen H, Guckian V, Giebisch G (1987) Independent effects of aldosterone and potassium on induction of potassium adaptation in rat kidney. J Clin Invest 79:198–206PubMedCrossRefPubMedCentral Stanton B, Pan L, Deetjen H, Guckian V, Giebisch G (1987) Independent effects of aldosterone and potassium on induction of potassium adaptation in rat kidney. J Clin Invest 79:198–206PubMedCrossRefPubMedCentral
86.
go back to reference Wingo CS, Seldin DW, Kokko JP, Jacobson HR (1982) Dietary modulation of active potassium secretion in the cortical collecting tubule of adrenalectomized rabbits. J Clin Invest 70:579–586PubMedCrossRefPubMedCentral Wingo CS, Seldin DW, Kokko JP, Jacobson HR (1982) Dietary modulation of active potassium secretion in the cortical collecting tubule of adrenalectomized rabbits. J Clin Invest 70:579–586PubMedCrossRefPubMedCentral
87.
go back to reference Young DB (1988) Quantitative analysis of aldosterone’s role in potassium regulation. Am J Physiol 255:F811–F822PubMed Young DB (1988) Quantitative analysis of aldosterone’s role in potassium regulation. Am J Physiol 255:F811–F822PubMed
88.
go back to reference Frindt G, Palmer LG (2009) K+ secretion in the rat kidney: Na+ channel-dependent and -independent mechanisms. Am J Physiol Renal Physiol 297:F389–F396 Frindt G, Palmer LG (2009) K+ secretion in the rat kidney: Na+ channel-dependent and -independent mechanisms. Am J Physiol Renal Physiol 297:F389–F396
89.
go back to reference Todkar A, Picard N, Loffing-Cueni D, Sorensen MV, Mihailova M, Nesterov V, Makhanova N, Korbmacher C, Wagner CA, Loffing J (2015) Mechanisms of renal control of potassium homeostasis in complete aldosterone deficiency. J Am Soc Nephrol 26:425–438PubMedCrossRef Todkar A, Picard N, Loffing-Cueni D, Sorensen MV, Mihailova M, Nesterov V, Makhanova N, Korbmacher C, Wagner CA, Loffing J (2015) Mechanisms of renal control of potassium homeostasis in complete aldosterone deficiency. J Am Soc Nephrol 26:425–438PubMedCrossRef
90.
go back to reference Van Buren PN, Adams-Huet B, Nguyen M, Molina C, Toto RD (2014) Potassium handling with dual renin-angiotensin system inhibition in diabetic nephropathy. Clin J Am Soc Nephrol 9:295–301PubMedCrossRefPubMedCentral Van Buren PN, Adams-Huet B, Nguyen M, Molina C, Toto RD (2014) Potassium handling with dual renin-angiotensin system inhibition in diabetic nephropathy. Clin J Am Soc Nephrol 9:295–301PubMedCrossRefPubMedCentral
91.
go back to reference Walsh M, Manns B, Garg AX, Bueti J, Rabbat C, Smyth A, Tyrwhitt J, Bosch J, Gao P, Devereaux PJ, Wald R (2015) The safety of eplerenone in hemodialysis patients: a noninferiority randomized controlled trial. Clin J Am Soc Nephrol 10:1602–1608PubMedCrossRefPubMedCentral Walsh M, Manns B, Garg AX, Bueti J, Rabbat C, Smyth A, Tyrwhitt J, Bosch J, Gao P, Devereaux PJ, Wald R (2015) The safety of eplerenone in hemodialysis patients: a noninferiority randomized controlled trial. Clin J Am Soc Nephrol 10:1602–1608PubMedCrossRefPubMedCentral
92.
go back to reference Bowling CB, Pitt B, Ahmed MI, Aban IB, Sanders PW, Mujib M, Campbell RC, Love TE, Aronow WS, Allman RM, Bakris GL, Ahmed A (2010) Hypokalemia and outcomes in patients with chronic heart failure and chronic kidney disease: findings from propensity-matched studies. Circ Heart Fail 3:253–260PubMedCrossRefPubMedCentral Bowling CB, Pitt B, Ahmed MI, Aban IB, Sanders PW, Mujib M, Campbell RC, Love TE, Aronow WS, Allman RM, Bakris GL, Ahmed A (2010) Hypokalemia and outcomes in patients with chronic heart failure and chronic kidney disease: findings from propensity-matched studies. Circ Heart Fail 3:253–260PubMedCrossRefPubMedCentral
93.
go back to reference Luo J, Brunelli SM, Jensen DE, Yang A (2016) Association between serum potassium and outcomes in patients with reduced kidney function. Clin J Am Soc Nephrol 11:90–100PubMedCrossRef Luo J, Brunelli SM, Jensen DE, Yang A (2016) Association between serum potassium and outcomes in patients with reduced kidney function. Clin J Am Soc Nephrol 11:90–100PubMedCrossRef
94.
go back to reference Kovesdy CP (2014) Management of hyperkalaemia in chronic kidney disease. Nat Rev Nephrol 10:653–662PubMedCrossRef Kovesdy CP (2014) Management of hyperkalaemia in chronic kidney disease. Nat Rev Nephrol 10:653–662PubMedCrossRef
95.
go back to reference Nguyen TQ, Maalouf NM, Sakhaee K, Moe OW (2011) Comparison of insulin action on glucose versus potassium uptake in humans. Clin J Am Soc Nephrol 6:1533–1539PubMedCrossRefPubMedCentral Nguyen TQ, Maalouf NM, Sakhaee K, Moe OW (2011) Comparison of insulin action on glucose versus potassium uptake in humans. Clin J Am Soc Nephrol 6:1533–1539PubMedCrossRefPubMedCentral
96.
go back to reference Sterns RH, Rojas M, Bernstein P, Chennupati S (2010) Ion-exchange resins for the treatment of hyperkalemia: are they safe and effective? J Am Soc Nephrol 21:733–735PubMedCrossRef Sterns RH, Rojas M, Bernstein P, Chennupati S (2010) Ion-exchange resins for the treatment of hyperkalemia: are they safe and effective? J Am Soc Nephrol 21:733–735PubMedCrossRef
98.
go back to reference Watson MA, Baker TP, Nguyen A, Sebastianelli ME, Stewart HL, Oliver DK, Abbott KC, Yuan CM (2012) Association of prescription of oral sodium polystyrene sulfonate with sorbitol in an inpatient setting with colonic necrosis: a retrospective cohort study. Am J Kidney Dis 60:409–416PubMedCrossRef Watson MA, Baker TP, Nguyen A, Sebastianelli ME, Stewart HL, Oliver DK, Abbott KC, Yuan CM (2012) Association of prescription of oral sodium polystyrene sulfonate with sorbitol in an inpatient setting with colonic necrosis: a retrospective cohort study. Am J Kidney Dis 60:409–416PubMedCrossRef
99.
go back to reference Lepage L, Dufour AC, Doiron J, Handfield K, Desforges K, Bell R, Vallee M, Savoie M, Perreault S, Laurin LP, Pichette V, Lafrance JP (2015) Randomized clinical trial of sodium polystyrene sulfonate for the treatment of mild hyperkalemia in CKD. Clin J Am Soc Nephrol 10:2136–2142PubMedCrossRefPubMedCentral Lepage L, Dufour AC, Doiron J, Handfield K, Desforges K, Bell R, Vallee M, Savoie M, Perreault S, Laurin LP, Pichette V, Lafrance JP (2015) Randomized clinical trial of sodium polystyrene sulfonate for the treatment of mild hyperkalemia in CKD. Clin J Am Soc Nephrol 10:2136–2142PubMedCrossRefPubMedCentral
100.
go back to reference Kessler C, Ng J, Valdez K, Xie H, Geiger B (2011) The use of sodium polystyrene sulfonate in the inpatient management of hyperkalemia. J Hosp Med 6:136–140PubMedCrossRef Kessler C, Ng J, Valdez K, Xie H, Geiger B (2011) The use of sodium polystyrene sulfonate in the inpatient management of hyperkalemia. J Hosp Med 6:136–140PubMedCrossRef
101.
go back to reference Fordjour KN, Walton T, Doran JJ (2014) Management of hyperkalemia in hospitalized patients. Am J Med Sci 347:93–100PubMedCrossRef Fordjour KN, Walton T, Doran JJ (2014) Management of hyperkalemia in hospitalized patients. Am J Med Sci 347:93–100PubMedCrossRef
102.
go back to reference Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S, RENAAL Study Investigators (2001) Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 345:861–869PubMedCrossRef Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S, RENAAL Study Investigators (2001) Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 345:861–869PubMedCrossRef
103.
go back to reference Lewis EJ, Hunsicker LG, Bain RP, Rohde RD (1993) The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med 329:1456–1462PubMedCrossRef Lewis EJ, Hunsicker LG, Bain RP, Rohde RD (1993) The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med 329:1456–1462PubMedCrossRef
104.
go back to reference Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz E, Atkins RC, Rohde R, Raz I, Collaborative Study Group (2001) Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med 345:851–860PubMedCrossRef Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz E, Atkins RC, Rohde R, Raz I, Collaborative Study Group (2001) Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med 345:851–860PubMedCrossRef
105.
go back to reference Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, Bittman R, Hurley S, Kleiman J, Gatlin M, Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study Investigators (2003) Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med 348:1309–1321PubMedCrossRef Pitt B, Remme W, Zannad F, Neaton J, Martinez F, Roniker B, Bittman R, Hurley S, Kleiman J, Gatlin M, Eplerenone Post-Acute Myocardial Infarction Heart Failure Efficacy and Survival Study Investigators (2003) Eplerenone, a selective aldosterone blocker, in patients with left ventricular dysfunction after myocardial infarction. N Engl J Med 348:1309–1321PubMedCrossRef
106.
go back to reference Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, Palensky J, Wittes J (1999) The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 341:709–717PubMedCrossRef Pitt B, Zannad F, Remme WJ, Cody R, Castaigne A, Perez A, Palensky J, Wittes J (1999) The effect of spironolactone on morbidity and mortality in patients with severe heart failure. Randomized Aldactone Evaluation Study Investigators. N Engl J Med 341:709–717PubMedCrossRef
107.
go back to reference Pitt B, Anker SD, Bushinsky DA, Kitzman DW, Zannad F, Huang IZ, PEARL-HF Investigators (2011) Evaluation of the efficacy and safety of RLY5016, a polymeric potassium binder, in a double-blind, placebo-controlled study in patients with chronic heart failure (the PEARL-HF) trial. Eur Heart J 32:820–828PubMedCrossRefPubMedCentral Pitt B, Anker SD, Bushinsky DA, Kitzman DW, Zannad F, Huang IZ, PEARL-HF Investigators (2011) Evaluation of the efficacy and safety of RLY5016, a polymeric potassium binder, in a double-blind, placebo-controlled study in patients with chronic heart failure (the PEARL-HF) trial. Eur Heart J 32:820–828PubMedCrossRefPubMedCentral
108.
go back to reference Weir MR, Bakris GL, Bushinsky DA, Mayo MR, Garza D, Stasiv Y, Wittes J, Christ-Schmidt H, Berman L, Pitt B, OPAL-HK Investigators (2015) Patiromer in patients with kidney disease and hyperkalemia receiving RAAS inhibitors. N Engl J Med 372:211–221PubMedCrossRef Weir MR, Bakris GL, Bushinsky DA, Mayo MR, Garza D, Stasiv Y, Wittes J, Christ-Schmidt H, Berman L, Pitt B, OPAL-HK Investigators (2015) Patiromer in patients with kidney disease and hyperkalemia receiving RAAS inhibitors. N Engl J Med 372:211–221PubMedCrossRef
109.
go back to reference Bakris GL, Pitt B, Weir MR, Freeman MW, Mayo MR, Garza D, Stasiv Y, Zawadzki R, Berman L, Bushinsky DA, AMETHYST-DN Investigators (2015) Effect of patiromer on serum potassium level in patients with hyperkalemia and diabetic kidney disease: the AMETHYST-DN randomized clinical trial. JAMA 314:151–161PubMedCrossRef Bakris GL, Pitt B, Weir MR, Freeman MW, Mayo MR, Garza D, Stasiv Y, Zawadzki R, Berman L, Bushinsky DA, AMETHYST-DN Investigators (2015) Effect of patiromer on serum potassium level in patients with hyperkalemia and diabetic kidney disease: the AMETHYST-DN randomized clinical trial. JAMA 314:151–161PubMedCrossRef
112.
go back to reference Stavros F, Yang A, Leon A, Nuttall M, Rasmussen HS (2014) Characterization of structure and function of ZS-9, a K+ selective ion trap. PLoS One 9:e114686PubMedCrossRefPubMedCentral Stavros F, Yang A, Leon A, Nuttall M, Rasmussen HS (2014) Characterization of structure and function of ZS-9, a K+ selective ion trap. PLoS One 9:e114686PubMedCrossRefPubMedCentral
113.
go back to reference Ash SR, Singh B, Lavin PT, Stavros F, Rasmussen HS (2015) A phase 2 study on the treatment of hyperkalemia in patients with chronic kidney disease suggests that the selective potassium trap, ZS-9, is safe and efficient. Kidney Int 88:404–411PubMedCrossRefPubMedCentral Ash SR, Singh B, Lavin PT, Stavros F, Rasmussen HS (2015) A phase 2 study on the treatment of hyperkalemia in patients with chronic kidney disease suggests that the selective potassium trap, ZS-9, is safe and efficient. Kidney Int 88:404–411PubMedCrossRefPubMedCentral
114.
go back to reference Packham DK, Rasmussen HS, Lavin PT, El-Shahawy MA, Roger SD, Block G, Qunibi W, Pergola P, Singh B (2015) Sodium zirconium cyclosilicate in hyperkalemia. N Engl J Med 372:222–231PubMedCrossRef Packham DK, Rasmussen HS, Lavin PT, El-Shahawy MA, Roger SD, Block G, Qunibi W, Pergola P, Singh B (2015) Sodium zirconium cyclosilicate in hyperkalemia. N Engl J Med 372:222–231PubMedCrossRef
115.
go back to reference Kosiborod M, Rasmussen HS, Lavin P, Qunibi WY, Spinowitz B, Packham D, Roger SD, Yang A, Lerma E, Singh B (2014) Effect of sodium zirconium cyclosilicate on potassium lowering for 28 days among outpatients with hyperkalemia: the HARMONIZE randomized clinical trial. JAMA 312:2223–2233PubMedCrossRef Kosiborod M, Rasmussen HS, Lavin P, Qunibi WY, Spinowitz B, Packham D, Roger SD, Yang A, Lerma E, Singh B (2014) Effect of sodium zirconium cyclosilicate on potassium lowering for 28 days among outpatients with hyperkalemia: the HARMONIZE randomized clinical trial. JAMA 312:2223–2233PubMedCrossRef
116.
go back to reference Kosiborod M, Peacock WF, Packham DK (2015) Sodium zirconium cyclosilicate for urgent therapy of severe hyperkalemia. N Engl J Med 372:1577–1578PubMedCrossRef Kosiborod M, Peacock WF, Packham DK (2015) Sodium zirconium cyclosilicate for urgent therapy of severe hyperkalemia. N Engl J Med 372:1577–1578PubMedCrossRef
117.
go back to reference Bushinsky DA, Williams GH, Pitt B, Weir MR, Freeman MW, Garza D, Stasiv Y, Li E, Berman L, Bakris GL (2015) Patiromer induces rapid and sustained potassium lowering in patients with chronic kidney disease and hyperkalemia. Kidney Int 88:1427–1433PubMedCrossRefPubMedCentral Bushinsky DA, Williams GH, Pitt B, Weir MR, Freeman MW, Garza D, Stasiv Y, Li E, Berman L, Bakris GL (2015) Patiromer induces rapid and sustained potassium lowering in patients with chronic kidney disease and hyperkalemia. Kidney Int 88:1427–1433PubMedCrossRefPubMedCentral
Metadata
Title
Potassium: friend or foe?
Author
Aylin R. Rodan
Publication date
01-07-2017
Publisher
Springer Berlin Heidelberg
Published in
Pediatric Nephrology / Issue 7/2017
Print ISSN: 0931-041X
Electronic ISSN: 1432-198X
DOI
https://doi.org/10.1007/s00467-016-3411-8

Other articles of this Issue 7/2017

Pediatric Nephrology 7/2017 Go to the issue