Skip to main content
Top
Published in: Pediatric Nephrology 2/2022

01-02-2022 | Review

Pathophysiological aspects of the thick ascending limb and novel genetic defects: HELIX syndrome and transient antenatal Bartter syndrome

Author: Rosa Vargas-Poussou

Published in: Pediatric Nephrology | Issue 2/2022

Login to get access

Abstract

The thick ascending limb plays a central role in human kidney physiology, participating in sodium reabsorption, urine concentrating mechanisms, calcium and magnesium homeostasis, bicarbonate and ammonium homeostasis, and uromodulin synthesis. This review aims to illustrate the importance of these roles from a pathophysiological point of view by describing the interactions of the key proteins of this segment and by discussing how recently identified and long-known hereditary diseases affect this segment. The descriptions of two recently described salt-losing tubulopathies, transient antenatal Bartter syndrome and HELIX syndrome, which are caused by mutations in MAGED2 and CLDN10 genes, respectively, highlight the role of new players in the modulation of sodium reabsorption the thick ascending limb.
Literature
2.
go back to reference Bankir L, Figueres L, Prot-Bertoye C, Bouby N, Crambert G, Pratt JH, Houillier P (2020) Medullary and cortical thick ascending limb: similarities and differences. Am J Physiol Ren Physiol 318:F422–F442CrossRef Bankir L, Figueres L, Prot-Bertoye C, Bouby N, Crambert G, Pratt JH, Houillier P (2020) Medullary and cortical thick ascending limb: similarities and differences. Am J Physiol Ren Physiol 318:F422–F442CrossRef
3.
go back to reference Bazua-Valenti S, Castaneda-Bueno M, Gamba G (2016) Physiological role of SLC12 family members in the kidney. Am J Physiol Ren Physiol 311:F131–F144CrossRef Bazua-Valenti S, Castaneda-Bueno M, Gamba G (2016) Physiological role of SLC12 family members in the kidney. Am J Physiol Ren Physiol 311:F131–F144CrossRef
4.
go back to reference Welling PA, Ho K (2009) A comprehensive guide to the ROMK potassium channel: form and function in health and disease. Am J Physiol Ren Physiol 297:F849–F863CrossRef Welling PA, Ho K (2009) A comprehensive guide to the ROMK potassium channel: form and function in health and disease. Am J Physiol Ren Physiol 297:F849–F863CrossRef
6.
go back to reference Castrop H, Schnermann J (2008) Isoforms of renal Na-K-2Cl cotransporter NKCC2: expression and functional significance. Am J Physiol Ren Physiol 295:F859–F866CrossRef Castrop H, Schnermann J (2008) Isoforms of renal Na-K-2Cl cotransporter NKCC2: expression and functional significance. Am J Physiol Ren Physiol 295:F859–F866CrossRef
7.
go back to reference Carota I, Theilig F, Oppermann M, Kongsuphol P, Rosenauer A, Schreiber R, Jensen BL, Walter S, Kunzelmann K, Castrop H (2010) Localization and functional characterization of the human NKCC2 isoforms. Acta Physiol (Oxford) 199:327–338 Carota I, Theilig F, Oppermann M, Kongsuphol P, Rosenauer A, Schreiber R, Jensen BL, Walter S, Kunzelmann K, Castrop H (2010) Localization and functional characterization of the human NKCC2 isoforms. Acta Physiol (Oxford) 199:327–338
8.
go back to reference Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Dominguez Rieg JA, Rieg T (2017) Renal tubular NHE3 is required in the maintenance of water and sodium chloride homeostasis. Kidney Int 92:397–414PubMedPubMedCentralCrossRef Fenton RA, Poulsen SB, de la Mora Chavez S, Soleimani M, Dominguez Rieg JA, Rieg T (2017) Renal tubular NHE3 is required in the maintenance of water and sodium chloride homeostasis. Kidney Int 92:397–414PubMedPubMedCentralCrossRef
9.
go back to reference Tsukita S, Tanaka H, Tamura A (2019) The claudins: from tight junctions to biological systems. Trends Biochem Sci 44:141–152PubMedCrossRef Tsukita S, Tanaka H, Tamura A (2019) The claudins: from tight junctions to biological systems. Trends Biochem Sci 44:141–152PubMedCrossRef
10.
go back to reference Gong Y, Hou J (2017) Claudins in barrier and transport function-the kidney. Pflugers Arch 469:105–113PubMedCrossRef Gong Y, Hou J (2017) Claudins in barrier and transport function-the kidney. Pflugers Arch 469:105–113PubMedCrossRef
11.
go back to reference Muto S (2017) Physiological roles of claudins in kidney tubule paracellular transport. Am J Physiol Ren Physiol 312:F9–F24CrossRef Muto S (2017) Physiological roles of claudins in kidney tubule paracellular transport. Am J Physiol Ren Physiol 312:F9–F24CrossRef
12.
go back to reference Prot-Bertoye C, Houillier P (2020) Claudins in renal physiology and pathology. Genes (Basel) 11:290CrossRef Prot-Bertoye C, Houillier P (2020) Claudins in renal physiology and pathology. Genes (Basel) 11:290CrossRef
13.
go back to reference Milatz S, Breiderhoff T (2017) One gene, two paracellular ion channels-claudin-10 in the kidney. Pflugers Arch 469:115–121PubMedCrossRef Milatz S, Breiderhoff T (2017) One gene, two paracellular ion channels-claudin-10 in the kidney. Pflugers Arch 469:115–121PubMedCrossRef
14.
go back to reference Plain A, Wulfmeyer VC, Milatz S, Klietz A, Hou J, Bleich M, Himmerkus N (2016) Corticomedullary difference in the effects of dietary Ca(2)(+) on tight junction properties in thick ascending limbs of Henle's loop. Pflugers Arch 468:293–303PubMedCrossRef Plain A, Wulfmeyer VC, Milatz S, Klietz A, Hou J, Bleich M, Himmerkus N (2016) Corticomedullary difference in the effects of dietary Ca(2)(+) on tight junction properties in thick ascending limbs of Henle's loop. Pflugers Arch 468:293–303PubMedCrossRef
15.
go back to reference Simon DB, Karet FE, Hamdan JM, DiPietro A, Sanjad SA, Lifton RP (1996) Bartter's syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotransporter NKCC2. Nat Genet 13:183–198PubMedCrossRef Simon DB, Karet FE, Hamdan JM, DiPietro A, Sanjad SA, Lifton RP (1996) Bartter's syndrome, hypokalaemic alkalosis with hypercalciuria, is caused by mutations in the Na-K-2Cl cotransporter NKCC2. Nat Genet 13:183–198PubMedCrossRef
16.
go back to reference Vargas-Poussou R, Feldmann D, Vollmer M, Konrad M, Kelly L, van den Heuvel LP, Tebourbi L, Brandis M, Karolyi L, Hebert SC, Lemmink HH, Deschenes G, Hildebrandt F, Seyberth HW, Guay-Woodford LM, Knoers NV, Antignac C (1998) Novel molecular variants of the Na-K-2Cl cotransporter gene are responsible for antenatal Bartter syndrome. Am J Hum Genet 62:1332–1340PubMedPubMedCentralCrossRef Vargas-Poussou R, Feldmann D, Vollmer M, Konrad M, Kelly L, van den Heuvel LP, Tebourbi L, Brandis M, Karolyi L, Hebert SC, Lemmink HH, Deschenes G, Hildebrandt F, Seyberth HW, Guay-Woodford LM, Knoers NV, Antignac C (1998) Novel molecular variants of the Na-K-2Cl cotransporter gene are responsible for antenatal Bartter syndrome. Am J Hum Genet 62:1332–1340PubMedPubMedCentralCrossRef
17.
go back to reference Simon DB, Karet FE, Rodriguez-Soriano J, Hamdan JH, DiPietro A, Trachtman H, Sanjad SA, Lifton RP (1996) Genetic heterogeneity of Bartter's syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet 14:152–156PubMedCrossRef Simon DB, Karet FE, Rodriguez-Soriano J, Hamdan JH, DiPietro A, Trachtman H, Sanjad SA, Lifton RP (1996) Genetic heterogeneity of Bartter's syndrome revealed by mutations in the K+ channel, ROMK. Nat Genet 14:152–156PubMedCrossRef
18.
go back to reference Simon DB, Bindra RS, Mansfield TA, Nelson-Williams C, Mendonca E, Stone R, Schurman S, Nayir A, Alpay H, Bakkaloglu A, Rodriguez-Soriano J, Morales JM, Sanjad SA, Taylor CM, Pilz D, Brem A, Trachtman H, Griswold W, Richard GA, John E, Lifton RP (1997) Mutations in the chloride channel gene, CLCNKB, cause Bartter's syndrome type III. Nat Genet 17:171–178PubMedCrossRef Simon DB, Bindra RS, Mansfield TA, Nelson-Williams C, Mendonca E, Stone R, Schurman S, Nayir A, Alpay H, Bakkaloglu A, Rodriguez-Soriano J, Morales JM, Sanjad SA, Taylor CM, Pilz D, Brem A, Trachtman H, Griswold W, Richard GA, John E, Lifton RP (1997) Mutations in the chloride channel gene, CLCNKB, cause Bartter's syndrome type III. Nat Genet 17:171–178PubMedCrossRef
19.
go back to reference Seys E, Andrini O, Keck M, Mansour-Hendili L, Courand PY, Simian C, Deschenes G, Kwon T, Bertholet-Thomas A, Bobrie G, Borde JS, Bourdat-Michel G, Decramer S, Cailliez M, Krug P, Cozette P, Delbet JD, Dubourg L, Chaveau D, Fila M, Jourde-Chiche N, Knebelmann B, Lavocat MP, Lemoine S, Djeddi D, Llanas B, Louillet F, Merieau E, Mileva M, Mota-Vieira L, Mousson C, Nobili F, Novo R, Roussey-Kesler G, Vrillon I, Walsh SB, Teulon J, Blanchard A, Vargas-Poussou R (2017) Clinical and genetic spectrum of Bartter syndrome type 3. J Am Soc Nephrol 28:2540–2552PubMedPubMedCentralCrossRef Seys E, Andrini O, Keck M, Mansour-Hendili L, Courand PY, Simian C, Deschenes G, Kwon T, Bertholet-Thomas A, Bobrie G, Borde JS, Bourdat-Michel G, Decramer S, Cailliez M, Krug P, Cozette P, Delbet JD, Dubourg L, Chaveau D, Fila M, Jourde-Chiche N, Knebelmann B, Lavocat MP, Lemoine S, Djeddi D, Llanas B, Louillet F, Merieau E, Mileva M, Mota-Vieira L, Mousson C, Nobili F, Novo R, Roussey-Kesler G, Vrillon I, Walsh SB, Teulon J, Blanchard A, Vargas-Poussou R (2017) Clinical and genetic spectrum of Bartter syndrome type 3. J Am Soc Nephrol 28:2540–2552PubMedPubMedCentralCrossRef
20.
go back to reference Birkenhager R, Otto E, Schurmann MJ, Vollmer M, Ruf EM, Maier-Lutz I, Beekmann F, Fekete A, Omran H, Feldmann D, Milford DV, Jeck N, Konrad M, Landau D, Knoers NV, Antignac C, Sudbrak R, Kispert A, Hildebrandt F (2001) Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet 29:310–314PubMedCrossRef Birkenhager R, Otto E, Schurmann MJ, Vollmer M, Ruf EM, Maier-Lutz I, Beekmann F, Fekete A, Omran H, Feldmann D, Milford DV, Jeck N, Konrad M, Landau D, Knoers NV, Antignac C, Sudbrak R, Kispert A, Hildebrandt F (2001) Mutation of BSND causes Bartter syndrome with sensorineural deafness and kidney failure. Nat Genet 29:310–314PubMedCrossRef
21.
go back to reference Schlingmann KP, Konrad M, Jeck N, Waldegger P, Reinalter SC, Holder M, Seyberth HW, Waldegger S (2004) Salt wasting and deafness resulting from mutations in two chloride channels. N Engl J Med 350:1314–1319PubMedCrossRef Schlingmann KP, Konrad M, Jeck N, Waldegger P, Reinalter SC, Holder M, Seyberth HW, Waldegger S (2004) Salt wasting and deafness resulting from mutations in two chloride channels. N Engl J Med 350:1314–1319PubMedCrossRef
22.
go back to reference Laghmani K, Beck BB, Yang SS, Seaayfan E, Wenzel A, Reusch B, Vitzthum H, Priem D, Demaretz S, Bergmann K, Duin LK, Gobel H, Mache C, Thiele H, Bartram MP, Dombret C, Altmuller J, Nurnberg P, Benzing T, Levtchenko E, Seyberth HW, Klaus G, Yigit G, Lin SH, Timmer A, de Koning TJ, Scherjon SA, Schlingmann KP, Bertrand MJ, Rinschen MM, de Backer O, Konrad M, Komhoff M (2016) Polyhydramnios, transient antenatal Bartter's syndrome, and MAGED2 mutations. N Engl J Med 374:1853–1863PubMedCrossRef Laghmani K, Beck BB, Yang SS, Seaayfan E, Wenzel A, Reusch B, Vitzthum H, Priem D, Demaretz S, Bergmann K, Duin LK, Gobel H, Mache C, Thiele H, Bartram MP, Dombret C, Altmuller J, Nurnberg P, Benzing T, Levtchenko E, Seyberth HW, Klaus G, Yigit G, Lin SH, Timmer A, de Koning TJ, Scherjon SA, Schlingmann KP, Bertrand MJ, Rinschen MM, de Backer O, Konrad M, Komhoff M (2016) Polyhydramnios, transient antenatal Bartter's syndrome, and MAGED2 mutations. N Engl J Med 374:1853–1863PubMedCrossRef
23.
go back to reference Bongers E, Shelton LM, Milatz S, Verkaart S, Bech AP, Schoots J, Cornelissen EAM, Bleich M, Hoenderop JGJ, Wetzels JFM, Lugtenberg D, Nijenhuis T (2017) A novel hypokalemic-alkalotic salt-losing tubulopathy in patients with CLDN10 mutations. J Am Soc Nephrol 28:3118–3128PubMedPubMedCentralCrossRef Bongers E, Shelton LM, Milatz S, Verkaart S, Bech AP, Schoots J, Cornelissen EAM, Bleich M, Hoenderop JGJ, Wetzels JFM, Lugtenberg D, Nijenhuis T (2017) A novel hypokalemic-alkalotic salt-losing tubulopathy in patients with CLDN10 mutations. J Am Soc Nephrol 28:3118–3128PubMedPubMedCentralCrossRef
24.
go back to reference Hadj-Rabia S, Brideau G, Al-Sarraj Y, Maroun RC, Figueres ML, Leclerc-Mercier S, Olinger E, Baron S, Chaussain C, Nochy D, Taha RZ, Knebelmann B, Joshi V, Curmi PA, Kambouris M, Vargas-Poussou R, Bodemer C, Devuyst O, Houillier P, El-Shanti H (2018) Multiplex epithelium dysfunction due to CLDN10 mutation: the HELIX syndrome. Genet Med 20:190–201PubMedCrossRef Hadj-Rabia S, Brideau G, Al-Sarraj Y, Maroun RC, Figueres ML, Leclerc-Mercier S, Olinger E, Baron S, Chaussain C, Nochy D, Taha RZ, Knebelmann B, Joshi V, Curmi PA, Kambouris M, Vargas-Poussou R, Bodemer C, Devuyst O, Houillier P, El-Shanti H (2018) Multiplex epithelium dysfunction due to CLDN10 mutation: the HELIX syndrome. Genet Med 20:190–201PubMedCrossRef
25.
go back to reference Klar J, Piontek J, Milatz S, Tariq M, Jameel M, Breiderhoff T, Schuster J, Fatima A, Asif M, Sher M, Mabert K, Fromm A, Baig SM, Gunzel D, Dahl N (2017) Altered paracellular cation permeability due to a rare CLDN10B variant causes anhidrosis and kidney damage. PLoS Genet 13:e1006897PubMedPubMedCentralCrossRef Klar J, Piontek J, Milatz S, Tariq M, Jameel M, Breiderhoff T, Schuster J, Fatima A, Asif M, Sher M, Mabert K, Fromm A, Baig SM, Gunzel D, Dahl N (2017) Altered paracellular cation permeability due to a rare CLDN10B variant causes anhidrosis and kidney damage. PLoS Genet 13:e1006897PubMedPubMedCentralCrossRef
26.
go back to reference Hou J, Renigunta A, Konrad M, Gomes AS, Schneeberger EE, Paul DL, Waldegger S, Goodenough DA (2008) Claudin-16 and claudin-19 interact and form a cation-selective tight junction complex. J Clin Invest 118:619–628PubMedPubMedCentral Hou J, Renigunta A, Konrad M, Gomes AS, Schneeberger EE, Paul DL, Waldegger S, Goodenough DA (2008) Claudin-16 and claudin-19 interact and form a cation-selective tight junction complex. J Clin Invest 118:619–628PubMedPubMedCentral
27.
go back to reference Gong Y, Renigunta V, Himmerkus N, Zhang J, Renigunta A, Bleich M, Hou J (2012) Claudin-14 regulates renal Ca(+)(+) transport in response to CaSR signalling via a novel microRNA pathway. EMBO J 31:1999–2012PubMedPubMedCentralCrossRef Gong Y, Renigunta V, Himmerkus N, Zhang J, Renigunta A, Bleich M, Hou J (2012) Claudin-14 regulates renal Ca(+)(+) transport in response to CaSR signalling via a novel microRNA pathway. EMBO J 31:1999–2012PubMedPubMedCentralCrossRef
28.
go back to reference Dimke H, Desai P, Borovac J, Lau A, Pan W, Alexander RT (2013) Activation of the Ca(2+)-sensing receptor increases renal claudin-14 expression and urinary Ca(2+) excretion. Am J Physiol Ren Physiol 304:F761–F769CrossRef Dimke H, Desai P, Borovac J, Lau A, Pan W, Alexander RT (2013) Activation of the Ca(2+)-sensing receptor increases renal claudin-14 expression and urinary Ca(2+) excretion. Am J Physiol Ren Physiol 304:F761–F769CrossRef
29.
go back to reference Gong Y, Hou J (2014) Claudin-14 underlies Ca(+)(+)-sensing receptor-mediated Ca(+)(+) metabolism via NFAT-microRNA-based mechanisms. J Am Soc Nephrol 25:745–760PubMedCrossRef Gong Y, Hou J (2014) Claudin-14 underlies Ca(+)(+)-sensing receptor-mediated Ca(+)(+) metabolism via NFAT-microRNA-based mechanisms. J Am Soc Nephrol 25:745–760PubMedCrossRef
30.
go back to reference Simon DB, Lu Y, Choate KA, Velazquez H, Al-Sabban E, Praga M, Casari G, Bettinelli A, Colussi G, Rodriguez-Soriano J, McCredie D, Milford D, Sanjad S, Lifton RP (1999) Paracellin-1, a renal tight junction protein required for paracellular Mg2+ resorption. Science 285:103–106PubMedCrossRef Simon DB, Lu Y, Choate KA, Velazquez H, Al-Sabban E, Praga M, Casari G, Bettinelli A, Colussi G, Rodriguez-Soriano J, McCredie D, Milford D, Sanjad S, Lifton RP (1999) Paracellin-1, a renal tight junction protein required for paracellular Mg2+ resorption. Science 285:103–106PubMedCrossRef
31.
go back to reference Konrad M, Schaller A, Seelow D, Pandey AV, Waldegger S, Lesslauer A, Vitzthum H, Suzuki Y, Luk JM, Becker C, Schlingmann KP, Schmid M, Rodriguez-Soriano J, Ariceta G, Cano F, Enriquez R, Juppner H, Bakkaloglu SA, Hediger MA, Gallati S, Neuhauss SC, Nurnberg P, Weber S (2006) Mutations in the tight-junction gene claudin 19 (CLDN19) are associated with renal magnesium wasting, renal failure, and severe ocular involvement. Am J Hum Genet 79:949–957PubMedPubMedCentralCrossRef Konrad M, Schaller A, Seelow D, Pandey AV, Waldegger S, Lesslauer A, Vitzthum H, Suzuki Y, Luk JM, Becker C, Schlingmann KP, Schmid M, Rodriguez-Soriano J, Ariceta G, Cano F, Enriquez R, Juppner H, Bakkaloglu SA, Hediger MA, Gallati S, Neuhauss SC, Nurnberg P, Weber S (2006) Mutations in the tight-junction gene claudin 19 (CLDN19) are associated with renal magnesium wasting, renal failure, and severe ocular involvement. Am J Hum Genet 79:949–957PubMedPubMedCentralCrossRef
32.
go back to reference Bardet C, Courson F, Wu Y, Khaddam M, Salmon B, Ribes S, Thumfart J, Yamaguti PM, Rochefort GY, Figueres ML, Breiderhoff T, Garcia-Castano A, Vallee B, Le Denmat D, Baroukh B, Guilbert T, Schmitt A, Masse JM, Bazin D, Lorenz G, Morawietz M, Hou J, Carvalho-Lobato P, Manzanares MC, Fricain JC, Talmud D, Demontis R, Neves F, Zenaty D, Berdal A, Kiesow A, Petzold M, Menashi S, Linglart A, Acevedo AC, Vargas-Poussou R, Muller D, Houillier P, Chaussain C (2016) Claudin-16 deficiency impairs tight junction function in ameloblasts, leading to abnormal enamel formation. J Bone Miner Res 31:498–513PubMedCrossRef Bardet C, Courson F, Wu Y, Khaddam M, Salmon B, Ribes S, Thumfart J, Yamaguti PM, Rochefort GY, Figueres ML, Breiderhoff T, Garcia-Castano A, Vallee B, Le Denmat D, Baroukh B, Guilbert T, Schmitt A, Masse JM, Bazin D, Lorenz G, Morawietz M, Hou J, Carvalho-Lobato P, Manzanares MC, Fricain JC, Talmud D, Demontis R, Neves F, Zenaty D, Berdal A, Kiesow A, Petzold M, Menashi S, Linglart A, Acevedo AC, Vargas-Poussou R, Muller D, Houillier P, Chaussain C (2016) Claudin-16 deficiency impairs tight junction function in ameloblasts, leading to abnormal enamel formation. J Bone Miner Res 31:498–513PubMedCrossRef
33.
go back to reference Yamaguti PM, Neves FA, Hotton D, Bardet C, de La Dure-Molla M, Castro LC, Scher MD, Barbosa ME, Ditsch C, Fricain JC, de La Faille R, Figueres ML, Vargas-Poussou R, Houillier P, Chaussain C, Babajko S, Berdal A, Acevedo AC (2017) Amelogenesis imperfecta in familial hypomagnesaemia and hypercalciuria with nephrocalcinosis caused by CLDN19 gene mutations. J Med Genet 54:26–37PubMedCrossRef Yamaguti PM, Neves FA, Hotton D, Bardet C, de La Dure-Molla M, Castro LC, Scher MD, Barbosa ME, Ditsch C, Fricain JC, de La Faille R, Figueres ML, Vargas-Poussou R, Houillier P, Chaussain C, Babajko S, Berdal A, Acevedo AC (2017) Amelogenesis imperfecta in familial hypomagnesaemia and hypercalciuria with nephrocalcinosis caused by CLDN19 gene mutations. J Med Genet 54:26–37PubMedCrossRef
34.
go back to reference Hannan FM, Babinsky VN, Thakker RV (2016) Disorders of the calcium-sensing receptor and partner proteins: insights into the molecular basis of calcium homeostasis. J Mol Endocrinol 57:R127–R142PubMedPubMedCentralCrossRef Hannan FM, Babinsky VN, Thakker RV (2016) Disorders of the calcium-sensing receptor and partner proteins: insights into the molecular basis of calcium homeostasis. J Mol Endocrinol 57:R127–R142PubMedPubMedCentralCrossRef
35.
go back to reference Watanabe S, Fukumoto S, Chang H, Takeuchi Y, Hasegawa Y, Okazaki R, Chikatsu N, Fujita T (2002) Association between activating mutations of calcium-sensing receptor and Bartter's syndrome. Lancet 360:692–694PubMedCrossRef Watanabe S, Fukumoto S, Chang H, Takeuchi Y, Hasegawa Y, Okazaki R, Chikatsu N, Fujita T (2002) Association between activating mutations of calcium-sensing receptor and Bartter's syndrome. Lancet 360:692–694PubMedCrossRef
36.
go back to reference Vargas-Poussou R, Huang C, Hulin P, Houillier P, Jeunemaitre X, Paillard M, Planelles G, Dechaux M, Miller RT, Antignac C (2002) Functional characterization of a calcium-sensing receptor mutation in severe autosomal dominant hypocalcemia with a Bartter-like syndrome. J Am Soc Nephrol 13:2259–2266PubMedCrossRef Vargas-Poussou R, Huang C, Hulin P, Houillier P, Jeunemaitre X, Paillard M, Planelles G, Dechaux M, Miller RT, Antignac C (2002) Functional characterization of a calcium-sensing receptor mutation in severe autosomal dominant hypocalcemia with a Bartter-like syndrome. J Am Soc Nephrol 13:2259–2266PubMedCrossRef
37.
go back to reference Thorleifsson G, Holm H, Edvardsson V, Walters GB, Styrkarsdottir U, Gudbjartsson DF, Sulem P, Halldorsson BV, de Vegt F, d'Ancona FC, den Heijer M, Franzson L, Christiansen C, Alexandersen P, Rafnar T, Kristjansson K, Sigurdsson G, Kiemeney LA, Bodvarsson M, Indridason OS, Palsson R, Kong A, Thorsteinsdottir U, Stefansson K (2009) Sequence variants in the CLDN14 gene associate with kidney stones and bone mineral density. Nat Genet 41:926–930PubMedCrossRef Thorleifsson G, Holm H, Edvardsson V, Walters GB, Styrkarsdottir U, Gudbjartsson DF, Sulem P, Halldorsson BV, de Vegt F, d'Ancona FC, den Heijer M, Franzson L, Christiansen C, Alexandersen P, Rafnar T, Kristjansson K, Sigurdsson G, Kiemeney LA, Bodvarsson M, Indridason OS, Palsson R, Kong A, Thorsteinsdottir U, Stefansson K (2009) Sequence variants in the CLDN14 gene associate with kidney stones and bone mineral density. Nat Genet 41:926–930PubMedCrossRef
38.
go back to reference Guha M, Bankura B, Ghosh S, Pattanayak AK, Ghosh S, Pal DK, Puri A, Kundu AK, Das M (2015) Polymorphisms in CaSR and CLDN14 genes associated with increased risk of kidney stone disease in patients from the eastern part of India. PLoS One 10:e0130790PubMedPubMedCentralCrossRef Guha M, Bankura B, Ghosh S, Pattanayak AK, Ghosh S, Pal DK, Puri A, Kundu AK, Das M (2015) Polymorphisms in CaSR and CLDN14 genes associated with increased risk of kidney stone disease in patients from the eastern part of India. PLoS One 10:e0130790PubMedPubMedCentralCrossRef
39.
go back to reference Corre T, Olinger E, Harris SE, Traglia M, Ulivi S, Lenarduzzi S, Belge H, Youhanna S, Tokonami N, Bonny O, Houillier P, Polasek O, Deary IJ, Starr JM, Toniolo D, Gasparini P, Vollenweider P, Hayward C, Bochud M, Devuyst O (2017) Common variants in CLDN14 are associated with differential excretion of magnesium over calcium in urine. Pflugers Arch 469:91–103PubMedCrossRef Corre T, Olinger E, Harris SE, Traglia M, Ulivi S, Lenarduzzi S, Belge H, Youhanna S, Tokonami N, Bonny O, Houillier P, Polasek O, Deary IJ, Starr JM, Toniolo D, Gasparini P, Vollenweider P, Hayward C, Bochud M, Devuyst O (2017) Common variants in CLDN14 are associated with differential excretion of magnesium over calcium in urine. Pflugers Arch 469:91–103PubMedCrossRef
40.
go back to reference Good DW (1993) The thick ascending limb as a site of renal bicarbonate reabsorption. Semin Nephrol 13:225–235PubMed Good DW (1993) The thick ascending limb as a site of renal bicarbonate reabsorption. Semin Nephrol 13:225–235PubMed
41.
go back to reference Wang T, Hropot M, Aronson PS, Giebisch G (2001) Role of NHE isoforms in mediating bicarbonate reabsorption along the nephron. Am J Physiol Ren Physiol 281:F1117–F1122CrossRef Wang T, Hropot M, Aronson PS, Giebisch G (2001) Role of NHE isoforms in mediating bicarbonate reabsorption along the nephron. Am J Physiol Ren Physiol 281:F1117–F1122CrossRef
43.
go back to reference Hart TC, Gorry MC, Hart PS, Woodard AS, Shihabi Z, Sandhu J, Shirts B, Xu L, Zhu H, Barmada MM, Bleyer AJ (2002) Mutations of the UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet 39:882–892PubMedPubMedCentralCrossRef Hart TC, Gorry MC, Hart PS, Woodard AS, Shihabi Z, Sandhu J, Shirts B, Xu L, Zhu H, Barmada MM, Bleyer AJ (2002) Mutations of the UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet 39:882–892PubMedPubMedCentralCrossRef
44.
go back to reference Devuyst O, Olinger E, Rampoldi L (2017) Uromodulin: from physiology to rare and complex kidney disorders. Nat Rev Nephrol 13:525–544PubMedCrossRef Devuyst O, Olinger E, Rampoldi L (2017) Uromodulin: from physiology to rare and complex kidney disorders. Nat Rev Nephrol 13:525–544PubMedCrossRef
45.
go back to reference Mutig K, Kahl T, Saritas T, Godes M, Persson P, Bates J, Raffi H, Rampoldi L, Uchida S, Hille C, Dosche C, Kumar S, Castaneda-Bueno M, Gamba G, Bachmann S (2011) Activation of the bumetanide-sensitive Na+,K+,2Cl- cotransporter (NKCC2) is facilitated by Tamm–Horsfall protein in a chloride-sensitive manner. J Biol Chem 286:30200–30210PubMedPubMedCentralCrossRef Mutig K, Kahl T, Saritas T, Godes M, Persson P, Bates J, Raffi H, Rampoldi L, Uchida S, Hille C, Dosche C, Kumar S, Castaneda-Bueno M, Gamba G, Bachmann S (2011) Activation of the bumetanide-sensitive Na+,K+,2Cl- cotransporter (NKCC2) is facilitated by Tamm–Horsfall protein in a chloride-sensitive manner. J Biol Chem 286:30200–30210PubMedPubMedCentralCrossRef
46.
go back to reference Renigunta A, Renigunta V, Saritas T, Decher N, Mutig K, Waldegger S (2011) Tamm–Horsfall glycoprotein interacts with renal outer medullary potassium channel ROMK2 and regulates its function. J Biol Chem 286:2224–2235PubMedCrossRef Renigunta A, Renigunta V, Saritas T, Decher N, Mutig K, Waldegger S (2011) Tamm–Horsfall glycoprotein interacts with renal outer medullary potassium channel ROMK2 and regulates its function. J Biol Chem 286:2224–2235PubMedCrossRef
47.
go back to reference Bachmann S, Mutig K, Bates J, Welker P, Geist B, Gross V, Luft FC, Alenina N, Bader M, Thiele BJ, Prasadan K, Raffi HS, Kumar S (2005) Renal effects of Tamm–Horsfall protein (uromodulin) deficiency in mice. Am J Physiol Ren Physiol 288:F559–F567CrossRef Bachmann S, Mutig K, Bates J, Welker P, Geist B, Gross V, Luft FC, Alenina N, Bader M, Thiele BJ, Prasadan K, Raffi HS, Kumar S (2005) Renal effects of Tamm–Horsfall protein (uromodulin) deficiency in mice. Am J Physiol Ren Physiol 288:F559–F567CrossRef
48.
go back to reference Graham LA, Padmanabhan S, Fraser NJ, Kumar S, Bates JM, Raffi HS, Welsh P, Beattie W, Hao S, Leh S, Hultstrom M, Ferreri NR, Dominiczak AF, Graham D, McBride MW (2014) Validation of uromodulin as a candidate gene for human essential hypertension. Hypertension 63:551–558PubMedCrossRef Graham LA, Padmanabhan S, Fraser NJ, Kumar S, Bates JM, Raffi HS, Welsh P, Beattie W, Hao S, Leh S, Hultstrom M, Ferreri NR, Dominiczak AF, Graham D, McBride MW (2014) Validation of uromodulin as a candidate gene for human essential hypertension. Hypertension 63:551–558PubMedCrossRef
49.
go back to reference Devuyst O, Olinger E, Weber S, Eckardt KU, Kmoch S, Rampoldi L, Bleyer AJ (2019) Autosomal dominant tubulointerstitial kidney disease. Nat Rev Dis Primers 5:60PubMedCrossRef Devuyst O, Olinger E, Weber S, Eckardt KU, Kmoch S, Rampoldi L, Bleyer AJ (2019) Autosomal dominant tubulointerstitial kidney disease. Nat Rev Dis Primers 5:60PubMedCrossRef
50.
go back to reference Legrand A, Treard C, Roncelin I, Dreux S, Bertholet-Thomas A, Broux F, Bruno D, Decramer S, Deschenes G, Djeddi D, Guigonis V, Jay N, Khalifeh T, Llanas B, Morin D, Morin G, Nobili F, Pietrement C, Ryckewaert A, Salomon R, Vrillon I, Blanchard A, Vargas-Poussou R (2018) Prevalence of novel MAGED2 mutations in antenatal Bartter syndrome. Clin J Am Soc Nephrol 13:242–250PubMedCrossRef Legrand A, Treard C, Roncelin I, Dreux S, Bertholet-Thomas A, Broux F, Bruno D, Decramer S, Deschenes G, Djeddi D, Guigonis V, Jay N, Khalifeh T, Llanas B, Morin D, Morin G, Nobili F, Pietrement C, Ryckewaert A, Salomon R, Vrillon I, Blanchard A, Vargas-Poussou R (2018) Prevalence of novel MAGED2 mutations in antenatal Bartter syndrome. Clin J Am Soc Nephrol 13:242–250PubMedCrossRef
51.
go back to reference Arthuis CJ, Nizon M, Komhoff M, Beck BB, Riehmer V, Bihouee T, Bruel A, Benbrik N, Winer N, Isidor B (2018) A step towards precision medicine in management of severe transient polyhydramnios: MAGED2 variant. J Obstet Gynaecol 39:395–397PubMedCrossRef Arthuis CJ, Nizon M, Komhoff M, Beck BB, Riehmer V, Bihouee T, Bruel A, Benbrik N, Winer N, Isidor B (2018) A step towards precision medicine in management of severe transient polyhydramnios: MAGED2 variant. J Obstet Gynaecol 39:395–397PubMedCrossRef
53.
go back to reference Florke Gee RR, Chen H, Lee AK, Daly CA, Wilander BA, Fon Tacer K, Potts PR (2020) Emerging roles of the MAGE protein family in stress response pathways. J Biol Chem 295:16121–16155PubMedPubMedCentralCrossRef Florke Gee RR, Chen H, Lee AK, Daly CA, Wilander BA, Fon Tacer K, Potts PR (2020) Emerging roles of the MAGE protein family in stress response pathways. J Biol Chem 295:16121–16155PubMedPubMedCentralCrossRef
55.
go back to reference Valino-Rivas L, Cuarental L, Agustin M, Husi H, Cannata-Ortiz P, Sanz AB, Mischak H, Ortiz A, Sanchez-Nino MD (2019) MAGE genes in the kidney: identification of MAGED2 as upregulated during kidney injury and in stressed tubular cells. Nephrol Dial Transplant 34:1498–1507PubMedCrossRef Valino-Rivas L, Cuarental L, Agustin M, Husi H, Cannata-Ortiz P, Sanz AB, Mischak H, Ortiz A, Sanchez-Nino MD (2019) MAGE genes in the kidney: identification of MAGED2 as upregulated during kidney injury and in stressed tubular cells. Nephrol Dial Transplant 34:1498–1507PubMedCrossRef
56.
go back to reference Breiderhoff T, Himmerkus N, Stuiver M, Mutig K, Will C, Meij IC, Bachmann S, Bleich M, Willnow TE, Muller D (2012) Deletion of claudin-10 (Cldn10) in the thick ascending limb impairs paracellular sodium permeability and leads to hypermagnesemia and nephrocalcinosis. Proc Natl Acad Sci U S A 109:14241–14246PubMedPubMedCentralCrossRef Breiderhoff T, Himmerkus N, Stuiver M, Mutig K, Will C, Meij IC, Bachmann S, Bleich M, Willnow TE, Muller D (2012) Deletion of claudin-10 (Cldn10) in the thick ascending limb impairs paracellular sodium permeability and leads to hypermagnesemia and nephrocalcinosis. Proc Natl Acad Sci U S A 109:14241–14246PubMedPubMedCentralCrossRef
57.
go back to reference Van Itallie CM, Rogan S, Yu A, Vidal LS, Holmes J, Anderson JM (2006) Two splice variants of claudin-10 in the kidney create paracellular pores with different ion selectivities. Am J Physiol Ren Physiol 291:F1288–F1299CrossRef Van Itallie CM, Rogan S, Yu A, Vidal LS, Holmes J, Anderson JM (2006) Two splice variants of claudin-10 in the kidney create paracellular pores with different ion selectivities. Am J Physiol Ren Physiol 291:F1288–F1299CrossRef
58.
go back to reference Gunzel D, Stuiver M, Kausalya PJ, Haisch L, Krug SM, Rosenthal R, Meij IC, Hunziker W, Fromm M, Muller D (2009) Claudin-10 exists in six alternatively spliced isoforms that exhibit distinct localization and function. J Cell Sci 122:1507–1517PubMedCrossRef Gunzel D, Stuiver M, Kausalya PJ, Haisch L, Krug SM, Rosenthal R, Meij IC, Hunziker W, Fromm M, Muller D (2009) Claudin-10 exists in six alternatively spliced isoforms that exhibit distinct localization and function. J Cell Sci 122:1507–1517PubMedCrossRef
59.
go back to reference Inai T, Sengoku A, Guan X, Hirose E, Iida H, Shibata Y (2005) Heterogeneity in expression and subcellular localization of tight junction proteins, claudin-10 and -15, examined by RT-PCR and immunofluorescence microscopy. Arch Histol Cytol 68:349–360PubMedCrossRef Inai T, Sengoku A, Guan X, Hirose E, Iida H, Shibata Y (2005) Heterogeneity in expression and subcellular localization of tight junction proteins, claudin-10 and -15, examined by RT-PCR and immunofluorescence microscopy. Arch Histol Cytol 68:349–360PubMedCrossRef
60.
go back to reference Meyers N, Nelson-Williams C, Malaga-Dieguez L, Kaufmann H, Loring E, Knight J, Lifton RP, Trachtman H (2019) Hypokalemia associated with a claudin 10 mutation: a case report. Am J Kidney Dis 73:425–428PubMedCrossRef Meyers N, Nelson-Williams C, Malaga-Dieguez L, Kaufmann H, Loring E, Knight J, Lifton RP, Trachtman H (2019) Hypokalemia associated with a claudin 10 mutation: a case report. Am J Kidney Dis 73:425–428PubMedCrossRef
62.
go back to reference Breiderhoff T, Himmerkus N, Drewell H, Plain A, Gunzel D, Mutig K, Willnow TE, Muller D, Bleich M (2018) Deletion of claudin-10 rescues claudin-16-deficient mice from hypomagnesemia and hypercalciuria. Kidney Int 93:580–588PubMedCrossRef Breiderhoff T, Himmerkus N, Drewell H, Plain A, Gunzel D, Mutig K, Willnow TE, Muller D, Bleich M (2018) Deletion of claudin-10 rescues claudin-16-deficient mice from hypomagnesemia and hypercalciuria. Kidney Int 93:580–588PubMedCrossRef
63.
64.
go back to reference Pedersen AML, Sorensen CE, Proctor GB, Carpenter GH, Ekstrom J (2018) Salivary secretion in health and disease. J Oral Rehabil 45:730–746PubMedCrossRef Pedersen AML, Sorensen CE, Proctor GB, Carpenter GH, Ekstrom J (2018) Salivary secretion in health and disease. J Oral Rehabil 45:730–746PubMedCrossRef
67.
go back to reference Delporte C (2009) Aquaporins in secretory glands and their role in Sjogren's syndrome. Handb Exp Pharmacol 190:185–201CrossRef Delporte C (2009) Aquaporins in secretory glands and their role in Sjogren's syndrome. Handb Exp Pharmacol 190:185–201CrossRef
68.
go back to reference Sato T, Courbebaisse M, Ide N, Fan Y, Hanai JI, Kaludjerovic J, Densmore MJ, Yuan Q, Toka HR, Pollak MR, Hou J, Lanske B (2017) Parathyroid hormone controls paracellular Ca(2+) transport in the thick ascending limb by regulating the tight-junction protein Claudin14. Proc Natl Acad Sci U S A 114:E3344–E3353PubMedPubMedCentralCrossRef Sato T, Courbebaisse M, Ide N, Fan Y, Hanai JI, Kaludjerovic J, Densmore MJ, Yuan Q, Toka HR, Pollak MR, Hou J, Lanske B (2017) Parathyroid hormone controls paracellular Ca(2+) transport in the thick ascending limb by regulating the tight-junction protein Claudin14. Proc Natl Acad Sci U S A 114:E3344–E3353PubMedPubMedCentralCrossRef
69.
go back to reference Kompatscher A, de Baaij JHF, Aboudehen K, Farahani S, van Son LHJ, Milatz S, Himmerkus N, Veenstra GC, Bindels RJM, Hoenderop JGJ (2018) Transcription factor HNF1beta regulates expression of the calcium-sensing receptor in the thick ascending limb of the kidney. Am J Physiol Ren Physiol 315:F27–F35CrossRef Kompatscher A, de Baaij JHF, Aboudehen K, Farahani S, van Son LHJ, Milatz S, Himmerkus N, Veenstra GC, Bindels RJM, Hoenderop JGJ (2018) Transcription factor HNF1beta regulates expression of the calcium-sensing receptor in the thick ascending limb of the kidney. Am J Physiol Ren Physiol 315:F27–F35CrossRef
70.
go back to reference Tokonami N, Olinger E, Debaix H, Houillier P, Devuyst O (2018) The excretion of uromodulin is modulated by the calcium-sensing receptor. Kidney Int 94:882–886PubMedCrossRef Tokonami N, Olinger E, Debaix H, Houillier P, Devuyst O (2018) The excretion of uromodulin is modulated by the calcium-sensing receptor. Kidney Int 94:882–886PubMedCrossRef
71.
go back to reference Hou J, Renigunta V, Nie M, Sunq A, Himmerkus N, Quintanova C, Bleich M, Renigunta A, Wolf MTF (2019) Phosphorylated claudin-16 interacts with Trpv5 and regulates transcellular calcium transport in the kidney. Proc Natl Acad Sci U S A 116:19176–19186PubMedPubMedCentralCrossRef Hou J, Renigunta V, Nie M, Sunq A, Himmerkus N, Quintanova C, Bleich M, Renigunta A, Wolf MTF (2019) Phosphorylated claudin-16 interacts with Trpv5 and regulates transcellular calcium transport in the kidney. Proc Natl Acad Sci U S A 116:19176–19186PubMedPubMedCentralCrossRef
72.
go back to reference Muller D, Kausalya PJ, Claverie-Martin F, Meij IC, Eggert P, Garcia-Nieto V, Hunziker W (2003) A novel claudin 16 mutation associated with childhood hypercalciuria abolishes binding to ZO-1 and results in lysosomal mistargeting. Am J Hum Genet 73:1293–1301PubMedPubMedCentralCrossRef Muller D, Kausalya PJ, Claverie-Martin F, Meij IC, Eggert P, Garcia-Nieto V, Hunziker W (2003) A novel claudin 16 mutation associated with childhood hypercalciuria abolishes binding to ZO-1 and results in lysosomal mistargeting. Am J Hum Genet 73:1293–1301PubMedPubMedCentralCrossRef
73.
go back to reference Bockenhauer D, Feather S, Stanescu HC, Bandulik S, Zdebik AA, Reichold M, Tobin J, Lieberer E, Sterner C, Landoure G, Arora R, Sirimanna T, Thompson D, Cross JH, van't Hoff W, Al Masri O, Tullus K, Yeung S, Anikster Y, Klootwijk E, Hubank M, Dillon MJ, Heitzmann D, Arcos-Burgos M, Knepper MA, Dobbie A, Gahl WA, Warth R, Sheridan E, Kleta R (2009) Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med 360:1960–1970PubMedPubMedCentralCrossRef Bockenhauer D, Feather S, Stanescu HC, Bandulik S, Zdebik AA, Reichold M, Tobin J, Lieberer E, Sterner C, Landoure G, Arora R, Sirimanna T, Thompson D, Cross JH, van't Hoff W, Al Masri O, Tullus K, Yeung S, Anikster Y, Klootwijk E, Hubank M, Dillon MJ, Heitzmann D, Arcos-Burgos M, Knepper MA, Dobbie A, Gahl WA, Warth R, Sheridan E, Kleta R (2009) Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med 360:1960–1970PubMedPubMedCentralCrossRef
74.
go back to reference Scholl UI, Choi M, Liu T, Ramaekers VT, Hausler MG, Grimmer J, Tobe SW, Farhi A, Nelson-Williams C, Lifton RP (2009) Seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by mutations in KCNJ10. Proc Natl Acad Sci U S A 106:5842–5847PubMedPubMedCentralCrossRef Scholl UI, Choi M, Liu T, Ramaekers VT, Hausler MG, Grimmer J, Tobe SW, Farhi A, Nelson-Williams C, Lifton RP (2009) Seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by mutations in KCNJ10. Proc Natl Acad Sci U S A 106:5842–5847PubMedPubMedCentralCrossRef
75.
go back to reference Zhang C, Wang L, Su XT, Lin DH, Wang WH (2015) KCNJ10 (Kir4.1) is expressed in the basolateral membrane of the cortical thick ascending limb. Am J Physiol Ren Physiol 308:F1288–F1296CrossRef Zhang C, Wang L, Su XT, Lin DH, Wang WH (2015) KCNJ10 (Kir4.1) is expressed in the basolateral membrane of the cortical thick ascending limb. Am J Physiol Ren Physiol 308:F1288–F1296CrossRef
76.
go back to reference de Baaij JH, Dorresteijn EM, Hennekam EA, Kamsteeg EJ, Meijer R, Dahan K, Muller M, van den Dorpel MA, Bindels RJ, Hoenderop JG, Devuyst O, Knoers NV (2015) Recurrent FXYD2 p.Gly41Arg mutation in patients with isolated dominant hypomagnesaemia. Nephrol Dial Transplant 30:952–957PubMedCrossRef de Baaij JH, Dorresteijn EM, Hennekam EA, Kamsteeg EJ, Meijer R, Dahan K, Muller M, van den Dorpel MA, Bindels RJ, Hoenderop JG, Devuyst O, Knoers NV (2015) Recurrent FXYD2 p.Gly41Arg mutation in patients with isolated dominant hypomagnesaemia. Nephrol Dial Transplant 30:952–957PubMedCrossRef
77.
go back to reference Schlingmann KP, Bandulik S, Mammen C, Tarailo-Graovac M, Holm R, Baumann M, Konig J, Lee JJY, Drogemoller B, Imminger K, Beck BB, Altmuller J, Thiele H, Waldegger S, Van't Hoff W, Kleta R, Warth R, van Karnebeek CDM, Vilsen B, Bockenhauer D, Konrad M (2018) Germline de novo mutations in ATP1A1 cause renal hypomagnesemia, refractory seizures, and intellectual disability. Am J Hum Genet 103:808–816PubMedPubMedCentralCrossRef Schlingmann KP, Bandulik S, Mammen C, Tarailo-Graovac M, Holm R, Baumann M, Konig J, Lee JJY, Drogemoller B, Imminger K, Beck BB, Altmuller J, Thiele H, Waldegger S, Van't Hoff W, Kleta R, Warth R, van Karnebeek CDM, Vilsen B, Bockenhauer D, Konrad M (2018) Germline de novo mutations in ATP1A1 cause renal hypomagnesemia, refractory seizures, and intellectual disability. Am J Hum Genet 103:808–816PubMedPubMedCentralCrossRef
78.
go back to reference Konrad M, Hou J, Weber S, Dotsch J, Kari JA, Seeman T, Kuwertz-Broking E, Peco-Antic A, Tasic V, Dittrich K, Alshaya HO, von Vigier RO, Gallati S, Goodenough DA, Schaller A (2008) CLDN16 genotype predicts renal decline in familial hypomagnesemia with hypercalciuria and nephrocalcinosis. J Am Soc Nephrol 19:171–181PubMedPubMedCentralCrossRef Konrad M, Hou J, Weber S, Dotsch J, Kari JA, Seeman T, Kuwertz-Broking E, Peco-Antic A, Tasic V, Dittrich K, Alshaya HO, von Vigier RO, Gallati S, Goodenough DA, Schaller A (2008) CLDN16 genotype predicts renal decline in familial hypomagnesemia with hypercalciuria and nephrocalcinosis. J Am Soc Nephrol 19:171–181PubMedPubMedCentralCrossRef
79.
go back to reference Godron A, Harambat J, Boccio V, Mensire A, May A, Rigothier C, Couzi L, Barrou B, Godin M, Chauveau D, Faguer S, Vallet M, Cochat P, Eckart P, Guest G, Guigonis V, Houillier P, Blanchard A, Jeunemaitre X, Vargas-Poussou R (2012) Familial hypomagnesemia with hypercalciuria and nephrocalcinosis: phenotype–genotype correlation and outcome in 32 patients with CLDN16 or CLDN19 mutations. Clin J Am Soc Nephrol 7:801–809PubMedPubMedCentralCrossRef Godron A, Harambat J, Boccio V, Mensire A, May A, Rigothier C, Couzi L, Barrou B, Godin M, Chauveau D, Faguer S, Vallet M, Cochat P, Eckart P, Guest G, Guigonis V, Houillier P, Blanchard A, Jeunemaitre X, Vargas-Poussou R (2012) Familial hypomagnesemia with hypercalciuria and nephrocalcinosis: phenotype–genotype correlation and outcome in 32 patients with CLDN16 or CLDN19 mutations. Clin J Am Soc Nephrol 7:801–809PubMedPubMedCentralCrossRef
80.
go back to reference Olinger E, Hofmann P, Kidd K, Dufour I, Belge H, Schaeffer C, Kipp A, Bonny O, Deltas C, Demoulin N, Fehr T, Fuster DG, Gale DP, Goffin E, Hodanova K, Huynh-Do U, Kistler A, Morelle J, Papagregoriou G, Pirson Y, Sandford R, Sayer JA, Torra R, Venzin C, Venzin R, Vogt B, Zivna M, Greka A, Dahan K, Rampoldi L, Kmoch S, Bleyer AJ Sr, Devuyst O (2020) Clinical and genetic spectra of autosomal dominant tubulointerstitial kidney disease due to mutations in UMOD and MUC1. Kidney Int 98:717–731PubMedCrossRef Olinger E, Hofmann P, Kidd K, Dufour I, Belge H, Schaeffer C, Kipp A, Bonny O, Deltas C, Demoulin N, Fehr T, Fuster DG, Gale DP, Goffin E, Hodanova K, Huynh-Do U, Kistler A, Morelle J, Papagregoriou G, Pirson Y, Sandford R, Sayer JA, Torra R, Venzin C, Venzin R, Vogt B, Zivna M, Greka A, Dahan K, Rampoldi L, Kmoch S, Bleyer AJ Sr, Devuyst O (2020) Clinical and genetic spectra of autosomal dominant tubulointerstitial kidney disease due to mutations in UMOD and MUC1. Kidney Int 98:717–731PubMedCrossRef
81.
go back to reference Pearce SH, Williamson C, Kifor O, Bai M, Coulthard MG, Davies M, Lewis-Barned N, McCredie D, Powell H, Kendall-Taylor P, Brown EM, Thakker RV (1996) A familial syndrome of hypocalcemia with hypercalciuria due to mutations in the calcium-sensing receptor. N Engl J Med 335:1115–1122PubMedCrossRef Pearce SH, Williamson C, Kifor O, Bai M, Coulthard MG, Davies M, Lewis-Barned N, McCredie D, Powell H, Kendall-Taylor P, Brown EM, Thakker RV (1996) A familial syndrome of hypocalcemia with hypercalciuria due to mutations in the calcium-sensing receptor. N Engl J Med 335:1115–1122PubMedCrossRef
Metadata
Title
Pathophysiological aspects of the thick ascending limb and novel genetic defects: HELIX syndrome and transient antenatal Bartter syndrome
Author
Rosa Vargas-Poussou
Publication date
01-02-2022
Publisher
Springer Berlin Heidelberg
Published in
Pediatric Nephrology / Issue 2/2022
Print ISSN: 0931-041X
Electronic ISSN: 1432-198X
DOI
https://doi.org/10.1007/s00467-021-05019-6

Other articles of this Issue 2/2022

Pediatric Nephrology 2/2022 Go to the issue