Regulation of magnesium balance: lessons learned from human genetic disease

Jeroen H F de Baaij, Joost G J Hoenderop, René J M Bindels, Jeroen H F de Baaij, Joost G J Hoenderop, René J M Bindels

Abstract

Magnesium (Mg(2+)) is the fourth most abundant cation in the body. Thus, magnesium homeostasis needs to be tightly regulated, and this is facilitated by intestinal absorption and renal excretion. Magnesium absorption is dependent on two concomitant pathways found in both in the intestine and the kidneys: passive paracellular transport via claudins facilitates bulk magnesium absorption, whereas active transcellular pathways mediate the fine-tuning of magnesium absorption. The identification of genes responsible for diseases associated with hypomagnesaemia resulted in the discovery of several magnesiotropic proteins. Claudins 16 and 19 form the tight junction pore necessary for mass magnesium transport. However, most of the causes of genetic hypomagnesaemia can be tracked down to transcellular magnesium transport in the distal convoluted tubule. Within the distal convoluted tubule, magnesium reabsorption is a tightly regulated process that determines the final urine magnesium concentration. Therefore, insufficient magnesium transport in the distal convoluted tubule owing to mutated magnesiotropic proteins inevitably leads to magnesium loss, which cannot be compensated for in downstream tubule segments. Better understanding of the molecular mechanism regulating magnesium reabsorption will give new opportunities for better therapies, perhaps including therapies for patients with chronic renal failure.

Keywords: TRPM6; human genetic disease; hypomagnesaemia; magnesium homeostasis.

Figures

Fig. 1.
Fig. 1.
Magnesium homeostasis. Panels represent the daily amount of Mg2+ intake and excretion. A daily net intake of ∼100 mg in the intestine results in a balanced 100 mg excretion in the kidney. In times of Mg2+ shortage, other tissues such as bone and muscle provide Mg2+ to restore blood Mg2+ levels. See also “Magnesium basics” in this supplement [5]. The conversion factor from milligrams to millimole is 0.04113.
Fig. 2.
Fig. 2.
A schematic overview of magnesium absorption pathways in the intestine, showing proteins associated with Mg2+ transport in enterocytes. In the intestinal epithelia, paracellular Mg2+ transport occurs via unidentified claudins, occuring concurrently with transcellular Mg2+ transport via transient receptor potential channel melastatin member 6 (TRPM6) and TRPM7 to facilitate Mg2+ absorption.
Fig. 3.
Fig. 3.
Magnesium reabsorption along the nephron. The glomerulus filters the blood and facilitates thereby the entrance of Mg2+ into the tubular system that subsequently mediates the reabsorption of 90–95% of Mg2+. Approximately 10–25% of Mg2+ is reabsorbed in the proximal tubule (PT). Bulk transport (50–70%) of Mg2+ is achieved along the thick ascending limb (TAL) of the loop of Henle. The final Mg2+ concentration in urine is determined in the distal convoluted tubule (DCT) where only 10% of Mg2+ is reabsorbed [28]. CNT, connecting tubule; CD, collecting duct.
Fig. 4.
Fig. 4.
Schematic overview of Mg2+ transport pathways in the thick ascending limb of the loop of Henle. The majority of Mg2+ is transported in this part of the nephron. Mg2+ absorption takes place in a paracellular fashion via claudins-16 and -19 of the tight junction complex. The driving force behind Mg2+ transport in the thick ascending limb is the transepithelial voltage gradient.
Fig. 5.
Fig. 5.
Ion transport pathways in the distal convoluted tubule. The final possibility of Mg2+ reabsorption is the tightly regulated transcellular transport in the distal convoluted tubule. In this schematic overview, all the proteins whose mutations cause hypomagnesaemia are shown. Mg2+ enters the cell via the TRPM6 Mg2+ channel that is regulated by EGF. The Kv1.1 K+ channel maintains transmembrane voltage that is the driving force for Mg2+ transport. The key molecule at the basolateral membrane is the Na+/K+-ATPase, whose expression is regulated by transcription factor HNF1B (not shown). The Na+/K+-ATPase activity is stimulated by its γ–subunit. Kir4.1 is responsible for recycling of K+ at the basolateral site of the cell. The basolateral Mg2+ transporter remains to be identified. Gitelman's-associated proteins NCC and ClC-Kb are responsible for Na+ and Cl− transport in the distal convoluted tubule.

References

    1. Musso CG. Magnesium metabolism in health and disease. Int Urol Nephrol. 2009;41:357–362.
    1. Quamme GA. Recent developments in intestinal magnesium absorption. Curr Opin Gastroenterol. 2008;24:230–235.
    1. Pham PC, Pham PM, Pham SV, et al. Hypomagnesemia in patients with type 2 diabetes. Clin J Am Soc Nephrol. 2007;2:366–373.
    1. Dimke H, Hoenderop JG, Bindels RJ. Hereditary tubular transport disorders: implications for renal handling of Ca2+ and Mg2+ . Clin Sci (Lond) 2010;118:1–18.
    1. Jahnen-Dechent W, Ketteler M. Magnesium basics. Clin Kidney J. 2012;5(Suppl 1):i3–i14.
    1. Geiger H, Wanner C. Magnesium in disease. Clin Kidney J. 2012;5(Suppl 1):i25–i38.
    1. Knoers NV. Inherited forms of renal hypomagnesemia: an update. Pediatr Nephrol. 2009;24:697–705.
    1. Huang CL, Kuo E. Mechanism of hypokalemia in magnesium deficiency. J Am Soc Nephrol. 2007;18:2649–2652.
    1. Kesteloot H, Joossens JV. The relationship between dietary intake and urinary excretion of sodium, potassium, calcium and magnesium: Belgian Interuniversity Research on Nutrition and Health. J Hum Hypertens. 1990;4:527–533.
    1. Kopple JD, Coburn JW. Metabolic studies of low protein diets in uremia. II. Calcium, phosphorus and magnesium. Medicine (Baltimore) 1973;52:597–607.
    1. Spencer H, Lesniak M, Gatza CA, et al. Magnesium absorption and metabolism in patients with chronic renal failure and in patients with normal renal function. Gastroenterology. 1980;79:26–34.
    1. Alfrey AC, Miller NL, Trow R. Effect of age and magnesium depletion on bone magnesium pools in rats. J Clin Invest. 1974;54:1074–1081.
    1. Graham L, Caesar J, Burgen A. Gastrointestinal absorption and excretion of Mg28 in man. Metabolism. 1960;9:646–659.
    1. Kayne LH, Lee DB. Intestinal magnesium absorption. Miner Electrolyte Metab. 1993;19:210–217.
    1. Aliaga IL, Miller DL, Wilson HD, et al. Effects of resection on absorption and secretion of divalent cations by small intestine of rat. Am J Clin Nutr. 1990;52:867–871.
    1. Amasheh S, Fromm M, Gunzel D. Claudins of intestine and nephron - a correlation of molecular tight junction structure and barrier function. Acta Physiol (Oxf) 2011;201:133–140.
    1. Hou J, Renigunta A, Gomes AS, et al. Claudin-16 and claudin-19 interaction is required for their assembly into tight junctions and for renal reabsorption of magnesium. Proc Natl Acad Sci USA. 2009;106:15350–15355.
    1. Groenestege WM, Thebault S, van der WJ, et al. Impaired basolateral sorting of pro-EGF causes isolated recessive renal hypomagnesemia. J Clin Invest. 2006;117:2260–2267.
    1. Romani A. Regulation of magnesium homeostasis and transport in mammalian cells. Arch Biochem Biophys. 2007;458:90–102.
    1. Schmulen AC, Lerman M, Pak CY, et al. Effect of 1,25-(OH)2D3 on jejunal absorption of magnesium in patients with chronic renal disease. Am J Physiol. 1980;238:G349–G352.
    1. Brannan PG, Vergne-Marini P, Pak CY, et al. Magnesium absorption in the human small intestine. Results in normal subjects, patients with chronic renal disease, and patients with absorptive hypercalciuria. J Clin Invest. 1976;57:1412–1418.
    1. Mawer EB, Taylor CM, Backhouse J, et al. Failure of formation of 1,25-dihydroxycholecalciferol in chronic renal insufficiency. Lancet. 1973;1:626–628.
    1. Fujita H, Sugimoto K, Inatomi S, et al. Tight junction proteins claudin-2 and -12 are critical for vitamin D-dependent Ca2+ absorption between enterocytes. Mol Biol Cell. 2008;19:1912–1921.
    1. McCance RA, Widdowson EM, Lehmann H. The effect of protein intake on the absorption of calcium and magnesium. Biochem J. 1942;36:686–691.
    1. Verbeek MJ, Van den Berg GJ, Lemmens AG, et al. High protein intake raises apparent but not true magnesium absorption in rats. J Nutr. 1993;123:1880–1887.
    1. Potter JD, Robertson SP, Johnson JD. Magnesium and the regulation of muscle contraction. Fed Proc. 1981;40:2653–2656.
    1. Rude RK, Gruber HE. Magnesium deficiency and osteoporosis: animal and human observations. J Nutr Biochem. 2004;15:710–716.
    1. Le Grimellec C. Micropuncture study along the proximal convoluted tubule. Electrolyte reabsorption in first convolutions. Pflugers Arch. 1975;354:133–150.
    1. Hou J, Renigunta A, Konrad M, et al. Claudin-16 and claudin-19 interact and form a cation-selective tight junction complex. J Clin Invest. 2008;118:619–628.
    1. Hou J, Shan Q, Wang T, et al. Transgenic RNAi depletion of claudin-16 and the renal handling of magnesium. J Biol Chem. 2007;282:17114–17122.
    1. Greenberg A. Diuretic complications. Am J Med Sci. 2000;319:10–24.
    1. Cohen N, Alon I, Almoznino-Sarafian D, et al. Metabolic and clinical effects of oral magnesium supplementation in furosemide-treated patients with severe congestive heart failure. Clin Cardiol. 2000;23:433–436.
    1. Glaudemans B, Knoers NV, Hoenderop JG, et al. New molecular players facilitating Mg2+ reabsorption in the distal convoluted tubule. Kidney Int. 2010;77:17–22.
    1. Voets T, Nilius B, Hoefs S, et al. TRPM6 forms the Mg2+ influx channel involved in intestinal and renal Mg2+ absorption. J Biol Chem. 2004;279:19–25.
    1. Nijenhuis T, Vallon V, van der Kemp AW, et al. Enhanced passive Ca2+ reabsorption and reduced Mg2+ channel abundance explains thiazide-induced hypocalciuria and hypomagnesemia. J Clin Invest. 2005;115:1651–1658.
    1. Ikari A, Okude C, Sawada H, et al. TRPM6 expression and cell proliferation are up-regulated by phosphorylation of ERK1/2 in renal epithelial cells. Biochem Biophys Res Commun. 2008;369:1129–1133.
    1. McNair P, Christiansen C, Transbol I. Effect of menopause and estrogen substitutional therapy on magnesium metabolism. Miner Electrolyte Metab. 1984;10:84–87.
    1. Simon DB, Lu Y, Choate KA, et al. Paracellin-1, a renal tight junction protein required for paracellular Mg2+ resorption. Science. 1999;285:103–106.
    1. Konrad M, Schaller A, Seelow D, et al. 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. 2006;79:949–957.
    1. Hou J, Goodenough DA. Claudin-16 and claudin-19 function in the thick ascending limb. Curr Opin Nephrol Hypertens. 2010;19:483–488.
    1. Jeck N, Schlingmann KP, Reinalter SC, et al. Salt handling in the distal nephron: lessons learned from inherited human disorders. Am J Physiol Regul Integr Comp Physiol. 2005;288:R782–R795.
    1. Hebert SC. Bartter syndrome. Curr Opin Nephrol Hypertens. 2003;12:527–532.
    1. Ellison DH. Divalent cation transport by the distal nephron: insights from Bartter's and Gitelman's syndromes. Am J Physiol Renal Physiol. 2000;279:F616–F625.
    1. Walder RY, Landau D, Meyer P, et al. Mutation of TRPM6 causes familial hypomagnesemia with secondary hypocalcemia. Nat Genet. 2002;31:171–174.
    1. Schlingmann KP, Weber S, Peters M, et al. Hypomagnesemia with secondary hypocalcemia is caused by mutations in TRPM6, a new member of the TRPM gene family. Nat Genet. 2002;31:166–170.
    1. Chubanov V, Waldegger S, Schnitzler M, et al. Disruption of TRPM6/TRPM7 complex formation by a mutation in the TRPM6 gene causes hypomagnesemia with secondary hypocalcemia. Proc Natl Acad Sci USA. 2004;101:2894–2899.
    1. Schmitz C, Dorovkov MV, Zhao X, et al. The channel kinases TRPM6 and TRPM7 are functionally nonredundant. J Biol Chem. 2005;280:37763–37771.
    1. Li M, Jiang J, Yue L. Functional characterization of homo- and heteromeric channel kinases TRPM6 and TRPM7. J Gen Physiol. 2006;127:525–537.
    1. Cao G, Thebault S, van der WJ, et al. RACK1 inhibits TRPM6 activity via phosphorylation of the fused alpha-kinase domain. Curr Biol. 2008;18:168–176.
    1. Woudenberg-Vrenken TE, Sukinta A, van der Kemp AW, et al. Transient receptor potential melastatin 6 knockout mice are lethal whereas heterozygous deletion results in mild hypomagnesemia. Nephron Physiol. 2011;117:11–19.
    1. Li M, Du J, Jiang J, et al. Molecular determinants of Mg2+ and Ca2+ permeability and pH sensitivity in TRPM6 and TRPM7. J Biol Chem. 2007;282:25817–25830.
    1. Chubanov V, Schlingmann KP, Waring J, et al. Hypomagnesemia with secondary hypocalcemia due to a missense mutation in the putative pore-forming region of TRPM6. J Biol Chem. 2007;282:7656–7667.
    1. Schrag D, Chung KY, Flombaum C, et al. Cetuximab therapy and symptomatic hypomagnesemia. J Natl Cancer Inst. 2005;97:1221–1224.
    1. Groenestege WM, Hoenderop JG, van den HL, et al. The epithelial Mg2+ channel transient receptor potential melastatin 6 is regulated by dietary Mg2+ content and estrogens. J Am Soc Nephrol. 2007;17:1035–1043.
    1. Tejpar S, Piessevaux H, Claes K, et al. Magnesium wasting associated with epidermal-growth-factor receptor-targeting antibodies in colorectal cancer: a prospective study. Lancet Oncol. 2007;8:387–394.
    1. Thebault S, Alexander RT, Tiel Groenestege WM, et al. EGF increases TRPM6 activity and surface expression. J Am Soc Nephrol. 2009;20:78–85.
    1. Ikari A, Sanada A, Okude C, et al. Up-regulation of TRPM6 transcriptional activity by AP-1 in renal epithelial cells. J Cell Physiol. 2010;222:481–487.
    1. Dimke H, van der WJ, Alexander TR, et al. Effects of the EGFR inhibitor erlotinib on magnesium handling. J Am Soc Nephrol. 2010;21:1309–1316.
    1. Gamba G, Saltzberg SN, Lombardi M, et al. Primary structure and functional expression of a cDNA encoding the thiazide-sensitive, electroneutral sodium-chloride cotransporter. Proc Natl Acad Sci USA. 1993;90:2749–2753.
    1. Simon DB, Nelson-Williams C, Bia MJ, et al. Gitelman's variant of Bartter's syndrome, inherited hypokalaemic alkalosis, is caused by mutations in the thiazide-sensitive Na-Cl cotransporter. Nat Genet. 1996;12:24–30.
    1. Glaudemans B, van der WJ, Scola RH, et al. A missense mutation in the Kv1.1 voltage-gated potassium channel-encoding gene KCNA1 is linked to human autosomal dominant hypomagnesemia. J Clin Invest. 2009;119:936–942.
    1. Gomez-Hernandez JM, Lorra C, Pardo LA, et al. Molecular basis for different pore properties of potassium channels from the rat brain Kv1 gene family. Pflugers Arch. 1997;434:661–668.
    1. van der Wijst J, Glaudemans B, Venselaar H, et al. Functional analysis of the Kv1.1 N255D mutation associated with autosomal dominant hypomagnesemia. J Biol Chem. 2010;285:171–178.
    1. Smart SL, Lopantsev V, Zhang CL, et al. Deletion of the K(V)1.1 potassium channel causes epilepsy in mice. Neuron. 1998;20:809–819.
    1. Meij IC, Koenderink JB, van BH, et al. Dominant isolated renal magnesium loss is caused by misrouting of the Na+/ K+-ATPase gamma-subunit. Nat Genet. 2000;26:265–266.
    1. Ferre S, Veenstra GJ, Bouwmeester R, et al. HNF-1B specifically regulates the transcription of the gamma a-subunit of the Na+/K+-ATPase. Biochem Biophys Res Commun. 2011;404:284–290.
    1. Katz AI, Doucet A, Morel F. Na+/K+-ATPase activity along the rabbit, rat, and mouse nephron. Am J Physiol. 1979;237:F114–F120.
    1. Cairo ER, Friedrich T, Swarts HG, et al. Impaired routing of wild type FXYD2 after oligomerisation with FXYD2-G41R might explain the dominant nature of renal hypomagnesemia. Biochim Biophys Acta. 2008;1778:398–404.
    1. Sha Q, Pearson W, Burcea LC, et al. Human FXYD2 G41R mutation responsible for renal hypomagnesemia behaves as an inward-rectifying cation channel. Am J Physiol Renal Physiol. 2008;295:F91–F99.
    1. Adalat S, Woolf AS, Johnstone KA, et al. HNF1B mutations associate with hypomagnesemia and renal magnesium wasting. J Am Soc Nephrol. 2009;20:1123–1131.
    1. Ferraro TN, Golden GT, Smith GG, et al. Fine mapping of a seizure susceptibility locus on mouse Chromosome 1: nomination of Kcnj10 as a causative gene. Mamm Genome. 2004;15:239–251.
    1. Bockenhauer D, Feather S, Stanescu HC, et al. Epilepsy, ataxia, sensorineural deafness, tubulopathy, and KCNJ10 mutations. N Engl J Med. 2009;360:1960–1970.
    1. Scholl UI, Choi M, Liu T, et al. Seizures, sensorineural deafness, ataxia, mental retardation, and electrolyte imbalance (SeSAME syndrome) caused by mutations in KCNJ10 . Proc Natl Acad Sci USA. 2009;106:5842–5847.
    1. Ito M, Inanobe A, Horio Y, et al. Immunolocalization of an inwardly rectifying K+ channel, K(AB)-2 (Kir4.1), in the basolateral membrane of renal distal tubular epithelia. FEBS Lett. 1996;388:11–15.
    1. Cha SK, Huang C, Ding Y, et al. Calcium-sensing receptor decreases cell surface expression of the inwardly rectifying K+ channel Kir4.1. J Biol Chem. 2011;286:1828–1835.
    1. Neusch C, Rozengurt N, Jacobs RE, et al. Kir4.1 potassium channel subunit is crucial for oligodendrocyte development and in vivo myelination. J Neurosci. 2001;21:5429–5438.
    1. Marcus DC, Wu T, Wangemann P, et al. KCNJ10 (Kir4.1) potassium channel knockout abolishes endocochlear potential. Am J Physiol Cell Physiol. 2002;282:C403–C407.
    1. Sala-Rabanal M, Kucheryavykh LY, Skatchkov SN, et al. Molecular mechanisms of EAST/SeSAME syndrome mutations in Kir4.1 (KCNJ10) J Biol Chem. 2010;285:36040–36048.
    1. Ferre S, Hoenderop JG, Bindels RJ. Insight into renal Mg2+ transporters. Curr Opin Nephrol Hypertens. 2011;20:169–176.

Source: PubMed

3
Abonnere