Reviews

؉ ؉ Epithelial Ca2 and Mg2 Channels in Health and Disease

Joost G.J. Hoenderop and Rene´J.M. Bindels Department of Physiology, Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen Medical Center, the Netherlands

A near constancy of the extracellular Ca2؉ and Mg2؉ concentration is required for numerous physiologic functions at the organ, tissue, and cellular levels. This suggests that minor changes in the extracellular concentration of these divalents must be detected to allow the appropriate correction by the homeostatic systems. The maintenance of the Ca2؉ and Mg2؉ balance is controlled by the concerted action of intestinal absorption, renal excretion, and exchange with bone. After years of research, rapid progress was made recently in identification and characterization of the Ca2؉ and Mg2؉ transport proteins that contribute to the delicate balance of divalent cations. Expression-cloning approaches in combination with knockout mice models and genetic studies in families with a disturbed Mg2؉ balance revealed novel Ca2؉ and Mg2؉ gatekeeper proteins that belong to the super family of the transient receptor potential (TRP) channels. These epithelial Ca2؉ (TRPV5 and TRPV6) and Mg2؉ channels (TRPM6 and TRPM7) form prime targets for hormonal control of the active Ca2؉ and Mg2؉ flux from the urine space or intestinal lumen to the blood compartment. This review describes the characteristics of epithelial Ca2؉ and Mg2؉ transport in general and highlights in particular the distinctive features and the physiologic relevance of these new epithelial .Ca2؉ and Mg2؉ channels in (patho)physiologic situations J Am Soc Nephrol 16: 15–26, 2005. doi: 10.1681/ASN.2004070523

ϩ ϩ ؉ a2 and Mg2 are of great physiologic importance by Ca2 (Re)absorption ϩ their intervention in many enzymatic systems and The major part of the Ca2 reabsorption takes place along the C their function in neural excitability, muscle contrac- proximal tubule (PT) and thick ascending limb of Henle’s loop tion, blood coagulation, bone formation, hormone secretion, (TAL) through a paracellular and, therefore, passive pathway ϩ and cell adhesion. The human body is equipped with an effi- (5). Fine-tuning of the Ca2 excretion occurs in the distal part of cient negative feedback system that counteracts variations of the nephron, where approximately15% of the filtered load of ϩ ϩ 2ϩ the Ca2 and Mg2 balance. This system encompasses parathy- Ca is reabsorbed. This section consists of the distal convo- roid glands, bone, intestine, and kidneys. These divalents are luted tubule (DCT), the connecting tubule (CNT), and the initial maintained within a narrow range by the small intestine and portion of the cortical collecting duct (CCD; Figure 1). In these 2ϩ kidney, which both increase their fractional (re)absorption un- latter nephron segments (DCT, CNT, CCD), Ca reabsorption der conditions of deprivation (1,2). If depletion continues, then is active and occurs against the existing electrochemical gradi- ent. Together with the fact that here the tight junctions are the bone store assists to maintain appropriate serum concen- ϩ ϩ impermeable for Ca2 ions, this substantiates that Ca2 is trations by exchanging part of its content with the extracellular 2ϩ reabsorbed through an active transcellular pathway. Active fluid. The Ca -sensing receptor (CaSR) represents the molec- ϩ Ca2 reabsorption is generally envisaged as a multistep process ular mechanism by which parathyroid cells detect changes in 2ϩ 2ϩ 2ϩ that consists of passive entry of Ca across the luminal or the ionized Ca and Mg concentration and modulate para- ϩ apical membrane, cytosolic diffusion of Ca2 bound to vitamin thyroid hormone (PTH) secretion (3,4). In addition to the effects D3-sensitive calcium-binding proteins (calbindin-D28K and/or of these divalents on PTH secretion, this hormone in turn 2ϩ ϩ ϩ calbindin-D9K), and active extrusion of Ca across the oppo- 2 2 ϩ ϩ regulates directly the Ca and Mg balance by modulating site basolateral membrane by a Na -Ca2 exchanger (NCX1) ϩ bone resorption, renal reabsorption, and indirectly intestinal and/or Ca2 -ATPase (PMCA1b) (6) (Figure 1, top). This active ␣ ϩ absorption by stimulating 1 -hydroxylase activity and conse- transcellular Ca2 transport is under hormonal control of PTH quently 1,25-dihydroxyvitamin D (1,25-(OH) D ) synthesis to 3 2 3 (7,8), 1,25-(OH)2D3 (7,9–12,103), and calcitonin (13) but also 2ϩ 2ϩ ϩ maintain serum Ca and Mg levels within a narrow phys- estrogen (14,15), androgen (16), and dietary Ca2 (10) are pri- iologic range. mary regulators.

؉ Mg2 (Re)absorption ϩ Regulation of the total body Mg2 balance principally resides

Published online ahead of print. Publication date available at www.jasn.org. within the kidney that tightly matches the intestinal absorption ϩ of Mg2 . In the kidney, approximately 80% of the total plasma Address correspondence to: Dr. Rene´J.M. Bindels, 160 Cell Physiology, Radboud 2ϩ University Nijmegen Medical Center, P.O. Box 9101, 6500 HB Nijmegen, the Neth- Mg is ultrafiltrated across the glomerular membrane and erlands. Phone: ϩ31-24-3614211; Fax: ϩ31-24-3616413; E-mail: [email protected] subsequently reabsorbed in consecutive segments of the

Copyright © 2005 by the American Society of Nephrology ISSN: 1046-6673/1601-0015 16 Journal of the American Society of Nephrology J Am Soc Nephrol 16: 15–26, 2005

ϩ ϩ ϩ ϩ Figure 1. Transcellular Ca2 and Mg2 transport. Active Ca2 and Mg2 transport is carried out as a three-step process in the distal ϩ part of the nephron. (Top) After entry of Ca2 in distal convoluted tubule (DCT2) and connecting tubule (CNT) through the ϩ ϩ epithelial Ca2 channels TRPV5 and TRPV6, Ca2 bound to calbindin diffuses to the basolateral membrane. At the basolateral ϩ ϩ ϩ ϩ membrane, Ca2 is extruded via an ATP-dependent Ca2 -ATPase (PMCA1b) and an Na -Ca2 –exchanger (NCX1). (Bottom) ϩ ϩ ϩ Apical Mg2 entry in DCT via TRPM6 (and TRPM7). As extrusion mechanisms are postulated a basolateral Na /Mg2 exchanger ϩ ϩ ϩ ϩ and/or ATP-dependent Mg2 pump. The Na ,K -ATPase complex including the ␥-subunit controls this transepithelial Mg2 ϩ ϩ transport. In this way, there is net Ca2 and Mg2 reabsorption from the luminal space to the extracellular compartment.

ϩ ؉ nephron (1). Approximately 10 to 20% of Mg2 is reabsorbed Search for Epithelial Ca2 Channels ϩ ϩ by the PT. However, the bulk amount of Mg2 (50 to 70%) is Several genes involved in the process of transepithelial Ca2 ϩ ϩ reabsorbed by the TAL, which likely mediates Mg2 reabsorp- transport have now been identified, but the Ca2 influx mech- tion via paracellular transport. The final urinary excretion of anism remained unknown for a long time. An expression- ϩ ϩ Mg2 is mainly determined by active reabsorption of Mg2 in cloning approach using Xenopus laevis oocytes revealed the ϩ DCT, because virtually no reabsorption takes places beyond molecular identity of the Ca2 influx systems (19,20). The first this segment (Figure 1) (1). Microperfusion studies have shown member, named TRPV5, was cloned from primary cultures of ϩ that Mg2 is reabsorbed in the superficial DCT, but little knowl- rabbit renal distal tubules that are primarily involved in active 2ϩ 2ϩ edge has been gained concerning the cellular mechanisms of transcellular Ca transport and encodes a Ca channel that ϩ ϩ transcellular Mg2 reabsorption (1,17,18). Speculatively, Mg2 belongs to the TRP family (19). Likewise, a homologous mem- can passively enter the DCT cell across the luminal membrane ber of this family, known as TRPV6, was successfully cloned from rat duodenum (20). driven by a favorable plasma membrane voltage (Figure 1, bottom). The molecular identity of the responsible influx pro- tein was unknown, however, and previous studies hypothe- 2ϩ Search for Genes Involved in sized that a Mg -specific is a possible candidate 2؉ ϩ Mg Homeostasis (1). Subsequently, Mg2 will be transported through the cytosol During the past few years, several genes that encode proteins ϩ and extruded at the opposing basolateral membrane by an that are either directly or indirectly involved in renal Mg2 active mechanism given the existing electrochemical gradient. handling have been identified following a positional cloning Again, the identity of responsible transport proteins remains to strategy in families with hereditary hypomagnesemia. The first 2ϩ be defined, and candidate mechanisms are Mg -binding pro- gene involved, PCLN-1 (or CLDN16), encodes the protein para- ϩ 2ϩ 2ϩ teins, Na /Mg exchanger and/or an ATP-dependent Mg cellin-1 (or claudin-16) (21). This protein is specifically ex- pump (Figure 1, bottom). pressed in the TAL and shows sequence and structural homol- ϩ ϩ J Am Soc Nephrol 16: 15–26, 2005 Ca2 and Mg2 Physiology 17 ogy to the claudin family of tight junction proteins. Paracellin-1 in autosomal recessive HSH, previously mapped to chromo- is mutated in patients who have hypomagnesemia, hypercalci- some 9q22. The TRPM6 protein is a new member of the long uria, and nephrocalcinosis (HHN; MIM 248250). In this auto- TRP channel (TRPM) family and is similar to TRPM7 (also ϩ somal recessive disorder, there is profound renal Mg2 and known as TRP-PLIK), a unique bifunctional protein known as a ϩ ϩ Ca2 wasting. The hypercalciuria often leads to nephrocalcino- Mg2 -permeable cation channel properties with protein kinase sis, resulting in progressive renal failure (22,23). Other symp- activity (33–35). TRPM6 and TRPM7 are distinct from all other toms that have been reported in patients with HHN include ion channels in that they are composed of a channel linked to a urinary tract infections, nephrolithiasis, incomplete distal tubu- protein kinase domain recently abbreviated as chanzymes (36). ϩ lar acidosis, and ocular abnormalities (22,24). Immunohisto- These chanzymes are essential for Mg2 homeostasis, which is logic studies have shown that claudin-16 co-localizes with oc- critical for human health and cell viability (37,38). cludin in intercellular junctions of human kidney sections, In summary, a variety of approaches, including a genetic indicating that it is a tight junction protein (21). The second screen in patients with primary hypomagnesemia and expres- ϩ ϩ ϩ gene, FXYD2, encodes the ␥-subunit of the Na ,K -ATPase sion cloning in Ca2 -transporting epithelial cells, revealed the pump, which is predominantly expressed in the kidney, with identification of TRP cation channels as potential gatekeepers in ϩ ϩ the highest expression levels in DCT and medullary TAL (25). the maintenance of the Ca2 and Mg2 balance. The TRP FXYD2 is mutated in patients with autosomal dominant renal superfamily is a newly discovered family of cation-permeable hypomagnesemia associated with hypocalciuria (IDH; MIM ion channels (33). There are at least three previously recognized 154020). Hypomagnesemia in these patients can be severe subfamilies of proteins—TRPC (conical), TRPV (vanilloid), and (Ͻ0.40 mM) and cause convulsions. Remarkably, in some af- TRPM (metastatin)—that are expressed throughout the animal ϩ fected individuals, there are no symptoms of Mg2 deficiency kingdom (http://clapham.tch.harvard.edu/trps/). Recently, except for chondrocalcinosis at adult age. The molecular mech- the polycystins were also included in the TRP superfamily ϩ anism for renal Mg2 loss in this autosomal dominant type of abbreviated as TRPP (polycystin) (39). Each of the proteins primary hypomagnesemia remains to be elucidated. The third seems to be a cation channel composed of six transmembrane- gene involved, SLC12A3, encodes the thiazide-sensitive sodium spanning domains and a conserved pore-forming region (Fig- chloride co-transporter (NCC) in DCT and is mutated in pa- ure 2) (6,33,40). Most members of the TRPC have been charac- ϩ tients with Gitelman syndrome (MIM 263800) (26). This auto- terized as Ca2 -permeable cation channels playing a role in ϩ somal recessive disorder is a frequent hereditary tubular dis- Ca2 signaling (41). The functional characterization of other ϩ order that affects renal Mg2 handling, which is characterized TRP members, including TRPV5 and TRPV6, and TRPM6 and by hypokalemia, hypomagnesemia, and hypocalciuria. Hypo- TRPM7, has recently been started. magnesemia is found in most patients with Gitelman syndrome and is assumed to be secondary to the primary defect in NCC, TRPV5 and TRPV6 but the mechanisms underlying hypomagnesemia are poorly TRPV5 and TRPV6 belong to the TRPV subfamily. These understood. homologues channel proteins are composed of approximately Although these linkage analyses revealed the identification 730 amino acids, whereas the corresponding genes consist of 15 ϩ of genes involved in Mg2 homeostasis, the key molecules that exons juxtaposed on 7q35 (42–44). In human em- ϩ represent the mechanisms for luminal Mg2 influx and baso- bryonic kidney 293 (HEK293) cells heterogeneously expressing ϩ ϩ lateral Mg2 extrusion in the process of transcellular Mg2 TRPV5 or TRPV6, currents can be activated under conditions of ϩ transport are still elusive. Importantly, Walder et al. (27) re- high intracellular buffering of Ca2 . In addition, the current is ported that hypomagnesemia associated with secondary hy- increased by hyperpolarizing voltage steps, which enhances the ϩ pocalciuria (HSH; MIM 602014) is an autosomal recessive dis- driving force for Ca2 (45,46). Outward currents are extremely ease that is genetically linked to chromosome 9p22. This disease small, indicating that these channels are inwardly rectifying ϩ ϩ is primarily due to defective intestinal Mg2 absorption, and (Table 1, Figure 3). TRPV5 and TRPV6 are subject to Ca2 - affected individuals show neurologic symptoms of hypomag- dependent feedback inhibition (46,47). Both channels rapidly nesemic hypocalcemia, including seizures and muscle spasms inactivate during hyperpolarizing voltage steps, and this inac- ϩ ϩ ϩ during infancy (28–30). Because passive Mg2 absorption is not tivation is reduced when Ba2 or Sr2 is used as a charge ϩ ϩ affected, the disease can be treated by high dietary Mg2 intake carrier, confirming the Ca2 dependence (47). Currents also ϩ (31). Renal Mg2 conservation has been reported to be normal diminish during repetitive activation by short hyperpolarizing in most patients. In some cases, however, a renal leak has been pulses (46). TRPV5 and TRPV6 are so far the only known highly ϩ ϩ reported, suggesting impaired renal Mg2 reabsorption. Pa- Ca2 -selective channels in the TRP superfamily. It has been ϩ tients who were studied by Konrad et al. and others (28,32) demonstrated that the molecular determinants of the Ca2 ϩ showed inappropriately high fractional Mg2 excretion rates selectivity and permeation of TRPV5 and TRPV6 reside at a ϩ with respect to their low serum Mg2 levels. When untreated, single aspartate residue (TRPV5D542 [48,49] and TRPV6D541 the disease may be fatal or may lead to severe neurologic [50], respectively) present in the predicted pore-forming region. damage. Hypocalcemia is secondary to parathyroid failure re- Neutralization of these negatively charged residues affects the ϩ ϩ sulting from Mg2 deficiency. Using a positional candidate high Ca2 selectivity of these channels. Therefore, it was sug- ϩ gene-cloning approach, two groups headed by Konrad and gested that Ca2 selectivity in TRPV5 and TRPV6 depends on Sheffield (28,29) independently identified mutations in TRPM6 a ring of four aspartate residues in the channel pore, similar to 18 Journal of the American Society of Nephrology J Am Soc Nephrol 16: 15–26, 2005

Figure 2. Structural organization of TRPV5/6 and TRPM6/7. TRPV5 and TRPV6 contain a cytosolic amino- and carboxyl-terminus containing ankyrin (ANK) repeats (A). TRPM6 and TRPM7 belong to the largest TRP channels consisting of approximately 2000 amino acids, including very large cytosolic amino- and carboxyl-termini including an atypical protein kinase domain (B). The six-transmembrane unit is one of four identical or homologous subunits presumed to surround the central pore (C). The gate and selectivity filter are formed by the four two-transmembrane domains (TM5–pore loop–TM6) facing the center of the channel. Cations are selected for permeation by the extracellular-facing pore loop, held in place by the TM5 and TM6 ␣-helices.

ϩ ϩ the ring of four negative residues (aspartates and/or gluta- bust calciuresis compared with wild-type (TRPV5 / ) litter- ϩ Ϫ Ϫ mates) in the pore of voltage-gated Ca2 channels (48,50). Re- mates. Serum analysis showed that TRPV5 / mice have ϩ cently, the substituted cysteine accessibility method was used normal plasma Ca2 concentrations but significantly elevated to map the pore region of TRPV5 (51) and TRPV6 (50). On the 1,25-(OH)2D3 levels (54). For locating the defective site of the ϩ basis of the permeability of the TRPV6 channel to organic Ca2 reabsorption along the nephron, in vivo micropuncture cations, a pore diameter of 5.4 Å was estimated (50). Mutating studies were performed. Collections of tubular fluid revealed ϩ D541 2 ϩ Ϫ Ϫ TRPV6 , a residue involved in high-affinity Ca binding, unaffected Ca2 reabsorption in TRPV5 / mice up to the last ϩ altered the apparent pore diameter, indicating that this residue surface loop of the late proximal tubule. In contrast, Ca2 indeed lines the narrowest part of the pore (50). delivery to puncturing sites within DCT and CNT were signif- The renal expression profile of TRPV5 has been studied in Ϫ/Ϫ icantly enhanced in TRPV5 mice. It is interesting that poly- great detail (Table 1). In different species, it was demonstrated Ϫ/Ϫ ϩ uria and polydipsia were consistently observed in TRPV5 that TRPV5 co-localizes in the kidney with the other Ca2 mice compared with control littermates. Polyuria facilitates the transport proteins, including calbindin-D and the extrusion ϩ 28K excretion of large quantities of Ca2 by reducing the potential proteins PMCA1b and NCX1 in DCT2 and CNT, with the ϩ risk of Ca2 precipitations. This hypercalciuria-induced poly- highest immunochemical abundance in DCT2, and a gradual uria has been observed in humans and animal models (55,56). decrease along CNT (52,53). A minority of cells along CNT Ϫ/Ϫ 2ϩ Furthermore, TRPV5 - lacked immunopositive staining for TRPV5 and the other Ca - mice produced urine that was signif ϩ/ϩ transporting proteins. These negative cells were identified as icantly more acidic compared with TRPV5 littermates. Acidification of the urine is known to prevent renal stone intercalated cells (52). Taken together, these findings suggest ϩ ϩ 2 that the major sites of transcellular Ca2 transport are DCT2 formation in hypercalciuria, because the formation of Ca and, probably to a lesser extent, CNT. Recently, Hoenderop et precipitates is less likely at an acidic pH (57). A significant Ϫ Ϫ 2ϩ al. (54) generated TRPV5 null (TRPV5 / ) mice by genetic increase in the rate of Ca absorption was observed in Ϫ Ϫ ablation of TRPV5 to investigate the function of TRPV5 in renal TRPV5 / mice compared with wild-type littermates, indicat- ϩ and intestinal Ca2 (re)absorption. It is interesting that meta- ing a compensatory role of the small intestine. Expression stud- Ϫ Ϫ Ϫ Ϫ bolic studies demonstrated that TRPV5 / mice exhibit a ro- ies using quantitative real-time PCR in TRPV5 / mice dem- ϩ ϩ J Am Soc Nephrol 16: 15–26, 2005 Ca2 and Mg2 Physiology 19 references 103,105) 59,61,103) 79,82) 62,63,82,83) (38,52,59,61) (35,37,38,45,46, (63,83) (58,75-78) (28,29,38,52, (9-11,14-16,72, (38,45,46,79,82) (38,54,60,82,83) (28,29,54,60, homeostasis 2 ϩ 2 ϩ TRPM7 Ca Ͼ 2 ϩ arrest of cells, lethality ND Not detected ND Cellular Mg Mg Knockout cells: growth

Figure 3. Current-voltage relationship of TRP channels. Repre-

2 ϩ sentative transmembrane currents in response to a voltage TRPM6

Ca ramp (I–V relation) of TRPV5/6 and TRPM6/7 channels. Ͼ (re)absorption 2 ϩ 2 ϩ with secondary hypocalciuria ND Luminal membraneHomo/heteromultimer Homo/heteromultimer Membrane (66,70,83) Outward rectifier Outward rectifier TRPM6a,b,c Kidney (DCT), intestine Widely ND Mg Mg Patient: hypomagnesemia onstrated increased TRPV6 and calbindin-D9K levels in ϩ duodenum consistent with Ca2 hyperabsorption. Immunohistochemical studies indicated that TRPV6, origi- nally cloned from duodenum, is localized to the brush-border membrane of the small intestine. In enterocytes, TRPV6 is co- androgen expressed with calbindin-D9K and PMCA1b (58,59). It is inter- malabsorption 2 ϩ esting that Hediger and co-workers (60) studied the functional 2 ϩ ϩ role of TRPV6 in Ca2 absorption by inactivation of the mouse

2 ϩ Ϫ Ϫ TRPV6 TRPV6 gene. These TRPV6 null (TRPV6 / ) mice were placed ϩ 3 Mg onaCa2 -deficient diet and subsequently challenged in a D

2 ϩ Ϫ Ϫ 45Ca2 absorption assay. TRPV6 / mice showed a consistent 2ϩ ϾϾϾ absorption decrease in Ca absorption over time. From these initial data, Ϫ/Ϫ 2ϩ 2 ϩ 2 ϩ it was concluded that TRPV6 mice show a significant Ca dietary Ca expression correlates with prostate carcinoma tumor grade S100A10, calmodulin Inward rectifier (Brushborder) membrane 2kb-cDNA in testis Knockout mice: Ca Widely Ca 1,25-(OH) Ca

a malabsorption, suggesting that TRPV6 is indeed the rate-limit- 2ϩ ing step in 1,25-OH2D3–dependent Ca absorption (60). Re- cently, it was found that TRPV6 is expressed in the mouse kidney along the apical domain of the late portion of the DCT (DCT2) through inner medullary collecting duct (61). TRPV6 ϩ co-localizes with TRPV5 and the other Ca2 transport proteins ϩ 2 ϩ in DCT2, suggesting a role in Ca2 reabsorption. In addition, PTH estradiol TRPV5 3 2 ϩ Mg the protein is detected in the intercalated cells and the inner D 2 medullary collecting duct that are not involved in transepithe- hyperabsorption, low 2ϩ

ϾϾϾ reabsorption lial Ca transport, pointing to additional functions of TRPV6. 2 ϩ 2ϩ

2 ϩ 2 ϩ Thus, the precise role of this epithelial Ca channel in kidney bone mineral density Ca dietary Ca intestine Ca Homo/heterotetramer Homo/heterotetramer Kidney (DCT2,CNT), bone, Ca remains to be established, but given the widespread distribu- tion of TRPV6 throughout the nephron segments, functions of ϩ ϩ TRPV6 could involve Ca2 reabsorption, Ca2 signaling, and Ϫ Ϫ others. Detailed characterization of the TRPV6 / mice will address these important questions shortly. It is interesting that quantitative PCR measurements indicated that the mouse pros- tate contains high expression levels of TRPV6 (61). Although TRPV5/6 and TRPM6/7 share several features, including expression profiling, transport function of divalent ions, and structural organization. It is interesting that

a the exact function in this organ remains to be elucidated, pre- Associated proteinsFunction S100A10, 80KH Hormonal regulation 1,25-(OH) Malfunction Knockout mice: hypercalciuria, LocalizationStructure Luminal membrane Expression Splice variants Not detected Selectivity Channel properties Inward rectifier one of the ion(TRPV5 channel and pairs TRPM6). is Elusiveof widely information, malfunction expressed such needs (TRPV6 as to and the be TRPM7), hormonal addressed whereas regulation in the of the homologous TRPM6/7; future. member mouse DCT, shows knockout distal a models convoluted more of tubule; restricted TRPV6, CNT, expression TRPM6, connecting pattern and tubule; in TRPM7; ND, epithelial and not tissues clinical determined. consequences For explanation, see text. Table 1 . Characteristics of TRPV5/6 and TRPM6/7 vious reports have suggested that TRPV6 expression correlates 20 Journal of the American Society of Nephrology J Am Soc Nephrol 16: 15–26, 2005 with prostate carcinoma tumor grade (16,62,63). Together, these which are described in detail (6,70). Recently, the effect of PTH findings indicate that TRPV6 expression is associated with was studied on the renal expression of TRPV5 (71). The para- prostate cancer progression and, therefore, represents a prog- thyroid glands play a key role in maintaining the extracellular ϩ nostic marker and a promising target for new therapeutic strat- Ca2 concentration through their capacity to sense minute ϩ egies to treat advanced prostate cancer (63). TRPV5 and TRPV6 changes in the level of blood Ca2 (4). Early studies using share several functional properties, including the permeation micropuncture and cell preparations demonstrated that PTH ϩ profile for monovalent and divalent cations, anomalous mole stimulates active Ca2 reabsorption in the distal part of the ϩ fraction behavior, and Ca2 -dependent inactivation (47,64). nephron via a dual signaling mechanism involving protein However, detailed comparison of the amino- and carboxyl- kinase A– and protein kinase C–dependent processes termini of the TRPV5 and TRPV6 channels illustrates signifi- (7,8,72,73). Preliminary studies demonstrated that parathyroid- cant differences, which may account for the unique electro- ectomy in rats resulted in decreased serum PTH levels and physiologic properties of these homologous channels (65). The hypocalcemia, which were accompanied by decreased levels of initial inactivation is faster in TRPV6 compared with TRPV5, TRPV5, calbindin-D28K, and NCX1. Supplementation with PTH ϩ ϩ ϩ and the kinetic differences between Ca2 and Ba2 currents are restored serum Ca2 concentrations and abundance of these ϩ more pronounced for TRPV6 than for TRPV5 (66,67). It is Ca2 transporters in kidney. These data suggest that long-term ϩ interesting that structural determinants of these functional dis- treatment with PTH affects renal Ca2 handling through the ϩ similarities are not located in either the amino- or carboxyl- regulation of the expression of the Ca2 transport proteins, terminus but in the TM2-TM3 linker (67). It is intriguing that including TRPV5 (71). Promoter analysis should reveal the TRPV5 has a 100-fold higher affinity for the potent channel molecular mechanism of this PTH-mediated increase in TRPV5 blocker than TRPV6 (65). Physiologic conse- expression. In addition, several regulatory proteins that interact quences of these functional differences remain to be established with TRPV5 and/or TRPV6 have been identified, including and are of interest with respect to the structural organization of calmodulin (74–76), S100A10-annexin 2 (58), and 80K-H (77) these channels (66). Cross-linking studies, co-immunoprecipi- (Table 1). These newly identified associated proteins have fa- tations, and molecular mass determination of TRPV5/6 com- cilitated the elucidation of important molecular pathways mod- plexes using sucrose gradient sedimentation showed that ulating transport activity. Calmodulin and 80K-H both have ϩ TRPV5 and TRPV6 form homo- and heterotetrameric channel been implicated as Ca2 sensors. Disturbance of the EF-hand complexes (Figure 2C). Hetero-oligomerization of TRPV5 and structures in these proteins directly affects TRPV5/6 channel TRPV6 might influence the functional properties of the formed activity. Interaction of TRPV5/6 with the S100A10-annexin 2 ϩ ϩ Ca2 channel complex. As TRPV5 and TRPV6 exhibit different complex is critical for trafficking of these epithelial Ca2 chan- ϩ channel kinetics with respect to Ca2 -dependent inactivation nels toward the plasma membrane. ϩ and Ba2 selectivity and sensitivity for the inhibitor ruthenium red, the influence of the heterotetramer composition on these TRPM6 and TRPM7 channel properties was investigated. Concatemers that con- TRPM6 is a protein of approximately 2000 amino acids en- sisted of four TRPV5 and/or TRPV6 subunits that were config- coded by a large gene that contains 39 exons (28,29,33). TRPM6 ured in a head-to-tail manner were constructed. A different shows approximately 50% sequence homology with TRPM7, ϩ ϩ ratio of TRPV5 and TRPV6 subunits in these concatemers which forms a Ca2 - and Mg2 -permeable cation channel (38). showed that the phenotype resembles the mixed properties of Unlike other members of the TRP family, TRPM6 and TRPM7 TRPV5 and TRPV6. An increased number of TRPV5 subunits in contain long carboxyl-terminal domains with similarity to the such a concatemer displayed more TRPV5-like properties, in- ␣-kinases (Figure 2B) (35). The combination of channel and dicating that the stoichiometry of TRPV5/6 heterotetramers enzyme domains in TRPM6 and TRPM7 is unique among influences the channel properties (66). Consequently, regula- known ion channels and raises intriguing questions concerning tion of the relative expression levels of TRPV5 and TRPV6 may the function of the enzymatic domains and physiologic role of ϩ be a mechanism to fine-tune the Ca2 transport kinetics in these chanzymes. The identification of TRPM6 as the gene TRPV5/6–co-expressing tissues (68). It is interesting that Ni- mutated in HSH represents the first case in which a human emeyer and colleagues (69) identified the third ankyrin (ANK) disorder has been attributed to a channel kinase. However, the repeat as being a stringent requirement for physical assembly precise function of this kinase domain remains to be estab- of TRPV6 subunits. Deletion of this repeat or mutation of lished. To date, TRPM7 regulation has received most of the critical residues within this repeat renders nonfunctional chan- attention. TRPM7 is ubiquitously expressed and implicated in ϩ nels that do not co-immunoprecipitate or form tetramers. It was cellular Mg2 homeostasis, whereas TRPM6 has a more re- proposed that the third ANK repeat initiates a molecular zip- stricted expression pattern predominantly present in absorbing pering process that proceeds past the fifth ANK repeat and epithelia (28,29,38). Although in HSH the defect at the level of creates an intracellular anchor that is necessary for functional the intestine is established, there is also evidence for impaired ϩ subunit assembly (69). renal Mg2 reabsorption (28,32). The renal expression of Previous studies in animal and cell models demonstrated TRPM6, in addition to the renal leak in patients with HSH, ϩ that TRPV5 and TRPV6 channels are tightly regulated at the stresses the potential important role of TRPM6 in renal Mg2 transcriptional level by various hormones, including 1,25- reabsorption. In kidney, TRPM6 is expressed in DCT, known as 2ϩ 2ϩ (OH)2D3, estradiol, androgen, and also dietary Ca intake, the main site of active transcellular Mg reabsorption along ϩ ϩ J Am Soc Nephrol 16: 15–26, 2005 Ca2 and Mg2 Physiology 21

ϩ the nephron (38). In line with the expected function of being the (1). Because the Ca2 concentration is in the millimolar range, ϩ ϩ gatekeeper of Mg2 influx, TRPM6 was predominantly local- the apical Mg2 influx pathway should exhibit a higher affinity ϩ ϩ ized along the apical membrane of these immunopositive tu- for Mg2 than for Ca2 . It is interesting that dose-response ϩ bules. Immunohistochemical studies of TRPM6 and NCC, curves for the Na current block at Ϫ80 mV indicated four 2ϩ 2ϩ which were used as specific markers for DCT, indicated a times higher KD values for Ca compared with Mg (38). complete co-localization of these transport proteins in the kid- These data suggest that the pore of the TRPM6 has a higher ϩ ϩ ϩ ney (38). Until now, specific Mg2 -binding proteins have not affinity for Mg2 than for Ca2 . In this way, TRPM6 comprises ϩ ϩ been identified, but it is interesting to mention that the Ca2 - a unique channel because all known Ca2 -permeable channels, ϩ binding proteins parvalbumin and calbindins also bind Mg2 including members of the TRP superfamily, generally display a ϩ ϩ (78). Importantly, TRPM6 co-localized with parvalbumin in 10 to 1000 times lower affinity for Mg2 than for Ca2 .Itis

DCT1 and with calbindin-D28K in DCT2 (38). In addition to the interesting that HEK293 cells transfected with the TRPM6 mu- DCT segment, Schlingmann et al. (28,38) reported the presence tants identified in HSH patients (TRPM6Ser590X and of TRPM6 mRNA by nephron segment–specific PCR analysis in TRPM6Arg736fsX737) displayed currents with similar amplitude the proximal tubule, which was not confirmed by immunohis- and activation kinetics as nontransfected HEK293 cells, indicat- tochemistry. In small intestine, absorptive epithelial cells ing that these mutant proteins are nonfunctional, in line with ϩ stained positively for TRPM6 detected by in situ hybridization the postulated function of TRPM6 being Mg2 influx step in ϩ and immunohistochemistry (28,38). In these cells, TRPM6 was epithelial Mg2 transport (38). The observation that TRPM7 ϩ localized along the brush-border membrane (38). conducts Mg2 and is required for cell viability suggested that ϩ ϩ To functionally characterize TRPM6, the protein was hetero- the TRPM7-mediated Mg2 influx is essential for cellular Mg2 ϩ geneously expressed in HEK293 cells. TRPM6-transfected homeostasis rather than the extracellular Mg2 homeostasis HEK293 cells perfused with an extracellular solution that con- (37). It is interesting that Schmitz et al. (82) demonstrated that ϩ ϩ ϩ tained 1 mM Mg2 or Ca2 exhibited characteristic outwardly Mg2 supplementation of cells that lack TRPM7 expression rectifying currents upon establishment of the whole-cell con- rescued growth arrest and cell lethality that was caused by figuration as was demonstrated for TRPM7 (Figure 3) TRPM7 inactivation (Table 1). Although TRPM7 is permeable ϩ ϩ ϩ ϩ (37,38,79). It is intriguing that at physiologic membrane poten- for Ca2 , as well as trace divalents such as Zn2 ,Ni2 ,Ba2 , ϩ tials of the DCT cell (Ϫ80 mV), small but significant inward and Co2 , supplementation with these cations was ineffective, ϩ currents were observed in TRPM6-expressing HEK293 cells indicating the specific effect of Mg2 on these cellular pro- with all tested divalent cations as the sole charge carrier. How- cesses. ever, mutations in TRPM6 are linked directly to HSH, empha- Recently, it was postulated that TRPM6 requires assembly sizing that this channel is an essential component of the epi- with TRPM7 to form functional channel complexes in the ϩ thelial Mg2 uptake machinery. It is possible that the TRPM6- plasma membrane and that disruption of multimer formation ϩ mediated Mg2 inward current is more pronounced in native by a mutated TRPM6 variant, TRPM6S141L, results in HSH (83). DCT and intestinal cells as a result of specific co-factors, such as In this study, TRPM6S141L was not directed to the cell surface by ϩ intracellular Mg2 buffers, that are missing in overexpression TRPM7 and failed to interact with the coexpressed TRPM7. systems such as HEK293 cells. Remarkably, in contrast to TRPM7, Gudermann and co-work- The unique permeation rank order determined from the in- ers (83) found that TRPM6 expression in Xenopus oocytes and ward current amplitude at Ϫ80 mV was comparable to TRPM7 HEK293 cells did not entail significant ion currents. In contrast, ϩ ϩ ϩ ϩ (Ba2 Ն Ni2 Ͼ Mg2 Ͼ Ca2 ) (35,38). Experiments using the Voets et al. (38) measured significantly larger currents in ϩ Mg2 -sensitive radiometric fluorescent dye Magfura-2 demon- TRPM6-transfected HEK293 cells compared with control cells. strated a coherent relationship between the applied extracellu- An explanation for this discrepancy might be the existence of ϩ ϩ lar Mg2 concentration and the measured intracellular Mg2 specific TRPM6 splice variants with different functional prop- ϩ level in TRPM6-expressing cells. Intracellular Mg2 was ele- erties. Chubanov et al. (83) demonstrated that 5Ј rapid amplifi- vated further when the plasma membrane was hyperpolarized cation of cDNA ends revealed three short alternative 5Ј exons, to the physiologic level of Ϫ80 mV, consistent with influx called 1A, 1B, and 1C, that were found to be individually through the TRPM6 channel. For evaluating the effect of intra- spliced onto exon 2, suggesting that the TRPM6 gene harbors a ϩ ϩ cellular Mg2 on TRPM6 activity, the Mg2 concentration was promoter with alternative transcription start sites. These cDNA altered directly in a spatially uniform manner using flash pho- have been named accordingly TRPM6a, TRPM6b, and TRPM6c, ϩ tolysis of the photolabile Mg2 chelator DM-nitrophen. Eleva- and additional functional measurements are needed to explain ϩ tion of intracellular Mg2 by a flash of ultraviolet light reduced possible biophysical differences. the TRPM6-induced current, indicating that the channel is reg- A key question concerns the nature of mechanisms underly- ulated by the intracellular concentration of this ion. Likewise, ing the activation and regulation of TRPM6 and TRPM7. In ϩ TRPM7 channel activity is strongly suppressed by Mg2 -ATP particular, what is the function of the atypical protein ␣-kinase concentrations in the millimolar range (37,80). Kozak and Ca- domain located in the carboxyl terminus? ␣-Kinases are a re- ϩ halan (81) demonstrated that internal Mg2 rather than ATP cently discovered family of proteins that have no detectable inhibits channel activity. sequence homology to conventional protein kinases (84). To Micropuncture studies have demonstrated that the luminal characterize the TRPM7 kinase activity in vitro, we performed ϩ concentration of free Mg2 in DCT ranges from 0.2 to 0.7 mM studies in which the catalytic domain was expressed in bacteria 22 Journal of the American Society of Nephrology J Am Soc Nephrol 16: 15–26, 2005

(85). This kinase is able to undergo autophosphorylation and to phosphorylate substrates such as myelin basic protein and histone H3 on serine and threonine residues. The kinase is ϩ ϩ specific for ATP and requires Mg2 or Mn2 for optimal activ- ity. Clapham and co-workers (35) found that kinase activity is

necessary for TRPM7 channel function. Although kinases have Disturbance

long been known to modulate ion channels, TRPM7 is unusual 2 ϩ

in that the channel has its own kinase. Future studies will Mg Homeostasis address the question of whether the kinase, present in TRPM6 Hypermagnesemia and TRPM7, has specific cellular targets that might modulate 2 ϩ ϩ ion channel activity and, therefore, the Mg2 balance. and Mg 2؉

Mutual Disturbance of the Ca and 2 ϩ 2؉ Mg Balance Ca ϩ ϩ A tight coupling of the Ca2 and Mg2 balance is frequently Disturbance 2 ϩ

observed in patients and animal models (86,87). In hypomag- hypercalcemia Ca nesemia with secondary hypocalcemia, Simon et al. (21) proposed Hypocalciuria Hypomagnesemia ϩ that paracellin-1 is involved in controlling both the Ca2 and ϩ Mg2 permeability of the paracellular pathway in TAL. Immuno- localization studies demonstrated that this tight junction protein is ϩ ϩ expressed in TAL. Defective paracellular Ca2 and Mg2 absorp-

tion by inactive paracellin-1 explains the observed hypomag- -ATPase nesemia and hypercalciuria in patients with HHN (Table 2). ϩ 2ϩ -K Mutations in the Ca -sensing receptor (CaSR) are also asso- ϩ ϩ ciated with disturbed Mg2 handling (Table 2). Mutations in this receptor are identified in autosomal dominant hypopara-

thyroidism, which is characterized by hypercalcemia and hy- a pocalciuria (88). Hypomagnesemia is observed in up to half of -subunit Na the patients (89). Mutations in the parathyroid and kidney CaSR ␥ 2ϩ 2ϩ result in a lower set point for plasma Ca and Mg on PTH balance 2ϩ 2ϩ secretion (86). Consequently, renal Ca and Mg reabsorption 2 ϩ is suppressed, and the disease is characterized by inappropri- ϩ ϩ ately low serum PTH and increased Ca2 and Mg2 excretion.

Furthermore, mutations in CaSR were identified in patients and Mg

with hypercalcemic disorders of familial benign (hypocalciuric) 2 ϩ hypercalcemia and neonatal severe primary hyperparathyroid- TALDCT CLDN16, paracellin 1 TRPM6, TRPM6 Hypercalciuria Hypomagnesemia Hypocalciuria Hypomagnesemia

ism (90). Inactivation of the CaSR likely leads to inappropriate Location Defect Affected Gene/Transporter ϩ ϩ ϩ reabsorption of Ca2 and Mg2 in the TAL (91) and Mg2 ϩ transport in DCT (92). Therefore, renal excretion of Ca2 and ϩ Mg2 is reduced, which leads to hypercalcemia and in some cases hypermagnesemia (93). It should be noted that the CaSR ϩ ϩ plays a role in controlling renal Ca2 and Mg2 secretion independent of its role in regulating PTH release. Recent stud- ies using double knockout mice for CaSR and PTH showed a ϩ much wider range of values for serum Ca2 and renal excretion ϩ Ϫ Ϫ of Ca2 than those in control (PTH / ) littermates, despite the absence of any circulating PTH (94,95). ϩ Patients with mutations in TRPM6, the ␥-subunit of the Na - ϩ K -ATPase, or NCC exhibit besides hypomagnesemia also hy- pocalciuria (Table 2). Expression of these affected genes is restricted to the DCT. However, an interaction between trans- ϩ ϩ cellular Ca2 and Mg2 pathways in the distal part of the nephron is still unclear. There is limited overlap in expression 2ϩ between the Ca transport proteins and TRPM6, ␥-subunit, or Inherited disorders with mutual disturbance in Ca

NCC (38,53,96). It is interesting that hypocalcemia in HSH nephrocalcinosis hypercalciuria TAL, thick ascending loop; NCC, sodium chloride co-transporter. Hypomagnesemia with hypercalciuria and Autosomal dominant hypoparathyroidismHypomagnesemia with secondary Gitelman TAL, syndrome DCTIsolated dominant hypomagnesemia CASR, CaSR DCT Hypocalciuria DCT SLC12A3, NCC Hypocalciuria Hypomagnesemia

patients can be corrected only by administration of high dietary a 2ϩ Mg content. Several studies reported that normalization of Table 2. ϩ ϩ J Am Soc Nephrol 16: 15–26, 2005 Ca2 and Mg2 Physiology 23

ϩ the hypomagnesemia by dietary supplementation resulted in a and Mg2 balance (Table 1). To date, several studies have prompt release of PTH and subsequent correction of the hy- focused on the regulation of TRPV5 and TRPV6, whereas many pocalcemia (97–99). These findings suggest that hypocalcemia questions remain to be investigated for TRPM6 and TRPM7. ϩ ϩ in HSH is caused by a disturbance in PTH-mediated Ca2 For instance, the hormonal regulation of these Mg2 channels ϩ reabsorption. The factors that determine whether Mg2 defi- has not been studied yet. The next step is to clarify the cellular ϩ ϩ ciency will result in inhibition of PTH release, a lack of response events in epithelial Ca2 and Mg2 transport. For instance, the ϩ of the bone to PTH, or both remain to be clarified. mechanisms by the DCT cells to sense the extracellular Ca2 ϩ Gitelman syndrome in adults is characterized by consistent and Mg2 concentration and appropriately adapt the transport hypomagnesemia, hypocalciuria, and hypokalemic metabolic rates are fertile areas for future research. The continued use of alkalosis (Table 2). The affected NCC gene results in loss of molecular and cell physiologic techniques to probe the consti- ϩ ϩ normal thiazide function, and the phenotype is identical to tutive and congenital disturbances of Ca2 and Mg2 metabo- patients with chronic use of thiazide diuretics. Likewise, hy- lism will increase further our understanding of renal electrolyte pocalciuria is observed when animals receive long-term treat- transport and provide new insights into the way in which renal ment with thiazides (100). Recently, it was postulated that diseases are diagnosed and managed. thiazides induce hypovolemia, which stimulates proximal elec- trolyte reabsorption, explaining the observed hypocalciuria Acknowledgments (100). This finding is in line with the observation that thiazide This work was supported by the Dutch Organization of Scientific exposure leads to structural degeneration of DCT, resulting in Research (Zon-Mw 016.006.001, Zon-Mw 902.18.298, NWO-ALW 2ϩ downregulation of Ca transport proteins, arguing an in- 810.38.004, NWO-ALW 805-09.042, NWO-ALW 814-02.001, NWO 812- 2ϩ creased transcellular Ca transport in this segment (100,101). 08.002), the Stomach Liver Intestine Foundation (MWO 03-19), Human This increased rate of apoptosis might reduce the absorptive Frontiers Science Program (RGP32/2004), and the Dutch Kidney Foun- surface area of the DCT in general and, therefore, could explain dation (C10.1881 and C03.6017). the observed hypomagnesemia. ϩ ϩ Furthermore, mutations in the ␥-subunit of Na -K -ATPase are the cause of IDH (Table 2). A cell model that hypothesized References that the mutated ␥-subunit manipulates the activity of the 1. Dai LJ, Ritchie G, Kerstan D, Kang HS, Cole DE, Quamme ϩ ϩ Na ,K -ATPase by disturbing routing of the total protein com- GA: Magnesium transport in the renal distal convoluted plex to the plasma membrane has been proposed (25). As a tubule. Physiol Rev 81: 51–84, 2001 ϩ 2. Hoenderop JG, Nilius B, Bindels RJ: Molecular mechanism consequence, the reduced intracellular K concentration, in- 2ϩ ϩ of active Ca reabsorption in the distal nephron. Annu creased intracellular Na concentration, or depolarization of ϩ Rev Physiol 64: 529–549, 2002 the membrane may subsequently lead to reduced Mg2 influx 2ϩ 3. Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, through the apical TRPM6 channel, resulting in Mg wasting Kifor O, Sun A, Hediger MA, Lytton J, Hebert SC: Cloning ϩ (25). However, the exact molecular mechanism of decreased and characterization of an extracellular Ca2 -sensing re- 2ϩ Mg reabsorption and the associated hypocalciuria remains to ceptor from bovine parathyroid. Nature 366: 575–580, 1993 be elucidated. 4. Brown EM, MacLeod RJ: Extracellular calcium sensing and ϩ ϩ Taken together, many diseases in which Ca2 and Mg2 extracellular calcium signaling. Physiol Rev 81: 239–297, disturbances are linked have been reported (Table 2). In some 2001 ϩ cases, there is an explanation for the mutual disorder in Ca2 5. Suki WN: Calcium transport in the nephron. Am J Physiol ϩ and Mg2 handling, but in the majority of the diseases, the (Lond) 237: F1–F6, 1979 6. Hoenderop JG, Nilius B, Bindels RJ: Molecular mecha- origin of this coupling is still unclear. Particularly, the limited 2ϩ ϩ ϩ nisms of active Ca reabsorption in the distal nephron. overlap between the Ca2 transport (DCT2-CNT) and Mg2 Annu Rev Physiol 64: 529–549, 2002 transport (DCT1-DCT2) machinery indicated that additional 7. Bindels RJ, Hartog A, Timmermans J, Van Os CH: Active ϩ mechanisms might be involved. Ca2 transport in primary cultures of rabbit kidney CCD:

Stimulation by 1,25-dihydroxyvitamin D3 and PTH. Am Research Directions J Physiol (Lond) 261: F799–F807, 1991 This review has focused on the identification, function, and 8. Friedman PA, Coutermarsh BA, Kennedy SM, Gesek FA: ϩ ϩ regulation of the Ca2 and Mg2 transport proteins. In the past Parathyroid hormone stimulation of calcium transport is few years, significant advances were achieved in our knowl- mediated by dual signaling mechanisms involving protein ϩ ϩ edge about the maintenance of the Ca2 and Mg2 balance. The kinase A and protein kinase C. Endocrinology 137: 13–20, ϩ identification of the epithelial Ca2 (TRPV5 and TRPV6) and 1996 ϩ 9. Van Cromphaut SJ, Dewerchin M, Hoenderop JG, Stock- Mg2 (TRPM6 and TRPM7) channels provided insight in a new ϩ ϩ mans I, Van Herck E, Kato S, Bindels RJ, Collen D, Carme- molecular concept of Ca2 and Mg2 influx in specialized liet P, Bouillon R, Carmeliet G: Duodenal calcium absorp- epithelia and other cell systems in which these channels facili- tion in vitamin D receptor-knockout mice: Functional and 2ϩ 2ϩ tate Ca and Mg transport. There are striking similarities molecular aspects. Proc Natl Acad Sci U S A 98: 13324– between the characteristics of the TRPV5/6 and TRPM6/7 13329, 2001 channel pairs, such as expression profiling, structural organi- 10. Hoenderop JG, Dardenne O, van Abel M, van der Kemp ϩ zation, and function with respect to the maintenance the Ca2 AW, Van Os CH, St-Arnaud R, Bindels RJ: Modulation of 24 Journal of the American Society of Nephrology J Am Soc Nephrol 16: 15–26, 2005

ϩ ϩ renal Ca2 transport protein genes by dietary Ca2 and 26. Simon DB, Nelson-Williams C, Bia MJ, Ellison D, Karet FE, ␣ 1,25-dihydroxyvitamin D3 in 25-hydroxyvitamin D3-1 - Molina AM, Vaara I, Iwata F, Cushner HM, Koolen M, hydroxylase knockout mice. FASEB J 16: 1398–1406, 2002 Gainza FJ, Gitleman HJ, Lifton RP: Gitelman’s variant of 11. Hoenderop JG, Muller D, Van Der Kemp AW, Hartog A, Bartter’s syndrome, inherited hypokalaemic alkalosis, is Suzuki M, Ishibashi K, Imai M, Sweep F, Willems PH, Van caused by mutations in the thiazide-sensitive Na-Cl co- Os CH, Bindels RJ: Calcitriol controls the epithelial calcium transporter. Nat Genet 12: 24–30, 1996 channel in kidney. J Am Soc Nephrol 12: 1342–1349, 2001 27. Walder RY, Shalev H, Brennan TM, Carmi R, Elbedour K,

12. Friedman PA, Gesek FA: Vitamin D3 accelerates PTH- Scott DA, Hanauer A, Mark AL, Patil S, Stone EM, Shef- dependent calcium transport in distal convoluted tubule field VC: Familial hypomagnesemia maps to chromosome cells. Am J Physiol (Lond) 265: F300–F308, 1993 9q, not to the X chromosome: Genetic linkage mapping and 13. Gesek FA, Friedman PA: Calcitonin stimulates calcium analysis of a balanced translocation breakpoint. Hum Mol transport in distal convoluted tubule cells. Am J Physiol Genet 6: 1491–1497, 1997 (Lond) 264: F744–F751, 1993 28. Schlingmann KP, Weber S, Peters M, Niemann Nejsum L, 14. Van Abel M, Hoenderop JG, Dardenne O, St Arnaud R, Vitzthum H, Klingel K, Kratz M, Haddad E, Ristoff E, Van Os CH, Van Leeuwen HJ, Bindels RJ: 1,25-dihy- Dinour D, Syrrou M, Nielsen S, Sassen M, Waldegger S,

droxyvitamin D3-independent stimulatory effect of estro- Seyberth HW, Konrad M: Hypomagnesemia with second- gen on the expression of ECaC1 in the kidney. JAmSoc ary hypocalcemia is caused by mutations in TRPM6, a new Nephrol 13: 2102–2109, 2002 member of the TRPM gene family. Nat Genet 31: 166–170, 15. Van Cromphaut SJ, Rummens K, Stockmans I, Van Herck 2002 E, Dijcks FA, Ederveen AG, Carmeliet P, Verhaeghe J, 29. Walder RY, Landau D, Meyer P, Shalev H, Tsolia M, Bo- Bouillon R, Carmeliet G: Intestinal calcium transporter rochowitz Z, Boettger MB, Beck GE, Englehardt RK, Carmi genes are upregulated by estrogens and the reproductive R, Sheffield VC: Mutation of TRPM6 causes familial hypo- cycle through vitamin D receptor-independent mecha- magnesemia with secondary hypocalcemia. Nat Genet 31: nisms. J Bone Miner Res 18: 1725–1736, 2003 171–174, 2002 16. Peng JB, Zhuang L, Berger UV, Adam RM, Williams BJ, 30. Paunier L, Radde IC, Kooh SW, Conen PE, Fraser D: Pri- Brown EM, Hediger MA, Freeman MR: CaT1 expression mary hypomagnesemia with secondary hypocalcemia in correlates with tumor grade in prostate cancer. Biochem an infant. Pediatrics 41: 385–402, 1968 Biophys Res Commun 282: 729–734, 2001 31. Shalev H, Phillip M, Galil A, Carmi R, Landau D: Clinical 17. Quamme GA, Dirks JH: Magnesium transport in the presentation and outcome in primary familial hypomag- nephron. Am J Physiol (Lond) 239: F393–F401, 1980 nesaemia. Arch Intern Med 78: 127–130, 1998 18. Quamme GA, Dirks JH: Intraluminal and contraluminal 32. Matzkin H, Lotan D, Boichis H: Primary hypomagnesemia magnesium on magnesium and calcium transfer in the rat with a probable double magnesium transport defect. nephron. Am J Physiol (Lond) 238: F187–F198, 1980 Nephron 52: 83–86, 1989 19. Hoenderop JG, van der Kemp AW, Hartog A, van de Graaf 33. Clapham DE, Runnels LW, Strubing C: The TRP ion chan- SF, van Os CH, Willems PH, Bindels RJ: Molecular identi- nel family. Nat Rev Neurosci 2: 387–396, 2001 ϩ fication of the apical Ca2 channel in 1, 25-dihydroxyvita- 34. Montell C, Birnbaumer L, Flockerzi V, Bindels RJ, Bruford

min D3-responsive epithelia. J Biol Chem 274: 8375–8378, EA, Caterina MJ, Clapham D, Harteneck C, Heller S, Julius 1999 D, Kojima I, Mori Y, Penner R, Prawitt D, Scharenberg AM, 20. Peng JB, Chen XZ, Berger UV, Vassilev PM, Tsukaguchi H, Schultz G, Shimizu S, Zhu MX: A unified nomenclature for Brown EM, Hediger MA: Molecular cloning and character- the superfamily of TRP cation channels. Mol Cell 9: 229– ization of a channel-like transporter mediating intestinal 231, 2002 calcium absorption. J Biol Chem 274: 22739–22746, 1999 35. Runnels LW, Yue L, Clapham DE: TRP-PLIK, a bifunc- 21. Simon DB, Lu Y, Choate KA, Velazquez H, Al-Sabban E, tional protein with kinase and ion channel activities. Sci- Praga M, Casari G, Bettinelli A, Colussi G, Rodriguez- ence 291: 1043–1047, 2001 ϩ ϩ Soriano J, McCredie D, Milford D, Sanjad S, Lifton RP: 36. Montell C: Mg2 homeostasis: The Mg2 nificent TRPM Paracellin-1, a renal tight junction protein required for chanzymes. Curr Biol 13: R799–R801, 2003 ϩ paracellular Mg2 resorption. Science 285: 103–106, 1999 37. Nadler MJ, Hermosura MC, Inabe K, Perraud AL, Zhu Q, 22. Manz F, Scharer K, Janka P, Lombeck J: Renal magnesium Stokes AJ, Kurosaki T, Kinet JP, Penner R, Scharenberg wasting, incomplete tubular acidosis, hypercalciuria and AM, Fleig A: LTRPC7 is a Mg.ATP-regulated divalent nephrocalcinosis in siblings. Eur J Pediatr 128: 67–79, 1978 cation channel required for cell viability. Nature 411: 590– 23. Nicholson JC, Jones CL, Powell HR, Walker RG, McCredie 595, 2001 DA: Familial hypomagnesaemia—hypercalciuria leading 38. Voets T, Nilius B, van der Kemp AW, Droogmans G, ϩ to end-stage renal failure. Pediatr Nephrol 9: 74–76, 1995 Bindels RJ, Hoenderop JG: TRPM6 forms the Mg2 influx ϩ 24. Torralbo A, Pina E, Portoles J, Sanchez-Fructuoso A, Bar- channel involved in intestinal and renal Mg2 absorption. rientos A: Renal magnesium wasting with hypercalciuria, J Biol Chem 279: 19–25, 2004 nephrocalcinosis and ocular disorders. Nephron 69: 472– 39. Montell C: Physiology, phylogeny, and functions of the 475, 1995 TRP superfamily of cation channels. Sci STKE 2001: RE1, 25. Meij IC, Koenderink JB, van Bokhoven H, Assink KF, Groe- 2001 nestege WT, de Pont JJ, Bindels RJ, Monnens LA, van den 40. Clapham DE: TRP channels as cellular sensors. Nature 426: Heuvel LP, Knoers NV: Dominant isolated renal magne- 517–524, 2003 ϩ ϩ sium loss is caused by misrouting of the Na ,K -ATPase 41. Minke B, Cook B: TRP channel proteins and signal trans- gamma-subunit. Nat Genet 26: 265–266, 2000 duction. Physiol Rev 82: 429–472, 2002 ϩ ϩ J Am Soc Nephrol 16: 15–26, 2005 Ca2 and Mg2 Physiology 25

ϩ 42. Muller D, Hoenderop JG, Merkx GF, van Os CH, Bindels Functional expression of the epithelial Ca2 channels RJ: Gene structure and chromosomal mapping of human (TRPV5 and TRPV6) requires association of the S100A10- epithelial . Biochem Biophys Res Commun annexin 2 complex. EMBO J 22: 1478–1487, 2003 275: 47–52, 2000 59. Zhuang L, Peng JB, Tou L, Takanaga H, Adam RM, Hedi- 43. Peng JB, Brown EM, Hediger MA: Structural conservation ger MA, Freeman MR: Calcium-selective ion channel, of the genes encoding CaT1, CaT2, and related cation CaT1, is apically localized in gastrointestinal tract epithelia channels. Genomics 76: 99–109, 2001 and is aberrantly expressed in human malignancies. Lab 44. Weber K, Erben RG, Rump A, Adamski J: Gene structure Invest 82: 1755–1764, 2002 and regulation of the murine epithelial calcium channels 60. Bianco S, Peng JB, Takanaga H, Kos CH, Crescenzi A, ECaC1 and 2. Biochem Biophys Res Commun 289: 1287–1294, Brown EM, Hediger MA: Mice lacking the epithelial cal- 2001 cium channel CaT1 (TRPV6) show a deficiency in intestinal 45. Hoenderop JG, Vennekens R, Muller D, Prenen J, Droog- calcium absorption despite high plasma levels of 1,25- mans G, Bindels RJ, Nilius B: Function and expression of dihydroxy vitamin D. FASEB J 18: A706, 2004 ϩ the epithelial Ca2 channel family: Comparison of the 61. Nijenhuis T, Hoenderop JG, van der Kemp AW, Bindels RJ: ϩ ϩ mammalian epithelial Ca2 channel 1 and 2. J Physiol Localization and regulation of the epithelial Ca2 channel (Lond) 537: 747–761, 2001 TRPV6 in the kidney. J Am Soc Nephrol 14: 2731–2740, 2003 46. Vennekens R, Hoenderop JG, Prenen J, Stuiver M, Willems 62. Wissenbach U, Niemeyer BA, Fixemer T, Schneidewind A, PH, Droogmans G, Nilius B, Bindels RJ: Permeation and Trost C, Cavalie A, Reus K, Meese E, Bonkhoff H, Flockerzi ϩ gating properties of the novel epithelial Ca2 channel. V: Expression of CaT-like, a novel calcium-selective chan- J Biol Chem 275: 3963–3969, 2000 nel, correlates with the malignancy of prostate cancer. J Biol 47. Nilius B, Prenen J, Vennekens R, Hoenderop JG, Bindels Chem 276: 19461–19468, 2001 RJ, Droogmans G: Modulation of the epithelial calcium 63. Fixemer T, Wissenbach U, Flockerzi V, Bonkhoff H: Ex- ϩ ϩ channel, ECaC, by intracellular Ca2 . Cell Calcium 29: 417– pression of the Ca2 -selective cation channel TRPV6 in 428, 2001 human prostate cancer: A novel prognostic marker for 48. Nilius B, Vennekens R, Prenen J, Hoenderop JG, Droog- tumor progression. Oncogene 22: 7858–7861, 2003 mans G, Bindels RJ: The single pore residue D542 deter- 64. Vennekens R, Prenen J, Hoenderop JG, Bindels RJ, Droog- ϩ ϩ mines Ca2 permeation and Mg2 block of the epithelial mans G, Nilius B: Pore properties and ionic block of the ϩ Ca2 channel. J Biol Chem 276: 1020–1025, 2001 rabbit epithelial calcium channel expressed in HEK 293 49. Jean K, Bernatchez G, Klein H, Garneau L, Sauve´R, Parent cells. J Physiol (Lond) 530: 183–191, 2001 ϩ L: The role of aspartate residues in Ca2 affinity and per- 65. Hoenderop JGJ, Vennekens R, Mu¨ller D, Prenen J, Droog- meation of the distal ECaC1 channel. Am J Physiol (Lond) mans G, Bindels RJM, Nilius B: Function and expression of ϩ 282: C665–C672, 2002 the epithelial Ca2 channel family: Comparison of the epithelial ϩ 50. Voets T, Janssens A, Droogmans G, Nilius B: Outer pore Ca2 channel 1 and 2. J Physiol (Lond) 537: 747–761, 2001 ϩ architecture of a Ca2 -selective TRP channel. J Biol Chem 66. Hoenderop JG, Voets T, Hoefs S, Weidema AF, Prenen J, 279: 15223–15230, 2004 Nilius B, Bindels RJ: Homo- and heterotetrameric architec- ϩ 51. Dodier Y, Banderali U, Klein H, Topalak O, Dafi O, Simoes ture of the epithelial Ca2 channels, TRPV5 and TRPV6. M, Bernatchez G, Sauve R, Parent L: Outer pore topology EMBO J 22: 776–785, 2003 of the ECaC-TRPV5 channel by cysteine scan mutagenesis. 67. Nilius B, Prenen J, Hoenderop JG, Vennekens R, Hoefs S, J Biol Chem 279: 6853–6862, 2004 Weidema AF, Droogmans G, Bindels RJ: Fast and slow ϩ 52. Hoenderop JG, Hartog A, Stuiver M, Doucet A, Willems inactivation kinetics of the Ca2 channels ECaC1 and ϩ PH, Bindels RJ: Localization of the epithelial Ca2 channel ECaC2 (TRPV5 and 6): Role of the intracellular loop lo- in rabbit kidney and intestine. J Am Soc Nephrol 11: 1171– cated between transmembrane segment 2 and 3. J Biol Chem 1178, 2000 277: 30852–30858, 2002 53. Loffing J, Loffing-Cueni D, Valderrabano V, Klausli L, 68. Song Y, Peng X, Porta A, Takanaga H, Peng JB, Hediger MA, Hebert SC, Rossier BC, Hoenderop JG, Bindels RJ, Kai- Fleet JC, Christakos S: Calcium transporter 1 and epithelial ssling B: Organization of the mouse distal nephron: Distri- calcium channel messenger ribonucleic acid are differentially

butions of transcellular calcium and sodium transport regulated by 1,25 dihydroxyvitamin D3 in the intestine and pathways. Am J Physiol Renal Physiol 281: F1021–F1027, kidney of mice. Endocrinology 144: 3885–3894, 2003 2001 69. Erler I, Hirnet D, Wissenbach U, Flockerzi V, Niemeyer ϩ 54. Hoenderop JG, van Leeuwen JP, van der Eerden B, Kersten BA: Ca2 -selective TRPV channel architecture and func- F, van der Kemp AW, Me´rrliat A, Waarsing E, Rossier B, tion require a specific ankyrin repeat. J Biol Chem 279: ϩ Vallon V, Hummler E, Bindels RJ: Renal Ca2 wasting, 34456–34463, 2004 ϩ hyperabsorption, and reduced bone thickness in mice lack- 70. Peng JB, Brown EM, Hediger MA: Epithelial Ca2 entry ϩ ing TRPV5. J Clin Invest 112: 1906–1914, 2003 channels: Transcellular Ca2 transport and beyond. 55. Frick KK, Bushinsky DA: Molecular mechanisms of pri- J Physiol (Lond) 551[Suppl]: 729–740, 2003 mary hypercalciuria. J Am Soc Nephrol 14: 1082–1095, 2003 71. van Abel M, Hoenderop JG, Van Leeuwen HJ, Bindels RJ: 56. Miller LA, Stapleton FB: Urinary volume in children with Down-regulation of calcium transporters in kidney and urolithiasis. J Urol 141: 918–920, 1989 duodenum by the calcimimetic compound NPS R-467 [Ab- 57. Baumann JM: Stone prevention: Why so little progress? stract]. J Am Soc Nephrol 14: 459A, 2003 Urol Res 26: 77–81, 1998 72. Greger R, Lang F, Oberleithner H: Distal site of calcium 58. van de Graaf SF, Hoenderop JG, Gkika D, Lamers D, reabsorption in the rat nephron. Pflugers Arch 374: 153–157, Prenen J, Rescher U, Gerke V, Staub O, Nilius B, Bindels RJ: 1978 26 Journal of the American Society of Nephrology J Am Soc Nephrol 16: 15–26, 2005

73. Hoenderop JG, De Pont JJ, Bindels RJ, Willems PH: Hor- drome of hypocalcemia with hypercalciuria due to muta- ϩ mone-stimulated Ca2 reabsorption in rabbit kidney cor- tions in the calcium-sensing receptor. N Engl J Med 335: tical collecting system is cAMP-independent and involves 1115–1122, 1996 a phorbol ester-insensitive PKC isotype. Kidney Int 55: 90. Pollak MR, Brown EM, Chou YH, Hebert SC, Marx SJ, 225–233, 1999 Steinmann B, Levi T, Seidman CE, Seidman JG: Mutations ϩ 74. Niemeyer BA, Bergs C, Wissenbach U, Flockerzi V, Trost in the human Ca2 -sensing receptor gene cause familial ϩ C: Competitive regulation of CaT-like-mediated Ca2 en- hypocalciuric hypercalcemia and neonatal severe hyper- try by protein kinase C and calmodulin. Proc Natl Acad Sci parathyroidism. Cell 75: 1297–1303, 1993 USA98: 3600–3605, 2001 91. Hebert SC: Extracellular calcium-sensing receptor: Impli- 75. Lambers TT, Weidema AF, Nilius B, Hoenderop JG, Bind- cations for calcium and magnesium handling in the kid- ϩ els RJ: Regulation of the mouse epithelial Ca2 channel ney. Kidney Int 50: 2129–2139, 1996 ϩ TRPV6, by the Ca2 -sensor calmodulin. J Biol Chem 279: 92. Bapty BW, Dai LJ, Ritchie G, Canaff L, Hendy GN, ϩ ϩ 28855–28861, 2004 Quamme GA: Mg2 /Ca2 sensing inhibits hormone-stim- ϩ 76. Hirnet D, Olausson J, Fecher-Trost C, Bodding M, Nas- ulated Mg2 uptake in mouse distal convoluted tubule tainczyk W, Wissenbach U, Flockerzi V, Freichel M: The cells. Am J Physiol (Lond) 275: F353–F360, 1998 TRPV6 gene, cDNA and protein. Cell Calcium 33: 509–518, 93. Kristiansen JH, Brochner Mortensen J, Pedersen KO: Fa- 2003 milial hypocalciuric hypercalcaemia I: Renal handling of 77. Gkika D, Mahieu F, Nilius B, Hoenderop JG, Bindels RJ: calcium, magnesium and phosphate. Clin Endocrinol (Oxf) ϩ 80K-H as a new Ca2 sensor regulating the activity of the 22: 103–116, 1985 ϩ epithelial Ca2 channel transient receptor potential cation 94. Kos CH, Karaplis AC, Peng JB, Hediger MA, Goltzman D, channel V5 (TRPV5). J Biol Chem 279: 26351–26357, 2004 Mohammad KS, Guise TA, Pollak MR: The calcium-sens- 78. Yang W, Lee HW, Hellinga H, Yang JJ: Structural analysis, ing receptor is required for normal calcium homeostasis identification, and design of calcium-binding sites in pro- independent of parathyroid hormone. J Clin Invest 111: teins. Proteins 47: 344–356, 2002 1021–1028, 2003 79. Runnels LW, Yue L, Clapham DE: The TRPM7 channel is 95. Tu Q, Pi M, Karsenty G, Simpson L, Liu S, Quarles LD:

inactivated by PIP2 hydrolysis. Nat Cell Biol 4: 329–336, 2002 Rescue of the skeletal phenotype in CasR-deficient mice by 80. Hermosura MC, Monteilh-Zoller MK, Scharenberg AM, Pen- transfer onto the Gcm2 null background. J Clin Invest 111: ner R, Fleig A: Dissociation of the store-operated calcium 1029–1037, 2003 current ICRAC and the Mg-nucleotide-regulated metal ion 96. Wetzel RK, Sweadner KJ: Immunocytochemical localiza- current MagNuM. J Physiol (Lond) 539: 445–458, 2002 tion of Na-K-ATPase alpha- and gamma-subunits in rat 81. Kozak JA, Cahalan MD: MIC channels are inhibited by kidney. Am J Physiol Renal Physiol 281: F531–F545, 2001 internal divalent cations but not ATP. Biophys J 84: 922–927, 97. Garty R, Alkalay A, Bernheim JL: Parathyroid hormone 2003 secretion and responsiveness to parathyroid hormone in 82. Schmitz C, Perraud AL, Johnson CO, Inabe K, Smith MK, primary hypomagnesemia. Isr J Med Sci 19: 345–348, 1983 Penner R, Kurosaki T, Fleig A, Scharenberg AM: Regula- 98. Rude RK, Oldham SB, Sharp CF Jr, Singer FR: Parathyroid ϩ tion of vertebrate cellular Mg2 homeostasis by TRPM7. hormone secretion in . J Clin Endo- Cell 114: 191–200, 2003 crinol Metab 47: 800–806, 1978 83. Chubanov V, Waldegger S, Mederos y Schnitzler M, Vitz- 99. Anast CS, Forte LF: Parathyroid function and magnesium thum H, Sassen MC, Seyberth HW, Konrad M, Gudermann depletion in the rat. Endocrinology 113: 184–189, 1983 T: Disruption of TRPM6/TRPM7 complex formation by a 100. Nijenhuis T, Hoenderop JG, Loffing J, van der Kemp AW, mutation in the TRPM6 gene causes hypomagnesemia Van Os C, Bindels RJ: Thiazide-induced hypocalciuria is ϩ with secondary hypocalcemia. Proc Natl Acad Sci U S A 101: accompanied by a decreased expression of Ca2 transport- 2894–2899, 2004 ing proteins in the distal tubule. Kidney Int 64: 555–564, 84. Drennan D, Ryazanov AG: Alpha-kinases: Analysis of the 2003 family and comparison with conventional protein kinases. 101. Loffing J, Loffing-Cueni D, Hegyi I, Kaplan MR, Hebert SC, Prog Biophys Mol Biol 85: 1–32, 2004 Le Hir M, Kaissling B: Thiazide treatment of rats provokes 85. Ryazanova LV, Dorovkov MV, Ansari A, Ryazanov AG: apoptosis in distal tubule cells. Kidney Int 50: 1180–1190, Characterization of the protein kinase activity of TRPM7/ 1996 ChaK1, a protein kinase fused to the transient receptor 102. Peng JB, Chen XZ, Berger UV, Weremowicz S, Morton CC, potential ion channel. J Biol Chem 279: 3708–3716, 2004 Vassilev PM, Brown EM, Hediger MA: Human calcium 86. Cole DE, Quamme GA: Inherited disorders of renal mag- transport protein CaT1. Biochem Biophys Res Commun 278: nesium handling. J Am Soc Nephrol 11: 1937–1947, 2000 326–332, 2000 87. Konrad M, Schlingmann KP, Gudermann T: Insights into 103. Hoenderop JG, Chon H, Gkika D, Bluyssen HA, Holstege the molecular nature of magnesium homeostasis. Am FC, St-Arnaud R, Braam B, Bindels RJ: Regulation of gene ϩ J Physiol Renal Physiol 286: F599–F605, 2004 expression by dietary Ca2 in kidneys of 25-hydroxyvita- ␣ 88. Pollak MR, Brown EM, Estep HL, McLaine PN, Kifor O, min D3-1 -hydroxylase knockout mice. Kidney Int 65: 531– Park J, Hebert SC, Seidman CE, Seidman JG: Autosomal 539, 2004 ϩ dominant hypocalcaemia caused by a Ca2 -sensing recep- 104. van Abel M, Hoenderop JG, van der Kemp AW, van Leeu- ϩ tor gene mutation. Nat Genet 8: 303–307, 1994 wen JP, Bindels RJ: Regulation of the epithelial Ca2 chan- 89. Pearce SH, Williamson C, Kifor O, Bai M, Coulthard MG, nels in small intestine as studied by quantitative mRNA Davies M, Lewis-Barned N, McCredie D, Powell H, Ken- detection. Am J Physiol Gastroinstest Liver Physiol 285: G78– dall-Taylor P, Brown EM, Thakker RV: A familial syn- G85, 2003 Erratum

Corrected text: Correction ϩ ϩ 2ϩ Mutations in the Ca2 /Mg2 -sensing receptor (CaSR) are also Hoenderop JGJ, Bindels RJM: Epithelial Ca and 2ϩ 2ϩ associated with disturbed Mg handling (Table 2). Mutations in this Mg Channels in Health and Disease. JAmSoc receptor are identified in autosomal dominant hypocalcemia with Nephrol 16:15–26, 2005 hypercalciuria (ADHH) (88). Hypomagnesemia is frequently ob- In this article from the January 2005 issue of JASN, the authors regret served in these patients (89). an error in terminology in Table 2 and in the text referring to Table 2 (see page 22). The corrected table and text are shown below.

ϩ ϩ Table 2. Inherited disorders with mutual disturbance in Ca2 and Mg2 balancea

Ca2ϩ and Mg2ϩ Homeostasis Affected Gene/ Location Defect ϩ ϩ Transporter Ca2 Mg2 Disturbance Disturbance

Hypomagnesemia with hypercalciuria TAL CLDN16, paracellin 1 Hypercalciuria Hypomagnesemia and nephrocalcinosis (HHN) Autosomal dominant hypocalcemia TAL, DCT CASR, CaSR Hypocalcemia Hypomagnesemia with hypercalciuria (ADHH) hypercalciuria Hypomagnesemia with secondary DCT TRPM6, TRPM6 Hypocalcemia Hypomagnesemia hypocalcemia (HSH) hypocalciuria Gitelman syndrome (GS) DCT SLC12A3, NCC Hypocalciuria Hypomagnesemia Isolated dominant hypomagnesemia DCT ␥-subunit Naϩ-Kϩ-ATPase Hypocalciuria Hypomagnesemia (IDH)

aTAL, thick ascending loop; NCC, sodium chloride co-transporter.