Sie sind auf Seite 1von 8

BRIEF REVIEW www.jasn.

org

Molecular Determinants of Magnesium Homeostasis:


Insights from Human Disease
R. Todd Alexander, Joost G. Hoenderop, and René J. Bindels
Department of Physiology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre,
Nijmegen, Netherlands

ABSTRACT
The past decade has witnessed multiple advances in our understanding of mag- that the molecular determinants of this
nesium (Mg2⫹) homeostasis. The discovery that mutations in claudin-16/paracel- process have become clearer. Much of
lin-1 or claudin-19 are responsible for familial hypomagnesemia with hypercalciuria this insight derives from the study of pa-
and nephrocalcinosis provided insight into the molecular mechanisms governing tients, often children, with disorders of
paracellular transport of Mg2⫹. Our understanding of the transcellular movement Mg2⫹ wasting. From these studies, the
of Mg2⫹ was similarly enhanced by the realization that defects in transient receptor molecular determinants of Mg2⫹ ho-
potential melastatin 6 (TRPM6) cause hypomagnesemia with secondary hypocal- meostasis have started to be unraveled.
cemia. This channel regulates the apical entry of Mg2⫹ into epithelia. In so doing,
TRPM6 alters whole-body Mg2⫹ homeostasis by controlling urinary excretion.
Consequently, investigation into the regulation of TRPM6 has increased. Acid-base INTESTINAL UPTAKE
status, 17␤ estradiol, and the immunosuppressive agents FK506 and cyclosporine
affect plasma Mg2⫹ levels by altering TRPM6 expression. A mutation in epithelial Typically, 300 mg of Mg2⫹ is ingested
growth factor is responsible for isolated autosomal recessive hypomagnesemia, daily, 24 to 75% of which is absorbed, a
and epithelial growth factor activates TRPM6. A defect in the ␥-subunit of the process dependent on body stores and
Na,K-ATPase causes isolated dominant hypomagnesemia by altering TRPM6 ac- dietary content.8 –10 The entire length of
tivity through a decrease in the driving force for apical Mg2⫹ influx. We anticipate the bowel is capable of absorbing Mg2⫹.
that the next decade will provide further detail into the control of the gatekeeper As occurs in the nephron, intestinal ab-
TRPM6 and, therefore, overall whole-body Mg2⫹ balance. sorption proceeds in both a passive para-
cellular and an active transcellular man-
J Am Soc Nephrol 19: 1451–1458, 2008. doi: 10.1681/ASN.2008010098
ner.8,11 Mg2⫹ absorption from the small
bowel occurs predominately in a paracel-
lular manner.12,13 Given the appropriate
Magnesium (Mg2⫹) is the second most meostasis is multifold. Hypermag- driving force, significant paracellular ab-
common intracellular cation.1 Its abun- nesemia can cause neurologic and car- sorption can also take place in the co-
dance facilitates multiple roles that it diac sequelae, including lethargy, lon.12 The kinetics of this movement are
plays in common, essential intracellular confusion, coma, a prolongation in the governed by active absorption of sodium
processes. It is a co-factor in multiple en- PR interval, widened QRS, complete (Na⫹) followed by water.14 Mg2⫹ and
zymatic reactions, including those in- heart block, and cardiac arrest.4 Hypo- other ions flow down their concentra-
volving energy metabolism and DNA magnesemia results in similar clinical tion gradient from bowel lumen to peri-
and protein synthesis, and it participates manifestations that include tetany, sei- intestinal capillary. It is noteworthy that
in the regulation of ion channels.2 Mg2⫹ zures, and cardiac arrhythmias.4,5 Al-
homeostasis is therefore fundamental to tered plasma Mg2⫹ levels can in turn af- Published online ahead of print. Publication date
the existence of life. Mg2⫹ balance in the fect calcium (Ca2⫹) and potassium (K⫹) available at www.jasn.org.
body is controlled by a dynamic interplay levels.4 –7 Thus, understanding Mg2⫹ ho- Correspondence: Dr. René J. Bindels, 286 Phys-
among intestinal absorption, exchange meostasis is important not only for the iology, Nijmegen Centre for Molecular Life Sci-
with bone, and renal excretion.3 This last treatment of these disorders but also for ences, Radboud University Nijmegen Medical
Centre, P.O. Box 9101, 6500 HB Nijmegen, Neth-
process is where the greatest regulation the understanding and management of erlands. Phone: ⫹31-24-3614211; Fax: ⫹31-24-
occurs and consequently is the major fo- other electrolyte abnormalities. Al- 3616413; E-mail: r.bindels@ncmls.ru.nl
cus of this review. though Mg2⫹ homeostasis has been Copyright 䊚 2008 by the American Society of
The consequence of altered Mg2⫹ ho- studied for decades, it is only recently Nephrology

J Am Soc Nephrol 19: 1451–1458, 2008 ISSN : 1046-6673/1908-1451 1451


BRIEF REVIEW www.jasn.org

under conditions producing a luminal BONE: A MG2ⴙ RESERVOIR tween 3 and 5%35; however, in the pres-
driving force, such as diarrheal states, ence of hypomagnesemia, this can be de-
Mg2⫹ can be secreted into the lumen of The majority of the body’s Mg2⫹ creased to 0.5 to 1% to conserve Mg2⫹
the gut along with water and electro- (⬎50%) resides within the skeleton, as stores.36 The majority of filtered Mg2⫹ is
lytes.11 part of the hydroxyapatite crystalline absorbed in the proximal tubule (ap-
Active transcellular absorption of structure.1,9,25 As is the case for Ca2⫹, proximately 20%) and thick ascending
Mg2⫹ occurs almost exclusively in the bone is thought to provide a buffer for limb (TAL) of the loop of Henle (ap-
colon.5,8 The rare monogenetic disorder plasma Mg2⫹, leaching Mg2⫹ when proximately 70%) by a passive paracellu-
hypomagnesemia with secondary hy- plasma levels drop and facilitating the lar mechanism (Figure 1).37,38 That two
pocalcemia (HSH) provides molecular synthesis of new bone when the circulat- thirds of filtered Mg2⫹ is absorbed in the
insight into this process. Children with ing level is plentiful.26 Consistent with TAL and not the proximal tubule is
this disease have seizures and tetany, sec- this notion, animals fed Mg2⫹-deficient unique for renal tubular ion transport.
ondary to extremely low Mg2⫹ levels.15,16 diets have a bone Mg2⫹ content that is All ions studied to date, except for Mg2⫹,
Their hypomagnesemia is due to a failure reduced by 30 to 40% and a reduced are reabsorbed to a greater extent in the
in the active transcellular (re)absorption bone mineral density.27–29 This is medi- proximal tubule. The remaining 10% of
of Mg2⫹ from both the gut and the kid- ated by a decrease in the number of os- filtered Mg2⫹ is absorbed by an active
ney.17–20 A mutation in the transient re- teoblasts, with inhibited function,9,27,28 transcellular mechanism in DCT.6 This
ceptor potential (TRP) channel transient and an increase in both the number and latter process ultimately controls the
receptor potential melastatin 6 (TRPM6) function of osteoclasts.9,27–29 Finally, amount of Mg2⫹ excreted in the urine,
was found to be responsible for this dis- current data, although by no means con- because no Mg2⫹ reabsorption occurs
ease.21,22 Localization studies demon- clusive, suggest that Mg2⫹ deficiency distal to this segment.6 The DCT is there-
strated this channel in the colon and dis- predisposes an individual to osteoporo- fore the predominant site of specifically
tal convoluted tubule (DCT), which is sis.29 –32 Unfortunately, our understand- regulated Mg2⫹ excretion. Insights gleaned
the site of active renal transcellular reab- ing of the molecular details governing from the delineation of monogenetic
sorption.23,24 At the subcellular level, the incorporation of Mg2⫹ into bone by Mg2⫹ wasting disorders have proved
TRPM6 is predominantly expressed api- osteoblasts and its retrieval by osteoclasts highly valuable in deciphering the mo-
cally.23 Hence, elucidation of this rare ge- is minimal. lecular events governing renal regulated
netic disorder resulted in the discovery of excretion (Figure 1, Table 1).39,40
the apical entry mechanism for Mg2⫹
into epithelia. The molecular identity of RENAL REGULATION OF MG2ⴙ Paracellular Transport: The
the protein responsible for the basolat- EXCRETION Proximal Tubule and Loop of Henle
eral exit of Mg2⫹ from the epithelial cell Reabsorption of Mg2⫹ from the lumen
remains unidentified. Furthermore, Approximately 80% of total plasma of the proximal tubule and TAL occurs in
whether there exists an intracellular Mg2⫹ is filtered by the glomeruli,33,34 the a passive, paracellular manner.38 Mg2⫹
chaperone that facilitates transcellular vast majority of which is absorbed along flows between the epithelial cells down
diffusion of Mg2⫹, as occurs for Ca2⫹, is the course of the nephron.6 On a normal its electrochemical gradient.39 The exact
also not known. diet, fractional excretion of Mg2⫹ is be- determinants that govern paracellular
Figure 1. Renal regulation of Mg2⫹ ho-
TAL DCT DCT meostasis. A total of 80% of Mg2⫹ is fil-
10%
PT
Tight junction tered at the glomeruli, 15% of which is
15%
2+ absorbed proximally, 70% in the TAL, and
Claudin-16/19 Mg
EGFR 15% in the DCT, leaving 3 to 5% to be
γ +
NKCC2 Na– α Na, K Rack1 excreted in the urine. The TAL is the main
β ATPase – EGF
TRPM6 Rac site of passive paracellular reabsorption of
Cl– CICKb ?
K +
– + Mg2⫹, a process mediated by claudin-16
ROMK TAL Mg2+
and -19. This paracellular reabsorption de-
70% γ
α Na, K pends on the active reabsorption of Na⫹,
CaSR β ATPase
which is mediated by apical entry through
–70 mV
Ca2+ sodium potassium chloride cotransporter
Estradiol
(NKCC) and efflux via the Na⫹,K⫹-ATPase.
Apical
+15 mV
Basolateral
Urine
Apical Basolateral
Efflux of chloride (Cl⫺) occurs through
3–5%
CLCKb, and K⫹ is recycled back into the
lumen via ROMK. The Ca -sensing receptor (CaSR) acts to inhibit this process and prevent both paracellular Ca2⫹ and Mg2⫹
2⫹

reabsorption. In the DCT, luminal Mg2⫹ enters via TRPM6. The mediator of its efflux is unknown. Intracellular Mg2⫹ and RACK1 inhibit
TRPM6. EGF, cleaved from the basolateral membrane, activates TRPM6, and its expression is increased by estradiol. EGFR, EGF
receptor; PT, proximal tubule.

1452 Journal of the American Society of Nephrology J Am Soc Nephrol 19: 1451–1458, 2008
www.jasn.org BRIEF REVIEW

Table 1. Proteins implicated in the molecular control of Mg2⫹ homeostasisa


Associated Human
Protein Gene Localization Function Reference
Disease
Claudin-16/ CLDN16 TAL, tight junction Permissive of paracellular FHHNC 41–44,53
paracellin-1 permeability
Claudin-19 CLDN19 TAL, tight junction Permissive of paracellular FHHNC 49,50,53
permeability
NCC SLC12A3 DCT, apical membrane Sodium chloride co-transporter Gitelman syndrome 54,55
TRPM6 TRPM6 DCT, apical membrane, colon, lung Selective Mg2⫹ channel, apical HSH 17–23
entry in transcellular
transport
Na,K-ATPase FXYD2 PT, MD/DCT, ?TAL, medulla Alters kinetics of Na⫹ and K⫹ Isolated dominant 76–80,82
␥-subunit exchange hypomagnesemia
EGF EGF Adrenal, brain, heart, kidney (DCT), Increases TRPM6 activity Isolated recessive 87
salivary gland, spleen, thymus, hypomagnesemia
intestine, thyroid, and uterus
a
The gene name, renal localization, function, and associated human disease of proteins known to be involved in the molecular control of Mg2⫹ homeostasis.
PT, proximal tubule.

Mg2⫹ movement are, as yet, unknown. mutations in claudin-19, which is also where it occurs in an active transcellular
Surprising, whereas ⬎60% of filtered located at the tight junction of the manner.6 There has been significant re-
Na⫹ and water is absorbed in the proxi- TAL.49,50 That claudin-16 affects para- cent progress in understanding the mo-
mal tubule, only approximately 15% of cellular permeability is supported by lecular details governing this process.
Mg2⫹ is reabsorbed in this segment.37,38 heterologous expression studies dem- Study of the affected protein, TRPM6, in
This is in direct contrast to Ca2⫹, another onstrating increased paracellular flux patients with HSH reveals its localization
divalent cation reabsorbed by the para- of cations.51,52 A recent study extended to the DCT.23 Moreover, Mg2⫹ loading
cellular route, which is absorbed in a this observation to demonstrate a func- experiments demonstrated that these in-
similar ratio to Na⫹ and water.40 The tional interaction between claudin-16 dividuals not only failed to absorb Mg2⫹
majority of Mg2⫹ is absorbed in the TAL, and -19 in renal epithelia at the tight from the gut but also had a renal leak.17
approximately 70%.39 This observation junction, which increases cation selec- As with intestinal epithelia, subcellular
suggests that something facilitates para- tivity above that of claudin-16 in isola- localization of TRPM6 in DCT is apical,
cellular Mg2⫹ absorption in the TAL that tion.53 supporting a role for this channel in lu-
is absent in the proximal tubule.37 A molecular enigma is presented by the minal Mg2⫹ influx.23 In parallel to the
It was through study of a rare disor- most common monogenetic disorder re- gut, the mechanism of basolateral efflux
der of Mg2⫹ wasting, familial hypo- sulting in Mg2⫹ wasting, Gitelman syn- of Mg2⫹ is unknown, although it is spec-
magnesemia with hypercalciuria and drome. Patients with this disorder have a ulated this occurs through exchange with
nephrocalcinosis (FHHNC), that the mutation in the thiazide-sensitive Na⫹/ Na⫹ in a secondarily active process (Fig-
molecular identity of this paracellular Cl⫺ co-transporter (NCC).54,55 Their dis- ure 1).59 Because the apical membrane
mediator was identified. Patients with ease is characterized by a hypokalemic potential in the DCT is approximately
FHHNC have mutations in claudin-16/ metabolic alkalosis with hypomagnesemia ⫺70 mV or greater,60,61 the Nernst po-
paracellin-1,41– 44 a protein localized and hypocalciuria.56 Inhibition of NCC tential favors Mg2⫹ influx. In fact, be-
exclusively to the tight junction of epi- with thiazide diuretics results in a similar cause intracellular and extracellular
thelia in the TAL.43,44 Claudins are tet- phenotype.57 Mice genetically engineered Mg2⫹ concentrations are compara-
raspanning, transmembrane proteins with null alleles of the gene encoding NCC ble,62,63 membrane potential is likely the
localized to the tight junction by zona or those treated with thiazide diuretics major determinant of apical Mg2⫹ en-
occludens proteins.45,46 Their localiza- have decreased expression of TRPM6.58 try.26 Consequently, energy must be ex-
tion and both intracellular and inter- This channel is the protein responsible for pended to effect its efflux at the basolat-
cellular interactions are postulated to apical entry of luminal Mg2⫹ and provides eral membrane.6 As such, it is possible
form pores that regulate paracellular an explanation for the Mg2⫹ wasting ob- that Mg2⫹ efflux is mediated by an ATP-
movement of ions.47,48 Absent or faulty served in this disease23; however what sig- dependent Mg2⫹ pump.6
claudin-16 activity could therefore nals the decrease in abundance of TRPM6 It is unclear whether intracellular
prevent the reabsorption of Mg2⫹ (as in the absence of NCC activity is unclear. Mg2⫹ is buffered by a chaperone in the
well as Ca2⫹) and result in FHHNC. kidney; if so, then the identity of such a
Consistent with this hypothesis is the Transcellular Transport: The DCT chaperone is also a matter of specula-
recent observation that a similar phe- The DCT is the last site of Mg2⫹ reab- tion.64 In DCT, parvalbumin and, to a
notype is observed in individuals with sorption in the nephron and the only site lesser extent, calbindin-D28K have over-

J Am Soc Nephrol 19: 1451–1458, 2008 Magnesium Homeostasis 1453


BRIEF REVIEW www.jasn.org

lapping expression with TRPM6.23,65 try of Mg2⫹ into epithelia; consequently, required for plasma membrane localiza-
Both proteins have an affinity for Mg2⫹ the amount of information with respect to tion and TRPM6-specific currents.69,72,73
that favors binding under physiologic its regulation is limited (Table 2). Its loca- These authors used electrochemical, bio-
conditions.66 Recently, investigators tion and function position it to be a key chemical, and immunofluorescent tech-
made a parvalbumin null mouse.65 The modulator of Mg2⫹ homeostasis. As such, niques to show that a direct interaction
null animal has polydipsia and polyuria this is an area of active research with several between TRPM6 and TRPM7 is required
as a result of a decrease in NCC expres- recent, interesting results. for plasma membrane localization of
sion. Ca2⫹ excretion is somehow de- TRPM6.69,72,73 What the actual composi-
creased, whereas renal Mg2⫹ excretion is Structure of TRPM6 tion of the functional unit of channels is
unaltered, neither confirming nor ex- TRPM6 is predicted to share structural in native epithelia remains unclear.
cluding parvalbumin from a role in DCT homology to other TRP channels. It is
Mg2⫹ reabsorption. Whether these ani- composed of a large intracellular amino- Function of TRPM6
mals will inappropriately excrete Mg2⫹ terminus, six membrane-spanning do- TRPM6 and TRPM7 are unique chan-
when deprived remains unclear. Regard- mains that make up the channel pore, nels. They conduct Mg2⫹ preferentially
less, intracellular concentrations of and a large intracellular carboxy-termi- over Ca2⫹23,68 and contain a functional
Mg2⫹ in the millimolar range are not nal domain. Fused to the carboxy-termi- ␣-kinase domain. This has dubbed them
detrimental to a cell (as would be the case nus is an ␣-kinase domain.67 The func- chanzymes.67 The ␣-kinase domain plays
for Ca2⫹), and, unlike Ca2⫹ that is used tional unit is thought to be a homo- or a role in regulating channel activity. This
as a dynamic signaling molecule, the in- heterotetramer with TRPM7.68,69 The domain is not necessary for basal func-
tracellular concentration of Mg2⫹ is not exact composition is debated in the liter- tion; however, as with TRPM7, auto-
known to fluctuate. This obviates the ne- ature. We have been able to express phosphorylation is a mechanism regulat-
cessity for a Mg2⫹ chaperone; however TRPM6 successfully in mammalian cells ing channel activity.74,75 TRPM6 is a
such a chaperone may increase the rate at and characterize the electrophysiologic cation-selective channel with strong out-
which Mg2⫹ can diffuse from the apical properties of the channel without coex- ward rectification. It is inhibited by ru-
to basolateral cell surface. Alternatively, pressing TRPM7.23 Consistent with this, thenium red in a voltage-dependent
because intracellular Mg2⫹ is known to Li et al.68 detected TRPM6-specific cur- manner, and intracellular Mg2⫹ acts as a
inhibit TRPM6,23 such a chaperone rents from the plasma membrane of cells negative regulator of channel activity.23
would relieve the Mg2⫹-dependent inhi- in the presence and absence of TRPM7. Furthermore, an acidic extracellular pH
bition of TRPM6. Both of these possibil- Furthermore, two separate studies dem- inhibits the conductance of TRPM6, a
ities would increase the efficiency of onstrated by heterologous expression characteristic dependent on specific res-
transcellular Mg2⫹ transport. This op- (without coexpressing TRPM7) that mu- idues in the pore region.71 Together these
tion remains speculative, and further re- tation of a single residue (E1024) in the properties provide a means to regulate
search will elucidate whether this mech- pore region of TRPM6 alters cation selec- the apical entry of Mg2⫹ from tubular
anism exists in vivo. tivity of the channel70,71 and its sensitiv- fluid and ultimately regulate whole-body
ity to extracellular pH.71 Together, these Mg2⫹ homeostasis.
studies suggested that TRPM6 can func- Recently, a new TRPM6-interacting
TRPM6 tion as a homotetramer. This is in con- protein, receptor for activated C-ki-
trast to findings in Xenopus oocytes and nase 1 (RACK1), has been described.75
TRPM6 has only recently been confirmed in inducible mammalian cell culture sys- This protein interacts directly with the
as the channel responsible for the apical en- tems, where coexpression of TRPM7 is ␣-kinase domain inhibiting channel

Table 2. Effectors of Mg2⫹ homeostasisa


Effector Effect on TRPM6 Effect on Mg2ⴙ Homeostasis Reference
EGF 1 Activity, mechanism unknown Hypomagnesuria (⫾hypermagnesemia) 87–89
RACK1 1 Activity, via association and nd 75
phosphorylation of the ␣ kinase domain
Mg2⫹ (intracellular) 2 Activity 1 Urinary Mg2⫹ excretion 23,24
Mg2⫹ (extracellular) 2 TRPM6 expression 1 Urinary Mg2⫹ excretion 24
Acidosis 2 TRPM6 expression/2 activity 1 Urinary Mg2⫹ excretion (⫾hypomagnesemia) 71,94
Alkalosis 1 TRPM6 expression 2 Urinary Mg2⫹ excretion (⫾hypermagnesemia) 94
17␤-estradiol 1 TRPM6 expression 2 Urinary Mg2⫹ excretion 24
FK506/cyclosporine 2 TRPM6 expression 1 Urinary Mg2⫹ excretion (⫾hypomagnesemia) 95,96
Thiazide diuretic 2 TRPM6 expression 1 Urinary Mg2⫹ excretion (⫾hypomagnesemia) 58
a
The known effectors of TRPM6 and their effect on TRPM6 activity and the mechanism (if known) and their affect on magnesium (Mg2⫹) homeostasis.
nd, not determined.

1454 Journal of the American Society of Nephrology J Am Soc Nephrol 19: 1451–1458, 2008
www.jasn.org BRIEF REVIEW

activity. The interaction itself is inde- Local Regulation of TRPM6 eumagnesemic secondary to an in-
pendent of the phosphorylation state Recently, the identification of the caus- creased renal excretion of Mg2⫹.24
of the kinase domain; however, ative mechanism for another Mg2⫹- 17␤-Estradiol also increased the ex-
RACK1-mediated inhibition requires wasting disease, isolated autosomal re- pression of TRPM6.24 Indeed, ovariecto-
autophosphorylation of residue T1851 cessive hypomagnesemia (IRH), mized rats showed a decrease in levels of
in the kinase domain.75 Furthermore, provided further insight into the regula- TRPM6 (and magnesuria) that was nor-
the inhibition of TRPM6 activity by in- tion of TRPM6 and identified the first malized by administration of the hor-
tracellular Mg2⫹ depends on this auto- magnesiotropic hormone.87 A family mone.24 This is in contrast to vitamin D
phosphorylation. It is possible, there- with hypomagnesemia was found to and parathyroid hormone, which are un-
fore, that TRPM6-mediated Mg2⫹ have a mutation in the gene encoding ep- able to alter TRPM6 expression in vivo;
influx induces phosphorylation of ithelial growth factor (EGF) that is ex- however, they both increased Mg2⫹ in-
T1851 located in the ␣-kinase domain, pressed in DCT along with TRPM6. The flux in a DCT cell culture model as mea-
a process that activates the inhibitory mutation was in the cytosolic carboxy- sured by radiometric imaging using
effect of RACK1. This last step may act terminus, within a conserved basolat- Mag-fura.3,6 These results can be recon-
as an intracellular feedback mechanism eral-sorting motif (PXXP), prompting us ciled as an effect mediated by altered ac-
controlling TRPM6-mediated Mg2⫹ to suggest that trafficking to the basolat- tivity, not by expression level of the chan-
influx and preventing Mg2⫹ overload eral membrane and consequent release nel. Given the presumed clarity of these
during renal epithelial Mg2⫹ transport. into the extracellular space are inhibited. later studies characterizing the affect of
The notion that transcellular Mg2⫹ Consistent with our hypothesis, the ap- hormones on Mg2⫹ influx in cell culture
transport, specifically Mg2⫹ flux through plication of EGF to cells expressing and the available micropuncture and mi-
TRPM6, is dependent on membrane po- TRPM6 increased the activity of this croperfusion data,6,92 it is likely that our
tential is supported by the finding that a channel. Furthermore, when culture me- understanding of the hormonal regula-
defect in the ␥-subunit of the Na,K-AT- dium from cells expressing wild-type tion of TRPM6 activity will grow even
Pase causes isolated dominant hypomag- EGF was applied to TRPM6, channel ac- further.
nesemia.76 The exact location of this kid- tivity increased, whereas medium from The acid-base status of an individual
ney-specific subunit of the Na,K-ATPase cells expressing mutant EGF did not affects the body’s handling of Mg2⫹.92,93
is debated, although it has been localized stimulate TRPM6.87 Several other obser- This occurs through an alteration in lev-
to the same part of the nephron as vations support a role for EGF in regulat- els of TRPM6. Mice with chronic meta-
TRPM6, the DCT,77– 80 as well as the re- ing TRPM6 activity. Lactating ewes, bolic acidosis display a reduced renal ex-
nal medulla.81 The null mouse has nearly when administered EGF, have a decrease pression of TRPM6, increased excretion
a 50% increase in Mg2⫹ excretion,78 and in fractional excretion of Mg2⫹ and de- of Mg2⫹, and decreased plasma Mg2⫹,94
although not significantly different from velop hypermagnesemia.88 Patients treated whereas metabolic alkalosis results in the
wild-type mice, it certainly suggests a with the anticancer agent cetuximab, a opposite effect.94 Finally, long-term ad-
role for this protein in renal Mg2⫹ han- mAb that blocks the EGF receptor, de- ministration of the immunosuppressive
dling. Expression with the other subunits velop hypomagnesemia secondary to in- agent FK506 commonly causes hypo-
alters the kinetics of the pump such that creased renal wasting.89 Taken together, magnesemia. Both this compound and
it has an increased affinity for K⫹ at neg- these findings suggest that renal EGF acts cyclosporine mediate this effect by de-
ative membrane potentials,77,82 a de- in an autocrine or a paracrine manner to creasing the expression of TRPM6, ex-
creased affinity for Na⫹,77,83 and altered increase TRPM6 activity. This stimulates plaining this common clinical complica-
affinity for ATP.84 Coexpression of the the reabsorption of Mg2⫹ from DCT and tion of kidney transplantation.95,96
mutant subunit with wild-type protein consequently decreases the fractional ex-
prevented trafficking to the plasma cretion of Mg2⫹ (Figure 1).90
membrane,76,85 although association be- CONCLUSIONS
tween wild-type and mutant subunits is Systemic Regulation of TRPM6
not observed with synthetic peptides.86 Reduction in dietary Mg2⫹ results in hy- Studies on monogenetic disorders of
Regardless, decreased or absent ␥-sub- pomagnesemia. This in turn stimulates Mg2⫹ wasting have unraveled the molec-
units affect pump activity and conse- Mg2⫹ reabsorption along the DCT.35,91 ular details of renal and intestinal Mg2⫹
quently alter intracellular K⫹ concentra- Rodents fed a diet deficient in Mg2⫹ absorption and consequently whole-
tion. This could, in turn, inhibit demonstrated an increase in colonic, ce- body Mg2⫹ homeostasis. Renal paracel-
transcellular transport of Mg2⫹ because cal, and DCT expression of TRPM6.24 lular transport of Mg2⫹ in the TAL oc-
of an altered membrane potential. The Coincident with this increase in TRPM6, curs in the presence of claudin-16/19,
exact mechanism causing increased uri- their urinary excretion of Mg2⫹ (and and loss of functioning claudin-16/19
nary Mg2⫹ excretion and hypomag- Ca2⫹) diminished. Conversely, animals causes FHHNC as a result of Mg2⫹ wast-
nesemia has yet to be determined but is fed a diet high in Mg2⫹ paradoxically up- ing. Gitelman syndrome, a disease char-
an area of active research. regulated colonic TRPM6 yet remained acterized by renal salt loss because of a

J Am Soc Nephrol 19: 1451–1458, 2008 Magnesium Homeostasis 1455


BRIEF REVIEW www.jasn.org

defect in NCC, results in hypokalemic 4. Topf JM, Murray PT: Hypomagnesemia and mann Nejsum L, Vitzthum H, Klingel K, Kratz
hypermagnesemia. Rev Endocr Metab Dis- M, Haddad E, Ristoff E, Dinour D, Syrrou M,
metabolic alkalosis and hypomag-
ord 4: 195–206, 2003 Nielsen S, Sassen M, Waldegger S, Seyberth
nesemia. The exact mechanism of this 5. Tong GM, Rude RK: Magnesium deficiency HW, Konrad M: Hypomagnesemia with sec-
latter finding remains unknown; how- in critical illness. J Intensive Care Med 20: ondary hypocalcemia is caused by muta-
ever, absent or inhibited NCC activity 3–17, 2005 tions in TRPM6, a new member of the TRPM
leads to the downregulation of TRPM6, 6. Dai LJ, Ritchie G, Kerstan D, Kang HS, Cole gene family. Nat Genet 31: 166 –170, 2002
DE, Quamme GA: Magnesium transport in 22. Walder RY, Landau D, Meyer P, Shalev H,
the DCT channel responsible for apical
the renal distal convoluted tubule. Physiol Tsolia M, Borochowitz Z, Boettger MB, Beck
entry of Mg2⫹. This channel plays a cen- Rev 81: 51– 84, 2001 GE, Englehardt RK, Carmi R, Sheffield VC:
tral role in Mg2⫹ homeostasis by regulat- 7. Huang CL, Kuo E: Mechanism of hypokale- Mutation of TRPM6 causes familial hypo-
ing the luminal entry of Mg2⫹ in both the mia in magnesium deficiency. J Am Soc magnesemia with secondary hypocalcemia.
DCT and the intestine. The absence or Nephrol 18: 2649 –2652, 2007 Nat Genet 31: 171–174, 2002
8. Schweigel M, Martens H: Magnesium trans- 23. Voets T, Nilius B, Hoefs S, van der Kemp
malfunction of TRPM6 results in severe
port in the gastrointestinal tract. Front Biosci AW, Droogmans G, Bindels RJ, Hoenderop
hypomagnesemia and is responsible for 5: D666 –D677, 2000 JG: TRPM6 forms the Mg2⫹ influx channel
HSH. TRPM6 is regulated at the tran- 9. Rude RK, Gruber HE: Magnesium deficiency involved in intestinal and renal Mg2⫹ ab-
scriptional level by acid-base status, 17␤- and osteoporosis: Animal and human obser- sorption. J Biol Chem 279: 19 –25, 2004
estradiol, and both FK506 and cyclo- vations. J Nutr Biochem 15: 710 –716, 2004 24. Groenestege WM, Hoenderop JG, van den
10. Kayne LH, Lee DB: Intestinal magnesium ab- Heuvel L, Knoers N, Bindels RJ: The epithe-
sporine. We are just beginning to
sorption. Miner Electrolyte Metab 19: 210 – lial Mg2⫹ channel transient receptor poten-
understand how its activity is regulated 217, 1993 tial melastatin 6 is regulated by dietary
in a shorter time scale through traffick- 11. Fine KD, Santa Ana CA, Porter JL, Fordtran Mg2⫹ content and estrogens. J Am Soc
ing to the plasma membrane or from al- JS: Intestinal absorption of magnesium from Nephrol 17: 1035–1043, 2006
terations in channel kinetics. To this end, food and supplements. J Clin Invest 88: 25. Wallach S: Effects of magnesium on skeletal
396 – 402, 1991 metabolism. Magnes Trace Elem 9: 1–14,
both EGF and RACK1 are implicated in
12. Behar J: Magnesium absorption by the rat 1990
its acute regulation. Indeed, mutations in ileum and colon. Am J Physiol 227: 334 – 26. van de Graaf SF, Bindels RJ, Hoenderop JG:
the former protein have been found to 340, 1974 Physiology of epithelial Ca2⫹ and Mg2⫹
cause IRH. 13. Brannan PG, Vergne-Marini P, Pak CY, Hull transport. Rev Physiol Biochem Pharmacol
AR, Fordtran JS: Magnesium absorption in the 158: 77–160, 2007
human small intestine: Results in normal sub- 27. Gruber HE, Rude RK, Wei L, Frausto A, Mills
jects, patients with chronic renal disease, and BG, Norton HJ: Magnesium deficiency: Ef-
ACKNOWLEDGMENTS
patients with absorptive hypercalciuria. J Clin fect on bone mineral density in the mouse
Invest 57: 1412–1418, 1976 appendicular skeleton. BMC Musculoskelet
The laboratory of R.J.B. and J.G.H. is sup- 14. Frizzell RA, Schultz SG: Ionic conductances Disord 4: 7, 2003
ported by the Netherlands Organization for of extracellular shunt pathway in rabbit il- 28. Rude RK, Gruber HE, Wei LY, Frausto A,
Scientific Research (Zon-Mw 016.006.001, eum: Influence of shunt on transmural so- Mills BG: Magnesium deficiency: Effect on
Zon-Mw 9120.6110), a EURYI award from dium transport and electrical potential dif- bone and mineral metabolism in the mouse.
ferences. J Gen Physiol 59: 318 –346, 1972 Calcif Tissue Int 72: 32– 41, 2003
the European Science Foundation, Human 15. Paunier L, Radde IC, Kooh SW, Conen PE, 29. Rude RK, Kirchen ME, Gruber HE, Meyer
Frontiers Science Program (RGP32/2004), Fraser D: Primary hypomagnesemia with MH, Luck JS, Crawford DL: Magnesium de-
and the Dutch Kidney foundation (C02.2030, secondary hypocalcemia in an infant. Pedi- ficiency-induced osteoporosis in the rat: Un-
C03.6017). R.T.A. is supported by a phase I, atrics 41: 385– 402, 1968 coupling of bone formation and bone re-
CIHR Clinician Scientist award and a KRES- 16. Dudin KI, Teebi AS: Primary hypomagne- sorption. Magnes Res 12: 257–267, 1999
saemia: A case report and literature review. 30. Rude RK: Magnesium deficiency: A cause of
CENT postdoctoral award from the Kidney Eur J Pediatr 146: 303–305, 1987 heterogeneous disease in humans. J Bone
Foundation of Canada. 17. Matzkin H, Lotan D, Boichis H: Primary hy- Miner Res 13: 749 –758, 1998
pomagnesemia with a probable double 31. Gur A, Colpan L, Nas K, Cevik R, Sarac J,
magnesium transport defect. Nephron 52: Erdogan F, Duz MZ: The role of trace min-
DISCLOSURES 83– 86, 1989 erals in the pathogenesis of postmeno-
None. 18. Skyberg D, Stromme JH, Nesbakken R, Har- pausal osteoporosis and a new effect of
naes K: Neonatal hypomagnesemia with se- calcitonin. J Bone Miner Metab 20: 39–43,
lective malabsorption of magnesium: A clin- 2002
ical entity. Scand J Clin Lab Invest 21: 355– 32. Brodowski J: Levels of ionized magnesium in
REFERENCES 363, 1968 women with various stages of postmeno-
19. Stromme JH, Skyberg D, Nesbakken R: A pausal osteoporosis progression evaluated
1. Elin RJ: Magnesium: The fifth but forgotten selective intestinal malabsorption of magne- on the basis of densitometric examinations
electrolyte. Am J Clin Pathol 102: 616 – 622, sium in an infant. Scand J Clin Lab Invest [in Polish]. Przegl Lek 57: 714 –716, 2000
1994 Suppl 100: 103, 1967 33. Brunette MG, Crochet ME: Fluormetric
2. Flatman PW: Magnesium transport across 20. Milla PJ, Aggett PJ, Wolff OH, Harries JT: method for the determination of magnesium
cell membranes. J Membr Biol 80: 1–14, Studies in primary hypomagnesaemia: Evi- in renal tubular fluid. Anal Biochem 65: 79 –
1984 dence for defective carrier-mediated small 88, 1975
3. Quamme GA: Renal handling of magne- intestinal transport of magnesium. Gut 20: 34. Grimellec CL, Poujeol P, Rouffignia C: 3H-
sium: Drug and hormone interactions. Mag- 1028 –1033, 1979 inulin and electrolyte concentrations in Bow-
nesium 5: 248 –272, 1986 21. Schlingmann KP, Weber S, Peters M, Nie- man’s capsule in rat kidney: Comparison

1456 Journal of the American Society of Nephrology J Am Soc Nephrol 19: 1451–1458, 2008
www.jasn.org BRIEF REVIEW

with artificial ultrafiltration. Pflugers Arch dins in determining paracellular charge selec- 60. Cohen B, Giebisch G, Hansen LL, Teuscher
354: 117–131, 1975 tivity. Proc Am Thorac Soc 1: 38–41, 2004 U, Wiederholt M: Relationship between
35. Quamme GA: Laboratory evaluation of 48. Van Itallie CM, Anderson JM: Claudins and peritubular membrane potential and net
magnesium status: Renal function and free epithelial paracellular transport. Annu Rev fluid reabsorption in the distal renal tubule
intracellular magnesium concentration. Clin Physiol 68: 403– 429, 2006 of Amphiuma. J Physiol 348: 115–134,
Lab Med 13: 209 –223, 1993 49. Konrad M, Schaller A, Seelow D, Pandey AV, 1984
36. Sutton RA, Domrongkitchaiporn S: Abnor- Waldegger S, Lesslauer A, Vitzthum H, Su- 61. Hansen LL, Schilling AR, Wiederholt M: Ef-
mal renal magnesium handling. Miner Elec- zuki Y, Luk JM, Becker C, Schlingmann KP, fect of calcium, furosemide and chlorothia-
trolyte Metab 19: 232–240, 1993 Schmid M, Rodriguez-Soriano J, Ariceta G, zide on net volume reabsorption and baso-
37. Murayama Y, Morel F, Le Grimellec C: Phos- Cano F, Enriquez R, Juppner H, Bakkaloglu lateral membrane potential of the distal
phate, calcium and magnesium transfers in SA, Hediger MA, Gallati S, Neuhauss SC, tubule. Pflugers Arch 389: 121–126, 1981
proximal tubules and loops of Henle, as Nurnberg P, Weber S: Mutations in the 62. Grubbs RD: Intracellular magnesium and
measured by single nephron microperfusion tight-junction gene claudin 19 (CLDN19) are magnesium buffering. Biometals 15: 251–
experiments in the rat. Pflugers Arch 333: associated with renal magnesium wasting, 259, 2002
1–16, 1972 renal failure, and severe ocular involvement. 63. Romani AM, Maguire ME: Hormonal regula-
38. Quamme GA: Control of magnesium trans- Am J Hum Genet 79: 949 –957, 2006 tion of Mg2⫹ transport and homeostasis in
port in the thick ascending limb. Am J 50. Angelow S, El-Husseini R, Kanzawa SA, Yu eukaryotic cells. Biometals 15: 271–283,
Physiol 256: F197–F210, 1989 AS: Renal localization and function of the 2002
39. de Rouffignac C, Quamme G: Renal magne- tight junction protein, claudin-19. Am J 64. Hoenderop JG, Bindels RJ: Epithelial Ca2⫹
sium handling and its hormonal control. Physiol Renal Physiol 293: F166 –F177, 2007 and Mg2⫹ channels in health and disease.
Physiol Rev 74: 305–322, 1994 51. Hou J, Paul DL, Goodenough DA: Paracel- J Am Soc Nephrol 16: 15–26, 2005
40. Hoenderop JG, Nilius B, Bindels RJ: Calcium lin-1 and the modulation of ion selectivity of 65. Belge H, Gailly P, Schwaller B, Loffing J,
absorption across epithelia. Physiol Rev 85: tight junctions. J Cell Sci 118: 5109 –5118, Debaix H, Riveira-Munoz E, Beauwens R,
373– 422, 2005 2005 Devogelaer JP, Hoenderop JG, Bindels RJ,
41. Sanjad SA, Hariri A, Habbal ZM, Lifton RP: A 52. Ikari A, Hirai N, Shiroma M, Harada H, Sakai Devuyst O: Renal expression of parvalbumin
novel PCLN-1 gene mutation in familial hy- H, Hayashi H, Suzuki Y, Degawa M, Takagi K: is critical for NaCl handling and response to
pomagnesemia with hypercalciuria and Association of paracellin-1 with ZO-1 aug- diuretics. Proc Natl Acad Sci U S A 104:
atypical phenotype. Pediatr Nephrol 22: ments the reabsorption of divalent cations in 14849 –14854, 2007
503–508, 2007 renal epithelial cells. J Biol Chem 279: 66. Yang W, Lee HW, Hellinga H, Yang JJ: Struc-
42. Simon DB, Lu Y, Choate KA, Velazquez H, 54826 –54832, 2004 tural analysis, identification, and design of
Al-Sabban E, Praga M, Casari G, Bettinelli A, 53. Hou J, Renigunta A, Konrad M, Gomes AS, calcium-binding sites in proteins. Proteins
Colussi G, Rodriguez-Soriano J, McCredie Schneeberger EE, Paul DL, Waldegger S, 47: 344 –356, 2002
D, Milford D, Sanjad S, Lifton RP: Paracel- Goodenough DA: Claudin-16 and clau- 67. Montell C: Mg2⫹ homeostasis: The
lin-1, a renal tight junction protein required din-19 interact and form a cation-selective Mg2⫹nificent TRPM chanzymes. Curr Biol
for paracellular Mg2⫹ resorption. Science tight junction complex. J Clin Invest 118: 13: R799 –R801, 2003
285: 103–106, 1999 619 – 628, 2008 68. Li M, Jiang J, Yue L: Functional characteriza-
43. Weber S, Schneider L, Peters M, Misselwitz 54. Simon DB, Nelson-Williams C, Bia MJ, Elli- tion of homo- and heteromeric channel ki-
J, Ronnefarth G, Boswald M, Bonzel KE, son D, Karet FE, Molina AM, Vaara I, Iwata F, nases TRPM6 and TRPM7. J Gen Physiol
Seeman T, Sulakova T, Kuwertz-Broking E, Cushner HM, Koolen M, Gainza FJ, Gitle- 127: 525–537, 2006
Gregoric A, Palcoux JB, Tasic V, Manz F, man HJ, Lifton RP: Gitelman’s variant of Bar- 69. Chubanov V, Waldegger S, Mederos y
Scharer K, Seyberth HW, Konrad M: Novel tter’s syndrome, inherited hypokalaemic al- Schnitzler M, Vitzthum H, Sassen MC, Sey-
paracellin-1 mutations in 25 families with fa- kalosis, is caused by mutations in the berth HW, Konrad M, Gudermann T: Disrup-
milial hypomagnesemia with hypercalciuria thiazide-sensitive Na-Cl cotransporter. Nat tion of TRPM6/TRPM7 complex formation
and nephrocalcinosis. J Am Soc Nephrol 12: Genet 12: 24 –30, 1996 by a mutation in the TRPM6 gene causes
1872–1881, 2001 55. Mastroianni N, Bettinelli A, Bianchetti M, hypomagnesemia with secondary hypocal-
44. Weber S, Hoffmann K, Jeck N, Saar K, Colussi G, De Fusco M, Sereni F, Ballabio A, cemia. Proc Natl Acad Sci U S A 101: 2894 –
Boeswald M, Kuwertz-Broeking E, Meij II, Casari G: Novel molecular variants of the 2899, 2004
Knoers NV, Cochat P, Sulakova T, Bonzel Na-Cl cotransporter gene are responsible 70. Topala CN, Groenestege WT, Thebault S,
KE, Soergel M, Manz F, Schaerer K, Sey- for Gitelman syndrome. Am J Hum Genet van den Berg D, Nilius B, Hoenderop JG,
berth HW, Reis A, Konrad M: Familial hypo- 59: 1019 –1026, 1996 Bindels RJ: Molecular determinants of per-
magnesaemia with hypercalciuria and 56. Simon DB, Lifton RP: The molecular basis of meation through the cation channel TRPM6.
nephrocalcinosis maps to chromosome inherited hypokalemic alkalosis: Bartter’s Cell Calcium 41: 513–523, 2007
3q27 and is associated with mutations in the and Gitelman’s syndromes. Am J Physiol 71. Li M, Du J, Jiang J, Ratzan W, Su LT, Runnels
PCLN-1 gene. Eur J Hum Genet 8: 414 – 422, 271: F961–F966, 1996 LW, Yue L: Molecular determinants of Mg2⫹
2000 57. Moore MJ: Thiazide-induced hypomag- and Ca2⫹ permeability and pH sensitivity in
45. Umeda K, Ikenouchi J, Katahira-Tayama S, nesemia. JAMA 240: 1241, 1978 TRPM6 and TRPM7. J Biol Chem 282:
Furuse K, Sasaki H, Nakayama M, Matsui T, 58. Nijenhuis T, Vallon V, van der Kemp AW, 25817–25830, 2007
Tsukita S, Furuse M, Tsukita S: ZO-1 and Loffing J, Hoenderop JG, Bindels RJ: En- 72. Schlingmann KP, Gudermann T: A critical
ZO-2 independently determine where clau- hanced passive Ca2⫹ reabsorption and re- role of TRPM channel-kinase for human
dins are polymerized in tight-junction strand duced Mg2⫹ channel abundance explains magnesium transport. J Physiol 566: 301–
formation. Cell 126: 741–754, 2006 thiazide-induced hypocalciuria and hypo- 308, 2005
46. Balkovetz DF: Claudins at the gate: Deter- magnesemia. J Clin Invest 115: 1651–1658, 73. Chubanov V, Schlingmann KP, Waring J,
minants of renal epithelial tight junction 2005 Heinzinger J, Kaske S, Waldegger S, Schnit-
paracellular permeability. Am J Physiol Re- 59. Gunther T: Mechanisms and regulation of zler MM, Gudermann T: Hypomagnesemia
nal Physiol 290: F572–F579, 2006 Mg2⫹ efflux and Mg2⫹ influx. Miner Elec- with secondary hypocalcemia due to a mis-
47. Van Itallie CM, Anderson JM: The role of clau- trolyte Metab 19: 259 –265, 1993 sense mutation in the putative pore-forming

J Am Soc Nephrol 19: 1451–1458, 2008 Magnesium Homeostasis 1457


BRIEF REVIEW www.jasn.org

region of TRPM6. J Biol Chem 282: 7656 – 81. Pihakaski-Maunsbach K, Vorum H, Honore lactating ewes (Ovis aries). Comp Biochem
7667, 2007 B, Tokonabe S, Frokiaer J, Garty H, Karlish Physiol Comp Physiol 103: 687–693, 1992
74. Schmitz C, Perraud AL, Johnson CO, Inabe SJ, Maunsbach AB: Locations, abundances, 89. Tejpar S, Piessevaux H, Claes K, Piront P,
K, Smith MK, Penner R, Kurosaki T, Fleig A, and possible functions of FXYD ion transport Hoenderop JG, Verslype C, Van Cutsem E:
Scharenberg AM: Regulation of vertebrate regulators in rat renal medulla. Am J Physiol Magnesium wasting associated with epider-
cellular Mg2⫹ homeostasis by TRPM7. Cell Renal Physiol 291: F1033–F1044, 2006 mal-growth-factor receptor-targeting anti-
114: 191–200, 2003 82. Beguin P, Wang X, Firsov D, Puoti A, Claeys bodies in colorectal cancer: A prospective
75. Cao G, Thébault S, van der Wijst J, van der D, Horisberger JD, Geering K: The gamma study. Lancet Oncol 8: 387–394, 2007
Kemp A, Lasonder E, Bindels RJ, Hoen- subunit is a specific component of the Na,K- 90. Muallem S, Moe OW: When EGF is offside,
derop JG: RACK1 inhibits TRPM6 activity Via ATPase and modulates its transport func- magnesium is wasted. J Clin Invest 117:
phosphorylation of the fused alpha-kinase tion. EMBO J 16: 4250 – 4260, 1997 2086 –2089, 2007
domain. Curr Biol 18: 168 –176, 2008 83. Beguin P, Crambert G, Guennoun S, Garty 91. Shils ME: Experimental human magnesium
76. Meij IC, Koenderink JB, van Bokhoven H, H, Horisberger JD, Geering K: CHIF, a mem- depletion. Medicine (Baltimore) 48: 61– 85,
Assink KF, Groenestege WT, de Pont JJ, ber of the FXYD protein family, is a regulator 1969
Bindels RJ, Monnens LA, van den Heuvel LP, of Na,K-ATPase distinct from the gamma- 92. Martin HE, Jones R: The effect of ammonium
Knoers NV: Dominant isolated renal magne- subunit. EMBO J 20: 3993– 4002, 2001 chloride and sodium bicarbonate on the uri-
sium loss is caused by misrouting of the 84. Therien AG, Karlish SJ, Blostein R: Expres- nary excretion of magnesium, calcium, and
Na(⫹),K(⫹)-ATPase gamma-subunit. Nat sion and functional role of the gamma sub- phosphate. Am Heart J 62: 206 –210, 1961
Genet 26: 265–266, 2000 unit of the Na, K-ATPase in mammalian cells. 93. Wong NL, Quamme GA, Dirks JH: Effects of
77. Arystarkhova E, Wetzel RK, Asinovski NK, J Biol Chem 274: 12252–12256, 1999 acid-base disturbances on renal handling of
Sweadner KJ: The gamma subunit modulates 85. Cairo ER, Friedrich T, Swarts HG, Knoers NV, magnesium in the dog. Clin Sci (Lond) 70:
Na(⫹) and K(⫹) affinity of the renal Na,K-AT- Bindels RJ, Monnens LA, Willems PH, De 277–284, 1986
Pase. J Biol Chem 274: 33183–33185, 1999 Pont JJ, Koenderink JB: Impaired routing of 94. Nijenhuis T, Renkema KY, Hoenderop JG,
78. Jones DH, Li TY, Arystarkhova E, Barr KJ, wild type FXYD2 after oligomerisation with Bindels RJ: Acid-base status determines the
Wetzel RK, Peng J, Markham K, Sweadner FXYD2–G41R might explain the dominant renal expression of Ca2⫹ and Mg2⫹ trans-
KJ, Fong GH, Kidder GM: Na,K-ATPase nature of renal hypomagnesemia. Biochim port proteins. J Am Soc Nephrol 17: 617–
from mice lacking the gamma subunit Biophys Acta 1778: 398 – 404, 2007 626, 2006
(FXYD2) exhibits altered Na⫹ affinity and 86. Pu HX, Scanzano R, Blostein R: Distinct regu- 95. Nijenhuis T, Hoenderop JG, Bindels RJ:
decreased thermal stability. J Biol Chem latory effects of the Na,K-ATPase gamma sub- Downregulation of Ca(2⫹) and Mg(2⫹)
280: 19003–19011, 2005 unit. J Biol Chem 277: 20270–20276, 2002 transport proteins in the kidney explains ta-
79. Arystarkhova E, Wetzel RK, Sweadner KJ: 87. Groenestege WM, Thebault S, van der Wijst crolimus (FK506)-induced hypercalciuria and
Distribution and oligomeric association of J, van den Berg D, Janssen R, Tejpar S, van hypomagnesemia. J Am Soc Nephrol 15:
splice forms of Na(⫹)-K(⫹)-ATPase regula- den Heuvel LP, van Cutsem E, Hoenderop 549 –557, 2004
tory gamma-subunit in rat kidney. Am J JG, Knoers NV, Bindels RJ: Impaired baso- 96. Ikari A, Okude C, Sawada H, Takahashi T,
Physiol Renal Physiol 282: F393–F407, 2002 lateral sorting of pro-EGF causes isolated Sugatani J, Miwa M: Down-regulation of
80. Wetzel RK, Sweadner KJ: Immunocytochem- recessive renal hypomagnesemia. J Clin In- TRPM6-mediated magnesium influx by cy-
ical localization of Na-K-ATPase alpha- and vest 117: 2260 –2267, 2007 closporin A. Naunyn Schmiedebergs Arch
gamma-subunits in rat kidney. Am J Physiol 88. Gow CB, Silvapulle MJ, Moore GP: Epidermal Pharmacol November 17, 2007 [epub ahead
Renal Physiol 281: F531–F545, 2001 growth factor alters the electrolyte profile of of print]

1458 Journal of the American Society of Nephrology J Am Soc Nephrol 19: 1451–1458, 2008

Das könnte Ihnen auch gefallen