You are on page 1of 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/221695400

Drug-induced alterations in Mg homoeostasis

Article  in  Clinical Science · July 2012


DOI: 10.1042/CS20120045 · Source: PubMed

CITATIONS READS

56 723

4 authors, including:

Anke L L Lameris Leo L A Monnens


Radboud University Medical Centre (Radboudumc) Radboud University Medical Centre (Radboudumc)
12 PUBLICATIONS   438 CITATIONS    505 PUBLICATIONS   14,011 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Anke L L Lameris on 19 January 2015.

The user has requested enhancement of the downloaded file.


www.clinsci.org
Clinical Science (2012) 123, 1–14 (Printed in Great Britain) doi:10.1042/CS20120045 1

R E V I E W

Drug-induced alterations in
Mg2 + homoeostasis

Anke L. LAMERIS, Leo A. MONNENS, René J. BINDELS and Joost G. J. HOENDEROP


Department of Physiology, Radboud University Nijmegen Medical Centre, 6500 HB Nijmegen, The Netherlands

A B S T R A C T

Clinical Science
Magnesium (Mg2 + ) balance is tightly regulated by the concerted actions of the intestine, bone
and kidneys. This balance can be disturbed by a broad variety of drugs. Diuretics, modulators of
the EGFR (epidermal growth factor receptor), proton pump inhibitors, antimicrobials, calcineurin
inhibitors and cytostatics may all cause hypomagnesaemia, potentially leading to tetany, seizures and
cardiac arrhythmias. Conversely, high doses of Mg2 + salts, frequently administered as an antacid
or a laxative, may lead to hypermagnesaemia causing various cardiovascular and neuromuscular
abnormalities. A better understanding of the molecular mechanisms underlying the adverse effects
of these medications on Mg2 + balance will indicate ways of prevention and treatment of these
adverse effects and could potentially provide more insight into Mg2 + homoeostasis.

INTRODUCTION MAGNESIUM HOMOEOSTASIS


Mg2 + , the second most abundant intracellular divalent Mg2 + transport can take place via two routes: an
cation, is of central importance for a broad variety of active transcellular pathway or a passive paracellular
physiological processes, including intracellular signalling, route. The transcellular transport of Mg2 + is less well
neuronal excitability, muscle contraction, bone formation defined than that of for instance Ca2 + . TRPM (transient
and enzyme activation. Its overall balance is tightly receptor potential melastatin) 6 and 7 cation channels
regulated by the concerted actions of the intestine, are thought to be responsible for Mg2 + transport into
bones and kidneys, keeping plasma Mg2 + levels within the cell. TRPM6 is most abundantly expressed in the
the range of 0.70 to 1.10 mmol/l. Disturbance of this intestine, kidney and lung, whereas TRPM7 is expressed
balance can have serious consequences. Symptoms of ubiquitously [1]. Transport of Mg2 + from the tubular or
hypomagnesaemia include tetany, seizures and cardiac intestinal lumen into the cell is, therefore, considered to
arrhythmias, whereas hypermagnesaemia may cause be mediated predominantly via TRPM6, whereas TRPM7
various cardiovascular and neuromuscular abnormalities. appears responsible for cellular Mg2 + homoeostasis.
Drugs can interfere with Mg2 + homoeostasis in multiple A role for TRPM7 and even for TRPM6–TRPM7
ways. The aim of the present review is to provide an heterotetrameric complexes in the apical transport of
overview of the various drugs that are known to influence Mg2 + has, however, also been suggested [2,3]. A recent
Mg2 + homoeostasis and to give some insights into the study by Ryazanova et al. [4] has shown that heterozygote
underlying physiological mechanisms. TRPM7-deficient mice develop hypomagnesaemia due

Key words: drug induction, hypermagnesaemia, hypomagnesaemia, ion channel, kidney, magnesium.
Abbreviations: AGA, aminoglycoside antibiotic; CaSR, Ca2 + -sensing receptor; CLC-Kb, Cl − channel Kb; CsA, cyclosporin A;
DCT, distal convoluted tubule; EGF, epidermal growth factor; EGFR, EGF receptor; NCC, Na + –Cl − co-transporter; NKCC2,
Na + –K + –2Cl − co-transporter; OMIM, Online Mendelian Inheritance in Man; PPI, proton pump inhibitor; PT, proximal tubulus;
PTH, parathyroid hormone; ROMK, renal outer medullary K + channel; TAL, thick ascending limb; TRPM, transient receptor
potential melastatin.
Correspondence: Professor Joost G.J. Hoenderop (email j.hoenderop@fysiol.umcn.nl).


C The Authors Journal compilation 
C 2012 Biochemical Society
2 A. L. Lameris and others

to a defect in intestinal absorption of Mg2 + , whereas Kidney


renal Mg2 + excretion is appropriately low, indicating In the kidney, approximately 2.5 g of Mg2 + is filtered by
TRPM7 does play an important role in intestinal Mg2 + the glomeruli on a daily basis. Approximately 95 % of this
homoeostasis. Specific Mg2 + -binding proteins involved filtered load is reabsorbed along the different parts of the
in cytosolic diffusion have not been identified to date. The nephron via both paracellular and transcellular processes
calbindin proteins, primarily responsible for cytosolic (Figure 1) [14]. In the PT (proximal tubulus), 10–30 %
Ca2 + transport, have been shown to also bind Mg2 + , but is reabsorbed [15,16]. Generally, this occurs via a para-
the physiological relevance of this observation remains cellular process, most likely depending on Na + -driven
to be determined [5]. The molecular identity of the water transport that increases the intraluminal Mg2 +
basolateral extrusion mechanism for Mg2 + remains to be concentration of the pro-urine along its way through the
defined as well. Extrusion of Mg2 + must occur against an PT; the small lumen-positive potential in the PT also adds
electrochemical gradient, implicating its dependence on driving force [17–19]. The exact mechanism, however,
a primary or secondary active transport process, a Mg2 + remains to be elucidated. Paracellular Mg2 + transport in
pump or a Na + /Mg2 + exchanger are most frequently the nephron predominates in the TAL (thick ascending
suggested [6,7]. Alternatively, Mg2 + can be absorbed limb) of the nephron, where 40–70 % of the filtered Mg2 +
passively via paracellular transport route. For this latter is reabsorbed [20]. Here, Mg2 + reabsorption is mainly
process, tight-junction proteins from the claudin family driven by the lumen-positive transepithelial voltage.
are important, as they can determine both size and charge NKCC2 (Na + –K + –2Cl − co-transporter) serves as an
selectivity of the paracellular transport [8]. apical entry mechanism for Na + , K + and Cl − . ROMK
(renal outer medullary K + channel) 2 allows for apical
Intestine recycling of K + back into the tubular lumen, thereby
In the intestine, Mg2 + is primarily derived from dietary generating a lumen-positive voltage of approximately
sources, such as green leafy vegetables, nuts and whole 5–10 mV. Cl − exits via the basolateral membrane via
grains. Approximately 300 mg of Mg2 + is ingested CLC-Kb (Cl − channel Kb). At the basolateral side, the
on a daily basis, of which 25–75 % is absorbed in Na + /K + -ATPase constitutes the initial driving force
the adult, depending on the availability and needs for Na + transport in the TAL. Here, the paracellular
of the body [6,9]. When Mg2 + intake is normal, the Mg2 + reabsorption depends on the lumen-positive
transcellular pathway, involving TRPM6, is responsible transepithelial voltage and is facilitated by claudin-16 and
for approximately 30 % of absorption, a fraction that -19, which are two tight-junction proteins that operate
increases when dietary intake is low [10]. Paracellular together to form a cation-permeable channel [21]. At the
transport depends on the transepithelial electrical voltage, basolateral side, CaSR (Ca2 + -sensing receptor) can mod-
which is approximately 5 mV lumen-positive with respect ulate transport in response to changes in concentration of
to blood. Luminal Mg2 + concentrations range from extracellular cations, reducing transport upon stimulation
1 to 5 mmol/l depending on dietary contents, providing [22]. In the DCT (distal convoluted tubule) the final 5–
a transepithelial chemical concentration gradient which 10 % of Mg2 + is reabsorbed in an active transcellular
further enhances absorption [10]. Paracellular transport manner [23]. The tubular epithelium in this part of the
takes place via tight junctions and is responsible for nephron consists of a high-resistance epithelium with a
approximately 70 % of Mg2 + absorption when luminal lumen-negative voltage of approximately − 5 mV. Apical
concentrations are high. For the intestine, Mg2 + -specific reabsorption occurs via TRPM6, which can be stimulated
paracellular channels have not yet been found; however, via the basolateral EGFR [EGF (epidermal growth factor)
the observation that, for instance, claudin-2 and -12 are receptor] [24]. Kv1.1, an apically located K + channel,
vitamin D-regulated indicates that it is likely that a similar establishes a favourable luminal membrane potential
pathway is involved in intestinal paracellular transport of facilitating an increase in the driving force for Mg2 +
divalent cations [11]. reabsorption via TRPM6 [25]. The Na + /K + -ATPase at
the basolateral membrane provides the Na + gradient that
Bone is utilized by the apical NCC (Na + –Cl − co-transporter)
Approximately 50 % of the total Mg2 + body content to transport NaCl into the cell, while K + that enters at
resides within the skeleton as part of hydroxyapatite the basolateral side in this process is recycled via Kir4.1.
crystals. The stores in bone serve as a buffer from which The Mg2 + reabsorption in the DCT defines the final
Mg2 + can be released when plasma levels are low and urinary Mg2 + excretion, as there is no significant
into which Mg2 + can be stored when circulating Mg2 + reabsorption of Mg2 + beyond the DCT.
levels are excessive [12]. In animals maintained on a low-
Mg2 + diet, bone Mg2 + content is reduced, as is bone
HYPOMAGNESAEMIA
mineral density [13]. To date, our understanding of the
mechanisms responsible for the mobilization of Mg2 + Hypomagnesaemia is defined as a serum Mg2 +
into and out of these skeletal stores remains insufficient. concentration of less than 0.70 mmol/l. Muscle cramps,


C The Authors Journal compilation 
C 2012 Biochemical Society
Drug-induced alterations in Mg2 + homoeostasis 3

Figure 1 Schematic representation of the nephron and electrolyte handling in the TAL of Henlē and DCT
(A) Approximately 95 % of the Mg2 + filtered in the glomerulus is reabsorbed from the pro-urine along the different segments of the nephron. Of this filtered Mg2+
load, 10–30 % is reabsorbed in the PT, followed by an additional 40–70 % in the TAL. The DCT is responsible for the transport of the final 5–10 % of Mg2 + back
into the blood. CNT, connecting tubule; TDL, thin desending limb. (B) In the TAL, NKCC2 allows apical entry of Na + , K + and Cl − . ROMK2 recycles K + back into
the tubular lumen. Cl − exits via the basolateral membrane via CLC-Kb. At the basolateral side, the Na + /K + -ATPase constitutes the initial driving force for Na +
transport in the TAL. Paracellular Mg2 + absorption is facilitated by claudin-16 and claudin-19 (CLDN16/19). CaSR will reduce the absorption upon stimulation by
extracellular cations. (C) In the DCT, Mg2 + absorption via TRPM6 depends on the membrane potential, which is set by Kv1.1 and can be stimulated via EGFR. The
Na + /K + -ATPase at the basolateral membrane provides an Na + gradient that is used by NCC on the apical cell membrane. K + that enters the cell in this process
is recycled via Kir4.1.

tetany and muscular weakness are the main complaints of hypomagnesaemia [27–29]. In isolated autosomal-
of hypomagnesaemic patients [26]. Cardiovascular dominant hypomagnesaemia due to mutations in KCNA1
abnormalities, such as arrhythmias and convulsions, have (encoding Kv1.1), serum Mg2 + levels <0.40 mmol/l
also been described. Hypomagnesaemia is, however, were found in an index patient suffering from recurrent
often associated with multiple biochemical abnormalities muscle cramps, tetanic episodes, tremor and muscular
such as hypokalaemia and hypocalcaemia. It is, therefore, weakness. Serum Mg2 + values of 0.37 and 0.25 mmol/l
difficult to ascribe specific clinical manifestations solely with normal serum Ca2 + and K + values were reported
to hypomagnesaemia. In this regard, important lessons in affected family members during severe attacks of
can be learned from patients with isolated forms of cramps and tetany [25]. In another form of dominant
hypomagnesaemia, such as isolated dominant [OMIM hypomagnesaemia caused by mutations in the CNNM2
(Online Mendelian Inheritance in Man) #154020, gene (encoding cyclin M2), the index patient had a
#176260 and #613882] and isolated recessive (OMIM serum Mg2 + level of 0.36 mmol/l and suffered from
#611718) hypomagnesaemia. It must, however, be noted weakness of the limbs, vertigo and headaches, but no
that these cases reflect only the chronically Mg2 + other electrolyte abnormalities were described [30,31].
-depleted state. Recessive hypomagnesaemia resulting from mutations in
Isolated dominant hypomagnesaemia can be caused the EGF gene also leads to isolated hypomagnesaemia.
by mutations in the FXYD2 gene, encoding the γ - Two sisters were described with generalized seizures
subunit of the Na + /K + -ATPase. The two index patients during infancy, and were found to have serum Mg2 +
described, with serum Mg2 + levels of approximately 0.4 levels of 0.53–0.66 mmol/l with no other clear clinical
mmol/l, suffered from generalized convulsions and severe symptoms [32].
mental retardation was present in one of the patients. Chronic depletion of Mg2 + , as reflected by the
Remarkably, other family members with low serum low serum Mg2 + levels, generally becomes clinically
Mg2 + levels (0.32–0.65 mmol/l) showed no symptoms evident in patients with serum Mg2 + concentrations


C The Authors Journal compilation 
C 2012 Biochemical Society
4 A. L. Lameris and others

of <0.40 mmol/l. As mentioned above, hypocalcaemia animal models they can be useful and are, therefore,
(muscle cramps and tetany) and hypokalaemia (muscle frequently applied. The use of radioactive Mg2 + isotopes
weakness), when occurring secondary to hypomag- for balance studies is unfortunately limited by the short
nesaemia, will contribute to the clinical phenotype. Mg2 + isotope half-lives, which range from 21 h to just
Hypokalaemia associated with hypomagnesaemia is milliseconds. Stable Mg2 + isotopes such as 25 Mg or 26 Mg
often refractory to treatment by K + and can only can be distinguished from 24 Mg using inductively coupled
be properly corrected when Mg2 + is co-administered. plasma mass spectrometry. Owing to their relatively high
The decreased intracellular Mg2 + concentration resulting natural abundance, high doses of the enriched isotopes
from Mg2 + deficiency, releases the Mg2 + -mediated are required for analytical precision, making this type of
inhibition of ROMKs, thereby increasing K + excretion experiments not only technically demanding, but also
[33]. Hypocalcaemia secondary to hypomagnesaemia has relatively expensive [39].
been hypothesized to originate from inappropriately low In conclusion, a simple, rapid and accurate test
PTH (parathyroid hormone) secretion, the release of to assess total body Mg2 + status is lacking. Until
which depends on an intracellular signalling cascade that adequate alternatives become available, the combined
requires Mg2 + . In addition, the responsiveness of target determination of serum Mg2 + and urinary Mg2 +
organs to PTH stimulation seems to be reduced [34,35]. excretion remains the only practical test to assess
Hypocalcaemia secondary to hypomagnesaemia with low Mg2 + status.
PTH secretion is mostly seen when serum Mg2 + levels
are extremely low [36]. Treatment is only successful
when hypomagnesaemia is addressed together with the
Treatment of hypomagnesaemia
The route of administration of Mg2 + supplements
hypocalcaemia, as Ca2 + supplementation alone will not
depends on the severity of clinical findings and the origin
suffice [34].
of the hypomagnesaemia. Patients can be treated with
oral supplements, with intramuscular injections or
Assessment of Mg2 + status with intravenous infusions [40]. Intravenous adminis-
In clinical practice total serum Mg2 + is most commonly
tration is the most effective in restoring serum Mg2 +
used to assess the Mg2 + status in patients. This
to normal levels and is, therefore, the best treatment in
parameter does not necessarily reflect the true total
cases of severe hypomagnesaemia. The disadvantage is
body Mg2 + content, as normal serum Mg2 + levels
that patients have to visit the hospital on a regular basis if
may be present despite intracellular depletion. Using
frequent treatment is required. Oral supplements do not
ion-selective electrodes it is possible to measure solely
require hospital visits; however, not all patients tolerate
ionized Mg2 + . Some studies indicate that ionized Mg2 +
oral supplementation of Mg2 + due to their cathartic
is a better representative of the Mg2 + status compared
effect at higher dosages. Solubility and bioavailability
with total serum Mg2 + , whereas others find both
vary greatly between various Mg2 + supplements. In
parameters equally representative. The Mg2 + content
general, organic Mg2 + salts have a higher solubility
of erythrocytes, leucocytes and platelets has also been
and bioavailability compared with inorganic Mg2 + salts,
suggested as potential biomarkers for the assessment of
making them more suitable for oral Mg2 + replacement
Mg2 + status. However, their usefulness and reliability are
therapy. It should be noted that, in cases of severe
currently under debate. Alternatives such as measuring
hypomagnesaemia or Mg2 + deficiency due to intestinal
Mg2 + content of bone, teeth, muscle, hair or nails are
malabsorption, oral Mg2 + supplements may not be
either too invasive, too laborious or not reliable to use in
sufficient to restore serum Mg2 + levels to normal.
daily clinical practice [37]. An easy non-invasive method
is to analyse urinary Mg2 + excretion. As renal Mg2 +
excretion will decrease in response to deficiency, this is
an important parameter during assessment of Mg2 + status DRUG-INDUCED HYPOMAGNESAEMIA
[37].
A summary of the drugs described below is provided in
The Mg2 + status can also be evaluated in balance
Table 1 and Figure 2.
studies by determining Mg2 + faecal as well as renal
excretion after oral loading or intravenous infusion.
Since the results of these tests are partly dependent Diuretics
on the renal function, the risk of false-negative and Diuretics increase urinary output and are widely used in
false-positive outcomes should be taken into account. the treatment of hypertension, heart failure and kidney
Owing to these and other limitations, balance studies diseases. They may cause hypermagnesuria, leading
are not frequently performed [38]. For some minerals, to possible hypomagnesaemia and Mg2 + depletion.
radioactive isotopes can be used to assess the mineral Three types of diuretics are known to influence Mg2 +
balance. For safety reasons radioactive isotopes are not homoeostasis, namely osmotic diuretics, loop diuretics
often employed for balance studies in humans, but in and thiazide-type diuretics.


C The Authors Journal compilation 
C 2012 Biochemical Society
Drug-induced alterations in Mg2 + homoeostasis 5

Table 1 Drug-induced changes in Mg2 + homoeostasis

Hypomagnesaemia Hypermagnesaemia

Drug/compound Reference(s) Drug/compound References

Diuretics Epsom salt poisoning [103,106–108]


Osmotic diuretics [41] Overdose using Mg2 + as a cathartic [108,110,111]
Loop diuretics [41,42] Antacids [112–115]
Thiazide-type diuretics [43–49] Laxative abuse [119,121,122]
EGFR modulators Enemas containing Mg2 + [107,123,124]
Cetuximab [32,50–55]
Erlotinib [56]
PPIs [57–65]
Antimicrobials
AGAs [66–73]
Pentamidine [74–78]
Sirolimus [52,79,80]
Amphotericin B [81,82]
Foscarnet [83,84b]
Calcineurin inhibitors
CsA [86–91]
FK506 [92–95]
Cisplatin/carboplatin [96–101]

Figure 2 Overview of drugs causing hypomagnesaemia


Overview of the six classes of drugs described causing hypomagnesaemia. Effects of individual drugs on key players in Mg2 + homoeostasis in cells of the TAL and
DCT are displayed. Pentamidine and cisplatin are not know to have an effect on specific proteins involved in Mg2 + homoeostasis, but have been shown to be toxic
to/accumulate in DCT cells respectively.


C The Authors Journal compilation 
C 2012 Biochemical Society
6 A. L. Lameris and others

Osmotic diuretics inappropriately high fractional excretion of Mg2 + . They


Osmotic diuretics (such as mannitol) are solutes that are identified a mutation in the pro-EGF gene that, upon
not absorbed or only partly absorbed from the renal closer investigation, was found to cause a defect in the
tubular lumen. They diminish salt and water reabsorption sorting of the pro-EGF, resulting in impaired basolateral
by increasing the flow rate thereby reducing, among sorting. Owing to the resulting lack of release of the
others, serum Mg2 + levels [41]. hormone on the basolateral side, the EGFR in the
DCT could not be stimulated. This study demonstrated
Loop diuretics that TRPM6 activity was increased upon binding of
Loop diuretics (such as furosemide, bumetanide, EGF to EGFR, making EGF the first magnesiotropic
torsemide and ethacrynic acid) act in the TAL by hormone. The increased activity results from an
competing for the Cl − site on NKCC2. Inhibition increase in the mobility and cell-surface expression
of apical Cl − reabsorption and diminished basolateral of TRPM6 [52]. These findings matched perfectly
Cl − efflux lead to a loss of lumen-positive potential, with earlier observations that certain EGFR-targeting
thus resulting in a diminished driving force for chemotherapeutic agents cause hypomagnesaemia. There
paracellular cation reabsorption [42]. Therefore the use are actually two main classes of anticancer agents that
of loop diuretics may lead to renal Mg2 + wasting and affect the EGFR: those targeting the extracellular ligand-
hypomagnesaemia due to reduced paracellular Mg2 + binding domain (such as cetuximab and panitumumab)
reabsorption via claudin-16 and -19 [41]. and drugs that block the intracellular tyrosine kinase
domain (such as erlotinib and gefitinib).
Thiazide-type diuretics
Thiazide-type diuretics comprise a class of diuretics de- Cetuximab (erbitux)
rived from benzothiadiazine, but also drugs with similar Cetuximab is a chimaeric (mouse/human) monoclonal
action that do not have the thiazide chemical structure antibody, which functions as an EGFR antagonist by
such as chlortalidone and metolazone. Thiazides enhance preventing the binding of EGF and triggering the
renal Na + excretion by inhibiting NCC in the DCT cells internalization of the receptor, consequently leading
[43]. Thiazide treatment leads to hypomagnesaemia as to inhibition of EGFR activation. Approximately
well as hypocalciuria [44,45]. In contrast with the effects 1.5 years after its release by the U.S. FDA (Food
of thiazide-type diuretics on Ca2 + balance, which seem and Drug Administration) in 2004, a warning was
to differ in short- and long-term treatment, the effect of released concerning the relationship between severe
thiazides on Mg2 + reabsorption only becomes apparent hypomagnesaemia and cetuximab treatment. Several
upon chronic treatment [46]. Hypomagnesaemia has been groups have performed retrospective studies to determine
suggested to result from K + deficiency, increased passive the incidence of cetuximab-induced hypomagnesaemia
Mg2 + secretion or decreased active Mg2 + reabsorption [53,54]. These studies are, however, hindered by the
in the DCT [47–49]. Chronic administration of thiazides fact that Mg2 + levels are not commonly determined
enhances renal Mg2 + excretion and specifically reduced in all patients. In a prospective study, it was shown
renal expression levels of TRPM6 in a mouse model. In that in most patients serum Mg2 + concentrations during
the same study, it was shown that TRPM6 expression EGFR-targeting treatment had decreased compared with
levels were severely decreased in NCC-knockout mice. baseline measurements [55]. In total, 54 % of the treated
Therefore the pathogenesis of hypomagnesaemia in patients developed hypomagnesaemia (defined as serum
chronic thiazide treatment appears to result from specific Mg2 + levels <0.65 mmol/l), and in 6 % of all patients
TRPM6 down-regulation in the DCT [46]. hypomagnesaemia was severe (serum Mg2 + levels <0.4
mmol/l). Clinically significant hypocalcaemia was rare
EGFR modulators and not progressive upon treatment. Careful monitoring
EGF is an important growth factor that is synthesized of serum Mg2 + levels is, therefore, essential during
as a pre-proprotein, which is cleaved and subsequently treatment.
processed into the mature EGF [50–51a]. EGF acts
by binding with high affinity to EGFR, which is Erlotinib (tarceva)
generally localized at the basolateral cell surface, thereby Erlotinib is a tyrosine kinase inhibitor that acts
stimulating the intrinsic protein tyrosine kinase activity on EGFR. It binds in a reversible fashion to the
of the receptor and activating intracellular signalling ATP-binding site of the receptor. For the signal to
cascades. EGFR is overexpressed in a broad variety of be transmitted, two members of the EGFR family
tumour cells, making it an effective target for anticancer need to dimerize. The two halves of the dimerized
treatments. In the nephron, EGFR is expressed along the EGFR then autophosphorylate each other using one
TAL and DCT [32]. ATP molecule. The autophosphorylation causes a
In 2007, Groenestege et al. [32] presented two conformational change in the intracellular structure of
sisters with low serum Mg2 + concentrations and an EGFR, which is essential for the activation of the


C The Authors Journal compilation 
C 2012 Biochemical Society
Drug-induced alterations in Mg2 + homoeostasis 7

intracellular signalling cascade that takes place upon Several explanations have been suggested in the pub-
binding of EGF to EGFR. By inhibiting the binding lished case reports. First, Mg2 + absorption might depend
of ATP, autophosphorylation is not possible and the on the stomach’s acidity for solubilization. Achlorhydria
signal is blocked. To date, reports on erlotinib-induced caused by PPIs could, therefore, reduce Mg2 +
hypomagnesaemia in patients are lacking. A recent study availability for absorption, resulting in diminished Mg2 +
by Dimke et al. [56] determined the effect of erlotinib on absorbed in the intestine. A second explanation would
Mg2 + homoeostasis. In vivo studies showed a small, but be an increase in intestinal Mg2 + loss via secretion;
significant, effect on serum Mg2 + levels; furthermore, considerable amounts of Mg2 + can be excreted in the
mice failed to decrease renal fractional excretion of intestine, and thus a shift in the balance between Mg2 +
Mg2 + in reaction to reduced Mg2 + levels. Although absorption and secretion in the intestine could result in
TRPM6 expression was lowered at the mRNA level, no net Mg2 + loss. Thirdly, active transcellular or passive
changes were found at the protein level. Erlotinib failed paracellular Mg2 + absorption could be impaired. TRPM6
to inhibit EGF-induced changes in TRPM6 activity at activity is positively regulated by extracellular protons,
physiological concentrations [56]. Taken together, these and an inhibitory effect of PPIs on the colonic H + /K + -
observations indicate that the effect of erlotinib on Mg2 + ATPase (a homologue of the gastric H + /K + -ATPase that
homoeostasis is minimal. Nevertheless, in the case of is inhibited by PPIs) could reduce TRPM6 activity due to
erlotinib treatment in patients predisposed to developing a decreased level of excreted protons. Recently, Thongon
hypomagnesaemia, extra caution is warranted. and Krishnamra [65] have shown that omeprazole
The dissimilarity between anti-EGFR antibodies and treatment of Caco-2 monolayers can decrease paracellular
EGFR tyrosine kinase inhibitors on Mg2 + handling is cation permeability leading to suppression of passive
still not fully understood; most probably differences Mg2 + absorption. A clear molecular explanation for this
in the bioavailability of erlotinib and cetuximab play observation is still lacking.
a decisive role in the aetiology of EGFR modulator- More studies need to be performed to determine
induced hypomagnesaemia [56]. the incidence of PPI-induced hypomagnesaemia and
to unravel the underlying mechanisms. Meanwhile,
PPIs (proton pump inhibitors) physicians should be aware of this serious side effect
PPIs belong to the most widely prescribed classes of of PPIs. Patients suffering from hypomagnesaemia due
drugs in the world. In 2006, two cases of PPI-induced to PPIs can be treated with supplements or switched to a
hypomagnesaemia were published by Epstein et al. [57]. histamine H2 receptor antagonist.
Since then, several groups have described similar findings
in patients using PPIs [58–62]. The exact prevalence Antimicrobials
remains unknown; however, the growing number of case
reports indicates that these patients might represent the AGAs (aminoglycoside antibiotics)
‘tip of an iceberg’. AGAs such as gentamycin, tobramycin and amikacin are
Patients generally suffer from hypomagnesaemia and widely used in the treatment of severe bacterial infections
often have secondary hypocalcaemia and hypokalaemia. of the abdomen and urinary tract because of their high
Long-term use seems to play a role, as all patients efficacy. Side effects can, however, be serious, with
were using PPIs for more then 1 year. Remarkably, nephrotoxicity and ototoxicity occurring in up to 35 %
patients recover relatively quickly upon discontinuation of the treated patients. As many as 25 % of the patients
and relapse within days when medication is restarted. will develop hypomagnesaemia due to renal Mg2 + loss
This could indicate that PPIs might have a relatively [66]. The effect of AGAs on Mg2 + balance is not directly
subtle effect on Mg2 + homoeostasis, which only becomes dose-dependent, but seems to be related to the cumulative
overt when Mg2 + stores are completely depleted. doses of AGAs received by the patient during treatment.
Indeed, Mg2 + retention upon intravenous infusion is Onset of hypomagnesaemia can take up to 2 weeks and
high, suggesting severe depletion of the body’s Mg2 + can persist after cessation of AGA treatment for several
stores. Urinary Mg2 + excretion is low, indicating intact months [67]. Hypermagnesuria is generally accompanied
renal function. Taken together, these findings point by hypercalciuria, which suggests that AGAs exert an
towards an intestinal abnormality as the most likely effect on renal solute transport in those tubular segments
problem underlying PPI-induced hypomagnesaemia. where both Ca2 + and Mg2 + are reabsorbed, i.e. the TAL
Studies examining the effect of PPIs on the intestinal and DCT. AGAs do not accumulate in the more distal
absorption of various salts have not identified differences parts of the nephron, but up to 10 % of the administered
in the absorption of Mg2 + [63,64]. All studies were, dose accumulates in the renal cortex, especially in the
however, performed after short-term PPI use and were PT cells, where intracellular accumulation of AGAs
based on faecal excretion analysis; a long-term effect or leads to damage as indicated by the increase in specific
a subtle effect for which this technique is not sensitive biomarkers [68,69]. Animal experiments have shown
enough and can, therefore, not be excluded. renal Mg2 + wasting in the absence of renal failure


C The Authors Journal compilation 
C 2012 Biochemical Society
8 A. L. Lameris and others

or tubular morphology abnormalities [70]. In a recent in TRPM6 expression by reducing mRNA stability.
study, Tzovaras et al. [71] found no correlation between Clinical implications of these findings remain to be
changes in Mg2 + and Ca2 + excretion with alterations in determined; however, they indicate hypomagnesaemia
urine lactate, alanine or hippurate used as markers for due to rapamycin treatment is the result of a defect in
nephrotoxicity. Together, these findings indicate that the Mg2 + absorption by DCT cells.
proximal and distal renal tubular dysfunction as a result of
AGA treatment is caused by two independent processes. Amphotericin B
AGAs can function as polyvalent cations and can thus Amphotericin B is an antifungal drug that is mainly
stimulate the CaSR [72,73]. Activation of the CaSR by used to treat various systemic fungal infections in
polyvalent cations could inhibit the passive reabsorption immunocompromised patients or for parasitic protozoan
of Ca2 + and Mg2 + in the TAL and active absorption infections. Amphotericin B causes renal injury leading
in the DCT. Indeed, Sassen et al. [70] have shown to renal insufficiency, urinary K + wasting and
a decreased expression of NKCC2 and an increased hypokalaemia, Mg2 + wasting and hypomagnesaemia,
expression of CaSR in the TAL of gentamycin-treated metabolic acidaemia due to distal renal tubular acidosis,
rats. The reduced NKCC2 expression would result in a and polyuria [81]. Barton et al. [82] found reduced serum
decrease in the lumen-positive membrane voltage and is, Mg2 + levels and an increase in renal Mg2 + excretion upon
therefore, likely to contribute to the increased urinary treatment with amphotericin B. They also showed that
Ca2 + and Mg2 + excretion resembling the response to the effect is reversible and Mg2 + homoeostasis returns to
furosemide treatment. normal baseline levels upon discontinuation of treatment
[82]. To date, no molecular data explaining amphotericin
Pentamidine B-induced hypomagnesaemia are available.
Pentamidine is an antimicrobial used in the prevention
or treatment of pneumocystic pneumonia in immuno- Foscarnet (trisodium phosphonoformate hexahydrate)
compromised patients. Treatment with pentamidine can Foscarnet is a pyrophosphate analogue that inhibits many
lead to severe symptomatic hypomagnesaemia secondary viral DNA polymerases [83]. It is used to treat cytomeg-
to renal Mg2 + wasting, typically in association with alovirus and acyclovir-resistant mucocutaneous herpes
hypocalcaemia, which has been observed to continue simplex virus disease in immunocompromised patients.
for up to 2 months after cessation of pentamidine Its side effects include hypomagnesaemia, hypocalcaemia
treatment [74–76]. The exact molecular mechanism and hypokalaemia [84]. Being a pyrophosphate analogue,
underlying this renal Mg2 + leakage is unknown; however, foscarnet is a potent chelator of divalent cations. Thus
autopsy studies in AIDS patients has shown pentamidine complexes readily form with Mg2 + and Ca2 + , and a
accumulation in renal tissue from 1 day to 1 year after linear relationship exists between serum divalent ions and
the completion of therapy [77,78]. In addition, the circulating plasma foscarnet concentrations [84a,84b].
epithelium of the DCT was degenerated, suggesting that This suggests that foscarnet-induced hypomagnesaemia
pentamidine-induced hypomagnesaemia might be the is solely the result of the physical interaction between
result of diminished active Mg2 + reabsorption in foscarnet and Mg2 + . It would be interesting, however,
the kidney. to investigate whether adaptational changes occur in
expression of, for example, TRPM6.
Rapamycin (sirolimus)
Rapamycin is a macrolide antibiotic derived from fungus. Calcineurin inhibitors
It was the first inhibitor of the mTOR (mammalian Calcineurin inhibitors such as CsA (cyclosporin A;
target of rapamycin) pathway approved by the FDA ciclosporin) and FK506 are immunosuppressant drugs
and it is frequently used to prevent post-transplant that are widely prescribed in post-transplantation
organ rejection. Treatment with rapamycin is associated immunosuppression and for numerous immunological
with increased renal Mg2 + excretion resulting in disorders. Calcineurin inhibitors are known to cause
hypomagnesaemia [79]. da Silva et al. [80] showed a renal injury, hypertension and tubular dysfunction
marked decrease in both plasma K + and Mg2 + in with disturbances of mineral metabolism. Both drugs
sirolimus-treated rats. In addition, increased urinary commonly lead to hypomagnesaemia due to renal Mg2 +
excretion of Na + , K + and Mg2 + was found, whereas the wasting, hypercalciuria and hypokalaemia [85].
glomerular filtration rate remained unchanged. Sirolimus
treatment reduced the expression of NKCC2 and AQP2 CsA
(aquaporin 2), whereas TRPM6 expression was increased, CsA is known to cause nephrotoxicity in a dose-
which could represent either a direct stimulatory effect dependent and reversible manner [86–88]. CsA treatment
of sirolimus or a compensatory response [80]. Recently, also frequently results in a disruption of mineral
Ikari et al. [52] demonstrated in an in vitro study homoeostasis, with 10–36 % of CsA-treated patients
that rapamycin inhibits the EGF-mediated increase developing hypomagnesaemia [88,89]. In vitro, CsA


C The Authors Journal compilation 
C 2012 Biochemical Society
Drug-induced alterations in Mg2 + homoeostasis 9

significantly reduces the expression of claudin-16 and Table 2 Symptoms of hypermagnesaemia


TRPM6, but not TRPM7, at both the mRNA and protein
Serum Mg2 + level (mmol/l) Symptoms
levels; for TRPM6, the decreased expression is mediated
by the inhibition of c-Fos transcription [89,90]. A recent 0.7–1.1 None; normal serum level
study has described an increase in the fractional excretion 1.1–2.0 Asymptomatic
of Mg2 + in rats as a result of CsA treatment [91]. 2.0–3.0 Nausea, vomiting, cutanous vasodilation,
The study also showed a reduction in the expression headaches, hyporeflexia and lethargy
levels of TRPM6, TRPM7, NCC and EGF, whereas 3.0–5.0 Unresponsiveness, loss of deep tendon
reduced expression of EGFR or claudin-16 was not found reflexes, electrocardiographic changes,
[91]. Together, these findings indicate CsA reduces renal such as prolongation of QRS, PR and QT
Mg2 + reabsorption in the DCT, possibly via the down- intervals, bradycardia and hypotension
regulation of TRPM6. >5.0 Complete heart block, respiratory paralysis,
coma and shock
FK506 (tacrolimus) >8.0 Asystole and death
FK506 treatment is frequently associated with an
inappropriately high fractional excretion of Mg2 + and
Ca2 + [92]. Nijenhuis et al. [93] found decreased renal enhance each other in a vicious circle in which cisplatin
levels of TRPV5 (transient receptor potential vanilloid treatment causes renal Mg2 + loss and the resulting
5), calbindin-D28K and TRPM6 mRNA expression in hypomagnesaemia stimulates cisplatin accumulation in
the kidneys of rats treated with FK506. The expression the kidney [101].
of specific DCT markers was unaltered, excluding the
possibility that the reduction in Mg2 + reabsorption is
the result of a general toxic effect of FK506 on DCT HYPERMAGNESAEMIA
cells [93]. It has been shown that FK506 treatment
results in decreased intracellular Mg2 + concentrations in Hypermagnesaemia causes a broad variety of symptoms
human osteoblast cells, suggesting that changes in Mg2 + depending on the serum Mg2 + levels (Table 2)
homoeostasis might also be involved in the reduced bone [26,102,103]. In the case of a relatively mild degree of
mineralization caused by FK506 treatment [94]. A recent hypermagnesaemia, patients may suffer from nausea,
study has shown that FK506 can suppress VDR (vitamin vomiting and flushing. Within the asymptomatic
D receptor) expression; the renal loss of both Mg2 + and mildly symptomatic stages of hypermagnesaemia
and Ca2 + occurring despite increased vitamin D (between 1.1 and 3.0 mmol/l), there is also the therapeutic
levels suggests a possible aetiological role of range in which hypermagnesaemia can be used to
vitamin D resistance [95]. Further studies are needed to control convulsions [104]. When the plasma Mg2 +
unravel the underlying molecular mechanisms. levels increase further, symptoms become increasingly
dangerous, eventually leading to a comatose state and/or
Cisplatin asystole. As the renal response to high Mg2 + levels
Cisplatin (cis-diammine dichloroplatinum) is widely used is extremely efficient, hypermagnesaemia is relatively
to treat various solid tumours. Among the various rare and primarily observed in patients with renal
side effects, nephrotoxicity is generally considered the insufficiency. It has, however, also been detected in
most important since it is the main dose-limiting factor individuals with normal renal function as a result of
in the treatment of patients [96]. Hypomagnesaemia exogenous administration via oral, intravenous or rectal
due to renal Mg2 + wasting is common in cisplatin- routes [105]. A summary of the effects of the compounds
treated patients; an additional effect of reduced intestinal described below is provided in Table 1.
absorption of Mg2 + cannot be excluded as the
absorption of several electrolytes has been shown to Epsom salt poisoning
be reduced by cisplatin treatment [97]. The incidence The heptahydrate of MgSO4 , also known as Epsom
of Mg2 + deficiency is >30 % and is augmented with salt, is often used as bath salt in flotation therapy,
increased dosage or prolonged duration of treatment but it is also applied as a home remedy for numerous
[98]. Interestingly, hypomagnesaemia caused by cisplatin ailments, including abdominal pain, constipation, sprains
treatment seems to be Mg2 + -specific, with hypocalcaemia and muscle strains. Accidental excessive ingestion can
and hypokalaemia only occurring with prolonged and lead to severe hypermagnesaemia. Several fatal or almost
severe Mg2 + deficiency [97]. Cisplatin predominantly fatal cases of hypermagnesaemia due to Epsom salt
accumulates in the PT and DCT, but effects of cisplatin poisoning have been reported in which patients used
may persist long after all inorganic platinum has Epsom salt gargles, Epsom salt enemas or simply ingested
disappeared from renal tissue [99,100]. In addition, several tablespoons of Epsom salt in an attempt to heal
hypomagnesaemia and cisplatin accumulation appear to various ailments [103,106–108].


C The Authors Journal compilation 
C 2012 Biochemical Society
10 A. L. Lameris and others

Overdose using Mg2 + as a cathartic [107,123,124]. As several safe alternatives are available
Gastrointestinal decontamination after a toxic ingestion nowadays, the use of Mg2 + -containing enemas is no
typically involves three actions, all aiming to minimize longer recommended.
absorption of ingested poisons: gastric emptying by
Treatment of hypermagnesaemia
aspiration and lavage, administration of an absorbing
Treatment of hypermagnesaemia should primarily be
agent such as active charcoal to bind the toxic agent, and
aimed at lowering plasma Mg2 + levels. However,
catharsis [109]. Cathartics decrease absorption of toxic
when serious complications of Mg2 + intoxication are
substances by accelerating the transport of both the toxic
present, intravenous administration of low doses of
substance and the charcoal–substance complex through
Ca2 + may quickly reduce the severity of symptoms
the intestine, thereby reducing the bodies exposure
due to the antagonistic effect of Ca2 + on Mg2 + . It is
time to the toxins. Mg3 (C6 H5 O7 )2 (magnesium citrate)
therefore the advised initial treatment in the case of life-
is often used as a cathartic after oral poisoning or
threatening situations. In addition, haemodialysis may
overdosing, and is considered safe in patients with
be used to quickly reduce serum Mg2 + levels in severe
normal renal function. Upon multiple doses, however,
cases [125,126]. Next, oral intake and/or intravenous
this treatment can lead to severe hypermagnesaemia
administration of Mg2 + salts should be discontinued
[108,110,111]. Careful monitoring of serum Mg2 + levels
immediately to avoid further intoxication. Subsequently,
during treatment of patients with multiple doses of
renal Mg2 + excretion could be stimulated by intravenous
Mg3 (C6 H5 O7 )2 is, therefore, strongly advised.
administration of furosemide and saline to ensure high
urine output and to prevent volume depletion [127].
Antacids
Many types of antacids contain Mg(OH)2 . Available as
SUMMARY
over-the-counter drugs, Mg2 + -based antacids such as
milk of magnesia are generally considered to be safe. Tightly controlled Mg2 + balance is essential for ample
The Mg2 + load posed by these drugs can indeed, under physiological processes and consequently disturbance
normal circumstances, easily be excreted via the kidneys. of this balance can have serious clinical consequences.
However, when renal function is compromised or when There is a large variety of drugs that influence
extreme amounts are ingested, hypermagnesaemia may Mg2 + homoeostasis in various ways, causing either
develop [112–115]. hypo- or hyper-magnesaemia. A better understanding
of the mechanisms that are underlying the drug-
Laxative abuse induced changes in Mg2 + homoeostasis could allow the
Various compounds containing Mg2 + [such as development of drugs with less side effects and thus
Mg3 (C6 H5 O7 )2 , Mg(OH)2 and MgSO4 ] can be used better patient care. The discovery of novel key players
as laxatives. They increase the faecal water content, in Mg2 + homoeostasis could help to further elucidate the
leading to softer stools and eventually diarrhoea. This molecular pathways involved in (re)absorption of Mg2 +
has long-been ascribed to the increased osmotic pressure as well as its regulation.
in the colon due to large amounts of unabsorbed Mg2 +
and SO4 2 − Two recent studies, however, have found ACKNOWLEDGEMENT
that the Mg2 + -induced diarrhoea is not solely the result
of increased osmotic pressure. Upon treatment with We thank Bob Glaudemans for his help in preparation of
Mg2 + salts, aquaporin-3 expression in the Caco-2 cell the Figures.
line increases in a Mg2 + -specific manner, inducing an
increase in water permeability [116,117]. This increase in FUNDING
aquaporin-3 expression was also observed in rats [118].
High dosages and impaired renal function increase the Our work was supported by the Netherlands Organiza-
risk of hypermagnesaemia due to the use of laxatives tion for Scientific Research [grant numbers TOP ZonMw
[119,120]. In addition in children and laxative abusers, 91208026, NWO-ALW 818.02.001], a EURYI award
such as for instance patients with eating disorders, from the European Science Foundation and the Dutch
laxatives can lead to hypermagnesaemia [119,121,122]. Kidney Foundation [grant number C08.2252].

Enemas REFERENCES
In the past, Mg2 + -containing enemas were applied in
cases of obstipation. We now know that in the large 1 Groenestege, W. M., Hoenderop, J. G., van den Heuvel,
L., Knoers, N. and Bindels, R. J. (2006) The epithelial
intestine substantial quantities of Mg2 + can be absorbed,
Mg2 + channel transient receptor potential melastatin 6 is
explaining the numerous cases of fatal hypermagnesaemia regulated by dietary Mg2 + content and estrogens. J. Am.
occurring even in patients with normal renal function Soc. Nephrol. 17, 1035–1043


C The Authors Journal compilation 
C 2012 Biochemical Society
Drug-induced alterations in Mg2 + homoeostasis 11

2 Chubanov, V., Waldegger, S., Mederos y Schnitzler, M., 23 Brunette, M. G., Vigneault, N. and Carriere, S. (1974)
Vitzthum, H., Sassen, M. C., Seyberth, H. W., Konrad, M. Micropuncture study of magnesium transport along the
and Gudermann, T. (2004) Disruption of TRPM6/ nephron in the young rat. Am. J. Physiol. 227, 891–896
TRPM7 complex formation by a mutation in the TRPM6 24 Thebault, S., Alexander, R. T., Tiel Groenestege, W. M.,
gene causes hypomagnesemia with secondary Hoenderop, J. G. and Bindels, R. J. (2009) EGF increases
hypocalcemia. Proc. Natl. Acad. Sci. U.S. A. 101, TRPM6 activity and surface expression. J. Am. Soc.
2894–2899 Nephrol. 20, 78–85
3 Chubanov, V., Schlingmann, K. P., Waring, J., Heinzinger, 25 Glaudemans, B., van der Wijst, J., Scola, R. H.,
J., Kaske, S., Waldegger, S., Mederos y Schnitzler, M. and Lorenzoni, P. J., Heister, A., van der Kemp, A. W.,
Gudermann, T. (2007) Hypomagnesemia with secondary Knoers, N. V., Hoenderop, J. G. and Bindels, R. J. (2009)
hypocalcemia due to a missense mutation in the putative A missense mutation in the Kv1.1 voltage-gated
pore-forming region of TRPM6. J. Biol. Chem. 282, potassium channel-encoding gene KCNA1 is linked to
7656–7667
4 Ryazanova, L. V., Rondon, L. J., Zierler, S., Hu, Z., Galli, human autosomal dominant hypomagnesemia. J. Clin.
J., Yamaguchi, T. P., Mazur, A., Fleig, A. and Ryazanov, Invest. 119, 936–942
26 Topf, J. M. and Murray, P. T. (2003) Hypomagnesemia
A. G. (2010) TRPM7 is essential for Mg2 + homeostasis in and hypermagnesemia. Rev. Endocr. Metab. Disord. 4,
mammals. Nat. Commun. 1, 109 195–206
5 Berggard, T., Miron, S., Onnerfjord, P., Thulin, E.,
Akerfeldt, K. S., Enghild, J. J., Akke, M. and Linse, S. 27 Meij, I. C., Koenderink, J. B., van Bokhoven, H., Assink,
(2002) Calbindin D28k exhibits properties characteristic K. F., Groenestege, W. T., de Pont, J. J., Bindels, R. J.,
Monnens, L. A., van den Heuvel, L. P. and Knoers, N. V.
of a Ca2 + sensor. J. Biol. Chem. 277, 16662–16672 (2000) Dominant isolated renal magnesium loss is caused
6 Schweigel, M. and Martens, H. (2000) Magnesium
transport in the gastrointestinal tract. Front. Biosci. 5, by misrouting of the Na + ,K + -ATPase γ -subunit. Nat.
D666–D677 Genet. 26, 265–266
7 Flatman, P. W. (1991) Mechanisms of magnesium 28 Meij, I. C., Saar, K., van den Heuvel, L. P., Nuernberg, G.,
transport. Annu. Rev. Physiol. 53, 259–271 Vollmer, M., Hildebrandt, F., Reis, A., Monnens, L. A.
8 Amasheh, S., Fromm, M. and Gunzel, D. Claudins of and Knoers, N. V. (1999) Hereditary isolated renal
intestine and nephron – a correlation of molecular tight magnesium loss maps to chromosome 11q23. Am. J.
junction structure and barrier function. Acta Physiol. 201, Hum. Genet. 64, 180–188
133–140 29 Geven, W. B., Monnens, L. A., Willems, H. L., Buijs,
9 Fine, K. D., Santa Ana, C. A., Porter, J. L. and Fordtran, W. C. and ter Haar, B. G. (1987) Renal magnesium
J. S. (1991) Intestinal absorption of magnesium from food wasting in two families with autosomal dominant
and supplements. J. Clin. Invest. 88, 396–402 inheritance. Kidney Int. 31, 1140–1144
10 Quamme, G. A. (2008) Recent developments in intestinal 30 Stuiver, M., Lainez, S., Will, C., Terryn, S., Gunzel, D.,
magnesium absorption. Curr. Opin. Gastroenterol. 24, Debaix, H., Sommer, K., Kopplin, K., Thumfart, J.,
230–235 Kampik, N. B. et al. (2011) CNNM2, encoding a
11 Fujita, H., Sugimoto, K., Inatomi, S., Maeda, T., Osanai, basolateral protein required for renal Mg2 + handling, is
M., Uchiyama, Y., Yamamoto, Y., Wada, T., Kojima, T., mutated in dominant hypomagnesemia. Am. J. Hum.
Yokozaki, H. et al. (2008) Tight junction proteins Genet. 88, 333–343
claudin-2 and -12 are critical for vitamin D-dependent
31 Meyer, T. E., Verwoert, G. C., Hwang, S. J., Glazer, N. L.,
Ca2 + absorption between enterocytes. Mol. Biol. Cell 19, Smith, A. V., van Rooij, F. J., Ehret, G. B., Boerwinkle, E.,
1912–1921 Felix, J. F., Leak, T. S. et al. (2011) Genome-wide
12 Wallach, S. (1990) Effects of magnesium on skeletal association studies of serum magnesium, potassium, and
metabolism. Magnes. Trace Elem. 9, 1–14 sodium concentrations identify six loci influencing serum
13 Rude, R. K., Kirchen, M. E., Gruber, H. E., Meyer, M. H.,
magnesium levels. PLoS Genet. 6, e1001045
Luck, J. S. and Crawford, D. L. (1999) Magnesium
deficiency-induced osteoporosis in the rat: uncoupling of 32 Groenestege, W. M., Thébault, S., van der Wijst, J., van
bone formation and bone resorption. Magnes. Res. 12, den Berg, D., Janssen, R., Tejpar, S., van den Heuvel, L. P.,
257–267 van Cutsem, E., Hoenderop, J. G., Knoers, N. V. and
14 Quamme, G. A. (1993) Laboratory evaluation of Bindels, R. J. (2007) Impaired basolateral sorting of
magnesium status. Renal function and free intracellular pro-EGF causes isolated recessive renal hypomagnesemia.
magnesium concentration. Clin. Lab. Med. 13, 209–223 J. Clin. Invest. 117, 2260–2267
15 Le Grimellec, C., Roinel, N. and Morel, F. (1973) 33 Huang, C. L. and Kuo, E. (2007) Mechanism of
Simultaneous Mg, Ca, P, K, Na and Cl analysis in rat hypokalemia in magnesium deficiency. J. Am. Soc.
tubular fluid. II. During acute Mg plasma loading. Nephrol. 18, 2649–2652
Pflugers Arch. 340, 197–210 34 Yamamoto, M., Yamaguchi, T., Yamauchi, M., Yano, S.
16 Le Grimellec, C., Roinel, N. and Morel, F. (1973) and Sugimoto, T. (2011) Acute-onset hypomagnesemia-
Simultaneous Mg, Ca, P, K, Na and Cl analysis in rat induced hypocalcemia caused by the refractoriness of
tubular fluid. I. During perfusion of either inulin or bones and renal tubules to parathyroid hormone. J. Bone
ferrocyanide. Pflugers Arch. 340, 181–196 Miner. Metab. 29, 752–755
17 Greger, R. (1985) Ion transport mechanisms in thick 35 Anast, C. S., Mohs, J. M., Kaplan, S. L. and Burns, T. W.
ascending limb of Henle’s loop of mammalian nephron. (1972) Evidence for parathyroid failure in magnesium
Physiol. Rev. 65, 760–797 deficiency. Science 177, 606–608
18 Quamme, G. A. and Smith, C. M. (1984) Magnesium 36 Schlingmann, K. P., Sassen, M. C., Weber, S., Pechmann,
transport in the proximal straight tubule of the rabbit. U., Kusch, K., Pelken, L., Lotan, D., Syrrou, M., Prebble,
Am. J. Physiol. 246, F544–F550 J. J., Cole, D. E. et al. (2005) Novel TRPM6 mutations in
19 Le Grimellec, C. (1975) Micropuncture study along the 21 families with primary hypomagnesemia and secondary
proximal convoluted tubule. Electrolyte reabsorption in hypocalcemia. J. Am. Soc. Nephrol. 16, 3061–3069
first convolutions. Pflugers Arch. 354, 133–150
20 de Rouffignac, C., Corman, B. and Roinel, N. (1983) 37 Arnaud, M. J. (2008) Update on the assessment of
Stimulation by antidiuretic hormone of electrolyte magnesium status. Br. J. Nutr. 99 (Suppl. 3), S24–S36
tubular reabsorption in rat kidney. Am. J. Physiol. 244, 38 Abrams, S. A. and Wen, J. P. (1999) Methodologies for
F156–F164 using stable isotopes to assess magnesium absorption and
21 Hou, J., Paul, D. L. and Goodenough, D. A. (2005) secretion in children. J. Am. Coll. Nutr. 18, 30–35
Paracellin-1 and the modulation of ion selectivity of tight 39 Feillet-Coudray, C., Coudray, C., Tressol, J. C., Pepin,
junctions. J. Cell Sci. 118, 5109–5118 D., Mazur, A., Abrams, S. A. and Rayssiguier, Y. (2002)
22 Desfleurs, E., Wittner, M., Simeone, S., Pajaud, S., Moine, Exchangeable magnesium pool masses in healthy women:
G., Rajerison, R. and Di Stefano, A. (1998) effects of magnesium supplementation. Am. J. Clin. Nutr.
Calcium-sensing receptor: regulation of electrolyte 75, 72–78
transport in the thick ascending limb of Henle’s loop. 40 Agus, Z. S. (1999) Hypomagnesemia. J. Am. Soc.
Kidney Blood Press. Res. 21, 401–412 Nephrol. 10, 1616–1622


C The Authors Journal compilation 
C 2012 Biochemical Society
12 A. L. Lameris and others

41 Quamme, G. A. and de Rouffignac, C. (2000) Epithelial 60 Kuipers, M. T., Thang, H. D. and Arntzenius, A. B.
magnesium transport and regulation by the kidney. Front. (2009) Hypomagnesaemia due to use of proton pump
Biosci. 5, D694–D711 inhibitors: a review. Neth. J. Med. 67, 169–172
42 Quamme, G. A. (1981) Effect of furosemide on calcium 61 Hoorn, E., Van Der Hoek, J., De Man, R., Kuipers, E.,
and magnesium transport in the rat nephron. Am. J. Bolwerk, C. and Zietse, R. (2010) A case series of proton
Physiol. 241, F340–F347 pump inhibitor-induced hypomagnesemia. Am. J. Kidney
43 Monroy, A., Plata, C., Hebert, S. C. and Gamba, G. Dis. 56, 112–116
(2000) Characterization of the thiazide-sensitive 62 Furlanetto, T. W. and Faulhaber, G. A. (2011)
Na + -Cl − cotransporter: a new model for ions and Hypomagnesemia and proton pump inhibitors: below the
diuretics interaction. Am. J. Physiol. Renal Physiol. 279, tip of the iceberg. Arch. Intern. Med. 171, 1391–1392
F161–F169 63 Insogna, K. (2009) The effect of proton pump-inhibiting
44 Hollifield, J. W. (1986) Thiazide treatment of drugs on mineral metabolism. Am. J. Gastroenterol. 104
hypertension. Effects of thiazide diuretics on serum (Suppl. 2), S2–S4
potassium, magnesium, and ventricular ectopy. Am. J. 64 Wright, M., Sullivan, R., Gaffney-Stomberg, E., Caseria,
Med. 80, 8–12 D., O’Brien, K., Proctor, D., Simpson, C., Kerstetter, J.
45 Grieff, M. and Bushinsky, D. A. (2011) Diuretics and and Insogna, K. (2010) Inhibiting gastric acid production
disorders of calcium homeostasis. Semin. Nephrol. 31, does not affect intestinal calcium absorption in young,
535–541 healthy individuals: a randomized, crossover, controlled
46 Nijenhuis, T., Vallon, V., van der Kemp, A. W., Loffing, J., clinical trial. J. Bone Miner. Res. 25, 2205–2211
Hoenderop, J. G. and Bindels, R. J. (2005) Enhanced 65 Thongon, N. and Krishnamra, N. (2011) Omeprazole
passive Ca2 + reabsorption and reduced Mg2 + channel decreases magnesium transport across Caco-2
abundance explains thiazide-induced hypocalciuria and monolayers. World J. Gastroenterol. 17, 1574–1583
hypomagnesemia. J. Clin. Invest. 115, 1651–1658 66 Shah, G. M. and Kirschenbaum, M. A. (1991) Renal
47 Dai, L. J., Friedman, P. A. and Quamme, G. A. (1997) magnesium wasting associated with therapeutic agents.
Cellular mechanisms of chlorothiazide and cellular Miner. Electrolyte Metab. 17, 58–64
potassium depletion on Mg2 + uptake in mouse distal 67 Wilkinson, R., Lucas, G. L., Heath, D. A., Franklin, I. M.
convoluted tubule cells. Kidney Int. 51, 1008–1017 and Boughton, B. J. (1986) Hypomagnesaemic tetany
48 Quamme, G. A. (1997) Renal magnesium handling: new associated with prolonged treatment with
aminoglycosides. Br. Med. J. 292, 818–819
insights in understanding old problems. Kidney Int. 52,
68 Sieber, M., Hoffmann, D., Adler, M., Vaidya, V. S.,
1180–1195
Clement, M., Bonventre, J. V., Zidek, N., Rached, E.,
49 Reilly, R. F. and Ellison, D. H. (2000) Mammalian distal
Amberg, A., Callanan, J. J. et al. (2009) Comparative
tubule: physiology, pathophysiology, and molecular
analysis of novel noninvasive renal biomarkers and
anatomy. Physiol. Rev. 80, 277–313
metabonomic changes in a rat model of gentamicin
50 Gray, A., Dull, T. J. and Ullrich, A. (1983) Nucleotide nephrotoxicity. Toxicol. Sci. 109, 336–349
sequence of epidermal growth factor cDNA predicts a 69 Mingeot-Leclercq, M. P. and Tulkens, P. M. (1999)
128,000-molecular weight protein precursor. Nature 303, Aminoglycosides: nephrotoxicity. Antimicrob. Agents
722–725 Chemother. 43, 1003–1012
51 Le Gall, S. M., Menton, P., Manduit, P. and Dreux, C. 70 Sassen, M. C., Kim, S. W., Kwon, T. H., Knepper, M. A.,
(2004) The sequential cleavage of membrane anchored Miller, R. T., Frøkiaer, J. and Nielsen, S. (2006)
pro-EGF requires a membrane serine protease other than Dysregulation of renal sodium transporters in
kallikrein in rat kidney. Regul. Pept. 122, 119–129 gentamicin-treated rats. Kidney Int. 70, 1026–1037
51a Breyer, J. A. and Coher, S. (1990) The epidermal growth 71 Tzovaras, V., Tsimihodimos, V., Kostara, C., Mitrogianni,
factor precursor isolated from murine kidney membranes. Z. and Elisaf, M. (2011) Aminoglycoside-induced
Chemical characterization and biological properties. nephrotoxicity studied by proton magnetic resonance
J. Biol. Chem. 265, 16564–16570 spectroscopy of urine. Nephrol. Dial. Transplant. 26,
52 Ikari, A., Sanada, A., Sawada, H., Okude, C., Tonegawa, 3219–3224
C. and Sugatani, J. (2011) Decrease in transient receptor 72 Ward, D. T., Maldonado-Perez, D., Hollins, L. and
potential melastatin 6 mRNA stability caused by Riccardi, D. (2005) Aminoglycosides induce acute cell
rapamycin in renal tubular epithelial cells. Biochim. signaling and chronic cell death in renal cells that express
Biophys. Acta 1806, 1502–1508 the calcium-sensing receptor. J. Am. Soc. Nephrol. 16,
53 Fakih, M. G., Wilding, G. and Lombardo, J. (2006) 1236–1244
Cetuximab-induced hypomagnesemia in patients with 73 Ward, D. T., McLarnon, S. J. and Riccardi, D. (2002)
colorectal cancer. Clin. Colorectal Cancer 6, 152–156 Aminoglycosides increase intracellular calcium levels and
54 Cao, Y., Liao, C., Tan, A., Liu, L. and Gao, F. (2010) ERK activity in proximal tubular OK cells expressing the
Meta-analysis of incidence and risk of hypomagnesemia extracellular calcium-sensing receptor. J. Am. Soc.
with cetuximab for advanced cancer. Chemotherapy 56, Nephrol. 13, 1481–1489
459–465 74 Mani, S. (1992) Pentamidine-induced renal magnesium
55 Tejpar, S., Piessevaux, H., Claes, K., Piront, P., wasting. AIDS 6, 594–595
Hoenderop, J. G., Verslype, C. and van Cutsem, E. (2007) 75 Shah, G. M., Alvarado, P. and Kirschenbaum, M. A.
Magnesium wasting associated with epidermal- (1990) Symptomatic hypocalcemia and hypomagnesemia
growth-factor receptor-targeting antibodies in colorectal with renal magnesium wasting associated with
cancer: a prospective study. Lancet Oncol. 8, pentamidine therapy in a patient with AIDS. Am. J. Med.
387–394 89, 380–382
56 Dimke, H., van der Wijst, J., Alexander, T. R., Meijer, I. 76 Gradon, J. D., Fricchione, L. and Sepkowitz, D. (1991)
M., Mulder, G. M., van Goor, H., Tejpar, S., Hoenderop, Severe hypomagnesemia associated with pentamidine
J. G. and Bindels, R. J. (2010) Effects of the EGFR therapy. Rev. Infect. Dis. 13, 511–512
inhibitor erlotinib on magnesium handling. J. Am. Soc. 77 Wang, J. J., Freeman, A. I., Gaeta, J. F. and Sinks, L. F.
Nephrol. 21, 1309–1316 (1970) Unusual complications of pentamidine in the
57 Epstein, M., McGrath, S. and Law, F. (2006) treatment of Pneumocystis carinii pneumonia. J. Pediatr.
Proton-pump inhibitors and hypomagnesemic 77, 311–314
hypoparathyroidism. N. Engl. J. Med. 355, 1834–1836 78 Lachaal, M. and Venuto, R. C. (1989) Nephrotoxicity and
58 Cundy, T. and Dissanayake, A. (2008) Severe hyperkalemia in patients with acquired
hypomagnesaemia in long-term users of proton-pump immunodeficiency syndrome treated with pentamidine.
inhibitors. Clin. Endocrinol. 69, 338–341 Am. J. Med. 87, 260–263
59 Regolisti, G., Cabassi, A., Parenti, E., Maggiore, U. and 79 Andoh, T. F., Burdmann, E. A., Fransechini, N.,
Fiaccadori, E. (2010) Severe hypomagnesemia during Houghton, D. C. and Bennett, W. M. (1996) Comparison
long-term treatment with a proton pump inhibitor. Am. J. of acute rapamycin nephrotoxicity with cyclosporine and
Kidney Dis. 56, 168–174 FK506. Kidney Int. 50, 1110–1117


C The Authors Journal compilation 
C 2012 Biochemical Society
Drug-induced alterations in Mg2 + homoeostasis 13

80 da Silva, C. A., de Bragança, A. C., Shimizu, M. H., 96 Lajer, H. and Daugaard, G. (1999) Cisplatin and
Sanches, T. R., Fortes, M. A., Giorgi, R. R., Andrade, L. hypomagnesemia. Cancer Treat. Rev. 25, 47–58
and Seguro, A. C. (2009) Rosiglitazone prevents 97 Bearcroft, C. P., Domizio, P., Mourad, F. H., Andre, E. A.
sirolimus-induced hypomagnesemia, hypokalemia, and and Farthing, M. J. (1999) Cisplatin impairs fluid and
downregulation of NKCC2 protein expression. Am. J. electrolyte absorption in rat small intestine: a role for
Physiol. Renal Physiol. 297, F916–F922 5-hydroxytryptamine. Gut 44, 174–179
81 Laniado-Laborı́n, R. and Cabrales-Vargas, M. N. (2009) 98 Mavichak, V., Coppin, C. M., Wong, N. L., Dirks, J. H.,
Amphotericin B: side effects and toxicity. Rev. Iberoam Walker, V. and Sutton, R. A. (1988) Renal magnesium
Micol. 26, 223–227 wasting and hypocalciuria in chronic cis-platinum
82 Barton, C. H., Pahl, M., Vaziri, N. D. and Cesario, T. nephropathy in man. Clin. Sci. 75, 203–207
(1984) Renal magnesium wasting associated with 99 Markmann, M., Rothman, R., Reichman, B., Hakes, T.,
amphotericin B therapy. Am. J. Med. 77, 471–474 Lewis, Jr, J. L., Rubin, S., Jones, W., Almadrones, L. and
83 Crumpacker, C. S. (1992) Mechanism of action of Hoskins, W. (1991) Persistent hypomagnesemia following
foscarnet against viral polymerases. Am. J. Med. 92, 3S–7S cisplatin chemotherapy in patients with ovarian cancer. J.
84 Huycke, M. M., Naguib, M. T., Stroemmel, M. M., Blick, Cancer Res. Clin. Oncol. 117, 89–90
K., Monti, K., Martin-Munley, S. and Kaufman, C. (2000) 100 Kroning, R., Lichtenstein, A. K. and Nagami, G. T. (2000)
A double-blind placebo-controlled crossover trial of Sulfur-containing amino acids decrease cisplatin
intravenous magnesium sulfate for foscarnet-induced
cytotoxicity and uptake in renal tubule epithelial cell
ionized hypocalcemia and hypomagnesemia in patients
lines. Cancer Chemother. Pharmacol. 45, 43–49
with AIDS and cytomegalovirus infection. Antimicrob.
Agents Chemother. 44, 2143–2148 101 Yokoo, K., Murakami, R., Matsuzaki, T., Yoshitome, K.,
84a Stünzi, H. and Perrin, D. D. (1979) Stability constants of Hamada, A. and Saito, H. (2009) Enhanced renal
metal complexes of phosphonoacetic acid. J. Inorg. accumulation of cisplatin via renal organic cation
Chem. 10, 309–319 transporter deteriorates acute kidney injury in
84b Noormohamed, F. H., Youte, M. S., Tang, B., hypomagnesemic rats. Clin. Exp. Nephrol. 13, 578–584
Martin-Munley, S., Gazzard, B. G. and Lant, A. F. (1996) 102 Kutsal, E., Aydemir, C., Eldes, N., Demirel, F., Polat, R.,
Foscarnet-induced changes in plasma concentrations of Taspnar, O. and Kulah, E. (2007) Severe hypermagnesemia
total and ionized calcium and magnesium in HIV-positive as a result of excessive cathartic ingestion in a child
patients. Antivir. Ther. 1, 172–179 without renal failure. Pediatr. Emerg. Care 23, 570–572
85 Kim, H. C., Hwang, E. A., Han, S. Y., Park, S. B., Kim, 103 Birrer, R. B., Shallash, A. J. and Totten, V. (2002)
H. T. and Cho, W. H. (2004) Primary Hypermagnesemia-induced fatality following epsom salt
immunosuppression with tacrolimus in kidney gargles. J. Emerg. Med. 22, 185–188
transplantation: three-year follow-up in a single center. 104 Lu, J. F. and Nightingale, C. H. (2000) Magnesium sulfate
Transplant. Proc. 36, 2082–2083 in eclampsia and pre-eclampsia: pharmacokinetic
86 June, C. H., Thompson, C. B., Kennedy, M. S., Nims, J. principles. Clin. Pharmacokinet. 38, 305–314
and Thomas, E. D. (1985) Profound hypomagnesemia 105 Clark, B. A. and Brown, R. S. (1992) Unsuspected morbid
and renal magnesium wasting associated with the use of hypermagnesemia in elderly patients. Am. J. Nephrol. 12,
cyclosporine for marrow transplantation. Transplantation 336–343
39, 620–624 106 Nordt, S. P., Williams, S. R., Turchen, S., Manoguerra, A.,
87 Ahmadi, F., Naseri, R. and Lessan-Pezeshki, M. (2009) Smith, D. and Clark, R. F. (1996) Hypermagnesemia
Relation of magnesium level to cyclosporine and following an acute ingestion of Epsom salt in a patient
metabolic complications in renal transplant recipients. with normal renal function. J. Toxicol. Clin. Toxicol. 34,
Saudi J. Kidney Dis. Transpl. 20, 766–769 735–739
88 Sabbagh, F., El Tawil, Z., Lecerf, F., Hulin, A., Maurois, 107 Tofil, N. M., Benner, K. W. and Winkler, M. K. (2005)
P., Dartevelle, P., Bac, P. and German-Fattal, M. (2008) Fatal hypermagnesemia caused by an Epsom salt enema: a
Impact of cyclosporine A on magnesium homeostasis: case illustration. South. Med. J. 98, 253–256
clinical observation in lung transplant recipients and 108 Gerard, S. K., Hernandez, C. and Khayam-Bashi, H.
experimental study in mice. Transplantation 86, 436–444 (1988) Extreme hypermagnesemia caused by an overdose
89 Chang, C. T., Hung, C. C., Tian, Y. C., Yang, C. W. and of magnesium-containing cathartics. Ann. Emerg. Med.
Wu, M. S. (2007) Ciclosporin reduces paracellin-1 17, 728–731
expression and magnesium transport in thick ascending 109 Weber, C. A. and Santiago, R. M. (1989)
limb cells. Nephrol. Dial. Transplant. 22, 1033–1040 Hypermagnesemia. A potential complication during
90 Ikari, A., Okude, C., Sawada, H., Takahashi, T., Sugatani,
treatment of theophylline intoxication with oral activated
J. and Miwa, M. (2008) Down-regulation of
charcoal and magnesium-containing cathartics. Chest 95,
TRPM6-mediated magnesium influx by cyclosporin A.
Naunyn. Schmiedebergs Arch. Pharmacol. 377, 333–343 56–59
91 Ledeganck, K. J., Boulet, G. A., Horvath, C. A., Vinckx, 110 Smilkstein, M. J., Smolinske, S. C., Kulig, K. W. and
M., Bogers, J. J., Van Den Bossche, R., Verpooten, G. A. Rumack, B. H. (1988) Severe hypermagnesemia due to
and De Winter, B. Y. (2011) Expression of renal distal multiple-dose cathartic therapy. West. J. Med. 148,
tubule transporters TRPM6 and NCC in a rat model of 208–211
cyclosporine nephrotoxicity and the effect of EGF 111 Smilkstein, M. J., Steedle, D., Kulig, K. W., Marx, J. A.
treatment. Am. J. Physiol. Renal Physiol. 301, F486–F493 and Rumack, B. H. (1988) Magnesium levels after
92 Lote, C. J., Thewles, A., Wood, J. A. and Zafar, T. (2000) magnesium-containing cathartics. J. Toxicol. Clin.
The hypomagnesaemic action of FK506: urinary Toxicol. 26, 51–65
excretion of magnesium and calcium and the role of 112 Herzog, P. and Holtermuller, K. H. (1982) Antacid
parathyroid hormone. Clin. Sci. 99, 285–292 therapy: changes in mineral metabolism. Scand. J.
93 Nijenhuis, T., Hoenderop, J. G. and Bindels, R. J. (2004) Gastroenterol. Suppl. 75, 56–62
Downregulation of Ca2 + and Mg2 + transport proteins 113 Whang, R. (1978) Antacid-induced hypermagnesemia.
in the kidney explains tacrolimus (FK506)-induced West. J. Med. 129, 256
hypercalciuria and hypomagnesemia. J. Am. Soc. 114 Humphrey, M., Kennon, S. and Pramanik, A. K. (1981)
Nephrol. 15, 549–557 Hypermagnesemia from antacid administration in a
94 Jeon, S. H., Kim, S. J., Kim, J. S. and Kang, H. S. (2009) newborn infant. J. Pediatr. 98, 313–314
Immunosuppressant FK506 decreases the intracellular 115 McLaughlin, S. A. and McKinney, P. E. (1998)
magnesium in the human osteoblast cell by inhibiting the Antacid-induced hypermagnesemia in a patient with
ERK1/2 pathway. Life Sci. 84, 23–27 normal renal function and bowel obstruction. Ann.
95 Lee, C. T., Ng, H. Y., Lien, Y. H., Lai, L. W., Wu, M. S., Pharmacother. 32, 312–315
Lin, C. R. and Chen, H. C. (2011) Effects of cyclosporine, 116 Okahira, M., Kubota, M., Iguchi, K., Usui, S. and Hirano,
tacrolimus and rapamycin on renal calcium transport and K. (2008) Regulation of aquaporin 3 expression by
vitamin D metabolism. Am. J. Nephrol. 34, 87–94 magnesium ion. Eur. J. Pharmacol. 588, 26–32


C The Authors Journal compilation 
C 2012 Biochemical Society
14 A. L. Lameris and others

117 Ikarashi, N., Mochiduki, T., Takasaki, A., Ushiki, T., 122 Gren, J. and Woolf, A. (1989) Hypermagnesemia
Baba, K., Ishii, M., Kudo, T., Ito, K., Toda, T., Ochiai, W. associated with catharsis in a salicylate-intoxicated
and Sugiyama, K. (2011) A mechanism by which the patient with anorexia nervosa. Ann. Emerg. Med. 18,
osmotic laxative magnesium sulphate increases the 200–203
intestinal aquaporin 3 expression in HT-29 cells. Life Sci. 123 Ashton, M. R., Sutton, D. and Nielsen, M. (1990) Severe
88, 194–200 magnesium toxicity after magnesium sulphate enema in a
118 Ikarashi, N., Ushiki, T., Mochizuki, T., Toda, T., Kudo, chronically constipated child. Br. Med. J. 300, 541
T., Baba, K., Ishii, M., Ito, K., Ochiai, W. and Sugiyama, 124 Brown, A. T. and Campbell, W. A. (1978) Hazards of
K. (2011) Effects of magnesium sulphate administration hypertonic magnesium enema therapy. Arch. Dis. Child.
on aquaporin 3 in rat gastrointestinal tract. Biol. Pharm. 53, 920
Bull. 34, 238–242 125 Shah, G. M., Winer, R. L., Cutler, R. E., Arieff, A. I.,
119 Zaman, F. and Abreo, K. (2003) Severe hypermagnesemia Goodman, W. G., Lacher, J. W., Schoenfeld, P. Y.,
as a result of laxative use in renal insufficiency. South. Coburn, J. W. and Horowitz, A. M. (1987) Effects of a
Med. J. 96, 102–103 magnesium-free dialysate on magnesium metabolism
120 Qureshi, T. and Melonakos, T. K. (1996) Acute during continuous ambulatory peritoneal dialysis. Am. J.
hypermagnesemia after laxative use. Ann. Emerg. Med. Kidney Dis. 10, 268–275
28, 552–555 126 Weisinger, J. R. and Bellorin-Font, E. (1998) Magnesium
121 Tatsuki, M., Miyazawa, R., Tomomasa, T., Ishige, T., and phosphorus. Lancet 352, 391–396
Nakazawa, T. and Arakawa, H. (2011) Serum magnesium 127 Kraft, M. D., Btaiche, I. F., Sacks, G. S. and Kudsk, K. A.
concentration in children with functional constipation (2005) Treatment of electrolyte disorders in adult patients
treated with magnesium oxide. World J. Gastroenterol. in the intensive care unit. Am. J. Health. Syst. Pharm. 62,
17, 779–783 1663–1682

Received 24 January 2012; accepted 31 January 2012


Published on the Internet 12 March 2012, doi:10.1042/CS20120045


C The Authors Journal compilation 
C 2012 Biochemical Society
View publication stats

You might also like