You are on page 1of 11

18 Nephrology Self-Assessment Program - Vol 22, No 1, April 2023

Article
Calcium Regulation and Management of Hypo- and
Hypercalcemia
Silvia M. Titan, MD, PhD
Nephrology & Hypertension Division, Mayo Clinic, Rochester, Minnesota
Nephrology Division, Faculdade de Medicina da Universidade de S~ao Paulo, S~ao Paulo, SP, Brazil
Rosa M. A. Moyse s, MD, PhD
Laboratorio de Investigaç~ao Medica 16 do Hospital das Clınicas da Faculdade de Medicina da Universidade de S~ao
Paulo, S~ao Paulo, SP, Brazil

of calcium to the loop of Henle predisposes to nephrocalcinosis, and


Learning Objectives nephrolithiasis. In addition, these animals have increased net intesti-
nal absorption, due to reduced intestinal calcium secretion. The authors
1. To identify the main processes related to calcium transport in also evaluated common genetic variants using a genome-wide associa-
the kidney, as well as the main transporters and hormones tion study in 11,130 kidney stone cases and 187,639 controls and
found that nine single nucleotide polymorphisms in the noncoding
mediating these processes
region of the claudin-2 gene (CLDN2) were associated with nephro-
2. To recognize new knowledge related to the role of claudins in lithiasis risk (4). Further analysis suggested that these single nucleotide
calcium tubular transport polymorphisms (SNPs) are able to decrease claudin-2 expression.
Therefore, this paper corroborates the role of claudin-2 in nephro-
3. To recognize new knowledge related to the role of CaSRs in the
lithiasis and identifies a potential target for therapy in the future.
kidneys In the TALH, calcium transport is also paracellular and depen-
4. To recognize causes of hypo and hypercalcemia related to dent on the gradient created by active Na1 and Cl2 transport
through Na1-K1-2Cl2 cotransporter (NKCC2) in the luminal
kidney disease
membrane, Na1/K1-ATPase in the basolateral membrane and K1
recirculation through renal outer medullary potassium channel in
the apical membrane. The molecular basis for the paracellular trans-
Physiology port in the TALH was not clear until recently, when mutations in
Approximately 60% of calcium in the blood is not bound to albu- claudins 16 and 19 causing familial hypomagnesemia with hypercal-
min and is filtered at the glomerulus, either as free ionic calcium ciuria and nephrocalcinosis were identified (5,6). Subsequently,
or complexed with bicarbonate, citrate, or phosphate. Under physi- claudin-14 was reported to modulate calcium transport by reducing
ologic conditions, of the total amount filtered (filtered load of cal- cation permeability of claudin-16 (7), and mutations in claudin-14
cium), only a minor percentage is excreted. Therefore, although have been associated with increased risk of kidney stones and
the glomerular filtration rate (GFR) is a major determinant of the reduced bone mineral density in a genome-wide association study
urinary calcium excretion, it is the tubular handling of calcium the (8). Acquired defects related to claudins may also be responsible for
one responsible for fine tuning of the final amount excreted. About disturbances in calcium and magnesium transport. In a recent publi-
65% of the filtered calcium is reabsorbed at the proximal tubule, cation, antibodies against claudin-16 have been described in a
with the remaining 20%, 10%, and 5% handled in the thick patient presenting severe magnesium wasting, hypocalcemia, and
ascending loop of Henle (TALH), distal tubule, and connecting tubulo-interstitial chronic nephropathy (9). Intravenous administra-
tubule, respectively (1). tion of his serum caused severe urinary waste in rats. Of note, the
In the proximal tubule, calcium transport occurs mainly patient received diagnosis 3 years later of a renal carcinoma expres-
through a paracellular pathway due to a favorable electrochemical sing high levels of claudin-16 mRNA. Claudins are a family of at
gradient secondary to sodium and chloride reabsorption (solvent least 26 proteins that interact with each other either in the same cell
drag) and to a lesser extent, transcellular transport. The paracellular or across cells, forming tight junctions (10). Claudins have four
transport is facilitated by claudin-2, and mutations in this protein transmembrane domains and the first domain is related to paracellu-
are associated with hypercalciuria in mice (2,3) and implicated in lar permeability, acting as barrier or pore depending on its proper-
the risk of kidney stones in human (4). Curry et al. (4) recently ties. The kidney expresses many different forms of claudins,
showed that claudin-2-null mice have decreased renal tubular reab- predominantly in the tubule but also in the epithelial cell of Bow-
sorption and papillary nephrocalcinosis. The increased distal delivery man capsule and in podocyte.

nephsap.org Copyright # 2023 by the American Society of Nephrology


Nephrology Self-Assessment Program - Vol 22, No 1, April 2023 19

with autosomal dominant hypocalcemia type 1 (ADH1) present


4 Claudins are directly involved in the molecular hypocalcemia but variable calciuria. Therapy with calcium and vita-
basis of paracellular transport of calcium (and min D may lead to hypercalciuria in these patients, which has been
magnesium) in the proximal tubule, through linked to the risk of nephrocalcinosis and renal injury. By studying
mouse models and one ADH1 patient, van Megen et al. suggest cal-
claudin-2, and in the TALH, through claudins 14, ciuria is being modulated by renal CaSR response to calcemia (14).
16, and 19. Claudin mutations have been associ- Gain-of-function genetic variants of CaSRs are now known to cause
ated with increased risk of kidney stones (claudins Bartter syndrome (15).
2 and 14), familial hypomagnesemia, hypercal- Calciotropic hormones regulate calcium excretion through com-
bined effects on bone, intestine, kidney, and parathyroid glands. PTH
ciuria and nephrocalcinosis (claudins 16 and 19), release is triggered by low serum calcium level through the absence of
and decreased bone mineral density (claudin 14). activation of CaSRs in the parathyroid cells. PTH exerts its multiple
effects in several cell types, leading to an integrated response that
releases calcium back to the circulation and restores normocalcemia.
In the distal tubule, Ca21 reabsorption is an active transcellular In osteoblasts, PTH binds to its receptor, causing an increase in
process, involving the permeable ion channel (TRPV5) at the apical RANKL (receptor activator of nuclear factor kB ligand) and a
membrane, intracellular binding to calbindins and extrusion to the decrease in osteoprotegerin expression, which collectively activate
circulation through Na1/Ca21 exchanger, plasma membrane Ca21- osteoclasts by RANK-RANKL, resulting in an increase in bone
ATPase (PMCA), and Na1-Ca21-K1 exchanger (NaCKX) localized resorption. PTH also has noncellular effects and promotes calcium
at the basolateral membrane. KCTD1 gene codes for the potassium buffering through increased expression of calcium-binding proteins,
channel tetramerization domain containing 1 protein and promotes osteocalcin, and osteonectin in the bone surface (16). In addition to
differentiation of TAL and DCT cells. KCTD1 has been shown to its bone effects, PTH increases tubular reabsorption of calcium by
modulate magnesium and calcium transport in these segments by activation of adenylyl cyclase systems in the proximal tubule and,
reducing NCC cotransporter expression in the DCT and by reduc- more importantly, through increased activity and expression of
ing NKCC2/claudin-16/claudin-19 and increasing claudin-14 TRPV5, increased expression of calbindin-D28k and reduced expres-
expression in the TAL (11). At the collecting duct, calcium is not sion of Na/Cl cotransporter in the distal tubule. PTH also reduces the
reabsorbed. Figure 1 summarizes the molecular basis for calcium availability of Na/Pi cotransporter in the proximal tubule and increases
transport along the tubule. the activity of 1a-hydroxylase. Despite the effect of PTH to augment
Emerging evidence from the last decade supports the concept calcium tubular reabsorption, excess of PTH, such as seen in primary
that the kidney is a calcium-sensing organ, with expression of hyperparathyroidism, leads to hypercalciuria, with increased risk of kid-
calcium-sensing receptors (CaSRs) along the nephron, particularly ney stones and nephrocalcinosis. This effect is a result of increased
in the TALH. The actions of kidney CaSRs are still not completely calcium filtered load secondary to bone resorption and increased natri-
understood, but some of the roles include counter-regulation of uresis secondary to downregulation of proteins related to sodium trans-
parathyroid hormone (PTH) effects (by upregulation of the sodium port, particularly NHE-3. FGF-23 and klotho have indirect effects on
hydrogen antiporter-3 [NHE-3] and inhibition of phosphaturia) calcium handling, by inhibiting the synthesis of PTH and 1,25-vitamin
and 1.25-vitamin D effects (by mitigation of the calcemic response), D (the latter independently of the former). However, FGF-23 and
inhibition of apical K1 recycling in the TALH with limitation klotho have also been shown to regulate TRPV-5 independently of
of paracellular calcium and magnesium transport, modulation PTH, thus promoting calcium reabsorption (17).
of claudin-14 (which regulates claudin 16 and 19), reduced Whether 1,25-vitamin D has effects on tubular calcium trans-
vasopressin-mediated trafficking and expression of aquaporin-2 port independent of its effect on calcium intestinal absorption in
(which explains the polyuria seen with hypercalcemia and modulates humans is not clear. Several animal studies have shown that 1,25-
the risk of kidney stones), and stimulation of renin release by the vitamin D enhances the expression of TRPV5, TRPV6, calbindins
juxtaglomerular cells, among others (12). CaSRs also cause an and PMCA in distal tubules and collecting duct. Results from a
increase in the sodium-chloride cotransporter activity in the distal double knock-out model for TRPV5 and 1a-hydroxylase also sug-
tubule (13). This effect contributes to the hypercalciuric effect of gest that 1,25-vitamin D has direct tubular effects, increasing cal-
the kidney CaSRs. Overall, these findings point to an integrated cium reabsorption (18). In addition, recent evidence suggests that
action of kidney CaSRs in promoting urinary calcium excretion, a sclerostin, an osteocyte-derived hormone is another component of
likely counter-regulatory response to increased tubular calcium traf- bone metabolism. Sclerostin inhibits the Wnt-signaling pathway
fic. CaSRs can be downregulated by PTH and phosphorus. Patients and has antianabolic effects on osteoblasts. Mutations in its gene or
transcription factor are associated with increased bone mass (scleros-
teosis and van Buchem disease, respectively) (19). Sclerostin produc-
4 The kidney is a calcium-sensing organ. When acti- tion is inhibited by PTH, mechanical load, and estrogen among
others and stimulated by calcitonin and steroids. Experimental mod-
vated by increased calcium load, the kidney CaSR
els have shown that sclerostin has direct and indirect effects on
promotes an integrated response that decreases 1,25-vitamin D synthesis. Knock-out mice for sclerostin have
calcium reabsorption and thus increases calcium reduced urinary calcium excretion and fractional calcium excretion,
excretion. as well as evidence of increased 1,25-vitamin D synthesis, which was
reverted by sclerostin infusion (20).
20 Nephrology Self-Assessment Program - Vol 22, No 1, April 2023

Figure 1. Schematic representation of calcium transport along the renal tubule. Calcium (Ca11) is filtered at the glomerulus, and 65% of its
ultrafiltrable fraction is reabsorbed at the proximal tubule (PT). The remaining 20%, 10%, and 5% are handled in the thick ascending loop of
Henle (TALH), distal convoluted tubule (DCT), and connecting tubule, respectively. In the PT, Ca11 transport occurs mainly through a para-
cellular pathway due to a favorable electrochemical gradient secondary to sodium (Na) and to a lesser extent, due to transcellular transport. Clau-
dins (Cl) regulate paracellular transport in PT (Cl2) and TAL (Cl14 regulates Cl16 and 19). In the TALH, Ca11 transport is also paracellular,
dependent on the gradient created by active Na1 and Cl2 transport through Na1-K1-2Cl2 cotransporter in the luminal membrane, Na1/K1-
ATPase in the basolateral membrane and K1 recirculation through renal outer medullary potassium channel in the apical membrane. In the dis-
tal tubule, Ca21 reabsorption becomes an active transcellular process, involving the permeable ion channel (TRPV5) at the apical membrane,
intracellular binding to calbindins (CB28) and final extrusion to the circulation through Na1/Ca21 exchanger, plasma membrane Ca21-ATPase
(PMCA), and Na1-Ca21-K1 exchanger (NaCKX) localized at the basolateral membrane. There is no calcium reabsorption at the collecting
duct (CD), but apical calcium sensing receptor (CaSR) senses urinary calcium concentration, inhibiting water reabsorption (through the inhibi-
tion of aquaporin-2; AQP2) and stimulating urine acidification. Hormones that regulate calcium transport across the nephron are parathyroid
hormone, calcitriol, calcitonin, and estradiol. There is new evidence that sclerostin, Klotho, and FGF-23 also influence tubular calcium
reabsorption.

There are several other factors that regulate calcium transport, These abnormalities were accompanied by decreased calcium
which may be independent or mediated by calciotropic hormones. reabsorption in the proximal tubule and TALH, likely mediated by
A recent study evaluated the effects of high versus normal sodium reduced expression of the sodium-proton exchanger (NHE3),
and low versus normal potassium diet on urinary calcium and NKCC2, and claudin-16. In addition, the authors described an
phosphate excretion in mice (21). The high-sodium diet increased increased calbindin-28k expression, suggesting the presence of a com-
urinary calcium and phosphate excretion regardless of the dietary pensatory response in the distal tubule that is not capable of revers-
potassium intake. However, serum calcium was lower with a high ing the calcium urinary loss.
sodium but low potassium diet suggesting an additional effect of Hypercalciuria is a well-known epiphenomenon of acidemia.
low-potassium diet in tubular calcium handling. Calcium phosphate is released from the skeleton as consequence of
Nephrology Self-Assessment Program - Vol 22, No 1, April 2023 21

H1 buffering, causing an increase in the calcium (and phosphate)- with 1-25-vitamin D levels, which may explain the increased frac-
filtered load. On top of this short-term effect, studies have shown tional calcium excretion despite the enhanced paracellular calcium
that cell-mediated effects also occur with a reduction in osteoblast reabsorption.
activity and an increase in osteoclast activity, resulting in reduced Adiponectin is an insulin-sensitizing adipokine known to be
bone formation and increased resorption (22). In addition, particu- reduced in obesity and associated with several beneficial effects,
larly in renal tubular acidosis type 1, other specific mechanisms may including renoprotection in experimental models. On the other
be operative (urine pH, citrate concentration), and kidney stones hand, adiponectin has been associated with bone loss. Rutkowski
and nephrocalcinosis are common phenotypes in type 1 renal tubu- et al. evaluated mineral metabolism in adiponectin knock-out, wild-
lar acidosis. Imenez Silva et al. examined the effect of decreased type, and transgenic mice models (28). Overexpression of adiponec-
expression of protein-activated ovarian cancer G-protein (OGR1) tin was related to lower bone mass, higher serum calcium, higher
on acidosis-induced calcium excretion in mice (23). OGR1 is a pro- calcium fractional excretion, lower FGF-23, with no changes in
tein normally expressed in the glomerulus, proximal tubule, and PTH and 1.25 vitamin D, whereas the knock-out model presented
endothelial cells and has been shown to reduce NHE-3 expression similar FGF-23 levels in comparison to the wild type but higher cir-
in the proximal tubule. NH4Cl-induced metabolic acidosis increased culating klotho levels. When challenged with a phosphate-enriched
H1 excretion in both animals. However, whereas the wild-type ani- diet (which results in increased phosphate and decreased calcium
mals demonstrated a positive association between H1 excretion and fractional excretion due to reduced intestinal absorption), transgenic
calciuria with metabolic acidosis, the same effect was not observed mice showed higher calcium excretion, increased FGF-23, and frac-
in knock-out mice, which showed higher expression of NHE-3, tional phosphate excretion than the wild type. Knock-out mice
TRPV5, and calbindin-D28k in comparison to the wild type. These showed a smaller increase in PTH and lower FGF-23 levels. When
data suggest that OGR may detect low pH and modulate resulting challenged with a calcium-enriched diet, all animals increased cal-
hypercalciuria. cium fractional excretion, but this effect was smaller in knock-out
There are sex differences in the urinary calcium and phosphate and higher in transgenic mice. Overall, the findings from this study
handling. Androgen and estrogen receptors are expressed along the suggest adiponectin modulates tubular transport of calcium and
nephron (24), and both experimental and human studies have phosphate, which is partially mediated by klotho expression. Levels
shown that urinary Ca21 excretion is higher in adult male compared of adiponectin are known to be increased in CKD, and studies eval-
to adult females. This observation could be related to differences in uating the effect of this adipokine in klotho expression in CKD-
diet, higher osteoclastic activity in men, and higher calcium filtered MBD are needed.
load (since men have higher absolute GFR values than women),
among others. In animal models, male mice present a reduced renal
expression of Ca21 transport proteins independently of calciotropic Pathophysiology
hormones (25), and estrogens stimulate calcium reabsorption and Proteins involved in tubular calcium transport and intracellular cal-
phosphate excretion (24). During childhood and particularly at cium trafficking are affected by a number of renal pathological con-
puberty, sex is also related to differences in bone mass gain and ditions, such as kidney stones, tubulopathies, and CKD, among
development driven by sex steroids. A study evaluated the effects of others.
testosterone on tubular calcium handling according to aging and sex
(26). Testosterone upregulates expression of SMP-30 (renal senes- Familial Hypomagnesemia with Hypercalciuria and
cence marker protein-30, a calcium-binding protein present in renal
Nephrocalcinosis
proximal tubular cells) and increases tubular calcium reabsorption in
mice, an effect mediated by increased plasma membrane calcium Familial hypomagnesemia with hypercalciuria and nephrocalcinosis
ATPase-4 (PMCA4) expression. Conversely, aging was associated is a monogenic disease caused by mutations in claudin-16 and in
with a reduction in testosterone and in SMP30 expression, as well as claudin-19 (29,30). The calcium and magnesium wasting seen in
an increase in urinary calcium excretion. Male rats presented higher the disease is generally attributable to impairment in the paracellular
SMP30 expression in comparison to female rats of the same age, transport in the TALH. However, one specific mutation (missense
and ovariectomy followed by testosterone replacement increased mutation c.908C.G, p.T303R) of claudin-16 is associated with a
SMP30 expression and reduced the hypercalciuric effect of ovariec- calcium-specific abnormality, with no significant magnesium wast-
tomy. This effect was reversed with the use of an androgen-receptor ing. Knowing that claudin-16 mRNA is expressed in the DCT,
inhibitor. All together, these findings suggest androgen-dependent Hou et al. hypothesized that this mutation is related to disturbance
SMP30 expression may underlie sex differences in calcium handling of a specific function of claudin-16 in the DCT. Through a series of
during puberty and bone loss in andropause. experiments, the authors showed that phosphorylation of claudin-16
Lactation is also related to important changes in renal and causes its expression in the apical membrane of DCT, along with an
intestinal calcium transport driven by the calcium transfer needed increase in TRPV5 abundance and channel conductance (31).
for milk production. A recent animal study compared calcium and Knock-down of claudin-16 expression in transgenic mouse kidney
magnesium excretion and protein expression in nonpregnant control caused delocalization of TRPV5 from the apical membrane of
mice, lactating mice, or mice undergoing involution (27). There was DCT. In addition, the authors showed that a calcium-enriched diet
increased fractional calcium excretion and increased expression of reduced the phosphorylation of claudin-16 through PTH-signaling.
TRPV5 and calbindin-1 during lactation, with no change in the Therefore, these results not only provide evidence for a combined
expression of claudins. In addition, expression of renal 1a- TALH and DCT defect in calcium reabsorption in certain mutations
hydroxylase and 24-OHase were increased during lactation, along of familial hypomagnesemia with hypercalciuria and nephrocalcinosis,
22 Nephrology Self-Assessment Program - Vol 22, No 1, April 2023

but also identified a novel regulatory effect of PTH in calcium tubu- also showed that mechanical stretch, compression, stiffness or TGF-
lar handling. Patients carrying CLDN19 mutations frequently exhibit b1 increased Piezo1, leading to profibrotic responses in cultured
associated congenital ocular defects leading to variable visual impair- tubular cells, which were prevented by inhibition or silence of
ment (29,32). Piezo1. Importantly, the Piezo1-mediated calcium response was
dependent on extracellular calcium levels. When extracellular cal-
Autosomal Dominant Tubulointerstitial Kidney Disease cium was chelated by EGTA, the profibrotic changes were signifi-
A subtype of autosomal dominant tubulointerstitial kidney disease cantly attenuated (40).
(ADTKD) is caused by mutations in the transcription factor, hepa-
tocyte nuclear factor 1b (HNF1b) (33). HNF1b mutations are Autosomal Dominant Polycystic Kidney Disease
associated with a phenotype including tubulointerstitial disease, Polycystin 2 has been characterized as a transient receptor potential
renal cysts, maturity-onset diabetes of the young, hypomagnesemia, channel, playing a role in normal Ca21 signaling in the tubular cell
hypokalemia, and hypocalciuria. Kompatscher et al. evaluated the (41). It has been hypothesized that calcimimetics may play a thera-
effect of HNF1b in the TALH, showing that HNF1b causes an peutic role in ADPKD. An in vitro study showed reduced basal rest-
activation of CaSRs, whereas a mutant protein abolished CaSR ing Ca21 concentration and higher cAMP and mTOR levels in
expression (34). In addition, HNF1b knock-down in immortalized PKD1-knock-down human proximal tubular cells in comparison to
kidney cell was associated with reduced CaSRs, reduction in wild-type cells. CaSR expression by these cells was confirmed and
claudin-14, and upregulation of claudin-10, whereas kidney-specific selective CaSR activation with a calcimimetic reversed the abnormal-
HNF1b knock-down was associated with important reduction in ities described (42). In a second study using the same cell lines, the
CaSR expression, along with reduction in claudin-19 and claudin- authors showed that PKD1 knock-down cells had lower mitochon-
10b. These findings provide evidence for the mechanisms underly- drial calcium levels and a severe reduction in ATP production in
ing the hypocalciuric effects of ADTKD-HNF1b. ADTKD is also comparison to the wild type, findings that were also reversed with a
observed in individuals with missense mutations in the SEC61A1 calcimimetic (43). Mitochondrial TRPC3 upregulation has also
gene, and impaired protein transport of renin is observed, along been shown in polycystin2 knock-down cells, causing an aggravation
with reduction of the expression of calcium transporters. Using an of the mitochondrial abnormalities and cell proliferation (44). How-
in vitro model of renal tubular cells, Sicking and colleagues observed ever, a previous in vivo study has shown that significant hypocalce-
that administration of phenylbutyrate reversed the defective trans- mia prevented the use of calcimimetics in animal models of
port of renin and calcium (35), opening an opportunity for pharma- ADPKD. A recent study, using a combination of small doses of a
cological management of this genetic disease. calcimimetic (R-568) and a vasopressin V2 receptor antagonist (lixi-
vaptan) showed a reduction in cyst progression in two animal mod-
Renal Fibrosis els of human ADPKD (45).
Interstitial fibrosis related to extracellular collagen deposition under-
lies progression of many kidney diseases. Proteins related to calcium Diabetic Kidney Disease
transport have recently been implicated in the development of renal Podocyte lesions and loss of podocytes are common features in dia-
fibrosis. One study evaluated whether the CaSR was involved in col- betic kidney disease and calcium signaling disturbances have been
lagen expression of fibrosis induced by dietary adenine in mice (36). implicated in mediation of kidney diseases presenting podocytopa-
The use of calcimimetic attenuated collagen deposition and abro- thy (46). Using the streptozotocin-induced diabetes model in
gated the collagen upregulation induced by TGF-b1, whereas block- knock-out mice for NADPH oxidase 4, TRPC6, and double knock-
ade of CaSRs enhanced collagen deposition induced by TGF-b1. In out for TRPC5 and TRPC6, Ilatovskaya et al. showed that
another study, Orai-1 (calcium release-activated calcium channel NADPH oxidase 4-deficient mice exhibited lower basal intracellular
protein 1) knock-down prevented renal fibrosis and decreased calcium in podocytes and less damage, with blunting of the
expression of proteins related to fibrosis in models of high-fat diet angiotensin-induced calcium flux in glomeruli (47). H2O2 stimu-
and unilateral ureteral obstruction (37), with further experiments lated calcium flux in wild-type animals, but this response was mini-
suggesting a modulatory effect of Orai-1 over TGF-b1-induced mized in TRPC6 knock-out models. The same group, in a more
pathways. In addition, it has been hypothesized that sympathetic recent study, identified that renal purinergic signaling is also
activation has a role in CKD progression (38). The N-type Ca21 involved in diabetic kidney disease. They showed an increase in
channel is expressed in nerves innervating the kidney, but its role is ATP-mediated calcium influx in the podocytes, through the recep-
not clear. Mishima et al., using a knock-out mouse for the N-type tors P2X4 and P2X7. This disruption in intracellular calcium
Ca21 channel submitted to unilateral ureteral obstruction, showed homeostasis contributes to podocytes dysfunction and aggravation
that (1) the N-type Ca21 channel is increased in the obstructed of diabetic kidney disease progression (48).
fibrotic kidneys and (2) the knock-out mice presented a significantly
attenuated fibrotic response in comparison at least partly mediated Chronic Kidney Disease
by reduction in renal sympathetic activation (39). More recently, CKD is associated with a dramatic decrease in urinary calcium
Piezo1, a mechanosensitive nonselective cation channel, was also excretion. However, the determinants of calciuria are not completely
related to kidney fibrosis. Zhao et al. showed that Piezo1 protein understood, neither the effects of some medications commonly used
expression was markedly upregulated in human fibrotic kidneys. In in this scenario, such as diuretics. Data from observational or retro-
two different CKD models (unilateral ureter obstruction and folic spective studies have shown that furosemide increases calciuria,
acid treatment), there was an increase in Piezo1 expression. They whether thiazides have the opposite effect. Also, higher serum PTH
Nephrology Self-Assessment Program - Vol 22, No 1, April 2023 23

is seen in furosemide users, although this effect could not be bone disease, or conditions such as immobilization. Denosumab (a
completely explained by the increase in urinary calcium excretion. fully humanized monoclonal antibody against RANKL) inhibits
Vasco et al. prospectively evaluated the effects of furosemide and bone resorption, is metabolized by the liver and does not require
hydrochlorothiazide in a randomized trial in patients with stage 3 dose adjustment in CKD. It has been increasingly used in patients
CKD for 1 year. At the follow-up, eGFR decreased, and PTH and with eGFR ,30 ml/min per 1.73 m2, in whom bisphosphonates
FGF-23 increased in both groups. However, furosemide was associ- are not recommended or are contraindicated. However, denosumab
ated with a more significant increase in PTH, 1a,25-vitamin D, is associated with increased risk of hypocalcemia, and this risk may
procollagen type I N-propeptide (P1NP) and collagen type I be higher in those with low GFR, which is also associated with
C-telopeptide (CTX), whereas hydrochlorothiazide attenuated the lower calcium levels. In most patients, hypocalcemia is mild and
PTH rise and caused a reduction in 1a,25-vitamin D and in these asymptomatic; however, symptomatic hypocalcemia occurs in small
bone turnover markers. This effect could not be explained by the percentage of patients (55).
changes in calciuria, highlighting the different effects of these drugs Miyaoka et al. evaluated risk factors for low corrected calcium
on vitamin D metabolism and bone remodeling (49). levels after denosumab treatment in 77 patients with osteoporosis,
Another question that arises is whether calcium supplementa- of whom 35% presented an eGFR of 30–59 ml/min per 1.73 m2
tion in this population would increase calcium uptake by the skele- (56). The nadir of calcium levels occurred at day 7 after treatment.
ton or increase soft tissue deposition. Further understanding on this Although none of the participants presented symptomatic hypocal-
matter is limited by technical difficulties in performing and assessing cemia during the study, asymptomatic hypocalcemia (defined as a
calcium balance. One alternative is using the 44/42Ca ratio. The nat- corrected calcium level less than 8.5 mg/dl) occurred in 10.4%. Low
urally occurring isotopically light 42Ca is preferentially incorporated baseline corrected calcium, eGFR ,60 ml/min per 1.73 m2,
into bone, whereas heavier 44Ca is excreted. The ratio increases tartrate-resistant acid phosphatase-5b, bone alkaline phosphatase,
when bone formation exceeds resorption, so the higher the ratio, the and pretreatment with antiresorptive agents were factors associated
greater the skeleton positive balance. Shroff et al. have previously with the absolute reduction in corrected calcium from baseline to
shown that healthy children have a higher serum ratio than adults. day 7. In a post hoc analysis of the FREEDOM trial (57), the safety
Specifically in CKD, they have shown that 44/42Ca ratio positively and efficacy of the use of denosumab in women with mild to mod-
correlated with serum calcium, 25-hydroxyvitamin D, alkaline phos- erate CKD were evaluated during the trial phase and in the open-
phatase, and total bone mineral content measured by peripheral label 7-year extension study. BMP gain over time and adverse effect
quantitative computed tomography and inversely with PTH and rates were similar among the CKD subgroups (normal, CKD G2,
G3a, and G3b).
bone resorption markers. More impressive was the finding that in
In a series with 47 patients on dialysis, 25% presented acute
children with CKD, the 44/42Ca ratio was 157% lower than that of
hypocalcemia following denosumab administration and tartrate-
age-matched children and 29% lower than levels in elderly women
resistant acid-phosphatase 5b was the best predictor of hypocalcemia,
with osteoporosis, when adjusted for calcium containing medica-
along with bone alkaline phosphatase (58). Thongprayoon et al. per-
tions and kidney function (50).
formed a meta-analysis evaluating the incidence of hypocalcemia in
patients with ESKD treated with denosumab (59). The authors
Renal Cell Cancer
included six observational studies and 84 patients and describe a
CaSR has been implicated in the development of bone metastasis in pooled incidence of hypocalcemia of 42% (95% CI, 29 to 55), occur-
renal cell carcinoma and other cancers (51) and polymorphisms of ring initially between 7 and 20 days and reaching a nadir between
CaSR have been associated with increased risk of several cancers 2 weeks and 2 months after the first dose. There was a significant
(52). CaSR activates Rac and cell migration via the Gbg-PI3K- increase in bone density parameters and reduction in alkaline phos-
mTORC2 pathway (53). Using a renal cell carcinoma cell line phatase and PTH, with no significant changes in serum calcium or
transfected with CaSR, Frees et al. showed that calcium-treated cells phosphate after 3 months. The same group published another meta-
presented an increase in adhesion, production of extracellular pro- analysis restricted to kidney transplant, identifying five eligible studies
teins, cell migration, and proliferation, and these effects were (one clinical trial and four observational studies) comprising 162 kid-
abrogated by a CaSR antagonist (54). In addition, in vivo mice stud- ney transplant patients (60), with the majority having an eGFR
ies showed that bone metastasis was increased after injection of .30 ml/min per 1.73 m2. During denosumab treatment, risk of
CaSR-transfected cells, raising the hypothesis that targeting CaSRs hypocalcemia was greater in the clinical trial and reported as small and
might be beneficial, at least in those with high CaSR expression. mild in observational studies (1.7%, 95% CI, 0.4 to 6.6). In addition,
after treatment, denosumab was related to increase in bone mineral
density and no change in Ca or PTH after treatment. Use of denosu-
Hypocalcemia mab is likely safer in patients presenting hypercalcemia pretreatment,
Hypocalcemia is commonly seen because of diseases related to cal- as suggested by a study evaluating the effects of denosumab in 14
cium transport or secondary to the use of drugs. We will review here hypercalcemic kidney transplant patients with osteoporosis (61).
recent studies related to hypocalcemia, particularly related to drugs In people with cancer, a retrospective study by Kanbayashi et al.
that are increasingly being used in the CKD population. evaluated 327 patients with advanced disease and found that con-
comitant use of vonoprazan (a potassium-competitive acid blocker)
Denosumab or dexamethasone, and pretreatment low serum calcium and hemo-
Several case reports and studies have addressed the risk of hypocalce- globin, as well as high alkaline phosphatase were significant predic-
mia related to denosumab treatment for osteoporosis, metastatic tors for the development of denosumab-induced hypocalcemia (62).
24 Nephrology Self-Assessment Program - Vol 22, No 1, April 2023

In patients who are candidates to denosumab therapy, irrespec- patients that were submitted to parathyroidectomy between 2008
tively of the indication being for metastatic cancer or osteoporosis, and 2019. Zhao et al. found that, beside the predictors previously
calcium and vitamin D should be corrected before denosumab mentioned, the mass of excised parathyroid gland and age were also
administration. Hypomagnesemia, when present, should also be associated with hypocalcemia, which was severe in 36.2% of the
corrected. In those patients with CKD, is it recommended to check patients. Curiously, hyperkalemia, a well-described, but not well-
calcium and PTH 1–2 weeks after administration and correcting explained, phenomenon was found in 24.6% of the patients (72).
hypocalcemia, when present.
Calcimimetics
PD-1 Inhibitors Hypocalcemia is a common side effect of calcimimetics, and a recent
Programmed cell death protein 1 (PD-1) inhibitors are used in the meta-analysis reported an increase in the risk of hypocalcemia by ten
treatment of several tumors, such as melanoma, non–small cell lung times in people with CKD when compared to control (73). In this
cancer, renal cell carcinoma, bladder and urothelial cancer, breast same study, calcimimetic use was associated with improvement in
cancer, among others. These drugs are associated with the risk of biochemical parameters, but not with all-cause or cardiovascular
urinary tract infections, electrolyte abnormalities and acute renal fail- mortality (73). It has been used also in the pediatric population
ure. A meta-analysis of 48 clinical trials including a total of 11,482 (74,75) with apparent safety and efficacy, although larger studies are
participants evaluated the risk of AKI and electrolyte abnormalities needed in this population. In addition, it is often used for manage-
in patients treated with PD-1 inhibitors (63). Of the 48 clinical ment of hyperparathyroidism post-transplant (76), although there is
trials, 16 RCTs were used in the analysis assessing the risk of AKI controversy about the optimal treatment of this condition. A retro-
(n513) and electrolyte abnormalities (n55) and 39 clinical trials spective analysis of the EVOLVE study has shown that, among the
were included in the analysis evaluating the incidence of electrolyte 1938 patients randomized to cinacalcet, 1130 (58.3%) developed at
abnormalities. The overall pooled relative risks of AKI and all elec- least one hypocalcemia episode during the first 16 weeks, and 356
trolyte abnormalities in patients treated with PD-1 inhibitors were (18.4%) were categorized as severe hypocalcemia (77). Risk factors
1.86 (95% CI, 0.95 to 3.64) and 1.67 (95% CI, 0.89 to 3.12), for hypocalcemia were high PTH and alkaline phosphatase, as well
respectively. In a subanalysis using patients treated with non- as low serum calcium. Other studies have also identified high PTH
nephrotoxic drugs as controls, the pooled RR of AKI in patients as a risk factor for calcimimetic-associated hypocalcemia (78,79).
treated with PD-1 inhibitors was 4.19 (95% CI, 1.57 to 11.18). This observation raises the question of whether hypocalcemia is
PD-1 inhibitors were associated with hypocalcemia, with a pooled being triggered by a similar mechanism as seen in postparathyroi-
RR of 10.87 (95% CI, 1.40 to 84.16), and other specific electrolyte dectomy hungry bone syndrome, which should be treated with cal-
disorders were not associated to treatment with PD-1 inhibitors. cium and calcitriol supplementation, or if it is predominantly caused
The pooled estimated incidence rates of AKI and hypocalcemia in by reduced bone resorption related to more severe forms of hyper-
patients treated with PD-1 inhibitors were 2.2% (95% CI, 1.5 to parathyroidism, therefore more likely to ultimately need parathy-
3.0) and 1.0% (95% CI, 0.6 to 1.8), respectively. In addition, the roidectomy (80).
pooled estimated rate of interstitial nephritis was 16.6% (95% CI,
10.2 to 26.0). Several mechanisms have been hypothesized to medi- Dialysate Calcium
ate the associations of PD-1 inhibitors and electrolyte disturbances The 2017 KDIGO Guideline on CKD-MBD suggests the use of a
(64). Of, note, one study suggests that the incidence of hypocalce- dialysate calcium concentration between 1.25 and 1.50 mmol/L
mia reported with PD inhibitors may be related to the reporting of (2.5 and 3.0 mEq/L) in CKD stage 5D. The rationale for this rec-
serum calcium not corrected for serum albumin (65). ommendation is to reduce the positive balance of calcium that often
occurs during years of dialysis treatment and likely reduce vascular
Tyrosine Kinase Inhibitors calcification and low turnover bone disease (81), although large
Hypocalcemia is a recognized potential side effect of this class of studies with hard clinical outcomes are still lacking. However, one
drugs. According to a meta-analysis of seven studies, severe hypocal- drawback of using low calcium dialysate is the increased risk of
cemia is not uncommon, reaching almost 10% in patients that hypocalcemia (82), which can result in increased risk of cardiac
received sorafenib (66). Although the exact mechanism is not arrythmias, sudden death, and higher PTH. Brunelli et al. have eval-
known, it seems to be more frequent in patients with thyroid cancer uated the consequences of the conversion to low calcium dialysate
and those with previous, but compensated hypoparathyroidism (67). (,2.5 mEqL) in a hemodialysis organization in the US reporting
greater rates of hospitalization for decompensated heart failure,
Parathyroidectomy hypocalcemia, and intradialytic hypotension, with no differences in
Hypocalcemia is a well-known severe complication after parathy- all-cause mortality or total hospitalization rate (83). Hypocalcemia
roidectomy, particularly in severe hyperparathyroidism, with a need has also been associated with increased risk of hypotension in
for massive supplementation of calcium and vitamin D in the first CRRT (84). The conversion to lower calcium dialysate was associ-
weeks following surgery (hungry bone syndrome) (68–70). A recent ated with lower serum calcium levels, increased serum phosphate
meta-analysis done by Gao et al. compiling 2990 patients with and PTH levels, and increased use of phosphate binders, vitamin D,
secondary hyperparathyroidism from 13 studies has shown that pre- and calcimimetics (83). Similar biochemical and drug use trends
operative PTH, alkaline phosphatase and calcium were significant were reported elsewhere, whereas others have found 2.5 mEq/L-cal-
predictors of hypocalcemia (71). Similar findings were described in cium dialysate use was not associated with increased risk of hypocal-
a single-center, retrospective study, which analyzed 1500 CKD cemia when compared to the use of 3 mEq/L dialysate (85). Further
Nephrology Self-Assessment Program - Vol 22, No 1, April 2023 25

studies are necessary and should assess not only the biochemical submitted to this procedure, particularly those with increased age,
parameters but also hard clinical outcomes. CKD, and immobilization (91).

Silicone and Polymethylmethacrylate


Hypercalcemia Recently, hypercalcemia related to silicone and polymethylmetha-
Hypercalcemia is a common reason for nephrology consult and is crylate injections for cosmetic purposes has been described
most often related to underlying cancer, parathyroid disorders, and (92,93). A systematic review by Tachamo et al. identified 23
drugs. Rarely, it can be caused by genetic conditions and loss-of- patients with this condition. Polymethylmethacrylate induces an
function variants in CYP24A1 have been associated with inflammatory response with appearance of activated macrophages
24-hydroxylase deficiency with concurrent hypercalcemia, nephro- in granulomas that produce 1-alpha-hydroxylase, increasing extra-
lithiasis, and nephrocalcinosis (86). renal calcitriol production and leading to hypercalcemia, and fre-
quently, AKI (94). Although this procedure is not Food and Drug
Malignancy Administration approved, it is commonly used in Latin America
Usually, hypercalcemia is mediated by the production of PTH- and other countries (95).
related peptide (PTHrP, which accounts for 80% of the cases) or
due to calcium release from bone by osteolytic metastases. Less than Immobilization
1% of malignancy-related hypercalcemias are explained by increased Prolonged immobilization is another less common cause for hyper-
calcitriol or PTH. In the first case, there is some debate about which calcemia. However, its prevalence is likely increasing, considering
is the source of excessive calcitriol synthesis, the tumor cells or adja- the expansion in number of patients and hospitalization days at
cent macrophages. A recent published case report of a patient with ICUs worldwide. This entity was described decades ago in patients
diffuse large B cell lymphoma and hypercalcemia documented the immobilized by traumatic spinal-cord injury (96) but is also seen in
increased expression of 1a-hydroxylase by lymphoma cells, confirm- critically ill patients. It is characterized by calcium release from the
ing that these cells can produce the active form of vitamin D (87). skeleton and mediated by increased osteocyte synthesis of sclerostin,
In the late, parathyroid carcinoma, which accounts for less than 1% which simultaneously inhibits bone formation and stimulates bone
of primary hyperparathyroidism, is the source of PTH. A review of resorption (97,98). The massive release of calcium from the skeleton
1057 cases from the US National Cancer Database showed that, inhibits PTH, leading to the usual finding of suppressed PTH.
although rare, this is a disease with high morbidity and mortality Those who have normal renal function can increase calcium excre-
(10-year survival rate of 57%), and in many cases related to the tion and remain normocalcemic or show only very mild hypercalce-
degree of hypercalcemia (88). Finally, ectopic production of PTH, mia. Conversely, when patients have CKD or AKI develops, the
such as by ovarian carcinoma or neuroendocrine tumors, is reduced GFR impairs the ability to excrete calcium and hypercalce-
described in anecdotical cases (89). mia ensues. It is important to note that in patients who are on
continuous renal replacement therapy, particularly those using
Fungal Infection citrate-anticoagulation, calcium removal may mitigate or abolish
There are several case reports of pulmonary fungal infections (Pneu- the hypercalcemia, increasing the risk of underdiagnosis of the
mocystic jirovecii and Histoplasma capsulatum) in immunosuppressed immobilization syndrome (99,100). During the first wave of the
patients, accompanied by hypercalcemia. These patients present COVID-19 pandemic, several patients remained heavily sedated,
high serum levels of 1a,25-vitamin D and low PTH, a finding com- with neuromuscular blockage for weeks. Many of them also needed
patible with extra-renal synthesis of 1a,25-vitamin D (from the continuous renal replacement therapy. An impressive bone loss was
granulomas). In an interesting report of hypercalcemia, the authors documented in one study (101). A case report described a patient
described a case of an immunocompromised person affected by who presented hypercalcemia only after renal function recovery,
invasive muscular microsporidial infection. The microsporidia was emphasizing the need of a greater attention to bone loss in patients
surrounded by mononuclear inflammatory cells, mainly macro- with prolonged immobilization (102).
phages, likely the potential source of 1a,25 vitamin D (90).
References
Antibiotic Impregnated Calcium Sulfate Beads 1. Friedman PA: Mechanisms of renal calcium transport. Exp Nephrol 8:
343–350, 2000 10.1159/000020688 PubMed
There have been some reports of hypercalcemia after use of calcium 2. Muto S, Hata M, Taniguchi J, Tsuruoka S, Moriwaki K, Saitou M,
sulfate beads impregnated with antibiotics, which are occasionally et al.: Claudin-2-deficient mice are defective in the leaky and cation-
used for treatment of osteomyelitis. A retrospective analysis of an selective paracellular permeability properties of renal proximal tubules.
orthopedic service in Switzerland, evaluated 215 patients who were Proc Natl Acad Sci USA 107: 8011–8016, 2010 10.1073/pnas.
treated with antibiotic-loaded calcium sulfate beads. They found 0912901107 PubMed
symptomatic hypercalcemia in only two of them, and the increase in 3. Curry JN, Yu ASL: Paracellular calcium transport in the proximal
tubule and the formation of kidney stones. Am J Physiol Renal Physiol
serum calcium could also be explained by other factors, such as
316: F966–F969, 2019 10.1152/ajprenal.00519.2018 PubMed
immobilization and chronic kidney disease. However, serum cal- 4. Curry JN, Saurette M, Askari M, Pei L, Filla MB, Beggs MR, et al.:
cium was not systematically measured, and asymptomatic hypercal- Claudin-2 deficiency associates with hypercalciuria in mice and human
cemia could not be ruled out in many cases. Therefore, calcium kidney stone disease. J Clin Invest 130: 1948–1960, 2020 10.1172/
monitorization is recommended in those patients that will be JCI127750 PubMed
26 Nephrology Self-Assessment Program - Vol 22, No 1, April 2023

5. Simon DB, Lu Y, Choate KA, Velazquez H, Al-Sabban E, Praga M, 22. Bushinsky DA, Krieger NS: Effects of acid on bone. Kidney Int 101:
et al.: Paracellin-1, a renal tight junction protein required for paracellu- 1160–1170, 2022 10.1016/j.kint.2022.02.032 PubMed
lar Mg21 resorption. Science 285: 103–106, 1999 10.1126/science.285. 23. Imenez Silva PH, Katamesh-Benabbas C, Chan K, Pastor Arroyo EM,
5424.103 PubMed Kn€opfel T, Bettoni C, et al.: The proton-activated ovarian cancer
6. Konrad M, Schaller A, Seelow D, Pandey AV, Waldegger S, Lesslauer A, G protein-coupled receptor 1 (OGR1) is responsible for renal calcium
et al.: Mutations in the tight-junction gene claudin 19 (CLDN19) are loss during acidosis. Kidney Int 97: 920–933, 2020 10.1016/j.kint.
associated with renal magnesium wasting, renal failure, and severe ocular 2019.12.006 PubMed
involvement. Am J Hum Genet 79: 949–957, 2006 10.1086/508617 24. Khalil R, Kim NR, Jardi F, Vanderschueren D, Claessens F, Decallonne B:
PubMed Sex steroids and the kidney: Role in renal calcium and phosphate handling.
7. Gong Y, Renigunta V, Himmerkus N, Zhang J, Renigunta A, Bleich M, Mol Cell Endocrinol 465: 61–72, 2018 10.1016/j.mce.2017.11.011
et al.: Claudin-14 regulates renal Ca11 transport in response to CaSR PubMed
signalling via a novel microRNA pathway. EMBO J 31: 1999–2012, 25. Hsu YJ, Dimke H, Schoeber JP, Hsu SC, Lin SH, Chu P, et al.: Tes-
2012 10.1038/emboj.2012.49 PubMed tosterone increases urinary calcium excretion and inhibits expression of
8. Thorleifsson G, Holm H, Edvardsson V, Walters GB, Styrkarsdottir U, renal calcium transport proteins. Kidney Int 77: 601–608, 2010 10.
Gudbjartsson DF, et al.: Sequence variants in the CLDN14 gene associ- 1038/ki.2009.522 PubMed
ate with kidney stones and bone mineral density. Nat Genet 41: 926– 26. Lin PH, Jian CY, Chou JC, Chen CW, Chen CC, Soong C, et al.:
930, 2009 10.1038/ng.404 PubMed Induction of renal senescence marker protein-30 (SMP30) expression
9. Figueres L, Bruneau S, Prot-Bertoye C, Brideau G, Neel M, Griveau C, by testosterone and its contribution to urinary calcium absorption in
et al.: Hypomagnesemia, hypocalcemia, and tubulointerstitial nephropa- male rats. Sci Rep 6: 32085, 2016 10.1038/srep32085 PubMed
thy caused by claudin-16 autoantibodies. J Am Soc Nephrol 33: 1402– 27. Beggs MR, Appel I, Svenningsen P, Skjødt K, Alexander RT, Dimke H:
1410, 2022 10.1681/asn.2022010060 PubMed Expression of transcellular and paracellular calcium and magnesium trans-
10. Gong Y, Hou J: Claudins in barrier and transport function-the kidney. port proteins in renal and intestinal epithelia during lactation. Am J Phy-
Pflugers Arch 469: 105–113, 2017 10.1007/s00424-016-1906-6 PubMed siol Renal Physiol 313: F629–F640, 2017 10.1152/ajprenal.00680.2016
11. Marneros AG: Magnesium and calcium homeostasis depend on PubMed
KCTD1 function in the distal nephron. Cell Rep 34: 108616, 2021 10. 28. Rutkowski JM, Pastor J, Sun K, Park SK, Bobulescu IA, Chen CT,
1016/j.celrep.2020.108616 PubMed et al.: Adiponectin alters renal calcium and phosphate excretion through
12. Riccardi D, Valenti G: Localization and function of the renal calcium- regulation of klotho expression. Kidney Int 91: 324–337, 2017 10.
sensing receptor. Nat Rev Nephrol 12: 414–425, 2016 10.1038/nrneph. 1016/j.kint.2016.09.016 PubMed
2016.59 PubMed 29. Vall-Palomar M, Madariaga L, Ariceta G: Familial hypomagnesemia
13. Baz ua-Valenti S, Rojas-Vega L, Casta~ neda-Bueno M, Barrera-Chimal J, with hypercalciuria and nephrocalcinosis. Pediatr Nephrol 36: 3045–
Bautista R, Cervantes-Perez LG, et al.: The calcium-sensing receptor 3055, 2021 10.1007/s00467-021-04968-2 PubMed
increases activity of the renal NCC through the WNK4-SPAK pathway. 30. Vall-Palomar M, Burballa C, Claverie-Martın F, Meseguer A, Ariceta G:
J Am Soc Nephrol 29: 1838–1848, 2018 10.1681/ASN.2017111155 Heterogeneity is a common ground in familial hypomagnesemia with
PubMed hypercalciuria and nephrocalcinosis caused by CLDN19 gene muta-
14. van Megen WH, Tan RSG, Alexander RT, Dimke H: Differential para- tions. J Nephrol 34: 2053–2062, 2021 10.1007/s40620-021-01054-6
thyroid and kidney Ca21-sensing receptor activation in autosomal dom- PubMed
inant hypocalcemia 1. EBioMedicine 78: 103947, 2022 10.1016/j. 31. Hou J, Renigunta V, Nie M, Sunq A, Himmerkus N, Quintanova C,
ebiom.2022.103947 PubMed et al.: Phosphorylated claudin-16 interacts with Trpv5 and regulates
15. Stevenson M, Pagnamenta AT, Mack HG, et al.: The Bartter-Gitelman transcellular calcium transport in the kidney. Proc Natl Acad Sci USA
spectrum: 50-year follow-up with revision of diagnosis after whole- 116: 19176–19186, 2019 10.1073/pnas.1902042116 PubMed
genome sequencing. J Endocr Soc 6: bvac079, 2022 10.1210/jendso/ 32. Kang TS, Siegel LM: Congenital macular scars in siblings from
bvac079PubMed CLDN19 mutations. J AAPOS 25: 316–318, 2021 10.1016/j.jaapos.
16. Talmage RV, Matthews JL, Mobley HT, Lester GE: Calcium homeo- 2021.05.008 PubMed
stasis and bone surface proteins, a postulated vital process for plasma 33. Eckardt KU, Alper SL, Antignac C, Bleyer AJ, Chauveau D, Dahan K,
calcium control. J Musculoskelet Neuronal Interact 3: 194–200, 2003 et al; Kidney Disease: Improving Global Outcomes: Autosomal domi-
PubMed nant tubulointerstitial kidney disease: Diagnosis, classification, and
17. Andrukhova O, Smorodchenko A, Egerbacher M, Streicher C, Zeitz U, management: A KDIGO consensus report. Kidney Int 88: 676–683,
Goetz R, et al.: FGF23 promotes renal calcium reabsorption through 2015 10.1038/ki.2015.28 PubMed
the TRPV5 channel. EMBO J 33: 229–246, 2014 10.1002/embj. 34. Kompatscher A, de Baaij JHF, Aboudehen K, Farahani S, van Son
201284188 PubMed LHJ, Milatz S, et al.: Transcription factor HNF1b regulates expression
18. Renkema KY, Nijenhuis T, van der Eerden BC, van der Kemp AW, of the calcium-sensing receptor in the thick ascending limb of the kid-
Weinans H, van Leeuwen JP, et al.: Hypervitaminosis D mediates com- ney. Am J Physiol Renal Physiol 315: F27–F35, 2018 10.1152/ajprenal.
pensatory Ca21 hyperabsorption in TRPV5 knockout mice. J Am Soc 00601.2017 PubMed
Nephrol 16: 3188–3195, 2005 10.1681/ASN.2005060632 PubMed 35.  a M, Bhadra P, Baresova V, Tirincsi A, Hadzibeganovic D,
Sicking M, Zivn
19. Baron R, Kneissel M: WNT signaling in bone homeostasis and disease: et al.: Phenylbutyrate rescues the transport defect of the Sec61a mutations
From human mutations to treatments. Nat Med 19: 179–192, 2013 10. V67G and T185A for renin. Life Sci Alliance 5: e202101150, 2022 10.
1038/nm.3074 PubMed 26508/lsa.202101150 PubMed
20. Ryan ZC, Ketha H, McNulty MS, McGee-Lawrence M, Craig TA, 36. Wu M, Feng Y, Ye GX, Han YC, Wang SS, Ni HF, et al.: Calcium-
Grande JP, et al.: Sclerostin alters serum vitamin D metabolite and sensing receptor activation attenuates collagen expression in renal proxi-
fibroblast growth factor 23 concentrations and the urinary excretion of mal tubular epithelial cells. Am J Physiol Renal Physiol 316: F1006–
calcium. Proc Natl Acad Sci USA 110: 6199–6204, 2013 10.1073/pnas. F1015, 2019 10.1152/ajprenal.00413.2018 PubMed
1221255110 PubMed 37. Mai X, Shang J, Liang S, Yu B, Yuan J, Lin Y, et al.: Blockade of orai1
21. van der Wijst J, Tutakhel OAZ, Bos C, Danser AHJ, Hoorn EJ, Hoen- store-operated calcium entry protects against renal fibrosis. J Am Soc
derop JGJ, et al.: Effects of a high-sodium/low-potassium diet on renal Nephrol 27: 3063–3078, 2016 10.1681/ASN.2015080889 PubMed
calcium, magnesium, and phosphate handling. Am J Physiol Renal Phy- 38. Hering L, Rahman M, Hoch H, Marko L, Yang G, Reil A, et al.: a2A-
siol 315: F110–F122, 2018 10.1152/ajprenal.00379.2017 PubMed adrenoceptors modulate renal sympathetic neurotransmission and
Nephrology Self-Assessment Program - Vol 22, No 1, April 2023 27

protect against hypertensive kidney disease. J Am Soc Nephrol 31: 783– 56. Miyaoka D, Imanishi Y, Ohara M, Hayashi N, Nagata Y, Yamada S,
798, 2020 10.1681/ASN.2019060599 PubMed et al.: Impaired residual renal function predicts denosumab-induced
39. Mishima K, Nakasatomi M, Takahashi S, Ikeuchi H, Sakairi T, Kaneko Y, serum calcium decrement as well as increment of bone mineral density
et al.: Attenuation of renal fibrosis after unilateral ureteral obstruction in in non-severe renal insufficiency. Osteoporos Int 30: 241–249, 2019 10.
mice lacking the N-type calcium channel. PLoS One 14: e0223496, 2019 1007/s00198-018-4688-1 PubMed
10.1371/journal.pone.0223496 PubMed 57. Broadwell A, Chines A, Ebeling PR, Franek E, Huang S, Smith S, et al.:
40. Zhao X, Kong Y, Liang B, Xu J, Lin Y, Zhou N, et al.: Mechanosensi- Denosumab safety and efficacy among participants in the FREEDOM
tive Piezo1 channels mediate renal fibrosis. JCI Insight 7: e152330, extension study with mild to moderate chronic kidney disease. J Clin
2022 10.1172/jci.insight.152330 PubMed Endocrinol Metab 106: 397–409, 2021 10.1210/clinem/dgaa851
41. Liu X, Tang J, Chen XZ: Role of PKD2 in the endoplasmic reticulum PubMed
calcium homeostasis. Front Physiol 13: 962571, 2022 10.3389/fphys. 58. Hiramatsu R, Ubara Y, Sawa N, Sakai A: Hypocalcemia and bone min-
2022.962571 PubMed eral changes in hemodialysis patients with low bone mass treated with
42. Di Mise A, Tamma G, Ranieri M, Centrone M, van den Heuvel L, denosumab: A 2-year observational study. Nephrol Dial Transplant 36:
Mekahli D, et al.: Activation of calcium-sensing receptor increases intra- 1900–1907, 2021 10.1093/ndt/gfaa359 PubMed
cellular calcium and decreases cAMP and mTOR in PKD1 deficient 59. Thongprayoon C, Acharya P, Acharya C, Chenbhanich J, Bathini T,
cells. Sci Rep 8: 5704, 2018 10.1038/s41598-018-23732-5 PubMed Boonpheng B, et al.: Hypocalcemia and bone mineral density changes
43. Di Mise A, Ranieri M, Centrone M, Venneri M, Tamma G, Valenti D, following denosumab treatment in end-stage renal disease patients: A
et al.: Activation of the calcium-sensing receptor corrects the impaired meta-analysis of observational studies. Osteoporos Int 29: 1737–1745,
mitochondrial energy status observed in renal polycystin-1 knockdown 2018 10.1007/s00198-018-4533-6 PubMed
cells modeling autosomal dominant polycystic kidney disease. Front Mol 60. Thongprayoon C, Acharya P, Aeddula NR, Torres-Ortiz A, Bathini T,
Biosci 5: 77, 2018 10.3389/fmolb.2018.00077 PubMed Sharma K, et al.: Effects of denosumab on bone metabolism and bone
44. Li Z, Zhou J, Li Y, Yang F, Lian X, Liu W: Mitochondrial TRPC3 mineral density in kidney transplant patients: A systematic review and
promotes cell proliferation by regulating the mitochondrial calcium and meta-analysis. Arch Osteoporos 14: 35, 2019 10.1007/s11657-019-0587-0
metabolism in renal polycystin-2 knockdown cells. Int Urol Nephrol 51: PubMed
1059–1070, 2019 10.1007/s11255-019-02149-7 PubMed 61. Nanmoku K, Shinzato T, Kubo T, Shimizu T, Yagisawa T: Effects of
45. Di Mise A, Wang X, Ye H, Pellegrini L, Torres VE, Valenti G: Pre- denosumab on hypercalcemia and bone mineral density loss in kidney
clinical evaluation of dual targeting of the GPCRs CaSR and V2R as transplant recipients. Clin Nephrol 92: 1–8, 2019 10.5414/CN109723
therapeutic strategy for autosomal dominant polycystic kidney disease. PubMed
FASEB J 35: e21874, 2021 10.1096/fj.202100774R PubMed 62. Kanbayashi Y, Sakaguchi K, Hongo F, Ishikawa T, Tabuchi Y, Uki-
46. Ning B, Guo C, Kong A, Li K, Xie Y, Shi H, et al.: Calcium signaling mura O, et al.: Predictors for development of denosumab-induced
mediates cell death and crosstalk with autophagy in kidney disease. Cells hypocalcaemia in cancer patients with bone metastases determined by
10: 3204, 2021 10.3390/cells10113204 PubMed ordered logistic regression analysis. Sci Rep 11: 978, 2021 10.1038/
47. Ilatovskaya DV, Blass G, Palygin O, Levchenko V, Pavlov TS, Grzy- s41598-020-80243-y PubMed
bowski MN, et al.: A NOX4/TRPC6 pathway in podocyte calcium reg- 63. Manohar S, Kompotiatis P, Thongprayoon C, Cheungpasitporn W,
ulation and renal damage in diabetic kidney disease. J Am Soc Nephrol Herrmann J, Herrmann SM: Programmed cell death protein 1 inhibitor
29: 1917–1927, 2018 10.1681/ASN.2018030280 PubMed treatment is associated with acute kidney injury and hypocalcemia:
48. Palygin O, Klemens CA, Isaeva E, Levchenko V, Spires DR, Dissa- Meta-analysis. Nephrol Dial Transplant 34: 108–117, 2019 10.1093/
nayake LV, et al.: Characterization of purinergic receptor 2 signaling in ndt/gfy105 PubMed
podocytes from diabetic kidneys. iScience 24: 102528, 2021 10.1016/j. 64. Uppal NN, Workeneh BT, Rondon-Berrios H, Jhaveri KD: Electrolyte
isci.2021.102528 PubMed and acid-base disorders associated with cancer immunotherapy. Clin J
49. Vasco RFV, Takayama L, Pereira RMR, Moyses RMA, Elias RM: Am Soc Nephrol 17: 922–933, 2022 10.2215/cjn.14671121 PubMed
Effects of diuretics furosemide and hydrochlorothiazide on CKD-MBD: 65. Nalluru SS, Piranavan P, Ning Y, Ackula H, Siddiqui AD, Trivedi N:
A prospective randomized study. Bone Rep 14: 100746, 2021 10.1016/ Hypocalcemia with immune checkpoint inhibitors: The disparity
j.bonr.2021.100746 PubMed among various reports. Int J Endocrinol 2020: 7459268, 2020 10.1155/
50. Shroff R, Lalayiannis AD, Fewtrell M, Schmitt CP, Bayazit A, Askiti V, 2020/7459268 PubMed
et al.: Naturally occurring stable calcium isotope ratios are a novel bio- 66. Yu ST, Ge JN, Luo JY, Wei ZG, Sun BH, Lei ST: Treatment-related
marker of bone calcium balance in chronic kidney disease. Kidney Int adverse effects with TKIs in patients with advanced or radioiodine
102: 613–623, 2022 10.1016/j.kint.2022.04.024 PubMed refractory differentiated thyroid carcinoma: A systematic review and
51. Das S, Clezardin P, Kamel S, Brazier M, Mentaverri R: The CaSR in meta-analysis. Cancer Manag Res 11: 1525–1532, 2019 10.2147/cmar.
pathogenesis of breast cancer: A new target for early stage bone metasta- S191499 PubMed
ses. Front Oncol 10: 69, 2020 10.3389/fonc.2020.00069 PubMed 67. Berends AMA, van der Horst-Schrivers ANA, Oosting SF, Kapiteijn EW,
52. Jeong S, Kim JH, Kim MG, Han N, Kim IW, Kim T, et al.: Genetic de Groot JWB, Links TP: Hypocalcemia induced by tyrosine kinase inhi-
polymorphisms of CASR and cancer risk: Evidence from meta-analysis bitors: Targeted treatment with ‘untargeted’ side effects. Acta Oncol 59:
and HuGE review. OncoTargets Ther 9: 655–669, 2016 10.2147/ott. 726–729, 2020 10.1080/0284186x.2020.1726455 PubMed
S97602 PubMed 68. Ho LY, Wong PN, Sin HK, Wong YY, Lo KC, Chan SF, et al.: Risk
53. Ordu~ na-Castillo LB, Del-Rıo-Robles JE, Garcıa-Jimenez I, Zavala- factors and clinical course of hungry bone syndrome after total parathy-
Barrera C, Beltran-Navarro YM, Hidalgo-Moyle JJ, et al.: Calcium roidectomy in dialysis patients with secondary hyperparathyroidism.
sensing receptor stimulates breast cancer cell migration via the Gbg- BMC Nephrol 18: 12, 2017 10.1186/s12882-016-0421-5 PubMed
AKT-mTORC2 signaling pathway. J Cell Commun Signal 16: 239–252, 69. Ge Y, Yang G, Wang N, Zha X, Yu X, Mao H, et al.: Bone metabolism
2022 10.1007/s12079-021-00662-y PubMed markers and hungry bone syndrome after parathyroidectomy in dialysis
54. Frees S, Breuksch I, Haber T, Bauer HK, Chavez-Munoz C, Raven P, patients with secondary hyperparathyroidism. Int Urol Nephrol 51:
et al.: Calcium-sensing receptor (CaSR) promotes development of bone 1443–1449, 2019 10.1007/s11255-019-02217-y PubMed
metastasis in renal cell carcinoma. Oncotarget 9: 15766–15779, 2018 70. Jain N, Reilly RF: Hungry bone syndrome. Curr Opin Nephrol Hyper-
10.18632/oncotarget.24607 PubMed tens 26: 250–255, 2017 10.1097/MNH.0000000000000327 PubMed
55. Rosner MH: Hypocalcemia in a patient with cancer. Clin J Am Soc 71. Gao D, Lou Y, Cui Y, Liu S, Cui W, Sun G: Risk factors for hypocal-
Nephrol 12: 696–699, 2017 10.2215/cjn.13241216 PubMed cemia in dialysis patients with refractory secondary hyperparathyroidism
28 Nephrology Self-Assessment Program - Vol 22, No 1, April 2023

after parathyroidectomy: A meta-analysis. Ren Fail 44: 503–512, 2022 D-mediated hypercalcemia disorders. J Endocr Soc 5: bvab119, 2021 10.
10.1080/0886022x.2022.2048856 PubMed 1210/jendso/bvab119 PubMed
72. Zhao S, Gan W, Xie W, Cao J, Zhang L, Wen P, et al.: A single-center 87. Ogawa M, Morikawa M, Kobatake M, Murakami T, Yamamoto Y,
experience of parathyroidectomy in 1500 cases for secondary hyperpara- Watanabe R, et al.: Hypercalcemia associated with the ectopic expression
thyroidism: A retrospective study. Ren Fail 44: 23–29, 2022 10.1080/ of 25-hydroxyvitamin D3-1a-hydroxylase in diffuse large B-cell lym-
0886022x.2021.2016445 PubMed phoma. Intern Med 61: 2489–2495, 2022 10.2169/internalmedicine.
73. Sun Y, Tian B, Sheng Z, Wan P, Xu T, Yao L: Efficacy and safety of 8933-21 PubMed
cinacalcet compared with other treatments for secondary hyperparathy- 88. Goldner E, Fingeret A: Parathyroid carcinoma: A National Cancer
roidism in patients with chronic kidney disease or end-stage renal dis- Database analysis. J Surg Res 281: 57–62, 2023 10.1016/j.jss.2022.08.
ease: A meta-analysis. BMC Nephrol 21: 316, 2020 10.1186/s12882- 017 PubMed
019-1639-9 PubMed 89. Quinn CD, Chaudhary F, Gould-Simon A, Chen B, Bhandal HS,
74. Bernardor J, Flammier S, Salles JP, Amouroux C, Castanet M, Lien- Chaudhary U: A case of a rare parathyroid hormone (PTH)-producing
hardt A, et al.: Off-label use of cinacalcet in pediatric primary hyper- neuroendocrine tumor. Am J Case Rep 23: e935783, 2022 10.12659/
parathyroidism: A French multicenter experience. Front Pediatr 10: ajcr.935783 PubMed
926986, 2022 10.3389/fped.2022.926986 PubMed 90. Watson AL, Matic M, Robertson T, Stewart AGA: Microsporidial myo-
75. Sheerah AA, Al-Ahmed RA, El-Desoky SM, Alhasan KA, Albanna AS, sitis, keratitis and hypercalcaemia in a cystic fibrosis lung transplant
Shalaby MA, et al.: Cinacalcet for severe secondary hyperparathyroidism recipient. BMJ Case Rep 15: e250643, 2022 10.1136/bcr-2022-250643
in children with end-stage kidney disease. Saudi J Kidney Dis Transpl PubMed
32: 1628–1636, 2021 10.4103/1319-2442.352423 PubMed 91. Vallon F, Meier C, Gautier E, Wahl P: The incidence of severe
76. Wang AX, Liu S, Montez-Rath ME, Chertow GM, Lenihan CR: Para- hypercalcaemia-induced mental status changes in patients treated with
thyroidectomy and cinacalcet use in medicare-insured kidney transplant antibiotic-loaded calcium sulphate depot for orthopaedic infections.
recipients. Am J Kidney Dis 81: 270–280.e1, 2023 10.1053/j.ajkd.2022. J Clin Med 11: 4900, 2022 10.3390/jcm11164900 PubMed
07.015 PubMed 92. Friedmann DP, Kurian A, Fitzpatrick RE: Delayed granulomatous reac-
77. Floege J, Tsirtsonis K, Iles J, Drueke TB, Chertow GM, Parfrey P: Inci- tions to facial cosmetic injections of polymethylmethacrylate micro-
dence, predictors and therapeutic consequences of hypocalcemia in spheres and liquid injectable silicone: A case series. J Cosmet Laser Ther
patients treated with cinacalcet in the EVOLVE trial. Kidney Int 93: 18: 170–173, 2016 10.3109/14764172.2015.1114642 PubMed
1475–1482, 2018 10.1016/j.kint.2017.12.014 PubMed 93. Manfro AG, Lutzky M, Dora JM, Kalil MAS, Manfro RC: Case reports
78. Palmer SC, Mavridis D, Johnson DW, Tonelli M, Ruospo M, Strippoli of hypercalcemia and chronic renal disease due to cosmetic injections of
GFM: Comparative effectiveness of calcimimetic agents for secondary polymethylmethacrylate (PMMA). J Bras Nefrol. Aug 10 2020;doi:10.
hyperparathyroidism in adults: A systematic review and network meta- 1590/2175-8239-JBN-2020-0044
analysis. Am J Kidney Dis 76: 321–330, 2020 10.1053/j.ajkd.2020.02. 94. Tachamo N, Donato A, Timilsina B, Nazir S, Lohani S, Dhital R, et al.:
439 PubMed Hypercalcemia associated with cosmetic injections: A systematic review.
79. Louie KS, Erhard C, Wheeler DC, Stenvinkel P, Fouqueray B, Floege J: Eur J Endocrinol 178: 425–430, 2018 10.1530/EJE-17-0938 PubMed
Cinacalcet-induced hypocalcemia in a cohort of European haemodialysis 95. Manfro AG, Lutzky M, Dora JM, Kalil MAS, Manfro RC: Case reports
patients: Predictors, therapeutic approaches and outcomes. J Nephrol 33: of hypercalcemia and chronic renal disease due to cosmetic injections of
803–816, 2020 10.1007/s40620-019-00686-z PubMed polymethylmethacrylate (PMMA). J Bras Nefrol 43: 288–292, 2021 10.
80. Evenepoel P, Shroff R: Facing cinacalcet-induced hypocalcemia: Sit 1590/2175-8239-jbn-2020-0044 PubMed
back and relax? Kidney Int 93: 1275–1277, 2018 10.1016/j.kint.2018. 96. Stewart AF, Adler M, Byers CM, Segre GV, Broadus AE: Calcium
01.038 PubMed homeostasis in immobilization: An example of resorptive hypercalciuria.
81. Ok E, Asci G, Bayraktaroglu S, Toz H, Ozkahya M, Yilmaz M, et al.: N Engl J Med 306: 1136–1140, 1982 10.1056/NEJM198205133061903
Reduction of dialysate calcium level reduces progression of coronary PubMed
artery calcification and improves low bone turnover in patients on 97. Spatz JM, Fields EE, Yu EW, Divieti Pajevic P, Bouxsein ML, Sibonga JD,
hemodialysis. J Am Soc Nephrol 27: 2475–2486, 2016 10.1681/ASN. et al.: Serum sclerostin increases in healthy adult men during bed rest.
2015030268 PubMed J Clin Endocrinol Metab 97: E1736–E1740, 2012 10.1210/jc.2012-1579
82. Yoshikawa M, Takase O, Tsujimura T, Sano E, Hayashi M, Takato T, PubMed
et al.: Long-term effects of low calcium dialysates on the serum calcium 98. Osipov B, Emami AJ, Christiansen BA: Systemic bone loss after frac-
levels during maintenance hemodialysis treatments: A systematic review ture. Clin Rev Bone Miner Metab 16: 116–130, 2018 10.1007/s12018-
and meta-analysis. Sci Rep 8: 5310, 2018 10.1038/s41598-018-23658-y 018-9253-0 PubMed
PubMed 99. Wang PL, Meyer MM, Orloff SL, Anderson S: Bone resorption and
83. Brunelli SM, Sibbel S, Do TP, Cooper K, Bradbury BD: Facility dialy- “relative” immobilization hypercalcemia with prolonged continuous
sate calcium practices and clinical outcomes among patients receiving renal replacement therapy and citrate anticoagulation. Am J Kidney Dis
hemodialysis: A retrospective observational study. Am J Kidney Dis 66: 44: 1110–1114, 2004 10.1053/j.ajkd.2004.09.001 PubMed
655–665, 2015 10.1053/j.ajkd.2015.03.038 PubMed 100. Madureira RM, Callas SH, Caires RA, Ferraz Crispilho S, Ayroza
84. Kelly YP, Sharma S, Mothi SS, McCausland FR, Mendu ML, McMa- Galv~ao PC, Moyses RMA: Continuous renal replacement therapy might
hon GM, et al.: Hypocalcemia is associated with hypotension during mask immobilization-induced hypercalcemia in critically ill patients.
CRRT: A secondary analysis of the Acute Renal Failure Trial Network Blood Purif 49: 129–131, 2020 10.1159/000502679 PubMed
Study. J Crit Care 65: 261–267, 2021 10.1016/j.jcrc.2021.07.008 101. Berktaş BM, G€okçek A, Hoca NT, Koyuncu A: COVID-19 illness and
PubMed treatment decrease bone mineral density of surviving hospitalized
85. Wen Y, Gan H, Li Z, Sun X, Xiong Y, Xia Y: Safety of low-calcium patients. Eur Rev Med Pharmacol Sci 26: 3046–3056, 2022 10.26355/
dialysate and its effects on coronary artery calcification in patients eurrev_202204_28636 PubMed
undergoing maintenance hemodialysis. Sci Rep 8: 5941, 2018 10.1038/ 102. Mesland JB, Collienne C, Laterre PF, Hantson P: Immobilization-
s41598-018-24397-w PubMed related hypercalcemia in a COVID-19 patient with prolonged intensive
86. Azer SM, Vaughan LE, Tebben PJ, Sas DJ. 24-Hydroxylase deficiency care unit stay. Am J Phys Med Rehabil 101: 61–63, 2022 10.1097/phm.
due to CYP24A1 Sequence variants: Comparison with other vitamin 0000000000001907 PubMed

You might also like