Comprehensive Physiology Wiley Online Library

Calcium Homeostasis in Health and in Kidney Disease

Full Article on Wiley Online Library



ABSTRACT

Calcium is an important ion in cell signaling, hormone regulation, and bone health. Its regulation is complex and intimately connected to that of phosphate homeostasis. Both ions are maintained at appropriate levels to maintain the extracellular to intracellular gradients, allow for mineralization of bone, and to prevent extra skeletal and urinary calcification. The homeostasis involves the target organs intestine, parathyroid glands, kidney, and bone. Multiple hormones converge to regulate the extracellular calcium level: parathyroid hormone, vitamin D (principally 25(OH)D or 1,25(OH)2D), fibroblast growth factor 23, and α‐klotho. Fine regulation of calcium homeostasis occurs in the thick ascending limb and collecting tubule segments via actions of the calcium sensing receptor and several channels/transporters. The kidney participates in homeostatic loops with bone, intestine, and parathyroid glands. Initially in the course of progressive kidney disease, the homeostatic response maintains serum levels of calcium and phosphorus in the desired range, and maintains neutral balance. However, once the kidneys are no longer able to appropriately respond to hormones and excrete calcium and phosphate, positive balance ensues leading to adverse cardiac and skeletal abnormalities. © 2016 American Physiological Society. Compr Physiol 6:1781‐1800, 2016.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1. Normal calcium balance. The average dietary intake of calcium is 20 mmol of which 16 mmol is excreted in stool. A net of approximately 4 mmol is absorbed (accounting for the obligatory secretion). With a normal glomerular filtration rate, the kidney filters approximately 270 mmol (10 g), and reabsorbs 98%. In neutral balance, the assumption is that the calcium entering the bone and leaving the bone is in steady state, and therefore the kidney maintains neutral balance by excreting any excess calcium.
Figure 2. Figure 2. Regulation of parathyroid hormone secretion and metabolism. Secondary hyperparathyroidism induced acutely by reducing calcium concentration or chronically by reducing calcium and vitamin D intake leads to an increase in PTH(1‐84), a relative decrease in C‐PTH fragment formation, and a lower C‐PTH fragments/PTH(1‐84) ratio. Half parathyroidectomy in dogs leads to a similar situation by decreasing PTH(1‐84) less than C‐PTH fragments. Acute calcium infusion, correction of secondary hyperparathyroidism or injection of 1,25(OH)2D without changing calcium concentration leads to the reverse situation with a high C‐PTH fragments/PTH(1‐84) ratio. Reprinted, with permission, from (46), Figure 2.
Figure 3. Figure 3. Overview of vitamin D metabolism. Vitamin D is obtained from dietary sources, and is metabolized via UVB from 7‐DHC in the skin. Both sources (diet and skin) of vitamin D2 and vitamin D3 bind to DBP and circulate to the liver. In the liver, vitamin D is hydroxylated by CYP27A1 to 25(OH)D, commonly referred to as calcidiol. Calcidiol is then further metabolized to calcitriol by the 1‐αhydroxylase enzyme (CYP27B1) at the level of the kidney. The active metabolite 1,25(OH)2D (calcitriol) acts principally on the target organs of intestine, parathyroid gland, bone cell precursors, and the kidney. Calcitriol is metabolized to the inert 1,24,25(OH)3D through the action of the 24,25‐hydroxylase enzyme (CYP24). Calcidiol is similarly hydroxylated to 24,25(OH)2D (134).
Figure 4. Figure 4. The bone‐kidney‐parathyroid endocrine axes mediated by FGF23 and klotho. Active form of vitamin D (1,25‐dihydroxyvitamin D3) binds to VDR in the bone (osteocytes). The ligand‐bound VDR forms a heterodimer with a nuclear receptor RXR and transactivates expression of the FGF23 gene. FGF23 secreted from bone acts on the klotho‐FGFR complex expressed in the kidney (the bone‐kidney axis) and parathyroid gland (the bone‐parathyroid axis). In the kidney, FGF23 suppresses synthesis of active vitamin D by down‐regulating expression of the Cyp27b1 gene and promotes its inactivation by upregulating expression of the Cyp24 gene, thereby closing a negative feedback loop for vitamin D homeostasis. In the parathyroid gland, FGF23 suppresses production and secretion of PTH. PTH binds to PTHR expressed on renal tubular cells, leading to upregulation of the Cyp27b1 gene expression. Thus, suppression of PTH by FGF23 reduces expression of the Cyp27b1 gene and serum levels of 1,25‐dihydroxyvitamin D3. This closes another long negative feedback loop for vitamin D homeostasis. Reprinted, with permission, from (106).
Figure 5. Figure 5. Location of CaSR in the kidney. CaSR expression along the nephron. For each segment (Glom, glomerulus; MTALH, medullary thick ascending limb of Henle; CTALH, cortical thick ascending limb of Henle; and MD, macula densa; ), the location within the cells of each segment is shown along with the physiologic targets and cell‐specific functions of the CaSR. Localization in red indicates that morphologic studies do not uniformly recognize the CaSR in these locations, but that supporting functional data exist. Localization in green indicates that most or all studies find the CaSR in this location and that physiologic data support the localization. Podo, podocyte; intercal, intercalated. Reprinted, with permission, from (199), Figure 1.
Figure 6. Figure 6. Functions of the CaSR in the thick ascending limb of Henle's loop. The apical electroneutral NKCC2 transporter and ROMK channel (which is the rate‐limiting step for apical K+ recycling) generate a lumen‐positive transepithelial potential difference. This creates the driving force for nonselective, paracellular reabsorption of Ca2 cations. The paracellular transport is through a complex of claudin 16, 19 that is inhibited through physical interaction by a third tight junction protein claudin 14. Activation of the CaSR on the basolateral side of the cell increases claudin 14 expression (yellow) through NFAT signaling, and thus inhibits paracellular transport. Furthermore, activation of the CaSR also inhibits the ROMK channel to further inhibit calcium reabsorption in the setting of hypercalemia, although the mechanism is not understood. Reprinted, with permission, from (195), Figure 1.
Figure 7. Figure 7. Calcium active transport in the DCT. The late part of the DCT and CNT play an important role in fine‐tuning renal excretion of Ca2+. The ephosphatethelial Ca2+ channel (TRPV5) is primarily expressed aphosphatecally in these segments and co‐localizes with calbindin‐D28K (28K), Na+/Ca2+ exchanger (NCX1), and the plasma membrane ATPase (PMCA1b). Upon entry via TRPV5, Ca2+ is buffered by 28K and diffuses to the basolateral membrane, where it is released and extruded by a concerted action of NCX1 and PMCA1b. In addition, the basolateral membrane exposes a parathyroid hormone receptor (PTHR) and the Na+/K+‐ATPase consisting of the α‐, β‐, and γ‐subunit. PTHR activation by PTH stimulates TRPV5 activity, and entered Ca2+ can subsequently control the expression level of the Ca2+ transporters. At the aphosphatecal membrane, there is a bradykinin receptor (BK2) that is activated by urinary TK to activate TRPV5‐mediated Ca2+ influx. In the cell, entered Ca2+ acts as a negative feedback on channel activity, and 28K plays a regulatory role by association with TRPV5 under low intracellular Ca2+ concentrations. Extracellular urinary klotho directly stimulates TRPV5 at the aphosphatecal membrane by modification of the N‐glycan, whereas intracellular klotho enhances Na+/K+‐ATPase surface expression that in turn activates NCX1‐mediated Ca2+ efflux. Reprinted, with permission, from (19).
Figure 8. Figure 8. Physiologic roles of klotho on solute channels and transporters and vitamin D metabolism in the kidney klotho is prominently expressed in DCTs, and less in the proximal convoluted tubules (PCT). In PCT, membrane klotho at the basolateral side functions as coreceptor of FGFRs and drive FGF23 signal transduction to inhibit NaPi cotransporters (NaPi: 2a/c and Pit2) and to suppress cyp27β1 encoding for 1‐hydroxylase,and to stimulate cyp24α1 encoding for 24‐hydoxylase. The role of klotho in the cytoplasm of renal tubules is unclear. Whether membrane klotho at luminal side inhibits NaPi cotransporters (2a/c and Pit2) in an autocrine mode is not known (dash line). In DCT, membrane klotho at basolateral side4 functions as coreceptor of FGFRs to induce FGF23 signal transduction. What intermediate(s) are released from DCT and how intermediate(s) affect on PCT in paracrine mode is not known. One possible candidate is klotho release from the DCT to act on the PCT. Whether membrane klotho at luminal side directly regulates TRPV5 in autocrine mode is unknown. Soluble Klotho in luminal urine derived from either blood or urine exerts regulatory action on NaPi cotransporters in PCT; and on TRPV5 in DCT. Ca: calcium ion; FGF23: NaPi cotransporter: sodium‐phosphate‐dependent cotransporter; Pi: phosphate; 1,25 VD3: 1,25‐(OH)2 vitamin D3, Dash line: suspected action. Reprinted, with permission, from (89).
Figure 9. Figure 9. Overview of calcium and phosphate homeostasis. As (phosphate) levels increase (or there is a chronic phosphate load), both PTH and FGF23 are increased. Both the elevated PTH and FGF23 increase urinary phosphate excretion. The two hormones differ in respect to their effects on the vitamin D axis. PTH stimulates 1‐alpha hydroxylase activity thereby increasing the production of 1,25(OH)2D, which in turn negatively feeds back on the parathyroid gland to decrease PTH secretion. In contrast, FGF23 inhibits 1‐alpha hydroxylase activity, thereby decreasing the production of 1,25(OH)2D feeding back to stimulate further secretion of FGF23. FGF23 and PTH also regulate each other. Finally, low calcium levels stimulate PTH, whereas high calcium levels stimulate FGF23. Lastly, there is some evidence that FGF23 also inhibits PTH secretion (solid line = stimulates; dashed line = inhibits). Reprinted, with permission, from Moe SM, Sprague SM. Chronic kidney disease‐mineral bone disorder (134).
Figure 10. Figure 10. Calcium balance in stage 3/4 CKD patients with and without calcium carbonate. Calcium balance was greater with calcium carbonate compared with placebo. Ca intake was experimentally controlled and statistical analysis does not apply. White bars = placebo; Black bars = calcium carbonate; Ca = calcium; NS = not significant (P > 0.05). Data are presented as least squares mean ± pooled SEM. Reprinted, with permission, from (85).


Figure 1. Normal calcium balance. The average dietary intake of calcium is 20 mmol of which 16 mmol is excreted in stool. A net of approximately 4 mmol is absorbed (accounting for the obligatory secretion). With a normal glomerular filtration rate, the kidney filters approximately 270 mmol (10 g), and reabsorbs 98%. In neutral balance, the assumption is that the calcium entering the bone and leaving the bone is in steady state, and therefore the kidney maintains neutral balance by excreting any excess calcium.


Figure 2. Regulation of parathyroid hormone secretion and metabolism. Secondary hyperparathyroidism induced acutely by reducing calcium concentration or chronically by reducing calcium and vitamin D intake leads to an increase in PTH(1‐84), a relative decrease in C‐PTH fragment formation, and a lower C‐PTH fragments/PTH(1‐84) ratio. Half parathyroidectomy in dogs leads to a similar situation by decreasing PTH(1‐84) less than C‐PTH fragments. Acute calcium infusion, correction of secondary hyperparathyroidism or injection of 1,25(OH)2D without changing calcium concentration leads to the reverse situation with a high C‐PTH fragments/PTH(1‐84) ratio. Reprinted, with permission, from (46), Figure 2.


Figure 3. Overview of vitamin D metabolism. Vitamin D is obtained from dietary sources, and is metabolized via UVB from 7‐DHC in the skin. Both sources (diet and skin) of vitamin D2 and vitamin D3 bind to DBP and circulate to the liver. In the liver, vitamin D is hydroxylated by CYP27A1 to 25(OH)D, commonly referred to as calcidiol. Calcidiol is then further metabolized to calcitriol by the 1‐αhydroxylase enzyme (CYP27B1) at the level of the kidney. The active metabolite 1,25(OH)2D (calcitriol) acts principally on the target organs of intestine, parathyroid gland, bone cell precursors, and the kidney. Calcitriol is metabolized to the inert 1,24,25(OH)3D through the action of the 24,25‐hydroxylase enzyme (CYP24). Calcidiol is similarly hydroxylated to 24,25(OH)2D (134).


Figure 4. The bone‐kidney‐parathyroid endocrine axes mediated by FGF23 and klotho. Active form of vitamin D (1,25‐dihydroxyvitamin D3) binds to VDR in the bone (osteocytes). The ligand‐bound VDR forms a heterodimer with a nuclear receptor RXR and transactivates expression of the FGF23 gene. FGF23 secreted from bone acts on the klotho‐FGFR complex expressed in the kidney (the bone‐kidney axis) and parathyroid gland (the bone‐parathyroid axis). In the kidney, FGF23 suppresses synthesis of active vitamin D by down‐regulating expression of the Cyp27b1 gene and promotes its inactivation by upregulating expression of the Cyp24 gene, thereby closing a negative feedback loop for vitamin D homeostasis. In the parathyroid gland, FGF23 suppresses production and secretion of PTH. PTH binds to PTHR expressed on renal tubular cells, leading to upregulation of the Cyp27b1 gene expression. Thus, suppression of PTH by FGF23 reduces expression of the Cyp27b1 gene and serum levels of 1,25‐dihydroxyvitamin D3. This closes another long negative feedback loop for vitamin D homeostasis. Reprinted, with permission, from (106).


Figure 5. Location of CaSR in the kidney. CaSR expression along the nephron. For each segment (Glom, glomerulus; MTALH, medullary thick ascending limb of Henle; CTALH, cortical thick ascending limb of Henle; and MD, macula densa; ), the location within the cells of each segment is shown along with the physiologic targets and cell‐specific functions of the CaSR. Localization in red indicates that morphologic studies do not uniformly recognize the CaSR in these locations, but that supporting functional data exist. Localization in green indicates that most or all studies find the CaSR in this location and that physiologic data support the localization. Podo, podocyte; intercal, intercalated. Reprinted, with permission, from (199), Figure 1.


Figure 6. Functions of the CaSR in the thick ascending limb of Henle's loop. The apical electroneutral NKCC2 transporter and ROMK channel (which is the rate‐limiting step for apical K+ recycling) generate a lumen‐positive transepithelial potential difference. This creates the driving force for nonselective, paracellular reabsorption of Ca2 cations. The paracellular transport is through a complex of claudin 16, 19 that is inhibited through physical interaction by a third tight junction protein claudin 14. Activation of the CaSR on the basolateral side of the cell increases claudin 14 expression (yellow) through NFAT signaling, and thus inhibits paracellular transport. Furthermore, activation of the CaSR also inhibits the ROMK channel to further inhibit calcium reabsorption in the setting of hypercalemia, although the mechanism is not understood. Reprinted, with permission, from (195), Figure 1.


Figure 7. Calcium active transport in the DCT. The late part of the DCT and CNT play an important role in fine‐tuning renal excretion of Ca2+. The ephosphatethelial Ca2+ channel (TRPV5) is primarily expressed aphosphatecally in these segments and co‐localizes with calbindin‐D28K (28K), Na+/Ca2+ exchanger (NCX1), and the plasma membrane ATPase (PMCA1b). Upon entry via TRPV5, Ca2+ is buffered by 28K and diffuses to the basolateral membrane, where it is released and extruded by a concerted action of NCX1 and PMCA1b. In addition, the basolateral membrane exposes a parathyroid hormone receptor (PTHR) and the Na+/K+‐ATPase consisting of the α‐, β‐, and γ‐subunit. PTHR activation by PTH stimulates TRPV5 activity, and entered Ca2+ can subsequently control the expression level of the Ca2+ transporters. At the aphosphatecal membrane, there is a bradykinin receptor (BK2) that is activated by urinary TK to activate TRPV5‐mediated Ca2+ influx. In the cell, entered Ca2+ acts as a negative feedback on channel activity, and 28K plays a regulatory role by association with TRPV5 under low intracellular Ca2+ concentrations. Extracellular urinary klotho directly stimulates TRPV5 at the aphosphatecal membrane by modification of the N‐glycan, whereas intracellular klotho enhances Na+/K+‐ATPase surface expression that in turn activates NCX1‐mediated Ca2+ efflux. Reprinted, with permission, from (19).


Figure 8. Physiologic roles of klotho on solute channels and transporters and vitamin D metabolism in the kidney klotho is prominently expressed in DCTs, and less in the proximal convoluted tubules (PCT). In PCT, membrane klotho at the basolateral side functions as coreceptor of FGFRs and drive FGF23 signal transduction to inhibit NaPi cotransporters (NaPi: 2a/c and Pit2) and to suppress cyp27β1 encoding for 1‐hydroxylase,and to stimulate cyp24α1 encoding for 24‐hydoxylase. The role of klotho in the cytoplasm of renal tubules is unclear. Whether membrane klotho at luminal side inhibits NaPi cotransporters (2a/c and Pit2) in an autocrine mode is not known (dash line). In DCT, membrane klotho at basolateral side4 functions as coreceptor of FGFRs to induce FGF23 signal transduction. What intermediate(s) are released from DCT and how intermediate(s) affect on PCT in paracrine mode is not known. One possible candidate is klotho release from the DCT to act on the PCT. Whether membrane klotho at luminal side directly regulates TRPV5 in autocrine mode is unknown. Soluble Klotho in luminal urine derived from either blood or urine exerts regulatory action on NaPi cotransporters in PCT; and on TRPV5 in DCT. Ca: calcium ion; FGF23: NaPi cotransporter: sodium‐phosphate‐dependent cotransporter; Pi: phosphate; 1,25 VD3: 1,25‐(OH)2 vitamin D3, Dash line: suspected action. Reprinted, with permission, from (89).


Figure 9. Overview of calcium and phosphate homeostasis. As (phosphate) levels increase (or there is a chronic phosphate load), both PTH and FGF23 are increased. Both the elevated PTH and FGF23 increase urinary phosphate excretion. The two hormones differ in respect to their effects on the vitamin D axis. PTH stimulates 1‐alpha hydroxylase activity thereby increasing the production of 1,25(OH)2D, which in turn negatively feeds back on the parathyroid gland to decrease PTH secretion. In contrast, FGF23 inhibits 1‐alpha hydroxylase activity, thereby decreasing the production of 1,25(OH)2D feeding back to stimulate further secretion of FGF23. FGF23 and PTH also regulate each other. Finally, low calcium levels stimulate PTH, whereas high calcium levels stimulate FGF23. Lastly, there is some evidence that FGF23 also inhibits PTH secretion (solid line = stimulates; dashed line = inhibits). Reprinted, with permission, from Moe SM, Sprague SM. Chronic kidney disease‐mineral bone disorder (134).


Figure 10. Calcium balance in stage 3/4 CKD patients with and without calcium carbonate. Calcium balance was greater with calcium carbonate compared with placebo. Ca intake was experimentally controlled and statistical analysis does not apply. White bars = placebo; Black bars = calcium carbonate; Ca = calcium; NS = not significant (P > 0.05). Data are presented as least squares mean ± pooled SEM. Reprinted, with permission, from (85).
References
 1.Alexander RT, Dimke H, Cordat E. Proximal tubular NHEs: Sodium, protons and calcium? Am J Physiol Renal Physiol 305: F229‐F236, 2013.
 2.Alfrey AC, LeGendre GR, Kaehny WD. The dialysis encephalopathy syndrome. Possible aluminum intoxication. N Engl J Med 294: 184‐188, 1976.
 3.Andress DL, Norris KC, Coburn JW, Slatopolsky EA, Sherrard DJ. Intravenous calcitriol in the treatment of refractory osteitis fibrosa of chronic renal failure [see comments]. N Engl J Med 321: 274‐279, 1989.
 4.Ba J, Brown D, Friedman PA. Calcium‐sensing receptor regulation of PTH‐inhibitable proximal tubule phosphate transport. Am J Physiol Renal Physiol 285: F1233‐F1243, 2003.
 5.Bacchetta J, Sea JL, Chun RF, Lisse TS, Wesseling‐Perry K, Gales B, Adams JS, Salusky IB, Hewison M. Fibroblast growth factor 23 inhibits extrarenal synthesis of 1,25‐dihydroxyvitamin D in human monocytes. J Bone Miner Res 28: 46‐55, 2013.
 6.Bacic D, Lehir M, Biber J, Kaissling B, Murer H, Wagner CA. The renal Na+/phosphate cotransporter NaPi‐IIa is internalized via the receptor‐mediated endocytic route in response to parathyroid hormone. Kidney Int 69: 495‐503, 2006.
 7.Baker LR, Abrams SM, Roe CJ, Faugere MC, Fanti P, Subayti Y, Malluche HH. Early therapy of renal bone disease with calcitriol: A prospective double‐blind study. Kidney Int Suppl 27: S140‐S142, 1989.
 8.Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: From human mutations to treatments. Nat Med 19: 179‐192, 2013.
 9.Bellido T, Saini V, Pajevic PD. Effects of PTH on osteocyte function. Bone 54: 250‐257, 2013.
 10.Belov AA, Mohammadi M. Molecular mechanisms of fibroblast growth factor signaling in physiology and pathology. Cold Spring Harb Perspect Biol 5: pii: a015958, 2013.
 11.Ben‐Dov IZ, Galitzer H, Lavi‐Moshayoff V, Goetz R, Kuro‐o M, Mohammadi M, Sirkis R, Naveh‐Many T, Silver J. The parathyroid is a target organ for FGF23 in rats. J Clin Invest 117: 4003‐4008, 2007.
 12.Benn BS, Ajibade D, Porta A, Dhawan P, Hediger M, Peng JB, Jiang Y, Oh GT, Jeung EB, Lieben L, Bouillon R, Carmeliet G, Christakos S. Active intestinal calcium transport in the absence of transient receptor potential vanilloid type 6 and calbindin‐D9k. Endocrinology 149: 3196‐3205, 2008.
 13.Bergsland KJ, Coe FL, Gillen DL, Worcester EM. A test of the hypothesis that the collecting duct calcium‐sensing receptor limits rise of urine calcium molarity in hypercalciuric calcium kidney stone formers. Am J Physiol Renal Physiol 297: F1017‐F1023, 2009.
 14.Berndt TJ, Schiavi S, Kumar R. “Phosphatonins” and the regulation of phosphorus homeostasis. Am J Physiol Renal Physiol 289: F1170‐F1182, 2005.
 15.Bleyer AJ, Burke SK, Dillon M, Garrett B, Kant KS, Lynch D, Rahman SN, Schoenfeld P, Teitelbaum I, Zeig S, Slatopolsky E. A comparison of the calcium‐free phosphate binder sevelamer hydrochloride with calcium acetate in the treatment of hyperphosphatemia in hemodialysis patients. Am J Kidney Dis 33: 694‐701, 1999.
 16.Block GA, Klassen PS, Lazarus JM, Ofsthun N, Lowrie EG, Chertow GM. Mineral metabolism, mortality, and morbidity in maintenance hemodialysis. J Am Soc Nephrol 15: 2208‐2218, 2004.
 17.Block GA, Martin KJ, de Francisco AL, Turner SA, Avram MM, Suranyi MG, Hercz G, Cunningham J, Abu‐Alfa AK, Messa P, Coyne DW, Locatelli F, Cohen RM, Evenepoel P, Moe SM, Fournier A, Braun J, McCary LC, Zani VJ, Olson KA, Drueke TB, Goodman WG. Cinacalcet for secondary hyperparathyroidism in patients receiving hemodialysis. N Engl J Med 350: 1516‐1525, 2004.
 18.Block GA, Spiegel DM, Ehrlich J, Mehta R, Lindbergh J, Dreisbach A, Raggi P. Effects of sevelamer and calcium on coronary artery calcification in patients new to hemodialysis. Kidney Int 68: 1815‐1824, 2005.
 19.Boros S, Bindels RJ, Hoenderop JG. Active Ca(2+) reabsorption in the connecting tubule. Pflugers Arch 458: 99‐109, 2009.
 20.Boskey AL, Gadaleta S, Gundberg C, Doty SB, Ducy P, Karsenty G. Fourier transform infrared microspectroscopic analysis of bones of osteocalcin‐deficient mice provides insight into the function of osteocalcin. Bone 23: 187‐196, 1998.
 21.Brennan SC, Davies TS, Schepelmann M, Riccardi D. Emerging roles of the extracellular calcium‐sensing receptor in nutrient sensing: Control of taste modulation and intestinal hormone secretion. Br J Nutr 111, S16‐S22, 2014.
 22.Bronner F. Recent developments in intestinal calcium absorption. Nutr Rev 67: 109‐113, 2009.
 23.Brown AJ, Ritter CS, Knutson JC, Strugnell SA. The vitamin D prodrugs 1alpha(OH)D2, 1alpha(OH)D3 and BCI‐210 suppress PTH secretion by bovine parathyroid cells. Nephrol Dial Transplant 21: 644‐650, 2006.
 24.Brown EM. Four‐parameter model of the sigmoidal relationship between parathyroid hormone release and extracellular calcium concentration in normal and abnormal parathyroid tissue. J Clin Endocrinol Metab 56: 572‐581, 1983.
 25.Brown EM. Extracellular Ca2+ sensing, regulation of parathyroid cell function, and role of Ca2+ and other ions as extracellular (first) messengers. Physiol Rev 71: 371‐411, 1991.
 26.Brown EM, Gamba G, Riccardi D, Lombardi M, Butters R, Kifor O, Sun A, Hediger MA, Lytton J, Hebert SC. Cloning and characterization of an extracellular Ca(2+)‐sensing receptor from bovine parathyroid. Nature 366: 575‐580, 1993.
 27.Brown EM, Katz C, Butters R, Kifor O. Polyarginine, polylysine, and protamine mimic the effects of high extracellular calcium concentrations on dispersed bovine parathyroid cells. J Bone Miner Res 6: 1217‐1225, 1991.
 28.Brown EM, Pollak M, Chou YH, Seidman CE, Seidman JG, Hebert SC. Cloning and functional characterization of extracellular Ca(2+)‐sensing receptors from parathyroid and kidney. Bone 17: 7S‐11S, 1995.
 29.Brownstein CA, Adler F, Nelson‐Williams C, Iijima J, Li P, Imura A, Nabeshima Y, Reyes‐Mugica M, Carpenter TO, Lifton RP. A translocation causing increased alpha‐klotho level results in hypophosphatemic rickets and hyperparathyroidism. Proc Natl Acad Sci U S A 105: 3455‐3460, 2008.
 30.Canalejo R, Canalejo A, Martinez‐Moreno JM, Rodriguez‐Ortiz ME, Estepa JC, Mendoza FJ, Munoz‐Castaneda JR, Shalhoub V, Almaden Y, Rodriguez M. FGF23 fails to inhibit uremic parathyroid glands. J Am Soc Nephrol 21: 1125‐1135, 2010.
 31.Capuano P, Bacic D, Stange G, Hernando N, Kaissling B, Pal R, Kocher O, Biber J, Wagner CA, Murer H. Expression and regulation of the renal Na/phosphate cotransporter NaPi‐IIa in a mouse model deficient for the PDZ protein PDZK1. Pflugers Arch 449: 392‐402, 2005.
 32.Carpenter TO, Insogna KL, Zhang JH, Ellis B, Nieman S, Simpson C, Olear E, Gundberg CM. Circulating levels of soluble klotho and FGF23 in X‐linked hypophosphatemia: Circadian variance, effects of treatment, and relationship to parathyroid status. J Clin Endocrinol Metab 95: E352‐E357, 2010.
 33.Cha SK, Jabbar W, Xie J, Huang CL. Regulation of TRPV5 single‐channel activity by intracellular pH. J Membr Biol 220: 79‐85, 2007.
 34.Cha SK, Kim JH, Huang CL. Flow‐induced activation of TRPV5 and TRPV6 channels stimulates Ca(2+)‐activated K(+) channel causing membrane hyperpolarization. Biochim Biophys Acta 1833: 3046‐3053, 2013.
 35.Chang Q, Hoefs S, van der Kemp AW, Topala CN, Bindels RJ, Hoenderop JG. The beta‐glucuronidase klotho hydrolyzes and activates the TRPV5 channel. Science 310: 490‐493, 2005.
 36.Charytan C, Coburn JW, Chonchol M, Herman J, Lien YH, Liu W, Klassen PS, McCary LC, Pichette V. Cinacalcet hydrochloride is an effective treatment for secondary hyperparathyroidism in patients with CKD not receiving dialysis. Am J Kidney Dis 46: 58‐67, 2005.
 37.Chertow GM, Burke SK, Raggi P. Sevelamer attenuates the progression of coronary and aortic calcification in hemodialysis patients. Kidney Int 62: 245‐252, 2002.
 38.Coburn JW, Hartenbower DL, Massry SG. Intestinal absorption of calcium and the effect of renal insufficiency. Kidney Int 4: 96‐104, 1973.
 39.Coburn JW, Koppel MH, Brickman AS, Massry SG. Study of intestinal absorption of calcium in patients with renal failure. Kidney Int 3: 264‐272, 1973.
 40.Cochran M, Bulusu L, Horsman A, Stasiak L, Nordin BE. Hypocalcaemia and bone disease in renal failure. Nephron 10: 113‐140, 1973.
 41.Conigrave AD, Ward DT. Calcium‐sensing receptor (CaSR): Pharmacological properties and signaling pathways. Best Pract Res Clin Endocrinol Metab 27: 315‐331, 2013.
 42.Cozzolino M, Dusso AS, Liapis H, Finch J, Lu Y, Burke SK, Slatopolsky E. The effects of sevelamer hydrochloride and calcium carbonate on kidney calcification in uremic rats. J Am Soc Nephrol 13: 2299‐2308, 2002.
 43.Cozzolino M, Staniforth ME, Liapis H, Finch J, Burke SK, Dusso AS, Slatopolsky E. Sevelamer hydrochloride attenuates kidney and cardiovascular calcifications in long‐term experimental uremia. Kidney Int 64: 1653‐1661, 2003.
 44.Cross HS. Extrarenal vitamin D hydroxylase expression and activity in normal and malignant cells: Modification of expression by epigenetic mechanisms and dietary substances. Nutr Rev 65: S108‐S112, 2007.
 45.Crouthamel MH, Lau WL, Leaf EM, Chavkin NW, Wallingford MC, Peterson DF, Li X, Liu Y, Chin MT, Levi M, Giachelli CM. Sodium‐dependent phosphate cotransporters and phosphate‐induced calcification of vascular smooth muscle cells: Redundant roles for PiT‐1 and PiT‐2. Arterioscler Thromb Vasc Biol 33: 2625‐2632, 2013.
 46.D'Amour P. Acute and chronic regulation of circulating PTH: Significance in health and in disease. Clin Biochem 45: 964‐969, 2012.
 47.D'Haese PC, Spasovski GB, Sikole A, Hutchison A, Freemont TJ, Sulkova S, Swanepoel C, Pejanovic S, Djukanovic L, Balducci A, Coen G, Sulowicz W, Ferreira A, Torres A, Curic S, Popovic M, Dimkovic N, De Broe ME. A multicenter study on the effects of lanthanum carbonate (Fosrenol) and calcium carbonate on renal bone disease in dialysis patients. Kidney Int Suppl S73‐S78, 2003.
 48.Danese MD, Halperin M, Lowe KA, Bradbury BD, Do TP, Block GA. Refining the definition of clinically important mineral and bone disorder in hemodialysis patients. Nephrol Dial Transplant 30: 1336‐1344, 2015.
 49.de Borst MH, Vervloet MG, ter Wee PM, Navis G. Cross talk between the renin‐angiotensin‐aldosterone system and vitamin D‐FGF‐23‐klotho in chronic kidney disease. J Am Soc Nephrol 22: 1603‐1609, 2011.
 50.Deliot N, Hernando N, Horst‐Liu Z, Gisler SM, Capuano P, Wagner CA, Bacic D, O'Brien S, Biber J, Murer H. Parathyroid hormone treatment induces dissociation of type IIa Na+‐P(i) cotransporter‐Na+/H +exchanger regulatory factor‐1 complexes. Am J Physiol Cell Physiol 289: C159‐C167, 2005.
 51.Denda M, Finch J, Slatopolsky E. Phosphorus accelerates the development of parathyroid hyperplasia and secondary hyperparathyroidism in rats with renal failure. Am J Kidney Dis 28: 596‐602, 1996.
 52.Dewberry K, Fox J, Stewart J, Murray J, Hutchison AJ. Lanthanum carbonate: A novel non‐calcium containing phosphate binder. J Amer Soc Neph 8: A2610, 1997.
 53.Di Marco GS, Reuter S, Kentrup D, Grabner A, Amaral AP, Fobker M, Stypmann J, Pavenstadt H, Wolf M, Faul C, Brand M. Treatment of established left ventricular hypertrophy with fibroblast growth factor receptor blockade in an animal model of CKD. Nephrol Dial Transplant 29: 2028‐2035, 2014.
 54.Diaz de Barboza G, Guizzardi S, Tolosa de Talamoni N. Molecular aspects of intestinal calcium absorption. World J Gastroenterol 21: 7142‐7154, 2015.
 55.Dimke H, Desai P, Borovac J, Lau A, Pan W, Alexander RT. Activation of the Ca(2+)‐sensing receptor increases renal claudin‐14 expression and urinary Ca(2+) excretion. Am J Physiol Renal Physiol 304: F761‐F769, 2013.
 56.Divieti P, John MR, Juppner H, Bringhurst FR. Human PTH‐(7‐84) inhibits bone resorption in vitro via actions independent of the type 1 PTH/PTHrP receptor. Endocrinology 143: 171‐176, 2002.
 57.Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G. Osf2/Cbfa1: A transcriptional activator of osteoblast differentiation [see comments]. Cell 89: 747‐754, 1997.
 58.Dunlay R, Hruska K. PTH receptor coupling to phospholipase C is an alternate pathway of signal transduction in bone and kidney. Am J Physiol 258: F223‐F231, 1990.
 59.Dusso AS, Finch J, Brown A, Ritter C, Delmez J, Schreiner G, Slatopolsky E. Extrarenal production of calcitriol in normal and uremic humans. J Clin Endocrinol Metab 72: 157‐164, 1991.
 60.Dusso AS, Negrea L, Gunawardhana S, Lopez‐Hilker S, Finch J, Mori T, Nishii Y, Slatopolsky E, Brown AJ. On the mechanisms for the selective action of vitamin D analogs. Endocrinology 128: 1687‐1692, 1991.
 61.Egbuna O, Quinn S, Kantham L, Butters R, Pang J, Pollak M, Goltzman D, Brown E. The full‐length calcium‐sensing receptor dampens the calcemic response to 1alpha,25(OH)2 vitamin D3 in vivo independently of parathyroid hormone. Am J Physiol Renal Physiol 297: F720‐F728, 2009.
 62.Farrow EG, Davis SI, Summers LJ, White KE. Initial FGF23‐mediated signaling occurs in the distal convoluted tubule. J Am Soc Nephrol 20: 955‐960, 2009.
 63.Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutierrez OM, Aguillon‐Prada R, Lincoln J, Hare JM, Mundel P, Morales A, Scialla J, Fischer M, Soliman EZ, Chen J, Go AS, Rosas SE, Nessel L, Townsend RR, Feldman HI, St John Sutton M, Ojo A, Gadegbeku C, Di Marco GS, Reuter S, Kentrup D, Tiemann K, Brand M, Hill JA, Moe OW, Kuro OM, Kusek JW, Keane MG, Wolf M. FGF23 induces left ventricular hypertrophy. J Clin Invest 121: 4393‐4408, 2011.
 64.Feng JQ, Clinkenbeard EL, Yuan B, White KE, Drezner MK. Osteocyte regulation of phosphate homeostasis and bone mineralization underlies the pathophysiology of the heritable disorders of rickets and osteomalacia. Bone 54: 213‐221, 2013.
 65.Galitzer H, Ben‐Dov IZ, Silver J, Naveh‐Many T. Parathyroid cell resistance to fibroblast growth factor 23 in secondary hyperparathyroidism of chronic kidney disease. Kidney Int 77: 211‐218, 2010.
 66.Gardiner EM, Baldock PA, Thomas GP, Sims NA, Henderson NK, Hollis B, White CP, Sunn KL, Morrison NA, Walsh WR, Eisman JA. Increased formation and decreased resorption of bone in mice with elevated vitamin D receptor in mature cells of the osteoblastic lineage. FASEB J 14: 1908‐1916, 2000.
 67.Gattineni J, Bates C, Twombley K, Dwarakanath V, Robinson ML, Goetz R, Mohammadi M, Baum M. FGF23 decreases renal NaPi‐2a and NaPi‐2c expression and induces hypophosphatemia in vivo predominantly via FGF receptor 1. Am J Physiol Renal Physiol 297: F282‐F291, 2009.
 68.Gauci C, Moranne O, Fouqueray B, de la Faille R, Maruani G, Haymann JP, Jacquot C, Boffa JJ, Flamant M, Rossert J, Urena P, Stengel B, Souberbielle JC, Froissart M, Houillier P. Pitfalls of measuring total blood calcium in patients with CKD. J Am Soc Nephrol 19: 1592‐1598, 2008.
 69.Giachelli CM. Vascular calcification: In vitro evidence for the role of inorganic phosphate. J Am Soc Nephrol 14: S300‐S304, 2003.
 70.Gkika D, Topala CN, Chang Q, Picard N, Thebault S, Houillier P, Hoenderop JG, Bindels RJ. Tissue kallikrein stimulates Ca(2+) reabsorption via PKC‐dependent plasma membrane accumulation of TRPV5. EMBO J 25: 4707‐4716, 2006.
 71.Glendenning P. It is time to start ordering ionized calcium more frequently: Preanalytical factors can be controlled and postanalytical data justify measurement. Ann Clin Biochem 50: 191‐193, 2013.
 72.Goltzman D. Inferences from genetically modified mouse models on the skeletal actions of vitamin D. J Steroid Biochem Mol Biol 148: 219‐224, 2015.
 73.Gong Y, Hou J. Claudin‐14 underlies Ca(+)(+)‐sensing receptor‐mediated Ca(+)(+) metabolism via NFAT‐microRNA‐based mechanisms. J Am Soc Nephrol 25: 745‐760, 2014.
 74.Gong Y, Renigunta V, Himmerkus N, Zhang J, Renigunta A, Bleich M, Hou J. Claudin‐14 regulates renal Ca(+)(+) transport in response to CaSR signalling via a novel microRNA pathway. EMBO J 31: 1999‐2012, 2012.
 75.Grabner A, Amaral AP, Schramm K, Singh S, Sloan A, Yanucil C, Li J, Shehadeh LA, Hare JM, David V, Martin A, Fornoni A, Di Marco GS, Kentrup D, Reuter S, Mayer AB, Pavenstadt H, Stypmann J, Kuhn C, Hille S, Frey N, Leifheit‐Nestler M, Richter B, Haffner D, Abraham R, Bange J, Sperl B, Ullrich A, Brand M, Wolf M, Faul C. Activation of cardiac fibroblast growth factor receptor 4 causes left ventricular hypertrophy. Cell Metab 22: 1020‐1032, 2015.
 76.Graciolli FG, Neves KR, dos Reis LM, Graciolli RG, Noronha IL, Moyses RM, Jorgetti V. Phosphorus overload and PTH induce aortic expression of Runx2 in experimental uraemia. Nephrol Dial Transplant 24: 1416‐1421, 2009.
 77.Groschel C, Tennakoon S, Kallay E. Cytochrome P450 vitamin D hydroxylases in inflammation and cancer. Adv Pharmacol 74: 413‐458, 2015.
 78.Gunzel D, Haisch L, Pfaffenbach S, Krug SM, Milatz S, Amasheh S, Hunziker W, Muller D. Claudin function in the thick ascending limb of Henle's loop. Ann N Y Acad Sci 1165: 152‐162, 2009.
 79.Gutierrez OM, Januzzi JL, Isakova T, Laliberte K, Smith K, Collerone G, Sarwar A, Hoffmann U, Coglianese E, Christenson R, Wang TJ, deFilippi C, Wolf M. Fibroblast growth factor 23 and left ventricular hypertrophy in chronic kidney disease. Circulation 119: 2545‐2552, 2009.
 80.Gutierrez OM, Mannstadt M, Isakova T, Rauh‐Hain JA, Tamez H, Shah A, Smith K, Lee H, Thadhani R, Juppner H, Wolf M. Fibroblast growth factor 23 and mortality among patients undergoing hemodialysis. N Engl J Med 359: 584‐592, 2008.
 81.Healy MD, Malluche HH, Goldstein DA, Singer FR, Massry SG. Effects of long‐term therapy with calcitriol in patients with moderate renal failure. Arch Intern Med 140: 1030‐1033, 1980.
 82.Heaney R, Skillman T. Secretion and excretion of calcium by the human gastrointestinal tract. J Lab & Clin Med 64: 29‐41, 1964.
 83.Hendy GN, D'Souza‐Li L, Yang B, Canaff L, Cole DE. Mutations of the calcium‐sensing receptor (CASR) in familial hypocalciuric hypercalcemia, neonatal severe hyperparathyroidism, and autosomal dominant hypocalcemia. Hum Mutat 16: 281‐296, 2000.
 84.Hendy GN, Hruska KA, Mathew S, Goltzman D. New insights into mineral and skeletal regulation by active forms of vitamin D. Kidney Int 69: 218‐223, 2006.
 85.Hill KM, Martin BR, Wastney ME, McCabe GP, Moe SM, Weaver CM, Peacock M. Oral calcium carbonate affects calcium but not phosphorus balance in stage 3‐4 chronic kidney disease. Kidney Int 83: 959‐966, 2012.
 86.Hoenderop JG, van Leeuwen JP, van der Eerden BC, Kersten FF, van der Kemp AW, Merillat AM, Waarsing JH, Rossier BC, Vallon V, Hummler E, Bindels RJ. Renal Ca2+ wasting, hyperabsorption, and reduced bone thickness in mice lacking TRPV5. J Clin Invest 112: 1906‐1914, 2003.
 87.Hoenderop JG, Voets T, Hoefs S, Weidema F, Prenen J, Nilius B, Bindels RJ. Homo‐ and heterotetrameric architecture of the epithelial Ca2+ channels TRPV5 and TRPV6. EMBO J 22: 776‐785, 2003.
 88.Hoorn EJ, Zietse R. Disorders of calcium and magnesium balance: A physiology‐based approach. Pediatr Nephrol 28: 1195‐1206, 2013.
 89.Hu MC, Kuro‐o M, Moe OW. Renal and extrarenal actions of Klotho. Semin Nephrol 33: 118‐129, 2013.
 90.Hu MC, Shi M, Cho HJ, Adams‐Huet B, Paek J, Hill K, Shelton J, Amaral AP, Faul C, Taniguchi M, Wolf M, Brand M, Takahashi M, Kuro OM, Hill JA, Moe OW. Klotho and phosphate are modulators of pathologic uremic cardiac remodeling. J Am Soc Nephrol 26: 1290‐1302, 2015.
 91.Hu MC, Shi M, Zhang J, Pastor J, Nakatani T, Lanske B, Razzaque MS, Rosenblatt KP, Baum MG, Kuro‐o M, Moe OW. Klotho: A novel phosphaturic substance acting as an autocrine enzyme in the renal proximal tubule. FASEB J 24: 3438‐3450, 2010.
 92.Investigators ET, Chertow GM, Block GA, Correa‐Rotter R, Drueke TB, Floege J, Goodman WG, Herzog CA, Kubo Y, London GM, Mahaffey KW, Mix TC, Moe SM, Trotman ML, Wheeler DC, Parfrey PS. Effect of cinacalcet on cardiovascular disease in patients undergoing dialysis. N Engl J Med 367: 2482‐2494, 2012.
 93.Isakova T, Gutierrez OM, Chang Y, Shah A, Tamez H, Smith K, Thadhani R, Wolf M. Phosphorus binders and survival on hemodialysis. J Am Soc Nephrol 20: 388‐396, 2009.
 94.Isakova T, Wahl P, Vargas GS, Gutierrez OM, Scialla J, Xie H, Appleby D, Nessel L, Bellovich K, Chen J, Hamm L, Gadegbeku C, Horwitz E, Townsend RR, Anderson CA, Lash JP, Hsu CY, Leonard MB, Wolf M. Fibroblast growth factor 23 is elevated before parathyroid hormone and phosphate in chronic kidney disease. Kidney Int 79: 1370‐1378, 2011.
 95.Isakova T, Xie H, Yang W, Xie D, Anderson AH, Scialla J, Wahl P, Gutierrez OM, Steigerwalt S, He J, Schwartz S, Lo J, Ojo A, Sondheimer J, Hsu CY, Lash J, Leonard M, Kusek JW, Feldman HI, Wolf M. Fibroblast growth factor 23 and risks of mortality and end‐stage renal disease in patients with chronic kidney disease. JAMA 305: 2432‐2439, 2011.
 96.Jiang Y, Ferguson WB, Peng JB. WNK4 enhances TRPV5‐mediated calcium transport: Potential role in hypercalciuria of familial hyperkalemic hypertension caused by gene mutation of WNK4. Am J Physiol Renal Physiol 292: F545‐554, 2007.
 97.Jones G. Extrarenal vitamin D activation and interactions between vitamin D(2), vitamin D(3), and vitamin D analogs. Annu Rev Nutr 33: 23‐44, 2013.
 98.Kantham L, Quinn SJ, Egbuna OI, Baxi K, Butters R, Pang JL, Pollak MR, Goltzman D, Brown EM. The calcium‐sensing receptor (CaSR) defends against hypercalcemia independently of its regulation of parathyroid hormone secretion. Am J Physiol Endocrinol Metab 297: E915‐923, 2009.
 99.Katsumata K, Kusano K, Hirata M, Tsunemi K, Nagano N, Burke SK, Fukushima N. Sevelamer hydrochloride prevents ectopic calcification and renal osteodystrophy in chronic renal failure rats. Kidney Int 64: 441‐450, 2003.
 100.KDIGO. Clinical practice guidelines for the management of CKD‐MBD. Kidney Int 76: S1‐S130, 2009.
 101.Kestenbaum B, Sampson JN, Rudser KD, Patterson DJ, Seliger SL, Young B, Sherrard DJ, Andress DL. Serum phosphate levels and mortality risk among people with chronic kidney disease. J Am Soc Nephrol 16: 520‐528, 2005.
 102.Kolek OI, Hines ER, Jones MD, LeSueur LK, Lipko MA, Kiela PR, Collins JF, Haussler MR, Ghishan FK. 1alpha,25‐Dihydroxyvitamin D3 upregulates FGF23 gene expression in bone: The final link in a renal‐gastrointestinal‐skeletal axis that controls phosphate transport. Am J Physiol Gastrointest Liver Physiol 289: G1036‐G1042, 2005.
 103.Kos CH, Karaplis AC, Peng JB, Hediger MA, Goltzman D, Mohammad KS, Guise TA, Pollak MR. The calcium‐sensing receptor is required for normal calcium homeostasis independent of parathyroid hormone. J Clin Invest 111: 1021‐1028, 2003.
 104.Krajisnik T, Olauson H, Mirza MA, Hellman P, Akerstrom G, Westin G, Larsson TE, Bjorklund P. Parathyroid Klotho and FGF‐receptor 1 expression decline with renal function in hyperparathyroid patients with chronic kidney disease and kidney transplant recipients. Kidney Int 78: 1024‐1032, 2010.
 105.Krupp K, Madhivanan P. FGF23 and risk of all‐cause mortality and cardiovascular events: A meta‐analysis of prospective cohort studies. Int J Cardiol 176: 1341‐1342, 2014.
 106.Kuro‐o M. Overview of the FGF23‐Klotho axis. Pediatr Nephrol 25: 583‐590, 2010.
 107.Kuro‐o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, Ohyama Y, Kurabayashi M, Kaname T, Kume E, Iwasaki H, Iida A, Shiraki‐Iida T, Nishikawa S, Nagai R, Nabeshima YI. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 390: 45‐51, 1997.
 108.Lambers TT, Oancea E, de Groot T, Topala CN, Hoenderop JG, Bindels RJ. Extracellular pH dynamically controls cell surface delivery of functional TRPV5 channels. Mol Cell Biol 27: 1486‐1494, 2007.
 109.Lambert PW, Stern PH, Avioli RC, Brackett NC, Turner RT, Greene A, Fu IY, Bell NH. Evidence for extrarenal production of 1 alpha,25‐dihydroxyvitamin D in man. J Clin Invest 69: 722‐725, 1982.
 110.Lameris AL, Nevalainen PI, Reijnen D, Simons E, Eygensteyn J, Monnens L, Bindels RJ, Hoenderop JG. Segmental transport of Ca(2)(+) and Mg(2)(+) along the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 308: G206‐G216, 2015.
 111.Lavi‐Moshayoff V, Wasserman G, Meir T, Silver J, Naveh‐Many T. PTH increases FGF23 gene expression and mediates the high‐FGF23 levels of experimental kidney failure: A bone parathyroid feedback loop. Am J Physiol Renal Physiol 299: F882‐F889, 2010.
 112.Lavi‐Moshayoff V, Wasserman G, Meir T, Silver J, Naveh‐Many T. PTH increases FGF23 gene expression and mediates the high FGF23 levels of experimental kidney failure: A bone parathyroid feedback loop. Am J Physiol Renal Physiol 299: F882‐F889, 2010.
 113.Lederer ED, Khundmiri SJ, Weinman EJ. Role of NHERF‐1 in regulation of the activity of Na‐K ATPase and sodium‐phosphate co‐transport in epithelial cells. J Am Soc Nephrol 14: 1711‐1719, 2003.
 114.Lee J, Cha SK, Sun TJ, Huang CL. PIP2 activates TRPV5 and releases its inhibition by intracellular Mg2+. J Gen Physiol 126: 439‐451, 2005.
 115.Lee M, Partridge NC. Parathyroid hormone signaling in bone and kidney. Curr Opin Nephrol Hypertens 18: 298‐302, 2009.
 116.Lee SM, Riley EM, Meyer MB, Benkusky NA, Plum LA, DeLuca HF, Pike JW. 1,25‐Dihydroxyvitamin D3 controls a cohort of vitamin D receptor target genes in the proximal intestine that is enriched for calcium‐regulating components. J Biol Chem 290: 18199‐18215, 2015.
 117.Li Y, Song YH, Rais N, Connor E, Schatz D, Muir A, Maclaren N. Autoantibodies to the extracellular domain of the calcium sensing receptor in patients with acquired hypoparathyroidism [see comments]. J Clin Invest 97: 910‐914, 1996.
 118.Lieben L, Benn BS, Ajibade D, Stockmans I, Moermans K, Hediger MA, Peng JB, Christakos S, Bouillon R, Carmeliet G. Trpv6 mediates intestinal calcium absorption during calcium restriction and contributes to bone homeostasis. Bone 47: 301‐308, 2010.
 119.Liu N, Nguyen L, Chun RF, Lagishetty V, Ren S, Wu S, Hollis B, DeLuca HF, Adams JS, Hewison M. Altered endocrine and autocrine metabolism of vitamin D in a mouse model of gastrointestinal inflammation. Endocrinology 149: 4799‐4808, 2008.
 120.Lu P, Boros S, Chang Q, Bindels RJ, Hoenderop JG. The beta‐glucuronidase klotho exclusively activates the epithelial Ca2+ channels TRPV5 and TRPV6. Nephrol Dial Transplant 23: 3397‐3402, 2008.
 121.Malluche H, Werner E, Ritz E. Intestinal absorption of calicum and whole‐body calcium retention in incipient and advanced renal failure. Miner Electrolyte Metab 1: 263‐270, 1978.
 122.Martino NA, Reshkin SJ, Ciani E, Dell'Aquila ME. Calcium‐sensing receptor‐mediated osteogenic and early‐stage neurogenic differentiation in umbilical cord matrix mesenchymal stem cells from a large animal model. PLoS One 9: e111533, 2014.
 123.Masuyama R. Role of local vitamin D signaling and cellular calcium transport system in bone homeostasis. J Bone Miner Metab 32: 1‐9, 2014.
 124.Mathew S, Lund RJ, Strebeck F, Tustison KS, Geurs T, Hruska KA. Reversal of the adynamic bone disorder and decreased vascular calcification in chronic kidney disease by sevelamer carbonate therapy. J Am Soc Nephrol 18: 122‐130, 2007.
 125.Matkovic V, Goel PK, Badenhop‐Stevens NE, Landoll JD, Li B, Ilich JZ, Skugor M, Nagode LA, Mobley SL, Ha EJ, Hangartner TN, Clairmont A. Calcium supplementation and bone mineral density in females from childhood to young adulthood: A randomized controlled trial. Am J Clin Nutr 81: 175‐188, 2005.
 126.Moe S, Drueke T, Cunningham J, Goodman W, Martin K, Olgaard K, Ott S, Sprague S, Lameire N, Eknoyan G, and Kidney Disease: Improving Global O. Definition, evaluation, and classification of renal osteodystrophy: A position statement from Kidney Disease: Improving Global Outcomes (KDIGO). Kidney Int 69: 1945‐1953, 2006.
 127.Moe SM, Abdalla S, Chertow GM, Parfrey PS, Block GA, Correa‐Rotter R, Floege J, Herzog CA, London GM, Mahaffey KW, Wheeler DC, Dehmel B, Goodman WG, Drueke TB, and Evaluation of Cinacalcet HTtLCETI. Effects of cinacalcet on fracture events in patients receiving hemodialysis: The EVOLVE trial. J Am Soc Nephrol 26: 1466‐1475, 2015.
 128.Moe SM, Chen NX, Newman CL, Gattone VH, II, Organ JM, Chen X, Allen MR. A comparison of calcium to zoledronic acid for improvement of cortical bone in an animal model of CKD. J Bone Miner Res 29: 902‐910, 2014.
 129.Moe SM, Chen NX, Seifert MF, Sinders RM, Duan D, Chen X, Liang Y, Radcliff JS, White KE, Gattone VH, II. A rat model of chronic kidney disease‐mineral bone disorder. Kidney Int 75: 176‐184, 2009.
 130.Moe SM, Chertow GM. The case against calcium‐based phosphate binders. Clin J Am Soc Nephrol 1: 697‐703, 2006.
 131.Moe SM, Chertow GM, Parfrey PS, Kubo Y, Block GA, Correa‐Rotter R, Drueke TB, Herzog CA, London GM, Mahaffey KW, Wheeler DC, Stolina M, Dehmel B, Goodman WG, Floege J, and Evaluation of Cinacalcet HTtLCETI. Cinacalcet, fibroblast growth factor‐23, and cardiovascular disease in hemodialysis: The evaluation of cinacalcet HCl therapy to lower cardiovascular events (EVOLVE) trial. Circulation 132: 27‐39, 2015.
 132.Moe SM, Duan D, Doehle BP, O'Neill KD, Chen NX. Uremia induces the osteoblast differentiation factor Cbfa1 in human blood vessels. Kidney Int 63: 1003‐1011, 2003.
 133.Moe SM, Seifert MF, Chen NX, Sinders RM, Chen X, Duan D, Henley C, Martin D, Gattone VH, II. R‐568 reduces ectopic calcification in a rat model of chronic kidney disease‐mineral bone disorder (CKD‐MBD). Nephrol Dial Transplant 24: 2371‐2377, 2009.
 134.Moe SM, Sprague S. Mineral bone disorders in chronic kidney disease. In: Brenner B, editor. The Kidney. Philadelphia, PA: Elsevier, 2016, pp. 1822‐1853.
 135.Nagano N, Miyata S, Abe M, Wakita S, Kobayashi N, Wada M. Effects of intermittent treatment with sevelamer hydrochloride on parathyroid hyperplasia and vascular calcification in rats with chronic kidney disease. Clin Calcium 15(Suppl 1): 35‐39; discussion 39‐40, 2005.
 136.Naveh‐Many T, Friedlaender MM, Mayer H, Silver J. Calcium regulates parathyroid hormone messenger ribonucleic acid (mRNA), but not calcitonin mRNA in vivo in the rat. Dominant role of 1,25‐dihydroxyvitamin D. Endocrinology 125: 275‐280, 1989.
 137.Naveh‐Many T, Silver J. Regulation of parathyroid hormone gene expression by hypocalcemia, hypercalcemia, and vitamin D in the rat. J Clin Invest 86: 1313‐1319, 1990.
 138.Nechama M, Ben‐Dov IZ, Silver J, Naveh‐Many T. Regulation of PTH mRNA stability by the calcimimetic R568 and the phosphorus binder lanthanum carbonate in CKD. Am J Physiol Renal Physiol 296: F795‐F800, 2009.
 139.Negri AL. The klotho gene: A gene predominantly expressed in the kidney is a fundamental regulator of aging and calcium/phosphorus metabolism. J Nephrol 18: 654‐658, 2005.
 140.Neves KR, Graciolli FG, dos Reis LM, Graciolli RG, Neves CL, Magalhaes AO, Custodio MR, Batista DG, Jorgetti V, Moyses RM. Vascular calcification: Contribution of parathyroid hormone in renal failure. Kidney Int 71: 1262‐1270, 2007.
 141.Nigwekar SU, Tamez H, Thadhani RI. Vitamin D and chronic kidney disease‐mineral bone disease (CKD‐MBD). BoneKEy Rep 3: 2014.
 142.Nijenhuis T, Renkema KY, Hoenderop JG, Bindels RJ. Acid‐base status determines the renal expression of Ca2+ and Mg2+ transport proteins. J Am Soc Nephrol 17: 617‐626, 2006.
 143.Nikolov IG, Joki N, Nguyen‐Khoa T, Guerrera IC, Maizel J, Benchitrit J, Machado dos Reis L, Edelman A, Lacour B, Jorgetti V, Drueke TB, Massy ZA. Lanthanum carbonate, like sevelamer‐HCl, retards the progression of vascular calcification and atherosclerosis in uremic apolipoprotein E‐deficient mice. Nephrol Dial Transplant 27: 505‐513, 2012.
 144.Nilius B, Prenen J, Vennekens R, Hoenderop JG, Bindels RJ, Droogmans G. Modulation of the epithelial calcium channel, ECaC, by intracellular Ca2+. Cell Calcium 29: 417‐428, 2001.
 145.Nordal KP, Dahl E. Low dose calcitriol versus placebo in patients with predialysis chronic renal failure. J Clin Endocrinol Metab 67: 929‐936, 1988.
 146.Olauson H, Lindberg K, Amin R, Sato T, Jia T, Goetz R, Mohammadi M, Andersson G, Lanske B, Larsson TE. Parathyroid‐specific deletion of Klotho unravels a novel calcineurin‐dependent FGF23 signaling pathway that regulates PTH secretion. PLoS Genet 9: e1003975, 2013.
 147.Palmer SC, Hayen A, Macaskill P, Pellegrini F, Craig JC, Elder GJ, Strippoli GF. Serum levels of phosphorus, parathyroid hormone, and calcium and risks of death and cardiovascular disease in individuals with chronic kidney disease: A systematic review and meta‐analysis. JAMA 305: 1119‐1127, 2011.
 148.Parfitt AM. Plasma calcium control at quiescent bone surfaces: A new approach to the homeostatic function of bone lining cells. Bone 10: 87‐88, 1989.
 149.Parfitt AM. Targeted and nontargeted bone remodeling: Relationship to basic multicellular unit origination and progression. Bone 30: 5‐7, 2002.
 150.Parfrey PS, Chertow GM, Block GA, Correa‐Rotter R, Drueke TB, Floege J, Herzog CA, London GM, Mahaffey KW, Moe SM, Wheeler DC, Dehmel B, Trotman ML, Modafferi DM, Goodman WG. The clinical course of treated hyperparathyroidism among patients receiving hemodialysis and the effect of cinacalcet: The EVOLVE trial. J Clin Endocrinol Metab 98: 4834‐4844, 2013.
 151.Parfrey PS, Drueke TB, Block GA, Correa‐Rotter R, Floege J, Herzog CA, London GM, Mahaffey KW, Moe SM, Wheeler DC, Kubo Y, Dehmel B, Goodman WG, Chertow GM, and Evaluation of Cinacalcet HTtLCETI. The effects of cinacalcet in older and younger patients on hemodialysis: The evaluation of cinacalcet HCl therapy to lower cardiovascular events (EVOLVE) trial. Clin J Am Soc Nephrol 10: 791‐799, 2015.
 152.Pearce SH, Williamson C, Kifor O, Bai M, Coulthard MG, Davies M, Lewis‐Barned N, McCredie D, Powell H, Kendall‐Taylor P, Brown EM, Thakker RV. A familial syndrome of hypocalcemia with hypercalciuria due to mutations in the calcium‐sensing receptor. N Engl J Med 335: 1115‐1122, 1996.
 153.Qi Q, Monier‐Faugere MC, Geng Z, Malluche HH. Predictive value of serum parathyroid hormone levels for bone turnover in patients on chronic maintenance dialysis. Am J Kidney Dis 26: 622‐631, 1995.
 154.Qunibi W, Moustafa M, Muenz LR, He DY, Kessler PD, Diaz‐Buxo JA, Budoff M. A 1‐year randomized trial of calcium acetate versus sevelamer on progression of coronary artery calcification in hemodialysis patients with comparable lipid control: The Calcium Acetate Renagel Evaluation‐2 (CARE‐2) study. Am J Kidney Dis 51: 952‐965, 2008.
 155.Raggi P, Bommer J, Chertow GM. Valvular calcification in hemodialysis patients randomized to calcium‐based phosphorus binders or sevelamer. J Heart Valve Dis 13: 134‐141, 2004.
 156.Ramirez JA, Goodman WG, Gornbein J, Menezes C, Moulton L, Segre GV, Salusky IB. Direct in vivo comparison of calcium‐regulated parathyroid hormone secretion in normal volunteers and patients with secondary hyperparathyroidism. J Clin Endocrinol Metab 76: 1489‐1494, 1993.
 157.Ren S, Nguyen L, Wu S, Encinas C, Adams JS, Hewison M. Alternative splicing of vitamin D‐24‐hydroxylase: A novel mechanism for the regulation of extrarenal 1,25‐dihydroxyvitamin D synthesis. J Biol Chem 280: 20604‐20611, 2005.
 158.Renkema KY, Velic A, Dijkman HB, Verkaart S, van der Kemp AW, Nowik M, Timmermans K, Doucet A, Wagner CA, Bindels RJ, Hoenderop JG. The calcium‐sensing receptor promotes urinary acidification to prevent nephrolithiasis. J Am Soc Nephrol 20: 1705‐1713, 2009.
 159.Reynolds JL, Joannides AJ, Skepper JN, McNair R, Schurgers LJ, Proudfoot D, Jahnen‐Dechent W, Weissberg PL, Shanahan CM. Human vascular smooth muscle cells undergo vesicle‐mediated calcification in response to changes in extracellular calcium and phosphate concentrations: A potential mechanism for accelerated vascular calcification in ESRD. J Am Soc Nephrol 15: 2857‐2867, 2004.
 160.Riccardi D, Brennan SC, Chang W. The extracellular calcium‐sensing receptor, CaSR, in fetal development. Best Pract Res Clin Endocrinol Metab 27: 443‐453, 2013.
 161.Ritter CS, Armbrecht HJ, Slatopolsky E, Brown AJ. 25‐Hydroxyvitamin D(3) suppresses PTH synthesis and secretion by bovine parathyroid cells. Kidney Int 70: 654‐659, 2006.
 162.Rodriguez‐Ortiz ME, Lopez I, Munoz‐Castaneda JR, Martinez‐Moreno JM, Ramirez AP, Pineda C, Canalejo A, Jaeger P, Aguilera‐Tejero E, Rodriguez M, Felsenfeld A, Almaden Y. Calcium deficiency reduces circulating levels of FGF23. J Am Soc Nephrol 23: 1190‐1197, 2012.
 163.Rossol M, Pierer M, Raulien N, Quandt D, Meusch U, Rothe K, Schubert K, Schoneberg T, Schaefer M, Krugel U, Smajilovic S, Brauner‐Osborne H, Baerwald C, Wagner U. Extracellular Ca2+ is a danger signal activating the NLRP3 inflammasome through G protein‐coupled calcium sensing receptors. Nat Commun 3: 1329, 2012.
 164.Rudnicki MA, Williams BO. Wnt signaling in bone and muscle. Bone 80: 60‐66, 2015.
 165.Russo D, Miranda I, Ruocco C, Battaglia Y, Buonanno E, Manzi S, Russo L, Scafarto A, Andreucci VE. The progression of coronary artery calcification in predialysis patients on calcium carbonate or sevelamer. Kidney Int 72: 1255‐1261, 2007.
 166.Saint‐Criq V, Rapetti‐Mauss R, Yusef YR, Harvey BJ. Estrogen regulation of epithelial ion transport: Implications in health and disease. Steroids 77: 918‐923, 2012.
 167.Satoh M, Nagasu H, Morita Y, Yamaguchi TP, Kanwar YS, Kashihara N. Klotho protects against mouse renal fibrosis by inhibiting Wnt signaling. Am J Physiol Renal Physiol 303: F1641‐F1651, 2012.
 168.Shalhoub V, Shatzen EM, Ward SC, Davis J, Stevens J, Bi V, Renshaw L, Hawkins N, Wang W, Chen C, Tsai MM, Cattley RC, Wronski TJ, Xia X, Li X, Henley C, Eschenberg M, Richards WG. FGF23 neutralization improves chronic kidney disease‐associated hyperparathyroidism yet increases mortality. J Clin Invest 122: 2543‐2553, 2012.
 169.Shanahan CM, Crouthamel MH, Kapustin A, Giachelli CM. Arterial calcification in chronic kidney disease: Key roles for calcium and phosphate. Circ Res 109: 697‐711, 2011.
 170.Shi Y, Gou BD, Shi YL, Zhang TL, Wang K. Lanthanum chloride suppresses hydrogen peroxide‐enhanced calcification in rat calcifying vascular cells. Biometals 22: 317‐327, 2009.
 171.Shimada T, Hasegawa H, Yamazaki Y, Muto T, Hino R, Takeuchi Y, Fujita T, Nakahara K, Fukumoto S, Yamashita T. FGF‐23 is a potent regulator of vitamin D metabolism and phosphate homeostasis. J Bone Miner Res 19: 429‐435, 2004.
 172.Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, Fukumoto S, Tomizuka K, Yamashita T. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J Clin Invest 113: 561‐568, 2004.
 173.Shimada T, Mizutani S, Muto T, Yoneya T, Hino R, Takeda S, Takeuchi Y, Fujita T, Fukumoto S, Yamashita T. Cloning and characterization of FGF23 as a causative factor of tumor‐induced osteomalacia. Proc Natl Acad Sci U S A 98: 6500‐6505, 2001.
 174.Shimizu T, Yoshitomi K, Nakamura M, Imai M. Effects of PTH, calcitonin, and cAMP on calcium transport in rabbit distal nephron segments. Am J Physiol 259: F408‐F414, 1990.
 175.Shroff RC, McNair R, Figg N, Skepper JN, Schurgers L, Gupta A, Hiorns M, Donald AE, Deanfield J, Rees L, Shanahan CM. Dialysis accelerates medial vascular calcification in part by triggering smooth muscle cell apoptosis. Circulation 118: 1748‐1757, 2008.
 176.Silva BC, Bilezikian JP. Parathyroid hormone: Anabolic and catabolic actions on the skeleton. Curr Opin Pharmacol 22: 41‐50, 2015.
 177.Silver J, Naveh‐Many T. Phosphate and the parathyroid. Kidney Int 75: 898‐905, 2009.
 178.Silver J, Naveh‐Many T, Mayer H, Schmelzer HJ, Popovtzer MM. Regulation by vitamin D metabolites of parathyroid hormone gene transcription in vivo in the rat. J Clin Invest 78: 1296‐1301, 1986.
 179.Slatopolsky E, Bricker NS. The role of phosphorus restriction in the prevention of secondary hyperparathyroidism in chronic renal disease. Kidney Int 4: 141‐145, 1973.
 180.Slatopolsky E, Finch J, Denda M, Ritter C, Zhong M, Dusso A, MacDonald PN, Brown AJ. Phosphorus restriction prevents parathyroid gland growth. High phosphorus directly stimulates PTH secretion in vitro. J Clin Invest 97: 2534‐2540, 1996.
 181.Slatopolsky E, Weerts C, Lopez‐Hilker S, Norwood K, Zink M, Windus D, Delmez J. Calcium carbonate as a phosphate binder in patients with chronic renal failure undergoing dialysis. N Engl J Med 315: 157‐161, 1986.
 182.Slatopolsky E, Weerts C, Norwood K, Giles K, Fryer P, Finch J, Windus D, Delmez J. Long‐term effects of calcium carbonate and 2.5 mEq/liter calcium dialysate on mineral metabolism. Kidney Int 36: 897‐903, 1989.
 183.Slatopolsky E, Weerts C, Thielan J, Horst R, Harter H, Martin KJ. Marked suppression of secondary hyperparathyroidism by intravenous administration of 1,25‐dihydroxy‐cholecalciferol in uremic patients. J Clin Invest 74: 2136‐2143, 1984.
 184.Smith RC, O'Bryan LM, Farrow EG, Summers LJ, Clinkenbeard EL, Roberts JL, Cass TA, Saha J, Broderick C, Ma YL, Zeng QQ, Kharitonenkov A, Wilson JM, Guo Q, Sun H, Allen MR, Burr DB, Breyer MD, White KE. Circulating alphaKlotho influences phosphate handling by controlling FGF23 production. J Clin Invest 122: 4710‐4715, 2012.
 185.Song S, Gao P, Xiao H, Xu Y, Si LY. Klotho suppresses cardiomyocyte apoptosis in mice with stress‐induced cardiac injury via downregulation of endoplasmic reticulum stress. PLoS One 8: e82968, 2013.
 186.Speer MY, Yang HY, Brabb T, Leaf E, Look A, Lin WL, Frutkin A, Dichek D, Giachelli CM. Smooth muscle cells give rise to osteochondrogenic precursors and chondrocytes in calcifying arteries. Circ Res 104: 733‐741, 2009.
 187.Spence LA, Lipscomb ER, Cadogan J, Martin B, Wastney ME, Peacock M, Weaver CM. The effect of soy protein and soy isoflavones on calcium metabolism in postmenopausal women: A randomized crossover study. Am J Clin Nutr 81: 916‐922, 2005.
 188.Sprague SM, Moe SM. The case for routine parathyroid hormone monitoring. Clin J Am Soc Nephrol 8: 313‐318, 2012.
 189.Stevens LA, Djurdjev O, Cardew S, Cameron EC, Levin A. Calcium, phosphate, and parathyroid hormone levels in combination and as a function of dialysis duration predict mortality: Evidence for the complexity of the association between mineral metabolism and outcomes. J Am Soc Nephrol 15: 770‐779, 2004.
 190.Sutton RA, Dirks JH. The renal excretion of calcium: A review of micropuncture data. Can J Physiol Pharmacol 53: 979‐988, 1975.
 191.Takasu H, Guo J, Bringhurst FR. Dual signaling and ligand selectivity of the human PTH/PTHrP receptor. J Bone Miner Res 14: 11‐20, 1999.
 192.Takeshita K, Fujimori T, Kurotaki Y, Honjo H, Tsujikawa H, Yasui K, Lee JK, Kamiya K, Kitaichi K, Yamamoto K, Ito M, Kondo T, Iino S, Inden Y, Hirai M, Murohara T, Kodama I, Nabeshima Y. Sinoatrial node dysfunction and early unexpected death of mice with a defect of klotho gene expression. Circulation 109: 1776‐1782, 2004.
 193.Talmage DW, Talmage RV. Calcium homeostasis: How bone solubility relates to all aspects of bone physiology. J Musculoskelet Neuronal Interact 7: 108‐112, 2007.
 194.Tentori F, Blayney MJ, Albert JM, Gillespie BW, Kerr PG, Bommer J, Young EW, Akizawa T, Akiba T, Pisoni RL, Robinson BM, Port FK. Mortality risk for dialysis patients with different levels of serum calcium, phosphorus, and PTH: The Dialysis Outcomes and Practice Patterns Study (DOPPS). Am J Kidney Dis 52: 519‐530, 2008.
 195.Toka HR, Pollak MR, Houillier P. Calcium sensing in the renal tubule. Physiology (Bethesda) 30: 317‐326, 2015.
 196.Tokumoto M, Mizobuchi M, Finch JL, Nakamura H, Martin DR, Slatopolsky E. Blockage of the renin‐angiotensin system attenuates mortality but not vascular calcification in uremic rats: Sevelamer carbonate prevents vascular calcification. Am J Nephrol 29: 582‐591, 2009.
 197.Torres PU, Prie D, Molina‐Bletry V, Beck L, Silve C, Friedlander G. Klotho: An antiaging protein involved in mineral and vitamin D metabolism. Kidney Int 71: 730‐737, 2007.
 198.Touchberry CD, Green TM, Tchikrizov V, Mannix JE, Mao TF, Carney BW, Girgis M, Vincent RJ, Wetmore LA, Dawn B, Bonewald LF, Stubbs JR, Wacker MJ. FGF23 is a novel regulator of intracellular calcium and cardiac contractility in addition to cardiac hypertrophy. Am J Physiol Endocrinol Metab 304: E863‐E873, 2013.
 199.Tyler Miller R. Control of renal calcium, phosphate, electrolyte, and water excretion by the calcium‐sensing receptor. Best Pract Res Clin Endocrinol Metab 27: 345‐358, 2013.
 200.Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, Fujita T, Fukumoto S, Yamashita T. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature 444: 770‐774, 2006.
 201.van de Graaf SF, Boullart I, Hoenderop JG, Bindels RJ. Regulation of the epithelial Ca2+ channels TRPV5 and TRPV6 by 1alpha,25‐dihydroxy vitamin D3 and dietary Ca2+. J Steroid Biochem Mol Biol 89‐90: 303‐308, 2004.
 202.van de Graaf SF, Rescher U, Hoenderop JG, Verkaart S, Bindels RJ, Gerke V. TRPV5 is internalized via clathrin‐dependent endocytosis to enter a Ca2+‐controlled recycling pathway. J Biol Chem 283: 4077‐4086, 2008.
 203.Wetmore JB, Santos PW, Mahnken JD, Krebill R, Menard R, Gutta H, Quarles LD. Elevated FGF23 levels are associated with impaired calcium‐mediated suppression of PTH in ESRD. J Clin Endocrinol Metab 96: E57‐64, 2011.
 204.White KE, Hum JM, Econs MJ. Hypophosphatemic rickets: Revealing novel control points for phosphate homeostasis. Curr Osteoporos Rep 12: 252‐262, 2014.
 205.Woudenberg‐Vrenken TE, Lameris AL, Weissgerber P, Olausson J, Flockerzi V, Bindels RJ, Freichel M, Hoenderop JG. Functional TRPV6 channels are crucial for transepithelial Ca2+ absorption. Am J Physiol Gastrointest Liver Physiol 303: G879‐G885, 2012.
 206.Xie J, Cha SK, An SW, Kuro OM, Birnbaumer L, Huang CL. Cardioprotection by Klotho through downregulation of TRPC6 channels in the mouse heart. Nat Commun 3: 1238, 2012.
 207.Xue Y, Karaplis AC, Hendy GN, Goltzman D, Miao D. Exogenous 1,25‐dihydroxyvitamin D3 exerts a skeletal anabolic effect and improves mineral ion homeostasis in mice that are homozygous for both the 1alpha‐hydroxylase and parathyroid hormone null alleles. Endocrinology 147: 4801‐4810, 2006.
 208.Xue Y, Xiao Y, Liu J, Karaplis AC, Pollak MR, Brown EM, Miao D, Goltzman D. The calcium‐sensing receptor complements parathyroid hormone‐induced bone turnover in discrete skeletal compartments in mice. Am J Physiol Endocrinol Metab 302: E841‐E851, 2012.
 209.Yeh BI, Kim YK, Jabbar W, Huang CL. Conformational changes of pore helix coupled to gating of TRPV5 by protons. EMBO J 24: 3224‐3234, 2005.
 210.Yeh BI, Sun TJ, Lee JZ, Chen HH, Huang CL. Mechanism and molecular determinant for regulation of rabbit transient receptor potential type 5 (TRPV5) channel by extracellular pH. J Biol Chem 278: 51044‐51052, 2003.
 211.Yu AS. Claudins and the kidney. J Am Soc Nephrol 26: 11‐19, 2015.
 212.Zella LA, Shevde NK, Hollis BW, Cooke NE, Pike JW. Vitamin D‐binding protein influences total circulating levels of 1,25‐dihydroxyvitamin D3 but does not directly modulate the bioactive levels of the hormone in vivo. Endocrinology 149: 3656‐3667, 2008.
 213.Zhou YB, Jin SJ, Cai Y, Teng X, Chen L, Tang CS, Qi YF. Lanthanum acetate inhibits vascular calcification induced by vitamin D3 plus nicotine in rats. Exp Biol Med (Maywood) 234: 908‐917, 2009.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Sharon M. Moe. Calcium Homeostasis in Health and in Kidney Disease. Compr Physiol 2016, 6: 1781-1800. doi: 10.1002/cphy.c150052