Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Calcium Extrusion Pump PMCA4: A New Player in Renal Calcium Handling?

  • Ellen P. M. van Loon,

    Affiliation Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands

  • Robert Little,

    Affiliation Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom

  • Sukhpal Prehar,

    Affiliation Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom

  • René J. M. Bindels,

    Affiliation Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands

  • Elizabeth J. Cartwright,

    Affiliation Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom

  • Joost G. J. Hoenderop

    Joost.Hoenderop@radboudumc.nl

    Affiliation Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands

Abstract

Calcium (Ca2+) is vital for multiple processes in the body, and maintenance of the electrolyte concentration is required for everyday physiological function. In the kidney, and more specifically, in the late distal convoluted tubule and connecting tubule, the fine-tuning of Ca2+ reabsorption from the pro-urine takes place. Here, Ca2+ enters the epithelial cell via the transient receptor potential vanilloid receptor type 5 (TRPV5) channel, diffuses to the basolateral side bound to calbindin-D28k and is extruded to the blood compartment via the Na+/Ca2+ exchanger 1 (NCX1) and the plasma membrane Ca2+ ATPase (PMCA). Traditionally, PMCA1 was considered to be the primary Ca2+ pump in this process. However, in recent studies TRPV5-expressing tubules were shown to highly express PMCA4. Therefore, PMCA4 may have a predominant role in renal Ca2+ handling. This study aimed to elucidate the role of PMCA4 in Ca2+ homeostasis by characterizing the Ca2+ balance, and renal and duodenal Ca2+-related gene expression in PMCA4 knockout mice. The daily water intake of PMCA4 knockout mice was significantly lower compared to wild type littermates. There was no significant difference in serum Ca2+ level or urinary Ca2+ excretion between groups. In addition, renal and duodenal mRNA expression levels of Ca2+-related genes, including TRPV5, TRPV6, calbindin-D28k, calbindin-D9k, NCX1 and PMCA1 were similar in wild type and knockout mice. Serum FGF23 levels were significantly increased in PMCA4 knockout mice. In conclusion, PMCA4 has no discernible role in normal renal Ca2+ handling as no urinary Ca2+ wasting was observed. Further investigation of the exact role of PMCA4 in the distal convoluted tubule and connecting tubule is required.

Introduction

Calcium (Ca2+) is involved in several important processes in the body, including muscle contraction, bone mineralization and as a second messenger in multiple signal transduction pathways [1]. As a consequence, the plasma Ca2+ concentration is tightly controlled via absorption of dietary Ca2+ at the intestine, storage in bone and reabsorption by the kidney [2, 3]. In the kidney the majority of filtered Ca2+ is passively reabsorbed in the proximal part of the nephron, though fine-tuning occurs in the late distal convoluted tubule (DCT) and the connecting tubule (CNT) [4]. This is an active process where Ca2+ reabsorption can be regulated by hormones including parathyroid hormone (PTH) and active vitamin D (1,25(OH)2D3) [57]. In the late DCT and CNT Ca2+ enters the cell from the pro-urine via the apically expressed transient receptor potential vanilloid channel type 5 (TRPV5) [8, 9]. Subsequently, Ca2+ binds to calbindin-D28k (CaBP28k) and/or calbindin-D9k (CaBP9k) and this complex diffuses to the basolateral side, where Ca2+ is extruded via the Na+/Ca2+ exchanger (NCX1) or plasma membrane Ca2+ ATPase (PMCA) [10, 11].

PMCA is a member of the P-type ATPase family, and is related to the sarcoplasmatic/endoplasmatic reticulum Ca2+ ATPase pumps. Four different genes encode for PMCA 1 to 4. Both PMCA1 and 4 are ubiquitously expressed and have been suggested to have a housekeeping function, extruding Ca2+ from the cell [12]. PMCA1 knockout (KO) mice are embryonically lethal, whereas the PMCA4 KO mice are viable and appear healthy [13, 14]. This suggests that PMCA4 might have a more specific role than PMCA1. Indeed, PMCA4 has been shown to play a role in Ca2+ signaling in sperm motility, B-lymphocytes and cardiac nitric oxide signaling [1316].

In the kidney, PMCA1 and 4 transcripts and protein are found in the proximal tubule, but higher expression has been shown at the distal part of the nephron [17, 18]. PMCA1 is considered as the predominant PMCA responsible for transcellular Ca2+ transport in the late DCT and CNT [18, 19]. Recently however, Alexander et al. investigated the exact localization of PMCA4 in the kidney, by co-staining the different tubular segments with representative markers [20]. They verified that PMCA4 was expressed highest in tubules that also expressed TRPV5. In addition, PMCA4 was decreased in TRPV5 KO mice, as were NCX1 and CaBP28k [21, 22]. On the contrary, PMCA1 was not changed [22]. This suggests that PMCA4 may be the predominant PMCA form involved in distal transcellular Ca2+ reabsorption.

In this study we assessed the role of PMCA4 in renal Ca2+ handling. To this end, the PMCA4 KO mouse model was used and its Ca2+ balance was compared to wild type (WT) and heterozygous (HZ) littermates. There were no significant differences in serum Ca2+ levels or urinary Ca2+ excretion between groups or changes in expression of Ca2+-related genes.

Material and Methods

Ethics statement

This study was carried out in strict compliance with the United Kingdom Animals (Scientific Procedures) Act 1986. All experimental procedures were approved by the University of Manchester Ethics Committee (permit-no: 40/3625) and all efforts were made to minimize suffering of animals. A completed ARRIVE guidelines checklist is included in S1 Checklist.

Animals

PMCA4 germline null mutant mice, as previously described [14], were maintained in a pathogen-free facility, housed under a 12 hour light/dark cycle, with ad libitum access to food and water. Experiments were performed with male WT (n = 10), HZ (n = 7) and KO (n = 10) mice when animals were aged 27–31 weeks old.

Immunofluorescence

Immunofluorescence staining was used to detect PMCA4 protein expression in kidney sections, and was performed as described previously [23]. In short, PMCA4 staining was performed on 5 μm thick sections of 1% (w/v) periodate-lysine-paraformaldehyde-fixed mouse kidney samples. Sections were incubated overnight at 4°C with mouse anti-PMCA4 (1:100, ab2783, Abcam, Cambridge, UK) and incubated with Alexa Fluor 488 conjugated secondary antibody (1:300, Invitrogen, Carlsbad, CA, USA) for 2 hours. Images were collected with an AxioCam camera and AxioVision software (Zeiss, Sliedrecht, The Netherlands).

Metabolic cage study

To collect urine, mice were housed individually in metabolic cages for 24 hours (from 10am to 10am) with ad libitum access to food and drinking water. The amount of water and food provided to each mouse was recorded at the start and after the 24-hour period, to determine amounts ingested. Following the metabolic cage housing, blood was collected via the jugular vein under isoflurane anesthesia and stored overnight at 4°C. All animals were humanely sacrificed by cervical dislocation. Kidneys were isolated, decapsulated, subdivided and snap frozen in liquid nitrogen. Duodenum segments were extracted, cleaned with PBS and snap frozen in liquid nitrogen. All samples were stored at -80°C until required. Serum was obtained following centrifugation of whole blood samples at 500g for 10 min, and frozen at -80°C until required. One KO mouse presented with fluid filled kidney cysts, and was not included in data analysis.

Analytical procedures

Urinary osmolality was measured using an Advanced® Model 3320 Micro-Osmometer (Advanced instruments Inc., Norwood, MA, USA). A colorimetric assay was used to determine serum and urine Ca2+ concentrations as described previously [22]. Ca2+ measurements were verified using an internal control, which was a commercial serum standard (Precinorm U, Roche, Basel, Switzerland). Serum and urinary phosphate (Pi) concentrations were determined by in-hospital services using automatic biochemical analyzers. Serum PTH levels were determined using the mouse PTH 1–84 ELISA kit (Immunotopics international, San Clemente, CA, USA) and serum Fibroblast Growth Factor (FGF)23 levels were determined with the mouse C-terminal FGF23 ELISA assay (Immunotopics International).

Quantitative real-time PCR

Total RNA was isolated from kidneys and duodenum using TRIzol® reagent (Invitrogen) according to the manufacturer’s protocol and dissolved in 150 μl diethylpyrocarbonate (DEPC)-treated deionized water, and stored at -80°C. Thereafter, RNA was treated with RQ1 DNase (1U, Promega, Madison, WI, USA) and reverse transcription was performed using 1.5 μg RNA and Moloney murine leukemia virus reverse transcriptase (200 U, Invitrogen) for 1.5 hours following the manufacturer’s protocol, except that RNasin® (Promega) was used as an RNase inhibitor [22]. The obtained cDNA was used to determine mRNA levels of several Ca2+-related genes, as well as glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as an endogenous control [22]. All the primers used (Biolegio, Nijmegen, The Netherlands) are listed in S1 Table. SYBR® Green mastermix (Bio-Rad, Veenendaal, The Netherlands) was used to perform quantitative real-time PCR according to manufacturer’s protocol and samples were measured on a CFX96 Bio-Rad analyzer (Bio-Rad). Gene expression was normalized for Gapdh expression and calculated according to the ΔΔCT method [24]. The PMCA4 real-time PCR product from WT, HZ and KO samples were run on a 2% (w/v) agarose gel electrophoresis.

Protein isolation from kidney and immunoblotting

Total kidney protein lysates were prepared as described previously [25] and 30 μg protein was loaded to either 8% (w/v) or 12% (w/v) SDS-PAGE gel for NCX1 and CaBP28k, respectively and blotted to a PVDF membrane (Millipore, Billerica, MA, USA). Immunoblots were incubated overnight at 4°C either with mouse anti-CaBP28k (1:5,000, clone CB-955, Sigma-Aldrich, Zwijndrecht, The Netherlands), mouse anti-NCX1 (1:500, ab6495, Abcam) or mouse anti-beta-actin (1:10,000, clone AC-15, Sigma-Aldrich). Immunoblots were enhanced using chemiluminescence (ECL, Pierce, Etten-Leur, The Netherlands) and analyzed using a ChemiDoc XRS (Bio-Rad) system. Semi-quantification was performed as described previously [26].

Statistics

Values are expressed as mean ± S.E.M. Differences between the WT, HZ and PMCA4 KO mice were tested using a one-way ANOVA with Tukey post-hoc test. Differences were considered significantly different when P<0.05. Analysis of the dataset was performed using GraphPad Prism, version 6.0.

Results

Similar serum Ca2+ levels and urinary Ca2+ excretion in WT and PMCA4 KO mice

The role of PMCA4 in renal Ca2+ handling was investigated using PMCA4 WT, HZ and KO mice. By quantitative real-time PCR and immunofluorescence staining of the kidney cortex, PMCA4 mRNA and protein were not detected in kidneys from PMCA4 KO mice (Fig 1). Before the start of the animal experiment mice of the different groups were of similar body weight (data not shown). Subsequently, mice were housed in metabolic cages to collect 24-hour urine. There was no significant difference in body weight, kidney weight, food intake, diuresis or osmolality between the different groups (Table 1). Water intake was significantly decreased between WT and KO (Table 1). To examine if PMCA4 KO mice develop disturbances in their Ca2+ homeostasis, serum Ca2+ levels and 24-hour urinary Ca2+ excretion were measured; however, both were similar in all three groups (Fig 2).

thumbnail
Fig 1. Confirmation of PMCA4 knockout in the kidney.

(A) Renal relative mRNA expression levels (corrected for GAPDH) of PMCA4 in wild type (WT, n = 10), heterozygous (HZ, n = 7) and knockout (KO, n = 10) mice. Data represents mean ± S.E.M. *P<0.05 compared to WT. Real-time PCR products were subjected to 2% (w/v) agarose gel electrophoresis. (B-D) Representative immunofluorescence image for PMCA4 expression in mouse kidney cortex of wild type (B) and PMCA4 knockout mice (C). (D) Negative control without the primary antibody.

https://doi.org/10.1371/journal.pone.0153483.g001

thumbnail
Fig 2. No difference in serum and urinary Ca2+ levels between the different PMCA4 genotypes.

Serum Ca2+ (A) and 24-hour urinary Ca2+ excretion (B) in wild type (WT, n = 10), heterozygous (HZ, n = 7) and PMCA4 knockout (KO, n = 10) mice. Data represents mean ± S.E.M.

https://doi.org/10.1371/journal.pone.0153483.g002

thumbnail
Table 1. Characteristics of PMCA4 wild type, heterozygous and knockout mice.

https://doi.org/10.1371/journal.pone.0153483.t001

No renal or duodenal compensation of Ca2+-related genes in PMCA4 KO mice

Renal mRNA expression of Ca2+-related genes was analyzed to determine possible compensation mechanisms following PMCA4 ablation. Expression of the epithelial Ca2+ channel TRPV5, the so-called Ca2+ gatekeeper of the late DCT and CNT [27], was not significantly different between groups (Fig 3A). In addition, other genes involved in Ca2+ reabsorption in the late DCT and CNT, including NCX1, PMCA1, CaBP28k and CaBP9k, were similar between the genotypes (Fig 3B–3E). In addition, renal protein expression of CaBP28k and NCX1 was not significantly different between groups (Fig 3F and 3G). Besides kidney, duodenal mRNA expression of Ca2+-related genes was measured to establish if potential compensation occurred in the duodenum. No difference was found for TRPV6, the active Ca2+ absorption channel in the duodenum [28] (Fig 4A). Moreover, NCX1, PMCA1 and CaBP9k were comparable between the three genotypes (Fig 4B–4D).

thumbnail
Fig 3. No effect of PMCA4 ablation on mRNA or protein levels of Ca2+-related genes in the kidney.

Relative mRNA expression levels (corrected for GAPDH) of TRPV5 (A), NCX1 (B), PMCA1 (C), calbindin-D28k (CaBP28k, D) and calbindin-D9k (CaBP9k, E) were determined in kidneys of wild type (WT, n = 10), heterozygous (HZ, n = 7) and PMCA4 knockout mice (KO, n = 10). (F) Representative immunoblot of renal CaBP28k and NCX1 protein expression in the three groups. Beta-actin was used as loading control. (G) Semi-quantification of immunoblots of CaBP28k and NCX1, corrected for beta-actin and compared to WT. Data represents mean ± S.E.M.

https://doi.org/10.1371/journal.pone.0153483.g003

thumbnail
Fig 4. No effect of PMCA4 ablation on mRNA expression of Ca2+-related genes in duodenum.

TRPV6 (A), NCX1 (B), PMCA1 (C) and calbindin-D9k (CaBP9k, D) relative mRNA expression levels (corrected for GAPDH) were analyzed in the duodenum of wild type (WT, n = 10), heterozygous (HZ, n = 7) and PMCA4 knockout mice (KO, n = 10). Data represents mean ± S.E.M.

https://doi.org/10.1371/journal.pone.0153483.g004

PMCA4 KO mice show increased FGF23 levels

In order to investigate whether hormonal Ca2+ regulation is changed in PMCA4 KO mice, PTH was measured in serum. No significant difference was present between the three groups of mice (Fig 5A). Moreover, renal mRNA expression of vitamin D activating (Cyp27b1) and breakdown (Cyp24a1) enzymes was investigated, and their expression was found not to significantly differ between genotypes (Fig 5B and 5C). Serum FGF23 levels were, however, significantly higher in PMCA4 KO mice compared to WT (Fig 4D). Since FGF23 is involved in regulating Pi homeostasis, serum Pi and 24-hour urinary Pi excretion were determined. There were, however, no significant differences between groups (S1 Fig). In addition, renal expression of klotho and Na+/Pi co-transporters NaPi-IIa and NaPi-IIc were determined, though also here no significant differences were observed (S1 Fig).

thumbnail
Fig 5.

PMCA4 ablation does not affect serum PTH but increases FGF23 serum levels (A) Serum PTH levels were measured in wild type (WT, n = 10), heterozygous (HZ, n = 7) and PMCA4 knockout mice (KO, n = 10). Relative mRNA expression levels (corrected for GAPDH) of the renal vitamin D activating enzyme Cyp27b1 (B) and vitamin D degrading enzyme Cyp24a1 (C). (D) Serum FGF23 levels were significantly higher in KO mice compared to WT. Data represents mean ± S.E.M, *P<0.05 compared to WT.

https://doi.org/10.1371/journal.pone.0153483.g005

Discussion

Importantly, for the first time, this study aimed to characterize the renal role of PMCA4. PMCA4 has recently been localized in Ca2+ transporting epithelial cells, promoting speculation about its functional role in transcellular Ca2+ transport at the distal nephron [20, 22]. In this study we show that ablation of PMCA4 has no significant effect on serum Ca2+ level or on renal Ca2+ excretion. Moreover, PMCA4 KO mice did not show Ca2+-related mRNA compensation in the kidney or duodenum.

The Ca2+ concentration in the body needs to be tightly regulated for multiple physiological processes [3, 29]. Disturbances in Ca2+ homeostasis can result in osteoporosis, heart failure or kidney stones [30, 31]. The late DCT and CNT are responsible for regulated active Ca2+ uptake, via the transcellular pathway. On the apical side, Ca2+ enters the cell via TRPV5, whereas NCX1 and PMCA1 and/or 4, located basolaterally, are responsible for Ca2+ extrusion [10,11, 22]. It was considered that PMCA1 was responsible for late DCT and CNT Ca2+ extrusion, since in the duodenum it is the principal Ca2+ efflux mechanism [19, 32]. However, Van der Hagen et al. showed that there was relatively more mRNA expression of PMCA4 in the late DCT and CNT compared to total kidney, whereas PMCA1 was not enriched in this segment [22]. This was confirmed at the protein level, where the most intense expression of PMCA4 in the kidney was observed in TRPV5 expressing tubules [20, 22]. In addition, the TRPV5 KO mouse showed a decrease in mRNA levels of CaBP28k, NCX1 and PMCA4, but not PMCA1 [22]. Furthermore, parathyroidectomized rats, klotho KO mice and 1α-hydroxylase KO mice showed co-regulation of TRPV5, CaBP28k and NCX1, but not of PMCA1 [5, 6, 33]. Therefore, we hypothesized that PMCA4, rather than PMCA1, is the predominant pump for renal Ca2+ homeostasis in the late DCT and CNT. However, in this study the PMCA4 KO mice did not show an increase in urinary Ca2+ excretion. PMCA1 and/or NCX1 might have compensated for the ablation of PMCA4, normalizing serum Ca2+ and urinary Ca2+ excretion. However, there was no significant upregulation of NCX1 at gene and protein level, or increased mRNA expression for PMCA1 in PMCA4 KO mice. Total ablation of either NCX1 or PMCA1 is embryonic lethal in mice, hence their role in Ca2+ extrusion in late DCT and CNT cannot be determined by using a global KO [13, 34]. On the contrary, TRPV5 KO mice are viable but present with severe hypercalciuria [21, 35]. Mice with depletion of the Ca2+ shuttling protein CaBP28k show varying results concerning their Ca2+ excretion, with no difference or increased Ca2+ excretion compared to WT mice, depending on the study [27, 3638]. In addition, Gkika et al. treated CaBP28k KO mice with a low and high Ca2+ diet (0.02% w/w or 2% w/w), and did not observe a disturbance in Ca2+ homeostasis, even though CaBP28k is involved in transcellular Ca2+ transport [27]. This suggests that we cannot necessarily rule out PMCA4 having a role in renal Ca2+ handling even though the PMCA4 KO mouse does not show a changed Ca2+ homeostasis.

At the intestinal level the PMCA4 KO mice did not show a difference in the Ca2+-regulating genes, indicating that no compensation occurred. Recently Alexander et al., have shown that PMCA4 is located in the smooth muscle layer of the intestine, and not in the enterocytes, where PMCA1 was more highly detected [20], suggesting that PMCA1, rather than PMCA4, is primarily involved in transcellular Ca2+ transport in the duodenum [20, 28, 39]. An earlier study has shown that PMCA4 KO mice presented with lower trabecular bone volume, lower bone mineral density and an increase in osteoclast surface area compared to WT mice [40]. Bone could be a source of Ca2+ to maintain a normal serum Ca2+ level; however, there was no indication of renal Ca2+ wasting in our mice studied here.

Ca2+ homeostasis is mainly regulated by PTH and 1,25(OH)2D3 in response to low serum Ca2+ levels [41]. The PMCA4 KO mice do not show differences in Cyp27b1 or Cyp24a1 mRNA, nor in serum PTH, compared to WT mice. This is consistent with our other observations where PTH- and 1,25(OH)2D3-regulated genes, such as TRPV5 and TRPV6, are not changed [5, 6, 42]. Moreover, PMCA4 KO mice presented with significantly lower water intake. However, since there were no changes in body weight or urinary excretion, this could have been caused by biological variation. In addition, we know of no reports indicating an involvement of PMCA4 in thirst and/or water homeostasis. Interestingly, FGF23 levels were increased in PMCA4 KO mice. In the kidney, FGF23 decreases NaPi-IIa and NaPi-IIc expression, resulting in increased urinary phosphate excretion [43]. In addition, it downregulates Cyp27b1 and increases Cyp24a1 expression [44]. These consequences were not observed in PMCA4 KO mice. This suggests that PMCA4 might be involved in FGF23 production and/or signaling, although more research is necessary to confirm this. Besides its role as general Ca2+ extruder, others have already shown the involvement of PMCA4 in signaling transducing pathways as well [45, 46].

In conclusion, PMCA4 does not seem to be vital for basal renal Ca2+ handling. However, the presence of PMCA4 in specific segments of the kidney points to the molecule having a different or additional role compared to PMCA1 and therefore, their exact roles should be investigated in more detail.

Supporting Information

S1 Fig. No difference in serum Pi or 24-hour urinary Pi excretion between the different genotypes.

Serum Pi (A) and 24-hour urinary Pi excretion (B) in wild type (WT, n = 10), heterozygous (HZ, n = 7) and knockout (KO, n = 10) mice. Relative mRNA expression of NaPi-IIa (C), NaPi-IIc (D) and klotho (E) were determined in the kidney. Data represents mean ± S.E.M.

https://doi.org/10.1371/journal.pone.0153483.s003

(TIF)

S1 Table. Primer sequences used for real-time PCR.

https://doi.org/10.1371/journal.pone.0153483.s004

(DOCX)

Author Contributions

Conceived and designed the experiments: EPML RL RJMB EJC JGJH. Performed the experiments: EPML RL SP. Analyzed the data: EPML RL RJMB EJC JGJH. Contributed reagents/materials/analysis tools: EPML RL RJMB EJC JGJH. Wrote the paper: EPML RL RJMB EJC JGJH.

References

  1. 1. Hoenderop JG, Bindels RJ. Epithelial Ca2+ and Mg2+ channels in health and disease. J Am Soc Nephrol. 2005;16(1):15–26. Epub 2004/12/03. pmid:15574510.
  2. 2. Bushinsky DA. Contribution of intestine, bone, kidney, and dialysis to extracellular fluid calcium content. Clin J Am Soc Nephrol. 2010;5 Suppl 1:S12–22. Epub 2010/02/03. pmid:20089498.
  3. 3. Renkema KY, Alexander RT, Bindels RJ, Hoenderop JG. Calcium and phosphate homeostasis: concerted interplay of new regulators. Ann Med. 2008;40(2):82–91. Epub 2008/02/23. pmid:18293139.
  4. 4. Dimke H, Hoenderop JG, Bindels RJ. Hereditary tubular transport disorders: implications for renal handling of Ca2+ and Mg2+. Clin Sci (Lond). 2010;118(1):1–18. Epub 2009/09/29. pmid:19780717.
  5. 5. Hoenderop JG, Dardenne O, Van Abel M, Van Der Kemp AW, Van Os CH, St -Arnaud R, et al. Modulation of renal Ca2+ transport protein genes by dietary Ca2+ and 1,25-dihydroxyvitamin D3 in 25-hydroxyvitamin D3-1alpha-hydroxylase knockout mice. FASEB J. 2002;16(11):1398–406. Epub 2002/09/03. pmid:12205031.
  6. 6. van Abel M, Hoenderop JG, van der Kemp AW, Friedlaender MM, van Leeuwen JP, Bindels RJ. Coordinated control of renal Ca(2+) transport proteins by parathyroid hormone. Kidney Int. 2005;68(4):1708–21. Epub 2005/09/17. pmid:16164647.
  7. 7. Lambers TT, Bindels RJ, Hoenderop JG. Coordinated control of renal Ca2+ handling. Kidney Int. 2006;69(4):650–4. Epub 2006/03/07. pmid:16518325.
  8. 8. Hoenderop JG, van der Kemp AW, Hartog A, van de Graaf SF, van Os CH, Willems PH, et al. Molecular identification of the apical Ca2+ channel in 1, 25-dihydroxyvitamin D3-responsive epithelia. J Biol Chem. 1999;274(13):8375–8. Epub 1999/03/20. pmid:10085067.
  9. 9. Nilius B, Prenen J, Hoenderop JG, Vennekens R, Hoefs S, Weidema AF, et al. Fast and slow inactivation kinetics of the Ca2+ channels ECaC1 and ECaC2 (TRPV5 and TRPV6). Role of the intracellular loop located between transmembrane segments 2 and 3. J Biol Chem. 2002;277(34):30852–8. Epub 2002/06/22. pmid:12077127.
  10. 10. Hoenderop JG, Nilius B, Bindels RJ. Molecular mechanism of active Ca2+ reabsorption in the distal nephron. Annu Rev Physiol. 2002;64:529–49. Epub 2002/02/05. pmid:11826278.
  11. 11. Brini M, Carafoli E. The plasma membrane Ca(2)+ ATPase and the plasma membrane sodium calcium exchanger cooperate in the regulation of cell calcium. Cold Spring Harb Perspect Biol. 2011;3(2). Epub 2011/03/23. pmid:21421919; PubMed Central PMCID: PMC3039526.
  12. 12. Stauffer TP, Hilfiker H, Carafoli E, Strehler EE. Quantitative analysis of alternative splicing options of human plasma membrane calcium pump genes. J Biol Chem. 1993;268(34):25993–6003. Epub 1993/12/05. pmid:8245032.
  13. 13. Okunade GW, Miller ML, Pyne GJ, Sutliff RL, O'Connor KT, Neumann JC, et al. Targeted ablation of plasma membrane Ca2+-ATPase (PMCA) 1 and 4 indicates a major housekeeping function for PMCA1 and a critical role in hyperactivated sperm motility and male fertility for PMCA4. J Biol Chem. 2004;279(32):33742–50. Epub 2004/06/05. pmid:15178683.
  14. 14. Schuh K, Cartwright EJ, Jankevics E, Bundschu K, Liebermann J, Williams JC, et al. Plasma membrane Ca2+ ATPase 4 is required for sperm motility and male fertility. J Biol Chem. 2004;279(27):28220–6. Epub 2004/04/14. pmid:15078889.
  15. 15. Mohamed TM, Oceandy D, Zi M, Prehar S, Alatwi N, Wang Y, et al. Plasma membrane calcium pump (PMCA4)-neuronal nitric-oxide synthase complex regulates cardiac contractility through modulation of a compartmentalized cyclic nucleotide microdomain. J Biol Chem. 2011;286(48):41520–9. Epub 2011/10/04. pmid:21965681; PubMed Central PMCID: PMC3308863.
  16. 16. Chen J, McLean PA, Neel BG, Okunade G, Shull GE, Wortis HH. CD22 attenuates calcium signaling by potentiating plasma membrane calcium-ATPase activity. Nat Immunol. 2004;5(6):651–7. Epub 2004/05/11. pmid:15133509.
  17. 17. White KE, Gesek FA, Nesbitt T, Drezner MK, Friedman PA. Molecular dissection of Ca2+ efflux in immortalized proximal tubule cells. J Gen Physiol. 1997;109(2):217–28. Epub 1997/02/01. pmid:9041450; PubMed Central PMCID: PMC2220068.
  18. 18. Van Baal J, Yu A, Hartog A, Fransen JA, Willems PH, Lytton J, et al. Localization and regulation by vitamin D of calcium transport proteins in rabbit cortical collecting system. Am J Physiol. 1996;271(5 Pt 2):F985–93. Epub 1996/11/01. pmid:8945992.
  19. 19. Hoenderop JG, Willems PH, Bindels RJ. Toward a comprehensive molecular model of active calcium reabsorption. Am J Physiol Renal Physiol. 2000;278(3):F352–60. Epub 2000/03/11. pmid:10710538.
  20. 20. Alexander RT, Beggs MR, Zamani R, Marcussen N, Frische S, Dimke H. Ultrastructural and Immunohistochemical Localization of the Plasma Membrane Ca2+-ATPase 4 (PMCA4) in Ca2+-Transporting Epithelia. Am J Physiol Renal Physiol. 2015:ajprenal 00651 2014. Epub 2015/07/17. pmid:26180241.
  21. 21. Hoenderop JG, van Leeuwen JP, van der Eerden BC, Kersten FF, van der Kemp AW, Merillat AM, et al. Renal Ca2+ wasting, hyperabsorption, and reduced bone thickness in mice lacking TRPV5. J Clin Invest. 2003;112(12):1906–14. Epub 2003/12/18. pmid:14679186; PubMed Central PMCID: PMC297001.
  22. 22. van der Hagen EA, Lavrijsen M, van Zeeland F, Praetorius J, Bonny O, Bindels RJ, et al. Coordinated regulation of TRPV5-mediated Ca(2)(+) transport in primary distal convolution cultures. Pflugers Arch. 2014;466(11):2077–87. Epub 2014/02/22. pmid:24557712.
  23. 23. Hoenderop JG, Hartog A, Stuiver M, Doucet A, Willems PH, Bindels RJ. Localization of the epithelial Ca(2+) channel in rabbit kidney and intestine. J Am Soc Nephrol. 2000;11(7):1171–8. Epub 2000/06/23. pmid:10864572.
  24. 24. van Loon EP, Pulskens WP, van der Hagen EA, Lavrijsen M, Vervloet MG, van Goor H, et al. Shedding of klotho by ADAMs in the kidney. Am J Physiol Renal Physiol. 2015;309(4):F359–68. Epub 2015/07/15. pmid:26155844.
  25. 25. Pulskens WP, Verkaik M, Sheedfar F, van Loon EP, van de Sluis B, Vervloet MG, et al. Deregulated Renal Calcium and Phosphate Transport during Experimental Kidney Failure. PLoS One. 2015;10(11):e0142510. Epub 2015/11/14. pmid:26566277; PubMed Central PMCID: PMC4643984.
  26. 26. Huybers S, Apostolaki M, van der Eerden BC, Kollias G, Naber TH, Bindels RJ, et al. Murine TNF(DeltaARE) Crohn's disease model displays diminished expression of intestinal Ca2+ transporters. Inflamm Bowel Dis. 2008;14(6):803–11. Epub 2008/02/13. pmid:18266230.
  27. 27. Gkika D, Hsu YJ, van der Kemp AW, Christakos S, Bindels RJ, Hoenderop JG. Critical role of the epithelial Ca2+ channel TRPV5 in active Ca2+ reabsorption as revealed by TRPV5/calbindin-D28K knockout mice. J Am Soc Nephrol. 2006;17(11):3020–7. Epub 2006/09/29. pmid:17005931.
  28. 28. Lameris AL, Nevalainen PI, Reijnen D, Simons E, Eygensteyn J, Monnens L, et al. Segmental transport of Ca(2)(+) and Mg(2)(+) along the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol. 2015;308(3):G206–16. Epub 2014/12/06. pmid:25477372.
  29. 29. Woudenberg-Vrenken TE, Bindels RJ, Hoenderop JG. The role of transient receptor potential channels in kidney disease. Nat Rev Nephrol. 2009;5(8):441–9. Epub 2009/06/24. pmid:19546862.
  30. 30. Hoorn EJ, Zietse R. Disorders of calcium and magnesium balance: a physiology-based approach. Pediatr Nephrol. 2013;28(8):1195–206. Epub 2012/11/13. pmid:23142866.
  31. 31. Renkema KY, Velic A, Dijkman HB, Verkaart S, van der Kemp AW, Nowik M, et al. The calcium-sensing receptor promotes urinary acidification to prevent nephrolithiasis. J Am Soc Nephrol. 2009;20(8):1705–13. Epub 2009/05/28. pmid:19470676; PubMed Central PMCID: PMC2723980.
  32. 32. Howard A, Legon S, Walters JR. Human and rat intestinal plasma membrane calcium pump isoforms. Am J Physiol. 1993;265(5 Pt 1):G917–25. Epub 1993/11/01. pmid:7694502.
  33. 33. Alexander RT, Woudenberg-Vrenken TE, Buurman J, Dijkman H, van der Eerden BC, van Leeuwen JP, et al. Klotho prevents renal calcium loss. J Am Soc Nephrol. 2009;20(11):2371–9. Epub 2009/08/29. pmid:19713312; PubMed Central PMCID: PMC2799169.
  34. 34. Cho CH, Kim SS, Jeong MJ, Lee CO, Shin HS. The Na+ -Ca2+ exchanger is essential for embryonic heart development in mice. Mol Cells. 2000;10(6):712–22. Epub 2001/02/24. pmid:11211878.
  35. 35. Renkema KY, Nijenhuis T, van der Eerden BC, van der Kemp AW, Weinans H, van Leeuwen JP, et al. Hypervitaminosis D mediates compensatory Ca2+ hyperabsorption in TRPV5 knockout mice. J Am Soc Nephrol. 2005;16(11):3188–95. Epub 2005/09/09. pmid:16148038.
  36. 36. Zheng W, Xie Y, Li G, Kong J, Feng JQ, Li YC. Critical role of calbindin-D28k in calcium homeostasis revealed by mice lacking both vitamin D receptor and calbindin-D28k. J Biol Chem. 2004;279(50):52406–13. Epub 2004/10/01. pmid:15456794.
  37. 37. Lee CT, Huynh VM, Lai LW, Lien YH. Cyclosporine A-induced hypercalciuria in calbindin-D28k knockout and wild-type mice. Kidney Int. 2002;62(6):2055–61. Epub 2002/11/13. pmid:12427129.
  38. 38. Sooy K, Kohut J, Christakos S. The role of calbindin and 1,25dihydroxyvitamin D3 in the kidney. Curr Opin Nephrol Hypertens. 2000;9(4):341–7. Epub 2000/08/05. pmid:10926169.
  39. 39. Ryan ZC, Craig TA, Filoteo AG, Westendorf JJ, Cartwright EJ, Neyses L, et al. Deletion of the intestinal plasma membrane calcium pump, isoform 1, Atp2b1, in mice is associated with decreased bone mineral density and impaired responsiveness to 1, 25-dihydroxyvitamin D. Biochem Biophys Res Commun. 2015. Epub 2015/09/24. pmid:26392310.
  40. 40. Kim HJ, Prasad V, Hyung SW, Lee ZH, Lee SW, Bhargava A, et al. Plasma membrane calcium ATPase regulates bone mass by fine-tuning osteoclast differentiation and survival. J Cell Biol. 2012;199(7):1145–58. Epub 2012/12/26. pmid:23266958; PubMed Central PMCID: PMC3529522.
  41. 41. Boden SD, Kaplan FS. Calcium homeostasis. Orthop Clin North Am. 1990;21(1):31–42. Epub 1990/01/01. pmid:2404236.
  42. 42. Bouillon R, Van Cromphaut S, Carmeliet G. Intestinal calcium absorption: Molecular vitamin D mediated mechanisms. J Cell Biochem. 2003;88(2):332–9. Epub 2003/01/10. pmid:12520535.
  43. 43. Gattineni J, Bates C, Twombley K, Dwarakanath V, Robinson ML, Goetz R, et al. FGF23 decreases renal NaPi-2a and NaPi-2c expression and induces hypophosphatemia in vivo predominantly via FGF receptor 1. Am J Physiol Renal Physiol. 2009;297(2):F282–91. Epub 2009/06/12. pmid:19515808; PubMed Central PMCID: PMC2724258.
  44. 44. Bai X, Miao D, Li J, Goltzman D, Karaplis AC. Transgenic mice overexpressing human fibroblast growth factor 23 (R176Q) delineate a putative role for parathyroid hormone in renal phosphate wasting disorders. Endocrinology. 2004;145(11):5269–79. Epub 2004/07/31. pmid:15284207.
  45. 45. Buch MH, Pickard A, Rodriguez A, Gillies S, Maass AH, Emerson M, et al. The sarcolemmal calcium pump inhibits the calcineurin/nuclear factor of activated T-cell pathway via interaction with the calcineurin A catalytic subunit. J Biol Chem. 2005;280(33):29479–87. Epub 2005/06/16. pmid:15955804.
  46. 46. Cartwright EJ, Oceandy D, Austin C, Neyses L. Ca2+ signalling in cardiovascular disease: the role of the plasma membrane calcium pumps. Sci China Life Sci. 2011;54(8):691–8. Epub 2011/07/26. pmid:21786192.