ASN's Mission

To create a world without kidney diseases, the ASN Alliance for Kidney Health elevates care by educating and informing, driving breakthroughs and innovation, and advocating for policies that create transformative changes in kidney medicine throughout the world.

learn more

Contact ASN

1401 H St, NW, Ste 900, Washington, DC 20005

email@asn-online.org

202-640-4660

The Latest on X

Kidney Week

Abstract: FR-PO286

TRPC1 Gene Deletion Disturbs Homeostasis of Intracellular Free Ca ([Ca 2+]i), Produces Hyperparathyroidism, Hypercalcemia, Hyperphosphatemia, and Increased Bone Mass Due to Renal Ca and P Retention

Session Information

Category: Mineral Disease

  • 1202 Mineral Disease: Vitamin D, PTH, FGF-23

Authors

  • Eby, Bonnie, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
  • Onopiuk, Marta, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
  • Humphrey, Marybeth, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
  • Tsiokas, Leonidas, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
  • Lau, Kai, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
Background

Mice deleted of the gene for transient receptor potential canonical channel 1 (TRPC1) have higher PTH (313 vs 218 pg/ml), hypercalcemia (11.3 vs.10.2 mg %) & hypocalciuria (1.2 vs 2.2 mg/d), indicating TRPC1 deficiency mimics familial hypocalciuric hypercalcemia phenotypes from CaSR inactivating mutations. Micro-CT shows increased tibia bone volume:tissue volume (11 vs 6 %). Their hind limbs were heavier (189 vs.151 mg). We now studied the potential mechanisms.

Methods

We measured Ca & P balance & clearance by standard methods, calciotropic hormones by ELISA, and [Ca 2+]i in cultured cells by fura 2.

Results

In rat parathyroid (PT) cells, [Ca 2+]i stimulation by CaSR allosteric agonists was markedly blunted by TRPC1 siRNA, showing CaSR signaling pathway depends on this store-operated Ca entry (SOCE). Without TRPC1, [Ca 2+]i is down & PTH secretion is up. In contrast, high medium [Ca] raises [Ca 2+]i & reduces PTH secretion. Transfection with plasmid overexpressing TRPC1 also inhibits PTH. The null mouse phenotypes cannot be attributed to the comparable serum calcitriol (301 vs.308 pg/ml) & calcitonin (21.9 vs.22.3 pg/ml). Renal cells transfected with TRPC1 siRNA & osteocytes from null mice also have blunted [Ca 2+]i response to CaSR agonists, indicating a generalized signaling defect in TRPC1-deficient cells. Since FGF23 synthesis &/or secretion are known to change directionally with [Ca 2+]I, we tested the thesis that P excretion is reduced in null mice, due to putatively low FGF23 from reduced [Ca 2+]i in osteocytes. At similar diet intake, 24 h urine P was similar (3.5 vs. 4.1 mg/d). Fasting serum P was elevated (7.4 vs. 6.3 mg %) due to concurrently decreased P clearance (48 vs. 67 µl/min).

Conclusion

Conclusions: 1. Our data support the model that, activated by upstream CaSR signaling, TRPC1 functions as a SOCE channel to control PT [Ca 2+]i and regulate PTH secretion. 2. Increased bone mineral accretion in TRPC1 deletion is mediated by greater renal Ca & P retention, the latter likely due to low FGF23 from reduced osteocyte [Ca 2+]i. 3. The apparent skeletal PTH resistance is explicable by the hypercalcemia, hyperphosphatemia, &/or the known impaired osteoclast proliferation, differentiation & function due to loss of TRPC1 functions.

Funding

  • NIDDK Support