1. Academic Validation
  2. Kidney glycolysis serves as a mammalian phosphate sensor that maintains phosphate homeostasis

Kidney glycolysis serves as a mammalian phosphate sensor that maintains phosphate homeostasis

  • J Clin Invest. 2023 Feb 23;e164610. doi: 10.1172/JCI164610.
Wen Zhou 1 Petra Simic 2 Iris Y Zhou 3 Peter Caravan 3 Xavier Vela Parada 1 Donghai Wen 1 Onica L Washington 1 Maria Shvedova 2 Kerry A Pierce 4 Clary B Clish 4 Michael Mannstadt 2 Tatsuya Kobayashi 2 Marc N Wein 2 Harald Jüppner 2 Eugene P Rhee 1
Affiliations

Affiliations

  • 1 Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, United States of America.
  • 2 Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, United States of America.
  • 3 Department of Radiology, Massachusetts General Hospital, Boston, United States of America.
  • 4 Metabolite Profiling Initiative, The Broad Institute of MIT and Harvard, Cambridge, United States of America.
Abstract

How phosphate levels are detected in mammals is unknown. The bone-derived hormone Fibroblast Growth Factor 23 (FGF23) lowers blood phosphate by reducing kidney phosphate reabsorption and 1,25(OH)2D production, but phosphate does not directly stimulate bone FGF23 expression. Using PET scanning and LC-MS, we show that phosphate increases kidney-specific glycolysis and synthesis of glycerol-3-phosphate (G-3-P), which then circulates to bone to trigger FGF23 production. Further, we find that glycerol-3-phosphate dehydrogenase 1 (Gpd1), a cytosolic Enzyme that synthesizes G-3-P and oxidizes NADH to NAD+, is required for phosphate-stimulated G-3-P and FGF23 production and prevention of hyperphosphatemia. In proximal tubule cells, we find that phosphate availability is substrate-limiting for glycolysis and G-3-P production, and that increased glycolysis and Gpd1 activity are coupled through cytosolic NAD+ recycling. Finally, we show that the type II sodium-dependent phosphate co-transporter Npt2a, which is expressed exclusively in the proximal tubule, confers kidney specificity to phosphate-stimulated G-3-P production. Importantly, exogenous G-3-P stimulates FGF23 production when Npt2a or Gpd1 are absent, confirming that it is the key circulating factor downstream of glycolytic phosphate sensing in the kidney. Together, these findings place glycolysis at the nexus of mineral and energy metabolism and identify a kidney-bone feedback loop that controls phosphate homeostasis.

Keywords

Calcium; Chronic kidney disease; Endocrinology; Glucose metabolism; Nephrology.

Figures
Products
Inhibitors & Agonists
Other Products