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

Please note that you are viewing an archived section from 2022 and some content may be unavailable. To unlock all content for 2022, please visit the archives.

Abstract: TH-PO347

WNK1 Is a Chloride-Sensing Scaffold That Potently Regulates mTORC2 Activity and ENaC

Session Information

Category: Fluid‚ Electrolyte‚ and Acid-Base Disorders

  • 1001 Fluid‚ Electrolyte‚ and Acid-Base Disorders: Basic

Authors

  • Saha, Bidisha, University of California San Francisco, San Francisco, California, United States
  • Leite-Dellova, Deise C A, University of California San Francisco, San Francisco, California, United States
  • Takagi, Enzo, University of California San Francisco, San Francisco, California, United States
  • Demko, John E., University of California San Francisco, San Francisco, California, United States
  • Shabbir, Waheed, University of California San Francisco, San Francisco, California, United States
  • Pearce, David, University of California San Francisco, San Francisco, California, United States
Background

mTORC2 is a multiprotein signaling complex that regulates a variety of cellular processes induced by hormones and growth factors. Recent evidence suggests that mTORC2 is also involved in cell autonomous action, as it responds to local extracellular K+ concentration to rapidly stimulate SGK1 phosphorylation and downstream target, ENaC, an epithelial sodium channel, in renal epithelial cells. Here we show that WNK1 has a pivotal role in mediating cell autonomous responses to local K+ concentration that activates mTORC2.

Methods

We generated WNK1 knockout mpkCCD cells and WNK1 and SIN1 (a component of mTORC2 complex) double knockout HEK293 cells using CRISPR/Cas9 system. mpkCCD cells were grown on Transwell filters and incubated in media containing different [K+], with or without WNK kinase inhibitor. Amiloride-sensitive current was measured before the cells were processed for immunoblot analysis. For intracellular Cl- measurement, HEK293 cells were transfected with mCl-YFP plasmid and fluorescence signals were measured and analyzed. For modulation of intracellular [Cl-], cells were incubated in media containing different [Cl-] and ionophores. Cells were then processed for immunoblot and Co-IP analysis.

Results

Our results showed that the effect of extracellular K+ on SGK1 phosphorylation and ENaC activity is dependent on elevated intracellular [Cl-] and on presence of chloride-binding protein kinase WNK1, but not on its kinase activity. Intracellular Cl- stimulates the scaffolding activity of WNK1, which physically interacts with both SGK1 and mTORC2 and enhances SGK1 recruitment to mTORC2. This results in a selective increase in SGK1 phosphorylation, and activation of ENaC which drives K+ secretion.

Conclusion

These findings establish a novel mode of chloride-regulated WNK1 action that regulates mTORC2 activity selectively towards SGK1 phosphorylation and ENaC activation, independently of its kinase activity and reveals a dual role of WNK1’s Cl--sensing property on epithelial ion transport regulation: one, low [Cl-]-dependent activation of NCC through catalytic mechanism and the second, high [Cl-]-dependent activation of ENaC through non-catalytic scaffolding mechanism; the coupling of both pathways is significantly implicated in DCT2, controlling K+ secretion.

Funding

  • NIDDK Support