SGK1 is a key component of the signaling machinery that regulates kidney tubule ion transport. It is under dual regulation by aldosterone, and the master kinase mTOR, which phosphorylates a specific serine (S422) within SGK1. The physiologically important signals that control mTOR-dependent SGK1 phosphorylation are not clear, and the regulatory mechanisms are unknown. Our preliminary data support the idea that both angiotensin II (Ang II) and local extracellular K+ concentration ([K+]) are important activators of SGK1, which act through one of the multi-subunit mTOR complexes, mTORC2, to stimulate SGK1 S422 phosphorylation and hence regulate ion transporters, particularly ENaC. The major goal of the present project is to elucidate the molecular mechanisms underlying this regulation, and to understand its physiological implications. We will: 1: We will first examine Ang II-stimulated selective regulation of SGK1 by mTORC2. We will identify residues within mTORC2 components SIN1 and Rictor that are phosphorylated in response to Ang II in cultured cells using mass spectrometry and immunoblot methods. We will then test mutants at these sites for their ability to support mTORC2-dependent SGK1 phosphorylation. We will examine the effect of Ang II on SGK1 subcellular localization and its interaction with mTORC2. 2: Characterize the effects of K+ on mTORC2-dependent SGK1 phosphorylation and its role in modulating ENaC and ROMK in cultured cells. Our preliminary data in cultured CCD cells and intact collecting duct support the idea that K+ modulates mTORC2 phosphorylation of SGK1 to regulate ENaC. We will explore both the mechanism and physiological implications of these findings in cultured cells, and extend to ROMK. We will examine the effect of altering [K+] within the physiologic range on mTORC2-dependent SGK1 phosphorylation and ENaC and ROMK currents in mpkCCD collecting duct cells grown on Transwell filters. We will also perform patch clamp on these cells to look directly at channel function in the apical membrane. We will characterize the signaling mechanisms implicated in K+ regulation of SGK1. 3: Characterize in vivo the role of mTORC2 in regulating Na+ and K+ excretion. In order to test key concepts from our in vitro experiments, and resolve discrepancies between recent publications, we will perform a series of in vivo experiments using pharmacologic inhibitors in WT and Rictor KO mice. We will compare the effects of pharmacologic inhibition of mTOR on Na+ and K+ handling in WT vs. distal nephron Rictor KO mice, and reconcile divergent results using electrolyte balance studies and patch clamp to assess ENaC and ROMK currents. Finally, we will examine the effects of acute vs. chronic loss of mTORC2 using an inducible KO model to compare acute and chronic Rictor deletion. These studies will shed new light on hormonal regulation of renal ion handling, and elucidate a novel mechanism for K+ to control its own excretion through direct effects in the aldosterone sensitive distal nephron/cortical collecting duct.

Public Health Relevance

The kidneys play an essential role in adjusting fluids and electrolytes and controlling blood pressure (BP). Hormonal systems are essential for this process, and when functioning normally permit adaptation to enormous variations in environmental water and salt, however, defects can result in severe abnormalities in BP, and cause edema. Our work will shed new light on how these systems are controlled, and suggest avenues toward intervention to correct abnormalities.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK056695-16
Application #
9727953
Study Section
Kidney Molecular Biology and Genitourinary Organ Development (KMBD)
Program Officer
Ketchum, Christian J
Project Start
2018-06-20
Project End
2022-03-31
Budget Start
2019-04-01
Budget End
2020-03-31
Support Year
16
Fiscal Year
2019
Total Cost
Indirect Cost
Name
University of California San Francisco
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
094878337
City
San Francisco
State
CA
Country
United States
Zip Code
94118
Morze, Cornelius von; Allu, Prasanna K R; Chang, Gene Y et al. (2018) Non-invasive detection of divergent metabolic signals in insulin deficiency vs. insulin resistance in vivo. Sci Rep 8:2088
von Morze, Cornelius; Chang, Gene-Yuan; Larson, Peder E Z et al. (2017) Detection of localized changes in the metabolism of hyperpolarized gluconeogenic precursors13C-lactate and13C-pyruvate in kidney and liver. Magn Reson Med 77:1429-1437
Rashmi, Priyanka; Colussi, GianLuca; Ng, Michael et al. (2017) Glucocorticoid-induced leucine zipper protein regulates sodium and potassium balance in the distal nephron. Kidney Int 91:1159-1177
Marco-Rius, Irene; Cao, Peng; von Morze, Cornelius et al. (2017) Multiband spectral-spatial RF excitation for hyperpolarized [2-13C]dihydroxyacetone13C-MR metabolism studies. Magn Reson Med 77:1419-1428
Lang, Florian; Pearce, David (2016) Regulation of the epithelial Na+ channel by the mTORC2/SGK1 pathway. Nephrol Dial Transplant 31:200-5
Gleason, Catherine E; Frindt, Gustavo; Cheng, Chih-Jen et al. (2015) mTORC2 regulates renal tubule sodium uptake by promoting ENaC activity. J Clin Invest 125:117-28
Suarez, Philippe Emmanuel; Rodriguez, Elena Gonzalez; Soundararajan, Rama et al. (2012) The glucocorticoid-induced leucine zipper (gilz/Tsc22d3-2) gene locus plays a crucial role in male fertility. Mol Endocrinol 26:1000-13
Soundararajan, Rama; Lu, Ming; Pearce, David (2012) Organization of the ENaC-regulatory machinery. Crit Rev Biochem Mol Biol 47:349-59
Soundararajan, Rama; Pearce, David; Ziera, Tim (2012) The role of the ENaC-regulatory complex in aldosterone-mediated sodium transport. Mol Cell Endocrinol 350:242-7
Schmid, Evi; Bhandaru, Madhuri; Nurbaeva, Meerim K et al. (2012) SGK3 regulates Ca(2+) entry and migration of dendritic cells. Cell Physiol Biochem 30:1423-35

Showing the most recent 10 out of 39 publications