scholarly journals Epithelial Sodium Channel Inhibition by AMP-activated Protein Kinase in Oocytes and Polarized Renal Epithelial Cells

2005 ◽  
Vol 280 (18) ◽  
pp. 17608-17616 ◽  
Author(s):  
Marcelo D. Carattino ◽  
Robert S. Edinger ◽  
Heather J. Grieser ◽  
Rosalee Wise ◽  
Dietbert Neumann ◽  
...  
1999 ◽  
Vol 274 (33) ◽  
pp. 23286-23295 ◽  
Author(s):  
Jonathan B. Zuckerman ◽  
Xiyin Chen ◽  
Joely D. Jacobs ◽  
Baofeng Hu ◽  
Thomas R. Kleyman ◽  
...  

2005 ◽  
Vol 83 (11) ◽  
pp. 977-987 ◽  
Author(s):  
Toshiyuki Yamagata ◽  
Yuko Yamagata ◽  
Chantal Massé ◽  
Marie-Claude Tessier ◽  
Emmanuelle Brochiero ◽  
...  

Although the amiloride-sensitive epithelial sodium channel (ENaC) plays an important role in the modulation of alveolar liquid clearance, the precise mechanism of its regulation in alveolar epithelial cells is still under investigation. Protein kinase C (PKC) has been shown to alter ENaC expression and activity in renal epithelial cells, but much less is known about its role in alveolar epithelial cells. The objective of this study was to determine whether PKC activation modulates ENaC expression and transepithelial Na+ transport in cultured rat alveolar epithelial cells. Alveolar type II cells were isolated and cultured for 3 to 4 d before they were stimulated with phorbol 12-myristate 13-acetate (PMA 100 nmol/L) for 4 to 24 h. PMA treatment significantly decreased α, β, and γENaC expression in a time-dependent manner, whereas an inactive form of phorbol ester had no apparent effect. This inhibitory action was seen with only 5-min exposure to PMA, which suggested that PKC activation was very important for the reduction of αENaC expression. The PKC inhibitors bisindolylmaleimide at 2 µmol/L and Gö6976 at 2 µmol/L diminished the PMA-induced suppression of αENaC expression, while rottlerin at 1 µmol/L had no effect. PMA elicited a decrease in total and amiloride-sensitive current across alveolar epithelial cell monolayers. This decline in amiloride-sensitive current was not blocked by PKC inhibitors except for a partial inhibition with bisindolylmaleimide. PMA induced a decrease in rubidium uptake, indicating potential Na+-K+-ATPase inhibition. However, since ouabain-sensitive current in apically permeabilized epithelial cells was similar in PMA-treated and control cells, the inhibition was most probably related to reduced Na+ entry at the apical surface of the cells. We conclude that PKC activation modulates ENaC expression and probably ENaC activity in alveolar epithelial cells. Ca2+-dependent PKC is potentially involved in this response.Key words: alveolar epithelial cells, Na+ transport, Na+ channel, ENaC, protein kinase C, Na+-K+-ATPase, amiloride, gene expression.


2004 ◽  
Vol 279 (31) ◽  
pp. 32071-32078 ◽  
Author(s):  
Savita Mohan ◽  
Jennifer R. Bruns ◽  
Kelly M. Weixel ◽  
Robert S. Edinger ◽  
James B. Bruns ◽  
...  

2010 ◽  
Vol 299 (6) ◽  
pp. F1308-F1319 ◽  
Author(s):  
Rodrigo Alzamora ◽  
Fan Gong ◽  
Christine Rondanino ◽  
Jeffrey K. Lee ◽  
Christy Smolak ◽  
...  

The KCNQ1 K+ channel plays a key role in the regulation of several physiological functions, including cardiac excitability, cardiovascular tone, and body electrolyte homeostasis. The metabolic sensor AMP-activated protein kinase (AMPK) has been shown to regulate a growing number of ion transport proteins. To determine whether AMPK regulates KCNQ1, we studied the effects of AMPK activation on KCNQ1 currents in Xenopus laevis oocytes and collecting duct epithelial cells. AMPK activation decreased KCNQ1 currents and channel surface expression in X. laevis oocytes, but AMPK did not phosphorylate KCNQ1 in vitro, suggesting an indirect regulatory mechanism. As it has been recently shown that the ubiquitin-protein ligase Nedd4-2 inhibits KCNQ1 plasma membrane expression and that AMPK regulates epithelial Na+ channels via Nedd4-2, we examined the role of Nedd4-2 in the AMPK-dependent regulation of KCNQ1. Channel inhibition by AMPK was blocked in oocytes coexpressing either a dominant-negative or constitutively active Nedd4-2 mutant, or a Nedd4-2 interaction-deficient KCNQ1 mutant, suggesting that Nedd4-2 participates in the regulation of KCNQ1 by AMPK. KCNQ1 is expressed at the basolateral membrane in mouse polarized kidney cortical collecting duct (mpkCCDc14) cells and in rat kidney. Treatment with the AMPK activators AICAR (2 mM) or metformin (1 mM) reduced basolateral KCNQ1 currents in apically permeabilized polarized mpkCCDc14 cells. Moreover, AICAR treatment of rat kidney slices ex vivo induced AMPK activation and intracellular redistribution of KCNQ1 from the basolateral membrane in collecting duct principal cells. AICAR treatment also induced increased ubiquitination of KCNQ1 immunoprecipitated from kidney slice homogenates. These results indicate that AMPK inhibits KCNQ1 activity by promoting Nedd4-2-dependent channel ubiquitination and retrieval from the plasma membrane.


2011 ◽  
Vol 287 (7) ◽  
pp. 4451-4461 ◽  
Author(s):  
Hak Joo Lee ◽  
Meenalakshmi M. Mariappan ◽  
Denis Feliers ◽  
Rita C. Cavaglieri ◽  
Kavithalakshmi Sataranatarajan ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document