Rotavirus infection induces increased chloride secretion, altered barrier function and epidermal growth factor receptor (EGF-R) polyubiquitination in intestinal epithelial cells (IEC)

2001 ◽  
Vol 120 (5) ◽  
pp. A704-A705
Author(s):  
S RESTALENERT ◽  
K BARRETT
2011 ◽  
Vol 301 (5) ◽  
pp. C1008-C1016 ◽  
Author(s):  
Byong Kwon Yoo ◽  
Peijian He ◽  
Sei-Jung Lee ◽  
C. Chris Yun

Na+ absorption is a vital process present in all living organisms. We have reported previously that lysophosphatidic acid (LPA) acutely stimulates Na+ and fluid absorption in human intestinal epithelial cells and mouse intestine by stimulation of Na+/H+ exchanger 3 (NHE3) via LPA5 receptor. In the current study, we investigated the mechanism of NHE3 activation by LPA5 in Caco-2bbe cells. LPA5-dependent activation of NHE3 was blocked by mitogen-activated protein kinase kinase (MEK) inhibitor PD98059 and U0126, but not by phosphatidylinositol 3-kinase inhibitor LY294002 or phospholipase C-β inhibitor U73122. We found that LPA5 transactivated the epidermal growth factor receptor (EGFR) and that inhibition of EGFR blocked LPA5-dependent activation of NHE3, suggesting an obligatory role of EGFR in the NHE3 regulation. Confocal immunofluorescence and surface biotinylation analyses showed that LPA5 was located mostly in the apical membrane. EGFR, on the other hand, showed higher expression in the basolateral membrane. However, inhibition of apical EGFR, but not basolateral EGFR, abrogated LPA-induced regulation of MEK and NHE3, indicating that LPA5 selectively activates apical EGFR. Furthermore, transactivation of EGFR independently activated the MEK-ERK pathway and proline-rich tyrosine kinase 2 (Pyk2). Similarly to MEK inhibition, knockdown of Pyk2 blocked activation of NHE3 by LPA. Furthermore, we showed that RhoA and Rho-associated kinase (ROCK) are involved in activation of Pyk2. Interestingly, LPA5 did not directly activate RhoA but was required for transactivation of EGFR. Together, these results unveil a pivotal role of apical EGFR in NHE3 regulation by LPA and show that the RhoA-ROCK-Pyk2 and MEK-ERK pathways converge onto NHE3.


2007 ◽  
Vol 293 (3) ◽  
pp. G599-G606 ◽  
Author(s):  
George Sheng ◽  
Jun Guo ◽  
Brad W. Warner

Previous studies have demonstrated that the proapoptotic protein Bax plays an important role in the elevated enterocyte apoptosis that occurs during the intestinal adaptation response to massive small bowel resection (SBR). Additionally, epidermal growth factor receptor (EGFR) activation prevents SBR-induced enterocyte apoptosis. The present study aims to delineate the relationship between EGFR activity and intestinal epithelial cell apoptosis. Treatment of model intestinal epithelial cells (RIEC-18) with both a selective EGFR inhibitor (ZD1839) and EGFR small interfering RNA knockdown resulted in a dramatic increase in apoptosis, accompanied by rapid phosphorylation of p38α. Concurrently, Bax underwent conformational changes consistent with activation and translocated to mitochondria. In contrast, EGF stimulation enhanced cell survival by attenuating p38α phosphorylation, Bax conformational change, mitochondrial trafficking, and apoptosis. These results demonstrate that that diminished EGFR activity initiates the intrinsic pathway of apoptosis through p38α-dependent Bax activation in intestinal epithelial cells. These finding provide mechanistic insight into the role that EGFR signaling plays in the regulation of enterocyte apoptosis following massive intestinal loss.


Sign in / Sign up

Export Citation Format

Share Document