Obligatory role for phospholipase C-γ1 in villin-induced epithelial cell migration

2007 ◽  
Vol 292 (5) ◽  
pp. C1775-C1786 ◽  
Author(s):  
Yaohong Wang ◽  
Alok Tomar ◽  
Sudeep P. George ◽  
Seema Khurana

While there is circumstantial evidence to suggest a requirement for phospholipase C-γ1 (PLC-γ1) in actin reorganization and cell migration, few studies have examined the direct mechanisms that link regulators of the actin cytoskeleton with this crucial signaling molecule. This study was aimed to examine the role that villin, an epithelial cell-specific actin-binding protein, and its ligand PLC-γ1 play in migration in intestinal and renal epithelial cell lines that endogenously or ectopically express human villin. Basal as well as epidermal growth factor (EGF)-stimulated cell migration was accompanied by tyrosine phosphorylation of villin and its association with PLC-γ1. Inhibition of villin phosphorylation prevented villin-PLC-γ1 complex formation as well as villin-induced cell migration. The absolute requirement for PLC-γ1 in villin-induced cell migration was demonstrated by measuring cell motility in PLC-γ1−/− cells and by downregulation of endogenous PLC-γ1. EGF-stimulated direct interaction of villin with the Src homology domain 2 domain of PLC-γ1 at the plasma membrane was demonstrated in living cells by using fluorescence resonance energy transfer. These results demonstrate that villin provides an important link between the activation of phosphoinositide signal transduction pathway and epithelial cell migration.

2001 ◽  
Vol 9 (2) ◽  
pp. 86-94 ◽  
Author(s):  
James D Firth ◽  
Edward E Putnins ◽  
Hannu Larjava ◽  
Veli-Jukka Uitto

2006 ◽  
Vol 281 (42) ◽  
pp. 31972-31986
Author(s):  
Alok Tomar ◽  
Sudeep George ◽  
Pallavi Kansal ◽  
Yaohong Wang ◽  
Seema Khurana

2008 ◽  
Vol 295 (6) ◽  
pp. C1499-C1509 ◽  
Author(s):  
Jaladanki N. Rao ◽  
Stephen V. Liu ◽  
Tongtong Zou ◽  
Lan Liu ◽  
Lan Xiao ◽  
...  

Intestinal mucosal restitution occurs as a consequence of epithelial cell migration and reseals superficial wounds after injury. This rapid reepithelialization is mediated in part by a phospholipase C-γ1 (PLC-γ1)-induced Ca2+ signaling, but the exact mechanism underlying such signaling and its regulation remains elusive. The small GTP-binding protein Rac1 functions as a pivotal regulator of several signaling networks and plays an important role in regulating cell motility. The current study tests the hypothesis that Rac1 modulates intestinal epithelial cell migration after wounding by altering PLC-γ1-induced Ca2+ signaling. Inhibition of Rac1 activity by treatment with its inhibitor NSC-23766 or Rac1 silencing with small interfering RNA decreased store depletion-induced Ca2+ influx and suppressed cell migration during restitution, whereas ectopic overexpression of Rac1 increased Ca2+ influx and promoted cell migration. Rac1 physically interacted with PLC-γ1 and formed Rac1/PLC-γ1 complex in intestinal epithelial cells. PLC-γ1 silencing in cells overexpressing Rac1 prevented stimulation of store depletion-induced Ca2+ influx and cell migration after wounding. Polyamine depletion inhibited expression of both Rac1 and PLC-γ1, decreased Rac1/PLC-γ1 complex levels, reduced Ca2+ influx, and repressed cell migration. Overexpression of Rac1 alone failed to rescue Ca2+ influx after store depletion and cell migration in polyamine-deficient cells, because it did not alter PLC-γ1 levels. These results indicate that Rac1 promotes intestinal epithelial cell migration after wounding by increasing Ca2+ influx as a result of its interaction with PLC-γ1.


2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Simon Müller ◽  
Sonja Djudjaj ◽  
Janina Lange ◽  
Mihail Iacovescu ◽  
Margarete Goppelt-Struebe ◽  
...  

2007 ◽  
Vol 292 (1) ◽  
pp. G335-G343 ◽  
Author(s):  
Jaladanki N. Rao ◽  
Lan Liu ◽  
Tongtong Zou ◽  
Bernard S. Marasa ◽  
Dessy Boneva ◽  
...  

Intestinal mucosal restitution occurs by epithelial cell migration, rather than by proliferation, to reseal superficial wounds after injury. Polyamines are essential for the stimulation of intestinal epithelial cell (IEC) migration during restitution in association with their ability to regulate Ca2+ homeostasis, but the exact mechanism by which polyamines induce cytosolic free Ca2+ concentration ([Ca2+]cyt) remains unclear. Phospholipase C (PLC)-γ1 catalyzes the formation of inositol ( 1 , 4 , 5 )-trisphosphate (IP3), which is implicated in the regulation of [Ca2+]cyt by modulating Ca2+ store mobilization and Ca2+ influx. The present study tested the hypothesis that polyamines are involved in PLC-γ1 activity, regulating [Ca2+]cyt and cell migration after wounding. Depletion of cellular polyamines by α-difluoromethylornithine inhibited PLC-γ1 expression in differentiated IECs (stable Cdx2-transfected IEC-6 cells), as indicated by substantial decreases in levels of PLC-γ1 mRNA and protein and its enzyme product IP3. Polyamine-deficient cells also displayed decreased [Ca2+]cyt and inhibited cell migration. Decreased levels of PLC-γ1 by treatment with U-73122 or transfection with short interfering RNA specifically targeting PLC-γ1 also decreased IP3, reduced resting [Ca2+]cyt and Ca2+ influx after store depletion, and suppressed cell migration in control cells. In contrast, stimulation of PLC-γ1 by 2,4,6-trimethyl- N-( meta-3-trifluoromethylphenyl)-benzenesulfonamide induced IP3, increased [Ca2+]cyt, and promoted cell migration in polyamine-deficient cells. These results indicate that polyamines are absolutely required for PLC-γ1 expression in IECs and that polyamine-mediated PLC-γ1 signaling stimulates cell migration during restitution as a result of increased [Ca2+]cyt.


2018 ◽  
Vol 33 (suppl_1) ◽  
pp. i107-i107
Author(s):  
Sonja Djudjaj ◽  
Simon Müller ◽  
Janina Lange ◽  
Margarete Goppelt-Struebe ◽  
Peter Boor

2006 ◽  
Vol 281 (42) ◽  
pp. 31972-31986 ◽  
Author(s):  
Alok Tomar ◽  
Sudeep George ◽  
Pallavi Kansal ◽  
Yaohong Wang ◽  
Seema Khurana

PLoS ONE ◽  
2013 ◽  
Vol 8 (2) ◽  
pp. e56731 ◽  
Author(s):  
Ching-Yuang Lin ◽  
Tze-Yi Lin ◽  
Min-Chun Lee ◽  
Shih-Chieh Chen ◽  
Jeng-Shou Chang

Sign in / Sign up

Export Citation Format

Share Document