The ERM proteins ezrin, radixin and moesin are adaptor proteins that link plasma membrane receptors to the actin cytoskeleton. Ezrin and moesin have been implicated in cell polarization and cell migration, but little is known about the involvement of radixin in these processes. Here we show that radixin is required for migration of PC3 prostate cancer cells, and that radixin, but not ezrin or moesin, depletion by RNA interference increases cell spread area and cell–cell adhesion mediated by adherens junctions. Radixin depletion also alters actin organization, and distribution of active phosphorylated ezrin and moesin. Similar effects were observed in MDA-MB-231 breast cancer cells. The phenotype of radixin-depleted cells is similar to that induced by constitutively active Rac1, and Rac1 is required for the radixin knockdown phenotype. Radixin depletion also increases the activity of Rac1 but not Cdc42 or RhoA. Analysis of Rac guanine nucleotide exchange factors (GEFs) suggests that radixin affects the activity of Vav GEFs. Indeed, Vav GEF depletion reverses the phenotype of radixin knockdown and reduces the effect of radixin knockdown on Rac1 activity. Our results indicate that radixin plays an important role in promoting cell migration by regulating Rac1-mediated epithelial polarity and formation of adherens junctions through Vav GEFs.

The ezrin/radixin/moesin (ERM) family of proteins act as reversible linkers between cell surface receptors and the actin cytoskeleton, and participate in signal transduction pathways regulating cell migration and cell polarity (Fehon et al., 2010). They contribute to the formation of dynamic actin filament-containing membrane structures such as filopodia, uropods, lamellipodia and membrane ruffles, the cleavage furrow in dividing cells, and cell–cell adhesions (Bretscher et al., 2002; Gautreau et al., 2002; Niggli and Rossy, 2008).

Ezrin, radixin and moesin are closely related ∼80-kDa proteins that consist of three domains: an N-terminal FERM (four point one-ezrin–radixin–moesin) domain linked to a C-terminal F-actin-binding domain through an α-helical domain (Fehon et al., 2010; Neisch and Fehon, 2011). The FERM domain interacts either directly with membrane receptors such as CD44, CD43, and intercellular adhesion proteins (ICAMs) or indirectly through scaffold proteins such as EBP50 (ezrin binding protein 50). It also binds to phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2] (Bretscher et al., 2002; Gautreau et al., 2000; Gautreau et al., 2002; Matsui et al., 1998; Nakamura et al., 1995; Oshiro et al., 1998). ERM proteins can exist in an inactive ‘closed’ conformation, where the C-terminal domain binds to the N-terminal FERM domain. In this conformation they are found in the cytosolic fraction of cells and the binding sites for plasma membrane proteins and F-actin are masked. ERM proteins are activated through conformational changes induced by binding to PtdIns(4,5)P2 and subsequent phosphorylation of a conserved C-terminal threonine residue [T567 in ezrin, T564 in radixin and T558 in moesin] (Brown et al., 2003; Gautreau et al., 2000; Nakamura et al., 1995; Roch et al., 2010; Suda et al., 2011). Several kinases, including members of the protein kinase C, ROCK and MRCK families have been reported to regulate ERM function through phosphorylation of this threonine (Baeyens et al., 2010; Belkina et al., 2009; Hebert et al., 2008; Matsui et al., 1998; Nakamura et al., 2000; Ng et al., 2001; Parisiadou et al., 2009; Yamane et al., 2011).

ERM proteins can act both upstream and downstream of Rho GTPases, which are key regulators of cell polarity and migration (Hall, 2009; Hughes and Fehon, 2007). Rho GTPases, cycle between an active GTP-bound and an inactive GDP-bound conformation. They are activated by guanine nucleotide exchange factors (GEFs) and inactivated by GTPase activating proteins (GAPs). Some Rho GTPases are also regulated by binding to GDP dissociation inhibitors (GDIs) or by phosphorylation (Hirao et al., 1996; Ivetic and Ridley, 2004; Mackay et al., 1997). RhoGDI can associate with ERM proteins, thereby reducing RhoGDI interaction with GTPases and consequently promoting their activation (Dovas and Couchman, 2005). Ezrin has been reported to interact with PLEKHG6 (pleckstrin homology domain containing family G with RhoGEF domain member 6), a GEF for RhoG and Rac1, to induce morphological changes in the apical region of epithelial cells (D’Angelo et al., 2007). In vitro studies have also reported a direct interaction between the RhoGEF Dbl and radixin (Prag et al., 2007; Takahashi et al., 1998). In this report it is suggested that this interaction would compete with RhoGDI (Takahashi et al., 1998). On the other hand, RhoA and Rac stimulate ERM activity by increasing the levels of PtdIns(4,5)P2 and/or by stimulating C-terminal phosphorylation, leading to induction of microvilli enriched in activated ERM proteins (Auvinen et al., 2007; Shaw et al., 1997; Yonemura et al., 2002). ERM proteins also bind to the cytoplasmic domain of the transmembrane receptor CD44 and this interaction is enhanced by RhoA activity (del Pozo et al., 1999; Lee et al., 2004; Tsuda et al., 2004). Finally, Rac1 activation in T-lymphocytes leads to inactivation of ERM proteins via dephosphorylation of the C-terminal threonine (Cernuda-Morollon et al., 2010).

The expression of ERM proteins has been investigated in several cancer types, including prostate cancer. For example, ezrin expression has been correlated with adverse prognosis, showing high levels of expression in both high grade prostate intraepithelial neoplasia and prostate adenocarcinoma (Pang et al., 2004; Valdman et al., 2005). Androgen-induced cell invasion requires hormonal regulation of ezrin phosphorylation in the prostate cancer cell line, LNCaP (Chuan et al., 2006). Recently, a study analysing radixin and moesin expression in normal prostate, high grade prostate intraepithelial neoplasia and prostatic adenocarcinoma tissues showed a slight decrease in radixin levels in adenocarcinomas (Bartholow et al., 2011). On the other hand, radixin has been implicated in promoting breast cancer metastasis in an experimental metastasis model (Valastyan et al., 2009).

Although the functions of ezrin and moesin have been extensively studied in cell migration, a role for radixin has not been described. Here we show that radixin depletion severely impairs migration of prostate cancer PC3 cells. This inhibition is accompanied by a change in cell shape and increased spread area concomitant with reorganization of the actin cytoskeleton, focal contacts and cell–cell contacts. We demonstrate that Rac1, which is activated by radixin depletion, mediates these changes. Our results indicate that radixin levels regulate the transition from a non-migratory epithelial morphology with cell–cell adhesions to a migratory morphology by altering Rac1 activity.

Radixin regulates cell migration

To elucidate the role of radixin in cell migration, we depleted ERM protein expression in prostate carcinoma-derived PC3 cells by transient transfection with siRNA (sequences in supplementary material Table S1). Radixin, ezrin and moesin were each efficiently knocked down by 4 different siRNAs (Fig. 1A) Radixin depletion did not affect the levels of ezrin or moesin (Fig. 1B).

Fig. 1.

Radixin depletion inhibits migration and increases cell area. (A) PC3 cells were transfected with four different siRNAs targeting radixin, ezrin, moesin or control siRNA, and lysed after 48 hours. Cell lysates were processed for further analysis by western blot using specific antibodies against radixin. Control: non-transfected cells. Blots were re-probed with antibodies to MLC as a loading control. (B) Western blot analysis of ezrin, radixin and moesin expression in radixin-depleted cells. PC3 cells were transfected with siRNA to radixin, lysed after 48, 72 and 96 hours and processed for western blot analysis using antibodies against the indicated proteins. (C) Cells were transfected with radixin siRNA or control siRNA. After 48 hours cells were trypsinised, replated and analysed by phase-contrast time-lapse microscopy. Images are shown from movies of control siRNA-transfected cells (top) or radixin siRNA-2-transfected cells (bottom). Similar results were obtained with radixin siRNA-3 (not shown). The time after the start of the time-lapse movies (h) is indicated. Scale bar: 50 µm. (D) Quantification of migration speed of PC3 cells transfected with four different siRNAs to radixin (1–4) or control siRNA. (E) High magnification and high contrast images of cells from supplementary material Movies 1 and 2. Membrane ruffles are indicated by arrows in control cells and arrowheads in radixin knockdown cells. Scale bar: 10 µm. (F) Quantification of cell spread area, determined by staining for F-actin, of cells transfected with radixin siRNA-2, ezrin siRNA-2, moesin siRNA-2 or control siRNA. The spread area is expressed relative to that of control cells. Scale bar: 10 µm. Values are means ± s.e.m. from at least three independent experiments. Analysis of variance (ANOVA) was performed to assess the significance of the results shown in each graph. Subsequently a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knocked down populations (***P<0.001).

Fig. 1.

Radixin depletion inhibits migration and increases cell area. (A) PC3 cells were transfected with four different siRNAs targeting radixin, ezrin, moesin or control siRNA, and lysed after 48 hours. Cell lysates were processed for further analysis by western blot using specific antibodies against radixin. Control: non-transfected cells. Blots were re-probed with antibodies to MLC as a loading control. (B) Western blot analysis of ezrin, radixin and moesin expression in radixin-depleted cells. PC3 cells were transfected with siRNA to radixin, lysed after 48, 72 and 96 hours and processed for western blot analysis using antibodies against the indicated proteins. (C) Cells were transfected with radixin siRNA or control siRNA. After 48 hours cells were trypsinised, replated and analysed by phase-contrast time-lapse microscopy. Images are shown from movies of control siRNA-transfected cells (top) or radixin siRNA-2-transfected cells (bottom). Similar results were obtained with radixin siRNA-3 (not shown). The time after the start of the time-lapse movies (h) is indicated. Scale bar: 50 µm. (D) Quantification of migration speed of PC3 cells transfected with four different siRNAs to radixin (1–4) or control siRNA. (E) High magnification and high contrast images of cells from supplementary material Movies 1 and 2. Membrane ruffles are indicated by arrows in control cells and arrowheads in radixin knockdown cells. Scale bar: 10 µm. (F) Quantification of cell spread area, determined by staining for F-actin, of cells transfected with radixin siRNA-2, ezrin siRNA-2, moesin siRNA-2 or control siRNA. The spread area is expressed relative to that of control cells. Scale bar: 10 µm. Values are means ± s.e.m. from at least three independent experiments. Analysis of variance (ANOVA) was performed to assess the significance of the results shown in each graph. Subsequently a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knocked down populations (***P<0.001).

Cell migration was analysed by tracking cells in movies generated by time-lapse microscopy (Fig. 1C; supplementary material Movies 1 and 2). PC3 cells normally grow and migrate as single cells with a mesenchymal morphology, and migrate with a mean speed of 1.34±0.04 µm/minute. In contrast, radixin-depleted PC3 cells had a migration speed of approximately 38% of that of control siRNA-transfected cells, observed with 4 independent siRNAs targeting radixin (Fig. 1D; supplementary material Movies 1 and 2). The inhibition of migration following radixin suppression appeared to be independent of the substrate as similar results were obtained on uncoated plastic and on collagen-I or fibronectin-coated surfaces (data not shown).

Radixin-depleted cells were flatter with a more circular shape than control cells; they also showed persistent membrane ruffling around their whole periphery, rather than the small and localised membrane ruffles in discrete regions observed in control cells (Fig. 1E; supplementary material Movies 3 and 4). In addition, we observed a fourfold increase in the cell spread area of radixin-depleted cells compared to control cells, either quantified from fixed cells stained for F-actin (Fig. 1F) or from analysis of cells in time-lapse movies (data not shown). In contrast, depletion of moesin or ezrin did not affect cell area (Fig. 1F). Although most of the control cells had a migratory phenotype with a defined leading edge, we observed some cells in the whole population with a similar flat shape to radixin-depleted cells. We therefore quantified the number of cells that had a circular, flat shape in both control and radixin knockdown cells. We observed that almost 90% of the radixin knockdown cells were circular and flat but just a 13% of control cells showed a similar phenotype (Table 1).

Table 1.

Quantification of cell–cell contacts in radixin-depleted cells

 Elongated cells Flat cells 
siRNA Proportion Cell–cell contacts Proportion Cell–cell contacts 
Control 87% 5% 13% 3.5% 
Radixin 11% 1.8% 89% 79% 
 Elongated cells Flat cells 
siRNA Proportion Cell–cell contacts Proportion Cell–cell contacts 
Control 87% 5% 13% 3.5% 
Radixin 11% 1.8% 89% 79% 

PC3 cells were transfected with siRNA-2 to radixin or control siRNA. After 72 hours, cells were fixed and stained for F-actin (to reveal their shape) and for N-cadherin (to reveal cell–cell contacts). Cells were quantified for their morphology: they were defined as elongated when they had a polarized morphology with a front and a back, or as flat when it was not possible to define a front and a back and the cell had a circular morphology; and for cell–cell contacts, defined as positive staining for N-cadherin between two cells.

Radixin regulates actin filament and focal contact distribution

Since actin cytoskeleton and cell adhesion dynamics are required for cell migration, we investigated whether radixin knockdown was accompanied by changes to the actin cytoskeleton and cell adhesions. We observed that radixin-depleted PC3 cells had more peripheral actin filament bundles than control cells, mainly localised close to the plasma membrane. This actin reorganization was accompanied by a re-localisation of the focal contact marker paxillin (Schaller, 2001; Schaller, 2004) from a uniform distribution around the cell periphery to larger focal contacts that mainly localised at the tips of actin filament bundles (Fig. 2A). Active, C-terminally phosphorylated ERM proteins (p-ERMs) often localise to membrane ruffles and lamellipodia (Niggli and Rossy, 2008). In control PC3 cells, p-ERMs were primarily localised to filopodia and other cell protrusions. In contrast, p-ERMs had a more punctate distribution on the dorsal surface in radixin-depleted cells, similar to the dorsal distribution of F-actin (Fig. 2B), a characteristic of microvilli of epithelial cells (Fehon et al., 2010; Lan et al., 2006). This suggests that radixin-depleted PC3 cells have a more epithelial phenotype. Quantification indicated that more than 80% of the radixin-depleted cells were positive for punctate F-actin and p-ERM in the top 20% of the cell height whereas only 12% of the control cells had similar F-actin/p-ERM distribution at that height (Fig. 2C). Radixin therefore clearly regulates p-ERM distribution, although it did not alter total levels of p-ERM, as determined by western blotting (supplementary material Fig. S1).

Fig. 2.

Radixin affects p-ERM, actin cytoskeleton and cell adhesion distribution. (A) Redistribution of actin filaments and phosphorylated paxillin (p-Paxillin) in radixin-depleted cells through transfection of siRNA-2 to radixin (similar results were obtained with radixin siRNA-3; not shown). PC3 cells were fixed and stained for F-actin and with antibodies to phosphorylated paxillin. Phosphorylated paxillin localises at the tips of F-actin bundles in radixin-depleted cells (magnification of boxed regions in far right panels). Scale bar: 20 µm. (B) Distribution of F-actin and phosphorylated ERM proteins (p-ERM) in radixin-depleted cells through transfection of radixin siRNA-2 (similar results were obtained with radixin siRNA-3, not shown). Cells were stained for actin filaments and with antibodies to p-ERM. Far right panels show magnifications of the boxed regions. Scale bar: 20 µm. (C) Quantification of F-actin and p-ERM staining in the top of cells grown in high-density cultures. Cells were maintained at confluence for 48 hours. Images were obtained by confocal microscopy (z-stacks) and quantification was performed analysing positive staining in those images for actin and p-ERM in the top 20% of the cell height. Values are means ± s.e.m. from at least three independent experiments. ANOVA was performed to assess the significance of the results. Subsequently, a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knockdown populations (***P<0.001).

Fig. 2.

Radixin affects p-ERM, actin cytoskeleton and cell adhesion distribution. (A) Redistribution of actin filaments and phosphorylated paxillin (p-Paxillin) in radixin-depleted cells through transfection of siRNA-2 to radixin (similar results were obtained with radixin siRNA-3; not shown). PC3 cells were fixed and stained for F-actin and with antibodies to phosphorylated paxillin. Phosphorylated paxillin localises at the tips of F-actin bundles in radixin-depleted cells (magnification of boxed regions in far right panels). Scale bar: 20 µm. (B) Distribution of F-actin and phosphorylated ERM proteins (p-ERM) in radixin-depleted cells through transfection of radixin siRNA-2 (similar results were obtained with radixin siRNA-3, not shown). Cells were stained for actin filaments and with antibodies to p-ERM. Far right panels show magnifications of the boxed regions. Scale bar: 20 µm. (C) Quantification of F-actin and p-ERM staining in the top of cells grown in high-density cultures. Cells were maintained at confluence for 48 hours. Images were obtained by confocal microscopy (z-stacks) and quantification was performed analysing positive staining in those images for actin and p-ERM in the top 20% of the cell height. Values are means ± s.e.m. from at least three independent experiments. ANOVA was performed to assess the significance of the results. Subsequently, a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knockdown populations (***P<0.001).

Radixin depletion similarly induced cell spreading, increased peripheral actin filament bundles and accumulation of p-ERM on the dorsal surface in MDA-MB-231 breast cancer cells, indicating that the phenotype is not PC3-cell specific (supplementary material Fig. S2).

Radixin depletion increases cell–cell contacts

From time-lapse movies, it was apparent that radixin-depleted cells formed more stable cell–cell contacts than control cells (Fig. 3A; supplementary material Movies 5 and 6). This was quantified by determining the proportion of cells that maintained contact with each other during the duration of each movie: over 90% of radixin-depleted cells maintained contact with their neighbour(s) over 16 h (Fig. 3B). In order to investigate in more detail the basis for this radixin-regulated change in cell–cell adhesion we analysed the distribution of adherens junctions, which are known to be important for establishing and maintaining cell–cell contacts (Millan et al., 2010; Shapiro and Weis, 2009). PC3 cells express N-cadherin but not E-cadherin (supplementary material Fig. S3A), and radixin knockdown did not induce E-cadherin expression (supplementary material Fig. S3B). In control PC3 cells N-cadherin was predominantly distributed throughout the cytoplasm with some localisation at the plasma membrane. After radixin knockdown, N-cadherin accumulated at cell–cell contact regions (Fig. 3C). Similarly, β-catenin, which binds directly to cadherin cytoplasmic tails (Shapiro and Weis, 2009; Sung et al., 2008), was localised to sites of cell–cell contact in radixin-depleted cells, whereas in control cells it was homogenously distributed throughout the cytoplasm (Fig. 3C).

Fig. 3.

Radixin depletion increases adherens junctions. (A) PC3 cells cultured at low-density were transfected with siRNA-2 to radixin or control siRNA. The images are from movies of control siRNA-transfected cells (top) and radixin siRNA-transfected cells (bottom). White arrows indicate an example of a cell group that remains as such through the whole movie. Similar results were obtained with siRNA-3 to radixin (not shown). The time after the start of the time-lapse movies (h) is indicated. Scale bars: 50 µm. (B) Quantification of cells keeping and/or establishing cell–cell contacts, determined from movies such as those shown in supplementary material Movies 5 and 6, of cells that were establishing and keeping a colony-like structure (more than two cells together through the whole movie). In both control and radixin knockdown cells the number of cells that remained as a colony through the whole movie (black portion of the column) and the ones that scattered (grey portion of the column) was quantified and the percentage of each type was calculated. (C) PC3 cells were transfected with radixin siRNA-2 or control siRNA and analysed after 72 hours. Cells were fixed and stained for N-cadherin and β-catenin. β-catenin and N-cadherin colocalise in cell–cell contact regions in radixin-depleted cells (right panels and magnification of boxed region in far right panels). Scale bar: 100 µm. (D) Quantification of cell height in high-density cultures. Cells were maintained at confluence for 48 hours and examined by confocal microscopy to determine the cell height. Five different fields containing at least 30 cells from each of three independent experiments were quantified using the calibrated ruler of the 3-D tool from the ZEN software (Zeiss). ANOVA was performed to assess the significance of the results. Subsequently, a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knockdown populations (***P<0.001).

Fig. 3.

Radixin depletion increases adherens junctions. (A) PC3 cells cultured at low-density were transfected with siRNA-2 to radixin or control siRNA. The images are from movies of control siRNA-transfected cells (top) and radixin siRNA-transfected cells (bottom). White arrows indicate an example of a cell group that remains as such through the whole movie. Similar results were obtained with siRNA-3 to radixin (not shown). The time after the start of the time-lapse movies (h) is indicated. Scale bars: 50 µm. (B) Quantification of cells keeping and/or establishing cell–cell contacts, determined from movies such as those shown in supplementary material Movies 5 and 6, of cells that were establishing and keeping a colony-like structure (more than two cells together through the whole movie). In both control and radixin knockdown cells the number of cells that remained as a colony through the whole movie (black portion of the column) and the ones that scattered (grey portion of the column) was quantified and the percentage of each type was calculated. (C) PC3 cells were transfected with radixin siRNA-2 or control siRNA and analysed after 72 hours. Cells were fixed and stained for N-cadherin and β-catenin. β-catenin and N-cadherin colocalise in cell–cell contact regions in radixin-depleted cells (right panels and magnification of boxed region in far right panels). Scale bar: 100 µm. (D) Quantification of cell height in high-density cultures. Cells were maintained at confluence for 48 hours and examined by confocal microscopy to determine the cell height. Five different fields containing at least 30 cells from each of three independent experiments were quantified using the calibrated ruler of the 3-D tool from the ZEN software (Zeiss). ANOVA was performed to assess the significance of the results. Subsequently, a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knockdown populations (***P<0.001).

Although cell spreading in radixin-depleted cells would be expected to increase cell–cell contact, very few control PC3 cells that interacted with each other showed N-cadherin and β-catenin accumulation or colocalisation in the regions of contact. Quantification showed that 79% of radixin-depleted cells had cell–cell contacts that were positive for N-cadherin, whereas only 3.5% of the control cells showing a similar flat phenotype had N-cadherin in the cell–cell contact regions (Table 1). The interaction of control cells did not seem to depend on whether they had an elongated or spread morphology, whereas for radixin knockdown cells there was a significant correlation between the presence of N-cadherin in cell–cell contacts and a spread phenotype. This indicates that the spread phenotype observed in some control cells is not likely to be related to that induced by radixin knockdown.

Interestingly, when cells were grown to very high confluence, the radixin-depleted cells showed a significant increase in cell height (Fig. 3D), indicating that cell–cell contacts are likely to be more mature. In these high-density cultures, the cadherin-associated protein p120-catenin (Carnahan et al., 2010) localised to cell–cell contacts of radixin-depleted cells but not those of control cells (supplementary material Movies 7 and 8).

Effects of radixin depletion are mediated by Rac1

Rho GTPases are known to be important both for cell migration and for formation of adherens junctions in epithelial cells (Parri and Chiarugi, 2010; Popoff and Geny, 2009; van Duijn et al., 2010). Since radixin knockdown inhibited migration and enhanced adherens junction formation, we investigated whether radixin affected Rho GTPase activity. Radixin depletion led to an increase in Rac1 activity, but no change in Cdc42 or RhoA activity was observed (Fig. 4A). Total levels of Rac1, Cdc42 and RhoA were not changed. Consistent with a role for Rac1 in mediating the phenotypic changes induced by radixin suppression, expression of constitutively active Rac1 (GFP–Rac1–L61) but not dominant negative Rac1 (GFP–Rac1–N17) stimulated cell spreading and increased N-cadherin levels at sites of cell–cell contact (Fig. 4B; supplementary material Fig. S4). GFP–Rac1–L61 colocalised with N-cadherin at cell–cell contacts (Fig. 4B).

Fig. 4.

Morphological effects of radixin depletion are Rac1-dependent. (A) PC3 cells were transfected with siRNAs to radixin or control siRNA. Cells were lysed after 72 hours, and Rac1, Cdc42 and RhoA activity were determined by pull-down assays. A representative western blot is shown of active and total Rac1, Cdc42, and RhoA (left), and quantification of relative activity from three independent experiments (right graph). Values are means ± s.e.m. Two-way ANOVA was performed to assess the significance of the results. Subsequently, a Bonferroni’s multiple comparison test was used to determine whether the differences were significant between control and knocked down populations for each one of the Rho GTPases (***P<0.001). (B) PC3 cells were transfected with the constitutively active form (GFP–Rac1–L61) or the dominant-negative form (GFP–Rac1–N17) of Rac1, and 48 hours later were fixed and stained for actin filaments and with antibodies to N-cadherin (magnifications of boxed regions are shown in far right panels). (C) PC3 cells were sequentially transfected with siRNAs to Rac1 and radixin or vice versa. Cells were then fixed and stained for actin filaments. The left panel shows a representative image of control cells; the middle panels show images of the first round of transfection with siRNA to either Rac1 (top) or radixin (bottom); the right panels show images of the cells after the second round of transfection with siRNA to either radixin (top) or Rac1 (bottom). Scale bars: 100 µm.

Fig. 4.

Morphological effects of radixin depletion are Rac1-dependent. (A) PC3 cells were transfected with siRNAs to radixin or control siRNA. Cells were lysed after 72 hours, and Rac1, Cdc42 and RhoA activity were determined by pull-down assays. A representative western blot is shown of active and total Rac1, Cdc42, and RhoA (left), and quantification of relative activity from three independent experiments (right graph). Values are means ± s.e.m. Two-way ANOVA was performed to assess the significance of the results. Subsequently, a Bonferroni’s multiple comparison test was used to determine whether the differences were significant between control and knocked down populations for each one of the Rho GTPases (***P<0.001). (B) PC3 cells were transfected with the constitutively active form (GFP–Rac1–L61) or the dominant-negative form (GFP–Rac1–N17) of Rac1, and 48 hours later were fixed and stained for actin filaments and with antibodies to N-cadherin (magnifications of boxed regions are shown in far right panels). (C) PC3 cells were sequentially transfected with siRNAs to Rac1 and radixin or vice versa. Cells were then fixed and stained for actin filaments. The left panel shows a representative image of control cells; the middle panels show images of the first round of transfection with siRNA to either Rac1 (top) or radixin (bottom); the right panels show images of the cells after the second round of transfection with siRNA to either radixin (top) or Rac1 (bottom). Scale bars: 100 µm.

In order to determine whether Rac1 was required for the phenotype of radixin-depleted cells, cells were sequentially transfected with siRNAs to Rac1 then radixin, or radixin then Rac1 (Fig. 4C). Rac1 suppression both prevented the increase in cell spread area induced by radixin depletion and reverted the radixin knockdown phenotype to that of control cells. The changes induced by radixin depletion are therefore dependent on Rac1.

Radixin depletion acts through Vav GEF activation to regulate Rac1 and cell spreading

To determine how radixin affected Rac1 activity, we first tested whether there was an interaction between radixin and regulators of Rac1, including Dbl and RhoGDI, which have previously been reported to interact with radixin in vitro (Takahashi et al., 1998), and Tiam1, which is known to promote adherens junction formation via Rac1 (Buongiorno et al., 2008). We could not detect interaction between endogenous proteins in any case by immunoprecipitation (data not shown). The GEF activity of Vav GEFs is known to be activated by phosphorylation of a conserved tyrosine (Y174) in their acidic regions, adjacent to the PH-DH GEF domains (Lopez-Lago et al., 2000). Interestingly, we observed an increase in Y174 tyrosine phosphorylation of Vav GEFs only in radixin-depleted cells but not ezrin- or moesin-depleted cells (Fig. 5A). In order to determine whether Vav GEFs were required for the phenotype of radixin-depleted cells, cells were transfected with siRNAs to radixin or Vav GEFs alone or together (Fig. 5B). Vav depletion prevented the increase in cell-spread area induced by radixin depletion, although Vav depletion alone did not alter cell morphology.

Fig. 5.

Radixin regulates Vav tyrosine phosphorylation. (A) PC3 cells were transfected with the indicated siRNAs to radixin, ezrin or moesin or control siRNA. After 72 hours, cells were lysed and cell lysates were analysed by western blotting for radixin, ezrin, moesin, phospho-Tyr174 Vav and total Vav. Values are means ± s.e.m. from at least three independent experiments. ANOVA was performed to assess the significance of the results shown in each graph. Subsequently a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knockdown populations (***P<0.001). (B) PC3 cells were transfected with siRNAs to radixin, Vav GEFs (Vav1, Vav2 and Vav3), both radixin and Vav GEFs, or control siRNA. After 72 hours, cells were fixed and stained for actin filaments. Representative images are shown. Scale bar: 10 µm.

Fig. 5.

Radixin regulates Vav tyrosine phosphorylation. (A) PC3 cells were transfected with the indicated siRNAs to radixin, ezrin or moesin or control siRNA. After 72 hours, cells were lysed and cell lysates were analysed by western blotting for radixin, ezrin, moesin, phospho-Tyr174 Vav and total Vav. Values are means ± s.e.m. from at least three independent experiments. ANOVA was performed to assess the significance of the results shown in each graph. Subsequently a Dunnet’s multiple comparison test was used to determine whether the differences were significant between control and knockdown populations (***P<0.001). (B) PC3 cells were transfected with siRNAs to radixin, Vav GEFs (Vav1, Vav2 and Vav3), both radixin and Vav GEFs, or control siRNA. After 72 hours, cells were fixed and stained for actin filaments. Representative images are shown. Scale bar: 10 µm.

The increase in Vav phosphorylation upon radixin depletion suggests that Vav GEFs could be responsible for the increase in Rac1 activity. Depletion of Vav GEFs significantly reduced the increase in Rac1 activity induced by radixin depletion (Fig. 6). No change in Rac1 activity was observed upon Vav GEF depletion in the absence of radixin knockdown (Fig. 6). Together, these results indicate that the Vav GEF activation following radixin depletion is required for Rac1-mediated cell spreading.

Fig. 6.

Vav GEFs mediate the radixin-dependent increase of Rac1 activity. PC3 cells were transfected as indicated with siRNAs to radixin, siRNAs to Vav GEFs (Vav1, Vav2 and Vav3) or control siRNAs. Cells were lysed after 72 hours, and Rac1 activity was determined by pull-down assays. A representative western blot is shown of active and total Rac1 and quantification of relative activity from three independent experiments (graph). Values are means ± s.e.m. ANOVA was performed to assess the significance of the results. Subsequently, a Bonferroni’s multiple comparison test was used to determine whether the differences were significant between each condition (*P<0.05; ***P<0.001).

Fig. 6.

Vav GEFs mediate the radixin-dependent increase of Rac1 activity. PC3 cells were transfected as indicated with siRNAs to radixin, siRNAs to Vav GEFs (Vav1, Vav2 and Vav3) or control siRNAs. Cells were lysed after 72 hours, and Rac1 activity was determined by pull-down assays. A representative western blot is shown of active and total Rac1 and quantification of relative activity from three independent experiments (graph). Values are means ± s.e.m. ANOVA was performed to assess the significance of the results. Subsequently, a Bonferroni’s multiple comparison test was used to determine whether the differences were significant between each condition (*P<0.05; ***P<0.001).

Compared to ezrin and moesin, little is known of radixin function. Here we show for the first time that radixin regulates cell shape, migration and cell–cell adhesion, and that it acts by altering Rac1 activity. The inhibition of migration in radixin-depleted cells is accompanied by an increase in cell area and cadherin-mediated contacts. Our results indicate that the different effects of ezrin, moesin or radixin depletion are not simply explained by tissue-selective expression of the 3 isoforms. Only radixin depletion but not ezrin or moesin depletion has a profound effect on cell morphology in PC3 and MDA-MB-231 cells, even though ezrin and moesin are both expressed.

Radixin depletion induces changes in PC3 cell shape and adhesion similar to acquisition of a more epithelial morphology, despite the fact that they do not re-express E-cadherin. However, mesenchymal cells (including fibroblasts and astrocytes) frequently form N-cadherin-mediated adherens junctions (Li et al., 2001; Zhu et al., 2010), and thus the junctions formed in radixin-depleted PC3 cells are likely to resemble these. The changes induced by radixin depletion are dependent on Rac1 since Rac1 knockdown inhibits the radixin knockdown-induced increase of cell area, and expression of constitutively active Rac1 induces cell spreading and N-cadherin accumulation at cell–cell contacts. The role of Rac1 in inducing and maintaining adherens junctions is well established (Hage et al., 2009; Ray et al., 2007), and thus increased Rac1 activity could explain the high level of adherens junctions in radixin-depleted cells. Several RacGEFs have been implicated in adherens junction assembly. For example, RacGEF Tiam1 stimulates adherens junction assembly in MDCK epithelial cells (Malliri et al., 2004). Vav GEFs have also been implicated in adherens junction signalling to Rac1 (Fukuyama et al., 2006). Both Tiam1 and Vav have been implicated downstream of phosphoinositide 3-kinases in regulating adherens junctions (Rivard, 2009). The increased Rac1 activity in radixin-depleted PC3 cells might be due to Vav GEFs rather than Tiam1, since tyrosine phosphorylation of Vav GEFs was increased following radixin knockdown and they were required for the increase in cell size and Rac1 activity observed upon radixin depletion.

Both ezrin and moesin are implicated in regulating cell–cell adhesion, but radixin has not previously been linked to this process. For example, in the mouse intestinal epithelium, ezrin deficiency results in fusion of villi, accompanied by the appearance of elongated cell–cell junctions (Saotome et al., 2004). In Caenorhabditis elegans, the single ERM protein erm-1 is required for lumen formation and positioning of adherens junctions (Gobel et al., 2004). In zebrafish, moesin and the endothelium-specific VE-cadherin both regulate endothelial lumen formation during development of blood vessels (Zhu et al., 2010).

Radixin localises to bile canicular membranes in mouse liver, and radixin-deficient mice accumulate high levels of bilirubin in their blood, possibly because they are unable to secrete it into the bile (Kikuchi et al., 2002). However, this phenotype is dependent on the genetic background of the mice (Fukumoto et al., 2007), and the mice grow normally and are fertile. The underlying defect is therefore unclear, although it could relate to a decrease in microvilli in the apical bile canalicular membranes (Kikuchi et al., 2002). Radixin-null mice also become deaf due to progressive degeneration of cochlear stereocilia, which are actin filament-based apical projections on hair cells (Kitajiri et al., 2004). Cadherin-23 mutations lead to deafness in humans (Muller, 2008), and thus it would be interesting to know whether radixin affects cadherin-23 localisation in the cochlea, given that we observe that radixin regulates N-cadherin localisation in PC3 cells.

Increased ezrin expression is a well-known prognostic marker for tumour aggressiveness in a variety of cancers, and influences tumour survival and progression (Hunter, 2004). Moesin expression is also upregulated in some cancers, correlating with progression (Cui et al., 2009). So far little is known about radixin in cancer, although interestingly it is a target of the microRNA miR-31, which inhibits breast cancer metastasis in animal models (Valastyan et al., 2009). Depletion of the miR-31 targets radixin, RhoA and integrin α5 together phenocopies the effects of miR-31 expression in suppressing metastasis (Valastyan et al., 2010). The relative contribution of each of these three genes is not known, however. Our data are consistent with a role for radixin in promoting cancer cell migration, which could contribute to cancer progression.

Cell lines and reagents

PC3 and DU145 prostate cancer cells were grown in RPMI containing 25 mM Hepes and 2 mM glutamine supplemented with 10% FCS, 100 µg/ml streptomycin, and 100 units/ml penicillin (5% CO2, 37°C). MDA-MB-231 breast cancer cells were grown in DMEM containing 25 mM Hepes and 2 mM glutamine supplemented with 10% FCS, 100 µg/ml streptomycin, and 100 units/ml penicillin (5% CO2, 37°C). Antibodies were obtained from the following sources: radixin from Sigma-Aldrich (cat. no. R3653); ezrin, moesin, phospho-ERM, MLC and phospho-paxillin from Cell Signalling (cat. no. 3145, 3142, 2466, 3672 and 2541, respectively); N-cadherin, E-cadherin, p120-catenin, β-catenin from BD Transduction Laboratories (cat. no. 610921, 610404, 610134 and 610154, respectively); Rac1 clone 23A8 from Millipore (cat. no. 05-389); Cdc42 from Millipore (cat. no. 07-1466); RhoA from Cytoskeleton (cat. no. ARH03-A); Vav and phospho-Vav from Santa Cruz (cat. no. sc-132 and sc-16408-R, respectively); GAPDH from Sigma-Aldrich (cat. no. G-8795); actin filaments were visualized with Alexa-Fluor-546- or Alexa-Fluor-633-labelled phalloidin from Invitrogen (cat. no. A22283 and A22284, respectively). Alexa Fluor 488 anti-rabbit, Alexa Fluor 546 anti-mouse, and Alexa Fluor 633 anti-mouse secondary antibodies were from Invitrogen (cat. no. A11008, A11103 and A21126, respectively). HRP-anti-rabbit and HRP-anti-mouse from GE-Healthcare (cat. no. NA934 and NA931, respectively). Except when specifically stated, all the chemical reagents were obtained from Sigma-Aldrich.

siRNA and DNA transfections

All siRNAs were from Dharmacon (Thermo Scientific); esiRNA (a mixture of siRNAs that all target the same mRNA sequence) for vav1, vav2 and vav3 were from Sigma-Aldrich; their sequences are specified in supplementary material Table S1. Cells were reverse transfected with 50 nM siRNA or esiRNA using Lipofectamine 2000 (Invitrogen) in complete medium without antibiotics. Medium was changed to complete medium 14 hours after transfection and samples were collected. For experiments involving radixin knockdown, radixin 2 siRNA (as specified in supplementary material Table S1) was used unless otherwise indicated.

For DNA transfection cells were transfected using indicated plasmids. 1 µg of DNA in 100 µl of serum-free medium and 3 µl of Lipofectamine 2000 (Invitrogen) in 100 µl of serum-free medium were mixed within 5 minutes and the mixture added into cells in medium without antibiotics for 8 hours before replacement with normal growth medium.

Western blotting

Cell lysates for western blot analysis were prepared at the specified times after treatment using lysis buffer (50 mM Tris-HCl pH 8, 0.5 mM EDTA, 150 mM NaCl, 1% Triton X-100 plus protease and phosphatase inhibitors cocktails). Lysates were mixed with 2× Final Sample Buffer (FSB; Invitrogen) and resolved on 4–12% polyacrylamide gels (Invitrogen), transferred to nitrocellulose membranes and blotted with the indicated antibodies.

Immunofluorescence

PC3 cells grown on 13-mm diameter coverslips were fixed with 4% paraformaldehyde for 20 min. Cells were permeabilised with 0.1% Triton X-100, blocked with blocking buffer (1% BSA, 2% FCS in PBS) and incubated with the indicated primary antibodies diluted in blocking buffer for 1 hour. After three washes with PBS, cells were incubated with fluorochrome-conjugated secondary antibody for 45 minutes. After five washes with PBS, coverslips were mounted in slides using fluorescent mounting medium (Dako).

Time-lapse microscopy

Images of PC3 cells were acquired at 37°C and 5% CO2 using a fully motorized, multi-field time-lapse microscope (Eclipse TE 2000-E; Nikon) with a charge-coupled device camera (ORCA; Hamamatsu Photonics). Images were acquired at different intervals using a 20× or 40× objective. Images were acquired using MetaMorph software (MDS Analytical Technologies), and cells were tracked using ImageJ software (National Institute of Health). Time-lapse videos were acquired for 16 to 24 hours with images taken at 10-min intervals. Analysis of cell speed was performed using the Chemotaxis plug in from Integrated BioDiagnostics GmbH (IBIDI).

Confocal microscopy

Images were acquired at room temperature using a confocal microscope (LSM 510 META; Carl Zeiss, Inc.) with three single photomultiplier tube detectors mounted on a inverted microscope (AxioObserver Z1; Carl Zeiss, Inc.) using a 40× 1.3 NA oil immersion objective (Carl Zeiss, Inc.). Images were acquired using Zen software (Carl Zeiss, Inc.) and processed using Zen and Photoshop (Adobe) software. Image analysis for cell height and morphology was performed using Zen and Image J software.

Rho GTPase activity assays

Cells treated with siRNA against radixin or with siRNA control were scrapped and processed for the Rac/Cdc42 activation assay or the RhoA activation assay according to the manufacturer's instructions (Upstate/Millipore and Cytoskeleton Inc, respectively) using the following lysis buffer: 50 mM Tris-HCl, pH 7.5; 1mM EDTA; 500 mM NaCl, 10 mM MgCl2, 1% (v/v) Triton X-100; 0.5% sodium deoxycholate; 0.1% SDS, 10% (v/v) glycerol; 0.5% (v/v) 2-mercaptoethanol; protease inhibitor cocktail complete (from Roche) and phosphatase inhibitor cocktail II and IV (from Calbiochem). Total input and bound samples were subjected to SDS-PAGE and immunoblot analysis to determine the total and GTP bound Rac1, Cdc42 or RhoA levels.

We are grateful to the members of the Ridley laboratory for their insights during the development of the manuscript and to the Microscopy Facility at St George's University of London for their help during the acquisition of some of the images included in the manuscript.

Funding

This work was supported by Cancer Research UK [grant number C6620/A8833 to A.J.R.]; the Ludwig Institute for Cancer Research (core funding to A.J.R.); the Bettencourt-Schueller Foundation (Prize for the Life Sciences to A.J.R.); King’s College London British Heart Foundation Centre of Excellence [grant number RE/08/003]; and St George’s University of London (start-up grant to F.V.).

Auvinen
E.
,
Kivi
N.
,
Vaheri
A.
(
2007
).
Regulation of ezrin localization by Rac1 and PIPK in human epithelial cells.
Exp. Cell Res.
313
,
824
833
.
Baeyens
N.
,
Horman
S.
,
Vertommen
D.
,
Rider
M.
,
Morel
N.
(
2010
).
Identification and functional implication of a Rho kinase-dependent moesin-EBP50 interaction in noradrenaline-stimulated artery.
Am. J. Physiol. Cell Physiol.
299
,
C1530
C1540
.
Bartholow
T. L.
,
Chandran
U. R.
,
Becich
M. J.
,
Parwani
A. V.
(
2011
).
Immunohistochemical staining of radixin and moesin in prostatic adenocarcinoma.
BMC Clin. Pathol.
11
,
1
.
Belkina
N. V.
,
Liu
Y.
,
Hao
J. J.
,
Karasuyama
H.
,
Shaw
S.
(
2009
).
LOK is a major ERM kinase in resting lymphocytes and regulates cytoskeletal rearrangement through ERM phosphorylation.
Proc. Natl. Acad. Sci. USA
106
,
4707
4712
.
Bretscher
A.
,
Edwards
K.
,
Fehon
R. G.
(
2002
).
ERM proteins and merlin: integrators at the cell cortex.
Nat. Rev. Mol. Cell Biol.
3
,
586
599
.
Brown
M. J.
,
Nijhara
R.
,
Hallam
J. A.
,
Gignac
M.
,
Yamada
K. M.
,
Erlandsen
S. L.
,
Delon
J.
,
Kruhlak
M.
,
Shaw
S.
(
2003
).
Chemokine stimulation of human peripheral blood T lymphocytes induces rapid dephosphorylation of ERM proteins, which facilitates loss of microvilli and polarization.
Blood
102
,
3890
3899
.
Buongiorno
P.
,
Pethe
V. V.
,
Charames
G. S.
,
Esufali
S.
,
Bapat
B.
(
2008
).
Rac1 GTPase and the Rac1 exchange factor Tiam1 associate with Wnt-responsive promoters to enhance beta-catenin/TCF-dependent transcription in colorectal cancer cells.
Mol. Cancer
7
,
73
.
Carnahan
R. H.
,
Rokas
A.
,
Gaucher
E. A.
,
Reynolds
A. B.
(
2010
).
The molecular evolution of the p120-catenin subfamily and its functional associations.
PLoS ONE
5
,
e15747
.
Cernuda–Morollón
E.
,
Millán
J.
,
Shipman
M.
,
Marelli–Berg
F. M.
,
Ridley
A. J.
(
2010
).
Rac activation by the T-cell receptor inhibits T cell migration.
PLoS ONE
5
,
e12393
.
Chuan
Y. C.
,
Pang
S. T.
,
Cedazo–Minguez
A.
,
Norstedt
G.
,
Pousette
A.
,
Flores–Morales
A.
(
2006
).
Androgen induction of prostate cancer cell invasion is mediated by ezrin.
J. Biol. Chem.
281
,
29938
29948
.
Cui
Y.
,
Wu
J.
,
Zong
M.
,
Song
G.
,
Jia
Q.
,
Jiang
J.
,
Han
J.
(
2009
).
Proteomic profiling in pancreatic cancer with and without lymph node metastasis.
Int. J. Cancer
124
,
1614
1621
.
D’Angelo
R.
,
Aresta
S.
,
Blangy
A.
,
Del Maestro
L.
,
Louvard
D.
,
Arpin
M.
(
2007
).
Interaction of ezrin with the novel guanine nucleotide exchange factor PLEKHG6 promotes RhoG-dependent apical cytoskeleton rearrangements in epithelial cells.
Mol. Biol. Cell
18
,
4780
4793
.
del Pozo
M. A.
,
Vicente–Manzanares
M.
,
Tejedor
R.
,
Serrador
J. M.
,
Sánchez–Madrid
F.
(
1999
).
Rho GTPases control migration and polarization of adhesion molecules and cytoskeletal ERM components in T lymphocytes.
Eur. J. Immunol.
29
,
3609
3620
.
Dovas
A.
,
Couchman
J. R.
(
2005
).
RhoGDI: multiple functions in the regulation of Rho family GTPase activities.
Biochem. J.
390
,
1
9
.
Fehon
R. G.
,
McClatchey
A. I.
,
Bretscher
A.
(
2010
).
Organizing the cell cortex: the role of ERM proteins.
Nat. Rev. Mol. Cell Biol.
11
,
276
287
.
Fukumoto
K.
,
Kikuchi
S.
,
Itoh
N.
,
Tamura
A.
,
Hata
M.
,
Yamagishi
H.
,
Tsukita
S.
,
Tsukita
S.
(
2007
).
Effects of genetic backgrounds on hyperbilirubinemia in radixin-deficient mice due to different expression levels of Mrp3.
Biochim. Biophys. Acta
1772
,
298
306
.
Fukuyama
T.
,
Ogita
H.
,
Kawakatsu
T.
,
Inagaki
M.
,
Takai
Y.
(
2006
).
Activation of Rac by cadherin through the c-Src-Rap1-phosphatidylinositol 3-kinase-Vav2 pathway.
Oncogene
25
,
8
19
.
Gautreau
A.
,
Louvard
D.
,
Arpin
M.
(
2000
).
Morphogenic effects of ezrin require a phosphorylation-induced transition from oligomers to monomers at the plasma membrane.
J. Cell Biol.
150
,
193
204
.
Gautreau
A.
,
Louvard
D.
,
Arpin
M.
(
2002
).
ERM proteins and NF2 tumor suppressor: the Yin and Yang of cortical actin organization and cell growth signaling.
Curr. Opin. Cell Biol.
14
,
104
109
.
Göbel
V.
,
Barrett
P. L.
,
Hall
D. H.
,
Fleming
J. T.
(
2004
).
Lumen morphogenesis in C. elegans requires the membrane-cytoskeleton linker erm-1.
Dev. Cell
6
,
865
873
.
Hage
B.
,
Meinel
K.
,
Baum
I.
,
Giehl
K.
,
Menke
A.
(
2009
).
Rac1 activation inhibits E-cadherin-mediated adherens junctions via binding to IQGAP1 in pancreatic carcinoma cells.
Cell Commun. Signal.
7
,
23
.
Hall
A.
(
2009
).
The cytoskeleton and cancer.
Cancer Metastasis Rev.
28
,
5
14
.
Hébert
M.
,
Potin
S.
,
Sebbagh
M.
,
Bertoglio
J.
,
Bréard
J.
,
Hamelin
J.
(
2008
).
Rho-ROCK-dependent ezrin-radixin-moesin phosphorylation regulates Fas-mediated apoptosis in Jurkat cells.
J. Immunol.
181
,
5963
5973
.
Hirao
M.
,
Sato
N.
,
Kondo
T.
,
Yonemura
S.
,
Monden
M.
,
Sasaki
T.
,
Takai
Y.
,
Tsukita
S.
,
Tsukita
S.
(
1996
).
Regulation mechanism of ERM (ezrin/radixin/moesin) protein/plasma membrane association: possible involvement of phosphatidylinositol turnover and Rho-dependent signaling pathway.
J. Cell Biol.
135
,
37
51
.
Hughes
S. C.
,
Fehon
R. G.
(
2007
).
Understanding ERM proteins--the awesome power of genetics finally brought to bear.
Curr. Opin. Cell Biol.
19
,
51
56
.
Hunter
K. W.
(
2004
).
Ezrin, a key component in tumor metastasis.
Trends Mol. Med.
10
,
201
204
.
Ivetic
A.
,
Ridley
A. J.
(
2004
).
Ezrin/radixin/moesin proteins and Rho GTPase signalling in leucocytes.
Immunology
112
,
165
176
.
Kikuchi
S.
,
Hata
M.
,
Fukumoto
K.
,
Yamane
Y.
,
Matsui
T.
,
Tamura
A.
,
Yonemura
S.
,
Yamagishi
H.
,
Keppler
D.
,
Tsukita
S.
, et al. 
. (
2002
).
Radixin deficiency causes conjugated hyperbilirubinemia with loss of Mrp2 from bile canalicular membranes.
Nat. Genet.
31
,
320
325
.
Kitajiri
S.
,
Fukumoto
K.
,
Hata
M.
,
Sasaki
H.
,
Katsuno
T.
,
Nakagawa
T.
,
Ito
J.
,
Tsukita
S.
,
Tsukita
S.
(
2004
).
Radixin deficiency causes deafness associated with progressive degeneration of cochlear stereocilia.
J. Cell Biol.
166
,
559
570
.
Lan
M.
,
Kojima
T.
,
Murata
M.
,
Osanai
M.
,
Takano
K.
,
Chiba
H.
,
Sawada
N.
(
2006
).
Phosphorylation of ezrin enhances microvillus length via a p38 MAP-kinase pathway in an immortalized mouse hepatic cell line.
Exp. Cell Res.
312
,
111
120
.
Lee
J. H.
,
Katakai
T.
,
Hara
T.
,
Gonda
H.
,
Sugai
M.
,
Shimizu
A.
(
2004
).
Roles of p-ERM and Rho-ROCK signaling in lymphocyte polarity and uropod formation.
J. Cell Biol.
167
,
327
337
.
Li
G.
,
Satyamoorthy
K.
,
Herlyn
M.
(
2001
).
N-cadherin-mediated intercellular interactions promote survival and migration of melanoma cells.
Cancer Res.
61
,
3819
3825
.
López–Lago
M.
,
Lee
H.
,
Cruz
C.
,
Movilla
N.
,
Bustelo
X. R.
(
2000
).
Tyrosine phosphorylation mediates both activation and downmodulation of the biological activity of Vav.
Mol. Cell. Biol.
20
,
1678
1691
.
Mackay
D. J.
,
Esch
F.
,
Furthmayr
H.
,
Hall
A.
(
1997
).
Rho- and rac-dependent assembly of focal adhesion complexes and actin filaments in permeabilized fibroblasts: an essential role for ezrin/radixin/moesin proteins.
J. Cell Biol.
138
,
927
938
.
Malliri
A.
,
van Es
S.
,
Huveneers
S.
,
Collard
J. G.
(
2004
).
The Rac exchange factor Tiam1 is required for the establishment and maintenance of cadherin-based adhesions.
J. Biol. Chem.
279
,
30092
30098
.
Matsui
T.
,
Maeda
M.
,
Doi
Y.
,
Yonemura
S.
,
Amano
M.
,
Kaibuchi
K.
,
Tsukita
S.
,
Tsukita
S.
(
1998
).
Rho-kinase phosphorylates COOH-terminal threonines of ezrin/radixin/moesin (ERM) proteins and regulates their head-to-tail association.
J. Cell Biol.
140
,
647
657
.
Millán
J.
,
Cain
R. J.
,
Reglero–Real
N.
,
Bigarella
C.
,
Marcos–Ramiro
B.
,
Fernández–Martín
L.
,
Correas
I.
,
Ridley
A. J.
(
2010
).
Adherens junctions connect stress fibres between adjacent endothelial cells.
BMC Biol.
8
,
11
.
Müller
U.
(
2008
).
Cadherins and mechanotransduction by hair cells.
Curr. Opin. Cell Biol.
20
,
557
566
.
Nakamura
F.
,
Amieva
M. R.
,
Furthmayr
H.
(
1995
).
Phosphorylation of threonine 558 in the carboxyl-terminal actin-binding domain of moesin by thrombin activation of human platelets.
J. Biol. Chem.
270
,
31377
31385
.
Nakamura
N.
,
Oshiro
N.
,
Fukata
Y.
,
Amano
M.
,
Fukata
M.
,
Kuroda
S.
,
Matsuura
Y.
,
Leung
T.
,
Lim
L.
,
Kaibuchi
K.
(
2000
).
Phosphorylation of ERM proteins at filopodia induced by Cdc42.
Genes Cells
5
,
571
581
.
Neisch
A. L.
,
Fehon
R. G.
(
2011
).
Ezrin, Radixin and Moesin: key regulators of membrane-cortex interactions and signaling.
Curr. Opin. Cell Biol.
23
,
377
382
.
Ng
T.
,
Parsons
M.
,
Hughes
W. E.
,
Monypenny
J.
,
Zicha
D.
,
Gautreau
A.
,
Arpin
M.
,
Gschmeissner
S.
,
Verveer
P. J.
,
Bastiaens
P. I.
, et al. 
. (
2001
).
Ezrin is a downstream effector of trafficking PKC-integrin complexes involved in the control of cell motility.
EMBO J.
20
,
2723
2741
.
Niggli
V.
,
Rossy
J.
(
2008
).
Ezrin/radixin/moesin: versatile controllers of signaling molecules and of the cortical cytoskeleton.
Int. J. Biochem. Cell Biol.
40
,
344
349
.
Oshiro
N.
,
Fukata
Y.
,
Kaibuchi
K.
(
1998
).
Phosphorylation of moesin by rho-associated kinase (Rho-kinase) plays a crucial role in the formation of microvilli-like structures.
J. Biol. Chem.
273
,
34663
34666
.
Pang
S. T.
,
Fang
X.
,
Valdman
A.
,
Norstedt
G.
,
Pousette
A.
,
Egevad
L.
,
Ekman
P.
(
2004
).
Expression of ezrin in prostatic intraepithelial neoplasia.
Urology
63
,
609
612
.
Parisiadou
L.
,
Xie
C.
,
Cho
H. J.
,
Lin
X.
,
Gu
X. L.
,
Long
C. X.
,
Lobbestael
E.
,
Baekelandt
V.
,
Taymans
J. M.
,
Sun
L.
, et al. 
. (
2009
).
Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis.
J. Neurosci.
29
,
13971
13980
.
Parri
M.
,
Chiarugi
P.
(
2010
).
Rac and Rho GTPases in cancer cell motility control.
Cell Commun. Signal.
8
,
23
.
Popoff
M. R.
,
Geny
B.
(
2009
).
Multifaceted role of Rho, Rac, Cdc42 and Ras in intercellular junctions, lessons from toxins.
Biochim. Biophys. Acta
1788
,
797
812
.
Prag
S.
,
Parsons
M.
,
Keppler
M. D.
,
Ameer–Beg
S. M.
,
Barber
P.
,
Hunt
J.
,
Beavil
A. J.
,
Calvert
R.
,
Arpin
M.
,
Vojnovic
B.
, et al. 
. (
2007
).
Activated ezrin promotes cell migration through recruitment of the GEF Dbl to lipid rafts and preferential downstream activation of Cdc42.
Mol. Biol. Cell
18
,
2935
2948
.
Ray
R. M.
,
Vaidya
R. J.
,
Johnson
L. R.
(
2007
).
MEK/ERK regulates adherens junctions and migration through Rac1.
Cell Motil. Cytoskeleton
64
,
143
156
.
Rivard
N.
(
2009
).
Phosphatidylinositol 3-kinase: a key regulator in adherens junction formation and function.
Front. Biosci.
14
,
510
522
.
Roch
F.
,
Polesello
C.
,
Roubinet
C.
,
Martin
M.
,
Roy
C.
,
Valenti
P.
,
Carreno
S.
,
Mangeat
P.
,
Payre
F.
(
2010
).
Differential roles of PtdIns(4,5)P2 and phosphorylation in moesin activation during Drosophila development.
J. Cell Sci.
123
,
2058
2067
.
Saotome
I.
,
Curto
M.
,
McClatchey
A. I.
(
2004
).
Ezrin is essential for epithelial organization and villus morphogenesis in the developing intestine.
Dev. Cell
6
,
855
864
.
Schaller
M. D.
(
2001
).
Paxillin: a focal adhesion-associated adaptor protein.
Oncogene
20
,
6459
6472
.
Schaller
M. D.
(
2004
).
FAK and paxillin: regulators of N-cadherin adhesion and inhibitors of cell migration?
J. Cell Biol.
166
,
157
159
.
Shapiro
L.
,
Weis
W. I.
(
2009
).
Structure and biochemistry of cadherins and catenins.
Cold Spring Harb. Perspect. Biol.
1
,
a003053
.
Shaw
L. M.
,
Rabinovitz
I.
,
Wang
H. H.
,
Toker
A.
,
Mercurio
A. M.
(
1997
).
Activation of phosphoinositide 3-OH kinase by the alpha6beta4 integrin promotes carcinoma invasion.
Cell
91
,
949
960
.
Suda
J.
,
Zhu
L.
,
Karvar
S.
(
2011
).
Phosphorylation of radixin regulates cell polarity and Mrp-2 distribution in hepatocytes.
Am. J. Physiol. Cell Physiol.
300
,
C416
C424
.
Sung
S. Y.
,
Hsieh
C. L.
,
Law
A.
,
Zhau
H. E.
,
Pathak
S.
,
Multani
A. S.
,
Lim
S.
,
Coleman
I. M.
,
Wu
L. C.
,
Figg
W. D.
, et al. 
. (
2008
).
Coevolution of prostate cancer and bone stroma in three-dimensional coculture: implications for cancer growth and metastasis.
Cancer Res.
68
,
9996
10003
.
Takahashi
K.
,
Sasaki
T.
,
Mammoto
A.
,
Hotta
I.
,
Takaishi
K.
,
Imamura
H.
,
Nakano
K.
,
Kodama
A.
,
Takai
Y.
(
1998
).
Interaction of radixin with Rho small G protein GDP/GTP exchange protein Dbl.
Oncogene
16
,
3279
3284
.
Tsuda
M.
,
Makino
Y.
,
Iwahara
T.
,
Nishihara
H.
,
Sawa
H.
,
Nagashima
K.
,
Hanafusa
H.
,
Tanaka
S.
(
2004
).
Crk associates with ERM proteins and promotes cell motility toward hyaluronic acid.
J. Biol. Chem.
279
,
46843
46850
.
Valastyan
S.
,
Benaich
N.
,
Chang
A.
,
Reinhardt
F.
,
Weinberg
R. A.
(
2009
).
Concomitant suppression of three target genes can explain the impact of a microRNA on metastasis.
Genes Dev.
23
,
2592
2597
.
Valastyan
S.
,
Chang
A.
,
Benaich
N.
,
Reinhardt
F.
,
Weinberg
R. A.
(
2010
).
Concurrent suppression of integrin α5, radixin, and RhoA phenocopies the effects of miR-31 on metastasis.
Cancer Res.
70
,
5147
5154
.
Valdman
A.
,
Fang
X.
,
Pang
S. T.
,
Nilsson
B.
,
Ekman
P.
,
Egevad
L.
(
2005
).
Ezrin expression in prostate cancer and benign prostatic tissue.
Eur. Urol.
48
,
852
857
.
van Duijn
T. J.
,
Anthony
E. C.
,
Hensbergen
P. J.
,
Deelder
A. M.
,
Hordijk
P. L.
(
2010
).
Rac1 recruits the adapter protein CMS/CD2AP to cell-cell contacts.
J. Biol. Chem.
285
,
20137
20146
.
Yamane
J.
,
Ohnishi
H.
,
Sasaki
H.
,
Narimatsu
H.
,
Ohgushi
H.
,
Tachibana
K.
(
2011
).
Formation of microvilli and phosphorylation of ERM family proteins by CD43, a potent inhibitor for cell adhesion: cell detachment is a potential cue for ERM phosphorylation and organization of cell morphology.
Cell Adhes. Migr.
5
,
119
132
.
Yonemura
S.
,
Matsui
T.
,
Tsukita
S.
,
Tsukita
S.
(
2002
).
Rho-dependent and -independent activation mechanisms of ezrin/radixin/moesin proteins: an essential role for polyphosphoinositides in vivo.
J. Cell Sci.
115
,
2569
2580
.
Zhu
H.
,
Wang
H.
,
Zhang
X.
,
Hou
X.
,
Cao
K.
,
Zou
J.
(
2010
).
Inhibiting N-cadherin-mediated adhesion affects gap junction communication in isolated rat hearts.
Mol. Cells
30
,
193
200
.

Supplementary information