During metastatic progression, an aberrant epithelial-to-mesenchymal transformation (EMT) that is most often driven by the loss of the cell–cell adhesion molecule E-cadherin generates noncohesive tumor cells that are highly invasive. We used mesenchymally transformed, E-cadherin–negative MDA-MB-231 breast carcinoma cells in a natural product screen and determined that the triterpenoid saponin sarasinoside A1 inhibited their invasion and the invasion of a number of other tumor cell lines. Sarasinoside A1 also caused MDA-MB-231 cells to become cohesive in a three-dimensional basement membrane and collagen gel cultures. In two-dimensional culture, sarasinoside A1 initiated a morphologic re-epithelialization of MDA-MB-231 cells wherein preexisting nonepithelial cadherins and the junction-associated proteins β-catenin and ZO-1 all relocalized to sites of cell–cell contact. In addition, the intercellular space between neighboring cells narrowed considerably, the stability of polymerized actin at cell-cell contact sites increased, and there was a recruitment and stabilization of nectin-based adhesion complexes to these sites, all of which strongly suggested that functional cell–cell junctions had formed. Importantly, sarasinoside A1 induced nascent cell–cell junction formation that did not require changes in gene expression and was not associated with an induction of E-cadherin but resulted in increased activation of Rap GTPases. Therefore, our findings with sarasinoside A1 suggest that it may be possible to re-epithelialize metastatic tumor cells with phenotypic consequence even when E-cadherin is completely absent. Mol Cancer Res; 11(5); 530–40. ©2013 AACR.

This article is featured in Highlights of This Issue, p. 441

Tumor cell invasion of the extracellular matrix (ECM) is an important driver of metastasis (1). Although target-based approaches have been used to identify a number of compounds with anti-invasive properties (2), we and others have developed phenotypic screens to identify structurally diverse invasion inhibitors that most often act by modulating cellular interactions with the ECM (3). In this report, we describe a newly identified invasion inhibitor that is also capable of facilitating cell–cell interactions.

The overwhelming majority of solid human tumors arise within epithelial tissues that consist of cohesive, stationary cells. Normally, highly regulated epithelial-to-mesenchymal transformations (EMT) generate noncohesive migratory cells that facilitate morphogenetic tissue rearrangements during organ development. However, as solid tumors progress, this process is often co-opted to generate noncohesive, mesenchymal tumor cells that are not subject to normal regulatory controls and are thus highly invasive (4).

Most approaches to controlling tumor invasion have focused on controlling the motile phenotype in relative isolation. Thus, highly selective inhibitors of potential motility factors such as TGF-β1, EGF, and endothelin have been developed (5–7). In addition, compounds that specifically disrupt integrin-dependent interactions with the ECM have been used with widely varying degrees of preclinical and clinical efficacy (2). In an effort to identify compounds that inhibit invasion by novel means, we developed a cell-based phenotypic screen that eliminates false-positives due to cytotoxicity (8). We then used this assay to identify several structurally diverse invasion inhibitors that act by modulating cellular interactions with the ECM without directly affecting integrin–ligand interactions (8–13). In this report, we describe a newly identified invasion inhibitor, the triterpenoid saponin sarasinoside A1.

During normal development, an EMT is initiated by a transcriptionally mediated downregulation of E-cadherin expression that leads to a dismantling of cell–cell junctions. Thus, considerable effort has been expended to develop strategies that can specifically reverse this change in gene expression in tumor cells (4). However, in a number of tumor contexts, E-cadherin is permanently lost due to mutation or stable epigenetic silencing. Thus, identifying compounds that can suppress the mesenchymal phenotype in the absence of E-cadherin could facilitate the development of new antimetastatic therapeutic strategies. Here, we report that sarasinoside A1 may be just such a compound as it re-epithelializes mesenchymally transformed tumor cells and initiates the formation of nascent cell–cell junctions in an E-cadherin–independent manner.

Isolation of sarasinoside A1

The marine sponge Melophlus sarassinorum was collected by hand, using scuba, from Papua New Guinea. Samples were deep frozen on site, transported on dry ice and methanol extracts were prepared. Sarasinoside A1 was isolated from the sponge extract by assay-guided fractionation, using the invasion assay described below. Briefly, the aqueous fraction from the original methanol extract was concentrated, taken up in H2O, and extracted with nBuOH. The resulting active fraction was subjected to LH20 chromatography in MeOH to give a cream solid which was further purified by elution through a C18 sep-pak (gradient of CH3CN in 0.05% trifluoracetic acid, TFA). Active fractions were combined and subjected to reverse-phase high-performance liquid chromatography (HPLC; Inertsil C18 column, 40% CH3CN in 0.05% TFA). This generated sarasinoside A1 (Fig. 1A) as a white amorphous solid that was soluble in dimethyl sulfoxide (DMSO) and which generated NMR data consistent with literature values (14–16).

Figure 1.

Sarasinoside A1 inhibits cellular invasion and migration. A, structure of the triterpenoid saponin sarasinoside A1. B, inhibition of invasion of MDA-MB-231 cells into a reconstituted basement membrane gel (Matrigel) matrix by sarasinoside A1 (SaraA). Viable, noninvasive cells are quantified by MTT assay (OD 570) such that invasion inhibition is a positive readout. Shown are averages of triplicates ± SD. **, P < 0.005 compared with 0 μg/mL as determined by 2-tailed Student t test. C, confluent 2-D cell monolayers were wounded with a sterile pipette tip, and cells were allowed to migrate into the wound over an 18-hour period in the absence or presence of 20 μg/mL sarasinoside A1 and imaged, live, by phase microscopy. Scale bar, 50 μm.

Figure 1.

Sarasinoside A1 inhibits cellular invasion and migration. A, structure of the triterpenoid saponin sarasinoside A1. B, inhibition of invasion of MDA-MB-231 cells into a reconstituted basement membrane gel (Matrigel) matrix by sarasinoside A1 (SaraA). Viable, noninvasive cells are quantified by MTT assay (OD 570) such that invasion inhibition is a positive readout. Shown are averages of triplicates ± SD. **, P < 0.005 compared with 0 μg/mL as determined by 2-tailed Student t test. C, confluent 2-D cell monolayers were wounded with a sterile pipette tip, and cells were allowed to migrate into the wound over an 18-hour period in the absence or presence of 20 μg/mL sarasinoside A1 and imaged, live, by phase microscopy. Scale bar, 50 μm.

Close modal

Cell culture and reagents

MDA-MB-231 cells were originally acquired from Erik Thompson (University of Melbourne, Melbourne Australia) and were routinely tested for an absence of epithelial markers, including E-cadherin, and mesenchymal invasiveness as shown here under control conditions (see Figs. 1–3). A375 cells were from American Type Culture Collection (ATCC). Both of these cell lines were maintained in monolayer culture in Dulbecco's Modified Eagle's Medium (DMEM; Invitrogen) containing 10% FBS (Hyclone). MCF7 cells were grown in DMEM/F12 (Sigma) with 5% FBS and SKOV3 cells were maintained in 199/105 medium (Sigma) with 5% FBS. All cells were grown at 37°C in a humidified 5% CO2 incubator. The pIRM2.1-IRES-dsFP593 vector encoding FLAG-Rap1V12 was from Michiyuki Matsuda (Kyoto University, Kyoto, Japan). The pEGFP-N1-Nectin2 construct was from Julian Guttman (Simon Fraser University, Burnaby, Canada), actin-GFP was from Invitrogen, and paxillin-GFP was kindly provided by Robert Nabi (University of British Columbia, Vancouver, BC, Canada). Cells were transfected transiently using Lipofectamine 2000 (Invitrogen).

Figure 2.

Sarasinoside A1 inhibits MDA-MB-231 cell invasion and increases cell–cell cohesion in 3-D culture. The morphology of MDA-MB-231 cells plated on (A) and within (B) a reconstituted basement membrane gel (Matrigel) in the absence (DMSO vehicle) or presence (SaraA) of 20 μg/mL of sarasinoside A1 was assessed by live phase microscopy (Scale bar, 50 μm). C, F-actin- and DAPI (nuclear)-labeled cells were maintained in the absence or presence of sarasinoside A1 within collagen gels and their elongated, invasive morphology was assessed by confocal microscopy. D, F-actin- and CMFDA dye (cytoplasm)-labeled cells where plated on collagen gels and their morphology and invasion into the gel in the absence or presence of sarasinoside A1 were assessed by sectioning and staining the gels followed by microscopic assessment in the xy orientation. The site of the collagen gel is marked by the bar on left.

Figure 2.

Sarasinoside A1 inhibits MDA-MB-231 cell invasion and increases cell–cell cohesion in 3-D culture. The morphology of MDA-MB-231 cells plated on (A) and within (B) a reconstituted basement membrane gel (Matrigel) in the absence (DMSO vehicle) or presence (SaraA) of 20 μg/mL of sarasinoside A1 was assessed by live phase microscopy (Scale bar, 50 μm). C, F-actin- and DAPI (nuclear)-labeled cells were maintained in the absence or presence of sarasinoside A1 within collagen gels and their elongated, invasive morphology was assessed by confocal microscopy. D, F-actin- and CMFDA dye (cytoplasm)-labeled cells where plated on collagen gels and their morphology and invasion into the gel in the absence or presence of sarasinoside A1 were assessed by sectioning and staining the gels followed by microscopic assessment in the xy orientation. The site of the collagen gel is marked by the bar on left.

Close modal
Figure 3.

Sarasinoside A1 morphologically epithelializes MDA-MB-231 cells. MDA-MB-231 cells were maintained in 2-D-monolayer culture and were treated for 4 h or 24 h with sarasinoside A1 (SaraA) and their morphology was assessed, live, by phase microscopy (A, upper panel, scale bar = 50 μm). Alternatively, cells were fixed, stained for either f-actin (A, middle panel) or E-cadherin (A, lower panel), and imaged by confocal microscopy. MDA-MB-231 cells were also stably transfected with E-cadherin (MDA231/ECad) and used as an epithelialized control in these experiments (A, far right images). Whole cell lysates from cells treated as described above were subjected to Western blotting for either E-cadherin or f-actin as a loading control (B).

Figure 3.

Sarasinoside A1 morphologically epithelializes MDA-MB-231 cells. MDA-MB-231 cells were maintained in 2-D-monolayer culture and were treated for 4 h or 24 h with sarasinoside A1 (SaraA) and their morphology was assessed, live, by phase microscopy (A, upper panel, scale bar = 50 μm). Alternatively, cells were fixed, stained for either f-actin (A, middle panel) or E-cadherin (A, lower panel), and imaged by confocal microscopy. MDA-MB-231 cells were also stably transfected with E-cadherin (MDA231/ECad) and used as an epithelialized control in these experiments (A, far right images). Whole cell lysates from cells treated as described above were subjected to Western blotting for either E-cadherin or f-actin as a loading control (B).

Close modal

Assay for invasion inhibition

Invasion inhibition assays were conducted as previously described and validated (8). Briefly, tumor cells were plated on reconstituted basement membrane gels (Matrigel; BD Biosciences) along with sarasinoside A1 or DMSO as a vehicle control. After 2.5 hours, noninvasive cells were recovered from the top surface of the Matrigel, and the number of viable cells was quantified using a MTT assay.

Cell viability and proliferation assays

To measure cell viability, cells were plated in 96-well plates to near confluency. Sarasinoside A1 was added at various concentrations in triplicate, and cell viability was measured 24 hours later by MTT assay. To determine cell proliferation, cells were plated in 96-well plates at 5 × 103 cells/well and incubated with different concentrations of sarasinoside A1, with daily change of medium and drug. Cell proliferation was measured at 0, 24, and 72 hours using an MTT assay.

Three-dimensional invasion assays

For basement membrane invasion, cells were plated on top of, or were embedded within, reconstituted basement membrane gels (Matrigel, BD Biosciences) as previously described (17) in the absence or presence of sarasinoside A1 as indicated and then imaged, live, by phase microscopy. For collagen invasion, CMFDA-labeled cells (Invitrogen) were plated on collagen I (2 mg/mL in PBS; BD Biosciences) that was polymerized on Transwell filters (BD Falcon). The lower chamber contained media with 55% FBS as a chemoattractant. After 20 hours, filters were fixed with paraformaldehyde, embedded in optimal cutting temperature compound (OCT), cryosectioned vertically, stained for actin (see below), and z-axis images were acquired as shown.

Immunofluorescence staining

Cells were plated on coverslips in serum-containing medium and maintained for 48 hours. Medium containing sarasinoside A1 or DMSO was then added to the cells for the specified lengths of time before fixation. Cells were fixed with 3% paraformaldehyde for 15 minutes at room temperature, permeabilized with 0.3% Triton X-100 in PBS for 15 minutes, and blocked with 3% bovine serum albumin (BSA; Sigma) in PBS for 30 minutes, followed by incubation with the following primary antibodies: mouse clone 14 β-catenin antibody (BD Transduction) at 1:100 dilution, rabbit polyclonal pan-cadherin antibody (Abcam ab16505) at 1:250, mouse clone 36 E-cadherin antibody (BD Transduction) at 1:100, and rabbit polyclonal ZO-1 antibody (Invitrogen) at 1:100, incubated overnight at 4°C. An Alexa Fluor 488–conjugated goat secondary antibody (Molecular Probes) was used at a dilution of 1 in 150. To visualize F-actin, cells were stained with a 165 nmol/L solution of rhodamine-phalloidin (Invitrogen). Nuclei were stained with a 500 ng/mL solution of 4′,6-diamidino-2-phenylindole (DAPI) dihydrochloride (Sigma). All images were obtained using the Uplan Apochromat 60×/1.35 numerical aperture objective on an Olympus FV1000 confocal microscope and captured using Olympus FluoView v1.6 software.

Immunoblotting

Cells were treated with either DMSO or 20 μg/mL sarasinoside A1 for the indicated times. Cells were lysed in radioimmunoprecipitation (RIPA) buffer containing protease inhibitors [50 mmol/L Tris, pH 7.5, 150 mmol/L NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 20 μg/mL aprotinin, 10 μg/mL leupeptin, 100 μmol/L phenylmethanesulfonylfluoride (PMSF), 10 μg/mL pepstatin A, 2.5 mmol/L EDTA] at 4°C for 30 minutes with agitation and then clarified through centrifugation at 10,000 × g for 15 minutes at 4°C. All samples for electrophoresis were quantified with a BCA protein assay (Pierce) and equalized for total protein before electrophoresis. The following primary antibodies were used: mouse clone AC-40 actin (Sigma) at 1:500 dilution, mouse clone 14 β-catenin (BD Transduction) at 1:500 dilution, rabbit polyclonal pan-cadherin (Abcam ab16505) at 1:500, mouse clone 36 E-cadherin (BD Transduction) at 1:1,000, and rabbit polyclonal ZO-1 (Invitrogen) at 1:500.

Transmission electron microscopy

MDA-MB-231 cells on 1- μm pore culture inserts (Falcon 35-3104) were treated with 20 μg/mL sarasinoside A1 or DMSO for 24 hours. Cells were fixed (1.5% paraformaldehyde, 1.5% glutaraldehyde, 0.1 mol/L sodium cacodylate, pH 7.3) at room temperature for 3 hours, placed in buffer (0.1 mol/L sodium cacodylate, pH 7.3) overnight at room temperature, post-fixed for 1 hour on ice (1:1 mixture of 2% OsO4 in ddH2O and 0.2 mol/L sodium cacodylate, pH 7.3), washed 3× with ddH2O at room temperature, stained en bloc for 1 hour (1% aqueous uranyl acetate), and washed again 3× with ddH2O. Finally, samples were dehydrated through a graded series of ethyl alcohol, passed through propylene oxide into a 1:1 mixture of propylene oxide and EMbed-812 (Electron Microscopy Sciences), followed by overnight infiltration and embedding in EMbed-812. Thin sections were stained with uranyl acetate and lead citrate and photographed on a Philips 300 electron microscope operated at 60 kV.

Fluorescence recovery after photobleaching

Fluorescence recovery after photobleaching (FRAP) was conducted on MDA-MB-231 cells expressing actin-GFP, paxillin-GFP, or Nectin2-GFP seeded on chamber slides (Ibidi). The indicated regions of interest were photobleached using a 405-nm laser (100% intensity, 0.1 seconds) on an Olympus FV1000 confocal microscope and captured using Olympus FluoView v1.6 software. Fluorescence recovery was followed at 4- or 5-second time intervals for at least 1 minute. The fluorescence signal was normalized to the prebleach intensity, and single exponential fit curves of the data were generated as previously described (18).

GTPase activation assays

Activated, GTP-bound Rho proteins were isolated by precipitation with the Rho-binding domain of rhotekin and quantified by Western blotting according to the manufacturer's instructions (Millipore). Rap1-GTP was precipitated from cell lysates using a glutathione S-transferase–RalGDS fusion protein and detected by immunoblotting with a Rap1 antibody as described (19).

Isolation and characterization of sarasinoside A1 as a tumor cell invasion inhibitor

We previously developed a cell-based phenotypic screening assay and used it to identify a number of tumor cell invasion inhibitors (8–13). In this report, we used this screen to identify the triterpenoid saponin sarasinoside A1 as an invasion inhibitor based on the assay-guided fractionation of a marine sponge extract (Fig. 1A). Specifically, sarasinoside A1 inhibited the invasion of highly metastatic and mesenchymal MDA-MB-231 breast cancer cells into a reconstituted basement membrane matrix (i.e., Matrigel) at a concentration of 20 μg/mL (Fig. 1B). The same concentration also inhibited MDA-MB-231 cell migration on a planar, rigid tissue culture plastic substratum in 2-dimensional (2-d) monolayer culture (Fig. 1C). At this dose, sarasinoside A1 was not cytotoxic, although it was reversibly antiproliferative (Supplementary Fig. S1A). It is important to note, however, that higher concentrations of sarasinoside A1 did cause some cytotoxicity (LC50 = 53 μg/mL; Supplementary Fig. S1B) as has been reported previously in studies in which its effect on cell movement was not assessed (16, 20).

Sarasinoside A1 inhibits cell elongation and facilitates cohesive cell cluster formation in 3-D culture

When they were plated on top of prepolymerized Matrigel, mesenchymal MDA-MB-231 cells elongated as they began to invade the matrix (Fig. 2A, DMSO vehicle control). In contrast, sarasinoside A1–treated MDA-MB-231 cells remained rounded on top of the gel (Fig. 2A, SaraA image). This anti-invasive, morphologic effect was also observed when highly invasive A375 melanoma and SKOV3 ovarian carcinoma cells were plated on top of Matrigel in the presence of sarasinoside A1 (Supplementary Fig. S2).

When they were embedded within the Matrigel, MDA-MB-231 cells also elongated as they became invasive and mesenchymal (Fig. 2B, DMSO vehicle control). Interestingly, under these conditions, the sarasinoside A1–treated cells did not elongate. Instead, they clustered together and formed spheroidal structures which is a characteristic of nonmetastatic, epithelialized tumor cells (Fig. 2B, SaraA image; ref. 21). In our experience, this cell clustering in 3-D culture, which is suggestive of increased cell–cell adhesion, is not induced by any of the other tumor cell invasion inhibitors we have identified (data not shown).

To determine whether the anti-invasive cell clustering described above was restricted to highly flexible, low-compliance matrices such as Matrigel, MDA-MB-231 cells were also embedded within more rigid type I collagen gels (Fig. 2C). Under the latter conditions, sarasinoside A1 also inhibited invasive cell elongation and led to the formation of small cohesive cell clusters (Fig. 2C, arrowhead). When untreated MDA-MB-231 cells were instead plated on top of the collagen gels and their invasive behavior was directionally stimulated by a chemoattractant (FBS) delivered from below, the cells pulled apart from each other and moved into the gel as occurs at the invasive front of mesenchymally transformed tumor masses in vivo (Fig. 2D, DMSO vehicle, invasion occurred downward into the collagen gel represented by the bar). In contrast, sarasinoside A1–treated cells formed a cohesive cell mass that resembled a stratified epithelium (Fig. 2D, SaraA image), and they remained stationary and noninvasive on top of the gel.

Sarasinoside A1 induces an epithelial reversion of MDA-MB-231 cells maintained in 2-D monolayer culture

When MDA-MB-231 cells maintained as 2-D monolayers on tissue culture plastic were treated with sarasinoside A1, the mesenchymal, spindle-shaped cells underwent a flattening and a morphologic epithelialization (i.e., they formed “cobblestone”-like monolayers; Fig. 3A). As a result, after 24 hours, the sarasinoside A1–treated monolayers resembled the monolayers that were formed by MDA-MB-231cells that had been reverted to an epithelial phenotype in response to the stable, forced expression of exogenous E-cadherin (Fig. 3A, top phase, MDA231/E-Cad stable transfectants).

In untreated MDA-MB-231 cells, considerable filamentous actin was concentrated at the terminal, lamellar edges of the migratory, mesenchymal cells (Fig. 3A, f-actin, left image, arrowheads). Sarasinoside A1 treatment eliminated these structures, and the majority of the F-actin instead relocalized to sites of cell–cell interaction (Fig. 3A, F-actin panels after 4- and 24-hour treatment). This F-actin relocalization was not associated with an increase or accumulation of endogenous E-cadherin in the sarasinoside A1–treated cells (Fig. 3A; E-cadherin; MDA231/ECad transfectants served as a positive control). This lack of endogenous E-cadherin upregulation in sarasinoside A1–treated cells was confirmed by Western blotting (Fig. 3B). Importantly, the sarasinoside A1–induced epithelialization of MDA-MB-231 cells also occurred in the presence of cycloheximide or actinomycin D, inhibitors of protein translation and transcription, respectively (data not shown). Thus, sarasinoside A1 appears to have caused a morphologic mesenchymal-to-epithelial reversion that did not require global changes in gene expression or a specific upregulation of E-cadherin.

Sarasinoside A1 initiates cell–cell junction formation in MDA-MB-231 cells

MDA-MB-231 cells do not express the classical epithelial cell–cell junction molecule E-cadherin due to epigenetic silencing (22). They do, however, express other cadherins, including R-cadherin and cadherin-11. As often occurs in mesenchymally transformed tumor cells, these cadherins do not initiate cell–cell junction formation in MDA-MB-231 cells. Thus, as expected, immunofluorescent staining indicated that these nonepithelial cadherins were diffusely localized in control cells (Fig. 4A, DMSO vehicle control). In contrast, sarasinoside A1 treatment caused these endogenously expressed cadherins to relocalize to sites of cell–cell contact together with the cell junction–associated proteins β-catenin and ZO-1, none of which exhibited a demonstrable change in their steady-state protein levels (Fig. 4A and B). These observations reinforce the notion that the sarasinoside A1–mediated re-epithelialization does not rely on changes in gene expression.

Figure 4.

Cells relocalize junctional proteins to cell–cell contact sites upon treatment with sarasinoside A1. A, images of MDA-MB-231 cells in 2-D monolayer culture treated without (DMSO, vehicle) or with 20 μg/mL sarasinoside A1 (SaraA). Cells were fixed and costained for nuclei using DAPI (blue) together with pan-cadherin, β-catenin, or ZO-1 by immunofluorescence (green). Alternatively, cells were transiently transfected with nectin-2-GFP and localization of this cell junction–associated molecule was assessed by GFP fluorescence (green). Images were generated by confocal microscopy (scale bar = 20 μm). B, lysates of cells maintained without and with sarasinoside A1 were subjected to Western blotting to show that the steady-state levels of endogenous cadherins, β-catenin, and ZO-1 do not change with sarasinoside A1 treatment. Actin was used as a loading control. C, cells in monolayer without (DMSO) and with sarasinoside A1 (SaraA) culture were subjected to transmission electron microscopy and sites of close cell–cell proximity are noted within the boxes. Arrowheads indicate dual nuclear membranes for an internal distance control. Scale bar, 0.5 μm.

Figure 4.

Cells relocalize junctional proteins to cell–cell contact sites upon treatment with sarasinoside A1. A, images of MDA-MB-231 cells in 2-D monolayer culture treated without (DMSO, vehicle) or with 20 μg/mL sarasinoside A1 (SaraA). Cells were fixed and costained for nuclei using DAPI (blue) together with pan-cadherin, β-catenin, or ZO-1 by immunofluorescence (green). Alternatively, cells were transiently transfected with nectin-2-GFP and localization of this cell junction–associated molecule was assessed by GFP fluorescence (green). Images were generated by confocal microscopy (scale bar = 20 μm). B, lysates of cells maintained without and with sarasinoside A1 were subjected to Western blotting to show that the steady-state levels of endogenous cadherins, β-catenin, and ZO-1 do not change with sarasinoside A1 treatment. Actin was used as a loading control. C, cells in monolayer without (DMSO) and with sarasinoside A1 (SaraA) culture were subjected to transmission electron microscopy and sites of close cell–cell proximity are noted within the boxes. Arrowheads indicate dual nuclear membranes for an internal distance control. Scale bar, 0.5 μm.

Close modal

Nectins are Ca2+-independent Ig-like cell adhesion molecules that facilitate cell–cell adhesion in diverse homotypic and heterotypic cell–cell interactions (23). Nectin-based adhesion complexes, which are often disrupted in tumor cells, promote the initiation and stabilization of cadherin-mediated adherens junctions by facilitating the homophilic binding of cadherins expressed on the surface of juxtaposed cells. To determine whether sarasinoside A1 could re-localize exogenous nectins, nectin2-GFP was expressed in MDA-MB-231 cells. Similar to endogenous cell junction proteins in these tumor cells, nectin2-GFP was diffusely localized in control cells but it was recruited to sites of cell–cell contact after sarasinoside A1 treatment (Fig. 4A). In addition, transmission electron microscopy indicated that neighboring sarasinoside A–treated cells had very closely apposed plasma membranes compared with controls (Fig. 4C), morphologic evidence of adhesion between cells. Taken together, these data suggest that sarasinoside A1 initiates nectin-based cell–cell junction formation in MDA-MB-231 cells.

Sarasinoside A1 activates RapGTPases and stabilizes nectin-based adhesion complexes

The formation of nectin-based adhesion complexes can result in activation of the Rap1 GTPase, an event that is required for adherens junction formation (24). The activation of Rap has also been shown to stabilize both- cadherin (25) and integrin-based adhesions (18) in an actin-dependent manner, which reduces the invasive capacity of many types of tumor cells. In contrast, activation of Rho GTPases can drive tumor cell migration and invasion by increasing cellular contractility and focal adhesion dynamics (26, 27). We therefore asked whether treating MDA-MB-231 cells with sarasinoside A1 altered the activation of Rho and Rap GTPases. We found that sarasinoside A1 had no effect on Rho activation but substantially increased the amount of activated Rap1 in the cells (Fig. 5A, B).

Figure 5.

Sarasinoside A1 activates Rap GTPases and stabilizes nectin-based adhesion complexes. A and B, MDA-MB-231 cells were maintained in 2-D monolayer culture and were treated for 30 minutes or 4 hours with sarasinoside A1 (SaraA), 0.1 mg/mL of the Rho activator calpeptin (A), or 500 nmol/L of the Rap activator PMA (B). Whole-cell lysates from cells were subject to GST-pulldown with the Rho-binding domain of rhotekin (A) or RalGDS (B) to detect active forms of Rho and Rap GTPases. Lysates for total Rho and Rap were used as input controls. C, MDA-MB-231 cells transiently expressing nectin2-GFP alone or with Rap1V12 (inset) were either maintained without (DMSO) or treated with 20 μg/mL of sarasinoside A (SaraA) for 4 hours in monolayer culture. Regions of interest (red boxes on large image and individual small images below) that represent sites of cell–cell junctions were photobleached and the FRAP, which is a measure of the mobility of the nectin2-GFP, was followed over time (in small images PB = “pre-bleached” region which is followed by the same region shown every 4 seconds after photobleaching beginning at time 0). Fluorescence recovery was quantified by assessing nectin-GFP emission intensity normalized to the prebleached intensity over time (i.e., mobile fraction). Note the increased contact sites (arrows) and decreased mobile fraction of nectin2-GFP in SaraA-treated and Rap1V12-expressing MDA-MB-231 cells.

Figure 5.

Sarasinoside A1 activates Rap GTPases and stabilizes nectin-based adhesion complexes. A and B, MDA-MB-231 cells were maintained in 2-D monolayer culture and were treated for 30 minutes or 4 hours with sarasinoside A1 (SaraA), 0.1 mg/mL of the Rho activator calpeptin (A), or 500 nmol/L of the Rap activator PMA (B). Whole-cell lysates from cells were subject to GST-pulldown with the Rho-binding domain of rhotekin (A) or RalGDS (B) to detect active forms of Rho and Rap GTPases. Lysates for total Rho and Rap were used as input controls. C, MDA-MB-231 cells transiently expressing nectin2-GFP alone or with Rap1V12 (inset) were either maintained without (DMSO) or treated with 20 μg/mL of sarasinoside A (SaraA) for 4 hours in monolayer culture. Regions of interest (red boxes on large image and individual small images below) that represent sites of cell–cell junctions were photobleached and the FRAP, which is a measure of the mobility of the nectin2-GFP, was followed over time (in small images PB = “pre-bleached” region which is followed by the same region shown every 4 seconds after photobleaching beginning at time 0). Fluorescence recovery was quantified by assessing nectin-GFP emission intensity normalized to the prebleached intensity over time (i.e., mobile fraction). Note the increased contact sites (arrows) and decreased mobile fraction of nectin2-GFP in SaraA-treated and Rap1V12-expressing MDA-MB-231 cells.

Close modal

To test the idea that sarasinoside A1 alters the dynamics of de novo adhesion formation in a Rap-dependent manner, we expressed nectin2-GFP and monitored FRAP at the membrane. Nectin-based contacts between DMSO-treated cells were rare and, when formed, were highly dynamic (Fig. 5C). These contacts often disassembled during the imaging period (data not shown) and those that were maintained showed a high recovery of nectin2-GFP that is indicative of either trafficking-mediated turnover or lateral mobility of nectin within the membrane (Fig. 5C, left). In contrast, in sarasinoside A1–treated cells, we frequently observed nectin2-GFP at cell–cell contacts and the nectin2-GFP at these adhesions was stable, that is, it exhibited a reduced mobile fraction when bleached compared with the nectin2-GFP in control cells (Fig. 5C, middle). Because sarasinoside A1 potently activated the Rap GTPases, we asked whether expressing a constitutively active form of Rap1 (Rap1V12) was sufficient for stabilizing nectin-based adhesions between MDA-MB-231 cells. Indeed, expressing Rap1V12 also led to a stabilization (i.e., reduced mobile fraction) of nectin2-GFP in the membrane at these contact sites, and this stabilization was not further enhanced by treating the cells with sarasinoside A1 (Fig. 5C, right). This suggests that sarasinoside A1 stabilizes nectin-based junctional complexes, at least in part, due to its ability to activate Rap1.

Sarasinoside A1 stabilizes F-actin at putative cell–cell junctions

When cell–cell junctions form, networks of filamentous actin (F-actin) are stabilized at the cytoplasmic face of the junctional complex where they act to physically link neighboring cells across the junction (28). To assess F-actin localization and stability, we used FRAP. Actin-GFP was expressed in MDA-MB-231 cells and regions of actin at sites of cell–cell contact were bleached. In control cells, actin-GFP was rarely incorporated into F-actin filaments that accumulated at small punctate sites of cell–cell interaction (Fig. 6A, DMSO, vehicle control). In contrast, sarasinoside A1–treated cells exhibited long, continuous accumulations of F-actin at sites of cell–cell interaction (Fig. 6A). In addition, FRAP-based recovery of actin-GFP intensity was significantly reduced at these sites in sarasinoside A1–treated cells, which is indicative of increased stability due to the maintenance of the photobleached actin (Fig. 6A). Interestingly, sarasinoside A1 did not increase the stability of cell–ECM contacts (i.e., focal adhesions), as there was no difference in the fluorescence recovery after bleaching of transfected paxillin-GFP, an actin-binding structural component of the focal adhesions (Fig. 6B). Taken together, these data show that sarasinoside A1 selectively increased the stability of F-actin networks that underlie the de novo cell–cell adhesions that formed between cells that would normally have been mesenchymal and invasive.

Figure 6.

Sarasinoside A increases the stability of cell–cell junction–associated actin. MDA-MB-231 cells were transiently expressing either actin-GFP or paxillin-GFP were maintained without (DMSO) or with 20 μg/mL of sarasinoside A (SaraA) for 4 hours in monolayer culture. Regions of interest (red boxes on large image and individual small images below) that represent sites of cell–cell junctions (A, actin-GFP) or cell–ECM/focal adhesion junctions (B, paxillin GFP) were photobleached and the FRAP, which is a measure of the mobility of the GFP-tagged molecule of interest, was followed over time (in small images PB = “pre-bleached” region which is followed by the same region shown every 5 seconds after photobleaching beginning at time 0). Fluorescence recovery was quantified by assessing actin-GFP emission intensity for cell-cell junctions in (A) or paxillin-GFP emission intensity for focal adhesions in (B). Note the decrease in mobility of actin-GFP in SaraA–treated cell–cell junctions, but not in focal adhesions, which is an indicator of increased stability.

Figure 6.

Sarasinoside A increases the stability of cell–cell junction–associated actin. MDA-MB-231 cells were transiently expressing either actin-GFP or paxillin-GFP were maintained without (DMSO) or with 20 μg/mL of sarasinoside A (SaraA) for 4 hours in monolayer culture. Regions of interest (red boxes on large image and individual small images below) that represent sites of cell–cell junctions (A, actin-GFP) or cell–ECM/focal adhesion junctions (B, paxillin GFP) were photobleached and the FRAP, which is a measure of the mobility of the GFP-tagged molecule of interest, was followed over time (in small images PB = “pre-bleached” region which is followed by the same region shown every 5 seconds after photobleaching beginning at time 0). Fluorescence recovery was quantified by assessing actin-GFP emission intensity for cell-cell junctions in (A) or paxillin-GFP emission intensity for focal adhesions in (B). Note the decrease in mobility of actin-GFP in SaraA–treated cell–cell junctions, but not in focal adhesions, which is an indicator of increased stability.

Close modal

The loss of E-cadherin initiates an EMT in many cancers that arise within the epithelia. In the specific case of lobular breast carcinoma, genetic mutation followed by LOH causes a loss of E-cadherin and the generation of invasive tumor cells that move into the surrounding tissue stroma in a single file pattern that is highly characteristic of mesenchymal invasion (29). In contrast, in ductal breast carcinoma, E-cadherin expression is more often downregulated by transcriptional repressors such as Snail and Slug followed by epigenetic silencing (30). In the mesenchymally transformed MDA-MB-231 breast carcinoma cells used in this study, E-cadherin silencing has been achieved through hypermethylation of the E-cadherin promoter (22). While it is possible to reverse this stable silencing by genetic manipulation (31), it has proven difficult to do so, particularly in a gene-specific manner, by pharmacologic means.

The natural product sarasinoside A1 was first isolated from a Palauan marine sponge more than 20 years ago when it was originally described as being mildly cytotoxic (14, 16, 20). We confirmed this cytotoxicity at high doses. However, at lower, noncytotoxic doses, sarasinoside A1 inhibited MDA-MB-231 tumor cell invasion, caused a morphologic epithelial reversion, and it initiated cell–cell junction formation. Importantly, all of these effects occurred in the absence of any upregulation of E-cadherin.

When epithelially derived tumor cells undergo a mesenchymal transformation, the expression of nonepithelial cadherins often increase as E-cadherin levels decrease. This process has been termed “cadherin switching: (32). Importantly, the upregulated cadherins rarely initiate stable junction formation between the tumor cells, and in some situations, they have been found to contribute to the invasive process by facilitating dynamic, migratory interactions with surrounding stromal and endothelial cells (32). Mesenchymally transformed MDA-MB-231 tumor cells express a number of nonepithelial cadherins (33) which we found to be diffusely localized throughout the cell cytoplasm under control conditions. Sarasinoside A1 treatment caused these cadherins to relocalize to sites of cell–cell interaction which, together with electron microscopic evidence, suggested that the compound causes nonepithelial cadherins to participate in cell–cell adhesion.

Cell–cell adhesion is initiated when the extracellular domains of transmembrane cadherins on the surface of neighboring cells form weak interactions at sites of cell–cell contact. These interactions are strengthened by the recruitment of nectin molecules which interact with ligand-engaged cadherins in the plane of the membrane and facilitate their stable clustering at the nascent cell–cell junction (34). Recruitment of nectin-based adhesion complexes are thought to enhance cadherin-mediated adhesion, in part by, binding F-actin via the afadin/AF-6 scaffolding protein (35) and by activating the Rap GTPases (24). We found that sarasinoside A1 facilitated the recruitment and stabilization of exogenously expressed nectin-2-GFP to these sites in MDA-MB-231 cells, suggesting that the compound acts, at least in part, by enhancing nascent junction stabilization. Sarasinoside A1 also selectively activated the Rap GTPases, which may have occurred because of an increase in nectin-based adhesion or the formation of more stable nectin-based cell–cell junctions, resulting in the activation of signaling pathways that promote Rap activation. Moreover, expressing an activated form of Rap1 was sufficient to stabilize nectin in the membrane of contacting MDA-MB-231 cells, suggesting a positive feedback loop in which nectin-based junctional complexes activate Rap1, which in turn acts to further stabilize these nectin-based adhesion complexes.

As nascent cell–cell junctions begin to mature, cytoplasmic scaffolding proteins help generate a physical linkage between the transmembrane cadherins and F-actin at the junctional site (28). Two such scaffolding proteins, β-catenin and ZO-1, were all prominent at sites of cell–cell contact in sarasinoside A1–treated cells. Sarasinoside A1 treatment also stimulated the accumulation of stable F-actin at sites of cell–cell interaction, which is a biophysical indicator of functional cell–cell junction formation (28). Importantly, this stabilization was selective; sarasinoside A1 did not increase the stability of the actin-binding protein paxillin molecule at cell–ECM junctions (i.e., focal adhesions; ref. 36). Thus, the compound does not appear to act as a general F-actin stabilizer such as, for example, jasplakinolide which also inhibits tumor cell invasion but has other substantial deleterious effects (37, 38). This raises the possibility that sarasinoside A1 may act by selectively regulating the activity of F-actin modulators at cell–cell junctions. Interestingly, a number of anti-invasive compounds that act by selectively altering preexisting cell–ECM or cell–cell junction dynamics have been identified in other screens (9, 11, 39, 40).

The Src kinase inhibitor SKI-606 (bosutinib) inhibits mesenchymal breast tumor cell migration and invasion (41), perhaps by decreasing the phosphorylation of focal adhesion proteins (42, 43). Interestingly, SKI-606 also facilitates cell–cell adhesion in a manner that, like sarasinoside A1, does not require either the presence or upregulation of E-cadherin in MDA-MB-231 cells (43). This is very different than the natural compounds 3,3′-diindolylmethane and isoflavone, which also epithelialize tumor cells but do so by upregulating E-cadherin expression in a manner that depends on modulating microRNA levels (44). Thus, sarasinoside A1 may impinge upon Src signaling to re-epithelialize cells. However, this would likely be an indirect effect because, unlike SKI-606 (41, 43), sarasinoside A1 does not inhibit downstream ERK activation (data not shown).

Preventing tumor cell invasion is an important goal of many target-based antimetastatic drug development efforts. While compounds of potential therapeutic value have been identified using this approach, few have been shown to specifically reverse a stable EMT. However, like the targeted molecule SKI-606, the natural compound sarasinoside A1, appears to initiate such a reversion. Importantly, the fact that SKI-606 and sarasinoside A1 both initiate this reversion in an E-cadherin–independent manner opens the door to the further development of novel antimetastatic therapies that suppress the mesenchymal phenotype, at least in part, by inducing cell–cell adhesion using preexisting adhesion molecules on the surface of metastatic tumor cells.

No potential conflicts of interest were disclosed.

Conception and design: P. Austin, R.J. Andersen, M. Roberge, C.D. Roskelley

Development of methodology: P. Austin, S.A. Freeman

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S.A. Freeman, C.A. Gray, A.W. Vogl, R.J. Andersen

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): P. Austin, S.A. Freeman, C.A. Gray, C.D. Roskelley

Writing, review, and/or revision of the manuscript: P. Austin, R.J. Andersen, M. Roberge, C.D. Roskelley

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.A. Gray

Study supervision: R.J. Andersen, C.D. Roskelley

Collected the electron microscopic images: A.W. Vogl

This research was supported by grant #018344 from the Canadian Cancer Society Research Institute (M. Roberge and C.D. Roskelley) and by funding from the infrastructure program of the Michael Smith Foundation for Health Research to the Solid Tumor Progression Research Unit (C.D. Roskelley, M. Roberge, R.J. Andersen).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Calvo
F
,
Sahai
E
. 
Cell communication networks in cancer invasion
.
Curr Opin Cell Biol
2011
;
23
:
621
9
.
2.
Chua
KN
,
Poon
KL
,
Lim
J
,
Sim
WJ
,
Huang
RY
,
Thiery
JP
. 
Target cell movement in tumor and cardiovascular diseases based on the epithelial-mesenchymal transition concept
.
Adv Drug Deliv Rev
2011
;
63
:
558
67
.
3.
Schumacher
M
,
Kelkel
M
,
Dicato
M
,
Diederich
M
. 
Gold from the sea: marine compounds as inhibitors of the hallmarks of cancer
.
Biotechnol Adv
2011
;
29
:
531
47
.
4.
Thiery
JP
,
Acloque
H
,
Huang
RY
,
Nieto
MA
. 
Epithelial-mesenchymal transitions in development and disease
.
Cell
2009
;
139
:
871
90
.
5.
Zhang
B
,
Halder
SK
,
Zhang
S
,
Datta
PK
. 
Targeting transforming growth factor-beta signaling in liver metastasis of colon cancer
.
Cancer Lett
2009
;
277
:
114
20
.
6.
Koppikar
P
,
Choi
SH
,
Egloff
AM
,
Cai
Q
,
Suzuki
S
,
Freilino
M
, et al
Combined inhibition of c-Src and epidermal growth factor receptor abrogates growth and invasion of head and neck squamous cell carcinoma
.
Clin Cancer Res
2008
;
14
:
4284
91
.
7.
Rosano
L
,
Spinella
F
,
Di Castro
V
,
Nicotra
MR
,
Dedhar
S
,
de Herreros
AG
, et al
Endothelin-1 promotes epithelial-to-mesenchymal transition in human ovarian cancer cells
.
Cancer Res
2005
;
65
:
11649
57
.
8.
Roskelley
CD
,
Williams
DE
,
McHardy
LM
,
Leong
KG
,
Troussard
A
,
Karsan
A
, et al
Inhibition of tumor cell invasion and angiogenesis by motuporamines
.
Cancer Res
2001
;
61
:
6788
94
.
9.
McHardy
LM
,
Sinotte
R
,
Troussard
A
,
Sheldon
C
,
Church
J
,
Williams
DE
, et al
The tumor invasion inhibitor dihydromotuporamine C activates RHO, remodels stress fibers and focal adhesions, and stimulates sodium-proton exchange
.
Cancer Res
2004
;
64
:
1468
74
.
10.
Warabi
K
,
McHardy
LM
,
Matainaho
L
,
Van Soest
R
,
Roskelley
CD
,
Roberge
M
, et al
Strongylophorine-26, a new meroditerpenoid isolated from the marine sponge Petrosia (Strongylophora) corticata that exhibits anti-invasion activity
.
J Nat Prod
2004
;
67
:
1387
9
.
11.
McHardy
LM
,
Warabi
K
,
Andersen
RJ
,
Roskelley
CD
,
Roberge
M
. 
Strongylophorine-26, a Rho-dependent inhibitor of tumor cell invasion that reduces actin stress fibers and induces nonpolarized lamellipodial extensions
.
Mol Cancer Ther
2005
;
4
:
772
8
.
12.
Diaz-Marrero
AR
,
Austin
P
,
Van Soest
R
,
Matainaho
T
,
Roskelley
CD
,
Roberge
M
, et al
Avinosol, a meroterpenoid-nucleoside conjugate with antiinvasion activity isolated from the marine sponge Dysidea sp
.
Org Lett
2006
;
8
:
3749
52
.
13.
Austin
P
,
Heller
M
,
Williams
DE
,
McIntosh
LP
,
Vogl
AW
,
Foster
LJ
, et al
Release of membrane-bound vesicles and inhibition of tumor cell adhesion by the peptide Neopetrosiamide A
.
PLoS One
2010
;
5
:
e10836
.
14.
Kitagawa
I
,
Kobayashi
M
,
Okamoto
Y
,
Yoshikawa
M
,
Hamamoto
Y
. 
Structures of sarasinosides A1, B1, and C1; new norlanostane-triterpenoid oligoglycosides from the Palauan marine sponge Asteropus sarasinosum
.
Chem Pharm Bull (Tokyo)
1987
;
35
:
5036
9
.
15.
Kobayashi
M
,
Okamoto
Y
,
Kitagawa
I
. 
Marine natural products. XXVIII. The structures of sarasinosides A1, A2, A3, B1, B2, B3, C1, C2, and C3, nine new norlanostane-triterpenoidal oligoglycosides from the Palauan marine sponge Asteropus sarasinosum
.
Chem Pharm Bull (Tokyo)
1991
;
39
:
2867
77
.
16.
Schmitz
FJ
,
Ksebati
MB
,
Gunasekera
SP
,
Agarwal
S
. 
Sarasinoside-A1 - a saponin containing amino-sugars isolated from a sponge
.
J Org Chem
1988
;
53
:
5941
7
.
17.
Roskelley
CD
,
Wu
C
,
Somasiri
AM
. 
Analysis of mammary gland morphogenesis
.
Methods Mol Biol
2000
;
136
:
27
38
.
18.
Freeman
SA
,
McLeod
SJ
,
Dukowski
J
,
Austin
P
,
Lee
CC
,
Millen-Martin
B
, et al
Preventing the activation or cycling of the Rap1 GTPase alters adhesion and cytoskeletal dynamics and blocks metastatic melanoma cell extravasation into the lungs
.
Cancer Res
2010
;
70
:
4590
601
.
19.
McLeod
SJ
,
Shum
AJ
,
Lee
RL
,
Takei
F
,
Gold
MR
. 
The Rap GTPases regulate integrin-mediated adhesion, cell spreading, actin polymerization, and Pyk2 tyrosine phosphorylation in B lymphocytes
.
J Biol Chem
2004
;
279
:
12009
19
.
20.
Lee
HS
,
Seo
Y
,
Cho
KW
,
Rho
JR
,
Shin
J
,
Paul
VJ
. 
New triterpenoid saponins from the sponge Melophlus isis
.
J Nat Prod
2000
;
63
:
915
9
.
21.
Roskelley
CD
,
Bissell
MJ
. 
The dominance of the microenvironment in breast and ovarian cancer
.
Semin Cancer Biol
2002
;
12
:
97
104
.
22.
Wong
AS
,
Gumbiner
BM
. 
Adhesion-independent mechanism for suppression of tumor cell invasion by E-cadherin
.
J Cell Biol
2003
;
161
:
1191
203
.
23.
Rikitake
Y
,
Mandai
K
,
Takai
Y
. 
The role of nectins in different types of cell-cell adhesion
.
J Cell Sci
2012
;
125
:
3713
22
.
24.
Fukuyama
T
,
Ogita
H
,
Kawakatsu
T
,
Fukuhara
T
,
Yamada
T
,
Sato
T
, et al
Involvement of the c-Src-Crk-C3G-Rap1 signaling in the nectin-induced activation of Cdc42 and formation of adherens junctions
.
J Biol Chem
2005
;
280
:
815
25
.
25.
Noda
K
,
Zhang
J
,
Fukuhara
S
,
Kunimoto
S
,
Yoshimura
M
,
Mochizuki
N
. 
Vascular endothelial-cadherin stabilizes at cell-cell junctions by anchoring to circumferential actin bundles through alpha- and beta-catenins in cyclic AMP-Epac-Rap1 signal-activated endothelial cells
.
Mol Biol Cell
2010
;
21
:
584
96
.
26.
Olson
MF
,
Sahai
E
. 
The actin cytoskeleton in cancer cell motility
.
Clin Exp Metastasis
2009
;
26
:
273
87
.
27.
Joshi
B
,
Strugnell
SS
,
Goetz
JG
,
Kojic
LD
,
Cox
ME
,
Griffith
OL
, et al
Phosphorylated caveolin-1 regulates Rho/ROCK-dependent focal adhesion dynamics and tumor cell migration and invasion
.
Cancer Res
2008
;
68
:
8210
20
.
28.
Hartsock
A
,
Nelson
WJ
. 
Adherens and tight junctions: structure, function and connections to the actin cytoskeleton
.
Biochim Biophys Acta
2008
;
1778
:
660
9
.
29.
Cleton-Jansen
AM
. 
E-cadherin and loss of heterozygosity at chromosome 16 in breast carcinogenesis: different genetic pathways in ductal and lobular breast cancer?
Breast Cancer Res
2002
;
4
:
5
8
.
30.
Yoder
BJ
,
Wilkinson
EJ
,
Massoll
NA
. 
Molecular and morphologic distinctions between infiltrating ductal and lobular carcinoma of the breast
.
Breast J
2007
;
13
:
172
9
.
31.
Farias
EF
,
Petrie
K
,
Leibovitch
B
,
Murtagh
J
,
Chornet
MB
,
Schenk
T
, et al
Interference with Sin3 function induces epigenetic reprogramming and differentiation in breast cancer cells
.
Proc Natl Acad Sci U S A
2010
;
107
:
11811
6
.
32.
Wheelock
MJ
,
Shintani
Y
,
Maeda
M
,
Fukumoto
Y
,
Johnson
KR
. 
Cadherin switching
.
J Cell Sci
2008
;
121
:
727
35
.
33.
Li
DM
,
Feng
YM
. 
Signaling mechanism of cell adhesion molecules in breast cancer metastasis: potential therapeutic targets
.
Breast Cancer Res Treat
2011
;
128
:
7
21
.
34.
Takai
Y
,
Miyoshi
J
,
Ikeda
W
,
Ogita
H
. 
Nectins and nectin-like molecules: roles in contact inhibition of cell movement and proliferation
.
Nat Rev Mol Cell Biol
2008
;
9
:
603
15
.
35.
Takai
Y
,
Ikeda
W
,
Ogita
H
,
Rikitake
Y
. 
The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin
.
Annu Rev Cell Dev Biol
2008
;
24
:
309
42
.
36.
Deakin
NO
,
Turner
CE
. 
Paxillin comes of age
.
J Cell Sci
2008
;
121
:
2435
44
.
37.
Kiuchi
T
,
Nagai
T
,
Ohashi
K
,
Mizuno
K
. 
Measurements of spatiotemporal changes in G-actin concentration reveal its effect on stimulus-induced actin assembly and lamellipodium extension
.
J Cell Biol
2011
;
193
:
365
80
.
38.
Matsuki-Fukushima
M
,
Hashimoto
S
,
Murakami
M
,
Ogata
Y
,
Fujita-Yoshigaki
J
,
Narita
T
, et al
The actin-specific reagent jasplakinolide induces apoptosis in primary rat parotid acinar cells
.
Arch Oral Biol
2012
;
57
:
567
76
.
39.
Tang
F
,
Wang
D
,
Duan
C
,
Huang
D
,
Wu
Y
,
Chen
Y
, et al
Berberine inhibits metastasis of nasopharyngeal carcinoma 5-8F cells by targeting Rho kinase-mediated Ezrin phosphorylation at threonine 567
.
J Biol Chem
2009
;
284
:
27456
66
.
40.
Harma
V
,
Knuuttila
M
,
Virtanen
J
,
Mirtti
T
,
Kohonen
P
,
Kovanen
P
, et al
Lysophosphatidic acid and sphingosine-1-phosphate promote morphogenesis and block invasion of prostate cancer cells in three-dimensional organotypic models
.
Oncogene
2012
;
31
:
2075
89
.
41.
Jallal
H
,
Valentino
ML
,
Chen
G
,
Boschelli
F
,
Ali
S
,
Rabbani
SA
. 
A Src/Abl kinase inhibitor, SKI-606, blocks breast cancer invasion, growth, and metastasis in vitro and in vivo
.
Cancer Res
2007
;
67
:
1580
8
.
42.
Golas
JM
,
Lucas
J
,
Etienne
C
,
Golas
J
,
Discafani
C
,
Sridharan
L
, et al
SKI-606, a Src/Abl inhibitor with in vivo activity in colon tumor xenograft models
.
Cancer Res
2005
;
65
:
5358
64
.
43.
Vultur
A
,
Buettner
R
,
Kowolik
C
,
Liang
W
,
Smith
D
,
Boschelli
F
, et al
SKI-606 (bosutinib), a novel Src kinase inhibitor, suppresses migration and invasion of human breast cancer cells
.
Mol Cancer Ther
2008
;
7
:
1185
94
.
44.
Li
Y
,
VandenBoom
II TG
,
Kong
D
,
Wang
Z
,
Ali
S
,
Philip
PA
, et al
Upregulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells
.
Cancer Res
2009
;
69
:
6704
12
.