Abstract
Self-renewal maintains the long-term clonogenic growth that is required for cancer relapse and progression, but the cellular processes regulating this property are not fully understood. In many diseases self-renewal is enhanced in cancer stem cells (CSCs), and in pancreatic ductal adenocarcinoma (PDAC) CSCs are characterized by the surface expression of CD44. In addition to cell adhesion, CD44 impacts cell shape and morphology by modulating the actin cytoskeleton via Ezrin, a member of the Ezrin/Radixin/Moesin (ERM) family of linker proteins. We examined the expression of Ezrin in PDAC cells and found higher levels of both total and activated Ezrin in CSCs compared to bulk tumor cells. We also found that the knock-down of Ezrin in PDAC cells decreased clonogenic growth, self-renewal, cell migration, and CSC frequency in vitro as well as tumor initiation in vivo. These effects were associated with cytoskeletal changes that are similar to those occurring during the differentiation of normal stem cells, and the inhibition of actin remodeling reversed the impact of Ezrin loss. Finally, targeting Ezrin using a small molecule inhibitor limited the self-renewal of clinically derived low-passage PDAC xenografts. Our findings demonstrate that Ezrin modulates CSCs properties and may represent a novel target for the treatment of PDAC.
Keywords: Ezrin, pancreas cancer, cancer stem cells, cytoskeleton
Implications
Our findings demonstrate that Ezrin modulates CSCs properties and may represent a novel target for the treatment of PDAC.
Introduction
Cancer stem cells (CSCs) have been identified in many cancers based on their enhanced tumor-initiating and self-renewal potential that implicates an important role in long-term disease maintenance, progression, and relapse (1, 2). Multiple groups have identified CSCs in pancreatic ductal adenocarcinoma (PDAC) that are enriched in migration, invasion, and drug resistance in addition to clonogenic growth and self-renewal (3–5). Importantly, CSCs appear to be clinically relevant as their presence in primary tumors is associated with the development of metastatic disease and shorter overall survival (5). Thus, novel strategies targeting CSCs may improve outcomes in PDAC.
In many solid tumors, including PDAC, CSCs express CD44 and HGFR/c-Met, and the expression of these antigens are associated with poor survival (3, 6–8). Several signaling events occur downstream of CD44, including cytoskeletal remodeling that involves Ezrin, a member of the Ezrin/Radixin/Moesin (ERM) family of membrane-cytoskeletal linker proteins that bridges the cytoplasmic tail of CD44 with F-actin (9–14). Ezrin expression is associated with decreased overall survival and the development of distant metastases in osteosarcoma, soft tissue sarcomas, as well as colorectal, breast, and hepatocellular carcinomas (15–19). In PDAC, Ezrin activation is associated with shorter overall survival (20, 21) and impacts the growth and motility of PDAC cell lines (22, 23). Despite these findings, the role of Ezrin in PDAC CSC biology is unknown.
We found that PDAC CSCs express higher levels of Ezrin relative to bulk tumor cells. The inhibition of Ezrin using knock-down studies or a small molecule antagonist decreased CSC frequency, tumor-initiating potential, and self-renewal. In addition, the effects of Ezrin loss were inhibited by blocking actin polymerization. These results demonstrate that Ezrin impacts stem cell properties in PDAC and may represent a novel target in this disease.
Materials and Methods
Cell culture
MIA PaCa-2 cells were obtained from the American Type Culture Collection (Manassas, VA) and authenticated by short tandem repeat profiling at the Johns Hopkins Genetics Core Resources Facility. L3.6pl cell were a kind gift of J. Fidler (24). E3LZ 10.7 cells have been previously described as Pa03C (25). All cell lines were cultured in complete media consisting of high glucose Dulbecco’s modified Eagle medium (Invitrogen, Carlsbad, CA) supplemented with 10% fetal bovine serum (Sigma-Aldrich, St. Louis, MO), 1% penicillin-streptomycin (Invitrogen), and1% L-glutamine (Invitrogen). Cells were treated with NSC305787 (NCI Developmental Therapeutics Program, Bethesda, MD), Cytochalasin D (Tocris Biosciences, Bristol, United Kingdom), or Y-27632 (Sigma-Aldrich) for 72 hours. Tumor colony formation in methylcellulose was carried out as previously described (5).
Ezrin knock-down and over-expression
Short hairpin RNA (shRNA) oligonucleotides were synthesized based on siRNA sequences against Ezrin (GE Dharmacon, Lafayette, CO, siRNA1 cat. #: D-017370–05-0002, siRNA2 cat. #: D-017370–06-0002) or firefly luciferase (ref – FAK paper?) and cloned into the Tet-pLKO-puro lentiviral vector (Addgene, Cambridge, MA). Following transduction, cells were selected with puromycin (2μg/ml, Thermo Fisher Scientific, Waltham, MA). For over-expression studies, cDNA encoding the Ezrin mutant T567D that contains a threonine to aspartic acid substitution at residue 567 (kindly provided by Dr. Chand Khanna, NIH, Bethesda, MD) was cloned into the pInucer20 lentiviral vector using the Gateway cloning system (Thermo Fisher Scientific) (26). A self-ligated empty pInducer20 construct was used as a control. Cell lines were transduced, and stable lines were selected using G418 sulfate (1.2 mg/ml, Corning).
Animal care, low passage derived patient xenografts, and tumor sphere formation
All animal experiments were approved by the Johns Hopkins University Animal Care and Use Committee. Low passage xenografts were generated from primary surgical specimens collected at the Johns Hopkins Hospital, serially passaged in nude mice, and prepared as single cell suspensions as previously described (5, 27). Tumor spheres enriched in CSCs were generated by culturing cells (200,000 cells/ml) in 60-mm ultralow attachment plates (Corning) and DMEM/F-12 (1/1) media (Invitrogen) supplemented with 1X B-27 supplement (Thermo Fisher Scientific), 20ng/ml bFGF (Invitrogen), 1% penicillin-streptomycin and 1% L-glutamine. Tumor cell differentiation was induced by culturing cells in complete media containing 10% FBS.
Tumor-initiating cell (TIC) frequency was determined by injecting L3.6pl cells (100,000) in serum-free DMEM and Matrigel (BD Biosciences) subcutaneously into the flanks of NSG mice. Following tumor formation, mice were provided doxycycline (2mg/ml, Sigma-Aldrich) in 5% sucrose. Following 9 days of treatment, tumors were collected, dissociated, and depleted of mouse cells then injected (50, 100, 250, and 500 cells) into secondary recipients. Tumor formation was monitored for 30 days, and (TIC) frequency was calculated using extreme limiting dilution analysis (ELDA) (28).
Flow cytometry
Cells were stained for surface antigen expression with anti-CD44-APC (BD Biosciences), anti-CD24-PE/FITC (BD Biosciences), anti-c-Met-PE (R&D Systems, Minneapolis, MN), and anti CD133-APC (Milteniy Biotec). Cells were analyzed using a BD FACScalibur (BD Biosciences) flow cytometer to identify CSC populations (8, 29, 30). Intracellular staining, was carried out using the Cytofix/Cytoperm kit (BD Biosciences) and anti-Ezrin (Cell signaling, Danvers, MA) or anti-pERM (Cell signaling) antibodies, goat anti-rabbit-AlexaFluor488 (Invitrogen), phalloidin-TRITC/FITC (Sigma-Aldrich), or DNAse1-AlexaFluor488 (Sigma-Aldrich).
Quantitative real-time PCR
Total mRNA was extracted and depleted of genomic DNA using the RNeasy PlusMini Kit (Qiagen, Germantown, MD) and reverse-transcribed with SuperScript III reverse transcriptase (Invitrogen). qRT-PCR was performed using Taqman Probes (GAPDH probe #: hs099999905, Ezrin probe #:hs00931653) (Applied Biosystems, Foster City, CA).
Cell migration assay
Cells were cultured in 24-well inserts with 8-μm pores (BD Biosciences) for 24 hours. Cells migrating through the filters were stained with DAPI followed by quantification using a Nikon Eclipse Ti fluorescent microscope (Nikon, Minato, Tokyo, Japan) and the NIS Elements software as previously described (5).
Immunofluorescence microscopy
Cells were grown on LAB-Tek II glass slides (Thermo Fisher Scientific) or cytospun on Shendon glass slides then fixed with 4% Formalin, permeabilized with 0.3% Triton-X (Sigma-Aldrich), and stained with phalloidin-TRITC and DNAse1-Alexafluor488, and DAPI. Slides were viewed using Nikon EZ-C1 confocal microscope (Nikon) and images processed with the NIS Elements software.
Immunoprecipitation and western blotting
Whole cell lysates were prepared and cleared as previously described (31). Immunoprecipitation was performed with anti-Ezrin antibody and recombinant protein A agarose beads (Thermo Scientific) then separated using NuPAGE Novex 4–12% Bis-Tris Protein Gels (Life technologies) and transferred to PVDF membranes. Protein detection was carried out using rabbit polyclonal anti-Ezrin, rabbit polyclonal anti-pERM (Cell Signaling Technology), anti-Actin, or anti-GAPDH (Abcam) antibodies. Densitometry was carried out using a GS-900 densitometer and Image Lab software (Bio-Rad, Hercules, Ca).
Gene expression profiling
RNA was isolated using the RNeasy mini kit from L3.6pl cells with Ezrin or control shRNA following treatment with doxycycline for 72hrs. Gene expression was quantified using Illumina HT12_4 microarrays, and gene set enrichment analysis (GESA) was performed with JAVA-based GSEA software.
Statistical analysis
Statistical differences between non-normalized groups were analyzed using the two-tailed unpaired Student’s t test, and between normalized groups using the one sample t test (GraphPad Prism Software). Statistical differences in tumor initiation frequency between groups were analyzed using the ELDA software. P values <0.05 were considered significant.
Results
Ezrin is overexpressed by PDAC CSCs
Ezrin is expressed in PDAC (20, 21), and we studied its expression in CSCs. Using flow cytometry, we examined PDAC cells from low-passage human xenografts derived from primary clinical specimens and found that the CD24hi/CD44hi, CD44hi/c-Methi, and CD44hi CSC populations all expressed significantly higher levels of Ezrin compared to bulk tumor cells (Fig. 1A, Supplementary Fig. S1A)(3, 8, 27). Similarly, in the L3.6pl cell line Ezrin was expressed at higher levels in CSCs compared to non-CSCs (Fig. 1B). Following activation, Ezrin binds to actin (32–35), and we quantified the co-precipitation of actin with Ezrin in CSCs and differentiated PDAC cells. For these studies, we examined low-passage xenografts and enriched for CSCs by culturing as tumor spheres in serum-free media on low attachment plates and differentiated cells by exposing spheres to serum (3) (Fig. 1C). We found that Ezrin activation was relatively higher in CSC-enriched tumor spheres compared to differentiated cells as evidenced by increased actin binding to Ezrin and expression of phosphorylated ERM proteins (Figs. 1D, 1E; Supplementary Fig. 1B). Therefore, Ezrin expression and activation are enhanced in PDAC CSCs.
Ezrin regulates CSC functions
To determine the functional role of Ezrin in PDAC, we inhibited its expression in PDAC cell lines using doxycycline-inducible shRNA constructs (Supplementary Fig. 2A). Clonogenic growth and self-renewal are hallmarks of CSCs, and we found that the inhibition of Ezrin expression for 7 days in L3.6pl cells significantly decreased subsequent tumor colony formation by >90% (Fig. 2A). Furthermore, this loss of clonogenic potential was not due to the induction of cell death or changes in proliferation (Fig. 2B; Supplementary Fig. S2B). To quantify self-renewal, we harvested and serially re-plated tumor colonies and found that the inhibition of clonogenic growth was maintained by L3.6pl cells containing the Ezrin shRNA vector despite the lack of doxycycline treatment during either the primary or secondary plating (Fig. 2A). Similarly, loss of Ezrin expression inhibited the formation of tumor spheres (Supplementary Fig. S2C). This loss of clonogenic growth was associated with a significant decrease in the frequency of CD24+/CD44+ (1.8% vs. 0.6 %,) and CD44+cMet+ (41% vs. 15%) CSCs (Fig. 2C). Similarly, clonogenic growth and self-renewal were significantly inhibited in E3LZ10.7 and MIA PaCa-2 cells (Supplementary Figs. S2D, S2E).
In addition to enhanced clonogenic growth, CSCs in many solid tumors have enhanced migratory capabilities that implicate a role in metastasis, and CSCs are increased in PDAC metastatic lesions (2, 5). Ezrin regulates cell migration in several malignancies (18, 19), and we found that the loss of Ezrin expression significantly inhibited the migration of L3.6pl cells by >99% (Fig. 2D) consistent with other reports (22, 23). We also carried out gain-of-function studies and over-expressed the constitutively active Ezrin mutant T567D in L3.6pl cells and observed significantly increased colony formation (1.9 fold) and cell migration (2.2 fold) compared to cells transduced with an empty vector (Fig. 2E; Supplementary Fig. S2F). Therefore, Ezrin modulates clonogenic growth, cell migration, and the frequency of phenotypically defined PDAC CSCs.
Ezrin regulates actin dynamics in PDAC cells
Ezrin links the cytoplasmic tail of CD44 to F-actin leading to cytoskeletal remodeling (14), and changes in actin cytoskeletal dynamics and cell shape can direct stem cell differentiation (36–38). For example, in normal keratinocytes, self-renewal is dependent on the maintenance of a lower ratio of polymerized (F-actin) to free actin (G-actin) (36). Similarly, the inhibition of actin polymerization maintains embryonic stem cells in an undifferentiated state without the need for Leukemia Inhibiting Factor (37). Therefore, we examined whether Ezrin regulates the actin cytoskeleton in PDAC. In L3.6pl cells, the loss of Ezrin increased cortical F-actin organization, decreased nuclear G-actin staining, and induced a flattened morphology (Fig 3A) similar to the changes seen during stem cell differentiation in normal systems (36–38). Moreover, we quantified F-actin and G-actin by flow cytometry and western blotting and observed that Ezrin knock-down significantly increased the F/G actin ratio, similar to the differentiation of epithelial stem cells (Fig. 3B; Supplementary Fig. S3A) (36).
We also examined cell morphology and actin dynamics in tumor cells from low-passage PDAC xenografts and found that CSC-enriched tumor spheres were similar to control L3.6pl cells in both their morphology and actin organization (Supplementary Fig. S3B). Following serum-induced differentiation, the morphologic changes and increase in the F/G actin ratio (2.7 in CSCs vs 4.3 in differentiated cells) were similar to L3.6pl cells following Ezrin knock-down (Supplementary Fig. S3C). We also compared CD24hi/CD44hi CSCs and bulk tumor cells directly isolated from low-passage PDAC xenografts and found that they significantly exhibited a lower F/G actin ratio than bulk cells in two of three tumors (Supplementary Fig. S3D). Therefore, PDAC CSCs are associated with specific actin cytoskeletal changes that can be modulated by Ezrin.
To determine whether Ezrin maintains clonogenic growth by modulating actin dynamics, we treated L3.6pl cells with Y-27632, a small molecule that indirectly decreases actin polymerization by blocking the activity of Rho-associated kinase (ROCK) (39, 40). Following Ezrin knock-down, treatment with Y-27632 significantly inhibited the loss of clonogenic growth and restored CSC-associated actin features (Fig. 3C; Supplementary Fig. S4A). In addition, we found that the inhibition of actin polymerization with Y-27632 or Cytochalasin D significantly increased the clonogenic growth of L3.6pl cells without impacting cell growth (Supplementary Fig. S4B, S4C)(39, 40). In contrast, Y-27632 treatment did not impact the clonogenic recover of L3.6pl cells expressing the constitutively active Ezrin mutant T567D (Supplementary Fig. S4D) suggesting that the modulation of ROCK activity and subsequent inhibition of actin polymerization maintains CSCs.
ROCK can block the differentiation of mesenchymal stem cells to chondrocytes by inhibiting Sox9 expression and activity (41). Since Sox9 is also required for pancreatic development (42, 43), we studied the impact of Sox9 on Ezrin function in PDAC. We found that Ezrin knock-down resulted in a significant loss of Sox9 expression in L3.6pl cells (Supplementary Fig. S4E). However, Sox9 overexpression did not inhibit the effects of Ezrin loss on clonogenic growth (Supplementary Fig. S4F). In order to better understand the impact of Ezrin loss in L6.3pl cells, we carried out gene expression profiling studies and found that the loss of Ezrin expression impacted gene sets associated with the epithelial to mesenchymal transition and active c-Met signaling that have been associated with CSCs (Supplementary Fig. S4G) (8, 44). Therefore, Ezrin maintains CSCs, at least in part, by regulating actin polymerization through ROCK inhibition, but independently of Sox9.
Pharmacological Ezrin inhibition decreases PDAC self-renewal
We recently we identified several small molecules that inhibit the phosphorylation of Ezrin at T567 and its interaction with actin (45). These compounds inhibited Ezrin-mediated invasion and survival of osteosarcoma cells in vitro and lung metastasis in vivo. We studied one of these compounds, NSC305787, in PDAC cells and found that it significantly decreased colony formation in L3.6pl (28%), E3LZ 10.7 (24%), and MIA PaCa-2 cells (57%) without inhibiting cell viability as well as inhibited the binding of Ezrin to actin (Fig. 4A; Supplementary Fig. S5). We also treated tumor spheres from low-passage PDAC xenografts with NSC305787 and found it significantly inhibited subsequent sphere formation by 17–40% in Panc265, Panc185, and JH102 tumors (Fig. 4B) (27). Therefore, the pharmacological inhibition of Ezrin may represent a novel strategy to target pancreatic CSCs
Loss of Ezrin expression limits in vivo self-renewal
To extend our in vitro findings, we examined the impact of Ezrin loss on tumor initiation in vivo. We injected L3.6pl cells with the inducible Ezrin shRNA construct subcutaneously into immunodeficient NSG mice, and following tumor formation, administered doxycycline for 9 days (Fig. 5A; Supplementary Fig. 6A). Doxycycline treatment did not significantly impact tumor growth, although the loss of Ezrin expression resulted in histological changes with clusters of differentiated epithelial cells as well as the inhibition of in vitro clonogenic growth (Figs. 5B, 5C; Supplementary Fig. S6B). We then collected tumors and re-transplanted cells in limiting dilution into naïve recipients. In the absence of additional doxycycline treatment of the recipient mice, the tumor initiating cell frequency during secondary engraftment of cells carrying the Ezrin shRNA construct was significantly lower than cells with the control shRNA (Table 1). Thus, transient Ezrin knock-down impairs long-term clonogenic growth and self-renewal in vivo.
Table 1.
1000 cells | 500 cells | 250 cells | 100 cells | p-value | |
---|---|---|---|---|---|
shControl | 8/8 | 12/12 | 12/12 | 4/4 | |
EZRsh1 | 7/8 | 11/12 | 5/12 | 2/4 | 2.1*10^-6 |
Discussion
We found that PDAC CSCs are characterized by increased Ezrin expression and activation. The induction of Ezrin activity increases the colony-forming ability PDAC cells, whereas the loss of Ezrin expression or activity markedly decreases migration and self-renewal in vitro and tumor initiation in vivo. We also found that a small molecule inhibitor of Ezrin decreased the self-renewal of clinically derived PDAC samples. Therefore, Ezrin may represent a novel means of inhibiting CSCs.
We observed a decreased polymerized to free actin ratio in CSCs compared to differentiated cells, and these findings suggest that actin polymerization impacts CSC biology. In keratinocytes, the balance between free and polymerized actin can influence cell fate decisions and specific gene expression by regulating the availability of transcriptional co-factors (36). Although the precise mechanisms responsible for the relationship between the cytoskeleton and stem cell is unclear, it is also possible that CSCs self–renew through asymmetrical cell division, and specific cytoskeletal changes could regulate the balance between symmetric and asymmetric divisions by controlling cellular polarity (46). Alternatively, actin dynamics could serve to organize different membrane domains and modulate activity of various cell surface receptors (47, 48).
Cytoskeletal dynamics might also play a role in the interaction between tumor cells and the surrounding stroma. The deposition of extracellular matrix proteins can generate a large compressive force, and it is possible that these forces impact cytoskeletal organization leading to changes in stem cell properties. Such effects are seen during skeletal development where compressive forces lead to differentiation of progenitor cells into cartilage (49, 50). In PDAC models, the loss of stromal elements leads to disease acceleration and increased tumor-initiating cell frequency, suggesting that pressure from stroma might limit CSC self-renewal (51, 52). Thus, the maintenance of a specific actin filament organization by CSCs by restrictive pressure arising from the stroma may contribute to CSC self-renewal.
In PDAC, distinct populations of CSCs have been identified, but targeting these cells using strategies based on the expression of specific surface antigens may be limited by differential expression on some, but not all, CSCs (2). If the regulation of cytoskeltal rearrangements is necessary for the maintenance of CSCs regardless of their phenyotype, then targeting actin dynamics could lead to a successful elimination of all sources of self-renewal in the tumor. Notably, we found that Ezrin was expressed in multiple, phenotypically defined CSCs. A further examination of the potential mechanisms of CSC regulation by the cytoskeleton may yield more targets for therapeutic intervention.
Small molecules that directly bind to actin and affect its polymerization have been widely used as experimental tools, including the study of normal stem cells (36, 38). However, these compounds lack specificity to a particular type of actin, and their potential effects on cardiac and skeletal muscle actin fibers limits their clinical potential (39). Thus, pharmacological strategies to inhibit Ezrin may be a relatively safer way to modulate actin dynamics in cancer cells. We demonstrated the utility of Ezrin inhibitors in decreasing the self-renewal of PDAC cells. Since Ezrin is overexpressed in multiple tumor types but typically not in the corresponding normal tissue of origin (15–19), it may be a promising target for pharmacological intervention.
Supplementary Material
Acknowledgments
The work was supported by NIH grants RO1CA150142 and R01 CA193887 (WM)
Footnotes
No potential conflicts of interest
References
- 1.Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645–8. [DOI] [PubMed] [Google Scholar]
- 2.Penchev VR, Rasheed ZA, Maitra A, Matsui W. Heterogeneity and targeting of pancreatic cancer stem cells. Clinical cancer research : an official journal of the American Association for Cancer Research. 2012;18:4277–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identification of pancreatic cancer stem cells. Cancer research. 2007;67:1030–7. [DOI] [PubMed] [Google Scholar]
- 4.Hermann PC, Huber SL, Herrler T, Aicher A, Ellwart JW, Guba M, et al. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell stem cell. 2007;1:313–23. [DOI] [PubMed] [Google Scholar]
- 5.Rasheed ZA, Yang J, Wang Q, Kowalski J, Freed I, Murter C, et al. Prognostic significance of tumorigenic cells with mesenchymal features in pancreatic adenocarcinoma. Journal of the National Cancer Institute. 2010;102:340–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100:3983–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Dalerba P, Dylla SJ, Park I-K, Liu R, Wang X, Cho RW, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci USA. 2007;104:10158–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Li C, Wu JÄ, Hynes M, Dosch J, Sarkar B, Welling TH, et al. c-Met Is a Marker of Pancreatic Cancer Stem Cells and Therapeutic Target. Gastroenterology. 2011;141:2218–27.e5. [DOI] [PubMed] [Google Scholar]
- 9.Tsukita S, Oishi K, Sato N, Sagara J, Kawai A, Tsukita S. ERM family members as molecular linkers between the cell surface glycoprotein CD44 and actin-based cytoskeletons. The Journal of cell biology. 1994;126:391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wakayama Y, Miura K, Sabe H, Mochizuki N. EphrinA1-EphA2 signal induces compaction and polarization of Madin-Darby canine kidney cells by inactivating Ezrin through negative regulation of RhoA. The Journal of biological chemistry. 2011;286:44243–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Parisiadou L, Xie C, Cho HJ, Lin X, Gu XL, Long CX, et al. Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2009;29:13971–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Manchanda N, Lyubimova A, Ho HY, James MF, Gusella JF, Ramesh N, et al. The NF2 tumor suppressor Merlin and the ERM proteins interact with N-WASP and regulate its actin polymerization function. The Journal of biological chemistry. 2005;280:12517–22. [DOI] [PubMed] [Google Scholar]
- 13.Orian-Rousseau V, Chen L, Sleeman JP, Herrlich P, Ponta H. CD44 is required for two consecutive steps in HGF/c-Met signaling. Genes & development. 2002;16:3074–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Orian-Rousseau V, Morrison H, Matzke A, Kastilan T, Pace G, Herrlich P, et al. Hepatocyte growth factor-induced Ras activation requires ERM proteins linked to both CD44v6 and F-actin. Molecular biology of the cell. 2007;18:76–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kang YK, Hong SW, Lee H, Kim WH. Prognostic implications of ezrin expression in human hepatocellular carcinoma. Molecular carcinogenesis. 2010;49:798–804. [DOI] [PubMed] [Google Scholar]
- 16.Elzagheid A, Korkeila E, Bendardaf R, Buhmeida A, Heikkila S, Vaheri A, et al. Intense cytoplasmic ezrin immunoreactivity predicts poor survival in colorectal cancer. Human pathology. 2008;39:1737–43. [DOI] [PubMed] [Google Scholar]
- 17.Weng WH, Ahlen J, Astrom K, Lui WO, Larsson C. Prognostic impact of immunohistochemical expression of ezrin in highly malignant soft tissue sarcomas. Clinical cancer research : an official journal of the American Association for Cancer Research. 2005;11:6198–204. [DOI] [PubMed] [Google Scholar]
- 18.Elliott BE, Meens JA, SenGupta SK, Louvard D, Arpin M. The membrane cytoskeletal crosslinker ezrin is required for metastasis of breast carcinoma cells. Breast cancer research : BCR. 2005;7:R365–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Khanna C, Wan X, Bose S, Cassaday R, Olomu O, Mendoza A, et al. The membrane-cytoskeleton linker ezrin is necessary for osteosarcoma metastasis. Nature medicine. 2004;10:182–6. [DOI] [PubMed] [Google Scholar]
- 20.Cui Y, Li T, Zhang D, Han J. Expression of Ezrin and phosphorylated Ezrin (pEzrin) in pancreatic ductal adenocarcinoma. Cancer investigation. 2010;28:242–7. [DOI] [PubMed] [Google Scholar]
- 21.Oda Y, Aishima S, Morimatsu K, Hayashi A, Shindo K, Fujino M, et al. Differential ezrin and phosphorylated ezrin expression profiles between pancreatic intraepithelial neoplasia, intraductal papillary mucinous neoplasm, and invasive ductal carcinoma of the pancreas. Human pathology. 2013;44:1487–98. [DOI] [PubMed] [Google Scholar]
- 22.Zhong ZQ, Song MM, He Y, Cheng S, Yuan HS. Knockdown of Ezrin by RNA interference reverses malignant behavior of human pancreatic cancer cells in vitro. Asian Pacific journal of cancer prevention : APJCP. 2012;13:3781–9. [DOI] [PubMed] [Google Scholar]
- 23.Meng Y, Lu Z, Yu S, Zhang Q, Ma Y, Chen J. Ezrin promotes invasion and metastasis of pancreatic cancer cells. Journal of translational medicine. 2010;8:61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bruns CJ, Harbison MT, Kuniyasu H, Eue I, Fidler IJ. In Vivo Selection and Characterization of Metastatic Variants from Human Pancreatic Adenocarcinoma by Using Orthotopic Implantation in Nude Mice. Neoplasia. 1999;1:50–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Jones S, Zhang X, Parsons DW, Lin JC-H, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Meerbrey KL, Hu G, Kessler JD, Roarty K, Li MZ, Fang JE, et al. The pINDUCER lentiviral toolkit for inducible RNA interference in vitro and in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:3665–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hu Y, Smyth GK. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. Journal of immunological methods. 2009;347:70–8. [DOI] [PubMed] [Google Scholar]
- 29.Ishizawa K, Rasheed ZA, Karisch R, Wang Q, Kowalski J, Susky E, et al. Tumor-initiating cells are rare in many human tumors. Cell stem cell. 2010;7:279–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Li C, Heidt DG, Dalerba P, Burant CF, Zhang L, Adsay V, et al. Identification of pancreatic cancer stem cells. Cancer Res. 2007;67:1030–7. [DOI] [PubMed] [Google Scholar]
- 31.Begum A, Ewachiw T, Jung C, Huang A, Norberg KJ, Marchionni L, et al. The extracellular matrix and focal adhesion kinase signaling regulate cancer stem cell function in pancreatic ductal adenocarcinoma. PloS one. 2017;12:e0180181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Turunen O, Wahlström T, Vaheri A. Ezrin has a COOH-terminal actin-binding site that is conserved in the ezrin protein family. The Journal of Cell Biology. 1994;126:1445–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yao X, Cheng L, Forte JG. Biochemical Characterization of Ezrin-Actin Interaction. J Biol Chem. 1996;271:7224–9. [DOI] [PubMed] [Google Scholar]
- 34.Gungor-Ordueri NE, Tang EI, Celik-Ozenci C, Cheng CY. Ezrin is an Actin Binding Protein That Regulates Sertoli Cell and Spermatid Adhesion During Spermatogenesis. Endocrinology. 2014;155:3981–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bosk S, Braunger Julia A, Gerke V, Steinem C Activation of F-Actin Binding Capacity of Ezrin: Synergism of PIP2 Interaction and Phosphorylation. Biophys J. 2011;100:1708–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Connelly JT, Gautrot JE, Trappmann B, Tan DW, Donati G, Huck WT, et al. Actin and serum response factor transduce physical cues from the microenvironment to regulate epidermal stem cell fate decisions. Nature cell biology. 2010;12:711–8. [DOI] [PubMed] [Google Scholar]
- 37.Murray P, Prewitz M, Hopp I, Wells N, Zhang H, Cooper A, et al. The self-renewal of mouse embryonic stem cells is regulated by cell-substratum adhesion and cell spreading. The international journal of biochemistry & cell biology. 2013;45:2698–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rowena McBeath DMP, Celeste M. Nelson KB, Chen aCS. Cell Shape, Cytoskeletal Tension, and RhoA Regulate Stem Cell Lineage Commitment. Developmental Cell. 2004;6. [DOI] [PubMed] [Google Scholar]
- 39.Bonello TT, Stehn JR, Gunning PW. New approaches to targeting the actin cytoskeleton for chemotherapy. Future medicinal chemistry. 2009;1:1311–31. [DOI] [PubMed] [Google Scholar]
- 40.Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, et al. Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science. 1999;285:895–8. [DOI] [PubMed] [Google Scholar]
- 41.Woods A, Wang G, Beier F. RhoA/ROCK signaling regulates Sox9 expression and actin organization during chondrogenesis. The Journal of biological chemistry. 2005;280:11626–34. [DOI] [PubMed] [Google Scholar]
- 42.Matsumoto Y, Inden M, Tamura A, Hatano R, Tsukita S, Asano S. Ezrin mediates neuritogenesis via down-regulation of RhoA activity in cultured cortical neurons. PLoS ONE. 2014;9:e105435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Seymour PA, Freude KK, Tran MN, Mayes EE, Jensen J, Kist R, et al. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:1865–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Mani S, Evans K, Hollier B, Guo W, Weinberg R. Generation of Stem-like Cells via EMT: A New Twist in Cancer Initiation and Progression. AACR Education Book. 2009;2009:173. [Google Scholar]
- 45.Bulut G, Hong SH, Chen K, Beauchamp EM, Rahim S, Kosturko GW, et al. Small molecule inhibitors of ezrin inhibit the invasive phenotype of osteosarcoma cells. Oncogene. 2012;31:269–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dard N, Louvet-Vallee S, Santa-Maria A, Maro B. Phosphorylation of ezrin on threonine T567 plays a crucial role during compaction in the mouse early embryo. Developmental biology. 2004;271:87–97. [DOI] [PubMed] [Google Scholar]
- 47.Donatello S, Babina IS, Hazelwood LD, Hill AD, Nabi IR, Hopkins AM. Lipid raft association restricts CD44-ezrin interaction and promotion of breast cancer cell migration. The American journal of pathology. 2012;181:2172–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yakovich AJ, Huang Q, Du J, Jiang B, Barnard JA. Vectorial TGFbeta signaling in polarized intestinal epithelial cells. Journal of cellular physiology. 2010;224:398–404. [DOI] [PubMed] [Google Scholar]
- 49.Rooney P, Archer CW. The development of the perichondrium in the avian ulna. Journal of anatomy. 1992;181 (Pt 3):393–401. [PMC free article] [PubMed] [Google Scholar]
- 50.Long F, Linsenmayer TF. Regulation of growth region cartilage proliferation and differentiation by perichondrium. Development. 1998;125:1067–73. [DOI] [PubMed] [Google Scholar]
- 51.Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer cell. 2014;25:719–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer cell. 2014;25:735–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.