Next Article in Journal / Special Issue
SARS-CoV-2 Mpro Inhibitors: Achieved Diversity, Developing Resistance and Future Strategies
Previous Article in Journal
Three-Dimensional Cell Culture Methods in Infectious Diseases and Vaccine Research
Previous Article in Special Issue
Persistence of Second and Third-Line Biologics in Inflammatory Bowel Disease: A Multi-Centre Cohort Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Perspectives for Targeting Ezrin in Cancer Development and Progression

Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
*
Author to whom correspondence should be addressed.
Future Pharmacol. 2023, 3(1), 61-79; https://doi.org/10.3390/futurepharmacol3010005
Submission received: 23 November 2022 / Revised: 30 December 2022 / Accepted: 4 January 2023 / Published: 9 January 2023
(This article belongs to the Special Issue Feature Papers in Future Pharmacology)

Abstract

:
Recent advances have been made in understanding molecular markers involved in cancer malignancy, resulting in better tumor staging and identifying new potential therapeutic targets. Ezrin (EZR), a member of the ezrin, radixin, moesin (ERM) protein family, is essential for linking the actin cytoskeleton to the cell membrane and participates in the signal transduction of key signaling pathways such as Rho GTPases and PI3K/AKT/mTOR. Clinical and preclinical studies in a wide variety of solid and hematological tumors indicate that (i) EZR is highly expressed and predicts an unfavorable clinical outcome, and (ii) EZR inhibition reduces proliferation, migration, and invasion in experimental models. The development of pharmacological inhibitors for EZR (or the signaling mediated by it) has opened a new round of investigation, but studies are still limited. The scope of the present review is to survey studies on the expression and clinical impact of EZR in cancer, as well as studies that perform interventions on the function of this gene/protein in cancer cells, providing proof-of-concept of its antineoplastic potential.

Graphical Abstract

1. Introduction

Ezrin (EZR) is a protein member of the ezrin, radixin, moesin (ERM) family, and its function, when phosphorylated, is essential for linking the actin cytoskeleton to the cell membrane. The FERM domain is composed of three structural modules (F1, F2, and F3), which together form a compact clover-shaped structure and bind to integral membrane proteins, adhesion molecules, multidrug resistance proteins, scaffold proteins, Rho-related proteins, and tyrosine kinase proteins, and are involved in epithelial organization, villus morphogenesis, and PIP2 signaling. EZR plays an essential role as an activator of notable signal transduction pathways involved in cancer progression, such as PI3K/AKT/mTOR signaling, which provides a mechanism to anchor PI3K in the proximity of its substrate recruiting the p85 regulatory subunit. In contrast, the phosphorylation of EZR at Y353 is also essential to activate the PI3K signaling [1,2,3,4,5]. The activation of AKT protects cells from apoptosis by phosphorylating and inactivating BAD, a proapoptotic member of the BCL2 family, and increasing cell proliferation [6]. Furthermore, ERM phosphorylation, which appears to be regulated positively by Rho and (possibly) negatively by RAC, may activate downstream signaling of Rho proteins (including RAC) that are required for membrane ruffling and lamellipodium extension and CDC42 that induces the formation of filopodia; both GTPases are essential for cell migration and invasion [7,8] (Figure 1). Thus, EZR may serve as a proliferation- and metastasis-related oncogene by modulating multiple cellular processes [9]. EZR is widely expressed in normal tissues, but its expression is time-specific during human development. For example, EZR immunoreactivity is positive in early embryo stages but undetectable during later development and postnatal stages [10]. Recently, reports have shown that EZR is upregulated in several human tumors, as described in the following.
The EZR C-terminal domain, especially 34 amino acid residues, is highly conserved among ERM proteins and binds to the actin cytoskeleton. A coiled-coil structure connects the N-terminal FERM domain and the C-terminal actin-binding domain. The primary protein structure of EZR and its inactive and active conformations are illustrated in Figure 2.

2. EZR Activation

In the closed inactive state, the FERM domain is tightly associated with the ~80 residues of the C-terminal domain (CTD) from EZR, hiding the membrane association and F-actin-binding sites, and the change from closed to open ERMs requires phosphorylation of a specific threonine residue (T567 in ezrin, T564 in radixin, and T558 in moesin) [12]. Activation of EZR has been proposed to follow a two-step mechanism. The first activation occurs via phosphatidylinositol-4,5-bisphosphate (PIP2) binding at the membrane, which seems to facilitate the binding of EZR to membrane proteins. In other words, PIP2 may activate the dormant protein and expose the membrane binding site. Through this PIP2 binding, T567 in the CTD-ERM association domain becomes accessible for phosphorylation by ROCK, LOK, SLK, and some PKC isoforms [13,14,15,16].

3. Genomics of EZR

The entire EZR gene is approximately 53.6 Kb (start: 158765741 and end: 158819368bp; orientation: reverse strand). In the NCBI database (https://www.ncbi.nlm.nih.gov/gene, accessed on 2 August 2021), there are two transcript variants for EZR that encode for the same protein (586 aa). Transcript variant 1 (transcript length: 3069 bp) represents the longer transcript, while transcript variant 2 (transcript length: 3052 bp) differs from transcript variant 1 in the 5′ UTR region. In the Ensembl database (http://www.ensembl.org/, accessed on 2 August 2021), there is one additional transcript variant for EZR (ENST00000392177.8: 2933 bp), which also encodes for the same protein (586 aa).
Recurrent mutations in the EZR gene are rare, and most are variants of uncertain significance. Using The Cancer Genome Atlas (TCGA) cohorts (10,953 patients in 32 studies), a total of 192 patients (2%) presented EZR genetic alterations (mutations, amplifications, deep deletions, and multiple alterations), as reported on cBioPortal (http://www.cbioportal.org, accessed on 10 October 2022) (Figure 3A). A total of 110 somatic mutations were found: 84 missense substitutions, 16 truncating, one splice mutation, and nine gene fusions (Figure 3B). Among these mutations, two alterations stand out for having driver mutation potential: EZR::ROS1 and ARID1B::EZR gene fusions.

4. Expression and Functions for EZR in Human Cancers

4.1. Breast Cancer

EZR is upregulated in breast cancer tissues compared with normal breast tissues, especially in metastatic breast cancer, and also may be used as a biomarker in overall survival (OS) [17]. Indeed, it had been demonstrated that EZR was required for the invasion and metastasis of mammary carcinoma cells [18]. In metastatic breast cancer cells, MDA-MB-231, EZR silencing by shRNA reduced cell motility and invasion, c-Src signaling, and increased E-cadherin expression, a vital component of the adherent junctions [19]. EZR interacts with AKT in breast cancer cells, induces its downstream signaling pathway, and promotes tumor progression [9]. PI3K and proto-oncogene c-Src activities were modulated by EZR and are required for EZR-dependent cell invasion [18]. The co-expression of EZR with CD44, a glycoprotein involved in cell–cell interactions, adhesion, and migration, may also be a potential biomarker for the initiation, progression, and differentiation of breast cancer tumors [20].

4.2. Melanoma

Ilmonen et al. [21] showed that EZR expression was found in 76 out of 95 melanoma samples analyzed (80%), with 48 weakly positive and 28 strongly positive, as evidenced by an immunohistochemistry assay. Notably, EZR immunoreactivity of metastatic tumors was more substantial than in primary tumors, and none of the metastasis samples were negative for EZR expression (two samples were weakly positive, and ten were strongly positive). Additionally, EZR expression was associated with tumor proliferation index and tumor growth in primary melanoma [21]. Federici et al. [22] aimed to correlate EZRs molecular interactions with major actors of the metastatic behavior of tumor cells (i.e., CD44) and showed that an EZR deletion mutant (146 N-terminal amino acids) abolished the in vivo metastatic dissemination of a melanoma model. In melanoma cells, EZR expression was also associated with phagocytic machinery and phagocytosis through actin cytoskeleton modulation [23]. Interestingly, miR-183 may serve as a tumor suppressor since it reduces migratory activity and invasiveness of A375 human melanoma cells. In this context, EZR was identified as a target for miR-183 [24].

4.3. Cervical Carcinoma

EZR protein was highly expressed in all cervical cancer cell lines and tissues compared with the normal cervical tissues and predicted poor prognosis in cervical cancer [25,26]. In HPV-associated squamous intraepithelial lesions, EZR expression was associated with the severity of the disease. Intense staining for EZR with diffuse intracellular distribution was also observed in in situ adenocarcinoma. In contrast, the distribution in normal endocervical tissue was strongly polarized to the apical side of columnar epithelial cells [27]. In a previous study, the authors observed slightly reduced EZR expression due to HPV 16 E5, an oncogene facilitating the early events in the neoplastic development, suggesting that increased expression of EZR would rather be a consequence of E6 and/or E7 oncogene expression [28]. In HeLa and SiHa cells, EZR silencing by siRNA reduced colony formation and cervical cancer cells ability to cross the chorioallantoic membrane surface and infiltrate the underlying stroma, consequently attenuating migration, invasion, mesenchymal marker expression, and PI3K/AKT activation [25].

4.4. Colorectal Cancer

Through tissue microarray and immunohistochemistry, Patara et al. [29] concluded that a higher cytoplasmatic EZR expression correlates with higher tumor aggressiveness and poor prognosis. Moreover, its expression is an important prognostic factor in colorectal cancer patients. One of the major causes of poor prognosis in colorectal cancer is lymph node metastasis initiated by epithelial–mesenchymal transition (EMT) due to the activation of several signaling pathways by EZR, which plays a significant role in protein signal transmission. Indeed, many growth factors and cytokines have been proven essential in stimulating EMT, including EGF. Li et al. [30] showed that the knockout of EZR inhibits EGF-induced lung metastasis of colorectal cancer xenografts. Abnormal activation of EZR and NFκB are related to colorectal cancer metastasis and poor prognosis. EZR expression was also associated with the degree of tumor differentiation, lymph node metastasis, and Dukes stage [29,31]. Gavert et al. [32] reported that the NFκB-mediated signaling participates in cellular changes induced by L1 (immunoglobulin-like cell-adhesion receptors) that lead to invasive phenotype in colorectal cancer. Moreover, the same authors found that NFκB- and EZR-mediated signaling are essential for the ability of L1 to induce metastasis in colorectal cancer cells. The decrease in NFκB transactivation, EZR levels, or an L1 mutant with an altered EZR-binding domain blocked the ability of L1 to induce liver metastasis. Accordingly, colorectal cancer patients with moderate to robust EZR expression (by immunohistochemistry) presented a reduced five-year overall survival rate compared to patients with a negative or low-intensity EZR expression [29]. Increased EZR phosphorylation at T567 was also reported in samples from colorectal cancer patients, which was associated with the IGF1R signaling pathway and expression of XIAP and BIRC5, both apoptosis inhibitors [33].

4.5. Endometrial Cancer

In endometrial cancer, EZR expression is higher in the high-metastasis Ishikawa subclone (named mEIIL) compared to its parental low-metastasis cell line. Treatment with EZR antisense phosphorothioate oligonucleotides reduced invasiveness, but not proliferation, in mEIIL cells [34]. High EZR protein expression was reported in uterine endometrioid adenocarcinomas. EZR protein levels were also significantly increased in endometrial hyperplasias that more frequently progressed to invasive cancer and metastatic lesions than in the matched primary lesions. In endometrioid carcinomas, EZR was at least focally expressed in 93% of cases, which was quite heterogeneous, with a wide range from only a few to nearly 100% positive tumor cells [35].

4.6. Gastric Cancer

EZR immunostaining was positive in 81.1% of intestinal-type and 40.9% of diffuse-type adenocarcinoma cases, suggesting that low EZR expression indicates the loss of adhesion in diffuse carcinomas. Furthermore, EZR overexpression was related to Helicobacter pylori infection [36]. In another study, EZR expression was detected, at low levels, in 11.2% of non-tumor mucosae and, at elevated levels, in 59.2% of gastric cancer samples. EZR overexpression was associated with age, tumor size, location, differentiation stage, depth of invasion, vessel invasion, lymph node and distant metastasis, and TNM stage, and it was an independent prognostic factor in patients with gastric carcinoma [37]. Jin et al. reported similar findings [38]. The high frequency of EZR expression suggests a central role in gastric cancer biology. Thus, EZR protein expression could be used as an early diagnostic marker of gastric cancer and its precancerous disease, and its overexpression may be a predictor of poor prognosis [38].

4.7. Head and Neck Squamous Cell Carcinoma

In head and neck squamous cell carcinoma patients, high cytoplasmic EZR expression was observed in 92% of the tumors and independently associated with a worse prognosis [39]. These data were validated in a large cohort in which the high EZR expression was associated with poor survival outcomes. Notably, tumors with high cytoplasmic EZR display a more invasive phenotype [40].

4.8. Hepatocellular Carcinoma

EZR expression was higher in tumor samples than hepatic tissues and associated with metastasis and differentiation degree [41,42]. High EZR levels have been significantly associated with advanced TNM stage, poor Edmondsons histological grade, macroscopic portal vein invasion, tumor recurrence, and extrahepatic recurrence, and are independently associated with poor overall survival [43,44]. Increased levels of EZR were observed in hepatocarcinoma cell lines with higher metastatic potential, and EZR inhibition by siRNA reduced clonogenicity, cell proliferation, motility, and invasion [44,45]. EZR hyperphosphorylation has been identified as responsible for the invasion of hepatocellular cells, promoting intrahepatic metastasis in vivo and cell migration in vitro [46,47]. EZR overexpression promotes hepatocellular carcinoma cell proliferation, EMT, angiogenesis, and glycolysis reprogramming [46,47]. In hepatocellular carcinoma patients, EZR gene expression was significantly decreased after treatment with arsenic trioxide [48], suggesting a potential approach for pharmacological intervention on EZR expression.

4.9. Kidney Cancer

In renal cell cancer, the absence of EZR expression was an independent prognostic factor of disease-specific survival. EZR expression was also associated with incidental tumors, clinical stage, pT stage, synchronic metastasis, and ISUP histological grade [49,50].

4.10. Leukemia

In T-cell acute lymphoblastic leukemia cells, ERM phosphorylation has been found to promote invasion [51], and EZR deregulation has been related to the progression of mouse preleukemia erythroblasts toward malignancy [52]. In acute myeloid leukemia cells, functional studies have shown that EZR promotes survival and acts as an effector protein in cell signaling mediated by FLT3-ITD and mutated KIT receptors [52,53,54]. In a comprehensive analysis of cytoskeleton regulatory genes, Lipreri da Silva et al. [55] identified that high EZR expression is an independent marker of worse outcomes in acute myeloid leukemia patients, and EZR pharmacological inhibition reduced viability, proliferation, autonomous clonal growth, and cell cycle progression in leukemia cells. In chronic lymphocytic leukemia (CLL), EZR is highly expressed and associated with molecular signatures relevant to the diseases development and maintenance, including TP53, PI3K/AKT/mTOR, NFκB, and MAPK pathways [56]. Pharmacological EZR inhibition with NSC305787 reduced viability, clonogenicity, and cell cycle progression and induced apoptosis in CLL primary and cell lines [56].

4.11. Lymphoma

In diffuse large B-cell lymphoma, EZR is essential to B-cell antigen receptor organization at the cell membrane and supports proliferation and survival signals. EZR inhibition by siRNA or pharmacological agents reduced cell viability in lymphoma cells [57]. Moreover, it has been shown that ERM proteins regulate B- and T-cell activation by controlling BCR and TCR dynamics, scaffolding protein assembly, and membrane-associated intracellular signaling [58].

4.12. Lung Cancer

EZR expression was associated with late-stage tumors, pleural invasion, and poor survival outcomes in non-small cell lung carcinomas [59]. EZR phosphorylation at T567 and Y353 was significantly upregulated in non-small cell lung carcinomas compared with normal tissues, which was also correlated with poor differentiation and late clinical stage. However, only EZR phosphorylation at T567 overexpression was associated with the presence of lymph node metastasis and overall survival [60]. EZR may also be a possible predictor to assess the prognosis in patients with non-small cell lung carcinoma since its gene expression was associated with lymphatic metastasis [61].

4.13. Malignant Pleural Mesothelioma

Pleural mesothelioma expresses activated ERM proteins, with EZR being the principal component of this family of proteins associated with proliferation and migration in this disease [62].

4.14. Oral Squamous Cell Carcinoma

EZR expression was significantly associated with N-classification, UICC stage, and lymphangitic carcinomatosis [63]. Another study, using tissue microarray, observed that EZR is overexpressed in primary tongue squamous cell carcinoma and may be implicated in these cells growth, migration, and invasiveness, possibly regulating the E-cadherin/β-catenin complex [64]. Evaluation of EZR expression in human tongue cancer tissues by immunohistochemical staining showed that this protein was highly expressed in invasive squamous cell carcinoma compared to in situ carcinoma [65]. Another study also suggested that EZR may be involved in the progression of tongue carcinoma in situ to squamous cell carcinoma [66]. Immunoexpression results confirmed a correlation between EZR expression in squamous cell carcinoma of the lip, suggesting cooperative participation of these proteins in cell movement and invasion [67]. In tongue squamous cell carcinoma clinical samples, EZR activation at Y353 is associated with metastases and poor patient prognosis [68].

4.15. Ovarian Cancer

In OVCA cells, estradiol increased EZR expression, which was associated with invasive behavior [69]. In ovarian carcinoma patients, EZR expression presents a high predictive value for tumor-related death, including late stage [70]. EZR regulates OVCA cell proliferation, migration, and invasion by modulating EMT and inducing the formation of actin stress fibers by regulating Rho GTPase activity [71]. In three-dimensional cultured ovarian carcinoma cell lines, reduced expression of EZR by shRNA impaired invasiveness and cell branching ability [72].

4.16. Pancreatic Cancer

Zhou et al. [73] showed that the levels of p-EZR T567/p-EZR Y353 protein expression in the cytoplasm of pancreatic cancer cells increased with the TNM stage of human pancreatic cancers. The survival time of the group positive for p-EZR T567/p-EZR Y353 protein expression was shorter than that of the negative group [73]. EZR levels were significantly upregulated in pancreatic cancer patients samples and correlated with poor prognosis [74]. Two active phosphorylated sites of EZR, Y353 and T567, were differentially expressed in pancreatic cancer tissues. High EZR phosphorylation at Y353, but not T567, was associated with malignant progression and clinical outcomes of pancreatic cancer patients [75]. In pancreatic cancer cells, EZR was involved in the cytoskeleton modulation, protrusions, and microvilli, which were related to migration and invasion [76]. Bioinformatic tools showed that EZR expression predicted worse survival outcomes in the pancreatic adenocarcinoma cohort, and gene signatures were associated with EZR expression, including apoptosis, PI3K/AKT/mTOR signaling, estrogen response early, NOTCH signaling, estrogen response late, and pancreatic beta cells [77]. Pharmacological EZR inhibitor, NSC305787, reduced viability, clonogenicity, migration, and induced apoptosis in pancreatic cancer cells [77].

4.17. Prostate Cancer

EZR expression was moderate or strong in samples from prostate cancer patients and negative or weakly expressed in benign cases [78]. EZR staining was more intense in high-grade prostatic intraepithelial neoplasia than in other prostate cancer cells [79]. EZR was found to be an essential intracellular mediator of cell invasion in hormone-sensitive and hormone-resistant prostate cancer cells. Androgen treatment induces EZR expression and phosphorylation at T567 and Y353 residues. Overexpression of mutated EZR variants (T567A and Y353F) or EZR silencing by siRNA inhibited androgen-induced invasion [80]. In metastatic prostate cancer cell lines (22RV1 and PC-3), EZR overexpression promoted migratory and invasive abilities. Accordingly, EZR expression was significantly increased in circulating tumor cells from metastatic prostate cancer patients [81].
After radical prostatectomy, circulating tumor cells from EZR-positive prostate cancer patients were susceptible to tumor metastases [81]. In nude mice bearing prostate cancer, treatment with baicalein decreased tumor volume and weight, which were much more pronounced in those with in vivo EZR knockdown [82]. Furthermore, baicalein treatment suppresses proliferative capacity in prostate cancer cell lines, interrupts the cell cycle progression, and stimulates apoptosis through EZR downregulation [82].

5. Concluding Remarks and Pharmacological Advances for EZR Inhibition

In most tumors studied to date, EZR is highly expressed and predicts an unfavorable clinical outcome. Studies with genetic intervention in experimental models highlight the importance of EZR in maintaining the malignant phenotype and directing this gene/protein (or the signaling mediated by it) as a potential target for antineoplastic therapy.
Two synthetic compounds that act as selective EZR inhibitors (NSC668394 and NSC305787) have been evaluated in preclinical studies (Figure 4). These compounds directly bind to EZR, inhibiting T567 phosphorylation with low micromolar affinity [83]. It has also been demonstrated that these EZR inhibitors have distinct target-binding profiles. For example, NSC305787 bound and dissociated more quickly from EZR than NSC668394 [83]. Another point worth mentioning is that NSC305787 and NSC668394 are well tolerated in murine models without any apparent acute toxicity [83]. Regarding pharmacokinetics, NSC305787 has the most prolonged plasma half-life (>6 h by intraperitoneal (i.p.) injection) compared to NSC668394 (<0.5 h i.p.) [84]. Although these compounds have potent antineoplastic activity in models of osteosarcomas, lung cancer, leukemia, and pancreatic cancer, their underlying molecular mechanisms and potential application in other cancer models remain poorly explored [55,77,83].
A summary of EZR expression in cancer is presented in Table 1, and a summary of EZR functional assays in cancer cells is shown in Table 2.

Author Contributions

Conceptualization, J.C.L.d.S. and J.A.M.-N.; writing—original draft preparation, J.C.L.d.S. and J.A.M.-N.; writing—review and editing, H.P.V.; visualization, J.C.L.d.S. and H.P.V.; supervision, J.A.M.-N.; project administration, J.A.M.-N.; funding acquisition, J.A.M.-N. All authors have read and agreed to the published version of the manuscript.

Funding

J.C.L.d.S. and H.P.V. received fellowships from the São Paulo Research Foundation (FAPESP; grants #2020/12909-7 and #2021/01460-1). This study was supported by grants #2019/23864-7 and #2021/11606-3 from the FAPESP. This study was financed in part by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, Brasil (CAPES), Finance Code 001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Neisch, A.L.; Fehon, R.G. Ezrin, Radixin and Moesin: Key regulators of membrane-cortex interactions and signaling. Curr. Opin. Cell Biol. 2011, 23, 377–382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Kawaguchi, K.; Asano, S. Pathophysiological Roles of Actin-Binding Scaffold Protein, Ezrin. Int. J. Mol. Sci. 2022, 23, 3246. [Google Scholar] [CrossRef] [PubMed]
  3. Clucas, J.; Valderrama, F. ERM proteins in cancer progression. J. Cell Sci. 2014, 127, 267–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Krishnan, K.; Bruce, B.; Hewitt, S.; Thomas, D.; Khanna, C.; Helman, L.J. Ezrin mediates growth and survival in Ewing’s sarcoma through the AKT/mTOR, but not the MAPK, signaling pathway. Clin. Exp. Metastasis 2006, 23, 227–236. [Google Scholar] [CrossRef]
  5. Gautreau, A.; Poullet, P.; Louvard, D.; Arpin, M. Ezrin, a plasma membrane-microfilament linker, signals cell survival through the phosphatidylinositol 3-kinase/Akt pathway. Proc. Natl. Acad. Sci. USA 1999, 96, 7300–7305. [Google Scholar] [CrossRef] [Green Version]
  6. Del Peso, L.; Gonzalez-Garcia, M.; Page, C.; Herrera, R.; Nunez, G. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt. Science 1997, 278, 687–689. [Google Scholar] [CrossRef]
  7. Ivetic, A.; Ridley, A.J. Ezrin/radixin/moesin proteins and Rho GTPase signalling in leucocytes. Immunology 2004, 112, 165–176. [Google Scholar] [CrossRef]
  8. Ridley, A.J. Rho family proteins: Coordinating cell responses. Trends Cell Biol. 2001, 11, 471–477. [Google Scholar] [CrossRef]
  9. Li, N.; Kong, J.; Lin, Z.; Yang, Y.; Jin, T.; Xu, M.; Sun, J.; Chen, L. Ezrin promotes breast cancer progression by modulating AKT signals. Br. J. Cancer 2019, 120, 703–713. [Google Scholar] [CrossRef] [Green Version]
  10. Xie, J.J.; Zhang, F.R.; Tao, L.H.; Lu, Z.; Xu, X.E.; Jian, S.; Xu, L.Y.; Li, E.M. Expression of ezrin in human embryonic, fetal, and normal adult tissues. J. Histochem. Cytochem. 2011, 59, 1001–1008. [Google Scholar] [CrossRef]
  11. Michie, K.A.; Bermeister, A.; Robertson, N.O.; Goodchild, S.C.; Curmi, P.M.G. Two Sides of the Coin: Ezrin/Radixin/Moesin and Merlin Control Membrane Structure and Contact Inhibition. Int. J. Mol. Sci. 2019, 20, 1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Pelaseyed, T.; Viswanatha, R.; Sauvanet, C.; Filter, J.J.; Goldberg, M.L.; Bretscher, A. Ezrin activation by LOK phosphorylation involves a PIP(2)-dependent wedge mechanism. Elife 2017, 6, e22759. [Google Scholar] [CrossRef] [PubMed]
  13. Shabardina, V.; Kramer, C.; Gerdes, B.; Braunger, J.; Cordes, A.; Schafer, J.; Mey, I.; Grill, D.; Gerke, V.; Steinem, C. Mode of Ezrin-Membrane Interaction as a Function of PIP2 Binding and Pseudophosphorylation. Biophys. J. 2016, 110, 2710–2719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Herrig, A.; Janke, M.; Austermann, J.; Gerke, V.; Janshoff, A.; Steinem, C. Cooperative adsorption of ezrin on PIP2-containing membranes. Biochemistry 2006, 45, 13025–13034. [Google Scholar] [CrossRef] [PubMed]
  15. Bretscher, A.; Reczek, D.; Berryman, M. Ezrin: A protein requiring conformational activation to link microfilaments to the plasma membrane in the assembly of cell surface structures. J. Cell Sci. 1997, 110 Pt 24, 3011–3018. [Google Scholar] [CrossRef] [PubMed]
  16. Viswanatha, R.; Ohouo, P.Y.; Smolka, M.B.; Bretscher, A. Local phosphocycling mediated by LOK/SLK restricts ezrin function to the apical aspect of epithelial cells. J. Cell Biol. 2012, 199, 969–984. [Google Scholar] [CrossRef] [Green Version]
  17. Zhang, R.; Zhang, S.; Xing, R.; Zhang, Q. High expression of EZR (ezrin) gene is correlated with the poor overall survival of breast cancer patients. Thorac. Cancer 2019, 10, 1953–1961. [Google Scholar] [CrossRef] [Green Version]
  18. Elliott, B.E.; Meens, J.A.; SenGupta, S.K.; Louvard, D.; Arpin, M. The membrane cytoskeletal crosslinker ezrin is required for metastasis of breast carcinoma cells. Breast. Cancer Res. 2005, 7, R365. [Google Scholar] [CrossRef] [Green Version]
  19. Li, Q.; Wu, M.; Wang, H.; Xu, G.; Zhu, T.; Zhang, Y.; Liu, P.; Song, A.; Gang, C.; Han, Z.; et al. Ezrin silencing by small hairpin RNA reverses metastatic behaviors of human breast cancer cells. Cancer Lett. 2008, 261, 55–63. [Google Scholar] [CrossRef]
  20. Ma, L.; Jiang, T. Clinical implications of Ezrin and CD44 coexpression in breast cancer. Oncol. Rep. 2013, 30, 1899–1905. [Google Scholar] [CrossRef]
  21. Ilmonen, S.; Vaheri, A.; Asko-Seljavaara, S.; Carpen, O. Ezrin in primary cutaneous melanoma. Mod. Pathol. 2005, 18, 503–510. [Google Scholar] [CrossRef] [Green Version]
  22. Federici, C.; Brambilla, D.; Lozupone, F.; Matarrese, P.; de Milito, A.; Lugini, L.; Iessi, E.; Cecchetti, S.; Marino, M.; Perdicchio, M.; et al. Pleiotropic function of ezrin in human metastatic melanomas. Int. J. Cancer 2009, 124, 2804–2812. [Google Scholar] [CrossRef] [PubMed]
  23. Lugini, L.; Lozupone, F.; Matarrese, P.; Funaro, C.; Luciani, F.; Malorni, W.; Rivoltini, L.; Castelli, C.; Tinari, A.; Piris, A.; et al. Potent phagocytic activity discriminates metastatic and primary human malignant melanomas: A key role of ezrin. Lab. Invest. 2003, 83, 1555–1567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zhang, Y.; Wang, G. MicroRNA-183 inhibits A375 human melanoma cell migration and invasion by targeting Ezrin and MMP-9. Oncol. Lett. 2019, 17, 548–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kong, J.; Di, C.; Piao, J.; Sun, J.; Han, L.; Chen, L.; Yan, G.; Lin, Z. Ezrin contributes to cervical cancer progression through induction of epithelial-mesenchymal transition. Oncotarget 2016, 7, 19631–19642. [Google Scholar] [CrossRef] [Green Version]
  26. Kong, J.; Li, Y.; Liu, S.; Jin, H.; Shang, Y.; Quan, C.; Lin, Z. High expression of ezrin predicts poor prognosis in uterine cervical cancer. BMC Cancer 2013, 13, 520. [Google Scholar] [CrossRef] [Green Version]
  27. Auvinen, E.; Carpen, O.; Korpela, T.; Ronty, M.; Vaheri, A.; Tarkkanen, J. Altered expression of ezrin, E-Cadherin and beta-Catenin in cervical neoplasia. Neoplasma 2013, 60, 56–61. [Google Scholar] [CrossRef] [Green Version]
  28. Greco, D.; Kivi, N.; Qian, K.; Leivonen, S.K.; Auvinen, P.; Auvinen, E. Human papillomavirus 16 E5 modulates the expression of host microRNAs. PLoS ONE 2011, 6, e21646. [Google Scholar] [CrossRef] [Green Version]
  29. Patara, M.; Santos, E.M.; Coudry Rde, A.; Soares, F.A.; Ferreira, F.O.; Rossi, B.M. Ezrin expression as a prognostic marker in colorectal adenocarcinoma. Pathol. Oncol. Res. 2011, 17, 827–833. [Google Scholar] [CrossRef]
  30. Li, Y.; Lin, Z.; Chen, B.; Chen, S.; Jiang, Z.; Zhou, T.; Hou, Z.; Wang, Y. Ezrin/NF-kB activation regulates epithelial- mesenchymal transition induced by EGF and promotes metastasis of colorectal cancer. Biomed. Pharmacother. 2017, 92, 140–148. [Google Scholar] [CrossRef]
  31. Wang, H.J.; Zhu, J.S.; Zhang, Q.; Sun, Q.; Guo, H. High level of ezrin expression in colorectal cancer tissues is closely related to tumor malignancy. World J. Gastroenterol. 2009, 15, 2016–2019. [Google Scholar] [CrossRef] [PubMed]
  32. Gavert, N.; Ben-Shmuel, A.; Lemmon, V.; Brabletz, T.; Ben-Ze’ev, A. Nuclear factor-kappaB signaling and ezrin are essential for L1-mediated metastasis of colon cancer cells. J. Cell Sci. 2010, 123, 2135–2143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Leiphrakpam, P.D.; Rajput, A.; Mathiesen, M.; Agarwal, E.; Lazenby, A.J.; Are, C.; Brattain, M.G.; Chowdhury, S. Ezrin expression and cell survival regulation in colorectal cancer. Cell Signal 2014, 26, 868–879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ohtani, K.; Sakamoto, H.; Rutherford, T.; Chen, Z.; Satoh, K.; Naftolin, F. Ezrin, a membrane-cytoskeletal linking protein, is involved in the process of invasion of endometrial cancer cells. Cancer Lett. 1999, 147, 31–38. [Google Scholar] [CrossRef]
  35. Kobel, M.; Langhammer, T.; Huttelmaier, S.; Schmitt, W.D.; Kriese, K.; Dittmer, J.; Strauss, H.G.; Thomssen, C.; Hauptmann, S. Ezrin expression is related to poor prognosis in FIGO stage I endometrioid carcinomas. Mod. Pathol. 2006, 19, 581–587. [Google Scholar] [CrossRef] [Green Version]
  36. Bal, N.; Yildirim, S.; Nursal, T.Z.; Bolat, F.; Kayaselcuk, F. Association of ezrin expression in intestinal and diffuse gastric carcinoma with clinicopathological parameters and tumor type. World J. Gastroenterol. 2007, 13, 3726–3729. [Google Scholar] [CrossRef]
  37. Li, L.; Wang, Y.Y.; Zhao, Z.S.; Ma, J. Ezrin is associated with gastric cancer progression and prognosis. Pathol. Oncol. Res. 2011, 17, 909–915. [Google Scholar] [CrossRef]
  38. Jin, J.; Jin, T.; Quan, M.; Piao, Y.; Lin, Z. Ezrin overexpression predicts the poor prognosis of gastric adenocarcinoma. Diagn. Pathol. 2012, 7, 135. [Google Scholar] [CrossRef] [Green Version]
  39. Madan, R.; Brandwein-Gensler, M.; Schlecht, N.F.; Elias, K.; Gorbovitsky, E.; Belbin, T.J.; Mahmood, R.; Breining, D.; Qian, H.; Childs, G.; et al. Differential tissue and subcellular expression of ERM proteins in normal and malignant tissues: Cytoplasmic ezrin expression has prognostic signficance for head and neck squamous cell carcinoma. Head Neck 2006, 28, 1018–1027. [Google Scholar] [CrossRef]
  40. Schlecht, N.F.; Brandwein-Gensler, M.; Smith, R.V.; Kawachi, N.; Broughel, D.; Lin, J.; Keller, C.E.; Reynolds, P.A.; Gunn-Moore, F.J.; Harris, T.; et al. Cytoplasmic ezrin and moesin correlate with poor survival in head and neck squamous cell carcinoma. Head Neck Pathol. 2012, 6, 232–243. [Google Scholar] [CrossRef]
  41. Bakheet, A.M.H.; Mahmoud, S.A.; Huang, Y.; Zhang, J.; Wang, J.; Wei, Y.; Gamallat, Y.; Awadasseid, A.; Owusu, L.; Khidir, Y.; et al. Ezrin as a possible diagnostic and/or prognostic biomarker in mice lymphatic metastatic hepatocellular carcinoma in vivo. Biofactors 2017, 43, 662–672. [Google Scholar] [CrossRef] [PubMed]
  42. Pan, D.; Wang, S.; Ye, H.; Xu, S.; Ye, G. Ezrin expression in the primary hepatocellular carcinoma patients and associated with clinical, pathological characteristics. J. Cancer Res. Ther. 2016, 12, 291–294. [Google Scholar]
  43. Kang, Y.K.; Hong, S.W.; Lee, H.; Kim, W.H. Prognostic implications of ezrin expression in human hepatocellular carcinoma. Mol. Carcinog. 2010, 49, 798–804. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, X.; Li, N.; Han, A.; Wang, Y.; Lin, Z.; Yang, Y. Ezrin promotes hepatocellular carcinoma progression by modulating glycolytic reprogramming. Cancer Sci. 2020, 111, 4061–4074. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, Y.; Hu, M.Y.; Wu, W.Z.; Wang, Z.J.; Zhou, K.; Zha, X.L.; Liu, K.D. The membrane-cytoskeleton organizer ezrin is necessary for hepatocellular carcinoma cell growth and invasiveness. J. Cancer Res. Clin. Oncol. 2006, 132, 685–697. [Google Scholar] [CrossRef] [PubMed]
  46. Du, S.; Song, X.; Li, Y.; Cao, Y.; Chu, F.; Durojaye, O.A.; Su, Z.; Shi, X.; Wang, J.; Cheng, J.; et al. Celastrol inhibits ezrin-mediated migration of hepatocellular carcinoma cells. Sci. Rep. 2020, 10, 11273. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, Y.; Wang, D.; Guo, Z.; Zhao, J.; Wu, B.; Deng, H.; Zhou, T.; Xiang, H.; Gao, F.; Yu, X.; et al. Rho kinase phosphorylation promotes ezrin-mediated metastasis in hepatocellular carcinoma. Cancer Res. 2011, 71, 1721–1729. [Google Scholar] [CrossRef] [Green Version]
  48. Lu, W.; Yang, C.; Du, P.; Zhang, J.L.; Zhang, J.C. Effects of arsenic trioxide on the expression of ezrin in hepatocellular carcinoma. Medicine 2017, 96, e7602. [Google Scholar] [CrossRef]
  49. Cetin, B.; Gonul, I.I.; Gumusay, O.; Afsar, B.; Bilgetekin, I.; Ozet, A.; Uner, A. Ezrin is a prognostic biomarker in patients with clear cell metastatic renal cell carcinoma receiving sunitinib. J. Cancer Res. Ther. 2021, 17, 408–413. [Google Scholar] [CrossRef]
  50. Ferrari, M.V.O.; da Costa, W.H.; Matushita, M.A.M.; Meduna, R.R.; Brazao, E.S., Jr.; Bezerra, S.M.; da Cunha, I.W.; Zequi, S.C. Immunohistochemical negative expression of ezrin predicts poor prognosis in clear cell renal cell carcinoma. Urol. Oncol. 2020, 38, 75.e1–75.e7. [Google Scholar] [CrossRef]
  51. Altaf, E.; Huang, X.; Xiong, J.; Yang, X.; Deng, X.; Xiong, M.; Zhou, L.; Pan, S.; Yuan, W.; Li, X.; et al. NHE1 has a notable role in metastasis and drug resistance of T-cell acute lymphoblastic leukemia. Oncol. Lett. 2017, 14, 4256–4262. [Google Scholar] [CrossRef] [PubMed]
  52. Habif, G.; Grasset, M.F.; Kieffer-Jaquinod, S.; Kuhn, L.; Mouchiroud, G.; Gobert-Gosse, S. Phosphoproteome analyses reveal specific implications of Hcls1, p21-activated kinase 1 and Ezrin in proliferation of a myeloid progenitor cell line downstream of wild-type and ITD mutant Fms-like tyrosine kinase 3 receptors. J. Proteomics 2013, 78, 231–244. [Google Scholar] [CrossRef] [PubMed]
  53. Corcoran, A.; Cotter, T.G. FLT3-driven redox-modulation of Ezrin regulates leukaemic cell migration. Free Radic Res. 2013, 47, 20–34. [Google Scholar] [CrossRef] [PubMed]
  54. Monni, R.; Haddaoui, L.; Naba, A.; Gallais, I.; Arpin, M.; Mayeux, P.; Moreau-Gachelin, F. Ezrin is a target for oncogenic Kit mutants in murine erythroleukemia. Blood 2008, 111, 3163–3172. [Google Scholar] [CrossRef] [Green Version]
  55. Lipreri da Silva, J.C.; Coelho-Silva, J.L.; Lima, K.; Vicari, H.P.; Lazarini, M.; Costa-Lotufo, L.V.; Traina, F.; Machado-Neto, J.A. Comprehensive analysis of cytoskeleton regulatory genes identifies ezrin as a prognostic marker and molecular target in acute myeloid leukemia. Cell Oncol. 2021, 44, 1105–1117. [Google Scholar] [CrossRef]
  56. Lipreri da Silva, J.C.; Saldanha-Araujo, F.; de Melo, R.C.B.; Vicari, H.P.; Silva-Carvalho, A.E.; Rego, E.M.; Buccheri, V.; Machado-Neto, J.A. Ezrin is highly expressed and a druggable target in chronic lymphocytic leukemia. Life Sci. 2022, 311, 121146. [Google Scholar] [CrossRef]
  57. Pore, D.; Bodo, J.; Danda, A.; Yan, D.; Phillips, J.G.; Lindner, D.; Hill, B.T.; Smith, M.R.; Hsi, E.D.; Gupta, N. Identification of Ezrin-Radixin-Moesin proteins as novel regulators of pathogenic B-cell receptor signaling and tumor growth in diffuse large B-cell lymphoma. Leukemia 2015, 29, 1857–1867. [Google Scholar] [CrossRef] [Green Version]
  58. Pore, D.; Gupta, N. The ezrin-radixin-moesin family of proteins in the regulation of B-cell immune response. Crit. Rev. Immunol. 2015, 35, 15–31. [Google Scholar] [CrossRef] [Green Version]
  59. Lee, H.W.; Kim, E.H.; Oh, M.H. Clinicopathologic implication of ezrin expression in non-small cell lung cancer. Korean J. Pathol. 2012, 46, 470–477. [Google Scholar] [CrossRef]
  60. Jin, T.; Jin, J.; Li, X.; Zhang, S.; Choi, Y.H.; Piao, Y.; Shen, X.; Lin, Z. Prognostic implications of ezrin and phosphorylated ezrin expression in non-small cell lung cancer. BMC Cancer 2014, 14, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Kolegova, E.S.; Kakurina, G.V.; Kostromitskiy, D.N.; Dobrodeev, A.Y.; Kondakova, I.V. Increases in mRNA and Protein Levels of the Genes for the Actin-Binding Proteins Profilin, Fascin, and Ezrin Promote Metastasis in Non-Small Cell Lung Cancer. Mol. Biol. 2020, 54, 285–292. [Google Scholar] [CrossRef]
  62. Pignochino, Y.; Dell’Aglio, C.; Inghilleri, S.; Zorzetto, M.; Basirico, M.; Capozzi, F.; Canta, M.; Piloni, D.; Cemmi, F.; Sangiolo, D.; et al. The combination of sorafenib and everolimus shows antitumor activity in preclinical models of malignant pleural mesothelioma. BMC Cancer 2015, 15, 374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Safi, A.F.; Nickenig, H.J.; Rothamel, D.; Zirk, M.; Thiele, O.; Grandoch, A.; Scheer, M.; Zinser, M.; Zoller, J.; Drebber, U.; et al. Expression of ezrin in oral squamous cell carcinoma: Prognostic impact and clinicopathological correlations. J. Craniomaxillofac. Surg. 2015, 43, 1899–1905. [Google Scholar] [CrossRef] [PubMed]
  64. Saito, S.; Yamamoto, H.; Mukaisho, K.; Sato, S.; Higo, T.; Hattori, T.; Yamamoto, G.; Sugihara, H. Mechanisms underlying cancer progression caused by ezrin overexpression in tongue squamous cell carcinoma. PLoS ONE 2013, 8, e54881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Noi, M.; Mukaisho, K.I.; Yoshida, S.; Murakami, S.; Koshinuma, S.; Adachi, T.; Machida, Y.; Yamori, M.; Nakayama, T.; Yamamoto, G.; et al. ERK phosphorylation functions in invadopodia formation in tongue cancer cells in a novel silicate fibre-based 3D cell culture system. Int. J. Oral. Sci. 2018, 10, 30. [Google Scholar] [CrossRef] [PubMed]
  66. Noi, M.; Mukaisho, K.I.; Murakami, S.; Koshinuma, S.; Machida, Y.; Yamori, M.; Nakayama, T.; Ogawa, T.; Nakata, Y.; Shimizu, T.; et al. Expressions of ezrin, ERK, STAT3, and AKT in tongue cancer and association with tumor characteristics and patient survival. Clin. Exp. Dent. Res. 2020, 6, 420–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Assao, A.; Nonogaki, S.; Lauris, J.R.P.; Carvalho, A.L.; Pinto, C.A.L.; Soares, F.A.; Kowalski, L.P.; Oliveira, D.T. Podoplanin, ezrin, and Rho-A proteins may have joint participation in tumor invasion of lip cancer. Clin. Oral. Investig. 2017, 21, 1647–1657. [Google Scholar] [CrossRef]
  68. Wang, Y.; Lin, Z.; Sun, L.; Fan, S.; Huang, Z.; Zhang, D.; Yang, Z.; Li, J.; Chen, W. Akt/Ezrin Tyr353/NF-kappaB pathway regulates EGF-induced EMT and metastasis in tongue squamous cell carcinoma. Br. J. Cancer 2014, 110, 695–705. [Google Scholar] [CrossRef] [Green Version]
  69. Song, J.; Fadiel, A.; Edusa, V.; Chen, Z.; So, J.; Sakamoto, H.; Fishman, D.A.; Naftolin, F. Estradiol-induced ezrin overexpression in ovarian cancer: A new signaling domain for estrogen. Cancer Lett. 2005, 220, 57–65. [Google Scholar] [CrossRef]
  70. Kobel, M.; Gradhand, E.; Zeng, K.; Schmitt, W.D.; Kriese, K.; Lantzsch, T.; Wolters, M.; Dittmer, J.; Strauss, H.G.; Thomssen, C.; et al. Ezrin promotes ovarian carcinoma cell invasion and its retained expression predicts poor prognosis in ovarian carcinoma. Int. J. Gynecol. Pathol. 2006, 25, 121–130. [Google Scholar] [CrossRef]
  71. Li, M.J.; Xiong, D.; Huang, H.; Wen, Z.Y. Ezrin Promotes the Proliferation, Migration, and Invasion of Ovarian Cancer Cells. Biomed. Environ. Sci. 2021, 34, 139–151. [Google Scholar] [PubMed]
  72. Horwitz, V.; Davidson, B.; Stern, D.; Trope, C.G.; Tavor Re’em, T.; Reich, R. Ezrin Is Associated with Disease Progression in Ovarian Carcinoma. PLoS ONE 2016, 11, e0162502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Zhou, J.; Feng, Y.; Tao, K.; Su, Z.; Yu, X.; Zheng, J.; Zhang, L.; Yang, D. The expression and phosphorylation of ezrin and merlin in human pancreatic cancer. Int. J. Oncol. 2014, 44, 2059–2067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Quan, C.; Sun, J.; Lin, Z.; Jin, T.; Dong, B.; Meng, Z.; Piao, J. Ezrin promotes pancreatic cancer cell proliferation and invasion through activating the Akt/mTOR pathway and inducing YAP translocation. Cancer Manag. Res. 2019, 11, 6553–6566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Cui, Y.; Li, T.; Zhang, D.; Han, J. Expression of Ezrin and phosphorylated Ezrin (pEzrin) in pancreatic ductal adenocarcinoma. Cancer Invest. 2010, 28, 242–247. [Google Scholar] [CrossRef]
  76. Meng, Y.; Lu, Z.; Yu, S.; Zhang, Q.; Ma, Y.; Chen, J. Ezrin promotes invasion and metastasis of pancreatic cancer cells. J. Transl. Med. 2010, 8, 61. [Google Scholar] [CrossRef] [Green Version]
  77. Lipreri da Silva, J.C.; Carvalho, M.F.L.; de Miranda, L.B.L.; de Almeida, B.O.; Lima, K.; Machado-Neto, J.A. NSC305787, a pharmacological ezrin inhibitor, exhibits antineoplastic activity in pancreatic cancer cells. Invest. New Drugs 2022, 40, 728–737. [Google Scholar] [CrossRef]
  78. Valdman, A.; Fang, X.; Pang, S.T.; Nilsson, B.; Ekman, P.; Egevad, L. Ezrin expression in prostate cancer and benign prostatic tissue. Eur. Urol. 2005, 48, 852–857. [Google Scholar] [CrossRef]
  79. Pang, S.T.; Fang, X.; Valdman, A.; Norstedt, G.; Pousette, A.; Egevad, L.; Ekman, P. Expression of ezrin in prostatic intraepithelial neoplasia. Urology 2004, 63, 609–612. [Google Scholar] [CrossRef]
  80. Chuan, Y.C.; Pang, S.T.; Cedazo-Minguez, A.; Norstedt, G.; Pousette, A.; Flores-Morales, A. Androgen induction of prostate cancer cell invasion is mediated by ezrin. J. Biol. Chem. 2006, 281, 29938–29948. [Google Scholar] [CrossRef] [Green Version]
  81. Chen, Z.; Wang, J.; Lu, Y.; Lai, C.; Qu, L.; Zhuo, Y. Ezrin expression in circulating tumor cells is a predictor of prostate cancer metastasis. Bioengineered 2022, 13, 4076–4084. [Google Scholar] [CrossRef] [PubMed]
  82. Ma, S.C.; Chen, R.; Yang, T.N.; Zhang, F.; Zhao, D. Baicalein inhibits the proliferative activity of human prostate cancer cell line PC3 by downregulating Ezrin. J. Biol. Regul. Homeost. Agents 2020, 34, 885–892. [Google Scholar] [PubMed]
  83. Bulut, G.; Hong, S.H.; Chen, K.; Beauchamp, E.M.; Rahim, S.; Kosturko, G.W.; Glasgow, E.; Dakshanamurthy, S.; Lee, H.S.; Daar, I.; et al. Small molecule inhibitors of ezrin inhibit the invasive phenotype of osteosarcoma cells. Oncogene 2012, 31, 269–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Celik, H.; Bulut, G.; Han, J.; Graham, G.T.; Minas, T.Z.; Conn, E.J.; Hong, S.H.; Pauly, G.T.; Hayran, M.; Li, X.; et al. Ezrin Inhibition Up-regulates Stress Response Gene Expression. J. Biol. Chem. 2016, 291, 13257–13270. [Google Scholar] [CrossRef] [Green Version]
  85. Ohtani, K.; Sakamoto, H.; Rutherford, T.; Chen, Z.; Kikuchi, A.; Yamamoto, T.; Satoh, K.; Naftolin, F. Ezrin, a membrane-cytoskeletal linking protein, is highly expressed in atypical endometrial hyperplasia and uterine endometrioid adenocarcinoma. Cancer Lett. 2002, 179, 79–86. [Google Scholar] [CrossRef]
Figure 1. Ezrin-mediated signaling pathways and cellular processes. When phosphorylated, ezrin (EZR) may bind membrane proteins such as G protein-coupled receptors (GPCRs) and other receptors allowing the binding of F-actin (actin filaments) with the membrane, promoting reorganization of the cytoskeleton. EZR also binds to several transmembrane receptors, such as growth factor or cytokine receptors, promoting the activation of multiple signaling pathways. Among the main signaling pathways triggered by EZR, we highlight the PI3K/AKT/mTOR pathway, which promotes protein synthesis and cell proliferation, and the Rho GTPase pathway, which promotes migration, invasion, and actin polymerization. Abbreviations: NHE3, Na+/H+ exchanger; CFTR, cystic fibrosis transmembrane conductance regulator; ICAM1, intercellular adhesion molecule 1; CD44, cell surface adhesion receptor; GF, growth factor; TKR, tyrosine kinase receptor; P, phosphorylation; GTP, guanosine triphosphate; PI3K, phosphatidylinositol 3-kinase; AKT, protein kinase B; mTOR, mammalian target of rapamycin; S6RP, ribosomal protein S6; 4EBP1, eukaryotic initiation factor 4E-binding protein 1; Rho, homologous Ras proteins; ROCK, Rho kinase; MLC, myosin light chain; LIMK, LIM kinase.
Figure 1. Ezrin-mediated signaling pathways and cellular processes. When phosphorylated, ezrin (EZR) may bind membrane proteins such as G protein-coupled receptors (GPCRs) and other receptors allowing the binding of F-actin (actin filaments) with the membrane, promoting reorganization of the cytoskeleton. EZR also binds to several transmembrane receptors, such as growth factor or cytokine receptors, promoting the activation of multiple signaling pathways. Among the main signaling pathways triggered by EZR, we highlight the PI3K/AKT/mTOR pathway, which promotes protein synthesis and cell proliferation, and the Rho GTPase pathway, which promotes migration, invasion, and actin polymerization. Abbreviations: NHE3, Na+/H+ exchanger; CFTR, cystic fibrosis transmembrane conductance regulator; ICAM1, intercellular adhesion molecule 1; CD44, cell surface adhesion receptor; GF, growth factor; TKR, tyrosine kinase receptor; P, phosphorylation; GTP, guanosine triphosphate; PI3K, phosphatidylinositol 3-kinase; AKT, protein kinase B; mTOR, mammalian target of rapamycin; S6RP, ribosomal protein S6; 4EBP1, eukaryotic initiation factor 4E-binding protein 1; Rho, homologous Ras proteins; ROCK, Rho kinase; MLC, myosin light chain; LIMK, LIM kinase.
Futurepharmacol 03 00005 g001
Figure 2. Protein structure and activation model for ezrin (EZR). (A) The N-terminus consists of a domain called FERM (band 4.1 protein, ezrin, radixin, moesin) formed by three subunits F1, F2, and F3, after which there is a helical domain called α1H, α3H, and α3H followed by a C-terminal domain (CTD) subunit, where the ezrin phosphorylation sites (Y353 and T567) are found. (B) Representation of active and inactive forms of EZR. It can interact intermolecularly with itself when inactive, forming a monomer or even a dimer with another protein. When it experiences the action of kinases and is phosphorylated, ezrin is activated, “extends”, and passes to a new conformation. The action of phosphatases reverses this conformation. This illustration was adapted from Michie et al. [11]. The three subunits F1 (blue lobe), F2 (purple lobe), and F3 (green lobe) for the FERM domain, helical domain (orange line), and C-terminal domain (pink) are illustrated. (C) The 3D reconstitution of the EZR protein was constructed using the SWISS-MODEL platform (https://swissmodel.expasy.org/, accessed on 2 August 2021), and cartoon and surface versions of the protein are illustrated. Alpha-helix structures are shown in purple, beta-pleated sheet structures are in green, and other structures are in white.
Figure 2. Protein structure and activation model for ezrin (EZR). (A) The N-terminus consists of a domain called FERM (band 4.1 protein, ezrin, radixin, moesin) formed by three subunits F1, F2, and F3, after which there is a helical domain called α1H, α3H, and α3H followed by a C-terminal domain (CTD) subunit, where the ezrin phosphorylation sites (Y353 and T567) are found. (B) Representation of active and inactive forms of EZR. It can interact intermolecularly with itself when inactive, forming a monomer or even a dimer with another protein. When it experiences the action of kinases and is phosphorylated, ezrin is activated, “extends”, and passes to a new conformation. The action of phosphatases reverses this conformation. This illustration was adapted from Michie et al. [11]. The three subunits F1 (blue lobe), F2 (purple lobe), and F3 (green lobe) for the FERM domain, helical domain (orange line), and C-terminal domain (pink) are illustrated. (C) The 3D reconstitution of the EZR protein was constructed using the SWISS-MODEL platform (https://swissmodel.expasy.org/, accessed on 2 August 2021), and cartoon and surface versions of the protein are illustrated. Alpha-helix structures are shown in purple, beta-pleated sheet structures are in green, and other structures are in white.
Futurepharmacol 03 00005 g002
Figure 3. EZR genomic alterations in cancer. (A) Distribution of EZR genetic alterations in The Cancer Genome Atlas (TCGA) cohorts (10953 patients in 32 studies). (B) A total of 110 somatic mutations were found: 84 missense substitutions, 16 truncating, one splice mutation, and nine gene fusions. The figures were obtained using the cBioPortal (http://www.cbioportal.org, accessed on 2 August 2021).
Figure 3. EZR genomic alterations in cancer. (A) Distribution of EZR genetic alterations in The Cancer Genome Atlas (TCGA) cohorts (10953 patients in 32 studies). (B) A total of 110 somatic mutations were found: 84 missense substitutions, 16 truncating, one splice mutation, and nine gene fusions. The figures were obtained using the cBioPortal (http://www.cbioportal.org, accessed on 2 August 2021).
Futurepharmacol 03 00005 g003
Figure 4. Chemical structures of putative pharmacological EZR inhibitors NSC668394 (A) and NSC305787 (B).
Figure 4. Chemical structures of putative pharmacological EZR inhibitors NSC668394 (A) and NSC305787 (B).
Futurepharmacol 03 00005 g004
Table 1. Summary of EZR expression in cancer.
Table 1. Summary of EZR expression in cancer.
PublicationsCancer TypeCell LinesPrimary TissuesTechniquesNotes
Zhang et al. [17]Breast cancerNoYes, 120 samples from breast cancer patients.IHCEZR was upregulated in breast cancer and may be used as a potential biomarker for overall survival.
Li et al. [19]Breast cancerYes, MCF-7, MDA-MB-453, MDA-MB-435s, and MDA-MB-231Yes, tumor samples from 23 patients with invasive human ductal breast cancer.RT-PCR, IHCEZR protein expression was significantly higher in primary cancer tissues than in lymph node metastases.
Ilmonen et al. [21]MelanomaNoYes, 95 samples from patients with primary melanoma of the skin. IHCEZR expression correlated with tumor thickness and level of invasion.
Kong et al. [26]Cervical cancerNoYes, 586 samples were collected from routinely processed and diagnosed uterine cervical lesion tissues from pretreatment surgical resections.qRT-PCR, IHCEZR was a potentially effective predictor of the poor prognosis of cervical cancer patients, especially those with early-stage disease. The determination of EZR expression levels may help to identify high-risk cervical cancer.
Patara et al. [29]Colorectal cancerNoYes, samples from tumors and construction of a TMA. TMA, IHCIncreased cytoplasmatic EZR expression correlated with higher tumor aggressiveness and a worse prognosis in colorectal cancer patients.
Gavert et al. [32]Colorectal cancerYes, Ls174T and SW620Yes, samples from tumor and normal colon tissues. Invading tumor cells expressed high EZR levels.
Leiphrakpam et al. [33].Colorectal cancerYes, GEO, GEORI, CBS, HCT166, and HCT166bYes, samples of tumors and construction of a TMA.TMA, IHCIncreased expression of p-EZRT567 was found in liver metastasis of orthotopic implantation studies in vivo and IHC studies of human CRC patient specimens.
Ohtani et al. [85]Endometrial cancerNoYes, 20 samples from uterine endometrioid adenocarcinoma, 7 samples from simple endometrial hyperplasias, 7 samples from complex endometrial hyperplasias, 7 samples from atypical endometrial hyperplasias, and 12 samples of normal endometrium.IHCEZR protein was explicitly expressed in uterine endometrioid adenocarcinoma and its precursor lesions. EZR protein expression in metastatic lesions increased and shifted from the cytosol to the membrane.
Bal et al. [36].Gastric cancerNoYes, samples from 53 intestinal-type adenocarcinoma and 22 diffuse-type carcinoma patients.IHCH. pylori-infected gastric carcinomas presented increased EZR expression.
Li et al. [37]Gastric cancerNoYes, samples from 436 gastric cancer patients.TMA, IHCEZR was upregulated in gastric cancer tissues compared with normal gastric tissues and correlated significantly with prognosis. In addition, high levels of EZR expression were associated with age, tumor size, location, depth of invasion, vessel invasion, lymph node and distant metastasis, and TNM stage.
Schlecht et al. [40]Head and neck squamous cell carcinomaNoYes, samples from 128 primary HNSCC were collected at initial diagnosis and treatment.TMA, global RNA expressionNo association with prognosis for HNSCC was found due to the inherent heterogeneity in disease management.
Lee et al. [59]Lung cancerNoYes, 112 NSCLC specimens. TMA, IHCEZR was overexpressed in tumor tissues. EZR expression correlated with pleural invasion and pathological stage. The negative group for EZR expression presented more prolonged survival.
Jin et al. [60]Lung cancerNoYes, samples from 150 NSCLC tumors and 14 normal lungs.IHC, qRT-PCREZR was upregulated in NSCLC. Increased EZR levels correlated with NSCLC late stage and poor differentiation. p-EZRT567 correlated with the presence of lymph node metastases. EZR was associated with reduced survival time for patients with early-stage NSCLC.
Kolegova et al. [61]Lung cancerNoYes, samples from 46 NSCLC patientsqRT-PCR, Western blottingIncreased EZR gene and protein expression was found in patients with distant metastasis.
Lipreri da Silva et al. [55]Acute myeloid leukemiaYes, Kasumi-1 and MOLM-13Yes, the cohort deposited in TCGA.BioinformaticsEZR expression was a prognostic marker in AML. In intermediate-risk AML patients, high EZR expression distinguished a group with a poor prognosis.
Lipreri da Silva et al. [56]Chronic lymphocytic leukemiaYes, MEC-1Yes, samples from 56 CLL patients and ten age-matched healthy donors.Bioinformatics, cDNA microarray, qPCREZR was highly expressed and positively associated with relevant signaling pathways related to CLL development and progression, including TP53, PI3K/AKT/mTOR, NFκB, and MAPK.
Safi et al. [63]Oral squamous cell carcinomaNoYes, resection specimens from 80 treatment-naive OSCC patients.IHCEZR expression had a significant impact on overall survival. Increased EZR expression was associated with malignant progression, leading to a higher risk of metastases.
Saito et al. [64]Oral squamous cell carcinomaYes, HSC-3Yes, 79 samples (10 normal tongue tissues and 69 tongue SCC tissues)IHC, microarrayEZR was overexpressed in 46.4% of the tumors examined. EZR expression was correlated with proliferative activity.
Noi et al. [65]Oral squamous cell carcinomaYes, HSC-3 and HSC-4 (3D culture)Yes, human tongue cancer tissue (CIS (12 cases) and invasive SCC of the tongue (T1: ten cases, T2: three cases)).IHCHigher expression of EZR was found in invasive squamous cell carcinoma than in carcinoma in situ.
Noi et al. [66]Oral squamous cell carcinomaNoYes, in situ tongue carcinoma patients (CIS, n = 17) and tongue squamous cell carcinoma patients (SCC, n = 46).IHCEZR expression was highly expressed in the cell membrane. EZR appears to be involved in the progress from in situ carcinoma in the tongue into squamous cell carcinoma.
Assao et al. [58]Oral squamous cell carcinomaNoYes, samples of 91 patients with primary squamous cell carcinoma of the lower lip.IHCEZR expression in squamous cell carcinoma of the lip suggests the participation of this protein in cell movement and invasion.
Wang et al. [68]Oral squamous cell carcinomaYes, SCC9 and SCC25Yes, primary tongue carcinomas collected from 63 patients.IHCEZR may be a therapeutic target to reverse EMT in tongue cancers and prevent TSCC progression. EZR activation was associated with metastasis and poor patient survival in TSCCs.
Köbel et al. [70]Ovarian cancerYes, SKOV-3Yes, ovarian carcinoma samples from 105 patients.IHCEZR was overexpressed in 49% of the samples. EZR expression correlated with reduced overall survival and appeared as an independent risk factor.
Li et al. [71]Ovarian cancerYes, SKOV3 and CaOV3No.Western blotting, qPCREZR was overexpressed in either CaOV3 or SKOV3.
Horwitz et al. [72].Ovarian cancerYes, ES2 and OVCAR3Yes, 93 effusions (76 peritoneal, 17 pleural) from 93 patients diagnosed with high-grade serous carcinoma (HGSC).qRT-PCRHigh expression of EZR was found in tumors. EZR expression in effusions was unrelated to clinical outcome.
Zhou et al. [73]Pancreatic cancerYes, SW1990Yes, 19 samples were obtained from patients undergoing surgical resection.IHC, plasmid transfection, target gene expressionThe expression of phosphorylated EZR proteins was related to pancreatic cancer’s clinical and pathological features. Phosphorylated EZR induced a positive regulatory role in growth, adhesion, and invasion.
Liprei da Silva et al. [77]Pancreatic cancerNoYes, the cohort was deposited in the TCGA.BioinformaticsHigh EZR expression predicted worse survival outcomes in the pancreatic adenocarcinoma cohort.
Valdman et al. [78]Prostate cancerNoYes, 103 radical prostatectomy specimens.IHCEZR immunoreactivity in prostate cancers was moderate or strong in 70% of specimens. EZR expression was correlated with Gleason score and seminal vesicle invasion.
Pang et al. [79]Prostate cancerNoYes, 19 high-grade prostatic intraepithelial neoplasia (HGPIN) samples obtained from radical prostatectomy specimens.IHCImmunoreactivity for EZR was absent or weak in benign prostatic epithelial cells. The immunostaining was moderate or strong in all HGPIN samples.
Chen et al. [81]Prostate cancerYes, 22RV1 and PC-3Yes, samples from 80 prostate cancer patients.qRT-PCR and IHCEzrin was significantly increased in prostate cancer tissues and 22RV1 and PC-3 cell samples. EZR expression in CTCs from patients with prostate cancer was significantly increased with metastatic grade.
Abbreviations: EZR, ezrin; IHC, immunohistochemistry; RT-PCR, reverse transcription polymerase chain reaction; qRT-PCR, quantitative reverse transcription polymerase chain reaction; TMA, tissue microarray; TNM, TNM classification of malignant tumors; HNSCC, head and neck squamous cell carcinoma; NSCLC, non-small cell lung carcinoma; AML, acute myeloid leukemia; CLL, chronic lymphocytic leukemia; OSCC, oral squamous cell carcinoma; TSCC, tongue squamous cell carcinoma; HGSC, high-grade serous carcinoma; CTC, circulating tumor cells.
Table 2. Summary of EZR functional assays in cancer cells.
Table 2. Summary of EZR functional assays in cancer cells.
PublicationCancer TypeCell LinesApproachActivityNotes
Li et al. [19]Breast cancerMCF-7, MDA-MB-453, MDA-MB-435s, and MDA-MB-231.shRNAEZR downregulationDecreased EZR expression reversed metastatic behaviors of human breast cancer cells by inducing c-SRC-mediated E-cadherin expression.
Federici et al. [22]Melanoma MM1, MM2, MM3, and HeLa.Stable transfectionEZR deletion mutantExpression of EZR deletion mutant comprising 146 N-terminal amino acids abolished metastatic dissemination.
Zhang et al. [24]MelanomaA375.MiR-183 overexpression and knockdownEZR downregulationmiR-183 inhibits A375 human melanoma cell migration and invasion, possibly through the downregulation of EZR.
Kong et al. [25]Cervical carcinomaHeLa, SiHa, C33A, and CaSki.RNAiEZR downregulation EZR silencing inhibited the proliferation, migration, and invasion of uterine cervical cancer cells through epithelial–mesenchymal transition inhibition.
Li et al. [30]Colorectal cancerSW480 and SW116.siRNA and shRNAEZR downregulationInhibition of EZR reduced EGF-induced epithelial–mesenchymal transition and lung metastasis of colorectal cancer.
Leiphrakpam et al. [3]Colorectal cancerGEO, GEORI, CBS, HCT166, and HCT166b.NSC668394 and NSC305787Pharmacological inhibition of EZR phosphorylationEZR inhibitors were effective in dephosphorylating EZR at T567 and decreased XIAP levels in metastatic colorectal cancer cells.
Ohtani et al. [34].Endometrial cancerIshikawa and mEIIL.ePONsEZR downregulationInhibitory effect of ePONs on invasiveness results from EZR suppression.
Lipreri da Silva et al. [55]Acute myeloid leukemiaKasumi-1 and MOLM-13.NSC305787 and NSC668394Pharmacological inhibition of EZR phosphorylationEZR inhibition reduced cell viability, clonogenicity, phosphorylation of the AKT signaling pathway and increased apoptosis in acute myeloid leukemia cell lines.
Lipreri da Silva et al. [56]Chronic lymphocytic leukemiaMEC-1 and primary patients’ cells.NSC305787Pharmacological inhibition of EZR phosphorylationNSC305787 reduced viability, clonogenicity, and cell cycle progression and induced apoptosis in chronic lymphocytic leukemia cells. Pharmacological EZR inhibition also attenuated ERK, S6RP, and NFκB activation.
Saito et al. [64]Oral squamous cell carcinomaHSC-3.RNAiEZR downregulationEZR depletion significantly reduced cell proliferation, migration, and invasiveness and disturbed actin reorganization during podia formation.
Noi et al. [5]Oral squamous cell carcinomaYes, HSC-3 and HSC-4 (3D culture).siRNAEZR downregulationNo marked morphological differences were observed upon EZR silencing.
Wang et al. [68].Oral squamous cell carcinomaSCC9 and SCC25.siRNA and lentivirus-mediated shRNAEZR downregulationEZR silencing reduced the invasion and migration of SCC9 and SCC25 cells. Downregulation of EZR also inhibited EGF-induced EMT in tongue squamous carcinoma cells.
Song et al. [69]Ovarian cancerSKOV3 and DOV13.Estradiol treatmentEstrogen-induced EZR overexpressionEstrogen induced EZR over-expression and the invasiveness of OVCA cells in culture.
Köbel et al. [70]Ovarian cancerSKOV-3.siRNAEZR downregulationEZR silencing reduced cell invasion in vitro.
Li et al. [71]Ovarian cancerSKOV3 and CaOV3.siRNA and FLAG-EZR overexpression plasmidEZR downregulation and EZR overexpressionEZR ectopic expression increased cell proliferation, invasiveness, and epithelial–mesenchymal transition. EZR knockdown prevented cell proliferation, invasiveness, and epithelial–mesenchymal transition.
Horwitz et al. [72].Ovarian cancerES2 and OVCAR3.Lentivirus-mediated shRNAEZR downregulationReduced EZR expression impaired ovarian cancer cells’ invasion ability and branching capacity.
Zhou et al. [73].Pancreatic cancerSW1990.Recombinant plasmidsEZR overexpressionOverexpression of T567D ezrin (p-EZR), a mutant that mimics permanent phosphorylation, in SW1990 cells increased proliferative capacity and growth rate.
Liprei da Silva et al. [77]Pancreatic cancerPANC-1, AsPC-1, and MIA PaCa-2.NSC305787Pharmacological inhibition of EZR phosphorylationInhibition of EZR favors a molecular network that reduces proliferation and induces apoptosis in pancreatic cancer cells.
Chuan et al. [80]Prostate cancerLNCaP-FGC, PC-3 and PCa.Androgen treatment, siRNA and VSV-G-tagged human wild-type EZR and Y353F-EZR mutantAndrogen-induced EZR expression, EZR downregulation, and EZR overexpressionAndrogen treatment induces EZR expression and phosphorylation of ezrin in T567 and Y353. EZR inhibition function using short interference RNA or the overexpression of T567A and Y353F-EZR mutants significantly reduces androgen-induced Matrigel invasion. Androgens regulate EZR at transcriptional and posttranscriptional levels.
Chen et al. [81]Prostate cancer22RV1 and PC-3.EZR overexpression and si-EZR plasmidsEZR downregulation and EZR overexpressionEZR overexpression promoted the migratory and invasive abilities of 22RV1 and PC-3 cells.
Abbreviations: EZR, ezrin; RNAi, RNA interference; siRNA, small interfering RNA; shRNA, short hairpin RNA; ePONs, antisense phosphorothioate oligonucleotides.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lipreri da Silva, J.C.; Vicari, H.P.; Machado-Neto, J.A. Perspectives for Targeting Ezrin in Cancer Development and Progression. Future Pharmacol. 2023, 3, 61-79. https://doi.org/10.3390/futurepharmacol3010005

AMA Style

Lipreri da Silva JC, Vicari HP, Machado-Neto JA. Perspectives for Targeting Ezrin in Cancer Development and Progression. Future Pharmacology. 2023; 3(1):61-79. https://doi.org/10.3390/futurepharmacol3010005

Chicago/Turabian Style

Lipreri da Silva, Jean Carlos, Hugo Passos Vicari, and João Agostinho Machado-Neto. 2023. "Perspectives for Targeting Ezrin in Cancer Development and Progression" Future Pharmacology 3, no. 1: 61-79. https://doi.org/10.3390/futurepharmacol3010005

Article Metrics

Back to TopTop