Next Article in Journal
Destined to Die: Apoptosis and Pediatric Cancers
Next Article in Special Issue
In Vivo Evidence for Voltage-Gated Sodium Channel Expression in Carcinomas and Potentiation of Metastasis
Previous Article in Journal
RAD52 Functions in Homologous Recombination and Its Importance on Genomic Integrity Maintenance and Cancer Therapy
Previous Article in Special Issue
Size and Shape Filtering of Malignant Cell Clusters within Breast Tumors Identifies Scattered Individual Epithelial Cells as the Most Valuable Histomorphological Clue in the Prognosis of Distant Metastasis Risk
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Pivotal Roles of the Epithelial Membrane Protein Family in Cancer Invasiveness and Metastasis

by
Mohammad Khusni B. Ahmat Amin
1,2,
Akio Shimizu
1 and
Hisakazu Ogita
1,*
1
Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu 520-2192, Japan
2
Translational Research Unit, Department of International Collaborative Research, Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan
*
Author to whom correspondence should be addressed.
Cancers 2019, 11(11), 1620; https://doi.org/10.3390/cancers11111620
Submission received: 2 September 2019 / Revised: 8 October 2019 / Accepted: 21 October 2019 / Published: 23 October 2019
(This article belongs to the Special Issue Cancer Invasion and Metastasis)

Abstract

:
The members of the family of epithelial membrane proteins (EMPs), EMP1, EMP2, and EMP3, possess four putative transmembrane domain structures and are composed of approximately 160 amino acid residues. EMPs are encoded by the growth arrest-specific 3 (GAS3)/peripheral myelin protein 22 kDa (PMP22) gene family. The GAS3/PMP22 family members play roles in cell migration, growth, and differentiation. Evidence indicates an association of these molecules with cancer progression and metastasis. Each EMP has pro- and anti-metastatic functions that are likely involved in the complex mechanisms of cancer progression. We have recently demonstrated that the upregulation of EMP1 expression facilitates cancer cell migration and invasion through the activation of a small GTPase, Rac1. The inoculation of prostate cancer cells overexpressing EMP1 into nude mice leads to metastasis to the lymph nodes and lungs, indicating that EMP1 contributes to metastasis. Pro-metastatic properties of EMP2 and EMP3 have also been proposed. Thus, targeting EMPs may provide new insights into their clinical utility. Here, we highlight the important aspects of EMPs in cancer biology, particularly invasiveness and metastasis, and describe recent therapeutic approaches.

1. Introduction

Cancer metastasis describes the lethal process of the journey of cancer cells from the original site of tumor formation to other major organs, such as the kidney, liver, and lung [1,2]. The metastatic cascade proceeds through systematic steps, including invasion into the adjacent tissues, intravasation, circulation through blood or lymphatic vessels, anchoring at a secondary site, extravasation, and the formation of metastatic lesions in distant organs [3,4]. Supported by Paget’s theory [5], the tumor microenvironment, which harbors components such as cancer-associated fibroblasts (CAFs), myofibroblasts, endothelial cells, immune cells, and adipocytes, greatly influences the phenotypes of the metastatic cells [6,7,8,9]. CAFs reside in the stromal compartment and are associated with certain malignancies [10]. Most recently, the direct interaction of cancer cells with CAFs has been found to enhance the clonogenic growth and migration of pancreatic ductal adenocarcinoma cells [11]. The transition of cancer cells from a quiescent state to uncontrolled growth is driven by alterations of their local environment, which depends on stromal support [12,13]. The cross-talk between invasive and metastatic cancer cells and stromal tissue components is illustrated in Figure 1. Further, the outgrowth of latent breast carcinoma cells into the extracellular matrix (ECM) to colonize the lung is induced by specific ECM components, such as tenascin C and periostin [14,15].
The local environment surrounding the tumor, called the tumor microenvironment, plays an important role in the complex biological process of cancer metastasis. Associated factors include the influence of growth factors and the functional role of stromal fibroblasts, particularly in the reciprocal response to CAFs [16,17]. Each factor fundamentally regulates tumor growth and progression. Moreover, the changes in the expression of metastasis-related genes, which usually occur during the interaction of cancer cells with the tumor microenvironment, become the key regulators of a metastatic event. The interactions promote the aggressive phenotypes of certain cancers, such as those of the bladder, endometrium, lung, urinary bladder, and breast cancer through programming the epithelial-mesenchymal transition (EMT) [18,19,20,21].
We have recently revealed that the expression of a transmembrane protein, epithelial membrane protein 1 (EMP1), is upregulated in co-cultures of human prostate cancer cells and prostate stromal cells [22]. This co-culture system was utilized to mimic a tumor microenvironment. In an animal model, EMP1 significantly enhances the migration of cancer cells and the formation of metastatic lesions in the lymph nodes, lungs, or both. Increased EMP1 expression in patients with prostate cancer occurs at the invasive front and is associated with malignancy. In this context, we highlight in this review the functional aspects of the EMP family members EMP1, EMP2, and EMP3 related to their pathophysiology, particularly where associated with cancer metastasis. EMPs belong to the growth arrest-specific 3 (GAS3)/peripheral myelin protein 22 kDa (PMP22) gene family. The family members possess topologies similar to those of members of the tetraspanin superfamily, also known as the transmembrane 4 superfamily (TM4SF) [23]. TM4SF is comprised of the connexin, tetraspanin, claudin, pannexin, and GAS3/PMP22 families. EMPs lack sequence similarities and key structural features characteristic of the tetraspanins, which contain four to eight cysteine residues in the second extracellular domain to form disulfide bonds with a highly conserved Cys–Cys–Gly motif [24,25]. There is an increasing number of studies focused on EMPs, which have certain physiological functions. Moreover, in the pathological regard, the abnormal expression of EMPs in cancer cells may indicate a connection with cancer progression. Despite insights into the biological and physiological roles of EMPs, detailed knowledge of their effects at the molecular level associated with pathological conditions remains elusive. In the last part of this review, we consider future therapeutic approaches designed to improve the prognosis of cancers according to the molecular properties of EMPs.

2. The Structures, Expressions, and Functions of EMPs

EMPs are hydrophobic proteins of approximately 160 amino acid residues with four predicted transmembrane domains, two extracellular domains, and small intracellular regions (Figure 2). The molecular structures of EMP2 and EMP3 share features such as protein kinase C phosphorylation sites, but EMP1 lacks these sites. EMP1 and EMP2 contain casein kinase-2 phosphorylation sites. All of the EMPs have N-glycosylation sites. The amino acid sequence of human PMP22 is 35%, 39%, and 41% identical to those of human EMP1, EMP2, and EMP3, respectively [26]. PMP22 is mainly expressed in the peripheral nervous system, particularly in Schwann cells [27], and regulates cell proliferation and apoptotic activity [28,29]. Similar to EMPs, the atypical activity of PMP22 is correlated with metastatic progression in certain cancers [30,31,32]. The amino acid sequences of the predicted transmembrane domains are highly conserved among the EMP family members.

2.1. EMP1

Analysis of the HPA, GTEx, and FANTOM5 datasets shows that EMP1 mRNA levels are high in the esophagus, followed by adipose tissue, and the gallbladder [33]. Further, the levels of EMP1 protein are highest in the stomach, but low in other tissues [34].
EMP1 is expressed during the development of the central and peripheral nervous systems, suggesting its association with neurogenesis [35]. Moreover, EMP1 is involved in the regulation of the cell cycle, cell-to-cell interactions, and cell death [26]. During the growth of fibroblasts and Schwann cells, the expression of EMP1 is inversely regulated by PMP22 [34,36], and EMP1 is a novel tight junction protein of the blood-brain barrier [37].

2.2. EMP2

EMP2 is most commonly expressed in the lung, skin, and esophagus and is least likely to be expressed in the pancreas and brain according to the Human Protein Atlas [38]. EMP2 is expressed in multiple structures of the eye, such as the cornea, ciliary body, and the epithelium of retina [39]. The transcript level of EMP2 is elevated in the fetal lung and kidney, but not in the adult thymus and peripheral leukocytes [40].
EMP2 localizes within lipid raft domains and is likely to modulate the plasma membrane trafficking activities of integrins and major histocompatibility complex class 1 proteins [41,42]. EMP2 regulates the physiological function of several integrins during blastocyst implantation, cell division, adhesion, and migration [41,43,44,45]. In a retinal epithelial cell line, EMP2 induces intracellular collagen gel contraction through the activation of focal adhesion kinase (FAK) at two phosphorylation sites Tyr576 and Tyr577 [46]. EMP2 and the activation of FAK are closely associated with integrin β1 subunits [41,47,48]. The enhancement of cellular adhesion to collagens types I and IV, the increase in expression of α-smooth muscle actin, and the activation of F-actin, particularly at the cell periphery, are detected in retinal epithelial cells that overexpress EMP2. These findings indicate that EMP2 may function in the reorganization of the actin cytoskeleton and enhance cellular contractility and adhesion [46].

2.3. EMP3

EMP3 is expressed in numerous organs, as indicated in the Human Protein Atlas [49]. Unlike EMP2, the levels of EMP3 mRNA are high in the adult peripheral leukocytes and relatively strong in the fetal lungs, liver, and kidneys [40]. However, lower levels are expressed in the fetal and adult brain.
Recently, published data have shown that EMP3 plays a role in the immune system. Overexpression of EMP3 in macrophages inhibits CD8+ cytotoxic T lymphocytes (CTLs) [50], which are involved in tumor progression, viral infection, and type IV allergy [51]. In contrast, knockdown of EMP3 expression enhances the induction of CTLs, secretion of interferon-γ, and the expression of IL-2 receptor α by CD8+ T cells. These findings represent the unique function of EMP3 in regulating the immune system via the inhibition of CTL induction.

3. Involvement of EMPs in Cancer Metastasis

After the discovery of the GAS3/PMP22 family members [52,53,54], numerous studies revealed the role of these membrane proteins in certain chronic diseases. These proteins possess pathological properties via interaction with their binding partners such as integrins. Integrins are cell surface receptors composed of α and β subunits, which influence the tumor microenvironment [55]. The EMP family members are expressed and implicated in numerous cancers, indicating their role in oncogenesis and tumor progression. Further, EMPs have become emerging molecules to be associated with cancer invasion and metastasis because of their abilities to regulate various signaling molecules (Figure 2). The interaction with the specific receptors of ECM, including integrins, can regulate a dramatic consequence at the molecular level with respect to tumor progression. Among the EMP family members, EMP2 is expressed in the endometrium as a complex with integrin αvβ3, which resides on the apical side of the luminal epithelium [44,56,57,58]. EMP3 enhances the expression of integrins, particularly α1, α2, α3, α5, α6, αV, and β1, to promote the proliferation and migration of urothelial carcinoma cells. The differential expression of EMPs in cancer cells may be associated with the regulation of certain structural and signaling molecules to perform fundamental effects on the multistep processes of cancer invasion and metastasis. However, the roles of EMPs in invasion and metastasis in diverse cancers are controversial.

3.1. EMP1

3.1.1. Pro-Metastatic Roles of EMP1

EMP1 contributes to the metastasis of glioblastoma multiforme (GBM). The mRNA levels of EMP1 are upregulated in GBM, compared with those in normal brain tissues, which may explain the poor clinical outcome [59,60]. Silencing EMP1 expression in glioblastoma cells inhibits their proliferation and invasiveness, as well as the expression of CD44 and matrix metalloprotease (MMP)-2 [59,60]. CD44 isoforms, which are translated from alternatively spliced mRNAs, stimulate the uncontrolled growth of glioma stem cells [61]. Signaling through the phosphatidyl inositol-3 kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and its downstream molecules is inhibited in the EMP1-ablated cells. Other consequences of the inhibition of dysregulated CD44 by silencing EMP1 expression include the suppression of the activities of transcription factors such as OCT4, SOX2, and Nanog [59]. Moreover, the implantation of EMP1-knockdown P3 glioma cells in athymic nude mice showed remarkably smaller tumor formations than that of control cells. In the mice implanted with EMP1-knockdown cells, the levels of a mitotic marker Ki-67 and MMPs were reduced, and overall survival was increased. Because the clinical outcome of this aggressive malignant brain tumor remains very poor and an estimated relative survival of 5 years after diagnosis is only 5.6% [62], the findings of biomarkers and pro- or anti-metastatic factors for GBM may be highly helpful to develop novel therapeutics and improve its prognosis.
In uveal melanoma, the most common intraocular malignancy, EMP1 mRNA levels are higher in class 2 tumors than in class 1 tumors [63]. A recent study found that treatment with ICG-001, a molecule that can impair the WNT/β-catenin signaling pathway [64], has strong anti-cancer activity against uveal melanoma by inhibiting the expression of EMP1 and the mTOR and mitogen-activated protein kinase (MAPK) signaling pathways [65].
Tumor tissues from non-small cell lung cancer (NSCLC) patients contain higher levels of EMP1 mRNA than the benign tissues [66]. Overexpression of EMP1 in lung cancer cells increases the rate of cell proliferation through the activation of the PI3K/AKT signaling pathway [67]. This also accelerates tumor progression in athymic mice. In addition, EMP1 mRNA expression is correlated with the acquisition of gefitinib resistance in patients with NSCLC, and the correlation is independent of gefitinib-sensitizing mutations in epidermal growth factor receptor [68].
The significance of EMP1 expression has been reported in acute lymphoblastic leukemia (ALL). Increased expression of EMP1 mRNA positively correlates with prednisolone-resistant leukemic cells [69]. Silencing EMP1 expression in prednisolone-resistant leukemic cell lines induces apoptosis, suppresses cell migration and adhesion, decreases the proliferation rate, and sensitizes cells to prednisolone. These phenomena are mediated by impaired activities of Src family kinases (Src, Yes, Fgr, Blk, and Hck) and their downstream effectors such as JNK, STAT3, STAT5, CREB, and NF-κB.

3.1.2. Anti-Metastatic Roles of EMP1

In contrast to the above tumors, the anti-tumor effects of EMP1 are observed in nasopharyngeal cancer, in which EMP1 protein levels are increased in normal tissues compared with the tumor lesions [70]. These findings are supported by observations that patients with the negative or weak expression of EMP1 protein in malignant lesions have lower 5-year survival rates than those with higher EMP1 levels. Overexpression of EMP1 in nasopharyngeal carcinoma cells decreases cell viability, migration and invasion, and induces apoptosis.
EMP1 also suppresses the progression of gastrointestinal cancers. EMP1 mRNA levels are low in 15 pairs of the esophageal cancer lesions compared with the adjacent normal areas, and overexpression of EMP1 in esophageal carcinoma cells inhibits their proliferation [71]. In agreement with this finding, cell cycle arrest at the S and G1 phases is prolonged in EMP1-overexpressed cells. Consistent with the tumor suppressive activity of EMP1 in esophageal cancer, EMP1 protein expression is lower in gastric cancer than in normal tissues [72]. In human colorectal cancer and normal adjacent colorectal tissues, EMP1 protein expression is significantly lower in the former (39.7% tissues positive) than the latter (90.3% tissues positive) [73]. Patients with high levels of EMP1 experience a significantly better survival rate than those with low levels of EMP1. Moreover, overexpression of EMP1 reduces the proliferation rate of colorectal adenocarcinoma cells, induces early apoptosis with the augmented caspase-9 level, and significantly diminishes cell migration and invasion.
EMP1 protein expression is reduced in high-grade serous ovarian cancer, compared with benign ovarian tumors [74]. The research group seems to consider that EMP1 may negatively regulate the progression of ovarian cancer and decrease the severity of the cancer, although the detailed evidence is not provided.

3.1.3. Opposing Roles of EMP1 in Cancer Invasiveness and Metastasis in the Same Type of Cancer

The functions of EMP1 in breast and prostate cancers are reported to be controversial [22,75,76,77]. The reason for the opposing activities of EMP1 in cancer progression in these types of cancer should be determined in the future. Further, the exact mechanism of EMP1-mediated signal transduction pathways in the regulation of cancer metastasis is largely unanswered. EMP1 serves as a reliable marker that distinguishes the two most common histological types of breast cancer: invasive ductal and lobular carcinomas [75]. The mRNA level of EMP1 is significantly higher in the latter (93.1%) than the former (16.3%). Lobular tumors often resist neoadjuvant therapy [78], and patients with these tumors experience a lower survival rate than those with ductal tumors [79,80]. These data may suggest the tumor-progressive function of EMP1. On the other hand, reduced protein expression of EMP1 is associated with a poor survival rate [76], suggesting that EMP1 acts as a tumor suppressor. Overexpression of EMP1 in breast cancer cells inhibits proliferation through reducing the expression of vascular endothelial growth factor (VEGF)-C, and attenuates cell migration and invasion.
Similar to breast cancer, the opposing roles of EMP1 in cancer invasion and metastasis operate in prostate cancer. We have recently found that EMP1 enhances the progression of prostate cancer in vitro and in vivo, and that the mRNA and protein levels of EMP1 are upregulated in co-cultures of human prostate cancer and stromal cells [22]. EMP1 mRNA levels are lower in low grade cancer cell lines. Overexpression of EMP1 in LNCaP prostate cancer cells enhances cell migration and invasion, similar to the findings for MCF7 breast cancer cells and Caco2 colon adenocarcinoma cells. The inoculation of LNCaP cells overexpressing EMP1 in the prostate glands of nude mice promotes tumor metastasis to the lymph nodes and lungs, while control LNCaP cells do not metastasize. The sizes of the primary tumor in the prostate glands of the engrafted mice are identical between control and EMP1-overexpressed LNCaP cells, suggesting that EMP1 does not affect the proliferation of prostate cancer cells. Further, we successfully explored the mechanism of EMP1-induced cancer metastasis. The direct binding of copine-III to the intracellular region of EMP1 is critically involved in cancer invasion. The binding activates the signaling cascade of a tyrosine kinase Src, a Rac guanine nucleotide exchange factor Vav2, and a small GTPase Rac1, leading to enhanced cell migration and invasiveness. The similar effects of EMP1 are also observed in other types of cancer cell lines, MCF7 breast cancer cells and Caco2 colon cancer cells. EMP1 expression is significantly increased in the prostate cancer samples with higher Gleason scores, compared with those with lower scores. Our model of EMP1 function in cancer cells is displayed in Figure 3. In contrast, others uncovered the potential role of EMP1 as a suppressor of prostate cancer [77]. In this study, the levels of EMP1 protein are higher in normal tissues than in prostate tumors. Overexpression of EMP1 in PC3 prostate cancer cells significantly decreases cell migration and invasiveness. Thus, future studies are required to explain the discrepancy of EMP1 functions in breast and prostate cancers.
The summarized information about the pro- and anti-metastatic roles of EMP1 in cancers is shown in Table 1.

3.2. EMP2

3.2.1. Pro-Metastatic Roles of EMP2

Studies on gene expression profiling elucidate that EMP2 mRNA expression is upregulated in GBM [81,82]. The inoculation of GBM cells overexpressing EMP2 into nude mice accelerates tumor formation, mainly through the growing vasculature of intracranial tumors [83,84]. EMP2 and integrin αvβ3 interact in GBM cells to reciprocally stimulate their expression [83], which can augment cell migration and invasion through Src activation. STAT3 also enhances EMP2 expression and induces glioma-initiating cells to cause the progression and recurrence of GBM [85].
Transcriptional profiling analyses detected the dysregulation of EMP2 mRNA expression in breast cancers [86,87,88,89]. EMP2 levels are significantly increased during the progression from non-malignant glandular to ductal carcinoma in situ, invasive ductal carcinoma, and lymph node metastasis [90], suggesting the potential role of EMP2 in the late stage of breast cancer in association with tumor invasion and metastasis [91]. EMP2 mRNA expression might be negatively regulated by breast cancer-related receptors (estrogen receptor, progesterone receptor, and ErbB2), because the expression is relatively high in the triple-negative breast cancer cells [92]. The level of EMP2 protein is positively associated with cancer progression, which is supported by findings that EMP2 is expressed in 67% of lymph node metastases and is linked with lymphovascular invasion [90]. Recently, EMP2 protein seems to have been identified as a novel biomarker to effectively capture circulating tumor cells (CTCs) in blood samples from patients with breast cancer [93]. In addition, CTCs are more effectively detected in the samples from the pulmonary vein than the peripheral vein, even in the early stage of the cancer [94]. Because CTCs play a role in the development of cancer metastasis [95,96], earlier and effective detection of them is important.
In gynecologic cancers, approximately 70% of serous and endometrioid ovarian tumors express higher levels of EMP2 protein [97]. The protein level of EMP2 positively correlates with the progression of endometrial cancer [98,99,100], gradually increasing in the order benign, hyperplasia, atypical hyperplasia, and endometrial adenocarcinoma [101]. EMP2 mediates angiogenesis in endometrial cancer by regulating the expression of VEGF and hypoxia-induced transcription factors-1α through Src [99]. These findings suggest that EMP2 may serve as a potential candidate for pharmacological treatment of gynecologic cancers.

3.2.2. Anti-Metastatic Roles of EMP2

EMP2 possesses anti-tumor properties by suppressing the growth of nasopharyngeal carcinoma cells [102]. EMP2 protein is expressed at low levels or is undetectable in most high-grade tumors (grades III and IV) in nasopharynx cancer. The clinical outcome of nasopharynx cancer is significantly worse for patients who do not detectably express EMP2. Ectopic expression of EMP2 in nasopharynx cancer cells in vitro impairs cell growth, enhances the efficiency of radiotherapy, induces cell cycle arrest at S phase, and increases apoptosis at both early and late stages.
Similar to nasopharyngeal cancer, EMP2 has been discovered to act as a novel biomarker to suppress cutaneous melanoma [103]. The apoptosis in cultured melanoma cells is significantly induced by EMP2. EMP2 also functions as a tumor suppressor in urothelial carcinoma [104]. EMP2 protein expression inversely correlates with the histologic grades of uroepithelial cancer cells, and overexpression of EMP2 impairs cancer cell proliferation and inhibits tumor development.
The summarized information about the pro- and anti-metastatic roles of EMP2 in cancers is shown in Table 2.

3.3. EMP3

3.3.1. Pro-Metastatic Roles of EMP3

EMP3 expression is upregulated to promote the progression of brain tumors [105,106,107], particularly in the mesenchymal subtype [107,108,109]. The expression of EMP3 and CD44 in GBM is relatively parallel [109]. CD44 is a biomarker for the mesenchymal subtype of GBM and is significantly related to unfavorable prognosis of patients with GBM [61,110]. In patients above age 50, high expression of EMP3 mRNA is markedly associated with poor prognosis [108,111]. The overall survival rates of GBM patients with high EMP3 levels are reduced compared with those with low EMP3 levels [112]. Depletion of EMP3 protein attenuates proliferation and colony formation of GBM cells and induces apoptosis [109]. The interaction between EMP3 and transforming growth factor (TGF)-β receptor 2 was discovered in CD44-high GBM cells, leading to the activation of TGF-β receptor/Smad2/3 signaling. The inoculation of EMP3-depleted GBM cells in nude mice showed smaller tumor formation than that of EMP3-overexpressed cells. The expression of caspase-3, a marker of apoptosis, is upregulated in the tumor lysates derived from EMP3-depleted cells.
EMP3 contributes to the malignant phenotype of oral squamous carcinoma [113]. The expression of EMP3 is likely modulated by miR-765 in oral squamous carcinoma cells, and the depletion of EMP3 markedly stimulates the expression of p66Shc to impair the migratory activity of the cells.
Further, overall survival and the progression-free period are very poor in patients with gastric cancer with high levels of EMP3 [114]. EMP3 functions downstream of TWIST1/2 to induce EMT through the regulation of expression of an anti-EMT protein E-cadherin and an EMT marker vimentin. Similar to gastric cancer, EMP3 is highly expressed in HCC and negatively correlates with the degree of tumor differentiation [115]. Knockdown of EMP3 protein in hepatic cancer cells inhibits proliferation by arresting cell cycle at the G1 phase. The knockdown enhances the expression of cyclin-dependent kinase inhibitors p21 and p27, and reduces the levels of cyclins E and D1. The effects of high EMP3 expression on the malignant phenotype of HCC cells are associated with the increment of cell migration and invasion, as well as the proteolytic activities of MMP-9 and urokinase.
EMP3 mRNA is significantly upregulated in lymph node metastases of breast carcinomas, and is related to the levels of ErbB2 expression [116,117]. EMP3 expression in breast cancer cells is negatively regulated by miR-765, which can bind to the 3′-UTR of EMP3 mRNA. Recent bioinformatics analysis indicates that EMP3 contributes to the pathogenesis of endometrial cancer [118]. EMP3 may be associated with estrogen receptor α-mediated signaling pathways during the development of endometrial cancer.
The phenotypes of cells overexpressing EMP3 protein in upper urinary tract urothelial carcinoma are consistent with the conclusion that EMP3 confers the malignant characteristic. EMP3 and ErbB2 levels are high in high-grade urothelial carcinoma cells [119]. Cell proliferation and migratory activity are enhanced in urothelial carcinoma cells overexpressing EMP3, which increases signaling through the PI3K/AKT and FAK/Src pathways, leading to the activation of RhoA and ROCK1/2 and the upregulation of the expression of certain integrins. These findings highlight the importance of the EMP3−ErbB2 association as a potential indicator of progression and metastasis of upper urinary tract urothelial carcinoma.

3.3.2. Anti-Metastatic Roles of EMP3

EMP3 protein expression negatively correlates with malignancy in esophageal squamous cell carcinoma [120]. The overexpression of EMP3 in esophageal cancer cells inhibits their growth and facilitates their death. The expression levels of EMP3 and telomerase reverse transcriptase inversely correlate in several esophageal cancer cell lines and tissue samples. The survival rate of patients with esophageal cancer after recurrence is significantly lower in patients with low levels of EMP3.
Recently, EMP3 has been reported to suppress the progression of gallbladder cancer, and its low expression is strongly associated with poor prognosis [121]. Knockdown of EMP3 protein in gallbladder cancer cells that possess high levels of endogenous EMP3 increases proliferation, while overexpression of EMP3 inhibits migratory activity and promotes high apoptotic activity by increasing the expression of caspase-3 and caspase-9. The inoculation of gallbladder cancer cells overexpressing EMP3 into nude mice induces significantly smaller and lower weight tumors than that of EMP3-depleted cancer cells. The level of miR-663a is augmented in human gallbladder cancer tissues, and knockdown of this miRNA reciprocally increases EMP3 expression. Downregulation of EMP3 in gallbladder cancer activates the MAPK/ERK signaling pathway to promote cancer progression.
EMP3 suppresses NSCLC, consistent with findings that EMP3 protein levels are significantly lower in malignant lesions than in normal tissues [122]. The EMP3 levels in the tumor inversely correlate with the Ki67 expression, suggesting increased cell proliferation in the absence of EMP3 in malignant tumors.
The summarized information about the pro- and anti-metastatic roles of EMP3 in cancers is shown in Table 3.

3.4. PMP22

As described above, both PMP22 and EMPs belong to the GAS3/PMP22 family. Although mutations and loss of functions of PMP22 are related to demyelinating peripheral neuropathies [28,123], PMP22 is also reported to increase the aggressiveness of various types of cancers. According to the immunohistochemical and gene expression analyses of human breast cancer tissue samples, the mRNA level of PMP22 is highly upregulated and contributes to poor overall survival and disease-free survival [32]. The PMP22 mRNA is also increased in pancreatic ductal adenocarcinoma, compared with normal pancreatic tissues [124]. This pathological feature appears to be associated with the early development of malignancies. The ectopic expression of PMP22 mRNA in tumor specimens of patients with gastric cancer is correlated with recurrence after perioperative chemotherapy, suggesting a crucial role of PMP22 in the resistance of chemotherapy [30]. This finding is supported by the experiments using xenograft model mice and PMP22-knockdown gastric cancer cells. The suppression of PMP22 expression in lung cancer cells by miR-29 impairs cell proliferation and invasion [125], and this result may suggest the involvement of PMP22 in progression of lung cancer.

4. Therapeutic Implications of EMPs

4.1. Development of Monoclonal Antibodies

The first therapeutic monoclonal antibody (mAb) rituximab was approved for clinical use in 1997 to treat non-Hodgkin’s lymphoma [126]. Rituximab treatment, which significantly improves the prognosis of patients with lymphoma [127], opened the era of biological cancer therapeutics, as evidenced by the numerous clinically-approved mAbs used to treat diverse cancers [128]. The current development of mAbs that target EMP family members to prevent cancer progression and metastasis is very challenging because of the clinicopathological heterogeneity and biological complexity of cancers. The anti-EMP2 recombinant bivalent antibody fragments (diabodies) reduce the aggressiveness of endometrial cancer cells [129]. These diabodies, KS49 and KS83, recognize human and mouse EMP2 peptides, respectively. Treatment of ovarian and endometrial cancer cells with the anti-EMP2 diabodies inhibits cell growth and increases apoptosis [97,129]. Moreover, treatment of breast cancer cells with anti-EMP2 IgG1 promotes cell death and inhibits cancer cell invasion [100]. In an animal model using BALB/c mice, anti-EMP2 treatment actually reduces the number of metastatic lesions generated by xenografted mammary gland tumor cells that express high levels of EMP2.
Despite current beneficial outcomes of pre-clinical trials in treatment with anti-EMP2, additional studies are required to assess acute and long-term adverse effects. Improvements in the pre-clinical implementation and the delivery method are necessary to optimize the treatment for future clinical use.

4.2. Perturbation of Protein−Protein Interactions

Targeting protein−protein interactions as a therapeutic intervention is very challenging, because the binding interfaces of proteins vary, thereby hampering efforts to predict the consequences of the targeting. Further studies are required to completely identify the effects on signal transduction networks induced by inhibitors of the relevant protein−protein interactions. If such studies are not well performed, the inhibitors may provoke the unexpected disturbance of the intricate balance inside the cell, leading to severe adverse effect.
Examples of studies designed to inhibit protein–protein interactions in order to prevent cancer progression are as follows: (1) An inhibitor of the interactions of bromodomain and extra-terminal family member BRD4 with other proteins in an incurable subtype of human squamous carcinoma [130]; (2) a competitor of the pro-survival protein Bcl-XL [131]; (3) an inhibitor that interferes with binding of MDM2 to P53 [132]; and (4) an inhibitor that blocks the binding of VEGF to its receptor to prevent angiogenesis [133]. Further, as described above, we have found that copine-III binds to EMP1, which enhances signal transduction to influence the invasiveness of cancer cells [22]. Therefore, developing a specific inhibitor to block the binding of copine-III to EMP1 would terminate the invasiveness. The direct interaction between EMP1 and copine-III is mediated through a contiguous stretch of nine amino acid residues within the intracellular region of EMP1. To perturb the interaction, a potential inhibitor must enter the cell, possibly by adding a cell-penetrating sequence of the human immunodeficiency virus TAT protein. This technique was used to inhibit the activation of RhoA to prevent the uncontrollable proliferation of cancer cells via aberrant signaling through the VEGF-A/NRP1 signaling pathway [134].

5. Conclusions

The research reviewed here provides compelling evidence that members of the EMP family mediate cancer progression and metastasis. Further studies are clearly warranted to characterize in detail, the tumor progressive and suppressive activities of EMPs, to design treatment strategies in order to improve the prognosis of patients with cancer. Important examples presented here include the development of anti-EMP monoclonal antibodies and inhibitors of the binding of EMPs to their effectors.

Author Contributions

M.K.B.A.A., A.S., and H.O. wrote and edited the manuscript.

Funding

This research was funded by Grants-in-aid for Scientific Research <KAKENHI> from Japan Society for the Promotion of Science for A.S. and H.O., and Takeda Science Foundation for H.O.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Steeg, P.S. Targeting metastasis. Nat. Rev. Cancer 2016, 16, 201–218. [Google Scholar] [CrossRef] [PubMed]
  2. Mehlen, P.; Puisieux, A. Metastasis: A question of life or death. Nat. Rev. Cancer 2006, 6, 449–458. [Google Scholar] [CrossRef] [PubMed]
  3. Bacac, M.; Stamenkovic, I. Metastatic cancer cell. Annu. Rev. Pathol. 2008, 3, 221–247. [Google Scholar] [CrossRef] [PubMed]
  4. Duffy, M.J.; McGowan, P.M.; Gallagher, W.M. Cancer invasion and metastasis: Changing views. J. Pathol. 2008, 214, 283–293. [Google Scholar] [CrossRef]
  5. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef]
  6. Andarawewa, K.L.; Motrescu, E.R.; Chenard, M.P.; Gansmuller, A.; Stoll, I.; Tomasetto, C.; Rio, M.C. Stromelysin-3 is a potent negative regulator of adipogenesis participating to cancer cell-adipocyte interaction/crosstalk at the tumor invasive front. Cancer Res. 2005, 65, 10862–10871. [Google Scholar] [CrossRef]
  7. Duda, D.G.; Cohen, K.S.; Kozin, S.V.; Perentes, J.Y.; Fukumura, D.; Scadden, D.T.; Jain, R.K. Evidence for incorporation of bone marrow-derived endothelial cells into perfused blood vessels in tumors. Blood 2006, 107, 2774–2776. [Google Scholar] [CrossRef]
  8. Levin, I. Changes in the tissue surrounding a growing tumor and the significance of the “precancerous state”. J. Exp. Med. 1912, 16, 149–154. [Google Scholar] [CrossRef]
  9. Lyden, D.; Hattori, K.; Dias, S.; Costa, C.; Blaikie, P.; Butros, L.; Chadburn, A.; Heissig, B.; Marks, W.; Witte, L.; et al. Impaired recruitment of bone-marrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat. Med. 2001, 7, 1194–1201. [Google Scholar] [CrossRef]
  10. Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
  11. Begum, A.; McMillan, R.H.; Chang, Y.T.; Penchev, V.R.; Rajeshkumar, N.V.; Maitra, A.; Goggins, M.G.; Eshelman, J.R.; Wolfgang, C.L.; Rasheed, Z.A.; et al. Direct interactions with cancer-associated fibroblasts lead to enhanced pancreatic cancer stem cell function. Pancreas 2019, 48, 329–334. [Google Scholar] [CrossRef] [PubMed]
  12. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [PubMed]
  13. Wan, L.; Pantel, K.; Kang, Y. Tumor metastasis: Moving new biological insights into the clinic. Nat. Med. 2013, 19, 1450–1464. [Google Scholar] [CrossRef] [PubMed]
  14. Malanchi, I.; Santamaria-Martínez, A.; Susanto, E.; Peng, H.; Lehr, H.A.; Delaloye, J.F.; Huelsken, J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 2011, 481, 85–89. [Google Scholar] [CrossRef] [PubMed]
  15. Oskarsson, T.; Acharyya, S.; Zhang, X.H.; Vanharanta, S.; Tavazoie, S.F.; Morris, P.G.; Downey, R.J.; Manova-Todorova, K.; Brogi, E.; Massagué, J. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat. Med. 2011, 17, 867–874. [Google Scholar] [CrossRef] [Green Version]
  16. Lau, E.Y.; Lo, J.; Cheng, B.Y.; Ma, M.K.; Lee, J.M.; Ng, J.K.; Chai, S.; Lin, C.H.; Tsang, S.Y.; Ma, S.; et al. Cancer-associated fibroblasts regulate tumor-initiating cell plasticity in hepatocellular carcinoma through c-Met/FRA1/HEY1 signaling. Cell Rep. 2016, 15, 1175–1189. [Google Scholar] [CrossRef]
  17. Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10+ GPR77+ cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 2018, 172, 841–856. [Google Scholar] [CrossRef]
  18. Goulet, C.R.; Champagne, A.; Bernard, G.; Vandal, D.; Chabaud, S.; Pouliot, F.; Bolduc, S. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of bladder cancer cells through paracrine IL-6 signalling. BMC Cancer 2019, 19, 137. [Google Scholar] [CrossRef]
  19. Wang, X.; Zhang, W.; Sun, X.; Lin, Y.; Chen, W. Cancer-associated fibroblasts induce epithelial-mesenchymal transition through secreted cytokines in endometrial cancer cells. Oncol. Lett. 2018, 15, 5694–5702. [Google Scholar] [CrossRef]
  20. Yu, Y.; Xiao, C.H.; Tan, L.D.; Wang, Q.S.; Li, X.Q.; Feng, Y.M. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. Br. J. Cancer 2014, 110, 724–732. [Google Scholar] [CrossRef]
  21. Zhuang, J.; Lu, Q.; Shen, B.; Huang, X.; Shen, L.; Zheng, X.; Huang, R.; Yan, J.; Guo, H. TGFβ1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT. Sci. Rep. 2015, 5, 11924. [Google Scholar] [CrossRef] [PubMed]
  22. Ahmat Amin, M.K.B.; Shimizu, A.; Zankov, D.P.; Sato, A.; Kurita, S.; Ito, M.; Maeda, T.; Yoshida, T.; Sakaue, T.; Higashiyama, S.; et al. Epithelial membrane protein 1 promotes tumor metastasis by enhancing cell migration via copine-III and Rac1. Oncogene 2018, 37, 5416–5434. [Google Scholar] [CrossRef] [PubMed]
  23. Wright, M.D.; Tomlinson, M.G. The ins and outs of the transmembrane 4 superfamily. Immunol. Today 1994, 15, 588–594. [Google Scholar] [CrossRef]
  24. Seigneuret, M.; Delaguillaumie, A.; Lagaudrière-Gesbert, C.; Conjeaud, H. Structure of the tetraspanin main extracellular domain. A partially conserved fold with a structurally variable domain insertion. J. Biol. Chem. 2001, 276, 40055–40064. [Google Scholar] [CrossRef] [PubMed]
  25. Stipp, C.S.; Kolesnikova, T.V.; Hemler, M.E. Functional domains in tetraspanin proteins. Trends Biochem. Sci. 2003, 28, 106–112. [Google Scholar] [CrossRef]
  26. Jetten, A.M.; Suter, U. The peripheral myelin protein 22 and epithelial membrane protein family. Prog. Nucleic Acid Res. Mol. Biol. 2000, 64, 97–129. [Google Scholar] [PubMed] [Green Version]
  27. Suter, U.; Snipes, G.J. Peripheral myelin protein 22: Facts and hypotheses. J. Neurosci. Res. 1995, 40, 145–151. [Google Scholar] [CrossRef] [PubMed]
  28. Fabbretti, E.; Edomi, P.; Brancolini, C.; Schneider, C. Apoptotic phenotype induced by overexpression of wild-type gas3/PMP22: Its relation to the demyelinating peripheral neuropathy CMT1A. Genes Dev. 1995, 9, 1846–1856. [Google Scholar] [CrossRef]
  29. Zoidl, G.; Blass-Kampmann, S.; D’Urso, D.; Schmalenbach, C.; Müller, H.W. Retroviral-mediated gene transfer of the peripheral myelin protein PMP22 in Schwann cells: Modulation of cell growth. EMBO J. 1995, 14, 1122–1128. [Google Scholar] [CrossRef]
  30. Cai, W.; Chen, G.; Luo, Q.; Liu, J.; Guo, X.; Zhang, T.; Ma, F.; Yuan, L.; Li, B.; Cai, J. PMP22 regulates self-renewal and chemoresistance of gastric cancer cells. Mol. Cancer Ther. 2017, 16, 1187–1198. [Google Scholar] [CrossRef]
  31. Liu, S.; Chen, Z. The functional role of PMP22 gene in the proliferation and invasion of osteosarcoma. Med. Sci. Monit. 2015, 21, 1976–1982. [Google Scholar] [PubMed]
  32. Tong, D.; Heinze, G.; Pils, D.; Wolf, A.; Singer, C.F.; Concin, N.; Hofstetter, G.; Schiebel, I.; Rudas, M.; Zeillinger, R. Gene expression of PMP22 is an independent prognostic factor for disease-free and overall survival in breast cancer patients. BMC Cancer 2010, 10, 682. [Google Scholar] [CrossRef] [PubMed]
  33. EMP1 in The Human Protein Atlas. Available online: https://www.proteinatlas.org/ENSG00000134531-EMP1/tissue (accessed on 8 October 2019).
  34. Taylor, V.; Welcher, A.A.; Program, A.E.; Suter, U. Epithelial membrane protein-1, peripheral myelin protein 22, and lens membrane protein 20 define a novel gene family. J. Biol. Chem. 1995, 270, 28824–28833. [Google Scholar] [CrossRef] [PubMed]
  35. Wulf, P.; Suter, U. Embryonic expression of epithelial membrane protein 1 in early neurons. Brain Res. Dev. Brain Res. 1999, 116, 169–180. [Google Scholar] [CrossRef]
  36. Gnirke, A.U.; Weidle, U.H. Investigation of prevalence and regulation of expression of progression associated protein (PAP). Anticancer Res. 1998, 18, 4363–4369. [Google Scholar] [PubMed]
  37. Bangsow, T.; Baumann, E.; Bangsow, C.; Jaeger, M.H.; Pelzer, B.; Gruhn, P.; Wolf, S.; von Melchner, H.; Stanimirovic, D.B. The epithelial membrane protein 1 is a novel tight junction protein of the blood-brain barrier. J. Cereb. Blood Flow Metab. 2008, 28, 1249–1260. [Google Scholar] [CrossRef]
  38. EMP2 in The Human Protein Atlas. Available online: https://www.proteinatlas.org/ENSG00000213853-EMP2/tissue (accessed on 8 October 2019).
  39. Wadehra, M.; Sulur, G.G.; Braun, J.; Gordon, L.K.; Goodglick, L. Epithelial membrane protein-2 is expressed in discrete anatomical regions of the eye. Exp. Mol. Pathol. 2003, 74, 106–112. [Google Scholar] [CrossRef]
  40. Taylor, V.; Suter, U. Epithelial membrane protein-2 and epithelial membrane protein-3: Two novel members of the peripheral myelin protein 22 gene family. Gene 1996, 175, 115–120. [Google Scholar] [CrossRef]
  41. Wadehra, M.; Iyer, R.; Goodglick, L.; Braun, J. The tetraspan protein epithelial membrane protein-2 interacts with β1 integrins and regulates adhesion. J. Biol. Chem. 2002, 277, 41094–41100. [Google Scholar] [CrossRef]
  42. Wadehra, M.; Su, H.; Gordon, L.K.; Goodglick, L.; Braun, J. The tetraspan protein EMP2 increases surface expression of class I major histocompatibility complex proteins and susceptibility to CTL-mediated cell death. Clin. Immunol. 2003, 107, 129–136. [Google Scholar] [CrossRef]
  43. Wadehra, M.; Goodglick, L.; Braun, J. The tetraspan protein EMP2 modulates the surface expression of caveolins and glycosylphosphatidyl inositol-linked proteins. Mol. Biol. Cell 2004, 15, 2073–2083. [Google Scholar] [CrossRef] [PubMed]
  44. Wadehra, M.; Forbes, A.; Pushkarna, N.; Goodglick, L.; Gordon, L.K.; Williams, C.J.; Braun, J. Epithelial membrane protein-2 regulates surface expression of αvβ3 integrin in the endometrium. Dev. Biol. 2005, 287, 336–345. [Google Scholar] [CrossRef] [PubMed]
  45. Wadehra, M.; Dayal, M.; Mainigi, M.; Ord, T.; Iyer, R.; Braun, J.; Williams, C.J. Knockdown of the tetraspan protein epithelial membrane protein-2 inhibits implantation in the mouse. Dev. Biol. 2006, 292, 430–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Morales, S.A.; Mareninov, S.; Wadehra, M.; Zhang, L.; Goodglick, L.; Braun, J.; Gordon, L.K. FAK activation and the role of epithelial membrane protein 2 (EMP2) in collagen gel contraction. Investig. Ophthalmol. Vis. Sci. 2009, 50, 462–469. [Google Scholar] [CrossRef]
  47. Parsons, J.T.; Martin, K.H.; Slack, J.K.; Taylor, J.M.; Weed, S.A. Focal adhesion kinase: A regulator of focal adhesion dynamics and cell movement. Oncogene 2000, 19, 5606–5613. [Google Scholar] [CrossRef]
  48. Schlaepfer, D.D.; Hauck, C.R.; Sieg, D.J. Signaling through focal adhesion kinase. Prog. Biophys. Mol. Biol. 1999, 71, 435–478. [Google Scholar] [CrossRef] [Green Version]
  49. EMP3 in The Human Protein Atlas. Available online: https://www.proteinatlas.org/ENSG00000142227-EMP3/tissue (accessed on 8 October 2019).
  50. Kusumoto, Y.; Okuyama, H.; Shibata, T.; Konno, K.; Takemoto, Y.; Maekawa, D.; Kononaga, T.; Ishii, T.; Akashi-Takamura, S.; Saitoh, S.I.; et al. Epithelial membrane protein 3 (Emp3) downregulates induction and function of cytotoxic T lymphocytes by macrophages via TNF-α production. Cell. Immunol. 2018, 324, 33–41. [Google Scholar] [CrossRef]
  51. Smith-Garvin, J.E.; Koretzky, G.A.; Jordan, M.S. T cell activation. Annu. Rev. Immunol. 2009, 27, 591–619. [Google Scholar] [CrossRef]
  52. Hotta, H.; Ross, A.H.; Huebner, K.; Isobe, M.; Wendeborn, S.; Chao, M.V.; Ricciardi, R.P.; Tsujimoto, Y.; Croce, C.M.; Koprowski, H. Molecular cloning and characterization of an antigen associated with early stages of melanoma tumor progression. Cancer Res. 1988, 48, 2955–2962. [Google Scholar]
  53. Oren, R.; Takahashi, S.; Doss, C.; Levy, R.; Levy, S. TAPA-1, the target of an antiproliferative antibody, defines a new family of transmembrane proteins. Mol. Cell. Biol. 1990, 10, 4007–4015. [Google Scholar] [CrossRef]
  54. Wright, M.D.; Henkle, K.J.; Mitchell, G.F. An immunogenic Mr 23,000 integral membrane protein of Schistosoma mansoni worms that closely resembles a human tumor-associated antigen. J. Immunol. 1990, 144, 3195–3200. [Google Scholar] [PubMed]
  55. Cabodi, S.; Di Stefano, P.; Leal, M.P.; Tinnirello, A.; Bisaro, B.; Morello, V.; Damiano, L.; Aramu, S.; Repetto, D.; Tornillo, G.; et al. Integrins and signal transduction. Adv. Exp. Med. Biol. 2010, 674, 43–54. [Google Scholar] [PubMed]
  56. Basak, S.; Dhar, R.; Das, C. Steroids modulate the expression of α4 integrin in mouse blastocysts and uterus during implantation. Biol. Reprod. 2002, 66, 1784–1789. [Google Scholar] [CrossRef] [PubMed]
  57. Lessey, B.A.; Castelbaum, A.J.; Wolf, L.; Greene, W.; Paulson, M.; Meyer, W.R.; Fritz, M.A. Use of integrins to date the endometrium. Fertil. Steril. 2000, 73, 779–787. [Google Scholar] [CrossRef]
  58. Lessey, B.A.; Gui, Y.; Apparao, K.B.; Young, S.L.; Mulholland, J. Regulated expression of heparin-binding EGF-like growth factor (HB-EGF) in the human endometrium: A potential paracrine role during implantation. Mol. Reprod. Dev. 2002, 62, 446–455. [Google Scholar] [CrossRef]
  59. Miao, L.; Jiang, Z.; Wang, J.; Yang, N.; Qi, Q.; Zhou, W.; Feng, Z.; Li, W.; Zhang, Q.; Huang, B.; et al. Epithelial membrane protein 1 promotes glioblastoma progression through the PI3K/AKT/mTOR signaling pathway. Oncol. Rep. 2019, 42, 605–614. [Google Scholar] [CrossRef]
  60. Wang, J.; Li, X.; Wu, H.; Wang, H.; Yao, L.; Deng, Z.; Zhou, Y. EMP1 regulates cell proliferation, migration, and stemness in gliomas through PI3K-AKT signaling and CD44. J. Cell. Biochem. 2019, 120, 17142–17150. [Google Scholar] [CrossRef]
  61. Wang, H.H.; Liao, C.C.; Chow, N.H.; Huang, L.L.; Chuang, J.I.; Wei, K.C.; Shin, J.W. Whether CD44 is an applicable marker for glioma stem cells. Am. J. Transl. Res. 2017, 9, 4785–4806. [Google Scholar]
  62. Ostrom, Q.T.; Gittleman, H.; Truitt, G.; Boscia, A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2011-2015. Neuro Oncol. 2018, 20, iv1–iv86. [Google Scholar] [CrossRef]
  63. Onken, M.D.; Ehlers, J.P.; Worley, L.A.; Makita, J.; Yokota, Y.; Harbour, J.W. Functional gene expression analysis uncovers phenotypic switch in aggressive uveal melanomas. Cancer Res. 2006, 66, 4602–4609. [Google Scholar] [CrossRef]
  64. Emami, K.H.; Nguyen, C.; Ma, H.; Kim, D.H.; Jeong, K.W.; Eguchi, M.; Moon, R.T.; Teo, J.L.; Oh, S.W.; Kim, H.Y.; et al. A small molecule inhibitor of β-catenin/CREB-binding protein transcription [corrected]. Proc. Natl. Acad. Sci. USA 2004, 101, 12682–12687. [Google Scholar] [CrossRef] [PubMed]
  65. Kaochar, S.; Dong, J.; Torres, M.; Rajapakshe, K.; Nikolos, F.; Davis, C.M.; Ehli, E.A.; Coarfa, C.; Mitsiades, N.; Poulaki, V. ICG-001 exerts potent anticancer activity against uveal melanoma cells. Investig. Ophthalmol. Vis. Sci. 2018, 59, 132–143. [Google Scholar] [CrossRef] [PubMed]
  66. Lai, S.; Wang, G.; Cao, X.; Li, Z.; Hu, J.; Wang, J. EMP-1 promotes tumorigenesis of NSCLC through PI3K/AKT pathway. J. Huazhong Univ. Sci. Technolog. Med. Sci. 2012, 32, 834–838. [Google Scholar] [CrossRef] [PubMed]
  67. De Marco, C.; Laudanna, C.; Rinaldo, N.; Oliveira, D.M.; Ravo, M.; Weisz, A.; Ceccarelli, M.; Caira, E.; Rizzuto, A.; Zoppoli, P.; et al. Specific gene expression signatures induced by the multiple oncogenic alterations that occur within the PTEN/PI3K/AKT pathway in lung cancer. PLoS ONE 2017, 12, e0178865. [Google Scholar] [CrossRef]
  68. Jain, A.; Tindell, C.A.; Laux, I.; Hunter, J.B.; Curran, J.; Galkin, A.; Afar, D.E.; Aronson, N.; Shak, S.; Natale, R.B.; et al. Epithelial membrane protein-1 is a biomarker of gefitinib resistance. Proc. Natl. Acad. Sci. USA 2005, 102, 11858–11863. [Google Scholar] [CrossRef] [Green Version]
  69. Ariës, I.M.; Jerchel, I.S.; van den Dungen, R.E.; van den Berk, L.C.; Boer, J.M.; Horstmann, M.A.; Escherich, G.; Pieters, R.; den Boer, M.L. EMP1, a novel poor prognostic factor in pediatric leukemia regulates prednisolone resistance, cell proliferation, migration and adhesion. Leukemia 2014, 28, 1828–1837. [Google Scholar] [CrossRef]
  70. Sun, G.G.; Lu, Y.F.; Fu, Z.Z.; Cheng, Y.J.; Hu, W.N. EMP1 inhibits nasopharyngeal cancer cell growth and metastasis through induction apoptosis and angiogenesis. Tumour Biol. 2014, 35, 3185–3193. [Google Scholar] [CrossRef]
  71. Wang, H.T.; Kong, J.P.; Ding, F.; Wang, X.Q.; Wang, M.R.; Liu, L.X.; Wu, M.; Liu, Z.H. Analysis of gene expression profile induced by EMP-1 in esophageal cancer cells using cDNA Microarray. World J. Gastroenterol. 2003, 9, 392–398. [Google Scholar] [CrossRef]
  72. Sun, G.; Zhao, G.; Lu, Y.; Wang, Y.; Yang, C. Association of EMP1 with gastric carcinoma invasion, survival and prognosis. Int. J. Oncol. 2014, 45, 1091–1098. [Google Scholar] [CrossRef] [Green Version]
  73. Sun, G.G.; Wang, Y.D.; Cui, D.W.; Cheng, Y.J.; Hu, W.N. Epithelial membrane protein 1 negatively regulates cell growth and metastasis in colorectal carcinoma. World J. Gastroenterol. 2014, 20, 4001–4010. [Google Scholar] [CrossRef]
  74. Demirag, G.G.; Kefeli, M.; Kemal, Y.; Yucel, I. Epithelial membrane protein 1 expression in ovarian serous tumors. Oncol. Lett. 2016, 11, 2140–2144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Turashvili, G.; Bouchal, J.; Baumforth, K.; Wei, W.; Dziechciarkova, M.; Ehrmann, J.; Klein, J.; Fridman, E.; Skarda, J.; Srovnal, J.; et al. Novel markers for differentiation of lobular and ductal invasive breast carcinomas by laser microdissection and microarray analysis. BMC Cancer 2007, 7, 55. [Google Scholar] [CrossRef] [PubMed]
  76. Sun, G.G.; Wang, Y.D.; Lu, Y.F.; Hu, W.N. EMP1, a member of a new family of antiproliferative genes in breast carcinoma. Tumour Biol. 2014, 35, 3347–3354. [Google Scholar] [CrossRef] [PubMed]
  77. Sun, G.G.; Wang, Y.D.; Cui, D.W.; Cheng, Y.J.; Hu, W.N. EMP1 regulates caspase-9 and VEGFC expression and suppresses prostate cancer cell proliferation and invasion. Tumour Biol. 2014, 35, 3455–3462. [Google Scholar] [CrossRef]
  78. Newman, L.A.; Buzdar, A.U.; Singletary, S.E.; Kuerer, H.M.; Buchholz, T.; Ames, F.C.; Ross, M.I.; Hunt, K.K. A prospective trial of preoperative chemotherapy in resectable breast cancer: Predictors of breast-conservation therapy feasibility. Ann. Surg. Oncol. 2002, 9, 228–234. [Google Scholar] [CrossRef]
  79. Arpino, G.; Bardou, V.J.; Clark, G.M.; Elledge, R.M. Infiltrating lobular carcinoma of the breast: Tumor characteristics and clinical outcome. Breast Cancer Res. 2004, 6, R149–R156. [Google Scholar] [CrossRef]
  80. Mersin, H.; Yildirim, E.; Gülben, K.; Berberoğlu, U. Is invasive lobular carcinoma different from invasive ductal carcinoma. Eur. J. Surg. Oncol. 2003, 29, 390–395. [Google Scholar] [CrossRef]
  81. Chung, L.K.; Pelargos, P.E.; Chan, A.M.; Demos, J.V.; Lagman, C.; Sheppard, J.P.; Nguyen, T.; Chang, Y.L.; Hojat, S.A.; Prins, R.M.; et al. Tissue microarray analysis for epithelial membrane protein-2 as a novel biomarker for gliomas. Brain Tumor Pathol. 2018, 35, 1–9. [Google Scholar] [CrossRef]
  82. Freije, W.A.; Castro-Vargas, F.E.; Fang, Z.; Horvath, S.; Cloughesy, T.; Liau, L.M.; Mischel, P.S.; Nelson, S.F. Gene expression profiling of gliomas strongly predicts survival. Cancer Res. 2004, 64, 6503–6510. [Google Scholar] [CrossRef]
  83. Qin, Y.; Fu, M.; Takahashi, M.; Iwanami, A.; Kuga, D.; Rao, R.G.; Sudhakar, D.; Huang, T.; Kiyohara, M.; Torres, K.; et al. Epithelial membrane protein-2 (EMP2) activates Src protein and is a novel therapeutic target for glioblastoma. J. Biol. Chem. 2014, 289, 13974–13985. [Google Scholar] [CrossRef]
  84. Qin, Y.; Takahashi, M.; Sheets, K.; Soto, H.; Tsui, J.; Pelargos, P.; Antonios, J.P.; Kasahara, N.; Yang, I.; Prins, R.M.; et al. Epithelial membrane protein-2 (EMP2) promotes angiogenesis in glioblastoma multiforme. J. Neurooncol. 2017, 134, 29–40. [Google Scholar] [CrossRef] [PubMed]
  85. Ganguly, D.; Fan, M.; Yang, C.H.; Zbytek, B.; Finkelstein, D.; Roussel, M.F.; Pfeffer, L.M. The critical role that STAT3 plays in glioma-initiating cells: STAT3 addiction in glioma. Oncotarget 2018, 9, 22095–22112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Kao, J.; Salari, K.; Bocanegra, M.; Choi, Y.L.; Girard, L.; Gandhi, J.; Kwei, K.A.; Hernandez-Boussard, T.; Wang, P.; Gazdar, A.F.; et al. Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery. PLoS ONE 2009, 4, e6146. [Google Scholar] [CrossRef] [PubMed]
  87. Li, Y.; Wang, Y. Bioinformatics analysis of gene expression data for the identification of critical genes in breast invasive carcinoma. Mol. Med. Rep. 2017, 16, 8657–8664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Ma, X.J.; Dahiya, S.; Richardson, E.; Erlander, M.; Sgroi, D.C. Gene expression profiling of the tumor microenvironment during breast cancer progression. Breast Cancer Res. 2009, 11, R7. [Google Scholar] [CrossRef]
  89. Watson, M.A.; Ylagan, L.R.; Trinkaus, K.M.; Gillanders, W.E.; Naughton, M.J.; Weilbaecher, K.N.; Fleming, T.P.; Aft, R.L. Isolation and molecular profiling of bone marrow micrometastases identifies TWIST1 as a marker of early tumor relapse in breast cancer patients. Clin. Cancer Res. 2007, 13, 5001–5009. [Google Scholar] [CrossRef]
  90. Fu, M.; Maresh, E.L.; Helguera, G.F.; Kiyohara, M.; Qin, Y.; Ashki, N.; Daniels-Wells, T.R.; Aziz, N.; Gordon, L.K.; Braun, J.; et al. Rationale and preclinical efficacy of a novel anti-EMP2 antibody for the treatment of invasive breast cancer. Mol. Cancer Ther. 2014, 13, 902–915. [Google Scholar] [CrossRef]
  91. Obermayr, E.; Sanchez-Cabo, F.; Tea, M.K.; Singer, C.F.; Krainer, M.; Fischer, M.B.; Sehouli, J.; Reinthaller, A.; Horvat, R.; Heinze, G.; et al. Assessment of a six gene panel for the molecular detection of circulating tumor cells in the blood of female cancer patients. BMC Cancer 2010, 10, 666. [Google Scholar] [CrossRef]
  92. Christgen, M.; Geffers, R.; Kreipe, H.; Lehmann, U. IPH-926 lobular breast cancer cells are triple-negative but their microarray profile uncovers a luminal subtype. Cancer Sci. 2013, 104, 1726–1730. [Google Scholar] [CrossRef] [Green Version]
  93. Chen, Q.; Yao, L.; Burner, D.; Minev, B.; Lu, L.; Wang, M.; Ma, W. Epithelial membrane protein 2: A novel biomarker for circulating tumor cell recovery in breast cancer. Clin. Transl. Oncol. 2019, 21, 433–442. [Google Scholar] [CrossRef]
  94. Murlidhar, V.; Reddy, R.M.; Fouladdel, S.; Zhao, L.; Ishikawa, M.K.; Grabauskiene, S.; Zhang, Z.; Lin, J.; Chang, A.C.; Carrott, P.; et al. Poor prognosis indicated by venous circulating tumor cell clusters in early-stage lung cancers. Cancer Res. 2017, 77, 5194–5206. [Google Scholar] [CrossRef] [PubMed]
  95. Dasgupta, A.; Lim, A.R.; Ghajar, C.M. Circulating and disseminated tumor cells: Harbingers or initiators of metastasis. Mol. Oncol. 2017, 11, 40–61. [Google Scholar] [CrossRef] [PubMed]
  96. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [PubMed]
  97. Fu, M.; Maresh, E.L.; Soslow, R.A.; Alavi, M.; Mah, V.; Zhou, Q.; Iasonos, A.; Goodglick, L.; Gordon, L.K.; Braun, J.; et al. Epithelial membrane protein-2 is a novel therapeutic target in ovarian cancer. Clin. Cancer Res. 2010, 16, 3954–3963. [Google Scholar] [CrossRef] [PubMed]
  98. Fu, M.; Rao, R.; Sudhakar, D.; Hogue, C.P.; Rutta, Z.; Morales, S.; Gordon, L.K.; Braun, J.; Goodglick, L.; Wadehra, M. Epithelial membrane protein-2 promotes endometrial tumor formation through activation of FAK and Src. PLoS ONE 2011, 6, e19945. [Google Scholar] [CrossRef] [PubMed]
  99. Gordon, L.K.; Kiyohara, M.; Fu, M.; Braun, J.; Dhawan, P.; Chan, A.; Goodglick, L.; Wadehra, M. EMP2 regulates angiogenesis in endometrial cancer cells through induction of VEGF. Oncogene 2013, 32, 5369–5376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Kiyohara, M.H.; Dillard, C.; Tsui, J.; Kim, S.R.; Lu, J.; Sachdev, D.; Goodglick, L.; Tong, M.; Torous, V.F.; Aryasomayajula, C.; et al. EMP2 is a novel therapeutic target for endometrial cancer stem cells. Oncogene 2017, 36, 5793–5807. [Google Scholar] [CrossRef] [Green Version]
  101. Habeeb, O.; Goodglick, L.; Soslow, R.A.; Rao, R.G.; Gordon, L.K.; Schirripa, O.; Horvath, S.; Braun, J.; Seligson, D.B.; Wadehra, M. Epithelial membrane protein-2 expression is an early predictor of endometrial cancer development. Cancer 2010, 116, 4718–4726. [Google Scholar] [CrossRef] [Green Version]
  102. Tang, M.; Liu, R.Y.; Zhou, C.; Yuan, M.Z.; Wu, D.M.; Yuan, Z.; Zhang, P.; Lang, J.Y. EMP2 re-expression inhibits growth and enhances radiosensitivity in nasopharyngeal carcinoma. Tumour Biol. 2017, 39, 1010428317695972. [Google Scholar] [CrossRef]
  103. Wang, M.; Li, S.; Zhang, P.; Wang, Y.; Wang, C.; Bai, D.; Jiang, X. EMP2 acts as a suppressor of melanoma and is negatively regulated by mTOR-mediated autophagy. J. Cancer 2019, 10, 3582–3592. [Google Scholar] [CrossRef] [Green Version]
  104. Wang, Y.W.; Li, W.M.; Wu, W.J.; Chai, C.Y.; Chang, T.Y.; Sun, Y.; Cheng, C.J.; Shiue, Y.L.; Su, S.J.; Cheng, H.L.; et al. Epithelial membrane protein 2 is a prognostic indictor for patients with urothelial carcinoma of the upper urinary tract. Am. J. Pathol. 2013, 183, 709–719. [Google Scholar] [CrossRef] [PubMed]
  105. Scrideli, C.A.; Carlotti, C.G.; Okamoto, O.K.; Andrade, V.S.; Cortez, M.A.; Motta, F.J.; Lucio-Eterovic, A.K.; Neder, L.; Rosemberg, S.; Oba-Shinjo, S.M.; et al. Gene expression profile analysis of primary glioblastomas and non-neoplastic brain tissue: Identification of potential target genes by oligonucleotide microarray and real-time quantitative PCR. J. Neurooncol. 2008, 88, 281–291. [Google Scholar] [CrossRef] [PubMed]
  106. Shu, C.; Wang, Q.; Yan, X.; Wang, J. Whole-genome expression microarray combined with machine learning to identify prognostic biomarkers for high-grade glioma. J. Mol. Neurosci. 2018, 64, 491–500. [Google Scholar] [CrossRef] [PubMed]
  107. Yue, H.; Xu, Q.; Xie, S. High EMP3 expression might independently predict poor overall survival in glioblastoma and its expression is related to DNA methylation. Medicine 2018, 97, e9538. [Google Scholar] [CrossRef]
  108. Gao, Y.F.; Zhu, T.; Mao, C.X.; Liu, Z.X.; Wang, Z.B.; Mao, X.Y.; Li, L.; Yin, J.Y.; Zhou, H.H.; Liu, Z.Q. PPIC, EMP3 and CHI3L1 are novel prognostic markers for high grade glioma. Int. J. Mol. Sci. 2016, 17, 1808. [Google Scholar] [CrossRef]
  109. Jun, F.; Hong, J.; Liu, Q.; Guo, Y.; Liao, Y.; Huang, J.; Wen, S.; Shen, L. Epithelial membrane protein 3 regulates TGF-β signaling activation in CD44-high glioblastoma. Oncotarget 2017, 8, 14343–14358. [Google Scholar] [CrossRef]
  110. Bhat, K.P.L.; Balasubramaniyan, V.; Vaillant, B.; Ezhilarasan, R.; Hummelink, K.; Hollingsworth, F.; Wani, K.; Heathcock, L.; James, J.D.; Goodman, L.D.; et al. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell 2013, 24, 331–346. [Google Scholar] [CrossRef]
  111. Guo, X.X.; Su, J.; He, X.F. A 4-gene panel predicting the survival of patients with glioblastoma. J. Cell. Biochem. 2019, 120, 16037–16043. [Google Scholar] [CrossRef]
  112. Ernst, A.; Hofmann, S.; Ahmadi, R.; Becker, N.; Korshunov, A.; Engel, F.; Hartmann, C.; Felsberg, J.; Sabel, M.; Peterziel, H.; et al. Genomic and expression profiling of glioblastoma stem cell-like spheroid cultures identifies novel tumor-relevant genes associated with survival. Clin. Cancer Res. 2009, 15, 6541–6550. [Google Scholar] [CrossRef]
  113. Zheng, Z.; Luan, X.; Zha, J.; Li, Z.; Wu, L.; Yan, Y.; Wang, H.; Hou, D.; Huang, L.; Huang, F.; et al. TNF-α inhibits the migration of oral squamous cancer cells mediated by miR-765-EMP3-p66Shc axis. Cell. Signal. 2017, 34, 102–109. [Google Scholar] [CrossRef]
  114. Han, M.; Xu, W. EMP3 is induced by TWIST1/2 and regulates epithelial-to-mesenchymal transition of gastric cancer cells. Tumour Biol. 2017, 39, 1010428317718404. [Google Scholar] [CrossRef] [PubMed]
  115. Hsieh, Y.H.; Hsieh, S.C.; Lee, C.H.; Yang, S.F.; Cheng, C.W.; Tang, M.J.; Lin, C.L.; Lin, C.L.; Chou, R.H. Targeting EMP3 suppresses proliferation and invasion of hepatocellular carcinoma cells through inactivation of PI3K/Akt pathway. Oncotarget 2015, 6, 34859–34874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Hong, X.C.; Fen, Y.J.; Yan, G.C.; Hong, H.; Yan, C.H.; Bing, L.W.; Zhong, Y.H. Epithelial membrane protein 3 functions as an oncogene and is regulated by microRNA-765 in primary breast carcinoma. Mol. Med. Rep. 2015, 12, 6445–6450. [Google Scholar] [CrossRef] [PubMed]
  117. Zhou, W.; Jiang, Z.; Li, X.; Xu, F.; Liu, Y.; Wen, P.; Kong, L.; Hou, M.; Yu, J. EMP3 overexpression in primary breast carcinomas is not associated with epigenetic aberrations. J. Korean Med. Sci. 2009, 24, 97–103. [Google Scholar] [CrossRef] [PubMed]
  118. Liu, A.; Zhang, D.; Yang, X.; Song, Y. Estrogen receptor α activates MAPK signaling pathway to promote the development of endometrial cancer. J. Cell. Biochem. 2019, 120, 17593–17601. [Google Scholar] [CrossRef]
  119. Wang, Y.W.; Li, W.M.; Wu, W.J.; Chai, C.Y.; Liu, H.S.; Lai, M.D.; Chow, N.H. Potential significance of EMP3 in patients with upper urinary tract urothelial carcinoma: Crosstalk with ErbB2-PI3K-Akt pathway. J. Urol. 2014, 192, 242–251. [Google Scholar] [CrossRef]
  120. Fumoto, S.; Hiyama, K.; Tanimoto, K.; Noguchi, T.; Hihara, J.; Hiyama, E.; Noguchi, T.; Nishiyama, M. EMP3 as a tumor suppressor gene for esophageal squamous cell carcinoma. Cancer Lett. 2009, 274, 25–32. [Google Scholar] [CrossRef]
  121. Ma, Q.; Zhang, Y.; Liang, H.; Zhang, F.; Liu, F.; Chen, S.; Hu, Y.; Jiang, L.; Hao, Y.; Li, M.; et al. EMP3, which is regulated by miR-663a, suppresses gallbladder cancer progression via interference with the MAPK/ERK pathway. Cancer Lett. 2018, 430, 97–108. [Google Scholar] [CrossRef]
  122. Xue, Q.; Zhou, Y.; Wan, C.; Lv, L.; Chen, B.; Cao, X.; Ju, G.; Huang, Y.; Ni, R.; Mao, G. Epithelial membrane protein 3 is frequently shown as promoter methylation and functions as a tumor suppressor gene in non-small cell lung cancer. Exp. Mol. Pathol. 2013, 95, 313–318. [Google Scholar] [CrossRef]
  123. Pareek, S.; Notterpek, L.; Snipes, G.J.; Naef, R.; Sossin, W.; Laliberté, J.; Iacampo, S.; Suter, U.; Shooter, E.M.; Murphy, R.A. Neurons promote the translocation of peripheral myelin protein 22 into myelin. J. Neurosci 1997, 17, 7754–7762. [Google Scholar] [CrossRef]
  124. Li, J.; Kleeff, J.; Esposito, I.; Kayed, H.; Felix, K.; Giese, T.; Büchler, M.W.; Friess, H. Expression analysis of PMP22/Gas3 in premalignant and malignant pancreatic lesions. J. Histochem. Cytochem. 2005, 53, 885–893. [Google Scholar] [CrossRef] [PubMed]
  125. Qu, H.; Zhu, M.; Tao, Y.; Zhao, Y. Suppression of peripheral myelin protein 22 (PMP22) expression by miR29 inhibits the progression of lung cancer. Neoplasma 2015, 62, 881–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Coulson, A.; Levy, A.; Gossell-Williams, M. Monoclonal antibodies in cancer therapy: Mechanisms, successes and limitations. West. Indian Med. J. 2014, 63, 650–654. [Google Scholar] [PubMed]
  127. Salles, G.; Barrett, M.; Foà, R.; Maurer, J.; O’Brien, S.; Valente, N.; Wenger, M.; Maloney, D.G. Rituximab in B-cell hematologic malignancies: A review of 20 years of clinical experience. Adv. Ther. 2017, 34, 2232–2273. [Google Scholar] [CrossRef]
  128. Strohl, W.R. Current progress in innovative engineered antibodies. Protein Cell 2018, 9, 86–120. [Google Scholar] [CrossRef]
  129. Shimazaki, K.; Lepin, E.J.; Wei, B.; Nagy, A.K.; Coulam, C.P.; Mareninov, S.; Fu, M.; Wu, A.M.; Marks, J.D.; Braun, J.; et al. Diabodies targeting epithelial membrane protein 2 reduce tumorigenicity of human endometrial cancer cell lines. Clin. Cancer Res. 2008, 14, 7367–7377. [Google Scholar] [CrossRef]
  130. Filippakopoulos, P.; Qi, J.; Picaud, S.; Shen, Y.; Smith, W.B.; Fedorov, O.; Morse, E.M.; Keates, T.; Hickman, T.T.; Felletar, I.; et al. Selective inhibition of BET bromodomains. Nature 2010, 468, 1067–1073. [Google Scholar] [CrossRef] [Green Version]
  131. Smith, B.J.; Lee, E.F.; Checco, J.W.; Evangelista, M.; Gellman, S.H.; Fairlie, W.D. Structure-guided rational design of α/β-peptide foldamers with high affinity for BCL-2 family prosurvival proteins. ChemBioChem 2013, 14, 1564–1572. [Google Scholar] [CrossRef]
  132. Zhao, Y.; Aguilar, A.; Bernard, D.; Wang, S. Small-molecule inhibitors of the MDM2−p53 protein−protein interaction (MDM2 inhibitors) in clinical trials for cancer treatment. J. Med. Chem. 2015, 58, 1038–1052. [Google Scholar] [CrossRef]
  133. Haase, H.S.; Peterson-Kaufman, K.J.; Lan Levengood, S.K.; Checco, J.W.; Murphy, W.L.; Gellman, S.H. Extending foldamer design beyond α-helix mimicry: α/β-peptide inhibitors of vascular endothelial growth factor signaling. J. Am. Chem. Soc. 2012, 134, 7652–7655. [Google Scholar] [CrossRef]
  134. Yoshida, A.; Shimizu, A.; Asano, H.; Kadonosono, T.; Kondoh, S.K.; Geretti, E.; Mammoto, A.; Klagsbrun, M.; Seo, M.K. VEGF-A/NRP1 stimulates GIPC1 and Syx complex formation to promote RhoA activation and proliferation in skin cancer cells. Biol. Open 2015, 4, 1063–1076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of the tumor microenvironment in cancer progression. When cancer cells derived from the original lesion travel across the basement membrane and invade the surrounding stromal tissue, they can interact with cells such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages, and myofibroblasts to generate the tumor microenvironment. The characteristics of cancer cells are often affected by the tumor microenvironment, which increases the expression of cell surface proteins, including integrins and epithelial membrane proteins (EMPs), and several soluble factors, such as VEGF. These molecules associate with each other and may also regulate the cancer phenotype, such as invasiveness and angiogenic ability. Thus, understanding the tumor microenvironment through studies of cell surface molecules and their related molecules will likely contribute to the development of strategies designed to target cancer cells and to develop novel anti-cancer therapies.
Figure 1. Schematic representation of the tumor microenvironment in cancer progression. When cancer cells derived from the original lesion travel across the basement membrane and invade the surrounding stromal tissue, they can interact with cells such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages, and myofibroblasts to generate the tumor microenvironment. The characteristics of cancer cells are often affected by the tumor microenvironment, which increases the expression of cell surface proteins, including integrins and epithelial membrane proteins (EMPs), and several soluble factors, such as VEGF. These molecules associate with each other and may also regulate the cancer phenotype, such as invasiveness and angiogenic ability. Thus, understanding the tumor microenvironment through studies of cell surface molecules and their related molecules will likely contribute to the development of strategies designed to target cancer cells and to develop novel anti-cancer therapies.
Cancers 11 01620 g001
Figure 2. Structural difference between EMPs, and signal transduction molecules of which expression and/or activation are affected by EMPs in the regulation of cancer invasion and metastasis. The number and position of N-glycosylation and the kinase-mediated phosphorylation are different between EMPs. Numerous signaling molecules and cell surface proteins are associated with the role of EMPs. They mediate the EMP-induced cellular functions, such as proliferation and migration, that regulate the invasiveness of cancer cells and subsequent metastatic events. Red arrows: increase or activation of the molecules or phenomena; blue arrows: decrease or inactivation of the molecules or phenomena.
Figure 2. Structural difference between EMPs, and signal transduction molecules of which expression and/or activation are affected by EMPs in the regulation of cancer invasion and metastasis. The number and position of N-glycosylation and the kinase-mediated phosphorylation are different between EMPs. Numerous signaling molecules and cell surface proteins are associated with the role of EMPs. They mediate the EMP-induced cellular functions, such as proliferation and migration, that regulate the invasiveness of cancer cells and subsequent metastatic events. Red arrows: increase or activation of the molecules or phenomena; blue arrows: decrease or inactivation of the molecules or phenomena.
Cancers 11 01620 g002
Figure 3. A model for the promotion of cancer invasion and metastasis by EMP1. Interaction of cancer cells with stromal cells, such as CAFs, myofibroblasts, and tumor-associated macrophages, upregulates the expression of EMP1, which activates Rac1 through the copine-III-mediated intracellular signal transduction. This enhances cancer cell migration to promote cancer invasion and metastasis. Red arrow: activation of Rac1.
Figure 3. A model for the promotion of cancer invasion and metastasis by EMP1. Interaction of cancer cells with stromal cells, such as CAFs, myofibroblasts, and tumor-associated macrophages, upregulates the expression of EMP1, which activates Rac1 through the copine-III-mediated intracellular signal transduction. This enhances cancer cell migration to promote cancer invasion and metastasis. Red arrow: activation of Rac1.
Cancers 11 01620 g003
Table 1. Roles of EMP1 in cancer metastasis.
Table 1. Roles of EMP1 in cancer metastasis.
Type of CancerIn Vitro ModelIn Vivo ModelPatient SamplesRemarks
mRNAProteinmRNAProteinmRNAProtein
Pro-metastatic property
GBM[59,60][59,60] [59][59,60][59,60]Promotion of cancer cell proliferation and invasion; Correlation with poor clinical outcome
Uveal melanoma[65][65][65] [63] Possible relationship with a high risk for metastatic death
NSCLC[66][66,67] [67][68][67]Increase in cancer cell proliferation; Impairment of drug sensitivity
ALL[69][69] [69] Decrease in apoptosis; Increase in cancer cell migration, adhesion and proliferation; Impairment of drug sensitivity
Anti-metastatic property
Nasopharyngeal cancer[70][70] [70]Inhibition of cancer cell migration and invasion; Increase in apoptosis; Improvement of the patients’ survival rates
Esophageal cancer[71] [71] Decrease in cancer cell proliferation
Gastric cancer[72][72] [72]Correlation with reduced cancer invasion and metastasis and with elongation of the patients’ survival
Colorectal cancer[73][73] [73]Decrease in cancer cell proliferation; Increase in apoptosis; Improvement of the patients’ survival rates
Ovarian cancer [74]Possible association with reduction of the severity of the cancer
Paradoxical effect on metastasis
Breast cancer[76][76] [75,76]Possible correlation with promotion of cancer invasion; Biomarker to distinguish the histological types of the cancer
Inhibition of cancer cell migration, proliferation and invasion; Improvement of the patients’ survival rates
rostate cancer[22,77][22,77] [22][22][22,77]Promotion of cancer cell migration, invasion and metastasis
Inhibition of cancer cell migration and invasion
[ ] indicates the number of reference.
Table 2. Roles of EMP2 in cancer metastasis.
Table 2. Roles of EMP2 in cancer metastasis.
Type of CancerIn Vitro ModelIn Vivo ModelPatient SamplesRemarks
mRNAProteinmRNAProteinmRNAProtein
Pro-metastatic property
GBM[84][83,84] [83,84][82][81,83]Promotion of cancer cell migration, invasion and angiogenesis
Breast cancer[92][90,93] [92][86,87,88,89,91][90,93]Promotion of cancer cell invasion and metastasis
Ovarian cancer [97] [97] [97]Decrease in cancer cell death; Association with the malignant type of the cancer
Endometrial cancer[99,100][98,99,100] [98,99,100] [101]Promotion of angiogenesis; Correlation with cancer progression
Anti-metastatic property
Nasopharyngeal cancer[102][102] [102]Decrease in cancer cell growth; Enhancement of the sensitivity of radiotherapy; Improvement of the clinical outcome
Cutaneous melanoma[103][103] [103]Increase in apoptosis
Urothelial cancer[104][104] [104] [104]Decrease in cancer cell proliferation
[ ] indicates the number of reference.
Table 3. Roles of EMP3 in cancer metastasis.
Table 3. Roles of EMP3 in cancer metastasis.
Type of CancerIn Vitro ModelIn Vivo ModelPatient SamplesRemarks
mRNAProteinmRNAProteinmRNAProtein
Pro-metastatic property
GBM[109][109] [109][105,106,107,108,109,111,112][112]Increase in cancer cell proliferation; Decrease in apoptosis; Correlation with poor clinical outcome
Oral squamous cancer [113] [113][113]Increase in cancer cell migration
Gastric cancer[114][114] [114] Induction of EMT; Correlation with poor clinical outcome
Hepatocellular cancer [115] [115] [115]Promotion of cancer cell proliferation, migration and invasion; Negative correlation with tumor differentiation
Breast cancer[116][116] [116,117][116]Promotion of cancer cell proliferation, invasion and metastasis
Endometrial cancer [118] Possible correlation with development of the cancer
Urothelial cancer[119][119] [119][119]Increase in cancer cell proliferation and migration
Anti-metastatic property
Esophageal cancer[120][120] [120][120]Increase in cancer cell death; Improvement of the survival rate
Gallbladder cancer[121][121] [121][121][121]Inhibition of cancer cell proliferation, migration and invasion; Improvement of the patients’ survival rates
NSCLC [122] [122]Decrease in cancer cell proliferation
[ ] indicates the number of reference.

Share and Cite

MDPI and ACS Style

Ahmat Amin, M.K.B.; Shimizu, A.; Ogita, H. The Pivotal Roles of the Epithelial Membrane Protein Family in Cancer Invasiveness and Metastasis. Cancers 2019, 11, 1620. https://doi.org/10.3390/cancers11111620

AMA Style

Ahmat Amin MKB, Shimizu A, Ogita H. The Pivotal Roles of the Epithelial Membrane Protein Family in Cancer Invasiveness and Metastasis. Cancers. 2019; 11(11):1620. https://doi.org/10.3390/cancers11111620

Chicago/Turabian Style

Ahmat Amin, Mohammad Khusni B., Akio Shimizu, and Hisakazu Ogita. 2019. "The Pivotal Roles of the Epithelial Membrane Protein Family in Cancer Invasiveness and Metastasis" Cancers 11, no. 11: 1620. https://doi.org/10.3390/cancers11111620

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop