Open access peer-reviewed chapter

METCAM/MUC18: A Novel Tumor Suppressor for Some Cancers

Written By

Guang-Jer Wu

Submitted: 01 November 2018 Reviewed: 10 April 2019 Published: 18 May 2019

DOI: 10.5772/intechopen.86271

From the Edited Volume

Genes and Cancer

Edited by Guy-Joseph Lemamy

Chapter metrics overview

1,157 Chapter Downloads

View Full Metrics

Abstract

METCAM/MUC18, a component of cellular membrane, is a cell adhesion molecule (CAM) in the Ig-like gene super-family. It is capable of carrying out general functions of CAMs, such as performing intercellular interactions and interaction of cell with extracellular matrix in tumor microenvironment, interacting with various signaling pathways, and regulating social behaviors of cells. METCAM/MUC18 plays the tumor suppressor function in some cancers, such as colorectal cancer, nasopharyngeal carcinoma type I, one mouse melanoma subline K1735-9, ovarian cancer, pancreatic cancer, prostate cancer PC-3 cell line, and perhaps hemangioma. Possible mechanism in the METCAM/MUC18-mediated tumor suppression is proposed. By taking advantage of the tumor suppressor function of METCAM/MUC18, recombinant METCAM/MUC18 proteins and other derived products may be used as therapeutic agents to treat these cancers.

Keywords

  • METCAM/MUC18
  • Ig-like CAM
  • in vivo tumor suppression
  • colorectal cancer
  • nasopharyngeal carcinoma
  • mouse melanoma
  • ovarian cancer
  • pancreatic cancer
  • prostate cancer
  • mouse models

1. Introduction: tumor initiation and malignant progression is mainly controlled by two sets of genes as well as CAMs

Tumor/cancer is a genetic disease due to accumulated mutations or epigenetic alterations in our genetic material, DNA [1]. 80–90% of cancer risk comes from environmental factors and the remaining 10–20% risk from hereditary factors [2]. Environment in a broad sense includes both the physical containment and the social and cultural environment and its associated effects on our lifestyle choices. The environmental factors in the physical containment include chemicals (from polluted drinking water, air and soil, and diet), physical agents (UV and environmental radiation and medical radiation), biological agents (tumor viruses, bacteria, and parasites), and the lifestyle. These agents aim to attack our DNA in the somatic cells and resulting in accumulation of mutations and epigenetic alterations in our genes throughout our life time. Hereditary factors (lineage specific cues) include both the inherited genetic mutations and epigenetic imprinting in the germ cells that pass on from generation to generation. Tumor initiation and malignant progression are mainly caused by two sets of genes, such as the tumor-promoting genes (oncogenes) and the tumor suppressor genes, thus, mutations and epigenetic alterations in these two sets of genes are doom to be responsible for the tumorigenic process [2, 3, 4].

In addition to exogenous chemical agents, physical agents, and biological agents in the environment that cause mutations in the genes, endogenous metabolic processes and chronic inflammation from our lifestyle choices produce free radicals that directly attack our DNA also resulting in mutations [5]. The major sources of free radicals are reactive oxygen species (ROS), which is a collective term for the unstable, reactive, partially reduced oxygen derivatives that are the normal by-products of our metabolic processes. They include hydrogen peroxide (H2O2), superoxide anion (O2), hypochlorous acid (HOCl), singlet oxygen (1O2), and hydroxyl radical (HO). ROS are also produced by the inflammatory macrophages and neutrophils and are spilled out to attack the DNA of bystander cells. ROS acts as the secondary messengers in cell signaling and essential for various biological processes in both normal and cancer cells and as both tumor-promoting and tumor suppressing agents. To keep the system in check, ROS is balanced by intracellular anti-oxidant enzymes, that produce a number of anti-oxidants, such as glutathione (GSH) and thioredoxin (Txn), which are also present in our foodstuffs, to remove ROS. ROS production is a mechanism shared by most chemotherapeutics to trigger cell-death in cancer cells and unfortunately also to some extent in normal cells. Thus, ROS has conflicting roles as a secondary messenger in cancer cells as well as cancer-killers during cancer chemotherapy.

Most of the mutations in the oncogenes are dominant and thus manifest obvious phenotypes of increased proliferation and survival of tumor cells (gain-of-functions). In contrast, most of mutations in the tumor suppressor genes are recessive and thus do not manifest any phenotype until both copies of the gene are mutated or altered epigenetically (loss-of-functions). Some tumor suppressor genes are gate-keepers that directly affect proliferation and death, thus directly open to tumor formation. But some tumor suppressor genes are care-takers that affect DNA repair functions and genomic stability, thus increase mutation rate of all genes and indirectly affect proliferation [2, 6].

Epigenetic alterations may change the extent of methylation (either hypo- or hyper-methylation) in the regulatory regions of both oncogenes and tumor-suppressor genes, thus affect the transcriptionally regulatory region of the genes and directly regulate transcriptional expression of the genes. Epigenetic alterations may also modify histones and non-histone proteins that affect chromosome remodeling, thus indirectly affect the transcription of the genes. Epigenetic alterations may also affect post-transcription processes (namely translational process or stability of mRNA) of the genes via microRNAs [7].

Besides the above traditional two sets of genes, other genes, such as CAMs, also contribute directly to the tumor initiation and progression or orchestrate the tumor microenvironment to affect the tumor progression [8]. CAMs are involved in several biological functions, such as tissue architecture, organ formation, blood vessel generation and angiogenesis, immune and inflammatory reactions, wound healing and social behaviors [8]. An altered expression of CAMs may have implications in tumorigenesis, since CAMs govern cellular social behaviors by directly contributing to cell adhesion and cross-talk with the intracellular signal transduction pathways [8]. As a consequence, an aberrant expression of CAMs is capable of changing mobility and invasiveness, influencing outlasting ability and proliferation of tumor cells, and altering new blood vessel formation [8]. It also affects distant organ-dissemination of carcinoma cells, because CAMs orchestrate complex interactions of tumor cells with various stromal cells in the tumor microenvironment, resulting in augmentation or reduction of the spreading potential of carcinoma cells [8]. Effects of the aberrant expression of the following CAMs on tumorigenesis and malignant progression are better studied, such as cadherin [9], integrins [10], CD44 [11], CEACAM [12], mucins [13], L1CAM [14], EpCAM [15], ALCAM [16] and METCAM/MUC18 [17]. Over the past several years, our team investigated the role of METCAM/MUC18 in several types of tumors, such as melanoma, breast, nasopharyngeal, ovarian and prostate cancers [18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36]. The resulting data showed a dual role of METCAM/MUC18 as a tumor promotor or suppressor in these cancers [17, 37].

Advertisement

2. METCAM/MUC18: an immunoglobulin-like (Ig-like) CAM

Originally, METCAM/MUC18 was first demonstrated to be abundantly expressed on the cellular membrane of most malignant human melanomas, hence named as MUC18. It has been implicated to play a pivotal role in the malignant progression of human melanoma, hence was named as MCAM and Mel-CAM) [38]. However, METCAM/MUC18 was found in subsequent studies not to be exclusively expressed in melanoma, and it did not initiate the transformation of normal cutaneous melanocytes to melanoma either [39]. Instead, METCAM/MUC18 was also expressed in other epithelial tumors and it could initiate or promote the transformation of other epithelial cells into carcinomas [40]. Thus, METCAM/MUC18 also bears other names, such as S-endo1, CD146, A32, or METCAM [40, 41]. Later METCAM/MUC18 was found to be able to suppress tumorigenesis in some cancer cell lines [17, 37, 40].

The human METCAM/MUC18 is a cell adhesion molecule (CAM) belonging to the Ig-like gene superfamily. The naked human METCAM/MUC18 is a single chain transmembrane protein of 65–72 kDa consisting in 646 amino acids with an extracellular N-terminal domain of 558 amino acids, a 24 amino acids-transmembrane domain and a cytoplasmic domain of 64 residues (Figure 1) [38, 42].

Figure 1.

The human METCAM/MUC18 (huMETCAM/MUC18). The figure represents the protein structure of huMETCAM/MUC18 with its 3 domains: (1) a large extracellular domain showing a signal peptide (SP), the five Ig-like variables (V1 and V2) and conserved (C1, C2, C2′ and C2″) domains, each of which held together by a disulfide bond, and one X domain; six conserved N-glycosylation sites indicated as wavy lines in V1, the interdomain C2′/C2″, C2″ and X domains; (2) a short transmembrane domain (TM); and (3) a cytoplasmic domain containing five potential phosphorylation sites (P).

Figure 1 shows that the N-terminal extra-cellular domain of the protein is composed of a signal peptide sequence (SP) and five immunoglobulin-like domains and one X domain [37, 42]. The intracellular cytoplasmic domain has one, three, and one protein kinase consent sequences that are potentially to be phosphorylated by PKA, PKC, and CK2, respectively [37, 38, 42]. In addition, the METCAM/MUC18 usually has an apparent molecular weight of 110–150,000 because it is heavily glycosylated in all cell types. The amino acid sequence of huMETCAM/MUC18 reveals nine possible N-glycosylation sites, of which six are conserved between human and mouse proteins, in the extracellular domain. METCAM/MUC18 is conserved in mouse, in which the amino acid sequences of mouse METCAM/MUC18 (moMETCAM/MUC18) are 72.6% identical to the huMETCAM/MUC18 [43]. Therefore, both human and mouse METCAM/MUC18’s are capable of performing similar general functions of CAMs, such as controlling cellular social behaviors by impacting the adhesion status of cells and modulating signaling. Furthermore, over-expression of both human and mouse METCAM/MUC18’s similarly affected tumor cells in in vitro motility and invasiveness, in vitro and in vivo tumorigenesis, and in vivo metastasis [42, 43].

The huMETCAM/MUC18 is expressed in at least 10 normal tissues: hair follicular cells, smooth muscle cells, endothelial cells, cerebellum, basal cells of the lung, activated T cells, intermediate trophoblasts [44], breast epithelium [18, 19], nasopharyngeal epithelium [23], and ovarian epithelium [27]. The protein is also expressed in several carcinomas, such as breast carcinoma, intermediate trophoblast tumors, melanoma, prostate adenocarcinoma, osteosarcoma, and others [17, 44]. Our studies also indicate that over-expression of METCAM/MUC18 augments tumorigenesis of breast carcinoma [18, 19, 20], nasopharyngeal carcinoma type III [24, 26], and prostate adenocarcinoma [34], but it does not have an obvious effect on tumorigenesis of most melanoma cell lines [21]. METCAM/MUC18 over-expression also initiates the distant organ-dissemination of prostate cancer [32, 33] and augments the distant organ-dissemination of melanoma [21] and breast carcinoma [45].

In contrast, over-expression of METCAM/MUC18 represses tumorigenesis of a mouse melanoma cell line, K1735-9 [22], nasopharyngeal carcinoma type I [24, 25] and perhaps hemangiomas [46]. METCAM/MUC18 over-expression also represses the distant organ-dissemination of the mouse melanoma cell line, K1735-9 [22].

Advertisement

3. METCAM/MUC18: a tumor suppressor in several types of cancer

3.1 Mouse melanoma

Over-expression of moMETCAM/MUC18 in one mouse melanoma cell line K1735 clone 10 (or K1735-10 subline) has no effect and that in another cell line K1735 clone 3 a slight suppression effect on subcutaneous tumorigenesis [21], but in K1735 clone 9 (or K1735-9 subline) it completely suppresses the subcutaneous tumorigenesis [22]. Thus, METCAM/MUC18 definitely acts as a tumor suppressor for the K1735-9 subline, but may have a less obvious effect on two other K1735 sublines, K1735-3 and K1735-10. In addition to its effect on tumorigenesis, over-expression of moMETCAM/MUC18 in K1735-9 also completely suppressed lung nodule formation in immunocompetent syngeneic C3H brown mouse model. In contrast, over-expression of moMETCAM/MUC18 in K1735-3 and K1735-10 subline has an opposite effect (namely promotion) on lung nodule formation. In conclusion, moMETCAM/MUC18 acts as a tumor suppressor with a different severity on different cell lines in a syngenetic mouse model [21, 22].

3.2 Nasopharyngeal carcinoma

Nasopharyngeal carcinoma (NPC) occurs in the non-lymphomatous, squamous epithelial lining of the posterior nasopharynx [24]. Histologically, three subtypes of NPC are defined according to World Health Organization (WHO) classification: WHO type I (keratinizing squamous cell carcinomas), WHO type II (non-keratinizing squamous cell carcinomas), and WHO type III (undifferentiated carcinomas) [24]. Three major risk factors suggested by epidemiological studies, such as genetic predisposition, dietary and environmental factors, and the Epstein Barr virus (EBV) infection, may cause the unusual occurrence of NPC in endemic areas [24, 25, 26]. However, the biological mechanisms of their involvement in cancer initiation, development or malignant progression are not well understood. Nevertheless, it could be hypothesized that altered cell adhesion molecules (CAMs) in NPC lead to tumorigenesis and malignant progression, since aberrant expression of CAMs, such as CD44, connexin 43, E-cadherin, and ICAM, has been associated with the progression of NPC [23]. In order to test this hypothesis, we previously studied the possible role of altered METCAM/MUC18 expression in nasopharyngeal carcinoma [23, 24].

Therefore, we used immunohistochemistry method to determine gene expression at the protein level in seven tissue specimens of normal nasopharynx, 97 specimens of three different types of NPC and also used immunoblot method to determine that in several cell lines established from type I and type III NPC [23]. The results showed a weak expression of the protein METCAM/MUC18 in 27% of the NPC tissues in contrast to all the normal nasopharynx tissues which exhibited high expression of the protein. According to these results, we suggested that METCAM/MUC18 may play a tumor suppressor function in the development of NPC during the progression of the disease [23]. We then tested the hypothesis by transfecting the cDNA into two NPC cell lines which weakly expressed the protein and isolated the high-expressing clones for examining the effect of METCAM/MUC18 over-expression on in vitro cellular behavior and in vivo tumorigenesis of the two NPC cell lines in athymic nude mice. Consistent with the hypothesis, we indeed observed that METCAM/MUC18 over-expression suppressed the tumor growth of NPC-TW01 cells, which were established from type I NPC [47], as previously shown [24, 25]. We thus conclude that METCAM/MUC18 plays a tumor suppressor role in the development of the type I NPC [24, 25].

Surprisingly, when a different cell line, NPC-TW04, was used for the similar set of the experiments we observed a completely opposite effect of METCAM/MUC18. We observed that over-expression of METCAM/MUC18 promoted in vitro and in vivo tumor growth of NPC-TW04 cells, which were established from type III NPC [47], as previously reported [24, 26]. We thus conclude that METCAM/MUC18 plays a tumor promoter role in the development of the type III NPC [24, 26].

Taken together we hypothesized that METCAM/MC18 plays a dual suppressor and promotor role in the different types of NPC.

3.3 Ovarian carcinoma

Two independent groups showed that METCAM/MUC18 expression is correlated with the progression of ovarian cancer [27, 48], and it affects the in vitro behaviors of ovarian carcinoma cells [49]. However, the role of METCAM/MUC18 in the progression of epithelial ovarian cancer has not been directly tested in animal models. To investigate this, we initiated the studies by testing the effect of over-expression of METCAM/MUC18 on the in vitro cellular behaviors and in vivo tumorigenesis and malignant progression of human ovarian cancer cell lines in nude mice. First, we used a human ovarian cell line, SK-OV-3, for testing the effects of METCAM/MUC18 expression on their in vitro motility and invasiveness, and in vivo tumor formation after subcutaneous (SC) injection and also in vivo progression after intraperitoneal (IP) injection in athymic nude mice. We observed that over-expression of METCAM/MUC18 reduced in vitro motility and invasiveness [28] and suppressed in vivo tumorigenesis and malignant progression of the human ovarian cancer cell line SK-OV-3 [28]. Then, we used the other human ovarian cancer cell line, BG-1, for similar tests and also observed similar phenomenon [50].

In summary, we supplied in vitro and in vivo evidence to definitely support the conclusion that METCAM/MUC18 plays a suppressor role in the tumorigenesis and malignant progression of two human ovarian cancer cell lines [28, 50]. Our results suggest that METCAM/MUC18 is a strong candidate as a new tumor and metastasis suppressor in human ovarian cancer cells.

3.4 Prostate cancer

For the previous two decades, we have firmly established the notion that over-expression of METCAM/MUC18 promotes the tumorigenesis and metastasis of human prostate cancer cell line LNCaP, which was established from lymphatic lesions [31, 32, 33, 34, 35, 36]. To check if the conclusion is also extended to another human prostate cancer cell line DU145, we recently tested the effect of knocking down the endogenously expressed METCAM/MUC18 on tumorigenesis in a nude mouse system, since DU145 endogenously expressed a high level of METCAM/MUC18 [51]. We found that knocking down of the endogenously expressed METCAM/MUC18 with three shRNAs decreased the subcutaneous tumorigenesis in male nude mice in comparison to a control shRNA, as shown in Figure 2. We thus concluded that METCAM/MUC18 expression in DU145 cell line, which was established from brain lesions, plays a positive role in tumorigenesis (and perhaps metastasis) similar to in LNCaP cells.

Figure 2.

Tumorigenicity of four shRNA-knockdown clones of DU145. Effect of METCAM/MUC18 expression on in vivo tumorigenicity (Left) and final tumor weight (Right). (Left) Average tumor volumes from five mice S.C. injected with each of the 46 (control), 72, 24, and 27 clones/cells, which were transfected with four corresponding shRNAs in pGIPZ vector, were plotted against time. (Right) Average final tumor weights from five mice S.C. injected with the same clones/cells and standard deviations were plotted at the end point of experiment. P values are shown in the figure by comparing the data to the control clone [51].

In contrast, we recently used the similar knocking down strategy to test the effect of decreased the endogenous METCAM/MUC18 expression on in vivo tumorigenesis of another human prostate cancer cell line, PC-3, which was established from bone lesions, surprisingly we found that knocking down the endogenously expressed METCAM/MUC18 increased the tumor proliferation of PC-3 cells (which was opposite to that of DU145, as shown above in Figure 2), suggesting that expression of METCAM/MUC18 suppressed the tumorigenesis of the human prostate cancer cell line PC-3 [52], as shown in Figure 3.

Figure 3.

Tumorigenicity of four shRNA-knockdown clones of PC-3. Effect of METCAM/MUC18 expression on in vivo tumorigenicity (Left) and final tumor weight (Right). (Left) Average tumor volumes from five mice S.C. injected with each of the 46 (control), 72, 24, and 27 clones/cells, which were transfected with the four corresponding shRNAs in pGIPZ vector, were plotted against time. (Right) Average final tumor weights from five mice S.C. injected with the same clones/cells and standard deviations were plotted at the end point of experiment. P values are shown in the figure by comparing the data to the control clone [52].

We thus conclude that METCAM/MUC18 serves as a tumor suppressor in the PC-3 cell line, different from its role in two other prostate cancer cell lines (LNCaP and DU145), suggesting that prostate cancer cell lines established from different organs may have different intrinsic factors that modulate the function of METCAM/MUC18.

3.5 Colorectal cancer, hemangioma and pancreatic cancer

The protein METCAM/MUC18 is also expressed others cancers, such as angiosarcoma, gestational trophoblastic tumors, Kaposi’s sarcoma, leiomyosarcoma, some lung squamous and small cell carcinomas, and some neuroblastoma [44]. However, its role in the development of most of these cancers is not well known. Recent meta-analysis suggests that high METCAM/MUC18 expression in many solid tumors appears to be associated with poor prognosis and patient survival [53]. However, in contrast to the conclusion, reduced expression of METCAM/MUC18 associates with the malignant progression of hemangioma [46]. Likewise, recent results of the effects of METCAM/MUC18 expression on tumorigenesis of colorectal cancer and pancreatic cancer also appear to support the similar conclusion, as described next. Reduced expression of METCAM/MUC18 promotes tumorigenesis and stemness of colorectal cancer [54]. Targeting soluble METCAM/MUC18 with a neutralizing antibody inhibits vascularization, growth and survival of METCAM/MUC18-positive pancreatic tumors [55]. Furthermore, attenuation of METCAM/MUC18 promotes pancreatic cancer progression [56]. Thus, the possible tumor and metastasis suppressor role of METCAM/MUC18 in solid tumors appear to extend from mouse melanoma K1735–9 subline, ovarian cancer, and NPC type I, to colorectal cancer [54] and pancreatic cancer [55, 56], and perhaps hemangioma [46]. Table 1 summarizes the negative role of METCAM/MUC18 in the tumor formation and/or cancer metastasis of seven tumors/cancers.

Tumor/cancer cell lines Tumorigenesis Metastasis References
Colorectal cancer human cell lines HT-29, SW480, SW948, SW620, colo205, Lovo320, P6C Suppression Not determined [54]
Hemangioma human cell lines HemEC, HDMEC Possible suppression Not determined [46]
Mouse melanoma cell line K1735-9 Suppression Suppression [22]
Mouse melanoma cell lines K1735-3, K1735-10 No effect or slight suppression Increasing and affecting the late stage [21]
Nasopharyngeal carcinoma type I cell line NPC-TW01 Suppression Not determined [24, 25]
Ovarian cancer cell lines SK-OV3, BG-1 Suppression Suppression [28, 50]
Pancreatic cancer human cell lines, UACC-1273, PANC1, C81-61, KP-2, SUIT-2, MIAPaca-2, HS766T and primary CAFs Suppression Suppression [55, 56]
Prostate cancer human cell line PC-3 Suppression Not determined [52]

Table 1.

The negative role of METCAM/MUC18 in tumor formation and/or cancer metastasis of seven tumor/cancer cell lines.

Advertisement

4. METCAM/MUC18: a tumor promoter in most solid tumors

In contrast to the above functions of METCAM/MUC18, recent work done on other solid tumors appears to be consistent with the meta-analysis results of solid tumors [53], as described next. For example, METCAM/MUC18 expression correlates with the epithelial-mesenchymal transition (EMT) markers and a poor prognosis in gastric cancer [57]. Tumor up-take of glioma in an orthotopic xenograft mouse model correlates with the expression level of METCAM/MUC18 [58]. METCAM/MUC18 promotes metastasis and predicts poor prognosis of hepatocellular carcinoma [59]. Increased expression of METCAM/MUC18 has been found in hepatocellular carcinoma (HCC) tumor tissues as compared with the matched adjacent normal liver tissues and the METCAM/MUC18+ cells purified from HCC tumors and cells have significantly increased colony-forming capacity consistent with the cancer stem cells or the tumor-initiating cells [60]. METCAM/MUC18 expression has been shown to express in 51% of non-small cell lung carcinoma (NSCLC) and positive expression of METCAM/MUC18 has been associated with a shorter survival of patients with adenocarcinomas and used to predict the poor overall survival in patients with lung adenocarcinomas [61, 62, 63]. METCAM/MUC18 expression mediates acquisition of cancer stemness and enhances tumor invasion and metastasis in a mouse model [64]. High expression of METCAM/MUC18 correlates with intrapulmonary metastasis of NSCLC cells in a mouse model [65]. Consistent with the results, we showed in Figure 4 (Guang-Jer Wu, unpublished data) that METCAM/MUC18 is expressed in a lung type II alveolar epithelial cell carcinoma cell, A549, and highly expressed in an adenocarcinoma cell line, H838, in comparison with its no expression in an immortalized normal embryonic WI38 cell line.

Figure 4.

Expression of METCAM/MUC18 in normal lung tissue (SV40-immortalized normal lung cells (WI38, lane 2) and lung type II alveolar epithelial cell carcinoma cell (A549, lane 3) and lung primary adenocarcinoma (H838, lane 4) (from Guang-Jer Wu, unpublished data).

Furthermore, METCAM/MUC18 mediates chemoresistance of small cell lung carcinoma (SCLC) [66]. METCAM/MUC18 is expressed in osteosarcoma cell lines, but not in normal osteoblast cells [67]. Osteosarcoma is effectively treated with METCAM/MUC18 monoclonal antibodies [68, 69]. Transcription factor MEIS1 activates METCAM/MUC18 expression to promote migration of mouse pancreatic tumor cell lines [70]. METCAM/MUC18 very likely promotes the formation of angiosarcoma, as supported by our preliminary results as described next. Mouse METCAM/MUC18 was expressed in one angiosarcoma clone, SVR, which was transfected with H-Ras, at a higher level than in the control cell line, an immortalized normal endothelial cell line, MS-1 [71]. Furthermore, the tumorigenicity of the SVR cell line was higher than the control cell line, thus in direct association with the higher expression level of moMETCAM/MUC18 [40, 71]. This suggests that METCAM/MUC18 very likely promotes the formation of angiosarcoma [40, 71]. Hence, the positive role played by the METCAM/MUC18 in the progression of solid tumors have been extended from breast cancer, human and mouse melanoma, prostate cancer to angiosarcoma [40, 71], gastric cancer [57], glioma [58], hepatocellular carcinoma [59, 60], non-small cell lung adenocarcinoma [61, 62, 63, 64, 65], small cell lung cancer [66], osteosarcoma [67, 68, 69], and mouse pancreatic cancer [70]. Taken together, METCAM/MCU18 appears to be more prevalently in playing a positive role than a negative role in the tumorigenesis of solid tumors. Table 2 summarizes the positive role of METCAM/MUC18 in the tumor formation and/or cancer metastasis of various tumors/cancers.

Tumor/cancer tissues or cell lines Tumorigenesis Metastasis References
Angiosarcoma human cell lines MS1, SVR Increasing Not determined [40, 71]
Human breast cancer cell line MCF-7 Promotion Not determined [18]
Human breast cancer cell line SK-BR-3 Promotion Not determined [19, 20]
Human breast cancer cell lines MDA-MB-231 and MDA-MB-468 Promotion Promotion [19, 45]
Gastric cancer human tissues Promotion Not determined [57]
Glioma cell lines U87MG, U251 Promotion Not Determined [58]
Hepatocellular carcinoma human cell lines PLC/PRF/5, Huh7, MHCC97H& 97 L HepG2, SMMC-7721, focus, YY-8103, LM3, HLF and primary HCC cell lines; normal liver cell line LO2 Promotion Not determined [59, 60]
Non-small cell lung cancer human cell lines A549, H23, H358, H460, H522, H838, HCC4006, H1650/ER, PC-9, PC9GR, and adenocarcinoma tissues Promotion Promotion [61, 62, 63, 64, 65], our unpublished results
Small cell lung cancer human cell lines H69, H69AR, H82, H196, H209, DMS79 Promotion Not determined [66]
Clinical melanoma tissues and human melanoma cell lines SB-2, SK, XP-44 No effect Increasing and affecting the late stage [38, 72, 73]
Mouse melanoma cell lines K1735-3, K1735-10 No effect or slight suppression Increasing and affecting the late stage [21]
Nasopharyngeal carcinoma type III human cell line NPC-TW04 Promotion Not determined [24, 26]
Osteosarcoma human cell lines CR9, MNNG-HOS, OHS, KPDX, KRIB, MG-63, shYY1, SaOS, SaOS-2, TE85, U20S Promotion Augmentation [67, 68, 69]
Pancreatic cancer mouse cell lines ptf1a, LSL-Kras, LSL-Trp53, Pdx1, Promotion Possible augmentation [70]
Clinical prostate cancer human tissues Increasing Increasing and affecting initiation in the early stage (PIN) [31]
Human prostate cancer cell line LNCaP Increasing Increasing and affecting initiation in the early stage [32, 34, 35, 36]
Human prostate cancer cell line DU145 Increasing Not determined [51]
Prostate adenocarcinoma in TRAMP mice Increasing Increasing and affecting initiation in the early stage [33]

Table 2.

The positive role of METCAM/MUC18 in tumor formation and/or cancer metastasis of various tumors/cancers.

In conclusion, METCAM/MUC18 appears to play a dual role in the tumorigenesis and perhaps also in metastasis of solid tumors. At this point, it is not clear why METCAM/MUC18 plays a dual role in this aspect. Since METCAM/MUC18 only plays a dual role in different cell lines from the same type of cancer or in different type of cancers, but never in the same cancer cell line. It is logical to suggest a possible explanation that the intrinsic properties of each cancer cell line may provide specific co-factors or heterophilic ligands that may positively or negatively modulate the METCAM/MUC18-mediated tumorigenesis and metastasis. This can be readily scrutinized by identifying these specific intrinsic co-factors or heterophilic ligands by using immunological co-precipitation method in the future studies. This approach is feasible as described in one of the following sections, Section 5.1.

Advertisement

5. Putative mechanisms

Since the huMETCAM/MUC18 was first discovered in the 1980s, three groups have worked on the role of huMETCAM/MUC18 in melanoma metastasis [38, 39, 72, 73], another group on the role of huMETCAM/MUC18 in the biology of endothelial cells [41], and our group joined in the effort to study the role of huMETCAM/MUC18 in the progression of mouse melanoma [43] and prostate cancer [31, 32, 33, 34, 35, 36, 51, 52], and later breast cancer [18, 19, 20], ovarian cancer [27, 28, 29, 30], and NPC [23, 24, 25, 26], as described above. Recently, more groups have participated in further exploring the possible role of METCAM/MUC18 in other solid tumors in different organs, such as colorectum [54], gastro-organ [57], glioma [58], liver [59, 60], lung [61, 62, 63, 64, 65, 66], pancreas [55, 56, 70], and bone [67, 68, 69]. Preliminary work in leiomyosarcoma, esophagus squamous cell carcinoma, clear cell renal sarcoma, and gallbladder adenocarcinoma are also beginning to emerge [53].

After many decades of group effort, we are beginning to understand the biology of METCAM/MUC18-mediated tumor progression. However, the biological mechanisms describing the role of METCAM/MUC18 in tumorigenesis and malignant progression are still not well clarified such as: the protein’s domain involved in cell adhesion, the domain which mediates the interactions of tumor cells with the tumor microenvironment leading to tumor progression and in the METCAM/MUC18-mediated tumorigenesis and malignant progression, and the effects of N-glycosylation on the functions of METCAM/MUC18 in tumorigenesis. Though the huMETCAM/MUC18-mediated outside-in and inside-out signaling in endothelial cells are understood to some extent, and the METCAM/MUC18-mediated signaling, which is leading to the progression of various cancer cells, are not much known. How METCAM/MUC18 is positively or negatively regulated at the level of transcription in different cancer cells remains minimally known. As such, the following five important aspects are much needed for immediate future studies, such as different kinds or quantities of co-factors or heterophilic ligand(s) in different cancer cell lines, contributions of different domains of the protein, different signaling pathways involved, differential regulation at the transcription level in tumors of different organs, and possible different extent of N-glycosylation in different cancer cell lines, which may critically modulate the function of METCAM/MUC18 in tumor progression.

5.1 The heterophilic ligands of METCAM/MUC18

The heterophilic ligands of METCAM/MUC18 may play an important role in the cell-cell and cell-extra-cellular matrix interactions and cancer metastasis. Our preliminary results suggest that the 72 kDa protein identified by immunoprecipitation method may be one of the heterophilic ligands for METCAM/MUC18, as shown in Figure 5 [40].

Figure 5.

Putative heterophilic ligand of METCAM/MUC18 in PC-3 and DU145 cell lines.

As shown in Figure 5, the putative heterophilic ligand 72 kDa is highly expressed in the PC-3 cell line, but much less in the DU145 cell line. This may reveal a possible explanation for the different role of huMETCAM/MUC18 in the tumorigenicity of the two prostate cancer cell lines [40].

5.2 The domains of huMETCAM/MUC18 required for tumorigenesis and metastasis

The relation of the protein structure of huMETCAM/MUC18 to its functions in tumorigenesis and metastasis have not been systematically defined. To begin addressing this question, we have generated mutants deleted different domains of huMETCAM/MUC18 by using a special PCR method [74] and used them to determine their contribution to tumorigenesis. Surprisingly, our results showed that the ecto-domain of huMETCAM/MUC18 induced tumorigenesis in LNCaP cells in nude mice, as well as the whole wild type of cDNA. These preliminary results suggested the key role of the ecto-domain in tumorigenesis induction in prostate cancer cells in vivo. This may implicate a puzzling notion that the cytoplasmic domain was not essential for this process (Guang-Jer Wu, data not shown). However, the critical direct test of using only the cytoplasmic domain for inducing tumor has not been performed for LNCaP cells. From the above puzzling observation, it is very clear that a systematic study has also to be performed in other cancer cell lines before a definitive conclusion can be drawn.

5.3 Signaling pathways in the METCAM/MUC18-mediated tumorigenesis and cancer metastasis

The huMETCAM/MUC18 contains three sites which are potentially phosphorylated by PKC, PKA and CK2 in the cytoplasmic tail [38, 42]. However, these putative phosphorylation sites have not been biochemically proven. Thus, the immediate question to be answered is that how many sites in the cytoplasmic tail of the native METCAM/MUC18 protein, which are to be isolated from different cancer cell lines, are actually phosphorylated? Which protein kinase is responsible for the phosphorylation? After this is answered, then we can further study how METCAM/MUC18 mediates crosstalk and networking with different signal pathways and to see if it is similar to or different from the cytoplasmic tails of other CAMs [41, 75, 76, 77]. Knowledge learned from other CAMs seem point to one aspect that METCAM/MUC18, as an integral membrane protein and a cell adhesion molecule, should mediate inside-in, inside-out, and outside-in signals to participate in intercellular communication and interaction of cell with extra-cellular matrix, which results in impacting cell motility and invasiveness [78, 79]. Furthermore, its interaction with co-factors or cognate heterophilic ligand(s) may alter these signals, which in turn should affect intrinsic tumor proliferation or impact tumor angiogenesis and/or mediate targeting to specific organs and promoting metastasis. Moreover, METCAM/MUC18 may interact with various hormonal receptors, growth or anti-growth factors/receptors, various chemokines/receptors, and the Ca2+-mediated signaling members, which in turn affect the process of tumor progression. Figure 6 summarizes the possible preliminary crosstalk of huMETCAM/MUC18 with many members of signal transduction pathways that may affect its function during tumor initiation and development and malignant progression.

Figure 6.

METCAM/MUC18-mediated signal transduction in tumorigenesis and malignant progression.

5.4 Regulation of the huMETCAM/MUC18 gene transcription

The mechanism of transcriptional control of METCAM/MUC18 gene is minimally studied [17]. Up to now, only the 900 bp sequences in the core promoter region of the huMETCAM/MUC18 gene are well-characterized [80]. This core promoter is rich in GC sequences but does not contain a TATA box. It includes many consensus sequences presumably as putative binding sites for various transcription regulatory factors, such as SP-1, CREB [81], AP-2 [82, 83], c-Myb [84], N-Oct2 (Brn2) [85], Ets [86], CArG [87], and Egr-1 [88]. In addition, it also contains three insulin responsive elements (one Ets and two E-box motifs) [89], suggesting that huMETCAM/MUC18 gene expression may respond to the cue of various growth signals [37, 40], as shown in Figure 7.

Figure 7.

Putative transcription factor-recognized motifs in the 900 bp core promoter and 5–10 kilo bp up-stream region of the huMETCAM/MUC18 gene.

In addition, some sequences upstream of the minimal core promoter sequences should also be expected for conferring the tissue-specific expression of the huMETCAM/MUC18 gene [90]. Recently this notion has been definitely supported by a finding that Ets sequence in the 10 kilo-bp up-stream region is involved in the regulation of the expression of huMETCAM/MUC18 gene [91]. We have also engaged in this task by searching the sequence of the up-stream region of the huMETCAM/MUC18 promoter in the Celera or other web sites. By taking advantage of the known sequence searched, we designed many pairs of primers to screening a genomic library and obtained several phage clones which contain at least 4 kilo-bp of the gene for future studies (Guang-Jer Wu, unpublished data).

The epigenetic control of the expression of huMETCAM/MUC18 gene has not been extensively studied in NPC, though it has been implicated [92]. This is because huMETCAM/MUC18 gene is located at the locus of human chromosome 11q23.3 that has been shown to be hypermethylated in NPC, suggesting that the expression of this gene may be regulated by epigenetic controls [93]. To support this notion, our preliminary results of treating NPC cell lines with 5-Aza-2′-deoxycytidine (Aza-C) showed that after the treatment with Aza-C, METCAM/MUC18 expression was somewhat elevated in the NPC-TW01 cell line, but not in the NPC-TW04 cell line (Guang-Jer Wu, unpublished data). METCAM/MUC18 has also been shown to be methylated in most of the early stage of prostate cancer [94]. Further systematic studies in this aspect should be very interesting and rewarding in the future.

5.5 The possible roles of glycosylation on the protein of METCAM/MUC18 in tumorigenesis and tumor progression

Glycosylation of a protein may affect the proper folding, stability, and/or activity of a protein [95], however, the possible roles of glycosylation in the function of MCETCAM/MUC18 protein have not been explored. The glycosylation of METCAM/MUC18 may also affect its ability in inducing/promoting or suppressing the metastasis of cancer cells [95, 96, 97, 98, 99]. Both huMETCAM/MUC18 and moMETCAM/MUC18 may very likely to be heavily glycosylated, sialylated, and post-translationally modified, because both have an apparent molecular weight of about 110–150 kDa, which is much more than the naked protein with a molecular weight of about 65–70 kDa [100]. To initiate the study, we subjected the huMETCAM/MUC18, which was expressed in a human cancer cell line, to the digestion with N-glycosidase F, neuraminidase (sialidase), O-glycosidase, or endoglycosidase H, and we observed that the apparent molecular weight of the protein was decreased after digestion with N-glycosidase F and neuraminidase (sialidase), but not with O-glycosidase or endoglycosidase H [37, 40]. From this, we suggested that both sialic acid and N-glycans are probably the major carbohydrate side chains of huMETCAM/MUC18. It is also possible that glycosylation may differ depending on the type of cancers. Thus, we suggested that different N-glycans at the N-glycosylation sites of huMETCAM/MUC18 may differ in different cancer cell lines, which may have significant positive or negative impacts on their EMT abilities as well as tumorigenesis and metastasis. According to our hypothesis, a recent study described GCNT3 as an up-stream regulator of METCAM/MUC18. Moreover, GCNT3 glycosylates METCAM/MUC18 and extends its half-life which results in further elevation of S100A8/A9-mediated cellular motility in melanoma cells [101].

By searching in the primary sequence of the human huMETCAM/MUC18 protein, nine potential N-glycosylation sites (Asn-X-Ser/Thr or N-X-S/T sites) have been revealed [37, 38, 40, 42], whereas only seven sites found in the mouse METCAM/MUC18 [43]. Six N-glycosylation sites are conserved between the two proteins: 56/58 NL/FS, 418/420NRT, 449/451NLS, 467NGT/469NGS, 507NTS/509NTT, and 544/546NST [37, 38, 40, 42]. We suggest that only these six conserved N-glycosylation sites are actually glycosylated, because the apparent molecular weights of human METCAM/MUC18 and mouse METCAM/MUC18 are similar in the SDS gel. All the N-glycosylation sites are located in the external region of the protein, such as the domains of V1, C′, C″ and X. First, all these six sites should be biochemically identified before further molecular genetic task. Then, we will use genetic tools to alter the N-glycosylation sites. The mutants will be transfected back into cancer cell lines without the endogenous expression of the protein. The clones, which only express these mutated METCAM/MUC18, will be used for various in vitro and in vivo experiments to test the effect of N-glycosylation on the function huMETCAM/MUC18. They also will be used for testing effects on in vitro cell–cell aggregation and cell-extracellular matrix adhesion and on in vivo tumorigenesis and metastasis of human cancer cells. We anticipate that systematic studies on this aspect should be very informative to reveal the essential role of N-glycosylation played in the METCAM/MUC18-mediated tumor progression.

Advertisement

6. Conclusions

METCAM/MUC18 plays a key role in suppressing the progression of colorectal cancer, one mouse melanoma cell line, NPC type I, ovarian cancer, pancreatic cancer, prostate cancer PC-3 cell line, and perhaps hemangioma and possibly in other cancers. On the other hand, METCAM/MUC18 also play a key positive function in the progression of breast cancer, gastric cancer, hepatocellular carcinoma, lung cancer, melanoma, NPC type III, pancreatic cancer, and prostate cancer. To further understand its role in these processes, it is essential to further identify its co-factor regulators and cognate heterophilic ligands, define its functional domains, and study its crosstalk with members of various signal transduction pathways, the regulation of its expression at the level of transcription, and effects of N-glycosylation on the functions of the protein.

Advertisement

7. Research perspectives and clinical applications

7.1 Research perspectives

The current studies have laid an important biological basis for inspiring future intense investigation to further understand the detailed knowledge of METCAM/MUC18-mediated suppression of tumorigenesis and metastasis of various cancer cell lines. For this purpose besides those have been described above, other future endeavors may include: (a) understanding three major mechanisms involved in METCAM/MUC18-induced tumor and metastasis dormancy, such as key players participated in inhibition of intrinsic growth capability, key chemokines and cytokines participated in suppression of immunological responses, and key pro-angiogenic and anti-angiogenic factors participated in the reduction of angiogenesis [102], (b) identification of possible miRNAs and non-coding RNAs participated in the process upstream and downstream of METCAM/MUC18 [103], and (c) possible clinical applications should be explored. Precaution should be taken that a complete picture may only be possibly constructed after all the above studies are successfully executed.

7.2 Clinical applications

The majority of the cancer-associated mortality is due to dissemination of primary tumor to distant organs (metastasis). If we are able to decrease or stop the metastatic propensity of cancer cells and keep them stayed only at the primary site, it should be a major success in cancer therapy. Alternatively, it is also a major success if we are able to control cancer cells at the state of dormancy or remaining them at the stage of micro-metastatic lesions [104]. Thus, similar to other tumor and metastasis suppressors, such as KISS1, KAI1, nm23, MAP2K4, and some micro-RNAs, METCAM/MUC18 may be used as a new therapeutic target for some clinical cancer treatments [105]. Strategically four major approaches may be taken for this purpose: (a) use gene therapeutic method to restore the functional copy of the suppressor genes or use epigenetic method to re-activate the genes. For gene therapy, the METCAM/MUC18 cDNA gene may be transported by an adenovirus-associated virus vector or a replication-defective adenovirus [106]. The human METCAM/MUC18 gene, located on 11q23-3 chromosome may be targeted with clinical reagents to reverse epigenetic repression, like Aza-C [107], or to change histone modifications to induce remodeling of the chromosome [108], (b) dispense recombinant proteins directly to the patients. For this approach, a complete copy or a partial portion of the METCAM/MUC18 recombinant protein, oligopeptides, or small molecule mimetics of METCAM/MUC18 may be directly dispensed to cancer patients, (c) target at downstream key members in the signaling pathways which are activated by the loss of the suppressor function, and (d) the co-factors or the cognate heterophilic ligand(s) of METCAM/MUC18 may be targeted. The above strategies may be used in single, or better in combination for treating the patients for the purpose of holding tumor cells at the primary sites, stopping them in a dormant state, or keeping the disseminating cancer cells at the state of micro-metastases. However, the dual role of METCAM/MUC18 in cancer progression may limit the above clinical applications to only cancers exhibiting an anti-tumor activity mediated by METCAM/MUC18.

Advertisement

Acknowledgments

I thank the support of grants from National Research Council, Taiwan.

Advertisement

Conflict of interests

The author has no conflict of interests.

References

  1. 1. Jackson M, Marks L, May GHW, Wilson JB. The genetic basis of disease. Essays in Biochemistry. 2018;62:643-723
  2. 2. Klinsmith LJ. Principles of Cancer Biology. San Francisco: Pearson Education Press; 2006
  3. 3. Weinberg RA. The Biology of Cancer. 1st ed. New York, USA and Abington, UK: Garland Science; 2007
  4. 4. Baylin SB, Jones PA. Epigenetic determinants in cancer. Cold Spring Harbor Perspectives in Biology. 2016;8(a019505):1-35
  5. 5. Yang H, Villani RM, Wang H, Simpson MJ, Roberts MS, Tang M, et al. The role of cellular reactive oxygen species in chemotherapy. Journal of Experimental and Clinical Cancer Research. 2018;37(266):1-10
  6. 6. Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell. 2011;144:646-674. DOI: 10.1016/j.cell.2011.02.013
  7. 7. Ramassone A, Pagotto S, Veronese A, Visone R. Epigenetics and MicroRNA in cancer. International Journal of Molecular Sciences. 2018;19(459):1-28
  8. 8. Gkretsi V, Stylianopoulos T. Cell adhesion and matrix stiffness: Coordinating cancer cell invasion and metastasis. Frontiers in Oncology. 2018;8:145. DOI: 10.3389/fonc.2018.00145
  9. 9. Mendonsa A, Na TY, Gumbiner BM. E-cadherin in contact inhibition and cancer. Oncogene. 2018;37(35):4769-4780
  10. 10. Hamidi H, Ivaska J. Every step of the way: Integrins in cancer progression and metastasis. Nature Reviews. Cancer. 2018;18(9):533-548. DOI: 10.1038/s41568-018-0038-z
  11. 11. Wang Z, Zhao K, Hackert T, Zoller M. CD44/CD44v6 a reliable companion in cancer-initiating cell maintenance and tumor progression. Frontiers in Cell and Development Biology. 2018;6:97. DOI: 10.3389/fcell.2018.00097
  12. 12. Calinescu A, Turcu G, Nedelcu RI, Brinzea A, Hodorogea A, Antohe M, et al. On the dual role of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) in human malignancies. Journal of Immunology Research. 2018;2018:ID7169081. 8p. DOI: 10.1155/2018/7169081
  13. 13. Bhatia P, Gautqm SK, Cannon A, Thompson C, Hall BR, Aithal A, et al. Cancer-associated mucins: Role in immune modulation and metastasis. Cancer Metastasis Review. 9 Jan 2019. 14p. DOI:10.1007/s10555-018-09775-0
  14. 14. Altevogt P, Doberstein K, Fogel M. L1CAM in human cancer. International Journal of Cancer. 2016;138:1565-1571
  15. 15. Yahyazadeh Mashhadi SM, Kazemimanesh M, Arashkia A, Azadmanesh K, Meshkat Z, Golichenari B, et al. Shedding light on the EpCAM: An overview. Journal of Cellular Physiology. 2019;234(4):12569-12580
  16. 16. Weidle UH, Eggle D, Klostermann S, Swart GWM. ALCAM/CD166: Cancer-related issues. Cancer Genomics and Proteomics. 2010;7(5):231-243
  17. 17. Wu GJ. Chapter 13. Dual role of METCAM/MUC18 expression in the progression of cancer cells. In: Uchiumi F, editor. Gene Expression and Regulation in Mammalian Cells-Transcription from General Aspects. University Campus STeP Ri, Rijeka, Croatia: InTech Open Access Publisher; 2018. pp. 257-289. ISBN 978-953-51-3856-3, Print ISBN 978-953-51-3855-6
  18. 18. Zeng GF, Cai SX, Wu GJ. Up-regulation of METCAM/MUC18 promotes motility, invasion, and tumorigenesis of human breast cancer cells. BMC Cancer. 2011;11:113. DOI: 10.1186/1471-2407-11-113
  19. 19. Zeng GF, Cai SX, Liu Y, Wu GJ. METCAM/MUC18 augments migration, invasion, and tumorigenicity of human breast cancer SK-BR-3 cells. Gene. 2012;492:229-238
  20. 20. Huang CY, Wu GJ. METCAM/MUC18 promoted tumorigenesis of human breast cancer SK-BR-3 cells in a dosage-specific manner. Taiwanese Journal of Obstetrics and Gynecology. 2016;55(2):202-212. DOI: 10.1016/j.tjog.2016.02.010
  21. 21. Wu GJ, Fu P, Wang SW, Wu MWH. Enforced expression of MCAM/MUC18 increases in vitro motility and invasiveness and in vivo metastasis of two mouse melanoma K1735 sublines in a syngeneic mouse model. Molecular Cancer Research. 2008;6(11):1666-1677
  22. 22. Wu GJ. Ectopic expression of MCAM/MUC18 increases in vitro motility and invasiveness, but decreases in vivo tumorigenesis and metastasis of a mouse melanoma K1735-9 subline in a syngeneic mouse model. Clinical & Experimental Metastasis. 2016;33(8):817-828. DOI: 10.1007/s10585-016-9812-z
  23. 23. Lin JC, Chiang CF, Wang SW, Wang WY, Kwuan PC, Wu GJ. Significance and expression of human METCAM/MUC18 in nasopharyngeal carcinoma (NPC) and metastatic lesions. Asian Pacific Journal of Cancer Prevention. 2014;15(1):245-252
  24. 24. Liu YC. Putative roles of huMETCAM in modulating the development and progression of nasopharyngeal carcinoma [thesis]. Chung Yuan Christian University; 2014. Available from: http://www.lib.cycu.edu.tw/thesis
  25. 25. Liu YC, Chen YR, Wu GJ. METCAM/MUC18 plays a tumor suppressor role in the development of nasopharyngeal carcinoma type I. 2019. (Submitted)
  26. 26. Liu YC, Ke CC, Chen YR, Wu GJ. METCAM/MUC18 plays a tumor promoter role in the development of nasopharyngeal carcinoma type III. 2019. (Submitted)
  27. 27. Wu GJ, Dickerson EB. Frequent and increased expression of human METCAM/MUC18 in cancer tissues and metastatic lesions associates with the clinical progression of human ovarian carcinoma. Taiwanese Journal of Obstetrics and Gynecology. 2014;53:509-517
  28. 28. Wu GJ, Zeng GF. METCAM/MUC18 is a novel tumor and metastasis suppressor for the human ovarian cancer SKOV3 cells. BMC Cancer. 2016;16:136. DOI: 10.1186/S12885-016-2181-9
  29. 29. Wu GJ. METCAM/MUC18 plays a novel tumor and metastasis suppressor role in the progression of human ovarian cancer cells. Obstetrics & Gynecology International Journal. 2017;6(4):00210, pp. 1-8
  30. 30. Wu GJ. METCAM/MUC18 decreases the malignant propensity of human ovarian carcinoma cells. International Journal of Molecular Sciences. 2018;19:02976
  31. 31. Wu GJ, Varma VA, Wu MWH, Yang H, Wang SWC, Liu Z, et al. Expression of a human cell adhesion molecule, MUC18, in prostate cancer cell lines and tissues. The Prostate. 2001;48:305-315
  32. 32. Wu GJ, Peng Q , Fu P, Chiang CF, Wang SWC, Dillehay DL, et al. Ectopical expression of human MUC18 increases metastasis of human prostate cancer LNCaP cells. Gene. 2004;327:201-213
  33. 33. Wu GJ, Chiang CF, Fu P, Hess W, Greenberg N, Wu MWH. Increased expression of MUC18 correlates with the metastatic progression of mouse prostate adenocarcinoma in the (TRAMP) model. Journal of Urology. 2005;173:1778-1783
  34. 34. Wu GJ, Wu MWH, Liu Y. Enforced expression of human METCAM/MUC18 increases the tumorigenesis of human prostate cancer cells in nude mice. Journal of Urology. 2011;185:1504-1512
  35. 35. Wu GJ. Human METCAM/MUC18 as a novel biomarker to drive and its specific SiRNAs to block the malignant progression of prostate cancer. Journal of Cell Science and Therapy. 2015;6(5):1000227
  36. 36. Wu GJ. Human METCAM/MUC18 is a new diagnostic marker of and a driver for promoting and its specific siRNAs, derived oligopeptides and antibodies be used for decreasing the malignant progression of prostate cancer. Journal of Stem Cell Research & Therapeutics. 2016;1(5):00035
  37. 37. Wu GJ. Dual role of METCAM in the progression of different cancers. Journal of Oncology. 2012;2012:853797. DOI: 10.1155/2012/853797
  38. 38. Lehmann JM, Reithmuller G, Johnson JP. MUC18, a marker of tumor progression in human melanoma. Proceedings of the National Academy of Sciences of the United States of America. 1989;86:9891-9895
  39. 39. Meier F, Caroli U, Satyamoorthy K, Schittek B, Bauer J, Berking C, et al. Fibroblast growth factor-2 but not Mel-CAM and/or β3 integrin promotes progression of melanocytes to melanoma. Experimental Dermatology. 2003;12:296-306
  40. 40. Wu GJ. METCAM/MUC18 expression and cancer metastasis. Current Genomics. 2005;6:333-349
  41. 41. Anfosso F, Bardin N, Frances V, Vivier E, Camoin-Jau L, Sampol J, et al. Activation of human endothelial cells via S-Endo-1 antigen (CD146) stimulates the tyrosine phosphorylation of focal adhesion kinase p125FAK. The Journal of Biological Chemistry. 1998;273:26852-26858
  42. 42. Wu GJ, Wu MWH, Wang SW, Liu Z, Peng Q , Qu P, et al. Isolation and characterization of the major form of human MUC18 cDNA gene and correlation of MUC18 over-expression in prostate cancer cells and tissues with malignant progression. Gene. 2001;279:17-31
  43. 43. Yang H, Wang SWC, Liu Z, Wu MWH, McAlpine B, Ansel J, et al. Isolation and characterization of murine MUC18 cDNA gene, and correlation of MUC18 expression in murine melanoma cell lines with metastatic ability. Gene. 2001;265:133-145
  44. 44. Shih IM. The role of CD146 (Mel-CAM) in biology and pathology. Journal of Pathology. 1999;189:4-11
  45. 45. Zeng Q , Li W, Lu D, Wu Z, Duan H, Luo Y, et al. CD146, an epithelial-mesenchymal transition inducer, is associated with triple-negative breast cancer. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(4):1127-1132
  46. 46. Li Q , Yu Y, Bischoff J, Milliken JB, Olsen BR. Differential expression of CD146 in tissues and endothelial cells derived from infantile hemangioma and normal human skin. Journal of Pathology. 2003;201:296-302
  47. 47. Lin CT, Wong CI, Chan WY, et al. Establishment and characterization of two nasopharyngeal carcinoma cell lines. Laboratory Investigation. 1990;62:713-724
  48. 48. Aldovini D, Demichelis F, Doglioni C, Di Vizio D, Galligioni E, et al. M-CAM expression as marker of poor prognosis in epithelial ovarian cancer. International Journal of Cancer. 2006;119(8):1920-1926
  49. 49. Wu Z, Wu ZY, Li J, Yang X, Wang Y, et al. MCAM is a novel metastasis marker and regulates spreading, apoptosis and invasion of ovarian cancer cells. Tumor Biology. 2012;33:1619-1628
  50. 50. Wu GJ. Enforced expression of METCAM/MUC18 decreases in vitro motility and invasiveness and tumorigenesis and in vivo tumorigenesis of human ovarian cancer BG-1 cells. In: Schatten H, editor. Ovarian Cancer: Molecular & Diagnostic Imaging and Treatment Strategies, Advances in Experimental Medicine and Biology. Humana Press (Springer Science + Business Media LLC); 2019. (In press)
  51. 51. Wu G-J, Chang YR, Chu JT. METCAM/MUC18 plays a positive role in the tumorigenesis of human prostate cancer DU145 cells: Knockdown effects shRNAs decreasing tumorigenicity in nude mice. 2019. (Submitted)
  52. 52. Wu G-J, Chang YR, Chu JT. METCAM/MUC18 plays a negative role in the tumorigenesis of human prostate cancer PC-3 cells: Knockdown effects shRNAs increasing tumorigenicity in nude mice. 2019. (Submitted)
  53. 53. Zeng P, Li H, Lu PH, Zhou LN, Tang M, Liu CY, et al. Prognostic value of CD146 in solid tumor: A systematic review and meta-analysis. Scientific Reports. 2017;7(1):4223. DOI: 10.1038/s41598-017-01061-3
  54. 54. Liu D, Du L, Chen D, Ye Z, Duan H, Tu T, et al. Reduced CD146 expression promotes tumorigenesis and cancer stemness in colorectal cancer through activating Wnt/β-catenin signaling. Oncotarget. 2016;7(26):40704-40718
  55. 55. Stalin J, Nollet M, Garigue P, Fernandez S, Vivavancos L, Essaah A, et al. Targeting soluble CD146 with a neutralizing antibody inhibits vascularization, growth, and survival of CD146 positive tumors. Oncogene. 2016;35:5489-5500
  56. 56. Zheng B, Ohuchida K, Chijiiwa Y, Zhao M, Mizuuchi Y, Cui L, et al. CD146 attenuation in cancer-associated fibroblasts promotes pancreatic cancer progression. Molecular Carcinogenesis. 2016;55(11):1560-1572
  57. 57. Liu WF, Ji SR, Sun JJ, Zhang Y, Liu ZY, Liang AB, et al. Gastric cancer CD146 expression correlates with epithelial-mesenchymal transition markers and a poor prognosis in gastric cancer. International Journal of Molecular Sciences. 2012;13:6399-6406
  58. 58. Yang Y, Hernandez R, Rao J, Yin L, Qu Y, Wu J, et al. Targeting CD146 with a 64Cu-labeled antibody enables in vivo immunoPET imaging of high-grade gliomas. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(47):E6525-E6534
  59. 59. Jiang G, Zhang L, Zhu Q , Bai D, Zhang C, Wang X. CD146 promotes metastasis and predicts poor prognosis of hepatocellular carcinoma. Journal of Experimental & Clinical Cancer Research. 2016;35:38. DOI: 10.1186/s13046-016-0313-3
  60. 60. Chen K, Ding A, Ding Y, Ghanekar A. High-throughput flow cytometry screening of human hepatocellular carcinoma reveals CD146 to be a novel marker of tumor-initiating cells. Biochemistry and Biophysics Reports. 2016;8:107-113
  61. 61. Kristiansen G, Yu Y, Schlüns K, Sers C, Dietel M, Petersen I. Expression of the cell adhesion molecule CD146/MCAM in non-small cell lung cancer. Analytical Cellular Pathology. 2003;25:77-81
  62. 62. Oka S, Uramoto H, Chikaishi Y, Tanaka F. The expression of CD146 predicts a poor overall survival in patients with adenocarcinoma of the lung. Anticancer Research. 2012;32:861-864
  63. 63. Zhang X, Wang Z, Kang Y, Li X, Ma X, Ma L. MCAM expression is associated with poor prognosis in non-small cell lung cancer. Clinical and Translational Oncology. 2014;16:178-183
  64. 64. Zhang F, Wang J, Wang X, Wei N, Liu H, Zhang X. CD146-mediated acquisition of stemness phenotype enhances tumor invasion and metastasis after EGFR-TKI resistance in lung cancer. Clinical Respiratory Journal. 2019;13(1):23-33
  65. 65. England CG, Jiang D, Hernandez R, Sun H, Valdovinos HF, Ehlerding EB, et al. ImmunoPET imaging of CD146 in murine models of intrapulmonary metastasis of non-small cell lung cancer. Molecular Pharmaceutics. 2017;14(10):3239-3247
  66. 66. Tripathi SC, Fahrmann JF, Celiktas M, Aguilar M, Marini KD, Jolly MK, et al. A novel mechanism of chemoresistance in small cell lung cancer mediated by MCAM via PI3K/AKT/SOX2 signaling pathway. Cancer Research. 2017;77(16):4414-44252
  67. 67. Schiano C, Grimaldi V, Casamassimi A, Infante T, Esposito A, Giovane A, et al. Different expression of CD146 in human normal and osteosarcoma cell lines. Medical Oncology. 2012;29(4):2998-3002
  68. 68. McGary EC, Heimberger A, Mills L, Weber K, Thomas GW, Shtivelband M, et al. A fully human anti-melanoma cellular adhesion molecule/MUC18 antibody inhibits spontaneous pulmonary metastasis of osteosarcoma cells in vivo. Clinical Cancer Research. 2003;9:6560-6566
  69. 69. Westrøm S, Bønsdorff TB, Abbas N, Bruland ØS, Jonasdottir TJ, Mælandsmo GM, et al. Evaluation of CD146 as target for radioimmunotherapy against osteosarcoma. PLoS One. 2016;11(10):e0165382. DOI: 10.1371/journal.pone.0165382
  70. 70. von Burstin J, Bachhuber F, Paul M, Schmid RM, Rustgi AK. The TALE homeodomain transcription factor MEIS1 activates the pro-metastatic melanoma cell adhesion molecule MCAM to promote migration of pancreatic cancer cells. Molecular Carcinogenesis. 2017;56(3):936-944
  71. 71. LaMontagne KR Jr, Moses MA, Wiederschain D, Mahajan S, Holden J, Ghazizadeh H, et al. Inhibition of MAP kinase causes morphological reversion and dissociation between soft agar growth and in vivo tumorigenesis in angiosarcoma cells. American Journal of Pathology. 2000;157:1937-1945
  72. 72. Xie S, Luca M, Huang S, Gutman M, Reich R, Johnson JP, et al. Expression of MCAM/MCU18 by human melanoma cells leads to increased tumor growth and metastasis. Cancer Research. 1997;57:2295-2303
  73. 73. Schlagbauer-Wadl H, Jansen B, Muller M, Polterauer P, Wolff K, Eichler HG, et al. Influence of MUC18/MCAM/CD146 expression on human melanoma growth and metastasis in SCID mice. International Journal of Cancer. 1999;81:951-955
  74. 74. Geiser M, Cebe R, Drewello D, Schmitz R. Integration of PCR fragments at any specific site within cloning vectors without the use of restriction enzymes and DNA ligase. BioTechniques. 2001;31:88-92
  75. 75. Morgan MR, Thomas GJ, Russell A, Hart IR, Marshall JF. The integrin cytoplasmic tail motif EKQKVDLSTDC is sufficient to promote tumor cell invasion mediated by matrix metalloproteinase MMP-2 or MMP-9. The Journal of Biological Chemistry. 2004;279:26533-26539
  76. 76. Heiz M, Grunberg J, Schubiger PA, Novak-Hofer I. Hepatocyte growth factor-induced ectodomain shedding of cell adhesion molecule L1-role of the cytoplasmic domain. The Journal of Biological Chemistry. 2004;279:31149-31156
  77. 77. Anfosso F, Bardin N, Vivier E, Sabatier F, Sampol J, Dignat-George F. Outside-in signaling pathway linked to CD146 engagement in human endothelial cells. The Journal of Biological Chemistry. 2001;276:1564-1569
  78. 78. Sastry SK, Horwitz AF. Integrin cytoplasmic domains: Mediators of cytoskeletal linkages and extra- and intracellular initiated transmembrane signaling. Current Opinion in Cell Biology. 1993;5:819-831
  79. 79. Schoenwaelder SM, Burridge K. Bidirectional signaling between the cytoskeleton and integrins. Current Opinion in Cell Biology. 1999;11:274-286
  80. 80. Sers C, Kirsch K, Rothbacher U, Riethmuller G, Johnson JP. Genomic organization of the melanoma-associated glycoprotein MUC18: Implications for the evolution of the immunoglobulin domains. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:8514-8518
  81. 81. Rummel MM, Sers C, Johnson JP. Phorbol ester and cyclic AMP-mediated regulation of the melanoma-associated cell adhesion molecule MUC18/MCAM. Cancer Research. 1996;56:2218-2223
  82. 82. Jean D, Gershenwald JE, Huang S, Luca M, Hudson MJ, Tainsky MA, et al. Loss of AP-2 results in up-regulation of CAM/MUC18 and an increase in tumor growth and metastasis of human melanoma cells. The Journal of Biological Chemistry. 1998;273:16501-16508
  83. 83. Ruiz M, Pettaway C, Song R, Stoeltzing O, Ellis L, Bar-Eli M. Activator protein 2a inhibits tumorigenicity and represses vascular endothelial growth factor transcription in prostate cancer cells. Cancer Research. 2004;64:631-638
  84. 84. Ness SA. The Myb oncoprotein: Regulating a regulator. Biochimica et Biophysica Acta. 1996;1299:F123-F139
  85. 85. Thomson JAF, Murphy K, Baker E, Sutherland GR, Parsons PG, Sturm RA. The brn-2 gene regulates the melanocytic phenotype and tumorigenic potential of human melanoma cells. Oncogene. 1995;11:691-700
  86. 86. Yordy JS, Li R, Sementchenko VI, Pei H, Muise-Helmericks RC, Watson DK. S100 expression modulates ETS1 transcriptional activity and inhibits cell invasion. Oncogene. 2004;23:6654-6665
  87. 87. L’honore A, Lamb NJ, Vandromme M, Turowski P, Carnac G, Fernandez A. MyoD distal regulatory region contains an SF binding CArG element required for MyoD expression in skeletal myoblasts and during muscle regeneration. Molecular Biology of the Cell. 2003;14:2151-2162
  88. 88. Baron V, Duss S, Rhim J, Mercola D. Antisense to the early growth response-1 gene (Egr-1) inhibits prostate tumor development in TRAMP mice. Annals of the New York Academy of Sciences. 2003;1002:197-216
  89. 89. O’Brien RM, Streeper RS, Ayala JE, Stadelmaier BT, Hornbuckle LA. Insulin-regulated gene expression. Biochemical Society Transactions. 2001;29:552-558
  90. 90. Johnson JP, Rummel MM, Rothbacher U, Sers C. MUC18: A cell adhesion molecule with a potential role in tumor growth and tumor cell dissemination. Current Topics in Microbiology and Immunology. 1997;213:95-106
  91. 91. Sechler M, Parrish JK, Birks DK, Jedlicka P. The histone demethylase KDM3A, and its downstream target MCAM, promote Ewing Sarcoma cell migration and metastasis. Oncogene. 2017;36(29):4150-4160
  92. 92. Lung HL, Cheng Y, Kumaran MK, et al. Fine mapping of the 11Q22-23 tumor suppressive region and involvement of TSLC1 in nasopharyngeal carcinoma. International Journal of Cancer. 2004;112:628-635
  93. 93. Wu GJ. MCAM (melanoma cell adhesion molecule). Atlas of Genetics and Cytogenetics in Oncology and Haematology (Open Access Journal). 2012. DOI: 10.4267/2042/47418. ID41314ch11q23. The manually annotated Biomax Human Genome Database Version 4.0, Biomax Informatics AG, www.biomax.com, the Biomax Solutions Inc., Falmouth, MA, USA
  94. 94. Brait M, Banerjee M, Maldonado L, Ooki A, Loyo M, Guida E, et al. Promoter methylation of MCAM, ERα and ERβ in serum of early stage prostate cancer patients. Oncotarget. 2017;8(9):15431-15440. DOI: 10.18632/oncotarget.14873
  95. 95. Parodi A. Protein glycosylation and its role in protein folding. Annual Review of Biochemistry. 2000;69:69-93
  96. 96. Varki A. Glycosylation changes in tumor cells. In: Varki A, Cumming R, Esko J, Freeze H, Hart G, Marth J, editors. Essentials of glycobiology. NY: Cold Spring Harbor Laboratory Press; 1999. pp. 537-549
  97. 97. Fukuda M. Possible roles of tumor-associated carbohydrate antigens. Cancer Research. 1996;56:2237-2244
  98. 98. Hakomori S. Tumor malignancy defined by aberrant glycosylation and sphingo(glyco) lipid metabolism. Cancer Research. 1996;56:5309-5318
  99. 99. Yamamoto H, Oviedo A, Sweeley C, Saito T, Moskal JR. α2,6-sialylation of cell-surface N-glycans inhibits glioma formation in vivo. Cancer Research. 2001;61:6822-6829
  100. 100. Lehmann JM, Holzmann B, Breitbart EW, Schmiegelow P, Riethmuller G, Johnson JP. Discrimination between benign and malignant cells of melanocytic lineage by two novel antigens, a glycoprotein with a molecular weight of 113,000 and a protein with a molecular weight of 76,000. Cancer Research. 1987;47:841-845
  101. 101. Sumardika W, Youyi C, Kondo E, Inoue Y, Ruma MW, Murata H, et al. β-1,3-galactosyl-O-glycosyl-glycoprotein β-1,6-N-acetylglucosaminyltransferase 3 increases MCAM stability, which enhances S100A8/A9-mediated cancer motility. Oncology Research. 2018;26(3):431-444
  102. 102. Aguirre-Chiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nature Reviews. Cancer. 2007;7:834-846
  103. 103. Kasinski AL, Slack FJ. MicroRNAs en route to the clinic: Progress in validating and targeting microRNAs for cancer therapy. Nature Reviews. Cancer. 2011;11:849-864
  104. 104. Wikman H, Vessella R, Pantel K. Cancer micro-metastasis and tumor dormancy. APMIS. 2008;116:754-770
  105. 105. Smith SC, Theodorescu D. Learning therapeutic lessons from metastasis suppressor proteins. Nature Reviews. Cancer. 2009;9(4):253-264
  106. 106. Naldini L. Gene therapy returns to center stage. Nature. 2015;526(7573):351-360. DOI: 10.1038/nature15818
  107. 107. Liang G, Weisenberger DJ. DNA methylation aberrancies as a guide for surveillance and treatment of human cancers. Epigenetics. 2017;12(6):416-432
  108. 108. Shanmugam MK, Arfuso F, Arumugam S, Chinnathambi A, Jinsong B, Warrier S, et al. Role of novel histone modifications in cancer. Oncotarget. 2018;9(11):11414-11426

Written By

Guang-Jer Wu

Submitted: 01 November 2018 Reviewed: 10 April 2019 Published: 18 May 2019