Abstract
Objective: In recent years, overexpression of FAM3C protein has been proved to contribute to epithelial to mesenchymal transition (EMT) and correlate with poor prognosis in several malignant tumors. However, the role of FAM3C in gastric cancer (GC) is still not clear. Thus, we detected the expression of FAM3C by immunohistochemistry (IHC) and determined the association of FAM3C expression with EMT, clinicopathologic characteristics, and prognosis in GC. Methods: We detected the expression of FAM3C, PDGFR-β, E-cadherin, and vimentin in 150 patients with GC by tissue chip technology and IHC methods. All statistical analyses were conducted using SPSS 22.0 software. Results: FAM3C expression in gastric carcinoma tissues was significantly higher than in matched adjacent normal tissues (P = 0.037). The expression of FAM3C positively correlated with vimentin expression and negatively correlated with E-cadherin expression (P = 0.045 and 0.029, respectively). However, there was no correlation between expression of FAM3C and PDGFR-β (P = 0.095). FAM3C overexpression was significantly associated with depth of invasion, lymph node metastasis and TNM stage (P = 0.004, 0.016 and 0.022, respectively). Multivariate analysis revealed that high expression of FAM3C is an independent prognostic factor for poor prognosis in GC patients (P = 0.007). Conclusions: Overexpression of FAM3C is a potential marker for EMT and predicts poor outcome in gastric cancer.
Keywords: FAM3C, gastric cancer, prognosis, epithelial-mesenchymal transition
Introduction
Although the incidence of gastric cancer/(GC)has been decreasing, it still remains the fifth most common cancer and the third leading cause of cancer-related death in the world [1]. In GC, metastasis is the main cause of cancer-related death [2]. Tumor metastasis is a multi-step process in which malignant tumor cells move from the primary site to the other parts throughlymphatic, vascular and other pathways. In this metastatic process, epithelial-to-mesenchymal transition (EMT) has been proven to play an important role [3,4]. Epithelial-mesenchymal transition (EMT), a developmental process in which epithelial cells acquire a mesenchymal cell phenotype, may promote tumor development, invasion or metastasis. In EMT, cells gradually lose the epithelial characteristics of intercellular adhesion, thereby acquiring migratory fibroblastoid properties, and becoming resistant to apoptosis [5]. EMT is characterized by downregulation of epithelial markers such as E-cadherin and upregulation of mesenchymal markers such as vimentin [6,7]. Accumulating evidence suggests that loss of E-cadherin expression and positive expression of vimentin play important roles in cancer invasion and metastasis, and EMT may predict a relatively poor prognosis [8-10]. EMT may be induced by secreted factors of diverse cells which a re recruited into the tumor microenvironment by tumor cells [11].
FAM3C, also called interleukine-like EMT inducer (ILEI), is one of the important secreted factors which can induce EMT [12]. FAM3C belongs to the FAM3 cytokine family. All four members of the family (FAM3A, B, C and D) can encode a protein with a hydrophobic leader sequence, which contains 224-235 amino acids [13]. FAM3C gene is located on chromosome 7 (7q31), which was initially considered as a candidate gene for autosomal recessive non-syndromic hearing loss [14]. Moreover, FAM3C is also considered to be associated with the occurrence and development of tumor [15]. And high levels of FAM3C have been found in human esophagus, breast, colon, prostate, lung, liver, head and neck tumors [16]. Recently, Zhu et al. [17] showed that FAM3C expression is upregulated in esophageal squamous cell carcinoma and is associated with aggressive tumor behavior, metastasis and poor clinical outcome. Moreover, Gao et al. [18] demonstrated that overexpression of FAM3C is significantly associated with tumor metastasis and poor prognosis in colorectal cancer. However, the expression and clinical significance of FAM3C in GC patients remains not yet well-documented. Moreover, several works have shown that FAM3C was a key regulator of EMT and contributed to metastatic progression in both human and animal models [16,19]. Moreover, downregulation of FAM3C could reduce the induction of EMT [19,20]. Furthermore, Lahsnig et al. [21] revealed that FAM3C could stimulate the upregulation of PDGFR-β and govern hepatocellular EMT through mechanisms involving PDGFR-β signaling in liver carcinoma. Guo et al. [22] have revealed that PDGFR-β was found to be highly expressed in gastric cancer and PDGFR-β signaling might induce EMT to promote metastasis of gastric carcinoma. However, whether FAM3C expression has a connection with PDGFR-β and EMT in gastric cancer remains unclear.
In the present study, immunohistochemistry (IHC) was used to evaluate the expression of FAM3C, PDGFR-β, E-cadherin and vimentin. We used downregulation of E-cadherin and upregulation of vimentin to evaluate the occurrence of EMT. Ultimately, we aim to investigate the correlation of FAM3C expression with PDGFR-β, EMT, clinicopathological characteristics, and prognosis in a number of GC cases.
Materials and methods
Patients and tissue samples
GC tissues and matched adjacent normal tissues were obtained from 150 GC patients who underwent surgical resection at Zhongnan Hospital of Wuhan University from 2010 to 2012. No patient had received preoperative chemotherapy or radiotherapy. The clinicopathological information of all the GC patients, including age, gender, tumor size, tumor location, histological grade, depth of invasion, lymph node metastasis status, and pathological TNM stage (pTNM stage), was collected from the Department of Oncology, Zhongnan Hospital of Wuhan University (Table 1). Pathological diagnoses were performed independently by two experienced pathologists. TNM staging of GC was conducted based on American Joint Committee on Cancer [23]. The study was approved by the ethics committee of Zhongnan Hospital of Wuhan University. Written informed consent for the use of tumor tissues was obtained from all the GC patients before the surgical resections. All the patients were followed up by telephone enquiry or mail communications. The final follow-up was completed in December 2017. The follow-up time ranged from 3 to 96 months (mean 57 months).
Table 1.
Correlation between FAM3C, PDGFR, E-cadherin, and vimentin expression and clinicopathological characteristics of patients with gastric carcinoma
| Variables | n | FAM3C | P value | PDGFR | P value | E-cadherin | P value | Vimentin | P value | ||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
|
|
|
|
|
||||||||||
| IS High (>6) | IS Low (≤6) | IS High (>6) | IS Low (≤6) | IS High (>4) | IS Low (≤4) | Positive | Negative | ||||||
| Age | 0.943 | 0.507 | 0.062 | 0.079 | |||||||||
| <60 | 74 | 52 | 22 | 41 | 33 | 24 | 50 | 18 | 56 | ||||
| ≥60 | 76 | 53 | 23 | 38 | 38 | 36 | 40 | 10 | 66 | ||||
| Gender | 0.741 | 0.309 | 0.537 | 0.254 | |||||||||
| Male | 93 | 66 | 27 | 52 | 41 | 39 | 54 | 20 | 73 | ||||
| Female | 57 | 39 | 18 | 27 | 30 | 21 | 36 | 8 | 49 | ||||
| Tumor size | 0.093 | 0.393 | 0.385 | ||||||||||
| <5 cm | 71 | 45 | 26 | 40 | 31 | 31 | 40 | 16 | 55 | 0.279 | |||
| ≥5 cm | 79 | 60 | 19 | 39 | 40 | 29 | 50 | 12 | 67 | ||||
| Histological grade | 0.969 | 0.536 | 0.001 | 0.088 | |||||||||
| Well and moderate | 47 | 33 | 14 | 23 | 24 | 28 | 19 | 5 | 42 | ||||
| Poor and others | 103 | 72 | 31 | 56 | 47 | 32 | 71 | 23 | 80 | ||||
| Tumor location | 0.406 | 0.607 | 0.368 | 0.7 | |||||||||
| Upper | 24 | 17 | 7 | 13 | 11 | 12 | 12 | 4 | 20 | ||||
| Middle | 51 | 39 | 12 | 24 | 27 | 17 | 34 | 8 | 43 | ||||
| Lower | 75 | 49 | 26 | 42 | 33 | 31 | 44 | 16 | 59 | ||||
| Depth of invasion | 0.004 | 0.002 | 0.029 | 0.12 | |||||||||
| T1+T2 | 45 | 24 | 21 | 15 | 30 | 24 | 21 | 5 | 40 | ||||
| T3+T4 | 105 | 81 | 24 | 64 | 41 | 36 | 69 | 23 | 82 | ||||
| Lymph node metastasis | 0.016 | 0.004 | 0.047 | 0.038 | |||||||||
| Absent | 58 | 34 | 24 | 22 | 36 | 29 | 29 | 6 | 52 | ||||
| Present | 92 | 71 | 21 | 57 | 35 | 31 | 61 | 22 | 70 | ||||
| TNM stage | 0.022 | <0.0001 | 0.029 | 0.032 | |||||||||
| I-II | 59 | 35 | 24 | 18 | 41 | 30 | 29 | 6 | 53 | ||||
| III-IV | 91 | 70 | 21 | 61 | 30 | 30 | 61 | 22 | 69 | ||||
IS, immunoreactivity score; T1, tumor invades lamina propria, muscularis mucosae, or submucosa; T2, tumor invasion of the muscularis propria; T3, tumor invasion subserosal connective tissue; T4, tumor invasion serosal or adjacent structures.
Preparation of tissue chips
150 cases of GC paraffin tissue blocks and 40 cases of matched adjacent normal paraffin tissue blocks were sent to Shanghai Outdo Biotech Company and made into tissue microarray. Through HE stained tumor sections, the donor wax blocks were selected and marked. Two different sampling sites were selected for each sample, and the donor tissue cores were placed in the receptor wax block through punching needle. Then, a dot matrix was arranged on the receptor wax block in the order of prior design, and then they were routinely sliced and stained with HE, which was confirmed by the senior pathologist.
Antibodies and reagents
The primary antibodies used were rabbit polyclonal anti-human FAM3C (14247-1-AP, Proteintech, Wuhan, China), rabbit polyclonal anti-human PDGFR-β (3169T, Cell Signaling Technology, Danvers, USA), rabbit polyclonal anti-human E-cadherin (20874-1-AP, Proteintech, Wuhan, China) and rabbit polyclonal anti-human vimentin (10366-1-AP, Proteintech, Wuhan, China).
Immunohistochemistry
We used immunohistochemistry (IHC) to analyze the expression of FAM3C, PDGFR-β, E-cadherin and vimentin in human GC. Briefly, the sections were dewaxed in xylene and then dehydrated in ascending concentrations of ethanol. Next, the slices were heated for 15 min to repair the antigen by using electric pottery furnace and then cooled down to room temperature. Subsequently, the sections were rinsed with phosphate-buffered saline (PBS) (3×3 min) and then blocked with 3% peroxide for 15 min at room temperature for endogenous peroxidase ablation. After being blocked with normal goat serum for 30 min at room temperature, sections were incubated overnight at 4°C with primary antibodies at the following dilutions: anti-FAM3C, 1:50; anti-PDGFR-β, 1:100; anti-E-cadherin, 1:100; and anti-vimentin, 1:100. After being washed with PBS (3×3 min), sections were incubated with horseradish peroxidase-labeled secondary antibodies (goat anti-rabbit for FAM3C, PDGFR-β, E-cadherin and vimentin) for 20 min at 37°C. Then, the sections were immersed in 3,3-diaminobenzidin (DAB) for 10 min at room temperature without light. Finally, all the sections were counterstained with hematoxylin and mounted. PBS was used instead of the primary antibody as a negative control.
Evaluation of immunostaining
All the sections were evaluated repetitively and independently by two experienced pathologists who were blind to both clinical and pathological data. All of the disagreements were resolved by the third investigator. According to those established methods, both of the stain intensity and proportion of the positive cells were evaluated. The staining intensity was scored on a 0-3 scale: 0, no staining; 1, weak staining; 2, moderate staining; 3, strong staining. The percentage of positive cells was scored on a 0-4 scale: 0, ≤5% positive tumor cells; 1, 6-25% positive tumor cells; 2, 26-50% positive tumor cells; 3, 51-75% positive tumor cells; 4, 76-100% positive tumor cells. We assessed the percentage of positive cells through calculating the percentage of positive tumor cells among the entire carcinoma-involved area. All the slides were examined by light microscope and five random fields (400× magnification) were selected. The immunoreactivity score (IS; 0-12) was generated by multiplying the intensity and reactivity rates [24,25]. For FAM3C and PDGFR-β, an IS score of 06 was considered low expression, and scores above 6 were high expression [18,26]. For E-cadherin, an overall staining score of 4 or less was considered as down-regulated, and scores above 4 were high [18]. However, expression of vimentin was considered positive when more than 10% of tumor cells showed positive vimentin staining and negative when 10% or less of tumor cells showed positive vimentin staining according to previously published methods [27,28].
Statistical analysis
All statistical analyses were conducted using SPSS 22.0 software. The differences in the protein expression patterns between gastric cancerous and paracancerous tissues were analyzed by the chi-square test. The chi-square test was also adopted for analysis of associations between FAM3C expression and clinicopathological parameters of GCs. The associations of FAM3C expression with PDGFR-β, E-cadherin and vimentin expression were analyzed with the nonparametric Spearman rank correlation coefficient. Survival curves were assessed by the Kaplan-Meier method and intergroup differences were analyzed with the log-rank test. Univariate and multivariate analysis were performed to evaluate the independence of significant prognostic factors by Cox proportional hazards regression model which was used to calculate hazard ratios (HR) of mortality for multiple clinicopathological variables and protein markers. Two-tailed p-value <0.05 was considered statistically significant.
Result
Expression of FAM3C, PDGFR-β, E-cadherin and vimentin in GC
IHC was conducted to analyze expression of FAM3C in 150 cases of GC tissues and 40 cases of matched adjacent normal tissues. As shown in Figure 1, positive staining of the FAM3C protein was most obviously detected in the cytoplasm of tumor cells. The proportion of high FAM3C expression was 70% (105/150) in GC and 52.5% (21/40) in matched adjacent normal tissues (Table 2). The chi-square test indicated that FAM3C expression in GC tissues was significantly higher than that in matched adjacent normal tissues (P<0.05).
Figure 1.

IHC staining of FAM3C in GC and corresponding paracancerous tissue. The staining of FAM3C protein (brown) was mainly located in the cytoplasm of GC tumor cells (×200): (A) High FAM3Cexpression in GC, (B) High FAM3C expression in non-neoplastic tissue, (C) Low FAM3C expression in GC, and (D) Low FAM3C expression in non-neoplastic tissue. GC, gastric cancer; IHC, immunohistochemical.
Table 2.
Expression of FAM3C in gastric cancer and normal gastric mucosa
| FAM3C expression | P value | ||
|---|---|---|---|
|
| |||
| High (%) | Low (%) | ||
| Gastric cancerous tissue (n = 150) | 105 (70%) | 45 (30%) | 0.037 |
| Paracancerous tissue (n = 40) | 21 (52.5%) | 19 (47.5%) | |
Immunohistochemical staining for PDGFR-β was mainly detected in the cytoplasm of cancer cells and surrounding stromal cells (Figure 2). PDGFR expression in GC tissues was significantly upregulated. Among the GC cases, 79 of 150 (52.7%) showed high PDGFR expression.
Figure 2.
Immunohistochemical staining for FAM3C, PDGFR-β, E-cadherin and vimentin in gastric cancer(×200): (A) High FAM3C expression, (B) High PDGFR-β expression, (C) High E-cadherin expression and (D) Positive vimentin expression; (E) Low FAM3C expression, (F) Low PDGFR-β expression, (G) Low E-cadherin expression and (H) Negative vimentin expression.
As shown in Figure 2, immunoreactivity for E-cadherin was present predominantly at the cell membrane. Expression of E-cadherin in GC tissues was significantly downregulated. The proportion of E-cadherin low expression was 60% (90/150) in GC tissues.
Vimentin was predominantly observed in the cytoplasm of GC cells. The percentage of vimentin positive expression was 18.7% (28/150) in GC tissues. Representative images are shown in Figure 2.
Associations of FAM3C expression with PDGFR-β, E-cadherin and vimentin expression in GC
As shown in Table 3, the Spearman rank test showed that high expression of FAM3C had obviously positive correlation with vimentin expression (r = 0.164, P = 0.045) and negative correlation with E-cadherin expression (r = -0.178, P = 0.029). However, there is no correlation between expression of FAM3C and PDGFR-β (r = 0.137, P = 0.095).
Table 3.
Correlation between FAM3C, PDGFR, E-cadherin, and vimentin expression in gastric cancer
| Variable | FAM3C expression | r | P | |
|---|---|---|---|---|
|
| ||||
| High | Low | |||
| PDGFR expression | 0.137 | 0.095 | ||
| High | 60 | 19 | ||
| Low | 45 | 26 | ||
| E-cadherin expression | -0.178 | 0.029 | ||
| High | 36 | 24 | ||
| Low | 69 | 21 | ||
| Vimentin expression | 0.164 | 0.045 | ||
| Positive | 24 | 4 | ||
| Negative | 81 | 41 | ||
Correlations of clinicopathologic characteristics with FAM3C, PDGFR-β, E-cadherin, and vimentin expression
The relationships between FAM3C, PDGFR-β, E-cadherin, and vimentin expression and clinicopathologic characteristics are shown in Table 1. High expression of FAM3C and PDGFR-β was significantly associated with depth of invasion (P = 0.004 and 0.002, respectively), lymph node metastasis (P = 0.016 and 0.004, respectively), and TNM stage (P = 0.022 and P<0.0001, respectively), but not with tumor size, tumor location or histologic grade (P>0.05). The down-regulated expression of E-cadherin was significantly associated with histological grade (P = 0.001), depth of invasion (P = 0.029), lymph node metastasis, (P = 0.047) and TNM stage (P = 0.029), but not with tumor size or location (P>0.05). The positive expression of vimentin was significantly associated with lymph node metastasis (P = 0.038) and TNM stage (P = 0.032), but not correlated with tumor size, tumor location, histological grade, or depth of invasion (P>0.05).
Survival analysis
Patients with high expression of FAM3C had significantly lower overall 1, 3, 5-year survival rates (92.4%, 72.4% and 58.1%, respectively) than those expressing low levels of FAM3C (97.8%, 91.1% and 84.4%, respectively). As shown in Figure 3, Kaplan-Meier analysis indicated that overall survival (OS) of patients with high FAM3C expression was significantly shorter than those with low FAM3C expression (log-rank, P = 0.001). Patients displaying high PDGFR-β expression had a shorter survival duration compared to those with low expression (log-rank, P = 0.008). Similarly, the OS of patients with positive vimentin expression was significantly lower than those with negative expression (log-rank, P = 0.009). However, low E-cadherin expression had no significant impact on OS of GC patients (log-rank, P = 0.112).
Figure 3.

Analysis of OS relative to FAM3C, PDGFR-β, E-cadherin and vimentin expression levels: A. FAM3C expression: low versus high expression (P = 0.001). B. PDGFR-β expression: low versus high (P = 0.008). C. E-cadherin expression: low versus high (P = 0.112). D. Vimentin expression: negative versus positive (P = 0.009).
Univariate analysis indicated that FAM3C expression (P = 0.002), PDGFR-β expression (P = 0.009) and vimentin expression (P = 0.011) could serve as prognostic factors for GC patients (Table 4).
Table 4.
Univariate and multivariate analyses of survival in gastric cancer
| Variable | Patients (n) | Univariate analysis | Multivariate analysis | ||
|---|---|---|---|---|---|
|
|
|
||||
| HR (95% CI) | P | HR (95% CI) | P | ||
| Age | 0.806 (0.493-1.317) | 0.389 | |||
| <60 | 74 | ||||
| ≥60 | 76 | ||||
| Gender | 0.614 (0.359-1.049) | 0.074 | |||
| Male | 93 | ||||
| Female | 57 | ||||
| Tumor size | 1.455 (0.886-2.389) | 0.138 | |||
| <5 cm | 71 | ||||
| ≥5 cm | 79 | ||||
| Histological grade | 1.701 (0.964-3.002) | 0.067 | |||
| Well and moderate | 47 | ||||
| Poor and others | 103 | ||||
| Tumor location | 0.786 (0.575-1.075) | 0.132 | |||
| Upper | 24 | ||||
| Middle | 51 | ||||
| Lower | 75 | ||||
| Depth of invasion | 3.807 (1.87-7.747) | <0.0001 | |||
| T1+T2 | 45 | ||||
| T3+T4 | 105 | ||||
| Lymph node metastasis | 2.395 (1.372-4.183) | 0.002 | |||
| Absent | 58 | ||||
| Present | 92 | ||||
| TNM stage | 3.139 (1.748-5.635) | <0.0001 | 0.35 (0.194-0.63) | <0.0001 | |
| I-II | 59 | ||||
| III-IV | 91 | ||||
| FAM3C expression | 0.346 (0.176-0.678) | 0.002 | 0.393 (0.199-0.773) | 0.007 | |
| High | 105 | ||||
| Low | 45 | ||||
| PDGFR expression | 0.5 (0.297-0.842) | 0.009 | |||
| High | 79 | ||||
| Low | 71 | ||||
| E-cadherin expression | 1.52 (0.902-2.56) | 0.116 | |||
| High | 60 | ||||
| Low | 90 | ||||
| Vimentin expression | 0.481 (0.274-0.843) | 0.011 | |||
| Positive | 28 | ||||
| Negative | 122 | ||||
CI, Confidence interval; HR, Hazard ratio.
Multivariate analysis by using forward stepwise Cox regression analysis revealed that high expression of FAM3C (P = 0.007) and TNM stage (P<0.0001) were independent prognostic factors in GC patients (Table 4).
Discussion
In the current study, it was observed that overexpression of FAM3C was closely correlated with poor outcomes for GC patients and could serve as a novel marker for EMT. This is the first research investigating the relationship between FAM3C and EMT for its clinical significance in GC.
Recently, several works have shown that high levels of FAM3C were detected in many epithelial tumors and may be involved in tumor initiation, progression, and metastasis, which indicated that overexpression of FAM3C might be closely associated with the genesis and development of cancer [17,29]. Importantly, FAM3C overexpression has been proven to be an independent predictive factor for poor survival in various malignant tumors, such as esophageal squamous cell carcinoma [17] and colorectal cancer [18]. However, there was no prior research concerning the expression pattern and clinical significance of FAM3C in GC. In our study, we found that FAM3C expression in GC tissues was significantly higher than that in matched adjacent normal tissues. Statistical analysis showed that higher levels of FAM3C expression were detected in tumors with aggressive clinicopathological features including deeper invasion, lymph node metastasis and advanced TNM stage, indicating an underlying promotion of FAM3C in GC formation, progression, and metastasis. Furthermore, our study showed that high expression of FAM3C was significantly associated with relatively poor outcome for GC patients. Patients with high expression of FAM3C displayed a relatively lower rate of 5-year OS than those expressing low levels of FAM3C. Using multivariate analyses, our data showed that high FAM3C expression is an independent factor of poor prognosis for GC. Therefore, these findings suggested that FAM3C expression could act as a predictive factor for poor outcomes or a potential therapeutic target for the clinical management of GC.
Metastasis is one of the decisive factors that affect outcomes of cancer patients [30]. EMT has been found to be involved in many aspects of tumor metastasis [31]. Moreover, an increasing number of articles have demonstrated that FAM3C overexpression can induce EMT, thereby promoting tumor growth and metastasis in vitro and in vivo [16,21]. Previous research found that only covalent FAM3C self-assembly is essential for EMT induction, elevated tumor growth and metastasis [32]. Therefore, we have also investigated the expression of EMT-related proteins (E-cadherin and vimentin) in GC tissues by immunohistochemistry and have analyzed the relationship between FAM3C and these proteins. E-cadherin is a member of the epithelial cadherin family of glycoproteins, which play important roles in maintaining cell adhesion and intercellular connections [33]. The down-regulated expression of E-cadherin could result in an advanced histologic grade, tumor invasion, and metastasis [34]. Vimentin is a major component of intermediate-sized filaments present in the cytoskeleton, which is essentially type III intermediate filament protein [35]. Vimentin plays a significant role in maintaining cell shape and the integrity of the cytoplasm and stabilizing cytoskeletal interactions [36]. Several studies have demonstrated that positive expression of vimentin could be correlated with poor prognosis and high frequency of metastasis in patients with GC [37,38]. In this study, we found that increased vimentin and reduced E-cadherin were closely linked to metastasis, advanced TNM stage and poor outcomes in GC. Moreover, for the first time, we found that high expression of FAM3C had obviously positive correlation with vimentin expression and negative correlation with E-cadherin expression, which indicated that FAM3C could be a novel marker for epithelial-mesenchymal transition for GC. The result is consistent with some other studies of colorectal cancer [18], breast cancer [16] and liver carcinoma [21]. Howeverthe precise mechanisms of FAM3C involved in EMT are still not completely clear.
In recent investigations, several studies have demonstrated that FAM3C could mediate the occurrence of EMT through TGF-β signaling in murine epithelial cells [19,20]. FAM3C was required but not sufficient to induce TGF-β-mediated EMT [19]. However, Lahsnig et al. [21] demonstrated that FAM3C required cooperation with oncogenic Ras to induce and maintain EMT of hepatocytes in a TGF-β-independent fashion in human hepatocytes. Moreover, Waerner et al. [16] revealed that overexpression of FAM3C alone was enough to induce EMT and promote tumor growth and metastasis by TGF-β-independent signaling in murine mammary epithelial cell EpH4. These results indicated that overexpression of FAM3C could induce tumor cells to undergo EMT and the molecular mechanisms of FAM3C involved in the process of EMT might vary with the type of epithelial cell. Interestingly, upregulation of PDGF/PDGFR induced by FAM3C may be involved in both murine mammary epithelial EMT and hepatocellular EMT [16,21]. Therefore, we also detected the expression of PDGFR in GC tissues and assessed their potential relationship with FAM3C. The results showed that PDGFR-β was highly expressed in GC, and the high expression was significantly associated with depth of invasion, lymph node metastasis and TNM stage. However, we found that there was no correlation between expression of FAM3C and PDGFR-β in GC. Similarly, differing associations between FAM3C and PDGFR-β expression have been shown in different kinds of cancer, suggesting that FAM3C might induce and maintain EMT by PDGFR-β-independent signaling in GC. Further work regarding the association between FAM3C and PDGFR-β in GC isstill needed.
In conclusion, FAM3C is a potential marker of EMT in GC. Moreover, high expression of FAM3C may promote GC invasion and metastasis through the EMT, and is an independent predictive factor for poor prognosis in GC patients.
Acknowledgements
This research was supported by Natural Foundation of Hubei Province (Grant No. 2013CFB267) and Wuhan Science and Technology Key Project (Grant No. 2013060602010248).
Disclosure of conflict of interest
None.
References
- 1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108. doi: 10.3322/caac.21262. [DOI] [PubMed] [Google Scholar]
- 2.Zhong J, Chen Y, Wang LJ. Emerging molecular basis of hematogenous metastasis in gastric cancer. World J Gastroenterol. 2016;22:2434–2440. doi: 10.3748/wjg.v22.i8.2434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–890. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
- 4.Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest. 2009;119:1420–1428. doi: 10.1172/JCI39104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kim HP, Han SW, Song SH, Jeong EG, Lee MY, Hwang D, Im SA, Bang YJ, Kim TY. Testican-1-mediated epithelial-mesenchymal transition signaling confers acquired resistance to lapatinib in HER2-positive gastric cancer. Oncogene. 2014;33:3334–3341. doi: 10.1038/onc.2013.285. [DOI] [PubMed] [Google Scholar]
- 6.Wang Y, Yao B, Wang Y, Zhang M, Fu S, Gao H, Peng R, Zhang L, Tang J. Increased FoxM1 expression is a target for metformin in the suppression of EMT in prostate cancer. Int J Mol Med. 2014;33:1514–1522. doi: 10.3892/ijmm.2014.1707. [DOI] [PubMed] [Google Scholar]
- 7.Zhang H, Liu L, Wang Y, Zhao G, Xie R, Liu C, Xiao X, Wu K, Nie Y, Zhang H, Fan D. KLF8 involves in TGF-beta-induced EMT and promotes invasion and migration in gastric cancer cells. J Cancer Res Clin Oncol. 2013;139:1033–1042. doi: 10.1007/s00432-012-1363-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhu X, Li Y, Zhou R, Wang N, Kang S. Knockdown of E-cadherin expression of endometrial epithelial cells may activate Wnt/beta-catenin pathway in vitro. Arch Gynecol Obstet. 2018;297:117–123. doi: 10.1007/s00404-017-4560-0. [DOI] [PubMed] [Google Scholar]
- 9.Richardson AM, Havel L, Koyen AE, Konen JM, Shupe JA, Wiles WG, Martin WD, Grossniklaus H, Sica GL, Gilbert-Ross M, Marcus A. Vimentin is required for lung adenocarcinoma metastasis via heterotypic tumor cell-cancer-associated fibroblast interactions during collective invasion. Clin Cancer Res. 2018;24:420–432. doi: 10.1158/1078-0432.CCR-17-1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Iwatsuki M, Mimori K, Yokobori T, Ishi H, Beppu T, Nakamori S, Baba H, Mori M. Epithelial-mesenchymal transition in cancer development and its clinical significance. Cancer Sci. 2010;101:293–299. doi: 10.1111/j.1349-7006.2009.01419.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127:679–695. doi: 10.1016/j.cell.2006.11.001. [DOI] [PubMed] [Google Scholar]
- 12.Kral M, Klimek C, Kutay B, Timelthaler G, Lendl T, Neuditschko B, Gerner C, Sibilia M, Csiszar A. Covalent dimerization of interleukin-like epithelial-to-mesenchymal transition (EMT) inducer (ILEI) facilitates EMT, invasion, and late aspects of metastasis. FEBS J. 2017;284:3484–3505. doi: 10.1111/febs.14207. [DOI] [PubMed] [Google Scholar]
- 13.Zhu Y, Xu G, Patel A, McLaughlin MM, Silverman C, Knecht K, Sweitzer S, Li X, McDonnell P, Mirabile R, Zimmerman D, Boyce R, Tierney LA, Hu E, Livi GP, Wolf B, Abdel-Meguid SS, Rose GD, Aurora R, Hensley P, Briggs M, Young PR. Cloning, expression, and initial characterization of a novel cytokine-like gene family. Genomics. 2002;80:144–150. doi: 10.1006/geno.2002.6816. [DOI] [PubMed] [Google Scholar]
- 14.Greinwald JJ, Wayne S, Chen AH, Scott DA, Zbar RI, Kraft ML, Prasad S, Ramesh A, Coucke P, Srisailapathy CR, Lovett M, Van Camp G, Smith RJ. Localization of a novel gene for nonsyndromic hearing loss (DFNB17) to chromosome region 7q31. Am J Med Genet. 1998;78:107–113. [PubMed] [Google Scholar]
- 15.Halberg N, Sengelaub CA, Navrazhina K, Molina H, Uryu K, Tavazoie SF. PITPNC1 Recruits RAB1B to the Golgi Network to Drive Malignant Secretion. Cancer Cell. 2016;29:339–353. doi: 10.1016/j.ccell.2016.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Waerner T, Alacakaptan M, Tamir I, Oberauer R, Gal A, Brabletz T, Schreiber M, Jechlinger M, Beug H. ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell. 2006;10:227–239. doi: 10.1016/j.ccr.2006.07.020. [DOI] [PubMed] [Google Scholar]
- 17.Zhu YH, Zhang B, Li M, Huang P, Sun J, Fu J, Guan XY. Prognostic significance of FAM3C in esophageal squamous cell carcinoma. Diagn Pathol. 2015;10:192. doi: 10.1186/s13000-015-0424-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gao ZH, Lu C, Wang ZN, Song YX, Zhu JL, Gao P, Sun JX, Chen XW, Wang MX, Dong YL, Xu HM. ILEI: a novel marker for epithelial-mesenchymal transition and poor prognosis in colorectal cancer. Histopathology. 2014;65:527–538. doi: 10.1111/his.12435. [DOI] [PubMed] [Google Scholar]
- 19.Chaudhury A, Hussey GS, Ray PS, Jin G, Fox PL, Howe PH. TGF-beta-mediated phosphorylation of hnRNP E1 induces EMT via transcript-selective translational induction of Dab2 and ILEI. Nat Cell Biol. 2010;12:286–293. doi: 10.1038/ncb2029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hussey GS, Chaudhury A, Dawson AE, Lindner DJ, Knudsen CR, Wilce MC, Merrick WC, Howe PH. Identification of an mRNP complex regulating tumorigenesis at the translational elongation step. Mol Cell. 2011;41:419–431. doi: 10.1016/j.molcel.2011.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lahsnig C, Mikula M, Petz M, Zulehner G, Schneller D, van Zijl F, Huber H, Csiszar A, Beug H, Mikulits W. ILEI requires oncogenic Ras for the epithelial to mesenchymal transition of hepatocytes and liver carcinoma progression. Oncogene. 2009;28:638–650. doi: 10.1038/onc.2008.418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Guo Y, Yin J, Zha L, Wang Z. Clinicopathological significance of platelet-derived growth factor B, platelet-derived growth factor receptor-beta, and E-cadherin expression in gastric carcinoma. Contemp Oncol (Pozn) 2013;17:150–155. doi: 10.5114/wo.2013.34618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17:3077–3079. doi: 10.1245/s10434-010-1362-z. [DOI] [PubMed] [Google Scholar]
- 24.Sinicrope FA, Ruan SB, Cleary KR, Stephens LC, Lee JJ, Levin B. bcl-2 and p53 oncoprotein expression during colorectal tumorigenesis. Cancer Res. 1995;55:237–241. [PubMed] [Google Scholar]
- 25.Zhu JL, Song YX, Wang ZN, Gao P, Wang MX, Dong YL, Xing CZ, Xu HM. The clinical significance of mesenchyme forkhead 1 (FoxC2) in gastric carcinoma. Histopathology. 2013;62:1038–1048. doi: 10.1111/his.12132. [DOI] [PubMed] [Google Scholar]
- 26.Treggiari E, Ressel L, Polton GA, Benoit J, Desmas I, Blackwood L. Clinical outcome, PDGFRbeta and KIT expression in feline histiocytic disorders: a multicentre study. Vet Comp Oncol. 2017;15:65–77. doi: 10.1111/vco.12142. [DOI] [PubMed] [Google Scholar]
- 27.Wu Y, Yamada S, Izumi H, Li Z, Shimajiri S, Wang KY, Liu YP, Kohno K, Sasaguri Y. Strong YB-1 expression is associated with liver metastasis progression and predicts shorter disease-free survival in advanced gastric cancer. J Surg Oncol. 2012;105:724–730. doi: 10.1002/jso.23030. [DOI] [PubMed] [Google Scholar]
- 28.Kouso H, Yano T, Maruyama R, Shikada Y, Okamoto T, Haro A, Kakeji Y, Maehara Y. Differences in the expression of epithelial-mesenchymal transition related molecules between primary tumors and pulmonary metastatic tumors in colorectal cancer. Surg Today. 2013;43:73–80. doi: 10.1007/s00595-012-0344-0. [DOI] [PubMed] [Google Scholar]
- 29.Sun Y, Jia X, Gao Q, Liu X, Hou L. The ubiquitin ligase UBE4A inhibits prostate cancer progression by targeting interleukin-like EMT inducer (ILEI) IUBMB Life. 2017;69:16–21. doi: 10.1002/iub.1585. [DOI] [PubMed] [Google Scholar]
- 30.Marx V. Tracking metastasis and tricking cancer. Nature. 2013;494:133–136. doi: 10.1038/494131a. [DOI] [PubMed] [Google Scholar]
- 31.Diepenbruck M, Christofori G. Epithelial-mesenchymal transition (EMT) and metastasis: yes, no, maybe? Curr Opin Cell Biol. 2016;43:7–13. doi: 10.1016/j.ceb.2016.06.002. [DOI] [PubMed] [Google Scholar]
- 32.Kral M, Klimek C, Kutay B, Timelthaler G, Lendl T, Neuditschko B, Gerner C, Sibilia M, Csiszar A. Covalent dimerization of interleukin-like epithelial-to-mesenchymal transition (EMT) inducer (ILEI) facilitates EMT, invasion, and late aspects of metastasis. FEBS J. 2017;284:3484–3505. doi: 10.1111/febs.14207. [DOI] [PubMed] [Google Scholar]
- 33.Wong S, Fang CM, Chuah LH, Leong CO, Ngai SC. E-cadherin: Its dysregulation in carcinogenesis and clinical implications. Crit Rev Oncol Hematol. 2018;121:11–22. doi: 10.1016/j.critrevonc.2017.11.010. [DOI] [PubMed] [Google Scholar]
- 34.Zhou Y, Li G, Wu J, Zhang Z, Wu Z, Fan P, Hao T, Zhang X, Li M, Zhang F, Li Q, Lu B, Qiao L. Clinicopathological significance of E-cadherin, VEGF, and MMPs in gastric cancer. Tumour Biol. 2010;31:549–558. doi: 10.1007/s13277-010-0068-y. [DOI] [PubMed] [Google Scholar]
- 35.Dave JM, Bayless KJ. Vimentin as an integral regulator of cell adhesion and endothelial sprouting. Microcirculation. 2014;21:333–344. doi: 10.1111/micc.12111. [DOI] [PubMed] [Google Scholar]
- 36.Eriksson JE, Dechat T, Grin B, Helfand B, Mendez M, Pallari HM, Goldman RD. Introducing intermediate filaments: from discovery to disease. J Clin Invest. 2009;119:1763–1771. doi: 10.1172/JCI38339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen J, Wang T, Zhou YC, Gao F, Zhang ZH, Xu H, Wang SL, Shen LZ. Aquaporin 3 promotes epithelial-mesenchymal transition in gastric cancer. J Exp Clin Cancer Res. 2014;33:38. doi: 10.1186/1756-9966-33-38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kim MA, Lee HS, Lee HE, Kim JH, Yang HK, Kim WH. Prognostic importance of epithelial-mesenchymal transition-related protein expression in gastric carcinoma. Histopathology. 2009;54:442–451. doi: 10.1111/j.1365-2559.2009.03247.x. [DOI] [PubMed] [Google Scholar]

