Expression of transmembrane protein 41A is associated with metastasis via the modulation of E‑cadherin in radically resected gastric cancer

  • Authors:
    • Bin Lin
    • Yingming Xue
    • Chao Qi
    • Xiangjie Chen
    • Weizheng Mao
  • View Affiliations

  • Published online on: July 3, 2018     https://doi.org/10.3892/mmr.2018.9241
  • Pages: 2963-2972
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Gastric cancer (GC) is one of the most commonly occurring malignancies worldwide, and metastasis is one of the key processes affecting the prognosis of GC. TMEM41A, which belongs to a group of transmembrane proteins that participate in signaling pathways and tumor development, is a 264‑amino acid protein encoded by a gene mapped to human chromosome The exact role of TMEM41A in GC has not been determined to date. In the present study, the expression of TMEM41A in 147 cases of GC was analyzed with immunohistohemistry and the prognoses of these patients were analyzed. It was revealed that TMEM41A was highly expressed in GC tissues, and may be associated with the progression of GC and poor prognosis. The expression of TMEM41A was observed to be correlated with lymph node metastasis, distant metastasis and advanced tumor, node and metastasis stages. Knockdown of TMEM41A in vitro and in vivo decreased the GC cell migration ability by regulating epithelial‑to‑mesenchymal transition and cell autophagy, via the upregulation of E‑cadherin and downregulating N‑cadherin expression in GC cells by reverse transcription‑quantitative polymerase chain reaction (PCR), semi‑PCR and western blotting. Furthermore, TMEM41A upregulation was associated with the upregulation of p62 and altered the conversion of light chain (LC)3‑1 into LC3‑2 by western blotting. Knockdown of TMEM41A was also observed to affect tumor metastasis in nude mice. Therefore, TMEM41A may be considered as a novel therapeutic target for the treatment of GC‑associated metastasis.

Introduction

Recent advances in surgical techniques and developments in anticancer agents with improved therapeutic efficacies have been made; however, gastric cancer (GC) remains to be the most prevalent type of malignancy and is the second most common cause of cancer-associated mortality worldwide (1,2). Metastasis is one of the key molecular steps affecting the prognosis of GC, but as a complex process, further investigation is required (3). Identifying novel diagnostic or prognostic biomarkers of GC may be considered a major objective for the treatment of GC.

Epithelial-to-mesenchymal transition (EMT) is one of the key molecular steps in the process associated with distant metastasis (4), during which tumor cells exhibiting epithelial characteristics acquire mesenchymal characteristics by modulating cellular polarity and adhesion, and via the loss of cell-cell junctions (5). Throughout the process of EMT, cancer cells lose the expression of cellular adhesion proteins, such as E-cadherin, which has been considered as a hallmark of EMT; cells also acquire the expression of mesenchymal markers, including vimentin (5,6). EMT permits the invasion and migration in a variety of cancer types, including gastric and colon cancer (5).

The transmembrane (TMEM) protein superfamily is a group of transmembrane proteins that participate in particular signaling pathways and tumor development associated with established oncogenes, particularly TMEM16a (7,8), TMEM17a and TMEM17b (9). The associated proteins serve roles in colorectal, ovarian and bladder cancer. TMEM41A, a member of the TMEM41 family, is a 264-amino acid protein encoded by a gene mapped to human chromosome 3 (10,11). Chromosome 3 consists of ~214 million bases, encoding <1,100 genes (11), and possesses various human tumor-associated loci, as well as a chemokine receptor gene cluster (12). Particular areas of the chromosome 3 short arm tend to be lost in numerous types of tumor cells (13,14).

At present, the exact roles of TMEM41A in GC have not been determined. In the present study, the expression of TMEM41A and the prognoses of 147 patients with GC, and the functional role of TMEM41A in GC-associated metastasis were evaluated to investigate the potential therapeutic role of TMEM41A in the treatment of GC.

Materials and methods

Patient specimens and cell lines

Tumor tissues and matched adjacent normal gastric tissues were obtained from 147 patients (31–84-years-old) who underwent radical GC surgery between April 2007 and June 2010 at the Qingdao Municipal Hospital (Qingdao, China). Patients that received neoadjuvant chemotherapy or irregular therapy (standard therapy treatments, followed by 6 months of chemotherapy) were excluded from the present study. The adjacent normal tissues were laterally resected at a distance of ≥5 cm from the tumor margin. None of the subjects underwent neoadjuvant chemoradiotherapy. All the resected samples were immediately frozen in liquid nitrogen and maintained at −80°C until use.

Written informed consent was provided by all patients prior to enrollment into the present study, and the study protocols were approved by the Ethics Committee of Qingdao University (Qingdao, China). The clinical data collected from the subjects are summarized in Table I, including patient age and sex, tumor location, size and differentiation, lymph node involvement, distant metastasis, and tumor, node and metastasis (TNM) classification. Postoperative follow-up was performed every 3 months for a minimum of 5 years of 104 patients; 43 patients did not undergo follow-up due to exclusion criteria that arose in the present study, including patients withdrawal and failure to adhere to courses of standard treatment.

Table I.

Association between TMEM41A expression and clinicopathologic features.

Table I.

Association between TMEM41A expression and clinicopathologic features.

VariableLow TMEM41A expressionHigh TMEM41A expressionP-value
Total3965
Sex 0.545
  Male2135
  Female1830
Age (years) 0.491
  ≤501526
  >502439
Tumor size (cm) 0.566
  ≤52540
  >51425
Histology 0.488
  Adenocarcinoma3457
  Mucinous58
Local tumor invasion 0.584
  Early stage3661
  Late stage34
Lymph node metastasis 0.0026
  Negative2522
  Positive1443
Distant metastasis 0.0179
  Negative3443
  Positive522
TNM stage 0.00238
  I–II2725
  III–IV1240

[i] TMEM41A, transmembrane protein 41A; TNM, tumor, node and metastasis.

The human GC cell lines MKN-45, MGC-803, NCI-N87, SNU-5, KATO III, HGC-27, BGC-823, SGC-7901 and AGS were obtained from the American Type Culture Collection (Manassas, VA, USA), and were cultured in Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA), McCoy's 5A complete medium or RPMI-1640 (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany), supplemented with 10% heat-inactivated fetal bovine serum (FBS; Sigma-Aldrich; Merck KGaA). All cells were maintained in a humidified atmosphere with 5% CO2 at 37°C.

RNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis

TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) was employed to isolate total RNA from cell lines according to the manufacturer's protocol. cDNA was obtained from 2 µg RNA using the PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China). The mRNA expression levels of TMEM41A were determined by semi-qPCR with GoTaq polymerase (Promega Corporation, Madison, WI, USA) following the manufacturer's protocols. qPCR was followed by amplification with SYBR® Premix Ex Taq (Takara Biotechnology Co., Ltd.) using an ABI 7500HT system (ABI; Thermo Fisher Scientific, Inc.). The thermocycling program was set as follows: Initial denaturation for 5 min at 95°C; 40 cycles at 95°C for 15 sec and 60°C for 30 sec. The primers used were as follows: TMEM41A forward, 5′-CTGCTGTGCTGTGTGTTGAC-3′ and reverse, 5′-GTGTCCCAGGAGAAAAGAGCA-3′; and β-actin forward, 5′-AGTGTGACGTGGACATCCGCAAAG-3′ and reverse, 5′-ATCCACATCTGCTGGAAGGTGGAC-3′. The 2−ΔΔCq method was used to quantify the expression levels of RNA (15). For semi-qPCR, the DNA products were run on a 1.5% agarose gel containing ethidium bromide. The bands were quantified using a UV illuminating instrument (Bio-Rad Laboratories, Inc., Hercules, CA, MA, USA).

Cell transfection

The small interfering (si)-negative control (NC; cat. no. CON077) and si-TMEM41A (cat. no. 38422-2) oligonucleotides were synthesized by Shanghai GenePharma Co., Ltd., (Shanghai, China) and transfected at 20 nM. the oligonucleotides sequence of the siRNA-TMEM41A (5′-3′) is: Ccgg-(stem)gcGGAAGTAGCTTGCCTCACTCTCGAG (loop)-AGTGAGGCAAGCTACTTCCGC(stem)-TTTTTg. The control (pCMV6-NC) or TMEM41A overexpression (pCMV6-TMEM41A) vectors were constructed by OriGene Technologies, Inc. (Rockville, MD, USA). Oligonucleotide transfection was performed using 0.5–1.0 µl siRNA/vector with Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol; successful transfection was confirmed via western blotting as described below. Cells were collected at 48 h following transfection.

Immunohistochemistry

Immunohistochemistry with the gastric tumor and adjacent normal tissues was performed as previously described (16). A TMEM41A rabbit polyclonal antibody (1:50; sc-103285; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) was utilized as the primary antibody (overnight, 4°C); a secondary antibody (1:1,000; GK600705; Gene Tech Biotechnology Co., Ltd. (Shanghai, China) was applied for 1 h at room temperature. Subsequently, sections were counterstained with hematoxylin at room temperature for 40 min and analyzed under a light microscope (magnification, ×10). The sections were scored semi-quantitatively by two observers independently, in a blinded manner and analyzed under a light microscope. The scoring system was as follows: 0, 0% immunoreactive cells; 1, <5% immunoreactive cells; 2, 5–50% immunoreactive cells and 3, >50% immunoreactive cells. Scores of 0 or 1 were considered as low expression levels, and scores of 2 or 3 were considered as high expression levels.

Cell proliferation analysis

Cell proliferation assays of transfected cells were performed using a Cell Counting kit-8 (Dojindo Molecular Technologies, Inc., Kumamoto, Japan) according to the manufacturer's protocols; absorbance measurements at 450 nm were conducted to determine the cell proliferative ability in each well. The cells were seeded in a dish at a density of 1×105 cells/dish and cultured for 24 h at 37°C prior to transfection; detection was performed at 24, 48 and 72 h post-transfection.

Wound-healing assay

A wound-healing assay was employed to assess the migration capacity of transfected cells in a defective monolayer. In brief, cells were seeded in 6-well plates and a 100-µl pipette tip was used to conduct scratches. The medium was discarded, the cells were washed with PBS and fresh medium (RPMI-1640/DMEM and 10% FBS) was added, followed by the analysis of scratch closure under a light microscope at 18 h. Calculations were performed using the following formula: (Sx time 0-Sx time point/Sx time 0) ×100, where S indicated the distance in µm.

Cell migration assays

A cell migration assay was performed using Costar Transwell inserts (Thermo Fisher Scientific, Inc.) with a diameter of 6.5 mm and a pore size of 8.0 µm. Briefly, cells (1×104) resuspended in 300 µl serum-free medium were seeded into the upper Transwell chamber, and the bottom chamber was supplemented with 10% FBS. The cells were incubated for 24 h. Subsequently, the upper chamber was stained with Diff-Quik stain (Polysciences, Inc., Warrington, PA, USA) according to the manufacturer's protocol. Cell numbers were counted in 10 fields of view under an inverted microscope (magnification, ×40; DMI600B; Leica Microsystems GmbH, Wetzlar, Germany), and the proportion of migrating cells was determined after normalization to control cells.

Cell immunofluorescence for cytoskeleton stain and autophagy

Cells were seeded on coverslips when a single monolayer was achieved from culture, fixed in 4% paraformaldehyde for 10 min at room temperature, permeabilized in 0.1% Triton X-100 in PBS for 4 min. Subsequently, cells were blocked with 1% BSA for 20 min at room temperature, and then incubated at room temperature for 30 min with rhodamine-conjugated phalloidin (Sigma-Aldrich; Merck KGaA) at 1:200 in blocking solution (1% BSA in 0.1% Triton X-100 in PBS). The nuclei were counterstained with DAPI for 20 min at room temperature. Images were captured with a fluorescence microscope.

Autophagosome formation was detected by red fluorescent protein (RFP)-LC3 puncta incorporation into autophagic vacuoles. Sterile coverslips were seeded with 5×105 cells into 6-well plates. Following adherence and washing twice with PBS, cells were cultured in serum-free medium for 24 h. Subsequently, the cells were transiently transfected with RFP-LC3B-expressing GFP-vector (OriGene Technologies, Inc.) for 32 h with Lipofectamine® 2000, followed by washing with cold-ice PBS and fixation with 4% formaldehyde in PBS at room temperature for 20 min. The nuclei were counterstained with DAPI for 20 min at room temperature. Images were captured with a fluorescence microscope (magnification, ×100).

Tumor formation in nude mice

Stably siRNA-transfected BGC-823 cells selected via incubation with 1–3 µg/l puromycin and were then suspended in 0.2 ml PBS and injected into the caudal vein of 12 5-week-old female BALB/c nude mice (20–22 g) (5×106 cells/mouse). The mice housed under a 12 h light/dark cycle with free access to food and water at 20–24°C, 50% humidity; mice were monitored for 70 days and then sacrificed. The tumor size was recorded and tumor volume was calculated using the formula: (length × width2)/2. Tumors formed in vivo were collected, sectioned and stained with hematoxylin and eosin (H&E) at room temperature for 40 min and analyzed under a light microscope.

Western blot analysis

Briefly, for western blotting, after washing with cold-ice PBS, total protein of transfected cells was isolated by incubation in radioimmunoprecipitation assay buffer (Cell Signaling Technology, Inc., Danvers, MA, USA) containing complete protease inhibitor cocktail (Roche Diagnostics, Basel, Switzerland) for 1 h on ice. Following centrifugation (15,000 × g at 4°C for 20 min), the supernatants were collected. Protein concentration was determined using a bicinchoninic acid protein assay kit (Thermo Fisher Scientific, Inc.). Samples (2–10 mg) were subjected to 8–12% SDS-PAGE according to protein mass loaded and transferred to polyvinylidene difluoride membranes activated with 100% methanol. After blocking in 5% fat-free milk for 1 h, the membranes were incubated with a primary antibody against TMEM41A (dilution, 1:200; Santa Cruz Biotechnology, Inc.), E-cadherin (ab15148), N-cadherin (ab18203), p62 (ab91526), LC3-I/II (ab51520), and β-actin (ab8227) (dilution, 1:1,000; Abcam, Cambridge, MA, USA) overnight on ice and incubated with the indicated secondary antibody (in-house; 1:1,000) for 2 h at room temperature. The membrane was then washed with tris-buffered saline with 1:1,000 Tween-20 in v/v. prior to treatment with an enhanced chemiluminescent reagent and placed in a dark room to allow the reaction to run to completion. β-actin was used as a positive control.

Bioinformatic analysis

Investigation with The Cancer Genome Atlas (TCGA; http://cancergenome.nih.gov/) dataset was conducted to analyze the expression levels of TMEM41A in normal gastric tissues; Protein Atlas (www.proteinatlas.org) also revealed that the mRNA and protein expression levels of TMEM41A may be inconsistent.

Statistical analysis

Data were analyzed using SPSS version 18 (SPSS Inc., Chicago, IL, USA). The results are presented as the mean ± standard error of the mean. Kaplan-Meier analysis was conducted, followed by a log-rank test. All experiments were performed in triplicate. The differences between groups were analyzed using one-way analysis of variance followed by the Least Significant Difference t-test. Categorical data was analyzed with a χ2 test. In all cases, P<0.05 was considered to indicate a statistically significant difference and P<0.01 was considered to indicate a highly statistically significant difference.

Results

Expression of TMEM41A in human GC cell lines

The expression levels of TMEM41A in the nine human GC cell lines were determined. As presented in Fig. 1A and B (F=5.381>F0.05; P<0.05), the expression levels of TMEM41A were the highest in BGC-823 cells and the lowest in MKN-45 cells (Fig. 1B). Therefore, BGC-823 cells were transfected with siRNA against TMEM41A, and MKN-45 cells were transfected with a TMEM41A-overexpression plasmid to investigate the effects of siRNA and plasmid transfection on the expression levels of TMEM41A. Western blot analysis demonstrated that si-TMEM41A markedly decreased the expression levels of TMEM41A in BGC-823 cells, and that pCMV6-TMEM41A notably increased the expression levels of TMEM41A in MKN-45 cells (Fig. 1C and D).

TMEM41A expression in human GC cells and its association with clinicopathological characteristics

To investigate the potential role of TMEM41A in GC-associated metastasis, RT-qPCR was used to assess TMEM41A expression in 147 cases of GC and paired adjacent normal tissues, which revealed the overexpression of TMEM41A mRNA in the GC tissue exhibited by 123/147 (83.67%) of the pairs, with a mean fold change of 2.22 for the cancer tissues; expression levels were significantly higher within the tumor tissues compared with in that of the normal tissues (Fig. 2A). In addition, the relative mean expression levels of TMEM41A were significantly higher in subjects with lymph node involvement compared with those without (P<0.01; Fig. 2B). Consistently, TMEM41A expression was significantly higher in subjects with distant metastasis compared to those without (P<0.01; Fig. 2C). The associations of TMEM41A expression with the clinicopathological characteristics of patients with GC are summarized in Table I. TMEM41A expression was observed to be significantly associated with advanced TNM stages (III and IV), lymph node involvement and distant metastasis (P<0.01; Fig. 2B-D); however, TMEM41A expression was not significantly associated with other clinicopathological characteristics, including patient age, sex, tumor location, differentiation or size (all P>0.05).

TMEM41A overexpression is correlated with lymph node metastasis, distant metastasis and advanced TNM stage in human GC tissues

Subsequently, the present study determined the expression levels of TMEM41A protein in the same pathological specimens. TMEM41A protein was suggested to be localized on the cell plasma membranes of GC samples (Fig. 2E). As presented in Table I, of the 147 patients with GC, 104 (selected based on follow-up, staining, and other factors, including variations in therapy) were divided into two subgroups based on the expression levels of TMEM41A. The high-expression group included 65 patients with a higher expression levels of TMEM41A in the carcinoma tissue compared with in adjacent normal gastric tissues, whereas in the low-expression group (n=39), the mean TMEM41A staining intensity obtained during semi-quantitative analysis of carcinoma tissue samples decreased compared with the adjacent normal gastric tissues (data not shown).

Survival analyses were performed on patients with GC. Patients in the TMEM41A high-expression group exhibited poorer overall and disease-free survival compared with the low-expression group (Fig. 2F and G).

There were no significant differences in the 1-year overall survival rate between the two groups; however, the 3- and 5-year overall survival rates were poorer in the TMEM41A high-expression group. The 5-year overall survival rates for the high- and low-expression groups were 75.6 and 45.8%, respectively (P=0.012; Table II). This finding suggested that high expression levels of TMEM41A in GC may be considered as a predictor of poor prognoses of GC.

Table II.

Association between TMEM41A expression and survival rate.

Table II.

Association between TMEM41A expression and survival rate.

TMEM41A density

Survival measurement (%)Low-TMEM41AHigh-TMEM41AP-value
1-year overall survival95.6±4.089.5±4.4
3-year overall survival90.4±8.875.5±8.7
5-year overall survival75.6±6.045.8±6.80.012a

a P=0.012 across all survival measurements. TMEM41A, transmembrane protein 41A.

TMEM41A does not promote the proliferation of GC cells

The present study performed Cell Counting kit-8 assays to investigate the effects of TMEM41A on GC cell proliferation. The Cell Counting kit-8 assays demonstrated that the stable knockdown of TMEM41A mediated by siRNA was not associated with variations in BGC-823 cell proliferation (P>0.0; Fig. 3A). Similar results were observed in TMEM41A-overexpressing MKN-45 cells (P>0.05; Fig. 3B).

Association between TMEM41A expression and the migration of human GC cells

In order to investigate the role of TMEM41A in cell migration, wound-healing and Transwell assays were performed. The results indicated that knockdown of TMEM41A significantly decreased the migration ability of BGC-823 cells (Fig. 4A; P<0.01). Conversely, a significant increase in the rate of migration was observed when TMEM41A was overexpressed in MKN-45 cells compared with in the control (Fig. 4B; P<0.01). Transwell assays indicated that the migration ability of BGC-823 cells was significantly decreased following the silencing of endogenous TMEM41A compared with in the control (Fig. 4C; P<0.01). Conversely, the overexpression of TMEM41A significantly increased the migration ability of MKN-45 cells compared with in the control (Fig. 4D; P<0.01).

Different expression of TMEM41A leads to cytoskeletal rearrangement and inhibition of cell adhesion and spreading

To determine the role of TMEM41A in the regulation of cytoskeletal dynamics, rhodamine-conjugated phalloidin staining was applied (Fig. 5A). Inside the cells, actin filaments were distributed in a disorderly manner, and no evident stress fiber formation was observed in TMEM41A knockdown cells, indicating that the knockdown of TMEM41A may inhibit stress fiber formation. On the cell membranes, membrane ruffling and the formation of pseudopodia, reflective of cell migration, were markedly observed in control and siTMEM41A transfected cells; in the overexpression group, opposing findings were observed. These results indicate that high expression levels of TMEM41A may promote metastatic properties of GC cells, consistent with the wound-healing assay as aforementioned.

TMEM41A may induce autophagy within GC cells

To further verify the hypothesis of the present study, RFP-LC3B plasmids were transiently transfected into the cells. Punctate fluorescence demonstrated the induction of autophagy. As presented in Fig. 5B, no notable punctate fluorescence was observed in si-TMEM41A-treated BGC-823 cells, but not in control vector-transfected or TMEM41A-overexpressing cells. These results suggested that TMEM41A may be associated with autophagy.

Effect of TMEM41A downregulation on E-cadherin expression and autophagy in GC cells

EMT is crucial for the invasion and migration of tumor cells, and the development of metastasis. To investigate the mechanisms underlying the effects of TMEM41A on cell migration, the effects of TMEM41A knockdown or overexpression on EMT were examined by analyzing the expression levels of EMT-associated factors (Fig. 6A). The knockdown or overexpression of TMEM41A affected the expression levels of E-cadherin and N-cadherin, and the autophagy-associated factors p62 and LC3-1/2, indicating that the knockdown or overexpression of TMEM41A may affect EMT and the autophagy process (Fig. 6A).

Knockdown of TMEM41A affects tumor metastasis in nude mice

To further determine the effect of TMEM41A on GC tumor metastasis in vivo, the present study employed an siRNA-mediated stable knockdown cell model. H&E staining indicated that tumors were formed in vivo at a lower rate in the si-TMEM41A group compared with in the si-NC group (Fig. 6B). Both groups exhibited successful tumor formation in the lung, and tumor size of the si-TMEM41A group was significantly smaller compared with in the si-NC group (Fig. 6C; P<0.01). These results indicated alterations in the expression of TMEM41A and EMT-associated markers may be associated with metastatic behavior in vivo.

Discussion

To retain normal organ size and specific organ morphology, cell numbers in tissues of high proliferative potential are strictly controlled by the balance between cell proliferation and apoptosis during organ development (17). This suggests that the dysregulation of genes associated with these processes may be detected in human malignancy.

Metastasis is one of the key processes affecting the prognosis of GC, but it is a complex process which requires further investigation (3). A total of ~70% of patients with GC develop metastasis (18). Advances in treatments for GC have been made; however, patients with advanced or metastatic GC exhibit poor prognosis (19). Therefore, the identification of novel diagnostic or prognostic biomarkers for GC is one of the major goals in this field.

The TMEM protein superfamily comprises >310 different members, including membrane proteins, which are extensively expressed on cell and lysosomal membranes, and the endoplasmic reticulum, are strongly associated with membrane integrity, transport and signaling pathways (20). An increasing number of studies have investigated the TMEM superfamily and tumor-associated genes, particularly the oncogenes, TMEM16a (7,8), TMEM17a and TMEM17b (9) and the tumor suppressor gene TMEM100 (21,22). Certain TMEM genes have been reported to be involved in the development and metastasis of GC, such as TMEM16a (20).

TMEM41A has been considered to be a potential cancer-associated gene, as it is located on the long arm of chromosome 3 (10,11). Various human cancer-associated gene loci are present on chromosome 3. One of the most common abnormalities in human cancers is the loss of the short arm of chromosome 3 (23). Therefore, the tumor suppressor gene(s) located on 3p may serve a key role in the development of numerous types of cancer (13,14). In addition, the amplification of chromosome 3q is associated with cancer; the amplification of the distal portion of chromosome 3q has been reported to be an important signature associated with lung squamous cell carcinoma (24). The role of TMEM41A in cancer has not been previously investigated. The present study revealed that the TMEM41A expression levels were high in patients with GC. Increased TMEM41A expression levels were positively associated with distant metastasis, advanced TNM stage and lymph node involvement, as observed in the present study. Patients with low TMEM41A expression levels exhibited improved overall survival compared with those with high TMEM41A expression levels. To the best of the authors' knowledge, the present study is the first to investigate the prognostic potential of TMEM41A in patients with GC.

An increasing number of studies (2528) have investigated the mechanisms underlying the aberrant expression of TMEM41A; however, the regulation of TMEM41A protein expression remains unclear. It has previously been demonstrated that its expression may be regulated by numerous mechanisms, including chromosomal abnormalities (25), polymorphisms (26), epigenetic mechanisms (27) and histone modifications (28). Further investigation in identifying the underlying mechanisms of irregular TMEM41A expression in GC is required. In particular, whether the upregulation of TMEM41A may be due to chromosomal abnormalities requires further investigation.

EMT is a series of events including the alteration of cell-cell and cell-extracellular matrix interactions, and cancer cell transformation from an epithelial to a mesenchymal phenotype (29). These alterations are often accompanied with increased cell motility. Therefore, EMT is a hallmark of cancer, and is associated with a poor clinical outcome and metastasis (30,31). During tumor progression and metastasis, EMT may be induced by autonomous oncogenic activation or inactivation of signaling molecules, in the presence or absence of external stimuli (32). In this process, markers of epithelial differentiation are downregulated, including E-cadherin; however, N-cadherin, Slug, vimentin, Twist-related protein-1 and Snail are upregulated (33). EMT is a key process in the metastasis of GC (34). The findings of present study suggested that TMEM41A may facilitate the metastasis of GC cells via EMT. Furthermore, TMEM41A may participate in the regulation of E-cadherin and N-cadherin expression; however, further experiments may be conducted in the future to elucidate the association between TMEM41A and the EMT signaling pathway.

It was previously suggested that autophagy may contribute towards the regulation of the migration and invasive abilities of cancer cells, and the activation of autophagy has been associated with the inhibition of EMT (3537). It has been reported that autophagy accelerates the invasion of glioblastoma stem cells by regulating DNA damage-regulated autophagy modulator 1 and p62 (38). The present study demonstrated similar results, as the upregulation of E-cadherin was accompanied with that of p62 and the altered conversion of LC3-I to LC3-II. In addition, immunofluorescence analysis in the present study was not conducted with a control and the pCMV-TMEM41A group. Therefore, further analysis in the future is required to verify the findings of the present study. However, whether TMEM41A regulates EMT via autophagy requires further investigation.

Additionally, the data presented in The Cancer Genome Atlas dataset, revealed that TMEM41A was highly expressed in normal gastric tissues; however, immunohistochemistry analysis did not demonstrate such expression in tumor samples in the present study. Furthermore, analysis with Protein Atlas (www.proteinatlas.org) also revealed that the mRNA and protein expression levels of TMEM41A may be inconsistent.

As the expression levels of TMEM41A at the gene and protein levels have not been fully investigated, contradictory findings may be reported. The present study reported that the mRNA expression levels of TMEM41A were similar at the protein level; however, the protein expression levels revealed by immunohistochemistry were contradictory. Therefore, the present study suggested that the dilution of antibody (1:50 vs. 1:400), post-translational modulation, or other unknown components may be associated with these differing results (data not shown). Conversely, the aforementioned databases have demonstrated a high correlation between TMEM41A mRNA expression and poor prognoses in endometrial, liver and pancreatic cancers, but not gastric cancer. Therefore, further investigation is required to understand these differing expression profiles.

The present study demonstrated a reduction in TMEM41A expression levels in a pair of GC cell lines, and revealed that high TMEM41A expression levels may promote GC-associated metastasis, which may be mediated by the downregulation of E-cadherin expression. The present study aimed to highlight the potential of TMEM41A as a novel target against GC-associated metastasis and suggest that TMEM41A may be considered to be an important oncogene in GC. Further studies examining the mechanisms underlying the regulation of TMEM41A expression are required to investigate the possible association between TMEM41A with E-cadherin expression in GC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

BL and YX made substantial contributions to the design of the present study and wrote the manuscript. CQ made substantial contributions to the design of the present study and performed the animal experiment, XC conducted software analysis and revised the manuscript and WM directed and managed the design of the present study. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Qingdao University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

GC

gastric cancer

TMEM41A

transmembrane protein 41A

EMT

epithelial-to-mesenchymal transition

PBS

phosphate-buffered saline

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Yuan DD, Zhu ZX, Zhang X and Liu J: Targeted therapy for gastric cancer: Current status and future directions (Review). Oncol Rep. 35:1245–1254. 2016. View Article : Google Scholar : PubMed/NCBI

3 

Jin X, Zhu Z and Shi Y: Metastasis mechanism and gene/protein expression in gastric cancer with distant organs metastasis. Bull Cancer. 101:E1–E12. 2014.PubMed/NCBI

4 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Bates RC and Mercurio AM: The epithelial-mesenchymal transition (EMT) and colorectal cancer progression. Cancer Biol Ther. 4:365–370. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Ferrera L, Caputo A and Galietta LJ: TMEM16A protein: A new identity for Ca(2+)-dependent Cl- channels. Physiology (Bethesda). 25:357–363. 2010.PubMed/NCBI

7 

Zeng X, Huang P, Chen M, Liu S, Wu N, Wang F and Zhang J: TMEM16A regulates portal vein smooth muscle cell proliferation in portal hypertension. Exp Ther Med. 15:1062–1068. 2018.PubMed/NCBI

8 

Zhang X, Zhang Y, Miao Y, Zhou H, Jiang G and Wang E: TMEM17 depresses invasion and metastasis in lung cancer cells via ERK signaling pathway. Oncotarget. 8:70685–70694. 2017.PubMed/NCBI

9 

Huber MA, Kraut N and Beug H: Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol. 17:548–558. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Strausberg RL, Feingold EA, Grouse LH, Derge JG, Klausner RD, Collins FS, Wagner L, Shenmen CM, Schuler GD, Altschul SF, et al: Generation and initial analysis of more than 15,000 full-length human and mouse cDNA sequences. Proc Natl Acad Sci USA. 99:16899–16903. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Ota T, Suzuki Y, Nishikawa T, Otsuki T, Sugiyama T, Irie R, Wakamatsu A, Hayashi K, Sato H, Nagai K, et al: Complete sequencing and characterization of 21,243 full-length human cDNAs. Nat Genet. 36:40–45. 2004. View Article : Google Scholar : PubMed/NCBI

12 

Maestro R, Gasparotto D, Vukosavljevic T, Barzan L, Sulfaro S and Boiocchi M: Three discrete regions of deletion at 3p in head and neck cancers. Cancer Res. 53:5775–5779. 1993.PubMed/NCBI

13 

Ricciardi R, Burks E, Schoetz DJ, Verma Y, Kershnar E, Kilpatrick MW, Tsipouras P and Walat RJ: Is there a gain in chromosome 3q in the pathway to anal cancer? Dis Colon Rectum. 57:1183–1187. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Wu CL, Sloan P, Read AP, Harris R and Thakker N: Deletion mapping on the short arm of chromosome 3 in squamous cell carcinoma of the oral cavity. Cancer Res. 54:6484–6488. 1994.PubMed/NCBI

15 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Feng X, Zhu K, Liu J, Chen J, Tang J, Liang Y, Jin R, Liang X and Cai X: The evaluative value of Sema3C and MFN2 co-expression detected by immunohistochemistry for prognosis in hepatocellular carcinoma patients after hepatectomy. Onco Targets Ther. 9:3213–3221. 2016.PubMed/NCBI

17 

Kumar R, Vadlamudi RK and Adam L: Apoptosis in mammary gland and cancer. Endocr Relat Cancer. 7:257–269. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Qinghai Z, Yanying W, Yunfang C, Xukui Z and Xiaoqiao Z: Effect of interleukin-17A and interleukin-17F gene polymorphisms on the risk of gastric cancer in a Chinese population. Gene. 537:328–332. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Park S, Ha S, Kwon HW, Kim WH, Kim TY, Oh DY, Cheon GJ and Bang YJ: Prospective evaluation of changes in tumor size and tumor metabolism in patients with advanced gastric cancer undergoing chemotherapy: Association and clinical implication. J Nucl Med. 58:899–904. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Zeng X, Huang P, Chen M, Liu S, Wu N, Wang F and Zhang J: TMEM16A regulates portal vein smooth muscle cell proliferation in portal hypertension. Exp Ther Med. 15:1062–1068. 2018.PubMed/NCBI

21 

Han Z, Wang T, Han S, Chen Y, Chen T, Jia Q, Li B, Li B, Wang J, Chen G, et al: Low-expression of TMEM100 is associated with poor prognosis in non-small-cell lung cancer. Am J Transl Res. 9:2567–2578. 2017.PubMed/NCBI

22 

Ou D, Yang H, Hua D, Xiao S and Yang L: Novel roles of TMEM100: Inhibition metastasis and proliferation of hepatocellular carcinoma. Oncotarget. 6:17379–17390. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Al-Hussain T, Ali A and Akhtar M: Wilms tumor: An update. Adv Anat Pathol. 21:166–173. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Haga Y, Hiroshima K, Iyoda A, Kohno H, Shibuya K, Iizasa T, Fujisawa T and Ohwada H: Frequency of loss of heterozygosity at 3 p, 9 p, 13 q and 17 p is related to proliferative activity in smokers with stage I non-small cell lung cancer. Thorac Cardiovasc Surg. 53:114–117. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Calin GA and Croce CM: MicroRNAs and chromosomal abnormalities in cancer cells. Oncogene. 25:6202–6210. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Calvo SE, Pagliarini DJ and Mootha VK: Upstream open reading frames cause widespread reduction of protein expression and are polymorphic among humans. Proc Natl Acad Sci USA. 106:7507–7512. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Jaenisch R and Bird A: Epigenetic regulation of gene expression: how the genome integrates intrinsic and environmental signals. Nat Genet. 33 Suppl:S245–S254. 2003. View Article : Google Scholar

28 

Fahrner JA, Eguchi S, Herman JG and Baylin SB: Dependence of histone modifications and gene expression on DNA hypermethylation in cancer. Cancer Res. 62:7213–7282. 2002.PubMed/NCBI

29 

Prieto-García E, Díaz-García CV, García-Ruiz I and Agulló-Ortuño MT: Epithelial-to-mesenchymal transition in tumor progression. Med Oncol. 34:1222017. View Article : Google Scholar : PubMed/NCBI

30 

Lefevre M, Rousseau A, Rayon T, Dalstein V, Clavel C, Beby-Defaux A, Pretet JL, Soussan P, Polette M, Saint Lacau Guily J and Birembaut P: Papillophar Study Group: Epithelial to mesenchymal transition and HPV infection in squamous cell oropharyngeal carcinomas: The papillophar study. Br J Cancer. 116:362–369. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Wang Y and Zhou BP: Epithelial-mesenchymal transition-A hallmark of breast cancer metastasis. Cancer Hallm. 1:38–49. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Scheel C, Eaton EN, Li SH, Chaffer CL, Reinhardt F, Kah KJ, Bell G, Guo W, Rubin J, Richardson AL and Weinberg RA: Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell. 145:926–940. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Zhao Y, Guo Q, Chen J, Hu J, Wang S and Sun Y: Role of long non-coding RNA HULC in cell proliferation, apoptosis and tumor metastasis of gastric cancer: A clinical and in vitro investigation. Oncol Rep. 31:358–364. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Fu M, Huang Z, Zang X, Pan L, Liang W, Chen J, Qian H, Xu W, Jiang P and Zhang X: Long noncoding RNA LINC00978 promotes cancer growth and acts as a diagnostic biomarker in gastric cancer. Cell Prolif. 2018. View Article : Google Scholar

35 

Gugnoni M, Sancisi V, Manzotti G, Gandolfi G and Ciarrocchi A: Autophagy and epithelial-mesenchymal transition: An intricate interplay in cancer. Cell Death Dis. 7:e25202016. View Article : Google Scholar : PubMed/NCBI

36 

Li J, Yang B, Zhou Q, Wu Y, Shang D, Guo Y, Song Z, Zheng Q and Xiong J: Autophagy promotes hepatocellular carcinoma cell invasion through activation of epithelial-mesenchymal transition. Carcinogenesis. 34:1343–1351. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Lv Q, Wang W, Xue J, Hua F, Mu R, Lin H, Yan J, Lv X, Chen X and Hu ZW: DEDD interacts with PI3KC3 to activate autophagy and attenuate epithelial-mesenchymal transition in human breast cancer. Cancer Res. 72:3238–3250. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Catalano M, D'Alessandro G, Lepore F, Corazzari M, Caldarola S, Valacca C, Faienza F, Esposito V, Limatola C, Cecconi F and Di Bartolomeo S: Autophagy induction impairs migration and invasion by reversing EMT in glioblastoma cells. Mol Oncol. 9:1612–1625. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 18 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin B, Xue Y, Qi C, Chen X and Mao W: Expression of transmembrane protein 41A is associated with metastasis via the modulation of E‑cadherin in radically resected gastric cancer. Mol Med Rep 18: 2963-2972, 2018
APA
Lin, B., Xue, Y., Qi, C., Chen, X., & Mao, W. (2018). Expression of transmembrane protein 41A is associated with metastasis via the modulation of E‑cadherin in radically resected gastric cancer. Molecular Medicine Reports, 18, 2963-2972. https://doi.org/10.3892/mmr.2018.9241
MLA
Lin, B., Xue, Y., Qi, C., Chen, X., Mao, W."Expression of transmembrane protein 41A is associated with metastasis via the modulation of E‑cadherin in radically resected gastric cancer". Molecular Medicine Reports 18.3 (2018): 2963-2972.
Chicago
Lin, B., Xue, Y., Qi, C., Chen, X., Mao, W."Expression of transmembrane protein 41A is associated with metastasis via the modulation of E‑cadherin in radically resected gastric cancer". Molecular Medicine Reports 18, no. 3 (2018): 2963-2972. https://doi.org/10.3892/mmr.2018.9241