Open Access

lncRNA LIFR‑AS1 inhibits gastric carcinoma cell proliferation, migration and invasion by sponging miR‑4698

  • Authors:
    • Jiangqiao Zhao
    • Xiaoning Li
    • Liping Fu
    • Na Zhang
    • Jiaping Yang
    • Jianhui Cai
  • View Affiliations

  • Published online on: December 20, 2020     https://doi.org/10.3892/mmr.2020.11792
  • Article Number: 153
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The vital functions of long non‑coding (lnc)RNAs have been verified in gastric carcinoma (GC). However, as a novel cancer‑related lncRNA, the influence of leukemia inhibitory factor receptor antisense RNA 1 (LIFR‑AS1) in GC cell biological behaviors remains unreported. The present study explored the biological effects of lncRNA LIFR‑AS1 on GC progression. Reverse transcription‑quantitative PCR was performed to examine lncRNA LIFR‑AS1 expression in GC tissues and cells. Cell Counting Kit‑8, 5‑ethynyl‑2'‑deoxyuridine incorporation, cell wound healing and Transwell invasion assays were used to assess the functions of lncRNA LIFR‑AS1 in GC cell proliferation, migration and invasion. Additionally, associations among lncRNA LIFR‑AS1, microRNA (miR)‑4698 and microtubule‑associated tumor suppressor 1 (MTUS1) were investigated via bioinformatics software and a luciferase reporter system. In addition, western blotting was used to examine the expression of MEK and ERK. Decreased lncRNA LIFR‑AS1 expression was observed in GC tissues and cells. Upregulated lncRNA LIFR‑AS1 inhibited GC cell proliferation, migration and invasion. Upregulated miR‑4698 and downregulated MTUS1 were identified in GC tissues and cells. The inhibitory interaction between lncRNA LIFR‑AS1 and miR‑4698 was confirmed. Additionally, MTUS1 was predicted as a target gene of miR‑4698 positively regulated by lncRNA LIFR‑AS1. The MEK/ERK pathway was inhibited by lncRNA LIFR‑AS1 via regulating MTUS1. These findings revealed the inhibitory functions of lncRNA LIFR‑AS1 in GC cell proliferation, migration and invasion. The process was mediated via miR‑4698, MTUS1 and the MEK/ERK pathway.

Introduction

Gastric carcinoma (GC) is a malignant tumor originating from the gastric mucosa (1). The incidence rate of GC is second only to lung carcinoma and is significantly higher in men than in women (2). Due to the GC tumor invasion and metastasis, GC affects the liver, kidney and respiratory functions and endangers life (3). The majority of patients with early GC generally have no distinct symptoms. However, with the growth of tumor, obvious symptoms appear as the gastric function is affected (4). In clinical practice, surgical treatment, chemotherapy, traditional Chinese medicine treatment and other comprehensive therapies are used to ameliorate the symptoms of patients with GC and prolong the survival (5,6). Nevertheless, the prognosis of patients with GC in advanced-stage is still unsatisfactory. Thus, it is important to investigated a tumor marker and develop a new curative method for GC.

Long non-coding (lnc)RNAs are transcripts >200 nucleotides in length (7). Increasing evidence has uncovered significant functions of lncRNAs in various types of cancer (8,9). Notably, a number of GC-associated lncRNAs such as H19 (10), HOXA11-AS (11) and AK058003 (12) have been reported to modulate the proliferative, migratory and invasive capacities of GC cells. The cancer-related lncRNA LIFR-AS1 has been studied in breast (13,14) and colorectal cancer (15). These studies have shown that lncRNA LIFR-AS1 is linked to the high survival rate of breast cancer and can inhibit breast cancer cell proliferation and migration; in addition, it can regulate the resistance of colorectal cancer to photodynamic therapy (14,15). However, the functions of lncRNA LIFR-AS1 in GC remain to be elucidated.

Micro (mi)RNAs are encoded by endogenous genes with a length of ~22 nucleotides and possess multiple vital regulating effects in a variety of cells (16,17). Accumulating evidence has established that lncRNAs work as miRNAs sponge to serve functions in the progression of various types of cancer, including GC (18,19). miR-4698 is situated on chromosome 12, which is linked to metastatic melanoma (20). Notably, Kalhori et al (21) revealed that miR-4698 can stop glioblastoma cell proliferation by controlling the PI3K/AKT pathway (21). Whether miR-4698 takes part in mediating the functions of lncRNA LIFR-AS1 in GC cells is the focus of the present study.

Microtubule-associated tumor suppressor 1 (MTUS1) is a tumor suppressor gene at chromosome 8p21.3–22, encoding a mitochondrial protein and controlling cellular proliferation (22). Previous studies have demonstrated that the expression status of MTUS1 is altered in several types of tumors, such as fibroadenoma and breast cancer (23), colorectal carcinoma (24) and bladder cancer (25). Thus far, no study has investigated whether MTUS1 participates in mediating the functions of lncRNA LIFR-AS1 in GC cells.

According to the above research, the present study preliminarily investigated the functions of lncRNA LIFR-AS1 in GC cell proliferation, migration and invasion and determined the molecular mechanisms involved. The results from the present study might offer a new research orientation for GC treatment from the aspect of lncRNA, miRNA and mRNA axis.

Materials and methods

Collection of samples from patients with GC

The GC tissues and paired adjacent tissues needed for the experiments were collected from 41 patients with GC (Table I). No treatment was performed on these patients prior to surgery. Patients were excluded if they had other clinical disorders. The obtained samples were frozen in liquid nitrogen and stored at −80°C. All patients personally signed an informed consent. The protocol of the present study was approved by the Ethics Committee of of Cangzhou People's Hospital (approval number AF/SC-08/02.0).

Table I.

Correlation between LRFR-AS1 expression and clinical characteristics.

Table I.

Correlation between LRFR-AS1 expression and clinical characteristics.

VariableN (%)Relative LRFR-AS1 expression (2−ΔΔCq method)P-value
Age (year)59.2±10.5/
Sex 0.2584
  Male27 (65.8%)0.466±0.185
  Female14 (34.2%)0.541±0.223
Histologic type 0.2711
  Adenocarcinoma35 (85.4%)0.477±0.187
  Others6 (14.6%)0.575±0.264
Differentiation degree 0.0383a
  Well/Moderately18 (43.9%)0.564±0.209
  Poorly23 (56.1%)0.435±0.176
Depth of invasion 0.1190
  T1 and T216 (39.0%)0.552±0.215
  T3 and T425 (61.0%)0.452±0.182
Nodal status 0.0045b
  pN018 (43.9%)0.588±0.184
  pN1-323 (56.1%)0.416±0.180
TNM stage 0.0011b
  I, II26 (63.4%)0.565±0.187
  III, IV15 (36.6%)0.364±0.153

a P<0.05

b P<0.01. TNM, TNM Classification of Malignant Tumors.

Cell culture

GC cell lines (MKN45 and AGS) and the gastric mucosal epithelial cell line (GES-1) were obtained from the Cell Bank of the Chinese Academy of Sciences. RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc.) was used to culture these cells. The culture conditions were 5% CO2 and 95% air at 37°C.

Cell transfection

The vectors of pcDNA-LIFR-AST and paired pcDNA negative control (NC), as well as miR-4698 mimics, miR-4698 inhibitors and their corresponding controls (mimics NC and inhibitors NC) used in this study were synthesized by GenePharma. The small interfering (si)RNA targeting microtubule-associated tumor suppressor 1 (MTUS1) was constructed in U6/GFP/Neo vectors (GenePharma) to silence MTUS1 expression. A scrambled siRNA was included as NC. The MKN45 and AGS cells were pre-incubated on 6-well plates until they reached ~60% confluence and the transfection was conducted with Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.). A total of 50 nM pcDNA-LIFR-AST or pcDNA NC, 20 nM miR-4698 mimics or mimics NC, 20 nM miR-4698 inhibitors or inhibitors NC and 45 nM si-MTUS1 or si-NC were used. Cells were harvested 48 h post-transfection for further studies.

Detection of cell proliferation

When cell transfection was concluded, Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2′- deoxyuridine (EdU) incorporation assays (Abcam) were used to assess cell proliferation. Transfected MKN45 and AGS cells (5×103 cells/well) were cultivated at 37°C for 1, 2, 3 and 4 days. CCK-8 solution (10 µl) was added into each well for another 2 h incubation. A microplate reader (BioTek Instruments, Inc.) was used for examining the absorbance at 450 nm wavelength. For the EdU assay, transfected cells were grown in 24-well plates at 37°C. The cells were then stained with 40 µM EdU solution at 37°C for 2 h and then 4% formaldehyde and 10X Triton X-100 buffer (Sigma-Aldrich; Merck KGaA) were added at room temperature for 15 min incubation. The reaction mix was added to fluorescently labelled EdU and was incubated at 37°C for 30 min. Finally, the EdU positive cells were analyzed by a fluorescence microscope (Olympus Corporation, magnification, ×100). Images were captured in five random fields.

Wound healing assay

The transfected MKN45 and AGS cells were cultured in 6-well plates to produce a cell monolayer (100% confluence). The cell wounds were created using a 100 µl pipette tip. After washing three times with sterile PBS, cells were further cultured at 37°C for 24 h and then the width of scratch was observed with an inverted microscope (Olympus Corporation, magnification, ×100). Additionally, images were captured at 0 and 24 h after scratching. ImageJ software (v1.8.0.112, National Institutes of Health) was used to analyze the images. Images were captured in five random fields.

Cell invasion

The transfected MKN45 and AGS cells in 200 µl serum-free RPMI-1640 medium were added to the upper Transwell chambers (Corning Inc.; 24-well insert, pore size 8 mm). The Transwell membrane was pre-coated with Matrigel (BD Biosciences). Meanwhile, the bottom Transwell chambers were filled with 600 µl RPMI-1640 medium supplemented with 10% FBS. After 24 h incubation at 37°C, the cells were fixed and subsequently stained with 0.5% crystal violet solution (Sigma-Aldrich; Merck KGaA) for 15 min at room temperature. The invasive cells were counted under a light microscope (Olympus Corporation, magnification, ×100). ImageJ software (v1.8.0.112, National Institutes of Health) was used to analyze the images. Images were captured in five random fields.

Luciferase reporter assay

The lncRNA LIFR-AS1 fragment or the 3′-untranslated region (UTR) of MTUS1 was amplified by performing PCR. Then, the pmirGLO luciferase vector (Promega Corporation) was used to establish LIFR-AS1 wild type (WT) and MTUS1-WT. A site-directed mutagenesis kit (Thermo Fisher Scientific, Inc.) was employed to generate LIFR-AS1 mutant (MUT) or MTUS1-MUT. All the above constructed vectors and miR-4698 mimics (500 ng) were co-transfected into MKN45 and AGS cells using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.). After 48 h transfection, the relative luciferase activity was assessed using a dual luciferase reporter assay system (Promega Corporation). Renilla luciferase activity was used for normalization.

Reverse transcription-quantitative (RT-q) PCR

Total RNA was isolated from GC tissues and cell lines (5×106 cells/ml) using TRIzol® reagent (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. A Reverse Transcription kit (Promega Corporation) was used to synthesize cDNA according to the manufacturer's protocols. SYBR® Green PCR Kit (Qiagen, Inc.) was used to observe lncRNA LIFR-AS1 and MTUS1 expression. All reactions were performed in a 10 µl reaction volume in triplicate. PCR were with conditions of 95°C for 50 sec, followed by 40 cycles of 95°C for 12 sec and 55.5°C for 30 sec. For detecting miR-4698 expression, TaqMan Universal Master Mix II (Takara Biotechnology Co., Ltd.) was employed for RT-qPCR procedure. β-actin and U6 served as internal control for standardization of the data. All the data were analyzed by the 2−ΔΔCq method (26). Primer sequences are listed in Table II.

Table II.

Primers used for reverse transcription-quantitative PCR.

Table II.

Primers used for reverse transcription-quantitative PCR.

Primer nameSequence
LIFR-AS1Forward 5′-GCAAATACTGTGTATTAGTCC-3′
Reverse 5′-CCGCTTCCTTGTGAAGAAGGT-3′
miR-4698Forward 5′-TGGTACTGATGTGATGGACT-3′
Reverse 5′-TCATATCACACAGCACCGAT-3′
MTUS1Forward 5′-GAGCTGAGCACTTACAGCAACAA-3′
Reverse 5′-TTCAACTGCATTAAGAGCTGTAA-3′
U6Forward 5′-CTCGCTTCGGCAGCACA-3′
Reverse 5′-AACGCTTCACGAATTTGCGT-3′
β-actinForward 5′-CCTGACGCCAACACACTGC-3′
Reverse 5′-ATACTCCTGCTTGGTGATCC-3′
Western blot assay

Following cell transfection, proteins were extracted with RIPA buffer with phenylmethanesulfonyl fluoride (Beyotime Institute of Biotechnology). The protein content was assessed using the BCA method. Afterwards, total protein (30 µg) was separated via 10% SDS-PAGE (Beyotime Institute of Biotechnology). The separated proteins were transferred to polyvinylidene fluoride (PVDF) membranes (EMD Millipore). Following blocking with 5% skimmed milk powder for 2 h at room temperature, the primary antibodies of MTUS1 (1:1,000, cat. no. sc-393120, Santa Cruz Biotechnology Inc.), MEK (1:1,000, cat. no. ab32091), phosphorylated (p-)MEK (1:1,000, cat. no. ab96379), ERK (1:1,000, cat. no. ab184699), p-ERK (1:1,000, cat. no. ab201015), Cell division cycle-25 (Cdc25)B (1:1,000, cat. no. ab124819), cyclin-dependent kinase (Cdk)1 (1:1,000, cat. no. ab133327), p-Cdk1 (1:1,000, cat. no. ab201008) and GAPDH (1:2,000, cat. no. ab8245; all from Abcam) were incubated with the PVDF membranes overnight at 4°C. Next, the appropriate secondary antibodies [1:5,000; goat anti-mouse IgG H&L (HRP) cat. no. ab205719 or goat anti-rabbit IgG H&L (HRP) cat. no. ab6721, Abcam] were used to incubate the above membranes for additional 1 h at room temperature. The protein levels were monitored by enhanced chemiluminescence reagents (Amersham Biosciences; Cytiva). The protein bands were quantified using Quantity One software (v4.6.6, Bio-Rad Laboratories, Inc.).

Bioinformatics analysis

The expression levels of lncRNA LIFR-AS1 in GC tumors and normal tissues was downloaded from The Cancer Genome Atlas (TCGA; http://portal.gdc.cancer.gov/). DIANA tools (http://carolina.imis.athena-innovation.gr/diana_tools/web/index.php) was used to predict potential binding miRNAs for lncRNA LIFR-AS1. The potential target gene of miRNA was predicted by TargetScan software, version 7.2 (http://www.targetscan.org/vert_72/).

Statistical analysis

All data are presented as the mean ± standard deviation. SPSS 19.0 statistical software (IBM Corp.) was used for statistical analyses. Relevance between lncRNA LIFR-AS1 and miR-4698 or MTUS1 was assessed by using Pearson correlation analysis. Comparisons between different groups were analyzed by using Student's t-test or one-way ANOVA followed by Tukey's multiple comparisons test. P<0.05 was considered to indicate a statistically significant difference.

Results

Downregulation of lncRNA LIFR-AS1 in GC tissues and cell lines

To investigate the biological role of LIFR-AS1 in GC, data from normal and tumor samples were obtained from TCGA and it was found that LIFR-AS1 was significantly downregulated in GC tissues compared with normal tissues (P=0.017; Fig. 1A). Expression of lncRNA LIFR-AS1 was examined in 41 GC tissues and the adjacent tissues using RT-qPCR. The low expression of lncRNA LIFR-AS1 was clearly presented in GC tissues (P<0.001, Fig. 1B). The relationship between lncRNA LIFR-AS1 expression and clinical characteristics is shown in Table I. In the GC tissues of patients with poorly differentiation degree, III and IV TNM stage and pN1-3 nodal status, the expression of lncRNA LIFR-AS1 was significantly reduced (P<0.05, P<0.01; Fig. 1C-E). Furthermore, the expression of lncRNA LIFR-AS1 was also explored in GES-1 and GC cell lines (MKN45 and AGS). It was identified that expression of lncRNA LIFR-AS1 was lower in MKN45 and AGS cells compared with that in GES-1 cells (P<0.01; Fig. 1F). These results demonstrated that lncRNA LIFR-AS1 was downregulated in GC tissues and cells.

Overexpression of lncRNA LIFR-AS1 restrains GC cell proliferation and movement

The overexpressed vector of lncRNA LIFR-AS1 (pcDNA-LIFR-AS1) was transfected into MKN45 and AGS cells. Expression of lncRNA LIFR-AS1 was distinctly upregulated in pcDNA-LIFR-AS1-transfected MKN45 and AGS cells (P<0.01; Fig. 2A). CCK-8 experimental results revealed that overexpressed lncRNA LIFR-AS1 reduced cell viability at days 2, 3 and 4 in MKN45 and AGS cells (P<0.05, P<0.01; Fig. 2B). In addition, the number of EdU positive cells was also reduced by overexpressed lncRNA LIFR-AS1 (P<0.01; Fig. 2C). Cell migratory and invasive ability was then evaluated by wound and cell invasion assays. It was noted that overexpressed lncRNA LIFR-AS1 significantly decreased the wound healing areas in MKN45 and AGS cells (P<0.01; Fig. 2D). Similary, the capacity of cell invasion was also inhibited by overexpressed lncRNA LIFR-AS1 (P<0.01; Fig. 2E). All above results suggested that lncRNA LIFR-AS1 suppressed GC cell proliferation, migration and invasion.

Negative correlation between lncRNA LIFR-AS1 and miR-4698

Fig. 3A shows the predicted binding site of miR-4698 in lncRNA LIFR-AS1. To verify the predicted sequence binding sites between lncRNA LIFR-AS1 and miR-4698, luciferase reporter gene assay was performed to further confirm their association. It was observed that the luciferase activity was reduced in MKN45 and AGS cells with LIFR-AS1-WT and miR-4698 mimics co-transfection (P<0.05; Fig. 3B). It was noted that overexpressed lncRNA LIFR-AS1 decreased miR-4698 expression in MKN45 and AGS cells (P<0.01; Fig. 3C). Notably, upregulated miR-4698 expression was found in GC tissues (P<0.001; Fig. 3D) in addition to MKN45 and AGS cells (P<0.01; Fig. 3E). A Pearson correlation test revealed a negative correlation between lncRNA LIFR-AS1 and miR-4698 (P<0.05; Fig. 3F). All these findings demonstrated the negative correlation between lncRNA LIFR-AS1 and miR-4698 and that lncRNA LIFR-AS1 worked as a sponge of miR-4698 in GC cells.

MTUS1 is a target of miR-4698 and is positively associated with lncRNA LIFR-AS1

TargetScan revealed that the 3′-UTR of MTUS1 contained the complementary site for the seed region of miR-4698 (Fig. 4A). Following co-transfection with miR-4698 mimics and the fragments of MTUS1-WT or MTUS1-MUT, the change in the luciferase activity was evaluated. The results demonstrated inhibited luciferase activity in miR-4698 mimics and MTUS1-WT co-transfected cells (P<0.05; Fig. 4B). The overexpressed and suppressed vectors of miR-4698 (miR-4698 mimics and miR-4698 inhibitors) were transfected into GC cells and the correlation between miR-4698 and MTUS1 was monitored. Expression of miR-4698 was clearly enhanced in miR-4698 mimics-transfected cells (P<0.01), but restricted in miR-4698 inhibitors-transfected cells (P<0.05; Fig. 4C). Western blotting results revealed that MTUS1 expression was inhibited by miR-4698 overexpression and increased by miR-4698 inhibition (P<0.05; Fig. 4D). Fig. 4E and F demonstrate the downregulation of MTUS1 in GC tissues (P<0.001) and in GC (MKN45 and AGS) cell lines (P<0.01). A Pearson correlation test further confirmed the negative correlation between MTUS1 and miR-4698 (Fig. 4G) and the positive correlation between MTUS1 and lncRNA LIFR-AS1 (Fig. 4H). Together, these results demonstrated that MTUS1 was a new predicated target gene of miR-4698 and was also positively regulated by lncRNA LIFR-AS1.

MEK/ERK pathway is counteracted by overexpression of lncRNA LIFR-AS1 via regulation of MTUS1

The signaling pathway regulation was assessed when cells were transfected with the expression vectors of pcDNA-LIFR-AS1 and si-MTUS1. It was noted that overexpressed lncRNA LIFR-AS1 inhibited p-MEK and p-ERK expression in GC (MKN45 and AGS) cell lines (P<0.05; Fig. 5A). si-MTUS1 vector was transfected into GC cells to inhibit MTUS1 expression and the relationship between lncRNA LIFR-AS1 and MTUS1 explored. It was observed that overexpressed lncRNA LIFR-AS1 upregulated MTUS1 expression in MKN45 and AGS cells (P<0.01; Fig. 5B). By contrast, silencing of MTUS1 increased p-MEK and p-ERK expression (P<0.05, Fig. 5C). Following pcDNA-LIFR-AS1 and si-MTUS1 co-transfection, the suppressed p-MEK and p-ERK expression in GC (MKN45 and AGS) cell lines was reversed by si-MTUS1 (P<0.01; Fig. 5C). On the basis of these results, it was hypothesized that the MEK/ERK pathway was hindered by overexpressed lncRNA LIFR-AS1 modulating MTUS1.

Overexpression of lncRNA LIFR-AS1 inhibits Cdc25B and inactivates Cdk1 signaling

The present study identified that overexpressed lncRNA LIFR-AS1 inhibited Cdc25B expression in GC (MKN45 and AGS) cell lines. The tyrosine phosphorylation of Cdk1 was induced by overexpression of lncRNA LIFR-AS1 in GC (MKN45 and AGS) cell lines (P<0.05; Fig. 6A). Following pcDNA-LIFR-AS1 and si-MTUS1 co-transfection, the suppressed Cdc25B expression and the induced tyrosine phosphorylation of Cdk1 in GC (MKN45 and AGS) cell lines was reversed by si-MTUS1 (P<0.01; Fig. 6B). These results demonstrated that the inhibition of Cdc25B activity by lncRNA LIFR-AS1 could contribute to Cdk1 tyrosine phosphorylation.

Discussion

GC has become one of the leading causes of mortality in individuals and its pathogenesis is still unclear (27). In the treatment of GC priority is given to surgical operation, whereas the unfavorable prognosis remains an enormous challenge in clinical practice. Due to their clinical potential in cancer treatment, lncRNAs have been extensively studied in GC (28). Wu et al (29) reported that lncRNA ZEB2-AS1 is upregulated in gastric cancer and affects cell proliferation and invasion via miR-143-5p/HIF-1α axis. Xiao et al (30) found that lncRNA MALAT1 increases the stemness of gastric cancer cells via enhancing SOX2 mRNA stability. However, the influence of lncRNA LIFR-AS1 in GC remains to be elucidated. The present study first investigated the functions of lncRNA LIFR-AS1 in GC cell proliferation and movement and uncovered the potential molecular mechanisms. Decreased expression of lncRNA LIFR-AS1 was identified in GC tissues and cell lines. Further study revealed that lncRNA LIFR-AS1 inhibited GC cell proliferation, migration and invasion through sponging miR-4698. In addition, MTUS1 was verified to be a target gene of miR-4698 and positively regulated by lncRNA LIFR-AS1. Furthermore, lncRNA LIFR-AS1 blocked the MEK/ERK pathway by mediating MTUS1.

lncRNA LIFR-AS1 is derived from the antisense transcription of the LIFR gene located on human chromosome 5p13.1, which serves a crucial role in the pathogenesis of a number of illnesses (31). A previous study revealed that lncRNA LIFR-AS1 negatively correlates with tumor size in uterine fibroids (32). Additionally, Wang et al (13) found that lncRNA LIFR-AS1 is downregulated in breast cancer. In accordance with that study, the present study also observed decreased lncRNA LIFR-AS1 expression in GC tissues and cell lines. Xu et al (14) explored the effects of lncRNA LIFR-AS1 on breast cancer cell proliferation and migration and verify its inhibitory role in breast cancer progression. On the basis of that research, the functions of lncRNA LIFR-AS1 in GC cell proliferation, migration and invasion were further investigated. The present study also demonstrated the suppressed functions of lncRNA LIFR-AS1 in GC cell proliferation, migration and invasion. These findings suggested that lncRNA LIFR-AS1 participated in the control of the GC progression.

Previous studies have demonstrated the vital regulatory functions of lncRNA-miRNA network in types of human cancers, such as diabetic pancreatic cancer and colorectal cancer (33,34). In the field of GC, lncRNA gastric cancer-related lncRNA1 has been reported to modulate GC cell proliferation and metastasis by sponging miR-885-3p (35). lncRNA XIST promotes GC cell growth and invasion by regulating miR-497 (36). miR-4698 is a novel miRNA and its downregulation is reported in chronic obstructive pulmonary disease (37). Similarly, a profile of miRNAs by microarrays in Liu et al (38) demonstrates that miR-4698 expression is upregulated in gastric cancer stem cells. Considering its abnormal expression in different diseases, the present study attempted to uncover the influence of miR-4698 in lncRNA LIFR-AS1-affected GC cell proliferation, migration and invasion. Upregulated expression of miR-4698 was observed in GC tissues and cell lines. In addition, a reciprocal suppression between miR-4698 and lncRNA LIFR-AS1 was identified. Thus, it was concluded that lncRNA LIFR-AS1 exerted inhibitory functions in GC cell proliferation, migration and invasion through sponging miR-4698.

MTUS1 is reported to locate on the reverse strand of the chromosome 8p22, which can encode angiotensin II AT2 receptor-interacting proteins (39). As a tumor suppressor, MTUS1 has been investigated in colon and prostate cancer and its aberrant expression is associated with the development of these cancers (40,41). The present study also observed the downregulated expression of MTUS1 in GC tissues and cell lines. In a previous study, silencing of MTUS1 facilitates GC cell growth and metastasis (42). The results of the present study confirmed that MTUS1 was a target gene of miR-4698. In addition, a positive correlation between MTUS1 and lncRNA LIFR-AS1 was identified. Given the relationship of lncRNA LIFR-AS1, miR-4698 and MTUS1, it was hypothesized that MTUS1 probably joined in modulating the functions of lncRNA LIFR-AS1 in GC.

MEK/ERK is a highly conserved cell signaling pathway, existing in eukaryotic cells, which regulates diverse cell behaviors in human cancers (43,44). The MEK/ERK pathway participates in adjusting the cisplatin resistance in GC cells (45) and inactivation of the MEK/ERK pathway can affect the proliferative ability of GC cells (46). Several lncRNAs affect cancer cell proliferation and metastasis by controlling the MEK/ERK pathway (47,48). However, whether lncRNA LIFR-AS1 regulates the MEK/ERK pathway to restrain GC cell proliferation and movement remains unreported. The present study showed that overexpressed lncRNA LIFR-AS1 inhibited the MEK/ERK pathway. The process was reversed by silencing of MTUS1. These findings corroborated that the MEK/ERK pathway was inhibited by overexpression of lncRNA LIFR-AS1 via modulating MTUS1.

Cdc25 serves an important role in transitions between cell-cycle phases by dephosphorylating and activating Cdks (49). ERK-MAP kinases are directly involved in activating Cdc25 during the G(2)/M transition (50). A previous study reported that small-molecule Cdc25 inhibitors inhibit Cdc25 enzymatic activities and induce tyrosine phosphorylation of the Cdc25-targeted Cdks (51). The present study identified that lncRNA LIFR-AS1 inhibited Cdc25B activity and contributed to Cdk1 tyrosine phosphorylation and cell growth inhibition.

The present study had some limitations. First, animal experiments to investigate the roles of lncRNA LIFR-AS1 in GC were not performed. Second, the role of lncRNA LIFR-AS1 on tumor cell apoptosis was unknown. Third, the underlying molecular mechanism of lncRNA LIFR-AS1 downregulation in GC cells was not elucidated.

In summary, the present study showed that lncRNA LIFR-AS1 was downregulated in GC tissues and was associated with the prognosis of patients with GC. lncRNA LIFR-AS1 inhibited GC cell proliferation, migration and invasion. Results revealed that lncRNA LIFR-AS1 interacted with miR-4698 to regulate MTUS1 expression. These results provided a new lncRNA-directed therapeutics for GC. However, the detailed effects of miR-4698 and MTUS1 on GC cell behaviors remain to be elucidated.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

JC and JZ conceived and designed the study. JZ, XL, LF, NZ and JY performed the study, collected the data and analyzed the data. JC wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All patients personally signed an informed consent. The protocol of the present study was approved by the Ethics Committee of Cangzhou People's Hospital (approval number AF/SC-08/02.0).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhong Q, Sun Q, Xu GF, Fan XQ, Xu YY, Liu F, Song SY, Peng CY and Wang L: Differential analysis of lymph node metastasis in histological mixed-type early gastric carcinoma in the mucosa and submucosa. World J Gastroenterol. 24:87–95. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Tanahashi T, Yoshida K and Yamaguchi K: Current status and future perspective of the chemotherapy for gastric cancer. Nihon Shokakibyo Gakkai Zasshi. 115:500–506. 2018.(In Japanese). PubMed/NCBI

3 

Li T, Gao X, Han L, Yu J and Li H: Identification of hub genes with prognostic values in gastric cancer by bioinformatics analysis. World J Surg Oncol. 16:1142018. View Article : Google Scholar : PubMed/NCBI

4 

Dreznik A, Purim O, Idelevich E, Kundel Y, Sulkes J, Sulkes A and Brenner B: Gastric cancer: Biology and clinical manifestations in Israel. J Surg Oncol. 105:316–322. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Sasaki T: Discussion for gastric cancer treatment guidelines in Japan. Nihon Rinsho. 61:13–18. 2003.(In Japanese).

6 

Kim H, Kim H, Kim SY, Kim TY, Lee KW, Baek SK, Kim TY, Ryu MH, Nam BH and Zang DY: Second-line chemotherapy versus supportive cancertreatment in advanced gastric cancer: A meta-analysis. Ann Oncol. 24:2850–2854. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Cong P, Zhang G, Chen Z, Chen Y and Zhang L: lncRNApred: Classification of long non-coding RNAs and protein-coding transcripts by the ensemble algorithm with a new hybrid feature. PLoS One. 11:e01545672016. View Article : Google Scholar : PubMed/NCBI

8 

Cao MX, Jiang YP, Tang YL and Liang XH: The crosstalk between lncRNA and microRNA in cancer metastasis: Orchestrating the epithelial-mesenchymal plasticity. Oncotarget. 8:12472–12483. 2015. View Article : Google Scholar

9 

Chen D, Sun Q, Cheng X, Zhang L, Song W, Zhou D, Lin J and Wang W: Genome-wide analysis of long noncoding RNA (lncRNA) expression in colorectal cancer tissues from patients with liver metastasis. Cancer Med. 5:1629–1639. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Li H, Yu B, Li J, Su L, Yan M, Zhu Z and Liu B: Overexpression of lncRNA H19 enhances carcinogenesis and metastasis of gastric cancer. Oncotarget. 5:2318–2329. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Sun M, Nie F, Wang Y, Zhang Z, Hou J, He D, Xie M, Xu L, De W, Wang Z and Wang J: lncRNA HOXA11-AS promotes proliferation and invasion of gastric cancer by scaffolding the chromatin modification factors PRC2, LSD1 and DNMT1. Cancer Res. 76:6299–6310. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Wang Y, Liu X, Zhang H, Sun L, Zhou Y, Jin H, Zhang H, Zhang H, Liu J, Guo H, et al: Hypoxia-inducible lncRNA-AK058003 promotes gastric cancer metastasis by targeting γ-synuclein. Neoplasia. 16:1094–1106. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Wang L, Li J, Zhao H, Hu J, Ping Y, Li F, Lan Y, Xu C, Xiao Y and Li X: Identifying the crosstalk of dysfunctional pathways mediated by lncRNAs in breast cancer subtypes. Mol Biosyst. 12:711–720. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Xu F, Li H and Hu C: LIFR-AS1 modulates Sufu to inhibit cell proliferation and migration by miR-197-3p in breast cancer. Biosci Rep. 39:BSR201805512019. View Article : Google Scholar : PubMed/NCBI

15 

Liu K, Yao H, Wen Y, Zhao H, Zhou N, Lei S and Xiong L: Functional role of a long non-coding RNA LIFR-AS1/miR-29a/TNFAIP3 axis in colorectal cancer resistance to pohotodynamic therapy. Biochim Biophys Acta Mol Basis Dis. 1864:2871–2880. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Yanaihara N and Harris C: MicroRNA involvement in human cancers. Clin Chem. 59:1811–1812. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Matamala N, Vargas MT, González-Cámpora R, Miñambres R, Arias JI, Menéndez P, Andrés-León E, Gómez-López G, Yanowsky K, Calvete-Candenas J, et al: Tumor MicroRNA expression profiling identifies circulating MicroRNAs for Early breast cancer detection. Clin Chem. 61:1098–1096. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Yu Y, Nangia-Makker P, Farhana L and Majumdar A: A novel mechanism of lncRNA and miRNA interaction: CCAT2 regulates miR-145 expression by suppressing its maturation process in colon cancer cells. Mol Cancer. 16:1552017. View Article : Google Scholar : PubMed/NCBI

19 

Hao NB, He YF, Li XQ, Wang K and Wang RL: The role of miRNA and lncRNA in gastric cancer. Oncotarget. 8:81572–81582. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Mikkelsen LH, Andersen MK, Andreasen S, Larsen AC, Tan Q, Toft PB, Wadt K and Heegaard S: Global microRNA profiling of metastatic conjunctival melanoma. Melanoma Res. 29:465–473. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Kalhori MR, Arefian E, Fallah Atanaki F, Kavousi K and Soleimani M: miR-548× and miR-4698 controlled cell proliferation by affecting the PI3K/AKT signaling pathway in glioblastoma cell lines. Sci Rep. 10:15582020. View Article : Google Scholar : PubMed/NCBI

22 

Seibold S, Rudroff C, Weber M, Galle J, Wanner C and Marx M: Identification of a new tumor suppressor gene located at chromosome 8p21.3-22. FASEB J. 17:1180–1182. 2003. View Article : Google Scholar : PubMed/NCBI

23 

Kara M, Kaplan M, Bozgeyik I, Ozcan O, Celik OI, Bozgeyik E and Yumrutas O: MTUS1 tumor suppressor and its miRNA regulators in fibroadenoma and breast cancer. Gene. 587:173–177. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Ozcan O, Kara M, Yumrutas O, Bozgeyik E, Bozgeyik I and Celik OI: MTUS1 and its targeting miRNAs in colorectal carcinoma: Significant associations. Tumour Biol. 37:6637–6645. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Xiao J, Chen JX, Zhu YP, Zhou LY, Shu QA and Chen LW: Reduced expression of MTUS1 mRNA is correlated with poor prognosis in bladder cancer. Oncol Lett. 4:113–118. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Livak K and Schmittgen T: 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

27 

Van Cutsem E, Sagaert X, Topal B, Haustermans K and Prenen H: Gastric cancer. Lancet. 388:2654–2664. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Zhang G, Li S, Lu J, Ge Y, Wang Q, Ma G, Zhao Q, Wu D, Gong W, Du M, et al: lncRNA MT1JP functions as a ceRNA in regulating FBXW7 through competitively binding to miR-92a-3p in gastric cancer. Mol Cancer. 17:872018. View Article : Google Scholar : PubMed/NCBI

29 

Wu F, Gao H, Liu K, Gao B, Ren H, Li Z and Liu F: The lncRNA ZEB2-AS1 is upregulated in gastric cancer and affects cell proliferation and invasion via miR-143-5p/HIF-1α axis. Onco Targets Ther. 12:657–667. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Xiao Y, Pan J, Geng Q and Wang G: lncRNA MALAT1 increases the stemness of gastric cancer cells via enhancing SOX2 mRNA stability. FEBS Open Bio. 9:1212–1222. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Frey HA, Stout MJ, Pearson LN, Tuuli MG, Cahill AG, Strauss JF III, Gomez LM, Parry S, Allsworth JE and Macones GA: Genetic variation associated with preterm birth in African-American women. Am J Obstet Gynecol. 215:235.e1–e8. 2016. View Article : Google Scholar

32 

Aissani B, Zhang K and Wiener H: Genetic determinants of uterine fibroid size in the multiethnic NIEHS uterine fibroid study. Int J Mol Epidemiol Genet. 6:9–19. 2015.PubMed/NCBI

33 

Yao K, Wang Q, Jia J and Zhao H: A competing endogenous RNA network identifies novel mRNA, miRNA and lncRNA markers for the prognosis of diabetic pancreatic cancer. Tumor Biol. 39:10104283177078822017. View Article : Google Scholar

34 

Matboli M, Shafei A and Ali M, El-Din Ahmed TS, Naser M, Abdel-Rahman T, Anber N and Ali M: Role of extracellular lncRNA-SNHG14/miRNA-3940-5p/NAP12 mRNA in colorectal cancer. Arch Physiol Biochem. 1–7. 2019.doi: 10.1080/13813455.2019.1650070 (Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

35 

Lin Z, Zhou Z, Guo H, He Y, Pang X, Zhang X, Liu Y, Ao X, Li P and Wang J: Long noncoding RNA gastric cancer-related lncRNA1 mediates gastric malignancy through miRNA-885-3p and cyclin-dependent kinase 4. Cell Death Dis. 9:6072018. View Article : Google Scholar : PubMed/NCBI

36 

Ma L, Zhou Y, Luo X, Gao H, Deng X and Jiang Y: Long non-coding RNA XIST promotes cell growth and invasion through regulating miR-497/MACC1 axis in gastric cancer. Oncotarget. 8:4125–4135. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Shen Z, Tang W, Guo J and Sun S: miR-483-5p plays a protective role in chronic obstructive pulmonary disease. Int J Mol Med. 40:193–200. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Liu J, Ma L, Wang Z, Wang L, Liu C, Chen R and Zhang J: MicroRNA expression profile of gastric cancer stem cells in the MKN-45 cancer cell line. Acta Biochim Biophys Sin (Shanghai). 46:92–99. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Di Benedetto M, Bièche I, Deshayes F, Vacher S, Nouet S, Collura V, Seitz I, Louis S, Pineau P, Amsellem-Ouazana D, et al: Structural organization and expression of human MTUS1, a candidate 8p22 tumor suppressor gene encoding a family of angiotensin II AT2 receptor-interacting proteins, ATIP. Gene. 380:127–136. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Zuern C, Heimrich J, Kaufmann R, Richter KK, Settmacher U, Wanner C, Galle J and Seibold S: Down-regulation of MTUS1 in human colon tumors. Oncol Rep. 23:183–189. 2010.PubMed/NCBI

41 

Louis SN, Chow L, Rezmann L, Krezel MA, Catt KJ, Tikellis C, Frauman AG and Louis WJ: Expression and function of ATIP/MTUS1 in human prostate cancer cell lines. Prostate. 70:1563–1574. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Li X, Liu H, Yu T, Dong Z, Tang L and Sun X: Loss of MTUS1 in gastric cancer promotes tumor growth and metastasis. Neoplasma. 61:128–135. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Zhang X, Liu G, Ding L, Jiang T, Shao S, Gao Y and Lu Y: HOXA3 promotes tumor growth of human colon cancer through activating EGFR/Ras/Raf/MEK/ERK signaling pathway. J Cell Biochem. 119:2864–2874. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Hu G, Zhang J, Xu F, Deng H, Zhang W, Kang S and Liang W: SLP-2 inhibits cisplatin-induced apoptosis through MEK/ERK signaling and mitochondrial apoptosis pathway in cervical cancer cells. Cancer Sci. 109:1357–1368. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Fu X, Feng J, Zeng D, Ding Y, Yu C and Yang B: PAK4 confers cisplatin resistance in gastric cancer cells via PI3K/Akt- and MEK/ERK-dependent pathways. Biosci Rep. 34:e000942014. View Article : Google Scholar : PubMed/NCBI

46 

Lin Z, Zhang C, Zhang M, Xu D, Fang Y, Zhou Z, Chen X, Qin N and Zhang X: Targeting Cadherin-17 Inactivates Ras/Raf/MEK/ERK signaling and inhibits cell proliferation in gastric cancer. PLoS One. 9:e852962014. View Article : Google Scholar : PubMed/NCBI

47 

Zhuang R, Zhang X, Lu D, Wang J, Zhuo J, Wei X, Ling Q, Xie H, Zheng S and Xu X: lncRNA DRHC inhibits proliferation and invasion in hepatocellular carcinoma via c-Myb-regulated MEK/ERK signaling. Mol Carcinog. 58:366–375. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Wang Y, Gu J, Lin X, Yan W, Yang W and Wu G: lncRNA BANCR promotes EMT in PTC via the Raf/MEK/ERK signaling pathway. Oncol Lett. 15:5865–5870. 2018.PubMed/NCBI

49 

Sur S and Agrawal DK: Phosphatases and kinases regulating CDC25 activity in the cell cycle: Clinical implications of CDC25 overexpression and potential treatment strategies. Mol Cell Biochem. 416:33–46. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Wang R, He G, Nelman-Gonzalez M, Ashorn CL, Gallick GE, Stukenberg PT, Kirschner MW and Kuang J: Regulation of Cdc25C by ERK-MAP kinases during the G2/M transition. Cell. 128:1119–1132. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Peyregne VP, Kar S, Ham SW, Wang M, Wang Z and Carr BI: Novel hydroxyl naphthoquinones with potent Cdc25 antagonizing and growth inhibitory properties. Mol Cancer Ther. 4:595–602. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 23 Issue 2

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
Zhao J, Li X, Fu L, Zhang N, Yang J and Cai J: lncRNA LIFR‑AS1 inhibits gastric carcinoma cell proliferation, migration and invasion by sponging miR‑4698. Mol Med Rep 23: 153, 2021
APA
Zhao, J., Li, X., Fu, L., Zhang, N., Yang, J., & Cai, J. (2021). lncRNA LIFR‑AS1 inhibits gastric carcinoma cell proliferation, migration and invasion by sponging miR‑4698. Molecular Medicine Reports, 23, 153. https://doi.org/10.3892/mmr.2020.11792
MLA
Zhao, J., Li, X., Fu, L., Zhang, N., Yang, J., Cai, J."lncRNA LIFR‑AS1 inhibits gastric carcinoma cell proliferation, migration and invasion by sponging miR‑4698". Molecular Medicine Reports 23.2 (2021): 153.
Chicago
Zhao, J., Li, X., Fu, L., Zhang, N., Yang, J., Cai, J."lncRNA LIFR‑AS1 inhibits gastric carcinoma cell proliferation, migration and invasion by sponging miR‑4698". Molecular Medicine Reports 23, no. 2 (2021): 153. https://doi.org/10.3892/mmr.2020.11792