Open Access

LncRNA GAS6 antisense RNA 1 facilitates the tumorigenesis of clear cell renal cell carcinoma by regulating the AMP‑activated protein kinase/mTOR signaling pathway

Retraction in: /10.3892/ol.2024.14259

  • Authors:
    • Xiaoyun Guo
    • Hongjun Li
    • Mei Zhang
    • Rong Li
  • View Affiliations

  • Published online on: August 10, 2021     https://doi.org/10.3892/ol.2021.12988
  • Article Number: 727
  • Copyright: © Guo 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 role of GAS6 antisense RNA 1 (GAS6‑AS1) in clear cell renal cell carcinoma (ccRCC) remains unclear. The aim of the present study was to investigate the role and molecular mechanisms of GAS6‑AS1 in the progression of ccRCC. GAS6‑AS1 was found to be upregulated in ccRCC tissues and cell lines, and patients with high GAS6‑AS1 expression levels exhibited a poor prognosis. Small interfering (si)RNA GAS6‑AS1 inhibited the activity, colony formation, invasiveness and glycolysis of OSRC‑2 and SW839 cells, while GAS6‑AS1 overexpression promoted these functions. Moreover, si‑GAS6‑AS1 increased the phosphorylation level of AMP‑activated protein kinase (AMPK) and decreased that of mTOR, as well as decreasing proliferating cell nuclear antigen (PCNA), MMP‑2 and hexokinase‑2 (HK2) expression, which were reversed by inhibiting AMPK or mTOR. In addition, the silencing of GAS6‑AS1 suppressed the growth of xenografted tumors and attenuated the expression of PCNA, MMP‑2 and HK2 in tumor tissues. These findings conclude that GAS6‑AS1 regulated the proliferation, invasiveness and glycolysis of ccRCC cells by regulating the AMPK/mTOR signaling pathway, and suggest that GAS6‑AS1 may be a potential therapeutic target for ccRCC.

Introduction

Renal cell carcinoma (RCC) is the most common malignant tumor of the kidney, accounting for 2–3% of all malignant tumors in adults (1). Clear cell renal cell carcinoma (ccRCC) is the most common and malignant subtype of RCC, accounting for 75–80% of RCC cases (2). Radical nephrectomy is the primary and most effective treatment for ccRCC. However, the recurrence rate after treatment is high, and the prognosis is poor (3). Therefore, it is paramount to elucidate the molecular mechanisms underlying the occurrence and development of ccRCC, and to identify early diagnostic molecular markers and new therapeutic targets.

LncRNAs are the most highly-researched type of non-coding RNA in mammals, and play important roles in various biological processes (4,5). The expression of lncRNA is tissue-specific and influences cell cycle regulation, cellular proliferation, the immune response and cellular pluripotency (68). Notably, some lncRNAs are associated with the regulation of multiple tumor-related pathways in the occurrence and development of tumors (9,10). With the rapid development of biotechnology, lncRNAs have shown promise as treatments and novel targets for cancer therapy (11,12). In ccRCC, the downregulation of lncRNA FOXO induced long non-coding RNA 1 (FILNC1) inhibited apoptosis induced by energy deficit, promoting cellular proliferation (13). Furthermore, lncRNA HOX transcript antisense RNA (HOTAIR) was found to promote the development of ccRCC through the microRNA (miR)-217/hypoxia-inducible factor 1α/AXL signaling pathway (14), and lncRNA colon cancer associated transcript 1 was found to promote cellular proliferation and invasiveness by regulating Wnt/β-catenin signaling in ccRCC (15). LncRNA metastatic renal cell carcinoma-associated transcript 1 promoted metastasis in ccRCC by activating the p38 MAPK signaling pathway (16).

GAS6 antisense RNA 1 (GAS6-AS1) is an antisense RNA downstream of GAS6, located on human chromosome 13q34 (17). GAS6-AS1 is reportedly highly expressed in hepatocellular carcinoma and gastric cancer, where it plays an oncogenic role (17,18). However, to the best of our knowledge, there are few studies on the relationship between GAS6-AS1 and ccRCC. Thus the biological effects and possible mechanisms of GAS6-AS1 in ccRCC require further clarification.

As an important nodal molecule in cellular metabolism, AMP-activated protein kinase (AMPK) plays a key role in regulating energy balance and maintaining ATP/AMP concentrations (19). Studies have found that AMPK kinase can directly or indirectly affect various downstream signaling pathways of AMPK, regulating intracellular oxidative stress (20), autophagy (21), proliferation (22), apoptosis (23) and mitochondrial function (24). Moreover, numerous reports have shown a reciprocal relationship between GAS6 and AMPK (25,26). Consequently, we hypothesized that GAS6-AS1 may affect the proliferation, invasiveness and glycolysis of RCC cells by influencing the activity of AMPK.

In the present study, The Cancer Genome Atlas (TCGA) database analysis indicated that GAS6-AS1 was highly expressed in ccRCC. At the same time, experimental results showed that the expression of GAS6-AS1 in ccRCC tissues and cells was significantly higher than that in adjacent-normal tissues and non-ccRCC cells, respectively, and was negatively associated with poor prognosis in patients with ccRCC. On this basis, the effects of GAS6-AS1 on the proliferation, invasion, glycolysis and activity of AMPK kinase in ccRCC were investigated both in vitro and in vivo.

Materials and methods

Tissue samples

The ccRCC and adjacent-normal tissue samples (at least 2 cm from the tumor margins) were obtained from 40 patients (30 males and 10 females, aged from 45 to 65 years) with ccRCC between June 2017 and September 2018 in the Second Hospital of Tianjin Medical University (Tianjin, China), and immediately stored in liquid nitrogen. The inclusion criteria were: i) Patients with ccRCC confirmed by pathology, ii) suitable formalin-fixed, paraffin-embedded tissue specimens and iii) patients undergoing surgical resection. Exclusion criteria were as follows: i) Any treatment within 3 months before admission and ii) complications of other malignancies. All patients were followed up for 5 years. The present study was approved by the Second Hospital of Tianjin Medical University Ethics Committee [approval no. SYXK (JIN) 2019-0004], and patient consent was sought prior to surgery. Oral informed consent was obtained from all patients, and written consents were signed by all patients.

Dataset analysis

Gene Expression Profiling Interactive Analysis (GEPIA) (http://gepia.cancer-pku.cn/index.html) were used to analyze the expression of PIG11 mRNA in GC. GEPIA is a newly developed interactive web server for analyzing the RNA sequencing expression data of 9,736 tumors and 8,587 normal samples from TCGA and the GTEx projects, using a standard processing pipeline (27).

Cell culture

ccRCC cell lines (OSRC-2, Caki-1, SN12-PM6, A498 and SW839) and normal renal HK-2 cells were obtained from the American Type Culture Collection, and cultured in RPMI-1640 medium containing 10% fetal bovine serum and 0.01% Penicillin-Streptomycin. The cells were maintained in an incubator (37°C) with 5% CO2, and were passaged at 80–90% confluence. The medium was replaced every 2 days.

Transfection

GAS6-AS1-specific small interfering RNA (si-GAS6-AS1#1, 3′-GAAATACATUACAGAGGAAAT-5′; si-GAS6-AS1#2 3′-GAACCTGAACGCTTAATTAUU-5′), siRNA negative control (si-NC, 3′-CUACAACAGCCACAACGUCTT-5′), pcDNA3.1-GAS6-AS1 (GAS6-AS1) and the empty vector pcDNA3.1 (vector) were purchased from Shanghai GenePharma Co., Ltd. Si-NC was used as the control for si-GAS6AS1 transfection, and ‘vector’ was used as the control for GAS6AS1 overexpression. Cells in the logarithmic growth phase were counted and seeded into 24-well culture plates, and recultured until reaching ~60-70% confluent. Then, the cells were transfected with si-GAS6-AS1, si-NC, GAS6-AS1 or vector at 37°C for 6 h using Lipofectamine® 3000 (Thermo Fisher Scientific, Inc.) per the manufacturer's instructions; siRNAs were used at a concentration of 100 nM, and the mass of vectors was 2 µg. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to verify transfection efficiency after 24 h.

Compound C was purchased from Calbiochem; Merck KGaA. mhy1485 was obtained from Sigma-Aldrich; Merck KGaA. After transfection, cells were treated with 40 µM compound C or mhy1485 for 24 h at room temperature.

Cell Counting Kit-8 (CCK-8) assay

After transfection for 24, 48 or 72 h, the medium was discarded, a mixture of 90 µl medium and 10 µl CCK-8 reagent (Beyotime institute of Biotechnology) was added to each well, and the cells were incubated at 37°C for 1 h. The absorbance value at 450 nm was measured using a microplate reader (Bio-Rad Laboratories, Inc.).

Colony formation assay

After transfection for 24 h, the cells were harvested and resuspended in culture medium, prior to seeding at ~1,000 cells per culture dish. The proliferation status of the cells was observed every day until visible cell colonies were present. Subsequently, 5 ml 4% paraformaldehyde was added to each culture dish to fix the cells for 2 h. After washing twice with PBS, the cells were incubated with 0.1% crystal violet staining solution for 20 min at room temperature (20±5°C), and then slowly washed with water. Images were captured under an inverted microscope (magnification, ×100).

Transwell assay

Matrigel was diluted to 1 mg/ml with precooled serum-free medium; 100 µl Matrigel (EMD Millipore) was added to the lower chamber of the Transwell inserts, and then incubated at 37°C for 1 h. After culturing for 24 h in serum-free medium, the cells were collected and added to the upper chamber, and medium containing 20% FBS was added to the lower chamber. After 24 h, the inserts were washed twice with PBS and fixed with 100% methanol for 10 min at room temperature. Then, the cells in the upper chamber were removed with cotton swabs, and the remaining cells were stained with 0.1% crystal violet solution for 20 min at room temperature. The cells were counted under a light microscope (magnification, ×50). At least 5 random fields were observed, from which the average value was recorded.

RT-qPCR

Total RNA was extracted from the tissues and cells using the minimal universal RNA Extraction Kit (Takara Bio, Inc.,). The purity and yield of total RNA were determined by measuring the absorbance values at 260 and 280 nm, and the ratio of A280/260 was 1.8/2.1. The PrimeScript™ RT Master Mix kit (Takara Bio, Inc.,) was used for one-step reverse transcription per the following conditions: 37°C for 15 min, and 85°C for 5 sec. qPCR was performed using SYBR Premix EX taq™ II (Promega Corporation) on the LightCycler 96 system (Roche Diagnostics). U6 was used as the internal reference gene. The reaction conditions were 42°C for 30 sec, 95°C for 10 sec, 95°C for 5 sec, and 60°C for 30 sec, for a total of 45 cycles. The results were calculated using the 2−ΔΔCq method (12). The following primers were synthesized by Sangon Biotech (Shanghai) Co., Ltd.: GAS6-AS1, forward 5′-ATGCAAGGACGGAACCACACCT-3′ and reverse 5′-GCAGAGGTTTCTGTCTCCATCG-3′; U6, forward 5-CTCGCTTCGGCAGCACA-3′ and reverse 5′-AACGCTTCACGAATTTGCGT-3′.

Glucose uptake, lactate production and ATP production assays

Glucose content, lactate production and ATP level were detected with the corresponding ELISA kits from Nanjing Jiangcheng Bioengineering Institute (28). For glucose uptake (cat. no. F006-1-1), after transfection, the cells were seeded into a 6-well plate at a density of 3×105/ml, and cultured in a 37°C incubator for 48 h. The supernatant was collected and mixed with the working solution at the ratio of 1:100. After incubation at 37°C for 15 min, 100 µl/well supernatant was added to a 96-well plate, and the absorbance value was detected at 505 nm using a microplate reader (Bio-Rad Laboratories, Inc.). The 0-h reading was used as the background value. The protein concentration was determined using a BCA Protein Assay Kit (Beyotime Institute of Biotechnology), and glucose concentration was determined using the following formula: (Background concentration-determination concentration)/sample protein concentration.

For lactate production (cat. no. A121-1-1), 3×105 cells were seeded into a 6-well plate and cultured for 48 h at 37°C. The lactate content in the cells and media were detected. The cells were lysed on ice by ultrasonic fragmentation (45 kHz for 3 sec, and stopped for 2 sec), and the supernatants were harvested by centrifugation (8,050 × g) at 4°C for 10 min. Lactate production was measured in the supernatants according to the manufacturer's instructions, and absorbance was measured at 565 nm using a microplate reader (Bio-Rad Laboratories, Inc.). Finally, the protein concentration was determined using a BCA kit (Beyotime Institute of Biotechnology) to calibrate the lactate production value in each group. The method of quantification was as follows: Determination concentration/sample protein concentration.

To determine the ATP level (cat. no. A095-1-1), 3×105 cells were seeded into a 6-well plate and cultured at 37°C for 48 h. The ATP levels in the cells and media were detected. Ultrasonic fragmentation was used to obtain cell homogenates as aforementioned, and the homogenates were centrifuged at 8,050 × g for 10 min at 4°C. The ATP level in the supernatant was detected using a microplate reader (Bio-Rad Laboratories, Inc.) according to manufacturer's recommendations at an absorbance value of 636 nm.

Xenograft tumor experiment

Male BALB/c-nude mice (n=12, 15–20 g, 4-weeks old) were purchased from Shanghai Lab. Animal Research Center. All the nude mice were housed at 25°C with relative humidity of 45–55% and 12-h light/dark cycle (lights turned on at 8:00 a.m every day and turned off at 8:00 p.m. The illumination was adjusted for 12 h. Mice were free access to food and water. GAS6-AS1 short hairpin RNA (sh-GAS6-AS1, 5′-GGAATGCAGCTGAAAGATTCC-3′) and scrambled sh-Control (sh-NC, 5′-UUUCCGAACGUGUCACGUdTdT-3′) were synthesized by Shanghai GenePharma Co., Ltd. Sh-GAS6-AS1 (50 mM) or sh-NC (50 mM) was transfected into OSRC-2 cells using Lipofectamine® 2000 reagent (Thermo Fisher Scientific, Inc.) for 12 h at 37°C. Subsequent experiments were performed 24 h after transfection. Cells in the logarithmic phase were collected and resuspended in 1 ml PBS. The mice were then subcutaneously injected with 200 µl cell suspension (1×107 cells) at the axilla (2931). The mice were observed every 7 days, and the tumor weight and volume were recorded. After 35 days, the mice were euthanized with intraperitoneal injections of pentobarbital sodium (120 mg/kg), and the xenograft tumors was dissected, weighed, photographed and stored for further analysis. All animal-related experiments were carried out to meet the requirements of the Animal Ethics Committee of the Second Hospital of Tianjin Medical University. The animal protocols were approved by the Animal Care and Use Committee of the Second Hospital of Tianjin Medical University [SYXK (JIN) 2019-0004].

Immunohistochemistry

Mouse tumor samples were used to prepare 4-µm paraffin-embedded sections. After dewaxing, the slices were immersed in citrate antigen repair solution (pH=6). After boiling, the slices were cooled to room temperature; the boiling and cooling procedure was repeated three times. After washing the slices 3 times with PBS, excess liquid was removed with filter paper, and then a circle was drawn around the tissue with histochemical pen. Next, the tissues were blocked with 5% goat serum (Beyotime Institute of Biotechnology) for 40 min at room temperature. After washing 3 times with PBS, the tissues were incubated for 1 h at 37°C with anti-proliferating cell nuclear antigen (PCNA) (1:4,000; cat. no. ab18197,), anti-hexokinase-2 (HK2) (1:250; cat. no. ab104836) and anti-MMP2 (1:100; cat. no. ab235167) antibodies (all Abcam). After washing 3 times with PBS, the tissues were incubated for 30 min at 37° with an HRP-conjugated goat anti-rabbit antibody (Abcam, cat. no. ab6721; 1:100) or HRP-conjugated goat anti-mouse antibody (Abcam, cat. no. ab6721; 1:250), and then washed with PBS once again. The tissues were then incubated with DAB solution in dark for 8 min at room temperature, and washed again before staining with hematoxylin for 5 min at room temperature. After washing, the stained slices were dehydrated in an ascending series of ethanol concentrations (70, 80, 90 and 100% ethanol; 5 min each), and sealed with neutral resin. Images of three areas per section were captured using an optical microscope (magnification, ×200).

Western blotting

Cells were lysed and total protein was extracted using RIPA reagent (Beyotime Institute of Biotechnology). A BCA Protein Assay Kit was used to detect protein concentration, and the concentration was adjusted to the same level in each sample. The protein samples (20 µg per lane) were mixed with 5X loading buffer and boiled at 100°C for 5 min, followed by separation via 12% SDS-PAGE, and transfer to PVDF membranes. The membranes were blocked at room temperature for 2 h with 5% skimmed milk, and then incubated overnight at 4°C with the corresponding primary antibodies. After washing 3 times with PBST, the membranes were incubated for 2 h at room temperature with the corresponding secondary antibody. After washing 3 more times with PBST (with 0.1% Tween-20), the membranes were incubated with ECL reagent (Thermo Fisher Scientific, Inc.), and the gray value of the bands was determined using ImageJ software version 1.8.0 (National Institutes of Health). All antibodies [anti-p-AMPK (cat. no. ab92701; 1:1,000), anti-AMPK (cat. no. ab79885; 1:1,000), anti-p-mTOR (cat. no. ab84400; 1:1,000), anti-mTOR (cat. no. ab2732, 1:1,000), anti-PCNA (cat. no. ab18197; 1:1,000), anti-MMP2 (cat. no. ab92536; 1:1,000), anti-HK2 (cat. no. ab104836; 1:1,000) and anti-GAPDH (cat. no. ab9485; 1:2,000)] were obtained from Abcam. Expression levels were normalized to that of GAPDH.

Statistical analysis

All data were analyzed using GraphPad prism 6 (GraphPad Software, Inc.), and are expressed as the mean ± SD. Multiple comparisons were performed by one-way ANOVA followed by Tukey's post hoc test, and pairwise comparisons were performed using paired t-test (Fig. 1) and unpaired t-test was used (Figs. 3 and 5). Kaplan-Meier analysis and log-rank test were used to perform survival analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

GAS6-AS1 is highly expressed in ccRCC tissues and cell lines

The ccRCC data collected from TCGA (523 tumor tissues and 100 adjacent-normal kidney tissues) were analyzed with R software. The results showed that the expression of GAS6-AS1 in tumor tissues was significantly higher than that in adjacent-normal kidney tissues (Fig. 1A). The results were verified by RT-qPCR, which was used to detect the expression of GAS6-AS1 in ccRCC tissues and matched normal kidney tissues. The results indicated that the expression of GAS6-AS1 in carcinoma tissues was higher than that in adjacent-normal renal tissues (Fig. 1B). Moreover, the expression of GAS6-AS1 in ccRCC cell lines (OSRC-2, Caki-1, SN12-PM6, A498 and SW839) was higher than that in normal renal tubular epithelial cells (HK-2; Fig. 1C). Finally, the OSRC-2 and SW839 cell lines, with the highest relative expression levels of GAS6-AS1, were selected for further experiments. The effect of GAS6-AS1 expression on overall survival was assessed by Kaplan-Meier analysis. The results demonstrated that the prognosis of patients with high GAS6-AS1 expression was significantly lower than that of patients with low expression (Fig. 1D). The results indicated that GAS6-AS1 might be an oncogene in ccRCC.

GAS6-AS1-knockdown inhibits the proliferation, invasiveness and glycolysis of ccRCC cells

In order to investigate the biological functions of GAS6-AS1 in ccRCC, GAS6-AS1-knockdown cells (OSRC-2 and SW839) were constructed using small interfering RNAs. The results indicated that expression of GAS6-AS1 was significantly decreased in the si-GAS6-AS1 groups (Fig. 2A). The results of CCK-8 and colony formation assays showed that compared with the si-NC group, the viability and proliferation of OSRC-2 and SW839 cells were reduced in the si-GAS6-AS1 groups (Fig. 2B and C). Furthermore, the results of Transwell analysis revealed that the invasive capacity of OSRC-2 and SW839 cells transfected with si-GAS6-AS1 was significantly decreased compared with that of si-NC-transfected cells (Fig. 2D). Compared with the control group, the glucose consumption, lactate production and ATP production of OSRC-2 and SW839 cells transfected with si-GAS6-AS1 were significantly decreased (Fig. 2E). In addition, western blotting revealed that si-GAS6-AS1 significantly decreased the expression of PCNA, MMP-2 and HK2 in OSRC-2 and SW839 cells (Fig. 2F). These findings showed that si-GAS6-AS1 inhibited proliferation, invasion and glycolysis in ccRCC cells.

GAS6-AS1 overexpression promotes the proliferation, invasiveness and glycolysis of ccRCC cells

OSRC-2 and SW839 cells overexpressing GAS6-AS1 were constructed, and the expression of GAS6-AS1 was significantly upregulated in the GAS6-AS1 group compared with the negative control vector group (Fig. 3A). The results of CCK-8 analysis indicated that the overexpression of GAS6-AS1 enhanced OSRC-2 and SW839 cell proliferation (Fig. 3B), and colony formation experiments revealed that GAS6-AS1 overexpression increased the number of OSRC-2 and SW839 cell clones (Fig. 3C). The results of the Transwell assay showed that the invasive ability of cells transfected with the GAS6-AS1 expression plasmid was enhanced (Fig. 3D). In addition, the detection of glycolysis-related indicators revealed that GAS6-AS1 overexpression upregulated glucose consumption, lactic acid production and ATP production in OSRC-2 and SW839 cells (Fig. 3E). Furthermore, western blotting demonstrated that the expression of PCNA, MMP-2 and HK2 was increased in cells overexpressing GAS6-AS1 (Fig. 3F). These results suggested that GAS6-AS1 may be involved in the development of ccRCC in an oncogenic manner. These findings showed that GAS6-AS1 promoted proliferation, invasion and glycolysis in ccRCC cells.

GAS6-AS1 regulates the AMPK/mTOR signaling pathway in ccRCC cells

The activation of AMPK can directly or indirectly suppress mTOR activity, inhibiting the phosphorylation of downstream proteins, thus affecting cellular physiological and biochemical processes and inhibiting tumor growth (32). In the current study, the relationship between the AMPK/mTOR signaling pathway and GAS6-AS1 in OSRC-2 and SW839 cells was determined by western blotting. The results showed that GAS6-AS1 silencing increased AMPK phosphorylation while decreasing mTOR phosphorylation in OSRC-2 and SW839 cells (Fig. 4A). Moreover, the expression of downstream proteins of the AMPK/mTOR signaling pathway (PCNA, MMP-2 and HK2) was downregulated in OSRC-2 and SW839 cells transfected with si-GAS6-AS1. However, an AMPK inhibitor (compound C) and mTOR inhibitor (mhy1485) reversed the effect of si-GAS6-AS1 on the expression of AMPK/mTOR pathway proteins in OSRC-2 and SW839 cells (Fig. 4B and C). The data demonstrated that GAS6-AS1 promoted expression of PCNA, MMP-2 and HK2 by modulating AMPK/mTOR pathway.

GAS6-AS1-knockdown inhibits xenografted tumor growth in vivo

A xenograft tumor model was used to determine the effects of GAS6-AS1 on tumor growth in vivo. The results showed that the volume and weight of tumors transfected with an sh-GAS6-AS1 expression plasmid were significantly reduced compared with those of the control group (Fig. 5A and B), and that the expression level of GAS6-AS1 in tumor tissues was decreased in the sh-GAS6-AS1 group (Fig. 5C). Furthermore, western blotting demonstrated that the level of p-AMPK was increased, and the levels of p-mTOR, PCNA, MMP-2 and HK2 were decreased in the sh-GAS6-AS1 group (Fig. 5D). The immunohistochemistry results were consistent with those of the western blot analyses (Fig. 5E). These results suggested that GAS6-AS1-knockdown significantly inhibited the tumor formation ability of ccRCC cells.

Discussion

ccRCC is one of the most common malignant tumors of the urinary system (33), for which targeted therapy may be a promising new treatment option. Corresponding targeted drugs have been developed, such as sunitinib, acetinib, pazopanib and the mTOR inhibitor everolimus (34,35). These compounds can effectively prolong the survival of patients with ccRCC. Therefore, it is necessary to further explore the mechanisms underlying development, in order to identify effective and reliable biomarkers and therapeutic targets for ccRCC.

In previous years, the biological functions of lncRNAs in the development of ccRCC have attracted increasing attention (36). Xiao et al (13) found that a deficiency in lncRNA FILNC1 reduced apoptosis induced by energy deficit, and thus significantly promoted ccRCC progression. The underlying mechanism was to increase the glucose uptake of tumor cells and increase the production of lactic acid by upregulating c-myc. Other studies have found that HOTAIR directly combines with protein Salvador homolog 1 to promote histone H3K27 methylation, resulting in the loss of its function, thus activating Hippo signaling and promoting the proliferation of ccRCC cells (37).

GAS6-AS1 is an antisense RNA downstream of Gas6. Han et al (38) found that the expression level of GAS6-AS1 in non-small cell lung cancer (NSCLC) was significantly lower than that in adjacent-normal tissues, indicating that the deletion of GAS6-AS1 was an important reason for the occurrence and development of NSCLC. However, Zhang et al (39) showed that GAS6-AS1 was upregulated in gastric cancer tissues, promoting cellular proliferation and invasiveness by regulating the GAS6/AXL signaling pathway in vivo and in vitro. GAS6-AS1 was also found to be upregulated in kidney renal papillary cell carcinoma compared with normal tissues, where it was positively correlated with patient prognosis (40). However, the correlation between GAS6-AS1 and ccRCC has not been reported. In the present study, TCGA database was analyzed and the expression of GAS6-AS1 in ccRCC tissues was found to be higher than in adjacent-normal tissues, which was consistent with the results of RT-qPCR. Furthermore, GAS6-AS1 expression was negatively correlated with the prognosis of patients with ccRCC. Further studies showed that GAS6-AS1 regulated the proliferation, invasiveness and glucose metabolism of ccRCC cells, and that GAS6-AS1-knockdown suppressed the growth of xenograft tumors. These results suggested that GAS6-AS1 may act as an oncogene in ccRCC.

PCNA plays an important role in the repair of nuclear damage. The number of PCNA-positive cells detected by monoclonal antibodies against PCNA has been used to predict the possibility of primary tumor recurrence and metastasis (41,42). MMPs are highly conserved zinc-dependent endopeptidases, which can degrade the basement membrane and extracellular matrix components. The upregulation of MMP-2 was found to increase tumor invasion and metastasis (43). HK2 is a key enzyme in glycolysis; the activation of mTORC1 upregulated the expression of HK2 protein, promoting glucose absorption and increasing the level of lactate secreted by cells (44). Hence, in the present study, the expression PCNA, MMP-2 and HK2 was assessed to determine the effects of GAS6-AS1 on proliferation, invasiveness and glycolysis of ccRCC cells. The results demonstrated that si-GAS6-AS1 inhibited the expression of PCNA, MMP-2 and HK2, while GAS6-AS1 increased the expression of these proteins.

mTOR is an integral component of multiple signaling pathways. Activated AMPK regulates mTOR via two pathways, directly phosphorylating Raptor (the direct downstream molecule of AMPK) to attenuate mTORC1 activity, and activating tuberous sclerosis complex 1/2 to inhibit mTORC1 activity (45,46). The inhibition of mTOR activity directly results in disordered glucose and protein synthesis, which subsequently affects the bioavailability of energy and nutrients (such as glucose) in cells, which eventually leads to the inhibition of tumor cell proliferation (47,48). Lung cancer associated lncRNA1 prevented the activation of AMPK, and then promoted glycolysis and restrained autophagy of lung cancer cells through the AMPK/mTOR/S6K axis, so as to promote the rapid proliferation of tumor cells and enhance tumorigenicity (49). miR-138 inhibited the proliferation, invasiveness and migration of lung cancer cells, and reduced the level of mTOR phosphorylation by activating the AMPK signaling pathway (50). In the present study, the effect of GAS6-AS1 on the AMPK/mTOR signaling pathway was assessed. The results indicated that si-GAS6-AS1 increased the level of p-AMPK, reduced the level of p-mTOR, and reduced the expression of PCNA, MMP-2 and HK2. Moreover, a p-AMPK inhibitor (compound C) and an p-mTOR inhibitor (MHY1485) attenuated the effects of si-GAS6-AS1 on the expression of PCNA, MMP-2 and HK2. These findings demonstrated that GAS6-AS1 may be involved in the progression of ccRCC by regulating AMPK/mTOR signaling pathway.

In conclusion, the results of the present study demonstrated that GAS6-AS1 was highly expressed in ccRCC and negatively correlated with the prognosis of patients. GAS6-AS1 influenced the proliferation, invasiveness and glycolysis of ccRCC cells by regulating the AMPK/mTOR signaling pathway, and GAS6-AS1-knockdown inhibited the growth of xenografted tumors. These findings suggest that GAS6-AS1 functions as an oncogene in ccRCC and may act as a molecular marker for the diagnosis and targeted treatment of patients ccRCC.

Supplementary Material

Supporting Data

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 upon reasonable request.

Authors' contributions

XG and RL designed experiments. XG, HL and MZ performed experiments and collected data. RL analyzed data and drafted manuscript. XG and RL confirmed the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Second Hospital of Tianjin Medical University ethics committee [approval no. SYXK (JIN) 2019-0004], and patient consent was obtained prior to surgery.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Vermassen T, De Meulenaere A, Van de Walle M and Rottey S: Therapeutic approaches in clear cell and non-clear cell renal cell carcinoma. Acta Clin Belg. 72:12–18. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Chandrasekaran D, Sundaram S, N K and R P: Programmed death ligand 1; An Immunotarget for renal cell carcinoma. Asian Pac J Cancer Prev. 20:2951–2957. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Barata PC and Rini BI: Treatment of renal cell carcinoma: Current status and future directions. CA Cancer J Clin. 67:507–524. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Quinn JJ and Chang HY: Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 17:47–62. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Peng WX, Koirala P and Mo YY: LncRNA-mediated regulation of cell signaling in cancer. Oncogene. 36:5661–5667. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Liu M, Zhang H, Li Y, Wang R, Li Y, Zhang H, Ren D, Liu H, Kang C and Chen J: HOTAIR, a long noncoding RNA, is a marker of abnormal cell cycle regulation in lung cancer. Cancer Sci. 109:2717–2733. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, Byron M, Monks B, Henry-Bezy M, Lawrence JB, et al: A long noncoding RNA mediates both activation and repression of immune response genes. Science. 341:789–792. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Yan P, Luo S, Lu JY and Shen X: Cis- and trans-acting lncRNAs in pluripotency and reprogramming. Curr Opin Genet Dev. 46:170–178. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Yao F, Wang Q and Wu Q: The prognostic value and mechanisms of lncRNA UCA1 in human cancer. Cancer Manag Res. 11:7685–7696. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Liu N, Liu Z, Liu X and Chen H: Comprehensive analysis of a competing endogenous RNA network identifies Seven-lncRNA signature as a prognostic biomarker for melanoma. Front Oncol. 9:9352019. View Article : Google Scholar : PubMed/NCBI

11 

Chao Y and Zhou D: lncRNA-D16366 is a potential biomarker for diagnosis and prognosis of hepatocellular carcinoma. Med Sci Monit. 25:6581–6586. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, Liu Y, Zhang N and Yang Q: LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. J Cell Physiol. 234:9105–9117. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Xiao ZD, Han L, Lee H, Zhuang L, Zhang Y, Baddour J, Nagrath D, Wood CG, Gu J, Wu X, et al: Energy stress-induced lncRNA FILNC1 represses c-Myc-mediated energy metabolism and inhibits renal tumor development. Nat Commun. 8:7832017. View Article : Google Scholar : PubMed/NCBI

14 

Hong Q, Li O, Zheng W, Xiao WZ, Zhang L, Wu D, Cai GY, He JC and Chen XM: LncRNA HOTAIR regulates HIF-1α/AXL signaling through inhibition of miR-217 in renal cell carcinoma. Cell Death Dis. 8:e27722017. View Article : Google Scholar : PubMed/NCBI

15 

Huang JL, Liao Y, Qiu MX, Li J and An Y: Long non-coding RNA CCAT2 promotes cell proliferation and invasion through regulating Wnt/β-catenin signaling pathway in clear cell renal cell carcinoma. Tumour Biol. 39:10104283177113142017. View Article : Google Scholar : PubMed/NCBI

16 

Li JK, Chen C, Liu JY, Shi JZ, Liu SP, Liu B, Wu DS, Fang ZY, Bao Y, Jiang MM, et al: Long noncoding RNA MRCCAT1 promotes metastasis of clear cell renal cell carcinoma via inhibiting NPR3 and activating p38-MAPK signaling. Mol Cancer. 16:1112017. View Article : Google Scholar : PubMed/NCBI

17 

Zhou J, Zhang S, Chen Z, He Z, Xu Y and Li Z: CircRNA-ENO1 promoted glycolysis and tumor progression in lung adenocarcinoma through upregulating its host gene ENO1. Cell Death Dis. 10:8852019. View Article : Google Scholar : PubMed/NCBI

18 

Ai J, Sun J, Zhou G, Zhu T and Jing L: Long non-coding RNA GAS6-AS1 acts as a ceRNA for microRNA-585, thereby increasing EIF5A2 expression and facilitating hepatocellular carcinoma oncogenicity. Cell Cycle. 19:742–757. 2020. View Article : Google Scholar : PubMed/NCBI

19 

Ke R, Xu Q, Li C, Luo L and Huang D: Mechanisms of AMPK in the maintenance of ATP balance during energy metabolism. Cell Biol Int. 42:384–392. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Gupta A, Anjomani-Virmouni S, Koundouros N, Dimitriadi M, Choo-Wing R, Valle A, Zheng Y, Chiu YH, Agnihotri S, Zadeh G, et al: PARK2 depletion connects energy and oxidative stress to PI3K/Akt activation via PTEN S-Nitrosylation. Mol Cell. 65:999–1013.e7. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Kosztelnik M, Kurucz A, Papp D, Jones E, Sigmond T, Barna J, Traka MH, Lorincz T, Szarka A, Banhegyi G, et al: Suppression of AMPK/aak-2 by NRF2/SKN-1 down-regulates autophagy during prolonged oxidative stress. FASEB J. 33:2372–2387. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Liu M, Zhang Z, Wang H, Chen X and Jin C: Activation of AMPK by metformin promotes renal cancer cell proliferation under glucose deprivation through its interaction with PKM2. Int J Biol Sci. 15:617–627. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Kim HI, Hong SH, Ku JM, Lim YS, Lee SJ, Song J, Kim TY, Cheon C and Ko SG: Scutellaria radix promotes apoptosis in non-small cell lung cancer cells via induction of AMPK-dependent autophagy. Am J Chin Med. 47:691–705. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Hart PC, Mao M, de Abreu AL, Ansenberger-Fricano K, Ekoue DN, Ganini D, Kajdacsy-Balla A, Diamond AM, Minshall RD, Consolaro ME, et al: MnSOD upregulation sustains the Warburg effect via mitochondrial ROS and AMPK-dependent signalling in cancer. Nat Commun. 6:60532015. View Article : Google Scholar : PubMed/NCBI

25 

Bordoloi J, Ozah D, Bora T, Kalita J and Manna P: Gamma-glutamyl carboxylated Gas6 mediates the beneficial effect of vitamin K on lowering hyperlipidemia via regulating the AMPK/SREBP1/PPARα signaling cascade of lipid metabolism. J Nutr Biochem. 70:174–184. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Son BK, Akishita M, Iijima K, Kozaki K, Maemura K, Eto M and Ouchi Y: Adiponectin antagonizes stimulatory effect of tumor necrosis factor-alpha on vascular smooth muscle cell calcification: Regulation of growth arrest-specific gene 6-mediated survival pathway by adenosine 5′-monophosphate-activated protein kinase. Endocrinology. 149:1646–1653. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Gao L, Li J, He J, Liang L, He Z, Yue C, Jin X, Luo G and Zhou Y: CD90 affects the biological behavior and energy metabolism level of gastric cancer cells by targeting the PI3K/AKT/HIF-1α signaling pathway. Oncol Lett. 21:1912021. View Article : Google Scholar : PubMed/NCBI

29 

Chen JF, Wu P, Xia R, Yang J, Huo XY, Gu DY, Tang CJ, De W and Yang F: STAT3-induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal-mediated inhibition of autophagy. Mol Cancer. 17:62018. View Article : Google Scholar : PubMed/NCBI

30 

Luo K, Geng J, Zhang Q, Xu Y, Zhou X, Huang Z, Shi KQ, Pan C and Wu J: LncRNA CASC9 interacts with CPSF3 to regulate TGF-β signaling in colorectal cancer. J Exp Clin Cancer Res. 38:2492019. View Article : Google Scholar : PubMed/NCBI

31 

Yang B, Zhang L, Cao Y, Chen S, Cao J, Wu D, Chen J, Xiong H, Pan Z, Qiu F, et al: Overexpression of lncRNA IGFBP4-1 reprograms energy metabolism to promote lung cancer progression. Mol Cancer. 16:1542017. View Article : Google Scholar : PubMed/NCBI

32 

Wang Z, Wang N, Liu P and Xie X: AMPK and Cancer. Exp Suppl. 107:203–226. 2016.PubMed/NCBI

33 

Chen Z, Xiao K, Chen S, Huang Z, Ye Y and Chen T: Circular RNA hsa_circ_001895 serves as a sponge of microRNA-296-5p to promote clear cell renal cell carcinoma progression by regulating SOX12. Cancer Sci. 111:713–726. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Chowdhury S, Matrana MR, Tsang C, Atkinson B, Choueiri TK and Tannir NM: Systemic therapy for metastatic non-clear-cell renal cell carcinoma: Recent progress and future directions. Hematol Oncol Clin North Ama. 25:853–869. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Hakimi AA, Voss MH, Kuo F, Sanchez A, Liu M, Nixon BG, Vuong L, Ostrovnaya I, Chen YB, Reuter V, et al: Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer: Data from a randomized Phase III trial. Cancer Discov. 9:510–525. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Qu L, Wang ZL, Chen Q, Li YM, He HW, Hsieh JJ, Xue S, Wu ZJ, Liu B, Tang H, et al: Prognostic value of a long non-coding RNA signature in localized clear cell renal cell carcinoma. Eur Urol. 74:756–763. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Jiang LP, Zhu ZT, Zhang Y and He CY: Downregulation of MicroRNA-330 correlates with the radiation sensitivity and prognosis of patients with brain metastasis from lung cancer. Cell Physiol Biochem. 42:2220–2229. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Han L, Kong R, Yin DD, Zhang EB, Xu TP, De W and Shu YQ: Low expression of long noncoding RNA GAS6-AS1 predicts a poor prognosis in patients with NSCLC. Med Oncol. 30:6942013. View Article : Google Scholar : PubMed/NCBI

39 

Zhang P, Dong Q, Zhu H, Li S, Shi L and Chen X: Long non-coding antisense RNA GAS6-AS1 supports gastric cancer progression via increasing GAS6 expression. Gene. 696:1–9. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Yang F, Song Y, Ge L, Zhao G, Liu C and Ma L: Long non-coding RNAs as prognostic biomarkers in papillary renal cell carcinoma. Oncol Lett. 18:3691–3697. 2019.PubMed/NCBI

41 

Boehm EM, Gildenberg MS and Washington MT: The many roles of PCNA in eukaryotic DNA replication. Enzymes. 39:231–254. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Cardano M, Tribioli C and Prosperi E: Targeting proliferating cell nuclear antigen (PCNA) as an effective strategy to inhibit tumor cell proliferation. Curr Cancer Drug Targets. 20:240–252. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Shamsara J: Identification of Non-Zinc binding inhibitors of MMP-2 through virtual screening and subsequent rescoring. Drug Res (Stuttg). 68:529–535. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Wu H, Pan L, Gao C, Xu H, Li Y, Zhang L, Ma L, Meng L, Sun X and Qin H: Quercetin inhibits the proliferation of glycolysis-addicted HCC cells by reducing hexokinase 2 and Akt-mTOR pathway. Molecules. 24:19932019. View Article : Google Scholar : PubMed/NCBI

45 

Alers S, Löffler AS, Wesselborg S and Stork B: Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: Cross talk, shortcuts, and feedbacks. Mol Cell Biol. 32:2–11. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Ren P, Ren X, Cheng L and Xu L: Frankincense, pine needle and geranium essential oils suppress tumor progression through the regulation of the AMPK/mTOR pathway in breast cancer. Oncol Rep. 39:129–137. 2018.PubMed/NCBI

47 

Mossmann D, Park S and Hall MN: mTOR signalling and cellular metabolism are mutual determinants in cancer. Nature reviews. Nat Rev Cancer. 18:744–757. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Han J, Zhang L, Guo H, Wysham WZ, Roque DR, Willson AK, Sheng X, Zhou C and Bae-Jump VL: Glucose promotes cell proliferation, glucose uptake and invasion in endometrial cancer cells via AMPK/mTOR/S6 and MAPK signaling. Gynecol Oncol. 138:668–675. 2015. View Article : Google Scholar : PubMed/NCBI

49 

Li JY and Luo ZQ: LCAL1 enhances lung cancer survival via inhibiting AMPK-related antitumor functions. Mol Cell Biochem. 457:11–20. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Ye Z, Fang B, Pan J, Zhang N, Huang J, Xie C, Lou T and Cao Z: miR-138 suppresses the proliferation, metastasis and autophagy of non-small cell lung cancer by targeting Sirt1. Oncol Rep. 37:3244–3252. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2021
Volume 22 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo X, Li H, Zhang M and Li R: LncRNA GAS6 antisense RNA 1 facilitates the tumorigenesis of clear cell renal cell carcinoma by regulating the AMP‑activated protein kinase/mTOR signaling pathway Retraction in /10.3892/ol.2024.14259. Oncol Lett 22: 727, 2021
APA
Guo, X., Li, H., Zhang, M., & Li, R. (2021). LncRNA GAS6 antisense RNA 1 facilitates the tumorigenesis of clear cell renal cell carcinoma by regulating the AMP‑activated protein kinase/mTOR signaling pathway Retraction in /10.3892/ol.2024.14259. Oncology Letters, 22, 727. https://doi.org/10.3892/ol.2021.12988
MLA
Guo, X., Li, H., Zhang, M., Li, R."LncRNA GAS6 antisense RNA 1 facilitates the tumorigenesis of clear cell renal cell carcinoma by regulating the AMP‑activated protein kinase/mTOR signaling pathway Retraction in /10.3892/ol.2024.14259". Oncology Letters 22.4 (2021): 727.
Chicago
Guo, X., Li, H., Zhang, M., Li, R."LncRNA GAS6 antisense RNA 1 facilitates the tumorigenesis of clear cell renal cell carcinoma by regulating the AMP‑activated protein kinase/mTOR signaling pathway Retraction in /10.3892/ol.2024.14259". Oncology Letters 22, no. 4 (2021): 727. https://doi.org/10.3892/ol.2021.12988