- Research
- Open access
- Published:
The PVT1-214/miR-671-5p/SLC45A4 signaling axis regulates cell proliferation in human gastric cancer
World Journal of Surgical Oncology volume 23, Article number: 158 (2025)
Abstract
Background
Long non-coding RNA PVT1 (lncRNA PVT1) serves as a carcinogenic regulatory factor in several cancers; however, the expression and function of its transcriptional isomer PVT1-214 in gastric cancer (GC) are poorly understood.
Methods
PVT1-214 and miR-671-5p levels in GC cells and tissues were analyzed through quantitative real-time polymerase chain reaction (qRT-PCR). Western blotting (WB) was used to detect Homo sapiens solute carrier family 45 member 4 (SLC45A4) expression in GC cells. Thereafter, the relationship between PVT1-214 and miR-671-5p was evaluated through dual-luciferase reporter assays and RNA immunoprecipitation (RIP) analysis. Additionally, the biological activities of PVT1-214, miR-671-5p and SLC45A4 in GC cells were analyzed through Cell Counting Kit-8 (CCK-8), 5-ethyl-20-deoxyuridine (EdU), colony formation and Transwell assays. The effect of PVT1-214 on GC was studied in vivo via a nude mouse tumor xenograft model.
Results
PVT1-214 overexpression in GC cells and tissues was positively related to tumor size, malignancy grade, lymphatic metastasis and clinical stage. PVT1-214 knockdown suppressed cell growth, invasion and migration in vitro, whereas PVT1-214 overexpression promoted tumor proliferation in vivo. In addition, PVT1-214 positively regulates SLC45A4 expression through its competitive endogenous RNA (ceRNA) activity against miR-671-5p. SLC45A4 expression was positively related to PVT1-214 expression. PVT1-214 competes with endogenous RNA (ceRNA) by binding to miR-671-5p. When miR-671-5p is inhibited, SLC45A4 is released from the complementary binding complex, thereby increasing SLC45A4 protein levels in GC cells.
Conclusions
The present work revealed the candidate ceRNA regulatory pathway by which PVT1-214 regulates SLC45A4 expression in GC cells, which is achieved through competitive binding to endogenous miR-671-5p. The results of this study can shed novel light on new molecular targets for treating GC.
Background
Gastric cancer (GC), which has high morbidity and mortality rates globally, is a public health problem with different epidemiological characteristics across different regions and ethnic groups [1]. GC ranks fifth among cancers in terms of morbidity and fourth among the factors associated with cancer-mortality [2]. Owing to its atypical early symptoms, the diagnosis is usually made at an advanced stage, and chemotherapy resistance is often the main clinical problem [3]. Therefore, new research and strategies may improve advanced GC treatment. The implementation of more rational, comprehensive and personalized treatment plans is the key to enabling maximum clinical benefits for patients [4] with gastric cancer.
Long non-coding RNAs (lncRNAs), noncoding RNAs (ncRNAs) over 200 nucleotides in length, cannot encode proteins but are involved in various biological processes. LncRNAs are essential for modulating disease progression. For example, LINC01207 is upregulated in gastric cancer tissues and promotes disease progression by regulating the miR-671-5p/DDX5 axis [5], the lncRNA GATA2AS acts as a regulator by affecting chromatin regulatory patterns and binding of erythroid transcription factors [6], and the lncRNA KCNQ1OT1 can inhibit the expression of Tcf7 and induce myocardial hypertrophy [7]. LncRNAs are also crucial in a variety of tumors. The lncRNA SNHG1 promotes cisplatin resistance and tumor progression in breast cancer [8], aberrant LINC00173.v1 upregulation serves as a risk factor for lung squamous cell carcinoma [9], while lncRNA HNF1A-AS1 downregulation can inhibit GC metastasis [10]. Plasmacytoma variant translocation 1 (PVT1) is a long non-coding RNA locus located adjacent to the c-myc locus on human chromosome 8q24.21 and is considered to be a candidate oncogene [11].
PVT1 has been shown to be important in the progression of several types of cancer, such as stomach cancer [12], liver cancer [13], pancreatic cancer [14], and so on. As a transcript of PVT1, the function of PVT1-214 is rarely reported. PVT1-214 can accelerate colorectal cancer proliferation and metastasis, suggesting its carcinogenic role in tumor biology [15]. Nonetheless, the exact mechanism underlying GC remains largely unknown.
MicroRNAs (miRNAs), ncRNAs of 18–25 nucleotides long, can regulate different biological processes at the post-transcriptional level, such as signaling pathways, immunity, and inflammation [16]. According to Šustr F et al., elevated plasma hsa-miR-206 expression predicts the recurrence of atrial fibrillation (AF) [17]. miR-221-3p can inhibit inflammatory responses in diabetic patients while promoting skin wound healing [18]. The levels of miR-92a-3p in extracapsular vesicles within the brain tissue of patients with major depressive disorder are significantly decreased [19]. In oncology, the plasma miR-126 levels are a candidate predictor of the efficacy of chemotherapeutic immunotherapy or first-line immunotherapy among advanced non-small cell lung cancer (NSCLC) patients [20]. miR-149-5p overexpression markedly accelerates the apoptosis of colorectal cancer cells, whereas miR-149-5p knockdown significantly suppresses their apoptosis [21]. MiR-133 A-1 can affect glutamine transport in gastric cancer by regulating SLC1A4 expression, and further regulate the occurrence and development of gastric cancer [22]. MiR-671-5p has crucial effects not only on regulating inflammatory response-mediated cell damage but also on modulating the occurrence and development of tumors. Therefore, targeting the miR-6715p/MFAP3L signaling pathway is a candidate anti-NSCLC strategy [23]. In breast cancer, miR-671-5p expression is related to oncogenic transformation and radiochemoresistance [24]. This study aims to explore the effects of miR-671-5p and PVT1-214 on gastric cancer cells, and assuming that lncRNA PVT1-214 can regulate the proliferation, invasion and migration of gastric cancer cells through modulating the miR-671-5p.
Materials and methods
Clinical tissue sampling
In this study, GC tissues were obtained from GC patients from Lu'an Hospital of Anhui Medical University from May 2018 to May 2019. Forty-three GC tissue samples were randomly selected for the present work. Control samples were taken from one patient’s paracancerous tissue (≥ 5 cm away from the surgical margin), in which no cancer cells could be detected by pathological analysis. After removal, the samples were immersed in liquid nitrogen at -196 °C. Informed consent was obtained from each patient. The Ethics Review Committee of Anhui Medical University Lu’an Hospital approved the present work. The study protocols were conducted in accordance with the tenets of the Declaration of Helsinki.
Cells and culture
SGC7901, MKN45, BGC-823 and AGS GC cells and healthy human gastric epithelial GES-1 cells were obtained from the Central Laboratory of the First Affiliated Hospital of Anhui Medical University. The cells were maintained in RPMI-1640 medium (Thermo Fisher Scientific, Cambridge, MA, USA) containing 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Cambridge, MA, USA) and 1% penicillin-streptomycin (Invitrogen, Carlsbad, CA, USA) at 37 °C with 5% CO2.
Cell transfection
The LINC PVT1-214 overexpression plasmid, LINC PVT1-214-siRNA (si-LINC PVT1-214), miR-671-5p mimics, miR-671-5p inhibitors and the respective controls were prepared by GenePharma (Shanghai, China). Lipofectamine® 3000 (Invitrogen, Carlsbad, CA, USA) was used for cell transfection following specific protocols.
Quantitative real-time polymerase chain reaction (qRT–PCR)
TRIzol reagent (Thermo Fisher Scientific, Shanghai, China) was used to obtain total cellular and tissue RNA, which was then reverse transcribed into cDNA using cDNA Synthesis SuperMix and EasyScript One-Step gDNA Removal (TransGen Biotech, China) according to specific protocols. The qRT-PCR mixture was pre-mixed in a 22331 Hamburg instrument (Eppendorf, Germany) and analysed using an ABI 7500 real-time fluorescence quantitative PCR system (LightCycler 96, Roche, Switzerland). We chose GAPDH and U6 as endogenous references for LINC PVT1-214, miR-671-5p and SLC45A4 and analysed gene expression using the 2-ΔΔCt approach. Table 1 shows the sequences of the primers used.
Western blotting
RIPA lysis buffer containing 1% PMSF was used to lyse the cells. The sample was then centrifuged at high speed to collect the supernatant. Protein content was quantified by the Bradford method. After 10 min heating in a 100 °C water bath, protein separation was achieved by SDS-PAGE before transfer to polyvinylidene fluoride (PVDF) membranes. The membranes were then immersed in 5% defatted milk at room temperature for 1 h. Anti-SLC45A4 antibody (YN3368) (1:1000; ImmunoWay, America) and anti-GAPDH antibody (K106389P) (1:1000; Solarbio, China) were added to the membranes and incubated overnight at 4 °C. After washing with TBST solution, the membranes were probed with secondary antibodies (goat anti-rabbit Beyotime, Shanghai, China) for a further 1 h at room temperature and rinsed three times. Finally, hypersensitive ECL (Biossci Biotechnology Co., Ltd., Wuhan, China) was used for colour development.
Cell counting Kit-8 (CCK-8) assay
The logarithmic AGS and SGC7901 cells were prepared as single cell suspensions using 0.25% trypsin. After counting, the cells were seeded in 96-well plates (2000/well) and incubated in 5% CO2 at 37 °C for 0, 12, 24, 36, 48–60 h. CCK-8 solution (10 µL) was then added to each well (Beyotime, Shanghai, China) to culture the cells for a further 1 h. The absorbance of each well was then detected at 450 nm using a microplate reader.
Transwell analyses
We performed transwell assays to analyse the invasion and migration of GC cells. For the invasion assay, we inoculated 2×104 cells/well (FBS-free) into the upper Transwell chamber (Corning, NY, USA), which was covered by an 8 μm membrane aperture, and poured RPMI-1640 medium containing 10% FBS into the lower chamber. At 24 h post-culture, non-migrated cells were removed with a cotton swab, and cells penetrating the membrane were immobilised with 4% paraformaldehyde and later stained with 0.1% crystal violet before imaging, drying and counting. A Matrigel (BD, San Jose, CA, USA) coated Transwell membrane was used for the invasion assay. A total of 2 × 104 cells/well (FBS-free) were then inoculated into the upper Transwell chamber (Corning, NY, USA) with a membrane aperture of 8 μm, and RPMI-1640 medium containing 10% FBS was poured into the lower cavity. After 24 h of culture, non-migrated cells were removed with a cotton swab. After fixation with 4% paraformaldehyde, membrane-penetrating cells were stained with 0.1% crystal violet, imaged, dried and counted. For the migration assay, the Matrigel membrane was removed and the remaining procedures were identical to those used for the invasion assay.
Luciferase reporter assay
This study carried out a luciferase reporter gene assay to verify the targeting relationship of PVT1-214 with miR-671-5p. Briefly, SGC7901 cells (5 × 105/well) were inoculated in 24-well plates, followed by cloning and insertion of luciferin reporter vectors that contained mutant (Mut) or wild-type (WT) PVT1-214 sequences in psiCHECK2 vectors (GenePharma, China). These vectors were then cotransfected with miR-671-5p mimics or negative controls in the above cells. After 48 h of transfection, we used a dual-luciferase reporter gene assay system (PerkinElmer, USA) to measure luciferase activity in accordance with specific protocols.
RNA immunoprecipitation (RIP) assay
To determine the relationship between PVT1-214 and miR-671-5p, we conducted a RIP assay with an EZMagna RIP kit (Millipore, Billerica, MA, USA) following specific protocols. Briefly, RIP cleavage buffer containing protease/RNase inhibitors was added to lyse SGC7901 cells at the logarithmic growth stage, followed by 1 h of incubation of the lysates with an anti-Ago2 antibody or mouse IgG-labeled magnetic beads at ambient temperature. Following protein removal with protease K, qRT‒PCR was conducted on the extracted immunoprecipitated RNA.
Colony formation assay
First, 1×104 cells from each group were inoculated into a 35-mm culture dish. After reaching 80% confluence, cell transfection was performed, and the experimental groups were set as follows: siRNA-NC, siRNA-PVT1-214, PCDNA3.1-no-load, and pCDNA3.1-PVT1-214. Cell digestion was completed after 48 h, followed by inoculation (1 × 103 cells/well) into a new 24-well plate, and 3 replicate wells were used for each group. After the culture medium was discarded, cells were fixed with 4% paraformaldehyde for 15 min, stained with 0.1% crystal violet for 10 min, imaged and colony counted.
5-Ethynyl-2′-deoxyuridine (EdU) assay
Cells were seeded in 96-well plates for the EdU assay, followed by fixation, permeabilization and incubation with 50 µM EdU (Sigma) for 3 h then 1 µg/mL (DAPI) (Sigma) for 10 min. The cells were observed and photographed using a fluorescence microscope (Leica, Germany).
Mouse experiments
Animal experimentation was approved by the Ethics Review Committee of the First Affiliated Hospital of Anhui Medical University. The animal protocols followed the UK Cancer Research Coordinating Committee (UKCCCR) guidelines for animal welfare in experimental oncology. Fourteen 6-week-old female SPF BALB/c nude mice were randomised to the NC or PVT1-214 groups. Mice in the NC group were inoculated subcutaneously with SGC7901 cell suspensions (100 µL, containing approximately 2 × 106 cells), and those in the PVT1-214 group were inoculated with SGC7901 cells overexpressing PVT1-214. The longest and shortest tumour diameters were then measured once a day with a caliper until the tumours were removed 2 weeks later. Tumour weight and volume were calculated as follows: volume = (length × width 2 × 0.5).
Statistical analysis
SPSS 17.0 (SPSS Inc., Chicago, IL, USA) was used for the statistical analyses. The measurement results are presented as the means ± standard deviations (x ± s). One-way ANOVA or a t test was used to analyze between-group differences. P < 0.05 indicated statistical significance.
Results
PVT1-214 is upregulated in GC tissues and cells
To confirm the effect of PVT1-214 on GC, 43 GC and 43 paired non-cancerous tissues were analysed. As a result, PVT1-214 expression was significantly upregulated in the GC samples compared to the non-cancer samples (P < 0.001; Fig. 1a). We then investigated whether PVT1-214 expression was sensitive and specific in discriminating between GC and non-cancerous samples using receiver operating characteristic (ROC) curve analysis. PVT1-214 showed significant predictive performance and the area under the curve was 0.765 [95% CI, 0.665–0.866; P < 0.001; Fig. 1b]. Kaplan-Meier analysis showed that tumour-related PVT1-214 expression was positively associated with overall survival (P = 0.0274; Fig. 1c). We examined the expression of PVT1-214 in GC cells (AGS, BGC-823, SGC7901 and MKN45 cells) and healthy gastric mucosa GES-1 cells. Compared to GES-1 cells, GC cells showed significantly increased PVT1-214 expression (Fig. 1d). Taken together, these results demonstrate that PVT1-214 is upregulated in GC.
PVT1-214 is upregulated in human GC tissues and is related to a dismal prognostic outcome. (a) LncRNA PVT1-214 expression in GC and paired noncarcinoma tissue samples detected through qRT‒PCR. (b) ROC curve analysis was conducted to analyze how PVT1-214 expression affects CRC prediction. (c) Kaplan‒Meier analysis was carried out to analyze the relationship of GC patient survival with PVT1-214 expression. (d) PVT1-214 expression in GC cells (AGS, BGC-823, SGC-7901 and MKN-45 cells) and healthy gastric mucosal GES-1 cells. *, ** and *** P < 0.05, P < 0.01 and P< 0.001, respectively
To understand the ability of PVT1-214 expression to predict GC, the relationships between PVT1-214 expression and clinical features in GC patients were investigated. On the basis of the median PVT1-214 expression, we classified the 43 patients into high (n = 22) or low PVT1-214 expression groups (n = 21). Our results revealed that high PVT1-214 expression in GC tissue samples was significantly related to T stage (P = 0.019), tumor size (P = 0.003), and lymph node metastasis (P = 0.009) but not to age, differentiation status or gender (Table 2).
PVT1-214 regulates GC cell growth, invasion and migration
To confirm the biological role of PVT1-214 in GC progression, this study constructed PVT1-214 overexpression and knockdown models using SGC7901 and AGS cells separately (Fig. 2a). As suggested by the results of the CCK-8 assay, compared with NC transfection, PVT1-214 siRNA transfection apparently decreased GC cell viability; moreover, PVT1-214 overexpression dramatically promoted GC cell proliferation relative to that in the NC group (Fig. 2b). To elucidate how PVT1-214 affects the metastasis of GC cells, a transwell assay was conducted. As a result, PVT1-214 knockdown decreased GC cell invasion and migration, whereas PVT1-214 overexpression had the opposite effects (Fig. 2c). With respect to the enhanced cell proliferation phenotype, EdU immunofluorescence staining verified that the proportions of EdU-positive AGS-PVT1-214 and SGC7901-PVT1-214 cells (79.2% and 62.2%, respectively) were greater than those of control cells (57.1% and 47.6%, respectively; Fig. 2d), indicating new DNA synthesis. In addition, according to the results of the colony formation assay, PVT1-214 knockdown suppressed colony formation in GC cells, whereas PVT1-214 overexpression promoted colony formation (Fig. 2e).
PVT1-214 Promotes GC cell growth, invasion, migration, and colony formation. (a) SGC7901 and AGS cells were used to construct the PVT1-214 overexpression and knockdown models separately, and successful construction was validated via qRT‒PCR. (b) A CCK-8 assay was conducted to analyze cell proliferation, compared with NC transfection, PVT1-214 siRNA transfection apparently decreased GC cell viability; PVT1-214 overexpression promoted GC cell proliferation relative to that in the NC group. (c) AGS and SGC7901 GC cell invasion and migration were measured through Transwell assays. (d-e) GC cell proliferation was detected through EdU (d) and colony formation (e) assays. *P < 0.05, **P < 0.01, ***P < 0.001, respectively
Overexpression of the PVT1-214 gene promotes GC proliferation in vivo
To verify the ability of PVT1-214 to accelerate GC proliferation in vivo, this study constructed a nude mouse model of subcutaneous xenograft tumors. PVT1-214-overexpressing xenografts presented an increased growth rate compared with xenograft tumors, accompanied by markedly increased tumor size, as evidenced by increased average tumor volume and weight (Fig. 3a–d). Additionally, the overexpression of PVT1-214 decreased miR-671-5p expression within tumor tissues (Fig. 3e).
MiR-671-5p overexpression suppresses GC cell growth and invasion
To explore how miR-671-5p affects GC cells, we constructed overexpression and knockdown models (Fig. 4a). According to our results, miR-671-5p inhibitors promoted AGS and SGC7901 cell proliferation, but miR-671-5p mimic treatment suppressed their proliferation (Fig. 4b). As expected, the miR-671-5p inhibitor promoted GC cell invasion and migration, whereas the miR-671-5p mimic suppressed invasion and migration (Fig. 4c). EdU staining (Fig. 4d) and colony formation assays (Fig. 4e) yielded results similar to those obtained by the CCK-8 assay. Taken together, these findings indicate that suppressing miR-671-5p expression promoted GC cell proliferation in vitro.
MiR-671-5p inhibits GC cell growth, invasion, migration, and colony Fformation. (a) SGC7901 and AGS cells were used to construct the miR-671-5p overexpression and knockdown models, respectively, and the results were confirmed through qRT‒PCR. (b) GC cell growth was analyzed via a CCK-8 assay. (c) Transwell assays were carried out to detect AGS and SGC7901 cell invasion and migration. (d-e) GC cell proliferation was detected through EdU (d) and colony formation (e) assays. *P < 0.05, **P < 0.01, ***P < 0.001
PVT1-214 adversely modulates miR-671-5p expression
To explore the mechanism underlying PVT1-214, the BiBiServ database (https://bibiserv.cebitec.uni-bielefeld.de) was used to predict the downstream targets of PVT1-214. As a result, miR-671-5p was among the candidate targets (Fig. 5a). Next, qRT-PCR was performed to measure miR-671-5p expression in GC and matched non-cancerous samples. As a result, miR-671-5p expression appeared to decrease in GC tissues (Fig. 5b). PVT1-214 expression was negatively associated with miR-671-5p levels in 43 of the aforementioned GC tissues (Fig. 5c). In addition, RIP experiments revealed that in SGC7901 cells, PVT1-214 and miR-671-5p expression were significantly increased in the anti-Ago2 group compared to the anti-IgG group (Fig. 5d). As suggested by the qRT-PCR results, PVT1-214 overexpression inhibited miR-671-5p, whereas PVT1-214 knockdown increased miR-671-5p expression in GC cells (Fig. 5e). To clarify the relationship between PVT1-214 and miR-671-5p, a luciferase activity assay was performed in this study. The miR-671-5p mimic dramatically suppressed the luciferase activity of the wild-type PVT1-214 sequence, whereas PVT1-214 overexpression remarkably promoted PVT1-214 luciferase activity. Furthermore, the mutant PVT1-214 sequence was not significantly affected (Fig. 5f). Based on these results, miR-671-5p was identified as a target of PVT1-214.
PVT1-214 Targets miR-671-5p. (a) The PVT1-214 downstream target was estimated on the basis of the BiBiServ database. The binding site in PVT1-214 for miR-671-5p is displayed. (b) miR-671-5p levels in GC and paired noncarcinoma tissues were detected through qRT‒PCR. (c) The relationship between PVT1-214 and miR-671-5p expression in GC tissues was examined through Pearson’s correlation analysis. (d) RIP assays validated the targeting relationship of PVT1-214 with miR-671-5p. (e) qRT‒PCR was performed to measure miR-671-5p expression following the upregulation or downregulation of PVT1-214 expression in GC cells. (f) Luciferase activity assays verified the relationship of PVT1-214 with miR-671-5p. ** P < 0.01, ***P < 0.001. ns indicates not significant
MiR-671-5p negatively regulates SLC45A4 expression
The BiBiServ database (https://bibiserv.cebitec.uni-bielefeld.de) was used to predict the downstream targets of miR-671-5p, as a result, SLC45A4 was among the candidate targets. According to the results of the Transwell assays, SLC45A4 knockdown suppressed GC cell invasion and migration, whereas SLC45A4 overexpression had the opposite effect (Fig. 6a). Additionally, according to the results of the EdU assay, SLC45A4 overexpression enhanced AGS cell growth, whereas SLC45A4 knockdown suppressed SGC7901 cell growth (Fig. 6b). To investigate the regulatory effects of PVT1-214 and miR-671-5p on SLC45A4 at the mRNA level, alterations in SLC45A4 mRNA and protein expression after PVT1-214 overexpression or knockdown and after miR-671-5p mimic or inhibitor treatment were detected through qRT‒PCR and Western blotting. PVT1-214 overexpression increased SLC45A4 mRNA and protein levels in GC cells, whereas PVT1-214 knockdown had the opposite effect (Fig. 6c). Our results also revealed that miR-671-5p expression was negatively related to SLC45A4 within GC tissues (Fig. 6d). Therefore, SLC45A4 expression is negatively modulated via miR-671-5p but positively modulated via PVT1-214.
PVT1-214 promotes SLC45A4 expression, whereas miR-671-5p represses SLC45A4 expression. (a) The miR-671-5p downstream target was estimated on the basis of the BiBiServ database. The binding site in SLC45A4 for miR-671-5p is displayed. (b) SGC7901 cells was used to construct the SLC45A4 knockdown models, and successful construction was validated via qRT‒PCR. (c) Transwell assays were implemented to detect AGS and SGC7901 cell invasion and migration. (d) GC cell proliferation was detected through EdU. (e) qRT‒PCR and Western blotting were implemented to detect SLC45A4 expression after PVT1-214 overexpression or knockdown in GC cells. (f) qRT‒PCR and western blotting were carried out to detect SLC45A4 expression following miR-671-5p overexpression or knockdown in GC cells. *P < 0.05, **P < 0.01
Discussion
Recently, many lncRNA transcripts have been shown to be overexpressed in tumor tissues and confirmed to be related to the biological behaviors of tumors. LncRNAs are currently known to exert important effects on many cellular processes, such as cell differentiation, metabolism, the cell cycle, and different disorders [25].
PVT1 is related to the regulation of tumors, including colorectal [26], gallbladder [27], and ovarian cancers [28]; however, the function of the PVT1-214 transcript (Ensembl ID: ENST00000522875) has rarely been reported.
The present study reports that the PVT1-214 transcript level was elevated in human GC tissues (see Fig. 1a and b). Furthermore, PVT1-214 expression was verified in GC clinical samples, and it was found that PVT1-214 upregulation was associated with tumour size, late pathological stage, and distant metastasis. Furthermore, PVT1-214 expression was found to be a valuable prognostic indicator, with the potential to independently predict patient outcomes.In addition, gain- and loss-of-function assays revealed that PVT1-214 significantly promoted GC cell growth, invasion, and migration in both in vivo and in vitro models. This is the first study to demonstrate that the PVT1-214 transcript is a potential oncogene for GC and has a significant effect on the pathogenic mechanism of GC, and the functional experiments demonstrate that PVT1-214 overexpression markedly enhances the malignant phenotypes of GC cells, while PVT1-214 knockdown suppresses their malignant phenotypes, thus suggesting a promotion of GC occurrence by PVT1-214. This study is the first to reveal the effect of PVT1-214 on GC.
MiRNAs exert crucial regulatory effects on GC. For example, regulating miR-1-3p expression may promote GC development [29], and miR-1343-3p promotes GC progression by targeting MAP3K6/MMP [30]. miR-671-5p is suggested to significantly affect the development of different cancer types. Similarly, miR-671-5p expression decreases within GC tissues, and its expression affects GC cell growth, migration and invasion [31]. According to our results, miR-671-5p expression decreases within GC tissue relative to its matched noncarcinoma counterpart. Suppressing miR-671-5p expression promoted GC cell growth, invasion and migration, whereas miR-671-5p overexpression had the opposite effect. Therefore, miR-671-5p suppresses GC progression.
LncRNAs are reported to regulate mRNA expression by acting as competitive endogenous RNAs (ceRNAs), which regulate protein levels by combining with 3ʹUTRs in mRNAs. This association has an important effect on GC development. For example, PVT1-214 functions as a ceRNA for suppressing autophagy-induced chemoresistance through the inhibition of miR-128 in CRC [32], and the lncRNA MIR200CHG suppresses epithelial‒mesenchymal transition (EMT) in GC by protecting miR-200c against target-directed miRNA degradation [33]. Our results revealed that PVT1-214 expression was negatively related to miR-671-5p in GC tissues. Furthermore, qRT‒PCR, RIP and luciferase reporter assays verified that PVT1-214 directly bound to miR-671-5p, the RNA-binding oncoprotein responsible for promoting cancer development.
Solute carrier family 45 (SLC45) belongs to the putative sugar transporter family. It comprises four members, SLC45A1–A4, which are not only involved in sugar transport and reabsorption but also may play important roles in tumors. Higher sugar absorption is a hallmark of cancer cells; moreover, abnormal expression levels of SGLT and GLUT are detected in numerous cancers [34]. The SLC45 family can transport sugars H+-dependently; therefore, cancer cells can channel a greater number of sugars in glycolysis, thus gaining benefits from increased sugar adsorption [35]. Consequently, the SLC45 family is a key target for cancer through restricting the energy access of cancer cells. For example, the expression of SLC45 family members is a good prognostic indicator for melanoma [36], and SLC45A4 can prevent autophagy through the AMPK/ULK1 axis in TP53-mutant pancreatic ductal adenocarcinoma (PDAC) and is a candidate biomarker and therapeutic target for TP53-mutant PDAC [37].
Our results revealed that SLC45A4 expression increased in GC tissues and was negatively correlated with miR-671-5p expression. Because miR-671-5p can target SLC45A4 in GC, we further determined whether SLC45A4 expression was modulated via the PVT1-214/miR-671-5p axis. According to our results, miR-671-5p mimics or PVT1-214 knockdown inhibited SLC45A4, whereas miR-671-5p inhibitor or PVT1-214 overexpression increased SLC45A4 expression. Consequently, PVT1-214 modulates GC development by regulating the miR-671-5p/SLC45A4 axis.
Conclusions
Taken together, these findings indicate that the lncRNA PVT1-214 is vital for GC cell proliferation in vitro and for tumor proliferation in vivo. As confirmed by mechanistic analysis, PVT1-214 regulates the miR-671-5p/SLC45A4 axis to modulate the development of gastric cancer. In conclusion, the results of this study elucidate the mechanisms of gastric cancer progression and lay a theoretical foundation for the diagnosis and treatment of the disease.
Data availability
No datasets were generated or analysed during the current study.
References
López MJ, Carbajal J, Alfaro AL, Saravia LG, Zanabria D, Araujo JM, Quispe L, Zevallos A, Buleje JL, Cho CE, Sarmiento M, Pinto JA, Fajardo W. Characteristics of gastric cancer around the world. Crit Rev Oncol Hematol. 2023;181:103841. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.critrevonc.2022.103841. Epub 2022 Oct 11. PMID: 36240980.
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F, Global Cancer S. 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71(3):209–249. https://doiorg.publicaciones.saludcastillayleon.es/10.3322/caac.21660. Epub 2021 Feb 4. PMID: 33538338.
Tekin C, Ercelik M, Dunaev P, Galembikova A, Tezcan G, Aksoy SA, Budak F, Isık O, Ugras N, Boichuk S, Tunca B. Leaf Extract from European Olive (Olea europaea L.) post-transcriptionally suppresses the epithelial–mesenchymal transition and sensitizes gastric cancer cells to chemotherapy. Biochemistry (Mosc). 2024;89(1):97–115. https://doiorg.publicaciones.saludcastillayleon.es/10.1134/S0006297924010061. PMID: 38467548.
Song Z, Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumor Biol. 2017;39(7):1010428317714626. https://doiorg.publicaciones.saludcastillayleon.es/10.1177/10104283177146-26.PMID: 28671042.
Liu H, Liu X. LINC01207 is upregulated in gastric cancer tissues and promotes disease progression by regulating miR-671-5p/DDX5 axis. J Biochem. 2021;170(3):337–347. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jb/mvab050. PMID: 33856490.
Liu G, Kim J, Nguyen NH, Zhou L, Dean A. Long noncoding RNA GATA2AS influences human erythropoiesis by transcription factor and chromatin landscape regulation. Blood. 2024 Mar 6:blood.2023021287. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.2023021287. Epub ahead of print. PMID: 38447046.
E M, Ren F, Yu Y, Li H, Shen C. The role of lncRNAKCNQ1OT1/miR-301b/Tcf7 axis in cardiac hypertrophy. Cardiol Young. 2024;1–13. https://doiorg.publicaciones.saludcastillayleon.es/10.1017/S1047951124000155. Epub ahead of print. PMID: 38456301.
Zhang M, Yang L, Hou L, Tang X. LncRNA SNHG1 promotes tumor progression and cisplatin resistance through epigenetically Silencing miR-381 in breast cancer. Bioengineered. 2021;12(2):9239–50. PMID: 34806925; PMCID: PMC8809974.
Chen J, Liu A, Wang Z, Wang B, Chai X, Lu W, Cao T, Li R, Wu M, Lu Z, Pang W, Xiao L, Chen X, Zheng Y, Chen Q, Zeng J, Li J, Zhang X, Ren D, Huang Y. LINC00173.v1 promotes angiogenesis and progression of lung squamous cell carcinoma by sponging miR-511-5p to regulate VEGFA expression. Mol Cancer. 2020;19(1):98. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-020-01217-2. PMID: 32473645; PMCID: PMC7260858.
Jiang L, Zhang Y, Su P, Ma Z, Ye X, Kang W, Liu Y, Yu J. Long noncoding RNA HNF1A-AS1 induces 5-FU resistance of gastric cancer through miR-30b-5p/EIF5A2 pathway. Transl Oncol. 2022;18:101351. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tranon.2022.101351. Epub 2022 Jan 26. PMID: 35092904; PMCID: PMC8802127.
Lu J, Xu F, Lu H. LncRNA PVT1 regulates ferroptosis through miR-214-mediated TFR1 and p53. Life Sci. 2020;260:118305. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.lfs.2020.118305. Epub 2020 Aug 20. PMID: 32827544.
Xin L, Lu H, Liu C, Zeng F, Yuan YW, Wu Y, Wang JL, Wu DZ, Zhou LQ. Methionine deficiency promoted mitophagy via LncRNA PVT1-mediated promoter demethylation of BNIP3 in gastric cancer. Int J Biochem Cell Biol. 2021;141:106100. Epub 2021 Oct 20. PMID: 34678458.
He GN, Bao NR, Wang S, Xi M, Zhang TH, Chen FS. Ketamine induces ferroptosis of liver Cancer cells by targeting LncRNA PVT1/miR-214-3p/GPX4. Drug Des Devel Ther. 2021;15:3965–78. PMID: 34566408; PMCID: PMC8458041.
Sun C, Ye Y, Tan Z, Liu Y, Li Y, Hu W, Liang K, Egranov SD, Huang LA, Zhang Z, Zhang Y, Yao J, Nguyen TK, Zhao Z, Wu A, Marks JR, Caudle AS, Sahin AA, Gao J, Gammon ST, Piwnica-Worms D, Hu J, Chiao PJ, Yu D, Hung MC, Curran MA, Calin GA, Ying H, Han L, Lin C, Yang L. Tumor-associated nonmyelinating Schwann cell-expressed PVT1 promotes pancreatic cancer kynurenine pathway and tumor immune exclusion. Sci Adv. 2023;9(5):eadd6995. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.add6995. Epub 2023 Feb 1. PMID: 36724291; PMCID: PMC9891701.
He F, Song Z, Chen H, Chen Z, Yang P, Li W, Yang Z, Zhang T, Wang F, Wei J, Wei F, Wang Q, Cao J. Long noncoding RNA PVT1-214 promotes proliferation and invasion of colorectal cancer by stabilizing Lin28 and interacting with miR-128. Oncogene. 2019;38(2):164–179. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41388-018-0432-8. Epub 2018 Aug 3. Erratum in: Oncogene. 2022;41(3):459. PMID: 30076414; PMCID: PMC6329639.
Hong X, Jiang M, Kho AT, Tiwari A, Guo H, Wang AL, McGeachie MJ, Weiss ST, Tantisira KG, Li J. Circulating MiRNAs associate with historical childhood asthma hospitalization in different serum vitamin D groups. Respir Res. 2024;25(1):118. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12931-024-02737-x. PMID: 38459594; PMCID: PMC10921757.
Šustr F, Macháčková T, Pešl M, Svačinova J, Trachtová K, Stárek Z, Kianička B, Slabý O, Novák J. Identification of plasmatic microRNA-206 as new predictor of early recurrence of atrial fibrillation after catheter ablation using next-generation sequencing. Mol Diagn Ther. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40291-024-00698-x. Epub ahead of print. PMID: 38459249.
Hu K, Liu L, Tang S, Zhang X, Chang H, Chen W, Fan T, Zhang L, Shen B, Zhang Q. MicroRNA-221-3p inhibits the inflammatory response of keratinocytes by regulating the DYRK1A/STAT3 signaling pathway to promote wound healing in diabetes. Commun Biol. 2024;7(1):300. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s42003-024-05986-0. PMID: 38461326; PMCID: PMC10924844.
Ibrahim P, Denniston R, Mitsuhashi H, Yang J, Fiori LM, Żurawek D, Mechawar N, Nagy C, Turecki G. Profiling small RNA from brain extracellular vesicles in individuals with depression. Int J Neuropsychopharmacol. 2024;27(3):pyae013. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/ijnp/pyae013. PMID: 38457375; PMCID: PMC10946232.
Grenda A, Kuźnar-Kamińska B, Kalinka E, Krawczyk P, Sawicki M, Filip A, Chmielewska I, Frąk M, Krzyżanowska N, Milanowski J. MicroRNA-126 selected with broad-spectrum analysis of microRNAs - a new predictive factor for the effectiveness of immunotherapy or chemoimmunotherapy in advanced NSCLC patients? Front Immunol. 2024;15:1344858. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2024.1344858. PMID: 38469304; PMCID: PMC10925701.
Feng PF, Zhu LX, Sheng N, Li XS, Liu PG, Chen XF. CircXRN2 accelerates colorectal cancer progression through regulating miR-149-5p/MACC1 axis and EMT. Sci Rep. 2024;14(1):2448. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-52257-3. PMID: 38291092; PMCID: PMC10828403.
Chakraborty A, Patnaik J, Sinha A, Parida N, Parija T, Patnaik S. In Silico analysis shows slc1a4 as a potential target of hsa-mir-133a for regulating glutamine metabolism in gastric cancer. Int J Biol Macromol. 2024;282(Pt 5):136974. Epub 2024 Oct 31. PMID: 39486714.
Ye J, Luo W, Luo L, Zhai L, Huang P. MicroRNA–671–5p inhibits cell proliferation, migration and invasion in non–small cell lung cancer by targeting MFAP3L. Mol Med Rep. 2022;25(1):30. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/mmr.2021.12546. Epub 2021 Nov 29. Retraction in: Mol Med Rep. 2024;29(6): PMID: 34841435; PMCID: PMC8669681.
Tan X, Li Z, Ren S, Rezaei K, Pan Q, Goldstein AT, Macri CJ, Cao D, Brem RF, Fu SW. Dynamically decreased miR-671-5p expression is associated with oncogenic transformation and radiochemoresistance in breast cancer. Breast Cancer Res. 2019;21(1):89. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13058-019-1173-5. PMID: 31391072; PMCID: PMC6686561.
Bridges MC, Daulagala AC, Kourtidis A. LNCcation: LncRNA localization and function. J Cell Biol. 2021;220(2):e202009045. https://doiorg.publicaciones.saludcastillayleon.es/10.1083/jcb.202009045. PMID: 33464299; PMCID: PMC7816648.
Guo H, Zhuang K, Ding N, Hua R, Tang H, Wu Y, Yuan Z, Li T, He S. High-fat diet induced Cyclophilin B enhances STAT3/lncRNA-PVT1 feedforward loop and promotes growth and metastasis in colorectal cancer. Cell Death Dis. 2022;13(10):883. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-022-05328-0. PMID: 36266267; PMCID: PMC9584950.
Jin L, Cai Q, Wang S, Wang S, Wang J, Quan Z. Long noncoding RNA PVT1 promoted gallbladder cancer proliferation by epigenetically suppressing miR-18b-5p via DNA methylation. Cell Death Dis. 2020;11(10):871. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41419-020-03080-x. PMID: 33067424; PMCID: PMC7568542.
Wu Y, Gu W, Han X, Jin Z. LncRNA PVT1 promotes the progression of ovarian cancer by activating TGF-β pathway via miR-148a-3p/AGO1 axis. J Cell Mol Med. 2021;25(17):8229–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jcmm.16700. Epub 2021 Jul 21. PMID: 34288373; PMCID: PMC8419181.
Deng C, Huo M, Chu H, Zhuang X, Deng G, Li W, Wei H, Zeng L, He Y, Liu H, Li J, Zhang C, Chen H. Exosome circATP8A1 induces macrophage M2 polarization by regulating the miR-1-3p/STAT6 axis to promote gastric cancer progression. Mol Cancer. 2024;23(1):49. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12943-024-01966-4. PMID: 38459596; PMCID: PMC10921793.
Wang X, Zhang Z, Cao X. Salidroside inhibited the proliferation of gastric cancer cells through upregulating tumor suppressor miR-1343-3p and downregulating MAP3K6/MMP24 signal molecules. Cancer Biol Ther. 2024;25(1):2322206. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/15384047.2024.2322206. Epub 2024 Mar 4. PMID: 38436092; PMCID: PMC10913707.
Song H, Xu Y, Xu T, Fan R, Jiang T, Cao M, Shi L, Song J. CircPIP5K1A activates KRT80 and PI3K/AKT pathway to promote gastric cancer development through sponging miR-671-5p. Biomed Pharmacother. 2020;126:109941. Epub 2020 Mar 10. PMID: 32169757.
Tong D, Zou E, Bai L, Ma J, Guo N, Wang H, Jiang L. IRF-1 mediated long noncoding RNA PVT1-214 promotes oxaliplatin resistance of colorectal cancer via miR-128 Inhibition. J BUON. 2020;25(5):2205–14. PMID: 33277837.
Zhu Y, Huang C, Zhang C, Zhou Y, Zhao E, Zhang Y, Pan X, Huang H, Liao W, Wang X. LncRNA MIR200CHG inhibits EMT in gastric cancer by stabilizing miR-200c from target-directed MiRNA degradation. Nat Commun. 2023;14(1):8141. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-43974-w. PMID: 38065939; PMCID: PMC10709323.
Bartölke R, Heinisch JJ, Wieczorek H, Vitavska O. Proton-associated sucrose transport of mammalian solute carrier family 45: an analysis in Saccharomyces cerevisiae. Biochem J. 2014;464(2):193–201. https://doiorg.publicaciones.saludcastillayleon.es/10.1042/BJ20140572. PMID: 25164149.
Gerweck LE, Seetharaman K. Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancer. Cancer Res. 1996;56(6):1194-8. PMID: 8640796.
Xie J, Ruan S, Zhu Z, Wang M, Cao Y, Ou M, Yu P, Shi J. Database mining analysis revealed the role of the putative H+/sugar transporter solute carrier family 45 in skin cutaneous melanoma. Channels (Austin). 2021;15(1):496–506. PMID: 34334114; PMCID: PMC8331014.
Chen W, Huang F, Huang J, Li Y, Peng J, Zhuang Y, Huang X, Lu L, Zhu Z, Zhang S. SLC45A4 promotes Glycolysis and prevents AMPK/ULK1-induced autophagy in TP53 mutant pancreatic ductal adenocarcinoma. J Gene Med. 2021;23(9):e3364. Epub 2021 Jun 2. PMID: 34010493; PMCID: PMC8459293.
Acknowledgements
Not applicable.
Funding
The study was financially supported by the Anhui Medical University (grant no. 2022xkt240).
Author information
Authors and Affiliations
Contributions
Wei Yan and yong-xiang Li contributions to the experimental concept and design, Yong Zhao and Hui-zheng Wang contributions to data acquisition or data analysis and interpretation; Wei-tong Zhang participated in drafting the article or strictly revised the content of important knowledge content. All authors have agreed to be responsible for all aspects of the work.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Ethical approval for this study (Ethical Approval NO. 2023LL041) was provided by the Ethics Committee of Lu’an Affiliated Hospital of Anhui Medical University, China, on 22 December 2023. Written informed consent was obtained from each patient.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Yan, W., Wang, H., Zhao, Y. et al. The PVT1-214/miR-671-5p/SLC45A4 signaling axis regulates cell proliferation in human gastric cancer. World J Surg Onc 23, 158 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12957-025-03805-2
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12957-025-03805-2