2025 Volume 50 Issue 5 Pages 235-244
With a fourth-place death rate among all malignancies, gastric cancer (GC) is one of the most prevalent tumors globally. As a primary malignant characteristic of GC, metastasis contributes substantially to a high death rate and unfavorable prognosis. miRNA-214-3p can influence cell apoptosis since it is an autophagy-regulating molecule. Its significance in GC malignant development has not, however, been investigated in terms of mechanism. qRT-PCR was utilized to confirm expression of miRNA-214-3p in GC tissues and cells. Bioinformatics analysis was then implemented to examine BNIP3 expression in GC as well as binding interaction between BNIP3 and miRNA-214-3p. The targeting capability of miRNA-214-3p on BNIP3 was confirmed using the dual-luciferase assay. Capacities of cells to proliferate, migrate, and invade were assayed using Transwell assays and colony formation. In order to determine if GC cells were capable of autophagy, immunofluorescence and western blot were employed. In GC, miRNA-214-3p was substantially expressed in GC tissues and cells, but BNIP3 was downregulated, as shown by bioinformatics analysis and verified by cell tests. MiRNA-214-3p targeted BNIP3, as shown by further bioinformatics analysis, and dual-luciferase experiment verified this binding connection. MicroRNA-214-3p facilitated cell invasion, migration, and proliferation, as shown by Transwell tests and colony formation. MiRNA-214-3p accelerated malignant development of GC by targeting BNIP3 to impact autophagy, as demonstrated by immunofluorescence and western blot analyses. By targeting BNIP3 to affect autophagy, miRNA-214-3p aided in the malignant growth of GC. This suggested that miRNA-214-3p may function as a likely therapeutic target or biomarker for the disease, with significant implications for early diagnosis and treatment of patients.
Situated fifth in incidence and fourth in death among all malignancies, gastric cancer (GC) is one of the most prevalent malignant tumors globally (Sung et al., 2021). A significant strain on public health was placed on China’s health system in 2022, when there were an estimated 509,000 newly diagnosed cases of GC there, along with a death toll of around 400,000 (Xia et al., 2022). When individuals seek medical attention for GC, most are already in late stages since early-stage patients often do not exhibit visible symptoms (Feng et al., 2019). The mainstay of treatment for GC is still surgery; however, for individuals whose cancer has progressed locally, it can also be coupled with novel adjuvant treatments, chemotherapy, and radiation (Tan, 2019). However, advanced GC patients frequently experience grim treatment results due to lymph node metastases, with a median overall survival of 8 months (Tan, 2019; Li et al., 2021). It is worth mentioning that targeted therapy has attained headway in cancer treatment, including GC (Guan et al., 2023). The search for novel biomarkers to use as therapy targets, as well as the investigation of putative processes behind GC development, will present patients with new and effective treatment alternatives.
Non-coding RNAs have emerged as a new study subject in recent years, playing an essential role in controlling onset and progression of varying malignancies. MicroRNAs, which are functional non-coding RNAs involved in regulation and possible biomarkers for illness prediction, have been extensively investigated (Yan and Bu, 2021; Ren and Wang, 2021). miRNA-214-3p has been proposed as a likely therapeutic target or prognostic marker for cancer, although its effect on different forms of cancer is inconsistent. Some studies claimed that miRNA-214-3p, as a possible biomarker for cancer, has the ability to curb tumor malignant progression, such as endometrial cancer (Fang et al., 2019), colorectal cancer (Zhou et al., 2020), hepatocellular carcinoma (Li et al., 2017). On the contrary, there are also studies suggesting that miRNA-214-3p can serve as a tumor predictor, being highly expressed in cancers such as epithelial ovarian cancer (Yang et al., 2019) and bladder cancer (Cheng et al., 2024), in turn promoting tumor progression. Moreover, multiple studies have demonstrated upregulation of miRNA-214-3p, which is associated with malignant progression of GC (He et al., 2018; Jiang et al., 2021). The findings of the preceding studies suggest that miRNA-214-3p may be a viable therapeutic target for treatment and prognosis of GC patients. However, the underlying processes require additional exploration.
Autophagy is a set of primary systems that transport intracellular substances to lysosomes for degradation and recycling (Debnath et al., 2023). In various diseases, such as Parkinson’s disease (Dong et al., 2024), atherosclerosis (Qian et al., 2023), and Alzheimer’s disease (Zhou et al., 2021), autophagy is often suppressed by overexpressed miRNA-214-3p, which in turn affects the development of the disease. For example, miRNA-214-3p affects autophagy by targeting ATG3 to promote Parkinson’s disease pathogenesis (Dong et al., 2024). Furthermore, in early cancer metastasis, autophagy acts as a mode of programmed cell death to inhibit tumorigenesis (Gundamaraju et al., 2022). Chai et al. reported that AIM2 represses malignant progression of renal cancer by boosting autophagy induction (Chai et al., 2018). In head and neck squamous cell carcinoma, silencing LSD1 inhibits tumor progression by inducing autophagy and cellular pyroptosis (Wang and Liu, 2023). Similarly, in a study by Zhao et al., miRNA-29b-3p mimic facilitates GC progression by upregulating autophagy-related proteins (Zhao et al., 2021). However, no study has yet elucidated the role played by miRNA-214-3p-mediated autophagy in GC. Therefore, this study focused on potential mechanism of miRNA-214-3p in malignant progression of GC.
We demonstrated high expression of miRNA-214-3p in GC tissues and cells and delved into the mechanisms involved in GC malignant progression mediated by miRNA-214-3p. Our results demonstrated that miRNA-214-3p drove GC malignant progression by targeting BNIP3 to influence autophagy. In conclusion, our research offers novel therapeutic targets or prognostic indicators for GC patients and reveals possible roles of miRNA-214-3p in the malignant course of the disease.
This study enrolled patients diagnosed with GC at Fujian Cancer Hospital from January 2023 to August 2023. GC tissues (n=10) and adjacent normal gastric tissues (n=10) were collected from the patients. The obtained fresh tissue samples were rapidly frozen in liquid nitrogen and stored at -80°C. This study obtained approval from the Fujian Cancer Hospital Ethics Committee (approval number: K2024-287-01) and written informed consent from all subjects.
Bioinformatics analysisBioinformatics analysis was completed to determine BNIP3 expression in GC tissues and adjacent normal tissues, as well as to predict binding relationship between miRNA-214-3p and BNIP3. Databases used in this study are publicly available and can be used reasonably.
Cell culture, transfection, and reagentsHuman embryonic kidney cells (293T), normal human gastric epithelial cells (GES1), and human GC cell lines (SNU-1, AGS) were all purchased from BNCC (China), and human GC cell lines (Fu97, HGC-27) were purchased from CoBior Biosciences Co., Ltd. (China). 293T, GES1, and Fu97 cells were cultured in DMEM-H medium. The medium for Fu97 cells also contained 10 μg/mL insulin. SNU-1 and HGC-27 cells were cultured in RPMI-1640 medium. AGS cells were cultured in F-12K medium. All mediums contained 10% FBS and 1% penicillin/streptomycin. All cells were cultured in a cell incubator at 37°C with 5% CO2. miRNA-214-3p mimic, miRNA-214-3p inhibitor, oe-BNIP3 (pcDNA3.1), and corresponding negative controls were purchased from Ribobio. The sequences used are shown in Supplementary Table 1. They were transfected into cells at approximately 80% confluence in the logarithmic growth phase using Lipofectamine 2000 reagent (Thermo Fisher Scientific, USA). Cells were then subjected to further experiments after 48 hr of transfection.
qRT-PCRClinical samples and cells were treated with TRIzol reagent (Thermo Fisher Scientific, USA) to isolate total RNA. Using NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, USA), RNA concentration and purity were ascertained. Total RNA was synthesized into cDNA using PrimeScript™ RT reagent Kit (Takara, Japan). Next, qRT-PCR assays were performed using AceQ qPCR SYBR Green Master Mix (Vazyme, China) on an Applied Biosystems 7500 Rapid Real-Time Fluorescence Quantitative PCR System (Thermo Fisher Scientific, USA). For miRNA-214-3p, specific stem-loop primers were used for qPCR. The relative expression levels of miRNA-214-3p and BNIP3 were normalized to internal reference U6 (miRNA-214-3p) and GAPDH (BNIP3) using 2-∆∆Ct method. The primer sequences used were as follows:
miRNA-214-3p stem-loop primer: 5-CTCAACTGGTGTCGTGGAGTCGGCAATTCAGTTGAGACTGCCTG-3.
miRNA-214-3p F: 5-TCGGCAGGACAGCAGGCA-3, R: 5-CTCAACTGGTGTCGTGGAGTCG-3.
U6 F: 5-CTCGCTTCGGCAGCACA-3, R: 5-AACGCTTCACGAATTTGCGT-3.
BNIP3 F: 5-GCCCACCTCGCTCGCAGACAC-3, R: 5-CAATCCGATGGCCAGCAAATGAGA-3.
GAPDH F: 5-AAGGTCGGAGTCAACGGATTTG-3, R: 5-CCATGGGTGGAATCATATTGGAA-3.
Colony formation assay for cell proliferationCells were assayed by colony formation assay 24 hr after transfection. In brief, cells from different treatment groups were seeded in 12-well plates (200 cells/well) and incubated for 10 days, with mediums replaced every 3-4 days. Finally, cells were fixed with 4% paraformaldehyde and stained with 0.5% crystal violet staining solution for 30 min.
Transwell assay for cell migration and invasionMigration and invasion abilities of cells were assayed using the Transwell assays. For cell migration assay, cells (2×104 cells) were seeded in the upper chamber of a 24-well plate (Corning, USA) with 8 μm pore size, and medium containing 10% FBS was added to the lower chamber. After incubation in a cell culture incubator for 24 hr, cells that did not migrate through pores were carefully wiped off with a cotton swab. For cell invasion assay, Matrigel (BD Biosciences, USA) was diluted with serum-free cell culture medium to a volume of 50 μL and coated on the chamber. The coated Matrigel was allowed to polymerize into a thin film by incubating at 37°C for 4 hr. Then, 200 μL (2×104 cells) of serum-free medium was added to the upper chamber, and 600 μL of medium containing 10% FBS was filled into the lower chamber. Following incubation at 37°C for 48 hr, cells were fixed with 4% paraformaldehyde and stained with crystal violet staining solution for 30 min. Random fields were captured using a microscope (Olympus Corp, Japan), and the number of migrated or invaded cells was counted.
Immunofluorescence stainingTransfected cells were seeded onto 6-well plates with coverslips and cultured for 24 hr, followed by 10-min fixing with 4% paraformaldehyde, 10-min permeabilization with 0.25% Triton X-100, and 1-hr blockage with 3% albumin from bovine serum (BSA). Cells were incubated with anti-LC3B (1:2000) overnight at 4°C. After washing cells with PBS three times, they were incubated with goat anti-rabbit IgG H&L (Alexa Fluor® 594) (Abcam, UK) at 37°C for 1 hr, washed, and stained with DAPI solution for 3 min. Anti-fade mounting medium (Beyotime, China) was instrumental to mount coverslips. LC3-positive autophagosome vesicles were counted and observed using a Zeiss LSM880 inverted confocal microscope.
Western blot (WB)Protein expressions were examined using WB. In brief, cells were lysed in RIPA lysis buffer containing protease and phosphatase inhibitors. The lysates were centrifuged at 12,000 rpm for 15 min at 4°C, and the supernatants were collected. Total protein concentration of samples was determined using BCA protein assay kit (Beyotime, China). The protein samples were loaded at 25 μg onto a 10% SDS-polyacrylamide gel for electrophoresis and then transferred to a PVDF membrane. The membranes were washed three times for 5 min each using TBST solution and closed for 1 hr at room temperature in 5% skimmed milk powder. Subsequently, primary antibodies rabbit anti-human: LC3B (Abcam, UK), P62 (Abcam, UK), GAPDH (Abcam, UK) were added for incubation at 4°C overnight. The membrane was rinsed three times to remove unbound primary antibody for 5 min each time, then incubated with secondary antibody goat anti-rabbit IgG H&L (HRP) (Abcam, UK) for 1 hr at room temperature, followed by washing the membrane three times to remove unbound secondary antibody for 5 min each time. ECL kit (Beyotime, China) was instrumental for color development. Blot images were captured using the ChemiScope6000 chemiluminescence imaging system (Clinx, China). Quantitative analysis of the WB image results was performed using ImageJ software.
Dual-luciferase reporter gene assayFollowing the steps described in reference (Li et al., 2020), interaction between miRNA-214-3p and BNIP3 was assessed using a dual luciferase assay. First, BNIP3 3’UTR WT or BNIP3 3’-UTR Mut was inserted into the pmirGLO vector (Promega, USA) to construct recombinant plasmid, which was co-transfected with inhibitor-miRNA-214-3p/inhibitor-NC into 293T cells using Lipofectamine 2000 (Thermo Fisher Scientific, USA). 48 hr after transfection, fluorescence values were detected on Dual-Luciferase Reporter Assay System (Promega, USA).
Statistical analysisEvery experiment was conducted thrice, and the results were expressed as mean ± standard deviation. With GraphPad Prism 8.0 (GraphPad Software, USA), statistical analysis and graphing were carried out. To compare the differences between the two groups, the student’s t-test was employed. ANOVA analysis was applied to examine the variations between multiple groups. Moreover, a significant difference with p<0.05 is indicated by *.
It has been documented that miRNA-214-3p is highly expressed in GC (He et al., 2018; Jiang et al., 2021). First, we isolated GC tissues (n=10) and adjacent normal tissues (n=10) from patients and determined expression of miRNA-214-3p via qRT-PCR. Consistent with previous studies, expression of miRNA-214-3p in GC tissues was significantly higher than in adjacent normal tissues (Fig. 1A). Furthermore, we examined expression of miRNA-214-3p in human normal gastric epithelial cells (GES1) and GC cell lines (SNU-1, AGS, Fu97, HGC-27) by qRT-PCR. It was significantly upregulated in GC cell lines but not in human normal epithelial cells (Fig. 1B). Taken together, these results indicate that miRNA-214-3p was highly expressed in GC tissues and cells.
High expression of miRNA-214-3p in GC. (A) qRT-PCR for miRNA-214-3p expression in 10 pairs of GC tissues and adjacent normal tissues; (B) qRT-PCR for miRNA-214-3p expression in human normal gastric epithelial cells (GES1) and human GC cell lines (SNU-1, Fu97, HGC-27, AGS); *p<0.05.
miRNA-214-3p has been theorized to be associated with cell autophagy (Qian et al., 2023; Zhou et al., 2021). To demonstrate the role of miRNA-214-3p in facilitating GC malignant progression through autophagy, we selected HGC-27 cell line with high expression of miRNA-214-3p and transfected inhibitor-NC and inhibitor-miRNA-214-3p into HGC-27 cell line. qRT-PCR confirmed the efficacy of transfection. The findings demonstrated a considerable suppression of miRNA-214-3p expression in the inhibitor-miRNA-214-3p group as compared to the inhibitor-NC group (Fig. 2A). Colony formation assay unveiled that proliferation potential of cells in the inhibitor-miRNA-214-3p group was significantly stamped down compared to the inhibitor-NC group (Fig. 2B). According to results of Transwell assay, cells in the inhibitor-miRNA-214-3p group had much less capacity for invasion and migration than cells in the inhibitor-NC group (Fig. 2C). These results suggested that suppressing expression of miRNA-214-3p significantly restrained proliferation, migration, and invasion capabilities of HGC-27 cells. To investigate whether miRNA-214-3p drives malignant progression of GC through autophagy, immunofluorescence staining was employed for assessment of cells in the inhibitor-NC group and the inhibitor-miRNA-214-3p group. Compared to the inhibitor-NC group, cells in the inhibitor-miRNA-214-3p group formed multiple bright red fluorescence spots under fluorescence microscope, and the counting results also indicated that autophagy activity in the inhibitor-miRNA-214-3p group was significantly higher than that in the inhibitor-NC group (Fig. 2D). Through further detecting of the changes of LC3II/I and P62 protein by WB, we found that LC3II/I was significantly up-regulated and P62 was significantly reduced in the inhibitor-miRNA-214-3p group compared with the inhibitor-NC group. Meanwhile, in the groups treated with the autophagy inhibitor (3-Methyladenine, 3-MA), the LC3II/I ratio was significantly reduced, and the expression level of P62 protein was markedly increased (Fig. 2E). This indicated that inhibitor-miRNA-214-3p promoted autophagy in GC. In summary, miRNA-214-3p drove malignant progression of GC through autophagy.
miRNA-214-3p promotes malignant progression of GC through autophagy. (A) qRT-PCR detection of the expression level of miRNA-214-3p in the cells of the inhibitor-NC and inhibitor-miRNA-214-3p groups; (B) Colony formation assay of the proliferative ability of the cells in the above subgroups; (C) Transwell assessment of the migration and invasion of the cells in the above subgroups; (D) Immunofluorescence staining for detection of the number of the LC3-positive autophagosome vesicles; (E) WB detection of changes in LC3 II/I and P62. 3-MA was purchased from MCE (USA). *p<0.05.
Using bioinformatics analysis, we first discovered that the BNIP3 level in GC tissues was significantly lower than that in adjacent normal tissues (Fig. 3A). Using qRT-PCR, we were able to identify differences in BNIP3 expression between human normal gastric epithelial cells (GES1) and GC cells (SNU-1, AGS, Fu97, HGC-27), therefore validating the results of the bioinformatics analysis. BNIP3 mRNA was significantly downregulated in GC cells compared with human normal gastric epithelial cells (Fig. 3B). Moreover, bioinformatics research demonstrated that BNIP3 was a target gene of miRNA-214-3p because it could bind to the miRNA-214-3p at the 660–666 position of its 3’UTR (Fig. 3C). To validate this prediction, miRNA-214-3p mimic and luciferase reporter vectors containing wild-type BNIP3 3’UTR or mutant BNIP3 3’UTR were co-transfected into 293T cells. Dual-luciferase reporter gene assay unveiled that relative fluorescence values in the WT group transfected with inhibitor-miRNA-214-3p increased, while the values in the MUT group remained unchanged, indicating a target binding relationship between the two (Fig. 3D). Finally, inhibitor-NC and inhibitor-miRNA-214-3p were transfected into HGC-27 cells, and qRT-PCR was applied to test expression changes of BNIP3. Compared to the inhibitor-NC group, the addition of inhibitor-miRNA-214-3p significantly upregulated BNIP3 mRNA expression in the cells (Fig. 3E). In conclusion, miRNA-214-3p can target bind to BNIP3 3’UTR and repress BNIP3 expression.
miRNA-214-3p targets BNIP3. (A) Bioinformatics analysis of differential expression of BNIP3 in GC and adjacent tissues; (B) qRT-PCR detection of differential expression of BNIP3 in normal gastric epithelial cells (GES1) and GC cells (SNU-1, AGS, Fu97, HGC-27); (C) Bioinformatics analysis of binding sites between miRNA-214-3p and BNIP3; (D) Dual-luciferase assay of the binding relationship between miRNA-214-3p and BNIP3; (E) After transfection of inhibitor-NC and inhibitor-miRNA-214-3p into HGC-27 cells, qRT-PCR was employed for detection of BNIP3 expression changes; *p<0.05.
To further explore the mechanism that miRNA-214-3p targeted BNIP3 to affect autophagy and thus facilitate malignant progression of GC, firstly, mimic-NC and mimic-miRNA-214-3p were transfected into HGC-27 cells, and qRT-PCR was implemented to detect transfection efficiency, with its results confirming successful transfection of miRNA-214-3p (Fig. 4A). Secondly, oe-NC and oe-BNIP3 plasmids were transfected into the above cells, and qRT-PCR revealed that compared with the mimic-NC+oe-NC group, BNIP3 mRNA expression in the miRNA-214-3p mimic+oe-NC group was significantly inhibited, while the addition of overexpressed BNIP3 plasmid reversed this result (Fig. 4A). Next, the proliferative capacity of the above grouped cells was tested through colony formation assay. Compared to the mimic-NC+oe-NC group, the cell proliferation potential of the miRNA-214-3p mimic+oe-NC group was significantly promoted, while the addition of overexpressed BNIP3 plasmid reversed this phenomenon (Fig. 4B). Migration and invasion abilities of cells in the above groups were measured by Transwell assay, and results illustrated that these cell abilities of miRNA-214-3p mimic+oe-NC group were significantly higher than those of the control group, but overexpression of BNIP3 curbed these abilities promoted by miRNA-214-3p mimic (Fig. 4C). According to the data, transfection of the miRNA-214-3p mimic significantly enhanced the capacity of HGC-27 cells to proliferate, migrate, and invade; however, this behavior was reversed by transfection of an overexpression BNIP3 plasmid. To test whether miRNA-214-3p targets BNIP3 to affect the malignant progression of GC through autophagy. We performed immunofluorescence staining on cells from the mimic-NC+oe-NC group, miRNA-214-3p mimic+oe-NC group, and miRNA-214-3p mimic+oe-BNIP3 group. Compared to the control group, cells in the miRNA-214-3p mimic+oe-NC group exhibited significantly suppressed red fluorescence spots under fluorescence microscope, and the counting results also suggested that autophagic activity was significantly lower in the miRNA-214-3p mimic+oe-NC group than in the control group. On this basis, overexpression of BNIP3 resulted in the opposite effect (Fig. 4D). This suggested that miRNA-214-3p mimic could repress autophagosome accumulation, while transfection with the overexpression plasmid of BNIP3 reversed this phenomenon. By detecting expression of LC3II/I and P62 proteins through WB, we found that compared to the control group, the miRNA-214-3p mimic+oe-NC group showed downregulation of LC3II/I and a significant increase in P62 protein expression. This effect was reversed by transfection with the overexpression plasmid of BNIP3. It is noteworthy that in the groups treated with the autophagy inhibitor (3-MA), the LC3II/I ratio was significantly reduced, while the expression level of P62 protein was markedly increased (Fig. 4E). This indicated that miRNA-214-3p could repress autophagy in GC by targeting BNIP3. In summary, miRNA-214-3p promoted GC malignant progression by targeting BNIP3 to affect autophagy.
miRNA-214-3p targets BNIP3 to affect autophagy and promote the malignant progression of GC. (A) mimic-NC and miRNA-214-3p mimic were transfected into cells, and then oe-NC and oe-BNIP3 plasmids were further transfected, with transfection efficiency detected by qRT-PCR. (B) Colony formation assay of proliferation ability of cells in the above groups. (C) Transwell assay of migration and invasion of cells in the above groups. (D) Immunofluorescence staining for detection of the number of LC3-positive autophagosomes. (E) WB test of changes in LC3 II/I and P62. *p<0.05.
Non-coding RNAs have emerged as a new area of investigation in recent years, and miRNAs are key modulators of onset and progression of varying malignancies (Ren and Wang, 2021). Since numerous miRNAs have a role in gastric tumorigenesis, they are regarded as useful indicators for diagnosis and potential treatment targets (Christodoulidis et al., 2024). The high expression of miRNA-214-3p in GC tissues and cells was validated in our study, in line with other research results (Jiang et al., 2021; He et al., 2018). According to this, miRNA-214-3p may be a target for treating GC patients, as well as a prognostic sign.
Notably, there is discrepancy in the literature on the function of miRNA-214-3p in various malignancies. For instance, miRNA-214-3p is involved in favoring tumor growth in bladder cancer (Cheng et al., 2024) but preventing the malignant evolution of cancer in colorectal cancer (Zhou et al., 2020). Through cell studies, this study verified that suppressing expression of miRNA-214-3p can limit capacity of GC cells to proliferate, migrate, and invade. Furthermore, autophagy has been shown to considerably reduce metastasis and prevent cancer from developing in the first place (Gundamaraju et al., 2022). Based on Peng et al.’s research, circCUL2 controls the miRNA-142-3p/ROCK2 axis, which in turn controls autophagy activation-mediated GC malignant transformation (Peng et al., 2020). Thus, we hypothesized that miRNA-214-3p inhibited autophagy, thereby driving malignant growth of GC cells. We have corroborated this finding with our experiments. The results from WB and immunofluorescence analyses demonstrated that transfection of inhibitor-miRNA-214-3p markedly increased autophagy in GC cells.
Furthermore, our investigation of bioinformatics revealed that BNIP3 was downregulated in GC, a finding that was corroborated by cell tests. As a mitophagy-related protein, BNIP3 is crucial for the development of malignancies (Yu et al., 2023). According to research by Niu et al. (2019) and Zhang et al. (2020), suppressing BNIP3 can encourage the metastasis of cancer and its malignant development. In this work, cell tests and bioinformatics analysis verified that miRNA-214-3p may repress BNIP3 expression. Additionally, Xin et al.’s work showed that GC cell growth may be hindered by BNIP3-mediated mitophagy activation (Xin et al., 2021). We hypothesized, based on the aforementioned studies, that miRNA-214-3p introduced the malignant growth of GC by repressing cell autophagy through its targeting of BNIP3. Our experimental findings verified that by blocking autophagy, miRNA-214-3p mimic fostered GC cell malignant behaviors; BNIP3 overexpression counteracted this effect. From a mechanistic perspective, miR-214-3p binds to the 3’UTR region of BNIP3 mRNA, thereby inhibiting the stability and translation of BNIP3 transcripts. Consequently, the reduced expression of BNIP3 diminishes the association of damaged organelles with autophagosomes, leading to a decrease in autophagy levels (Gao et al., 2020; Gorbunova et al., 2020). The imbalance in autophagic homeostasis ultimately results in altered metabolic patterns and stem-like characteristics in tumor cells (Panigrahi et al., 2020), further promoting the progression of GC.
In conclusion, our research demonstrated that via targeting BNIP3 and influencing autophagy, miRNA-214-3p facilitated GC proliferation, migration, and invasion. These results offer GC patients a cutting-edge and effective treatment avenue and revealed that miRNA-214-3p may be a viable therapeutic target for the disease.
FundingThis work was supported by Startup Fund for Scientific Research, Fujian Medical University (Grant number:2021QH1145).
Conflict of interestThe authors declare that there is no conflict of interest.