- Research
- Open access
- Published:
Inhibition of esophageal squamous cell carcinoma progression by MIR210HG and activation of the P53 signaling pathway to promote apoptosis and autophagy
European Journal of Medical Research volume 30, Article number: 269 (2025)
Abstract
Background
Esophageal squamous cell carcinoma (ESCC) stands among the frequently occurring malignancies. The lack of efficient early detection methods and therapeutic approaches leads to a high mortality rate for ESCC. The long noncoding RNA MIR210HG is strongly related to various malignant tumors. However, its involvement in ESCC remains unexplored. Thus, this investigation aimed to assess the involvement of MIR210HG in ESCC development.
Methods
The MIR210HG expression was analyzed in numerous tumor types through pan-cancer analysis of The Cancer Genome Atlas(TCGA) database. This research investigated the MIR210HG role in the survival and prognosis of individuals with ESCC. The biological functions of MIR210HG were examined by enrichment analyses, including GO, GSEA, and KEGG. Moreover, immune cell infiltration, tumor microenvironment (TME) characteristics, and immune checkpoint expression levels associated with MIR210HG were explored. To get more insight into the connection between MIR210HG and ESCC, we assessed related gene and protein expression using Western blotting and qRT-PCR. To evaluate the proliferation, invasion, migration, apoptosis, and autophagy of ESCC cells, various techniques were employed, including EdU proliferation tests, monodansylcadaverine (MDC) staining, wound healing assays, cell colony formation, transwell assays, flow cytometry, and an established xenograft mouse model.
Results
MIR210HG exhibited low expression levels in ESCC. High expression of MIR210HG correlated with a higher survival rate among patients. The elevated expression of MIR210HG hindered the ESCC cell's ability to proliferate, invade, and migrate, both in vivo and in vitro settings. Furthermore, a positive correlation between MIR210HG and the P53 signaling pathway was observed, which could affect autophagy and apoptosis in ESCC cells.
Conclusions
MIR210HG emerges as a pivotal gene in ESCC, influencing both the immunity and prognosis of patients. Moreover, it may affect autophagy and apoptosis via the P53 signaling pathway. Overall, these outcomes present novel ideas for ESCC treatment.
Introduction
Esophageal cancer (EC) is known as the sixth significant contributor to cancer-related deaths, with esophageal squamous cell carcinoma (ESCC) being the most prevalent EC subtype [1]. ESCC stands as one of the most widespread malignant tumors globally, with over 450,000 new cases diagnosed annually and more than 400,000 deaths reported worldwide [2, 3]. The lack of effective early diagnostic and therapeutic approaches results in a survival rate of less than 20% over 5 years for ESCC [4]. Hence, there is a pressing need for the development of new therapeutic mechanisms and the exploration of effective diagnostic and prognostic approaches for ESCC.
Recently, rising transcriptomic investigations have demonstrated that instead of proteins, non-coding RNAs (ncRNAs) were encoded by the majority of human genes [5]. In general, ncRNAs are divided into two main groups: housekeeping and regulatory ncRNAs. Furthermore, regulatory ncRNAs are subdivided into micro RNAs (miRNAs, 18–25 nt), small-interfering RNAs (siRNAs, < 200 nt), long ncRNAs (lncRNAs, > 200 nt), and piwi-interacting RNAs (piRNAs, < 200 nt) [6]. The exploration of the potential involvement of ncRNAs in the treatment of ESCC has also been carried out. In particular, lncRNA genes are closely linked to the ESCC development. For example, lncSUMO1P3 further worsens ESCC through miR- 486 - 5p/PHF8/CD151 [7]. The tumorigenicity of ESCC is facilitated by lncRNA G077640 via the H2 AX–HIF1α–glycolysis axis [8]. LncRNA LOC146880 promotes ESCC progression through miR- 328 - 5p/FSCN1/MAPK axis [9]. Functional studies of these lncRNAs present novel strategies for ESCC treatment.
The transcript of MIR210HG, located on chromosome 11, consists of 1,965 nucleotides [10]. MIR210HG has been implicated in numerous human tumors. For instance, it facilitates breast cancer development through IGF2BP1-mediated m6 A modification [11]. MIR210HG facilitates ovarian cancer carcinogenesis by suppressing HIF- 1α degradation [12]. MIR210HG facilitates the progression of non-small cell lung cancer via direct regulation of the miR- 874/STAT3 axis [13]. Nevertheless, the involvement of MIR210HG in ESCC remains unexplored.
Recently, explorations utilizing signaling pathways and high-throughput sequencing data have offered novel insights into cancer research. In this research, MIR210HG expression was assessed in numerous cancers through pan-cancer analysis on the basis of the TCGA database. Furthermore, this analysis was validated by the GSE161533 data set and gene expression profiling interactive analysis (GEPIA) database. This research also assessed the involvement of MIR210HG in the prognosis and survival of individuals having esophageal squamous cancer. The biological functions of MIR210HG were investigated by enrichment analyses, encompassing gene set enrichment analysis (GSEA), Kyoto Encyclopedia of Genomes (KEGG), and Gene Ontology (GO). Moreover, validation was carried out using an external data set, GSE53624. The findings of the aforementioned analyses revealed the involvement of MIR210HG in the modulation of tumor metabolism and tumor immune-related signaling pathways. Moreover, the study investigated immune cell infiltration, tumor microenvironment (TME) characteristics, and immune checkpoint expression levels associated with MIR210HG. These results indicated the strong correlation between MIR210HG and the immune function of ESCC. Finally, various in vitro and in vivo analyses, encompassing flow cytometry, quantitative real-time fluorescence polymerase chain reaction (qRT-PCR), Western blotting, migration, invasion, and colony formation, monodansylcadaverine (MDC) staining, and xenograft experiments, were performed for the elucidation of the involvement of MIR210HG in ESCC, providing a new theoretical reference for ESCC treatment. In summary, the results of this research present a basis for designing current and future treatment approaches for ESCC.
Methodology
Retrieval of data
The Transcripts Per Million (TPM) sequencing data from the ESCC data set were processed after being retrieved through the TCGA database (https://www.example.com). Sample clinical information from 93 cases of ESCC was obtained for further analysis, following the exclusion of data samples lacking prognosis information and duplicates (Table S1). The data sets, such as GSE161533 (comprising 28 normal esophageal squamous epithelial samples and 28 ESCC samples) [14] and GSE53624 (encompassing 119 normal esophageal epithelial and 119 ESCC samples), were accessed at the GEO database (https://www.ncbi.nlm.nih.gov/geo/) [15]. Furthermore, patient clinical information included in the GSE53624 data set was retrieved (Table S2).
Survival analysis
The Kaplan–Meier plots were drawn on the basis of TCGA–ESCC data using the survival ROC package to assess whether MIR210HG expression was associated with survival among patients with ESCC [16]. Validation was conducted using the external data set GSE53624. In addition, MIR210HG localization on chromosomes was performed using the RCircos package [17].
Functional enrichment analysis
The most relevant 200 genes of MIR210HG were transferred to the Database for Annotation, Visualization and Integrated Discovery (DAVID) (https://david.ncifcrf.gov/) database for analysis and annotation using the GSE53624 and TCGA–ESCC data sets. Enrichment analysis results for GO and KEGG were obtained using the official gene names as identifiers and Homo sapiens as the species. Finally, the top 7 enrichment results were presented in descending order of P value (P < 0.05).
Gene set enrichment analysis
Utilizing the TCGA–ESCC data set, the samples were categorized into low-expression and high-expression groups to assess the impact of gene expression on tumors. Subsequently, GSEA was conducted to analyze gene expression patterns [18]. GSEA, a computational approach, is commonly utilized to assess statistical differences between two biological states exhibited by a specific gene set. It estimates pathway changes and bioprocess activity within expression data set samples. In addition, the GSE53624 data set was employed for validation.
ESTIMATE immunoreactivity analysis
Estimation of STromal and Immune cells in MAlignant Tumour tissues using Expression data (ESTIMATE) determines the content of tumor cells along with infiltrating stromal and immune cells by transcriptional profiling of cancer samples. It can also calculate the abundance or proportion of immune, tumor, and stromal cells in tumor tissues associated with the tumor microenvironment (TME). Based on the TCGA–ESCC data set, immune and stromal scores in ESTIMATE were obtained for all patients using the ESTIMATE package for R [19]. In addition, the GSE53624 data set was used for validation.
Immune cell infiltration analysis
Immune infiltration analysis was carried out utilizing CIBERSORTx to assess the association between immune cells and MIR210HG. The CIBERSORT package [20] utilizes the linear support vector regression principle for the deconvolution of the transcriptome expression matrix, thus evaluating the abundance and composition of immune cells within mixed cells. Utilizing the CIBERSORT algorithm, samples with an output P < 0.05 were filtered based on the gene expression feature set of 22 immune cell subtypes, thereby estimating the abundance and composition of immune cells within mixed cells. Considering the growing significance of immunotherapy in oncology, several common immune checkpoints were analyzed.
Cell line culture
Four human ESCC cell lines (Eca9706, KYSE150, KYSE510, and KYSE30) with different degrees of differentiation along with a normal esophageal epithelial cell line (HEEC) were acquired from the cell bank of the Shanghai Research Institute of the People's Republic of China. In a humidified incubator, Eca9706, KYSE150, KYSE510, and KYSE30 cells underwent cultivation at 37 °C with 5% CO2 in RPMI- 1640 medium (Kibbutz Beit HaEmek BI) enriched with 10% fetal bovine serum (FBS). HEEC cells were then cultivated in a humidified incubator at 37 ℃ with 5% CO2 in a DMEM medium (Gibco) containing 10% FBS. All cells were passaged no more than 15 times.
Cellular lentiviral transduction and processing
Negative control lentiviral vectors (Vector) and MIR210HG-overexpressing lentiviral vectors (OE-MIR210HG) were obtained from GenePharma. Lentiviral vectors were transduced into KYSE30 and KYSE150 cells, respectively, and subsequently screened utilizing a medium containing 5 µg/mL puromycin for obtaining stably transfected cell lines. Cells in the respective subgroups were treated with PFT-α (p53 inhibitor; MedChemExpress 40 μmol/L) following the provided protocol.
Quantitative real-time fluorescence polymerase chain reaction
Cells underwent total RNA extraction utilizing RNAiso Plus (TaKaRa, Dalian, China). Subsequently, reverse transcription was conducted utilizing the RT Reagent Kit (TaKaRa, Dalian, China) to produce cDNA. Subsequently, SYBR Green Real-Time PCR Master Mix (TaKaRa, Dalian, China) was employed to perform quantitative PCR (qPCR), with GAPDH serving as the endogenous reference gene. The following are the PCR primers employed in this research:
MIR210HG:
Forward primer: 5'-TGTTCCCTTTGTGTGCTCCAG- 3'.
Reverse primer: 5'-GCCCTAGATCATGGGGGTCTT- 3'.
GAPDH:
Forward primer: 5'-AGAGCCTCGCCTTTGCCGATCC- 3'.
Reverse primer: 5'-ATACACCCGCTGCTCCGGGGTC- 3'.
Partial separation of nucleocytoplasmic RNA
The Cytoplasmic and nuclear rna purification kit (Norgen, Cat. No 21000, Canada) was employed to separate cytoplasmic and nuclear RNA from KYSE30 and KYSE150 cells. qPCR was conducted to assess the relative amount of MIR210HG in the cytoplasm and nucleus of the cells. U6 and GAPDH were utilized as the endogenous controls in the nucleus and cytoplasm, respectively, to elucidate the corresponding proportion of MIR210HG expression in the nucleus and cytoplasm. The U6 primer sequences are mentioned below:
Forward primer: 5'-CTCGCTTCGGCAGCACA- 3'.
Reverse primer: 5'-AACGCTTCACGAATTTGCGT- 3'.
Cell invasion and migration assays
The Transwell chamber (24-well) is composed of lower and upper compartments, divided by a polycarbonate membrane featuring an 8 μm opening. Cell invasion was detected using a matrigel (Corning)-coated polycarbonate membrane, while cell migration was assessed by utilizing an uncoated polycarbonate membrane. A solution of exponential phase lentiviral vectors-transfected cells was introduced into the upper compartment of the aforementioned chamber, whereas a medium enriched with 20% FBS was introduced to the lower compartment. After 12 h of incubation with 5% CO2 at 37 °C, cells were allowed to traverse the polycarbonate membrane, underwent fixation with 4% paraformaldehyde, and then subjected to crystal violet staining. The number of stained cells passing through the polycarbonate membrane into the lower compartment was counted under an inverted microscope.
Colony formation assay
Different groups of stably transfected cells were seeded into six-well plate cell culture plates (1000 per well) and incubated for 12 days. The formed cell colonies were subjected to fixation by 4% paraformaldehyde and subsequently underwent 0.1% crystal violet staining. Ultimately, the number of colonies exceeding 50 cells was counted.
Wound healing assay
Each group of cells was inoculated uniformly (density: 4 × 105 cells/well) in 6-well plates and subsequently cultured in a cell culture incubator. Once the cells adhered to the wall and reached a density of 90–95%, a 200 μL tip was employed for the creation of a straight linear scratch on the cell monolayer. After gently washing the detached cells with PBS, a serum-free medium was introduced, and the scratches were photographed at 0 h under a microscope to calculate the initial wound area. Subsequently, the cells were put back into the incubator to continue incubation for 24 h. After this period, the wound area was photographed under the microscope and then measured. ImageJ was utilized to quantify the wound area at the same location. The following formula was utilized to assess the cell migration rate:
Western blotting analysis
The lysis of cells and tissues was carried out using radioimmunoprecipitation assay (RIPA) lysis buffer enriched with phenylme thylsulfonyl fluoride (PMSF) and phosphatase inhibitors for the extraction of proteins. Sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS–PAGE) was employed for the separation of the protein samples. Subsequently, they were transferred onto a polyvinylidene difluoride (PVDF) membrane. After that, 5% BSA was employed to block the membrane for 1 h. Afterward, the membrane underwent overnight incubation at 4 °C with primary antibodies, including anti-Bax (HUABIO Catalog# ET1603 - 34 Dilution ratio 1:20000), anti-Bcl- 2 (HUABIO Catalog# ET1702 - 53 Dilution ratio 1:2000), anti-Beclin- 1 (Proteintech Cat No: 66665 - 1-Ig Dilution ratio 1:2000), anti-C-caspase3 (Abcam ab2302 Dilution ratio 1:100), anti-GAPDH (Abcam ab181602 Dilution ratio 1:10000), anti-LC3B-I/II (Abcam ab192890 Dilution ratio 1:2000), anti-P53 (HUABIO Catalog# ET7107 - 33 Dilution ratio 1:1000), anti-P62 (HUABIO Catalog# HA721171 Dilution ratio 1:5000), anti-AMPKa (Abcam ab207442 Dilution ratio 1:1000), and anti-P-AMPKa (Abcam ab314032 Dilution ratio 1:1000). Subsequently, the membrane was incubated again for 1 h at room temperature with the specified secondary antibodies and enhanced chemiluminescence (ECL) was utilized for visualizing the membrane. The relative expression of target proteins was assessed with GAPDH serving as an internal reference.
EdU cell proliferation assay
Stably transfected ESCC cells in the logarithmic growth phase were placed into 96-well plates (5000 cells/well). Following this, the plates underwent incubation for 24 h. Subsequently, the cells were exposed to EdU for a duration of 3 h. After that, they were labeled, washed, and fixed with paraformaldehyde for 30 min. Following fixation, they underwent destaining through osmotic decolorization for 10 min with 0.5% Triton X- 100. Ultimately, 4',6-diamidino- 2-phenylindole (DAPI) was employed for 30 min for staining the nuclei of the cells. Using a fluorescence microscope, the total number of cells and the number of EdU-positively stained cells were identified, and corresponding images were captured. The formula to calculate the cell proliferation rate is mentioned below:
Monodansylcadaverine (MDC) staining
Cellular autophagy was assessed through MDC staining. The cells from various treatment groups were washed once with 1xWash buffer, followed by incubation with MDC staining reagent (Solarbio) for 30 min in the dark. Subsequently, the cells were rinsed thrice with 1xWash buffer. Finally, the cells were covered with a collection buffer. MDC staining was observed, photographed, and counted using a fluorescence microscope. The formula to calculate the autophagy rate is mentioned below:
Apoptosis detection via flow cytometry
Apoptosis was detected via flow cytometry utilizing the PE Annexin V Apoptosis Detection Kit I (BD Biosciences). Test cells were prepared into cell suspensions by trypsin digestion. Samples were assessed by flow cytometry after double staining with P-phycoerythrin (PE) and 7-aminoactinomycin D (7-AAD).
ESCC xenograft model
A total of 10 female BALB/C mice (weight: 18–22 g and age: 4–6 weeks) were procured from Beijing Charles River Laboratory Animal Technology Co., Ltd. These mice underwent random allocation into two groups (5 mice/group). KYSE150 cells (100 μL, 7.5 × 105 cells), transfected with lentivirus from Vector and OE-MIR210HG, were administered into the axilla (right) of the mice by subcutaneous injection. The mice were kept at 20–26 °C in a specific pathogen-free (SPF) facility with a humidity range between 40 and 60%.When the transplanted tumor reached 100 mm3, its volume and size were assessed and recorded every 2 days. Eighteen day post-inoculation, all mice were euthanized by cervical dislocation. Subsequently, the subcutaneous grafted tumors were completely excised, and their weights were measured. Furthermore, the expression levels of relevant genes and proteins in the isolated tumor tissues were assessed using Western blotting analysis, qRT-PCR, and immunohistochemistry. Animal experiments were conducted in accordance with the ARRIVE guidelines, and the study protocols for animal experiments were reviewed and approved by the Ethical Review Committee of Shandong Provincial hospital affiliated to Shandong First Medical University.
Statistical analyses
All data processing and analyses were done through R software (version 4.1.1). For comparisons between two groups of continuous variables, statistical significance of normally distributed variables was estimated by independent Student t tests and differences between non-normally distributed variables were analysed by Mann–Whitney U tests (i.e., Wilcoxon rank sum tests). The chi-square test or Fisher exact test was used to compare and analyse the statistical significance between categorical variables in the two groups. The survival package of R was used to perform survival analyses, the Kaplan–Meier survival curves were used to show differences in survival, and the log-rank test (log-rank test) was used to assess the significance of the differences in the survival times of the two groups of patients. Both univariate and multivariate Cox analyses were based on the survival R package, and lasso analysis was based on the glmnet R package. All statistical P values were two-sided, with p < 0.05 considered statistically significant.
Results
Downregulated expression of MIR210HG in ESCC
Chromosomal localization analysis conducted with the RCircos package showed that the gene MIR210HG was located on chromosome 11 (Fig. 1A). According to the analysis conducted using the GEPIA database, the expression level of MIR210HG exhibited a significant rise in 286 normal tissues within the context of ESCC than in 182 cancer tissues (P < 0.01, Fig. 1B). The similar outcome was acquired from the analysis of the external data set GSE161533 (Fig. 1C), reinforcing the upregulation of MIR210HG in normal tissues in ESCC. Patients with ESCC in the TCGA database were categorized into high- and low-MIR210HG expression groups to determine whether MIR210HG expression impacts patient survival. Kaplan–Meier survival analysis depicted that individuals in the high-MIR210HG expression group exhibited a higher survival rate (Fig. 1D). Subsequently, the external data set GSE53624 validated the above outcome (Fig. 1E). These outcomes propose that high expression of MIR210HG may improve the prognosis of patients with ESCC. Pearson correlation analysis was employed to gain further insights into MIR210HG biological function in the TCGA–ESCC data set. The 300 genes with the strongest positive correlations were screened (Table S3). Subsequently, a heat map was generated to visualize the top 20 genes positively associated with MIR210HG (Fig. 1F). Similarly, Pearson correlation analysis was performed utilizing GSE161533 to screen the 300 genes with the strongest positive correlation (Table S4), followed by heat map plotting (Fig. 1G).
Expression levels of MIR210HG in ESCC. A Chromosomal localization of the MIR210HG gene. B Expression data for MIR210HG in the GEPIA database. C Expression of MIR210HG in tumor vs. normal tissues in the GSE161533 data set. D Overall survival rate of ESCC patients in TCGA, estimated by Kaplan–Meier. E Overall survival rate of ESCC patients in GSE53624, estimated by Kaplan–Meier. F Heatmap showing the top 20 genes positively correlated with MIR210HG in TCGA-ESCC. G Heatmap showing the top 20 genes positively correlated with MIR210HG in GSE53624. *p < 0.05, **p < 0.01, ***p < 0.001
Enrichment and functional analyses of MIR210HG-associated genes in ESCC
To explore the biological functions linked to MIR210HG, Pearson correlation analysis was conducted on the genes from the external data set GSE53624 and the TCGA database that exhibited the strongest correlation with MIR210HG (top 300). Subsequently, these genes were subjected to GSEA, KEGG, and GO analyses. In the TCGA database, the most correlated biological processes with MIR210HG encompassed response to hypoxia, epidermis development, and keratinization (Fig. 2A). The cellular component, mostly correlated with MIR210HG, was extracellular exosome (Fig. 2B), whereas the most correlated molecular function with MIR210HG was protein binding (Fig. 2C). The most correlated signaling pathways with MIR210HG included the P53 and HIF- 1 signaling pathways (Fig. 2D). Similar signaling pathways, biological processes, molecular functions, and cellular components correlated with MIR210HG were observed in the external data set GSE53624, aligning with the findings from the TCGA database (Fig. 2E–H). Significant results were observed in GSEA, and external data set validation of the P53 signaling pathway confirmed these findings (Fig. 2I, J). These outcomes propose that MIR210HG may exhibit its inhibitory impact on ESCC by activating the P53 signaling pathway.
Functional clustering of MIR210HG-related genes. A–C Biological processes (BP), cellular components (CC), and molecular functions associated with MIR210HG-related genes in the TGGA database. D Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of MIR210HG-related genes. E GSEA analysis of upregulated genes related to MIR210HG in the TGGA database. F GSEA analysis of downregulated genes related to MIR210HG in the TGGA database. E–G Biological processes (BP), cellular components (CC), and molecular functions of MIR210HG-related genes in the external data set GSE53624. H KEGG analysis of MIR210HG-related genes. I Correlation of MIR210HG-related genes with the P53 signaling pathway in the TCGA database. J Correlation of MIR210HG-related genes with the P53 signaling pathway in the external data set GSE53624
Relationship between immunity and MIR210HG
The analysis was aimed at examining the relationship between the tumor immune microenvironment and MIR210HG expression levels. As per the TCGA–ESCC database, the estimated, immune, and stromal scores were assessed per the MIR210HG expression (Fig. 3A), revealing a substantial difference (P < 0.05). The external data set GSE53624 exhibited similar results (Fig. 3B), confirming a notable impact of MIR210HG on the immune status within the TME of ESCC. As shown in Fig. 3C, CIBSCORT analysis depicted a significant correlation between MIR210HG expression and the infiltration of M0 macrophages, CD4 + T cells, M2 macrophages, NK cells, mast cells, and other immune cells in the TCGA–ESCC data set (P < 0.05), which was further validated in the GSE53624 data set (Fig. 3E). In this research, the correlation between the MIR210HG expressions and several immune checkpoints was investigated, considering the current widespread usage of immune checkpoint inhibitors in tumor immunotherapy (Fig. 3D, F). These results highlight a possible important role of MIR210HG in tumor immunity.
Relationship between MIR210HG and immunity. A Evaluation of immune cell stroma and abundance in TCGA-ESCC. B Evaluation of immune cell stroma and abundance in the data set GSE53624. C CIBERSORT assessment of immune cell composition between two groups in TCGA-ESCC. D Relationship of MIR210HG with conventional immune checkpoints in TCGA-ESCC. E CIBERSORT assessment of immune cell composition between two groups in the data set GSE53624. F Relationship of MIR210HG with conventional immune checkpoints in the data set GSE53624
MIR210HG is lowly expressed in ESCC cells and overexpression of MIR210HG suppresses the proliferative, invasive, and migratory capabilities of cancer cell
The qRT-PCR was employed to examine the MIR210HG expression levels in Eca9706, HEEC, KYSE150, KYSE510, and KYSE30 cell lines. The MIR210HG expression exhibited a significant reduction in cell lines, including Eca9706, KYSE510, KYSE150, and KYSE30 compared with that in HEEC, especially in KYSE150 and KYSE30 (Fig. 4A). Thus, these two aforementioned cell lines were selected for further experiment. The qRT-PCR was conducted to identify the overexpression effectiveness of MIR210HG in cell lines, such as KYSE30 and KYSE150 (Fig. 4B), presenting significant differences between the two. For subsequent clarification of the MIR210HG action mechanism in ESCC, the nuclear and cytoplasmic expression fractions of MIR210HG in these two cell lines were determined. MIR210HG exhibited significant enrichment in the nuclear fraction, indicating that MIR210HG may be involved in ESCC development through the nuclear fraction (Fig. 4C, D).
Expression of MIR210HG. A qRT-PCR detection of MIR210HG expression in esophageal squamous cell carcinoma cell lines (KYSE150, KYSE510, Eca9706 and KYSE30) and normal esophageal epithelial cell line (HEEC). B qRT-PCR detection of overexpression efficiency of MIR210HG in KYSE150 and KYSE30 cells. C, D Localization of MIR210HG in nuclear and cytoplasmic RNA fractions of KYSE150 and KYSE30 cells. E–G Effect of MIR210HG on the migration of KYSE150 and KYSE30 cells in wound healing assays. H–J Effect of MIR210HG on the proliferation of KYSE150 and KYSE30 cells in colony formation assays. *p < 0.05, **p < 0.01, ***p < 0.001. Scale bars: 100 μm
The wound healing assays were conducted to examine the impact of MIR210HG on the migration of the two above-mentioned cell lines. These cell lines reported a significant reduction in their healing rates due to MIR210HG overexpression (Fig. 4E–G). Colony formation assays exhibited that MIR210HG overexpression significantly diminished the proliferation capacity of KYSE30 and KYSE150 cell lines (Fig. 4H–J). Based on the findings of invasion and migration assays, MIR210HG overexpression significantly hindered the invasive and migratory capabilities of these cells compared to those in the controls (Fig. 5A, B). EdU cell proliferation assay also revealed that MIR210HG overexpression suppressed the proliferative ability of ESCC cell lines (Fig. 5C, D). These outcomes depicted that MIR210HG overexpression inhibited the invasive, proliferative, and migratory abilities of ESCC cells in vitro.
Overexpression of MIR210HG significantly inhibits the proliferation, invasion and migration of KYSE150 and KYSE30 cells. A, B Compared to control, overexpression of MIR210HG significantly inhibits the migration and invasion of esophageal squamous carcinoma cells. C, D EdU proliferation assays show that overexpression of MIR210HG can inhibit the proliferative capacity of ESCC lines. *p < 0.05, **p < 0.01, ***p < 0.001. Scale bars: 100 μm. ESCC, esophageal squamous carcinoma cell
Association of MIR210HG with the P53 signaling pathway and induction of apoptosis and autophagy in ESCC cells
GSEA enrichment analysis exhibited a positive correlation between the P53 signaling pathway and MIR210HG. In the Western blotting validation, MIR210HG overexpression in cell lines, including KYSE30 and KYSE150 resulted in a significant upregulation of intracellular P53 protein levels. This suggests the P53 signaling pathway activation, aligning with the findings of the above bioinformatics analysis (Fig. 6A–C). A significant number of scientific investigations have confirmed the close relationship between P53 and apoptosis and autophagy in cancer cells. Moreover, the downstream molecule of P53, AMPK, has been identified as a key activator of autophagy. Therefore, PFT-α, an inhibitor of the transcriptional activity of the P53 protein, was utilized to assess whether P53 exhibited any involvement concerning the influence of MIR210HG on the biological process of ESCC cells. In the Western blotting analysis, the cells were grouped into Vector, OE-MIR210HG, Vector + PFT-α, and OE-MIR210HG + PFT-α. In comparison with the cells in the Vector group, the level of proteins in Bax, Beclin- 1, C-caspase3, LC3B-II/I, and P-AMPKa exhibited substantial elevation and the protein levels of P62 and Bcl- 2 exhibited a reduction in the cells after MIR210HG overexpression. PFT-α effectively reversed the above-mentioned protein expression alterations in cells overexpressing MIR210HG (Fig. 6D–F). Flow cytometry further verified that overexpression of MIR210HG promoted apoptosis in KYSE30 and KYSE150 cells. Moreover, apoptosis induced by MIR210HG overexpression in ESCC cells was significantly attenuated after PFT-α treatment (Fig. 7A, B). According to subsequent MDC staining experiments, MIR210HG overexpression significantly increased cellular autophagy compared to cells in the Vector group. In addition, PFT-α treatment effectively attenuated the autophagy effect induced by MIR210HG overexpression in ESCC cells (Fig. 7C–F). Similarly, we again confirmed by colony assay that overexpression of MIR210HG significantly attenuated the proliferative capacity of KYSE150 and KYSE30 cells, and this result was effectively reversed using PFT-α (Fig. 8). The above results suggest that MIR210HG overexpression in ESCC cells triggers apoptosis through the P53 signaling pathway activation, which activates the downstream AMPK and significantly increases the phosphorylation level of AMPKα (Thr172) (increased expression of P-AMPKa), thereby inducing cell autophagy, and then inhibited the proliferation of esophageal squamous carcinoma cells.
Overexpression of MIR210HG promotes the p53 signaling pathway and is related to autophagy and apoptosis. A–C Changes in P53 protein expression in ESCC cells after overexpression of MIR210HG detected by western blot. D–F Changes in the expression of apoptosis and autophagy-related proteins in different subgroups of ESCC cells were detected by western blot in Vector, OE-MIR210HG, Vector + PFT-α, and OE-MIR210HG + PFT-α, respectively, and PFT-α was an inhibitor of the transcriptional activity of P53 protein. Error bars indicate the mean ± standard deviation of three independent experiments, ns (non-significant), *p < 0.05, **p < 0.01, ***p < 0.001
Overexpression of MIR210HG induces autophagy and apoptosis in ESCC cells. A, B Flow cytometry analysis demonstrated that overexpression of MIR210HG promoted apoptosis in KYSE150 and KYSE30 cells, and that the addition of PFT-α reversed this apoptotic enhancement effect. C–F Overexpression of MIR210HG resulted in increased green fluorescence of MDC in KYSE150 and KYSE30 cells, indicating enhanced autophagy, and the addition of PFT-α reversed this autophagy-enhancing effect. Error bars indicate the mean ± standard deviation of three independent experiments. *p < 0.05, **p < 0.01, ***p < 0.001. Scale bars: 100 μm
Colony assay confirms activation of p53 signaling pathway in ESCC cells after overexpression of MIR210HG to inhibit cell proliferation. A, B Overexpression of MIR210HG inhibited the proliferation of KYSE150 cells, and the addition of PFT-α reversed this proliferation inhibitory effect. C, D Overexpression of MIR210HG inhibited KYSE30 cell proliferation, and the addition of PFT-α reversed this proliferation inhibitory effect. Error bars indicate the mean ± standard deviation of three independent experiments. **p < 0.01
MIR210HG overexpression suppresses the xenograft tumor growth
For further investigation of the influence of MIR210HG overexpression on the proliferation of ESCC cells in vivo, KYSE150 cells underwent transfection with Vector and OE-MIR210HG. These cells were then inoculated subcutaneously into nude mice to develop ESCC-grafted tumor models. In comparison with the Vector group, OE-MIR210HG significantly reduced the tumor growth rate (Fig. 9A–C). Total cellular RNA was extracted from some transplanted tumor tissues of the Vector group and the OE-MIR210HG group for qRT-PCR. MIR210HG was substantially overexpressed in the OE-MIR210HG group than in the Vector group (Fig. 9D). After that, mouse transplanted tumor tissues were randomly selected from the Vector and OE-MIR210HG groups for Ki67 immunohistochemical staining. The immunohistochemical scores of the two groups exhibited significant differences, with the Ki67 expression in the OE-MIR210HG group notably reduced in comparison with the Vector group (Fig. 9E, F). Furthermore, Western blotting analysis was conducted on the tumor specimens of the two mice groups to validate the aforementioned analysis. MIR210HG overexpression in ESCC cells substantially elevated the expression of Bax, Beclin- 1, C-caspase3, and P53. Moreover, it significantly decreased the expression of P62, thereby promoting apoptosis and autophagy (Fig. 9G, H).
Overexpression of MIR210HG inhibits xenograft tumor growth. A–C Tumor volume size measurements were performed during subcutaneous inoculation of esophageal squamous carcinoma cells in nude mice for 18 days, and nude mice were necropsied after 18 days, and the tumors were exfoliated intact according to subgroups, weighed, and statistically counted; overexpression of MIR210HG significantly reduced the growth rate of the tumors as compared to the control group. D MIR210HG expression was detected by qRT-PCR in tumor specimens from mice in the Vector and OE-MIR210HG groups, respectively. E, F Immunohistochemical Ki67 staining was performed on tumor specimens from mice in the Vector and OE-MIR210HG groups, respectively, and scores were tallied. G, H Protein expression of P53, Bax, C-caspase3, Beclin- 1 and P62 in tumor specimens of mice in the Vector and OE-MIR210HG groups were further detected by Western blot, respectively, and statistically analyzed. *p < 0.05, **p < 0.01, ***p < 0.001
Discussion
ESCC presents as a severe and life-threatening malignant tumor characterized by high prevalence and easy metastasis to other sites in the early stage [21]. The limited effect of conventional antitumor drugs on ESCC leads to a high mortality rate and unpromising therapeutic outcomes [22]. EC can be detected early with endoscopy. However, the 5-year overall survival rate of patients is still less than 20%, necessitating new treatment modalities [23]. LncRNAs are closely related to human diseases, especially to malignant tumors [24,25,26]. Previous investigations have identified a strong correlation between cancer and MIR210HG. For example, MIR210HG facilitates the proliferative, migratory, and invasive abilities of lung cancer cells by suppressing the transcription of SH3GL3 [27]. In addition, MIR210HG promotes the malignant proliferation of glioma cells [28]. Nevertheless, the involvement of MIR210HG in ESCC is yet to be known.
In the current research, elevated expression of MIR210HG inhibited ESCC and was closely associated with patient survival. To elucidate the underlying mechanism of MIR210HG affecting ESCC, the 300 most correlated genes were screened and subjected to enrichment analyses. According to the findings, a positive correlation was observed between MIR210HG and the P53 signaling pathway. The TP53 gene, also known as the “guard gene”, stands among the most important cancer suppressor genes within the human body [29]. P53 activation promotes DNA repair or the controlled death of abnormal cells, thus preventing the onset and progression of cancer [30]. In addition, P53 can eliminate tumor immune evasion [31].
P53 exerts a crucial influence on numerous pathways, encompassing autophagy, cellular metabolism, ferroptosis, and metabolism [32]. In this study, when MIR210HG exhibited elevated expression in ESCC cells, the expression of P53 was significantly elevated, the phosphorylation levels of AMPKα (Thr172) were significantly increased, along with a substantial elevation observed in the expression of autophagy-associated proteins, such as LC3B-II and Beclin- 1, and a significant downregulation in the expression level of P62 proteins, suggesting enhanced autophagy levels in tumor cells. Subsequently, a p53 inhibitor (PFT-α) was used to validate whether the changes at autophagy levels in ESCC cells were induced by MIR210HG through P53 regulation. The addition of PFT-α to ESCC cells overexpressing MIR210HG effectively suppressed the elevated autophagy level. Similarly, we conducted a similar exploration for apoptosis. We further confirmed that overexpression of MIR210HG inhibited the proliferation of esophageal squamous carcinoma cells by activating the p53 signaling pathway through colony assay. The above results indicate that overexpression of MIR210HG in esophageal squamous carcinoma cells activated the p53 signaling pathway and then activated the downstream AMPK, and the phosphorylation level of AMPKα (Thr172) (expression of P-AMPKa) was significantly increased, which induced apoptosis and autophagy, and then inhibited the proliferation of esophageal squamous carcinoma cells. Finally, the above findings were further validated in mouse specimens. As per the assessed literature, this is the first demonstration that MIR210HG can influence autophagy and apoptosis in ESCC through the P53 signaling pathway. The in-depth study of the mechanism underlying the interaction between MIR210HG and autophagy and apoptosis promises to offer fresh insights and avenues for ESCC treatment.
The TME, encompassing various immune and stromal cells, exerts a crucial impact on cancer progression and treatment response [33, 34]. Treg cells can suppress tumor immunity during tumor progression. Therefore, the heightened infiltration of Treg cells in TME often predicts an unfavorable prognosis [35]. In this study, MIR210HG low-expression group exhibited more enriched immunosuppressive cells, encompassing Treg cells, which generate an immunosuppressive microenvironment and inhibit the tumor clearance process. At present, monoclonal antibodies targeting immune checkpoint molecules have made notable advancements in the field of cancer treatment. CCL2 and PD-L1 have shown promising outcomes in ESCC treatment [36]. In the present study, MIR210HG expression also showed significant differences among different immune checkpoints.
This study holds significant clinical significance, and strict screening criteria were applied throughout the analysis process. However, this research still encounters several limitations. First, the sample size was relatively small, necessitating a larger sample size and additional validation experiments. Finally, while this study has shed new light on the potential role of MIR210HG in ESCC, we acknowledge that we have not yet investigated the specific mechanisms by which MIR210HG affects the P53 signalling pathway. This is a crucial aspect that requires further investigation, as understanding these mechanisms could pave the way for novel therapeutic strategies targeting MIR210HG in ESCC. Therefore, further exploration is necessary.
Conclusion
In summary, this research revealed that MIR210HG exhibits low expression in ESCC. In addition, this low expression of MIR210HG demonstrated a strong correlation with the survival of patients with ESCC. MIR210HG overexpression suppressed the proliferative, invasive, and migratory capacities of ESCC cells. Highly expressed MIR210HG could promote autophagy and apoptosis of ESCC cells via the P53 signaling pathway. Furthermore, immune cell infiltration analyses were performed on individuals having ESCC with low and high expression of MIR210HG. Overall, the findings of this research present a theoretical foundation for individualized treatment and management of patients with ESCC.
Availability of data and materials
No datasets were generated or analysed during the current study.
References
Huang FL, Yu SJ. Esophageal cancer: risk factors, genetic association, and treatment. Asian J Surg. 2018;41:210–5.
Abnet CC, Arnold M, Wei WQ. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology. 2018;154:360–73.
Zeng H, Zheng R, Zhang S, Zuo T, Xia C, Zou X, Chen W. Esophageal cancer statistics in China, 2011: estimates based on 177 cancer registries. Thorac Cancer. 2016;7:232–7.
Lin DC, Dinh HQ, Xie JJ, Mayakonda A, Silva TC, Jiang YY, Ding LW, He JZ, Xu XE, Hao JJ, Wang MR, Li C, Xu LY, Li EM, Berman BP, Phillip KH. Identification of distinct mutational patterns and new driver genes in oesophageal squamous cell carcinomas and adenocarcinomas. Gut. 2018;67:1769–79.
Khorkova O, Stahl J, Joji A, Volmar CH, Wahlestedt C. Amplifying gene expression with RNA-targeted therapeutics. Nat Rev Drug Discov. 2023;22:539–61.
Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, Loewer A, Ziebold U, Landthaler M, Kocks C, le Noble F, Rajewsky N. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495:333–8.
Feng Q, Zhang H, Yao D, Chen WD, Wang YD. Emerging role of non-coding RNAs in esophageal squamous cell carcinoma. Int J Mol Sci. 2019;21:258.
Huang X, Liu C, Li H, Dai T, Luo G, Zhang C, Li T, Lu M. Hypoxia-responsive lncRNA G077640 promotes ESCC tumorigenesis via the H2AX-HIF1alpha-glycolysis axis. Carcinogenesis. 2023;44:383–93.
Tang J, Xu H, Liu Q, Zheng J, Pan C, Li Z, Wen W, Wang J, Zhu Q, Wang Z, Chen L. LncRNA LOC146880 promotes esophageal squamous cell carcinoma progression via miR-328-5p/FSCN1/MAPK axis. Aging (Albany NY). 2021;13:14198–218.
Wang Y, Li W, Chen X, Li Y, Wen P, Xu F. MIR210HG predicts poor prognosis and functions as an oncogenic lncRNA in hepatocellular carcinoma. Biomed Pharmacother. 2019;111:1297–301.
Shi W, Tang Y, Lu J, Zhuang Y, Wang J. MIR210HG promotes breast cancer progression by IGF2BP1 mediated m6A modification. Cell Biosci. 2022;12:38.
Liu P, Huang H, Qi X, Bian C, Cheng M, Liu L, Xue L, Zhao X, Yi T, Quan Y. Hypoxia-induced LncRNA-MIR210HG promotes cancer progression by inhibiting HIF-1alpha degradation in ovarian cancer. Front Oncol. 2021;11:701488.
Bu L, Zhang L, Tian M, Zheng Z, Tang H, Yang Q. LncRNA MIR210HG facilitates non-small cell lung cancer progression through directly regulation of miR-874/STAT3 Axis. Dose Response. 2020;18:1559325820918052.
He X, Wang J, Yu H, Lv W, Wang Y, Zhang Q, Liu Z, Wu Y. Clinical significance for diagnosis and prognosis of POP1 and its potential role in breast cancer: a comprehensive analysis based on multiple databases. Aging (Albany NY). 2022;14:6936–56.
Li J, Chen Z, Tian L, Zhou C, He MY, Gao Y, Wang S, Zhou F, Shi S, Feng X, Sun N, Liu Z, Skogerboe G, Dong J, Yao R, Zhao Y, Sun J, Zhang B, Yu Y, Shi X, Luo M, Shao K, Li N, Qiu B, Tan F, Chen R, He J. LncRNA profile study reveals a three-lncRNA signature associated with the survival of patients with oesophageal squamous cell carcinoma. Gut. 2014;63:1700–10.
Mandrekar JN. Receiver operating characteristic curve in diagnostic test assessment. J Thorac Oncol. 2010;5:1315–6.
Zhang H, Meltzer P, Davis S. RCircos: an R package for Circos 2D track plots. BMC Bioinform. 2013;14:244.
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, Mesirov JP. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–50.
Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres-Garcia W, Trevino V, Shen H, Laird PW, Levine DA, Carter SL, Getz G, Stemke-Hale K, Mills GB, Verhaak RG. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612.
Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol. 2018;1711:243–59.
He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B. 2021;11:3379–92.
Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72:7–33.
Xu L, Wei XF, Li CJ, Yang ZY, Yu YK, Li HM, Xie HN, Yang YF, Jing WW, Wang Z, Kang XZ, Zhang RX, Qin JJ, Xue LY, Bi N, Chen XK, Li Y. Pathologic responses and surgical outcomes after neoadjuvant immunochemotherapy versus neoadjuvant chemoradiotherapy in patients with locally advanced esophageal squamous cell carcinoma. Front Immunol. 2022;13:1052542.
Alsharoh H, Chiroi P, Nutu A, Raduly L, Zanoaga O, Berindan-Neagoe I. Vinorelbine alters lncRNA expression in association with EGFR mutational status and potentiates tumor progression depending on NSCLC cell lines’ genetic profile. Biomedicines. 2023;11:3298.
Liu HT, Gao ZX, Li F, Guo XY, Li CL, Zhang H, Zhao RN, Liu Y, Shi DB, Zhu WJ, Gao P. LncRNA LY6E-DT and its encoded metastatic-related protein play oncogenic roles via different pathways and promote breast cancer progression. Cell Death Differ. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41418-023-01247-5.
Wang M, Pan W, Xu Y, Zhang J, Wan J, Jiang H. Microglia-mediated neuroinflammation: a potential target for the treatment of cardiovascular diseases. J Inflamm Res. 2022;15:3083–94.
Zou F, Zhang ZH, Zou SS, Zhuang ZB, Ji Q, Chang R, Cao JH, Wang B. LncRNA MIR210HG promotes the proliferation, migration, and invasion of lung cancer cells by inhibiting the transcription of SH3GL3. Kaohsiung J Med Sci. 2023;39:1166–77.
Yu Z, Che N, He Y, Zhang B. ceRNA network of lncRNA MIR210HG/miR-377-3p/LMX1A in malignant proliferation of glioma cells. Genes Genomics. 2022;44:1445–55.
Aning OA, Cheok CF. Drugging in the absence of p53. J Mol Cell Biol. 2019;11:255–64.
Baslan T, Morris JP, Zhao Z, Reyes J, Ho YJ, Tsanov KM, Bermeo J, Tian S, Zhang S, Askan G, Yavas A, Lecomte N, Erakky A, Varghese AM, Zhang A, Kendall J, Ghiban E, Chorbadjiev L, Wu J, Dimitrova N, Chadalavada K, Nanjangud GJ, Bandlamudi C, Gong Y, Donoghue MTA, Socci ND, Krasnitz A, Notta F, Leach SD, Iacobuzio-Donahue CA, Lowe SW. Ordered and deterministic cancer genome evolution after p53 loss. Nature. 2022;608:795–802.
Zhou X, Singh M, Sanz Santos G, Guerlavais V, Carvajal LA, Aivado M, Zhan Y, Oliveira MMS, Westerberg LS, Annis DA, Johnsen JI, Selivanova G. Pharmacologic activation of p53 triggers viral mimicry response thereby abolishing tumor immune evasion and promoting antitumor immunity. Cancer Discov. 2021;11:3090–105.
Hernandez Borrero LJ, El-Deiry WS. Tumor suppressor p53: biology, signaling pathways, and therapeutic targeting. Biochim Biophys Acta Rev Cancer. 2021;1876:188556.
Pitt JM, Marabelle A, Eggermont A, Soria JC, Kroemer G, Zitvogel L. Targeting the tumor microenvironment: removing obstruction to anticancer immune responses and immunotherapy. Ann Oncol. 2016;27:1482–92.
Kang J, La Manna F, Bonollo F, Sampson N, Alberts IL, Mingels C, Afshar-Oromieh A, Thalmann GN, Karkampouna S. Tumor microenvironment mechanisms and bone metastatic disease progression of prostate cancer. Cancer Lett. 2022;530:156–69.
Izzi V, Davis MN, Naba A. Pan-cancer analysis of the genomic alterations and mutations of the matrisome. Cancers (Basel). 2020;12:2046.
Ko KP, Huang Y, Zhang S, Zou G, Kim B, Zhang J, Jun S, Martin C, Dunbar KJ, Efe G, Rustgi AK, Nakagawa H, Park JI. Key genetic determinants driving esophageal squamous cell carcinoma initiation and immune evasion. Gastroenterology. 2023;165(613–628):e620.
Acknowledgements
We thank Bullet Edits Limited for the linguistic editing and proofreading of the manuscript.
Funding
This study is supported by the Key Research and Development Program of Shandong Province (No. 2017GSF218096).
Author information
Authors and Affiliations
Contributions
Jianyu Wang conceived and designed the study; Zhenhu Zhang performed data analysis; Liang Song and Xiangyan Liu contributed analysis tools; Xiaopeng He wrote the paper.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
The study was approved by the Ethical Review Committee of Shandong Provincial hospital affiliated to Shandong First Medical University (approval number: No. NSFC 2024 - 613).
Consent for publication
Not applicable.
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.
Supplementary Information
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
Wang, J., Zhang, Z., Song, L. et al. Inhibition of esophageal squamous cell carcinoma progression by MIR210HG and activation of the P53 signaling pathway to promote apoptosis and autophagy. Eur J Med Res 30, 269 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40001-025-02512-8
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40001-025-02512-8