Identifcation and characteristics of SNHG15 in GC
Firstly, 50 paired GC tissues and matched adjacent non-tumor tissues were selected to carry out the RNA-seq to analyze any alternative expression profiles of lncRNAs. Among them, 20 lncRNAs that have signifcant diference expression in GC tissues were chosen (15 upregulated circRNAs, 5 downregulated circRNAs) (Fig. 1A) and the same tendency was also found in GC tissues derived from TCGA (Additional file 2: Fig. S1A, B). We further analyzed the expression of the upregulated lncRNAs in 10 GC and the paired non-GC patient’s primary tissues from the SHNPH database and found that SNHG15 was the most signifcantly upregulated in tumor tissues compared with adjacent non-tumor tissues, and the expression of other 5 lncRNAs were also consistent with those observed in RNA-Seq (Additional file 2: Fig. S2). Notably, the results suggested that the expression of SNHG15 in GC tissues was signifcantly increased compared with corresponding normal mucosa sample in GC patients in SHNPH cohort (Fig. 1B) and SYSUCC cohort (Fig. 1C). Te correlation analysis in 93 cases of GC patients with clinical characteristics revealed that SNHG15 expression was correlated with tumor differentiation, lymph node metastasis and TNM stage using Chi-squared test (Table 1). Subsequently, Kaplan–Meier survival analysis with the log-rank test was performed. The data from our center revealed that GC patients with higher SNHG15 expression had poorer overall survival rates than those with lower SNHG15 expression (Fig. 1D). Next, we compared the SNHG15 expression in a tissue microarray containing 93 GC samples using FISH and demonstrated that the expression of SNHG15 was signifcantly upregulated in GC tissues compared with normal mucosa tissues (Fig. 1E); the increased expression of SNHG15 was positively correlated with advanced AJCC TNM stage (Fig. 1F, G). Collectively, these results suggested that the aberrantly high expression of SNHG15 is signifcantly associated with tumor progression in GC. Additionally, the coding potential of SNHG15 was predicted using several prediction software and the NCBI ORF finder (https://www.ncbi.nlm.nih.gov/orffinder/). The results showed that SNHG15 could not code any protein, indicating it a non-coding RNA (Additional file 2: Fig. S3A–D).
SNHG15 functions as a tumor inducer in vitro and in vivo
To elucidate the function of SNHG15 on cell biological behavior, we first explored the expression level of SNHG15 in six GC cell lines compared with normal gastric epithelial cells GES1 (Fig. 2A). Then, we transfected MGC803 and AGS cells with two different siRNAs and each siRNA could effectively inhibit the SNHG15 expression (Fig. 2B). The whole length of SNHG15-pcDNA overexpression vector was transfected into MKN45 cell. Meanwhile, to explore the function of SNHG15 in GC, SNHG15 was also knocked down by two of lentivirus-mediated short hairpin RNAs (shRNAs) in GC, and ectopic SNHG15 was transfected into GC with a lentivirus-carried SNHG15 (Additional file 2: Fig. S4A, B). Following, qRT-PCR verified that the expression of SNHG15 could increase over about 100 fold (Fig. 2B). To investigate the biological roles of SNHG15 in GC progression, the cell proliferation was analyzed using CCK8 assay and clone formation. All results showed that MKN45 cell proliferative capability was increased by SNHG15 overexpression, while knockdown of SNHG15 remarkably attenuated the proliferative effects in MGC803 and AGS cells (Fig. 2C, D). It can be concluded that SNHG15 is effective in promoting GC cell growth. We also tested the influence of SNHG15 on GC cell-cycle progression. As shown in Fig. 2E, knock down of SNHG15 significantly blocked the cell cycle at the G1–S phase, while upregulation of SNHG15 slightly facilitated cell cycle progression.
We then tested the proliferation-inducing effect of SNHG15 on an nude mouse model by injecting SNHG15 stable knockdown MGC803 cells, SNHG15 overexpressing MKN45 cells, or corresponding control cells into nude mice to determine whether SNHG15 could affect GC cell tumorigenesis in vivo. The results suggested that tumors grown from SNHG15 knockdown cells were smaller than the control cells (Fig. 3A–C); however, tumors grown from SNHG15 overexpressing cells were larger than those grown from control cells (Fig. 3 E–G). We further explored the role of SNHG15 in peritoneal dissemination by inoculating cells directly into the abdominal cavity of nude mice, and found that the abdominal metastatic burden was decreased in SNHG15 stable knockdown MGC803 cells, and increased in SNHG15-overexpressing MKN45 cells (Fig. 3D, H). To analyze the physiological function of SNHG15, we also generated additional SNHG15 stable knockdown BGC823 cells, and validated these data in vivo assays (Additional file 2: Fig. S5A–C).
In summary, SNHG15 was confirmed to play important roles on GC cell migration, invasion, and peritoneal colonization, signifying that the effect of SNHG15 on GC patients prognosis might be probably associated with the role in metastasis.
Oncogenic transcription factors contribute to SNHG15 overexpression
To further identify the potential mechanism involved in SNHG15 overexpression in GC, we analyzed the data available from TCGA dataset by cBioPortal [26]. As shown in Fig. 4A, there is a gene copy number co-amplification among SNHG15, E2F1, and MYC. We hypothesized that TFs may play important roles in modulating SNHG15 gene transcription. To confirm this hypothesis, we first obtained ChIP-seq of TFs from the ENCODE project by using UCSC Genome Bioinformatics Site (http://genome.ucsc.edu/) and JASPAR database (https://jaspar.genereg.net/), and found high enrichment of oncogenic TFs E2F1 and MYC at the promoter of SNHG15. Meanwhile, correlation analysis revealed that SNHG15 was significantly and positively correlated with E2F1 and MYC in our cohort (Fig. 4B) and the TCGA dataset (Fig. 4C). In addition, our ChIP assays showed that E2F1 and MYC could bind to the SNHG15 promoter DNA regions (Fig. 4D, left). To further evaluate the biologic roles of E2F1 and MYC in SNHG15 expression, we performed gain-of-function method, and revealed that the overexpression (Fig. 4D, right) upregulated E2F1 and MYC enrichment on the SNHG15 promoter. Meanwhile, overexpression and downregulation of E2F1 and MYC could lead to upregulated or downregulated expression of SNHG15 in GC cells, respectively (Fig. 4E). In addition, luciferase report assays indicated that E2F1 bound to the B (−610 bp) binding site, and MYC bound to the A (−1354 bp) binding site (Fig. 4F).
E2F1, MYC and CDC25A are key downstream targets of SNHG15
To explore the biologic pathways involved in GC pathogenesis, SNHG15 expression level was stratified by the median, and GSEA analysis was applied in our cohort RNA-seq datasets. Enrichment plots of GSEA showed that the gene signatures of cell cycle, nucleotide excision repair, and DNA replication were more involved in GC patients with SNHG15 higher expression relative to patients with lower expression (Fig. 5A) and TCGA datasets (Additional file 2: Fig. S6A). To gain further insights into SNHG15-related pathway in regulating GC progression, we evaluated the gene profiles in GC tissues with altered SNHG15 expression. RNA transcriptome sequencing analysis was used to detect the altered gene expression profiles from high and low expression of SNHG15 in our cohort (Fig. 5B) and TCGA (Additional file 2: Fig. S6B). Gene Ontology analysis revealed that changes in gene sets were primarily related to cell cycle in high-expressed-SNHG15 tissues (Fig. 5C). Further qRT-PCR data and Western blot validated that alterations in SNHG15 expression markedly affected the key gene signatures involved in tumorigenesis and GC progression (Fig. 5D, E), indicating that SNHG15 maybe a significant regulator for modulating the malignant phenotypes of GC by regulating cell cycle.
To further elucidate the potential genes involved in the function of SNHG15, we analyzed the RNA-sequencing and qPCR data, and confirmed that alterations in SNHG15 expression greatly affected E2F1, MYC and CDC25A gene enrichment (Fig. 5B, D, E). Therefore, we tested their expressions in human GC samples. IHC and qRT-PCR showed that E2F1, MYC and CDC25A expressions were increased in GC tumors with high expression of SNHG15 compared with GC tumors with low SNHG15 expression (Fig. 6A). Meanwhile, correlation analysis demonstrated that SNHG15 was significantly and positively correlated with E2F1, MYC and CDC25A in our GC cohort (Fig. 6B). To determine whether E2F1, MYC and CDC25A participated in increased GC cell proliferation and invasion induced by SNHG15, we performed a rescue experiment by cotransfected MGC803 cells with SNHG15-OE, siE2F1, or siMYC, or siCDC25A (Fig. 6C). Transwell assays demonstrated that downregulation of E2F1, MYC, and CDC25A partially rescued the SNHG15-induced cell invasion (Fig. 6D), and colony formation assays and CCK8 also showed that interference of E2F1, MYC and CDC25A could partially rescue the SNHG15-induced cell proliferation in vitro (Fig. 6E, F).
SNHG15 regulates E2F1, MYC, and CDC25A expression by competing for miR-24-3p
To elaborate the mechanism for SNHG15-regulated E2F1, MYC, and CDC25A expression, we firstly examined SNHG15 distribution in GC cells using RNA-fluorescence in situ hybridization and subcellular distribution. It was found that SNHG15 RNAs were more distributed in the cytoplasm (Fig. 1E, F), suggesting that SNHG15 was more likely to play a post-transcriptional regulation function. Knowing that lncRNAs can function as natural “microRNA sponges” that protect mRNAs by competing for the targeting miRNAs, it is worth investigating whether SNHG15 playes a similar role. We predicted miRNAs targeting sites on SNHG15 using the starBase tools prediction algorithm [27]. Of these miRNAs, miR-17-3p, miR-31-5p, miR-103b, and miR-24-3p have been reported to be associated with tumor proliferation and invasion [28, 29], and with significant binding energy predicted by the DIANA tools (Fig. 7A). Next, we focused on these miRNAs and explored the interaction between SNHG15 in GC cell lines. Upregulation of SNHG15 significantly decreased the expression of miR-24-3p, but not the expression of other miRNAs in GC cells (Fig. 7B). Interestingly, miR-24-3p, which was reported to target E2F2, MYC, and other cell-cycle related genes [30, 31], was also found to act as a tumor suppressor in GC in our previous study [21]. To investigate whether SNHG15 acted as a sponge of miR-24-3p, we constructed luciferase reporter vectors containing the wild type known as pLUC-SNHG15-WT or mutated miR-24-3p binding sites known as pLUC-SNHG15-mut. It was found that the upregulation of miR-24-3p decreased the luciferase activities of the reporter vector pLUC-SNHG15-WT but not mutant reporter vector in 293T cells (Fig. 7C). To illustrate whether SNHG15 was modulated by miR-24-3p, we conducted transiently upregulation or inhibition of miR-24-3p by mimics or inhibitor in MGC803 cells. We found SNHG15 levels were inhibited or upregulated after overexpression or inhibition of miR-24-3p (Additional file 2: Fig. S7). These findings indicated that miR-24-3p was associated with the expression of SNHG15. To verify the ceRNA network in GC tissues, miR-24-3p expression level was also analyzed and Pearson analysis showed a negative correlation between the expression score of miR-24-3p and SNHG15 (Additional file 2: Fig. S8).
To determine whether SNHG15 regulated E2F1, MYC, and CDC25A expression by competing for miR-24-3p, we performed rescue assays. Ectopic expression of SNHG15 increased E2F1, MYC, and CDC25A protein levels and upregulation of miR-24-3p reduced this increase, while inhibition of SNHG15 decreased the expression of E2F1, MYC, and CDC25A, and the use of miR-24-3p inhibitor reversed this decrease (Fig. 7D). We also found that the expression of miR-24-3p was negatively correlated with the expression of E2F1, MYC, and CDC25A in our GC tissues (Additional file 2: Fig. S9). CCK8 also showed that miR-24-3p partially rescued the SNHG15-induced cell proliferation in vitro (Additional file 2: Fig. S10). Ectopic expression of miR-24-3p remarkably inhibited cell migration and invasion, and re-expression of miR-24-3p was able to reverse the effect of SNHG15 on cell migration, invasion and colony formation in vitro (Fig. 7E, F). The results illustrated that SNHG15 regulated E2F1, MYC, and CDC25A expression by interacting with miR-24-3p.
To test the effect of miR-24-3p on the predicted target genes, we constructed three luciferase reporters to examine miR-24-3p activity by inserting the 3′ untranslated region (UTR) of human E2F1, MYC and CDC25A into 3′UTR of Renilla luciferase. As expected, miR-24-3p could significantly decrease the luciferase reporter signals of 3′UTR of E2F1, MYC and CDC25A (Fig. 7G). Meanwhile, these results also suggest that miR-24-3p regulates SNHG15 expression by targeting E2F1 and MYC. Furthermore, we conducted FISH in GC cells to examine the subcellular localization of miR-24-3p and SNHG15, and found a large extent of overlap in the cytoplasm, suggesting co-localization of miR-24-3p and SNHG15 (Fig. 7H).
SNHG15 acts as a repressor of ferroptosis in GC cells
Gene Ontology analysis demonstrated that changes in gene sets were mainly correlated to the regulation of signal transduction by p53 in addition to cell cycle regulation in high-expressed-SNHG15 tissues (Fig. 5C). Given that tumor growth arrest, DNA repair and apoptosis are believed to constitute the core mechanisms of p53-dependent tumour suppression [32], we examined the effects of SNHG15 on apoptosis by flow cytometry. However, we found that upregulation or downregulation of SNHG15 had no effect on apoptosis of GC cells (Additional file 2: Fig. S11A, B). LncRNAs are involved in the regulation of reactive oxygen species (ROS) levels, and ferroptosis is a form of cell death resulting from the catastrophic accumulation of lipid ROS [33]. Further analyses in our cohort (Additional file 2: Fig. S12A, B) and the TCGA database (Additional file 2: Fig. S12C, D) revealed that SNHG15 expression was significantly correlated with key regulatory gene sets of ferroptosis. We further confirm the effect of SNHG15 on ferroptosis, and found that downregulation of SNHG15 increased erastin-induced ferroptosis (Fig. 8A), which was rescued by treatments of the cells with ferroptosis inhibitor ferrostatin-1 (Fig. 8B). Conversely, SNHG15 overexpressing GC cells were robustly resistant to erastin-induced ferroptosis (Fig. 8C). Moreover, downregulation of SNHG15 in GC cells exhibited an increase in MDA production, while SNHG15 overexpression decreased MDA production (Fig. 8D, E).
Although p53-mediated growth arrest and apoptosis serve as critical barriers to cancer development, emerging evidence suggests that ferroptosis regulation of p53 is also important [34]. As shown in Fig. 5C, p53 may play an important role in SNHG15-mediated carcinogenesis. To test the effect of miR-24-3p on the regulation of signal transduction by p53, we constructed luciferase reporter containing the 3′UTR of human MDM2, which is considered to be a negative regulator of p53 signaling pathway [35]. As expected, miR-24-3p could significantly decrease the luciferase reporter signals of 3′UTR of MDM2 (Fig. 8F). Previous study showed p53 sensitized cells to ferroptosis by repressing expression of SLC7A11, a key regulator of ferroptosis[34]. Rescue assays revealed that ectopic expression of SNHG15 increased MDM2 protein level and upregulation of miR-24-3p reduced this increase, and SNHG15 suppressed ferroptosis by miR-24-3p-mediated regulation of MDM2/p53/SLC7A11 signal axis (Fig. 8G). Meanwhile, we found that miR-24-3p inhibitor could deteriorate ferroptosis promotion induced by downregulation of SNHG15 (Fig. 8H), while upregulation of miR-24-3p could reverse the ferroptosis inhibition induced by SNHG15 (Fig. 8I). Taken together, these findings indicate that the SNHG15/miR-24-3p/MDM2/p53/SLC7A11 signaling inhibits ferroptosis.