Lnc-TSPAN12 promotes HCC cells metastasis in vitro and in vivo
Our previous study demonstrated the high expression of lnc-TSPAN12 in HCC and its association with unfavourable clinicopathological features and poor prognosis. To further determine the potential role of lnc-TSPAN12 in HCC, we knocked down or overexpressed lnc-TSPAN12 in Huh7 and Hep3B HCC cell lines by transfecting shRNA (pEX-2) or overexpressing a plasmid (pcDNA3.1) and confirmed the transfection efficiency by quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis (Additional file 1: Fig. S1A-B). Wound healing and transwell assays showed that knockdown of lnc-TSPAN12 evidently inhibited the migration and invasion of Huh7 and Hep3B cells in vitro when compared with the scramble control group (Fig. 1A-D, Additional file 1: Fig. S1C, D). By contrast, the ectopic expression of lnc-TSPAN12 enhanced migration and invasion of HCC cells (Fig. 1A-D, Additional file 1: Fig. S1C, D).
EMT has been recognized as a critical process in HCC metastasis [25, 26]. Thus, we aimed to determine whether lnc-TSPAN12 is involved in inducing EMT in HCC cells. As shown in Fig. 1E, F, knockdown of lnc-TSPAN12 significantly increased the expression of epithelial cell marker E-cadherin but decreased those of mesenchymal cell markers Vimentin and Fibronectin. Nevertheless, overexpression of lnc-TSPAN12 presented the opposite result. To better demonstrate this phenomenon, we used confocal microscopy after immunofluorescence staining. The expressions of E-cadherin and Vimentin in Huh7 cells were consistent with those observed in western blot analysis (Fig. 1G, H). Collectively, our data suggest that lnc-TSPAN12 may promote HCC metastasis by regulating EMT.
To further explore the promoting effects of lnc-TSPAN12 on HCC cell metastasis, we firstly constructed stable lnc-TSPAN12-knockdown Huh7 HCC cells (sh-lnc-TSPAN12#1, 2) and established patient-derived xenograft (PDX) models for liver metastasis by injection into the inferior hemispleen of nude mice. As shown in Fig. 2A-D, after knockdown of lnc-TSPAN12 in Huh7 cells, the numbers and luciferase activity of metastatic nodules in the liver significantly decreased compared with those of the control cells. Haematoxylin and eosin staining showed that the metastatic foci in the sh-lnc-TSPAN12#1,2 cells dramatically shrunk in the liver sections (Fig. 2E). Immunohistochemical staining analysis revealed that knockdown of lnc-TSPAN12 led to a reduced level of Vimentin and an increased level of E-cadherin in xenograft tumour tissues (Fig. 2F). Taken together, these findings suggest that lnc-TSPAN12 may function as an oncogenic driver in the metastasis of HCC.
Lnc-TSPAN12 specifically interacts with EIF3I
Given that the function of lncRNA is related to its subcellular localization, we firstly detected the distribution of lnc-TSPAN12 by RNA fluorescence in situ hybridization (FISH) in Huh7 cell. The result suggested that lnc-TSPAN12 was predominantly present in the cytoplasm (Additional file 1: Fig. S2A). To elucidate the potential molecular mechanisms of lnc-TSPAN12 in HCC metastasis, we performed RNA-seq analysis to investigate the difference in the gene expression profiles of Huh7 cell before and after lnc-TSPAN12 knockdown. Hierarchical clustering showed that a total of 4,505 genes (FDR<0.05 and|log2FC|> 1.0) were significantly altered in lnc-TSPAN12 knockdown Huh7 cell when compared with the control cell (Additional file 1: Fig. S2B, Table S4). KEGG enrichment analysis revealed that the enriched differential genes belonged to several key biological processes and signal pathways, including cell adhesion, gap junctions, extracellular matrix receptor interactions and TGF-β, which are closely associated with EMT and cancer metastasis (Additional file 2: Fig. S2C). These data suggest that lnc-TSPAN12 may be a critical modulator in HCC metastasis.
To further investigate the regulatory role of lnc-TSPAN12 in HCC metastasis, we performed RNA pull-down assay combined with mass spectrometry (MS) to screen lnc-TSPAN12-interacting proteins. In brief, the extracted proteins were subjected to silver staining and SDS-PAGE analysis, and several distinct bands were selected for mass spectrum analysis (Fig. 3A and Additional file 2: Table S5). We noted that EIF3I, the top ranked lnc-TSPAN12-interacting protein in band 1, has been shown to play critical roles in cancer progression and β-catenin pathway [27, 28]. Importantly, Wnt/β-catenin signalling is deemed as a powerful driver of EMT [26]. Therefore, we selected this protein for subsequent validation. We further identified EIF3I as specific lnc-TSPAN12-binding protein by western blot (Fig. 3B). In addition, RNA immunoprecipitation (RIP) assays were adopted with antibodies against EIF3I to demonstrate the association between EIF3I and lnc-TSPAN12 (Fig. 3C). Furthermore, lnc-TSPAN12 FISH followed by immunofluorescence of EIF3I verified the colocalization of lnc-TSPAN12 and EIF3I in the cytoplasm of Huh7 cell, further supporting their interaction (Fig. 3D). Taken together, these data suggest that lnc-TSPAN12 physically interacts with EIF3I.
To identify the binding region of lnc-TSPAN12 that interacts with EIF3I, we constructed a series of lnc-TSPAN12 truncated fragments based on the predicted secondary structure of lnc-TSPAN12 in the catRAPID database (http://service.tartaglialab.com/page/catrapid_group, Fig. 3E) [29] and then performed RNA pull-down assays with these deletion mutant constructs. It was shown that the 430–493 nt fragment of lnc-TSPAN12 was responsible for its interaction with EIF3I (Fig. 3F). EIF3I is a protein containing the WD40 domain, with seven WD40 repeats covering almost the entire sequence of EIF3I [30]. Next, to further determine the domain of EIF3I that accounted for its interaction with lnc-TSPAN12, we performed RIP assays with FLAG-tagged full-length and five truncated EIF3I. As shown in Fig. 3G, the deletion of RNA-binding domains (128–325 and 218–325 aa) of EIF3I dramatically abolished the association between this protein and lnc-TSPAN12, indicating that these domains may be essential for the interaction with lnc-TSPAN12.
Lnc-TSPAN12 increases the stability of EIF3I protein
To investigate whether a cross-talk exists between lnc-TSPAN12 and EIF3I, we firstly measured the EIF3I levels in lnc-TSPAN12-knockdown HCC cells. The mRNA levels of EIF3I remained unaltered after knockdown of lnc-TSPAN12 (Additional file 1: Fig. S3A). However, knockdown of lnc-TSPAN12 led to a remarkable decrease in EIF3I protein level, suggesting that lnc-TSPAN12 can increase the levels of EIF3I protein at the post-transcriptional level (Fig. 4A). Similarly, immunohistochemical staining analysis against previous xenograft tumour tissues displayed that lnc-TSPAN12 knockdown was also correlated with the decreased level of EIF3I (Additional file 1: Fig. S3B). Through treatment with the protein synthesis inhibitor cycloheximide (CHX), we observed that knockdown of lnc-TSPAN12 in Huh7 and Hep3B cells accelerated the degradation of EIF3I and shortened its half-life (Fig. 4B, C). Moreover, following treatment with the proteasome inhibitor MG132, the down-regulation of endogenous EIF3I in HCC cells induced by knockdown of lnc-TSPAN12 was prevented, and this finding was accompanied by increased EIF3I ubiquitination levels, which indicates that EIF3I degradation through the ubiquitin-proteasome pathway may be inhibited by lnc-TSPAN12 (Fig. 4D, Additional file 1: Fig. S3C). We further discovered that several E3 ligases can mediate the ubiquitination and degradation of EIF3I in the Ubibrowser database (Additional file 1: Fig. S3D) (http://ubibrowser.ncpsb.org/) [31].
Previous study has shown that the lysine at 298 served as the major ubiquitination site of EIF3I [32, 33]. We then mutated this lysine to alanine (K298A mutant) and evaluated whether this mutant can block the EIF3I ubiquitination degradation. We measured the ubiquitination and expression level of EIF3I in lnc-TSPAN12 knockdown HCC cells co-transfected with a lentiviral vector expressing a FLAG-tagged EIF3I K298A mutant (FLAG-EIF3I-K298A) and a lentiviral vector expressing a FLAG-tagged wild-type EIF3I (FLAG-EIF3I-WT). Indeed, knockdown of lnc-TSPAN12 can reduce the stability of EIF3I and facilitate its degradation via enhancing its ubiquitin modification, whereas K298A mutant significantly abolished the promoting effect of lnc-TSPAN12 knockdown on EIF3I ubiquitination degradation, thus increasing the expression level of EIF3I (Fig. 4E, F). These data indicated that lnc-TSPAN12 has a negative impact on the process of EIF3I ubiquitination. Therefore, we further determined the effect of lnc-TSPAN12 on the binding of EIF3I to its predicted ubiquitin E3 ligases, including SMURF1 and STUB1. Co-immunoprecipitation (co-IP) experiments showed that knockdown of lnc-TSPAN12 enhanced the interaction between EIF3I and SMURF1, but with no evident changes on the binding between EIF3I and STUB1 (Fig. 4G). Thus, lnc-TSPAN12 can block the ubiquitination and degradation of EIF3I by attenuating its binding to the ubiquitin E3 ligase SMURF1.
Lnc-TSPAN12 enhancement of SENP1-mediated EIF3I deSUMOylation inhibits its ubiquitination
We further investigated the mechanism of lnc-TSPAN12-mediated ubiquitination and degradation of EIF3I. SUMOylation is now recognized as a crucial ubiquitin-like, post-translational modification (PTM) of proteins by SUMO proteins, which can regulate the stability, subcellular localization and interaction of the targeted substrate proteins [34–36]. Intriguingly, previous studies have shown that SUMOylation and ubiquitination often occur at the same lysine residues of a substrate protein, and SUMOylation can sometimes promote the ubiquitination and degradation of modified proteins [37, 38]. Of note, apart from EIF3I, a SUMO1-specific protease SENP1 was also found in band 4. RNA pull-down followed by western blot and RIP assay were performed, and the results showed that SENP1 was also directly bind to lnc-TSPAN12 (Fig. 5A, B). FISH followed by immunofluorescence of SENP1 further verified the colocalization of lnc-TSPAN12 and SENP1 in the cytoplasm of Huh7 cell (Additional file 1: Fig. S4A). In addition, the 430–493 nt fragment of lnc-TSPAN12 was also confirmed to be responsible for its interaction with SENP1(Additional file 1: Fig. S4B). Given that SENP1 is responsible for the removal of SUMO family [39], we wondered whether SENP1 acts as a lncRNA-binding protein and gives lnc-TSPAN12 the capacity for deSUMOylation- and/or deubiquitination-related PTM on EIF3I. As expected, lnc-TSPAN12 knockdown had minimal effect on the expression of SENP1 protein (Additional file 1: Fig. S4C). However, co-IP assay results showed that knockdown of lnc-TSPAN12 decreased the combination of SENP1 and EIF3I, proposing that lnc-TSPAN12 may serve as a scaffold to promote the binding of SENP1 to EIF3I (Fig. 5C). Moreover, after knockdown of lnc-TSPAN12, the binding of EIF3I with SUMO1, SUMO2 and SUMO3 was enhanced to varying degrees, indicating the facilitation of lnc-TSPAN12 to EIF3I deSUMOylation (Fig. 5D).
We next investigated whether SUMOylation of EIF3I exerts influences on the combination of EIF3I with ubiquitination enzyme and ubiquitination of EIF3I. We found that silencing of SENP1 increased the combination of EIF3I with SMURF1 and SUMO1, as well as enhancement of EIF3I ubiquitination (Fig. 5E, F). On the other hand, the binding of EIF3I with SUMO1, SUMO2, SUMO3 and SENP1 remained unchanged after silencing of SMURF1 (Fig. 5G), implying that SUMOylation of EIF3I can mediate its ubiquitination, whereas the ubiquitination of EIF3I had no effect on its SUMOylation level. Coincidentally, K298 is also the underlying SUMOylation site according to the bioinformatics website prediction(http://ubibrowser.ncpsb.org/ubibrowser). To verify this prediction, we mutated K298 and detected the changes in EIF3I SUMOylation level after knockdown of lnc-TSPAN12 in Huh-7 cell. Accordingly, this mutant remarkably reduced the EIF3I SUMOylation (Fig. 5H). Similarly, the intracellular level of EIF3I was obviously decreased in sh-lnc-TSPAN12 + Flag-EIF3I-K298A group when compared with that of the FLAG-EIF3I-WT (Fig. 5H). Consequently, these findings show that lnc-TSPAN12 enhances SENP1-mediated deSUMOylation of EIF3I K298, thereby restraining the ubiquitination of EIF3I.
Lnc-TSPAN12 exerts EMT-promoting functions in HCC by regulating EIF3I/SENP1
Considering the effect of lnc-TSPAN12 on EIF3I stability, we hypothesized that lnc-TSPAN12 may exert its biological effects through interacting with EIF3I/SENP1 in HCC. We observed that specific shRNA for EIF3I/SENP1 remarkably reduced the migration and invasion abilities of Huh7 cell (Fig. 6A, B). In addition, the increased cell migration and invasion in lnc-TSPAN12-overexpressing HCC cell was reversed by EIF3I/SENP1 knockdown (Fig. 6A, B). These data suggest that knockdown of EIF3I/SENP1 can reverse the lnc-TSPAN12-induced metastasis in HCC.
To further elucidate the role of lnc-TSPAN12 in Wnt/β-catenin signalling, we firstly examined the effects of lnc-TSPAN12 on the expression of downstream target genes of the Wnt/β-catenin signalling. As shown in Fig. 6C, D, the mRNA and protein expression levels of cyclin D1, c-myc and Axin2 were significantly down-regulated in Huh-7 cells with lnc-TSPAN12 knockdown. We next investigated whether lnc-TSPAN12 can activate Wnt/β-catenin signalling through regulating the EIF3I/SENP1 activity to induce EMT in HCC. Consistent with these results, western blot findings further showed that ectopic lnc-TSPAN12 expression increased the expression levels of β-catenin along with its downstream target genes and induced EMT in HCC, whereas knockdown of EIF3I (Fig. 6E) or SENP1 (Fig. 6F) alleviated lnc-TSPAN12-mediated Wnt/β-catenin signalling activation. Collectively, our results demonstrate that EIF3I and SENP1 are required for lnc-TSPAN12 to activate Wnt/β-catenin signalling, which consequently induces EMT and promote metastasis in HCC.
m6A modification is responsible for lnc-TSPAN12 upregulation in HCC
Preliminary studies have reported that m6A modifications are widespread in mRNA and lncRNA and functionally regulate the transcriptome to affect RNA translation, export, localization and stability [40, 41]. We then determined whether m6A modification is associated with lnc-TSPAN12 upregulation in HCC. Methylated RIP qPCR analysis of HCC cells was performed. The results showed that the m6A methylation level of lnc-TSPAN12 was highly enriched in Huh7 and Hep3B cells (Fig. 7A). Our bioinformatics analysis suggested that METTL3, the crucial m6A methyltransferase, was evidently upregulated in HCC (Fig. 7B). Moreover, the overall survival (Fig. 7C) and disease-free survival (Fig. 7D) of HCC patients with high METTL3 expression were also significantly lower than those with low METTL3 expression, indicating its prognostic value in HCC.
To investigate the effects of METTL3 on lnc-TSPAN12 upregulation in HCC, we thus knocked down the expression of METTL3 using shRNA in Huh7 and Hep3B cells. As shown in Fig. S5A, the knockdown efficiency was verified by both western blot. We found that METTL3 knockdown was associated with remarkably decreased lnc-TSPAN12 expression level and m6A level of lnc-TSPAN12 when compared with the control group (Fig. 7E, F). Afterward, we treated two HCC cells with methylation inhibitor 3-DAA and observed that the expression level of lnc-TSPAN12 was significantly reduced (Fig. 7G). Finally, RNA FISH assay displayed that METTL3 and lnc-TSPAN12 were colocalized in the cytosol (Fig. 7H).
Furthermore, METTL3 knockdown suppressed the invasion and migration abilities of Hep3B cell (Additional file 1: Fig. S5B, C). Consistently, after knockdown of METTL3 in Hep3B and Huh7 cells, the expression of E-cadherin was significantly upregulated, whereas those of Vimentin and Fibronectin were significantly downregulated (Additional file 1: Fig. S5D). The above findings raised the possibility that the transcriptional stability of lnc-TSPAN12 was enhanced by METTL3-mediated m6A modification, which may partially be responsible for the upregulation of lnc-TSPAN12 in HCC.