LINC00313 is a novel TGFβ target in CCA
By gene expression profiling, we identified 103 non-redundant genes differentially expressed by TGFβ in both HuCCT1 and Huh28 CCA cell lines (Fig. 1A), including known (SERPINE1) and novel coding and non-coding TGFβ targets (Table S1). Notably, the long intergenic non-protein coding RNA 313 (LINC00313) was greatly induced by TGFβ in HuCCT1, Huh28 and NHC (Fig. 1A, B). LINC00313 was up-regulated by TGFβ in a dose-dependent manner (Fig. 1C) and appeared to be an intermediate-to-late and early responsive gene in HuCCT1 and Huh28 cells, respectively (Fig. 1D). Using exon-specific primers, we further confirmed the induction of LINC00313 by TGFβ in CCA cells (Fig. S1A). LINC00313 was also highly expressed in hepatocellular carcinoma (HCC) but TGFβ had no impact on its expression in HepG2 and Hep3B HCC cell lines (Fig. S1B). Finally, subcellular fractionation demonstrated that LINC00313 was mainly a nuclear lncRNA, similar to MALAT1 and SNORD48 (Fig. 1E).
TGFβ-induced LINC00313 expression requires TβRI/Smad- and p38-dependent pathways
TGFβ signals through Smad-dependent (canonical) or Smad-independent (non-canonical) pathways [4]. TβRI inhibitor LY2157299 abolished TGFβ-induced LINC00313 expression in TGFβ-responsive cells (Fig. 2A, S1A, S2A). Similarly, blocking SMAD3 activation using SIS3 inhibitor completely abolished the TGFβ-mediated up-regulation of LINC00313 in HuCCT1 and Huh28 cells (Fig. 2B). In HuCCT1, SMAD4 silencing reduced baseline and TGFβ-induced LINC00313 expression similar to the combined SMAD2/3/4 silencing (Fig. 2C, S2B). In Huh28, SMAD2 silencing also prevented TGFβ-induced LINC00313 expression (Fig. 2D, S2B).
As regard to the non-canonical TGFβ pathway, p38 inhibition decreased, while MEK inhibition increased LINC00313 expression. Inhibition of c-Jun N-terminal kinase (JNK) induced LINC00313 in the presence of TGFβ in HuCCT1 cells (Fig. 2E). The efficiency of MAPKs inhibition was verified (Fig. S3A). The enhanced LINC00313 expression upon MEK inhibition correlated with an enhanced TGFβ/SMAD response, as revealed by a SMAD-binding element (SBE) reporter assay (Fig. S3B). In Huh28 cells, LINC00313 remained unchanged after blocking MAPKs (Fig. 2E). Thus, both SMAD3 and SMAD4 and the kinase activity of p38 are required for the TGFβ-mediated up-regulation of LINC00313.
LINC00313 modulates expression of genes involved in Wnt pathway
Based on its nuclear localization, we hypothesized that LINC00313 acts at the transcriptional level to regulate gene expression (Fig. 3A). Gain-of-function tools were developed in HuCCT1 cells. An efficient LINC00313 over-expression and a nuclear abundance similarly to parental HuCCT1 cells was confirmed (Fig. S4A). RNA-seq analysis identified 334 up- and 316 down-regulated genes after LINC00313 gain-of-function (Fig. 3B, S4B, Table S2). Up-regulated genes were related to signalling pathways regulating pluripotency of stem cells (e.g. WNT5A, TCF7, AXIN2, FZD2 and ID1) (Fig. 3C). Gene Set Enrichment Analysis (GSEA) further highlighted signatures of Hippo, Wnt and TGFβ pathways in the gene expression profile of LINC00313 overexpressing cells (Fig. 3D, S4C). Induction of WNT5A, AXIN2 and SULF2 following LINC00313 gain-of-function was also validated (Fig. 3E, S4D).
Next, gene expression profiling after LINC00313 silencing was performed in the presence or absence of TGFβ (Fig. S5A, S5B and Table S3). GO analysis confirmed that TGFβ-induced genes participated in cell migration, extracellular matrix organization and cell differentiation (Fig. S5C). KEGG pathway analysis revealed that many genes repressed after LINC00313 silencing are associated with cancer signalling pathways regulating pluripotency of stem cells, including the Wnt signalling (Fig. S5D). In agreement with LINC00313 gain-of-function experiments, WNT5A, AXIN2 and SULF2 expression levels were decreased upon LINC00313 silencing in HuCCT1 cells (Fig. 3F). Expression of TCF7, SULF2 and WNT5A was also diminished in LINC00313-silenced Huh28 and TFK1 cells (Fig. S6A). These observations were confirmed in Huh28 using another siRNA in order to exclude possible siRNA off-target effects (Fig. S6B).
Nuclear LINC00313 alters chromatin accessibility at genomic loci of Wnt-related genes
ATAC-seq identified genome-wide alterations in chromatin accessibility, including 23,628 unique peaks in LINC00313 over-expressing cells (pcLINC00313) (Fig. 4A, S7A, S7B). No global alteration of the location of peaks, relative to genomic annotations was observed (Fig. S7C). Differential region analysis revealed 1657 genomic regions with increased accessibility and 2090 regions with decreased accessibility in pcLINC00313 cells (Fig. 4B and Table S4). HOMER motif enrichment analysis highlighted Fos-related antigen 1 (Fra1), an AP-1 transcription factor subunit, as the most significant enriched motif (Fig. S7D).
We then integrated ATAC-seq and RNA-seq data and pinpointed the common genes. We found 44 genes that show both altered chromatin accessibility and altered expression after LINC00313 over-expression (Fig. 4C). Interestingly, these genes were associated with the Wnt signalling pathway (Fig. 4D), known to play a key role in CCA progression [17]. Thus, we decided to investigate in deep the regulation of selected genes of the pathway, such as TCF7. Notably, we observed increased peak signal around the transcription start site and in the gene body of TCF7 locus (Fig. 4E), as well as elevated TCF7 mRNA and protein levels in LINC00313 over-expressing cells (Fig. 4F). Interestingly, TGFβ induced TCF7 expression whereas silencing LINC00313 reduced TCF7 mRNA and protein levels (Fig. 4G) in both control and TGF-β stimulated cells, suggesting that LINC0313 may promote chromatin opening and enhanced transcriptional activity at the TCF7 locus.
LINC00313 potentiates TCF/LEF transcriptional responses
Then, we investigated whether LINC00313 could modulate Wnt/β-catenin-dependent transcription. We first validated luciferase reporter assays in HuCCT1 cells treated with CHIR99021 (CHIR), a glycogen synthase kinase 3 inhibitor, which activates the Wnt signalling. In HuCCT1 cells stably expressing a TCF/LEF reporter, CHIR increased reporter activity but TGFβ had no impact (Fig. S8A). Silencing LINC00313 repressed CHIR-induced TCF/LEF reporter activity (Fig. 5A) and the associated target genes (Fig. 5B). LINC00313 over-expression further enhanced the baseline and CHIR-induced TCF/LEF-luciferase activity (Fig. 5C). Administration of CHIR increased the viability of control HuCCT1 cells, an effect that was more pronounced in cells over-expressing LINC00313 (Fig. S8B). Moreover, CHIR boosted colony formation of HuCCT1 control cells and to a larger magnitude of LINC00313 over-expressing cells (Fig. S8C).
XAV939, a Wnt pathway inhibitor, did not influence TCF/LEF responses in control cells, but reduced it in pcLINC00313 cells, which exhibit enhanced Wnt activity (Fig. 5D). Interestingly, XAV939 reduced colony formation (Fig. S8C). We confirmed the efficiency of the inhibitors by measuring AXIN2 expression, as a typical Wnt-target gene. Indeed, CHIR strongly induced AXIN2, an effect that was enhanced in LINC00313 over-expressing cells, whereas XAV939 modestly reduced AXIN2 (Fig. S8D). Also, CHIR potentiated TCF7 but not LINC00313, while XAV939 had no effect on their expression (Fig. S8D). β-catenin nuclear import is the major event that drives Wnt transcriptional responses. LINC00313 over-expression did not change the subcellular localization of β-catenin (Fig. S8E). Collectively, the data support a model whereby LINC00313 acts as a positive regulator of TCF/LEF-mediated signalling, but is dispensable for the initial steps of Wnt activation.
LINC00313 promotes CCA colony-forming capacities in vitro and tumour growth in vivo
Wnt signalling regulates cancer stem cell maintenance, cancer cell proliferation and migration [18]. In this context, we showed that LINC00313 boosted the ability of single cells to form colonies in vitro (Fig. 5E). Moreover, LINC00313 over-expression neutralized the TGFβ-mediated decrease in HuCCT1 cell viability (Fig. S9A). However, LINC00313 did not influence cell migration (Fig. S9B).
Interestingly, LINC00313 accelerated tumour growth in vivo when cells were xenografted in nude mice (Fig. 5F, S10A). Expression analysis revealed a positive correlation between LINC00313 and TCF7, SULF2 and AXIN2 mRNA levels in resected tumours (Fig. 5G). Moreover, we measured increased SULF2 and AXIN2 expression in LINC00313 xenograft tumours, compared to control (Fig. 5H). Although TCF7 mRNA levels did not change (Fig. 5H), we observed increased TCF7 protein expression in LINC00313 tumours (Fig. S10B). LINC00313 had no impact on the development of spontaneous metastases in mice (Fig. S10C) in agreement with the absence of effects on cell migration in vitro.
The SWI/SNF complex subunit ACTL6A is an interactor of LINC00313 lncRNA
RNA pull-down assay followed by mass spectrometry identified LINC00313 partners possibly involved in chromatin remodelling. In total, 1538 proteins were identified to interact with LINC00313 and 1840 proteins with the negative control firefly luciferase (f-luc) mRNA (Fig. 6A, Table S5). From the above, 121 proteins were specifically bound to LINC00313 (Fig. 6B). In order to single out interesting candidates, we set three sorting layers (Fig. 6C). First, we focused on nuclear proteins (51/121), considering the nuclear localisation of LINC00313. Protein network analysis revealed multiple physical and functional associations and clustered them in four groups. The largest cluster contained proteins involved in cell cycle regulation and the rest members of the transcription factor TFIID complex, the integrator complex and proteins with RNA helicase activity (Fig. 6D). Second, we predicted the binding of LINC00313 to 51 nuclear proteins using catRAPID [19]. Thirty two proteins were predicted to interact with LINC00313 lncRNA (interaction propensity ≥ 75, discriminative power ≥ 98%) (Table S5). Third, among the 32 proteins we searched for transcription factors or chromatin modifiers (Fig. 6E), including actin-like 6A (ACTL6A) (Fig. 6F), a subunit of the SWI/SNF chromatin remodeling complex [20]. Given that LINC00313 modulates chromatin state of certain loci, we investigated the physical and functional association between LINC00313 and ACTL6A deeper. Initially, we verified the nuclear localization of ACTL6A. TGFβ stimulation did not alter the nuclear abundance of ACTL6A (Fig. 6G). A specific interaction between LINC00313 lncRNA and ACTL6A was demonstrated in HuCCT1, especially in the presence of TGFβ (Fig. 6H). Similarly, in HA-tagged ACTL6A over-expressing HEK293T cells, an even stronger interaction between LINC00313 and HA-ACTL6A was observed (Fig. 6I). We also confirmed this interaction by RNA immunoprecipitation (RIP) assays. Interestingly, TGFβ stimulation increased enrichment of endogenous LINC00313 to immunoprecipitated ACTL6A (Fig. 6J). Overall, LINC00313 forms ribonucleoprotein complexes with several nuclear proteins that belong to transcriptional regulatory networks.
ACTL6A silencing or pharmacological inhibition of SWI/SNF complex diminishes TCF/LEF-mediated gene expression
Next, we evaluated the effects of ACTL6A on TCF/LEF-dependent transcription. Silencing ACTL6A attenuated TCF7 mRNA and protein levels but did not affect TGFβ-induced LINC00313 (Fig. 7A). ACTL6A silencing also resulted in decreased TCF/LEF luciferase activity and a drop in TCF7 and AXIN2 mRNA levels (Fig. 7B). Rescue experiments demonstrated that ACTL6A silencing dampened the LINC00313-mediated up-regulation of SULF2 mRNA (Fig. 7C) and TCF7 mRNA and protein levels (Fig. 7D).
Since ACTL6A is an accessory subunit of SWI/SNF, we hypothesized that blocking the catalytic activity of the complex may yield effects similar to ACTL6A silencing. Thus, we utilized two inhibitors, targeting different domains of the core catalytic subunits SMARCA2/SMARCA4. Notably, the bromodomain inhibitor PFI-3 did not affect SULF2 or TCF7 mRNA expression. In contrast, administration of BRM/BRG1 ATPase inhibitor significantly reduced SULF2 mRNA (Fig. 7E) and TCF7 expression both at the mRNA and protein levels (Fig. 7F), implying that these two genes are targets of SWI/SNF. Consistent with the effects on the individual genes, BRM/BRG1 ATPi, but not PFI-3 treatment resulted in a modest, but significant decrease of CHIR-induced TCF/LEF-luciferase reporter activity (Fig. 7G). Overall, we suggest a mechanism, whereby increased LINC00313 levels promote chromatin accessibility in an ACTL6A/SWI/SNF-dependent manner and facilitate transcription of TCF7 and SULF2, resulting in enhanced Wnt activation (Fig. 7H).
LINC00313 signature predicts poor prognosis in patients with CCA
From the TCGA dataset, LINC00313 was neither overexpressed in CCA tissues nor correlated with overall or disease-free survival in patients with CCA (Fig. S11A). On the other hand, the expression levels of ACTL6A, TCF7, WNT5A, AXIN2 and SULF2 were all increased in CCA human tumours (Fig. S11B). Interestingly, LINC00313 was significantly up-regulated in pancreatic ductal adenocarcinoma and correlated with lower disease-free survival (Fig. S11C).
Next, we hypothesized that if LINC00313 expression may not be a prognostic factor in CCA, its activity could better reflect its clinical relevance. Thus, we performed RNA-seq analysis in control and LINC00313 xenograft tumours to establish a signature reflecting LINC00313 activity. In total, 347 genes were up-regulated and 327 genes were down-regulated in LINC00313-overexpressing tumours (Fig. 8A). Then, we integrated this signature with the gene expression profiles of 255 cases of clinically annotated human CCA. Integrative transcriptomics using principal component analysis (PCA) showed that the four control samples clustered together and were distinct to the four LINC00313 samples (Fig. 8B). Dimension 2 of the PCA was related to LINC00313 activity and identified two main clusters of human CCA (Fig. 8B). Interestingly, the cluster associated with LINC00313 activity correlated with lower overall survival (Fig. 8C). Furthermore, IDH1 and IDH2 mutations were more prevalent in control cluster, while KRAS and TP53 mutations were significantly enriched in LINC00313 cluster (Fig. 8D). We also observed increased levels of carbohydrate antigen 19 − 9 and γ-glutamyltransferase and decreased levels of albumin, markers of hepatobiliary disease and of unfavourable prognosis in CCA, in the LINC00313 cluster (Fig. 8E). In addition, the LINC00313 cluster was characterized by significantly higher perineural invasion and regional lymph node metastasis (Fig. 8E). Collectively, we conclude that although LINC00313 expression per se is not a prognostic factor in CCA, the in vivo gene signature that reflects its activity exhibits a strong prognostic value in terms of survival outcome.