Identification of novel SE-associated oncogenic lncRNAs in glioma
A previous study has demonstrated that SE-associated protein-coding genes were characterized in integrative analysis of both sensitivities to transcriptional inhibition and their expression levels in glioma cells[19]. To investigate the implications of SE-associated lncRNAs in glioma, the publicly available H3K27Ac and MED1 ChIP-seq datasets of glioblastoma (GBM) cells were obtained and analyzed to generate a catalog of SEs (Fig. 1A-B). We identified 256 and 197 SE-associated lncRNAs upon occupancy of either H3K27Ac or MED1, respectively (Fig. 1C). Importantly, 176 SE-associated lncRNAs were overlapped (Fig. 1C). To prioritize these SE-associated lncRNAs for further study, we selected lncRNAs with (1) elevated expression in GBM tissue versus low-grade glioma (LGG) tissue (Fig. 1D-E) and (2) prognostic significance from the integrated analysis CGGA and TCGA databases, where high lncRNA expression was associated with poor glioma patient survival (Fig. 1F). A total of nine differentially expressed SE-lncRNAs were selected for further analyses (Fig. 1F). To validate data, we analyzed nine candidates' SE-lncRNAs expression in the GEPIA database. As shown in FigS.1A-F, LUCAT1, LINC00601, LOXL1-AS1, SBF2-AS1, PINK1-AS, NEAT1 were observed with no significant differences in glioma compared to normal tissues. PVT1 and DLGAP1-AS2 were a high expression in GBM, but not LGG compared to normal brain tissue (FigS.1G-H). Importantly, only LIMD1-AS1 was finally found to be significantly overexpressed in GBM and LGG, but other types of cancer, suggesting that LIMD1-AS1 may be especially involved in the progression of glioma (FigS.1I).
LIMD1-AS1 is an oncogenic lncRNA in glioma
To study the roles of LIMD1-AS1 in gliomas, two published datasets: CGGA and TCGA were analyzed. In CGGA datasets, LIMD1-AS1 was highly expressed in GBM samples compared to that in LGG, and associated with the degree of malignancy, IDH mut (Fig. 2A-C). The elevated level of LIMD1-AS1 expression indicated poor survival for glioma patients (Fig. 2D). Consistent with these results, the expression of LIMD1-AS1 in gliomas was related to the degree of malignancy, IDH mut, and associated with poor survival for glioma patients in the TCGA database (Fig. 2E-I). Our cohort confirmed that LIMD1-AS1 was the high expression in gliomas compared with normal brain tissue (Fig. 2J). RT-qPCR analysis showed that LIMD1-AS1 expression was higher in all 4 GBM compared with LGG and normal astrocyte cell lines (Fig. 2K). Therefore, LIMD1-AS1 might exert an important function in glioma progression.
We next explored the underlying mechanism by which LIMD1-AS1 is up-regulated in glioma. Abnormal activation of proto-oncogenes caused by genomic gain, amplification, and hypomethylation is a frequent occurrence in gliomas [20, 21]. Firstly, we analyzed the DNA methylation data of the LIMD1-AS1 gene in glioma from the TCGA database. There was no significant association between LIMD1-AS1 methylation and its expression (FigS.2A-B). We then focused on the copy number variation (CNV) data of glioma in TCGA as previously described [22]. Although the copy number of LIMD1-AS1 was frequently upregulated in GBM, the copy number of LIMD1-AS1 slightly correlated with LIMD1-AS1 expression in glioma samples (FigS.2C-D).
To explore the cellular function of LIMD1-AS1, we performed the loss or gain-of-function assay to study its biological role in glioma cells. Specifically, siRNAs were applied to LN-18 and T98G glioma cell lines exhibiting high LIMD1-AS1 expression levels, while an overexpression plasmid was used in low-expressing LIMD1-AS1 SF126 glioma cells (Fig. 2K, Fig. 3A). LIMD1-AS1 knockdown by siRNA significantly inhibited cell viability in both LN-18 and T98G glioma cells, while LIMD1-AS1 overexpression increased clonogenic ability of SF126 glioma cells (Fig. 3B-C, Fig. 3H-J). Importantly, LIMD1-AS1 knockdown markedly reduced the migration and invasion in T98G and LN-18 (Fig. 3D-G), while ectopic expression of LIMD1-AS1 promoted migration and invasion in SF126 glioma cells (Fig. 3K-N). These data identified LIMD1-AS1 as a functionally oncogenic lncRNA in glioma. To examine the function of LIMD1-AS1 in vivo, glioma cells were implanted into nude mice subcutaneously. Knockdown of LIMD1-AS1 dramatically reduced tumor growth in xenograft mouse tumor models (Fig. 4A-B). In support of the pro-tumor role of LIMD1-AS1, the Ki67, and PCNA staining showed that LIMD1-AS1 knockdown reduced tumor cell proliferation in vivo (Fig. 4C-D). Our data suggest that LIMD1-AS1 contributes to glioma progression.
LIMD1-AS1 is a SE-driven lncRNA in glioma
To characterize the transcriptional regulation of LIMD1-AS1, H3K27ac, MED1, and Pol II profiles in GBM cell lines were analyzed by ChIP-seq. An SE upstream of LIMD1-AS1 was found in GBM cell lines but was absent in normal human cell lines (Fig. 5A). Additionally, co-localized with H3K27ac and MED1 (known to identify enhancer elements) in the identified super-enhancer of LIMD1-AS1 (Fig. 5A). To further verify H3K27ac and MED1 binds the SE and promoter of LIMD1-AS1, ChIP analysis was performed to show that H3K27ac binds SE and promoter of LIMD1-AS1 in LN-18 and T98G glioma cells (Fig. 5B-C). As expected, MED1 binds only at the LIMD1-AS1 super-enhancer (Fig. 5D-E). We next cloned promoter and individual super-enhancer constituents (E4 and E5) of the LIMD1-AS1 into promoter-reporter and enhancer-reporter vectors, and then measured their activities by luciferase reporter assay. E4, E5, and promoter were active in LN-18 and T98G glioma cell lines (Fig. 5F-G). As shown in Fig. 5H-J, the recruitment of the dCas9-KRAB to interfere with LIMD1-AS1 SEs regions resulted in significant downregulation of LIMD1-AS1 expression in LN-18 and T98G glioma cells. Silencing of the super-enhancer component decreased the proliferation in LN-18 and T98G glioma cells (Fig. 5K-L). Collectively, these results suggest that increased LIMD1-AS1 is driven by super-enhancers to promote glioma cell growth.
CDK7 interacts with MED1 to regulate the super-enhancer of LIMD1-AS1
SE-complex that is involved in the effect of SE that functions as independent or interdependent components of these large transcription-regulating complexes to drive high-level expression of their associated gene [23–25]. Thus, we explored whether the components of the SE-complex are involved in the regulation of LIMD1-AS1 in glioma progression. To address this question, we selected SE-complex with (1) significantly correlated with the expression of LIMD1-AS1, (2) Top ten expressions in GBM tissue versus LGG tissue from integrated analyses of and (3) prognostic significance in TCGA and CGGA database, where high lncRNA expression was associated with poor glioma patient survival (Fig. 6A-C, FigS.3A-C). Thus, the top-ranked gene CDK7 was selected for further validation and analysis. Cyclin-Dependent Kinase 7 (CDK7) is a CDK-activating kinase (CAK), and as a component of the general transcription factor TFIIH, it mediates RNA polymerase-II-based transcription and contributes to tumor progression [26, 27]. THZ1, a CDK7 inhibitor, treatment markedly reduced LIMD1-AS1 expression in LN-18 and T98G glioma cells (Fig. 6D). Similarly, CDK7 knockdown also markedly decreased LIMD1-AS1 expression in LN-18 and T98G glioma cells (Fig. 6E, FigS.3D). Mechanistically, we observed that the reporter activity was prominently increased upon transfection of either the LIMD1-AS1 promoter or enhancer (E4, E5), whereas THZ1 treatment or CDK7 knockdown potently inhibited this reporter activity (Fig. 6F-H). Taken together, our findings support the hypothesis that the perturbation of SE-complex components may collectively suppress the transcriptional activation activity of LIMD1-AS1 in glioma.
A recent study showed that CDK7 inhibition selectively targets MED1-mediated oncogenic transcriptional amplification[28]. The mediator complex subunit 1 (MED1) is a component of the mediator complex and functions as a transcriptional coactivator specifically enriched in a novel class of transcription regulatory DNA regions called super-enhancers [29]. Because MED1 enriched in super-enhancers of LIMD1-AS1 (Fig. 5C), we sought to determine whether CDK7 recruits MED1 to the super enhancer of LIMD1-AS1. Firstly, the inhibition of CDK7 decreased the mRNA expression of MED1 in LN-18 and T98G glioma cells (FigS.3E). Additionally, CDK7 knockdown reduced p-MED1 levels in LN-18 and T98G glioma cells, but had no significant effect on MED1 protein expression (Fig. 6I-L). We examined the subcellular location of CDK7 and MED1 using immunofluorescence staining, and found that CDK7 mainly colocalized with MED1 in the nucleus of LN-18 and T98G glioma cells (Fig. 6M). Most importantly, CDK7 was also significantly correlated with MED1 expression in glioma tissues from the TCGA and CGGA database (Fig. 6N, FigS.3F). Furthermore, endogenous CDK7 coimmunoprecipitated with endogenous MED1 in LN-18 and T98G glioma cells (Fig. 6O). The interaction between MED1 and CDK7 was further demonstrated by reverse endogenous coimmunoprecipitation of MED1 with CDK7 (Fig. 6O), verifying the interaction between MED1 and CDK7 in vitro. These results prompted us to explore whether CDK7 is associated with super-enhancer of LIMD1-AS1 mediated by MED1, we conducted ChIP analysis. We found that the inhibition of CDK7 decreased the binding of MED1 across the LIMD1-AS1 super-enhancer, but not promoter, in LN-18 and T98G glioma cells (Fig. 6P-S). To further verify whether MED1 is involved in epigenetic activation of LIMD1-AS1 induced CDK7, we observed LIMD1-AS1 expression after co-transfection of si-MED1 and CDK7 to SF126 glioma cells. As shown in Fig. 6T, MED1 knockdown partially reduced LIMD1-AS1 expression induced by CDK7. Together, these data indicated that MED1 might be involved in the function of CDK7 as an epigenetic activator of LIMD1-AS1 in glioma cells.
A covalent CDK7 Inhibitor alleviated LIMD1-AS1-promoted the growth of glioma cells
To address the translational potential of LIMD1-AS1, we prioritized A covalent CDK7 Inhibitor, THZ1, for further investigation based upon the expression of LIMD1-AS1 in glioma. We found that LIMD1-AS1 is significantly enriched in patient-derived PN12 and PN16 glioblastoma cells, but not MES23, MES27 glioblastoma cells (Fig. 7A). PN12 and PN16 glioblastoma cells migration and invasion were associated with LIMD1-AS1 expression, with THZ1 showing an inhibitory effect (Fig. 7B-F). We next examined whether LIMD1-AS1 was involved in the anti-tumor effect of this THZ1 therapy by treating PN12 and PN16 glioblastoma cells. LIMD1-AS1 partially reversed THZ1-mediated reduction in glioma cell migration (Fig. 7G-H). Additionally, LIMD1-AS1 reversed the reduction in invasive glioma cells treated with THZ1 (Fig. 7I-J). These results indicated that glioma cells with high expression of LIMD1-AS1 are susceptible to disturbance by THZ1.
LIMD1-AS1-regulated malignant phenotype of glioma partially dependents on HSPA5
Since the molecular mechanism of lncRNA's effect depends on subcellular localization, we first observe the localization of LIMD1-AS1 in LN-18 and T98G glioma cells. RNA fluorescence in situ hybridization (RNA-FISH) showed that LIMD1-AS1 was distributed mainly in the cytoplasm (Fig. 8A). Nuclear/cytoplasmic fractionation followed by RT-qPCR analysis further confirmed that LIMD1-AS1 was predominantly localized in the cytoplasm, with only weak expression in the nucleus (Fig. 8B-C). Cytoplasmic lncRNAs interact with various protein partners thus regulating RNA stability, degradation, translation and splicing of mRNAs[30, 31]. To investigate this phenomenon, we performed RNA pull-down using in vitro synthesized LIMD1-AS1 coupled to biotin, then subjected the precipitants to mass spectrometry (MS) analysis (Fig. 8D). Among these identified proteins, we selected HSPA5 protein base on HSPA5 correlated with LIMD1-AS1, highly expressed in glioma, and associated with overall survival and disease-free survival in glioma patients (Fig. 8E-H, FigS.4A-C). Western blot confirmed that Heat Shock Protein Family A (Hsp70) Member 5 (HSPA5) (also known as GRP78) was co-precipitated with in vitro synthesized LIMD1-AS1 but not with antisense LIMD1-AS1 (Fig. 8I). Moreover, an IF and RNA-FISH assay results showed that LIMD1-AS and HSPA5 were predominantly colocalized in the cytoplasm of LN-18 and T98G glioma cells (Fig. 8J). Subsequently, RT-qPCR analysis following RNA immunoprecipitation assays confirmed the enrichment of LIMD1-AS1 in the complex with HSPA5, compared with IgG control (Fig. 8K-L).
Then, the regulation of LIMD1-AS1 on HSPA5 was evaluated. We found that LIMD1-AS1 knockdown markedly reduced the HSPA5 protein level in the LN-18 and T98G glioma cells (Fig. 8M-N). To further investigate the molecular mechanism by which LIMD1-AS1 up-regulates HSPA5 expression at the posttranscriptional level, the protein synthesis inhibitor cycloheximide (CHX) was used. It was found that overexpression of LIMD1-AS1 in SF126 cells enhanced HSPA5 protein stability (Fig. 8O). Additionally, HSPA5 knockdown partially alleviated the proliferation, migration, and invasion in SF126 glioma cells induced by LIMD1-AS1 overexpression (Fig. 8P-R). Therefore, our results suggest that LIMD1-AS1 promotes malignant phenotype of glioma cells by directly binding the HSPA5 protein and enhancing its stability.
LIMD1-AS1 regulated interferon signaling by HSPA5 in glioma
To further elucidate the potential signaling pathway of LIMD1-AS1 in promoting glioma progression, we performed RNA-seq analysis to compare the gene expression profiles of LIMD1-AS1 overexpression and control SF126 glioma cells. A total of 596 differential genes were detected after overexpression of LIMD1-AS1 in glioma cells (Fig. 9A). Reactome analysis revealed that these gene sets are mainly related to interferon signals including Interferon alpha/beta signaling, DDX58/IFIH1-mediated induction of interferon-alpha/beta, Interferon Signaling (Fig. 9B). Additionally, we confirmed that LIMD1-AS1 is positively associated with interferon signaling in the TCGA and CGGA database (FigS.5A-B). Next, we focus on the top-ranked differential genes in the three gene sets, and significantly correlated with the expression of LIMD1-AS1 in the TCGA and CGGA database (Fig. 9C, FigS.6A-J), indicating LIMD1-AS1 and interferon signaling were usually co-expressed in glioma. These genes of interferon signaling including DDX58, USP18, STAT1, IRF1, and TNFAIP3 (IFN signatures) are for further analyses. We found that the mRNA expression of IFN signatures was reduced in LIMD1-AS1 knockdown glioma cells (Fig. 9D-F). IFN signatures mRNA levels were significantly up-regulated in glioma samples compared with normal brain tissue, and upregulation of IFN signatures was associated with glioma malignancy (Fig. 9G). The highly the IFN signatures is significantly associated with poor survival in patients with glioma (Fig. 9H, FigS.7A-J). Next, we explore whether HSPA5 was involved in the downregulation of the interferon signaling caused by LIMD1-AS1 knockdown. We transfected HSPA5 siRNA into LIMD1-AS1-overexpressed SF126 glioma cells and found that knockdown of HSPA5 expression significantly reduced LIMD1-AS1-induced increase IFN signatures mRNA levels in SF126 glioma cells (Fig. 9I). Therefore, these data indicate that LIMD1-AS1's regulation of interferon signaling is at least partially dependent on HSPA5 in gliomas.