Increased DLGAP1-AS2 expression is associated with poor clinical outcomes in CRC patients
To identify potential CRC-related lncRNAs, we performed transcriptional profiling analyses using next-generation sequencing in nine paired CRC and NCTs. The top 50 differentially expressed lncRNAs were verified with the data from the TCGA CRC cohort, and DLGAP1-AS2 showed significant upregulation in both CRC cohorts (Fig.1A). The aberrant upregulation of DLGAP1-AS2 was further validated in four additional CRC cohorts(GSE8671, GSE32323, GSE18105 and GSE22598, Fig.1B-C, Fig.S1). Moreover, pancancer analyses revealed that DLGAP1-AS2 was highly expressed in other types of cancer, including stomach adenocarcinoma, esophageal carcinoma, pancreatic adenocarcinoma, cholangiocarcinoma and kidney renal papillary cell carcinoma, suggesting that DLGAP1-AS2 is a key cancer-related lncRNA (Fig.1D). Thus, we focused on DLGAP1-AS2 for subsequent study.
Further experimental validation using an independent CRC cohort we collected showed that 67% (67 of 101) of CRCs showed more than 1.5-fold upregulation of DLGAP1-AS2 in CRCs compared with the adjacent NCTs (Fig.1E-F). Kaplan–Meier survival analyses showed that high DLGAP1-AS2 expression was significantly correlated with poor overall survival (Fig.1G) and disease-free survival (Fig. 1H).Correlation analyses showed that DLGAP1-AS2 expression levels were correlated with tumor differentiation, lymph node metastasis and tumor stage (Table S6). Furthermore, univariate and multivariate Cox proportional hazard analyses identified DLGAP1-AS2 as an independent prognostic factor for CRC (Fig.1I-J).
Identification of a novel transcript of DLGAP1-AS2 in CRC
ThreeDLGAP1-AS2transcripts,923 bp, 1138 bp and 2261 bp in length, were listed in GENECODE or GenBank (Fig. 2A). Interestingly, when we cloned DLGAP1-AS2 based on the only transcript provided by GenBank (NR_119377), a novel transcript with an additional 58 bp in the second exon was identified, which we submitted to GenBank(MK336171, Fig.2B).Further analyses using qRT-PCR and semi quantitative RT-PCR revealed that the novel transcript was the predominant transcript in CRC cells (Fig. 2C-D).In addition, further analyses using the PhyloCSF and Coding Potential Assessment Tool (CPAT) indicated that DLGAP1-AS2 lacks protein-coding potential (Fig. S2A-B). Consequently, we focused on this new and predominant transcript for subsequent studies in CRC. DLGAP1-AS2 is highly expressed in different CRC cell lines (Fig.S2C) and distributed in both the cytoplasm and nucleus of CRC cells (Fig. S2D).
DLGAP1-AS2 promotes CRC growth and metastasis.
CRC cells with relatively higher(HCT116 and SW480) or lower (LoVo and DLD1) DLGAP1-AS2 expression were selected for gene knockdown or overexpression and subsequent functional assays, respectively.CCK-8 and colony formation assays demonstrated that DLGAP1-AS2 knockdown significantly inhibited, whereas ectopic DLGAP1-AS2 expression promoted the proliferation and colony formation abilities of CRC cells (Fig.3A-C).Transwell assays showed that DLGAP1-AS2 induction enhanced the migration and invasion of LoVo and DLD1 cells. In contrast, DLGAP1-AS2 knockdown drastically inhibited the migration and invasion activities of HCT116 and SW480 cells (Fig.3D-E).
To further explore the growth-promoting effects of DLGAP1-AS2 on CRC in vivo, we subcutaneously injected CRC cells with stable knockdown or overexpression of DLGAP1-AS2 into nude mice. Both the volumes and weights of the xenograft tumors in the knockdown group were markedly lower than those in the control group. In contrast, ectopic DLGAP1-AS2 expression significantly promoted CRC tumorigenesis (Fig. 3F).
We also used a mouse lung metastasis model to evaluate the effect of DLGAP1-AS2 on CRC metastasis. The results showed that DLGPA1-AS2 knockdown drastically inhibited, whereas DLGAP1-AS2 overexpression promoted CRC metastasis (Fig.3G, Fig. S3A-B).Taken together, these data demonstrate that DLGPA1-AS2 promotes CRC growth and metastasis.
DLGAP1-AS2 interacts with CPSF2, CSTF3 and ELOA in CRC cells
To explore the molecular mechanism underlying the oncogenic role of DLGAP1-AS2 in colorectal carcinogenesis, we performed RNA pull-down assays to identify the proteins associated with DLGAP1-AS2 in CRC cells. The retrieved proteins were subjected to SDS-PAGE electrophoresis, mass spectrum and subsequent western blotting analyses. The results showed that CPSF2, CSTF3 and ELOA were potential DLGAP1-AS2-associated proteins(Fig. 4A-B,Fig.S4A-C). Moreover, RIP assays further confirmed the associations between these three proteins and DLGAP1-AS2(Fig. 4C,Fig.S5A).
To identify the regions of DLGAP1-AS2 accounting for the binding of DLGAP1-AS2 to CPSF2, CSTF3 or ELOA, we constructed a series of DLGAP1-AS2 mutants based on their secondary structure as predicted by LNCipedia (http://www.lncipedia.org/) andcatRAPID (http://service. tartaglialab.com/page/catrapidgroup)(Fig.S6A-C). RNA pull-down assays with these mutants showed that the 1–379 nt, 621–981 nt or 70–981 nt fragment of DLGAP1-AS2 mediates its interaction with CPSF2,CSTF3 or ELOA, respectively(Fig. 4D).
We then constructed several deletion mutants of these three proteins for RIP assays. The results showed that the deletion of 376-728 aa of CPSF2 significantly abolished the association between CPSF2 and DLGAP1-AS2(Fig. 4E). Additionally, the 1-374 aa domain of CSTF3 mediates its association with DLGAP1-AS2 (Fig. 4F), and the 251-500 aa domain of ELOA physically associates with DLGAP1-AS2 in CRC cells(Fig.4G). Together, these data indicate that DLGAP1-AS2 specifically binds to CPSF2, CSTF3 and ELOA in CRC cells.
DLGAP1-AS2 promotes ELOA ubiquitination and degradation.
Although we revealed that DLGAP1-AS2 interacted withCPSF2, CSTF3 and ELOA in CRC cells, their underlying functional and mechanistic effects were unclear. We evaluated the effects of DLGAP1-AS2 on the expression of these targets, and no obvious changes were observed at either the protein or mRNA levels of CPSF2 and CSTF3. The mRNA levels of ELOA also did not change inDLGAP1-AS2-depleted or DLGAP1-AS2-overexpressing CRC cells (Fig.S7A-C). However, the protein levels of ELOA were dramatically increased in DLGAP1-AS2-depleted CRC cells and were notably reduced with DLGAP1-AS2 overexpression(Fig.5A).Moreover, ectopic DLGAP1-AS2 expression decreased the half-life of the ELOA protein in CRC cells treated with the protein synthesis inhibitor cycloheximide (CHX) (Fig.5B,Fig.S8A).DLGAP1-AS2-induceddownregulation of ELOA protein was blocked in CRC cells treated with the proteasome inhibitor MG132 (Fig.5C).Furthermore, we found that the ubiquitination levels of ELOA were significantly increased inDLGAP1-AS2-overexpressing CRC cells and were significantly decreased in DLGAP1-AS2-depleted CRC cells (Fig.5D).Taken together, these data suggest that DLGAP1-AS2 promotes the proteasome-dependent degradation of ELOA in CRC cells.
DLGAP1-AS2 promotes the interactions between ELOA and Trim21.
To investigate howDLGAP1-AS2 accelerates the ubiquitin-mediated proteasome degradation of ELOA, we screened out six E3 ligases using ELOA IP and subsequent mass spectrometry analyses(Fig.5E). Of these E3 ligases, Trim21 showed the highest abundance, and co-IP assays further confirmed the association between Trim21 and ELOA(Fig.5F). Immunofluorescence assays also suggested that ELOA and Trim21 colocalized with each other in CRC cells (Fig.S8B). The protein levels of ELOA were dramatically increased in Trim21-depleted CRC cells and were significantly reduced in Trim21-overexpressing CRC cells (Fig.5G).MG132 treatment rescued the Trim21-induced downregulation of ELOA(Fig.5H), suggesting that Trim21 promotes the proteasome-dependent degradation of ELOA in CRC cells. Moreover, Trim21 knockdown increased the half-life of ELOA in CRC cells treated with CHX(Fig.5I). Furthermore, we demonstrated that the ubiquitination levels of ELOA were significantly decreased in Trim21-depleted CRC cells and increased in Trim21-overexpressing CRC cells (Fig.5J).
RNA pull-down assays revealed that DLGAP1-AS2 bind to Trim21(Fig.5K), and RIP assays further indicated that DLGAP1-AS2was significantly enriched in the RNA-protein complexes precipitated with anti-Trim21 antibody in CRC cells (Fig.5L).We next examined whether DLGAP1-AS2 impacts the interactions between ELOA and Trim21 in CRC cells, and showed that DLGAP1-AS2 knockdown significantly impaired, whereas ectopic DLGAP1-AS2 expression significantly enhanced this association (Fig.5M). Collectively, these results indicate that DLGAP1-AS2 promotes Trim21-mediated ubiquitination degradation of ELOA by enhancing the interaction between Trim21 and ELOA.
ELOA inhibits CRC growth and metastasis
ELOA, a subunit of the transcription factor B (SIII) complex, is relatively understudied and its role in tumorigenesis and progression is unclear. Therefore, we studied the functional role of ELOA in CRC by using a series of in vitro and in vivo assays. We demonstrated that ELOA knockdown significantly promoted, whereas ectopic ELOA expression inhibited, CRC cell proliferation and colony formation (Fig.6A-C,Fig.S8C-D). Transwell assays demonstrated the inhibitory functions of ELOA on the migratory and invasive abilities of CRC cells (Fig. 6D-E).
We further confirmed the growth-suppressive effects of ELOA on CRC in vivo by using a xenograft nude mouse model (Fig. 6F).In addition, a mouse lung metastasis model was applied to evaluate the effect of ELOA on CRC metastasis. The results showed that the number of lung metastatic nodules was decreased in the ELOA-overexpressing group compared with the control group (Fig.6G). Collectively, the above results reveal that ELOA inhibits the growth and metastasis of CRC.
ELOA protein expression negatively correlates with DLGAP1-AS2 and is associated with good prognosis in CRC
To further evaluate the role of ELOA in CRC, we detected its expression in clinical CRC tissues using IHC (Fig. 7A). Kaplan-Meier survival analyses showed that low ELOA expression was significantly correlated with poor overall survival (Fig. 7B).Correlation analyses showed that ELOA expression levels were correlated with tumor differentiation, lymph node metastasis and tumor stage (Fig. 7C,Table S7).Furthermore, univariate and multivariate Cox proportional hazard analyses identified ELOA as a dependent prognostic factor for CRC (Fig. 7D).Importantly, a significant negative correlation was observed between the protein expression levels of DLGAP1-AS2 and ELOA (Fig. 7E).These results suggest that ELOA is involved in CRC tumorigenesis and is regulated by DLGAP1-AS2.
ELOA is a downstream functional target of DLGAP1-AS2
To verify whether ELOA is a functional target of DLGAP1-AS2, we performed a rescue assay. The results revealed that silencing ELOA expression restored the impaired proliferation and colony formation abilities induced by DLGAP1-AS2 knockdown, whereas ectopic expression of ELOA remarkably impaired the proliferation-promoting effects of DLGAP1-AS2 overexpression in HCT116cells.Transwell assays demonstrated the ELOA rescue the promoting effect of DLGAP1-AS2 on the invasive abilities of CRC cells (Fig.7F-G, Fig.S8E-F).These data suggest that ELOA is adownstream functional target of DLGAP1-AS2.
To further investigate the mechanistic association between DLGAP1-AS2 and ELOA, we compared the transcriptome profiles in HCT116 cells transfected with si-DLGAP1-AS2, ELOA plasmid or their corresponding control. A total of 682 DEGs, including 478upregulated genes and 204 downregulated genes, were identified in DLGAP1-AS2-silenced HCT116 cells compared with control cells. In contrast, a total of 1420 DEGs, including 768 downregulated genes and 652 upregulated genes, were observed in ELOA-overexpressing HCT116 cells. We performed GSEA pathway enrichment analyses using these 682 and 1420DEGs and found that the pathways potentially affected byDLGAP1-AS2 or ELOA highly overlapped, including the inflammatory response, mitotic spindle, PI3K/AKT, interferon and UV response pathways (Fig. 7H). Furthermore, some genes in these pathways potentially regulated by both DLGAP1-AS2 and ELOA were verified using qRT-PCR. We observed that ELOA overexpression significantly rescued the effects of DLGAP1-AS2 knockdown on the expression of several genes in the inflammatory response, AKT and interferon pathways (Fig. 7I), suggesting that DLGAP1-AS2 regulates the expression of its downstream genes through ELOA.
ELOA transcriptionally regulates LHPP expression by specifically binding to its promoter
To screen gene harboring the specific binding site of ELOA in their promoters, a ChIP-on-chip assay was employed in HCT116 cells using Nimblegen human 720K RefSeq promoter arrays. A total of 836 promoters were enriched by the ChIP-on-chip assay using an ELOA antibody (false discovery rate<0.05). By combining the results of the ChIP-on-chip assay with those of the above-mentioned transcriptome profile data, three genes (LHPP, IL7 and CMPK2) potentially regulated by ELOA were screened out. Of them, only LHPP, a tumor suppressor [16], appeared to be upregulated by ELOA and was selected for further validations. As expected, LHPP mRNA expression was significantly increased in ELOA-overexpressing CRC cells(Fig. 7J).
To validate the potential ELOA-binding region(-1655/-1134) revealed by the ChIP-on-chip assay, we constructed two mutants with deletion of -1034/-513 or-1655/-1134 (as a negative control) of the LHPP promoter. Luciferase assays showed that ELOA failed to stimulate the reporter expression of the constructs containing the -1034/-513 deletion of the LHPP promoter, suggesting that ELOA transcriptionally regulated LHPP expression by specifically binding to its promoter(Fig.7K).To validate it, we performed ChIP-qPCR assays using primers flanking the promoter segments of LHPP(-1034/-513). We observed a significant amount of ELOA bound to the -684/-513 region of the LHPP promoter (Fig.7L), confirming that ELOA enhances the transcription of LHPP by directly binding to its promoter.
Using MEME-ChIP database and Markov model, we searched the conserved sequences in the promoters of 836 genes screened by the ChIP-on-chip assay, and revealed two possible motif binding sequences of ELOA(GCTGGGATTACAGGC and CCAGCCTGGGCAACA).One of them existed in the -579/-565 region of the LHPP promoter (TGTCTGTCAGGGTGT, partially reverse complement to the sequence of CCAGCCTGGGCAACA).When this region was mutated, ELOA failed to induce luciferase expression of the recombinant reporter plasmids (Fig. 7M).Based on the above results, we conclude that ELOA promotes the transcription of LHPP by binding to the-579/-565 region of the LHPP promoter.
DLGAP1-AS2 activates the AKT pathway by regulating the ELOA/LHPP axis
We revealed thatDLGAP1-AS1 regulated several key cancer-related pathways, including PI3K/AKT (Fig. 7H-I). Interestingly, recent studies have reported that LHPP inhibits the PI3K/AKT signaling pathway in CRC[17, 18]. We found that LHPP was expressed at lower levels in CRC tissues compared with NCTs based on multiple public CRC cohorts(Fig. S9A). We also showed that LHPP expression was negatively correlated with DLGPA1-AS2, and was positively correlated with ELOA in CRC (Fig. 7N,Fig. S9B).To determine whether LHPP mediated the regulation of the PI3K/AKT pathway by ELOA in CRC, we measured the AKT activity(phosphorylated AKT at Ser473) in ELOA-overexpressing CRC cells and confirmed that ELOA increased LHPP expression and suppressed AKT pathway activity (Fig. S9C). What is more, the rescue assays revealed that ELOA overexpression inhibitedDLGAP1-AS2-inducedAKT activation (Fig. 7O).Taken together, these data demonstrate that DLGPA1-AS2 promotes CRC development and progression by regulating the ELOA/LHPP/AKT signaling axis in CRC.
CPSF2 and CSTF3bind to DLGAP1-AS2and increaseits stability
CPSF2 and CSTF3, which are part of the C/P machinery family, function as multi protein complexes to regulate RNA processing and stability. We have shown that both CPSF2 and CSTF3 are DLGAP1-AS2-associated proteins. Analyses of public CRC databases revealed that both CPSF2 and CSTF3 were upregulated in tumor tissues compared with paired NCTs, and higher CPSF2 and CSTF3 expression was associated with worse survival (Fig. S10A-D).We observed that DLGAP1-AS2 did not affect the expression of CPSF2 and CSTF3 in CRC cells, whereas CPSF2 or CSTF3 positively regulated the expression of DLGAP1-AS2 in CRC cells (Fig.8A).Moreover, actinomycin D (2ug/ml) treatment showed that both CPSF2 and CSTF3 increased the stability of DLGAP1-AS2 (Fig.8B). Functionally, the knockdown of CPSF2 or CSTF3 remarkably reduced, whereas the overexpression of CPSF2 or CSTF3 promoted the proliferation of CRC cells (Fig. S11A-D). Furthermore, DLGPA1-AS2 overexpression rescued the decreased cell proliferation induced by the knockdown of CPSF2 or CSTF3 in CRC cells (Fig. S12A-D).As expected, CPSF2 or CSTF3 negatively regulated the expression of ELOA through DLGAP1-AS2 in CRC cells (Fig. 8C).
We further revealed that CPSF2 was able to bind to CSTF3 (Fig.8D), and their interaction was enhanced by DLGAP1-AS2 in CRC cells(Fig.8E), suggesting that DLGAP1-AS2 functions as a molecular scaffold to enhance the association between them. In addition, we showed that CPSF2 and CSTF3 might work together to increase the levels of DLGA1-AS2 (Fig.8F).Interestingly, we found a CPSF2-specific binding motif (AAUAAA) in the 89-94 nt region of DLGAP1-AS2. When the motif was deleted (ΔAATAAA), the promoting effect of CPSF2 and CSTF3 on DLGAP1-AS2 was significantly suppressed(Fig. 8G). Taken together, these data suggest that CPSF2 and CSTF3 may work together to stabilizeDLGAP1-AS2 in CRC cells.