High expression of miRNAs in miR-23a/27a/24 − 2 cluster closely associated with disease progression and immune evasion in NSCLC
To examine the correlation between miRNAs expression level of miR-23a/27a/24 − 2 cluster and NSCLC progression, patients with high expression of all miRNAs of miR-23a/27a/24 − 2 cluster in tumor tissues compared to adjacent tissues are classified into the high expression group. In contrast, patients with low or same expression level of all miRNAs of miR-23a/27a/24 − 2 cluster in tumor tissues compared to adjacent tissues are classified into the low expression group. Clinical data show that in NSCLC patients, miRNAs expression levels of miR-23a/27a/24 − 2 cluster are negatively correlated with overall survival rate (Fig. 1A) and disease-free survival rate (Fig. 1B). In addition, we found that group with high expression of miRNAs in miR-23a/27a/24 − 2 cluster had a lower number of infiltrating CD8+ T cells in tumor tissues (Fig. 1C). Further, Gene Ontology (GO) using mRNA sequencing data from miR-23a/27a/24 − 2 cluster silenced H1299 cells and control cells showed silencing of miR-23a/27a/24 − 2 cluster affects T cell activation, differentiation (Fig. 1D). Importantly, Gene Set Enrichment Analysis (GSEA) show miRNAs expression levels of miR-23a/27a/24 − 2 cluster negatively correlated with T cell mediated immunity in NSCLC cells (Fig. 1E). Consistent results were observed from the analysis of GEO dataset. As showing in Fig. 1F, GEO dataset analysis showed that miR-23a/27a/24 − 2 cluster expression level was associated with T cell activation and differentiation in NSCLC. In addition, target-based miRNA functional analysis [18] also showed that miRNAs of miR-23a/27a/24 − 2 cluster were associated with immune system process (Fig. 1G). Taken together, these findings suggesting that upregulated miRNAs of miR-23a/27a/24 − 2 cluster contribute to NSCLC progression by inhibiting T cell mediated immunity.
miRNAs of miR-23a/27a/24 − 2 cluster inhibit T cell mediated immune response in NSCLC
To investigate whether miRNAs of miR-23a/27a/24 − 2 cluster directly regulates T cell mediated immune response, T cells were co-cultured with miR-23a/27a/24 − 2 cluster overexpressing NSCLC cells. That transfection of miR-23a/27a/24 − 2 cluster expressing construct significantly upregulated all miRNAs of miR-23a/27a/24 − 2 cluster in NSCLC cells (Supplementary Fig. S2A). In vitro results show that overexpression of all miRNAs in miR-23a/27a/24 − 2 cluster significantly reduced T cell released IFN-γ (Fig. 2A), T cell-induced NSCLC cell death (Fig. 2B and Supplementary Fig. S3), and T cell migration (Fig. 2C). In addition, that overexpression of all miRNAs in miR-23a/27a/24 − 2 cluster (Supplementary Fig. S2B) dramatically promoted tumor growth (Fig. 2D), inhibited CD8+ T cells infiltrate into tumor tissues (Fig. 2E), and reduced IFN-γ expression (Fig. 2F) in the C57BL/6J xenograft models that constructed using LLC cells. In contrast, silencing of miR-23a/27a/24 − 2 cluster in LLC cells (Supplementary Fig. S2B) inhibited tumor growth (Fig. 2G), increased CD8+ T cells infiltration (Fig. 2H) and IFN-γ expression (Fig. 2I). These results were further confirmed using other xenograft models. Consistent with results from C57BL/6J xenograft models that silencing of miR-23a/27a/24 − 2 cluster in H1299 cells (Supplementary Fig. S2C) significantly inhibited tumor growth when the cells were inoculated in SCID mice that treated with or without peripheral blood mononuclear cells (PBMCs) (Fig. 2J-L). Notably, that PBMCs treatment more effectively increased the tumor inhibition rate of miR-23a/27a/24 − 2 cluster silencing in SCID mice models (Fig. 2M). Together, these data indicating a potential role of miR-23a/27a/24 − 2 cluster miRNAs in the inhibition of T cell mediated antitumor immune responses.
miRNAs of miR-23a/27a/24 − 2 cluster upregulated PD-L1 and downregulated MHC-Ⅰ expression in NSCLC
To investigate which proteins are involved in the inhibition of miRNAs in miR-23a/27a/24 − 2 cluster on T cell immune response, we performed proteomics analysis using NSCLC cells with high expression of all miRNAs in the miR-23a/27a/24 − 2 cluster and their control cells. As showing in Fig. 3A, many proteins expression was affected by overexpression of all miRNAs in the miR-23a/27a/24 − 2 cluster, including PD-L1 and MHC-Ⅰ, and that GO analysis showed that miR-23a/27a/24 − 2 cluster overexpression was correlated with MHC-Ⅰ mediated antigen processing and presentation pathway. Because PD-L1 and MHC-Ⅰ are key molecules for immune evasion and ICB resistance, we chose PD-L1 and MHC-Ⅰ proteins as targets for further experiments. Consistent with proteomics analysis, Western blot (Fig. 3B) and Flow cytometry analysis (Fig. 3C) showed the PD-L1 expression was upregulated by overexpression of miR-23a/27a/24 − 2 cluster miRNAs in NSCLC cells. The inhibitory effect of miR-23a/27a/24 − 2 cluster on MHC-Ⅰ expression in NSCLC cells also confirmed by Western blot (Fig. 3B) and immunofluorescence (IF) assay (Fig. 3D). In addition, immunohistochemistry (IHC) analysis of tumor tissues from a lung cancer model constructed using LLC cells in C57BL/6J mice showed that high expression of PD-L1 and low expression of MHC-Ⅰ in miR-23a/27a/24 − 2 cluster miRNAs high expressing group (Fig. 3E). These results have also been further confirmed in human NSCLC specimens by IHC analysis (Fig. 3F). Collectively, these data suggesting that miR-23a/27a/24 − 2 cluster may inhibit T cell mediated tumor immune response by upregulating PD-L1 and downregulating MHC-Ⅰ in NSCLC.
miRNAs of miR-23a/27a/24 − 2 cluster upregulated the expression of PD-L1 in NSCLC by targeting CBLB
Then, we investigated the regulatory mechanism of miR-23a/27a/24 − 2 cluster miRNAs on PD-L1 expression in NSCLC. From proteomics analysis, we found that CBLB was downregulated in group of miR-23a/27a/24 − 2 cluster overexpression (Fig. 3A), which is a negative regulator of PD-L1 in NSCLC [19]. The inhibitory effect of miRNAs of miR-23a/27a/24 − 2 cluster on CBLB expression in NSCLC cells at mRNA (Fig. 4A) and protein levels (Fig. 4B) was further confirmed by qRT-PCR and Western blot, respectively. Notably, our data show that overexpression of CBLB suppressed miRNAs of miR-23a/27a/24 − 2 cluster-induced upregulation of PD-L1 in NSCLC cells (Fig. 4C), suggesting that miRNAs of miR-23a/27a/24 − 2 cluster upregulate PD-L1 by inhibiting CBLB. In fact, we using computational algorithm analysis (targetscan.org) predicted the 3`-untranlated region (3`-UTR) of CBLB contains sequences that can bind with miR-23a and miR-27a (Fig. 4D). Importantly, a luciferase report assay showed that overexpression of miR-23a/27a/24 − 2 cluster significantly suppressed the luciferase level that regulated by wild 3`-UTR of CBLB, but, did not affect the luciferase expression level that regulated by mutant 3`-UTR of CBLB (Fig. 4E). The negative and positive correlation of miR-23a/27a/24 − 2 cluster with CBLB and PD-L1 expression was further confirmed by IHC analysis in tumor tissues that from C57BL/6J lung cancer models constructed using miR-23a/27a/24 − 2 cluster overexpressing LLC cells (Fig. 4F) and NSCLC patients (Fig. 4G). Taken together, these findings indicating that miRNAs of miR-23a/27a/24 − 2 cluster upregulate PD-L1 expression in NSCLC through inhibiting CBLB by directly targeting 3`-UTR of CBLB.
miRNAs of miR-23a/27a/24 − 2 cluster inhibits the expression of MHC-Ⅰ in NSCLC by targeting MITF
We also investigated the regulatory mechanism of miR-23a/27a/24 − 2 cluster on MHC-Ⅰ expression. From proteomic results, we found that eIF3B was upregulated in miR-23a/27a/24 − 2 cluster overexpressed groups (Fig. 3A). In addition, computational algorithm analysis showed that 3`-UTR of MITF contains sequences that can bind with all miRNAs of miR-23a/27a/24 − 2 cluster (Fig. 5A). According to Santasusagna et al. report, that MITF inhibits the expression of eIF3B, while eIF3B inhibits the expression of MHC-Ⅰ [20], suggesting that miRNAs of miR-23a/27a/24 − 2 cluster in NSCLC may downregulate MHC-Ⅰ expression through upregulating eIF3B by targeting MITF. As expected, in vitro experiment results showed that overexpression of miR-23a/27a/24 − 2 cluster dramatically downregulated the expression of MITF and MHC-Ⅰ expression in NSCLC cells, while, upregulated eIF3B expression (Fig. 5B). Also, downregulated expression of MITF by miR-23/27a/24 − 2 cluster at mRNA level was demonstrated in NSCLC cells (Fig. 5C). Importantly, that overexpression of MITF or silencing eIF3B restored MHC-Ⅰ expression that inhibited by miR-23a/27a/24 − 2 cluster (Fig. 5D). The miR-23a/27a/24 − 2 cluster-induced upregulation of eIF3B was also inhibited by overexpression of MITF (Fig. 5D). Further, we using luciferase reporter assay investigated whether miRNAs in miR-23a/27a/24 − 2 cluster inhibit MITF expression by targeting 3`-UTR of MITF. As showing in Fig. 5E, overexpression of miR-23a/27a/24 − 2 cluster significantly suppressed the luciferase level that regulated by wild 3`-UTR of MITF, but, did not affect the luciferase expression level that regulated by mutant 3`-UTR of MITF. Finally, the correlation among the expression levels of miR-23a/27a/24 − 2 cluster, MITF, eIF3B and MHC-Ⅰ was further confirmed by IHC analysis of tumors from C57BL/6J xenograft models that constructed using miR-23a/27a/24 − 2 cluster overexpressing LLC cells (Fig. 5F) and patients with NSCLC (Fig. 5G). Collectively, these findings suggesting that miRNAs of miR-23a/27a/24 − 2 cluster inhibits MHC-Ⅰ expression in NSCLC through upregulating eIF3B by directly targeting MITF.
miRNAs of miR-23a/27a/24 − 2 cluster maintain its expression through β-catenin/TCF4 axis in NSCLC
To investigate the regulatory mechanism of miR-23a/27a/24 − 2 cluster miRNAs in NSCLC, we have screened candidate transcription factors that may be involved in the regulation of miR-23a/27a/24 − 2 cluster expression, and found that TCF4 can bind with promoter of miR-23a/27a/24 − 2 cluster (Fig. 6A). TCF4 is an effector of Wnt/β-catenin signaling and involved in miRNAs expression by binding to promoter region of miRNAs [21]. In addition, our previous study has shown that miRNAs of miR-23a/27a/24 − 2 cluster stimulate TCF/LEF complex regulated gene expression by activating Wnt/β-catenin signaling in NSCLC [9]. Thus, we proposed that miR-23a/27a/24 − 2 cluster maintenance its expression through β-catenin/TCF4 axis. To prove this hypothesis, we first confirmed that TCF4 whether regulates miR-23a/27a/24 − 2 cluster expression. Our results show that overexpression of TCF4 significantly upregulated expression of miR-23a, miR-27a and miR-24-2 (Fig. 6B) and luciferase transcription that regulated by miR-23a/27a/24 − 2 cluster promoter (Fig. 6C). In addition, EMSA results show that TCF4 directly interact with miR-23a/27a/24 − 2 cluster promoter site that containing TCF4 binding sequences, while, mutation of binding sequence significantly reduced the interaction between TCF4 and miR-23a/27a/24 − 2 cluster promoter (Fig. 6D). Collectively, these results indicating that TCF4 promotes miRNAs expression of miR-23a/27a/24 − 2 cluster by interacting to miR-23a/27a/24 − 2 cluster promoter. Then, we investigated that β-catenin whether promotes TCF4 mediated expression of miR-23a/27a/24 − 2 cluster miRNAs. The EMSA results show that overexpression of β-catenin increased the interaction between TCF4 and miR-23a/27a/24 − 2 cluster promoter (Fig. 6E), miR-23a/27a/24 − 2 cluster promoter regulated luciferase transcription (Fig. 6F), and miRNAs expression of miR-23a/27a/24 − 2 cluster (Fig. 6G). Notably, inhibition of the interaction between β-catenin and TCF4 by small molecule LF3 dramatically inhibited β-catenin overexpression-induced upregulation of miR-23a/27a/24 − 2 cluster miRNAs (Fig. 6H). Together, these findings indicating that miR-23a/27a/24 − 2 cluster maintains its expression through β-catenin/TCF4 axis in NSCLC.
Targeting eIF3B pathway dramatically enhanced therapeutic efficacy of PD-1/PD-L1 blockade in lung cancers with high expression of miRNAs in miR-23a/27a/24 − 2 cluster
Given that miRNAs of miR-23a/27a/24 − 2 cluster modulate the expression of key molecules associated with PD-1/PD-L1 blockade therapy efficacy, we investigated whether high expression of miRNAs in miR-23a/27a/24 − 2 cluster leads to resistance to PD-1/PD-L1 blockade. As expected that overexpression of miR-23/27a/24 − 2 cluster miRNAs reduced the sensitivity of tumors to PD-L1 mAb treatment (Fig. 7A) and suppressed the infiltration of CD8+ T cells into tumors in C57BL/6J mice models that constructed using LLC cells (Fig. 7B).
Further, given that miR-23a/27a/24 − 2 cluster maintains its expression through β-catenin/TCF4 axis and downregulates MHC-Ⅰ expression through eIF3B pathway, we investigated whether these axes could be pharmacologically targeted to enhance therapeutic efficacy of PD-1/PD-L1 blockade. Animal experiment results show that blockade of the interaction between β-catenin and TCF4 by LF3 (Fig. 7C) significantly suppressed growth of tumors with high expression of miR-23a/27a/24 − 2 cluster miRNAs compared to control group (Fig. 7D-F). Notably, compared to single drug treatment, combination of LF3 and mAb of PD-L1 more significantly inhibited tumor growth in miR-23a/27a/24 − 2 cluster high expressing lung cancers (Figs. 7D-F). In addition, LF3 treatment suppressed miRNAs expression of miR-23a/27a/24 − 2 cluster (Fig. 7G), PD-L1 expression, and upregulated MHC-Ⅰ expression (Fig. 7H). Notably, that combination of PD-L1 mAb and LF3 more significantly increased infiltration of CD8+ T cells into tumor tissues (Fig. 7I). Then, we compared the promoting effects of targeting the β-catenin/TCF4 axis and targeting the eIF3B pathway on PD-1/PD-L1 blockade therapy. Because, small molecule 4EGI-1 impedes eIF3B binding to the translation initiation assembly and inhibits MITF/eIF3B pathway [20, 22], we used 4EGI-1 as inhibitor of eIF3B pathway. The animal experiment results show that targeting of eIF3B pathway by 4EGI-1 more significantly enhanced PD-L1 mAb therapeutic efficacy in miR-23a/27a/24 − 2 cluster high expressing lung cancer than inhibiting β-catenin and TCF4 interaction by LF3 treatment (Fig. 7J). In addition, we demonstrated that combination of 4EGI-1 and PD-L1 mAb more significantly increased CD8+ T cells infiltrate into tumor tissues (Fig. 7K). We also demonstrated that 4EGI-1 treatment caused upregulation of MHC-Ⅰ expression, but did not suppress miR-23a/27a/24 − 2 cluster-induced high expression of PD-L1 (Fig. 7L). Taken together, these data indicating that targeting eIF3B can significantly enhance the therapeutic efficacy of PD-1/PD-L1 blockade in miR-23a/27a/24 − 2 cluster overexpressing lung cancer.