NAT10 is upregulated in ICC
Surfing in The Cancer Genome Atlas (TCGA) database, we found NAT10 was significantly upregulated in CCA (P < 0.01) (Fig. 1A), as well as in a variety of other solid tumors of the digestive system in TCGA, including stomach adenocarcinoma (STAD), colon adenocarcinoma (COAD), and rectum adenocarcinoma (READ) (Fig. 1B). There was no difference in overall survival (OS) or disease-free survival (DFS) between CCA patients with high and low NAT10 expression. However, patients with high NAT10 expression had significantly poorer OS and DFS than those with low expression in most solid tumors (Fig. 1C, Supplementary Fig. 1). The possible reasons for this result are the high degree of malignancy of CCA, late stage when the patient diagnosed with no chance of surgery, short survival time, and small number of samples in the database.
The tissue microarray (TMA) of 90 patients with CCA and corresponding adjacent tissues were used to further verify the expression of NAT10 and its clinicopathological features. Data shown that higher NAT10 expression was observed in CCA tissues (Fig. 1D, E), and NAT10 expression was significantly correlated with tumor location, histological grade, and primary tumor stage (Supporting Table 1). qRT-PCR data shown that NAT10 was upregulated in CCA cells (RBE, HuCCT1, and TFK-1) compared to bile duct epithelial cells (HIBEpiC) (Fig. 1F).
NAT10 promotes the growth of ICC in vivo and in vitro
To investigate the functional role of NAT10 in ICC, we constructed three independent short hairpin RNA (shRNA) sequences (shNAT10#1, shNAT10#2, and shNAT10#3). ShNAT10#1 and shNAT10#2 were selected for subsequent experiments own to their silencing efficiency (Fig. 2A, B). In addition, we constructed NAT10-overexpressing cell lines and validated them at the mRNA and protein levels (Supplementary Fig. 2A, B). The CCK8 assay showed that knockdown of NAT10 significantly inhibited the proliferation of ICC cells, whereas overexpression of NAT10 increased the proliferation of ICC cells (Fig. 2C, Supplementary Fig. 2C). Live-cell imaging was used to photograph the cells every 2 h for 120 h, and the data were consistent with the CCK8 (Fig. 2D, E, Supplementary Fig. 2D, E). Colony formation assay was performed to determine the long-term effects of NAT10 on ICC cell proliferation. After 8–12 days, NAT10 knockdown significantly reduced colony formation (Fig. 2F), whereas NAT10 overexpression significantly increased colony formation (Supplementary Fig. 2F). To evaluate the effects of NAT10 on ICC in vivo, we constructed an animal xenograft model by injecting HuCCT1 cells subcutaneously into the left forelimbs of nude mice. Consistent with the in vitro results, the growth rate of NAT10-knockdown xenografts was slower than that of control xenografts (Fig. 2G-I). NAT10 also promotes ICC growth in vivo.
Subcutaneous tumor tissues from xenograft animal models were stained with hematoxylin and eosin (H&E) (Supplementary Fig. 2G), and the knockdown of NAT10 was confirmed by WB and IHC (Fig. 2J, Supplementary Fig. 2H). Data shown that NAT10 promotes ICC proliferation and may be an oncogene of ICC.
CCL2 is a downstream target of NAT10
To explore the role of NAT10 in the development of ICC and identify its downstream targets, we conducted ONT full-length transcriptome sequencing to examine changes after NAT10 knockdown. In RBE cells, 9 and 39 genes were consistently upregulated and downregulated, respectively, by two independent shRNAs (Fig. 3A). We observed good agreement between the two independent shRNAs for the downregulated genes. Gene ontology (GO) analysis showed that differentially expressed genes were involved in apoptosis, cell surface receptor signaling pathways, immune response, secretion, and gene concentration in the extracellular regions and space, suggesting that NAT10 may have an important impact on ICC biology (Fig. 3B). CCL2 was concerned as the most prominent candidate target gene. qRT-PCR and Western blot verified that NAT10 knockdown decreased the mRNA and protein levels of CCL2 (Fig. 3C, D). Additionally, CCL2 expression was significantly decreased in NAT10-knockdown tumors in vivo (Fig. 3E). The data indicate that NAT10 promotes CCL2 expression in ICC both in vitro and in vivo. Based on the acetyltransferase properties of NAT10, we further determine the regulatory mechanism of NAT10 on CCL2. RNA immunoprecipitation-quantitative PCR (RIP-qPCR) and coimmunoprecipitation (COIP) was conducted. The data showed that NAT10 exhibited strong binding and interaction with CCL2 mRNA. The two proteins did not interact (Fig. 3F and G). In summary, CCL2 is a downstream target directly regulated by NAT10.
CCL2 promotes ICC growth in vitro and in vivo
To verify the tumor-promoting function of CCL2, we established stable CCL2-knockdown cell lines, which were validated by western blot (Fig. 4A). Using CCK8 and live-cell imaging assays, we found that CCL2 knockdown significantly inhibited the proliferation of ICC cells (Fig. 4B-D). Additionally, CCL2 knockdown significantly inhibited colony formation (Fig. 4E). To confirm the role of CCL2 in ICC in vivo, we used a xenograft animal model wherein HuCCT1 cells were inoculated subcutaneously into the right forelimbs of nude mice. Consistent with the in vitro results, the growth rate of subcutaneous tumors in the CCL2-knockdown group was slower than that of tumors in the control group (Fig. 4F-H). Therefore, CCL2 promotes ICC growth in vivo. We stained the subcutaneous tumor tissues of xenograft animal models with H&E (Fig. 4I), and the knockout of CCL2 was confirmed by IHC (Fig. 4J).
We next infected NAT10-knockdown ICC cells with a lentivirus carrying CCL2 (Supplementary Fig. 3A) and performed CCK8 assays and live-cell imaging, which showed that CCL2 overexpression partially rescued the loss of proliferation observed in NAT10-knockdown ICC cells (Supplementary Fig. 3B-D). Taken together, CCL2 is a downstream target of NAT10 and plays a role in promoting ICC growth both in vivo and in vitro.
NAT10 polarizes macrophages toward the M2 type through its regulation of CCL2
To study whether ICC cells can cause macrophage polarization and the type of polarization, we cultured RAW264.7 mouse macrophages with conditioned medium from ICC cells and performed qRT-PCR to detect the iNOS and Arg-1 levels after 24 h. Compared with control cells, co-cultured RAW264.7 was polarized (Fig. 5A). We also used a transwell chamber to co-culture ICC and RAW264.7 cells and obtained similar but more significant results (Fig. 5B). We also assessed this by flow cytometry. There was no difference in CD86 expression, representing M1 macrophages, after co-culture; however, there was a significant increase in CD206 expression, representing M2 macrophages (Fig. 5C). These results indicate that ICC cells could polarize RAW264.7 cells toward the M2 type.
CCL2 can polarize macrophages toward the M2 type. Therefore, to assess whether the polarization of RAW264.7 cells to M2 type by ICC cells was dependent on NAT10, we first conducted immunofluorescence staining of the NAT10-knockdown tumors, which showed that the expression of CD86, which represents M1 macrophages in mice, was higher in the knockdown tumors than in the control tumors. By contrast, the expression of CD163, which represents M2 macrophages, was decreased (Fig. 5D). We then knocked down NAT10 in ICC cells and used WB and enzyme-linked immunosorbent assay (ELISA) to detect the levels of CCL2 in the ICC cells and cell supernatants. NAT10 knockdown reduced the expression of CCL2 in both the cells and cell supernatants (Fig. 5E, F).
Lentivirus-mediated gene silencing was used to knock down CCL2 in RBE and HuCCT1 cells (Fig. 5G). Cells were co-cultured with RAW264.7 cells, and the levels of CD86 and CD206 were assessed by flow cytometry. Although ICC cells could still polarize RAW264.7 cells toward the M2 type after CCL2 knockdown, the effect was significantly reduced (Fig. 5H). We also performed immunofluorescence staining of the CCL2-knockdown tumors, and the results were consistent with those of the NAT10-knockdown tumors. The expression of CD86, representing M1 macrophages, was increased in CCL2-knockdown tumors, whereas the expression of CD163, representing M2 macrophages, was decreased (Fig. 5I). These results suggest that ICC cells can polarize macrophages toward the M2 type and that NAT10 plays an important role through its regulation of CCL2.
Berberine can target binding and inhibit CCL2
Based on the anti-ICC function of NAT10 and CCL2, we tried to screen their natural targeted inhibitors. Fortunately, a natural product, berberine (BBR) was concerned, which presents high anti-inflammatory and anticancer activity. Molecular docking shown that the binding affinity of BBR with NAT10 and CCL2. was − 8.3 kcal/mol and − 6.3 kcal/mol, respectively (Fig. 6A). After treated with BBR, HuCCT1 cells were performed using RNA sequencing. There was no significant change in NAT10 expression after BBR treatment, but CCL2 was significantly decreased (Fig. 6B). The data suggested that BBR might exert antitumor effects on ICC by inhibiting CCL2. qRT-PCR and Western blot also present a significantly down-regulation of CCL2 but not NAT10 in mRNA and protein level (Fig. 6C, D). To further verify whether BBR could specifically bind to CCL2, we used surface plasmon resonance (SPR) assay to detect its affinity. The data showed a strong binding of BBR to CCL2, and the concentration gradient trend was significant. There was specific binding with an affinity of 423.4 µM (Fig. 6E, F).
Antitumor effects of BBR on ICC in vitro and in vivo
To demonstrate the effect of BBR on the proliferation of ICC cells, we treated RBE and HuCCT1 cells with different concentrations of BBR (0, 10, 20, 40, 80, 160, and 200 µM) and performed CCK8 assays. BBR significantly reduced the viability of both cell lines. This inhibition was time- and concentration-dependent (Fig. 7A). BBR had a stronger effect on HuCCT1 cells, showing a significant inhibitory effect at 48 hours at a very low dose. We then treated these two cell lines with BBR and conducted live-cell imaging. There were significantly fewer cells in the BBR condition than in the DMSO condition, which was consistent with the results of the CCK8 assay (Fig. 7B, C).
To deeply uncovered the inhibitory effect of BBR on the proliferation of ICC cells, RBE and HuCCT1 cells were treated with BBR for 48 h, and changes in the cell cycle were detected using flow cytometry. After treatment with BBR, the percentage of GO/G1 phase cells increased, whereas the percentage of cells in the S and G2/M phases significantly decreased (Supplementary Fig. 4A, B). We also used flow cytometry to assess apoptosis in RBE and HuCCT1 cells after 48 h of BBR treatment. BBR induced apoptosis in both cell lines (Supplementary Fig. 4C, D). Taken together, the data suggest that BBR exerts antitumor effects by blocking ICC cells in the G0/G1 phase and inducing apoptosis.
To further verified the effect of BBR on tumor growth in vivo, HuCCT1 cells were subcutaneously transplanted into nude mice. When tumors appeared, the mice were randomly divided into two groups, with six tumor-bearing mice per group, and BBR (50 mg/kg/d) or sterile water was administered orally. After 18 days of continuous administration of BBR, tumor size and weight were significantly lower than those in the control group (Fig. 7D-F). The tumor tissues were stained with H&E (Supplementary Fig. 4E), and IHC was used to detect the expression of NAT10 and CCL2. There was no significant difference in the expression of NAT10 between the two groups; however, the expression of CCL2 in the tumors of the BBR group was significantly lower than that in tumors in the control group (Supplementary Fig. 4F), which was consistent with the results of the in vitro experiments. Finally, immunofluorescence was used to detect macrophages in the tumors. Compared to the DMSO group, the expression of CD86, representing M1, was increased in the BBR group, whereas the expression of CD163, representing M2, was significantly decreased (Supplementary Fig. 4G), which was consistent with what we observed in NAT10- and CCL2-knockdown tumors. In summary, BBR inhibits the proliferation of ICC and affects the polarization of macrophages by specifically binding to CCL2, thus playing an antitumor role.