High expression of TIGAR in NSCLC was associated with malignant proliferation and poor prognosis
To determine the clinical significance of TIGAR in patients with NSCLC, immunohistochemical staining was used to detect TIGAR expression in cancer tissues and adjacent tissues of 35 patients with NSCLC (Fig. 1a, b). The intensity of the immune response to TIGAR was generally higher in most cancer tissues than that in adjacent tissues. In addition, western blot analysis of TIGAR and Ki67 in cancer tissues and adjacent tissues of 10 lung cancer patients (Fig. 1c) showed that the expression of TIGAR in cancer tissues was significantly higher than that in adjacent tissues (Fig. 1d). As an antigen associated with proliferating cells, Ki67 is commonly used to characterize rapidly proliferating tumors. Correlation analysis showed that the expression of TIGAR and Ki67 was significantly correlated in cancer tissues and adjacent tissues, indicating that TIGAR might be related to the malignant proliferation of NSCLC (Fig. 1e). A search of the oncomine database also revealed that TIGAR gene copy numbers were significantly higher in both lung adenocarcinoma and lung squamous cell carcinoma than that in normal lung tissue (Fig. 1f). Moreover, high expression of TIGAR in lung cancer was significantly associated with shorter disease-free survival time (Fig. 1g). Taken together, these results indicated that TIGAR was significantly associated with malignant proliferation and poor prognosis of NSCLC.
TIGAR regulated proliferation and cell cycle of NSCLC cells in vitro
To further determine the role of TIGAR in the proliferation of NSCLC cells, the cell proliferation of A549 and PC9 cells was determined after knocking down TIGAR with shRNA viral particles for one week. As shown in Fig. 2a, the cell proliferation began to significantly decrease on the fifth day after TIGAR knockdown. Cell proliferation was also detected by colony formation and EdU incorporation, and the results showed that TIGAR knockdown in A549 and PC9 cells significantly decreased cell proliferation (Fig. 2c-g). Cell cycle analysis revealed that TIGAR knockdown resulted in cell cycle arrest at s-phase (Fig. 2b). Taken together, these results indicated that TIGAR played an important role in the regulation of tumor cell proliferation.
TIGAR regulated deoxyribonucleotide production in NSCLC cells
To investigate the underlying mechanism of TIGAR in regulating the proliferation of NSCLC cells, we established a stable TIGAR knockdown cell line by infecting A549 cells with virus particles LV-NC and sh-TIGAR, followed by 2 weeks of screening with puromycin (Fig. 3a). The knockdown of TIGAR in the cell line was confirmed by western blot (Fig. 3b). After harvesting cells, 266 differential metabolites were identified by LC-MS untargeted metabolomics analysis, among which 234 metabolites were up-regulated and 318 metabolites were down-regulated (Fig. 3c, Supplementary Table S3). As shown in Fig. 3d, the enrichment of metabolic pathways was plotted by GO and KEGG analysis, with the red line indicating a p-value of 0.01 and the blue line indicating a p-value of 0.05. All the signaling pathways listed are significantly different, with the most significant effects on purine and pyrimidine metabolism. Since the final metabolites of purine and pyrimidine metabolism are several deoxyribonucleotides, metabolomics data were further mined and discovered that the intracellular levels of several deoxyribonucleotides were significantly decreased after TIGAR knockdown compared with controls (Fig. 3e, f).
TIGAR regulated the gene expression in purine metabolism pathway
Furthermore, transcriptomic analysis was performed by reverse transcription of total RNA into cDNA extracted from the TIGAR knockdown stable A549 cell line. Principal component analysis diagram (PCA) showed the similarity degree among the samples of this sequencing, excluding the difference in expression abundance caused by the variation between the samples (Figure S1a). Cluster analysis of the genes screened by sequencing showed that 520 genes were down-regulated and 390 genes were up-regulated after TIGAR knockdown compared with controls (Fig. 4a, Supplementary Table S4). Since the results of metabolomics showed that TIGAR knockdown had the most significant effect on several metabolic pathways, transcriptome data were further used for Gene Set Enrichment Analysis (GSEA) on purine and pyrimidine metabolic pathways (Supplementary Table S5, Table S6). Interestingly, it was found that the genes related to purine metabolism pathway were significantly down-regulated after TIGAR knockdown (Fig. 4b), while the genes related to pyrimidine metabolism pathway were also down-regulated, but did not reach the statistical significant level (Figure S1b).
After being integrated, these results suggested that TIGAR may regulate the expression of genes involved in purine metabolism pathway to maintain intracellular deoxyribonucleotide levels. Therefore, we compared the most significant genes affected in purine metabolic pathway (Figure S1c), and further verified their expression in A549, PC9 and NCI-H1299 cells after TIGAR knockdown. The results showed that TIGAR knockdown decreased several genes in several cell lines, with the most significant effects on RNR subunit RRM2B and mitochondrial thymidine kinase TK2 (Fig. 4c-e).
Subsequently, protein levels were also determined and found that RRM2B was significantly down-regulated after TIGAR knockdown in several cell lines, while RRM1 and RRM2, two other subunits of RNR, were not significantly changed (Fig. 4f, g).
Studies have shown that RNR encoded by RRM2B is a key rate-limiting enzyme in de novo deoxyribonucleotide synthesis pathway in vivo [14]. RNR has a unique catalytic function and is the only catalytic enzyme that can convert ribonucleotides (NDPs) into deoxyribonucleotides (dNDPs) [14]. Moreover, deoxyribonucleotides are substrates for DNA replication, repair and synthesis in vivo. In summary, TIGAR regulated intracellular deoxynucleotide levels through RRM2B to maintain normal DNA replication and tumor proliferation.
TIGAR interacted with the transcription factor YBX1 and participated in its nuclear translocation to activate the transcriptional expression of RRM2B
To further determine the relationship between TIGAR and RRM2B, the correlation between TIGAR and RRM2B gene expression in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) databases was analyzed by the correlation analysis module of GEPIA2 (Gene Expression Profiling Interactive Analysis), and a strong positive correlation was found (Figure S2a, b). Moreover, the determination of TIGAR and RRM2B proteins in cancer tissues and adjacent tissues of lung cancer patients also found that they had a significant positive correlation (Figure S2c, d). The co-localization of TIGAR and RRM2B with high expression in lung cancer tissues was also confirmed with immunofluorescence (Figure S2e).
Since TIGAR knockdown reduced the mRNA level of RRM2B, we speculated that TIGAR could regulate the transcription of RRM2B. Therefore, luciferase reporter gene assay of RRM2B promoter was performed and found that RRM2B gene promoter activity was effectively reduced by TIGAR knockdown in A549, PC9 and HEK293T cells (Figure S3a-c).
However, existing studies have not shown that TIGAR acts as a transcription factor, so the hypothesis of its interaction with a transcription factor has been raised. To explore the potential, we isolated the TIGAR-interacting proteins in A549 cells with MS analysis (Figure S4a). The silver staining indicated that TIGAR was successfully isolated by immunoprecipitation under our experimental conditions and enriched to its interacting proteins (Figure S4b). After the purification of the TIGAR-interacting proteins, we performed MS analysis and identification proteins of two biological replicates by high-throughput LC-MS/MS. We used the label-free quantification (LFQ) incorporated in the MaxQuant software suite to quantify the relative protein abundance, and identified 307 interacting-proteins in total (Supplementary Table S7). The relative abundance (Log2(LFQCtrl/LFQExpt)) was used to construct a scatter plot, which was set to > 1 in both samples to indicate significant differences (Fig. 5a). Finally, 149 proteins were considered as potential TIGAR-interacting proteins, including the transcription factor YBX1.
Next, we used biochemical approaches to test whether the transcription factor YBX1 indeed interacts with TIGAR. To do so, we used co-immunoprecipitation of endogenous and exogenous proteins. Forward and reverse immunoprecipitation and subsequent immunoblotting of TIGAR and YBX1 in A549 cells were performed and found that they did interact with each other (Fig. 5b, Figure S4c). To further confirm their interaction, we performed immunoprecipitation for immunoblotting analysis by overexpressing exogenous tagged proteins. On the one hand, GFP-TIGAR was transfected into A549 cells and anti-YBX1 antibody magnetic beads were used for immunoprecipitation (Fig. 5c). On the other hand, after transfection of FLAG-TIGAR, anti-FLAG antibody magnetic beads were used for immunoprecipitation (Figure S4d). The forward and reverse immunoprecipitation and immunoblotting experiments clearly demonstrated that TIGAR interacted with the transcription factor YBX1. Meanwhile, immunofluorescent staining also showed the colocalization of endogenous TIGAR (green) and YBX1 (Red) in A549 cells (Fig. 5d).
Furthermore, considering that YBX1 activity was mainly determined by its phosphorylation status and the level of nuclear translocation [15, 16], participation of TIGAR in the phosphorylation and nuclear localization of YBX1 was examined. We examined the expression of YBX1 and its phosphorylated form at ser102 in several lung cancer cells after TIGAR knockdown. The results showed that TIGAR knockdown effectively reduced the expression of p-YBX1 without affecting its total protein in A549, PC9, and NCI-H1299 (Fig. 5e). Intriguingly, nucleus-cytoplasm fractionation assay showed that YBX1 was present in both nuclear and cytoplasmic compartments of A549 cells, and TIGAR knockdown significantly decreased YBX1 levels in the nucleus (Fig. 5f). Meanwhile, immunofluorescence staining confirmed that the nuclear staining of YBX1 decreased after TIGAR knockdown (Figure S4f).
Since the transcription factor YBX1 has been reported to be involved in the transcriptional expression of many genes in tumor cells, including TK1, RRM2, TYMS and so on [17]. However, whether the transcriptional expression of RRM2B is also regulated by YBX1 is still unknown. Therefore, we further confirmed that YBX1 can bind to the promoter region of RRM2B and promote its transcriptional activation by chromatin immunoprecipitation (Fig. 5g, h).
These results indicated that TIGAR interacted with YBX1 and participated in the regulation of its biological function and transcription of the downstream gene RRM2B.
The inhibitory effect of TIGAR on NSCLC cell proliferation was dependent on levels of RRM2B and intracellular deoxynucleotide
The previous results confirmed that TIGAR knockdown inhibited tumor cell proliferation and down-regulated the transcriptional expression of RRM2B. Therefore, we hypothesized that TIGAR regulated proliferation of NSCLC cells through limiting availability of deoxynucleotide. We then constructed a plasmid containing the RRM2B transcript sequence (EGFP-RRM2B) and transfected it into TIGAR knockdown A549 and PC9 cells to ensure that RRM2B had been overexpressed (Figure S5a). The proliferation ability of tumor cells was detected by fluorescence observation and flow cytometry analysis after EdU incorporation (Fig. 6a, Figure S5b), combined with colony formation assay (Fig. 6b, Figure S5c). These data showed that overexpression of RRM2B could effectively antagonize the effects of TIGAR knockdown on the proliferation of A549 and PC9 cells.
Since RRM2B functions as a small subunit of RNR through the production of deoxyribonucleotide and TIGAR knockdown reduced the intracellular level of deoxyribonucleotide, we assumed that exogenous supplementation of deoxyribonucleotide could rescued the proliferation of NSCLC cells. Subsequently, A549 and PC9 cells with TIGAR knockdown were supplemented with 50 and 100 µM dNTP for 24 h. The proliferation ability of tumor cells was detected by fluorescence observation and flow cytometry analysis after EdU incorporation (Fig. 6c, Figure S6a), combined with colony formation assay (Fig. 6d, Figure S6b). Similarly, these data showed that dNTP supplementation effectively rescued the proliferation inhibition caused by TIGAR knockdown in A549 and PC9 cells.
The aggravation of DNA damage caused by TIGAR knockdown was rescued by dNTP
Deoxyribonucleotides in cells provide the raw materials for DNA synthesis. Tumor cells with rapid proliferation demand a large supply for deoxyribonucleotide to meet the need for DNA replication and repair. Based on the above results, we considered whether DNA repair was also impaired after TIGAR knockdown. We selected a DNA damage model of lung cancer cells induced by moderate ionizing radiation (IR) to investigate the role of TIGAR in DNA repair after DNA damage. We first investigated the effects of different doses of IR (0, 2.5, 5, 10 Gy) on cell survival of A549 cells with TIGAR knockdown. The results showed that the survival of A549 cells was significantly inhibited after 48 h and 72 h of IR in a dose-dependent manner, and the damage was worsened after TIGAR knockdown (Figure S7a, b).
As a classic marker of DNA damage, single-cell gel electrophoresis is used to directly reflect the damage of single and double strand DNA breaks [18]. Studies have shown that the single and double strands of DNA were rapidly broken after irradiation with 10 Gy IR [19]. However, due to the cell repair mechanisms, most of the damage can be repaired within 2 h, and the rest of the damage can be completely repaired within 24 h. The present results showed that the length of comet tail gradually decreased with time after 10 Gy IR, while TIGAR knockdown increased the length of tail distance, prolong the cell repair time, and increase DNA damage (Fig. 7a, b). Meanwhile, the signature markers γH2AX and p-ATR in DNA damage signaling pathway were also significantly upregulated during DNA damage and gradually decreased with time, while TIGAR knockdown significantly increased the expression of γH2AX and p-ATR, indicating that the damage was aggravated (Fig. 7c, d).
Since dNTP can maintain normal DNA replication to maintain tumor cell proliferation, we predicted that dNTP might also protecte DNA repair. The single cell gel electrophoresis and DNA damage markers were used to detect DNA damage. The results showed that exogenous dNTP reduced the DNA damage caused by IR in a dose-dependent manner, reduced the comet tail distance (Fig. 8a, b), inhibited the up-regulation of γH2AX and p-ATR (Fig. 8c, d), and reduced the fluorescence intensity of γH2AX (Fig. 8e). In particular, it had a significant protective effect on TIGAR knockdown cells.
Taken together, these results suggested that TIGAR regulated the repair process of DNA damage by maintaining the production of deoxyribonucleotide in cells.
TIGAR deficiency inhibited the growth of lung cancer xenografts by reducing the expression of RRM2B in vivo
To investigate the effect of TIGAR on tumor growth in vivo, A549 cells were infected with LV-NC and sh-TIGAR to construct a stable TIGAR knockdown cells, and then subcutaneously xenografted tumor model was established in nude mice. Knockdown of TIGAR significantly inhibited the growth of xenograft tumors as compared with the controls (Figure S8a), with a significant difference in the growth rate of tumors beginning on day 36 (Figure S8c) and a significant reduction in the size and weight of tumors (Figure S8b, d). Immunofluorescence staining further showed that knockdown of TIGAR reduced the number of proliferating cells in the xenografted tumors as compared with the controls (Figure S8e).
Subsequently, we further examined whether RRM2B mediates the effect of TIGAR on xenograft tumor growth in vivo with overexpressing RRM2B in transplanted A549 cells. Overexpression of RRM2B in xenograft tumor cells significantly increased tumor growth rate, size and weight in TIGAR knockdown cells (Fig. 9a-c), which was consistent with the role of RRM2B in mediating TIGAR’s role NSCLC cell proliferation in vitro. These results suggest that TIGAR regulated tumor proliferation and growth through RRM2B in vivo.