B3galt5 is upregulated in HCC patients
We first examined the mRNA expression of b3galt5 in tumor tissues and adjacent normal tissues in HCC patients using RNA-seq. The mRNA level of b3galt5 in HCC tissues was significantly increased compared with that in paired normal tissues (Figure S1A). Analysis of b3galt5 expression in HCC from the Gene Expression Omnibus dataset GSE1898 also confirmed a consistently higher b3galt5 level in tumor tissues (Figure S1B). Furthermore, we determined the protein level of b3galt5 in HCC samples, which was also dramatically elevated, indicating the upregulation of b3galt5 in HCC tissues (Figure S1C). Importantly, Kaplan-Meier survival analysis revealed that HCC patients with high b3galt5 expression (n=259) were associated with poor overall survival, while low expression (n=111) prolonged overall survival (Figure S1D). Overall, these results indicated that b3galt5 might act as a tumor promoter in HCC.
B3galt5 promotes proliferation and inhibits apoptosis in HCC cells
To explore the biological functions of b3galt5 in HCC cells, we first screened two cell lines, HLE and HCCLM3, with high b3galt5 expression profiles from a series of HCC cell lines (Figure S2A). Then, we silenced b3galt5 in HLE and HCCLM3 cells using specific siRNA. Intriguingly, the proliferation of these two cell lines was significantly inhibited after b3galt5 knockdown, as determined by the CCK-8 assay (Figure S2B). The apoptotic rates of HLE and HCCLM3 cells with b3galt5 silencing were 12.83% ± 0.06% and 16.87% ± 0.22%, respectively, compared with rates of 7.60% ± 0.10% and 12.27% ± 1.83% in siRNA control cells, respectively (Figure S2C and D). Moreover, b3galt5 knockdown disrupted cell cycle progression and induced S-phase arrest (Figure S2E and F). Taken together, these results indicated that b3galt5 promoted proliferation and inhibited apoptosis in HCC cells.
B3galt5 deficiency attenuates DEN/TCPOBOP-induced hepatocarcinogenesis
To further investigate the role of b3galt5 in liver tumorigenesis, we attempted to silence b3galt5 in a DEN/TCPOBOP-induced mouse HCC model. C57BL/6 male mice were treated with DEN at day 14 followed by biweekly injections of TCPOBOP for 16 weeks starting at 4 weeks postpartum. Then, mice were given tail vein injections of shNC AAV or shb3 AAV at 18 weeks old (Figure S3A). As expected, b3galt5 expression was markedly decreased in shb3 AAV-treated mice (Figure S3B and C), and the incidence of liver cancer in shb3 AAV mice was lower than that in shNC AAV mice (Figure S3D). Histopathological analysis of livers showed that there were more fat deposits and more severe dysplasia in shNC AAV mice than in shb3 AAV mice (Figure S3E). Quantitative analysis showed that shb3 AAV mice had an ~60% decrease in tumor number (Figure S3F). In addition, although the difference was not statistically significant, the tumor volume showed a decreasing trend in shb3 AAV mice (Figure S3G). The liver weight/body weight ratio was similar in both groups of mice (Figure S3H). We further assessed the IHC scores of PCNA and cleaved caspase-3 in shNC AAV and shb3 AAV mice, which were 5.14 ± 1.10 vs 1.43 ± 0.78 and 2 ± 0 vs 7.83 ± 1.93, respectively, indicating massive remission of proliferation-positive cells and induction of apoptosis in the livers after b3galt5 silencing (Figure S3I). In addition, hepatic fibrosis was markedly alleviated in shb3 AAV mice with b3galt5 deficiency, as determined by α-smooth muscle actin (α-SMA) staining (Figure S3I).
Furthermore, we investigated liver tumorigenesis in b3galt5-/- (KO) and wild-type (WT) mice under DEN/TCPOBOP challenge (Figure 1A). Mice were sacrificed, and whole livers were collected and analyzed 18 weeks after DEN injection. As expected, the protein level of b3galt5 was effectively decreased in KO mice (Figure 1B and C). Notably, KO mice developed fewer tumors in the liver than WT mice (Figure 1D). The KO livers exhibited smaller neoplastic lesions and attenuated cirrhosis compared with control littermates, as shown by H&E staining (Figure 1E). Quantitative analysis revealed that the number of tumor nodules in KO mice was only 3.07 ± 0.50 per liver, which was significantly lower than that in WT mice (12.14 ± 2.83 per liver, Figure 1F). The tumor volume of KO mice was also markedly reduced compared to that of WT mice (30.33 ± 8.65 vs 131.9 ± 43.58 mm3, respectively) (Figure 1G), accompanied by an ~20% reduction in the liver/body weight ratio (Figure 1H). In addition, histopathological analysis demonstrated markedly inhibited proliferation, a significant induction of apoptosis, and attenuatedhepatic fibrosis in the livers of KO mice compared to WT mice (Figure 1I).
Collectively, these results revealed that b3galt5 deficiency attenuated DEN/TCPOBOP-induced HCC in mice.
B3galt5 promotes glycolysis in HCC
To investigate the underlying molecular mechanism of the tumorigenic effect of b3galt5, we performed metabolomics analysis of liver cancer cells from shb3 AAV and shNC AAV treated mice. The results revealed that the metabolites were significantly changed when b3galt5 was knocked down (Figure S4A). Kyoto Encyclopedia of Gene and Genomes (KEGG) enrichment analysis identified that multiple carbohydrate metabolism signaling pathways were enriched, including pyruvate metabolism, pentose and glucuronate interconversion, amino sugar and nucleotide sugar metabolism and OXPHOS (Figure S4B). Next, we performed proteomic sequencing of liver cancer cells from shb3 AAV- and shNC AAV-treated mice. A number of key proteins closely associated with OXPHOS, including Cox7a2l[23], Ndufaf3[24], Cmc1[25] and Sdhaf4[26], were significantly upregulated upon b3galt5 knockdown (Figure S4C). To further explore the function of the b3galt5-regulated proteins, we conducted KEGG enrichment analysis. Our results showed that proteins involved in amino nucleic acid sugar metabolism, glycolysis and gluconeogenesis, and OXPHOS were markedly enriched in b3galt5 knockdown mice (Figure S4D), suggesting the potential roles of b3galt5 in glucose metabolism.
Inspired by metabolomics analysis and proteomic sequencing, we then examined whether b3galt5 regulated glycolysis in HCC. Compared with control mice, knockout of b3galt5 resulted in a significant decrease in key glycolytic enzymes, including HK2, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) and LDHA (Figure 2A). Immunohistochemical staining also confirmed that the expression of LDHA, HK2, and PFKFB3 was decreased in the livers of KO mice (Figure S5A-C). Additionally, ablation of b3galt5 led to a significant reduction in lactate production and decreased lactate dehydrogenase, hexokinase, and phosphofructokinase activity in the serum of KO mice (Figure 2B-E). It should be noted that the absence of b3galt5 did not lead to significant changes in total pyruvate kinase activity (Figure 2F), which might be because pyruvate kinase has a variety of isoenzymes, while pyruvate kinase M2 is the main form expressed in liver tissue[27].
In HCCLM3 and HLE cells, silencing b3galt5 likewise decreased the expression of several key glycolytic enzymes, such as HK2, PFKFB3, LDHA and pyruvate dehydrogenase kinase 1 (PDK1) (Figure 2G). Conversely, overexpression of b3galt5 in LO2 and HUH7 cells increased the expression of these glycolytic enzymes (Figure 2H). Moreover, b3galt5 knockdown significantly reduced lactate production, while b3galt5 overexpression had the opposite effect (Figure 2I). Accordingly, we observed an obviously increased rate of ECAR, as indicated by the increased glycolysis rate and glycolysis capacity in b3galt5-overexpressing cells (Figure 2J and K). Knockdown of b3galt5 in HCCLM3 and HLE cells led to opposing effects (Figure 2L and M). In addition, we also detected the OCR in HCC cells. As expected, knockdown of b3galt5 in HCCLM3 and HLE cells led to a significant increase in the OCR. In particular, the maximal respiration in HLE cells increased from 42.1 ± 3.8 to 49.1 ± 6.2 pmol/min, and in HCCLM3 cells, it increased from 31.0 ± 3.9 to 64.7 ± 5.5 pmol/min (Figure 3A-C). In contrast, a significant reduction in maximal respiration in LO2 and PLC/PRF5 cells was observed after b3galt5 overexpression (Figure 3D-F). All these compelling results suggested that b3galt5 promoted glycolysis in HCC.
B3galt5 promotes glycolysis in HCC via the mTOR/p70s6k pathway
The mTOR/p70s6k pathway is one of the main signaling pathways that regulates glycolysis. To clarify its role in the promotion of glycolysis by b3galt5, we detected the total and phosphorylated protein levels of mTOR and its downstream target, p70s6k. Immunoblot analysis showed that both phosphorylated and total protein levels of mTOR, as well as p70s6k, were downregulated in b3galt5-KO liver cells (Figure 4A). Consistent results could also be observed in HLE and HCCLM3 cells (Figure 4B). Conversely, in b3galt5-overexpressing LO2 and HUH7 cells, total and phosphorylated mTOR and p70s6k levels were accordingly upregulated, except for phosphor-p70s6k levels in LO2 cells, which were undetectable possibly be due to its low endogenous expression (Figure 4C). Conversely, we observed that total and phosphorylated mTOR and p70s6k levels were upregulated in b3galt5-overexpressing LO2 and HUH7 cells, but phosphor-p70s6k levels were not detectable in LO2 cells (Figure 4C). As expected, these changes were in line with the expression of key glycolytic enzymes. To further verify the clinical correlation between b3galt5 and p-p70s6k, we performed immunohistochemistry staining in a 90-dot tissue microarray (Figure S6A). Encouragingly, a significant positive correlation between b3galt5 and p-p70s6k levels was confirmed in HCC samples (Figure S6B).
Then, we examined whether activation of mTOR/p70s6k mediated the effects of b3galt5 on glycolysis. For this purpose, we treated HCC cells with a p70s6k activator or inhibitor in combination with b3galt5. The results indicated that the expression of key glycolytic enzymes, including HK2, LDHA, PFKFB3, and PKM2, in b3galt5-silenced HLE cells could partially be rescued by 3BDO, an activator of p70s6k (Figure 5A). Conversely, the inhibition of p70s6k by PF-4708671 reduced the levels of these key glycolytic enzymes in b3galt5-overexpressing LO2 and PLC/PRF5 cells (Figure 5B and C). To further confirm the role of p70s6k in b3galt5-mediated glycolysis, we carried out live monitoring using a Seahorse XF Extracellular-Flux Analyzer for ECAR. Knockdown of p70s6k markedly inhibited the glycolysis rate and glycolysis capacity (Figure 5D-F). Overexpression of p70s6k in b3galt5 knockdown cells rescued the glycolysis rate and glycolysis capacity (Figure 5G and H). Collectively, these results illustrated that b3galt5 promoted glycolysis by activating the mTOR/p70s6k pathway.
B3galt5 regulates the N-linked glycosylation of mTOR/p70s6k
It has been reported that glycosylation mediates protein stability, function, and transport. We therefore investigated whether the activation of mTOR/p70s6k is regulated by glycosylation. RCA-lectin is a reagent that is generally used to identify the galactose or N-acetylgalactosamine residues of membrane glycoconjugates[28]. Immunoblot analysis revealed that b3galt5 knockdown significantly blocked the glycosylation modification of p-mTOR, p70s6k, and p-p70s6k, while overexpression of b3galt5 enhanced their glycosylation modification (Figure 6A and Figure S7A). In the immunofluorescence assay, we confirmed a high fluorescence intensity of p-mTOR and p-p70s6k after B3galt5 overexpression, accompanied by the high fluorescence intensity of RCA-lectin, indicating the presence of galactose in p-mTOR and p-p70s6k (Figure 6B and Figure S7B). In contrast, b3galt5 knockdown correspondingly suppressed the fluorescence intensity (Figure 6C and Figure S7C).
The main receptors of b3galt5 are GlcNAc and GalNAc, which are involved in N-linked and O-linked glycosylation. When treating HCC cells with an N-linked glycosylation inhibitor, namely, tunicamycin, or an O-linked glycosylation inhibitor, that is, benzyl-α-GalNAc, only tunicamycin treatment led to decreased expression and phosphorylation of p70s6k in LO2 and HUH7 cells (Figure 7A and B). Although tunicamycin enhanced mTOR expression in HUH7 cells, phosphorylation of mTOR was still decreased in both cell lines (Figure 7A and B). However, this suppressive effect was not observed when cells were treated with benzyl-α-GalNAc (Figure 7A and B).
Above all, these findings suggested that b3galt5 N-glycosylated and activated the mTOR/p70s6k pathway in HCC cells.