O -GlcNAcylation is up-regulated in CCA
We started by investigating the clinical relevance of O-GlcNAcylation in CCA. We first analyzed the O-GlcNAc, and the expression of OGT and OGA in 21 pairs of human resected tumor tissues and adjacent normal bile duct using immunohistochemistry (IHC) (Fig. 1A). The levels of O-GlcNAc and OGT were distinctively elevated in tumor tissues compared with normal tissues, consistent with the IHC staining scores in these CCA patients (P < 0.001, Fig. 1B). However, the level of OGA did not show distinctive changes between the two groups (Fig. 1A, 1B). We then analyzed the O-GlcNAc expression levels in additional 15 peritumoral/tumor tissue pairs from CCA patients. Again, O-GlcNAcylation and OGT were identified to be up-regulated in tumor tissues (Fig. 1C and Fig. S1), and quantitative analysis of the signals confirmed that the increases are statistically significant (P < 0.01 and 0.05, respectively by Student’s t-tests, two-tailed) (Fig. 1D). Notably, mRNA expression of OGT and OGA was escalated from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets (GSE32879, GSE107943, GSE119336, GSE76297) (Fig. S2A, 2B). CCA patients with high levels of OGT and low levels of OGA gene expression displayed worse overall survival (OS) according to the Kaplan-Meier survival analysis, indicating the pivotal role of O-GlcNAcylation in CCA (Fig. S2C, 2D). We further assessed the O-GlcNAc, OGT and OGA expressions in three human CCA cell lines, namely HCCC-9810, RBE and HuCCT1, and the Human Intrahepatic Biliary Epithelial Cell (HIBEpiC) control cell line (Fig. 1E). O-GlcNAcylation level increased in all CCA cells compared with HIBEpiC cells, while mRNA expression of OGT and OGA, as measured by quantitative real-time PCR (qRT-PCR), exhibited varied expression levels (Fig. 1F). These data altogether pinpointed the up-regulation of O-GlcNAcylation as a common event in CCA samples.
Global O -GlcNAcylation contributes to CCA cell proliferation
Previous reports by the Wongkham group indicated the significance of O-GlcNAcylation in controlling the metastatic ability of CCA cells via nuclear translocation of NF-κB and heterogeneous nuclear ribonucleoprotein-K (hnRNP-K).13–15 We therefore postulated that modulation of O-GlcNAc levels would alter CCA oncology phenotypes (i.e., proliferation, apoptosis, cell cycle), by targeting pathways known to regulate CCA progression. To do so, we first used chemical tools to inhibit OGT and OGA. Ac45SGlcNAc (designated as 5S thereof) is a known inhibitor of OGT that acts as a metabolic precursor to form uridine diphosphate activated precursor-5SGlcNAc (UDP-5SGlcNAc), and OGA can be selectively and effectively inhibited by Thiamet-G (designated as TMG thereof) (Fig. 2A).16,17 Treatment of all three CCA cell lines with 5S or TMG led to the robust decrease or increase of O-GlcNAc modification level on proteins, in a time- and dose-dependent manner (Fig. 2B, Fig. S3A, S4A and S5), in accordance with previous observations. At the same time, cellular viability was assessed using a cell counting kit-8 (CCK-8). The overall cytotoxicity in HuCCT1 cells was governed by the inhibition of OGT rather than OGA, suggesting that lowering O-GlcNAcylation will induce cell death (Fig. 2C). Similar effects were also observed in RBE and HCCC-9810 cells in a dose-dependent manner (Fig. S3B, S4B). Moreover, suppression of OGT promoted CCA cell apoptosis, as evidenced by the increased percentage of both early apoptosis (FITC+/PE−) and late apoptosis (FITC+/PE+) after OGT silencing upon 5S incubation (Fig. 2D, Fig. S3C). No remarkable effects of TMG as an anti-apoptotic inhibitor were detected (Fig. 2D, Fig. S3C). Mounting reports implied that O-GlcNAc plays a multifaceted role during the cell cycle, and incongruousness arises partly due to different physiological cues.18 We examined the cell cycle distribution of HuCCT1 and RBE cells using flow cytometric analysis after incubation with 5S or TMG at varied concentrations (Fig. 2E, Fig. S3D). Interestingly, cell cycle distribution of HuCCT1 cells was majorly found in the G2/M phase, while RBE cells were arrested in the S phase when treated with 5S, but not under TMG-treated scenarios (Fig. 2E, Fig. S3D). It is therefore sufficient to conclude that an appropriate O-GlcNAc level is critical for all cell cycle phases. Additionally, we blotted the classical biomarkers for cell cycle and apoptosis after perturbation of O-GlcNAcylation using 5S and TMG, respectively (Fig. 2F, 2J, Fig. S3E). There were no notable differences in these biomarkers for the TMG-treated group, yet the 5S-treated group presented global up-regulation on apoptosis markers and down-regulation on the cell cycle checkpoints (Fig. 2F, 2J, Fig. S3E). To further elucidate the correlation between O-GlcNAcylation and cancer progression, we complemented three independent small interfering RNAs (siRNAs) for OGT or OGA knockdown (Fig. 2G, Fig. S3F), all of which showed potent knockdown efficacy with the corresponding O-GlcNAc processing enzymes (i.e., OGT or OGA), meanwhile maintaining the desired catalytic activities (Fig. 2H, Fig. S3G). Clonogenic assay both qualitatively and quantitatively validated that knockdown of OGT in HuCCT1 and RBE cells clearly reduced cell proliferation and presented an anti-tumor effect (Fig. 2I, Fig. S3H). These biological characterizations reciprocally integrate with chemical tools, emphasizing that global O-GlcNAcylation may promote the development and progression of cholangiocarcinoma.
Chemical enrichment and profiling of intact O -GlcNAcylated glycopeptides in CCA
Since imbalanced O-GlcNAcylation impacts the process of cancer progression in CCA, we next explored to systematically enrich, identify and profile intact O-GlcNAcylated glycopeptides with matched information on glycosylation sites and glycan compositions. We adopted Click-iG, a comprehensive platform that amalgamates metabolic oligosaccharide engineering (MOE) of selected, clickable unnatural sugar probes for O-GlcNAcylated protein enrichment, and a customized pGlyco3 search engine for intact glycopeptide annotation.19,20 In brief, azidosugars were metabolically incorporated into various glycans. The azido-containing glycoproteins were reacted with a three-module alkyne-photocleavable linker-biotin tag (alkyne-PC-biotin), digested with trypsin, and enriched using streptavidin beads. After photocleavage with 365 nm ultraviolet (UV), the released click-labeled glycopeptides were subjected to liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis, with preferred glycopeptide fragmentation strategies, namely, stepped collision energy based higher-energy collisional dissociation (sceHCD) followed by product-dependent electron transfer/higher-energy dissociation (sceHCD-pd-EThcD) (Fig. 3A). 1,6-di-O-propionyl-N-azidoacetylgalactosamine (1,6-Pr2GalNAz), an optimized monosaccharide O-GlcNAc chemical reporter with minimal nonspecific S-glyco-modification and cytotoxicity, was employed in the experiment (Fig. S6).21 We first evaluated the metabolic efficacy of 1,6-Pr2GalNAz in the CCA and HIBEpiC cell lines and observed azidosugar incorporation in a dose- and time-dependent manner (Fig. 3B, Fig. S7A). Significant fluorescence labeling at nucleocytoplasmic regions was achieved when 1,6-Pr2GalNAz-treated cells were permeabilized and reacted with alkyne-AZDye-488, via the ligand-assisted copper (I)-catalyzed azide-alkyne cycloaddition (CuAAC), in agreement with the localization of O-GlcNAc glycosylation (Fig. 3C, Fig. S7B). Similar quantitative data on all four cell lines were observed by flow cytometry (Fig. 3D). In view of these results, HuCCT1 and HIBEpiC cells incubated with 200 µM 1,6-Pr2GalNAz for 48 h were used as the standardized condition for glycoproteomic analysis.
By using the established streamlined MS data analysis and annotation procedure (Fig. S8), we then mapped a total of 1170 O-GlcNAc sites on 368 O-GlcNAcylated proteins from three replicate experiments using the Click-iG workflow (Fig. 3A, 3E, Fig. S9A, S10A). In combination, the Click-iG strategy yielded pan-scale intact glycopeptide information (i.e., intact glycosites, glycosites, glycoproteins) on HuCCT1 and HIBEpiC cells (Fig. S10B). For instance, a total of 1872 intact glycosites were identified in HuCCT1 cells, of which 1094 overlapped with those identified in HIBEpiC cells, indicating the cell-type specific feature of protein glycosylation (Fig. S10B, left panel). By glycan classification, we identified a total of 3164 intact glycosites, which also consisted of 1421 intact N-linked glycosites and 573 intact mucin-type O-linked glycosites (Fig. S10C). Sequence visualized using a probabilistic approach around the identified glycosites revealed the typical sequon/motif for O-GlcNAcylation, mucin-type O-linked glycosylation and N-linked glycosylation (Fig. S9B, S10D). Remarkably, the Click-iG also provided in-depth glycan type and composition information from intact glycosites (Fig. S11A, S11B, S12A and S12B). Gene Ontology (GO) analysis showed that the identified O-GlcNAcylated proteins concentrated in the nucleocytoplasmic region (Fig. S9C). In addition, proteins carrying 23 up-regulated (fold change > 1.50, P < 0.05) and 36 down-regulated (fold change < 0.67, P < 0.05) O-GlcNAc sites in HuCCT1 cells were potently enriched using Click-iG (Fig. 3F, Table S1, S2), of which many protein regulators involved in the cell cycle and growth, as well as transcriptional processes, were identified to be O-GlcNAcylated (Fig. 3G, Fig. S13). These results, collectively, demonstrate that Click-iG enables simultaneous and comprehensive profiling of O-GlcNAcylation (and other types of glycosylation) in the CCA and HIBEpiC cell lines, with intact glycosite level resolution.
Keratin 18 is mainly O -GlcNAcylated at Ser 30
Keratins play a pivotal role in differentiation and tissue specialization and are highly regulated among various epithelia in a cell-specific manner.22 We noticed that Keratin 18 (K18) is on the list of up-regulated O-GlcNAcylated proteins in HuCCT1 cells compared to HIBEpiC control cells (Fig. 3F, Table S1, S2). Keratin 18 plays various roles in intracellular scaffolding and cellular processes and is highly associated with malignant phenotype in digestive epithelia.23,24 Post-translational modification (PTM) including sumoylation,25 acetylation/methylation,26O-GlcNAcylation27 and its reciprocal cross-talk with phosphorylation28 on K18 have been reported in previous works. To confirm the O-GlcNAcylation of K18, we incubated HuCCT1 and HIBEpiC cells with 1,6-Pr2GalNAz for 48 h, reacted with alkyne-biotin using CuAAC, and conducted pull-down procedures with streptavidin beads. Immunoblotting with anti-K18 demonstrated that K18 was modified with azidosugars (Fig. 4A, left panel). To further verify the labeling results, we treated both cell lysates with a permissive β-1,4-galactosyltransferase mutant (Y289L GalT1), which transfers N-azidoacetylgalactosamine (GalNAz) from its uridine diphosphate activated precursor (UDP-GalNAz) to O-GlcNAc residues.29 Subsequent chemoselective click reaction with alkyne-biotin followed by streptavidin enrichment proved O-GlcNAcylation occurred on endogenous K18 (Fig. 4A, right panel). The extent of O-GlcNAc modification on K18 in HuCCT1 cells was semi-quantitatively measured by labeling azides with alkynylated polyethylene glycol 5000 (alkyne-PEG5KD) in a mass shift assay. The stoichiometric ratio for O-GlcNAcylation, whether in O-β-N-azidoacetylglucosamine (O-β-GlcNAz) or in O-β-GlcNAc-β-1,4-GalNAz form, was approximately 50% (Fig. 4B). Based on Click-iG, we mapped four O-GlcNAc modification sites (Ser 15, Ser 18, Ser 30 and Ser 31) on K18, of which Ser 30 and Ser 31 were previously reported. Interestingly, O-GlcNAcylation on Ser 49 was previously reported yet not found in our experiment setting.27 Of note, all four sites identified are well conserved in Homo sapiens, Mus musculus and Rattus norvegicus. (Fig. S14A, S14B). To further determine the relative abundance of O-GlcNAc modification on these sites, the FLAG-tagged K18 mutants, K18S15A, K18S18A, K18S15/18A, K18S30A, K18S31A, K18S30/31A and K18S15/18/30/31A (K184A), were transfected into HEK293T cells. The incorporated azides, either via MOE with 1,6-Pr2GalNAz, or Y289L GalT1 chemoenzymatic labeling, were reacted with alkyne-biotin for streptavidin capture and immunoblot analysis. Ser 30 appears as the major O-GlcNAcylation site in K18 (Fig. 4C, 4D), in line with our chemical proteomic analysis results (Fig. 3F). Alternatively, we pulled down with anti-FLAG beads for K18 enrichment, and then measured the O-GlcNAc level for FLAG-K18WT or FLAG-K18S30A, using biotinylated signals introduced via the above-mentioned glyco-analytical methods. A significant loss of O-GlcNAcylation for FLAG-K18S30A compared with FLAG-K18WT was observed (Fig. 4E and 4F). In addition, sceHCD-pd-EThcD based LC-MS/MS analysis annotated the K18 peptide with amino acid 28–45 (PVSSAASVYAGAGGSGSR) as an O-GlcNAcylated peptide at Ser 30 (Fig. 4G).
O -GlcNAcylation of K18 promotes CCA proliferation and progression in vitro and in vivo
With the detailed glycosylation information for K18 at hand, we next asked whether K18 O-GlcNAc modification would impact cholangiocarcinoma phenotype(s). We first examined the K18 O-GlcNAcylation levels among typical human CCA cell lines and the normal HIBEpiC cell line. Elevated K18 O-GlcNAcylation was observed in all three CCA cell lines but not in HIBEpiC cells (Fig. 5A). To scrutinize the importance of Ser 30 O-GlcNAcylation, we generated stable CCA cell lines with three independent targeting-resistant short hairpin RNA (shRNA) for K18 knockdown (Fig. S15A, S15B, S16A and S17A), and then recovered K18 expression using either FLAG-K18-WT or FLAG-K18-S30A (Fig. 5B, Fig. S15C, S16B and S17B). A systematic evaluation of KRT18 mRNA expression and knockdown efficiency led to shK18-2 as the optimal construct (designated as shK18 thereof). CCK-8 and colony formation assays indicated that depletion of K18 gravely inhibited cell proliferation in HuCCT1 cells, which was rescued by re-expression of FLAG-K18-WT rather than FLAG-K18-S30A (Fig. 5C, 5D). Similar results were also observed in RBE and HCCC-9810 cells (Fig. S16C, S16D, S17C and S17D). These data imply that Ser 30 O-GlcNAcylation is functionally crucial in CCA proliferation in vitro. We next assessed the cell cycle distribution for each rescue cell line and found that FLAG-K18-S30A-rescued HuCCT1, RBE and HCCC-9810 cells showed an increase in S and G2/M phase arrest, partially in conformity with a previous report that O-GlcNAc on K18 is known to increase upon G2/M phase arrest (Fig. 5E, Fig. S16E, S17E).30 Furthermore, immunoblot analysis on cell cycle biomarkers for these rescued cell lines displayed a similar correlation (Fig. 5F, Fig. S16F, S17F).
Given that O-GlcNAcylation on K18 was reported to regulate its solubility, filament organization, and stability,31 we tested whether the stability of K18 is modulated by O-GlcNAcylation at Ser 30. Previous reports from the Rajiv lab exhibited that the half-life for K18 is regulated by O-GlcNAcylation in human hepatocytes (Chang) cell line.28,31 The CCA cell lines were treated with cycloheximide (CHX) to block protein synthesis for an immunoblot chase assay. OGT inhibition by 5S considerably accelerated the K18 degradation, while silencing of OGA by TMG poses minimal effect on its rate of decay (Fig. 5G, Fig. S16G, S17G). Likewise, FLAG-K18-WT was more stable than FLAG-K18-S30A after 8 h of protein lifespan (Fig. 5H, Fig. S16H, S17H). Correlatively, an increased level of ubiquitination was observed in FLAG-K18-S30A (Fig. 5I, Fig. S16I, S17I). These findings indicate that K18 stability was enhanced through up-regulation of its O-GlcNAcylation at Ser 30, as well as the inhibition of the corresponding ubiquitination.
To decipher the effect of K18 O-GlcNAcylation on tumor growth in vivo, we injected BALB/c nude mice with shRNA negative control with an empty vector (shNC + Mock), shK18, shK18 + WT, shK18 + S30A stable HuCCT1 cell lines, and quantitatively measured tumor formation. K18 depletion and its mutant at Ser 30 greatly repressed tumor growth rate, tumor size/weight, and the Ki67 positive percentage (a marker to determine cancer cell proliferation) (Fig. 5J-M, Fig. S18A, S18B). In parallel, tumor tissues dissected from HuCCT1 cells expressing FLAG-K18-WT showed a higher level of K18 O-GlcNAcylation than the corresponding cells expressing FLAG-K18-S30A (Fig. 5N). Luckily, we observed that both the protein expression level and the O-GlcNAcylation level for K18 were significantly enhanced when advancing toward the clinical CCA tumor tissues compared with adjacent normal tissues (Fig. 5O, Fig. S19). These results are able to recapitulate most of the effects observed in vitro and solidify the hypothesis that O-GlcNAcylation of K18 promotes CCA progression in vivo.
K18 O -GlcNAcylation promotes K18-Isocitrate dehydrogenase interaction to regulate the TCA cycle in CCA.
O-GlcNAcylation serves as a nutrient rheostat in a myriad of physiological contexts, especially in metabolically active organs such as the liver. The tight interconnection between O-GlcNAcylation and glucose metabolism prompted us to explore the mechanistic insight as to how K18 O-GlcNAcylation affects its interaction.32 We transfected HuCCT1 shK18 stable cell line with FLAG-K18WT or FLAG-K18S30A with equivalent protein expression level, co-immunoprecipitated the K18-interacting proteins with anti-FLAG beads and subjected to LC-MS/MS analysis (Fig. S20A, S20B). We identified 858 up-regulated (fold change > 1.50, P < 0.05) interacting proteins in FLAG-K18WT HuCCT1 cells compared to FLAG-K18S30A cells (Fig. 6A), the majority of which were closely related to the TCA cycle shown by the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis (Fig. 6B). To further elucidate the K18 binding partners in the TCA cycle, we analyzed 8 out of 13 up-regulated TCA enzymes, by immunoblotting with their corresponding antibodies (Table S3). We noticed an apparent signal decrease in isocitrate dehydrogenase (IDH) families, including IDH2, IDH3A, IDH3B and IDH3G, when O-GlcNAc modification at Ser 30 of K18 was functionally amputated (Fig. 6C). Similar results were observed in RBE-rescued cells (Fig. S21). The IDH enzymes are responsible for the oxidative decarboxylation of isocitrate. To further corroborate these results, we performed relative quantification of cellular metabolites using LC-MS/MS analysis. We found that in the absence of O-GlcNAcylation at Ser 30 on K18, the rescued cells accumulated higher levels of citrate, aconitate and isocitrate, but lower levels of pyruvate, α-ketoglutarate, succinate, fumarate and malate, compared to K18 wild type rescued cells (Fig. 6D). These data suggest a strong correlation between K18 O-GlcNAcylation and IDH catalytic activities. In toto, we conclude that in cholangiocarcinoma, up-regulated OGT level will inevitably enhance the K18 O-GlcNAcylation, which increases its interaction with isocitrate dehydrogenases in the TCA cycle, one of the major metabolic pathways to regulate glucose metabolism, to coordinate the cell proliferation and CCA progression (Fig. 6E).