TNF-related apoptosis-inducing ligand (TRAIL) resistance, including nongenetically acquired tolerance in cancer persister cells, is a major obstacle to translating TRAIL therapy into patients with cancer. However, the underlying mechanisms remain to be elucidated. Here, we show that DR4/TRAIL-R1 is O-GlcNAcylated at Ser424 in its death domain to mediate both apoptosis and necrosis upon TRAIL ligation. We found that DR4–Ser424 mutations, identified from our cell-based functional screen using a cancer patient–derived cDNA expression library and from The Cancer Genome Atlas, caused TRAIL resistance in various human cancer cell lines. Using O-GlcNAc transferase knockdown cells, DR4-preferred versus DR5-preferred cancer cells, and a DR5-neutralizing antibody, we evaluated the essential role of DR4-specific O-GlcNAc modification in TRAIL cytotoxicity. In contrast to DR4, DR5 was not O-GlcNAcylated by TRAIL treatment, discriminating DR4 from DR5-mediated signaling. Apart from genetic changes in DR4-Ser424, we further classified various cancer cell lines originated from stomach, colon, lung, and glioblastoma according to their sensitivity to and receptor preference upon TRAIL death signaling and generated TRAIL-tolerant persister-derived DLD-1PER cells. Among these, we discovered that DR4 was not modified by O-GlcNAc in most of the TRAIL-resistant cancer cells and DLD-1PER cells. Interestingly, promoting DR4 O-GlcNAcylation intentionally using 2-deoxy-d-glucose or a high concentration of glucose sensitized those resistant cancer cells to TRAIL. The O-GlcNAcylation–defective DR4 failed to form DISC/necrosome and could not translocate to aggregated platforms for receptor clustering. Our findings demonstrate that DR4 O-GlcNAcylation is crucial for TRAIL death signaling, providing new opportunities for TRAIL therapy overcoming TRAIL resistance in cancers.

Significance:

This study reports that a novel posttranslational modification by O-GlcNAcylation of one of the two human TRAIL receptors with a death domain, TRAIL-R1 (DR4), plays a crucial role in enabling both apoptotic and necroptotic cell death induction by TRAIL.

TNF-related apoptosis-inducing ligand (TRAIL) has an attractive ability to kill cancer cells selectively (1, 2). TRAIL research has expanded to the study of its cognate receptors and the development of various TRAIL receptor agonists (TRA) as therapeutic candidates for cancer prevention (3). However, clinical trials of TRAIL or TRAs have failed to achieve a beneficial anticancer activity because many primary cancers are resistant to TRAIL (4–6). In some cases, small subpopulations of drug-sensitive cancer cells also acquire a nonmutational drug-tolerant phenotype and enter a “persistent state” due to heterogeneity within cancer cell populations (7–9). Thus, the use of genetic or proteomic profiling to elucidate the mechanisms that cause TRAIL resistance in cancer cells (10) or the combination of TRAIL with radiotherapy or chemotherapy (11) has been explored in an effort to develop innovative TRAIL-based therapeutic concepts.

Unlike the single functional TRAIL receptor in murine (12), there are two TRAIL death receptors in humans, DR4/TRAIL-R1 and DR5/TRAIL-R2, which mediate TRAIL-induced apoptosis, necrosis, and gene-activating pathways including NF-κB signaling (13–17). Several posttranslational modifications (18, 19) and spatial dynamics (20) of the receptors have been reported. In addition, cancer cells show different preferences for receptor signaling in a cell type–dependent manner (21). Elucidating the differences between two receptors in terms of their activation and signal transduction within the pathophysiologic context is one of the main current concerns in the field of TRAIL research. Thus, the specific targeting and manipulating each receptor by using agonistic/antagonistic/neutralizing antibodies and TRAIL variants selectively binding and activating to either receptor have been considered for the TRAIL experiments and development of clinical therapies (3, 4, 22, 23). For these research avenues, the DR4-dominant (e.g., DLD-1 human colorectal adenocarcinoma cells) and/or DR5-dominant (e.g., SK-Hep1 human hepatic adenocarcinoma cells) type of cells has been employed.

The O-linked β-N-acetylglucosamine (O-GlcNAc) modification, which is termed O-GlcNAcylation, is a conserved posttranslational modification at serine/threonine residues (24). This modification serves as a nutrient sensor for metabolic status and is responsible for numerous cellular signaling events and disease processes (25–27). UDP-GlcNAc derived from glucose via the hexosamine biosynthetic pathway (HBP) is the high-energy donor that is added to target proteins by O-GlcNAc transferase (OGT) and is removed hydrolytically by O-GlcNAcase (25). Although O-GlcNAcylation occurs primarily on nuclear/cytoplasmic proteins (24), this modification can mediate critical signaling at the plasma membrane via the interaction of OGT with phosphatidylinositol trisphosphate (28). However, the modification of TRAIL death receptors by O-GlcNAcylation and its roles in tumorigenesis are unknown.

From a cell-based functional screen using a cancer cell–derived cDNA expression library, we unexpectedly isolated a DR4 mutant that suppressed TRAIL cytotoxicity in cancer cells. We characterized that O-GlcNAcylation of DR4-Ser424 is a crucial step to initiate DR4-mediated tumor-selective killing mechanisms and to determine TRAIL sensitivity in DR4-preferred cancer cells. Moreover, we propose prospective strategies for overcoming TRAIL resistance to facilitate the clinical application of TRAIL.

Cell culture

Human stomach cancer cells (SNU lines) were obtained from Cancer Research Institute of Seoul National Hospital, Seoul National University (Seoul, Korea; ref. 29), and all other cells were from the ATCC. All the cell lines, except SNU lines, were authenticated by short tandem repeat profiling and tested for Mycoplasma contamination using MycoAlert Mycoplasma Detection Kit (Lonza) more than once during this study. SNU lines were authenticated by their manufacturers. Cells were used within a maximum of 13 passages after thawing.

Reagents and antibodies

Reagents and specific antibodies used in this study are described in Supplementary Materials and Methods. IDN-6556 was kindly provided by Dr. C.-W. Chung (LG Biotech Inc., Korea).

Plasmids construction and transfection

The wtDR4 construct has been reported previously (30). All point mutations and SNPs of DR4 were generated by site-directed mutagenesis through PCR using specific primers containing the corresponding mutations. The sequences of primers are described in Supplementary Materials and Methods. Transfection was performed with iN-fect (iNtRON Biotechnology) or polyethylenimine (PEI; Sigma-Aldrich) following the manufacturer's instructions.

CRISPR/Cas9 gene editing for knockdown of OGT expression

For genomic engineering of OGT through CRISPR/Cas9 system, the single-guide RNA sequences (sgRNA oligos) targeting OGT (OGT sg; Supplementary Materials and Methods) were annealed and cloned into the BsmBI (Fermentas) sites of lentiCRISPR transfer plasmid (pXPR_001; Addgene) for virus production. Viral vectors were produced in HEK293T cells, and cells of interest were infected for OGT knockdown as described previously (31). After lentiviral delivery of both Cas9 and OGT sg, cells were selected with puromycin (Sigma-Aldrich). After single-cell cloning, the clones were screened by Western blotting.

Functional screen of a cancer cell line–derived cDNA expression library

Human full-length cDNA expression library was prepared by cloning full-length cDNA of mRNA sources from various cancer cell lines into pME18S-FL3 expression vector using a PCR-based oligo-capping method (Korea Research Institute of Bioscience & Biotechnology, KRIBB; https://genbank.kribb.re.kr). After cotransfection of a library with pEGFP-N1 into HeLa cells, cells were treated with TRAIL and the viability of GFP-positive cells was assessed as a readout under a fluorescence microscope (Olympus). Bcl-2 and c-FLIPL were used as positive controls to isolate negative regulators in TRAIL-induced cell death.

The Cancer Genome Atlas

The Cancer Genome Atlas (TCGA) consequence for S424L mutation of TNFRSF10A in uterine corpus endometrial carcinoma [TCGA-UCEC/TCGA-B5-A1MR] was accessed from Genomic Data Commons (GDC) Data Portal (https://portal.gdc.cancer.gov).

Cell death assays

Cell viability was examined by microscopic detection (32), FACS analysis, or Trypan blue exclusion assay. Briefly, nuclear chromatin was stained with 1 μg/mL Hoechst 33342 (Molecular Probes) and 1 μg/mL propidium iodide (PI; Sigma-Aldrich) for approximately 15 minutes. Apoptotic ratio was measured from cells with fragmented blue nuclei among PI-positive signals (n = 300–600 cells) under a fluorescent microscope. For the quantification of PI incorporation by FACS analysis, adherent and floating cells harvested by trypsinization and centrifugation, respectively, were resuspended in PI solution and analyzed by using a FACSCalibur (BD Biosciences). Necrotic cells were analyzed by flow cytometry after Annexin-V and 7AAD double-staining using FITC Annexin-V Detection Kit with 7AAD (640922, BioLegend) according to the manufacturer's instructions.

Detection of cell surface TRAIL receptors

For flow cytometry analysis, phycoerythrin (PE)-conjugated mouse anti-human DR4 (DJR1), anti-human DR5 (DJR2-4), and anti-IgG isotype control (MOPC-21) antibodies (BioLegend) were used according to the manufacturer's instructions. Briefly, cells were trypsinized and collected, washed with Cell Staining Buffer (420201, BioLegend), and incubated with PE-conjugated antibodies for 1 hour on ice. After being washed and resuspended with Cell Staining Buffer, cells were analyzed by flow cytometry using a FACSCalibur.

O-GlcNAcylation assays

For O-GlcNAc detection, cells were preincubated in a glucose-free medium for 10–30 minutes and then exposed to TRAIL in a glucose-supplemented medium. Cells were lysed with preheated 2.5% SDS-containing buffer: 10 mmol/L Tris-HCl (pH 8.0), 1 mmol/L EDTA, 2.5% SDS, and protease inhibitor cocktail (Quartett). After sonication (optional) and heat denaturing at 95°C, lysis buffer (50 mmol/L Tris-HCl pH 8.0, 150 mmol/L NaCl, 10% -glycerol, 1% Triton X-100, 1 mmol/L EDTA, 1 mmol/L EGTA, 10 μmol/L Thiamet-G, and protease inhibitor cocktail) was added for dilution of the final SDS concentration to under 1%. Samples were pipetted or incubated with rocking to relieve the viscous state. After centrifugation at 20,000 × g, O-GlcNAcylated proteins were immunopurified from the supernatant using an anti-DR4 antibody, an anti-Flag M2 affinity gel (Sigma-Aldrich), or GlcNAc-specific succinylated wheat germ (sWGA; Vector Laboratories).

Affinity purification of TRAIL-DISC and lipid rafts

Samples were prepared as described previously (33). Briefly, cells were preincubated with 500 ng/mL His-TRAIL (BML-SE721, Enzo Life Sciences) for 30 minutes at 4°C, and further incubated at 37°C for additional 30 minutes to 1 hour. For DISC purification, the resulting lysate was incubated with Ni-NTA agarose (QIAGEN) at 4°C for the enrichment of His-TRAIL–captured immunocomplexes. Lipid rafts were purified from the same cell lysates by discontinuous sucrose density gradient purification (33). For the isolation of TRAIL-DISC within lipid rafts, the resulting lipid raft fraction was further incubated with Ni-NTA agarose for ligand complex capture.

Statistical analysis

Statistical analysis was performed using Prism version 5.03 software (GraphPad Software). Data are expressed as mean ± SEM from at least three experiments. For statistical comparisons, data were analyzed by either one-way ANOVA, followed by Tukey post hoc test or two-way ANOVA, followed by Bonferroni post hoc test as appropriate, and P < 0.05 was considered statistically significant.

DR4-Ser424 mutants identified from a cancer-derived cDNA library and the database of patients with cancer lack cytotoxic activity

To isolate a genetic regulator of TRAIL-mediated cell death, we performed a gain-of-function screen using a cell death rescue assay. A total of 1,099 full-length cDNAs in a mammalian expression vector were prepared from patient-derived cancer cells and transfected into HeLa human cervical carcinoma cells, followed by TRAIL treatment (Fig. 1A, left). From the screen, four novel clones were identified to affect TRAIL cytotoxicity, and unexpectedly, DR4 was one of those clones (Supplementary Fig. S1A, TNFRSF10A). We found that this DR4 clone had four mutations (Fig. 1A and B, MUT), comprising three (L86R, H141R, and R209T) in the extracellular region and one (S424P) in the cytosolic death domain. Unlike wtDR4, DR4-MUT did not induce apoptotic morphologies, including cell shrinkage and membrane blebbing (Fig. 1A, graph). To identify which of these mutations caused the loss of cytotoxicity, we generated four revertants from DR4-MUT by individually substituting the mutations back into wild-type residues, so that each revertant harbored three mutations and one wild-type residue (Fig. 1B). Using reconstitution systems in DR4-null T98G cells, we first confirmed that all the revertants expressed equivalently on the cell surface as wtDR4 (Supplementary Fig. S1B). With ectopic expression assays in DLD-1 cells in which the TRAIL signaling is triggered dominantly through DR4, we found that only the DR4-MUTP424S revertant fully restored the cytotoxic activity equal to that of wtDR4 (Fig. 1C).

Figure 1.

Mutation at DR4-Ser424 identified from cancer cells loses its cytotoxicity. A, Representation of a gain-of-function screen strategy using a cell death rescue assay described in Materials and Methods. DR4-MUT here refers to the DR4 clone isolated from this screen. HeLa cells were cotransfected with pEGFP-N1 and empty vector (EV), wtDR4 (WT), or DR4-MUT (MUT) in the absence (NT) or presence of 10 μmol/L IDN-6556 (IDN). GFP-positive dying cells with shrinkage were assessed under a fluorescence microscope. B, Schematic representation depicting DR4 constructs. DR4-revertant possesses a single resubstituted WT residue as compared with DR4-MUT. Numbers indicate amino acid residues on DR4. DD, death domain; DMC, DR4-mediated cytotoxicity. C, DLD-1 cells coexpressing pEGFP-N1 with either empty vector or each of DR4 constructs were examined for the shrinked morphology with GFP signals under a fluorescence microscope (top). The expression levels of DR4 constructs were detected by immunoblotting (bottom). D, HeLa cells expressing empty vector or DR4-HA constructs were treated with 100 ng/mL TRAIL for 12 hours. Apoptosis was quantified (top) and the expression of DR4 constructs was detected by immunoblotting (bottom). E, DR4-null T98G and SNU-216 cells were reconstituted with wtDR4, DR4S424P, or DR4S424A and examined for apoptosis or analyzed by immunoblotting. F and G, HeLa/RIPK3-HA cells were transfected with empty vector, wtDR4, or various DR4-Ser424 mutants and then incubated with 500 ng/mL TRAIL for 16 hours (F) or 200 ng/mL TRAIL (T) for 4 hours (G) in the presence of 50 nmol/L SM-164 (S) and 10 μmol/L IDN-6556 (I). Cells were analyzed by flow cytometry using Annexin-V and 7AAD double-staining (F) or analyzed by immunoblotting (G). Numbers represent the rate of necrotic cell death by quantifying the signal density in top left quadrant (F). Arrowheads indicate upshifted and activated RIPK1 and RIPK3 (G). Bars, mean ± SEM (n = 3). The P values were calculated using either one-way ANOVA, followed by Tukey post hoc test (C; D, wtDR4 vs. DR4 mutants) or two-way ANOVA, followed by Bonferroni post hoc test (A). ***, P < 0.001.

Figure 1.

Mutation at DR4-Ser424 identified from cancer cells loses its cytotoxicity. A, Representation of a gain-of-function screen strategy using a cell death rescue assay described in Materials and Methods. DR4-MUT here refers to the DR4 clone isolated from this screen. HeLa cells were cotransfected with pEGFP-N1 and empty vector (EV), wtDR4 (WT), or DR4-MUT (MUT) in the absence (NT) or presence of 10 μmol/L IDN-6556 (IDN). GFP-positive dying cells with shrinkage were assessed under a fluorescence microscope. B, Schematic representation depicting DR4 constructs. DR4-revertant possesses a single resubstituted WT residue as compared with DR4-MUT. Numbers indicate amino acid residues on DR4. DD, death domain; DMC, DR4-mediated cytotoxicity. C, DLD-1 cells coexpressing pEGFP-N1 with either empty vector or each of DR4 constructs were examined for the shrinked morphology with GFP signals under a fluorescence microscope (top). The expression levels of DR4 constructs were detected by immunoblotting (bottom). D, HeLa cells expressing empty vector or DR4-HA constructs were treated with 100 ng/mL TRAIL for 12 hours. Apoptosis was quantified (top) and the expression of DR4 constructs was detected by immunoblotting (bottom). E, DR4-null T98G and SNU-216 cells were reconstituted with wtDR4, DR4S424P, or DR4S424A and examined for apoptosis or analyzed by immunoblotting. F and G, HeLa/RIPK3-HA cells were transfected with empty vector, wtDR4, or various DR4-Ser424 mutants and then incubated with 500 ng/mL TRAIL for 16 hours (F) or 200 ng/mL TRAIL (T) for 4 hours (G) in the presence of 50 nmol/L SM-164 (S) and 10 μmol/L IDN-6556 (I). Cells were analyzed by flow cytometry using Annexin-V and 7AAD double-staining (F) or analyzed by immunoblotting (G). Numbers represent the rate of necrotic cell death by quantifying the signal density in top left quadrant (F). Arrowheads indicate upshifted and activated RIPK1 and RIPK3 (G). Bars, mean ± SEM (n = 3). The P values were calculated using either one-way ANOVA, followed by Tukey post hoc test (C; D, wtDR4 vs. DR4 mutants) or two-way ANOVA, followed by Bonferroni post hoc test (A). ***, P < 0.001.

Close modal

Next, to examine whether DR4-Ser424 is essential for mediating TRAIL-induced cell death, we directly introduced a proline mutation at Ser424 of wtDR4 by substitution of the 1270th thymidine with cysteine. In addition to the DR4S424P mutant isolated in our screen, we also found another DR4-Ser424 mutation. From searching the database of patients with cancer in TCGA (TCGA-B5-A1MR-10A), we discovered a DR4S424L mutant in uterine corpus endometrial carcinoma, although the frequency was low (1 of 530 cases, 0.19%). Ectopic expression assays revealed that all these mutants showed loss of cytotoxicity after TRAIL stimulation and even suppressed TRAIL-induced apoptosis (Fig. 1D) despite of the equivalent membrane expressions to wtDR4 (Supplementary Fig. S1C). In addition, with reconstitution assays in DR4-null T98G human glioblastoma cells and SNU-216 human stomach adenocarcinoma cells, we confirmed a lack of caspase activation and PARP-1 cleavage by DR4-Ser424 mutants (Fig. 1E). Finally, because DR4 polymorphisms associated with cancer risk have been described in various human cancers (34), we examined the cytotoxicity of other DR4 polymorphisms (rs6557634, rs4871857, rs17088993, rs2230229) that have appeared at particularly high frequencies in cancers. Unlike DR41270T>C (DR4S424P), the other polymorphisms described above actively induced apoptosis in DLD-1 cells (Supplementary Fig. S1D), emphasizing the notion that mutations at DR4-Ser424 are responsible for the resistance of tumor cells to TRAIL-induced apoptosis.

Because TRAIL induces not only apoptosis but also programmed necrosis (15, 16), we decided to test whether DR4-Ser424 mutants could also affect necrosis. Owing to the requirement of RIPK3 for necroptosis, we employed two different cancer cell lines from analyses of apoptosis, SNU-16 human stomach adenocarcinoma cells expressing endogenous RIPK3 and HeLa cells expressing exogenous RIPK3-HA (HeLa/RIPK3-HA), and treated them with TRAIL in the presence of SMAC mimetic SM-164 and a pan-caspase inhibitor IDN-6556 (T/S/I in figures). Similar to the results for apoptosis, necroptosis was also suppressed by DR4-Ser424 mutants (Fig. 1F). In addition, the active forms of necroptosis mediators, such as phosphorylated RIPK1/RIPK3/MLKL, were not observed after the ectopic expression of DR4-Ser424 mutants (Fig. 1G). Thus, DR4-Ser424 is the key residue for triggering necroptosis as well as apoptosis in response to TRAIL.

DR4-Ser424 is O-GlcNAcylated to trigger TRAIL-mediated cell death

Serine generally undergoes posttranslational modifications, such as phosphorylation and O-GlcNAcylation (35). We thus hypothesized that DR4-Ser424 might undergo a posttranslational modification and monitored the phosphorylation and O-GlcNAcylation of DR4 upon TRAIL administration. Results from immunoprecipitation (IP) analysis using an anti-phosphoserine antibody showed that the overexpressed wtDR4 appeared to be phosphorylated (Fig. 2A, IP:p-Serine). However, the observation that DR4S424A also seemed to be phosphorylated as much as wtDR4 revealed that DR4 phosphorylation might occur at residues other than Ser424 (Fig. 2A). Because proline's unique cyclic structure commonly causes conformational changes in the secondary structure of proteins (36), the weak phosphorylation of DR4S424P may have been due to a structural perturbation (Fig. 2A). Meanwhile, we observed that wtDR4 was O-GlcNAcylated, but all DR4-Ser424 mutants, including DR4S424P, DR4S424A, and DR4S424L, were not (Fig. 2A, IP:HA). Using GlcNAc-specific agarose-conjugated sWGA to enrich O-GlcNAcylated proteins by affinity purification, we confirmed that DR4-Ser424 is the only site of O-GlcNAcylation and there was no additional Ser/Thr residue for O-GlcNAcylation (Fig. 2B) or the modulation of cytotoxicity (Supplementary Fig. S1E) within the peptides from 425th residue to the end of DR4. In contrast to DR4, we found that DR5 was not O-GlcNAcylated under the same conditions (Fig. 2C).

Figure 2.

DR4-Ser424 is modified by O-GlcNAc for TRAIL-induced cell death. A, HEK293T cells were transfected with empty vector (EV), wtDR4, or DR4-Ser424 mutants, and cell extracts were subjected to immunoprecipitation (IP) assay using antiphosphoserine antibody (p-Serine) or anti-HA antibody. Whole-cell lysates (Input) and the immunocomplex were analyzed by Western blotting. B, HeLa cells were transfected with wtDR4 or DR4 mutants for 24 hours and then subjected to affinity purification (AP) using sWGA as described in Materials and Methods. Thiamet-G (50 μmol/L) was treated for 6 hours prior to AP. C, HEK293T cells expressing DR4-Flag or DR5-Flag were treated with 100 ng/mL TRAIL for 1 hour and analyzed by affinity purification assay. D, DLD-1 cells were stimulated with either 100 ng/mL TRAIL for the indicated times (left) or the indicated doses of TRAIL for 1 hour (right). Cells were analyzed by O-GlcNAcylation assay. E, HeLa/RIPK3-HA control (OGT sg−) and HeLa/RIPK3-HA/OGT-knockdown (OGT sg+) cells were exposed to T/S/I for 1 hour and then analyzed by immunopurification. F and G, Immunoprecipitation assays to analyze DR4–OGT interactions. DLD-1 or SNU-16 cells were incubated with 50 ng/mL TRAIL or T/S/I for 1 hour, respectively. H, DLD-1 cells were pretreated with 20 μmol/L Thiamet-G, 20 μmol/L DON, or 5 μmol/L ST045849 for 30 minutes and stimulated with 200 ng/mL TRAIL for 24 hours. I, SK-Hep1/Control and SK-Hep1/OGT-knockdown (OGT sg #1, #2) cells were treated with 100 ng/mL TRAIL in the presence or absence of αDR5 for 12 hours. J, Cells were transfected with empty vector, wtDR4, DR4S424P, or wtDR5 and stimulated with 100 ng/mL TRAIL for 12 hours. Cells were subjected to cell death assay (top) and immunoblotting (bottom). Bars represent cell death rates (mean ± SEM, n = 3). The P values were calculated using one-way ANOVA, followed by Tukey test (H) or two-way ANOVA, followed by Bonferroni test (I and J). ns, not significant; **, P < 0.01; ***, P < 0.001.

Figure 2.

DR4-Ser424 is modified by O-GlcNAc for TRAIL-induced cell death. A, HEK293T cells were transfected with empty vector (EV), wtDR4, or DR4-Ser424 mutants, and cell extracts were subjected to immunoprecipitation (IP) assay using antiphosphoserine antibody (p-Serine) or anti-HA antibody. Whole-cell lysates (Input) and the immunocomplex were analyzed by Western blotting. B, HeLa cells were transfected with wtDR4 or DR4 mutants for 24 hours and then subjected to affinity purification (AP) using sWGA as described in Materials and Methods. Thiamet-G (50 μmol/L) was treated for 6 hours prior to AP. C, HEK293T cells expressing DR4-Flag or DR5-Flag were treated with 100 ng/mL TRAIL for 1 hour and analyzed by affinity purification assay. D, DLD-1 cells were stimulated with either 100 ng/mL TRAIL for the indicated times (left) or the indicated doses of TRAIL for 1 hour (right). Cells were analyzed by O-GlcNAcylation assay. E, HeLa/RIPK3-HA control (OGT sg−) and HeLa/RIPK3-HA/OGT-knockdown (OGT sg+) cells were exposed to T/S/I for 1 hour and then analyzed by immunopurification. F and G, Immunoprecipitation assays to analyze DR4–OGT interactions. DLD-1 or SNU-16 cells were incubated with 50 ng/mL TRAIL or T/S/I for 1 hour, respectively. H, DLD-1 cells were pretreated with 20 μmol/L Thiamet-G, 20 μmol/L DON, or 5 μmol/L ST045849 for 30 minutes and stimulated with 200 ng/mL TRAIL for 24 hours. I, SK-Hep1/Control and SK-Hep1/OGT-knockdown (OGT sg #1, #2) cells were treated with 100 ng/mL TRAIL in the presence or absence of αDR5 for 12 hours. J, Cells were transfected with empty vector, wtDR4, DR4S424P, or wtDR5 and stimulated with 100 ng/mL TRAIL for 12 hours. Cells were subjected to cell death assay (top) and immunoblotting (bottom). Bars represent cell death rates (mean ± SEM, n = 3). The P values were calculated using one-way ANOVA, followed by Tukey test (H) or two-way ANOVA, followed by Bonferroni test (I and J). ns, not significant; **, P < 0.01; ***, P < 0.001.

Close modal

We observed that a little proportion of endogenous DR4 proteins was modified by O-GlcNAc under basal conditions in untreated control cells, while the frequency of this modification markedly increased in response to treatment with either apoptotic TRAIL (Fig. 2D) or necrosis-inducing T/S/I (Fig. 2E). Accordingly, DR4 O-GlcNAcylation was abolished by the CRISPR/Cas9-mediated knockdown of OGT expression (Fig. 2E; Supplementary Fig. S2A). Furthermore, results from immunoprecipitation assays revealed that upon TRAIL stimulation, the binding of DR4 to OGT was enhanced in apoptotic DLD-1 and necrotic SNU-16 cells (Fig. 2F), or OGT bound to DR4 vice versa (Fig. 2G). Taken together, we concluded that DR4 was modified by O-GlcNAc upon TRAIL ligation in an OGT-dependent manner.

We further examined TRAIL-induced cell death under O-GlcNAcylation–deficient conditions. We found that TRAIL-induced apoptosis was accelerated in the presence of Thiamet-G, an inhibitor of O-GlcNAcase, but mitigated by ST045849, an OGT inhibitor, and by 6-diazo-5-oxo-l-norleucine (DON), a glutamine analogue that results in a decreased concentration of UDP-GlcNAc (Fig. 2H). In SK-Hep1 and HeLa cells, which transmit TRAIL signaling preferentially via DR5, blocking DR5 using a DR5-neutralizing antibody (αDR5) allowed only DR4-mediated TRAIL signaling. Under this condition, TRAIL/DR4-dependent cell death was undermined by OGT knockdown (Fig. 2I, TRAIL+αDR5). As expected, TRAIL-induced cytotoxicity was not influenced by OGT levels in the absence of DR5 blocking (Fig. 2I, TRAIL) because DR5 was not modified by O-GlcNAc (Fig. 2C). Consistently, the overexpression of wtDR4 evoked less apoptosis and caspase-8 activation in OGT-deficient SK-Hep1 cells than those of DR5, which was similar to the effect of O-GlcNAcylation-defective DR4S424P (Fig. 2J). Expressions and surface localization of DR4 and DR5 were not affected by OGT knockdown (Fig. 2J, Flag; Supplementary Fig. S2B). Upon TRAIL-necrotic stimulation combined with blocking DR5, the phosphorylation-dependent activation of RIPK1/RIPK3/MLKL was also suppressed in OGT-knockdown HeLa/RIPK3-HA cells (Supplementary Fig. S2C). These observations indicate again that O-GlcNAcylation specifically enhances DR4-mediated cell death.

O-GlcNAcylation facilitates DISC assembly and DR4 clustering within lipid rafts for the onset of TRAIL signaling

To examine the contribution of DR4 O-GlcNAcylation to the TRAIL-DISC formation, we used again Thiamet-G and ST045849 to potentiate and inhibit O-GlcNAcylation, respectively, and purified DISC. Because DR5 was not O-GlcNAcylated (Fig. 2C, I, and J), the amount of DR5 within the isolated DISC was not influenced by O-GlcNAcylation, while DR4 recruitment into the TRAIL-DISC was abolished by treatment of ST045849 (Fig. 3A). Levels of FADD and caspase-8 in the purified TRAIL-DISC were apparently increased after incubation with Thiamet-G, but were again diminished by treatment of ST045849 (Fig. 3A). Interestingly, OGT was also recruited into the purified DISC together with DR4. These results indicate that DR4 O-GlcNAcylation is required for DISC formation upon TRAIL stimulation.

Figure 3.

DR4-Ser424 O-GlcNAcylation facilitates DISC/necrosome assembly and clustering in lipid rafts. A and C, DLD-1 cells were pretreated with 50 μmol/L Thiamet-G or 10 μmol/L ST045849 for 1 hour, incubated with 500 ng/mL His-TRAIL for additional 30 minutes, lysed, and subjected to lipid raft fractionation with discontinuous sucrose density gradient purification (C). TRAIL-DISC was isolated using a His pull-down assay from total (A) or fractionated (C) lysates. DISC components were analyzed by immunoblotting. Arrowheads, active CASP8. B and D, HeLa cells were transfected with either empty vector (−), wtDR4-Flag (WT), or DR4S424P-Flag (SP) and stimulated with 500 ng/mL His-TRAIL for additional 1 hour. TRAIL-DISC and DR4-bound complex was isolated by a His pull-down or a Flag pull-down assay, respectively. Total DISC (B) and DISC within lipid rafts (D) were analyzed as in A and C. E and F, HeLa cells were transfected with either empty vector (EV), wtDR4-Flag (WT), DR4S424P-Flag (SP), DR4S424A-Flag (SA), or DR4S424L-Flag (SL) and stimulated with 100 ng/mL TRAIL for additional 2 hours. Non-raft and lipid raft fractions were purified and separated by SDS-PAGE under a reducing (E) or a nonreducing (F) condition for immunoblotting. G, Necrosome analysis. HeLa/RIPK3-HA cells transfected with empty vector, wtDR4, or DR4-Ser424 mutants were stimulated by TRAIL (100 ng/mL)/S/I for 4 hours. Cell extracts were subjected to immunoprecipitation (IP) using an anti-RIPK1 antibody. Arrowheads, activated RIPK1/RIPK3. H, HeLa/RIPK3-HA cells were transfected with wtDR4 (WT) or DR4S424P (SP) and then treated with T/S/I for 6 hours. Cells were separated by SDS-PAGE under nonreducing conditions for immunoblotting. White and black arrowheads correspond to protein monomers and oligomers, respectively. Data represent two or three independent experiments.

Figure 3.

DR4-Ser424 O-GlcNAcylation facilitates DISC/necrosome assembly and clustering in lipid rafts. A and C, DLD-1 cells were pretreated with 50 μmol/L Thiamet-G or 10 μmol/L ST045849 for 1 hour, incubated with 500 ng/mL His-TRAIL for additional 30 minutes, lysed, and subjected to lipid raft fractionation with discontinuous sucrose density gradient purification (C). TRAIL-DISC was isolated using a His pull-down assay from total (A) or fractionated (C) lysates. DISC components were analyzed by immunoblotting. Arrowheads, active CASP8. B and D, HeLa cells were transfected with either empty vector (−), wtDR4-Flag (WT), or DR4S424P-Flag (SP) and stimulated with 500 ng/mL His-TRAIL for additional 1 hour. TRAIL-DISC and DR4-bound complex was isolated by a His pull-down or a Flag pull-down assay, respectively. Total DISC (B) and DISC within lipid rafts (D) were analyzed as in A and C. E and F, HeLa cells were transfected with either empty vector (EV), wtDR4-Flag (WT), DR4S424P-Flag (SP), DR4S424A-Flag (SA), or DR4S424L-Flag (SL) and stimulated with 100 ng/mL TRAIL for additional 2 hours. Non-raft and lipid raft fractions were purified and separated by SDS-PAGE under a reducing (E) or a nonreducing (F) condition for immunoblotting. G, Necrosome analysis. HeLa/RIPK3-HA cells transfected with empty vector, wtDR4, or DR4-Ser424 mutants were stimulated by TRAIL (100 ng/mL)/S/I for 4 hours. Cell extracts were subjected to immunoprecipitation (IP) using an anti-RIPK1 antibody. Arrowheads, activated RIPK1/RIPK3. H, HeLa/RIPK3-HA cells were transfected with wtDR4 (WT) or DR4S424P (SP) and then treated with T/S/I for 6 hours. Cells were separated by SDS-PAGE under nonreducing conditions for immunoblotting. White and black arrowheads correspond to protein monomers and oligomers, respectively. Data represent two or three independent experiments.

Close modal

Confirmatively, compared with wtDR4, the O-GlcNAcylation–defective DR4S424P was little recruited into the DISC upon TRAIL stimulation and failed to form TRAIL-DISC in HeLa cells (Fig. 3B, His pull-down). DR4-Ser424 mutants also failed to bind to FADD (Supplementary Fig. S3A). While the similar level of DR5 was detected within TRAIL-DISC regardless of the endogenous DR4 O-GlcNAcylation (Fig. 3A), there was little DR5 within TRAIL-DISC in DR4S424P-expressing cells as compared with that of wtDR4-expressing cells (Fig. 3B, His pull-down). Because DR4S424P could also interact with DR5 upon TRAIL ligation (Supplementary Fig. S3B), we speculated that DR4S424P might capture DR5 like a decoy receptor and hinder DR5 from being properly recruited into the TRAIL-DISC. We tested this hypothesis by purifying and analyzing DR4-enriched complex. When the protein complexes in wtDR4-Flag- and DR4S424P-Flag–expressing cells were purified by a Flag pull-down, we found that OGT, FADD, and caspase-8 were not detected in the DR4S424P complex, whereas DR5 was contained in the DR4S424P complex as well as in the wtDR4 complex (Fig. 3B, Flag pull-down).

To further inquire whether O-GlcNAcylated DR4–containing DISC is indeed assembled within lipid rafts and whether DR4 O-GlcNAcylation is critical for DR4 translocation into and clustering within lipid rafts, we prepared lipid rafts using sucrose density gradient fractionation (33) and examined DISC formation, DR4 distribution, and receptor clustering. After TRAIL stimulation, DISC was formed within lipid rafts in DLD-1 cells (Fig. 3C) and wtDR4-expressing HeLa cells (Fig. 3D), but not in ST045849-treated cells (Fig. 3C) and DR4S424P-expressing cells (Fig. 3D). Accordingly, unlike DR4-Ser424 mutants, only wtDR4 could translocate into lipid rafts (Fig. 3E). Unlike wtDR4, DR4-Ser424 mutants also failed to self-aggregate in both non-raft and lipid raft fractions under nonreducing conditions (Fig. 3F). Together, DR4 O-GlcNAcylation is an essential prerequisite to initiate TRAIL signaling via facilitating DISC formation and DR4 clustering in lipid rafts. More, when we analyzed the impact of DR4 O-GlcNAcylation on necrosome formation, we found that, unlike those in wtDR4-expressing HeLa/RIPK3-HA cells, necrosome (Fig. 3G) and MLKL trimers (Fig. 3H; ref. 37) were not observed in DR4-Ser424 mutant–expressing cells upon TRAIL-necrosis stimulation.

Glucose level determines TRAIL sensitivity of cancer cells via DR4 O-GlcNAcylation

Given that O-GlcNAcylation is regulated according to the concentration of glucose (26), we assessed the effects of glucose on DR4 O-GlcNAcylation and TRAIL/DR4-dependent death. Compared with DLD-1 and HEK293T cells cultured in a glucose-rich medium, DR4 O-GlcNAcylation following TRAIL ligation was not observed in the same cell types cultured in a glucose-free medium (Fig. 4A–C). DR4 O-GlcNAcylation in HEK293T cells expressing wtDR4-Flag increased as the concentration of glucose was elevated and by treatment with Thiamet-G (Fig. 4B). Conversely, the modification was reduced by treatment with DON (Fig. 4B). Again, O-GlcNAcylation of DR4S424P was undetectable regardless of the presence or absence of glucose (Fig. 4C). Thus, glucose is a critical metabolic factor for DR4 O-GlcNAcylation as well.

Figure 4.

Glucose level contributes TRAIL susceptibility of cancer cells by modulating DR4 O-GlcNAcylation. A, DLD-1 cells were incubated in either a glucose-free (Glc−) or a complete (Glc+) medium for 1 hour, exposed to 100 ng/mL TRAIL for additional 1 hour, and analyzed by immunoprecipitation assay using anti-DR4 antibody. B, HEK293T cells expressing wtDR4-Flag were incubated with the increasing concentrations of glucose (0–25 mmol/L) in the presence of 50 μmol/L Thiamet-G or 100 μmol/L DON and subjected to affinity purification (AP) assay using anti-Flag M2 affinity gel. C, HEK293T cells transfected with empty vector (EV), wtDR4, or DR4S424P were cultured in a glucose-free or a complete medium in the presence of 100 ng/mL TRAIL, followed by affinity purification with sWGA. *, nonspecific signals. D, DLD-1 cells preincubated in a glucose-free or a complete medium for 30 minutes were left untreated or treated for 3 hours with 100 ng/mL TRAIL, 20 ng/mL TNFα plus 20 μmol/L cycloheximide (CHX), or 5 μmol/L A23187. Bars represent cell death rates with mean ± SEM (n = 3). E, DLD-1 cells preincubated in a glucose-free medium for 30 minutes were exposed to 500 ng/mL His-TRAIL for additional 1 hour in the presence or absence of glucose. TRAIL-DISC isolated by a His pull-down assay and input were analyzed by Western blotting. Arrowhead, active CASP8. F, DLD-1/control (Con-Glc, 25 mmol/L) and DLD-1/Low-Glc cells adapted to a low-glucose (5.5 mmol/L) condition were analyzed by flow cytometry after immunostaining using a phycoerythrin (PE)-conjugated isotype control antibody (gray) and a PE-conjugated anti-DR4 antibody (black). G and H, Cells were treated with 100 ng/mL TRAIL for 1 hour (G) or for the indicated times (H) and analyzed by O-GlcNAcylation assay (G) and Trypan blue exclusion assay (H). Values indicate cell death ratios with mean ± SEM (n = 3). The P values in D and H were calculated using two-way ANOVA, followed by Bonferroni test (ns, not significant; **, P < 0.01; ***, P < 0.001). Immunoblotting data represent three independent experiments of O-GlcNAcylation assays.

Figure 4.

Glucose level contributes TRAIL susceptibility of cancer cells by modulating DR4 O-GlcNAcylation. A, DLD-1 cells were incubated in either a glucose-free (Glc−) or a complete (Glc+) medium for 1 hour, exposed to 100 ng/mL TRAIL for additional 1 hour, and analyzed by immunoprecipitation assay using anti-DR4 antibody. B, HEK293T cells expressing wtDR4-Flag were incubated with the increasing concentrations of glucose (0–25 mmol/L) in the presence of 50 μmol/L Thiamet-G or 100 μmol/L DON and subjected to affinity purification (AP) assay using anti-Flag M2 affinity gel. C, HEK293T cells transfected with empty vector (EV), wtDR4, or DR4S424P were cultured in a glucose-free or a complete medium in the presence of 100 ng/mL TRAIL, followed by affinity purification with sWGA. *, nonspecific signals. D, DLD-1 cells preincubated in a glucose-free or a complete medium for 30 minutes were left untreated or treated for 3 hours with 100 ng/mL TRAIL, 20 ng/mL TNFα plus 20 μmol/L cycloheximide (CHX), or 5 μmol/L A23187. Bars represent cell death rates with mean ± SEM (n = 3). E, DLD-1 cells preincubated in a glucose-free medium for 30 minutes were exposed to 500 ng/mL His-TRAIL for additional 1 hour in the presence or absence of glucose. TRAIL-DISC isolated by a His pull-down assay and input were analyzed by Western blotting. Arrowhead, active CASP8. F, DLD-1/control (Con-Glc, 25 mmol/L) and DLD-1/Low-Glc cells adapted to a low-glucose (5.5 mmol/L) condition were analyzed by flow cytometry after immunostaining using a phycoerythrin (PE)-conjugated isotype control antibody (gray) and a PE-conjugated anti-DR4 antibody (black). G and H, Cells were treated with 100 ng/mL TRAIL for 1 hour (G) or for the indicated times (H) and analyzed by O-GlcNAcylation assay (G) and Trypan blue exclusion assay (H). Values indicate cell death ratios with mean ± SEM (n = 3). The P values in D and H were calculated using two-way ANOVA, followed by Bonferroni test (ns, not significant; **, P < 0.01; ***, P < 0.001). Immunoblotting data represent three independent experiments of O-GlcNAcylation assays.

Close modal

In general, nutrient deprivation, such as glucose starvation, is a stress that eventually triggers cell death. Thus, sustained exposure to a low concentration of glucose or the inhibition of glucose metabolism augments TRAIL cytotoxicity (38). Our experimental results from maintaining cells under glucose deprivation for a long duration coincided with those from the previous studies. When DLD-1 cells were exposed to TRAIL under a glucose-deprived condition for more than 12 hours, the cells became more vulnerable to TRAIL-induced cell death (Supplementary Fig. S4A). Unlike this long-lasting effect of glucose deprivation, however, when DLD-1 cells were incubated under glucose-free conditions for a short time, cell death triggered by TRAIL was markedly mitigated at such early time points (Fig. 4D). In contrast, there was no significant difference in the incidence of cell death triggered by other cytotoxic stimuli, such as TNFα plus cycloheximide, an extrinsic death signal, or a calcium ionophore A23187 that causes intrinsic death (Fig. 4D). Accordingly, subjecting cells to the short-term deprivation of glucose impaired the formation of DISC and subsequent caspase-8 activation under TRAIL stimulation (Fig. 4E; Supplementary Fig. S4B). To further evaluate the effect of glucose concentration on TRAIL efficacy in detail, we generated tumor cells adapted to a low-glucose medium. DLD-1 cells were cultured with a complete (25 mmol/L; DLD-1/Con-Glc) or a low-glucose (5.5 mmol/L; DLD-1/Low-Glc) medium for 1 month and tested for their TRAIL sensitivity. Although membrane expressions of DR4 in these cell lines were equivalent (Fig. 4F), DR4 O-GlcNAcylation triggered by TRAIL (Fig. 4G) and sensitivity to TRAIL (Fig. 4H) were significantly reduced in DLD-1/Low-Glc cells as compared with DLD-1/Con-Glc cells. This evidence suggests that cancer cells become less susceptible to TRAIL-mediated cell death under glucose-deficient conditions owing to the impairment of DR4 O-GlcNAcylation.

DR4 O-GlcNAcylation is dysregulated in TRAIL-resistant cancer cells, including cancer persister cells with acquired TRAIL tolerance

In cancer cells mainly using DR4 for the transduction of TRAIL signaling, TRAIL efficacy was highly associated with glucose concentrations and ultimately with DR4 O-GlcNAcylation. To inquire whether impaired O-GlcNAcylation could be a novel causative factor of TRAIL resistance in cancer cells, we sought to determine whether TRAIL sensitivity correlated with DR4 O-GlcNAcylation in various cancer cell lines. Total 15 cancer cell lines were assessed for their receptor expressions and sensitivity to TRAIL (Fig. 5A; Supplementary Fig. S5A–S5C). We classified them into TRAIL-sensitive (TRAIL-S) and -resistant (TRAIL-R; death rates ≤ 20%) groups and further classified the DR4-positive (DR4+)/TRAIL-sensitive cell lines into DR4-dominant (DR4*) or DR5-dominant (DR5*) groups according to their preferential receptor engagement. Receptor preference was classified by analyzing cell death after treatment with TRAIL and αDR5 (Fig. 5A; Supplementary Fig. S5B). Then, the status of DR4 O-GlcNAcylation in DR4-positive cancer cells was examined with O-GlcNAcylation assays using either an anti-DR4 antibody or sWGA. Surprisingly, DR4 O-GlcNAcylation was markedly promoted by TRAIL stimulation only in the TRAIL-sensitive cell lines, but not observed or not increased upon TRAIL treatment in all of the TRAIL-resistant cell lines we tested (Fig. 5B; Supplementary Fig. S5D). Thus, we suggest that the impairment of DR4 O-GlcNAcylation after TRAIL ligation would lead to TRAIL resistance in cancer cells.

Figure 5.

Impairment of DR4 O-GlcNAcylation leads to TRAIL resistance, including TRAIL-tolerant persistent state. A, Cell lines from stomach cancer (top left), lung cancer (top right), and glioblastoma (GBM; Supplementary Fig. S5B) were exposed to 200 ng/mL TRAIL for 24 hours in the presence or absence of αDR5 and analyzed by cell death assay. Bars represent the percentages of cell death with mean ± SEM (n = 3, two-way ANOVA, followed by Bonferroni test. ns, not significant; *, P < 0.05; ***, P < 0.001). Table shows the classification of cancer cells according to their receptor distribution (Supplementary Fig. S5A) and sensitivity to TRAIL. B, The correlation of TRAIL sensitivity with DR4 O-GlcNAcylation. DR4-positive stomach (left) and colon (right) cancer cells were incubated with 100 ng/mL TRAIL for 1 hour and then analyzed by O-GlcNAcylation assay. C, Schematic diagram showing TRAIL-tolerant (“persistent”) subpopulation from TRAIL-sensitive DLD-1 cells (DLD-1Naïve). Each cell population was established as described in Materials and Methods. D, DLD-1Naïve, DLD-1PER, and DLD-1PER-S cells were exposed to 100 ng/mL TRAIL for 12 hours and examined for apoptosis. Bars, mean ± SEM (n = 3, one-way ANOVA, followed by Tukey test; ***, P < 0.001). E, DLD-1Naïve and DLD-1PER cells were exposed to 100 ng/mL TRAIL for 1 hour, followed by O-GlcNAcylation assay. F, DLD-1Naïve, DLD-1PER, and DLD-1PER-S cells were exposed to 500 ng/mL His-TRAIL for 1 hour. TRAIL-DISC isolated by a His pull-down assay and input were analyzed by immunoblotting. Arrowhead, active CASP8. Western blotting data represent two or three independent experiments.

Figure 5.

Impairment of DR4 O-GlcNAcylation leads to TRAIL resistance, including TRAIL-tolerant persistent state. A, Cell lines from stomach cancer (top left), lung cancer (top right), and glioblastoma (GBM; Supplementary Fig. S5B) were exposed to 200 ng/mL TRAIL for 24 hours in the presence or absence of αDR5 and analyzed by cell death assay. Bars represent the percentages of cell death with mean ± SEM (n = 3, two-way ANOVA, followed by Bonferroni test. ns, not significant; *, P < 0.05; ***, P < 0.001). Table shows the classification of cancer cells according to their receptor distribution (Supplementary Fig. S5A) and sensitivity to TRAIL. B, The correlation of TRAIL sensitivity with DR4 O-GlcNAcylation. DR4-positive stomach (left) and colon (right) cancer cells were incubated with 100 ng/mL TRAIL for 1 hour and then analyzed by O-GlcNAcylation assay. C, Schematic diagram showing TRAIL-tolerant (“persistent”) subpopulation from TRAIL-sensitive DLD-1 cells (DLD-1Naïve). Each cell population was established as described in Materials and Methods. D, DLD-1Naïve, DLD-1PER, and DLD-1PER-S cells were exposed to 100 ng/mL TRAIL for 12 hours and examined for apoptosis. Bars, mean ± SEM (n = 3, one-way ANOVA, followed by Tukey test; ***, P < 0.001). E, DLD-1Naïve and DLD-1PER cells were exposed to 100 ng/mL TRAIL for 1 hour, followed by O-GlcNAcylation assay. F, DLD-1Naïve, DLD-1PER, and DLD-1PER-S cells were exposed to 500 ng/mL His-TRAIL for 1 hour. TRAIL-DISC isolated by a His pull-down assay and input were analyzed by immunoblotting. Arrowhead, active CASP8. Western blotting data represent two or three independent experiments.

Close modal

To verify a direct correlation and causality between DR4 O-GlcNAcylation and TRAIL susceptibility, we attempted to explore this association further focusing on the drug-tolerant “persistent state,” which is a recently proposed aspect of drug resistance (7–9). First, we obtained a small fraction of TRAIL-tolerant persisters (TTP) from TRAIL-sensitive DLD-1 cells (DLD-1Naïve) after exposure to TRAIL and further established expanded persistent populations (DLD-1PER) that grew in the continuous presence of TRAIL (Fig. 5C and D). We found that both DR4 O-GlcNAcylation and its interaction with OGT were abrogated in DLD-1PER cells showing acquired resistance to TRAIL, whereas the expression levels of OGT and death receptors were unchanged as compared with DLD-1Naïve cells (Fig. 5E; Supplementary Fig. S5E). Next, we abolished the TRAIL-tolerant state of DLD-1PER cells by propagating the cells in the absence of TRAIL (DLD-1PER-S) to resensitize them to TRAIL (Fig. 5C and D). Interestingly, we found that DR4 O-GlcNAcylation was restored in the DLD-1PER-S cells (Supplementary Fig. S5F). Consistently, TRAIL-DISC was not assembled in DLD-1PER cells, but was restored in DLD-1PER-S cells (Fig. 5F), suggesting that the deficiency of DR4 O-GlcNAcylation also renders TRAIL-sensitive cancer cells persistent to TRAIL.

TRAIL resistance is overcome by augmenting DR4 O-GlcNAcylation

Considering the significant influence of cellular glucose concentration on O-GlcNAcylation and the subsequent cytotoxicity of DR4, we hypothesized that increasing the glucose concentration could render TRAIL-resistant cancer cells susceptible to TRAIL through enhancing DR4 O-GlcNAcylation. To test this hypothesis, cancer cells were exposed to TRAIL together with a high concentration of glucose (high-Glc, 100 mmol/L; TRAIL+high-Glc) or a glucose analogue 2-deoxy-d-glucose (2-DG; TRAIL+2-DG). It is well-established that 2-DG increases TRAIL sensitivity in clinical studies (39) and also leads to the hyper-O-GlcNAcylation of certain proteins despite its inhibitory effect on glycolysis (40). Notably, TRAIL-resistant SNU-638 and SNU-719 human stomach adenocarcinoma cells became significantly vulnerable to TRAIL upon incubation with high-Glc or 2-DG (Fig. 6A and B; Supplementary Fig. S6A).

Figure 6.

DR4 O-GlcNAcylation in TRAIL-resistant cancer cells is restored by a high concentration of glucose or 2-DG application. A and B, SNU-638 cells were pretreated with αDR5, 20 μmol/L Thiamet-G, 2.5 μmol/L ST045849, or 20 μmol/L DON in the presence or absence of 5 mmol/L 4PBA for 1 hour and then stimulated by 200 ng/mL TRAIL and either 10 mmol/L 2-DG (A) or 100 mmol/L high-Glc (B) for 48 hours. Cell death was quantified by FACS analysis. C and D, SNU-638 cells were pretreated with 50 μmol/L Thiamet-G or 10 μmol/L ST045849 for 1 hour and stimulated by 100 ng/mL TRAIL and either 100 mmol/L high-Glc (C) or 10 mmol/L 2-DG (D) for 16 hours, and then subjected to O-GlcNAcylation assay. E, DLD-1PER cells were treated with 100 ng/mL TRAIL for 12 hours in the presence or absence of 100 mmol/L glucose or 10 mmol/L 2-DG and examined for cell death. F, The same cells in E were analyzed for DR4 O-GlcNAcylation by immunoprecipitation (IP) assay. Bars represent the percentages of cell death (mean ± SEM, n = 3). The P values were calculated using either one-way ANOVA, followed by Tukey test (A) or two-way ANOVA, followed by Bonferroni test (B and E). ns, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001. Immunoblotting data represent two or three independent experiments.

Figure 6.

DR4 O-GlcNAcylation in TRAIL-resistant cancer cells is restored by a high concentration of glucose or 2-DG application. A and B, SNU-638 cells were pretreated with αDR5, 20 μmol/L Thiamet-G, 2.5 μmol/L ST045849, or 20 μmol/L DON in the presence or absence of 5 mmol/L 4PBA for 1 hour and then stimulated by 200 ng/mL TRAIL and either 10 mmol/L 2-DG (A) or 100 mmol/L high-Glc (B) for 48 hours. Cell death was quantified by FACS analysis. C and D, SNU-638 cells were pretreated with 50 μmol/L Thiamet-G or 10 μmol/L ST045849 for 1 hour and stimulated by 100 ng/mL TRAIL and either 100 mmol/L high-Glc (C) or 10 mmol/L 2-DG (D) for 16 hours, and then subjected to O-GlcNAcylation assay. E, DLD-1PER cells were treated with 100 ng/mL TRAIL for 12 hours in the presence or absence of 100 mmol/L glucose or 10 mmol/L 2-DG and examined for cell death. F, The same cells in E were analyzed for DR4 O-GlcNAcylation by immunoprecipitation (IP) assay. Bars represent the percentages of cell death (mean ± SEM, n = 3). The P values were calculated using either one-way ANOVA, followed by Tukey test (A) or two-way ANOVA, followed by Bonferroni test (B and E). ns, not significant; *, P < 0.05; **, P < 0.01; ***, P < 0.001. Immunoblotting data represent two or three independent experiments.

Close modal

Because upregulation and self-aggregation of death receptors by 2-DG and/or ER stress have been reported (38, 39, 41, 42), we examined receptor levels after incubating SNU-638 cells with TRAIL+2-DG or TRAIL+high-Glc. When SNU-638 cells were stressed by treatment with TRAIL+2-DG, DR4 with a lower molecular weight was detected and DR5 was upregulated by 2-fold (Supplementary Fig. S6B and S6C). On the other hand, induction of DR5 expression and ER stress was not observed by treatment with TRAIL+high-Glc (Supplementary Fig. S6B, right). We found that 4PBA, an ER stress inhibitor, potently suppressed the increase of DR5 triggered by TRAIL+2-DG (Supplementary Fig. S6D). We then analyzed the effects of DR4-O-GlcNAcylation on TRAIL sensitivity in SNU-638 cells using both αDR5 and 4PBA. We found that TRAIL+2-DG-induced cell death via DR4 was potentiated by Thiamet-G but was alleviated by ST045849 in SNU-638 cells (Fig. 6A, white bars). The same results were observed when 2-DG–induced ER stress was inhibited by 4PBA (Fig. 6A, dark gray bars) and when SNU-638 cells were incubated in a high-Glc–containing culture medium (Fig. 6B), indicating that the sensitization of TRAIL-resistant SNU-638 cells to TRAIL by treatment with 2-DG and high-Glc is indeed due to DR4 O-GlcNAcylation.

Confirmatively, we found that DR4 O-GlcNAcylation by the treatment with TRAIL+2-DG or TRAIL+high-Glc was promoted in the presence of Thiamet-G, while this modification was not observed in the presence of ST045849 (Fig. 6C and D). Moreover, nongenetically TRAIL-resistant DLD-1PER cells were also resensitized to TRAIL in the presence of high-Glc or 2-DG (Fig. 6E), and DR4 O-GlcNAcylation in these DLD-1PER cells was recovered as well (Fig. 6F). Thus, enhancing DR4 O-GlcNAcylation improves TRAIL sensitivity in TRAIL-resistant cancer cells. Taken together, we propose again that DR4-Ser424 is the essential residue for the cytotoxic functioning of DR4 via O-GlcNAcylation in TRAIL signaling, providing clues for increasing the responsiveness to TRAIL therapy in patients with cancer.

Recent studies have highlighted the importance of sugar modifications, such as DR4 N-glycosylation (18) and DR5 O-glycosylation (19). Most of the reported modifications of TRAIL receptors, however, have focused only on the apoptotic elimination of cancer cells. In this study, we demonstrate that DR4 O-GlcNAcylation is important for the induction of not only apoptosis but also necrosis by TRAIL. Although necrosis is highly associated with inflammation and accelerates the pathogenesis of human diseases, including inflammatory diseases (43) and tumorigenesis (44), programmed necrosis works as a second barrier against tumor cells that avoid apoptosis (16). Notably, necrosis is the crucial step for the killing and elimination of stem-like cells (45) or cancer stem cells (46). Tumors that can evade both apoptotic and necrotic defenses tend to be aggressive and metastatic. Because O-GlcNAcylation is responsible for DR4-mediated necrosis as well as apoptosis, tumor cells harboring mutations at DR4-Ser424 have a high risk of becoming more tolerant against TRAIL therapy.

Tumor cells frequently display altered glucose metabolism, which is one of the hallmarks of cancer (47). O-GlcNAcylation is highly dependent on glucose and this modification generally becomes more abundant in various metabolic pathways in cancer cells (48). It appears that DR4 O-GlcNAcylation is a nutrient-dependent checkpoint that licenses DR4 to transmit death signals upon TRAIL stimulation. A low basal level of DR4 O-GlcNAcylation was observed in our experiments, but this may not reach the threshold to trigger cell death in nontransformed or unstimulated cells. In tumor cells, however, glucose uptake and HBP flux become greatly increased so that upregulated DR4 O-GlcNAcylation, as an ignition switch, may help TRAIL to selectively kill cancer cells. In addition, a sustained scarcity of glucose can create a favorable environment for malignant tumor cells (49) and consistently, an absence of DR4 O-GlcNAcylation under conditions of glucose shortage renders tumor cells more resistant to TRAIL. In short, the O-GlcNAcylation status of DR4 may determine the sensitivity of DR4-preferred cancer cells to TRAIL.

We also found that TRAIL efficacy was enhanced in TRAIL-resistant cancer cells, even in TRAIL-tolerant cancer persister cells, when we experimentally forced the O-GlcNAcylation of DR4 (Figs. 6 and 7). Our study addresses the mechanism of combined TRAIL chemotherapies in that treatment with a high concentration of glucose or 2-DG together with TRAIL encourages DR4 O-GlcNAcylation in TRAIL-resistant cancer cells, which overcomes their tolerance and restores the effectiveness of TRAIL. That is, we have demonstrated the importance of DR4 participation in the synergistic induction of cell death by TRAIL in combination with 2-DG, which is well-established to augment TRAIL sensitivity in TRAIL-resistant cancers (39). Therefore, we propose that manipulating the intracellular glucose level can provide a new strategy for chemotherapy against TRAIL-resistant cancer cells, although the safety of altering the glucose concentration must be carefully considered for clinical application in practice. In addition, the fast-migrating band on DR4 blot that we observed after 2-DG treatment (Fig. 6D; Supplementary Fig. S6B) remains to be characterized. It might be associated with the effects of 2-DG on ER stress and N-glycosylation (50) because it was similar with the size of unmodified DR4 deficient in N-glycosylation (18).

Figure 7.

Graphical summary depicting a significance of DR4 O-GlcNAcylation in TRAIL signaling. DR4-Ser424 is modified by O-GlcNAc in response to TRAIL ligation. O-GlcNAcylated DR4 is encouraged to translocate into compartmentalized lipid rafts at the initial step of TRAIL signaling. Subsequent clustering of DR4 in this active platform amplifies signaling cascade to induce apoptosis or necrosis via the formation of DISC or necrosome, respectively, in cancer cells. In contrast, in cancer cells tolerant to TRAIL, TRAIL fails to induce DR4 O-GlcNAcylation, resulting from DR4-Ser424 mutation, glucose shortage, or other unknown mechanisms. By utilizing this pathway of DR4 O-GlcNAcylation, however, TRAIL resistance of cancer cells can be overcome for TRAIL therapy. With the support of increased glucose or 2-DG application, the level of UDP-GlcNAc is upregulated to enhance OGT-dependent DR4 O-GlcNAcylation, enabling to ablate TRAIL-resistant cancer cells by TRAIL-mediated apoptosis or necrosis. A white “×” denotes the mutation of DR4-Ser424, such as DR4S424P and DR4S424L. G, O-GlcNAc.

Figure 7.

Graphical summary depicting a significance of DR4 O-GlcNAcylation in TRAIL signaling. DR4-Ser424 is modified by O-GlcNAc in response to TRAIL ligation. O-GlcNAcylated DR4 is encouraged to translocate into compartmentalized lipid rafts at the initial step of TRAIL signaling. Subsequent clustering of DR4 in this active platform amplifies signaling cascade to induce apoptosis or necrosis via the formation of DISC or necrosome, respectively, in cancer cells. In contrast, in cancer cells tolerant to TRAIL, TRAIL fails to induce DR4 O-GlcNAcylation, resulting from DR4-Ser424 mutation, glucose shortage, or other unknown mechanisms. By utilizing this pathway of DR4 O-GlcNAcylation, however, TRAIL resistance of cancer cells can be overcome for TRAIL therapy. With the support of increased glucose or 2-DG application, the level of UDP-GlcNAc is upregulated to enhance OGT-dependent DR4 O-GlcNAcylation, enabling to ablate TRAIL-resistant cancer cells by TRAIL-mediated apoptosis or necrosis. A white “×” denotes the mutation of DR4-Ser424, such as DR4S424P and DR4S424L. G, O-GlcNAc.

Close modal

Compared with the loss-of-function screen using siRNA library, our screen was effective to identify regulators displaying a gain-of-function in a signaling pathway. We discovered DR4-Ser424 mutations as a potent suppressor of TRAIL-induced death and characterized that DR4-Ser424 is O-GlcNAcylated to initiate TRAIL signaling. DR5 was not modified by O-GlcNAc, enabling us to discriminate a novel difference between DR4 and DR5. Despite the low frequency in TCGA data, it is interesting to note the power of DR4-Ser424 mutations because the impairment of DR4 O-GlcNAcylation even has an adverse effect not only on DR4 signaling but also on DR5-mediated cytotoxicity (Supplementary Fig. S7A and S7B). Given that death receptors can form a hetero-trimer upon TRAIL ligation (Supplementary Fig. S3B; ref. 23), the recruitment of the DR4-Ser424 mutant to DR5 can undermine DR5-mediated TRAIL cytotoxic signaling in a dominant-negative manner, which might contribute to tumorigenesis by enabling cancer cells to escape cell death. Thus, we propose that the DR4-Ser424 residue deserves careful consideration as a potential diagnostic marker for the genetic test of patients with cancer. However, we have to consider that DR4 O-GlcNAcylation is not totally responsible for TRAIL death signaling because the impairment of this modification by OGT knockdown and inhibition suppressed TRAIL death considerably but not entirely to the control level. Although further exploration is needed to determine whether DR4 O-GlcNAcylation has other roles in the immune system or NF-κB signaling and how TRAIL enhances DR4 O-GlcNAcylation specifically, our findings may be beneficial for the design of specific TRAIL treatments, bringing us closer to personalized medicine.

No potential conflicts of interest were disclosed.

Conception and design: H. Lee, Y.-K. Jung

Development of methodology: H. Lee, Y. Oh, Y.-J. Jeon, H. Kim, Y.-K. Jung

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Lee, Y. Oh, Y.-K. Jung

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): H. Lee, Y. Oh, Y.-J. Jeon, Y.-K. Jung

Writing, review, and/or revision of the manuscript: H. Lee, H.-J. Lee, Y.-K. Jung

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): H. Lee, Y.-J. Jeon, S.-Y. Lee, H. Kim, Y.-K. Jung Study supervision: Y.-K. Jung

This work was supported by a Global Research Laboratory grant (NRF-2010-00341 to Y.-K. Jung) and by the CRI grant (NRF-2016R1A2A1A05005304 to Y.-K. Jung) funded by the Ministry of Education, Science, and Technology. We thank Prof. J.-W. Cho (Yonsei University, Korea) for his kind discussion and for providing the human OGT plasmid and an anti-O-GlcNAc antibody that were used for our preliminary experiments.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Wiley
SR
,
Schooley
K
,
Smolak
PJ
,
Din
WS
,
Huang
CP
,
Nicholl
JK
, et al
Identification and characterization of a new member of the TNF family that induces apoptosis
.
Immunity
1995
;
3
:
673
82
.
2.
Walczak
H
,
Miller
RE
,
Ariail
K
,
Gliniak
B
,
Griffith
TS
,
Kubin
M
, et al
Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo
.
Nat Med
1999
;
5
:
157
63
.
3.
Ashkenazi
A
. 
Directing cancer cells to self-destruct with pro-apoptotic receptor agonists
.
Nat Rev Drug Discov
2008
;
7
:
1001
12
.
4.
Stuckey
DW
,
Shah
K
. 
TRAIL on trial: preclinical advances in cancer therapy
.
Trends Mol Med
2013
;
19
:
685
94
.
5.
von Karstedt
S
,
Conti
A
,
Nobis
M
,
Montinaro
A
,
Hartwig
T
,
Lemke
J
, et al
Cancer cell-autonomous TRAIL-R signaling promotes KRAS-driven cancer progression, invasion, and metastasis
.
Cancer Cell
2015
;
27
:
561
73
.
6.
LeBlanc
H
,
Lawrence
D
,
Varfolomeev
E
,
Totpal
K
,
Morlan
J
,
Schow
P
, et al
Tumor-cell resistance to death receptor–induced apoptosis through mutational inactivation of the proapoptotic Bcl-2 homolog Bax
.
Nat Med
2002
;
8
:
274
81
.
7.
Sharma
SV
,
Lee
DY
,
Li
B
,
Quinlan
MP
,
Takahashi
F
,
Maheswaran
S
, et al
A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations
.
Cell
2010
;
141
:
69
80
.
8.
Ramirez
M
,
Rajaram
S
,
Steininger
RJ
,
Osipchuk
D
,
Roth
MA
,
Morinishi
LS
, et al
Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells
.
Nat Commun
2016
;
7
:
10690
.
9.
Terai
H
,
Kitajima
S
,
Potter
DS
,
Matsui
Y
,
Quiceno
LG
,
Chen
T
, et al
ER stress signaling promotes the survival of cancer “persister cells” tolerant to EGFR tyrosine kinase inhibitors
.
Cancer Res
2018
;
78
:
1044
57
.
10.
Lemke
J
,
von Karstedt
S
,
Zinngrebe
J
,
Walczak
H
. 
Getting TRAIL back on track for cancer therapy
.
Cell Death Differ
2014
;
21
:
1350
64
.
11.
de Miguel
D
,
Lemke
J
,
Anel
A
,
Walczak
H
,
Martinez-Lostao
L
. 
Onto better TRAILs for cancer treatment
.
Cell Death Differ
2016
;
23
:
733
47
.
12.
Kayagaki
N
,
Yamaguchi
N
,
Nakayama
M
,
Takeda
K
,
Akiba
H
,
Tsutsui
H
, et al
Expression and function of TNF-related apoptosis-inducing ligand on murine activated NK cells
.
J Immunol
1999
;
163
:
1906
13
.
13.
MacFarlane
M
,
Ahmad
M
,
Srinivasula
SM
,
Fernandes-Alnemri
T
,
Cohen
GM
,
Alnemri
ES
. 
Identification and molecular cloning of two novel receptors for the cytotoxic ligand TRAIL
.
J Biol Chem
1997
;
272
:
25417
20
.
14.
Chaudhary
PM
,
Eby
M
,
Jasmin
A
,
Bookwalter
A
,
Murray
J
,
Hood
L
. 
Death receptor 5, a new member of the TNFR family, and DR4 induce FADD-dependent apoptosis and activate the NF-κB pathway
.
Immunity
1997
;
7
:
821
30
.
15.
Meurette
O
,
Rebillard
A
,
Huc
L
,
Le Moigne
G
,
Merino
D
,
Micheau
O
, et al
TRAIL induces receptor-interacting protein 1-dependent and caspase-dependent necrosis-like cell death under acidic extracellular conditions
.
Cancer Res
2007
;
67
:
218
26
.
16.
Voigt
S
,
Philipp
S
,
Davarnia
P
,
Winoto-Morbach
S
,
Röder
C
,
Arenz
C
, et al
TRAIL-induced programmed necrosis as a novel approach to eliminate tumor cells
.
BMC Cancer
2014
;
14
:
74
.
17.
Lafont
E
,
Kantari-Mimoun
C
,
Draber
P
,
De Miguel
D
,
Hartwig
T
,
Reichert
M
, et al
The linear ubiquitin chain assembly complex regulates TRAIL-induced gene activation and cell death
.
EMBO J
2017
;
36
:
1147
66
.
18.
Dufour
F
,
Rattier
T
,
Shirley
S
,
Picarda
G
,
Constantinescu
AA
,
Morlé
A
, et al
N-glycosylation of mouse TRAIL-R and human TRAIL-R1 enhances TRAIL-induced death
.
Cell Death Differ
2017
;
24
:
500
10
.
19.
Wagner
KW
,
Punnoose
EA
,
Januario
T
,
Lawrence
DA
,
Pitti
RM
,
Lancaster
K
, et al
Death-receptor O-glycosylation controls tumor-cell sensitivity to the proapoptotic ligand Apo2L/TRAIL
.
Nat Med
2007
;
13
:
1070
7
.
20.
Song
JH
,
Tse
MC
,
Bellail
A
,
Phuphanich
S
,
Khuri
F
,
Kneteman
NM
, et al
Lipid rafts and nonrafts mediate tumor necrosis factor related apoptosis-inducing ligand induced apoptotic and nonapoptotic signals in non small cell lung carcinoma cells
.
Cancer Res
2007
;
67
:
6946
55
.
21.
van Roosmalen
IA
,
Quax
WJ
,
Kruyt
FA
. 
Two death-inducing human TRAIL receptors to target in cancer: similar or distinct regulation and function?
Biochem Pharmacol
2014
;
91
:
447
56
.
22.
Bremer
E
,
de Bruyn
M
,
Samplonius
DF
,
Bijma
T
,
ten Cate
B
,
de Leij
LF
, et al
Targeted delivery of a designed sTRAIL mutant results in superior apoptotic activity towards EGFR-positive tumor cells
.
J Mol Med
2008
;
86
:
909
24
.
23.
Duiker
EW
,
de Vries
EG
,
Mahalingam
D
,
Meersma
GJ
,
Boersma-van Ek
W
,
Hollema
H
, et al
Enhanced antitumor efficacy of a DR5-specific TRAIL variant over recombinant human TRAIL in a bioluminescent ovarian cancer xenograft model
.
Clin Cancer Res
2009
;
15
:
2048
57
.
24.
Wells
L
,
Vosseller
K
,
Hart
GW
. 
Glycosylation of nucleocytoplasmic proteins: signal transduction and O-GlcNAc
.
Science
2001
;
291
:
2376
8
.
25.
Ruan
HB
,
Singh
JP
,
Li
MD
,
Wu
J
,
Yang
X
. 
Cracking the O-GlcNAc code in metabolism
.
Trends Endocrinol Metab
2013
;
24
:
301
9
.
26.
Guo
B
,
Liang
Q
,
Li
L
,
Hu
Z
,
Wu
F
,
Zhang
P
, et al
O-GlcNAc-modification of SNAP-29 regulates autophagosome maturation
.
Nat Cell Biol
2014
;
16
:
1215
26
.
27.
Wang
AC
,
Jensen
EH
,
Rexach
JE
,
Vinters
HV
,
Hsieh-Wilson
LC
. 
Loss of O-GlcNAc glycosylation in forebrain excitatory neurons induces neurodegeneration
.
Proc Natl Acad Sci U S A
2016
;
113
:
15120
5
.
28.
Yang
X
,
Ongusaha
PP
,
Miles
PD
,
Havstad
JC
,
Zhang
F
,
So
WV
, et al
Phosphoinositide signalling links O-GlcNAc transferase to insulin resistance
.
Nature
2008
;
451
:
964
9
.
29.
Park
JG
,
Frucht
H
,
LaRocca
RV
,
Bliss
DP
 Jr
,
Kurita
Y
,
Chen
TR
, et al
Characteristics of cell lines established from human gastric carcinoma
.
Cancer Res
1990
;
50
:
2773
80
.
30.
Oh
Y
,
Jeon
YJ
,
Hong
GS
,
Kim
I
,
Woo
HN
,
Jung
YK
. 
Regulation in the targeting of TRAIL receptor 1 to cell surface via GODZ for TRAIL sensitivity in tumor cells
.
Cell Death Differ
2012
;
19
:
1196
207
.
31.
Shalem
O
,
Sanjana
NE
,
Hartenian
E
,
Shi
X
,
Scott
DA
,
Mikkelson
T
, et al
Genome-scale CRISPR-Cas9 knockout screening in human cells
.
Science
2014
;
343
:
84
7
.
32.
Jouan-Lanhouet
S
,
Arshad
MI
,
Piquet-Pellorce
C
,
Martin-Chouly
C
,
Le Moigne-Muller
G
,
Van Herreweghe
F
, et al
TRAIL induces necroptosis involving RIPK1/RIPK3-dependent PARP-1 activation
.
Cell Death Differ
2012
;
19
:
2003
14
.
33.
Dickens
LS
,
Boyd
RS
,
Jukes-Jones
R
,
Hughes
MA
,
Robinson
GL
,
Fairall
L
, et al
A death effector domain chain DISC model reveals a crucial role for caspase-8 chain assembly in mediating apoptotic cell death
.
Mol Cell
2012
;
47
:
291
305
.
34.
Rai
R
,
Sharma
KL
,
Sharma
S
,
Misra
S
,
Kumar
A
,
Mittal
B
. 
Death receptor (DR4) haplotypes are associated with increased susceptibility of gallbladder carcinoma in north Indian population
.
PLoS One
2014
;
9
:
e90264
.
35.
Hart
GW
,
Slawson
C
,
Ramirez-Correa
G
,
Lagerlof
O
. 
Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease
.
Annu Rev Biochem
2011
;
80
:
825
58
.
36.
Buevich
AV
,
Dai
QH
,
Liu
X
,
Brodsky
B
,
Baum
J
. 
Site-specific NMR monitoring of cis-trans isomerization in the folding of the proline-rich collagen triple helix
.
Biochemistry
2000
;
39
:
4299
308
.
37.
Cai
Z
,
Jitkaew
S
,
Zhao
J
,
Chiang
HC
,
Choksi
S
,
Liu
J
, et al
Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis
.
Nat Cell Biol
2014
;
16
:
55
65
.
38.
Iurlaro
R
,
Püschel
F
,
León-Annicchiarico
CL
,
O'Connor
H
,
Martin
SJ
,
Palou-Gramón
D
, et al
Glucose deprivation induces ATF4-mediated apoptosis through TRAIL death receptors
.
Mol Cell Biol
2017
;
37
:
e00479
16
.
39.
Liu
H
,
Jiang
CC
,
Lavis
CJ
,
Croft
A
,
Dong
L
,
Tseng
HY
, et al
2-Deoxy-D-glucose enhances TRAIL-induced apoptosis in human melanoma cells through XBP-1-mediated up-regulation of TRAIL-R2
.
Mol Cancer
2009
;
8
:
122
.
40.
Kang
HT
,
Ju
JW
,
Cho
JW
,
Hwang
ES
. 
Down-regulation of Sp1 activity through modulation of O-glycosylation by treatment with a low glucose mimetic, 2-deoxyglucose
.
J Biol Chem
2003
;
278
:
51223
31
.
41.
Lam
M
,
Lawrence
DA
,
Ashkenazi
A
,
Walter
P
. 
Confirming a critical role for death receptor 5 and caspase-8 in apoptosis induction by endoplasmic reticulum stress
.
Cell Death Differ
2018
;
25
:
1530
1
.
42.
Muñoz-Pinedo
C
,
López-Rivas
A
. 
A role for caspase-8 and TRAIL-R2/DR5 in ER-stress-induced apoptosis
.
Cell Death Differ
2018
;
25
:
226
.
43.
Brost
S
,
Koschny
R
,
Sykora
J
,
Stremmel
W
,
Lasitschka
F
,
Walczak
H
, et al
Differential expression of the TRAIL/TRAIL-receptor system in patients with inflammatory bowel disease
.
Pathol Res Pract
2010
;
206
:
43
50
.
44.
Degenhardt
K
,
Mathew
R
,
Beaudoin
B
,
Bray
K
,
Anderson
D
,
Chen
G
, et al
Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis
.
Cancer Cell
2006
;
10
:
51
64
.
45.
Carlisi
D
,
Buttitta
G
,
Di Fiore
R
,
Scerri
C
,
Drago-Ferrante
R
,
Vento
R
, et al
Parthenolide and DMAPT exert cytotoxic effects on breast cancer stem-like cells by inducing oxidative stress, mitochondrial dysfunction and necrosis
.
Cell Death Dis
2016
;
7
:
e2194
.
46.
Singh
SR
,
Zeng
X
,
Zhao
J
,
Liu
Y
,
Hou
G
,
Liu
H
, et al
The lipolysis pathway sustains normal and transformed stem cells in adult Drosophila
.
Nature
2016
;
538
:
109
13
.
47.
Hanahan
D
,
Weinberg
RA
. 
Hallmarks of cancer: the next generation
.
Cell
2011
;
144
:
646
74
.
48.
Yang
X
,
Qian
K
. 
Protein O-GlcNAcylation: emerging mechanisms and functions
.
Nat Rev Mol Cell Biol
2017
;
18
:
452
65
.
49.
Yun
J
,
Rago
C
,
Cheong
I
,
Pagliarini
R
,
Angenendt
P
,
Rajagopalan
H
, et al
Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells
.
Science
2009
;
325
:
1555
9
.
50.
Berthe
A
,
Zaffino
M
,
Muller
C
,
Foulquier
F
,
Houdou
M
,
Schulz
C
, et al
Protein N-glycosylation alteration and glycolysis inhibition both contribute to the antiproliferative action of 2-deoxyglucose in breast cancer cells
.
Breast Cancer Res Treat
2018
;
171
:
581
91
.