Skip to main content

ORIGINAL RESEARCH article

Front. Oncol., 12 June 2024
Sec. Cancer Molecular Targets and Therapeutics

Auranofin and reactive oxygen species inhibit protein synthesis and regulate the level of the PLK1 protein in Ewing sarcoma cells

  • 1Carver College of Medicine, University of Iowa, Iowa City, IA, United States
  • 2Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Iowa, Iowa City, IA, United States

Novel therapeutic approaches are needed for the treatment of Ewing sarcoma tumors. We previously identified that Ewing sarcoma cell lines are sensitive to drugs that inhibit protein translation. However, translational and therapeutic approaches to inhibit protein synthesis in tumors are limited. In this work, we identified that reactive oxygen species, which are generated by a wide range of chemotherapy and other drugs, inhibit protein synthesis and reduce the level of critical proteins that support tumorigenesis in Ewing sarcoma cells. In particular, we identified that both hydrogen peroxide and auranofin, an inhibitor of thioredoxin reductase and regulator of oxidative stress and reactive oxygen species, activate the repressor of protein translation 4E-BP1 and reduce the levels of the oncogenic proteins RRM2 and PLK1 in Ewing and other sarcoma cell lines. These results provide novel insight into the mechanism of how ROS-inducing drugs target cancer cells via inhibition of protein translation and identify a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.

1 Introduction

Ewing sarcoma is an aggressive bone and soft-tissue tumor that is characterized by a recurrent chromosomal translocation involving the EWSR1 and FLI1 genes (13). The current treatment for Ewing sarcoma, which consists of cytotoxic chemotherapy combined with surgery and/or radiation, has changed very little in the past two decades and is associated with suboptimal outcomes (1, 4, 5). The EWS-FLI1 oncoprotein, which is required for tumorigenesis and only expressed in tumor cells, is an attractive therapeutic target in Ewing sarcoma, but has proven difficult to directly target (68). A complementary approach is to identify unique vulnerabilities incurred by the EWS-FLI1 oncoprotein (9, 10).

Recent studies identified protein translation as a potential therapeutic vulnerability and target in Ewing sarcoma tumors (11, 12). However, translational and therapeutic approaches to inhibit protein synthesis in tumors are lacking (13). Notably, reactive oxygen species (ROS), which are generated by a range of drugs via different mechanisms, have been linked to inhibition of protein synthesis in cancer cells (1417). Furthermore, Ewing sarcoma tumors are reported to be sensitive to reactive oxygen species and to the manipulation of oxidative stress (1824). For example, treatment of Ewing sarcoma tumors with fenretinide, a synthetic retinoid, causes ROS-dependent toxicity in vitro and in vivo in xenograft experiments (21, 22). Similarly, auranofin, an inhibitor of thioredoxin reductase and regulator of ROS, was identified using a multipronged screening approach as a drug with activity against Ewing sarcoma tumors (23). Moreover, from a mechanistic standpoint, SOX6, a transcriptional target of the EWS-FLI1 oncoprotein, was shown to increase ROS and oxidative stress in Ewing sarcoma cells via upregulation of thioredoxin interacting protein (TXNIP), an inhibitor of thioredoxin and inducer of ROS (24, 25). However, despite the sensitivity of Ewing sarcoma cells to ROS-inducing drugs, it is unknown whether ROS targets protein synthesis in this and other sarcoma types.

We began the current study with the aim to test the hypothesis that reactive oxygen species regulate protein synthesis in Ewing sarcoma cells. We found that both hydrogen peroxide and auranofin inhibit protein translation in Ewing and other sarcoma cells. Moreover, from a mechanistic standpoint, we also identified that ROS activates eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1), a repressor of protein translation that binds to the eukaryotic translation initiation factor eIF4E and prevents the formation of the translation initiation complex (26, 27). Activation of 4E-BP1 reduces the levels of the oncogenic proteins ribonucleotide reductase M2 (RRM2) and polo-like kinase 1 (PLK1) in Ewing and other sarcoma cell lines. Overall, these results define a link between ROS and protein synthesis in sarcoma cells and identify a mechanistic connection between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.

2 Materials and methods

2.1 Cell lines and culture

Cell lines were maintained at 37°C in a 5% CO2 atmosphere. The A673, TC71, and EW8 cell lines were provided by Dr. Kimberly Stegmaier (Dana-Farber Cancer Institute, Boston, MA), the CB-AGPN cell line was obtained from the Childhood Cancer Repository (Children’s Oncology Group), the HT1080 (RRID: CVCL_0317) and U2OS (RRID: CVCL_0042) cell lines were obtained from ATCC, and the RD cell line was provided by Dr. Munir Tanas (University of Iowa, Iowa City, IA). The non-transformed BJ-tert (RRID: CVCL_6573) and RPE-tert (RRID: CVCL_4388) cell lines were obtained from ATCC. Cells were grown in Dulbecco’s Modified Eagle’s Media (DMEM) supplemented with 10% FBS, 100 IU ml-1 penicillin and 100 µg ml-1 streptomycin. DNA fingerprinting confirmation of cell lines was performed using the short tandem repeat method and cell lines were used within 8–10 passages of thawing.

2.2 Chemical compounds

Auranofin and TAK-228 (sapanisertib) were obtained from MedChemExpress. N-acetylcysteine, hydrogen peroxide, and doxycycline were obtained from Sigma. Puromycin was obtained from ThermoFisher Scientific. Auranofin was dissolved in DMSO and then used at a 1:1000 dilution in assays. TAK-228 was also prepared in DMSO and used at a 1:1000 dilution in assays. N-acetylcysteine was prepared fresh for each experiment in DMEM media.

2.3 Cell viability and drug dose response assays

Cell proliferation was measured using the AlamarBlue (resazurin; Sigma) fluorescence assay, as previously described (11, 12, 28, 29). Briefly, approximately 5 x 104 cells were plated per well of a 96-well plate. Cells were then treated the next day with a range of drug concentrations for 72 hours. Fluorescence measurements were obtained, after adding the AlamarBlue reagent, using a FLUOstar Omega microplate reader (BMG Labtech). IC50 values were calculated using log-transformed and normalized data (GraphPad Prism 10.1.0). Cell viability was quantified using 0.4% Trypan Blue (ThermoFisher), which stains dead cells, with a DeNovix Cell Drop BF automated cell counter.

2.4 Puromycin labeling

Protein synthesis was assessed using puromycin labeling (SUnSET technique), as described (11, 12, 30, 31). For labeling of newly synthesized proteins, puromycin (2 μg/mL) was added to cells at a 1:400 dilution. The cells were then incubated with the puromycin for one hour before cell lysates were collected, as described in the Immunoblotting section. Protein loading for the immunoblots was normalized using cell number.

2.5 O-propargyl-puromycin labeling

Protein synthesis was assessed using O-propargyl-puromycin Click-iT labeling according to the manufacturer’s instructions (ThermoFisher Scientific) (32, 33). Flow cytometry was performed using a Becton Dickinson LSR II instrument. Briefly, cells were incubated with O-propargyl-puromycin (1:1000, 10 μM final concentration) for one hour. Cells were then collected using trypsin and fixed in a 4% paraformaldehyde fixative solution. Cells were washed by centrifugation and resuspended in Click-iT saponin-based permeabilization and wash reagent. Click-iT Plus reaction cocktail was added to the cell suspension for thirty minutes, after which the cells were washed by centrifugation. Flow cytometry was performed on a Becton Dickinson LSR II flow cytometer.

2.6 Protein isolation and immunoblotting

Protein extracts for immunoblotting were prepared by incubating cells in RIPA buffer (Boston BioProducts), supplemented with protease and phosphatase inhibitors (Halt Protease & Phosphatase Inhibitor Cocktail, EDTA-free; ThermoFisher Scientific), for 20 minutes. Supernatants were collected following a 15-minute centrifugation at 17,000 r.c.f. at 4°C. SDS-PAGE was used to separate proteins, which were then transferred to polyvinylidene difluoride membranes (Millipore). Antibodies to the following proteins were used in the immunoblots: puromycin (Millipore, #AF488, 1:2000; RRID: AB_2737590), 4E-BP1 (Cell Signaling, #9644, 1:1000; RRID: AB_2097841), p-4E-BP1–37/46 (Cell Signaling, #2855, 1:1000; RRID: AB_560835), RRM2 (Santa Cruz Biotechnology, #398294, 1:500; RRID: AB_2894824), PLK1 (Cell Signaling, #4513, 1:1000; RRID: AB_2167409), tubulin (Proteintech, #66031, 1:2000; RRID: AB_28834830), and vinculin (Proteintech, #26520, 1:5000; RRID: AB_2919877). Protein loading for the immunoblots was normalized using cell number.

2.7 Reactive oxygen species detection

ROS was quantified using a DCFDA (2’,7’-dichlorofluorescin diacetate) assay kit according to the manufacturer’s instructions (Abcam). Briefly, 1 x 104 cells were plated per well of a 96-well plate in phenol red-free media. Drugs were then added the next day and incubated for the amount of time indicated in the figure legends. The DCDFA reagent, which was dissolved in the kit buffer at a 20 mM concentration, was added to the wells at a 1:800 dilution and then incubated for 45 minutes at 37°C. Fluorescence measurements (excitation 485 nM/emission 535 nM) were obtained using a FLUOstar Omega microplate reader (BMG Labtech).

2.8 EdU labeling

Detection of DNA replication was performed in duplicate using a Click-iT EdU-488 kit (ThermoFisher Scientific) (34). Briefly, cells were labeled with 10 μM EdU for one hour and then harvested using trypsin and fixed using the Click-iT fixative buffer. Cells were then washed by centrifugation and resuspended in Click-iT saponin-based permeabilization and wash reagent. Click-iT Plus reaction cocktail was added to the cell suspension for thirty minutes, after which the cells were washed by centrifugation. Flow cytometry was performed on a Becton Dickinson LSR II flow cytometer.

2.9 Doxycycline-inducible 4E-BP1-Ala

The full-length 4E-BP1 cDNA, with alanine substitutions at Thr37, Thr46, Ser65, and Thr70 and a FLAG-tag, was obtained as a gene block (IDT; Coralville, IA) and cloned into a lentiviral vector as described in previous publications (12, 34).

2.10 Reverse phase protein array

RPPA analysis of cell lines were performed by the RPPA Core Facility at the MD Anderson Cancer Center. Cells were provided to the core facility as frozen pellets and the protein extraction, data normalization, and analysis were performed according to facility protocols (https://www.mdanderson.org/research/research-resources/core-facilities/functional-proteomics-rppa-core/education-and-references.html) (RRID: SCR_016649).

2.11 Statistical analysis

Student’s t-test two-tailed was used to calculate P-values for the comparison of two groups. Statistical analyses were conducted using GraphPad Prism 10.1.0 (RRID: SCR_002798).

3 Results

3.1 Hydrogen peroxide inhibits protein synthesis in sarcoma cell lines

To directly test the hypothesis that ROS inhibits protein synthesis in Ewing sarcoma cells we treated two cell lines, EW8 and TC71, with hydrogen peroxide for three hours and then measured nascent protein synthesis using puromycin labeling (SUnSET technique) and immunoblotting (30, 31). Figure 1A shows that hydrogen peroxide decreases protein synthesis in Ewing sarcoma cells and that this effect is attenuated by co-treatment of the cells with the ROS-scavenger n-acetylcysteine (NAC). We also used a flow cytometry approach (O-propargyl-puromycin) to quantify, in single cells, the effect of hydrogen peroxide on protein synthesis (32, 33). Notably, hydrogen peroxide reduced protein synthesis to the same extent as cycloheximide, a well-established inhibitor of protein synthesis that blocks translation elongation, and this effect was blocked by co-treatment with NAC (Figures 1B, C). We then tested additional sarcoma cell lines, HT1080 (fibrosarcoma), RD (rhabdomyosarcoma), and U2OS (osteosarcoma), that represent other sarcoma subtypes and found that hydrogen peroxide also inhibited protein synthesis, similar to the results obtained with the Ewing sarcoma cell lines (Figure 1D).

Figure 1
www.frontiersin.org

Figure 1 Hydrogen peroxide inhibits protein synthesis in sarcoma cell lines. (A) EW8 and TC71 cells were treated with hydrogen peroxide, NAC, or the combination for three hours. Cells were labeled with puromycin to quantify protein synthesis and then lysates were collected for immunoblotting. (B, C) EW8 (B) and TC71 (C) cells were treated with hydrogen peroxide (100 μM), NAC (5 mM), or the combination for three hours and then labeled with O-propargyl-puromycin for quantification of protein synthesis by flow cytometry. (D) Additional sarcoma cell lines were treated with hydrogen peroxide, NAC, or the combination for three hours and then labeled with puromycin for immunoblotting. (E) EW8 and TC71 cells were treated with hydrogen peroxide for one hour and then protein synthesis was assessed using puromycin. (F) Ewing sarcoma cells were treated with hydrogen peroxide (100 μM) for three hours and then allowed to recover for twenty-one hours before protein synthesis was assessed using puromycin.

Next, we treated the Ewing sarcoma cells lines with hydrogen peroxide for one hour, in contrast to the three hours used in the previous experiments, and identified a significant decrease in protein synthesis at this earlier time point, albeit to a lesser extent than at the later time point (Figure 1E). We also treated Ewing sarcoma cells lines with hydrogen peroxide for three hours and then removed the hydrogen peroxide and allowed the cells to recover for twenty-one hours before measuring nascent protein synthesis. Figure 1F shows that a three-hour treatment with hydrogen peroxide is sufficient to cause prolonged inhibition of protein synthesis, that extends after the hydrogen peroxide is removed. In addition, this more prolonged inhibition of protein synthesis also reduced the level of the protein loading control, tubulin. Furthermore, staining total protein with Ponceau S showed a significant reduction in total protein levels at the later time point. Of note, the loading for these and subsequent immunoblots were normalized using cell number and not total protein due to the effects of the drugs on protein synthesis.

3.2 Auranofin inhibits protein synthesis in Ewing sarcoma cell lines

Auranofin is an inhibitor of thioredoxin reductase and regulator of oxidative stress that is used for the treatment of rheumatoid arthritis (35, 36). Auranofin was previously identified, using a multipronged drug screening approach, as a drug that inhibits the growth of Ewing sarcoma cells in vitro and in vivo (23). Of note, this sensitivity of Ewing sarcoma cell lines to auranofin was dependent on expression of the EWS-FLI1 oncoprotein (23). Consistent with this result, analysis of Cancer Dependency Map data (Broad Institute) identifies that Ewing sarcoma cell lines are significantly more sensitive to CRISPR-mediate knockout of thioredoxin reductase than other cancer cell lines (Figure 2A) (37, 38). Figure 2B demonstrates that treatment of Ewing sarcoma cells with auranofin increases reactive oxygen species, as detected using the fluorescent ROS-detector DCFDA (2’,7’-dichlorofluorescin diacetate). Auranofin, as previously reported, also decreases the growth of Ewing sarcoma cell lines in a 72 hour growth assay (Figure 2C) (23). Next, we assessed the effect of auranofin dose on protein synthesis using a 24 hour drug treatment and identified that inhibition of protein synthesis occurred in the low (25) micromolar dose range (Supplementary Figure 1). To confirm that the effect of auranofin on protein synthesis is mediated by ROS we performed a co-treatment experiment with NAC and found that the ROS scavenger was able to block the effects of auranofin on nascent protein synthesis (Figure 2D). We also used flow cytometry (O-propargyl-puromycin) to investigate the effect of auranofin on protein synthesis in single cells and confirmed that the drug reduces protein synthesis (Figures 2E, F). The effect of auranofin on protein synthesis was then assessed at an earlier time point, 6 hours after drug addition, compared to the timepoint of 24 hours used in the previous experiments. Figure 2G demonstrates that auranofin inhibits protein synthesis in Ewing sarcoma cells as early as 6 hours after drug addition, although the effect is larger with a longer duration of drug treatment. We also used flow cytometry and O-propargyl-puromycin labeling to validate that auranofin inhibits protein synthesis at this earlier (6 hour) time point (Supplementary Figures 1B, C). Cell viability was not significantly compromised at either the 6 hour or 24 hour timepoints (Supplementary Figures 1D, E). We then tested additional sarcoma cell lines that represent other sarcoma subtypes and found that auranofin also inhibited protein synthesis, similar to the results obtained with the Ewing sarcoma cell lines (Figure 2H). Finally, we treated two non-transformed cell lines, BJ-tert and RPE-tert, with auranofin and then labeled newly synthesized proteins with puromycin. Supplementary Figure 2A shows that auranofin inhibits protein synthesis in these non-transformed cell lines, and the effect is rescued by treatment with NAC. However, in contrast to the Ewing sarcoma cell lines, auranofin had minimal effect on the growth of the BJ-tert and RPE-tert cell lines in dose response assays, suggesting that the differential toxicity of the drug with the Ewing sarcoma and non-transformed cells is due to an increased reliance on active protein synthesis in the cancer cells (Supplementary Figures 2B, C).

Figure 2
www.frontiersin.org

Figure 2 Auranofin inhibits protein synthesis in Ewing sarcoma cell lines. (A) Dependency Map data (https://depmap.org/portal/) showing the effect of CRISPR knockout of thioredoxin reductase on the growth of cancer cell lines. (B) EW8 and TC71 cells were treated with auranofin (5 μM) for twenty-four hours, or hydrogen peroxide (100 μM) for three hours, and then ROS was quantified using a DCFDA fluorescence assay and normalized to cells treated with DMSO. Error bars represent the mean ± SD of three independent experiments with six technical replicates per drug treatment. (C) Dose response curves for Ewing sarcoma cells treated with auranofin. Cell viability was assessed 72 hours after drug was added using the AlamarBlue assay. Error bars represent the mean ± SD of three technical replicates. The results are representative of two independent experiments. (D) EW8 and TC71 cells were treated with auranofin, NAC, or the combination for twenty-four hours. Cells were labeled with puromycin to quantify protein synthesis and then lysates were collected for immunoblotting. (E, F) EW8 (E) and TC71 (F) cells were treated with auranofin (5 μM) for twenty-four hours and then labeled with O-propargyl-puromycin for quantification of protein synthesis by flow cytometry. (G) EW8 and TC71 cells were treated with auranofin, NAC, or the combination for either six or twenty-four hours and then labeled with puromycin to quantify protein synthesis. (H) Additional sarcoma cell lines were treated with auranofin, NAC, or the combination for twenty-four hours and then labeled with puromycin for immunoblotting. Protein loading for the immunoblots was normalized using cell number. ****P < 0.0001.

3.3 Auranofin reduces the level of the RRM2 protein and blocks DNA replication Ewing sarcoma cells

In previous work, we identified that Ewing sarcoma cell lines require active protein synthesis to maintain a stable level of the ribonucleotide reductase M2 (RRM2) protein, which is a potential therapeutic target in Ewing sarcoma tumors (12, 34, 39, 40). Consequently, we next evaluated whether auranofin and ROS downregulate RRM2 levels in Ewing sarcoma cells. Figure 3A demonstrates that auranofin depletes RRM2 in Ewing sarcoma cell lines and that this effect is blocked by co-treatment of the cells with the ROS-scavenger NAC. Knockdown or inhibition of RRM2 in Ewing sarcoma is lethal and Figure 3B shows that auranofin significantly impairs the growth of four different Ewing sarcoma cell lines and that this effect is abrogated by co-treatment with NAC. RRM2 is a subunit of ribonucleotide reductase, which is the rate limiting complex in the synthesis of deoxyribonucleotides, and required for progression through S-phase (4143). Cell cycle analysis demonstrates that auranofin blocks DNA replication, as assessed using EdU labeling, and arrests cells throughout S-phase, as expected for a drug that inhibits ribonucleotide reductase activity (Figure 3C).

Figure 3
www.frontiersin.org

Figure 3 Auranofin reduces the level of the RRM2 protein and blocks DNA replication Ewing sarcoma cells. (A) EW8 and TC71 cells were treated with auranofin, NAC, or the combination for twenty-four hours and then cell lysate was collected for immunoblotting for RRM2. Protein loading for the immunoblots was normalized using cell number. (B) Ewing sarcoma cell lines were treated with auranofin (5 μM), NAC (5 mM), or the combination for 72 hours and then cell viability was quantified using AlamarBlue. Error bars represent the mean ± SD of four technical replicates. The results are representative of two independent experiments. (C) Ewing sarcoma cell lines were treated with auranofin for twenty-four hours and then the cell cycle distribution was analyzed using EdU labeling and propidium iodide. ****P < 0.0001.

3.4 Auranofin and reactive oxygen species activate the translational repressor 4E-BP1 in sarcoma cell lines

Reactive oxygen species are reported to inhibit protein synthesis via multiple mechanisms, including modification of cysteines in ribosomal proteins and activation of the translational repressor 4E-BP1 (1416). Specifically, 4E-BP1 binds to the eukaryotic translation initiation factor eIF4E and prevents the formation of the translation initiation complex (26, 27). Notably, in previous work, we identified a role for 4E-BP1, which is regulated by phosphorylation, in blocking protein synthesis in Ewing sarcoma cells (11, 12). Consequently, we tested whether ROS activates 4E-BP1 in Ewing sarcoma cells. Figure 4A shows that hydrogen peroxide decreases the phosphorylation of 4E-BP1, which reflects activation of the translational repressor, in Ewing sarcoma cell lines and this effect is blocked by co-treatment with NAC. Similarly, auranofin also inhibits the phosphorylation of 4E-BP1 and, thereby, activates the repressor of cap-dependent protein translation (Figure 4B). To investigate the effect of active 4E-BP1 we previously generated multiple sarcoma cell lines that express a doxycycline-inducible, constitutively-active 4E-BP1 protein with alanine substitutions at the phosphorylation sites Thr37, Thr46, Ser65, and Thr70 (TO-4E-BP1-Ala) (12, 44). Figure 4C shows that the expression of constitutively-active 4E-BP1-Ala in Ewing sarcoma cells inhibits protein synthesis and reduces the level of the RRM2 protein, which we previously showed is regulated by 4E-BP1 (12). To identify additional proteins that are regulated by 4E-BP1 we evaluated the effect of the inducible, constitutively-active 4E-BP1-Ala on the proteome of multiple sarcoma cell lines, described in an earlier publication, using reverse phase protein arrays (RPPA) (Figure 4D; Supplementary Table 1) (12). Overlap of the RPPA data for the five cell lines identified that the expression of active 4E-BP1-Ala in all of the sarcoma cell lines results in the downregulation of the level of the PLK1 protein (Figure 4E; Supplementary Table 2).

Figure 4
www.frontiersin.org

Figure 4 Auranofin and reactive oxygen species activate the translational repressor 4E-BP1 in sarcoma cell lines. (A) EW8 and TC71 cells were treated with hydrogen peroxide, NAC, or the combination for three hours and then cell lysates were collected for immunoblotting. Protein loading for the immunoblots was normalized using cell number. (B) EW8 and TC71 cells were treated with auranofin, NAC, or the combination for twenty-four hours and then cell lysates were collected for immunoblotting. Protein loading for the immunoblots was normalized using cell number. (C) TO-4E-BP1-Ala cell lines were treated with doxycycline for 48 hours to induce expression of 4E-BP1-Ala and then cell lysates were collected after labeling the cells with puromycin. (D) Comparison of protein expression (RPPA) in TO-4E-BP1-Ala sarcoma cell lines treated with vehicle or doxycycline for 48 hours. (E) Venn diagram showing the overlap of the RPPA data for proteins that are downregulated by >1.25-fold in the TO-4E-BP1-Ala sarcoma cell lines in the presence of doxycycline.

3.5 Auranofin and active 4E-BP1 reduce the level of the PLK1 protein in sarcoma cell lines

PLK1 is a well-described therapeutic target in multiple types of cancer, including Ewing sarcoma (4549). To validate PLK1 as a target of 4E-BP1 we treated the 4E-BP1-Ala cells with doxycycline and performed immunoblotting for PLK1. Figure 5A shows that the expression constitutively-active 4E-BP1 reduces the level of the PLK1 protein, consistent with the RPPA results. Auranofin also reduced the level of the PLK1 protein in Ewing (Figure 5B) and other (Figure 5C) sarcoma cell lines. In previous work, we identified that inhibition of mTORC1/2 activates 4E-BP1 and inhibits protein synthesis in sarcoma cell lines (12). Figure 5D shows that treatment of sarcoma cell lines with the mTORC1/2 inhibitor TAK-228 activates 4E-BP1 and downregulates the level of the PLK1 protein (Figure 5D).

Figure 5
www.frontiersin.org

Figure 5 Auranofin and active 4E-BP1 reduce the level of the PLK1 protein in sarcoma cell lines. (A) TO-4E-BP1-Ala cell lines were treated with doxycycline for 48 hours to induce expression of 4E-BP1-Ala and then cell lysates were collected for immunoblotting. (B) EW8 and TC71 cells were treated with auranofin for either six or twenty-four hours and then cell lysates were collected for immunoblotting. (C) Additional sarcoma cell lines were treated with auranofin, NAC, or the combination for twenty-four hours and then cell lysates were collected for immunoblotting for PLK1. (D) Sarcoma cell lines were treated with the mTORC1/2 inhibitor TAK-228 for twenty-four hours and then cell lysates were collected for immunoblotting. Protein loading for the immunoblots was normalized using cell number.

4 Discussion

Ewing sarcoma is an aggressive sarcoma of the bone and soft tissue that is treated with chemotherapy in combination with surgery and/or radiation (1, 2). Unfortunately, the standard treatment regimen for Ewing sarcoma is unchanged in over two decades and this therapy is associated with significant on- and off-treatment morbidities (4, 5). Consequently, there is an urgent need to identify new therapeutic approaches and targets in Ewing sarcoma tumors (8). This study investigated the effects of reactive oxygen species (ROS) on protein synthesis in Ewing sarcoma cell lines. We found that hydrogen peroxide or auranofin, an inhibitor of thioredoxin reductase and ROS inducer, inhibited protein synthesis, activated the repressor of cap-dependent protein translation 4E-BP1, and reduced the levels of the proteins RRM2 and PLK1, both of which promote tumorigenesis in Ewing sarcoma tumors (12, 34, 39, 40, 4547).

Our findings provide novel insight into the mechanism of how ROS-inducing drugs target cancer cells. It is well-established that ROS can damage DNA, leading to apoptosis or cell cycle arrest (50, 51). However, our results demonstrate that ROS can also target protein synthesis, which is another essential process for cell survival and proliferation (13). Of note, we found that auranofin also inhibits protein synthesis in non-transformed cells (BJ-tert and RPE-tert), but the drug had minimal effects on the growth and proliferation of these cells. This suggest that that the differential toxicity of auranofin between the Ewing sarcoma and non-transformed cells is due to an increased reliance on active protein synthesis in the cancer cells, which is well-described in the literature (11, 13, 26, 5257).

Although we identified that ROS activates 4E-BP1 we expect that the mechanism of how ROS inhibits protein synthesis is complex and not limited to only activation of 4E-BP1. For example, recent work identified that ROS selectively targets and modifies cysteine residues in ribosomal proteins and, thereby, reduces protein synthesis (15). Future work will focus on delineating the contributions of these different targets of ROS and their effects on protein synthesis. In addition, we also plan to investigate the mechanism of how ROS activates 4E-BP1 in sarcoma cell lines. Work in other cell types has shown that ROS can inhibit mTOR activity and activate 4E-BP1 via activation of the peroxisome-bound tuberous sclerosis complex (TSC) and this will be a focus of future investigation (16, 58, 59). Finally, future work will also investigate the impact of the different types of ROS on protein synthesis.

Auranofin, an inhibitor of thioredoxin reductase, is approved for the treatment of rheumatoid arthritis (35, 36). Notably, Pessetto et al. previously described in vitro and in vivo sensitivity of Ewing sarcoma cells to auranofin (23). In addition, the sensitivity of Ewing sarcoma cell lines to auranofin was dependent on the expression of the EWS-FLI1 oncoprotein, which suggests that thioredoxin reductase may target a unique vulnerability in Ewing sarcoma tumors (23). Other groups have also identified that Ewing sarcoma cells are sensitive to reactive oxygen species and to the manipulation of oxidative stress, using drugs such as fenretinide (18, 21, 22). Furthermore, a number of drugs commonly used in the clinic, including doxorubicin and cisplatin, are known to generate ROS, although it is currently unknown whether inhibition of protein synthesis may contribute to the mechanisms of action of these drugs (15). Recent work also identified that checkpoint kinase 1 (CHK1), which we and others previously identified as therapeutic target in Ewing sarcoma tumors, functions as a nuclear H2O2 sensor and regulates a cellular program that dampens ROS (15). Notably, CHK1 inhibition was shown to increase steady-state levels of nuclear H2O2, which suggests that combining auranofin, or other ROS-inducing drug, with a CHK1 inhibitor could enhance toxicity (15, 60). These drug combinations will be investigated in future work.

Overall, our data demonstrate that reactive oxygen species activate 4E-BP1, which blocks cap-dependent protein translation, and inhibit protein synthesis in sarcoma cells. We also identified a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author.

Ethics statement

Ethical approval was not required for the studies on humans in accordance with the local legislation and institutional requirements because only commercially available established cell lines were used.

Author contributions

JH: Data curation, Investigation, Methodology, Writing – review & editing. SK: Data curation, Investigation, Writing – review & editing. EG: Data curation, Investigation, Writing – review & editing. DG: Writing – original draft, Writing – review & editing, Conceptualization, Formal Analysis, Funding acquisition, Investigation, Project administration, Resources.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. DG is supported by The Matt Morrell and Natalie Sanchez Pediatric Cancer Research Foundation, the Stead Fund for Leadership Development & Innovation in Pediatric Medicine, Sammy’s Superheroes, Cal’s Angels, Unravel Pediatric Cancer, Hyundai Hope on Wheels, the Sarah Waul Family Fund, and NIH Grant R37-CA217910. This work was also supported by a grant (P30CA086862) from the NCI, administered through the Holden Comprehensive Cancer Center at The University of Iowa.

Acknowledgments

The authors would like to acknowledge use of the University of Iowa Flow Cytometry Facility and the DNA sequencing facility (Genomics Division of the Iowa Institute of Human Genetics) which are supported, in part, by the University of Iowa Carver College of Medicine and the Holden Comprehensive Cancer Center (NCI P30CA086862).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2024.1394653/full#supplementary-material

Nomenclature

ROS, reactive oxygen species; DCFDA, 2’,7’-dichlorofluorescin diacetate; RRM2, ribonucleotide reductase M2; PLK1, polo-like kinase 1; 4E-BP1, eukaryotic translation initiation factor 4E binding protein 1; TXNRD1, thioredoxin reductase; EdU, 5-ethynyl-2’-deoxyuridine; PI, propidium iodide; TSC, tuberous sclerosis complex; RPPA, reverse phase protein array; NAC, n-acetylcysteine; H2O2, hydrogen peroxide; OPP, O-propargyl-puromycin; RIPA, radioimmunoprecipitation assay.

References

1. Riggi N, Suvà ML, Stamenkovic I. Ewing’s sarcoma. N Engl J Med. (2021) 384:154–64. doi: 10.1056/NEJMra2028910

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Grünewald TGP, Cidre-Aranaz F, Surdez D, Tomazou EM, de Álava E, Kovar H, et al. Ewing sarcoma. Nat Rev Dis Primers. (2018) 4:5. doi: 10.1038/s41572-018-0003-x

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Balamuth NJ, Womer RB. Ewing’s sarcoma. Lancet Oncol. (2010) 11:184–92. doi: 10.1016/S1470-2045(09)70286-4

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Womer RB, West DC, Krailo MD, Dickman PS, Pawel BR, Grier HE, et al. Randomized controlled trial of interval-compressed chemotherapy for the treatment of localized Ewing sarcoma: a report from the Children’s Oncology Group. J Clin Oncol. (2012) 30:4148–54. doi: 10.1200/JCO.2011.41.5703

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Leavey PJ, Laack NN, Krailo MD, Buxton A, Randall RL, DuBois SG, et al. Phase III trial adding vincristine-topotecan-cyclophosphamide to the initial treatment of patients with nonmetastatic ewing sarcoma: A children’s oncology group report. J Clin Oncol. (2021) 39:4029–38. doi: 10.1200/JCO.21.00358

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Cote GM, Choy E. Update in treatment and targets in Ewing sarcoma. Hematol Oncol Clin North Am. (2013) 27:1007–19. doi: 10.1016/j.hoc.2013.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Grohar PJ, Helman LJ. Prospects and challenges for the development of new therapies for Ewing sarcoma. Pharmacol Ther. (2013) 137:216–24. doi: 10.1016/j.pharmthera.2012.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Kovar H, Amatruda J, Brunet E, Burdach S, Cidre-Aranaz F, de Alava E, et al. The second European interdisciplinary Ewing sarcoma research summit–A joint effort to deconstructing the multiple layers of a complex disease. Oncotarget. (2016) 7:8613–24. doi: 10.18632/oncotarget.v7i8

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Kovar H. Downstream EWS/FLI1 - upstream Ewing’s sarcoma. Genome Med. (2010) 2:8. doi: 10.1186/gm129

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Kovar H. Blocking the road, stopping the engine or killing the driver? Advances in targeting EWS/FLI-1 fusion in Ewing sarcoma as novel therapy. Expert Opin Ther Targets. (2014) 18:1315–28. doi: 10.1517/14728222.2014.947963

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Koppenhafer SL, Goss KL, Terry WW, Gordon DJ. mTORC1/2 and protein translation regulate levels of CHK1 and the sensitivity to CHK1 inhibitors in ewing sarcoma cells. Mol Cancer Ther. (2018) 17:2676–88. doi: 10.1158/1535-7163.MCT-18-0260

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Goss KL, Koppenhafer SL, Waters T, Terry WW, Wen K-K, Wu M, et al. The translational repressor 4E-BP1 regulates RRM2 levels and functions as a tumor suppressor in Ewing sarcoma tumors. Oncogene. (2021) 40:564–77. doi: 10.1038/s41388-020-01552-0

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kovalski JR, Kuzuoglu-Ozturk D, Ruggero D. Protein synthesis control in cancer: selectivity and therapeutic targeting. EMBO J. (2022) 41:e109823. doi: 10.15252/embj.2021109823

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Topf U, Suppanz I, Samluk L, Wrobel L, Böser A, Sakowska P, et al. Quantitative proteomics identifies redox switches for global translation modulation by mitochondrially produced reactive oxygen species. Nat Commun. (2018) 9:324. doi: 10.1038/s41467-017-02694-8

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Zhang J, Simpson CM, Berner J, Chong HB, Fang J, Ordulu Z, et al. Systematic identification of anticancer drug targets reveals a nucleus-to-mitochondria ROS-sensing pathway. Cell. (2023) 186:2361–79. doi: 10.1016/j.cell.2023.04.026

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Ye Q, Zhang Y, Cao Y, Wang X, Guo Y, Chen J, et al. Frenolicin B targets peroxiredoxin 1 and glutaredoxin 3 to trigger ROS/4E-BP1-mediated antitumor effects. Cell Chem Biol. (2019) 26:366–77. doi: 10.1016/j.chembiol.2018.11.013

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Pensabene KM, LaMorte J, Allender AE, Wehr J, Kaur P, Savage M, et al. Acute oxidative stress can paradoxically suppress human NRF2 protein synthesis by inhibiting global protein translation. Antioxid (Basel). (2023) 12:1735. doi: 10.3390/antiox12091735

CrossRef Full Text | Google Scholar

18. Smith DG, Magwere T, Burchill SA. Oxidative stress and therapeutic opportunities: focus on the Ewing’s sarcoma family of tumors. Expert Rev Anticancer Ther. (2011) 11:229–49. doi: 10.1586/era.10.224

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Grunewald TGP, Diebold I, Esposito I, Plehm S, Hauer K, Thiel U, et al. STEAP1 is associated with the invasive and oxidative stress phenotype of Ewing tumors. Mol Cancer Res. (2012) 10:52–65. doi: 10.1158/1541-7786.MCR-11-0524

PubMed Abstract | CrossRef Full Text | Google Scholar

20. El-Naggar AM, Somasekharan SP, Wang Y, Cheng H, Negri GL, Pan M, et al. Class I HDAC inhibitors enhance YB-1 acetylation and oxidative stress to block sarcoma metastasis. EMBO Rep. (2019) 20:e48375. doi: 10.15252/embr.201948375

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Myatt SS, Redfern CPF, Burchill SA. p38MAPK-Dependent sensitivity of Ewing’s sarcoma family of tumors to fenretinide-induced cell death. Clin Cancer Res. (2005) 11:3136–48. doi: 10.1158/1078-0432.CCR-04-2050

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Myatt SS, Burchill SA. The sensitivity of the Ewing’s sarcoma family of tumours to fenretinide-induced cell death is increased by EWS-Fli1-dependent modulation of p38(MAPK) activity. Oncogene. (2008) 27:985–96. doi: 10.1038/sj.onc.1210705

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Pessetto ZY, Chen B, Alturkmani H, Hyter S, Flynn CA, Baltezor M, et al. In silico and in vitro drug screening identifies new therapeutic approaches for Ewing sarcoma. Oncotarget. (2017) 8:4079–95. doi: 10.18632/oncotarget.v8i3

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Marchetto A, Ohmura S, Orth MF, Knott MML, Colombo MV, Arrigoni C, et al. Oncogenic hijacking of a developmental transcription factor evokes vulnerability toward oxidative stress in Ewing sarcoma. Nat Commun. (2020) 11:2423. doi: 10.1038/s41467-020-20017-2

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Marchetto A, Grünewald TGP. SOX6: a double-edged sword for Ewing sarcoma. Mol Cell Oncol. (2020) 7:1783081. doi: 10.1080/23723556.2020.1783081

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Musa J, Orth MF, Dallmayer M, Baldauf M, Pardo C, Rotblat B, et al. Eukaryotic initiation factor 4E-binding protein 1 (4E-BP1): a master regulator of mRNA translation involved in tumorigenesis. Oncogene. (2016) 35:4675–88. doi: 10.1038/onc.2015.515

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Qin X, Jiang B, Zhang Y. 4E-BP1, a multifactor regulated multifunctional protein. Cell Cycle. (2016) 15:781–6. doi: 10.1080/15384101.2016.1151581

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Koppenhafer SL, Goss KL, Voigt E, Croushore E, Terry WW, Ostergaard J, et al. Inhibitor of DNA binding 2 (ID2) regulates the expression of developmental genes and tumorigenesis in ewing sarcoma. Oncogene. (2022) 41(20):2873–84. doi: 10.1038/s41388-022-02310-0

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Waters T, Goss KL, Koppenhafer SL, Terry WW, Gordon DJ. Eltrombopag inhibits the proliferation of Ewing sarcoma cells via iron chelation and impaired DNA replication. BMC Cancer. (2020) 20:1171. doi: 10.1186/s12885-020-07668-6

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Schmidt EK, Clavarino G, Ceppi M, Pierre P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat Methods. (2009) 6:275–7. doi: 10.1038/nmeth.1314

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Goodman CA, Mabrey DM, Frey JW, Miu MH, Schmidt EK, Pierre P, et al. Novel insights into the regulation of skeletal muscle protein synthesis as revealed by a new nonradioactive in vivo technique. FASEB J. (2011) 25:1028–39. doi: 10.1096/fj.10-168799

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Nagelreiter F, Coats MT, Klanert G, Gludovacz E, Borth N, Grillari J, et al. OPP labeling enables total protein synthesis quantification in CHO production cell lines at the single-cell level. Biotechnol J. (2018) 13:e1700492. doi: 10.1002/biot.201700492

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Forester CM, Zhao Q, Phillips NJ, Urisman A, Chalkley RJ, Oses-Prieto JA, et al. Revealing nascent proteomics in signaling pathways and cell differentiation. Proc Natl Acad Sci USA. (2018) 115:2353–8. doi: 10.1073/pnas.1707514115

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Koppenhafer SL, Goss KL, Terry WW, Gordon DJ. Inhibition of the ATR-CHK1 pathway in ewing sarcoma cells causes DNA damage and apoptosis via the CDK2-mediated degradation of RRM2. Mol Cancer Res. (2020) 18:91–104. doi: 10.1158/1541-7786.MCR-19-0585

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Abdalbari FH, Telleria CM. The gold complex auranofin: new perspectives for cancer therapy. Discovery Oncol. (2021) 12:42. doi: 10.1007/s12672-021-00439-0

CrossRef Full Text | Google Scholar

36. Gamberi T, Chiappetta G, Fiaschi T, Modesti A, Sorbi F, Magherini F. Upgrade of an old drug: Auranofin in innovative cancer therapies to overcome drug resistance and to increase drug effectiveness. Med Res Rev. (2022) 42:1111–46. doi: 10.1002/med.21872

PubMed Abstract | CrossRef Full Text | Google Scholar

37. DepMap: The Cancer Dependency Map Project at Broad Institute (2020). Available online at: https://depmap.org/portal/.

Google Scholar

38. Corsello SM, Nagari RT, Spangler RD, Rossen J, Kocak M, Bryan JG, et al. Discovering the anti-cancer potential of non-oncology drugs by systematic viability profiling. Nat Cancer. (2020) 1:235–48. doi: 10.1038/s43018-019-0018-6

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Goss KL, Gordon DJ. Gene expression signature based screening identifies ribonucleotide reductase as a candidate therapeutic target in Ewing sarcoma. Oncotarget. (2016) 7:63003–19. doi: 10.18632/oncotarget.v7i39

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Ohmura S, Marchetto A, Orth MF, Li J, Jabar S, Ranft A, et al. Translational evidence for RRM2 as a prognostic biomarker and therapeutic target in Ewing sarcoma. Mol Cancer. (2021) 20:97. doi: 10.1186/s12943-021-01393-9

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Kolberg M, Strand KR, Graff P, Andersson KK. Structure, function, and mechanism of ribonucleotide reductases. Biochim Biophys Acta. (2004) 1699:1–34. doi: 10.1016/j.bbapap.2004.02.007

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Greene BL, Kang G, Cui C, Bennati M, Nocera DG, Drennan CL, et al. Ribonucleotide reductases: structure, chemistry, and metabolism suggest new therapeutic targets. Annu Rev Biochem. (2020) 89:45–75. doi: 10.1146/annurev-biochem-013118-111843

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Cerqueira NMFSA, Pereira S, Fernandes PA, Ramos MJ. Overview of ribonucleotide reductase inhibitors: an appealing target in anti-tumour therapy. Curr Med Chem. (2005) 12:1283–94. doi: 10.2174/0929867054020981

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Wang Z, Feng X, Molinolo AA, Martin D, Vitale-Cross L, Nohata N, et al. 4E-BP1 is a tumor suppressor protein reactivated by mTOR inhibition in head and neck cancer. Cancer Res. (2019) 79:1438–50. doi: 10.1158/0008-5472.CAN-18-1220

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Wei LM, Hugle M, Fulda S. Eribulin alone or in combination with the PLK1 inhibitor BI 6727 triggers intrinsic apoptosis in Ewing sarcoma cell lines. Oncotarget. (2017) 8:52445–56. doi: 10.18632/oncotarget.v8i32

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Weiß LM, Hugle M, Romero S, Fulda S. Synergistic induction of apoptosis by a polo-like kinase 1 inhibitor and microtubule-interfering drugs in Ewing sarcoma cells. Int J Cancer. (2016) 138:497–506. doi: 10.1002/ijc.29725

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Li J, Ohmura S, Marchetto A, Orth MF, Imle R, Dallmayer M, et al. Therapeutic targeting of the PLK1-PRC1-axis triggers cell death in genomically silent childhood cancer. Nat Commun. (2021) 12:5356. doi: 10.1038/s41467-021-25553-z

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Chiappa M, Petrella S, Damia G, Broggini M, Guffanti F, Ricci F. Present and future perspective on PLK1 inhibition in cancer treatment. Front Oncol. (2022) 12:903016. doi: 10.3389/fonc.2022.903016

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Stafford JM, Wyatt MD, McInnes C. Inhibitors of the PLK1 polo-box domain: drug design strategies and therapeutic opportunities in cancer. Expert Opin Drug Discovery. (2023) 18:65–81. doi: 10.1080/17460441.2023.2159942

CrossRef Full Text | Google Scholar

50. Srinivas US, Tan BWQ, Vellayappan BA, Jeyasekharan AD. ROS and the DNA damage response in cancer. Redox Biol. (2019) 25:101084. doi: 10.1016/j.redox.2018.101084

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Zhao Y, Ye X, Xiong Z, Ihsan A, Ares I, Martínez M, et al. Cancer metabolism: the role of ROS in DNA damage and induction of apoptosis in cancer cells. Metabolites. (2023) 13:796. doi: 10.3390/metabo13070796

CrossRef Full Text | Google Scholar

52. White-Gilbertson S, Kurtz DT, Voelkel-Johnson C. The role of protein synthesis in cell cycling and cancer. Mol Oncol. (2009) 3:402–8. doi: 10.1016/j.molonc.2009.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Knight JRP, Sansom OJ. Tuning protein synthesis for cancer therapy. Mol Cell Oncol. (2021) 8:1884034. doi: 10.1080/23723556.2021.1884034

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Ruggero D. The role of Myc-induced protein synthesis in cancer. Cancer Res. (2009) 69:8839–43. doi: 10.1158/0008-5472.CAN-09-1970

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Lee LJ, Papadopoli D, Jewer M, Del Rincon S, Topisirovic I, Lawrence MG, et al. Cancer plasticity: the role of mRNA translation. Trends Cancer. (2021) 7:134–45. doi: 10.1016/j.trecan.2020.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Li L, Halpert G, Lerner MG, Hu H, Dimitrion P, Weiss MJ, et al. Protein synthesis inhibitor omacetaxine is effective against hepatocellular carcinoma. JCI Insight. (2021) 6:e138197. doi: 10.1172/jci.insight.138197

CrossRef Full Text | Google Scholar

57. Howard TP, Oberlick EM, Rees MG, Arnoff TE, Pham M-T, Brenan L, et al. Rhabdoid tumors are sensitive to the protein-translation inhibitor homoharringtonine. Clin Cancer Res. (2020) 26:4995–5006. doi: 10.1158/1078-0432.CCR-19-2717

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Zhang J, Kim J, Alexander A, Cai S, Tripathi DN, Dere R, et al. A tuberous sclerosis complex signalling node at the peroxisome regulates mTORC1 and autophagy in response to ROS. Nat Cell Biol. (2013) 15:1186–96. doi: 10.1038/ncb2822

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Chen L, Xu B, Liu L, Luo Y, Yin J, Zhou H, et al. Hydrogen peroxide inhibits mTOR signaling by activation of AMPKalpha leading to apoptosis of neuronal cells. Lab Invest. (2010) 90:762–73. doi: 10.1038/labinvest.2010.36

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Zhang S, Zhao Y, Wang X, Qi C, Tian J, Zou Z. Synergistic lethality between auranofin-induced oxidative DNA damage and ATR inhibition in cancer cells. Life Sci. (2023) 332:122131. doi: 10.1016/j.lfs.2023.122131

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: reactive oxygen species, protein synthesis, auranofin, 4E-BP1, RRM2, plk1

Citation: Haight JA, Koppenhafer SL, Geary EL and Gordon DJ (2024) Auranofin and reactive oxygen species inhibit protein synthesis and regulate the level of the PLK1 protein in Ewing sarcoma cells. Front. Oncol. 14:1394653. doi: 10.3389/fonc.2024.1394653

Received: 01 March 2024; Accepted: 30 May 2024;
Published: 12 June 2024.

Edited by:

Carole Sourbier, United States Food and Drug Administration, United States

Reviewed by:

Rosa M. Andrade, University of California, Irvine, United States
Amit Kumar, Virginia Commonwealth University, United States
Mikael Molin, Chalmers University of Technology, Sweden

Copyright © 2024 Haight, Koppenhafer, Geary and Gordon. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: David J. Gordon, david-j-gordon@uiowa.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.