Low expression of MYCN promotes cisplatin resistance by suppressing cisplatin‑induced apoptosis in epithelial ovarian cancer

  • Authors:
    • Rao Yu
    • Hao Zhang
    • Rong Wang
    • Lin Xiao
  • View Affiliations

  • Published online on: October 11, 2022     https://doi.org/10.3892/ol.2022.13543
  • Article Number: 423
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Epithelial ovarian cancer (EOC) is the most common cause of gynecological cancer‑associated mortality. Cisplatin is one of the most effective chemotherapeutic drugs used in EOC; however, its use can lead to relapse due to cisplatin resistance. MYCN sensitizes neuroblastoma to undergo cisplatin‑induced apoptosis. However, to the best of our knowledge, there have been no studies to date on the association between MYCN and cisplatin resistance in EOC. Therefore, the present study assessed this association. Datasets from The Cancer Genome Atlas database were used. The overall survival (OS) of patients receiving platin‑based therapy was analyzed using Kaplan‑Meier Plotter software. RNA sequencing data of 300 patients with EOC were downloaded from cBioportal. The co‑expressed genes were subjected to ‘Kyoto Encyclopedia of Genes and Genomes’ analysis using DAVID software. For gene set enrichment analysis, the expression matrix was separated according to the median expression of MYCN, which was selected for hallmark gene set enrichment. Immunohistochemistry was used to assess MYCN expression in EOC tissue. Western blotting was used to evaluate MYCN, p53, Bax and Bcl‑2 protein expression levels in EOC cells. Cell viability and apoptosis were assessed using Cell Counting Kit-8 and flow cytometry, respectively. The results demonstrated that MYCN upregulation was associated with increased cisplatin sensitivity and prolonged OS of patients with EOC and patients receiving platin‑based therapy. Cisplatin downregulated MYCN expression in cisplatin‑sensitive, but not resistant, EOC cells. The genes co‑expressed with MYCN were primarily involved in pathways involved in ‘chemotherapeutic resistance’ and ‘apoptosis’. MYCN enriched the apoptosis and p53 signaling pathways in hallmark gene sets. Cells in which MYCN was knocked down demonstrated significantly increased cisplatin resistance; however, MYCN overexpression in cisplatin‑resistant cells restored cisplatin sensitivity. Collectively, the present study demonstrated that MYCN downregulation promoted cisplatin resistance by suppressing cisplatin‑induced apoptosis in EOC.

Introduction

Ovarian cancer (OC) is a gynecological cancer which contributes to a large number of deaths each year in industrialized countries; in 2020, it was estimated that there were 21,750 new cases and 13,940 associated deaths in the United States (1). It is estimated that 80% of patients with OC are eligible for the gold standard treatment of aggressive surgical debulking and platinum-based chemotherapy (2) and that 70% of these will develop platinum resistance and fatal disease following the long-term use of platinum (3). The outcomes of patients with platinum-resistant OC are poor, with a median overall survival (OS) rate of <12 months (4).

Resistance to platinum-based chemotherapy is a major clinical challenge in the treatment of OC, which results in a high mortality-to-incidence ratio (5). Epithelial OC (EOC) presents at an advanced stage globally and is the most common cause of gynecological cancer-associated mortality (6). In recent years, there have been notable achievements in the development of treatments of EOC, which have been validated by landmark clinical trials, such as a combination of surgery and systemic therapy, targeted therapy, chemotherapy and maximal surgical effort, of which the latter remains the mainstay (7). Therefore, overcoming platinum resistance is key to improving the prognosis of patients with EOC.

Numerous mechanisms and biological pathways underlying platinum resistance are being investigated. It has been reported that cisplatin functions by covalently binding to the DNA of tumor cells to form platinum-DNA adducts and induces cell apoptosis (8,9). One established mechanism for cisplatin resistance is evasion of cell apoptosis following long-term use of cisplatin (10), which results in resistance to cisplatin (11,12). Integrated genomic analysis of EOC reported one of the most common focal amplifications to be in the 8q24 region containing MYC (13). MYCC, MYCN and MYCL nuclear proteins are members of the Myc family that bind to and control ~15% of the human genome (14). It has been reported that downregulation of MYCN does not influence other MYC members such as MYCC and MYCL (15). MYC belongs to the Myc proto-oncogene family, which encodes basic helix-loop-helix/leucine zipper transcription factors, and functions in numerous types of human malignancy (14), including lung cancer (16) and mammary adenocarcinomas (17). Moreover, the Myc signaling pathway is one of the most commonly activated oncogenic pathways in human malignancy (18). Furthermore, previous studies have reported that Myc-mediated transcriptional networks are under tight regulation in normal cells and control numerous cellular processes, such as metabolic processes and cell proliferation, differentiation and apoptosis (14,19,20). As a member of the MYC family, MYCN is a type of short-lived transcription factor that is dysregulated in numerous types of human cancer (21), serves as a therapeutic target (22) and is associated with poor clinical outcome in multiple types of cancer (23). It has been reported that amplification of MYCN is associated with an aggressive phenotype and a poor prognosis in neuroblastoma; relapse of platinum-resistant neuroblastoma is the primary cause of mortality in patients with MYCN amplification (24). MYCN overexpression is significantly associated with poor outcomes in breast cancer (25). However, MYCN contributes to cisplatin sensitization in acute myelogenous leukemia (26).

The function of MYCN in EOC and chemotherapeutic resistance remains unclear. Therefore, the present study assessed the role of MYCN in EOC chemotherapeutic resistance.

Materials and methods

Bioinformatics analysis

The datasets used in the present study are available from The Cancer Genome Atlas (TCGA) database (tcga-data.nci.nih.gov/tcga) under TCGA-OV project (13). The analysis of the OS of patients receiving platin-based therapy was performed using Kaplan-Meier Plotter software (kmplot.com/analysis/index.php?p=service&cancer=ovar) with auto select best cut-off.

The GSE114206 dataset (27), which contains mRNA expression profiles of 12 patients with EOC (cisplatin-resistant patients, n=6; cisplatin-sensitive patients, n=6) was obtained from Gene Expression Omnibus (GEO) database (ncbi.nlm.nih.gov/geo/).

RNA sequencing data of 300 patients with OC (TCGA pan-cancer project) was downloaded from cBioportal (cbioportal.org/) (13). The co-expressed genes, assessed using Spearman's correlation analysis, were subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) (https://www.genome.jp/kegg/) analysis using DAVID software version 2.0 (https://david.ncifcrf.gov/). For gene set enrichment analysis (GSEA), the expression matrix was separated according to the median expression of MYCN. The expression matrix was used for Hallmark gene set enrichment using GSEA software (V.4.1.0) (gsea-msigdb.org/gsea/index.jsp).

Tissue samples

In total, 26 female patients with EOC who underwent primary surgery followed by cisplatin-based chemotherapy at the First Affiliated Hospital of Chongqing Medical University (Chongqing, China) between 2015 and 2019 were enrolled in the present study. None of these patients received radiotherapy before the surgery. The subtypes were assessed using histological examination performed by pathologists. A total of 22 patients (84.6%) were assessed as being serous and 4 (15.4%) were assessed as having mucinous EOC. Following primary chemotherapy, patients who relapsed within 6 months were assigned to the cisplatin-resistant group (n=13; mean age, 56 years; range, 37–70 years) and those who relapsed after 6 months or did not relapse were assigned to the cisplatin-sensitive group (n=13; mean age, 51 years; age range, 42–68 years). All patients provided written informed consent prior to inclusion in the study. The present study was approved by the Institutional Ethics Committee of The First Affiliated Hospital of Chongqing Medical University (Approval No. TFAHCQMU-2021-010). The characteristics of patients with EOC are presented in Table I.

Table I.

Characteristics of patients with epithelial ovarian cancer.

Table I.

Characteristics of patients with epithelial ovarian cancer.

Chemotherapy

CharacteristicSensitive, n=13.0Resistant, n=13P-value
Median age, years (range)51.0 (42.0-68.0)56.0 (37.0-70.0)0.198
Histology 0.296
  Serous (%)10.0 (76.9)12.0 (92.3)
  Mucinous (%)3.0 (23.1)1.0 (7.7)
FIGO stage 0.187
  I (%)2.0 (15.4)0.0 (0.0)
  II (%)3.0 (23.1)1.0 (7.7)
  III (%)6.0 (46.1)11.0 (84.6)
  IV (%)2.0 (15.4)1.0 (7.7)
Grade 0.500
  1/2 (%)5.0 (38.5%)6.0 (46.2%)
  3 (%)8.0 (61.5%)7.0 (53.8%)
Median CA125 at diagnosis, U/ml (range)474.0 (46.0-1,483.0)884.0 (28.5-3949)0.215

[i] FIGO, The International Federation of Gynecology and Obstetrics; CA125, cancer antigen 125.

Immunohistochemical analysis

Samples were fixed by 4% PFA solution, embedded by paraffin at 4°C overnight, sliced into 4 µm sections, and incubated at 60°C for 30 min. Following deparaffinization by xylene I and xylene II (each for 20 min) at room temperature, rehydration by alcohol series (100, 95%, 85%, and 75%), antigen retrieval by citric acid repair solution at oven for 5 min and endogenous peroxidase inhibition by 3%H2O2 at room temperature for 10 min, serous EOC sample slides were incubated with anti-MYCN antibody (1:100; cat. no. 10159-2-AP; ProteinTech Group, Inc.) at 4°C overnight. Slides were incubated with goat-anti-rabbit horseradish peroxidase-conjugated secondary antibodies (1:50; PR30009; ProteinTech Group, Inc.) for 1 h at room temperature, followed by assessment of peroxidase activity using diaminobenzidine for 10 min at room temperature. The tissue sections were visualized using a light microscope (40×). The statistical analysis was performed using histochemistry score (H-score) as previously reported (28).

Cells and cell culture

The human EOC SK-OV-3 cell line, which is commonly used in the study of cisplatin-resistant in serous EOC (29,30), was purchased from Jiangsu KeyGEN BioTECH Co., Ltd. The human EOC cisplatin-resistant SK-OV-3/DDP cell line was purchased from Shanghai Chuan Qiu Biotechnology Co., Ltd. Cells were cultured in RPMI-1640 medium (Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (PAN-Biotech GmbH) and 1% penicillin/streptomycin (Beyotime Institute of Biotechnology) in an incubator at 37°C with 5% CO2.

Cisplatin treatment

The SK-OV-3 and SK-OV-3/DDP cells were treated with a range of concentrations of cisplatin (0, 5 and 10 µM) for 24 h at 37°C.

Lentivirus construction and infection

The short hairpin (sh)RNA MYCN (LV-sh-MYCN; 5′-GCAGAAACCACAACATCCTGG-3′), negative control (LV-sh-NC; 5′-TTCTCCGAACGTGTCACGT-3′), MYCN-overexpressing (LV-MYCN) and NC lentivirus (LV-NC with a scrambled sequence) were purchased from Shanghai GenePharma Co., Ltd. The sequences were ligated into plko.1-puro plasmid. The lentivirus was packaged by transfection of 2 µg plko.1-puro, 1 µg psPAX2 and 2 µg pMD2.G into 293 cells for 24 h. The supernatant was collected for harvesting lentivirus particles. All lentiviruses contained GFP and puromycin resistance genes. At 72 h post-transduction, cells (MOI=10) were selected using puromycin (2 µg/ml) and maintained using puromycin (1 µg/ml) (Beyotime Institute of Biotechnology). The transfection efficiency in SK-OV-3 cells and SK-OV-3/DDP cells was assessed using western blotting.

Western blotting

SK-OV-3 and SK-OV-3/DDP cells were treated with cisplatin (0, 5 and 10 µM) for 24 h at 37°C and LV-sh-MYCN- SK-OV-3 cells and LV-MYCN SK-OV-3/DDP cells were treated with cisplatin (0 and 10 µM) for 24 h at 37°C, then harvested using PBS and lysed using RIPA buffer [Roche Diagnostics (Shanghai) Co., Ltd.] with protease inhibitor cocktail (Sigma-Aldrich; Merck KGaA). The protein concentration was evaluated using the BCA method (Beyotime Institute of Biotechnology). The extracted proteins (20 µg/lane) were separated using 10% (MYCN, p53 and β-actin) or 12% (Bax and Bcl2) SDS-PAGE, transferred to a PVDF membrane. Following blocking using 5% skimmed milk for 2 h at room temperature, PVDF membranes were incubated with primary antibodies overnight at 4°C and goat anti-rabbit (1:1,000; cat. no. 7074; Cell Signaling Technology, Inc.) and anti-mouse (1:1,000; cat. no. 7076; Cell Signaling Technology, Inc.) horseradish peroxidase-conjugated secondary antibodies (Cell Signaling Technology, Inc.) for 1 h at 37°C. The proteins were visualized using chemiluminescence (ECL Plus Western Blotting Detection system, Thermo Fisher). Primary antibodies were as follows: MYCN (1:1,000; cat. no. 10159-2-AP; ProteinTech Group, Inc.), MYCC (1:1,000; cat. no. 10828-1-AP; ProteinTech Group, Inc.), MYCL1 (1:1,000; cat no. PA5-109998; Thermo Fisher Scientific), β-actin (1:1,000; cat. no. 8457; Cell Signaling Technology, Inc.), p53 (1:1,000; cat. no. 2527; Cell Signaling Technology, Inc.), Bcl2 (1:1,000, cat. no. 15071; Cell Signaling Technology, Inc.) and Bax (1:1,000, cat. no. 5023; Cell Signaling Technology, Inc.). ImageJ software (version 1.8.0; National Institutes of Health) was used for densitometric analysis of the bands.

Cell Counting Kit-8 (CCK-8) assay

LV-sh-MYCN SK-OV-3 and LV-MYCN SK-OV-3/DDP cells (1×104 cells/well) were seeded into a 96-well plate and treated with 0, 2, 4, 6, 8, 10 and 15 µM cisplatin (Sigma-Aldrich; Merck KGaA) for 24 h at 37°C. Cell viability was determined using CCK-8 assay (Abcam), for which the cells were incubated at 37°C for 1 h. The absorbance was measured at 450 nm using an Infinite M200 PRO spectrophotometer (Tecan Group, Ltd.).

Apoptosis analysis

The number of apoptotic cells was quantified using Annexin V-FITC/propidium iodide (PI) staining. LV-sh-MYCN SK-OV-3 and LV-MYCN SK-OV-3/DDP cells (1×105 cells/well) were incubated in 6-well plates overnight at 37°C and treated with cisplatin (0 and 10 µM) for 24 h at 37°C. Following centrifugation at 2,000 × g for 3 min at room temperature, cells were suspended in 100 µl PBS, then mixed with 5 µl Annexin V-FITC (Beckman Coulter, Inc.) and 5 µl PI (Beckman Coulter, Inc.) prior to incubation for 15 min in the dark at room temperature. Cells at an early stage (FITC+/PI) and late stage (FITC+/PI+) were assessed as being apoptotic. The apoptotic cell percentage was assessed using a BD FACSCalibur™ flow cytometer (BD Biosciences). Data were analyzed using FlowJo software (version 7.6.3; FlowJo LLC).

Statistical analysis

Statistical analysis was performed using GraphPad Prism 8.0.1 (GraphPad Software, Inc.). Fisher's exact test was used for the analysis of histology, FIGO stage and grade. Unpaired Student's t test was used for analysis of age and CA125. H scores are presented as the median + interquartile range; all other data are from at least 3 independent experimental repeats, presented as the mean ± standard deviation. Comparisons between 2 groups were performed using Mann-Whitney test; for comparisons of ≥3 groups, Kruskal-Wallis followed by Dunn's post hoc test was used. P<0.05 was considered to indicate a statistically significant difference.

Results

High MYCN expression is positively associated with greater OS in patients receiving platin-based therapy

The data of 424 patients (low MYCN group, n=212; high MYCN group, n=212) obtained from TCGA database were assessed using OS analysis, which demonstrated that high expression of MYCN was associated with greater OS (Fig. 1A). As platin-based therapy was the first-line therapy approach for patients with OC, OS analysis was performed in patients receiving platin-based therapy, which indicated that high expression of MYCN was associated with a prolonged OS (Fig. 1B). The GSE114206 dataset, which contains mRNA expression profiles of 12 patients with EOC (cisplatin-resistant patients, n=6; cisplatin-sensitive patients, n=6), was obtained from the GEO database. A heatmap of the top 50 differentially expressed genes demonstrated that MYCN was increased in cisplatin-sensitive patients compared with cisplatin-resistant patients (Fig. 1C). The immunohistochemistry assessment of the tumor tissue collected in the present study demonstrated that MYCN protein expression in patients with cisplatin-sensitive EOC was significantly higher than that in cisplatin-resistant EOC (Fig. 1D and E).

Low MYCN protein expression levels are positively associated with cisplatin-resistance

Western blotting demonstrated that MYCN expression was significantly lower in SK-OV-3/DDP compared with SK-OV-3 cells (Fig. 2A). Moreover, cisplatin (0, 5 and 10 µM) significantly decreased MYCN protein expression in a dose-dependent manner in SK-OV-3 cells but not in SK-OV-3/DDP cells (Fig. 2B).

MYCN downregulation promotes cisplatin resistance in EOC cells

Western blotting demonstrated notable knockdown of MYCN by LV-sh-MYCN compared with LV-sh-NC in SK-OV-3 cells (Fig. 3A). The present study assessed the effect of MYCN knockdown on MYCC and MYCL protein expression levels, which demonstrated that LV-sh-MYCN did not affect MYCC and MYCL expression compared with LV-sh-NC in SK-OV-3 cells. CCK-8 assay demonstrated that cisplatin markedly decreased SK-OV-3 cell viability in a dose-dependent manner; furthermore, compared with LV-sh-NC, cell viability was significantly higher in the LV-sh-MYCN group following cisplatin treatment (Fig. 3B). Flow cytometry of SK-OV-3 cells demonstrated that cisplatin induced significant cell apoptosis in the LV-sh-NC group compared with the untreated LV-sh-NC group, but there was no significant difference between cisplatin treated group and cisplatin untreated group in the LV-sh-MYCN group. Furthermore, compared with LV-sh-NC, cisplatin-induced cell apoptosis was significantly decreased in the LV-sh-MYCN group. In groups without cisplatin treatment, there was significantly decreased cell apoptosis in the LV-sh-MYCN group compared with the LV-sh-NC group (Fig. 3C).

MYCN upregulation reverses cisplatin resistance of EOC cells

Western blotting demonstrated significant overexpression of MYCN in LV-MYCN SK-OV-3/DDP cells compared with LV-NC in SK-OV-3/DDP cells (Fig. 4A). CCK-8 assay in the SK-OV-3/DDP cells demonstrated that cisplatin markedly decreased cell viability in a dose-dependent manner; moreover, compared with LV-NC, cell viability was significantly decreased by cisplatin (≥4 µM) in the LV-MYCN group (Fig. 4B). Flow cytometry demonstrated that in the SK-OV-3/DDP cells, cisplatin induced significantly increased apoptosis in the LV-MYCN group compared with the LV-NC group; furthermore, compared with LV-NC, cisplatin-induced cell apoptosis was significantly increased in the LV-MYCN group. In the groups without cisplatin treatment, there was significantly increased cell apoptosis in the LV-MYCN group compared with the LV-NC group (Fig. 4C).

MYCN downregulation promotes cisplatin resistance by decreasing cisplatin-induced apoptosis

KEGG enrichment analysis of co-expressed genes of MYCN in the TCGA-OC dataset demonstrated that they were primarily involved in pathways that contributed to chemotherapeutic resistance and participated in cell apoptosis, such as ‘MAPK signaling pathway’, ‘PI3K/AKT signaling pathway’ and ‘p53 signaling pathway’ (Fig. 5A). Globally, GSEA demonstrated that MYCN was enriched in ‘apoptosis’ and ‘p53 pathway’ in the hallmark gene set (Fig. 5B-D).

To evaluate these results, p53, BAX and Bcl-2 protein expression levels were assessed using western blotting. In SK-OV-3 cells, treatment with cisplatin induced the significant upregulation of p53 protein expression levels and significantly increased the Bax/Bcl2 ratio in the LV-sh-NC group compared with the untreated group; however no significant difference was demonstrated in the LV-sh-MYCN group compared with the untreated group (Fig. 6A). Furthermore, compared with LV-sh-NC, cisplatin treatment induced significant upregulation of p53 protein expression levels and significantly decreased Bax/Bcl2 ratio in the LV-sh-MYCN group. Moreover, in the groups without cisplatin treatment, there was a significantly lower p53 protein expression and Bax/Bcl2 ratio in the LV-sh-MYCN group compared with LV-sh-NC group (Fig. 6A). However, in the SK-OV-3/DDP cells, cisplatin treatment induced significant upregulation of p53 protein expression levels and significantly increased Bax/Bcl2 ratio in the LV-MYCN group compared with the untreated group, however no significant difference was demonstrated in the LV-NC group compared with the untreated group. Furthermore, compared with LV-NC, cisplatin induced significant upregulation of p53 protein expression levels and significantly increased Bax/Bcl2 ratio in LV-MYCN group; moreover, in the groups without cisplatin treatment, there were significantly higher p53 protein expression levels and Bax/Bcl2 ratio in the LV-MYCN group compared with the LV-NC group (Fig. 6B).

Discussion

The present study was based on bioinformatics analysis, which demonstrated that patients with high MYCN expression had greater OS. High MYCN expression was associated with increased OS of patients receiving platin-based therapy; immunohistochemistry of tumor tissue collected in the present study demonstrated that there was significantly higher MYCN protein expression levels in cisplatin-sensitive EOC than cisplatin-resistant EOC. Therefore, it was hypothesized that MYCN was inhibited cisplatin resistance in EOC.

A previous study of expression profile of EOC reported that MYCN is overexpressed in C5 subtype tumors compared with three other molecular subtypes (C1, C2 and C4) of high-grade serous EOC (31), which indicated its aggressive role in high-grade serous EOC. Furthermore, MYCN overexpression has been reported to be predictive of an aggressive phenotype and poor prognosis in neuroblastoma (23), breast cancer (24) and spinal ependymoma (32); however, MYCN contributes to cisplatin sensitization in acute myelogenous leukemia (25). Consistent with this, the present study demonstrated that MYCN protein expression in SK-OV-3/DDP cells was significantly lower compared with that in SK-OV-3 cells and cisplatin significantly decreased MYCN protein expression levels in SK-OV-3 cells, but not in SK-OV-3/DDP cells. These results indicated that cisplatin functioned by suppressing expression of MYCN in EOC. However, the association between MYCN protein expression and cisplatin-induced cell behavior in EOC is unknown.

Apoptosis serves a key role in tissue homeostasis in response to numerous stimuli (33); decreased apoptosis associated with occurrence, development and drug resistance of tumors (34). Cisplatin functions by covalently binding to the DNA of tumor cells to form platinum-DNA adducts and induces cell apoptosis (8,9). Once the cisplatin-induced apoptotic pathway is blocked, tumor cells acquire resistance to the proapoptotic effects of cisplatin, thus decreasing its antitumor efficacy (35). In the present study, viability and apoptosis of SK-OV-3 and SK-OV-3/DDP cells following treatment with cisplatin was assessed, which demonstrated that the SK-OV-3 cells in which MYCN was knocked down exhibited a significantly decreased sensitivity to cisplatin-induced cell apoptosis compared with NC. Furthermore, SK-OV-3/DDP cells with MYCN overexpression exhibited a significantly increased sensitivity to cisplatin-induced cell apoptosis. Collectively, these results demonstrated that MYCN increased cisplatin-induced apoptosis and that apoptosis may be the primary mechanism by which MYCN inhibits cisplatin resistance in EOC. However, the molecules that mediate the role of MYCN in EOC remain to be elucidated.

The genes co-expressed with MYCN were primarily involved in pathways which contributed to chemotherapeutic resistance and participated in cell apoptosis, including ‘MAPK signaling pathway’, ‘PI3K/AKT signaling pathway’ and ‘p53 signaling pathway’. Globally, GSEA demonstrated that MYCN was enriched in ‘apoptosis’ and ‘p53 pathway’ in the hallmark gene sets. The tumor suppressor p53 is a transcription factor that regulates molecules in extrinsic (Bcl2 family) and intrinsic (mitochondrial) apoptotic pathways (3638). Balance of Bcl2 family members determines whether a cell undergoes apoptosis or survival (39). Cisplatin increases p53 levels and facilitates the apoptotic response in tumor cells (40); moreover, cisplatin activates Bax, decreases expression of Bcl2 and shifts the Bax/Bcl2 ratio in a pro-apoptotic direction in tumor cells (41). Furthermore, the emergence of p53 mutant cisplatin-resistant OC cells has been demonstrated following drug exposure (42) and patients with OC who have p53 mutations are more resistant to cisplatin-based therapy (43). In the present study, p53, Bax and Bcl-2 protein expression levels were assessed using western blotting, which demonstrated that following treatment with cisplatin, SK-OV-3 cells in which MYCN was knocked down exhibited significantly decreased p53 protein expression levels and Bax/Bcl2 ratio, whereas SK-OV-3/DDP cells with overexpressed MYCN exhibited significantly increased p53 protein expression levels and Bax/Bcl2 ratio. Therefore, it was hypothesized that MYCN affected cisplatin resistance by regulating p53 expression and ratio of Bax/Bcl2.

In conclusion, the present study suggested that MYCN served as a potential marker for cisplatin treatment in EOC. Specifically, the present study demonstrated that patients with high expression of MYCN were more sensitive to cisplatin, whereas patients with low expression of MYCN may be resistant to cisplatin. Furthermore, it may be hypothesized that the findings for cisplatin may be analogous to other chemotherapeutic drugs that lead to cell apoptosis. However, one weakness in current study is the use of only one EOC cell line and experiments should be replicated using another EOC cell line.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Chongqing (grant no. cstc2021jcyj-msxmX0120).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the GEO repository, accession number GSE114206.

Authors' contributions

RY, HZ and RW performed experiments and data analysis. LX conceived and supervised the study and wrote the manuscript. RY and LX confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All patients provided signed consent prior to their inclusion in the present study. The present study was approved by the Institutional Ethics Committee of the First Affiliated Hospital of Chongqing Medical University (approval No. TFAHCQMU-2021-010).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Markman M and Bookman MA: Second-line treatment of ovarian cancer. Oncologist. 5:26–35. 2000. View Article : Google Scholar : PubMed/NCBI

3 

Assis J, Pereira C, Nogueira A, Pereira D, Carreira R and Medeiros R: Genetic variants as ovarian cancer first-line treatment hallmarks: A systematic review and meta-analysis. Cancer Treat Rev. 61:35–52. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Marchetti C, De Felice F, Romito A, Iacobelli V, Sassu CM, Corrado G, Ricci C, Scambia G and Fagotti A: Chemotherapy resistance in epithelial ovarian cancer: Mechanisms and emerging treatments. Semin Cancer Biol. 77:144–166. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Khan MA, Vikramdeo KS, Sudan SK, Singh S, Wilhite A, Dasgupta S, Rocconi RP and Singh AP: Platinum-resistant ovarian cancer: From drug resistance mechanisms to liquid biopsy-based biomarkers for disease management. Semin Cancer Biol. 77:99–109. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Lheureux S, Gourley C, Vergote I and Oza AM: Epithelial ovarian cancer. Lancet. 393:1240–1253. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Kurnit KC, Fleming GF and Lengyel E: Updates and new options in advanced epithelial ovarian cancer treatment. Obstet Gynecol. 137:108–121. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Kelland L: The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer. 7:573–584. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Zhang J, Wei H, Liu X, Wang N, Qi Y, Zhang Y and Zhang S: Downregulation of phosphoglycerate dehydrogenase inhibits proliferation and enhances cisplatin sensitivity in cervical adenocarcinoma cells by regulating Bcl-2 and caspase-3. Cancer Biol Ther. 16:541–548. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Binju M, Amaya-Padilla MA, Wan G, Gunosewoyo H, Suryo Rahmanto Y and Yu Y: Therapeutic inducers of apoptosis in ovarian cancer. Cancers (Basel). 11:17862019. View Article : Google Scholar : PubMed/NCBI

11 

Kartalou M and Essigmann JM: Mechanisms of resistance to cisplatin. Mutat Res. 478:23–43. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Wernyj RP and Morin PJ: Molecular mechanisms of platinum resistance: Still searching for the Achilles' heel. Drug Resist Updat. 7:227–232. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Cancer Genome Altas Research Network, . Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Meyer N and Penn LZ: Reflecting on 25 years with MYC. Nat Rev Cancer. 8:976–990. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Neri F, Zippo A, Krepelova A, Cherubini A, Rocchigiani M and Oliviero S: Myc regulates the transcription of the PRC2 gene to control the expression of developmental genes in embryonic stem cells. Mol Cell Biol. 32:840–851. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Zajac-Kaye M: Myc oncogene: A key component in cell cycle regulation and its implication for lung cancer. Lung Cancer. 34 (Suppl 2):S43–S46. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Boxer RB, Jang JW, Sintasath L and Chodosh LA: Lack of sustained regression of c-MYC-induced mammary adenocarcinomas following brief or prolonged MYC inactivation. Cancer Cell. 6:577–586. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Bild AH, Yao G, Chang JT, Wang Q, Potti A, Chasse D, Joshi MB, Harpole D, Lancaster JM, Berchuck A, et al: Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature. 439:353–357. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Mo H, Vita M, Crespin M and Henriksson M: Myc overexpression enhances apoptosis induced by small molecules. Cell Cycle. 5:2191–2194. 2006. View Article : Google Scholar : PubMed/NCBI

20 

McMahon SB: MYC and the control of apoptosis. Cold Spring Harb Perspect Med. 4:a0144072014. View Article : Google Scholar : PubMed/NCBI

21 

Koach J, Holien JK, Massudi H, Carter DR, Ciampa OC, Herath M, Lim T, Seneviratne JA, Milazzo G, Murray JE, et al: Drugging MYCN oncogenic signaling through the MYCN-PA2G4 binding interface. Cancer Res. 79:5652–5667. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Beltran H: The N-myc oncogene: Maximizing its targets, regulation, and therapeutic potential. Mol Cancer Res. 12:815–822. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Jung M, Russell AJ, Liu B, George J, Liu PY, Liu T, DeFazio A, Bowtell DD, Oberthuer A, London WB, et al: A Myc activity signature predicts poor clinical outcomes in Myc-associated cancers. Cancer Res. 77:971–981. 2017. View Article : Google Scholar : PubMed/NCBI

24 

Nakagawara A, Li Y, Izumi H, Muramori K, Inada H and Nishi M: Neuroblastoma. Jpn J Clin Oncol. 48:214–241. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Mizukami Y, Nonomura A, Takizawa T, Noguchi M, Michigishi T, Nakamura S and Ishizaki T: N-myc protein expression in human breast carcinoma: Prognostic implications. Anticancer Res. 15:2899–2905. 1995.PubMed/NCBI

26 

Huang X, Qi L, Lu W, Li Z, Li W and Li F: MYCN contributes to the sensitization of acute myelogenous leukemia cells to cisplatin by targeting SRY-box transcription factor 4. Bioengineered. 2021. View Article : Google Scholar

27 

Veskimäe K, Scaravilli M, Niininen W, Karvonen H, Jaatinen S, Nykter M, Visakorpi T, Mäenpää J, Ungureanu D and Staff S: Expression analysis of platinum sensitive and resistant epithelial ovarian cancer patient samples reveals new candidates for targeted therapies. Transl Oncol. 11:1160–1170. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Schulz H, Kuhn C, Hofmann S, Mayr D, Mahner S, Jeschke U and Schmoeckel E: Overall survival of ovarian cancer patients is determined by expression of galectins-8 and −9. Int J Mol Sci. 19:3232018. View Article : Google Scholar : PubMed/NCBI

29 

Zhu H, Zou X, Lin S, Hu X and Gao J: Effects of naringin on reversing cisplatin resistance and the Wnt/β-catenin pathway in human ovarian cancer SKOV3/CDDP cells. J Int Med Res. 48:3000605198878692020. View Article : Google Scholar : PubMed/NCBI

30 

Zou GP, Yu CX, Shi SL, Li QG, Wang XH, Qu XH, Yang ZJ, Yao WR, Yan DD, Jiang LP, et al: Mitochondrial dynamics mediated by DRP1 and MFN2 contributes to cisplatin chemoresistance in human ovarian cancer SKOV3 cells. J Cancer. 12:7358–7373. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Helland Å, Anglesio MS, George J, Cowin PA, Johnstone CN, House CM, Sheppard KE, Etemadmoghadam D, Melnyk N, Rustgi AK, et al: Deregulation of MYCN, LIN28B and LET7 in a molecular subtype of aggressive high-grade serous ovarian cancers. PLoS One. 6:e180642011. View Article : Google Scholar : PubMed/NCBI

32 

Ghasemi DR, Sill M, Okonechnikov K, Korshunov A, Yip S, Schutz PW, Scheie D, Kruse A, Harter PN, Kastelan M, et al: MYCN amplification drives an aggressive form of spinal ependymoma. Acta Neuropathol. 138:1075–1089. 2019. View Article : Google Scholar : PubMed/NCBI

33 

de Oliveira CB, Comunello LN, Maciel ES, Giubel SR, Bruno AN, Chiela EC, Lenz G, Gnoatto SC, Buffon A and Gosmann G: The inhibitory effects of phenolic and terpenoid compounds from Baccharis trimera in Siha cells: Differences in their activity and mechanism of action. Molecules. 18:11022–11032. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Plati J, Bucur O and Khosravi-Far R: Dysregulation of apoptotic signaling in cancer: Molecular mechanisms and therapeutic opportunities. J Cell Biochem. 104:1124–1149. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Wang H, Luo Y, Qiao T, Wu Z and Huang Z: Luteolin sensitizes the antitumor effect of cisplatin in drug-resistant ovarian cancer via induction of apoptosis and inhibition of cell migration and invasion. J Ovarian Res. 11:932018. View Article : Google Scholar : PubMed/NCBI

36 

Green DR and Kroemer G: The pathophysiology of mitochondrial cell death. Science. 305:626–629. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Czabotar PE, Lessene G, Strasser A and Adams JM: Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Vousden KH: p53: Death star. Cell. 103:691–694. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Pistritto G, Trisciuoglio D, Ceci C, Garufi A and D'Orazi G: Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY). 8:603–619. 2016. View Article : Google Scholar : PubMed/NCBI

40 

An SH, Kang JH, Kim DH and Lee MS: Vitamin C increases the apoptosis via up-regulation p53 during cisplatin treatment in human colon cancer cells. BMB Rep. 44:211–216. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Li X, Mu J, Lin Y, Zhao J and Meng X: Combination of cyanidin-3-O-glucoside and cisplatin induces oxidative stress and apoptosis in HeLa cells by reducing activity of endogenous antioxidants, increasing bax/bcl-2 mRNA expression ratio, and downregulating Nrf2 expression. J Food Biochem. 45:e138062021. View Article : Google Scholar : PubMed/NCBI

42 

Righetti SC, Perego P, Corna E, Pierotti MA and Zunino F: Emergence of p53 mutant cisplatin-resistant ovarian carcinoma cells following drug exposure: Spontaneously mutant selection. Cell Growth Differ. 10:473–478. 1999.PubMed/NCBI

43 

Xie X, Lozano G and Siddik ZH: Heterozygous p53(V172F) mutation in cisplatin-resistant human tumor cells promotes MDM4 recruitment and decreases stability and transactivity of p53. Oncogene. 35:4798–806. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2022
Volume 24 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu R, Zhang H, Wang R and Xiao L: Low expression of MYCN promotes cisplatin resistance by suppressing cisplatin‑induced apoptosis in epithelial ovarian cancer. Oncol Lett 24: 423, 2022
APA
Yu, R., Zhang, H., Wang, R., & Xiao, L. (2022). Low expression of MYCN promotes cisplatin resistance by suppressing cisplatin‑induced apoptosis in epithelial ovarian cancer. Oncology Letters, 24, 423. https://doi.org/10.3892/ol.2022.13543
MLA
Yu, R., Zhang, H., Wang, R., Xiao, L."Low expression of MYCN promotes cisplatin resistance by suppressing cisplatin‑induced apoptosis in epithelial ovarian cancer". Oncology Letters 24.6 (2022): 423.
Chicago
Yu, R., Zhang, H., Wang, R., Xiao, L."Low expression of MYCN promotes cisplatin resistance by suppressing cisplatin‑induced apoptosis in epithelial ovarian cancer". Oncology Letters 24, no. 6 (2022): 423. https://doi.org/10.3892/ol.2022.13543