Zinc finger domain of p62/SQSTM1 is involved in the necroptosis of human cisplatin‑resistant ovarian cancer cells treated with sulfasalazine

  • Authors:
    • Nannan Liu
    • Shanshan Liu
    • Xueshuang Zhang
    • Wenzhu Tian
    • Heqiang Jia
    • Xin Ye
    • Xiaoyu Yan
    • Chunyan Yu
    • Huimei Yu
  • View Affiliations

  • Published online on: September 3, 2024     https://doi.org/10.3892/ol.2024.14662
  • Article Number: 529
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cisplatin resistance in ovarian cancer cells is mainly apoptosis resistant. Although other types of programmed cell death are highly involved in chemoresistance, which type can overcome cisplatin resistance remains unclear. The present study observed that cisplatin‑sensitive SKOV3 cells and cisplatin‑resistant SKOV3/DDP cells had different levels of sensitivity to sulfasalazine (SAS). The present study aimed to investigate the effect of SAS on necroptosis under the same inhibition rate in these two types of cells. Necroptosis inhibitor Necrostatin‑1 (Nec‑1) attenuated SAS‑induced SKOV3/DDP cytotoxicity. SAS decreased SKOV3/DDP cells survival rate, accompanied by decreased cell adhesion and spreading. SAS treatment activated necrosome formation in SKOV3/DDP cells, suggesting the possibility of necroptosis. p62/sequestosome‑1 (SQSTM1) protein expression levels were also increased over the same time period. The transfection of small interfering (si)‑p62 could decrease the ratios of phosphorylated (p)‑receptor‑interacting serine/threonine kinase 1 (RIP1)/RIP1, p‑receptor‑interacting serine/threonine kinase 3 (RIP3)/RIP3 and p‑mixed lineage kinase domain‑like protein (MLKL)/MLKL proteins in SKOV3/DDP cells. Under the si‑p62 condition, there was no increase in the rate of cell survival in Nec‑1 and SAS combination group compared with SAS. The zinc finger domain deletion of p62/SQSTM1 effectively decreased the expression levels of necroptosis‑related p‑proteins. Collectively, certain drugs were able to induce necroptosis in SKOV3/DDP, while p62/RIP1/RIP3/MLKL was associated with the induction of necroptosis and with increasing the sensitivity of cisplatin‑resistant ovarian cancer cells, which provided evidence for potential as a therapeutic target for overcoming resistance.

Introduction

It has been reported that in addition to apoptosis, other types of programmed cell death including ferroptosis, necroptosis and pyroptosis are highly related to chemoresistance (1). Inhibition of necroptosis by application of receptor-interacting serine/threonine kinase 1 (RIP1) activity inhibitor Necrostatin-1 (Nec-1) or knockdown of receptor-interacting serine/threonine kinase 3 (RIP3) significantly inhibits the cisplatin-induced loss of cell viability in apoptosis-resistant esophageal squamous cell carcinoma cells (2), suggesting that induction of necroptosis can increase the sensitivity of chemotherapeutic agents.

Reactive oxygen species (ROS) are considered to be a driving force for necroptosis (35). ROS contributes to increasing necroptosis in NF-κB-deficient cells induced by TNFα by detecting phosphorylated (p)-RIP1, RIP3 and mixed-lineage kinase domain-like (MLKL) (6). ROS production upon TNFα/BV6 treatment is involved in promoting the assembly of the RIP1/RIP3 necrosome in FADD-deficient Jurkat cells by immunoprecipitation of RIP1/RIP3 (4). Sulfasalazine (SAS), a potent cystine/glutamate transporter cystine-glutamate exchange (xCT) inhibitor that plays an important role in maintaining glutathione levels, impairs the ROS defense system and increases the therapeutic efficacy of anticancer therapies (79). Based on the previous results of a close correlation between necroptosis and ROS, a hypothesis of the present study was that SAS might be associated to the induction of necroptosis.

P62/sequestosome-1 (SQSTM1) is a multi-domain adapter protein that is highly expressed in various tumor cells, and it binds to multiple signaling molecules (10,11). However, the results of previous studies highlight that p62/SQSTM1 binds to RIP1 to assemble RIP3/MLKL on the autophagosome membrane in mouse prostate cells lacking Map3k7, suggesting that p62/SQSTM1 not only plays a role as an autophagy adaptor to degrade cellular material, but also may induce necroptosis by recruiting RIP1 (12,13). A previous study has indicated that deletion of the zinc finger (ZZ) domain of p62/SQSTM1 prevents the binding of RIP1 to p62/SQSTM1, and therefore inhibits the activation of the NF-κB signaling pathway (14). P62/SQSTM1 also exhibits pro-death functions through interaction with caspase-8, which plays a role in the progression of ovarian cancer (15). Notably, RIP1 activates NF-κB to promote cell survival, and binds to caspase-8 or RIP3 to play a role in the regulation of cell apoptosis and necroptosis (16). Thus the mechanism of necroptosis in SKOV3/DDP cells treated with SAS through p62/SQSTM1 remains to be elucidated.

The present study aimed to determine the effects of SAS on cisplatin-resistant ovarian cancer SKOV3/DDP cells. Another aim was to demonstrate that p62/SQSTM1 regulated the necroptosis of cisplatin-resistant ovarian cancer SKOV3/DDP cells treated with SAS, providing evidence for p62/SQSTM1 as a potential therapeutic target to overcome drug resistance.

Materials and methods

Reagents and antibodies

SAS, necroptosis inhibitor Nec-1 and 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) were purchased from Sigma-Aldrich; Merck KGaA. ViaFect™ transfection reagent was purchased from Promega Corporation. Anti-p62/SQSTM1 [cat. no. 66184-1-Ig; 1:5,000 for western blotting (WB)], anti-RIP1 (cat. no. 17519-1-AP; 1:500 for WB) and anti-β-actin (cat. no. 60008-1-Ig; 1:5,000 for WB) were purchased from ProteinTech Group, Inc. Anti-p-RIP1 (cat. no. 31122; 1:1,000 for WB) was purchased from Cell Signaling Technology, Inc. Anti-RIP3 (cat. no. ab305054; 1:1,000 for WB), anti-p-RIP3 (cat. no. ab222320; 1:2,000 for WB), anti-MLKL (cat. no. ab196436; 1:1,000 for WB) and anti-p-MLKL (cat. no. ab279863; 1:1,000 for WB) were purchased from Abcam.

Cell lines and cell culture

Cisplatin-sensitive HOCC SKOV3 cell lines were purchased from The Cell Bank of Type Culture Collection of The Chinese Academy of Sciences (cat. no. 1101HUM-PUMCO00027). SKOV3/DDP were added to the medium containing 0.02 µg/ml cisplatin, was gradually induced by increasing the concentration of cisplatin, and kept in medium containing 0.2 µg/ml cisplatin to maintain resistance. All cells were cultured in RPMI-1640 culture medium (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% fetal bovine serum (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C in a 5% CO2 incubator.

Cell transfection

p62/SQSTM1-small interfering (si)RNA (si-p62.1, si-p62.2 and si-p62.3) and non-targeting siRNA (scrambled control) were obtained from Shanghai GeneChem Co., Ltd. The target sequences for p62-siRNA were as follows: Si-p62.1 forward, 5′-CAGATGGAGTCGGATAACT-3′, and reverse, 5′-AGTTATCCGACTCCATCTG-3′; si-p62.2 forward, 5′-GTGACGAGGAATTGACAAT-3′, and reverse, 5′-ATTGTCAATTCCTCGTCAC-3′; si-p62.3 forward, 5′-GACATCTTCCGAATCTACA-3′, and reverse, 5′-TGTAGATTCGGAAGATGTC-3′; and scramble control sequences were forward, 5′-TTCTCCGAACGTGTCACGT-3′, and reverse, 5′-ACGTGACACGTTCGGAGAA-3′. The pcDNA3.1-∆ZZ-p62 plasmid (with a truncated mutation in the ZZ domain) and empty pcDNA3.1 vector (negative control; NC) were constructed by Sangon Biotech Co., Ltd. In total, 25×104 cells were plated into 6-well plates and cultured for 36 h. Following culturing, cells were transfected with 2 µg siRNA or plasmids per 6-well plate and 200 µl of total ViaFect™ transfection reagent complex at 37°C in a 5% CO2 incubator. Subsequent experimentation was carried out within 72 h.

Cell viability assay

In total, 8×103 cells/per well were seeded in 96-well plates and cultured for 36 h at 37°C with 5% CO2. Cells were treated with 0.5, 1.0, 2.0, 4.0 and 8.0 mM of SAS or 2.5, 5, 10, 25 and 50 µM of Nec-1. Cell viability was assessed using an MTT assay which dissolved the purple formazan by DMSO and measured at a wavelength of 570 nm using a microplate reader (Molecular Devices, LLC).

Real-time labeled cell function analysis (RTCA)

Cell suspensions were added to 96-well plates at a concentration of 1.5×104 cells in 100 µl medium per well. The 96-well plates were placed into the RTCA connection instrument (ACEA Bioscience, Inc.; Agilent), and cells were incubated at 37°C in 5% CO2 according to the manufacturer's protocol. When cells reached the logarithmic growth phase, the program was paused, the 96-well plate was removed and the original solution was discarded. Subsequently, 0.5 mM SAS and 10 µM Nec-1 were added to SKOV3 wells, and 2.0 mM SAS and 10 µM Nec-1 were added to SKOV3/DDP wells and the 96-well plate was returned to the RTCA connection instrument. The real-time growth of cells was observed and the program was completed when the pre-planned program ended or when cell proliferation met the requirements. Results were analyzed using the RTCA Data Analysis Software (version, 1.0; ACEA Bioscience Inc.).

Western blotting

Total protein was extracted from cells using RIPA lysis buffer (Beyotime Institute of Biotechnology) supplemented with protease and phosphatase inhibitors (2 mM AEBSF, 0.3 µM Aprotinin, 0.13 mM Bestatin, 0.014 mM E64 and 0.01 mM Leupeptin). Cells were centrifuged at 1,000 × g for 15 min at 4°C. The protein determination method was BCA assay and 30 µg protein per lane was separated using 10% SDS-PAGE. Separated proteins were subsequently transferred onto a PVDF membrane and blocked with 5% skimmed milk for 90 min at room temperature. Membranes were incubated with primary antibodies at 4°C overnight. Following primary incubation, membranes were incubated with HRP-conjugated secondary antibodies (cat. nos. SA00001-1 and SA00001-1; 1:2,000; ProteinTech Group, Inc.) for 2 h at room temperature. Protein bands were visualized using ECL reagent (Thermo Fisher Scientific, Inc.) and protein expression was quantified using a Syngene Bio Imaging system version 4.3.17 (Synoptics).

Immunoprecipitation assay

Cells were lysed in 200 µl NP40 lysis buffer (Beyotime Institute of Biotechnology) supplemented with protease inhibitors (8 µM Aprotinin, 0.5 mM Bestatin, 0.15 mM E64 and 0.2 mM Leupeptin) and were centrifuged at 12,000 × g and 4°C for 5 min. Subsequently, 30 µl of each cell supernatant were conducted as input. Equal amounts 1 mg of each cell supernatants were immunoprecipitated with 2 µg of the anti-RIP3 primary antibody overnight at 4°C. In total, 25 µl of protein A and G agarose (Beyotime Institute of Biotechnology) was used in each sample overnight at 4°C. The supernatants were isolated at 1,000 × g and 4°C for 5 min. Beads were washed three times with 1 ml PBS buffer, the magnetic were centrifugated at 1,000 × g and 4°C for 1 min, and eluted proteins were analyzed using anti-RIP1 primary antibody by western blotting.

Statistical analysis

Data are presented as the mean ± SEM and experimental repeats performed three times. Statistical analysis was performed using SPSS statistical software (version, 20.0; IBM Corp.). Comparisons between groups were conducted using one-way ANOVA followed by a post hoc test. Dunnett's t and Dunnett's T3 tests were used for comparisons using the same control group. LSD tests were used for comparisons between experimental, control and SAS groups. P<0.05 was considered to indicate a statistically significant difference.

Results

SAS treatment activates the necroptosis pathway in SKOV3/DDP cells

The survival rates of SKOV3 and SKOV3/DDP cells significantly decreased in a dose-dependent manner as the concentration of SAS increased, compared with the control group (Fig. 1A and B). Compared with the negative control SKOV3 cells, SKOV3/DDP cells have low sensitivity to SAS. The survival rate for SKOV3 cells treated with 0.5 mM SAS and 2 mM SAS were 81.9±1.4 and 69.3±2.43% respectively. The survival rate for SKOV3/DDP treated with 0.5 mM SAS and 2 mM SAS were 88.9±1.1 and 80.3±1.5% respectively. To observe the difference in cell death, in subsequent experiments, 0.5 mM SAS was selected to treat SKOV3 cells, and 2 mM SAS was selected to treat SKOV3/DDP cells with the same cell survival rate.

Notably, it has been reported that Nec-1, a RIP1-specific inhibitor, can block necroptosis (17). The present study tested the effect of Nec-1 on SAS-induced death in SKOV3/DDP cells. The present study intended to observe their cell survival rate, adhesion and spreading and cell morphology when treated with a combination of SAS with Nec-1. Results of the MTT assay demonstrated that, compared with the SAS group, a combination of Nec-1 and SAS did not increase the survival rate of SKOV3 cells, and a combination of Nec-1 and 5, 10, 25 µM and 2 mM SAS increased the survival rate of SKOV3/DDP cells (Fig. 1C and D). The cell survival rate was not altered in groups treated with different concentrations of Nec-1. The survival curve of cell index in RTCA indicates the cell number, and the slope of cell index indicates the adhesion and spreading, as well as cell morphology of cells. Compared with the SAS group, the survival curve of SKOV3 cells was low in the 10 µM Nec-1 and SAS combination group (Fig. 1E). In addition, the survival curve of SKOV3/DDP cells in the 10 µM Nec-1 and SAS combination group was high compared with the SAS group (Fig. 1F). Results of the RTCA demonstrated that in SKOV3 cells, the slope of the curve was decreased in the SAS group compared with the control. However, there was no significant difference between the slope of the curve in the SAS group, and that in the 10 µM Nec-1 and SAS combination group (Fig. 1G). In SKOV3/DDP cells, the slope of the curve was decreased in the SAS group, and showed a negative increase compared with the control. Moreover, the slope of the curve was increased in the 10 µM Nec-1 and SAS combination group compared with the SAS group (Fig. 1H). By contrast, the cell index changes between the Control and SAS groups were 4.7 and 3.7 in SKOV3 cells, respectively, and 4.0 and 0.5 in SKOV3/DDP cells respectively, indicating the adhesion and spreading of SKOV3/DDP cells decreased following SAS treatment in contrast to SKOV3 cells under the same cell survival rate (~80%).

SAS treatment activates necrosome formation in SKOV3/DDP cells

Expression levels of marker proteins associated with the necroptosis pathway were investigated in the present study. The expressions level of necroptosis-associated proteins; namely, p-RIP1, RIP3 and MLKL were significantly increased in SKOV3/DDP cells following SAS treatment for 12 h. On the other hand, in SKOV3 cells treated with SAS, the necroptosis-associated proteins p-RIP1, RIP3 and MLKL were not significantly altered at each observed time point (Fig. 2A-C). Notably, the complex formed following the phosphorylation of MLKL and the combination of RIP1 with RIP3 is the core component of necroptosis. Therefore, the SAS-induced precipitation of RIP3 was investigated in human ovarian epithelial carcinoma cells. Results of the co-immunoprecipitation analysis revealed that the expression of RIP1 and RIP3 complexes were increased in SKOV3/DDP cells compared with SKOV3 cells (Fig. 2D).

P62/SQSTM1 adaptor plays a role in SAS-induced necroptosis in SKOV3/DDP cells

Notably, p62/SQSTM1 protein expression levels were increased in SKOV3/DDP cells, compared with SKOV3 cells (18). During the SAS-induced death of SKOV3/DDP cells, p62/SQSTM1 expression was significantly increased following 12 h of SAS treatment compared with 0 h of treatment. These results are consistent with the increased expression of necroptosis marker proteins observed at the same time point. Results of the present study demonstrated that p62/SQSTM1 protein expression was minimal during the death of SKOV3 cells treated with SAS (Fig. 3A-C). RIP1 interacts directly with p62/SQSTM1 (19), suggesting a mechanism that by which p62/SQSTM1 could play a role in SAS-induced necroptosis. To test this, three si-p62 plasmids were used in transfection experiments, and si-p62.3 was selected for subsequent studies due to the highest level of transfection efficiency (Fig. 3D and E). Then, the present study immunoblotted necroptosis-related proteins after treatment with si-p62/SQSTM1 and found the ratios of p-RIP1/RIP1, p-RIP3/RIP3 and p-MLKL/MLKL proteins were decreased (Fig. 3F and G). Moreover, in contrast to the si-scramble group, which increased the rate of SKOV3/DDP cell survival after treatment with SAS and Nec-1 compared with SAS alone, following transfection with si-p62, there was no change in the rate of SKOV3/DDP cell survival in the 10 µM Nec-1 and SAS combination group compared with the SAS group (Fig. 3H).

Figure 3.

P62/SQSTM1 plays a role in SAS-induced necroptosis in SKOV3/DDP cells. (A) p62/SQSTM1 protein expression in SKOV3 and SKOV3/DDP cells induced by SAS at different time points. Relative protein expression levels of (B) SKOV3 and (C) SKOV3/DDP cells. Protein expression was normalized to the intensity of β-actin and results are expressed relative to the value of the control group. *P<0.05 vs. 0 h group. Values are displayed as the mean ± SEM (n=3). (D) p62/SQSTM1 protein expression in SKOV3/DDP cells following transfection with siRNA and (E) relative protein expression levels. Protein expression was normalized to the intensity of β-actin and results are expressed relative to the value of the scrambled group. *P<0.05 vs. Scramble group. Values are displayed as the mean ± SEM (n=3). (F) Protein expression levels of p-MLKL/MLKL, p-RIP3/RIP3 and p-RIP1/RIP1 in SKOV3/DDP cells transfected with siRNA against p62/SQSTM1 and the (G) relative protein expression levels. *P<0.05 vs. si-Scramble group. Values are displayed as the mean ± SEM (n=3). (H) Survival rate of SKOV3/DDP cells transfected with si-p62 following treatment with SAS and/or Nec-1. Results are expressed relative to the value of the control group. *P<0.05 vs. SAS group. Values are displayed as the mean ± SEM (n=3). SAS, sulfasalazine; SEM, standard error of the mean; siRNA, small interfering RNA; scrambled control, negative control; p-, phosphorylated; MLKL, mixed lineage kinase domain like protein; RIP, receptor interacting protein kinase.

ZZ domain of p62/SQSTM1 regulates necroptosis in SAS-induced SKOV3/DDP cells

The molecular structure of p62/SQSTM1 contains a ZZ domain that binds to RIP1. In the present study, p62/SQSTM1 protein with a ZZ domain deletion was overexpressed in ovarian carcinoma SKOV3 cells (Fig. 4A and B). Following the aforementioned overexpression, the ratios of p-RIP1/RIP1, p-RIP3/PIR3 and p-MLKL/MLKL were decreased (Fig. 4C and D).

Discussion

The modulation of necroptosis, a type of non-apoptotic programmed cell death, seems to be a promising approach to overcoming apoptotic drug resistance (20,21). To improve the response to acquired resistance, trastuzumab resistance is sensitized through the necroptosis pathway in breast cancer, and the survival rates of patients with breast cancer are affected (22). Results of a previous study demonstrated that SAS induces the apoptosis of cisplatin-tolerant small cell lung cancer cells (23), and may also induce apoptosis and autophagy in glioma U251 cells (24). In the present study, SKOV3/DDP cells exhibited a higher tolerance to SAS compared with SKOV3 cells. Results of the MTT assay and RTCA demonstrated that the response to Nec-1 also differed between SKOV3 and SKOV3/DDP cells. Notably, the survival rate of SKOV3/DDP cells was increased following treatment with a combination of SAS and Nec-1, while the survival rate of SKOV3 cells was not increased following treatment with a combination of SAS and Nec-1. In SKOV3/DDP cells, p- and non-p-proteins, RIP1 and RIP3, formed complexes, and these were increased following treatment with SAS. These results indicated that the necroptosis could be induced in SKOV3/DDP cells by SAS and increased the sensitivity of SKOV3/DDP cells to SAS.

The results of our previous study revealed that the multifunctional protein p62/SQSTM1 is expressed at low levels in SKOV3 cells, and expressed at high levels in SKOV3/DDP cells (18). Results of the present study demonstrated that the protein expression levels of p62/SQSTM1, RIP1 and RIP3 were increased in SKOV3/DDP cells following SAS treatment for 12 h. Although autophagy directly inhibits necroptosis through degradation of the kinase RIP1 (25), p62/SQSTM1 also co-immunoprecipitated with RIP1 and provided a scaffold for necroptosis signaling following treatment with tumor necrosis factor-related apoptosis-inducing ligand (19). The results of the present study also revealed that the expression levels of necroptosis-associated proteins, namely, p-RIP1, p-RIP3 and p-MLKL, were decreased following SAS treatment and p62/SQSTM1 knockdown in SKOV3/DDP cells. These results suggested that the p62/SQSTM1 protein may be required for RIP1 localization to autophagy machinery, leading to efficient assembly and activation of the necrosome in SAS-induced SKOV3/DDP cells. The ubiquitination and kinase activity of RIP1 dictates whether TNF signaling leads to activation of NF-κB; thus, protecting cells from apoptosis-mediated death (26), and triggering apoptosis (27) or necroptosis (28). In addition, TNF-α promotes necroptosis and apoptosis through various complexes involving RIP1, and the balance of protein activity controls caspase-dependent and -independent cell death (19). The results of the present study demonstrated that under transfection with si-p62, cell survival induced by Nec-1 and SAS was not increased compared with the survival of SKOV3/DDP cells following treatment with SAS. These results indicated that p62/SQSTM1 inhibition led to the activation of apoptosis, rather than necroptosis. The p62/RIP1/RIP3/MLKL pathway could be involved in necroptosis in SKOV3/DDP cells treated with SAS, and may increase the sensitivity of cisplatin-resistant ovarian cancer cells.

Notably, the multi-domain adapter protein, p62/SQSTM1, contains rich protein interaction sequences and domains, and plays certain functions in tumor progression depending on the interaction factors recruited. Mutations in the p62/SQSTM1 UBA domain increases the localization of hexokinase 2 on the mitochondria, and increases the phosphorylated ubiquitin form of Parkin (29). This stabilizes the mitophagy process in ovarian cancer A2780 cells, ultimately enabling cell survival. p62/SQSTM1 exhibits pro-death functions through the functional domain, ubiquitin-associated domain and LC3II-interacting region domain interacting with caspase-8 (15). In addition, the ZZ domain of p62/SQSTM1 binds to N-terminally arginylated proteins, and these are considered activators of the autophagic activity of p62/SQSTM1, rather than autophagy cargos (30). The results of a previous study demonstrated that SAS is a classic inhibitor of NF-κB (24). Notably, the ZZ domain of p62/SQSTM1 regulates K63-associated ubiquitination of RIP1, and loss of the ZZ domain from p62/SQSTM1 leads to poor proliferative capacity and high levels of apoptosis in SKOV3 cells. This ultimately leads to higher levels of cisplatin sensitivity (31). In the present study, the effects of necroptosis were determined using mutant p62/SQSTM1 molecules lacking the ZZ domain required for RIP1 interaction. Notably, the ZZ domain deletion of p62/SQSTM1 significantly inhibited p-RIP1, p-RIP3 and p-MLKL expression levels in SKOV3 cells.

The present study exhibits limitations, for example, there remains to be a lack of experiments on the colocalization of p62/SQSTM1 with the necrosome after the induction of SAS treatment, the interaction of ZZ with the necrosome and the two cells different response when treated with the same concentration of SAS. Moreover, in mammals, there are two autophagy receptors; namely, neighbor of BRCA1 gene 1 and p62/SQSTM1, and these both contain a ZZ domain. These results suggest that activation of the necrosome was regulated by the ZZ domain of p62/SQSTM1; however, the functional significance of the multi-specific binding ability of the aforementioned ZZ domains is yet to be fully established. Further studies should address these questions to fully clarify the function of p62/SQSTM1 by serving as a scaffold for necrosome in ovarian cancer.

Collectively, certain drugs were able to induce necroptosis in SKOV3/DDP, while p62/RIP1/RIP3/MLKL was associated with the induction of necroptosis and increased the sensitivity of cisplatin-resistant ovarian cancer cells. Thus, therapeutic targeting of necroptosis or its associated p62/RIP1/RIP3/MLKL pathway exhibits potential for restoring cisplatin sensitivity as a overcoming resistance therapeutic target.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from The Department of Science and Technology of Jilin Province (grant nos. YDZJ202101ZYTS090, 20230203073SF, 20220303003SF), The National Natural Science Foundation of China (grant no. 81541148) and The Graduate Innovation Program from Beihua University (grant no. 2022-014, 2024-022).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

NL, CY and HY contributed to design, plan and interpret the data. SL, XZ and WT contributed to acquisition of data. HJ, XYe and XYa carried out analysis and interpretation of data. NL and CY confirm the authenticity of all the raw data. All authors read and approved the final manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work (including the data) are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

MLKL

mixed lineage kinase domain-like protein

SQSTM1

sequestosome-1

RIP1

receptor-interacting serine/ threonine kinase 1

RIP3

receptor-interacting serine/threonine kinase 3

SAS

sulfasalazine

ZZ

zinc finger

References

1 

Zhang C and Liu N: Ferroptosis, necroptosis, and pyroptosis in the occurrence and development of ovarian cancer. Front Immunol. 13:9200592022. View Article : Google Scholar : PubMed/NCBI

2 

Xu Y, Lin Z, Zhao N, Zhou L, Liu F, Cichacz Z, Zhang L, Zhan Q and Zhao X: Receptor interactive protein kinase 3 promotes Cisplatin-triggered necrosis in apoptosis-resistant esophageal squamous cell carcinoma cells. PLoS One. 9:e1001272014. View Article : Google Scholar : PubMed/NCBI

3 

Zhang Y, Su SS, Zhao S, Yang Z, Zhong CQ, Chen X, Cai Q, Yang ZH, Huang D, Wu R and Han J: RIP1 autophosphorylation is promoted by mitochondrial ROS and is essential for RIP3 recruitment into necrosome. Nat Commun. 8:143292017. View Article : Google Scholar : PubMed/NCBI

4 

Schenk B and Fulda S: Reactive oxygen species regulate Smac mimetic/TNFα-induced necroptotic signaling and cell death. Oncogene. 34:5796–5806. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Hsu SK, Chang WT, Lin IL, Chen YF, Padalwar NB, Cheng KC, Teng YN, Wang CH and Chiu CC: The role of necroptosis in ROS-Mediated cancer therapies and its promising applications. Cancers (Basel). 12:21852020. View Article : Google Scholar : PubMed/NCBI

6 

Shindo R, Kakehashi H, Okumura K, Kumagai Y and Nakano H: Critical contribution of oxidative stress to TNFα-induced necroptosis downstream of RIPK1 activation. Biochem Biophys Res Commun. 436:212–216. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Narang VS, Pauletti GM, Gout PW, Buckley DJ and Buckley AR: Suppression of cystine uptake by sulfasalazine inhibits proliferation of human mammary carcinoma cells. Anticancer Res. 23:4571–4579. 2003.PubMed/NCBI

8 

Ji X, Qian J, Rahman SMJ, Siska PJ, Zou Y, Harris BK, Hoeksema MD, Trenary IA, Heidi C, Eisenberg R, et al: xCT (SLC7A11)-mediated metabolic reprogramming promotes non-small cell lung cancer progression. Oncogene. 37:5007–5019. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Guo W, Zhao Y, Zhang Z, Tan N, Zhao F, Ge C, Liang L, Jia D, Chen T, Yao M, et al: Disruption of xCT inhibits cell growth via the ROS/autophagy pathway in hepatocellular carcinoma. Cancer Lett. 312:55–61. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Jovanović L, Nikolić A, Dragičević S, Jović M and Janković R: Prognostic relevance of autophagy-related markers p62, LC3, and Beclin1 in ovarian cancer. Croat Med J. 63:453–460. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Adams O, Dislich B, Berezowska S, Schläfli AM, Seiler CA, Kröll D, Tschan MP and Langer R: Prognostic relevance of autophagy markers LC3B and p62 in esophageal adenocarcinomas. Oncotarget. 7:39241–39255. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Kharaziha P, Chioureas D, Baltatzis G, Fonseca P, Rodriguez P, Gogvadze V, Lennartsson L, Björklund AC, Zhivotovsky B, Grandér D, et al: Sorafenib-induced defective autophagy promotes cell death by necroptosis. Oncotarget. 6:37066–37082. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Goodall ML, Cramer SD and Thorburn A: Autophagy RIPs into cell death. Cell Cycle. 15:3014–3015. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Sanz L, Sanchez P, Lallena MJ, Diaz-Meco MT and Moscat J: The interaction of p62 with RIP links the atypical PKCs to NF-kappaB activation. EMBO J. 18:3044–3053. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Yan XY, Zhong XR, Yu SH, Zhang LC, Liu YN, Zhang Y, Sun LK and Su J: p62 aggregates mediated Caspase 8 activation is responsible for progression of ovarian cancer. J Cell Mol Med. 23:4030–4042. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Kaiser WJ, Daley-Bauer LP, Thapa RJ, Mandal P, Berger SB, Huang C, Sundararajan A, Guo H, Roback L, Speck SH, et al: RIP1 suppresses innate immune necrotic as well as apoptotic cell death during mammalian parturition. Proc Natl Acad Sci USA. 111:7753–7758. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on cell death 2018. Cell Death Differ. 25:486–541. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Yu H, Su J, Xu Y, Kang J, Li H, Zhang L, Yi H, Xiang X, Liu F and Sun L: p62/SQSTM1 involved in cisplatin resistance in human ovarian cancer cells by clearing ubiquitinated proteins. Eur J Cancer. 47:1585–1594. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Goodall ML, Fitzwalter BE, Zahedi S, Wu M, Rodriguez D, Mulcahy-Levy JM, Green DR, Morgan M, Cramer SD and Thorburn A: The Autophagy Machinery controls cell death switching between apoptosis and necroptosis. Dev Cell. 37:337–349. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Chen D, Yu J and Zhang L: Necroptosis: An alternative cell death program defending against cancer. Biochim Biophys Acta. 1865:228–236. 2016.PubMed/NCBI

21 

Basit F, Cristofanon S and Fulda S: Obatoclax (GX15-070) triggers necroptosis by promoting the assembly of the necrosome on autophagosomal membranes. Cell Death Differ. 20:1161–1173. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Dolapçi İB, Noyan S, Polat AY, Gürdal H and Dedeoğlu BG: miR-216b-5p promotes late apoptosis/necroptosis in trastuzumab-resistant SK-BR-3 cells. Turk J Biol. 47:199–207. 2023. View Article : Google Scholar : PubMed/NCBI

23 

Bagherpoor AJ, Shameem M, Luo X, Seelig D and Kassie F: Inhibition of lung adenocarcinoma by combinations of sulfasalazine (SAS) and disulfiram-copper (DSF-Cu) in cell line models and mice. Carcinogenesis. 44:291–303. 2023. View Article : Google Scholar : PubMed/NCBI

24 

Su J, Liu F, Xia M, Xu Y, Li X, Kang J, Li Y and Sun L: p62 participates in the inhibition of NF-κB signaling and apoptosis induced by sulfasalazine in human glioma U251 cells. Oncol Rep. 34:235–243. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Bray K, Mathew R, Lau A, Kamphorst JJ, Fan J, Chen J, Chen HY, Ghavami A, Stein M, DiPaola RS, et al: Autophagy suppresses RIP kinase-dependent necrosis enabling survival to mTOR inhibition. PLoS One. 7:e418312012. View Article : Google Scholar : PubMed/NCBI

26 

Li H, Kobayashi M, Blonska M, You Y and Lin X: Ubiquitination of RIP is required for tumor necrosis factor alpha-induced NF-kappaB activation. J Biol Chem. 281:13636–13643. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Tenev T, Bianchi K, Darding M, Broemer M, Langlais C, Wallberg F, Zachariou A, Lopez J, MacFarlane M, Cain K and Meier P: The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol Cell. 43:432–448. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL, Schneider P, Seed B and Tschopp J: Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol. 1:489–495. 2000. View Article : Google Scholar : PubMed/NCBI

29 

Yu S, Yan X, Tian R, Xu L, Zhao Y, Sun L and Su J: An experimentally induced mutation in the UBA Domain of p62 changes the sensitivity of cisplatin by Up-Regulating HK2 localisation on the mitochondria and increasing mitophagy in A2780 ovarian cancer cells. Int J Mol Sci. 22:39832021. View Article : Google Scholar : PubMed/NCBI

30 

Wang YY, Zhang J, Liu XM, Li Y, Sui J, Dong MQ, Ye K and Du LL: Molecular and structural mechanisms of ZZ domain-mediated cargo selection by Nbr1. EMBO J. 40:e1074972021. View Article : Google Scholar : PubMed/NCBI

31 

Yan XY, Zhang Y, Zhang JJ, Zhang LC, Liu YN, Wu Y, Xue YN, Lu SY, Su J and Sun LK: p62/SQSTM1 as an oncotarget mediates cisplatin resistance through activating RIP1-NF-κB pathway in human ovarian cancer cells. Cancer Sci. 108:1405–1413. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 28 Issue 5

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
Liu N, Liu S, Zhang X, Tian W, Jia H, Ye X, Yan X, Yu C and Yu H: Zinc finger domain of p62/SQSTM1 is involved in the necroptosis of human cisplatin‑resistant ovarian cancer cells treated with sulfasalazine. Oncol Lett 28: 529, 2024.
APA
Liu, N., Liu, S., Zhang, X., Tian, W., Jia, H., Ye, X. ... Yu, H. (2024). Zinc finger domain of p62/SQSTM1 is involved in the necroptosis of human cisplatin‑resistant ovarian cancer cells treated with sulfasalazine. Oncology Letters, 28, 529. https://doi.org/10.3892/ol.2024.14662
MLA
Liu, N., Liu, S., Zhang, X., Tian, W., Jia, H., Ye, X., Yan, X., Yu, C., Yu, H."Zinc finger domain of p62/SQSTM1 is involved in the necroptosis of human cisplatin‑resistant ovarian cancer cells treated with sulfasalazine". Oncology Letters 28.5 (2024): 529.
Chicago
Liu, N., Liu, S., Zhang, X., Tian, W., Jia, H., Ye, X., Yan, X., Yu, C., Yu, H."Zinc finger domain of p62/SQSTM1 is involved in the necroptosis of human cisplatin‑resistant ovarian cancer cells treated with sulfasalazine". Oncology Letters 28, no. 5 (2024): 529. https://doi.org/10.3892/ol.2024.14662