TOPK inhibition accelerates oxidative stress‑induced granulosa cell apoptosis via the p53/SIRT1 axis

  • Authors:
    • Jung‑Hwan Park
    • Sang‑Ah Park
    • Young‑Ju Lee
    • Na‑Rae Joo
    • Jongdae Shin
    • Sang‑Muk Oh
  • View Affiliations

  • Published online on: August 27, 2020     https://doi.org/10.3892/ijmm.2020.4712
  • Pages: 1923-1937
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

It has been suggested that oxidative stress involving reactive oxygen species (ROS) induces granulosa cell apoptosis, leading to follicular atresia, and that T‑lymphokine‑activated killer cell‑originated protein kinase (TOPK) suppresses cancer cell apoptosis induced by several stimuli. However, it remains to be determined whether TOPK affects oxidative stress‑induced granulosa cell apoptosis. The present study demonstrates that TOPK inhibition increases human granulosa COV434 cell apoptosis induced by hydrogen peroxide (H2O2). Co‑treatment with the TOPK inhibitor, OTS514, in combination with H2O2 increased p53 acetylation and its expression, whereas it decreased Sirtuin 1 (SIRT1) expression, contributing to the promotion of apoptosis. In addition, the SIRT1 activator, resveratrol, or the SIRT1 inhibitor, Ex527, reduced or elevated H2O2‑induced COV434 cell apoptosis, respectively. Furthermore, the p53 inhibitor, Pifithrin‑μ, diminished the augmentation in poly(ADP‑ribose) polymerase (PARP) cleavage induced by OTS514 plus H2O2, while the Mdm2 antagonist, Nutlin 3, increased PARP cleavage. Moreover, OTS514 further decreased the SIRT1 transcriptional activity decreased by H2O2, but promoted the H2O2‑induced p53 or p21 transcriptional activity. Notably, the expression of exogenous p53 reduced SIRT1 transcriptional activity. Taken together, the findings of the present study demonstrate that TOPK inhibition promotes p53‑mediated granulosa cell apoptosis through SIRT1 downregulation in response to H2O2. Therefore, it can be concluded that TOPK suppresses H2O2‑induced apoptosis through the modulation of the p53/SIRT1 axis, suggesting a potential role of TOPK in the regulation of human granulosa cell apoptosis, leading to the promotion of abnormal follicular development.

Introduction

The ovaries are known to be unique and crucial female organs which reproduce generations of eggs and secrete sex hormones for the female secondary sexual characteristics. However, the number of ovarian follicles secreted during a lifetime is limited. Follicles consist of several cells, including oocytes, granulosa cells and theca cells. The oocyte is surrounded by layers of granulosa cells and forms a direct connection with granulosa cells. It has been suggested that granulosa cells not only provide nutrient to oocytes, but also protect oocytes from oxidative stress during follicle development (1,2), and that reactive oxygen species (ROS)-mediated oxidative stress results in granulosa cell apoptosis to promote abnormal follicular development (3).

It has been reported that sirtuin 1 (SIRT1), a well-known NAD+ dependent deacetylase (class III histone deacetylase), influences not only histone proteins, but also non-histone proteins, and regulates various biological processes involving aging, apoptosis or the stress response (4,5). In addition, SIRT1 is known to induce the phosphorylation of extracellular signal-regulated kinase (ERK) and inhibit the translocation of nuclear factor (NF)-κB into the nucleus, leading to suppression of granulosa cell apoptosis (6). It has been shown that various microRNAs (miRNAs or miRs) such as miRNA-590-3P or miRNA-494 suppress SIRT1 activity to promote apoptosis (7,8). It has been suggested that SIRT1 deacetylates lysine 382 of p53, thereby reducing p53 transcriptional activity and p53-mediated apoptosis, whereas p53 expression suppresses the transcription of SIRT1 gene induced by c-Myc (9-11).

T-LAK cell originated protein kinase (TOPK), a type of serine/threonine kinase, has been shown to be strongly expressed in various solid cancers, including colorectal cancer (12-14), lung cancer (15,16), gastric cancer (17), prostate cancer (18,19), ovarian carcinoma (20), nasopharyngeal carcinoma (21) and esophageal squamous cell carcinoma (22). TOPK consists of 322 amino acids and has a functional similarity with MKK3/6, which activates p38 (23,24). However, it has been reported that TOPK phosphorylates ERK in EGF-stimulated HCT116 colorectal cancer cells or T47D cells (25,26) and phosphorylates JNK1 in ras-induced cell transformation or UVB-mediated signaling (27). In addition, TOPK has been shown to contribute to an increased metastasis of breast cancer cells by modulating the expression of MMP9 through the regulation of NF-κB activity in LPS signaling (28). On the other hand, it has been reported that TOPK directly binds to the DNA-binding domain of p53 and suppresses p53 transcriptional activity (29,30). However, whether TOPK affects oxidative stress-induced granulosa cell apoptosis remains to be determined.

In the present study, it was first revealed that TOPK inhibition increased p53-mediated granulosa cell apoptosis through SIRT1 downregulation upon H2O2 treatment. It was demonstrated that p53 negatively regulated SIRT1 transcriptional activity in response to H2O2, which was aggravated by TOPK inhibition. It was also found that TOPK inhibition promoted p53 stability, leading to an increase in H2O2-induced granulosa cell apoptosis. These findings provide evidence that TOPK suppresses H2O2-induced granulosa cell apoptosis through the regulation of the p53/SIRT1 axis.

Materials and methods

Cell culture and reagents

Human granulosa COV434 cells were purchased from Sigma-Aldrich; Merck KGaA. HeLa cells were purchased from the American Type Culture Collection (ATCC) and maintained in DMEM supplemented with 10% FBS, 2 mM L-glutamine, and 1% penicillin and streptomycin (Thermo Fisher Scientific, Inc.). COV434 cells were maintained in high glucose Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum, 2 mM L-glutamine, 1% penicillin and streptomycin at 37°C in a CO2 incubator with 5% CO2. Rabbit anti-TOPK (ab75987), rabbit anti-SIRT1 (ab32441), mouse anti-p53 (ab26), rabbit anti-phospho-p53 (Ser-15) (ab1431) and rabbit anti-acetyl p53 (K382) (ab75754) antibodies were purchased from Abcam. Rabbit anti-cleaved poly(ADP-ribose) polymerase (PARP) antibody (#9541) was purchased from Cell Signaling Technology, Inc. Mouse anti-β-actin antibody (A1975) was purchased from Sigma-Aldrich. OTS514, Z-VAD-FMK were purchased from Selleckchem; 2′,7′-dichloroflurescin diacetate, Nutlin-3, Pifithrin-μ, resveratrol and Ex527 were purchased Sigma-Aldrich; Merck KGaA. Annexin V-FITC antibody and propidium iodide were purchased from Thermo Fisher Scientific, Inc. The luciferase assay kit was purchased from Promega Corporation.

Plasmids and transfection

The SIRT1 promoter linked luciferase reporter gene, SIRT1-Luc construct was generated. Briefly, an approximately 1 kb promoter region containing nucleotides -1 to -1,026 of the human SIRT1 gene was amplified using the Takara Ex Taq polymerase (Takara Bio, Inc.) and human genomic DNA (Promega Corporation) as a template. The amplified product was cloned into the NheI and XhoI sites of the pGL3-basic vector (cat. no. E1751, Promega Corporation). The specific primer sequences were as follows: Forward, 5′-CCGGCTAACCCATACTAGGCTTAAGG-3′ and reverse, 5′-CCGCTCGAGCTTCCAACTGCCTCTCTGGC-3′. p53-GFP was a gift from Geoff Wahl (Addgene plasmid #11770). The pGL2-p53 promoter-Luc construct was a gift from Wafik El-Deiry (Addgene plasmid #16292) and the pGL2-p21 promoter-Luc plasmid was a gift from Martin Walsh (addgene plasmid #33021). The pcDNA3.1 or pEGFP-N1 vector was purchased from Invitrogen; Thermo Fisher Scientific, Inc. (cat. no. V79020) or Clontech (cat. no. 6085-1), respectively, and the V5-TOPK construct was previously described (31). Briefly, total RNA from HeLa cells was prepared, and cDNA synthesis and PCR were performed using Superscript III reverse transcriptase (Invitrogen; Thermo Fisher Scientific, Inc.) and primers, 5′-CGCGGATCCCGATGGAAGGGATCAGTAATTT-3′ harboring the BamHI site (forward) and 5′-CGCTCGAGCGGGACATCTGTTTCCAGAGCTT-3′ harboring the XhoI site (reverse). The PCR product digested with BamHI/XhoI was inserted into the BamHI/XhoI sites of pcDNA6/V5-His ABC (cat. no. V22020, Invitrogen; Thermo Fisher Scientific, Inc.), generating the V5-TOPK construct. Non-target siRNA and p53 siRNA were purchased from Dharmacon. COV434 cells growing in 12-well plates were transfected with 1 µg of pcDNA3.1 or V5-TOPK, 2 µg of pGL3-SIRT1 promoter-Luc, 1 µg of pEGFP-N1, pGL2-p53 promoter-Luc or pGL2-p21 promoter-Luc plasmids or 30 nM of non-target or p53 siRNA using Lipofectamine 3000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to manufacturer's instructions. At 24 h following transfection, cells were lysed, and luciferase assay was performed using cell lysate.

Flow cytometric analysis

A total of 1×106 of COV434 cells growing on 60 mm dishes were pre-treated with inhibitors or activators, OTS514 (20 nM), Z-VAD-FMK (50 µM), resveratrol (20 µM), Ex527 (30 nM), Pifithrin-μ (1 µM), or Nutlin-3 (10 µM) for 2 h and H2O2 (0.1 mM) was then added for 24 h. Apoptosis analysis was performed using Annexin V-FITC and propidium iodide according to manufacturer's instructions (Thermo Fisher Scientific, Inc.), and data were acquired using a FACScalibur (BD Biosciences).

2′,7′-Dichloroflurescin diacetate assay

COV434 cells growing on 60 mm dishes at 80% confluency were pre-treated with OTS514 (20 nM) for 2 h, and incubated at 37°C with H2O2 (0.1 mM) for 24 h. The conditioned medium was removed, and fresh medium containing 10 µM H2DCF-DA was added. The cells were then incubated at 37°C for 30 min, and then washed and trypsinized. Cells were resuspended using conditioned medium. DCF fluorescence was measured using a FACScalibur (BD Biosciences).

TUNEL assay

A total of 2×106 of COV434 cells were seeded in 6-well plates and pre-treated with inhibitor or activator, OTS514 (20 nM), Z-VAD-FMK (50 µM), resveratrol (20 µM), Ex527 (30 nM), Pifithrin-µ (1 µM), or Nutlin-3 (10 µM), for 2 h, and then H2O2 (0.1 mM) was added for 24 h. Apoptosis was determined using the DeadEnd™ Fluorometric TUNEL System (G3250, Promega Corporation). The cells were fixed using 4% formaldehyde for 25 min at 4°C, and then permeabi-lized by 0.2% Triton X-100 for 5 min at room temperature. The cells were labeled using 50 µl of TdT reaction mix and incubated for 60 min at 37°C and then data analysis was performed using a laser-scanning confocal microscope Zeiss LSM 710 (Zeiss AG).

Western blot analysis

Briefly, cells were harvested and lysed using lysis buffer containing 0.5% Triton X-100, 1 mM EDTA, 50 mM Tris-HCl, pH 7.4 and 40 mM NaCl. Briefly, 30 µg of cell lysate was separated on 8 or 10% SDS-PAGE and transferred to nitrocellulose membranes (Bio-Rad Laboratories, Inc.). The membranes were blocked with 5% skim milk in Tris-buffered saline and Tween-20 (TBST) for 1 h and then incubated with anti-TOPK (1:5×106 in 5% Skim milk), p53 (1:1,000 in TBST), phospho-p53 (1:1,000 in TBST), acetyl-p53 (1:1,000 in 5% Skim milk), cleaved PARP (1:1,000 in TBST), SIRT1 (1:1,000 in TBST) and β-actin (1: 20,000 in 5% Skim milk) antibodies at 4°C for overnight. The membranes were washed 3 times for 5 min using TBST, and then incubated at room temperature with goat anti-rabbi IgG, polyclonal (1:4,000 in 5% Skim milk, ADI-SAB-300-J, Enzo Life Sciences) or goat anti-Mouse polyclonal (1:4,000 in 5% Skim milk, ADI-SAB-100-J, Enzo Life Sciences) antibodies for 2 h. After washing, the blots were analyzed with SuperSignal West Pico chemiluminescent substrate (Pierce; Thermo Fisher Scientific, Inc.) and X-ray film. Each protein level was determined using Image J software (ver.1.52a; National Institute of Health).

Luciferase assay

A total of 1×105 of COV434 cells were seeded in 12-well plates. After 24 h, 1 µg of SIRT1, p53 or p21 promoter-luciferase reporter constructs plus 0.5 µg of the pRL-SV40 gene were transfected into the COV434 cells using Lipofectamine 3000 reagent. At 24 h following transfection, the cells were pre-treated with OTS514 (20 nM), resveratrol (20 µM), Ex527 (30 nM), Pifithrin-µ (1 µM) or Nutlin-3 (10 µM) for 2 h, and then incubated at 37°C with 0.1 mM H2O2 for 24 h. Cells were harvested and lysed. Firefly and Renilla luciferase activities were measured using Dual-Luciferase® Reporter assay system (E1910, Promega Corporation).

Protein stability assay

COV434 cells were seeded in 6-well plates. After 24 h, the cells were pre-treated with OTS514 (20 nM) for 2 h, and then treated with H2O2 (0.1 mM) for 24 h at 37°C. 24 h after H2O2 treatment, cells were incubated with cycloheximide (10 µg/ml) (Sigma-Aldrich) for 0, 2, or 8 h, and then were harvested, lysed and subjected to western blot analysis.

Statistical analysis

Results are presented as the means ± standard deviation (SD) for at least 3 independent experiments in duplicate. Statistical analysis was carried out by one-way ANOVA with Tukey's test or two-way ANOVA followed by Bonferroni's correction. P-values <0.05 were considered to indicate statistically significant differences.

Results

TOPK inhibition increases H2O2-induced human COV434 granulosa cell apoptosis

Previous studies have demonstrated that oxidative stress induces granulosa cell apoptosis and SIRT1 downregulation mediated by oxidative stress contributes to this apoptosis (32-34). In addition, it has been suggested that TOPK functions as a survival effector in several cancer cells (35-37). Based on these findings, the present study wished to determine whether TOPK inhibition affects H2O2-induced granulosa cell apoptosis or SIRT1 expression. COV434 cells were treated with H2O2, OTS514 or H2O2 plus OTS514 for 24 h. Treatment of human COV434 granulosa cells with the TOPK inhibitor, OTS514, increased the number of TUNEL-positive cells induced by H2O2 stimulation (Fig. 1A), and significantly elevated the H2O2-induced cleavage of PARP (Fig. 1B). Of note, OTS514 treatment dose-dependently decreased TOPK expression upregulated by H2O2 and abolished the expression of SIRT1 downregulated by H2O2. Furthermore, the total p53 level, as well as its phosphorylation and acetylation were markedly increased upon H2O2 stimulation, and these effects were enhanced by OTS514 co-treatment. These data suggest that TOPK plays an important role in the regulation of p53 or SIRT1 expression in response to H2O2.

Subsequently, the effects of TOPK overexpression on the cleavage of PARP induced by H2O2 were examined. For this purpose, the COV434 cells were transfected with empty vector or TOPK overexpression vector for 24 h, and then incubated with a combination of H2O2 or OTS514 for 24 h. TOPK overexpression blocked the H2O2-induced cleavage of PARP (Fig. 1C). Furthermore, the effects of TOPK inhibition on endogenous ROS generation were investigated by 2′,7′-dichlo-roflurescin diacetate assay. The results revealed that targeting TOPK by OTS514 treatment enhanced the H2O2-induced ROS generation in granulosa cells, accelerating H2O2-induced the apoptosis (Fig. 1D).

Caspase inhibitor blocks granulosa cell apoptosis induced by co-treatment with H2O2 and OTS514

The present study then examined whether granulosa cell apoptosis induced by H2O2 and OTS514 is dependent on caspase-dependent apoptotic signaling pathways. COV434 cells were exposed to H2O2 only, H2O2 plus OTS514 or H2O2 plus OTS514 in combination with Z-VAD-FMK for 24 h. Co-treatment of the COV434 cells with OTS514 and H2O2 markedly increased the number of TUNEL-positive cells, compared to the cells stimulated with H2O2 only. However, co-treatment with the cell-permeable, irreversible pan-caspase inhibitor, Z-VAD-FMK, significantly decreased the number of TUNEL-positive cells (Fig. 2A). Consistent with this finding, flow cytometric analysis indicated that Z-VAD-FMK effectively diminished the number of Annexin V-positive cells induced by treatment with H2O2 and OTS514 (Fig. 2B). These findings suggest that caspase-dependent apoptosis pathways play a pivotal role in granulosa cell apoptosis induced by H2O2 and OTS514.

SIRT1 plays an important role in the regulation of H2O2-induced granulosa cell apoptosis

Subsequently, whether SIRT1 regulates H2O2-induced granulosa cell apoptosis was investigated. For this purpose, the SIRT1 activator, resveratrol, or the SIRT1 inhibitor, Ex527, were employed. The COV434 cells were incubated with H2O2 only, or H2O2 plus resveratrol or Ex527. As was expected, co-treatment with resveratrol decreased H2O2-induced apoptosis (Fig. 3A), whereas co-treatment with Ex527 increased apoptosis. In addition, western blot analysis revealed that co-treatment with resveratrol decreased the level of cleaved PARP level, as well as the acetylated p53 level induced by H2O2 (Fig. 3B); however, co-treatment with Ex527 increased the level of cleaved PARP and acetylated p53 level (Fig. 3C). These findings demonstrate that SIRT1 activity affects H2O2-induced granulosa cell apoptosis and they p53 level or activity is a key factor for the suppression of apoptosis.

p53 inhibition suppresses H2O2 or OTS514-induced granulosa cell apoptosis

It has been suggested that tumor suppressor p53 is involved in granulosa cell apoptosis in response to oxidative stress or cAMP-mediated signaling (33,38). The present study then wished to determine whether p53 affects COV434 cell apoptosis upon H2O2 and OTS514 treatment. For this purpose, COV434 cells were treated with a combination of H2O2, OTS514 or the p53 inhibitor, Pifithrin-µ. The inhibition of p53 by Pifithrin-µ treatment decreased the H2O2-induced increase in the level of cleaved PARP (Fig. 4A). Co-treatment with OTS514 decreased TOPK expression, elevated the p53 level and decreased SIRT1 expression. In addition, TUNEL assay revealed that the inhibition of p53 decreased the H2O2-induced COV434 cell apoptosis by approximately 2-fold. However, co-treatment with OTS514 increased cell apoptosis (Fig. 4B). Taken together, these results demonstrate that p53 plays a key role in H2O2 or OTS514-induced granulosa cell apoptosis.

Inhibition of Mdm2 accelerates H2O2- and OTS514-induced granulosa cell apoptosis

To examine the effects of Mdm2 inhibition on H2O2 and OTS514-induced granulosa cell apoptosis, the Mdm2 antagonist, Nutlin 3, that inhibits the interaction of Mdm2 and p53 was employed. Treatment of the COV434 cells with Nutlin 3 elevated the levels of cleaved PARP, total p53, acetylated p53 or the phosphorylated p53 level, whereas it decreased the SIRT1 level upon H2O2 and OTS514 treatment (Fig. 5A). Moreover, flow cytometric analysis indicated that the inhibition of the interaction of Mdm2 and p53 markedly increased the number of Annexin V-positive cells (Fig. 5B). Subsequently, whether p53 knockdown affects the H2O2-induced decrease in SIRT1 expression was examined. As was expected, the results revealed that p53 knockdown markedly restored SIRT1 expression to decrease the H2O2-induced cleavage of PARP (Fig. 5C). These findings suggest that both the level and activity of p53 may function as important factors in the regulation of granulosa cell apoptosis.

To examine whether p53 stability is affected by TOPK inhibition in H2O2-exposed granulosa cells, a cycloheximide chase experiment was then performed. The COV434 cells were treated with H2O2, OTS514 and cycloheximide, an inhibitor of protein biosynthesis. The level of acetylated p53, as well as the total p53 or cleaved PARP level in the H2O2- and OTS514-treated cells was elevated, while the TOPK or SIRT1 level was almost abolished 8 h following cycloheximide treatment compared with the cells stimulated with H2O2 only (Fig. 6). These findings suggest that TOPK inhibitor positively regulates p53 stability in H2O2-stimulated granulosa cells.

TOPK inhibition decreases the SIRT1 transcriptional level downregulated by H2O2, but increases the H2O2-induced p53 transcriptional level

It has been suggested that p53 negatively regulates SIRT1 expression. It has also been suggested that p53 negatively regulates SIRT1 expression (11). The present study thus examined the effect of TOPK inhibition on the SIRT1 or p53 transcriptional level, and the effect of p53 expression on the SIRT1 transcriptional level upon H2O2 stimulation. COV434 cells were transfected with SIRT1 or a p53 promoter-driven luciferase reporter construct. At 24 h following transfection, the cells were incubated with a combination of H2O2 or OTS514. The results indicated that stimulation of the COV434 cells with H2O2 resulted in a decrease in the SIRT1 transcriptional level and co-treatment with OTS514 promoted this decrease (Fig. 7A). On the other hand, co-treatment with OTS514 promoted the level of p53 or the p53 target, p21 transcriptional level induced by H2O2 (Fig. 7B). Notably, the expression of GFP-p53 diminished the SIRT1 transcriptional level downregulated by H2O2 (Fig. 7A). These findings demonstrate that upon H2O2 exposure, TOPK inhibition suppresses the SIRT1 transcriptional level and enhances the p53 transcriptional level, and that p53 negatively regulates SIRT1 expression. In addition, schematic model indicated that TOPK inhibition enhanced p53-mediated granulosa cell apoptosis through SIRT1 downregulation in response to H2O2 (Fig. 7C), suggesting the role of TOPK in the SIRT1/p53 regulatory axis.

Discussion

Granulosa cells are well known to be a class of cells that constitute a follicle for oocyte maturation. It has been proposed that granulosa cells and oocytes form gap junctions through connexins, which transfer low molecular weight molecules, such as cGMP, cAMP and calcium, and that granulosa cells transfer nutrients and regulatory factors for the growth and differentiation of oocytes (39-43). In addition, it seems that granulosa cells protect oocytes from oxidative stress involving ROS during follicle development. ROS have been reported to carry out functions that induce follicle rupture, and to regulate the expression of genes which induce oocyte maturation; however, accumulated ROS levels have been shown to cause oxidative stress that damages oocytes and granulosa cells (44-47). ROS mainly produced from the mitochondria cause mitochondrial damage to increase Bax expression, decrease Bcl-2 expression, and to release cytochrome c that forms the apoptosome, leading to the activation of the caspase cascade (48,49).

In this study, it was determined whether TOPK can affect oxidative stress-induced human granulosa cell apoptosis. It has been suggested that OTS514, a thieno[2,3-c]quinolone compound, strongly suppresses the growth of TOPK-positive cancer cells, including ovarian cancer, lung cancer or leukemia with low IC50 values ranging from 1.5 to 14 nM (50-52). OTS514 was shown to inhibit TOPK expression as well as TOPK kinase activity. It has been suggested that OTS514 suppresses Forkhead box protein M1 (FOXM1) and FOXM1 transactivates the c-Myc promoter (52,53). Furthermore, c-Myc has been shown to positively regulate TOPK expression (54). In addition, FOXM1 enhances cyclin B1 that phosphorylates TOPK on the Thr-9 residue (55). Collectively, it seems that OTS514 inhibits TOPK activity and expression by suppressing FOXM1 or c-Myc. Based on these reports, an inhibitor was employed in the presents study to suppress TOPK expression or activity. The inhibition of TOPK activity or expression by the TOPK inhibitor, OTS514, exacerbated H2O2-induced COV434 granulosa cell apoptosis, which is dependent on caspase signaling pathways. It was also found that ROS production was markedly increased by co-treatment with H2O2 and TOPK inhibitor than with H2O2 only in COV434 granulosa cells. consistent with these findings, it has been previously demonstrated that TOPK inhibits the accumulation of H2O2 in RPMI7951 melanoma cells (56), induces the expression of manganese superoxide dismutase (MnSOD), an essential antioxidant enzyme localized in the mitochondria, in PC12 cells (57), and induces the expression of Nrf2, an antioxidant molecule, to reduce ROS (58). Collectively, the present study provides evidence that TOPK negatively regulates oxida-tive stress-induced granulosa cell apoptosis, thereby positively contributing to follicular development.

It has been suggested that oxidative stress induces tumor suppressor p53 to increase the expression of p53 target proteins, including p21, Bax and PUMA (9,59,60). In granulosa cells, oxidative stress has also been shown to result in PUMA expression, leading to the promotion of apoptosis through p53 induction (33). The findings of the presents study demonstrated that TOPK inhibition by OTS514 elevated the expression of total p53, acetylated p53 or the phosphorylated p53 level in granulosa cells. In addition, it was found that the inhibition of p53 degradation by Nutlin 3 accelerated H2O2-mediated granulosa cell apoptosis. It appears that the increase in the phosphorylated or acetylated p53 level, as well as the total p53 level contributed to the apoptosis. In agreement with these findings, it has been reported that TOPK binds to the p53 DNA binding domain, leading to the inhibition of expression of p53 and p53 target protein, p21, and thereby the induction of cell cycle arrest (29,30). Moreover, it has been proposed that the phosphorylation of p53 at the Ser-15 residue decreases its interaction with Mdm2, but increases its interaction with CBP/p300, thereby promoting p53 acetylation and activity (61,62). It is well known that p53-mediated apoptosis is due to the p53 translocation into the mitochondria. The present study confirmed that p53 inhibition by Pifithrin-µ, an inhibitor of p53 binding to the mitochondria alleviates granulosa cell apoptosis induced by H2O2 and OTS514. Previous studies have demonstrated that mitochondrial p53 accumulation promotes the opening of the mitochondrial permeability transition pore (MPTP), which is a protein channel for the regulation of mitochondrial membrane permeability (63-65). Mitochondrial p53 is known to induce Bak and Bax oligomerization and to suppress the anti-apoptotic activity of Bcl-xL and Bcl-2 (63,66,67).

On the other hand, it has been suggested that p53 deacetylation is mostly caused by HDACs, but SIRT1 specifically deacetylates p53 at the K382 residue (60,68,69). The present study found that the regulation of p53 acetylation by SIRT1 activator or inhibitor affected H2O2-induced granulosa cell apoptosis, and that H2O2-induced apoptosis was further decreased by resveratrol and Pifithrin-µ, and increased by Ex527 and Nutlin 3. Furthermore, TOPK inhibition diminished SIRT1 transcriptional activity downregulated by H2O2 treatment and p53 expression promoted the inhibitory effect of TOPK, which may be due to an increase in p53 expression induced by TOPK inhibition. In agreement with these findings, previous studies have suggested that p53 inhibits SIRT1 transcription by directly binding to the SIRT1 promoter, and that TOPK functions as a suppressor for p53 (11,30).

It has been suggested that SIRT1 binds to p53 and deacetylates the C-terminal p53 residue, Lys382, to negatively regulate the transcriptional activity of p53 (70,71). In the present study, the SIRT1 activator, resveratrol, or the SIRT1 inhibitor, Ex527, decreased or increased H2O2-induced TOPK expression, respectively. Although research on the regulation of the TOPK promoter remains elusive, p53 may transcriptionally modulate TOPK expression (16). Therefore, these findings may be due to the change in the H2O2 induction of SIRT1-mediated p53 transactivation, activity regulating TOPK expression. Taken together, these findings provide evidence for a novel role of TOPK in the p53/SIRT1 regulatory axis that mediates granu-losa cell apoptosis. Therefore, TOPK may be a novel target molecule for the improvement of follicular development or oocyte maturation.

Acknowledgements

The authors would like to thank the laboratory (Protein Expression Laboratory, Department of Biochemstry, Konyang University) members for their helpful discussion about this work.

Abbreviations:

TOPK

T-lymphokine-activated killer cell-originated protein kinase

SIRT1

sirtuin1

ROS

reactive oxygen species

H2O2

hydrogen peroxide

PARP

poly(ADP-ribose) polymerase

HDAC

histone deacetylase

MnSOD

manganese superoxide dismutase

Bax

Bcl-2-associated X protein

Funding

The present study was supported by the National Research Foundation of Korea (NRF) grants funded by the Korea government (MSIP) (NRF-2019R1I1A3A01063191). The present study was also supported by the Priority Research Centers Program through the NRF funded by the MEST (NRF-2017R1A6A1A03015713).

Availability of materials and data

The data that support the findings of the present study are available from the corresponding author on reasonable request.

Authors' contributions

JHP and SAP conducted experiments, collected data, analyzed data and wrote the manuscript. YJL and NRJ performed experiments. JS provided some reagents and made suggestions during the performing of the experiments and data analysis. SMO designed and supervised the study. All authors read and approved the final manuscript.

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.

References

1 

Adeldust H, Zeinoaldini S, Kohram H, Amiri Roudbar M and Daliri Joupari M: In vitro maturation of ovine oocyte in a modified granulosa cells co-culture system and alpha-tocopherol supplementation: Effects on nuclear maturation and cleavage. J Anim Sci Technol. 57:272015. View Article : Google Scholar : PubMed/NCBI

2 

Tripathi A, Shrivastav TG and Chaube SK: An increase of granu-losa cell apoptosis mediates aqueous neem (Azadirachta indica) leaf extract-induced oocyte apoptosis in rat. Int J Appl Basic Med Res. 3:27–36. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Devine PJ, Perreault SD and Luderer U: Roles of reactive oxygen species and antioxidants in ovarian toxicity. Biol Reprod. 86:272012.

4 

Tatone C, Di Emidio G, Vitti M, Di Carlo M, Santini S, D'Alessandro AM, Falone S and Amicarelli F: Sirtuin functions in female fertility: Possible role in oxidative stress and aging. Oxid Med Cell Longev. 2015:6596872015.PubMed/NCBI

5 

Yang H, Yan B, Liao D, Huang S and Qiu Y: Acetylation of HDAC1 and degradation of SIRT1 form a positive feedback loop to regulate p53 acetylation during heat-shock stress. Cell Death Dis. 6:e17472015.PubMed/NCBI

6 

Han Y, Luo H, Wang H, Cai J and Zhang Y: SIRT1 induces resistance to apoptosis in human granulosa cells by activating the ERK pathway and inhibiting NF-κB signaling with anti-inflammatory functions. Apoptosis. 22:1260–1272. 2017.PubMed/NCBI

7 

Abdolvahabi Z, Nourbakhsh M, Hosseinkhani S, Hesari Z, Alipour M, Jafarzadeh M, Ghorbanhosseini SS, Seiri P, Yousefi Z, Yarahmadi S and Golpou P: MicroRNA-590-3P suppresses cell survival and triggers breast cancer cell apoptosis via targeting sirtuin-1 and deacetylation of p53. J Cell Biochem. 120:9356–9368. 2019.

8 

Yu X, Zhang S, Zhao D, Zhang X, Xia C, Wang T, Zhang M, Liu T, Huang W and Wu B: SIRT1 inhibits apoptosis in in vivo and in vitro models of spinal cord injury via microRNA-494. Int J Mol Med. 43:1758–1768. 2019.PubMed/NCBI

9 

Han MK, Song EK, Guo Y, Ou X, Mantel C and Broxmeyer HE: SIRT1 regulates apoptosis and Nanog expression in mouse embryonic stem cells by controlling p53 subcellular localization. Cell Stem Cell. 2:241–251. 2008.PubMed/NCBI

10 

Kume S, Haneda M, Kanasaki K, Sugimoto T, Araki SI, Isono M, Isshiki K, Uzu T, Kashiwagi A and Koya D: Silent information regulator 2 (SIRT1) attenuates oxidative stress-induced mesangial cell apoptosis via p53 deacetylation. Free Radic Biol Med. 40:2175–2182. 2006.PubMed/NCBI

11 

Yuan F, Liu L, Lei Y and Tang P: P53 inhibits the upregulation of sirtuin 1 expression induced by c-Myc. Oncol Lett. 14:4396–4402. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Zlobec I, Molinari F, Kovac M, Bihl MP, Altermatt HJ, Diebold J, Frick H, Germer M, Horcic M, Montani M, et al: Prognostic and predictive value of TOPK stratified by KRAS and BRAF gene alterations in sporadic, hereditary and metastatic colorectal cancer patients. Br J Cancer. 102:151–161. 2010. View Article : Google Scholar :

13 

Zykova TA, Zhu F, Wang L, Li H, Bai R, Lim DY, Yao K, Bode AM and Dong Z: The T-LAK cell-originated protein kinase signal pathway promotes colorectal cancer metastasis. EBioMedicine. 18:73–82. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Su TC, Chen CY, Tsai WC, Hsu HT, Yen HH, Sung WW and Chen CJ: Cytoplasmic, nuclear, and total PBK/TOPK expression is associated with prognosis in colorectal cancer patients: A retrospective analysis based on immunohistochemistry stain of tissue microarrays. PLoS One. 13:e02048662018. View Article : Google Scholar : PubMed/NCBI

15 

Shih MC, Chen JY, Wu YC, Jan YH, Yang BM, Lu PJ, Cheng HC, Huang MS, Yang CJ, Hsiao M and Lai JM: TOPK/PBK promotes cell migration via modulation of the PI3K/PTEN/AKT pathway and is associated with poor prognosis in lung cancer. Oncogene. 31:2389–2400. 2012. View Article : Google Scholar

16 

Lei B, Qi W, Zhao Y, Li Y, Liu S, Xu X, Zhi C, Wan L and Shen H: PBK/TOPK expression correlates with mutant p53 and affects patients' prognosis and cell proliferation and viability in lung adenocarcinoma. Hum Pathol. 46:217–224. 2015. View Article : Google Scholar

17 

Ohashi T, Komatsu S, Ichikawa D, Miyamae M, Okajima W, Imamura T, Kiuchi J, Kosuga T, Konishi H, Shiozaki A, et al: Overexpression of PBK/TOPK relates to tumour malignant potential and poor outcome of gastric carcinoma. Br J Cancer. 116:218–226. 2017. View Article : Google Scholar :

18 

Sun H, Zhang L, Shi C, Hu P, Yan W, Wang Z, Duan Q, Lu F, Qin L, Lu T, et al: TOPK is highly expressed in circulating tumor cells, enabling metastasis of prostate cancer. Oncotarget. 6:12392–12404. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Pirovano G, Ashton TM, Herbert KJ, Bryant RJ, Verrill CL, Cerundolo L, Buffa FM, Prevo R, Harrap I, Ryan AJ, et al: TOPK modulates tumour-specific radiosensitivity and correlates with recurrence after prostate radiotherapy. Br J Cancer. 117:503–512. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Ikeda Y, Park JH, Miyamoto T, Takamatsu N, Kato T, Iwasa A, Okabe S, Imai Y, Fujiwara K, Nakamura Y, et al: T-LAK cell-originated protein kinase (TOPK) as a prognostic factor and a potential therapeutic target in ovarian cancer. Clin Cancer Res. 22:6110–6117. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Wang MY, Lin ZR, Cao Y, Zheng LS, Peng LX, Sun R, Meng DF, Xie P, Yang JP, Cao L, et al: PDZ binding kinase (PBK) is a theranostic target for nasopharyngeal carcinoma: Driving tumor growth via ROS signaling and correlating with patient survival. Oncotarget. 7:26604–26616. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Ohashi T, Komatsu S, Ichikawa D, Miyamae M, Okajima W, Imamura T, Kiuchi J, Nishibeppu K, Kosuga T, Konishi H, et al: Overexpression of PBK/TOPK contributes to tumor development and poor outcome of esophageal squamous cell carcinoma. Anticancer Res. 36:6457–6466. 2017. View Article : Google Scholar

23 

Abe Y, Matsumoto S, Kito K and Ueda N: Cloning and expression of a novel MAPKK-like protein kinase, lymphokine- activated killer T-cell-originated protein kinase, specifically expressed in the testis and activated lymphoid cells. J Biol Chem. 275:21525–21531. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Matsumoto S, Abe Y, Fujibuchi T, Takeuchi T, Kito K, Ueda N, Shigemoto K and Gyo K: Characterization of a MAPKK-like protein kinase TOPK. Biochem Biophys Res Commun. 325:997–1004. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Zhu F, Zykova TA, Kang BS, Wang Z, Ebeling MC, Abe Y, Ma WY, Bode AM and Dong Z: Bidirectional signals transduced by TOPK-ERK interaction increase tumorigenesis of HCT116 colorectal cancer cells. Gastroenterology. 133:219–231. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Aksamitiene E, Kholodenko BN, Kolch W, Hoek JB and Kiyatkin A: PI3K/Akt-Sensitive MEK-independent compensatory circuit of ERK activation in ER-positive PI3K-mutant T47D breast cancer cells. Cell Signal. 22:1369–1378. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Oh SM, Zhu F, Cho YY, Ki WL, Bong SK, Kim HG, Zykova T, Bode AM and Dong Z: T-Lymphokine-activated killer cell-originated protein kinase functions as a positive regulator of c-Jun-NH2-kinase 1 signaling and H-ras-induced cell transformation. Cancer Res. 67:5186–5194. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Seol MA, Park JH, Jeong JH, Lyu J, Han SY and Oh SM: Role of TOPK in lipopolysaccharide-induced breast cancer cell migration and invasion. Oncotarget. 8:40190–40203. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Nandi AK, Ford T, Fleksher D, Neuman B and Rapoport AP: Attenuation of DNA damage checkpoint by PBK, a novel mitotic kinase, involves protein-protein interaction with tumor suppressor p53. Biochem Biophy Res Commun. 358:181–188. 2007. View Article : Google Scholar

30 

Hu F, Gartenhaus RB, Eichberg D, Liu Z, Fang HB and Rapoport AP: PBK/TOPK interacts with the DBD domain of tumor suppressor p53 and modulates expression of transcriptional targets including p21. Oncogene. 29:5464–5474. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Park JH, Yoon DS, Choi HJ, Hahm DH and Oh SM: Phosphorylation of IκBα at serine 32 by T-lymphokine-activated killer cell-originated protein kinase is essential for chemoresistance against doxorubicin in cervical cancer cells. J Biol Chem. 288:3585–3593. 2013. View Article : Google Scholar

32 

Weng Q, Liu Z, Li B, Liu K, Wu W and Liu H: Oxidative stress induces mouse follicular granulosa cells apoptosis via JNK/FoxO1pathway. PLoS One. 11:e01678692016. View Article : Google Scholar

33 

Yang H, Xie Y, Yang D and Ren D: Oxidative stress-induced apoptosis in granulosa cells involves JNK, p53 and puma. Oncotarget. 8:25310–25322. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Zhang M, Zhang Q, Hu Y, Xu L, Jiang Y, Zhang C, Ding L, Jiang R, Sun J, Sun H and Yan G: MiR-181a increases FoxO1 acetylation and promotes granulosa cell apoptosis via SIRT1 downregulation. Cell Death Dis. 8:e30882017. View Article : Google Scholar : PubMed/NCBI

35 

Joel M, Mughal AA, Grieg Z, Murrell W, Palmero S, Mikkelsen B, Fjerdingstad HB, Sandberg CJ, Behnan J, Glover JC, et al: Targeting PBK/TOPK decreases growth and survival of glioma initiating cells in vitro and attenuates tumor growth in vivo. Mol Cancer. 14:1212015. View Article : Google Scholar : PubMed/NCBI

36 

Kim DJ, Li Y, Reddy K, Lee MH, Kim MO, Cho YY, Lee SY, Kim JE, Bode AM and Dong Z: Novel TOPK inhibitor HI-TOPK-032 effectively suppresses colon cancer growth. Cancer Res. 72:3060–3068. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Xu M and Xu S: PBK/TOPK overexpression and survival in solid tumors: A PRISMA-compliant meta-analysis. Medicine (Baltimore). 98:e147662019. View Article : Google Scholar

38 

Keren-Tal I, Suh BS, Dantes A, Lindner S, Oren M and Amsterdam A: Involvement of p53 expression in cAMP-mediated apoptosis in immortalized granulosa cells. Exp Cell Res. 218:283–295. 1995. View Article : Google Scholar : PubMed/NCBI

39 

Heller DT, Cahill DM and Schultz RM: Biochemical studies of mammalian oogenesis: Metabolic cooperativity between granu-losa cells and growing mouse oocytes. Dev Biol. 84:455–464. 1981. View Article : Google Scholar : PubMed/NCBI

40 

Brower PT and Schultz RM: Intercellular communication between granulosa cells and mouse oocytes: Existence and possible nutritional role during oocyte growth. Dev Biol. 90:144–153. 1982. View Article : Google Scholar : PubMed/NCBI

41 

Bruzzone R, White TW and Paul DL: Connections with connexins: The molecular basis of direct intercellular signaling. Eur J Biochem. 238:1–27. 1996. View Article : Google Scholar : PubMed/NCBI

42 

Kumar NM and Gilula NB: The gap junction communication channel. Cell. 84:381–388. 1996. View Article : Google Scholar : PubMed/NCBI

43 

Grazul-Bilska AT, Reynolds LP and Redmer DA: Gap junctions in the ovaries. Biol Reprod. 57:947–957. 1997. View Article : Google Scholar : PubMed/NCBI

44 

Yang HW, Hwang KJ, Kwon HC, Kim HS, Choi KW and Oh KS: Detection of reactive oxygen species (ROS) and apoptosis in human fragmented embryos. Hum Reprod. 13:998–1002. 1998. View Article : Google Scholar : PubMed/NCBI

45 

Hensley K, Robinson KA, Gabbita SP, Salsman S and Floyd RA: Reactive oxygen species, cell signaling, and cell injury. Free Radic Biol Med. 28:1456–1462. 2000. View Article : Google Scholar : PubMed/NCBI

46 

Dröge W: Free radicals in the physiological control of cell function. Physiol Rev. 82:47–95. 2002. View Article : Google Scholar : PubMed/NCBI

47 

Goud AP, Goud PT, Diamond MP, Gonik B and Abu-Soud HM: Reactive oxygen species and oocyte aging: Role of superoxide, hydrogen peroxide, and hypochlorous acid. Free Radic Biol Med. 44:1295–1304. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Li D, Ueta E, Kimura T, Yamamoto T and Osaki T: Reactive oxygen species (ROS) control the expression of Bcl-2 family proteins by regulating their phosphorylation and ubiquitination. Cancer Sci. 95:644–650. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Alarifi S, Ali H, Alkahtani S and Alessia MS: Regulation of apoptosis through bcl-2/bax proteins expression and DNA damage by nano-sized gadolinium oxide. Int J Nanomedicine. 12:4541–4551. 2017. View Article : Google Scholar : PubMed/NCBI

50 

Alachkar H, Mutonga M, Malnassy G, Park JH, Fulton N, Woods A, Meng L, Kline J, Raca G, Odenike O, et al: T-LAK cell-originated protein kinase presents a novel therapeutic target in FLT3-ITD mutated acute myeloid leukemia. Oncotarget. 6:33410–33425. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Matsuo Y, Park JH, Miyamoto T, Yamamoto S, Hisada S, Alachkar H and Nakamura Y: TOPK inhibitor induces complete tumor regression in xenograft models of human cancer through inhibition of cytokinesis. Sci Transl Med. 6:259ra1452014. View Article : Google Scholar : PubMed/NCBI

52 

Park JH, Inoue H, Kato T, Zewde M, Miyamoto T, Matsuo Y, Salgia R and Nakamura Y: TOPK (T-LAK cell-originated protein kinase) inhibitor exhibits growth suppressive effect on small cell lung cancer. Cancer Sci. 108:488–496. 2017. View Article : Google Scholar : PubMed/NCBI

53 

Wierstra I and Alves J: FOXM1c transactivates the human c-myc promoter directly via the two TATA boxes P1 and P2. FEBS J. 273:4645–4667. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Hu F, Gartenhaus RB, Zhao XF, Fang HB, Minkove S, Poss DE and Rapoport AP: C-Myc and E2F1 drive PBK/TOPK expression in high-grade malignant lymphomas. Leukemia Res. 37:447–454. 2013. View Article : Google Scholar

55 

Leung TW, Lin SS, Tsang AC, Tong CS, Ching JC, Leung WY, Gimlich R, Wong GG and Yao KM: Over-expression of foxM1 stimulates cyclin B1 expression. FEBS Lett. 507:59–66. 2001. View Article : Google Scholar

56 

Zykova TA, Zhu F, Vakorina TI, Zhang J, Higgins LA, Urusova DV, Bode AM and Dong Z: T-LAK cell-originated protein kinase (TOPK) phosphorylation of prx1 at ser-32 prevents UVB-induced apoptosis in RPMI7951 melanoma cells through the regulation of Prx1 peroxidase activity. J Biol Chem. 285:29138–29146. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Zhao H, Wang R, Tao Z, Gao L, Yan F, Gao Z, Liu X, Ji X and Luo Y: Ischemic postconditioning relieves cerebral ischemia and reperfusion injury through activating T-LAK cell-originated protein kinase/protein kinase b pathway in rats. Stroke. 45:2417–2424. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Liu Y, Liu H, Cao H, Song B and Zhang W and Zhang W: PBK/TOPK mediates promyelocyte proliferation via Nrf2-regulated cell cycle progression and apoptosis. Oncol Rep. 34:3288–3296. 2015. View Article : Google Scholar : PubMed/NCBI

59 

Hsieh TC, Juan G, Darzynkiewicz Z and Wu JM: Resveratrol increases nitric oxide synthase, induces accumulation of p53 and p21(WAF1/CIP1), and suppresses cultured bovine pulmonary artery endothelial cell proliferation by perturbing progression through S and G2. Cancer Res. 59:2596–2601. 1999.

60 

Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, Guarente L and Gu W: Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell. 107:137–148. 2001. View Article : Google Scholar : PubMed/NCBI

61 

Shieh SY, Ikeda M, Taya Y and Prives C: DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 91:325–334. 1997. View Article : Google Scholar : PubMed/NCBI

62 

Lambert PF, Kashanchi F, Radonovich MF, Shiekhattar R and Brady JN: Phosphorylation of p53 serine 15 increases interaction with CBP. J Biol Chem. 273:33048–33053. 1998. View Article : Google Scholar : PubMed/NCBI

63 

Marchenko ND and Moll UM: Mitochondrial death functions of p53. Mol Cell Oncol. 1:e9559952014. View Article : Google Scholar : PubMed/NCBI

64 

Chen LJ, Gao YQ, Li XJ, Shen DH and Sun FY: Melatonin protects against MPTP/MPP+ -induced mitochondrial DNA oxidative damage in vivo and in vitro. J Pineal Res. 39:34–42. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Yu W, Zhang X, Liu J, Wang X, Li S, Liu R, Liao N, Zhang T and Hai C: Cyclosporine A suppressed glucose oxidase induced P53 mitochondrial translocation and hepatic cell apoptosis through blocking mitochondrial permeability transition. Int J Biol Sci. 12:198–209. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Wolff S, Erster S, Palacios G and Moll UM: P53's mitochondrial translocation and MOMP action is independent of puma and bax and severely disrupts mitochondrial membrane integrity. Cell Res. 18:733–744. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Sykes SM, Mellert HS, Holbert MA, Li K, Marmorstein R, Lane WS and McMahon SB: Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol Cell. 24:841–851. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Brooks CL and Gu W: The impact of acetylation and deacetylation on the p53 pathway. Protein Cell. 2:456–462. 2011. View Article : Google Scholar : PubMed/NCBI

69 

Islam S, Abiko Y, Uehara O and Chiba I: Sirtuin 1 and oral cancer. Oncol Lett. 17:729–738. 2019.PubMed/NCBI

70 

Ong AL and Ramasamy TS: Role of sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res Rev. 43:64–80. 2018. View Article : Google Scholar : PubMed/NCBI

71 

van Leeuwen I and Lain S: Sirtuins and p53. Adv Cancer Res. 102:171–195. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 46 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Park JH, Park SA, Lee YJ, Joo NR, Shin J and Oh SM: TOPK inhibition accelerates oxidative stress‑induced granulosa cell apoptosis via the p53/SIRT1 axis. Int J Mol Med 46: 1923-1937, 2020
APA
Park, J., Park, S., Lee, Y., Joo, N., Shin, J., & Oh, S. (2020). TOPK inhibition accelerates oxidative stress‑induced granulosa cell apoptosis via the p53/SIRT1 axis. International Journal of Molecular Medicine, 46, 1923-1937. https://doi.org/10.3892/ijmm.2020.4712
MLA
Park, J., Park, S., Lee, Y., Joo, N., Shin, J., Oh, S."TOPK inhibition accelerates oxidative stress‑induced granulosa cell apoptosis via the p53/SIRT1 axis". International Journal of Molecular Medicine 46.5 (2020): 1923-1937.
Chicago
Park, J., Park, S., Lee, Y., Joo, N., Shin, J., Oh, S."TOPK inhibition accelerates oxidative stress‑induced granulosa cell apoptosis via the p53/SIRT1 axis". International Journal of Molecular Medicine 46, no. 5 (2020): 1923-1937. https://doi.org/10.3892/ijmm.2020.4712