Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway

  • Authors:
    • Huixiang Wang
    • Xiaoli Zhao
    • Chengxiang Ni
    • Yuyang Dai
    • Yan Guo
  • View Affiliations

  • Published online on: March 29, 2018     https://doi.org/10.3892/mmr.2018.8823
  • Pages: 7797-7806
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Increased endometrial stromal cell (ESC) survival and migration is responsible for the development and progression of endometriosis. However, little is known about the mechanisms underlying ESC survival and migration, and limited therapeutic strategies that are able to reverse these abnormalities are available. The present study investigated the effects of zearalenone (ZEA) on ESC survival and migration, particularly focusing on mitochondrial fission and the c‑Jun N‑terminal kinase (JNK)/dynamin‑related protein 1 (Drp1) pathway. The results revealed that ZEA induced ESC apoptosis in a dose‑dependent manner. Furthermore, ZEA treatment triggered excessive mitochondrial fission resulting in structural and functional mitochondrial damage, leading to the collapse of the mitochondrial membrane potential and subsequent leakage of cytochrome c into the cytoplasm. This triggered the mitochondrial pathway of apoptosis. Additionally, ZEA‑induced mitochondrial fission decreased ESC migration through F‑actin/G‑actin homeostasis dysregulation. ZEA also increased JNK phosphorylation and subsequently Drp1 phosphorylation at the serine 616 position, resulting in Drp1 activation. JNK/Drp1 pathway inhibition abolished the inhibitory effects of ZEA on ESC survival and migration. In summary, the present study demonstrated that ZEA reduced ESC survival and migration through the stimulation of mitochondrial fission by activation of the JNK/Drp1 pathway.

Introduction

Endometriosis is characterized by endometriotic tissue growth outside the uterine cavity. The pathophysiological mechanism of this disorder remains unknown (1). It is a common, chronic, benign, estrogen-dependent, gynecological disorder associated with pelvic pain and infertility (2). Numerous studies have indicated that endometriosis is associated with increased endometrial stromal cell (ESC) survival and invasion, suggesting a link between a heightened menstrual repair response and ectopic endometrial implant formation (35). ESC survival and migration is controlled by a complex array of hormones, growth factors, chemokines and inflammatory mediators, involving signaling through phosphatidylinositol-4,5-biphosphate 3-kinase/AKT and mitogen-activated protein kinase (MAPK) pathways, particularly MAPK/c-Jun N-terminal kinase (JNK) signaling (6). Therefore, therapies that inhibit ESC migration and promote apoptosis may have the potential to effectively treat endometriosis.

Mitochondria are central to a variety of cellular physiological processes, including bioenergetic regulation, cellular oxidation-reduction status maintenance and apoptosis induction (7). The mitochondrial network is associated with mitochondrial dynamics and the morphology ranges from a highly interconnected and elongated to a highly fragmented and punctated network (8). The primary mitochondrial dynamics machinery is responsible for mitochondrial membrane fusion and fission. Changes in mitochondrial fission have been reported as an early event occurring in cancer cell proliferation, apoptosis, metabolism, cell motility and migration (9,10). Mitochondrial fission contributes to the mitochondrial pathway of apoptosis via the induction of cardiolipin oxidation by mitochondrial reactive oxygen species (mROS) and mitochondrial permeability transition pore (mPTP) opening, induced by hexokinase 2 (HK2)/voltage-dependent anion channel 1 (VDAC1) dissociation (11). ESC apoptosis and migration are pathogenic factors in endometriosis (12,13). Cell motility is regulated by the balance of F-actin/G-actin. Notably, these actin proteins also facilitate the recruitment of dynamin-related protein 1 (Drp1) to the mitochondria and subsequent mitochondrial fission (14,15). Previous study has confirmed that the accumulation of actin filaments at future fission sites increases the rate of fission, indicating that mitochondrial fission is associated with the migration of malignant cells through the influence of actin homeostasis (16). However, the ability of mitochondrial fission to modulate the apoptosis and migration of ESCs remains unknown.

Zearalenone (ZEA) is a non-steroidal mycotoxin produced by several fungi of the genus Fusarium (17). Accumulating evidence has demonstrated that ZEA may regulate the cancer cell cycle (18), mitochondrial metabolism (19) and apoptosis (20). However, whether ZEA promotes ESC apoptosis through mitochondrial integrity modification remains unknown. Therefore, the role of mitochondrial fission in ESC apoptosis and migration were investigated in the present study. The protective mechanism of ZEA on ESCs was also examined. The results revealed that ZEA induces mitochondrial fission via amplification of the JNK/Drp1 pathway. Furthermore, increased mitochondrial fission caused ESC apoptosis by inducing functional and structural mitochondrial damage. ZEA-mediated mitochondrial fission also impaired ESC migration through the promotion of F-actin depolymerization.

Materials and methods

Ethics statement

The present study was conducted in accordance with the Declaration of Helsinki. The experimental protocol was approved by the Ethics Committee of the Department of Gynecology and Obstetrics, Beijing Tongren Hospital (Beijing, China).

Cell culture

The human ESCs was an American Type Culture Collection (ATCC; Manassas, VA, USA) culture (ATCC-CRL-4003; lot no. 3857441) purchased from LGC Promochem GmbH, Teddington, UK. ESCs were cultured according to ATCC recommendations in RPMI medium (Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) (21), 1% L-glutamine and 0.5% gentamycin (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) at 37°C in 5% CO2. Increasing concentrations of ZEA (1, 5, 10 and 20 µM; Z2125; Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) were applied to ESCs for 24 h once 70–80% confluence was reached (22).

Effects of ZEA on cell viability

To analyze the effects of various ZEA concentrations (1–20 µM) on ESC viability, an MTT assay was performed. ESCs were seeded into a 96-well plate (103 cells/well) (23) and 20 µl MTT (5 mg/ml PBS; pH 7.4; Sigma-Aldrich; Merck KGaA) was subsequently added to the medium for 4 h. The supernatant was discarded and 100 µl dimethyl sulfoxide was added to each well for 10 min. The optical density was measured at 490 nm (24,25).

Cell proliferation, migration and scratch assay

Cell proliferation was measured with a cell counting kit-8 (CCK-8) (Beyotime Institute of Biotechnology, Beijing, China) assay. Cell suspension (200 µl) was seeded in 96-well cell culture plates at a density of 1,000 cells/well and incubated at 37°C for 1–4 days, as previously described (26). Cell migration was analyzed using a Transwell chamber assay (1×105 cells; pore size, 8 µm) with a polycarbonate membrane, as previously described (27,28).

For the scratch assay, cells were cultured in serum-free medium for 24 h and were subsequently scratched with pipette tips, as described previously (29). Wound healing was observed for 48 h and images were captured using a light microscope (Olympus DX51; Olympus Corporation, Tokyo, Japan) every 24 h.

Western blot analysis

Cells were lysed with radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology) supplemented with phenylmethylsufonyl fluoride (Beyotime Institute of Biotechnology). Protein concentration was determined using a bicinchoninic acid protein assay. Protein (50 µg) was separated by 10% SDS-PAGE and transferred to polyvinylidene difluoride membranes. The membranes were subsequently blocked with 5% non-fat milk for 1 h at room temperature prior to incubation with the following primary antibodies: GAPDH (1:1,000; 5174; CST Biological Reagents Co., Ltd., Shanghai, China), caspase-3 (1:2,000; 9662; CST Biological Reagents Co., Ltd.), X-linked inhibitor of apoptosis (X–IAP; 1:1,000; 14334; CST Biological Reagents Co., Ltd.), phosphorylated (p-)JNK (1:1,000; 9225; CST Biological Reagents Co., Ltd.), p-Drp1 (1:500; ab193216; Abcam, Cambridge, MA, USA), B-cell lymphoma 2 (Bcl-2; 1:1,000; 15071; CST Biological Reagents Co., Ltd.) associated X protein (Bax; 1:2,000; ab32503), γ-actin (1:1,000; ab123034) and F-actin (1:1,000; ab205; all Abcam) overnight at 4°C (30). The membranes were washed in Tris-buffered saline with Tween-20 for 15 min and were subsequently incubated with horseradish peroxidase-conjugated secondary antibody (1:1,000; sc-2004/sc-2005; Santa Cruz Biotechnology, Inc., Dallas, TX, USA) for 1 h at room temperature. Blots were visualized with an enhanced chemiluminescence substrate kit (Thermo Fisher Scientific, Inc.) (31,32). The bands were scanned and quantified by Quantity One (version 4.6.2; Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Detection of lactate dehydrogenase (LDH) release, mitochondrial membrane potential (ΔΨm), adenosine triphosphate (ATP) production and mPTP opening

LDH released into the medium from injured cells was detected with an LDH cytotoxicity kit (Roche Diagnostics, Indianapolis, IN, USA). ΔΨm was analyzed with a JC-1 kit (Beyotime Institute of Biotechnology, Shanghai, China), according to the manufacturer's protocol (33,34). Cellular ATP was detected using a firefly luciferase-based ATP assay kit (Beyotime Institute of Biotechnology). mPTP opening was visualized as a rapid dissipation of tetramethylrhodamine ethyl ester fluorescence, as described previously (35).

Terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) staining

A TUNEL assay was performed using a one-step TUNEL kit (Beyotime Institute of Biotechnology, Haimen, China), according to the manufacturer's protocol. Briefly, cells were fixed for 1 h in 4% (w/v) paraformaldehyde at room temperature. Following specific labelling, the cells were exposed to DAPI (5 mg/ml) with PBS in the dark for 5 min at room temperature. TUNEL-positive cells were defined as those with fluorescein-dUTP staining present. Then 20 different fields were randomly selected under magnification, ×40 to count the number of apoptotic cells by confocal microscopy (FluoView 1000; Olympus Corporation) (36).

Determination of caspase-3 and caspase-9 activity

Caspase-3 and caspase-9 activities were detected with their respective activity assay kits (C1115 and C1157; Beyotime Institute of Biotechnology), according to the manufacturer's protocol. The assay was repeated three times (37).

Immunocytochemistry

To determine cytochrome c (cyt-c) localization and mitochondrial division, immunocytochemistry was performed (38). Cells (1,000 cells/well) were fixed in 4% paraformaldehyde in PBS for 15 min at room temperature, washed with PBS, and then permeabilized with 0.1% Triton X-100 in PBS for 5 min. Subsequently, the cells were blocked for 1 h with PBS containing 5% bovine serum albumin (Sigma-Aldrich) at room temperature incubated with primary antibodies against cyt-c (1:500; 12963; CST Biological Reagents Co., Ltd.) and translocase of outer mitochondrial membrane 20 (Tom20; 1:500; 42406; CST Biological Reagents Co., Ltd.) for 1 h at room temperature. Mitochondrial fission was observed via Tom20. F-actin was stained with rhodamin-phalloidin (1:100; Molecular Probes; Thermo Fisher Scientific, Inc.) in the dark for 15 min at room temperature. DAPI (1:100, Sigma-Aldrich; Merck KGaA) was used to stain the nuclei in the dark for 5 min at room temperature. Images of the immunostained cells were captured using a fluorescence microscope through a 50× objective (VANOX-S; Olympus Corporation).

ATP production and respiratory chain complex activities assays

The cellular ATP levels were measured using a firefly luciferase-based ATP assay kit (Beyotime, Shanghai, China). Complex I, II, and V activity was measured according to previous studies (7). Mitochondrial respiratory function was measured polarographically at 30°C using a Biological Oxygen Monitor System (Hansatech Instruments, King's Lynn, UK) and a Clarktype oxygen electrode (Hansatech DW1, Norfolk, UK).

Statistical analysis

All analyses were performed with SPSS software version 20.0 (SPSS Inc.; IBM Corp., Armonk, NY, USA). All experiments were repeated three times. All results are expressed as the mean ± standard deviation and statistical significance for each variable was estimated by a one-way analysis of variance followed by Tukey's test for the post hoc analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

ZEA reduces ESC viability and growth

The MTT assay revealed a dose-dependent decrease in cell viability following treatment with ZEA for 24 h (Fig. 1B), indicating that ZEA has a toxic effect on ESC viability. The CCK-8 assay was performed to assess the growth capacity of ESCs treated with ZEA. Compared with the control group, ZEA treatment significantly interfered with the proliferative capacity of ESCs. However, no statistical difference was noted between the groups in the first 12 h of treatment. The growth capacity of ESCs reduced as the ZEA concentration increased (Fig. 1A), suggesting that ZEA reduced ESC proliferation.

ZEA induces ESC apoptosis through mitochondrial fission

As no significant difference was observed in ESC proliferation following 12 h ZEA treatment, ESCs were treated with ZEA for 24 h in the subsequent experiments to exclude the influence of proliferation on cell numbers (Fig. 2). A TUNEL assay was performed to detect the apoptotic rate of ESCs treated with ZEA. The results demonstrated that ZEA significantly increased the apoptotic rate in ESCs compared with the rate observed in the control group (Fig. 2A and B). Approximately 8.35±2.73% of ESCs were TUNEL-positive in the control group. The percentage of TUNEL-positive cells was significantly increased by ZEA in a concentration-dependent manner: 1 µM/l, 19.35±4.64%; 5 µM/l, 36.58±3.29%; 10 µM/l, 58.37±4.26%; and 20 µM/l, 64.21±4.57% (Fig. 2B). These data indicate that ZEA had a lethal impact on ESCs. Caspase-3 activity was investigated as its activation represents an essential and final step in the apoptotic process, leading to the induction of DNA breakage and subsequent cellular apoptosis (7). Significantly increased caspase-3 activity was observed in the ZEA-treated cells compared with the level of activity in the control group (Fig. 2C). The largest pro-apoptotic effect was detected in the of 20 µM/l ZEA group. Thus, this concentration was used for subsequent experiments.

Several studies have identified that mitochondrial fission is a critical process in determining cellular survival and apoptosis (39,40). Changes in mitochondrial morphology were investigated to examine the role of mitochondrial fission in ZEA-mediated cellular apoptosis. The mitochondrial fission inducer, carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), and the inhibitor, Mdivi1, were used. Compared with the control group, the ZEA-treated group had a marked increase in the level of fragmented mitochondria, with evidence of rounder mitochondria and increased debris (Fig. 2D). The mitochondrial length was also significantly reduced following treatment with ZEA compared with the length in the control group (Fig. 2E). Mdivi1 application with ZEA markedly blocked the mitochondrial fragmentation mediated by ZEA. FCCP was used as the positive control group and induced substantial amounts of mitochondrial debris. These results indicate that ZEA promoted mitochondrial fission.

To establish whether ZEA induced ESC apoptosis by mitochondrial fission induction, LDH release and caspase-9 activity assays were conducted with and without Mdivi1. Inhibition of mitochondrial fission with Mdivi1 significantly reduced the concentration of LDH detected in the medium compared with the level in the ZEA group (Fig. 2G). Similarly, increased caspase-9 activity in the ZEA treatment group was significantly reduced with Mdivi1 application. Notably, FCCP not only promoted LDH release but also enhanced caspase-9 activity. Similar results were observed in the ZEA group (Fig. 2F). Taken together, these data suggest that ZEA induced ESC apoptosis through the upregulation of mitochondrial fission.

Mitochondrial fission contributes to mitochondrial dysfunction and structural damage

Changes in mitochondrial function and structure were examined to elucidate the mechanism by which mitochondrial fission activates cellular apoptosis (11). The primary function of the mitochondria is to produce adequate ATP to fuel cell metabolism (41,42). Significantly reduced ATP production was detected in the ZEA treatment group when compared with the control group, demonstrating that ZEA treatment reduces the ability of mitochondria to generate sufficient ATP (Fig. 3A). Mdivi1 was demonstrated to reverse the ZEA-mediated decrease in ATP, indicating that ZEA-induced mitochondrial fission is responsible for mitochondrial energy disorder. This functional change was induced by the ZEA-induced structural damage. ΔΨm is fundamental to ATP production (43). Over time, the mitochondrial membrane potential dissipates following ZEA treatment (Fig. 3B and C) and these changes were reversed with the addition of Mdivi1 (Fig. 3D and E). ΔΨm collapse is a marker of increased mitochondrial membrane permeability, which may lead to the leakage of mitochondrial proteins from the mitochondria into the cytoplasm (44). The addition of ZEA resulted in increased mitochondrial release of cyt-c, with evidence of dispersion into the cytoplasm and nucleus (Fig. 3F). Furthermore, ZEA treatment promoted excessive mPTP opening and decreased electron transport chain complex function (ETCx; Fig. 3G and H). Mdivi1 was observed to partially suppress cyt-c diffusion and mPTP opening.

Western blot analysis

Cyt-c release triggers the formation of the apoptosome, a complex comprising of apoptotic peptidase activating factor 1, cyt-c, dATP (deoxy-adenosine triphosphate) and pro-caspase-9 (45). The apoptosome triggers caspase-9 activation, which subsequently cleaves caspase-3 to its active form. Caspase-3 may then activate the mitochondrial cell death effectors (46). Western blot analysis was used to examine the downstream changes induced by cyt-c leakage. ZEA treatment significantly increased levels of the pro-apoptotic proteins, cleaved caspase-3 and Bax, and decreased the levels of anti-apoptotic proteins, X-IAP and Bcl-2, compared with the levels in the control group. Mdivi1 addition prevented the changes observed in the ZEA-treated cells (Fig. 3I and J). These results identified the potential pathological mechanism of ZEA on ESC apoptosis. ZEA-induced mitochondrial fission may have promoted the destruction of mitochondrial function and structure, which subsequently activated the mitochondrial pathway of apoptosis.

ZEA impairs ESC migration via the activation of mitochondrial fission

A Transwell migration assay was performed to investigate the association between ESC migration and endometriosis progression. As ZEA reduced the number of ESCs through apoptosis induction, ESCs were first treated with ZEA for 24 h. Equal numbers of ESCs (1×105) were collected and placed in the upper chamber of the Transwell plate. After 12 h, the number of ESCs that had migrated to the underside of the insert membranes was counted. As demonstrated in Fig. 4, ZEA treatment for 24 h significantly decreased the migratory ability of ESCs compared with the level observed in the control group. Mitochondrial fission inhibition increased ESC migration, suggesting that mitochondrial fission is involved in the migratory process. As F-actin is the primary stress fiber that is essential for cell mobility (47), it was hypothesized that the decreased migration may be a result of mitochondrial fission-induced F-actin depolymerization. ZEA treatment significantly reduced F-actin fluorescence compared with that observed in the control group, indicating a decrease in F-actin activity (Fig. 4C and D). Fission inhibition by Mdivi1 preserved the filamentary structure of F-actin. Additionally, the decrease in F-actin occurred in parallel with an accumulation of G-actin (Fig. 4E and G), an end-product of F-actin depolymerization (48), suggesting that mitochondrial fission stimulated the dissociation and disrupted the synthesis of F-actin. Mdivi1 reduced the conversion of F-actin to G-actin. These results demonstrate that the ZEA treatment interfered with ESC migration by increasing mitochondrial fission, which influenced the balance of F-actin/G-actin.

ZEA regulates mitochondrial fission by activation of the JNK/Drp1 pathway

JNK and Drp1 levels were analyzed to further elucidate the mechanism of mitochondrial fission regulation by ZEA. Drp1 is a large GTPase that controls mitochondrial fission in mammalian cells, and its active phosphorylation site is a serine at position 616 (Ser616) (40). Drp1 is activated by Ser616 phosphorylation, which may be performed by JNK (39). Therefore, it was speculated that the regulatory effects of ZEA on mitochondrial fission may be due to JNK/Drp1 pathway activation. As demonstrated in Fig. 5, the results revealed that ZEA significantly increased p-JNK levels compared with the levels in the control group, and this increase was inhibited by the JNK pathway inhibitor SP600125 (Fig. 5A and B). JNK inhibition also significantly reduced Drp1 phosphorylation at Ser616 compared with the level observed in the ZEA group, which resulted in a decrease in mitochondrial Drp1 and an increase in cytoplasmic Drp1 compared with that observed in the ZEA group (Fig. 5A, C and E). These data indicate that mitochondrial fission is modulated by the JNK/Drp1 pathway, which in turn is activated by ZEA.

Discussion

Endometriosis is a gynecological disease defined by ectopic endometrial tissue implantation that forms functional endometriotic lesions that are frequently located in the ovaries and peritoneum (49). Endometriosis affects ~10% of women of a reproductive age and may cause severe pelvic pain and infertility (50). It has also been associated with an elevated risk of ovarian cancer (51). The pathogenesis of endometriosis is poorly defined and there are a limited number of effective treatments to cure the disease or slow its progression (52). The relatively high incidence and lack of effective therapeutic options presents a requirement for a more in-depth understanding of the underlying mechanisms that influence the development and severity of endometriosis (53). It has been demonstrated that the increased migration and reduced apoptosis of ESCs contributes to the progression of endometriosis (38). Therefore, methods to reduce the mobility and enhance the apoptosis of ESCs are vital to improve the clinical outcomes of patients with endometriosis.

ZEA, also known as the F-2 toxin, is a non-steroidal mycotoxin produced by several species of Fusarium (54). It is a common fungal contaminant of cereal crops worldwide and is typically found in feed and grains, including maize, wheat and rye (55). ZEA is structurally similar to estrogen and competes with estradiol for binding to estrogen receptors, and stimulates estrogenic activity, which may cause several physiological alterations in the reproductive tract (56). In vitro study has indicated that ZEA may regulate metabolic processes, including cell proliferation, differentiation and apoptosis (57). The regulatory role of ZEA in endometriosis has also been demonstrated (58). In the present study, ZEA was revealed to induce the apoptosis and impair the migration of ESCs through regulation of the JNK/Drp1 pathway and mitochondrial fission.

The mitochondrion is present in all human body cells, excluding erythrocytes. It is the primary organelle for cell metabolism, signal transmission, cellular survival and apoptosis (59). In addition to the vital function of ATP production, it has also been identified that mitochondrial fission is a prerequisite for intrinsic apoptosis in some forms of cancer (60). An increase in mitochondrial length has been demonstrated to decrease mitochondrial fission and increase mitochondrial fusion, leading to the inhibition of apoptotic initiation and the downstream catabolic process of autophagy (6). In the present study, mitochondrial fission was revealed to be responsible for ESC apoptosis via the induction of structural and functional mitochondrial damage. ZEA-induced mitochondrial fission caused mitochondrial depolarization, which resulted in the leakage of cyt-c into the cytoplasm. An increase in pro-apoptotic and a decrease in anti-apoptotic proteins was detected, indicating that the mitochondrial pathway of apoptosis was activated as a consequence of cyt-c release.

Mitochondrial fission has been revealed to result in VDAC1 oligomerization and HK2 separation from the outer mitochondrial membrane, leading to mPTP opening that is responsible for a reduction in ΔΨm (11). Additionally, an increase of mROS in response to mitochondrial fission has been demonstrated to induce cardiolipin peroxidation, which mediates cyt-c release and the activation of the mitochondrial pathway of apoptosis (61). Similarly, the present study identified that mitochondrial fission was responsible for the apoptosis observed, via the induction of structural and functional mitochondrial damage. ZEA-induced mitochondrial fission was also demonstrated to be involved in ESC migration, and mitochondrial fission inhibition reduced ESC migration. These effects were independent of cellular apoptosis.

As F-actin is the primary stress fiber that directly modulates cellular migration, it was hypothesized that decreased migration was the result of a mitochondrial fission-mediated F-actin imbalance (62). The results confirmed that mitochondrial fission resulted in F-actin depolymerization to G-actin, thereby impeding cell migration. Furthermore, successful mitochondrial fission has been revealed to be dependent on Drp1, the Drp1 receptor and stress fibers (63). Previous study has suggested that the brief accumulation of intracellular F-actin on the surface of mitochondria is a prerequisite for subsequent mitochondrial division (7). Under physiological conditions, F-actin is regularly distributed to certain parts of the cytoplasm that control cell migration through directional cues. The initiation of mitochondrial fission transforms F-actin into G-actin at the outer mitochondrial membrane, through the formation of a contractile ring, which involves Drp1 and its receptor (64). Considering the indispensable nature of F-actin in fission, excessive fission would likely consume large amounts of cytoplasmic F-actin and cause uneven F-actin distribution, leading to the dysregulation of F-actin homeostasis and impaired migration. However, further evidence is required to support this hypothesis.

The JNK/Drp1 pathway was demonstrated to be involved in the regulatory effects of ZEA on mitochondrial fission in the present study. Previous study has identified that activated JNK contributes to Drp1 phosphorylation (65). Drp1 is a large GTPase that translocates into puncta on mitochondria, where it couples guanosine 5′-triphosphate hydrolysis with mitochondrial membrane constriction and fission (66). Drp1 activity is determined by its phosphorylation state. Drp1 phosphorylation at Ser616 results in Drp1 activation (67), and activated Drp1 translocates from the cytoplasm to the surface of the mitochondria to mediate mitochondrial fission. The present study demonstrated that ZEA increased p-JNK levels and p-Drp1. Notably, inhibition of JNK prevents this change. ZEA-induced JNK pathway activation was also responsible for an increase in mitochondrially-located Drp1 and a decrease in cytoplasmic Drp1, suggesting that JNK regulated Drp1 activity and subsequent mitochondrial fission.

Collectively, the results of the present study highlight the important role of mitochondrial fission in ESC apoptosis and migration via the JNK/Drp1 pathway. Thus, the regulation of mitochondrial fission may be an effective target for endometriosis therapy. These data also provide evidence for the use of ZEA as an effective method to increase mitochondrial fission. However, further understanding of the underlying mechanisms is required prior to potential clinical research and application.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81030002).

Availability of data and materials

All data generated or analyzed during the present study are included in this published article.

Authors' contributions

HW and XZ conceived the research; CN, YD and YG performed the experiments; all authors participated in discussing and revising the manuscript.

Ethics approval and consent to participate

All experimental protocols were approved by the Ethics Committee of the Department of Gynecology and Obstetrics, Beijing Tongren Hospital (Beijing, China). The ethics reference no. 2015TS1303525.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ricci AG, Olivares CN, Bilotas MA, Meresman GF and Baranao RI: Effect of vascular endothelial growth factor inhibition on endometrial implant development in a murine model of endometriosis. Reprod Sci. 18:614–622. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Crosignani P, Olive D, Bergqvist A and Luciano A: Advances in the management of endometriosis: An update for clinicians. Hum Reprod Update. 12:179–189. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Blitek A, Morawska E, Kiewisz J and Ziecik AJ: Effect of conceptus secretions on HOXA10 and PTGS2 gene expression and PGE2 release in co-cultured luminal epithelial and stromal cells of the porcine endometrium at the time of early implantation. Theriogenology. 76:954–966. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Daiber A, Oelze M, Steven S, Kroller-Schon S and Munzel T: Taking up the cudgels for the traditional reactive oxygen and nitrogen species detection assays and their use in the cardiovascular system. Redox Biol. 12:35–49. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Chen HH, Chen YT, Yang CC, Chen KH, Sung PH, Chiang HJ, Chen CH, Chua S, Chung SY, Chen YL, et al: Melatonin pretreatment enhances the therapeutic effects of exogenous mitochondria against hepatic ischemia-reperfusion injury in rats through suppression of mitochondrial permeability transition. J Pineal Res. 61:52–68. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Jin Q, Li R, Hu N, Xin T, Zhu P, Hu S, Ma S, Zhu H, Ren J and Zhou H: DUSP1 alleviates cardiac ischemia/reperfusion injury by suppressing the Mff-required mitochondrial fission and Bnip3-related mitophagy via the JNK pathways. Redox Biol. 14:576–587. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Shi C, Cai Y, Li Y, Li Y, Hu N, Ma S, Hu S, Zhu P, Wang W and Zhou H: Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways. Redox Biol. 14:59–71. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Zhou H, Zhu P, Guo J, Hu N, Wang S, Li D, Hu S, Ren J, Cao F and Chen Y: Ripk3 induces mitochondrial apoptosis via inhibition of FUNDC1 mitophagy in cardiac IR injury. Redox Biol. 13:498–507. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Zhu H, Jin Q, Li Y, Ma Q, Wang J, Li D, Zhou H and Chen Y: Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca2+]c/VDAC-[Ca2+]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperones. 23:101–113. 2015. View Article : Google Scholar

10 

Zhou H, Li D, Shi C, Xin T, Yang J, Zhou Y, Hu S, Tian F, Wang J and Chen Y: Effects of Exendin-4 on bone marrow mesenchymal stem cell proliferation, migration and apoptosis in vitro. Sci Rep. 5:128982015. View Article : Google Scholar : PubMed/NCBI

11 

Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, Ma Q, Tian F and Chen Y: Mff-dependent mitochondrial fission contributes to the pathogenesis of cardiac microvasculature ischemia/reperfusion injury via induction of mROS-mediated cardiolipin oxidation and HK2/VDAC1 Disassociation-involved mPTP opening. J Am Heart Assoc. 6:e0053282017. View Article : Google Scholar : PubMed/NCBI

12 

Chua S, Lee FY, Chiang HJ, Chen KH, Lu HI, Chen YT, Yang CC, Lin KC, Chen YL, Kao GS, et al: The cardioprotective effect of melatonin and exendin-4 treatment in a rat model of cardiorenal syndrome. J Pineal Res. 61:438–456. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Du K, Ramachandran A and Jaeschke H: Oxidative stress during acetaminophen hepatotoxicity: Sources, pathophysiological role and therapeutic potential. Redox Biol. 10:148–156. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Zhou H, Wang S, Zhu P, Hu S, Chen Y and Ren J: Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol. 15:335–346. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Zhou H, Wang J, Zhu P, Hu S and Ren J: Ripk3 regulates cardiac microvascular reperfusion injury: The role of IP3R-dependent calcium overload, XO-mediated oxidative stress and F-action/filopodia-based cellular migration. Cell Signal. 45:12–22. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Kasahara A and Scorrano L: Mitochondria: From cell death executioners to regulators of cell differentiation. Trends Cell Biol. 24:761–770. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Paterniani E: Selection for reproductive isolation between two populations of maize, zea mays l. Evolution. 23:534–547. 1969. View Article : Google Scholar : PubMed/NCBI

18 

Banjerdpongchai R, Kongtawelert P, Khantamat O, Srisomsap C, Chokchaichamnankit D, Subhasitanont P and Svasti J: Mitochondrial and endoplasmic reticulum stress pathways cooperate in zearalenone-induced apoptosis of human leukemic cells. J Hematol Oncol. 3:502010. View Article : Google Scholar : PubMed/NCBI

19 

Kowalska K, Habrowska-Gorczynska DE, Dominska K and Piastowska-Ciesielska AW: The dose-dependent effect of zearalenone on mitochondrial metabolism, plasma membrane permeabilization and cell cycle in human prostate cancer cell lines. Chemosphere. 180:455–466. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Bouaziz C, El Dein Sharaf O, El Golli E, Abid-Essefi S, Brenner C, Lemaire C and Bacha H: Different apoptotic pathways induced by zearalenone, T-2 toxin and ochratoxin A in human hepatoma cells. Toxicology. 254:19–28. 2008. View Article : Google Scholar : PubMed/NCBI

21 

He B, Zhao Y, Xu L, Gao L, Su Y, Lin N and Pu J: The nuclear melatonin receptor RORalpha is a novel endogenous defender against myocardial ischemia/reperfusion injury. J Pineal Res. 60:313–326. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Agorastos A and Linthorst AC: Potential pleiotropic beneficial effects of adjuvant melatonergic treatment in posttraumatic stress disorder. J Pineal Res. 61:3–26. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Kang JW, Hong JM and Lee SM: Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis. J Pineal Res. 60:383–393. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Lee HY and Back K: Melatonin is required for H2 O2-and NO-mediated defense signaling through MAPKKK3 and OXI1 in Arabidopsis thaliana. J Pineal Res. 62:2017. View Article : Google Scholar

25 

Carrasco-Pozo C, Tan KN, Reyes-Farias M, De La Jara N, Ngo ST, Garcia-Diaz DF, Llanos P, Cires MJ and Borges K: The deleterious effect of cholesterol and protection by quercetin on mitochondrial bioenergetics of pancreatic β-cells, glycemic control and inflammation: In vitro and in vivo studies. Redox Biol. 9:229–243. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Lam SM, Wang Z, Li J, Huang X and Shui G: Sequestration of polyunsaturated fatty acids in membrane phospholipids of Caenorhabditis elegans dauer larva attenuates eicosanoid biosynthesis for prolonged survival. Redox Biol. 12:967–977. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Gao Y, Xiao X, Zhang C, Yu W, Guo W, Zhang Z, Li Z, Feng X, Hao J, Zhang K, et al: Melatonin synergizes the chemotherapeutic effect of 5-fluorouracil in colon cancer by suppressing PI3K/AKT and NF-kappaB/iNOS signaling pathways. J Pineal Res. 62:2017. View Article : Google Scholar

28 

Chen W, Zou P, Zhao Z, Chen X, Fan X, Vinothkumar R, Cui R, Wu F, Zhang Q, Liang G and Ji J: Synergistic antitumor activity of rapamycin and EF24 via increasing ROS for the treatment of gastric cancer. Redox Biol. 10:78–89. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Zhou H, Yang J, Xin T, Zhang T, Hu S, Zhou S, Chen G and Chen Y: Exendin-4 enhances the migration of adipose-derived stem cells to neonatal rat ventricular cardiomyocyte-derived conditioned medium via the phosphoinositide 3-kinase/Akt-stromal cell-derived factor-1α/CXC chemokine receptor 4 pathway. Mol Med Rep. 11:4063–4072. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Kim YD, Hwang SL, Lee EJ, Kim HM, Chung MJ, Elfadl AK, Lee SE, Nedumaran B, Harris RA and Jeong KS: Melatonin ameliorates alcohol-induced bile acid synthesis by enhancing miR-497 expression. J Pineal Res. 62:2017.doi: 10.1111/jpi.12386. View Article : Google Scholar

31 

Cai SY, Zhang Y, Xu YP, Qi ZY, Li MQ, Ahammed GJ, Xia XJ, Shi K, Zhou YH, Reiter RJ, et al: HsfA1a upregulates melatonin biosynthesis to confer cadmium tolerance in tomato plants. J Pineal Res. 62:2017.doi: 10.1111/jpi.12387. View Article : Google Scholar : PubMed/NCBI

32 

Mailloux RJ and Treberg JR: Protein S-glutathionlyation links energy metabolism to redox signaling in mitochondria. Redox Biol. 8:110–118. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Zhou H, Yang J, Xin T, Li D, Guo J, Hu S, Zhou S, Zhang T, Zhang Y, Han T and Chen Y: Exendin-4 protects adipose-derived mesenchymal stem cells from apoptosis induced by hydrogen peroxide through the PI3K/Akt-Sfrp2 pathways. Free Radic Biol Med. 77:363–375. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Zhang Y, Zhou H, Wu W, Shi C, Hu S, Yin T, Ma Q, Han T, Zhang Y, Tian F and Chen Y: Liraglutide protects cardiac microvascular endothelial cells against hypoxia/reoxygenation injury through the suppression of the SR-Ca(2+)-XO-ROS axis via activation of the GLP-1R/PI3K/Akt/survivin pathways. Free Radic Biol Med. 95:278–292. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Feng D, Wang B, Wang L, Abraham N, Tao K, Huang L, Shi W, Dong Y and Qu Y: Pre-ischemia melatonin treatment alleviated acute neuronal injury after ischemic stroke by inhibiting endoplasmic reticulum stress-dependent autophagy via PERK and IRE1 signalings. J Pineal Res. 62:2017.doi: 10.1111/jpi.12395. View Article : Google Scholar

36 

Areti A, Komirishetty P, Akuthota M, Malik RA and Kumar A: Melatonin prevents mitochondrial dysfunction and promotes neuroprotection by inducing autophagy during oxaliplatin-evoked peripheral neuropathy. J Pineal Res. 62:2017.doi: 10.1111/jpi.12393. View Article : Google Scholar : PubMed/NCBI

37 

Fuhrmann DC and Brune B: Mitochondrial composition and function under the control of hypoxia. Redox Biol. 12:208–215. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Doskey CM, Buranasudja V, Wagner BA, Wilkes JG, Du J, Cullen JJ and Buettner GR: Tumor cells have decreased ability to metabolize H2O2: Implications for pharmacological ascorbate in cancer therapy. Redox Biol. 10:274–284. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Zhou H, Du W, Li Y, Shi C, Hu N, Ma S, Wang W and Ren J: Effects of melatonin on fatty liver disease: The role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission and mitophagy. J Pineal Res. 64:2018. View Article : Google Scholar

40 

Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F and Chen Y: Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res. 63:2017.doi: 10.1111/jpi.12413. View Article : Google Scholar :

41 

Brasacchio D, Alsop AE, Noori T, Lufti M, Iyer S, Simpson KJ, Bird PI, Kluck RM, Johnstone RW and Trapani JA: Epigenetic control of mitochondrial cell death through PACS1-mediated regulation of BAX/BAK oligomerization. Cell Death Differ. 24:961–970. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Zhang R and Sun Y, Liu Z, Jin W and Sun Y: Effects of melatonin on seedling growth, mineral nutrition and nitrogen metabolism in cucumber under nitrate stress. J Pineal Res. 62:2017.doi: 10.1111/jpi.12403. View Article : Google Scholar

43 

Banerjee K, Keasey MP, Razskazovskiy V, Visavadiya NP, Jia C and Hagg T: Reduced FAK-STAT3 signaling contributes to ER stress-induced mitochondrial dysfunction and death in endothelial cells. Cell Signal. 36:154–162. 2017. View Article : Google Scholar : PubMed/NCBI

44 

Dufour F, Rattier T, Shirley S, Picarda G, Constantinescu AA, Morlé A, Zakaria AB, Marcion G, Causse S, Szegezdi E, et al: N-glycosylation of mouse TRAIL-R and human TRAIL-R1 enhances TRAIL-induced death. Cell Death Differ. 24:500–510. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Murphy PS, Wang J, Bhagwat SP, Munger JC, Janssen WJ, Wright TW and Elliott MR: CD73 regulates anti-inflammatory signaling between apoptotic cells and endotoxin-conditioned tissue macrophages. Cell Death Differ. 24:559–570. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Park J, Tran Q, Mun K, Masuda K, Kwon SH, Kim SH, Kim DH, Thomas G and Park J: Involvement of S6K1 in mitochondria function and structure in HeLa cells. Cell Signal. 28:1904–1915. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Van Nostrand JL, Bowen ME, Vogel H, Barna M and Attardi LD: The p53 family members have distinct roles during mammalian embryonic development. Cell Death Differ. 24:575–579. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Perdiz D, Lorin S, Leroy-Gori I and Pous C: Stress-induced hyperacetylation of microtubule enhances mitochondrial fission and modulates the phosphorylation of Drp1 at 616Ser. Cell Signal. 39:32–43. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Xu S, Pi H, Zhang L, Zhang N, Li Y, Zhang H, Tang J, Li H, Feng M, Deng P, et al: Melatonin prevents abnormal mitochondrial dynamics resulting from the neurotoxicity of cadmium by blocking calcium-dependent translocation of Drp1 to the mitochondria. J Pineal Res. 60:291–302. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Laschke MW and Menger MD: Anti-angiogenic treatment strategies for the therapy of endometriosis. Hum Reprod Update. 18:682–702. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Lagana AS, Vitale SG, Salmeri FM, Triolo O, Ban Frangež H, Vrtačnik-Bokal E, Stojanovska L, Apostolopoulos V, Granese R and Sofo V: Unus pro omnibus, omnes pro uno: A novel, evidence-based, unifying theory for the pathogenesis of endometriosis. Med Hypotheses. 103:10–20. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Kowalska K, Habrowska-Gorczynska DE and Piastowska-Ciesielska AW: Zearalenone as an endocrine disruptor in humans. Environ Toxicol Pharmacol. 48:141–149. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Smith MC, Madec S, Coton E and Hymery N: Natural co-occurrence of mycotoxins in foods and feeds and their in vitro combined toxicological effects. Toxins. 8:942016. View Article : Google Scholar : PubMed/NCBI

54 

Danicke S and Winkler J: Invited review: Diagnosis of zearalenone (ZEN) exposure of farm animals and transfer of its residues into edible tissues (carry over). Food Chem Toxicol. 84:225–249. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Mally A, Solfrizzo M and Degen GH: Biomonitoring of the mycotoxin Zearalenone: Current state-of-the art and application to human exposure assessment. Arch Toxicol. 90:1281–1292. 2016. View Article : Google Scholar : PubMed/NCBI

56 

Hu J, Xu M, Dai Y, Ding X, Xiao C, Ji H and Xu Y: Exploration of Bcl-2 family and caspases-dependent apoptotic signaling pathway in Zearalenone-treated mouse endometrial stromal cells. Biochem Biophys Res Commun. 476:553–559. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Stopa E, Babinska I, Zielonka L, Gajecki M and Gajecka M: Immunohistochemical evaluation of apoptosis and proliferation in the mucous membrane of selected uterine regions in pre-pubertal bitches exposed to low doses of zearalenone. Pol J Vet Sci. 19:175–186. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Ellenrieder L, Rampelt H and Becker T: Connection of protein transport and organelle contact sites in mitochondria. J Mol Biol. 429:2148–2160. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Senft D and Ronai ZA: Regulators of mitochondrial dynamics in cancer. Curr Opin Cell Biol. 39:43–52. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Ribas V, Garcia-Ruiz C and Fernandez-Checa JC: Mitochondria, cholesterol and cancer cell metabolism. Clin Transl Med. 5:222016. View Article : Google Scholar : PubMed/NCBI

61 

Nomura K, Imai H, Koumura T, Kobayashi T and Nakagawa Y: Mitochondrial phospholipid hydroperoxide glutathione peroxidase inhibits the release of cytochrome c from mitochondria by suppressing the peroxidation of cardiolipin in hypoglycaemia-induced apoptosis. Biochem J. 351:183–193. 2000. View Article : Google Scholar : PubMed/NCBI

62 

Prieto-Dominguez N, Ordonez R, Fernandez A, Méndez-Blanco C, Baulies A, Garcia-Ruiz C, Fernández-Checa JC, Mauriz JL and González-Gallego J: Melatonin-induced increase in sensitivity of human hepatocellular carcinoma cells to sorafenib is associated with reactive oxygen species production and mitophagy. J Pineal Res. 61:396–407. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Zhou H, Ma Q, Zhu P, Ren J, Reiter RJ and Chen Y: Protective role of melatonin in cardiac ischemia-reperfusion injury: From pathogenesis to targeted therapy. J Pineal Res. 2018.doi: 10.1111/jpi.12471. View Article : Google Scholar

64 

Estaquier J and Arnoult D: Inhibiting Drp1-mediated mitochondrial fission selectively prevents the release of cytochrome c during apoptosis. Cell Death Differ. 14:1086–1094. 2007. View Article : Google Scholar : PubMed/NCBI

65 

Bereiter-Hahn J, Voth M, Mai S and Jendrach M: Structural implications of mitochondrial dynamics. Biotechnol J. 3:765–780. 2008. View Article : Google Scholar : PubMed/NCBI

66 

Lin C, Chao H, Li Z, Xu X, Liu Y, Hou L, Liu N and Ji J: Melatonin attenuates traumatic brain injury-induced inflammation: A possible role for mitophagy. J Pineal Res. 61:177–186. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Yang X, Wang H, Ni HM, Xiong A, Wang Z, Sesaki H, Ding WX and Yang L: Inhibition of Drp1 protects against senecionine-induced mitochondria-mediated apoptosis in primary hepatocytes and in mice. Redox Biol. 12:264–273. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2018
Volume 17 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang H, Zhao X, Ni C, Dai Y and Guo Y: Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway. Mol Med Rep 17: 7797-7806, 2018
APA
Wang, H., Zhao, X., Ni, C., Dai, Y., & Guo, Y. (2018). Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway. Molecular Medicine Reports, 17, 7797-7806. https://doi.org/10.3892/mmr.2018.8823
MLA
Wang, H., Zhao, X., Ni, C., Dai, Y., Guo, Y."Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway". Molecular Medicine Reports 17.6 (2018): 7797-7806.
Chicago
Wang, H., Zhao, X., Ni, C., Dai, Y., Guo, Y."Zearalenone regulates endometrial stromal cell apoptosis and migration via the promotion of mitochondrial fission by activation of the JNK/Drp1 pathway". Molecular Medicine Reports 17, no. 6 (2018): 7797-7806. https://doi.org/10.3892/mmr.2018.8823