Uterine leiomyoma is the most common benign smooth muscle tumor of uterus in women of reproductive age, with a high lifetime incidence. Nowadays, the exploration on the pharmacotherapies, such as progesterone receptor antagonist (PRA) requires more attention. Hence, the current study aimed to examine whether mifepristone, a PRA, influences the autophagy and apoptosis of uterine leiomyoma cells. Primary uterine leiomyoma cells were collected from 36 patients diagnosed with uterine leiomyoma to establish PR-M-positive (PR-M[+]) cells. The lentiviral vector overexpressing or silencing PR-M was subsequently delivered into one part of PR-M(+) cells in order to evaluate the role of PR-M in PR-M(+) cells. The results obtained revealed that cell viability was increased, while cell autophagy and apoptosis were diminished in the PR-M(+) cells treated with overexpressed PR-M, whereby the Bcl-2 level was elevated and the level of Beclin1 was reduced. An opposite trends were identified following treatment with knockdown of PR-M. Mifepristone at different concentrations (low, moderate, or high) was then applied to treat another part of the PR-M(+) cells. Mifepristone was identified to promote cell autophagy and apoptosis, decrease Bcl-2 level and increase Beclin1 level, accompanied by weakened interaction between Bcl-2 and Beclin1. Moreover, these effects of mifepristone on PR-M(+) cells were enhanced with increasing of the concentration. Taken together, the present study present evidence indicates the ability of PRA to regulate the Bcl-2/Beclin1 axis, ultimately promoting the autophagy and apoptosis of uterine leiomyoma cells, highlighting that PRA serves as a promising therapeutic target for the treatment of uterine leiomyoma.

Uterine leiomyoma, also commonly referred to as uterine fibroids, remains the most common type of benign tumor afflicting the smooth muscle of the uterus, with the lifetime prevalence exceeding approximately 70% in the general population [1]. Uterine leiomyoma is characterized by heavy or prolonged menstrual bleeding and resultant anemia generally affecting women of reproductive age, which may significant affect the quality of life and fertility [2]. At present, surgeries (e.g. hysterectomy and myomectomy by hysteroscopy or laparoscopy) and pharmacotherapies (e.g. oral contraceptives, progestins, and GnRH analogs) represent the first-line therapeutic approaches for patients diagnosed with uterine leiomyoma [3,4]. Emerging evidence has demonstrated that progesterone and its receptors (PRs) may act to promote the uterine leiomyomata cell viability [5]. Although surgical intervention has been reported as an effective measure, it is an approach that is financially damaging for most patients [3]. Therefore, in order to find an alternative measure to treat uterine leiomyoma and reduce the need for invasive surgery, treatments with anti-progestins and selective PR modulators (SPRMs), like progesterone receptor antagonist (PRA), require further investigation.

As a steroid hormone, progesterone represents a crucial element involved in the coordination of the reproductive physiology of normal mammalian female, which is predominately secreted by the corpus luteum developed in the ovulated ovary. Ovarian progesterone represents an essential regulator of uterine function, which elicits effects on numerous tissues and organs, including the breast uterus, as well as the ovary [6]. Evidence has been presented indicating that volume maintenance and growth of uterine leiomyoma are progesterone dependent events [7]. The action of progesterone on the female reproductive system is largely mediated via PR which is synthesized from a single gene and expressed as two main protein isoforms (PR-A and PR-B) [8]. Moreover, PRs, involving in the actions of the ovarian steroid progesterone, play a critical role in gonadotropin secretion, endometrium implantation, pregnancy maintenance, and breast tissue differentiation [9]. One truncated progesterone receptor (PR-M) only encodes exons 4 with 16 unique N-terminal amino acids and is involved in some diseases [10]. Two classical isoform of progesterone receptors, including PR-A and B, are integrate pattern, in which isoform A represents ligand-dependent transdominant repressor, while isoform B represents transcriptional activator of some hormone-related promoters [11,12]. However, little evidence determines the short receptor, PR-M, exhibit similar physiological function of PR-A or PR-B. Recent studies have demonstrated that PR-M localizes to outer membrane of mitochondrion and is a regulatory participant in cellular respiration [10]. A progesterone-dependent elevation in cellular respiration via PR-M is presented. Furthermore, a large number of studies have highlighted the role of progesterone and PR in various types of cancers [13]. PR-M has been identified in breast cancer cells and has a progesterone-dependent effect on proliferation of cancer cell [10,14]. In addition to its involvement in cancer diseases, a recent investigation indicated that PR-M is strongly associated with heart failure in transgenic mouse models. The aforementioned literature suggests that PR-M is a metabolic regulator capable of regulating cellular respiration. The role of SPRMs in the treatment of uterine leiomyoma has been evidenced in previous reports, highlighting SPRMs as an integrator of proliferation in uterine leiomyoma cells with potential benefit to those seeking to preserve future fertility [15,16]. Based on the Clinical Trial database (http://clinicaltrials.gov) (the US National Institutes of Health), several treatment options are under clinical trial, including mifepristone (MIF), asoprisnil, ulipristal, aromatase inhibitors, epigallocatechin gallate, and pirfenidone [17]. A prospective randomized placebo controlled trial has reported that MIF, a PRA, may offer an effective therapeutic option for women with uterine leiomyoma [18]. Recent reports have implicated PRs in the development of certain cancers via the modulation of the apoptosis-related factor Bcl-2 or autophagy-related factor Beclin1 [19–21]. In the current study, we employ MIF at varying concentrations to treat primary uterine leiomyoma cells isolated from patients diagnosed with uterine leiomyoma, in order to determine whether PRA could be a therapeutic option for the treatment of uterine leiomyoma. Hence, we examined the hypothesis that the underlying mechanism may be related to the Bcl-2/Beclin1 axis.

Study subjects

A total of 36 patients diagnosed with uterine leiomyoma, who had previously undergone a myomectomy by laparotomy at the Third Affiliated Hospital of Zhengzhou University (Zhengzhou, China) from July 2016 to January 2018 were enrolled for the present study. The enrolled patients were aged from 31 to 50 years old, with a mean age of 40.56 ± 6.41 years. All enrolled patients were confirmed not to be suffering from other hormone related diseases and had not received any steroid hormone medications at least 6 months before surgery. All specimens were diagnosed by pathological examination. The present study was conducted with the approval of the Ethics Committee of the Third Affiliated Hospital of Zhengzhou University (Zhengzhou, China) and in strict adherence with the Helsinki Declaration. Written informed consent was obtained from each patient prior to enrollment and collection of their respective primary uterine leiomyoma cells as well as endometrium biopsy samples.

Selection of the primary PR-M-positive uterine leiomyoma cells

The collected specimens were rinsed with phosphate buffer solution (PBS) containing penicillin/streptomycin for 3–5 min, and then treated with 0.2% type I collagen at 37°C for 2–3 h. The solution was then filtered, centrifuged at 1000 r/min for 10 min with supernatant discarded, followed by incubation with 10% complete medium in 5% CO2 at 37°C for 24 h. The cells observed to have adhered were passaged using 0.25% trypsin–EDTA solution, when cell confluence reached 80%. Cellular morphology was analyzed under an inverted phase contrast microscope (DM500/DM750, Leica, Wetzlar, Germany). The cells were subsequently identified by immunohistochemistry means. The cells at the third passage were then inoculated into six-well plates. When cells reached 80–90% confluence, the total protein was extracted and analyzed by Western blot analysis in order to identify the PR-M-positive (PR-M [+]) cells and PR-A/B-negative (PR-A/B [−]) cells.

Immunohistochemistry

The PR-M(+) cells were trypsinized and inoculated in six-well plates. Upon reaching 70–80% confluence, the cells were fixed in 4% paraformaldehyde for 30 min and blocked using normal goat serum for 1 h at room temperature. After the removal of goat serum, the cells were incubated with primary antibody anti-smooth muscle Actin (ab5694, 1:200, Abcam, Cambridge, U.K.) at 4°C overnight. The cells were then incubated with goat anti-rabbit secondary antibody immunoglobulin G (IgG) (1:5000, ab6721, Abcam, Cambridge, U.K.) at room temperature for 2 h under conditions void of light. The cells were then developed using 3,3′ diaminobenzidine (DAB; Zhongshan Golden Bridge Biotechnology Co., Ltd., Beijing, China) and analyzed under a microscope. The cells were subsequently counterstained with hematoxylin for 10 s.

Cell treatment

The lentivirus vectors constructed by Shanghai GenePharma Company (Shanghai, China) included pLV-EGFP-N (gene overexpression vector) and pSIH1-H1-copGFP (gene silencing vector). The lentivirus particles expressing PR-M and Bcl-2, and containing the allele specific shRNAs for PR-M were constructed by co-transfection of 293T cells with packaged and enveloped plasmids in a RPMI 1640 complete medium containing 10% FBS. Finally, the lentiviral particles (1 × 108 TU/ml) were transduced into the PR-M(+) cells, which were then made into cell suspension (5 × 104cells/ml) and inoculated in 6-well plates. After 24 h, the green fluorescence protein (GFP) expression efficiency was identified under a fluorescence microscope. The remaining PR-M(+) cells were then incubated in 96-well plates in 5% CO2 at 37°C. The cells were then treated with PRA mifepristone (MIF, Hy-Clone, Logan, Utah, U.S.A.) at different concentrations: 0 mol/l, 10−6 mol/l (low MIF, L-MIF), 10−5 mol/l (moderate MIF, M-MIF), and 10−4 mol/l (high MIF, H-MIF). The PR-M(+) cells were then infected with pLV-EGFP expressing Bcl-2 and treated with H-MIF.

Co-immunoprecipitation

The PR-M(+) cells were lysed in a lysis buffer containing proteinase (Hoffmann-La Roche Ltd, Basel, Switzerland) and phosphatase (Thermo Scientific, Inc., Waltham, Massachusetts, U.S.A.) inhibitor at 4°C for 30 min. After the cell lysates had been centrifuged at 16000 g at 4°C for 30 min, the supernatant was pre-treated with 60 μl protein G agarose for 2 h and incubated overnight with Bcl-2-agarose (1:1000, sc-7382, Santa Cruz Biotechnology, Santa Cruz, CA, U.S.A.), or with IgG (1:5000, ab6789, Abcam). The mixture was then re-suspended with 2 × sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS/PAGE) loading buffer and boiled for 5 min. Finally, the protein level of Beclin1 was determined by Western blot analysis.

Western blot analysis

The PR-M(+) cells were lysed in a lysis buffer containing proteinase and phosphatase inhibitor at 4°C for 30 min and centrifuged at 10,000 r/min for 15 min. The cell lysates were then separated using 10% PAGE followed by transfer onto a polyvinylidene fluoride (PVDF) membrane. Membrane was blocked using 5% skim milk for 1 h at room temperature, followed by incubation at 4°C overnight with Tris-buffer saline with Tween (TBST)-diluted primary antibodies: anti-p62 (1:1000, ab56416, Abcam), anti-LC3B (1:10000, L7543, Sigma–Aldrich, St. Louis, MO, U.S.A.), anti-Bcl-2 (1:1000, sc-7382, Santa Cruz), anti-Beclin1 (1:500, sc-48341, Santa Cruz), anti-β-actin (1:5000, sc-47778, Santa Cruz) as an internal control, anti-PR (A-2) (1:5000, sc-398898, Santa Cruz), anti-PR-M (C262; 1:1000, sc-53943, Santa Cruz), anti-PR-B (B-30; 1:2000, sc-811, Santa Cruz) and anti-caspase-3 (1:1000, ab2302, Abcam). The membrane was then incubated with the horseradish peroxidase (HRP)-conjugated secondary antibodies at room temperature for 1 h: goat anti-mouse IgG (1:5000, ab6789, Abcam) or anti-rabbit (1:5000, ab6721, Abcam). Finally, the membrane was washed six times with TSBT and developed using enhanced chemiluminescent (ECL), with the gray value of the protein bands quantified by ImageJ 1.48u software (Bio-Rad, Hercules, CA, U.S.A.). The experiments were repeated three times.

Monodansylcadaverine staining for cell autophagy

The PR-M(+) cells were incubated on coverslips coated with polylysine. The autophagosomes were subsequently labeled with 20 μM MDC for 30 min and analyzed under a fluorescence microscope. The fluorescence of MDC was observed to be in a punctate distribution within the cytoplasm when the autophagosome was formed. Image J software was applied for quantitative fluorescence analysis.

Cell counting kit-8 assay for cell viability

The PR-M(+) cells were treated with 0.25% trypsin and constructed into the single cell suspension with the density adjusted to 5 × 104 cells/ml. The cell suspension was then inoculated into 96-well plates (100 μl/well) and cultured in 5% CO2 at 37°C for 24 and 48 h, respectively, with five duplicated wells set for each group. After a 24- or 48-h incubation, each well was incubated with 100 μl of CCK-8 reagent (Beyotime Biotechnology, Shanghai, China) for a period of 1 h. Following the termination of the incubation process, a microplate reader (BD Bioscience, San Jose, CA, U.S.A.) was employed to determine the absorbance value of each well at 490 nm.

Flow cytometry

The PR-M(+) cells were treated with 0.5 ml of 0.25% trypsin and subsequently constructed into a cell suspension with the density adjusted to 1 × 106 cells/ml. The cell suspension (0.5 μl) was stained with 5 μl Annexin V-FITC and 10 μl propidium iodide (PI) (Beyotime, Biotechnology, Shanghai, China) under conditions void of light at room temperature for 15 min. Flow cytometry (BD Bioscience, U.S.A.) were subsequently applied to determine cell apoptosis at 488 nm. The experiment was repeated three times.

Statistical analysis

Statistical analysis was performed using the SPSS 21.0 software (IBM Corp. Armonk, NY, U.S.A.). The measurement data were expressed as mean ± standard deviation, with equality of variances and normal distribution tested. If the data subjected to normal distribution and equality of variances, comparisons among multiple groups were analyzed by one-way analysis of variance (ANOVA) or repeated-measures ANOVA, and the post hoc test was used for the pairwise comparison within a group. For the data with skew distribution or unequal variances, the Wilcoxon rank sum test was performed. A P-value < 0.05 was considered to be significantly different.

Successful establishment of PR-M(+) cells

The first step in our experiment was to establish the primary PR-M(+) uterine leiomyoma cells isolated from patients with uterine leiomyoma. Next, cellular morphology was observed and identified under a high-power microscope following immunohistochemistry. As depicted in Figure 1A, the cells were identified as uterine leiomyoma cells. The protein levels of PR-M, PR-A and PR-B were detected using Western blot analysis (Figure 1B), which revealed that in the PR-M(+) cells, PR-M was highly expressed and PR-A/B was not expressed. The normal endometrial stromal cells served as the control, with our investigations revealing that the PRA/B antibody was effective [22]. The results obtained were indicative of the successful establishment of primary uterine leiomyoma cells and PR-M(+) cells.

The PR-M(+) cells are successfully established

Figure 1
The PR-M(+) cells are successfully established

(A), Cell morphology observation under an inverted phase contrast microscope (a, 100×, scale bar = 100 μm; b, 200×, scale bar = 50 μm). (B), Protein levels of PR-M and PR-A/B tested using Western blot analysis.

Figure 1
The PR-M(+) cells are successfully established

(A), Cell morphology observation under an inverted phase contrast microscope (a, 100×, scale bar = 100 μm; b, 200×, scale bar = 50 μm). (B), Protein levels of PR-M and PR-A/B tested using Western blot analysis.

Close modal

PR-M inhibits the PR-M(+) cell autophagy and apoptosis

Following confirmation of the successful establishment of the PR-M(+) cells, we further investigated the effect of PR-M on cell autophagy and apoptosis of PR-M(+) cells. Initially, the PR-M(+) cells were treated with overexpressed PR-M treatment and then interference treatment. Next, reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis were conducted to identify the PR-M expression, the results of which revealed there to be no significant difference in the mRNA and protein expression of PR-M in PR-M(+) cells without treatment and that of the cells treated with sh-NC and oe-NC (P>0.05). In comparison with PR-M(+) cells treated with sh-NC, the mRNA and protein expression of PR-M were markedly decreased after treatment of sh-PR-M, while a notable increase was detected following treatment with oe-PR-M, relative to the oe-NC treatment (P<0.05) (Figure 2A,B). Next, CCK-8 assay, MDC staining assay, Western blot analysis and flow cytometry were adopted to detect the cell viability, cell autophagy, expression of autophagy-related proteins (p62, LC3-I, and LC3-II) and cell apoptosis, respectively as illustrated in Figure 2C displayed, the PR-M(+) cells with silenced PR-M exhibited down-regulated cell viability, which displayed an increased trend in those with overexpressed PR-M (P<0.05). The MDC staining results (Figure 2D) revealed that the punctate green fluorescence intensity around nucleus was enhanced in the PR-M(+) cells following PR-M knockdown treatment but reduced after PR-M overexpression treatment. The subsequent Western blot analysis (Figure 2E) revealed that silencing of PR-M down-regulated the p62 protein level and up-regulated the LC3-II/(LC3-I + LC3-II) level, which manifested with a reversed tendency following the overexpression of PR-M (all P<0.05). Subsequently, flow cytometry, combined with Western blot analysis, demonstrated that PR-M knockdown promoted cell apoptosis rate and expression of activated Caspase-3, while PR-M up-regulation was found to decrease the rate of cell apoptosis of PR-M(+) cells, accompanied with reduced expression of activated Caspase-3 (P<0.05, Figure 2F,G). The results obtained suggested that PR-M could exert an inhibitory role in the autophagy and apoptosis of primary uterine leiomyoma cells, which might be promoted by PR-M down-regulation.

The autophagy and apoptosis of PR-M(+) cells are suppressed by PR-M

Figure 2
The autophagy and apoptosis of PR-M(+) cells are suppressed by PR-M

The PR-M(+) cells used for following detection were cells without treatment or cells treated with sh-PR-M and oe-PR-M and their controls. (A), The mRNA expression of PR-M in cells with different treatments detected by RT-qPCR. (B), The protein expression of PR-M in cells with different treatments examined by Western blot analysis, with protein bands assessed. (C), The optical density (OD) value of cells at different time points determined using CCK-8 assay. (D), Cell autophagy observed using monodansylcadaverine staining (500×, scar bar = 20 μm), and the quantitative fluorescence analysis. (E), Protein levels of autophagy-related proteins determined using Western blot analysis, with protein bands assessed. (F), Cell apoptosis determined using flow cytometry. (G), The expression of activated Caspase-3 in cells detected by Western blot analysis, with protein bands assessed. Blank, PR-M(+) cells without treatment; sh-NC, PR-M(+) cells infected with pSIH1-H1-copGFP expressing irrelevant shRNA used as negative control; sh-PR-M, PR-M(+) cells infected with pSIH1-H1-copGFP expressing shRNA for PR-M; oe-NC, PR-M(+) cells infected with empty pLV-EGFP used as negative control; oe-PR-M, PR-M(+) cells infected with pLV-EGFP expressing PR-M. *P<0.05, vs cells with sh-NC treatment; #P<0.05, vs cells with oe-NC treatment. The measurement data were expressed as mean ± standard error, and were anzlyed by one-way ANOVA. The experiment was repeated three times.

Figure 2
The autophagy and apoptosis of PR-M(+) cells are suppressed by PR-M

The PR-M(+) cells used for following detection were cells without treatment or cells treated with sh-PR-M and oe-PR-M and their controls. (A), The mRNA expression of PR-M in cells with different treatments detected by RT-qPCR. (B), The protein expression of PR-M in cells with different treatments examined by Western blot analysis, with protein bands assessed. (C), The optical density (OD) value of cells at different time points determined using CCK-8 assay. (D), Cell autophagy observed using monodansylcadaverine staining (500×, scar bar = 20 μm), and the quantitative fluorescence analysis. (E), Protein levels of autophagy-related proteins determined using Western blot analysis, with protein bands assessed. (F), Cell apoptosis determined using flow cytometry. (G), The expression of activated Caspase-3 in cells detected by Western blot analysis, with protein bands assessed. Blank, PR-M(+) cells without treatment; sh-NC, PR-M(+) cells infected with pSIH1-H1-copGFP expressing irrelevant shRNA used as negative control; sh-PR-M, PR-M(+) cells infected with pSIH1-H1-copGFP expressing shRNA for PR-M; oe-NC, PR-M(+) cells infected with empty pLV-EGFP used as negative control; oe-PR-M, PR-M(+) cells infected with pLV-EGFP expressing PR-M. *P<0.05, vs cells with sh-NC treatment; #P<0.05, vs cells with oe-NC treatment. The measurement data were expressed as mean ± standard error, and were anzlyed by one-way ANOVA. The experiment was repeated three times.

Close modal

PR-M inhibits the PR-M(+) cell autophagy and apoptosis via regulation of Bcl-2/Beclin1 axis

As the inhibitory role of PR-M in cell autophagy and apoptosis of PR-M(+) cells was demonstrated in the former experiments, we subsequently clarified whether the Bcl-2/Beclin1 axis is involved this process. Initially, Western blot analysis was employed to determine the protein level of Bcl-2 and Beclin1 (Figure 3A). The results revealed that PR-M depletion decreased protein level of Bcl-2, and elevated the protein level of Beclin1 (P<0.05). In contrast, protein level of Bcl-2 was elevated in PR-M(+) cells with PR-M up-regulation, along with reduced protein level of Beclin1 (P<0.05). Second, a Co-immunoprecipitation (Co-IP) assay was performed to verify the interaction between Bcl-2 and Beclin1 in PR-M(+) cells. As depicted in Figure 3B, in the PR-M(+) cells infected with pSIH1-H1-copGFP expressing shRNA for PR-M, the interaction between Bcl-2 and Beclin1 was weakened, which was enhanced in those infected with pLV-EGFP expressing PR-M. Collectively, these results suggested that the Bcl-2/Beclin1 axis might involve in the inhibitory effect of PR-M on autophagy and apoptosis of primary uterine leiomyoma cells.

The cell autophagy and apoptosis of PR-M(+) cells are inhibited by PR-M via the Bcl-2/Beclin1 axis

Figure 3
The cell autophagy and apoptosis of PR-M(+) cells are inhibited by PR-M via the Bcl-2/Beclin1 axis

The PR-M(+) cells used for following detections were cells without treatment or cells treated with sh-PR-M and oe-PR-M and their controls. (A), Protein level of Bcl-2 and Beclin1 measured by Western blot analysis. (B), Interaction between Bcl-2 and Beclin1 verified by CO-IP assay. Blank, PR-M(+) cells without treatment; sh-NC, PR-M(+) cells infected with pSIH1-H1-copGFP expressing irrelevant shRNA used as negative control; sh-PR-M, PR-M(+) cells infected with pSIH1-H1-copGFP expressing shRNA for PR-M; oe-NC, PR-M(+) cells infected with empty pLV-EGFP used as negative control; oe-PR-M, PR-M(+) cells infected with pLV-EGFP expressing PR-M. *P<0.05, vs cells treated with sh-NC; #P<0.05, vs cells treated with oe-NC. The measurement data were expressed as mean ± standard error, and were analyzed by one-way ANOVA. The experiment was repeated three times.

Figure 3
The cell autophagy and apoptosis of PR-M(+) cells are inhibited by PR-M via the Bcl-2/Beclin1 axis

The PR-M(+) cells used for following detections were cells without treatment or cells treated with sh-PR-M and oe-PR-M and their controls. (A), Protein level of Bcl-2 and Beclin1 measured by Western blot analysis. (B), Interaction between Bcl-2 and Beclin1 verified by CO-IP assay. Blank, PR-M(+) cells without treatment; sh-NC, PR-M(+) cells infected with pSIH1-H1-copGFP expressing irrelevant shRNA used as negative control; sh-PR-M, PR-M(+) cells infected with pSIH1-H1-copGFP expressing shRNA for PR-M; oe-NC, PR-M(+) cells infected with empty pLV-EGFP used as negative control; oe-PR-M, PR-M(+) cells infected with pLV-EGFP expressing PR-M. *P<0.05, vs cells treated with sh-NC; #P<0.05, vs cells treated with oe-NC. The measurement data were expressed as mean ± standard error, and were analyzed by one-way ANOVA. The experiment was repeated three times.

Close modal

PRA promotes the PR-M(+) cell autophagy and apoptosis

With the results in the above section detailing the inhibitory role of PR-M in the PR-M(+) cell autophagy and apoptosis, we examined the hypothesis that PRA may have a reverse effect. In order to verify this, we treated the PR-M(+) cells using MIF at different concentrations, which is a type of PRA. Next, cell viability was evaluated using a CCK-8 assay (Figure 4A). Our results revealed that cell viability was significantly decreased in PR-M(+) cells with MIF treatment, which was decreased most obviously in those with H-MIF. Next, MDC staining revealed that MIF enhanced the punctate green fluorescence intensity around nucleus, which exhibited the most significant increase following H-MIF treatment (Figure 4B). Western blot analysis was subsequently employed to further determine the protein level of autophagy-related proteins (p62, LC3-I, and LC3-II). The results obtained (Figure 4C) indicated that L-MIF, M-MIF, and H-HIF down-regulated the protein level of p62 and up-regulated the protein level of LC3-II/(LC3-I + LC3-II), among which H-MIF induced the lowest protein level of p62 and highest protein level of LC3-II/(LC3-I + LC3-II) (P<0.05). What is more, in order to evaluate the effect of MIF on cell apoptosis of PR-M(+) cells and expression of activated Caspase-3, flow cytometry and Western blot analysis were performed, which demonstrated that PR-M(+) cells treated with L-MIF, M-MIF, and H-MIF exhibited increased cell apoptosis rate and elevated expression of activated caspase-3, among which the PR-M(+) cells with H-MIF showed highest cell apoptosis rate as well as the expression of activated caspase-3 (P<0.05, Figure 4D,E). Above findings highlighted a facilitation effect of PRA in autophagy and apoptosis of primary uterine leiomyoma cells, which might be enhanced with increasing of the concentration.

The cell autophagy and apoptosis of PR-M(+) cells are promoted by PRA

Figure 4
The cell autophagy and apoptosis of PR-M(+) cells are promoted by PRA

The PR-M(+) cells used for following detection were cells without treatment or cells treated with L-MIF, M-MIF, and H-MIF. (A), OD value of cells at different time points determined using CCK-8 assay. (B), Cell autophagy observed using monodansylcadaverine staining (500×, scar bar = 20 μm), and the quantitative fluorescence analysis. (C), Protein levels of autophagy-related proteins determined using Western blot analysis. (D and E), cell apoptosis and expression of Caspase-3 determined using flow cytometry and Western blot analysis, respectively. Control, PR-M(+) cells without treatment; L-MIF, low MIF, PR-M(+) cells treated with 1 × 10−6 mol/l; M-MIF, moderate MIF, PR-M(+) cells treated with 1 × 10−5 mol/l; H-MIF, high MIF, PR-M(+) cells treated with 1 × 10−4 mol/l. *P<0.05, vs cells without treatment; #P<0.05, vs cells treated with L-MIF; &P<0.05, vs cells treated with M-MIF. The measurement data were expressed as mean ± standard error, and were analyzed by one-way ANOVA. The experiment was repeated three times.

Figure 4
The cell autophagy and apoptosis of PR-M(+) cells are promoted by PRA

The PR-M(+) cells used for following detection were cells without treatment or cells treated with L-MIF, M-MIF, and H-MIF. (A), OD value of cells at different time points determined using CCK-8 assay. (B), Cell autophagy observed using monodansylcadaverine staining (500×, scar bar = 20 μm), and the quantitative fluorescence analysis. (C), Protein levels of autophagy-related proteins determined using Western blot analysis. (D and E), cell apoptosis and expression of Caspase-3 determined using flow cytometry and Western blot analysis, respectively. Control, PR-M(+) cells without treatment; L-MIF, low MIF, PR-M(+) cells treated with 1 × 10−6 mol/l; M-MIF, moderate MIF, PR-M(+) cells treated with 1 × 10−5 mol/l; H-MIF, high MIF, PR-M(+) cells treated with 1 × 10−4 mol/l. *P<0.05, vs cells without treatment; #P<0.05, vs cells treated with L-MIF; &P<0.05, vs cells treated with M-MIF. The measurement data were expressed as mean ± standard error, and were analyzed by one-way ANOVA. The experiment was repeated three times.

Close modal

PRA promotes the PR-M(+) cell autophagy and apoptosis via the Bcl-2/Beclin1 axis

After determining the role of PRA (MIF) in the autophagy and apoptosis of PR-M(+) cells, we then performed Western blot analysis and Co-IP in order to investigate whether Bcl-2/Beclin1 axis involved in this process. At first, Western blot analysis demonstrated that MIF treatment inhibited protein level of Bcl-2 and promoted protein level of Beclin1, which were changed most obviously in the PR-M(+) cells with H-MIF treatment (P<0.05, Figure 5A). Next, the interaction between Bcl-2 and Beclin1 in PR-M(+) cells treated with MIF was verified by Co-IP assay, which revealed that the interaction between Bcl-2 and Beclin1 was weakened in the PR-M(+) cells with L-MIF, M-MIF, and H-MIF, and weakened most significantly in those with H-MIF (Figure 5B). The results obtained highlighted the potential involvement of the Bcl-2/Beclin1 axis in the promotion of PRA regarding the autophagy and apoptosis of primary uterine leiomyoma cells.

The cell autophagy and apoptosis of PR-M(+) cells are enhanced by PRA via the Bcl-2/Beclin1 axis

Figure 5
The cell autophagy and apoptosis of PR-M(+) cells are enhanced by PRA via the Bcl-2/Beclin1 axis

The PR-M(+) cells used for following detections were cells without treatment or cells treated with L-MIF, M-MIF, and H-MIF. (A), Protein level of Bcl-2 and Beclin1 measured by Western blot analysis. (B), Interaction between Bcl-2 and Beclin1 verified by CO-IP assay. Control, PR-M(+) cells without treatment; L-MIF, low MIF, PR-M(+) cells treated with 1 × 10−6 mol/l; M-MIF, moderate MIF, PR-M(+) cells treated with 1 × 10−5 mol/l; H-MIF, high MIF, PR-M(+) cells treated with 1 × 10−4 mol/l. *P<0.05, vs cells without treatment; #P<0.05, vs cells treated with L-MIF; &P<0.05, vs cells treated with M-MIF. The measurement data were expressed as mean ± standard error, and were analyzed by one-way ANOVA. The experiment was repeated three times.

Figure 5
The cell autophagy and apoptosis of PR-M(+) cells are enhanced by PRA via the Bcl-2/Beclin1 axis

The PR-M(+) cells used for following detections were cells without treatment or cells treated with L-MIF, M-MIF, and H-MIF. (A), Protein level of Bcl-2 and Beclin1 measured by Western blot analysis. (B), Interaction between Bcl-2 and Beclin1 verified by CO-IP assay. Control, PR-M(+) cells without treatment; L-MIF, low MIF, PR-M(+) cells treated with 1 × 10−6 mol/l; M-MIF, moderate MIF, PR-M(+) cells treated with 1 × 10−5 mol/l; H-MIF, high MIF, PR-M(+) cells treated with 1 × 10−4 mol/l. *P<0.05, vs cells without treatment; #P<0.05, vs cells treated with L-MIF; &P<0.05, vs cells treated with M-MIF. The measurement data were expressed as mean ± standard error, and were analyzed by one-way ANOVA. The experiment was repeated three times.

Close modal

The PRA-modulated Bcl-2/Beclin1 axis involves in the PR-M(+) cell autophagy and apoptosis

In order to further clarify whether the PRA-modulated Bcl-2/Beclin1 axis is involved in the autophagy and apoptosis of PR-M(+) cells, the PR-M(+) cells were treated with combination of pLV-EGFP expressing Bcl-2 and H-MIF. RT-qPCR and Western blot analysis were employed to detect the transfection efficiency of Bcl-2, the results of which revealed that the mRNA and protein expression of Bcl-2 was significantly increased after oe-Bcl-2 treatment (P<0.05) (Figure 6A,B), highlighting the successful establishment of pLV-EGFP expressing Bcl-2, which were used for subsequent experimentation. The MDC staining results revealed there to be a reduction in the punctate green fluorescence intensity around nucleus in the PR-M(+) cells following Bcl-2 overexpression and H-MIF treatment in comparison to H-MIF treatment alone (Figure 6C). Moreover, Western blot analysis (Figure 6D) was performed to determine the protein level of autophagy-related proteins, while flow cytometry combined with Western blot analysis (Figure 6E,F) was used to determine the rate of cell apoptosis as well as the expression of activated Caspase-3. Our results revealed that compared with H-MIF treatment alone, co-treatment of overexpressed Bcl-2 and H-MIF increased the protein level of p62 and decreased levels of LC3-II/(LC3-I + LC3-II), coupled with a reduced cell apoptosis rate and activated Caspase-3 (P<0.05). Next, the protein level of Bcl-2 and Beclin1 and the interaction between Bcl-2 and Beclin1 were evaluated by Western blot analysis and Co-IP, respectively. The results obtained displayed that Bcl-2 was notably elevated, while that of Beclin1 was reduced in PR-M(+) cells after co-treatment of overexpressed Bcl-2 and H-MIF (P<0.05, Figure 6G). As displayed in Figure 6H, the interaction between Bcl-2 and Beclin1 was enhanced after the co-treatment of overexpressed Bcl-2 and H-MIF. Collectively, the above results provided further evidence that the PRA-modulated Bcl-2/Beclin1 axis could be involved in promoting the autophagy and apoptosis of primary uterine leiomyoma cells.

The cell autophagy and apoptosis of PR-M(+) cells are promoted by PRA-modulated Bcl-2/Beclin1 axis

Figure 6
The cell autophagy and apoptosis of PR-M(+) cells are promoted by PRA-modulated Bcl-2/Beclin1 axis

The PR-M(+) cells used for following detections were cells without treatment or cells treated H-MIF or in the presence with oe-Bcl-2. (A), The mRNA expression of Bcl-2 in cells detected by RT-qPCR. (B), The protein expression of Bcl-2 in cells examined by Western blot analysis, with protein bands assessed. (C), Cell autophagy observed using monodansylcadaverine staining (500×, scar bar = 20 μm), and the quantitative fluorescence analysis. (D), the protein levels of autophagy-related proteins determined using Western blot analysis. (E), cell apoptosis determined using flow cytometry. (F), The expression of activated Caspae-3 detected by Western blot analysis, with protein bands assessed. (G), Protein level of Bcl-2 and Beclin1 measured by Western blot analysis. (H), Interaction between Bcl-2 and Beclin1 verified by Co-immunoprecipitation assay. Control, PR-M(+) cells without treatment; H-MIF, high MIF, PR-M(+) cells treated with 1 × 10−4 mol/l; H-MIF + oe-Bcl-2, the PR-M(+) cells infected with pLV-EGFP expressing Bcl-2 and treated with H-MIF. *P<0.05, vs cells without treatment; #P<0.05, vs cells treated with H-MIF. The measurement data were expressed as mean ± standard error, and analyzed by one-way ANOVA. The experiment was repeated three times.

Figure 6
The cell autophagy and apoptosis of PR-M(+) cells are promoted by PRA-modulated Bcl-2/Beclin1 axis

The PR-M(+) cells used for following detections were cells without treatment or cells treated H-MIF or in the presence with oe-Bcl-2. (A), The mRNA expression of Bcl-2 in cells detected by RT-qPCR. (B), The protein expression of Bcl-2 in cells examined by Western blot analysis, with protein bands assessed. (C), Cell autophagy observed using monodansylcadaverine staining (500×, scar bar = 20 μm), and the quantitative fluorescence analysis. (D), the protein levels of autophagy-related proteins determined using Western blot analysis. (E), cell apoptosis determined using flow cytometry. (F), The expression of activated Caspae-3 detected by Western blot analysis, with protein bands assessed. (G), Protein level of Bcl-2 and Beclin1 measured by Western blot analysis. (H), Interaction between Bcl-2 and Beclin1 verified by Co-immunoprecipitation assay. Control, PR-M(+) cells without treatment; H-MIF, high MIF, PR-M(+) cells treated with 1 × 10−4 mol/l; H-MIF + oe-Bcl-2, the PR-M(+) cells infected with pLV-EGFP expressing Bcl-2 and treated with H-MIF. *P<0.05, vs cells without treatment; #P<0.05, vs cells treated with H-MIF. The measurement data were expressed as mean ± standard error, and analyzed by one-way ANOVA. The experiment was repeated three times.

Close modal

PRs are the most vital receptors that bind reproductive hormone, progesterone. They collaborate to help regulate menstruation, sexual development and reproduction in females [23]. PRs have various isoforms and have been shown to be capable of reversing hormone stimulation functions [11,12]. In the current study, our results revealed that MIF, as one antagonist of progesterone receptor, is capable of inhibiting the autophagy and apoptosis of PR-M(+) uterine leiomyoma cells by regulating Bcl/Beclin1 axis. The findings not only present a novel molecular function of PR-M in hysteromyoma, but also a potential therapeutic role that MIF could play against uterine leiomyoma.

The first critical finding in our research was that it was PR-M not PR-A/B expressed specifically in primary uterine leiomyoma cells. The next important finding was that PR-M inhibited the autophagy and apoptosis of uterine leiomyoma cells through the Bcl-2/Beclin1 axis. Bcl-2 is a regulator protein that control cellular apoptosis [24]. It has been demonstrated that damage to the Bcl-2 gene can result in a variety of cancers, including breast cancer, prostate cancer, lung cancer, melanoma and chronic lymphocytic leukemia. For example, the co-overexpression of Bcl-2 and Myc can result in aggressive B cell malignancies [25]. The overexpression of Bcl-2 has been shown to lead to drug-resistance of Borrelidin in breast cancer cells, underlining the prognostic value of Bcl-2 in cancer [26]. Beclin1 is an important protein implicated in autophagy of cells, which usually interacts with the Bcl-2 protein. Beclin1 is also a tumorigenic regulator protein. Ovarian cancer cells with up-regulated Beclin1 show increased level of cellular autophagy and are much more sensitive to chemotherapy than normal ones. In lung cancer cells, Beclin1 and LC3 have been reported to exhibit down-regulated levels with the result of decreased autophagy level of cancer cells [27]. Additionally, norcantharidin was demonstrated to up-regulate the expression of Beclin1 by suppressing miR-129-5p, which then leads to an increase in the autophagy of prostate cancer cells [28]. Collectively, PR-M can regulate programmed-death of cancer cell via Bcl-2/Beclin1 axis.

Finally, our results demonstrated that the antagonist of progesterone receptor, MIF, can boost programmed-death of PR-M-positive uterine leiomyoma through the Bcl-2/Beclin1 axis. Although PR-M is a truncated form of a progesterone receptor, and it still contains a hinge and hormone-binding domain, which grants progesterone-binding capability for itself [10]. Currently, MIF, which was initially employed as an abortion drug in early pregnancy, has been increasingly investigated as a potential cancer treatment option. MIF has been shown to repress the proliferation of cancer stem cells in triple-negative breast cancer via down-regulation of KLF5 [29]. Furthermore, MIF has been shown to combine with proteasome or lysosome inhibitors in addition to killing ovarian cancer cells by elevating autophagy-related proteins. Given the possible benefit in cancer treatment, a previous study presented a comprehensive report highlighting the safety and effectiveness of the simultaneous usage of aspirin, lysine, MIF, and doxycycline against cancer metastasis [30]. Hence, although MIF was found to block cancer cell cycle by binding with PR-M, a more accurate elucidation of the molecular regulation between PR-M and Bcl-2 has yet to be acquired. Further studies are required to elucidate the mechanism associated with MIF medication.

In conclusion, the key findings of the current study highlight the therapeutic value of MIF in the treatment of PR-M-positive uterine leiomyoma, which results in the autophagy and apoptosis of cancer cells by regulating Bcl-2/Beclin1 axis (Figure 7). However, considering that long term use of MIF may lead to inflammation of uterine or even un-pregnancy of women, it is necessary to conduct more studies to raise safety of MIF medication.

The schematic diagram depicts molecular basis of the effect of MIF on the autophagy and apoptosis of uterine leiomyoma cells

Figure 7
The schematic diagram depicts molecular basis of the effect of MIF on the autophagy and apoptosis of uterine leiomyoma cells

MIF inhibits the expression of Bcl-2 and weakens the interaction between of Bcl-2 and Beclin1, and then increases the Beclin1 level, thereby promoting the autophagy and apoptosis of PR-M–positive uterine leiomyoma cells.

Figure 7
The schematic diagram depicts molecular basis of the effect of MIF on the autophagy and apoptosis of uterine leiomyoma cells

MIF inhibits the expression of Bcl-2 and weakens the interaction between of Bcl-2 and Beclin1, and then increases the Beclin1 level, thereby promoting the autophagy and apoptosis of PR-M–positive uterine leiomyoma cells.

Close modal

This work was supported by Colleges and Universities in Henan Province Key Scientific Research Project Plan (China) [grant no. 16A320047].

L.Z. and S.C. conceived and designed the project, and wrote the manuscript. Q.F. performed experiments. Z.W. and P.L. analyzed data. All authors reviewed the manuscript.

The authors declare that there are no competing interests associated with the manuscript.

ANOVA

analysis of variance

Bcl-2

B-cell leukemia/lymphoma 2

CCK-8

cell counting kit 8

EGFP

enhanced green fluorescent protein

MDC

monodansylcadaverine

MIF

mifepristone

NC

negative control

OD

optical density

oe

overexpressed

PRA

progesterone receptor antagonist

PR-A/B(−)

PR-A/B-negative

PR-M(+)

PR-M-positive

RT-qPCR

reverse transcription quantitative polymerase chain reaction

sh

short hairpin RNA

SPRM

selective PR modulators

1.
Styer
A.K.
and
Rueda
B.R.
(
2016
)
The epidemiology and genetics of uterine leiomyoma
.
Best Pract. Res. Clin. Obstet. Gynaecol.
34
,
3
12
[PubMed]
2.
Stewart
E.A.
,
Laughlin-Tommaso
S.K.
,
Catherino
W.H.
,
Lalitkumar
S.
,
Gupta
D.
and
Vollenhoven
B.
(
2016
)
Uterine fibroids
.
Nat. Rev. Dis. Primers
2
,
16043
[PubMed]
3.
Donnez
J.
,
Arriagada
P.
,
Donnez
O.
and
Dolmans
M.M.
(
2018
)
Emerging treatment options for uterine fibroids
.
Expert Opin. Emerg. Drugs
23
,
17
23
[PubMed]
4.
Brazert
M.
,
Korman
M.P.
and
Pawelczyk
L.A.
(
2013
)
Applicability of selective progesterone receptor modulators in the treatment of uterine leiomyomata and their future role in the field of gynecology
.
Ginekol. Pol.
84
,
794
800
[PubMed]
5.
Szamatowicz
M.
and
Kotarski
J.
(
2013
)
Selective progesterone receptor modulator (ulipristal acetate–a new option in the pharmacological treatment of uterine fibroids in women
.
Ginekol. Pol.
84
,
219
222
[PubMed]
6.
Lee
K.
,
Jeong
J.
,
Tsai
M.J.
,
Tsai
S.
,
Lydon
J.P.
and
DeMayo
F.J.
(
2006
)
Molecular mechanisms involved in progesterone receptor regulation of uterine function
.
J. Steroid Biochem. Mol. Biol.
102
,
41
50
[PubMed]
7.
Ishikawa
H.
,
Ishi
K.
,
Serna
V.A.
,
Kakazu
R.
,
Bulun
S.E.
and
Kurita
T.
(
2010
)
Progesterone is essential for maintenance and growth of uterine leiomyoma
.
Endocrinology
151
,
2433
2442
[PubMed]
8.
Robinson-Rechavi
M.
,
Escriva Garcia
H.
and
Laudet
V.
(
2003
)
The nuclear receptor superfamily
.
J. Cell Sci.
116
,
585
586
[PubMed]
9.
Kim
J.J.
,
Kurita
T.
and
Bulun
S.E.
(
2013
)
Progesterone action in endometrial cancer, endometriosis, uterine fibroids, and breast cancer
.
Endocr. Rev.
34
,
130
162
[PubMed]
10.
Dai
Q.
,
Shah
A.A.
,
Garde
R.V.
,
Yonish
B.A.
,
Zhang
L.
,
Medvitz
N.A.
et al.
(
2013
)
A truncated progesterone receptor (PR-M) localizes to the mitochondrion and controls cellular respiration
.
Mol. Endocrinol.
27
,
741
753
[PubMed]
11.
Vegeto
E.
,
Shahbaz
M.M.
,
Wen
D.X.
,
Goldman
M.E.
,
O’Malley
B.W.
and
McDonnell
D.P.
(
1993
)
Human progesterone receptor A form is a cell- and promoter-specific repressor of human progesterone receptor B function
.
Mol. Endocrinol.
7
,
1244
1255
[PubMed]
12.
Wen
D.X.
,
Xu
Y.F.
,
Mais
D.E.
,
Goldman
M.E.
and
McDonnell
D.P.
(
1994
)
The A and B isoforms of the human progesterone receptor operate through distinct signaling pathways within target cells
.
Mol. Cell. Biol.
14
,
8356
8364
[PubMed]
13.
Check
J.H.
(
2017
)
The role of progesterone and the progesterone receptor in cancer
.
Expert Rev. Endocrinol. Metab.
12
,
187
197
[PubMed]
14.
Price
T.M.
,
Hansen
E.L.
and
Oliver
T.N.
(
2005
)
Immunofluorescent localization of a novel progesterone receptor(s) in a T47D-Y breast cancer cell line lacking genomic progesterone receptor expression
.
J. Soc. Gynecol. Investig.
12
,
610
616
[PubMed]
15.
Yin
P.
,
Lin
Z.
,
Reierstad
S.
,
Wu
J.
,
Ishikawa
H.
,
Marsh
E.E.
et al.
(
2010
)
Transcription factor KLF11 integrates progesterone receptor signaling and proliferation in uterine leiomyoma cells
.
Cancer Res.
70
,
1722
1730
[PubMed]
16.
Ali
M.
and
Al-Hendy
A.
(
2017
)
Selective progesterone receptor modulators for fertility preservation in women with symptomatic uterine fibroids
.
Biol. Reprod.
97
,
337
352
[PubMed]
17.
Islam
M.S.
,
Protic
O.
,
Giannubilo
S.R.
,
Toti
P.
,
Tranquilli
A.L.
,
Petraglia
F.
et al.
(
2013
)
Uterine leiomyoma: available medical treatments and new possible therapeutic options
.
J. Clin. Endocrinol. Metab.
98
,
921
934
[PubMed]
18.
Engman
M.
,
Granberg
S.
,
Williams
A.R.
,
Meng
C.X.
,
Lalitkumar
P.G.
and
Gemzell-Danielsson
K.
(
2009
)
Mifepristone for treatment of uterine leiomyoma. A prospective randomized placebo controlled trial
.
Hum. Reprod.
24
,
1870
1879
19.
De Amicis
F.
,
Guido
C.
,
Santoro
M.
,
Giordano
F.
,
Dona
A.
,
Rizza
P.
et al.
(
2016
)
Ligand activated progesterone receptor B drives autophagy-senescence transition through a Beclin-1/Bcl-2 dependent mechanism in human breast cancer cells
.
Oncotarget
7
,
57955
57969
[PubMed]
20.
Yin
P.
,
Lin
Z.
,
Cheng
Y.H.
,
Marsh
E.E.
,
Utsunomiya
H.
,
Ishikawa
H.
et al.
(
2007
)
Progesterone receptor regulates Bcl-2 gene expression through direct binding to its promoter region in uterine leiomyoma cells
.
J. Clin. Endocrinol. Metab.
92
,
4459
4466
[PubMed]
21.
Holmes
F.A.
,
Espina
V.
,
Liotta
L.A.
,
Nagarwala
Y.M.
,
Danso
M.
,
McIntyre
K.J.
et al.
(
2013
)
Pathologic complete response after preoperative anti-HER2 therapy correlates with alterations in PTEN, FOXO, phosphorylated Stat5, and autophagy protein signaling
.
BMC Res. Notes
6
,
507
[PubMed]
22.
Kaya
H.S.
,
Hantak
A.M.
,
Stubbs
L.J.
,
Taylor
R.N.
,
Bagchi
I.C.
and
Bagchi
M.K.
(
2015
)
Roles of progesterone receptor A and B isoforms during human endometrial decidualization
.
Mol. Endocrinol.
29
,
882
895
[PubMed]
23.
Bouchard
P.
(
1999
)
Progesterone and the progesterone receptor
.
J. Reprod. Med.
44
,
153
157
[PubMed]
24.
Real
P.J.
,
Cao
Y.
,
Wang
R.
,
Nikolovska-Coleska
Z.
,
Sanz-Ortiz
J.
,
Wang
S.
et al.
(
2004
)
Breast cancer cells can evade apoptosis-mediated selective killing by a novel small molecule inhibitor of Bcl-2
.
Cancer Res.
64
,
7947
7953
[PubMed]
25.
Otake
Y.
,
Soundararajan
S.
,
Sengupta
T.K.
,
Kio
E.A.
,
Smith
J.C.
,
Pineda-Roman
M.
et al.
(
2007
)
Overexpression of nucleolin in chronic lymphocytic leukemia cells induces stabilization of bcl2 mRNA
.
Blood
109
,
3069
3075
[PubMed]
26.
Gafiuc
D.
,
Weiss
M.
,
Mylonas
I.
and
Bruning
A.
(
2014
)
Borrelidin has limited anti-cancer effects in bcl-2 overexpressing breast cancer and leukemia cells and reveals toxicity in non-malignant breast epithelial cells
.
J. Appl. Toxicol.
34
,
1109
1113
[PubMed]
27.
Jiang
Z.F.
,
Shao
L.J.
,
Wang
W.M.
,
Yan
X.B.
and
Liu
R.Y.
(
2012
)
Decreased expression of Beclin-1 and LC3 in human lung cancer
.
Mol. Biol. Rep.
39
,
259
267
[PubMed]
28.
Xiao
W.
,
Dai
B.
,
Zhu
Y.
and
Ye
D.
(
2015
)
Norcantharidin induces autophagy-related prostate cancer cell death through Beclin-1 upregulation by miR-129-5p suppression
.
Tumour Biol.
29.
Liu
R.
,
Shi
P.
,
Nie
Z.
,
Liang
H.
,
Zhou
Z.
,
Chen
W.
et al.
(
2016
)
Mifepristone suppresses basal triple-negative breast cancer stem cells by down-regulating KLF5 expression
.
Theranostics
6
,
533
544
[PubMed]
30.
Wan
L.
,
Dong
H.
,
Xu
H.
,
Ma
J.
,
Zhu
Y.
,
Lu
Y.
et al.
(
2015
)
Aspirin, lysine, mifepristone and doxycycline combined can effectively and safely prevent and treat cancer metastasis: prevent seeds from gemmating on soil
.
Oncotarget
6
,
35157
35172
[PubMed]
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).