Open Access

MicroRNA‑202 inhibits endometrial stromal cell migration and invasion by suppressing the K‑Ras/Raf1/MEK/ERK signaling pathway

Retraction in: /10.3892/ijmm.2024.5375

  • Authors:
    • Di Zhang
    • Ling Wang
    • Hua‑Lei Guo
    • Zi‑Wei Zhang
    • Chong Wang
    • Ri‑Cheng Chian
    • Zhi‑Fen Zhang
  • View Affiliations

  • Published online on: October 9, 2020     https://doi.org/10.3892/ijmm.2020.4749
  • Pages: 2078-2088
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The enhanced migratory ability of endometrial stromal cells (ESCs) is a key factor in the formation of functional endometrium‑like tissues outside the uterine cavity during endometriosis (EMS). Although accumulating evidence has suggested the importance of microRNAs (miRNAs) in the pathogenesis of EMS, the role of particular miRNAs in the invasiveness of ESCs remain poorly understood. In the present study, the function of miRNAs in the invasiveness of ESCs, along with the associated underlying mechanism involved, were investigated. Initially, the expression patterns of miRNAs in the ectopic and eutopic endometrium isolated from patients with EMS were analyzed using microarray. MicroRNA‑202‑5p (miR‑202) was selected for further study due to its previously reported suppressive effects on the invasion in various types of cancers. The expression of miR‑202 and K‑Ras in eutopic and ectopic endometrioma tissues were detected using reverse transcription‑quantitative PCR, immunohistochemistry and western blotting. The migration and invasion ability of ESCs was determined using wound healing and Transwell invasion assays, respectively. Compared with that from healthy individuals, miR‑202 expression was demonstrated to be lower in the eutopic endometrium from patients with EMS, which was even lower in ectopic endometrium. Functional experiments in primary ESCs revealed that enhanced miR‑202 expression suppressed the cell invasion and migration abilities, which was also accompanied with increased E‑cadherin and reduced N‑cadherin expression in ESCs, suggesting its potentially suppressive role in epithelial‑mesenchymal transition. K‑Ras is a well‑known regulator of the ERK signaling pathway that was shown to be directly targeted and negatively regulated by miR‑202. In addition, K‑Ras expression was found to be upregulated in the ectopic endometrium, where it correlated negatively with that of miR‑202. Knocking down K‑Ras expression mimicked the anti‑invasive effects of miR‑202 overexpression on ESCs, whilst K‑Ras overexpression attenuated the inhibitory role of miR‑202 overexpression in ESC invasion. The K‑Ras/Raf1/MEK/ERK signaling pathway was also blocked by miR‑202 overexpression. These findings suggested that miR‑202 inhibited ESC migration and invasion by inhibiting the K‑Ras/Raf1/MEK/ERK signaling pathway, rendering miR‑202 a candidate for being a therapeutic target for EMS.

Introduction

Endometriosis (EMS) is a common gynecological disorder that is primarily characterized by the presence of endometrium-like tissue outside the uterine cavity (1). This disease affects 6-10% of the female population at reproductive age, where ~70% patients with EMS would suffer from infertility or chronic pelvic pain (2). Preliminary studies have shown that enhanced migratory and invasive capabilities of endometrial stromal cells (ESCs) may serve to be the critical factor in the occurrence of EMS (3,4). Therefore, inhibiting ESC invasion and migration may prove to be an effective therapeutic intervention strategy for patients with EMS.

MicroRNAs (miRNAs) are single-stranded non-coding RNAs that bind to the 3′-untranslated region (3′-UTR) of target mRNAs, resulting in translational inhibition or mRNA degradation (5,6). Accumulating evidence has reported that the expression profiles of miRNAs are frequently dysregulated during EMS, a pathogenic process in which they serve important roles (7,8). Okamoto et al (9) previously reported that miR-210 overexpression promoted the pathogenesis of EMS by activating STAT3, whereas Li et al (10) reported that transfection with the miR-451a inhibitor resulted in reductions in EMS lesion size in a mouse model. In addition, Hsu et al (11) revealed that overexpression of miR-199a-5p could reduce the size of endometriotic lesions in a mice model of endometriosis. miRNA expression profiles obtained from the sera and plasma of patients with EMS have been recently described, which have been reported to confer relatively high diagnostic power (12,13). Let-7b and miR-135a were previously found to be differentially expressed in the serum samples of patients with EMS compared with those without EMS, where they were demonstrated to be viable biomarkers of this disease (14,15). A previous study has also reported that plasma miR-17-5p, miR-20a and miR-22 are significantly lower in the ectopic endometrium compared with those in the eutopic endometrium (16). These previous findings aforementioned support the potential clinical utility of miRNAs for the diagnosis of this disease. However, the biological role of miRNAs in the regulation of ESC physiology and its under-lying molecular mechanism remain to be fully elucidated.

miR-202 is located within a chromosomal fragile site in the 10q26 locus that has been previously found to modulate the pathological processes of several malignancies, with marked effects on tumor cell proliferation, invasion and migration (17,18). Meng et al (19) found that miR-202 significantly inhibited cell migration and invasion in esophageal squamous cell carcinoma (ESCC) cell lines by targeting laminin α1. In another study, Ke et al (20) previously demonstrated that miR-202 inhibited the proliferation and invasion of colorectal carcinoma cells by downregulating the oncogenic protein SWI/SNF related, matrix associated, actin dependent regulator of chromatin subfamily C member 1. In addition, Lin et al (21) reported that miR-202 over-expression significantly inhibited tumor formation by HCT116 cells in the BALB/c nude mouse xenograft model. Although reductions in miR-202 expression have been previously identified in endometrial adenocarcinoma (22), the role of miR-202 in the invasiveness of ESCs have not been documented.

In the present study, miRNA profiles of the ectopic and eutopic endometrium were analyzed using miRNA micro-array, following which miR-202 was selected for subsequent experiments. The aim is to explore the role of miR-202 in the invasion and migration of primary ESCs and propose a possible mechanism. In addition, the cellular and molecular effects of miR-202 on ESCs, as well as potential downstream targets of miR-202, were explored.

Materials and methods

Clinical specimens

Eutopic and ectopic endometrial tissues were collected from 30 female patients (mean age, 41.28±4.01 years; age range, 25-48 years; stage I-II, n=13; stage III-IV, n=17) who underwent laparoscopic surgery for ovarian endometriosis between December 2016 and July 2018 in Hangzhou Women's Hospital (Hangzhou, China). Diagnosis was performed according to the revised American Fertility Society classification of endometriosis (23). There were 15 patients with proliferative phase endometriosis and 15 patients with secretory phase endometriosis. For the control group, 15 healthy endometrial tissues (mean age, 32.20±7.62 years; range 22-45 years) were obtained from women with tubal factor infertility without endometriosis who underwent hysteroscopic exploration for oviduct obstruction during the same period. All cases of endometriosis were confirmed by postoperative pathological diagnosis. Experiments in the present study were approved by The Ethics Committee of Hangzhou Women's Hospital (approval no. 2016-0231) and written informed consents for tissue donation were obtained from each patient for the research purposes only.

miRNA microarray analysis

Total miRNA was extracted from the eutopic (n=3) and ectopic (n=3) endometrium tissues which were distinct from those subjected to RT-qPCR, using the miRNeasy mini kit according to manufacturer's protocol (Qiagen, Inc.). Total RNA (200 ng) was labeled using the miRCURY LNA™ Hy3™/Hy5™ Power labeling kit (cat. no. 208032-A, Exiqon; Qiagen GmbH) and hybridized on the miRCURY™ LNA Array (v.16.0; cat. no. 208040 Exiqon; Qiagen GmbH) according to the manufacturer's protocol. Microarray images were taken using a Genepix 4000B scanner (Molecular Devices, LLC) and analyzed with Genepix Pro 6.0 software (Molecular Devices, LLC). The intensity of green signal was calculated after background subtraction before four replicated spots of each probe on the same slide were averaged. Median normalization was calculated using the following formula: Normalized Data=(Foreground-Background)/Median, where Median is the 50% quantile of miRNA intensity >50 in all samples after background correction. After normalization, significant differentially expressed miRNAs between the two groups were identified through fold change and P-value. Differentially expressed miRNAs between two samples were filtered by fold change. Finally, the heat map of the 49 miRNAs with most obvious differences was created using hierarchical clustering by GeneSpring GX, version 7.3 (Agilent Technologies, Inc.). The threshold of screening most differentially expressed miRNAs was set as a fold change >2.0 and a P-value <0.05.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the endometrial tissues or cultured cells using TRIzol® Reagent according to manufacturer's protocol (Invitrogen; Thermo Fisher Scientific, Inc.). RNA was then reverse transcribed into cDNA using the PrimeScript™ RT reagent kit (cat. no. RR047A; Takara Bio, Inc.) or the TaqMan™ MicroRNA Reverse Transcription kit (cat. no. 4366596; Thermo Fisher Scientific, Inc.), respectively. miR-202 and K-Ras expression were measured using SYBR® PrimeScript™ RT-PCR kit (cat. no. RR014A; Takara Bio, Inc.) in an Applied Biosystems 7300 Real-Time PCR System (Thermo Fisher Scientific, Inc.). The primer sequences used are listed as follows: miR-202 forward, 5′-CCT CCC AGG CTC ACG AGG CT-3′ and reverse, 5′-GGT GCA GGT GCACTG GTG CA-3′; U6 forward, 5′-GCT TCG GCA GCA CAT ATA CTA AAA T-3′ and reverse, 5′-CGC TTC ACG AAT TTG CGT GTC AT-3′; K-Ras forward, 5′-CAT GTT CTA ATA TAG TCA CA-3′ and reverse, 3′-GTT ATC TCC ATT TAG AAC AA-5′ and GAPDH forward, 5′-CGA GCC ACA TCG CTC AGA CA-3′ and reverse, 5′-GTG GTG AAG ACG CCA GTG GA-3′. The thermocycling conditions were as follows: Initial denaturation at 95°C for 10 min, followed by 40 cycles of 95°C for 15 sec, 55°C for 30 sec and 70°C for 30 sec. Each sample was run in triplicate for analysis. All samples were normalized to U6 or GAPDH and fold changes were calculated using the 2−∆∆Cq method (24). All experiments were performed three times and each in duplicate.

Cell culture and transfection

Endometriosis (EMS) is characterized by the presence of endometrium-like tissue outside the uterine cavity which are called ectopic endometrial tissues (25). Since the enhanced migratory and invasive capabilities of endometrial stromal cells (ESCs) may serve to be the critical factor in the occurrence of endometriosis EMS (26), primary endometrial stromal cells were isolated from three ectopic endometrial tissues using procedures described previously (27). The cells were maintained in DMEM/F12 (Gibco; Thermo Fisher Scientific, Inc.) containing with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 mg/ml streptomycin (Sigma-Aldrich; Merck KGaA) at 37°C in a 5% CO2 incubator. 293T cells (ATCC) used for luciferase assays were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) supplemented with 10% FBS under 37°C in a humidified atmosphere under 5% CO2. Cellular purity was examined by immunofluorescence staining for strong cytoplasmic immunostaining for vimentin (marker of stromal cells) and negative cellular staining for cytokeratin 7 (markers of epithelial cells) (28,29).

When ESCs (8.0×105 cells/well) in six-well plates reached ~80% confluence, miR-202 mimics (5′-UU CCU AUG CAU AUA CUU CUU UG-3′; 20 nM; Shanghai GenePharma Co., Ltd.), mimics NC (5′-UUC UCC GAA CGU GUC ACG UTT-3′; 20 nM), si-K-Ras (sense sequence, 5′-UGU GGA CGA AUA UGA UCC AAC AAU A-3′) or si-Scramble (sense sequence, 5′-UGU GCA UAA GUA CUA AAC ACG GAU A-3′; 50 nM; Shanghai GenePharma Co., Ltd.) were transfected into cells at 37°C for 24 h using Lipofectamine® 2000 according to manufacturer's protocol (Invitrogen; Thermo Fisher Scientific, Inc.).

To induce the overexpression of K-ras, K-Ras gene (NCBI Reference Sequence, NM_033360.4) was amplified by PCR using human cDNA isolated from ectopic endometrium tissues as the template. The coding region sequences of K-ras were then cloned into the pcDNA3.1(-) plasmid (Shanghai GenePharma Co., Ltd.), which were termed pcDNA3.1-Kras. The empty pcDNA3.1 was used as a negative control (NC).

Cell invasion assay

Cell invasion assay was performed as previously described (30). Briefly, the Transwell chambers (Corning, Inc.) were coated with Matrigel (BD Biosciences) and placed into the incubator at 37°C for 3 h. Freshly trypsinized and washed cells were suspended in serum free media and 200 µl cells (8×104 cells)/well were placed into the upper chambers 24 h after transfection, whilst DMEM/F12 medium with 20% FBS (600 µl) was added to the lower chamber. After 24 h incubation at 37°C, cells attached onto the upper side of the chamber membranes were removed using a cotton swab. The membranes were then fixed in 10% formaldehyde for 15 min at room temperature and stained with 0.2% crystal violet for 30 min at room temperature. The number of cells was counted in five randomly selected high-power fields per chamber by a light microscope (Olympus Corporation) at ×200 magnification.

Wound healing assay

Wound healing assay was performed as previously described (31). Briefly, ESCs were seeded onto 12-well plates at 2×105 cells/well, following which a scratch was made using a 10-µl pipette tip in the confluent cell mono-layer 24 h after transfection. The cells were then washed twice with PBS and incubated in serum-free DMEM at 37°C. The wound healing images (magnification, ×200) were taken at the time of wounding, defined as 0 and 24 h after scratching using a light microscope (Olympus Corporation). The wound area was photographed at 0 and 24 h and analyzed using the ImageJ software (version 1.46; National Institute of Health). The percentage of the wound healing rate was calculated using the following formula: % wound healing=specific day wound size/initial wound size ×100.

Plasmids and constructs

The predicted binding sites between miR-202 and K-Ras were obtained using TargetScan 7.0 (http://www.targetscan.org/vert_70/) and miRanda (v3.3a; http://www.microrna.org) online softwares. The full-length 3′-UTR of the wild-type (wt) human K-Ras gene was amplified by PCR (2X Phanta® Master Mix; Vazyme Biotech Co., Ltd.) using human cDNA isolated from ectopic endometrium tissues as the template using the following primers: Forward, 5′-CCG GGT ACC ATG ACT GAA TAT AAA CTT GTG G-3′ and reverse, 5′-CCG CTC GAG TTA CAT AAT TAC ACA CTT TGT C-3′. Thermocycling conditions consisted of an initial incubation at 95°C for 5 min, followed by 30 cycles of 94°C for 30 sec, 56°C for 30 sec and 72°C for 30 sec, followed by a final 10-min extension step at 72°C. The resultant amplicon was then cloned into the KpnI and XhoI sites of the pGL vector (Promega Corporation). The mutant (mt)-K-Ras vector was also constructed. This vector contains mutations in the major miR-202 binding site made using the GeneArt™ Site-Directed Mutagenesis Plus System according to the manufacturer's protocols (Thermo Fisher Scientific, Inc.). Successful plasmid construction was confirmed by Sanger sequencing by Sangon Biotech Co., Ltd.

Luciferase assay

In total, 8×104 293T cells were added into 24-well plates until ~90% confluence was reached, following which the cells were co-transfected with 0.12 mg either wt-K-Ras-3′UTR-pGL3 or mt-K-Ras-3′UTR-pGL3 reporter plasmids together with 40 nM miR-202 mimic or mimics NC using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.). Luciferase activity was measured using the Dual-luciferase® Reporter Assay System (Promega Corporation) 48 h after transfection according to manufacturer's protocol. All luciferase activity readings were normalized relative to the activity of the Renilla luciferase control. All experiments were carried out in triplicate.

Immunohistochemical (IHC) staining

IHC was performed to determine the expression of K-Ras. Briefly, eutopic and ectopic endometrial tissues were fixed in 4% paraformaldehyde at 4°C in PBS for 20 min, embedded in paraffin and cut into 4-µm sections. The sections were immersed in xylene twice for 10 min and then immersed in a descending ethanol gradient, before being incubated with 1% BSA (Sigma-Aldrich; Merck KGaA) at 20°C for 1 h and in hydrogen peroxide at 20°C for 30 min. Thereafter, they were probed with the anti-K-Ras primary antibody (1:200; cat. no. ab216890; Abcam) at 4°C overnight. Subsequently, the sections were washed with PBS three times, incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (1:2,000; cat. no. 7074; Cell Signaling Technology, Inc.) at 37°C for 30 min and then stained with 3′,3′-diaminobenzidine for 5-15 min at room temperature. The specimens were then counterstained with hematoxylin for 2 min at room temperature. Finally, the photographs of five fields of view per section were taken using a light microscope with a digital camera (VHX-5000; Keyence Corporation) at ×200 magnification.

Immunofluorescent staining

Prior to immunofluorescence staining, the primary ESCs in 6-well plates at 5×105 cells per well were rinsed three times with PBS and then fixed with 4% paraformaldehyde for 10 min at room temperature. Cells were blocked with 5% bovine serum albumin (Sigma-Aldrich; Merck KGaA) for 60 min at room temperature. Subsequently, the cells were incubated at 4°C overnight in a solution containing primary antibodies specific for vimentin (1:200, cat. no. ab92547; Abcam) or cytokeratin (1:200, cat. no. ab68459; Abcam). Subsequently, the cells were stained with an Alexa Fluor® 488-conjugated anti-rabbit antibody (1:200, cat. no. ab150117, Abcam) for 1 h at 37°C. Finally, images were captured by a fluorescence microscope (Olympus Corporation) at ×200 magnification.

Western blot analysis

Western blotting was performed as previously described (32). Briefly, total protein of endometrial stromal cells was isolated using NP-40 lysis buffer (cat. no. P0013F, Beyotime Institute of Biotechnology) and protein concentration was quantified using bicinchoninic acid protein assay (Beyotime Institute of Biotechnology). The protein samples (40 µg) were then separated by 10-12% SDS-PAGE and transferred onto PVDF membranes (EMD Millipore), followed by blocking with 5% skim milk for at 4°C overnight. The membranes were then probed with primary antibodies overnight at 4°C before being incubated with HRP-conjugated anti-rabbit IgG secondary antibodies (1:10,000; cat. no. 7074; Cell Signaling Technology, Inc.) for 1 h at room temperature. Antibodies against K-Ras (1:1,000; cat. no. 14429), E-cadherin (1:1,000; cat. no. 3195), N-cadherin (1:1,000; cat. no. 13116), total Raf1 (1:1,000; cat. no. 53745), phosphorylated (p)-Raf1 (1:1,000; cat. no. 9427), MEK1/2 (1:1,000; cat. no. 8727), p-MEK1/2 (1:1,000; cat. no. 3958), ERK1/2 (1:1,000; cat. no. 4695), p-ERK1/2 (1:1,000; cat. no. 4376) and β-actin (1:1,000; cat. no. 4970) were obtained from Cell Signaling Technology, Inc. Proteins bands were detected using the enhanced chemiluminescence (ECL) detection system (Cytiva) and quantified using ImageJ (version 1.46; National Institutes of Health).

Statistical analysis

Statistical analysis was performed using the SPSS 13.0 software package (SPSS Inc.). All data are presented as the mean ± SD from ≥ three experimental repeats. Pairwise comparisons was conducted using Student's t-test. Comparisons among multiple groups were performed by one-way ANOVA followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference. Spearman's rank correlation coefficient analysis was used for all correlation analyses.

Results

miR-202 expression is downregulated in the eutopic and ectopic endometrium

To examine the role of miRNAs in the progression of EMS, a microarray analysis was used to analyze the expression patterns of miRNAs in the eutopic and ectopic endometrium. Compared with those in eutopic endometrium, 19 downregulated miRNAs and 30 upregulated miRNAs were found in the ectopic endometrium (Fig. 1A). Notably, compared with that in the eutopic endometrium, miR-202 was found to be one of the most markedly downregulated miRNAs in the ectopic endometrium. Therefore, miR-202 was chosen for subsequent experiments.

To validate the expression profile of miR-202 obtained from the miRNA microarray assay, miR-202 expression in eutopic, ectopic and control endometria were measured. Compared with that in the control endometrium, miR-202 expression was found to be significantly lower in the eutopic endometrium from patients with EMS, which was even lower in ectopic endometrium (Fig. 1B). Subsequently, the expression levels of miR-202 were also measured in ESCs isolated from patients with ectopic endometriosis and control endometrium. miR-202 expression was revealed to be significantly downregulated in ESCs from patients with endometriosis compared with that in control ESCs (Fig. 1C). This finding suggesting that miR-202 may serve important roles in the pathogenesis of EMS.

Overexpression of miR-202 inhibits ESC invasion and migration

The purity of isolated endometrial stromal cells were determined by diffuse and strong cytoplasmic immunostaining for vimentin (marker of stromal cells) and weak cellular staining for cytokeratin 7 (marker of epithelial cells; Fig. 2A) (28). To examine the possible role of miR-202 in primary ESCs from patients with ectopic endometriosis, the migratory and invasive capabilities were measured using Transwell and wound healing assays, respectively, following transfection with miR-202 mimics. miR-202 expression was significantly increased in ESCs after transfection with miR-202 mimics compared with cells transfected with the mimics NC (Fig. 2B). Compared with cells in the mimics NC group, ESCs in the miR-202 mimics group exhibited significantly lower the invasive and migratory capabilities (Fig. 2C and D). Since epithelial-mesenchymal transition (EMT) has been previously reported to be a key step in the invasiveness of ESCs (33,34), the influence of miR-202 on EMT was next evaluated. According to western blotting results, E-cadherin expression, an epithelial marker, was found to be markedly increased, whilst N-cadherin, a mesenchymal marker, was revealed to be markedly reduced in ESCs following miR-202 mimics transfection (Fig. 2E). This suggests that miR-202 overexpression inhibited ESC invasion and migration by interfering with the EMT progress in ESCs.

K-Ras is a direct target of miR-202 in ESCs

To elucidate the molecular mechanism underlying the suppressive effects of miR-202 on ESC invasion, TargetScan 7.0 and miRanda were used to search for the predict targets of miR-202. This analysis revealed that K-Ras, an important regulator of Raf1/MEK/ERK pathway, was identified to be a predicted target of miR-202 (Fig. 3A). To verify experimentally if miR-202 can target K-Ras directly, luciferase reporter assay was performed. Transfection with the miR-202 mimics significantly reduced the luciferase activity of the wt-K-Ras 3′-UTR compared with cells transfected with mimics NC, but not the luciferase activity of the mut-K-Ras 3′-UTR construct (Fig. 3B). To assess if K-Ras expression is regulated by miR-202, the protein expression of K-Ras was measured in ESCs after transfection with miR-202 mimics. miR-202 mimics transfection led to a marked reduction in K-Ras expression in ESCs, compared with cells transfected with mimics NC (Fig. 3C). The expression levels of miR-202 mRNA were next compared in the eutopic and ectopic endometria by RT-qPCR. Consistent with previous reports (35), K-Ras expression was demonstrated to be significantly higher in the ectopic endometrium compared with that in the eutopic endometrium (Fig. 3D). Spearman's correlation analysis showed that there was an inverse relationship between K-Ras and miR-202 expression in the ectopic endometrium (r=−0.8244; P<0.01l; Fig. 3E). Next, the expression levels of K-Ras in eutopic and ectopic endometrium was assessed by IHC. (Fig. 3F), K-Ras staining was revealed to be markedly higher in the ectopic endometrium compared with that in the eutopic endometrium. Collectively, these findings suggest that miR-202 suppressed the expression of K-Ras mRNA by binding to its 3′-UTR.

K-Ras knockdown inhibits the invasion and migration of ESCs

To determine if K-Ras serves a role in the migration and invasion of ESCs, ESCs were transfected with either si-scrambled or si-k-Ras. Western Blot analysis revealed that K-Ras protein expression was significantly reduced in ESCs following si-K-Ras transfection (Fig. 4A). Subsequent Transwell and wound healing assay results showed that the ESC invasion and migration were significantly inhibited in ESCs transfected with si-K-Ras compared with those transfected with si-scrambled (Fig. 4B and C). In addition, it was also observed that K-Ras knockdown markedly increased E-cadherin whilst reducing N-cadherin expression in ESCs (Fig. 4D). All these observations suggest that downregulation of K-Ras confers an inhibitory effect on the invasion and migration of ESCs, which resembles the effects of miR-202 overexpression.

K-Ras is an intermediary in the suppression of invasion and migration of ESCs induced by miR-202 overexpression

To explore if K-Ras mediated the suppressive effects of miR-202 on ESC invasion, miR-202 mimics and pcDNA-K-Ras were co-transfected into ESCs. Initially, the expression of K-Ras was measured by western blotting 24 h following transfection with either pcDNA-K-Ras or pcDNA-empty vector. K-Ras protein expression was found to be significantly upregulated following transfection with pcDNA-K-Ras, compared with that in ESCs transfected with the pcDNA-empty vector (Fig. 5A). Transfection with pcDNA-K-Ras also markedly reversed the suppressive effects of miR-202 mimics on the expression of K-Ras in ESCs (Fig. 5B). Transwell and wound healing assays demonstrated that miR-202 mimics significantly suppressed the invasion and migration of ESCs compared with those in the control group, but the inhibitory effects of miR-202 mimics were found to be significantly reversed by K-Ras upregulation (Fig. 5C-F). These data suggest that miR-202 upregulation inhibited the invasion and migration of ESCs by targeting K-Ras.

miR-202 overexpression inhibits the activation of k-Ras/Raf1/MEK/ERK signaling in ESCs

Previous studies have reported that K-Ras regulated the Raf1/MEK/ERK signaling pathway, which is directly associated with cell invasion and migration (36,37). To determine further if the miR-202-medi-ated regulation of K-Ras expression affected the activation of Raf1/MEK/ERK signaling, expression and phosphorylation levels of the key components of this pathway, specifically Raf1, MEK and ERK, were measured. Western blot analysis data showed that overexpression of miR-202 significantly reduced Raf1, MEK and ERK phosphorylation compared with those in the control group, which was in turn significantly reversed by K-Ras overexpression (Fig. 6A and B). These findings suggest that the miR-202/K-Ras axis is accompanied with the regulation of the Raf1/MEK/ERK pathway, which may form the underlying mechanism of ESC invasiveness (Fig. 7).

Discussion

In the present study, miR-202 expression was found to be lower in the ectopic and eutopic endometrium isolated from patients with EMS. In addition, miR-202 overexpression significantly inhibited the invasion and migration of ESCs by blocking the activation of K-Ras/Raf1/MEK/ERK signaling. These findings suggest that miR-202 may serve as a potential therapeutic target for patients with EMS in the future.

Accumulating evidence has demonstrated the involvement of aberrantly expressed miRNAs in EMS pathogenesis (13,38). Zhu et al (39) previously showed that upregulation of miR-488 suppressed the migration and invasion of endometrial glandular endometrial cells isolated from mice by targeting the Frizzled-7 protein. Liang et al (40) found that injection with the miR-200c mimics significantly inhibited the growth of ectopic endometriotic lesions in a rat EMS model. In addition, Li et al (10) reported that miR-451a knockdown improved endometriosis lesion injury in a mouse model. In the present study, using microarray analysis, it was found that EMS altered the expression profiles of many miRNAs. Among these aberrantly expressed miRNAs, miR-451a and miR-33b were revealed to be increased, whilst miR-20a was decreased, consistent with previous reports (16,41,42), implicating reliability of the microarray analysis performed in the present study. In previous studies, miR-202 also was found to be aberrantly expressed in endometrial tissues from patients with EMS (13,38). In addition, miR-202 has been previous shown to exert suppressive roles on the invasive and migratory capacities of tumor cells (43-45). According to the aforementioned findings from the present and previous studies, it could be hypothesized that miR-202 can regulate EMS progression.

Previous studies have demonstrated that miR-202 mediates suppressive invasive roles in various types of human tumors (21,44). Xu et al (45) reported that miR-202 targeted Rho-associated protein kinase 1 to inhibit the invasive and migratory abilities of breast cancer. In addition, Zhang et al (18) found that miR-202 upregulation resulted in significant reductions in the migration and invasion of prostate cancer cells by targeting the phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α. Although the function of miR-202 is better characterized in tumors, information on the role of miR-202 in EMS, especially in the invasiveness of ESCs, remain elusive. In the present study, it was found that upregulation of miR-202 significantly inhibited the invasion and migration of primary ESCs from patients with EMS. Since EMT has been previously documented to be a step in mediating the invasion of ESCs (32), the effect of miR-202 on EMT was assessed in ESCs from patients with EMS. Overexpression of miR-202 led to the promotion of E-cadherin expression and reduced the expression of N-cadherin. All these data suggest that miR-202 inhibited the invasion and migration of primary ESCs by suppressing EMT.

The fundamental function of miRNA is to regulate target mRNAs by direct binding or by inhibition of protein synthesis. In the present study, the predicted target gene was found to be K-Ras, a positive regulator of the ERK signaling pathway (46). Luciferase activity assay and western blotting were subsequently used to verify that K-Ras is directly targeted by miR-202. In addition, a notable inverse correlation was observed between the expression of K-Ras and miR-202 in the ectopic endometrium. These results suggest that miR-202 directly interacts with and negatively regulate the expression of K-Ras.

K-Ras is one of the three members of the Ras oncogene family that has been previously implicated in the carcinogen-esis of many human cancers, where K-Ras knockdown has been shown to suppress tumor cell growth and invasion (47,48). Recently, a number of miRNAs, including miR-202, were reported to suppress K-Ras expression and functioned as a tumor suppressor (49). The present study found that K-Ras knockdown conferred an inhibitory effect on the invasion and migration of ESCs. By contrast, overexpression of K-Ras reversed the inhibitory effects of miR-202 overexpression on ESC invasion and migration. Therefore, miR-202 can be hypothesized to mediate effects on ESC invasion and migration in EMS, in a manner that is dependent on the expression of K-Ras. However, the underlying mechanism of this aspect requires further exploration.

Notably, K-Ras can activate downstream RAF protein kinases to in turn activate the RAS/RAF/MEK/ERK pathway, which serves as one of the most important signaling pathways in cancer (50). Recent studies showed that this pathway plays an important role in the initiation and progression of Ems (51,52). Liu et al (51) previously found that ERK signaling contributed to the enhanced invasiveness of ectopic ESCs, whilst Kim et al (53) reported that ERK1/2 signaling was involved in endometriotic cell migration induced by peritoneal fluid from individuals with EMS. These previous studies suggest that repressing the activation of this signaling pathway may prove to be a viable option for treating this condition. In the present study, it was found that the Raf-1/MEK/ERK signaling pathway was suppressed by miR-202 upregulation whereas K-Ras overexpression reactivated the Raf-1/MEK/ERK pathway, suggesting that miR-202 blocked the Raf-1/MEK/ERK pathway by targeting K-Ras.

The present study assessed the possibility of exploiting the changes in the miRNA expression profile in the ectopic endometrium of patients with EMS, compared with that in the eutopic endometrium, as potential biomarkers for the disease. The role of miR-202 in ESC migration and invasion were also examined. Examining the effects of altering miR-202 expression on EMS could contribute to facilitating diagnosis and developing treatment strategies of this pathology. However, a number of limitations remain associated with the present study. The clinical sample size used is relatively small. A larger number of samples would be required to verify further the possibility of using miR-202 as a diagnostic marker. Furthermore, off-target effects of miR-202 in the in vitro experiments cannot be ruled out. In the future, the direct relationship between miR-202 and K-Ras would need to be verified.

In conclusion, the present study demonstrated that down-regulation of miR-202 promoted ESCs migration and invasion by regulating K-Ras/Raf-1/MEK/ERK pathway, suggesting that it served an important role in the occurrence and development of EMS. This suggests that miR-202 can serve as a future therapeutic target of EMS.

Acknowledgments

Not applicable.

Funding

No funding was received.

Availability of data and materials

The data that support the findings of this study cannot be uploaded onto an online repository due to this research project being ongoing but are available from the corresponding author upon reasonable request.

Authors' contributions

DZ and LW performed the experiments, contributed to data analysis and wrote the paper. HLG, ZWZ and CW analyzed the data. RCC and ZFZ conceptualized the study design, contributed to data analysis and experimental materials. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

All individuals provided written informed consent for the use of human specimens for clinical research. The present study was approved by Hangzhou Women's Hospital Ethics Committees (approval no. 2016-0231; Hangzhou, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zhang A, Wang G, Jia L, Su T and Zhang L: Exosome-mediated microRNA-138 and vascular endothelial growth factor in endometriosis through inflammation and apoptosis via the nuclear factor-KB signaling pathway. Int J Mol Med. 43:358–370. 2019.

2 

Miller JE, Ahn SH, Monsanto SP, Khalaj K, Koti M and Tayade C: Implications of immune dysfunction on endometriosis associated infertility. Oncotarget. 8:7138–7147. 2017. View Article : Google Scholar :

3 

Sampson JA: Metastatic or embolic endometriosis, due to the menstrual dissemination of endometrial tissue into the venous circulation. Am J Pathol. 3:93–110. 1431927.PubMed/NCBI

4 

Guan YT, Huang YQ, Wu JB, Deng ZQ, Wang Y, Lai ZY, Wang HB, Sun XX, Zhu YL, Du MM, et al: Overexpression of chloride channel-3 is associated with the increased migration and invasion ability of ectopic endometrial cells from patients with endometriosis. Hum Reprod. 31:986–998. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Ibrahim SA, Hassan H and Gotte M: MicroRNA-dependent targeting of the extracellular matrix as a mechanism of regulating cell behavior. Biochim Biophys Acta. 1840:2609–2620. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Nematian SE, Mamillapalli R, Kadakia TS, Majidi Zolbin M, Moustafa S and Taylor HS: Systemic inflammation induced by microRNAs: Endometriosis-derived alterations in circulating microRNA 125b-5p and Let-7b-5p regulate macrophage cytokine production. J Clin Endocrinol Metab. 103:64–74. 2018. View Article : Google Scholar

8 

Joshi NR, Miyadahira EH, Afshar Y, Jeong JW, Young SL, Lessey BA, Serafini PC and Fazleabas AT: Progesterone resistance in endometriosis is modulated by the altered expression of MicroRNA-29c and FKBP4. J Clin Endocrinol Metab. 102:141–149. 2017.

9 

Okamoto M, Nasu K, Abe W, Aoyagi Y, Kawano Y, Kai K, Moriyama M and Narahara H: Enhanced miR-210 expression promotes the pathogenesis of endometriosis through activation of signal transducer and activator of transcription 3. Hum Reprod. 30:632–641. 2015. View Article : Google Scholar

10 

Li M, Zhou Y and Taylor HS: MiR-451a inhibition reduces established endometriosis Lesions in mice. Reprod Sci. 26:1506–1511. 2019. View Article : Google Scholar

11 

Hsu CY, Hsieh TH, Tsai CF, Tsai HP, Chen HS, Chang Y, Chuang HY, Lee JN, Hsu YL and Tsai EM: MiRNA-199a-5p regulates VEGFA in endometrial mesenchymal stem cells and contributes to the pathogenesis of endometriosis. J Pathol. 232:330–343. 2014. View Article : Google Scholar

12 

Wang WT, Zhao YN, Han BW, Hong SJ and Chen YQ: Circulating microRNAs identified in a genome-wide serum microRNA expression analysis as noninvasive biomarkers for endometriosis. J Clin Endocrinol Metab. 98:281–289. 2013. View Article : Google Scholar

13 

Hawkins SM, Creighton CJ, Han DY, Zariff A, Anderson ML, Gunaratne PH and Matzuk MM: Functional microRNA involved in endometriosis. Mol Endocrinol. 25:821–832. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Teague EM, Print CG and Hull ML: The role of microRNAs in endometriosis and associated reproductive conditions. Hum Reprod Update. 16:142–165. 2010. View Article : Google Scholar

15 

Filigheddu N, Gregnanin I, Porporato PE, Surico D, Perego B, Galli L, Patrignani C, Graziani A and Surico N: Differential expression of microRNAs between eutopic and ectopic endometrium in ovarian endometriosis. J Biomed Biotechnol. 2010:3695492010. View Article : Google Scholar : PubMed/NCBI

16 

Jia SZ, Yang Y, Lang J, Sun P and Leng J: Plasma miR-17-5p miR-20a and miR-22 are down-regulated in women with endometriosis. Hum Reprod. 28:322–330. 2013. View Article : Google Scholar

17 

Li C, Ma D, Yang J, Lin X and Chen B: MiR-202-5p inhibits the migration and invasion of osteosarcoma cells by targeting ROCK1. Oncol Lett. 16:829–834. 2018.PubMed/NCBI

18 

Zhang S, Cai J, Xie W, Luo H and Yang F: MiR-202 suppresses prostate cancer growth and metastasis by targeting PIK3CA. Exp Ther Med. 16:1499–1504. 2018.PubMed/NCBI

19 

Meng X, Chen X, Lu P, Ma W, Yue D, Song L and Fan Q: MicroRNA-202 inhibits tumor progression by targeting LAMA1 in esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 473:821–827. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Ke SB, Qiu H, Chen JM, Shi W and Chen YS: MicroRNA-202-5p functions as a tumor suppressor in colorectal carcinoma by directly targeting SMARCC1. Gene. 676:329–335. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Lin Y, Chen Z, Lin S, Zheng Y, Liu Y, Gao J and Chen S: MiR-202 inhibits the proliferation and invasion of colorectal cancer by targeting UHRF1. Acta Biochim Biophys Sin (Shanghai). 51:598–606. 2019. View Article : Google Scholar

22 

Deng X, Hou C, Liang Z, Wang H, Zhu L and Xu H: MiR-202 suppresses cell proliferation by targeting FOXR2 in endometrial adenocarcinoma. Dis Markers. 2017:28274352017. View Article : Google Scholar : PubMed/NCBI

23 

Revised American Society for Reproductive Medicine classification of endometriosis: 1996. Fertil Steril. 67:817–821. 1997. View Article : Google Scholar

24 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

25 

Sourial S, Tempest N and Hapangama DK: Theories on the pathogenesis of endometriosis. Int J Reprod Med. 2014:1795152014. View Article : Google Scholar : PubMed/NCBI

26 

Weimar CH, Macklon NS, Post Uiterweer ED, Brosens JJ and Gellersen B: The motile and invasive capacity of human endometrial stromal cells: Implications for normal and impaired reproductive function. Hum Reprod Update. 19:542–557. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Brosens JJ, Hayashi N and White JO: Progesterone receptor regulates decidual prolactin expression in differentiating human endometrial stromal cells. Endocrinology. 140:4809–4820. 1999. View Article : Google Scholar : PubMed/NCBI

28 

Chen C, Ye C, Xia J, Zhou Y and Wu R: Ezrin T567 phosphorylation regulates migration and invasion of ectopic endometrial stromal cells by changing actin cytoskeleton. Life Sci. 254:1176812020. View Article : Google Scholar : PubMed/NCBI

29 

Liu H, Zhang Z, Xiong W, Zhang L, Xiong Y, Li N, He H, Du Y and Liu Y: Hypoxia-inducible factor-1α promotes endometrial stromal cells migration and invasion by upregulating autophagy in endometriosis. Reproduction. 153:809–820. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Dai L, Gu L and Di W: MiR-199a attenuates endometrial stromal cell invasiveness through suppression of the IKKβ/NF-KB pathway and reduced interleukin-8 expression. Mol Hum Reprod. 18:136–145. 2012. View Article : Google Scholar

31 

Lv Y, Zhang L, Ma J, Fei X, Xu K and Lin J: CTHRC1 overexpression promotes ectopic endometrial stromal cell proliferation, migration and invasion via activation of the Wnt/β-catenin pathway. Reprod Biomed Online. 40:26–32. 2020. View Article : Google Scholar

32 

Meng X, Liu J, Wang H, Chen P and Wang D: MicroRNA-126-5p downregulates BCAR3 expression to promote cell migration and invasion in endometriosis. Mol Cell Endocrinol. 494:1104862019. View Article : Google Scholar : PubMed/NCBI

33 

Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, Aranda S, Palomo S, McCormick F, Izpisúa-Belmonte JC and de la Pompa JL: Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 18:99–115. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Proestling K, Birner P, Gamperl S, Nirtl N, Marton E, Yerlikaya G, Wenzl R, Streubel B and Husslein H: Enhanced epithelial to mesenchymal transition (EMT) and upregulated MYC in ectopic lesions contribute independently to endometriosis. Reprod Biol Endocrinol. 13:752015. View Article : Google Scholar : PubMed/NCBI

35 

Dinulescu DM, Ince TA, Quade BJ, Shafer SA, Crowley D and Jacks T: Role of K-ras and Pten in the development of mouse models of endometriosis and endometrioid ovarian cancer. Nat Med. 11:63–70. 2005. View Article : Google Scholar

36 

Zhou K, Luo X, Wang Y, Cao D and Sun G: MicroRNA-30a suppresses tumor progression by blocking Ras/Raf/MEK/ERK signaling pathway in hepatocellular carcinoma. Biomed Pharmacother. 93:1025–1032. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Dalpa E, Gourvas V, Soulitzis N and Spandidos DA: K-Ras, H-Ras, N-Ras and B-Raf mutation and expression analysis in Wilms tumors: Association with tumor growth. Med Oncol. 34:62017. View Article : Google Scholar

38 

Braza-Boils A, Mari-Alexandre J, Gilabert J, Sánchez-Izquierdo D, España F, Estellés A and Gilabert-Estellés J: MicroRNA expression profile in endometriosis: Its relation to angiogenesis and fibrinolytic factors. Hum Reprod. 29:978–988. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Zhu H, Cao XX, Liu J and Hua H: MicroRNA-488 inhibits endometrial glandular epithelial cell proliferation, migration, and invasion in endometriosis mice via Wnt by inhibiting FZD7. J Cell Mol Med. 23:2419–2430. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Liang Z, Chen Y, Zhao Y, Xu C, Zhang A, Zhang Q, Wang D, He J, Hua W and Duan P: MiR-200c suppresses endometriosis by targeting MALAT1 in vitro and in vivo. Stem Cell Res Ther. 8:2512017. View Article : Google Scholar : PubMed/NCBI

41 

Nothnick WB, Falcone T, Joshi N, Fazleabas AT and Graham A: Serum miR-451a levels are significantly elevated in women with endometriosis and recapitulated in baboons (Papio anubis) with experimentally-induced disease. Reprod Sci. 24:1195–1202. 2017. View Article : Google Scholar

42 

Zhang H, Li G, Sheng X and Zhang S: Upregulation of miR33b promotes endometriosis via inhibition of Wnt/β-catenin signaling and ZEB1 expression. Mol Med Rep. 19:2144–2152. 2019.PubMed/NCBI

43 

Zhao J, Ding D and Zhao G: Reduced miR-202 levels enhanced oral cancer development via targeting Sp1. Exp Ther Med. 18:489–496. 2019.PubMed/NCBI

44 

Chen J, Yin J, Liu J, Zhu RX, Zheng Y and Wang XL: MiR-202-3p functions as a tumor suppressor and reduces cell migration and invasion in papillary thyroid carcinoma. Eur Rev Med Pharmacol Sci. 23:1145–1150. 2019.PubMed/NCBI

45 

Xu F, Li H and Hu C: MiR-202 inhibits cell proliferation, invasion, and migration in breast cancer by targeting ROCK1 gene. J Cell Biochem. 120:16008–16018. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Guo L, Bai Y, Ji S and Ma H: MicroRNA98 suppresses cell growth and invasion of retinoblastoma via targeting the IGF1R/kRas/Raf/MEK/ERK signaling pathway. Int J Oncol. 54:807–820. 2019.PubMed/NCBI

47 

Deng M, Tang H, Zhou Y, Zhou M, Xiong W, Zheng Y, Ye Q, Zeng X, Liao Q, Guo X, et al: MiR-216b suppresses tumor growth and invasion by targeting KRAS in nasopharyngeal carcinoma. J Cell Sci. 124(Pt 17): 2997–3005. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Fang Y, Sun B, Li Z, Chen Z and Xiang J: MiR-622 inhibited colorectal cancer occurrence and metastasis by suppressing K-Ras. Mol Carcinog. 55:1369–1377. 2016. View Article : Google Scholar

49 

Sun W, Ping W, Tian Y, Zou W, Liu J and Zu Y: MiR-202 enhances the anti-tumor effect of cisplatin on non-small cell lung cancer by targeting the Ras/MAPK pathway. Cell Physiol Biochem. 51:2160–2171. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Steelman LS, Franklin RA, Abrams SL, Chappell W, Kempf CR, Bäsecke J, Stivala F, Donia M, Fagone P, Nicoletti F, et al: Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy. Leukemia. 25:1080–1094. 2011. View Article : Google Scholar : PubMed/NCBI

51 

Liu Z, Yi L, Du M, Gong G and Zhu Y: Overexpression of TGF-β enhances the migration and invasive ability of ectopic endometrial cells via ERK/MAPK signaling pathway. Exp Ther Med. 17:4457–4464. 2019.PubMed/NCBI

52 

Tang XL, Zhang FL, Jiang XJ and Yang XJ: Telocytes enhanced the proliferation, adhesion and motility of endometrial stromal cells as mediated by the ERK pathway in vitro. Am J Transl Res. 11:572–585. 2019.PubMed/NCBI

53 

Kim JH, Woo JH, Kim HM, Oh MS, Jang DS and Choi JH: Anti-endometriotic effects of pueraria flower extract in human endometriotic cells and mice. Nutrients. 9:2122017. View Article : Google Scholar :

Related Articles

Journal Cover

December-2020
Volume 46 Issue 6

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang D, Wang L, Guo HL, Zhang ZW, Wang C, Chian RC and Zhang ZF: MicroRNA‑202 inhibits endometrial stromal cell migration and invasion by suppressing the K‑Ras/Raf1/MEK/ERK signaling pathway Retraction in /10.3892/ijmm.2024.5375. Int J Mol Med 46: 2078-2088, 2020
APA
Zhang, D., Wang, L., Guo, H., Zhang, Z., Wang, C., Chian, R., & Zhang, Z. (2020). MicroRNA‑202 inhibits endometrial stromal cell migration and invasion by suppressing the K‑Ras/Raf1/MEK/ERK signaling pathway Retraction in /10.3892/ijmm.2024.5375. International Journal of Molecular Medicine, 46, 2078-2088. https://doi.org/10.3892/ijmm.2020.4749
MLA
Zhang, D., Wang, L., Guo, H., Zhang, Z., Wang, C., Chian, R., Zhang, Z."MicroRNA‑202 inhibits endometrial stromal cell migration and invasion by suppressing the K‑Ras/Raf1/MEK/ERK signaling pathway Retraction in /10.3892/ijmm.2024.5375". International Journal of Molecular Medicine 46.6 (2020): 2078-2088.
Chicago
Zhang, D., Wang, L., Guo, H., Zhang, Z., Wang, C., Chian, R., Zhang, Z."MicroRNA‑202 inhibits endometrial stromal cell migration and invasion by suppressing the K‑Ras/Raf1/MEK/ERK signaling pathway Retraction in /10.3892/ijmm.2024.5375". International Journal of Molecular Medicine 46, no. 6 (2020): 2078-2088. https://doi.org/10.3892/ijmm.2020.4749