Open Access

Activation of GPER by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR‑124/CD151 pathway

  • Authors:
    • Huaicheng Yang
    • Congyu Wang
    • Heqiang Liao
    • Qi Wang
  • View Affiliations

  • Published online on: March 31, 2021     https://doi.org/10.3892/ol.2021.12693
  • Article Number: 432
  • Copyright: © Yang 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

Breast cancer is one of the most common malignancies worldwide and is responsible for a high mortality rate. However, the underlying pathological mechanism of breast cancer remains unclear. MicroRNAs (miRNAs/miRs) play critical roles in the progression of breast cancer. Recent studies have reported that miR‑124/CD151 participates in the development of breast cancer. However, the exact molecular mechanism of miR‑124/CD151 action in 17β‑estradiol (E2)‑treated breast cancer cells remains unknown. Thus, the present study aimed to investigate miR‑124 and CD151 expression levels in MCF‑7 cells treated with E2 via reverse transcription‑quantitative PCR and western blot analyses. Bioinformatic analysis was performed to predict and identify whether CD151 is a potential target of miR‑124. The Cell Counting Kit‑8 and colony formation assays were performed to detect proliferation of MCF‑7 cells. In addition, the invasive and migratory abilities of MCF‑7 cells were assessed via the Transwell and wound healing assays, respectively. The results demonstrated that E2 downregulated miR‑124 expression, while upregulating G protein ‑coupled estrogen receptor (GPER) expression in MCF‑7 cells. Following treatment with the GPER antagonist, G15, miR‑124 expression was significantly enhanced and E2‑induced proliferation, invasion and migration of MCF‑7 cells were notably inhibited. In addition, CD151 was confirmed as a direct target of miR‑124. CD151 silencing remarkably suppressed the proliferation, invasion and migration of E2‑induced MCF‑7 cells. Taken together, these results suggest that upregulation of GPER expression induced by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR‑124/CD151 pathway. Thus, the results of the present study provide a potential novel method for the treatment and prognosis of breast cancer.

Introduction

Breast cancer is a lethal malignancy, which has become the second leading cause of mortality in females in the United States (1,2). A previous study has reported that the new cases of breast cancer have an increased incidence of ~30% compared with that noted in other types of cancer in women (3). Despite recent advancements in surgery, chemotherapy, radiation and hormone therapy, the 5-year survival rate of patients with breast cancer is 80–85% (46). This is mainly attributed to the lack of molecular mechanism analysis that can explain in detail the pathways responsible for breast cancer formation. Thus, detailed investigation of the pathogenesis of breast cancer is of considerable significance. It has been demonstrated that the prevention of metastasis is a vital factor for the effective reduction of breast cancer (7,8). Estrogen plays a crucial role, not only in the proliferation and initiation of breast cancer, but also in migration (9). Thus, it is essential to unravel the associated molecular mechanisms of breast cancer formation to improve the treatment and prognosis of breast carcinomas.

17β-estradiol (E2) is one of the three main self-producing estrogens and is considered an important hormone in women (10). E2 has a crucial influence on the growth and function of the female reproductive system, and of the mammary gland under physiological conditions (1113). E2 is involved in the induction of malignant transformation of several types of cancer cells via regulation of the associated biological processes, such as proliferation, invasion and migration (14,15). Most of the biological effects of estrogens are mediated via binding and activation of the classic estrogen receptors (ERs) (16). The G protein -coupled estrogen receptor (GPER) is also denoted as G protein receptor 30 (GPR30) and has been demonstrated to bind to E2 (17). GPER is responsible for interacting with multiple genomic signaling pathways in different types of cells, including breast, endometrial, ovarian and thyroid cancers (1824). Overexpression of GPER in breast cancer is positively associated with a metastatic phenotype (25). However, the molecular mechanism of proliferation, invasion and migration of breast cancer resulting from E2 binding to GPER remains largely unknown.

MicroRNAs (miRNAs/miRs) are small non-coding RNA molecules that serve as endogenous regulators of gene expression (26) and are involved in a variety of vital biological processes (2729). Previous studies have demonstrated that most miRNAs inhibit the metastasis of breast cancer and that their expression levels can be used as metastatic predictors. For example, miR-124 is considered an expression-rich miRNA (3033), which is usually expressed at low levels and is implicated in hematological malignancies and solid tumors, including breast cancer (3436). A previous study has reported that miR-124 expression is notably downregulated following treatment with E2 in ER positive breast cancer cells (37). CD151 is a member of tetraspanins that is characterized by 4 hydrophobic domains, and mediates signal transduction events associated with cell proliferation, activation and motility (38). It has been demonstrated that the CD151 protein can accelerate the progression of breast cancer (39). To the best of our knowledge, miR-124 is a negative regulator of breast cancer and it was hypothesized that E2 may regulate miR-124 and CD151 expression levels by GPER, which in turn will affect the proliferation, invasion and migration of breast cancer cells.

In the present study, the potential internal molecular mechanism was investigated with regards to the proliferation, invasion and migration of breast cancer cells. The experiments aimed to investigate the interaction between E2-GPER signaling and alteration in the expression levels of miR-124 and CD151, and the progression of breast cancer cell metastasis. The results of the present study may provide a potential novel approach to the treatment and prognosis of breast carcinomas.

Materials and methods

Cell culture and treatment

The human ER-positive breast cancer cell line, MCF-7, was purchased from Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China) and stored in liquid nitrogen. Cells were maintained in Dulbecco's modified Eagle's medium (DMEM, Invitrogen; Thermo Fisher Scientific, Inc.) supplemented with high glucose, 10% fetal bovine serum (FBS; HyClone; Cytiva) and 1% penicillin-streptomycin (Invitrogen; Thermo Fisher Scientific, Inc.), at 37°C with 5% CO2 in a humid environment.

The E2 (5, 10 or 100 nM, Sigma-Aldrich; Merck KGaA)-treated cells were maintained in complete medium with phenol red (Sigma-Aldrich; Merck KGaA) until they reached 60% confluence and subsequently replaced with 2.5% CS-FBS (HyClone; Cytiva) in DMEM containing phenol red-free for 24 h. The dose of E2 was in accordance with previous studies (40,41). Cells were cultured for 24 h at 37°C in serum-free and phenol-red free DMEM. Subsequently, a certain dose of E2 (5, 10 or 100 nM) was added into the medium. G15 was used as a well-established antagonist of GPER in this experiment (42). To determine whether E2-induced miR-124 downregulation was directly regulated by GPER, MCF-7 cells were pretreated with G15 (100 nM) for 6 h prior to addition of E2.

Cell transfection

MCF-7 cells were incubated for 24 h prior to transfection. miR-124 negative control (mimic-NC; forward, 5′-UUCUCCGAACGUGUCACGUTT−3′ and reverse, 5′-ACGUGACAGGUUCGGAGAATT−3′; 50 nM), miR-124 mimic (forward, 5′-UAAGGCACGCGGUGAAUGCCAA-3′ and reverse, 5′-CAUUCACCGCGUGCCUUAUU-3′; 50 nM), CD151-small interfering (si)RNA (50 nM) and negative control (siRNA-NC; 50 nM) sequences were synthesized by Shanghai GenePharma, Co., Ltd. Cells were seeded into 6-well plates and cultured until they reached 60–70% confluence. Transfection was performed using Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) at 37°C for 24 h, according to the manufacturer's instructions. Subsequent experiments were performed 24 h post-transfection.

Cell proliferation assay

Cell proliferation was assessed via the Cell Counting Kit-8 (CCK-8) assay (Dojindo Molecular Technologies, Inc.). The cell suspension was seeded into a 96-well plate and cultured for 6 h at 37°C. Subsequently, cells were incubated with 10 µl CCK-8 solution for 1 h and cell proliferation was measured at a wavelength of 450 nm (A450), using a microplate reader.

Colony formation assay

MCF-7 cells were seeded into 6-well plates at a density of 500 cells/well and cultured at 37°C in the presence of 5% CO2 for 7–12 days. Cells were subsequently fixed with 4% paraformaldehyde for 10 min at room temperature and stained with 0.2% crystal violet for 5 min at room temperature. The number of cell colonies were manually counted using ImageJ software (version 1.52r; National Institutes of Health).

Invasion assay

The Transwell assay was performed to assess cell invasion. A total of 1×106 MCF-7 cells were plated in the upper chambers of Transwell plates in serum-free medium (DMEM, Invitrogen; Thermo Fisher Scientific, Inc.). The Transwell membranes were pre-coated with 100 µl cell suspension and Matrigel (BD Biosciences) overnight at 4°C. DMEM supplemented with 10% FBS was plated in the lower chambers. Following incubation at 37°C with 5% CO2 for 24 h, cells that did not pass through the polycarbonate membrane were removed. The invasive cells were fixed with 4% paraformaldehyde for 30 min at room temperature, stained with 1% Giemsa for 15 min at room temperature, rinsed using phosphate-buffered saline (PBS) and air dried. All experiments were performed in triplicate. Stained cells were counted in five randomly selected fields using an inverted light microscope (Olympus Corporation; magnification, ×100) and the results were analyzed using ImageJ software (version 1.52r; National Institutes of Health).

Cell migration assay

The cell migratory ability was assessed via the wound healing assay. Briefly, cells were seeded into a 6-well plate (4×105 cells/well) and incubated at 37°C until they reached 80% confluence. Subsequently, the cell monolayers were scratched using a plastic scriber. Cells were washed with PBS to elute the debris and the media were replaced with serum-free medium. The average distance that the cells migrated into the wound surface was detected under an inverted light microscope (Olympus Corporation; magnification, ×100) after 48 h.

Reverse transcription-quantitative (RT-q)PCR

Total RNA was extracted from MCF-7 cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Single-stranded cDNA was synthesized at 42°C for 30 min using the one-step cDNA synthesis kit (cat. no. 210210; Qiagen, Inc.). The following thermocycling conditions were used: 40 cycles of pre- denaturation at 95°C for 10 min, denaturation at 95°C for 30 sec, annealing at 60°C for 20 sec and extension at 72°C for 35 sec. The following primer sequences were used: miR-124 forward, 5′-ACGTTGTGTAGCTTATCAGACTG-3′ and reverse, 5′-AATGGTTGTTCTCCACACTCTC-3′; CD151 forward, 5′-ACAGCCTACATCCTGGTGGT-3′ and reverse, 5′-TTCTCCTTGAGCTCCGTGTT-3′; U6 forward, 5′-ATTGGAACGATACAGAGAAGATT-3′ and reverse, 5′-GGAACGCTTCACGAATTTG-3′; and GAPDH forward, 5′-ACAACTTTGGTATCGTGGAAGG-3′ and reverse, 5′-GCCATCACGCCACAGTTTC-3′. Relative expression levels were calculated using the 2−ΔΔCq method (43).

Western blotting

Anti-GPER rabbit polyclonal antibody (cat. no. D161727; 1:350), goat anti-rabbit IgG antibody (cat. no. D111018; 1:8,000), anti-matrix metalloproteinase (MMP)2 (cat. no. D161447; 1:500), anti-MMP9 (cat. no. D162000; 1:500) and anti-GAPDH (cat. no. D110016; 1:500) antibodies were purchased from Sangon Biotech Co., Ltd. Cells were washed with PBS and total proteins were lysed from the cells using SDS lysis buffer (Beyotime Institute of Biotechnology) supplemented with proteinase inhibitor cocktail (Sigma-Aldrich, Merck KGaA). Total protein was quantified using the bicinchoninic acid kit (Beyotime Institute of Biotechnology) and 40 µg protein/lane was fractionated with 8% SDS-PAGE. The separated proteins were subsequently transferred onto polyvinylidene fluoride membranes (EMD Millipore) and subsequently incubated in PBS 0.2% Tween-20 containing 5% skimmed milk powder for 60 min at room temperature to block non-specific binding. Following washing, the blots were incubated overnight at 4°C with anti-GPER rabbit polyclonal antibody and GAPDH (cat. no. D110016; 1:500; Sangon Biotech Co., Ltd.) was used as the reference protein. Subsequently, the membranes were washed with PBS Tween-20 and incubated with goat anti-rabbit IgG antibody for 2 h at room temperature. Protein bands were visualized using an enhanced chemiluminescence system ChemiDoc MP (Bio-Rad Laboratories, Inc.) and quantified using ImageJ software (version 1.46; National Institutes of Health).

Dual-luciferase reporter assay

The target gene of miR-124 was predicted using the TargetScan database (http://www.targetscan.org/vert_71). A dual-luciferase reporter assay system (Promega Corporation) was used to detect luciferase activities, according to the manufacturer's instructions, using the Envision Multilabel Plate Reader (PerkinElmer, Inc.). The CD151 3′-untranslated region (UTR) was amplified from human cDNA by Shanghai GenePharma Co., Ltd., cloned into a pGL3 luciferase vector (Promega Corporation). The following sequences were used: i) Wild-type (WT) of CD151 3′-UTR forward, 5′-TCTAGAACCCAACTACTGAGCTGAGA-3′ and reverse, 5′-TCTAGAGTACAGCAGTGAACAAAACCA−3′; and ii) mutant type (MUT) of CD151 3′-UTR forward,5′-CTTCTTCCGAGTTTTGCTGCGCACCAATGC-3′ and reverse, 5′-AGCAAAACTCGGAAGAAGCTGCCTCTGAGGT-3′. Following seeding into 24-well plates and incubation overnight, cells were co-transfected with miR-124 mimic or mimic-NC for 48 h at 37°C, using Lipofectamine® 2000. The Renilla luciferase activities were normalized to the firefly luciferase activities to set the control samples for the transfection efficiency experiments. All experiments were performed in triplicate.

Statistical analysis

Statistical analysis was performed using GraphPad Prism 6.0 software. All experiments were performed in triplicate and data are presented as the mean ± standard deviation. Unpaired student's t-test was used to compare differences between two groups, while one way ANOVA and Tukey's post hoc test were used to compare differences between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

E2 induces miR-124 expression in MCF-7 cells

MCF-7 cells were treated with E2 at different time points and concentrations and miR-124 expression was detected. The results demonstrated that miR-124 expression significantly decreased following treatment with E2 compared with the control group (P<0.01, P<0.001; Fig. 1). The concentration of E2 was selected based on the lowest level of miR-124 expression. The dose-dependent experiments revealed that miR-124 expression considerably decreased in MCF-7 cells following treatment with 10 nM E2 for 8 h compared with the control group. Thus, 10 nM E2 was selected for subsequent experiments, which was in accordance with a previous study (44).

Induction of GPER by E2

MCF-7 cells were treated with 10 nM E2 at different time points (0, 6, 8 and 12 h) and GPER expression was detected. The results demonstrated that GPER expression was upregulated in MCF-7 cells following treatment with E2 for 6, 8 and 12 h, and maximum GPER expression was attained following treatment for 8 h (P<0.001; Fig. 2). Thus, cells treated with E2 for 8 h was selected for subsequent experiments.

GPER mediates the E2-induced decrease in miR-124 expression

The effects of GPER on miR-124 expression were investigated, which were induced by E2 in MCF-7 cells. G15 was used as a well-established antagonist of GPER (41) in this experiment. To determine whether E2-induced miR-124 downregulation was directly regulated by GPER, MCF-7 cells were pretreated with G15 (100 nM) for 6 h prior to addition of E2. The results demonstrated that miR-124 expression increased in the E2 + G15 group compared with the E2 group (P<0.05; Fig. 3A), suggesting that GPER mediates the E2-induced decrease of miR-124 expression.

GPER mediates the E2-induced proliferation, invasion and migration of MCF-7 cells

Cell proliferation was assessed via the CCK-8 assay. The results indicated that the proliferation of MCF-7 cells in the E2 + G15 group was significantly inhibited compared with the E2 group (P<0.01, P<0.001; Fig. 3B and C). In addition, the results of the Transwell assays demonstrated that the number of invasive cells in the E2 group was significantly higher compared with the control group, the effects of which were reversed following treatment with G15 (P<0.01; Fig. 3D and E). The wound healing assay was performed to assess cell migration. As presented in Fig. 4A and B, the ability of cell migration in the E2 group was notably enhanced compared with the control group, whereas G15 intervention attenuated the effect of E2 on cell migration. The assessment of the expression of migration-associated proteins, including matrix metalloproteinase (MMP)2 and MMP9 exhibited similar results to the cell invasion assay (Fig. 4A-C). Taken together, these results suggest that GPER mediates the E2-induced proliferation, invasion and migration of MCF-7 cells.

CD151 is a direct target of miR-124 in breast cancer cells

The TargetScan database was used to predict the target of miR-124. miR-124 was bound to the 3′-UTR of CD151 (Fig. 5A). Cells were transfected with miR-124 mimic to overexpress miR-124 (Fig. 5B). To validate whether CD151 was regulated by miR-124 in MCF-7 cells, reporter plasmids were generated (WT-CD151 3′-UTR or MUT-CD151 3′-UTR). The results demonstrated that the luciferase activity of CD151 decreased following co-transfection of miR-124 mimic with WT-CD151 3′-UTR in MCF-7 cells compared with the control mimic group. Notably, no significant differences were observed in the luciferase activity following co-transfection of miR-124 mimic with MUT-CD151 3′-UTR in MCF-7 cells (Fig. 5C). To investigate whether CD151 was regulated via the inhibition of miR-124 following GPER induced expression by E2 in MCF-7 cells, western blot and RT-qPCR analyses were performed to detect CD151 expression. In this experiment, MCF-7 cells were treated with 100 nM G15 for 2 h or pre-transfected with miR-124 mimic for 12 h prior to addition of 10 nM E2 for 8 h. The results demonstrated that CD151 protein and mRNA expression levels significantly increased in the E2 group compared with the control group (Fig. 5D and E). However, the effects of E2 on CD151 expression were blocked following pretreatment with miR-124 mimic or G15. Taken together, these results suggest that CD151 is a direct target gene of miR-124, which is consistent with previous findings (26).

Effects of CD151 on the proliferation, invasion and migration of MCF-7 cells following treatment with E2

To investigate the proliferation, invasion and migration of E2-treated MCF-7 cells, cells were transfected with CD151-siRNA. RT-qPCR analysis was performed to verify CD151 knockdown in MCF-7 cells and the results demonstrated that CD151 expression significantly decreased following transfection with CD151-siRNA (Fig. 6A). The effect of CD151 knockdown on cell proliferation was assessed via the CCK-8 assay. The results demonstrated that cell proliferation significantly decreased in the CD151-siRNA group compared with the control group (Fig. 6B and C). The invasive (Fig. 6D and E) and migratory (Fig. 7A and B) abilities of MCF-7 cells following CD151 interference were assessed via the Transwell and wound healing assays, respectively. The results demonstrated that cell invasion and migration significantly decreased following transfection with CD151-siRNA. Collectively, these results suggest that the proliferation, invasion and migration of E2-induced MCF-7 cells are inhibited following CD151 interference.

Discussion

Breast cancer is one of the most common malignancies worldwide that is responsible for a high number of fatalities (45). The number of new breast cancer cases is estimated at 272,400 in China, annually (46). The identification of novel and efficacious methods for early diagnosis and treatment of this disease remains a major challenge. In the present study, the molecular mechanism of the proliferation, invasion and migration of breast cancer cells was investigated.

Estrogens play an important role in breast cancer metastasis (4749). E2 is one of the typical estrogens that is responsible for the occurrence of 70–80% of human breast cancer and has been generally investigated for its contribution in the development of the human mammary gland (50). It is considered that GPER can mediate estrogenic signals in breast cancer (51). Previous studies have demonstrated that GPER is bound with E2, owing to its high affinity (20,52,53). Thus, it is important to investigate the effects of E2-GPER signaling on the proliferation, invasion and migration of breast cancer cells. A previous study reported that GPER expression is upregulated in patients with breast cancer (54). Another study indicated that E2-treated cancer-associated fibroblasts exhibit a positive feedback behavior, which involves GPER/EGFR/ERK signaling and E2 production, and contributes to the progression of breast cancer (55). Breast cancer progression is associated with overexpression of GPER and PM localization, whereas the lack of GPER is associated with the long-term prognosis of primary breast cancer in PM (56). However, the molecular mechanism underlying the proliferation, invasion and migration of breast cancer cells via E2-GPER signaling remains largely unknown. The results of the present study demonstrated that GPER-E2 signaling decreased miR-124 expression, suggesting that inhibition of this target may be used as a novel treatment in breast carcinoma.

miRNAs are important post-transcriptional regulators of gene expression that play a major role in carcinogenesis (28,29). It has been demonstrated that miR-124 acts as a tumor suppressor that can inhibit the proliferation, invasion and migration of breast cancer cells (37). A previous study reported that miR-124 targets Slug to regulate epithelial-to-mesenchymal transition and metastasis of breast cancer (57). Feng et al (58) discovered that downregulation of miR-124 results in breast cancer progression via regulation of long non-coding RNA-MALAT1 and activation of CDK4/E2F1 signaling. In addition, another study demonstrated that downregulation of miR-124-3p promotes breast cancer progression by targeting Beclin-1 (59). Thus, it is crucial to investigate the expression levels and regulation of miR-124 in breast cancer. Taken together, the results of the present study demonstrated that E2 inhibited miR-124 expression via the receptor, GPER. Furthermore, E2-GPER signaling promoted CD151 expression by suppressing miR-124 expression.

In conclusion, the present study demonstrated that activation of GPER was mediated by E2 and promoted the proliferation, invasion and migration of breast cancer cells via regulation of the miR-124/CD151 pathway. These findings may provide a novel therapeutic strategy for the treatment and prognosis of breast cancer. However, use of a single breast cancer cell line is a major limitation of the present study. Thus, prospective studies will focus on using different clinical types of breast cancer cells.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and or/analyzed during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

HY and CW designed and performed the experiments. HL drafted the initial manuscript. HY, HL and QW contributed to data collection, analysis and interpretation. QW reviewed and edited the manuscript. HY and QW confirmed the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

2 

DeSantis CE, Ma J, Goding Sauer A, Newman LA and Jemal A: Breast cancer statistics, 2017, racial disparity in mortality by state. CA Cancer J Clin. 67:439–448. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Bartelink H, Horiot JC, Poortmans P, Struikmans H, Van den Bogaert W, Barillot I, Fourquet A, Borger J, Jager J, Hoogenraad W, et al: Recurrence rates after treatment of breast cancer with standard radiotherapy with or without additional radiation. N Engl J Med. 345:1378–1387. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Mattiuzzi C and Lippi G: Current cancer epidemiology. J Epidemiol Glob Health. 9:217–222. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Clayforth C, Fritschi L, McEvoy SP, Byrne MJ, Ingram D, Sterrett G, Harvey JM, Joseph D and Jamrozik K: Five-year survival from breast cancer in Western Australia over a decade. Breast. 16:375–381. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Yuan J, Xiao C, Lu H, Yu H, Hong H, Guo C and Wu Z: miR-200b regulates breast cancer cell proliferation and invasion by targeting radixin. Exp Ther Med. 19:2741–2750. 2020.PubMed/NCBI

8 

Si CS, Yu Q and Yao YF: Effect of miR-146a-5p on proliferation and metastasis of triple-negative breast cancer via regulation of SOX5. Exp Ther Med. 15:4515–4521. 2018.PubMed/NCBI

9 

Yager JD and Davidson NE: Estrogen carcinogenesis in breast cancer. N Engl J Med. 354:270–282. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Holden MP and Hampson E: Endogenous variation in estradiol in women affects the weighting of metric and categorical information in spatial location memory. Horm Behav. 128:62021. View Article : Google Scholar

11 

Mantri AV, Allaway HCM, Brezicha JE, Hogan HA and Bloomfield SA: Oral estradiol impact on mitigating unloading-induced bone loss in ovary-intact rats. Aerosp Med Hum Perform. 92:65–74. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Fang GZ, Huang GY, Ying GG, Qiu SQ, Shi WJ, Xie L, Yang YY and Ma DD: Endocrine disrupting effects of binary mixtures of 17β-estradiol and testosterone in adult female western mosquitofish (Gambusia affinis). Ecotox Environ Safe. 208:1115662021. View Article : Google Scholar

13 

Huang S, Liu M, Fu F, Liu H, He B, Xiao D and Yang J: High serum estradiol reduces acute hepatotoxicity risk induced by epirubicin plus cyclophosphamide chemotherapy in premenopausal women with breast cancer. Front Pharmacol. 11:5724442021. View Article : Google Scholar : PubMed/NCBI

14 

Shang D, Li Z, Zhu Z, Chen H, Zhao L, Wang X and Chen Y: Baicalein suppresses 17-β-estradiol-induced migration, adhesion and invasion of breast cancer cells via the G protein -coupled receptor 30 signaling pathway. Oncol Rep. 33:2077–2085. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Ignatov A, Ignatov T, Roessner A, Costa SD and Kalinski T: Role of GPR30 in the mechanisms of tamoxifen resistance in breast cancer MCF-7 cells. Breast Cancer Res Treat. 123:87–96. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Muhammad JS, Guimei M, Jayakumar MN, Shafarin J, Janeeh AS, AbuJabal R, Eladl MA, Ranade AV, Ali A and Hamad M: Estrogen-induced hypomethylation and overexpression of YAP1 facilitate breast cancer cell growth and survival. Neoplasia. 23:68–79. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Feng G, Cai J, Huang Y, Zhu X, Gong B, Yang Z, Yan C, Hu Z, Yang L and Wang Z: G-protein-coupled estrogen receptor 1 promotes gender disparities in hepatocellular carcinoma via modulation of SIN1 and mTOR complex 2 activity. Mol Cancer Res. 18:1863–1875. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Maggiolini M, Vivacqua A, Fasanella G, Recchia AG, Sisci D, Pezzi V, Montanaro D, Musti AM, Picard D and Andò S: The G protein -coupled receptor GPR30 mediates c-fos up-regulation by 17beta-estradiol and phytoestrogens in breast cancer cells. J Biol Chem. 279:27008–27016. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Vivacqua A, Bonofiglio D, Recchia AG, Musti AM, Picard D, Andò S and Maggiolini M: The G protein - coupled receptor GPR30 mediates the proliferative effects induced by 17beta-estradiol and hydroxytamoxifen in endometrial cancer cells. Mol Endocrinol. 20:631–646. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Revankar CM, Cimino DF, Sklar LA, Arterburn JB and Prossnitz ER: A transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science. 307:1625–1630. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Filardo EJ, Quinn JA, Bland KI and Frackelton AR Jr: Estrogen-induced activation of Erk-1 and Erk-2 requires the G protein -coupled receptor homolog, GPR30, and occurs via trans-activation of the epidermal growth factor receptor through release of HB-EGF. Mol Endocrinol. 14:1649–1660. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Albanito L, Madeo A, Lappano R, Vivacqua A, Rago V, Carpino A, Oprea TI, Prossnitz ER, Musti AM, Andò S and Maggiolini M: G protein -coupled receptor 30 (GPR30) mediates gene expression changes and growth response to 17beta- estradiol and selective GPR30 ligand G-1 in ovarian cancer cells. Cancer Res. 67:1859–1866. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Vivacqua A, Bonofiglio D, Albanito L, Madeo A, Rago V, Carpino A, Musti AM, Picard D, Andò S and Maggiolini M: 17beta-estradiol, genistein, and 4-hydroxytamoxifen induce the proliferation of thyroid cancer cells through the G protein -coupled receptor GPR30. Mol Pharmacol. 70:1414–1423. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Thomas P, Pang Y, Filardo EJ and Dong J: Identity of an estrogen membrane receptor coupled to a G protein in human breast cancer cells. Endocrinology. 146:624–632. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Liu Q, Li JG, Zheng XY, Jin F and Dong HT: Expression of CD133, PAX2, ESA, and GPR30 in invasive ductal breast carcinomas. Chin Med J (Engl). 122:2763–2769. 2009.PubMed/NCBI

26 

Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell. 136:215–233. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Liu S, Chen X, Zhang S, Wang X, Du X, Chen J and Zhou G: miR-106b-5p targeting SIX1 inhibits TGF-β1-induced pulmonary fibrosis and epithelial-mesenchymal transition in asthma through regulation of E2F1. Int J Mol Med. 47:12021. View Article : Google Scholar

28 

Wan JH and Liu SS: miR-613 inhibits the proliferation of human ovarian granulosa cells by arresting cell cycle progression via the targeting of IGF-1. Mol Med Rep. 23:12021.

29 

Dong B, Li SY, Zhu SL, Yi M, Luo S and Wu K: miRNA- mediated EMT and CSCs in cancer chemoresistance. Exp Hematol Oncol. 10:122021. View Article : Google Scholar : PubMed/NCBI

30 

Cheng LC, Pastrana E, Tavazoie M and Doetsch F: miR-124 regulates adult neurogenesis in the subventricular zone stem cell niche. Nat Neurosci. 12:399–408. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Yu JY, Chung KH, Deo M, Thompson RC and Turner DL: MicroRNA miR-124 regulates neurite outgrowth during neuronal differentiation. Exp Cell Res. 314:2618–2633. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Makeyev EV, Zhang J, Carrasco MA and Maniatis T: The MicroRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Mol Cell. 27:435–448. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Mishima T, Mizuguchi Y, Kawahigashi Y and Takizawa T and Takizawa T: RT-PCR-based analysis of microRNA (miR-1 and-124) expression in mouse CNS. Brain Res. 1131:37–43. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Silber J, Lim DA, Petritsch C, Persson AI, Maunakea AK, Yu M, Vandenberg SR, Ginzinger DG, James CD, Costello JF, et al: miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells. BMC Med. 6:142008. View Article : Google Scholar : PubMed/NCBI

35 

Wong KY, So CC, Loong F, Chung LP, Lam WW, Liang R, Li GK, Jin DY and Chim CS: Epigenetic inactivation of the miR-124-1 in haematological malignancies. PLoS One. 6:e190272011. View Article : Google Scholar : PubMed/NCBI

36 

Xia J, Wu Z, Yu C, He W, Zheng H, He Y, Jian W, Chen L, Zhang L and Li W: miR-124 inhibits cell proliferation in gastric cancer through down-regulation of SPHK1. J Pathol. 227:470–480. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Jiang CF, Li DM, Shi ZM, Wang L, Liu MM, Ge X, Liu X, Qian YC, Wen YY, Zhen LL, et al: Estrogen regulates miRNA expression: Implication of estrogen receptor and miR-124/AKT2 in tumor growth and angiogenesis. Oncotarget. 7:36940–36955. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Wang J, Lei W, Li G, Ma H, Guo H and Li S: CD151 promotes proliferation and migration of SK-NEP-1 cells via the GSK-3β/P21/cyclinD signaling pathway. Pathol Res Pract. 215:329–334. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Han ZB, Yang Z, Chi Y, Zhang L, Wang Y, Ji Y, Wang J, Zhao H and Han ZC: MicroRNA-124 suppresses breast cancer cell growth and motility by targeting CD151. Cell Physiol Biochem. 31:823–832. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Li Q, Gao H, Yang H, Wei W and Jiang Y: Estradiol promotes the progression of ER+ breast cancer through methylation- mediated RSK4 inactivation. Onco Targets Ther. 12:5907–5916. 2019. View Article : Google Scholar : PubMed/NCBI

41 

Khan S, Abdelrahim M, Samudio I and Safe S: Estrogen receptor/Sp1 complexes are required for induction of cad gene expression by 17beta-estradiol in breast cancer cells. Endocrinology. 144:2325–2335. 2003. View Article : Google Scholar : PubMed/NCBI

42 

Dennis MK, Burai R, Ramesh C, Petrie WK, Alcon SN, Nayak TK, Bologa CG, Leitao A, Brailoiu E, Deliu E, et al: In vivo effects of a GPR30 antagonist. Nat Chem Biol. 5:421–427. 2009. View Article : Google Scholar : PubMed/NCBI

43 

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 : PubMed/NCBI

44 

Hayashi T, Hikichi M, Yukitake J, Harada N and Utsumi T: Estradiol suppresses phosphorylation of ERα serine 167 through upregulation of PP2A in breast cancer cells. Oncol Lett. 14:8060–8065. 2017.PubMed/NCBI

45 

Emens LA: Breast cancer immunotherapy: Facts and hopes. Clin Cancer Res. 24:511–520. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Chen WQ, Zheng RS, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Yuan S, Huang C, Ji X, Ma M, Rao K and Wang Z: Prediction of the combined effects of multiple estrogenic chemicals on MCF-7 human breast cancer cells and a preliminary molecular exploration of the estrogenic proliferative effects and related gene expression. Ecotoxicol Environ Saf. 160:1–9. 2018. View Article : Google Scholar : PubMed/NCBI

48 

Parl FF, Crooke PS, Plummer WD Jr and Dupont WD: Genomic-epidemiologic evidence that estrogens promote breast cancer development. Cancer Epidemiol Biomarkers Prev. 27:899–907. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Sampayo RG, Toscani AM, Rubashkin MG, Thi K, Masullo LA, Violi IL, Lakins JN, Cáceres A, Hines WC, Coluccio Leskow F, et al: Fibronectin rescues estrogen receptor a from lysosomal degradation in breast cancer cells. J Cell Biol. 217:2777–2798. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Meneses-Morales I, Tecalco-Cruz AC, Barrios-García T, Gómez-Romero V, Trujillo-González I, Reyes-Carmona S, García-Zepeda E, Méndez-Enríquez E, Cervantes-Roldán R, Pérez-Sánchez V, et al: SIP1/NHERF2 enhances estrogen receptor alpha transactivation in breast cancer cells. Nucleic Acids Res. 42:6885–6900. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Liang S, Chen Z, Jiang G, Zhou Y, Liu Q, Su Q, Wei W, Du J and Wang H: Activation of GPER suppresses migration and angiogenesis of triple negative breast cancer via inhibition of NF-κB/IL-6 signals. Cancer Lett. 386:12–23. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Moriarty K, Kim KH and Bender JR: Minireview: Estrogen receptor-mediated rapid signaling. Endocrinology. 147:5557–5563. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Prossnitz ER, Arterburn JB and Sklar LA: GPR30: A G protein -coupled receptor for estrogen. Mol Cell Endocrinol. 265-266:138–142. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Girgert R, Emons G and Gründker C: Inhibition of growth hormone receptor by Somavert reduces expression of GPER and prevents growth stimulation of triple-negative breast cancer by 17β-estradiol. Oncol Lett. 15:9559–9566. 2018.PubMed/NCBI

55 

Luo H, Yang G, Yu T, Luo S, Wu C, Sun Y, Liu M and Tu G: GPER-mediated proliferation and estradiol production in breast cancer-associated fibroblasts. Endocr Relat Cancer. 21:355–369. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Yu T, Liu M, Luo H, Wu C, Tang X, Tang S, Hu P, Yan Y, Wang Z and Tu G: GPER mediates enhanced cell viability and motility via non-genomic signaling induced by 17β- estradiol in triple-negative breast cancer cells. J Steroid Biochem Mol Biol. 143:392–403. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Liang YJ, Wang QY, Zhou CX, Yin QQ, He M, Yu XT, Cao DX, Chen GQ, He JR and Zhao Q: miR-124 targets Slug to regulate epithelial-mesenchymal transition and metastasis of breast cancer. Carcinogenesis. 34:713–722. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Feng T, Shao F, Wu Q, Zhang X, Xu D, Qian K, Xie Y, Wang S, Xu N, Wang Y and Qi C: miR-124 downregulation leads to breast cancer progression via LncRNA-MALAT1 regulation and CDK4/E2F1 signal activation. Oncotarget. 7:16205–16216. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Zhang F, Wang B, Long H, Yu J, Li F, Hou H and Yang Q: Decreased miR-124-3p expression prompted breast cancer cell progression mainly by targeting Beclin-1. Clin Lab. 62:1139–1145. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2021
Volume 21 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang H, Wang C, Liao H and Wang Q: Activation of GPER by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR‑124/CD151 pathway. Oncol Lett 21: 432, 2021
APA
Yang, H., Wang, C., Liao, H., & Wang, Q. (2021). Activation of GPER by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR‑124/CD151 pathway. Oncology Letters, 21, 432. https://doi.org/10.3892/ol.2021.12693
MLA
Yang, H., Wang, C., Liao, H., Wang, Q."Activation of GPER by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR‑124/CD151 pathway". Oncology Letters 21.6 (2021): 432.
Chicago
Yang, H., Wang, C., Liao, H., Wang, Q."Activation of GPER by E2 promotes proliferation, invasion and migration of breast cancer cells by regulating the miR‑124/CD151 pathway". Oncology Letters 21, no. 6 (2021): 432. https://doi.org/10.3892/ol.2021.12693