MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells

Retraction in: /10.3892/etm.2021.10603

  • Authors:
    • Qiongyan Zou
    • Wenjun Yi
    • Jianghai Huang
    • Fenfen Fu
    • Gannong Chen
    • Dewu Zhong
  • View Affiliations

  • Published online on: June 13, 2017     https://doi.org/10.3892/etm.2017.4593
  • Pages: 1198-1204
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs (miRs) are a type of small non-coding RNA that serve crucial roles in the development and progression of breast cancer. However, the exact role and underlying molecular mechanism of miR‑375 in mediating the growth and metastasis of breast cancer remains unknown. In the present study, reverse transcription‑quantitative polymerase chain reaction and western blot analysis were conducted to examine RNA and protein expression. A luciferase reporter assay was performed to determine the association between miR‑375 and paired box 6 (PAX6). The results of the current study indicate that the expression of miR‑375 was reduced in breast cancer tissues compared with matched adjacent normal tissues. Transfection with miR‑375 mimics led to a significant increase in levels of miR‑375 in human breast cancer Michigan Cancer Foundation (MCF)‑7 cells (P<0.05). The increase in miR‑375 expression caused a significant decrease in the viability, migration and invasion of MCF‑7 cells (P<0.05), accompanied by a reduced expression of matrix metalloproteinase (MMP) 2 and MMP9 proteins. Luciferase reporter assay identified PAX6 as a novel target of miR‑375 and miR‑375 in turn, negatively regulated the protein expression of PAX6 in MCF‑7 cells. By contrast, overexpression of PAX6 led to a significant increase in MCF‑7 cell viability (P<0.01) but did not affect the migration and invasion of MCF‑7 cells, suggesting that the inhibitory effect of miR‑375 on MCF‑7 cell viability may be occurring, in part, via the direct targeting of PAX6.

Introduction

In females, the most commonly diagnosed type of cancer is breast cancer, which accounts for 23% of total cancer cases and 14% of cancer-associated mortality (1,2). Due to improved therapeutic strategies, the mortality rate for breast cancer has decreased markedly. However, it remains a leading cause of cancer-related mortality in females worldwide (1,2). The deregulation of numerous oncogenes or tumor suppressors has been demonstrated to participate in the development and progression of breast cancer (3). Therefore, studying the specific roles of these genes, as well as their regulatory mechanisms, is promising for the development of effective therapeutic strategies to treat breast cancer.

MicroRNAs (miRs), a type of endogenous small non-coding RNA containing 22–25 nucleotides, are able to suppress the expression of genes by directly binding to 3′-untranslated region (UTR) of their target mRNAs, either causing translational repression or leading to mRNA degradation (4). By negatively mediating their target genes, miRs participate in a variety of cellular biological processes including cell growth and proliferation, survival and apoptosis, differentiation, migration and invasion (58). Furthermore, it has been demonstrated that the deregulation of certain miRs, such as miR-375, are involved in tumorigenesis and tumor metastasis (911). Shi et al (12) demonstrated that miR-375 served a suppressive role in osteosarcoma by targeting the PIK3CA gene. Shen et al (13) suggested that miR-375 may promote the epithelial-mesenchymal transition, resulting in the development of chemo-resistance in cervical cancer cells. Accordingly, miR-375 may exhibit the functions of an oncogene or tumor suppressor in different human cancers and its exact role is tumor-specific. Currently, the underlying molecular mechanism by which miR-375 affects the biological processes of breast cancer cells remains largely unknown.

Paired box (PAX) 6, a member of the PAX family, is an important transcription factor associated with the development of the eyes, pancreas and central nervous system (14,15). Furthermore, it has been established that deregulated PAX6 is involved different types of human cancer, including non-small cell lung cancer (16), retinoblastoma (17), glioma (18), colorectal cancer (19), prostate cancer (20) and breast cancer (21). Meng et al (21) demonstrated that miR-335 inhibited cell cycle progression, colony formation, proliferation and invasion by targeting PAX6 in breast cancer cells. This suggests that PAX6 may be used as a potential target for the treatment of breast cancer. Other miRs that are potentially involved in the regulation of breast cancer by targeting PAX6 have not yet been studied. Therefore, the present study aimed to investigate the role and molecular mechanism of miR-375 in the regulation of breast cancer growth and metastasis in vitro.

Materials and methods

Tissue collection

Breast cancer tissues (n=15) and their matched adjacent normal tissues (n=15) were obtained from 15 female breast cancer patients (44 to 67 years old) admitted to the Xiangya Hospital of Central South University (Changsha, China) between January and June 2014. Tissues were immediately snap-frozen in liquid nitrogen following surgical resection and stored at −80°C until use. The present study was approved by the Ethical Committee of Xiangya Hospital of Central South University and written informed consent was obtained from all patients.

Cell culture

The human breast cancer cell line Michigan Cancer Foundation (MCF)-7 was purchased from the American Type Culture Collection (Manassas, VA, USA) and cultured in passage in Dulbecco's modified Eagle's medium (DMEM; Thermo Fisher Scientific, Inc. Waltham, MA, USA) with 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Inc.) at 37°C with 5% CO2.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA of MCF-7 cells was isolated using TRIzol® Reagent (Thermo Fisher Scientific, Inc.), in accordance with the manufacturer's protocol. In order to detect miR-375 expression, total RNA was reverse transcribed using the miScript II RT kit (Qiagen, Hilden, Germany) in accordance with manufacturer's protocol. qPCR was then conducted using the miScript SYBR Green PCR kit (Qiagen). All reactions were performed using an ABI 7500 PCR thermocycler (Thermo Fisher Scientific, Inc.). Primers were purchased from Yearthbio, Changsha, China and the primer sequences were commercially unavailable. The reaction conditions were as follows: 95°C for 5 min, followed by 40 cycles of 95°C for 10 sec and 60°C for 30 sec. Relative mRNA expression levels were calculated using the 2−ΔΔCq method (22). The relative expression of miR was normalized using U6.

Cell transfection

MCF-7 cells were transfected with miR-375 mimics (Thermo Fisher Scientific, Inc.) or scrambled miR as a negative control (miR-NC; Thermo Fisher Scientific, Inc.) using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Briefly, MCF-7 cells were cultured to 70% confluence. miR-375 mimics, miR-NC, PAX6 plasmid (Yearthbio) or Lipofectamine® 2000 were diluted with serum-free DMEM, respectively. The diluted 100 nM Lipofectamine® 2000 was then mixed with the diluted miR and incubated at room temperature for 20 min prior to being added to the cell medium. Cells were incubated at 37°C in 5% CO2 for 6 h. The medium in each well of 6-well plates was then replaced by the normal serum-containing DMEM medium and cultured for 24 h prior to subsequent assays.

Western blot analysis

Cells were lysed and protein was isolated in radioimmunoprecipitation assay buffer (Thermo Fisher Scientific, Inc.). The concentration of protein was quantified using a bicinchoninic acid protein assay kit (Santa Cruz Biotechnology, Inc., Dallas, TX, USA), according to the manufacturer's instructions. A total of 50 µg protein from each sample was separated using 12% SDS-PAGE and transferred onto a polyvinylidene difluoride membrane (Thermo Fisher Scientific, Inc.). The membrane was incubated with Tris-buffered saline and Tween-20 containing 5% skimmed milk (China Mengniu Dairy Company Limited, Hong Kong, China) at 37°C for 2 h. The membrane was then incubated with mouse anti-PAX6 monoclonal antibody (1:100; ab197768; Abcam, Cambridge, MA, USA), mouse anti-MMP2 monoclonal antibody (1:50; ab86607; Abcam), mouse anti-MMP9 monoclonal antibody (1:50; ab58803; Abcam) or mouse anti-GAPDH monoclonal antibody (1:400; ab8245; Abcam) at room temperature for 3 h, respectively. The membrane was then washed four times with phosphate buffered saline (PBS) and Tween 20 (PBST; Sigma-Aldrich; Merck kGaA, Darmstadt, Germany), for 10 min. The membrane was incubated with the goat anti-mouse secondary antibody (1:5,000; A4416; Sigma-Aldrich; Merck kGaA) at 4°C overnight and washed a further four times for 10 min with PBST. An Enhanced ECL Chemiluminescent Substrate Kit (Pierce Chemical, Dallas, TX, USA) was used to perform chemiluminescent detection, according to the manufacturer's instructions. Image-Pro plus software version 6.0 (Media Cybernetics, Inc., Rockville, MD, USA) was used to analyze the relative protein expression, presented as the density ratio vs. GAPDH.

Luciferase reporter assay

The wild-type (WT) or mutant type (MUT) 3′-UTR of PAX6 was inserted downstream of the luciferase reporter gene in the pMIR-REPORT vector (Thermo Fisher Scientific, Inc.), generating WT-PAX6 and MUT-PAX6. MCF-7 cells were co-transfected with miR-375 mimics/miR-NC, WT-PAX6/MUT-PAX6 and pRL-SV40 (Promega Corporation, Madison, WI, USA) expressing Renilla luciferase. Following 48 h incubation at 37°C with 5% CO2, the luciferase activity was measured using the Dual-Luciferase® Reporter Assay System (Promega Corporation). Cells in the control group were co-transfected with WT-PAX6/MUT-PAX6 and pRL-SV40 expressing Renilla luciferase.

Cell viability analysis

To measure cell viability, an MTT assay was performed. At 72 h post-transfection, a 100 µl cell suspension (5,000 cells/ml) was seeded into 96-well plates and incubated at 37°C with 5% CO2 for 12, 24, 48 or 72 h. For the MTT assay, 100 µl fresh serum-free DMEM with 0.5 g/l MTT replaced the transfection medium in each well. Following 4 h incubation at 37°C, the MTT medium was removed by aspiration and 50 µl dimethyl sulfoxide was added to each well. Formazan production was detected following 10 min at room temperature using an ELx800 Absorbance Reader (BioTek Instruments, Inc., Winooski, VT, USA) to measure the optical density at a wavelength of 570 nm.

Cell migration assay

To examine the cell migratory capacity, 1×105 cells/well MCF-7 cells in each group were cultured in DMEM with 10% FBS to 100% confluence. Wounds of ~1 mm width were created with a plastic scriber and cells were washed once using PBS. Following 48 h culture at 37°C with 5% CO2, MCF-7 cells were observed under an inverted microscope (Olympus Corporation, Tokyo, Japan).

Cell invasion assay

Transwell Chambers pre-coated with Matrigel (BD Bioscience, San Jose, CA, USA) were used to examine cell invasion capacity. Briefly, a suspension of MCF-7 cells (5×105 cells/ml) in each group was prepared in DMEM. A total of 300 µl cell suspension was added to the upper chamber, while 500 µl DMEM with 10% FBS was added to the lower chamber. Following 24 h incubation at 37°C with 5% CO2, cells that did not invade through the pores were wiped out using a cotton-tipped swab. The filters were stained using 0.1% crystal violet and the invasive cell number was determined from five randomly selected fields under an inverted microscope (Olympus Corporation).

Statistical analysis

All data were expressed as mean ± standard deviation of triplicate experiments. Statistical analysis was performed using SPSS 17.0 statistical software (SPSS, Inc., Chicago, IL, USA). Data was analyzed by one-way analysis of variance followed by Tukey post hoc test or Student's t-test and P<0.05 was considered to represent a statistically significant difference.

Results

miR-375 is significantly downregulated in breast cancer

To reveal the exact role of miR-375 in breast cancer, RT-qPCR was conducted to examine the expression of miR-375 in breast cancer tissues as well as their matched normal adjacent tissues. It was determined that miR-203 expression was significantly decreased in breast cancer tissues compared with their matched normal adjacent tissues (P<0.01; Fig. 1), suggesting that miR-375 may serve a suppressive role in breast cancer.

miR-375 negatively mediates the viability, migration and invasion of breast cancer cells

MCF-7 cells were used to investigate the role of miR-375 in breast cancer in vitro. MCF-7 cells were transfected with miR-375 mimic or miR-375 inhibitor. Following transfection, RT-qPCR was conducted to examine levels of miR-375. Transfection with miR-375 mimic led to a significant increase in miR-375 levels (P<0.01), while transfection with miR-375 inhibitor led to a significant decrease in the level of miR-375 in MCF-7 cells (P<0.01; Fig. 2A).

An MTT assay was conducted to determine the cell viability rate in each group. Overexpression of miR-375 significantly inhibited MCF-7 cell viability (P<0.01). By contrast, downregulation of miR-375 significantly promoted MCF-7 cell viability (P<0.01; Fig. 2B), suggesting that miR-375 may serve a suppressive role in breast cancer growth.

Tumor cell migration and invasion are two key processes that occur during cancer metastasis, therefore the effects of miR-375 on breast cancer cell migration and invasion were further examined. Wound scratch assay data indicated that, compared with the control group, overexpression of miR-375 significantly suppressed MCF-7 cell migration (P<0.01), while inhibition of miR-375 led to a significant increase in the migration of MCF-7 cells (P<0.01; Fig. 3A). Furthermore, the transwell migration assay data indicated that overexpression of miR-375 significantly inhibited MCF-7 cell invasion (P<0.01), while knockdown of miR-375 markedly enhanced MCF-7 cell migration, when compared to the control group (P<0.05; Fig. 3B). This suggests that miR-375 may have a suppressive effect on breast cancer metastasis. Moreover, overexpression of miR-375 significantly reduced the MMP2 and MMP9 protein expression (P<0.01; Fig. 3C), while inhibition of miR-375 promoted the protein expression of MMP2 and MMP9 in MCF-7 cells (P<0.01; Fig. 3C).

PAX6 is a direct target of miR-375

As predicted by the online software Targetscan (http://www.targetscan.org/vert_61/) PAX6 is a putative target of miR-375 (Fig. 4A) and this targeting association is evolutionally conserved (Fig. 4B). To determine whether PAX6 was a direct target of miR-375, the WT 3′-UTR or MUT of PAX6 mRNA was inserted downstream of the luciferase reporter gene in an apMIR-REPORT vector (Fig. 4C). A luciferase reporter assay was then performed. In miR-375 and WT PAX6 3′-UTR co-transfected MCF-7 cells, the Renilla/firefly value of luciferase was significantly reduced compared with the control group (P<0.01; Fig. 4D). However, in MCF-7 cells co-transfected with miR-375 mimic and MUT 3′UTR of PAX6, the Renilla/firefly value of luciferase did not decrease (Fig. 4D), indicating that miR-375 does not bind to the MUT 3′UTR of PAX6. The results suggest that PAX6 is a novel target of miR-375.

miR-375 negatively regulates the protein expression of PAX6 in MCF-7 cells

As miRs typically inhibit the expression of their target genes at a post-transcriptional level, the effects of miR-375 overexpression and downregulation on the expression of PAX6 protein in MCF-7 cells was investigated. RT-qPCR and western blot analysis were performed to examine levels of PAX6 mRNA and protein in miR-375-upregulating or -downregulating MCF-7 cells. miR-375 did not affect expression of PAX6 mRNA in MCF-7 cells (Fig. 5A). However, levels of PAX6 protein were significantly reduced following upregulation of miR-375 (P<0.01) and significantly increased following miR-375 knockdown in MCF-7 cells (P<0.01; Fig. 5B). These findings indicate that miR-375 negatively mediates the expression of PAX6 at the post-transcriptional level in breast cancer cells.

Overexpression of PAX6 inhibits cell viability but has no effect on cell migration and invasion in MCF-7 cells

Based the aforementioned results of the present study, the inhibitory effects of miR-375 on MCF-7 cell viability, migration and invasion may occur via inhibition of PAX6. Thus, MCF-7 cells were transfected with a PAX6 plasmid to upregulate PAX6 expression. Following transfection, western blot analysis data indicated that the expression of PAX6 protein was significantly increased compared with the control group (P<0.01; Fig. 6A). Furthermore, MTT assay data indicated that overexpression of PAX6 significantly promoted MCF-7 cell viability compared to the control group (P<0.01; Fig. 6B). This indicates that PAX6 is involved in the regulation of miR-375-mediated MCF-7 cell viability. However, overexpression of PAX6 did not affect the migration and invasion of MCF-7 cells compared with the control group (Fig. 7). Therefore, PAX6 may primarily regulate breast cancer growth but not metastasis and the suppressive role of miR-375 in the regulation of MCF-7 cell migration and invasion may occur due to mediation of other targets.

Discussion

miRs act as key regulators in different types of human cancer by negatively mediating their target genes, which are oncogenes or tumor suppressors (911). Deregulated miR-375 has been identified in numerous cancer types, including laryngeal squamous cell carcinoma (23), head and neck squamous cell carcinoma (24), prostate cancer (25), hepatocellular carcinoma (26), lung cancer (27) and breast cancer (28). Wu et al (29) examined the circulating miR profile in patients with breast cancer by deep sequencing all circulating small RNAs. Circulating miR-375 and miR-122 demonstrated a strong association with the clinical outcome, including neoadjuvant chemotherapy responses and relapse with metastatic disease (29), suggesting that miR-375 is involved in breast cancer.

The suppressive role of miR-375 in breast cancer has been determined. Ye et al (30) demonstrated that miR-375 was downregulated in human epidermal growth factor receptor 2-positive breast cancer and its downregulation induced trastuzumab resistance by targeting insulin like growth factor 1. Furthermore, it was indicated that miR-375 inhibits the epithelial-to-mesenchymal transition in breast cancer by targeting the short stature homeobox 2 gene (31). However, the detailed molecular mechanism of miR-375 in the regulation of breast cancer remains largely unknown. In the present study, it was observed that miR-375 expression was reduced in breast cancer tissues compared with matched adjacent normal tissues. Overexpression of miR-375 significantly suppressed the viability, migration and invasion of breast cancer cells, suggesting that miR-375 acts as a tumor suppressor in breast cancer.

To further reveal the underlying molecular mechanism, the targets of miR-375 in breast cancer cells were investigated in the present study. PAX6 was predicted to be a putative target gene of miR-375. To clarify this prediction, a luciferase reporter assay was completed. This, to the best of our knowledge, was the first time PAX was identified as a direct target of the miR-375 gene. Furthermore, it was demonstrated that miR-375 negatively mediates the expression of PAX6 protein in breast cancer cells.

PAX6 is a member of the PAX gene family and acts as a key regulator in the development of the eyes, central nervous system and pancreas (15,32,33). Furthermore, it has been demonstrated that PAX6 serves an oncogenic role in breast cancer. Knockdown of PAX6 expression markedly suppresses the viability, DNA synthesis and colony formation of breast cancer MCF-7 and MDA-MB-231 cells, and significantly inhibits tumorigenesis in xenograft nude mice (34). Knockdown of PAX6 expression also led to an arrest at the G0/G1 phase in breast cancer cells (34). In the present study, overexpression of PAX6 significantly enhanced MCF-7 cell viability, indicating that PAX6 promotes the cell viability of breast cancer cells. As miR-375 negatively regulated the expression of PAX6 protein in MCF-7 cells, it is suggested that the inhibitory effect of miR-375 on MCF-7 cell viability may occur directly by inhibiting PAX6 expression. However, overexpression of PAX6 did not affect the migration and invasion of MCF-7 cells, suggesting that PAX6 has no effect on breast cancer metastasis. Thus, the suppressive role of miR-375 in the regulation of MCF-7 cell migration and invasion may occur via mediation of other targets. It has been demonstrated that PAX6 mediates cell invasion in different types of cancer, including lung cancer (16), glioma (18), glioblastoma (35,36) and colorectal cancer (19). Therefore, the role of PAX6 in cancer cell metastasis may potentially be tumor-specific.

In conclusion, to the best of our knowledge, the present study is the first to demonstrate that miR-375 is downregulated in breast cancer and negatively mediates the viability, migration and invasion of breast cancer cells. This inhibitory effect of miR-375 on breast cancer cell viability partly occurred via the direct targeting of PAX6. Knowledge of the molecular mechanism of miRs in breast cancer has been expanded and suggests that the miR-375/PAX6 axis may be a promising target for the treatment of breast cancer in the future.

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Wu Y, Sarkissyan M and Vadgama JV: Epithelial-mesenchymal transition and breast cancer. J Clin Med. 5(pii): E132016. View Article : Google Scholar : PubMed/NCBI

4 

Cullen BR: MicroRNAs as mediators of viral evasion of the immune system. Nat Immunol. 14:205–210. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Chen LJ, Lim SH, Yeh YT, Lien SC and Chiu JJ: Roles of microRNAs in atherosclerosis and restenosis. J Biomed Sci. 19:792012. View Article : Google Scholar : PubMed/NCBI

6 

Esau C, Kang X, Peralta E, Hanson E, Marcusson EG, Ravichandran LV, Sun Y, Koo S, Perera RJ, Jain R, et al: MicroRNA-143 regulates adipocyte differentiation. J Biol Chem. 279:52361–52365. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, Ivey KN, Bruneau BG, Stainier DY and Srivastava D: miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 15:272–284. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Yates LA, Norbury CJ and Gilbert RJ: The long and short of microRNA. Cell. 153:516–519. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Serpico D, Molino L and Di Cosimo S: microRNAs in breast cancer development and treatment. Cancer Treat Rev. 40:595–604. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Esquela-Kerscher A and Slack FJ: Oncomirs-microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Luo J, Wu J, Li Z, Qin H, Wang B, Wong TS, Yang W, Fu QL and Lei W: miR-375 suppresses IGF1R expression and contributes to inhibition of cell progression in laryngeal squamous cell carcinoma. Biomed Res Int. 2014:3745982014. View Article : Google Scholar : PubMed/NCBI

12 

Shi ZC, Chu XR, Wu YG, Wu JH, Lu CW, Lü RX, Ding MC and Mao NF: MicroRNA-375 functions as a tumor suppressor in osteosarcoma by targeting PIK3CA. Tumour Biol. 36:8579–8584. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Shen Y, Zhou J, Li Y, Ye F, Wan X, Lu W, Xie X and Cheng X: miR-375 mediated acquired chemo-resistance in cervical cancer by facilitating EMT. PLoS One. 9:e1092992014. View Article : Google Scholar : PubMed/NCBI

14 

Yamaoka T and Itakura M: Development of pancreatic islets (review). Int J Mol Med. 3:247–261. 1999.PubMed/NCBI

15 

Elso C, Lu X, Weisner PA, Thompson HL, Skinner A, Carver E and Stubbs L: A reciprocal translocation dissects roles of Pax6 alternative promoters and upstream regulatory elements in the development of pancreas, brain, and eye. Genesis. 51:630–646. 2013.PubMed/NCBI

16 

Luo J, Li H and Zhang C: MicroRNA-7 inhibits the malignant phenotypes of non-small cell lung cancer in vitro by targeting Pax6. Mol Med Rep. 12:5443–5448. 2015.PubMed/NCBI

17 

Meng B, Wang Y and Li B: Suppression of PAX6 promotes cell proliferation and inhibits apoptosis in human retinoblastoma cells. Int J Mol Med. 34:399–408. 2014.PubMed/NCBI

18 

Cheng Q, Cao H, Chen Z, Ma Z, Wan X, Peng R and Jiang B: PAX6, a novel target of miR-335, inhibits cell proliferation and invasion in glioma cells. Mol Med Rep. 10:399–404. 2014.PubMed/NCBI

19 

Li Y, Li Y, Liu Y, Xie P, Li F and Li G: PAX6, a novel target of microRNA-7, promotes cellular proliferation and invasion in human colorectal cancer cells. Dig Dis Sci. 59:598–606. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Shyr CR, Tsai MY, Yeh S, Kang HY, Chang YC, Wong PL, Huang CC, Huang KE and Chang C: Tumor suppressor PAX6 functions as androgen receptor co-repressor to inhibit prostate cancer growth. Prostate. 70:190–199. 2010.PubMed/NCBI

21 

Meng Y, Zou Q, Liu T, Cai X, Huang Y and Pan J: microRNA-335 inhibits proliferation, cell-cycle progression, colony formation, and invasion via targeting PAX6 in breast cancer cells. Mol Med Rep. 11:379–385. 2015.PubMed/NCBI

22 

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

23 

Hu A, Huang JJ, Xu WH, Jin XJ, Li JP, Tang YJ, Huang XF, Cui HJ, Sun GB, Li RL and Duan JL: MiR-21/miR-375 ratio is an independent prognostic factor in patients with laryngeal squamous cell carcinoma. Am J Cancer Res. 5:1775–1785. 2015.PubMed/NCBI

24 

Jimenez L, Sharma VP, Condeelis J, Harris T, Ow TJ, Prystowsky MB, Childs G and Segall JE: MicroRNA-375 suppresses extracellular matrix degradation and invadopodial activity in head and neck squamous cell carcinoma. Arch Pathol Lab Med. 139:1349–1361. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Costa-Pinheiro P, Ramalho-Carvalho J, Vieira FQ, Torres-Ferreira J, Oliveira J, Gonçalves CS, Costa BM, Henrique R and Jerónimo C: MicroRNA-375 plays a dual role in prostate carcinogenesis. Clin Epigenetics. 7:422015. View Article : Google Scholar : PubMed/NCBI

26 

Wen Y, Han J, Chen J, Dong J, Xia Y, Liu J, Jiang Y, Dai J, Lu J, Jin G, et al: Plasma miRNAs as early biomarkers for detecting hepatocellular carcinoma. Int J Cancer. 137:1679–1690. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Patnaik S, Mallick R, Kannisto E, Sharma R, Bshara W, Yendamuri S and Dhillon SS: MiR-205 and MiR-375 microRNA assays to distinguish squamous cell carcinoma from adenocarcinoma in lung cancer biopsies. J Thorac Oncol. 10:446–453. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Erbes T, Hirschfeld M, Rücker G, Jaeger M, Boas J, Iborra S, Mayer S, Gitsch G and Stickeler E: Feasibility of urinary microRNA detection in breast cancer patients and its potential as an innovative non-invasive biomarker. BMC Cancer. 15:1932015. View Article : Google Scholar : PubMed/NCBI

29 

Wu X, Somlo G, Yu Y, Palomares MR, Li AX, Zhou W, Chow A, Yen Y, Rossi JJ, Gao H, et al: De novo sequencing of circulating miRNAs identifies novel markers predicting clinical outcome of locally advanced breast cancer. J Transl Med. 10:422012. View Article : Google Scholar : PubMed/NCBI

30 

Ye XM, Zhu HY, Bai WD, Wang T, Wang L, Chen Y, Yang AG and Jia LT: Epigenetic silencing of miR-375 induces trastuzumab resistance in HER2-positive breast cancer by targeting IGF1R. BMC Cancer. 14:1342014. View Article : Google Scholar : PubMed/NCBI

31 

Hong S, Noh H, Teng Y, Shao J, Rehmani H, Ding HF, Dong Z, Su SB, Shi H, Kim J and Huang S: SHOX2 is a direct miR-375 target and a novel epithelial-to-mesenchymal transition inducer in breast cancer cells. Neoplasia. 16:279–290.e1-5. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Georgala PA, Carr CB and Price DJ: The role of Pax6 in forebrain development. Dev Neurobiol. 71:690–709. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Hanson IM: PAX6 and congenital eye malformations. Pediatr Res. 54:791–796. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Zong X, Yang H, Yu Y, Zou D, Ling Z, He X and Meng X: Possible role of Pax-6 in promoting breast cancer cell proliferation and tumorigenesis. BMB Rep. 44:595–600. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Huang BS, Luo QZ, Han Y, Li XB, Cao LJ and Wu LX: microRNA-223 promotes the growth and invasion of glioblastoma cells by targeting tumor suppressor PAX6. Oncol Rep. 30:2263–2269. 2013.PubMed/NCBI

36 

Mayes DA, Hu Y, Teng Y, Siegel E, Wu X, Panda K, Tan F, Yung WK and Zhou YH: PAX6 suppresses the invasiveness of glioblastoma cells and the expression of the matrix metalloproteinase-2 gene. Cancer Res. 66:9809–9817. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2017
Volume 14 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zou Q, Yi W, Huang J, Fu F, Chen G and Zhong D: MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells Retraction in /10.3892/etm.2021.10603. Exp Ther Med 14: 1198-1204, 2017
APA
Zou, Q., Yi, W., Huang, J., Fu, F., Chen, G., & Zhong, D. (2017). MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells Retraction in /10.3892/etm.2021.10603. Experimental and Therapeutic Medicine, 14, 1198-1204. https://doi.org/10.3892/etm.2017.4593
MLA
Zou, Q., Yi, W., Huang, J., Fu, F., Chen, G., Zhong, D."MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells Retraction in /10.3892/etm.2021.10603". Experimental and Therapeutic Medicine 14.2 (2017): 1198-1204.
Chicago
Zou, Q., Yi, W., Huang, J., Fu, F., Chen, G., Zhong, D."MicroRNA-375 targets PAX6 and inhibits the viability, migration and invasion of human breast cancer MCF-7 cells Retraction in /10.3892/etm.2021.10603". Experimental and Therapeutic Medicine 14, no. 2 (2017): 1198-1204. https://doi.org/10.3892/etm.2017.4593