Open Access

MicroRNA‑378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma

Retraction in: /10.3892/ol.2023.14196

  • Authors:
    • Peng Qiu
    • Tong‑Jiang Xu
    • Xiang‑Dong Lu
    • Wei Yang
    • Yu‑Bao Zhang
    • Guang‑Ming Xu
  • View Affiliations

  • Published online on: November 16, 2017     https://doi.org/10.3892/ol.2017.7431
  • Pages: 789-794
  • Copyright: © Qiu 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

MicroRNA-378 (miR-378) is dysregulated in multiple malignancies and is associated with tumor progression. However, the expression and mechanism of miR‑378 in pituitary adenoma (PA) remains to be elucidated. In the present study, the role and mechanism of miR‑378 in PA tumorigenesis and development was investigated. It was revealed that the levels of miR‑378 expression were markedly downregulated in PA tissues. CCK‑8 and wound healing assays revealed that transfection with miR‑378 mimics was able to markedly inhibit the proliferation and migration of GH3 cells. Furthermore, quantitative polymerase chain reaction analysis demonstrated that ring finger protein 31 (RNF31) was upregulated in PA specimens and the levels of RNF31 expression was negatively regulated by miR‑378. In addition, knockdown of RNF31 markedly suppressed cell proliferation and migration in GH3 cells. In conclusion, the present study provides a molecular basis for the function of miR‑378/RNF31 in the progression of human PA, indicating a potential novel target for the treatment of PA.

Introduction

Pituitary adenomas (PAs) are the third most common cranial tumors after meningioma and glioma, accounting for 10–15% of all cranial tumors (1). The major challenges of PAs are due to a lack of diagnostic markers and a poor response to available therapies (1,2). A number of genetic syndromes are known to be associated with the development of PAs (3). However, the molecular mechanism of PA tumorigenesis and progression remains poorly understood.

MicroRNAs (miRNAs) are a class of 17–27 nucleotides single-stranded small non-coding RNAs. Accumulating studies have demonstrated that miRNAs can regulate gene expression by binding to target mRNAs at the 3′ untranslated region (3′UTR) region and inhibit gene translation (4,5). MiRNAs, as intrinsic regulators, are known to be involved in various cellular processes, including cell proliferation, differentiation, invasion and apoptosis (6). Accumulating studies revealed that miRNAs function as oncogenes or tumor suppressors. MiRNAs are dysregulated in various types of cancer and are associated with the development and progression of various types of cancer, including breast cancer, colorectal carcinoma, colorectal cancer and ovarian cancer (710). Previously, dysregulated miRNAs were also reported in pituitary tumors (1,11). Trivellin et al (12) demonstrated that miR-107, as a tumor suppressor, is overexpressed in pituitary adenomas. Bottoni et al (13) revealed the downregulation of miR-15a and miR-16-1 in PA tissues. MiR-200c was overexpressed in PA tissues and inhibits cell apoptosis by targeting the phosphatase and tensin homolog/Akt signaling pathway (2). Liang et al (14) observed that miR-31 was downregulated in pituitary adenomas tissues. However, the expression and mechanism of miR-378 in PAs remains to be elucidated.

The atypical E3 ubiquitin ligase RNF31 is a RING finger protein, also termed HOIL-1 interacting protein (15,16), belonging to the RING-between ring-RING (RBR) protein family of E3 ubiquitin ligases. RNF31 was initially cloned from breast cancer cells based on its increased mRNA expression and constitute a valuable diagnostic tool and/or a drug target for ERα-positive breast cancer (17). Previously, several studies demonstrated that RNF31 is involved in tumorigenesis in breast (18) and prostate cancer (19) and adrenocortical carcinoma (20). However, the role of RNF31 in PAs remains unclear.

In the present study, the biological function and molecular mechanisms of miR-378 in regulating proliferation and migration of GH3 cells were investigated, and the underlying mechanism was elucidated, indicating miR-378 as a potential novel diagnostic or therapeutic target for PA.

Materials and methods

Tissue samples

A total of 25 tumor and adjacent non-tumor tissues samples were collected from patients (39–65 years old, with a mean age of 51 years old, including 11 men and 14 women) who had undergone surgical treatment in Shandong Provincial Hospital Affiliated to Shandong University (Shandong, China) between January 2013 and May 2015. None of the patients had received chemotherapy or radiotherapy prior to surgery. The present study was approved by the Ethics Committee of Shandong University, and written informed consent was obtained from each patient. Tissue fragments were immediately frozen in liquid nitrogen under ribonuclease-free conditions at the time of surgery and stored at −80°C.

Cell culture and transfection

The rat pituitary GH3 cell line was purchased from the Cell Resource Center of Shanghai Institutes for Biological Sciences (Shanghai, China) and cultured in Dulbecco's modified Eagle's medium (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) supplemented with 10% fetal bovine serum (Biosera Ltd., Ringmer, UK), penicillin (100 IU/ml) and streptomycin (100 mg/ml) (Sigma-Aldrich; Merck KGaA) in a humidified atmosphere at 37°C with 5% CO2.

GH3 cells (1×106) were seeded on 6-well plates and then transfected with 50 nM miR-378 mimics or inhibitors or mimics-control or inhibitor-control (GenePharma, Co., Ltd., Shanghai, China) using Lipofectamine® RNAiMAX (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) as previously described (21).

Cell proliferation analysis

GH3 cells (3×105) were seeded on 96-well microplates at 24 h post-transfection. Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan) was then used to detect cell proliferative ability at 0, 24, 48 and 72 h according to the manufacturer's instructions.

Cell migration assay

The wound healing assay was used to detect cell migratory capability according to a previous study (22).

Western blot analysis

Total protein was collected using cell lysis reagent (Sigma-Aldrich; Merck KGaA) and then 20 µg protein was loaded per lane and separated by 10% SDS-PAGE and transferred to polyvinylidene fluoride (PVDF) membranes (BD Bioscience, Franklin Lakes, NJ, USA). Subsequent to blocking with 5% bovine serum albumin (Gibco; Thermo Fisher Scientific, Inc.) at room temperature for 1 h, PVDF membranes were incubated with the primary antibodies anti-GAPDH (1:1,000; catalog no. ab8425) and anti-RNF31 (1:1,000; catalog no. ab46322) from Abcam (Cambridge, UK) at 4°C overnight. Subsequently, the membranes were incubated with horseradish peroxidase goat anti-rabbit secondary antibody (catalog no. BS10000; 1:2,500; Bioworld Technology, Inc., St. Louis Park, MN, USA) at room temperature for 1 h. The protein signals were detected by ECL detection systems (SuperSignal West Femto; Pierce; Thermo Fisher Scientific, Inc.).

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

RNA was extracted using TRIzol solution (Sigma-Aldrich; Merck KGaA), and the first strand of cDNA was synthesized from 1 µg RNA by RT using the PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd., Dalian, China) and the RT Primer mix was added at 37°C for 15 min, followed by 85°C for 5 sec. The expression of miRNA was detected by RT-qPCR analysis using the SYBR-Green detection system (Roche Applied Science, Penzberg, Germany) (18). Primer sequences for qPCR are as follows: miR-378 forward, 5′-AGGCAAGATGCTGGCATAGCT-3′. U6 was used as a control to normalize the expression level of miRNAs. The forward primer sequence of U6 is as follows: 5′-GCGAGCACAGAATTAATACGAC-3′. The sequences of additional primers used are as follows: RNF31 forward, 5′-ACCCCCTATTGAGAGAGATTGCT-3′ and reverse, 5′-TGGAGCCTGGGACAGAGG-3′; and GAPDH forward, 5′-TGTGGGCATCAATGGATTTGG-3′ and reverse, 5′-ACACCATGTATTCCGGGTCAAT-3′. GAPDH was used as a control to normalize the expression level of mRNA.

Dual luciferase reporter assay

MiRNA target prediction website (www.microRNA.org) was used to predict the target gene of miR-378. The RNF31-3′ untranslated region (3′UTR) containing the putative miR-378 target sites was amplified by PCR from the genomic DNA of a healthy control donor and cloned into the pGL3 Basic dual Luciferase reporter plasmid as described previously (23). The cells were co-transfected with either pGL3-WT-RNF31-3′-UTR (wild-type) or pGL3-Mut-RNF31-3′-UTR (mutant) vector (0.8 µg) and the miR-378 mimic (50 nM) with Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) and then detected luciferase activities by Dual-Luciferase Reporter Assay system (Promega Corporation, Madison, WI, USA) after 48 h.

Statistical analysis

Data are presented as the mean ± standard deviation. The differences were analyzed using Student's t-test or one-way analysis of variance followed by Tukey multiple comparison post-hoc analysis. All the analyses were performed using SPSS software (version 17.0; SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

miR-378 expression in prostate cancer

The levels of miR-378 expression were first analyzed in human PA tissues and adjacent normal tissues using qPCR assay (Fig. 1). Consistent with a previous study (14), the results revealed that miR-378 expression was significantly downregulated in PA tissues compared with the adjacent normal tissues (P<0.01). The results indicated that downregulated miR-378 may be involved in the tumorigenesis of PA.

Effect of miR-378 on proliferation of GH3 cells

To understand the role of miRNA downregulation in pituitary tumorigenesis, the effects of miR-378 on cell proliferation were analyzed. Rat pituitary GH3 cells were transfected with miR-378 mimics and miR-378 inhibitors. The proliferative ability of GH3 cells was detected by Cell Counting Kit-8. As presented in Fig. 2, overexpression of miR-378 was able to significantly inhibit the proliferative ability of GH3 cells compared with the control.

miR-378 inhibits migration of GH3 cells

The effects of miR-378 on cell migration were subsequently detected in GH3 cells. As presented in Fig. 3, transfection with miR-378 mimics was able to repress migration of GH3 cells, indicating that miR-378 may contribute to the inhibition of PA metastasis.

RNF31 is overexpressed in PA tissues and directly regulated by miR-378

In the present study, the expression levels of RNF31 in human PA tissues and adjacent normal tissues were analyzed by qPCR. As presented in Fig. 4A, the levels of RNF31 expression were significantly increased in PA tissues compared with the adjacent normal tissues (P<0.01), indicating that the upregulation of RNF31 may be involved in human PA carcinogenesis.

miRNA target prediction websites www.microRNA.org were used, and it was revealed that miR-378 was able to bind to the 3′UTR of RNF31 mRNA (Fig. 4B). To confirm that RNF31 is a target of miR-378, a dual-luciferase reporter assay was used. As presented in Fig. 4B, transfection with miR-378 mimic was able to significantly inhibit luciferase activity of the wild-type RNF31 3′UTR, whereas the luciferase activity of mutated target sites were unaffected, indicating that RNF31 is a direct target gene of miR-378.

The effect of miR-378 transfection on endogenous RNF31 mRNA and protein expression was subsequently evaluated in GH3 cells by western blot analysis and RT-qPCR. As presented in Fig. 4C and D, the expression of RNF31 mRNA and protein was markedly downregulated by miR-378 mimics compared with the control. Collectively, the present results demonstrated that miR-378 was able to downregulate the expression of RNF31 in GH3 cells.

Knockdown of RNF31 mimics miR-378 inhibition

To investigate the role of RNF31 in miR-378-mediated tumor suppression, siRNAs-RNF3 was used to silence RNF3 expression in GH3 cells. Western blot analysis revealed that transfection with siRNAs-RNF31 was able to downregulate the expression of RNF31 at the protein level (Fig. 5A). In addition, transfection with siRNA-RNF31 was able to significantly reduce the proliferative and migratory abilities of the cells (Fig. 5B and C), which was similar to the effects of the miR-378 mimics.

Discussion

Dysregulated miRNAs are often involved in the tumorigenesis and progression of various types of cancer. However, their potential roles in PAs remain to be elucidated. In the present study, the expression of miR-378 was analyzed in PA. It was revealed that miR-378 expression was downregulated in PA tissues compared with adjacent normal tissues. The effect of miR-378 on the downregulation of proliferation and migration of GH3 cells was also identified. In addition, RNF31 was identified as a direct target of miR-378 and is involved in miR-378-mediated proliferation and migration of GH3 cells.

MiR-378, as a tumor suppressor or oncogene, has been reported to be potentially important in the progression of cancer. Previous studies have confirmed that miR-378 was downregulated in patients with prostate cancer, and the downregulation of miR-378 was associated with higher Gleason score, tumors with a larger diameter (P=0.034) and elevated serum prostate-specific antigen levels (24). It has also been reported that miR-378 is able to suppress cell proliferation through downregulation of mitogen-activated protein kinase 1 (MAPK1) in prostate cancer (25,26). MiR-378 was also reported to inhibit progression of human gastric cancer by targeting MAPK1 (27). In addition, miR-378 was reported as an oncogene and promotes the migration of liver cancer cells (28). Taken together, these findings indicate that the role of miR-378 in human malignancies may be multifaceted, depending on the specific tissues involved. In the present study, it was revealed that miR-378 expression was downregulated in PA tissues and transfection with miR-378 mimics was able to suppress cell proliferation and migration in GH3 cells, indicating the potential role of miR-378 as a tumor suppressor in PA.

RNF31 (also known as HOIP and ZIBRA), which belongs to the RBR protein family of E3 ubiquitin ligases, was initially cloned from breast cancer cells based on its elevated mRNA expression and shown to exhibit higher expression in breast tumors compared with adjacent tissues (17). RNF31 and other RING finger protein family members have been demonstrated to be able to modify the levels of p53 protein and p53 signaling (18,29,30). RNF31 was also reported to stabilize estrogen receptor-α and modulate estrogen-stimulated breast cancer cell proliferation (31). miR-31 reduces growth of papillary thyroid carcinoma cells by targeting RNA-binding protein human antigen R (32). Ehrlund et al (20) demonstrated that RNF31 was associated with cholesterol metabolism and steroid hormone synthesis, strengthening its function as a co-regulator of splicing factor 1. In the present study, it was demonstrated that RNF31 expression was regulated by miR-378 in PA, using the luciferase reporter assay. Transfection with siRNA-RNF31 in GH3 cells was able to reduce proliferation and migration compared with the control, similar to the phenotype exhibited upon restoration of miR-378 expression.

In conclusion, a regulatory mechanism of miR-378 in proliferation and migration of GH3 cells and a downstream target, RNF31, was identified, providing an insight into the role of miR-378 in carcinogenesis and metastasis of PA. These findings may provide possible therapeutic targets for the treatment of metastatic PA.

Acknowledgements

The present study was funded by the Natural Science Foundation of Shandong Province (grant no. ZR2015HM015).

References

1 

Shi X, Tao B, He H, Sun Q, Fan C, Bian L, Zhao W and Lu YC: MicroRNAs-based network: A novel therapeutic agent in pituitary adenoma. Med Hypotheses. 78:380–384. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Liao C, Chen W, Fan X, Jiang X, Qiu L, Chen C, Zhu Y and Wang H: MicroRNA-200c inhibits apoptosis in pituitary adenoma cells by targeting the PTEN/Akt signaling pathway. Oncol Res. 21:129–136. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Sukumari-Ramesh S, Singh N, Jensen MA, Dhandapani KM and Vender JR: Anacardic acid induces caspase-independent apoptosis and radiosensitizes pituitary adenoma cells. J Neurosurg. 114:1681–1690. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Srivastava N, Manvati S, Srivastava A, Pal R, Kalaiarasan P, Chattopadhyay S, Gochhait S, Dua R and Bamezai RN: miR-24-2 controls H2AFX expression regardless of gene copy number alteration and induces apoptosis by targeting antiapoptotic gene BCL-2: A potential for therapeutic intervention. Breast Cancer Res. 13:R392011. View Article : Google Scholar : PubMed/NCBI

5 

Liu X, Wang A, Heidbreder CE, Jiang L, Yu J, Kolokythas A, Huang L, Dai Y and Zhou X: MicroRNA-24 targeting RNA-binding protein DND1 in tongue squamous cell carcinoma. FEBS Lett. 584:4115–4120. 2010. View Article : Google Scholar : PubMed/NCBI

6 

Xu XM, Qian JC, Deng ZL, Cai Z, Tang T, Wang P, Zhang KH and Cai JP: Expression of miR-21, miR-31, miR-96 and miR-135b is correlated with the clinical parameters of colorectal cancer. Oncol Lett. 4:339–345. 2012.PubMed/NCBI

7 

Chen S, Dai Y, Zhang X, Jin D, Li X and Zhang Y: Increased miR-449a expression in colorectal carcinoma tissues is inversely correlated with serum carcinoembryonic antigen. Oncol Lett. 7:568–572. 2014.PubMed/NCBI

8 

Lu Z, Ye Y, Jiao D, Qiao J, Cui S and Liu Z: miR-155 and miR-31 are differentially expressed in breast cancer patients and are correlated with the estrogen receptor and progesterone receptor status. Oncol Lett. 4:1027–1032. 2012.PubMed/NCBI

9 

Lei SL, Zhao H, Yao HL, Chen Y, Lei ZD, Liu KJ and Yang Q: Regulatory roles of microRNA-708 and microRNA-31 in proliferation, apoptosis and invasion of colorectal cancer cells. Oncol Lett. 8:1768–1774. 2014.PubMed/NCBI

10 

Luo J, Zhou J, Cheng Q, Zhou C and Ding Z: Role of microRNA-133a in epithelial ovarian cancer pathogenesis and progression. Oncol Lett. 7:1043–1048. 2014.PubMed/NCBI

11 

D'Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, Fedele M, Croce CM, Trouillas J and Fusco A: Altered microRNA expression profile in human pituitary GH adenomas: Down-regulation of miRNA targeting HMGA1, HMGA2 and E2F1. J Clin Endocrinol Metab. 97:E1128–E1138. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Trivellin G, Butz H, Delhove J, Igreja S, Chahal HS, Zivkovic V, McKay T, Patócs A, Grossman AB and Korbonits M: MicroRNA miR-107 is overexpressed in pituitary adenomas and inhibits the expression of aryl hydrocarbon receptor-interacting protein in vitro. Am J Physiol Endocrinol Metab. 303:E708–E719. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Bottoni A, Piccin D, Tagliati F, Luchin A, Zatelli MC and degli Uberti EC: miR-15a and miR-16-1 down-regulation in pituitary adenomas. J Cell Physiol. 204:280–285. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Liang S, Chen L, Huang H and Zhi D: The experimental study of miRNA in pituitary adenomas. Turk Neurosurg. 23:721–727. 2013.PubMed/NCBI

15 

Kirisako T, Kamei K, Murata S, Kato M, Fukumoto H, Kanie M, Sano S, Tokunaga F, Tanaka K and Iwai K: A ubiquitin ligase complex assembles linear polyubiquitin chains. EMBO J. 25:4877–4887. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Walczak H, Iwai K and Dikic I: Generation and physiological roles of linear ubiquitin chains. BMC Biol. 10:232012. View Article : Google Scholar : PubMed/NCBI

17 

Thompson HG, Harris JW, Lin L and Brody JP: Identification of the protein Zibra, its genomic organization, regulation and expression in breast cancer cells. Exp Cell Res. 295:448–459. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Zhu J, Zhao C, Zhuang T, Jonsson P, Sinha I, Williams C, Strömblad S and Dahlman-Wright K: RING finger protein 31 promotes p53 degradation in breast cancer cells. Oncogene. 35:1955–1964. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Yao WJ, Wang YL, Lu JG, Guo L, Qi B and Chen ZJ: MicroRNA-506 inhibits esophageal cancer cell proliferation via targeting CREB1. Int J Clin Exp Pathol. 8:10868–10874. 2015.PubMed/NCBI

20 

Ehrlund A, Jonsson P, Vedin LL, Williams C, Gustafsson JA and Treuter E: Knockdown of SF-1 and RNF31 affects components of steroidogenesis, TGFβ and Wnt/β-catenin signaling in adrenocortical carcinoma cells. PLoS One. 7:e320802012. View Article : Google Scholar : PubMed/NCBI

21 

Ehrlund A, Anthonisen EH, Gustafsson N, Venteclef N, Robertson Remen K, Damdimopoulos AE, Galeeva A, Pelto-Huikko M, Lalli E, Steffensen KR, et al: E3 ubiquitin ligase RNF31 cooperates with DAX-1 in transcriptional repression of steroidogenesis. Mol Cell Biol. 29:2230–2242. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Liang CC, Park AY and Guan JL: In vitro scratch assay: A convenient and inexpensive method for analysis of cell migration in vitro. Nat Protoc. 2:329–333. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Micale L, Fusco C, Fontana A, Barbano R, Augello B, De Nittis P, Copetti M, Pellico MT, Mandriani B, Cocciadiferro D, et al: TRIM8 downregulation in glioma affects cell proliferation and it is associated with patients survival. BMC Cancer. 15:4702015. View Article : Google Scholar : PubMed/NCBI

24 

Avgeris M, Stravodimos K and Scorilas A: Loss of miR-378 in prostate cancer, a common regulator of KLK2 and KLK4, correlates with aggressive disease phenotype and predicts the short-term relapse of the patients. Biol Chem. 395:1095–1104. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Kulyté A, Lorente-Cebrián S, Gao H, Mejhert N, Agustsson T, Arner P, Rydén M and Dahlman I: MicroRNA profiling links miR-378 to enhanced adipocyte lipolysis in human cancer cachexia. Am J Physiol Endocrinol Metab. 306:E267–E274. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Chen QG, Zhou W, Han T, Du SQ, Li ZH, Zhang Z, Shan GY and Kong CZ: MiR-378 suppresses prostate cancer cell growth through downregulation of MAPK1 in vitro and in vivo. Tumour Biol. 37:2095–2103. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Fei B and Wu H: MiR-378 inhibits progression of human gastric cancer MGC-803 cells by targeting MAPK1 in vitro. Oncol Res. 20:557–564. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Ma J, Lin J, Qian J, Qian W, Yin J, Yang B, Tang Q, Chen X, Wen X, Guo H and Deng Z: MiR-378 promotes the migration of liver cancer cells by down-regulating Fus expression. Cell Physiol Biochem. 34:2266–2274. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Wen W, Peng C, Kim MO, Ho Jeong C, Zhu F, Yao K, Zykova T, Ma W, Carper A, Langfald A, et al: Knockdown of RNF2 induces apoptosis by regulating MDM2 and p53 stability. Oncogene. 33:421–428. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Nie J, Xie P, Liu L, Xing G, Chang Z, Yin Y, Tian C, He F and Zhang L: Smad ubiquitylation regulatory factor 1/2 (Smurf1/2) promotes p53 degradation by stabilizing the E3 ligase MDM2. J Biol Chem. 285:22818–22830. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Zhu J, Zhao C, Kharman-Biz A, Zhuang T, Jonsson P, Liang N, Williams C, Lin CY, Qiao Y, Zendehdel K, et al: The atypical ubiquitin ligase RNF31 stabilizes estrogen receptor alpha and modulates estrogen-stimulated breast cancer cell proliferation. Oncogene. 33:4340–4351. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Wu D, Wang B, Shang J, Song J and Zhang H: miR-31 reduces cell growth of papillary thyroid carcinoma by RNA-binding protein HuR. Clin Lab. 61:1625–1634. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 15 Issue 1

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
Qiu P, Xu TJ, Lu XD, Yang W, Zhang YB and Xu GM: MicroRNA‑378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma Retraction in /10.3892/ol.2023.14196. Oncol Lett 15: 789-794, 2018
APA
Qiu, P., Xu, T., Lu, X., Yang, W., Zhang, Y., & Xu, G. (2018). MicroRNA‑378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma Retraction in /10.3892/ol.2023.14196. Oncology Letters, 15, 789-794. https://doi.org/10.3892/ol.2017.7431
MLA
Qiu, P., Xu, T., Lu, X., Yang, W., Zhang, Y., Xu, G."MicroRNA‑378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma Retraction in /10.3892/ol.2023.14196". Oncology Letters 15.1 (2018): 789-794.
Chicago
Qiu, P., Xu, T., Lu, X., Yang, W., Zhang, Y., Xu, G."MicroRNA‑378 regulates cell proliferation and migration by repressing RNF31 in pituitary adenoma Retraction in /10.3892/ol.2023.14196". Oncology Letters 15, no. 1 (2018): 789-794. https://doi.org/10.3892/ol.2017.7431