Open Access

Downregulation of BRAF‑activated non‑coding RNA suppresses the proliferation, migration and invasion, and induces apoptosis of hepatocellular carcinoma cells

  • Authors:
    • Jing Li
    • Jinfeng Wang
    • Wei Zhou
    • Shuqin Zhang
    • Yuan Le
    • Rong He
  • View Affiliations

  • Published online on: August 18, 2017     https://doi.org/10.3892/ol.2017.6770
  • Pages: 4751-4757
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

The long non‑coding RNA BRAF‑activated non‑coding RNA (BANCR) has been reported to serve essential roles in the progress of a various cancer types. The purpose of the present study was to investigate the effect of lncRNA BANCR in HCC cells. The expression of BANCR in the HCC cell line Huh7 and a normal liver cell line were determined using reverse transcription‑quantitative polymerase chain reaction. The expression of BANCR in Huh7 cells was downregulated by short hairpin (sh)RNA. Subsequently, the proliferation, apoptosis, migration and invasion rates were determined, along with the activity of MAPK/ERK kinase (MEK) and mitogen‑activated protein kinase signaling pathways. The results revealed that BANCR was overexpressed in Huh7 cells when compared with normal liver cells. The downregulation of BANCR significantly inhibited the proliferation and colony formation ability, and induced cell cycle arrest and apoptosis of Huh7 cells. The migration and invasion of Huh7 cells were also suppressed in BANCR shRNA‑transfected cells. The downregulation of BANCR significantly inhibited the activity of MEK, extracellular signal‑regulated kinase and janus kinase signaling pathways. Collectively, these findings demonstrated that the lncRNA BANCR is oncogenic in Huh7 cells, and may be a promising molecular target for HCC therapy.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common types of malignancy and is the third leading cause of cancer-associated mortality worldwide (1). There are ~500,000 cases diagnosed as HCC each year, which represents >5% of all cancer cases (2). The incidence of HCC has a considerable geographic variation, with the majority of the cases occurring in developing countries, including Southeast Asia and sub-Saharan Africa (3). However, in recent decades, epidemiological studies have also indicated a rising trend in the incidence and mortality of HCC in Western countries (4). Despite substantial and accelerated studies focused on the treatment of HCC, the five-year survival rate of advanced HCC remains poor (57). Thus, the development of novel and efficient therapy strategies for HCC are warranted.

Long non-coding RNA (lncRNA) is a group of noncoding RNAs that are >200 nucleotides in length, and serve regulatory roles in different physiological processes, including growth, differentiation, senescence and apoptosis (8). Recently, increasing evidence suggest that the dysregulation of lncRNAs are involved in diverse pathological conditions, in particular in various types of cancer (911). BRAF-activated non-coding RNA (BANCR), a 693-bp lncRNA on chromosome 9, was first identified to be overexpressed in melanoma cells and serve as a regulator in the migration of melanoma cells (12,13). Reportedly, BANCR is abnormally expressed in gastric tumor (14), papillary thyroid carcinoma (15), colorectal cancer (16), retinoblastoma (17), papillary thyroid carcinoma (18) and non-small cell lung cancer (19). Despite the majority of these studies indicating an oncogenic property of BANCR, Sun et al (19) and Shi et al (16) reported tumor suppressive activity of BANCR in non-small cell lung cancer, and colorectal carcinoma, respectively.

In the present study, the expression of BANCR in Huh7 cells was downregulated using short hairpin (sh)RNA, and the effect of BANCR on proliferation, apoptosis, migration and invasion of HCC cells was investigated in vitro. Furthermore, the underlying mechanisms of BANCR in HCC were explored.

Materials and methods

Cell lines, culture condition and treatment

The human hepatocellular carcinoma cell line Huh7 was purchased from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). Human normal liver L-02 cells were purchased from Zhongqiaoxinzhou Biotechnology Co., Ltd (Shanghai, China). Cells were maintained in Dulbecco's modified Eagle's medium or RPMI-1640 (both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and supplemented with 10% fetal bovine serum (Hyclone; GE Healthcare Life Sciences, Logan, UT, USA) in a humidified atmosphere of 5% CO2 at 37°C. Cells was transfected with BANCR shRNA or negative control (NC) shRNA using Lipofectamine™ 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The target sequences were as follows: BANCR shRNA, 5′-GGAGTGGCGACTATAGCAA-3′ and NC shRNA, 5′-TTCTCCGAACGTGTCACGT-3′.

Reverse transcription-quantitative polymerase chain reaction (PCR)

Total RNA was extracted using the RNApure total RNA extraction kit and reverse transcribed to cDNA using Super M-MLV reverse transcriptase at 25°C for 10 min followed by 42°C for 50 min. (both from Bioteke Corporation, Beijing, China). SYBR Green-based PCR was performed using an Exicycler™ 96 real-time PCR system (Bioneer Corporation, Daejeon, Korea) with 2X Power Taq PCR Master mix (Bioteke Corporation), the thermocycling conditions was as follows: 95°C for 10 min followed by 40 cycles of 95°C for 10 sec, 60°C for 20 sec and 72°C for 30 sec. The specific primers for BANCR were as follows: Forward, 5′-TCAGAAGAAACAAGAGGGAGG-3′ and reverse, 5′-AGCAGCATGAACTGGGAAAC-3′. The specific primers of β-actin were as follows: Forward, 5′-CTTAGTTGCGTTACACCCTTTCTTG-3′ and reverse, 5′-CTGTCACCTTCACCGTTCCAGTTT-3′. Each experiment was repeated for three times. The relative BANCR mRNA level was calculated using the 2−ΔΔCq method (20) and normalized to β-actin expression.

Western blot analysis

Protein from each group was extracted using the Whole Cell Lysis kit (Wanleibio, Shenyang, China). A total of 40 µg protein per lane were electrophoresed using 8, 10 or 13% SDS-PAGE and subsequently electrotransfered to polyvinylidene fluoride membranes (EMD Millipore, Billerica, MA, USA). After blocking in 5% non-fat milk at room temperature for 1 h, the membranes were incubated at 4°C overnight with the following primary antibodies: Bcl-2 (1:400 dilution, cat. no. BA0412), Bax (1:400 dilution, cat. no. BA0315) (both from Wuhan Boster Biological Technology, Ltd., Wuhan, China), cleaved caspase-3 (1:1,000 dilution; cat. no. ab2302, Abcam, Cambridge, MA, USA), phosphorylated (p)-extracellular signal-regulated kinase (p-ERK; 1:500 dilution; cat. no. bs-1522R), ERK (1:500 dilution; cat. no. bs-2637R), p-janus kinase (p-JNK; 1:500 dilution; cat. no. bs-1640R), JNK (1:500 dilution; cat. no. bs-10562R), p-P38 (1:500 dilution; cat. no. bs-5477R), P38 (1:500 dilution; cat. no. bs-0637R) (all from BIOSS, Beijing, China), p-MAPK/ERK kinase (p-MEK; 1:200 dilution; cat. no. sc-271914), MEK (1:200 dilution; cat. no. sc-219), and β-actin (1:1,000 dilution; cat. no. sc-47778) (all from Santa Cruz Biotechnology, Inc., Dallas, TX, USA). Subsequently, the membranes were incubated with horseradish peroxidase-conjugated goat anti rabbit (cat. no. WLA023) or goat anti mouse (cat. no. WLA024) secondary antibodies (Wanleibio) at 37°C for 45 min. The specific bands were visualized with the Immu-Plus ECL system (Wanleibio), and the protein levels were semi-quantified using densitometry analysis that performed by Gel-Pro Analyzer Version 3.0 (Media Cybernetics, Silver Spring, MD, USA). β-actin served as an internal control.

Cell viability assay

Cells were seeded in a 96-well plate at a density of 3×103 cells/well and cultivated at 37°C for 24 h. Subsequently, cells were transfected with BANCR shRNA or NC shRNA, and further incubated for 24, 48, 72 or 96 h. Cells were then incubated with 0.5 mg/ml MTT for 4 h, followed by the addition of 200 µl dimethyl sulfoxide. Lastly, the absorbance at 490 nm was measured using the ELX-800 microplate spectrophotometer (Biotek Instruments, Inc., Winooski, VT, USA). For colony formation, cells were seeded in a 35-mm culture dish (300 cells/dish), and maintained in culture media supplemented with 10% fetal bovine serum for two weeks. The cell clones were fixed with 4% paraformaldehyde at room temperature for 20 min, then stained with Wright-Giemsa dye at room temperature for 5 min. Colony formation rate was calculated as follows: (colony number/seeding number) ×100%.

Flow cytometry

Cells were collected 48 h post-transfection for cell cycle analysis, cells were incubated with 25 µl propidium iodide (Beyotime Institute of Biotechnology, Haimen, China) for 30 min at 37°C in the dark. For apoptosis detection, cells were incubated with 5 µl Annexin V-FITC and 5 µl propidium iodide following the manufacturer's protocol (Nanjing KeyGen Biotech Co., Ltd., Nanjing, China). Following staining, the cell cycle or apoptosis status was analyzed using flow cytometry (BD Accuri C6; BD Biosciences, Franklin Lakes, NJ, USA).

Hoechst staining

Cells were seeded on 12-well plates at a density of 5×104 cells/well, transfected with BANCR shRNA or NC shRNA. A total of 48 h after transfection, cells were fixed and stained with 2 µg/ml Hoeschst staining solution (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. Apoptotic nuclei were observed under a fluorescent microscope (BX53; Olympus Corporation, Tokyo, Japan).

Wound healing assay

Cells were allowed to grow until 80–90% confluence was achieved on 6-well plates, then transfected with BANCR shRNA or NC shRNA. A total of 48 h after transfection, cells were incubated with 1 µg/ml mitomycin C (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) for 1 h, and a wound was created with a 200-µl pipette tip. Subsequently, cells were cultured in a humidified atmosphere of 5% CO2 at 37°C for 24 h. The migration distance was measured under an inverted phase contrast microscope (AE31; Motic Instruments, Richmond, BC, Canada).

Transwell assays

Transwell chambers (Corning Incorporated, Corning, NY, USA) were pre-coated with 40 µl Matrigel (BD Biosciences) and placed on to 24-well plates. Cells from each group were seeded in the top chamber, 2×104 cells/well. A total of 800 µl cell culture medium supplemented with 20% fetal bovine serum was added to the lower chamber as a chemoattractant. The non-invading cells on the upper-side of the membrane were removed with cotton swabs 24 after incubation. The invasive cells were fixed with 4% paraformaldehyde at room temperature for 20 min and stained with 0.5% crystal violet solution at room temperature for 5 min. The number of invasive cells was counted under inverted phase-contrast microscope in a blinded manner.

Statistical analysis

The cell viability assay was repeated five times; all other experiments were repeated three times. All values are expressed as the mean ± standard deviation. Differences between groups were analyzed using one-way analysis of variance followed by Bonferroni's multiple comarisons test with SPSS 16.0 software (SPSS, Inc., Chicago, IL, USA). P<0.05 were considered to indicate a statistically significant difference.

Results

BANCR is overexpressed in HCC cells

To assess the biological role of BANCR in the progression of HCC, the expression level of BANCR in Huh7 HCC cells and normal liver cells was measured by using RT-PCR. As presented in Fig. 1A, BANCR was significantly overexpressed in the human HCC cell line Huh7 when compared with normal liver cells (P<0.001). Thus, Huh7 cells were transfected with BANCR shRNA or NC shRNA, the expression level of BANCR in transfected cells was also determined (Fig. 1B). The results revealed that transfection with BANCR shRNA resulted in a significant decrease (72.59±1.59%) in BANCR levels compared with NC shRNA-transfected Huh7 cells (P<0.001), suggesting that the expression of BANCR was significantly inhibited by BANCR shRNA.

Downregulation of BANCR suppresses the viability of HCC cells

To investigate whether BANCR was functionally involved in HCC cell viability, the proliferation of HCC cells was detected using an MTT assay. The results demonstrated that the proliferation of Huh7 cells transfected with BANCR shRNA were significantly decreased compared with NC shRNA-transfected cells (Fig. 2A; P<0.01). Simultaneously, the clonogenicity tests revealed that downregulation of BANCR resulted in a significantly decreased colony formation rate compared with the NC shRNA group (Fig. 2B; P<0.001). Furthermore, cell cycle analysis was performed to assess the role of BANCR in the regulation of cell proliferation, as presented in Fig. 2C, the G0/G1 phase cell proportion was increased significantly in shRNA BANCR-transfected cells compared with NC shRNA-transfected cells (P<0.05). Thus, the downregulation of BANCR may inhibit HCC cell proliferation by induction of cell cycle arrest.

Downregulation of BANCR induces the apoptosis of HCC cells

The apoptosis status of BANCR shRNA-transfected cells were determined using flow cytometry (Fig. 3A), it was demonstrated that the apoptosis cell proportion in Huh7 cells was significantly higher in BANCR shRNA-transfected cells compared with NC shRNA-transfected cells (P<0.001), and this was also confirmed by Hoechst staining. As presented in Fig. 3B, typical apoptotic bodies were notably observed in BANCR shRNA-transfected cells. In addition, the expression of apoptosis-associated proteins was measured (Fig. 3C). As expected, when compared with the NC group, downregulation of BANCR significantly inhibited the expression of anti-apoptotic protein Bcl-2, and this was accompanied by an increase in proapoptotic protein Bax and cleaved caspase-3. Taken together, downregulation of BANCR promoted apoptosis of HCC cells.

Downregulation of BANCR inhibits the migration and invasion of HCC cells

To clarify the effect of BANCR on migration and invasion of HCC cells, wound healing and Transwell assays were performed. As presented in Fig. 4A and B, when compared with the NC group, the migration rate, and invasive cells in the BANCR shRNA-transfected group were significantly decreased (P<0.05), indicating that downregulation of BANCR can inhibit the migration and invasion of HCC cells.

Downregulation of BANCR inactivates MEK, ERK and JNK, but does not affect the activity of P38 MAPK signaling pathway

The activation of MAPK and MEK pathway were examined using western blot analysis, to explore the potential underlying mechanisms of BANCR in the progress of HCC. It was revealed that the MEK, ERK and JNK signaling pathways were inactivated significantly in BANCR shRNA-transfected cells when compared with NC shRNA-transfected cells (Fig. 5A-C). However, the activity of the P38 pathway was not affected by the downregulation of BANCR (Fig. 5D). Thus, these results suggest that the MEK, ERK and JNK signaling pathways were involved in the BANC-associated malignance of HCC cells.

Discussion

LncRNAs are newly recognized RNAs, which may serve important roles in the development of cancer. Previous studies have reported that the lncRNA BANCR is abnormally expressed in various malignant tumors and participates in tumor development (1419). In the present study, it was demonstrated that the lncRNA BANCR was overexpressed in the HCC cell line Huh7 when compared with the normal liver cell line L-02. The downregulation of BANCR by shRNA significantly suppressed the proliferative capacity, clonogenicity and induced apoptosis of Huh7 cells. The migratory and invasive ability of Huh7 cells were also inhibited by BANCR shRNA. Furthermore, these tumor inhibitory effects appear to be associated with the repression of MEK, ERK and JNK signaling pathways. These findings demonstrated an essential role for BANCR in the regulation of proliferation, apoptosis, migration and invasion in HCC cells.

BANCR has been identified to be significantly upregulated in human malignant melanoma (21), papillary thyroid carcinoma (18), gastric tumor (14,22) and retinoblastoma tissues (23). Previous studies revealed that the knockdown of BANCR resulted in cell growth inhibition and cell cycle arrest in human thyroid cancer cells (15), and human melanoma cells (21). The aforementioned studies described a tumor promotional role of BANCR. However, other studies have reported that BANCR was expressed at a lower level in various cancer types, and its expression was inversely correlated with tumor malignancy, meanwhile, in vitro experiments on colorectal cancer, non-small cell lung cancer and lung carcinomas cells suggested that BANCR serves as an cancer suppressor gene (16,19,24). Thereby, BANCR serves distinct roles in different tumor types, which may be attributed to tumor heterogenicity. The present study demonstrated that BANCR was significantly overexpressed in Huh7 HCC cells when compared with normal liver cells, and the downregulation of BANCR significantly inhibited proliferation, colony formation and induced cell cycle arrest in Huh7 cells, suggesting a tumor promoter property of BANCR in HCC cells.

It was also demonstrated that the repression of BANCR significantly induced apoptosis of Huh7 cells. Given the important roles of apoptosis-associated proteins in the cell apoptosis signaling pathway (25,26), the expression of Bcl-2, Bax and cleaved caspase-3 was determined using western blotting. The results revealed that transfection with BANCR shRNA significantly inhibited the expression of Bcl-2, increased the level of Bax and cleaved caspase-3, further elucidating the role of BANCR in the regulation of apoptosis.

Flockhart et al (12) reported that BANCR was recurrently overexpressed in melanomas tissues, and knockdown of BANCR in melanoma cells significantly reduced cell migration. In addition, Guo et al (27) confirmed that BANCR contributes to the migration of colorectal cancer cells by the promotion of epithelial-mesenchymal transition. In the present study, it was revealed that the migration rate and invasive cell number of Huh7 cells were significantly reduced following BANCR shRNA transfection, these results consistently suggest that the downregulation of BANCR can suppress the migration and invasion of HCC cells.

It is accepted that the abnormal activation of numerous cellular and molecular signaling pathways are involved in the biological process of hepatocarcinogenesis (2832). MEK and MAPK signaling pathways are key pathways that participate in the regulation of cell proliferation, apoptosis, and differentiation (33,34). The suppression of MEK/ERK and MAPK signaling pathways using chemotherapeutic drugs has yielded major improvements in the management of HCC (35,36). In the present study, the downregulation of BANCR significantly inhibited the activity of MEK, ERK and JNK signaling pathways in Huh7 cells, but had no effect on P38 MAPK signaling. Thus, implying that MEK, ERK and JNK pathways may be implicated in BANCR-mediated tumor development, but not the P38 MAPK pathway. These findings are in line with previous studies demonstrating that the activation of MEK and MAPK signaling pathways are positively associated with the expression of BANCR (21,24,27).

Although the downregulation of lncRNA BANCR demonstrated a significant cancer inhibitory effect in the HCC cell line Huh7, the role of lncRNA BANCR on other HCC cell lines remains unclear. A limitation of the present study is that a single hepatocellular carcinoma cell line was used to investigate the effect of lncRNA BANCR. Thus, further studies are warranted to verify the function role of lncRNA BANCR in the development of HCC.

In conclusion, the results of the present study demonstrated that lncRNA BANCR was overexpressed in Huh7 cells when compared with normal liver cells L-02. Downregulation of BANCR significantly inhibited the proliferation, migration, invasion and aggravated apoptosis of Huh7 cells. Parallel to these biological changes, the downregulation of BANCR also suppressed the activity of MEK, ERK and JNK signaling pathways in HCC cells. The present study described a cancer promotion property of BANCR and provided a potential molecular target for the treatment of HCC.

References

1 

Knudsen ES, Gopal P and Singal AG: The changing landscape of hepatocellular carcinoma: Etiology, genetics and therapy. Am J Pathol. 184:574–583. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Ahn J and Flamm SL: Hepatocellular carcinoma. Dis Mon. 50:556–573. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Everson GT: Increasing incidence and pretransplantation screening of hepatocellular carcinoma. Liver Transpl. 6 (6 Suppl 2):S2–S10. 2000. View Article : Google Scholar : PubMed/NCBI

4 

El-Serag HB: Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology. 142:1264–1273.e1. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Bruix J and Llovet JM: Major achievements in hepatocellular carcinoma. Lancet. 373:614–616. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Olsen SK, Brown RS and Siegel AB: Hepatocellular carcinoma: Review of current treatment with a focus on targeted molecular therapies. Therap Adv Gastroenterol. 3:55–66. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Forner A and Bruix J: Biomarkers for early diagnosis of hepatocellular carcinoma. Lancet Oncol. 13:750–751. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Meseure D, Alsibai Drak K, Nicolas A, Bieche I and Morillon A: Long noncoding RNAs as new architects in cancer epigenetics, prognostic biomarkers, and potential therapeutic targets. Biomed Res Int. 2015:3202142015. View Article : Google Scholar : PubMed/NCBI

9 

Tsai MC, Spitale RC and Chang HY: Long intergenic noncoding RNAs: New links in cancer progression. Cancer Res. 71:3–7. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Wapinski O and Chang HY: Long noncoding RNAs and human disease. Trends Cell Biol. 21:354–361. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Spizzo R, Almeida MI, Colombatti A and Calin GA: Long non-coding RNAs and cancer: A new frontier of translational research? Oncogene. 31:4577–4587. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Flockhart RJ, Webster DE, Qu K, Mascarenhas N, Kovalski J, Kretz M and Khavari PA: BRAFV600E remodels the melanocyte transcriptome and induces BANCR to regulate melanoma cell migration. Genome Res. 22:1006–1014. 2012. View Article : Google Scholar : PubMed/NCBI

13 

McCarthy N: Epigenetics. Going places with BANCR. Nat Rev Cancer. 12:4512012. View Article : Google Scholar : PubMed/NCBI

14 

Zhang ZX, Liu ZQ, Jiang B, Lu XY, Ning XF, Yuan CT and Wang AL: BRAF activated non-coding RNA (BANCR) promoting gastric cancer cells proliferation via regulation of NF-κB1. Biochem Biophys Res Commun. 465:225–231. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Zheng H, Wang M, Jiang L, Chu H, Hu J, Ning J, Li B, Wang D and Xu J: BRAF-activated long non-coding RNA modulates papillary thyroid carcinoma cell proliferation through regulating thyroid stimulating hormone receptor. Cancer Res Treat. 48:698–707. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Shi Y, Liu Y, Wang J, Jie D, Yun T, Li W, Yan L, Wang K and Feng J: Downregulated long noncoding RNA BANCR promotes the proliferation of colorectal cancer cells via downregulation of p21 expression. PLoS One. 10:e01226792015. View Article : Google Scholar : PubMed/NCBI

17 

Su S, Gao J, Wang T, Wang J, Li H and Wang Z: Long non-coding RNA BANCR regulates growth and metastasis and is associated with poor prognosis in retinoblastoma. Tumour Biol. 36:7205–7211. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Wang Y, Guo Q, Zhao Y, Chen J, Wang S, Hu J and Sun Y: BRAF-activated long non-coding RNA contributes to cell proliferation and activates autophagy in papillary thyroid carcinoma. Oncol Lett. 8:1947–1952. 2014.PubMed/NCBI

19 

Sun M, Liu XH, Wang KM, Nie FQ, Kong R, Yang JS, Xia R, Xu TP, Jin FY, Liu ZJ, et al: Downregulation of BRAF activated non-coding RNA is associated with poor prognosis for non-small cell lung cancer and promotes metastasis by affecting epithelial-mesenchymal transition. Mol Cancer. 13:682014. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Li R, Zhang L, Jia L, Duan Y, Li Y, Bao L and Sha N: Long non-coding RNA BANCR promotes proliferation in malignant melanoma by regulating MAPK pathway activation. PLoS One. 9:e1008932014. View Article : Google Scholar : PubMed/NCBI

22 

Li L, Zhang L, Zhang Y and Zhou F: Increased expression of LncRNA BANCR is associated with clinical progression and poor prognosis in gastric cancer. Biomed Pharmacother. 72:109–112. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Su S, Gao J, Wang T, Wang J, Li H and Wang Z: Long non-coding RNA BANCR regulates growth and metastasis and is associated with poor prognosis in retinoblastoma. Tumour Biol. 36:7205–7211. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Jiang W, Zhang D, Xu B, Wu Z, Liu S, Zhang L, Tian Y, Han X and Tian D: Long non-coding RNA BANCR promotes proliferation and migration of lung carcinoma via MAPK pathways. Biomed Pharmacother. 69:90–95. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Zheng JH, Follis Viacava A, Kriwacki RW and Moldoveanu T: Discoveries and controversies in BCL-2 protein-mediated apoptosis. FEBS J. 283:2690–2700. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Brentnall M, Rodriguez-Menocal L, De Guevara RL, Cepero E and Boise LH: Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC Cell Biol. 14:322013. View Article : Google Scholar : PubMed/NCBI

27 

Guo Q, Zhao Y, Chen J, Hu J, Wang S, Zhang D and Sun Y: BRAF-activated long non-coding RNA contributes to colorectal cancer migration by inducing epithelial-mesenchymal transition. Oncol Lett. 8:869–875. 2014.PubMed/NCBI

28 

Galuppo R, Ramaiah D, Ponte OM and Gedaly R: Molecular therapies in hepatocellular carcinoma: What can we target? Dig Dis Sci. 59:1688–1697. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Zhou Q, Lui VW and Yeo W: Targeting the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Future Oncol. 7:1149–1167. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Bruix J and Sherman M: American Association for the Study of Liver Diseases: Management of hepatocellular carcinoma: An update. Hepatology. 53:1020–1022. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Sicklick JK, Li YX, Jayaraman A, Kannangai R, Qi Y, Vivekanandan P, Ludlow JW, Owzar K, Chen W, Torbenson MS and Diehl AM: Dysregulation of the Hedgehog pathway in human hepatocarcinogenesis. Carcinogenesis. 27:748–757. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Delire B and Stärkel P: The Ras/MAPK pathway and hepatocarcinoma: Pathogenesis and therapeutic implications. Eur J Clin Invest. 45:609–623. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Neuzillet C, Tijeras-Raballand A, de Mestier L, Cros J, Faivre S and Raymond E: MEK in cancer and cancer therapy. Pharmacol Ther. 141:160–171. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Wagner EF and Nebreda AR: Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 9:537–549. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Gao JJ, Shi ZY, Xia JF, Inagaki Y and Tang W: Sorafenib-based combined molecule targeting in treatment of hepatocellular carcinoma. World J Gastroenterol. 21:12059–12070. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Chen C and Wang G: Mechanisms of hepatocellular carcinoma and challenges and opportunities for molecular targeted therapy. World J Hepatol. 7:1964–1970. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2017
Volume 14 Issue 4

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
Li J, Wang J, Zhou W, Zhang S, Le Y and He R: Downregulation of BRAF‑activated non‑coding RNA suppresses the proliferation, migration and invasion, and induces apoptosis of hepatocellular carcinoma cells. Oncol Lett 14: 4751-4757, 2017
APA
Li, J., Wang, J., Zhou, W., Zhang, S., Le, Y., & He, R. (2017). Downregulation of BRAF‑activated non‑coding RNA suppresses the proliferation, migration and invasion, and induces apoptosis of hepatocellular carcinoma cells. Oncology Letters, 14, 4751-4757. https://doi.org/10.3892/ol.2017.6770
MLA
Li, J., Wang, J., Zhou, W., Zhang, S., Le, Y., He, R."Downregulation of BRAF‑activated non‑coding RNA suppresses the proliferation, migration and invasion, and induces apoptosis of hepatocellular carcinoma cells". Oncology Letters 14.4 (2017): 4751-4757.
Chicago
Li, J., Wang, J., Zhou, W., Zhang, S., Le, Y., He, R."Downregulation of BRAF‑activated non‑coding RNA suppresses the proliferation, migration and invasion, and induces apoptosis of hepatocellular carcinoma cells". Oncology Letters 14, no. 4 (2017): 4751-4757. https://doi.org/10.3892/ol.2017.6770