microRNA-195 functions as a tumor suppressor by inhibiting CBX4 in hepatocellular carcinoma

Retraction in: /10.3892/or.2021.8145

  • Authors:
    • Changli Zheng
    • Jingjing Li
    • Qi Wang
    • Wei Liu
    • Jianda Zhou
    • Rui Liu
    • Qinghai Zeng
    • Xiaowei Peng
    • Chenghui Huang
    • Peiguo Cao
    • Ke Cao
  • View Affiliations

  • Published online on: January 19, 2015     https://doi.org/10.3892/or.2015.3734
  • Pages: 1115-1122
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

microRNA-195 (miR-195) plays important roles in tumor metastasis and angiogenesis, yet its function and mechanism of action in hepatocellular carcinoma (HCC) remain to be elucidated. In this study, we aimed to confirm whether chromobox homolog 4 (CBX4) is a direct target gene of miR-195 and determine the functions of miR-195 through the CBX4 pathway. miR-195 expression was slightly lower in the HCC tissues compared with that in the adjacent normal tissues. In addition, western blotting and qRT-RCR results showed that both CBX4 mRNA and protein levels were downregulated upon miR-195 overexpression. Luciferase reporter assays revealed that CBX4 is a direct target gene of miR-195. Furthermore, overexpression of CBX4 significantly restored the proliferative, invasive and migratory capacities of the HepG2 cells. Finally, in vivo experiments confirmed that high expression of CBX4 in HepG2 cells promoted tumor growth. In conclusion, our study demonstrated that miR-195 acts as a tumor suppressor by directly targeting CBX4 in HCC. This finding suggests a potential novel strategy for therapeutic interventions of this disease.

Introduction

Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related mortality worldwide, with an increasing incidence and high death rates (1,2). HCC is a malignant tumor that is difficult to diagnose as early-stage disease. It is characterized by a high frequency of recurrence, metastasis following surgical resection, and resistance to common chemotherapy and radiotherapy, resulting in poor survival (3,4). Over the past few decades, there have been great advances in the treatment of HCC, yet overall patient survival remains low (5). Crucially, molecular mechanisms underlying tumor proliferation, invasion and migration may be therapeutic targets in HCC.

microRNAs (miRNAs), which regulate gene expression at the post-transcriptional level, are conserved non-coding RNAs of approximately 22 nucleotides (6). In mammals, mature miRNAs suppress protein expression by base-pairing with the 3′-untranslated region (3′-UTR) of targeted gene transcripts and directly degrading messenger RNA (mRNA) (7). The abnormal expression of miRNAs has been reported in many types of cancer, whereby miRNAs act as either suppressors or promoters (8). Recent studies have revealed that a series of miRNAs are involved in HCC tumor development. For example, miR-122 (9) and miR-29b (10) have been characterized to have anti-angiogenic and anti-metastatic functions in HCC. These miRNAs and their target genes can be treatment targets, or diagnostic and prognostic biomarkers for HCC clinical processing. However, miR-21 has been found to be a pro-metastatic miRNA in HCC (11). Interestingly, miR-195 is frequently downregulated and plays different roles in multiple cancer types (1216). A recent study found that miR-195 is associated with tumor metastasis and angiogenesis in HCC cells (17). As yet, the exact mechanisms of miR-195 in HCC development still remain largely unknown.

Chromobox homolog 4 (CBX4), also known as poly-comb 2 (PC2) or NBP16, is located on chromosome 17q25.3. It encodes a polycomb repressive complex 1 (PRC1)-associated protein (CBX4 protein) that is a member of the Polycomb group (PcG) proteins involved in chromatin remodeling and transcriptional regulation (18). The chromobox family includes five members: CBX2, CBX4, CBX6, CBX7 and CBX8 (19). PcG genes are transcriptional repressors related to cancer progression and stem cell maintenance (20). CBX7 is the best-studied of the Polycomb paralogs and is a master controller that extends cellular lifespan, delays senescence, promotes proliferation, and bestows pluripotency to adult and embryonic stem cells (2123). CBX4 protein is a SUMO E3 ligase that differs from other members of the CBX family (24). It exerts critical roles in biological functions by impacting numerous important proteins, such as HIPK2, CtBP and Bmi1 (25). CBX4 also functions as a pro-angiogenic gene, and is significantly correlated with hypoxia-induced VEGF expression and plays an important role in tumor angiogenesis by governing HIF-1α protein in HCC (26).

Based on these findings, we hypothesized that miR-195 may act as a tumor suppressor via downregulation of CBX4. Furthermore, miR-195 might directly target CBX4 as revealed by bioinformatics analysis software packages (Targetscan, PicTar, PITA, miRanda and miRDB). However, the relationship between miR-195 and CBX4 has not been previously reported.

In the present study, we demonstrated that overexpression of miR-195 reduced CBX4 expression at the transcriptional and protein level. We applied dual reporter gene assays to further confirm that CBX4 is a downstream target gene of miR-195. Upregulation of CBX4 markedly restored the proliferative, invasive, and migratory capacities of HCC cells in vitro. In vivo experiments also revealed that tumor growth capacity was recovered after CBX4 overexpression. Thus, miR-195 may be a promising molecular target for HCC therapy.

Materials and methods

Cell lines and tissue specimens

Human HCC tissues and matched normal tissues (located >5 cm away from the tumor) were collected from 10 patients who underwent HCC resection at Xiangya Hospital, Central South University. Informed consent was obtained from each patient, and the study was approved by the Ethics Committee of Xiangya Hospital, Central South University. Human HCC cell line HepG2 was purchased from ATCC (Manassas, VA, USA). HepG2 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Gibco, Grand Island, NY, USA), supplemented with 10% fetal bovine serum (FBS) at 37°C with 5% CO2 in a humidified atmosphere.

Plasmid transfection

A eukaryotic expression plasmid expressing fluorescently labeled miR-195 (hsa-mir-195, MI0000489) and a negative control (hsa-mir-195-NC, CON031) were purchased from GeneChem Biotechnology (Shanghai, China). The pcDNA3.1 plasmid (Invitrogen, Carlsbad, CA, USA) and psiCHECK™-2 vector (C8021; Promega, San Luis Obispo, CA, USA) were purchased from Auragene Biotechnology (Changsha, Hunnan, China). The pcDNA3.1 plasmid expresses CBX4 and that without CBX4 3′-UTR was constructed by Auragene Biotechnology (Changsha, Hunnan, China). psiCHECK™-2 vector constructs expressing either intact CBX4 or CBX4 with a mutation in the putative binding site for miR-195 were generated. The primers of CBX4 3′-UTR (forward 5′-CTCGAGAACTGCCTCACCG TTACTT-3′, reverse 5′-GCGGCCGCAATATTTACATTCA AGCAGG-3′) and mutated CBX4 3′-UTR (forward 5′-GCGG CCGCAACTGCCTCACCGTTACTT-3′, reverse 5′-CTC GAGAATATTTACATTCAAGCAGG-3′) were designed using Primer 5.0 software. The PCR amplified sequences were inserted into the psiCHECK™-2-vector within the XhoI/NotI sites. Plasmids were introduced into the HepG2 cells using RNAiMAX and Lipofectamine 2000 (both from Invitrogen) when cell confluency had reached 40–50% in 6-cm culture dishes. After 48 h of transfection, fluorescence microscopy and qRT-PCR were performed to check transfection efficiency.

Quantitative real-time RT-PCR

miR-195 and CBX4 were acquired from GenBank. The primers were designed using Primer 5.0 software, and the sequences were as follows: CBX4 forward, 5′-TGGAGTATCTGGTGAAATGGA-3′ and reverse, 5′-ACGACGGGCAAAGGTAGGCAC-3′; miR-195 forward, 5′-TAGCAGCACAGAAATATTGGC-3′ and reverse, 5′-GCG AGCACAGAATTAATACGAC-3′. Total RNA from cells and tissues was extracted with TRIzol reagent (Invitrogen) according to the manufacturer’s protocol. Reverse-transcribed cDNA was synthesized using a reverse transcription synthesis system (Toyobo, Osaka, Japan). Quantitative real-time PCR analyses were performed with MJ mini PCR (MiniOption; Bio-Rad Laboratories, CA, USA). Samples were compared using the relative CT method, where the relative expression of miR-195 was normalized to that of U6, while that of CBX4 used ACTB (β-actin) as an internal control for mRNA quantification.

Cell proliferation assay

The Cell Counting Kit-8 (CCK-8; Dojindo Laboratories, Kumamoto, Japan) colorimetric assay was used to measure cell proliferation. After transfection, cells were seeded in 96-well plates at a density of 5×103 cells/well. Cells were cultured for 24 h, and then the supernatant was removed and 100 μl of DMEM containing 10 μl of CCK-8 was added to each well for 3 h at 37°C. The absorbance at 450 nm was measured with a plate reader (Thermo Multiskan MK3 spectrophotometer; Thermo Fisher Scientific, Waltham, MA, USA). The OD value was determined and used to construct a growth curve to assess cell proliferation.

For the colony formation assay, HepG2 cells were seeded in 10-cm dishes (1,000/plate) after transfection and maintained in complete culture medium for 21 days. Next, cells were fixed in 4% paraformaldehyde for 15 min and stained with Giemsa dye. Images of cells were captured, and the number of clones was calculated.

Scratch migration and Transwell invasion assays

Cells were seeded in 24-well plates (1×105 cells/well) after transfection and cultured for 12 h. Upon reaching the appropriate confluency, the cell monolayer was scratched with a 10-μl tip. Images were captured at different time points (0 and 48 h) by microscopy to assess the rate of gap closure. The percentage of closure of the gap area was calculated using the following formula: GapΔ(%) = Gap48(%) − Gap0(%), where GapΔ is the occupied gap area after 48 h; Gap48 is the gap area at 48 h; Gap0 is the gap area at baseline.

Transwell invasion assays were performed to measure the invasive capacity of the transfected cells plated on 8-μm pore size Matrigel-coated membranes (1×105 cells/well in serum-free medium), which were in turn placed in the top chamber of 24-well Transwell plates. The bottom chamber contained 500 μl chemotactic factor. After 24 h, cells on the upper surface were removed, while cells attached to the membranes were fixed in 4% paraformaldehyde for 20 min and stained with hematoxylin. The results of the Transwell assay were imaged and the number of invasive cells was evaluated by the resultant OD value.

Western blotting

Western blotting was performed as described previously (27). Cells were harvested 48 h after transfection, and proteins were extracted and then quantified with a BCA protein assay kit and separated on 10% SDS-PAGE gels and transferred onto PVDF membranes (Millipore, Billerica, MA, USA). We, then, followed standard procedures using rabbit anti-human CBX4 polyclonal antibody (1:1,000; HPA008228; Sigma-Aldrich) and mouse anti-human β-actin monoclonal antibody (1:1,000; BS6007M; Bioworld Technology). Protein levels were quantified using β-actin as a loading control, and immunoreactive bands were visualized by electrochemiluminescence.

Dual luciferase reporter assays

Luciferase reporter assays were performed using the psiCHECK™-2-CBX4-3′-UTR vector. HepG2 cells were co-transfected with hsa-mir-195 or hsa-mir-195-NC (1 μg) followed by the psiCHECK™-2-CBX4-3′-UTR or psiCHECK2-CBX4-Mut-3′-UTR (1 μg). Cells were harvested 48 h after transfection and analyzed with the Dual-Luciferase reporter gene assay kit E1910 (Promega). The firefly luciferase values were normalized to Renilla lucif-erase values as an internal control.

Nude mouse xenograft studies

All experimental procedures were performed according to NIH Animal Care, and the entire experiment was approved by the Ethics Committee of Faculty of Experimental Animals, Central South University. Male BALB/c-nu/nu (aged 4–6 weeks) were purchased from the animal laboratory of Third Xiangya Hospital of Central South University and maintained under specific pathogen-free conditions. To clarify the effect of CBX4 in vivo, 9 nude mice were injected subcutaneously in the ventral trunk with 2×106 cells (NC, miR-195, and miR-195+CBX4) in 200 μl DMEM. Tumor volume was calculated using the formula V (mm3) = 0.5 × a × b2 where a is the maximum length to diameter; b is the maximum transverse diameter. Nude mice were sacrificed at 30 days after tumor implantation, and tumor volume and weight were measured.

Statistical analysis

All experiments were repeated at least three times, and the results are expressed as the mean ± SD (n=3). Briefly, statistical analyses were carried out using SPSS 16.0 software (SPSS, Chicago, IL, USA). The results were assessed by one-way ANOVA or the Student’s t-test. All statistical tests were two-sided, and a P-value of <0.05 was considered to indicate a statistically significant difference.

Results

miR-195 is aberrantly downregulated in HCC tissue samples

We used qRT-PCR to examine the miR-195 expression levels in 10 HCC tissue samples and corresponding normal tissues. As shown in Fig. 1A, compared with normal tissues, most HCC tissues showed significantly lower expression of miR-195, similar to the findings of Wang et al (3).

Since the expression of miR-195 was significantly downregulated compared with that in the matched normal tissues, we postulated that miR-195 may function as a tumor suppressor through downregulation of CBX4 in HCC. To investigate this hypothesis, we overexpressed miR-195 in HepG2 cells through lipofection with hsa-miR-195, and constructed a stably transfected cell line through hygromycin selection. The subgroups were as follows: HepG2, a blank group without any treatment; NC-HepG2, a control group transfected with empty plasmid. The transfection efficiency was evaluated by fluorescence microscopy and qRT-PCR (Fig. 1B). Successful overexpression of miR-195 was confirmed by qRT-PCR, whereas there was no difference in its expression level between the blank and negative control groups. Therefore, these results indicated that the transfection efficiency was satisfactory.

CBX4 is a target gene of miR-195 in HepG2 cells

CBX4 was recently identified to be a cancer-promoting gene (26). According to multiple microRNA target gene prediction software packages, such as TargetScan, CBX4 is highly predicted to be a target of miR-195. To confirm this hypothesis, we detected the endogenous expression levels of CBX4 in HepG2, NC-HepG2 and miR-195-HepG2 cell lines. Western blotting (Fig. 2A) and qRT-PCR (Fig. 2B) analyses revealed that a significant inverse correlation was observed between the expression of miR-195 and CBX4 protein. In addition, qRT-PCR (Fig. 3A) analysis showed that CBX4 had significantly higher expression in the HCC clinical samples. These results suggest that the overexpression of miR-195 may account for CBX4 downregulation in HCC.

To further explore this hypothesis, we amplified the CBX4 3′-UTR containing the target sequence in which there are two possible binding sites. This was either left intact or mutated, then inserted into a psiCHECK™-2 luciferase reporter vector (Fig. 2C). As shown in Fig. 2D, miR-195 suppressed the luciferase activity of the CBX4 3′-UTR, while mutation of the miR-195 binding sites had no visible effect on the HepG2 cells. When cells were transfected with the mutated CBX4 or transfected with the miR-195-NC or left non-treated, the luciferase activity was basically the same and exhibited no significant difference. These results suggest that miR-195 can bind to the 3′-UTR of CBX4. In summary, CBX4 is a miR-195 target gene that is inhibited by miR-195 at the post-transcriptional level.

Overexpression of CBX4 restores HepG2 cell proliferative, migration and invasion

HepG2 cells were divided into three groups: a control group transfected with an empty plasmid (NC), a group transfected with hsa-miR-195 (miR-195), and cells co-transfected with hsa-miR-195 and CBX4 (miR-195+CBX4). Western blotting (Fig. 3B) of CBX4 protein expression in HepG2 cells demonstrated that the transfection efficiency was satisfactory. To elucidate whether miR-195 affects HepG2 cell proliferation via downregulation of CBX4, CCK-8 assays were employed. The miR-195 group proliferated most slowly compared with either the control (NC) or miR-195+CBX4 group (Fig. 4A), whereas no obvious difference was detected between the NC and miR-195+CBX4 groups. Colony formation assays were also used to evaluate cell proliferation and plating efficiency after overexpression of miR-195 and CBX4. The results showed that the number of colonies formed when miR-195 was overexpressed was much lower than that exhibited by the NC and co-transfected (miR-195+CBX4) groups (Fig. 4B), whereas there was no obvious difference between the NC and miR-195+CBX4 groups. Our studies have therefore demonstrated that overexpression of miR-195 limits HepG2 proliferation, yet CBX4 upregulation significantly recovered this inhibitory effect, suggesting that miR-195 suppresses HCC growth mainly by inhibiting CBX4.

To verify the possible role of miR-195 in HCC metastasis, the effects of miR-195 and CBX4 on the migration and invasion of HepG2 cells were analyzed in vitro. Scratch migration assays were performed to test the migratory ability of HepG2 cells. The results showed that the scratched areas were occupied by the NC and miR-195+CBX4 groups to a much greater extent when compared to the miR-195 group (Fig. 4C). Transwell invasion assays were also performed to explore the effects on invasive capacity. As expected, the number of invading cells in the miR-195-overexpressing group was much lower than this number in the NC or miR-195+CBX4 group (Fig. 4D). In short, miR-195 inhibits HepG2 cell invasion and migration, indicating that it suppresses metastasis in HCC. However, overexpression of CBX4 markedly recovered tumor cell invasion and migration.

Overexpression of CBX4 restores tumor growth in nude mice

We further validated the effect of the upregulation of miR-195 and CBX4 in HepG2 cells on tumor growth in nude mice. Nude mice were s.c. inoculated with the HepG2 cells transfected with the blank empty vector, the miR-195 vector, or the miR-195+CBX4 vector. After 30 days, the mice were sacrificed. The average volume of the tumors in the NC and miR-195+CBX4 group was 233.751±54.535 and 144.794±22.460 mm3, which was notably higher than that in the miR-195 group (49.303±23.895 mm3) (Fig. 5A and B). The average tumor mass in the NC and miR-195+CBX4 group was 0.700±0.162 g and 0.445±0.023, which was also higher than that in the miR-195 group (0.152±0.065 g) (Fig. 5C).

These results suggest that miR-195 functions as a tumor suppressor through inhibition of CBX4, and overexpression of CBX4 recovers the tumor growth ability of HCC in vivo.

Discussion

Tumor overgrowth and metastasis are two of the most important hallmarks of malignant tumors, and metastasis is the major cause of tumor recurrence and patient death (28,29). Therefore, understanding the underlying molecular pathways involved in the process of tumor growth and metastasis is crucial.

miRNAs that possess antiproliferative or antimetastatic activities may provide novel targets for anticancer therapies. Increasing evidence suggests that the dysregulation of miRNAs participates in HCC progression (30,31). For example, Zhou et al (32) demonstrated that miR-625 suppressed HCC cell migration, invasion and metastasis in vitro and in vivo through downregulation of IGF2BP1. Shih et al (33) reported that low miR-214 expression is negatively correlated with that of hepatoma-derived growth factor (HDGF) and contributes to tumor angiogenesis in HCC.

CBX4 encodes a polycomb repressive complex 1 (PRC1) associated protein (CBX4) that is a member of the Polycomb group (PcG) proteins involved in chromatin remodeling and transcriptional regulation (18). It plays a critical role in tumor angiogenesis by governing HIF-1α protein in HCC (26). Although CBX4 protein was detectable in both the nuclei and cytoplasm of HCC tissues, only abnormal expression of CBX4 in cytoplasm was correlated with tumor progression, including tumor volume and weight, pathological differentiation, and tumor, node, metastasis classification system stages (3).

We identified that miR-195 may target CBX4 using bioinformatics analysis software packages (Targetscan, PicTar, PITA, miRanda and miRDB). miR-195 is frequently downregulated and plays different roles in multiple cancer types, including HCC, colorectal, breast and bladder cancer (1216). miR-195 has been shown to block the G1/S transition of the cell cycle by regulating the expression of CCND1/3, CDK4/6 and E2F3 (12,16) and to suppress tumor development by targeting BCL-2 and BCL-w (13,34). miR-195 directly regulates WEE1 expression in malignant melanoma (35), but as yet, the exact regulatory mechanisms of miR-195 in HCC development have not been explored.

In this study, we found that miR-195 and CBX4 mRNA were aberrantly expressed in most HCC clinical tissues. To test our hypothesis that miR-195 inhibits CBX4 expression, which in turn prevents the development of HCC, we used qRT-PCR and western blotting to measure CBX4 mRNA and protein expression levels following miR-195 overexpression in HepG2 cells. CBX4 expression was reduced when miR-195 was overexpressed, indicating that CBX4 is a target gene of miR-195 in vitro, which was confirmed by dual luciferase activity assays.

To further explore the inhibitory role of miR-195 in CBX4 expression, we overexpressed miR-195 and CBX4 in HepG2 cells. Upregulation of CBX4 markedly restored HepG2 cell proliferation, invasion and migration in vitro. The results of the Transwell invasion and scratch migration assays demonstrated that both migration and invasion were restored by CBX4, suggesting that it may function as a promoter of metastasis in HCC. Taken together, these findings suggest that miR-195 suppresses HCC development through inhibition of

CBX4

Nude mouse xenograft studies were performed to further understand the cancer-promoting function of CBX4 in vivo. The average tumor volume and mass in the miR-195+CBX4 and NC groups were significantly higher than these values in the miR-195 group. These results strongly revealed that overexpression of CBX4 restored tumor growth ability, which was regulated by miR-195 in HCC. However, the underlying mechanisms of the CBX4 pathway remain to be elucidated. In future studies, we will explore the miR-195 targeting of CBX4 downstream signaling pathways.

In summary, our study demonstrated that miR-195 expression was markedly decreased in most HCC tissues compared with that in matched normal tissues. Upregulation of miR-195 limited the expression of CBX4, which was confirmed as a target gene of miR-195. As a target gene of miR-195, overexpression of CBX4 restored HepG2 cell proliferation, invasion and migration in vitro. Moreover, we also confirmed that CBX4 recovered the tumor growth ability of HepG2 cells in vivo. Together with the findings from this study, the newly identified miR-195/CBX4 axis might contribute to the identification of a promising tumor suppressor and molecular target that provides a new strategy for anticancer clinical therapies in HCC.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (grant nos. 81372140, 81301688, 81272192, 81171882), Ph.D. Programs Foundation of Ministry of Education of China (nos. 20130162110050 and 20130162120093), Program for New Century Excellent Talents in University (NCET-11-0527), China Postdoctoral Science Foundation (2014M552167), Postdoctoral Foundation of Central South University (no. 131425), Project of the Nature Science Foundation of Hunan Province of China (no. 12JJ4088), Project of the Department of Science and Technology of Hunan Province (nos. 2013FJ6003, 2012FJ4344, 2014FJ3120), 125 Talent Project of the Third Xiangya Hospital of Central South University and the Freedom Explore Program of Central South University (no. 2011QNZT193).

References

1 

Forner A, Llovet JM and Bruix J: Hepatocellular carcinoma. Lancet. 379:1245–1255. 2012. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Wang B, Tang J, Liao D, et al: Chromobox homolog 4 is correlated with prognosis and tumor cell growth in hepatocellular carcinoma. Ann Surg Oncol. 20:S684–S692. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Aravalli RN, Steer CJ and Cressman EN: Molecular mechanisms of hepatocellular carcinoma. Hepatology. 48:2047–2063. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Tameda M, Sugimoto K, Shiraki K, et al: Collagen triple helix repeat containing 1 is overexpressed in hepatocellular carcinoma and promotes cell proliferation and motility. Int J Oncol. 45:541–548. 2014.PubMed/NCBI

6 

Lim LP, Lau NC, Garrett-Engele P, et al: Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 433:769–773. 2005. View Article : Google Scholar : PubMed/NCBI

7 

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

8 

Landgraf P, Rusu M, Sheridan R, et al: A mammalian microRNA expression atlas based on small RNA library sequencing. Cell. 129:1401–1414. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Bai S, Nasser MW, Wang B, et al: microRNA-122 inhibits tumorigenic properties of hepatocellular carcinoma cells and sensitizes these cells to sorafenib. J Biol Chem. 284:32015–32027. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Fang JH, Zhou HC, Zeng C, et al: MicroRNA-29b suppresses tumor angiogenesis, invasion, and metastasis by regulating matrix metalloproteinase 2 expression. Hepatology. 54:1729–1740. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Zhou L, Yang ZX, Song WJ, et al: MicroRNA-21 regulates the migration and invasion of a stem-like population in hepatocellular carcinoma. Int J Oncol. 43:661–669. 2013.PubMed/NCBI

12 

Xu T, Zhu Y, Xiong Y, Ge YY, Yun JP and Zhuang SM: MicroRNA-195 suppresses tumorigenicity and regulates G1/S transition of human hepatocellular carcinoma cells. Hepatology. 50:113–121. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Liu L, Chen L, Xu Y, Li R and Du X: microRNA-195 promotes apoptosis and suppresses tumorigenicity of human colorectal cancer cells. Biochem Biophys Res Commun. 400:236–240. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Li D, Zhao Y, Liu C, et al: Analysis of miR-195 and miR-497 expression, regulation and role in breast cancer. Clin Cancer Res. 17:1722–1730. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Lin Y, Wu J, Chen H, et al: Cyclin-dependent kinase 4 is a novel target in micoRNA-195-mediated cell cycle arrest in bladder cancer cells. FEBS Lett. 586:442–447. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Zhang QQ, Xu H, Huang MB, et al: MicroRNA-195 plays a tumor-suppressor role in human glioblastoma cells by targeting signaling pathways involved in cellular proliferation and invasion. Neuro Oncol. 14:278–287. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Wang R, Zhao N, Li S, et al: MicroRNA-195 suppresses angiogenesis and metastasis of hepatocellular carcinoma by inhibiting the expression of VEGF, VAV2, and CDC42. Hepatology. 58:642–653. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Luis NM, Morey L, Mejetta S, et al: Regulation of human epidermal stem cell proliferation and senescence requires polycomb- dependent and -independent functions of Cbx4. Cell Stem Cell. 9:233–246. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Bracken AP and Helin K: Polycomb group proteins: navigators of lineage pathways led astray in cancer. Nat Rev Cancer. 9:773–784. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Gieni RS and Hendzel MJ: Polycomb group protein gene silencing, noncoding RNA, stem cells, and cancer. Biochem Cell Biol. 87:711–746. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Gil J, Bernard D, Martinez D and Beach D: Polycomb CBX7 has a unifying role in cellular lifespan. Nat Cell Biol. 6:67–72. 2004. View Article : Google Scholar

22 

Bernard D, Martinez-Leal JF, Rizzo S, et al: CBX7 controls the growth of normal and tumor-derived prostate cells by repressing the Ink4a/Arf locus. Oncogene. 24:5543–5551. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Yap KL, Li S, Munoz-Cabello AM, et al: Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell. 38:662–674. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Kagey MH, Melhuish TA and Wotton D: The polycomb protein Pc2 is a SUMO E3. Cell. 113:127–137. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Ismail IH, Gagne JP, Caron MC, et al: CBX4-mediated SUMO modification regulates BMI1 recruitment at sites of DNA damage. Nucleic Acids Res. 40:5497–5510. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Li J, Xu Y, Long XD, et al: Cbx4 governs HIF-1α to potentiate angiogenesis of hepatocellular carcinoma by its SUMO E3 ligase activity. Cancer Cell. 25:118–131. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Hong J, Hu K, Yuan Y, et al: CHK1 targets spleen tyrosine kinase (L) for proteolysis in hepatocellular carcinoma. J Clin Invest. 122:2165–2175. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Hanahan D and Weinberg RA: Hallmarks of cancer: the next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Tang ZY: Hepatocellular carcinoma - cause, treatment and metastasis. World J Gastroenterol. 7:445–454. 2001.

30 

Lujambio A and Lowe SW: The microcosmos of cancer. Nature. 482:347–355. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Su H, Yang JR, Xu T, et al: MicroRNA-101, downregulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity. Cancer Res. 69:1135–1142. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Zhou X, Zhang CZ, Lu SX, et al: miR-625 suppresses tumour migration and invasion by targeting IGF2BP1 in hepatocellular carcinoma. Oncogene. Mar 17–2014.(Epub ahead of print). View Article : Google Scholar

33 

Shih TC, Tien YJ, Wen CJ, et al: MicroRNA-214 downregulation contributes to tumor angiogenesis by inducing secretion of the hepatoma-derived growth factor in human hepatoma. J Hepatol. 57:584–591. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Yang X, Yin J, Yu J, et al: miRNA-195 sensitizes human hepatocellular carcinoma cells to 5-FU by targeting BCL-w. Oncol Rep. 27:250–257. 2012.

35 

Bhattacharya A, Schmitz U, Wolkenhauer O, Schönherr M, Raatz Y and Kunz M: Regulation of cell cycle checkpoint kinase WEE1 by miR-195 in malignant melanoma. Oncogene. 32:3175–3183. 2013. View Article : Google Scholar

Related Articles

Journal Cover

March-2015
Volume 33 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zheng C, Li J, Wang Q, Liu W, Zhou J, Liu R, Zeng Q, Peng X, Huang C, Cao P, Cao P, et al: microRNA-195 functions as a tumor suppressor by inhibiting CBX4 in hepatocellular carcinoma Retraction in /10.3892/or.2021.8145. Oncol Rep 33: 1115-1122, 2015
APA
Zheng, C., Li, J., Wang, Q., Liu, W., Zhou, J., Liu, R. ... Cao, K. (2015). microRNA-195 functions as a tumor suppressor by inhibiting CBX4 in hepatocellular carcinoma Retraction in /10.3892/or.2021.8145. Oncology Reports, 33, 1115-1122. https://doi.org/10.3892/or.2015.3734
MLA
Zheng, C., Li, J., Wang, Q., Liu, W., Zhou, J., Liu, R., Zeng, Q., Peng, X., Huang, C., Cao, P., Cao, K."microRNA-195 functions as a tumor suppressor by inhibiting CBX4 in hepatocellular carcinoma Retraction in /10.3892/or.2021.8145". Oncology Reports 33.3 (2015): 1115-1122.
Chicago
Zheng, C., Li, J., Wang, Q., Liu, W., Zhou, J., Liu, R., Zeng, Q., Peng, X., Huang, C., Cao, P., Cao, K."microRNA-195 functions as a tumor suppressor by inhibiting CBX4 in hepatocellular carcinoma Retraction in /10.3892/or.2021.8145". Oncology Reports 33, no. 3 (2015): 1115-1122. https://doi.org/10.3892/or.2015.3734