Inhibition of cell proliferation and metastasis of human hepatocellular carcinoma by miR-137 is regulated by CDC42

  • Authors:
    • Ming Gao
    • Liying Liu
    • Shenglei Li
    • Xudong Zhang
    • Zhiwei Chang
    • Mingzhi Zhang
  • View Affiliations

  • Published online on: September 9, 2015     https://doi.org/10.3892/or.2015.4261
  • Pages: 2523-2532
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In the present study, we evaluated the mechanisms of CDC42 in association with the microRNA-137 (miR-137)-induced inhibition of human hepatocellular carcinoma (HCC). The gene expression levels of miR-137 were evaluated in HCC cell lines. Direct association of miR-137 with its downstream target, cell division control protein 42 (CDC42), was evaluated by dual-luciferase assay, western blot analysis and correlation analysis using clinical tumor samples. In the HCC HuH7 and MHCC97L cell lines, miR-137 was upregulated to inhibit cell proliferation and metastasis in vitro and tumor growth in vivo. CDC42 was overexpressed in the HuH7 and MHCC97L cells to evaluate its effect on the miR-137-mediated antitumor effects. Furthermore, possible crosstalk between CDC42 and another miR-137 target gene, AKT2, was evaluated by co-overexpressing CDC42 and AKT2 in the HuH7 and MHCC97L cells and examining their effects on miR-137-mediated HCC regulation. miR-137 was confirmed to be downregulated in the HCC cell lines. Dual‑luciferase assay showed that CDC42 was directly targeted by miR-137, and western blotting showed that CDC42 was downregulated by miR-137 upregulation in the HuH7 and MHCC97L cells. In human tumors, the expression levels of CDC42 and miR-137 were inversely correlated. The inhibitory effects of miR-137 on HCC proliferation, metastasis and in vivo tumor growth were all ameliorated by CDC42 overexpression. Furthermore, co-overexpression of AKT2 in addition to CDC42 additively reduced the inhibition of miR-137 on HCC proliferation and metastasis, suggesting two independent pathways of CDC42 and AKT2 in miR-137-mediated HCC regulation. Our study demonstrated that CDC42 independently regulated the antitumor effects of miR-137 in human HCC.

Introduction

Hepatocellular carcinoma (HCC) is the most common form of liver cancer in men and women over 50 years of age. In the past decade, despite the great advances in HCC diagnosis and treatment, the incidence rates, as well as the mortality rates of HCC patients worldwide are increasing (1). Particularly in the Asian or Chinese population, the numbers of HCC patients and HCC-related cancer deaths are almost twice these numbers in Caucasian patients, due to common infection with Helicobacter pylori or hepatitis B virus (2). Thus, it is critical to elucidate the underlying mechanisms of HCC proliferation and metastasis in order to provide more accurate diagnosis and advanced optimal treatment strategies for HCC patients.

MicroRNAs (miRNAs) are families of 18–22 nucleotide non-coding RNAs that bind to the 3′-untranslated regions (3′-UTR) of target mRNAs to negatively modulate gene and protein expression by DNA or protein degradation in both animals and humans (3). In the past few decades, mounting evidence demonstrates that miRNAs play a critical role in various stages of carcinogenesis, cancer proliferation and cancer metastasis of human cancers, including HCC (46). In HCC, numerous cancer-associated miRNAs act as either oncogenes, such as miR-494, miR-93 and miR-184 (79), or tumor-suppressors, such as miR-31, miR-29c and miR-148a (1012). A recent study found that miR-137 is a tumor-suppressor miRNA in HCC, as miR-137 was downregulated in HCC and its subsequent upregulation inhibited HCC proliferation and migration in in vivo xenografts (13). However, it is known that miR-137 may exert its cancer regulatory effects through multiple genes (14,15). Therefore, it is important to explore the full scope of the downstream genes associated with miR-137 to better understand its regulation in HCC.

One of the common target genes of miR-137 is cell division cycle 42 (CDC42), a GTPase of the Rho family (1618). CDC42 itself is also highly associated with cancer regulation in many types of human carcinomas (19,20). A mouse model of CDC42 deficiency showed that it induced the development of HCC (21). In humans, CDC42 was found to be weakly expressed in clinical tumor samples (22). While this body of evidence points to a tumor-suppressing role of CDC42 in HCC, it is notable that CDC42 also acts as an oncogenic factor as knockdown of CDC42 inhibited HCC migration in vitro (23). Therefore, this conflicting body of evidence suggests that complex signaling pathways may be associated with CDC42 regulation in HCC.

In the present study, we explored the possible molecular association between CDC42 and miR-137 in HCC regulation. We examined whether miR-137 directly targets CDC42 in HCC, and whether CDC42 exerts any regulatory effects on miR-137-induced inhibition of HCC proliferation and metastasis, the two key properties of human HCC. Furthermore, we examined whether there is crosstalk between CDC42 and other miR-137 target genes in HCC. The results of our study may help elucidate the molecular profile of miRNA regulation in human HCC.

Materials and methods

HCC cell lines

Human HCC cell lines, HuH7, BEL-7402, HEPG2 and HEP3B and a normal human hepatocyte cell line (THLE-2) were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). MHCC97L was kindly provided by the Liver Cancer Institute of Zhongshan Hospital at Fudan University (Shanghai, China). All cell lines were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (both from Sigma-Aldrich, USA), 100 IU/ml penicillin G and 100 µg/ml streptomycin at 37°C in 5% CO2. Once confluency was achieved, the cells were passaged with replenished medium every 3 or 4 days.

RNA isolation and quantitative real-time PCR (qRT-PCR)

To extract RNA, the cell or tumor samples were homogenized in TRIzol reagent (Invitrogen, USA). Reverse-transcription was carried out by regular PCR using a RT-PreMix kit (SBS Genetech, China) according to the manufacturer's protocol. To quantitatively measure the gene expression level of miR-137, miRNA qRT-PCR was carried out using a Hairpin-it™ miRNAs Real-Time PCR Quantitation kit (GenePharma, China) with internal control of U6 snRNA, according to the manufacturer's protocol. To quantify mRNA expression of CDC42 and AKT2, qRT-PCR was carried out using a SYBR Green PCR Master Mix (Applied Biosystems, USA) with internal control of the 18s gene, according to the manufacturer's protocol. All primer sets were purchased from SBS Genetech. Gene expression levels were quantified by 2−ΔΔct methods against internal control genes and are reported as relative fold-changes.

MicroRNA-137 upregulation

Lentiviruses carrying the mature hsa-miR-137 mimic (miR-137) or its negative control miRNA (miR-C) were obtained from RiboBio (China). Lentiviral transfection of the HuH7 and MHCC97L cells was carried out using Lipofectamine 2000 reagent (Invitrogen) according to the manufacturer's protocol for 24 h, followed by replenishment with fresh medium.

Dual-luciferase reporter assay

The 3′-UTR of human CDC42 including the putative miR-137 binding site was amplified from a human liver cDNA library and then cloned into the SpeI/HindIII site of a pMIR-REPORT luciferase vector to generate the wild-type (WT) CDC42 luciferase reporter (CDC42-WT; Ambion, USA). A mutant CDC42 3′UTR with a modified miR-137 binding site was generated using a Site-Directed Mutagenesis kit (SBS Genetech). It was also cloned into the pMIR-REPORT vector to generate the mutant CDC42 luciferase reporter (CDC42-MT). In both HuH7 and MHCC97L cells, miR-137 was co-transfected with CDC42-WT, CDC42-MT or a Renilla luciferase control vector (Luc-C) for 48 h. The luciferase activities were measured by a dual-luciferase reporter assay (Promega, USA), and normalized to the activity of the Renilla control.

Human tumor specimens

Thirteen human HCC specimens were collected by surgery between June 2013 and April 2015 at the Department of Oncology of The First Affiliated Hospital of Zhengzhou University, and the Department of Oncology of The First People's Hospital of Zhengzhou in Zhengzhou, China. Consent forms were signed by all patients. The clinical and laboratory protocols were approved by the Ethic Committees at the participating institutes.

Western blot analysis

HuH7 and MHCC97L cells were collected and lysed in a lysis buffer containing 50 mM Tris (pH 7.6), 150 mM NaCl, 1 mM EDTA, 10% glycerol, and 0.5% NP-40 and protease inhibitor cocktail (Millipore, USA). The extracted cell proteins were dissolved on 10% SDS-PAGE gel, transferred to nitrocellulose membranes, and incubated with a primary rabbit antibody against human CDC42 (1:200; Sigma-Aldrich) at 4°C overnight. On the second day, after washing with Tris-buffered saline (3 × 10 min), the membranes were incubated with a horseradish peroxidase-conjugated secondary antibody (Millipore). Each blot was visualized with enhanced chemiluminescence (Pierce, USA) according to the manufacturer's protocol.

Overexpression assay

Whole DNA sequences of CDC42 and AKT2 were amplified from a human liver cDNA library and confirmed with sequencing. The DNA sequences were then cloned into a recombinant plasmid eukaryotic expression vector pcDNA3.1 (Invitrogen) to construct overexpressing vectors of CDC42 (pcDNA3.1/CDC42), and AKT2 (pcDNA3.1/AKT2). An empty vector (pcDNA3.1/+) was used as control. The transfection was carried out using Lipofectamine 2000 reagent according to the manufacturer's protocol for 24 h, followed by replenishment with fresh medium.

In vitro proliferation assay

HuH7 and MHCC97L cells were initially transfected with the lentiviruses and/or the over-expressing vectors for 24 h. After 24 h, the culture medium was replenished and the proliferation of HCC cells was characterized using a 3-(4,5-dimethylthazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay for 5 consecutive days. Briefly, after washing the cells with PBS, 1 ml of 0.5 mg/ml MTT was added into the culture for 4 h. MTT was aspirated and 300 µl isopropanol was immediately added. The optical density (OD) was measured at a wavelength of 570 nm.

In vitro migration assay

Metastasis in the HuH7 and MHCC97L cells was measured using a QCM chemotaxis 96-well migration assay (Chemicon, USA). On the first day, HuH7 and MHCC97L cells were transfected with the lenti-viruses and/or the overexpressing vectors. During this time, the upper chamber of the QCM chemotaxis plate was coated with 0.1% gelatin (in PBS) overnight. On the second day, HuH7 and MHCC97L cells were resuspended and re-plated in the upper chamber with RPMI-1640 medium. The lower chamber was filled with RPMI-1640 medium with the addition of 10% FBS as a chemoattractant. After 24 h, the HCC cells that migrated into the lower chamber were fixed with 4% PFA and stained with hematoxylin and eosin (H&E). The relative migration rates were measured by averaging the cell numbers in 5 random 0.1 × 0.1 mm regions and normalizing to the cell number under control conditions.

In vivo transplantation assay

HuH7 cells were transfected with the lentiviruses and/or the overexpressing vectors. After 24 h, the cells were collected and resuspended. Approximately 1 million cells were subcutaneously inoculated into the left flank of 2-month-old female nude mice. The in vivo tumor growth assay was carried out by measuring the in vivo tumor volume based on the equation: length × width2/2. At the end of the 5-week transplantation, the tumors were extracted and Ki-67 immunostaining (BD Biosciences) was carried out on paraffin sections.

Statistical analysis

In the present study, data are presented as the mean ± standard deviation. Statistical analysis with the two-tailed Student's t-test (SPSS, version 11.0) was carried out to evaluate the difference between results. Differences were considered significant at P<0.05. All experiments were repeated in triplicates.

Results

MicroRNA-137 is downregulated in HCC cell lines and targets CDC42

A previous study demonstrated that miR-137 is downregulated in HCC cell lines (13). In the present study, we firstly verified this result by qRT-PCR. We found that the gene expression levels of miR-137 were lower in all 7 probed HCC cell lines, when compared with the expression level of miR-137 in THLE2, a normal human hepatocyte cell line (P<0.05, Fig. 1A). We then used a lentiviral vector to ectopically upregulate miR-137 in two HCC cell lines, HuH7 and MHCC97L. The efficiency of lentivirus-mediated miR-137 upregulation was confirmed by qRT-PCR (P<0.05, Fig. 1B).

In order to find the downstream molecular target of miR-137 in HCC, we explored several online miRNA target softwares, such as TargetScan (www.targetscan.org), microRNA (www.microRNA.org) and miTarget (www.cbit.snu.ac.kr/~miTarget) and found that CDC42 was a possible hit (Fig. 1C). We then carried out a dual-luciferase reporter assay and confirmed that in HuH7 and MHCC97L cells, CDC42 was the downstream target of miR-137 (P<0.05, Fig. 1D).

MicroRNA-137 is inversely correlated with CDC42 in both HCC tumors and cell lines

We then evaluated the correlation between miR-137 and CDC42 in both HCC tumors and cell lines. Firstly, 13 clinically obtained HCC tumor samples underwent gene expression analysis by qRT-PCR. We found that, through a linear regression method, the gene expression level of miR-137 was inversely correlated with the mRNA expression level of CDC42 in the human HCC tumors (Fig. 2A). We also examined the protein expression of CDC42 in HCC cell lines. We found that, while miR-137 was upregulated by lentiviral transfection, CDC42 protein levels were substantially downregulated in the HuH7 and MHCC97L cells (Fig. 2B). The downregulation of CDC42 by miR-137 upregulation in HCC cells was further confirmed by qRT-PCR (Fig. 2C and D; miR-C vs. miR-137, P<0.05).

After CDC42 was downregulated by miR-137 upregulation, we re-introduced CDC42, through the transection of an overexpression vector pcDNA3.1/CDC42, back into HuH7 and MHCC97L cells. In the control experiment, the HCC cells were transfected with an empty vector pcDNA3.1/+. We found that, while pcDNA3.1+ had no effect on CDC42 gene expression (Fig. 2C and D; miR-137 vs. miR-137+pcDNA3.1/+, P>0.05), pcDNA3.1/CDC42 significantly upregulated CDC42 mRNA in the HuH7 and MHCC97L cells (Fig. 2C and D; miR-137+pcDNA3.1/+ vs. miR-137+ pcDNA3.1/CDC42, P<0.05).

Inhibition of HCC proliferation by miR-137 is ameliorated by CDC42

Since we discovered that CDC42 was inversely regulated by miR-137 in HCC, we speculated that CDC42 may play a functional role in HCC. To test this hypothesis, we firstly upregulated miR-137 in the HuH7 and MHCC97L cells with an miR-137 lentivirus. Through a 5-day proliferation assay, we found that miR-137 upregulation significantly inhabited cancer growth in both the HuH7 and MHCC97L cells (P<0.05, Fig. 3A and C), in line with a previous study (13). Secondly, 24 h after lentiviral transfection to upregulate miR-137, CDC42 was overexpressed in the HuH7 and MHCC97L cells for 24 h. Another 5-day in vitro proliferation assay showed that re-introduction of CDC42 restored the growth of HuH7 and MHCC97L cells (P<0.05, Fig. 3B and D). Thus, our results showed that overexpression of CDC42 ameliorated the inhibitory effect of miR-137 on HCC proliferation.

Inhibition of HCC metastasis by miR-137 is ameliorated by CDC42

We speculated that CDC42 may play a functional role in miR-137-mediated HCC metastasis. Firstly, we confirmed the inhibitory effect of miR-137 on HCC metastasis (13). We transfected HuH7 and MHCC97L cells with a lentivirus of miR-137 or miR-C, followed by a migration assay to assess the metastasis in 24 h. The assay showed that in both HCC cell lines, the metastatic capability of the cancer cells was inhibited by miR-137 upregulation. Immunostaining showed that significantly less migrated HCC cells were noted in the lower chambers (Fig. 4A and C, miR-C vs. miR-137). Quantitative measurement showed that miR-137 upregulation reduced the percentages of migrated cells to <40% in both the HuH7 and MHCC97L cells (Fig. 4B and D; miR-C vs. miR-137, P<0.05).

Secondly, 24 h after miR-137 transfection, we carried out another transfection with either pcDNA3.1/CDC42 or pcDNA3.1/+ in the HuH7 and MHCC97L cells, followed by a migration assay to evaluate the effect of CDC42 overexpression on HCC metastasis. Immunostaining showed that, after miR-137 upregulation, re-introduction of CDC42 promoted more HCC cells to migrate into the lower chambers (Fig. 4A and C; miR-137 + pcDNA3.1/+ vs. miR-137 + pcDNA3.1/+CDC42). Furthermore, quantification demonstrated that CDC42 restored the percentages of migrated cells to ~60% of the original levels (Fig. 4B and D; miR-137 + pcDNA3.1/+ vs. miR-137 + pcDNA3.1/+CDC42, P<0.05). It is also worth noting that second transfection of the empty vector had no effect on HCC metastasis (Fig. 4B and D; miR-137 vs. miR-137 + pcDNA3.1/+).

Therefore, the results of our migration assay demonstrated that overexpression of CDC42 ameliorated the inhibitory effect of miR-137 on HCC metastasis.

Inhibition of in vivo HCC tumor growth by miR-137 is ameliorated by CDC42

We then examined the effect of the overexpression of CDC42 on miR-137-mediated inhibition of in vivo HCC tumor growth (13). Firstly, we transfected HuH7 cells with the lentivirus of miR-137 or miR-C. Twenty-four hours later, HuH7 cells were transplanted into 2-month-old null mice. The in vivo growth of tumors was monitored for 5 weeks, followed by immunostaining of Ki67 at the end of the in vivo assay. Both the in vivo assay (P<0.05, Fig. 5A) and Ki67 immunostaining (Fig. 5B) confirmed that miR-137 inhibited HCC tumor growth. Secondly, 24 h after miR-137 transfection, HuH7 cells were further transfected with either pcDNA3.1/CDC42 or pcDNA3.1/+, followed by the transplantation assay. Both in vivo tumor growth assay (P<0.05, Fig. 5C) and Ki67 immunostaining (Fig. 5D) showed that overexpression of CDC42 ameliorated the inhibitory effect of miR-137 on in vivo HCC tumor growth.

CDC42 and AKT2 are independently expressed in HCC

A previous study demonstrated that miR-137 inhibited HCC through AKT2 (13). Since we demonstrated that CDC42 was also involved in miR-137-mediated HCC inhibition, we aimed to ascertain whether CDC42 and AKT2 undergoes crosstalk in HCC. We firstly examined the effect of the overexpression of CDC42 on the mRNA expression level of AKT2 in the HuH7 and MHCC97L cells. The results of qRT-PCR showed that CDC42 did not alter the expression level of AKT2 in the HCC cells (P>0.05, Fig. 6A and B). We then transfected the HuH7 and MHCC97L cells with another set of overexpression vectors of pcDNA3.1/AKT2 and its control pcDNA3.1/+. qRT-PCR showed that, in both HuH7 and MHCC97L cells, the expression levels of AKT2 were significantly upregulated by pcDNA3.1/AKT2 (P<0.05, Fig. 6C and D), whereas expression levels of CDC42 were not changed (P>0.05, Fig. 6E and F).

AKT2 contributes additively to CDC42 in reversing the inhibitory effect of miR-137 on HCC proliferation and metastasis

We then investigated the correlation between AKT2 and CDC42 in the regulation of miR-137-induced HCC inhibition. Firstly, we studied the possible additive effect of AKT2 on HCC proliferation. We transfected HuH7 and MHCC97L cells with the miR-137 lentivirus. Twenty-four hours after that, we co-transfected the cells with pcDNA3.1/CDC42 and pcDNA3.1/AKT2. The control cells were co-transfected with pcDNA3.1/CDC42 and pcDNA3.1/+. The in vitro proliferation assay demonstrated that AKT2 overexpression, in addition to CDC42 overexpression, further rescued HCC proliferation from miR-137-induced inhibition (P<0.05, Fig. 7). Secondly, we evaluated the additive effect of the overexpression of AKT2 on HCC metastasis. Twenty-four hours after co-transfection of the double-overexpressing vectors, an in vitro migration assay was carried out. It showed that in both HCC cell lines, more cancer cells migrated into the lower chambers (Fig. 8A and C). Quantitative measurement also showed that AKT2 overexpression, in addition to CDC42 overexpression, further rescued HCC metastasis from miR-137-induced inhibition (P<0.05, Fig. 8B and D).

Discussion

It was recently reported that miR-137 is a new member of the tumor-suppressing miRNAs found in human HCC (13). Liu et al found that miR-137 was lowly expressed in HCC tumors and cell lines, and was strongly associated with survival in patients with HCC (13). In addition, forced miR-137 overexpression was able to exert inhibitory effects on HCC proliferation and migration, both in vitro and in vivo (13). In the present study, we firstly confirmed that the gene expression levels of miR-137 were low in 7 HCC lines, as compared to the expression level of miR-137 in a normal human hepatocyte cell line (THLE2). Importantly, one of the newly examined HCC cell lines in our study, MHCC97L, was specifically derived from Chinese HCC patients with low metastatic capability (24). Thus, our result showing that miR-137 was downregulated in MHCC97L cells, as in the other HCC cell lines, suggests that a low expression pattern of miR-137 may be universal regardless of ethic background.

Moreover, in our study, we used a lentivirus to ectopically upregulate miR-137 in the HuH7 and MHCC97L cells. We found that miR-137 upregulation inhibited HCC proliferation and metastasis in vitro and tumor growth in vivo (Figs. 3Figure 45), further confirming the antitumor effect of miR-137 in HCC as shown in a previous study (13). Moreover, we identified CDC42 as another downstream target gene of miR-137 in HCC. Dual-luciferase reporter and western blot assays showed that CDC42 was directly regulated by miR-137 in HCC cells. Most importantly, while we used an overexpression system to ectopically re-introduce CDC42 back into HCC cells after miR-137 upregulation, we were able to ameliorate or reverse the tumor-suppressive effects of miR-137 on HCC in vitro proliferation, migration and in vivo tumor growth. A previous study showed that CDC42 was lowly expressed in liver tumors when compared to the expression in non-tumor liver tissues (22), suggesting that CDC42 may act as an antitumor (or tumor suppressing) factor in HCC. Interestingly, another study showed that CDC42 indeed acted as an oncogene in HCC as CDC42 knockdown inhibited the migration of QGY-7703 cells (23). Although the results of our study supported the idea of CDC42 as an oncogene, caution shall be taken to draw such a conclusion as more complex signaling mechanisms may be associated with CDC42 to determine whether it is an oncogene or a tumor-suppressor in HCC.

Finally, we co-expressed CDC42 with AKT2, another known miR-137 target gene in HCC (13), in the HuH7 and MHCC97L cells. We found that AKT2 contributed additively to CDC42 to rescue the inhibition of miR-137 on HCC proliferation and migration (Figs. 7 and 8), suggesting that CDC42 and AKT2 may exert their oncogenic effects independently, although both are regulated by miR-137. Future study may help to elucidate the differential signaling pathways associated with CDC42 or AKT2 in HCC regulation.

In summary, our study revealed that CDC42 is an independent target gene of miR-137 in regulating HCC. These results may help to identify possible biomarkers and elucidate the underlying molecular mechanisms of human HCC.

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Alvarez-Garcia I and Miska EA: MicroRNA functions in animal development and human disease. Development. 132:4653–4662. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Sassen S, Miska EA and Caldas C: MicroRNA: Implications for cancer. Virchows Arch. 452:1–10. 2008. View Article : Google Scholar

5 

Takasaki S: Roles of microRNAs in cancers and development. Methods Mol Biol. 1218:375–413. 2015. View Article : Google Scholar

6 

Hung CH, Chiu YC, Chen CH and Hu TH: MicroRNAs in hepatocellular carcinoma: Carcinogenesis, progression, and therapeutic target. Biomed Res Int. 2014:4864072014. View Article : Google Scholar : PubMed/NCBI

7 

Lim L, Balakrishnan A, Huskey N, Jones KD, Jodari M, Ng R, Song G, Riordan J, Anderton B, Cheung ST, et al: MicroRNA-494 within an oncogenic microRNA megacluster regulates G1/S transition in liver tumorigenesis through suppression of mutated in colorectal cancer. Hepatology. 59:202–215. 2014. View Article : Google Scholar :

8 

Ohta K, Hoshino H, Wang J, Ono S, Iida Y, Hata K, Huang SK, Colquhoun S and Hoon DS: MicroRNA-93 activates c-Met/PI3K/Akt pathway activity in hepatocellular carcinoma by directly inhibiting PTEN and CDKN1A. Oncotarget. 6:3211–3224. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Gao B, Gao K, Li L, Huang Z and Lin L: miR-184 functions as an oncogenic regulator in hepatocellular carcinoma (HCC). Biomed Pharmacother. 68:143–148. 2014. View Article : Google Scholar

10 

Kim HS, Lee KS, Bae HJ, Eun JW, Shen Q, Park SJ, Shin WC, Yang HD, Park M, Park WS, et al: MicroRNA-31 functions as a tumor suppressor by regulating cell cycle and epithelial-mesenchymal transition regulatory proteins in liver cancer. Oncotarget. 6:8089–8102. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Bae HJ, Noh JH, Kim JK, Eun JW, Jung KH, Kim MG, Chang YG, Shen Q, Kim SJ, Park WS, et al: MicroRNA-29c functions as a tumor suppressor by direct targeting oncogenic SIRT1 in hepatocellular carcinoma. Oncogene. 33:2557–2567. 2014. View Article : Google Scholar

12 

Zhang JP, Zeng C, Xu L, Gong J, Fang JH and Zhuang SM: MicroRNA-148a suppresses the epithelial-mesenchymal transition and metastasis of hepatoma cells by targeting Met/Snail signaling. Oncogene. 33:4069–4076. 2014. View Article : Google Scholar

13 

Liu LL, Lu SX, Li M, Li LZ, Fu J, Hu W, Yang YZ, Luo RZ, Zhang CZ and Yun JP: FoxD3-regulated microRNA-137 suppresses tumour growth and metastasis in human hepatocellular carcinoma by targeting AKT2. Oncotarget. 5:5113–5124. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Luo C, Tetteh PW, Merz PR, Dickes E, Abukiwan A, Hotz-Wagenblatt A, Holland-Cunz S, Sinnberg T, Schittek B, Schadendorf D, et al: miR-137 inhibits the invasion of melanoma cells through downregulation of multiple oncogenic target genes. J Invest Dermatol. 133:768–775. 2013. View Article : Google Scholar

15 

Wright C, Turner JA, Calhoun VD and Perrone-Bizzozero N: Potential impact of miR-137 and its targets in schizophrenia. Front Genet. 4:582013. View Article : Google Scholar : PubMed/NCBI

16 

Chen Q, Chen X, Zhang M, Fan Q, Luo S and Cao X: miR-137 is frequently down-regulated in gastric cancer and is a negative regulator of Cdc42. Dig Dis Sci. 56:2009–2016. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Zhu X, Li Y, Shen H, Li H, Long L, Hui L and Xu W: miR-137 inhibits the proliferation of lung cancer cells by targeting Cdc42 and Cdk6. FEBS Lett. 587:73–81. 2013. View Article : Google Scholar

18 

Liu M, Lang N, Qiu M, Xu F, Li Q, Tang Q, Chen J, Chen X, Zhang S, Liu Z, et al: miR-137 targets Cdc42 expression, induces cell cycle G1 arrest and inhibits invasion in colorectal cancer cells. Int J Cancer. 128:1269–1279. 2011. View Article : Google Scholar

19 

Arias-Romero LE and Chernoff J: Targeting Cdc42 in cancer. Expert Opin Ther Targets. 17:1263–1273. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Stengel K and Zheng Y: Cdc42 in oncogenic transformation, invasion, and tumorigenesis. Cell Signal. 23:1415–1423. 2011. View Article : Google Scholar : PubMed/NCBI

21 

van Hengel J, D'Hooge P, Hooghe B, Wu X, Libbrecht L, De Vos R, Quondamatteo F, Klempt M, Brakebusch C and van Roy F: Continuous cell injury promotes hepatic tumori-genesis in cdc42-deficient mouse liver. Gastroenterology. 134:781–792. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Zhang Y, Takahashi S, Tasaka A, Yoshima T, Ochi H and Chayama K: Involvement of microRNA-224 in cell proliferation, migration, invasion, and anti-apoptosis in hepatocellular carcinoma. J Gastroenterol Hepatol. 28:565–575. 2013. View Article : Google Scholar

23 

Wang R, Zhao N, Li S, Fang JH, Chen MX, Yang J, Jia WH, Yuan Y and Zhuang SM: MicroRNA-195 suppresses angio-genesis 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

24 

Li Y, Tang ZY, Ye SL, Liu YK, Chen J, Xue Q, Chen J, Gao DM and Bao WH: Establishment of cell clones with different metastatic potential from the metastatic hepatocellular carcinoma cell line MHCC97. World J Gastroenterol. 7:630–636. 2001.

Related Articles

Journal Cover

November-2015
Volume 34 Issue 5

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
Gao M, Liu L, Li S, Zhang X, Chang Z and Zhang M: Inhibition of cell proliferation and metastasis of human hepatocellular carcinoma by miR-137 is regulated by CDC42. Oncol Rep 34: 2523-2532, 2015
APA
Gao, M., Liu, L., Li, S., Zhang, X., Chang, Z., & Zhang, M. (2015). Inhibition of cell proliferation and metastasis of human hepatocellular carcinoma by miR-137 is regulated by CDC42. Oncology Reports, 34, 2523-2532. https://doi.org/10.3892/or.2015.4261
MLA
Gao, M., Liu, L., Li, S., Zhang, X., Chang, Z., Zhang, M."Inhibition of cell proliferation and metastasis of human hepatocellular carcinoma by miR-137 is regulated by CDC42". Oncology Reports 34.5 (2015): 2523-2532.
Chicago
Gao, M., Liu, L., Li, S., Zhang, X., Chang, Z., Zhang, M."Inhibition of cell proliferation and metastasis of human hepatocellular carcinoma by miR-137 is regulated by CDC42". Oncology Reports 34, no. 5 (2015): 2523-2532. https://doi.org/10.3892/or.2015.4261