MicroRNA-616 promotes the migration, invasion and epithelial-mesenchymal transition of HCC by targeting PTEN

  • Authors:
    • Di Zhang
    • Peihua Zhou
    • Wei Wang
    • Xiaolong Wang
    • Junhui Li
    • Xuejun Sun
    • Li Zhang
  • View Affiliations

  • Published online on: October 16, 2015     https://doi.org/10.3892/or.2015.4334
  • Pages: 366-374
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

MicroRNAs, which can post-transcriptionally regulate gene expression by binding to the 3'-untranslated regions of the mRNAs, have been found to be the critical regulators of the development and progression of hepatocellular carcinoma (HCC). The present study demonstrated for the first time that microRNA-616 (miR-616) was markedly upregulated in HCC tissues, and was associated with the recurrence and metastasis of HCC. Elevated level of miR-616 was correlated with adverse clinicopathological features and poor prognosis of HCC patients. Gain- and loss-of-function studies revealed that miR-616 could potentiate the migration, invasion and the epithelial-mesenchymal transtion (EMT) phenotype of HCC cells. Phosphatase and tensin homolog (PTEN), the predicted target of miR-616 by bioinformatics analysis, was confirmed as a direct downstream target of miR-616 through western blotting, luciferase reporter and immunohistochemical assays. Furthermore, we demonstrated that miR-616 exerted the promoting effects on EMT and metastatic ability of HCC cells through suppressing PTEN expression. Based on these results, we conclude that miR-616 is a promising prognostic biomarker of HCC and targeting miR-616 may be a potential option to prevent the progression of HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most common human cancers worldwide and ranks third in cancer-related death (1). Although remarkable achievements have been made (2), the long-term prognosis of HCC patients remains poor, particularly for those in advanced stages, with the 5-year survival rate lower than 5% (3). Local and systemic metastasis are the critical reasons for the unsatisfactory survival of HCC patients in advanced stages (4). Elucidating the molecular mechanism of the metastasis of HCC is critical for identifying novel therapeutic targets and may significantly improve the prognosis of HCC patients.

MicroRNAs (miRNAs) are a group of short non-conding RNAs and serve as important post-transcriptional modulator of gene expression by binding to 3′ untranslated region (3′-UTR) of targeted mRNAs and resulting in the suppression of protein translation or the degradation of the target mRNAs (5,6). Numerous studies have demonstrated that miRNAs are involved in various cellular activities (79) including proliferation, apoptosis, differentiation and movement. Aberrant expression and function of miRNAs has been found to play fundamental roles in human malignancies (7,1012). In addition, miRNAs have become promising diagnostic and prognostic biomarkers and attractive therapeutic targets of human cancers (13,14).

Among numerous miRNAs, microRNA-616 (miR-616) has been found to be a novel cancer-associated miRNA. miRNA profiling of human gastric cancer showed that miR-616 was overexpressed in gastric cancer tissues (15). In addition, studies of lung (16) and prostate cancer (17) demonstrated that the level of miR-616 was significantly elevated in the serum of patients. Furthermore, miR-616 was found to play an important role in the development and maintenance of androgen-independent prostate cancer by regulating the expression of TFPI-2 (17). However, the expression and clinical significance of miR-616 in HCC tissues, and its functional role and the underlying mechanisms in HCC cells, are still undefined.

In the present study, we demonstrated that miR-616 was significantly upregulated in HCC tissues. Elevated expression of miR-616 was observed in patients with metastasis and recurrence. Additionally, elevated miR-616 expression was associated with adverse clinicopathological features and poor prognosis of HCC patients. In vitro studies showed that miR-616 promoted the invasive behavior and epithelial-mesenchymal transition (EMT) of HCC cells. Moreover, phosphatase and tensin homolog (PTEN) was identified to be the direct downstream target of miR-616 in HCC. miR-616 contributed to the metastasis and EMT of HCC by inhibiting the expression of PTEN.

Materials and methods

Human tissue specimens and cell lines

The paired HCC and adjacent non-tumor tissues were collected from 80 HCC patients who received surgical resection of primary HCC in the Department of General Surgery at the Second Affiliated Hospital of Xi'an Jiaotong University from January 2006 to December 2009. No local or systemic treatments had been performed before the surgical resection. Informed consent was obtained from each patient before collecting the clinical samples. The demographic and clinicopathological data are presented in Table I. All protocols in the present study were approved by the Xi'an Jiaotong University Ethics Committee according to the Declaration of Helsinki (as revised in Tokyo, 2004).

Table I

The association between the clinicopathological features and miR-616 expression in HCC.

Table I

The association between the clinicopathological features and miR-616 expression in HCC.

Clinicopathological featuresTotal no. of pts.No. of patients
P-value
miR-616High miR-616Low
Gender
 Male5931280.446
 Female21912
Age (years)
 <503418160.651
 ≥50462224
HBV infection
 Absent2815130.639
 Present522527
Cirrhosis
 Absent3318150.496
 Present472225
Diameter of tumor (cm)
 <53419150.366
 ≥5462125
No. of tumor nodules
 14424200.369
 >1361620
Venous infiltration
 Absent572433 0.026a
 Present23167
Edmondson-Steiner grading
 I+II602535 0.001a
 III+IV20155
TNM stage
 I+II582236 <0.001a
 III+IV22184

a Statistically significant. pts., patients.

Six HCC cell lines, Hep3B, HepG2, Huh7, MHCC97L HCCLM3 and MHCC97H, and the human immortalized normal hepatocyte cell line, LO2 (The Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China) were maintained in complete Dulbecco's modified Eagle's medium (DMEM; HyClone, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS, HyClone, Thermo Fisher Scientific, Australia) with 100 U/ml penicillin and 100 µg/ml streptomycin (Sigma, USA) in a humidified incubator containing of 5% CO2 at 37°C.

Quantitative real-time PCR (qPCR) analysis

Total RNA from tissues or cells was extracted using TRIzol (Life Technologies) according to the manufacturer's instructions. The PCR amplification for the quantification of the miR-616 was performed using the TaqMan microRNA assay kit (Applied Biosystems, Foster City, CA, USA). U6 was used as endogenous controls. The relative expression of miR-616 was normalized to that of U6.

Cell transfection

miR-616 expression vector (HmiR0186-MR04), the control vector for miR-616 (CmiR0001-MR04), miR-616 inhibitor (HmiR-AN0721-AM03) and the negative control for the miR-616 inhibitor (CmiR-AN0001-AM03), were purchased from GeneCopoeia (Guangzhou, China). Plasmids carrying wt SMAD7 (pCMV5-SMAD7) were obtained from Addgene (Cambridge, MA, USA). The vectors mentioned above were transfected into the HCC cells using Lipofectamine 2000 (Invitrogen, USA) following the manufacturer's protocol.

Migration and invasion assays

Cell migration and invasion assays were evaluated in chambers of 8-µm with Transwell inserts (Millipore, USA) according to the manufacturer's instructions. Cells (5×104) were seeded into the top chamber of each insert in serum-free medium and serum-containing medium was used in the lower chamber as the attractant. Specifically for the invasion assay, before seeding the HCC cells, each chamber was coated with mixture of DMEM and Matrigel (Becton-Dickinson Labware, USA) at a ratio of 6:1. After incubated at 37°C for 24 h, the cells adherent to the upper surface of the filter were removed using a cotton swab. The migrated or invaded cells were fixed and stained with 0.1% crystal violet, air-dried and photographed. Three independent experiments were performed.

Western blotting

HCC cells were washed twice with phosphate-buffered saline (PBS) and lysed on the culture dishes using RIPA lysis buffer (BioMed, China). Protein concentration was measured using the BCA kit (Pierce, USA) and 30 µg of each sample were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to PVDF membrane. Non-specific binding sites were blocked by incubating with TBST containing 5% skimmed milk/TBST for 2 h at room temperature. The blots were then incubated overnight at 4°C with the following primary antibodies: PTEN (1:1,000), AKT (1:1,000) (both from Santa Cruz, USA), p-AKT (1:1,000), E-cadherin (1:1,000), vimentin (1:1,000) and β-actin (1:1500) (all from Cell Signaling, USA). After washing the membranes with TBST, blots were incubated with horseradish peroxidase-conjugated goat anti-mouse or anti-rabbit secondary antibodies (1:10,000; Bio-Rad, USA), and were detected using the Bio-Rad Gel imaging system.

Luciferase reporter assay

The wild-type 3′-UTR sequence of PTEN (wt PTEN-3′-UTR) predicted to interact with miR-616 or the mutated sequence (mt PTEN-3′-UTR) within the predicted target sites was synthesized and inserted into the pGL3 control vector (Promega, USA). For the reporter assay, Hep3B cells were seeded into 24-well plates and were transfected with the above constructs and miR-616 expressing vector, miR-616 inhibitor, control vector or negative control. After 48 h, the cells were harvested and luciferase activity was measured using the Dual-Luciferase Reporter Assay system (Promega) according to the manufacturer's instructions. Results were obtained from three independent experiments performed in duplicate.

Immunohistochemical staining

After being deparaffinized in xylene and rehydrated in a graded alcohol series and distilled water, the paraffin sections were heated for antigen retrieval in sodium citrate buffer for 10 min, and were quenched for endogenous peroxidase activity in 3% hydrogen peroxide for 15 min. They were incubated with PTEN (1:100) or p-AKT (1:50) antibody at 4°C overnight. Then, sections were incubated with biotinylated secondary antibodies from Zhongshan Golden Bridge Biotechnology (Beijing, China) for 2 h. Sections were stained with the avidin-biotin-peroxidase complex (SABC) method and visualized with diaminobenzidine and counter-stained with hematoxylin.

The staining results for PTEN and p-AKT were semi-quantitatively calculated by multiplying the staining intensity and the percentage of positive staining. Staining intensity was assessed as four grades: 0, none; 1, weak; 2, moderate; and 3, strong. The percentage of positive staining was assessed as the following grades: 0, <5%; 1, 6–25%; 2, 26–50%; 3, 51–75%; and 4, >75%. Ten independent high magnification (×400) fields were assayed for each section. Two experienced pathologists independently evaluated the sections, and obtained the average scores of 10 fields.

Statistical analysis

Results are presented as mean ± SEM. The SPSS statistical package for Windows version 13 (SPSS, Inc., Chicago, IL, USA) and GraphPad Prism 5 software (GraphPad Software, Inc., USA) were employed to perform the statistical analysis. The Pearson's Chi-square test, the Spearman's rank correlation coefficient, the two-tailed Student's t-test, the Kaplan-Meier plot, the log-rank test or ANOVA was used in appropriate situation. Difference was considered to indicate a statistically significant result, at P<0.05.

Results

miR-616 is upregulated in HCC and is associated with the metastasis and recurrence of HCC

To determine the level of miR-616 expression in HCC tissues, the quantitative RT-PCR assay was performed in 40 pairs of HCC and adjacent non-tumor tissues. The expression level of miR-616 in HCC tissues was significantly elevated compared with that in the adjacent non-tumor liver tissues (P<0.01, Fig. 1A). Moreover, in the recurrent cases, the level of miR-616 was significantly higher than that in the non-recurrent cases (P<0.01, Fig. 1B). In addition, patients with metastasis had obviously increased level of miR-616 than those without metastasis (P<0.01, Fig. 1C). These results demonstrated that the expression level of miR-616 is significantly upregulated in HCC tissues and is associated with the recurrence and metastasis of HCC.

Then, we examined the expression level of miR-616 in a non-transformed hepatic cell line (LO2) and six types of HCC cell lines (Hep3B, HepG2, Huh7, MHCC97L, HCCLM3 and MHCC97H). Compared with LO2, all HCC cell lines had significantly higher expression level of miR-616 (P<0.01, Fig. 1D). Moreover, it was interesting to find that miR-616 expression in HCCLM3 and MHCC97H, two highly metastatic HCC cell lines, was obviously higher than that in the low metastatic HCC cell lines including Hep3B, HepG2, Huh7 and MHCC97L (P<0.01, Fig. 1D). These data indicated that miR-616 can probably potentiate the invasive ability of HCC cells.

Elevated expression of miR-616 was correlated with adverse clinicopathological features and poor prognosis of HCC patients

The obvious elevation of miR-616 in HCC tissues and its association with HCC recurrence and metastasis prompted us to examine the clinical significance of miR-616 in HCC. The expression level of miR-616 was assessed either low (n=40) or high (n=40) based on the cut-off value which was defined as the median level of miR-616 in the 80 patients cohort. As presented in Table II, elevated expression level of miR-616 in HCC patient was associated with the venous infiltration (P=0.026), high Edmondson-Steiner grading (P=0.001) and advanced TNM stage (P<0.001). Furthermore, Kaplan-Meier analysis was performed to evaluate the prognostic value of miR-616 in HCC. Patients with high-miR-616 expression level had significantly decreased overall survival (P<0.01, Fig. 2A) and disease-free survival (P<0.01, Fig. 2B). These data suggest miR-616 can serve as a prognostic predictor for HCC patients.

Table II

Multivariate Cox regression analysis of 5-year overall and disease-free survival.

Table II

Multivariate Cox regression analysis of 5-year overall and disease-free survival.

VariablesOverall survival
Disease-free survival
HR95% CIP-valueHR95% CIP-value
miR-6161.8081.044–3.134 0.035a1.7241.014–2.932 0.044a
Venous infiltration1.9411.012–3.725 0.046a2.1641.132–4.139 0.020a
Edmondson-Steiner grading1.7171.018–2.897 0.043a1.7301.019–2.939 0.042a
TNM stage2.2941.126–4.671 0.022a2.1161.040–4.303 0.038a

{ label (or @symbol) needed for fn[@id='tfn2-or-35-01-0366'] } HR, hazard ratio; CI, confidence interval.

a Statistically significant.

miR-616 promotes the migration, invasion and EMT of HCC cells

Next, we further explored the functional role of miR-616 in HCC cells. We transfected Hep3B cells with miR-616 mimics, and miR-616 expression level was significantly increased after transfection, as suggested by the qRT-PCR (P<0.01, Fig. 3A). Functionally, as suggested by the Transwell assay, Hep3B cells overexpressing miR-616 (Hep3B-miR-616 cells) showed increased ability of migration (P<0.01, Fig. 3B) and invasion (P<0.05, Fig. 3B). In contrast, miR-616 inhibitor significantly downregulated the expression level of miR-616 in MHCC97H cells (P<0.01, Fig. 3C), and resulted in an obviously reduced number of migrated (P<0.01, Fig. 3D) and invaded (P<0.01, Fig. 3D) MHCC97H cells. These results demonstrated that miR-616 can promote the invasive behavior of HCC cells.

Since the EMT process, which is regarded as a hallmark of metastasis (18), increased the migratory and invasive ability of HCC cells (19), we next investigated whether miR-616 could modulate EMT phenotype of HCC cells. Overexpression of miR-616 in Hep3B cells resulted in decreased level of E-cadherin (P<0.01, Fig. 3E) and increased expression of vimentin (P<0.01, Fig. 3E). In addition, inhibition of miR-616 in MHCC97H cells led to increased level of E-cadherin (P<0.01, Fig. 3E) and decreased expression of vimentin (P<0.01, Fig. 3E). These results suggested that miR-616 can promote EMT of HCC cells.

PTEN is a downstream target of miR-616 in HCC cells

To clarify the underlying mechanisms by which miR-616 regulates the invasive behavior and EMT of HCC cells, we used two publicly available databases (TargetScan 6.2 and miRanda) to search potential downstream target of miR-616. PTEN, which is a well-recognized tumor suppressor and an important regulator of HCC metastasis (20,21), was predicted as a downstream target of miR-616. As shown in Fig. 4A, the 3′-UTR of PTEN mRNA was found to contain the complementary sequence of miR-616. This suggests that miR-616 can potentially regulate the expression of PTEN by binding to the 3′-UTR of PTEN. To confirm this prediction, Hep3B cells transfected with miR-616 expression vector were subjected to western blotting. Overexpression of miR-616 in Hep3B cells resulted in significantly reduced expression of PTEN (P<0.05, Fig. 4B). It is well recognized that inhibiting PTEN expression can lead to increased phosphorylation of AKT (22). Accordingly, the phosphorylation of AKT was significantly increased after overexpressing miR-616 in Hep3B cells (P<0.05, Fig. 4B). Conversely, inhibiting the expression of miR-616 in MHCC97H cells resulted in significantly increased expression of PTEN (P<0.01, Fig. 4B) and reduced phosphorylation of AKT (P<0.01, Fig. 4B). Next, we performed the dual-luciferase reporter assay to elucidate whether miR-616 regulated PTEN expression by directly binding to its 3′-UTR. Overexpressing miR-616 significantly inhibited the luciferase activity of PTEN with a wild-type (wt) 3′-UTR (P<0.01, Fig. 4C), but had no influence on the activity of PTEN with a mutant (mt) 3′-UTR. Additionally, when anti-miR-616 was transfected, the luciferase activity of wt PTEN 3′-UTR obviously increased (P<0.01, Fig. 4C) while the luciferase activity of mt PTEN 3′-UTR remained unchanged.

To further validate the regulatory effect of miR-616 on PTEN expression, we compared the expression of PTEN and p-AKT in HCC tissues with low-miR-616 expression level with that of high-miR-616 expression. The IHC results showed that the PTEN expression in high-miR-616 expression group was significantly decreased when compared with that in the low-miR-616 expression group (P<0.01, Fig. 5A). Accordingly, the expression of p-AKT was significantly increased in high-miR-616 expression group compared to the low-miR-616 expression group (P<0.01, Fig. 5B). In all, these data demonstrated that PTEN is a direct downstream target of miR-616 and miR-616 can inhibit the expression of PTEN by binding to its 3′-UTR.

miR-616 promotes EMT and metastatic ability of HCC cells through inhibiting PTEN

To further clarify the functional significance of PTEN in miR-616-mediated EMT and metastatic ability of HCC cells, we transfected pCMV5-PTEN plasmids into Hep3B cells overexpressing miR-616 (Hep3B-miR-616 cells). The results of western blotting showed that transfection of pCMV5-PTEN plasmids resulted in significant increase of PTEN level in Hep3B-miR-616 cells (P<0.01, Fig. 6A), and led to upregulation of E-cadherin (P<0.05, Fig. 6A) and reduced expression of vimentin (P<0.05, Fig. 6A), indicating that restoring PTEN expression can reverse the EMT phenotype of Hep3B cells induced by miR-616 overexpression. Furthermore, forced expression of PTEN partly abrogated the promoting effects of miR-616 on the migration (P<0.05, Fig. 6B) and invasion (P<0.05, Fig. 6B) of HCC cells. These data indicated that miR-616 promotes EMT and metastatic ability of HCC cells by targeting PTEN.

Discussion

The initiation and progression of HCC is a complex process in which various molecules, and numerous signaling pathways are involved (23,24). Emerging studies have confirmed that abnormal expression and function of miRNAs play a critical role in this process (25,26). miR-616 is a novel cancer-associated miRNA which has been found to be abnormally expressed in gastric (15), lung (16) and prostate cancer (17,27). In the present study, we initially examined the expression status of miR-616 in clinical specimens of HCC. We confirmed for the first time that compared with adjacent non-tumor tissues, the expression level of miR-616 was significantly elevated in HCC tissues. In addition, patients with recurrence and metastasis had significantly higher miR-616 levels than those without recurrence and metastasis. The clinical analysis further demonstrated that increased expression level of miR-616 was associated with adverse clinicopathological features including venous infiltration, high Edmondson-Steiner grading and advanced TNM tumor stage. Importantly, Kaplan-Meier analysis showed that increased expression of miR-616 was associated with poorer prognosis of HCC patients including overall and disease-free survival. Moreover, the expression level of miR-616 in HCC cell lines, particularly in those with high metastatic ability (HCCLM3 and MHCC97H), was significantly higher than that in the non-transformed hepatic cell line LO2. Taken together, these results indicated that miR-616 is an important participant in the development and progression of HCC, and can serve as a prognostic marker of HCC patients.

The obvious elevation of miR-616 expression in HCC prompted us to examine the functional role of miR-616 in HCC. Since our results showed that the expression of miR-616 was associated with the recurrence and metastasis of HCC patients, we speculated that miR-616 probably influenced the migration and invasion of HCC cells. Transwell assay showed that over-expression of miR-616 promoted the migration and invasion of HCC cells while downregulation of miR-616 inhibited the metastatic behavior of HCC cells. These data suggested that miR-616 can play an active role in HCC by promoting the migration and invasion of HCC cells. EMT, a process in which epithelial cells lose cell polarity and intracellular junctions and acquire mesenchymal features (19,28), can potentiate the migratory and invasive ability of HCC cells (19,29). Increasing evidence has demonstrated that miRNAs, which could regulate the expression of EMT-related genes, played a critical role in the EMT process (28,30,31). In the present study, we found that overexpression of miR-616 reduced the expression of E-cadherin, the epithelial marker and increased the expression of vimentin, the mesenchymal marker. Downregulation of miR-616 resulted in elevated expression of E-cadherin and reduced expression of vimentin. These results suggested that miR-661 promotes the EMT of HCC cells. Altogether, miR-616 may increase the invasive ability of HCC cells by inducing EMT.

Phosphatase and tensin homolog (PTEN) is a well-recognized tumor suppressor which is frequently mutated or deleted in human cancers (22). However, instead of the mutation or the deletion of PTEN, abnormal expression of PTEN was confirmed in HCC (32) and was found to promote the progression of HCC (20). However, the molecular mechanisms responsible for the abnormal expression of PTEN remains unclear. PTEN has been found to be the downstream target of several miRNAs including miR-21 (20,33), miR-216a and 217 (21), miR-221 and 222 (34), miR29a (35) and miR-32 (36). In the present study, we confirmed that PTEN was a direct downstream target of miR-616 in HCC. Firstly, putative binding sequences of miR-616 were found in the 3′-UTR of PTEN after searching two publicly available databases (TargetScan and miRanda). Secondly, transfecting miR-616 mimics into Hep3B cells resulted in decreased expression of PTEN while downregulation of miR-616 in MHCC97H cells led to increased PTEN expression. The phosphorylation of Akt, which is inhibited by PTEN (37), was reduced after miR-616 overexpression and was increased after miR-616 suppression, supporting that miR-616 regulated the expression of PTEN. Additionally, the expression of PTEN in HCC tissues with high-miR-616 level was significantly lower than that in those with low-miR-616 level. This further validates the regulatory effect of miR-616 on PTEN expression. Furthermore, altering the expression level of miR-616 in Hep3B cells significantly influenced the luciferase activity of wt 3′-UTR of PTEN, while had no obvious influence on the luciferase activity of mt 3′-UTR of PTEN, suggesting that miR-616 can regulate the expression of PTEN by directly interacting with the 3′-UTR of PTEN.

Functionally, PTEN was able to influence the migration of glioma cells by modulating its downstream PI3K/Akt pathway (38). Therefore, we performed rescue experiments in Hep3B-miR-616 cells to clarify whether PTEN was involved in the promoting effect of miR-616 on the metastatic ability and EMT phenotype of HCC cells. We found that restoring the expression of PTEN abrogated the promoting effects of miR-616 on EMT and metastatic abilities of HCC cells. Therefore, our results suggest that PTEN is not only a direct target of miR-616 but also a functional mediator downstream of miR-616 in HCC. Of interest is that a study of prostate cancer (17) found TFPI-2 was a direct downstream target of miR-616, and miR-616 induced androgen-independent growth of prostate cancer cells by suppressing TFPI-2 expression. Therefore, the downstream targets and the functional significance of miRNAs in human cancers seem to be cancer-type specific.

In summary, our results demonstrated for the first time that miR-616 expression is elevated in HCC tissues, and its increased expression is associated with adverse prognostic features of HCC patients. Moreover, miR-616 is a valuable biomarker in predicting the prognosis of HCC patients. In vitro experiments demonstrated that miR-616 can promote the migration, invasion and EMT of HCC cells. Mechanistically, we found that PTEN is a downstream target of miR-616 in HCC and miR-616 exerts its functional significance on HCC cells by suppressing PTEN. Taken together, the findings from the present study indicated that miR-616 can serve as a valuable clinical biomarker, and may potentially be a promising therapeutic target in HCC.

References

1 

Bosch FX, Ribes J, Díaz M and Cléries R: Primary liver cancer: Worldwide incidence and trends. Gastroenterology. 127(Suppl 1): S5–S16. 2004. View Article : Google Scholar : PubMed/NCBI

2 

Karaman B, Battal B, Sari S and Verim S: Hepatocellular carcinoma review: Current treatment, and evidence-based medicine. World J Gastroenterol. 20:18059–18060. 2014.PubMed/NCBI

3 

El-Serag HB and Rudolph KL: Hepatocellular carcinoma: Epidemiology and molecular carcinogenesis. Gastro-enterology. 132:2557–2576. 2007. View Article : Google Scholar

4 

Roberts LR: Sorafenib in liver cancer - just the beginning. N Engl J Med. 359:420–422. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Rosa A and Brivanlou AH: MicroRNAs in early vertebrate development. Cell Cycle. 8:3513–3520. 2009. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Chang RM, Yang H, Fang F, Xu JF and Yang LY: MicroRNA-331-3p promotes proliferation and metastasis of hepatocellular carcinoma by targeting PH domain and leucine-rich repeat protein phosphatase. Hepatology. 60:1251–1263. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Budhu A, Jia HL, Forgues M, Liu CG, Goldstein D, Lam A, Zanetti KA, Ye QH, Qin LX, Croce CM, et al: Identification of metastasis-related microRNAs in hepatocellular carcinoma. Hepatology. 47:897–907. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Garzon R, Calin GA and Croce CM: MicroRNAs in Cancer. Annu Rev Med. 60:167–179. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Jansson MD and Lund AH: MicroRNA and cancer. Mol Oncol. 6:590–610. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Ladeiro Y, Couchy G, Balabaud C, Bioulac-Sage P, Pelletier L, Rebouissou S and Zucman-Rossi J: MicroRNA profiling in hepatocellular tumors is associated with clinical features and oncogene/tumor suppressor gene mutations. Hepatology. 47:1955–1963. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Giordano S and Columbano A: MicroRNAs: New tools for diagnosis, prognosis, and therapy in hepatocellular carcinoma? Hepatology. 57:840–847. 2013. View Article : Google Scholar

14 

Cho WC: MicroRNAs: Potential biomarkers for cancer diagnosis, prognosis and targets for therapy. Int J Biochem Cell Biol. 42:1273–1281. 2010. View Article : Google Scholar

15 

Yao Y, Suo AL, Li ZF, Liu LY, Tian T, Ni L, Zhang WG, Nan KJ, Song TS and Huang C: MicroRNA profiling of human gastric cancer. Mol Med Rep. 2:963–970. 2009.PubMed/NCBI

16 

Rani S, Gately K, Crown J, O'Byrne K and O'Driscoll L: Global analysis of serum microRNAs as potential biomarkers for lung adenocarcinoma. Cancer Biol Ther. 14:1104–1112. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Ma S, Chan YP, Kwan PS, Lee TK, Yan M, Tang KH, Ling MT, Vielkind JR, Guan XY and Chan KW: MicroRNA-616 induces androgen-independent growth of prostate cancer cells by suppressing expression of tissue factor pathway inhibitor TFPI-2. Cancer Res. 71:583–592. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Książkiewicz M, Markiewicz A and Zaczek AJ: Epithelial-mesenchymal transition: A hallmark in metastasis formation linking circulating tumor cells and cancer stem cells. Pathobiology. 79:195–208. 2012. View Article : Google Scholar

19 

Reichl P, Haider C, Grubinger M and Mikulits W: TGF-β in epithelial to mesenchymal transition and metastasis of liver carcinoma. Curr Pharm Des. 18:4135–4147. 2012. View Article : Google Scholar

20 

Meng F, Henson R, Wehbe-Janek H, Ghoshal K, Jacob ST and Patel T: MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology. 133:647–658. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Xia H, Ooi LL and Hui KM: MicroRNA-216a/217-induced epithelial-mesenchymal transition targets PTEN and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology. 58:629–641. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Song MS, Salmena L and Pandolfi PP: The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol. 13:283–296. 2012.PubMed/NCBI

23 

Roberts LR and Gores GJ: Hepatocellular carcinoma: Molecular pathways and new therapeutic targets. Semin Liver Dis. 25:212–225. 2005. View Article : Google Scholar : PubMed/NCBI

24 

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

25 

Jiang J, Gusev Y, Aderca I, Mettler TA, Nagorney DM, Brackett DJ, Roberts LR and Schmittgen TD: Association of microRNA expression in hepatocellular carcinomas with hepatitis infection, cirrhosis, and patient survival. Clin Cancer Res. 14:419–427. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Murakami Y, Yasuda T, Saigo K, Urashima T, Toyoda H, Okanoue T and Shimotohno K: Comprehensive analysis of microRNA expression patterns in hepatocellular carcinoma and non-tumorous tissues. Oncogene. 25:2537–2545. 2006. View Article : Google Scholar

27 

Haldrup C, Kosaka N, Ochiya T, Borre M, Høyer S, Orntoft TF and Sorensen KD: Profiling of circulating microRNAs for prostate cancer biomarker discovery. Drug Deliv Transl Res. 4:19–30. 2014. View Article : Google Scholar

28 

Thiery JP and Sleeman JP: Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 7:131–142. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Choi SS and Diehl AM: Epithelial-to-mesenchymal transitions in the liver. Hepatology. 50:2007–2013. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Bullock MD, Sayan AE, Packham GK and Mirnezami AH: MicroRNAs: Critical regulators of epithelial to mesenchymal (EMT) and mesenchymal to epithelial transition (MET) in cancer progression. Biol Cell. 104:3–12. 2012. View Article : Google Scholar

31 

Lee JM, Dedhar S, Kalluri R and Thompson EW: The epithelial-mesenchymal transition: New insights in signaling, development, and disease. J Cell Biol. 172:973–981. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Wan XW, Jiang M, Cao HF, He YQ, Liu SQ, Qiu XH, Wu MC and Wang HY: The alteration of PTEN tumor suppressor expression and its association with the histopathological features of human primary hepatocellular carcinoma. J Cancer Res Clin Oncol. 129:100–106. 2003.PubMed/NCBI

33 

Bao L, Yan Y, Xu C, Ji W, Shen S, Xu G, Zeng Y, Sun B, Qian H, Chen L, et al: MicroRNA-21 suppresses PTEN and hSulf-1 expression and promotes hepatocellular carcinoma progression through AKT/ERK pathways. Cancer Lett. 337:226–236. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Garofalo M, Di Leva G, Romano G, Nuovo G, Suh SS, Ngankeu A, Taccioli C, Pichiorri F, Alder H, Secchiero P, et al: miR-221&222 regulate TRAIL resistance and enhance tumorigenicity through PTEN and TIMP3 downregulation. Cancer Cell. 16:498–509. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Kong G, Zhang J, Zhang S, Shan C, Ye L and Zhang X: Upregulated microRNA-29a by hepatitis B virus X protein enhances hepatoma cell migration by targeting PTEN in cell culture model. PLoS One. 6:e195182011. View Article : Google Scholar : PubMed/NCBI

36 

Wu W, Yang J, Feng X, Wang H, Ye S, Yang P, Tan W, Wei G and Zhou Y: MicroRNA-32 (miR-32) regulates phosphatase and tensin homologue (PTEN) expression and promotes growth, migration, and invasion in colorectal carcinoma cells. Mol Cancer. 12:302013. View Article : Google Scholar : PubMed/NCBI

37 

Hers I, Vincent EE and Tavaré JM: Akt signalling in health and disease. Cell Signal. 23:1515–1527. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Dasari VR, Kaur K, Velpula KK, Gujrati M, Fassett D, Klopfenstein JD, Dinh DH and Rao JS: Upregulation of PTEN in glioma cells by cord blood mesenchymal stem cells inhibits migration via downregulation of the PI3K/Akt pathway. PLoS One. 5:e103502010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 35 Issue 1

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
Zhang D, Zhou P, Wang W, Wang X, Li J, Sun X and Zhang L: MicroRNA-616 promotes the migration, invasion and epithelial-mesenchymal transition of HCC by targeting PTEN. Oncol Rep 35: 366-374, 2016
APA
Zhang, D., Zhou, P., Wang, W., Wang, X., Li, J., Sun, X., & Zhang, L. (2016). MicroRNA-616 promotes the migration, invasion and epithelial-mesenchymal transition of HCC by targeting PTEN. Oncology Reports, 35, 366-374. https://doi.org/10.3892/or.2015.4334
MLA
Zhang, D., Zhou, P., Wang, W., Wang, X., Li, J., Sun, X., Zhang, L."MicroRNA-616 promotes the migration, invasion and epithelial-mesenchymal transition of HCC by targeting PTEN". Oncology Reports 35.1 (2016): 366-374.
Chicago
Zhang, D., Zhou, P., Wang, W., Wang, X., Li, J., Sun, X., Zhang, L."MicroRNA-616 promotes the migration, invasion and epithelial-mesenchymal transition of HCC by targeting PTEN". Oncology Reports 35, no. 1 (2016): 366-374. https://doi.org/10.3892/or.2015.4334