Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01.12.2018 | Research

RETRACTED ARTICLE: Integrin-β5, a miR-185-targeted gene, promotes hepatocellular carcinoma tumorigenesis by regulating β-catenin stability

verfasst von: Zhikun Lin, Ruiping He, Haifeng Luo, Chang Lu, Zhen Ning, Yuanhang Wu, Chuanchun Han, Guang Tan, Zhongyu Wang

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2018

Abstract

Background

The tumour microenvironment is essential for cancer progress and metastasis. Integrin-β5 (ITGB5), a member of the integrin family, has been implicated to mediate the interactions of cells with the extracellular matrix (ECM) and promote tumorigenesis in several malignancies. However, the role of ITGB5 in hepatocellular carcinoma (HCC) is still unknown.

Methods

The biological function of ITGB5 in HCC was investigated using migration, colony formation assays. The potential molecular mechanism of ITGB5 in regulating HCC tumorigenesis and β-catenin stabilization was investigated by western blotting, co-immunoprecipitation and ubiquitination assays. The expression level of ITGB5 mediated by miR-185 was confirmed by bioinformatic analysis, luciferase assay. The clinical significance of ITGB5 was based on human tissue microarray (TMA) analysis.

Results

Here, we found that the expression of ITGB5 is increased in HCC tissues. Elevated ITGB5 markedly facilitates HCC cell migration and tumorigenesis in vitro and in vivo. Further mechanistic studies revealed that ITGB5, as a partner of β-catenin, directly interacts with β-catenin and inhibits its degradation, thus leading to WNT/β-catenin activity. Subsequently, we also found that ITGB5 is a direct targeted gene of miR-185. The downregulation of miR-185 in HCC cells promotes an increase in ITGB5. An additional increase of ITGB5 is associated with β-catenin upregulation and a miR-185 decrease in HCC tissues.

Conclusions

Our data reveal that the miR-185-ITGB5-β-catenin pathway plays an important role in HCC tumorigenesis, and ITGB5 may be a promising specific target for HCC therapy.

Background

Hepatocellular carcinoma(HCC) accounts for most (85 to 90%) of the primary liver cancers and ranks as the second leading cause of cancer-related death in men. Globally, approximately 50% of the cases occur in China [13]. Historically, the high mortality rate of HCC is due to the advanced stage and frequent recurrence after surgical resection, and the majority of the patients are virtually limited to surgical treatment or chemotherapy [46]. Therefore, an in-depth study of the molecular pathogenesis could contribute by providing reliable predictors of HCC and a basis for effective therapeutic strategies, which is a promising approach to improve the survival rate [7].
Integrins are heterodimeric cell surface receptors consisting of 18 α-subunits and 8 β-subunits regulating diverse biological functions in cancers, including proliferation, adhesion, migration and invasion. Multifarious integrins involved in the disease progression and their correlation to oncogenes make them attractive targets for tumour therapy [8]. ITGB5,which associates only with ITGαV [9], has been linked to a variety of pathological conditions by several studies. ITGB5 enhances cell survival through cooperating with endothelial growth factor receptor 2(VEGF2) to inhibit extrinsic apoptosis [1012]. The contribution of ITGB5 to angiogenesis has been well established [1315]. Accumulating evidence supports the pivotal role of ITGB5 in facilitating cancer cell migration, invasion and transforming growth factor β (TGF-β)-induced epithelial-mesenchymal transition(EMT) [1619]. In 2015, a remarkable publication demonstrated that exosomal ITGB5 governed liver tropism, which was associated with liver metastasis in several malignancies, including colorectal cancer, pancreatic cancer and gastric cancer [20]. However, the role of ITGB5 in HCC is unknown.
The WNT/β-catenin signalling pathway mediates various cell biological functions in widely diverse cancer processes [2123]. β-catenin is a component of the cell surface cadherin protein complex, whose stabilization is indispensable for the activation of the WNT/β-catenin pathway [22]. A large body of evidence has indicated that alterations in β-catenin activity are correlated to the development of HCC, including the proliferation, migration and differentiation [23]. The association of integrin with β-catenin has also been published in relation to melanoma and colorectal cancer [24, 25]. Meanwhile, the interplay between ITGB5 and β-catenin can still not be elucidated in HCC.
The present study provides evidence that ITGB5 potently facilitates the tumorigenesis and progression of HCC cells in vitro and in vivo. For the first time, we focused on the relationship between ITGB5 and β-catenin in HCC, revealing that the expression of ITGB5 positively correlated with the stability of β-catenin leading to an increase in β-catenin. In addition, our findings implied that miR-185 participated in the mediation of the cell growth and migration in HCC by directly regulating ITGB5. Therefore, our data indicated that miR-185-ITGB5-β-catenin pathway may play an important role in HCC tumorigenesis.

Methods

Cell culture and reagents

The human HCC cell line Huh-7 was obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China), and the MHCC-97 Lcell line was obtained from Zhongshan Hospital, Fudan University (Shanghai, China). These cell lines were grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% foetal bovine serum (FBS), 1% penicillin, streptomycin and 0.1% Savelt ™. They were maintained in a humidified atmosphere with 5% CO2 at 37 °C. The following antibodies and reagents were used: the antibody to ITGB5 (1:500, Abcam, #ab15459), beta-catenin (1:1000, Proteintech Group, #51067-2-AP), beta-actin (1:1000, Proteintech Group, #60008-1-Ig), ubiquitin (1:1000,Cell Signalling Technology,#3393), Cyclin D1 (1:500, Cell Signaling Technology,#2978), c-Myc (1:500, Cell Signaling Technology,#13,987), MG132 (Calbiochem), puromycin (Mediatech), cycloheximide (CHX) (Inalco Spa Milano Italy), Savelt™ (Hanbio Co.LTD 1:1000) and polybrene (Sigma).

Western blot analysis and co-immunoprecipitation

For western blot analysis, cells were lysed with RIPA buffer (150 mM NaCl, 0.5% sodium deoxycholate, 0.1% SDS, 1% NP40, 1 mM EDTA, and 50 mM Tris (pH = 8.0)). After boiling with loading buffer for 15 min, the supernatants were loaded onto SDS–PAGE gels. Nitrocellulose membranes (Millipore) were incubated with primary antibodies, and then, they were incubated and visualized by HRP conjugate.
For immunoprecipitation (IP) experiments, cultured cells were washed in cold PBS and lysed in lysis buffer (20 mM Tris pH = 7.4, 0.15 M NaCl, 1 mM EDTA, 1 mM EGTA, 1% TritonX-100, 0.0025 MNa2P2O7, 1 mM β-glycerol phosphate, 1 mM Na3VO4, and 1 mM NaF) with an EDTA-free protease inhibitor mixture (Roche Applied Science) and PMSF. Cell lysates were then incubated on ice for 10 min, centrifuged, and precleared by incubation with protein A/G beads (Roche) for 1 h at 4 °C. For IP bead preparation, 30 μl of protein A/G beads was incubated with 5 μg of antibodies for 2 h at 4 °C. IPs were carried out overnight at 4 °C, and the beads were centrifuged, washed and subsequently boiled for 5 min in SDS protein loading buffer to elute bound protein. IP lysates were subjected to Western blotting with the indicated antibodies.

Colony formation assays

Huh-7 and MHCC-97 L cells with the treatment as indicated (1 × 103 cells per well) were plated into 6 well plates and cultured at 37 °C equipped with 5% CO2. Cells were fed with fresh growth medium every 3 days. Colonies were allowed to form for 2 weeks and were fixed with 4% paraformaldehyde, stained with crystal violet, washed with water to remove excess stain, and counted using Image J software. Each experiment was repeated three times.

Transwell assays

The transwell system was used for the determination of cell migration. Cells were seeded in serum-free medium in chambers (8.00 mm pores, BD, Biosciences), and then, they were allowed to migrate across the uncoated inserts using serum-containing medium for 24 h. Cells on the apical surface of the insert were scraped off, and the membranes invaded with cells were fixed with 1% paraformaldehyde and stained with crystal violet. Cell counts are expressed as the average number of cells per field of view. Three independent experiments were performed.

RNA interference and virus infection

RNA interference was performed as previously described [26, 27]. The shRNA was purchased from Sigma. The various targeting sequences were as follows:ITGB5-1,5-CCCGCTATGAAATGGCTTCAA-3; ITGB5-2, 5-GCATCCAACCAGATGGACTAT-3;and β-catenin, 5-AGGTGCTATCTGTCTGCTCTA-3. The primers 5′- gcGAATTCATGCCGCGGGCCCCGGCG.
-3′ and 5′- gcGGATCCTCAGTCCACAGTGCCATT-3′ were used to generate plasmids encoding full-length ITGB5. ITGB5 cDNA was purchased from Genecopoeia,Inc. (A8639). To generate lentivirus expressing ITGB5, HEK 293 T cells grown on a 6 cm dish were transfected with 2 μg pCDH-ITGB5 or control vector (pCDH), 1.5 μg psPax2, and 0.5 μg pMD2G. 24 h after the transfection, cells were cultured with DMEM containing 10% FBS for an additional 24 h. The culture medium containing lentiviral particles was centrifuged at 1000 g for 5 min. Viral particles collected in the supernatant were used for infection. In order to establish the stable cell line, the puromycin was used as a selection marker for the infected cells. The expression efficiency was evaluated by western blot analysis.

Quantitative real time polymerase chain reaction assay (Q-PCR)

The total RNA was isolated using Trizol (Invitrogen). One microgram of total RNA was used to synthesize cDNA using the PrimeScript™ RT reagent kit (Takara, RR047A) according to the manufacturer’s instructions. The primers were as follows:ITGB5up: 5-GTCTGCTAATCCACCCAAAATG-3,dn:5- TCTCTATCTCACCTCCACAGC-3;and actin up: 5-GACCTGACTGACTACCTCATGAAGAT-3, dn: 5-GTCACACTTCATGATGGAGTTGAAGG-3.The primers for mature miR-185 were purchased from Takara.

Introduction of microRNA mimics and inhibitors

Mimics and inhibitors of miRNA-185 were synthesized by the Genepharma Company (Shanghai, People’s Republic of China). For each transfection in a six-well plate, 100 nM miRNA mimics, mimic control or inhibitor, or inhibitor control were used. The transfection of melanoma cells by oligofectamine (Invitrogen) was performed according to the manufacturer’s instructions.

Protein stability assay and in vivo ubiquitination assay

To assess protein stability, cells were treated with cycloheximide (CHX) (20 mmol/L) for the indicated times(2,4, or 6 h). Cell extracts were prepared, and the expression levels of the indicated proteins were detected with western blotting. The ubiquitination assay was performed as previously described [28, 29].

Promoter reporters and dual-luciferase assay

MHCC-97 L and Huh-7 cells were transfected with the TOP-FLASH and FOP-FLASH reporter plasmids together with pRL-TK. Luciferase activity was measured in a 1.5-ml Eppendorf tube with the Promega Dual-Luciferases Reporter Assay kit (Promega E1980) according to manufacturer’s protocols after transfection. Relative renilla luciferase activity was normalized to firefly luciferase activity. The assay was performed as previously described [2931].

Tissue microarray, immunohistochemistry (IHC) and histological studies

The HCC tissue microarrays containing 61 HCC tissues and 35 normal tissues were purchased from Shanghai Outdo Biotech (Shanghai, China). The characteristics of the patients and their tumors were collected through review of medical records and pathologic reports. Informed consent with approval of the ethics committee of Taizhou Hospital of Zhejiang Province was obtained. All of the methods in this study were in accordance with the approved guidelines. Sections were stained with Masson’s trichrome and H&E (haematoxylin and eosin) for histopathological examination. For immunohistochemistry, sections were subjected to antigen retrieval using microwave heating at 95 °C in citrate buffer (pH = 6.0, for β-catenin) or boiling in Tris-EDTA buffer(pH = 9.0, for ITGB5). The indicated antibodies specific forITGB5 (1:100, abcam, #ab15459) and β-catenin (1:50, Proteintech Group, #51067-2-AP) were diluted according to the manufacturer’s instructions.
The degrees of immunostaining were reviewed and scored by two independent observers. The proportion of the stained cells and the extent of the staining were used as criteria of evaluation. For each case, at least 1000 tumor cells were analyzed. For each sample, the proportion of ITGB5 and β-catenin -expressing cells varied from 0 to 100%, and the intensity of staining varied from weak to strong. One score was given according to the percentage of positive cells as:< 5% of the cells:1 point; 6–35% of the cells:2 point; 36–70% of the cells:3 point; > 70% of the cells: 4 point. Another score was given according to the intensity of staining as: negative staining: 1 point; weak staining (light yellow): 2 point; moderate staining (yellowish brown): 3 point; and strong staining (brown): 4 point. A final score was then calculated by multiple the above two scores. If the final score was equal or bigger than four, the protein expression in the tumor was considered high; otherwise, the protein expression in the tumor was considered low. The high expression of miR-185 in HCC tissues was defined to more than 2 fold higher than in normal tissues. On the contrary, we defined it low expression.

Animal experiments

Animal studies were carried out in accordance with the National Institute of Health’s Guide for the Care and Use of Laboratory Animals, with the approval of the Animal Research Committee of Dalian Medical University. Male nude mice (4–6 weeks old, 18–20 g) were obtained from the SPF Laboratory Animal Centre of Dalian Medical University (Dalian, China). The mice were used for experiments after they had been acclimatized for 1 week. Stable MHCC-97 L cells (1 × 107) that were suspended in 200 μl of PBS were subcutaneously inoculated in mice. Five mice (n = 5) was used in each of the experiments. After 5 weeks, all animals were killed by cervical decapitation, the tumour weights were measured and the tumour tissues were excised aseptically. The protocol was approved by the Animal Care and Ethics Committee of Dalian Medical University.

Mass spectrometry

Immunoprecipitates were resolved on 12% SDS page. Gels were minimally stained with Coomassie Brilliant Blue to differentiate IgG bands. Each lane was then cut into three molecular weight regions. These bands were digested with 100 ng of trypsin overnight. LTQ Orbitrap Velos (Thermo Fisher Scientific) was run in a data-dependent mode, where each sample was eluted in 5–30% acetonitrile gradient. Spectral data were then searched against the human protein RefSeq database in Proteome Discoverer1.3 Suites with Mascot software.

Statistical analysis

All results are shown as the mean ± S.D. of multiple independent experiments, not technical replicates. Detailed P values for each panel in the figures are stated in the corresponding legends. A Student’s t-test, a Mann-Whitney test (for two group comparisons) or a Kruskal-Wallis one-way ANOVA followed by Dunn’s multiple comparison tests (for more than two group comparisons) was used for statistical analyses. All statistical analyses were performed with GraphPad Prism 5 and SPSS 19.0 software. All statistical tests were two-sided, and P values < 0.05 were considered to be statistically significant.

Results

ITGB5 promotes HCC tumorigenesis

To investigate the role of ITGB5 in HCC tumorigenesis, we used Huh-7 and MHCC-97 L cell lines and knocked down ITGB5 expression with 2 independent shRNAs. As shown in Fig. 1a, stable cell lines expressing these shRNAs showed significantly reduced ITGB5 levels and the percent of ITGB5 knockdown was nearly 70% in Huh-7 and 90% in MHCC-97 L. In the subsequent colony formation assays, the numbers of colonies formed by Huh-7 and MHCC-97 L cells with ITGB5 knockdown were remarkably reduced compared to control cells (Fig. 1b and c). Furthermore, in transwell assays, the migratory capabilities of Huh-7 and MHCC-97 L cells depleted of ITGB5 were also apparently inhibited (Fig. 1d, e, and f). We then turn to the opposite approach to investigate the effects of ITGB5 overexpression on HCC cells and generated Huh-7 cells stably overexpressing ITGB5 (Fig. 1g). Consistent with data from knockdown studies, Huh-7 cells overexpressing ITGB5 showed increased colony numbers in colony formation assays and enhanced migratory ability in transwell assays (Fig. 1h–k). To examine the involvement of ITGB5 expression in HCC tumorigenesis in vivo, we implanted MHCC-97 L cells stably expressing control shRNA or shRNA targeting ITGB5 into nude mice. As illustrated in Fig. 1l and m, the size and weight of xenograft tumours were significantly reduced by ITGB5 knockdown. Tumour tissues dissected were also subjected to immunohistochemistry analysis that confirmed the efficiency of ITGB5 depletion (Fig. 1n).

High expression of ITGB5 in HCC

To reveal the clinical relevance of ITGB5 expression in HCC, we collected 61 HCC specimens and 35 normal tissue samples adjacent to tumours. Through immunohistochemistry assays, we examined the expression levels of ITGB5 in these samples. Results showed that ITGB5 was significantly overexpressed in tumour tissues compared to control normal samples (Fig. 2b), and representative immunohistochemistry data are shown in Fig. 2a. The associations of ITGB5 expression with the age, gender, tumour size, and clinical stage of the patients were also analysed. Positive correlations of ITGB5 expression to patient age, tumour size, and the clinical stage were observed, which showed that ITGB5 was significantly overexpressed in patients with ages of over 50, with tumour sizes of greater than 5 cm, and those classified as in advanced stages (Fig. 2c–f).

ITGB5 interacts with β-catenin and enhances its stability

To gain more mechanistic insights into the ITGB5 regulation of HCC tumorigenesis, we carried out mass spectrometry analysis to study the partners interacting with ITGB5 and identified β-catenin as a top candidate (Additional file 1: Figure S1A). To validate the interaction between β-catenin and ITGB5, we immunoprecipitated endogenous β-catenin and ITGB5 separately from Huh-7 and MHCC-97 L cells. In both HCC cell lines, β-catenin co-immunoprecipitated with ITGB5 and vice versa (Fig. 3a and b). Interestingly, when we knocked down ITGB5 expression in Huh-7 and MHCC-97 L cells, the protein levels of β-catenin were significantly reduced, although its mRNA levels remained stable (Fig. 3c–f). On the contrary, the overexpression of ITGB5 in MHCC-97 L cells led to an upregulation of β-catenin without changing its mRNA levels (Fig. 3g and h). These findings collectively suggest that ITGB5 regulates β-catenin levels at the post-transcriptional level. Therefore, we speculated that ITGB5 might affect the degradation of β-catenin protein. We treated Huh-7 and MHCC-97 L cells with the proteasome inhibitor MG132 to stop proteasomal protein degradation and observed the protein levels of β-catenin in these cells expressing control or ITGB5-targeting shRNAs. As shown in Fig. 3i and j, MG132 treatment efficiently rescued the protein levels of β-catenin in Huh-7 and MHCC-97 L cells depleted of ITGB5. Furthermore, we treated MHCC-97 L cells stably expressing control or ITGB5-targeting shRNAs with cycloheximide to stop protein synthesis and then examined the turnover of β-catenin. It appeared that β-catenin was degraded at a much faster rate in cells with ITGB5 knockdown than in control shRNA-expressing cells (Fig. 3k). Meanwhile, the overexpression of ITGB5 in MHCC-97 L cells stabilized the protein levels of β-catenin under cycloheximide treatment conditions (Fig. 3l). Since the proteasomal degradation of cellular proteins is frequently mediated by ubiquitination, we measured the ubiquitination status of β-catenin in cells with ITGB5 knockdown or overexpression. As shown in Fig. 3m, a greatly enhanced ubiquitin signal was detected β-catenin immunoprecipitated from ITGB5 knockdown cells compared to that from control-treated cells. We also immunoprecipitated β-catenin from Huh-7 and MHCC-97 L cells with or without ITGB5 overexpression. Consistent with data from knockdown cells, in both HCC cell lines, the overexpression of ITGB5 greatly diminished the ubiquitination of β-catenin (Fig. 3n).

ITGB5 promotes HCC tumorigenesis via regulating β-catenin

To understand the functional role of this ITGB5-β-catenin interaction, we first evaluated the effect of ITGB5 on the transcriptional activity of b-catenin/TCF/LEF by using a TCF/LEF-responsive luciferase (TOP-FLASH) and non-responsive (FOP-FLASH) reporter. We found that overexpression of ITGB5 induced an approximately three-fold higher transcription from the b-catenin/TCF/LEF-dependent promoter (TOP-FLASH) and had little effect on FOP-FLASH (Fig. 4a). Whereas, inhibition of ITGB5 notably decreased the b-catenin/TCF/LEF-dependent promoter activity (Fig. 4b). Next we analyzed the expression of the downstream target genes of β-catenin and found overexpression of ITGB5 elevated the protein and mRNA levels of Cyclin D1 and c-Myc (Fig. 4c and d). Conversely, depletion of ITGB5 reduced the expression of Cyclin D1 and c-Myc (Fig. 4e and f).
β-catenin has been indicted to play an important role in tumorigenesis. To investigate whether the effects of ITGB5 on HCC tumorigenesis were relied on the regulation of β-catenin. We stably overexpressed β-catenin in MHCC-97 L cells expressing control or ITGB5 targeting shRNAs. As shown in Fig. 4g, exogenous overexpression elevated β-catenin levels in both control MHCC-97 L cells and those depleted of ITGB5. Colony formation assays were performed using these β-catenin overexpressing cells, with results showing increased colony numbers with β-catenin overexpression despite of ITGB5 knockdown (Fig. 4h and i). Additionally, in transwell assays, β-catenin overexpression also rescued the defect of cell migration caused by ITGB5 depletion (Fig. 4j and k). We then used these β-catenin overexpressing cells together with corresponding parental ones to grow xenograft tumors in nude mice. Relative to MHCC-97 L control and ITGB5 knockdown cells, β-catenin overexpression led to significantly increased size and weight of tumors formed by MHCC-97 L cells with or without ITGB5 knockdown (Fig. 4l and m).

miR-185 regulates ITGB5 expression

Having identified β-catenin as the downstream target of ITGB5 in HCC tumorigenesis, we sought to investigate upstream regulators of ITGB5 expression. Through combined analyses from web-based miRNA resources (miRanda and TargetScan), we identified 6 top candidate miRNAs that might regulate ITGB5 expression, including miR-122, miR-486-5p, miR-139, miR-340, miR-271, and miR-185 (Fig. 5a). To examine whether these candidates regulate the expression of ITGB5, we constructed the 3′UTR region of ITGB5 onto the luciferase reporter system and carried out luciferase assays. The results revealed that both miR-271 and miR-185 suppressed the expression of luciferase, with miR-185 showing the strongest inhibitory effect (Fig. 5b). To examine the influence of miR-271 and miR-185 on endogenous ITGB5 levels, we treated Huh-7 cells with the corresponding miRNA mimics. Both mimics repressed the expression of ITGB5 protein (Fig. 5c). As miR-185 showed the strongest inhibition on ITGB5 levels, we picked this miRNA for further studies. Consistently, in MHCC-97 L cells, the miR-185 mimic also efficiently suppressed ITGB5 levels (Fig. 5d). Furthermore, treatment with the miR-185 inhibitor resulted in elevated ITGB5 expression in both Huh-7 and MHCC-97 L cells (Fig. 5e and f).
We then investigated the binding site of miR-185 to the 3′UTR region of ITGB5. In doing this, we split the 3′UTR region of ITGB5 into two fragments (0–500 and 500–1166) and constructed these fragments together with the full length region (0–1166) onto pSICHEK2 vectors (Fig. 5g). Luciferase activity assays were performed using these constructs with or without miR-185 mimic treatment. The results showed that miR-185 mimic significantly inhibited the luciferase activities in the full length and 0–500 fragment groups, but the activity was not altered in the 500–1166 fragment group (Fig. 5h). These observations suggest that the binding site of miR-185 to the 3′UTR region of ITGB5 is located within the sequence 0–500 of the ITGB5 3′UTR region. By the comparison of sequence similarity between miR-185 and the sequence 0–500 of the ITGB5 3′UTR region, we speculated the binding site of miR-185 and generated the corresponding binding mutant for miR-185 on the pSICHEK2-luciferase construct (Fig. 5i). In subsequent luciferase activity assays, the binding mutations conferred this construct resistance to miR-185 mimic-mediated inhibition of the luciferase activity (Fig. 5j and k). On the contary, inhibition of endogenus miR-185 elevated the luciferase activity (Fig. 5l).

miR-185 regulates β-catenin through ITGB5

We next examined whether miR-185 regulates the levels of β-catenin. When Huh-7 and MHCC-97 L cells were treated with the miR-185 mimic, a concurrent decrease of ITGB5, β-catenin protein levels and the transcript activity of β-catenin was observed (Fig. 6a–c). In these assays, the miR-185 mimic dramatically upregulated the levels of miR-185 (Additional file 1: Figure S1B). Furthermore, treatment with the miR-185 inhibitor in Huh-7 and MHCC-97 L cells increased expression of both ITGB5 and β-catenin, leading to the transcription activity of β-catenin upregulation. However, when ITGB5 expression was silenced, the miR-185 inhibitor failed to increase the levels of β-catenin in Huh-7 and MHCC-97 L cells, suggesting an ITGB5-dependent mechanism of the miR-185 regulation of β-catenin expression (Fig. 6d, e, and f). In these assays, the miR-185 inhibitor also efficiently suppressed the expression of miR-185 (Additional file 1: Figures S1C and S1D). In the following colony formation assays, the miR-185 inhibitor increased the number of colonies. However, when the expression of ITGB5 or β-catenin was knocked down, the stimulatory effect of the miR-185 inhibitor was compromised (Fig. 6g and h). Similarly, in transwell assays, the miR-185 inhibitor promoted cell migration, in which process the expression of ITGB5 and β-catenin was also required (Fig. 6i and j). These results indicate that miR-185 regulates the growth and migration of HCC cells through effects on ITGB5 and β-catenin.

Decreased miR-185 levels in HCC

We compared the expression levels of miR-185 in HCC specimens and adjacent normal tissues. As shown in Fig. 7a, the expression of miR-185 was significantly decreased in HCC tissues compared to in normal adjacent samples. Correspondingly, the immunohistochemistry analyses of these HCC samples showed elevated expressions of ITGB5 and β-catenin (Fig. 7b, c, and d). With these clinical data, we examined the association of miR-185 expression with the level of ITGB5 and observed significant reverse correlation (Fig. 7e). Meanwhile, the expression of β-catenin is positively related to the level of ITGB5 (Fig. 7f).

Discussion

HCC is the third most common cause of cancer mortality worldwide, and tumour recurrence due to de novo tumours and dissemination are the predominant causes leading to the high mortality [1, 4, 32, 33].The present study demonstrated that ITGB5 is a critical regulator of the tumorigenic potential of HCC, not only facilitating cancer cell growth but also promoting cell migration. In clinical data, we found that ITGB5 expression was frequently increased in tumour tissues compared to adjacent normal specimens correlating with older patients, a large tumour size and an advanced clinical stage. In functional experiments, the depletion of ITGB5 by shRNA markedly inhibits HCC cell clone formation, which reflects the ITGB5 function in the replicative potential of cancer cells. Importantly, the knock down of ITGB5 effectively attenuates the migratory ability of HCC cells and vice versa.
Canonically, integrins affect intracellular signalling by activating integrin-associated kinases due to lack of enzymatic activity [8, 9, 34], and mechanistic studies of ITGB5 mainly focus on activating Src-FAK and MEK-ERK signalling pathways, which profoundly contribute to the tumorigenic capacity of the malignancies [19]. In our present experiments, results show the novel role of ITGB5 in HCC, which interacted with β-catenin and upregulated β-catenin expression. When ITGB5 had dysregulated expression, the protein level of β-catenin was subsequently altered in HCC cells, which was consistent with the clinical specimen for immunohistochemistry correlation analyses. Interestingly, we noticed that the increase of β-catenin by ITGB5 is in a transcription-independent manner, and the mRNA level of β-catenin did not change when ITGB5 was overexpressed or knocked down, suggesting that ITGB5 is likely to control β-catenin in the phase of protein translation and/or degradation. A series of experiments revealed that the regulation of ITGB5 on β-catenin mainly relied on inhibiting its degradation. Using a protein stability assay and immunoprecipitation assays, we confirmed that ITGB5depletion triggers the strong ubiquitylation of β-catenin. However, the in-depth molecular basis by which ITGB5 regulates the ubiquitylation of β-catenin is still unclear, and the underlying mechanisms will be investigated in our future work.
The expression level of ITGB5 was increased in HCC. However, the mechanism is still unknown. Previous reports indicated that gene expression is partially regulated by a class of miRNAs, and identifying the target gene of miRNA that is involved in the malignancy behaviours is important for exploring the molecular mechanisms [35]. In our manuscript, we revealed novel candidate miRNAs that could regulate ITGB5 expression. Among the 6 miRNAs, both miR-271 and miR-185 suppressed the luciferase activity of ITGB5 3′UTR, and miR-185 decreased the protein level of ITGB5 most significantly. Previously, miR-185 has been demonstrated to be a key suppressor in HCC [36, 37], which was similar with our result. Nonetheless, the reason for the downregulation of miR-185 in HCC still remains to be discovered in the future.
Collectively, the current study provides a novel insight into the function of ITGB5 in HCC. ITGB5, negatively modulated by miR-185, promoted tumour growth and migration by regulating β-catenin stability in vitro and in vivo. An increase of ITGB5 was associated with β-catenin upregulation and a decrease in miR-185 in HCC tissues. Thus, our data suggested that ITGB5 may be a novel diagnostic biomarker and a potential therapeutic target in HCC.

Conclusions

In summary, our study indicates that ITGB5 was an important regulator in HCC and found that ITGB5, a miR-185 targeted gene, could directly interact with β-catenin and inhibit its degradation, leading to WNT/β-catenin activity. Thus, Our data uncover that the miR-185-ITGB5-β-catenin pathway plays an important role in HCC tumorigenesis, and ITGB5 may be a promising specific target for HCC therapy.

Acknowledgements

This research was supported by the National Nature Science Foundation of China (No.30870719 to Zhongyu Wang, No. 81402260 to Chuanchun Han) and the Liaoning Provincial Natural Science Foundation of China (No. 2015020309 to Chuanchun Han).

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 2007;132:2557–76.CrossRefPubMed El-Serag HB, Rudolph KL. Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology. 2007;132:2557–76.CrossRefPubMed
2.
Zurück zum Zitat Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.CrossRefPubMed Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87–108.CrossRefPubMed
3.
Zurück zum Zitat Han LL, Lv Y, Guo H, Ruan ZP, Nan KJ. Implications of biomarkers in human hepatocellular carcinoma pathogenesis and therapy. World J Gastroenterol. 2014;20:10249–61.CrossRefPubMedPubMedCentral Han LL, Lv Y, Guo H, Ruan ZP, Nan KJ. Implications of biomarkers in human hepatocellular carcinoma pathogenesis and therapy. World J Gastroenterol. 2014;20:10249–61.CrossRefPubMedPubMedCentral
4.
6.
Zurück zum Zitat Portolani N, Coniglio A, Ghidoni S, Giovanelli M, Benetti A, Tiberio GA, Giulini SM. Early and late recurrence after liver resection for hepatocellular carcinoma: prognostic and therapeutic implications. Ann Surg. 2006;243:229–35.CrossRefPubMedPubMedCentral Portolani N, Coniglio A, Ghidoni S, Giovanelli M, Benetti A, Tiberio GA, Giulini SM. Early and late recurrence after liver resection for hepatocellular carcinoma: prognostic and therapeutic implications. Ann Surg. 2006;243:229–35.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Liang Y, Feng Y, Zong M, Wei X, Lee J, Feng Y, Li H, Yang G, Wu ZJ, Fu XD, Feng GS. beta-Catenin Deficiency in Hepatocytes Aggravates Hepatocarcinogenesis Driven by Oncogenic beta-Catenin and MET. Hepatol. 20:377–495. Liang Y, Feng Y, Zong M, Wei X, Lee J, Feng Y, Li H, Yang G, Wu ZJ, Fu XD, Feng GS. beta-Catenin Deficiency in Hepatocytes Aggravates Hepatocarcinogenesis Driven by Oncogenic beta-Catenin and MET. Hepatol. 20:377–495.
9.
10.
Zurück zum Zitat Hood JD, Frausto R, Kiosses WB, Schwartz MA, Cheresh DA. Differential alphav integrin-mediated Ras-ERK signaling during two pathways of angiogenesis. J Cell Biol. 2003;162:933–43.CrossRefPubMedPubMedCentral Hood JD, Frausto R, Kiosses WB, Schwartz MA, Cheresh DA. Differential alphav integrin-mediated Ras-ERK signaling during two pathways of angiogenesis. J Cell Biol. 2003;162:933–43.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Alavi A, Hood JD, Frausto R, Stupack DG, Cheresh DA. Role of Raf in vascular protection from distinct apoptotic stimuli. Science. 2003;301:94–6.CrossRefPubMed Alavi A, Hood JD, Frausto R, Stupack DG, Cheresh DA. Role of Raf in vascular protection from distinct apoptotic stimuli. Science. 2003;301:94–6.CrossRefPubMed
12.
Zurück zum Zitat Tancioni I, Uryu S, Sulzmaier FJ, Shah NR, Lawson C, Miller NL, Jean C, Chen XL, Ward KK, Schlaepfer DD. FAK inhibition disrupts a beta5 integrin signaling axis controlling anchorage-independent ovarian carcinoma growth. Mol Cancer Ther. 2014;13:2050–61.CrossRefPubMedPubMedCentral Tancioni I, Uryu S, Sulzmaier FJ, Shah NR, Lawson C, Miller NL, Jean C, Chen XL, Ward KK, Schlaepfer DD. FAK inhibition disrupts a beta5 integrin signaling axis controlling anchorage-independent ovarian carcinoma growth. Mol Cancer Ther. 2014;13:2050–61.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Su G, Hodnett M, Wu N, Atakilit A, Kosinski C, Godzich M, Huang XZ, Kim JK, Frank JA, Matthay MA, Sheppard D, Pittet JF. Integrin alphavbeta5 regulates lung vascular permeability and pulmonary endothelial barrier function. Am J Respir Cell Mol Biol. 2007;36:377–86.CrossRefPubMed Su G, Hodnett M, Wu N, Atakilit A, Kosinski C, Godzich M, Huang XZ, Kim JK, Frank JA, Matthay MA, Sheppard D, Pittet JF. Integrin alphavbeta5 regulates lung vascular permeability and pulmonary endothelial barrier function. Am J Respir Cell Mol Biol. 2007;36:377–86.CrossRefPubMed
14.
Zurück zum Zitat Eliceiri BP, Puente XS, Hood JD, Stupack DG, Schlaepfer DD, Huang XZ, Sheppard D, Cheresh DA. Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol. 2002;157:149–60.CrossRefPubMedPubMedCentral Eliceiri BP, Puente XS, Hood JD, Stupack DG, Schlaepfer DD, Huang XZ, Sheppard D, Cheresh DA. Src-mediated coupling of focal adhesion kinase to integrin alpha(v)beta5 in vascular endothelial growth factor signaling. J Cell Biol. 2002;157:149–60.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Li F, Liu Y, Kan X, Li Y, Liu M, Lu JG. Elevated expression of integrin alphav and beta5 subunit in laryngeal squamous-cell carcinoma associated with lymphatic metastasis and angiogenesis. Pathol Res Pract. 2013;209:105–9.CrossRefPubMed Li F, Liu Y, Kan X, Li Y, Liu M, Lu JG. Elevated expression of integrin alphav and beta5 subunit in laryngeal squamous-cell carcinoma associated with lymphatic metastasis and angiogenesis. Pathol Res Pract. 2013;209:105–9.CrossRefPubMed
16.
Zurück zum Zitat Bianchi A, Gervasi ME, Bakin A. Role of beta5-integrin in epithelial-mesenchymal transition in response to TGF-beta. Cell Cycle. 2010;9:1647–59.CrossRefPubMed Bianchi A, Gervasi ME, Bakin A. Role of beta5-integrin in epithelial-mesenchymal transition in response to TGF-beta. Cell Cycle. 2010;9:1647–59.CrossRefPubMed
17.
Zurück zum Zitat Zhang H, Li Z, Viklund E-K, Strömblad S. p21-activated kinase 4 interacts with integrin αvβ5 and regulates αvβ5-mediated cell migration. J Cell Biol. 2002;158:1287–97.CrossRefPubMedPubMedCentral Zhang H, Li Z, Viklund E-K, Strömblad S. p21-activated kinase 4 interacts with integrin αvβ5 and regulates αvβ5-mediated cell migration. J Cell Biol. 2002;158:1287–97.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Majhen D, Stojanovic N, Speljko T, Brozovic A, De Zan T, Osmak M, Ambriovic-Ristov A. Increased expression of the coxsackie and adenovirus receptor downregulates alphavbeta3 and alphavbeta5 integrin expression and reduces cell adhesion and migration. Life Sci. 2011;89:241–9.CrossRefPubMed Majhen D, Stojanovic N, Speljko T, Brozovic A, De Zan T, Osmak M, Ambriovic-Ristov A. Increased expression of the coxsackie and adenovirus receptor downregulates alphavbeta3 and alphavbeta5 integrin expression and reduces cell adhesion and migration. Life Sci. 2011;89:241–9.CrossRefPubMed
19.
Zurück zum Zitat Bianchi-Smiraglia A, Paesante S, Bakin AV. Integrin beta5 contributes to the tumorigenic potential of breast cancer cells through the Src-FAK and MEK-ERK signaling pathways. Oncogene. 2013;32:3049–58.CrossRefPubMed Bianchi-Smiraglia A, Paesante S, Bakin AV. Integrin beta5 contributes to the tumorigenic potential of breast cancer cells through the Src-FAK and MEK-ERK signaling pathways. Oncogene. 2013;32:3049–58.CrossRefPubMed
20.
Zurück zum Zitat Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35.CrossRefPubMedPubMedCentral Hoshino A, Costa-Silva B, Shen TL, Rodrigues G, Hashimoto A, Tesic Mark M, Molina H, Kohsaka S, Di Giannatale A, Ceder S, Singh S, Williams C, et al. Tumour exosome integrins determine organotropic metastasis. Nature. 2015;527:329–35.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Tian Y, Mok MT, Yang P, Cheng AS. Epigenetic Activation of Wnt/beta-Catenin Signaling in NAFLD-Associated Hepatocarcinogenesis. Cancers. 2016;8:66–76. Tian Y, Mok MT, Yang P, Cheng AS. Epigenetic Activation of Wnt/beta-Catenin Signaling in NAFLD-Associated Hepatocarcinogenesis. Cancers. 2016;8:66–76.
22.
Zurück zum Zitat Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, Barroso-Sousa R, Kaplan JB, Chae YK, Giles FJ. Wnt/beta-catenin pathway: modulating anticancer immune response. J Hematol Oncol. 2017;10:101.CrossRefPubMedPubMedCentral Pai SG, Carneiro BA, Mota JM, Costa R, Leite CA, Barroso-Sousa R, Kaplan JB, Chae YK, Giles FJ. Wnt/beta-catenin pathway: modulating anticancer immune response. J Hematol Oncol. 2017;10:101.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Monga SP. Beta-catenin signaling and roles in liver homeostasis, injury, and tumorigenesis. Gastroenterology. 2015;148:1294–310.CrossRefPubMed Monga SP. Beta-catenin signaling and roles in liver homeostasis, injury, and tumorigenesis. Gastroenterology. 2015;148:1294–310.CrossRefPubMed
24.
Zurück zum Zitat Piva MBR, Jakubzig B, Bendas G. Integrin Activation Contributes to Lower Cisplatin Sensitivity in MV3 Melanoma Cells by Inducing the Wnt Signalling Pathway. Cancers 2017;9. Piva MBR, Jakubzig B, Bendas G. Integrin Activation Contributes to Lower Cisplatin Sensitivity in MV3 Melanoma Cells by Inducing the Wnt Signalling Pathway. Cancers 2017;9.
25.
Zurück zum Zitat Groulx JF, Giroux V, Beausejour M, Boudjadi S, Basora N, Carrier JC, Beaulieu JF. Integrin alpha6A splice variant regulates proliferation and the Wnt/beta-catenin pathway in human colorectal cancer cells. Carcinogenesis. 2014;35:1217–27.CrossRefPubMedPubMedCentral Groulx JF, Giroux V, Beausejour M, Boudjadi S, Basora N, Carrier JC, Beaulieu JF. Integrin alpha6A splice variant regulates proliferation and the Wnt/beta-catenin pathway in human colorectal cancer cells. Carcinogenesis. 2014;35:1217–27.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Han C, Gu H, Wang J, Lu W, Mei Y, Wu M. Regulation of L-threonine dehydrogenase in somatic cell reprogramming. Stem Cells. 2013;31:953–65.CrossRefPubMed Han C, Gu H, Wang J, Lu W, Mei Y, Wu M. Regulation of L-threonine dehydrogenase in somatic cell reprogramming. Stem Cells. 2013;31:953–65.CrossRefPubMed
27.
Zurück zum Zitat Zhang L, Wang Y, Li X, Xia X, Li N, He R, He H, Han C, Zhao W. ZBTB7A enhances Osteosarcoma Chemoresistance by Transcriptionally repressing lncRNALINC00473-IL24 activity. Neoplasia. 2017;19:908–18.CrossRefPubMedPubMedCentral Zhang L, Wang Y, Li X, Xia X, Li N, He R, He H, Han C, Zhao W. ZBTB7A enhances Osteosarcoma Chemoresistance by Transcriptionally repressing lncRNALINC00473-IL24 activity. Neoplasia. 2017;19:908–18.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Han C, Jin L, Mei Y, Wu M. Endoplasmic reticulum stress inhibits cell cycle progression via induction of p27 in melanoma cells. Cell Signal. 2013;25:144–9.CrossRefPubMed Han C, Jin L, Mei Y, Wu M. Endoplasmic reticulum stress inhibits cell cycle progression via induction of p27 in melanoma cells. Cell Signal. 2013;25:144–9.CrossRefPubMed
29.
Zurück zum Zitat Yang X, Du T, Wang X, Zhang Y, Hu W, Du X, Miao L, Han C. IDH1, a CHOP and C/EBPbeta-responsive gene under ER stress, sensitizes human melanoma cells to hypoxia-induced apoptosis. Cancer Lett. 2015;365:201–10.CrossRefPubMed Yang X, Du T, Wang X, Zhang Y, Hu W, Du X, Miao L, Han C. IDH1, a CHOP and C/EBPbeta-responsive gene under ER stress, sensitizes human melanoma cells to hypoxia-induced apoptosis. Cancer Lett. 2015;365:201–10.CrossRefPubMed
30.
Zurück zum Zitat Ma B, Yuan Z, Zhang L, Lv P, Yang T, Gao J, Pan N, Wu Q, Lou J, Han C, Zhang B. Long non-coding RNA AC023115.3 suppresses chemoresistance of glioblastoma by reducing autophagy. Biochim Biophys Acta. 2017;1864:1393–404.CrossRef Ma B, Yuan Z, Zhang L, Lv P, Yang T, Gao J, Pan N, Wu Q, Lou J, Han C, Zhang B. Long non-coding RNA AC023115.3 suppresses chemoresistance of glioblastoma by reducing autophagy. Biochim Biophys Acta. 2017;1864:1393–404.CrossRef
31.
Zurück zum Zitat Li X, Ma C, Zhang L, Li N, Zhang X, He J, He R, Shao M, Wang J, Kang L, Han C. LncRNAAC132217.4, a KLF8-regulated long non-coding RNA, facilitates oral squamous cell carcinoma metastasis by upregulating IGF2 expression. Cancer Lett. 2017;407:45–56.CrossRefPubMed Li X, Ma C, Zhang L, Li N, Zhang X, He J, He R, Shao M, Wang J, Kang L, Han C. LncRNAAC132217.4, a KLF8-regulated long non-coding RNA, facilitates oral squamous cell carcinoma metastasis by upregulating IGF2 expression. Cancer Lett. 2017;407:45–56.CrossRefPubMed
32.
Zurück zum Zitat Chen YJ, Yeh SH, Chen JT, Wu CC, Hsu MT, Tsai SF, Chen PJ, Lin CH. Chromosomal changes and clonality relationship between primary and recurrent hepatocellular carcinoma. Gastroenterology. 2000;119:431–40.CrossRefPubMed Chen YJ, Yeh SH, Chen JT, Wu CC, Hsu MT, Tsai SF, Chen PJ, Lin CH. Chromosomal changes and clonality relationship between primary and recurrent hepatocellular carcinoma. Gastroenterology. 2000;119:431–40.CrossRefPubMed
33.
Zurück zum Zitat Imamura H, Matsuyama Y, Tanaka E, Ohkubo T, Hasegawa K, Miyagawa S, Sugawara Y, Minagawa M, Takayama T, Kawasaki S, Makuuchi M. Risk factors contributing to early and late phase intrahepatic recurrence of hepatocellular carcinoma after hepatectomy. J Hepatol. 2003;38:200–7.CrossRefPubMed Imamura H, Matsuyama Y, Tanaka E, Ohkubo T, Hasegawa K, Miyagawa S, Sugawara Y, Minagawa M, Takayama T, Kawasaki S, Makuuchi M. Risk factors contributing to early and late phase intrahepatic recurrence of hepatocellular carcinoma after hepatectomy. J Hepatol. 2003;38:200–7.CrossRefPubMed
34.
Zurück zum Zitat Luo M, Guan JL. Focal adhesion kinase: a prominent determinant in breast cancer initiation, progression and metastasis. Cancer Lett. 2010;289:127–39.CrossRefPubMed Luo M, Guan JL. Focal adhesion kinase: a prominent determinant in breast cancer initiation, progression and metastasis. Cancer Lett. 2010;289:127–39.CrossRefPubMed
35.
Zurück zum Zitat Liu C, Chen Z, Hu X, Wang L, Li C, Xue J, Zhang P, Chen W, Jiang A. MicroRNA-185 downregulates androgen receptor expression in the LNCaP prostate carcinoma cell line. Mol Med Rep. 2015;11:4625–32.CrossRefPubMed Liu C, Chen Z, Hu X, Wang L, Li C, Xue J, Zhang P, Chen W, Jiang A. MicroRNA-185 downregulates androgen receptor expression in the LNCaP prostate carcinoma cell line. Mol Med Rep. 2015;11:4625–32.CrossRefPubMed
36.
Zurück zum Zitat Qadir XV, Han C, Lu D, Zhang J, Wu T. miR-185 inhibits hepatocellular carcinoma growth by targeting the DNMT1/PTEN/Akt pathway. Am J Pathol. 2014;184:2355–64.CrossRefPubMedPubMedCentral Qadir XV, Han C, Lu D, Zhang J, Wu T. miR-185 inhibits hepatocellular carcinoma growth by targeting the DNMT1/PTEN/Akt pathway. Am J Pathol. 2014;184:2355–64.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Zhou L, Liu S, Han M, Feng S, Liang J, Li Z, Li Y, Lu H, Liu T, Ma Y, Cheng J. MicroRNA-185 induces potent autophagy via AKT signaling in hepatocellular carcinoma. Tumour Biol. 2017;39:1010428317694313.CrossRefPubMed Zhou L, Liu S, Han M, Feng S, Liang J, Li Z, Li Y, Lu H, Liu T, Ma Y, Cheng J. MicroRNA-185 induces potent autophagy via AKT signaling in hepatocellular carcinoma. Tumour Biol. 2017;39:1010428317694313.CrossRefPubMed
Metadaten
Titel
RETRACTED ARTICLE: Integrin-β5, a miR-185-targeted gene, promotes hepatocellular carcinoma tumorigenesis by regulating β-catenin stability
verfasst von
Zhikun Lin
Ruiping He
Haifeng Luo
Chang Lu
Zhen Ning
Yuanhang Wu
Chuanchun Han
Guang Tan
Zhongyu Wang
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2018
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0691-9

Weitere Artikel der Ausgabe 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.