Activation of phosphatidylinositol 3-kinase/Akt signaling mediates sorafenib-induced invasion and metastasis in hepatocellular carcinoma

  • Authors:
    • Haiyong Wang
    • Litao Xu
    • Xiaoyan Zhu
    • Peng Wang
    • Huiying Chi
    • Zhiqiang Meng
  • View Affiliations

  • Published online on: July 23, 2014     https://doi.org/10.3892/or.2014.3352
  • Pages: 1465-1472
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Sorafenib, an antiangiogenic agent, can promote tumor invasion and metastasis. The phosphatidylinositol 3-kinase (PI3K)/Akt/Snail-dependent pathway plays an important role in tumor invasion and metastasis. Yet, little is known concerning the role of the PI3K/Akt/Snail-dependent pathway in sorafenib‑induced invasion and metastasis of hepatic carcinoma (HCC). A human HCC orthotopic xenograft model was established, and sorafenib (30 mg/kg/day) was administered orally. Tumor growth and intrahepatic metastasis were assessed, and immunohistochemistry was applied to analyze the activation of the PI3K/Akt/Snail-dependent pathway. HCC cell lines were treated with sorafenib (1, 5 and 10 µM), and proliferation, migration and invasion were assessed. Western blotting and real-time polymerase chain reaction (RT-PCR) were used to examine the related gene expression of epithelial-mesenchymal transition (EMT) markers and the PI3K/Akt/Snail-dependent pathway. Sorafenib inhibited tumor growth and promoted intrahepatic invasion and metastasis of the orthotopic tumors grown from SMMC7721-GFP cells in vivo. Additionally, sorafenib promoted EMT and invasion and metastasis of HCC cells in vitro. Importantly, sorafenib enhanced PI3K and Akt activation and upregulation of the expression of transcription factor Snail, a critical EMT mediator. The upregulation of transcription factor Snail expression by sorafenib may be related to activation of the PI3K/AKT signaling pathway. The PI3K/Akt/Snail-dependent pathway may mediate the pro-invasive and pro-metastatic effects of sorafenib on HCC by inducing EMT.

Introduction

Hepatic carcinoma (HCC) is the fifth most common malignancy worldwide and the second leading cause of cancer-related death in Asia generally and in China in particular (1). Currently, surgical resection and liver transplantation offer the best potential for treating HCC (24), but most HCC patients are diagnosed in advanced stages. At present, sorafenib, a multikinase inhibitor with antiangiogenic and antiproliferative effects, currently sets the new standard for advanced HCC (5,6). However, the survival benefit is only 2.8 months.

Antiangiogenic therapy has been thought to hold significant potential for the treatment of cancer (7). However, clinical and preclinical observations indicate that these therapies may have limited efficacy. Although these agents typically produce inhibition of primary tumor growth, lasting responses are rare, with only a moderate increase in progression-free survival and little benefit in overall survival (8). In addition, recent reports describe that treatment of tumor-bearing mice with antiangiogenic drugs leads to increased local tumor cell invasion and enhanced distant metastasis after prolonged treatment or after only short-term treatment (9,10). Notably, sorafenib, the only approved molecular-targeted drug for HCC, was found to promote invasion and metastasis of HCC by increased intrahepatic metastasis, lung metastasis, and circulating tumor cells in tumors with higher expression of HTATIP2 in xenograft models (11). Therefore, it is important to clarify the molecular mechanisms of invasion and metastasis caused by sorafenib from all aspects in HCC.

Epithelial-mesenchymal transition (EMT) plays a key role in tumor invasion and metastasis. During this process, epithelial cells lose their epithelial signatures while acquiring the characteristics of mesenchymal cells including morphology, cellular structure and biological function (12). Transcription factor Snail has also been shown to confer survival properties either concomitantly with induction of EMT or independent of EMT (1315). Snail, Slug and Twist transcription factors can act as E-box repressors and block E-cadherin transcription (16). In addition, Snail transcription factor can mediate an increase in expression of mesenchymal markers such as vimentin, fibronectin, matrix metalloproteinases (MMPs) and RhoA (1720). The overall effect of Snail is increased migration and invasion (18,19).

Numerous signaling pathways are involved in the regulation of EMT. PI3K/Akt signaling is an important survival/proliferative pathway involving various growth factors, cytokines and activation of receptors (21). In addition, the PI3K/Akt signaling pathway plays a key role in the control of cell invasion and metastasis and the activation of PI3K/AKT is a central feature of EMT in the development of cancer (2227). On the one hand, the PI3K/AKT signaling pathway can increase the expression of matrix metalloproteinases to induce EMT (28,29). On the other hand, the PI3K/AKT signaling pathway can upregulate the expression of transcription factor Snail to induce EMT (3032). Notably, activation of the PI3K/Akt signaling pathway plays a key role in mediating resistance to sorafenib. The combination of MK-2206, an Akt inhibitor, and sorafenib overcomes such resistance (33). Yet, little is known concerning the role of the PI3K/Akt signaling pathway on the invasion and metastasis induced by sorafenib in HCC.

In the present study, we tested and verified that sorafenib promotes invasion and metastasis of HCC by inducing EMT. More importantly, we showed that activation of the PI3K/Akt/Snail-dependent pathway may play a key role in this process.

Materials and methods

Reagents and antibodies

Sorafenib was purchased from Bayer Corporation (West Haven, CT, USA). Antibodies against E-cadherin, N-cadherin, vimentin, Snail and GAPDH were purchased from Epitomics (Burlingame, CA, USA); antibodies against p-PI3K and p-AKT were purchased from Bioworld Technology (Minneapolis, MN, USA).

Cell culture

The human HCC cell lines SMMC7721 and HCCLM3 originated from the American Type Culture Collection (ATCC) and were cultured in RPMI-1640 containing 10% fetal bovine serum (FBS; Biochrom, Berlin, Germany) in 5% CO2 at 37°C. SMMC7721-GFP cells were SMMC7721 cells transfected with green fluorescence protein (GFP) and were labeled as SMMC7721-GFP cells.

Cell proliferation, migration and invasion assays

Cell proliferation analysis was performed as previously described by us (34). Briefly, cells were plated at 5,000/well in 96-well microtiter plates and incubated overnight at 37°C in a humidified incubator containing 5% CO2. On the following day, various concentrations of sorafenib were added to the wells, and cultures were incubated for an additional 24, 48 and 72 h. Cell viability was determined using a Cell Counting Kit-8 (Dojindo, Gaithersburg, MD, USA) according to the manufacturer’s instructions. For cell migration assay, cell migration was assessed using the Transwell assay (Boyden chambers; Corning, Cambridge, MA, USA). Cells (5×104) were seeded in serum-free medium in the upper chamber and allowed to migrate toward the lower chamber that contained 10% FBS. After 48 h, cells that had traveled through and adhered to the underside of the membrane were counted at ×200 magnification. The cell invasion assay was carried out similarly, except that 50 μl Matrigel (BD Biosciences, Franklin Lakes, NJ, USA) diluted 1:6 with serum-free medium was added to each well overnight before cells (2×105) were seeded onto the membrane.

Animal models and treatments

Six-week-old BALBc nu/nu female mice were obtained from the Shanghai Institute of Material Medica, Chinese Academy of Science. All mice were bred in laminar flow cabinets under specific pathogen-free conditions. We followed internationally recognized guidelines on animal welfare. The study design was approved by the Animal Ethics Committee, and the experiments were undertaken in accordance with the Ethical Principles of Animal Experimentation of Fudan University. SMMC7721-GFP cells [5×106/0.2 ml phosphate-buffered saline (PBS)] were subcutaneously inoculated into the right flanks of 6-week-old BALBc nu/nu female mice. After 4 weeks, non-necrotic tumor tissue was cut into 1 mm3 pieces and orthotopically implanted into the liver. Treatment was started 2 weeks after orthotopic implantation of the tumors. Mice were randomly separated into two groups with 6 mice in each group. Mice in the experimental group received 30 mg/kg/day sorafenib, whereas the control mice received vehicle alone. Animal weight was measured twice a week for 4 weeks. At the end of the experiment, mice were sacrificed, tumors were excised from each mouse, weighed and snap-frozen for further analysis.

Detection of metastasis

Tumors were excised and their largest (a) and smallest (b) diameters were measured to calculate tumor volume (V = ab2/2). The livers were also excised, and green fluorescent protein-positive metastatic foci were imaged by Lumazone imaging system (Mag Biosystems, Tucson, AZ, USA). Hematoxylin and eosin staining (H&E) was further applied to detect liver metastasis.

Western blot analysis

Cells were washed with cold PBS and lysed in culture dishes using PhosphoSafe™ Extraction Reagent (Merck, Darmstadt, Germany) containing 1% protease inhibitor cocktail (EDTA-Free; Thermo, San Jose, CA, USA). Protein concentrations were then determined using Bio-Rad detergent compatible protein assays (Bio-Rad, Hercules, CA, USA). A total of 30 μg protein from each of the control and treated cell lysates was loaded on 8–12% gradient NuPAGE gels (Novex, San Diego, CA, USA), electrophoresed under reducing conditions, and transferred onto polyvinylidene difluoride membranes (0.22 Å; Millipore). Western blot analysis was carried out as previously described (34).

Immunohistochemistry

Procedures for the immunohistochemistry were previously described (35). Briefly, the tumor sections were stained with rabbit anti-p-Akt, and rabbit antip-PI3K at 4°C overnight. Goat anti-rabbit IgG/horseradish peroxidase was applied as the secondary antibody according to the standard protocols provided by the manufacturer. For negative controls, primary antibodies were replaced with PBS. The procedures were performed by two independent investigators, both of whom were blinded to the model/treatment type for the series of experiments.

Real-time polymerase chain reaction

RT-PCR analysis was performed as previously described by us (36). The following primers for amplification of human genes were used: E-cadherin forward, 5′-AGCCCCGCCTTATGATTCTCTG-3′ and reverse, 5′-TGCCCCATTCGTTCAAGTAGTCAT-3′; N-cadherin forward, 5′-CCACGCCGAGCCCCAGTAT-3′ and reverse, 5′-GGCCCCCAGTCGTTCAGGTAAT-3′; vimentin forward, 5′-CCTTGACATTGAGATTGCCACCTA-3′ and reverse, 5′-TCATCGTGATGCTGAGAAGTTTCG-3′; Snail forward, 5′-CAGCCTGGGTGCCCTCAAGAT-3′ and reverse, 5′-GCACACGCCTGGCACTGGTA-3′.

Statistical analysis

All analyses of the results were performed using the GraphPad Prism software version 5.0 (GraphPad Software, San Diego, CA, USA) and the SPSS 19.0 software package (SPSS, Inc., Chicago, IL, USA). Statistical analyses were performed using the Student’s t-test and analysis of variance (ANOVA) models. Differences were considered statistically significant at P<0.05.

Results

Sorafenib promotes invasion and migration in vivo

To elucidate the effects of sorafenib on HCC invasion and migration, mice were orthopically implanted with SMMC7721-GFP cells and treated with 30 mg/kg/day sorafenib. Tumor growth and metastasis were monitored. Our results showed that sorafenib substantially reduced the primary tumor growth compared with the control tumors. Tumor weight and volume were reduced in the sorafenib-treated mice (Fig. 1A and B). Additionally, sorafenib was well tolerated by the mice as no apparent weight loss was noted (Fig. 1C). Unfortunately, sorafenib-treated mice developed more intrahepatic metastatic lesions and exhibited irregular tumor margins as detected by green fluorescence imaging (Fig. 1D). To further explore the effect of sorafenib on the invasion and metastasis of HCC, liver metastatic nodules were evaluated by H&E staining as observed under a microscope. The number of metastatic nodules was then statistically analyzed. A higher number of intrahepatic metastatic nodules was detected in the sorafenib-treated mice (Fig. 1E).

Sorafenib promotes invasion and migration of HCC cells

As sorafenib promoted invasion and migration in vivo, we wanted to further validate whether sorafenib could promote the invasion and migration in vitro. Cell proliferation assay was applied to assess the proliferative effect on hepatoma cells after sorafenib treatment. The antiproliferative effect of sorafenib on HCC cells was dose- and time-dependent at a concentration of 1–10 μM in the SMMC7721 and HCCLM3 cells (Fig. 2A). Sorafenib at a concentration of 5 μM, with little effect on cell proliferation, was applied to assess the effect of sorafenib on the migration and invasion of HCC cells. Cells (5×104 or 2×105) were seeded in the upper chamber. A higher number of metastatic and invasive cells were detected in the sorafenib-treated HCC cells as assessed by Transwell assay (Fig. 2B and C).

Sorafenib promotes EMT in HCC cells

EMT is well known to closely correlate with cancer metastasis. To test and verify whether 5 μM sorafenib promotes the EMT process, we evaluated the expression of EMT markers in the sorafenib-treated and the control cells. As expected, SMMC7721 and HCCLM3 cells treated with 5 μM sorafenib underwent significant morphological changes and displayed the mesenchymal phenotype (Fig. 3A). Importantly, epithelial marker E-cadherin was downregulated and mesenchymal markers N-cadherin and vimentin were upregulated in the sorafenib-treated cells (Fig. 3B). RT-PCR assay further confirmed the decreased levels of epithelial marker E-cadherin and the increased levels of mesenchymal markers N-cadherin and vimentin in the SMMC7721 and HCCLM3 cells (Fig. 3C).

Sorafenib upregulates the expression of Snail in vitro

As zinc-finger transcriptional repressor Snail plays a key role in EMT-mediated tumor invasion and metastasis, we ascertained whether Snail is involved in sorafenib-mediated EMT. HCC cells were treated with 5 μM sorafenib and western blot analysis and RT-PCR were carried out to measure Snail expression. As anticipated, transcription factor Snail was upregulated in the SMMC7721 and HCCLM3 cells, when compared to the controls (Fig. 4A and B).

Sorafenib activates the PI3K/AKT signaling pathway in vivo and vitro

As a highly conserved cellular program, EMT has been documented to involve several important pathways. Accumulating research suggests that PI3K/Akt activation plays a pivotal role in tumor progression via induction of EMT. The PI3K/Akt/GSK-3β/Snail-dependent signaling pathway is involved in HCC. Thus, we detected the activity of PI3K/AKT in the sorafenib-induced invasion and metastasis of HCC. The results showed that the PI3K/AKT signaling pathway was activated in the HCC cells treated with 5 μM sorafenib (Fig. 5A). In addition, the marginal tissues of the xenografts were analyzed by immunohistochemical staining as described earlier. PI3K/AKT phosphorylation levels were also upregulated (Fig. 5B).

Discussion

As a result of the SHARP and ORIENTAL trials, sorafenib has become the new standard therapy for patients with advanced hepatic carcinoma (HCC) (5,6). However, the survival benefit is only a few months. Furthermore, tumors may progress during sorafenib treatment (911). In the present study, we demonstrated that sorafenib exerted an antitumor effect and inhibited tumor growth in mouse models of cancer. However, sorafenib also promoted invasion and metastasis of HCC in this tumor model by inducing EMT. Similar observations were reported by other authors. Importantly, we found that sorafenib upregulated the expression of transcription factor Snail and activated the PI3K/Akt signaling pathway.

In a previous study, increased local invasion and distant metastasis during or after treatment with sorafenib were observed (11). EMT plays a key role in tumor invasion and metastasis. EMT is also reported to be involved in the progression of HCC and is correlated with the prognosis of patients (37). In the present study, more metastatic lesions in the livers of nude mice were detected in the sorafenib treatment group. In addition, HCC cell lines, including SMMC7721 and HCCLM3, were treated with 5 μM sorafenib, with little effect on cell proliferation as confirmed by Cell Counting Kit-8. Surprisingly, morphology of the cells underwent significant changes and presented a mesenchymal phenotype after treatment for 72 h. Then EMT-related markers were analyzed. As anticipated, mesenchymal markers were significantly upregulated and epithelial markers were markedly decreased in the sorafenib-treated cells. Transwell assay was also used to analyze the ability of hepatoma cell invasion and migration. Invasion and migration capacity of the HCC cell lines was enhanced. Therefore, these data indicate that sorafenib may promote HCC invasion and metastasis by the induction of EMT, consistent with other reports.

The Snail transcription factor, a member of the Snail superfamily, is a zinc finger protein that can mediate EMT through downregulation of cell adhesion molecules such as E-cadherin by binding several E-boxes located in the promotor region (16). Snail has also been shown to confer survival properties either concomitantly with induction of EMT or independent of EMT. Snail plays an important role in inducing EMT in HCC cells (38). In cancer patients, an EMT-phenotype transcriptome profile, with increased Snail expression correlates with invasive tumors. Phosphorylation and subsequent degradation of Snail is controlled by GSK-3β, which is predominantly regulated by PI3K/Akt (39). The PI3K/Akt/GSK-3β/Snail-dependent signaling pathway can mediate invasion and metastasis of HCC (40,41). Increasing evidence also demonstrates that activation of the PI3K/Akt pathway plays a central role in the EMT process and correlates with an aggressive phenotype in several types of malignancies (2227). Several signaling pathways that induce EMT and metastasis often converge at or activate PI3K/Akt, which itself is frequently activated during tumor progression. Hyperactivation of Akt is closely associated with elevated invasion and metastasis, resulting in a poor prognosis and a greater probability of relapse in many different cancer types (4246). The PI3K/Akt signaling pathway plays a key role in invasion and metastasis of HCC. It was therefore of significance to investigate whether the PI3K/Akt/Snail-dependent signaling pathway participates in sorafenib-induced EMT. The PI3K/Akt signaling pathway was analyzed in the human HCC SMMC7721 and HCCLM3 cells. Notably, we found that 5 μM sorafenib activated the PI3K/Akt signaling pathway and upregulated the expression of transcription factor Snail. Immunohistochemical staining was then applied to the xenograft marginal tissues. As anticipated, these results were further confirmed in vivo.

In conclusion, the present study showed that sorafenib upregulated the expression of transcription factor Snail and activated the PI3K/AKT signaling pathway. Importantly, these may be associated with sorafenib-induced invasion and metastasis of HCC. Therefore, inhibition of the expression of transcription factor Snail or combined with PI3K/AKT signaling pathway inhibitors may enhance the effectiveness of sorafenib treatment of HCC. Currently, relevant studies are being carried out. The present study may provide the theoretical basis for the combined treatment of sorafenib and PI3K/AKT signaling pathway inhibitors to treat HCC.

Acknowledgements

We thank Te Liu (Shanghai Geriatric Institute of Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai) and Ning Zhang (Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai) for the experimental technical assistance.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

2 

Gao JJ, Song PP, Tamura S, et al: Standardization of perioperative management on hepato-biliary-pancreatic surgery. Drug Discov Ther. 6:108–111. 2012.PubMed/NCBI

3 

Belghiti J and Fuks D: Liver resection and transplantation in hepatocellular carcinoma. Liver Cancer. 1:71–82. 2012. View Article : Google Scholar

4 

Lee Cheah Y and Chow KHP: Liver transplantation for hepatocellular carcinoma: an appraisal of current controversies. Liver Cancer. 1:183–189. 2012.PubMed/NCBI

5 

Cheng AL, Kang YK, Chen Z, et al: Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 10:25–34. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Llovet JM, Ricci S, Mazzaferro V, et al: Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 359:378–390. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Folkman J: Tumor angiogenesis: therapeutic implications. N Engl J Med. 285:1182–1186. 1971. View Article : Google Scholar : PubMed/NCBI

8 

Bergers G and Hanahan D: Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 8:592–603. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG and Kerbel RS: Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell. 15:232–239. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Pàez-Ribes M, Allen E, Hudock J, et al: Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 15:220–231. 2009.PubMed/NCBI

11 

Zhang W, Sun HC, Wang WQ, et al: Sorafenib down-regulates expression of HTATIP2 to promote invasiveness and metastasis of orthotopic hepatocellular carcinoma tumors in mice. Gastroenterology. 143:1641–1649. 2012. View Article : Google Scholar : PubMed/NCBI

12 

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

13 

Barrallo-Gimeno A and Nieto MA: The Snail genes as inducers of cell movement and survival: implications in development and cancer. Development. 132:3151–3161. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Martínez-Alvarez C, Blanco MJ, Pérez R, et al: Snail family members and cell survival in physiological and pathological cleft palates. Dev Biol. 265:207–218. 2004.PubMed/NCBI

15 

Emadi Baygi M, Soheili ZS, Schmitz I, Sameie S and Schulz WA: Snail regulates cell survival and inhibits cellular senescence in human metastatic prostate cancer cell lines. Cell Biol Toxicol. 26:553–567. 2010.PubMed/NCBI

16 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Cano A, Pérez-Moreno MA, Rodrigo I, et al: The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol. 2:76–83. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Zhang AL, Wang QS, Zhong YH, et al: Effect of transcriptional factor snail on epithelial-mesenchymal transition and tumor metastasis. Ai Zheng. 24:1301–1305. 2005.(In Chinese).

19 

Jordà M, Olmeda D, Vinyals A, et al: Upregulation of MMP-9 in MDCK epithelial cell line in response to expression of the Snail transcription factor. J Cell Sci. 118:3371–3385. 2005.PubMed/NCBI

20 

Yokoyama K, Kamata N, Fujimoto R, et al: Increased invasion and matrix metalloproteinase-2 expression by Snail-induced mesenchymal transition in squamous cell carcinomas. Int J Oncol. 22:891–898. 2003.PubMed/NCBI

21 

Liu P, Cheng H, Roberts TM and Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8:627–644. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL and Arteaga CL: Phosphatidylinositol 3-kinase function is required for transforming growth factor β-mediated epithelial to mesenchymal transition and cell migration. J Biol Chem. 275:36803–36810. 2000.

23 

Grille SJ, Bellacosa A, Upson J, et al: The protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 63:2172–2178. 2003.PubMed/NCBI

24 

Altomare DA and Testa JR: Perturbations of the AKT signaling pathway in human cancer. Oncogene. 24:7455–7464. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Larue L and Bellacosa A: Epithelial-mesenchymal transition in development and cancer: role of phosphatidylinositol 3′ kinase/AKT pathways. Oncogene. 24:7443–7454. 2005.

26 

Wang H, Quah SY, Dong JM, Manser E, Tang JP and Zeng Q: PRL-3 down-regulates PTEN expression and signals through PI3K to promote epithelial-mesenchymal transition. Cancer Res. 67:2922–2926. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Song LB, Li J, Liao WT, et al: The polycomb group protein Bmi-1 represses the tumor suppressor PTEN and induces epithelial-mesenchymal transition in human nasopharyngeal epithelial cells. J Clin Invest. 119:3626–3636. 2009. View Article : Google Scholar

28 

Yoo YA, Kang MH, Lee HJ, et al: Sonic hedgehog pathway promotes metastasis via activation of AKT, EMT, and MMP-9 in gastric cancer. Cancer Res. 71:7061–7070. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Zuo JH, Zhu W, Li MY, et al: Activation of EGFR promotes squamous carcinoma SCC10A cell migration and invasion via inducing EMT-like phenotype change and MMP-9-mediated degradation of E-cadherin. J Cell Biochem. 112:2508–2517. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Qiao M, Sheng S and Pardee AB: Metastasis and AKT activation. Cell Cycle. 7:2991–2996. 2008. View Article : Google Scholar

31 

Emadi Baygi M, Soheili ZS, Essmann F, et al: Slug/SNAI2 regulates cell proliferation and invasiveness of metastatic prostate cancer cell lines. Tumour Biol. 31:297–307. 2010.PubMed/NCBI

32 

Bolós V, Peinado H, Pérez-Moreno MA, Fraga MF, Esteller M and Cano A: The transcription factor Slug represses E-cadherin expression and induces epithelial to mesenchymal transitions: a comparison with Snail and E47 repressors. J Cell Sci. 116:499–511. 2003.

33 

Chen KF, Chen HL, Tai WT, et al: Activation of phosphatidylinositol 3-kinase/Akt signaling pathway mediates acquired resistance to sorafenib in hepatocellular carcinoma cells. J Pharmacol Exp Ther. 337:155–161. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Gao Y, Li HX, Xu LT, et al: Bufalin enhances the anti-proliferative effect of sorafenib on human hepatocellular carcinoma cells through downregulation of ERK. Mol Biol Rep. 39:1683–1689. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Wang P, Chen Z, Meng ZQ, et al: Dual role of Ski in pancreatic cancer cells: tumor-promoting versus metastasis-suppressive function. Carcinogenesis. 30:1497–1506. 2009.

36 

Li H, Wang P, Gao Y, et al: Na+/K+-ATPase α3 mediates sensitivity of hepatocellular carcinoma cells to bufalin. Oncol Rep. 25:825–830. 2011.

37 

Yang MH, Chen CL, Chau GY, et al: Comprehensive analysis of the independent effect of twist and snail in promoting metastasis of hepatocellular carcinoma. Hepatology. 50:1464–1474. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Zucchini-Pascal N, Peyre L and Rahmani R: Crosstalk between beta-catenin and snail in the induction of epithelial to mesenchymal transition in hepatocarcinoma: role of the ERK1/2 pathway. Int J Mol Sci. 14:20768–20792. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Zhou BP, Deng J, Xia W, et al: Dual regulation of Snail by GSK-3β-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat Cell Biol. 6:931–940. 2004.PubMed/NCBI

40 

Wen W, Ding J, Sun W, et al: Cyclin G1-mediated epithelial-mesenchymal transition via phosphoinositide 3-kinase/Akt signaling facilitates liver cancer progression. Hepatology. 55:1787–1798. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Shi GM, Ke AW, Zhou J, et al: CD151 modulates expression of matrix metalloproteinase 9 and promotes neoangiogenesis and progression of hepatocellular carcinoma. Hepatology. 52:183–196. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Thiery JP and Sleeman JP: Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 7:13–42. 2006. View Article : Google Scholar

43 

Pérez-Tenorio G and Stål O; Southeast Sweden Breast Cancer Group. Activation of AKT/PKB in breast cancer predicts a worse outcome among endocrine treated patients. Br J Cancer. 86:540–545. 2002.PubMed/NCBI

44 

Scheid MP and Woodgett JR: PKB/AKT: functional insights from genetic models. Nat Rev Mol Cell Biol. 2:760–768. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

46 

Xue G, Restuccia DF, Lan Q, et al: Akt/PKB-mediated phosphorylation of Twist1 promotes tumor metastasis via mediating cross-talk between PI3K/Akt and TGF-β signaling axes. Cancer Discov. 2:248–259. 2012.PubMed/NCBI

Related Articles

Journal Cover

October 2014
Volume 32 Issue 4

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
Wang H, Xu L, Zhu X, Wang P, Chi H and Meng Z: Activation of phosphatidylinositol 3-kinase/Akt signaling mediates sorafenib-induced invasion and metastasis in hepatocellular carcinoma. Oncol Rep 32: 1465-1472, 2014
APA
Wang, H., Xu, L., Zhu, X., Wang, P., Chi, H., & Meng, Z. (2014). Activation of phosphatidylinositol 3-kinase/Akt signaling mediates sorafenib-induced invasion and metastasis in hepatocellular carcinoma. Oncology Reports, 32, 1465-1472. https://doi.org/10.3892/or.2014.3352
MLA
Wang, H., Xu, L., Zhu, X., Wang, P., Chi, H., Meng, Z."Activation of phosphatidylinositol 3-kinase/Akt signaling mediates sorafenib-induced invasion and metastasis in hepatocellular carcinoma". Oncology Reports 32.4 (2014): 1465-1472.
Chicago
Wang, H., Xu, L., Zhu, X., Wang, P., Chi, H., Meng, Z."Activation of phosphatidylinositol 3-kinase/Akt signaling mediates sorafenib-induced invasion and metastasis in hepatocellular carcinoma". Oncology Reports 32, no. 4 (2014): 1465-1472. https://doi.org/10.3892/or.2014.3352