Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma

H Wang, Q Wu, Z Liu, X Luo, Y Fan, Y Liu, Y Zhang, S Hua, Q Fu, M Zhao, Y Chen, W Fang, X Lv, H Wang, Q Wu, Z Liu, X Luo, Y Fan, Y Liu, Y Zhang, S Hua, Q Fu, M Zhao, Y Chen, W Fang, X Lv

Abstract

It is largely recognized that fibroblast activation protein (FAP) is expressed in cancer-associated fibroblasts (CAFs) of many human carcinomas. Furthermore, FAP was recently also reported to be expressed in carcinoma cells of the breast, stomach, pancreatic ductal adenocarcinoma, colorectum, and uterine cervix. The carcinoma cell expression pattern of FAP has been described in several types of cancers, but the role of FAP in oral squamous cell carcinoma (OSCC) is unknown. The role of endogenous FAP in epithelium-derived tumors and molecular mechanisms has also not been reported. In this study, FAP was found to be expressed in carcinoma cells of OSCC and was upregulated in OSCC tissue samples compared with benign tissue samples using immunohistochemistry. In addition, its expression level was closely correlated with overall survival of patients with OSCC. Silencing FAP inhibited the growth and metastasis of OSCC cells in vitro and in vivo. Mechanistically, knockdown of FAP inactivated PTEN/PI3K/AKT and Ras-ERK and its downstream signaling regulating proliferation, migration, and invasion in OSCC cells, as the inhibitory effects of FAP on the proliferation and metastasis could be rescued by PTEN silencing. Our study suggests that FAP acts as an oncogene and may be a potential therapeutic target for patients with OSCC.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
FAP expression in oral squamous cell carcinoma (OSCC) is associated with poor prognosis. (I) FAP expression examination in OSCC and control tissues (a) Negative FAP expression in oral cavity epithelium; (b, c) Negative expression of FAP in OSCC tissues; (d, e) Weakly positive expression of FAP in OSCC tissues; (f, g, and h) Positive and strong positive expression of FAP in OSCC tissues; (II) Immunofluorescent images of KB and Tca-8113 cells stained for FAP (green) and 4′,6-diamidino-2-phenylindole (DAPI; blue) (magnification, × 400); (III) Kaplan–Meier survival analysis of overall survival in all patients. The log-rank test was used to calculate P-values
Figure 2
Figure 2
Stable suppression of FAP attenuates the growth of OSCC cells in vitro and vivo. (a) Stably knocking down decreased the expression of FAP in shRNA-FAP-1 and -2 cells compared with shRNA-con cells by real-time PCR and western blotting. β-Actin was used as a loading control. (b) MTT cell viability assays were performed on days 1–6 of KB cells stably expressing the shRNA-FAP-1 and 2 or shRNA-con vectors. (c) Colony formation assay was performed on KB cells stably expressing the shRNA-FAP-1 and -2 or shRNA-con vectors for 2 weeks. (d) Cell-cycle analysis of KB cells stably expressing the shRNA-FAP-1 and 2 or shRNA-con vectors. (e) External whole-body fluorescence images of KB/shRNA-FAP-1 and -2 and KB/shRNA-con mice obtained 30 days after injection. Representative photographs of H&E staining of primary cancer tissues are shown. Data are presented as mean±S.D. for three independent experiments **P<0.01; ***P<0.001, compared with control
Figure 3
Figure 3
Transient suppression of FAP inhibits growth of OSCC cells in vitro. (a) Transiently knocking down FAP by siRNA reduced the expression of FAP by real-time PCR and western blotting. (b) MTT cell viability assays was performed on days 1–5 after the transfection of KB and Tca-8113 cells with either siRNA FAP-1 or the negative control. (c) Cell-cycle profiles were analyzed after KB and Tca-8113 cells were transfected with siRNA FAP-1 or the negative control for 48 h. Data are presented as mean±S.D for three independent experiments *P<0.05; **P<0.01; ***P<0.001, compared with control
Figure 4
Figure 4
Stable depletion of FAP suppresses cell migration and invasion in vitro and vivo. (a) Wound-healing assay showed KB cells stably expressing the shRNA-FAP-1 and -2 or shRNA-con vectors at 0 and 48 h after wounding. Bar chart showing the relative migration ability at 48 h. (b) Cell invasion assay showed decreased invasion from cells transfected with shRNA-FAP-1 and -2, with representative fields shown. (c) The ability of the cells to adhere to a solid surface was significantly increased in cells treated with shRNA-FAP-1 and -2, and a representative experiment is shown. Bar chart showing the adhesive rate quantified by MTT assay. (d). Phalloidin staining of F-actin in KB/shRNA-FAP-1 and KB/shRNA-con cells (magnification, × 1000). (e) External whole-body fluorescence images of liver were obtained 2 months after spleen injection, respectively. Metastatic cancer tissue (H&E staining, magnification, × 200). Data shown are means±S.D. of at least three independent experiments *P<0.05; **P<0.01, compared with control
Figure 5
Figure 5
Transient depletion of FAP reduces cell migration and invasion in vitro. (a) Transiently downregulated FAP dramatically inhibited KB and Tca-8113 cell migration in vitro. (b) Transiently suppressed FAP decreased in vitro invasiveness of KB and Tca-8113 cells. (c) Transient knockdown of FAP elevated the adhesion rate of KB and Tca-8113 cells. Data are presented as means±S.D. of at least three independent experiments *P<0.05; **P<0.01, compared with control
Figure 6
Figure 6
FAP regulates the expression of cell cycle, MMPs, and EMT-associated genes in OSCC through PTEN/PI3K/Akt and ERK signaling pathways. (a) Knocking down endogenous FAP expression reduced the activation of pRb(ser780), an oncogenic cell-cycle regulator including CCNE1, E2F1, and c-Myc, and elevated the expression of tumor suppressors including p27 and p21. However, the expression of CDK4, CCND1, and total Rb was not affected. (b) Suppressing FAP expression decreased the expression of MMP2, MMP9, and EMT-marker genes including Snail, Slug, N-cadherin, and Vimentin but increased E-cadherin expression. (c) Reduced FAP significantly decreased the expression of phosphorylated PI3K, AKT, MEK1/2, ERK1/2, and GSK3β, whereas total levels remained unchanged. β-Actin was used as a loading control
Figure 7
Figure 7
PTEN depletion or gain respectively elevates or decreases proliferation, migration, and invasion of cells with downregulated FAP. (a) Decreased PTEN mRNA and protein levels in KB cells with stably silenced FAP and transfected with siRNA were found by real-time PCR and western blotting. (b) MTT cell viability assays were performed on days 1–5 after the transfection of FAP knockdown KB cells with either siRNA PTEN-1 and -2 or the control. (c) Transiently downregulated PTEN dramatically enhanced the ability of KB cells treated with shRNA-FAP-1 invasion in vitro. (d) Transiently suppressed PTEN elevated in vitro migration of KB cells that had stable downregulated FAP. (e) MTT cell viability assays were performed on days 1–4 after the transfection of FAP knockdown KB cells with either the wild type (pEGFP-wt-PTEN) and the mutant type (pEGFP-G129R-PTEN) or empty vector (pEGFP-C1). (f) Introduction of PTEN decreased invasion of KB cells treated with shRNA-FAP-1. Data are presented as mean±S.D. for three independent experiments *P<0.05; **P<0.01; ***P<0.001, compared with control

References

    1. Markopoulos AK. Current aspects on oral squamous cell carcinoma. Open Dent J 2012; 6: 126–130.
    1. Jian SL, Hsieh HY, Liao CT et al. Galpha(1)(2) drives invasion of oral squamous cell carcinoma through up-regulation of proinflammatory cytokines. PLoS One 2013; 8: e66133.
    1. Albuquerque R, Lopez-Lopez J, Mari-Roig A et al. Oral tongue squamous cell carcinoma (OTSCC): alcohol and tobacco consumption versus non-consumption. A study in a Portuguese population. Braz Dent J 2011; 22: 517–521.
    1. Chaudhary A, Pandya S, Singh M et al. Identification of high-risk human papillomavirus-16 and -18 infections by multiplex PCR and their expression in oral submucous fibrosis and oral squamous cell carcinoma. Head Neck Oncol 2013; 5: 4.
    1. Hsieh CH, Chang JW, Hsieh JJ et al. Epidermal growth factor receptor mutations in patients with oral cavity cancer in a betel nut chewing-prevalent area. Head Neck 2011; 33: 1758–1764.
    1. Reis PP, Tomenson M, Cervigne NK et al. Programmed cell death 4 loss increases tumor cell invasion and is regulated by miR-21 in oral squamous cell carcinoma. Mol Cancer 2010; 9: 238.
    1. Li C, Fan J, Song X et al. Expression of angiopoietin-2 and vascular endothelial growth factor receptor-3 correlates with lymphangiogenesis and angiogenesis and affects survival of oral squamous cell carcinoma. PLoS One 2013; 8: e75388.
    1. Yang W, Han W, Ye S et al. Fibroblast activation protein-alpha promotes ovarian cancer cell proliferation and invasion via extracellular and intracellular signaling mechanisms. Exp Mol Pathol 2013; 95: 105–110.
    1. Lee HO, Mullins SR, Franco-Barraza J et al. FAP-overexpressing fibroblasts produce an extracellular matrix that enhances invasive velocity and directionality of pancreatic cancer cells. BMC Cancer 2011; 11: 245.
    1. Kelly T, Kechelava S, Rozypal TL et al. Seprase, a membrane-bound protease, is overexpressed by invasive ductal carcinoma cells of human breast cancers. Mod Pathol 1998; 11: 855–863.
    1. Shi M, Yu DH, Chen Y et al. Expression of fibroblast activation protein in human pancreatic adenocarcinoma and its clinicopathological significance. World J Gastroenterol 2012; 18: 840–846.
    1. Mori Y, Kono K, Matsumoto Y et al. The expression of a type II transmembrane serine protease (Seprase) in human gastric carcinoma. Oncology 2004; 67: 411–419.
    1. Iwasa S, Okada K, Chen WT et al. 'Increased expression of seprase, a membrane-type serine protease, is associated with lymph node metastasis in human colorectal cancer'. Cancer Lett 2005; 227: 229–236.
    1. Jin X, Iwasa S, Okada K et al. Expression patterns of seprase, a membrane serine protease, in cervical carcinoma and cervical intraepithelial neoplasm. Anticancer Res 2003; 23: 3195–3198.
    1. Peyton SR, Ghajar CM, Khatiwala CB et al. The emergence of ECM mechanics and cytoskeletal tension as important regulators of cell function. Cell Biochem Biophys 2007; 47: 300–320.
    1. Turley EA, Veiseh M, Radisky DC et al. Mechanisms of disease: epithelial-mesenchymal transition–does cellular plasticity fuel neoplastic progression? Nat Clin Pract Oncol 2008; 5: 280–290.
    1. Sarrio D, Rodriguez-Pinilla SM, Hardisson D et al. Epithelial-mesenchymal transition in breast cancer relates to the basal-like phenotype. Cancer Res 2008; 68: 989–997.
    1. Cross DA, Alessi DR, Cohen P et al. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995; 378: 785–789.
    1. Cohen P, Frame S. The renaissance of GSK3. Nat Rev Mol Cell Biol 2001; 2: 769–776.
    1. Hua X, Yu L, Huang X et al. Expression and role of fibroblast activation protein-alpha in microinvasive breast carcinoma. Diagn Pathol 2011; 6: 111.
    1. Wikberg ML, Edin S, Lundberg IV et al. High intratumoral expression of fibroblast activation protein (FAP) in colon cancer is associated with poorer patient prognosis. Tumour Biol 2013; 34: 1013–1020.
    1. Cohen SJ, Alpaugh RK, Palazzo I et al. Fibroblast activation protein and its relationship to clinical outcome in pancreatic adenocarcinoma. Pancreas 2008; 37: 154–158.
    1. Liao Y, Ni Y, He R et al. Clinical implications of fibroblast activation protein-alpha in non-small cell lung cancer after curative resection: a new predictor for prognosis. J Cancer Res Clin Oncol 2013; 139: 1523–1528.
    1. Huang L, Wang HY, Li JD et al. KPNA2 promotes cell proliferation and tumorigenicity in epithelial ovarian carcinoma through upregulation of c-Myc and downregulation of FOXO3a. Cell Death Dis 2013; 4: e745.
    1. Romano S, Staibano S, Greco A et al. FK506 binding protein 51 positively regulates melanoma stemness and metastatic potential. Cell Death Dis 2013; 4: e578.
    1. Hanai JI, Doro N, Seth P et al. ATP citrate lyase knockdown impacts cancer stem cells in vitro. Cell Death Dis 2013; 4: e696.
    1. Saxena M, Stephens MA, Pathak H et al. Transcription factors that mediate epithelial-mesenchymal transition lead to multidrug resistance by upregulating ABC transporters. Cell Death Dis 2011; 2: e179.
    1. He L, Zhou X, Qu C et al. Musashi2 predicts poor prognosis and invasion in hepatocellular carcinoma by driving epithelial-mesenchymal transition. J Cell Mol Med 2014; 18: 49–58.
    1. Zhao XL, Sun T, Che N et al. Promotion of hepatocellular carcinoma metastasis through matrix metalloproteinase activation by epithelial-mesenchymal transition regulator Twist1. J Cell Mol Med 2011; 15: 691–700.
    1. Van Tubergen EA, Banerjee R, Liu M et al. Inactivation or loss of TTP promotes invasion in head and neck cancer via transcript stabilization and secretion of MMP9, MMP2, and IL-6. Clin Cancer Res 2013; 19: 1169–1179.
    1. Liu J, van Mil A, Aguor EN et al. MiR-155 inhibits cell migration of human cardiomyocyte progenitor cells (hCMPCs) via targeting of MMP-16.J. Cell Mol Med 2012; 16: 2379–2386.
    1. Song G, Ouyang G, Mao Y et al. Osteopontin promotes gastric cancer metastasis by augmenting cell survival and invasion through Akt-mediated HIF-1alpha up-regulation and MMP9 activation. J Cell Mol Med 2009; 13: 1706–1718.
    1. Bullock MD, Pickard KM, Nielsen BS et al. Pleiotropic actions of miR-21 highlight the critical role of deregulated stromal microRNAs during colorectal cancer progression. Cell Death Dis 2013; 4: e684.
    1. 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 2012; 55: 1787–1798.
    1. Ye Y, Xiao Y, Wang W et al. ERalpha signaling through slug regulates E-cadherin and EMT. Oncogene 2010; 29: 1451–1462.
    1. Han L, Peng B, Ma Q et al. Indometacin ameliorates high glucose-induced proliferation and invasion via modulation of e-cadherin in pancreatic cancer cells. Curr Med Chem 2013; 20: 4142–4152.
    1. Dong-Dong L, Xi-Ran Z, Xiang-Rong C. Expression and significance of new tumor suppressor gene PTEN in primary liver cancer. J Cell Mol Med 2003; 7: 67–71.
    1. Luo H, Yang Y, Duan J et al. PTEN-regulated AKT/FoxO3a/Bim signaling contributes to reactive oxygen species-mediated apoptosis in selenite-treated colorectal cancer cells. Cell Death Dis 2013; 4: e481.
    1. Liu Z, Luo W, Zhou Y et al. Potential tumor suppressor NESG1 as an unfavorable prognosis factor in nasopharyngeal carcinoma. PLoS One 2011; 6: e27887.
    1. Yu X, Zhen Y, Yang H et al. Loss of connective tissue growth factor as an unfavorable prognosis factor activates miR-18b by PI3K/AKT/C-Jun and C-Myc and promotes cell growth in nasopharyngeal carcinoma. Cell Death Dis 2013; 4: e634.

Source: PubMed

3
Tilaa