Fibroblast activation protein in osteosarcoma cells promotes angiogenesis via AKT and ERK signaling pathways

  • Authors:
    • Chao Zeng
    • Ming Wen
    • Xiaomei Liu
  • View Affiliations

  • Published online on: February 12, 2018     https://doi.org/10.3892/ol.2018.8027
  • Pages: 6029-6035
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Although it is established as a marker of cancer-associated fibroblasts, the expression of fibroblast activation protein (FAP) is not restricted to stromal cells; its expression in multiple types of tumor cell and its pro‑tumor functions have been reported. However, the role of FAP in angiogenesis in osteosarcoma remains uncharacterized. In the present study, it was identified that the mRNA and protein expression levels of FAP and vascular endothelial growth factor‑A (VEGF‑A) corresponded to each other in MG63, U2‑OS and HOS osteosarcoma cells. Subsequent to upregulating FAP in MG63 cells, VEGF‑A mRNA and protein expression significantly increased; subsequent to downregulating FAP in U2‑OS cells, VEGF‑A mRNA and protein expression significantly declined. These changes in VEGF‑A level were also detected in the cell supernatant with ELISA. Conditioned medium (CM) from MG63 cells overexpressing FAP promoted the phosphorylation of AKT and extracellular signal‑regulated kinase (ERK) in human umbilical vein endothelial cells (HUVECs), as well as the proliferation rate. The CM from U2‑OS cells with FAP knockdown inhibited the proliferation rate of HUVECs. The phosphorylation of AKT and ERK was increased in MG63 cells overexpressing FAP, but reduced in U2‑OS cells with FAP knockdown. Furthermore, treatment with the AKT inhibitor LY294002 or the ERK inhibitor U0126 inhibited the upregulation of VEGF‑A induced by FAP expression. Collectively, the results suggest that FAP expression in osteosarcoma cells promotes angiogenesis.

Introduction

Osteosarcoma, which is derived from primitive bone-forming mesenchymal cells, is the most common type of malignant bone tumor, and predominantly occurs in children and young adults (1). Despite recent developments in medical technology, including surgery and chemotherapy, therapy outcomes remain unsatisfactory; this can be partially attributed to the complex interaction between the tumor cells and the tumor microenvironment (24).

Blood vessels are critical component of the tumor microenvironment, as they are not only essential for supplying tumor nutrition, but also for transporting tumor cells to distant sites (5). A growing body of cancer research is focusing on angiogenesis. A range of types of drugs targeting tumor vessels have been developed and applied clinically. Although such drugs improve the treatment outcome to some extent, drug resistance is ultimately induced during treatment, and so therapy outcomes remain unsatisfactory (6,7). Further endeavor is required to identify more effective targets for anti-angiogenesis therapy and improve the prognosis for cancer.

Fibroblast activation protein (FAP) was initially identified as a specific marker for cancer-associated fibroblasts (8). It was previously reported that stromal FAP expression was correlated with the progression and prognosis in a range of tumor types, including colon, breast and ovarian cancer (912). In view of the role of stromal FAP in cancer progression, its expression in tumor cells has attracted increasing attention. Mentlein et al (13) reported that FAP was highly expressed on the surface of bone and soft tissue tumor cells. Yuan et al (14) reported that the high expression of FAP in osteosarcoma cells was positively correlated with an advanced clinical stage, high histological grade, positive metastasis and poor prognosis. However, the role of FAP in angiogenesis in osteosarcoma has yet to be characterized.

In the present study, it was identified that FAP expression in osteosarcoma cells promoted the expression of vascular endothelial growth factor-A (VEGF-A). Furthermore, conditioned medium (CM) from osteosarcoma cells with altered FAP expression could activate AKT, ERK and proliferation in endothelial cells. Finally, it was demonstrated that the activation of the AKT and extracellular signal-regulated kinase (ERK) signaling pathways was associated with the VEGF-A expression induced by FAP.

Materials and methods

Cell lines and cell culture

HOS, U2-OS and MG63 human osteosarcoma cell lines, and human umbilical vein endothelial cells (HUVECs) were purchased from American Type Culture Collection (Manassas, VA, USA). HOS, U2-OS and MG63 cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc.). HUVEC cells were cultured in endothelial cell medium supplemented with 5% FBS and 1% endothelial cell growth supplement (all from ScienCell Research Laboratories, Carlsbad, CA, USA). All cells were cultured in 5% CO2 at 37°C. To study potential molecular mechanism, the AKT inhibitor LY294002 or ERK inhibitor U0126 were added into osteosarcoma cells using FAP overexpression for 24 h at 37°C. The final concentrations of LY294002 and ERK were 50 and 20 µM, respectively.

Transfection

A total of 2×105 cells were plated in 6-well plates. After 24 h, 1.5 µg p-cDNA3.1/FAP-vector and 2 µl Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) were mixed for 20 min at room temperature. Then, the mixture was added in cells in order to overexpress FAP. A total of 1.5 µg p-GPU6/FAP-shRNA plasmids (Shanghai GenePharma Co., Ltd., Shanghai, China) and 2 µl Lipofectamine® 2000 were mixed for 20 min at room temperature. Then, the mixture was added in cells to silence FAP expression. The empty plasmids were used as a control. After 48 h, RNA and protein were extracted for further procedures. Subsequent to culture in DMEM without FBS for a further 48 h, the supernatant was collected as CM. The shRNA sequence is 5′-CCCTCAGACAGTTTGCTTATT-3′.

Reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA from cells was extracted with TRIzol (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. cDNA was synthesized using the PrimeScript RT-PCR kit (Takara Biotechnology Co., Ltd., Dalian, China) according to the manufacturer's protocol (25°C for 10 min, 42°C for 60 min, then 70°C for 10 min). The PCR was performed using specific primers and Universal PCR Master Mix (Thermo Fisher Scientific, Inc.) with the following thermocycler conditions: 4°C for 5 min, then 94°C for 30 sec, 57°C for 30 sec and 72°C for 30 sec, followed by a holding step at 72°C for 5 min. For the FAP PCR, this was repeated for 40 cycles. For the VEGF-A and GAPDH PCR, this was repeated for 36 cycles. PCR products were electrophoretically separated on 1.0% agarose gels. The results were analyzed by Labwork software, version 4.0 (UVP, Inc., Upland, CA, USA). The primers used were as follows: FAP forwards, 5′-TTAGTCTGACAAAGAGAAACACTG-3′ and reverse, 5′-ATGAAGACTTGGGTAAAAATCG-3′; VEGF-A forwards, 5′-CGGGCAGGAGGAAGGAGCCT-3′ and reverse, 5′-GTGATGGTGTGGTGGCGGCA-3′; GAPDH forward, 5′-AGAAGGCTGGGGCTCATTTG-3′ and reverse, 5′-AGGGGCCATCCACAGTCTTC-3′ GAPDH was used as an internal control.

Western blot analysis

Protein was extracted using radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Haimen, China) containing 1% protease inhibitor. Total protein was measured using a bicinchoninic acid assay kit (Pierce; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. Then, 300 µg/well protein was loaded in 5% acrylamide and separated by 10% separating gel, then transferred to a nitrocellulose membrane. Subsequent to blocking in TBS with 0.05% Tween-20 (ST825, Beyotime Institute of Biotechnology) containing 5% non-fat dried milk for 1 h at room temperature, the membrane was then incubated with primary antibodies (listed in Table I) at 4°C overnight, followed by incubation with peroxidase-linked goat anti-rabbit-IgG (cat. no. ab205718, dilution, 1:4,000; Abcam, Cambridge, MA, USA) at room temperature for 1 h. Signals were detected using enhanced chemiluminescence reagents (Pierce; Thermo Fisher Scientific, Inc.) and measured using Image-Pro software (version 5.1; Media Cybernetics, Inc., Rockville, MA, USA).

Table I.

Primary antibodies used for western blotting in the present study.

Table I.

Primary antibodies used for western blotting in the present study.

TargetSpecificityDilutionCat. no.Company
FAPRabbit anti-human1:1,000ab53066Abcam, Cambridge, MA, USA
VEGF-ARabbit anti-human1:1,000ab46154Abcam
Phospho-ERKRabbit anti-human1:1,0004370Cell Signaling Technology, Inc., Danvers, MA, USA
ERKRabbit anti-human1:1,0004695Cell Signaling Technology, Inc.
Phospho-AKTRabbit anti-human1:1,0004058Cell Signaling Technology, Inc.
AKTRabbit anti-human1:1,0004691Cell Signaling Technology, Inc.
GAPDHRabbit anti-human1:4,000ab128915Abcam
ELISA

VEGF-A in CM was detected using PGRN ELISA kit (cat. no. CSB-E11718h; CUSABIO Life Science, Wuhan, China) and VEGF-A ELISA Kit (CUSABIO Life Science) according to the manufacturer's instructions. The experiment was repeated at least three times.

HUVEC cell proliferation assay

HUVEC cells were plated in a 96-well plate at 5,000 cells per well, in triplicate. Once the cells had adhered to the well walls, the medium was replaced with CM from transfected MG63 or U2-OS cells. An MTT assay was performed at 0 and 48 h from the addition of CM. Cells were incubated with MTT for 4 h, then formazan was dissolved with 0.1% dimethyl sulfoxide. The absorbance of the wells was then measured at 490 nm. The proliferation rate was calculated as OD at 48 h/OD at 0 h.

Statistical analysis

Data was expressed as the mean ± standard deviation. SPSS software (version 11.0; SPSS, Inc., Chicago, IL, USA) was used for statistical analysis. Differences between groups were analyzed using one-way analysis of variance with Dunnett's post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

FAP and VEGF-A expression in osteosarcoma cells

RT-PCR and western blot analysis were performed to detect the expression of FAP and VEGF-A in osteosarcoma cells. As demonstrated in Fig. 1, U2-OS cells exhibited the highest expression of FAP at the mRNA (Fig. 1A and B) and protein (Fig. 1C and D) levels, which was significantly different from MG63 and HOS cells (P<0.05). MG63 cells exhibited the lowest FAP expression at the mRNA (Fig. 1A and B) and protein (Fig. 1C and D) levels, which were significantly different from U2-OS and HOS cells (P<0.05). The VEGF-A expression trend among these three cell lines was consistent with FAP expression at the mRNA (Fig. 1A and B) and protein (Fig. 1C and D) levels. This indicated a potential association between FAP and VEGF-A expression in osteosarcoma cells.

FAP promotes VEGF-A expression in osteosarcoma cells

To identify whether an association between FAP and VEGF-A existed, FAP expression was altered by transfection. As demonstrated in Fig. 2, when FAP in MG63 cells was overexpressed, VEGF-A expression was upregulated significantly at the mRNA (Fig. 2A and B) and protein (Fig. 2C and D) levels (P<0.05). Conversely, subsequent to silencing FAP expression in U2-OS cells, VEGF-A expression was significantly downregulated at the mRNA (Fig. 2E and F) and protein (Fig. 2G and H) levels. In addition, when FAP in MG63 cells was overexpressed, an increase in the secreted VEGF-A in supernatant was also identified using ELISA (Fig. 2I). When FAP in MG63 cells was silenced, a decline in the VEGF-A in supernatant was also identified with ELISA (Fig. 2J). This suggested that FAP could promote VEGF-A expression in osteosarcoma cells, implying its pro-angiogenic properties.

CM from osteosarcoma cells with alterations in FAP expression affects the angiogenic properties of HUVEC cells

To further confirm the effect of FAP on angiogenesis, the changes in the angiogenic properties of HUVEC cells were directly assessed. As demonstrated in Fig. 3, subsequent to treatment with CM from MG63 cells with over-expressed FAP, compared with the control group, the phosphorylation of AKT and ERK of HUVEC cells were activated (Fig. 3A). The proliferation rate of the HUVECs increased from 0.89±0.13 to 1.32±0.08, representing a significant difference (Fig. 3B; P<0.05). Conversely, subsequent to treatment with CM from U2-OS cells with silenced FAP for 24 h, the proliferation rate of HUVEC cells declined from 0.92±0.07 to 0.44±0.07, a significant difference (Fig. 3C; P<0.05). This further demonstrated that FAP expression promoted angiogenesis.

Phosphoinositide 3-kinase (PI3K)/AKT and ERK signaling pathways are required for the VEGF-A upregulation induced by FAP

To further investigate the potential molecular mechanisms of FAP in angiogenesis, AKT and ERK were selected as candidate targets. A western blot demonstrated that MG63 cells over-expressing FAP exhibited the activation (phosphorylation) of AKT and ERK (Fig. 4A), whereas in U2-OS cells with silenced FAP expression, the phosphorylation of AKT and ERK were inhibited (Fig. 4B), implying that AKT and ERK may be downstream targets of FAP. Subsequent to treatment with an ERK inhibitor (U0126) or AKT inhibitor (LY294002), the upregulation of VEGF-A expression induced by FAP overexpression was abrogated (Fig. 4C and D). These results collectively suggest that FAP regulated VEGF-A expression in osteosarcoma cells via the PI3K/AKT and ERK signaling pathways.

Discussion

Angiogenesis is a complicated process by which new vessels form from existing vasculature. This process is controlled by the balance between proangiogenic factors and antiangiogenic factors (7). VEGF-A is a crucial proangiogenic factor that is implicated in the carcinogenesis of multiple types of tumor (15). It stimulates signaling pathways in endothelial cells through binding to vessel endothelial growth factor receptor 2 (VEGFR-2) to enhance the proliferation, migration and tubule formation abilities of the cells, thus promoting angiogenesis (1618).

FAP, initially emerging as a specific marker of reactive fibroblasts in tumors, is a 170-kDa homodimeric glycoprotein composed of two 97-kDa subunits (19). It belongs to the serine protease family and has dipeptidyl peptidase and endopeptidase activity, allowing the degradation of gelatin and type I collagen (11). The roles of FAP in tumor progression have been widely studied. Jia et al (20) identified that overexpressing FAP significantly promoted the growth and motility of MDA-MB-213 human breast cancer cells in vitro. Shi et al (21) reported that FAP was highly expressed in carcinoma cells, and was associated with desmoplasia and a worse prognosis in pancreatic ductal adenocarcinoma. In the present study, FAP and VEGF-A expression levels were determined in MG63, U2-OS and HOS cells. It was identified that FAP expression was consistent with VEGF-A expression in the different osteosarcoma cell lines. MG63 cells exhibited the lowest expression of FAP and VEGF-A; U2-OS cells exhibited the highest expression. This may imply that FAP expression is correlated with VEGF-A expression. According to the FAP expression level, MG63 and U2-OS cells were selected for further research. Following the silencing of FAP expression in U2-OS cells, VEGF-A expression was significantly downregulated; likewise, over-expressing FAP significantly upregulated the expression of VEGF-A in MG63 cells. To the best of our knowledge, this confirmed the effect of FAP on promoting the expression of pro-angiogenic factors for the first time, further implying its pro-angiogenic role in osteosarcoma.

Vessel endothelial cells line the inner walls of blood vessels and serve a critical role in angiogenesis (22). Under the control of angiogenic factors derived from multiple types of cell, including pericytes and tumor cells, vessel endothelial cells began to proliferate and migrate, resulting in angiogenesis (7,23). In the present study, it was identified that VEGF-A in the supernatant was altered with changes in the FAP expression of osteosarcoma cells. Subsequent to culture in CM from MG63 cells overexpressing FAP, the AKT and ERK signal pathways in HUVEC cells were activated, and the proliferation rate was increased. Furthermore, CM from U2-OS cells with silenced FAP expression inhibited the proliferation rate of HUVEC cells. This demonstrated that the expression of FAP in osteosarcoma cells could, at least in part, promote angiogenesis through regulating the expression of VEGF-A. Whether other angiogenic factors may also be induced by FAP requires further investigation.

The phosphorylation of AKT and ERK has been reported in multiple cancers. They are members of critical signal pathways for maintaining the malignancy of tumor cells, and affect a range of biological behaviors in tumor cells, including proliferation, migration, differentiation, drug resistance, apoptosis and phenotype maintenance (2426). Liu et al (27) reported that microRNA-21 induced angiogenesis through the activation of AKT and ERK in DU145 human prostate cancer cells. Wang et al (28) identified that the downregulation of FAP suppressed cell proliferation and metastasis through the PI3K/AKT and ERK signaling pathways in oral squamous cell carcinoma. In the present study, it was identified that the phosphorylation of AKT and ERK were activated by FAP. The Akt inhibitor, LY294002, and the ERK inhibitor, U0126, each abrogated the upregulation of VEGF-A induced by FAP. This is consistent with previous studies (27,28). However, as there is complexity in the crosstalk between different signaling pathways, although the activation of AKT and ERK were confirmed in the present study, the detailed mechanism of regulation has yet to be fully elucidated.

In conclusion, FAP and VEGF-A expression levels corresponded with each other in osteosarcoma cells. The overexpression of FAP in osteosarcoma cells activated the PI3K/AKT and ERK signaling pathways, which may promote angiogenesis. This data may enrich the understanding of the tumor-promoting role of FAP, and supplies evidence to support the identification of novel therapy targets against osteosarcoma.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article

Authors' contributions

CZ designed the present study and wrote the paper; MW performed the cell experiments and XL performed the cell experiments and data analysis.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Savage SA, Mirabello L, Wang Z, Gastier-Foster JM, Gorlick R, Khanna C, Flanagan AM, Tirabosco R, Andrulis IL, Wunder JS, et al: Genome-wide association study identifies two susceptibility loci for osteosarcoma. Nat Genet. 45:799–803. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Bonuccelli G, Avnet S, Grisendi G, Salerno M, Granchi D, Dominici M, Kusuzaki K and Baldini N: Role of mesenchymal stem cells in osteosarcoma and metabolic reprogramming of tumor cells. Oncotarget. 5:7575–7588. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Lu J, Song G, Tang Q, Zou C, Han F, Zhao Z, Yong B, Yin J, Xu H, Xie X, et al: IRX1 hypomethylation promotes osteosarcoma metastasis via induction of CXCL14/NF-κB signaling. J Clin Invest. 125:1839–1856. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Garcia A and Singh H: Bevacizumab and ovarian cancer. Ther Adv Med Oncol. 5:133–141. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Bergers G and Benjamin LE: Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 3:401–410. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Rusckowski M, Wang Y, Blankenberg FG, Levashova Z, Backer MV and Backer JM: Targeted scVEGF/(177)Lu radiopharmaceutical inhibits growth of metastases and can be effectively combined with chemotherapy. EJNMMI Res. 6:42016. View Article : Google Scholar : PubMed/NCBI

7 

Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT and De Bruijn EA: Vascular endothelial growth factor and angiogenesis. Pharmacol Rev. 56:549–580. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Zi FM, He JS, Li Y, Wu C, Wu WJ, Yang Y, Wang LJ, He DH, Yang L, Zhao Y, et al: Fibroblast activation protein protects bortezomib-induced apoptosis in multiple myeloma cells through β-catenin signaling pathway. Cancer Biol Ther. 15:1413–1422. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Wikberg ML, Edin S, Lundberg IV, Van Guelpen B, Dahlin AM, Rutegård J, Stenling R, Oberg A and Palmqvist R: High intratumoral expression of fibroblast activation protein (FAP) in colon cancer is associated with poorer patient prognosis. Tumor Biol. 34:1013–1020. 2013. View Article : Google Scholar

10 

Hua X, Yu L, Huang X, Liao Z and Xian Q: Expression and role of fibroblast activation protein-alpha in microinvasive breast carcinoma. Diagn Pathol. 6:1112011. View Article : Google Scholar : PubMed/NCBI

11 

Liu R, Li H, Liu L, Yu J and Ren X: Fibroblast activation protein. Cancer Biol Ther. 13:123–129. 2014. View Article : Google Scholar

12 

Lai D, Ma L and Wang F: Fibroblast activation protein regulates tumor-associated fibroblasts and epithelial ovarian cancer cells. Int J Oncol. 41:541–550. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Mentlein R, Hattermann K, Hemion C, Jungbluth AA and Held-Feindt J: Expression and role of the cell surface protease seprase/fibroblast activation protein-α (FAP-α) in astroglial tumors. Biol Chem. 392:199–207. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Yuan D, Liu B, Liu K, Zhu G, Dai Z and Xie Y: Overexpression of fibroblast activation protein and its clinical implications in patients with osteosarcoma. J Surg Oncol. 108:157–162. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Zhao D, Pan C, Sun J, Gilbert C, Drews-Elger K, Azzam DJ, Picon-Ruiz M, Kim M, Ullmer W, El-Ashry D, et al: VEGF drives cancer-initiating stem cells through VEGFR-2/Stat3 signaling to upregulate Myc and Sox2. Oncogene. 34:3107–3119. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Ishigami SI, Arii S, Furutani M, Niwano M, Harada T, Mizumoto M, Mori A, Onodera H and Imamura M: Predictive value of vascular endothelial growth factor (VEGF) in metastasis and prognosis of human colorectal cancer. Br J Cancer. 78:1379–1384. 1998. View Article : Google Scholar : PubMed/NCBI

17 

Neufeld G, Cohen T, Gengrinovitch S and Poltorak Z: Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 13:9–22. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Cross MJ, Dixelius J, Matsumoto T and Claesson-Welsh L: VEGF-receptor signal transduction. Trends Biochem Sci. 28:488–494. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Kelly T, Huang Y, Simms AE and Mazur A: Fibrobl Activation Protein-α: A key modulator of the microenvironment in multiple pathologies. Int Rev Cell Mol Biol. 297:83–116. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Jia J, Martin TA, Ye L and Jiang WG: FAP-α (Fibroblast activation protein-α) is involved in the control of human breast cancer cell line growth and motility via the FAK pathway. BMC Cell Biol. 15:162014. View Article : Google Scholar : PubMed/NCBI

21 

Shi M, Yu DH, Chen Y, Zhao CY, Zhang J, Liu QH, Ni CR and Zhu MH: Expression of fibroblast activation protein in human pancreatic adenocarcinoma and its clinicopathological significance. World J Gastroenterol. 18:840–846. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Adeoye OO, Bouthors V, Hubbell MC, Williams JM and Pearce WJ: VEGF receptors mediate hypoxic remodeling of adult ovine carotid arteries. J Appl Physiol (1985). 117:777–787. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Wu Y, Huang A, Li T, Su X, Ding H, Li H, Qin X, Hou L, Zhao Q, Ge X, et al: MiR-152 reduces human umbilical vein endothelial cell proliferation and migration by targeting ADAM17. FEBS Lett. 588:2063–2069. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Steelman LS, Chappell WH, Abrams SL, Kempf RC, Long J, Laidler P, Mijatovic S, Maksimovic-Ivanic D, Stivala F, Mazzarino MC, et al: Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging (Albany NY). 3:192–222. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Sasore T and Kennedy B: Deciphering combinations of PI3K/AKT/mTOR pathway drugs augmenting anti-angiogenic efficacy in vivo. PLoS One. 9:e1052802014. View Article : Google Scholar : PubMed/NCBI

26 

Khan KH, Yap TA, Yan L and Cunningham D: Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin J Cancer. 32:253–265. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Liu LZ, Li C, Chen Q, Jing Y, Carpenter R, Jiang Y, Kung HF, Lai L and Jiang BH: miR-21 induced angiogenesis through AKT and ERK Activation and HIF-1α expression. PLoS One. 6:e191392011. View Article : Google Scholar : PubMed/NCBI

28 

Wang H, Wu Q, Liu Z, Luo X, Fan Y, Liu Y, Zhang Y, Hua S, Fu Q, Zhao M, et al: Downregulation of FAP suppresses cell proliferation and metastasis through PTEN/PI3K/AKT and Ras-ERK signaling in oral squamous cell carcinoma. Cell Death Dis. 5:e11552014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng C, Wen M and Liu X: Fibroblast activation protein in osteosarcoma cells promotes angiogenesis via AKT and ERK signaling pathways. Oncol Lett 15: 6029-6035, 2018
APA
Zeng, C., Wen, M., & Liu, X. (2018). Fibroblast activation protein in osteosarcoma cells promotes angiogenesis via AKT and ERK signaling pathways. Oncology Letters, 15, 6029-6035. https://doi.org/10.3892/ol.2018.8027
MLA
Zeng, C., Wen, M., Liu, X."Fibroblast activation protein in osteosarcoma cells promotes angiogenesis via AKT and ERK signaling pathways". Oncology Letters 15.4 (2018): 6029-6035.
Chicago
Zeng, C., Wen, M., Liu, X."Fibroblast activation protein in osteosarcoma cells promotes angiogenesis via AKT and ERK signaling pathways". Oncology Letters 15, no. 4 (2018): 6029-6035. https://doi.org/10.3892/ol.2018.8027