Broussonetia kazinoki modulates the expression of VEGFR-2 and MMP-2 through the inhibition of ERK, Akt and p70S6K‑dependent signaling pathways: Its implication in endothelial cell proliferation, migration and tubular formation

  • Authors:
    • Young-Rak Cho
    • Jae Hyeon Kim
    • Jin-Kyu Kim
    • Eun-Kyung Ahn
    • Hye-Jin Ko
    • Jae Kyung In
    • Sang-Jin Lee
    • Gyu-Un Bae
    • Yong Kee Kim
    • Joa Sub Oh
    • Dong-Wan Seo
  • View Affiliations

  • Published online on: August 4, 2014     https://doi.org/10.3892/or.2014.3380
  • Pages: 1531-1536
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Broussonetia kazinoki (BK) has been used as a traditional medicine to improve vision, as well as for inflammatory and infectious diseases. In the present study, we investigated the effects and molecular mechanism of the ethanolic extract of BK on cell proliferation, migration and tubular formation in vascular endothelial growth factor-A (VEGF-A)-treated human umbilical vein endothelial cells. BK treatment inhibited VEGF-A-stimulated endothelial cell proliferation through the downregulation of cell cycle-related proteins including cyclin-dependent kinases and cyclins. Moreover, BK treatment suppressed cell migration and tubular formation in response to VEGF-A. These anti-angiogenic activities of BK were associated with the inactivation of mitogenic signaling pathways including extracellular signal-regulated kinase, Akt and p70S6K, and the subsequent downregulation of VEGFR-2 and matrix metalloproteinase-2. Taken together, these findings suggest further evaluation and development of BK as a potential therapeutic agent for the treatment and prevention of angiogenesis-related diseases including cancer.

Introduction

Angiogenesis, the formation and recruitment of new blood vessels, is known to play important roles in cancer growth and progression, and is regulated by the production of several angiogenic and anti-angiogenic factors (1,2). Among angiogenic factors, vascular endothelial growth factor-A (VEGF-A) and VEGF receptor-2 (VEGFR-2) signaling pathways stimulate the secretion and activation of matrix metalloproteinases (MMPs) in endothelial cells which result in cell proliferation, migration and survival, and are widely appreciated as the therapeutic targets for a variety of angiogenesis-related diseases. The central role of the VEGF-A/VEGFR-2 in this process is evidenced by the development and approval of pegaptanib and ranibizumab for age-related macular degeneration as well as bevacizumab and sunitinib for cancer treatment (35). However, current approved anti-angiogenic drugs frequently lead to drug resistance and recurrence of cancer growth and progression. Thus, further understanding of the molecular mechanisms and targets of angiogenic responses may help to develop potential therapeutic strategies for the treatment of cancer as well as angiogenesis-related diseases.

MMPs belong to a family of zinc-dependent endopeptidases and are responsible for tissue remodeling by selective proteolytic degradation, resulting in the promotion of cancer growth and progression through alterations of cell adhesion, migration, epithelial to mesenchymal transition, tumor angiogenesis and release of growth factors (68). These MMPs are regulated by their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs) (9,10). In addition to MMP-inhibitory activity, TIMPs have been reported to regulate cell growth, migration and differentiation through the MMP-independent mechanism (1114). Identification of molecular mechanisms and targets in regulating expression and activities of MMPs and TIMPs within the tumor microenvironment has become an attractive strategy for therapeutic intervention in cancer growth and progression.

Broussonetia kazinoki (BK) Siebold (Moraceae) has been used as a traditional medicine for the amelioration of vision, inflammatory and infectious diseases as well as a material for production of paper in Northeastern Asia. Previous investigations have demonstrated that the extracts and bioactive phytochemicals of BK have anti-diabetic, anti-hyperglycemic, anti-inflammatory, anti-allergic and anticancer activities (1519). However, the effects and molecular mechanisms of BK on angiogenesis remain unknown. In the present study, we evaluated the effects and signaling pathways of BK on proliferation, migration and tubular formation in human umbilical vein endothelial cells.

Materials and methods

Cell culture conditions

Primary cultures of human umbilical vein endothelial cells (HUVECs) were purchased from Lonza (Walkersville, MD, USA) and used between passages 4 and 6 for all the experiments. The cells were cultured in EGM-2® BulletKit media, according to the manufacturer’s instructions (Lonza).

Reagents

The following pharmacological agents and antibodies were purchased from commercial sources: vascular endothelial growth factor-A (VEGF-A) and LY294002 (Merck Millipore, Billerica, MA, USA); rapamycin (Sigma-Aldrich, St. Louis, MO, USA); PD98059, anti-phospho-ERK (T202/Y204), anti-phospho-Akt (S473), anti-phospho-p70S6K (T421/S424), anti-phospho-p38MAPK (T180/Y182), anti-phospho-pRb (S780) and anti-MMP-2 (Cell Signaling Technology, Beverly, MA, USA); anti-VEGFR-2, anti-ERK, anti-Akt, anti-p70S6K, anti-p38MAPK, anti-TIMP-2, anti-Cdk4, anti-Cdk2, anti-cyclin D, anti-cyclin E, anti-actin antibodies and mouse and rabbit IgG-horseradish peroxidase conjugates (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA).

Preparation of Broussonetia kazinoki (BK) extract

The ethanol extract was prepared by mixing 100 g of twigs of BK with 1 liter of 100% ethanol and stirring for 72 h. The extract was then filtered through a filter paper (Advantec No. 1; Toyo Roshi Kaisha, Ltd., Tokyo, Japan), and the filtrate was concentrated using a rotary evaporator (Tokyo Rikakikai Co., Ltd., Tokyo, Japan) at 40°C under vacuum. The yield of the dried extract was ~1.37%.

Cell viability and proliferation assay

HUVECs, plated on 6-well plates (1×105 cells/well; BD Biosciences, Bedford, MA, USA), were serum-starved for 14 h to synchronize cells in the G1/G0 phase of the cell cycle, and pretreated with BK (0.1–10 μg/ml) for 30 min in the presence or absence of PD98059 (25 μM), LY294002 (10 μM) or rapamycin (50 nM) as indicated, and then incubated with VEGF-A (10 ng/ml) for 24 h. Following culture for 24 h, cell viability was determined by a Muse™ cell analyzer using a cell count and viability assay kit (Merck Millipore), and the cell proliferation was quantified as previously described (2022). The results from triplicate determinations (mean ± standard deviation) are presented as the number of cells per culture.

Western blot analysis

HUVECs in 100-mm dishes (1×106 cells/dish; BD Biosciences) were serum-starved for 14 h in endothelial cell basal medium (EBM-2; Lonza) and replaced with fresh media, followed by treatments for different time-points, as indicated in the figure legends, at 37°C. The cells were rinsed twice with ice-cold phosphate-buffered saline (PBS) and lysed by incubation in 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 10% glycerol, 1% Triton X-100, 1 mM EDTA, 100 μg/ml 4-(2-aminoethyl)benzenesulfonyl fluoride, 10 μg/ml aprotinin, 1 μg/ml pepstatin A, 0.5 μg/ml leupeptin, 80 mM β-glycerophosphate, 25 mM sodium fluoride and 1 mM sodium orthovanadate for 30 min at 4°C. Cell lysates were clarified at 12,500 × g for 20 min at 4°C, and the supernatants were subjected to western blot analysis as previously described (23,24). All the western blots were performed in triplicate and representative gels were shown.

Migration assay

Cell migration was quantified in the in vitro wound-healing assay as previously described (25,26). Cells were plated on 48-well plates (4×104 cells/well), grown to confluence, and a single wound was created in the center of the cell monolayer by the gentle removal of the attached cells with a sterile plastic pipette tip. Following serum starvation with EBM-2 for 2 h, the cells were pretreated with BK (0.1–10 μg/ml) for 30 min, followed by VEGF-A (10 ng/ml) stimulation for 16 h. The cells were fixed with methanol, and then stained with 0.04% Giemsa solution (Sigma-Aldrich). The migration of the cells into the wound was observed using images captured at the indicated time-points.

Tube formation assays

Matrigel® basement membrane matrix (10.4 mg/ml; BD Biosciences) was thawed overnight at 4°C, and each well of pre-chilled 24-well plates was coated with 200 μl Matrigel® and then incubated at 37°C for 30 min. Following serum starvation with EBM-2 for 2 h, the cells (4×104 cells/ml) were added to Matrigel®-coated plates and pretreated with BK (0.1–10 μg/ml) for 30 min, followed by VEGF-A (10 ng/ml) for 6 h. Tube formation was observed with an Olympus CKX41 inverted microscope (CAchN 10/0.25php objective) and ToupTek Toupview software (version x86, 3.5.563; Hangzhou ToupTek Photonics Co., Ltd., Zhejiang, China).

Zymogram analysis

Activities of MMPs were measured by zymography (27,28). Aliquots of conditioned medium collected from HUVECs treated with BK (10 μg/ml) and VEGF-A (10 ng/ml) for 16 h were diluted in sample buffer, and applied to 8% polyacrylamide gels containing 1 mg/ml gelatin (Sigma-Aldrich) as a substrate. After electrophoresis, the gels were incubated in 2.5% Triton X-100 for 1 h to remove SDS and allow the re-naturalization of MMPs, and then incubated in developing buffer containing 50 mM Tris-HCl (pH 7.5), 10 mM CaCl2, and 150 mM NaCl for 16 h at 37°C. The gels were stained with 0.5% Coomassie brilliant blue R-250 in 30% methanol-10% acetic acid for 3 h and followed by destaining with 30% methanol-10% acetic acid. Gelatinolytic activities were detected as unstained bands against the background of the Coomassie blue-stained gelatin.

Statistical analysis

Statistical analysis was performed using the Student’s t-test, and was based on at least three different experiments. The results were considered to be statistically significant when P<0.05.

Results

BK suppresses VEGF-A-stimulated endothelial cell proliferation by modulating the expression of cell cycle-related proteins

We first examined the ability of BK to regulate proliferation in HUVECs. BK treatment suppressed VEGF-A-stimulated cell proliferation in a dose-dependent manner (Fig. 1A) and did not alter cell viability (Fig. 1B), suggesting that BK inhibition of cell proliferation is not mediated by the induction of apoptosis or cytotoxicity. Based on these findings, we examined the regulatory effect of BK on cell cycle progression by analyzing the changes of cyclin-dependent kinases (Cdks) and cyclins. Cell cycle progression requires activation of Cdks through formation with cyclins and subsequent phosphorylation of retinoblastoma protein (pRb) (29). As shown in Fig. 1C, BK treatment markedly suppressed the expression of Cdks and cyclin D, resulting in pRb hypophosphorylation in VEGF-A-treated HUVECs. These findings indicate that BK downregulates the expression of cell cycle-related proteins, resulting in inhibition of cell cycle progression and proliferation.

BK inhibits VEGF-A-stimulated endothelial cell migration and capillary structure formation

Endothelial cell migration and tubular formation are coordinately controlled by the interactions from ECM molecules to the intracellular and intercellular components of cells, and play important roles in angiogenic responses associated with cancer growth and progression (2). BK treatment dose-dependently inhibited VEGF-A-stimulated cell migration and tubular formation in HUVECs (Figs. 2 and 3). Collectively, these findings suggested that the bioactive components from the ethanolic extracts of BK act on multiple targets and mechanisms involved in cell proliferation, migration and tubular formation, resulting in the regulation of angiogenic responses in vitro.

BK suppresses VEGFR-2 and MMP-2 expression in VEGF-A-treated HUVECs

The expression and activation of VEGFR-2 and MMP-2 have been reported to promote angiogenic responses including endothelial cell proliferation, migration, invasion and tubular formation (1,2,30). To understand the molecular mechanism underlying the regulatory effects of BK on endothelial cell proliferation, migration and tubular formation, we analyzed the changes in the expression of VEGFR-2, MMP-2 and TIMP-2, an endogenous inhibitor of MMPs, in VEGF-A-treated HUVECs. BK treatment markedly suppressed the VEGF-A-induced expression of VEGFR-2 and MMP-2 (Fig. 4A). As shown in Fig. 4B, BK treatment also inhibited VEGF-A-induced activation of MMP-2 in conditioned medium of HUVECs. By contrast, the levels of TIMP-2 in HUVECs were not altered by VEGF-A or BK treatment (Fig. 4A). Taken together, these findings indicates that the inhibitory effects of BK on endothelial cell proliferation, migration and tubular formation may be mediated at least in part through the suppression of VEGFR-2 and MMP-2 expression.

In vitro anti-angiogenic activities of BK are mediated through the inhibition of mitogenic signaling pathways and downregulation of VEGFR-2 and MMP-2

To investigate the molecular mechanisms by which BK regulates endothelial cell responses, we examined the changes in the activation of signaling pathways including extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (p38MAPK), phosphatidylinositol 3-kinase (PI3-K)/Akt and mammalian target of rapamycin (mTOR)/p70S6K, which play pivotal roles in cell fate (31). As shown in Fig. 5A, VEGF-A stimulation markedly increased the phosphorylation/activation of ERK, Akt and p70S6K, but not that of p38MAPK, when compared with unstimulated controls. By contrast, BK treatment significantly inhibited the VEGF-A-stimulated phosphorylation of ERK, Akt, and p70S6K in HUVECs. Pretreatment of cells with PD98059 (an inhibitor of the ERK pathway), LY294002 (an inhibitor of the PI3-K/Akt pathway) or rapamycin (an inhibitor of the mTOR/p70S6K pathway) suppressed the expression of VEGFR-2 and MMP-2 in response to VEGF-A stimulation, suggesting that BK may contain pharmacologically effective components similar to these inhibitors, and share the roles and mechanisms of action in regulating endothelial cell proliferation, migration and tubular formation (Fig. 5B). Taken together, these findings demonstrated that the suppression of endothelial cell proliferation, migration and tubular formation by BK ethanol extracts might be mediated through the inactivation of VEGFR-2 downstream signaling pathways and subsequent downregulation of VEGFR-2 and MMP-2.

Discussion

Broussonetia kazinoki (BK) has been used as a traditional medicine for improving vision, as well as in inflammatory and infectious diseases. These applications are well supported by previous investigations that BK possesses bioactive phytochemicals such as isoprenylated and prenylated flavans to reduce inflammation (15,17). In addition to anti-inflammatory activity, BK has been reported to have cytotoxicity against several different cell lines including liver, cervical, oral, colon, lung and gastric cancer (18,19,32). However, no effects and molecular mechanisms of BK on angiogenesis have been reported thus far.

VEGF-A/VEGFR-2 signaling pathways play pivotal roles in the formation and stability of blood vessels associated with cancer growth and progression (1,3,14,30,33). These angiogenic responses induced by VEGF-A/VEGFR-2 signaling pathways include the secretion and activation of MMPs, resulting in the degradation of the extracellular matrix and remodeling of the tumor microenvironment. Thus, selective inhibition of VEGF-A/VEGFR-2 activation or downstream signaling pathways is appreciated as a potent strategy, compared to conventional chemotherapy.

To the best of our knowledge, in the present study, we have shown for the first time that the ethanol extract of BK inhibits VEGF-A-stimulated proliferation, migration and tubular formation in HUVECs. These anti-angiogenic activities of BK were found to be mediated through the inhibition of VEGF-A-stimulated phosphorylation/activation of ERK, Akt and p70S6K, the downstream targets in VEGFR-2 signaling pathways, and downregulation of VEGFR-2 and MMP-2 as evidenced by using pharmacological inhibitors such as PD98059, LY294002 and rapamycin.

In conclusion, our findings provide important insights into the roles and pharmacological efficacy of BK in regulation of angiogenesis. However, further investigation on the development of BK for the treatment of a variety of diseases associated with angiogenesis should be conducted.

Acknowledgements

The present study was carried out with the support of ‘Forest Science and Technology Projects’ (Project no. S121313L070100) provided by Korea Forest Service.

References

1 

Carmeliet P and Jain RK: Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov. 10:417–427. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Folkman J: Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov. 6:273–286. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Jain RK, Duda DG, Clark JW and Loeffler JS: Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Prac Oncol. 3:24–40. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Ng EWM, Shima DT, Calias P, Cunningham ET, Guyer DR and Adamis AP: Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat Rev Drug Discov. 5:123–132. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Brown DM and Regillo CD: Anti-VEGF agents in the treatment of neovascular age-related macular degeneration: applying clinical trial results to the treatment of everyday patients. Am J Ophthalmol. 144:627–637. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Kessenbrock K, Plaks V and Werb Z: Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 141:52–67. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Stetler-Stevenson WG: Matrix metalloproteinases in angiogenesis: a moving target for therapeutic intervention. J Clin Invest. 103:1237–1241. 1999. View Article : Google Scholar : PubMed/NCBI

8 

Bourboulia D and Stetler-Stevenson WG: Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): positive and negative regulators in tumor cell adhesion. Semin Cancer Biol. 20:161–168. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Brew K and Nagase H: The tissue inhibitors of metalloproteinases (TIMPs): an ancient family with structural and functional diversity. Biochim Biophys Acta. 1803:55–71. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Stetler-Stevenson WG: Tissue inhibitors of metalloproteinases in cell signaling: metalloproteinase-independent biological activities. Sci Signal. 1:re62008. View Article : Google Scholar : PubMed/NCBI

11 

Seo DW, Li H, Guedez L, et al: TIMP-2 mediated inhibition of angiogenesis: an MMP-independent mechanism. Cell. 114:171–180. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Qi JH, Ebrahem Q, Moore N, et al: A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med. 9:407–415. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Jung KK, Liu XW, Chirco R, Fridman R and Kim HRC: Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein. EMBO J. 25:3934–3942. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Kim SH, Cho YR, Kim HJ, et al: Antagonism of VEGF-A-induced increase in vascular permeability by an integrin α3β1-Shp-1-cAMP/PKA pathway. Blood. 120:4892–4902. 2012.PubMed/NCBI

15 

Ryu JH, Ahn H and Jin Lee H: Inhibition of nitric oxide production on LPS-activated macrophages by kazinol B from Broussonetia kazinoki. Fitoterapia. 74:350–354. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Cha JY, Kim YT, Kim HS and Cho YS: Antihyperglycemic effect of stem bark powder from paper mulberry (Broussonetia kazinoki Sieb.) in type 2 diabetic Otsuka Long-Evans Tokushima fatty rats. J Med Food. 11:499–505. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Lee JK, Ha H, Lee HY, et al: Inhibitory effects of heartwood extracts of Broussonetia kazinoki Sieb on the development of atopic dermatitis in NC/Nga mice. Biosci Biotechnol Biochem. 74:1802–1806. 2010.PubMed/NCBI

18 

Ko HH, Yen MH, Wu RR, Won SJ and Lin CN: Cytotoxic isoprenylated flavans of Broussonetia kazinoki. J Nat Prod. 62:164–166. 1998. View Article : Google Scholar

19 

Zhang PC, Wang S, Wu Y, Chen RY and Yu DQ: Five new diprenylated flavonols from the leaves of Broussonetia kazinoki. J Nat Prod. 64:1206–1209. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Kim HJ, Cho YR, Kim SH and Seo DW: TIMP-2-derived 18-mer peptide inhibits endothelial cell proliferation and migration through cAMP/PKA-dependent mechanism. Cancer Lett. 343:210–216. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Seo DW, Cho YR, Kim W and Eom SH: Phytochemical linarin enriched in the flower of Chrysanthemum indicum inhibits proliferation of A549 human alveolar basal epithelial cells through suppression of the Akt-dependent signaling pathway. J Med Food. 16:1086–1094. 2013.PubMed/NCBI

22 

Cho YR, Kim JK, Kim J, Oh J and Seo DW: Ligularia fischeri regulates lung cancer cell proliferation and migration through down-regulation of epidermal growth factor receptor and integrin β1 expression. Genes Genom. 35:741–746. 2013. View Article : Google Scholar

23 

Seo DW, Kim SH, Eom SH, et al: TIMP-2 disrupts FGF-2-induced downstream signaling pathways. Microvasc Res. 76:145–151. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Seo DW, Li H, Qu CK, et al: Shp-1 mediates the antiproliferative activity of tissue inhibitor of metalloproteinase-2 in human microvascular endothelial cells. J Biol Chem. 281:3711–3721. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Cho YR, Kim S, Ko H, Kim MD, Choi S and Seo DW: Sepiapterin inhibits cell proliferation and migration of ovarian cancer cells via down-regulation of p70S6K-dependent VEGFR-2 expression. Oncol Rep. 26:861–867. 2011.PubMed/NCBI

26 

Yoon HJ, Cho YR, Joo JH and Seo DW: Knockdown of integrin α3β1 expression induces proliferation and migration of non-small cell lung cancer cells. Oncol Rep. 29:662–668. 2013.

27 

Cho YR, Choi S and Seo DW: The in vitro antitumor activity of Siegesbeckia glabrescens against ovarian cancer through suppression of receptor tyrosine kinase expression and the signaling pathways. Oncol Rep. 30:221–226. 2013.

28 

Lee HN, Joo JH, Oh JS, Choi SW and Seo DW: Regulatory effects of Siegesbeckia glabrescens on non-small cell lung cancer cell proliferation and invasion. Am J Chin Med. 42:453–463. 2014.

29 

Malumbres M and Barbacid M: Cell cycle, CDKs and cancer: a changing paradigm. Nat Rev Cancer. 9:153–166. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Ellis LM and Hicklin DJ: VEGF-targeted therapy: mechanisms of anti-tumour activity. Nat Rev Cancer. 8:579–591. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Lemmon MA and Schlessinger J: Cell signaling by receptor tyrosine kinases. Cell. 141:1117–1134. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Wei BL, Chen YC and Hsu HY: Kazinol Q from Broussonetia kazinoki enhances cell death induced by Cu(ll) through increased reactive oxygen species. Molecules. 16:3212–3221. 2011.PubMed/NCBI

33 

Olsson AK, Dimberg A, Kreuger J and Claesson-Welsh L: VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol. 7:359–371. 2006. View Article : Google Scholar : 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
Cho Y, Kim JH, Kim J, Ahn E, Ko H, In JK, Lee S, Bae G, Kim YK, Oh JS, Oh JS, et al: Broussonetia kazinoki modulates the expression of VEGFR-2 and MMP-2 through the inhibition of ERK, Akt and p70S6K‑dependent signaling pathways: Its implication in endothelial cell proliferation, migration and tubular formation. Oncol Rep 32: 1531-1536, 2014
APA
Cho, Y., Kim, J.H., Kim, J., Ahn, E., Ko, H., In, J.K. ... Seo, D. (2014). Broussonetia kazinoki modulates the expression of VEGFR-2 and MMP-2 through the inhibition of ERK, Akt and p70S6K‑dependent signaling pathways: Its implication in endothelial cell proliferation, migration and tubular formation. Oncology Reports, 32, 1531-1536. https://doi.org/10.3892/or.2014.3380
MLA
Cho, Y., Kim, J. H., Kim, J., Ahn, E., Ko, H., In, J. K., Lee, S., Bae, G., Kim, Y. K., Oh, J. S., Seo, D."Broussonetia kazinoki modulates the expression of VEGFR-2 and MMP-2 through the inhibition of ERK, Akt and p70S6K‑dependent signaling pathways: Its implication in endothelial cell proliferation, migration and tubular formation". Oncology Reports 32.4 (2014): 1531-1536.
Chicago
Cho, Y., Kim, J. H., Kim, J., Ahn, E., Ko, H., In, J. K., Lee, S., Bae, G., Kim, Y. K., Oh, J. S., Seo, D."Broussonetia kazinoki modulates the expression of VEGFR-2 and MMP-2 through the inhibition of ERK, Akt and p70S6K‑dependent signaling pathways: Its implication in endothelial cell proliferation, migration and tubular formation". Oncology Reports 32, no. 4 (2014): 1531-1536. https://doi.org/10.3892/or.2014.3380