Ligularia fischeri inhibits endothelial cell proliferation, invasion and tube formation through the inactivation of mitogenic signaling pathways and regulation of vascular endothelial cadherin distribution and matrix metalloproteinase expression

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

  • Published online on: May 20, 2015     https://doi.org/10.3892/or.2015.4000
  • Pages: 221-226
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ligularia fischeri (LF) has been used as an edible herb and traditional medicine for the treatment of inflammatory and infectious diseases. In the present study, we report the effects and molecular mechanism of the ethanolic extract of LF on cell proliferation, invasion and tube formation in human umbilical vein endothelial cells (HUVECs). LF-mediated inhibition of cell proliferation was accompanied by reduced expression of cell cycle-related proteins such as cyclin-dependent kinases (Cdks) and cyclins, leading to pRb hypophosphorylation and G1 phase cell cycle arrest. We also show that LF treatment inhibited cell invasion and tube formation in HUVECs. These anti-angiogenic activities of LF were associated with the inactivation of mitogenic signaling pathways, induction of vascular endothelial (VE)-cadherin distribution at cell-cell contacts and inhibition of matrix metalloproteinase (MMP) expression. Collectively, our findings demonstrate the pharmacological functions and molecular mechanisms of LF in regulating endothelial cell fates, and support further development as a potential therapeutic agent for the treatment and prevention of angiogenesis-related disorders including cancer.

Introduction

Angiogenesis, the formation of new blood vessels from pre-existing neighboring vessels, is essential for pathological conditions including cancer, ocular disorders, arthritis and obesity as well as physiological processes such as wound-healing, menstruation and ovulation (1,2). The process of angiogenesis includes endothelial cell proliferation, migration, adhesion, invasion, tube formation and recruitment of pericytes, and is tightly regulated by the complex interplay of angiogenic and anti-angiogenic factors within tissue microenvironment (35). Matrix metalloproteinases (MMPs) play important roles in tissue remodeling by degrading extracellular matrix (ECM) components and cell surface molecules, leading to angiogenic responses associated with cancer growth and progression (68). MMP-mediated cleavage of vascular endothelial (VE)-cadherin in cell surfaces may promote vascular permeability, proliferation, invasion and capillary-like structure formation by dissociating cadherin-catenin complex and disrupting cell-cell adhesion (914). The activities of these MMPs are regulated by endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs) (15). In addition to MMP-inhibitory activity, many investigations demonstrate that TIMPs regulate cell fates such as proliferation, migration, apoptosis and differentiation through MMP-independent mechanism (1621). Molecular mechanisms underlying regulation of expression and activities of MMPs and TIMPs may be attractive therapeutic targets and strategy for intervention in angiogenesis-related disorders including cancer. However, MMP inhibitors in multiple clinical trials show severe musculoskeletal pain and inflammation as well as limited efficacy, suggesting that the identification of the specific MMP target is absolutely required for realizing the clinical potential of MMP inhibitors (22,23).

Ligularia fischeri (LF) (Ledebour) Turczaninow var. spiciformis Nakai (Compositae), an edible herb distributed in Eastern Asia including Korea, China and Japan, has been used in traditional medicine to treat rheumatoid arthritis, erysipelas, scarlet fever and jaundice. Previous investigations demonstrate that the extracts and biologically active components of LF exert anti-inflammatory, anti-oxidative, anti-hepatotoxic and anti-obesity activities (2429). In addition, several studies show the anticancer activity of LF against various cancer cell lines including acute promyelocytic leukemia, oral cancer, breast cancer, lung cancer and ovarian cancer cells (3033). However, the biological effects of LF on angiogenesis associated with cancer growth and progression have not yet been explored. In the present study, we evaluated the regulatory effects and molecular mechanisms of LF on cell proliferation, invasion and capillary-like structure formation in human umbilical vein endothelial cells (HUVECs).

Materials and methods

Cell culture conditions

Primary cultures of HUVECs were purchased from Lonza (Walkersville, MD, USA) and used between passages 3 and 6 for all experiments. Cells were cultured in EGM-2® BulletKit containing endothelial basal medium-2 (EBM-2) and the following growth supplements (EGM-2® SingleQuots Kit: human epidermal growth factor, vascular endothelial growth factor, R3-insulin-like growth factor-1, human fibroblast growth factor, ascorbic acid, hydrocortisone, heparin, fetal bovine serum and gentamicin/amphotericin B) (designated as complete media), according to the manufacturer’s instructions (Lonza).

Reagents

The following pharmacological agents and antibodies were purchased from commercial sources: anti-phospho-extra-cellular signal-regulated kinase (ERK) (T202/Y204), anti-phospho-Akt (S473), anti-phospho-p70S6K (T421/S424), anti-phospho-p38MAPK (T180/Y182), anti-MMP-2, anti-MMP-9, anti-phospho-pRb (S780) and anti-phospho-pRb (S807/S811) (all from Cell Signaling Technology, Beverly, MA, USA); anti-p27Kip1 (BD Biosciences, Bedford, MA, USA); anti-ERK, anti-Akt, anti-p70S6K, anti-p38MAPK, anti-cyclin-dependent kinase (Cdk)4, anti-Cdk2, anti-cyclin D, anti-cyclin E, anti-actin antibodies and mouse and rabbit IgG-horseradish peroxidase conjugates (Santa Cruz Biotechnology, Santa Cruz, CA, USA).

Preparation of LF extract

Five hundred grams of LF were extracted with 2 liters of ethanol and stirring for 5 h. The extract of LF was obtained as previously reported (32).

Cell viability and proliferation assay

Subconfluent HUVECs, plated on 6-well plates (1×105 cells/well; BD Biosciences), were serum-starved for 14 h in EBM-2 media to synchronize cells in G1/G0 phase of cell cycle and incubated for 24 h in EGM-2 BulletKit media in the presence or absence of LF (1 and 10 µg/ml). Following culture for 24 h, cell viability was determined by a Muse™ Cell Analyzer using Cell Count and Viability Assay kit (Merck Millipore, Billerica, MA, USA), and the cell proliferation was quantified as previously described (34). The results from triplicate determinations (mean ± standard deviation) are presented as the fold-increase of the untreated controls or the percentage of viable cells of the total cell count.

Cell cycle analysis

Quiescent HUVECs were incubated for 24 h in EGM-2 BulletKit media in the presence or absence of LF (10 µg/ml). Cells were harvested with trypsin-EDTA, rinsed with phosphate-buffered saline (PBS, pH 7.4) and then fixed with ice-cold 70% ethanol for 3 h. After washing with PBS, cells were stained with Muse™ cell cycle reagent. The profile of cells in the G1/G0, S and G2/M phases of the cell cycle was analyzed with a Muse™ Cell Analyzer (Merck Millipore) (35).

Western blot analysis

Quiescent HUVECs in 100-mm dishes (1×106 cells/dish; BD Biosciences) were incubated for 15 min or 24 h in EGM-2 Bulletkit media in the presence or absence of LF (10 µg/ml). Cells were rinsed twice with ice-cold 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 described previously (3638). All western blot analyses are representative of at least three independent experiments. Bands of interest were integrated and quantified by the use of National Institutes of Health (NIH) ImageJ version 1.34s software.

Invasion assay

The upper side of the Transwell insert (Costar, 6.5-mm diameter insert, 8-µm pore size) (Corning Inc., Corning, NY, USA) was coated with 50 µl of 1 mg/ml Matrigel® basement membrane matrix (10.4 mg/ml; BD Biosciences) diluted in EBM-2. Aliquots (100 µl) of HUVECs (5×104 cells/ml) resuspended in EBM-2 were added to the upper compartment of the Matrigel-coated Transwell and 600 µl of EBM-2 was added to the lower compartment. After serum starvation with EBM-2 for 2 h, cells were incubated for 18 h in EGM-2 BulletKit media in the presence or absence of LF (1 and 10 µg/ml). The inserts were fixed with methanol and using a cotton-tipped swab the non-invasive cells were removed from the top of the membrane. After staining with 0.04% Giemsa solution (Sigma-Aldrich, St. Louis, MO, USA), the number of invasive cells was determined from six different fields using ×200 objective magnification.

Tube formation assay

Each well of pre-chilled 24-well plates was coated with 200 µl Matrigel (BD Biosciences). Following serum starvation with EBM-2 for 2 h, HUVECs (3×104 cells/ml) were added to Matrigel®-coated plates and incubated for 6 h in EGM-2 BulletKit media in the presence or absence of LF (1 and 10 µg/ml). Tube formation was observed with an Olympus CKX41 inverted microscope (CAchN 10/0.25php objective) and ToupTek Toupview software (version ×86, 3.5.563; Hangzhou ToupTek Photonics Co., Zhejiang, China) (39).

Immunofluorescence microscopy

Quiescent HUVECs on gelatin-coated coverslips in 12-well plates were incubated for 30 min in EGM-2 BulletKit media in the presence or absence of LF (10 µg/ml), fixed with 3.7% paraformaldehyde for 5 min, washed with PBS, permeabilized with 0.1% Triton X-100 for 10 min, washed with PBS and blocked with PBS containing 5% BSA for 1 h. Primary antibodies diluted 1:100 in 5% BSA-PBS were incubated for 2 h at room temperature, washed with PBS, and followed by Alexa Fluor 488-conjugated goat anti-mouse IgG (Life Technologies, Grand Island, NY, USA). Images were obtained with Carl Zeiss microscope (Axio Imager. M2) and AxioVision Rel. 4.8 software (Zeiss Co., Gottingen, Germany).

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 at P<0.05.

Results

LF suppresses endothelial cell proliferation through regulating the expression of cell cycle-related proteins

We first examined the ability of LF to modulate cell proliferation of HUVECs. LF treatment suppressed endothelial cell proliferation in a dose-dependent manner (Fig. 1A) and did not alter cell viability (Fig. 1B), indicating that LF inhibition of endothelial cell proliferation is not mediated by induction of apoptosis or cytotoxicity. This finding is similar to the patterns of LF in other cell types as previously reported (32,33). We next examined the effect of LF on the cell cycle by DNA content analysis (Fig. 2A). LF treatment prevented the increase in S phase (10.2 vs. 8.7%) and G2/M phase (26.0 vs. 21.5%) and the decrease in G1 phase (63.8 vs. 69.8%) associated with mitogenic stimulation, similar to those of untreated controls. These observations suggest that LF inhibits the transition from G1 to S phase, leading to G1 arrest, which is well correlated with inhibition of cell proliferation (Fig. 1A). We have previously reported that the ethanolic extract and ethyl caffeate, a natural phenolic compound isolated from LF, had growth-suppressive activity in different types of cancer cells including non-small cell lung and ovarian cancer, this inhibitory effect was found to be mediated by downregulation of Cdks and cyclins (32,33). Based on these findings, we analyzed the changes of cell cycle-related proteins such as Cdks, cyclins and Cdk inhibitor p27Kip1 in LF-treated HUVECs. As shown in Fig. 2B, LF treatment markedly reduced the expression of Cdks and cyclins, enhanced the levels of p27Kip1, leading to inhibition of pRb phosphorylation in response to mitogenic stimulation. These findings clearly show the regulatory effects of LF on cell cycle progression and proliferation in HUVECs.

LF inhibits endothelial cell invasion and tube formation

The release of a variety of biologically active molecules from ECM and cell surface components by MMP-mediated proteolytic degradation is associated with the regulation of cellular behavior such as cell adhesion, migration and invasion (68). Thus, we next examined the changes of cell invasion and MMP expression in LF-treated HUVECs. As shown in Fig. 3, LF treatment markedly inhibited the invasion and MMP expression in response to mitogenic stimuli, suggesting that the anti-invasive activity of LF may be mediated through downregulation of MMP expression. In addition, LF treatment completely suppressed mitogen-induced capillary-like structure formation to the levels observed in untreated controls (Fig. 4). Collectively, these findings clearly show the pharmacological roles of LF in regulating endothelial cell proliferation, invasion and tube formation.

Anti-angiogenic effects of LF are mediated through the down-regulation of mitogenic signaling pathways and VE-cadherin expression

Distribution of VE-cadherin at cell-cell contacts has been reported to enhance the stability of adherens junctions, resulting in maintenance of endothelial barrier function (9,10). Angiogenic factors such as VEGF-A disrupt the loss of VE-cadherin from the endothelial cell surfaces and induces endothelial cell responses such as permeability, proliferation, invasion and tube formation (13,14). Change of VE-cadherin function can be assessed by the levels of VE-cadherin detectable at cell-cell contacts. As shown in Fig. 5A, LF treatment prevented the mitogen-induced loss of VE-cadherin from cell-cell contacts, similar to untreated controls. To further investigate the molecular mechanisms by which LF modulates mitogen-induced endothelial cell responses, we examined the changes in activation of mitogenic signaling pathways including ERK, Akt, p70S6K and p38MAPK (40). As shown in Fig. 5B, LF treatment markedly inhibited the mitogen-induced phosphorylation/activation of ERK, Akt, p70S6K and p38MAPK in HUVECs. Collectively, these observations suggest that anti-angiogenic activities of LF may be mediated at least in part through the inactivation of mitogenic signaling pathways and redistribution of VE-cadherin at cell-cell contacts.

Discussion

Overexpressed receptor tyrosine kinases (RTKs) and dysregulation of RTK downstream signaling pathways are closely associated with pathological conditions including cancer (40). Selective inhibition or normalization of RTK-mediated signaling pathways has widely been appreciated as a rational therapeutic strategy. However, many drugs that target RTKs and the downstream signaling networks frequently lead to drug resistance and adverse effects in clinical trials or use. Therefore, natural products which act simultaneously on multiple molecular targets can sometimes be of therapeutic benefit in treating diseases. LF has been used for improving liver function, as well as in inflammatory and infectious disorders. These applications of LF may be mediated through anti-inflammatory, anti-oxidative and anti-hepatotoxic effects (2528). We have previously reported that the ethanolic extract of LF inhibits proliferation and migration of non-small cell lung cancer cells through the inactivation of signaling pathways such as ERK, Akt and p70S6K, and the downregulation of epidermal growth factor receptor, integrin β1 and integrin-linked kinase (ILK) (32). In addition, ethyl caffeate, a natural phenolic compound isolated from the ethanolic extract of LF, exerts anti-proliferative, anti-migratory and anti-invasive activities in ovarian cancer cells. The mechanism of these effects involves suppression of signaling pathways including ERK, Akt, p70S6K and p38MAPK, and downregulation of human epidermal growth factor receptor 2, fibroblast growth factor receptor-1, vascular endothelial growth factor receptor-2, integrin α3β1, ILK and N-cadherin (33).

In the present study, we demonstrate for the first time that LF inhibits mitogen-induced endothelial cell proliferation, invasion and tube formation. These anti-angiogenic activities of LF were found to be mediated through the inactivation of mitogenic signaling pathways, redistribution of VE-cadherin at cell-cell contacts and downregulation of MMP expression. Based on the regulatory effects of LF on MMP expression, we examined the ability of LF to alter the levels of TIMP-2, an endogenous inhibitor of MMPs which has been known to regulate cell proliferation and differentiation through MMP-dependent and/or MMP-independent mechanism (15,16,21). LF treatment showed little or no change of TIMP-2 expression in mitogen-treated HUVECs (data not shown). These findings indicate that anti-angiogenic effects of LF may be mediated through the regulation of MMP-2 and MMP-9, but not that of TIMP-2. However, it cannot exclude the possibility that LF may modulate the expression and activity of other MMP and TIMP family members.

In conclusion, the present study demonstrates the pharmacological roles and mechanisms of LF in the regulation of angiogenesis, and warrants further evaluation and development of LF for the prevention and treatment of pathological states associated with angiogenesis.

Acknowledgments

This study was supported by the High Value-added Food Technology Development Program (112060-3) through the Ministry of Agriculture, Food and Rural Affairs, and by the Basic Science Research Program (2010-0021913, 2014R1A1A2058015) through the National Research Foundation of Korea, Ministry of Education.

References

1 

Cristofanilli M, Charnsangavej C and Hortobagyi GN: Angiogenesis modulation in cancer research: novel clinical approaches. Nat Rev Drug Discov. 1:415–426. 2002. 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 

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

4 

Cook KM and Figg WD: Angiogenesis inhibitors: current strategies and future prospects. CA Cancer J Clin. 60:222–243. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Nyberg P, Xie L and Kalluri R: Endogenous inhibitors of angiogenesis. Cancer Res. 65:3967–3979. 2005. View Article : Google Scholar : PubMed/NCBI

6 

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

7 

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

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 

Dejana E, Orsenigo F and Lampugnani MG: The role of adherens junctions and VE-cadherin in the control of vascular permeability. J Cell Sci. 121:2115–2122. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Kim SH, Cho YR, Kim HJ, Oh JS, Ahn EK, Ko HJ, Hwang BJ, Lee SJ, Cho Y, Kim YK, et al: Antagonism of VEGF-A-induced increase in vascular per meability by an integrin α3β1-Shp-1-cAMP/PKA pathway. Blood. 120:4892–4902. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Herren B, Levkau B, Raines EW and Ross R: Cleavage of beta-catenin and plakoglobin and shedding of VE-cadherin during endothelial apoptosis: evidence for a role for caspases and metalloproteinases. Mol Biol Cell. 9:1589–1601. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Grazia Lampugnani M, Zanetti A, Corada M, Takahashi T, Balconi G, Breviario F, Orsenigo F, Cattelino A, Kemler R, Daniel TO, et al: Contact inhibition of VEGF-induced proliferation requires vascular endothelial cadherin, beta-catenin, and the phosphatase DEP-1/CD148. J Cell Biol. 161:793–804. 2003. View Article : Google Scholar : PubMed/NCBI

13 

George SJ and Dwivedi A: MMPs, cadherins, and cell proliferation. Trends Cardiovasc Med. 14:100–105. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Spring K, Chabot C, Langlois S, Lapointe L, Trinh NT, Caron C, Hebda JK, Gavard J, Elchebly M and Royal I: Tyrosine phosphorylation of DEP-1/CD148 as a mechanism controlling Src kinase activation, endothelial cell permeability, invasion, and capillary formation. Blood. 120:2745–2756. 2012. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Seo D-W, Li H, Guedez L, Wingfield PT, Diaz T, Salloum R, Wei BY and Stetler-Stevenson WG: TIMP-2 mediated inhibition of angiogenesis: an MMP-independent mechanism. Cell. 114:171–180. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Qi JH, Ebrahem Q, Moore N, Murphy G, Claesson-Welsh L, Bond M, Baker A and Anand-Apte B: 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

18 

Jung KK, Liu XW, Chirco R, Fridman R and Kim HR: 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

19 

Seo DW, Li H, Qu CK, Oh J, Kim YS, Diaz T, Wei B, Han JW and Stetler-Stevenson WG: 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 :

20 

Seo DW, Kim SH, Eom SH, Yoon HJ, Cho YR, Kim PH, Kim YK, Han JW, Diaz T, Wei BY, et al: TIMP-2 disrupts FGF-2-induced downstream signaling pathways. Microvasc Res. 76:145–151. 2008. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Coussens LM, Fingleton B and Matrisian LM: Matrix metalloproteinase inhibitors and cancer: trials and tribulations. Science. 295:2387–2392. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Vandenbroucke RE and Libert C: Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat Rev Drug Discov. 13:904–927. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Hwang BY, Lee JH, Koo TH, Kim HS, Hong YS, Ro JS, Lee KS and Lee JJ: Furanoligularenone, an eremophilane from Ligularia fischeri, inhibits the LPS-induced production of nitric oxide and prostaglandin E2 in macrophage RAW2647 cells. Planta Med. 68:101–105. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Lee KH and Choi EM: Analgesic and anti-inflammatory effects of Ligularia fischeri leaves in experimental animals. J Ethnopharmacol. 120:103–107. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Choi EM: Ligularia fischeri leaf extract prevents the oxidative stress in DBA/1J mice with type II collagen-induced arthritis. J Appl Toxicol. 27:176–182. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Shang YF, Kim SM, Song DG, Pan CH, Lee WJ and Um BH: Isolation and identification of antioxidant compounds from Ligularia fischeri. J Food Sci. 75:C530–C535. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Choi J, Park JK, Lee KT, Park KK, Kim WB, Lee JH, Jung HJ and Park HJ: In vivo antihepatotoxic effects of Ligularia fischeri var. spiciformis and the identification of the active component, 3,4-dicaffeoylquinic acid. J Med Food. 8:348–352. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Cha KH, Song DG, Kim SM and Pan CH: Inhibition of gastrointestinal lipolysis by green tea, coffee, and gomchui (Ligularia fischeri) tea polyphenols during simulated digestion. J Agric Food Chem. 60:7152–7157. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Jeong SH, Koo SJ, Choi JH, Park JH, Ha J, Park HJ and Lee KT: Intermedeol isolated from the leaves of Ligularia fischeri var. spiciformis induces the differentiation of human acute promyeocytic leukemia HL-60 cells. Planta Med. 68:881–885. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Xie WD, Li X, Weng CW, Liu SS and Row KH: Fischerisin A and B, cytotoxic sesquiterpenoid-geranylhydroquinones from Ligularia fischeri. Chem Pharm Bull (Tokyo). 59:511–514. 2011. View Article : Google Scholar

32 

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

33 

Lee HN, Kim JK, Kim JH, Lee SJ, Ahn EK, Oh JS and Seo DW: A mechanistic study on the anti-cancer activity of ethyl caffeate in human ovarian cancer SKOV-3 cells. Chem Biol Interact. 219:151–158. 2014. View Article : Google Scholar : PubMed/NCBI

34 

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

35 

Kim HJ, Ko HY, Choi SW and Seo DW: Anti-angiogenic effects of Siegesbeckia glabrescens are mediated by suppression of the Akt and p70S6K-dependent signaling pathways. Oncol Rep. 33:699–704. 2015.

36 

Cho YR, Choi SW 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.PubMed/NCBI

37 

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. View Article : Google Scholar : PubMed/NCBI

38 

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.

39 

Cho YR, Kim JH, Kim JK, Ahn EK, Ko HJ, In JK, Lee SJ, Bae GU, Kim YK, 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.PubMed/NCBI

40 

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

Related Articles

Journal Cover

July-2015
Volume 34 Issue 1

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
Kim JH, Kim H, Kim J, Ahn E, Ko H, Cho Y, Lee S, Bae G, Kim YK, Park JW, Park JW, et al: Ligularia fischeri inhibits endothelial cell proliferation, invasion and tube formation through the inactivation of mitogenic signaling pathways and regulation of vascular endothelial cadherin distribution and matrix metalloproteinase expression. Oncol Rep 34: 221-226, 2015
APA
Kim, J.H., Kim, H., Kim, J., Ahn, E., Ko, H., Cho, Y. ... Seo, D. (2015). Ligularia fischeri inhibits endothelial cell proliferation, invasion and tube formation through the inactivation of mitogenic signaling pathways and regulation of vascular endothelial cadherin distribution and matrix metalloproteinase expression. Oncology Reports, 34, 221-226. https://doi.org/10.3892/or.2015.4000
MLA
Kim, J. H., Kim, H., Kim, J., Ahn, E., Ko, H., Cho, Y., Lee, S., Bae, G., Kim, Y. K., Park, J. W., Oh, J. S., Seo, D."Ligularia fischeri inhibits endothelial cell proliferation, invasion and tube formation through the inactivation of mitogenic signaling pathways and regulation of vascular endothelial cadherin distribution and matrix metalloproteinase expression". Oncology Reports 34.1 (2015): 221-226.
Chicago
Kim, J. H., Kim, H., Kim, J., Ahn, E., Ko, H., Cho, Y., Lee, S., Bae, G., Kim, Y. K., Park, J. W., Oh, J. S., Seo, D."Ligularia fischeri inhibits endothelial cell proliferation, invasion and tube formation through the inactivation of mitogenic signaling pathways and regulation of vascular endothelial cadherin distribution and matrix metalloproteinase expression". Oncology Reports 34, no. 1 (2015): 221-226. https://doi.org/10.3892/or.2015.4000