Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
In Vivo
  • Other Publications
    • In Vivo
    • Anticancer Research
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
In Vivo

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Advertisers
    • Editorial Board
  • Other Publications
    • Anticancer Research
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
  • About Us
    • General Policy
    • Contact
  • Visit iiar on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

Fistein Suppresses Human Osteosarcoma U-2 OS Cell Migration and Invasion via Affecting FAK, uPA and NF-ĸB Signaling Pathway In Vitro

JR-KAI CHEN, SHU-FEN PENG, KUANG CHI LAI, HSIN-CHUNG LIU, YI-PING HUANG, CHIN-CHUNG LIN, AN-CHENG HUANG, FU-SHIN CHUEH and JING-GUNG CHUNG
In Vivo May 2019, 33 (3) 801-810; DOI: https://doi.org/10.21873/invivo.11542
JR-KAI CHEN
1Attending Physician of Orthopaedadics, Department of Chang Bing Show-Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
SHU-FEN PENG
2Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
3Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KUANG CHI LAI
4Department of Medical Laboratory Science and Biotechnology, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, Taiwan, R.O.C.
5Department of Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
HSIN-CHUNG LIU
2Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
YI-PING HUANG
6Department of Physiology, College of Medicine, China Medical University, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
CHIN-CHUNG LIN
7Department of Chinese Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Executive Yuan, Taichung, Taiwan, R.O.C.
8General Education Center, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
AN-CHENG HUANG
9Department of Nursing, St. Mary's Junior College of Medicine, Nursing and Management, Yilan, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
FU-SHIN CHUEH
10Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: jgchung@mail.cmu.edu.tw fushin@asia.edu.tw
JING-GUNG CHUNG
2Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
11Department of Biotechnology, Asia University, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: jgchung@mail.cmu.edu.tw fushin@asia.edu.tw
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Evidence has indicated that fisetin induces cytotoxic effects in human cancer cell lines, including the inhibition of cell migration and invasion, however, the exact molecular mechanism of action of fisetin in human osteosarcoma cells remains unclear. Materials and Methods: The anti-metastatic mechanisms of fisetin in human osteosarcoma U-2 OS cells were investigated in vitro. Results: Fisetin reduced the viability of cells at different concentrations (2.5, 5 and 10 μM) as measured by flow cytometric assay. Fisetin suppressed cell mobility, migration and invasion of U-2 OS cells, as shown by wound healing assay and transwell filter chambers, respectively. The gelatin zymography assay showed that fisetin inhibited MMP-2 activity in U-2 OS cells. Results from western blotting indicated that fisetin reduced the levels of pEGFR, SOS-1, GRB2, Ras, PKC, p-ERK1/2, p-JNK, p-p-38, VEGF, FAK, RhoA, PI3K, p-AKT, NF-ĸB, uPA, MMP-7, MMP-9, and MMP-13, but increased GSK3β and E-cadherin in U-2 OS cells after 48 h of treatment. Conclusion: Fisetin can be used in the future, as a target for the treatment of metastasis of human osteosarcoma cells.

  • Fisetin
  • human osteosarcoma U-2 OS cell
  • migration
  • invasion
  • metastasis

Osteosarcoma (OS) has been recognized to be one of the most common malignant bone tumors, which occurs frequently in children and adolescents and exhibits high destructive and metastatic potential (1, 2). OS has been reported to have an increased tendency to metastasize (1, 3, 4). In the past 30 years, the clinical outcome for OS patients with metastatic tumors has remained unchanged, thus, development of new therapeutic strategies for OS patients is urgent (5). Evidence has revealed that the regulation of oncogenes and tumor suppressor genes is vital in the development, progression and metastasis of OS (6-8).

Metastasis, the most important characteristic of malignant tumors, is a multistep process initiated by local invasion, plays a critical role in treatment efficacy and quality of life in patients with metastatic tumors (9) and accounts for more than 90% of cancer related deaths (10, 11).

A vital step in the process of metastasis is the degradation of the extracellular matrix (ECM) by matrix metalloproteinases (MMPs) (12). The MMP system includes MMPs, tissue inhibitor of metalloproteinases (TIMPs) and urokinase-type plasminogen activator (uPA). Furthermore, MMP upregulation promotes cancer metastasis (13-15). The inhibition of molecular pathways involved in cancer metastasis could be a potential strategy to inhibit metastasis (16, 17).

Currently, many chemotherapeutic drugs used in cancer patients are derived from natural sources (18). Fisetin, a dietary tetrahydroxyflavone, is found in natural plants including many fruits and vegetables. It has also been shown to have many biological activities, including inhibiting different types of cancer via the inhibition of multiple oncogenic pathways both in vitro and in vivo (19-21). In our earlier studies, fisetin induced cancer cell apoptosis of human oral cancer SCC-4 and HSC-3 cells by causing endoplasmic reticulum stress, and via caspase- and mitochondria-mediated signaling pathways (22, 23). Fisetin has also been shown to inhibit cell migration and invasion of Epstein-Barr virus latent membrane protein-1 (LMP1)-positive nasopharyngeal carcinoma (NPC) cells (24). Fisetin inhibited cell migration and invasion of cervical cancer cells by repressing uPA via interruption of the p38 MAPK-dependent NF-ĸB signaling pathway (25). Recently, it was also reported that fisetin inhibited the cell growth and migration of human lung cancer A549 cells by blocking the ERK1/2 pathway (26). However, there is no available information showing the effect of fisetin in the cell migration and invasion of human osteosarcoma cells, which was therefore the aim of the present study. The results showed that fisetin had a significant anti-migratory and anti-invasive effect on human osteosarcoma U-2 OS cells in vitro. These findings offer a deeper understanding of the anti-metastatic mechanisms of fisetin in osteosarcoma.

Materials and Methods

Test chemicals, reagents and culture medium. Fisetin, dimethyl sulfoxide (DMSO), Tris-HCl, trypan blue, trypsin, propidium iodide (PI), gelatin and Coomassie blue R-250 were purchased from Sigma Chemical Co. (St. Louis, MO, USA). McCoy's 5A medium, fetal bovine serum (FBS) and penicillin-streptomycin were purchased from Gibco®/Invitrogen Life Technologies (Carlsbad, CA, USA). Antibodies specific for MMP-7, MMP-9 and MMP-13 were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA) and antibodies for Ras, GRB2, Sos1, uPA, PKC, p-EGFRTyr1068, p-ERK1/2, p-JNK, p-P38, VEGF, FAK, Rho A, PI3K, p-AktTHr308, NF-ĸB, GSK3β, Snail, E-cadherin, β-catenin, N-cadherin, Vimentin, p-AktSer473, and peroxidase conjugated secondary antibodies were purchased from Cell Signaling Technology, Inc. (Beverly, MA, USA). Fisetin was dissolved in DMSO (carrier solvent) and 0.5% DMSO was used in control groups. Fisetin was further diluted in culture medium to reach the appropriate final concentrations.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Fisetin decreased cell viability of U-2 OS cells. Cells (1×105 cells/well) were incubated with fisetin (0, 2.5, 5, 10, 20 and 40 μM) for 48 h. Cells were collected for measurement of the percentage of total viable cells as described in Materials and Methods. *p<0.05, **p<0.01, ***p<0.001, significant difference between fisetin-treated groups and the control as analyzed by one-way ANOVA.

Cell line and culture. Human osteosarcoma U-2 OS cell line was obtained from the Food Industry Research and Development Institute (Hsinchu, Taiwan). U-2 OS cells were cultured in McCoy's 5A medium supplemented with 10% FBS, 2 mM L-glutamine, 10 g/l non-essential amino acids, 100 μg/ml streptomycin and 100 units/ml penicillin at 37°C in a humidified atmosphere containing a 5% CO2.

Cell viability assay. U-2 OS cells (1×105 cells/well) were placed onto 12-well plates with McCoy's 5A medium and were incubated with fisetin at the final concentrations 0, 2.5, 5, 10, 20 and 40 μM, in triplicate for 48 h. At the end of the incubation, cells were harvested, washed, counted and stained with PI (5 μg/ml) to measure the percentage of cell viability by using flow cytometry (Becton-Dickinson, San Jose, CA, USA) as previously described (27).

In vitro scratch wound healing assay for cell mobility. Scratch wound healing assay was used to examine cell mobility characteristics as previously described (28). Briefly, U-2 OS cells (1×105 cells/well) were grown in a 12-well plate until they reached a confluent monolayer. Medium was replaced with serum-free McCoy's 5A culture medium. Cell monolayers were scratched (wound) using a sterile 200 μl-pipette tip and PBS was used for washing and removing cell debris. Cells were incubated with various concentrations of fisetin (0, 2.5, 5 and 10 μM) for 24 h. In the denuded zone, the migrating cells were monitored and photographed under phase contrast microscopy and experiments were repeated three times. Image J software was used to quantify the relative wound size. Cell mobility inhibition (%)=new scratch width/original scratch width ×100% as previously described (28, 29).

Gelatin zymography assay for gelatinolytic activity. U-2 OS cells (1×105 cells/well) were placed in a 12-well plate for 24 h, after which medium was replaced with serum-free McCoy's 5A culture medium containing fisetin (0, 2.5, 5 and 10 μM) for 24 and 48 h. At the end of incubation, conditioned medium from each well was harvested and loaded onto 10% polyacrylamide gels and co-polymerized with 0.2% gelatin. Gel was soaked twice in 2.5% Triton X-100 in dH2O at 25°C for 30 min as previously described (28, 30). Then, gel was incubated in zymogen developing buffer (Sigma Aldrich), containing 50 mM Tris (pH 7.5), 200 mM NaCl, 5 mM CaCl2, 1 μM ZnCl2 and 0.02% Brij 35, overnight at 37°C. Band corresponding to MMP-2 activity was stained with 0.2% Coomassie blue in 10% acetic acid and 50% methanol and photographed. The band of gelatinolytic activity was measured using NIH Image J software, version 1.47 (National Institutes of Health, Bethesda, MA, USA), as previously described (30, 31).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Fisetin affected in vitro wound closure of U-2 OS cells. Cells (1×105 cells/well) were kept in a 12-well plate for 24 h, scratched (wounded), and incubated with fisetin (0, 2.5, 5 and 10 μM) for 24 h. The relative wound closures were photographed using phase contrast microscopy (A) and percentage of wound area was calculated (B) as described in Materials and Methods. *p<0.05, **p<0.01, significant difference between fisetin-treated groups and the control as analyzed by one-way ANOVA.

Cell migration and invasion analyzed with the Transwell assay. Cell migration and invasion in vitro were examined by using Collagen and Matrigel assay system as previously described (28, 30). Briefly, U-2 OS cells (5×104 cells/well) in serum-free McCoy's 5A culture medium containing different concentrations of fisetin (0, 2.5, 5 and 10 μM) were placed in the upper chamber (transwell insert) (8 μm pore size; Millipore, Temecula, CA, USA) which was coated with 50 μl collagen (for cell migration examination) overnight. In the lower chamber, 800 μl of McCoy's medium with 10% FBS were placed for 48 h. The non-migrated cells found on the upper surface of the membrane were removed. The migrated cells (those adhered to the lower surface of the membrane) were fixed with 4% formaldehyde in PBS, treated with methanol, stained with 2% crystal violet and all samples were photographed under light microscopy. The percentage of cells that migrated were calculated. The cell invasion assay was performed similarly to the cell migration assay, except that the membrane of the insert (upper chamber) was covered with Matrigel (Matrigel: serum-free medium 1:9) (28, 30).

Western blotting analysis for cell metastasis-associated protein expression. U-2 OS cells (1×106 cells/dish) were placed in 10-cm culture dishes and incubated with fisetin (0, 2.5, 5 and 10 μM) for 48 h. At the end of incubation, cells were collected and resuspended in lysis buffer (50 mM Tris-HCl pH 7.5, 400 mM NaCl, 2 mM EGTA, 1 mM EDTA, 1 mM DTT, and protease inhibitor cocktail) (Roche). The cell lysates were centrifuged at 10,000 × g at 4°C for 10 min and total protein concentration was measured using Bradford protein assay kit as previously described (28, 30). Each sample (30 μg of total protein) was separated by 12% SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred onto PVDF membrane (Millipore, Bedford, MA, USA). All membranes were blocked with 5% non-fat milk in TBS-T buffer (10 mM Tris–HCl, 150 mM NaCl, and 0.05% Tween-20, pH 7.8) for 1 h at room temperature. After washing with TBS-T buffer, membranes were incubated with primary antibodies against MMP-7, MMP-9, MMP-13, p-EGFRTyr1068, Ras, GRB2, Sos1, uPA, PKC, p-ERK1/2, p-JNK, p-p38, VEGF, FAK, Rho A, PI3K, p-AktThr308, p-AktSer473, NF-ĸB, GSK3β, Snail, E-cadherin, β-catenin, N-cadherin, Vimentin. Membranes were incubated with the diluted corresponding HRP-conjugated secondary antibodies (diluted 1:5000; Cell Signaling Technology) and developed with ECL (Amersham). The Biospectrum Imaging System (UVP, Inc., Upland, CA, USA) was used to detect the corresponding bands as previously described (28, 32).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Fisetin inhibited MMP-2 activity and suppressed cell migration and invasion of U-2 OS cells in vitro. Cells (1×105 cells/well) were placed in a 12-well plate and treated with fisetin (0, 2.5, 5 and 10 μM) for 24 and 48 h and then conditioned medium was harvested for gelatin zymography assay, as described in Materials and Methods. (A) Representative gelatin gel pictures. Cells (5×104 cells/well) were placed on a transwell insert coated with collagen for migration or Matrigel for invasion, and were treated with fisetin (0, 2.5, 5 and 10 μM) for 48 h. U-2 OS cells penetrated the lower surface of the transwell membrane for migration (B) and invasion (C) were stained with crystal violet and were photographed under a light microscope at 200×. Penetrated cells were counted as described in Materials and Methods. *p<0.05, **p<0.01, ***p<0.001, significant difference between fisetin-treated groups and the control, as analyzed by one-way ANOVA.

Statistical analysis. Data are presented as mean±SD and were statistically analyzed by one-way ANOVA analysis of variance. *p<0.05, **p<0.01, ***p<0.001 were determined as significant.

Results

Fisetin decreased viability of U-2 OS cells. After treated with various concentrations of fisetin, total viable cell number was measured by flow cytometry. As indicated in Figure 1, fisetin at a concentration of 2.5-5 μM did not show morphological changes and only slightly reduced the percentage of viable cells after 48 h of treatment in U-2 OS cells. However, fisetin at 10 μM induced cell morphological changes and reduced the percentage (about reduced 10%) of viable cells when compared to control groups.

Fisetin inhibited cell mobility in U-2 OS cells. As indicated in Figure 2A and B, fisetin at 5-10 μM inhibited the closure rate of the scratch in U-2 OS cells. After 24 h of incubation, the control cells but not the fisetin-treated cells covered the opening of the scratch. The inhibited effect was dose-dependent, since at the high dose (10 μM), the edge distance was significantly longer, compared to that observed at the low dose (2.5 μM) (Figure 2B). Based on these observations, fisetin significantly suppresses the cell mobility of U-2 OS cells in vitro.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Fisetin affected the levels of proteins associated with migration and invasion of U-2 OS cells. Cells (1×106 cells/dish) were treated with fisetin (0, 2.5, 5 and 10 μM) for 48 h and collected. The proteins were separated by SDS-PAGE as described in the Materials and Methods. The levels of p-EGFRTyr1068, Ras, GRB2, Sos1, uPA and PKC (A); p-ERK1/2, p-JNK, and p-P38 (B); VEGF, FAK, Rho A, PI3K, p-AktThr308, p-AKTThr473, NF-ĸB, MMP-7, MMP-9 and MMP-13 (C); GSK3β, Snail, E-cadherin, β-catenin, N-Cadherin and Vimentin (D) expressions were estimated by western blotting as described in Materials and Methods.

Fisetin affected matrix metalloproteinase activity and cell migration and invasion in U-2 OS cells. Each conditioned medium was collected from fisetin treated U-2 OS cells (2.5, 5 and 10 μM) for 24 and 48 h to measure MMP-2 activity by using gelatin zymography (Figure 3A). Fisetin treatment at 10 μM for 48 h significantly inhibited MMP-2 activity. The results of cell migration and cell invasion assays are presented in Figure 3B and C. Figure 3B indicates that fisetin (2.5, 5 and 10 μM) significantly inhibited the migration of U-2 OS cells by about 40-70% compared to control cells. Figure 3C indicates that fisetin (10 μM) significantly inhibited the invasion of U-2 OS cells about 30% compared to control cells.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

The possible signaling pathways involved in fisetin-induced inhibition of migration and invasion in U-2 OS cells in vitro.

Fisetin affected key metastasis-related proteins in U-2 OS cells. Western blotting results are presented in Figure 4. Results indicated that fisetin (5-10 μM) significantly decreased the levels of p-EGFRTyr1068, Ras, GRB2, Sos1, uPA and PKC (Figure 4A), p-ERK1/2, p-JNK, p-P38 (Figure 4B), VEGF, FAK, Rho A, PI3K, p-AktThr308, p-AktSer473, NF-ĸB, MMP-7, MMP-9 and MMP-13 (Figure 4C), however, it increased GSK3β and E-cadherin levels (Figure 4D) in U-2 OS cells after 48 h treatment. Based on these findings, fisetin suppresses cell metastasis of U-2 OS cells through multiple signaling pathways (Figure 5).

Discussion

Currently, preoperative chemotherapy, surgery, and adjuvant postoperative chemotherapy are the standard treatment strategies for OS patients. Although surgery (tumor excision) combined with adjuvant chemotherapy and radiotherapy have been used, the cure rate of OS patients remain still unsatisfied (33). The reason is due to metastatic lesions in the lungs, multi-drug resistance and the lack of better molecular biomarkers to detect OS at an early stage (34). Some of the chemotherapy drugs were developed to inhibit tumor cell proliferation, apoptosis or metastasis (35-37). Thus, blockage of the signaling pathways leading to cancer cell metastasis is one of best strategies against cancer cells (37, 38). To this end, natural products are being tested for the treatment of OS. Fisetin, which is present in fruits and vegetables, has been shown to induce apoptosis of cancer cells and to inhibit migration and invasion of latent membrane protein-1 (LMP1)-positive nasopharyngeal carcinoma (NPC) cells (24). Fisetin has been found to inhibit cervical cancer cell migration and invasion by repressing uPA via interruption of the p38 MAPK-dependent NF-ĸB signaling pathway (25). Herein, the effects of fisetin on migration and invasion in U-2 OS cells in vitro were investigated.

Treatment of U-2 OS cells with 20-40 μM fisetin for 48 h decreased their viability. Thus, in the wound healing assay, lower concentrations (2.5-10 μM) were used. Treatment of U-2 OS cells with 5-10 μM fisetin for 24 h suppressed cell mobility (Figure 2A and B) in a dose-dependent manner (Figure 2B). This is in agreement with another report showing that fisetin inhibited migration in MCF-7 cells in vitro (39). In order to further confirm this finding, transwell chambers assay was used to examine cell migration and invasion. Fisetin suppressed cell migration at 2.5-10 μM (Figure 3B) and inhibited cell invasion at 10 μM after 48 h treatment (Figure 3C) in U-2 OS cells. These results are also in agreement with another report indicating that fisetin suppressed cell migration and invasion in A549 cells (40). Our results indicate that fisetin suppresses mobility, migration and invasion of U-2 OS cells in vitro. Gelatin zymography was also used to measure the MMP-2 gelatinase activity of U-2 OS cells after treating them with 10 μM of fisetin for 48 h. Results showed that fisetin significantly reduced MMP-2 activity.

In order to understand the mechanism by which fisetin suppresses migration and invasion of U-2 OS cells, proteins associated with EMT and metastasized were analyzed (41). Western blotting analysis indicated that the levels of E-cadherin increased, while those of N-cadherin decreased. Both, E-cadherin and N-cadherin are well known to be involved in cancer cell migration and invasion (42, 43). Furthermore, fisetin was found to decrease the levels of Ras protein in U-2 OS cells (Figure 4A). It is well documented that Ras-related proteins regulate cell adhesion (44, 45). When cancer cells metastasize, decreased E-cadherin and increased N-cadherin expression are observed during EMT, that promotes adhesion to stroma and increased tumor cell motility and invasiveness (46, 47). It was reported that fisetin reduced the levels of Twist protein, an EMT regulator in LMP1-positive nasopharyngeal carcinoma cells (24). Our results showed that fisetin reduced the protein levels of the transcriptional factor Snail. Fisetin also suppressed the protein expression of MMP-7, -9 and -13 in U-2 OS cells (Figure 4C), that is in agreement with another report indicating that fisetin inhibits MMP-1, MMP-3, MMP-7, and MMP-9 in tumor cells (48). In addition, fisetin significantly reduced Rho A, FAK, p-AktThr308 and p-AktSer473 (Figure 4C) protein expression in U-2 OS cells. Rho A and activated PI3K/AKT signaling are associated with cancer cell invasion and oncogenesis (43, 49-51). Focal adhesion kinase (FAK) is involved in cell migration and invasion and the FAK-Rho A signaling pathway is also associated with cell motility (50). In the present study, fisetin was found to suppress the expression of uPA (Figure 4A), p-ERK1/2, p-JNK (Figure 4B), p-AktThr308, p-AktSer473 and NF-ĸB (Figure 4C) in U-2 OS cells. Activated AKT promoted cancer cell invasion and metastasis via stimulating secretion of MMPs (49). p-ERK1/2, and p-JNK have also been involved cell metastasis (52, 53), while NF-ĸB has been linked with tumor cell metastasis (54). Inhibition of NF-ĸB has been recognized as one of the strategies to inhibit cancer cell metastasis (55). The role of uPA has also been reported to be involved in cancer cell metastasis (1) and MMPs have been shown to be up-regulated by uPA and down-regulated by TIMPs (56). Our results suggest that fisetin suppresses migration in U-2 OS cells by inhibiting NF-ĸB and uPA.

In conclusion, fisetin significantly inhibited mobility, migration and invasion of U-2 OS cells by reducing FAK, Rho A, NF-ĸB and uPA protein levels and inhibiting MMP-2 and MMP-9 in vitro.

Acknowledgements

This work was supported by grant RD106003 from Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan, and by grant CMU107-ASIA-17 from China Medical University, Taichung, Taiwan. Experiments and data analysis were performed in part through the use of the Medical Research Core Facilities Center, Office of Research & Development at China medical University, Taichung, Taiwan.

Footnotes

  • Authors' Contributions

    J.K. Chen, F.S. Chueh and J.G. Chung conceived and designed the experiments; K.C. Lai, H.C. Liu performed the experiments; J.K. Chen, F.S. Chueh and H.C. Liu analyzed the data; Y.P. Huang, C.C. Lin. and A.C. Huang contributed reagents/materials/analysis tools; S.F. Peng, F.S. Chueh and J.G. Chung wrote the paper.

  • This article is freely accessible online.

  • Conflicts of Interest

    The Authors have no conflicts of interest to disclose.

  • Received January 30, 2019.
  • Revision received February 25, 2019.
  • Accepted February 26, 2019.
  • Copyright© 2019, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved

References

  1. ↵
    1. Moirangthem A,
    2. Bondhopadhyay B,
    3. Mukherjee M,
    4. Bandyopadhyay A,
    5. Mukherjee N,
    6. Konar K,
    7. Bhattacharya S,
    8. Basu A
    : Simultaneous knockdown of uPA and MMP9 can reduce breast cancer progression by increasing cell-cell adhesion and modulating EMT genes. Sci Rep 6: 21903, 2016. PMID: 26906973, DOI: 10.1038/srep21903
    OpenUrl
  2. ↵
    1. Jones KB,
    2. Salah Z,
    3. Del Mare S,
    4. Galasso M,
    5. Gaudio E,
    6. Nuovo GJ,
    7. Lovat F,
    8. LeBlanc K,
    9. Palatini J,
    10. Randall RL,
    11. Volinia S,
    12. Stein GS,
    13. Croce CM,
    14. Lian JB,
    15. Aqeilan RI
    : miRNA signatures associate with pathogenesis and progression of osteosarcoma. Cancer Res 72: 1865-1877, 2012. PMID: 22350417, DOI: 10.1158/0008-5472.can-11-2663
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. Gibbs CP,
    2. Kukekov VG,
    3. Reith JD,
    4. Tchigrinova O,
    5. Suslov ON,
    6. Scott EW,
    7. Ghivizzani SC,
    8. Ignatova TN,
    9. Steindler DA
    : Stem-like cells in bone sarcomas: implications for tumorigenesis. Neoplasia 7: 967-976, 2005. PMID: 16331882, DOI: 10.1002/cncr.30439
    OpenUrlCrossRefPubMed
  4. ↵
    1. Meyers PA,
    2. Healey JH,
    3. Chou AJ,
    4. Wexler LH,
    5. Merola PR,
    6. Morris CD,
    7. Laquaglia MP,
    8. Kellick MG,
    9. Abramson SJ,
    10. Gorlick R
    : Addition of pamidronate to chemotherapy for the treatment of osteosarcoma. Cancer 117: 1736-1744, 2011. PMID: 21472721, DOI: 10.1002/cncr.25744
    OpenUrlCrossRefPubMed
  5. ↵
    1. Kansara M,
    2. Teng MW,
    3. Smyth MJ,
    4. Thomas DM
    : Translational biology of osteosarcoma. Nat Rev Cancer 14: 722-735, 2014. PMID: 25319867, DOI: 10.1038/nrc3838
    OpenUrlCrossRefPubMed
  6. ↵
    1. Lu J,
    2. Song G,
    3. Tang Q,
    4. Zou C,
    5. Han F,
    6. Zhao Z,
    7. Yong B,
    8. Yin J,
    9. Xu H,
    10. Xie X,
    11. Kang T,
    12. Lam Y,
    13. Yang H,
    14. Shen J,
    15. Wang J
    : IRX1 hypomethylation promotes osteosarcoma metastasis via induction of CXCL14/NF-kappaB signaling. J Clin Invest 125: 1839-1856, 2015. PMID: 25822025, DOI: 10.1172/jci78437
    OpenUrl
    1. Hou CH,
    2. Lin FL,
    3. Hou SM,
    4. Liu JF
    : Cyr61 promotes epithelial-mesenchymal transition and tumor metastasis of osteosarcoma by Raf-1/MEK/ERK/Elk-1/TWIST-1 signaling pathway. Mol Cancer 13: 236, 2014. PMID: 25326651, DOI: 10.1186/1476-4598-13-236
    OpenUrlCrossRef
  7. ↵
    1. Tsai HC,
    2. Su HL,
    3. Huang CY,
    4. Fong YC,
    5. Hsu CJ,
    6. Tang CH
    : CTGF increases matrix metalloproteinases expression and subsequently promotes tumor metastasis in human osteosarcoma through down-regulating miR-519d. Oncotarget 5: 3800-3812, 2014. PMID: 25003330, DOI: 10.18632/oncotarget.1998
    OpenUrlCrossRefPubMed
  8. ↵
    1. Martin TA YL,
    2. Sanders AJ,
    3. Lane J,
    4. Jiang WG
    : Cancer invasion and metastasis: Molecular and cellular perspective, metastatic cancer clinical and biological perspectives, Austin, TX, USA: Landes Bioscience, 34, 2013.
  9. ↵
    1. Spano D,
    2. Heck C,
    3. De Antonellis P,
    4. Christofori G,
    5. Zollo M
    : Molecular networks that regulate cancer metastasis. Semin Cancer Biol 22: 234-249, 2012. PMID: 22484561, DOI: 10.1016/j.semcancer.2012.03.006
    OpenUrlCrossRefPubMed
  10. ↵
    1. Hainaut P,
    2. Plymoth A
    : Targeting the hallmarks of cancer: towards a rational approach to next-generation cancer therapy. Curr Opin Oncol 25: 50-51, 2013. PMID: 23150341, DOI: 10.1097/CCO.0b013e32835b651e
    OpenUrlCrossRefPubMed
  11. ↵
    1. Steeg PS
    : Tumor metastasis: mechanistic insights and clinical challenges. Nat Med 12: 895-904, 2006. PMID: 16892035, DOI: 10.1038/nm1469
    OpenUrlCrossRefPubMed
  12. ↵
    1. Babykutty S,
    2. S PP,
    3. J NR,
    4. Kumar MA,
    5. Nair MS,
    6. Srinivas P,
    7. Gopala S
    : Nimbolide retards tumor cell migration, invasion, and angiogenesis by downregulating MMP-2/9 expression via inhibiting ERK1/2 and reducing DNA-binding activity of NF-kappaB in colon cancer cells. Mol Carcinog 51: 475-490, 2012. PMID: 21678498, DOI: 10.1002/mc.20812
    OpenUrlCrossRefPubMed
    1. Deryugina EI,
    2. Quigley JP
    : Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev 25: 9-34, 2006. PMID: 16680569, DOI: 10.1007/s10555-006-7886-9
    OpenUrlCrossRefPubMed
  13. ↵
    1. Bendardaf R,
    2. Buhmeida A,
    3. Hilska M,
    4. Laato M,
    5. Syrjanen S,
    6. Syrjanen K,
    7. Collan Y,
    8. Pyrhonen S
    : MMP-9 (gelatinase B) expression is associated with disease-free survival and disease-specific survival in colorectal cancer patients. Cancer Invest 28: 38-43, 2010. PMID: 20001295, DOI: 10.3109/07357900 802672761
    OpenUrlCrossRefPubMed
  14. ↵
    1. Rosol TJ
    : Pathogenesis of bone metastases: role of tumor-related proteins. J Bone Miner Res 15: 844-850, 2000. PMID: 10804013, DOI: 10.1359/jbmr.2000.15.5.844
    OpenUrlCrossRefPubMed
  15. ↵
    1. Santini D,
    2. Galluzzo S,
    3. Zoccoli A,
    4. Pantano F,
    5. Fratto ME,
    6. Vincenzi B,
    7. Lombardi L,
    8. Gucciardino C,
    9. Silvestris N,
    10. Riva E,
    11. Rizzo S,
    12. Russo A,
    13. Maiello E,
    14. Colucci G,
    15. Tonini G
    : New molecular targets in bone metastases. Cancer Treat Rev 36: S6-s10, 2010. PMID: 21129612, DOI: 10.1016/s0305-7372(10) 70013-x
    OpenUrlCrossRefPubMed
  16. ↵
    1. Newman DJ,
    2. Cragg GM
    : Natural Products as Sources of New Drugs from 1981 to 2014. J Nat Prod 79: 629-661, 2016. PMID: 26852623, DOI: 10.1021/acs.jnatprod.5b01055
    OpenUrlCrossRefPubMed
  17. ↵
    1. Khan N,
    2. Asim M,
    3. Afaq F,
    4. Abu Zaid M,
    5. Mukhtar H
    : A novel dietary flavonoid fisetin inhibits androgen receptor signaling and tumor growth in athymic nude mice. Cancer Res 68: 8555-8563, 2008. PMID: 18922931, DOI: 10.1158/0008-5472.can-08-0240
    OpenUrlAbstract/FREE Full Text
    1. Adhami VM,
    2. Syed DN,
    3. Khan N,
    4. Mukhtar H
    : Dietary flavonoid fisetin: a novel dual inhibitor of PI3K/Akt and mTOR for prostate cancer management. Biochem Pharmacol 84: 1277-1281, 2012. PMID: 22842629, DOI: 10.1016/j.bcp.2012.07.012
    OpenUrlCrossRefPubMed
  18. ↵
    1. Mukhtar E,
    2. Adhami VM,
    3. Khan N,
    4. Mukhtar H
    : Apoptosis and autophagy induction as mechanism of cancer prevention by naturally occurring dietary agents. Curr Drug Targets 13: 1831-1841, 2012. PMID: 23140293.
    OpenUrl
  19. ↵
    1. Su CH,
    2. Kuo CL,
    3. Lu KW,
    4. Yu FS,
    5. Ma YS,
    6. Yang JL,
    7. Chu YL,
    8. Chueh FS,
    9. Liu KC,
    10. Chung JG
    : Fisetin-induced apoptosis of human oral cancer SCC-4 cells through reactive oxygen species production, endoplasmic reticulum stress, caspase-, and mitochondria-dependent signaling pathways. Environ Toxicol 32: 1725-1741, 2017. PMID: 28181380, DOI: 10.1002/tox.22396
    OpenUrl
  20. ↵
    1. Shih YL,
    2. Hung FM,
    3. Lee CH,
    4. Yeh MY,
    5. Lee MH,
    6. Lu HF,
    7. Chen YL,
    8. Liu JY,
    9. Chung JG
    : Fisetin induces apoptosis of HSC3 human oral cancer cells through endoplasmic reticulum stress and dysfunction of mitochondria-mediated signaling pathways. In Vivo 31: 1103-1114, 2017. PMID: 29102932, DOI: 10.21873/invivo.11176
    OpenUrlAbstract/FREE Full Text
  21. ↵
    1. Li R,
    2. Zhao Y,
    3. Chen J,
    4. Shao S,
    5. Zhang X
    : Fisetin inhibits migration, invasion and epithelial-mesenchymal transition of LMP1-positive nasopharyngeal carcinoma cells. Mol Med Rep 9: 413-418, 2014. PMID: 24297333, DOI: 10.3892/mmr.2013.1836
    OpenUrl
  22. ↵
    1. Chou RH,
    2. Hsieh SC,
    3. Yu YL,
    4. Huang MH,
    5. Huang YC,
    6. Hsieh YH
    : Fisetin inhibits migration and invasion of human cervical cancer cells by down-regulating urokinase plasminogen activator expression through suppressing the p38 MAPK-dependent NF-kappaB signaling pathway. PLoS One 8: e71983, 2013. PMID: 23940799, DOI: 10.1371/journal.pone.0071983
    OpenUrl
  23. ↵
    1. Wang J,
    2. Huang S
    : Fisetin inhibits the growth and migration in the A549 human lung cancer cell line via the ERK1/2 pathway. Exp Ther Med 15: 2667-2673, 2018. PMID: 29467859, DOI: 10.3892/etm.2017.5666
    OpenUrl
  24. ↵
    1. Lin CC,
    2. Lee MH,
    3. Lin JH,
    4. Lin ML,
    5. Chueh FS,
    6. Yu CC,
    7. Lin JP,
    8. Chou YC,
    9. Hsu SC,
    10. Chung JG
    : Crude extract of Rheum palmatum L. Induces cell cycle arrest S phase and apoptosis through mitochondrial-dependent pathways in U-2 OS human osteosarcoma cells. Environ Toxicol 31: 957-969, 2016. PMID: 25689151, DOI: 10.1002/tox.22105
    OpenUrl
  25. ↵
    1. Shih YL,
    2. Au MK,
    3. Liu KL,
    4. Yeh MY,
    5. Lee CH,
    6. Lee MH,
    7. Lu HF,
    8. Yang JL,
    9. Wu RS,
    10. Chung JG
    : Ouabain impairs cell migration, and invasion and alters gene expression of human osteosarcoma U-2 OS cells. Environ Toxicol 32: 2400-2413, 2017. PMID: 28795476, DOI: 10.1002/tox.22453
    OpenUrl
  26. ↵
    1. Huang YP,
    2. Chang NW
    : PPARalpha modulates gene expression profiles of mitochondrial energy metabolism in oral tumorigenesis. BioMedicine 6: 3, 2016. PMID: 26869356, DOI: 10.7603/s40681-016-0003-7
    OpenUrl
  27. ↵
    1. Ma YS,
    2. Hsiao YT,
    3. Lin JJ,
    4. Liao CL,
    5. Lin CC,
    6. Chung JG
    : Phenethyl isothiocyanate (PEITC) and benzyl isothiocyanate (BITC) inhibit human melanoma A375.S2 cell migration and invasion by affecting MAPK signaling pathway in vitro. Anticancer Res 37: 6223-6234, 2017. PMID: 29061805, DOI: 10.21873/anticanres.12073
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Chan CY,
    2. Lien CH,
    3. Lee MF,
    4. Huang CY
    : Quercetin suppresses cellular migration and invasion in human head and neck squamous cell carcinoma (HNSCC). BioMedicine 6: 15, 2016. PMID: 27510965, DOI: 10.7603/s40681-016-0015-3
    OpenUrl
  29. ↵
    1. Lin YJ,
    2. Ho TJ,
    3. Lin TH,
    4. Hsu WY,
    5. Huang SM,
    6. Liao CC,
    7. Lai CH,
    8. Liu X,
    9. Tsang H,
    10. Lai CC,
    11. Tsai FJ
    : P-coumaric acid regulates exon 12 splicing of the ATP7B gene by modulating hnRNP A1 protein expressions. BioMedicine 5: 1-9, 2015. PMID: 26048696, DOI: 10.7603/s40681-015-0010-0
    OpenUrl
  30. ↵
    1. Cao J,
    2. Han X,
    3. Qi X,
    4. Jin X,
    5. Li X
    : TUG1 promotes osteosarcoma tumorigenesis by upregulating EZH2 expression via miR-144-3p. Int J Oncol 51: 1115-1123, 2017. PMID: 28902349, DOI: 10.3892/ijo.2017.4110
    OpenUrl
  31. ↵
    1. Cai L,
    2. Lv J,
    3. Zhang Y,
    4. Li J,
    5. Wang Y,
    6. Yang H
    : The lncRNA HNF1A-AS1 is a negative prognostic factor and promotes tumorigenesis in osteosarcoma. J Cell Mol Med 21: 2654-2662, 2017. PMID: 28866868, DOI: 10.1111/jcmm.12944
    OpenUrl
  32. ↵
    1. Zhu W,
    2. Liang Q,
    3. Yang X,
    4. Yu Y,
    5. Shen X,
    6. Sun G
    : Combination of sorafenib and Valproic acid synergistically induces cell apoptosis and inhibits hepatocellular carcinoma growth via down-regulating Notch3 and pAkt. Am J Cancer Res 7: 2503-2514, 2017. PMID: 29312803.
    OpenUrl
    1. Zanotto-Filho A,
    2. Rajamanickam S,
    3. Loranc E,
    4. Masamsetti VP,
    5. Gorthi A,
    6. Romero JC,
    7. Tonapi S,
    8. Goncalves RM,
    9. Reddick RL,
    10. Benavides R,
    11. Kuhn J,
    12. Chen Y,
    13. Bishop AJR
    : Sorafenib improves alkylating therapy by blocking induced inflammation, invasion and angiogenesis in breast cancer cells. Cancer Lett 425: 101-115, 2018. PMID: 29608984, DOI: 10.1016/j.canlet. 2018.03.037
    OpenUrl
  33. ↵
    1. Yin P,
    2. Song G,
    3. Jiang Z
    : Cisplatin suppresses proliferation, migration and invasion of nasopharyngeal carcinoma cells in vitro by repressing the Wnt/beta-catenin/Endothelin-1 axis via activating B cell translocation gene 1. Cancer Chemother Pharmacol 81: 863-872, 2018. PMID: 29536130, DOI: 10.1007/s00280-018-3536-5
    OpenUrl
  34. ↵
    1. Shi C,
    2. Zhang N,
    3. Feng Y,
    4. Cao J,
    5. Chen X,
    6. Liu B
    : Aspirin inhibits IKK-beta-mediated prostate cancer cell invasion by targeting matrix metalloproteinase-9 and urokinase-type plasminogen activator. Cell Physiol Biochem 41: 1313-1324, 2017. PMID: 28278500, DOI: 10.1159/000464434
    OpenUrl
  35. ↵
    1. Noh EM,
    2. Park YJ,
    3. Kim JM,
    4. Kim MS,
    5. Kim HR,
    6. Song HK,
    7. Hong OY,
    8. So HS,
    9. Yang SH,
    10. Kim JS,
    11. Park SH,
    12. Youn HJ,
    13. You YO,
    14. Choi KB,
    15. Kwon KB,
    16. Lee YR
    : Fisetin regulates TPA-induced breast cell invasion by suppressing matrix metalloproteinase-9 activation via the PKC/ROS/MAPK pathways. Eur J Pharmacol 764: 79-86, 2015. PMID: 26101063, DOI: 10.1016/j.ejphar. 2015.06.038
    OpenUrl
  36. ↵
    1. Liao YC,
    2. Shih YW,
    3. Chao CH,
    4. Lee XY,
    5. Chiang TA
    : Involvement of the ERK signaling pathway in fisetin reduces invasion and migration in the human lung cancer cell line A549. J Agric Food Chem 57: 8933-8941, 2009. PMID: 19725538, DOI: 10.1021/jf902630w
    OpenUrlCrossRefPubMed
  37. ↵
    1. Wu Y,
    2. Zhou BP
    : New insights of epithelial-mesenchymal transition in cancer metastasis. Acta Biochim Biophys Sin (Shanghai) 40: 643-650, 2008. PMID: 18604456.
    OpenUrlCrossRefPubMed
  38. ↵
    1. Labernadie A,
    2. Kato T,
    3. Brugues A,
    4. Serra-Picamal X,
    5. Derzsi S,
    6. Arwert E,
    7. Weston A,
    8. Gonzalez-Tarrago V,
    9. Elosegui-Artola A,
    10. Albertazzi L,
    11. Alcaraz J,
    12. Roca-Cusachs P,
    13. Sahai E,
    14. Trepat X
    : A mechanically active heterotypic E-cadherin/N-cadherin adhesion enables fibroblasts to drive cancer cell invasion. Nat Cell Biol 19: 224-237, 2017. PMID: 28218910, DOI: 10.1038/ncb3478
    OpenUrlCrossRef
  39. ↵
    1. Kim D,
    2. Kim S,
    3. Koh H,
    4. Yoon SO,
    5. Chung AS,
    6. Cho KS,
    7. Chung J
    : Akt/PKB promotes cancer cell invasion via increased motility and metalloproteinase production. FASEB J 15: 1953-1962, 2001. PMID: 11532975, DOI: 10.1096/fj.01-0198com
    OpenUrlCrossRefPubMed
  40. ↵
    1. Mittal V,
    2. Linder ME
    : Biochemical characterization of RGS14: RGS14 activity towards G-protein alpha subunits is independent of its binding to Rap2A. Biochem J 394: 309-315, 2006. PMID: 16246175, DOI: 10.1042/bj20051086
    OpenUrlAbstract/FREE Full Text
  41. ↵
    1. Albright CF,
    2. Giddings BW,
    3. Liu J,
    4. Vito M,
    5. Weinberg RA
    : Characterization of a guanine nucleotide dissociation stimulator for a ras-related GTPase. Embo J 12: 339-347, 1993. PMID: 8094051.
    OpenUrlPubMed
  42. ↵
    1. Tran NL,
    2. Nagle RB,
    3. Cress AE,
    4. Heimark RL
    : N-Cadherin expression in human prostate carcinoma cell lines. An epithelial-mesenchymal transformation mediating adhesion withStromal cells. Am J Pathol 155: 787-798, 1999. PMID: 10487836, DOI: 10.1016/s0002-9440(10)65177-2
    OpenUrlCrossRefPubMed
  43. ↵
    1. Canel M,
    2. Serrels A,
    3. Frame MC,
    4. Brunton VG
    : E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci 126: 393-401, 2013. PMID: 23525005, DOI: 10.1242/jcs.100115
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Park JH,
    2. Jang YJ,
    3. Choi YJ,
    4. Jang JW,
    5. Kim JH,
    6. Rho YK,
    7. Kim IJ,
    8. Kim HJ,
    9. Leem MJ,
    10. Lee ST
    : Fisetin inhibits matrix metalloproteinases and reduces tumor cell invasiveness and endothelial cell tube formation. Nutr Cancer 65: 1192-1199, 2013. PMID: 24099040, DOI: 10.1080/01635581.2013.828090
    OpenUrl
  45. ↵
    1. Veit C,
    2. Genze F,
    3. Menke A,
    4. Hoeffert S,
    5. Gress TM,
    6. Gierschik P,
    7. Giehl K
    : Activation of phosphatidylinositol 3-kinase and extracellular signal-regulated kinase is required for glial cell line-derived neurotrophic factor-induced migration and invasion of pancreatic carcinoma cells. Cancer Res 64: 5291-5300, 2004. PMID: 15289335, DOI: 10.1158/0008-5472.can-04-1112
    OpenUrlAbstract/FREE Full Text
  46. ↵
    1. Senapati S,
    2. Rachagani S,
    3. Chaudhary K,
    4. Johansson SL,
    5. Singh RK,
    6. Batra SK
    : Overexpression of macrophage inhibitory cytokine-1 induces metastasis of human prostate cancer cells through the FAK-RhoA signaling pathway. Oncogene 29: 1293-1302, 2010. PMID: 19946339, DOI: 10.1038/onc.2009.420
    OpenUrlCrossRefPubMed
  47. ↵
    1. Shukla S,
    2. Maclennan GT,
    3. Hartman DJ,
    4. Fu P,
    5. Resnick MI,
    6. Gupta S
    : Activation of PI3K-Akt signaling pathway promotes prostate cancer cell invasion. Int J Cancer 121: 1424-1432, 2007. PMID: 17551921, DOI: 10.1002/ijc.22862
    OpenUrlCrossRefPubMed
  48. ↵
    1. Shih YL,
    2. Chou HM,
    3. Chou HC,
    4. Lu HF,
    5. Chu YL,
    6. Shang HS,
    7. Chung JG
    : Casticin impairs cell migration and invasion of mouse melanoma B16F10 cells via PI3K/AKT and NF-kappaB signaling pathways. Environ Toxicol 32: 2097-2112, 2017. PMID: 28444820, DOI: 10.1002/tox.22417
    OpenUrlCrossRefPubMed
  49. ↵
    1. Rosenberg L,
    2. Yoon CH,
    3. Sharma G,
    4. Bertagnolli MM,
    5. Cho NL
    : Sorafenib inhibits proliferation and invasion in desmoid-derived cells by targeting Ras/MEK/ERK and PI3K/Akt/mTOR pathways. Carcinogenesis 39: 681-688, 2018. PMID: 29538717, DOI: 10.1093/carcin/bgy038
    OpenUrl
  50. ↵
    1. Perkins ND
    : The diverse and complex roles of NF-kappaB subunits in cancer. Nat Rev Cancer 12: 121-132, 2012. PMID: 22257950, DOI: 10.1038/nrc3204
    OpenUrlCrossRefPubMed
  51. ↵
    1. Guo X,
    2. Zheng L,
    3. Jiang J,
    4. Zhao Y,
    5. Wang X,
    6. Shen M,
    7. Zhu F,
    8. Tian R,
    9. Shi C,
    10. Xu M,
    11. Li X,
    12. Peng F,
    13. Zhang H,
    14. Feng Y,
    15. Xie Y,
    16. Xu X,
    17. Jia W,
    18. He R,
    19. Xie C,
    20. Hu J,
    21. Ye D,
    22. Wang M,
    23. Qin R
    : Blocking NF-kappaB is essential for the immunotherapeutic effect of recombinant IL18 in pancreatic cancer. Clin Cancer Res 22: 5939-5950, 2016. PMID: 27297583, DOI: 10.1158/1078-0432.ccr-15-1144
    OpenUrlAbstract/FREE Full Text
  52. ↵
    1. Hsu HH,
    2. Hu WS,
    3. Lin YM,
    4. Kuo WW,
    5. Chen LM,
    6. Chen WK,
    7. Hwang JM,
    8. Tsai FJ,
    9. Liu CJ,
    10. Huang CY
    : JNK suppression is essential for 17beta-Estradiol inhibits prostaglandin E2-Induced uPA and MMP-9 expressions and cell migration in human LoVo colon cancer cells. J Biomed Sci 18: 61, 2011. PMID: 21859479, DOI: 10.1186/1423-0127-18-61
    OpenUrlPubMed
PreviousNext
Back to top

In this issue

In Vivo
Vol. 33, Issue 3
May-June 2019
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on In Vivo.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Fistein Suppresses Human Osteosarcoma U-2 OS Cell Migration and Invasion via Affecting FAK, uPA and NF-ĸB Signaling Pathway In Vitro
(Your Name) has sent you a message from In Vivo
(Your Name) thought you would like to see the In Vivo web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
2 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Fistein Suppresses Human Osteosarcoma U-2 OS Cell Migration and Invasion via Affecting FAK, uPA and NF-ĸB Signaling Pathway In Vitro
JR-KAI CHEN, SHU-FEN PENG, KUANG CHI LAI, HSIN-CHUNG LIU, YI-PING HUANG, CHIN-CHUNG LIN, AN-CHENG HUANG, FU-SHIN CHUEH, JING-GUNG CHUNG
In Vivo May 2019, 33 (3) 801-810; DOI: 10.21873/invivo.11542

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Fistein Suppresses Human Osteosarcoma U-2 OS Cell Migration and Invasion via Affecting FAK, uPA and NF-ĸB Signaling Pathway In Vitro
JR-KAI CHEN, SHU-FEN PENG, KUANG CHI LAI, HSIN-CHUNG LIU, YI-PING HUANG, CHIN-CHUNG LIN, AN-CHENG HUANG, FU-SHIN CHUEH, JING-GUNG CHUNG
In Vivo May 2019, 33 (3) 801-810; DOI: 10.21873/invivo.11542
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Sorafenib Induces Apoptosis and Inhibits NF-{kappa}B-mediated Anti-apoptotic and Metastatic Potential in Osteosarcoma Cells
  • Google Scholar

More in this TOC Section

  • Evaluation of the Relationship Between miRNA-22-3p and Gal-9 Levels in Glioblastoma
  • Metformin Inhibits the Estrogen-mediated Epithelial-Mesenchymal Transition of Ectopic Endometrial Stromal Cells in Endometriosis
  • MCC950 Ameliorates Acute Exogenous Lipoid Pneumonia Induced by Sewing Machine Oil in Rats via the NF-κB/NLRP3 Inflammasome Pathway
Show more Experimental Studies

Similar Articles

Keywords

  • Fisetin
  • human osteosarcoma U-2 OS cell
  • Migration
  • invasion
  • metastasis
In Vivo

© 2023 In Vivo

Powered by HighWire