Next Article in Journal
Physicochemical Properties and Oxidative Storage Stability of Milled Roselle (Hibiscus sabdariffa L.) Seeds
Next Article in Special Issue
Antiangiogenic Potential of Microbial Metabolite Elaiophylin for Targeting Tumor Angiogenesis
Previous Article in Journal
Two New Triterpenoids from the Roots of Codonopsis pilosula
Previous Article in Special Issue
Anti-Tumor and Radiosensitization Effects of N-Butylidenephthalide on Human Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Wogonin Suppresses the Activity of Matrix Metalloproteinase-9 and Inhibits Migration and Invasion in Human Hepatocellular Carcinoma

1
Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, 12 Jichang Road, Guangzhou 510405, China
2
College of mechanical engineering, Yangzhou University, 88 South University Ave., Yangzhou 225009, China
3
Department of Genome Engineering, Beijing Institute of Basic Medical Sciences, Taiping Road 27, Beijing 100850, China
*
Authors to whom correspondence should be addressed.
These authors contribute equally to this work.
Molecules 2018, 23(2), 384; https://doi.org/10.3390/molecules23020384
Submission received: 31 December 2017 / Revised: 22 January 2018 / Accepted: 7 February 2018 / Published: 11 February 2018
(This article belongs to the Collection Natural Products: Anticancer Potential and Beyond)

Abstract

:
As one of the major active ingredients in Radix Scutellariae, wogonin has been shown to be associated with various pharmacological activities on cancer cell growth, apoptosis, and cell invasion and migration. Here, we demonstrated that wogonin may harbor potential anti-metastatic activities in hepatocarcinoma (HCC). The anti-metastasis potential of wogonin and its underlying mechanisms were evaluated by ligand–protein docking approach, surface plasmon resonance assay, and in vitro gelatin zymography studies. Our results showed that wogonin (100 μM, 50 μM) suppressed MHCC97L and PLC/PRF/5 cells migration and invasion in vitro. The docking approach and surface plasmon resonance assay indicated that the potential binding affinity between wogonin and matrix metalloproteinase-9 (MMP-9) may lead to inhibition of MMP-9 activity and further leads to suppression of tumor metastasis. This conclusion was further verified by Western blot results and gelatin zymography analysis. Wogonin might be a potent treatment option for disrupting the tumor metastasis that favors HCC development. The potential active targets from computational screening integrated with biomedical study may help us to explore the molecular mechanism of herbal medicines.

Graphical Abstract

1. Introduction

Hepatocellular carcinoma (HCC), also called malignant hepatoma, is the most common type of hepatic cancer. Despite the great progress in novel drug therapy and hepatic surgical technologies, the five-year survival rates for HCC patients remained dismally below 20% due to tumor recurrence and distance metastasis after treatment [1,2]. Although the recently developed target-therapy drugs such as Sorafenib (a kinase inhibitor agent) have been used in clinic for prolonging survival in patients with advanced HCC, their therapeutic potential to date is still limited due to their high cost-effectiveness ratio in developing countries [3,4] and the significant intolerable side effects such as serious diarrhea, skin toxicities, and hypertension [5,6]. In this case, much effort has been put into exploring novel therapies to increase the efficacy of anti-HCC drugs and decrease side effects as well as improve quality of life. The extensive application of novel biomedical and phytochemical approaches in many herbal-derived ingredients has shown great potential in developing novel anti-HCC natural products [7,8,9].
Wogonin (5,7-dihydroxy-8-methoxyflavone), a flavonoid-like chemical compound found in Radix Scutellariae, is a well-known herbal agent which has shown remarkable anticarcinogenic and chemopreventive capacity in various studies [10,11,12]. In recent years, numerous evidences have demonstrated that wogonin effectively inhibited the invasion and migration of human gallbladder carcinoma [13] and breast cancer [14]. In addition, wogonin exhibit inhibition effects on intracellular MMPs activity in human aortic smooth muscle cells [15]. However, the potential effect of wogonin on hepatocellular carcinoma (HCC) has not been fully elucidated so far. Therefore, we would like to investigate the potential anti-migration and anti-invasion effect of wogonin on HCC and discover the molecular mechanism of action in the present study.
The rapid progresses in physic and computational technologies have provided effective strategies for exploring the molecular mechanism of herbal medicines. Molecular docking is such a rational approach for drug discovery, which is more effective than regular experimental drug screen. The molecular docking approach based on the theory of “lock-and-key” [16]. During the phytomedicine development, the binding affinity is an important indicator for screening active components or targets. Several docking programs such as Sybyl-X can predict the binding affinity between the ligand and target by using score function [17,18]. Most scoring functions are based on the energy of the pose within the binding sites. A well designed scoring function program is important for molecular docking and rapid screening of the potential ligands or protein targets [19]. The binding affinity between herbal ingredients and intracellular proteins play an important role in cell signal transduction, thus, it will be useful for predicting the mechanism of action of herbal medicines. In this study, we explored the mechanism of the possible anti-invasion and anti-migration effect of wogonin in HCC cells used a molecular docking-based screening approach. The candidate targets were further verified by surface plasmon resonance (SPR) assay in vitro. Furthermore, Western blot and gelatin zymography assay were investigated to explore the molecular mechanism account for the inhibition effects by wogonin.

2. Results

2.1. Wogonin Inhibits MHCC97L, PLC/PRF/5 Cells Migration and Invasion at Non-Toxic Dosage In Vitro

Based on previous studies, we first evaluated the dose-dependent cytotoxicity of wogonin on MHCC97L, PLC/PRF/5, and normal hepatocyte LO2 cell lines by methyl thiazolyl tetrazolium (MTT) assay. Wogonin decreased the cell viability in a dose-dependent manner, and the IC50 against MHCC97L, PLC/PRF/5 and LO2 cells at 48 h was 486 µM, 431 µM and 424 µM, respectively (Figure 1A). To examine the invasion and migration ability of HCC cells, we performed in vitro wound healing assay and transwell chambers assay. The wound area in 100 μM and 50 μM wogonin treatment group was significantly increased than that in the non-treated group after 48 h of incubation (p < 0.05, n = 5) (Figure 1B). Matrigel matrix invasion assay further indicated that wogonin are potent inhibitors for HCC cells invasion, which is independent to their cytotoxic effects (Figure 1C). These result suggested that the capacity of invasiveness was reduced when treated with a non-toxic dosage of wogonin.

2.2. Candidate Target Proteins Screening and Binding Affinity Measurement

After applying the molecular docking approach, four metastasis-related targets (Human matrix metalloprotein 9 (MMP-9), Intercellular Adhesion Molecule 1 (ICAM-1), Stromal interaction molecule 1 (STIM1), and Fibronectin 1 (FN1)) were mapped out due to its potential binding affinity to wogonin (Table 1). The protein candidates which exhibit strong interaction with wogonin may have the potential of inhibiting metastasis of HCC. According to the visual ligand–protein docking results, we used an in silico analysis to determine the binding site of wogonin with MMP-9. The inhibitory activities were confirmed with molecular docking analysis when considering crystallographic water molecules in the ligand-binding pocket of MMP-9 (Figure 2B). Based on these docking results, deeper investigation on the interaction between the candidate targets and wogonin were further explored by SPR-based binding analysis. As shown in Figure 2D,E and Table 2, MMP-9 bound to wogonin (KD = 16.27 μM) much tighter than other candidate targets. These results demonstrated that the binding of wogonin to the active site of MMP-9 might inhibit its activation and suppress HCC migration and invasion.

2.3. Wogonin Down-Regulates MMP-9 Activity without Effecting its Protein Expression Level

Based on our molecular docking and SPR results, the activity of MMPs was further detected with Gelatin Zymography assay. Wogonin could inhibit intracellular active MMP-9 activities in dose- and time-dependent manner in MHCC97L cells while no inhibition effects have been observed in pro-MMP-9 and MMP-2 activities. (Figure 3A). These observations indicated that the anti-migration and anti-invasion action of wogonin might mainly attributed by the suppression of secreted MMP-9 activity. However, wogonin had no significant effect on the protein expression of MMP-9 (Figure 3B). These results demonstrated that the potential binding affinity between wogonin and MMP-9 may lead to inhibition of MMP-9 activity and further leads to suppression of tumor metastasis.

3. Discussion

Wogonin have been shown to possess antitumor effects in various cancer cells, such as anti-proliferation, cell cycle arrest, induction autophagy-related apoptosis, and inhibition of angiogenesis. [20,21,22]. In the present study, we demonstrated that wogonin can significantly inhibit HCC cell migration and invasion in vitro. Although previous studies have shown that 80 µM wogonin may induce apoptosis in HCC cell lines [11], our study indicated that a lower dosage (50 µM) could also inhibit cell invasion and migration in vitro. In that case, we speculated that non-toxic concentration of wogonin may exhibit anti-cancer activity in HCC cells. The molecular docking approach, SPR assay, and gelatin zymography analysis indicated that the mechanism of action might partly proceed through suppressing the active site of MMP-9 in HCC cells. Pervious research results showed that wogonin could inhibit the invasion of human breast carcinoma cells by down-regulating the protein expression of MMP-9 [14]. However, in our study, wononin has no effect on the protein expression of MMP-9 in HCC cells. The docking results and SPR analysis indicated that wogonin may inhibit MMP-9 activity without decreasing protein expression. HCC cell metastasis involves multiple processes and various cytophysiological changes, such as the adhesive properties change between the extracellularmatrix (ECM) and cancer cells [23,24]. This involves proteolytic degradation and impaired intracellular interactions. Thus, the proteinases such as MMP-9 induced degradation of the ECM and components of the basement membrane play a critical role in HCC invasion and migration. The activity of MMP-9 can be modulated by various endogenous or exogenous inhibitors [25,26]. Our study suggested that wogonin might be a potential exogenous inhibitor for MMP-9. For future studies, further identification should be done to determine the effects of metabolites of wogonin in vivo. Orthotopical transplantation animal HCC model for evaluating the interaction of MMP-9 and wogonin in vivo are required in our future studies. In addition, the MMP-9 site directed mutagenesis or CRISPR/Cas9 experiments would be applied in our next study to deeply elaborate the anti-metastasis mechanism of this herbal agent.
As comprehensive analysis the active mechanisms of herbal products at molecular level are time consuming and expensive, computational approaches may compensate for such drawbacks [27,28]. In recent years, a large number of three dimensional structure data of chemical compounds and intracellular proteins have been revealed. As a result, novel drug discovery processes such as molecular docking studies have been widely applied in intracellular target screening of new discovered bioactive chemical agents. To the best of our knowledge, for the first time, we discovered the potential anti-metastasis mechanisms of wogonin by a biomedical investigation integrated with molecular docking. This in silico tool integrated with in vitro investigation may break through the bottleneck on the mechanism study of herbal products and promote the development of novel anti-HCC phytochemicals. In the future study, we will further explore the anti-cancer effects of wogonin and its underlying mechanism by an in-depth study such as using an orthotopic transplantation model to study the in vivo activity of wogonin.

4. Methods and Materials

4.1. Cell Culture and Cell Viability Assay

Human hepatocellular carcinoma cell line MHCC97L, PLC/PRF/5 and human normal hepatocyte line LO2 were subscribed from American Type Culture Collection (ATCC, Manassas, VA, USA) and preserved in liquid nitrogen. The cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Hyclone, Logan, UT, USA) supplemented with antibiotics (100 U/mL penicillin, 100 U/mL streptomycin) and 10% Fetal Bovine Serum (FBS), in a humidified incubator (Sanyo Electric, Osaka, Japan) at 37 °C containing 5% CO2. Cells were collected by disposing with trypsin (Hyclone, USA) after the medium has been refreshed for twice. The cells in the logarithmic growth phase were used for dectcting the cell viability by methyl thiazolyl tetrazolium (MTT) assay. The MHCC97L and LO2 cells were treated with serial concentrations of wogonin for 24 h. At the end of the incubation, 10 μL of MTT solution(Sigma-Aldrich, St. Louis, Missouri, MO, USA) was added to each well and incubated for another 2.5 h. Absorbance was detected at wavelength of 570 nm after 100 μL of DMSO was used to dissolve formazan crystal.

4.2. Cell Migration and Invasion Assay

Cell migration capacity was detected by wound healing assay. The MHCC97L cells and PLC/PRF/5 cells were grown to overspread the culture plates. Then, a 100 µL pipette tips was used to scratch the cell surface. The wounded monolayers were cultured in fetal bovine serum -free media with different doses of wogonin for 48 h after washed by PBS for twice. Wounded healing extent was observed at the indicated time intervals (0 h and 48 h) as previous describes [14]. Cell invasion ability was evaluated by invasion chambers assay [15]. A total of 2 × 105 of HCC cells treated with different dose of wogonin (0, 50, and 100 µM) were added separately onto the apical chambers (FBS negative). The basal chambers were filled with DMEM medium (500 µL) with 10% FBS. The non-invaded HCC cells in the top chamber were gently removed after incubating for 24 h. The invading HCC cells in the chamber bottom were fixed with 100% methanol and stained with 1% crystal violet solution for 15 min. Ten vision fields were chosen randomly per well under a 40 × microscope (Leica, Frankfurt, Germany) and the number of invaded HCC cells was counted. The Data were presented as means ± SD from two independent researchers.

4.3. Molecular Docking Assay

The cancer metastasis related proteins were searched from Potential Drug Target Database (http://www.dddc.ac.cn/pdtd/), DrugBank (http://www.drugbank.ca/), and Therapeutic Targets Database (http://bidd.nus.edu.sg/group/ttd/). A total of 123 candidate proteins related with cancer metastasis were shown in Supplementary Table S1. Before the docking process, the protein crystallographic structures were downloaded from RCSB Protein Data Bank (http://www.pdb.org/) and prepared with Sybyl-X 2.0 (Tripos, St. Louis, MO, USA) for the flexible docking studies [29]. An energy minimized 3D structure of Wogonin (PubChem: 5281703) was optimized from NCBI-PubChem (https://pubchem.ncbi.nlm.nih.gov/). The detailed docking process and reliability validated assay were strictly followed the operation instruction of Surflex-Dock module of Sybyl-X [30,31,32]. The intracellular target proteins with consensus docking score ≥6.0 were chose as the candidate proteins and further verified by AutoDock Vina (http://vina.scripps.edu/).

4.4. Binding Affinity Measurement Using Surface Plasmon Resonance

The candidate targets with satisfactory docking score were further verified by surface plasmon resonance (SPR) assay. SPR binding studies were conducted by Biacore T200 biosensor system (GE Healthcare, Stockholm, Sweden). All the SPR-based materials were acquired from GE Healthcare. Human matrix metalloprotein 9 (MMP-9), Intercellular Adhesion Molecule 1 (ICAM-1), Stromal interaction molecule 1 (STIM1), and Fibronectin 1 (FN1) were purchased from R&D systems (Minneapolis, MN, USA). The proteins were diluted in 10 mM sodium acetate buffer at pH 5.0 and immobilized on a CM5 chip using an amine coupling kit. The immobilization of protein targets on a CM5 chip was performed according to the BIA applications handbook. A total of 8419 RU of immobilized proteins was obtained. Sensor preparation and interaction analyses were performed at 25 °C in a PBS-P (10 mM phosphate buffer, 137 mM NaCl, 2.7 mM KCl, 0.05% P20 (pH 7.4)) running buffer containing 5% DMSO. Tested wogonin were prepared in a two-fold dilution concentration series by running buffer containing 5% DMSO. Solvent-correction procedures were included to compensate for any DMSO-related bulk refractive index variations. Biacore traces were baseline subtracted and the signal was presented in sensorgrams and measured in RU. Empirically in the BIAcore technology, 1 ng of analyte bound at the surface gives a response of 1000 RU. Equilibrium constants (KD) were calculated using the ‘affinity’ model in Biacore T200 evaluation software version 2.0.

4.5. Gelatin Zymography

The gelatin zymography was conducted to detect the activity of matrix metalloproteinases-9 (MMP-9) and matrix metalloproteinases-2 (MMP-2) (Abcam, London, UK). The cell-conditioned media was tested by gelatin zymography assay. Firstly, proteins were separated in 10% SDS-PAGE gelatin gel (2 mg/mL) and ran in running buffer for 20 min at 70 Volt and then 90 min at 120 Volt, after running, the gel was incubated in the 2.5% triton-X on a low-speed shaken platform for 20 min at room temperature. Then the gel was incubated in developing buffer (5 mM CaCl2, 0.2 M NaCl, 50 mM Tris-HCl) for 12 h on a low-speed shaken platform. At the last step, Coomassie Blue R-250 (Sigma-Aldrich, USA) was used as staining dye and washed the gel for 15 min after staining. The results were analyzed using a chemi-luminescence imaging system (Bio-Rad, Hercules, CA, USA).

4.6. Western Blot

MHCC97L and LO2 cells were processed with RIPA buffer on ice for 45 s and then centrifuged at 10,000 rpm at 4 °C for 10 min. The protein concentration of the supernatant of liver homogenate was detected by BSA assay. Equal amounts of protein were resolved by SDS-Polyacrylamide gel electrophoresis and transferred onto Immun-Blot® PVDF Membrane. Then, use 5% BSA in TBST as blocking buffer to block the membrane at room temperature for 4 h. The membrane was cut and each part was incubated with corresponding antibody at 4 °C for 12 h, then, after cleaning the membrane with TBST for 10 min for 3 times, the membrane was further incubated with corresponding secondary antibodies at 24 °C for 3 h. The immunoreactivities were tested by using Electrochemiluminescence Advanced kit (GE Healthcare, Little Chalfont, UK) and the results were analyzed with Bio-Rad chemi-luminescence imaging system. The protein expression levels of MMP-9 and MMP-2 were normalized with GAPDH and quantified by using ImageJ (National Institutes of Health, Bethesda, MD, USA).

4.7. Statistical Analysis

All the data obtained are presented as the mean ± SD, and analyzed by one-way analysis of variance (ANOVA) with Statistical Product and Service Solutions (SPSS) version 19 ( IBM, Armonk, NY, USA). The significance of two groups was examined using a Student’s t-test. Value of p < 0.05 was considered as statistically significant.

Supplementary Materials

The Supplementary Materials are available online.

Acknowledgments

We appreciate the financial support from the Young scientist’s research funding of Guangzhou University of Chinese Medicine (2017).

Author Contributions

Ming Hong and Honghui Cheng completed the experiment, and drafted the manuscript. Lei Song, Wencai Wang, Qi Wang and Donggang Xu analyzed the data. All authors revised and approved on the manuscript and discussed the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. El-Serag, H.B. Hepatocellular carcinoma. N. Engl. J. Med. 2011, 12, 1118–1127. [Google Scholar] [CrossRef] [PubMed]
  2. Hainaut, P.; Boyle, P. Curbing the liver cancer epidemic in Africa. Lancet 2008, 9610, 367–368. [Google Scholar] [CrossRef]
  3. Leung, H.W.; Liu, C.F.; Chan, A.L. Cost-effectiveness of sorafenib versus SBRT for unresectable advanced hepatocellular carcinoma. Radiat. Oncol. 2016, 11, 69. [Google Scholar] [CrossRef] [PubMed]
  4. Johnson, P.; Billingham, L. Sorafenib for liver cancer: The horizon broadens. Lancet Oncol. 2009, 1, 4–5. [Google Scholar] [CrossRef]
  5. Abdel-Rahman, O.; Lamarca, A. Development of sorafenib-related side effects in patients diagnosed with advanced hepatocellular carcinoma treated with sorafenib: A systematic-review and meta-analysis of the impact on survival. Expert. Rev. Gastroenterol. Hepatol. 2017, 1, 75–83. [Google Scholar] [CrossRef] [PubMed]
  6. Berk, V.; Kaplan, M.A.; Tonyali, O.; Buyukberber, S.; Balakan, O.; Ozkan, M.; Demirci, U.; Ozturk, T.; Bilici, A.; Tastekin, D.; et al. Efficiency and side effects of sorafenib therapy for advanced hepatocellular carcinoma: A retrospective study by the anatolian society of medical oncology. Asian Pac. J. Cancer Prev. 2013, 12, 7367–7369. [Google Scholar] [CrossRef]
  7. Tsai, T.Y.; Livneh, H.; Hung, T.H.; Lin, I.H.; Lu, M.C.; Yeh, C.C. Associations between prescribed Chinese herbal medicine and risk of hepatocellular carcinoma in patients with chronic hepatitis B: A nationwide population-based cohort study. BMJ Open 2017, 1, e014571. [Google Scholar] [CrossRef] [PubMed]
  8. Yu, Y.; Lang, Q.; Chen, Z.; Li, B.; Yu, C.; Zhu, D.; Zhai, X.; Ling, C. The efficacy for unresectable hepatocellular carcinoma may be improved by transcatheter arterial chemoembolization in combination with a traditional Chinese herbal medicine formula: A retrospective study. Cancer 2009, 22, 5132–5138. [Google Scholar] [CrossRef] [PubMed]
  9. Hu, Y.; Wang, S.; Wu, X.; Zhang, J.; Chen, R.; Chen, M.; Wang, Y. Chinese herbal medicine-derived compounds for cancer therapy: A focus on hepatocellular carcinoma. J. Ethnopharmacol. 2013, 3, 601–612. [Google Scholar] [CrossRef] [PubMed]
  10. Liu, X.; Tian, S.; Liu, M.; Jian, L.; Zhao, L. Wogonin inhibits the proliferation and invasion, and induces the apoptosis of HepG2 and Bel7402 HCC cells through NFkappaB/Bcl-2, EGFR and EGFR downstream ERK/AKT signaling. Int. J. Mol. Med. 2016, 4, 1250–1256. [Google Scholar] [CrossRef] [PubMed]
  11. Chen, X.M.; Bai, Y.; Zhong, Y.J.; Xie, X.L.; Long, H.W.; Yang, Y.Y.; Wu, S.G.; Jia, Q.; Wang, X.H. Wogonin has multiple anti-cancer effects by regulating c-Myc/SKP2/Fbw7alpha and HDAC1/HDAC2 pathways and inducing apoptosis in human lung adenocarcinoma cell line A549. PLoS ONE 2013, 11, e79201. [Google Scholar]
  12. Dai, Z.J.; Wang, B.F.; Lu, W.F.; Wang, Z.D.; Ma, X.B.; Min, W.L.; Kang, H.F.; Wang, X.J.; Wu, W.Y. Total flavonoids of Scutellaria barbata inhibit invasion of hepatocarcinoma via MMP/TIMP in vitro. Molecules 2013, 1, 934–950. [Google Scholar] [CrossRef] [PubMed]
  13. Dong, P.; Zhang, Y.; Gu, J.; Wu, W.; Li, M.; Yang, J.; Zhang, L.; Lu, J.; Mu, J.; Chen, L.; et al. Wogonin, an active ingredient of Chinese herb medicine Scutellaria baicalensis, inhibits the mobility and invasion of human gallbladder carcinoma GBC-SD cells by inducing the expression of maspin. J. Ethnopharmacol. 2011, 3, 1373–1380. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, P.; Lu, N.; Ling, Y.; Chen, Y.; Hui, H.; Lu, Z.; Song, X.; Li, Z.; You, Q.; Guo, Q. Inhibitory effects of wogonin on the invasion of human breast carcinoma cells by downregulating the expression and activity of matrix metalloproteinase-9. Toxicology 2011, 3, 122–128. [Google Scholar] [CrossRef] [PubMed]
  15. Lee, S.O.; Jeong, Y.J.; Yu, M.H.; Lee, J.W.; Hwangbo, M.H.; Kim, C.H.; Lee, I.S. Wogonin suppresses TNF-alpha-induced MMP-9 expression by blocking the NF-kappaB activation via MAPK signaling pathways in human aortic smooth muscle cells. Biochem. Biophys. Res. Commun. 2006, 1, 118–125. [Google Scholar] [CrossRef] [PubMed]
  16. Yuriev, E.; Ramsland, P.A. Latest developments in molecular docking: 2010–2011 in review. J. Mol. Recognit. 2013, 5, 215–239. [Google Scholar] [CrossRef] [PubMed]
  17. Thai, K.M.; Le, D.P.; Tran, N.V.; Nguyen, T.T.; Tran, T.D.; Le, M.T. Computational assay of Zanamivir binding affinity with original and mutant influenza neuraminidase 9 using molecular docking. J. Theor. Biol. 2015, 385, 31–39. [Google Scholar] [CrossRef] [PubMed]
  18. Joshi, S.D.; More, U.A.; Koli, D.; Kulkarni, M.S.; Nadagouda, M.N.; Aminabhavi, T.M. Synthesis, evaluation and in silico molecular modeling of pyrroyl-1,3,4-thiadiazole inhibitors of InhA. Bioorg. Chem. 2015, 59, 151–167. [Google Scholar] [CrossRef] [PubMed]
  19. Cheng, T.; Li, X.; Li, Y.; Liu, Z.; Wang, R. Comparative assessment of scoring functions on a diverse test set. J. Chem. Inf. Model. 2009, 4, 1079–1093. [Google Scholar] [CrossRef] [PubMed]
  20. Sun, Y.; Zhao, Y.; Wang, X.; Zhao, L.; Li, W.; Ding, Y.; Kong, L.; Guo, Q.; Lu, N. Wogonoside prevents colitis-associated colorectal carcinogenesis and colon cancer progression in inflammation-related microenvironment via inhibiting NF-kappaB activation through PI3K/Akt pathway. Oncotarget 2016, 23, 34300–34315. [Google Scholar]
  21. Wang, Y.; Yin, R.F.; Teng, J.S. Wogonoside induces cell cycle arrest and mitochondrial mediated apoptosis by modulation of Bcl-2 and Bax in osteosarcoma cancer cells. Int. J. Clin. Exp. Pathol. 2015, 1, 63–72. [Google Scholar]
  22. Zhang, L.; Wang, H.; Cong, Z.; Xu, J.; Zhu, J.; Ji, X.; Ding, K. Wogonoside induces autophagy-related apoptosis in human glioblastoma cells. Oncol. Rep. 2014, 3, 1179–1187. [Google Scholar] [CrossRef] [PubMed]
  23. Cheng, X.; Yang, Y.; Fan, Z.; Yu, L.; Bai, H.; Zhou, B.; Wu, X.; Xu, H.; Fang, M.; Shen, A.; et al. MKL1 potentiates lung cancer cell migration and invasion by epigenetically activating MMP9 transcription. Oncogene 2015, 44, 5570–5581. [Google Scholar] [CrossRef] [PubMed]
  24. Dong, D.D.; Zhou, H.; Li, G. ADAM15 targets MMP9 activity to promote lung cancer cell invasion. Oncol. Rep. 2015, 5, 2451–2460. [Google Scholar] [CrossRef] [PubMed]
  25. Nazdik, M.K.; Taheri, M.; Sajjadi, E.; Arsang-Jang, S.; Koohpar, Z.K.; Inoko, H.; Sayad, A. Increased expression ratio of matrix metalloproteinase-9 (MMP9) and tissue inhibitor of matrix metalloproteinase (TIMP-1) RNA levels in Iranian multiple sclerosis patients. Hum. Antibodies 2016, 3–4, 65–70. [Google Scholar] [CrossRef] [PubMed]
  26. Tong, W.; Wang, Q.; Sun, D.; Suo, J. Curcumin suppresses colon cancer cell invasion via AMPK-induced inhibition of NF-kappaB, uPA activator and MMP9. Oncol. Lett. 2016, 5, 4139–4146. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, L.; Du, J.; Dai, Q.; Zhang, H.; Pang, W.; Hu, J. Prediction of anti-tumor chemical probes of a traditional Chinese medicine formula by HPLC fingerprinting combined with molecular docking. Eur. J. Med. Chem. 2014, 83, 294–306. [Google Scholar] [CrossRef] [PubMed]
  28. Mo, S.L.; Liu, W.F.; Li, C.G.; Zhou, Z.W.; Luo, H.B.; Chew, H.; Liang, J.; Zhou, S.F. Pharmacophore, QSAR, and binding mode studies of substrates of human cytochrome P450 2D6 (CYP2D6) using molecular docking and virtual mutations and an application to chinese herbal medicine screening. Curr. Pharm. Biotechnol. 2012, 9, 1640–1704. [Google Scholar] [CrossRef]
  29. El-Saadi, M.W.; Williams-Hart, T.; Salvatore, B.A.; Mahdavian, E. Use of in-silico assays to characterize the ADMET profile and identify potential therapeutic targets of fusarochromanone, a novel anti-cancer agent. Silico Pharmacol. 2015, 1, 6. [Google Scholar] [CrossRef] [PubMed]
  30. Gao, J.; Liang, L.; Zhu, Y.; Qiu, S.; Wang, T.; Zhang, L. Ligand and Structure-Based Approaches for the Identification of Peptide Deformylase Inhibitors as Antibacterial Drugs. Int. J. Mol. Sci. 2016, 7, 1141. [Google Scholar] [CrossRef] [PubMed]
  31. Renner, S.; Derksen, S.; Radestock, S.; Morchen, F. Maximum common binding modes (MCBM): Consensus docking scoring using multiple ligand information and interaction fingerprints. J. Chem. Inf. Model. 2008, 2, 319–332. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, R.; Lu, Y.; Wang, S. Comparative evaluation of 11 scoring functions for molecular docking. J. Med. Chem. 2003, 12, 2287–2303. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are available from the authors.
Figure 1. Wogonin inhibited invasion and migration in a dose-dependent manner in MHCC97L and PLC/PRF/5 cell lines. (A) Wogonin decreased the cell viability in a dose-dependent manner, and the IC50 against MHCC97L and LO2 cells at 48 h was 486 µM and 424 µM, respectively; (B) HCC cells were scratched and incubated with wogonin (0, 50, and 100 µM). The scar closure area was detected by measuring the distance of the wound surface healing after treatment; (C) After treating with wogonin (0, 25, and 50 µM) for 24 h, the cell invasion ability was significantly inhibited in a dose-dependent manner (* p < 0.05 versus no wogonin treatment group).
Figure 1. Wogonin inhibited invasion and migration in a dose-dependent manner in MHCC97L and PLC/PRF/5 cell lines. (A) Wogonin decreased the cell viability in a dose-dependent manner, and the IC50 against MHCC97L and LO2 cells at 48 h was 486 µM and 424 µM, respectively; (B) HCC cells were scratched and incubated with wogonin (0, 50, and 100 µM). The scar closure area was detected by measuring the distance of the wound surface healing after treatment; (C) After treating with wogonin (0, 25, and 50 µM) for 24 h, the cell invasion ability was significantly inhibited in a dose-dependent manner (* p < 0.05 versus no wogonin treatment group).
Molecules 23 00384 g001
Figure 2. Molecular docking and Surface plasmon resonance measurement of the binding between wogonin and MMP-9. (A) The two-dimensional structure of wogonin; (B) Shows the favorable binding positions of wogonin with lower binding free energy and proper distances (more than 1Å) to water molecules in the ligand-binding cavity of MMP-9. The interaction of Wogonin (in dull-red sticks) to MMP-9 with labeled amino residues responsible for generating binding free energy. Surfaces represent amino residues responsible for hydrophobic contacts with ligands. Blue lines with respective distances represent H-bonding between ligands and amino acid residue; (C) The sensorgram of wogonin binding to MMP-9-immobilized chip. The wogonin concentrations were 0.75, 1.50, 3.0, 6.0, 12.0, and 24.0 μM (from bottom to top); (D) The fitted curve for different concentrations of wogonin binding to immobilized MMP-9 using the ‘Affinity’ model in the Biacore T200 evaluation software. The dot in different colors represents different concentrations of wogonin.
Figure 2. Molecular docking and Surface plasmon resonance measurement of the binding between wogonin and MMP-9. (A) The two-dimensional structure of wogonin; (B) Shows the favorable binding positions of wogonin with lower binding free energy and proper distances (more than 1Å) to water molecules in the ligand-binding cavity of MMP-9. The interaction of Wogonin (in dull-red sticks) to MMP-9 with labeled amino residues responsible for generating binding free energy. Surfaces represent amino residues responsible for hydrophobic contacts with ligands. Blue lines with respective distances represent H-bonding between ligands and amino acid residue; (C) The sensorgram of wogonin binding to MMP-9-immobilized chip. The wogonin concentrations were 0.75, 1.50, 3.0, 6.0, 12.0, and 24.0 μM (from bottom to top); (D) The fitted curve for different concentrations of wogonin binding to immobilized MMP-9 using the ‘Affinity’ model in the Biacore T200 evaluation software. The dot in different colors represents different concentrations of wogonin.
Molecules 23 00384 g002
Figure 3. Wogonin down-regulates MMP-9 activity without affect its protein expression. (A) Shows that wogonin could inhibit intracellular active MMP-9 activities in a dose dependent manner in MHCC97L cells while no inhibit effects have been observed in pro-MMP-9 and MMP-2 activities (B) The protein expression level was determined by Western blot, MMP-9 expression was decreased in the human normal hepatocyte line, LO2. However, no significant change on the protein expression of MMP-9 has been detected in MHCC97L cells. * p < 0.05 when compare with non-treated MHCC97L group. # p < 0.05 when compare with LO2 cell control.
Figure 3. Wogonin down-regulates MMP-9 activity without affect its protein expression. (A) Shows that wogonin could inhibit intracellular active MMP-9 activities in a dose dependent manner in MHCC97L cells while no inhibit effects have been observed in pro-MMP-9 and MMP-2 activities (B) The protein expression level was determined by Western blot, MMP-9 expression was decreased in the human normal hepatocyte line, LO2. However, no significant change on the protein expression of MMP-9 has been detected in MHCC97L cells. * p < 0.05 when compare with non-treated MHCC97L group. # p < 0.05 when compare with LO2 cell control.
Molecules 23 00384 g003
Table 1. Docking score values of candidate metastasis-related targets of wogonin.
Table 1. Docking score values of candidate metastasis-related targets of wogonin.
TargetPDB-IDConsensus Score aCrash bPolar c
Human matrix metalloprotein 9 (MMP-9)5I127.63−2.321.39
Intercellular Adhesion Molecule 1 (ICAM-1)1D3E7.42−2.521.56
Stromal interaction molecule 1 (STIM1)3TER7.06−2.184.34
Fibronectin 1 (FN1)1E886.44−1.691.92
a Consensus Score: total molecular docking score to reflect the binding affinity; b Crash: degree of unsuitable penetration by the ligand into the target protein and of interpenetration among ligand atoms that are isolated by rotatable bonds; c Polar: the polar non-hydrogen bonding connections devoting to the total score. PDB-ID: Protein Data Bank Identification.
Table 2. The binding parameters of the interaction between immobilized targets and wogonin.
Table 2. The binding parameters of the interaction between immobilized targets and wogonin.
TargetsPDB-IDKD (μM)
MMP-95I1216.27
ICAM-11D3E62.46
STIM13TER87.21
FN11E88>200

Share and Cite

MDPI and ACS Style

Hong, M.; Cheng, H.; Song, L.; Wang, W.; Wang, Q.; Xu, D.; Xing, W. Wogonin Suppresses the Activity of Matrix Metalloproteinase-9 and Inhibits Migration and Invasion in Human Hepatocellular Carcinoma. Molecules 2018, 23, 384. https://doi.org/10.3390/molecules23020384

AMA Style

Hong M, Cheng H, Song L, Wang W, Wang Q, Xu D, Xing W. Wogonin Suppresses the Activity of Matrix Metalloproteinase-9 and Inhibits Migration and Invasion in Human Hepatocellular Carcinoma. Molecules. 2018; 23(2):384. https://doi.org/10.3390/molecules23020384

Chicago/Turabian Style

Hong, Ming, Honghui Cheng, Lei Song, Wencai Wang, Qi Wang, Donggang Xu, and Weiwei Xing. 2018. "Wogonin Suppresses the Activity of Matrix Metalloproteinase-9 and Inhibits Migration and Invasion in Human Hepatocellular Carcinoma" Molecules 23, no. 2: 384. https://doi.org/10.3390/molecules23020384

Article Metrics

Back to TopTop