Osthole suppresses the proliferation and accelerates the apoptosis of human glioma cells via the upregulation of microRNA-16 and downregulation of MMP-9

  • Authors:
    • Kai Lin
    • Zhiyu Gao
    • Bin Shang
    • Shaohua Sui
    • Qiang Fu
  • View Affiliations

  • Published online on: June 15, 2015     https://doi.org/10.3892/mmr.2015.3929
  • Pages: 4592-4597
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Osthole (7-methoxy-8-isoamyl alkenyl coumarin) has been reported to exhibit marked anticancer effects on several types of cancer. The expression levels of matrix metalloproteinase-9 (MMP-9) are closely associated with the pathogenesis of glioma. Furthermore, it is reported that the upregulation of microRNA‑16 (miR‑16) by the MMP‑9 signaling pathway can restrain the proliferation of cancer cells. To examine whether osthole increases the anticancer effect on human glioma cells in the present study, the common glioma cell line, U87, was treated with osthole at concentrations of 0, 50, 100 and 200 µΜ. The effects of osthole on cell viability were determined using a 3‑(4,5‑dimethylthiazol‑2‑thiazolyl)‑2,5‑diphenyl‑tetrazolium bromide assay. The rate of cellular apoptosis was analyzed by measuring the activity of caspase‑3 and using flow cytometry. The expression of MMP‑9 was determined using gelatin zymography assays and the expression of miR‑16 was determined using reverse transcription‑quantitative polymerase chain reaction. The results demonstrated that osthole significantly suppressed the proliferation and accelerated the apoptosis of the U87 cells. Furthermore, increased expression levels of miR‑16 and reduced protein expression levels of MMP‑9 were found in the U87 cells. In addition, miR‑16 was found to regulate the expression of MMP‑9 in the U87 cells through transfection of miR‑16 precursor and anti‑miR‑16 into the U87 cells. In conclusion, these observations indicated that osthole suppressed the proliferation and accelerated the apoptosis of human glioma cells through upregulation of the expression of miR‑16 and downregulation of the expression of MMP-9.

Introduction

It is well known that glioma accounts for 44.6% of tumors of the central nervous system and has the characteristics of a high recurrence rate and high mortality rate (1). Although certain surgical comprehensive treatments can significantly improve the survival rates of patients with glioma, the prognosis remains poor due to the occurrence of chemotherapy drug resistance in glioma therapy (2).

Matrix metalloproteinases (MMPs) are reported to interact with the cellular adhesive molecular degradation of the extracellular matrix, promoting the growth of tumor cells into the surrounding brain tissue (3). Cell proliferation may be affected by regulating MMP-9 activity, and MMP-9 activity is controlled by several factors, including the quantity and activation of enzymes (4). MMP-9 is correlated with glioma and it has been demonstrated that the expression levels of MMP-9 directly reflect the prognosis of patients with glioma and is the preferred predictor of invasive glioma cell growth (5,6).

MicroRNAs (miRNAs) are a series of non-coding, small molecule RNAs, which regulates gene expression via sequence complementation. These small RNAs consist of between 19 and 25 nucleotides (7). Following transcription, they exert inhibitory effects on gene expression and are involved in several physiological processes, including cell differentiation, apoptosis and metabolism (8). Previous studies have demonstrated that miR-16 is important in tumors of various origins (911). In glioma growth and invasiveness, the importance of miR-16 as a tumor suppressor gene and a novel mechanism of miR-16 regulation through inhibition the nuclear factor (NF)-κB1/MMP-9 signaling pathway has been reported (11).

Osthole is a natural coumarin isolated from umbelliferae plant monomers (ripe fruit). A previous modern pharmacological investigation demonstrated that osthole has anti-inflammatory, antioxidant and other pharmacological properties (12,13). In addition, it was also revealed that osthole markedly decreases the activity and protein content of MMP-9, suggesting that this protective effect at the molecular level may be due to downregulation of the MMP-9 pathway (14). The aim of the present study was to investigate the anticancer potential of osthole against glioma cells and examine whether this mechanism is dependent on the upregulation of miR-16 and downregulation of MMP-9 expression.

Materials and methods

Reagents and chemicals

The chemical structure of osthole (Sigma-Aldrich, St. Louis, MO, USA; purity, ≥95%) is shown in Fig. 1. Osthole was dissolved in physiological saline (50–200 µM). Roswell Park Memorial Institute-1640 (RPMI-1640) medium, fetal calf serum (FCS) and Lipofectamine 2000 were purchased from Invitrogen Life Technologies (Carlsbad CA, USA). 3-(4,5-dimethylthiazol-2-thiazolyl)-2,5-diphenyl-tetrazolium bromide (MTT) was purchased from Beyotime Institute of Biotechnology, (Haimen, China). The Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) Double Staining kit was purchased from BestBio (Shanghai, China).

Cancer cell lines

The U87 glioma cell line was purchased from the Animal Experiments of Clinical School of Taishan Medical University (Taian, China). The U87 cells were cultured in RPMI-1640 medium, supplemented with 10% FCS, 100 U/ml penicillin and 100 mg/ml streptomycin, at 37°C and 5% CO2.

MTT viability assay

The effect of osthole on the proliferation of U87 cells was measured using an MTT assay. The U87 cells (5.0×103 cells/well) were seeded into 96-well culture plates at 95% confluence and incubated at 37°C and 5% CO2 in a humidified incubator for 24 h. Following incubation, the cells were treated with different concentrations of osthole (0, 50, 100 or 200 µΜ) for 0, 24, 48 or 72 h. MTT (~10 µl of 10 mg/ml) was added into each well and incubated at 37°C and 5% CO2 for 4 h. Subsequently, 150 µl dimethyl sulfoxide was added to each well and incubated for 20 min at room temperature with agitation. The absorbance of the plates was detected using an CM2600d spectrometer (Bio-Tek Instruments, Inc., Winooski, VT, USA) at 570 nm.

Measurement of caspase-3 activity

The activity of caspase-3 in the cells was measured using a Colorimetric Caspase-3 Assay kit (Beyotime Institute of Biotechnology). Following treatment with 100 µM osthole for 48 h, the U87 cells (1.0–2.0×106 cells/well) were centrifuged at 12,300 × g for 20 min at 4°C. The supernatant was collected and the protein concentration was quantified using a bicinchoninic acid (BCA) protein assay kit (Sangon Biotech, Shanghai, China). The protein extract (~50 µg) was incubated and added to a reaction buffer containing 90 µl 1X assay buffer and 10 µl Ac-DEVD-pNA caspase-3 substrate at 37°C for 6 h. The protein extract (~50 µg) was incubated at 37°C and added to a reaction buffer containing 90 µl 1X assay buffer (Beyotime Institute of Biotechnology) and 10 µl Ac-DEVD-pNA caspase-3 substrate (Beyotime Institute of Biotechnology) at 37°C for 6 h. The change was calculated using a CM2600d spectrometer at a wavelength of 405 nm (Bio-Tek Instruments, Inc.).

Annexin V/PI flow cytometric analysis

The apoptotic rates of the U87 cells were determined using flow cytometric analysis (BD Biosciences, Franklin Lakes, NJ, USA) using an Annexin V-FITC/PI apoptosis kit. Following treat ment with 100 µM osthole for 48 h, the U87 cells were collected and washed twice with phosphate-buffered saline. Annexin V-FITC (10 µl) was added to the U87 cells, following which the cells were stained with binding buffer (BestBio) for 30 min in the dark, according to the manufacturer's instructions. PI (10 µl) was added to the glioma cell samples, which were then incubated for 30 min at room temperature in the dark. Immediately following incubation, the samples were analyzed using flow cytometry.

MMP-9 measurement

To determine whether 100 µM osthole induced the expression of MMP-9 in the U87 cells, gelatin zymography assays were used. Following treatment with osthole for 48 h at 37°C, the U87 cells were harvested and the concentration of protein was determined using a BCA protein assay kit (Sangon Biotech). Equal quantities of protein were extracted and were subsequently electrophoresed on 10% SDS-PAGE gels (Beyotime Institute of Biotechnology), containing 1% gelatin. Following electrophoresis, the gel was rinsed in 2% Triton X-100 (Nanjing Senbeijia Biotech Company, Nanjing, China) for 1 h and subsequently washed in water. The gels were incubated in radioimmunoprecipitation assay buffer (pH 8.0; Sigma-Aldrich) at 37°C for 12 h and the gel was stained with 0.2% Coomassie Blue R-250 (Beyotime Institute of Biotechnology) for 1 h. The protein expression levels of MMP-9 were quantified using a MiniBis system (DNR Bio-Imaging Systems Ltd., Jerusalem, Israel) and prestained SDS-PAGE standards (Houbio Tech Co., Ltd., Fan Ling, Hong Kong).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) of the expression of miR-16

The present study investigated whether 100 µM osthole induced the expression of miR-16 in the U87 cells using RT-qPCR. Following treatment with osthole for 48 h at 37°C, the total RNA was extracted from the cells using TRIzol reagent (Invitrogen Life Technologies), according to manufacturer's instructions. Approximately 1 µg total RNA from U87 cells-treated was used to perform the first-strand cDNA synthesis. Subsequently, 2 ml RNA was transcribed to cDNA using random hexamers (Promega, Madison, WI, USA) according to the manufacturer's instructions. The cycling conditions were as follows: 10 min at 95°C, 40 cycles of 45 sec at 95°C, 45 sec at 58°C and 45 sec at 72°C. The quantification of the miR-16 level was conducted by real- time PCR using TransStart™ SYBR Green qPCR Supermix (TransGen Biotech, Beijing, China) and U6 small nuclear RNA was regarded as an endogenous reference gene. The U6 primer sequence was as follows: Forward 5′-CTCGCTTCGGCAGCACA-3′ and reverse 5′-AACGCTTCACGAATTTGCGT-3′. The miR-16 primer sequence was as follows: Forward 5′-TTCCATGCTGTTTTGGTCCC-3′ and reverse 5′-TGGGTGGAGGTTTGTTCGGA-3′. Primers were provided by Sangon Biotech Co., Ltd. (Shanghai, China). Relative quantification was carried out using the 2−ΔΔCt cycle threshold method.

miR-16 and anti-miR-16 transfection

The miR-16 precursor and the anti-miR-16 were obtained from KeyGen Biotech Co., Ltd. (Nanjing, China). The U87 cells (5×105 cells/well) were cultured in six-well plates and transfected with either miR-16 precursor or anti-miR-16 using Lipofectamine 2000 for 6 h at 37°C. The transfection media was replaced with RPMI-1640 containing 10% FCS without antibiotic in a humidified atmosphere at 37°C with 5% CO2 for 18 h.

Statistical analysis

All data are presented as the mean ± standard deviation. The data were analyzed using Student's t-test. Statistical analysis was performed using SPSS 17.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Osthole suppresses proliferation and increases caspase-3 activity in the U87 cells

To determine whether there is an association between osthole and U87 cells, the viability of U87 cells following treatment with different concentrations of osthole (0, 50, 100 or 200 µΜ) was determined using an MTT assay. As shown in Fig. 2A, the analysis revealed that treatment with 50 µΜ osthole for 72 h significantly reduced U87 cell viability, and treatment with 100 and 200 µΜ for 48 h or 72 h significantly reduced U87 cell viability. In addition, the decrease in U87 cell viability was found to occur in a time- and concentration-dependent manner in the osthole-treated cells. The activity of caspase-3 in the U87 cells was analyzed following treatment with osthole (0, 50, 100 or 200 µΜ) using a caspase-3 assay. Following treatment with 100 or 200 µΜ osthole for 48 h, the activity of caspase-3 was significantly increased in the U87 cells (P<0.05), and this increase in the activity of caspase-3 occurred in a concentration-dependent manner in the osthole-treated cells (Fig. 2B).

Flow cytometric analysis for the detection of cellular apoptosis

The U87 cells were exposed to osthole at concentrations of 0, 50, 100 or 200 µΜ for 48 h, and the effect of osthole on the augmentation of apoptosis was investigated. The results of this analysis using flow cytometry demonstrated that osthole inhibited the growth of the U87 cells in a dose-dependent manner (Fig. 3). Treatment with 100 and 200 µΜ osthole for 48 h significantly increased the apoptosis of U87 cells.

Inhibition of MMP-9 by osthole

To examine a potential association between the effect of osthole (0, 50, 100 of 200 µΜ) on the U87 cells and MMP-9, the protein expression levels of MMP-9 were determined using gelatin zymography assays. The data indicated that osthole inhibited the protein expression levels of MMP-9 in a dose-dependent manner (Fig. 4A). As shown in Fig. 4B, treatment with 100 and 200 µΜ of osthole for 48 h significantly reduced the protein expression levels of MMP-9 in the U87 cells.

Osthole activates the expression of miR-16

The present study aimed to determine the possible correlations between osthole (0, 50, 100 or 200 µΜ) and the expression levels of miR-16, which was assessed using RT-qPCR. The data indicated that osthole promoted the expression levels of miR-16 in a dose-dependent manner. As shown in Fig. 5, treatment with 100 and 200 µΜ osthole for 48 h significantly promoted the expression of miR-16 in the U87 cells (P<0.05).

Overexpression of miR-16 inhibits the protein expression of MMP-9

The protein expression levels of MMP-9 were examined following transfection of the U87 cells with miR-16. The results demonstrated that the protein expression levels of MMP-9 were reduced by overexpression of miR-16 in the U87 cells treated with 100 µΜ osthole. As shown in Fig. 6A and B, the miR-16 overexpressing cells inhibited the protein expression of MMP-9.

Anti-miR-16 reverses the effect of osthole

To confirm the functional role of osthole and of miR-16 in mediating the effects on the viability of the U87 cells, the present study examined the activity of caspase-3 and the protein expression levels of MMP-9 following transfection of the cells with anti-miR-16. The results indicated that transfection with the anti-miR-16 antibody efficiently penetrated into the U87 cells and significantly reduced the expression of miR-16 in the U87 cells (Fig. 7A). Following treatment with 100 µΜ osthole for 48 h, transfection with the anti-miR-16 antibody neutralized the effect of miR-16 on the U87 cells (Fig. 7B), inhibited the apoptotic effect on the U87 cells, indicated by downregulation in capsase-3 activity (Fig. 7C), and neutralized the inhibitory effect of osthole through downregulating the expression of MMP-9 (Fig. 7D).

Discussion

Glioma comprises 44.6% of tumors of the central nervous system and exhibit characteristics of a high recurrence rate and high mortality rate. With the development of molecular biology techniques, investigating glioma resistance mecha nism has been achieved at the molecular gene level and, through increased investigation, a deeper understanding of the resistance mechanism of glioma has been demonstrated (15). Several studies have reported certain natural chemical and compound products, which exert anticancer effects on a wide variety of tumor types (16,17). A previous study reported that osthole induces cell apoptosis and weakens cell migration in brain tumors (18). However, the detailed mechanisms underlying the anticancer effects of osthole on glioma remain to be fully elucidated. The present study demonstrated that osthole suppressed the proliferation and increased the activity of caspase-3 in the U87 cells. These results were consistent with those of previous studies (1921). For example, Ding et al (19) demonstrated that osthole inhibits the proliferation, migration and invasion, and induces the apoptosis of glioma cells.

Osthole, isolated from the rumbrelliferae plant monomers of the ripe fruit, Cnidium monnieri, possesses anti-inflammatory properties, improves learning and memory, and inhibits thrombosis (2224). In the present study, treatment of the U87 cells with osthole significantly increased the apoptosis of the cells. Osthole was previously observed to suppress cell proliferation and induce cellular apoptosis in NCI-H460 lung carcinoma cells, which corresponded with the present study (25).

The degradation of the extracellular matrix by MMPs via type II collagenase AB is associated with the invasive cell growth of glioma, with MMP-9 being the most closely associated with glioma and, to a certain extent, its expression can reflect the extent of glioma invasion within the body (26,27). The mRNA expression level of MMP-9 is closely associated with the degree of malignant glioma, and it has been reported that the intensity of MMP-9-positive staining is closely associated with the pathological levels of glioma (28). These reports indicate that MMP-9 can be used to determine the human glioma malignant phenotype (29). Using a nude mouse model to investigate the growth of glioma, it was determined that increased expression levels of MMP-9 are more marked, compared with the expression level of MMP-2, and indicates that MMP-9 is important in the growth of glioma (30). Consistently, the results of the present study demonstrated that treatment with osthole significantly reduced the protein expression levels of MMP-9. It was previously revealed that osthole suppresses the levels of MMP-2 and MMP-9 in A549 human lung cancer cells (31). In addition, osthole has been observed to inhibit NF-κB-MMP-9 in human lung adenocarcinoma (14).

In the present study, RT-qPCR was used to identify possible correlations between osthole (0, 50, 100 or 200 µΜ) and the expression of miR-16. The data indicated that osthole promoted the expression of miR-16 in a dose-dependent manner. The expression levels of miR-16-1 were markedly reduced in the human glioma cell lines, which suggested that miR-16-1 is involved in the proliferative, migratory and invasive abilities of highly-invasive glioma cells (32). The present study demonstrated that upregulation of the expression of miR-16 reduced the protein expression levels of MMP-9 in the (U87 cells. In addition, the results revealed that downregulation of the expression of miR-16, via anti-miR-16 transfection, promoted the protein expression of MMP-9 in the U87 cells and reduced the effect of osthole on the proliferation and apop-tosis of the U87 cells. In conclusion, the results of the present study suggested that osthole suppressed the proliferation and accelerated the apoptosis of U87 human glioma cells via upregulation of the expression of miR-16 and downregulation of the expression of MMP-9.

Acknowledgments

This study was supported by the Twelve-Five National Science and Technology Support Program (grant no. 2011BAI08B04).

References

1 

Sun YC, Wang J, Guo CC, Sai K, Wang J, Chen FR, Yang QY, Chen YS, Wang J, To TS, et al: MiR-181b sensitizes glioma cells to teniposide by targeting MDM2. BMC Cancer. 14:6112014. View Article : Google Scholar : PubMed/NCBI

2 

Cunha LC, Del Bel E, Pardo L, Stühmer W and Titze-DE-Almeida R: RNA interference with EAG1 enhances interferon gamma injury to glioma cells in vitro. Anticancer Res. 33:865–870. 2013.PubMed/NCBI

3 

Yang X, Lv S, Liu Y, Li D, Shi R, Tang Z, Fan J and Xu Z: The clinical utility of matrix metalloproteinase 9 in evaluating pathological grade and prognosis of glioma patients: a meta-analysis. Mol Neurobiol. Aug 10–2014.

4 

Sun C, Wang Q, Zhou H, Yu S, Simard AR, Kang C, Li Y, Kong Y, An T, Wen Y, et al: Antisense MMP-9 RNA inhibits malignant glioma cell growth in vitro and in vivo. Neurosci Bull. 29:83–93. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Asuthkar S, Velpula KK, Chetty C, Gorantla B and Rao JS: Epigenetic regulation of miRNA-211 by MMP-9 governs glioma cell apoptosis, chemosensitivity and radiosensitivity. Oncotarget. 3:1439–1454. 2012.PubMed/NCBI

6 

Xia H, Qi Y, Ng SS, Chen X, Li D, Chen S, Ge R, Jiang S, Li G, Chen Y, et al: microRNA-146b inhibits glioma cell migration and invasion by targeting MMPs. Brain Res. 1269:158–165. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Cui Y, Bai Y, Wang XD, Liu B, Zhao Z and Wang LS: Differential expression of miRNA in rat myocardial tissues under psychological and physical stress. Exp Ther Med. 7:901–906. 2014.PubMed/NCBI

8 

Yan X, Liang H, Deng T, et al: The identification of novel targets of miR-16 and characterization of their biological functions in cancer cells. Mol Cancer. 12:922013. View Article : Google Scholar : PubMed/NCBI

9 

Zhu Y, Xia Y, Niu H and Chen Y: MiR-16 induced the suppression of cell apoptosis while promote proliferation in esophageal squamous cell carcinoma. Cell Physiol Biochem. 33:1340–1348. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Aqeilan RI, Calin GA and Croce CM: miR-15a and miR-16-1 in cancer: discovery, function and future perspectives. Cell Death Differ. 17:215–220. 2010. View Article : Google Scholar

11 

Yang TQ, Lu XJ, Wu TF, Ding DD, Zhao ZH, Chen GL, Xie XS, Li B, Wei YX, Guo LC, et al: MicroRNA-16 inhibits glioma cell growth and invasion through suppression of BCL2 and the nuclear factor-kappaB1/MMP9 signaling pathway. Cancer Sci. 105:265–271. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Wang XY, Dong WP, Bi SH, Pan ZG, Yu H, Wang XW, Ma T, Wang J and Zhang WD: Protective effects of osthole against myocardial ischemia/reperfusion injury in rats. Int J Mol Med. 32:365–372. 2013.PubMed/NCBI

13 

Sun F, Xie ML, Zhu LJ, Xue J and Gu ZL: Inhibitory effect of osthole on alcohol-induced fatty liver in mice. Dig Liver Dis. 41:127–133. 2009. View Article : Google Scholar

14 

Kao SJ, Su JL, Chen CK, Yu MC, Bai KJ, Chang JH, Bien MY, Yang SF and Chien MH: Osthole inhibits the invasive ability of human lung adenocarcinoma cells via suppression of NF-kappaB-mediated matrix metalloproteinase-9 expression. Toxicol Appl Pharmacol. 261:105–115. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Yang X, Lv S, Zhou X, Liu Y, Li D, Shi R, Kang H, Zhang J and Xu Z: The clinical implications of transforming growth factor beta in pathological grade and prognosis of glioma patients: A meta-analysis. Mol Neurobiol. Aug 23–2014.

16 

Lu DY, Chang CS, Yeh WL, Tang CH, Cheung CW, Leung YM, Liu JF and Wong KL: The novel phloroglucinol derivative BFP induces apoptosis of glioma cancer through reactive oxygen species and endoplasmic reticulum stress pathways. Phytomedicine. 19:1093–1100. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Tsai CF, Yeh WL, Huang SM, Tan TW and Lu DY: Wogonin induces reactive oxygen species production and cell apoptosis in human glioma cancer cells. Int J Mol Sci. 13:9877–9892. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Tsai CF, Yeh WL, Chen JH, Lin C, Huang SS and Lu DY: Osthole suppresses the migratory ability of human glioblastoma multiforme cells via inhibition of focal adhesion kinase-mediated matrix metalloproteinase-13 expression. Int J Mol Sci. 15:3889–3903. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Ding D, Wei S, Song Y, Li L, Du G, Zhan H and Cao Y: Osthole exhibits anti-cancer property in rat glioma cells through inhibiting PI3K/Akt and MAPK signaling pathways. Cell Physiol Biochem. 32:1751–1760. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Wang L, Peng Y, Shi K, et al: Osthole inhibits proliferation of human breast cancer cells by inducing cell cycle arrest and apoptosis. J Biomed Res. 29:132–138. 2015.PubMed/NCBI

21 

Lin YC, Lin JC, Hung CM, et al: Osthole inhibits insulin-like growth factor-1-induced epithelial to mesenchymal transition via the inhibition of PI3K/Akt signaling pathway in human brain cancer cells. J Agric Food Chem. 62:5061–5071. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Liu J, Zhang W, Zhou L, Wang X and Lian Q: Anti-inflammatory effect and mechanism of osthole in rats. Zhong Yao Cai. 28:1002–1006. 2005.

23 

Zhang Q, Qin L, He W, Van Puyvelde L, Maes D, Adams A, Zheng H and De Kimpe N: Coumarins from Cnidium monnieri and their antiosteoporotic activity. Planta Med. 73:13–19. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Luszczki JJ, Andres-Mach M, Cisowski W, Mazol I, Glowniak K and Czuczwar SJ: Osthole suppresses seizures in the mouse maximal electroshock seizure model. Eur J Pharmacol. 607:107–109. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Xu XM, Zhang Y, Qu D, Liu HB, Gu X, Jiao GY and Zhao L: Combined anticancer activity of osthole and cisplatin in NCI-H460 lung cancer cells in vitro. Exp Ther Med. 5:707–710. 2013.PubMed/NCBI

26 

Singh MK, Bhattacharya D and Chaudhuri S, Acharya S, Kumar P, Santra P, Basu AK and Chaudhuri S: T11TS inhibits glioma angio-genesis by modulation of MMPs, TIMPs, with related integrin alphav and TGF-β1 expressions. Tumour Biol. 35:2231–2246. 2014. View Article : Google Scholar

27 

Kim MS, Kwak HJ, Lee JW, Kim HJ, Park MJ, Park JB, Choi KH, Yoo H, Shin SH, Shin WS, et al: 17-Allylamino-17-demethoxygeldanamycin down-regulates hyaluronic acid-induced glioma invasion by blocking matrix metalloproteinase-9 secretion. Mol Cancer Res. 6:1657–1665. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Huang HC, Huang CY, Lin-Shiau SY and Lin JK: Ursolic acid inhibits IL-1beta or TNF-alpha-induced C6 glioma invasion through suppressing the association ZIP/p62 with PKC-zeta and downregulating the MMP-9 expression. Mol Carcinog. 48:517–531. 2009. View Article : Google Scholar

29 

Gondi CS, Lakka SS, Dinh DH, Olivero WC, Gujrati M and Rao JS: Downregulation of uPA, uPAR and MMP-9 using small, interfering, hairpin RNA (siRNA) inhibits glioma cell invasion, angiogenesis and tumor growth. Neuron Glia Biol. 1:165–176. 2004. View Article : Google Scholar

30 

Pagliara V, Adornetto A, Mammi M, Masullo M, Sarnataro D, Pietropaolo C and Arcone R: Protease Nexin-1 affects the migration and invasion of C6 glioma cells through the regulation of urokinase Plasminogen Activator and Matrix Metalloproteinase-9/2. Biochim Biophys Acta. 1843:2631–2644. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Xu XM, Zhang Y, Qu D, Feng XW, Chen Y and Zhao L: Osthole suppresses migration and invasion of A549 human lung cancer cells through inhibition of matrix metalloproteinase-2 and matrix metallopeptidase-9 in vitro. Mol Med Rep. 6:1018–1022. 2012.PubMed/NCBI

32 

Li X, Ling N, Bai Y, Dong W, Hui GZ, Liu D, Zhao J and Hu J: MiR-16-1 plays a role in reducing migration and invasion of glioma cells. Anat Rec (Hoboken). 296:427–432. 2013. View Article : Google Scholar

Related Articles

Journal Cover

September-2015
Volume 12 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin K, Gao Z, Shang B, Sui S and Fu Q: Osthole suppresses the proliferation and accelerates the apoptosis of human glioma cells via the upregulation of microRNA-16 and downregulation of MMP-9. Mol Med Rep 12: 4592-4597, 2015
APA
Lin, K., Gao, Z., Shang, B., Sui, S., & Fu, Q. (2015). Osthole suppresses the proliferation and accelerates the apoptosis of human glioma cells via the upregulation of microRNA-16 and downregulation of MMP-9. Molecular Medicine Reports, 12, 4592-4597. https://doi.org/10.3892/mmr.2015.3929
MLA
Lin, K., Gao, Z., Shang, B., Sui, S., Fu, Q."Osthole suppresses the proliferation and accelerates the apoptosis of human glioma cells via the upregulation of microRNA-16 and downregulation of MMP-9". Molecular Medicine Reports 12.3 (2015): 4592-4597.
Chicago
Lin, K., Gao, Z., Shang, B., Sui, S., Fu, Q."Osthole suppresses the proliferation and accelerates the apoptosis of human glioma cells via the upregulation of microRNA-16 and downregulation of MMP-9". Molecular Medicine Reports 12, no. 3 (2015): 4592-4597. https://doi.org/10.3892/mmr.2015.3929