Hexane extract from Sargassum serratifolium inhibits the cell proliferation and metastatic ability of human glioblastoma U87MG cells

  • Authors:
    • Chang-Won Kang
    • Min-Seok Park
    • Nan-Hee Kim
    • Ji-Hyun Lee
    • Chul-Woong Oh
    • Hyeung-Rak Kim
    • Gun-Do Kim
  • View Affiliations

  • Published online on: August 21, 2015     https://doi.org/10.3892/or.2015.4222
  • Pages: 2602-2608
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study is the first to demonstrate the anticancer effects of a hexane extract from the brown algae Sargassum serratifolium (HES) on human cancer cell lines, including glioblastoma U87MG, cervical cancer HeLa and gastric cancer MKN-28 cells, as well as liver cancer SK-HEP 1 cells. Among these cancer cell lines, U87MG cells were most sensitive to the cell death induced by HES. HES exhibited a cytotoxic effect on U87MG cells at concentrations of 14-16 µg/ml, yet an effect was not observed in human embryonic kidney HEK293 cells. The antiproliferative effects of HES were regulated by inhibition of the MAPK/ERK signaling pathway which plays a pivotal role in the proliferation of glioblastoma U87MG cells. In addition, treatment with HES led to cell morphological changes and cell cytoskeleton degradation through regulation of actin dynamic signaling. Furthermore, migration and invasion of the U87MG cells were inhibited by HES via suppression of matrix metalloproteinase (MMP)-2 and -9 expression. Thus, our results suggest that HES is a potential therapeutic agent which has anticancer effects on glioblastoma.

Introduction

Human glioblastoma is one of the most common and malignant tumors, that originates from glial cells (1). Glioblastoma is characterized by rapid invasion and distant migration (2). Current therapeutic strategies for glioblastoma include surgery, radiation therapy and chemotherapy. However, the median survival of glioblastoma patients is extremely poor (3).

Mitogen-activated protein kinase (MAPK) signaling regulates diverse cellular functions, such as cell proliferation, the cell cycle, cell survival, angiogenesis and cell migration (4). The MAPK cascade is initiated by Ras activation which is regulated by receptor tyrosine kinases at the cell surface. Ras activation recruits C-Raf to the cell membrane, processing the phosphorylation of multiple co-factors (5). Downstream of the C-Raf proteins are MEK1 and MEK2, a dual serine/threo-nine and tyrosine kinase (6). MEK further transmits signals to extracellular regulated serine/threonine kinases (ERK), comprised of ERK1 and ERK2 (7). The cellular functions of ERK play diverse roles in the regulation of cell proliferation, survival, mitosis and migration (8).

The cytoskeleton plays pivotal roles in various signaling pathways related to cellular adhesion, cytokinesis, cell survival and transcriptional processes (9). Particularly, reorganization of the actin cytoskeleton is essential for the migration and metastasis of malignant tumors. Actin reorganization is regulated by Rac, the Wiskott-Aldrich syndrome protein (WASP) family and actin-related protein (Arp) (10). Rac is a Ras-related GTP binding protein that controls assembly and disassembly of actin cytoskeleton by transducing extracellular chemoattractive signals to downstream effectors such as WASP family verprolin-homologous protein-2 (WAVE-2) and Arp-2. WAVE-2 activates the Arp2/3 complex through their binding and consequently the Arp2/3 complex locates to actin filament branches and crosslinks into a branching network (11). In addition, they play a functional role in lamelli-podium formation that is related to the invasion and metastasis of malignant tumors (12,13).

Metastasis is a mechanism by which new tumors form in distant tissues from a primary tumor. Cell migration and invasion are required for cancer metastasis. Invasion is induced by matrix metalloproteinases (MMPs) that induce degradation of the extracellular matrix (ECM) and basement membrane (14,15). Thus, inhibition of MMPs is considered to be a therapeutic strategy for tumor metastasis.

Sargassum (Sargassaceae, Fucales) is a genus of brown seaweed that is found in all the oceans. Previous research on Sargassum spp. extracts have been reported to exhibit anticancer, antibacterial, antifungal, antiviral, anti-inflammatory, anticoagulant, antioxidant, hepatoprotective and neuroprotective activities (16). However, the pharmacological effect of extracts from Sargassum serratifolium (S. serratifolium) has not been thoroughly studied in glioblastoma. Therefore, the present study aimed to investigate the anticancer effects of an extract from S. serratifolium on U87MG glioblastoma cells.

Materials and methods

Preparation of the hexane extracts

S. serratifolium was collected from the coast of Busan, Korea, in April 2013. Taxonomic identification was confirmed by an algal taxonomist (C.K. Choi) at the Department of Ecological Engineering, Pukyong National University, Korea. The seaweed was rinsed in tap water to remove salt and air-dried under sunlight for 3 days. The dried powder (1.5 kg) of S. serratifolium underwent extraction three times with 96% (v/v) ethanol (EtOH) for 3 h at 70°C. The combined extracts were concentrated under reduced pressure to obtain the EtOH extract. For further fractionation of the EtOH extract, the extract was resuspended in water:EtOH (9:1) and partitioned successively with n-hexane and ethyl acetate. The n-hexane extract, which showed the highest cell cytotoxic activities against U87MG cells, was maintained at −20°C and was used for the present study.

Cell culture

Human glioblastoma cancer U87MG, human cervical cancer HeLa, human gastric cancer MKN-28 and human liver cancer SK-HEP 1 cells as well as human embryonic kidney HEK-293 cells, were obtained from the American Tissue Culture Collection (ATCC; Manassas, VA, USA). U87MG and SK-HEP 1 cells were incubated with minimum essential medium (MEM), MKN-28 cells were incubated with RPMI-1640 medium, and HeLa and HEK-293 cells were incubated in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 1% penicillin-streptomycin (PAA Laboratories GmbH, Pasching, Austria). The cells were cultured in a 5% CO2 incubator (binder) at 37°C in a humidified atmosphere. The culture was sub-cultured every 3 to 4 days and routinely checked under an inverted microscope for any contamination.

Cell cytotoxicity and morphology

The effects of the extracts of Sargassum serratifolium on cell viability were evaluated by WST-1 assay, based on the reduction of the number of metabolically active cells, and the results are expressed as a percentage of the control. Cells were seeded in 96-well micro-plates at a density of 1×104 cells/well and were cultured for 24 h. After 24 h, the cells were treated with the extracts at various concentrations and incubated for 12 and 24 h. Then, the media was replaced and 2-(4-iodophenyl)-3-(4-nitrophenyl)-5-(2,4-disulforphenyl)-2H-tetrazolium, monosodium salt (WST-1 solution) was added to each well at 10 µl, and the cells were incubated at 37°C for 3 h. Finally the optical density (OD) was measured with an ELISA reader (Molecular Devices, Silicon Valley, CA, USA) at 460 nm. The cytotoxic activity of the extract was expressed as an IC50 value, which is the concentration of the extract that caused 50% cell death. The extract with an IC50 value ≤16 µg/ml was considered active. Dimethylsulfoxide (DMSO) was used to dilute the extracts and the final concentration of DMSO in each well was not in excess of 0.5% (v/v). No adverse effect due to the presence of DMSO was observed.

Protein extraction and western blot analysis

U87MG cells were treated with Sargassum serratifolium. The treated cells were washed with ice-cold 1X phosphate-buffered saline (PBS) and collected in lysis buffer [(50 mM Tris-Cl (pH 7.5), 150 mM NaCl, 1 mM DTT, 0.5% NP-40, 1% Triton X-100, 1% deoxycholate, 0.1% SDS] and a proteinase inhibitor cocktail [PMSF, EDTA, aptotinin, leupeptin, prostatin A (Intron Biotechnology, Gyeonggi, Korea)] on ice. After incubation on ice for 30 min, the insoluble materials were removed by centrifugation at 14,000 rpm for 20 min. Protein contents of the cell lysates were determined by a protein quantification kit (Commasie Brilliant Blue solution®) (Dojindo Molecular Technologies, Rockville, MD, USA) with bovine serum albumin (BSA) as a standard. The absorbance was determined at 595 nm. An aliquot from each sample was boiled for 5 min and then resolved by 12% SDS-polyacrylamide gel electrophoresis (SDS-PAGE). Then, the proteins were electrotransferred to a nitrocellulose membrane (Pall Life Sciences, Pensacola, FL, USA) and blocked in PBST buffer (135 µM NaCl, 2.7 mM KCl, 4.3 mM NaPO4, 1.4 mM KH2PO4 and 0.5% Tween-20) containing 5% non-fat milk (w/v). After blocking, the membranes were then incubated with primary antibodies against GAPDH, integrin β1, Rac1/2/3, Ras, PI3K p110α, WAVE-2, Arp-2, MMP-2, MMP-9, phospho-C-RafSer338, phospho-MEK 1/2Ser217/221, phospho-ERK 1/2Thr202/Tyr204 overnight at 4°C. The membranes were next incubated with HRP-conjugated secondary antibodies (Cell Signaling Technology) for 60 min. All membranes were visualized using West Save Gold ECL (AbFrontier) and exposed to Hyperfilm (GE Healthcare). GAPDH was used as a loading control.

Wound healing assay

U87MG cells (5.0×105 cells/well) were seeded in the chamber of an Ibidi culture insert (Ibidi GmbH, Martinsried, Germany) consisting of two reservoirs separated by a 500 µm well and incubated at 37°C in an atmosphere of 5% CO2 for 24 h. After incubation, the inserts were gently removed, and the cells were cultured with medium to facilitate cell migration. Cell migration was recorded by phase contrast microscopy over a time course of 2h following treatment with the S. serratifolium extract. On-line based Wimasis image analysis was used to carry out quantitative analysis of the cell migration.

Cell invasion assay

The invasion of the tumor cells was assessed in Matrigel-coated Τranswell chambers with a 6.5-mm polyvinyl/pyrolidone-free polycarbonate filter with 8-µm pore size (Corning Life Sciences) as previously described (17). The U87MG cells (5×104 cells/well and test compounds at different concentrations were suspended in 100 µl of serum-free medium and placed in the upper Τranswell chamber and incubation was carried out for 24 h at 37°C. Then, the cells on the upper surface of the filter were completely wiped away with a cotton swab, and the cells on the lower surface were lysed with 4% formaldehyde and stained with crystal violet. After staining, cells on the lower surface were lysed with 2% SDS for 1 h and the lysate was measured using a microplate reader at 570 nm.

Immunofluorescence

Cultured U87MG cells on a cover glass-bottom dish were incubated for 30 min with 10 µg/ml HES. For this, the cells were pre-treated with 1 µg/ml DAPI for 20 min at 37°C, then fixed with 4% formaldehyde (Junsei Chemical Co., Ltd., Japan) for 15 min at 25°C and blocked for 1 h in a blocking solution, including 5% mouse and rabbit normal serum (Santa Cruz Biotechnology, Inc.) with 0.3% Triton X-100 (Sigma-Aldrich, St. Louis, Mo, USA). Fixed and blocked cells were incubated with the primary antibodies (β-actin, phospho-ERK1/2Thr202/Tyr204) for 3 h and washed three times with PBS buffer. After washing, the cells were treated with 0.1 µg/ml of anti-mouse IgG (H+L), F(ab′)2 fragment (Alexa Fluor® 555-conjugated) and anti-rabbit IgG (H+L), F(ab′)2 fragment (Alexa Fluor® 488-conjugated) for 1 h. Stained cells were mounted on a slide with ProLong Gold antifade reagent (Invitrogen, Grand Island, NY, USA) and fluorescence was determined under a Carl Zeiss LSM 710 confocal laser scanning microscope.

Statistical analysis

Data are presented as the mean ± standard deviation for the indicated number of separate experiments. The mean of the control was compared with the mean of each individual treatment group by one-way ANOVA followed by Tukey's test using the statistical software SigmaPlot v.12.3 s(Systat Software, Inc., San Jose, CA, USA), and a statistically significant difference was set at p<0.001.

Results

Effects of S. serratifolium extracts on cell proliferation

To investigate the antiproliferative effects of three types of extracts, hexane, ethanol and ethyl acetate, from S. serratifolium, we performed WST-1 assay on human glioblastoma cancer U87MG, human cervical cancer HeLa, human gastric cancer MKN-28, human liver cancer SK-HEP 1 and noncancerous HEK293 cells following treatments with the extracts. As shown in Fig. 1, each extract had a cytotoxic effect on the human cancer cell lines. Among these extracts, the hexane extract from S. serratifolium (HES) exhibited the most effective inhibition of cell proliferation. HES particularly decreased the cell viability of the U87MG cells to a higher degree than that observed in the other cancer cell lines. The IC50 value of HES was observed between 14 and 16 µg/ml HES (Fig. 2). In addition, morphological cell changes were observed in a dose-dependent manner (Fig. 4A). However, there was no inhibitory effect on cell proliferation of the noncancerous HEK293 cells, indicating that HES particularly has antiproliferation effects on U87MG cells (Fig. 2B).

Antiproliferative effect of HES is mediated through the MAPK/ERK pathway

The MAPK/ERK cascade is closely related to activation of transcription factors and leads to cell proliferation. In the present study, we investigated the effect of HES on the MAPK/ERK pathway using western blot analysis. As shown in Fig. 3A, the expression of Ras and phosphorylation of C-Raf, MEK and ERK were downregulated by HES in a time-dependent manner. Furthermore, the expression level of phosphorylated ERK and translocation to the nucleus were also decreased by HES (Fig. 3B). Thus, our results indicated that HES suppressed cell proliferation via regulation of the MAPK/ERK pathway in U87MG cells.

Effect of HES on the degradation of the cytoskeleton

As HES induced morphological cell change (Fig. 4A), we hypothesized that the expression of cytoskeleton-related proteins may be affected by HES. Thus, we measured protein expression associated with actin dynamic signaling using western blot analysis. As shown in Fig. 4B, actin dynamic signaling, including integrin β1, PI3K p110α, Rac, WAVE-2 and Arp-2, was decreased by HES in a dose-dependent manner. Therefore, these results showed that HES induced unstabilization of the cytoskeleton and affected cell survival through inactivation of actin dynamic signaling in U87MG cells.

HES inhibits the invasion and migration of U87MG cells

One of the most representative characteristics of glioblastoma is its invasive ability, diffusing into intact brain regions, which hinders elimination of the cancer by surgery (18). Thus, we investigated the inhibitory effects of HES on the metastatic ability of the U87MG cells. As shown in Fig. 5A, protein expression of MMP-2 and -9 was inhibited by HES in a dose-dependent manner. Additionally, wound healing assay results showed that the entire wound area in the HES-treated group was markedly decreased in comparison with the control group (Fig. 5B). In the Τranswell invasion assay, the number of invasive cells was also decreased in a dose-dependent manner (Fig. 5C). Hence, our findings indicated that HES inhibited cell invasion and migration of U87MG cells via negative regulation of MMP-2 and -9 expression.

Discussion

The present study is the first to demonstrate the anticancer potential of extracts from S. serratifolium. Among these extracts, a hexane extract from S. serratifolium (HES) sensitively induced inhibition of cell proliferation and morphological change in U87MG cells (Figs. 2B and 4A).

In glioblastoma, the Ras/Raf/MEK/ERK pathway is required for cell proliferation (18). Recently research has shown that various agents inhibiting this signaling pathway, such as sorafenib, an inhibitor of Raf, are able to inhibit proliferation and induce apoptosis in glioblastoma (20). This cascade signaling pathway is initiated by Ras activation, interacting with C-Raf and phosphorylating at serine (Ser) 338, tyrosine (Tyr) 341, threonine (Thr) 491 and Ser497 of C-Raf (21). Phosphorylated C-Raf directly activates MEK1 and MEK2, Tyr and Thr/Ser dual-specificity kinases, through phosphorylation at Ser217 and 221 (22). These kinases subsequently phosphorylate Thr202 and Tyr204 residues in p44/42 MAPK, ERK1/2, leading to phosphorylation of p90 ribosomal S6 kinase (p90RSK) which phosphorylates cAMP response element-binding protein (CREB) (23). ERK and CREB kinase phosphorylate transcription factors translocating to the nucleus for gene transcription (24). Our results demonstrated that the antiproliferative effects of HES were exerted by suppression of MAPK/ERK cascade signaling (Fig. 3).

The cytoskeleton is essential for morphological change, growth, survival, cell motility and cancer metastasis (9,25). Hence, regulation of the cell cytoskeleton and morphological changes may be used to treat metastatic tumors (26). Actin dynamic signaling is one of the crucial signaling pathways for regulation of the cell cytoskeleton (27). This signaling is initiated by integrin β1 which is located in the extracellular membrane and converts extracellular stimuli into intracellular signals (28). Integrin β1 regulates expression of phosphoinositide 3-kinase (PI3K) p110α and Rac (29,30). Rac, Rho-family of small G proteins, regulates the cytoskeleton through activation of WAVE-2 leading to activation of Arp-2, consequently facilitating morphological change (3133). This change regulates cytoskeletal stability, cell structure, cell motility and cell death (34,35). Morphological change in the U87MG cells was observed following treatment with HES (Fig. 4A). Western blot analysis results also revealed that HES suppressed reorganization of the cytoskeleton through regulation of actin dynamic signaling, including integrin β1, PI3K p110α, Rac1, WAVE-2 and Arp-2, resulting in inhibition of cell survival, growth and motility (Fig. 4B). Arp-2 and WAVE-2 are closely related to lamellipodium formation which regulates cell motility and invasion (36). In addition, degradation of the ECM by matrix metalloproteinases (MMPs) is required for the migration and invasion of metastatic cancer cells. MMPs, zinc-dependent endopeptidases, promote invasion and metastasis of cancer cells (37,38). Among the members of the MMP family, MMP-2 and -9 are crucial for degradation of ECM (39). Therefore, western blot analysis, Τranswell invasion and wound healing assays were performed to demonstrate the effects of HES on cell migration and invasion of the U87MG cells. As expected, the expression levels of MMP-2 and -9 were decreased in the glioblastoma cells following treatment with HES in a dose-dependent manner (Fig. 5A). Likewise, HES showed inhibitory effects on cell migration and invasion of the U87MG cells (Fig. 5B and C).

Taken together, the present study confirmed that HES has anticancer potential through regulation of the MAPK/ERK and actin dynamic pathways in human glioblastoma U87MG cells (Fig. 6). These findings demonstrated that HES may be used as a candidate anticancer agent for the treatment of human glioblastoma. To further analyze the detailed anticancer mechanisms of extracts from Sargarssuim serratifolium, we plan to use fractions of HES and investigate the effects of each fraction on glioblastoma.

Acknowledgments

The present study was supported by a Research Grant from KIOST-PKNU (Pukyong National University; 2015).

References

1 

Black PM: Brain tumors. Part 1. N Engl J Med. 324:1471–1476. 1991. View Article : Google Scholar : PubMed/NCBI

2 

Zaboronok A, Isobe T, Yamamoto T, Sato E, Takada K, Sakae T, Tsurushima H and Matsumura A: Proton beam irradiation stimulates migration and invasion of human U87 malignant glioma cells. J Radiat Res. 55:283–287. 2014. View Article : Google Scholar

3 

Papi A, Bartolini G, Ammar K, Guerra F, Ferreri AM, Rocchi P and Orlandi M: Inhibitory effects of retinoic acid and IIF on growth, migration and invasiveness in the U87MG human glioblastoma cell line. Oncol Rep. 18:1015–1021. 2007.PubMed/NCBI

4 

Liebmann C: Regulation of MAP kinase activity by peptide receptor signalling pathway: Paradigms of multiplicity. Cell Signal. 13:777–785. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Bernards A and Settleman J: GAP control: Regulating the regulators of small GTPases. Trends Cell Biol. 14:377–385. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Blalock WL, Weinstein-Oppenheimer C, Chang F, Hoyle PE, Wang XY, Algate PA, Franklin RA, Oberhaus SM, Steelman LS and McCubrey JA: Signal transduction, cell cycle regulatory, and anti-apoptotic pathways regulated by IL-3 in hematopoietic cells: Possible sites for intervention with anti-neoplastic drugs. Leukemia. 13:1109–1166. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Xing J, Ginty DD and Greenberg ME: Coupling of the RAS-MAPK pathway to gene activation by RSK2, a growth factor-regulated CREB kinase. Science. 273:959–963. 1996. View Article : Google Scholar : PubMed/NCBI

8 

Roux PP, Ballif BA, Anjum R, Gygi SP and Blenis J: Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase. Proc Natl Acad Sci USA. 101:13489–13494. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Fletcher DA and Mullins RD: Cell mechanics and the cytoskeleton. Nature. 463:485–492. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Yamazaki D, Kurisu S and Takenawa T: Regulation of cancer cell motility through actin reorganization. Cancer Sci. 96:379–386. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Mullins RD: How WASP-family proteins and the Arp2/3 complex convert intracellular signals into cytoskeletal structures. Curr Opin Cell Biol. 12:91–96. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Wu C, Asokan SB, Berginski ME, Haynes EM, Sharpless NE, Griffith JD, Gomez SM and Bear JE: Arp2/3 is critical for lamellipodia and response to extracellular matrix cues but is dispensable for chemotaxis. Cell. 148:973–987. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Machesky LM: Lamellipodia and filopodia in metastasis and invasion. FEBS Lett. 582:2102–2111. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Curran S and Murray GI: Matrix metalloproteinases in tumour invasion and metastasis. J Pathol. 189:300–308. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Lin HH, Chen JH, Chou FP and Wang CJ: Protocatechuic acid inhibits cancer cell metastasis involving the down-regulation of Ras/Akt/NF-κB pathway and MMP-2 production by targeting RhoB activation. Br J Pharmacol. 162:237–254. 2011. View Article : Google Scholar :

16 

Liu L, Heinrich M, Myers S and Dworjanyn SA: Towards a better understanding of medicinal uses of the brown seaweed Sargassum in Traditional Chinese Medicine: A phytochemical and pharmacological review. J Ethnopharmacol. 142:591–619. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Huang GJ, Yang CM, Chang YS, Amagaya S, Wang HC, Hou WC, Huang SS and Hu ML: Hispolon suppresses SK-Hep1 human hepatoma cell metastasis by inhibiting matrix metallo-proteinase-2/9 and urokinase-plasminogen activator through the PI3K/akt and ERK signaling pathways. J Agric Food Chem. 58:9468–9475. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Clarke J, Butowski N and Chang S: Recent advances in therapy for glioblastoma. Arch Neurol. 67:279–283. 2010.PubMed/NCBI

20 

Carra E, Barbieri F, Marubbi D, Pattarozzi A, Favoni RE, Florio T and Daga A: Sorafenib selectively depletes human glioblastoma tumor-initiating cells from primary cultures. Cell Cycle. 12:491–500. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Chong H, Lee J and Guan KL: Positive and negative regulation of Raf kinase activity and function by phosphorylation. EMBO J. 20:3716–3727. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Alessi DR, Saito Y, Campbell DG, Cohen P, Sithanandam G, Rapp U, Ashworth A, Marshall CJ and Cowley S: Identification of the sites in MAP kinase kinase-1 phosphorylated by p74raf-1. EMBO J. 13:1610–1619. 1994.PubMed/NCBI

23 

Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Bonni A, Brunet A, West AE, Datta SR, Takasu MA and Greenberg ME: Cell survival promoted by the Ras-MAPK signaling pathway by transcription-dependent and -independent mechanisms. Science. 286:1358–1362. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Colomba A and Ridley AJ: Analyzing the roles of Rho GTPases in cancer cell migration with a live cell imaging 3D-morphology-based assay. Methods Mol Biol. 1120:327–337. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Najm P and El-Sibai M: Palladin regulation of the actin structures needed for cancer invasion. Cell Adh Migr. 8:29–35. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Edwards KA, Demsky M, Montague RA, Weymouth N and Kiehart DP: GFP-moesin illuminates actin cytoskeleton dynamics in living tissue and demonstrates cell shape changes during morphogenesis in Drosophila. Dev Biol. 191:103–117. 1997. View Article : Google Scholar : PubMed/NCBI

28 

Harburger DS and Calderwood DA: Integrin signalling at a glance. J Cell Sci. 122:159–163. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Miyamoto S, Teramoto H, Coso OA, Gutkind JS, Burbelo PD, Akiyama SK and Yamada KM: Integrin function: Molecular hierarchies of cytoskeletal and signaling molecules. J Cell Biol. 131:791–805. 1995. View Article : Google Scholar : PubMed/NCBI

30 

Legate KR and Fässler R: Mechanisms that regulate adaptor binding to beta-integrin cytoplasmic tails. J Cell Sci. 122:187–198. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Kurisu S, Suetsugu S, Yamazaki D, Yamaguchi H and Takenawa T: Rac-WAVE2 signaling is involved in the invasive and metastatic phenotypes of murine melanoma cells. Oncogene. 24:1309–1319. 2005. View Article : Google Scholar

33 

Miki H, Suetsugu S and Takenawa T: WAVE, a novel WASP-family protein involved in actin reorganization induced by Rac. EMBO J. 17:6932–6941. 1998. View Article : Google Scholar : PubMed/NCBI

34 

Pollard TD and Cooper JA: Actin, a central player in cell shape and movement. Science. 326:1208–1212. 2009. View Article : Google Scholar : PubMed/NCBI

35 

Wang F, Liu DZ, Xu H, Li Y, Wang W, Liu BL and Zhang LY: Thapsigargin induces apoptosis by impairing cytoskeleton dynamics in human lung adenocarcinoma cells. Scientific World Journal. 2014:6190502014.PubMed/NCBI

36 

Small JV, Stradal T, Vignal E and Rottner K: The lamellipodium: Where motility begins. Trends Cell Biol. 12:112–120. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Chen JH, Lin HH, Chiang TA, Hsu JD, Ho HH, Lee YC and Wang CJ: Gaseous nitrogen oxide promotes human lung cancer cell line A549 migration, invasion, and metastasis via iNoS-mediated MMP-2 production. Toxicol Sci. 106:364–375. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Westermarck J and Kähäri VM: Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J. 13:781–792. 1999.PubMed/NCBI

39 

Parks WC and Shapiro SD: Matrix metalloproteinases in lung biology. Respir Res. 2:10–19. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 34 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kang C, Park M, Kim N, Lee J, Oh C, Kim H and Kim G: Hexane extract from Sargassum serratifolium inhibits the cell proliferation and metastatic ability of human glioblastoma U87MG cells. Oncol Rep 34: 2602-2608, 2015
APA
Kang, C., Park, M., Kim, N., Lee, J., Oh, C., Kim, H., & Kim, G. (2015). Hexane extract from Sargassum serratifolium inhibits the cell proliferation and metastatic ability of human glioblastoma U87MG cells. Oncology Reports, 34, 2602-2608. https://doi.org/10.3892/or.2015.4222
MLA
Kang, C., Park, M., Kim, N., Lee, J., Oh, C., Kim, H., Kim, G."Hexane extract from Sargassum serratifolium inhibits the cell proliferation and metastatic ability of human glioblastoma U87MG cells". Oncology Reports 34.5 (2015): 2602-2608.
Chicago
Kang, C., Park, M., Kim, N., Lee, J., Oh, C., Kim, H., Kim, G."Hexane extract from Sargassum serratifolium inhibits the cell proliferation and metastatic ability of human glioblastoma U87MG cells". Oncology Reports 34, no. 5 (2015): 2602-2608. https://doi.org/10.3892/or.2015.4222