• 검색 결과가 없습니다.

Fucoidan Induces Apoptosis in A2058 Cells through ROS-exposed Activation of MAPKs Signaling Pathway

N/A
N/A
Protected

Academic year: 2022

Share "Fucoidan Induces Apoptosis in A2058 Cells through ROS-exposed Activation of MAPKs Signaling Pathway"

Copied!
9
0
0

로드 중.... (전체 텍스트 보기)

전체 글

(1)

https://doi.org/10.20307/nps.2020.26.3.191

191

Fucoidan Induces Apoptosis in A2058 Cells through ROS-exposed Activation of MAPKs Signaling Pathway

Yea Seong Ryu1,2, Jin Won Hyun2,*, and Ha Sook Chung1,*

1Department of Food and Nutrition, College of Natural Sciences, Duksung Women’s University, Seoul 01369, Korea

2Department of Biochemistry, School of Medicine and Institute for Nuclear Science and Technology, Jeju National University, Jeju 63243, Korea

Abstract  Fucoidan, a natural component of brown seaweed, has various biological activities such as anti- cancer activity, anti-oxidant, and anti-inflammatory against various cancer cells. However, the fucoidan has been implicated in melanoma cells via apoptosis signaling pathway. Therefore, we investigated apoptosis with fucoidan in A2058 human melanoma cells with dose- and time-dependent manners. In our results, A2058 cells viability decreased at relatively short-time and low-concentration through fucoidan. This effects of fucoidan on A2058 cells appeared to be mediated by the induction of apoptosis, as manifested by morphological changes through DNA-binding dye Hoechst 33342 staining. When a dose of 80 µg/mL fucoidan was treated, the cells were observed: crescent or ring-like structure, chromatin condensation, and nuclear fragmentation. With the increase at 100 µg/mL fucoidan, the cell membrane is intact throughout the total process, including membrane blebbing and loss of membrane integrity as well as increase of sub-G1 DNA. Furthermore, to understand the exact mechanism of fucoidan-treated in A2058 cells, western blotting was performed to detect apoptosis-related protein expression. In this study, Bcl-2 family proteins can be regulated by fucoidan, suggesting that fucoidan- induced apoptosis is modulated by intrinsic pathway. Therefore, expression of Bcl-2 and Bax may result in altered permeability, activating caspase-3 and caspase-9. And the cleaved form of poly ADP-ribose polymerase was detected in fucoidan-treated A2058 cells. These results suggest that A2058 cells are highly sensitive to growth inhibition by fucoidan via apoptosis, as evidenced by activation of extracellular signal-regulated kinases/

p38/Bcl-2 family signaling, as well as alteration in caspase-9 and caspase-3.

Keywords  Fucoidan, apoptosis, ROS, MAPKs, A2058, melanoma cells

Introduction

Fucoidan, a sulfated polysaccharide, is found in various brown algae, such as Undaria pinnatifida, Fucus vesiculosus and Ecklonia cava.1 The structures and compositions of fucoidan are complex and vary from species to species. For example, fucoidan from species of Fucus vesiculosus, a common harvest crop of northern hemisphere kelp, is the most common source of experi- mental at present. It has basic composition, mainly being composed of 44.1% fucose, 26.3% sulfate and 31.1% ash, plus a little amino glucose2 and small quantities of

monosaccharides.3

Accumulating studies have presented that fucoidan shows the inhibition of coagulant,4 thrombotic,5 cancer,6 viral,7 and inflammatory activities.8 Also, fucoidan- mediated apoptosis in cancer cells via reactive oxygen species (ROS) production, which are responsible for the loss of Δψm and the down-regulation of Bcl-2 proteins.

Besides inflammation, ROS can also cause cell death in cancer by anti-tumor reagents. Fucoidan was found to block proliferation and induce apoptosis in human lymphoma HS-Sultan cell lines.9

Apoptosis is a self-destruction mechanism involved in developmental sculpturing, tissue homeostasis, and the removal of unwanted cells. Disruption of the regulation of apoptosis may result in various diseases, including cancer.10 An apoptotic signal transduction pathway appears to be as follows: many pro-apoptotic signals initially activate separate signaling pathways, which eventually converge into a common mechanism driven by caspases.11,12

*Author for correspondence

Jin Won Hyun, Department of Biochemistry, School of Medicine and Institute for Nuclear Science and Technology, Jeju National Uni- versity, Jeju 63243, Korea

Tel: 82-64-754-3838; E-mail: jinwonh@jejunu.ac.kr

Ha Sook Chung, Department of Food and Nutrition, College of Nat- ural Sciences, Duksung Women’s University, Seoul 01369, Korea Tel: +82-2-901-8593; E-mail: hasook@duksung.ac.kr

(2)

192 Natural Product Sciences This mechanism is negatively regulated by several sets

of genes, the best characterized of which are the Bcl-2 family.13 There is growing evidence for a role of mitochondria in apoptosis induction, leading to the oligomerization of adapter proteins and pro-caspases, resulting in auto activation of initiator caspases.14 Release of cytochrome c appears to be the central event, since it is critical for aggregation of the adapter molecule (Apaf1).

The pro-apoptotic member of the Bcl-2 family Bax can directly cause mitochondria to release cytochrome c.15 The ability of some Bcl-2 family members, including Bax, to form ion channels has suggested that these proteins open pores in the outer mitochondrial membrane, allowing exit of cytochrome c.14

Taken together, ROS induces the oxidation of mito- chondrial pores which contribute to cytochrome c release by mitochondrial membrane potential disruption.16 It seems that mitochondria are both source and target of ROS. In this study, we investigated the molecular mechanisms of fucoidan with cytotoxic effects through ERK/p38/Bcl-2 family signaling in A2058 human melanoma cells.

Experimental

General procedure  [3-(4,5-Dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium] bromide (MTT) and fucoidan (F8190) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), and streptomycin- penicillin were purchased from Gibco (Carlsbad, CA, USA). The primary p38, phosphor-p38, JNK, phopho- JNK, ERK1/2, phospho-ERK1/2, caspase-9, caspase-3 and PARP antibodies were purchased from Cell Signaling Technologies (Danvers, MA, USA). All other chemicals and reagents were of the highest analytical grade.

Cell culture and cell viability assay  The human keratinocyte cell line (HaCaT) was purchased from the American Type Culture Collection (Manassas, VA, USA) and A2058 cells was obtained from Professor Hyun, Jin Won, School of Medicine and Institute for Nuclear Science and Technology, JeJu National University, JeJu, Korea. Cells were maintained and MTT assay have been conducted as previously reported.17 For experiments, fucoidan was dissolved in dimethylsulfoxide (DMSO) at concentrations not exceeding 0.01% and was then directly applied to the culture medium.

Nuclear staining with Hoechst 33342  The nuclear morphology of the cells was observed using Hoechst 33342 DNA-specific blue fluorescent dye, followed by

previously reported.18,19 A fucoidan-treated A2058 cells were observed under a fluorescence microscope (Olympus Optical Co., Tokyo, Japan).

Immunocytochemistry  To obtain immunological detection on tissues, including those tied directly to primary antibodies, immunochemistry was performed as reported.17 The immune reacted primary antibody was detected by incubating cells with FITC-conjugated secon- dary antibody (Jackson Immuno Research Laboratories, West Grove, PA, USA). Images were collected using a Zeiss confocal microscope and Zeiss LSM 510 software (version 3.2; Zeiss AG, Oberkochen, Germany).

Measurement of intracellular ROS  The Intracellular ROS generation was assessed using the stable nonpolar dye DCF-DA (Sigma-Aldrich; St. Louis, MO, USA) followed by published method.20 ROS production was measured by FACSCalibur flowcytometry (Becton Dickinson, San Jose, CA, USA).

Cell cycle analysis  Cell cycle analysis was carried out to determine the proportion of apoptotic sub-G1 hypodiploid cells followed by previously reported.21 Fluorescence emitted from the PI-DNA complex was quantified using FACS Calibur flow cytometer (BD Biosciences, San Jose, CA, USA).

Apoptosis analysis  Apoptosis analysis was performed as published reports.22 Briefly, Annexin V/PI double staining assay was then carried out in order to differentiate between early and late apoptosis stages.

Protein extraction and western blot analysis  Western blot analysis performed as previously described.22 Fluorescence was quantified using FACSCalibur flow cytometer. The membranes detected by an enhanced chemiluminescence western blotting detection kit (Bio- Rad Laboratories, Inc., Hercules, CA, USA). β-Actin and TBP were used as loading controls for the nuclear and whole cell extracts, respectively. The values for the specific protein levels are presented as the fold-change relative to the control and densitometry was performed using Image J (National Institutes of Health, Bethesda, MD, USA).

Statistical analysis  All measurements made in triplicate, and all values are given as the mean ± the standard deviation. The results were subjected to an analysis of variance followed by Tukey’s test in order to analyze differences between conditions. In each case, a p- value of < 0.05 was determined statistically significant.

Results and discussion

To examine anti-proliferative activities in A2058 cells

(3)

exposed fucoidan at concentrations of 40, 80, and 100 µg/

mL for 24, 48, and 72 h, and cell viability was determined by MTT assay. As shown in Fig. 1A, fucoidan inhibited the proliferation of A2058 cells in dose- and time- dependent manners. Treatment for 72 h, cell viability of fucoidan-treated A2058 cells was decreased by 77.7%, 62.6%, and 49.7% at the concentration of 40, 80, and 100 µg/mL, respectively with IC50 values of 100 µg/mL. At 150 µg/mL and 200 µg/mL, fucoidan markedly decreased cell proliferation 20.9% and 7.3%, respectively. Human normal keratinocytes (HaCaT) were treated dose-depen- dently for 72 h to compare the cytotoxic effects. (Fig.

1B). These results indicate that HaCaT cells were more resistant to the fucoidan-treated melanoma cells. Therefore, the concentrations of 40, 80, and 100 µg/mL were used for the experiments.

Apoptosis is characterized by a series of morphology changes and chromatin condensation of cellular nucleus by distinct mechanisms. In order to determine whether the fucoidan-induced growth inhibition in A2058 cells was associated with the induction of apoptosis, DNA-binding dye Hoechst 33342 staining was performed. As shown in Fig. 2, the nuclei showed homogeneously stained and an intact structure in control cells. After the treatment with 20 µg/mL fucoidan, cells exhibited nearly no change (data not shown). When a dose 40 µg/mL of fucoidan was incubated, there was a slight cell shrinkage and reduction in cell numbers. However, with the increase at 80 µg/mL fucoidan, the marked apoptotic morphologic alterations were observed: membrane blebbing, nuclear fragmentation, chromatin condensation, increased fluorescent intensity.

Especially, crescent-shaped nuclei, which are one of the typical characteristics of apoptotic cells, were also found.

At the higher dose 100 µg/mL of fucoidan, these characteristics were even more showed and the cells exhibited partial detachment. These results suggest that fucoidan showed anti-proliferative activities in a dose- dependent manner, and is consistent with MTT results.

In case of mitochondrial-dependent apoptosis, it is largely regulated by direct or indirect intracellular ROS.16 The ROS production was examined to investigate the possible mechanisms of fucoidan-induced apoptosis using Fig. 1. (A) Cytotoxic effects of fucoidan in A2058 cells. Cell

viability at the indicated concentrations of fucoidan at 24, 48, and 72 h were assessed by MTT assay. *p < 0.05, significantly different from control cells for 24 h; **p < 0.05, significantly different from control cells for 48 h; #p < 0.05, significantly different from control cells for 72 h. (B) Cytotoxic effects of fucoidan in HaCaT human keraninocytes. Cell viability at the indicated concentrations of fucoidan at 72 h were assessed by MTT assay. *p < 0.05, significantly different from control cells.

Fig. 2. Microscopy image of fucoidan treated A2058 cells. After incubation with various concentrations of fucoidan for 72 h, the cells were observed by fluorescent microscopy using Hoechst 33342 staining (arrow indicates the formation of .bodies).

(4)

194 Natural Product Sciences

specific fluorescence probes H2DCF-DA. The treatment with fucoidan caused a right shift of the H2DCF-DA, indicating the increase of hydroxyl radicals and hydrogen peroxide levels, followed by cell death (Fig. 3A). However, the treatment of fucoidan and N-acetyl-L-cysteine (NAC) slightly attenuated relative to only fucoidan treatment, suggesting that NAC, antioxidant, might be ROS scavenger (Fig. 3B). These results presented that ROS might be involved in fucoidan-mediated apoptotic process.

The regulation of cell cycle is critical process for the growth and division of cancer. Flow cytometry was performed on A2058 cells which were treated with 40,

80, and 100 µg/mL of fucoidan to investigate the effect on cell cycle progression. As shown in Figs. 4A and 4B, control cells showed 11.1% of sub-G1 phase, whereas fucoidan-treated cells caused a dose-dependent accumu- lation of cells by 57.1%, 63.8%, and 83.2% at 40, 80, and 100 µg/mL. Moreover, it shows the significant decrease in the S phase by 12.6%, 11.0%, and 6.4%, and in G2/M phase by 8.4%, 7.6%, and 3.4% at 40, 80, and 100 µg/

mL. This is in agreement with a previous study of cell viability assay and cellular phenotypic changes, such as cell fragmentation. These results present that fucoidan induce apoptosis in A2058 cells in dose dependently.

To further investigated the apoptotic cells, Annexin V and PI double staining were used and were analyzed by flow cytometry analysis. The Annexin V+/PI staining represent the early apoptotic cells, since the high affinity of Annexin V-FITC with phosphatidylserine (PS). The Fig. 3. (A) Changes in ROS levels in fucoidan-treated A2058

cells. DCF-DA staining was conducted to measure changes in ROS level. After incubation with NAC and fucoidan for 72 h, ROS levels were measured by flow cytometry. (B) Statistical analysis for ROS levels. *p < 0.05, significantly different from control cells; **p < 0.05, significantly different from fucoidan- treated cells.

Fig. 4. Effects of fucoidan on the cell cycle progression in A2058 cells. (A) After incubation with various concentrations of fucoidan for 72 h, sub-G1 cells were detected by flow cytometry after propidium iodide staining. (B) Statistical analysis for apoptosis. *p < 0.05, significantly different from control cells in Sub-G1 phase; **p < 0.05, significantly different from control cells in G1 phase; ***p < 0.05, significantly different from control cells in S phase; #p < 0.05, significantly different from control cells in G2/M phase.

(5)

plasma membrane alteration: translocation of PS from the inner side of the plasma membrane to the outer layer is the characteristic features of early apoptotic cells. The Annexin V/PI+ staining represent the necrotic cells, due to a membrane-impermeant DNA binding dye that cannot penetrate intact cell membrane. During necrosis, Propidium iodide (PI) enters dead cells with comprised membranes and stains dead nucleated cells to generate fluorescence.23 And the Annexin V-/PI-population represents viable cells and Annexin V+/PI+ as late apoptotic cells.

The results showed that the fucoidan-treated cells significantly increased apoptotic cells populations, com- pared to untreated control cells (Fig. 5A). The early apoptotic cells were increased 10.8%, 13.1%, 33.7% at 40, 80, and 100 µg/mL of fucoidan, respectively, compared with 2.1% for the control. The late apoptotic cells were

also increased 21.2%, 20.5%, and 28.1% at 40, 80, and 100 µg/mL, respectively, compared with 7.48% for the control. The total apoptotic cells were increased from 9.6% in control cells to 32.0%, 33.6%, and 61.8% at 40, 80, and 100 µg/mL of fucoidan, respectively (Fig. 5B).

Also, the early apoptotic cells were increased than late apoptotic cells at 40 and 80 µg/mL. However, the late apoptotic cells were much greater than that of early apoptotic cells. These results present fucoidan induced anti-proliferative activities against A2058 cells, which were caused by inducing apoptosis in a dose-dependent fashion.

Caspases are a family of cysteine protease that plays essential roles in a cascade by apoptosis signaling, this cascade is proteins cleavage in cell. The intrinsic mitochondrial pathway is controlled by the Bcl-2 family proteins, which prevent mitochondrial permeability transition pores from opening during apoptosis.24 To study whether fucoidan induced apoptosis through the cascade- dependent pathway, western blotting was conducted on cells which were treated with fucoidan. The A2058 melanoma cells exposed to fucoidan showed a concentra- tion-dependent reduction in the level of Bcl-2 protein, with a concomitant increase in the level of Bax. With the change of Bcl-2 family proteins, the activation of caspase- 9 and caspase-3, an effective caspase, was accompanied.

Moreover, poly ADP-ribose polymerase (PARP) cleavage, Fig. 5. Effects of fucoidan on apoptosis in A2058 cells. (A) Flow

cytometric analysis of A2058 cells incubated with fucoidan for 72 h. The right bottom quadrant represents Annexin V-stained cells (early-phase apoptotic cells). The top right quadrant represents PI- and Annexin V-stained cells (late-phase apoptotic cells). (B) Statistical analysis of apoptosis. *p < 0.05, significantly different from control cells in intact cells; **p < 0.05, significantly different from control cells in early apoptotic; ***p < 0.05, significantly different from control cells in late apoptotic; #p <

0.05, significantly different from control cells in total apoptotic.

Fig. 6. Effects of fucoidan on expression of apoptosis-related proteins in A2058 cells. Cells were treated with fucoidan (0, 40, 80, and 100 µg/mL) for 72 h. The cell lysates were electro- phoresed, and western blotting with Bax, Bcl-2, caspase-9, caspase- 3, and cleaved PARP antibodies.

(6)

196 Natural Product Sciences

an indicator of apoptosis, was also elevated (Fig. 6).

These results presented that fucoidan can lead to apoptosis by caspase-related protein family and might be through intrinsic mitochondrial apoptosis pathway.

Mitogen-activated protein kinases (MAPKs) signaling cascades, including c-Jun amino-terminal kinases (JNKs), extracellular signal-regulated kinases (ERKs), and p38 kinase, are found in all eukaryotes and play a central role in regulating cell proliferation, differentiation and apoptosis.25 As shown in Fig. 7, the expressions of non- phosphorylated JNK, ERK1/2 and p38 MAPK were not changed upon treatment with fucoidan. By contrast, the accumulation of phosphorylated ERK and p38 MAPK were significantly elevated in a time-dependent manner, whereas no significant changes of phosphorylation of JNK were observed. These results indicated that 100 µg/

mL of fucoidan induces apoptosis through ERK and p38 MAPK in A2058 cells.

To further determine whether ERK and p38 are involved in the 100 µg/mL of fucoidan-induced A2058 cytotoxicity, the kinase-specific inhibitors, PD98059, or SB203580, were incubated. As shown in Fig. 8A, the level of phosphorylated p-ERK significantly decreased in response to co-treatment with PD98059 and fucoidan.

Moreover, fucoidan-induced cell death significantly attenuated relative to only focoidan treatment (Fig. 8B).

Likewise, co-treatment with fucoidan and SB203580 effectively blocked fucoidan mediated p-p38 up-regula- tion (Fig. 8C). Also fucoidan-induced cell death was slightly decreased by SB203580 (Fig. 8D). The cumulative

results suggested that ERK and p38 MAPK might involve in fucoidan-induced mitochondrial apoptosis.

In the present study, fucoidan showed the apoptotic effects of on A2058 human melanoma cells via ERK and p38 signal pathway. This study implies that fucoidan might be promising candidates for anti-tumor on melanoma cells.

Apoptosis is commonly called physiological pro- grammed cell death and is generally distinguished from necrosis, which is considered to be a toxic process. In normal cells, apoptosis plays an important role in the growth and death and as a cellular homeostatic mechanism to maintain cell populations. Typically, apoptosis is characterized by morphological hallmarks; chromatin condensation, cell shrinkage, nuclear fragmentation and, which are accompanied by rounding up of the cell and retraction of pseudopods.26

In the early stages of apoptosis, there are alterations of PS, which translocate the outer layer from the inner side of the plasma membrane. This allows early recognition of dead cells by which PS exposes at the external surface of the cell.27 This is followed by an enabling characteristic parabens can cause DNA damage in the short term, but more study is needed to figure out long-term and low- dose mixtures.28

Recently, apoptotic DNA fragmentation may be a cooperative activity between caspase-activated deoxyri- bonuclease (DNase) and PARP-regulated DNAS1L3, an endoplasmic reticulum-localized endonuclease that translocate to the nucleus.29 The study discovered the anti- death activity of Bcl-2 in a system where apoptosis was induced by deprivation of IL-3.30. Bcl-2 was shown to prevent apoptosis by serum deprivation, heat shock, and chemotherapeutic reagents.31

There are two main groups of the Bcl-2 family proteins, namely the pro-apoptotic proteins and the anti-apoptotic proteins. The anti-apoptotic proteins prevent apoptosis by blocking cytochrome c release from mitochondria. The pro-apoptotic proteins regulate by promoting such release.

It is not the absolute quantity but rather the ratio between the anti-apoptotic and pro-apoptotic proteins of the Bcl-2 family may determine the membrane potential loss and the membrane permeability transition.32 Thus, once cytochrome c released from mitochondria, cytochrome c directly activated caspases. Caspases are cysteine-yl aspartate proteinases (cysteine proteases that cleave their substrates following an Asp residue), which is essential phase in apoptosis. While the upstream caspases for the intrinsic pathway is caspase-9, of the extrinsic pathway is caspase-8. The intrinsic and extrinsic pathways converge Fig. 7. Regulations of MAPKs in treated with 100 µg/mL of

fucoidan in A2058 cells. Equal amounts of cell lysates were electrophoresed and JNK, ERK and p38 and their phosphorylated expression form were detected by western blotting analysis with corresponding antibodies.

(7)

to caspase-3. Caspase-3, then cleaves the inhibitor of the caspase-activated DNase, which is responsible for nuclear apoptosis. In this pathway, cell death signals lead to cytochrome c release from the mitochondria, which binds and facilitates the formation of the apoptosome structure that recruits and activates caspase-9.33,34 Caspase-3 is the main downstream effector that cleaves the majority of the cellular substrates in apoptotic cells. It is activated following cleavage by caspase-8 or caspase-9, but not by caspase-2.35 Recent evidence has demonstrated that brown algae induce apoptosis by triggering accumulation of ROS.

Especially, several reports suggested that the inhibitory effects of fucoidan in hepatocellular carcinoma cells and breast cancer cells, thereby offering potential chemo- preventive activities.36-38 In the present study, we examined the basics of hypothesis testing, and investigated apoptosis

in A2058 human melanoma cells, which were treated with various concentrations of fucoidan (0-100 μg/mL).

The MTT assay for viability of A2058 cells provided proliferation inhibitory in a dose- and time-dependent manner and the IC50 of cytotoxicity was determined to be 100 μg/mL. The non-malignant HaCaT cell line exhibits lower sensitivity to fucoidan treatment than the malignant cell lines.

A study reported that cell proliferation inhibitory of MCF-7 and MDB-MB-231 breast cancer cells decreased 60% and 41% when exposed to 410 μg/mL fucoidan for 96 h. And the growth inhibitory of HeLa epithelial adenocarcinoma cells and HT1080 fibro-sarcoma cells decreased 52% and 40% for the same dose and time periods.36 In this study, the A2058 melanoma cell viability with decreased at a relatively short-time and low–

Fig. 8. Role of ERK and p38 MAPK in treated with 100 µg/mL of fucoidan on apoptosis. (A) Role of ERK in fucoidan-induced apoptosis in A2058 cells. The cells were stimulated with 100 µg/mL of fucoidan after pre-treatment with PD98059. Equal amounts of cell lysates were electrophoresed and ERK protein expression form was detected by western blotting analysis with corresponding antibodies.

Relative abundance of proteins was calculated for the p-ERK/ERK ratios. (B) Microscopy images of fucoidan-induced apoptosis by ERK inhibitor in A2058 cells. The cells were stimulated with 100 µg/mL of fucoidan after pre-treatment with PD98059. Cells were observed by fluorescent microscopy using Hoechst 33342 staining (arrow indicates the formation of apoptotic bodies). (C) Role of p38 in fucoidan- induced apoptosis in A2058 cells. The cells were stimulated with 100 μg/mL of fucoidan after pre-treatment with SB203580. Equal amounts of cell lysates were electrophoresed and p38 protein expression form was detected by western blotting analysis with corresponding antibodies. Relative abundance of proteins was calculated for the p-p-38/p38 ratios. Microscopy images of fucoidan- induced apoptosis by p38 inhibitors in A2058 cells. The cells were stimulated with 100 µg/mL of fucoidan after pre-treatment with SB203580. Cells were observed by fluorescent microscopy using Hoechst 33342 staining (arrow indicates the formation of apoptotic bodies). (D) Microscopy images of fucoidan-induced apoptosis by p38 inhibitors in A2058 cells. The cells were stimulated with 100 µg/

mL of fucoidan after pre-treatment with SB203580. Cells were observed by fluorescent microscopy using Hoechst 33342 staining (arrow indicates the formation of apoptotic bodies)

(8)

198 Natural Product Sciences concentration, indicating that melanoma cells were more

sensitive than the other cell lines to fucoidan. This effects of fucoidan on melanoma cells appeared to be mediated by the induction of apoptosis, as manifested by mor- phological changes through DNA-binding dye Hoechst 33342 staining. The cell was not affected by 20 µg/mL fucoidan, but with increasing at a 40 µg/mL fucoidan, the population of live cells dropped slightly and morphology of the cells changed by reduction in cellular volume (pyknosis). When a dose of 80 µg/mL fucoidan was treated, the cells were observed: crescent or ring-like structure, chromatin condensation, and nuclear fragmen- tation. With the increase at a dose of 100 µg/mL fucoidan, the cell membrane is intact throughout the total process, including membrane blebbing and loss of membrane integrity. Many observations suggest that cell cycle and apoptosis are an important to maintain homoeostasis between cell proliferation and cell death. Therefore, deregulation of the cell cycle is one of the most frequent alterations during tumor development.39 The inhibition of cell cycle progression is considered an effective strategy for the control of tumor growth.40 The present study demonstrated that fucoidan-treated with A2058 cells was confirmed by observations of sub-G1 DNA accumulation, whereas the S phase and G2/M phase were decreased, leading to apoptosis. A previous study has shown the induction of apoptosis by fucoidan on the MCF-7 breast cancer cell line via sub-G1 arrest.36 To understand the mechanism of fucoidan in A2058 cells, western blotting was performed to detect the expression of apoptosis- associated proteins. The mitochondrial-related pathway is regulated by anti-apoptotic (Bcl-2, Bcl-x, and Bcl-XL) and pro-apoptotic members (Bax, Bak, Bid, Bad and Bim) of the Bcl-2 family. The anti-apoptotic proteins on the outer membranes of mitochondria maintain the integrity of the mitochondria, through inhibiting apoptosis in the presence of various apoptotic stimuli.41 In response to an apoptotic stimulus, the pro-apoptotic proteins reside in the cytosol and translocate to the mitochondria, which leads to the formation of membrane pores at the mitochondrial membranes and the activation of caspase cascade.42,43 Therefore, the ratio of Bax and Bcl-2 protein determines the susceptibility of cells to cell survival as well as death.44 Caspase-3 is one of the key protagonists of apoptosis, because it is either partially or completely responsible for the proteolytic cleavage of many key proteins. PARP is important for cell viability, but its cleavage facilitates cellular disassembly and serves as a marker of cells undergoing apoptosis.42,45 Therefore, the western blotting experiments indicated that caspase-9 and

caspase-3 appear to be activated in fucoidan-induced A2058 cells. And the cleaved form of PAPR was detected in fucoidan-treated A2058 cells. These data indicate that fucoidan induced apoptosis via the mitochondrial pathway.

These results suggest that A2058 human melanoma cells are highly sensitive to growth inhibition by fucoidan via the activation of apoptosis, as evidenced by activation of Bcl-2-mediated signaling, as well as alteration in caspase- 9 and caspase-3.

Conflict of interest The authors declare no conflict of interest.

References

(1) Patankar, M. S.; Oehninger, S.; Barnett, T.; Williams, R. L.; Clark, G. F. J. Biol. Chem. 1993, 268, 21770-21776.

(2) Nishino, T.; Nishioka, C.; Ura, H.; Nagumo, T. Carbohydr. Res.

1994, 255, 213-224.

(3) Bilan, M. I.; Grachev, A. A.; Ustuzhanina, N. E.; Shashkov, A. S.;

Nifantiev, N. E.; Usov, A. I. Carbohydr. Res. 2002, 337, 719-730.

(4) Kim, K. J.; Lee, O. H.; Lee, H. H.; Lee, B. Y. Toxicology 2010, 267, 154-158.

(5) Ren, R.; Azuma, Y.; Ojima, T.; Hashimoto, T.; Mizuno, M.;

Nishitani, Y.; Yoshida, M.; Azuma, T.; Kanazawa, K. Br. J. Nutr. 2013, 110, 880-890.

(6) Cho, T. M.; Kim, W. J.; Moon, S. K. Food Chem. Toxicol. 2014, 64, 344-352.

(7) Zhang, W.; Oda, T.; Yu, Q.; Jin, J. O. Mar. Drugs 2015, 13, 1084- 1104.

(8) Kim, K. J.; Yoon, K. Y.; Lee, B. Y. Fitoterapia 2012, 83, 1628-1635.

(9) Aisa, Y.; Miyakawa, Y.; Nakazato, T.; Shibata, H.; Saito, K.; Ikeda, Y.; Kizaki, M. Am. J. Hematol. 2005, 78, 7-14.

(10) Portt, L.; Norman, G.; Clapp, C.; Greenwood, M.; Greenwood, M.

T. Biochim. Biophys. Acta 2011, 1813, 238-259.

(11) Igney, F. H.; Krammer, P. H. Nat. Rev. Cancer 2002, 2, 277-288.

(12) Martinvalet, D.; Zhu, P.; Lieberman, J. Immunity 2005, 22, 355- 370.(13) Adams, J. M.; Cory, S. Science 1998, 281, 1322-1326.

(14) Green, D. R.; Reed, J. C. Science 1998, 281, 1309-1312.

(15) Park, H. Y.; Kim, G. Y.; Kwon, T. K.; Hwang, H. J.; Kim, N. D.;

Yoo, Y. H.; Choi, Y. H. Mutat. Res. 2013, 751, 101-108.

(16) Dalla Via, L.; García-Argáez, A. N.; Martínez-Vázquez, M.;

Grancara, S.; Martinis, P.; Toninello, A. Curr. Pharm. Des. 2014, 20, 223- 244.(17) Ryu, M. J.; Chung, H. S. Mol. Med. Rep. 2016, 14, 3255-3260.

(18) Matsumoto, T.; Tabata, K.; Suzuki, T. Biol. Pharm. Bull. 2014, 37, 633-641.

(19) Visagie, M. H.; Joubert, A. M. Mol. Cell Biochem. 2011, 357, 343- 352.(20) Ramyaa, P.; Krishnaswamy, R.; Padma, V. V. Biochim. Biophys.

Acta 2014, 1840, 681-692.

(21) Yu, H. Y.; Kim, S. O.; Jin, C. Y.; Kim, G. Y.; Kim, W. J.; Yoo, Y.

H.; Choi, Y. H. Biomol. Ther. (Seoul) 2014, 22, 184-192.

(22) Guon, T. E.; Chung, H. S. Oncol. Lett. 2016, 11, 2463-2470.

(23) Tripathi, P.; Chandra, M.; Misra, M. K. Indian J. Clin. Biochem.

2010, 25, 302-306.

(24) Armstrong, J. S. Br. J. Pharmacol. 2007, 151, 1154-1165.

(9)

(25) Kim, E. K.; Choi, E. J. Arch. Toxicol. 2015, 89, 867-882.

(26) Kumar, V.; Abbas, A. K.; Fausto, N.; Aster, J. C. Robins and Cotran: Pathologic Basis and Disease. 8 ed; Elsevier : Philadelphia, 2010, pp 25-32.

(27) Lee, S. H.; Meng, X. W.; Flatten, K. S.; Loegering, D. A.;

Kaufmann, S. H. Cell Death Differ. 2013, 20, 64-76.

(28) Darbre, P. D.; Harvey, P. W. J. Appl. Toxicol. 2014, 34, 925-938.

(29) Errami, Y.; Naura, A. S.; Kim, H.; Ju, J.; Suzuki, Y.; El-Bahrawy, A. H.; Ghonim, M. A.; Hemeida, R. A.; Mansy, M. S.; Zhang, J.; Xu, M.;

Smulson, M. E.; Brim, H.; Boulares, A. H. J. Biol. Chem. 2013, 288, 3460-3468.

(30) Ning, Y. B.; Du, Z. Q. Cell Biol. Int. 2015, 39, 577-583.

(31) Gong, X.; Fan, G.; Wang, W.; Wang, G. Cell. Physiol. Biochem.

2014, 34, 2245-2255.

(32) Reed, J. C. Semin. Hematol. 1997, 34, 9-19.

(33) Li, P.; Nijhawan, D.; Budihardjo, I.; Srinivasula, S. M.; Ahmad, M.; Alnemri, E. S.; Wang, X. Cell 1997, 91, 479-489.

(34) Acehan, D.; Jiang, X.; Morgan, D. G.; Heuser, J. E.; Wang, X.;

Akey, C. W. Mol. Cell. 2002, 9, 423-432.

(35) Shan, H.; Yan, R.; Diao, J.; Lin, L.; Wang, S.; Zhang, M.; Zhang, R.; Wei. J. J. Trace Elem. Med. Biol. 2015, 31, 85-91.

(36) Zhang, Z.; Teruya, K.; Eto, H.; Shirahata, S. PLoS One 2011, 6, e27441.

(37) Yang, L.; Wang, P.; Wang, H.; Li, Q.; Teng, H.; Liu, Z.; Yanq, W.;

Hou, L.; Zou, X. Mar. Drugs 2013, 11, 1961-1976.

(38) Ahn, J. H.; Yang, Y. I.; Lee, K. T.; Choi, J. H. J. Cancer Res. Clin.

Oncol. 2015, 141, 255-268.

(39) de Andrea, C. E.; Zhu, J. F.; Jin, H.; Bovée, J. V. M. G.; Jones, K.

B. J. Pathol. 2015, 236, 210-218.

(40) Al-Sheddi, E. S.; Al-Oqail, M. M.; Saquib, Q.; Siddiqui, M. A.;

Musarrat, J.; Al-Khedhairy. A. A.; Farshori, N. N. Molecules 2015, 20, 8181-8197.

(41) Nagappan, A.; Park, K. I.; Park, H. S.; Kim, J. A.; Hong, G. E.;

Kang, S. R.; Lee, D. H.; Kim, E. H.; Lee, W. S.; Won, C. K.; Kim, G. S.

Food Chem. 2012, 135, 1920-1928.

(42) Trebinska, A.; Högstrand, K.; Grandien, A.; Grzybowska, E. A.;

Fadeel, B. FEBS Lett. 2014, 588, 2921-2927.

(43) Kon, A.; Yuan, B.; Hanazawa, T.; Kikuchi, H.; Sato, M.; Furutani, R.; Takagi, N.; Toyoda, H. Oncol. Rep. 2013, 30, 1965-1970.

(44) Sun, L.; Fan, H.; Yang, L.; Shi, L.; Liu, Y. Molecules 2015, 20, 3758-3775.

(45) Liu, X.; Li, W.; Geng, S.; Meng, Q. G.; Bi, Z. G. Mol. Med. Rep.

2015, 12, 1183-1188.

Received March 05, 2020 Revised June 29, 2020 Accepted July 20, 2020

참조

관련 문서

Thus, the main purposes of this study were to investigate whether lovastatin induces apoptotic cell death in HepG2 human hepatoma cells and to elucidate the mechanism

The aim of the present study was to assess the role of tumor necrosis factor-producing MSC on apoptosis of B-CLL cells resistant to fludarabine-based chemotherapy, and showed

Anthocyanins have been shown to induce apoptosis in different cancer cells via a mitochondrial pathway (Banjerdpongchai et al., 2013) and human leukemia U937 cells via

RNases like RNase L shows antiviral and antitumour activities against virus infected cells and cancer cells through 2’-5’ oligo adenylate pathway and

It was found that the snake venom toxin from Vipera lebentina turnica induces apoptotic cell death of ovarian cancer cells through the inhibition of NF-kB and

The present study demonstrated the role of TRPM7 in PCa and TRPM7 inhibitors could enhance the apoptosis of PC-3 cells induced by TRAIL might be mediated by the

DUSP23 specifically dephosphorylates and inhibits ERK, and NSC-87877 inhibits the action of DUSP23 on ERK (A) Active recombinant MAPKs (p38, ERK, or JNK) were incubated in vitro

12) Maestu I, Gómez-Aldaraví L, Torregrosa MD, Camps C, Llorca C, Bosch C, Gómez J, Giner V, Oltra A, Albert A. Gemcitabine and low dose carboplatin in the treatment of