Next Article in Journal
Analysis of Sensory Properties in Foods: A Special Issue
Next Article in Special Issue
Elderberry (Sambucus nigra L.) Fruit Extract Alleviates Oxidative Stress, Insulin Resistance, and Inflammation in Hypertrophied 3T3-L1 Adipocytes and Activated RAW 264.7 Macrophages
Previous Article in Journal
Patagonian Berries: Healthy Potential and the Path to Becoming Functional Foods
Previous Article in Special Issue
Anti-Obesity Effect of Extract from Nelumbo Nucifera L., Morus Alba L., and Raphanus Sativus Mixture in 3T3-L1 Adipocytes and C57BL/6J Obese Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Fermented Sea Tangle (Laminaria japonica Aresch) Suppresses RANKL-Induced Osteoclastogenesis by Scavenging ROS in RAW 264.7 Cells

1
Nakdonggang National Institute of Biological Resources, Sangju 37242, Korea
2
Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Korea
3
Anti-Aging Research Center, Dong-eui University, Busan 47227, Korea
4
Department of Marine Life Sciences, Jeju National University, Jeju 63243, Korea
5
Department of Molecular Biology, College of Natural Sciences, Dong-eui University, Busan 47340, Korea
6
Marine Bioprocess Co. Ltd., Busan 46048, Korea
7
Department of Oral Pathology and Regenerative Medicine, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
8
Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63243, Korea
*
Author to whom correspondence should be addressed.
Submission received: 2 July 2019 / Revised: 23 July 2019 / Accepted: 24 July 2019 / Published: 26 July 2019

Abstract

:
Sea tangle (Laminaria japonica Aresch), a brown alga, has been used for many years as a functional food ingredient in the Asia-Pacific region. In the present study, we investigated the effects of fermented sea tangle extract (FST) on receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-stimulated osteoclast differentiation, using RAW 264.7 mouse macrophage cells. FST was found to inhibit the RANKL-stimulated activation of tartrate-resistance acid phosphatase (TRAP) and F-actin ring structure formation. FST also down-regulated the expression of osteoclast marker genes like TRAP, matrix metalloproteinase-9, cathepsin K and osteoclast-associated receptor by blocking RANKL-induced activation of NF-κB and expression of nuclear factor of activated T cells c1 (NFATc1), a master transcription factor. In addition, FST significantly abolished RANKL-induced generation of reactive oxygen species (ROS) by activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and its transcriptional targets. Hence, it seems likely that FST may have anti-osteoclastogenic potential as a result of its ability to inactivate the NF-κB-mediated NFATc1 signaling pathway and by reducing ROS production through activation of the Nrf2 pathway. Although further studies are needed to inquire its efficacy in vivo, FST appears to have potential use as an adjunctive or as a prophylactic treatment for osteoclastic bone disease.

1. Introduction

Bone remodeling is an active physiological process involving bone deposition and bone resorption by osteoblast and osteoclast, respectively. Imbalance of these processes in favor of resorption may lead to the formation of osteolytic lesions and an increase in bone disease-related disorders and morbidity [1,2,3]. Receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) are cytokines that play important roles in osteoclast differentiation and maturation. RANKL belongs to the tumor necrosis factor (TNF) superfamily and is regarded as the key promoter of osteoclastogenesis. M-CSF by contrast, is involved in the maintenance of mature osteoclast survival and mobility [4,5]. The binding of RANKL to its receptor RANK results in the activation of various signaling pathways, including the NF-κB pathway [6,7], which then enhances the activation of nuclear factor of activated T cell c1 (NFATc1), which then in turn promotes osteoclast formation by up-regulating the expression of osteoclast-specific genes [8,9]. In addition, a number of previous studies have shown that reactive oxygen species (ROS) are also critical messengers for osteoclast differentiation [10,11] and increased activity of the Nrf2 signaling system can block this activation [12,13,14]. These findings suggest that suppression of ROS production in combination with increasing activity of Nrf2 may provide a means to block osteoclast activity. Although various drugs have been used clinically to inhibit bone resorption, all have severe side effects when used long-term [15] and as a result, research into the prevention and treatment of osteolytic diseases using natural products has greatly increased in recent years.
Many marine algae extract or components of these extracts have been shown to exhibit potential for preventing and treating bone resorption related diseases [16,17] and fermented marine algal extracts have attracted the attention of the food and medical care industries [17,18]. The sea tangle, Laminaria japonica Aresch, is one of the most well-known edible brown seaweeds and has long been used as an important food supplement in Pacific and Asian countries [19]. This seaweed is rich in polysaccharides, dietary fiber, minerals, carbohydrates, polyphenols and proteins [20,21] and has been reported to protect against obesity, inflammation and cancer [22,23,24,25]. Interestingly, Lee et al. [26] developed a fermented form of sea tangle using Lactobacillus brevis with high antioxidant activity and showed that a fermented sea tangle extract (FST) protected against liver damage better than a non-fermented sea tangle extract [27,28]. They speculated that glutamate in the sea tangle which converted to gamma-aminobutyric acid through the fermentation process, was the reason behind the increased antioxidant capacity. It has been reported that FST supplementation reduce obesity and improve stress management [29]. Furthermore, previous studies have shown that FST can protect against age-associated short-term memory loss and reduced physical functioning [30,31,32]. However, the effect of FST on bone has not previously been investigated and therefore we decided to investigate whether FST had any inhibitory effect on RANKL-stimulated osteoclast differentiation using RAW 264.7 mouse macrophage cells.

2. Materials and Methods

2.1. Reagents and Antibodies

Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS) and other reagents for cell culture were purchased from WelGENE Inc. (Daegu, Republic of Korea). RANKL and osteoprotegerin (OPG) were obtained from Abcam (Cambridge, MA, USA) and Peprotech (Rocky Hill, NJ, USA), respectively. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), tartrate-resistant acid phosphatase (TRAP) assay kit, bovine serum albumin (BSA), 4′,6-diamidino-2-phenylindole (DAPI), 5,6-carboxy-2′,7′-dichlorofluorescein diacetate (DCF-DA) and N-acetyl cysteine (NAC) were purchased from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA). NE-PERTM nuclear and cytoplasmic extraction reagents kit and polyvinylidene difluoride (PVDF) membranes were obtained from Pierce Biotechnology (Rockford, IL, USA) and Schleicher & Schuell (Keene, NH, USA), respectively. Fluorescein isothiocyanate (FITC)-phalloidin solution was purchased from Thermo Fisher Scientific (Waltham, MA, USA). Primary and secondary antibodies were obtained from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA), Cell Signaling Technology Inc. (Beverly, MA, USA), Abcam, Novus (Novus Biologicals, LLC., Littleton, CO, USA), Thermo Fisher Scientific and R&D system. Appropriate horseradish-peroxidase (HRP)-linked secondary antibodies and enhanced chemiluminescence (ECL) detection solution were purchased from Amersham Corp. (Arlington Heights, IL, USA). All reagents not specifically identified were purchased from Sigma-Aldrich Chemical Co. (St. Louis, MO, USA).

2.2. Preparation of FST

FST received from Marine Bioprocess Co. Ltd. (Busan, Korea) was extracted as previously described [30]. In brief, yeast extract and glucose were added to water at a ratio of 1:15 (w/v) and sea tangle (L. japonica Aresch) was then added and sterilized in an autoclave at 121 °C for 30 min. After autoclaving, culture broth of L. brevis BJ20 (accession no. KCTC 11377BP) was added to the mix at a concentration of 1.2% (v/v) and the mixture was incubated at 37 °C for 2 days. The fermented product was obtained by filtration and lyophilized. The dried extract (FST) so obtained was dissolved in Milli-Q Water to produce a 10 mg/mL stock solution.

2.3. Cell Culture and Viability Analysis

RAW 264.7 cell line was purchased from the American Type Culture Collection (Manassas, VA, USA). The cells were cultured in DMEM containing 10% heat inactivated FBS, penicillin (100 units/mL) and streptomycin (100 g/mL) at 37 °C in a humidified 5% CO2 atmosphere and subcultured every 3 days. The viability of the cells was assessed by MTT assay as previously described [14]. Briefly, the cells were treated with the desired concentrations of FST with or without 100 ng/mL RANKL for 72 h and then incubated with 50 μg/mL MTT solution for 3 h. Formazan crystals were dissolved in DMSO and the absorbance was measured using an enzyme-linked immunosorbent assay (ELISA) microplate reader (Dynatech Laboratories, Chantilly, VA, USA) at 540 nm.

2.4. Osteoclast Differentiation and TRAP Assay

Osteoclast formation was measured by quantifying cells positively stained by TRAP. Briefly, the cells were fixed in 4% paraformaldehyde (pH 7.4) at room temperature for 10 min and then stained with commercial TRAP staining kit according to the manufacturer’s instructions. Osteoclasts were defined as TRAP-positive multinuclear cells containing 3 or more nuclei, under a phase-contrast microscope (Carl Zeiss, Oberkochen, Germany). TRAP activity was determined in culture media using a TRAP assay kit, in accordance with the manufacturer’s instructions. TRAP activities were expressed as percentages of control activities.

2.5. F-Actin Ring Staining

As described previously, evaluation of actin ring formation was performed [14]. Briefly, the cells were fixed with 4% paraformaldehyde, permeabilized with 0.1% Triton X-100 in PBS for 5 min and then stained with an anti-actin antibody at 4 °C overnight. After washing with PBS, the cells were incubated with FITC-conjugated phalloidin for 30 min at 37 °C and then counterstained with 2.5 μg/mL DAPI for 20 min. F-actin rings were analyzed by fluorescence microscopy (Carl Zeiss, Oberkochen, Germany).

2.6. Western Blot Analysis

As described previously, total protein was extracted from the cells using the Bradford Protein assay kit [14]. Nuclear and cytosolic proteins were prepared using a NE-PER nuclear and cytoplasmic extraction reagents kit according to the manufacturer’s instructions. Equal amounts of protein from samples were loaded and separated by sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis and transferred onto PVDF membranes. The membranes were blocked with 5% non-fat skim milk in trisbuffered saline containing 0.1% Triton X-100 (TBST) for 1 h and probed with specific primary antibodies at 4 °C overnight (Table 1). After washing three times with TBST, the membranes were incubated with the appropriate HRP-conjugated secondary antibodies for 2 h. Protein expression was detected by an ECL kit and visualized by Fusion FX Image system (Vilber Lourmat, Torcy, France).

2.7. Immunofluorescence Staining for NF-κB

RAW 264.7 cells were seeded on gelatin-coated glass coverslips. After it was cultured for 24 h, cells were treated with RANKL in the presence or absence of various concentrations of FST for 24 h, fixed in 4% paraformaldehyde for 15 min, permeabilized with 0.2% Triton X-100 in PBS for 15 min and blocked with PBS containing 5% BSA. Cells were stained with primary antibody against phosphoNF-κ B p65 at 4 °C overnight and incubated with a fluorescein-conjugated anti-rat IgG in the dark at 37 °C for 1 h. Cells were mounted on slides and then analyzed by fluorescence microscope.

2.8. Measurement of Intracellular ROS Levels

The production of intracellular ROS was measured by a flow cytometer with DCF-DA as described previously [14]. Briefly, the cells were treated with FST in the presence or absence of 100 ng/mL RANKL. In the last 20 min of treatment, 10 μM DCF-DA was added to the incubated cells in the dark. Following incubation, the cells were washed twice with PBS and 10,000 cells were analyzed for intracellular ROS content by BD Accuri C6 software in a flow cytometer (BD Biosciences) at 480/520 nm. To observe ROS generation by fluorescence microscopy, cells were stimulated with RANKL in the presence or absence of FST for 1 h. Cells were then stained with DCF-DA and then fixed with 4% paraformaldehyde for 2.

2.9. Statistical Analysis

All experiments were performed at least three times. Data were analyzed using GraphPad Prism software (version 5.03; GraphPad Software, Inc., La Jolla, CA, USA) and expressed as the mean ± standard deviation (SD). Differences between groups were assessed using analysis of variance followed by ANOVA-Tukey’s post hoc test and p < 0.05 was considered to indicate a statistically significant difference.

3. Results

3.1. Effect of FST on Cell Viability in RAW 264.7 Cells

RAW 264.7 cells were treated with various concentrations of FST for 72 h and then 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed. Figure 1A shows that FST had no cytotoxicity on the cells at concentrations up to 800 μg/mL but relatively cytotoxic effect was observed in the 1000 μg/mL treatment group as compared with untreated controls. In the presence of 100 ng/mL RANKL or 100 ng/mL osteoprotegerin (OPG), a decoy receptor for RANKL that inhibits osteoclastogenesis [33,34], cell viability was not significantly reduced by FST at concentrations up to 800 ng/mL compared to that of control groups (Figure 1B). Hence, non-toxic concentrations (<800 μg/mL) were used to investigate the effect of FST on RANKL-induced osteoclast differentiation.

3.2. FST Suppresses RANKL-Induced Osteoclastogenesis in RAW 264.7 Cells

In order to examine the effect of FST on RANKL-induced osteoclastogenesis, RAW 264.7 cells were treated with FST in the presence of different concentrations of RANKL. As shown in Figure 2A, FST treatment markedly inhibited RANKL-induced osteoclast-like morphological changes. TRAP staining demonstrated that FST suppressed cell fusion and the conversion of RAW 264.7 cells into osteoclasts (Figure 2B FST suppressed numbers of TRAP-positive osteoclasts as compared with RANKL treated cells, dose-dependent manner (Figure 2C). These reductions in TRAP-positive osteoclast number were paralleled by the inhibition of TRAP activity (Figure 2D). As expected, RANKL-induced osteoclast differentiation and TRAP activity were completely suppressed in the presence of OPG.

3.3. FST Disrupts RANKL-Induced Formation of F-Actin Rich Adhesive Structures in RAW 264.7 Mouse Macrophage-Like Cells

Formation of the F-actin rich adhesive structures by osteoclasts is an essential step in bone resorption [35,36]. Figure 3 indicated that staining with FITC-conjugated phalloidin showed RANKL (100 ng/mL) stimulation increased well-defined F-actin sealing rings with a higher intensity ring height. However, the size of rings formed by RANKL-treated cells was remarkably and concentration-dependently reduced in cells co-treated with FST. Furthermore, OPG treatment complementally inhibited the F-actin sealing ring formation in RANKL-stimulated cells.

3.4. FST Inhibits the RANKL-Induced Nuclear Translocation of NF-κB and IκBα Degradation in RAW 264.7 Cells

Activation of NF-κB through nuclear translocation by RANKL is an essential step for initiation of osteoclast differentiation [6,7]. Therefore, we assessed whether FST affected the activation of NF-κB induced by RANKL. As shown in Figure 4A,B, our immunoblotting results reveal that the expression of NF-κB was markedly increased in the nuclei of RANKL treated cells but the expression of IκBα was reduced in the cytoplasm, which suggested that RANKL stimulated activation of NF-κB. However, FST suppressed the RANKL-mediated degradation of IκBα and the subsequent nuclear accumulation of NF-κB. Furthermore, immunofluorescence studies produced similar results. More specifically, phosphorylated NF-κB p65 was predominantly located in nuclei in RANKL-stimulated cells but not in FST and RANKL co-treated cells (Figure 4C).

3.5. FST Down-Regulates RANKL-Induced Osteoclast-Associated Gene Expression in RAW 264.7 Cells

NFATc1 is considered to be the most important regulator of the transcriptional activation of osteoclast differentiation-associated genes by RANKL [8,9]. To examine in more detail the mechanism of FST-mediated inhibition of osteoclastogenesis, we assessed the expression of NFATc1 in RANKL-stimulated RAW 264.7 cells. Consistent with previous studies, the expression of NFATc1 was significantly increased by RANKL but was down-regulated in a concentration-dependent manner by FST (Figure 5). In addition, we investigated the effects of FST on the levels of specific marker for osteoclast such as TRAP, cathepsin (CTSK), matrix metallopeptidase-9 (MMP-9) and osteoclast-associated receptor (OSCAR). Figure 5 showed that RANKL markedly up-regulated levels of these osteoclast-specific markers, which were effectively attenuated by the addition of FST. Co-treatment with OPG also completely prevented increases in these protein markers.

3.6. FST Attenuates RANKL-Induced Intracellular ROS Accumulation Associated with Activation of Nrf2 in RAW 264.7 Mouse Macrophage-Like Cells

Overproduction of intracellular ROS plays a critical step in RANKL-mediated osteoclastogenesis [12,13,14], thereby we examined whether FST inhibits the generation of ROS during RANKL-mediated osteoclastogenesis using DCF-DA, a cell permeant redox-sensitive dye. We demonstrated by flow cytometry that ROS levels were significantly increased by RANKL and that these up-regulation were abolished by FST (Figure 6A,D). Moreover, this effect of FST was supported by our fluorescence microscopic examination (Figure 6B) and further, co-treatment with N-acetyl cysteine (NAC), an intensive ROS scavenger, completely alleviated RANKL-induced ROS generation and F-actin ring formation (Figure 6C). In addition, Figure 6E,F shows that FST has the efficacy of equivalence and/or superiority compared with NAC and it was suggested that FST is a powerful anti-oxidant, thereby it has a suppressed RANKL-mediated ROS generation.
In addition, we show that FST increased the expression and phosphorylation of Nrf2 in RANKL-stimulated cells, which was associated with an increase in typical Nrf2-dependent cytoprotective enzymes such as heme oxygenase-1 (HO-1) and NAD(P)H: Quinone oxidoreductase 1 (NQO-1) (Figure 7A,B). Furthermore, we observed that Nrf2 translocation to the nucleus was promoted by FST treatment (Figure 7C,D).

4. Discussion

Osteoclasts are multinucleated cells of hematopoietic origin which are derived from the monocyte/macrophage in their ability to resorb bone, whereas osteoblast are derived from pluripotent mesenchymal stem cells and are involved in bone formation [1,2]. Since excessive bone resorption by osteoclasts causes an imbalance in bone regeneration and induces osteolytic diseases, osteoclasts are considered prime targets for the management and treatment of bone diseases [2,3]. RANKL is a pro-osteoclastogenic cytokine and plays a crucial role in promoting osteoclastogenesis from osteoclast progenitor cells [4,5]. As has been well established in many earlier studies, RANKL binds to RANK expressed on the plasma membrane of osteoclast precursors and activates complex signaling cascades including NF-κB and NFATc1 for osteoclast differentiation [9,10]. Differentiation through activation of these signal transduction systems by RANKL is characterized by the formation of multinucleated giant cells [5,7]. This is a preliminary step in the maintenance, formation and function of the F-actin loop structure, which plays an important role in seal zone formation and resorption of bone mineral matrix in osteoclasts by activated TRAP [37,38]. According to the present findings, FST effectively inhibited RANKL-induced TRAP activation and F-actin ring formation without causing any significant cytotoxicity in RAW 264.7 cells, implying that FST suppressed osteoclast differentiation from osteoclast precursors at an early stage.
NF-κB, a transcription factor that plays a key role in inducing osteoclast differentiation, complexes with cytoplasmic IκB-α in the absence of osteoclastogenic induction signals and keeps it in an inactive form that tightly regulates its transcriptional activity for osteoclast differentiation [4,37]. However, the interaction of RANKL and RANK promotes the activation of the IκB kinase (IKK) complex, which phosphorylates IκB-α leading to ubiquitin-dependent degradation [6,7]. As a result, free NF-κB translocates to the nucleus and activates transcription of various genes involved in osteoclastogenesis [5]. Our results demonstrated that RANKL promoted the degradation of IκB-α in the cytoplasm and induced the translocation of NF-κB into the nucleus, both of which are essential for the activation of NF-κB but that these changes were completely inhibited by FST.
In the early stages of NF-κB activation and osteoclast differentiation, NFATc1 acts as a master regulator that enhances transcription of various osteoclast marker genes, which are highly expressed in the terminal differentiation stage to promote bone resorption [5,39]. In addition to blocking RANKL-induced NF-κB activation, FST inhibited NFATc1 expression in RANKL-treated cells. OPG treatment also completely blocked the expression of NFATc1. As further proof that FST was effectively inhibiting osteoclastogenesis, we showed that it attenuated RANKL-induced up-regulation of osteoclast marker genes such as TRAP, MMP-9, CTSK and OSCAR to levels seen in the control and OPG co-treated groups. Although further experiments are required to determine whether NFATc1 inhibition is the direct result of NF-κB inactivation, the present results indicate that inactivation of the NF-κB signaling pathway and inhibition of the expression of osteoclast marker genes associated with a decrease in NFATc1 expression are involved as important mechanisms in the anti-osteoclastogenic effect of FST.
A number of previous studies have shown that ROS, as specific secondary messengers, play a key role in the initiation of RANKL-stimulated osteoclast differentiation and bone resorption through similar pathways involving the activation of NF-κB and NFATc1 [10,12]. However, the accumulation of excessive ROS due to oxidative stress blocks osteoblast differentiation, suppresses osteoblast survival and acts to promote bone loss [12,13]. It has also been reported that a variety of natural products with antioxidant activity inhibit osteoclast differentiation by inhibiting ROS production [6,10,11]. Therefore, ROS can be considered a potential target for inhibition of osteoclast differentiation and prevention of bone loss. The present results showed that FST significantly suppressed ROS production by RANKL. Moreover, consistent with the results of previous studies [12,40], RANKL-induced osteoclast differentiation was completely inhibited when production of ROS was artificially blocked using NAC, indicating that FST blocks osteoclast differentiation by acting as a scavenger or inhibitor of ROS. In order to reduce the damage from oxidative stress in the face of excess production of ROS in cells, several transcription factors are known to be activated to increase the expression of downstream antioxidant enzymes [41,42]. One of these redox sensitive transcription factors, Nrf2 has recently been reported to attenuate osteoclast differentiation through the regulation of ROS production [42,43]. For example, Nrf2 deficiency improved RANKL-induced osteoclast differentiation [44], whereas local induction of nuclear Nrf2 weakened RANKL-mediated osteoclastogenesis [45]. Under normal conditions, Nrf2 is sequestered by Kelch-like ECH-associated protein 1 (Keap1) to the cytoplasm but becomes separated from Keap1 by oxidative or electrophilic stress and translocated into the nucleus. In the nucleus, Nrf2 binds to the antioxidant response elements to induce the transcription of target antioxidants and detoxifying enzymes including HO-1 and NQO-1 [42,43]. In this study, FST significantly increased expression of Nrf2 and its transcriptional targets, including HO-1 and NQO-1 in RANKL-treated RAW 264.7 cells. We also observed that FST increased phosphorylation and nuclear translocation of Nrf2 compared to the RANKL-alone stimulated group. The results presented, indicate that FST attenuates osteoclast differentiation by decreasing RANKL-induced oxidative stress in osteoclast precursor cells through the activation of Nrf2 and its downstream genes.

5. Conclusions

To assume the effect of FST on RANKL-mediated osteoclast differentiation, recombinant RANKL protein was used to differentiate murine monocyte/macrophage RAW 264.7 cells as osteoclast precursor cells into osteoclasts. Present results demonstrated that FST inhibited RANKL-induced osteoclastogenesis and reduced the expression of several key osteoclast-regulatory genes through the inactivation of NF-κB. In addition, FST blocked RANKL-induced oxidative stress, which was associated with the activation of Nrf2 signaling pathway. Although the present study provides new insights into the inhibition of osteoclastogenesis by FST, further investigation of the molecular mechanisms underlying this process as well as identification of the bioactive constituents of FST are needed.

Author Contributions

Y.H.C., Y.-J.J., C.P. and B.-J.L. conceived and designed the experiments; J.-W.J., H.L., S.H.H. and C.P. performed the experiments; S.Y.J., H.L., G.-Y.K., E.K.P. and J.W.H. analyzed the data; J.-W.J. wrote the paper and Y.H.C. edited the paper.

Funding

This research was a part of the project titled ‘Development of functional food products with natural materials derived from marine resources (20170285)’, funded by the Ministry of Oceans and Fisheries.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kikuta, J.; Ishii, M. Osteoclast migration, differentiation and function: Novel therapeutic targets for rheumatic diseases. Rheumatology 2013, 52, 226–234. [Google Scholar] [CrossRef] [PubMed]
  2. Teitelbaum, S.L.; Ross, F.P. Genetic regulation of osteoclast development and function. Nat. Rev. Genet. 2003, 4, 638–649. [Google Scholar] [CrossRef] [PubMed]
  3. Galson, D.L.; Roodman, G.D. Pathobiology of Paget’s disease of bone. J. Bone Metab. 2014, 21, 85–98. [Google Scholar] [CrossRef] [PubMed]
  4. Ono, T.; Nakashima, T. Recent advances in osteoclast biology. Histochem. Cell Biol. 2018, 149, 325–341. [Google Scholar] [CrossRef] [PubMed]
  5. Park, J.H.; Lee, N.K.; Lee, S.Y. Current understanding of RANK signaling in osteoclast differentiation and maturation. Mol. Cells 2017, 40, 706–713. [Google Scholar] [PubMed]
  6. Bi, H.; Chen, X.; Gao, S.; Yu, X.; Xiao, J.; Zhang, B.; Liu, X.; Dai, M. Key triggers of osteoclast-related diseases and available strategies for targeted therapies: A review. Front. Med. 2017, 4, 234. [Google Scholar] [CrossRef] [PubMed]
  7. Kuroda, Y.; Matsuo, K. Molecular mechanisms of triggering, amplifying and targeting RANK signaling in osteoclasts. World J. Orthop. 2012, 3, 167–174. [Google Scholar] [CrossRef]
  8. Sundaram, K.; Nishimura, R.; Senn, J.; Youssef, R.F.; London, S.D.; Reddy, S.V. RANK ligand signaling modulates the matrix metalloproteinase-9 gene expression during osteoclast differentiation. Exp. Cell Res. 2007, 313, 168–178. [Google Scholar] [CrossRef]
  9. Asagiri, M.; Sato, K.; Usami, T.; Ochi, S.; Nishina, H.; Yoshida, H.; Morita, I.; Wagner, E.F.; Mak, T.W.; Serfling, E.; et al. Autoamplification of NFATc1 expression determines its essential role in bone homeostasis. J. Exp. Med. 2005, 202, 1261–1269. [Google Scholar] [CrossRef] [Green Version]
  10. Lee, S.H.; Jang, H.D. Scoparone attenuates RANKL-induced osteoclastic differentiation through controlling reactive oxygen species production and scavenging. Exp. Cell Res. 2015, 331, 267–277. [Google Scholar] [CrossRef]
  11. Domazetovic, V.; Marcucci, G.; Iantomasi, T.; Brandi, M.L.; Vincenzini, M.T. Oxidative stress in bone remodeling: Role of antioxidants. Clin. Cases Miner. Bone Metab. 2017, 14, 209–216. [Google Scholar] [CrossRef] [PubMed]
  12. Li, Z.; Chen, C.; Zhu, X.; Li, Y.; Yu, R.; Xu, W. Glycyrrhizin suppresses RANKL-Induced osteoclastogenesis and oxidative stress through inhibiting NF-κB and MAPK and activating AMPK/Nrf2. Calcif. Tissue Int. 2018, 103, 324–337. [Google Scholar] [CrossRef] [PubMed]
  13. Thummuri, D.; Naidu, V.G.M.; Chaudhari, P. Carnosic acid attenuates RANKL-induced oxidative stress and osteoclastogenesis via induction of Nrf2 and suppression of NF-κB and MAPK signalling. J. Mol. Med. 2017, 95, 1065–1076. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, H.J.; Park, C.; Kim, G.Y.; Park, E.K.; Jeon, Y.J.; Kim, S.; Hwang, H.J.; Choi, Y.H. Sargassum serratifolium attenuates RANKL-induced osteoclast differentiation and oxidative stress through inhibition of NF-κB and activation of the Nrf2/HO-1 signaling pathway. Biosci. Trends 2018, 12, 257–265. [Google Scholar] [CrossRef] [PubMed]
  15. Khan, M.; Cheung, A.M.; Khan, A.A. Drug-related adverse events of osteoporosis therapy. Endocrinol. Metab. Clin. 2017, 46, 181–192. [Google Scholar] [CrossRef] [PubMed]
  16. Koyama, T. Extracts of marine algae show inhibitory activity against osteoclast differentiation. Adv. Food Nutr. Res. 2011, 64, 443–454. [Google Scholar] [PubMed]
  17. Venkatesan, J.; Kim, S.K. Osteoporosis treatment: Marine algal compounds. Adv. Food Nutr. Res. 2011, 64, 417–427. [Google Scholar] [PubMed]
  18. De Jesus Raposo, M.F.; De Morais, A.M.; De Morais, R.M. Emergent sources of prebiotics: Seaweeds and microalgae. Mar. Drugs 2016, 14, E27. [Google Scholar] [CrossRef] [PubMed]
  19. Shirosaki, M.; Koyama, T. Laminaria japonica as a food for the prevention of obesity and diabetes. Adv. Food Nutr. Res. 2011, 64, 199–212. [Google Scholar]
  20. Gao, J.; Lin, L.; Sun, B.; Zhao, M. A comparison study on polysaccharides extracted from Laminaria japonica using different methods: Structural characterization and bile acid-binding capacity. Food Funct. 2017, 8, 3043–3052. [Google Scholar] [CrossRef]
  21. Machu, L.; Misurcova, L.; Ambrozova, J.V.; Orsavova, J.; Mlcek, J.; Sochor, J.; Jurikova, T. Phenolic content and antioxidant capacity in algal food products. Molecules 2015, 20, 1118–1133. [Google Scholar] [CrossRef] [PubMed]
  22. Kim, Y.M.; Jang, M.S. Anti-obesity effects of Laminaria japonica fermentation on 3T3-L1 adipocytes are mediated by the inhibition of C/EBP-α/β and PPAR-γ. Cell. Mol. Biol. 2018, 64, 71–77. [Google Scholar] [CrossRef] [PubMed]
  23. Jang, E.J.; Kim, S.C.; Lee, J.H.; Lee, J.R.; Kim, I.K.; Baek, S.Y.; Kim, Y.W. Fucoxanthin, the constituent of Laminaria japonica, triggers AMPK-mediated cytoprotection and autophagy in hepatocytes under oxidative stress. BMC Complement. Altern. Med. 2018, 18, 97. [Google Scholar] [CrossRef] [PubMed]
  24. Je, J.Y.; Park, S.Y.; Ahn, C.B. Antioxidant and cytoprotective activities of enzymatic extracts from Rhizoid of Laminaria japonica. Prev. Nutr. Food Sci. 2017, 22, 312–319. [Google Scholar] [CrossRef] [PubMed]
  25. Zeng, M.; Wu, X.; Li, F.; She, W.; Zhou, L.; Pi, B.; Xu, Z.; Huang, X. Laminaria Japonica polysaccharides effectively inhibited the growth of nasopharyngeal carcinoma cells in vivo and in vitro study. Exp. Toxicol. Pathol. 2017, 69, 527–532. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, B.J.; Senevirathne, M.; Kim, J.S.; Kim, Y.M.; Lee, M.S.; Jeong, M.H.; Kang, Y.M.; Kim, J.I.; Nam, B.H.; Ahn, C.B.; et al. Protective effect of fermented sea tangle against ethanol and carbon tetrachloride-induced hepatic damage in Sprague-Dawley rats. Food Chem. Toxicol. 2010, 48, 1123–1128. [Google Scholar] [CrossRef] [PubMed]
  27. Kang, Y.M.; Lee, B.J.; Kim, J.I.; Nam, B.H.; Cha, J.Y.; Kim, Y.M.; Ahn, C.B.; Choi, J.S.; Choi, I.S.; Je, J.Y. Antioxidant effects of fermented sea tangle (Laminaria japonica) by Lactobacillus brevis BJ20 in individuals with high level of γ-GT: A randomized, double-blind, and placebo-controlled clinical study. Food Chem. Toxicol. 2012, 50, 1166–1169. [Google Scholar] [CrossRef]
  28. Cha, J.Y.; Jeong, J.J.; Yang, H.J.; Lee, B.J.; Cho, Y.S. Effect of fermented sea tangle on the alcohol dehydrogenase and acetaldehyde dehydrogenase in Saccharomyces cerevisiae. J. Microbiol. Biotechnol. 2011, 21, 791–795. [Google Scholar] [CrossRef]
  29. You, J.S.; Sung, M.J.; Chang, K.J. Evaluation of 8-week body weight control program including sea tangle (Laminaria japonica) supplementation in Korean female college students. Nutr. Res. Pract. 2009, 3, 307–314. [Google Scholar] [CrossRef]
  30. Choi, W.C.; Reid, S.N.S.; Ryu, J.K.; Kim, Y.; Jo, Y.H.; Jeon, B.H. Effects of γ-aminobutyric acid-enriched fermented sea tangle (Laminaria japonica) on brain derived neurotrophic factor-related muscle growth and lipolysis in middle aged women. Algae 2016, 31, 175–187. [Google Scholar] [CrossRef]
  31. Reid, S.N.S.; Ryu, J.K.; Kim, Y.; Jeon, B.H. GABA-enriched fermented Laminaria japonica improves cognitive impairment and neuroplasticity in scopolamine- and ethanol-induced dementia model mice. Nutr. Res. Pract. 2018, 12, 199–207. [Google Scholar] [CrossRef] [PubMed]
  32. Reid, S.N.S.; Ryu, J.K.; Kim, Y.; Jeon, B.H. The effects of fermented Laminaria japonica on short-term working memory and physical fitness in the elderly. Evid. Based Complement. Altern. Med. 2018, 2018, 8109621. [Google Scholar]
  33. Khosla, S. Minireview: The OPG/RANKL/RANK system. Endocrinology 2001, 142, 5050–5055. [Google Scholar] [CrossRef] [PubMed]
  34. Hofbauer, L.C. Osteoprotegerin ligand and osteoprotegerin: Novel implications for osteoclast biology and bone metabolism. Eur. J. Endocrinol. 1999, 141, 195–210. [Google Scholar] [CrossRef] [PubMed]
  35. Kong, X.; Yang, Y.; Wu, W.; Wan, H.; Li, X.; Zhong, M.; Su, X.; Jia, S.; Lin, N. Triterpenoid saponin W3 from Anemone flaccida suppresses osteoclast differentiation through inhibiting activation of MAPKs and NF-κB pathways. Int. J. Biol. Sci. 2015, 11, 1204–1214. [Google Scholar] [CrossRef]
  36. Hong, S.; Huh, J.E.; Lee, S.Y.; Shim, J.K.; Rhee, S.G.; Jeong, W. TRP14 inhibits osteoclast differentiation via its catalytic activity. Mol. Cell. Biol. 2014, 34, 3515–3524. [Google Scholar] [CrossRef] [PubMed]
  37. Soysa, N.S.; Alles, N. Osteoclast function and bone-resorbing activity: An overview. Biochem. Biophys. Res. Commun. 2016, 476, 115–120. [Google Scholar] [CrossRef]
  38. Hayman, A.R. Tartrate-resistant acid phosphatase (TRAP) and the osteoclast/immune cell dichotomy. Autoimmunity 2008, 41, 218–223. [Google Scholar] [CrossRef]
  39. Kim, Y.W.; Baek, S.H.; Lee, S.H.; Kim, T.H.; Kim, S.Y. Fucoidan, a sulfated polysaccharide, inhibits osteoclast differentiation and function by modulating RANKL signaling. Int. J. Mol. Sci. 2014, 15, 18840–18855. [Google Scholar] [CrossRef]
  40. Zhang, Y.; Zhang, Y.; Kou, J.; Wang, C.; Wang, K. Role of reactive oxygen species in angiotensin II: Induced receptor activator of nuclear factor-κB ligand expression in mouse osteoblastic cells. Mol. Cell. Biochem. 2014, 396, 249–255. [Google Scholar] [CrossRef]
  41. Lee, N.K.; Choi, Y.G.; Baik, J.Y.; Han, S.Y.; Jeong, D.W.; Bae, Y.S.; Kim, N.; Lee, S.Y. A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood 2005, 106, 852–859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Kang, K.A.; Hyun, J.W. Oxidative stress, Nrf2, and epigenetic modification contribute to anticancer drug resistance. Toxicol. Res. 2017, 33, 1–5. [Google Scholar] [CrossRef] [PubMed]
  43. Sun, Y.X.; Xu, A.H.; Yang, Y.; Li, J. Role of Nrf2 in bone metabolism. J. Biomed. Sci. 2015, 22, 101. [Google Scholar] [CrossRef] [PubMed]
  44. Hyeon, S.; Lee, H.; Yang, Y.; Jeong, W. Nrf2 deficiency induces oxidative stress and promotes RANKL-induced osteoclast differentiation. Free Radic. Biol. Med. 2013, 65, 789–799. [Google Scholar] [CrossRef] [PubMed]
  45. Kanzaki, H.; Shinohara, F.; Kajiya, M.; Fukaya, S.; Miyamoto, Y.; Nakamura, Y. Nuclear Nrf2 induction by protein transduction attenuates osteoclastogenesis. Free Radic. Biol. Med. 2014, 77, 239–248. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effects of fermented sea tangle extract (FST) and receptor of activator of nuclear factor kappa-B ligand (RANKL) on the viability of RAW 264.7 mouse macrophage-like cells. Cells were treated with desired concentrations of FST in the absence (A) or presence (B) of 100 ng/mL RANKL and/or 100 ng/mL OPG for 72 h. H2O2 was used as a positive control. Cell viabilities were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Relative cell viability is expressed as percentages compared to treatment of naïve control cells. Results are presented as means ± SD of three independent experiments. * p < 0.05 and *** p < 0.005 indicates significant difference compared to the untreated control cells. OPG: osteoprotegerin; +: cells treated the reagent; -: cells untreated the reagent.
Figure 1. Effects of fermented sea tangle extract (FST) and receptor of activator of nuclear factor kappa-B ligand (RANKL) on the viability of RAW 264.7 mouse macrophage-like cells. Cells were treated with desired concentrations of FST in the absence (A) or presence (B) of 100 ng/mL RANKL and/or 100 ng/mL OPG for 72 h. H2O2 was used as a positive control. Cell viabilities were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Relative cell viability is expressed as percentages compared to treatment of naïve control cells. Results are presented as means ± SD of three independent experiments. * p < 0.05 and *** p < 0.005 indicates significant difference compared to the untreated control cells. OPG: osteoprotegerin; +: cells treated the reagent; -: cells untreated the reagent.
Foods 08 00290 g001
Figure 2. Inhibition of RANKL-stimulated osteoclast differentiation by FST in RAW 264.7 mouse macrophage-like cells. Cells were stimulated with 100 ng/mL RANKL in the presence or absence of FST or 100 ng/mL OPG for 5 days. (A) Representative photographs of the morphological changes are presented. (B) Cells were fixed and stained for TRAP and examined under an inverted microscope. (C) TRAP-positive multinucleated cells were counted to determine osteoclast numbers. (D) Supernatants were collected from cells grown under the same conditions and TRAP activities were measured using an ELISA reader. Results are presented as means ± SD of three independent experiments. * p < 0.05, ** p < 0.01 and *** p < 0.001 indicates significant difference compared to RANKL-treated cells. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; OPG: osteoprotegerin; TRAP: tartrate-resistance acid phosphatase; +: cells treated the reagent; -: cells untreated the reagent.
Figure 2. Inhibition of RANKL-stimulated osteoclast differentiation by FST in RAW 264.7 mouse macrophage-like cells. Cells were stimulated with 100 ng/mL RANKL in the presence or absence of FST or 100 ng/mL OPG for 5 days. (A) Representative photographs of the morphological changes are presented. (B) Cells were fixed and stained for TRAP and examined under an inverted microscope. (C) TRAP-positive multinucleated cells were counted to determine osteoclast numbers. (D) Supernatants were collected from cells grown under the same conditions and TRAP activities were measured using an ELISA reader. Results are presented as means ± SD of three independent experiments. * p < 0.05, ** p < 0.01 and *** p < 0.001 indicates significant difference compared to RANKL-treated cells. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; OPG: osteoprotegerin; TRAP: tartrate-resistance acid phosphatase; +: cells treated the reagent; -: cells untreated the reagent.
Foods 08 00290 g002
Figure 3. Suppression of F-actin ring formation by FST in RANKL-induced RAW 264.7 mouse macrophage-like cells. The cells were co-treated with 100 ng/mL RANKL in the presence or absence of FST or 100 ng/mL OPG for 5 days and stained for F-actin rich adhesive structures with fluorescein isothiocyanate (FITC)-phalloidin and 4′,6-diamidino-2-phenylindole (DAPI). The photographs are representative of the morphological changes observed under a fluorescence microscope. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract.
Figure 3. Suppression of F-actin ring formation by FST in RANKL-induced RAW 264.7 mouse macrophage-like cells. The cells were co-treated with 100 ng/mL RANKL in the presence or absence of FST or 100 ng/mL OPG for 5 days and stained for F-actin rich adhesive structures with fluorescein isothiocyanate (FITC)-phalloidin and 4′,6-diamidino-2-phenylindole (DAPI). The photographs are representative of the morphological changes observed under a fluorescence microscope. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract.
Foods 08 00290 g003
Figure 4. Effects of FST on the RANKL-induced activation of NF-κB in RAW 264.7 mouse macrophage-like cells. (A) After co-treating cells with 100 ng/mL RANKL in the presence or absence of FST for 1 h, nuclear and cytosolic proteins were isolated. The expression of NF-κB and IκB-α were determined by Western blotting. Lamin B and β-actin were used as internal controls for the nuclear and cytosolic fractions, respectively. (B) Densitometry quantifications of protein expressions were measured by ImageJ. Statistical analyses were conducted using analysis of variances between groups. *** p < 0.0001 when compared to control. (C) Cells grown on gelatin-coated glass coverslips were co-treated with 800 μg/mL FST with or without 100 ng/mL RANKL. Localization of phospho-NF-κB p65 was observed under a fluorescence microscope following staining with anti-phospho-NF-κB p65 antibody (red) and DAPI (nuclear stain; blue). Original magnification ×400. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; IκBα: inhibitory proteins of kappa B, alpha; NF-κB: nuclear factor-kappa B; +: cells treated the reagent; -: cells untreated the reagent.
Figure 4. Effects of FST on the RANKL-induced activation of NF-κB in RAW 264.7 mouse macrophage-like cells. (A) After co-treating cells with 100 ng/mL RANKL in the presence or absence of FST for 1 h, nuclear and cytosolic proteins were isolated. The expression of NF-κB and IκB-α were determined by Western blotting. Lamin B and β-actin were used as internal controls for the nuclear and cytosolic fractions, respectively. (B) Densitometry quantifications of protein expressions were measured by ImageJ. Statistical analyses were conducted using analysis of variances between groups. *** p < 0.0001 when compared to control. (C) Cells grown on gelatin-coated glass coverslips were co-treated with 800 μg/mL FST with or without 100 ng/mL RANKL. Localization of phospho-NF-κB p65 was observed under a fluorescence microscope following staining with anti-phospho-NF-κB p65 antibody (red) and DAPI (nuclear stain; blue). Original magnification ×400. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; IκBα: inhibitory proteins of kappa B, alpha; NF-κB: nuclear factor-kappa B; +: cells treated the reagent; -: cells untreated the reagent.
Foods 08 00290 g004
Figure 5. Inhibition of the RANKL-induced expressions of osteoclast-regulatory genes by FST in RAW 264.7 mouse macrophage-like cells. Cells were co-treated with various concentrations of FST or 100 ng/mL OPG in the presence or absence of 100 ng/mL RANKL for 5 days. (A) The expression levels of osteoclast-regulatory proteins were assessed by Western blot analysis. β-actin was used as the internal control. The results shown are representative of three independent experiments. (B) Densitometry quantifications of protein expression were measured by ImageJ. Statistical analyses were conducted using analysis of variances. * p < 0.05 and *** p < 0.0001 when compared to control. ### p < 0.0001 when compared to RANKL treatment. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; OPG: osteoprotegerin; CTSK: cathepsin K; MMP-9: matrix metalloproteinase-9; NFATc1: nuclear factor of activated T cells c1; OSCAR: osteoclast-associated receptor; +: cells treated the reagent; -: cells untreated the reagent.
Figure 5. Inhibition of the RANKL-induced expressions of osteoclast-regulatory genes by FST in RAW 264.7 mouse macrophage-like cells. Cells were co-treated with various concentrations of FST or 100 ng/mL OPG in the presence or absence of 100 ng/mL RANKL for 5 days. (A) The expression levels of osteoclast-regulatory proteins were assessed by Western blot analysis. β-actin was used as the internal control. The results shown are representative of three independent experiments. (B) Densitometry quantifications of protein expression were measured by ImageJ. Statistical analyses were conducted using analysis of variances. * p < 0.05 and *** p < 0.0001 when compared to control. ### p < 0.0001 when compared to RANKL treatment. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; OPG: osteoprotegerin; CTSK: cathepsin K; MMP-9: matrix metalloproteinase-9; NFATc1: nuclear factor of activated T cells c1; OSCAR: osteoclast-associated receptor; +: cells treated the reagent; -: cells untreated the reagent.
Foods 08 00290 g005
Figure 6. Effect of FST on RANKL-induced reactive oxygen species (ROS) generation in RAW 264.7 mouse macrophage-like cells. Cells were co-treated with 100 ng/mL RANKL for 1 h in the presence or absence of 800 μg/mL FST or 10 mM NAC. (A,D) Cells were stained with 5,6-carboxy-2′,7′-dichlorofluorescein diacetate (DCF-DA) and DCF fluorescence was measured by flow cytometry. Results are means of two independent experiments. (B) After staining with DCF-DA, images were obtained using a fluorescence microscope. Images are representative of at least three independent experiments. (C) Cells cultured under the conditions used to induce osteoclast differentiation were fixed and stained for F-actin ring with FITC-phalloidin solution and imaged under a fluorescence microscope. Representative photographs of the morphological changes observed are presented. (E) Cellular proteins were isolated from cells and the expression of NFATc1, phospho-NF-κB and phosphor-Nrf2 by Western blot analysis. β-actin was used as the internal control. The results shown are representative of three independent experiments. (F) Statistical analyses were conducted using analysis of variances. * p < 0.05 and *** p < 0.001 when compared to control. ### p < 0.0001 when compared to RANKL treatment. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; NAC: N-acetyl cysteine osteoprotegerin; NFATc1: nuclear factor of activated T cells c1; p- NF-κB p65: phosphorylated nuclear factor-kappa B p65; p-Nrf2: phosphorylated nuclear factor-erythroid 2-related factor 2; +: cells treated the reagent; -: cells untreated the reagent.
Figure 6. Effect of FST on RANKL-induced reactive oxygen species (ROS) generation in RAW 264.7 mouse macrophage-like cells. Cells were co-treated with 100 ng/mL RANKL for 1 h in the presence or absence of 800 μg/mL FST or 10 mM NAC. (A,D) Cells were stained with 5,6-carboxy-2′,7′-dichlorofluorescein diacetate (DCF-DA) and DCF fluorescence was measured by flow cytometry. Results are means of two independent experiments. (B) After staining with DCF-DA, images were obtained using a fluorescence microscope. Images are representative of at least three independent experiments. (C) Cells cultured under the conditions used to induce osteoclast differentiation were fixed and stained for F-actin ring with FITC-phalloidin solution and imaged under a fluorescence microscope. Representative photographs of the morphological changes observed are presented. (E) Cellular proteins were isolated from cells and the expression of NFATc1, phospho-NF-κB and phosphor-Nrf2 by Western blot analysis. β-actin was used as the internal control. The results shown are representative of three independent experiments. (F) Statistical analyses were conducted using analysis of variances. * p < 0.05 and *** p < 0.001 when compared to control. ### p < 0.0001 when compared to RANKL treatment. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; NAC: N-acetyl cysteine osteoprotegerin; NFATc1: nuclear factor of activated T cells c1; p- NF-κB p65: phosphorylated nuclear factor-kappa B p65; p-Nrf2: phosphorylated nuclear factor-erythroid 2-related factor 2; +: cells treated the reagent; -: cells untreated the reagent.
Foods 08 00290 g006
Figure 7. Activation of Nrf2 signaling pathway by FST in RAW 264.7 mouse macrophage-like cells. Cells were treated with FST with or without 100 ng/mL RANKL for 5 days. (A) Total cellular proteins were isolated from cells and the expression levels of Nrf2 and its regulatory proteins were assessed by Western blot analysis. β-actin was used as the internal control. (C) The expression of nuclear and cytosol Nrf2 were determined by Western blotting. Lamin B and β-actin were used as internal controls for the nuclear and cytosolic fractions, respectively. The results shown are representative of three independent experiments. (B,D) Statistical analyses were conducted using analysis of variances between groups. * p < 0.05 and *** p < 0.0001 when compared to control. # p < 0.05 and ### p < 0.0001 when compared to RANKL treatment. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; Nrf2: nuclear factor-erythroid 2-related factor 2; p-Nrf2: phosphorylated nuclear factor-erythroid 2-related factor 2; HO-1: heme oxygenase-1; NQO-1: NAD(P)H quinone oxidoreductase 1; +: cells treated the reagent; -: cells untreated the reagent.
Figure 7. Activation of Nrf2 signaling pathway by FST in RAW 264.7 mouse macrophage-like cells. Cells were treated with FST with or without 100 ng/mL RANKL for 5 days. (A) Total cellular proteins were isolated from cells and the expression levels of Nrf2 and its regulatory proteins were assessed by Western blot analysis. β-actin was used as the internal control. (C) The expression of nuclear and cytosol Nrf2 were determined by Western blotting. Lamin B and β-actin were used as internal controls for the nuclear and cytosolic fractions, respectively. The results shown are representative of three independent experiments. (B,D) Statistical analyses were conducted using analysis of variances between groups. * p < 0.05 and *** p < 0.0001 when compared to control. # p < 0.05 and ### p < 0.0001 when compared to RANKL treatment. RANKL: receptor of activator of nuclear factor kappa-B ligand; FST: fermented sea tangle extract; Nrf2: nuclear factor-erythroid 2-related factor 2; p-Nrf2: phosphorylated nuclear factor-erythroid 2-related factor 2; HO-1: heme oxygenase-1; NQO-1: NAD(P)H quinone oxidoreductase 1; +: cells treated the reagent; -: cells untreated the reagent.
Foods 08 00290 g007
Table 1. Information of primary and secondary antibodies.
Table 1. Information of primary and secondary antibodies.
AntibodyManufacturerItem No.
β-actinSanta Cruzsc-1615
CTSKSanta Cruzsc-48353
HO-1Millipore374090
IkBαSanta Cruzsc-371
Lamin BSanta Cruzsc-6216
MMP-9Abcam38898
NFATc1Santa Cruzsc-7294
NF-κB p65Santa Cruzsc-109
Phospho- NF-κB p65Cell signaling3033
Nrf2Santa Cruzsc-13032
phospho-Nrf2Abcam76026
NQO-1NovusNB200-209
OSCARR&D systemMAB1633
TRAPThermo Fisher ScientificPA5-42729
Goat anti-mouse IgG-HRPSanta Cruzsc-2005
Goat anti-rabbit IgG-HRPSanta Cruzsc-2004
Bovine anti-goat IgG-HRPSanta Cruzsc-2350
Mouse anti-rabbit igG-TRSanta CruzSc-3917
CTSK: cathepsin K; HO-1: heme oxygenase-1; IκBα: inhibitory proteins of kappa B, alpha; MMP-9: matrix metalloproteinase-9; NFATc1: nuclear factor of activated T cells c1; NF-κB: nuclear factor-kappa B; Nrf2: nuclear factor-erythroid 2-related factor 2; NQO-1: NAD(P)H quinone oxidoreductase 1; OSCAR: osteoclast-associated receptor; TRAP: tartrate-resistance acid phosphatase; HRP: horseradish-peroxidase.

Share and Cite

MDPI and ACS Style

Jeong, J.-W.; Ji, S.Y.; Lee, H.; Hong, S.H.; Kim, G.-Y.; Park, C.; Lee, B.-J.; Park, E.K.; Hyun, J.W.; Jeon, Y.-J.; et al. Fermented Sea Tangle (Laminaria japonica Aresch) Suppresses RANKL-Induced Osteoclastogenesis by Scavenging ROS in RAW 264.7 Cells. Foods 2019, 8, 290. https://0-doi-org.brum.beds.ac.uk/10.3390/foods8080290

AMA Style

Jeong J-W, Ji SY, Lee H, Hong SH, Kim G-Y, Park C, Lee B-J, Park EK, Hyun JW, Jeon Y-J, et al. Fermented Sea Tangle (Laminaria japonica Aresch) Suppresses RANKL-Induced Osteoclastogenesis by Scavenging ROS in RAW 264.7 Cells. Foods. 2019; 8(8):290. https://0-doi-org.brum.beds.ac.uk/10.3390/foods8080290

Chicago/Turabian Style

Jeong, Jin-Woo, Seon Yeong Ji, Hyesook Lee, Su Hyun Hong, Gi-Young Kim, Cheol Park, Bae-Jin Lee, Eui Kyun Park, Jin Won Hyun, You-Jin Jeon, and et al. 2019. "Fermented Sea Tangle (Laminaria japonica Aresch) Suppresses RANKL-Induced Osteoclastogenesis by Scavenging ROS in RAW 264.7 Cells" Foods 8, no. 8: 290. https://0-doi-org.brum.beds.ac.uk/10.3390/foods8080290

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop