Open Access

Anti‑osteoclastogenic effect of fermented mealworm extract by inhibiting RANKL‑induced NFATc1 action

  • Authors:
    • Ju Ri Ham
    • Mi-Kyung Lee
  • View Affiliations

  • Published online on: February 6, 2024     https://doi.org/10.3892/etm.2024.12418
  • Article Number: 130
  • Copyright: © Ham et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Augmented osteoclast activity and differentiation can lead to destructive bone diseases, such as arthritis and osteoporosis. Therefore, modulating osteoclastogenesis and differentiation may serve to be a possible strategy for treating such diseases. Tenebrio molitor larvae, also known as mealworms, are considered a good source of protein with nutritional value, digestibility, flavor and functional properties, such as antioxidant, anti‑diabetic and anti‑obesity effects. However, the role of mealworms in osteoclastogenesis remains poorly understood. The present study therefore investigated the effects of fermented mealworm extract (FME) on receptor activator of nuclear factor κB ligand (RANKL)‑induced osteoclastogenesis in bone marrow‑derived macrophages (BMMs) whilst also attempting to understand the underlying mechanism, if any. The cells treated with RANKL were used as the negative control. To prepare FME, defatted mealworm powder was fermented with a Saccharomyces cerevisiae strain, and then extracted with fermented alcohol. Cell viability of BMMs isolated from 5‑week‑old Institute of Cancer Research mice was measured using Cell Counting Kit‑8 assay. Subsequently, the effects of FME on osteoclast differentiation were measured using tartrate‑resistant acid phosphatase (TRAP) staining. In addition, expression of markers associated with osteoclast differentiation was assessed by reverse transcription‑quantitative PCR. Expression of nuclear factor of activated T‑cells cytoplasmic 1 (NFATc1) was assessed by western blotting. TRAP staining revealed that FME inhibited osteoclast differentiation in a dose‑dependent manner (10‑100 µg/ml) without causing cytotoxicity. Specifically, the formation of osteoclasts appear to have been suppressed by FME as indicated by the reduction in the number of TRAP‑positive multinucleated cells observed. Furthermore, FME treatment significantly decreased the mRNA expression of c‑Fos, whilst also significantly decreasing the expression of NFATc1 on both protein and mRNA levels. c‑Fos and NFATc1 are transcription factors that can regulate osteoclast differentiation. FME treatment also reduced the expression of genes associated with osteoclast differentiation and function, including dendritic cell‑specific transmembrane protein, osteoclast associated Ig‑like receptor, Cathepsin K and TRAP, compared with that in the control group. Subsequently, FME was found to effectively suppress RANKL‑induced osteoclast differentiation compared with that by the non‑fermented mealworm extract. These findings suggest that FME may confer anti‑osteoclastogenic effects, providing insights into its potential application in treatment of osteoporosis.

Introduction

Osteoporosis is a metabolic bone disorder that is characterized by a progressive decline in bone mass and reduction in bone strength, leading to fragile bones and an increased in the risk of fracture (1). Physiologically, it typically occurs due to an imbalance between osteoblast and osteoclast activity, with osteoclast activity becoming excessive and the resultant bone resorption outpacing bone formation during remodeling (1). According to results from a previous meta-analysis from 2000 to 2020, the worldwide prevalence of osteoporosis was 18.3% (95% CI, 16.2-20.7) (2). In particular, this disease is especially common among the elderly (aged >65 years), placing a problematic health burden on this population (3). Therefore, inhibition of osteoclastic activity has been proposed to be a potential strategy for the treatment or even prevention of osteoporosis (4). Hormone replacement therapy, bisphosphonates and inhibitor of the receptor activator of nuclear factor κB ligand (RANKL) are the most commonly applied methods for osteoporosis treatment (5). However, due to the side effects (such as muscle pain and dyspepsia) of these aforementioned drugs, efforts are being made to substitute them with naturally occurring compounds such as triphenyl hexene and methoxsalen (4,6,7).

Mealworms (Tenebrio molitor larvae) are important edible insects and serve as a potential protein source for animals and humans (8,9). Previous studies have shown that mealworms can exert anti-obesity (10), anti-diabetic (11) and antioxidant (12) activities. In another previous study, Ham et al (13) found that the fermented extract of defatted mealworm (FME) had higher amounts of free amino acids and essential amino acids compared with those in their non-fermented (ME) counterparts. Mealworms are particularly rich in branched-chain amino acids (BCAAs), such as leucine, isoleucine and valine, which were previously found to be exceeded in quantity by FME (14). It has also been reported that FME treatment in in vivo models, including rats or mice, resulted in the attenuation of alcoholic and non-alcoholic hepatic steatosis and type 2 diabetes (13-15). In addition, Cui et al (16) reported that serum leucine and valine levels in patients were positively associated with total body bone mineral density (BMD) in Mendelian randomization analyses using a summary-level genome-wide association study. Therefore, the present study aimed to investigate the impact of FME on bone health, with specific focus on osteoclast differentiation. In addition, the present study aimed to elucidate its underlying mechanism of action.

Materials and methods

Preparation of ME and FME

ME and FME were provided from the Suncheon Research Center for Bio Health Care. Briefly, fat was first removed from freeze-dried mealworms for 5 days at -125˚C using edible hexane. Edible hexane (1 l) was added to freeze-dried mealworms (200 g) and stirred at 60 rpm for 24 h at 24˚C. This lipid extraction process was repeated three times. This extract was then filtered using polypropylene and the remaining solution was evaporated using a rotary evaporator (customized for a large capacity) at 24-25˚C and powdered. To prepare FME, defatted mealworm powder was fermented with a Saccharomyces cerevisiae strain (cat. no. KCTC 17299) provided by the Korean Collection for Type Cultures. The medium was prepared by mixing yeast, defatted mealworm powder and dextrose in a ratio of 1:2:2 (w/w). Specifically, yeast was inoculated (1x106 CFU/ml) into the medium and it was fermented for 72 h at 30±2˚C with 150 rpm shaking. The solvent used was distilled water, and the total volume per reaction was 1 l. Fermented alcohol (Ethanol Supplies World Co., Ltd.) was then added to the cultured medium at a ratio of 7:3 (fermented ethanol: cultured medium, v/v), and then the mixture was extracted under reflux cooling for 3 h at 85˚C and filtered under pressure (4.08 bar/60 PSI) at 24-25˚C using an ADVANTEC No. 2 filter paper (Toyo Roshi Kaisha, Ltd.). The resulting extract was evaporated with a rotary vacuum concentrator and freeze-dried for 72 h at -125˚C to obtain the extraction powder.

For the non-fermented ME, yeast, defatted mealworm powder and dextrose were mixed in a ratio of 1:2:2 (w/w) without any incubation, and then fermented alcohol was immediately added to the mixture to proceed with the fermentation extraction process (Fig. 1).

The freeze-dried powder was dissolved in DMSO (300 µg/ml) and used for subsequent experiments.

Collection of bone marrow-derived macrophages (BMM) and osteoclast differentiation

BMMs were obtained from the femurs and tibiae of 5-week-old male ICR mice (n=2; weight, 25.85±0.35 g) purchased from Raon Bio (Yongin, Korea), as described in previous studies (17,18). The mice were housed in a controlled environment at 22±2˚C and 50±5% humidity and maintained in a 12-h light/dark cycle. The mice had free access to food and water. The mice were sacrificed by cervical dislocation, before their femurs and tibiae were carefully cleared of adherent soft tissues. The tip of each bone was then removed with forceps and the marrow was extracted by inserting a 1-ml syringe needle into the end of the bone and flushed with α-minimum essential medium (α-MEM) supplemented with 100 U/ml penicillin/streptomycin (all from Thermo Fisher Scientific, Inc.). After flushing, the cells were centrifuged at 400 x g for 5 min at 4˚C. Using Red Blood Cell Lysis Buffer (Merck KGaA), red blood cells were removed using centrifugation (400 x g for 5 min at 4˚C). After this step, the supernatant was removed, and α-MEM containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) was added. After filtering through a 40-µm nylon mesh filter (BD Biosciences), the cells were immediately placed in the culture medium (α-MEM containing 10% FBS; Gibco; Thermo Fisher Scientific, Inc.) and cultured under 37˚C in a humidified atmosphere containing 5% CO2. The cells from the two mice were pooled into the same culture. Previous studies have reported a typical yield of 7.5x107-1.5x108 cells per mouse, although variations may occur depending on the specific conditions (17,19). The BMMs extracted from mouse bones were cultured for 3-7 days, during which the osteoclasts adhere whilst other substances and cells that do not attach fall off. During this period, cells were rinsed with fresh medium every 2 or 3 days and checked for cell status under a microscope. This process allows for isolation of pure osteoclasts (17,19). Animal experiments were conducted according to the ethical guidelines of the Sunchon National University Institutional Animal Care and Use Committee (approval no. SCNU_IACUC-2021-06).

For osteoclast differentiation, BMMs were seeded at a density of 1x104 cells/well in a 96-well plate. The cells were incubated with 30 ng/ml recombinant murine macrophage colony-stimulating factor (M-CSF) provided by PeproTech, Inc., in α-MEM containing 10% FBS at 37˚C. The following day, the cells were treated with 10 ng/ml RANKL (R&D Systems, Inc.) along with either non-fermented ME or FME (1, 3, 10, 30 or 100 µg/ml) for 3 days at 37˚C. DMSO was used as a vehicle control in the RANKL-only treated cells. The concentrations used in the present study were based on a previous study (20).

Detection of osteoclast differentiation using tartrate-resistant acid phosphatase (TRAP) staining assay

After the differentiation process, cells in the 96-well plate were washed with PBS and subsequently fixed with formalin (3.7%) for 5 min at room temperature. Following fixation, the cells were treated with Triton X-100 (0.1%) for 10 min. Subsequently, the cells were treated with a TRAP solution (Sigma-Aldrich; Merck KGaA) for 10 min in a dark room at room temperature. TRAP-positive multinucleated cells with three or more nuclei were identified and counted as mature osteoclasts using light microscopy. The average of TRAP-positive multinucleated cells was determined from five locations per well and calculated.

Determining the cytotoxicity of FME on the BMMs

BMMs (1x104 cells/well) were cultured in 10% FBS-supplemented α-MEM containing M-CSF (30 ng/ml) for 24 h at 37˚C. The next day, FME (10, 30 or 100 µg/ml) was added and incubated with the cells for 3 days at 37˚C, before cell viability was evaluated using the Cell Counting Kit-8 (CCK-8) assay (Dojindo Laboratories, Inc.). In each well, 100 µl of the culture supernatant was mixed with 10 µl of CCK-8 reagent and incubated for 4 h at 37˚C. Absorbance was measured at 450 nm at 1-h intervals. The sample treatment period was determined based on previous studies (21-23).

Reverse transcription-quantitative PCR (RT-qPCR)

BMMs (3x105 cells/well) were incubated for 16-24 h at 37˚C with α-MEM containing 10% FBS. Following the initial incubation, the cells were treated with RANKL (10 ng/ml), M-CSF (30 ng/ml) and FME (30 or 100 µg/ml) at the same time, before being incubated for an additional 72 h at 37˚C. The total RNA was extracted from the BMM cells using the TRIzol reagent (Thermo Fisher Scientific Inc.) For the reverse transcription of the total RNA into cDNA, the ReverTra Ace qPCR RT master mix (Toyobo Life Science) was utilized. cDNA was synthesized by reacting 5X RT Master Mix, RNA template (1 µg/µl) and nuclease-free water following the manufacturer's protocol for 15 min at 37˚C, 5 min at 50˚C and 5 min at 98˚C. The mRNA expression levels were quantified using QuantiTect SYBR® Green RT-PCR kits (Qiagen GmbH) in a CFX96 real-time system (Bio-Rad Laboratories, Inc.). Amplification was performed as follows: Initial denaturation at 95˚C for 15 min, followed by 40 cycles of 94˚C for 15 sec, 50˚C for 30 sec and 72˚C for 30 sec. The primers used for each gene are shown in Table I. An internal housekeeper gene GAPDH was used as described by Stephens et al (24). The mRNA expression was determined using the 2-ΔΔCq method (25).

Table I

Primers used for reverse transcription-quantitative PCR.

Table I

Primers used for reverse transcription-quantitative PCR.

Gene nameForward/reverse (5'-3')Species specificityAmplicon sizeMelting temperature, ˚CRNA-seq identification No.
c-Fos CCAGTCAAGAGCATCAGCAA/AAGTAGTGCAGCCCGGAGTAMouse24760NM_010234.3
Cathepsin K ACTCCAGTCAAGAACCAGGG/TCTTCTTGAGTTGGCCCTCCMouse8259NM_007802.4
Dendritic cell-specific transmembrane protein CCAAGGAGTCGTCCATGATT/ GGCTGCTTTGATCGTTTCTCMouse25559NM_029422.4
Glyceraldehyde-3- phosphate dehydrogenase AAGGTCATCCCAGAGCTGAA/ CTGCTTCACCACCTTCTTGAMouse13859NM_001411843.1
Nuclear factor-activated T cells c1 AGGACCCGGAGTTCGACTT/ GTCGAGGTGACACTAGGGGAMouse10660NM_001164112.1
Osteoclast-associated Ig-like receptor GTCCTGTCGCTGATACTCCA/ CGCTGTTGGTGTGAAGTCAGMouse8759NM_001290377.1
Tartrate-resistant acid phosphatase AGGAAGAGCCTTCAAGTAAGTG/ CCACCCATGAATCCATCTTCTMouse8957NM_001102405.1
Western blot analysis

BMMs (3x105 cells/well) were seeded into a 6-well plate and incubated with α-MEM containing 10% FBS at 37˚C for 16-24 h. Following the initial incubation, the cells were treated with M-CSF (30 ng/ml), RANKL (10 ng/ml) and FME (30 or 100 µg/ml) at the same time and incubated for an additional 72 h at 37˚C. At the end of the incubation period, the cells were washed with PBS and lysed in a lysis buffer (50 mM HEPES, 150 mM NaCl, 1 mM EDTA, 2 mM EGTA and 1% Triton) containing protease inhibitor (25 µg/ml leupeptin, 2 µg/ml aprotinin and 50 mM sodium fluoride) for 1 h at 4˚C. After lysis, the cells were centrifuged at 18,928 x g for 5 min at 4˚C and the supernatants were collected for western blotting. The protein concentrations in the supernatants were determined using the Bradford method (26). A total of 10 µg protein samples were separated using 10% SDS-PAGE and transferred onto nitrocellulose membranes. Subsequently, a blocking step was carried out at room temperature for 1 h using 5% bovine serum albumin (Sigma-Aldrich; Merck KGaA). The membranes were then incubated overnight at 4˚C with antibodies against nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1; cat no. sc-7294; 1:200; Santa Cruz Biotechnology Inc.) and β-actin (cat no. A2066; 1:2,000; Sigma-Aldrich). Subsequently, the membranes were incubated with anti-mouse monoclonal (cat no. 7076) and anti-rabbit IgG (cat no. 7074) secondary antibodies (1:10,000; Cell Signaling Technology, Inc.) for 2 h at room temperature. The protein bands were visualized using a Western Blotting Luminol Reagent (Santa Cruz Biotechnology, Inc.), and the visualization of protein bands was assessed using a chemiluminescence image analyzer (Alliance Q9 advanced chemiluminescence imager). Densitometry analysis of the visualized protein bands was performed using the UVITEC Alliance Q9 advanced system (version 4.3.8; UVITEC) to quantify the protein expression.

Statistical analysis

All data are presented as the means ± SEM of three independent tests. Statistical analysis was performed using the SPSS version 26 software (IBM Corp.). The data were analyzed by two-way or one-way ANOVA followed by a Holm-Sidak post hoc test to examine the differences among the groups. The mRNA levels of TRAP, cathepsin K (CTSK), osteoclast-associated Ig-like receptor (OSCAR) and dendritic cell-specific transmembrane protein (DC-STAMP) were compared using the unpaired Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

RANKL-induced osteoclast differentiation is inhibited more effectively by FME compared with non-fermented mealworm extract

FME was found to inhibit osteoclast differentiation more effectively compared with non-fermented ME at concentrations of 10 and 30 µg/ml. This difference was small at 1 and 3 µg/ml between FME- and ME-treated cells (Fig. 2A). However, the number of TRAP-positive multinucleated cells with three or more nuclei was reduced more effectively in FME-treated cells compared with that in the non-fermented ME-treated cells at concentrations of 1 to 30 µg/ml (Fig. 2B). To further evaluate the effects of FME on osteoclast differentiation, FME concentrations of 10, 30 and 100 µg/ml were next tested, to assess if these concentrations were capable of inducing cytotoxicity. These FME concentrations did not indicate cytotoxicity. Furthermore, the cell viability increased at 100 µg/ml FME compared with the control cells that were not treated with FME, although the difference was not statistically significant (Fig. 3B). FME exhibited a dose-dependent suppression of osteoclast differentiation (Fig. 3A). Notably, treatment with FME at concentrations >10 µg/ml significantly reduced the formation of osteoclasts, as evidenced by the significantly decreased number of TRAP-positive multinucleated cells with three or more nuclei (Fig. 3A).

RANKL-induced NFATc1 activation is suppressed by FME

To evaluate the mechanism underlying the anti-osteoclastogenic activity of the FME, the levels of the expression of key transcription factors involved in osteoclastogenesis, namely c-Fos and NFATc1, were examined. As shown in Fig. 4A, FME (100 µg/ml) significantly downregulated the expression of both transcription factors on day 1 compared with that in the RANKL-only group. Additionally, compared with that in the RANKL-only group, FME (30 µg/ml) significantly reduced NFATc1 expression on day 0 (2-h reaction). Furthermore, compared with that in the RANKL-only group, c-Fos mRNA expression levels in FME 100 µg/ml showed a statistical difference on day 1 but none thereafter, whereas NFATc1 mRNA expression levels showed a statistical difference on days 0, 1 and 3 at 100 µg/ml of FME (Fig. 4A). Therefore, the present study next evaluated the impact of FME on the expression of the NFATc1 protein. The results revealed that FME exerted a dose-dependent downregulation of NFATc1 protein expression on each of the 3 days. In particular, there was a significant difference in concentration on day 2 (Fig. 4B).

Expression of osteoclast-specific marker genes is downregulated by FME treatment

NFATc1 has been previously reported to promote the expression of osteoclast-associated genes, including TRAP, CTSK, OSCAR and DC-STAMP (27,28). Therefore, the present study examined the effect of FME (100 µg/ml) on the mRNA expression of such genes. RANKL stimulation led to a time-dependent increase in the expression of the TRAP, CTSK, OSCAR and DC-STAMP (Fig. 5). However, treatment with FME significantly suppressed the mRNA expression of these genes compared with that in their RANKL-only counterparts on each of the 3 days (Fig. 5). These findings suggest that FME can suppressed the expression of osteoclast-associated genes, which are regulated by the NFATc1 signaling pathway.

Discussion

Osteoclasts are multinucleated cells of the macrophage lineage that can be derived from BMMs (29). They serve an important role in bone resorption (30). The process of osteoclast formation is regulated by genetic, humoral and mechanical signals (31). Among these, M-CSF and RANKL serve pivotal roles in stimulating osteoclast differentiation (30). The present study therefore induced osteoclast differentiation in BMMs using M-CSF (30 ng/ml) and RANKL (10 ng/ml). FME, compared with the non-fermented ME, was found to effectively suppressed the RANKL-stimulated osteoclast differentiation, as evidenced by TRAP staining. To assess whether the anti-osteoclastogenic effect of FME was associated with cytotoxicity, a cell viability assay was conducted for 3 days when stable cell growth was observed under the microscope. Given that the experiment period for differentiation inhibition spanned 3 days, the present study verified the absence of cytotoxicity throughout this duration. Although parameters in cell experiments can vary, such as the environment and cell condition, previous similar studies (21-23,32) also treated cells for 3 days using 1x104 cells in 96-well plates under the same conditions and found no cytotoxicity at concentrations ≤100 µg/ml. FME exhibited a dose-dependent inhibition of osteoclast differentiation in the concentration range of 10-100 µg/ml, suggesting that it has an anti-osteoclastogenic effect without causing cytotoxicity.

The RANKL signaling pathway activates several transcription factors, such as the NF-κB, activator protein 1(33) and NFATc1(34). NFATc1 functions as a master transcription factor downstream of RANKL-RANK signaling and serves a critical role in osteoclast differentiation, formation and bone resorption (35,36). Therefore, the present study investigated whether FME can alter the activity of NFATc1 during RANKL-stimulated osteoclastogenesis. FME dose-dependently downregulated NFATc1 gene and protein expression in response to RANKL stimulation. In addition, FME significantly downregulated c-Fos gene expression on day 1. c-Fos is an important transcription factor that is involved in the early stage of osteoclast differentiation whilst promoting NFATc1 action (37). These findings suggest that FME can inhibit the expression of key transcription factors involved in osteoclastogenesis, including c-Fos and NFATc1. In particular, the mRNA expression of NFATc1 continued to increase until day 3, whereas the corresponding protein expression levels peaked on day 2 and decreased on day 3. These observations are consistent with the results reported by Kim et al (38), which reported that the levels of NFATc1 protein expression decreased during late stage osteoclastogenesis due to its translocation from the cytosol into the nucleus in the presence of RANKL. They also suggested that NFATc1 continued to be transcribed until the end of osteoclastogenesis (38). However, a limitation of the present study is that it did not perform NFATc1 knockdown or overexpression.

Subsequent to the activation of NFATc1, a continuous upregulation in the expression of various factors has been previously reported, including CTSK, TRAP, DC-STAMP and OSCAR (39-42). CTSK, TRAP, DC-STAMP and OSCAR contribute to the differentiation of osteoclasts into mature osteoclasts and potentially worsen bone disorders (39-42). The present study revealed that RANKL stimulation markedly increased the mRNA expression levels of CTSK, TRAP, DC-STAMP and OSCAR in a time-dependent manner. However, FME treatment significantly reduced the expression of these genes from day 0 to 3. This suggest that FME has the ability to suppress the expression of osteoclast-associated genes involved in bone resorption and fusion of mononuclear pre-osteoclasts. Notably, FME treatment at concentrations of 30 and 100 µg/ml resulted in a significant reduction in the number of TRAP-positive multinucleated cells. The fusion of mononuclear pre-osteoclasts is a critical step in the formation of mature multinucleated osteoclasts (27,43), which can lead to increased bone resorption activity and decrease in bone mass (27,44). Specifically, DC-STAMP serve an essential role in the fusion of mononuclear osteoclasts (27,45). The reduction observed in the number of TRAP-positive multinucleated cells suggests that FME may interfere with the fusion process and consequently inhibit osteoclast maturation and bone resorption.

FME was previously found to have higher amino acid content compared with non-fermented ME, with leucine and alanine being the most abundant amino acids present (14). A recent study reported a negative correlation between plasma valine, leucine, isoleucine and alanine levels with BMD in elderly women (46). By contrast, in men, plasma tryptophan levels exhibited an inverse correlation with BMD (46). Another study previously demonstrated that although BCAAs can activate osteoclast differentiation, at higher concentrations BCAAs were actually found to inhibit this process (47). This suggests the presence of a negative feedback mechanism for BCAAs (47). However, the effects of amino acids on osteoclast differentiation remain poorly understood and further investigation is required to determine the specific mechanism and impact of different amino acids on osteoclast differentiation. Another limitation of the present study is that it did not identify specific compounds and their potential effects on osteoporosis. Additionally, to further substantiate the anti-osteoclastogenic effects of FME and their effects on NFATc1 activity, experiments using NFATc1 inhibitors as opposed to expression manipulation should be conducted.

Taken together, results from the present study suggest that FME has a potential inhibitory effect on osteoclast differentiation and formation. This effect may be associated with the downregulation of NFATc1 activity and the suppression of osteoclastogenic marker expression, such as CTSK, TRAP, DC-STAMP and OSCAR. These results suggest that FME is a promising intervention method for the prevention or treatment of osteoporosis. However, since the present study was only conducted in vitro, it is necessary to evaluate its efficacy and safety through in vivo studies and clinical trials in animal and human subjects.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the National Research Foundation of Korea grant funded by the Korea government (grant nos. NRF-2020R1A6A3A01095872 and NRF-2023R1A2C1003276).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

MKL conceived the study and edited the manuscript. JRH carried out all of the assays in the study and drafted the manuscript. All authors have read and approved the final manuscript. JRH and MKL confirm the authenticity of all the raw data.

Ethics approval and consent to participate

The experimental protocols were approved by the Sunchon National University Institutional Animal Care and Use Committee (approval no. SCNU_IACUC-2021-06; Suncheon, Korea) and were carried out on the basis of relevant national and international guidelines.

Patient consent for publication

Not applicable.

Competing interests

The authors declared that they have no competing interests.

References

1 

Baron R and Kneissel M: WNT signaling in bone homeostasis and disease: From human mutations to treatments. Nat Med. 19:179–192. 2013.PubMed/NCBI View Article : Google Scholar

2 

Salari N, Ghasemi H, Mohammadi L, Behzadi MH, Rabieenia E, Shohaimi S and Mohammadi M: The global prevalence of osteoporosis in the world: A comprehensive systematic review and meta-analysis. J Orthop Surg Res. 16(609)2021.PubMed/NCBI View Article : Google Scholar

3 

Chen X, Wang C, Qiu H, Yuan Y, Chen K, Cao Z, Xiang Tan R, Tickner J, Xu J and Zou J: Asperpyrone A attenuates RANKL-induced osteoclast formation through inhibiting NFATc1, Ca2+ signalling and oxidative stress. J Cell Mol Med. 23:8269–8279. 2019.PubMed/NCBI View Article : Google Scholar

4 

He J, Chen K, Deng T, Xie J, Zhong K, Yuan J, Wang Z, Xiao Z, Gu R, Chen D, et al: Inhibitory effects of rhaponticin on osteoclast formation and resorption by targeting RANKL-induced NFATc1 and ROS activity. Front Pharmacol. 12(645140)2021.PubMed/NCBI View Article : Google Scholar

5 

Tu KN, Lie JD, Wan CKV, Cameron M, Austel AG, Nguyen JK, Van K and Hyun D: Osteoporosis: A review of treatment options. P T. 43:92–104. 2018.PubMed/NCBI

6 

Lee Y, Lee HJ, Shin HB, Ham JR, Lee MK, Lee MJ and Son YJ: Triphenyl hexene, an active substance of Betaone barley water extract, inhibits RANKL-induced osteoclast differentiation and LPS-induced osteoporosis. J Funct Foods. 92(105037)2022.

7 

Ham JR, Choi RY, Yee ST, Hwang YH, Kim MJ and Lee MK: Methoxsalen supplementation attenuates bone loss and inflammatory response in ovariectomized mice. Chem Biol Interact. 278:135–140. 2017.PubMed/NCBI View Article : Google Scholar

8 

Hong J, Han T and Kim YY: Mealworm (Tenebrio molitor larvae) as an alternative protein source for monogastric animal: A review. Animals (Basel). 10(2068)2020.PubMed/NCBI View Article : Google Scholar

9 

Veldkamp T and Bosch G: Insects: A protein-rich feed ingredient in pig and poultry diets. Anim Front. 5:45–50. 2015.

10 

Seo M, Goo T, Chung MY, Baek M, Hwang J, Kim M and Yun E: Tenebrio molitor larvae inhibit adipogenesis through AMPK and MAPKs signaling in 3T3-L1 adipocytes and obesity in high-fat diet-induced obese mice. Int J Mol Sci. 18(518)2017.PubMed/NCBI View Article : Google Scholar

11 

Kim SY, Park JE and Han JS: Tenebrio molitor (mealworm) extract improves insulin sensitivity and alleviates hyperglycemia in C57BL/Ksj-db/db mice. Kor J Life Sci. 29:570–579. 2019.

12 

Baek M, Kim M, Kwon Y, Hwang J, Goo T, Jun M and Yun E: Effects of processing methods on nutritional composition and antioxidant activity of mealworm (Tenebrio molitor) larvae. Entomol Res. 49:284–293. 2019.

13 

Ham JR, Choi RY, Lee Y and Lee MK: Effects of edible insect Tenebrio molitor larva fermentation extract as a substitute protein on hepatosteatogenesis and proteomic changes in obese mice induced by high-fat diet. Int J Mol Sci. 22(3615)2021.PubMed/NCBI View Article : Google Scholar

14 

Choi RY, Ham JR, Ryu HS, Lee SS, Michelle AM, Paik MJ, Ji M, Park KW, Kang KY, Lee HI, et al: Defatted Tenebrio molitor larva fermentation extract modifies steatosis, inflammation and intestinal microflora in chronic alcohol-fed rats. Nutrients. 12(1426)2020.PubMed/NCBI View Article : Google Scholar

15 

Turkyilmaz A, Lee Y and Lee MK: Fermented extract of mealworm (Tenebrio molitor larvae) as a dietary protein source modulates hepatic proteomic profiles in C57BLKS/J-db/db mice. J Insects Food Feed. 9:1199–1210. 2023.

16 

Cui Z, Feng H, He B, He J and Tian Y: Relationship between serum amino acid levels and bone mineral density: A mendelian randomization study. Front Endocrinol (Lausanne). 12(763538)2021.PubMed/NCBI View Article : Google Scholar

17 

Zhang X, Goncalves R and Mosser DM: The isolation and characterization of murine macrophages. Curr Protoc Immunol Chapter. 14:14.1.1–14.1.14. 2008.PubMed/NCBI View Article : Google Scholar

18 

Huang S, Xu L, Sun Y, Wu T, Wang K and Li G: An improved protocol for isolation and culture of mesenchymal stem cells from mouse bone marrow. J Orthop Transl. 3:26–33. 2014.PubMed/NCBI View Article : Google Scholar

19 

Kim HJ, Kang WY, Seong SJ, Kim SY, Lim MS and Yoon YR: Follistatin-like 1 promotes osteoclast formation via RANKL-mediated NF-κB activation and M-CSF-induced precursor proliferation. Cell Signal. 28:1137–1144. 2016.PubMed/NCBI View Article : Google Scholar

20 

Kim KJ, Lee Y, Son SR, Lee H, Son YJ, Lee MK and Lee M: Water extracts of hull-less waxy barley (Hordeum vulgare L.) cultivar ‘Boseokchal’ inhibit RANKL-induced osteoclastogenesis. Molecules. 24(3735)2019.PubMed/NCBI View Article : Google Scholar

21 

Jeong DH, Kwak SC, Lee MS, Yoon KH, Kim JY and Lee CH: Betulinic acid inhibits RANKL-induced osteoclastogenesis via attenuating Akt, NF-κB, and PLCγ2-Ca2+ signaling and prevents inflammatory bone loss. J Nat Prod. 83:1174–1182. 2020.PubMed/NCBI View Article : Google Scholar

22 

Park GD, Cheon YH, Eun SY, Lee CH, Lee MS, Kim JY and Cho HJ: β-Boswellic acid inhibits RANKL-induced osteoclast differentiation and function by attenuating NF-κB and PLCγ2 signaling pathways. Molecules. 26(2665)2021.PubMed/NCBI View Article : Google Scholar

23 

Nam HH, Lee AY, Seo YS, Park I, Yang S, Chun JM, Moon BC, Song JH and Kim JS: Three Scrophularia species (Scrophularia buergeriana, S. koreaiensis, and S. takesimensis) inhibit RANKL-induced osteoclast differentiation in bone marrow-derived macrophages. Plants (Basel). 9(1656)2020.PubMed/NCBI View Article : Google Scholar

24 

Stephens AS, Stephens SR and Morrison NA: Internal control genes for quantitative RT-PCR expression analysis in mouse osteoblasts, osteoclasts and macrophages. BMC Res Notes. 4(410)2011.PubMed/NCBI View Article : Google Scholar

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

26 

Bradford MM: A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 72:248–254. 1976.PubMed/NCBI View Article : Google Scholar

27 

Zeng Xz, He L, Wang S, Wang K, Zhang YY, Tao L, Li XJ and Liu SW: Aconine inhibits RANKL-induced osteoclast differentiation in RAW264.7 cells by suppressing NF-κB and NFATc1 activation and DC-STAMP expression. Acta Pharmacol Sin. 37:255–263. 2016.PubMed/NCBI View Article : Google Scholar

28 

Negishi-Koga T and Takayanagi H: Ca2+-NFATc1 signaling is an essential axis of osteoclast differentiation. Immunol Rev. 231:241–256. 2009.PubMed/NCBI View Article : Google Scholar

29 

Wang J, Guan H, Liu H, Lei Z, Kang H, Guo Q, Dong Y, Liu H, Sun Y, Fang Z and Li F: Inhibition of PFKFB3 suppresses osteoclastogenesis and prevents ovariectomy-induced bone loss. J Cell Mol Med. 24:2294–2307. 2020.PubMed/NCBI View Article : Google Scholar

30 

Zhou Z, Immel D, Xi CX, Bierhaus A, Feng X, Mei L, Naworth P, Stern DM and Xiong WC: Regulation of osteoclase function and bone mass by RAGE. J Exp Med. 203:1067–1080. 2006.PubMed/NCBI View Article : Google Scholar

31 

Jin H, Yao L, Chen K, Liu Y, Wang Q, Wang Z, Liu Q, Cao Z, Kenny J, Tickner J, et al: Evodiamine inhibits RANKL-induced osteoclastogenesis and prevents ovariectomy-induced bone loss in mice. J Cell Mol Med. 23:522–534. 2019.PubMed/NCBI View Article : Google Scholar

32 

Kwak HB, Lee BK, Oh J, Yeon JT, Choi SW, Cho HJ, Lee MS, Kim JJ, Bae JM, Kim SH and Kim HS: Inhibition of osteoclast differentiation and bone resorption by rotenone, through down-regulation of RANKL-induced c-Fos and NFATc1 expression. Bone. 46:724–731. 2010.PubMed/NCBI View Article : Google Scholar

33 

Jimi E and Ghosh S: Role of nuclear factor-kappaB in the immune system and bone. Immunol Rev. 208:80–87. 2005.PubMed/NCBI View Article : Google Scholar

34 

Kim JH, Kim K, Kim I, Seong S, Kim SW and Kim N: Role of anoctamin 5, a gene associated with gnathodiaphyseal dysplasia, in osteoblast and osteoclast differentiation. Bone. 120:432–438. 2019.PubMed/NCBI View Article : Google Scholar

35 

Xiao D, Zhou Q, Gao Y, Cao B, Zhang Q, Zeng G and Zong S: PDK1 is important lipid kinase for RANKL-induced osteoclast formation and function via the regulation of the Akt-GSK3β-NFATc1 signaling cascade. J Cell Biochem. 121:4542–4557. 2020.PubMed/NCBI View Article : Google Scholar

36 

Takayanagi H, Sunhwa K, Koga T, Nishina H, Isshiki M, Yoshida H, Saiura A, Isobe M, Yokochi T, Inoue J, et al: Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell. 3:889–901. 2002.PubMed/NCBI View Article : Google Scholar

37 

Kim JH and Kim N: Regulation of NFATc1 in osteoclast differentiation. J Bone Metab. 21:233–241. 2014.PubMed/NCBI View Article : Google Scholar

38 

Kim JH, Kim K, Jin HM, Song I, Youn BU, Lee SH, Choi Y and Kim N: Negative feedback control of osteoclast formation through ubiquitin-mediated down-regulation of NFATc1. J Biol Chem. 285:5224–5231. 2010.PubMed/NCBI View Article : Google Scholar

39 

Lee Y, Kantayos V, Kim JS, Rha ES, Son YJ and Baek SH: Inhibitory effects of protopanaxadiol-producing transgenic rice seed extracts on RANKL-induced osteoclast differentiation. Life (Basel). 12(1886)2022.PubMed/NCBI View Article : Google Scholar

40 

Suh KS, Chon S, Jung WW and Choi EM: Effects of methylglyoxal on RANKL-induced osteoclast differentiation in RAW264.7 cells. Chem Biol Interact. 296:18–25. 2018.PubMed/NCBI View Article : Google Scholar

41 

Hwang YH, Kim T, Kim R and Ha H: Magnolol inhibits osteoclast differentiation via suppression of RANKL expression. Molecules. 23(1598)2018.PubMed/NCBI View Article : Google Scholar

42 

Song C, Yang X, Lei Y, Zhang Z, Smith W, Yan J and Kong L: Evaluation of efficacy on RANKL induced osteoclast from RAW264.7 cells. J Cell Physiol. 234:11969–11975. 2019.PubMed/NCBI View Article : Google Scholar

43 

Zeng Z, Zhang C and Chen J: Lentivirus-mediated RNA interference of DC-STAMP expression inhibits the fusion and resorptive activity of human osteoclasts. J Bone Miner Metab. 31:409–416. 2013.PubMed/NCBI View Article : Google Scholar

44 

Islam R, Bae HS, Yoon WJ, Woo KM, Baek JH, Kim HH, Uchida T and Ryoo HM: Pin1 regulates osteoclast fusion through suppression of the master regulator of cell fusion DC-STAMP. J Cell Physiol. 229:2166–2174. 2014.PubMed/NCBI View Article : Google Scholar

45 

Zhang C, Dou CE, Xu J and Dong S: DC-STAMP, the key fusion-mediating molecule in osteoclastogenesis. J Cell Physiol. 229:1330–1335. 2014.PubMed/NCBI View Article : Google Scholar

46 

Panahi N, Fahimfar N, Roshani S, Arjmand B, Gharibzadeh S, Shafiee G, Migliavacca E, Breuille D, Feige JN, Grzywinski Y, et al: Association of amino acid metabolites with osteoporosis, a metabolomic approach: Bushehr elderly health program. Metabolomics. 18(63)2022.PubMed/NCBI View Article : Google Scholar

47 

Go M, Shin E, Jang SY, Nam M, Hwang G and Lee SY: BCAT1 promotes osteoclast maturation by regulating branched-chain amino acid metabolism. Exp Mol Med. 54:825–833. 2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2024
Volume 27 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ham J and Ham J: Anti‑osteoclastogenic effect of fermented mealworm extract by inhibiting RANKL‑induced NFATc1 action. Exp Ther Med 27: 130, 2024
APA
Ham, J., & Ham, J. (2024). Anti‑osteoclastogenic effect of fermented mealworm extract by inhibiting RANKL‑induced NFATc1 action. Experimental and Therapeutic Medicine, 27, 130. https://doi.org/10.3892/etm.2024.12418
MLA
Ham, J., Lee, M."Anti‑osteoclastogenic effect of fermented mealworm extract by inhibiting RANKL‑induced NFATc1 action". Experimental and Therapeutic Medicine 27.4 (2024): 130.
Chicago
Ham, J., Lee, M."Anti‑osteoclastogenic effect of fermented mealworm extract by inhibiting RANKL‑induced NFATc1 action". Experimental and Therapeutic Medicine 27, no. 4 (2024): 130. https://doi.org/10.3892/etm.2024.12418