Next Article in Journal
Investigation of Synechocystis sp. CPCC 534 Motility during Different Stages of the Growth Period in Active Fluids
Next Article in Special Issue
Synthesis, Molecular Docking, Molecular Dynamics Studies, and In Vitro Biological Evaluation of New Biofunctional Ketoprofen Derivatives with Different N-Containing Heterocycles
Previous Article in Journal
Post-Fire Analysis of Thermally Sprayed Coatings: Evaluating Microstructure, Mechanical Integrity, and Corrosion Behavior
Previous Article in Special Issue
Novel Non-Toxic Highly Antibacterial Chitosan/Fe(III)-Based Nanoparticles That Contain a Deferoxamine—Trojan Horse Ligands: Combined Synthetic and Biological Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Sustainable Bioactive Composite of Glehnia littoralis Extracts for Osteoblast Differentiation and Bone Formation

1
Research Institute of Biotechnology, Hwajin Bio Cosmetic, Chuncheon 24232, Republic of Korea
2
Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Gwangjin, Seoul 05029, Republic of Korea
3
Department of Agricultural Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
4
Department of Bio-Resource Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Processes 2023, 11(5), 1491; https://doi.org/10.3390/pr11051491
Submission received: 20 February 2023 / Revised: 31 March 2023 / Accepted: 10 May 2023 / Published: 15 May 2023

Abstract

:
Different bone-related diseases are mostly caused by the disruption of bone formation and bone resorption, including osteoporosis. Traditional medicinal literature has reported the possible anti-osteoporotic properties of Glehnia littoralis. However, the chemical compounds in extracts that are responsible for bone metabolism are poorly understood. The present study aimed to explore and compare the coumarin-based compounds present in G. littoralis extracts, the antioxidant activities, and the anti-osteoporotic properties of different extracts of G. littoralis (leaf and stem, fruit, whole plant, and root extracts) on bone metabolism. This study analyzed G. littoralis extract effects on the proliferation and osteoblastic differentiation of MC3T3-E1 osteoblasts. Among the different tested samples, stem extracts had the highest scopoletin (53.0 mg/g), and umbelliferone (1.60 mg/g). The significantly (p < 0.05) highest amounts of imperatorin (31.9 mg/g) and phellopterin (2.3 mg/g), were observed in fruit and whole plant extracts, respectively. Furthermore, the results confirmed alkaline phosphatase activity, collagen synthesis, mineralization, osteocalcin content, and osterix and RUNX2 expression. G. littoralis extracts at concentrations greater than 20 µg/mL had particularly adverse effects on MC3T3-E1 cell viability and proliferation. Notably, cell proliferation was significantly elevated at lower G. littoralis concentrations. Comparatively, 0.5 µg/mL stem had a higher osteocalcin content. Of the four extract types, stem showed a higher collagen synthesis effect at concentrations of 0.5–5 µg/mL. Except for fruit extracts, G. littoralis extract treatment significantly elevated osterix gene expression. All G. littoralis extracts increased RUNX2 gene expression. The results described here indicate that G. littoralis ethanolic extracts can effectively prevent osteoporosis.

1. Introduction

Osteoporosis is an osteometabolic disorder caused by an imbalance in bone formation by osteoblasts and bone resorption by osteoclasts that make up bones [1,2], which leads to fragility, increased risk of fractures, and threatens mobility in the elderly [3]. According to the International Osteoporosis Foundation (IOF), approximately 200 million people worldwide suffer from osteoporosis [4,5]. Recent studies suggest that increases in inflammation-related cytokine secretion, the number of macrophages, and leukotriene B4, an inflammation-inducing factor [6,7,8,9], cause this disease. In women, osteoporosis is associated with a sharp decline in estrogen secretion in the postmenopausal stage, known as postmenopausal osteoporosis [10]. Synthetic therapeutic agents, such as parathyroid hormones, bisphosphonates, selective estrogen receptor modulators (SERMs), and hormone replacement therapy (HRT), are used to treat osteoporosis [11]. However, these drugs can have side effects, including hypercalcemia and osteosarcoma in postmenopausal women [12], esophageal gastric irritation, and cancer [13], and they can increase the chance of strokes, breast cancer, and coronary heart diseases [14]. Thus, it would be beneficial to discover natural anti-osteoporotic agents that minimize bone loss in postmenopausal women.
Osteoblast cells are critical in bone metabolism and are responsible for bone matrix synthesis and mineralization [15]. Cell culture is widely used to assess the activity of a substance in vitro, such as its osteoinductive potential. In particular, MC3T3 pre-osteoblasts have methodological advantages and facilities [16,17], meaning the differentiation of these cells into mature osteoblasts can be easily recognized by markers of osteoblastic metabolism, such as alkaline phosphatase (ALP), and by the degree of extracellular matrix (ECM) mineralization [18]. ALP is the most widely used biochemical marker for estimating osteoblastic activity [19]. This enzyme is associated with the ECM mineralization process and osteoblastic differentiation, as it is responsible for the maturation of the matrix, which will later be mineralized [18,20].
Current research focuses on natural materials and phytoestrogens for bone formation and resorption pathways related to bone metabolism. There is a constant search for alternatives that can help bone healing, in cases of injury, or favor rehabilitating the quality of the bone tissue. Several recent reports have indicated that phytoestrogen compounds in food and plants can effectively suppress the secretion of inflammation-inducing factors and inflammation-related cytokines associated with osteoporosis [21,22], and enhance or stimulate osteoblast activity [23]. Phytoestrogens act like estrogen and exist in the form of flavonoids, lignins, isoflavones, and coumestans, which share structural and functional similarities with synthetic estrogens [24,25]. The present study aimed to research therapeutic alternatives to treat osteoporosis based on drugs obtained from natural sources, mainly by observing their lower cost and incidence of adverse effects when compared to synthetic drugs.
Glehnia littoralis Fr. Schmidt et Miquel, a perennial marine herbaceous plant belonging to the Apiaceae family, is native to the sandy coastal area of eastern Asia, mainly Japan, Korea, China, Manchuria, Sakhalin, Okhotsk, the Kuril Islands, and North America from California to Alaska [26,27]. The leaves and flower buds are edible, their rhizomes and roots are used traditionally to treat lung diseases, tuberculosis, coughs, hemoptysis, and dyspnea [28,29], and they have diaphoretic, antipyretic, and analgesic effects [30,31]. Previous studies have reported the presence of phytochemicals such as phenolic acids, flavonoids, pyranocoumarins, and polysaccharides in G. littoralis extracts, which have various biological activities, including antitumor, antimicrobial, antioxidant, blood circulation-promoting and immunomodulatory properties [29]. Moreover, several previous studies have shown that these phytochemicals are effective in bone formation [32]. To our knowledge, no studies have reported the use of G. littoralis extracts as potential natural therapeutic agents to prevent osteoporosis.
Therefore, the main objectives of the present study were to identify and select the toxicity of different G. littoralis plant parts for use in the differentiation analysis of an MC3T3-E1 cell culture through a cell viability assay. In addition, this study investigated the proliferative potential of MC3T3-E1 cells treated with G. littoralis using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. This study also compared the degree of osteoblastic differentiation between control cells and those treated with different G. littoralis concentrations by quantifying ALP. Finally, we evaluated the degree of matrix mineralization formed by MC3T3-E1 cells treated with different G. littoralis plant parts and concentrations to confirm its applicability as a natural material for osteoblast differentiation and bone formation.

2. Materials and Methods

2.1. Chemicals

All the chemicals and solvents used in the experiments were of analytical grade. All chemicals were obtained from Sigma-Aldrich (St. Louis, MO, USA).

2.2. Plant Material and Extract Preparation

The G. littoralis used in the experiment was grown and harvested from a field in Gangneung-si, Gangwon-do (Slonaemall, Gangneung, Republic of Korea) at 37°45′06″ N latitude and 128°52′38″ E longitude, in October 2020. The leaves, roots, fruits, and stems of the G. littoralis were washed with purified water and separated. The collected samples were dried at room temperature for 72 h. Approximately 2 g of the finely ground samples and placed in a conical flask containing 40 mL of 80% (v/v) ethanol. The mixture was filtered through filter paper to remove debris, and the solvents were evaporated at 41 °C in a rotary vacuum evaporator (Eyela, SB-1300, Shanghai Eyela Co., Ltd., Shanghai, China) and then lyophilized using a freeze dryer (PVTFD 300R) (IlShinBioBase, Yangju, Republic of Korea). The collected extracts were mixed with 80% methanol and stored in a refrigerator at 4 °C until further analysis.

2.3. Determination of Total Phenolic Content (TPC)

The total phenolic content of different samples was measured by following the Folin-Ciocalteu procedure, the method Singleton and Rossi [33] described previously. An aliquot of 100 µL of the plant extract (at a concentration of 1 mg mL−1) and 500 µL of the Folin-Ciocalteu reagent (1:3 v/v) were mixed with 500 µL distilled water in a test tube and shaken for 5 min at room temperature. Then, 500 µL sodium carbonate (10%) was added to the solution, and the mixture was left to rest for 1 h. Deionized distilled water served as a blank. Then, the absorbance of the obtained mixture was taken using a ultraviolet-visible (UV-Vis) spectrophotometer (Jasco V530 UV-VIS spectrophotometer, Tokyo, Japan) at 725 nm against the blank. The results are expressed as mg of the gallic acid equivalent (GAE) per g of the dry weight (DW). A calibration curve was prepared using 20–500 mg/L gallic acid (R2 = 0.9980).

2.4. Determination of Total Flavonoid Content (TFC)

Total flavonoid content was measured following the method described by Moreno et al. [34]. An aliquot of 500 µL of plant extract (at a concentration of 1 mg mL−1) was mixed with 100 µL KCH3COO (1 M) and 100 µL of 10% Al(NO3)3 in a 10 mL test tube and homogenized manually, and incubated for 50 min at room temperature. Then, the absorbance was measured using a spectrophotometer (Jasco V530 UV-VIS spectrophotometer, Tokyo, Japan) at 415 nm against the blank. The experiment was carried out in triplicate, and the results are expressed as the mean ± standard deviation in mg of the quercetin equivalent (QE) per g of the dry sample. A calibration curve was prepared using 20–500 mg/L quercetin (R2 = 0.9970).

2.5. LC/UVD Quantitative Analysis of Coumarin-Based Compounds

Quantitative analysis of coumarin-based compounds was performed by using LC/UVD with various solvents. An UltiMate 3000 HPLC system (Thermo Fisher Scientific Lin., San Jose, CA, USA) coupled with a UV detector (Thermo Fisher Scientific, San Jose, CA, USA) was applied for quantitative analysis of coumarin-based compounds. The separation of each compound was achieved by a column (5 μm, 250 mm × 4.6 mm, Bischoff Analysentechnik und-geräte GmbH., Leonberg, Garmany). The mobile phase comprised water (A) and acetonitrile (B). The gradient elution conditions were: 0–5 min, 80% A; 5–60 min, 80–0% A; 60–70 min, 0% A; 70–71 min, 0–80% A; 71–80 min, 80% A. The temperature of the column oven was maintained at 30 °C. A flow rate of 1.0 mL/min was used and the injection volume was 10 μL. The chromatograms of the compounds were acquired at 203 nm. Standard chemical compounds such as scopoletin, umbelliferone, phellopterin, and imperatorin were obtained from Sigma-Aldrich (St. Louis, MO, USA). Stock standard chemicals (1000 ng/mL) were prepared in 100% methanol. The stock solutions were maintained at 4 °C and used to construct calibration curves after appropriate dilution. Amounts of different quantified compounds were calculated as mean values from HPLC analyses based on the calibration curves of the corresponding standard compounds. The regression equation of the standard compounds were as follows.
Scopoletin, y = 0.482x + 0.077, R2 = 0.999
Umbelliferone, y = 1.005x + 0.027, R2 = 0.999
Imperatorin, y = 0.996x + 0.017, R2 = 0.999
Phellopterin, y = 0.993x + 0.010, R2 = 0.999

2.6. Antioxidant Activity

2.6.1. Evaluating the 2,2-Diphenyl-1-picrylhydrazyl (DPPH) Radical Scavenging Assay

The free radical scavenging capacity of G. littoralis samples was measured using a DPPH radical scavenging assay, following the method Chung et al. [35] described previously. In triplicate, an aliquot of 200 µL of different sample extracts was mixed with 4.5 mL DPPH (0.004% in methanol). The reaction mixture was homogenized manually and incubated at room temperature (25 °C) for 40 min. The mixture was shaken and kept in dark conditions for 45 min. Then, the absorbance value was taken using a spectrophotometer (Jasco V530 UV-VIS spectrophotometer, Tokyo, Japan) at 517 nm. A blank was prepared by replacing DPPH with 80% methanol in the reaction medium. BHT was used as the positive control.
The free radical scavenging ability of the sample was measured from the following equation:
DPPH scavenging activity (%) = (Abscontrol − Abssample)/Abscontrol × 100%
where Abscontrol represent the absorbance of the mixture + methanol, and Abssample represent the absorbance of the mixture + plant extract. The antioxidant activity was expressed as the capacity to scavenge or reduce the DPPH radical by 50%; that is, the amount of antioxidant compounds required to scavenge or reduce the initial concentration of DPPH by 50%.

2.6.2. Evaluation of the 2,2′-Azino-bis(3-ethylbenzothiazoline-6-sulfonic Acid (ABTS) Assay

The antioxidant activity was determined using the ABTS radical method described elsewhere [35]. Briefly, the ABTS solution used in the experiments was formed by mixing 7.4 mM ABTS and 2.6 mM potassium persulphate (1:1, v/v). The reaction solution was incubated for 12 h at room temperature. The solution was then diluted with 80% methanol until a solution with an absorbance of 0.70 ± 0.01 was achieved. Then, 1 mL diluted ABTS was mixed with 100 mL of the sample. Then, the absorbance was taken using a spectrophotometer (Jasco V530 UV-VIS spectrophotometer, Tokyo, Japan) at 734 nm. Trolox was used as a positive control. The standard curve was prepared from various concentration of trolox (500 µM, 600 µM, 700 µM, 800 µM, 900 µM, and 1000 µM).
The ABTS radical scavenging ability of the samples was measured from the following equation:
ABTS scavenging activity = (Abscontrol − Abssample)/Abscontrol × 100
where Abscontrol represent the absorbance of the ABTS solution + methanol, and Abssample represent the absorbance of the ABTS solution + test sample.

2.7. Cell Culture

Osteoblast MC3T3-E1 cells derived from mouse bones were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). Osteoblast MC3T3-E1 cells were prepared in α-minimum essential medium (α-MEM, Gibco, Grand Island, NE, USA) with 10% fetal bovine serum (FBS; Gibco, Grand Island, NE, USA), 100 U/mL penicillin (CO2 incubator (MCO-230AIC-PK, Panasonic, Kadoma, Japan) using a culture medium supplemented with Gibco (Grand Island, NE, USA)), and 100 U/mL streptomycin (Gibco, Grand Island, NE, USA) at 37 °C, 5% CO2, and 95% air. The cells were washed with phosphate-buffered saline (PBS; pH 7.4; Gibco, Grand Island, NE, USA) and 0.25% trypsin-2.65 mM EDTA (Gibco, Grand Island, NE, USA), and sub-cultured by changing the culture medium every two days.

2.8. Cell Viability

Cell viability was analyzed according to the protocol described by Denizot and Land [36] by dispensing osteoblast MC3T3-E1 cells in a 96-well plate at 2 × 103 cells/well and subsequently pre-incubating them in a CO2 incubator for 24 h. After incubation, the MC3T3-E1 cells were treated with different concentrations of plant sample for 72 h. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT solution) at 5 mg·mL−1 was added to each well and maintained at 37 °C for 4 h. Then, the resultant formazan crystals were dissolved in dimethyl sulfoxide (DMSO). The absorbance of the mixture was taken using a microplate reader (Thermo Fisher Scientific Instrument Co., Ltd., Shanghai, China) at 570 nm.

2.9. Cytotoxicity

The cytotoxicity of different plant parts was evaluated according to a previously described protocol [37,38]. Osteoblast MC3T3-E1 cells were dispensed in a 96-well plate at a density of 2 × 103 cells/mL, then pre-incubated in a CO2 incubator for 24 h. After incubation, the cells were treated with various concentrations of samples (ranging from 0 to200 µg/mL) and incubated for 48 h. The cell culture solution (10 µL) was added to a 96-well plate containing 40 μL PBS. After placing the LDH reagent in each well, the mixture was placed in a dark place for 45 min at 25 °C. After the reaction was terminated by mixing 50 μL of stop solution, the cytotoxicity was evaluated by quantifying the plasma membrane damage. LDH is a stable enzyme present in all cell types and is rapidly released into the culture medium when the plasma membrane is damaged. Cell membrane integrity was evaluated by measuring the LDH leakage levels from the cells using the LDH Cytotoxicity Assay Kit (BioVision, Inc., Milpitas, CA, USA). The absorbance value of the samples was measured using a microplate reader (Thermo Fisher Scientific Instrument Co., Ltd., Shanghai, China) at 492 nm.

2.10. ALP Activity

MC3T3-E1 cells were seeded as described above, and the ALP activity of the media was measured by following the method described by Liu et al. [39]. Initially, MC3T3-E1 cells were dispensed in a 48-well plate at 5 × 104 cells/well and pre-incubated in a CO2 incubator for 24 h. Then, the MC3T3-E1 cells were treated with different sample concentrations (0–20 μg/mL). Subsequently, the culture medium was replaced and supplemented with 100 μL chemiluminescent substrate for alkaline phosphatase (CSPD; Roche, Basel, Switzerland), added to 20 μL total cell lysate, and reacted for 30 min. The amount of protein in the total cell lysate was measured using CSPD (Life Technologies, Carlsbad, CA, USA) according to the Protein Assay Kit (Bio-Rad, Hercules, CA, USA), and the ALP activity value was expressed as the fold change per µg of total protein.

2.11. Collagen Synthesis Rate

Collagen content was quantified by a Sirius Red-based colorimetric assay as described by Park et al. [40]. The MC3T3-E1 cells were cultured in an osteogenic medium containing 10 mM β-glycerophosphate, 5 nM dexamethasone, 50 g/mL ascorbic acid, and G. littoralis (0 μg/mL and 20 μg/mL). After seven days, the cells were cultured in a medium containing bovine serum albumin (BSA) (3%) for two days. The cultured cells were gently washed twice with PBS, followed by fixation with Bouin’s fluid (8.3% formaldehyde and 4.8% acetic acid in saturated aqueous picric acid, (O2N)3C6H2OH) for 1 h. After fixation, the fixative fluids were removed, and the cultured cells were washed with tap water for 10 min. The cultured cells were air-dried and stained with Sirius red dye (0.1% saturated picric acid) for 1 h on a shaker. The stained cells were washed with PBS and observed under a microscope. For quantitative analysis, the stained cell layer was washed extensively with 0.01 N HCl to remove the non-bound dye. The stained cells were dissolved in 0.2 mL of 0.1 N and shaken for 30 min. Then, the absorbance was measured at 540 nm.

2.12. Mineralization Content

Calcium content was measured in osteoblasts using the method described by Zakłos-Szyda et al. [41]. Osteoblast MC3T3-E1 cells were aliquoted in a 24-well plate at 2 × 104 cells/well in a CO2 incubator at 37 °C. The cells were treated with plant samples at a concentration of 0–20 μg/mL and placed in an incubator for seven and 14 days to induce mineral deposition. The culture medium was changed at three-day intervals. For harvesting, the MC3T3-E1 cells were washed with PBS twice and fixed with 4% paraformaldehyde for 2 h. The cells were then stained with 40 mM Alizarin Red S (pH 4.5). The stained cells were then rinsed four times with distilled water and observed under an inverted microscope. Calcified nodules appearing bright red were confirmed, and 100 mM cetylpyridinium chloride (Sigma) solution was added to each well to elute the stain. The eluted stain (100 μL) was added to a 96-well microplate. The absorbance of solubilized calcium-bound Alizarin Red S was taken at 570 nm using a spectrophotometer (Eppendorf AG 22331; Hamburg, Germany). Calcium deposition was expressed as the molar equivalent of calcium.

2.13. Osteocalcin Content

Osteocalcin concentration was determined by the test method of the OCN ELISA kit (Takara Bio, Kusatsu, Japan) by following the process described by Bukhari et al. [42]. Initially, 100 μL of the cell culture solution was added to a 96-well plate coated with antibodies and incubated for 1 h at 37 °C. After removing unbound material with washing buffer (50 mM Tris, 200 mM NaCl, and 0.2% Tween 20), horseradish peroxidase (HRP)-conjugated streptavidin was added to each well and incubated at 37 °C for 1 h to bind to the antibodies. After washing the wells five times, tetramethylbenzidine (TMB) solution was added to each well and incubated at room temperature for 20 min in the dark. HRP catalyzed the conversion of a chromogenic substrate (TMB) to a colored solution, with a color intensity proportional to the amount of protein present in the sample. After adding the stop solution, the absorbance of the sample in each well was measured at 450 nm. Results are presented as the percentage change in activity compared to the untreated control. The concentration of osteocalcin in the serum was measured using the same method as that used for cultured cell samples.

2.14. mRNA Expression Rate

Total RNA was isolated from cells at specific times using TRIzol reagent (Invitrogen, Waltham, MA, USA) with DNase treated with RNase-free DNase (35 U/mL) for 50 min at 37 °C according to the method described by Liu et al. [43] and Matsubara et al. [44]. The total RNA present in each sample was quantified by measuring the absorbance at 260 nm using a spectrophotometer (Eppendorf AG 22331; Hamburg, Germany). Gene expression was measured by adding cDNA to a PCR mixture containing EXPRESS SYBR Green qPCR Supermix (Bio Prince, Seongsu, Seoul, Republic of Korea). Real-time PCR was performed using the Rotor-Gene Q (Qiagen, Düsseldorf, Germany). The reaction was carried out at 95 °C for 20 s, 60 °C for 20 s, and 72 °C for 25 s for 40 cycles of amplification. Relative mRNA expression of specific genes was standardized with glyceraldehyde 3-phosphate dehydrogenase (GAPDH), and sequencing was performed using PCR primer sequences (Table 1).

2.15. Statistical Processing

The results were presented as the mean ± standard deviations of three trials independent (n=3). The data was compared by one-way ANOVA using the SPSS program (Statistical Package for Social Science, Version 24), followed by Duncan’s multiple range test, considering significant differences at P values < 0.05.

3. Results

3.1. The Total Phenolic Contents (TPCs) and Total Flavonoid Contents (TFCs)

The total phenolic contents (TPCs) and total flavonoid contents (TFCs) of stems, leaves, roots, and whole plant extracts of G. littoralis extracts were calculated from the linear regression equation of the gallic acid standard calibration curve. The TPC of different plant parts ranged from 4.27 ± 0.03 to 23.29 ± 0.43 mgGAE/g dry sample. As Figure 1 shows, the leaf and stem (GLSE) and root (GLRE) extracts had the highest and lowest TPCs, respectively. The total flavonoid contents (TFCs) of stems, leaves, roots, and whole plant extracts of G. littoralis extracts were determined using colorimetric analysis. The flavonoid content of different G. littoralis extract parts ranged from 0.28 ± 0.04 to 4.93 ± 0.30 mgQE/g dry sample. The results showed that fruit (GLFE) extracts contained a higher TFC, followed by GLFE, whole plant (GLAE), and GLRE extracts.

3.2. LC/UVD Quantitative Analysis of Coumarin-Based Compounds

LC/UVD quantitative analysis of four coumarin components varied widely with the leaf, stem, fruit, whole plant, and root extracts of G. littoralis (Table 2). Among the different tested samples, GLSE extracts had the highest scopoletin (53.0 mg/g), and umbelliferone (1.6 mg/g). The lowest amount of scopoletin was observed in GLRE (8.5 mg/g), and of umbelliferone, in GLFE (0.8 mg/g). The significantly (p < 0.05) highest amount of imperatorin (31.9 mg/g) and phellopterin (2.3 mg/g) were observed in GLFE and GLAE extracts, respectively (Table 2). It was confirmed that the total content of four types of coumarin was higher in the order of GLSE, GLRE, GLAE, and GLFE (Figure 2).

3.3. Antioxidant Activity

Figure 3 presents the radical scavenging potential of the GLSE, GLFE, GLAE, and GLRE extracts. The results showed that the antioxidant activity of different plant parts varied significantly and appeared in a concentration-dependent manner. Likewise, GLAE extracts had higher antioxidant properties, as represented by the lower half-maximal inhibitory concentration (IC50) (718.40 ± 14.025 µg mL−1). Furthermore, all other plant part extracts, except GLRE extracts, possessed a better radical scavenging potential, indicating the presence of higher amounts of antioxidant compounds. The results showed that the ABTS+ radical scavenging potential of different G. littoralis extract parts varied significantly (p < 0.05), with GLAE extracts exhibiting higher ABTS radical scavenging activity (16,348.41 ± 1315.26 µg mL−1). Comparatively, the GLRE extract showed the lowest antioxidant activity (Figure 3). Two different antioxidant assays were carried out, and these antioxidant assays were positively and significantly correlated with TPC and TFC of G. littoralis (Table S1).

3.4. Cell Viability Test

We used osteoblastic precursor cell lines (MC3T3-E1 cells) derived from Mus musculus to induce the expression of osteoblast markers and investigate the effect of G. littoralis on bone metabolism. In this study, we attempted to determine the effect of G. littoralis extract on in vitro osteogenic induction using MC3T3-E1 cells. We performed a pilot study in which G. littoralis extract concentrations varied from 0.5 to 200 μg/mL to determine the optimal concentration. Cell viability studies were performed for different G. littoralis parts, including GLSE, GLFE, GLAE, and GLRE extracts. Initially, the purified G. littoralis ethanol extracts were re-suspended in 70% ethanol, and various plant extract concentrations were added to the MC3T3-E1 cell culture. The viability of cells varied considerably among the different plant parts used. The viable cell number increased in up to 5 μg/mL of G. littoralis treatment. Then, the higher concentrations showed a significant decrease in the cell population in a concentration-dependent manner (Figure 4), indicating G. littoralis’s cellular toxicity properties. Lower concentrations of the plant extracts (between 0 and 2 μg/mL) did not significantly affect the cell viability. Among the different extraction samples, GLSE at a concentration of 0.5–10 μg/mL showed 88.5 ± 1.0% to 96.6 ± 3.9% cellular viability, while these values varied in GLFE (90.7 ± 1.2% to 94.5 ± 0.9%), GLAE (85.8 ± 1.6% to 95.4 ± 1.2%), and GLRE (80.0 ± 0.7% to 97.7 ± 0.4%) at the same concentrations (Figure 4). In addition, the lactate dehydrogenase (LDH) cytotoxicity assay proved that plant extracts beyond 5 μg/mL were toxic to the cell lines (Figure 5). We used 5 μg/mL of G. littoralis extract in the subsequent experiments to avoid cytotoxicity and promote MC3T3-E1 cell growth.

3.5. ALP Activity

We evaluated ALP activity to assess how G. littoralis extracts affect osteogenic induction (Figure 6). All the sample extracts (GLSE, GLAE, GLRE, and GLFE) significantly increased ALP activity in MC3T3-E1 cells. Comparatively, higher ALP activities were observed at sample concentrations of 0.5 μg/mL. Increasing the treated sample concentration resulted in a decrease in ALP activity. Among the treated samples, GLSE at a concentration of 0.5–20 μg/mL showed higher ALP activity ranging from 158.4 ± 7.9% to 125.9 ± 11.5%, respectively, while the lowest ALP activity was observed in GLFE at a concentration of 0.5–20 μg/mL ranging from 127.4 ± 2.9% to 84.1 ± 3.7%, respectively. The results indicate that G. littoralis extract enhanced the ALP activity required for osteoblast formation and ECM mineralization in MC3T3-E1 cells.

3.6. Collagen Synthesis Rate

GLSE, at a concentration of 0.5–5 μg/mL, showed a higher collagen synthesis effect than the other extract types (Figure 7). The results indicate that the GLFE methanolic extract had phytochemicals that inhibited the collagen synthesis effect. As Figure 1 shows, GLFE extracts showed lower collagen synthesis effects at concentrations ranging from 1 to 20 μg/mL, which decreased in a concentration-dependent manner. GLFE extract concentrations higher than 2 μg/mL did not affect collagen synthesis. However, no significant difference was observed between GLAE and GLRE extracts in collagen synthesis at any of the concentrations used in the experiment.

3.7. GL Extracts Enhanced Osteoblast Mineralization

After seven days of treatment with G. littoralis extracts, we measured the mineralization content in MC3T3-E1 cells. Among the different samples, GLAE showed a higher mineral content (131.9 ± 4.7%), followed by GLSE (130.8 ± 2.9%), GLRE (127.8 ± 3.4%), and GLFE (119.9 ± 5.6%), at a concentration of 0.5 μg/mL. G. littoralis extracts at a concentration of 0.5 μg/mL induced higher mineralization levels in all the groups compared to 20 μg/mL treatments (Figure 8). GLFE showed lower mineralization and decreased in a concentration-dependent manner. GLAE showed higher mineralization at all concentrations used (0.5–20 μg/mL), indicating that the whole plant extracts contain more bioactive compounds responsible for osteoblast formation.

3.8. Osteocalcin Content

We investigated the effects of G. littoralis extracts on the degree of osteocalcin production during the late stage of osteoblast differentiation. As Figure 9 shows, G. littoralis extract significantly increased osteocalcin production in MC3T3-E1 cells. This is the first report describing the inhibitory osteoporotic properties of G. littoralis using the MC3T3-E1 in vitro system. Osteocalcin content levels varied with different concentrations of the tested plant parts. Except in the case of GLAE, an increase in the sample concentration resulted in reduced osteocalcin content. Comparatively, 0.5 μg/mL GLSE resulted in a higher osteocalcin content, indicating that the phytochemical responsible for producing the protein in osteoblast cells is more present in this extract. However, increasing the GLFE concentration resulted in a decrease in osteocalcin content in a concentration-dependent manner. Comparatively, 20 μg/mL GLFE displayed lower osteocalcin production than the negative control, indicating that a higher GLFE concentration is cytotoxic and inhibits osteoblast formation.

3.9. mRNA Expression Rate

We confirmed how G. littoralis extracts affect osteoblast differentiation by analyzing the expression patterns of prominent osteoblast marker genes Table 3. After seven days of treatment with different G. littoralis extracts, a real-time polymerase chain reaction (PCR) was performed to investigate the effect on osterix and RUNX2 mRNA expression. As the results show, the expression patterns of both osteoblastic genes changed when treated with the G. littoralis extracts. The findings showed that, except in the case of GLFE extracts, G. littoralis extract treatment significantly elevated osterix gene expression, and RUNX2 gene expression was increased by all G. littoralis extracts. Comparatively, RUNX2 expression significantly increased after treatment with lower G. littoralis extract concentrations (ranging from 0.5 to 2 μg/mL).

4. Discussion

Plants contain various antioxidants; these antioxidants play an important role in protecting plants from oxidative stress and signals, when ingested, and they can act as natural antioxidants to help prevent disease [45]. Among these compounds, the phenolic compounds present in the plants are mostly responsible for the antioxidant properties [46]. In the present study, the DPPH and ABTS radical scavenging assays presented wide variations in antioxidant activity values for different plant parts. We studied the relationship between the different parameters by obtaining Pearson’s correlation coefficients. The antioxidant potentials estimated by both assays were somewhat different and showed a high correlation between them (p < 0.05, r = 0.894). The variation in antioxidant potential could be due to the various antioxidant compound responses to the different radicals present in each assay. Excessive reactive oxygen species (ROS) production can cause lipid and protein oxidation, damage DNA integrity, and simultaneously cause tissue damage [47]. Several previous studies have shown the involvement of ROS in bone remodeling by enhancing osteoclastic bone resorption and decreasing osteoblast cell formation [48,49]. In the present study, all G. littoralis extracts effectively scavenged the DPPH and ABTS radicals, indicating that the G. littoralis extracts had potential antioxidant activity and protected the MC3T3-E1 cells from degeneration and death. Moreover, Pearson’s correlation analysis revealed a significant correlation between DPPH and TPC and TFC in G. littoralis extracts. A similar correlation was also observed between ABTS and TFC. Phenolic compounds, such as coumarins and their derivatives, have been reported as dominant G. littoralis phytochemical components [48,49]. In G. littoralis extracts, phenolic compounds, including caffeic acid, vanillic acid, ferulic acid, chlorogenic acid, rutin, quercetin, kaempferol, and coumarins and their derivatives, have been identified [50,51,52]. In the present study, coumarin-based flavonoids such as scopoletin, umbelliferone, imperatorin, phellopterin have been detected in G. littoralis extracts. These compounds are mostly responsible for the antioxidant properties. For instance, scopoletin has been involved in considerable antioxidant activities by scavenging ROS, especially hydrogen peroxide (H2O2) scavenging activity, ferrous ion (Fe2+) chelating activity, and activity against superoxide anion radicals (O2•−), and OH-radicals [53]. Moreover, Um et al. [53] isolated scopoletin and umbelliferone from Glehnia littoralis and demonstrated a reactive oxygen species (ROS) scavenging ability of about 60% or more compared to a control. Imperatorin (IMP) has been reported in several plants with antioxidant properties [54,55,56,57]. Methanolic extracts containing umbelliferone have been shown to exhibit an efficient pro-oxidant activity [58] and inhibit lipid peroxidation [59]. Others observed that the treatment of umbelliferone has been shown to inhibit the intracellular ROS production in irradiated lymphocytes and effectively restore the mitochondrial membrane and inhibited gamma radiation-induced DNA damage [60].
Numerous studies observed that oxidative stress enhances the differentiation and function of osteoclasts [61]. ROS-induced oxidative stress has been shown to involve the suppression of bone formation and the stimulation of osteoclast resorption [62]. Present results indicate that phenolic compounds such as scopoletin, umbelliferone, imperatorin, and phellopterin present in the G. littoralis extracts inhibited ROS formation to suppress the excessive bone breakdown by osteoclasts. Moreover, several studies have reported the anti-osteoporotic properties of coumarins by suppressing the interaction of advanced glycation end-products (AGE) and their receptors [63]. Treatment using scopoletin prevented bone loss in diabetic mice by increasing bone turnover of bone-degrading osteoclasts and bone-forming osteoblasts. It has been shown that treatment with imperatorin in rats promoted osteogenesis and suppressed the osteoclast differentiation [64]. The authors found that the imperatorin activates AKT that leads to the inactivation of GSK3β that causes the activation of β-catenin and accumulation of β-catenin in the nucleus [65,66,67,68]. It was believed that the activation of β-catenin plays an important role in the suppression of osteoblast differentiation [69]. Thus, it can be inferred that imperatorin could induce osteogenesis via the AKT/GSK3β/β-catenin pathway [64], indicating that imperatorin present in the GL extracts could be responsible for bone growth and inhibition of resorption. Furthermore, umbelliferone prevented trabecular bone matrix degradation and osteoclast formation in bone tissue [70]. The authors reported that umbelliferone is closely associated with the dysfunction of osteoclasts attributed to defects in osteoclast survival and differentiation [65]. In addition, Li et al. [71] reported that phellopterin inhibits Ca2+ influx induced by the stimulation of voltage-gated and receptor-dependent calcium channels [72,73]. Therefore, in the present study, these compounds, together with the other polyphenols present in the G. littoralis extracts, strongly favored MC3T3-E1 cell differentiation. Furthermore, it can be suggested that G. littoralis effectively contributes to the prevention of oxidative damage to bone tissues via antioxidant action and its phytochemicals.
ALP, a typical protein product, is associated with osteoblast growth and differentiation and is expressed and increased during the active matrix maturation phase immediately after the cell proliferation period [18,20,74]. Although the exact ALP mechanism of action is poorly understood, it is believed that these enzymes are responsible for bone mineralization [75]. Therefore, it is important to examine the effect of G. littoralis extracts on MS3T3-E1 cell ALP activity. In the present study, all G. littoralis extracts effectively accelerated ALP activity in a dose-dependent manner. Moreover, some G. littoralis extracts showed higher ALP activity than the positive control, indicating that the different phytochemicals present in the extracts may be necessary for osteoblast differentiation. We hypothesize that G. littoralis extracts are associated with osteoblast proliferation and differentiation from a newly synthesized protein component.
In this study, maximum ALP activity was observed at the lowest G. littoralis extract concentration (0.5 μg/mL), which was confirmed in GLSE and GLAE extracts. Similar to our findings, the aqueous extracts of rooibos promoted ALP activity and mineralization [76]. Moreover, there is abundant evidence that dietary phytochemicals have osteoprotective effects. Caffeic acid regulates bone remodeling by inhibiting osteoclastogenesis, bone resorption, and osteoblast apoptosis [77]. Chlorogenic acid extracted from Cortex Eucommiae inhibited a decrease in bone mineral density [78]. Quercetin inhibits osteoblast apoptosis, osteoclastogenesis, and oxidative stress [79]. Jang et al. [80] reported similar results in A. rugosa, stating that some of the phenolic compounds present in A. rugosa effectively suppressed osteoclasts [81,82]. Flavonoids, such as orientin, quercetin, and luteolin, have shown blastogenic effects by increasing ALP activity and mineralization in rooibos [83,84]. Treating osteoblast cells with various phenolic compounds increases ALP synthesis and decreases the expression of antigens involved in osteoblast immune functions, which may improve bone mineral density [85]. Flavonoids, such as icariin and naringin, were found to regenerate bone tissues by increasing ALP activity and osteopontin content [86,87,88,89]. Another study observed increases in osteoblast proliferation, and several other reports have provided convincing data about phytochemicals and their association with osteoclast formation in vitro [90]. Although this study did not determine the exact composition of phenolic compounds in G. littoralis, the phenolic compounds from G. littoralis could be crucial in modulating the bone formation process through the osteoblast formation process and ALP production.
It has been reported that osteoblasts produce biochemical markers, such as type I collagen, ALP, and osteopontin, which are important components for matrix maturation and mineralization [74]. In the present study, phenotypic markers, such as collagen and osteocalcin, mainly associated with the later stage of osteoblast differentiation and were elevated in the MS3T3-E1 cells treated with G. littoralis extract. This indicated that G. littoralis extracts were vital in osteoblast differentiation. GLSE showed greater collagen synthesis and upregulated osteoblastic MC3T3-E1 cell proliferation and differentiation by enhancing ALP activity and mineralization compared to the other extract types. There is increasing interest in both in vitro and in vivo research that phenolic compounds may favorably improve osteoporosis. Sparse experimental data show that phenolic acids may have in vitro estrogenic activity. Bioactive compounds, such as β-estradiol, reportedly significantly increase osteoblastic cell proliferation, DNA and protein content, and ALP activity [91]. Phenolic acids may act on osteoblasts by binding to their estrogen receptors, found in osteoblastic cells [92].
Bone mineralization refers to the deposition of calcium and minerals in cells. It acts as a reservoir for calcium and phosphorus in the bone, maintains bone elasticity and flexibility, and provides compactness and mechanical resistance to the bone [93]. During the postmenopausal period, estrogen deficiency causes a decrease in the absorption of micronutrients in the body [94]. In the present study, different GL extracts showed various degrees of elevation in calcium levels, possibly due to affecting calcium absorption and contributing to matrix deposition during osteogenesis [95]. In this test, the maximum mineralization content was observed at the lowest concentration (0.5 μg/mL) of GLSE, GLFE, and GLRE extracts. Our results are consistent with those reported by Yun et al. [96], who observed an increase in calcium deposition in osteoblast MC3T3-E1 cells treated with lower concentrations of chrysanthemum extract. Prak et al. [97] reported similar results in 10 μg/mL of seaweed extracts. Osteocalcin is a non-collagenous protein in the bone secreted into osteoblasts and used as a biochemical marker for bone formation [98]. Osteocalcin is associated with changes in bone turnover rate in bone metabolism and is reflected in the rate of bone formation. The osteocalcin carboxyl group is removed and released into the circulation due to pH acidification of the bone when osteoclasts resorbed it [99].
Several transcription factors are involved in osteoblast differentiation. Most importantly, RUNX2 and osterix are genes that differentiate mesenchymal stem cells into immature osteoblasts and are osteoblast-specific transcription factors required for osteoblast differentiation and bone formation [100]. RUNX2, the earliest identifiable marker, is known as “a master gene” for osteoblast differentiation and is associated with ALP and osterix upregulation [101,102]. It has been argued that RUNX2 triggers osteocalcin expression by binding to the cis-acting elements of the osteocalcin promoter region of osteogenic genes to initiate the expression of ALP, osteopontin, bone sialoprotein (BSP), and osteocalcin [103,104]. Osterix is an osteoblast-specific transcription factor containing a zinc finger. It maintains strong expression in mesenchymal cells and the periosteum and is expressed in cells, such as chondrocytes and the bone matrix [105].
In the present study, we determined the gene expression patterns of osteoblast differentiation markers to understand how G. littoralis extracts induce mineralization. The results showed that G. littoralis extract treatments significantly elevated osterix and RUNX2 gene expression and enhanced the production of proteins involved in osteoblast production, such as type 1 collagen and osteocalcin. RUNX2 gene upregulation by the cells and their ALP activity and mineralization have also been reported in Davallia formosana extracts [106]. Previous studies have shown that phenolic compounds of different plant species extracts can induce the proliferative capacity and maturation of osteoblastic cells by improving ALP activity and increasing calcium ion deposition in the ECM [107,108]. It has been reported that changes in osteoblastic cell activity by phenolic compounds occur through the modulation of different transcription factors, such as osterix, osteocalcin, and bone morphogenic proteins (BMPs), by activating the genes involved [108]. The phenolic compounds of various plant species induce osteoblast cell differentiation through the expression of osterix and RUNX2 markers, which are associated with bone maturation [109,110,111,112,113]. In another study, daidzein, present in soy, acted as a phytoestrogen via osteoblast proliferation and differentiation by activating the BMP/Smad signaling pathway [114]. In the present study, all the GL extracts showed higher expression levels of mRNA expression rate of RUNX2 than control. Furthermore, it has been shown that imperatorin promotes the maturation and differentiation of osteoblast by increasing the expression of RUNX2; thus, it is closely associated with early stage osteogenic differentiation [115,116,117]. In the present study, all G. littoralis extracts increased RUNX2 gene expression. The results described here indicate that G. littoralis ethanolic extracts can effectively prevent osteoporosis. These results indicate that the phenolic compounds in GL extracts may synergistically induce osteoblastic cell proliferation to a greater extent than a single compound. Moreover, the results further suggest that phytochemicals other than phenolic compounds may be present in the G. littoralis extracts, causing the osteoblastic proliferation of MC3T3-E1 cells. Because the extracts of GLAE with stronger antioxidant activity show stronger anticancer activities, it is implied that the contents of flavonoids in GL are responsible not only for its antioxidant activities but also effectively prevent osteoporosis.

5. Conclusions

This study is the first to demonstrate that G. littoralis extracts can enhance osteoblast cell formation. The data produced at the molecular level suggest that G. littoralis extracts effectively induced osteoblast cell ALP production and mineralization. However, further research is required to establish the detailed mechanism involved in G. littoralis’s anti-osteoporotic potential by identifying the active components present in G. littoralis extracts. This study suggests that G. littoralis extracts have phytoestrogenic properties that may enable the development of therapeutic agents to prevent osteoporosis.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pr11051491/s1. Table S1. Pearson’s correlation coefficient among antioxidant activities, total phenolic contents, and total flavonoid contents.

Author Contributions

C.J.K. and B.K.G. contributed by doing experiments and writing the manuscript. C.Y.Y. supervised the experiments. S.K.C. and J.G.L. contributed by analyzing phenolic compounds and editing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funding from Hwajin Bio Cosmetic, Chuncheon 24232, Korea. Also, this work was supported by funding from the KU research professor program.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rachner, T.D.; Khosla, S.; Hofbauer, L.C. Osteoporosis: Now and the future. Lancet 2011, 377, 1276–1287. [Google Scholar] [CrossRef]
  2. Rodan, G.A.; Martin, T.J. Therapeutic approaches to bone diseases. Science 2000, 289, 1508–1514. [Google Scholar] [CrossRef]
  3. Kung, A. Management of osteoporosis in Hong Kong. Clin. Calcium 2004, 14, 108–111. [Google Scholar]
  4. Kanis, J.A. WHO Technical Report; University of Sheffield: Sheffield, UK, 2007; p. 66. [Google Scholar]
  5. IOF (International Osteoporosis Foundation). Facts and Statistics. International Osteoporosis Foundation Website. Available online: http://www.iofbonehealth.org/factsstatistics (accessed on 18 January 2014).
  6. Udagawa, N.; Takahashi, N.; Jimi, E.; Matsuzaki, K.; Tsurukai, T.; Itoh, K.; Nakagawa, N.; Yasuda, H.; Goto, M.; Tsuda, E.; et al. Osteoblasts/stromal cells stimulate osteoclast activation through expression of osteoclast differentiation factor/RANKL but not macrophage colony-stimulating factor. Bone 1999, 25, 517–523. [Google Scholar] [CrossRef]
  7. Suda, T.; Ueno, Y.; Fujii, K.; Shinki, T. Vitamin D and bone. J. Cell. Biochem. 2003, 88, 259–266. [Google Scholar] [CrossRef]
  8. Meghji, S.; Sandy, J.R.; Scutt, A.M.; Harvey, W.; Harris, M. Stimulation of bone resorption by lipoxygenase metabolites of arachidonic acid. Prostaglandins 1988, 36, 139–149. [Google Scholar] [CrossRef]
  9. Garcia, C.; Boyce, B.F.; Gilles, J.; Dallas, M.; Qiao, M.; Mundy, G.R.; Bonewald, L.F. Leukotriene B4 stimulates osteoclastic bone resorption both in vitro and in vivo. J. Bone Miner. Res. 1996, 11, 1619–1627. [Google Scholar] [CrossRef] [PubMed]
  10. Gruber, H.E.; Ivey, J.L.; Baylink, D.L.; Mathews, M.; Nelp, W.B.; Sisom, B.; Chestnut, C.H. Long-term calcitonin therapy in post-menopausal osteoporosis. Metabolism 1984, 33, 295–303. [Google Scholar] [CrossRef] [PubMed]
  11. Tasadduq, R.; Gordon, J.; AL-Ghanim, K.A.; Lian, J.B.; Wijnen, A.J.V.; Stein, J.L.; Stein, G.S.; Shakoori, A.R. Ethanol Extract of Cissus quadrangularis Enhances Osteoblast Differentiation and Mineralization of Murine Pre-Osteoblastic MC3T3-E1 Cells. J. Cell. Physiol. 2017, 232, 540–547. [Google Scholar] [CrossRef] [PubMed]
  12. Hamadeh, I.S.; Ngwa, B.A.; Gong, Y. Drug induced osteonecrosis of the jaw. Cancer Treat. Rev. 2015, 41, 455–464. [Google Scholar] [CrossRef]
  13. Abrahamsen, B. Adverse effects of bisphosphonates. Calcif. Tissue Int. 2010, 86, 421–435. [Google Scholar] [CrossRef] [PubMed]
  14. Rossouw, J.E.; Anderson, G.L.; Prentice, R.L.; LaCroix, A.Z.; Kooperberg, C.; Stefanick, M.L.; Jackson, R.D.; Beresford, S.A.; Howard, B.V.; Johnson, K.C.; et al. Writing group for the Women’s Health Initiative Investigators. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: Principal results from the women’s health Initiative randomized controlled trial. J. Am. Med. Assoc. 2002, 288, 321–333. [Google Scholar]
  15. Adluri, R.S.; Zhan, L.; Bagchi, M.; Maulik, N.; Maulik, G. Comparative effects of a novel plant-based calcium supplement with two common calcium salts on proliferation and mineralization in human osteoblast cells. Mol. Cell. Biochem. 2010, 340, 73–80. [Google Scholar] [CrossRef]
  16. Kodama, H.; Amagai, Y.; Sudo, H.; Kasai, S.; Yamamoto, S. Establishment of a clonal osteogenic cell line from newborn mouse calvaria. Jpn. J. Oral Biol. 1981, 23, 899–901. [Google Scholar] [CrossRef]
  17. Quarles, L.D.; Yohay, D.L.; Lever, L.W.; Caton, R.; Wenstrup, R.J. Distinct proliferative and differentiated stages of murine MC3T3-E1 cells in culture: An in vitro model of osteoblast development. J. Bone Miner. Res. 1992, 7, 683–692. [Google Scholar] [CrossRef] [PubMed]
  18. Golub, E.E.; Boesze-Battaglia, K. The role of alkaline phosphatase in mineralization. Curr. Opin. Orthop. 2007, 18, 444–448. [Google Scholar] [CrossRef]
  19. Jeong, J.C.; Lee, J.W.; Yoon, C.H.; Lee, Y.C.; Chung, K.H.; Kim, M.G.; Cheorl-Ho Kim, C.H. Stimulative effects of Drynariae Rhizoma extracts on the proliferation and differentiation of osteoblastic MC3T3-E1 Cells. J. Ethnopharmacol. 2005, 96, 489–495. [Google Scholar] [CrossRef]
  20. Lian, J.B.; Stein, G.S. Concepts of osteoblast growth and differentiation: Basis for modulation of bone cell development and tissue formation. Crit. Rev. Oral Biol. Med. 1992, 3, 269–305. [Google Scholar] [CrossRef]
  21. Ammon, H.P.T.; Mack, T.; Singh, G.B.; Safayhi, H. Inhibition of Leukotriene B4 Formation in Rat Peritoneal Neutrophils by an Ethanolic Extract of the Gum Resin Exudate of Boswellia serrata. Planta Med. 1991, 57, 203–207. [Google Scholar] [CrossRef]
  22. Jun, A.Y.; Kim, H.J.; Park, K.K.; Son, K.H.; Lee, D.H.; Woo, M.H.; Kim, Y.S.; Lee, S.K.; Chung, W.Y. Extract of Magnoliae Flos inhibits ovariectomy-induced osteoporosis by blocking osteoclastogenesis and reducing osteoclast-mediated bone resorption. Fitoterapia 2012, 83, 1523–1531. [Google Scholar] [CrossRef]
  23. Mohan, S.; Kutilek, S.; Zhang, C.; Shen, H.G.; Kodama, Y.; Srivastava, A.K.; Wergedal, J.E.; Beamer, W.G.; Baylink, D.J. Comparison of bone formation responses to parathyroid hormone (1–34), (1–31), and (2–34) in mice. Bone 2000, 27, 471–478. [Google Scholar] [CrossRef]
  24. Anderson, J.; Garner, S. Phytoestrogens and bone Bailieres. Clin. Endocrinol. Metab. 1998, 12, 543–557. [Google Scholar]
  25. Fitzpatrick, L. Selective estrogen receptor modulators and phytoestrogens: New therapies for the postmenopausal women. Mayo Clin. Proc. 1999, 74, 601–607. [Google Scholar] [CrossRef] [PubMed]
  26. Jing, Y.; Li, J.; Zhang, Y.; Zhang, R.; Zheng, Y.; Hu, B.; Wu, L.; Zhang, D. Structural characterization and biological activities of a novel polysaccharide from Glehnia littoralis and its application in preparation of nano-silver. Int. J. Biol. Macromol. 2021, 183, 1317–1326. [Google Scholar] [CrossRef] [PubMed]
  27. Rozema, J.; Bijwaard, P.; Prast, G.; Broekman, R. Ecophysiological adaptation of coastal halophytes from foredunes and salt marshes. Vegetatio 1985, 62, 499–521. [Google Scholar] [CrossRef]
  28. Cassileth, B.R.; Rizvi, N.; Deng, G.; Yeung, K.S.; Vickers, A.; Guillen, S.; Woo, D.; Coleton, M.; Kris, M.G. Safety and pharmacokinetic trial of docetaxel plus an Astragalus-based herbal formula for non-small cell lung cancer patients. Cancer Chemother. Pharmacol. 2009, 65, 67–71. [Google Scholar] [CrossRef]
  29. Ng, T.B.; Liu, F.; Wang, H.X. The antioxidant effects of aqueous and organic extracts of Panax quinquefolium, Panax notoginseng, Codonopsis pilosula, Pseudostellaria heterophylla, and Glehnia littoralis. J. Ethnopharmacol. 2004, 93, 285–288. [Google Scholar] [CrossRef]
  30. Chiang Su New Medicinal College (de.). Dictionary of Chinese Crude Drug’; Shanghai Scientific Technologic Publisher: Shanghai, China, 1977; p. 644. [Google Scholar]
  31. Masuda, T.; Takasugi, M.; Anetai, M. Psoralen and other linear furanocoumarins as phytoalexins in Glehnia littoralis. Phytochemistry 1998, 47, 13–16. [Google Scholar] [CrossRef]
  32. Hwang, Y.H.; Ha, H.; Kim, R.; Cho, C.W.; Song, Y.R.; Hong, H.D.; Kim, T. Anti-Osteoporotic Effects of Polysaccharides Isolated from Persimmon Leaves via Osteoclastogenesis Inhibition. Nutrients 2018, 10, 901. [Google Scholar] [CrossRef]
  33. Singleton, V.L.; Rossi, J.A., Jr. Colorimetry of total phenolics with phosphomolybdic-phosphotungstic acid reagents. Am. J. Enol. Vitic. 1965, 16, 144–158. [Google Scholar] [CrossRef]
  34. Moreno, M.I.; Isla, M.I.; Sampietro, A.R.; Vattuone, M.A. Comparison of the free radical-scavenging activity of propolis from several regions of Argentina. J. Ethnopharmacol. 2000, 71, 109–114. [Google Scholar] [CrossRef] [PubMed]
  35. Chung, I.-M.; Chelliah, R.; Oh, D.-H.; Kim, S.-H.; Yu, C.Y.; Ghimire, B.K. Tupistra nutans wall. root extract, rich in phenolics, inhibits microbial growth and α-glucosidase activity, while demonstrating strong antioxidant potential. Braz. J. Bot. 2019, 42, 383–397. [Google Scholar] [CrossRef]
  36. Denizot, F.; Lang, R. Rapid colorimetric assay for cell growth and survival: Modifications to the tetrazolium dye procedure giving improved sensitivity and reliability. J. Immunol. Methods 1986, 89, 271–277. [Google Scholar] [CrossRef] [PubMed]
  37. Kim, B.M.; Kim, G.T.; Kim, E.J.; Lim, E.G.; Kim, S.Y.; Kim, Y.M. Extract from Artemisia annua Linné induces apoptosis through the mitochondrial signaling pathway in HepG2 cells. J. Korean Soc. Food Sci. Nut. 2016, 45, 1708–1716. [Google Scholar] [CrossRef]
  38. Sewing, S.; Boess, F.; Moisan, A.; Bertinetti-Lapatki, C.; Minz, T.; Hedtjaern, M.; Tessier, Y.; Schuler, F.; Singer, T.; Roth, A.B. Establishment of a predictive in vitro assay for assessment of the hepatotoxic potential of oligonucleotide drugs. PLoS ONE 2016, 11, 159431. [Google Scholar] [CrossRef] [PubMed]
  39. Liu, X.W.; Ma, B.; Zi, Y.; Xiang, L.B.; Han, T.Y. Effects of rutin on osteoblast MC3T3-E1 differentiation, ALP activity and Runx2 protein expression. Eur. J. Histochem. 2021, 65, 3195. [Google Scholar] [CrossRef]
  40. Park, E.K.; Jin, H.S.; Cho, D.Y.; Kim, J.H.; Kim, M.C.; Choi, C.W.; Jin, Y.L.; Lee, J.W.; Park, J.H.; Chung, Y.S.; et al. The effect of Lycii radicis cortex extract on bone formation in vitro and in vivo. Molecules 2014, 19, 19594–19609. [Google Scholar] [CrossRef]
  41. Zakłos-Szyda, M.; Nowak, A.; Pietrzyk, N.; Podsedek, A. Viburnum opulus L. juice phenolic compounds influence osteogenic differentiation in human osteosarcoma saos-2 cells. Int. J. Mol. Sci. 2020, 21, 4909. [Google Scholar] [CrossRef]
  42. Bukhari, S.N.A.; Hussain, F.; Thu, H.E.; Hussain, Z. Synergistic effects of combined therapy of curcumin and Fructus Ligustri Lucidi for treatment of osteoporosis: Cellular and molecular evidence of enhanced bone formation. J. Integr. Med. 2019, 17, 38–45. [Google Scholar] [CrossRef]
  43. Techaniyom, P.; Tanurat, P.; Sirivisoot, S. Osteoblast differentiation and gene expression analysis on anodized titanium samples coated with graphene oxide. Applied Surface Science 2020, 526, 146646. [Google Scholar] [CrossRef]
  44. Matsubara, T.; Kida, K.; Yamaguchi, A.; Hata, K.; Ichida, F.; Meguro, H.; Aburatani, H.; Nishimura, R.; Yoneda, T. BMP2 regulates Osterix through Msx2 and Runx2 during osteoblast differentiation. J. Biol. Chem. 2008, 283, 29119–29144. [Google Scholar] [CrossRef] [PubMed]
  45. Kasote, D.M.; Katyare, S.S.; Hegde, M.V.; Bae, H. Significance of Antioxidant Potential of Plants and its Relevance to Therapeutic Applications. Int. J. Biol. Sci. 2015, 11, 982–991. [Google Scholar] [CrossRef] [PubMed]
  46. Womeni, H.M.; Djikeng, F.T.; Tiencheu, B.; Linder, M. Antioxidant potential of methanolic extracts and powders of some Cameroonian spices during accelerated storage of soybean oil. Adv. Biol. Chem. 2013, 3, 304–313. [Google Scholar] [CrossRef]
  47. Naka, K.; Muraguchi, T.; Hoshii, T.; Hirao, A. Regulation of reactive oxygen species and genomic stability in hematopoietic stem cells. Antioxid. Redox Signal 2008, 10, 1883–1894. [Google Scholar] [CrossRef] [PubMed]
  48. Bai, X.C.; Lu, D.; Liu, A.L.; Zhang, Z.M.; Li, X.M.; Zou, Z.P.; Zeng, W.S.; Cheng, B.L.; Luo, S.Q. Reactive oxygen species stimulates receptor activator of NF-kappaB ligand expression in osteoblast. J. Biol. Chem. 2005, 280, 17497–17506. [Google Scholar] [CrossRef]
  49. 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]
  50. Kitajima, J.; Okamura, C.; Ishikawa, T.; Tanaka, Y. Coumarin glycosides of Glehnia lifforalis root and rhizoma. Chem. Pharm. Bull. 1998, 46, 1404–1407. [Google Scholar] [CrossRef]
  51. Lee, J.W.; Lee, C.; Jin, Q.; Yeon, E.T.; Lee, D.; Kim, S.Y.; Han, S.B.; Hong, J.T.; Lee, M.K.; Hwang, B.Y. Pyranocoumarins from Glehnia littoralis inhibit the LPS-induced NO production in macrophage RAW 264.7 cells. Bioorganic Med. Chem. Lett. 2014, 24, 2717–2719. [Google Scholar] [CrossRef]
  52. Malik, A.; Kushnoor, A.; Saini, V.; Singhal, S.; Kumar, S.; Yadav, Y.C. In vitro antioxidant properties of Scopolet in. J. Chem. Pharm. Res. 2011, 3, 659–665. [Google Scholar]
  53. Um, Y.R.; Lee, J.I.; Lee, J.L.; Kim, H.J.; Yea, S.S.; Seo, Y.W. Chemical constituents of the halophyte Glehnia littoralis. J. Korean Chem. Soc. 2010, 54, 701–706. [Google Scholar] [CrossRef]
  54. Nasser, M.I.; Zhu, S.; Hu, H.; Huang, H.; Guo, M.; Zhu, P. Effects of imperatorin in the cardiovascular system and cancer. Biomed. Pharmacother. 2019, 120, 109401. [Google Scholar] [CrossRef] [PubMed]
  55. Bertina, R.; Chena, Z.; Martínez-Vázquez, M.; García-Argaéz, A.; Froldi, G. Vasodilation and radical-scavenging activity of imperatorin and selected coumarinic and flavonoid compounds from genus Casimiroa. Phytomedicine 2014, 21, 586–594. [Google Scholar] [CrossRef] [PubMed]
  56. Shen, D.Y.; Chao, C.H.; Chan, H.H.; Huang, G.J.; Hwang, T.L.; Lai, C.Y.; Lee, K.H.; Thang, T.D.; Wu, T.S. Bioactive constituents of Clausena lansium and a method for discrimination of aldose enantiomers. Phytochemistry 2012, 82, 110–117. [Google Scholar] [CrossRef] [PubMed]
  57. Adebajo, A.C.; Iwalewa, E.O.; Obuotor, E.M.; Ibikunle, G.F.; Omisore, N.O.; Adewunmi, C.O.; Obaparusi, O.O.; Klaes, M.; Adetogun, G.E.; Schmidt, T.J.; et al. Pharmacological properties of the extract and some isolated compounds of Clausena lansium stem bark: Anti-trichomonal, antidiabetic, antiinflammatory, hepatoprotective and antioxidant effects. J. Ethnopharmacol. 2009, 122, 10–19. [Google Scholar] [CrossRef] [PubMed]
  58. Kassim, N.K.; Rahmanil, M.; Ismail, A.; Sukari, M.A.; Ee, G.C.L.; Nasir, N.M.; Awang, K. Antioxidant activity-guided separation of coumarins and lignan from Melicope glabra (Rutaceae). Food Chem. 2013, 139, 87–92. [Google Scholar] [CrossRef]
  59. Singh, R.; Singh, B.; Singh, S.; Kumar, N.; Kumar, S.; Arora, S. Umbelliferone-An antioxidant isolated from Acacia nilotica (L.) Willd. Ex. Del. Food Chem. 2010, 120, 825–830. [Google Scholar] [CrossRef]
  60. Kanimozhi, G.; Prasad, N.R.; Ramachandran, S.; Pugalendi, K.V. Umbelliferone modulates gamma-radiation induced reactive oxygen species generation and subsequent oxidative damage in human blood lymphocytes. Eur. J. Pharmacol. 2011, 672, 20–29. [Google Scholar] [CrossRef]
  61. Luyen, B.T.L.; Tai, B.H.; Thao, N.P.; Lee, S.H.; Jang, H.D.; Lee, Y.M.; Kim, Y.H. Evaluation of the Anti-osteoporosis and Antioxidant Activities of Phenolic Compounds from Euphorbia maculate. J. Korean Soc. Appl. Biol. Chem. 2014, 57, 573–579. [Google Scholar] [CrossRef]
  62. Zhang, J.K.; Yang, L.; Meng, G.L.; Yuan, Z.; Fan, J.; Li, D.; Chen, J.Z.; Shi, T.Y.; Hu, H.M.; Wei, B.Y.; et al. Protection by salidroside against bone loss via inhibition of oxidative stress and bone-resorbing mediators. PLoS ONE 2013, 8, e57251. [Google Scholar] [CrossRef]
  63. Lee, E.J.; Kang, Y.H. Coumarin Boosts Optimal Bone Remodeling Through Blocking AGE-RAGE Interaction in Diabetic Osteoblasts and Osteoclasts | Current Developments in Nutrition | Oxford Academic. Curr. Dev. Nutr. 2020, 4, 395. [Google Scholar]
  64. Yan, D.Y.; Tang, J.; Chen, L.; Wang, B.; Weng, S.; Xie, Z.; Wu, Z.Y.; Shen, Z.; Bai, B.; Yang, L. Imperatorin promotes osteogenesis and suppresses osteoclast by activating AKT/GSK3 β/β-catenin pathways. J. Cell. Mol. Med. 2020, 24, 2330–2341. [Google Scholar] [CrossRef] [PubMed]
  65. Lin, F.X.; Zheng, G.Z.; Chang, B.O.; Chen, R.C.; Zhang, Q.H.; Xie, P.; Li, X.D. Connexin 43 modulates osteogenic differentiation of bone marrow stromal cells through GSK-3beta/Beta-catenin signaling pathways. Cell. Physiol. Biochem. 2018, 47, 161–175. [Google Scholar] [CrossRef]
  66. Galli, C.; Piemontese, M.; Lumetti, S.; Manfredi, E.; Macaluso, G.M.; Passeri, G. GSK3b-inhibitor lithium chloride enhances activation of Wnt canonical signaling and osteoblast differentiation on hydrophilic titanium surfaces. Clin. Oral Implant. Res. 2013, 24, 921–927. [Google Scholar] [CrossRef]
  67. Wang, J.; Guan, X.; Guo, F.; Zhou, J.; Chang, A.; Sun, B.; Cai, Y.; Ma, Z.; Dai, C.; Li, X.; et al. miR-30e reciprocally regulates the differentiation of adipocytes and osteoblasts by directly targeting low-density lipoprotein receptor-related protein 6. Cell Death Dis. 2013, 4, e845. [Google Scholar] [CrossRef] [PubMed]
  68. Li, J.P.; Zhuang, H.T.; Xin, M.Y.; Zhou, Y.L. MiR-214 inhibits human mesenchymal stem cells differentiating into osteoblasts through targeting β-catenin. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4777–4783. [Google Scholar]
  69. Wei, W.; Zeve, D.; Suh, J.M.; Wang, X.; Du, Y.; Zerwekh, J.E.; Dechow, P.C.; Graff, J.M.; Wan, Y. Biphasic and dosage-dependent regulation of osteoclastogenesis by β-catenin. Mol. Cell. Biol. 2011, 31, 4706–4719. [Google Scholar] [CrossRef]
  70. Kwak, S.C.; Baek, J.M.; Lee, C.H.; Yoon, K.H.; Lee, M.S.; Kim, J.Y. Umbelliferone Prevents Lipopolysaccharide-Induced Bone Loss and Suppresses RANKL-Induced Osteoclastogenesis by Attenuating Akt-c-Fos-NFATc1 Signaling. Int. J. Biol. Sci. 2019, 15, 2427–2437. [Google Scholar] [CrossRef]
  71. Li, H.T.; He, L.; Qiu, J.B. Effects of the Chinese herb component phellopterin on the increase in cytosolic free calcium in PC12 cells. Drug Dev. Res. 2007, 68, 79–83. [Google Scholar] [CrossRef]
  72. Ryu, S.Y.; Kim, J.C.; Kim, Y.S.; Kim, H.T.; Kim, S.K.; Chi, G.J.; Kim, J.S.; Lee, S.W.; Heor, J.H.; Cho, K.Y. Antifungal activities of coumarins isolated from Angelica gigas and Angelica dahurica against plant pathogenic fungi. Korean J. Pestic. Sci. 2001, 5, 26–35. [Google Scholar]
  73. Kontogiorgis, C.A.; Hadjipavlou-Litina, D.J. Synthesis and antiinflammatory activity of coumarin derivatives. J. Med. Chem. 2005, 48, 6400–6408. [Google Scholar] [CrossRef]
  74. Stein, G.S.; Lian, J.B.; Owen, T.A. Relationship of cell-growth to the regulation of tissue-specific gene-expression during osteoblast differentiation. FASEB J. 1990, 4, 3111–3123. [Google Scholar] [CrossRef] [PubMed]
  75. Evans, D.B.; Bunning, R.A.D.; Russell, R.G.G. The effects of recombinant human interleukin-1 on cellular proliferation and the production of prostaglandin E2, plasminogen activator, osteocalcin and alkaline phosphatase by osteoblast-like cells derived from human bone. Biochem. Biophys. Res. Commun. 1990, 166, 208–216. [Google Scholar] [CrossRef]
  76. Nash, L.A.; Ward, W.E. Comparison of black, green and rooibos tea on osteoblast activity. Food Funct. 2016, 7, 1166–1175. [Google Scholar] [CrossRef]
  77. Ekeuku, S.O.; Pang, K.L.; Chin, K.Y. Effects of Caffeic Acid and Its Derivatives on Bone: A Systematic Review. Drug Des. Dev. Ther. 2021, 15, 259–275. [Google Scholar] [CrossRef] [PubMed]
  78. Zhou, R.P.; Lin, S.J.; Wan, W.B.; Zuo, H.L.; Yao, F.F.; Ruan, H.B.; Xu, J.; Song, W.; Zhou, Y.C.; Wen, S.Y.; et al. Chlorogenic Acid Prevents Osteoporosis by Shp2/PI3K/Akt Pathway in Ovariectomized Rats. PLoS ONE 2016, 11, e0166751. [Google Scholar] [CrossRef] [PubMed]
  79. Wong, S.K.; Chin, K.Y.; Ima-Nirwana, S. Quercetin as an Agent for Protecting the Bone: A Review of the Current Evidence. Int. J. Mol. Sci. 2020, 21, 6448. [Google Scholar] [CrossRef]
  80. Jang, S.A.; Hwang, Y.H.; Kim, T.; Yang, H.; Lee, J.; Seo, Y.H.; Park, J.I.; Ha, H. Water Extract of Agastache rugosa Prevents Ovariectomy-Induced Bone Loss by Inhibiting Osteoclastogenesis. Foods 2020, 9, 1181. [Google Scholar] [CrossRef]
  81. Goto, T.; Hagiwara, K.; Shirai, N.; Yoshida, K.; Hagiwara, H. Apigenin inhibits osteoblastogenesis and osteoclastogenesis and prevents bone loss in ovariectomized mice. Cytotechnology 2015, 67, 357–365. [Google Scholar] [CrossRef]
  82. Kim, T.H.; Jung, J.W.; Ha, B.G.; Hong, J.M.; Park, E.K.; Kim, H.J.; Kim, S.Y. The effects of luteolin on osteoclast differentiation, function in vitro and ovariectomy induced bone loss. J. Nutr. Biochem. 2011, 22, 8–15. [Google Scholar] [CrossRef]
  83. Nash, L.A.; Sullivan, P.J.; Peters, S.J.; Ward, W.E. Rooibos flavonoids, orientin and luteolin, stimulate mineralization in human osteoblasts through the Wnt pathway. Mol. Nutr. Food Res. 2015, 59, 443–453. [Google Scholar] [CrossRef]
  84. Wattel, A.; Kamel, S.; Mentaverri, R.; Lorget, F.; Prouillet, C.; Petit, J.P.; Brazier, M. Potent inhibitory effect of naturally occurring flavonoids quercetin and kaempferol on in vitro osteoclastic bone resorption. Biochem. Pharmacol. 2003, 65, 35–42. [Google Scholar] [CrossRef]
  85. Melguizo-Rodríguez, L.; Manzano-Moreno, F.J.; De Luna-Bertos, E.; Rivas, A.; Ramos-Torrecillas, J.; Ruiz, C.; García-Martínez, O. Effect of olive oil phenolic compounds on osteoblast differentiation. Eur. J. Clin. Investig. 2018, 48, e12904. [Google Scholar] [CrossRef]
  86. Xu, B.; Wang, X.; Wu, C.; Zhu, L.; Chen, O.; Wang, X. Flavonoid compound icariin enhances BMP-2 induced differentiation and signalling by targeting to connective tissue growth factor (CTGF) in SAMP6 osteoblasts. PLoS ONE 2018, 13, e0200367. [Google Scholar] [CrossRef] [PubMed]
  87. Liang, W.; Lin, M.; Li, X.; Li, C.; Gao, B.; Gan, H.; Yang, Z.; Lin, X.; Liao, L.; Yang, M. Icariin promotes bone formation via the BMP-2/Smad4 signal transduction pathway in the hFOB 1.19 human osteoblastic cell line. Int. J. Mol. Med. 2012, 30, 889–895. [Google Scholar] [CrossRef]
  88. Wu, J.-B.; Fong, Y.-C.; Tsai, H.-Y.; Chen, Y.-F.; Tsuzuki, M.; Tang, C.-H. Naringin-induced bone morphogenetic protein-2 expression via PI3K, Akt, c-Fos/c-Jun and AP-1 pathway in osteoblasts. Eur. J. Pharmacol. 2008, 588, 333–341. [Google Scholar] [CrossRef] [PubMed]
  89. Gaoli, X.; Yi, L.; Lili, W.; Qiutao, S.; Guang, H.; Zhiyuan, G. Effect of naringin combined with bone morphogenetic protein-2 on the proliferation and differentiation of MC3T3-E1 cells. Hua Xi Kou Qiang Yi Xue Za Zhi Huaxi Kouqiang Yixue Zazhi West China. J. Stomatol. 2017, 35, 275–280. [Google Scholar]
  90. Hofbauer, L.C.; Kuhne, C.A.; Viereck, V. The OPG/RANKL/RANK system in metabolic bone diseases. J. Musculoskelet. Neuronal Interact. 2004, 4, 268–275. [Google Scholar]
  91. Sugimoto, E.; Yamaguchi, M. Stimulatory effect of Daidzein in osteoblastic MC3T3-E1 cells. Biochem. Pharmacol. 2000, 59, 471–475. [Google Scholar] [CrossRef] [PubMed]
  92. Eriksen, E.F.; Colvard, D.S.; Berg, N.J.; Graham, M.L.; Mann, K.G.; Spelsberg, T.C.; Riggs, B.L. Evidence of estrogen receptors in normal human osteoblast-like cells. Science 1998, 241, 84–86. [Google Scholar] [CrossRef]
  93. Shin, C.S.; Cho, H.Y. Bone remodeling and mineralization. J. Korean Soc. Endocrinol. 2005, 20, 543–555. [Google Scholar] [CrossRef]
  94. Bonjour, J.P. Calcium and phosphate: A duet of ions playing for bone health. J. Am Coll. Nutr. 2011, 30, 438S–448S. [Google Scholar] [CrossRef]
  95. Mo, X.M.; Zeng, Y.; Hong, J. Biochemical characteristics of an ovariectomized female rat model of osteoporosis. J. Tradit. Complement Med. 1999, 526–528. [Google Scholar]
  96. Yun, J.H.; Hwang, E.S.; Kim, G.H. Effects of Chrysanthemum indicum L. extract on the growth and differentiation of osteoblastic MC3T3-E1 cells. J. Korean Soc. Food Sci. Nutr. 2011, 40, 1384–1390. [Google Scholar] [CrossRef]
  97. Park, J.H.; Lee, J.W.; Kim, H.J.; Lee, I.S. Effects of Solidago virga-aurea var. gigantea Miq. root extracts on the activity and differentiation of MC3T3- E1 osteoblastic cell. J. Korean Soc. Food Sci. Nutr. 2005, 34, 929–936. [Google Scholar]
  98. Akiko, M.; Tomoyo, K.Y.; Masato, H. Osteocalcin and its endocrine functions. Biochem. Pharmacol. 2017, 132, 1–8. [Google Scholar]
  99. Manolagas, S.C. Osteocalcin promotes bone mineralization but is not a hormone. PLoS Genet. 2020, 16, e1008714. [Google Scholar] [CrossRef] [PubMed]
  100. Komori, T. Signaling networks in RUNX2-dependent bone development. J. Cell. Biochem. 2011, 112, 750–755. [Google Scholar] [CrossRef]
  101. Bronckers, A.L.; Sasaguri, K.; Engelse, M.A. Transcription and immunolocalization of Runx2/Cbfa1/Pebp2alphaA in developing rodent and human craniofacial tissues: Further evidence suggesting osteoclasts phagocytose osteocytes. Microsc. Res. Tech. 2003, 61, 540–548. [Google Scholar] [CrossRef]
  102. Lorenzo, J.A.; Teitelbaum, S.; Faccio, R.; Takayanagi, H.; Choi, Y.; Horowitz, M.; Takayanagi, H. (Eds.) Chapter 6: The Signaling Pathways Regulating Osteoclast Differentiation; Academic Press: London, UK, 2011. [Google Scholar]
  103. Bruderer, M.; Richards, R.G.; Alini, M.; Stoddart, M.J. Role and regulation of RUNX2 in osteogenesis. Eur. Cell Mater. 2014, 28, 269–286. [Google Scholar] [CrossRef]
  104. Franceschi, R.T.; Xiao, G.; Zh Jiang, D.; Gopalakrishnan, R.; Yang Sh, Y.; Reith, E. Multiple signaling pathways converge on the Cbfa1/Runx2 transcription factor to regulate osteoblast differentiation. Connect. Tissue Res. 2003, 44, 109–116. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, J.E. Function of runx2 and osterix in osteogenesis and teeth. J. Korean Assoc. Oral Maxillofac. Surg. 2007, 33, 381–385. [Google Scholar]
  106. Wu, C.F.; Lin, Y.S.; Lee, S.C.; Chen, C.Y.; Wu, M.C.; Lin, J.S. Effects of Davallia formosana Hayata Water and Alcohol Extracts on Osteoblastic MC3T3-E1 Cells. Phytother. Res. 2017, 31, 1349–1356. [Google Scholar] [CrossRef]
  107. Hagiwara, K.; Goto, T.; Araki, M.; Miyazaki, H.; Hagiwara, H. Olive polyphenol hydroxytyrosol prevents bone loss. Eur. J. Pharmacol. 2011, 662, 78–84. [Google Scholar] [CrossRef] [PubMed]
  108. Dai, Z.; Li, Y.; Quarles, L.D.; Song, T.; Pan, W.; Zhou, H.; Xiao, Z. Resveratrol enhances proliferation and osteoblastic differentiation in human mesenchymal stem cells via ER-dependent ERK1/2 activation. Phytomedicine 2007, 14, 806–814. [Google Scholar] [CrossRef] [PubMed]
  109. Satué, M.; del Mar Arriero, M.; Monjo, M.; Ramis, J.M. Quercitrin and taxifolin stimulate osteoblast differentiation in MC3T3-E1 cells and inhibit osteoclastogenesis in RAW 264.7 cells. Biochem. Pharmacol. 2013, 86, 1476–1486. [Google Scholar] [CrossRef]
  110. Srivastava, S.; Bankar, R.; Roy, P. Assessment of the role of flavonoids for inducing osteoblast differentiation in isolated mouse bone marrow derived mesenchymal stem cells. Phytomedicine 2013, 20, 683–690. [Google Scholar] [CrossRef] [PubMed]
  111. Xiao, H.H.; Gao, Q.G.; Zhang, Y.; Wong, K.C.; Dai, Y.; Yao, X.S.; Wong, M.S. Vanillic acid exerts oestrogen-like activities in osteoblast-like UMR 106 cells through MAP kinase (MEK/ERK)-mediated ER signaling pathway. J. Steroid Biochem. Mol. Biol. 2014, 144 Pt B, 382–391. [Google Scholar] [CrossRef]
  112. Kim, M.B.; Song, Y.; Hwang, J.K. Kirenol stimulates osteoblast differentiation through activation of the BMP and Wnt/β-catenin signaling pathways in MC3T3-E1 cells. Fitoterapia 2014, 98, 59–65. [Google Scholar] [CrossRef] [PubMed]
  113. Xiao, H.H.; Fung, C.Y.; Mok, S.K.; Wong, K.C.; Ho, M.X.; Wang, X.L.; Yao, X.S.; Wong, M.S. Flavonoids from Herba epimedii selectively activate estrogen receptor alpha (ERα) and stimulate ER-dependent osteoblastic functions in UMR-106 cells. J. Steroid Biochem. Mol. Biol. 2014, 143, 141–151. [Google Scholar] [CrossRef]
  114. Hu, B.; Yu, B.; Tang, D.; Li, S.; Wu, Y. Daidzein promotes osteoblast proliferation and differentiation in OCT1 cells through stimulating the activation of BMP-2/Smads pathway. Genet. Mol. Res. 2016, 15, 1–10. [Google Scholar] [CrossRef]
  115. Isaac, J.; Erthal, J.; Gordon, J.; Gordon, J.; Duverger, O.; Sun, H.-W.; Lichtler, A.C.; Stein, G.S.; Lian, J.B.; Morasso, M.I. DLX3 regulates bone mass by targeting genes supporting osteoblast differentiation and mineral homeostasis in vivo. Cell Death Differ. 2014, 21, 1365–1376. [Google Scholar] [CrossRef] [PubMed]
  116. Khrimian, L.; Obri, A.; Karsenty, G. Modulation of cognition and anxiety-like behavior by bone remodeling. Mol. Metab. 2017, 6, 1610–1615. [Google Scholar] [CrossRef] [PubMed]
  117. Yang, Y.; Bai, Y.; He, Y.; Zhao, Y.; Chen, J.; Ma, L.; Pan, Y.; Hinten, M.; Zhang, J.; Karnes, R.J.; et al. PTEN Loss promotes intratumoral androgen synthesis and tumor microenvironment remodeling via aberrant activation of RUNX2 in castration-resistant prostate cancer. Clin Cancer Res. 2018, 24, 834–846. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (a) Total phenolic contents (TPC) and (b) total flavonoid contents (TFC) in G. littoralis extracts. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 1. (a) Total phenolic contents (TPC) and (b) total flavonoid contents (TFC) in G. littoralis extracts. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g001
Figure 2. Representative chromatograms of compounds of extracts from G. littoralis.
Figure 2. Representative chromatograms of compounds of extracts from G. littoralis.
Processes 11 01491 g002
Figure 3. Antioxidant activity (DPPH and ABTS radical assay) in G. littoralis extracts. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 3. Antioxidant activity (DPPH and ABTS radical assay) in G. littoralis extracts. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g003
Figure 4. Cell viability of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 4. Cell viability of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g004
Figure 5. Cell cytotoxicity of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 5. Cell cytotoxicity of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g005
Figure 6. ALP activity of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 6. ALP activity of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g006
Figure 7. Collagen synthesis effects of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 7. Collagen synthesis effects of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g007
Figure 8. Mineralization contents of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 8. Mineralization contents of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is the mean ± standard deviation of nine replicate tests. Mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g008
Figure 9. Osteocalcin contents of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is mean ± standard deviation of nine replicate tests. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Figure 9. Osteocalcin contents of extracts from each part of G. littoralis in osteoblastic MC3T3-E1 cell line. Each value is mean ± standard deviation of nine replicate tests. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM). GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Processes 11 01491 g009
Table 1. Forward and reverse primers sequences used in this study.
Table 1. Forward and reverse primers sequences used in this study.
Primer NameSequence
OsterixForward5′-AGCGACCACTTGAGCAAACAT-3′
Reverse5′-GCGGCTGATTGGCTTCTTCT-3′
RUNX2Forward5′-CGGCCCTCCCTGAACTCT-3′
Reverse5′-TGCCTGCCTRGGGATCTGTA-3′
Table 2. LC/UVD quantitative analysis compounds of extracts from G. littoralis (Unit: mg/g, dry weight).
Table 2. LC/UVD quantitative analysis compounds of extracts from G. littoralis (Unit: mg/g, dry weight).
Sample 1ScopoletinUmbelliferoneImperatorinPhellopterinTotal
GLSE53.0 ± 0.2 d1.6 ± 0.0 d2.0 ± 0.0 aN.D. 256.6 ± 0.2 d
GLFE17.7 ± 0.0 b0.8 ± 0.0 a8.9 ± 0.1 b1.1 ± 0.4 b28.5 ± 0.5 a
GLAE24.5 ± 1.1 c1.0 ± 0.0 b15.1 ± 0.6 c0.6 ± 0.1 a41.1 ± 1.8 b
GLRE8.5 ± 0.0 a1.4 ± 0.1 c31.9 ± 0.1 d2.3 ± 0.0 c44.2 ± 0.1 c
1 GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis plant extracts, GLRE: G. littoralis root extracts. 2 N.D.: not detected. Each value is means ± SD of three replicate tests. The mean values followed by the same letter are not significantly different based on the DMRT (p < 0.05).
Table 3. Effect of G. littoralis extracts on Osterix and RUNX2 mRNA expression in osteoblastic MC3T3-E1 cells.
Table 3. Effect of G. littoralis extracts on Osterix and RUNX2 mRNA expression in osteoblastic MC3T3-E1 cells.
SampleConcentration (μg/mL)mRNA Expression Rate (Fold)
Osterix 1RUNX2
P.C.AA (50 μg/mL) + BGP (100 mM)1.820 ± 0.072 c2.531 ± 0.050 a
Control-1.000 ± 0.000 ki1.000 ± 0.000 k
GLSE0.51.510 ± 0.046 ef2.327 ± 0.023 ab
11.530 ± 0.041 de2.220 ± 0.090 b
21.620 ± 0.033 d2.190 ± 0.250 b
51.820 ± 0.053 c1.940 ± 0.150 c
101.520 ± 0.021 ef1.533 ± 0.029 fg
201.430 ± 0.003 f1.410 ± 0.017 gh
GLFE0.50.990 ± 0.020 ki1.920 ± 0.080 cd
10.970 ± 0.070 lm1.830 ± 0.070 cde
20.960 ± 0.010 lm1.800 ± 0.090 cde
50.937 ± 0.055 lm1.700 ± 0.120 def
100.900 ± 0.070 lm1.310 ± 0.040 ghij
200.880 ± 0.050 m1.110 ± 0.050 jk
GLAE0.51.280 ± 0.020 gh1.280 ± 1.113 jk
11.250 ± 0.070 gh1.250 ± 1.517 fg
21.240 ± 0.080 gh1.240 ± 1.660 ef
51.200 ± 0.080 hi1.200 ± 1.353 ghi
101.130 ± 0.060 ij1.130 ± 1.190 hijk
201.080 ± 0.020 jk1.080 ± 1.073 jk
GLRE0.51.080 ± 0.050 jk1.150 ± 0.030 jk
11.320 ± 0.030 g1.060 ± 0.040 k
21.430 ± 0.080 f1.230 ± 0.120 hijk
51.850 ± 0.020 c1.190 ± 0.150 hijk
102.020 ± 0.120 b1.080 ± 0.070 jk
202.220 ± 0.050 a1.010 ± 0.030 k
1 Each value is mean ± standard deviation of nine replicate tests. Mean values within a column followed by the same letter are not significantly different based on the DMRT (p < 0.05). P.C.: Positive control (ascorbic acid (50 μg/mL), β-glycerophosphate (100 mM), AA: Ascorbic acid, BGP: β-glycerophosphate. GLSE: G. littoralis leaf, stem extracts, GLFE: G. littoralis fruit extracts, GLAE: G. littoralis all extracts, GLRE: G. littoralis root extracts.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, C.J.; Ghimire, B.K.; Choi, S.K.; Yu, C.Y.; Lee, J.G. Sustainable Bioactive Composite of Glehnia littoralis Extracts for Osteoblast Differentiation and Bone Formation. Processes 2023, 11, 1491. https://doi.org/10.3390/pr11051491

AMA Style

Kim CJ, Ghimire BK, Choi SK, Yu CY, Lee JG. Sustainable Bioactive Composite of Glehnia littoralis Extracts for Osteoblast Differentiation and Bone Formation. Processes. 2023; 11(5):1491. https://doi.org/10.3390/pr11051491

Chicago/Turabian Style

Kim, Chul Joong, Bimal Kumar Ghimire, Seon Kang Choi, Chang Yeon Yu, and Jae Geun Lee. 2023. "Sustainable Bioactive Composite of Glehnia littoralis Extracts for Osteoblast Differentiation and Bone Formation" Processes 11, no. 5: 1491. https://doi.org/10.3390/pr11051491

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