Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteoblasts

  • Authors:
    • Fang‑Bing Zhu
    • Jian‑Yue Wang
    • Ying‑Liang Zhang
    • Yun‑Gen Hu
    • Zhen‑Shuang Yue
    • Lin‑Ru Zeng
    • Wen‑Jie Zheng
    • Qiao Hou
    • Shi‑Gui Yan
    • Ren‑Fu Quan
  • View Affiliations

  • Published online on: October 31, 2016     https://doi.org/10.3892/mmr.2016.5908
  • Pages: 5377-5384
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Monotropein, the primary iridoid glycoside isolated from Morindacitrifolia, has been previously reported to possess potent antioxidant and antiosteoporotic properties. However, there is no direct evidence correlating the antiosteoporotic effect of monotropein with its observed antioxidant capacity, and the molecular mechanisms involved in mediating these processes remain unclear. Therefore, the aim of the present study was to investigate the protective effects of monotropein against oxidative stress in osteoblasts and the mechanisms involved in mediating this process. Osteoblast viability was evaluated using the MTT assay. The mitochondrial membrane potential and reactive oxygen species were detected by flow cytometry analyses. Western blotting and enzyme‑linked immunosorbent assays were performed to detect protein expression levels. A significant reduction in osteoblast viability was observed at 24 h following exposure to various concentrations (100‑1,000 µM) of H2O2 compared with untreated osteoblasts. The cytotoxic effect of H2O2 was notably reversed when osteoblasts were pretreated with 1‑10 µg/ml monotropein. Pretreatment with 1-10 µg/ml monotropein increased the mitochondrial membrane potential and reduced the generation of reactive oxygen species in osteoblasts following exposure to H2O2. In addition, the H2O2‑induced increase in apoptotic markers (caspase-3 and caspase-9) and H2O2-induced reduction in sirtuin 1 levels were significantly reversed following pretreatment of cells with monotropein. Furthermore, monotropein significantly reduced H2O2‑induced stimulation of NF‑κB expression, in addition to the expression of a number of proinflammatory mediators. These results indicate that monotropein suppresses apoptosis and the inflammatory response in H2O2‑induced osteoblasts through the activation of the mitochondrial apoptotic signaling pathway and inhibition of the NF‑κB signaling pathway.

Introduction

Osteoporosis is a key public health issue that affects millions of people worldwide, and predominantly occurs in postmenopausal women (1,2). It is a chronic progressive disease characterized by porous bones and the microarchitectural deterioration of bones (3). Over the last 20 years, various pharmacological agents, including alendronate, risedronate, estrogen and glucocorticoids, have been used for the prevention and treatment of osteoporosis (46). The primary aim of pharmacological therapy is to reduce the risk of fractures that may occur as a result of osteoporosis.

Estrogen deficiency is one of the main risk factors for osteoporosis, and has been associated with the enhanced production of reactive oxygen species (ROS) (7). Excessive levels of ROS (oxidative stress) have been demonstrated to be an important contributing factor in the etiology of various degenerative diseases, including atherosclerosis, osteoporosis and cancer, where the levels of markers associated with oxidative stress are markedly increased (8). At the cellular level, oxidant-induced injury confers a wide range of responses, including cell proliferation, differentiation arrest and apoptosis, through the activation of nuclear factor-κB (NF-κB), p53, c-Jun N-terminal kinase and extracellular signal-related kinase (ERK) signaling pathways (9). Previous studies have demonstrated that a strong correlation exists between oxidative stress and the pathogenesis of osteoporosis (10,11). Oxidative stress induced by hydrogen peroxide (H2O2) inhibits the differentiation of mouse MC3T3-E1 osteoblast precursor cells and M2-10B4 bone marrow cells (12,13). In addition, aged osteoporotic women have been demonstrated to exhibit a marked reduction in plasma antioxidant levels (14), and a biochemical association between increased oxidative stress and reduced bone mineral density was observed in aged women and men (15). Therefore, ROS may be considered as a target for the prevention of bone density loss, and may be used as a potential candidate for the treatment of osteoporosis.

Morindacitrifolia (M. citrifolia), also known as noni, is a tree in the Rubiaceae coffee family indigenous to the Hawaiian and Tahitian islands, and anthraquinones, flavonoids, iridoids and oligosaccharides have been isolated from M. citrifolia (16). The roots of M. officinalis, which are known to possess similar pharmacological effects to M. citrifolia, have been widely used in traditional Asian medicine to treat rheumatoid arthritis and diabetes (17). Monotropein is the active compound isolated from M. officinalis, and its molecular structure is shown in Fig. 1A. Previous studies demonstrated the anti-inflammatory effects of monotropein in rats with carrageenan-induced edema, in addition to in RAW 264.7 macrophages (17,18). However, there is no direct evidence to correlate the antiosteoporotic effects of monotropein with its antioxidant effects, and the associated molecular mechanisms remain unclear.

In the present study, the effects of monotropein on osteoblast viability and differentiation, and the generation of ROS in osteoblasts in response to 400 µM H2O2 were investigated. The results demonstrated that monotropein promoted cell differentiation and protected osteoblasts from H2O2-induced oxidative damage by inhibiting the expression of apoptosis-associated markers and the activation of the NF-κB signaling pathway.

Materials and methods

Cell culture

The current study was approved by the Ethics Committee of the Xiaoshan Traditional Chinese Medical Hospital (Hangzhou, China). A total of 30 male Sprague-Dawley rats (age, 3 days; weight, 180 g), purchased from the Experimental Animal Center of the Xiaoshan Traditional Chinese Medical Hospital (Hangzhou, China), were housed in the animal facility in individual cages at 25°C and 60–70% humidity with 12 h light/dark cycles and free access to food and water. The rats were anesthetized by intraperitoneal injection of 3% sodium pentobarbital (40 mg/kg; Sigma-Aldrich; Merck Millipore, Darmstadt, Germany). Primary osteoblasts were prepared according to the methods described previously (19). Osteoblasts were isolated from the calvarias of newborn rats. Briefly, five calvarias were minced and incubated with 0.25% trypsin (Gibco; Thermo Fisher Scientific, Waltham, MA, USA) for 10 min at 37°C, and 4 mg/ml collagenase I (0.4%; National Biochemicals Corporation, Twinsburg, OH, USA) for 90 min at 37°C three consecutive times. Cells isolated from the last four to six digests were cultured in Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) containing 10% fetal bovine serum (FBS; Hangzhou Sijiqing Biological Engineering Materials Co., Ltd., Hangzhou, China) and antibiotics (300,000 U/l penicillin/streptomycin; Invitrogen; Thermo Fisher Scientific, Inc.). After reaching 80–90% confluence, the cells were removed from each flask and pooled together to produce a single osteoblast culture. Osteoblasts were subsequently collected by centrifugation (1,000 × g/min for 10 min) at 4°C and resuspended in DMEM containing 10% FBS and 300,000 U/l penicillin/streptomycin.

H2O2 treatment

Osteoblasts were harvested and randomly divided into the following 5 groups: The untreated control group; the H2O2-treated group; and three monotropein plus H2O2-treated groups, which were treated with 1, 5 and 10 µg/ml monotropein, respectively. Osteoblasts in the H2O2 group were incubated for 24 h in DMEM containing 400 µM H2O2. In the monotropein plus H2O2-treated groups, the cells were pre-incubated with the various concentrations of monotropein for 24 h, prior to incubation with 400 µM H2O2 for 24 h.

Monotropein

Monotropein (98% purity) was isolated from M. officinalis (Nanjing Zelang Medical Technological Co., Ltd., Nanjing, China). It was dissolved in 150 µl of dimethylsulfoxide (DMSO) and diluted to the desired concentrations prior to utilization, with the final concentration of DMSO maintained below 0.5%.

Cell viability assay

Osteoblast viability was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay as described previously (20). In brief, cells (5×104 cells/ml) were first seeded in 96-well culture plates and treated with or without H2O2, in the absence or presence of monotropein (1, 5 or 10 µg/ml) for 24, 48 and 72 h. Cell viability was subsequently evaluated using an MTT Cell Proliferation assay kit (cat. no. 4890-025-K; Wuhan Amyjet Scientific Co., Ltd., Wuhan, China). The absorbance was measured at 490 nm with an automated Bio-Rad 550 microtiter plate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Analysis of alkaline phosphatase (ALP) activity

Osteoblasts (1×104 cells/well) were seeded and cultured in DMEM containing 10% FBS for 4 days, prior to treatment of the cells with or without H2O2 and in the absence or presence of monotropein (1, 5 and 10 µg/ml) for a further 2 days. ALP activity was measured at the end of the treatment period using p-nitrophenylphosphate as a substrate in 0.05 M 2-amino-2-methylpropanol and 2 mM MgCl2 (pH 10.5), according to the methods described previously (21). The amount of p-nitrophenol released was estimated by measuring the absorbance at 410 nm. The total protein concentration was determined using the Bradford protein assay as described previously (22).

Mitochondrial membrane potential (MMP)

Rhodamine-123 dye (Sigma Aldrich; Merck Millipore) was used to measure alterations in osteoblast MMP levels. Cells (1×104 cells/well) were seeded in a 24-well plate. Following treatment with or without H2O2, in the absence or presence of monotropein (1, 5 and 10 µg/ml) for 24 h, cells were washed with PBS, incubated with Rho-123 (10 mg/ml) and subsequently subjected to flow cytometry analysis using the BD Accuri C6 flow cytometer (BD Biosciences, San Jose, CA, USA; excitation wavelength, 480 nm; emission wavelength, 525 nm).

Detection of ROS

Detection of ROS was performed using flow cytometric analysis as described previously (23). In brief, osteoblasts (1×104 cells/well) were first seeded in a 24-well plate. Following treatment with or without H2O2, in the absence or presence of monotropein (1, 5 and 10 µg/ml) for 24 h, cells were washed with PBS, resuspended in complete medium and incubated with 0.5 µM dihydrorhodamine 123 (Sigma Aldrich; Merck Millipore) for 30 min at 37°C. ROS fluorescence intensity was determined by flow cytometric analysis, with excitation at 490 nm and emission at 520 nm.

Western blot analysis

Cells were seeded at a density of 1×105 cells/well in 6-well plates, incubated overnight and then treated with or without H2O2 in the absence or presence of monotropein (1, 5 and 10 µg/ml) for 24 h. Cells were lysed using radioimmunoprecipitation buffer supplemented with protease inhibitor (Beyotime Institute of Biotechnology, Shanghai, China). The protein concentration was estimated using a bicinchoninic acid assay kit (Thermo Fisher Scientific, Inc.). Cell protein lysates (50 µg) were separated on 10% sodium dodecyl sulfate-polyacrylamide gels and electroblotted onto a polyvinylidene fluoride membrane (Roche Diagnostics GmbH, Mannheim, Germany). Membranes were blocked in fat-free milk overnight at 4°C. Membranes were then incubated with the following primary antibodies for 2 h at 25°C: Polyclonal rabbit anti-caspase-3 (dilution, 1:200; cat. no. ab2302; Abcam, Cambridge, MA, USA); anti-caspase-9 (dilution, 1:500; cat. no. ab69514; Abcam); anti-cyclooxygenase-2 (COX-2; dilution, 1:500; cat. no. ab15191; Abcam); anti-inducible nitric oxide synthase (iNOS; dilution, 1:800; cat. no. ab3523; Abcam); anti-NF-κB p65 (dilution, 1:1,000; cat. no. ab16502; Abcam); and monoclonal mouse anti-sirtuin 1 (SIRT1; dilution, 1:800; cat. no. ab110304; Abcam). Mouse anti-histone protein 3 (dilution, 1:1,000; cat. no. ab1220; Abcam) or anti-GAPDH (dilution, 1:1,000; cat. no. ab8245; Abcam) monoclonal antibodies were used as loading controls. After washing, the membranes were incubated with horseradish peroxidase-conjugated goat anti-rabbit (cat. no. A0208) or goat anti-mouse IgG (cat. no. A0216) secondary antibodies (dilution, 1:1,000; Beyotime Institute of Biotechnology) at 37°C for 1 h. The blots were visualized using enhanced chemiluminescence (EMD Millipore, Billerica, MA, USA) and signal intensity was determined using ImageJ software (version 1.46; National Institutes of Health, Bethesda, MD, USA).

Enzyme-linked immunosorbent assay (ELISA)

The protein levels of rat tumor necrosis factor-a (TNF-α), interleukin (IL)-1β, IL-6 and macrophage-colony stimulating factor (M-CSF) in osteoblasts were determined using Quantikine murine-specific sandwich ELISA kits (cat. nos. RTA00, RLB00, R6000B and MMC00, respectively; R&D Systems, Inc., Minneapolis, MN, USA) according to the manufacturer's instructions. Absorbance was read at 570 nm using an EL301 Microwell Strip Reader (Omega Bio-Tek, Inc., Norcross, GA, USA).

Statistical analysis

Data are expressed as the mean ± standard deviation. Differences between groups were analyzed using a two-tailed Student's t-test. The SPSS statistical software program (version, 13.0; SPSS, Inc., Chicago, IL, USA) was used for analysis. P<0.05 was considered to indicate a statistically significant difference.

Results

Effect of monotropein on the viability of osteoblasts

To determine whether H2O2 may exhibit cytotoxic effects on osteoblasts in vitro, the effect of H2O2 exposure on osteoblast viability was determined using an MTT assay. Osteoblasts were treated with 0–1,000 µM H2O2 for 24, 48 and 72 h. As presented in Fig. 1B, treatment with >400 µM H2O2 significantly reduced cell viability in a dose and time-dependent manner (P=0.001). Therefore, 400 µM H2O2 was used in all subsequent experiments. As presented in Fig. 1C, 1, 5 and 10 µg/ml monotropein significantly inhibited the H2O2-induced suppression in osteoblast viability (P=0.011; 13.2±1.63, 27.9±2.65 and 37.5±2.32% viability increase compared with osteoblasts treated with H2O2 alone, respectively).

Effect of monotropein on the differentiation of osteoblasts

ALP activation is the earliest marker of osteoblast differentiation (24). In addition, the M-CSF cytokine is constitutively expressed during the growth phase of osteoblasts (25). As presented in Fig. 2A, a significant reduction in ALP activity was observed following incubation of osteoblasts with >200 µM H2O2 (P<0.05). Notably, pretreatment of cells with monotropein (1, 5 and 10 µg/ml) for 24 h significantly attenuated the H2O2-mediated downregulation of ALP activity (21.7±2.23, 34.2±2.02 and 45.1±1.35% activity increase, compared with osteoblasts treated with H2O2 alone, respectively; 1 µg/ml, P=0.005; 5 µg/ml, P=0.0002; 10 µg/ml, P=1.16×10−5; Fig. 2B). In addition, pretreatment with 1, 5 and 10 µg/ml monotropein significantly increased M-CSF expression compared with H2O2-only treated osteoblasts (27.1±1.83, 46.7±1.52 and 61.4±1.35%, respectively; 1 µg/ml, P=0.0003; 5 µg/ml, P=2.78×10−5; 10 µg/ml, P=1.21×10−5; Fig. 2C).

Effect of monotropein on MMP and ROS levels in H2O2-induced osteoblasts

Destruction of the MMP is the initial process of mitochondrial-induced apoptosis (26). To elucidate the possible mechanisms by which monotropein prevented the H2O2-induced decrease in cell viability and ALP activity in osteoblasts, the MMP and intracellular ROS levels in H2O2-treated osteoblasts with or without monotropein pretreatment were investigated. The MMP level in H2O2-induced osteoblasts was significantly decreased compared with that of the untreated control osteoblasts (Fig. 3A and C; P=9.98×10−7). However, osteoblasts pretreated with monotropein (1, 5 and 10 µg/ml) exhibited a significant dose-dependent increase in MMP levels (Fig. 3A and C; 1 µg/ml, P=9.59×10−5; 5 µg/ml, P=7.86×10−6; 10 µg/ml, P=2.72×10−6). MMP levels were increased by 3.1, 5.1 and 8.4-fold following pretreatment with 1, 5 and 10 µg/ml monotropein, respectively, when compared with that of H2O2-only treated osteoblasts. Similarly, ROS generation in monotropein-treated osteoblasts was significantly reduced in a dose-dependent manner when compared with H2O2-only treated controls (Fig. 3B and D; 1 µg/ml, P=0.0095; 5 µg/ml, P=0.0007; 10 µg/ml, P=0.0002). ROS levels were reduced by 27.9±1.26, 58.2±2.16 and 79.7±1.51% following pretreatment with 1, 5 and 10 µg/ml monotropein, compared with H2O2-only treated osteoblasts.

Effect of monotropein on the expression of apoptosis-associated proteins

In order to investigate the mechanisms underlying the anti-apoptotic effects of monotropein in H2O2-induced osteoblasts, the protein expression levels of apoptosis-associated molecules were determined by western blot analysis. As presented in Fig. 4, the protein expression levels of caspase-3 (P=7.58×10−5) and caspase-9 (P=8.57×10−6) were significantly increased following H2O2 treatment for 24 h compared with untreated controls, whereas SIRT1 (P=8.14×10−6) protein expression was significantly reduced. However, pretreatment of osteoblasts with monotropein (1, 5 and 10 µg/ml) for 24 h significantly attenuated the H2O2-induced upregulation of caspase-3 (1 µg/ml, P=0.007; 5 µg/ml, P=0.0018; 10 µg/ml, P=5.77×10−5) and caspase-9 (1 µg/ml, P=0.0489; 5 µg/ml, P=0.009; 10 µg/ml, P=0.0007) protein expression levels and the H2O2-induced downregulation in SIRT1 (1 µg/ml, P=0.001; 5 µg/ml, P=0.0005; 10 µg/ml, P=3.89×10−5) protein expression (Fig. 4).

Effect of monotropein on NF-κB p65, iNOS and COX-2 expression levels

In order to determine whether signaling pathways downstream of NF-κB p65 were affected by monotropein treatment, the protein expression levels of NF-κB, iNOS and COX-2 in osteoblasts following pretreatment with 0, 1, 5 or 10 µg/ml monotropein and exposure to H2O2 were examined. As presented in Fig. 5A and B, the protein expression levels of NF-κB p65 (P=1.35×10−5), iNOS (P=9.76×10−5) and COX-2 (P=7.89×10−6) were significantly increased in H2O2-induced osteoblasts compared with untreated controls. Following monotropein treatment, osteoblasts exhibited a significant reduction in the protein expression levels of NF-κB p65 (1 µg/ml, P=0.001; 5 µg/ml, P=0.0002; 10 µg/ml, P=5.53×10−5), iNOS (1 µg/ml, P=0.0482; 5 µg/ml, P=0.0162; 10 µg/ml, P=0.0023) and COX-2 (1 µg/ml, P=0.0084; 5 µg/ml, P=0.0017; 10 µg/ml, P=0.0001) compared with H2O2-only-treated osteoblasts (Fig. 5A and B). These data suggest that H2O2 may induce osteoblast injury through activating NF-κB and increasing the expression of downstream signaling pathways involving iNOS and COX-2.

Effect of monotropein on the protein expression levels of pro-inflammatory mediators

In order to determine whether inflammation was induced by H2O2, the protein expression levels of TNF-α, IL-1β and IL-6 in osteoblasts following incubation with H2O2 and in the presence or absence of monotropein were determined. As presented in Fig. 5C-E, the protein expression levels of TNF-α (P=3.59×10−6), IL-1β (P=9.19×10−6) and IL-6 (P=3.73×10−6) were significantly increased in H2O2-induced osteoblasts compared with untreated controls. Following monotropein treatment, osteoblasts exhibited a significant reduction in TNF-α (1 µg/ml, P=0.0002; 5 µg/ml, P=4.64×10−5; 10 mg/ml, P=9.79×10−6), IL-1β (1 µg/ml, P=0.0024; 5 µg/ml, P=0.0002; 10 µg/ml, P=4.76×10−5) and IL-6 (1 µg/ml, P=0.0009; 5 µg/ml, P=9.96×10−5; 10 µg/ml, P=3.48×10−5) protein expression levels compared with H2O2-only treated osteoblasts (Fig. 5C-E). These data suggest that H2O2 induces osteoblast injury through stimulating inflammatory responses and increasing the expression of proinflammatory mediators, including TNF-α, IL-1β and IL-6.

Discussion

ROS is known to contribute to the pathogenesis of a number of diseases, such as osteoporosis (27). H2O2 is one of the major sources of ROS, which disperses across cell membranes and generates highly reactive hydroxyl radicals that cause various types of oxidative damage by attacking cellular components (28). H2O2-induced apoptosis and inflammation have been reported to occur in several types of cells including mesenchymal stem cells, cardiomyocytes and alveolar epithelial cells (2931). In the present study, the effect of different concentrations of H2O2 on osteoblast viability was investigated. Treatment with H2O2 for 24 h significantly repressed the viability of osteoblasts at doses ranging from 100 to 1,000 µM when compared with untreated controls, which indicates that H2O2 may inhibit the viability of osteoblasts. In a previous study, pretreatment with curculigoside, one of the main bioactive phenolic compounds isolated from the rhizome of Curculigoorchioides Gaertn, markedly protected against the H2O2-induced inhibition of osteoblast viability (32). Consistent with these observations, pretreatment of osteoblasts with 1–10 µg/ml monotropein for 24 h in the present study, significantly suppressed cell injury following exposure to 400 µM H2O2. Taking these results into account, 400 µM H2O2 was considered to be sufficient for the induction of oxidative stress, and 1–10 µg/ml monotropein was selected to examine the effects of monotropein on osteoblast function.

ALP is widely expressed in various organs, including the liver, kidney, placenta and bone (33,34). ALP serves an important role in bone formation and remodeling through promoting mineralization of the matrix (35). Previous studies observed H2O2-induced suppression of osteoblast differentiation in bone marrow stem cells and MC3T3-E1 cells (36,37). M-CSF, also known as CSF1, is released by osteoblasts and is involved in the proliferation, differentiation and survival of bone marrow progenitor cells (38,39). In the present study, ALP activity and M-CSF release was observed to be significantly suppressed in H2O2-induced osteoblasts; the levels of which recovered following monotropein treatment. These observations suggest that monotropein may promote osteoblast differentiation.

Previous studies have demonstrated that activation of ERK is important for H2O2-induced apoptosis in cardiomyocytes, endothelial cells and osteoblasts (4042). H2O2 treatment increased Bax expression and led to hyperpolarization of the mitochondrial membrane potential in MC3T3-E1 mouse osteoblastic cells (33). This effect was prevented by treating cells with an inhibitor of the ERK upstream kinase mitogen activated protein kinase kinase1/2 (PD98059) (43). Consistent with these observations, the results presented in the current study demonstrated that monotropein could significantly reverse the H2O2-induced reduction in MMP levels and the H2O2-induced increase in ROS production. In addition, the protein expression levels of apoptotic markers in H2O2-induced osteoblasts were investigated, and the results suggested that the proapoptotic genes, caspase-3 and caspase-9, were significantly increased and the anti-apoptotic gene SIRT1 was significantly reduced. Notably, treatment with monotropein significantly reversed the effects of H2O2 on the expression of apoptosis-associated proteins in osteoblasts.

NF-κB has been demonstrated to participate in the regulation of cell survival genes, and mediate the expression of proinflammatory cytokines, including COX-2, iNOS, TNF-α, IL-1β and IL-6 (44,45). In the present study, the protein expression level of nuclear NF-κB p65 was examined, in order to determine the activity of NF-κB. The results demonstrated that the protein expression levels of nuclear NF-κB p65, COX-2, iNOS, TNF-α, IL-1β and IL-6 were significantly increased in H2O2-induced osteoblasts. In addition, the expression of these proinflammatory factors was attenuated by pretreatment of cells with monotropein, which suggests that monotropein presents a possible approach for the treatment of various inflammatory diseases.

In conclusion, the results of the present study demonstrate that monotropein suppresses the functional impairment of osteoblasts as a result of H2O2-induced oxidative stress, and its antioxidant properties may be responsible for these antioxidative effects. Furthermore, the observations of the present study indicate that the protective effects of monotropein may be mediated by the inhibition of apoptosis-associated markers and the activation of the NF-κB pathway. These results provide a novel insight into the protective effects of monotropein in osteoblasts via reducing ROS generation, and suggest that monotropein may be a potential therapeutic agent for the treatment of osteoporosis.

Acknowledgements

This study was funded by Xiaoshan Science and Technology Bureau Funds (grant no. 2013304).

References

1 

Reginster JY and Burlet N: Osteoporosis: A still increasing prevalence. Bone. 38(2): Suppl 1. S4–S9. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Landfeldt E, Ström O, Robbins S and Borgström F: Adherence to treatment of primary osteoporosis and its association to fractures-the Swedish Adherence Register Analysis (SARA). Osteoporosis Int. 23:433–443. 2012. View Article : Google Scholar

3 

Burghardt AJ, Kazakia GJ, Sode M, de Papp AE, Link TM and Majumdar S: A longitudinal HR-pQCT study of alendronate treatment in postmenopausal women with low bone density: Relations among density, cortical and trabecular microarchitecture, biomechanics and bone turnover. J Bone Miner Res. 25:2558–2571. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Kothawala P, Badamgarav E, Ryu S, Miller RM and Halbert RJ: Systematic review and meta-analysis of real-world adherence to drug therapy for osteoporosis. Mayo Clin Proc. 82:1493–1501. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Cramer J, Gold D, Silverman S and Lewiecki E: A systematic review of persistence and compliance with bisphosphonates for osteoporosis. Osteoporosis Int. 18:1023–1031. 2007. View Article : Google Scholar

6 

Siris ES, Harris ST, Rosen CJ, Barr CE, Arvesen JN, Abbott TA and Silverman S: Adherence to bisphosphonate therapy and fracture rates in osteoporotic women: Relationship to vertebral and nonvertebral fractures from 2 US claims databases. Mayo Clin Proc. 81:1013–1022. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Lean JM, Jagger CJ, Kirstein B, Fuller K and Chambers TJ: Hydrogen peroxide is essential for estrogen-deficiency bone loss and osteoclast formation. Endocrinology. 146:728–735. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Cencioni C, Spallotta F, Martelli F, Valente S, Mai A, Zeiher AM and Gaetano C: Oxidative stress and epigenetic regulation in ageing and age-related diseases. Int JMol Sci. 14:17643–17663. 2013. View Article : Google Scholar

9 

Martindale JL and Holbrook NJ: Cellular response to oxidative stress: Signaling for suicide and survival. J Cell Physiol. 192:1–15. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Manolagas SC: From estrogen-centric to aging and oxidative stress: A revised perspective of the pathogenesis of osteoporosis. Endocr Rev. 31:266–300. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Sánchez-Rodríguez MA, Ruiz-Ramos M, Correa-Muñoz E and Mendoza-Núñez VM: Oxidative stress as a risk factor for osteoporosis in elderly Mexicans as characterized by antioxidant enzymes. BMC Musculoskelet Disord. 8:1242007. View Article : Google Scholar : PubMed/NCBI

12 

Mody N, Parhami F, Sarafian TA and Demer LL: Oxidative stress modulates osteoblastic differentiation of vascular and bone cells. Free Radical Biol Med. 31:509–519. 2001. View Article : Google Scholar

13 

Lee DH, Lim BS, Lee YK and Yang HC: Effects of hydrogen peroxide (H2O2) on alkaline phosphatase activity and matrix mineralization of odontoblast and osteoblast cell lines. Cell Biol Toxicol. 22:39–46. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Maggio D, Barabani M, Pierandrei M, Polidori MC, Catani M, Mecocci P, Senin U, Pacifici R and Cherubini A: Marked decrease in plasma antioxidants in aged osteoporotic women: Results of a cross-sectional study. J Clin Endocri Metab. 88:1523–1527. 2003. View Article : Google Scholar

15 

Lee YJ, Hong JY, Kim SC, Joo JK, Na YJ and Lee KS: The association between oxidative stress and bone mineral density according to menopausal status of Korean women. Obstet Gyn Sci. 58:46–52. 2015. View Article : Google Scholar

16 

Ho CT and Zheng QY: Quality management of nutraceuticals. Am Chem Soc. 8032002.

17 

Choi J, Lee K, Choi MY, Nam JH, Jung HJ, Park SK and Park HJ: Antinociceptive anti-inflammatory effect of monotropein isolated from the root of Morinda officinalis. Biol Pharm Bull. 28:1915–1918. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Shin JS, Yun KJ, Chung KS, Seo KH, Park HJ, Cho YW, Baek NI, Jang D and Lee KT: Monotropein isolated from the roots of Morinda officinalis ameliorates proinflammatory mediators in RAW 264.7 macrophages and dextran sulfate sodium (DSS)-induced colitis via NF-κB inactivation. Food Chem Toxicol. 53:263–271. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Ishizuya T, Yokose S, Hori M, Noda T, Suda T, Yoshiki S and Yamaguchi A: Parathyroid hormone exerts disparate effects on osteoblast differentiation depending on exposure time in rat osteoblastic cells. J Clin Invest. 99:2961–2970. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Wang YK, Hong YJ, Wei M, Wu Y, Huang ZQ, Chen RZ and Chen HZ: Curculigoside attenuates human umbilical vein endothelial cell injury induced by H2O2. J Ethnopharmacol. 132:233–239. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Owen TA, Aronow M, Shalhoub V, Barone LM, Wilming L, Tassinari MS, Kennedy MB, Pockwinse S, Lian JB and Stein GS: Progressive development of the rat osteoblast phenotype in vitro: Reciprocal relationships in expression of genes associated with osteoblast proliferation and differentiation during formation of the bone extracellular matrix. J Cell Physiol. 143:420–430. 1990. View Article : Google Scholar : PubMed/NCBI

22 

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. View Article : Google Scholar : PubMed/NCBI

23 

Amer J, Goldfarb A and Fibach E: Flow cytometric measurement of reactive oxygen species production by normal and thalassaemic red blood cells. Eur J Haematol. 70:84–90. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Horii A, Wang X, Gelain F and Zhang S: Biological designer self-assembling peptide nanofiber scaffolds significantly enhance osteoblast proliferation, differentiation and 3-D migration. PLoS One. 2:e1902007. View Article : Google Scholar : PubMed/NCBI

25 

Mancino AT, Klimberg VS, Yamamoto M, Manolagas SC and Abe E: Breast cancer increases osteoclastogenesis by secreting M-CSF and upregulating RANKL in stromal cells. J Surg Res. 100:18–24. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Ly JD, Grubb DR and Lawen A: The mitochondrial membrane potential (deltapsi(m)) in apoptosis; an update. Apoptosis. 8:115–128. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Finkel T and Holbrook NJ: Oxidants, oxidative stress and the biology of ageing. Nature. 408:239–247. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Cabiscol E, Tamarit J and Ros J: Oxidative stress in bacteria and protein damage by reactive oxygen species. Int Microbiol. 3:3–8. 2000.PubMed/NCBI

29 

Cremers NA, Lundvig D, van Dalen S, Schelbergen RF, van Lent PL, Szarek WA, Regan RF, Carels CE and Wagener FA: Curcumin-induced heme oxygenase-1 expression prevents H2O2-induced cell death in wild type and heme oxygenase-2 knockout adipose-derived mesenchymal stem cells. Int J Mol Sci. 15:17974–17999. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Li K, Yang B and Zhao C: Transforming growth factor-β-activated kinase 1 enhances H2O2-induced apoptosis independently of reactive oxygen species in cardiomyocytes. J Cardiovasc Med (Hagerstown). 15:565–571. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Wei L, Yamaguchi H, Takeuchi R, Matsumoto H and Shibutani K: Propofol reduces hydrogen peroxide-induced apoptosis through down-regulating bim expression in alveolar epithelial cells. Int J Oral Med Sci. 11:274–279. 2013. View Article : Google Scholar

32 

Wang Y, Zhao L, Wang Y, Xu J, Nie Y, Guo Y, Tong Y, Qin L and Zhang Q: Curculigoside isolated from Curculigo orchioides prevents hydrogen peroxide-induced dysfunction and oxidative damage in calvarial osteoblasts. Acta Bioch Bioph Sin (Shanghai). 44:431–441. 2012. View Article : Google Scholar

33 

Ðokić-Lišanin M, Pantović V, Jovanović Z, Samardžić G and Jurišić V: Values of alkaline phosphathase and their isoenzyme profiles in patients with cancer in respect to bone and liver metastasis. Arch Oncol. 21:14–16. 2013. View Article : Google Scholar

34 

Peters E, Heemskerk S, Masereeuw R and Pickkers P: Alkaline phosphatase: A possible treatment for sepsis-associated acute kidney injury in critically ill patients. Am J Kidney Dis. 63:1038–1048. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Sargeant TD, Aparicio C, Goldberger JE, Cui H and Stupp SI: Mineralization of peptide amphiphile nanofibers and its effect on the differentiation of human mesenchymal stem cells. Acta Biomater. 8:2456–2465. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Zhou L, Chen X, Liu T, Gong Y, Chen S, Pan G, Cui W, Luo ZP, Pei M, Yang H and He F: Melatonin reverses H2 O2 -induced premature senescence in mesenchymal stem cells via the SIRT1-dependent pathway. J Pineal Res. 59:190–205. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Fu C, Xu D, Wang CY, Jin Y, Liu Q, Meng Q, Liu KX, Sun HJ and Liu MZ: Alpha-lipoic acid promotes osteoblastic formation in H2O2-treated MC3T3-E1 cells and prevents bone loss in ovariectomized rats. J Cell Physiol. 230:2184–2201. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Hume DA and MacDonald KP: Therapeutic applications of macrophage Colony-Stimulating Factor-1 (CSF-1) and antagonists of CSF-1 receptor (CSF-1R) signaling. Blood. 119:1810–1820. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Gow DJ, Garceau V, Kapetanovic R, Sester DP, Fici GJ, Shelly JA, Wilson TL and Hume DA: Cloning and expression of porcine Colony Stimulating Factor-1 (CSF-1) and Colony Stimulating Factor-1 Receptor (CSF-1R) and analysis of the species specificity of stimulation by CSF-1 and Interleukin 34. Cytokine. 60:793–805. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Sun B, Sun GB, Xiao J, Chen RC, Wang X, Wu Y, Cao L, Yang ZH and Sun XB: Isorhamnetin inhibits H2O2-induced activation of the intrinsic apoptotic pathway in H9c2 cardiomyocytes through scavenging reactive oxygen species and ERK inactivation. J Cell Biochem. 113:473–485. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Polidoro L, Properzi G, Marampon F, Gravina GL, Festuccia C, Di Cesare E, Scarsella L, Ciccarelli C, Zani BM and Ferri C: Vitamin D protects human endothelial cells from H2O2 oxidant injury through the Mek/Erk-Sirt1 axis activation. J Cardiovasc Transl Res. 6:221–231. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Liang D, Yang M, Guo B, Cao J, Yang L, Guo X, Li Y and Gao Z: Zinc inhibits H(2)O(2)-induced MC3T3-E1 cells apoptosis via MAPK and PI3K/AKT pathways. BiolTrace Elem Res. 148:420–429. 2012. View Article : Google Scholar

43 

Park BG, Yoo CI, Kim HT, Kwon CH and Kim YK: Role of mitogen-activated protein kinases in hydrogen peroxide-induced cell death in osteoblastic cells. Toxicology. 215:115–125. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Peng C, Perera PK, Li YM, Fang WR, Liu LF and Li FW: Anti-inflammatory effects of Clematis chinensis Osbeck extract(AR-6) may be associated with NF-κB, TNF-α and COX-2 in collagen-induced arthritis in rat. Rheumatol Int. 32:3119–3125. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Li M, Zhang L, Cai RL, Gao Y and Qi Y: Lipid-soluble extracts from Salvia miltiorrhiza inhibit production of LPS-induced inflammatory mediators via NF-κB modulation in RAW 264.7 cells and perform antiinflammatory effects in vivo. Phytother Res. 26:1195–1204. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 14 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu FB, Wang JY, Zhang YL, Hu YG, Yue ZS, Zeng LR, Zheng WJ, Hou Q, Yan SG, Quan RF, Quan RF, et al: Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteoblasts. Mol Med Rep 14: 5377-5384, 2016
APA
Zhu, F., Wang, J., Zhang, Y., Hu, Y., Yue, Z., Zeng, L. ... Quan, R. (2016). Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteoblasts. Molecular Medicine Reports, 14, 5377-5384. https://doi.org/10.3892/mmr.2016.5908
MLA
Zhu, F., Wang, J., Zhang, Y., Hu, Y., Yue, Z., Zeng, L., Zheng, W., Hou, Q., Yan, S., Quan, R."Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteoblasts". Molecular Medicine Reports 14.6 (2016): 5377-5384.
Chicago
Zhu, F., Wang, J., Zhang, Y., Hu, Y., Yue, Z., Zeng, L., Zheng, W., Hou, Q., Yan, S., Quan, R."Mechanisms underlying the antiapoptotic and anti-inflammatory effects of monotropein in hydrogen peroxide-treated osteoblasts". Molecular Medicine Reports 14, no. 6 (2016): 5377-5384. https://doi.org/10.3892/mmr.2016.5908