Open Access

Tanshinone‑IIA attenuates the deleterious effects of oxidative stress in osteoporosis through the NF‑κB signaling pathway

Retraction in: /10.3892/mmr.2023.13140

  • Authors:
    • Shaowen Zhu
    • Wanfu Wei
    • Zhiwei Liu
    • Yang Yang
    • Haobo Jia
  • View Affiliations

  • Published online on: March 14, 2018     https://doi.org/10.3892/mmr.2018.8741
  • Pages: 6969-6976
  • Copyright: © Zhu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Osteoclasts are responsible for bone resorption caused by bone microstructural damage and bone­related disorders. Evidence shows that tanshinone IIA (Tan‑IIA), a traditional Chinese medicine, is used clinically as a drug for the treatment of cardiovascular and cerebrovascular diseases. However, the efficacy and mechanism underlying the effect of Tan‑IIA on the viability of osteoclasts remain to be fully elucidated. The present study investigated the therapeutic effects of Tan‑IIA on osteoblast differentiation and oxidative stress in vitro and in vivo. Cell viability was analyzed and oxidative stress was examined in the osteoblasts. Wnt1sw/sw mice were used to investigate the therapeutic effects of Tan‑IIA on spontaneous tibia fractures and severe osteopenia. The bone strength, collagen and mineral were examined in the tibia. Osteoblast activity was also analyzed in the experimental mice. The Tan‑IIA‑induced differentiation of osteoclasts and the mechanism of action were investigated in osteocytes. The data showed that Tan‑IIA treatment improved cell viability. The data also demonstrated that Tan‑IIA decreased the levels of H2O2, accumulation of reactive oxygen species and apoptosis of osteoblasts. Tan‑IIA inhibited the deleterious outcomes triggered by oxidative stress. In addition, Tan‑IIA inhibited the activation of nuclear factor (NF)‑κB and its target genes, tumor necrosis factor (TNF)‑α, inducible nitric oxide synthase and cyclooxygenase 2, and increased the levels of TNF receptor‑associated factor 1 and inhibitor of apoptosis protein‑1/2 in the osteocytes. Furthermore, it was shown that Tan‑IIA reduced the propensity to fractures and severe osteopenia in mice with osteoporosis. Tan‑IIA also exhibited improved bone strength, mineral and collagen in the bone matrix of the experimental mice. It was found that the Tan‑IIA‑mediated benefits on osteoblast activity and function were through the NF‑κB signaling pathway. Taken together, the data obtained in the present study suggested that Tan‑IIA had protective effects against oxidative stress in osteoblastic differentiation in mice with osteoporosis by regulating the NF‑κB signaling pathway.

Introduction

Osteoporosis is a set of bone diseases caused by variety of factors featuring normal calcification, normal proportions of calcium salt and substrate, decreased bone quantity per unit volume, and metabolic bone lesions (1). Reports have shown that osteoporosis affects a higher proportion of individuals in the elderly population (2). Statistics indicate that osteoporosis is becoming a serious health problem in the aging population due to weakened bone structure and fractures in elderly individuals (3,4). The results of pathological investigations indicate that osteoclast activity and subtypes are crucial for the function of physiology and pathological implications for future treatment of osteoporosis (5). Osteoporosis readily leads to hip fractures, which are associated with functional impairment, maintenance of bone damage, loss of independence and increased mortality rates (6,7). Furthermore, clinical practice guidelines recommend that all patients with osteoporosis receive chronic oral glucocorticoid treatment. However, the therapeutic efficacy is limited in patients with osteoporosis who receive osteoporosis pharmacotherapy in terms of bone mineral density testing and bone strength. Therefore, examining more efficient molecular agents for the clinical treatment of osteoporosis is urgently required.

Tanshinone IIA (Tan-IIA) is a traditional Chinese medicine extracted from Danshen, which has been used clinically for the treatment of various human diseases (8,9). A previous study indicated that Tan-IIA decreased the protein expression of epidermal growth factor receptor and insulin-like growth factor 1 receptor, inhibiting the phosphoinositide 3-kinase/Akt/mammalian target of rapamycin pathway in gastric carcinoma in vitro and in vivo (10). In addition, Tan-IIA was shown to exert inhibitory effects on the hypoxia-induced enhancement of store-operated calcium entry in pulmonary arterial smooth muscle cells through the protein kinase G-proliferator-activated receptor-γ signaling pathway (11). The pharmacological and therapeutic properties of Tan-IIA in the cardiovascular system have attracted interest (12). A previous study showed that Tan-IIA treatment alleviated rat gingival connective tissue overgrowth induced by cyclosporine A (13). Emerging experimental investigations and clinical trials have demonstrated that Tan-IIA prevents cardiac injury, hypertrophy and atherogenesis through estrogen receptor-α-induced oxidative stress (14). Although the efficacy of Tan-IIA has been observed in different diseases (15,16), the efficacy and molecular mechanism underlying the effect of Tan-IIA have not been reported previously. In the present study, the effects and molecular mechanisms underlying the effects of Tan-IIA on osteocytes were evaluated in vitro and in vivo. The present study also examined the efficacy of Tan-IIA on anti-apoptotic effects in the treatment of osteoporosis. Current evidence suggests that Tan-IIA is an efficient anti-apoptotic agent in osteoporosis.

Oxidative and reductive stress are essential for the dynamic phases experienced by cells undergoing adaptation towards an endogenous or exogenous noxious stimulus (17). Mitochondrial malfunction is the common denominator arising from the aberrant functioning of the rheostat, which maintains homeostasis between oxidative and reductive stress in osteoblasts (18,19). Previous evidence suggests that the mechanism underlying osteoporosis is important for osteoclast activation in the treatment of osteoporosis. Maladaptation during oxidative stress may be pivotal in the pathophysiology of osteoporosis (20,21). A previous study provided evidence that targeting antioxidant drug therapy is an efficient clinical regimen for reducing the deterioration of osteoporosis in the aforementioned oxidative stress signaling pathway (22,23). Evidence has also revealed that endoplasmic reticulum stress is a key risk factor in the pathogenic progression of osteoporosis. Therefore, the present study investigated Tan-IIA-mediated oxidative stress in osteoblasts from mice with osteoporosis. The data showed that Tan-IIA may be an efficient anti-osteoporotic agent in vitro and in vivo.

The present study also investigated whether Tan-IIA has an anti-apoptotic and protective role in the treatment of osteoporosis in a mouse model. The data suggested that Tan-IIA ameliorated osteoblast activity and functioned through the nuclear factor (NF)-κB signaling pathway. Taken together, the data suggested that Tan-IIA exerted protective effects against oxidative stress in osteoblastic differentiation of mice with osteoporosis via regulation of the NF-κB signaling pathway and endoplasmic reticulum stress-dependent pathways, which may contribute to the treatment of osteoporosis.

Materials and methods

Ethics statement

The present study was performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of Taihe Hospital Affiliated to Hubei University of Medicine (Hubei, China). All experimental protocols on animals were performed in accordance with the National Institutes of Health (24) and approved by the Committee on the Ethics of Animal Experiments Defence Research of Tianjin Medical University General Hospital (Tianjin, China). All surgery and euthanasia were made to minimize suffering.

Animal experiments

A total of 64 female Wnt1sw/sw mice (10-week-old, 32–40 g) with osteoporosis were purchased from the Jackson Laboratory (Ben Harbor, ME, USA) on a mixed C57BL/6 background. All animals were fed under pathogen-free conditions. All animals were housed under controlled temperatures in a 12 h light/dark cycle with free access to food and water. The Wnt1sw/sw mice with osteoporosis were randomly divided into three groups (n=32 in each experimental group) and received treatment with Tan-IIA (10 mg/kg), Alendronate (ADN) or PBS. Treatments were performed via an intraperitoneal injection manner (200 µl) once a day. The detailed procedures were according to those of a previous report (25). The therapeutic efficacies of Tan-IIA were analyzed according to a previous study (26).

Reverse transcription-quantitative-polymerase chain reaction (RT-qPCR) analysis

Osteoblasts were isolated from experimental mice following treatment with Tan-IIA, ADN or PBS as described previously (27). Total RNA from the osteoblasts was extracted using an RNAeasy Mini kit (Qiagen Sciences, Inc., Gaithersburg, MD, USA) according to the manufacturer's protocols. The expression levels of B-cell lymphoma (Bcl)-2, alkaline phosphatase (ALP), p53, NF-κB, inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)2, tumor necrosis factor (TNF) receptor-associated factor 1 (TRAF-1), TNF-α, Caspase-3, Apaf-1, and inhibitor of apoptosis protein (IAP) in the osteoblasts were determined using RT-qPCR analysis (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). All forward and reverse primers were synthesized by Invitrogen; Thermo Fisher Scientific, Inc. (Table I). RT-qPCR was performed using a TaqPath™ 1-Step RT-qPCR Master Mix RT-qPCR kit (cat. no. A15300; Thermo Fisher Scientific, Inc.). Thermocycling conditions included 45 amplification cycles, denaturation at 95°C for 120 sec, primer annealing at 62.5°C for 30 sec with touchdown to 56.5°C for 45 sec and applicant extension at 72°C for 60 sec). Changes in relative mRNA expression levels were calculated using the 2−ΔΔCq method (28). The results are expressed as the n-fold change, compared with the control (β-actin).

Table I.

Sequences of primers in the present study.

Table I.

Sequences of primers in the present study.

Sequence

Gene nameReverseForward
P53 5′-TTAAGCTTTTTGCGTTCGGGCTGG-3′ 5-ATGGTGGCATGAACCTGTGG-3G
Bcl-2 5-CGTCATAACTAAAGACACCCC-3′ 5-TTCATCTCCAGTATCCGACT-3′
Caspase-3 5′-ATGGAGAACAACAAAACCTCAGT-3′ 5-TTGCTCCCATGTATGGTCTTTAC-3′
Apaf-1 5-AGCAAGTTGGTGTCATCCTCCGAT-3r 5-ATAGCAACAAAGCTCTCCGGTGGA-3r
ALP 5-CACCAATCACCTGCGGTACA-3G 5-CAGATCACGTCATCGCAC-3-
TNF-α 5′-CAATCCCTTTATTACCC-3′ 3′-GTCTTCTCAAGTCCTGC-3′
iNOS 5′-CTGCAGGTCTTTGACGCTCGG-3′ 5′-GTGGAACACAGGGGTGATG-3′
COX2 5′-TGAACACGGACTTGCTCACTTTG-3′ 5′-AGGCCTTTGCCACTGCTTGTA-3′
TRAF-1 5′-AGAACCCGAGGAATGGCGA-3′ 5′-TGAAGGAGCAGCCGACACC-3′
IAP 5′-GGCAGATTATGAAGCACGGATC-3′ 5′-GGCTTCCAATCAGTTAGCCCTC-3′
β-actin 5-ACGGTCAGGTCATCACTATCG-3′ 5′-GGCATAGAGGTCTTTACGGATG-3′

[i] Apaf-1, apoptotic protease-activating factor 1; ALP, alkaline phosphatase; Bcl-2, B-cell lymphoma-2; COX2, cyclooxygenase 2; IAP, inhibitor of apoptosis protein; iNOS, inducible nitric oxide synthase; TNF-α, tumor necrosis factor-α; TRAF-1, TNF receptor-associated factor 1.

Western blot analysis

The osteoblasts from the experimental mice with osteoporosis treated with Tan-IIA, ADN and PBS were homogenized in lysate buffer containing protease-inhibitor and were centrifuged at 8,000 × g at 4°C for 10 min. The supernatant of the mixture was used for analysis of proteins of interest. For detection of proteins, the transmembrane proteins were extracted using a transmembrane protein extraction kit (Qiagen Sciences, Inc.) according to the manufacturer's protocols. Protein concentrations were determined using a Bicinchoninic Acid protein assay (Pierce; Thermo Fisher Scientific, Inc.) and protein samples (40 µg) was separated by 12.5% polyacrylamide gel electrophoresis. Proteins were transferred onto a nitrocellulose membrane (Bio-Rad Laboratories, Inc., Hercules, CA, USA). The SDS assays were performed as previously described (29). For the western blot analysis, primary antibodies: p65 (1:1,000, ab16502; Abcam, Cambridge, UK), IKK-β (1:1,000, ab178870; Abcam), IκBα (1:1,000, ab109300; Abcam) and β-actin (1:1,000, ab8227; Abcam) were added following blocking (5% skimmed milk) for 1 h at 37°C, following which they were incubated with horseradish peroxidase-conjugated immunoglobulin G (cat. no. STAR206P; Bio-Rad Laboratories, Inc.) was used at a 1:5,000 dilution for 24 h at 4°C. The results were visualized using a chemiluminescence detection system.

Immunohistochemical staining

Bone tissues were obtained from experimental mice after treatment as described previously (30). Immunohistochemical staining was performed using an avidin-biotin-peroxidase technique. Paraffin-embedded tissue sections (4 µm) were prepared and epitope retrieval was performed for further analysis. The paraffinized sections were subjected to H2O2 (3%) for 10–15 min, and were subsequently blocked in regular blocking solution for 10–15 min at 37°C. Finally, the sections were incubated in anti-cluster of differentiation 31 (1:1,200, ab28364; Abcam), and anti-Ki67 (1:1,200, ab15580; Abcam), respectively, at 4°C for 12 h following blocking. All sections were washed three times and incubated with horseradish peroxidase-conjugated Immunoglobulin G (1:2,000, product code: ab97057; Abcam) for 1 h at 37°C and were observed in six randomly selected views under a fluorescence microscope at 488 nm (Olympus BX41; Olympus Corporation, Tokyo, Japan).

Oxidative stress assay

The examination of intracellular oxidative stress was performed using flow cytometry and DCFH-DA. The osteoblasts were harvested from the experimental mice on day 60, and trypsinized and labeled with DCFH-DA for 45 min at 37°C. Subsequently, the osteoblasts were treated with H2O2 in the presence Tan-IIA, ADN or PBS for 30 min at room temperature. The data were further analyzed with CellQuest Pro software (version 3.2; BD Biosciences, San Jose, CA, USA).

Phenotypic characterization of osteoblast differentiation

The osteoblasts were seeded at a density of 1×105 cells/cm2 for 12 h at 37°C. At 85% confluence, the cells were cultured in osteogenic medium (Invitrogen; Thermo Fisher Scientific, Inc.) containing 5% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) with Tan-IIA, ADN or PBS (1.5 mg/ml) in the presence of the indicated reagents. The procedures used for characterizing osteoblast differentiation were as described in a previous study (31).

Statistical analysis

All data are presented as the mean ± standard error of the mean. Unpaired data were analyzed using Student's t-test. Comparisons of data between multiple groups were analyzed using one-way analysis of variance followed by a Bonferroni post hoc test. Statistical analyses were performed using SPSS 19.0 (IBM SPSS, Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Tan-IIA ameliorates the decreased viability of osteoblasts and inhibits osteoclast differentiation

To investigate the effects of Tan-IIA on osteocyte growth, osteoblasts and osteoclasts were treated with Tan-IIA for 24 h. As shown in Fig. 1A and B, the results showed that Tan-IIA treatment increased the viability of the osteoblasts in a dose-dependent manner, compared with ADN and PBS. The viability of osteoblasts was also improved by Tan-IIA following treatment with increasing concentrations of H2O2 between 0.025 and 1.2 mM for 24 h (Fig. 1C). The present study also investigated the effects of Tan-IIA on osteoclastogenesis. The results (Fig. 1D) showed that Tan-IIA treatment inhibited osteoclast differentiation. The morphology of osteoclasts also confirmed the efficacy of Tan-IIA on osteoclast differentiation (Fig. 1E). Collectively, these results suggested that Tan-IIA not only ameliorated the decreased viability of osteoblasts, but effectively inhibited osteoclast differentiation.

Tan-IIA treatment inhibits the bone-resorbing activity and apoptosis of osteoclasts in vitro

The present study also investigated whether Tan-IIA affects the function of osteoblasts and osteoclasts. It was found that Tan-IIA treatment inhibited the apoptosis of osteoblasts induced by receptor activator of NF-κB ligand (RANKL), whereas the apoptosis of osteoclasts was promoted by treatment with Tan-IIA (Fig. 2A). In addition, the expression levels of caspase-3 and apoptotic protease-activating factor 1 were significantly upregulated in osteoclasts, but downregulated in osteoblasts (Fig. 2B and C). It was also observed that the expression levels of the anti-apoptotic genes Bcl-2 and p53 were increased and decreased in osteoblasts, respectively (Fig. 2D and E). Furthermore, Tan-IIA treatment significantly increased the bone-resorbing activity for osteoblasts, however, no effects were observed in osteoclasts in vitro (Fig. 2F). Taken together, these data suggested that Tan-IIA treatment was beneficial in bone-resorbing activity through regulation of the apoptosis of osteoblasts and osteoclasts.

Tan-IIA treatment shows beneficial effects in mice with osteoporosis

The biomechanical properties of Wnt1sw/sw bones were examined to assess the in vivo effects of Tan-IIA on osteoporosis. First, bone strength was assessed in experimental mice treated with Tan-IIA, ADN and PBS. The data showed that ADN increased bone strength, compared with that in the PBS group, however, Tan-IIA increased the bone strength of mice with osteoporosis, compared with that in the ADN and PBS groups (Fig. 3A-C). In addition, the present study analyzed the bone mineral and matrix composition of the experimental mice treated with Tan-IIA, ADN or PBS. The results (Fig. 3D) showed that Tan-IIA treatment led to an increasing ratio of proline to amide I. The relative mineral content was calculated by the ratio of phosphate to amide I, which was also increased in the Tan-IIA-treated Wnt1sw/sw mice (Fig. 3E). The evidence showed that Tan-IIA treatment increased the ratio of phosphate to proline in the Wnt1sw/sw mice (Fig. 3F). Overall, these data suggested that Tan-IIA was beneficial for the treatment of mice with osteoporosis by decreasing the mineral and collagen composition of the bone matrix.

Tan-IIA improves osteoporosis by regulating oxidative stress in osteoblasts from experimental mice

In order to analyze the efficacy of Tan-IIA in osteoblasts and osteoclasts from experimental mice treated with Tan-IIA, ADN and PBS, the present study analyzed oxidative stress in the mice with osteoporosis. The mRNA expression of ALP, a biomarker of osteoblastogenesis, was downregulated by Tan-IIA treatment for 24 h (Fig. 4A). The levels of H2O2 and accumulation of ROS were also decreased in the osteoblasts from the experimental mice (Fig. 4B and C). In addition, the results revealed that the expression levels of superoxide dismutase (SOD) and thiobarbituric acid reactive substances (TBARS) were decreased in the osteoblasts from the experimental mice treated with Tan-IIA, compared with those in mice treated with ADN or PBS (Fig. 4D and E). The results also demonstrated that the levels of reactive nitrogen species (RNS) were decreased in Tan-IIA-mediated oxidative stress in osteoblasts from the experimental mice (Fig. 4F). These data suggested that Tan-IIA inhibited the deleterious effects on the osteoblasts of experimental mice triggered by oxidative stress.

Tan-IIA improves osteoporosis via regulation of the NF-κB signaling pathway

To understand the mechanism underlying the Tan-IIA-induced suppression of oxidative stress in osteoblasts, the present study first analyzed the activity of NF-κB in osteocytes. The results (Fig. 5A) showed that Tan-IIA treatment inhibited the activity of NF-κB in osteocytes. Tan-IIA treatment also suppressed the activation of NF-κB phosphorylation in the osteoblasts (Fig. 5B). The levels of NF-κB target genes (TNF-α, iNOS and COX2) were decreased (Fig. 5C-E), and the levels of TRAF-1 and IAP were increased (Fig. 5F and G) in osteoblasts. The results of the western blot analysis revealed that the levels of p65, inhibitor of NF-κB (IκB)α and IκB kinase (IKK)-β were downregulated following treatment with Tan-IIA (Fig. 5H). Collectively, these results indicated that Tan-IIA improved osteoporosis by regulating the NF-κB signaling pathway.

Discussion

Osteoporosis is a comprehensive disease occurring in skeletal tissues, which is increasing in the aging population (32). The clinical consequences of osteoporosis include fractures of the upper extremities, hip and even spine, resulting in loss of function and independence, impaired quality of life, and increasing morbidity and mortality rates (33). Therefore, understanding the molecular mechanism of osteoporosis and identifying more efficient drugs for the treatment of osteoporosis are important for the clinical treatment of osteoporosis. In the present study, the efficacy and mechanism of Tan-IIA on osteocytes were investigated using a mice model of osteoporosis. In addition, osteocyte viability and osteoclast differentiation were examined to identify the therapeutic effects of Tan-IIA on osteoporosis. The effects of Tan-IIA on oxidative stress were investigated in vitro and in vivo. Notably, the mechanism underlying the Tan-IIA-mediated signaling pathway was analyzed. The data obtained suggested that Tan-IIA exhibited protective effects against oxidative stress in osteoblast differentiation in mice with osteoporosis via regulation of the NF-κB signaling pathway. The results also indicated that oxidative stress was important in the progression of osteoporosis and may be an efficient target, consistent with previous reports (34,35).

Previous reports have shown that the excessive accumulation of ROS and the subsequent activation of oxidative stress act as contributory functions in the development and progression of osteoporosis in the skeletal system (36,37). The involvement of the production of ROS in age-related osteoporosis and glucocorticoid-induced osteoporosis have been well documented (23). In addition, evidence has indicated that oxidative stress can damage various cellular components of osteoblasts, and contribute to the aggravation of osteoporosis (22,38). Oxidative stress is also considered an important pathogenic factor on bone mineral density and bone loss (20). Increasing evidence has shown that cell autophagy is important in the response to oxidative stress (21), and oxidative damage to osteoblasts can be considered a target for alleviating the condition of osteoporosis through the endoplasmic reticulum stress pathway (39). In the present study, the efficacy of Tan-IIA on the improvement of osteoblasts was confirmed, and it was also shown that Tan-IIA promoted osteoblastic differentiation through oxidative stress, which may be associated with changes in bone matrix formation and bone mineralization in the progression bone rarefaction.

Previous investigations have been performed to understand the Tan-IIA-associated therapeutic regimen via targeting specific molecules and disrupting the dopaminergic system, leading to various symptoms of cardiovascular diseases (40,41). However, the molecular mechanism of Tan-IIA in mediating the treatment of osteoporosis has not been investigated previously. Lee et al (42) suggested that the activation of NF-κB was associated with osteoclast differentiation, and their investigations indicated that the downregulation of RANKLinduced osteoclast differentiation assisted in the recovery of osteoporosis through inhibiting IκB degradation. In the present study, the findings indicated that Tan-IIA treatment may be a potential drug candidate for the treatment of osteoporosis. The results suggested that Tan-IIA inhibited the deleterious outcomes triggered by oxidative stress. In addition, Tan-IIA inhibited the activation of NF-κB and its target genes, TNF-α, iNOS and COX2, and increased the expression of TRAF-1 and IAP1/IAP2 in osteocytes.

In conclusion, the present study aimed to elucidate the underlying mechanisms responsible for Tan-IIA-mediated anti-apoptotic effects, and its effects on oxidative stress and signaling pathways. The investigation was extended to understand the Tan-IIA-mediated improvement of osteoporosis in a mouse model. The results confirmed the protective effects of Tan-IIA against oxidative stress and its beneficial effect on osteoblast differentiation. Extensive evidence has indicated oxidative stress as a novel mechanism, which contributes to the development and progression of osteoporosis, with its increase leading to degenerative disease in osteoporosis (43,44). The results of the present study confirmed those of previous reports, demonstrating that Tan-IIA ameliorated the apoptosis of osteoblasts and improved osteoblast function through the NF-κB signaling pathway. Future investigations of Tan-IIA may clarify the most promising outcomes to be investigated as a potential agent for osteoporosis.

Acknowledgements

Not applicable.

Funding

This study was supported by grant from the Natural Science Foundation of Tianjin (grant no. 2011TJH1104PU).

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

SZ is the executor for this paper and performed most of the experiments. WW designed the experiments. ZL, YY and HJ prepared the figures and conducted the data statistics.

Ethics approval and consent to participate

All experimental protocols on animals were performed in accordance with the National Institutes of Health (44) and approved by the Committee on the Ethics of Animal Experiments Defence Research of Tianjin Medical University General Hospital (Tianjin, China). All surgery and euthanasia were made to minimize suffering.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Cornelius C, Koverech G, Crupi R, Di Paola R, Koverech A, Lodato F, Scuto M, Salinaro AT, Cuzzocrea S, Calabrese EJ and Calabrese V: Osteoporosis and alzheimer pathology: Role of cellular stress response and hormetic redox signaling in aging and bone remodeling. Front Pharmacol. 5:1202014. View Article : Google Scholar : PubMed/NCBI

2 

Kondo T, Endo I and Matsumoto T: The pathology, diagnosis and therapy on osteoporosis. Nihon Rinsho. 72:1774–1779. 2014.(In Japanese). PubMed/NCBI

3 

Scott LJ: Denosumab: A review of its use in postmenopausal women with osteoporosis. Drugs Aging. 31:555–576. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Yu S, Liu F, Cheng Z and Wang Q: Association between osteoporosis and benign paroxysmal positional vertigo: A systematic review. BMC Neurol. 14:1102014. View Article : Google Scholar : PubMed/NCBI

5 

Henriksen K, Bollerslev J, Everts V and Karsdal MA: Osteoclast activity and subtypes as a function of physiology and pathology-implications for future treatments of osteoporosis. Endocr Rev. 32:31–63. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Sadat-Ali M, Al-Omran A, Al-Bakr W, Azam MQ, Tantawy A and Al-Othman A: Established osteoporosis and gaps in the management: Review from a teaching hospital. Ann Med Health Sci Res. 4:198–201. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Miyazaki T, Tokimura F and Tanaka S: A review of denosumab for the treatment of osteoporosis. Patient Prefer Adherence. 8:463–471. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Li F, Han G and Wu K: Tanshinone IIA alleviates the AD phenotypes in APP and PS1 transgenic mice. Biomed Res Int. 2016:76318012016.PubMed/NCBI

9 

Kim EO, Kang SE, Im CR, Lee JH, Ahn KS, Yang WM, Um JY, Lee SG and Yun M: Tanshinone IIA induces TRAIL sensitization of human lung cancer cells through selective ER stress induction. Int J Oncol. 48:2205–2212. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Su CC and Chiu TL: Tanshinone IIA decreases the protein expression of EGFR and IGFR blocking the PI3K/Akt/mTOR pathway in gastric carcinoma AGS cells both in vitro and in vivo. Oncol Rep. 36:1173–1179. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Jiang Q, Lu W, Yang K, Hadadi C, Fu X, Chen Y, Yun X, Zhang J, Li M, Xu L, et al: Sodium tanshinone IIA sulfonate inhibits hypoxia-induced enhancement of SOCE in pulmonary arterial smooth muscle cells via the PKG-PPAR-γ signaling axis. Am J Physiol Cell Physiol. 311:C136–C149. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Mao C, Zhang Y, Cao L, Shao H, Wang L, Zhu L and Xu Z: The effect of tanshinone IIA on the cardiovascular system in ovine fetus in utero. AM J Chin Med. 37:1031–1044. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Ma S, Liu W, Liu P, Liu J, Chen L and Qin C: Tanshinone IIA treatment alleviated the rat gingival connective tissue overgrowth induced by cyclosporine A. J Periodontal Res. 51:567–576. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Li DC, Bao XQ, Sun H and Zhang D: Research progress in the study of protective effect of tanshinone IIA on cerebral ischemic stroke. Yao Xue Xue Bao. 50:635–639. 2015.(In Chinese). PubMed/NCBI

15 

Ouyang DS, Huang WH, Chen D, Zhang W, Tan ZR, Peng JB, Wang YC, Guo Y, Hu DL, Xiao J and Chen Y: Kinetics of cytochrome P450 enzymes for metabolism of sodium tanshinone IIA sulfonate in vitro. Chin Med. 11:112016. View Article : Google Scholar : PubMed/NCBI

16 

Bai Y, Zhang L, Fang X and Yang Y: Tanshinone IIA enhances chemosensitivity of colon cancer cells by suppressing nuclear factor-κB. Exp Ther Med. 11:1085–1089. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Kurian GA, Rajagopal R, Vedantham S and Rajesh M: The role of oxidative stress in myocardial ischemia and reperfusion injury and remodeling: Revisited. Oxid Med Cell Longev. 2016:16564502016. View Article : Google Scholar : PubMed/NCBI

18 

Saito M: On ‘2015 guidelines for prevention and treatment of osteoporosis’. The mechanism of bone fragility. Clin Calcium. 25:1301–1306. 2015.(In Japanese). PubMed/NCBI

19 

Koga T and Takayanagi H: On ‘2015 guidelines for prevention and treatment of osteoporosis’. Cellular mechanism and etiology of osteoporosis. Clin Calcium. 25:1293–1300. 2015.(In Japanese). PubMed/NCBI

20 

Wu Q, Zhong ZM, Pan Y, Zeng JH, Zheng S, Zhu SY and Chen J: Advanced oxidation protein products as a novel marker of oxidative stress in postmenopausal osteoporosis. Med Sci. 21:2428–2432. 2015.

21 

Yang YH, Li B, Zheng XF, Chen JW, Chen K, Jiang SD and Jiang LS: Oxidative damage to osteoblasts can be alleviated by early autophagy through the endoplasmic reticulum stress pathway-implications for the treatment of osteoporosis. Free Radic Biol Med. 77:10–20. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Spilmont M, Leotoing L, Davicco MJ, Lebecque P, Mercier S, Miot-Noirault E, Pilet P, Rios L, Wittrant Y and Coxam V: Pomegranate and its derivatives can improve bone health through decreased inflammation and oxidative stress in an animal model of postmenopausal osteoporosis. Eur J Nutr. 53:1155–1164. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Wilson C: Bone: Oxidative stress and osteoporosis. Nat Rev Endocrinol. 10:32014. View Article : Google Scholar

24 

Couzin-Frankel J: National Institutes of Health. Needed: More females in animal and cell studies. Science. 344:6792014. View Article : Google Scholar : PubMed/NCBI

25 

Kushwaha P, Khedgikar V, Ahmad N, Karvande A, Gautam J, Kumar P, Maurya R and Trivedi R: A neoflavonoid dalsissooal isolated from heartwood of Dalbergia sissoo Roxb. has bone forming effects in mice model for osteoporosis. Eur J Pharmacol. 788:65–74. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Shum LC, White NS, Nadtochiy SM, Bentley KL, Brookes PS, Jonason JH and Eliseev RA: Cyclophilin D knock-out mice show enhanced resistance to osteoporosis and to metabolic changes observed in aging bone. PLoS One. 11:e01557092016. View Article : Google Scholar : PubMed/NCBI

27 

Gartland A, Rumney RM, Dillon JP and Gallagher JA: Isolation and culture of human osteoblasts. Methods Mol Biol. 806:337–355. 2012. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Wai-Hoe L, Wing-Seng L, Ismail Z and Lay-Harn G: SDS-PAGE-based quantitative assay for screening of kidney stone disease. Biol Proced Online. 11:pp. 145–160. 2009; View Article : Google Scholar : PubMed/NCBI

30 

Williams EL, White K and Oreffo RO: Isolation and enrichment of Stro-1 immunoselected mesenchymal stem cells from adult human bone marrow. Methods Mol Biol. 1035:67–73. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Elsafadi M, Manikandan M, Dawud RA, Alajez NM, Hamam R, Alfayez M, Kassem M, Aldahmash A and Mahmood A: Transgelin is a TGFβ-inducible gene that regulates osteoblastic and adipogenic differentiation of human skeletal stem cells through actin cytoskeleston organization. Cell Death Dis. 7:e23212016. View Article : Google Scholar : PubMed/NCBI

32 

Lambert MF, Cook JV, Roelant E, Bradshaw C, Foy R and Eccles MP: Feasibility of applying review criteria for depression and osteoporosis national guidance in primary care. Prim Health Care Res Dev. 15:396–405. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Ghosh M and Majumdar SR: Antihypertensive medications, bone mineral density and fractures: A review of old cardiac drugs that provides new insights into osteoporosis. Endocrine. 46:397–405. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Sanchez-Rodriguez 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

35 

Chavan SN, More U, Mulgund S, Saxena V and Sontakke AN: Effect of supplementation of vitamin C and E on oxidative stress in osteoporosis. Indian J Clin Biochem. 22:101–105. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Yin H, Shi ZG, Yu YS, Hu J, Wang R, Luan ZP and Guo DH: Protection against osteoporosis by statins is linked to a reduction of oxidative stress and restoration of nitric oxide formation in aged and ovariectomized rats. Eur J Pharmacol. 674:200–206. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Baek KH, Oh KW, Lee WY, Lee SS, Kim MK, Kwon HS, Rhee EJ, Han JH, Song KH, Cha BY, et al: Association of oxidative stress with postmenopausal osteoporosis and the effects of hydrogen peroxide on osteoclast formation in human bone marrow cell cultures. Calcif Tissue Int. 87:226–235. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Cervellati C, Bonaccorsi G, Cremonini E, Romani A, Fila E, Castaldini MC, Ferrazzini S, Giganti M and Massari L: Oxidative stress and bone resorption interplay as a possible trigger for postmenopausal osteoporosis. Biomed Res Int. 2014:5695632014. View Article : Google Scholar : PubMed/NCBI

39 

Yilmaz N and Eren E: Homocysteine oxidative stress and relation to bone mineral density in post-menopausal osteoporosis. Aging Clin Exp Res. 21:353–357. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Puntmann VO, Taylor PC and Mayr M: Coupling vascular and myocardial inflammatory injury into a common phenotype of cardiovascular dysfunction: Systemic inflammation and aging-a mini-review. Gerontology. 57:295–303. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Ramachandran A, Jha S and Lefer DJ: Review paper: Pathophysiology of myocardial reperfusion injury: The role of genetically engineered mouse models. Vet Pathol. 45:698–706. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Lee WS, Lee EG, Sung MS, Choi YJ and Yoo WH: Atorvastatin inhibits osteoclast differentiation by suppressing NF-κB and MAPK signaling during IL-1β-induced osteoclastogenesis. Korean J Intern Med. Mar 28–2017.(Epub ahead of print).

43 

Handzlik-Orlik G, Holecki M, Wilczynski K and Dulawa J: Osteoporosis in liver disease: Pathogenesis and management. Ther Adv Endocrinol Metab. 7:128–135. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Kim IH, Chung MY, Shin JY and Han D: Protective effects of black hoof medicinal mushroom from Κorea, Phellinus linteus (higher basidiomycetes), on osteoporosis in vitro and in vivo. Int J Med Mushrooms. 18:39–47. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2018
Volume 17 Issue 5

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 S, Wei W, Liu Z, Yang Y and Jia H: Tanshinone‑IIA attenuates the deleterious effects of oxidative stress in osteoporosis through the NF‑κB signaling pathway Retraction in /10.3892/mmr.2023.13140. Mol Med Rep 17: 6969-6976, 2018
APA
Zhu, S., Wei, W., Liu, Z., Yang, Y., & Jia, H. (2018). Tanshinone‑IIA attenuates the deleterious effects of oxidative stress in osteoporosis through the NF‑κB signaling pathway Retraction in /10.3892/mmr.2023.13140. Molecular Medicine Reports, 17, 6969-6976. https://doi.org/10.3892/mmr.2018.8741
MLA
Zhu, S., Wei, W., Liu, Z., Yang, Y., Jia, H."Tanshinone‑IIA attenuates the deleterious effects of oxidative stress in osteoporosis through the NF‑κB signaling pathway Retraction in /10.3892/mmr.2023.13140". Molecular Medicine Reports 17.5 (2018): 6969-6976.
Chicago
Zhu, S., Wei, W., Liu, Z., Yang, Y., Jia, H."Tanshinone‑IIA attenuates the deleterious effects of oxidative stress in osteoporosis through the NF‑κB signaling pathway Retraction in /10.3892/mmr.2023.13140". Molecular Medicine Reports 17, no. 5 (2018): 6969-6976. https://doi.org/10.3892/mmr.2018.8741