Open Access

Crocin protects against dexamethasone‑induced osteoblast apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway

  • Authors:
    • Zhigang Nie
    • Shuang Deng
    • Lei Zhang
    • Sen Chen
    • Qiang Lu
    • Hao Peng
  • View Affiliations

  • Published online on: May 22, 2019     https://doi.org/10.3892/mmr.2019.10267
  • Pages: 401-408
  • Copyright: © Nie 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

Osteoblast apoptosis has been identified as an important event in the development of glucocorticoid (GC)‑induced osteoporosis and osteonecrosis of the femoral head. Crocin, a bioactive ingredient of saffron, has been demonstrated to induce antiapoptotic effects on numerous types of cell in vitro; however, the effects of crocin on the dexamethasone (Dex)‑induced apoptosis of osteoblasts remain unclear. In the present study, the protective effects of crocin during Dex‑induced apoptosis of MC3T3‑E1 osteoblasts, and the underlying mechanisms, were investigated. MTT and Annexin V‑FITC/PI flow cytometry assays were performed to evaluate the viability and apoptosis of cells, respectively. The mitochondrial transmembrane potential, reactive oxygen species (ROS), intracellular Ca2+ levels and apoptosis‑associated protein expression were assessed via flow cytometry, fluorescence microscopy and western blotting. It was demonstrated that crocin pretreatment inhibited Dex‑induced apoptosis of osteoblasts in a dose‑dependent manner. Crocin reversed Dex‑induced decreases in the mitochondrial transmembrane potential, and increases in ROS and intracellular Ca2+ levels. Furthermore, crocin upregulated the expression levels of B‑cell lymphoma-2 (Bcl‑2) and mitochondrial cytochrome c (Cyt C), and downregulated those of cleaved caspase‑9, cleaved caspase‑3, Bcl‑2‑associated X protein and cytoplasmic Cyt C. N‑acetylcysteine, a ROS inhibitor, and 1,2‑bis(2‑aminophenoxy)ethane‑N,N,N',N'‑tetraacetic acid, a calcium chelator, attenuated Dex‑induced osteoblast apoptosis, whereas H2O2 and ionomycin, a calcium ionophore that increases intracellular calcium levels, reversed the antiapoptotic effects of crocin on Dex‑treated osteoblasts. These results indicated that crocin may protect osteoblasts from Dex‑induced apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway, thus suggesting that crocin has potential value as a treatment for GC‑induced bone diseases.

Introduction

Glucocorticoids (GCs) are widely used as treatments for various diseases (1), including systemic lupus erythematosus, idiopathic thrombocytopenic purpura and nephrotic syndrome; however, GC use can lead to numerous complications, the most serious of which are osteoporosis and osteonecrosis of the femoral head (ONFH) (2). Osteoblast apoptosis is regarded as an important pathogenic mechanism underlying these two complications (36). Consistent with these findings, previous studies have detected a large number of TUNEL-positive osteoblasts (apoptotic cells) in the femoral head of GC-treated rats (7,8). Therefore, the development of novel treatments that inhibit osteoblast apoptosis is required.

The role of reactive oxygen species (ROS) in osteoblast apoptosis has received considerable attention from researchers. Dai et al (9) revealed that H2O2 induces apoptosis in the Saos-2 osteoblastic cell line, which is attenuated by curcumin via increased protein kinase B-glycogen synthase kinase 3β signaling and preservation of mitochondrial function. Additionally, Linares et al (10) confirmed that apoptosis is induced in MC3T3-E1 cells by H2O2 and revealed that the effect is regulated by glutaredoxin 5. Li et al (11) reported that aluminum induces osteoblast apoptosis via the oxidative stress-mediated c-Jun N-terminal kinase (JNK) pathway. ROS serve roles in promoting apoptosis by inducing cytochrome c (Cyt C) release from the mitochondria to the cytosol (12). Furthermore, ROS have been reported to induce apoptosis of osteoblasts via activation of a protein kinase Cβ/p66shc/JNK signaling cascade (13). Intracellular Ca2+ is also involved in inducing apoptosis of various cell types (14,15); however, the role of Ca2+ in osteoblasts remains unclear. At present, only Nam et al (16) has reported that H2O2 increases intracellular Ca2+ levels in osteoblasts, subsequently inducing cell death.

Crocin (Fig. 1A) is a major bioactive component extracted from saffron, which has been reported to possess anticancer, anti-inflammatory, antioxidant and antiapoptotic properties (1720). As revealed by Santhosh et al (21), crocin provides notable protection against Vipera russelli venom-induced oxidative stress and neutrophil apoptosis. Additionally, Oruc et al (22) reported that crocin exhibits antiapoptotic and antioxidant effects on ischemia-reperfusion injury induced by four-vessel occlusion. The effects of crocin on intracellular Ca2+ signaling have received limited attention, with the exception of a study by Liu et al (23), which revealed that crocin decreases the L-type Ca2+ current and inhibits Ca2+ entry into cardiomyocytes, thereby exerting cardioprotective effects. Notably, crocin has been demonstrated to protect against ovariectomy-induced osteoporosis by inhibiting oxidative stress in a rat model (24). Therefore, it has been suggested that crocin may serve a protective role in osteoblasts. This study hypothesized that crocin may suppress dexamethasone (Dex)-induced osteoblast apoptosis by inhibiting the ROS/Ca2+-mediated mitochondrial pathway.

In the present study, the effects of crocin on Dex-induced osteoblast apoptosis and its underlying mechanisms were investigated. ROS and intracellular Ca2+ levels, and the activity of the mitochondrial apoptotic pathway, were determined following crocin administration in Dex-treated MC3T3-E1 osteoblasts.

Materials and methods

Materials

Crocin (cat. no. 17304), MTT (cat. no. M2128), N-acetyl-L-cysteine (NAC, cat. no. A7250), 1,2-bis(2-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid (BAPTA-AM; cat. no. 14510), H2O2 (cat. no. 88597), ionomycin (Ion; cat. no. 407952), and dimethyl sulfoxide (cat. no. 156914) were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). The purity of crocin was determined to be 98.06% via high-performance liquid chromatography conducted by the Department of Pharmacology of Wuhan University (Wuhan, China). Dex was acquired from Shanghai Aladdin Bio-Chem Technology Co., Ltd. (Shanghai, China, cat. no. D137736). An Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) kit was purchased from Nanjing KeyGen Biotech Co., Ltd. (Nanjing, China; cat. no. KGA108). JC-1 Assay (cat. no. C2006), ROS Assay (cat. no. S0033), Mitochondria Isolation (cat. no. C3601), Bicinchoninic Acid (BCA; cat. no. P0010) Assay and Caspase-3 Activity Assay kits (cat. no. C1116), and phenylmethylsulfonyl fluoride (PMSF; cat. no. ST506) were acquired from Beyotime Institute of Biotechnology (Shanghai, China). A Fluo-3 AM kit was purchased from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan; cat. no. F026). B-cell lymphoma-2 (Bcl-2; cat. no. 4223S), Bcl-2-associated X protein (Bax; cat. no. 2772T), cleaved caspase-3 (cat. no. 9664T), −8 (cat. no. 8592) and −9 (cat. no. 9509) antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Cyt C antibody was obtained from Wuhan Sanying Biotechnology (Wuhan, China; cat. no. 10993-1-AP). GAPDH antibody was acquired from Hangzhou Goodhere Biotechnology Co., Ltd. (Hangzhou, China; cat. no. AB-P-R 001). Cyt C oxidase IV (COX IV; cat. no. ab16056) antibody was purchased from Abcam (Cambridge, UK). Horseradish peroxidase-conjugated secondary antibodies were acquired from Boster Biological Technology (Pleasanton, CA, USA; cat. no. BA1054).

Cell culture

MC3T3-E1 osteoblasts were obtained from Wuhan Biofavor Biotech Services Co., Ltd. (Wuhan, China). Cells were cultured in α-minimal essential medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin at 37°C in an atmosphere containing 5% CO2.

Cell viability assay

Cells were cultured in 96-well plates at a density of 5×103 cells/well. Increasing concentrations of crocin (0, 5, 25, 100, 400, 1,000 and 4,000 µM) were added to the wells, and cells were incubated at 37°C for 24 or 48 h. Then, nontoxic concentrations of crocin were determined using an MTT assay and were selected for subsequent experiments. Three concentrations (5, 25 and 100 µM) were then used to investigate the protective effects of crocin against 1 µM Dex-induced cytotoxicity using an MTT assay. Cells were pretreated with 5, 25 and 100 µM crocin for 1 h, and then treated with 1 µM Dex for a further 24 or 48 h. Cells were incubated at 37°C. The MTT assay was conducted as follows: Following aforementioned treatment and incubation, MTT reagent (10 µl) was added to wells, and the plates were incubated at 37°C for 4 h. The medium was then discarded, and 150 µl dimethyl sulfoxide was added to the wells to dissolve the formazan crystals. The absorbance was detected at 568 nm using a microplate reader (Thermo Fisher Scientific, Inc.).

Apoptosis assay

An Annexin V-FITC/PI assay was used to determine the apoptosis of osteoblasts. Cells were pretreated with 5, 25 and 100 µM crocin for 1 h, and then treated with 1 µM Dex for a further 24 h. Cells were incubated at 37°C. Following treatment, cells were washed twice with PBS, and were then incubated with 5 µl Annexin V and 5 µl PI in the dark at room temperature for 15 min. Subsequently, the cells were subjected to flow cytometry (Beckmancoulter, Brea, CA, USA), and CytExpert 2.0 software (Beckmancoulter) was used to determine the percentage of apoptotic cells. Annexin V+/PI cells were designated as early apoptotic cells, whereas Annexin V+/PI+ cells were identified as late apoptotic cells. The total percentage of apoptotic cells was calculated by adding the percentage of early apoptotic cells to the percentage of late apoptotic cells.

Effects of increase and decrease of ROS and Ca2+

NAC, H2O2, BAPTA-AM, and Ion were added to cells to observe the effects of increases and decreases in ROS and Ca2+ on the mitochondrial transmembrane potential (Δψm), caspase-3 activity, ROS levels, Ca2+ levels and apoptotic rate of osteoblasts. Cells were pretreated with 100 µM Cro, 2 mM NAC, 20 µM BAP, 100 µM H2O2 or 0.5 µM Ion for 1 h at 37°C prior to treatment with 1 µM Dex for 24 h at 37°C. The effects of NAC and BAP on Dex-induced mitochondrial membrane potential (Δψm) changes, caspase-3 activation, osteoblast apoptosis, and ROS and Ca2+ levels were evaluated. The effects of H2O2 and Ion on the protective effects of Cro against Dex-induced Δψm changes, caspase-3 activation, osteoblast apoptosis, and ROS and Ca2+ levels were also evaluated.

Measurement of the Δψm

The Δψm was measured using the JC-1 Assay kit, according to the manufacturer's protocol. Briefly, following treatment, cells were incubated with JC-1 solution (500 µl) at 37°C for 20 min and were then centrifuged at 13,500 × g for 3 min at 4°C. Subsequently, the cells were washed and resuspended in 1X incubation buffer (provided in the assay kit) three times. Finally, the Δψm was determined by flow cytometry. The JC-1 polymer/monomer fluorescence ratio was used to quantify the Δψm.

ROS detection

ROS levels were determined via two methods using the ROS Assay kit: Flow cytometry and fluorescence microscopy. Briefly, following treatment, cells were incubated with dichlorodihydrofluorescein diacetate solution (10 µM) at 37°C for 20 min. Cells were then washed three times with serum-free medium and washed twice with PBS. Finally, a flow cytometer was used to quantify the fluorescence intensity as a measure of ROS production, and data were analyzed using CytExpert 2.0 software. A fluorescence microscope (Olympus Corporation, Tokyo, Japan) and cellSens Entry 1.17 software (Olympus Corporation) was used to observe intracellular ROS fluorescence.

Intracellular Ca2+ detection

The Ca2+ dye Fluo-3 AM was used to determine intracellular Ca2+ levels. Two methods, flow cytometry and fluorescence microscopy, were employed. Following treatment, cells were incubated with Fluo-3 AM solution (final concentration, 5 µM) at 37°C for 30 min. The cells were then washed twice with PBS, and the Ca2+-dependent fluorescence intensity was determined using a flow cytometer and CytExpert 2.0 software. Fluorescence images were visualized under a fluorescence microscope (Olympus Corporation, Tokyo, Japan) using cellSens Entry 1.17 software.

Caspase-3 activity assay

Caspase-3 activity in cells was determined using a Caspase-3 Activity Assay kit, according to the manufacturer's protocols. Luminescence was measured at 405 nm using a microplate reader (Thermo Fisher Scientific, Inc.).

Western blotting

A Mitochondria Isolation kit was used to isolate mitochondria for analysis of mitochondrial Cyt C expression, according to the manufacturer's protocol. Following treatment, cells were homogenized on ice in cell lysis buffer (RIPA buffer; Beyotime Institute of Biotechnology) containing PMSF and centrifuged at 13,500 × g for 15 min at 4°C. Subsequently, protein concentrations were determined using a BCA kit. Equal quantities of total protein (50 µg/lane) were separated by SDS-PAGE (separation gel, 15%; stacking gel, 5%) and transferred to polyvinylidene fluoride membranes. Membranes were blocked with 5% non-fat dried milk in TBS-0.1% Tween-20 at room temperature for 2 h. incubated with primary antibodies against Bax (1:1,000), Bcl-2 (1:1,000), cleaved caspase-3 (1:1,000), cleaved caspase-8 (1:1,000), cleaved caspase-9 (1:1,000), Cyt C (1:1,000), COX IV (1:2,000) and GAPDH (1:1,000) overnight at 4°C. Subsequently, membranes were incubated with horseradish peroxidas-conjugated secondary antibodies (1:50,000) at 37°C for 2 h. Protein bands were visualized using enhanced chemiluminescence (Thermo Fisher Scientific, Inc.) and the optical density of protein bands was detected using BandScan 5.0 software (Glyko, Inc.; BioMarin Pharmaceutical, Inc., Novato, CA, USA).

Statistical analysis

Data are presented as the means ± standard deviation of three independent experiments. Data were analyzed using GraphPad Prism 5.0 software (GraphPad Software, Inc., La Jolla, CA, USA). Statistical significance was evaluated by one-way analysis of variance followed by a Tukey-Kramer test for post hoc comparisons. P<0.05 was considered to indicate a statistically significant difference.

Results

Crocin protects osteoblasts against Dex-induced cytotoxicity and apoptosis

The viability of MC3T3-E1 osteoblasts following treatment with various concentrations of crocin was investigated to determine a nontoxic concentration range. As presented in Fig. 1B, crocin did not exhibit cytotoxic effects on osteoblasts at concentrations ≤400 µM. Subsequently, the protective effects of crocin against Dex-treated MC3T3-E1 osteoblasts were determined. As presented in Fig. 1C, the viability of osteoblasts at 24 h was increased from 59.9±1.6% following treatment with 1 µM Dex alone, to 65.1±6.7, 76.2±5.0 and 85.8±4.9% following treatment with Dex + 5, 25 and 100 µM crocin, respectively (P<0.05). There was no notable difference in cell viability following incubation for 24 or 48 h. Similarly, it was revealed that the percentage of apoptotic cells at 24 h was significantly decreased from 27.0±2.9% following incubation with 1 µM Dex alone, to 17.6±1.6, 11.6±1.4 and 8.97±0.9% following treatment with Dex + 5, 25 and 100 µM crocin, respectively (Fig. 1D; P<0.05).

Crocin protects osteoblasts against Dex-induced apoptosis by inhibiting the mitochondrial apoptotic pathway

The Δψm and expression of mitochondrial apoptotic pathway-associated proteins were investigated. As presented in Fig. 2A and B, Dex significantly reduced the Δψm (JC-1 polymer/monomer fluorescence ratio) compared with in the control group; however, crocin pretreatment reversed the effects of Dex in a dose-dependent manner. Additionally, Dex significantly increased the expression levels of cleaved caspase-9 and cleaved caspase-3 compared with in the control groups; these effects were significantly attenuated by crocin pretreatment. Conversely, Dex and crocin did not induce a significant effect on cleaved caspase-8 expression (Fig. 2C and D). Mitochondrial Cyt C levels were significantly decreased and Cyt C levels were significantly increased following Dex treatment compared with in the control group (Fig. 2E and F); crocin significantly reversed these effects. The relative expression levels of Bax and Bcl-2 exhibited similar alterations; Bax expression was increased and Bcl-2 expression was decreased by Dex, whereas these effects were reversed by crocin.

ROS and intracellular Ca2+ are involved in the protective effects of crocin on Dex-treated osteoblasts

The roles of ROS and intracellular Ca2+ in the protective effects of crocin on Dex-treated osteoblasts were investigated. It was demonstrated that Dex significantly increased ROS and intracellular Ca2+ levels compared with in the control group, whereas crocin pretreatment significantly inhibited these effects in a dose-dependent manner (Fig. 3).

Crocin induces antiapoptotic effects on Dex-treated osteoblasts via ROS/Ca2+ signaling

As presented in Fig. 4A-C, treatment with NAC or BAPTA-AM attenuated Dex-induced apoptosis, loss of the Δψm and activation of caspase-3 in osteoblasts. Furthermore, it was demonstrated that H2O2 and Ion attenuated the protective effects of crocin on Dex-induced apoptosis, alterations in the Δψm and caspase-3 activation (Fig. 4D-F). The results indicated that the protective effects of crocin were mediated via alterations in intracellular Ca2+ and ROS levels.

Association between ROS and intracellular Ca2+ in Dex- and crocin-treated osteoblasts

The association between ROS and intracellular Ca2+ in Dex- and crocin-treated osteoblasts was further investigated. As presented in Fig. 5A-C, NAC and BAPTA-AM significantly decreased Dex-induced ROS generation and intracellular Ca2+ accumulation compared with Dex treatment alone. Additionally, as presented in Fig. 5D-F, H2O2 and Ion treatment significantly attenuated the protective effects of crocin on Dex-induced ROS generation and intracellular Ca2+ accumulation. The results suggested that ROS and intracellular Ca2+ levels may be associated and collectively contribute to apoptosis.

Discussion

Osteoblast apoptosis remains a significant cause of GC-induced osteoporosis and ONFH (25,26). Crocin has been reported to exert antioxidative and antiapoptotic effects (27,28). Cao et al (24) revealed that crocin ameliorates ovariectomy-induced osteoporosis in rats by inhibiting oxidative stress; however, it is yet to be determined whether crocin exerts protective effects against Dex-induced osteoblast apoptosis. In the present study, it was observed that crocin significantly inhibited Dex-induced osteoblast apoptosis in a dose-dependent manner, thus suggesting that crocin may be considered a potential natural treatment for GC-induced bone diseases.

Numerous studies have reported that the antiapoptotic effects of crocin protect various tissues and organs (19,2931), whereas others have observed that its proapoptotic effects promote apoptosis in tumor cells (3234). Therefore, crocin appears to exhibit antiapoptotic and proapoptotic properties; however, the dose ranges of crocin used in these studies may be responsible for these varied effects, as doses <500 µM tend to induce antiapoptotic effects, whereas those >500 µM induce proapoptotic effects. The present findings were similar; concentrations ≤400 µM did not exhibit toxicity, whereas those >1,000 µM significantly reduced osteoblast viability.

To identify the mechanisms underlying the antiapoptotic effects of crocin on Dex-induced apoptosis of osteoblasts, the mitochondrial apoptotic pathway was investigated. The results revealed that Dex exposure decreased the Δψm, whereas crocin treatment reversed this effect in a dose-dependent manner. In addition, Dex activated caspase-9, but did not alter caspase-8 activity, suggesting that the mitochondrial pathway, but not the death receptor-mediated pathway, contributed to Dex-induced osteoblast apoptosis. These results were consistent with the findings of Li et al (35). Furthermore, it was demonstrated that crocin treatment attenuated Dex-induced caspase-9 activation, suggesting that crocin inhibited the mitochondrial apoptotic pathway. Loss of the Δψm is associated with release of Cyt C from the mitochondria to the cytosol, subsequently leading to the activation of caspase-3 and apoptosis (36). A decrease in the Bcl-2/Bax ratio can induce loss of the Δψm (37,38). Consistent with these findings, the results of the present study indicated that Cyt C translocated from the mitochondria to the cytosol following Dex treatment, and that crocin attenuated this effect. The expression levels of Bcl-2, Bax and cleaved caspase-3 in the present study also supported the hypothesis that crocin may suppress the mitochondrial apoptosis pathway in Dex-treated osteoblasts.

ROS, which are primarily generated in the mitochondria, induce loss of the Δψm and serve an important role in osteoblast apoptosis (11,39). Almeida et al (13) observed elevated ROS levels and increased apoptosis in Dex-treated UAMS-32 osteoblasts; however, these effects are inhibited by the antioxidant NAC. The present study also revealed that ROS was involved in Dex-induced osteoblast apoptosis, and that crocin attenuated ROS generation. Inhibition of ROS with NAC suppressed Dex-induced apoptosis. Furthermore, H2O2 suppressed the antiapoptotic effects of crocin on Dex-treated osteoblasts. Intracellular Ca2+ overload has also been reported to lead to loss of the Δψm and the induction of apoptosis (40,41). Pretreatment with the calcium chelator BAPTA-AM partially suppresses apoptosis (42). Similarly, it was observed in the present study that Dex increased intracellular Ca2+ concentrations, and that crocin reversed the effect. Notably, BAPTA-AM also suppressed Dex-induced apoptosis, whereas the calcium ionophore Ion reversed the antiapoptotic effects of crocin on Dex-treated osteoblasts. Zhang et al (43) reported that NAC and BAPTA-AM suppress the eicosapentaenoic acid-induced apoptosis of HepG2 cells, and suggested the involvement of the ROS-Ca2+-JNK mitochondrial pathways. Based on the present findings, it was hypothesized that crocin may induce antiapoptotic effects on Dex-induced osteoblasts by inhibiting the ROS/Ca2+-mediated mitochondrial pathway.

The results of the present study suggested that ROS and intracellular Ca2+ levels are associated in Dex-treated cells or Dex- and crocin-treated cells. Notably, treatment with H2O2 or NAC also affected intracellular Ca2+ levels, whereas treatment with Ion or BAPTA-AM also affected ROS levels. Furthermore, a number of studies have reported that ROS contributes to intracellular Ca2+ overload (16,44), and other studies have demonstrated that intracellular Ca2+ overload leads to increased ROS production (45,46). Wang et al (44) suggested that oxidative stress decreases the efficiency of ATPase, thus contributing to voltage-gated calcium ion influx and subsequently apoptosis. Lipton and Nicotera (45) suggested that cytosolic Ca2+ overload leads to depolarization of the mitochondria, subsequently contributing to the accumulation of ROS; however, the potential mechanisms are complex and requires further investigation.

In conclusion, crocin exerted protective effects against apoptosis in Dex-induced MC3T3-E1 osteoblasts. Inactivation of the ROS/Ca2+-mediated mitochondrial pathway may be involved in the inhibitory effects of crocin on osteoblast apoptosis. The present study may promote further investigation into the application of crocin as a treatment for GC-induced osteoporosis and ONFH.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the National Natural Science Foundation of China (grant no. 81672154).

Availability of data and materials

All data generated and/or analyzed during this study are included in this published article.

Author's contributions

ZN, SD and HP designed the study. ZN, SD, LZ, QL and SC performed the experiments. ZN, SD and LZ performed data analysis. ZN drafted the manuscript. ZN, SD, LZ and HP revised the manuscript. All authors reviewed the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Deng S, Dai G, Chen S, Nie Z, Zhou J, Fang H and Peng H: Dexamethasone induces osteoblast apoptosis through ROS-PI3K/AKT/GSK3β signaling pathway. Bio Pharmacother. 110:602–608. 2019. View Article : Google Scholar

2 

Feng Z, Zheng W, Tang Q, Cheng L, Li H, Ni W and Pan X: Fludarabine inhibits STAT1-mediated up-regulation of caspase-3 expression in dexamethasone-induced osteoblasts apoptosis and slows the progression of steroid-induced avascular necrosis of the femoral head in rats. Apoptosis. 22:1001–1012. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Weinstein RS, Jilka RL, Parfitt AM and Manolagas SC: Inhibition of osteoblastogenesis and promotion of apoptosis of osteoblasts and osteocytes by glucocorticoids. Potential mechanisms of their deleterious effects on bone. J Clin Invest. 102:274–282. 1998. View Article : Google Scholar : PubMed/NCBI

4 

Zalavras C, Shah S, Birnbaum MJ and Frenkel B: Role of apoptosis in glucocorticoid-induced osteoporosis and osteonecrosis. Crit Rev Eukaryot Gene Expr. 13:221–235. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Kerachian MA, Séguin C and Harvey EJ: Glucocorticoids in osteonecrosis of the femoral head: A new understanding of the mechanisms of action. J Steroid Biochem Mol Biol. 114:121–128. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Chen F, Zhang L, OuYang Y, Guan H, Liu Q and Ni B: Glucocorticoid induced osteoblast apoptosis by increasing E4BP4 expression via up-regulation of Bim. Calcif Tissue Int. 94:640–647. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Chen S, Li J, Peng H, Zhou J and Fang H: Administration of erythropoietin exerts protective effects against glucocorticoid-induced osteonecrosis of the femoral head in rats. Int J Mol Med. 33:840–848. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Zheng H, Yang E, Peng H, Li J, Chen S, Zhou J, Fang H, Qiu B and Wang Z: Gastrodin prevents steroid-induced osteonecrosis of the femoral head in rats by anti-apoptosis. Chin Med J (Engl). 127:3926–3931. 2014.PubMed/NCBI

9 

Dai P, Mao Y, Sun X, Li X, Muhammad I, Gu W, Zhang D, Zhou Y, Ni Z, Ma J and Huang S: Attenuation of oxidative stress-induced osteoblast apoptosis by curcumin is associated with preservation of mitochondrial functions and increased Akt-GSK3β signaling. Cell Physiol Biochem. 41:661–677. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Linares GR, Xing W, Govoni KE, Chen ST and Mohan S: Glutaredoxin 5 regulates osteoblast apoptosis by protecting against oxidative stress. Bone. 44:795–804. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Li X, Han Y, Guan Y, Zhang L, Bai C and Li Y: Aluminum induces osteoblast apoptosis through the oxidative stress-mediated JNK signaling pathway. Biol Trace Elem Res. 150:502–508. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Ding G, Zhao J and Jiang D: Allicin inhibits oxidative stress-induced mitochondrial dysfunction and apoptosis by promoting PI3K/AKT and CREB/ERK signaling in osteoblast cells. Exp Ther Med. 11:2553–2560. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Almeida M, Han L, Ambrogini E, Weinstein RS and Manolagas SC: Glucocorticoids and tumor necrosis factor alpha increase oxidative stress and suppress Wnt protein signaling in osteoblasts. J Biol Chem. 286:44326–44335. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Li L, Tan H, Gu Z, Liu Z, Geng Y, Liu Y, Tong H, Tang Y, Qiu J and Su L: Heat stress induces apoptosis through a Ca2+-mediated mitochondrial apoptotic pathway in human umbilical vein endothelial cells. PLoS One. 9:e1110832014. View Article : Google Scholar : PubMed/NCBI

15 

Wang CL, Xia Y, Nie JZ, Zhou M, Zhang RP, Niu LL, Hou LH and Cao XH: Musca domestica larva lectin induces apoptosis in BEL-7402 cells through a Ca(2+)/JNK-mediated mitochondrial pathway. Cell Biochem Biophys. 66:319–329. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Nam SH, Jung SY, Yoo CM, Ahn EH and Suh CK: H2O2 enhances Ca2+ release from osteoblast internal stores. Yonsei Med J. 43:229–235. 2002. View Article : Google Scholar : PubMed/NCBI

17 

Hoshyar R and Mollaei H: A comprehensive review on anticancer mechanisms of the main carotenoid of saffron, crocin. J Pharm Pharmacol. 69:1419–1427. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Yarijani ZM, Pourmotabbed A, Pourmotabbed T and Najafi H: Crocin has anti-inflammatory and protective effects in ischemia-reperfusion induced renal injuries. Iran J Basic Med Sci. 20:753–759. 2017.PubMed/NCBI

19 

Ben Salem I, Boussabbeh M, Kantaoui H, Bacha H and Abid-Essefi S: Crocin, the main active saffron constituent, mitigates dichlorvos-induced oxidative stress and apoptosis in HCT-116 cells. Biomed Pharmacother. 82:65–71. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Yang X, Huo F, Liu B, Liu J, Chen T, Li J, Zhu Z and Lv B: Crocin inhibits oxidative stress and pro-inflammatory response of microglial cells associated with diabetic retinopathy through the activation of PI3K/Akt signaling pathway. J Mol Neurosci. 61:581–589. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Santhosh MS, Sundaram MS, Sunitha K, Jnaneshwari S, Devaraja S, Kemparaju K and Girish KS: Propensity of crocin to offset Vipera russelli venom induced oxidative stress mediated neutrophil apoptosis: A biochemical insight. Cytotechnology. 68:73–85. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Oruc S, Gönül Y, Tunay K, Oruc OA, Bozkurt MF, Karavelioğlu E, Bağcıoğlu E, Coşkun KS and Celik S: The antioxidant and antiapoptotic effects of crocin pretreatment on global cerebral ischemia reperfusion injury induced by four vessels occlusion in rats. Life Sci. 154:79–86. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Liu T, Chu X, Wang H, Zhang X, Zhang Y, Guo H, Liu Z, Dong Y, Liu H, Liu Y, et al: Crocin, a carotenoid component of Crocus cativus, exerts inhibitory effects on L-type Ca(2+) current, Ca(2+) transient, and contractility in rat ventricular myocytes. Can J Physiol Pharmacol. 94:302–308. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Cao PC, Xiao WX, Yan YB, Zhao X, Liu S, Feng J, Zhang W, Wang J, Feng YF and Lei W: Preventive effect of crocin on osteoporosis in an ovariectomized rat model. Evid Based Complement Alternat Med. 2014:8251812014. View Article : Google Scholar : PubMed/NCBI

25 

Zhang Z, Jin A and Yan D: MicroRNA206 contributes to the progression of steroidinduced avascular necrosis of the femoral head by inducing osteoblast apoptosis by suppressing programmed cell death 4. Mol Med Rep. 17:801–808. 2018.PubMed/NCBI

26 

Yun SI, Yoon HY, Jeong SY and Chung YS: Glucocorticoid induces apoptosis of osteoblast cells through the activation of glycogen synthase kinase 3beta. J Bone Miner Metab. 27:140–148. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Dianat M, Radan M, Badavi M, Mard SA, Bayati V and Ahmadizadeh M: Crocin attenuates cigarette smoke-induced lung injury and cardiac dysfunction by anti-oxidative effects: The role of Nrf2 antioxidant system in preventing oxidative stress. Respir Res. 19:582018. View Article : Google Scholar : PubMed/NCBI

28 

Razavi BM, Hosseinzadeh H, Abnous K, Khoei A and Imenshahidi M: Protective effect of crocin against apoptosis induced by subchronic exposure of the rat vascular system to diazinon. Toxicol Ind Health. 32:1237–1245. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Yousefsani BS, Mehri S, Pourahmad J and Hosseinzadeh H; Crocin prevents sub-cellular organelle damage, proteolysis apoptosis in rat hepatocytes, : A justification for its hepatoprotection. Iran J Pharm Res. 17:553–562. 2018.PubMed/NCBI

30 

Boussabbeh M, Prola A, Ben Salem I, Guilbert A, Bacha H, Lemaire C and Abis-Essefi S: Crocin and quercetin prevent PAT-induced apoptosis in mammalian cells: Involvement of ROS-mediated ER stress pathway. Environ Toxicol. 31:1851–1858. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Thushara RM, Hemshekhar M, Santhosh MS, Jnaneshwari S, Nayaka SC, Naveen S, Kemparaju K and Girish KS: Crocin, a dietary additive protects platelets from oxidative stress-induced apoptosis and inhibits platelet aggregation. Mol Cell Biochem. 373:73–83. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Amin A, Bajbouj K, Koch A, Gandesiri M and Schneider-Stock R: Defective autophagosome formation in p53-null colorectal cancer reinforces crocin-induced apoptosis. Int J Mol Sci. 16:1544–1561. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Rezaee R, Jamialahmadi K, Riahi Zanjani B, Mahmoudi M, Abnous K, Zamani Taghizadeh Rabe S, Tabasi N, Zali M, Rezaee M, Amin B and Karimi G: Crocin effects on human myeloma cells regarding intracellular redox state, DNA fragmentation, and apoptosis or necrosis profile. Jundishapur J Nat Pharm Prod. 9:e201312014. View Article : Google Scholar : PubMed/NCBI

34 

Hoshyar R, Bathaie SZ and Sadeghizadeh M: Crocin triggers the apoptosis through increasing the Bax/Bcl-2 ratio and caspase activation in human gastric adenocarcinoma, AGS, cells. DNA Cell Biol. 32:50–57. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Li J, He C, Tong W, Zou Y, Li D, Zhang C and Xu W: Tanshinone IIA blocks dexamethasone-induced apoptosis in osteoblasts through inhibiting Nox4-derived ROS production. Int J Clin Exp Pathol. 8:13695–13706. 2015.PubMed/NCBI

36 

Bak DH, Kim HD, Kim YO, Park CG, Han SY and Kim JJ: Neuroprotective effects of 20(S)-protopanaxadiol against glutamate-induced mitochondrial dysfunction in PC12 cells. Int J Mol Med. 37:378–386. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Lv R, Du L, Lu C, Wu J, Ding M, Wang C, Mao N and Shi Z: Allicin protects against H2O2-induced apoptosis of PC12 cells via the mitochondrial pathway. Exp Ther Med. 14:2053–2059. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Giménez-Cassina A and Danial NN: Regulation of mitochondrial nutrient and energy metabolism by BCL-2 family proteins. Trends Endocrinol Metab. 26:165–175. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Gan X, Huang S, Yu Q, Yu H and Yan SS: Blockade of Drp1 rescues oxidative stress-induced osteoblast dysfunction. Biochem Biophys Res Commun. 468:719–725. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Assaf H, Azouri H and Pallardy M: Ochratoxin A induces apoptosis in human lymphocytes through down regulation of Bcl-xL. Toxicol Sci. 79:335–344. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Liu L, Wang D, Wang J and Wang S: The nitric oxide prodrug JS-K induces Ca(2+)-mediated apoptosis in human hepatocellular carcinoma HepG2 cells. J Biochem Mol Toxicol. 30:192–199. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Wang J, Zhu H, Liu X and Liu Z: Oxidative stress and Ca(2+) signals involved on cadmium-induced apoptosis in rat hepatocyte. Biol Trace Elem Res. 161:180–189. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Zhang Y, Han L, Qi W, Cheng D, Ma X, Hou L, Cao X and Wang C: Eicosapentaenoic acid (EPA) induced apoptosis in HepG2 cells through ROS-Ca(2+)-JNK mitochondrial pathways. Biochem Biophys Res Commun. 456:926–932. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Wang W, Zheng LL, Wang F, Hu ZL, Wu WN, Gu J and Chen JG: Tanshinone IIA attenuates neuronal damage and the impairment of long-term potentiation induced by hydrogen peroxide. J Ethnopharmacol. 134:147–155. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Lipton SA and Nicotera P: Calcium, free radicals and excitotoxins in neuronal apoptosis. Cell Calcium. 23:165–171. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Chen H, Gao W, Yang Y, Guo S, Wang H, Wang W, Zhang S, Zhou Q, Xu H, Yao J, et al: Inhibition of VDAC1 prevents Ca2+-mediated oxidative stress and apoptosis induced by 5-aminolevulinic acid mediated sonodynamic therapy in THP-1 macrophages. Apoptosis. 19:1712–1726. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2019
Volume 20 Issue 1

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
Nie Z, Deng S, Zhang L, Chen S, Lu Q and Peng H: Crocin protects against dexamethasone‑induced osteoblast apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway. Mol Med Rep 20: 401-408, 2019
APA
Nie, Z., Deng, S., Zhang, L., Chen, S., Lu, Q., & Peng, H. (2019). Crocin protects against dexamethasone‑induced osteoblast apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway. Molecular Medicine Reports, 20, 401-408. https://doi.org/10.3892/mmr.2019.10267
MLA
Nie, Z., Deng, S., Zhang, L., Chen, S., Lu, Q., Peng, H."Crocin protects against dexamethasone‑induced osteoblast apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway". Molecular Medicine Reports 20.1 (2019): 401-408.
Chicago
Nie, Z., Deng, S., Zhang, L., Chen, S., Lu, Q., Peng, H."Crocin protects against dexamethasone‑induced osteoblast apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway". Molecular Medicine Reports 20, no. 1 (2019): 401-408. https://doi.org/10.3892/mmr.2019.10267