Hostname: page-component-76fb5796d-2lccl Total loading time: 0 Render date: 2024-04-26T07:05:40.831Z Has data issue: false hasContentIssue false

Genistein induces oestrogen receptor-α gene expression in osteoblasts through the activation of mitogen-activated protein kinases/NF-κB/activator protein-1 and promotes cell mineralisation

Published online by Cambridge University Press:  05 July 2013

Mei-Hsiu Liao
Affiliation:
College of Medicine, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Xing Street, Taipei 110, Taiwan
Yu-Ting Tai
Affiliation:
Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
Yih-Giun Cherng
Affiliation:
Department of Anesthesiology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan
Shing-Hwa Liu
Affiliation:
College of Medicine, Institute of Toxicology, National Taiwan University, Taipei, Taiwan
Ya-An Chang
Affiliation:
Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
Pei-I Lin
Affiliation:
College of Medicine, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Xing Street, Taipei 110, Taiwan
Ruei-Ming Chen*
Affiliation:
College of Medicine, Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Xing Street, Taipei 110, Taiwan Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan Anesthetics and Toxicology Research Center, Taipei Medical University Hospital, Taipei, Taiwan
*
*Corresponding author: R.-M. Chen, fax +886 2 86621119, email rmchen@tmu.edu.tw
Rights & Permissions [Opens in a new window]

Abstract

Oestrogen and oestrogen receptors (ER) play critical roles in the maintenance of bone remodelling. Genistein, structurally similar to 17β-oestradiol, is a phyto-oestrogen that may be beneficial for treating osteoporosis. In the present study, we evaluated the effects of genistein on the regulation of ERα gene expression and osteoblast mineralisation using MC3T3-E1 cells and primary rat calvarial osteoblasts as our experimental models. Exposure of MC3T3-E1 cells and primary rat osteoblasts to genistein at ≤ 10 μm for 24 h did not affect the cell morphology or viability. However, treatment of MC3T3-E1 cells with 10 μm-genistein enhanced the phosphorylation of extracellular signal-regulated kinase 1/2, p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase 1/2 in a time-dependent manner. Sequentially, genistein increased the translocation of NF-κB and c-Jun from the cytoplasm to the nucleus. Consequently, exposure of MC3T3-E1 cells to genistein induced ERα mRNA expression in concentration- and time-dependent manners. In parallel, the amounts of cytosolic and nuclear ERα in MC3T3-E1 cells were increased following genistein administration. Additionally, genistein also increased the levels of ERα mRNA and nuclear ERα protein in rat calvarial osteoblasts. A bioinformatic search revealed that there are several ERα-specific DNA-binding elements in the 5′-promoter regions of the bone morphogenetic protein-6, collagen type I and osteocalcin genes. As a result, genistein could induce the expressions of these osteoblast differentiation-related genes in primary rat osteoblasts. Co-treatment with genistein and traditional differentiation reagents synergistically increased osteoblast mineralisation. Therefore, the present study showed that genistein can induce ERα gene expression via the activation of MAPK/NF-κB/activator protein-1 and accordingly stimulates differentiation-related gene expressions and osteoblast mineralisation.

Type
Full Papers
Copyright
Copyright © The Authors 2013 

Bone structure is maintained by a dynamic balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption( Reference Seeman and Delmas 1 , Reference Chen, Lin and Chou 2 ). In particular, osteoblasts, differentiated from mesenchymal stem cells, play critical roles in bone formation( Reference Takeda and Karsenty 3 ). During osteogenesis, stem and primitive osteoprogenitors that are differentiated from stromal stem cells can replace osteoblasts, which promote bone turnover and fracture healing( Reference Engin and Lee 4 ). Additionally, sequential differentiation of these precursor cells leads to osteoblast maturation and mineralisation( Reference Takeda and Karsenty 3 , Reference Engin and Lee 4 ). A complicated network of various proteins is involved in the regulation of osteogenesis( Reference Vandenput and Ohlsson 5 , Reference Hung, Chen and Liao 6 ). For example, endogenous bone morphogenetic proteins (BMP) are multifunctional growth factors that participate in matrix differentiation and bone formation( Reference Kugimiya, Kawaguchi and Kamekura 7 , Reference Trzeciakiewicz, Habauzit and Mercier 8 ). Osteocalcin (OCN) can act as an early osteoblast marker that controls osteoblast function and bone extracellular matrix mineralisation( Reference van Leeuwen, van Driel and van den Bemd 9 ). In addition, collagen type I (Col I), an extracellular matrix protein, stimulates osteoblast adhesion and differentiation( Reference Mizuno and Kuboki 10 , Reference Mathews, Bhonde and Gupta 11 ). The expressions of all these differentiation-related genes can be regulated by several local and systemic factors, including oestrogen, growth factors and cytokines( Reference Hung, Chen and Liao 6 , Reference Fiorelli and Brandi 12 , Reference Hsu, Liao and Tai 13 ).

Oestrogen acts as the main regulator of skeletal growth and maintenance by adjusting osteoblast differentiation and metabolism( Reference Fiorelli and Brandi 12 ). The levels of serum oestrogen are significantly diminished with ageing and concurrently bring about an imbalance of osteoblastogenesis and osteoclastogenesis( Reference Fiorelli and Brandi 12 , Reference Krum 14 ). As a result, reduced serum oestrogen concentrations may cause systemic diseases such as osteoporosis, leading to increased risks of bone fracture( Reference Manolagas 15 , Reference Eastell and Hannon 16 ). In the clinic, oestrogen replacement therapy has been applied to treat osteoporosis, but many side effects, including a certain cancer incidence, have been reported( Reference Modugno, Laskey and Smith 17 ). As an alternative, phyto-oestrogens are plant-derived compounds that exhibit effects similar to those of mammalian estrogens; they have been widely studied to explore their potential to treat osteoporosis( Reference Usui 18 ). Genistein is a non-steroidal phyto-oestrogen found in a number of plants, including lupin, fava beans and soyabeans( Reference Coward, Barnes and Setchell 19 ). Because of a structural similarity to 17β-oestradiol, genistein has been proposed as a potential therapeutic agent for preventing postmenopausal bone loss( Reference Poulsen and Kruger 20 ). The major mechanism of genistein's action is the control of osteoblast metabolism through oestrogen receptor (ER)-dependent pathways( Reference Wang, Sathyamoorthy and Phang 21 ).

ER are a family of intracellular receptors, including ERα and ERβ( Reference Dahlman-Wright, Cavailles and Fuqua 22 ). ERα has been reported to be the major regulator mediating the effects of oestrogen on bone metabolism( Reference Sims, Clément-Lacroix and Minet 23 ). According to the classical mechanism, oestrogen binds to ERα to form a receptor dimer, and then this dimer is translocated from the cytoplasm to the nucleus where it associates with co-regulatory proteins( Reference Levy, Zhao and Tang 24 ). After binding to its specific oestrogen-response elements, this ER–oestrogen complex can initiate certain gene expressions and control osteoblast differentiation( Reference Gruber, Gruber and Gruber 25 ). Genistein may have a noteworthy impact on the prevention and therapy of postmenopausal bone loss( Reference Poulsen and Kruger 20 ). One of the mechanisms of genistein's action is the stimulation of osteoblast maturation via an ER-dependent induction of cell proliferation-related gene expressions( Reference Pan, Quarles and Song 26 ). Other mechanisms may be the promotion of osteogenesis and repression of adipogenic differentiation of human primary bone marrow stromal cells( Reference Heim, Frank and Kampmann 27 ). In addition, genistein can induce ERα mRNA expression in bone marrow cells( Reference Liao, Xiao and Qin 28 ). ERα plays an important role in the mediation of genistein-involved regulation of osteoblast differentiation and mineralisation. However, the effects of genistein on the regulation of ERα gene expression in osteoblasts are still not very clear. Therefore, the present study attempted to evaluate the mechanisms of genistein-induced ERα gene expression and the effects of genistein on the regulation of osteoblast differentiation and mineralisation using MC3T3-E1 cells and primary rat calvarial osteoblasts as the experimental models.

Materials and methods

Cell culture and drug treatment

Mouse MC3T3-E1 cells were purchased from American Type Culture Collection. Primary rat osteoblasts were prepared from neonatal rat calvaria following a sequential enzymatic digestion method described previously( Reference Hung, Chen and Liao 6 ). All procedures were performed according to the National Institutes of Health Guidelines for the Use of Laboratory Animals and approved by the Institutional Animal Care and Use Committee of Taipei Medical University (Taipei, Taiwan). Briefly, after anaesthesia, the neonatal rats were killed, and the calvarias were collected, washed and cut into pieces. The calvarial pieces were incubated in a digestion solution (0·25 % trypsin, 2 g/l collagenases and 0·1 % EDTA) at 37°C. After centrifugation, the primary osteoblasts were washed and cultured. MC3T3-E1 cells and primary rat osteoblasts were seeded in Dulbecco's modified Eagle's medium (Gibco-BRL) supplemented with 10 % fetal bovine serum, l-glutamine, penicillin (65 mg/l) and streptomycin (100 μg/ml) in 75 cm2 flasks at 37°C in a humidified atmosphere of 5 % CO2. Genistein purchased from Sigma was freshly dissolved in dimethyl sulphoxide. These two types of bone cells were exposed to various concentrations of genistein for different time intervals. Control cells were exposed to only dimethyl sulphoxide.

Assay of cell viability

The cytotoxicity of genistein towards osteoblasts was evaluated by analyses of cell viability and morphologies( Reference Hsu, Liao and Tai 13 ). Cell viability was determined by a colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Briefly, 104 cells were seeded in ninety-six-well tissue culture plates overnight. After genistein treatment, the osteoblasts were cultured with a new medium containing 0·5 mg/ml of MTT for a further 3 h. Blue formazan products in the osteoblasts were dissolved in dimethyl sulphoxide and spectrophotometrically measured at a wavelength of 550 nm. Cell morphologies were observed and photographed using an inverted phase-contrast microscope (Nikon).

Immunodetection of phosphorylated extracellular signal-regulated kinase 1/2, p38 and c-Jun N-terminal kinase and β-actin proteins

After genistein treatment, cell lysates were prepared in an ice-cold radioimmunoprecipitation assay buffer (25 mm-Tris–HCl (pH 7·2), 0·1 % SDS, 1 % Triton X-100, 1 % sodium deoxycholate, 0·15 m-NaCl and 1 mm-EDTA) as described previously( Reference Chang, Chen and Tai 29 ). Protein concentrations were quantified using a bicinchoninic acid protein assay kit (Pierce). Proteins (100 μg per well) were subjected to SDS–PAGE and transferred onto nitrocellulose membranes. After blocking, phosphorylated extracellular signal-regulated kinase (ERK) 1/2, p38 and c-Jun N-terminal kinase (JNK; Cell Signaling Technology) were immunodetected. β-Actin was detected using a mouse monoclonal antibody (Sigma) as the internal control. The bands of these proteins were quantified using a digital imaging system (UVtec).

Extraction of nuclear proteins and immunodetection

Nuclear components were extracted, and immunodetection was carried out following the method of Wu et al. ( Reference Wu, Chen and Ueng 30 ). After genistein treatment, the nuclear extracts of osteoblasts were prepared. Protein concentrations were quantified using a bicinchoninic acid protein assay kit (Pierce). Nuclear proteins (50 μg/well) were subjected to SDS–PAGE and transferred onto nitrocellulose membranes. After blocking, nuclear ERα, NF-κB, c-Jun and c-Fos were immunodetected using specific antibodies (Santa Cruz Biotechnology). Proliferating cell nuclear antigen was immunodetected (Santa Cruz Biotechnology) as the internal standard. The intensities of the immunoreactive bands were determined using a digital imaging system (UVtec).

RT-PCR and real-time PCR assays

For the RT-PCR analyses of ERα, Col I, BMP-6, OCN and β-actin mRNA, mRNA from MC3T3-E1 cells and primary rat osteoblasts were prepared according to a previously described method( Reference Lin, Chen and Hong 31 ). Oligonucleotide primers were designed and synthesised by Clontech Laboratories. The oligonucleotide sequences of the respective upstream and downstream primers for analyses of these mRNA were as follows: 5′-TCCTTCTAGACCCTTCAGTGAAGCC-3′ and 5′-ACATGTCAAAGATCTCCACCATGCC-3′ for ERα; 5′-AAAGCCAAGAGAAACGGTGGGCAT-3′ and 5′-GCCAATCATGTGCACCAGTTCCTT-3′ for ERβ; 5′-CTTGTCCTCATGGCTGTGAAAC-3′ and 5′-TATTGCTGGTGCTCCTGGCTTC-3′ for Col I; 5′-AGGATGGGGTGTCAGAGGGAGA-3′ and 5′-GTTGTGCTGCGGTGTCACCA-3′ for BMP-6; 5′-ATGAGGACCCTCTCTCTGCTC-3′ and 5′-GTGGTGCCATAATGCGCTTG-3′ for OCN and 5′-GTGGGCCGCTCTAGGCACCAA-3′ and 5′-CTCTTTGATGTCACGCACGATTTC-3′ for rat β-actin. The PCR products were loaded onto a 1·8 % agarose gel containing 0·1 μg/ml ethidium bromide and electrophoretically separated. DNA bands were visualised and photographed under UV-light exposure. The intensities of the DNA bands in the agarose gel were quantified with the aid of a digital imaging system (Uvtec). Real-time PCR analyses were carried out using the iQSYBR Green Supermix (Bio-Rad) and the MyiQ Single-Color Real-Time PCR Detection System (Bio-Rad).

Assays of osteoblast mineralisation

Osteoblast maturation was determined by evaluating cell mineralisation using the Alizarin red S dye-staining protocol( Reference Hess, Angel and Schorpp-Kistner 32 ). Primary rat osteoblasts were treated with genistein, a differentiation reagent (DR; 10 nm-dexamethasone, 100 μg/ml ascorbic acid and 10 mm-β-glycerophosphate) and a combination of genistein and the DR for 21 d. After genistein treatment, rat osteoblasts were washed with ice-cold PBS (0·14 m-NaCl, 2·6 mm-KCl, 8 mm-Na2HPO4 and 1·5 mm-KH2PO4) and then fixed in ice-cold 10 % formalin for 20 min. The fixed osteoblasts were thoroughly rinsed and then incubated in 1 % alcian blue (pH 2·5; Fisher Scientific) for 12 h. The sections were then incubated in Alizarin red S (Fisher Scientific) for 8 min, briefly dehydrated in xylene and covered with a coverslip in Permount (Fisher Scientific). Mineralised nodules were visualised and counted under an inverted microscope. Each experiment was performed in duplicate wells and repeated three times.

Statistical analysis

Statistical differences between the control and drug-treated groups were considered significant when the P value of Duncan's multiple-range test was < 0·05. Statistical analysis between the drug-treated groups was carried out using a two-way ANOVA.

Results

The cytotoxicity of genistein towards MC3T3-E1 cells was determined (Fig. 1). Exposure of MC3T3-E1 cells to 0·01, 0·1, 1, 10 and 100 μm-genistein for 24 h did not influence cell viability (Fig. 1(a)). Additionally, after treatment of MC3T3-E1 cells with 1, 5 and 10 μm-genistein for 24 h, the cell morphologies did not change (Fig. 1(b)). Exposure of MC3T3-E1 cells to 10 μm-genistein for 0·5, 1, 3, 6 and 24 h did not affect the cell morphologies (Fig. 1(c)). After exposure to 0·01, 0·1, 1, 10 and 100 μm-genistein for 24 h, the viability of primary rat calvarial osteoblasts was not influenced (data not shown).

Fig. 1 Cytotoxicity of genistein towards MC3T3-E1 cells. (a) MC3T3-E1 cells were exposed to 0·01, 0·1, 1, 10 and 100 μm-genistein for 24 h, and cell viability was determined using a colorimetric method. The morphologies of MC3T3-E1 cells were observed after exposure to (b) 1, 5 and 10 μm-genistein for 24 h or to (c) 10 μm-genistein for 0·5, 1, 3, 6 and 24 h. Values are means (n 6), with their standard errors represented by vertical bars. OD550 nm, optical density at 550 nm.

The effects of genistein on the phosphorylation of mitogen-activated protein kinase (MAPK) and the translocation of NF-κB and activator protein-1 (AP-1) were immunodetected (Fig. 2). In the untreated MC3T3-E1 cells, low levels of phosphorylated ERK1/2, p38 MAPK and JNK1/2 were detected (Fig. 2(a), top three panels, lane 1). After exposure to 10 μm-genistein for 0·5, 1, 3 and 6 h, the levels of phosphorylated ERK1/2 were increased in a time-dependent manner (Fig. 2(a), top panel, lanes 2–5). The phosphorylation of p38 MAPK in MC3T3-E1 cells was also time dependently enhanced following exposure to 10 μm-genistein for 0·5, 1, 3 and 6 h (lanes 2–5). When MC3T3-E1 cells were treated with 10 μm-genistein for 6 h, the amounts of phosphorylated JNK1/2 increased (lane 5). β-Actin was immunodetected as the internal control (Fig. 2(a)). Exposure of MC3T3-E1 cells to 10 μm-genistein for 0·5 h increased the levels of nuclear NF-κB (Fig. 2(b), top panel, lane 2). After exposure for 1 and 3 h, genistein enhanced the increases in the amounts of nuclear NF-κB (lanes 3 and 4). The levels of c-Jun and c-Fos in the nuclei were immunodetected (Fig. 2(b)). Treatment of MC3T3-E1 cells with genistein for 0·5, 1 and 3 h led to significant increases in the levels of nuclear c-Jun and c-Fos (lanes 2 and 3). Nuclear proliferating cell nuclear antigen was analysed as the internal control (Fig. 2(b)).

Fig. 2 Effects of genistein on the phosphorylation (p) of extracellular signal-regulated kinase (ERK) 1/2, p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) 1/2 and the translocation of NF-κB, c-Jun and c-Fos. MC3T3-E1 cells were exposed to 10 μm-genistein for 0·5, 1, 3 and 6 h. (a) p-ERK1/2, p-p38 MAPK and p-JNK1/2 were immunodetected. The amounts of β-actin were analysed as internal controls. (b) The levels of nuclear (n) NF-κB, nc-Jun and nc-Fos were determined. The amounts of nuclear proliferating cell nuclear antigen (PCNA) were analysed as internal controls (bottom).

Analyses of RNA were performed to determine the effects of genistein on ERα and ERβ mRNA expressions (Fig. 3). Exposure of MC3T3-E1 cells to 1 μm-genistein for 6 h did not affect ERα mRNA expression (Fig. 3(a), top panel, lane 2). When the concentration reached 5 μm, genistein slightly induced ERα mRNA synthesis (lane 3). In contrast, 10 μm-genistein induced a strong ERα mRNA expression (lane 4). Treatment of MC3T3-E1 cells with 10 μm-genistein for 1 and 3 h did not influence ERα mRNA expression (Fig. 3(b), top panel, lanes 2 and 3). However, exposure for 6 and 24 h significantly induced ERα mRNA expression (lanes 4 and 5). The amounts of β-actin mRNA were analysed as internal controls (Fig. 3(a) and (b)). Real-time PCR analyses showed that genistein significantly induced ERα and ERβ mRNA expressions by 230 and 190 %, respectively (Fig. 3(c)).

Fig. 3 Effects of genistein on the regulation of the expressions of oestrogen receptor (ER) α and ERβ mRNA. MC3T3-E1 cells were exposed to (a) 1, 5 and 10 μm-genistein for 6 h or to (b) 10 μm-genistein for 1, 3, 6 and 24 h. (a, b) Analysis of ERα mRNA was conducted using a RT-PCR. β-Actin mRNA was analysed as an internal control. (c) Real-time PCR analyses were conducted to confirm the effects of genistein on the regulation of ERα and ERβ mRNA expressions. Values are means (n 4), with their standard errors represented by vertical bars. * Mean value was significantly different from that of the control (P< 0·05). M, marker of 100 bp DNA ladder.

Immunoblotting analyses were performed to determine the effects of genistein on ERα protein synthesis (Fig. 4). Exposure of MC3T3-E1 cells to 1 μm-genistein for 24 h did not affect the levels of ERα (Fig. 4(a), top panel, lane 2). In contrast, when the concentrations reached 5 and 10 μm, genistein increased ERα synthesis (lanes 3 and 4). Exposure of MC3T3-E1 cells to 10 μm-genistein for 0·5, 1, 3, 6 and 24 h time dependently enhanced the levels of ERα (Fig. 4(b)). The amounts of β-actin were immunodetected as internal controls (Fig. 4(a) and (b)).

Fig. 4 Effects of genistein on the synthesis of oestrogen receptor (ER) α protein. MC3T3-E1 cells were exposed to (a) 1, 5 and 10 μm-genistein for 24 h or to (b) 10 μm-genistein for 0·5, 1, 3, 6 and 24 h. (a, b) ERα was immunodetected. The amounts of β-actin were analysed as internal controls.

Nuclear proteins were isolated and analysed to determine the effects of genistein on the translocation of ERα from the cytoplasm to the nucleus (Fig. 5). Exposure of MC3T3-E1 cells to 1 μm-genistein for 24 h increased the levels of nuclear ERα (Fig. 5(a), top panel, lane 2). After treatment with 5 and 10 μm-genistein, the amounts of ERα in the nuclei were significantly increased (lanes 3 and 4). Exposure of MC3T3-E1 cells to 10 μm-genistein for 0·5, 1, 3, 6, and 24 h enhanced the nuclear ERα levels in a time-dependent manner (Fig. 5(b), top panel, lanes 2–6). The amounts of β-actin were immunodetected as internal controls (Fig. 5(a) and (b)).

Fig. 5 Effects of genistein on the translocation of oestrogen receptor (ER) α from the cytoplasm to the nucleus (n). MC3T3-E1 cells were exposed to (a) 1, 5 and 10 μm-genistein for 24 h or to (b) 10 μm-genistein for 0·5, 1, 3, 6 and 24 h. The levels of nERα were immunodetected. The amounts of nuclear proliferating cell nuclear antigen (PCNA) were analysed as internal controls.

Rat calvarial osteoblasts were prepared to evaluate the effects of genistein on the expressions of ERα and cell differentiation-related genes (Fig. 6). Exposure of rat osteoblasts to 10 μm-genistein for 6 h induced ERα mRNA expression (Fig. 6(a), top panel, lane 2). The levels of nuclear ERα in rat osteoblasts were significantly increased following treatment with 10 μm-genistein for 24 h (Fig. 6(b), top panel, lane 2). The amounts of β-actin mRNA and protein were analysed as internal controls (Fig. 6(a) and (b)). A bioinformatic approach was performed, and the results indicated that 9, 10 and 10 ERα-specific DNA-binding elements, respectively, existed in the 5′-promoter regions of the BMP-6, Col I and OCN genes (Fig. 6(c)). After exposure to 10 μm-genistein for 6 h, the expressions of BMP-6, Col I and OCN mRNA were significantly induced (Fig. 6(d), top three panels, lane 2). β-Actin mRNA was analysed as the internal control.

Fig. 6 Effects of genistein on the regulation of the expressions of the oestrogen receptor (ER) α, bone morphogenetic protein (BMP) 6, collagen type I (Col I) and osteocalcin (OCN) genes in primary rat osteoblasts. Rat osteoblasts prepared from rat calvaria were exposed to 10 μm-genistein for (a, d) 6 h and (b) 24 h. (a, d) The levels of ERα, BMP-6, Col I and OCN mRNA were determined using a RT-PCR. (a, d) β-Actin mRNA was analysed as an internal control. (b) The amounts of nuclear ERα (nERα) were immunodetected. Nuclear proliferating cell nuclear antigen (PCNA) was analysed as an internal control. A bioinformatic approach was used to search the ERα-specific DNA-binding elements in the 5′-promoter regions of the BMP-6, Col I and OCN genes. M, marker of 100 bp DNA ladder.

After exposure to a combined mixture of dexamethasone, ascorbic acid, and β-glycerophosphate or genistein for 21 d, osteoblast mineralisation was determined (Fig. 7). Treatment of primary rat calvarial osteoblasts with genistein alone did not affect cell mineralisation (Fig. 7(a)). However, when the osteoblasts were exposed to a mixture of dexamethasone, ascorbic acid and β-glycerophosphate for 21 d, mineralised nodules were obviously increased (Fig. 7(a)). In contrast, co-treatment with the DR and genistein synergistically increased osteoblast mineralisation (Fig. 7(a)). These mineralised nodules were quantified and analysed (Fig. 7(b)). In the control and genistein-treated rat osteoblasts, mineralised nodules were not observed. After exposure to the DR, the nodules were significantly produced. Compared with the DR-treated group, co-treatment with genistein and the DR resulted in a significant twofold increase in osteoblast mineralisation (Fig. 7(b)).

Fig. 7 Effects of genistein on the mineralisation of primary rat osteoblasts. Rat osteoblasts prepared from rat calvaria were exposed to 10 μm-genistein (G), a differentiation reagent (DR, including 10 nm-dexamethasone (Dex), 100 μg ascorbic acid (AA)/ml and 10 mm-β-glycerophosphate (GP)), and a combination of genistein and the DR for 21 d. The drugs were renewed every 2 d. (a) Osteoblast mineralisation was determined using the Alizarin red S dye-staining protocol. (b) The amounts of mineralised nodules were quantified and analysed.

Discussion

Genistein can induce ERα gene expression. The present study showed that after exposure to genistein the levels of ERα in MC3T3-E1 cells were increased in concentration- and time-dependent manners. At the same time, ERα mRNA expression was significantly increased by genistein. Thus, genistein-induced ERα gene expression occurred at least by a pre-translational mechanism. The genistein-induced ERα mRNA expression was also confirmed in primary osteoblasts isolated from rat calvaria. In addition, we demonstrated that genistein under the present administered conditions did not cause cytotoxicity in osteoblasts. Liao et al. ( Reference Liao, Xiao and Qin 28 ) reported that genistein can stimulate ERα mRNA expression in bone marrow cells. In the present study, we further showed that genistein can induce ERα mRNA and protein expressions in MC3T3-E1 cells and primary rat calvarial osteoblasts. We also demonstrated that ERβ mRNA expression can be induced by genistein. ERα and ERβ play vital roles in the mediation of oestrogen-involved regulation of skeletal growth and maintenance( Reference Sims, Clément-Lacroix and Minet 23 ). However, the levels of human serum oestrogen decrease with ageing( Reference Fiorelli and Brandi 12 , Reference Krum 14 ). Genistein, an isoflavone-type phyto-oestrogen, has been reported to have the potential to prevent and treat osteoporosis( Reference Usui 18 , Reference Coward, Barnes and Setchell 19 ). Osteoblasts contribute to bone formation( Reference Seeman and Delmas 1 , Reference Chen, Lin and Chou 2 ). Therefore, the present study further showed that genistein can induce ERα gene expression and possibly participate in the regulation of osteoblast activities and bone metabolism.

The translocation of NF-κB and AP-1 is involved in genistein-induced ERα gene expression. The amounts of nuclear NF-κB and c-Jun in the osteoblasts were time dependently up-regulated following genistein administration. c-Jun and c-Fos can bind to each other to form the heterodimeric AP-1, a transcription factor( Reference Hess, Angel and Schorpp-Kistner 32 ). The results of our bioinformatic search revealed that both NF-κB- and AP-1-specific DNA-binding elements exist in the 5′-promoter region of the ERα gene. Thus, genistein-induced ERα gene expression was due to the improved translocation of NF-κB and AP-1 from the cytoplasm to the nucleus. The increased translocation of these two transcription factors simultaneously increases their transactivation activities and induces ERα gene expression. NF-κB and AP-1 are two typical transcription factors that participate in the regulation of osteoblast differentiation and mineralisation( Reference Kim, Lee and Kim 33 , Reference Tan, Huang and Chang 34 ). Targeting NF-κB- and AP-1-transduced signals has been reported to be a promising strategy for treating bone diseases( Reference Wu, Li and Yang 35 ). Genistein-induced translocation of NF-κB and AP-1 can concurrently explain the induction of the expression of the ERα gene and its effects on the promotion of osteoblast activities and bone metabolism.

Genistein can trigger the phosphorylation of MAPK and enhance the translocation of NF-κB and AP-1. The present results revealed that exposure of osteoblasts to genistein caused time-dependent increases in the phosphorylation of ERK1/2, JNK1/2 and p38 MAPK. MAPK are serine/threonine-specific protein kinases containing three major enzymes, namely ERK1/2, JNK1/2 and p38 MAPK( Reference Manning, Whyte and Martinez 36 ). After phosphorylation, ERK1/2 and p38 MAPK can activate NF-κB and trigger the translocation of this transcription factor into nuclei, which subsequently induces the expressions of certain genes( Reference Karin 37 ). In contrast, the activated JNK1/2 can phosphorylate c-Jun and induce the transactivation activity of AP-1( Reference Shaulian 38 ). Accordingly, one of the major reasons explaining the translocation of NF-κB and AP-1 stimulated by genistein is the phosphorylation of ERK1/2, p38 MAPK and JNK1/2 by this phyto-oestrogen. In oestrogen-involved signalling, the activated ERK1/2 promotes ERα-mediated intracellular signal-transducing events( Reference Driggers and Segars 39 ). In addition, the phosphorylation of MAPK contributes to the regulation of bone remodelling( Reference Wu, Li and Yang 35 ). Therefore, genistein-induced phosphorylation of ERK1/2, JNK1/2 and p38 MAPK and subsequent activation of the transcription factors NF-κB and AP-1 could have multiple functions in the regulation of ERα gene expression and protein activation and improvement of osteoblast maturation.

Genistein stimulates the expressions of the BMP-6, Col I and OCN genes through the activation of ERα in primary rat osteoblasts. MC3T3-E1 is an osteoblast-like cell line. To confirm the effects of genistein on primary cells, osteoblasts were isolated from neonatal rat calvaria. After exposure to genistein, ERα mRNA and protein expressions were significantly induced. Thus, genistein can also induce ERα gene expression in primary osteoblasts as that observed in osteoblast-like MC3T3-E1 cells. ERα can act as a transcription factor in the regulation of osteoblast differentiation-related genes, including BMP-6, Col I and OCN ( Reference Dahlman-Wright, Cavailles and Fuqua 22 ). The present study showed that in parallel to the increase in the translocation of ERα into the nuclei, the expressions of BMP-6, Col I and OCN mRNA in primary rat osteoblasts were simultaneously induced. We also showed that several ERα-specific DNA-binding elements are present in the 5′-promoter regions of the BMP-6, Col I and OCN genes. Therefore, in primary osteoblasts, genistein can induce ERα mRNA expression and protein activation and stimulate the expressions of the BMP-6, Col I and OCN genes.

Genistein has synergistic effects on the stimulation of mineralisation of primary rat osteoblasts. The present study demonstrated that although genistein alone cannot induce the mineralisation of primary rat osteoblasts, co-treatment with a traditional DR synergistically promotes osteoblast maturation. Osteoblast differentiation and maturation are regulated by a complicated network of various proteins, including BMP, Col I and OCN( Reference Vandenput and Ohlsson 5 ). In osteogenesis, BMP-6 and Col I participate in bone extracellular matrix differentiation and mineralisation( Reference Trzeciakiewicz, Habauzit and Mercier 8 , Reference van Leeuwen, van Driel and van den Bemd 9 ). Col I contributes to osteoblast adhesion and differentiation( Reference Mathews, Bhonde and Gupta 11 ). Increases in the syntheses of these proteins will trigger osteoblast differentiation and mineralisation( Reference Vandenput and Ohlsson 5 , Reference Kugimiya, Kawaguchi and Kamekura 7 , Reference Mizuno and Kuboki 10 ). In the present study, genistein was shown to induce the expressions of the BMP-6, Col I and OCN genes. Therefore, genistein can promote osteoblast differentiation and bone metabolism by inducing the expressions of these cell differentiation-related genes.

In summary, the present study showed that exposure of MC3T3-E1 cells and primary rat osteoblasts to genistein did not cause cytotoxicity. In contrast, genistein induced ERα mRNA and protein expressions in MC3T3-E1 cells in concentration- and time-dependent manners. As regards the mechanisms, treatment with genistein time dependently increased the phosphorylation of ERK1/2, p38 MAPK and JNK1/2. Sequentially, the levels of nuclear NF-κB and c-Jun were significantly enhanced after genistein administration. In primary neonatal rat calvarial osteoblasts, the effects of genistein on ERα mRNA and protein expressions were also confirmed. Genistein can trigger the translocation of ERα from the cytoplasm to the nucleus. The results of a bioinformatic approach revealed that several ERα-specific DNA-binding elements exist in the 5′-promoter regions of the osteoblast differentiation-related BMP-6, Col I and OCN genes. In parallel, genistein induced the expressions of BMP-6, Col I and OCN mRNA in primary rat osteoblasts. Consequently, exposure to genistein synergistically improved the traditional ER-induced osteoblast mineralisation. In summary, the present study showed that genistein can induce ERα gene expression via the activation of MAPK/NF-κB/AP-1 and stimulate osteoblast differentiation and maturation through the ERα-dependent induction of the expressions of the BMP-6, Col I and OCN genes. The molecular mechanisms of genistein-involved regulation of the expressions of osteoblast differentiation-related genes, including BMP-6, Col I and OCN, are being studied in our laboratory. The effects of genistein on bone healing using an animal model of bone defects will be our next research focus.

Acknowledgements

The present study was supported by Wan-Fang Hospital (100-wf-eva-10) and the National Science Council (NSC101-2314-B-038-003-MY3), Taipei, Taiwan. All authors participated in the design of the present study. M.-H. L. and P.-I. L. carried out the cell culture and drug treatment. M.-H. L., Y.-T. T. and Y.-A. C. conducted the assays of cell viability and immunodetection. M.-H. L., Y.-T. T. and Y.-G. C. contributed to the RT-PCR analyses and osteoblast maturation. S.-H. L. and R.-M. C. participated in the data analyses. M.-H. L. and R.-M. C. wrote the initial manuscript draft. R.-M. C. assembled the final version. All authors read and approved the final manuscript. None of the authors has any conflict of interest to declare.

References

1 Seeman, E & Delmas, PD (2006) Bone quality – the material and structural basis of bone strength and fragility. N Engl J Med 354, 22502261.CrossRefGoogle ScholarPubMed
2 Chen, RM, Lin, YL & Chou, CW (2010) GATA-3 transduces survival signals in osteoblasts through upregulation of bcl-x L gene expression. J Bone Miner Res 25, 21932204.CrossRefGoogle ScholarPubMed
3 Takeda, S & Karsenty, G (2001) Central control of bone formation. J Bone Miner Metab 19, 195198.CrossRefGoogle ScholarPubMed
4 Engin, F & Lee, B (2010) NOTCHing the bone: insights into multi-functionality. Bone 46, 274280.Google Scholar
5 Vandenput, L & Ohlsson, C (2009) Estrogens as regulators of bone health in men. Nat Rev Endocrinol 5, 437443.CrossRefGoogle ScholarPubMed
6 Hung, TY, Chen, TL, Liao, MH, et al. (2010) Drynaria fortunei J. Sm. promotes osteoblast maturation by inducing differentiation-related gene expression and protecting against oxidative stress-induced apoptotic insults. J Ethnopharmacol 131, 7077.Google Scholar
7 Kugimiya, F, Kawaguchi, H, Kamekura, S, et al. (2005) Involvement of endogenous bone morphogenetic protein (BMP) 2 and BMP6 in bone formation. J Biol Chem 280, 3570435712.Google Scholar
8 Trzeciakiewicz, A, Habauzit, V, Mercier, S, et al. (2010) Hesperetin stimulates differentiation of primary rat osteoblasts involving the BMP signalling pathway. J Nutr Biochem 21, 424431.Google Scholar
9 van Leeuwen, JP, van Driel, M, van den Bemd, GJ, et al. (2001) Vitamin D control of osteoblast function and bone extracellular matrix mineralization. Crit Rev Eukaryot Gene Expr 11, 199226.Google Scholar
10 Mizuno, M & Kuboki, Y (2001) Osteoblast-related gene expression of bone marrow cells during the osteoblastic differentiation induced by type I collagen. J Biochem 129, 133138.Google Scholar
11 Mathews, S, Bhonde, R, Gupta, PK, et al. (2012) Extracellular matrix protein mediated regulation of the osteoblast differentiation of bone marrow derived human mesenchymal stem cells. Differentiation 84, 185192.Google Scholar
12 Fiorelli, G & Brandi, ML (1999) Skeletal effects of estrogens. J Endocrinol Invest 22, 589593.Google Scholar
13 Hsu, CK, Liao, MH, Tai, YT, et al. (2011) Nanoparticles prepared from the water extract of Gusuibu (Drynaria fortunei J. Sm.) protects osteoblasts against insults and promotes cell maturation. Int J Nanomedicine 6, 14051413.Google Scholar
14 Krum, SA (2011) Direct transcriptional targets of sex steroid hormones in bone. J Cell Biochem 112, 401408.Google Scholar
15 Manolagas, SC (2000) Birth and death of bone cells: basic regulatory mechanisms and implications for pathogenesis and treatment of osteoporosis. Endocr Rev 21, 115137.Google Scholar
16 Eastell, R & Hannon, RA (2008) Biomarkers of bone health and osteoporosis risk. Proc Nutr Soc 67, 157162.Google Scholar
17 Modugno, F, Laskey, R, Smith, AL, et al. (2012) Hormone response in ovarian cancer: time to reconsider as a clinical target? Endocr Relat Cancer 19, 255279.Google Scholar
18 Usui, T (2006) Pharmaceutical prospects of phytoestrogens. Endocr J 53, 720.Google Scholar
19 Coward, L, Barnes, NC, Setchell, KDR, et al. (1993) Genistein, daidzein, and their β-glycoside conjugates: antitumor isoflavones in soybean foods from American and Asian diets. J Agric Food Chem 41, 19611967.Google Scholar
20 Poulsen, RC & Kruger, MC (2008) Soy phytoestrogen: impact on postmenopausal bone loss and mechanisms of action. Nutr Rev 66, 359374.Google Scholar
21 Wang, TT, Sathyamoorthy, N & Phang, JM (1996) Molecular effects of genistein on estrogen receptor mediated pathways. Carcinogenesis 17, 271275.CrossRefGoogle ScholarPubMed
22 Dahlman-Wright, K, Cavailles, V, Fuqua, SA, et al. (2006) International Union of Pharmacology. LXIV. Estrogen receptors. Pharmacol Rev 58, 773781.Google Scholar
23 Sims, NA, Clément-Lacroix, P, Minet, D, et al. (2003) A functional androgen receptor is not sufficient to allow estradiol to protect bone after gonadectomy in estradiol receptor-deficient mice. J Clin Invest 111, 13191327.Google Scholar
24 Levy, N, Zhao, X, Tang, H, et al. (2007) Multiple transcription factor elements collaborate with estrogen receptor alpha to activate an inducible estrogen response element in the NKG2E gene. Endocrinology 148, 34493458.Google Scholar
25 Gruber, CJ, Gruber, DM, Gruber, IM, et al. (2004) Anatomy of the estrogen response element. Trends Endocrinol Metab 15, 7378.Google Scholar
26 Pan, W, Quarles, LD, Song, LH, et al. (2005) Genistein stimulates the osteoblastic differentiation via NO/cGMP in bone marrow culture. J Cell Biochem 94, 307316.CrossRefGoogle ScholarPubMed
27 Heim, M, Frank, O, Kampmann, G, et al. (2004) The phytoestrogen genistein enhances osteogenesis and represses adipogenic differentiation of human primary bone marrow stromal cells. Endocrinology 145, 848859.Google Scholar
28 Liao, QC, Xiao, ZS, Qin, YF, et al. (2007) Genistein stimulates osteoblastic differentiation via p38 MAPK-Cbfa1 pathway in bone marrow culture. Acta Pharmacol Sin 28, 15971602.Google Scholar
29 Chang, HC, Chen, TG, Tai, YT, et al. (2011) Resveratrol attenuates oxidized LDL-evoked Lox-1 signaling and consequently protects against apoptotic insults to cerebrovascular endothelial cells. J Cereb Blood Flow Metab 31, 842854.Google Scholar
30 Wu, GJ, Chen, TL, Ueng, YF, et al. (2008) Ketamine inhibits tumor necrosis factor-α and interleukin-6 gene expressions in lipopolysaccharide-stimulated macrophages through suppression of toll-like receptor 4-mediated c-Jun N-terminal kinase phosphorylation and activator protein-1 activation. Toxicol Appl Pharmacol 228, 105113.Google Scholar
31 Lin, JW, Chen, JT, Hong, CY, et al. (2012) Honokiol traverses the blood–brain barrier and induces apoptosis of neuroblastoma cells via an intrinsic Bax-mitochondrion-cytochrome c-caspase protease pathway. Neuro Oncol 14, 302314.CrossRefGoogle ScholarPubMed
32 Hess, J, Angel, P & Schorpp-Kistner, M (2004) AP-1 subunits: quarrel and harmony among siblings. J Cell Sci 117, 59655973.Google Scholar
33 Kim, JM, Lee, SU, Kim, YS, et al. (2008) Baicalein stimulates osteoblast differentiation via coordinating activation of MAP kinases and transcription factors. J Cell Biochem 104, 19061917.CrossRefGoogle ScholarPubMed
34 Tan, TW, Huang, YL, Chang, JT, et al. (2012) CCN3 increases BMP-4 expression and bone mineralization in osteoblasts. J Cell Physiol 227, 25312541.Google Scholar
35 Wu, X, Li, Z, Yang, Z, et al. (2012) Caffeic acid 3,4-dihydroxy-phenethyl ester suppresses receptor activator of NF-kappa B ligand-induced osteoclastogenesis and prevents ovariectomy-induced bone loss through inhibition of mitogen-activated protein kinase/activator protein 1 and Ca2+-nuclear factor of activated T-cells cytoplasmic 1 signaling pathways. J Bone Miner Res 27, 12981308.Google Scholar
36 Manning, G, Whyte, DB, Martinez, R, et al. (2002) The protein kinase complement of the human genome. Science 298, 19121934.Google Scholar
37 Karin, M (2005) Inflammation-activated protein kinases as targets for drug development. Proc Am Thorac Soc 2, 386390.Google Scholar
38 Shaulian, E (2010) AP-1 – the Jun proteins: oncogenes or tumor suppressors in disguise? Cell Signal 22, 894899.Google Scholar
39 Driggers, PH & Segars, JH (2002) Estrogen action and cytoplasmic signaling pathways. Part II: the role of growth factors and phosphorylation in estrogen signaling. Trends Endocrinol Metab 13, 422427.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1 Cytotoxicity of genistein towards MC3T3-E1 cells. (a) MC3T3-E1 cells were exposed to 0·01, 0·1, 1, 10 and 100 μm-genistein for 24 h, and cell viability was determined using a colorimetric method. The morphologies of MC3T3-E1 cells were observed after exposure to (b) 1, 5 and 10 μm-genistein for 24 h or to (c) 10 μm-genistein for 0·5, 1, 3, 6 and 24 h. Values are means (n 6), with their standard errors represented by vertical bars. OD550nm, optical density at 550 nm.

Figure 1

Fig. 2 Effects of genistein on the phosphorylation (p) of extracellular signal-regulated kinase (ERK) 1/2, p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) 1/2 and the translocation of NF-κB, c-Jun and c-Fos. MC3T3-E1 cells were exposed to 10 μm-genistein for 0·5, 1, 3 and 6 h. (a) p-ERK1/2, p-p38 MAPK and p-JNK1/2 were immunodetected. The amounts of β-actin were analysed as internal controls. (b) The levels of nuclear (n) NF-κB, nc-Jun and nc-Fos were determined. The amounts of nuclear proliferating cell nuclear antigen (PCNA) were analysed as internal controls (bottom).

Figure 2

Fig. 3 Effects of genistein on the regulation of the expressions of oestrogen receptor (ER) α and ERβ mRNA. MC3T3-E1 cells were exposed to (a) 1, 5 and 10 μm-genistein for 6 h or to (b) 10 μm-genistein for 1, 3, 6 and 24 h. (a, b) Analysis of ERα mRNA was conducted using a RT-PCR. β-Actin mRNA was analysed as an internal control. (c) Real-time PCR analyses were conducted to confirm the effects of genistein on the regulation of ERα and ERβ mRNA expressions. Values are means (n 4), with their standard errors represented by vertical bars. * Mean value was significantly different from that of the control (P< 0·05). M, marker of 100 bp DNA ladder.

Figure 3

Fig. 4 Effects of genistein on the synthesis of oestrogen receptor (ER) α protein. MC3T3-E1 cells were exposed to (a) 1, 5 and 10 μm-genistein for 24 h or to (b) 10 μm-genistein for 0·5, 1, 3, 6 and 24 h. (a, b) ERα was immunodetected. The amounts of β-actin were analysed as internal controls.

Figure 4

Fig. 5 Effects of genistein on the translocation of oestrogen receptor (ER) α from the cytoplasm to the nucleus (n). MC3T3-E1 cells were exposed to (a) 1, 5 and 10 μm-genistein for 24 h or to (b) 10 μm-genistein for 0·5, 1, 3, 6 and 24 h. The levels of nERα were immunodetected. The amounts of nuclear proliferating cell nuclear antigen (PCNA) were analysed as internal controls.

Figure 5

Fig. 6 Effects of genistein on the regulation of the expressions of the oestrogen receptor (ER) α, bone morphogenetic protein (BMP) 6, collagen type I (Col I) and osteocalcin (OCN) genes in primary rat osteoblasts. Rat osteoblasts prepared from rat calvaria were exposed to 10 μm-genistein for (a, d) 6 h and (b) 24 h. (a, d) The levels of ERα, BMP-6, Col I and OCN mRNA were determined using a RT-PCR. (a, d) β-Actin mRNA was analysed as an internal control. (b) The amounts of nuclear ERα (nERα) were immunodetected. Nuclear proliferating cell nuclear antigen (PCNA) was analysed as an internal control. A bioinformatic approach was used to search the ERα-specific DNA-binding elements in the 5′-promoter regions of the BMP-6, Col I and OCN genes. M, marker of 100 bp DNA ladder.

Figure 6

Fig. 7 Effects of genistein on the mineralisation of primary rat osteoblasts. Rat osteoblasts prepared from rat calvaria were exposed to 10 μm-genistein (G), a differentiation reagent (DR, including 10 nm-dexamethasone (Dex), 100 μg ascorbic acid (AA)/ml and 10 mm-β-glycerophosphate (GP)), and a combination of genistein and the DR for 21 d. The drugs were renewed every 2 d. (a) Osteoblast mineralisation was determined using the Alizarin red S dye-staining protocol. (b) The amounts of mineralised nodules were quantified and analysed.