Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jul 1.
Published in final edited form as: J Biomed Mater Res A. 2013 Oct 17;102(7):2417–2426. doi: 10.1002/jbm.a.34909

Effects of SiO2, SrO, MgO and ZnO dopants in TCP on osteoblastic Runx2 expression

Gary A Fielding 1, Will Smoot 1, Susmita Bose 1,1
PMCID: PMC3979499  NIHMSID: NIHMS547289  PMID: 23946240

Abstract

Calcium phosphate materials share a compositional similarity to natural bone which makes them excellent for use in orthopedic applications. Although these materials are osteoconductive, they lack strong osteoinductive capabilities and recent research has focused on the addition of biologics and pharmacologics with varying successes. In this study, trace elements that have been proven to play important roles in bone health and bone formation were incorporated into β-tricalcium phosphate compacts in their oxide forms (SiO2, ZnO, SrO, and MgO). Cell material interactions were characterized using human fetal preosteoblastic cells. An MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was used to evaluate cellular proliferation. Cellular differentiation was evaluated using an enzymatic colorimetric alkaline phosphatase assay as well as immunohistochemistry for Runt-related transcription factor 2 (Runx2) expression. Results prove ZnO and MgO to be effective mitogenic factors and SiO2, ZnO and SrO to be capable of inducing rapid cellular differentiation. MgO was found to have little effect on the modulation of osteoblastic differentiation, likely due to more aggressive inherent cellular regulation of Mg2+. In addition to the results from the study, a signaling mechanism is proposed as to the action of the dopants for further consideration.

Keywords: bone tissue engineering, calcium phosphates, magnesium, strontium, zinc, silicon

1. Introduction

Calcium phosphate (CaP) ceramics are amongst the most widely studied materials for use in orthopedic implant applications due to their excellent biocompatibility, tailorable bioresorbability and compositional similarity to natural bone [13]. Current clinical uses include, but are not limited to, coatings on metallic implants, grafting materials and bone cement applications. [4]. The physical properties of CaPs are largely dependent on composition and phases present in the material. Of particular interest are the CaPs with a calcium-to-phosphorus ratio of 1.5:1, known as tricalcium phosphates (TCP). TCP has a solubility product (Ksp = 2.83 × 10−30) that allows it to degrade over the course of months under physiological conditions, allowing it to provide mechanical support while the natural healing process can take place [1,5]. Our group and others have shown that these degradation kinetics as well as the overall material strength can be further tailored by the addition of dopants or sintering additives [68]. Additionally, by choosing dopants that can play important biological roles in the bone healing process, such as strontium, magnesium, silicon and zinc, the biological interaction between the material and natural bone can be enhanced [911].

Strontium is a non-essential element that has the ability to replace calcium in natural bone tissue, so it is said to have bone seeking behavior[12]. It is thought to displace Ca2+ ions in osteoblastic mediated processes. Researchers have identified that strontium likely stimulates bone formation by activating the calcium sensing receptor (CaSR) [13,14] and simultaneously increasing osteoprotegerin (OPG) production and inhibiting receptor activator of nuclear factor kappa B ligand (RANKL) expression in osteoblasts [15]. OPG is a protein that inhibits RANKL induced osteoclastogenesis by operating as a decoy receptor for RANKL [16]. The OPG/RANKL ratio, then, can be a powerful regulator of bone resorption and osteoclastogenesis. Phase III clinical trials that began in 2000 investigated the efficacy of strontium ranelate in reducing vertebral fractures and peripheral fractures, including hip fractures. After 3 years, patients treated with strontium ranelate showed significant reduction in vertebral fractures (41%) and hip fractures (36%) compared with patients treated with placebo [17].

Magnesium is one of the most abundant cations in the human body, with about 65% of it being contained in bone and teeth [18]. It has the ability to influence intracellular Ca2+ homeostasis via the parathyroid hormone pathway [19]. Furthermore, studies have shown that magnesium deficiency can result in reduced bone growth, bone formation and mineralization [20,21].

Silicon has been proven to be an important trace element in bone and connective tissue formation and can stimulate biological activity by increasing the solubility of the material, generating a more electronegative surface and creating a finer microstructure resulting in transformation of the material surface to a biologically equivalent apatite [22]. Recent studies have also demonstrated that silicate containing bioceramics have the added benefit of inducing angiogenesis, which is a crucial aspect of bone defect healing [23,24].

Zinc, also an important trace element in bone formation, has been shown to have stimulatory effects on bone formation when added to CaP [25]. Zinc is released during skeletal breakdown and has shown to inhibit osteoclastic bone resorption [26]. It does this by inhibiting osteoclast-like cell formation from bone marrow cells and inducing apoptosis of mature osteoclast [27]. Osteoporotic patients have also been shown to have lower levels of skeletal zinc than that of control groups [28]. Zinc has also been shown to induce osteoblastogenesis, osteoblastic differentiation and mineralization [29,30].

While many benefits have been attributed to these particular elements in healthy bone biology, the mechanisms of action to which they may influence bone and the healing process is still largely a mystery. The aim of this study is to identify potential signaling pathways by which strontium, silicon, magnesium and zinc may play a role in osteoblastic differentiation. Runt related transcription factor 2 (Runx2) is essential for osteoblastic differentiation and skeletal morphogenesis and acts as a scaffold for nucleic acids and regulatory factors involved in skeletal gene expression [31]. It is vital for the maturation of osteoblasts and both intramembranous and endochondral ossification [32]. Alkaline phosphatase (ALP) has long been identified as an osteoblast differentiation marker, signaling the maturation of the extracellular matrix (ECM) produced by osteoblasts; the final step before terminal differentiation or apoptosis for osteoblast cells. By studying these two targets, which are seemingly unrelated in the differentiation process, insight may be gained as to which signaling pathways should be studied further.

2. Methods

2.1 Sample preparation

β-tricalcium phosphate powder (β-TCP) was obtained from Berkeley Advanced Biomaterials (Berkeley, CA) with an average particle size of 550 nm. High purity strontium oxide (SrO) (99.9% purity) was purchased from Sigma Aldrich ( St. Louis, MO) and magnesium oxide (MgO, 99.998%) was procured from Alfa Aesar, MA, USA. Silicon dioxide (SiO2) (99%+ purity) and zinc oxide (ZnO) (99.9%+ purity) were purchased from Fisher Scientific (Fair Lawn, NJ). Samples were prepared by mixing 50 g of β-TCP powder and appropriate amounts of dopants (1 wt% SrO, 1 wt% MgO, 0.5 wt% SiO2 and 0.25 wt% ZnO) in 250 mL polypropylene Nalgene bottles containing 75 mL of anhydrous ethanol and 100 g zirconia milling media with 5mm diameter. Dopant concentrations were chosen based on previous optimization research [6,7,9,10] The mixtures were then milled for 6 h at 70 rpm to minimize the formation of agglomerates and increase homogeneity. After milling, powder was dried in an oven at 60 °C for 72 h and pressed to discs (12 mm diameter and 2.5 mm thickness) using a uniaxial press at 145 MPa. Green compacts were then cold isostatically pressed at 414 MPa for 5 min and sintered at 1250 °C for 2 h in a muffle furnace.

2.2 Sample Density

Density was measured using Archimedes method. Samples were weighed initially dry and then submerged in boiling water for 3 minutes to remove any excess air that may be trapped in the porous structure. The samples were then transferred from the boiling water to room temperature water, where the weight was recorded again (n=3).

2.2 Cell culture

All samples were sterilized by autoclaving at 121 °C for 20 min. In this study, established human preosteoblast cell line hFOB 1.19 (ATCC, Manassas, VA) were used. Cells were seeded onto the samples in 24-well plates at a density of 105 cells/sample. The base medium for this cell line was a 1:1 mixture of Ham's F12 Medium and Dulbecco's Modified Eagle's Medium (DMEM/F12, Sigma, St. Louis, MO), with 2.5 mM L-glutamine (without phenol red). The medium was supplemented with 10% fetal bovine serum (HyClone, Logan, UT) and 0.3 mg/ml G418 (Sigma, St. Louis, MO). Cultures were maintained at 34 °C under an atmosphere of 5% CO2 as recommended by ATCC for this particular cell line. Medium was changed every 2 days for the duration of the experiment.

2.4 Cellular proliferation

MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was used to evaluate cell proliferation. The MTT (Sigma, St. Louis, MO) solution of 5 mg/ml was prepared by dissolving MTT in sterile filtered PBS. 10% MTT solution was then added to each sample in 24-well plates. After 2 h of incubation, 1 ml of solubilization solution made up of 10% Triton X-100, 0.1N HCl and isopropanol was added to dissolve the formazan crystals. 100 μl of the resulting supernatant was transferred into a 96-well plate, and read by a plate reader at 570 nm. Triplicate samples were used in MTT assay experiments to insure reproducibility. Data was normalized to the absorbance values for the pure TCP control.

2.4 Cellular morphology

Samples for testing were removed from culture after 3 days of incubation. All samples for SEM observation were fixed with 2% paraformaldehyde/2% glutaraldehyde in 0.1M cacodylate buffer overnight at 4 °C. Post-fixation was performed with 2% osmium tetroxide (OsO4) for 2 h at room temperature. The fixed samples were then dehydrated in an ethanol series (30%, 50%, 70%, 95% and 100% three times), followed by a hexamethyldisilane (HMDS) drying procedure. After gold coating, the samples were observed under field emission scanning electron microscope (FESEM) (FEI 200F, FEI Inc., OR, USA) for cell morphologies.

2.5 Alkaline phosphatase activity

The alkaline phosphatase (ALP) activity of the cells was determined by a spectrophotometric endpoint assay that determined the conversion of colorless p-nitrophenyl phosphate to colored p-nitrophenol. Cell media was aspirated from each sample at 7 and 11 days and samples were subsequently transferred to new 24 well plates. 350 μl 2-amino-2-methyl-propanol buffer and 500 μl p-nitrophenyl phosphate (4 mg/ml) were added. Samples were incubated for 15 min at 37° C and stopped by the addition of 500 μl of 2M NaOH to each well. The solution was then transferred to a 96 well plate in triplicate and absorbance was read at 405 nm. Triplicates of each sample were examined to ensure reproducibility. Data was normalized to MTT data to account differences in cellular proliferation/expression.

2.6 Runx2 activity

Samples were fixed in 3.7% paraformaldehyde/ phosphate buffered solution with a pH of 7.4 at room temperature for 10 min. Samples were then washed in PBS 3 times (5 min each) and cells were permeabilized with 0.1% Triton X-100 (in PBS) for 4 min at room temperature. Next, samples were rinsed in TBST 3 times (5 minutes each) and incubated in TBST-BSA (Tris-buffered saline with 1% bovine serum albumin, 250 mM NaCl, pH 8.3) blocking solution for 1h at room temperature. Primary antibody against Runx2 (Abcam, Cambridge, MA) was added at a 1:100 dilution and incubated at room temperature for 2 h and kept at 4 °C overnight. Samples were then washed with TBST 3 times (10 min each). The secondary antibody, goat anti-mouse (GAM) Oregon green 488 (Molecular Probes, Eugene, OR), was diluted 1:100 in TBST and was used to incubate the cells for 1h. After rinsing three times for 10 minutes each with TBST the samples placed in 24 well plates with Vectashield mounting medium (Vector Labs, Burlingame, CA) with propidium iodide (PI). Samples were immediately analyzed by use of fluorescent area scans using a 5×5 grid pattern on a Synergy Hybrid multi-mode microplate reader (Biotek, Winooski, VT) with excitation wavelength of 488 nm and emission wavelength of 540 nm. Data was normalized to account for differences in cellular proliferation/expression. Afterwards, samples were placed on glass coverslips and confocal micrographs were taken using a Zeiss 510 laser scanning microscope (LSM 510 META, Carl Zeiss MicroImaging, Inc., NY, USA).

2.7 Statistical analysis

Statistical analysis was performed using a one way ANOVA and p<0.05 was considered statistically significant.

3. Results

3.1 Density

All samples exhibited very high density, >95%, as seen in Table 1. None of the samples demonstrated a significant difference in density in the group.

Table 1.

Relative densities of sintered samples

Sample Relative Density
Pure TCP 96.17 ± 0.89
Si – TCP 97.02 ± 1.47
Zn – TCP 98.07 ± 0.52
Sr - TCP 95.02 ± 1.39
Mg - TCP 98.61 ± 0.97

3.1 Cellular proliferation

At day 3 (Figure 4), all samples showed decreased cellular proliferation when compared to the pure TCP control with ZnO-TCP samples having significantly lower living cells. At day 7 (Figure 1), both ZnO-TCP and MgO-TCP samples had significantly greater amount of live cells when compared to the pure TCP controls, while the SiO2-TCP and SrO-TCP samples where comparable in live cell density to the pure TCP controls. By day 11 (Figure 2/5) SiO2-TCP and SrO-TCP had lower live cell density, ZnO-TCP had comparable cell density and MgO-TCP showed increased cellular density when compared to pure TCP controls.

Figure 4.

Figure 4

MTT measurements for cellular proliferation and Runx2 fluorescent measurements for cellular differentiation at day 3 for SiO2, ZnO, SrO, MgO doped and pure TCP samples. Data is normalized to pure TCP control. Runx2 data is normalized to MTT measurements. *P < 0.05 (n=3)

Figure 1.

Figure 1

MTT measurements for cellular proliferation and ALP measurements for cellular differentiation at day 7 for SiO2, ZnO, SrO, MgO doped and pure TCP samples. Data is normalized to pure TCP control. ALP data is normalized to MTT measurements. *P < 0.05 (n=3)

Figure 2.

Figure 2

MTT measurements for cellular proliferation and ALP measurements for cellular differentiation at day 11 for SiO2, ZnO, SrO, MgO doped and pure TCP samples. Data is normalized to pure TCP control. ALP data is normalized to MTT measurements. *P < 0.05 (n=3)

Figure 5.

Figure 5

MTT measurements for cellular proliferation and Runx2 fluorescent measurements for cellular differentiation at day 11 for SiO2, ZnO, SrO, MgO doped and pure TCP samples. Data is normalized to pure TCP control. Runx2 data is normalized to MTT measurements. *P < 0.05 (n=3)

3.2 Cellular morphology

Micrographs depicting cellular morphology are given in Figure 3. At day 3 all samples demonstrated a barely visible confluent monolayer of cells. At day 7, a second layer of cells was reaching confluency on all samples. By day 11, all samples showed multiple layers of cells with evidence of early stages of mineralization.

Figure 3.

Figure 3

FESEM micrographs depicting hFOB cell morphology after 3, 7 and 11 days in culture

3.3 Alkaline phosphatase activity

Expression of ALP was evaluated for hFOB cells cultured on SiO2-TCP, ZnO-TCP, SrO-TCP, MgO-TCP and pure TCP samples. Results after 7 and 11 days of culture are shown in Figure 1 and Figure 2, respectively. At day 7, ALP activity was significantly lower in all doped samples when compared to the pure TCP samples. At day 11, ZnO-TCP and SrO-TCP samples demonstrated higher ALP activity, while SiO2-TCP samples had decreased activity and MgOTCP samples had comparable activity to the pure TCP control samples.

3.4 Runx2 activity

Expression of Runx2 in osteoblast cells cultured on doped and pure TCP samples were evaluated at days 3 and 11. Results for fluorescent area scans for day 3 are given in Figure 4 and results for day 11 are given in Figure 5. Confocal micrographs for each day are shown in Figure 6, where cellular nuclei fluoresce in red and green indicates Runx2 expression. At day 3, SiO2-TCP, ZnO-TCP and SrO TCP all showed increased Runx2 expression when compared to pure TCP samples, with SiO2-TCP and ZnO-TCP samples being significantly greater. MgO doped samples showed less Runx2 activity, but not at a significant level. At day 11, samples containing SiO2 and ZnO had comparable Runx2 expression to pure TCP control samples, while both SrOTCP and MgO-TCP samples had decreased Runx2 expression. The MgO-TCP samples demonstrated significantly lower Runx2 expression than the pure TCP samples. In general, when compared with day 3 data, Runx2 expression decreased for all samples with respect to pure TCP. Confocal micrographs confirm the results of the quantitative fluorescent area scans, showing similar patterns of Runx2 expression.

Figure 6.

Figure 6

Confocal micrographs showing Runx2 expression in hFOB cells at 3 and 11 days. Green fluorescence indicates active Runx2; red fluorescence indicates propidium iodide bound to the nuclei of cells.

4. Discussion

Previous studies have well characterized the inherent material properties associated with these dopants [6,7,9,10]. The use of isostatic pressing, eliminates most of the inherent density differences associated with dopant addition, resulting in a very high average density ranging from 95% - 98% (Table 1) with no statistically significant differences detected. Studies have also shown that the addition of any single dopant used in this study has the ability to reduce α phase formation associated with a high sintering temperature when compared to the pure TCP samples [6,7,9,10]. These two properties are extremely important when thinking about potential differences in dissolution between sample types. The pure control composition has higher alpha phase so its dissolution rate will be higher than its doped experimental samples if the densities are similar, however, dissolution rate will be minimal considering the time frame of the study as seen in previous studies [33,34]. It is well documented that increased Ca2+ and P5+ can positively affect cellular differentiation and proliferation [3537], but the results observed in this study demonstrated that samples containing dopants had significant positive effects when compared to the pure composition. It is then very likely, then, that the cells are responding to the dopants rather than Ca2+ or P5+ due to differences in dissolution.

The osteoblastic cell cycle is a process comprised of three distinct stages (Figure 7): proliferation, maturation and termination. During the proliferation stage and part of the maturation stage, Runx2 plays a stimulatory role in differentiation of mesenchymal stem cells (MSCs), osteochondrogenic precursors and preosteoblasts. During the latter portion of the maturation stage and the terminal differentiation stage, Runx2 plays an inhibitory role in osteoblastic differentiation [38]. It is, therefore, necessary for the upregulation of Runx2 initially and the downregulation of Runx2 during the latter part of the cell cycle to obtain mature bone growth. Results from the MTT assay at day 3 (Figure 4) suggest an expeditive transition from the proliferation stage of the osteoblast life cycle to the maturation stage in samples containing SiO2, ZnO and MgO as evidenced by the decrease in proliferation rates. Samples doped with SrO are comparable to that of the pure TCP samples. Research has shown that Sr is able to augment proliferation of osteoblast cells while maintaining increased differentiation rates [39], so it is possible that cells seeded on the SrO doped samples are also in the maturation phase. SEM micrographs (Figure 3) of cells at day 3 show characteristics of mature osteoblasts in all samples manifested by the broadening and flattening of cells as well as the extension of cellular processes and cellular aggregation. At day 7 (Figure 1), MTT results show increased proliferation in samples containing ZnO and MgO compared to pure TCP. The other samples had similar values to pure TCP, indicating that cells on the pure TCP samples had reached advanced maturity and the resultant slowing of proliferation. The increased level of viable cells in the MgO and ZnO doped samples can be attributed to the fact that Zn2+ and Mg2+ are both mitogenic factors and may prevent terminal apoptosis of osteoblast cells [40,41]. SEM micrographs show the formation of multiple layers of mature osteoblast cells on all samples. Day 11 MTT results (Figure 2/5) indicate an affinity for active terminal differentiation in samples containing SiO2 and SrO demonstrated by decreased cellular viability (increased terminal apoptosis). Samples containing MgO had increased proliferation rates, while ZnO doped samples were comparable to pure TCP indicating a lasting effect of Mg2+ as a mitogenic factor. SEM images (Figure 3) for day 11 show all doped samples demonstrate advanced characteristics of cellular differentiation when compared to pure TCP samples, embodied by the sheet-like formation of bone lining cells, where single cells are often difficult to distinguish, as well as apoptosis and mineralization.

Figure 7.

Figure 7

Diagram depicting the osteoblast cell lifecycle and influence of Runx2 [52,53].

Results from the ALP assay reveal that at day 7 (Figure 1), all samples show significantly lower activity when compared to pure TCP. At day 11 (Figure 2), ALP activity in SiO2 doped samples was less than pure TCP, MgO doped samples comparable and ZnO and SrO doped samples significantly greater. Alkaline phosphatases are glycosyl-phosphatidylinositol anchored, Zn2+ metallated glycoproteins that are released during the beginning of the termination phase of the osteoblastic cell life cycle and help to catalyze the hydrolysis of phosphomonoesters into inorganic phosphates [42]. They create an alkaline environment that favors the mineralization of these inorganic phosphates. These results suggest a prolonged maturation phase in the ZnO and MgO doped samples through day 7, likely caused by increased Runx2 activity in the samples through this time period. Researchers have also shown that the Zn2+ binding sites in ALP may be easily substituted by other cations, where differing cationic radii and charge can act as destabilizing forces in the enzyme causing decreased functionality [43]. This phenomenon becomes apparent when considering day 11 results. ZnO and SrO have a large increase in functional enzymatic ALP activity while SiO2 is continuing to show destabilization when compared to the pure TCP samples. Zn2+, being the native cation in ALP, is expected to show increased activity. While the ionic radii of Sr2+ is dissimilar to Zn2+ (132pm and 88 pm, respectively), Sr2+ seems to act as an agonist for ALP activity. Si4+, likely due to vast differences in ionic radii (54 pm) and charge, may cause destabilization of the ALP enzyme. Mg2+ is very similar in size (86 pm) and charge to Zn2+ and has been demonstrated to have very little effect on the stability of ALP [43], so it was expected that results would be similar to samples doped with ZnO. This was not the observation, however. It is apparent that MgO doped samples are only producing as much ALP as the pure samples at day 11where we expected to see an increase in activity. Two different mechanisms may be responsible for these findings. The first, we still see significant increase in proliferation rates with samples doped with MgO, so it may be that samples remain in the early maturation stage. Runx2 findings, however, do not correlate well with this. They should remain at high levels during states where proliferation is prevailing. The second mechanism would suggest that Mg2+ is well regulated along the differentiation pathways (as we still see increases in cellular proliferation). This seems to be the more likely scenario as Mg2+ is one of the most abundantly available cations in the human body and is expected to be more highly regulated.

Results from this study indicate that at day 3 (Figure 4), Runx2 expression is significantly upregulated in samples containing SiO2 and ZnO. SrO doped samples also showed upregulation of Runx2, but not quite at a significant level. At day 11 (Figure 5), samples containing SiO2 and ZnO show Runx2 expression to be about the same as pure TCP. Samples containing MgO and SrO show downregulation of Runx2, with MgO doped samples giving significant results. Confocal microscopy (Figure 6) confirms these findings. Overall results signify that samples containing SiO2, ZnO and SrO can appropriately modulate Runx2 activity leading to accelerated differentiation of osteoblast cells when compared to pure TCP samples. A proposed signaling mechanism for the effect of dopants on Runx2 expression is offered in Figure 8. One of the major regulators of osteoblastic cellular differentiation, and in particular Runx2, is the canonical Wnt signaling pathway[32,44]. The non-canonical Wnt5 calcium-dependent signaling pathway may also play an important role in the osteoblastic differentiation process and has been shown to cross paths with other Wnt isoform signaling pathways [45]. Several studies have presented that dopants, such as Sr2+, can interact with the calcium sensing receptor (CaSR) in osteoblastic cells to moderate cellular processes [13,46]. In the presence of high extracellular Ca2+, Wnt5 is activated through the CaSR and binds to the Frizzled G protein-coupled receptor and starts the intracellular Ca2+ signaling cascade [47]. There are extensive reviews available concerning this cascade [44,48]. Briefly, once Wnt is bound, the cytosolic G protein breaks off into its α and β/γ subunits. The β/γ then activates membrane bound phospholipase C (PLC) which, in turn, hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into second messengers inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). IP3 then diffuses through the cytosol and activates calcium channels located in the cellular membrane and endoplasmic reticulum causing an influx of intracellular calcium. Studies have shown that calcium channels can be non-selective to certain ions, including Sr2+, which may allow entrance of the dopants into the cell[49]. Other studies also suggest that the endoplasmic reticulum may also allow for the storage of cations such as Mg2+, Sr2+ and Zn2+ [5052]. The influx of Ca2+ into the cell leads to the activation of calcineurin (CaN) and Ca2+-calmodulin-dependent protein kinase II (CamKII), while DAG activates protein kinase C (PKC). PKC activates the translocation of cyclic adenosine monophosphate (cAMP) response element-binding (CREB) and nuclear factor kappa B (NFκB) from the cytoplasm to the nucleus. CamKII also plays a role in the translocation of NFκB. CaN is responsible for the permutation of Nuclear factor of activated T-cells (NFAT) into the nucleus. CREB, NFκB and NFAT have all been shown to play important roles in the regulation of Runx2 [5355]. The ability of cations to act as a competitive agonist with Ca2+ for the activation of CaN or CamKII has been established with strontium. Fromique et al. demonstrated that strontium ranelate induced NFATc1 nuclear translocation and was completely abrogated with CaN inhibitors[56]. It is likely that other dopants may interact in a similar manner, although confirmation studies are still required to confirm this.

Figure 8.

Figure 8

Activation of the Wnt/Fz ligand-receptor leads to the production of the second messengers IP3 and DAG from membrane-bound PIP2 via the action of membrane-bound enzyme PLC. IP3 causes release of Ca2 from the ER and extracellular Ca2+ influx through transmembrane Ca2+ channels; CaN and CamKII are activated which in turn activate NFAT and NFkB. DAG is also activated by increased inctracellular Ca2+, which activates PKC. PKC activates NFkB and CREB. NFAT, NFkB, and CREB translocate to the nucleus and transcribe downstream regulatory genes such as Runx2.

A recapitulating theme throughout this study is the apparent inactivity of Mg2+ in the osteoblastic cellular differentiation process as evidenced by results from Runx2 and ALP analysis. In the human body, Mg2+ is the second most abundant cation and is known to be a cofactor in over 300 enzymatic reactions ranging from energy metabolism to nucleic acid synthesis [57]. Because it is so abundant, it is also highly regulated and can even act as a Ca2+ ion channel blocker [58]. Another study demonstrated that while some calcium ion channels may be non-selective to some cations, Mg2+ efflux remained low through these same channels [49]. While dietary deficiencies have been linked to osteoporosis, this study provides no evidence that an abundance of Mg2+ will help in the maturation and differentiation of osteoblastic cells. On the other hand, significant results were seen in cellular proliferation at days 7 and 11, confirming Mg2+ as a valuable mitogenic element. SrO, ZnO and SiO2 all seemed to have positive effects on Runx2 regulation, increasing early expression and decreasing at the latter stages of the cell life cycle indicating the ability for accelerated differentiation when compared to pure TCP samples. The mineralization capabilities of the osteoblast cells were also positively affected by the ZnO and SrO dopants, demonstrated by ALP activity at day 11. Overall results suggest that ZnO, SrO and SiO2 dopants can have profound effects on the differentiation capabilities of osteoblast cells in vitro and likely act through the intracellular Ca2+ signaling pathway as a competitive agonist with calcium to activate key proteins in this pathway. While Si4+ was found to cause functional destabilization of ALP, a key enzyme in the mineralization process of osteoblasts, it has also been shown that it is an important trace element in bone and connective tissue formation and can stimulate biological activity by increasing the solubility of the CaP material, generating a more electronegative surface and it can also create a finer microstructure resulting in a nucleation site that is favorable for apatite formation [22,59].

5. Conclusions

CaP materials have been widely studied for use as an osteogenic bone replacement material and are effectively used in several medical and dental applications today. In this study, by incorporating the dopants SiO2, ZnO, and SrO into TCP, the important transcription factor Runx2 was able to be modulated in a beneficial manner as to induce accelerated bone cell differentiation, with high expression in the early stages of cell maturation and low expression in the terminal stage. ALP activity was increased with the introduction of ZnO and SrO into the samples when compared to pure TCP indicating increased affinity for matrix mineralization. ZnO and MgO dopants both demonstrated favorable mitogenic properties, but Mg did not have any significant effect on the differentiation markers. In addition to these results a signaling mechanism was proposed as to the action of these dopants for consideration of further study and understanding of the system.

Acknowledgements

Authors would like to acknowledge financial support from the National Institutes of Health, NIBIB (Grant # NIH-R01-EB-007351). The authors wish to acknowledge the help rendered by Will Smoot, School of Mechanical and Materials Engineering, as well as the Franceschi Microscopy & Imaging Center.

References

  • 1.Bose S, Tarafder S. Calcium phosphate ceramic systems in growth factor and drug delivery for bone tissue engineering: A review. Acta Biomater. 2012;8:1401–21. doi: 10.1016/j.actbio.2011.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bai X, Sandukas S, Appleford MR, et al. Deposition and investigation of functionally graded calcium phosphate coatings on titanium. Acta Biomater. 2009;5:3563–72. doi: 10.1016/j.actbio.2009.05.013. [DOI] [PubMed] [Google Scholar]
  • 3.Ginebra MP, Espanol M, Montufar EB, et al. New processing approaches in calcium phosphate cements and their applications in regenerative medicine. Acta Biomater. 2010;6:2863–73. doi: 10.1016/j.actbio.2010.01.036. [DOI] [PubMed] [Google Scholar]
  • 4.Brown PW. Encyclopedia of Materials: Science and Technology. Second Edition Elsevier; Oxford: 2001. Calcium Phosphates in Biomedical Engineering. pp. 893–7. [Google Scholar]
  • 5.Saikia K, Bhattacharya T, Bhuyan S, et al. Calcium phosphate ceramics as bone graft substitutes in filling bone tumor defects. Indian J Orthop. 2008;42:169–72. doi: 10.4103/0019-5413.39588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bandyopadhyay A, Bernard S, Xue W, et al. Calcium Phosphate-Based Resorbable Ceramics: Influence of MgO, ZnO, and SiO2 Dopants. J Am Ceram Soc. 2006;89:2675–88. [Google Scholar]
  • 7.Xue W, Dahlquist K, Banerjee A, et al. Synthesis and characterization of tricalcium phosphate with Zn and Mg based dopants. J Mater Sci Mater Med. 2008;19:2669–77. doi: 10.1007/s10856-008-3395-4. [DOI] [PubMed] [Google Scholar]
  • 8.Bohner M. Silicon-substituted calcium phosphates–a critical view. Biomaterials. 2009;30:6403–6. doi: 10.1016/j.biomaterials.2009.08.007. [DOI] [PubMed] [Google Scholar]
  • 9.Banerjee SS, Tarafder S, Davies NM, et al. Understanding the influence of MgO and SrO binary doping on the mechanical and biological properties of [beta]-TCP ceramics. Acta Biomater. 2010;6:4167–74. doi: 10.1016/j.actbio.2010.05.012. [DOI] [PubMed] [Google Scholar]
  • 10.Bose S, Tarafder S, Banerjee SS, et al. Understanding in vivo response and mechanical property variation in MgO, SrO and SiO2 doped β-TCP. Bone. 2011;48:1282–90. doi: 10.1016/j.bone.2011.03.685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Castellani C, Lindtner RA, Hausbrandt P, et al. Bone–implant interface strength and osseointegration: Biodegradable magnesium alloy versus standard titanium control. Acta Biomater. 2011;7:432–40. doi: 10.1016/j.actbio.2010.08.020. [DOI] [PubMed] [Google Scholar]
  • 12.Bärenholdt O, Kolthoff N, Nielsen SP. Effect of long-term treatment with strontium ranelate on bone strontium content. Bone. 2009;45:200–6. doi: 10.1016/j.bone.2009.04.196. [DOI] [PubMed] [Google Scholar]
  • 13.Coulombe J, Faure H, Robin B, et al. In vitro effects of strontium ranelate on the extracellular calcium-sensing receptor. Biochem Biophys Res Commun. 2004;323:1184–90. doi: 10.1016/j.bbrc.2004.08.209. [DOI] [PubMed] [Google Scholar]
  • 14.Brown EM. Is the calcium receptor a molecular target for the actions of strontium on bone? Osteoporos Int. 2003;14:25–34. doi: 10.1007/s00198-002-1343-6. [DOI] [PubMed] [Google Scholar]
  • 15.Tat SK, Pelletier JP, Mineau F, et al. Strontium ranelate inhibits key factors affecting bone remodeling in human osteoarthritic subchondral bone osteoblasts. Bone. 2011 doi: 10.1016/j.bone.2011.06.005. [DOI] [PubMed] [Google Scholar]
  • 16.Kostenuik PJ. Osteoprotegerin A Physiological and Pharmacological Inhibitor of Bone Resorption. Curr Pharm Des. 2001;7:613–35. doi: 10.2174/1381612013397807. others. [DOI] [PubMed] [Google Scholar]
  • 17.Meunier PJ, Roux C, Seeman E, et al. The effects of strontium ranelate on the risk of vertebral fracture in women with postmenopausal osteoporosis. N Engl J Med. 2004;350:459–68. doi: 10.1056/NEJMoa022436. [DOI] [PubMed] [Google Scholar]
  • 18.Rude RK, Gruber HE, Norton HJ, et al. Dietary magnesium reduction to 25% of nutrient requirement disrupts bone and mineral metabolism in the rat. Bone. 2005;37:211–9. doi: 10.1016/j.bone.2005.04.005. [DOI] [PubMed] [Google Scholar]
  • 19.Paunier L. Effect of magnesium on phosphorus and calcium metabolism. Monatsschrift Kinderheilkd Organ Dtsch Ges Für Kinderheilkd. 1992;140:S17–20. [PubMed] [Google Scholar]
  • 20.Stendig-Lindberg G, Koeller W, Bauer A, et al. Experimentally induced prolonged magnesium deficiency causes osteoporosis in the rat. Eur J Intern Med. 2004;15:97–107. doi: 10.1016/j.ejim.2004.01.003. [DOI] [PubMed] [Google Scholar]
  • 21.Eisinger JCD. Effects of silicon, fluoride, etidronate and magnesium on bone mineral density: a retrospective study. Magnes Res. 1993;6:247–9. [PubMed] [Google Scholar]
  • 22.Pietak AM, Reid JW, Stott MJ, et al. Silicon substitution in the calcium phosphate bioceramics. Biomaterials. 2007;28:4023–32. doi: 10.1016/j.biomaterials.2007.05.003. [DOI] [PubMed] [Google Scholar]
  • 23.Zhai W, Lu H, Chen L, et al. Silicate bioceramics induce angiogenesis during bone regeneration. Acta Biomater. 2012;8:341–9. doi: 10.1016/j.actbio.2011.09.008. [DOI] [PubMed] [Google Scholar]
  • 24.Alt V, Kögelmaier DV, Lips KS, et al. Assessment of angiogenesis in osseointegration of a silica-collagen biomaterial using 3D-nano-CT. Acta Biomater. 2011;7:3773–9. doi: 10.1016/j.actbio.2011.06.024. [DOI] [PubMed] [Google Scholar]
  • 25.Porter AE, Patel N, Skepper JN, et al. Effect of sintered silicate-substituted hydroxyapatite on remodelling processes at the bone–implant interface. Biomaterials. 2004;25:3303–14. doi: 10.1016/j.biomaterials.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 26.Yamaguchi M. Role of nutritional zinc in the prevention of osteoporosis. Mol Cell Biochem. 2009;338:241–54. doi: 10.1007/s11010-009-0358-0. [DOI] [PubMed] [Google Scholar]
  • 27.Murray EJ, Messer HH. Turnover of bone zinc during normal and accelerated bone loss in rats. J Nutr. 1981;111:1641–7. doi: 10.1093/jn/111.9.1641. [DOI] [PubMed] [Google Scholar]
  • 28.Reginster J, Saltman O, Franchimont P. Trace elements and postmenopausal osteoporosis: a preliminary study of decreased serum manganese. J Nutr. 1981;111:1641–7. [Google Scholar]
  • 29.Kwun IS, Cho YE, Lomeda RAR, et al. Zinc deficiency suppresses matrix mineralization and retards osteogenesis transiently with catch-up possibly through Runx 2 modulation. Bone. 2010;46:732–41. doi: 10.1016/j.bone.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • 30.Yamaguchi M, Goto M, Uchiyama S, et al. Effect of zinc on gene expression in osteoblastic MC3T3-E1 cells: enhancement of Runx2, OPG, and regucalcin mRNA expressions. Mol Cell Biochem. 2008;312:157–66. doi: 10.1007/s11010-008-9731-7. [DOI] [PubMed] [Google Scholar]
  • 31.Long F. Building strong bones: molecular regulation of the osteoblast lineage. Nat Rev Mol Cell Biol. 2011;13:27–38. doi: 10.1038/nrm3254. [DOI] [PubMed] [Google Scholar]
  • 32.Lin GL, Hankenson KD. Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation. J Cell Biochem. 2011;112:3491–501. doi: 10.1002/jcb.23287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Roy M, Fielding GA, Bandyopadhyay A, et al. Effects of zinc and strontium substitution in tricalcium phosphate on osteoclast differentiation and resorption. Biomater Sci. 2012;1:74–82. doi: 10.1039/C2BM00012A. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Roy M, Bose S. Osteoclastogenesis and osteoclastic resorption of tricalcium phosphate: Effect of strontium and magnesium doping. J Biomed Mater Res A. 2012;100A:2450–61. doi: 10.1002/jbm.a.34181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Maeno S, Niki Y, Matsumoto H, et al. The effect of calcium ion concentration on osteoblast viability, proliferation and differentiation in monolayer and 3D culture. Biomaterials. 2005;26:4847–55. doi: 10.1016/j.biomaterials.2005.01.006. [DOI] [PubMed] [Google Scholar]
  • 36.Eklou-Kalonji E, Denis I, Lieberherr M, et al. Effects of extracellular calcium on the proliferation and differentiation of porcine osteoblasts in vitro. Cell Tissue Res. 1998;292:163–71. doi: 10.1007/s004410051046. [DOI] [PubMed] [Google Scholar]
  • 37.Ma S, Yang Y, Carnes DL, et al. Effects of dissolved calcium and phosphorous on osteoblast responses. J Oral Implantol. 2005;31:61–7. doi: 10.1563/0-742.1. [DOI] [PubMed] [Google Scholar]
  • 38.Komori T. Regulation of bone development and extracellular matrix protein genes by RUNX2. Cell Tissue Res. 2010;339:189–95. doi: 10.1007/s00441-009-0832-8. [DOI] [PubMed] [Google Scholar]
  • 39.Sila-Asna M, Bunyaratvej A, Maeda S, et al. Osteoblast differentiation and bone formation gene expression in strontium-inducing bone marrow mesenchymal stem cell. Kobe J Med Sci. 2007;53:25–35. [PubMed] [Google Scholar]
  • 40.Leidi M, Dellera F, Mariotti M, et al. Nitric oxide mediates low magnesium inhibition of osteoblast-like cell proliferation. J Nutr Biochem. doi: 10.1016/j.jnutbio.2011.06.016. doi:10.1016/j.jnutbio.2011.06.016. [DOI] [PubMed] [Google Scholar]
  • 41.Guo B, Yang M, Liang D, et al. Cell apoptosis induced by zinc deficiency in osteoblastic MC3T3-E1 cells via a mitochondrial-mediated pathway. Mol Cell Biochem. 2012;361:209–16. doi: 10.1007/s11010-011-1105-x. [DOI] [PubMed] [Google Scholar]
  • 42.Fielding GA, Roy M, Bose S, et al. Antibacterial and biological characteristics of plasma sprayed silver and strontium doped hydroxyapatite coatings. Acta Biomater. 2012 doi: 10.1016/j.actbio.2012.04.004. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chlebowski JF, Mabrey S, Falk MC. Calorimetry of Alkaline Phosphatase. Stability of the Monomer and Effect of Metal Ion and Phosphate Binding on Dimer Stability. J Biol Chem. 1979;254:5745–53. [PubMed] [Google Scholar]
  • 44.Yavropoulou MP, Yovos JG. The role of the Wnt signaling pathway in osteoblast commitment and differentiation. Horm-Athens. 2007;6:279. doi: 10.14310/horm.2002.1111024. others. [DOI] [PubMed] [Google Scholar]
  • 45.Bradley EW, Drissi MH. WNT5A Regulates Chondrocyte Differentiation through Differential Use of the CaN/NFAT and IKK/NF- B Pathways. Mol Endocrinol. 2010;24:1581–93. doi: 10.1210/me.2010-0037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chattopadhyay N, Quinn SJ, Kifor O, et al. The calcium-sensing receptor (CaR) is involved in strontium ranelate-induced osteoblast proliferation. Biochem Pharmacol. 2007;74:438–47. doi: 10.1016/j.bcp.2007.04.020. [DOI] [PubMed] [Google Scholar]
  • 47.MacLeod RJ, Hayes M, Pacheco I. Wnt5a Secretion Stimulated by the Extracellular Calcium-Sensing Receptor Inhibits Defective Wnt Signaling in Colon Cancer Cells. Am J Physiol - Gastrointest Liver Physiol. 2007;293:G403–G411. doi: 10.1152/ajpgi.00119.2007. [DOI] [PubMed] [Google Scholar]
  • 48.De A. Wnt/Ca2+ Signaling Pathway: A Brief Overview. Acta Biochim Biophys Sin. 2011;43:745–56. doi: 10.1093/abbs/gmr079. [DOI] [PubMed] [Google Scholar]
  • 49.Chen X-Z, Vassilev PM, Basora N, et al. Polycystin-L is a calcium-regulated cation channel permeable to calcium ions. Nature. 1999;401:383–6. doi: 10.1038/43907. [DOI] [PubMed] [Google Scholar]
  • 50.Hogstrand C, Kille P, Nicholson RI, et al. Zinc transporters and cancer: a potential role for ZIP7 as a hub for tyrosine kinase activation. Trends Mol Med. 2009;15:101–11. doi: 10.1016/j.molmed.2009.01.004. [DOI] [PubMed] [Google Scholar]
  • 51.Rasgado-Flores H, Sanchez-Armass S, Blaustein MP, et al. Strontium, barium, and manganese metabolism in isolated presynaptic nerve terminals. Am J Physiol. 1987;252:C604–610. doi: 10.1152/ajpcell.1987.252.6.C604. [DOI] [PubMed] [Google Scholar]
  • 52.Günther T. Functional compartmentation of intracellular magnesium. Magnesium. 1986;5:53–9. [PubMed] [Google Scholar]
  • 53.Fan Q-M, Yue B, Bian Z-Y, et al. The CREB-Smad6-Runx2 axis contributes to the impaired osteogenesis potential of bone marrow stromal cells in fibrous dysplasia of bone. J Pathol Published Online First. 2012 Mar 27; doi: 10.1002/path.4033. doi:10.1002/path.4033. [DOI] [PubMed] [Google Scholar]
  • 54.Cho HH, Shin KK, Kim YJ, et al. NF-κB activation stimulates osteogenic differentiation of mesenchymal stem cells derived from human adipose tissue by increasing TAZ expression. J Cell Physiol. 2010;223:168–77. doi: 10.1002/jcp.22024. [DOI] [PubMed] [Google Scholar]
  • 55.Koga T, Matsui Y, Asagiri M, et al. NFAT and Osterix cooperatively regulate bone formation. Nat Med. 2005;11:880–5. doi: 10.1038/nm1270. [DOI] [PubMed] [Google Scholar]
  • 56.Fromigué O, Haÿ E, Barbara A, et al. Essential Role of Nuclear Factor of Activated T Cells (NFAT)-Mediated Wnt Signaling in Osteoblast Differentiation Induced by Strontium Ranelate. J Biol Chem. 2010;285:25251–8. doi: 10.1074/jbc.M110.110502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Elin RJ. Magnesium: the fifth but forgotten electrolyte. Am J Clin Pathol. 1994;102:616–22. doi: 10.1093/ajcp/102.5.616. [DOI] [PubMed] [Google Scholar]
  • 58.Kubota T, Shindo Y, Tokuno K, et al. Mitochondria are intracellular magnesium stores: investigation by simultaneous fluorescent imagings in PC12 cells. Biochim Biophys Acta Bba - Mol Cell Res. 2005;1744:19–28. doi: 10.1016/j.bbamcr.2004.10.013. [DOI] [PubMed] [Google Scholar]
  • 59.Wei X, Ugurlu O, Ankit A, et al. Dissolution behavior of Si,Zn-codoped tricalcium phosphates. Mater Sci Eng C. 2009;29:126–35. [Google Scholar]

RESOURCES