Vous êtes sur la page 1sur 11

Chemical Engineering Journal 222 (2013) 49–59

Contents lists available at SciVerse ScienceDirect

Chemical Engineering Journal


journal homepage: www.elsevier.com/locate/cej

Strontium substituted hydroxyapatite porous microspheres:


Surfactant-free hydrothermal synthesis, enhanced biological
response and sustained drug release
Kaili Lin a,1, Peiyi Liu b,1, Li Wei b, Zhaoyong Zou a, Weibin Zhang b, Ying Qian b, Yuhui Shen b,⇑, Jiang Chang a,⇑
a
State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, China
b
Department of Orthopaedics, Shanghai Institute of Orthopaedics & Traumatology, Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China

h i g h l i g h t s g r a p h i c a l a b s t r a c t

" The mesoporous strontium


substituted hydroxyapatite (SrHAp)
microspheres were fabricated.
" The ionic extracts of the
microspheres promoted the
biological responses.
" The architecture of SrHAp resulted in
favorable drug-loading and
sustained release properties.

a r t i c l e i n f o a b s t r a c t

Article history: In the absence of any surfactants, organic solvents or template-directing reagents, the strontium substi-
Received 5 December 2012 tuted hydroxyapatite (SrHAp) microspheres with hierarchically mesoporous structures and 1.17–
Received in revised form 6 February 2013 5.60 mol.% of Sr substitution were successfully fabricated via hydrothermal method. The morphology
Accepted 12 February 2013
observation showed that the fabricated SrHAp porous microspheres in diameters of 50–65 lm were
Available online 21 February 2013
assembled by two-dimensional single-crystal nano-sheets with 30–70 nm in thickness, and up to 3 lm
in width and length. The ionic extracts of SrHAp porous microspheres promoted the proliferation, oste-
Keywords:
ogenic differentiation and angiogenic factor expression of human osteoblast-like cells (MG-63), and the
Hydroxyapatite
Strontium
microspheres with 3.22 mol.% of Sr substitution showed the best potentially therapeutic applications in
Porous microspheres bone regeneration field. In addition, the novel 3D architectures of SrHAp resulted in favorable drug load-
Synthesis ing and sustained release properties. Our study indicated that the fabricated multifunctional SrHAp por-
Biological response ous microspheres might be a potential candidate as bioactive bone-regeneration and drug-delivery
Drug delivery and release material.
Ó 2013 Elsevier B.V. All rights reserved.

1. Introduction als [1]. Calcium phosphate (Ca–P) materials, such as hydroxyapa-


tite [Ca10(PO4)6(OH)2, HAp], b-tricalcium phosphate [b-Ca3(PO4)2,
The material with initiative stimulation capacity in tissue b-TCP] and calcium phosphate cement (CPC), are widely used in
regeneration is the major character for next generation biomateri- biomedical fields due to their good biocompatibility, osteo-con-
ductivity and similarity to the inorganic component of bone tissues
[2]. However, the traditional Ca–P based materials, including the
⇑ Corresponding authors. Tel.: +86 21 52412810; fax: +86 21 52413903. HAp, are lack of the ability to stimulate the formation of new bone,
E-mail addresses: yuhuiss@163.com (Y. Shen), jchang@mail.sic.ac.cn (J. Chang). which hinder their clinical applications [3]. Several method are
1
These authors contributed equally to this article.

1385-8947/$ - see front matter Ó 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.cej.2013.02.037
50 K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59

applied to solve this problem, such as loading bioactive growth fac- and Sr5-HAp, respectively. In the synthesis process, the aqueous
tors [4], grain size and surface morphology/topography design [5], solutions containing 0.05 mol (Ca2+ + Sr2+), 0.03 mol HPO24 and
and the incorporation of the functional trace elements [6,7]. In 0.15 mol urea were prepared by dissolving Ca(NO3)2, Sr(NO3)2,
which, the incorporation of the functional trace inorganic elements NH4H2PO4 and urea in 100 mL distilled water, and 0.1 mol/L
into the biomaterials to improve the biological responses has been HNO3 solution was used to adjust the pH to 2.41–2.45 to obtain
aroused great interest due to its simplicity and low cost. Recently, clear solutions. Then 17 mL of the obtained solution were trans-
strontium (Sr) ranelate, a newly developed drug treating osteopo- ferred into 20 mL Teflon autoclaves and heated at 120 °C for 1–
rosis, has been shown to have dual effects of stimulation osteoblast 72 h, followed by cooling to room temperature naturally. It is well
differentiation and inhibiting osteoclast activity and bone resorp- known that, with the increase of the hydrothermal temperatures,
tion, which could reduce the incidence of fractures in osteoporotic the (NH2)2CO continuously decomposes to form CO2 and aqueous
patients [8]. In addition, the partial substitution of Ca by Sr can ammonia species based on the following equation:
apparently improve the biological properties of Ca–P materials [9].
ðNH2 Þ2 CO þ H2 O ! 2NH3 þ CO2
Recently, the nano-structured HAp porous microspheres have
attracted great interests due to their high specific surface area In which, the released NH3 easily dissolves in water and in-
and novel three-dimensional (3D) hierarchical architectures, creases the pH value to alkaline condition in the reaction solution,
which make it possible to incorporate higher dosages of drugs into wherein HAp becomes the more thermodynamically stable cal-
the materials and release them at a control rate [6,10]. Further- cium orthophosphate compound, and is thus formed [22]. There-
more, the 3D architectured HAp microporous materials can be used fore, the continuous and homogeneous decomposition of urea
as injectable bone regeneration biomaterials and cell/drug loaded generates the driving force toward the nucleation and growth of
implants, which is superior to particles [11,12]. Thus, comparing HAp crystals under moderate supersaturation conditions [22]. In
2
with traditional particles, the Sr-substituted HAp (SrHAp) porous addition, the CO2 dissolves in water to form HCO 3 and CO3 , and
microspheres might possess better biological properties for guid- then incorporates as carbonate in the HAp lattice [23]. After hydro-
ing bone tissue regeneration, and have great potential application thermal reaction, the obtained suspension was filtrated and
for drug delivery. Up to now, the SrHAp porous microspheres with washed with distilled water and anhydrous ethanol for three
hierarchical nano-architectures are rarely reported. times, respectively. The obtained powders were dried at 120 °C
Furthermore, the traditional strategy to synthesize the 3D for 24 h.
architectured materials is focused on the organic solvent, surfac- To investigate the roles of the chemical composition (Sr-substi-
tant and chelating ligand assistant assembled approach [13–20]. tution) and porous morphologies on the biological performance
Ma and Zhu synthesized HAp hollow microspheres via solvother- and drug loading/release properties, respectively, the pure HAp
mal method using N,N-dimethylformamide (DMF) as the solvent nanoparticles labeled as Sr0-HAp were prepared as the control
[16]. Zhang et al. fabricated HAp microspheres and microflowers sample. The pure HAp nanoparticles can well play dual roles as
via hydrothermal method using hexadecyltrimethylammonium chemical component control for biological response, and as mor-
bromide (CTAB) as the surfactant [17]. He et al. reported that in phology control for drug loading/release properties. First, 0.5 M
the hexane–water–bis(2-ethylhexyl) sulfosuccinate (AOT) system Ca(NO3)2 solution and 0.3 M (NH4)2HPO4 solution were obtained
hollow HAp microspheres were presented [18]. The HAp materials by dissolving Ca(NO3)2 and (NH4)2HPO4 in distilled water, respec-
with flower-like morphology assembled from nanosheets consist- tively, and the pH of both solutions was adjusted to 10.5 using
ing of nanorod building blocks was synthesized by Ma using potas- ammonia solution. The reactant molar ratio of Ca/P was kept at
sium sodium tartrate as chelating ligand and template molecule 1.67. The solution of (NH4)2HPO4 was added dropwise into the
[19]. Veljovic et al. prepared the HAp hollow microspheres assem- Ca(NO3)2 solution to obtain a suspension, and the pH of the sus-
bled by rod-shaped nanoparticles using Na2EDTA as chelating li- pension was maintained at 10.5 using ammonia solution. Then
gand and template molecule [20]. However, in most of these the suspension was transferred into autoclaves and heated at
cases, large amounts of template and/or organic solvents were 120 °C for 72 h, followed by cooling to room temperature naturally.
widely used, which might be hazardous to health and environ- After the hydrothermal reaction, the obtained suspensions were
ment. Up to now, self-assembly of nano-units into the 3D architec- filtrated and washed with distilled water and anhydrous ethanol
tures in the absence of any surfactants, template-supports or for three times, respectively. The obtained powders were dried at
structure-directing reagents are still a major challenge [21]. Re- 120 °C for 24 h.
cently, Neira et al. developed a repeated stepwise hydrothermal The obtained powders were characterized by X-ray diffraction
method to synthesize micrometer-sized HAp particles with a sharp (XRD: D/max 2550 V, Rigaku, Japan) with mono-chromated Cu
faceted hexagonal prism-like morphology [22]. Ka radiation and Fourier transform infrared spectroscopy (FTIR:
Herein, the SrHAp porous microspheres with 1.17–5.60 mol.% of Nicolet Co., USA). The lattice constants were calculated from the
Sr substitution was synthesized via hydrothermal method. Then well determined positions of the intense XRD diffractions that
the effect of Sr substitution on osteoblast cell (MG-63) prolifera- were processed by MDI Jade 6.1 software [24]. To evaluate the
tion, osteogenic differentiation and angiogenic factor expression crystallinity degree, the reflection (2 1 1) of HAp powders at around
of the microspheres, and the effect of the 3D architectured nano- 2h = 31.8° (JCPDS No. 09-0432) was selected as the diffraction
structures on drug loading and release capacities were further plane for crystallinity measurement to calculate the relative index
investigated. of crystallinity (IOC, %) defined as follows [25]:
IOC ¼ ðIc =Io Þ  100%
2. Materials and methods where Ic and Io are the peak intensity of the selected diffraction
plane in the obtained products and the control sample, respectively.
2.1. Synthesis and characterization of SrHAp porous microspheres In present study, the fully crystallized HAp structure prepared by
wet chemical precipitation method and then calcined in furnace
The SrHAp porous microspheres with designed Sr2+/(Ca2+ + Sr2+) at 900 °C for 1.5 h was used as the control sample [26]. The mor-
molar ratios of 0.01, 0.03 and 0.05 were hydrothermally synthe- phology and size of the obtained powders were characterized by
sized using urea ((NH2)2CO) as homogeneous precipitation re- scanning electron microscopy (SEM: JSM-6700F, JEOL, Japan) and
agent. The obtained products were labeled as Sr1-HAp, Sr3-HAp transmission electron microscopy (TEM: JEM-2100F, JEOL, Japan).
K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59 51

The surface area and nitrogen sorption isotherm for the samples Table 1
were measured on a Micrometitics Tristar 3000 system. The chem- Primer sequences for real-time polymerase chain reaction (PCR).

ical compositions of the products were quantified by inductively Gene Primer sequences
coupled plasma – optical emission spectrometry (ICP-OES, 715-ES, ALP Forward: GGGAACGAGGTCACCTCCAT
Varian, USA). Reverse: TGGTCACAATGCCCACAGAT
Col-I Forward: CCTGCGTGTACCCCACTCA
2.2. Effect of ionic product from SrHAp porous microspheres on the Reverse: ACCAGACATGCCTCTTGTCCTT
Cbfa1 Forward: AGTGATTTAGGGCGCATTCCT
proliferation, osteogenic differentiation and angiogenic factor Reverse: GGAGGGCGGCGTGGGTTCT
expression of human osteoblast-like cells VEGF Forward: AGGAGGAGGGCAGAATCATCA
Reverse: CTCGATTGGATGGCAGTAGCT
The ionic extract method is widely used as the international
standard method to evaluate the effect of the chemical composi- keeping gene, human ACTIN (DCt = Ct gene of interest – Ct ACTIN).
tions on cell biological responses [27], which can effectively avoid Results are reported as relative gene expression (2DDCt). All exper-
the additional effects coming from the material morphologies by iments were done in triplicate to obtain the average data.
directly incubation the materials with cells. In present study, the
human osteoblast-like cells (MG-63 cell, Cell bank, Shanghai, Chi-
na) were used, and the cells were cultured in the Dulbecco’s mod- 2.3. In vitro study of drug loading and release properties
ified Eagle’s medium (DMEM, Invitrogen) containing 10% fetal
bovine serum (FBS) (Hyclone) and 1% penicillin–streptomycin 2.3.1. Release measurements
(Invitrogen). To prepare the extracts, a stock solution of 200 mg/ To investigate the influence of the porous structures on drug
mL was first prepared by immersing each test HAp powders in loading and release properties, the antibiotic vancomycin (New
DMEM culture medium. After incubation at 37 °C for 24 h, the mix- Drug Research and Development CO., Ltd., North China Pharmaceu-
ture was centrifuged and the supernatant was collected. Then a se- tical Group) and the HAp nanoparticles were used as the drug
rial diluted extracts with concentrations of 100, 50, 25 and model and the material control sample, respectively. Vancomycin
12.5 mg/mL were prepared by diluting from the stock solution of release measurements were carried out using UV/VIS spectroscopy
200 mg/mL with serum-free DMEM. Subsequently, these extracts at a wavelength of 280 nm. Calibration curve (correlation coeffi-
were sterilized by filtration through 0.22 lm filter membrane for cient R2 = 0.9999) was made for each set of measurements and
cell culture experiments. The ion concentrations of the extracts determined by taking absorbance vs. vancomycin concentration
were measured by ICP-OES. The proliferation of MG-63 was carried between 0 and 1.0 mg/mL as parameters. Three samples from each
out using Cell Counting Kit-8 (CCK-8) assay [28]. group were measured to obtain the average data of the drug load-
The MG-63 cells was seeded in 96-well tissue culture plates at a ing and release amount.
density of 5  103 cells/well for 1 and 3 days of culture by incuba-
tion at 37 °C for 24 h with 5% CO2, 95% air at 100% RH. The medium
in the well was then replaced by the prepared extracts. After cell 2.3.2. Drug loading procedures
culture, removed the supernatant then added 100 lL DMEM plus Typically, 0.25 g of SrHAp products were added into 1.0 mL of
10 lL CCK-8 into each well. After additional incubation for 1 h aqueous solution with a vancomycin concentration of 10 mg/mL
and then 20 s of slow shaking, the absorbance was read at at 37 °C, and soaked for 24 h under stirring in a vial that was sealed
450 nm against the reference value at 600 nm (BIO-RAD Model to prevent the evaporation of aqueous solution. The vancomycin-
680, USA), and the results were expressed as optical density loaded SrHAp samples were separated by centrifugation, and then
(OD). All experiments were done in triplicate to obtain the average dried in vacuum at 37 °C for 24 h. The amount of loaded vancomy-
data. cin was measured by the depletion method, by determining the
Based on the proliferation results, the optimal extract concen- difference in vancomycin concentration in the loading medium be-
tration of the SrHAp sample with strongest stimulation property fore and after loading. The amount of drug loading was calculated
was selected to further examine the effect of the Sr-substitution according to the following formula:
on osteogenic differentiation and angiogenic factor expression of
the osteoblasts using real-time polymerase chain reaction (PCR) Amount of drug loaded ¼ ðA  BÞ=A  100%
analysis. The extract of the pure HAp nanoparticles with same con-
centration was used as control. After culture MG-63 in 96-well tis- where A and B represented the initial and final drug concentrations,
sue culture plates at a density of 1  105 cells/well for 5, 7 and respectively.
10 days, the total RNA was isolated and then the cDNA was synthe-
sized using a Prime-ScriptTM RT reagent kit (Takara Bio, Shiga, Ja-
pan) according to manufacturer’s instructions. Highly purified gene 2.3.3. In vitro drug release
specific primers (Table 1) for alkaline phosphatase (ALP), collagen 0.25 g of drug-loaded SrHAp samples were placed into 1.0 mL of
type 1 (Col I), core-binding factor subunit alpha-1 (Cbfa1), vascular phosphate buffer solution (PBS) at pH 7.4 and agitated in a hori-
endothelial growth factor (VEGF) and the house-keeping gene, AC- zontally shaking water bath at 37 °C. At the predetermined time
TIN, were synthesized commercially (Shengong, Co., Ltd. Shanghai, intervals, the solutions were centrifuged, and then the release
China). Quantification of all cDNA of bone marker genes was tested medium was withdrawn and replaced with fresh release medium
using Applied Biosystems-7500 real-time PCR system. For quanti- (0.50 mL). The amount of the drug release was determined by
tative PCR, 10 lL SYBR Premix Ex Taq™ 6 lL ddH2O, 0.8 lL of each UV/VIS spectroscopy.
forward and reverse primer, 0.4 lL Rox and 2.0 lL cDNA template
were used in a final reaction volume of 20 lL. The thermal profile
for all reactions was as follows: 30 s at 95 °C, followed by 40 cycles 2.4. Statistical analysis
of 5 s at 95 °C, 34 s at 60 °C and 30 s at 72 °C. Data collection was
enabled at 72 °C in each cycle. CT (threshold cycle) values were cal- Data were analyzed for statistical significance using an analysis
culated using the Applied Biosystems 7500 software v 2.0.5 sys- of variance. Differences at p values of <0.05 were considered
tem. Each mRNA value was normalized to that of the house- significant.
52 K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59

3. Results and discussion only a small amount of the product was obtained. As shown in
Fig. 4A, the morphology of the obtained product in the early stage
3.1. Characterization of SrHAp porous microspheres was small sheetlike shape with sizes of about 0.1–0.5 lm, and was
free of aggregation. Then the crystal growth followed, and bigger
At the end of the hydrothermal process, the pH value of the particles grew at the expense of smaller crystals, as described by
reaction solutions reached 7.46–7.48 which was favorable for the the Gibbs–Thomson equation [30]. With the increase of the hydro-
nucleation and growth of HAp crystals. Fig. 1 shows the morphol- thermal time to 3 h, the obtained powders grew wider and longer
ogy images of the control sample and the synthetic SrHAp products than those in the early stage at the expense of the small crystal-
via hydrothermal treatment at 120 °C for 72 h. It is clear that the lites. The small sheets almost completely disappeared. Due to the
obtained SrHAp products were in 3D architectured porous micro- high surface energy of the nano-crystals, the as-formed nano-
spheres with similar morphology. The diameters of the porous sheets aggregated (Fig. 4B) to reduce their surface energy in aque-
microspheres were between 50 and 65 lm and the porous micro- ous solution. The optimum shape for minimizing the surface free
spheres consisted of 2D nano-sheets. The high magnification energy is a spherical structure due to the lowest surface area of
images (Fig. 1B, D and F) showed that the nano-sheet had smooth the spheres [31], which favors the self-assembling of the 2D
surface with a thickness of about 30–70 nm, and width and length nano-sheets into microspheres rather than other shape of aggrega-
of up to 3 lm. As expected, the control sample was mono-disper- tions. Further increase of the reaction time to 6 h, these sheets
sive in the absence of aggregations and had smooth surface and assembled as ‘‘petals’’ into 3D structures (Fig. 4C). The morphology
rod-like shape, 25–30 nm in diameter and 25–100 nm in length of the final product is shown in Fig. 4D. It was clear that much
(Fig. 1G). In general, the simply hydrothermal treatment of the more sheets were assembled into the clusters, and the typical 3D
precipitated HAp particles in aqueous solution without using any architecture of porous microspheres formed. The degree of assem-
template-directing reagents is usually dispersive rod-like nanopar- bly increased with the increase of hydrothermal time. From
ticles because of the preferred orientation growth of HAp crystal Fig. 4A–D, one can clearly see the process of crystalline nucleation
along the c-axis [2]. and growth, and self-assembling growth from nano-sheets to 3D
Fig. 2A presents the XRD patterns of the control sample and the microsphere morphology. Based on the results of time-dependent
synthetic SrHAp porous microspheres. It is clear that all of the prod- experiments and the morphology observations (Fig. 4), the process
ucts were identified as pure HAp phase (JCPDS card: No. 09-0432). of the morphology evolution of HAp porous microspheres is sum-
The crystallinity of the hydrothermally synthetic HAp products marized in Fig. 5. The similar phenomenon of nucleation–dissolu-
was about 69–86% calculated by the area-integration method from tion–recrystallization–self-assembly growth process has been
XRD patterns [25,26], indicating the relatively high crystallinity of widely observed in the synthesis of metal oxide and hydroxide,
the products. The small angle XRD scanning results (Fig. 2B) clearly sulfide and other kinds of materials [32–35]. However, further
confirmed that the corresponding peaks of the synthetic SrHAp por- work needs to be done to completely illuminate the self-assemble
ous microspheres significantly shifted to lower degree when com- mechanism for a better understand the role of all parameters in
pared with the pure HAp materials, indicating an increase of the structure regulation, such as the ion concentration, pH value,
lattice constants [29]. The shifting degree increased with the in- hydrothermal temperature and period.
crease of the Sr substitution amount. The lattice constants were fur- The crystalline structure of the apatite porous microspheres
ther calculated based on the XRD characterization and the results was further confirmed by selected-area electron diffraction (SAED)
were listed in Table 2. It is clear that most of the lattice constants and high-resolution transmission electron microscopy (HRTEM)
of the synthetic SrHAp products were larger than that of the pure investigations. Fig. 6 showed the representative SAED (Fig. 6B)
HAp. The increasing of the lattice constants were due to the replace- and HRTEM image (Fig. 6C) taken from the single nano-sheet
ment of the Ca ions by bigger atomic diameter of Sr [29]. In present (Fig. 6A) which was sonicated from the microspheres. The discrete
study, the deviation of lattice constants suggested that Sr2+ replaced SAED spots (Fig. 6B) revealed that the final product was well-crys-
and occupied the Ca2+ crystal sites of the HAp. tallized hexagonal single crystal. A typical HRTEM image of the sin-
Fig. 3 presents the FITR spectra of the control sample and the gle nano-sheet in Fig. 6C apparently showed the lattice fringes,
synthetic SrHAp porous microspheres. The broad and strong bimo- with interplanar spacings of 0.46 and 0.34 nm, corresponding to
dal peaks at 1458 and 1418 cm1, and the peak at 872 cm1 were the (1 1 0) and (0 0 2) planes of HAp, respectively. The HRTEM im-
attributed to carbonate ions (CO2 3 ) in B-site [21]. The peaks at 475
age further confirmed that the nano-sheet as the assembled com-
(m2), 559 (m4), 602 (m4), 958 (m1), 1032 (m3) and 1106 (m3) cm1 were ponent for the microspheres was single crystal.
the characteristic bands for PO3 4 , and the peaks at 3448 and
The elemental analyses of the synthetic HAp powders were pre-
1636 cm1 were assigned to the bending mode of the adsorbed sented in Table 2. The results showed that the (Ca + Sr)/P molar ra-
water. In addition, the characteristic OH bands of HAp at around tio of the fabricated SrHAp porous microspheres was similar to that
3571 cm1 (mOH) and 633 cm1 (dOH) were not clearly visible from of pure HAp (Ca/P = 1.67). In addition, the Sr substitution level
the synthetic SrHAp porous microspheres because of the presence could be facilely tailored by regulation the initial molar ratios of
of carbonate ions [21]. Carbonate substitution induces crystal lat- Sr2+/(Ca2+ + Sr2+).
tice aberrance, and the harmonic oscillator unit of the equilateral Moreover, the aggregates of the nanocrystals in the synthetic
triangle 3Ca(II)–OH is disordered, which means that the resonance SrHAp porous microspheres led to higher specific surface area
effect of harmonic vibration in OH channels is disturbed and the (SBET) and formation of mesopores, which were confirmed by the
higher the content of carbonate is, the more this disturbance vari- nitrogen adsorption–desorption isotherm and the density func-
able is. Therefore, the structured OH peaks will disappear in case of tional theory (DFT) pore size distribution curves (Table 3 and
high content of carbonate [21]. Fig. 7). The SBET of the SrHAp porous microspheres was about
To investigate the formation mechanism of the microspheres, 2–2.6 times higher than that of the control HAp nanoparticles
the morphology of the products (using the precursors of Sr3- (Table 3). According to the International Union of Pure and Applied
HAp) obtained at different hydrothermal time points such as 1, 3, Chemistry, the data shown in Fig. 7 suggested that the fabricated
6 and 72 h (final product) at reaction temperature of 120 °C were microspheres exhibit a type H4 hysteresis loop deriving from
characterized by SEM (Fig. 4). After 1 h of hydrothermal treatment, particle aggregates with slit-shaped pores in the range of 0.45–
K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59 53

Fig. 1. The morphologies of the synthetic SrHAp porous microspheres: Sr1-HAp (A and B), Sr3-HAp (C and D) and Sr5-HAp (E and F), and the control Sr0-HAp nanoparticles
(G).
54 K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59

the values in present study. It is well known that the amount of Sr


in bone tissue is much higher than that in blood. Furthermore,
99.1% of the absorbed Sr is deposited in bone (36–140 mg/kg)
[37,41]. Therefore, it can be indicated that a certain high dose of
Sr may stimulate the proliferation of osteoprogenitor cells and
benefit new bone regeneration.
Base on the cell proliferation results, the extract concentration
of 100 mg/mL from Sr3-HAp porous microspheres was further
selected to examine the osteogenic differentiation and angiogenic
factor expression of the Sr substituted HAp materials, and the
Sr0-HAp was used as the control sample (Fig. 9). The real-time
PCR analysis results showed that the expression levels of osteo-
blastic markers, such as ALP, Col I and Cbfa1, and the VEGF were
Fig. 2. XRD patterns of the control HAp sample and the synthetic SrHAp porous markedly higher for Sr3-HAp porous microspheres than those for
microspheres. the control Sr0-HAp material at all the culture time points, except
for cbfa1 on day 5. For the Sr3-HAp extract, the expression level of
ALP decreased slightly from day 5 to day 7, and then maintained
1.0 P/P0, which was typical of a mesoporous structure [36]. Calcu- with the further increase of the culture time to day 10, while those
lated from the adsorption branches of the isotherms using the DFT of Col I, Cbfa1 and VEGF increased sustainably. As for the control
method, the pore size is distributed from meso- to macro-scale in a Sr0-HAp extract, the expression levels of ALP, Col I, Cbfa1 and VEGF
range of about 2–100 nm (Fig. 7). reached the peak at day 7, and then decreased apparently.
Especially, the Cbfa1 known as an osteoblast-specific transcription
factor was remarkably upregulated by SrHAp materials, which
3.2. Effect of ionic product from SrHAp porous microspheres on MG-63 plays an important role as an upstream regulator of osteoprogeni-
proliferation, osteogenic differentiation and angiogenic factor tor differentiation [42]. Traditionally, the osteogenic differentiation
expression of osteoblast cell lines is initiated by the addition of dexametha-
sone, b-glycerophosphate and ascorbic acid, etc. [43]. The CCK-8
The CCK-8 evaluation results revealed no appreciable toxicity and PCR analysis showed that the Sr substitution can stimulate
when the human osteoblast-like cells (MG-63) were subjected to the proliferation and osteogenic differentiation of MG-63 cells.
control HAp nanoparticles and the SrHAp porous microspheres in Furthermore, the angiogenic factor of VEGF can also be upregu-
the extract concentration range of 200–12.5 mg/mL (Fig. 8). All lated by the Sr incorporation. The angiogenesis is extraordinarily
extracts of SrHAp porous microspheres exhibited none cytotoxic- important during bone regeneration [44]. Especially for bone
ity. The cell proliferation was evident for all samples over the defects with critical size in centimeter size, the difficulty of
duration of the culture time. A stimulatory effect of the ionic inducing rapid vascular ingrowths is harmful for bone regenera-
product from Sr3-HAp porous microspheres on MG-63 prolifera- tion, which limits the diffusion of oxygen and nutrients from the
tion was extraordinarily higher than other materials at all the surrounding tissue to ensure the viability and function of cells
extract concentration range of 200–12.5 mg/mL in culture periods. [45]. Up to now, the VEGF has been widely used as growth factor
In addition, the extract concentration of 100 mg/mL both for to stimulate angiogenesis in tissue engineered constructs [12,46].
Sr1-HAp and Sr5-HAp also favors to stimulate MG-63 proliferation. However, due to the complex of in vivo environment and short
The results suggested that the component of Sr3-HAp porous half-life of proteins, the release behavior of proteins in vivo cannot
microspheres might be the best potentially therapeutic applica- be easily controlled, which may also result in possible toxicity ef-
tions for bone regeneration. Previous studies have confirmed that fects [47]. Furthermore, the cost of the growth factors is extraordi-
the Sr2+ ion plays the most important role in biological perfor- narily high. All of these concerns limit the further application of
mances in the extracts of Sr-containing bioceramics [8,9]. The growth factors in clinical fields. Up to now, little work has been
ICP-OES analysis showed that the Sr2+ ion concentrations of the done on the influences of Sr on angiogenesis. Herein, our primarily
Sr3-HAp extracts (200–12.5 mg/mL) for cell culture was 0.1342– study suggested that the low cost incorporation of inorganic Sr ion
0.0084 mmol/L. Furthermore, the effects of Sr on bone cells were into HAp bioceramics could induce higher angiogenic expression
dose-dependent [37]. Canalis et al. found that the divalent stron- level, suggesting that the substitution of the functional Sr element
tium salt S12911 at 103 mmol/L increased the replication of pre- may be extremely useful in designing osteogenic and angiogenic
osteoblastic cells by 30–50% after 24 h and by 60% after 96 h of bioactive materials for bone regeneration and bone tissue engi-
treatment [38]. Bonnelye et al. revealed that strontium ranelate neering. The enhancement of Sr component on angiogenic expres-
with Sr2+ between 0.1 and 1 mmol/L could stimulate osteoblast sion was rarely reported up to now. However, the mechanism of
formation [39]. It has been reported that the concentration of Sr promoted angiogenic behaviors initiated by Sr component remains
in normal serum varies between 10 lg/L (1.14  104mmol/L) unknown, which needs to be further investigated in details.
and 217 lg/L (2.48  103 mmol/L) [40], which is much lower than

Table 2
Crystallinity, lattice constants, 2h value for (2 1 1) diffraction and chemical composition of the synthetic HAp products.

Samples Crystallinity (%) Lattice constantsa 2h (°) for (2 1 1) reflectiona Chemical composition
a (Å) c (Å) Ca replacement by Sr (mol.%) (Ca + Sr)/P molar ratio
Sr0-HAp 69 9.496 6.93 31.52 0 1.66
Sr1-HAp 86.5 9.496 6.954 31.5 1.17 1.64
Sr3-HAp 73.2 9.507 6.916 31.48 3.22 1.66
Sr5-HAp 82.4 9.507 6.989 31.44 5.6 1.67
a
Hydroxyapatite ICDD PDF # 09-0432: a = 9.418 Å, c = 6.884 Å, 2h for (2 1 1) diffraction is 31.86°.
K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59 55

Fig. 5. Schematic illustration of the formation and morphology evolution of 3D


architectured HAp microspheres in the synthetic process.

specific surface areas, mesopores and interconnected macropores


Fig. 3. The FTIR spectra of the control sample and the synthetic SrHAp porous
(Table 3 and Fig. 7). Therefore, the synthetic SrHAp microspheres
microspheres. with hierarchically mesoporous structures have great potential
applications for drug delivery system. Herein, the vancomycin
model was used to investigate the capability of the synthetic SrHAp
3.3. In vitro study of drug-loading and release properties microspheres as drug carriers, and the results are listed in Table 3.
The drug-loading amount (DLA: mloaded drug/mcarrier, mg/g) and
Comparing with traditional nano-particles, the SrHAp micro- drug-loading efficiency (DLE: mloaded drug/mtotal drug amount, %) were
spheres with hierarchically mesoporous structures possess larger apparently higher than that of the traditional HAp nanoparticles.

Fig. 4. SEM images of the obtained products after hydrothermal treatment at 120 °C for 1 h (A), 3 h (B), 6 h (C) and 72 h (D).
56 K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59

Fig. 8. The effect of ionic product from control Sr0-HAp and SrHAp porous
Fig. 6. A single nano-sheet (A) was selected for undertaking intensive character- microspheres on proliferation of MG-63 after culture for 1 and 3d. The experi-
ization by SAED (B) and HRTEM (C). mental group compared with the control group of Sr0-HAp nanoparticles at the
same concentration and culture time, p < 0.05.

Table 3
The specific surface area (SBET), drug-loading amount (DLA) and drug-loading molecules and HAp crystals [6]. The higher specific surface area
efficiency (DLE) of the nanoparticles (controlling sample Sr0-HAp) and the synthetic (SBET) of the SrHAp porous microspheres possess higher amount
SrHAp porous microspheres. of OH groups, which will improve the interactions between adsor-
Samples SBET (m2/g) DLA (mg/g) DLE (%) bents and vancomycin molecules, and ultimately increase the DLA
(Table 3).
Sr0-HAp 11.17 19.08 ± 2.70 47.70 ± 6.75
Sr1-HAp 22.77 34.00 ± 0.42 84.99 ± 1.05 It is well known that the physical interaction of hydrogen bond-
Sr3-HAp 20.81 33.56 ± 0.22 83.90 ± 0.54 ing is relatively weak comparing with the chemical bonding. After
Sr5-HAp 29.71 33.36 ± 1.33 83.41 ± 3.32 soaking the drug-loaded SrHAp in aqueous solution, the loaded
drug will be released into the medium due to the dissociation of
hydrogen bonding [48]. Fig. 10 shows the cumulative release re-
The DLA of the SrHAp porous microspheres reached 33.36– sults of vancomycin from the SrHAp porous microspheres and con-
34.00 mg/g, which was about 74.8–78.2% higher than that of the trol HAp nanoparticles in PBS. It is clear that the vancomycin
HAp nanoparticles. The high DLA and DLE were attributed to the no- showed similar release behavior during the whole period, which
vel 3D hierarchically nanostructures with mesoporous structures. had obvious two-step release behavior, an initial fast release and
The mesoporous structures in the SrHAp microspheres increase a relatively slow subsequent release. However, the drug release
the specific surface area (SBET), which is a vital factor for high rate of HAp nanoparticles was significantly faster than that of
DLA. Simultaneity, the mesopores among the microspheres not only SrHAp porous microspheres. For HAp nanoparticles, the drug re-
decreased the diffusion limitations for loading drug molecules, but lease rate was remarkably higher than that of SrHAp porous micro-
also provided space for attracting the vancomycin. In addition, the spheres, around 9 wt.% of total drug loading in the first 3 h. After
DLA is greatly related to the intrinsic nature of carrier materials. 5 days, the amount of released vancomycin reached 24.28 wt.%.
Vancomycin can adsorb on HAp through the hydrogen bonding While for SrHAp porous microspheres, an initial burst release in
interactions between the OH group, which both existed in the drug the first 3 h was around 6 wt.% in PBS of total amount of vancomy-
cin. The subsequent release rate remarkably decreased with time,
and the cumulative release crept during the next 5 days, reaching
a maximum value of 16.07–17.68 wt.% in PBS. The lower release
rate was due to the novel 3D hierarchically nanostructures with
mesoporous structures. In clinical field, the minimum inhibitory
concentration, the minimum bactericidal concentration and the
breakpoint sensitivity of vancomycin for Staphylococcus aureus
were 1.01, 1.98 and 5 lg/mL, respectively [49]. The vancomycin
concentrations released from SrHAp porous microspheres during
the research period was around 280–580 lg/mL (Fig. 11), which
was significantly higher than that of the breakpoint sensitivity of
vancomycin and the published data [49–51]. Therefore, it can be
believed that the released concentration of the objective drug is
high enough for the local therapeutic need. However, the in vivo
study needs to be done to further confirm the therapeutics of
SrHAp porous microspheres.
Our results show the most promising applications of the fabri-
cated SrHAp porous microspheres in the fields of drug delivery sys-
tem at a sustained release rate [6,10], injectable bone regeneration
biomaterials and cell/drug loaded implants due to the higher spe-
Fig. 7. The DFT pore size distribution of the synthetic SrHAp porous microspheres. cific surface area and novel 3D hierarchical architectures, which is
K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59 57

Fig. 9. Real-time PCR analysis of the expression levels of ALP (A), Col I (B), Cbfa1 (C) and VEGF (D) mRNA after culture of MG-63 cells in the extract concentration of 100 mg/
mL from control Sr0-HAp nanoparticles and Sr3-HAp porous microspheres, p < 0.05.

Fig. 11. The vancomycin concentrations released from HAp nanoparticles and
Fig. 10. The cumulative vancomycin release ratio from HAp nanoparticles and
SrHAp porous microspheres in PBS.
SrHAp porous microspheres in PBS.

superior to particles [11,12]. In addition, the SrHAp porous micro-


spheres might be used as the component to fabricate the compos-
directing reagents. Comparing with the traditional HAp nanoparti-
ite biomaterials because of the excellent biological performances.
cles, the ionic product of SrHAp porous microspheres could appar-
ently stimulate the proliferation and osteogenic differentiation of
4. Conclusions MG-63 at certain concentrations of Sr2+ ion. In which, 3 mol.% Sr
dosage was optimal for bone regeneration. Moreover, the interest-
Herein, the Sr-substituted mesoporous HAp microspheres ing phenomenon of the incorporation of Sr into HAp bioceramics
(SrHAp) were successfully synthesized via hydrothermal method leading to the promoting of angiogenic behaviors was observed.
in the absence of any surfactants, organic solvents or template- Therefore, SrHAp might be favorable in vivo angiogenesis during
58 K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59

bone repair, and can be used as a promising scaffold in bone regen- [19] M.G. Ma, Hierarchically nanostructured hydroxyapatite: hydrothermal
synthesis, morphology control, growth mechanism, and biological activity,
eration field. In addition, the synthetic SrHAp porous microspheres
Int. J. Nanomed. 7 (2012) 1781–1791.
exhibited higher drug-loading capacity and sustained drug release [20] D. Veljovic, E. Palcevskis, A. Dindune, S. Putic, I. Balac, R. Petrovic, D.
properties due to their novel 3D hierarchical mesoporous struc- Janackovic, Microwave sintering improves the mechanical properties of
tures, high specific surface area, and hydrogen bonding interac- biphasic calcium phosphates from hydroxyapatite microspheres produced
from hydrothermal processing, J. Mater. Sci. 45 (2010) 3175–3183.
tions between the carriers and drugs. The present study suggests [21] K.L. Lin, J. Chang, Y.J. Zhu, W. Wu, G.F. Cheng, Y. Zeng, M.L. Ruan, A facile one-
that the fabricated SrHAp porous microspheres might be greatly step surfactant-free and low-temperature hydrothermal method to prepare
suitable for applications as new bioactive materials in bone regen- uniform 3D structured carbonated apatite flowers, Cryst. Growth Des. 9 (2009)
177–181.
eration and drug-delivery applications. [22] I.S. Neira, Y. Kolen’ko, O.I. Lebedev, G.V. Tendeloo, H.S. Gupta, F. Guitian, M.
Yoshimura, An effective morphology control of hydroxyapatite crystals via
hydrothermal synthesis, Cryst. Growth Des. 9 (2009) 466–474.
Acknowledgements [23] S.P. Parthiban, I.Y. Kim, K. Kikuta, C. Ohtsuki, Effect of urea on formation of
hydroxyapatite through double-step hydrothermal processing, Mater. Sci. Eng.
The authors gratefully acknowledge the support of Natural Sci- 31C (2011) 1383–1388.
[24] S. Koutsopoulos, Synthesis and characterization of hydroxyapatite crystals: a
ence Foundation of China (Nos.: 81171458, 81150007, 81190132, review study on the analytical methods, J. Biomed. Mater. Res. 62 (2002) 600–
81170655), Funds of the Shanghai Institute of Ceramics, Chinese 612.
Academy of Sciences for Innovation of Science and Technology [25] C.Y. Yang, B.C. Wang, E. Chang, J.D. Wu, The influences of plasma spraying
parameters on the characteristics of hydroxyapatite coatings: a quantitative
(No.: Y26ZC1110G), and Shanghai Municipal Education Commis-
study, J. Mater. Sci.: Mater. Med. 6 (1995) 249–257.
sion (No.: 12YZ039). [26] H. Li, K.A. Khor, P. Cheang, Titanium dioxide reinforced hydroxyapatite
coatings deposited by high velocity oxy-fuel (HVOF) spray, Biomaterials 23
(2002) 85–91.
References [27] Biological evaluation of medical devices – Part 1. Evaluation and testing within
a risk management process, ISO 10993-1, 2009.
[1] L.L. Hench, J.M. Polak, Third-generation biomedical materials, Science 295 [28] L.Z. Zhao, L. Liu, Z.F. Wu, Y.M. Zhang, P.K. Chu, Effects of micropitted/
(2002) 1014–1017. nanotubular titania topographies on bone mesenchymal stem cell osteogenic
[2] J.C. Elliott, Structure and chemistry of the apatites and other calcium differentiation, Biomaterials 33 (2012) 2629–2641.
orthophosphates, Elsevier, Amsterdam, 1994. [29] Z.Y. Li, W.M. Lam, C. Yang, B. Xu, G.X. Ni, S.A. Abbah, K.M.C. Cheung, K.D.K. Luk,
[3] H.P. Yuan, H. Fernandes, P. Habibovic, J. De Boer, A.M.C. Barradas, A. De Ruiter, W.W. Lu, Chemical composition, crystal size and lattice structural changes
W.R. Walsh, C.A. Van Blitterswijk, J.D. De Bruijn, Osteoinductive ceramics as a after incorporation of strontium into biomimetic apatite, Biomaterials 28
synthetic alternative to autologous bone grafting, Proc. Natl. Acad. Sci. USA 107 (2007) 1452–1460.
(2010) 13614–13619. [30] R. Viswanatha, D.D. Sarma, Growth of Nanocrystals in Solution, in: C.N.R. Rao,
[4] N. Saranya, S. Saravanan, A. Moorthi, B. Ramyakrishna, N. Selvamurugan, A. Muller, A.K. Cheetham (Eds.), Nanomaterials Chemistry, WILEY-VCH Verlag
Enhanced osteoblast adhesion on polymeric nano-scaffolds for bone tissue GmbH & Co, KGaA, Weinheim, 2007.
engineering, J. Biomed. Nanotechnol. 7 (2011) 238–244. [31] D. Vanmaekelbergh, Self-assembly of colloidal nanocrystals as route to novel
[5] K.L. Lin, L. Chen, J. Chang, Fabrication of dense hydroxyapatite nano- classes of nanostructured materials, Nano Today 6 (2011) 419–437.
bioceramics with enhanced mechanical properties via two-step sintering [32] J.S. Xu, Y.J. Zhu, C-Fe2O3 and Fe3O4 magnetic hierarchically nanostructured
process, Int. J. Appl. Ceram. Technol. 9 (2012) 479–485. hollow microspheres: preparation, formation mechanism, magnetic property,
[6] K.L. Lin, Y.L. Zhou, Y. Zhou, H.Y. Qu, F. Chen, Y.J. Zhu, J. Chang, Biomimetic and application in water treatment, J. Colloid Interface Sci. 385 (2012) 58–65.
hydroxyapatite porous microspheres with co-substituted essential trace [33] J. Zhang, S. Liu, J. Lin, H. Song, J. Luo, E.M. Elssfah, E. Ammar, Y. Huang, X. Ding,
elements: surfactant-free hydrothermal synthesis, enhanced degradation and J. Gao, S. Qi, C. Tang, Self-assembly of flowerlike AlOOH (boehmite) 3D
drug release, J. Mater. Chem. 21 (2011) 16558–16565. nanoarchitectures, J. Phys. Chem. 110B (2006) 14249–14252.
[7] K.L. Lin, J. Chang, X.G. Liu, L. Chen, Y.L. Zhou, Synthesis of the element- [34] B.X. Li, Y. Xie, Y. Xue, Controllable synthesis of CuS nanostrucutres from self-
substituted hydroxyapatite with controllable morphologies and chemical assembled precursors with biomolecule assistance, J. Phys. Chem. 111C (2007)
compositions using calcium silicate as precursor, CrystEngComm 13 (2011) 12181–12187.
4850–4855. [35] S. Thirumalairajan, K. Girija, I. Ganesh, D. Mangalaraj, C. Viswanathan, A.
[8] E. Landi, A. Tampieri, G. Celotti, S. Sprio, M. Sandri, G. Logroscino, Sr- Balamurugan, N. Ponpandian, Controlled synthesis of perovskite LaFeO3
substituted hydroxyapatites for osteoporotic bone replacement, Acta microsphere composed of nanoparticles via self-assembly process and their
Biomater. 3 (2007) 961–969. associated photocatalytic activity, Chem. Eng. J. 209 (2012) 420–428.
[9] C. Capuccini, P. Torricelli, F. Sima, E. Boanini, C. Ristoscu, B. Bracci, Strontium- [36] K.S.W. Sing, Reporting physisorption data for gas solid systems-with special
substituted hydroxyapatite coatings synthesized by pulsed-laser deposition: reference to the determination of surface-area and porosity, Pure Appl. Chem.
in vitro osteoblast and osteoclast response, Acta Biomater. 4 (2008) 1885– 54 (1982) 2201–2218.
1893. [37] M.L. Zhang, W.Y. Zhai, K.L. Lin, H.B. Pan, W. Lu, J. Chang, Synthesis, in vitro
[10] L. Wu, Y.D. Dou, K.L. Lin, W.Y. Zhai, W.G. Cui, J. Chang, Hierarchically structured hydroxyapatite forming ability, and cytocompatibility of strontium silicate
nanocrystalline hydroxyapatite assembled hollow fibers as a promising powders, J. Biomed. Mater. Res. 93B (2010) 252–257.
protein delivery system, Chem. Commun. 47 (2011) 11674–11676. [38] E. Canalis, M. Hott, P. Deloffre, Y. Tsouderos, P.J. Marie, The divalent strontium
[11] X.Q. Gong, S.L. Peng, W.J. Wen, P. Sheng, W.H. Li, Design and fabrication of salt S12911 enhances bone cell replication and bone formation in vitro, Bone
magnetically functionalized core/shell microspheres for smart drug delivery, 18 (1996) 517–523.
Adv. Funct. Mater. 19 (2009) 292–297. [39] E. Bonnelye, A. Chabadel, F. Saltel, P. Jurdic, Dual effect of strontium ranelate:
[12] H.J. Chung, T.G. Park, Surface engineered and drug releasing pre-fabricated stimulation of osteoblast differentiation and inhibition of osteoclast formation
scaffolds for tissue engineering, Adv. Drug Deliv. Rev. 59 (2007) 249–262. and resorption in vitro, Bone 42 (2008) 129–138.
[13] G. Wang, H. Wang, Z.Y. Ren, J.T. Bai, J.B. Bai, PVP-assisted assembly of [40] W.E. Cabrera, I. Schrooten, M.E. De Broe, P.C.D. Haese, Strontium and bone, J.
lanthanum carbonate hydroxide with hierarchical architectures and their Bone Miner. Res. 14 (1999) 661–668.
luminescence properties, Chem. Eng. J. 214 (2013) 386–393. [41] S.C. Skoryna, Metabolic aspects of the pharmacologic use of trace elements in
[14] D.H. Jiang, W.B. Hu, H.R. Wang, B. Shen, Y.D. Deng, Synthesis, formation human subjects with specific reference to stable strontium, Trace Subst.
mechanism and photocatalytic property of nanoplate-based copper sulfide Environ. Health 18 (1984) 3–20.
hierarchical hollow spheres, Chem. Eng. J. 189–190 (2012) 443–450. [42] C. Banerjee, L.R. McCabe, J.Y. Choi, Runt homology domain proteins in
[15] L.H. Ai, Y. Zeng, Hierarchical porous NiO architectures as highly recyclable osteoblast differentiation: AML3/CBFA1 is a major component of a bone-
adsorbents for effective removal of organic dye from aqueous solution, Chem. specific complex, J. Cell Biochem. 66 (1997) 1–8.
Eng. J. 215–216 (2013) 269–278. [43] S.J. Roberts, Y. Chen, M. Moesen, J. Schrooten, F.P. Luyten, Enhancement of
[16] M.G. Ma, J.F. Zhu, Solvothermal synthesis and characterization of osteogenic gene expression for the differentiation of human periosteal derived
hierarchically nanostructured hydroxyapatite hollow spheres, Eur. J. Inorg. cells, Stem Cell Res. 7 (2011) 137–144.
Chem. 36 (2009) 5522–5526. [44] S.K.L. Levengood, M.J. Poellmann, S.G. Clark, D.A. Ingram, M.C. Yoder, A.J.W.
[17] C.M. Zhang, J. Yang, Z.W. Quan, P.P. Yang, C.X. Li, Z.Y. Hou, J. Lin, Johnson, Human endothelial colony forming cells undergo vasculogenesis
Hydroxyapatite nano- and microcrystals with multiform morphologies: within biphasic calcium phosphate bone tissue engineering constructs, Acta
controllable synthesis and luminescence properties, Cryst. Growth Des. 9 Biomater. 7 (2011) 4222–4228.
(2009) 2725–2733. [45] E.C. Novosel, C. Kleinhans, P.J. Kluger, Vascularization is the key challenge in
[18] W.H. He, J.H. Tao, H.H. Pan, X.R. Xu, R.K. Tang, A size-controlled synthesis of tissue engineering, Adv. Drug Deliver. Rev. 63 (2011) 300–311.
hollow apatite nanospheres at water–oil interfaces, Chem. Lett. 39 (2010) [46] P. Tayalia, D.J. Mooney, Controlled growth factor delivery for tissue
674–675. engineering, Adv. Mater. 21 (2009) 3269–3285.
K. Lin et al. / Chemical Engineering Journal 222 (2013) 49–59 59

[47] A.A. Gorustovich, C. Perio, J.A. Roether, A.R. Boccaccini, Effect of bioactive [50] X. Liu, Z.P. Xie, C.Q. Zhang, H.B. Pan, M.N. Rahaman, X. Zhang, Q. Fu, W.H.
glasses on angiogenesis: a review of in vitro and in vivo evidences, Tissue. Eng. Huang, Bioactive borate glass scaffolds: in vitro and in vivo evaluation for use
16 (2010) 199–207. as a drug delivery system in the treatment of bone infection, J. Mater. Sci.:
[48] J.X. Gu, F. Xia, Y. Wu, X.Z. Qu, Z.Z. Yang, L. Jiang, Programmable delivery of Mater. Med. 21 (2010) 575–582.
hydrophilic drug using dually responsive hydrogel cages, J. Control. Release [51] B. Li, K.V. Brown, J.C. Wenke, S.A. Guelcher, Sustained release of vancomycin
117 (2007) 396–402. from polyurethane scaffolds inhibits infection of bone wounds in a rat femoral
[49] S.W.N. Ueng, L.J. Yuan, N. Lee, S.S. Lin, S.J. Liu, E.C. Chan, J.H. Weng, In vivo segmental defect model, J. Control. Release 145 (2010) 221–230.
study of hot compressing molded 50:50 poly (DL-lactide-co-glycolide)
antibiotic beads in rabbits, J. Orthop. Res. 10 (2002) 654–661.

Vous aimerez peut-être aussi