You are on page 1of 15

Materials Today Chemistry 29 (2023) 101410

Contents lists available at ScienceDirect

Materials Today Chemistry


journal homepage: www.journals.elsevier.com/materials-today-chemistry/

Enhancing osteoinduction and bone regeneration of biphasic calcium


phosphate scaffold thought modulating the balance between
pro-osteogenesis and anti-osteoclastogenesis by zinc doping
T. Lu a, b, X. Yuan a, b, L. Zhang a, b, F. He d, X. Wang c, J. Ye a, b, e, *
a
School of Materials Science and Engineering and Key Laboratory of Biomedical Materials of Ministry of Education, South China University of Technology,
Guangzhou 510641, PR China
b
National Engineering Research Center for Tissue Restoration and Reconstruction, Guangzhou 510006, PR China
c
Medical Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510080, China
d
School of Electromechanical Engineering, Guangdong University of Technology, Guangzhou 510006, PR China
e
Key Laboratory of Biomedical Engineering of Guangdong Province and Innovation Center for Tissue Restoration and Reconstruction, South China
University of Technology, Guangzhou 510641, PR China

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

Article history: Biphasic calcium phosphate (BCP) is a promising bone regeneration material with an adjustable
Received 23 November 2022 degradation rate, and ions doping is an effective method to improve its bone repair effect. However, few
Received in revised form studies have focused on the role of ions doping-mediated osteogenesis and osteoclastogenesis balance in
20 January 2023
bone regeneration. In order to improve the bone repair effect of BCP and emphasize the role of osteo-
Accepted 20 January 2023
genesis and osteoclastogenesis balance regulated by ions doping to promote bone regeneration. Here,
Available online 18 February 2023
zinc (Zn), an essential trace element with pro-osteogenesis and anti-osteoclastogenesis, was dopped into
BCP scaffolds, and its effects on the physicochemical properties, in vitro cytological responses, in vivo
Keywords:
Biphasic calcium phosphate
osteoinduction, and bone defect regeneration of BCP were systematically investigated. Results showed
Zinc doping that Zn was successfully introduced into the crystal lattice of BCP. The differentiation of both osteoblasts
Osteoclastogenesis and osteoclasts showed a doping content-dependent behavior. By increasing the doping content (0
Osteogenesis e5 mol.%), the anti-osteoclastogenesis effect of BCP gradually increased, while its pro-osteogenesis
performance first increased and then decreased. ThoughBCP with a doping content of 5 mol.% could
promote osteogenic differentiation of stem cells and reduced fibrous capsule encapsulation, it could not
enhance ectopic bone formation or bone regeneration due to its excessive anti-osteoclastogenesis. BCP
with a doping content of 2.5 mol.% remarkably enhanced the osteoinduction activity and accelerated new
bone formation, which were associated with its highest pro-osteogenesis and appropriate anti-
osteoclastogenesis activity. This study highlights on regulating the balance between pro-osteogenesis
and anti-osteoclastogenesis by Zn doping and paves the way for the development of a more osteoin-
ductive calcium phosphate-based material for bone regeneration.
© 2023 Elsevier Ltd. All rights reserved.

1. Introduction b-tricalcium phosphate (b-TCP) and hydroxyapatite (HA) are most


extensively studied. HA and b-TCP can not only be used as the
Bone defect is a common clinical disease, usually caused by matrix of bone repair materials, but can also be used alone or in
functional degeneration, tumor, and trauma, and brings great combination with nanomaterials to modify metal or other matrix
tribulation to patients. Calcium phosphate-based bone regenera- materials for preparing intelligent antibacterial or antitumor im-
tion biomaterials have attracted much attention due to their plants and endow the implants with good angiogenesis [3] or
excellent bioactivity and perfect osteoconductivity [1,2]. Thereinto, osteogenesis [4e6]. The mixture of b-TCP and HA named biphasic
calcium phosphate (BCP) with an adjustable degradation rate is
considered to be a promising bone regeneration material [7,8].
Furthermore, according to the literature, it is found that a BCP
* Corresponding author. ceramic possessed an apparent osteoinduction activity compared
E-mail address: jdye@scut.edu.cn (J. Ye).

https://doi.org/10.1016/j.mtchem.2023.101410
2468-5194/© 2023 Elsevier Ltd. All rights reserved.
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

to a pure HA ceramic and b-TCP ceramic [9]. In addition, it was also eagle's medium (H-DMEM) was offered by Gibco (USA). Fetal
found that osteoinductive materials possessed better bone defect bovine serum (FBS) was bought from Biological Industries (Israel).
repair effects than non-osteoinductive materials [10]. A cell counting kit-8 solution was purchased from Dojindo (Japan).
Bone defect healing is a temporally coordinated and highly The rabbit-anti-mouse iNOS primary antibody, rabbit-anti-mouse
complex biological process that requires a series of interactions CD163 primary antibody, Cy3-labeled anti-rabbit secondary anti-
among a range of different cell types [11]. When a bone is injured, body, and 488-labeled anti-rabbit secondary antibody were offered
damaged blood vessels lead to the formation of blood clots to by Abcam (UK).
provide hemostasis. The clotting is followed by an inflammatory
response. Next is the phase of fibrovascular healing, when 2.2. Preparation of BCP powder
mesenchymal stem cells (MSCs) are recruited into the temporary
callus. These recruited MSCs subsequently differentiate into oste- A BCP powder was synthesized by a chemical precipitation
oblasts and/or chondrocytes, which eventually form new bones method, as described in our previous work [14]. Briefly, Ca(N-
through the mechanism of entochondrostosis and/or intra- O3)2$4H2O and (NH4)2HPO4 were separately dissolved in deionized
membranous ossification. Finally, the newly forming bones are water to prepare solution A (1 mol/L) and B (0.6 mol/L), respec-
remodeled by the coordination between osteoclasts to resorb tively. Then, solution B was dropwise added to solution A by a
woven bones and osteoblasts to form mature lamellar bones. This peristaltic pump under stirring. The pH of the reaction solution was
remodeling stage eventually leads to bone regeneration. It would adjusted to 10.2 by NH4OH after finishing the addition. The reaction
be feasible to target any of these healing stages to enhance bone solution was stirred for 2 h and then aged for 24 h. After that, the
regeneration [12]. reaction product was washed, centrifuged, frozen, and lyophilized.
Zinc (Zn) is one of the essential trace elements, which could Then, the obtained powder was treated at 900  C for 2 h. For the
inhibit the activities of osteoclasts while stimulating the osteogenic preparation of the Zn-doped BCP(Zn-BCP) powder, the Ca(N-
differentiation of stem cells [13]. Zn doping has been used to O3)2$4H2O solution was exchanged by a mixed solution (1 mol/L)
improve the bone defect repair effects of calcium phosphate ce- composed of Zn(NO3)2$6H2O and Ca(NO3)2$4H2O. The molar ratio
ramics [14e16]. However, previous research mainly focused on of (Ca þ Zn) to P was 1.67 in the initial reaction solution. The initial
promoting the osteogenic differentiation of MSCs [14,15] or inhib- doping content of Zn was in the range of 0e6 mol.%. Zn-BCP
iting the formation of osteoclasts as well as their bone resorption powders were denoted as ZnX (X ¼ Zn/(Zn þ Ca)  100). The in-
activity [17]. Few studies were concerned about the balance be- gredients and initial reaction solutions of Zn-BCP powders are
tween osteogenesis and osteoclastogenesis in bone regeneration shown in Table S1.
mediated by bioactive ions doping. According to bone healing
processes, osteoclasts also play vital roles in bone healing, espe- 2.3. Fabrication of Zn-BCP scaffolds
cially in a bone remolding phase. In addition, the effects of Zn on
osteogenesis and osteoclastogenesis are concentration dependent Zn-BCP scaffolds were fabricated by an extrusion 3D printing
[18e20]. Hence, we speculated that designing a kind of BCP ceramic method. A ceramic paste was prepared by mixing 8 g of the Zn-BCP
scaffold with the balance of promoting osteogenesis and inhibiting powder with 7 g of a 8 wt% polyvinyl alcohol solution under me-
osteoclastogenesis by Zn doping at an appropriate content would chanical stirring for 3 min. The scaffold models were designed by
facilitate bone defect healing. CAD. The prepared paste was extruded through a plastic nozzle
In the present study, Zn doping was used to improve the bone (inside diameter ¼ 400 mm), with an interval between adjacent
defect repair effects of BCP ceramic scaffolds by mediating the fibers of 800 mm at about 20  C, and successively dried at 37  C and
balance between osteogenesis and anti-osteoclastogenesis. The 60  C for 2 days and 1 day, respectively. The green scaffolds were
effects of Zn doping on the physicochemical properties, in vitro sintered at 1100  C for 2 h to obtain the final scaffolds. Zn-BCP
cytological responses and in vivo osteoinduction as well as bone scaffolds were denoted as ZnX-S.
defect regeneration of BCP were systematically investigated. We
aim to explore the effect of Zn doping content on the osteogenic 2.4. Phase composition and morphology characterization
and osteoclastic properties of BCP, and to further screen out the
appropriate doping content of Zn, which endows BCP with an X-ray diffraction (X'Pert, PANalytical, the Netherlands) with a
excellent bone defect repair effect. This study would pave the way CuKa radiation was used to analyze the phase of Zn-BCP scaffolds.
for the development of a more practical calcium phosphate-based Rietveld refinements were performed with the program of General
material for bone regeneration in the clinic. Structure Analysis System to calculate the phase composition of Zn-
BCP scaffolds. A refinement strategy was carried out according to
2. Materials and method the guidelines for structure refinement by using the Rietveld
method [21]. The morphology and structure of scaffolds were
2.1. Raw materials and reagents observed by a field emission scanning electron microscope (Merlin,
Carl Zeiss AG, Germany). Scanning electron microscope was
NH4OH solution (25%), (NH4)2HPO4, Ca(NO3)2$4H2O, and coupled with an energy dispersive spectrometer (EDS), which was
Zn(NO3)2$6H2O were commercially obtained from Guangzhou used to analyze the distribution of elements. The determination of
Chemical Reagent Factory (China). A DAPI staining solution, a BCIP/ Ca, P, O, and Zn elements were also analyzed by using an X-ray
NBT alkaline phosphatase (ALP) color development kit and a RIPA photoelectron spectroscope (Thermo Scientific K-Alpha, USA) with
lysis buffer were purchased from Biyotime (China). A Live/Dead Al Ka radiation.
staining solution was offered by Biotium (USA). An F-actin Labeling
Kit was offered by AAT Bioquest (USA). A receptor activator of nu- 2.5. In vitro degradation and ions release behaviors
clear factor-kappa B ligand (RANKL) was offered by R&D System
(USA). Disodium 4-nitrophenylphosphate, sodium b-glycer- An in vitro degradation experiment was conducted under acidic
ophosphate, vitamin C, and dexamethasone were bought from conditions (pH ¼ 4.5) because in a bone remodeling process, the
Sigma-Aldrich (USA). A micro-BCA protein assay kit was purchased ruffled border of osteoclasts has a pH in the range of 4.0e5.0 [22].
from Thermo Scientific (USA). A high-glucose Dulbecco's modified Each Scaffold (weight ¼ 0.2 g, diameter ¼ 7.5 mm, and
2
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

height ¼ 1.5 mm) was placed in a container with 10 mL of an ac- TRAP staining of the cells cultured for 5 days was also conducted
etate-sodium acetate buffer solution (pH ¼ 4.5) and incubated in a and observed by a fluorescence inverted microscope.
table concentrator (37  C, 60 rpm) for 1e12 weeks. The buffer so-
lution was exchanged by new ones every week. After immersion for 2.6.6. In vitro immune osteogenesis
setting time points (1, 4, 8, and 12 w), scaffolds were taken out and Raw 264.7 was cultured with scaffolds for 3 days, then collected
dried at 80  C for 3 days and weighted. The ion concentrations of the culture medium. The collected medium was filtrated by a
the soaking solution immersion with scaffolds for 1, 4, 8, and 12 w 0.2 mm filter membrane, followed by mixing with a fresh culture
were measured by an inductively coupled plasma atomic emission medium (25 vol% vs 75 vol%), and a macrophage-conditioned me-
spectroscope (Optimal 5300DV, Perkin Elmer, USA). dium was obtained. mBMSCs were first cultured for 12 h in a 48-
well plate, then the culture medium was replaced by the macro-
2.6. In vitro cell responses evaluation phage-conditioned medium mentioned above and refreshed every
other day. After culturing for 7 and 14 days, the osteogenic differ-
2.6.1. Cell cultivation entiation of mBMSCs tests including ALP activity, ALP staining, and
RAW264.7 (Cobioer, China) and mouse bone mesenchymal stem genes expression were conducted to evaluate the in vitro immune
cells (mBMSCs, ATCC, USA) were used as cell models in this study. A osteogenesis.
cell culture medium was composed of 10 vol% fetal bovine serum
and 90 vol% high-glucose Dulbecco's modified eagle's medium . The 2.7. In vivo experiments
osteoclast differentiation culture medium and osteogenic inductive
medium were obtained by further adding an osteoclast induction 2.7.1. In vivo angiogenesis and macrophage polarization assessment
factor (50 ng/mL of RANKL) and osteogenic inductive components Animal experiments were approved by the South China Univer-
(0.1 mM of dexamethasone, 50 mg/L of vitamin C, and 10 mM of sity of Technology Ethics Committee. Zn0-S, Zn2.5-S, and Zn5.0-S
sodium b-glycerophosphate), respectively. were used in this experiment. A total of 18 rats were divided into
three groups, and scaffolds (diameter ¼ 6 mm and height ¼ 3 mm)
were implanted into the abdominal skin. After seven days and 14
2.6.2. Cell proliferation
days after implantation, the scaffolds with surrounding tissues were
2  104 of mBMSCs in 500 mL of the culture medium were
retrieved, fixed, decalcified, and embedded in paraffin. Samples were
cultured with scaffolds (diameter ¼ 7.5 mm and height ¼ 1.5 mm)
sectioned with a thickness of 5 mm. The generation and development
in a 48-well plate. After being cultured for 1, 3, and 7 days, 250 mL of
of blood vessels were assessed by the immunohistochemical staining
a cell counting kit-8 working solution was added to each well and
of CD31. The effects of the scaffolds on macrophage polarization were
incubated in a cell incubator for 1 h. Then, 100 mL of the reaction
investigated using immunofluorescent staining. Cell nuclei were
solution of each well was exchanged to a 96-plate well. Finally, its
stained by DAPI. M1 macrophages were labeled by iNOS primary
optical density value at 450 nm wave length was measured by an
antibody and Cy3-labeled secondary antibody. M2 macrophages
ELISA reader (Thermo 3001, Thermo, USA) to determine the num-
were labeled by CD163 primary antibody and 488-labeled secondary
ber of cells.
antibody. The stained samples were then captured by a digital pa-
thology system (P250 Flash, 3D Histech, Hungary).
2.6.3. Cell activity and adhesion
After culturing for 3 days, the cells on scaffold were stained by
2.7.2. In vivo osteoinduction estimation
the Live/Dead staining kit for 15 min in a dark place and observed
Zn0-S, Zn2.5-S, and Zn5.0-S were used for the in vivo osteoin-
by a fluorescence inverted microscope (Axio Observer 7, ZEISS,
duction experiment. Four healthy adult mongrel dogs (male,
Germany). For the observation of cell morphologies, the cells
20e25 kg) were offered by the Guangdong Medical Laboratory
cultured for 1 and 7 days were stained with the F-actin Labeling Kit
Animal Center. Incisions (8 mm) over the paraspinal muscle were
and DAPI for 8 h and 5 min, respectively, before observing by a laser
first made on both sides of the spine along the dorsal midline, then
scanning confocal microscope (TCPS SP8, Leica, Germany).
scaffolds (diameter ¼ 6 mm and height ¼ 3 mm) were implanted.
After the implantation for 4 and 8 weeks, the dogs were sacrificed
2.6.4. In vitro osteogenesis and the scaffolds within them were harvested and immersed in
The effects of Zn doping on the in vitro osteogenesis were paraformaldehyde. Tissue sections were prepared as described
evaluated by ALP staining, ALP activities, and osteogenesis-related above. The sections were conducted by TRAP staining, toluidine
genes expression of mBMSCs cultured with scaffolds for 3, 7, and blue staining, Masson's Trichrome staining as well as hematoxylin
14 days. The genes expression was determined by a quantitative and eosin (HE) staining.
real-time polymerase chain reaction. GAPDH was used as a
housekeeping gene. The osteogenesis-related genes included ALP, 2.7.3. In vivo bone regeneration evaluation
collagen type I (Col-I), bone sialoprotein (BSP), osteocalcin (OCN), Zn0-S, Zn2.5-S, and Zn5.0-S were used for the in vivo bone
and runt-related transcription factor 2 (Runx2). The primer se- regeneration experiment. A total of 27 New Zealand white rabbits
quences are summarized in Table S2. (male, 2.5e3.0 kg) were used for this experiment. Scaffolds
(diameter ¼ 6 mm and height ¼ 8 mm) were implanted into the
2.6.5. In vitro osteoclastogenesis critical-size bone defect (diameter ¼ 6.2 mm and height ¼ 8 mm) in
Raw 264.7 at a cell density of 4  104 cell/mL was cultured with the condyles of the femur. After the implantation for 4, 8, and 12
a scaffold in a 48-well plate with a 500 mL osteoclast differentiation weeks, the femora together with the implants of the sacrificed
culture medium. The osteoclasts differentiation-related genes used rabbits was retrieved. Then, samples were evaluated by micro
in this study included c-Fos, tartrate-resistant acid phosphatase computed tomography imaging (Aloka Latheta LCT-200, HITACHI,
(TRAP), and nuclear factor of activated T-cells 1 (NFATc1). The Japan). Next, tissue sections were prepared as described above. The
primer sequences are provided in Table S2. Cells cultured in the obtained sections were stained with HE and Masson's trichrome
osteoclast differentiation culture medium for 5 days were stained and examined by a digital pathology system. The bone regeneration
with DAPI and F-actin Labeling Kit and observed by a laser scanning ratio according to the photographs of the HE staining sections was
confocal microscope. To further evaluate the osteoclastogenesis, quantitatively analyzed by an Image-Pro Plus 6.0 software.
3
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

2.8. Statistical analysis and HA (JCPDS #090432) can be observed, manifesting the suc-
cessful synthesis of BCP. The diffraction peaks of b-TCP shifted to
Quantitative data are presented as mean ± standard deviations. higher angles, while those of HA did not deviate with Zn doping.
A comparison between two means was made using the student's t- This indicated that in the presence of both b-TCP and HA, Zn
test, with statistical significance set as p<0.05 (‘*’ p<0.05, ‘#’ p<0.01, preferred entering the lattice structure of b-TCP. Zn ions with
and ‘$’ p<0.001). smaller radius substituted for Ca ions with larger radius [23],
resulted in the lattice contraction of b-TCP and led to the shift of
3. Results and discussion diffraction peaks to higher angles. The phase composition calcu-
lated by Rietveld refinements using the program of General
3.1. Phase composition, morphology and in vitro degradation of Zn- Structure Analysis System is showed in Fig. 1B. The mass ratio of b-
BCP scaffolds TCP and HA of all scaffolds fluctuated around 0.5 with the Zn doping
content increase, and it was in the range of 0.4e0.6. This indicated
Fig. 1A shows the X-ray diffraction patterns of Zn-BCP scaffolds. that we had successfully prepared BCP scaffolds with a gradient
Only the diffraction peaks corresponding to b-TCP (JCPDS #090169) variation of Zn doping but a uniformly phase ratio. Because the

Fig. 1. X-ray diffraction patterns (A), phase compositions (B), X-ray photoelectron spectroscope survey spectra (C) and macroscopic features (D) of scaffolds. D1: fiber diameter, D2:
fibers distance in X- and Y-direction, D3: fibers distance in Z-direction.

4
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

phase ratio of BCP plays a vital role in the osteoinduction perfor- Zn  5 mol.%. After seven days of culture, stem cells have covered
mance and bone defect repair effect of BCP [24,25], it is necessary to the whole scaffolds, indicating a long-term biocompatibility of BCP
control the phase ratio of BCP. X-ray photoelectron spectroscope scaffolds. Fig. 2CeE exhibit the ion concentration of the cell culture
and EDS were added to analyze the element composition and dis- medium immersed with scaffolds for seven days. Zn and Ca con-
tribution of the scaffold, and the results are shown in Fig. 1C and centrations in the culture medium increased as the Zn doping
Fig. S1, respectively. According to Fig. 1C, we could find that several content increased. The concentration of Zn showed a decreasing
deconvolution peaks were identified at 532, 350, 285, and 132 eV, trend as the immersion time prolonged, while the concentration of
which were attributed to O1s, Ca2p, C1s, and P2p, respectively. New Ca showed an opposite trend. The P concentrations in the culture
peaks attributed to Zn2p (1045 eV and 1022 eV) were found on medium of all scaffolds were in the range of 5e10 ppm, except for
Zn2.5-S and Zn5.0-S, and the peaks of Zn2p in Zn5.0-S were more Zn6.0-S. It is obvious that the ion concentrations of Zn6.0-S were
obvious than those in Zn2.5-S. Similar results can be seen in EDS remarkably higher than those of the others, and its Zn concentra-
spectra (Fig. S1). In addition, the results of element distribution tion on day 1 was about 37 ppm, which was significantly higher
show that Zn ions were uniformly distributed. than the toxic concentration (9.75 ppm) for cells [28]. This would be
The fabricated scaffolds were three-dimensionally connected the reason for the apoptosis of mBMSCs cultured on Zn6.0-S (Fig. 2A
(Fig. 1D) with a fiber diameter of about 350 mm. The fiber distance in and B).
XY-direction and Z-direction was about 400 mm and 170 mm, Fig. 3A and B show the ALP activity and ALP staining of mBMSCs
respectively (Fig. 1D, Table S3). As the sintering temperature cultured on scaffolds for 7 and 14 days, respectively. Zn doping can
increased, the scaffolds became denser, and fewer micropores on significantly up-regulate the ALP activity of mBMSCs both on days 7
free surfaces and facture surfaces can be observed (Fig. S2). When and 14, and cells cultured with Zn2.0-S and Zn2.5-S had the highest
the sintering temperature rose to 1150  C, the micropores almost ALP activity (Fig. 3A). The ALP activity analysis agreed with the ALP
disappeared. A study by Yuan et al. [26] found that micropores staining result. ALP was stained in blue, and the deeper staining
played a dominant role on ectopic bone formation. However, the color on Zn2.0-S and Zn2.5-S indicated a more secretion content of
existence of micropores was harmful to the mechanical strength of the ALP by the cells cultured on them (Fig. 3B).
scaffolds. Hence, taking the osteoinductivity and the mechanical The expression level of the osteogenesis-related genes of
strength into consideration, the sintering temperature of BCP mBMSCs cultured with scaffolds for 3, 7 and 14 days is exhibited in
scaffolds was set as 1100  C. Fig. 3CeG. For Runx2, only Zn2.5-S significantly enhanced its
Scanning electron microscope images of Zn-BCP scaffolds after expression on day 3. Zn-BCP scaffolds doped with Zn  1.0 mol.%
sintering are exhibited in Fig. S3. Many micropores can be observed remarkably promoted the expression level of Runx2 on day 7. Zn-
on the free surfaces and fracture surfaces of scaffolds. Compared to BCP scaffolds doped with Zn  2.0 mol.% could still enhance the
a bare BCP scaffold, the crystal grains on free surfaces of Zn-BCP expression level of Runx2 on day 14. It should be noted that, cells
scaffolds bonded more tightly with each other, especially for the cultured with Zn2.5-S expressed the highest level of Runx2 on
scaffolds with a doping content 2.0 mol.%. This indicated that Zn every detected time point. For Col-I and ALP, Zn-BCP scaffolds with
doping promoted the sintering and densification of BCP scaffolds a doping range of 1.0e3.5 mol.% significantly promoted their
sintered at 1100  C for 2 h, which was consistent with the result expression on day 3 and day 7. In addition, Zn1.5-S, Zn2.0-S, Zn2.5-
found by Sopyan et al. [27]. The denser structure of Zn-BCPs S, Zn3.0-S, and Zn3.5-S still could enhance the expression level of
resulted in their lower degradation rate than a bare BCP scaffold. ALP and Col-I on day 14. It is worth noting that, Zn5.0-S down-
The in vitro degradation behaviors of Zn-BCP scaffolds immersed in regulated the expression level of Col-I on day 14 as well as the
an acetic acid/sodium acetate buffer solution (pH ¼ 4.5) for 12 expression level of ALP on day 3 and day 7. For BSP and OCN, cells
weeks are shown in Fig. S4. The weight loss of all scaffolds cultured on Zn-BCP scaffolds with a doping range of 1.5e3.5 mol.%
increased as the immersion time prolonged. Compared to a bare expressed remarkably a higher level of BSP and OCN than those
BCP scaffold (Zn0-S), Zn-BCP scaffolds showed lower weight loss, cultured on Zn0-S (control group) at every detected time point.
whose values were about 15 wt% after immersion for 12 weeks Cells cultured on Zn2.5-S expressed the highest level of BSP and
(Fig. S4A). With the increase of the Zn doping content, the Ca and P OCN on day 14. Taking the expression level of all these
concentrations released by BCP scaffolds showed a decreasing osteogenesis-related genes into consideration, we could find that:
trend in the first four weeks of immersion, while the Zn concen- 1) Zn0.5-S did not significantly enhance the osteogenic differenti-
trations showed an increasing trend (Fig. S4BeD). The decrease of ation of mBMSCs; 2) Zn-BCP scaffolds with a doping range of
the Ca and P concentrations was related to the decreased degra- 1.0e5.0 mol.% could effectively promote osteogenic differentiation
dation rate of BCP scaffolds caused by Zn doping, while the increase compared to bare BCP scaffolds; 3) Zn2.0-S and Zn2.5-S showed the
of the Zn release concentration was related to the increase of the Zn most effective osteogenic differentiation accelerated ability.
doping content. It should be noticed that Zn-BCP scaffolds with a The ions, especially Zn, released from scaffolds played vital roles
doping content 2.5 mol.% could release Zn ions for a long time (at in stem cells survival, proliferation, and differentiation. In addition,
least for 12 weeks, Fig. S4D). many bioactive ions, including Zn, mediated cell actions in a
concentration-dependent manner [18e20]. Taking the prolifera-
3.2. In vitro cell compatibility and osteogenic differentiation tion, ALP activity, and the expression level of osteogenesis-related
genes into consideration, we could find that Zn doping mediated
The optical density value of mBMSCs cultured on the surfaces of the activity and osteogenic differentiation of mBMSCs in a doping
scaffolds increased as time prolonged, except for those cultured content-dependent manner because the released concentration of
with Zn6.0-S. Zn-BCP scaffolds with an appropriate Zn doping Zn depended on its doping content (Fig. 2C). Although we have not
content (1.0e3.5 mol.%) could promote the proliferation of mBMSCs thoroughly explored the molecular mechanism of the Zn-doped
at both day 3 and day 7 (Fig. 2A). This was consistent with the live/ BCP regulating the osteogenic differentiation of stem cells, a large
dead cell staining results for three days exhibited in Fig. 2B, for number of studies have proved that Zn may promote osteogenic
more green live cells can be observed on the surfaces of Zn-BCP differentiation of BMSCs through various cellular molecular path-
scaffolds, with a doping content of 1.0e3.5 mol.%. The cellular ways, such as MAPK pathway, cAMP pathway, and TGF-b/BMP
cytoskeleton staining for 1 day in Fig. S5 shows that mBMSCs could pathway [29]. Gao et al. [30] found that extracellular Zn entered
adhere and spread well on the surfaces of scaffolds doped with BMSCs by endocytosis, after internalization, activating the MAPK/
5
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 2. cell compatibility and ions concentration. The cell proliferation (A; on days 1, 3 and 7) and live/dead staining (B; on day 3) of mouse bone mesenchymal stem cells cultured
on the surfaces of scaffolds; the concentrations of Zn (C), Ca (D) and P (E) in a cell cultured medium.

6
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 3. alkaline phosphatase activity (A), alkaline phosphatase staining (B) and osteogenesis-related genes expression (CeG) of mouse bone mesenchymal stem cells cultured with
scaffolds.

ERK pathway and eventually promoting osteogenesis. A study by with RANKL (50 ng/mL). NFATc1 serves as a master regulator of
Zhu et al. [31] confirmed that extracellular Zn was transferred into osteoclast differentiation. The activation of c-Fos and NFATc1 is
cells by cellular receptors, and then activated the cAMP-PKA necessary for the sufficient differentiation of osteoclast [35,36].
pathway and triggered Ca responses in the cytoplasm, leading to For the osteoclast-related genes expression (Fig. 4AeC), Zn doping
the activation of the MAPK pathway. Zn was also found to promote significantly inhibited the expression level of NFATc1 on day 3.
the differentiation of BMSCs to mature matrix-secreting osteoblasts While on day 5, the expression level of NFATc1 in cells cultured
through the TGF-b/BMP signaling pathway [32]. Our in vitro oste- with different scaffolds showed no statistically significant differ-
ogenic differentiation results illustrated that the released Zn con- ence but decreased in the order of Zn0-S, Zn1.0-S, Zn2.5-S, and
centration from scaffolds doped with Zn  0.5 mol.% was not Zn5.0-S. Zn-BCP scaffolds could inhibit the expression level of c-
enough to stimulate osteoblast differentiation, while that from Fos on day 3 and day 5, and thereinto, Zn2.5-S and Zn5.0-S
scaffolds doped with Zn  6 mol.% was exorbitant and resulted in remarkably down-regulated the expression level of c-Fos on day
cell apoptosis. Zn-BCP scaffolds with a doping content of 2.5 mol.% 5. NFATc1 regulates a number of osteoclast specific genes such as
possessed the highest osteogenic differentiation property. Hence, TRAP through cooperation with c-Fos [37]. Zn2.5-S and Zn5.0-S
we chose Zn0-S, Zn1.0-S, Zn2.5-S, and Zn5.0-S to further explore inhibited the expression level of TRAP both on days 3 and 5, and
the effect of Zn doping on osteoclast differentiation. Zn1.0-S down-regulated the expression level of TRAP only on day
5.
3.3. In vitro osteoclastogenesis The monocyte/macrophage lineage fuses to form TRAP-positive
multinucleated cells (osteoclasts), and cells that have two or more
Bone matrix can be absorbed by multinucleated cells named nuclei are considered to be osteoclasts. According to cytoskeleton
osteoclasts [33], which differentiate from the macrophage/ staining images (Fig. 4D), we could easily find that there were
monocyte lineage when stimulated by RANKL [34]. Fig. 4 shows multinuclei cells which were osteoclasts, generating when cultured
the osteoclast differentiation of Raw 264.7 cultured with scaffolds with Zn0-S and Zn1.0-S. Not only that, the size of osteoclasts on

7
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 4. osteoclast differentiation of RAW 264.7. The osteoclast-related genes expression of RAW 264.7 co-cultured with scaffolds (AeC); the cytoskeleton staining (D) and tartrate-
resistant acid phosphatase staining (E) of RAW 264.7 cultured with scaffolds and in scaffold extracts for 5 days, respectively. The red circles in Fig. 4D and E labeled multinuclear
osteoclasts.

Zn0-S was larger than that on Zn1.0-S. However, there were few Calcineurin-NFATc1 signaling pathway. An animal study by Hie et al.
osteoclasts generating on the surfaces of Zn2.5-S and Zn5.0-S. TRAP [39] showed that Zn-administration decreased osteoclastogenesis by
staining of cells cultured in the extracts of scaffolds, showed in inhibiting RANK expression through the suppression of the pro-
Fig. 4E, also indicated that there were fewer TRAP positive cells in duction of reactive oxygen species and ERK activation in Zn-adequate
the Zn-doped scaffold extracts, especially in the extracts of Zn2.5-S rats. Further, Yamaguchi et al. [40] revealed that Zn suppressed
and Zn5.0-S. osteoclast differentiation and promoted osteoblast mineralization
Concerning the genes expression level of TRAP, c-Fos, and NFATc1 and did indeed act as a potent NF-kappa B activation antagonist in
as well as the fusion and formation of osteoclasts, we concluded that both osteoclast and osteoblast precursors. Our in vitro osteoclasto-
Zn inhibited osteoclastogenesis in a doping content-dependent genesis results revealed that Zn doping with a content 2.5 mol.%
manner. Higher Zn doping content resulted in a stronger inhibition could effectively suppress osteoclast differentiation. Hence, we chose
of osteoclastogenesis. Park et al. [38] found that Zn treatment during Zn0-S, Zn2.5-S, and Zn5.0-S to further explore the effects of Zn
osteoclast differentiation decreased RANKL-induced osteoclast for- doping on the in vivo osteoinduction and bone regeneration effects of
mation in a dose-dependent manner by suppressing the Ca2þ- BCP scaffolds.

8
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 5. polarization of macrophages surrounding scaffolds implanted for 14 days (A) and the osteogenesis of mouse bone mesenchymal stem cells cultured in the macrophage-
conditioned medium for 7 and 14 days (BeE). alkaline phosphatase activity (B), alkaline phosphatase staining images (C) and osteogenesis-related genes expression at day 7
(D) and day 14 (E).
9
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

3.4. Immunomodulation and angiogenesis beneficial to the migration, tube formation and angiogenesis
during a chicken embryo development. However, a high concen-
It was found that immune system also played vital roles in bone tration of Zn treatment had no positive effect on the endothelial
defect repair process [41,42], especially in the inflammation phase. activity and even had side-effects. Their results were similar to
Much literature indicated that promoting macrophages polariza- ours. Therefore, it is reasonable to believe that Zn2.5-S could
tion into M2 phenotype, which could secrete anti-inflammatory promote angiogenesis in vivo by releasing an appropriate con-
and pro-healing factors, was beneficial to the reduction of inflam- centration of Zn, while the excessive concentration of Zn released
mation and the enhancement of defect repair [43]. Fig. 5A displays by Zn5.0-S had no promoting effect on angiogenesis.
the macrophage phenotypes around scaffolds after implanted for
14 days. iNOS and CD163 positive cells were M1 and M2 macro- 3.5. In vivo osteoinduction
phages, respectively. It was obvious that, the distribution density of
M2 around Zn-BCP scaffolds, especially Zn2.5-S, was much higher When implanted in the mongrel dog dorsal muscle for four
than that around bare BCP scaffolds (Zn0-S). However, lower dis- weeks, numerous connective tissues were filled into the macro-
tribution density of M1 was found around Zn2.5-S and Zn5.0-S. pores of all scaffolds (Fig. S7). There were multinuclear macro-
These indicated that Zn doping contributed to the M2 polarization phages spreading on the interface between tissues and macropores.
while inhibited M1 polarization. This would result in reducing Inside the macropores, new blood vessels appeared. However, few
inflammation and facilitating defects repair. On the one hand, the materials degraded and few ectopic bones generated. TRAP staining
thickness of fibrous capsules around Zn2.5-S and Zn5.0-S was images (Fig. 7A) show that numerous TRAP positive cells (osteo-
thinner than that around Zn0-S (Fig. S6). On the other hand, the clasts) spread on the interface between tissues and macropores in
macrophage-conditioned medium extracted from Zn2.5-S and Zn0-S, and its distribution density was higher in the place closer to
Zn5.0-S was beneficial to the osteogenic differentiation of mBMSCs. the margins of macropores. Fewer osteoclasts were observed in
The macrophage-conditioned medium of Zn2.5-S and Zn5.0-S Zn2.5-S, and hardly any TRAP positive cells could be found in Zn5.0-
effectively enhanced the ALP activity of cells (Fig. 5B and C). S. This manifested that Zn doping could also inhibit the osteoclasts
Furthermore, the conditioned medium of Zn2.5-S up-regulated the formation in vivo, and the inhibition effect showed a doping
expression level of osteogenesis-related genes like ALP, BSP and content-dependent manner, which agreed with the in vitro exper-
OCN both on days 7 and 14 (Fig. 5D and E). Though the conditioned iments (Fig. 4).
medium of Zn5.0-S down-regulated the expression level of Runx2, When implanted for eight weeks, newly forming osteoids could
Col-I, ALP and OCN on day 7, it up-regulated the genes expression be observed in all groups (Fig. 7B). The osteoids were mainly
level of Col-I, ALP, BSP and OCN on day 14 (Fig. 5D and E). Therefore, distributed around the macropores of scaffolds. Osteoblasts with
Zn-doped BCP scaffolds, especially Zn2.5-S, showed the potential to stringy shape were lined in the interface between the osteoids and
promote osteogenesis by providing an anti-inflammation and pro- inner tissues, while osteocytes with circular shape were imbedded
healing microenvironment for cells involved in defect repair in the osteoids. These observations indicated that the osteoids
through promoting M2 macrophage polarization. initiated around the margins of macropores and infiltrated into the
Vascularization is necessary for bone defect repair. Blood internal gradually with the passage of implantation time. Toluidine
vessels not only can bring oxygen and nutrients to cells and blue staining images in Fig. S8 show that at week four, there were
remove metabolic waste but also transport the cells involved in toluidine blue positive chondrocytes, indicating that the formation
repair to the bone defect sites. Hence, promoting early vascular- of osteoids included entochondrostosis. The stained chondrocytes
ization can accelerate the repair process of bone defect [44]. Fig. 6 were mainly distributed in the margins of macropores, which also
shows the CD31 staining images (Fig. 6A) and the relevant sta- manifested that the osteoids initiated from the margins of mate-
tistical analysis of the amount (Fig. 6B) and diameter (Fig. 6C and rials. The newly osteoids formation ratio in Zn0-S was about 12%,
D) of vessels around scaffolds after implanted for 7 and 14 days. while it increased to about 25% in Zn2.5-S and dropped to 10% in
The CD31 positive brown circles were blood vessels. On day 7, Zn5.0-S (Fig. S9). According to the results of the Masson staining
numerous new blood vessels could be seen generating around all shown in Fig. S10, we found that the maturity of the generated
scaffolds, and few blood vessels distributing in the macropores in osteoids decreased in the order of Zn0-S, Zn2.5-S, and Zn5.0-
the interior of Zn0-S as well as Zn5.0-S. However, a few vessels S because the osteoids in Zn0-S showed comparable characteris-
had infiltrated into the macropores of Zn2.5-S. In addition, the tics of mature trabecular bone (lamellar bone), while the new bones
number of blood vessels around of Zn2.5-S was remarkably larger in Zn2.5-S and Zn5.0-S, especially Zn5.0-S, showed higher sectors of
than that of Zn0-S (Fig. 6B). When after implanted for 14 days, fibroreticular bone (woven bone).
blood vessels started to infiltrate into the inner macropores of The inhibition of osteoclasts formation and activity by Zn doping
Zn0-S and Zn5.0-S (Fig. 6A). More new blood vessels could be would result in the reduction of the active degradation rate of
found around all scaffolds, and there were still more blood vessels materials mediated by osteoclasts. Furthermore, Zn doping could
distributing on the periphery as well as in the interior of Zn2.5-S also reduce the passive degradation rate of BCP ceramic scaffolds
(Fig. 6A and B). Although the size of vessels around Zn2.5-S (Fig. S4). These two statuses led to a lower release concentration of
showed no significant difference from those around Zn0-S on Ca and P ions around scaffolds when implanted in vivo. The released
postoperative day 7 (Fig. 6C), we could easily see some larger Ca and P ions from materials were necessary for the formation of an
vessels around Zn2.5-S (Fig. 6A). On postoperative day 14, the size ectopic bone. When the concentration of Ca and P was higher than
of blood vessels around Zn2.5-S was significantly larger than their corresponding saturation concentration, ectopic bones would
those around Zn0-S (Fig. 6D). However, vessels around Zn5.0-S more easily generate [47]. In addition, the formation of an ectopic
were comparable to those around Zn0-S, both in number and bone and osteoclastogenesis would be blocked by macrophage
size (Fig. 6BeD). Recently, extracellular Zn has been shown to depletion [48]. These all manifested that the material active
promote an endothelial activity by triggering the upregulation of degradation mediated by osteoclasts and the material passive
pro-angiogenic cytokines, such as PDGF-BB and VEGF, in a Zn- degradation in body fluid by dissolving were both essential to the
sensing receptor ZnR/GPR39-dependent manner and through the induction of ectopic bones. We thought that, the higher degrada-
downstream Gaq-PLC pathways [45]. A study by Sai A et al. [46] tion rate of Zn0-S made ectopic bones initiate in it earlier and
showed that an appropriate concentration of Zn treatment was resulted in more mature ectopic bones. Zn5.0-S had adverse effects
10
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 6. blood vessel analysis on 7- and 14-day after surgery. CD31 immunohistochemical staining images of tissues around samples (A). The statistical analysis of the amount (B) and
diameter (CeD) of vessels. M: materials; CT: connective tissues; blue arrows: blood vessels.

on ectopic bone formation, which probably due to its obvious interconnected pores. Much more new bones could be found
inhibitory effect on osteoclastogenesis and low dissolution. While generating and infiltrating in Zn2.5-S. New bones were fully filled
Zn2.5-S could effectively promote osteogenesis, facilitating the in the macropores of Zn2.5-S at week 8 and week 12, while the new
formation and infiltration of blood vessels, which eventually made bones in the inner pores of Zn0-S and Zn5.0-S were under filled and
it possess the highest ectopic new bone ratio among all scaffolds. mainly spread around the margins of pores.
Fig. 9A displays the HE staining images of rabbit femur bones
3.6. In vivo bone regeneration implanted with scaffolds. When implanted for four weeks, the
macropores of all scaffolds were filled with tissues, with a few new
Fig. 8 exhibits the micro computed tomography images of rabbit bones which appeared around the margins of macropores. New
femur bones implanted with scaffolds. We can see that the bones gradually infiltrated into the inner macropores of Zn0-S and
implanted scaffolds degraded gradually as implanted time went Zn5.0-S at week 8 and week 12. It should be noticed that new bones
on as the diameter of scaffolds decreased gradually. The degrada- not only distributed around the margins of pores but also filled in
tion of scaffolds did not damage the structure of scaffolds. This some of the macropores of Zn2.5-S at week 4 and week 8. At week
made scaffolds still in a whole after implantation for 12 weeks. 12, with the degradation of the material, new bones further entered
With the degradation of materials, new bones (stained in red) were the interior of Zn2.5-S through the degradation pores/channels. In
generated and infiltrated into the inner pore of scaffolds through addition, the new bone area in Zn2.5-S was obviously larger than

11
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 7. tartrate-resistant acid phosphatase (A; 4 weeks) and hematoxylin and eosin (B; 8 weeks) staining images of scaffolds implanted in a mongrel dog dorsal muscle. In Fig. 7A,
while arrows: multinuclear osteoclasts. In Fig. 7B, M: materials; NB: new bones (osteoids); blue arrows: osteoblasts; black arrows: osteocytes.

Fig. 8. micro computed tomography images of the femur bone defect of rabbits after the implantation of scaffolds for 4, 8, and 12 weeks (New bones were staining in red).

12
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Fig. 9. hematoxylin and eosin staining images (A) and the new bone ratio (B) of scaffolds after implantation in the femur bone defect of rabbits for 4, 8, and 12 weeks. HB: host
bones, NB: new bones, CT: connective tissues, M: materials.

that in Zn0-S and Zn5.0-S. The new bone ratio calculation according at the late implantation state (12 w). Zn2.5-S could effectively
to the HE staining results is shown in Fig. 9B, it increased with the promote osteoblast differentiation, facilitate new blood vessels
passage of implanted time in all scaffolds. The new bone ratio of formation and infiltration, promote M2 polarization, and reduce
Zn2.5-S was the highest at all-time points. At weeks 4 and 8, the fibrous encapsulation, which made sure it possessed the highest
new bone ratio of Zn5.0-S was slightly higher than that of Zn0-S but new bone ratio among all scaffolds at early implantation stages. In
with no statistical difference. addition, Zn2.5-S inhibited osteoclasts differentiation and activity
Bone remodeling has been considered to consist of four to an appropriate degree, and we thought that this appropriate
sequential processes [49], including: 1) osteoclast progenitors are inhibition on osteoclastogenesis would not be harmful to bone
recruited; 2) osteoclast progenitors differentiate into mature oste- remolding. Namely, the balance between the pro-osteogenesis and
oclasts and resorb damaged bones; 3) osteoclasts apoptosis and anti-osteoclastogenesis mediated by Zn2.5-S made it possess the
osteoblast progenitors are recruited; 4) osteoblast progenitors highest new bone ratio at late implantation stages.
differentiate into mature osteoblasts, and produce new bone matrix
and then mineralize [50,51]. Almost all new bone formation was 4. Conclusion
observed in areas where bone resorption had previously occurred,
known as basic multicellular units in different anatomical struc- In this study, BCP scaffolds with different Zn doping contents but
tures [52]. Under homeostatic conditions, the balance between similar phase compositions were prepared. The effects of Zn doping
osteoblast-mediated bone formation and osteoclast-mediated bone on the osteogenesis and osteoclastogenesis of BCP scaffolds were
resorption was strictly regulated without a major alteration. systematically investigated in vitro and in vivo. The osteoblast dif-
Disruption of this balance would result in abnormal bone regen- ferentiation of mBMSCs and osteoclast differentiation of RAW264.7
eration. Most anti-resorptive agents suppress osteoclasts formation both showed a Zn doping content-dependent behavior. By
and activity while inhibiting bone formation simultaneously. increasing the Zn doping content (0e5 mol.%), the anti-
Analogously, the induced bone formation activity of anabolic osteoclastogenesis effect of Zn-BCP scaffolds gradually increased.
agents is modulated by increasing bone resorption at the same time While the pro-osteogenesis property of Zn-BCP scaffolds first
[53]. increased and then decreased, and the most effective doping con-
Zn5.0-S could promote osteogenic differentiation, relieve tent of Zn for promoting osteoblast differentiation was 2.5 mol.%.
inflammation, and reduce fibrous capsule formation, making it The BCP scaffold with a Zn doping content of 2.5 mol.% possessed
promote new bone formation at early implantation stages (4 w and the highest osteoinduction activity and fastest new bone formation,
8 w). While its excessive suppression effect on osteoclastogenesis which would be associated with the balance between pro-osteo-
was not conducive to bone remolding, and its low dissolution rate genesis and anti-osteoclastogenesis mediated by the appropriate
also did not match with the infiltration rate of new bone tissues. Zn doping content. Hence, Zn2.5-S would be a promising bioma-
These two factors made Zn5.0-S not facilitate new bone formation terial for bone defect regeneration.

13
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

Author contributions [11] K.D. Hankenson, G. Zimmerman, R. Marcucio, Biological perspectives of


delayed fracture healing, Injury 45 (Suppl 2) (2014) S8eS15, https://doi.org/
10.1016/j.injury.2014.04.003.
Teliang Lu: conceptualization, methodology, investigation, and [12] K.D. Hankenson, K. Gagne, M. Shaughnessy, Extracellular signaling molecules
writing e original draft. to promote fracture healing and bone regeneration, Adv. Drug Delivery Rev.
Xinyuan Yuan: investigation and formal analysis. 94 (2015) 3e12, https://doi.org/10.1016/j.addr.2015.09.008.
[13] J.P. O'Connor, D. Kanjilal, M. Teitelbaum, S.S. Lin, J.A. Cottrell, Zinc as a ther-
Luhui Zhang: formal analysis. apeutic agent in bone regeneration, Materials 13 (10) (2020), https://doi.org/
Fupo He: writing e review & editing and validation. 10.3390/ma13102211.
Xiaolan Wang: validation and formal analysis. [14] T. Lu, X. Yuan, L. Zhang, F. He, X. Wang, Y. Zhang, J. Ye, High throughput
synthesis and screening of zinc-doped biphasic calcium phosphate for bone
Jiandong Ye: conceptualization, supervision, writing - review & regeneration, Appl. Mater. Today 25 (2021), 101225, https://doi.org/10.1016/
editing, and funding acquisition. j.apmt.2021.101225.
[15] X. Huang, D.H. Huang, T. Zhu, X.H. Yu, K.C. Xu, H.Y. Li, H. Qu, Z.Y. Zhou,
K. Cheng, W.J. Wen, Z.M. Ye, Sustained zinc release in cooperation with CaP
Declaration of competing interest
scaffold promoted bone regeneration via directing stem cell fate and trig-
gering a pro-healing immune stimuli, J. Nanobiotechnol. (1) (2021) 19,
The authors declare that they have no known competing https://doi.org/10.1186/s12951-021-00956-8.
[16] R. Cruz, J. Calasans-Maia, S. Sartoretto, V. Moraschini, A.M. Rossi, R.S. Louro,
financial interests or personal relationships that could have
J.M. Granjeiro, M.D. Calasans-Maia, Does the incorporation of zinc into cal-
appeared to influence the work reported in this paper. cium phosphate improve bone repair? A systematic review, Ceram. Int. 44 (2)
(2018) 1240e1249, https://doi.org/10.1016/j.ceramint.2017.10.157.
Data availability [17] D.V. Shepherd, K. Kauppinen, R.A. Brooks, S.M. Best, An in vitro study into the
effect of zinc substituted hydroxyapatite on osteoclast number and activity,
J. Biomed. Mater. Res. Part A 102 (11) (2014) 4136e4141, https://doi.org/
Data will be made available on request. 10.1002/jbm.a.35089.
[18] S. An, Q. Gong, Y. Huang, Promotive effect of zinc ions on the vitality,
migration, and osteogenic differentiation of human dental pulp cells, Biol.
Acknowledgments
Trace Elem. Res. 175 (1) (2017) 112e121, https://doi.org/10.1007/s12011-
016-0763-7.
This work was supported by the National Key R&D Program of [19] J.M. Yu, L.Z. Xu, N. Xie, K. Li, Y.H. Xi, X.L. Liu, X.B. Zheng, X.S. Chen, X.J. Ye,
D.X. Wei, Optimal Zn-modified Ca-Si-based ceramic nanocoating with Zn ion
China (Grant No. 2016YFB0700803) and Guangdong Basic and
release for osteoblast promotion and osteoclast inhibition in bone tissue en-
Applied Basic Research Foundation (Grant No. 2022A1515110274). gineering, J. Nanomater. 2017 (2017), https://doi.org/10.1155/2017/7374510.
[20] H.J. Seo, Y.E. Cho, T. Kim, H.I. Shin, I.S. Kwun, Zinc may increase bone for-
Appendix A. Supplementary data mation through stimulating cell proliferation, alkaline phosphatase activity
and collagen synthesis in osteoblastic MC3T3-E1 cells, Nutr. Res. Pract. 4 (5)
(2010) 356e361, https://doi.org/10.4162/nrp.2010.4.5.356.
Supplementary data to this article can be found online at [21] L.B. McCusker, R.B. Von Dreele, D.E. Cox, D. Louer, P. Scardi, Rietveld refine-
https://doi.org/10.1016/j.mtchem.2023.101410. ment guidelines, J. Appl. Crystallogr. 32 (1999) 36e50, https://doi.org/
10.1107/s0021889898009856.
[22] N.C. Andres, N.L. D'Elia, J.M. Ruso, A.E. Campelo, V.L. Massheimer, P.V. Messina,
References Manipulation of Mg2þ-Ca2þ switch on the development of bone mimetic
hydroxyapatite, ACS Appl. Mater. Interfaces 9 (18) (2017) 15698e15710,
[1] S. Samavedi, A.R. Whittington, A.S. Goldstein, Calcium phosphate ceramics in https://doi.org/10.1021/acsami.7b02241.
bone tissue engineering: a review of properties and their influence on cell [23] E. Boanini, M. Gazzano, A. Bigi, Ionic substitutions in calcium phosphates
behavior, Acta Biomater. 9 (9) (2013) 8037e8045, https://doi.org/10.1016/ synthesized at low temperature, Acta Biomater. 6 (6) (2010) 1882e1894,
j.actbio.2013.06.014. https://doi.org/10.1016/j.actbio.2009.12.041.
[2] N. Eliaz, N. Metoki, Calcium phosphate bioceramics: a review of their history, [24] Z.R. Tang, Y.F. Tan, Y.L. Ni, J. Wang, X.D. Zhu, Y.J. Fan, X.N. Chen, X. Yang,
structure, properties, coating technologies and biomedical applications, Ma- X.D. Zhang, Comparison of ectopic bone formation process induced by four
terials 10 (4) (2017), https://doi.org/10.3390/ma10040334. calcium phosphate ceramics in mice, Mater. Sci. Eng. C 70 (2017) 1000e1010,
[3] Y. Li, X.M. Xu, X.M. Liu, B. Li, Y. Han, Y.F. Zheng, D.F. Chen, K.W.W. Yeung, https://doi.org/10.1016/j.msec.2016.06.097.
Z.D. Cui, Z.Y. Li, Y.Q. Liang, S.L. Zhu, X.B. Wang, S.L. Wu, Photoelectrons [25] C.S. Shao, L.J. Chen, R.M. Tang, B. Zhang, J.J. Tang, W.N. Ma, Degradation and
mediating angiogenesis and immunotherapy through heterojunction film for biological performance of porous osteomimetic biphasic calcium phosphate
noninvasive disinfection, Adv. Sci. 7 (17) (2020), https://doi.org/10.1002/ in vitro and in vivo, Rare Met. 41 (2) (2022) 457e468, https://doi.org/10.1007/
advs.202000023. s12598-021-01814-0.
[4] K. Zhang, Y. Zhou, C. Xiao, W.L. Zhao, H.F. Wu, J.Q. Tang, Z.T. Li, S. Yu, X.F. Li, [26] H.P. Yuan, K. Kurashina, J.D. de Bruijn, Y.B. Li, K. de Groot, X.D. Zhang,
L. Min, Z.T. Yu, G. Wang, L. Wang, K. Zhang, X. Yang, X.D. Zhu, C.Q. Tu, A preliminary study on osteoinduction of two kinds of calcium phosphate
X.D. Zhang, Application of hydroxyapatite nanoparticles in tumor-associated ceramics, Biomaterials 20 (19) (1999) 1799e1806, https://doi.org/10.1016/
bone segmental defect, Adv. Sci. 5 (8) (2019), https://doi.org/10.1126/ s0142-9612(99)00075-7.
sciadv.aax6946. [27] I. Sopyan, Gunawan, Q.H. Shah, M. Mel, Fabrication and sintering behavior of
[5] J.N. Fu, X.M. Liu, L. Tan, Z.D. Cui, Y.F. Zheng, Y.Q. Liang, Z.Y. Li, S.L. Zhu, zinc-doped biphasic calcium phosphate bioceramics, Mater. Manuf. Process.
K.W.K. Yeung, X.B. Feng, X.B. Wang, S.L. Wu, Photoelectric-responsive extra- 31 (6) (2016) 713e718, https://doi.org/10.1080/10426914.2015.1048361.
cellular matrix for bone engineering, ACS Nano 13 (11) (2019) 13581e13594, [28] W. Watjen, H. Haase, M. Biagioli, D. Beyersmann, Induction of apoptosis in
https://doi.org/10.1021/acsnano.9b08115. mammalian cells by cadmium and zinc, Environ. Health Perspect. 110 (s5)
[6] J.N. Fu, W.D. Zhu, X.M. Liu, C.Y. Liang, Y.F. Zheng, Z.Y. Li, Y.Q. Liang, D. Zheng, (2002) 865e867, https://doi.org/10.1289/ehp.110-1241262.
S.L. Zhu, Z.D. Cui, S.L. Wu, Self-activating anti-infection implant, Nat. Commun. [29] H.Y. Li, M.Z. Li, X. Ran, J.C. Cui, F. Wei, G.L. Yi, W. Chen, X.L. Luo, Z.W. Chen, The
12 (1) (2021), https://doi.org/10.1038/s41467-021-27217-4. role of zinc in bone mesenchymal stem cell differentiation, Cell Reprogram. 24
[7] S.E. Kim, K. Park, Recent advances of biphasic calcium phosphate bioceramics (2) (2022) 80e94, https://doi.org/10.1089/cell.2021.0137.
for bone tissue regeneration, in: H.J. Chun, R.L. Reis, A. Motta, G. Khang (Eds.), [30] X.M. Gao, Y.Y. Xue, Z. Zhu, J.Y. Chen, Y.H. Liu, X.T. Cheng, X. Zhang, J. Wang,
Biomimicked Biomaterials: Advances in Tissue Engineering and Regenerative X.B. Pei, Q.B. Wan, Nanoscale zeolitic imidazolate framework-8 activator of
Medicine, vol. 1250, 2020, pp. 177e188. canonical MAPK signaling for bone repair, ACS Appl. Mater. Interfaces 13 (1)
[8] J.M. Bouler, P. Pilet, O. Gauthier, E. Verron, Biphasic calcium phosphate ce- (2021) 97e111, https://doi.org/10.1021/acsami.0c15945.
ramics for bone reconstruction: a review of biological response, Acta Bio- [31] D.H. Zhu, Y.C. Su, M.L. Young, J. Ma, Y.F. Zheng, L.P. Tang, Biological responses
mater. 53 (2017) 1e12, https://doi.org/10.1016/j.actbio.2017.01.076. and mechanisms of human bone marrow mesenchymal stem cells to Zn and
[9] J. Wang, Y. Chen, X. Zhu, T. Yuan, Y. Tan, Y. Fan, X. Zhang, Effect of phase Mg biomaterials, ACS Appl. Mater. Interfaces 9 (33) (2017) 27453e27461,
composition on protein adsorption and osteoinduction of porous calcium https://doi.org/10.1021/acsami.7b06654.
phosphate ceramics in mice, J. Biomed. Mater. Res. Part A 102 (12) (2014) [32] K. Yusa, O. Yamamoto, M. Iino, H. Takano, M. Fukuda, Z.W. Qiao, T. Sugiyama,
4234e4243, https://doi.org/10.1002/jbm.a.35102. Eluted zinc ions stimulate osteoblast differentiation and mineralization in
[10] H. Yuan, C.A. van Blitterswijk, K. de Groot, J.D. de Bruijn, A comparison of bone human dental pulp stem cells for bone tissue engineering, Arch. Oral Biol. 71
formation in biphasic calcium phosphate (BCP) and hydroxyapatite (HA) (2016) 162e169, https://doi.org/10.1016/j.archoralbio.2016.07.010.
implanted in muscle and bone of dogs at different time periods, J. Biomed. [33] B.J. Kim, J.M. Koh, Coupling factors involved in preserving bone balance, Cell.
Mater. Res. Part A 78A (1) (2006) 139e147, https://doi.org/10.1002/ Mol. Life Sci. 76 (7) (2019) 1243e1253, https://doi.org/10.1007/s00018-018-
jbm.a.30707. 2981-y.

14
T. Lu, X. Yuan, L. Zhang et al. Materials Today Chemistry 29 (2023) 101410

[34] H. Takayanagi, RANKL as the master regulator of osteoclast differentiation, Funct. Mater. 30 (44) (2020), 1909874, https://doi.org/10.1002/
J. Bone Miner. Metabol. 39 (1) (2021) 13e18, https://doi.org/10.1007/s00774- adfm.201909874.
020-01191-1. [45] D.H. Zhu, Y.C. Su, Y.F. Zheng, B.M. Fu, L.P. Tang, Y.X. Qin, Zinc regulates vascular
[35] J.H. Park, N.K. Lee, S.Y. Lee, Current understanding of RANK signaling in endothelial cell activity through zinc-sensing receptor ZnR/GPR39, Am. J.
osteoclast differentiation and maturation, Mol. Cells 40 (10) (2017) 706e713, Physiol. 314 (4) (2018) C404eC414, https://doi.org/10.1152/ajpcell.00279.2017.
https://doi.org/10.14348/molcells.2017.0225. [46] S.A. Sreenivasamurthy, F.F. Akhter, A. Akhter, Y.C. Su, D.H. Zhu, Cellular
[36] J.Y. Kang, N. Kang, Y.M. Yang, J.H. Hong, D.M. Shin, The role of Ca2þ-NFATc1 mechanisms of biodegradable zinc and magnesium materials on promoting
signaling and its modulation on osteoclastogenesis, Int. J. Mol. Sci. 21 (10) angiogenesis, Biomater. Adv. (2022) 139, https://doi.org/10.1016/
(2020), https://doi.org/10.3390/ijms21103646. j.bioadv.2022.213023.
[37] J.H. Kim, N. Kim, Regulation of NFATc1 in osteoclast differentiation, J. Bone [47] A.M.C. Barradas, H.P. Yuan, C.A. van Blitterswijk, P. Habibovic, Osteoinductive
Metabol. 21 (4) (2014) 233e241, https://doi.org/10.11005/jbm.2014.21.4.233. biomaterials: current knowledge of properties, experimental models and
[38] K.H. Park, B. Park, D.S. Yoon, S.H. Kwon, D.M. Shin, J.W. Lee, H.G. Lee, J.H. Shim, biological mechanisms, Eur. Cells Mater. 21 (2011) 407e429, https://doi.org/
J.H. Park, J.M. Lee, Zinc inhibits osteoclast differentiation by suppression of 10.22203/eCM.v021a31.
Ca2þ-Calcineurin-NFATc1 signaling pathway, Cell Commun. Signal. 11 (2013), [48] N.L. Davison, A.-L. Gamblin, P. Layrolle, H. Yuan, J.D. de Bruijn, F. Barrere-de
https://doi.org/10.1186/1478-811x-11-74. Groot, Liposomal clodronate inhibition of osteoclastogenesis and osteoin-
[39] M. Hie, I. Tsukamoto, Administration of zinc inhibits osteoclastogenesis duction by submicrostructured beta-tricalcium phosphate, Biomaterials 35
through the suppression of RANK expression in bone, Eur. J. Pharmacol. 668 (19) (2014) 5088e5097, https://doi.org/10.1016/j.biomaterials.2014.03.013.
(1e2) (2011) 140e146, https://doi.org/10.1016/j.ejphar.2011.07.003. [49] R. Hattner, B.N. Epker, H.M. Frost, Suggested sequential mode of control of
[40] M. Yamaguchi, M.N. Weitzmann, Zinc stimulates osteoblastogenesis and changes in cell behaviour in adult bone remodelling, Nature 206 (983) (1965)
suppresses osteoclastogenesis by antagonizing NF-kappa B activation, Mol. 489e490, https://doi.org/10.1038/206489a0.
Cell. Biochem. 355 (1e2) (2011) 179e186, https://doi.org/10.1007/s11010- [50] B. Langdahl, S. Ferrari, D.W. Dempster, Bone modeling and remodeling: po-
011-0852-z. tential as therapeutic targets for the treatment of osteoporosis, Ther. Adv.
[41] T. Zhang, Y. Yao, Effects of inflammatory cytokines on bone/cartilage repair, Musculoskel. Dis. 8 (6) (2016) 225e235, https://doi.org/10.1177/
J. Cell. Biochem. 120 (5) (2018) 6841e6850, https://doi.org/10.1002/ 1759720x16670154.
jcb.27953. [51] J.C. Crockett, M.J. Rogers, F.P. Coxon, L.J. Hocking, M.H. Helfrich, Bone
[42] H. Liu, D. Li, Y. Zhang, M. Li, Inflammation, mesenchymal stem cells and bone remodelling at a glance, J. Cell Sci. 124 (7) (2011) 991e998, https://doi.org/
regeneration, Histochem. Cell Biol. 149 (4) (2018) 393e404, https://doi.org/ 10.1242/jcs.063032.
10.1007/s00418-018-1643-3. [52] A.M. Parfitt, The coupling of bone formation to bone resorption: a critical
[43] W. Liu, J. Li, M. Cheng, Q. Wang, K.W.K. Yeung, P.K. Chu, X. Zhang, Zinc- analysis of the concept and of its relevance to the pathogenesis of osteopo-
modified sulfonated polyetheretherketone surface with immunomodulatory rosis, Metab. Bone Dis. Relat. Res. 4 (1) (1982) 1e6, https://doi.org/10.1016/
function for guiding cell fate and bone regeneration, Adv. Sci. 5 (10) (2018), 0221-8747(82)90002-9.
1800749, https://doi.org/10.1002/advs.201800749. [53] J.-M. Kim, C. Lin, Z. Stavre, M.B. Greenblatt, J.-H. Shim, Osteoblast-osteoclast
[44] A.R. Armiento, L.P. Hatt, G.S. Rosenberg, K. Thompson, M.J. Stoddart, Func- communication and bone homeostasis, Cells 9 (9) (2020), https://doi.org/
tional biomaterials for bone regeneration: a lesson in complex biology, Adv. 10.3390/cells9092073.

15

You might also like