Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Acta Biomaterialia 10 (2014) 2518–2528

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Study of bilineage differentiation of human-bone-marrow-derived


mesenchymal stem cells in oxidized sodium alginate/N-succinyl
chitosan hydrogels and synergistic effects of RGD modification
and low-intensity pulsed ultrasound
Yingying Wang a, Wenzhen Peng b, Xia Liu a, Minghua Zhu c, Tao Sun c, Qiang Peng c, Yi Zeng c, Bo Feng a,
Wei Zhi a, Jie Weng a, Jianxin Wang a,⇑
a
Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, People’s
Republic of China
b
Department of Biochemistry and Molecular Biology, College of Basic and Forensic Medicine, Sichuan University, Chengdu 610041, People’s Republic of China
c
Sichuan Centre for Disease Control and Prevention, Chengdu 610041, People’s Republic of China

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

Article history: The level of formation of new bone and vascularization in bone tissue engineering scaffold implants is
Received 19 July 2013 considered as a critical factor for clinical application. In this study, an approach using an RGD-grafted oxi-
Received in revised form 5 December 2013 dized sodium alginate/N-succinyl chitosan (RGD–OSA/NSC) hydrogel as a scaffold and low-intensity
Accepted 26 December 2013
pulsed ultrasound (LIPUS) as mechanical stimulation was proposed to achieve a high level of formation
Available online 3 January 2014
of new bone and vascularization. An in vitro study of endothelial and osteogenic differentiations of
human-bone-marrow-derived mesenchymal stem cells (hMSCs) was conducted to evaluate it. The results
Keywords:
showed that RGD–OSA/NSC composite hydrogels presented good biological properties in attachment,
Bone tissue engineering
Hydrogel
proliferation and differentiation of cells. The MTT cell viability assay showed that the total number of
LIPUS cells increased more significantly in the LIPUS-stimulated groups with RGD than that in the control ones;
Osteogenic differentiation similar results were obtained for alkaline phosphatase activity/staining and mineralized nodule forma-
Endothelial differentiation tion assay of osteogenic induction and immunohistochemical test of endothelial induction. The positive
synergistic effect of LIPUS and RGD on the enhancement of proliferation and differentiation of hMSCs was
observed. These findings suggest that the hybrid use of RGD modification and LIPUS might provide one
approach to achieve a high level of formation of new bone and vascularization in bone tissue engineering
scaffold implants.
Ó 2013 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction injected to fill defects of any size and can homogenously suspend
cells within a 3-D environment. Physically cross-linked hydrogels
Bone tissue engineering provides a potential approach for the do not readily meet the basic demands for tissue engineering due
production of three-dimensional (3-D) implants [1–3]. However, to their rapid degradation property. Chemically cross-linked com-
there are still some challenges associated with 3-D bone regenera- posite hydrogels (prepared without using chemical cross-linking
tion, including creating regenerated tissue with similar structural agents like formaldehyde), which exhibit excellent properties
and mechanical properties to those of the natural bone and achiev- of biocompatibility, degradation and mechanical strength, are
ing successful integration of scaffolds with the host bone [4–6]. The regarded as optimal scaffold materials for tissue engineering
levels of formation of new bone and vascularization are considered [13]. However, hydrophilic surfaces of hydrogels are not beneficial
to be critical factors for the clinical application of bone tissue engi- for the adsorption of protein and are not conducive to the adhesion
neering [7,8]. Both synthetic and natural materials have been ex- and growth of cells [14] and, in turn, to the formation of new bone
plored as potential scaffolds for bone regeneration [9–12]. Of and vascularization. One approach to overcome this is to use RGD
these materials, the most commonly explored is hydrogels, which to modify hydrophilic surfaces. The Schiff base reaction between
are particularly attractive because they can be non-invasively amine groups and aldehyde groups offers the possibility of modifi-
cation of RGD on the hydrophilic surfaces of hydrogels, whereas
⇑ Corresponding author. polysaccharides can provide aldehyde groups for the Schiff base
E-mail addresses: j.wang63@gmail.com, jwang@swjtu.edu.cn (J. Wang). reaction by oxidization of linear polysaccharides, and their source

1742-7061/$ - see front matter Ó 2013 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.actbio.2013.12.052
Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528 2519

is very rich [14]. It is for these reasons that polysaccharide hydro- 2.1. Materials
gels are receiving increasing attention. In this study, we proposed
to use an RGD-grafted oxidized sodium alginate/N-succinyl chito- Sodium alginate (SA, Mw: 612 kDa) and chitosan (CS, Mw:
san (RGD–OSA/NSC) polysaccharide composite hydrogel as a scaf- 209 kDa) were purchased from Maichao Chemical Reagent (Shang-
fold and to employ low-intensity pulsed ultrasound (LIPUS) as hai, China) and Aokang Biological Technology Co. Ltd (Shandong,
mechanical stimulation so as to achieve good integration of scaf- China), respectively. All other chemicals and solvents were pur-
folds with the host bone by improving the formation of new bone chased from Kelong Chemical Reagent Factory (Chengdu, China)
and vascularization in bone tissue engineering scaffolds [15–18]. with reagent grade or better, without further purification.
Angiogenesis is a process of endothelial migration and prolifera-
tion to form a capillary network while endothelial differentiation 2.2. Preparation of composite hydrogels
of marrow-derived mesenchymal stem cells (MSCs) is very impor-
tant for vascularization in scaffolds when implanted. Vasculariza- Oxidized sodium alginate (OSA) was prepared according to the
tion in scaffolds will be conducive to the formation of new bone. method previously reported [19,20]. Aqueous solutions of RGD
Additionally, the ability of scaffold materials to induce osteogenic (4 ml, 10 mg ml–1) and OSA (2 ml, 10% w/v) were mixed and stirred
differentiation of MSCs determines the level of formation of new in the dark at room temperature for 36 h and then the obtained
bone in scaffold materials. In the present study, an in vitro study RGD–OSA product was lyophilized at –80 °C for use. N-succinyl
of endothelial and osteogenic differentiation of MSCs in RGD– chitosan (NSC) was prepared according to the previously reported
OSA/NSC hydrogels is employed to evaluate the ability of scaffold method [21,22]. The final formation of RGD-oxidized sodium algi-
materials to induce the formation of new bone and vascularization. nate (RGD–OSA)/N-succinyl chitosan (NSC) hydrogels was based
The synergistic effect of LIPUS and RGD on the proliferation and on the Schiff base reaction. 1 g of sterilized NSC was dissolved in
differentiation of human mesenchymal stem cells (hMSCs) is also 40 ml sterilized phosphate buffer solution (PBS, pH = 7.4). Simi-
investigated. To the authors’ knowledge, there has been little pre- larly, 2 g of sterilized RGD–OSA was dissolved in 20 ml PBS. Steril-
vious research of this topic. ized NSC and RGD–OSA solutions were mixed by stirring
vigorously. Subsequently, the mixed solution became highly vis-
cous and was then used to coat glass discs using a spinning coater.
2. Materials and methods The coating finally formed into an RGD–OSA/NSC gel on the glass
discs within a few minutes at 37 °C. All the hydrogel-coated glass
A flow chart for the preparation of materials and cell culture in discs for cell culture were immersed in 75% ethanol for 2 h for fur-
this study is shown in Fig. 1. ther sterilization and then rinsed three times with sterilized PBS

Fig. 1. Flow chart for preparation of materials and cell culture.


2520 Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528

before cell culture. All the hydrogel-coated glass discs were placed equipped with a digital camera and Image Pro-plus (Radiance
in 24-well plates for cell culture. 2100; Biorad Laboratories, Hercules, CA, USA).

2.3. Cell culture 2.4.4. Endothelial differentiation assay


After endothelial induction of 28 days, endothelial differentia-
hMSCs were purchased from Sichuan University and cultured in tion of hMSCs was examined by immunofluorescence staining, as
25 cm2 flasks in D-minimum essential medium (DMEM) supple- described by Wang et al. [26]. In brief, cells were rinsed twice with
mented with 10% fetal bovine serum (FBS) and 100 U ml–1 penicil- PBS and fixed in 4% paraformaldehyde for 20 min at room temper-
lin/streptomycin antibiotics. The cell culture medium was replaced ature and then incubated with 1% Triton X-100 in PBS to permea-
with fresh medium every 3 days. The hMSCs were purified by con- bilize cell membrane for 10 min at room temperature. After that,
trolling the passage time [23]. The average passage time was these cells were incubated overnight at 4 °C with primary antibody
2 days. The fifth passage cells, which grew stably and kept their (Anti-CD31, 1:25, Beijing Biosynthesis Biotechnology Co. Ltd) and
primary characteristics, were used for later induction experiments. then the non-specific bindings were blocked by washing three
Passage cells were harvested with 0.25% trypsin-ethylenediamine- times with the blocking buffer of PBS containing 4% bovine serum
tetraacetic acid solution and then they were resuspended in com- albumin. After the removal of the blocking buffer, these cells were
plete medium for later seeding when reaching 80% confluence. The incubated again with secondary antibodies (fluorescein isothiocy-
viability of passage cells was measured by the Alamer Blue test. anate (FITC)-conjugated goat-anti-rabbit immunoglobulin G,
When the activity of the cells reached 90%, the passage cells were 1:100, Beijing Zhongshan Glodenbridge Biotechnology Co. Ltd)
seeded on glass discs with OSA/NSC, RGD–OSA/NSC coatings and for 1 h at room temperature in the dark and then rinsed three
blank glass discs in 24-well plates at a density of 0.5  105 cells times with the blocking buffer. The nuclei were counterstained
per well, and then they were cultured and differentiated. All the with 40 -6-diamidino-2-phenylindole (DAPI) for visualization. For
culture experiments were divided into two groups with and with- CD34 staining, the process was the same as described above, ex-
out LIPUS. The cells in the groups with LIPUS were exposed to the cept that cells were incubated with the different primary antibody
pulsed ultrasound with an intensity of 200 mW cm–2, a duty cycle (Anti-CD34, 1:50, Beijing Biosynthesis Biotechnology Co. Ltd).
of 20% and a repetition rate of 1 MHz for 10 min each day. Microscopy observation was performed using an Olympus fluores-
cence microscope equipped with a digital camera and Image Pro-
2.4. In vitro endothelial and osteogenic differentiation of hMSCs plus. Fluorescent intensity was also used to quantitatively measure
the expression of CD31 and CD34 by using Image Pro-plus.
2.4.1. Cell culture for endothelial and osteogenic differentiation of
hMSCs
2.4.5. Osteogenic differentiation assay
Endothelial induction was performed using low glucose DMEM
At days 3, 7, 10 and 14 of osteogenic induction, the qualitative
(L-DMEM) supplemented with 10% FBS, 2 mM L-glutamine,
and quantitative examination of osteogenic differentiation of
100 U ml–1 antibiotic, 50 ng ml–1 vascular endothelial growth fac-
hMSCs was conducted by monitoring the activity of alkaline phos-
tor (VEGF165), 100 U ml–1 antibiotics and 10 ng ml–1 bone morpho-
phatase (ALP) using an alkaline phosphatase kit (Jiancheng Bioen-
genetic protein-2 (BMP-2). While osteogenic induction was
gineering Institute, Nanjing, China) as well as staining with a BCIP/
conducted using high glucose DMEM (H-DMEM) supplemented
NBT alkaline phosphatase kit (R&D Systems, Minneapolis, MN,
with 10% (v/v) FBS, 50 lM L-ascorbic acid, 10 mM b-glycerophos-
USA) according to the manufacturer’s instructions. Additionally,
phate, 0.1 lM dexamethasone and 0.1 mM dexamethasone. All
at days 14, 21 and 28 of osteogenic induction, mineralization dur-
the cells were cultured in a humidified atmosphere containing
ing osteogenic differentiation was analyzed by von Kossa staining
5% CO2 and 95% air at 37 °C. The medium was renewed every
according to the protocol described previously [27]. Before von
3 days [24,25].
Kossa staining, cells were rinsed twice with PBS and then fixed in
4% paraformaldehyde for 2 h at room temperature. In the mean-
2.4.2. Proliferation assay of cells during endothelial and osteogenic
time, calcium content was measured quantitatively using a Cal-
differentiation of hMSCs
cium C-Test kit (Wako Pure Chemical Industries, Ltd) as
The proliferation of cells was determined using the MTT
described by Takayama et al. [28].
(3-{4, 5-dimethylthiazol-2yl}-2, 5-diphenyl-2H-tetrazoliumbromide)
assay. The original medium was removed and 20 ll of 0.5% MTT
added to each well. Incubation of 4 h required a humidified 2.5. Statistical analysis
atmosphere containing 5% CO2 and 95% air at 37 °C until MTT
was taken up by active cells and reduced in the mitochondria to Each experiment was repeated three times. Statistical signifi-
insoluble purple formazan granules. Subsequently, the medium cances of all data were determined using Student‘s t-test. The dif-
was discarded and the precipitated formazan was dissolved in ference is considered significant if the p-value is less than 0.05.
dimethyl sulfoxide (50 ll per well). Optical density of the solution Indicated error bars correspond to the standard deviation (SD).
was evaluated using an ELISA reader at a wavelength of 570 nm.
Three wells were examined for each type sample each time. The 3. Results
investigated time points were days 1, 3, 7, 10, 14 and 21.
3.1. Observation of proliferative density and morphology of cells
2.4.3. Morphological observation during endothelial and osteogenic
differentiation of hMSCs Cell proliferation was determined by the optical density (OD)
Fluorescence staining using acridine orange (AO) was employed values of the MTT assay in our study. The OD values of the MTT
for cell morphological analysis in our study. After 2, 3 and 4 weeks assay for all the groups exhibited a similar increasing tendency
of induction, cells were rinsed twice with PBS and fixed in 4% para- over time, as shown in Fig. 2. It can be seen that cell proliferation
formaldehyde solution for 2 h at room temperature. The fixed cells was low during the first 3 days. The main reason for this might
were then incubated in 0.1 ml PBS containing 1% AO for 15 min at be because that hydrophilic hydrogel surfaces were not beneficial
37 °C and rinsed with PBS to remove excess dye. Microscopy obser- for the adsorption of proteins and this, in turn, was not conducive
vation was performed using an Olympus fluorescence microscope to the adhesion and growth of cells. It can also be seen that LIPUS
Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528 2521

Fig. 2. Cell proliferation by MTT assays on days 1, 3, 7, 10, 14 and 21 during (A) endothelial induction and (B) osteogenic induction. Results were presented as the mean ± SD,
and experiments were performed in triplicate. ⁄Significantly different (p < 0.05, n = 3) to the control group at the same time point; ⁄⁄significantly different (p < 0.01, n = 3) to
the control group at the same time point. The OSA/NSC groups, RGD–OSA/NSC groups and blank control groups are denoted as ON, R-ON and Blank, respectively, whilst these
groups with LIPUS are named as LIPUS-ON, LIPUS-R-ON and LIPUS-Blank, respectively.

led to lower cell proliferation during the first 3 days compared It is generally accepted that MSCs are mainly fusiform and
with the groups without LIPUS, demonstrating that LIPUS is not fibroblast-like cells and have the ability to differentiate into a vari-
beneficial for the early adsorption of adhesive proteins. However, ety of tissue cells. The morphologies of cells have a close relation-
the groups with RGD modification showed higher cell proliferation ship with stem cell differentiation and their mechanical properties.
compared with that in the control groups and the groups without Studies revealed that differentiation can result in the change of
RGD modification, indicating a positive effect of RGD on enhancing morphologies of cells while they, in turn, affect differentiation
the adhesion and growth of cells. A significant increase in cell pro- [29–33]. Thus, the change of morphologies of cells may be used
liferation was observed in all the groups from day 7 to day 14 for as evidence for differentiation of MSCs.
osteogenic induction and from day 7 to day 21 for endothelial After cell culture of 5 days, the proliferation of the passage cells
induction. The use of LIPUS or RGD promoted cell proliferation. reached up to 90% confluence. Their volume was larger than that of
From Table 1, it can be seen that the enhancement of cell prolifer- the primary cells and they had an irregular polygon shape, like
ation caused by the use of both LIPUS and RGD was much larger fibroblastic cells; their nuclei were oval or round and consisted
than that caused by LIPUS only plus that caused by RGD only, dem- of multiple nucleoli. After induction of 28 days, it was seen that
onstrating the synergistic effect of LIPUS and RGD on the enhance- most of the hMSCs presented totally different morphologies from
ment of the adhesion and growth of cells. The strongest synergistic those of mesenchymal stem cells, demonstrating that most of them
effect of LIPUS and RGD on the enhancement of cell proliferation have differentiated. Most cells for endothelial induction presented
occurred at day 14 for osteogenic induction and at day 21 for endo- a typical flat polygonal appearance, like endothelial cells, as
thelial induction. shown in Fig. 3A, while those for osteogenic induction exhibited

Table 1
Cell proliferations determined by means of the MTT assay during osteogenic (endothelial) differentiation.

Time (D) ON R-ON LIPUS-ON LIPUS-R-ON LIPUS-Blank


1 D-value 0.0001 (–0.0001) 0.0004 (0.0003) –0.0004 (–0.0007) –0.0003 (–0.0003) –0.0006 (–0.0006)
D/B (%) 1.08 (–1.09) 4.35 (3.26) –4.35 (–7.61) 3.26 (3.26) –6.52 (–6.52)
p 0.4915 (0.4840) 0.0601 (0.4299) 0.3718 (0.0185) 0.1930 (0.0015) 0.3536 (0.1190)
3 D-value 0.0003 (–0.0005) 0.0025 (0.001) –0.0057 (–0.0013) 0.0029 (0.0029) –0.0109 (–0.0006)
D/B (%) 0.17 (–0.28) 1.42 (0.56) –3.23 (–0.73) 1.64 (0.85) –6.18 (–0.34)
p 0.2569 (0.4681) 0.4642 (0.4680) 0.4105 (0.3661) 0.3986 (0.2614) 0.0098 (0.4123)
7 D-value –0.0082 (–0.0003) 0.0154 (0.0131) 0.1208 (0.1089) 0.1618 (0.2011) 0.1234 (0.1285)
D/B (%) –1.72 (–0.06) 3.23 (2.74) 25.33 (22.73) 33.93 (42.05) 25.88 (26.87)
p 0.0646 (0.4065) 0.1507 (0.0783) 0.0062 (0.0153) 0.0107 (0.0122) 0.0192 (0.0120)
10 D-value –0.0105 (–0.0105) 0.0254 (0.0154) 0.1080 (0.0982) 0.1903 (0.1805) 0.1450 (0.1068)
D/B (%) –1.31 (–1.30) 3.18 (1.91) 13.52 (12.16) 23.81 (22.36) 18.15 (13.23)
p 0.0919 (0.1102) 0.0114 (0.0117) 0.0013 (0.0019) 0.0009 (0.0004) 0.0042 (0.0003)
14 D-value –0.011 (–0.0118) 0.0229 (0.0406) 0.2079 (0.1656) 0.3298 (0.2284) 0.2703 (0.1912)
D/B (%) –1.36 (–1.30) 2.83 (4.46) 25.69 (18.21) 40.75 (25.11) 33.40 (21.02)
p 0.0235 (0.0986) 0.0042 (0.0120) 0.0003 (0.0015) <0.0001 (0.0002) 0.0018 (0.0038)
21 D-value –0.0197 (–0.016) 0.0088 (0.0323) 0.2080 (0.165) 0.3267 (0.2287) 0.2182 (0.1982)
D/B (%) –2.49 (–1.70) 1.11 (3.41) 26.26 (17.40) 41.24 (24.12) 23.69 (20.90)
p 0.0478 (0.2220) 0.0579 (0.1006) 0.0006 (0.0017) 0.0002 (0.0026) 0.0053 (0.0044)

B: The blank control group; D-value: the difference in the OD between the samples and the blank control group; p: p-value determined using Student’s t-test (n = 3).
2522 Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528

Fig. 3. Microscopic observation of the cell morphology that was stained with acridine orange (AO) after 28 days of (A) endothelial induction and (B) osteogenic induction.
Original magnification: 40; the insets represent a corresponding original magnification of 100.

spindle-shaped morphologies similar to osteoblasts, as shown in and RGD and it was much higher than the totality caused by LIPUS
Fig. 3B. Compared with the control groups, the cells in the groups and RGD alone shown in Table 2, indicating the synergistic effect of
with LIPUS grew more intensively and showed stronger character- LIPUS and RGD on the enhancement of the endothelial differentia-
istics of endothelial and osteoblasts cells, demonstrating that LIPUS tion of hMSCs.
was conducive to proliferation and differentiation of hMSCs.
3.3. Osteogenic induction
3.2. Endothelial induction
ALP activity is the most widely recognized biochemical marker
Angiogenesis is a process of endothelial migration and prolifer- for osteoblastic activity in the early stage. It is a typical protein for
ation to form a capillary network. Endothelial differentiation of osteoblast-phenotype and osteoblast differentiation [34].
MSCs is very important for vascularization in implant scaffolds Fig. 7A shows the results of ALP staining of the cultured cells at
when implanted. In our study, endothelial differentiation of hMSCs day 3, 7, 10 and 14. The expression of alkaline phosphatase was
on OSA/NSC and RGD–OSA/NSC hydrogels were conducted to eval- observed at day 7, which then increased with increasing the
uate their ability to induce vascularization. cultivation time and reached a peak by day 10 and then started
The immunofluorescence staining can provide a direct evidence to decrease [35]. The initial increase in ALP activity is a marker
for endothelial differentiation of the cells. The differentiated cells of the commitment towards osteoblastic lineage while the
can be detected for the expression of endothelial cell markers such subsequent decrease should be attributed to the formation of the
as CD31 and CD34 can be visualized by FITC-conjugated secondary advanced matrix mineralization and more mature phenotype
antibody by immunofluorescence staining. We chose two endothe- [36–40]. A repeatable positive expression of alkaline phosphatase
lial cell markers, CD31 and CD34, to assay the endothelial differen- in the groups with RGD could be observed, although it was not
tiation of hMSCs and found that all the cells expressed both specific significant from a statistical point of view; however, it was
endothelial markers of CD31 and CD34 at day 28 and could be visu- pronounced when LIPUS was used, indicating that there existed a
alized by FITC-conjugated secondary antibody, demonstrating that possible positive enhancement effect of RGD and LIPUS on the
the hMSCs had been successfully induced to differentiate into osteogenic differentiation of hMSCs. Further work needs to be done
endothelial cells (Figs. 4 and 5). The cells in the groups with LIPUS to prove it.
showed a much higher level of the positive expression of CD31 and The similar results were also detected in the measurement of
CD34 compared with those in the groups without LIPUS, revealing ALP activity in Fig. 7B and Table 3. The use of LIPUS led to a much
that the use of LIPUS is very useful for promoting the endothelial higher level of the ALP activity compared with that in the controls,
differentiation of hMSCs. Fluorescent intensity has been used to revealing that LIPUS did enhance the differentiation of cultured
quantitatively assay the expression of CD31 and CD34 (Fig. 6). Sim- hMSCs towards osteoblasts, as indicated by the increase in the
ilar results were observed. The expression of CD31 and CD34 was ALP activity. Moreover, it can be seen from Table 3 that the syner-
remarkably enhanced when LIPUS was used. The highest expres- gistic effect of LIPUS and RGD on the enhancement of the bone ma-
sion of CD31 and CD34 was detected in the group with both LIPUS trix proteins ALP was relatively low compared with proliferation of
Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528 2523

Fig. 4. Images of immunofluorescence staining of cells after endothelial induction of 28 days. The differentiated cells were detected for the expression of (red) endothelial cell
markers CD31 and visualized by FITC-conjugated secondary antibody. DAPI counter stained nuclei in blue. Original magnification: 200.

hMSCs and a high level in the synergistic effect was observed at The calcium content at different induction time points was
day 14. measured using a Calcium C-Test kit so as to quantitatively assay
Alizarin red S (AR-S) as a dye can bind selectively to calcium the formation of bone matrix. It can be seen that the change of cal-
salts and hence is widely used for calcium mineral histochemistry cium content presented the same tendency over time as Alizarin
and the assay of formation of new bone. This kind of histological red S staining shown in Fig. 8C and Table 4. It can be seen that
staining is based on the capacity of alizarin red to specifically stain the use of LIPUS or RGD could induce a higher level of calcium
matrix containing calcium and its positive appearance is consid- deposition compared with that in the controls, further revealing
ered an expression of bone matrix deposition [41]. The ability of that LIPUS and RGD can enhance the differentiation of cultured
cells to form mineralized matrix is essential with regard to the hMSCs towards osteoblasts, as indicated by the increase in the cal-
development of materials for the formation of new bone [41]. Nor- cium content. In comparison, LIPUS led to a much higher level of
mally, the measurement of ALP activity alone is not sufficient for calcium deposition and hence had a much stronger enhancement
determining bone formation. Hence, calcium nodulation is consid- effect on osteogenic induction than RGD. The synergistic effect of
ered as another specific marker of extracellular mineralized depos- LIPUS and RGD on the enhancement of calcium nodules was also
it in the late stage. We examined the formation of mineralized observed but was not very singnificant compared with the prolifer-
nodules in the presence and absence of transient LIPUS stimulation ation of hMSCs, as shown in Tables 1 and 4.
by staining with alizarin red, shown in Fig. 8A, and microscopy
observation of the living cells, shown in Fig. 8B. Alizarin red posi-
tive nodular aggregates have already been observed at day 14, indi- 4. Discussion
cating the formation of bone mineralization matrix in all the
groups. At the same time point, the number and size of Alizarin 4.1. The effect of RGD modification on the proliferation and
red positive nodular aggregates were larger in the groups with differentiation of hMSCs
RGD and with LIPUS compared with in the controls, demonstrating
the enhancement of both LIPUS and RGD on calcium deposition Due to good biocompatibility and the properties of degradation
and formation of bone matrix. The increased number and size of and release, as well as appropriate mechanical properties, hydro-
Alizarin red positive nodules with increasing induction time indi- gels have good prospects for use in biomedical applications. How-
cated that more and more calcium deposition had occurred. ever, hydrophilic surfaces for hydrogels are not beneficial for the
2524 Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528

Fig. 5. Images of immunofluorescence staining of cells after endothelial induction of 28 days. The differentiated cells were detected for the expression of (green) endothelial
cell markers CD34 and visualized by FITC-conjugated secondary antibody. DAPI counter stained nuclei in blue. Original magnification: 200.

Fig. 6. Quantitative analysis of fluorescence intensity of (A) CD31 and (B) CD34 weas performed and values are expressed as IOD. The results are presented as mean ± SD from
experiments performed in triplicate. ⁄Significantly different (p < 0.05, n = 3) to the control group at the same time point; ⁄⁄significantly different (p < 0.01, n = 3) to the control
group at the same time point. The OSA/NSC groups, RGD–OSA/NSC groups and blank control groups are denoted as ON, R-ON and Blank, respectively, whilst these groups with
LIPUS are named as LIPUS-ON, LIPUS-R-ON and LIPUS-Blank, respectively.
Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528 2525

Table 2
Integrated optical density (IOD) of images by CD31 (CD34) staining.

28D ON R-ON LIPUS-ON LIPUS-R-ON LIPUS-Blank


D-value D/B (%) p –37563 (–37518.6) 31481.7 (31509.5) 83754.4 (93800.9) 233853.5 (233936) 117030.8 (127092.4)
–14.98 (–14.90) 12.56 (12.52) 33.39 (37.26) 93.24 (92.93) 46.66 (50.49)
0.0058 (0.0003) 0.0002 (0.0025) <0.0001 (0.0014) 0.0001 (<0.0001) <0.0001 (0.0009)

B: The blank control group; D-value: the difference in the OD between the samples and the blank control group; p: p-value determined using Student’s t-test (n = 3).

Fig. 7. (A) ALP staining assay after osteogenic induction of 3, 7, 10 and 14 days. Original magnification: 100. (B) ALP activity assay after osteogenic induction of 3, 7, 10 and
14 days. Results are presented as mean ± SD from experiments were performed in triplicate. ⁄Significantly different (p < 0.05, n = 3) to the control group at the same time
point; ⁄⁄significantly different (p < 0.01, n = 3) to the control group at the same time point. The OSA/NSC groups, RGD–OSA/NSC groups and blank control groups are denoted
as ON, R-ON and Blank, respectively, whilst these groups with LIPUS are named as LIPUS-ON, LIPUS-R-ON and LIPUS-Blank, respectively.

Table 3
ALP activity assay of osteogenic induction.

Time (D) ON R-ON LIPUS-ON LIPUS-R-ON LIPUS-Blank


3 D-value – 0.0002 –0.0008 0.0026 0.0005
D/B (%) – 1.20 –1.30 4.21 0.81
p 0.4976 0.4914 0.4420 0.3338 0.4738
7 D-value –0.0038 0.0067 0.0316 0.0413 0.0338
D/B (%) –2.66 4.69 22.11 28.90 23.65
p 0.2732 0.1427 0.0132 0.0147 0.0081
10 D-value –0.0013 0.0066 0.0525 0.0618 0.0545
D/B (%) –0.46 2.31 18.41 21.68 19.11
p 0.4190 0.0877 <0.0001 0.0027 0.0026
14 D-value –0.0013 0.0040 0.0267 0.0364 0.0283
D/B (%) –0.71 2.18 14.56 19.85 15.43
p 0.1423 0.0558 0.0011 0.0059 0.0112

B: The blank control group; D-value: the difference in the OD between the samples and the blank control group; p: p-value determined using Student’s t-test (n = 3).

adsorption of protein and not conducive to the adhesion and in the 3-D dense collagen scaffolds can affect the degree of osteo-
growth of cells; hence these will limit the biomedical applications blastic cell proliferation, differentiation and some aspects of matrix
of hydrogels. The crucial factors for adherent cell survival are the remodeling activity [42]. Wang et al.’s study found that a higher
stability of the substrate’s adherent surface and its ability to sup- density of human umbilical cord mesenchymal stromal cells can
port cell adhesion. Hence, the processes of material functionaliza- result in better osteogenic differentiation and more matrix biosyn-
tion and modification have become very important in tissue thesis than lower densities [43]. Hence, we speculate that the
engineering. In this study, the modification of hydrogels by graft- promoted effect of RGD on the differentiation of hMSCs might be
ing adhesive molecules such as RGD onto them has been proved attributed to a high cell density caused by RGD.
to be a promising approach to overcome these limitations. The
results showed that the RGD modification of hydrogels could 4.2. The effect of LIPUS with 1 MHz and 200 mW cm–2 on the
improve the adhesion, growth and proliferation of hMSCs to a proliferation and differentiation of hMSCs
certain extent, although not more significant than expected. RGD
was also found to promote the differentiation of hMSCs in this LIPUS led to lower cell proliferation during the first 3 days com-
study. Moreira et al.’s study showed that the seeded cell population pared with the groups without LIPUS, indicating that LIPUS is not
2526 Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528

Fig. 8. (A) Images of mineralized nodule formation by staining with alizarin red S after osteogenic induction of 14, 21 and 28 days. Original magnification 100. (B) Images of
living cells after osteogenic induction of 28 days, showing mineralized nodule formation. Original magnification 400. (C) Quantitative analysis of calcium content after
osteogenic induction of 14, 21 and 28 days. Results are presented as the mean ± SD from experiments performed in triplicate. ⁄Significantly different (p < 0.05, n = 3) to the
control group at the same time point; ⁄⁄significantly different (p < 0.01, n = 3) to the control group at the same time point. The OSA/NSC groups, RGD–OSA/NSC groups and
blank control groups are denoted as ON, R-ON and Blank, respectively, whilst these groups with LIPUS are named as LIPUS-ON, LIPUS-R-ON and LIPUS-Blank, respectively.

Table 4
Mineralized nodule formation of osteogenic induction.

Time (D) ON R-ON LIPUS-ON LIPUS-R-ON LIPUS-Blank


14 D-value –0.1 0.4 1.1 2.0 1.2
D/B (%) –2.38 9.52 26.19 47.62 28.57
p 0.4075 0.0889 0.0030⁄⁄ 0.0162⁄ 0.0587
21 D-value –0.2 0.5 2.1 3.0 2.2
D/B (%) –2.06 5.15 21.65 30.93 22.68
p 0.0435 0.0189 0.0172 0.0044 0.0161
28 D-value –0.3 0.4 1.9 2.9 2.1
D/B (%) –2.04 2.72 12.93 19.73 14.29
p 0.3169 0.2346 0.0217 0.0101⁄ 0.0003

B: The blank control group; D-value: the difference in the OD between the samples and the blank control group; p: p-value determined using Student’s t-test (n = 3).

Significantly different (p < 0.05, n = 3) to the control group at the same time point.

Significantly different (p < 0.01, n = 3) to the control group at the same time point.

beneficial for the early adsorption of adhesive proteins. However, results showed that 1 MHz and 200 mW cm–2 might be a suitable
the enhancement of cell proliferation caused by LIPUS was frequency and intensity beneficial for the proliferation of cells.
detected after culture of 3 days and it was very strong. The present study shows that LIPUS can enhance osteogenic dif-
Whether LIPUS can promote cell proliferation or not remains ferentiation of hMSCs by stimulating ALP activity and promoting
debatable. Takayama et al. reported that transient LIPUS stimula- mineralized nodule formation. Similar results have been reported
tion (1.5 MHz, 30 mW cm–2, 20 min day–1) did not affect the rate elsewhere [16,46–50]. Warden et al. found that LIPUS could stim-
of cell proliferation [28]. Parvizi et al. reported that pulsed ultra- ulate the expression of the immediate early response genes c-fos
sound stimulates (1 kHz) had no effect on cell proliferation [44]. and COX-2, and elevate mRNA levels for the bone matrix proteins
However, Zhang et al. reported that LIPUS influenced chondrocyte ALP and OC, suggesting that LIPUS has a direct effect on bone for-
proliferation in an intensity-dependent manner [45]. We speculate mation [16]. They also found that the cells could be induced by the
that bio-effects of cells caused by LIPUS depends on ultrasound differentiation of hMSCs to osteoblasts in the absence of LIPUS.
dosage and ultrasound stimulates have no effect on cell prolifera- Hence, these suggest that soluble factors secreted by hMSCs might
tion under too low an intensity and frequency and can inhibit pro- induce the differentiation of hMSCs in vitro while LIPUS only
liferation of cells under too high an intensity and frequency. Our enhanced this process rather than triggering it. Chen et al. found
Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528 2527

Fig. 9. Schematic diagram (based on the model proposed by Chien [53]) showing the signaling pathways during the differentiation of hMSCs.

that bone marrow mesenchymal stem cells responded to LIPUS by RGD, cytokine and LIPUS, which may be regarded as different
increasing extracellular signal-regulated kinase phosphorylation ‘‘chemical ligands’’ responsible for binding with different receptors.
and enhancing osteogenic differentiation [49]. Jiang et al. Like chemical ligands, LIPUS can stimulate hMSCs by activating
investigated the osteogenic in vitro effect of LIPUS on SD rat adi- some special mechanosensors while these sensors can trigger sig-
pose-derived stem cells (ADSCs) and found that LIPUS-treated naling pathways to activate transcription factors to bind with the
ADSCs presented higher mRNA expression levels of runt-related appropriate cis elements so as to modulate the expression of the
transcription factor 2 (Runx2), osteocalcin (OCN), ALP and bone gene and then the protein [53]. The changes in gene and then pro-
sialoprotein (BSP) genes and higher protein levels of Runx2 and tein expressions, in turn, modulate the function of the hMSCs and
BSP than the controls, suggesting that LIPUS can also induce the finally result in the differentiation of hMSCs [53]. As the receptors
osteogenic differentiation of ADSCs in vitro [50]. These findings of RGD, integrins are responsible for not only regulating the attach-
are consistent with those observed by us. ment between cells and materials but also passing chemical and
In our study, the cells in the groups with LIPUS showed a much mechanical signals of the ECM into the cells to activate the signal-
higher level of the positive expression of CD31 and CD34 compared ing pathways [54,55]. During the differentiation of hMSCs, some of
with those in the groups without LIPUS, revealing that LIPUS can the same receptors might simultaneously respond to a number of
also promote the endothelial differentiation of hMSCs. Reher different ‘‘chemical ligands’’; for example, integrins respond not
et al.’s study demonstrated that ultrasound stimulates the produc- only to RGD but also to LIPUS. Moreover, LIPUS can increase the
tion of angiogenic factors such as IL-8, bFGF and VEGF [47]. Hence, permeability of the cell membrane and accelerate signal transmis-
we speculate that endothelial differentiation might be attributed sion and enhance the role of cytokines and RGD, promoting the dif-
to the non-thermal effects of LIPUS via a similar approach. ferentiation of hMSCs [56]. Hence, a positive synergistic effect of
Although in vitro investigations possess limitations, they do LIPUS and RGD can be expected in this process, as we have seen
provide important insight into the potential benefits of LIPUS and in our experiments.
the mechanisms by which these effects are produced for the
non-thermal effects of LIPUS [51,52]. We speculate that LIPUS
might stimulate the release of some growth factors and extracellu- 5. Conclusion
lar matrices (ECMs) from hMSCs during induction and as a result,
promoted the proliferation and differentiation of hMSCs. To further An RGD–OSA/NSC hydrogel was fabricated. An in vitro study of
investigate it, further in vitro study is required in the future. endothelial and osteogenic differentiation of marrow-derived
MSCs was conducted on hydrogels and the effect of RGD modifica-
4.3. Synergistic effect of LIPUS and RGD on the proliferation and tion and LIPUS on the proliferation and differentiation of hMSCs
differentiation of hMSCs was investigated. The results demonstrated that cell proliferation
was upregulated, and osteogenic and endothelial differentiation
In our study, LIPUS and RGD enhanced the proliferation and dif- of hMSCs was enhanced by the use of RGD modification and LIPUS,
ferentiation of hMSCs. This is a little-studied phenomenon. We which might be the result of LIPUS increasing the permeability of
found that the synergistic effect of LIPUS and RGD on the enhance- the cell membrane and accelerating signal transmission and
ment of proliferation of hMSCs was different to the effect on hMSC enhancing the role of cytokines and RGD. The synergistic effect
differentiation. We speculate that the synergistic effect on the of LIPUS and RGD on the enhancement of proliferation and
enhancement of proliferation is achieved by RGD promoting cell differentiation of hMSCs was observed. These findings suggest
proliferation to create a higher cell density and LIPUS stimulating the hybrid use of RGD modification and LIPUS might provide one
hMSCs to release ECMs, which further enhance the proliferation approach to achieve a high level of formation of new bone and
of hMSCs. Similarly, for the synergistic effect on the enhancement vascularization in bone tissue engineering scaffold implants.
of differentiation of hMSCs, the use of RGD can promote cell prolif-
eration to achieve a high cell density for differentiation, thus en-
abling LIPUS to stimulate more hMSCs to release growth factors Acknowledgements
to form a microenvironment with high concentrations of growth
factors where differentiation of hMSCs can be successfully induced. This work was supported by the National Basic Research Pro-
Based on the model proposed by Chien [53], possible signaling gram of China (973 Program, 2012CB933600), the National Natural
pathways of endothelial and osteogenic differentiation of hMSCs Science Foundation of China (No. 51072167 and No. 31370966),
were presented in Fig. 9. A series of signaling pathways plays a cru- Funds from the Health Department of Sichuan Province
cial role in the differentiation of hMSCs, where in our experiments (No. 110281) and Fundamental Research Funds for the Central
there are three kinds of sources of extracellular signaling including Universities (SWJTU11ZT05).
2528 Y. Wang et al. / Acta Biomaterialia 10 (2014) 2518–2528

References [30] Choi CB, Cho YK, Prakash KVB, Jee BK, Han CW, Paik YK, et al. Analysis of
neuron-like differentiation of human bone marrow mesenchymal stem cells.
Biochem Biophys Res Commun 2006;350:138–46.
[1] Seidi A, Ramalingam M, Elloumi-Hannachi I, Ostrovidov S, Khademhosseini A.
[31] Gade N, Pratheesh M, Nath A, Dubey P, Sharma B, Saikumar G, et al. Molecular
Gradient biomaterials for soft-to-hard interface tissue engineering. Acta
and cellular characterization of buffalo bone marrow-derived mesenchymal
Biomaterialia 2011;7:1441–51.
stem cells. Reprod Domestic Animals 2012;48:358–67.
[2] Thein-Han W, Misra R. Biomimetic chitosan–nanohydroxyapatite composite
[32] Zheng HZ, Liu C, Ou YF, Zhang YJ, Fu XK. Total saponins of Panax notoginseng
scaffolds for bone tissue engineering. Acta Biomaterialia 2009;5:1182–97.
enhance VEGF and relative receptors signals and promote angiogenesis
[3] Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design
derived from rat bone marrow mesenchymal stem cells. J Ethnopharmacol
variables and applications. Biomaterials 2003;24:4337–51.
2013;147:595–602.
[4] He FP, Ye JD. In vitro degradation, biocompatibility, and in vivo osteogenesis of
[33] Kim BS, Kim JS, Chung YS, Sin YW, Ryu KH, Lee J, et al. Growth and osteogenic
poly(lactic-co-glycolic acid)/calcium phosphate cement scaffold with
differentiation of alveolar human bone marrow-derived mesenchymal stem
unidirectional lamellar pore structure. J Biol Mater Res 2012;100A:3239–50.
cells on chitosan/hydroxyapatite composite fabric. J Biol Mater Res
[5] Boccaccini AR, Kneser U, Arkudas A. Scaffolds for vascularized bone
2013;101:1550–8.
regeneration: advances and challenges. Expert Rev Med Dev 2012;9:457–60.
[34] Ehrlich PJ, Lanyon LE. Mechanical strain and bone cell function: a review.
[6] Le Visage C, Gournay O, Benguirat N, Hamidi S, Chaussumier L, Mougenot N,
Osteoporos Int 2002;13:688–700.
et al. Mesenchymal stem cell delivery into rat infarcted myocardium using a
[35] Magnusson P, Larsson L, Magnusson M, Davie MW, Sharp CA. Isoforms of bone
porous polysaccharide-based scaffold: a quantitative comparison with
alkaline phosphatase: characterization and origin in human trabecular and
endocardial injection. Tissue Eng part A 2012;18:35–44.
cortical bone. Bone Miner Res 1999;14:1926–33.
[7] Urist MR. Bone: formation by autoinduction. Science 1965;150:893–9.
[36] Anderson H. Mechanism of mineral formation in bone. J technical methods and
[8] Dzau VJ, Gnecchi M, Pachori AS, Morello F, Melo LG. Therapeutic potential of
pathology. 1989;60:320–30.
endothelial progenitor cells in cardiovascular diseases. Hypertension
[37] Gerstenfeld LC, Chipman SD, Glowacki J, Lian JB. Expression of differentiated
2005;46:7–18.
function by mineralizing cultures of chicken osteoblasts. Dev Biol 1987;122:
[9] Griffon DJ, Sedighi MR, Schaeffer DV, Eurell JA, Johnson AL. Chitosan scaffolds:
49–60.
interconnective pore size and cartilage engineering. Acta Biomaterialia
[38] Sugawara Y, Suzuki K, Koshikawa M, Ando M, Iida J. Necessity of enzymatic
2006;2:313–20.
activity of alkaline phosphatase for mineralization of osteoblastic cells.
[10] Whang K, Healy KE, Elenz DR, Nam EK, Tsai DC, Thomas CH, et al. Engineering
Japanese J Pharmacol 2002;88:262–9.
bone regeneration with bioabsorbable scaffolds with novel microarchitecture.
[39] Aubin J, Liu F, Malaval L, Gupta A. Osteoblast and chondroblast differentiation.
Tissue Eng 1999;5:35–51.
Bone 1995;17:S77–83.
[11] Rezwan K, Chen QZ, Blaker JJ, Boccaccini AR. Biodegradable and bioactive
[40] Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP. Osteogenic differentiation of
porous polymer/inorganic composite scaffolds for bone tissue engineering.
purified, culture-expanded human mesenchymal stem cells in vitro. J Cell
Biomaterials 2006;27:3413–31.
Biochem 1997;64:295–312.
[12] Li ZS, Ramay HR, Hauch KD, Xiao DM, Zhang MQ. Chitosan–alginate hybrid
[41] Wang D, Haile A, Jones LC. Dexamethasone-induced lipolysis increases the
scaffolds for bone tissue engineering. Biomaterials 2005;26:3919–28.
adverse effect of adipocytes on osteoblasts using cells derived from human
[13] Rogalsky AD, Kwon HJ, Lee-Sullivan P. Compressive stress–strain response of
mesenchymal stem cells. Bone 2013;53:520–30.
covalently crosslinked oxidized-alginate/N-succinyl-chitosan hydrogels. J
[42] Moreira SM, Andrade FK, Domingues L, Gama M. Development of a strategy to
Biomed Mater Res A 2011;99:367–75.
functionalize a dextrin-based hydrogel for animal cell cultures using a starch-
[14] Köllmer M, Keskar V, Hauk TG, Collins JM, Russell B, Gemeinhart RA. Stem cell-
binding module fused to RGD sequence. BMC Biotechnol 2008;8:78.
derived extracellular matrix enables survival and multilineage differentiation
[43] Wang L, Dormer NH, Bonewald LF, Detamore MS. Osteogenic differentiation of
within superporous hydrogels. Biomacromolecules 2012;13:963–73.
human umbilical cord mesenchymal stromal cells in polyglycolic acid
[15] Spadaro JA, Albanese SA. Application of low-intensity ultrasound to growing
scaffolds. Tissue Eng 2010;16:1937–48.
bone in rats. Ultrasound Med Biol 1998;24:567–73.
[44] Parvizi J, Wu CC, Lewallen DG, Greenleaf JF, Bolander ME. Low-intensity
[16] Warden SJ, Favaloro JM, Bennell KL, McMeeken JM, Ng KW, Zajac JD, et al. Low-
ultrasound stimulates proteoglycan synthesis in rat chondrocytes by
intensity pulsed ultrasound stimulates a bone-forming response in UMR-106
increasing aggrecan gene expression. J Orthop Res 1999;17:488–94.
cells. Biochem Biophys Res Commun 2001;286:443–50.
[45] Zhang ZJ, Huckle J, Francomano CA, Spencer RG. The effects of pulsed low-
[17] Naruse K, Mikuni-Takagaki Y, Azuma Y, Ito M, Oota T, Kameyama K, et al.
intensity ultrasound on chondrocyte viability, proliferation, gene expression
Anabolic response of mouse bone-marrow-derived stromal cell clone ST2 cells
and matrix production. Ultrasound Med Biol 2003;29:1645–51.
to low-intensity pulsed ultrasound. Biochem Biophys Res Commun
[46] Li L, Yang Z, Zhang H, Chen W, Chen M, Zhu Z. Low-intensity pulsed ultrasound
2000;268:216–20.
regulates proliferation and differentiation of osteoblasts through osteocytes.
[18] Ito M, Azuma Y, Ohta T, Komoriya K. Effects of ultrasound and 1,25-
Biochem Biophys Res Commun 2012;418:296–300.
dihydroxyvitamin D3 on growth factor secretion in co-cultures of
[47] Reher P, Harris M, Whiteman M, Hai H, Meghji S. Ultrasound stimulates nitric
osteoblasts and endothelial cells. Ultrasound Med. Biol 2000;26:161–6.
oxide and prostaglandin E2 production by human osteoblasts. Bone
[19] Sun S, Wang A. Adsorption properties of N-succinyl-chitosan and cross-linked
2002;31:236–41.
N-succinyl-chitosan resin with Pb(II) as template ions. Sep Purif Technol
[48] Korstjens C, Nolte P, Burger E, Albers G, Semeins C, Aartman I, et al. Stimulation
2006;51:409–15.
of bone cell differentiation by low-intensity ultrasound – a histomorphometric
[20] Yamaguchi R, Araj Y, Itoh T, Hirano S. Preparation of partially N-succinylated
in vitro study. J Orthop Res 2004;22:495–500.
chitosans and their cross-linked gels. Carbohyd Res 1981;88:172–5.
[49] Chen YJ, Wang CJ, Yang KD, Chang PR, Huang HC, Huang YT, et al. Pertussis
[21] Fan LH, Pan XR, Zhou Y, Chen LY, Xie WG, Long ZH, et al. Preparation and
toxin-sensitive GKi protein and ERK-dependent pathways mediate ultrasound
characterization of crosslinked carboxymethyl chitosan-oxidized sodium
promotion of osteogenic transcription in human osteoblasts. FEBS Lett
alginate hydrogels. J App Poly Sci 2011;122:2331–7.
2003;554:154–8.
[22] Gao CM, Liu MZ, Chen J, Zhang X. Preparation and controlled degradation of
[50] Jiang T, Xu T, Gu F, Chen A, Xiao Z, Zhang D. Osteogenic effect of low intensity
oxidized sodium alginate hydrogel. Polym Degrad Stabil 2009;94:1405–10.
pulsed ultrasound on rat adipose-derived stem cells in vitro. J Huazhong Univ
[23] Liu X, Peng WZ, Wang YY, Zhu MH, Sun T, Peng Q, et al. Synthesis of an RGD-
Sci Technol-Med Sci 2012;32:75.
grafted oxidized sodium alginate–N-succinyl chitosan hydrogel and an in vitro
[51] Chen SH, Chiu CY, Yeh JM, Wang SH, Effect of low intensity ultrasounds on the
study of endothelial and osteogenic differentiation. J Mater Chem B 2013;1:
growth of osteoblasts, Engineering in Medicine and Biology Society, 2007.
4484–92.
EMBS 2007. 29th Annual International Conference of the IEEE. IEEE, 2007,
[24] Cao Y, Sun Z, Liao LM, Meng Y, Han Q, Zhao RCH. Human adipose tissue-derived
5833-5836.
stem cells differentiate into endothelial cells in vitro and improve postnatal
[52] Doan N, Reher P, Meghji S, Harris M. In vitro effects of therapeutic ultrasound
neovascularization in vivo. Biochem Biophys Res Commun 2005;332:370–9.
on cell proliferation, protein synthesis, and cytokine production by human
[25] Schneider RK, Puellen A, Kramann R, Raupach K, Bornemann J, Knuechel R,
fibroblasts, osteoblasts, and monocytes. J Oral Maxillofac Surg 1999;57:
et al. The osteogenic differentiation of adult bone marrow and perinatal
409–19.
umbilical mesenchymal stem cells and matrix remodelling in three-
[53] Chien S. Mechanotransduction and endothelial cell homeostasis: the wisdom
dimensional collagen scaffolds. Biomaterials 2010;31:467–80.
of the cell. Am J Physiol Heart Circ Physiol 2007;292:H1209–24.
[26] Wang MK, Su YP, Sun HQ, Wang T, Yan GH, Ran XZ, et al. Induced endothelial
[54] Chun C, Lim HJ, Hong K-Y, Park K-H, Song S-C. The use of injectable,
differentiation of cells from a murine embryonic mesenchymal cell line C3H/
thermosensitive poly (organophosphazene)–RGD conjugates for the
10T1/2 by angiogenic factors in vitro. Differentiation 2010;79:21–30.
enhancement of mesenchymal stem cell osteogenic differentiation.
[27] Zhang W, Yang N, Shi XM. Regulation of mesenchymal stem cell osteogenic
Biomaterials 2009;30:6295–308.
differentiation by glucocorticoid-induced leucine zipper (GILZ). J Biol Chem
[55] Taubenberger AV, Woodruff MA, Bai H, Muller DJ, Hutmacher DW. The effect
2008;283:4723–9.
of unlocking RGD-motifs in collagen I on pre-osteoblast adhesion and
[28] Takayama T, Suzuki N, Ikeda K, Shimada T, Suzuki A, Maeno M, et al. Low-
differentiation. Biomaterials 2010;31:2827–35.
intensity pulsed ultrasound stimulates osteogenic differentiation in ROS 17/
[56] Dinno M, Dyson M, Young S, Mortimer A, Hart J, Crum L. The significance of
2.8 cells. Life Sci 2007;80:965–71.
membrane changes in the safe and effective use of therapeutic and diagnostic
[29] Zeng R, Wang LW, Hu ZB, Guo WT, Wei JS, Lin H, et al. Differentiation of human
ultrasound. Phys Med Biol 1989;34:1543.
bone marrow mesenchymal stem cells into neuron-like cells in vitro. Spine
2011;36:997–1005.

You might also like