He 2019

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Received: 21 February 2019 Revised: 10 May 2019 Accepted: 13 May 2019

DOI: 10.1002/jbm.a.36723

ORIGINAL ARTICLE

An injectable scaffold based on temperature-responsive


hydrogel and factor-loaded nanoparticles for application in
vascularization in tissue engineering

Dan He1 | An-Sha Zhao1 | Hong Su1 | Yan Zhang3 | Ya-Nan Wang1 | Dan Luo1 |
Yuan Gao1 | Jing-An Li1,2 | Ping Yang1

1
Key Laboratory for Advanced Technologies of
Materials, School of Material Science and Abstract
Engineering, Southwest Jiaotong University, Controlled release of functional factors contributes to target migration of therapeutic
Ministry of Education, Chengdu, People’s
Republic of China cells and plays a crucial role in the in situ vascularization of tissue repair and regener-
2
School of Material Science and Engineering, ation. A biomedical application requires the selective release of multiple factors
Zhengzhou University, Zhengzhou, People’s
which will guide the synergy of the cells. Here, we developed an injectable system
Republic of China
3
Department of Cardiology, Chengdu Military based on a temperature-responsive hydrogel and stromal cell-derived factor-1 (SDF-
General Hospital, Chengdu, People’s Republic 1)/vascular endothelial growth factor (VEGF) loaded into two types of nanoparticles
of China
to induce migration and rapid proliferation of mesenchymal stem cells (MSCs) and
Correspondence endothelial cells (ECs) via selective SDF-1/VEGF release. Series of in vitro and in vivo
An-Sha Zhao and Jing-An Li, Key Laboratory
for Advanced Technologies of Materials, experiments demonstrate that our composited system can accurately guide MSCs
School of Material Science and Engineering, and ECs for vascularization. In addition, the properties of the nanoparticles and
Southwest Jiaotong University, Ministry of
Education, Chengdu 610031, People’s hydrogel, including micro/nanoscales, characteristic of charge, and biocompatibility,
Republic of China. played crucial roles for the selective release and cells behavior (target migration and
Emails: anshazhao@home.swjtu.edu.cn,
4828713@qq.com rapid proliferation).

Funding information KEYWORDS


Key Project and Special Foundation of
hydrogels, nanoparticles, selective release, tissue engineering scaffolds, vascularization
Research, Development and Promotion in
Henan province, Grant/Award Number:
182102310076; National Natural Science
Foundation of China, Grant/Award Number:
NSFC 81771988; The Joint Fund for Fostering
Talents of NCIR-MMT & HNKL-MMT, Grant/
Award Number: MMT2017-01; Top Doctor
Program of Zhengzhou University, Grant/
Award Number: 32210475

1 | I N T RO D UC T I O N However, only a few engineered tissues or organs have clinical appli-


cation. The successful examples, such as skin, cartilage, and trachea,
Nowadays, tissue engineering is widely recognized as a hot topic in are thin-walled tissues that require less vascularization; the thicker
the field of biomedical science, and it is playing more and more crucial essential organs, such as the liver and kidney, require a functional vas-
roles for organ/tissue repair and replacement (Caccavo, Cascone, cular network to achieve graft survival in vivo (Griffith & Naughton,
Lamberti, & Barba, 2018). Current technologies have achieved the 2002). Thus, insufficient vascularization has become a bottleneck hin-
aims of implanting the biomaterials loaded with the massive in vitro dering the clinical application of the tissue engineering.
cultured cells into the human body in order to restore, replace, main- Stem cells homing and vascular endothelial cells (VECs) target
tain, or improve the function of tissues or organs (Zhang et al., 2018). migration, as well as their rapid proliferation in situ play important

J Biomed Mater Res. 2019;107A:2123–2134. wileyonlinelibrary.com/journal/jbma © 2019 Wiley Periodicals, Inc. 2123
2124 HE ET AL.

parts in the vascularization process: wherein stem cells homing, rapid chitosan solution. The fucoidan solution 0.05% (w/v) was prepared in
proliferation, and oriented differentiation mainly contribute to the distilled water (dH2O) in the same way. The nanoparticles were
regeneration of vascular tissue structure. VECs migration and prolifer- obtained by mixing chitosan and fucoidan solutions and then collected
ation are inclined to vascular functions like relaxation, patency, and at 14,000 rpm for 4 min. Nanoparticles prepared by chitosan and
signal molecule release (e.g., nitric oxide and prostatic cyclin), and so fucoidan with volume ratios of 5:5 were labeled as C5F5 and selected
forth (Laiva, Raftery, Keogh, & O'Brien, 2018; Li et al., 2014). Stromal for loading SDF-1 (the SDF-1-loaded C5F5 was labeled as C5F5@SDF-
cell-derived factor-1 (SDF-1) is a cytokine with small molecule weight, 1), while nanoparticles prepared by chitosan and fucoidan with vol-
which affects homing of mesenchymal stem cells (MSCs) to the lesion ume ratios of 2:5 were labeled as C2F5 and selected for loading VEGF
location via SDF-1/CXCR4 axis (Park, Lee, Kim, Park, & Song, 2018). (the VEGF-loaded C2F5 was labeled as C2F5@VEGF).
Vascular endothelial growth factor (VEGF) is a specific protein which The chemical compositions of the C5F5 and C2F5 were examined
promotes the VECs migration and proliferation, and it is also the pref- by transmission mode of Fourier-transform infrared (FTIR) spectrome-
erable factor on inducing angiogenesis in vivo (Hu et al., 2018). Cer- ter (ST-IR20SX, Nicolet, Madison, Wisconsin, US) after freeze-drying
tain studies also suggest a positive contribution of VEGF to the for 12 hr. The particle size, zeta potential, and polymer dispersity
oriented differentiation of MSCs in the specific microenvironment, index (PDI) of C5F5, C2F5, C5F5@SDF-1, and C2F5@VEGF were mea-
such as bone, vascular, and nervous (Lü, Deegan, Musa, Xu, & Yang, sured by a nanoscale laser particle size analyzer (Zetasizer Nano-
2018; Luo, Zhang, Zhang, & Jin, 2012; Wang et al., 2018). ZS90, Malvern Ltd, United Kingdom). The microstructures of C5F5,
Hence, loading and sustained release of SDF-1 and VEGF at the C2F5, C5F5@SDF-1, and C2F5@VEGF nanoparticles were observed by
focus of the lesion are effective methods for in situ vascularization in transmission electron microscopy (TEM, JEM-2100F, Hitachi, Japan).
tissue engineering scaffolds. Yet, the traditional method of direct load-
ing of the factors into the scaffolds has a disadvantage of uncontrolled
2.2 | Preparation and characterization of the
release, which may result in disturbed biological response or loss of
temperature-responsive hydrogel
function of the loaded factors (Davoodi et al., 2018).
Nanoparticles have been successfully used as carriers for the Chitosan was dissolved in 0.1 mol/L aqueous acetic acid at room temper-
delivery of bioactive substances to cells and in tissue engineering (Liu, ature (RT) using a magnetic stirrer (85-1, Shanghai Sile Instrument Co.,
Jiang, & Hunziker, 2016). Co-loading of nanoparticles with molecules Ltd, China) for 5 h to obtain a 2.2% (w/v) solution. Gelatin (Sigma-Aldrich,
of different charge, VEGF (negative charge), and SDF-1 (positive Germany) was dissolved in the dH2O at 60 C to give a 1% solution (w/v).
charge) can improve the controllable and selective release of the β-Glycerophosphate (Sigma) was also dissolved in the dH2O at RT to give
whole delivery system. The development of environmental-responsive a 56% (w/v) solution. Gelatin solution was introduced to the chitosan
hydrogels with response to temperature, pH, enzymes, and so forth solution with a volume ratio of 1:10, then the β-glycerophosphate solu-
allows for better targeting of tissue engineering scaffolds (Bao et al., tion was added drop by drop with stirring on ice. The volume ratio of the
2017; Tang et al., 2017; Zhong et al., 2018). In particular, injectable chitosan solution and the β-glycerophosphate solution was 5:1. After this
hydrogel scaffolds may avoid surgical trauma and infection as much as process, the hydrogel was obtained.
possible, to further reduce the risk of surgery, and simultaneously The entire hydrogel at 20 C and 37 C was directly inspected and
improve in situ the precision of the gel at the focus of the lesion photographed with a Huawei Mate10 camera equipment. A stress-
(Rose & Laura, 2018). controlled rheometer (DRH-1, TA Instrument) was applied to detect
In this contribution, we developed an injectable hydrogel with the the storage modulus (G0 ), loss modulus (G00 ), phase angle, and complex
natural cationic polysaccharide chitosan, sodium β-glycerophosphate, viscosity of the hydrogel during the gelatination process. A temperature
and gelatin as a matrix, to co-load chitosan/fucoidan/VEGF nano- scan with a heating rate of 2/min from 17 C to 90 C was performed,
particles and chitosan/fucoidan/SDF-1 nanoparticles for in situ vascu- the analysis frequency was 1 rad/s, and the change curves of the G0 ,
larization. This composite system was temperature responsive and G00 , phase angle, and complex viscosity were recorded; the inter-
improved MSCs homing/VEC migration as well as their proliferation. section of G0 and G00 is defined as gelation temperatures (Tgel, Chenite,
The chick chorioallantoic membrane (CAM) model was applied to 2000). The morphology of the gelling hydrogel was observed by scan-
observe the vascular regeneration via injecting the composite system. ning electron microscopy (QUANTA200, Holland FEI, The Netherlands)
We hope this study may provide a novel research direction for regener- after freeze-drying and gold spraying (Zhang, Shi, et al., 2018).
ative medical biomaterials that require vascularization in situ during
implantation.
2.3 | SDF-1 and VEGF release from the
nanocomposite system
2 | MATERIALS AND METHODS
C5F5@SDF-1 and C2F5@VEGF nanoparticles were distributed into the
hydrogel to prepare the nanocomposite system (the concentration of
2.1 | Nanoparticles preparation and characterization
both SDF-1 and VEGF solution was 50 ng/mL, the samples were
Chitosan (degree of deacetylation 9 ≥95%; Aladdin) was added to labeled as hydrogel@NPVEGF&SDF-1), and then placed onto a desktop
0.2% (v/v) aqueous acetic acid with stirring to get a 0.05% (w/v) concentrator at 37 C for gelling. A control was prepared with SDF-1
HE ET AL. 2125

and VEGF directly introduced into the hydrogel, labeled as hydro- investigation, the fifth passage of the HUVEC was seeded onto the
gel@VEGF&SDF-1 (the concentration of both SDF-1 and VEGF solu- samples with a density of 5 × 104 cells/mL and then incubated at
tion was also 50 ng/mL). Hydrogels loaded with single C5F5@SDF-1 37 C for 1 and 3 days; after fixation with 4% paraformaldehyde
and C2F5@VEGF were also fabricated as control samples, and they (Sigma) for 2 hr at RT, the HUVECs of each group were stained by
were labeled as hydrogel@NPSDF-1 and hydrogel@NPVEGF, respectively. phalloidin (Sigma) and for 15 min and 40 ,6-diamino-2-phenylindole
All the gelling hydrogels were immersed in phosphate buffer solu- (DAPI, Sigma) for 5 min, and finally observed by a fluorescence micro-
tion (PBS, pH 7.4) at 37 C and the concentration of the released SDF-1 scope (DMRX, Leica, Germany). A typical CCK-8 assay was performed
and VEGF from each hydrogel was detected via a typical enzyme-linked to investigate the HUVEC proliferation on each sample (Chen et al.,
immuno sorbent assay at the first, third, fifth, and seventh days, respec- 2018). The HUVEC migration experiment was performed according to
tively (Li et al., 2015). the protocol of MSCs homing which was described in Section 2.4.
To investigate the apoptosis of HUVEC of each group, cell-

2.4 | MSCs homing and proliferation permeable AO) dye in combination with propidium iodide (PI), a
plasma membrane-impermeable Deoxyribonucleic acid (DNA)-binding
The MSCs were isolated from bone marrow of stromal cell-derived dye, was applied. AO and PI excite green and orange-red fluores-
rats as described in our previous work and its third passage was cence, respectively, after intercalation into DNA. The HUVEC apopto-
applied for investigating the ability of each sample on improving hom- sis assay was carried out as follows: after cultured for 1 and 3 days,
ing and proliferation (Li et al., 2016). The protocol of MSCs homing
respectively, the cells were stained with a 1:1 mixture of AO
was carried out using a Millipore transwell (pore size: 8.0 μm). In this
(100 mg/mL) and PI (100 mg/mL) at 37 C for 3 min, and then immedi-
experiment, 200 μL of each hydrogel was filled in a 12-well plate and
ately inspected with a fluorescence microscope. The vital ratios of
incubated at 37 C for gelling. For the test, fresh culture medium was
HUVEC on each sample were calculated from at least 15 fields
initially added to the hydrogels in the 12-well plate (700 μL/well). The
(Li et al., 2017).
transwell was subsequently inserted into the plate, and 500 μL of
MSCs suspension (5 × 104 cells/mL) was added. After culturing at
37 C and 5% CO2 for 12 hr, the MSCs adhering to the upper surface 2.6 | Establishment of Chick embryo CAM model
of the insert were removed using a swab, and the MSCs across the
Chick embryo CAM is a thin and transparent membrane attached to
bottom and those that had migrated to the undersurface were stained
the eggshell, and it will be in large quantities during the development of
using 0.1% crystal violet (Liu et al., 2017). The homing MSCs were
chick embryos. The CAM model was used in the study of angiogenesis
observed using optical microscopy (Carl Zeiss A2). The numbers of the
for the first time by Auerbach, Kubai, Knighton, & Folkman, (1974) for
homing MSCs of each group were counted from at least 15 visual
CAM takes on abundant blood vessels, enabling observation of micro-
fields (Li et al., 2015).
vascular structure changes without special equipment (Auerbach et al.,
For the MSCs proliferation assay, the third passage of MSCs was
1974). In this study, we applied the CAM model to investigate the con-
suspended in the hydrogel@NPVEGF&SDF-1, hydrogel@VEGF&SDF-1,
tribution of each hydrogel group to the angiogenesis. In brief, fertilized
hydrogel@NPSDF-1, hydrogel@NPVEGF, and the single hydrogel con-
chicken eggs (ChengDu Muxing Poultry Industry Limited Liability Com-
trol, respectively, with a concentration of 5 × 104 cells/mL at 20 C,
pany, China) were incubated at the standard conditions of 37 C and
and cultured using fresh Dulbecco's Modified Eagle Media/Nutrient
55% humidity and turned 3–5 times daily. After 7 days of incubation,
Mixture F-12 (DMEM/F12) complete medium (with 10% fetal bovine
the eggs were gently cleaned with a 2% benzalkonium bromide solu-
serum) at 37 C in a humidified atmosphere containing 95% air and 5%
tion, and a 1.5 cm × 1.5 cm hole was opened at the end of the chick
CO2. Cells were fed with freshly prepared growth medium every
24 hr. After culturing for 1 and 3 days, the MSCs-loaded hydrogels embryo chamber by tweezers to remove eggshell and inner eggshell

were stained with acridine orange (AO) solution (DMEM/F12 = 1:50) membrane. Then, the gas chamber membrane was picked up by twee-

at 37 C for 10 min and observed under a confocal laser scanning zers, the upper chamber membrane was removed, and the CAM film on

microscope (CLSM, Nikon C2 Plus, Japan) in Z-axis scanning mode to the lower layer was finally exposed. An amount of 100 μL of hydroge-

investigate the MSCs proliferation, and a Cell Counting Kit-8 (CCK-8) l@NPVEGF&SDF-1, hydrogel@VEGF&SDF-1, hydrogel@NPSDF-1, hydro-
assay was also applied for the quantitative characterization (Zhang, gel@NPVEG, and the single hydrogel control were placed on the large
Shi, et al., 2018). vascular branches of the CAM (each group has five parallel CAM, n = 5)
at 20 C; the hole was sealed with medical wound dressing and the eggs
were returned to the incubator for continuous culture. After 3 days cul-
2.5 | Endothelial cells migration, proliferation, and
ture, 4% paraformaldehyde was added from the hole for a 15-min fixed
apoptosis
step, and then the CAM covered by the hydrogels were cut off and
In this experiment, the 12-well plate was also modified with each rinsed with PBS. Paraffin-embedded sections were stained with
hydrogel as descripbed in Section 2.4. Human umbilical vein endothe- hematoxylin-eosin (HE) to observe each hydrogel of blood vessel
lial cells (HUVECs) obtained from newborn umbilical cord (Huaxi Hos- ingrowth (Zou et al., 2018). The operation process of the CAM model is
pital, Chengdu, China) were applied in this study: for the proliferation displayed in Figure 1.
2126 HE ET AL.

F I G U R E 1 The operation process of


the chorioallantoic membrane (CAM)
model aiming at studying angiogenesis

2.7 | Statistical analysis indicates that both the factors-loaded nanoparticles (C5F5@SDF-1
and C2F5@VEGF) and the single nanoparticles (C5F5 and C2F5) had
The data were statistically evaluated using ANOVA by homogeneity
moderately stable structure and distribution because the absolute
test of variances firstly, and the post hoc test was subsequently per-
values of their zeta potentials were more than 30 mV, and this param-
formed using the least significant difference method for comparison.
eter point is considered as a minimum value for the stability of
The data are expressed as mean ± SD. The probability value p < 0.05
nanoparticles. As Liu et al., (2017) reported, PDI is an important indi-
was considered as a significant difference. The data analysis was car-
cator to evaluate the size distribution of the particles, and PDI values
ried out using the software SPSS 11.5 (Chicago, IL).
smaller than 0.2 indicate good uniformity. The C5F5@SDF-1 and
C2F5@VEGF not only showed PDI values below 0.2 but also
3 | RESULTS maintained their original charge property (C5F5@SDF-1: positive
charge; C2F5@VEGF: negative charge), which may contribute to their
3.1 | Characterization of the factor-loaded better uniformity and orderly delivery.
chitosan/fucoidan nanoparticles
To confirm the successful preparation of the chitosan/fucoidan 3.2 | Characterization of the temperature-responsive
nanoparticles, the chemical structures of chitosan/fucoidan nanoparticles, hydrogels
as well as the chitosan and fucoidan controls, were analyzed by FTIR Figure 3a displays the entire chitosan/gelatin/β-glycerophosphate
spectrophotometry. As shown in Figure 2a, the chitosan/fucoidan hydrogel at different temperatures: the hydrogel presented as a limpid
nanoparticles, chitosan, and fucoidan controls had a peak around solution at 20 C, and when the temperature went up to 37 C, it
1,560/cm ascribed to the bending vibration of N─H, while the converted into a milky white colloid. Figure 3b shows the dynamic vis-
chitosan/fucoidan nanoparticles and the chitosan control presented a coelasticity of thermosensitive hydrogel: G0 represents elastic proper-
characteristic peak at 1150/cm ascribed to the stretching vibration of ties, G00 represents viscous properties, and the tangent of the phase
C─O─C. Only the chitosan/fucoidan nanoparticles and the fucoidan angle equals to G00 /G0 and represents the degree of viscoelasticity of
control displayed the peaks at 1160–1260/cm (stretching vibration of the system. At 37 C, the hydrogel became solid, its G00 was less than G0
S─O) and 845/cm (stretching vibration of C─O─S), indicating that the and it, therefore, had a lower phase angle that indicated elastic property
chitosan/fucoidan nanoparticles were successfully prepared. in the range of frequency measurement. Figure 3c presented the G0
The formation of chitosan/fucoidan nanoparticles was mainly and G00 changes of the thermosensitive hydrogel during the tempera-
driven by the electrostatic interaction between negatively charged ture rise: G0 reflects the solid-state properties of the hydrogel's rheolog-
fucoidan and positively charged chitosan. The charge properties of ical properties, while G00 reflects the fluid properties of the hydrogel's
the chitosan/fucoidan nanoparticles could be controlled by changing rheological properties. So, at the low temperature, the G0 maintained a
the ratios of these two molecules, wherein the C5F5 nanoparticles relatively low value, while it rose rapidly when the temperature went
were positively charged and selected for loading SDF-1 (negative up to a certain point, and G0 even exceeded G00 as the temperature con-
charge), while the C2F5 nanoparticles possessed negative charge and tinued to rise. The intersection point of G00 and G00 is defined as gelation
were selected for loading VEGF (positive charge). Figure 2b–d temperatures (Tgel), and the hydrogel's Tgel was 35 C from
HE ET AL. 2127

F I G U R E 2 (a) Fourier-transform infrared


spectra of chitosan/fucoidan nanoparticles,
chitosan, and fucoidan controls;
(b) transmission electron microscopy images
and (c) size measurements of C5F5, C2F5,
C5F5@SDF-1, and C2F5@VEGF
nanoparticles; (d) sizes, polymer dispersity
index and zeta potential values of different
nanoparticles

Figure 3c. The composite viscosity reflects the injectable properties of with uniform pore diameters and the pore size in the range from 30 to
the thermosensitive hydrogel (Figure 3d): smaller composite viscosity 50 μm; this structure is conducive to the transportation of water and
indicated better flow property of the hydrogel which meant more con- nutrients; besides, introducing nanoparticles did not change the
ductivity for the in situ injection, while larger composite viscosity microstructure of the single hydrogel. Figure 4b shows the VEGF
expressed gelling. Figure 3d shows that the composite viscosity release of the hydrogel@NPVEGF&SDF-1, hydrogel@NPVEGF, and hydro-
maintained a lower and stable value when the temperature was lower gel@VEGF&SDF-1 samples: obviously, the hydrogel@NPVEGF&SDF-1
than the Tgel, suggesting the hydrogel's injectable property; however, samples released less VEGF compared with the hydrogel@NPVEGF and
when the temperature exceeded the Tgel, the composite viscosity rap- hydrogel@VEGF&SDF-1 samples, suggesting better stability of the
idly increased, and this meant the gelation of the hydrogel. VEGF loaded in the hydrogel@NPVEGF&SDF-1 samples. VEGF plays a
crucial role in EC proliferation and angiogenesis. Thus, its better stabil-
ity in the hydrogel scaffold will help for rapid vascularization in the
3.3 | SDF-1 and VEGF release
tissue/organ repair and regeneration. Figure 4c presents the SDF-1
Figure 4a depicts the morphology of the gelling single hydrogel and release curves of the hydrogel@NPVEGF&SDF-1, hydrogel@NPSDF-1, and
the hydrogel@NPVEGF&SDF-1 samples. Both the hydrogel control and hydrogel@VEGF&SDF-1 samples within 7 days, and displays an oppo-
the hydrogel@NPVEGF&SDF-1 samples had a porous network structure site trend compared to the VEGF release: the hydrogel@NPVEGF&SDF-1
2128 HE ET AL.

F I G U R E 3 (a) The entire


object of the hydrogel at 20 C
and 37 C; (b) G0 , G00 and phase
angle of the hydrogel at 37 C;
(c) G0 and G00 change of the
hydrogel with the temperature
changing; (d) complex viscosity
change of the hydrogel with the
temperature changing

F I G U R E 4 (a) Scanning
electron microscopy images of
the gelling single hydrogel and
the hydrogel@NPVEGF&SDF-1
samples; (b) VEGF release of
hydrogel@NPVEGF&SDF-1,
hydrogel@NPVEGF and
hydrogel@VEGF&SDF-1
samples (mean ± SD, n = 5);
(c) SDF-1 release of
hydrogel@NPVEGF&SDF-1,
hydrogel@NPSDF-1 and
hydrogel@VEGF&SDF1 samples
(mean ± SD, n = 5). NP,
nanoparticle; SDF-1, stromal
cell-derived factor-1; VEGF,
vascular endothelial growth
factor

samples released more SDF-1 compared with the hydrogel@NPSDF-1 3.4 | MSCs homing and proliferation
and hydrogel@VEGF&SDF-1 samples, indicating that more MSCs
The chemotactic effect of each hydrogel on MSCs is shown in
might be induced by the nanocomposite system because SDF-1 is a
Figure 5. The crystal violet stains showed that all the hydrogels
chemokine for the MSCs homing.
HE ET AL. 2129

F I G U R E 5 (a) Crystal violet staining of MSCs migrated to the underside of bottom membrane; (b) amounts of migrated MSCs (mean ± SD,
n = 3, *p < .05 compared with the other samples, **p < .05 compared with hydrogel@NPSDF-1, hydrogel@NPVEGF, and the single hydrogel control,
***p < .05 compared with hydrogel@NPVEGF and the single hydrogel control); (c) CLSM images of MSCs distributed in different hydrogels;
(d) proliferation of MSCs loaded in different hydrogels detected by CCK-8 assay (mean ± SD, n = 3, *p < .05 compared with the other samples,
**p < .05 compared with hydrogel@NPSDF-1, hydrogel@NPVEGF and the single hydrogel control, ***p < .05 compared with hydrogel@NPSDF-1 and
the single hydrogel control, ****p < .05 compared with the single hydrogel control). CCK-8, Cell Counting Kit-8; CLSM, confocal laser scanning
microscope; MSCs, mesenchymal stem cell; NP, nanoparticle; SDF-1, stromal cell-derived factor-1; VEGF, vascular endothelial growth factor

induce MSCs migration through the semipermeable membrane due indicating that the amounts of the SDF-1 released from the samples
to their good biocompatibility (Figure 5a). Wherein the hydrogel@ determined their functions on inducing MSCs homing.
NPVEGF&SDF-1 presented the largest MSCs number, suggesting better Figure 5c,d presents the MSCs distribution and proliferation in the
ability on inducing MSCs homing compared with other samples, and hydrogel@NPVEGF&SDF-1, hydrogel@VEGF&SDF-1, hydrogel@NPSDF-1,
the hydrogel@VEGF&SDF-1 and hydrogel@NPSDF-1 exhibited slightly hydrogel@NPVEGF, and single hydrogel control. The CLSM images
inferior capability successively, but they were still superior to the in a Z-axis mode showed that the hydrogel@NPVEGF&SDF-1 and
hydrogel@NPVEGF and single hydrogel control (Figure 5b). The MSCs hydrogel@VEGF&SDF-1 samples possessed more homogeneous
homing displayed a consistent result with the SDF-1 release, MSCs distribution and higher MSCs density compared with other
2130 HE ET AL.

samples (Figure 5c). The CCK-8 results showed that all the hydrogels 3.5 | HUVEC migration, proliferation, and apoptosis
promoted MSCs proliferation, wherein the designed hydroge-
The effect of different hydrogels on HUVEC migration behavior was
l@NPVEGF&SDF-1 system exhibited better functions (Figure 5d). Among
investigated via corresponding to the MSCs homing, and the results
the single-nanoparticle hydrogels, the hydrogel@NPVEGF contributed
are shown in Figure 6a,b. Consistent with the MSCs homing, the
significantly more to MSCs proliferation then the hydrogel@NPSDF-1,
which may be due to the direct promotion of the loaded VEGF factor hydrogel@NPVEGF&SDF-1 possessed the largest migrated HUVEC num-

and the preferable scale of the VEGF nanoparticles (154 nm). Our pre- ber; the second was the hydrogel@VEGF&SDF-1, followed by the

vious work proved that the nanoscale around 100 nm contributed to hydrogel@NPSDF-1, hydrogel@NPVEGF, and single hydrogel control.

better proliferation and oriented proliferation of the MSCs According to reports elsewhere, VEGF promotes angiogenesis and EC
(Li et al., 2016). growth (Ferrara, Gerber, & LeCouter, 2003; Leung, Cachianes, Kuang,

F I G U R E 6 (a) Crystal violet staining of HUVEC migrated to the underside of bottom membrane; (b) amounts of migrated HUVEC (mean ± SD,
n = 3, *p < .05 compared with the other samples, **p < .05 compared with hydrogel@NPSDF-1, hydrogel@NPVEGF, and the single hydrogel control,
***p < .05 compared with hydrogel@NPVEGF and the single hydrogel control, ****p < .05 compared with the single hydrogel control);
(c) phalloidin and DAPI staining of HUVEC on different samples surfaces; (d) HUVEC number detection via a typical CCK-8 assay (mean ± SD,
n = 3, *p < .05 compared with the other samples, **p < .05 compared with hydrogel@NPSDF-1, hydrogel@NPVEGF and the single hydrogel control,
***p < .05 compared with hydrogel@NPSDF-1 and the single hydrogel control, ****p < .05 compared with the single hydrogel control). CCK-8, Cell
Counting Kit-8; DAPI, 40 ,6-diamino-2-phenylindole; HUVEC, human umbilical vein endothelial cell, NP, nanoparticle
HE ET AL. 2131

Goeddel, & Ferrara, 1989; Shweiki, Itin, Soffer, & Keshet, 1992), how- 3.6 | Blood vessel ingrowth
ever, according to the results in Figure 6a,b, the migrated HUVEC
To further investigate the stimulating effect of the composite system
number on hydrogel@NPVEGF samples was less than the HUVEC num-
on in situ vascularization, we injected the hydrogel@NPVEGF&SDF-1,
ber on the single hydrogel control. The hydrogel@NPSDF-1 samples
hydrogel@VEGF&SDF-1, hydrogel@NPSDF-1, hydrogel@NPVEGF, and
presented larger HUVEC numbers than the hydrogel@NPVEGF sam-
the single hydrogel control in the CAM model to observe the induc-
ples. In this experiment, two major factors may influence the HUVEC
tion of blood vessel ingrowth. Figure 8 shows the HE staining of each
migration: SDF-1 also promoted the HUVEC migration, and simulta-
group: the purple area represents vascular tissue, and the red area
neously it induced the migrated HUVEC-secreted VEGF (Liu et al.,
indicates the hydrogels. There was a clear boundary between the pur-
2017); SDF-1 is a chemotactic factor, but VEGF is a growth factor.
ple area and red area in the single hydrogel control, suggesting little
Thus, the former plays an important role in directed migration,
blood vessel ingrowth. The hydrogel@NPSDF-1 group showed more
homing, and mobilization of cells (Orimo et al., 2005), while the
blood vessel ingrowth compared with the single hydrogel control, but
latter mainly contributes to cell proliferation and activity (Ferrara &
still less than the hydrogel@NPVEGF group. Obviously, VEGF plays a
DavisSmyth, 1997).
critical role in in situ vascularization. The hydrogel@NPVEGF&SDF-1
To investigate the effect of different hydrogels on HUVEC prolifer-
group showed most blood vessel of ingrowth; the blood vessels
ation, fluorescence staining and CCK-8 characterization were per-
filled almost the whole hydrogels. On the contrary, the hydrogel@
formed for HUVEC after 1 and 3 days culture on each hydrogel
VEGF&SDF-1 group induced very little blood vessel ingrowth, merely
(Figure 6c,d). Figure 6c shows that all the hydrogels supported the
as much as the hydrogel@NPSDF-1 group. The blood vessel ingrowth
HUVEC growth, wherein the hydrogel@NPVEGF&SDF-1 obviously pres-
in the CAM model demonstrated that the hydrogel@NPVEGF&SDF-1
ented larger HUVEC coverage compared with the other controls,
system induced better in situ vascularization depending on its selec-
suggesting better endothelial compatibility. In addition, more blue dots
tive release of VEGF/SDF-1, which was controlled by the co-loaded
stained by DAPI appeared on the hydrogel@NPVEGF&SDF-1 group, which
amphoteric nanoparticle carriers.
meant more DNA production in this group, and this phenomenon indi-
cated that the hydrogel@NPVEGF&SDF-1 sample had the better ability on
improving HUVEC proliferation. The CCK-8 result (Figure 6d) showed 4 | DISCUSSION
a consistent trend with the fluorescence images: the hydrogel@
NPVEGF&SDF-1 group exhibited larger HUVEC number compared with Tissue repair and regeneration has always been the key technology of
the other groups, suggesting better functions of improving HUVEC pro- clinical treatment and the focus of basic medical research (Wei, Zhan,
liferation. Although the HUVECs on all hydrogels presented increased Yu, & Chen, 2017). During this process, tissue vascularization plays a
number after 3 days culture, which indicated improved proliferation, crucial role for both the structure and function of vascular networks,
the HUVEC number showed a significant order on each sample: hydro- as well as nutrition and signal transmission through the blood flow
gel@NPVEGF&SDF-1 > hydrogel@VEGF&SDF-1 > hydrogel@ (Zhang et al., 2018). The amount and behavior of stem cells and VECs
NPVEGF > hydrogel@NPSDF-1 > single hydrogel control. Compared with decide the vascularization speed: numerous and active cells will
the VEGF release curve in Figure 4b, this result showed a negative cor- undoubtedly accelerate vascularization in tissue engineering, thereby
relation, that is, more HUVEC grew on the less VEGF release contributing to tissue repair and regeneration (Rafii & Lyden, 2003).
(or more VEGF remaining in the hydrogel) samples. As mentioned previ- Thus, the rapid recruitment and proliferation of stem cells and VECs
ously, VEGF is the specific growth factor for endothelial cells (ECs), thus to the lesion site induced by the scaffold are the key to the vasculari-
more VEGF loading contributed to quicker HUVEC growth. In addition, zation of early tissue repair and regeneration.
the SDF-1-loaded hydrogels (hydrogel@NPVEGF&SDF-1, hydrogel@ Temperature-responsive injectable hydrogels recently attracted
VEGF&SDF-1, and hydrogel@NPSDF-1) also possessed the better func- high attention for biomedical application, because their handling is
tion of improving HUVEC proliferation compared with single hydrogel more straightforward, minimally invasive, they can adapt to various
control which probably did from the VEGF released by the HUVEC via kinds of damaged tissues and organs, and they allow better contact
an SDF-1/CXCR4 pathway. with the host tissue (Ekenseair et al., 2012; Park et al., 2009). Our
The HUVEC apoptosis rate on each sample was investigated by a study also indicated that the pore sizes of the injectable hydrogels
live/dead staining method and further calculated. In the fluorescence was controlled by the materials parameters (such as feed ratios or
images, the green dots indicate the live cells and the red dots indicate molecular weight) and allowed tuning of the cell behaviors. Figure 4a
the dead cells. Figure 7a shows that all the hydrogels had lots of green shows that the hydrogels had a porous network structure with uni-
cells and very few red cells, suggesting good ability to inhibit HUVEC form pore size between 30 and 50 μm. The previous studies found
apoptosis. Quantification of vital cells from at least 15 images showed that pore sizes between 20 and 30 μm regulate single VECs to main-
that the rate of vital HUVEC on each sample was beyond 90%, the tain their physiological functions (Li et al., 2013; Wu et al., 2015), but
VEGF and SDF-1-loaded hydrogel had a higher rate, and the hydroge- multicellular regulation may need a little bigger sizes.
l@NPVEGF&SDF-1 further enhanced the values, which indicated a better Selective release of multiple biological factors from scaffolds may
inhibition of HUVEC apoptosis in the composite system (Figure 7b). guide multicellular synergy and precise therapy in clinical application.
2132 HE ET AL.

F I G U R E 7 (a) HUVEC viability


on the hydrogel@NPVEGF&SDF-1,
hydrogel@NPSDF-1,
hydrogel@NPVEGF,
hydrogel@VEGF&SDF-1, and
single hydrogel samples. (b) Ratio
of vital HUVEC grown on the
hydrogel@NPVEGF&SDF-1,
hydrogel@NPSDF-1,
hydrogel@NPVEGF,
hydrogel@VEGF&SDF-1, and
single hydrogel samples, which
were calculated from at least
15 images (*p < 0.05 compared
with all the other samples,
**p < 0.05 compared with the
single hydrogel control, mean
± SD, n = 15); green color indicates
vital cells, red color indicate dead
cells. HUVEC, human umbilical
vein endothelial cell; SDF-1,
stromal cell-derived factor-1;
VEGF, vascular endothelial growth
factor

Clinical scientists always hold in esteem on stem cells which are usually The MSCs migration experiment (Figure 5a,b) proved that the scaf-
chemotactic for SDF-1 to the scaffold to support more rapid recruit- fold with selective SDF-1/VEGF release (hydrogel@NPVEGF&SDF-1)
ment and proliferation in vascularization (Tachibana et al., 1998; Tang induced better MSCs homing compared with the nonselective one
et al., 2015), while VECs tend to migrate slowly to the target location. (hydrogel@VEGF&SDF-1). The scaffold with selective SDF-1/VEGF
The VEGF release may accelerate cell migration (Mohsen, Hatef, release also enhanced cell proliferation (Figures 5c,d and 6c,d) and cau-
Hamid, Ali, & Ghasem, 2018; Torio-Padron et al., 2007). However, it sed less cell apoptosis (Figure 7) compared to the nonselective scaffold.
seems that keeping VEGF in the scaffold supports vascularization In the typical CAM model, the SDF-1/VEGF selective-released scaffold
because it not only enhances the proliferation of cells (Carpenter et al., obviously possessed more areas of blood vessel ingrowth compared
2005; Zhou et al., 2018), but also regulates the interaction between with the nonselective scaffold, which confirmed better vascularization.

stem cells and ECs (such as directional differentiation of stem cells


induced by ECs and pore size in the scaffold) and promotes angiogene- 5 | C O N CL U S I O N S
sis (Adibfar et al., 2018; Cho, Moon, Park, Lee, & Seo, 2018; Zhou et al.,
2018). Thus, in the present study, we designed two kinds of In this study, we developed an injectable system composed of a
nanoparticles to load SDF-1 and VEGF, respectively. These molecules temperature-responsive hydrogel and factors-loaded nanoparticles for
had distinct charge properties (C5F5@SDF-1 nanoparticles were posi- potential application of vascularization in tissue engineering. This
tively charged, and C2F5@VEGF nanoparticles were negatively charged, composite system can control the selective release of MSCs
Figure 2d) and regulated the scaffold to release more SDF-1 (Figure 4c) chemokines (SDF-1) and cell growth factor (VEGF; more SDF-1
and less VEGF (Figure 4b) via electrostatic interaction with the hydrogel release and less VEGF release) via co-loading with two different
(the chitosan-based hydrogel was positively charged). nanoparticles. The SDF-1 nanoparticles possessed positive charge and
HE ET AL. 2133

F I G U R E 8 Histological analysis results: typical optical photographs of the HE-stained cross-sectional slices of the chick embryo blood vessel
with hydrogel@NPVEGF&SDF-1, hydrogel@NPSDF-1, hydrogel@NPVEGF, hydrogel@VEGF&SDF-1, and single hydrogel samples (the area between
two black dotted lines is the blood vessels in growth; the purple area indicates chick embryo blood vessel, and the red area indicates each
hydrogel). HE, hematoxylin-eosin; HUVEC, human umbilical vein endothelial cell; SDF-1, stromal cell-derived factor-1; VEGF, vascular endothelial
growth factor

bigger scales (304 nm), while the VEGF nanoparticles possessed nega- RE FE RE NCE S
tive charge and smaller scales (154 nm); wherein the smaller dimen-
Adibfar, A., Amoabediny, G., Eslaminejad, M. B., Mohamadi, J.,
sion and the loaded VEGF contributed to in situ cell growth, while the Bagheri, F., & Doulabi, B. Z. (2018). VEGF delivery by smart polymeric
sustained SDF-1 release supported the MSCs homing. The CAM PNIPAM nanoparticles affects both osteogenic and angiogenic capaci-
ties of human bone marrow stem cells. Materials Science and Engineer-
model further demonstrated that the composite system significantly
ing: C, 93, 790–799.
improved angiogenesis of the hydrogel scaffold. We expect that our Auerbach, R., Kubai, L., Knighton, D., & Folkman, J. (1974). A simple proce-
research facilitates the in situ vascularization during application in tis- dure for the long-term cultivation of chicken embryos. Developmental
sue or organ repair and regeneration. Future studies will focus on fur- Biology, 41(2), 391–394.
Bao, B., Hao, J., Bian, X., Zhu, X., Xiao, K., Liao, J., … Jiang, L. (2017). 3D
ther fine-tuning the parameters of the functional nanoparticles, such
porous hydrogel/conducting polymer heterogeneous membranes with
as dimensions, degradability, and more factors, aiming at more rapid electro-/pH-modulated ionic rectification. Advanced Materials, 29(44),
vascularization. 1702926.
Caccavo, D., Cascone, S., Lamberti, G., & Barba, A. A. (2018). Hydrogels:
Experimental characterization and mathematical modelling of their
mechanical and diffusive behaviour. Chemical Society Reviews, 47(7),
ACKNOWLEDGMENTS 2357–2373.
Carpenter, B., Lin, Y. K., Stoll, S., Raffai, R. L., McCuskey, R., & Wang, R.
We sincerely thank Professor Manfred F. Maitz for his help in this
(2005). VEGF is crucial for the hepatic vascular development required
work. This work was supported by the National Natural Science Foun- for lipoprotein uptake. Development, 132(14), 3293–3303.
dation of China (NSFC 81771988), Key Project and Special Founda- Chen, L., Li, J. A., Chang, J. W., Jin, S. B., Wu, D., Yan, H. H., … Guan, S. K.
tion of Research, Development and Promotion in Henan province (2018). Mg-Zn-Y-Nd coated with citric acid and dopamine by layer-by-
layer self-assembly to improve surface biocompatibility. Science China
(No. 182102310076), the Joint Fund for Fostering Talents of NCIR-
Technological Sciences, 61(8), 1228–1237.
MMT and HNKL-MMT (No. MMT2017-01), and Top Doctor Program Cho, H. D., Moon, K. D., Park, K. H., Lee, Y. S., & Seo, K. I. (2018). Effects
of Zhengzhou University (No. 32210475). of auriculasin on vascular endothelial growth factor (VEGF)-induced
angiogenesis via regulation of VEGF receptor 2 signaling pathways
in vitro and in vivo. Food and Chemical Toxicology, 121, 612–621.
Davoodi, P., Lee, L. Y., Xu, Q., Sunil, V., Sun, Y., Soh, S., & Wang, C. H.
CONF LICT OF IN TE RE ST
(2018). Drug delivery systems for programmed and on-demand
release. Advanced Drug Delivery Reviews, 132, 104–138.
The authors declare no potential conflict of interest.
Ekenseair, A. K., Boere, K. W. M., Tzouanas, S. N., Vo, T. N.,
Kasper, F. K., & Mikos, A. G. (2012). Synthesis and characterization of
thermally and chemically gelling injectable hydrogels for tissue engi-
ORCID neering. Biomacromolecules, 13(6), 1908–1915.
Ferrara, N., & DavisSmyth, T. (1997). The biology of vascular endothelial
Jing-An Li https://orcid.org/0000-0002-0305-6929 growth factor. Endocrine Reviews, 18(1), 4–25.
2134 HE ET AL.

Ferrara, N., Gerber, H. P., & LeCouter, J. (2003). The biology of VEGF and Park, K. M., Lee, S. Y., Joung, Y. K., Na, J. S., Lee, M. C., & Park, K. D. (2009).
its receptors. Nature Medicine, 9(6), 669–676. Thermosensitive chitosan-pluronic hydrogel as an injectable cell delivery
Griffith, L. G., & Naughton, G. (2002). Tissue engineering-current chal- carrier for cartilage regeneration. Acta Biomaterialia, 5(6), 1956–1965.
lenges and expanding opportunities. Science, 295(5557), 1009–1014. Rafii, S., & Lyden, D. (2003). Therapeutic stem and progenitor cell transplantation
Hu, H. T., Hao, L. X., Yan, B., Li, X. M., Zhu, Y. X., Yao, J., … Jiang, Q. for organ vascularization and regeneration. Nature Medicine, 9(6), 702–712.
(2018). Gefitinib inhibits retina angiogenesis by affecting VEGF signal- Rose, J. C., & Laura, L. D. (2018). Hierarchical design of tissue regenerative
ing pathway. Biomedicine & Pharmacotherapy, 102, 115–119. constructs. Advanced Health Care Materials, 7(6), 1701067.
Laiva, A. L., Raftery, R. M., Keogh, M. B., & O'Brien, F. J. (2018). Pro- Shweiki, D., Itin, A., Soffer, D., & Keshet, E. (1992). Vascular endothelial
angiogenic impact of SDF-1α gene-activated collagen-based scaffolds growth factor induced by hypoxia may mediate hypoxia-initiated
in stem cell driven angiogenesis. International Journal of Pharmaceutics, angiogenesis. Nature, 359(6398), 843–845.
544(2), 372–379. Tachibana, K., Hirota, S., Iizasa, H., Yoshida, H., Kawabata, K., Kataoka, Y., …
Leung, D. W., Cachianes, G., Kuang, W. J., Goeddel, D. V., & Ferrara, N. Nagasawa, T. (1998). The chemokine receptor CXCR4 is essential for vas-
(1989). Vascular endothelial growth factor is a secreted angiogenic cularization of the gastrointestinal tract. Nature, 393(6685), 591–594.
mitogen. Science, 246(4935), 1306–1309. Tang, J., Cui, X., Caranasos, T. G., Hensley, M. T., Vandergriff, A. C.,
Li, J. A., Qin, W., Zhang, K., Wu, F., Yang, P., He, Z. K., … Huang, N. (2016). Hartanto, Y., … Cheng, K. (2017). Heart repair using nanogel-
Controlling mesenchymal stem cells differentiate into contractile encapsulated human cardiac stem cells in mice and pigs with myocar-
smooth muscle cells on a TiO2 micro/nano interface: Towards benign dial infarction. ACS Nano, 11(10), 9738–9749.
pericytes environment for endothelialization. Colloids and Surfaces B: Tang, T., Jiang, H., Yu, Y., He, F., Ji, S. Z., Liu, Y. Y., … Xia, Z. F. (2015). A
Biointerfaces, 145, 410–419. new method of wound treatment: Targeted therapy of skin wounds
Li, J. A., Zhang, K., Wu, F., He, Z. K., Yang, P., & Huang, N. (2015). Con- with reactive oxygen species-responsive nanoparticles containing
structing bio-functional layers of hyaluronan and type IV collagen on SDF-1 alpha. International Journal of Nanomedicine, 10, 6571–6585.
titanium surface for improving endothelialization. Journal of Materials Torio-Padron, N., Borges, J., Momeni, A., Mueller, M. C.,
Science, 50, 3226–3236. Tegtmeier, F. T., & Stark, G. B. (2007). Implantation of VEGF trans-
Li, J. A., Zhang, K., Wu, J. J., Zhang, L. J., Yang, P., Tu, Q. F., & Huang, N. fected preadipocytes improves vascularization of fibrin implants on
(2015). Tailoring of the titanium surface by preparing cardiovascular the cylinder chorioallantoic membrane (CAM) model. Minimally Inva-
endothelial extracellular matrix layer on the hyaluronic acid micro- sive Therapy & Allied Technologies, 16(3), 155–162.
pattern for improving biocompatibility. Colloids and Surfaces B: Bio- Wang, C., Li, Y., Yang, M., Zou, Y., Liu, H., Liang, Z., … Zhang, B. (2018).
interfaces, 128, 201–210. Efficient differentiation of bone marrow mesenchymal stem cells into
Li, J. A., Zhang, K., Xu, Y., Chen, J., Yang, P., Zhao, Y. C., … Huang, N. (2014). endothelial cells in vitro. European Journal of Vascular and Endovascular
A novel co-culture models of human vascular endothelial cells and Surgery, 55(2), 257–265.
smooth muscle cells by hyaluronic acid micro-pattern on titanium sur- Wei, T., Zhan, W., Yu, Q., & Chen, H. (2017). Smart biointerface with photo-
face. Journal of Biomedical Materials Research: Part A, 102A, 1950–1960. switched functions between bactericidal activity and bacteria-releasing
Li, J. A., Zhang, K., Yang, P., Qin, W., Li, G. C., Zhao, A. S., & Huang, N. ability. ACS Applied Materials & Interfaces, 9(31), 25767–25774.
(2013). Human vascular endothelial cell morphology and functional Wu, J. J., Li, J. A., Wu, F., He, Z. K., Yang, P., & Huang, N. (2015). Effect of
cytokine secretion influenced by different size of HA micro-pattern on micropatterned TiO2 nanotubes thin film on the deposition of endo-
titanium substrate. Colloids and Surfaces B: Biointerfaces, 110, 199–207. thelial extracellular matrix: For the purpose of enhancing surface bio-
Li, J. A., Zou, D., Zhang, K., Luo, X., Yang, P., Jing, Y. Y., … Huang, N. compatibility. Biointerphases, 10, 04A302.
(2017). Strong multi-functions based on conjugating chondroitin sul- Zhang, H., Zhou, Y., Zhang, W., Wang, K., Xu, L., Ma, H., & Deng, Y. (2018).
fate on amine-rich surface direct vascular cells fate for cardiovascular Construction of vascularized tissue-engineered bone with a double-
implanted devices. Journal of Materials Chemistry B, 5, 8299–8313. cell sheet complex. Acta Biomaterialia, 77, 212–227.
Liu, K., Jiang, X., & Hunziker, P. (2016). Carbohydrate-based amphiphilic Zhang, K., Shi, Z. Q., Zhou, J. K., Xing, Q., Ma, S. S., Li, Q. H., … Guan, F. X.
nano delivery systems for cancer therapy. Nanoscale, 8, 16091–16156. (2018). Potential application of an injectable hydrogel scaffold loaded
Liu, T., Wang, X., Tang, X., Gong, T., Ye, W., Pan, C., … Wang, Q. M. (2017). with mesenchymal stem cells for treating traumatic brain injury. Jour-
Surface modification with ECM-inspired SDF-1α/laminin-loaded nal of Materials Chemistry B, 6, 2982–2992.
nanocoating for vascular wound healing. ACS Applied Materials & Inter- Zhong, R., Tang, Q., Wang, S., Zhang, H., Zhang, F., Xiao, M., … Pei, H. (2018).
faces, 9, 30373–30386. Self-assembly of enzyme-like nanofibrous G-molecular hydrogel for printed
Lü, L., Deegan, A., Musa, F., Xu, T., & Yang, Y. (2018). The effects of bio- flexible electrochemical sensors. Advanced Materials, 30(12), 1706887.
mimetically conjugated VEGF on Osteogenesis and angiogenesis of Zhou, L., Niu, X., Liang, J., Li, J., Li, J., Cheng, Y., … Zhang, K. (2018). Effi-
MSCs (human and rat) and HUVECs co-culture models. Colloids and cient differentiation of vascular endothelial cells from dermal-derived
Surfaces B: Biointerfaces, 167, 550–559. mesenchymal stem cells induced by endothelial cell lines conditioned
Luo, H., Zhang, Y., Zhang, Z., & Jin, Y. (2012). The protection of MSCs from medium. Acta Histochemica, 120(8), 734–740.
apoptosis in nerve regeneration by TGFβ1 through reducing inflamma- Zou, D., Luo, X., Li, J. A., Wang, S., Zhang, K., Sun, J. Y., … Zhang, C. J.
tion and promoting VEGF-dependent angiogenesis. Biomaterials, 33 (2018). Investigating blood compatibility and tissue compatibility of a
(17), 4277–4287. biomimetic ECM layer on cardiovascular biomaterials. Journal of Bio-
Mohsen, K., Hatef, S. A., Hamid, A., Ali, G., & Ghasem, M. (2018). Mesen- materials and Tissue Engineering, 8(5), 640–646.
chymal stem cells differentiate to endothelial cells using recombinant
vascular endothelial growth factor-A. Reports of Biochemistry & Molec-
ular Biology, 6(2), 144.
Orimo, A., Gupta, P. B., Sgroi, D. C., Arenzana-Seisdedos, F., Delaunay, T., How to cite this article: He D, Zhao A-S, Su H, et al. An
Naeem, R., … Weinberg, R. A. (2005). Stromal fibroblasts present in inva- injectable scaffold based on temperature-responsive hydrogel
sive human breast carcinomas promote tumor growth and angiogenesis and factor-loaded nanoparticles for application in vascularization
through elevated SDF-1/CXCL12 secretion. Cell, 121(3), 335–348.
in tissue engineering. J Biomed Mater Res. 2019;107A:
Park, H. J., Lee, W. Y., Kim, J. H., Park, C., & Song, H. (2018). Expression
patterns and role of SDF-1/CXCR4 axis in boar spermatogonial stem 2123–2134. https://doi.org/10.1002/jbm.a.36723
cells. Theriogenology, 113, 221–228.

You might also like