Sunteți pe pagina 1din 10

Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641

Contents lists available at ScienceDirect

Colloids and Surfaces B: Biointerfaces


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

A comparison of nanoscale and multiscale PCL/gelatin scaffolds


prepared by disc-electrospinning
Dawei Li a,b , Weiming Chen b , Binbin Sun b , Haoxuan Li a , Tong Wu b , Qinfei Ke a,c,∗∗ ,
Chen Huang a , Hany EI-Hamshary d,e , Salem S. Al-Deyab d , Xiumei Mo a,b,∗
a
Engineering Research Center of Technical Textiles, College of Textiles, Donghua University, Shanghai 201620, China
b
State Key Lab. for Modification of Chemical Fibers & Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua
University, Shanghai 201620, China
c
Shanghai Normal University, Shanghai 200234, China
d
Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
e
Department of Chemistry, Faculty of Science, Tanta University, Tanta 31527, Egypt

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

Article history: Electrospinning is a versatile and convenient technology to generate nanofibers suitable for tissue engi-
Received 3 February 2016 neering. However, the low production rate of traditional needle electrospinning hinders its applications.
Received in revised form 23 May 2016 Needleless electrospinning is a potential strategy to promote the application of electrospun nanofiber in
Accepted 4 July 2016
various fields. In this study, disc-electrospinning (one kind of needleless electrospinning) was conducted
Available online 7 July 2016
to produce poly(␧-caprolactone)/gelatin (PCL/GT) scaffolds of different structure, namely the nanoscale
structure constructed by nanofiber and multiscale structure consisting of nanofiber and microfiber. It
Keywords:
was found that, due to the inhomogeneity of PCL/GT solution, disc-electrospun PCL-GT scaffold pre-
Disc-electrospinning
Three-dimensional scaffolds sented multiscale structure with larger pores than that of the acid assisted one (PCL-GT-A). Scanning
Acetic acid electron microscopy images indicated the PCL-GT scaffold was constructed by nanofibers and microfibers.
Multiscale structure Mouse fibroblasts and rat bone marrow stromal cells both showed higher proliferation rates on multiscale
Nanofiber scaffold than nanoscale scaffolds. It was proposed that the nanofibers bridged between the microfibers
enhanced cell adhesion and spreading, while the large pores on the three dimensional (3D) PCL-GT scaf-
fold provide more effective space for cells to proliferate and migrate. However, the uniform nanofibers
and densely packed structure in PCL-GT-A scaffold limited the cells on the surface. This study demon-
strated the potential of disc-electrospun PCL-GT scaffold containing nanofiber and microfiber for 3D
tissue regeneration.
© 2016 Elsevier B.V. All rights reserved.

1. Introduction (3D), high porosity, and interconnected pores which could facilitate
cellular infiltration and transport of nutrition and waste [2,3].
Tissue engineering is a promising approach for the repair of Electrospinning has gained popularity in the fabrication of
tissue defects [1]. An effective scaffold selected for tissue regener- nanofibrous scaffolds from diverse natural and synthetic materi-
ation should provide suitable micro-environment for cell adhesion als, owning to its simplicity and cost-effectiveness. The nanoscale
and proliferation [1]. Various types of scaffolds have been invented structure of electrospun nanofiber mimics the structure of native
to meet the demand of tissue regeneration, among which fibrous extracellular matrix (ECM), which is crucial for cell adhesion,
scaffolds are widely used due to their three dimensional structure spreading, and proliferation [4]. Thus, electrospun nanofiber scaf-
folds have been widely applied in bone, skin, vascular, nerve, and
cartilage tissue regeneration [2,5–8]. However, it is accepted that
the highly compacted nanofibers may often result in increased cell
spreading and limited cellular infiltration, while cells require full
∗ Corresponding author at: State Key Lab. for Modification of Chemical Fibers &
context of 3D matrix to maintain their morphology and estab-
Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology,
lish natural behavior [3,9]. Thus, different approaches have been
Donghua University, Shanghai 201620, China.
∗∗ Corresponding author at: Engineering Research Center of Technical Textiles, invented to obtain electrospun structure with large pore [6,10–13].
College of Textiles, Donghua University, Shanghai 201620, China Kidoaki et al. attempted to remove part of the component in a mul-
E-mail address: xmm@dhu.edu.cn (X. Mo).

http://dx.doi.org/10.1016/j.colsurfb.2016.07.009
0927-7765/© 2016 Elsevier B.V. All rights reserved.
D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641 633

tilayered bicomponent scaffold to increase the pore size [14]. Salt


leaching was also applied to create large pores [15]. However, the
structure of scaffold would be affected by the leaching out pro-
cess. A second method was to increase the diameter of electrospun
fibers to several micrometers, leading to larger pore size and higher
porosity [16], but the method also led to a negative effect on cell
attachment and proliferation.
Recently, combination of nanofibers and microfibers became a
promising alternative for fabrication a 3D scaffold. The microfibers
formed a porous structure large enough for cell migration through
the scaffold, whereas the nanofibers could enhance protein absorp-
tion, improving protein-mediated interaction with the cell surface
so promoting cell adhesion and improving cell viability [17]. Soli-
man et al. electrospun scaffolds of microscale, nanoscale, and
multiscale fibers [18]. In vitro experiment showed the multiscale
scaffold boosted cell motility, survival, and proliferation, indicat-
ing that the 3D multiscale scaffold could allow different cell types
to coexist for the study of cell therapy of complex tissues. Pham
et al. manufactured bilayered constructs consisting of microfiber
scaffold with various thickness of nanofibers [19]. The presence
of nanofiber could enhance cell spreading. However, the layered
structure need a two step process, and the nanofibrous layers
always show a sheet-like structure over microstructure. To solve
this problem, Levorson et al. used a duel spinneret system to elec-
trospin different scale fibers and a cylindrical rotating mandrel to
collect the uniformly mixed multiscale scaffold [20]. It was reported
that the presence of nanoscale fiber could promote the produc-
tion and distribution of glycosaminoglycan, which revealed that
the scaffold was more closely resembling native ECM components.
An alternative strategy is demonstrated in this work in which Fig. 1. PCL/gelatin solution with and without acid. PCL/gelatin and PCL/gelatin/acid
needleless electrospinning is applied to generate 3D scaffold with solution left stand for 0 H (a) and 4 H (b). (c) Schematic of disc-electrospinning
multiscale structure. setup, consisting of a rotating metal disc spinneret, a solution vessel, a high-voltage
To fabricate a scaffold that can properly mimic the native ECM, direct-current power generator supply and a rotary collector.

the components always play a vital role. Synthetic materials pos-


sesse controllable properties, excellent mechanical strength but
poor biocompatibility, while natural materials could make up the DEMO Medical Tech. Co. Ltd. (China). All chemicals were used with-
drawbacks and promote cell attachment and proliferation but weak out further purification.
in mechanical properties [21]. Needle electrospinning was used to
produce Poly(␧-caprolactone) (PCL)/gelatin nanofiber to construct
tissue engineering scaffold [22–24]. The bicomponent nanofiber 2.2. Disc-electrospinning
possessed the merits of both PCL and gelatin, creating a favorable
environment for cell attachment and proliferation. 2.5 g PCL and 2.5 g porcine gelatin was dissolved in 50 mL
In the present study, PCL/gelatin fibrous scaffolds were fabri- TFE to generate 10% w/v PCL/gelatin solution. 500 ␮L acetic
cated via disc-electrospinning. An increased fiber diameter and a acid was added into another 50 mL PCL/gelatin solution to form
looser structure was achieved when acetic acid was absent in the 10% PCL/gelatin/acid solution. Disc-electrospinning was conducted
electrospinning solution. Fiber morphology, pore size and mechan- using a homemade needleless electrospinning setup described
ical properties of the two types of scaffolds were compared. FTIR previously (Fig. 1(c)) [25]. Briefly, the disc-electrospinning setup
and XRD were employed to assess the compositional and crys- consists a rotating metal disc spinneret, a solution vessel, a
talline characters. Fibroblasts and bMSCs were cultured to study high-voltage direct-current power generator supply (Gamma High
the biocompatibility. Voltage Research, USA) and a rotary collector. The applied voltage,
collecting distance, and the rotating speed of the disc was set as
50 kV, 25 cm, and 10 rpm, respectively. After disc-electrospinning,
the collected nanofiber was kept in vacuum at room temperature
2. Materials and experiments for 72 h to remove the solvent residue.

2.1. Materials
2.3. EDC/NHS crosslinking
PCL (average Mw 80,000) was purchased from Sigma-Aldrich
(USA). 2,2,2-Trifluoroethanol (TFE) was purchased from Shanghai Before in vitro experiments, EDC/NHS solution was used to
Darui Finechemical Co., Ltd. (China). Porcine gelatin was pur- crosslink the gelatin in PCL/gelatin scaffolds. 5.76 g EDC and 3.75 g
chased from MP Biomedicals, LLC (France). Acetic acid of analytical NHS was dissolved in 200 mL of 95% ethanol solution. The final
reagent was supplied by Kunshan JingKe Microelectronics Mate- concentration of EDC and NHS was 0.15 mol−1 and 0.16 mol−1 ,
rial Co., Ltd. (China). The culture medium and related reagents respectively. The crosslinking process was conducted by immers-
were purchased from Gibco Life Technologies Co., Ltd. (USA). 1-(3- ing the PCL/gelatin scaffolds in the EDC/NHS solution at 4 ◦ C for
Dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC) 12 h. After the crosslinking process, the scaffolds were rinsed with
and N-Hydroxysuccinimide (NHS) was purchased from Shanghai 75% ethanol solution for three time, and dried for 48 h at 37 ◦ C.
634 D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641

2.4. Characterizations folds at predetermined time were fixed, dehydrated, and followed
by freeze-drying for 24 h. Then the samples were sputter-coated
Fiber morphology of the disc-electrospun PCL/Gelatin nanofiber with gold and visualized by SEM.
was examined by scanning electron microscopy (SEM, TM 3000, Confocal laser microscopy imaging was applied to observe the
Hitachi, Japan). The disc-electrospun nanofiber was briefly sputter- distribution and morphology of cells on the scaffolds [25]. Cells on
coated with gold for 30 s. Images were acquired using an SEM the scaffolds were fixed and the cytoskeletons and nuclei of cells
operating at an accelerating voltage of 15 kV. Diameter of fibers were stained with 25 ␮g mL−1 rhodamine-conjugated phalloidin
were measured based on the SEM figures by image visualization (Invitrogen, USA) and 10 ␮g mL−1 4 ,6 -diamidino-2-phenylindole
software Image J (National Institutes of Health, USA). 100 fibers hydrochloride (DAPI, Invitrogen, USA) for 30 min and 5 min, respec-
were measured for each sample. tively. Subsequently, the cells were observed using confocal laser
The pore size of the scaffolds was determined as described scanning microscopy (CLSM) (Carl Zeiss, LSM 700, Germany).
previously [25]. Briefly, the PCL/gelatin scaffolds was cut into Cell infiltration was analysed with z-stacked confocal
30 mm × 30 mm squares, which were wetted by the wetting agent, microscopy. For each sample, 20 slices were photoed at one
calwick (PMI Porous Materials Int.), with a surface tension of field of vision by CLSM. Thereafter, imaging software ZEN 2008
21 dyn cm−1 . Subsequently, a CFP-1100-AI capillary flow porom- (Carl Zeiss, Germany) was employed to provide 3D views of the
eter (PMI Porous Materials Int.) was used to determine the pore stained cells growing on the scaffolds using depth coding.
size and pore distribution.
Tensile properties of disc-electrospun nanofiber were ana- 2.6. Statistical analysis
lysed by stressing uniaxially until the scaffolds failed. Various
disc-electrospun PCL/gelatin nanofiber scaffolds (before and after All the data were obtained at least in triplicate and all values
crosslinking) were tested (n = 5). All samples were cut into same were presented as the mean and standard deviation (SD). Statistical
size of 50 mm × 10 mm and tested using a universal materials tester analysis was performed by the one-way analysis of variance using
(H5 K-S, Hounsfield, UK) with a 50 N load cell at ambient temper- Origin 8.0 (OriginLab Inc., USA). The statistical difference between
ature of 20 ◦ C and humidity of 65%. The effective length of the test two sets of data was considered when * p < 0.05, ** p < 0.01, and ***
was 30 mm with a stretching speed of 10 mm/min. p < 0.001.
Chemical analysis of PCL/gelatin nanofiber scaffolds (before and
after crosslinking) was carried out by Fourier transform infrared 3. Results
spectroscopy (FTIR). A FTIR spectrophotometer (Avatar380, USA)
was used to obtain FTIR spectra of the disc-electrospun PCL/gelatin 3.1. Phase separation and disc-electrospinning
nanofiber scaffolds over the range of 500–4000 cm−1 at scanning
resolution of 2 cm−1 . Although both PCL and gelatin could be dissolved in TFE to
X-ray diffraction spectroscopy was conducted on a D/max-2550 form transparent solution, when being mixed together, the mixed
PC (Rigaku, Japan) with Cu K␣ radiation. The operating voltage PCL/gelatin solution transformed to emulsion and became opaque.
and current was set as 40 kV and 200 mA. The disc-electrospun Phase separation occurred and two different layer of solution was
PCL/gelatin nanofiber scaffolds were examined between 0 and formed after left to stand for hours [28]. The addition of acetic
60◦ (2␪) at a scanning rate of 1◦ per minute. acid could transform the opaque solution clear again. The com-
parison of the two kinds of solution was shown in Fig. 1(a). After
2.5. In vitro cell culture 4 h of standing, the PCL/gelatin solution became layered, while
the PCL/gelatin/acid solution was still transparent (Fig. 1(b)). Feng
Mouse fibroblasts (L929) were supplied by the Institute of Bio- et al. confirmed that in the bottom layer, gelatin was dominant,
chemistry and Cell Biology of the Chinese Academy of Sciences while PCL was mainly in the top layer [28]. In the initial stage of
(Shanghai, China). L929 were maintained and expanded in high the phase separation, PCL phase was continuous in the PCL/gelatin
glucose Dulbecco’s modified eagle medium (DMEM) medium con- solution while the gelatin was micro balloon sphere dispersed in
taining 10% fetal serum, 100 units/mL penicillin and 100 units/mL the PCL phase. The gelatin sphere gradually increased and aggre-
streptomycin, incubated in humidified atmosphere with 5% CO2 at gated which resulted in layered solution after hours. As traditional
37 ◦ C. The culture medium was refreshed every other day. electrospinning always lasts for several hours, phase separation
Rat bone marrow stromal cells (bMSCs) were isolated using affects the morphology and composition of generated fiber. Disc-
density gradient centrifugation as described previously [26]. The electrospinning always last for only several minutes, during which
isolated bMSCs were maintained and expanded in DMEM/F12 the properties of the solution will not change evidently. The prop-
medium containing 10% fetal serum, 100 units/mL penicillin and erties of obtained fiber would keep same during the process. During
100 units/mL streptomycin. The culture medium was refreshed disc-electrospinning, the production for PCL-GT is 9.47 ± 0.56 g h−1
every other day. and for PCL-GT A is 9.80 ± 0.55 g h−1 .
The in vitro biocompatibility assessment was performed as
described previously [25]. L929 and bMSCs were seeded on the pre- 3.2. Morphology of disc-electrospun nanofiber
pared scaffolds. For L929, 2 × 104 cells were seeded on the scaffolds
with tissue culture plates (TCPs) as control. The culture medium Fig. 2 illustrates the morphology and micro structure of the disc-
was refreshed every other day. After 1, 3, 5 and 7 days of culture, electrospun scaffolds. It could be seen that the PCL-GT scaffold
the culture medium was removed and the samples were rinsed was fluffy and loosely packed while the PCL-GT-A scaffold showed
with PBS buffer for three times to remove the unattached cells and a dense appearance (Fig. 2(a–c)). The average diameter of fibers
the culture medium. Subsequently, the amount of cells was deter- for PCL-GT and PCL-GT-A was 592.3 ± 598.0 and 316.1 ± 163.7 nm,
mined by the standard MTT assay described previously [25]. For respectively. The diameter of fibers in PCL-GT scaffolds was ran-
bMSCs, 10 × 104 cells were seeded on the scaffolds, and the MTT domly ranged from ∼100 nm to several micron (Fig. 2(d), (e) and
assay was performed on 3, 7, 14 days post seeding. The experiment (h)). The pores between fibers were much larger than those of
was conducted with 6 parallel samples. PCL-GT-A. The microfibers built up the backbone, constructing the
Cell morphology on the scaffolds was determined followed the support of large pore. The nanofibers formed webs and bridged
method reported by Wu et al. [27]. Briefly, cells cultured on the scaf- between the microfibers, filling into the large pores (Fig. 2(e)). In the
D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641 635

Fig. 2. (a) Appearance of disc-electrospun PCL-GT and PCL-GT-A scaffold. Side view of the PCL-GT (b) and PCL-GT-A (c) scaffold. SEM images of PCL-GT (d), (e) and PCL-GT-A
(f), (g) scaffold. Fiber size distribution of PCL-GT (h) and PCL-GT-A (i) scaffold.

PCL-GT-A scaffold, the diameter of fibers was thinner and showed tension and viscosity would work against the electric field force
a smaller distribution, resulted in a flat and densely packed film and thus restrict the stretching, resulting in thicker fiber diameter
(Fig. 2(f), (g) and (i)). in PCL-GT scaffold. And the increase of conductivity also facilitated
Feng et al. reported that the addition of acetic acid could change the stretching to avoid the microfibers [29]. Moreover, the inhomo-
the solution properties by reducing the pH value, surface tension geneity and phase separation of the solution caused the uniformity
and viscosity, and increasing the conductivity [28]. Higher surface of the surface of polymer jets. As the PCL phase was continuous and
636 D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641

Fig. 3. Pore size distribution of PCL-GT (a) and PCL-GT-A (b) scaffold. The average pore diameter for PCL-GT and PCL-GT-A scaffolds was 13.39 ␮m and 2.40 ␮m, respectively.

the gelatin formed particles dispersed in the PCL phase. Current bond, respectively [31]. The addition of acetic acid showed no influ-
experimental phenomena suggested that PCL occupied the body ence in the components in the disc-electrospun PCL/gelatin fibers.
of the polymer jet generated from the disc, while the gelatin part XRD results suggested that, compared to the PCL-GT scaffold,
formed spherical droplets inserted in the PCL polymer jets, causing the characteristic diffraction peaks of PCL at 21.56 and 23.84◦ were
the undulating surface of the electrified jet. Forced by the surface significantly sharpened in the PCL-GT-A scaffold, indicating a much
tension, electrostatic and coulombic repulsion force, gelatin might higher crystallinity. During needle electrospinning, the addition of
be generated into branches from the jet body. After stretching, acetic acid could improve the miscibility of PCL and gelatin solu-
the branches were collected and form nanofiber web between the tion. Thus the better miscibility of blended nanofibers decrease
microfibers. It was reported that gelatin in TFE solution was neg- the crystallinity [28]. However, in the disc-electrospun PCL-GT and
atively charged, which may also caused the formation of ultrafine PCL-GT-A scaffolds a reverse result was obtained. The fibers in PCL-
fiber of ∼100 nm. GT-A were thinner than that in PCL-GT scaffold, demonstrating
a stronger stretching was performed during disc-electrospinning.
Tim et al. found that the smaller diameter fibers was resulted from
sufficient crystallization of polymer chain before the polymer jets
3.3. Physical property of disc-electrospun nanofiber
reaches the collector, along with extra elongation of the jets [32].
Fig. 3 illustrates the pore size distribution in two scaffolds. It
could be seen in the PCL-GT scaffold, the diameter of pores under- 3.4. Cell proliferation on scaffolds
went a wider distribution ranging from 9 ␮m to 19 ␮m and the
average diameter was 13.39 ␮m. In the PCL-GT-A scaffold, the aver- Biocompatibility such as cell proliferation, adhesion, spreading,
age pore diameter was 2.40 ␮m, concentrating between 1.7 ␮m and and migration on the scaffolds is an important aspect to evaluate
2.9 ␮m. The large pores were constructed by the loosely distributed the fitness for tissue regeneration. For tissue engineering scaffold,
microfibers, indicated in Fig. 2(d) and (e). While the densely packed surface properties and structure are major factors in regulating cell
nanofibers formed a compact structure, reducing the pore size as behavior [9]. L929 cells and BMSCs were cultured on two types
in Fig. 2(f) and (g). of scaffolds to determine the biocompatibility. As the proliferation
The mechanical property was characterized by stretching 5 rate of BMSCs was much slower than that of L929, the culturing
specimens of each scaffold. Fig. 4(a) show the stress-strain behavior periods were set as 1, 3, 7 days and 3, 7, 14 days for L929 and
of the disc-electrospun PCL-GT and PCL-GT-A scaffold. The aver- BMSCs, respectively. MTT assay was conducted at predetermined
age tensile strength, strain at break, and Young’s modulus were time point and the result was illustrated in Fig. 5. In Fig. 5(a), it
illustrated in Fig. 4(b)–(d), respectively. It could be seen from these could be found that during the 7 day long culturing, cells on PCL-
results that the PCL-GT-A scaffold showed higher tensile strength GT-A scaffold were less than that on PCL-GT. After 3 days of culture,
and Young’s modulus than those of PCL-GT scaffold. However, it L929 on TCP were higher than on PCL-GT scaffold. Interestingly, the
was not statistically different for their strain at break. In the PCL- trend was reversed after 7 days of culture. Similar trend occurred
GT scaffold, the density of fibers was less than that of PCL-GT-A to the BMSCs during the 14 days culturing period (Fig. 5(b)). BMSCs
scaffold. Therefore, the contact points for a single fiber with other cultured on the PCL-GT scaffolds showed significant increase over
fibers were also less in PCL-GT scaffold, which reduced the mechan- time and performed the fastest growth rate throughout the cultur-
ical strength and increased the motion facility. Thus, the PCL-GT ing time. After 14 days, BMSCs on the PCL-GT scaffolds increased
scaffold showed weaker strength and Young’s modulus. from 100 thousand to over 500 thousand, which was twice as many
ATR-FTIR and XRD results were shown in Fig. 4e and f. FTIR as that on PCL-GT-A (about 250 thousand).
spectra for the two scaffolds were just the same. The PCL related These results indicated that PCL-GT scaffold with large pores
stretching modes were observed in PCL-GT and PCL-GT-A scaffolds, performed better than the PCL-GT-A scaffold with small pore in
including 2946 cm−1 (asymmetric CH2 stretching), 2868 cm−1 long period cell proliferation. In our previous study, L929 showed
(symmetric CH2 stretching), 1727 cm−1 (carbonyl stretching), and high proliferation rate on the disc-electrospun PCL scaffold with 3D
1293 cm−1 (C O and C C stretching in the crystalline phase) [30]. structure [25]. It was concluded that the large pores in the PCL-GT
Typical protein related bonds were found at 1652 cm− 1 (amide I) scaffold provided another direction for cell spreading and migra-
and 1549 cm−1 (amide II), corresponding to C O stretching vibra- tion [33]. The interconnected pores could promote the nutrition
tion, and coupling of bending of N H bond and stretching of C N transportation and the removal of metabolic waste.
D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641 637

Fig. 4. Mechanical properties of PCL-GT and PCL-GT-A scaffold. (a) Stress-strain curve, (b) Tensile strength, (c) Stress at break, and (d) Young’s modulus. FTIR spectra (a) and
X-ray diffraction (b) of PCL-GT and PCL-GT-A scaffolds.

Fig. 5. Cell proliferation of L929 (a) and BMSCs (b) on PCL-GT, PCL-GT-A scaffolds and TCP quantitatively measured by MTT assay. * indicates statistically difference for
p < 0.05; ** indicates statistical difference for p < 0.01; *** indicates statistical difference for p < 0.001.

3.5. Morphology of cells on scaffolds found not only on the surface, but also inside the scaffold. Three
days later, number of cells in the scaffolds increased. Some cells
The structure of scaffolds was treated as a priority for cellular have migrated into the PCL-GT scaffolds, as labeled by the white
colonization in tissue regeneration, which would affect the inter- arrows in Fig. 6(b), (b’). However, for the PCL-GT-A scaffolds, the
action of cells with the scaffolds. The morphology of cells on two pore size between fiber was much smaller than cell, restricting the
scaffolds was observed by SEM (Fig. 6). L929 on both scaffolds cells on the surface (Fig. 6(e), (e’)).
showed a significantly increase over time from day 1 to day 7. On the After 7 days of culture, L929 on PCL-GT-A scaffolds further
first day after cell seeding, cells adhered to the fibers and formed a spread and connected with each other, forming a thin layer
typical spindle shape (Fig. 6(a), (a’), (d), (d’)). On the PCL-GT scaffold, (Fig. 6(f), (f’)). While for the PCL-GT scaffold, cells could spread in
cells were thinner than that on PCL-GT-A scaffold. L929 cells was the thickness direction. Most cells kept single and distributed dis-
638 D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641

Fig. 6. SEM images of L929 on two scaffolds. L929 cultured on PCL-GT scaffolds after 1 day (a), (a’), 3 days (b), (b’), and 7 days (c), (c’). L929 cultured on PCL-GT-A scaffolds
after 1 day (d), (d’), 3 days (e), (e’), and 7 days (f), (f’) (Bar: 100 ␮m).

persedly (Fig. 6(c), (c’)). Only part of the surface was covered by Fig. 7(g–j) illustrates the depth of fibroblasts cultured on PCL-GT
cells, leaving efficient space for further proliferation. Round cells and PCL-GT-A scaffolds, respectively. The stained cells were pho-
were observed on PCL-GT-A scaffolds in Fig. 6(f), (f’) (indicated by tographed by LSCM in Z-stacked mode. The obtained images was
white circles). This suggested that the insufficient living space lim- stacked up and translated into depth code where different colors
ited the spreading of cells. In contrast, most cells on the PCL-GT represent an increasing depth from red to blue. It could be seen that
scaffolds kept the spindle shaped or starlike, which was considered cells could be observed in a depth of 140 ␮m from the upper sur-
as a signal of health. face in the PCL-GT scaffold (Fig. 7(g) and (i)), which indicated that
The morphology of cells was also observed by LSCM (Fig. 7). cells had infiltrated into the scaffold for a 140 ␮m long distance.
On the first day, cells on both scaffolds showed spindle or starlike While on the PCL-GT-A scaffold, only a thin layer of cells, ranging
shapes, indicating normal spreading (Fig. 7(a), (d)). After 3 days, from 20 ␮m to 70 ␮m (Fig. 7(h) and (j)), could be seen, meaning
the whole surface of the PCL-GT-A scaffold was occupied by cells that cells were restricted on the surface of the scaffold.
(Fig. 7(e)). On the 7th day, part of the cells turned round (Fig. 7(f)),
which was caused by the lack of living space. In contrast, after 4. Discussion
7 days, gaps between cells could still be observed on the PCL-GT
scaffold (Fig. 7(c)). The spindle shaped cells were dominant, demon- Electrospinning has been widely used in the fabrication of
strating the potential for further proliferation. scaffolds with nanoscale structure, while the low production rat
D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641 639

Fig. 7. Confocal images of L929 cells on scaffolds. L929 on PCL-GT scaffolds after (a) 1 day, (b) 3 days, (c) 7 days of growth, on PCL-GT-A after (d) 1 day, (e) 3 days, (f) 7 days
of culture. Cell penetration on (g) PCL-GT scaffold, (h) PCL-GT-A scaffold, and side view of the cells distribution in PCL-GT (i) and PCL-GT-A scaffold (j). visualized by confocal
images in z-stack mode. Different colors represent an increasing depth from red to blue. (For interpretation of the references to colour in this figure legend, the reader is
referred to the web version of this article.)

of traditional needle electrospinning limit the application Gen- Electrospinning PCL/gelatin blends into nanofiber has been
erally, the production rate for the conventional electrospinning extensively studied in previous literatures [22–24,28]. Nanofiber
varies from 0.1 to 1.0 g/h. The development of needleless elec- constructed by PCL and gelatin possessed the merits of both
trospinning raised an valid solution to scale up the production of synthetic and natural materials. Gelatin could improve the bio-
nanofiber. In this study, disc-electrospinning was used to fabri- compatibility, while the PCL strengthen the mechanical properties.
cate PCL/gelatin scaffold with homogeneous and inhomogeneous However, the obtained PCL/gelatin scaffold was constructed by
PCL/gelatin blends. The properties of both scaffolds was char- fibers of nanoscale, which reduced the pore size to several microm-
acterized and in vitro experiment was conducted to study the eters between fibers in the scaffold, inhibiting the cells on the very
biocompatibility.
640 D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641

Fig. 8. The architecture of electrospun nanofibrous scaffold affects the distribution and morphology of cells. (a) Cells adhere and along a single microfiber. (b) and (d) Cells
spread on the surface of nanofiber scaffold. (c) and (e) Cells adhere on both microfiber and nanofiber inserted in the microstructure.

surface. Thus the cells could only form a thin layer without migra- ture period, even better than the TCP (Fig. 5). As the culturing
tion into the scaffold. period extended, the superiority of the multiscale scaffold became
On the other hand, to generate the nanofiber into a 3D scaffold more obvious. Which could attribute to the more space provided
allowing cell infiltration, several methods were invented, includ- by the large pore structure of the PCL-GT scaffold. Moreover, the
ing salt leaching, freeze drying, water bath collecting. All these interconnected pore structure in the PCL-GT scaffold facilitated the
methods generated large pore un-uniformly distributed in the scaf- transport of nutrition and metabolic waste.
fold. Some parts formed voids, while the rest was still densely Scaffolds constructed by nanofiber with nanoscale architectures
packed nanofibers. Recently, the combination of nanofiber and were believed to have larger surface areas to absorb proteins, pre-
microfiber provide a promising method to solve the existing prob- senting more binding sites for cell attachment [9]. Electrospun
lems. Microfiber formed the backbone of the scaffold while the nanofiber was widely applied in the fabrication of various tissue
nanofibers bridged between microfibers. Experiments had verified engineering scaffolds. In present, it has been verified that cells
that the multiscale scaffold could boost cell motility, survival, and culture on microfiber scaffold always adhere and spread along a
proliferation. However, the existing methods were based on needle single fiber (Fig. 8(a)) [18,34]. Without support, cells could not col-
electrospinning, which always need a dual needle system. onize in the space between fibers. Compared with the microfiber,
Here, we compared the nanoscale and multiscale PCL/Gelatin electrospun nanofiber could promote cells attachment, spreading,
scaffold prepared via disc-electrospinning. Experiments has indi- and proliferation [19]. The high porosity would facilitate nutri-
cated that phase separation would happen when PCL and gelatin tion transport and removal of the waste. However, as illustrated
dissolved in TFE simultaneously. Gelatin would form micro spheres in Fig. 8(b) and (d), the small pore size in the scaffold limited the
distributed in the PCL solution. In contrast, the addition of acetic cell migration into the scaffold [5,35,36].
acid could reduce the pH of the solution and thus eliminate the Nanofibers and microfibers were combined to form 3D struc-
phase separation. During disc-electrospinning, phase separation ture with large pores and efficient nanoscale binding sites. Pham
would help generate multiscale fibers form the inhomogeneous et al. manufactured bilayered constructs consisting of microfiber
solution (Figs. 1 and 2). Compared with the uniform nanofiber scaffold with various thickness of nanofibers [19]. The presence
of PCL-GT-A scaffold (Fig. 2(f) and (g)), multiscale PCL-GT scaf- of nanofiber could enhance cell spreading, while increasing the
fold shows larger pore size. The nanofiber of hundreds nanometer thickness of nanofiber layer reduced cell infiltration. Soliman et al.
could be found bridged between the microfibers, forming nets in produced the multiscale 3D scaffolds containing fibers with aver-
the pores (Fig. 2(d) and (e)). The pore size distribution verified the age diameter of 3.3 ␮m and 0.6 ␮m via multimodal electrospinning
difference (Fig. 3). It could be found that the pore size was signifi- [18]. Results indicated that nanofibers properly inserted in the
cantly raised in the PCL-GT scaffold. The pore size is quite important microfiber scaffold could crucially enhance the interaction between
for the cell colonization in the scaffold. Migration of cells into the cells and scaffold (Fig. 8(c)).
scaffold is crucial for the success of the graft and the overall healing In this study, multiscale scaffold constructed by PCL and
of defect. Greater pore size always leads to enhanced cell infiltra- Gelatin was prepared via disc-electrospinning. In this scaffold, the
tion. However, the multiscale PCL-GT scaffold is loosely packed, microfibers acted as the backbone and supported the 3D structure.
which reduces the mechanical strength and increased the motion Nanofibers inserted among the microfiber framework, bridging
facility (Fig. 4). The addition of acetic acid have no influence on the adjacent microfibers. As shown in Fig. 8(e), fibroblasts cultured on
components but increased the crystallinity of the obtained fiber the PCL-GT scaffold were dispersedly distributed in a 3D manner.
(Fig. 4). Some cells adhered and spread along a single microfiber (white
To study the biocompatibility, L929 and BMSCs were cultured lines). Some cells partly attached to a microfiber and spread out
on the multiscale scaffold. Results indicated that cells culture on along nanofibers two types of adhesion points (white arrows). The
PCL-GT scaffold showed a higher proliferation rate during the cul-
D. Li et al. / Colloids and Surfaces B: Biointerfaces 146 (2016) 632–641 641

others adhered on the nanoweb constructed by nanofibers among [11] C. Vaquette, J. Cooper-White, A simple method for fabricating 3-D
microfibers (hollow arrows). multilayered composite scaffolds, Acta Biomater. 9 (2013) 4599–4608.
[12] C.R. Wittmer, A. Hébraud, S. Nedjari, G. Schlatter, Well-organized 3D
nanofibrous composite constructs using cooperative effects between
5. Conclusion electrospinning and electrospraying, Polymer 55 (2014) 5781–5787.
[13] S.J. Kim, D.H. Jang, W.H. Park, B.-M. Min, Fabrication and characterization of
3-dimensional PLGA nanofiber/microfiber composite scaffolds, Polymer 51
We prepared multiscale PCL-GT scaffold containing nanofiber (2010) 1320–1327.
and microfiber via disc-electrospinning. Due to the inhomogene- [14] S. Kidoaki, I.K. Kwon, T. Matsuda, Mesoscopic spatial designs of nano-and
microfiber meshes for tissue-engineering matrix and scaffold based on newly
ity of the PCL-GT in TFE solution, microfibers and nanofibers were devised multilayering and mixing electrospinning techniques, Biomaterials
generated simultaneously and formed multiscale scaffolds with 26 (2005) 37–46.
large pores. Compared with the acetic acid assisted PCL-GT-A scaf- [15] J. Nam, Y. Huang, S. Agarwal, J. Lannutti, Improved cellular infiltration in
electrospun fiber via engineered porosity, Tissue Eng. 13 (2007) 2249–2257.
fold constructed by uniform nanofibers, PCL-GT scaffold possessed [16] Q.P. Pham, U. Sharma, A.G. Mikos, Electrospun poly(␧-caprolactone)
larger pores with a diameter of 13.39 ␮m. In vitro experiments indi- microfiber and multilayer nanofiber/microfiber scaffolds: characterization of
cated that the L929 and BMSCs cultured on PCL-GT scaffold showed scaffolds and measurement of cellular infiltration, Biomacromolecules 7
(2006) 2796–2805.
higher proliferation rate. PCL-GT also enhanced the cell infiltration
[17] K. Shalumon, N. Binulal, M. Deepthy, R. Jayakumar, K. Manzoor, S. Nair,
into the scaffold while the nanofibrous structure of PCL-GT-A scaf- Preparation, characterization and cell attachment studies of electrospun
fold inhibited the cell on the surface. In the PCL-GT scaffold, cells multi-scale poly(caprolactone) fibrous scaffolds for tissue engineering, J.
Macromol. Sci. Part A 48 (2010) 21–30.
seemed to settle and align on the microfibers but bridge between
[18] S. Soliman, S. Pagliari, A. Rinaldi, G. Forte, R. Fiaccavento, F. Pagliari, O.
microfibers through the nanoweb constructed by the nanoscale Franzese, M. Minieri, P. Di Nardo, S. Licoccia, E. Traversa, Multiscale
fibers, distributing throughout the whole scaffold in a 3D manner. three-dimensional scaffolds for soft tissue engineering via multimodal
Results in this study demonstrated the potential of disc-electrospun electrospinning, Acta Biomater. 6 (2010) 1227–1237.
[19] Quynh P. Pham, Upma Sharma, Antonios G. Mikos, Electrospun
PCL-GT scaffold with multiscale structure in the application of tis- poly(␧-caprolactone) microfiber and multilayer nanofiber/microfiber
sue engineering. scaffolds: characterization of scaffolds and measurement of cellular
infiltration, Biomacromolecule 7 (10) (2006) 2796–2805.
[20] E.J. Levorson, P. Raman Sreerekha, K.P. Chennazhi, F.K. Kasper, S.V. Nair, A.G.
Acknowledgements Mikos, Fabrication and characterization of multiscale electrospun scaffolds
for cartilage regeneration, Biomed. Mater. 8 (2013) 014103.
[21] X. Wu, L. Black, G. Santacana-Laffitte, C.W. Patrick Jr., Preparation and
This research was supported by National Natural Science Foun- assessment of glutaraldehyde-crosslinked collagen-chitosan hydrogels for
dation of China (31470941, 31271035, and 51403033), Science adipose tissue engineering, J. Biomed. Mater. Res. Part A 81 (2007) 59–65.
and Technology Commission of Shanghai Municipality Program [22] Y. Zhang, H. Ouyang, C.T. Lim, S. Ramakrishna, Z.M. Huang, Electrospinning of
gelatin fibers and gelatin/PCL composite fibrous scaffolds, J. Biomed. Mater.
(14JC1492100), Ph.D. Programs Foundation of Ministry of Edu- Res. Part B: Appl. Biomater. 72 (2005) 156–165.
cation of China (20130075110005) and light of textile project [23] W. Ji, F. Yang, J. Ma, M.J. Bouma, O.C. Boerman, Z. Chen, J.J. van den Beucken,
(J201404), The authors would like to extend their sincere apprecia- J.A. Jansen, Incorporation of stromal cell-derived factor-1alpha in PCL/gelatin
electrospun membranes for guided bone regeneration, Biomaterials 34 (2013)
tion to the Deanship of Scientific Research at King Saud University 735–745.
for its funding of this research through the research group project [24] L. Ghasemi-Mobarakeh, M.P. Prabhakaran, M. Morshed, M.-H. Nasr-Esfahani,
NO. RGP-201, and the Fundamental Research Funds for the Central S. Ramakrishna, Electrospun poly(␧-caprolactone)/gelatin nanofibrous
scaffolds for nerve tissue engineering, Biomaterials (29 2008) 4532–4539.
Universities (CUSF-DH-D-2014011, and 2232014D3-15).
[25] D. Li, T. Wu, N. He, J. Wang, W. Chen, L. He, C. Huang, H.A. Ei-Hamshary, S.S.
Al-Deyab, Q. Ke, X. Mo, Three-dimensional polycaprolactone scaffold via
needleless electrospinning promotes cell proliferation and infiltration,
Appendix A. Supplementary data Colloids Surf. B: Biointerfaces 121 (2014) 432–443.
[26] X. Li, Y. Zhang, G. Qi, Evaluation of isolation methods and culture conditions
Supplementary data associated with this article can be found, in for rat bone marrow mesenchymal stem cells, Cytotechnology 65 (2013)
323–334.
the online version, at http://dx.doi.org/10.1016/j.colsurfb.2016.07.
[27] J. Wu, S. Liu, L. He, H. Wang, C. He, C. Fan, X. Mo, Electrospun nanoyarn scaffold
009. and its application in tissue engineering, Mater. Lett. 89 (2012) 146–149.
[28] B. Feng, H. Tu, H. Yuan, H. Peng, Y. Zhang, Acetic-acid-mediated miscibility
toward electrospinning homogeneous composite nanofibers of GT/PCL,
References Biomacromolecules 13 (2012) 3917–3925.
[29] D.H. Reneker, A.L. Yarin, Electrospinning jets and polymer nanofibers,
[1] S. Yang, K.-F. Leong, Z. Du, C.-K. Chua, The design of scaffolds for use in tissue Polymer (Guildf) 49 (2008) 2387–2425.
engineering. Part I. Traditional factors, Tissue Eng. 7 (2001) 679–689. [30] S.A. Catledge, W.C. Clem, N. Shrikishen, S. Chowdhury, A.V. Stanishevsky, M.
[2] Q.P. Pham, U. Sharma, A.G. Mikos, Electrospinning of polymeric nanofibers for Koopman, Y.K. Vohra, An electrospun triphasic nanofibrous scaffold for bone
tissue engineering applications: a review, Tissue Eng. 12 (2006) 1197–1211. tissue engineering, Biomed. Mater. 2 (2007) 142–150.
[3] J. Rnjak-Kovacina, A.S. Weiss, Increasing the pore size of electrospun scaffolds, [31] C.S. Ki, D.H. Baek, K.D. Gang, K.H. Lee, I.C. Um, Y.H. Park, Characterization of
Tissue Eng. Part B: Rev. 17 (2011) 365–372. gelatin nanofiber prepared from gelatin-formic acid solution, Polymer 46
[4] D.R. Nisbet, J.S. Forsythe, W. Shen, D.I. Finkelstein, M.K. Horne, Review paper: (2005) 5094–5102.
a review of the cellular response on electrospun nanofibers for tissue [32] C.T. Lim, E.P.S. Tan, S.Y. Ng, Effects of crystalline morphology on the tensile
engineering, J. Biomater. Appl. 24 (2009) 7–29. properties of electrospun polymer nanofibers, Appl. Phys. Lett. 92 (2008)
[5] W. He, Z. Ma, W.E. Teo, Y.X. Dong, P.A. Robless, T.C. Lim, S. Ramakrishna, 141908.
Tubular nanofiber scaffolds for tissue engineered small-diameter vascular [33] K. Sisson, C. Zhang, M.C. Farach-Carson, D.B. Chase, J.F. Rabolt, Fiber diameters
grafts, J. Biomed. Mater. Res. Part A 90 (2009) 205–216. control osteoblastic cell migration and differentiation in electrospun gelatin,
[6] P. Causin, R. Sacco, M. Verri, A multiscale approach in the computational J. Biomed. Mater. Res. Part A 94 (2010) 1312–1320.
modeling of the biophysical environment in artificial cartilage tissue [34] Y. Tang, C. Wong, H. Wang, A. Sutti, M. Kirkland, X. Wang, T. Lin,
regeneration, Biomech. Model. Mechanobiol. 12 (2013) 763–780. Three-dimensional tissue scaffolds from interbonded
[7] M.P. Prabhakaran, J.R. Venugopal, T.T. Chyan, L.B. Hai, C.K. Chan, A.Y. Lim, S. poly(epsilon-caprolactone) fibrous matrices with controlled porosity, Tissue
Ramakrishna, Electrospun biocomposite nanofibrous scaffolds for neural Eng. Part C: Methods 17 (2011) 209–218.
tissue engineering, Tissue Eng. Part A 14 (2008) 1787–1797. [35] C. Vaquette, J.J. Cooper-White, Increasing electrospun scaffold pore size with
[8] R.A. Franco, Y.K. Min, H.M. Yang, B.T. Lee, Fabrication and biocompatibility of tailored collectors for improved cell penetration, Acta Biomater. 7 (2011)
novel bilayer scaffold for skin tissue engineering applications, J. Biomater. 2544–2557.
Appl. 27 (2013) 605–615. [36] A. Yin, K. Zhang, M.J. McClure, C. Huang, J. Wu, J. Fang, X. Mo, G.L. Bowlin, S.S.
[9] M.M. Stevens, J.H. George, Exploring and engineering the cell surface Al-Deyab, M. El-Newehy, Electrospinning collagen/chitosan/poly(l-lactic
interface, Science 310 (2005) 1135–1138. acid-co-epsilon-caprolactone) to form a vascular graft: mechanical and
[10] I.K. Shim, M.R. Jung, K.H. Kim, Y.J. Seol, Y.J. Park, W.H. Park, S.J. Lee, Novel biological characterization, J. Biomed. Mater. Res. Part A 101 (2013)
three-dimensional scaffolds of poly(l-lactic acid) microfibers using 1292–1301.
electrospinning and mechanical expansion: fabrication and bone
regeneration, J. Biomed. Mater. Res. Part B Appl. Biomater. 95 (2010) 150–160.

S-ar putea să vă placă și