ArticlePDF Available

Design and Fabrication of Injectable Microcarriers Composed of Acellular Cartilage Matrix and Chitosan

Authors:
  • The Ohio State University Comprehensive Cancer Center-The James

Abstract and Figures

Cartilage is an avascular tissue with limited self-repair ability. Since the methods for treatment of cartilage defects have not been effective, new therapies based on tissue engineering are considered over the recent years. In this study, human cartilage tissue was decellularized and porous injectable microcarriers (MCs) composed of acellularextracellular matrix (ECM) of cartilage tissue and chitosan(CS), with different ECM weight ratios, were fabricated by electrosprayingtechnique to be used in the treatment of articular cartilage defects. Various properties of ECM/CS MCs such as microstructure, mechanical strength, water uptake behaviour, and biodegradability rate were investigated. MCs with 1% ECM and 2% CS show appropriate characteristics in terms of pore size, density, porosity, and mechanical properties. MTT cytotoxicity assays performed on chondrocyte cells cultured on ECM/CS MCs with various amounts of ECM showed that the sample with 1% ECM content had the greatest cell attachment. Overall, it can be concluded that the prepared MCs with 1% ECM could be a potential candidate to be used in cartilage tissue engineering applications.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=tbsp20
Journal of Biomaterials Science, Polymer Edition
ISSN: 0920-5063 (Print) 1568-5624 (Online) Journal homepage: http://www.tandfonline.com/loi/tbsp20
Design and fabrication of injectable microcarriers
composed of acellular cartilage matrix and
chitosan
Farzane Sivandzade & Shohreh Mashayekhan
To cite this article: Farzane Sivandzade & Shohreh Mashayekhan (2018) Design and fabrication
of injectable microcarriers composed of acellular cartilage matrix and chitosan, Journal of
Biomaterials Science, Polymer Edition, 29:6, 683-700, DOI: 10.1080/09205063.2018.1433422
To link to this article: https://doi.org/10.1080/09205063.2018.1433422
Accepted author version posted online: 26
Jan 2018.
Published online: 02 Feb 2018.
Submit your article to this journal
Article views: 22
View related articles
View Crossmark data
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION, 2018
VOL. 29, NO. 6, 683700
https://doi.org/10.1080/09205063.2018.1433422
Design and fabrication of injectable microcarriers composed
of acellular cartilage matrix and chitosan
FarzaneSivandzade and ShohrehMashayekhan
Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
ABSTRACT
Cartilage is an avascular tissue with limited self-repair ability. Since the
methods for treatment of cartilage defects have not been eective,
new therapies based on tissue engineering are considered over the
recent years. In this study, human cartilage tissue was decellularized
and porous injectable microcarriers (MCs) composed of acellular
extracellular matrix (ECM) of cartilage tissue and chitosan (CS), with
dierent ECM weight ratios, were fabricated by electrospraying
technique to be used in the treatment of articular cartilage defects.
Various properties of ECM/CS MCs such as microstructure, mechanical
strength, water uptake behaviour, and biodegradability rate were
investigated. MCs with 1% ECM and 2% CS show appropriate
characteristics in terms of pore size, density, porosity, and mechanical
properties. MTT cytotoxicity assays performed on chondrocyte cells
cultured on ECM/CS MCs with various amounts of ECM showed that
the sample with 1% ECM content had the greatest cell attachment.
Overall, it can be concluded that the prepared MCs with 1% ECM could
be a potential candidate to be used in cartilage tissue engineering
applications.
1. Introduction
e limited self-repair capacity of cartilage has posed signicant clinical challenges to the
elds of orthopedic surgery. Most of cartilage defects require the replacement or regenera-
tion of damaged articular cartilage [1]. Although dierent methods of treatments, such as
mosaicplasty, autologous chondrocyte implantation (ACI), osteochondral allogra trans-
plantation, microfracture, and prosthetic joint replacement, have been devised to repair car-
tilage defects over recent decades, there are still several limitations in producing long-lasting
cartilage with full biological activity [2,3]. Cartilage tissue engineering remains a promising
approach to repair cartilage defects in a functional manner [4]. For such applications, pri-
mary chondrocytes are combined with various scaolds designed with specic architectures
and made of dierent biomaterials, such as natural and synthetic polymers [5–8].
KEYWORDS
Cartilage tissue engineering;
extracellular matrix; chitosan;
microcarriers
ARTICLE HISTORY
Received 27 November 2017
Accepted24 January 2018
© 2018 Informa UK Limited, trading as Taylor & Francis Group
CONTACT Shohreh Mashayekhan mashayekhan@sharif.edu
684 F. SIVANDZADE AND S. MASHAYEKHAN
Recent studies have indicated that decellularized extracellular matrix (ECM) derived
from various tissues can be used to provide bioactive scaolds. Henrique et al. studied a
novel scaold derived from cartilaginous ECM that was used as a growth factor delivery
system to promote chondrogenesis of stem cells in articular cartilage regeneration [9]. In
another study, Cheng et al. investigated the cell growth, matrix accumulation and mechan-
ical properties of neocartilage formed by porcine articular chondrocytes on a porcine car-
tilage-derived matrix for using in cartilage tissue engineering [10]. Kang et al. studied
human cartilage ECM-derived 3D porous acellular scaold using autologous adipose-de-
rived mesenchymal stem cells (MSCs) for cartilage defect repair in in vivo cartilage tissue
engineering [11]. Yang et al. developed a cartilage ECM-derived porous scaold which
showed suitable biocompatibility and 3-D interconnected structure in both in vitro and
in vivo studies. eir results demonstrated appropriate cell repopulation and neocartilage
formation for ECM-derived scaolds [7]. It would be advantageous for a bioactive scaold
to support chondrocyte activity both in vitro and in vivo. is accelerates tissue growth and
improves associated functional biomechanical properties of the engineered construct [12].
Such tissue-derived scaolds may provide a natural microenvironment for cell migration
and dierentiation to promote cartilage tissue formation [13].
Chitosan (CS) is a deacetylated derivative of chitin, the second most abundant polysac-
charide in nature [14,15]. Because of its attractive properties including low immunogenicity,
antibacterial activity, biocompatibility, biodegradability and low cost, CS is recommended
as a suitable material among the applied natural polymers used in diverse biomedical appli-
cations such as tissue engineering, wound healing and drug delivery [14,16–18]. Dierent
blends of synthetic and natural polymer composed of CS have been investigated and showed
promising results both in vitro and in vivo studies in cartilage tissue engineering [19,20].
As for the scaold structure, microcarriers (MCs) have been recently used in several
approaches successfully to expand, redierentiate, and dierentiate chondrocytes because
of their ability to ll irregularly shaped defects by minimally invasive procedures [21].
Schrobback et al. investigated the proliferation and subsequent chondrogenic dierenti-
ation of freshly isolated adult human articular chondrocytes on porous gelatin MCs and
compared them to those expanded using traditional monolayers [22]. Hong et al. fabricated
an injectable scaold by combining collagen-coated polylactide (PLA) MCs and crosslink-
able CS hydrogel and demonstrated the potential application of the composite scaold
in orthopaedics [23]. Georgi et al. tested the use of MSC or chondrocyte laden MCs as
building blocks for cartilage tissue engineering. It led to a compact and ecient macrotis-
sue formation for engineered cartilage tissue [24]. Liao et al. developed a new injectable
three-dimensional alginate hydrogel loaded with biodegradable porous microspheres as a
promising approach for cartilage tissue engineering [21]. In fact, cell expansion on MCs
is a new culture method for chondrocytes leading to the enhancement of proliferation
as well as the conservation of chondrogenic phenotype. Moreover, the cells expanded on
MCs can be used as building units for cartilage tissue engineering constructs [24]. While
recent studies have either focused on non-ECM-derived MCs for engineered cartilage tissue
[23,24]or aimed at fabricating macro-scale cartilage ECM-derived scaolds [14], no study
has been carried out to fabricate and evaluate ECM-derived MCs in the eld of cartilage
tissue engineering. is is despite the fact that the application of such MCs is probably more
promising in cartilage regeneration due to both beneting from various aspects of MCs
potential in tissue engineering reviewed elsewhere [22–25]. Moreover, ECM incorporation
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 685
provides informational signals and certain unique composition, which mimick the natural
tissue environment. is, consequently, leads to proper tissue regeneration [7,11].
In our previous study, a hybrid macro-scale CS/ECM scaold, which showed desirable
properties with promissing potentials as a biomimetic matrix for articular cartilage tissue
engineering, was proposed [14]. e aim of this study was to fabricate porous MCs com-
posed of articular cartilage ECM and CS using ionic electrospraying technique. en, their
characteristics were evaluated in terms of physico-mechanical properties, in vitro enzymatic
biodegradation, and biocompatibility.
2. Materials and methods
2.1. Decellularization of articular cartilage for matrix preparation
e decellularization method was performed according to a previous report [14]. In sum-
mary, articular cartilage was obtained from adult human knee cartilage (Baqiyatallah
Hospital, Tehran, Iran). e samples were cut to 2×2mm slices, rinsed with phosphate
buer saline) PBS) (Sigma–Aldrich) immediately, and transferred to a freezer at −20°C
prior to use. Human articular cartilage pieces were rinsed with PBS, pH 7.4. en, they were
lyophilized for 24h at −50°C, 0.6mbar. e samples were then ground into ne powder for
40min with Mixer/Mill device. e cartilage powder was agitated in 0.25% (w/v) trypsin/
EDTA (Gibco) at 37°C for 24h and the solution was changed every 8h. Having rinsed
the samples with PBS for 30min, they were treated with 10mM hypotinic tris(hydroxym-
ethyl)aminomethane hydrochloride (Tris-HCl) (Sigma–Aldrich) solution for 24h followed
by an incubation in Triton X-100 (1% v/v in PBS) (Dae Jung) for another 24h. Finally, the
samples were rinsed in PBS for 24h and the PBS was changed every 8h. e decellularized
cartilage powder was stored at -20°C prior to use. All the solutions contained 1% penicillin/
streptomycin (Sigma–Aldrich) during the decellularization process.
2.2. Preparation of porous ECM/CSMCs
Various hybrid ECM/CS MCs were fabricated with the same diameter (400±50μm) using
CS (medium molecular weight chitosan powder with Mw= 280 kDa (Sigma–Aldrich))
and ECM solution as presented in Table 1. ECM solution was prepared by enzymatic
digestion according to our previous report [14]. In summary ECM powder was briey
digested in pepsin solution (1mg/ml in 0.1M HCl). In order to get a homogeneous solu-
tion, the suspension went under constant stirring for about 60h at room temperature to
complete the digestion process. CS solution (4% w/v acetic acid (Merck)) was prepared
by dissolving CS in 2% (v/v) acetic acid and stirring overnight. en, solubilized ECM
and CS were mixed in certain quantities along with the carbodiimide solution containing
40 mM1-ethyl-3-[3 dimethylaminopropyl] carbodiimide (EDC) (Sigma–Aldrich), 20mM
Table 1.Preparation data of ECM/CS MCs.
Sample C2E0 C2E0.5 C2E1 C2E1.5
CS solution in 2% acetic acid (w/v) 2% 4% 4% 4%
ECM solution in pepsin (w/v) 0% 1% 2% 3%
Final solution for making MCs 2% CS, 0% ECM 2% CS, 0.5% ECM 2% CS, 1% ECM 2% CS, 1.5%ECM
686 F. SIVANDZADE AND S. MASHAYEKHAN
(N-hydroxysuccinimide) (NHS) (Sigma–Aldrich), and 50mM 2-[N-morpholine] ehanesul-
fonic acid (MES) (Sigma–Aldrich) followed by stirring for 20h to reach a homogenous
mixture. e homogeneous solution was dropped into a crosslinker solution containing 3%
w/v sodium tripolyphosphate) TPP ((Sigma–Aldrich) through 16-gauge needle driven by
a syringe pump under a high voltage electrostatic eld to produce spherical shaped MCs.
e needle was electrically connected to the positive electrode of a high voltage electrostatic
generator, the negative electrode of which was electrically connected to an annular stainless
steel plate xed under the coagulation solution (Figure 1). e optimized potential between
the needle and the stainless steel plate and the pumping rate was obtained between 7 and
11 kV and 300–700 μl/min, respectively. e fabricated MCs were frozen overnight at
−20°C, followed by lyophilisation for 24h. en, the MCs were immersed into 1N NaOH
solution for 30min, rinsed by deionized water, and lyophilized again for 24h.
2.3. Dynamic cell culturing
Human primary chondrocyte cells were derived from the adult human articular cartilage.
Isolation of the cells was carried out as per the instructions by Eslaminejad et al. [26].
Chondrocytes were cultured in dulbecco’s modied eagle’s medium (DMEM) (Sigma–
Aldrich) supplemented with 10% fetal bovine serum (FBS) (Sigma–Aldrich). Cells were
maintained in a humidied CO
2
incubator at 37°C until reaching 90% conuence and were
fed by fresh medium every two days. e prepared porous MCs were sterilized using ethanol
(70%) and rinsed three times with PBS solution for 10min. en, they were incubated with
Figure 1.Schematic illustration of ECM/CS MCs fabrication by electrospraying technique.
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 687
DMEM for 24h at 37°C and 5% CO2. A 10mg mass of the sterilized MCs was immersed
in 1ml DMEM and added to a 24-well plate. e cell suspension with a concentration of
2×105 cells/cm2was also added to the plate. e cell-seeded MCs were incubated for 3h at
37°C and 5% CO2 [24]. e plate was then removed and the culture medium was replaced
with a fresh one. e plate was placed on a mini-rocker at 15rpm to facilitate the cell
expansion. Aerwards, it was incubated again at 37°C and 5% CO2 for a specied period
of time. e culture medium was replaced daily with a fresh one.
2.4. Characterization
2.4.1. Evaluation of the decellularization process
Parts of the cartilage samples (native and decellularized) were xed for 24h in 10% neutral
buered formalin solution in PBS (pH 7.4) at room temperature. Subsequently, the sam-
ples were rinsed anddehydrated in graded series of ethanol. en, they were embedded in
paran and sectioned with a thickness of 5μm using microtome. Hematoxylin and eosin
(H&E) staining was performed aer deparanization by xylene and rehydrationto validate
the chondrocytes removal from the decellularized cartilage tissue [7,24]. Moreover, total
DNA content was quantied in samples of native and decellularized tissue according tothe
protocol proposed by Rajabi-Zeleti et al. [27]. e amount of DNA was expressed in ng/
mg dry weight of samples.
2.4.2. Fourier transform infrared spectroscopy
e Fourier transform infrared spectroscopy (FTIR) spectra were recorded using a Shimadzu
FTIR spectrometer. All spectra were obtained from 400 to 4000cm−1 at a 4cm−1 resolution
at room temperature.
2.4.3. Morphological characterization
e morphology of porous MCs were investigated by scanning electron microscopy (SEM,
Seron Technology, AIS-2100, Korea). e porous MCs were coated with gold before obser-
vation. Image J analysis soware was used to determine the pore sizes. ree planned
locations were selected for three samples of MCs and a total of 40 measurements were
recorded at each location.
2.4.4. Porosity and density
To obtain the porosity (P) and density (ρs) of MCs, the total volume of MCs (Vs) and pore
volume (Vp) were determined. First, 20 MCs were randomly weighed (W0) and immersed
in ethanol (density ρ). en, the samples were dried in vacuum for 20 min and the excess
ethanol on the surface was removed with blotting paper. en the MCs were immediately
re-weighed (We). Porosity and density were calculated as follows:
(1)
P
=Vp
/
V
s
×
100
(2)
Vp
=
(
WeW0
)/𝜌
688 F. SIVANDZADE AND S. MASHAYEKHAN
2.4.5. Mechanical properties
Mechanical strength: In order to investigate the mechanical strength of MCs, 100 random
MCs were added to 40mL of water and stirred for 30min (400rpm). en, fragmented
MCs were separated and the residual (unfragmented) MCs were recounted. e mechanical
strength was estimated by noting the percentage of residual MCs [28].
Young’s modulus: e attened samples, which had the same concentrations as prepared
MCs, were generated. In summary, ECM powder was digested in pepsin solution (0.1mg/
100ml in 0.1N HCl) for 60h at 37°C. CS (4% w/v acetic acid) was prepared by dissolving
CS in 2% (v/v) acetic acid and stirring overnight. en, solubilised ECM and CS were
mixed in certain quantities along with carbodiimide solution (40mM EDC, 20mM NHS
and 50mM MES) and stirred for 20h to reach a homogenous mixture. Aerwards, 1ml of
the prepared mixture was poured to each chamber of the 24-well plates. en, they were
frozen overnight at -20°C and lyophilized for 24h. e dried porous scaolds were then
immersed into 1N NaOH solution for 30min, smoothly rinsed by deionized water and
lyophilized again for 24h. To measure the compressive properties, the lyophilized scaolds
were immersed in PBS for 1h at room temperature. en, the mechanical properties of
the scaolds were measured by compressing the hydrated scaolds up to 70% strain using
Hounseld H10KS mechanical tester with a load cell of 1 kN. e loading rate was 0.5mm/
min. e Young’s modulus was determined from the slope of the initial linear section of
stress-strain diagram.
2.4.6. Water Uptake behaviour and in vitro enzymatic biodegradation test
To assess water uptake ratio, 100 MCs of each samples were dried in a vacuum until a con-
stant weight was reached (W0) and then immersed in PBS for 3days at 37°C for pre-dened
time intervals (15, 30min, 1, 2, 4, 6, 8, 24, 48, and 72h). Having removed the excess PBS
on the surface with blotting paper, the MCs were removed from the PBS and weighed (Ww)
immediately. e water uptake ratio (WR) was dened as follows:
e in vitro enzymatic biodegradation test was examined by exposing the MCs in a PBS
solution containing 1.5 μg/ml lysozyme from chicken egg white (Sigma–Aldrich) at 37°C
for various periods up to 4weeks. In summary, the dried MCs (W0) were weighed and
incubated at 37°C in the enzymatic solution. Aerwards, the sample was weighed (Wd)
followed by drying at 60°C for 24h [17]. e extent of in vitro biodegradation rate (BR)
was dened as follows:
(3)
s=
0
V
(4)
WR
(%)=
W
w
W
0
W
0
×
100
(5)
BR
(%)=
W
0
W
d
W
0
×
100
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 689
2.4.7. In vitro cell culture studies
In order to determine the viability and proliferation of the cells cultured on dierent MCs,
3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide) MTT (assay was performed
on the1st, 3rd, 5th, and 7th days of culture. Briey, aer a specied period of incubation of
the cells on MCs, the culture medium in each well were replaced with a 10 μl MTT solution
(1mg/mL) with 400 μl Roswell Park Memorial Institute (RPMI) 1640 medium (Sigma–
Aldrich). Aer a 4h incubation at 37°C with5% of CO2, the medium in all wells were
replaced with 14.95ml isopropyl alcohol and 50 μl of HCl and incubated again for 30min
to ensure solubilizing of crystals. Absorbance was determined using a spectrophotometer
(BioPhotometer, Eppendorf, Germany) at 570nm [17].
2.4.8. Statistical analysis
Each experiment was replicated three times. Values were presented as the means of rep-
licates±standard error of the mean. Mean values were compared by one-way analysis of
variance (ANOVA) using SPSS.18 statistical soware package. P values less than 0.01 were
considered statistically signicant.
3. Results
3.1. Evaluation of decellularization process
Histological ndings from hematoxylin and eosinstaining conrmed that the decellulari-
zation process was successful as there were no visible cell nuclei, as compared to the intact
cartilage tissue (Figure 2(A)). Moreover, DNA quantication showed complete removal of
DNA following mechanical and chemical treatments with signicant dierences in DNA
content observed between native and decellularized tissue (Figure 2(B)).
3.2. FTIR spectroscopy
Figure 3 illustrates the results of the FTIR spectra of ECM, CS, and cross-linked C2E1 MCs.
e FTIR spectrum of CS shows characteristic absorption peaks at 3437, 1811, 1558,1471,
1400, 1205, 1076 and 856cm−1. e broad absorption peak at 3437and 3712cm−1 are
attributed to the amino (–NH2) and hydroxyl (–OH) bond, respectively. e peaks at 1076
and 859cm−1correspond to the saccharide structure of CS [29]. e characteristic peak at
1558cm−1 represents the amide I absorption band, while those at 1400 and 1205cm−1rep-
resent the amide III absorption band, which suggests CS is a partially deacetylated product
[17,30]. e ECM displays absorption peaks at 1680, 1550, and 1290cm−1, which repre-
sent the absorption bands of amide I (C=O bond), amide II (N–H and C–N bonds) and
amide III (C–N bond), respectively. Moreover, the ECM shows the presence of carboxyl
and amine (–NH2) groups at 3420cm−1. e comparison between the graphs of CS and
C2E1 MCs demonstrated signicant reduction in intensity of N-H peaks. is might be
due to the reaction between amino groups in CS and carboxyl group in ECM during the
cross-linking process by EDC/NHS. e intermolecular interaction between CS and ECM
is also conrmed by the peak between 1630 and 1690cm1that indicates the stretching
690 F. SIVANDZADE AND S. MASHAYEKHAN
band of C=O in amide group. Moreover, the absorption peaks of PO4
−3 in the region of
900–500cm−1 shows the existence of TPP, the ionic cross-linker in the prepared MCs [31].
3.3. Characterization of the prepared MCs in terms of physical and mechanical
properties
In this study, various hybrid ECM/CS MCs were fabricated with the same diameter
(400±50μm) using CS and ECM solution (Table 1). It is noteworthy that the MCs con-
taining more than 2% CS did not show spherical shape. MCs for tissue engineering should
have a highly porous structure to provide enough space for cell attachment and prolifera-
tion [32]. e eect of ECM on the pore parameters of the MCs are shown in Figure 4 and
the results are summarized in Table 2. Figure 4 indicates good porosity and pore-to-pore
interconnectivity within the MCs’ structure. e mean pore size of the MCs decreased
with the increase in ECM content. However, C2E1.5 MCs show the largest pore size and
non-uniform structure. is might be attributed to inecient mixing of the polymers due
to high ECM concentration. Moreover, the mechanical behaviour of the prepared MCs was
investigated in the equilibrium swelling state and the results are summarized in Table 2. By
adding ECM (from 0 to 1.5 wt%), Young’s modulus and mechanical strength decreased. As
shown in Table 2, by an increase in ECM, density of the fabricated MCs decreased, while
porosity increased.
3.4. Water uptake behaviour and in vitro biodegradation test
Water uptake behaviour is one of the most important properties of the scaold that depends
on pore size and interconnectivity between the pores to provide nutrients ow for the cells
[33,34]. e results of the water uptake ratio of the prepared MCs are illustrated in Figure 5.
e weight of all the MCs increased in the rst hours of imersion. Aer 8 h, all the samples
reached equilibrium. C2E1 MCs showed the highest water uptake ratio, compared with
other groups. C2E1.5 MCs showed lower water uptake ratio than other hybrid MCs. is
might be attributed to non-uniform porosity and lost structure.
As the eld of tissue engineering aims to regenerate damaged tissues, the scaold deg-
radation rate should match the new tissue formation rate to allow optimal regeneration.
e results of the in vitro biodegradation rate of the prepared MCs are illustrated in Figure
6. C2E1 MCs showed the highest degradation rate in comparison with the other groups.
ey reached their maximum weight loss of about 55% aer 28days of incubation. As
shown in Figure 6, the biodegradation rate increased with the increase in ECM content.
is might be due to higher porosity, better accessibility of cleavage sites by the enzymes,
or the increased hydrophilicity of the MCs caused by incorporation of higher ECM content.
However, C2E1.5 MCs showed lower biodegradation rate compared with the other hybrid
MCs. is might be attributed to non-uniform porosity and lost structure. Since C2E1.5
MCs demonstrated inappropriate mechanical and structural properties, in the next step,
other groups were examined for the biocompatibility analysis.
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 691
3.5. Cell proliferation and cytotoxicity analysis
e biocompatibility of the ECM/CSMCs was conrmed using primary chondrocyte cells
derived from human articular cartilage (Figure 7). e number of attached cells increased
over time during 7days on all MCs due to the high porosities and presence of ECM. e
number of cells on C2E1 MCs increased over time during 7-day culture more than the
other groups. Although the number of attached cells on 2D tissue cell plate (TCP) was
higher than those on all other groups of MCs until day5, it showed a decrease in attached
cells on day7due tothe limited area for cell attachment. In contrast, the MCs provided 3D
porous structure enhancing the overall surface area for cell adhesion and proliferation. By
comparing results of dynamic and static culture (Figure 7), it can be observed that dynamic
culture has a signicant inuence on the growth and proliferation of the attached cells. e
attached cells on CS/ECM MCs aer the 1st day of culture was examined by SEM which
further conrms that the fabricated MCs are nontoxic and biocompatible (data not shown).
4. Discussion
e fundamental concepts guiding the development of tissue-engineered scaolds depend
on the selected biomaterial or the method of scaold production [35]. New trends in tis-
sue engineering have focused on scaold materials mimicking the structure, morphology,
and biological cues of the natural environment [36]. Although various natural and syn-
thetic materials have been explored recently for cartilage tissue engineering, they have not
been considerably capable of mimicking the native ECM environment [37–41]. Herein, we
fabricated porous MCs composed of cartilage ECM and CS designed for cartilage tissue
engineering due to the high potential of biomimeticallyenhanced decellularized cartilage
tissue as a chondroinductive material for cartilage regeneration [7,9,10]. Cartilage ECM is
composed of type II collagen and an interlocking mesh of brous proteins, proteoglycans,
hyaluronic acid, and chondroitin sulfate, which provides cell retention, migration, prolifer-
ation and dierentiation [42]. In fact, the cartilage morphogenetic proteins retained in the
ECM, could serve as a potential source of endogenous biologically active molecules that can
promote cartilage formation [10]. In this study, we demonstrated the complete removal of
cellular contents through a decellularization protocol using SDS, trypsin, and triton-100X
[14,27]. e successful decellularization process was conrmed by H&E staining and DNA
quantication analysis (Figure 2). EDC and NHS were used as cross-linking agents to
Figure 2.(A) H&E staining of cartilage tissue. Cells and empty lacunasin native and decellularized tissue,
respectively are shown by arrows (×20 mag), (B) DNA quantification in native and decellularized tissue.
692 F. SIVANDZADE AND S. MASHAYEKHAN
form amide bonds between the primary amino group of CS and a carboxylic group of
ECM existing in collagen II [43] and glycosaminoglycan (GAG) [44] as recently described
elsewhere [14,45,46]. In order to generate an interconnected structure, hybrid MCs made
of decellularized cartilage matrix and CS were fabricated using an ionic electrospraying
technique. All the prepared MCs have a spherical shape providing a high contact surface for
cell attachment, hence increasing cell growth rate [47]. We have recently reported to apply
the same technique to fabricate porous gelatin-chitosan MCs for MSCs proliferation [48].
e chemical composition of ECM/CS MCs was characterized by FTIR analysis and
compared with its components, which include CS and ECM (Figure 4). e FTIR analysis of
MCs showed a signicant reduction in the free amino group (–NH
2
) with broad absorption
Table 2.Characteristics of the prepared porous MCs.
Sample
The average pore
sizes (μm) Porosity(%) Density (g/cm3)
Mechanical
strength(%)
Young’s modulus
(kPa)
C2E0 85±10 77%±4 1.19±0.01 90%±5 140±10
C2E0.5 50±5 81%±3 1.12±0.02 93%±4 150±10
C2E1 40±5 87%±3 1.06±0.04 84%±3 50±5
C2E1.5 150±15 91%±2 1.02±0.01 81%±3 10±1
Figure 3.FTIR spectra of the samples. (a) ECM, (b) C2E1 MCs, (c) CS.
Figure 4.SEM images of the MCs. (a) C2E0, (b) C2E0.5, (c) C2E1, (d) C2E1.5.
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 693
peaks at 3437cm
−1
and 3712cm
−1
, compared with CS. is might be due to the crosslinking
of (–NH
2
) groups of CS with the (–COOH) groups of ECM during the MCs formation. e
intermolecular interaction between CS and ECM is also conrmed by the peak between 1630
and 1690cm−1. is indicates the stretching band of C=O in the amide group. Moreover,
the results of the ECM and cross-linked samples are highly similar. erefore, it can be
concluded that the chemical reaction between the polymers does not disturb the original
ECM structure aer cross-linking.
Mean pore size and its distribution, pore interconnectivity, pore shape, and pore wall
roughness are important parameters that should be considered for designing appropriate
MCs [49]. In other words, the scaold must possess a highly porous structure with an open,
Figure 5.The result of water uptake behaviour test of the MCs.
Figure 6.The result of in vitro biodegradation test of the MCs.
694 F. SIVANDZADE AND S. MASHAYEKHAN
fully interconnected geometry which provides a large surface area allowing cell in growth
and uniform cell distribution. It should also facilitate the neovascularization of the con-
struct [50]. Moreover, the porous architecture can protect cells grown inside the pore from
physical damage exerted by the shear stress that occurs during cell culture or injection [51].
Hence, the pore size can play an important role in the biological function of MCs [10,52].
Moreover, it was discovered that water content, evaporation rate, and evaporation time of
the solvent considerably aect the pore parameters of the MCs [33,53,54]. Hence, all the
prepared MCs were kept in PBS solution for 48h to reach the equilibrium swelling state,
then freeze-dried at the same evaporation rate and time. e only variable is this process
was the amount of ECM. As Figure 4 shows, the SEM images of the fabricated MCs indi-
cated desirable porosity and pore-to-pore interconnectivity. e mean pore size of the MCs
decreased with the increase in ECM content. is can be due to an increase in the amide
band between (–NH2) groups of CS and (–COOH) groups of ECM existing in collagen II
[43] and glycosaminoglycan (GAG) [44]. Similarly, Vrana et al. indicated that amide band
between (–NH2) groups of CS and (–COOH) groups of collagen causes a reduction in the
pore size of collagen-based scaolds [55].
In this study, the eect of ECM content on MCs density and porosity was investigated.
Corresponding results are presented in Table 2. e density and porosity of the prepared
MCs decreased and increased with the increase in the ECM content, respectively. is can
be due to the stronger intermolecular interactions between molecules in hybrid MCs [55,56].
Scaolds with high porosity provide a high surface area for cell-matrix interactions and act
as a temporary template for the new tissues growth and reorganization. is, consequently,
increases cell adhesion, sucient space for ECM regeneration, and uniform and ecient
cell seeding [48]. e ideal porosity varies for dierent cells and tissues for a more ecient
cell/tissue in growth and regeneration [57]. In previous studies, the porosity percentage
was reported 70–92% in cartilage tissue engineering [49,58]. As illustrated in Table.2, the
porosity values of all fabricated MCs are in the range of 77–91%, which highly comply with
Figure 7.The results of the MTT assay for the cells cultured on the prepared MCs in dynamic culture(*
p<0.01 statistically significant).
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 695
the reported range. Moreover, it was postulated that in dynamic cell culturing, the MCs
density is an important factor. It is due to the fact that a buoyant density of between 1.03
and 1.10g/cm3 allows MCs suspension with slow agitation [25]. At a density lower than
1g/cm3, the MCs would be oating in the culture medium and the cell attachment would
not be uniform.
Mechanical stability of the MCs are of the most crucial properties for successful gra
uptake by the host tissues. Besides, the MCs must be compatible with and harmless to the
surrounding tissues. ey should be also capable of resisting structural collapse during in
vitro culture and in vivo implantation, which may cause necrosis and inammation [59]. e
eect of ECM content on the MCs mechanical properties was investigated since the main
purpose of the prepared MCs is to provide mechanical and structural support for attach-
ment and proliferation of the cells in a 3D environment which mimicks the native cartilage.
All the prepared MCs showed the ability to reabsorb water and retain their original shape
aer removing the mechanical load. is is an essential feature for engineered tissue since
it makes it capable of tolerating the pressures during physiological high repetitive loading
[60]. Both the mechanical strength and Young’s moduli of the prepared MCs decreased with
the increase in ECM content (Table 2). As anticipated, CS has a tightening eect. In fact,
as CS to ECM ratio increases, the mechanical strength and Young’s moduli of the prepared
MCs improves. Another study by Correia et al. showed a similar eect of hyaluronic acid
on the mechanical properties of the fabricated scaolds which decreased with increasing
hyaluronic acid content [61]. It should be noted that the biomechanical properties of MCs
would increase dramatically following in vivo implantation. It is due to cellular microen-
vironment changes and ECM secretion by chondrocytes in tissue-engineered device [37].
Overall, C2E0.5 and C2E1 MCs show appropriate characteristics in terms of pore size,
density, porosity, and mechanical properties.
Fluid absorption and exchange of nutrients and waste are critical features of scaolds
in tissue engineering applications [48,62]. e water uptake ratio depends on both the
scaold’s hydrophilic nature and microstructure [63]. As Figure 5 shows, the ratio of CS to
ECM considerably aected the MCs’ water uptake ratio andC2E1 MCs showed the highest
water uptake ratio. is might be due to an increase in hydrophilicity of the MCs caused
by incorporation of higher ECM content. In a similar study, Correia et al. showed the
eect of the collagen and hyaluronic acid presence in scaolds. e eects were similar to
those of ECM presence, leading to a signicant increase in absorption of water due to high
hydrophilicity of collagen and hyaluronic acid [61]. Although C2E1.5 contained a higher
amount of ECM in its component, it showed less PBS absorption, compared to C2E1 and
C2E0.5. In homogeneity of the polymeric solution caused poor mechanical properties and a
collapsed pore network which prevented retention of PBS in the MCs. erefore, it showed
a lower water uptake ratio in comparison with the other hybrid samples.
Scaolds need to provide an appropriate condition for the cells to attach, proliferate,
and synthesize their own matrix molecules. Upon forming new ECM by the cells during in
vivo tissue regeneration the scaold should degrade or be absorbed by the body gradually.
erefore, to have an optimal regeneration, the rate of degradation should keep up with
the speed of new tissue formation [64]. e rst four weeks following graing is a critical
period for cartilage regeneration [10]. erefore, the in vitro biodegradation of the prepared
MCs in PBS/lysozyme solution during four weeks was evaluated and the corresponding
results are presented in Figure 6. It can be observed that C2E1 MCs showed the highest
696 F. SIVANDZADE AND S. MASHAYEKHAN
rate of degradation. is might be due to increased hydrophilicity caused by the incorpo-
ration of greater ECM content, and therefore caused substantial degradation of the MCs
as it enhances the interaction of the biomaterial with the enzymatic solution. e increase
in the biodegradation rate of chitosan by adding hydrophilic material, such as gelatin [37]
and hyaluronic acid [61], was shown in previous studies. Correia et al. reported the same
eect for adding hyaluronic acid to CS on biodegradability as that of adding ECM to CS.
ey also indicated that the scaolds containing more hyaluronic acid show a higher rate
of biodegradation compared to other samples due to their higher porosity, accessibility
of cleavage sites by the enzymes, and the biodegradable nature of hyaluronic acid [61].
Although the ECM content of C2E1.5 was higher than other hybrid MCs, the percentage
of weight loss was not higher due to reduced uid uptake and lost structure leading to less
accessibility for enzymatic solution, hence less biodegradation.
Previous studies regarding primary chondrocyte cultures performed in monolayer and
pellet culture may have limitations in their applications while applying these studies to func-
tional tissue engineering in a human clinical setting [65,66]. Despite the numerous studies
on primary chondrocytes seeded on 3D scaolds [10,14,38], they may be less applicable
compared to MCs due to the fact that they require a surgery process to be transplanted in a
human body. us in the present study, the possibility of using MCs for primary chondrocyte
cultivation derived from human articular cartilage was investigated [10]. Despite the limited
access to primary chondrocytes and diculties of the isolation process, they are applicable
in tissue engineering and regenerative medicine due to their potential to avoid allogenic
rejection following transplantation [67] Herein, the ability of porous MCs to support the
cell growth without inducing cytotoxicity was evaluated using the MTT proliferation assay
during 7days of culture and the results are presented in Figure 7. As illustrated, all the MCs
showed a rising trend in all time intervals, meaning that MCs supported the attachment
and proliferation of chondrocytes. A signicant increase in the number of attached cells
during culturing on the MCs, compared to TCP, conrmed the unique properties of MCs,
such as higher porosity. e greatest attached cell number was that of C2E1 MCs during the
7-day dynamic culture. Similarly, Chang et al. observed that an increase in the trend of cell
attachment for a 3D porous scaold composed of polycaprolactone improved by collagen
for cartilage tissue engineering. is is due to the increase in the amount of collagen as a
biological material providing appropriate cell binding site and environmental cues to the
cells [39]. O’Brien et al. showed that pore size has a signicant eect on mechanical and
structural properties, and cell adhesion in collagen-GAG scaolds since pore size should
be large enough to help cell migration and small enough to provide sucient surface [68].
e collapsed oversized pore structure of C2E1.5 led to smaller adhesion area and, hence,
less cell viability, compared to other hybrid MCs. erefore, it seems that the biocompatible
properties of the ECM not only play a pivotal role in mimicking the native cartilage envi-
ronment for primary chondrocytes attachment and proliferation, but also in the incorpo-
ration of higher ECM content leading to favourable biochemical and structural properties
enhancing the biocompatible properties of MCs [33,48,69,70]. Hence, the proposed C2E1
MCs provide the most desirable biocompatible properties and support cell attachment and
proliferation in vitro.
While in our previous study, 2D macro-scale CS/ECM scaolds were fabricated, in this
study, porous MCs composed of the same materials were designed and fabricated in order
to provide injectable MCs for 3D cell cultivation in cartilage tissue engineering [14]. Overall,
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 697
the ndings of this study reveal that MCs composed of 1% ECM and 2% CS (C2E1 MCs)
can be considered as a potential candidate to be used in cartilage tissue engineering due
to their appropriate biocompatibility, biodegradability, and structural properties. Ongoing
studies focus on using fabricated porous, cell-laden MCs as building blocks for cartilage
tissue engineering.
5. Conclusion
In this study, we successfully designed and fabricated porous MCs composed of acellular
cartilage-derived ECM and CS due to their potential application in the regeneration of a
damaged cartilage following injury. MCs for cartilage regeneration must have key charac-
teristics, including appropriate mechanical strength, porous microstructure, and pore size
as well as controlled biodegradability and biocompatibility. Here, the eect of CS and ECM
content on various characteristics of the prepared MCs was investigated. Overall, based on
our observations, C2E1 MCs can be considered as a potential candidate to be used in car-
tilage tissue engineering applications due to appropriate biodegradation, suitable structural
properties, and favourable biocompatibility.
Acknowledgment
e authors are grateful to Dr Hasanzadeh, the head of Persian Orthopedic Trauma Association,
for providingthe required articular cartilage specimens during total knee operations. We are also
thankful to Mr Fallahfor isolating MSCs from the human articular cartilage specimens and Mr Liles
for help with the nal editing.
Disclosure statement
No potential conict of interest was reported by the authors.
References
[1] Lynn A, Brooks R, Boneld W, et al. Repair of defects in articular joints. J Bone Joint Surg.
2004;86:1093–1099.
[2] Iwasa J, Engebretsen L, Shima Y, et al. Clinical application of scaolds for cartilage tissue
engineering. Knee Surg Sports Traumatol Arthrosc. 2009;17:561–577.
[3] Eslaminejad MB , Zomorodian E, Bagheri F. Mesenchymal stem cells in bone and cartilage
regeneration. Regenerative medicine and cell therapy. Totowa (NJ): Humana Press; 2013.
p. 131–153.
[4] S Dhinsa BS, B Adesida AB. Current clinical therapies for cartilage repair, their limitation and
the role of stem cells. Curr Stem Cell Res er. 2012;7:143–148.
[5] Doulabi AH, Mequanint K, Mohammadi H. Blends and nanocomposite biomaterials for
articular cartilage tissue engineering. Materials. 2014;7:5327–5355.
[6] Yoon IS, Chung CW, Sung JH, et al. Proliferation and chondrogenic dierentiation of human
adipose-derived mesenchymal stem cells in porous hyaluronic acid scaold. J Biosci Bioeng.
2011;112:402–408.
[7] Yang Q, Peng J, Guo Q, et al. A cartilage ECM-derived 3-D porous acellular matrix scaold for
in vivo cartilage tissue engineering with PKH26-labeled chondrogenic bone marrow-derived
mesenchymal stem cells. Biomaterials. 2008;29:2378–2387.
[8] Park H, Choi B, Hu J, et al. Injectable chitosan hyaluronic acid hydrogels for cartilage tissue
engineering. Acta Biomater. 2013;9:4779–4786.
698 F. SIVANDZADE AND S. MASHAYEKHAN
[9] Henrique V, Almeida YL, Grinne M, et al. Controlled release of transforming growth factor-b3
from cartilage extra-cellular-matrix-derived scaolds to promote chondrogenesis of human-
joint-tissue-derived stem cells. Acta Biomater.. 2014;10:4400–4409.
[10] Cheng NC, Estes BT, Young TH, et al. Engineered cartilage using primary chondrocytes cultured
in a porous cartilage-derived matrix. Regenerat Med. 2011;6:81–93.
[11] Kang H, Peng J, Lu S, et al. In vivo cartilage repair using adipose-derived stem cell-loaded
decellularized cartilage ECM scaolds. J Tissue Eng Regenerat Med. 2014;8:442–453.
[12] Park H, Lim D-J, Lee S-H, et al. Nanobrous mineralized electrospun scaold as a substrate for
bone tissue regeneration. J Biomed Nanotechnol. 2016;12:2076–2082.
[13] Ponce Márquez SP, Martínez VS, McIntosh Ambrose WM, et al. Decellularization of bovine
corneas for tissue engineering applications. Acta Biomater. 2009;5:1839–1847.
[14] Nasiri B, Mashayekhan S. Fabrication of porous scaolds with decellularized cartilage matrix
for tissue engineering application. Biologicals. 2017;48:39–46.
[15] Wang Q, Xu J, Jin H, et al. Articial periosteum in bone defect repair – a review. Chin Chem
Lett. 2017;28:1801–1807.
[16] Prabaharan M. Chitosan-based nanoparticles for tumor-targeted drug delivery. Int J Biol
Macromol. 2015;72:1313–1322.
[17] Baniasadi H. Fabrication and characterization of conductive chitosan/gelatin-based scaolds
for nerve tissue engineering. Int J Biol Macromol. 2015;74:360–366.
[18] Archana D, Dutta PK, Dutta J. Chitosan: a potential therapeutic dressing material for wound
healing. In Chitin and Chitosan for Regenerative Medicine. India: Springer; 2016. p. 193–227.
[19] Lao L, Tan H, Wang Y, et al. Chitosan modied poly(l-lactide) microspheres as cell microcarriers
for cartilage tissue engineering. Colloids Surf B. 2008;66:218–225.
[20] Yan J, Li X, Liu L, et al. Potential use of collagen-chitosan-hyaluronan tri-copolymer scaold for
cartilage tissue engineering. Articial Cells Blood Substit Biotechnol. 2006;34:27–39.
[21] Liao J, Wang B, Huang Y, et al. Injectable alginate hydrogel cross-linked by calcium gluconate-
loaded porous microspheres for cartilage tissue engineering. ACS Omega. 2017;2:443–454.
[22] Schrobback K, Klein TJ, Schuetz M, et al. Adult human articular chondrocytes in a microcarrier-
based culture system: expansion and redierentiation. J Orthop Res. 2011;29:539–546.
[23] Hong Y, Gong Y, Gao C, et al. Collagen-coated polylactide microcarriers/chitosan hydrogel
composite: injectable scaold for cartilage regeneration. J Biomed Mater Res Part A.
2008;85A:628–637.
[24] Georgi N, Blitterswijk C, Karperien M. MSC or chondrocyte seeded microcarriers as building
blocks for cartilage tissue engineering. Tissue Eng Part A. 2013;20:2513–2523.
[25] Chen A, Chen XiaoliL, Choo Andre Boon HwaX, et al. Critical microcarrier properties aecting
the expansion of undierentiated human embryonic stem cells. Stem Cell Res. 2011;7:97–111.
[26] E slaminejad MB, Taghiyar L, Falahi F. Quantitative analysis of the proliferation and dierentiation
of rat articular chondrocytes in alginate 3D culture. Iranian Biomed J. 2009;13:153.
[27] Rajabi-Zeleti S, Jalili-Firoozinezhad S, Azarnia M, et al. e behavior of cardiac progenitor cells
on macroporous pericardium-derived scaolds. Biomaterials. 2014;35:970–982.
[28] Guo T, Xia Y, Hao G, et al. Adsorptive separation of hemoglobin by molecularly imprinted
chitosan beads. Biomaterials. 2004;25:5905–5912.
[29] akur VK, akur MK. Handbook of sustainable polymers. Boca Raton (FL): CRC Press; 2016.
[30] Yan LP, Wang YJ, Ren L, et al. Genipin-cross-linked collagen/chitosan biomimetic scaolds for
articular cartilage tissue engineering applications. J Biomed Mater Res Part A. 2010;95A:465–
475.
[31] Stoch A, Jastrzębski W, Brożek A, et al. FTIR absorption–reection study of biomimetic growth
of phosphates on titanium implants. J Mol Struct. 2000;555:375–382.
[32] Matsiko A, Levingstone TJ, O’Brien FJ. Advanced strategies for articular cartilage defect repair.
Materials. 2013;6:637–668.
[33] Baniasadi H, Ramazani SAA, Mashayekhan S, et al. Design, fabrication, and characterization
of novel porous conductive scaolds for nerve tissue engineering. Int J Polym Mater Polym
Biomater. 2015;64:969–977.
JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 699
[34] Peng Z, Peng Z, Shen Y. Fabrication and properties of gelatin/chitosan composite hydrogel.
Polymer-Plast Technol Eng. 2011;50:1160–1164.
[35] Spector M. Biomaterials-based tissue engineering and regenerative medicine solutions to
musculoskeletal problems. Swiss Med Wkly. 2006;136:293–301.
[36] Lutolf M, Hubbell J. Synthetic biomaterials as instructive extracellular microenvironments for
morphogenesis in tissue engineering. Nat Biotechnol. 2005;23:47–55.
[37] Whu SW, Hung K-C, Hsieh K-H, et al. In vitro and in vivo evaluation of chitosan–gelatin
scaolds for cartilage tissue engineering. Mater Sci Eng C. 2013;33:2855–2863.
[38] Mallick SP, Pal K, Rastogi A, et al. Evaluation of poly (L-lactide) and chitosan composite scaolds
for cartilage tissue regeneration. Des Monomers Polym. 2016;19:271–282.
[39] Chang KY, Hung LH, Chu I, et al. e application of type II collagen and chondroitin sulfate
graed PCL porous scaold in cartilage tissue engineering. J Biomed Mater Res Part A.
2010;92A:712–723.
[40] Kuo Y-C, Ku H-F, Rajesh R. Chitosan/γ-poly (glutamic acid) scaolds with surface-modied
albumin, elastin and poly-l-lysine for cartilage tissue engineering. Mater Sci Eng C. 2017;78:265–
277.
[41] Kim DK, In Kim JI, Sim BR, et al. Bioengineered porous composite curcumin/silk scaolds for
cartilage regeneration. Mater Sci Eng C. 2017;78:571–578.
[42] Gao Y, Liu S, Huang J, et al. e ECM-cell interaction of cartilage extracellular matrix on
chondrocytes. Biomed Res Int. 2014;2014. Article ID: 648459.
[43] Mendoza-Novelo B, Cauich-Rodríguez JV. Decellularization, stabilization and functionalization
of collagenous tissues used as cardiovascular biomaterials. Biomaterials-Physics and
Chemistry.China:InTech; 2011.
[44] Prestwich GD, Shu XZ, Liu Y, et al. Injectable synthetic extracellular matrices for tissue
engineering and repair. Tissue engineering.Boston (MA): Springer; 2006.p. 125–133.
[45] Olde Damink LO, Dijkstra P, van Luyn M, et al. Cross-linking of dermal sheep collagen using
a water-soluble carbodiimide. Biomaterials. 1996;17:765–773.
[46] Pieper J, Oosterhof A, Dijkstra P, et al. Preparation and characterization of porous crosslinked
collagenous matrices containing bioavailable chondroitin sulphate. Biomaterials. 1999;20:847–
858.
[47] Frederick C. Biomaterials aspects of porous microcarriers for animal cell culture. Trends
Biotechnol. 1990;8:131–136.
[48] Karimian S.A. M, Mashayekhan S, Baniasadi H. Fabrication of porous gelatin-chitosan
microcarriers and modeling of process parameters via the RSM method. Int J Biol Macromol.
2016;88:288–295.
[49] Matsiko AL, Levingstone TJ, O'Brien FergalJ. Advanced strategies for articular cartilage defect
repair. Materials. 2013;6:637–668.
[50] Amoabediny G, Salehi-Nik N, Heli B. e role of biodegradable engineered scaold in tissue
engineering. Biomaterials Science and Engineering. China:InTech; 2011.
[51] Choi S-W, Zhang Y, Yeh Y-C, et al. Biodegradable porous beads and their potential applications
in regenerative medicine. J Mater Chem. 2012;22:11442–11451.
[52] KIM TK, YOON JJ, LEE DS, et al. Gas foamed open porous biodegradable polymeric
microspheres. Biomaterials. 2006;27:152–159.
[53] Wan Y, Wu H, Wen D. Porous-conductive chitosan scaolds for tissue engineering, 1. Macromol
Biosci. 2004;4:882–890.
[54] Bhardwaj N, Nguyen QT, Chen AC, et al. Potential of 3-D tissue constructs engineered from
bovine chondrocytes/silk broin-chitosan for in vitro cartilage tissue engineering. Biomaterials.
2011;32:5773–5781.
[55] Vrana N, Builles N, Kocak H, et al. EDC/NHS cross-linked collagen foams as scaolds for
articial corneal stroma. J Biomater Sci. 2007;18:1527–1545.
[56] Fan J, Shang Y, Yuan Y, et al. Preparation and characterization of chitosan/galactosylated
hyaluronic acid scaolds for primary hepatocytes culture. J Mater Sci Mater Med. 2010;21:319–
327.
700 F. SIVANDZADE AND S. MASHAYEKHAN
[57] Dhandayuthapani B, Yoshida Y, Maekawa T, et al. Polymeric scaolds in tissue engineering
application: a review. Int J Polym Sci. 2011;2011:19. Article ID: 290602.
[58] Yan LP, Oliveira JM, Oliveira AL, et al. Macro/microporous silk broin scaolds with potential
for articular cartilage and meniscus tissue engineering applications. Acta Biomater. 2012;8:289–
301.
[59] Gao S, Wang S, Ramakrishna S. Development of brous biodegradable polymer conduits for
guided nerve regeneration. J Mater Sci Mater Med. 2005;16:367–375.
[60] Ge Z, Li C, Heng BC, et al. Functional biomaterials for cartilage regeneration. J Biomed Mater
Res Part A. 2012;100:2526–2536.
[61] Correia CR, Moreira Teixeira LS, Moroni L, et al. Chitosan scaolds containing hyaluronic acid
for cartilage tissue engineering. Tissue Eng Part C Methods. 2011;17:717–730.
[62] Yan LP, Wang YJ, Ren L, et al. Genipin-cross-linked collagen/chitosan biomimetic scaolds for
articular cartilage tissue engineering applications. J Biomed Mater Res Part A. 2010;95A:465–
475.
[63] Ma L, Gao C, Mao Z, et al. Collagen/chitosan porous scaolds with improved biostability for
skin tissue engineering. Biomaterials. 2003;24:4833–4841.
[64] Alizadeh M, Abbasi F, Khoshfetrat A, et al. Microstructure and characteristic properties of
gelatin/chitosan scaold prepared by a combined freeze-drying/leaching method. Mater Sci
Eng C. 2013;33:3958–3967.
[65] Giannoni P, Crovace A, Malpeli M, et al. Species variability in the dierentiation potential of
in vitro -expanded articular chondrocytes restricts predictive studies on cartilage repair using
animal models. Tissue Eng. 2005;11:237–248.
[66] Schulze-Tanzil G, Müller R, Kohl B, et al. Diering in vitro biology of equine, ovine, porcine
and human articular chondrocytes derived from the knee joint: an immunomorphological
study. Histochem Cell Biol. 2009;131:219–229.
[67] Adkisson HD IV, Martin JA, Amendola RL, et al. e potential of human allogeneic juvenile
chondrocytes for restoration of articular cartilage. Am J Sports Med. 2010;38:1324–1333.
[68] O’Brien FJ, Harley B, Yannas IV, et al. e eect of pore size on cell adhesion in collagen-GAG
scaolds. Biomaterials. 2005;26:433–441.
[69] Choi B, Kim S, Lin B, et al. Cartilaginous extracellular matrix-modied chitosan hydrogels for
cartilage tissue engineering. ACS Appl Mater Interfaces. 2014;6:20110–20121.
[70] Almeida H, Eswaramoorthy R, Cunnie G, et al. Fibrin hydrogels functionalized with cartilage
extracellular matrix and incorporating freshly isolated stromal cells as an injectable for cartilage
regeneration. Acta Biomater. 2016;36:55–62.
... Most of today's tissue engineering treatments for articular cartilage defects require opening the joint cavity to implant bioscaffolds, a process that damages the tissue surrounding the joint. Because of its small diameter, microcarriers can be implanted into the cartilage defect site by injection, thereby reducing the damage to the periarticular tissue caused by the surgical incision [48]. ...
... In addition, some studies [37] have shown that microcarriers whose structures are more similar to the cartilage tissue's native ECM perform better in repairing articular cartilage damage. In addition, other studies [48] have shown that there is a higher rate of cell attachment if the cartilage tissue microcarriers contain ECM. Therefore, we reasoned that if the native ECM could be directly incorporated or generated in microcarriers with appropriate physical characteristics, the effect of cartilage repair would be improved. ...
Chapter
Full-text available
Articular cartilage plays an important role in daily joint activities. With the aging of the social population, the degenerative cartilage injury and the sports injury caused by inappropriate exercise of young patients, etc., the incidence rate of articular cartilage injury is constantly rising, and the injured patients tend to be younger. Although articular cartilage has its corresponding metabolic activities, it is difficult to recover and regenerate itself once it is damaged due to lack of nerve, blood vessel, and lymphatic tissue Common articular cartilage injuries can be divided into three types according to the degree of injury: partial cartilage injury, full-thickness cartilage injury, and osteochondral defect. If partial cartilage damage and full-thickness cartilage damage are not found and treated in time in the early stage, further deterioration will lead to serious osteochondral defects. After the corresponding subchondral bone injury, the upward invasion of the upper cartilage layer will also cause the overall osteochondral injury. Therefore, whether the osteochondral injury caused by the top-down or the osteochondral injury caused by the bottom-up, it seriously affects the normal activities of human joints. It not only brings great inconvenience to the daily life of patients, but also causes huge economic and psychological burden to patients. At the same time, it also consumes a large number of social public medical resources. Therefore, seeking an effective osteochondral repair strategy is not only the urgent need and hope of the society, but also one of the clinical scientific problems that clinicians and scientists urgently need to solve.
... The findings from bioink CLSM characterisation suggested that the ECM protein composition can be integrated into biomimetic tissue constructs, serving as a potential microenvironment for the proliferation of chondrocytes and BMSCs. A recent investigation of porous injectable micro-carriers (MCs) composed of acellular ECM from cartilage tissue and chitosan demonstrated that MCs with 1% ECM exhibited optimal cell attachment, suggesting that these MCs could be potential candidates for cartilage tissue engineering applications 24 . This finding supports the current work involving the integration of dECM bioink with the biopolymers such as, alginate and with alginate-chitosan composite, representing a strategic 3D approach in cartilage tissue engineering. ...
Article
Full-text available
In vitro use of articular cartilage on an organ-on-a-chip (OOAC) via microfluidics is challenging owing to the dense extracellular matrix (ECM) composed of numerous protein moieties and few chondrocytes, which has limited proliferation potential and microscale translation. Hence, this study proposes a novel approach for using a combination of biopolymers and decellularised ECM (dECM) as a bioink additive in the development of scalable OOAC using a microfluidic platform. The bioink was tested with native chondrocytes and mesenchymal stem cell-induced chondrocytes using biopolymers of alginate and chitosan composite hydrogels. Two-dimensional (2D) and three-dimensional (3D) biomimetic tissue construction approaches have been used to characterise the morphology and cellular marker expression (by histology and confocal laser scanning microscopy), viability (cell viability dye using flow cytometry), and genotypic expression of ECM-specific markers (by quantitative PCR). The results demonstrated that the bioink had a significant impact on the increase in phenotypic and genotypic expression, with a statistical significance level of p < 0.05 according to Student’s t-test. The use of a cell-laden biopolymer as a bioink optimised the niche conditions for obtaining hyaline-type cartilage under culture conditions, paving the way for testing mechano-responsive properties and translating these findings to a cartilage-on-a-chip microfluidics system.
... The group first solubilized dECM with acetic acid and then electrospray dECM droplets onto aluminum foil and collected them with a smaller size distribution of 225 ± 67 nm and a higher zeta potential (10 ± 1.6 mV), The dECM particles/powder produced by electrospray have a natural fiber-like structure, contain various ECM proteins, and are easily resuspended in the buffer and gelatin solution [74]. Farzane Sivandzade et al. adopted crosslinking techniques to obtain injectable microcarriers with fibrous structure by creating an enzymatically digested ECM solution mixed with a Chitosan solution containing crosslinking agents, which was then dropped into a crosslinker solution through a 16-gauge needle driven by a syringe pump under a high-voltage electrostatic field to produce spherical-shaped microcarriers [75]. The microdroplets have key characteristics, including appropriate mechanical strength, porous microstructure, and pore size, as well as controlled biodegradability and biocompatibility, and are therefore promising in skeletal tissue engineering. ...
Article
Full-text available
The decellularized extracellular matrix (dECM) is capable of promoting stem cell proliferation, migration, adhesion, and differentiation. It is a promising biomaterial for application and clinical translation in the field of periodontal tissue engineering as it most effectively preserves the complex array of ECM components as they are in native tissue, providing ideal cues for regeneration and repair of damaged periodontal tissue. dECMs of different origins have different advantages and characteristics in promoting the regeneration of periodontal tissue. dECM can be used directly or dissolved in liquid for better flowability. Multiple ways were developed to improve the mechanical strength of dECM, such as functionalized scaffolds with cells that harvest scaffold-supported dECM through decellularization or crosslinked soluble dECM that can form injectable hydrogels for periodontal tissue repair. dECM has found recent success in many periodontal regeneration and repair therapies. This review focuses on the repairing effect of dECM in periodontal tissue engineering, with variations in cell/tissue sources, and specifically discusses the future trend of periodontal regeneration and the future role of soluble dECM in entire periodontal tissue regeneration.
... Apart from solid spherical particles, octopus-shaped [31], asymmetric dumbbell-shaped [32], and porous microcarriers [33,34] are also manufactured for large-scale cell cultures. The advantages of these microcarrierbased expansion systems include: (i) ease of scale up [29,[35][36][37], (ii) ability to precisely control cell growth conditions within large-scale bioreactors [38,39], (iii) reduction in bioreactor volumes and the floor spaces required for given-sized manufacturing operations [40], (iv) reduction in technician labor [37,39], and (v) more natural culture environments for cell proliferation and differentiation [38,41]. All these benefits have enabled the microcarrierbased culture technology to be generally employed for industrial production of mammalian cells [37,42], protein-based therapeutics [29], and for research purposes [35]. ...
Article
Full-text available
Developmental engineering (DE) aims to culture mammalian cells on corresponding modular scaffolds (scale: micron to millimeter), then assemble these into functional tissues imitating natural developmental biology processes. This research intended to investigate the influences of polymeric particles on modular tissue cultures. When poly(methyl methacrylate) (PMMA), poly(lactic acid) (PLA) and polystyrene (PS) particles (diameter: 5–100 µm) were fabricated and submerged in culture medium in tissue culture plastics (TCPs) for modular tissue cultures, the majority of adjacent PMMA, some PLA but no PS particles aggregated. Human dermal fibroblasts (HDFs) could be directly seeded onto large (diameter: 30–100 µm) PMMA particles, but not small (diameter: 5–20 µm) PMMA, nor all the PLA and PS particles. During tissue cultures, HDFs migrated from the TCPs surfaces onto all the particles, while the clustered PMMA or PLA particles were colonized by HDFs into modular tissues with varying sizes. Further comparisons revealed that HDFs utilized the same cell bridging and stacking strategies to colonize single or clustered polymeric particles, and the finely controlled open pores, corners and gaps on 3D-printed PLA discs. These observed cell–scaffold interactions, which were then used to evaluate the adaptation of microcarrier-based cell expansion technologies for modular tissue manufacturing in DE.
... The main cartilage regeneration approaches include microfracture [30,31], molecular delivery [32,33], tissue grafting [34,35], chondrocyte or stem cell transplantation, either with or without scaffold [36,37], and various acellular biomaterial approaches [38,39]. Autogenous chondrocyte implantation, mosaicplasty, or bone marrow stimulating procedures, such as microfracture and multiple drilling, are favored alternatives to joint replacement therapy in young patients with knee cartilage defects. ...
Article
Cartilage tissue engineering is becoming increasingly popular for repairing cartilage defects. However, lack of nerves and blood vessels, as well as low cell density cause inadequate cartilage tissue regeneration. As a result, it is critical to create an appropriate microenvironment for chondrocyte proliferation. Hydrogels, as three-dimensional cross-linked polymeric networks, are considered promising alternatives to cartilage tissue due to their high-water content, viscoelasticity, and resemblance to the articular cartilage extracellular matrix. Silk fibroin hydrogels of Bombyx mori, a natural polymer, have recently been widely used to regenerate cartilage due to their ability to mimic the extracellular matrix of cartilage and endurability. For this purpose, silk fibroin is frequently combined with other materials. Composite silk fibroin hydrogels have shown strong potential for cartilage regeneration. This review explains various composite silk fibroin hydrogels studied for cartilage regeneration, as well as their advantages over single SF scaffolds.
Article
Tissue engineering and regenerative medicine have shown potential in the repair and regeneration of tissues and organs via the use of engineered biomaterials and scaffolds. However, current constructs face limitations in replicating the intricate native microenvironment and achieving optimal regenerative capacity and functional recovery. To address these challenges, the utilization of decellularized tissues and cell-derived extracellular matrix (ECM) has emerged as a promising approach. These biocompatible and bioactive biomaterials can be engineered into porous scaffolds and grafts that mimic the structural and compositional aspects of the native tissue or organ microenvironment, both in vitro and in vivo. Bioactive dECM materials provide a unique tissue-specific microenvironment that can regulate and guide cellular processes, thereby enhancing regenerative therapies. In this review, we explore the emerging frontiers of decellularized tissue-derived and cell-derived biomaterials and bio-inks in the field of tissue engineering and regenerative medicine. We discuss the need for further improvements in decellularization methods and techniques to retain structural, biological, and physicochemical characteristics of the dECM products in a way to mimic native tissues and organs. This article underscores the potential of dECM biomaterials to stimulate in situ tissue repair through chemotactic effects for the development of growth factor and cell-free tissue engineering strategies. The article also identifies the challenges and opportunities in developing sterilization and preservation methods applicable for decellularized biomaterials and grafts and their translation into clinical products.
Article
The growth and repair of skeletal muscle are due in part to activation of muscle precursor cells, commonly known as satellite cells or myoblasts. In order to acquire enough cells for neoskeletal muscle regeneration, it is urgent to develop microcarriers for skeletal myoblasts proliferation with a considerable efficiency. The current study was thus proposed to develop a microfluidic technology to manufacture porous poly(l-lactide-co-ε-caprolactone) (PLCL) microcarriers of high uniformity, and porosity was manipulated via camphene to suit the proliferation of C2C12 cells. A co-flow capillary microfluidic device was first designed to obtain PLCL microcarriers with different porosity. The attachment and proliferation of C2C12 cells on these microcarriers were evaluated and the differentiation potential of expanded cells were verified. The obtained porous microcarriers were all uniform in size with a high mono-dispersity (CV < 5 %). The content of camphene rendered effects on the size, porosity, and pore size of microcarriers, and porous structure addition produced a softening of their mechanical properties. The one of 10 % camphene (PM-10) exhibited the superior expansion for C2C12 cells with the number of cells after 5 days of culture reached 9.53 times of the adherent cells on the first day. The expanded cells from PM-10 still retained excellent myogenic differentiation performance as the expressions of MYOD, Desmin and MYH2 were intensively enhanced. Hence, the current developed porous PLCL microcarriers could offer as a promising type of substrates not only for in vitro muscular precursor cells expansion without compromising any multipotency but also have the potential as injectable constructs to mediate muscle regeneration.
Article
Full-text available
Articular cartilage (AC) is the thin tissue that covers the long bone ends in the joints and that ensures the transmission of forces between adjacent bones while allowing nearly frictionless movements between them. AC repair is a technologic and scientific challenge that has been addressed with numerous approaches. A major deadlock is the capacity to take in account its complex mechanical properties in repair strategies. In this review, we first describe the major mechanical behaviors of AC for the non‐specialists. Then, we show how researchers have progressively identified specific mechanical parameters using mathematical models. There are still gaps in our understanding of some of the observations concerning AC biomechanical properties, particularly the differences in extracellular matrix stiffness measured at the microscale and at the millimetric scale. Nevertheless, for bioengineering applications, AC repair strategies must take into account what are commonly considered the main mechanical features of cartilage: its ability to withstand high stresses through three main behaviors (elasticity, poroelasticity and swelling). Finally, we emphasize that future studies need to investigate AC mechanical properties at different scales, particularly the gradient of mechanical properties around cells and across the cartilage depth, and the differences in mechanical properties at different scales. This multi‐scale approach could greatly enhance the success of AC restorative approaches.
Article
Full-text available
Chitosan microcapsules draw attention due to their biodegradability, biocompatibility, antibacterial behavior, low cost, easy processing, and the capability to be used for different applications. This study utilized the electrospraying technique for the chitosan microcapsules formulation. As a novel cross-linking agent, a mixture of oxalic acid and sodium phosphate dibasic was utilized as a collecting solution for the first time in the electrospraying of chitosan microcapsules. Scanning Electron Microscopy (SEM) was utilized to optimize the spherical morphology and size of the experimentally obtained microcapsules. The different parameters, including chitosan concentration, applied voltage, flow rate, and tip-to-collector (TTC) distance, affecting the microcapsules’ size, sphericity, yield, and combined effects were optimized using Surface Responses Methodology (RSM). The Analysis of Variance (ANOVA) was utilized to obtain the impact of each parameter on the process responses. Accordingly, the results illustrated the significant impact of the voltage parameter, with the highest F-values and least p-values, on the capsule size, sphericity, and yield. The predicted optimum conditions were determined as 5 wt% chitosan concentration, 7 mL/h flow rate, 22 kV, and 8 cm TTC distance. The predicted responses at the optimized conditions are 389 µm, 0.72, and 80.6% for the capsule size, sphericity, and yield, respectively. While the validation of the model prediction was conducted experimentally, the obtained results were 369.2 ± 23.5 µm, 0.75 ± 0.04, and 87.3 ± 11.4%, respectively. The optimization process was successfully examined for the chitosan microcapsules manufacturing.
Article
Full-text available
A great interest has been shown in the injectable scaffolds for cartilage tissue regeneration because it can fill irregularly shaped defects easily through minimally invasive surgical treatments. Herein, we developed a new injectable three-dimensional (3D) alginate hydrogel loaded with biodegradable porous poly(ε-caprolactone)–b-poly(ethylene glycol)–b-poly(ε-caprolactone) microspheres (MPs/Alg) as the calcium gluconate container to cross-link alginate. Suspensions of chondrocytes/alginate and porous microspheres turned into a gel because of the release of calcium gluconate; thus, the injectable composite hydrogels give a 3D scaffold to fit the defects perfectly and integrate the extracellular-matrix-mimicking architecture to efficiently accommodate cartilage cells in situ. Tissue repair in a full-thickness cartilage defect model was controlled at 6, 12, and 18 weeks after the implant by micro-CT and immunohistochemistry to evaluate the healing status. The defect in the MPs/Alg+ cells group achieved an almost complete repair at 18 weeks, and the repaired chondrocytes regained a normal tissue structure. Moreover, the MPs/Alg+ cells-treated group increased the quality of tissue formed, including the accumulated glycosaminoglycan and the uniformly deposited type II collagen. The results point out the promising application of the injectable MPs/Alg-chondrocytes system for cartilage tissue engineering.
Article
Full-text available
Statement of significance: An alternative to the use of in vitro expanded cells in regenerative medicine is the use of freshly isolated stromal cells, where a bioactive scaffold or hydrogel is used to provide an environment that enhances their proliferation and tissue-specific differentiation in vivo. The objective of this study was to develop an injectable fibrin hydrogel functionalized with cartilage ECM micro-particles and the growth factor TGF-β3 as a therapeutic for articular cartilage regeneration. This study demonstrates that freshly isolated stromal cells generate cartilage tissue in vivo when incorporated into such a fibrin hydrogels functionalized with cartilage ECM particles. These findings open up new possibilities for intheatre, single-stage, cell-based therapies for joint regeneration.
Article
Periosteum is a thin membrane that encases the surfaces of most bones. It is composed of an outer fibrous layer contains longitudinally oriented cells and collagen fibers and an inner cambial layer that consist of multipotent mesenchymal stem cells (MSCs) and osteogenic progenitor cells. Periosteum has a function of regulating cell and collagen arrangement, which is important to the integrity, modelling and remodelling of bone, particularly during bone defect repair. Apart from autograft and allograft, artificial periosteum, or tissue-engineered periosteum mimicking native periosteum in structure or function, made up of small intestinal submucosa, acellular dermis, induced membrane, cell sheets, and polymeric scaffolds, and so on, has been developed to be used in bone defect repair. In this review, we classify the artificial periosteum into three approaches based on the material source, that is, native tissues, scaffold-free cell sheets and scaffold-cell composites. Mechanisms, methods and efficacy of each approach are provided. Existing obstacles and enabling technologies for future directions are also discussed.
Article
Due to the avascular nature of articular cartilage, damaged tissue has little capacity for spontaneous healing. Three-dimensional scaffolds have potential for use in tissue engineering approach for cartilage repair. In this study, bovine cartilage tissue was decellularized and chemically crosslinked hybrid chitosan/extracellular matrix (ECM) scaffolds were fabricated with different ECM weight ratios by simple freeze drying method. Various properties of chitosan/ECM scaffolds such as microstructure, mechanical strength, swelling ratio, and biodegradability rate were investigated to confirm improved structural and biological characteristics of chitosan scaffolds in the presence of ECM. The results indicated that by introducing ECM to chitosan, pore sizes in scaffolds with 1% and 2% ECM decreased and thus the mechanical properties were improved. The presence of ECM in the same scaffolds also improved the swelling ratio and biodegradation rate in the hybrid scaffolds. MTT cytotoxicity assays performed on chondrocyte cells cultured on chitosan/ECM scaffolds having various amounts of ECM showed that the greatest cell attachment belongs to the sample with intermediate ECM content (2% ECM). Overall, it can be concluded from all obtained results that the prepared scaffold with intermediate concentration of ECM could be a proper candidate for use in cartilage tissue engineering.
Book
Sustainable polymers play an indispensable role in the emergence of green materials, and the 21st century is an era of sustainable polymeric materials. Sustainable polymer-based materials have attracted considerable interest because of the energy crisis and ecological concerns as well as the potential to substitute certain petroleum-derived materials. The use of biorenewable resources is becoming increasingly important and is related to natural bioresources, agricultural production, and new developments in the bio-based materials. This book seeks to address the fundamental understanding of the structure and chemistry of sustainable polymers. It covers the fundamentals of sustainable polymer and presents guidelines in a logical and clear manner for students and researchers to follow. It is a milestone that will help accelerate the progress and advancement in the field of sustainable polymers. The text explores the structure and chemistry of various sustainable polymers, such as cellulose, hemicellulose, lignin, chitosan, starch, guar gum, pectin, and protein, for possible development of green sustainable materials. Each chapter is authored by highly accomplished experts in the field of sustainable polymers and discusses in detail the chemistry and structure of sustainable polymers procured from different natural resources.
Article
Cartilage has limited ability to self-repair due to the absence of blood vessels and nerves. The application of biomaterial scaffolds using biomimetic extracellular matrix (ECM)-related polymers has become an effective approach to production of engineered cartilage. Chitosan/γ-poly(glutamic acid) (γ-PGA) scaffolds with different mass ratios were prepared using genipin as a cross-linker and a freeze-drying method, and their surfaces were modified with elastin, human serum albumin (HSA) and poly-l-lysine (PLL). The scaffolds were formed through a complex between NH3⁺ of chitosan and COO⁻ of γ-PGA, confirmed by Fourier transform infrared spectroscopy, and exhibited an interconnected porous morphology in field emission scanning electron microscopy analysis. The prepared chitosan/γ-PGA scaffolds, at a 3:1 ratio, obtained the required porosity (90%), pore size (≥ 100 μm), mechanical strength (compressive strength > 4 MPa, Young's modulus > 4 MPa) and biodegradation (30–60%) for articular cartilage tissue engineering applications. Surface modification of the scaffolds showed positive indications with improved activity toward cell proliferation (deoxyribonucleic acid), cell adhesion and ECM (glycoaminoglycans and type II collagen) secretion of bovine knee chondrocytes compared with unmodified scaffolds. In caspase-3 detection, elastin had a higher inhibitory effect on chondrocyte apoptosis in vitro, followed by HSA, and then PLL. We concluded that utilizing chitosan/γ-PGA scaffolds with surface active biomolecules, including elastin, HSA and PLL, can effectively promote the growth of chondrocytes, secrete ECM and improve the regenerative ability of cartilaginous tissues.
Article
Articular cartilage repair is a challenge due to its limited self-repair capacity. Cartilage tissue engineering supports to overcome following injuries or degenerative diseases. Herein, we fabricated the scaffold composed of curcumin and silk fibroin as an appropriate clinical replacement for defected cartilage. The scaffolds were designed to have adequate pore size and mechanical strength for cartilage repair. Cell proliferation, sulfated glycosaminoglycan (sGAG) content and mRNA expression analysis indicated that chondrocytes remained viable and showed its growth ability in the curcumin/silk scaffolds. Especially, in 1 mg/ml curcumin/silk scaffold showed higher cell viability rate and extracellular matrix formation than other experimental groups. Furthermore, curcumin/silk scaffold showed its biocompatibility and favorable environment for cartilage repair after transplantation in vivo, as indicated in histological examination results. Overall, the functional composite curcumin/silk scaffold can be applied in cartilage tissue engineering and promising substrate for cartilage repair.
Article
Fibrous scaffolds that reconstruct the extracellular matrix (ECM) have been utilized for tissue regeneration demonstrated potential for guiding stem cell differentiation. Nanofibrous scaffolds fabricated by a unique electrospinning method enabled us to create tailored, functional scaffolds on-demand. Several patterned electrospun poly(ε-caprolactone) (PCL) scaffolds were prepared, and then utilized for creating a hybrid composite in which bone-like hydroxyapatite (b-HA) was deposited onto the unique electrospun scaffolds. The mineral deposits onto the patterned PCL scaffolds was confirmed by scanning electron microscope (SEM). When culturing human adipose-derived stem cells (hASC) onto the different SBF-treated electrospun PCL scaffolds, it was found that the hybrid composite can support hASC differentiated into osteoblasts under osteogenic differentiation conditions. Image analysis and alamar blue assay indicated a significant increase of hASC adhesion and proliferation on the SBF-treated PCL scaffolds. Subsequent analysis of osteogenic potential by via gene expression analysis and alkaline phosphatase (ALP) activity also demonstrated that the SBF-treated electrospun PCL made by the modified electrospinning process is more favorable for the osteogenic differentiation hASCs. Additionally, results of alizarin red S staining and ALP staining at days 7 and 14 showed improved deposition of mineralized matrix on the SBF-treated PCL. Therefore, this study indicates that the facile scaffold fabrication method described in this study is promising approach to prepare osteoconductive scaffold for bone tissue engineering.
Chapter
A wide variety of polymers have been used over decades for the preparation of dressing materials for wound healing applications. But the dressing materials based on polysaccharides such as chitosan (CS) have received tremendous attention of the worldwide researchers as a consequence of its important properties like anti-infectional activity, biocompatibility, biodegradability, nontoxicity to mention a few. CS helps in every phase of wound healing such as acting as barrier against microbes, absorbing exudates, accelerates the infiltration of inflammatory cells like neutrophils and helps in healing without scar formation. A reason behind the popularity of CS is that not only it can easily be processed as gels, films, fibers, and scaffolds but also can be blended with natural as well as synthetic polymers to reduce price and improve properties like mechanical, wettability, gas permeability, and handling. Apart from natural and synthetic polymers, CS is also blended with nanoparticles and growth factors to which it shows better antibacterial activity and reduce time span for wound healing. The present chapter aims to focus on feasibility of combining natural polymers, synthetic polymers, nanoparticles, and growth factors with CS for the preparation of wound dressings as basic healthcare materials for regenerative medicine.