Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

International Journal of Biological Macromolecules 158 (2020) 452–460

Contents lists available at ScienceDirect

International Journal of Biological Macromolecules

journal homepage: http://www.elsevier.com/locate/ijbiomac

Advanced gellan gum-based glycol chitosan hydrogel for cartilage tissue


engineering biomaterial
Sumi Lee 1, Joo Hee Choi 1, Ain Park, Mina Rim, Jina Youn, Wonchan Lee, Jeong Eun Song, Gilson Khang ⁎
Department of BIN Convergence Technology, Department of Polymer Nano Science & Technology Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-
do, Republic of Korea

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

Article history: Gellan gum (GG), a nature-derived polysaccharide, is one of the materials widely used in cartilage tissue engi-
Received 17 January 2020 neering (TE). Glycol chitosan (GC), a derivative of chitosan, is a water-soluble natural polymer that has excellent
Received in revised form 24 February 2020 biocompatibility and biodegradability as well as cell adhesion. Herein, GG was physically blended with GC to en-
Accepted 18 April 2020
hance the mechanical properties and microenvironment of the GG to apply in cartilage TE. The study was con-
Available online 23 April 2020
ducted with a hydrogel model which is similar to the extracellular matrix (ECM) of cartilage tissue. The
Keywords:
physicochemical studies were carried out with morphological study, swelling ratio, weight loss, and sol fraction.
Hydrogel The mechanical characterization was conducted with compression test and rheological study to confirm avail-
Gellan gum ability in cartilage TE material. Furthermore, in vitro studies such as morphology investigation, viability assay,
Glycol chitosan GAG content, qRT-PCR, and histological study were performed to verify biocompatibility and chondrogenesis
Cartilage of the material. The mechanical and biological properties improved with a proper amount of GC. Overall results
Chondrocyte verify the potential of the material and can be further used for the cartilage TE.
Regeneration © 2020 Published by Elsevier B.V.

1. Introduction

Gellan gum hydrogel (GG) exhibited cell suitability for various cell of methods have recently been proposed to enhance the characters of
types and allowed cell encapsulation in many studies. The GG forms a hydrogels by using a simple method.
random coil at high temperatures, but at low temperatures it forms a In this study, composite matrix of GG and glycol chitosan (GC) was
double helix which leads to physical crosslinking [1,2]. Furthermore, ad- manufactured to evaluate the effect of the composite in chondrogenesis.
dition of cations such as Ca2+, Mg2+, Na+, and K+ or controlling the Chitosan is widely investigated as a suitable composite for bio-
concentration of GG allows tunable mechanical performance [3]. How- applications [10,11]. However, its solubility in water is poor and re-
ever, despite its merits in tissue engineering application, GG fails to quires aqueous solution containing acid which lead to the protonation
meet the conditions of cell delivery system due to the disadvantages of amino groups (R-NH+ 3 ) [12]. This may induce toxicity in vitro and
such low mechanical property and lack of cell binding sites for cells in vivo [13]. Therefore, chitosan was modified into new properties by
growth and migration. These drawbacks may lead to a major cell modifying the backbone. Among many chitosan derivatives, GC is
death, low proliferation, hinder growth and differentiation [4]. water-soluble, biocompatible, biodegradable, and easily available on
Many studies have been conducted to solve these problems. The GG the market [14]. Furthermore, the presence of –NH2 can increase the
backbone was modified chemically with biological factor or GG itself to cell adhesion which may enhance the microenvironment of the matrix
decrease the gelation temperature, enhance the microenvironment, and [15,16]. The composite hydrogel of GG and GC (G/C) was characterized
include adhesive sites [5,6]. Another approach to improve mechanical, chemically and mechanically and evaluated in vitro to confirm its appli-
physiological, and biological properties of the GG is synthesizing with cability in cartilage tissue engineering.
different types of polymers [7,8]. Many strategies using chemical modi-
fications have been proposed, but they have the disadvantage that they
can be time consuming and highly toxic in vivo [9]. Therefore, a number 2. Materials and methods

⁎ Corresponding author. 2.1. Preparation of G/C hydrogels


E-mail addresses: sumilee@jbnu.ac.kr (S. Lee), zooheechoi@jbnu.ac.kr (J.H. Choi),
ainpark@jbnu.ac.kr (A. Park), miiing95@jbnu.ac.kr (M. Rim), yjina@jbnu.ac.kr (J. Youn),
lwc95@jbnu.ac.kr (W. Lee), songje@jbnu.ac.kr (J.E. Song), gskhang@jbnu.ac.kr (G. Khang). Low-acyl gellan gum (GG, Gelzan™, Sigma-Aldrich, St. Louis, MO,
1
First co-authors. USA) was used in this experiment. First, 1% (w/v) GG was dissolved in

https://doi.org/10.1016/j.ijbiomac.2020.04.135
0141-8130/© 2020 Published by Elsevier B.V.
S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460 453

90 °C deionized water (DW) to fabricate homogeneous GG hydrogel so- 2.2.6. SEM analysis
lution and CaCl2 was added to the solution. The temperature was The porous structure of the hydrogels was studied by Bio-LV scan-
lowered to 60 °C and the glycol chitosan (GC, Sigma-Aldrich, USA) ning electron microscope (Bio-LV SEM, Hitachi, SN-3000 Hitachi,
was added to the solution at the amount of 0%, 0.05%, 0.075%, 0.1%, Japan). The hydrogels were frozen in a deep-freezer (−60 °C). The spec-
and 0.2% (w/v); GG, G/C0.05, G/C0.075, G/C0.1, and G/C0.2, respectively. imens were cross-sectioned and gold sputtered using a vacuum sputter
The solutions were stirred until the GC was completely dissolved. The (SC500k, Emscope, UK) for the evaluation.
homogeneous solutions were poured into petri dish and left at room
temperature for gelation. The fabricated hydrogels were punched with 2.3. In vitro study
a biopsy punch (Kai Medical, Honolulu, HI, USA) to make 3 mm height
and 6 mm diameter cylindrical disk and were used for further studies. 2.3.1. Animal experiment
All the animal experiments were performed following the guidelines
2.2. Characterization and approval of Chonbuk National University Animal Care Committee,
Jeonju, Republic of Korea (CBNU 2016-50).
2.2.1. FT-IR analysis
The chemical structures of GG, GC, and G/C hydrogel were character- 2.3.2. Isolation and culture of chondrocytes from rabbit model
ized by a Fourier transform infrared spectroscopy (FTIR, Perkin Elmer, All materials were autoclaved before the experiment. Chondrocytes
USA) at the wavelength range of 400–4000 cm−1. The samples were were isolated from a 4 weeks old female New Zealand white rabbits
measured in a dried state. (Hanil laboratory animal center, Wanju, Korea) following the lab
protocol [20]. Briefly, flesh was removed from the extracted legs and
2.2.2. Swelling analysis washed with a sterilized PBS to eliminate residual blood and flesh. A car-
The specimens were prepared with the method of Section 2.1. Initial tilage tissue was peeled off using a blade and digested it in a 0.02% col-
weight of the hydrogel was measured (Mf) and then placed in a 24-well lagenase A (Roche, Germany) for overnight. Separated cells were
plate. A phosphate-buffered saline (PBS, pH 7.4, Gibco, Big Cabin, OK, centrifuged in a 1200 rpm at 4 °C for 3 min. The centrifuged
USA) was added and stored in a 37 °C incubator until at a specific time chondrocytes were resuspended in a Dulbecco's Modified Eagle Me-
point. At a certain point, the PBS was removed and the residual solution dium Nutrient Mixture F-12 (DMEM /F12, Gibco, Big Cabin, OK, USA)
was removed using a filter paper. The weight of the samples was re- supplemented with 10% of Fetal Bovine Serum (FBS, Gibco, USA) and
corded (mw) and the samples were rinsed with a DW 3 times. The 1% of Penicillin/Streptomycin (PS, Gibco, USA) and cultured in cell cul-
hydrogels were dried in a 60 °C oven for 3 h and the weight was mea- ture dishes (Eppendorf, Hamburg, Germany) under standard conditions
sured (mf). The swelling ratio and the weight loss were calculated (5% CO2 and 37 °C). The media was changed every 3 days and primary
using the Eqs. (1) and (2) respectively [17,18]. The study was evaluated passage 2 of chondrocytes were used for this study.
on 1, 7, 14, 21, and 28 days. PBS was replaced every 3 days.
2.3.3. Cell culture in the hydrogels
Swelling ratio ð%Þ ¼ ðmw −m f Þ=m f  100 ð1Þ All materials were sterilized with autoclave and under ultraviolet
(UV) lamp before the experiment. The hydrogels were fabricated fol-
Weight loss ð%Þ ¼ ðmi −m f Þ=mi  100 ð2Þ lowing the 2.1. Preparation of G/C hydrogels method. The manufactured
solutions were filtered through a 0.45 μm filter (Sigma-Aldrich,
Germany). The trypsinized chondrocytes (2 × 106 cells/mL) were care-
fully mixed with the hydrogels at the temperature before the gelation
2.2.3. Sol fraction
occur (35–37 °C). The co-cultured solutions were cast into a petri dish
The weight of freeze-dried hydrogels was measured (mi). The mea-
and left at RT for gelation. The stable hydrogels were punched into
sured samples were immersed in a DW (good solvent) and agitated on a
3 mm height and 6 mm diameter size with a biopsy punch. Each hydro-
shaker for 1 h. The DW was removed and the hydrogels were freeze-
gel was transferred to a 24-well plate and cultured in a 1 mL of DMEM/
dried again. The weight of the samples was recorded (mf). The sol frac-
F12 cell culture media. The media was changed every 3 days.
tion was calculated by following Eq. (3) [19].
2.3.4. Cell viability assay
Sol fraction ð%Þ ¼ ðmi −m f Þ=mi  100% ð3Þ Cell viability study was conducted with MTT (3-[4,-dimethylthiazol-
2-yl]-2,5-diphenylterazolium bromide; thiazolyl blue, Amresco, TX,
USA) assay. The evaluation was carried out on 3, 7, 14, and 21 days of
2.2.4. Compressive strength culture. The existing medium was replaced with a fresh medium and
The compression test was carried out under an unconfined condi- added MTT solution (5 mg/mL in PBS) to make a 10% reagent. The
tion. The prepared cylindrical shape hydrogels were placed on a cylin- treated samples were stored in a humidified 5% CO2 and 37 °C environ-
der plate of a Texture Analyzer (FTC, Sterting, Virginia, USA). The ment for 3 h. The supernatant was removed and the samples were ho-
hydrogels were compressed at the speed of 2 mm/min and load cell of mogenized with dimethyl sulfoxide (DMSO, Samchun chemical, South
10 N. Before characterization, the height, and diameter of the samples Korea) using a glass tissue grinder (WHEATON, USA) dissolve formazan
were measured using a caliper (Mitutoyo, South Korea) to calculate crystal. The solutions were transferred into a 96-well plate and mea-
the accurate compressive strength. sured under an absorbance of 570 nm with a microplate reader (Syn-
ergy MX, Biotek, Vernusky, VT, USA).
2.2.5. Rheological evaluation
The rheological character and gelation temperature of the hydrogels 2.3.5. Live/dead assay
were characterized with a viscometer (AMETEK Brookfield, Middleboro, A live/dead cell imaging kit (Invitrogen, Carlsbad, CA, USA) was used
MA, USA). The water circulation bath (VCB-07, JONGRO Industrial Co., in this study according to the kit protocol. Briefly, the hydrogels were
Ltd., South Korea) was preheated and the 5 mL of a premade hydrogel washed with PBS and sliced into half. The samples were transferred to
solution was transferred to the viscometer. The temperature was grad- a coverglass bottom dish (SPL, Lifescience, Korea). A mixed reagent
ually lowered until the circulation bath reached 18 °C. The router speed which contains calcein AM (green) and ethidium homodimer (red)
was set at 0.5 rpm and cone and plate spindle (SC-32 spindle, AMETEK were added on top of the specimens. The treated samples were incu-
Brookfield, USA) were used for this study. bated under standard conditions (5% CO2 and 37 °C). The image was
454 S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460

2.3.7. Histological analysis


The specimens were cultured for 21 days and washed. At specific
time point, the samples were washed with PBS 3 times and fixed with
a 4% formaldehyde (Sigma-Aldrich, USA). The specimens were frozen
with a CryoMatrix (Thermo Fisher Scientific, USA) in a liquid nitrogen.
The prepared samples were sectioned in a 10 μm thickness using a
cryomicrotome (Thermo Fisher Scientific, USA). Hematoxylin & Eosin
(H&E) and Safranin-O staining were carried out according to the stan-
dard histological methods. All the reagents were bought from Sigma-
Aldrich except for hematoxylin which was obtained from Fisher Scien-
tific, Hampton, USA.

2.3.8. GAG quantitative analysis


Glycosaminoglycan (GAG) analysis was performed at specific time
points. The samples were rinsed with PBS and were lyophilized. The
weight of the samples was recorded and papain solution was added
and treated for 16 h in a 60 °C oven to perform enzymatic digestion.
The papain digestion solution was prepared by dissolving 125 μg/mL
Fig. 1. FT-IR analysis of GG, GC and G/C hydrogel in the wavelength range of of papain (Sigma-Aldrich, USA) and 10 mM L-cystein (Sigma-Aldrich,
400–4000 cm−1. USA) into a premade PBE buffer (pH 6.5) which constituted of
100 mM Na2HPO4 (SHOWA, Japan) and 10 mM EDTA (Sigma-Aldrich,
USA). GAG content was characterized with a 1,9-dimethylmethylene
taken under a super resolution confocal laser scanning microscope (LSM blue (DMMB, Sigma-Aldrich, USA) dye-binding assay with a chondroi-
880 with Airyscan, Carl Zeiss, Germany) using a Z-stack method. The tin sulfate from bovine trachea (Sigma-Aldrich, USA) as a standard
fluorescence intensity was measured by using an image J program [21]. [22]. The biosynthetic activity of the cells was assessed by normalizing
the GAG quantification to the double-stranded DNA (dsDNA) content
which was studied by a Qunati-iT PicoGreen reagent (Life Technologies,
2.3.6. Morphology observation Carlsbad, CA, USA) according to the product protocol. Standard curves
The cell encapsulated hydrogels were cultured for 21 days. The spec- were prepared at concentrations ranging from 0 to 2 μg/mL. The mea-
imens were washed with PBS 3 times and fixed with 2.5% glutaralde- surement was conducted with a microplate reader at a wavelength for
hyde (Sigma-Aldrich, USA) for 24 h in 4 °C. The fixed samples were GAG content (525 nm) and the fluorescence intensities at an excitation
stored in −60 °C deep-freezer for 24 h and were lyophilized for 48 h wavelength of 485/20 nm and an emission wavelength of 528/20 nm for
to remove water. The SEM image of the prepared samples was observed the dsDNA quantification.
by following Section 2.2.6.

Fig. 2. (A) Swelling ratio (%) (B) weight loss (%) and (C) sol fraction (%) of the hydrogels (values are mean ± SD, n = 3, P b 0.05 (*), P b 0.01 (**), P b 0.001 (***)).
S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460 455

Fig. 3. Morphological analysis under SEM.

2.3.9. Quantitative real-time polymerase chain reaction (qRT-PCR) 2.4. Statistics


A quantitative real-time polymerase chain reaction (qRT-PCR) was
carried out to investigate gene expression of chondrocytes cultured in All the numerical results are presented by the mean ± standard de-
the hydrogels. On 21 days of culture, hydrogels were washed 3 times viation (SD). The GraphPad Prism 5.0 software (GraphPad Software, La
with PBS and cells were frozen in −60 °C deep freezer for overnight. Jolla, CA, USA) was utilized to perform the statistical analysis. The stud-
Cell lysis was conducted by homogenizing the cell encapsulated ies were analyzed employing one-way analysis of variance (one-way
hydrogels with a Trizol (Invitrogen, USA) in the glass tissue grinder. ANOVA test), and the differences were considered significant at
The solution was transferred to a 1.5 mL Eppendorf tube (EP tube). A P b 0.05(*), P b 0.01(**), and P b 0.001(***).
chloroform (SAMCHUN chemicals, South Korea) was included and the
solution was shaken well to mix and centrifuged at 12,000 rpm in 4 °C 3. Result and discussion
for 15 min. The supernatant was carefully transferred to another EP
tube and mixed with isopropanol (Sigma-Aldrich, USA) to sink ex- 3.1. Characterization
tracted mRNA. The samples were stored in 4 °C overnight and centri-
fuged at 12,000 rpm in 4 °C for 5 min. The supernatant was removed 3.1.1. FT-IR analysis
and washed with a 75% ethanol. The samples were centrifuged at The chemical composition of GG, GC, and interaction among GG and
7500 rpm in 4 °C for 10 min and the supernatant was removed. Ultra- were evaluated with FT-IR analysis (Fig. 1). The specific peak of GG was
Pure DNase/RNase-free distilled water (Invitrogen, USA) was added to observed at 3300 cm−1 a stretching band of –OH groups, at 2914 cm−1
the extracted samples. The concentration of mRNA was determined which corresponds to stretching vibrations of the –CH groups, at
using a Biospectrophotometer (Eppendorf, USA). cDNA was synthesized 1618 cm−1 due to the presence of C_O stretching vibration from car-
by using TOPscriptTM RT DryMIX (dT18 plus) (Enzynomics, South bonyl groups, and at 1030 cm−1 which is caused by –COC stretching.
Korea). qRT-PCR was performed by StepOnePlus Real-Time PCR system The FTIR spectrum of GC showed characteristic peaks associated with
(Applied Biosystems, USA) using a SYBR Green Master Mix (Applied C_O stretching amide I, –NH bending of primary amines, and amide
Biosystems, USA). Aggrecan (AGG), Collagen type 2 (COL2), and SOX9 II. The blended network exhibited shifted frequency band at
were applied for the study and GAPDH were used as a housekeeping 3375 cm−1, 2943 cm−1, 1619 cm−1, and 1041 cm−1 [23]. This phenom-
gene. enon appeared due to the electrostatic interaction of carboxylate groups

Fig. 4. (A) Compressive strength and (B) rheological characteristics (values are mean ± SD, n = 3, P b 0.05 (*), P b 0.01 (**), P b 0.001 (***)).
456 S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460

hydrophilicity of glycol group in the GC (Fig. 2A). The weight loss of


the hydrogels also showed the similar aspect as the swelling kinetic
(Fig. 2B). This may be due to difference in the osmotic pressure with
the physiological aqueous solution. The difference in an osmotic pres-
sure leads hydrogels to absorb water and swell and reach equilibrium
state. However, as the matrix degrade, the elastic pressure decreases
and cause more swollen state [25]. The initial state of all the specimens
showed fast degradation which may be due to the release of free GG and
GC chains [20]. The swelling and weight loss rate did not show signifi-
cant differences between each specimen until 14 days of measurement.
However, after 14 days, GG and G/C0.05 showed a progressive decrease
in the rate of degradation, while the rest of the groups continued to in-
crease. This may be due to the lower polymers that are involved in the
matrix by the inhibition of crosslinking from GC incorporation as
shown in the sol fraction result (Fig. 2C) [26]. The morphological analy-
sis of all the hydrogels showed continuous porous structures which re-
sult from ice crystal formation from freeze-drying step [27]. The pore
size increased as the GC content and displayed lamellar porous struc-
Fig. 5. MTT assay exhibited in optical density values (values are mean ± SD, n = 3).
tures (Fig. 3). It is assumed that the entanglement between the two net-
works allowed interconnected porous structures.
and amide groups in GG and GC lead to shifting in the specific peak of
GG peak in the G/C hydrogel [24]. Besides the characteristic peaks corre- 3.1.3. Mechanical study
sponding to GG and GC, a new absorption peak did not appear, implying The compressive strength and viscosity of the fabricated materials
that the network is formed with the physical interaction. were characterized to investigate influence of GC. The compressive
strength of the hydrogel is an essential property for cartilage tissue engi-
3.1.2. Physicochemical characterization of GC/GG neering materials in order to sustain the external force [28]. The compres-
The swelling and degradation kinetics were carried out for 28 days. sive modulus increased as the GC was included in the GG (Fig. 4A). The
The swelling rate of the hydrogel is important because it is related to viscosity of the hydrogels displayed higher level when GC was added in
nutrient uptake and in vivo fluid retention in cell culture media the matrix (Fig. 4B). This may be due to the higher interaction between
in vitro [19]. The swelling ratio of the hydrogels showed gradual in- two matrices [29]. The gelation temperature was measured according to
crease as the time passed which may be due to the increased the temperature change. It is important for gelation to occur at proper

Fig. 6. (A) LSM image of chondrocytes encapsulated in the hydrogels analyzed with a Z-stack mode and (B) relative intensity of LSM images (scale bar = 50 μm) (values are mean ± SD,
n = 3, P b 0.05 (*), P b 0.01 (**), P b 0.001 (***)).
S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460 457

Fig. 7. SEM images of chondrocytes encapsulated in the hydrogels.

temperature which is not too high or low which may affect the survival of 3.2.3. Biochemical study
the cells [30]. Also, it is important for hydrogel to solidify after cells are GAG is a significant component of cartilaginous ECM [39,40]. The
uniformly suspended in the hydrogels [31,32]. Each hydrogel showed ge- results of the biosynthesis of chondrocytes encapsulated in
lation temperature at 32.1 °C, 32.2 °C, 33.6 °C, 33.7 °C, and 34.2 °C in GG, hydrogels showed increasing amount over time (Fig. 9). The GAG
G/C0.05, G/C0.075, G/C0.1, and G/C0.2, respectively. The gelation temper- synthesis showed significantly higher level in the G/C0.075 con-
ature was not significantly affected of inclusion of GC which verifies the structs when compared to the GG but decreased when higher
applicability in cell culturing material [33]. amount of GC was incorporated in the GG matrix. Especially on 14
and 21 days, G/C0.075 showed about 2 times higher content com-
pared to the GG. These results show the positive effect of the appro-
3.2. In vitro study
priate amount of GC blended GG from the complementary effect of
the two substances which provided enhanced microenvironment to
3.2.1. Biocompatibility
form GAGs in hydrated macrostructures. Furthermore, the expres-
The biocompatibility is an important factor to consider in choosing
sion of cartilage-related gene markers such as AGG, COL2, and
tissue engineering materials. The MTT assay which confirms the viabil-
SOX9 was confirmed and standardized based on GAPDH, a house
ity of cells by metabolic activity showed that all the groups showed sim-
keeping gene. SOX9 is one of the most important transcription fac-
ilar optical density (Fig. 5). However, live/dead staining of the cells
tors that is crucial for chondrocyte differentiation and cartilage for-
encapsulated hydrogels showed higher amounts of dead cells in the
mation. COL2 is the main constituent of cartilage tissue and AGG
GG group while the viability of the cells increased in the GC incorpo-
plays an important role in a cartilage function [41–43]. All the
rated hydrogels (Fig. 6). This may be due to the higher attachment site
genes showed higher level in the GC incorporated hydrogels when
from GC and improved mechanical property enhanced the viability of
compared to the pristine GG (Fig. 10). Especially, G/C0.075 matrix
the cells [16,34].
exhibited a significantly highest amount. As it is specified in the pre-
vious experiments, when appropriate amount of GC was added, the
3.2.2. Morphological and histological observation cell proliferation and growth were active which allowed proper
Chondrocytes encapsulated hydrogels were incubated for 21 days chondrogenesis by providing the enhanced microenvironment.
and examined under SEM to observe ECM formation, cell morphology
and growth. It is noted that the spherical shape is favorable for chondro-
genesis [35]. All the encapsulated cells displayed roundel shape. How- 4. Conclusion
ever, while GG showed cells tangled in the matrix rather than being
presented in the pores, higher amounts of cells existing in the porous In this study, GG and GC composite was proposed as an effective ma-
structure was detected in hydrogels containing GC (Fig. 7). H&E and terial for cartilage TE. The GG was manufactured with various amount of
Safranin-O staining were performed on day 21 to evaluate morphology GC with a simple method. GG acted as a platform for the entire support,
and expression of cartilaginous matrix. The H&E which stains nucleus while GC improved the mechanical properties of the hydrogel and en-
and possible secreted cytoplasm around cells were examined. The hanced the microenvironment. The physicochemical and mechanical
cells were well distributed in the matrix and some of the cells showed properties of the hydrogels were improved as the GC was incorporated.
noticeable cytoplasm around the cells (Fig. 8A). The G/C0.075 showed In vitro results showed higher cell viability and proliferation. The bio-
cytoplasm stretching out of the cells (shown in arrows) which indicate chemical studies exhibited higher level of GAG synthesis and mRNA ex-
that the inclusion of GC enhances the microenvironment of the hydro- pression of cartilage-specific genes in G/C0.075. The hydrogel was
gel and has a beneficial effect on the metabolism and growth of the fabricated with a non-toxic crosslinker in a pH-neutral condition. Also,
cells [36,37]. The Safranin-O which stains GAG exhibited higher positive material can be improved with the use of stem cells, growth factors,
red staining around the cells in GC loaded hydrogel (shown in arrows) genes, therapeutic drugs, and biomolecules [44]. Overall, with the ap-
(Fig. 8B). These results show that the GC has positive effect on improv- propriate amount of GC in the GG-based hydrogel, G/C hydrogel is ex-
ing microenvironment [38]. pected to be applicable in cartilage TE.
458 S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460

Fig. 8. Histological images of (A) H&E and (B) Safranin-O.

Funding

This research was supported by the Korea Health Technology R&D


Project through the Korea Health Industry Development Institute
(KHIDI), funded by the Ministry of Health & Welfare, Republic of
Korea (HI15C2996) and the International Research & Development
Program of the National Research Foundation of Korea (NRF) funded
by the Ministry of Science, ICT & Future Planning (NRF-
2017K1A3A7A03089427).

Author contributions

We believe that these individuals should be listed as authors be-


cause: S. Lee and J.H. Choi designed and conceived the study. S. Lee, J.
Yoon, and W. Lee synthesized and characterized the materials. S. Lee,
J.H. Choi and A. Park performed in vitro study. S. Lee, A. Park, and M.
Fig. 9. GAG quantitative analysis of chondrocytes encapsulated in the hydrogels (values Rim performed the Histology. S. Lee and J.H. Choi wrote the manuscript.
are mean ± SD, n = 3). J.E. and G. contributed reagents and materials for this study.
S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460 459

Declaration of competing interest encapsulated rabbit marrow mesenchymal stem cells in vitro, Biomacromolecules
(2009)https://doi.org/10.1021/bm801197m.
[20] W.K. Kim, J.H. Choi, M.E. Shin, J.W. Kim, P.Y. Kim, N. Kim, J.E. Song, G. Khang, Evalu-
We wish to confirm that there are no know conflicts of interest asso- ation of cartilage regeneration of chondrocyte encapsulated gellan gum-based
ciated with this publication and there has been no significant financial hyaluronic acid blended hydrogel, Int. J. Biol. Macromol. (2019)https://doi.org/10.
1016/j.ijbiomac.2019.08.176.
support for this work that could have influenced its outcome. [21] M.D. Abràmofff, P.J. Magalhães, S.J. Ram, Image processing with image J part II,
Biophoton. Int. 11 (2005) 36–43, https://doi.org/10.1117/1.3589100.
Acknowledgements [22] M. Adamczyk, J.C. Gebler, J. Wu, Papain digestion of different mouse IgG subclasses
as studied by electrospray mass spectrometry, J. Immunol. Methods (2000)https://
doi.org/10.1016/S0022-1759(00)00135-6.
This research was supported by the Korea Health Technology R&D [23] V.C. Dumont, H.S. Mansur, A.A.P. Mansur, S.M. Carvalho, N.S.V. Capanema, B.R.
Project through the Korea Health Industry Development Institute Barrioni, Glycol chitosan/nanohydroxyapatite biocomposites for potential bone tis-
sue engineering and regenerative medicine, Int. J. Biol. Macromol. 93 (2016)
(KHIDI), funded by the Ministry of Health &Welfare, Republic of Korea
1465–1478, https://doi.org/10.1016/j.ijbiomac.2016.04.030.
(HI15C2996) and the International Research & Development Program [24] J.W. Jang, J. Kim, M.S. Kook, K.Y. Lee, Evaluation of gellan gum/glycol chitosan
of the National Research Foundation of Korea (NRF) funded by the Min- bioabsorbable membrane for guided bone regeneration, Polym (2018)https://doi.
istry of Science, ICT & Future Planning (NRF-2017K1A3A7A03089427). org/10.7317/pk.2018.42.5.874.
[25] K. Hayashi, F. Okamoto, S. Hoshi, T. Katashima, D.C. Zujur, X. Li, M. Shibayama, E.P.
Gilbert, U. Il Chung, S. Ohba, T. Oshika, T. Sakai, Fast-forming hydrogel with ultralow
References polymeric content as an artificial vitreous body, Nat. Biomed. Eng. (2017)https://
doi.org/10.1038/s41551-017-0044.
[1] D.F. Coutinho, S.V. Sant, H. Shin, J.T. Oliveira, M.E. Gomes, N.M. Neves, A. [26] S. Khunmanee, Y. Jeong, H. Park, Crosslinking method of hyaluronic-based hydrogel
Khademhosseini, R.L. Reis, Modified Gellan Gum hydrogels with tunable physical for biomedical applications, J. Tissue Eng. (2017)https://doi.org/10.1177/
and mechanical properties, Biomaterials (2010)https://doi.org/10.1016/j. 2041731417726464.
biomaterials.2010.06.035. [27] S. Sornkamnerd, M.K. Okajima, T. Kaneko, Tough and porous hydrogels prepared by
[2] A.H. Bacelar, J. Silva-Correia, J.M. Oliveira, R.L. Reis, Recent progress in gellan gum simple Lyophilization of LC gels, ACS Omega (2017)https://doi.org/10.1021/
hydrogels provided by functionalization strategies, J. Mater. Chem. B (2016) acsomega.7b00602.
https://doi.org/10.1039/c6tb01488g. [28] A. Vedadghavami, F. Minooei, M.H. Mohammadi, S. Khetani, A. Rezaei Kolahchi, S.
[3] F. Wang, Y. Wen, T. Bai, The composite hydrogels of polyvinyl alcohol–gellan gum- Mashayekhan, A. Sanati-Nezhad, Manufacturing of hydrogel biomaterials with con-
Ca2+ with improved network structure and mechanical property, Mater. Sci. Eng. C trolled mechanical properties for tissue engineering applications, Acta Biomater.
(2016)https://doi.org/10.1016/j.msec.2016.06.084. (2017)https://doi.org/10.1016/j.actbio.2017.07.028.
[4] H. Shin, B.D. Olsen, A. Khademhosseini, Gellan gum microgel-reinforced cell-laden [29] J. Jancar, J.F. Douglas, F.W. Starr, S.K. Kumar, P. Cassagnau, A.J. Lesser, S.S. Sternstein,
gelatin hydrogels, J. Mater. Chem. B (2014)https://doi.org/10.1039/c3tb20984a. M.J. Buehler, Current issues in research on structure-property relationships in poly-
[5] R. López-Cebral, A. Civantos, V. Ramos, B. Seijo, J.L. López-Lacomba, J.V. Sanz-Casado, mer nanocomposites, Polymer (Guildf) (2010)https://doi.org/10.1016/j.polymer.
A. Sanchez, Gellan gum based physical hydrogels incorporating highly valuable 2010.04.074.
endogen molecules and associating BMP-2 as bone formation platforms, Carbohydr. [30] L. Klouda, A.G. Mikos, Thermoresponsive hydrogels in biomedical applications, Eur.
Polym. (2017)https://doi.org/10.1016/j.carbpol.2017.03.049. J. Pharm. Biopharm. (2008)https://doi.org/10.1016/j.ejpb.2007.02.025.
[6] L.P. Da Silva, M.T. Cerqueira, R.A. Sousa, R.L. Reis, V.M. Correlo, A.P. Marques, Engi- [31] I.M. El-Sherbiny, M.H. Yacoub, Hydrogel scaffolds for tissue engineering: Progress and
neering cell-adhesive gellan gum spongy-like hydrogels for regenerative medicine challenges, Glob. Cardiol. Sci. Pract. (2013)https://doi.org/10.5339/gcsp.2013.38.
purposes, Acta Biomater. (2014)https://doi.org/10.1016/j.actbio.2014.07.009. [32] R. Dimatteo, N.J. Darling, T. Segura, In situ forming injectable hydrogels for drug de-
[7] K.M. Zia, S. Tabasum, M.F. Khan, N. Akram, N. Akhter, A. Noreen, M. Zuber, Recent livery and wound repair, Adv. Drug Deliv. Rev. (2018)https://doi.org/10.1016/j.addr.
trends on gellan gum blends with natural and synthetic polymers: a review, Int. J. 2018.03.007.
Biol. Macromol. (2018)https://doi.org/10.1016/j.ijbiomac.2017.11.099. [33] U.J. Kim, J. Park, C. Li, H.J. Jin, R. Valluzzi, D.L. Kaplan, Structure and properties of silk
[8] T.E.L. Douglas, W. Piwowarczyk, E. Pamula, J. Liskova, D. Schaubroeck, S.C.G. hydrogels, Biomacromolecules (2004)https://doi.org/10.1021/bm0345460.
Leeuwenburgh, G. Brackman, L. Balcaen, R. Detsch, H. Declercq, K. Cholewa- [34] M. Khoshgoftar, K. Ito, C.C. Van Donkelaar, The influence of cell-matrix attachment
Kowalska, A. Dokupil, V.M.J.I. Cuijpers, F. Vanhaecke, R. Cornelissen, T. Coenye, A.R. and matrix development on the micromechanical environment of the chondrocyte
Boccaccini, P. Dubruel, Injectable self-gelling composites for bone tissue engineering in tissue-engineered cartilage, Tissue Eng. - Part A. (2014)https://doi.org/10.1089/
based on gellan gum hydrogel enriched with different bioglasses, Biomed. Mater. ten.tea.2013.0676.
(2014)https://doi.org/10.1088/1748-6041/9/4/045014. [35] C.G. Jeong, S.J. Hollister, Mechanical and biochemical assessments of three-
[9] A.M.M. Edeerozey, H.M. Akil, A.B. Azhar, M.I.Z. Ariffin, Chemical modification of dimensional poly (1,8-octanediol-co-citrate) scaffold pore shape and permeability
kenaf fibers, Mater. Lett. (2007)https://doi.org/10.1016/j.matlet.2006.08.006. effects on in vitro chondrogenesis using primary chondrocytes, Tissue Eng. - Part
[10] J. Venkatesan, S.K. Kim, Chitosan composites for bone tissue engineering — an over- A. (2010)https://doi.org/10.1089/ten.tea.2010.0103.
view, Mar. Drugs (2010)https://doi.org/10.3390/md8082252. [36] Y.M. Kook, Y. Jeong, K. Lee, W.G. Koh, Design of biomimetic cellular scaffolds for co-
[11] S.J. Yoon, Y. Yoo, S.E. Nam, H. Hyun, D.W. Lee, S. Um, S.Y. Kim, S.O. Hong, D.H. Yang, culture system and their application, J. Tissue Eng. (2017)https://doi.org/10.1177/
H.J. Chun, The cocktail effect of BMP-2 and TGF-β1 loaded in visible light-cured gly- 2041731417724640.
col chitosan hydrogels for the enhancement of bone formation in a rat tibial defect [37] H.S. Yu, J.J. Kim, H.W. Kim, M.P. Lewis, I. Wall, Impact of mechanical stretch on the
model, Mar. Drugs. (2018)https://doi.org/10.3390/md16100351. cell behaviors of bone and surrounding tissues, J. Tissue Eng. (2016)https://doi.
[12] K. Sun, Z.H. Li, Preparations, properties and applications of chitosan based nanofi- org/10.1177/2041731415618342.
bers fabricated by electrospinning, Express Polym Lett (2011)https://doi.org/10. [38] Z. Liu, H. Wang, Y. Wang, Q. Lin, A. Yao, F. Cao, D. Li, J. Zhou, C. Duan, Z. Du, Y. Wang,
3144/expresspolymlett.2011.34. C. Wang, The influence of chitosan hydrogel on stem cell engraftment, survival and
[13] H.J. Chun, G.W. Kim, C.H. Kim, Fabrication of porous chitosan scaffold in order to im- homing in the ischemic myocardial microenvironment, Biomaterials (2012)https://
prove biocompatibility, J. Phys. Chem. Solids (2008)https://doi.org/10.1016/j.jpcs. doi.org/10.1016/j.biomaterials.2011.12.044.
2007.10.104. [39] H.V. Almeida, A.D. Dikina, K.J. Mulhall, F.J. O’Brien, E. Alsberg, D.J. Kelly, Porous scaf-
[14] B.G. Amsden, A. Sukarto, D.K. Knight, S.N. Shapka, Methacrylated glycol chitosan as a folds derived from devitalized tissue engineered cartilaginous matrix support chon-
photopolymerizable biomaterial, Biomacromolecules (2007)https://doi.org/10. drogenesis of adult stem cells, ACS Biomater. Sci. Eng (2017)https://doi.org/10.
1021/bm700691e. 1021/acsbiomaterials.7b00019.
[15] H.Y. Nam, S.M. Kwon, H. Chung, S.Y. Lee, S.H. Kwon, H. Jeon, Y. Kim, J.H. Park, J. Kim, [40] Y.N. Wu, Z. Yang, J.H.P. Hui, H.W. Ouyang, E.H. Lee, Cartilaginous ECM component-
S. Her, Y.K. Oh, I.C. Kwon, K. Kim, S.Y. Jeong, Cellular uptake mechanism and intracel- modification of the micro-bead culture system for chondrogenic differentiation of
lular fate of hydrophobically modified glycol chitosan nanoparticles, J. Control. Re- mesenchymal stem cells, Biomaterials (2007)https://doi.org/10.1016/j.
lease (2009)https://doi.org/10.1016/j.jconrel.2009.01.018. biomaterials.2007.05.039.
[16] T.T. Yu, F.Z. Cui, Q.Y. Meng, J. Wang, D.C. Wu, J. Zhang, X.X. Kou, R.L. Yang, Y. Liu, Y.S. [41] K.R. Brodkin, A.J. García, M.E. Levenston, Chondrocyte phenotypes on different ex-
Zhang, F. Yang, Y.H. Zhou, Influence of surface chemistry on adhesion and osteo/ tracellular matrix monolayers, Biomaterials (2004)https://doi.org/10.1016/j.
odontogenic differentiation of dental pulp stem cells, ACS Biomater. Sci. Eng. 3 biomaterials.2004.01.044.
(2017) 1119–1128, https://doi.org/10.1021/acsbiomaterials.7b00274. [42] M.A. Pratta, W. Yao, C. Decicco, M.D. Tortorella, R.Q. Liu, R.A. Copeland, R. Magolda,
[17] L. Liu, S. Li, H. Garreau, M. Vert, Selective enzymatic degradations of poly (L-lactide) R.C. Newton, J.M. Trzaskos, E.C. Arner, Aggrecan protects cartilage collagen from pro-
and poly(∈-caprolactone) blend films, Biomacromolecules (2000)https://doi.org/ teolytic cleavage, J. Biol. Chem. (2003)https://doi.org/10.1074/jbc.M303737200.
10.1021/bm000046k. [43] J. Dudhia, Aggrecan, aging and assembly in articular cartilage, Cell. Mol. Life Sci.
[18] Biological evaluation of medical devices, Biomed. Saf. Stand. (1996)https://doi.org/ (2005)https://doi.org/10.1007/s00018-005-5217-x.
10.1097/00149078-199604150-00011. [44] J.L. Drury, D.J. Mooney, Hydrogels for tissue engineering: scaffold design variables
[19] H. Park, X. Guo, J.S. Temenoff, Y. Tabata, A.I. Caplan, F.K. Kasper, A.G. Mikos, Effect of and applications, Biomaterials (2003)https://doi.org/10.1016/S0142-9612(03)
swelling ratio of injectable hydrogel composites on chondrogenic differentiation of 00340-5.
460 S. Lee et al. / International Journal of Biological Macromolecules 158 (2020) 452–460

Fig. 10. RT-PCR analysis of (A) AGG (B) COL2 and (C) SOX9 normalized by GAPDH (values are mean ± SD, n = 3, P b 0.05 (*), P b 0.01 (**), P b 0.001 (***)).

You might also like