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

Journal of

Materials Chemistry B
View Article Online
PAPER View Journal | View Issue

The preparation and biocompatible evaluation


of injectable dual crosslinking hyaluronic acid
Cite this: J. Mater. Chem. B, 2019,
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

7, 4413 hydrogels as cytoprotective agents


Wanxu Cao,a Junhui Sui,a Mengcheng Ma,a Yang Xu,a Weimin Lin, b
Yafang Chen,a Yi Man,b Yong Sun, *a Yujiang Fan a and Xingdong Zhanga

Injectable hydrogels attract a great deal of attention due to their in situ formation in vivo, which
minimizes the risk of major trauma during the surgical procedure as compared to traditional hydrogel
scaffolds. Click chemistry is widely used in the preparation of injectable hydrogels, although some of
them require photoinitiators or catalysts that may be cytotoxic and impair the proliferation of
encapsulated cells. In the present study, an injectable dual crosslinking hydrogel was designed by a
thiol–ene click reaction and conversion between sulfhydryl and disulfide bonds at a neutral pH. The
sulfhydryl group and acrylate group were successfully grafted onto HA and characterized using FT-IR
1
and H-NMR. The gelation time, morphology, injection force, swelling, degradation, mechanical
properties and drug release behavior of the hydrogel varied with the molecular weight of the hydrogel
(Mw = 0.1, 1.0, 2.0 MDa). MTT, FDA/PI staining and scanning electron microscopy (SEM) showed that the
encapsulated L929 proliferated well in different molecular weight hydrogels. It was also observed that in
a low molecular weight (0.1 MDa) hydrogel system, the cells were better distributed and secreted more
Received 29th April 2019, extracellular matrix. Subcutaneous injection in mice also demonstrated that hydrogels could support the
Accepted 2nd June 2019 proliferation of encapsulated L929 cells in vivo, and no significant infiltration of peripheral cells into the
DOI: 10.1039/c9tb00839j interior of the material was observed. These results indicate that this injectable hyaluronan-based
hydrogel is an excellent cell protectant and exhibits good biocompatibility, with potential for biomedical
rsc.li/materials-b applications such as cell delivery and postoperative anti-adhesion.

1. Introduction biochemical and mechanical properties could also improve cell


survival and retention, and promote cell-mediated tissue matrix
Direct injection delivery of cell suspensions has been intensively production, vascularization, and integration with endogenous
used in the domains of clinical application due to its simple tissues.3,4 Tissue engineering scaffolds based on biomaterials
operation and low invasiveness. However, such an approach are mainly independent of implantable pre-fabricated and inject-
might bring about low cell survival and retention due to cell able in situ consolidation forming technology.5 The implantable
shearing during injection, hypoxic microenvironments with scaffolds require invasive surgery for transplantation and initiate
limited nutrient supply and dynamic mechanical stress in the the risk of external trauma during the implantation procedure. In
recipient tissue, and may even generate unsatisfactory therapeutic addition, the pre-fabricated process might influence uniform
efficiency. Compared with individual cell injection, biomaterials cell distribution in most individuals, and even cause a certain
could serve as 3D tissue engineering scaffolds for cell delivery, amount of cell death. However, injectable scaffolds, as an
which could protect cells from unwanted injection shearing, emerging strategy for therapeutic drugs and cells delivery, have
provide cell growth spatial structural support, and improve been widely explored for biomedical applications.6 They are self-
therapeutic efficiency.1,2 In addition, biomaterials with optimized adaptable in matching complex defect tissue models under
physiological conditions, they could guarantee intensive mixing
between injectable biomaterials and encapsulate cells or drugs,
a
National Engineering Research Center for Biomaterials, Sichuan University, minimize the surgical trauma, and facilitate wound healing via a
29 Wangjiang Road, Chengdu 610064, P. R. China.
minimally invasive manner to reduce patient pain and medical
E-mail: sunyong8702@scu.edu.cn; Fax: +86-28-85417654; Tel: +86-28-85417654
b
Diseases and Department of Oral Implantology, Department of Oral Implantology,
expenses.7–10
West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Biocompatibility is one of the major issues associated with
P. R. China implanted materials.11 Synthetic hydrogels are rich in chemical

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 7, 4413--4423 | 4413
View Article Online

Paper Journal of Materials Chemistry B

modification sites and are capable of providing better mechanical were investigated in this study. Finally, the biocompatibility of
properties. However, some chemical reactions involving the cross- these hydrogels and the influence of molecular weight on
linking of hydrogels interfere with the biological processes of cells, cytoprotective were studied in vitro and in vivo.
and the degradation products of certain scaffolds exhibit varying
degrees of cytotoxicity, which may lead to a sharp or long-term
decline in cell survival.12,13 Naturally derived hydrogels, such as 2. Materials and methods
collagen and Matrigel, have excellent cell compatibility and bio- 2.1. Materials
degradability but they may cause inflammatory responses associated
with their immunogenicity.14 Hyaluronic acid (HA) derivatives as Hyaluronic acid (HA, cosmetic grade, Mw = 0.1, 1.0, 2.0 MDa)
injectable scaffolds have received widespread attention because of was obtained from Bloomage Freda Biopharm Corporation
(Shandong, China). 1-Ethyl-3-(3-dimethyllaminopropyl)carbodiimide
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

their excellent biocompatibility, moisture capacity, viscoelasticity,


lubricity and non-immunogenicity.15,16 Various chemical and hydrochloride (EDCI, 99%), N-hydroxysuccinimide (NHS, 99%),
physical modification methods have been applied to obtain sodium chloride (NaCl, 99%), ethylenediaminetetraacetic acid
hyaluronan-based hydrogels,17,18 such as enzymatically cross- (EDTA, 99%), cysteamine hydrochloride (CSAHCl, 99%), adipic
linking,16 photo polymerization,19 disulfide bond formation20 acid dihydrazide (ADH, 99%), N-acryloxysuccinimide (NHS-Ac),
and click chemistry-mediated hydrogels21 for chemical cross- dithiothreitol (DTT, 99%) and 4-(2-hydroxyethyl)-1-piperazine
linking, together with freeze–thaw-induced22 for physical cross- ethanesulfonic acid (HEPES) were purchased from Best-Reagent
linking. Click chemistry has been intensively demonstrated in Corporation (Chengdu, China). 3-(4,5-Dimethyl-2-thiazolyl)-2,5-
cross-linking hydrogel networks due to their significant features diphenyl-2-H-tetrazolium bromide (MTT, 98%), DMEM medium
such as high reaction speed, high selectivity, and mild reaction (Hyclone) and phosphate buffered saline (PBS, 0.0067 M) were
conditions.23,24 Several click reactions have been greatly expanded purchased from Thermo Fisher Scientific Corporation (USA). Fetal
in the drug delivery and tissue engineering field including azide– bovine serum (FBS, Gibco, South America origin) was bought from
alkyne cycloadditions,25 thiol–ene/yne conjugation reactions,26 Life Technologies Corporation (USA). All other chemicals were
Diels–Alder and inverse electron demand Diels–Alder cyclo- used as-received unless otherwise specified.
additions,27,28 strain-promoted azide–alkyne cycloadditions,29
tetrazine–norbornene reactions,30 and so on. Nevertheless, some 2.2. Fabrication of the HA-SH–Ac-HA hydrogel
of these click reactions require transition-metal catalysts, UV light 2.2.1. Preparation and characterization of hyaluronic acid
irradiation, or complex of precursor synthetic approaches, which derivatives (HA-SH, HA-ADH, and HA-Ac). Thiolated hyaluronic
complicate the experimental operations and restrict the applications. acid (HA-SH) was synthesized through the following process as
Compared to other traditional cross-linking methods of fabricating a representative example. Firstly, HA (1 mmol) and NHS
hyaluronic acid hydrogel, the Michael addition reaction conditions (2 mmol) were dissolved thoroughly in 80 mL of deionized
of thiol–ene are relatively mild, and no initiator or cross-linking water under stirring at room temperature. Then, EDCI (3 mmol)
agents are required, which guarantee the biocompatibility of the in solid form was added into the mixture solution and reacted
injectable scaffolds.31–33 Moreover, hyaluronic acid lacks cell for 2 h to activate the carboxylic group of HA. Subsequently,
adhesion sites and thus, it is difficult for cells to obtain good CSAHCl (3 mmol) was dissolved in 10 mL of deionized water
adhesion; as such, hyaluronan-based biomaterials are widely and added to the mixture solution. The pH of the mixture
used for postoperative anti-adhesion, as cytoprotective agents solution was adjusted to 4.75 by adding 1.0 M NaOH or 1.0 M
and carriers, and in other biomedical applications.34–36 HCl during the reaction process. After stirring for 24 h, the
In previous studies, Huang et al. designed a pH-responsive solution mixture was transferred to the dialysis tubing (8000–
hydrogel via a spontaneous amino–yne click reaction,37 which 14 000 MWCO) and dialyzed exhaustively against dilute HCl
had more serious cytotoxicity in high concentration. Further- solution (pH 3.5) containing NaCl (concentration gradient from
more, Segura et al. used acrylated hyaluronic acid and MMP 100 mM to 0) for 72 h. The acidified solution was then
peptide to build an injectable hydrogel, but the strength of the lyophilized to yield HA-SH as a spongy solid. The chemical
hydrogel was weak and its range of application is limited to brain structures of HA and HA-SH were determined by 1H-NMR
repair.38 Herein, we have designed an in situ-formed injectable (400 MHz, Bruker AMX-400, USA) using D2O as a solvent and
dual crosslinking hydrogel via a thiol–ene click reaction and FT-IR (Nicolet 6700, USA).
conversion between sulfhydryl and disulfide bonds. The use Acrylated hyaluronic acid (HA-Ac) was prepared using a two-
of click chemistry ensured that the hydrogel could be formed in step synthesis through the following process. Firstly, HA was
a relatively short period of time, while the dual crosslinking activated to get the HA-NHS solution as described above. This
structure endowed the gel with more excellent stability and was then added into 50 mL of ADH (10 mmol) deionized
intelligent responsiveness to cell proliferation. Acrylated hyaluronic solution, and the pH value of the mixture solution was kept at
acid (HA-Ac) and thiolated hyaluronic acid (HA-SH) were synthe- 4.75. After 24 h, the solution was transferred to the dialysis tubing
sized and characterized by 1H-NMR and FT-IR. Also, the effects of (8000–14 000 MWCO) and dialyzed against NaCl solution
molecular weight and concentrations of the polymer precursors on (concentration gradient from 100 mM to 0) for 72 h. The purified
the gelation time, morphology, injection force, swelling properties, intermediate HA-ADH was lyophilized and stored at 20 1C. Next,
degradation rates, mechanical properties and drug release behavior HA-ADH (1 mmol) was reacted with NHS-Ac (5 mmol) in HEPES

4414 | J. Mater. Chem. B, 2019, 7, 4413--4423 This journal is © The Royal Society of Chemistry 2019
View Article Online

Journal of Materials Chemistry B Paper

buffer (10 mM HEPES, 150 mM NaCl, 10 mM EDTA, pH 7.2) for measured in three replicates. The swelling rate of the hydrogel
12 h at room temperature, and then dialyzed for 72 h under the was calculated by the following formula:
same conditions. The solution was then lyophilized to yield HA-Ac as
Swelling ratio = (W1  W0)/W0
a spongy solid. The chemical structures of HA-ADH and HA-Ac were
determined by FT-IR and 1H-NMR using D2O as a solvent.
2.3.3. The degradation performance. The six groups of
2.2.2. The fabrication of HA-SH–Ac-HA hydrogel. The
prepared hydrogels were weighed (W0) as mentioned above
mixed solution of HA-SH and HA-Ac can rapidly crosslink under
and then the degradation performances were measured using
mild conditions by the Michael addition reaction between thiol
two methods. On the one hand, they were immersed in PBS
groups and double bonds, while the free sulfhydryl groups
solution containing hyaluronidase (50 mg mL1) and placed in a
can also undergo oxidation reactions to form disulfide bonds
constant temperature shaker at 90 rpm at 37 1C. On the other
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

by self-crosslinking. Equal volumes of lyophilized HA-SH and


hand, they were immersed in PBS solution containing DTT
HA-Ac polymer (Mw = 0.1, 1.0, 2.0 MDa) were dissolved in water
(0.1 mM) and placed in a constant temperature shaker at 90 rpm at
at pH 3.5 and 7, respectively, and then mixed to form a
37 1C. To calculate the weight loss percentage, the hydrogels were
homogeneous acidic solution. To fabricate the hydrogel, the
taken out at a certain time point, washed in distilled water and
pH value of the mixture solution was adjusted to 7.4 to get a
weighed again (Wr). Every sample was measured in three replicates.
neutral precursor solution by adding 1.0 M NaOH. Afterwards,
The degradation behavior of the hydrogels was expressed as the
the freshly prepared neutral precursor solution was immediately
percentage of weight loss and was calculated as follows:
injected into the self-made annular molds (diameter 8.5 mm,
height 3.0 mm) to acquire the HA-SH–Ac-HA hydrogel. The percentage of the remaining hydrogel weight = Wr/W0  100%
2.3. Characterization of the HA-SH–Ac-HA hydrogel
2.3.4. Dynamic mechanical analysis (DMA). Hydrogel samples
2.3.1. The gelation time, morphology and injection force. of six groups were prepared as mentioned above for mechanical
Rheology measurements were performed to analyze the accurate analysis. The storage modulus (G0 ) and loss modulus (G00 ) of the
gelation time using a TA Discovery DHR-2 rheometer with hydrogels were measured using a Dynamic Mechanical Analyzer
parallel-plate geometry (40 mm) and a 1.0 mm gap. The dynamic (DMA, TA-Q800, USA) instrument in the multi-frequency mode
strain sweep was operated with constant ramp strain of 10% and (amplitude of 40 mm, a preload force of 0.002 N and a force track
frequency of 1 Hz at time sweep in 250 s. Meanwhile, an integrated of 105%) with a fixed frequency of 1–10 Hz at room temperature.
temperature controller was used to maintain the temperature at Every sample was measured in three replicates.
37 1C. To investigate the influence of the molecular weight of HA 2.3.5. The simulation of albumen release behavior in the
(Mw = 0.1, 1.0, 2.0 MDa) and polymer concentration (10 mg mL1, hydrogel in vitro. Hydrogel samples of six groups were prepared
20 mg mL1) on the gelation time of the hydrogel, a series of as mentioned above for in vitro simulation of the controlled
HA-SH–Ac-HA solutions were prepared and the gelation time release behavior. BSA albumen (1 mg) was added to a certain
was recorded. The gross view of each sample was investigated concentration of acidic HA-SH solution, shaken evenly with a
using a digital camera. Subsequently, the hydrogel was rinsed vortex shaker, and then an equal volume of HA-Ac solution was
three times with deionized H2O and immersed in liquid nitro- added. After forming a homogeneous solution, the pH of the gel
gen immediately before lyophilization. The cross-section of the was adjusted by adding 1.0 M NaOH. After lyophilization, six dry
hydrogel was further sputter-coated with a gold layer to observe hydrogels loaded with BSA were obtained at the final concentration.
the morphology and internal structure by scanning electron Six dry BSA-loaded hydrogels were immersed in 5 mL of PBS buffer
microscopy (SEM, HITACHI S-800, Japan). The pore size of the in a 37 1C water bath to study the in vitro release behavior of the
hydrogel was analysed by ImageJ software (1.51j8, National BSA. At the set time point, 0.1 mL of the release solution was taken
Institutes of Health) based on SEM images. The injection force while the same amount of PBS solution was added to the release
was measured using an electromechanical universal testing system. Next, 5 mL of Coomassie Brilliant Blue G250 solution was
machine (Shimadzu Autograph AGS-X, Japan) with a 30 mm min1 added to 0.1 mL of the release solution, the absorbance was
crosshead displacement up to a maximum load of 500 N. A 1 ml measured at 595 nm using an ultraviolet spectrophotometer, and
syringe (needle gauge: 26 G (0.45 mm); 7 mm diameter and 146 mm set in parallel three times to calculate the cumulative release of BSA
in length) fitted with a cannula of 5 mm inner diameter and 75 mm according to the standard absorption curve. The cumulative release
in length was applied. Each sample was measured in five replicates. of BSA (Mt) was calculated as follows:
2.3.2. The swelling properties. Six groups of hydrogel
solution (pH = 7.4) were immediately injected into self-made The cumulative release of BSA (Mt) = ct  5
annular molds (diameter 8.5 mm, height 3.0 mm) to form
+ (c1 + c2 + c3 +    + ct1)  0.1
hydrogels at 37 1C. The hydrogels were freeze-dried and weighed
(W0) and then immersed into 10 mL PBS buffer (pH = 7.4) and The percentage of the cumulative release of BSA (CR) was
placed in a constant temperature shaker (ZHWY-2012C, Shanghai calculated as follows:
Zhicheng, China) at 90 rpm and 37 1C. To calculate the swelling
rate, the hydrogels were weighed (W1) at a certain time interval The percentage of the cumulative release of BSA (CR)
until swelling equilibrium was reached. Every sample was = Mt/M0  100%

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 7, 4413--4423 | 4415
View Article Online

Paper Journal of Materials Chemistry B

where Mt is the cumulative release of BSA at time t, ct is the fixed in 2.5% glutaraldehyde solution for 2.5 h at room temperature.
concentration of BSA released in the medium at time t, and M0 They were subsequently dehydrated against a graded series of
is the initial negative initial loading of BSA in the drug-loaded alcohol (20%, 40%, 60%, 80%, 100%, 100%, 100%) for 15 minutes,
hydrogel. Every sample was measured in three replicates. and dried by critical point drying for 2 hours, and then cryo-
fractured in liquid nitrogen to observe the internal structure by
2.4. Cytotoxicity, cell viability and proliferation in vitro SEM. Cytoskeletal F-actin stain was performed by using rhodamine–
2.4.1. Cytotoxicity in vitro. MTT experiments were performed phalloidin (red, Ex = 496 nm, Em = 516 nm) and DAPI (blue, Ex =
to evaluate the cytotoxicity of the hydrogel precursor polymers 364 nm, Em = 454 nm) after incubation for 4, 7, 14 days. More
(HA-SH, HA-ADH, HA-Ac) on mice fibroblast cells (L929) and human specifically, the cell-hydrogel constructs were immersed in PBS to
breast cancer cells (MCF-7). L929 cells and MCF-7 cells were cultured remove the culture medium, and then were treated with 4.0%
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

using standard procedures in DMEM (containing 1% penicillin– formaldehyde to fix cells. Subsequently, the cell membranes were
streptomycin and 10% FBS) and subsequently suspended and ruptured by 0.5% Triton X-100 solution, and the cytoskeletal F-actin
placed in a 96-well plate with a concentration of 2000 cells per well of cells was stained by rhodamine–phalloidin for 40 minutes, next,
for 48 h in an incubator (37 1C, 5% CO2). Meanwhile, the hydrogel the DAPI solution was used to stain cell nucleus for 1 minute. The
precursor polymers with different molecular weights (Mw = 0.1, fluorescence pictures of L929 cells were taken by CLSM.
1.0, 2.0 MDa) were dissolved in PBS and diluted with cell
culture medium to gradient concentrations from 4 mg mL1 to 2.5. Biocompatibility studies in vivo
2500 mg mL1. Afterwards, the cell culture medium was removed All animal studies were approved by the Sichuan University
from the wells and replaced with hydrogel precursor polymer Medical Ethics Committee. All animal procedures were performed
solutions. After 48 h, the MTT assay was carried out. Finally, the in accordance with the guidelines for the care and use of
absorbance was measured using a Thermo Scientific Varioskan Laboratory Animals of Sichuan University. Female BALB/c mice,
Flash Multiscan Spectrum at the wavelength of 490 nm. aged 5–6 weeks, were supplied by the Experimental Animal
2.4.2. Fabrication of 3D cell-hydrogel constructs. For cell Center in Chengdu Dashuo Corporation (Chengdu, China).
encapsulation, spongy HA-SH and HA-Ac solids were soaked in They were fed ad libitum in a standard animal facility with
75% ethanol for 5 h, and then the HA-SH and HA-Ac polymers 12 h light–dark cycles and kept at a constant temperature of
were dried overnight on a clean bench. Thereafter, the dried 20–22 1C and a relative humidity of 50–60%. The sterilized
HA-SH and HA-Ac were dissolved in serum-free DMEM medium, precursor polymer solution was prepared by the method as
and 1.0 M NaOH and 1.0 M HCl solutions (0.22 mm filter to sterilize) described above. Subsequently, 0.15 mL sterilized precursor
were prepared to adjust the pH value to 7.4. L929 suspension mixture solutions (Mw = 0.1, 1.0, 2.0 MDa, n = 3 per time point)
(100 mL) was immediately added to the polymer solution to were subcutaneously injected separately into the back of the
obtain the mixture solution with a cell concentration of 2  female BALB/c mice using a syringe with a 26 gauge needle. On
106 cells per mL. The freshly prepared mixture solution was the 7th, 14th, 21st, and 28th days, the mice were euthanized,
immediately injected into a cylindrical mold (8.5 mm in diameter and the hydrogels were removed and recorded with a digital
and 3.0 mm in length). After several minutes, the hydrogels were camera, and were promptly stained with FDA/PI and then
detached from the mold and immersed into DMEM medium observed by CLSM to evaluate the cell viability in vivo as
containing 1% penicillin–streptomycin and 10% FBS on 6-well mentioned in the experimental procedure in Section 2.4.3. For
tissue culture plate at 37 1C in 5% CO2 (MCO-15AC, SANYO, Japan). histopathological evaluation, the freshly acquired hydrogels
2.4.3. Cell viability and proliferation. After the cell-hydrogel were washed twice with PBS solution, and then fixed by 4%
constructs were cultured for 1, 4, 7, 14, 21 and 28 days in vitro, paraformaldehyde solution for 48 h at 4 1C. The hydrogel pieces
respectively, the constructs were removed. Live/dead assays were were embedded in optimal cutting temperature compound (OCT,
used to observe the cell viability and distribution of encapsulated Tissue-Tek) and were then sliced into a thickness of 5–10 mm. These
cells. The cell-hydrogel constructs were immersed in PBS solution samples were stained by hematoxylin–eosin (H&E) and DAPI.
containing 1 mg mL1 of fluorescein diacetate (FDA) (live, green) and
1 mg mL1 of propidium iodide (PI) (dead, red) for 2 min to stain the 2.6. Statistical analysis
cells, and then washed with PBS solution again. The viability and
The statistically significant differences among groups were
distribution of L929 in hydrogels were observed on a Leica TCS SP5
verified using Student’s t-test. Differences were considered
confocal laser scanning microscope (CLSM). The proliferation of
significant if P o 0.05 (*), P o 0.01 (**) or P o 0.001 (***)
L929 cells was quantified by MTT assays. After 4, 7, 14, 21 and
and were marked accordingly in the figures.
28 days of cultivation, the proliferative capacity of L929 was
measured. The optical absorbance value of the solution in each
well was measured at 490 nm using a Multiscan Spectrum 3. Results and discussion
(Varioskan Flash, Thermo Fisher Scientific, USA). Each sample
was replicated three times. 3.1. Synthesis and characterization of HA-SH/HA-ADH/HA-Ac
2.4.4. Cell morphology and cytoskeletal F-actin staining. Chan et al. believed that the sequence of reaction efficiency was
The morphology of L929 encapsulated in hydrogels was observed propylmaleimide 4 diethyl fumarate 4 diethyl maleate 4
using a scanning electron microscope (SEM). The samples were dimethylacrylamide 4 acrylonitrile 4 ethyl crotonate 4 ethyl

4416 | J. Mater. Chem. B, 2019, 7, 4413--4423 This journal is © The Royal Society of Chemistry 2019
View Article Online

Journal of Materials Chemistry B Paper

cinnamate 4 ethyl methacrylate.39 Besides, Morgan et al. studied group and the hydroxyl group. Comparatively, HA-ADH had an
the influence of the molecular structure reaction of thiol–ene by absorption peak at 1650 cm1, which was due to the stretching
experimental means. It was believed that the reaction rate was vibration of –CQN, while two absorption peaks at 1607 cm1 and
mainly determined by the addition rate of sulfhydryl and double 1560 cm1 were attributed to the stretching vibration of –CQO.
bond, and the decrease in the charge density of the CQC double At 1660 cm1, HA-Ac had an absorption peak, which was due to
bond resulted in the lower reaction efficiency.40 According to the the stretching vibration of the double bond. The characterization
above theory, the designed Michael addition reaction in this of thiolated hyaluronic acid was conducted as previously
paper had a relatively high reaction efficiency. The synthesis reported.20,41 In the HA-SH spectrum, a sharp peak appeared at
protocol for the hyaluronic acid derivatives is shown in Fig. 1A. At 1665 cm1 as compared with HA, showing the existence of the
first, the carboxylic group of HA was reacted with NHS to prepare amide I band. Due to the presence of the III band, peak splitting
occurred at 1260 cm1. The above results confirmed the success-
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

the HA-NHS active ester using EDCI as the condensing agent.


Then, HA-NHS was reacted with ADH/CSAHCl to synthesize ful modification of ADH, Ac and SH with hyaluronic acid.
the HA-ADH/HA-SH polymer. Subsequently, to obtain the pure The 1H-NMR spectrum was used to confirm the existence of
product, HA-SH raw product was dialyzed exhaustively against the conjugated thiol groups, and the typical spectra of native
dilute HCl aqueous solution (pH = 3.5) which prevented the thiol HA and thiolated HA are shown in Fig. 1C. Compared with the
groups from reacting with each other to form the disulfide bond, spectrum of native HA, it was obviously seen that new resonant
and the addition of NaCl could decrease the viscosity of the peaks of HA-SH appeared at 2.8 ppm, which represented the
solution. The HA-ADH was prepared by a carbodiimide-mediated methylene protons of the –CH2CH2SH.42 The degree of sub-
coupling reaction between HA and ADH, and was dialyzed against stitution could be calculated by taking the ratio of peaks at
100 mM NaCl solution to acquire a pure product. HA-Ac was 2.8 ppm to the singlet peak of the acetyl methyl protons in HA
prepared via the nucleophilic substitution of NHS-Ac with HA-ADH (1.88 ppm). The 1H-NMR spectrum of HA-ADH showed new
in HEPES buffer, and the product was purified as HA-Ac as resonant peaks at 1.6 ppm and 2.3 ppm, representing the eight
above. The reaction efficiency of the thiol–ene click reaction was hydrogens of the methylene groups on the ADH. The degree of
significantly affected by the different double bond activities on substitution could be calculated by taking the ratio of peaks at
different molecular structures. 1.6 ppm and 2.3 ppm to the singlet peak of the acetyl methyl
As shown in Fig. 1D, HA-ADH has a broad association peak protons in HA (1.88 ppm).43 The 1H-NMR spectrum of HA-Ac
at 3300–3400 cm1. As a derivative of HA, an amino group was showed new resonant peaks at 5.65 ppm and 6.2 ppm, representing
added to the HA-ADH chain, and the amino group has an the cis and trans acrylate hydrogens, respectively. The degree of
absorption peak at 3300–3400 cm1. However, the hydroxyl group substitution was calculated by taking the ratio of peaks at 5.65 and
on the chain also has an absorption peak at 3200–3650 cm1, so 6.2 ppm to the singlet peak of the acetyl methyl protons in HA
that a broad association peak appeared, which was as a result of (1.88 ppm);44 the higher the molecular weight of HA was, the
the superposition of the stretching vibration peaks of the amino lower the degree of substitution was (Fig. 1B). The possible

Fig. 1 The synthesis protocol (A), degree of substitution (B), 1H-NMR (C), and FT-IR (D) of the hyaluronic acid derivatives (HA-SH, HA-ADH and HA-Ac)
are presented.

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 7, 4413--4423 | 4417
View Article Online

Paper Journal of Materials Chemistry B

reason is that the high viscosity for high molecular weight HA Meanwhile, the increase in the hyaluronic acid concentration
could result in lower liquidity,45 which might decrease the ensured greater hydrogen bond content.
opportunity for the reaction substrates to touch each other The in vitro degradation profile of hydrogels is shown in
and cause lower reaction activity. Fig. 3A and B. With the molecular weight and concentration
increasing, the degradation rate gradually decreased in PBS
3.2. Preparation and characterization of the dual crosslinking solution containing hyaluronidase. This was due to the fact that
hydrogels the glycosidic bonds in the molecular structure of hyaluronic
The fast thiol/acrylate Michael addition reaction contributed to acid hydrogel with low molecular weight were more likely to be
the establishment of the primary network. The disulfide bonding exposed and then be degraded. It was noteworthy that the early
network as a secondary network could be slowly and spontaneously degradation rate (from 0 h to 40 h) was slightly higher than that
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

formed by the oxidation of thiol groups. Meanwhile, the secondary of the late stage. However, in PBS solution containing DTT, only
network further stabilized and strengthened the primary network. the disulfide bonds in the hydrogel were degraded. In Fig. 3B,
3.2.1. Gelation time. In the rheological test, the intersec- the hydrogels degraded faster before 8 h, and then their masses
tion of the storage modulus and the loss modulus indicates the gradually stabilized. This phenomenon was due to the rapid
gelation time at the molecular level as shown in Fig. 2A and B. collapse of the secondary structure constructed by the S–S bond
The molecular weight and solution concentration of hyaluronic in the first 8 h hydrogel under the action of DTT. After the DTT
acid both quite significantly affect the gelation time, but the completely degraded the S–S bond, the six groups of hydrogels
concentration exerted a greater impact on the gelation time. were equivalent to degradation in PBS buffer alone; this process
Specifically, at a molecular weight of 2.0 MDa, the gelation can was very slow and required a long degradation time.
be quickly completed at a minimum of 10 s; with the decrease in 3.2.5. DMA. The storage modulus (G 0 ) and loss modulus
molecular weight, the gelation time gradually increased to about 2 (G00 ) of hydrogels represent the energy stored or consumed per
minutes. Compared with the lower concentration, increasing the unit of strain, respectively. As shown in Fig. 3D and E, with the same
precursor solution concentration to 20 mg mL1 enhanced the concentration of hydrogels, the higher the molecular weight, the
probability of the Michael addition reaction between reactive greater the storage modulus, and the storage modulus (G0 ) slightly
functional groups per unit volume, which accelerated the increased with increasing frequency. When the concentration of
formation of 3D networks. hyaluronic acid increased from 10 mg mL1 to 20 mg mL1, the
3.2.2. Morphology. The macroscopic pictures and internal storage modulus increased by about twofold. On the other hand, the
network structures of the hydrogels with different molecular loss modulus of the hydrogel progressively declined along with
weights and concentrations of the hydrogels are shown in increasing frequency. This trend showed that the hydrogels could
Fig. 2B and D. It can be seen that all the hydrogels are transparent. increase the storage modulus (G0 ) and reduce the loss modulus (G00 )
Pore size is an important parameter of three-dimensional porous when subjected to periodic higher frequency shock.
scaffolds; it has a great impact on material exchange inside and 3.2.6. The simulation of albumen release behavior in the
outside the scaffold, affecting cell proliferation and regulating cell hydrogel in vitro. Fig. 3F shows the release profile of BSA in six
physiology.46 A typical uniform sponge-like porous structure with a groups of hydrogels at 37 1C in PBS buffer. It can be seen from
pore size of 5 mm to 30 mm was observed. With the decrease in the figure that in the PBS buffer solution the BSA was released
molecular weight and concentration, the porosity of the hydrogel faster in the first 48 hours in the six groups of hydrogels, and the
network increased as shown in Fig. 2E. cumulative release percentage of BSA in the high concentration
3.2.3. Injection force. The HA-Ac and HA-SH were dis- group (20 mg mL1) of hydrogels was about 40%; for hydrogels in
solved in deionized water and their final concentrations were the low concentration group (10 mg mL1), the cumulative
adjusted to 20 mg mL1 and 10 mg mL1, respectively by release percentage of BSA was up to 75%. After 48 hours, the
mixing. The result (Fig. 2C) showed that the injection force of release rate of BSA from six groups hydrogels was slowed down. The
20 mg mL1 (8–13 N) was significantly higher than that of cumulative release percentage of BSA in the high concentration
10 mg mL1 (2–4 N), while the molecular weight had little effect group (20 mg mL1) hydrogel was about 60%, while in the low
on the injection force. One possible reason was that the increase concentration group (10 mg mL1), the hydrogel was about 83% in
in hyaluronic acid concentration resulted in the increase in the 240 hours. The hydrogel from the high concentration group
covalent cross-linking sites in the dual crosslinking hydrogels. (20 mg mL1) had a lower release rate than the low concentration
3.2.4. Swelling and degradation. As shown in Fig. 3C, all group (10 mg mL1) because of its crosslink density. The higher
the hydrogels reached the equilibrium of swelling in about crosslink density of the hydrogel and the smaller average molecular
10 hours, and the maximal value of the swelling ratio was up to distance between adjacent cross-linking points prevented the diffu-
24 (0.1 MDa, 10 mg mL1). Generally speaking, the lower the sion of BSA molecules from the hydrogel. However, even in the
concentration and molecular weight of hyaluronic acid were, lowest cross-linking hydrogel group D (0.1 MDa, 10 mg mL1), BSA
the greater the equilibrium swelling rate of the gel was. With was not completely released in 240 hours, probably because a partial
the molecular weight increasing, the single molecular chains Michael addition reaction occurs between the amino on the BSA
formed many hydrogen bonds, they were more likely to become and the CQC double bond on the HA-Ac during BSA loading,
entangled, and at the same time, the cross-linking points were which allows the BSA bond in the gel network. These results
densely added, resulting in a decrease in the swelling ratio. indicate that the injectable hydrogel has a good sustained

4418 | J. Mater. Chem. B, 2019, 7, 4413--4423 This journal is © The Royal Society of Chemistry 2019
View Article Online

Journal of Materials Chemistry B Paper


Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

Fig. 2 Rheology measurements (A), gelation time and gross view of each sample (B), injection force (C), microstructure (D) and mean pore size (E) of
hyaluronic acid hydrogels are presented; scale bar: 10 mm. Error bars represent standard deviation (N = 5) in (C) and standard error of mean in (E).
(*P o 0.05, **P o 0.01, ***P o 0.001).

release of BSA in PBS solution, which could be applied in long- These features facilitate the application of injectable dual cross-
term controlled release medium and protective agents for linking HA-Ac–SH-HA hydrogels as scaffold materials for cell, drug
proteins or cells, and effectively maintain their structure and and bioactive substance delivery.
function. The above results indicate that the interior structure,
gelation time, injection force, swelling and degradation properties, 3.3. Cell viability and proliferation in vitro
mechanical modulus and drug release behavior of dual crosslinking In order to explore the cytotoxicity of the prepared dual cross-
hyaluronan-based hydrogels were precisely controlled by adjusting linking hydrogels, normal cells (L929) and cancer cells (MCF-7)
the concentrations and molecular weights of HA-Ac and HA-SH. were co-cultured with low-concentration aqueous solutions of

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 7, 4413--4423 | 4419
View Article Online

Paper Journal of Materials Chemistry B


Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

Fig. 3 The in vitro simulated degradation behavior under different environmental response conditions with hyaluronidase (A) and DTT (B). The swelling
ratio (C), storage modulus (D), loss modulus (E), and simulation of albumen release behaviour (F) of dual crosslinking hyaluronic acid hydrogels with
different molecular weights.

hydrogel precursor polymers (HA-SH, HA-Ac, HA-ADH) in 96- concentration increasing (from 4 to 2500 mg mL1), but the
well plates, respectively. The results of the MTT assay showed viability of all materials was higher than 80% against L929 cells
that the cell activity presented a slight decline with solution (Fig. 4A) and MCF-7 cells (Fig. 4B), indicating good biocompatibility.

Fig. 4 Biocompatibility of hydrogel precursor polymers (HA-SH, HA-ADH, HA-Ac) against L929 (A) and MCF-7 (B). Live/dead staining of cell-hydrogel constructs
after 1 d, 4 d, 7 d, 14 d, 21 d and 28 d of culture are presented, where the green stain represents the live cells and the red stain represents dead cells (C). The
proliferative capacity of the L929 in hydrogel (D). The scale bar: 100 mm. Error bars represent standard deviation (N = 3) in (D). (*P o 0.05, **P o 0.01, ***P o 0.001).

4420 | J. Mater. Chem. B, 2019, 7, 4413--4423 This journal is © The Royal Society of Chemistry 2019
View Article Online

Journal of Materials Chemistry B Paper

Furthermore, FDA/PI staining and MTT assays were applied the cells and the scaffold. Similarly, the agglomeration proliferation
to observe the cell proliferation profiles in hydrogels. As shown phenomenon was also detected. In addition, the phalloidin/DAPI
in Fig. 4C, all the L929 cells were homogeneously distributed in staining (Fig. 5B) could visually confirm the morphology of the
HA-Ac–SH-HA hydrogels with different molecular weights, and cytoskeleton and nucleus. These phenomena were consistent
had a prominent effect of agglomeration proliferation with the with the previous results.
extension of co-culture time. Interestingly, the HA-Ac–SH-HA
hydrogels with high molecular weights displayed more remark- 3.4. Biocompatibility studies in vivo
able cell agglomeration as compared to that of low molecular The injection force of the 20 mg mL1 gel precursor solution
weights, just as shown in Fig. 4C (2.0 MDa, 28 d). This was obviously higher than that of the 10 mg mL1 gel precursor
was consistent with the conclusion of other similar studies.20 solution, which would inconvenience the actual injection operation
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

Also, the cell displayed a circular morphology, probably because due to a reduction in the actual operation time. Meanwhile, the
hyaluronan-based materials lack cell adhesion sites, which inhibit higher the concentration of the gel precursor solution, the
the stretching of encapsulated cells. In Fig. 4D, the quantitative faster the gelation speed. Therefore, the final concentration
analysis of cell proliferation was determined by MTT assay. It of the 10 mg mL1 was selected for injection. At 1, 2, 3 and 4
could be seen that the number of cells in all groups increased with weeks after injection, no obvious swelling phenomenon was
time, which indicated that the hydrogels could effectively support detected on the back of the mice. After euthanizing the mice, these
the proliferation of the encapsulated cells. hydrogels were removed and presented excellent morphology
To further observe the morphology of the cells and the (Fig. 6A). FDA/PI staining was performed to observe the cell
secretion of the matrix, SEM and cytoskeletal staining were viability by CLSM. As shown in Fig. 6B, compared with blank
performed. The results of SEM and phalloidin/DAPI staining hydrogel groups (1.0 MDa as control), a large number of living
showed that almost all cells exhibited spherical growth. cells with agglomerating growth were observed, and the cell
The SEM (Fig. 5A) showed the abundant extracellular matrix density was obviously increased after 4 weeks of culture.
that was secreted by the encapsulated cells with increased H&E (Fig. 7A) staining results provided a histological
incubation time, which was mainly distributed on the surface of overview of the tissue response at the site of the injection.

Fig. 5 Cell morphologies of cell-hydrogel constructs after 1 d, 4 d, 7 d, 14 d, 21 d and 28 d of culture are presented (A); scale bar: 10 mm. Cytoskeletal
F-actin staining cell-hydrogel constructs after 4 d, 7 d and 14 d of culture are presented (B); scale bar: 25 mm.

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 7, 4413--4423 | 4421
View Article Online

Paper Journal of Materials Chemistry B


Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

Fig. 6 Observations of the subcutaneous injection of hydrogel in BALB/c mice (A), and live/dead staining of cell-hydrogel constructs after 7 d, 14 d, 21 d and 28 d
of subcutaneous injection are presented, where the green stain represents the live cells and the red stain represents dead cells (B). Scale bar: 500 mm and 100 mm.

Fig. 7 HE staining (A) and DAPI staining (B) of cell-hydrogel constructs with different molecular weights after 7 d, 14 d, 21 d and 28 d of subcutaneous
injection; scale bar: 500 mm.

These L929 cells encapsulated in hydrogels were distributed process. Therefore, the injectable dual crosslinking HA-Ac–SH-HA
uniformly after subcutaneous injection. An orderly proliferation hydrogel could rapidly form a 3D structure in situ while minimizing
phenomenon was obvious 4 weeks after injection. Relatively few or invasion and allowing it to remain in the injection site for long
no cells were noticed in the blank hydrogel group from 1 week to 4 periods of time. Thus, the in situ formed HA-Ac–SH-HA hydrogel has
weeks. DAPI (Fig. 7B) staining was adopted to confirm this the potential for applications in postoperative anti-adhesion and
observation. On prolonging the time to 4 weeks, the intensity of other applications that delaminate tissue interfaces.
the internal fluorescence staining of the hydrogels gradually
increased, implying that the encapsulated cells were continuously
proliferating over time. Likewise, the blank hydrogel group only 4. Conclusions
showed a faint fluorescence. These results indicated that injectable
dual crosslinking HA-Ac–SH-HA hydrogel was of ideal biocompat- The injectable dual crosslinking HA-Ac–SH-HA hydrogel was
ibility and could support cell proliferation. It should be noted that prepared by a fast thiol/acrylate Michael addition reaction as
rather high levels of infiltrating inflammatory cells were found at the primary network, and the spontaneous oxidation reaction
early time points (at days 7 and 14) and gradually decreased with of thiol groups as the secondary network. The interior structure,
extending the time to 28 days; they were mainly localized at the gelation time, injection force, swelling and degradation properties,
boundary between surrounding tissue and the hydrogel. The mechanical modulus and drug release behavior of the dual cross-
infiltration of inflammatory cells was hardly observed in the linking hyaluronan-based hydrogel were precisely controlled by
inner areas of the hydrogel, suggesting that the hydrogel did not adjusting the concentrations and molecular weights of HA-Ac and
induce a local inflammatory reaction in the long-term implantation HA-SH. The research results in vivo and in vitro indicate that the

4422 | J. Mater. Chem. B, 2019, 7, 4413--4423 This journal is © The Royal Society of Chemistry 2019
View Article Online

Journal of Materials Chemistry B Paper

HA-Ac–SH-HA hydrogel can effectively support and protect the 20 S. Bian, M. He, J. Sui, H. Cai, Y. Sun, J. Liang, Y. Fan and
agglomeration proliferation of the encapsulated L929 cells and X. Zhang, Colloids Surf., B, 2016, 140, 392–402.
present wonderful biocompatibility. It also has the ability to prevent 21 Y. K. Du, U. P. Shinde, B. Yeon and B. Jeong, Prog. Polym.
cell adhesion and infiltration, which opens up huge possibilities Sci., 2013, 38, 672–701.
and great application potential as a novel surgical anti-adhering 22 Z. Cai, F. Zhang, Y. Wei and H. Zhang, Macromolecules,
material, cytoprotective agent and bioinert tissue filler. 2017, 50, 6647–6658.
23 T. Murakami, H. R. Brown and C. J. Hawker, J. Polym. Sci.,
Part A: Polym. Chem., 2016, 54, 1459–1467.
Conflicts of interest 24 K. S. Anseth and H. A. Klok, Biomacromolecules, 2016, 17, 1–3.
There are no conflicts to declare. 25 S. S. Han, H. Y. Yoon, J. Y. Yhee, M. O. Cho, H. E. Shim,
Published on 11 June 2019. Downloaded by University of Exeter on 1/13/2021 6:26:31 PM.

J. E. Jeong, D. E. Lee, K. Kim, H. Guim, J. H. Lee, K. M. Huh


and S. W. Kang, Polym. Chem., 2018, 9, 20–27.
Acknowledgements 26 Y. Xu, H. Xu, X. Jiang and J. Yin, Adv. Funct. Mater., 2014, 24,
This work was sponsored by National Key Technology R&D Plan 1679–1686.
(Grant No. 2018YFC1105900), National Natural Science Foundation 27 V. X. Truong, M. P. Ablett, S. M. Richardson, J. A. Hoyland
of China (51573113), Sichuan Provincial Key Technology R&D and A. P. Dove, J. Am. Chem. Soc., 2015, 137, 1618–1622.
Program (2016SZ0003, 2019YFS0007), Young Elite Scientists 28 F. Yu, X. Cao, J. Du, G. Wang and X. Chen, ACS Appl. Mater.
Sponsorship Program by CAST (2017QNRC001), and Sichuan Interfaces, 2015, 7, 24023–24031.
university Innovation Spark Project (2018SCUH0089). 29 J. W. Xu, T. M. Filion, F. Prifti and J. Song, Chem. – Asian J.,
2011, 6, 2730–2737.
30 D. L. Alge, M. A. Azagarsamy, D. F. Donohue and K. S. Anseth,
References Biomacromolecules, 2013, 14, 949–953.
1 A. Khademhosseini and R. Langer, Nat. Protoc., 2016, 11, 1775. 31 D. P. Nair, M. Podgorski, S. Chatani, T. Gong, W. Xi, C. R.
2 R. Langer and J. P. Vacanti, Science, 1993, 260, 920–926. Fenoli and C. N. Bowman, Chem. Mater., 2014, 26, 724–744.
3 E. S. Place, N. D. Evans and M. M. Stevens, Nat. Mater., 2009, 32 B. D. Mather, K. Viswanathan, K. M. Miller and T. E. Long,
8, 457–470. Prog. Polym. Sci., 2006, 31, 487–531.
4 M. P. Lutolf and J. A. Hubbell, Nat. Biotechnol., 2005, 23, 47–55. 33 M. H. Lee, Z. Yang, C. W. Lim, Y. H. Lee, S. Dongbang,
5 X. M. Tong and F. Yang, Adv. Healthcare Mater., 2018, 7, e1701065. C. Kang and J. S. Kim, Chem. Rev., 2013, 113, 5071–5109.
6 J. A. Yang, J. Yeom, B. W. Hwang, A. S. Hoffman and 34 Z. Z. Khaing and S. K. Seidlits, J. Mater. Chem. B, 2015, 3,
S. K. Hahn, Prog. Polym. Sci., 2014, 39, 1973–1986. 7850–7866.
7 C. He, S. Kim and D. Lee, J. Controlled Release, 2008, 127, 35 S. Amorim, A. Martins, N. M. Neves, L. R. Rui and R. A. Pires,
189–207. J. Mater. Chem. B, 2014, 2, 6939–6946.
8 L. Yu and J. Ding, Chem. Soc. Rev., 2008, 39, 1473–1481. 36 T. Lee, H. Y. Son, Y. Choi, Y. Shin, S. Oh, J. Kim, Y. M. Huh
9 Y. Yan, M. Li, D. Yang, Q. Wang, F. Liang, X. Qu, D. Qiu and and S. Haam, J. Mater. Chem. B, 2017, 5, 1400–1407.
Z. Yang, Biomacromolecules, 2017, 18, 2128–2138. 37 J. C. Huang and X. L. Jiang, ACS Appl. Mater. Interfaces, 2018,
10 J. Radhakrishnan, A. Subramanian, U. M. Krishnan and 10, 361–370.
S. Sethuraman, Biomacromolecules, 2017, 18, 1–26. 38 P. Moshayedi, L. R. Nih, I. L. Llorente, A. R. Berg,
11 D. F. Williams, ACS Biomater. Sci. Eng., 2017, 3, 2–35. J. Cinkornpumin, W. E. Lowry, T. Segura and S. T. Carmichael,
12 M. K. Nguyen and E. Alsberg, Prog. Polym. Sci., 2014, 39, Biomaterials, 2016, 105, 145–155.
1236–1265. 39 J. W. Chan, C. E. Hoyle, A. B. Lowe and M. Bowman,
13 E. S. Place, J. H. George, C. K. Williams and M. M. Stevens, Macromolecules, 2010, 43, 6381–6388.
Chem. Soc. Rev., 2009, 38, 1139–1151. 40 C. R. Morgan, F. Magnotta and A. D. Ketley, J. Polym. Sci.,
14 W. Zhao, X. Jin, Y. Cong, Y. Liu and J. Fu, J. Chem. Technol. Polym. Chem. Ed., 1977, 15, 627–645.
Biotechnol., 2013, 88, 327–339. 41 M. M. He, J. H. Sui, Y. F. Chen, S. Q. Bian, Y. N. Cui,
15 C. X. Liu, K. H. Bae, A. Yamashita, J. E. Chung and C. C. Zhou, Y. Sun, J. Liang, Y. J. Fan and X. D. Zhang,
M. Kurisawa, Biomacromolecules, 2017, 18, 3143–3155. J. Mater. Chem. B, 2017, 5, 4852–4862.
16 G. Huang and H. Huang, J. Controlled Release, 2018, 278, 42 Y. J. Jie, W. Di, W. D. Cheng and Y. Y. Zi, Chem. Commun.,
122–126. 2013, 49, 6057–6059.
17 C. E. Schanté, G. Zuber, C. Herlin and T. F. Vandamme, 43 T. Pouyani, G. S. Harbison and G. D. Prestwich, J. Am. Chem.
Carbohydr. Polym., 2011, 85, 469–489. Soc., 1994, 116, 7515–7522.
18 M. Hemshekhar, R. M. Thushara, S. Chandranayaka, 44 C. Cam, S. Zhu, N. F. Truong, P. O. Scumpia and T. Segura,
L. S. Sherman, K. Kemparaju and K. S. Girish, Int. J. Biol. J. Mater. Chem. B, 2015, 3, 7986–7992.
Macromol., 2016, 86, 917–928. 45 E. Fouissac, M. Milas and M. Rinaudo, Macromolecules,
19 X. P. Li, Y. Y. Teng, J. Liu, H. Lin, Y. J. Fan and X. D. Zhang, 1993, 26, 6945–6951.
J. Mater. Chem. B, 2017, 5, 5109–5119. 46 D. Seliktar, Science, 2012, 336, 1124–1128.

This journal is © The Royal Society of Chemistry 2019 J. Mater. Chem. B, 2019, 7, 4413--4423 | 4423

You might also like