You are on page 1of 11

Feature Article

www.mrc-journal.de

Transparent Conductive Supramolecular Hydrogels with


Stimuli-Responsive Properties for On-Demand Dissolvable
Diabetic Foot Wound Dressings

Yue Zhao, Zuhao Li, Qiuju Li, Longfei Yang, Hou Liu, Ruyue Yan, Lizhi Xiao, He Liu,*
Jingcheng Wang,* Bai Yang, and Quan Lin*

1. Introduction
Diabetic foot ulcers (DFU) remain a very considerable health care burden,
and their treatment is difficult. Hydrogel-based wound dressings are Diabetic foot ulcer (DFU) is a common
appealing to provide an optimal environment for wound repair. However, complication in patients with diabetic
hyperglycemia, which leaves patients at
the currently available hydrogel dressings still need surgical or mechanical
increased high risk of morbidity, infec-
debridement from the wound, causing reinjury of the newly formed tissues, tion, nontraumatic limb amputations,
wound infection, delayed healing time, and personal suffering. Addition- and even early death.[1–5] Among various
ally, to meet people’s increasing demand, hydrogel wound dressings with efforts to address this urgent issue, wound
improved performance and multifunctionality are urgently required. Here, dressings are effective strategies to pro-
a new multifunctional supramolecular hydrogel for on-demand dissolvable vide an optimal environment for wound
repair.[6,7] Traditional wound dressings
diabetic foot wound dressings is designed and constructed. Based on multi-
for DFU treatment typically cover rubber,
hydrogen bonds between hydrophilic polymers, the resultant supramolecular electrospun nanofiber, cotton wool, nat-
hydrogels present controlled and excellent properties, such as good transpar- ural or synthetic bandages, and gauzes.[8,9]
ency, antibacterial ability, conductive, and self-healing properties. Thus, the Although these dry dressings are conven-
supramolecular hydrogels improve the new tissue formation and provide a ient to control the initial state of wound
healing, they tend to adhere to the wound
significant therapeutic effect on DFU by inducing angiogenesis, enhancing
area once the absorbed blood and exudate
collagen deposition, preventing bacterial infection, and controlling wound dry out.[7] Besides, the dry dressings still
infection. Remarkably, the resultant hydrogels also exhibit stimuli-responsive suffer from limitations of maintaining a
ability, which renders its capability to be dissolved on-demand, allowing for a moisture environment, allowing gaseous
facile DFU dressing removal. This multifunctional supramolecular hydrogel exchange, and preventing infection.[6,7] In
may provide a novel concept in the design of on-demand dissolvable wound this context, a wound dressing that can
deal with the above shortcomings would
dressings.
be ideal to speed up diabetic foot wounds
healing and improve treatment outcomes.
In recent years, hydrogels have gener-
ated tremendous interest in wound healing applications.[10–16]
Dr. Y. Zhao, Dr. H. Liu, Prof. B. Yang, Prof. Q. Lin As water-based soft materials, hydrogels can facilitate wound
State Key Lab of Supramolecular Structure and Materials, College of
Chemistry healing by absorbing wound exudate, preventing wound desic-
Jilin University cation, and isolating the wound from the environment, which
Changchun 130012, P. R. China makes them the best choice for wound healing.[17,18] However,
E-mail: linquan@jlu.edu.cn the currently available hydrogel dressings still require to be
Dr. Z. Li, Q. Li, Dr. L. Yang, Dr. H. Liu, Prof. J. Wang changed frequently, which is a laborious process and inevitably
Orthopaedic Medical Center
The Second Hospital of Jilin University
cause reinjury of the wounds, wound infection, delayed healing
Changchun 130041, P. R. China time, and personal suffering.[19] To this end, on-demand dis-
E-mail: heliu@jlu.edu.cn; wangjinc@jlu.edu.cn solvable hydrogels represent a new class of emerging “smart”
R. Yan, L. Xiao wound dressings that can be readily operated and painlessly
Jilin Ginseng Academy removed.[19–24] Generally, this type of hydrogels can form in
Changchun University of Chinese Medicine situ and dissolve on-demand via physical crosslinking cases
Changchun 130117, P. R. China
and chemical crosslinking cases. The dissolution of physi-
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/marc.202000441.
cally crosslinked hydrogels is based on physical interactions,
such as molecular entanglements and/or secondary forces
DOI: 10.1002/marc.202000441 (e.g., ionic, H-bonding, and hydrophobic associations).[19,20,22,23]

Macromol. Rapid Commun. 2020, 2000441 2000441  (1 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

These hydrogels possess dynamic networks that can be formed dissolvable wound dressings, allowing for a facile DFU wound
and destroyed in response to environmental stimuli such as a dressing removal. In vivo results suggest that this hydrogel
change in pH, magnetic, temperature, ionic strength of the can effectively induce angiogenesis, enhance collagen deposi-
solution, or light. The dissolution of chemical crosslinked tion, prevent bacterial infection, and control wound infection in
hydrogels is achieved by using cleavable moieties that undergo DFU animal models, which in turn promote wound recovery.
ester hydrolysis or enzymatic degradation. There has also Therefore, the transparent conductive supramolecular hydro-
been growing interest in the preparation of dissolvable chemi- gels can act as an on-demand dissolvable wound dressing for
cally crosslinked hydrogels by incorporation of thiol–disulfide wound repair, which may also have a positive therapeutic effect
exchange, retro-Michael-type, and retro-Diels-Alder reactions, on other types of wounds beyond DFU.
and so on.[21] Despite much progress on on-demand dissolvable
hydrogels have been achieved, the majority of them are involved
with the complicated preparation procedures or the introduction 2. Results and Discussion
of potentially toxic crosslinking agents, which may greatly thwart
the large-scale fabrication of on-demand dissolvable hydrogel 2.1. Fabrication and Characterization of the
dressings. Additionally, the function of developed on-demand PVA-CEC-AGA/Ag Hydrogel
dissolvable hydrogel dressings is too simple that they could meet
people’s increasing demand for wound dressings with improved The synthesis procedures of transparent conductive supramo-
performance and multifunctionality. When the on-demand dis- lecular hydrogel and its multinetwork structures are depicted
solvable hydrogel dressings are applied for practical application, in Scheme 1. To allow proper wound healing, we firstly selected
it is desirable to maintain their basic properties while having hydrogel components that are safe and compatible with the
other enhanced characteristics, such as transparency, antibacte- biological system, and hydrogels with multinetwork structures
rial ability, conductivity, self-healing properties, and so on. were directly fabricated. In brief, the solution of Na2B4O7 was
In this study, we developed a new transparent conductive injected into the mixture solution of PVA, agarose, CEC, and
supramolecular hydrogel based on a multihydrogen bond net- silver nanowires (Ag NWs) (Figure S1, Supporting Information)
work structure, abbreviated as PVA-CEC-AGA/Ag (polyvinyl under vigorously stirring at 90 °C. The transparent conductive
alcohol-N-carboxyethyl chitosan-agarose/Ag). The resultant supramolecular hydrogel was instantly formed. The backbone
supramolecular hydrogel not only possesses good transpar- networks of hydrogels were attributed to the strong hydrogen
ency, antibacterial ability, conductive, and self-healing proper- bonds (hydrogen bond I) between N-carboxyethyl chitosan-aga-
ties, but also exhibit stimuli-responsive ability. The macroscopic rose, N-carboxyethyl chitosan-PVA, and agarose-PVA.[25] These
structure of stimuli-responsive hydrogels can undergo gel-sol strong hydrogen bonds endow hydrogels with stable mechanical
transformation triggered by physical stimulus (temperature or properties. In addition, the reversible networks were also gener-
ultrasound) or chemical stimulus (pH or diol-containing mole- ated due to the dynamic reactions of PVA-borax-PVA and PVA-
cules). In addition, the PVA-CEC-AGA/Ag hydrogels are non- glycerol-PVA (hydrogen bonds II).[26] It is worth noting that the
toxic and have good in vitro cytocompatibility. These properties conductive Ag NWs were uniformly dispersed in the hydrogel
render the supramolecular hydrogel useful for on-demand because of the static hydrogen bond network (hydrogen

Scheme 1.  The synthesis procedures of transparent conductive supramolecular hydrogel and its multinetwork structures.

Macromol. Rapid Commun. 2020, 2000441 2000441  (2 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

bond III) between PVA and the nanowires. The sol-to-gel phase light in the wavelength range 400–700  nm for hydrogels with
transformation of hydrogels is displayed in Figure  1a, named different concentrations of Ag NWs. The hydrogel’s transpar-
PVA-CEC-AGA/Ag. One of the essential requirements for ency could be altered by changing the concentration of Ag
hydrogel wound dressings is their porosity.[27,28] The porosity NWs. The average transmittance per millimeter of thickness of
of wound dressings promises the transport of nutrition and the hydrogel at 660 nm wavelength decreased from 93% to 74%,
gases, thereby enhancing cell migration and proliferation. The when the Ag NWs content increased from 0 to 0.2 mg mL−1. In
porosity and microarchitectural features of hydrogels were addition, when hydrogels were covered on a cardboard, the text
observed by scanning electron microscope (SEM). Overall, of “SKLJLU” on the cardboard was clearly observed indicating
based on multihydrogen bonds between hydrophilic polymers, their high transparency (Figure  2b). This may be due to Ag
the supramolecular hydrogels showed a heterogeneous struc- NWs are well-dispersed in the hydrogel to form light-passible
ture (Figure  1b) with pore diameters ranging from 2.5 to 45 nanoscale networks, which allows light to pass through and
µm (Figure S2, Supporting Information). These porosities on endows hydrogel with high transparency.
hydrogels could enable cellular penetration, which are signifi-
cant to the new tissue formation.
The swelling ability was measured by immersing the PVA- 2.3. Mechanical Strength and Self-Healing Performance of the
CEC-AGA/Ag hydrogels into an excess of phosphate buffered PVA-CEC-AGA/Ag Hydrogel
saline (PBS) (pH  =  7.4) for different periods of time.[19,29,30]
As illustrated in Figure  1c, the hydrogel swelled to 200% in The mechanical properties of PVA-CEC-AGA/Ag could be easily
180  min and 250% in 360  min. After reaching equilibrium, adjusted by varying the individual components (Figure S3,
the hydrogel was stable and retained its integrity. These results Supporting Information). We constructed the resultant supra-
indicated that the swelling could not cause the breakage or dis- molecular hydrogels with crosslinking networks based on mul-
solution of hydrogels. In addition, the hydrogel could contain tihydrogen bonds to provide the tunable mechanical properties.
a large amount of water, which is beneficial to remove wound In addition, the hydrogen bonds between PVA-borax-PVA and
exudate and maintain a moist environment for the wound beds. PVA-glycerol-PVA can be reversibly associated and disassoci-
ated at room temperature, thereby providing good self-healing
properties.[26] To evaluate the self-healing property of hydrogels,
2.2. Transparency of the PVA-CEC-AGA/Ag Hydrogel a strain amplitude sweep test was first performed. As shown in
Figure  3a, in the low strain region, both the storage modulus
Transparency is also an important requirement for hydrogel (G′) and loss modulus (G″) maintained constant values. With
dressings when doctors want to monitor the condition of enhanced strain, G′ and G″ curves intersected at a strain of
wounds and place dressings on the wound area for a long 13.5%, which is the critical strain value indicating the transition
time.[31–34] Figure 2a exhibits the plots of the transmittance of of the gel network to a liquid state. To further increase the

Figure 1.  a) Digital photos of the gelation progress. The mixture of prepared Ag NWs, PVA, and chitosan solution presented in the sol state, and it
experienced sol-gel transition after adding Na2B4O7. b) SEM images of the hydrogel. c) The swelling kinetics of the hydrogel incubated in distilled
water for different times. The hydrogel was prepared with PVA (5 wt%), agarose (0.025 g), CEC (0.025 g), glycerin (0.2 mL), Ag NWs (0.2 mg mL−1),
and Na2B4O7 (0.04 mol L−1, 10 mL).

Macromol. Rapid Commun. 2020, 2000441 2000441  (3 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

Figure 2.  a) Light transmittance spectra of the hydrogel with different concentration of Ag NWs in air. b) Optical images of the hydrogel with different
concentration of Ag NWs. The hydrogel was prepared with PVA (5 wt%), agarose (0.025 g), CEC (0.025 g), glycerin (0.2 mL), Ag NWs (0, 0.05, 0.10,
0.15, 0.20 mg mL−1), and Na2B4O7 (0.04 mol L−1, 10 mL).

strain to 100%, both G′ and G″ decreased dramatically because the self-healing behaviors of hydrogels. The continuous shear
of the collapse of the hydrogel networks. strain was alternated between 200% and 0.01% for 150 s at a
Based on the results of oscillatory shear rheology, the step- fixed angular frequency of 10 rad s−1, respectively. As displayed
strain sweeps were then performed to quantitatively measure in Figure  3b, when the hydrogel was subjected at a higher

Figure 3.  a) Strain-dependent oscillatory shear rheology of the PVA/chitosan/Ag hydrogel hydrogels at a fixed angular frequency (10 rad s−1). b) Using
alternating low (1%)–high (500%) shear strain to measure the self-healing property of PVA/chitosan/Ag hydrogel. Each strain interval was kept as
150 s and the fixed frequency was kept as 10 rad s−1. c) The hydrogels were cut into two pieces and then healed under ambient conditions. Rhodamine
B and methylene blue were present in the hydrogel for visualization. d) Macroscopic photographs of the restored electrical conductivity by a healed
hydrogel connected to purple LED bulbs. Methylene orange and blue were present in the hydrogel for visualization. The hydrogel was prepared with
PVA (10 wt%), agarose (0.025 g), CEC (0.025 g), glycerin (0.2 mL), Ag NWs (0.20 mg mL−1), and Na2B4O7 (0.04 mol L−1, 10 mL).

Macromol. Rapid Commun. 2020, 2000441 2000441  (4 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

strain of 200%, the gel failure occurred where G″ was larger at room temperature. After 2 h, we found that the hydrogels
than G′. Subsequently, when the hydrogel was applied with a healed completed with an indistinguishable interface, and the
lower strain of 0.01%, the G′ and G″ values were instantane- LED bulbs could be lighted up again.
ously fully restored to their original values even after repeated
multiple times. The mechanical healing ability of hydrogels was
also demonstrated by the macroscopic observation. According 2.4. Stimuli-Responsive and On-Demand Dissolvable Abilities of
to Figure  3c, the hydrogels with different colors were physi- the PVA-CEC-AGA/Ag Hydrogel
cally cut into two halves of the same size. Then, the different
parts of the hydrogel were put together. After 5  min, the two Intelligent soft materials have attracted increasing attention
halves of the hydrogels could be automatically reconstructed, during the past decades whose macroscopic structure and
and they could maintain integrity. Apart from the mechanical physical properties show a significant change in response to
self-healing property, the electrical properties of the hydrogel specific stimuli.[35–39] With the reversible linkages in hydrogels
could also be restored. According to Figure 3d, when the hydro- demonstrated above, the prepared hydrogels are promising as
gels were cut into two parts, the LED bulbs were extinguished. stimuli-responsive macroscopic structures. As illustrated in
Afterward, the separated two parts were brought into contact Figure 4a, the hydrogel network could be completely destroyed

Figure 4.  a) The gel-sol phase transitions of the hydrogel triggered by different stimuli, such as fructose, glucose, dopamine, and Vitamin B6 and
HCl (0.1 m). b) Schematic of hydrogel’s multiresponses to different stimuli. c) Photographs of the on-demand dissolution of hydrogel after treatment
with a glucose solution (0.1 m, pH = 7.0). The hydrogel was prepared with PVA (10 wt%), agarose (0.025 g), CEC (0.025 g), glycerin (0.2 mL), Ag NWs
(0.2 mg mL−1), and Na2B4O7 (0.04 mol L−1, 10 mL).

Macromol. Rapid Commun. 2020, 2000441 2000441  (5 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

Figure 5.  a) Proliferation of L929 cells cultured for 1, 4, and 7 d (n = 3). b) Live/Dead staining of L929 cells for 3 d. The hydrogel was prepared with PVA
(5, 7.5, 10, 12.5 wt%), agarose (0.025 g), CEC (0.025 g), glycerin (0.2 mL), Ag NWs (0.2 mg mL−1), and Na2B4O7 (0.04 mol L−1, 10 mL).

by increasing the temperature or applying ultrasound. In addi- AGA/Ag hydrogel to be dissolved on-demand, which is prom-
tion to physical stimulation, reducing solution pH or adding ising as a desirable alternative to debridement of the dressing.
competitive diol-containing molecules (such as fructose, glu-
cose, dopamine, and vitamin B6) could also quickly trigger the
breakdown of the hydrogel. This stimuli-responsive property 2.5. Biocompatibility Evaluation of the PVA-CEC-AGA/Ag
was mainly due to the reversible nature of boronate formation Hydrogel
between Na2B4O7 and PVA. More specifically, the equilibrium
of boronate-based reactions could be shifted by heating, ultra- The in vitro cytotoxicity test (Cell Counting Kit 8 (CCK-8) assay)
sound, reducing pH, and diol-containing molecules. The diol was carried out by using the fibroblast (L929) cell line, and the
groups in free molecules are more reactive than PVA polymer, corresponding results are illustrated in Figure  5a. The control
which can act as a competitor of PVA to react with the Na2B4O7 cells were incubated in a culture dish, and other hydrogel group
(Figure  4b).[40] Therefore, the PVA-CEC-AGA/Ag hydrogels cells were incubated with different components of hydrogel
break down and dissolve in the presence of molecules con- extract solutions. The L929 cells were proliferating with the pro-
taining diol groups (such as fructose, glucose, dopamine, and longation of culture time, and there was no significant differ-
vitamin B6). ence between each hydrogel group and the control group. The
Usually, traditional available dressings will adhere to the CCK-8 assay results clearly demonstrated that the PVA-CEC-
wound surface, requiring surgical and mechanical removal AGA/Ag hydrogel had good biocompatibility. In addition, Live/
from the wound. The repeated dressing changes not only cause Dead staining revealed that L929 cells maintained excellent
additional trauma to the newly formed tissues but also bring viability whether in the hydrogel group or the control group
personal suffering in the injured patient.[21–23] Thus, hydrogels (Figure 5b). Thus, PVA-CEC-AGA/Ag hydrogels exhibited good
with on-demand dissolvable property are particularly appealing in vitro cytocompatibility, which have great potential for use in
to improve treatments and reduce patient pain. It is anticipated biomedical applications.
the PVA-CEC-AGA/Ag hydrogels have the capability to be dis-
solved on-demand via the reversible nature of boronate forma-
tion. The on-demand dissolution of the developed hydrogels 2.6. PVA-CEC-AGA/Ag Hydrogel for Diabetic Foot Wound
by glucose was investigated. As displayed in Figure  4ci,ii, the Dressing
PVA-CEC-AGA/Ag hydrogels were coated in an artificial wound
on a diabetic rat. After a glucose-soaked gauze was applied to DFUs are always regarded as one of the most serious compli-
half of the hydrogel for about 20 min, the covered hydrogel cations of diabetes associated with amputation. The impaired
part dissolution occurred (Figure 4ciii,iv). If the glucose-soaked DFUs typically always have features of extensive size, irregular
gauze was administered to the whole hydrogel, the hydrogel shape, difficult-to-access areas, massive wound exudate, etc.[7]
completely dissolved as time elapsed (Figure  4cv–viii). The Based on this situation, the development of benign and efficient
on-demand dissolvable capability of the hydrogels was further diabetic foot wound dressings is urgent to be explored. Com-
quantitatively analyzed by recording G′ and G″ in time sweep pared with other traditional wound dressings, hydrogel-based
mode (Figure S4, Supporting Information). Obviously, the wound dressing has more ideal properties including removal
hydrogel remained stable when in contact with air. In contrast, of wound exudate, providing a moist wound environment, pre-
when in contact with glucose (0.1 m, pH  =  7.0), the G′ and G″ vention of secondary infections, and promotion of cell adhesion
of the hydrogel decreased and G″ became larger than G′ indi- and proliferation.[11,13,16] In addition to the above advantages, our
cating that the hydrogel experienced a sol-gel transformation. designed PVA-CEC-AGA/Ag hydrogels can be easily detached
Thus, these results demonstrated the ability of the PVA-CEC- from the wound by an on-demand dissolvable property. This

Macromol. Rapid Commun. 2020, 2000441 2000441  (6 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

nonharmful change characteristic can create an optimal was suppurative wound surface in the control group. In addi-
environment for promoting diabetic foot wound healing. To tion, the quantitative analysis showed that the healing rate of
examine the PVA-CEC-AGA/Ag hydrogel treatment of infec- the wounds treated with hydrogels was significantly faster than
tive diabetic foot wounds, we established diabetic rat models that of the control group, especially at the late stage of treat-
by injecting streptozotocin (STZ). In order to observe the rate ment (10, 15, and 20 d post-treatment) (p < 0.01) (Figure 6b). At
of wound healing, the general wound closure results were 20 d post-treatment, the wounds covered with hydrogels were
recorded and the residual wound area calculated. According almost fully healed, whereas about 35.73  ±  7.33% of unhealed
to Figure 6a, the wounds treated with hydrogel showed a rapid wound areas had remained in the control group. During the
contraction and healed in a well-organized manner, while there whole experiment, the blood glucose level of rats was also

Figure 6.  a) Representative photos of the diabetic foots after treated with PBS and hydrogel. b) Quantification of wound residual area. c) Hematoxylin
and eosin (H&E) staining of wound tissues around foots. d) Quantification of inflammatory cells. e) Masson trichrome staining of wound tissues
around foots. f) Quantitative analysis of the percentage of collagen deposition. g) Neovascularization in granulation tissue was assessed with CD31
immunohistochemical staining. h) Quantification of blood vessels. i) Giemsa staining for bacteria in wound tissue (*P < 0.05, **P < 0.01, n = 3). The
hydrogel was prepared with PVA (10 wt%), agarose (0.025 g), CEC (0.025 g), glycerin (0.2 mL), Ag NWs (0.2 mg mL−1), and Na2B4O7 (0.04 mol L−1,
10 mL).

Macromol. Rapid Commun. 2020, 2000441 2000441  (7 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

measured, and there was a drop in the blood glucose level after increased on the 5th and 15th day, which were 31.47 ± 2.87 and
the wounds were treated with PVA-CEC-AGA/Ag hydrogel, 33.4  ±  3.85  ×  109  L−1, respectively. This result further demon-
especially in the first 5 d after treatment (p < 0.05) (Figure S5, strates that this effective sterilization dressing can avoid sys-
Supporting Information). temic infection, and remarkably accelerate wound closure and
To further distinguish the quality of regenerated skin in enhance the quality of the healed wound as described above.
the diabetic foot wounds, histological analysis was performed.
Consistent with the optical image results, H&E staining
(Figure  6c) revealed that wounds treated by PVA-CEC-AGA/ 3. Conclusions
Ag hydrogel displayed well-organized granulation tissue for-
mation on the 5th day. On the 15th day, more granulation tis- In summary, we have designed a new transparent conductive
sues were observed in the hydrogel group. In contrast, necrotic supramolecular hydrogel through multihydrogen bond net-
tissue, foreign bodies, less granulation tissues, and excessive works using biocompatible macromonomers. The developed
inflammatory cells were formed in the control group during hydrogels exhibit many desirable features including tunable
the whole healing process. Additionally, the aggregation and mechanical, good transparency, antibacterial ability, conduc-
activation of appropriate inflammatory cells are essential to the tive, and self-healing properties. It is worth mentioning that
transition from inflammation to repair phases, while excessive the resultant hydrogels can dissolve on-demand via the revers-
inflammatory cells impair wound repair. In the control group, ible nature of boronate formation, which renders significant
a large number of inflammatory cells were observed at the promise for a more effective treatment for DFU patients. In
wound site on the 5th day, and the inflammation was still vitro results demonstrate that the resultant hydrogels are bio-
serious on the 15th day. Compared with the control group, the compatible. In vivo DFU animal experiments result further
wounds covered with hydrogel showed a mild inflammatory demonstrates that this hydrogel dressing can induce angio-
reaction (p < 0.05), which maintained an appropriate inflamma- genesis, enhance collagen deposition, avoid systemic infection,
tory environment and did not cause a foreign body response remarkably accelerate wound closure, and enhance the quality
(Figure  6d). Collagen deposition is another critical factor for of the healed wound. This work provides a unique approach to
wound contraction and scar formation.[41] Masson’s trichrome design and construct multifunctional hydrogels with controlled
(MT) staining was performed to detect the newborn collagen dissolution ability.
deposition in the wound (Figure  6e,f). On the 5th day, almost
no collagen deposition was observed in the control group, while
limited collagen deposition occurred in the hydrogel group. On 4. Experimental Section
the 15th day, the collagen deposition of the hydrogel group was
Materials: Silver nitrate (AgNO3, ≈99.8%) was purchased from
further increased and extended to the central of wounds with Sinopharm Chemical Reagent Co., Ltd. (Shanghai, P. R. China).
regular arrangement, which was 1.75-fold higher than that of Hydrochloric acid (HCl, 36–38%), sodium hydroxide (NaOH, ≈99.0%),
the control group. ethanol (C2H5OH, ≈99.7%), ethylene glycol (≈99%), and sodium
It is well-known that some crucial substances (e.g., growth tetraborate (Na2B4O7·10H2O) were purchased from Beijing Chemical
factors, oxygen, nutrients, etc.) are delivered and infiltrated into Works (Beijing, P. R. China). Polyvinyl pyrrolidone (PVPON, Mw ≈
the wounded tissue via blood vessels.[5] As a result, neovascu- 40 000), PVA (≈99% hydrolyzed, Mw ≈ 130 000), dopamine hydrochloride
(DA, ≈98%), STZ, glucose, and fructose were obtained from
larization plays a critical role in the tissue remodeling process, Sigma-Aldrich. Chitosan (degree of deacetylation ≈ 95%, 100–200 mps)
and wounds may fail to heal due to inadequate local vascular and vitamin B6 hydrochloride were purchased from Aladdin. Acrylic
supply. As shown in CD31 immunohistochemical staining, acid (≈99%) and paraformaldehyde (≈99%) were from Xilong Chemical
more neovascularization (green arrows) was generated after Co., Ltd. (Guangzhou, P. R. China). Copper chloride (CuCl2·2H2O)
treated with hydrogel, which was 4.76-fold and 2.2-fold higher was from Tianjin Yongsheng Fine Chemical Co., Ltd. (Tianjin, P. R.
than the control group on the 5th and 15th days post-treatment China). Dulbecco’s modified Eagle’s medium (DMEM, low glucose)
was purchased from HyClone (Beijing, P. R. China). Fetal bovine
(p < 0.05), respectively (Figure 6g,h).
serum (FBS) and streptomycin-penicillin were purchased from Gibco
Diabetic wounds are easily infected by bacteria and facili- Life Technologies (USA). CCK-8 was obtained from Dojindo Molecular
tated bacterial growth because of local high blood glucose Technology (Japan). Calcein-AM/PI was obtained from Biobetimes
levels and exposure to air. An infectious environment exerts an Biotechnology Co., Ltd. (Changsha, P. R. China). Hematoxylin and
obvious negative impact on wound healing, even causing bac- eosin, Masson’s trichrome (MT), and Giemsa stain were purchased
teremia.[41] Giemsa staining was used to investigate bacterial from Thermo Fisher Scientific Co., Ltd. (Shanghai, P. R. China). Triton
X-100 was purchased from Dingguo Changsheng Biotechnology
contamination on the wound surface. As shown in Figure  6i,
Co. Ltd. (Beijing, P. R. China). Antibody CD31 (ab28364) used for
the wounds in the control group had a large number of bac- immunohistochemistry was purchased from Abcam (UK). The ultrapure
teria on the 5th and 15th days. However, in the hydrogel group, water used throughout the experiments was purified with a Milli-Q A10
owing to the antibacterial properties of Ag NWs, it was difficult filtration system (Millipore, Billerica, MA). The other chemicals were
to find colonizing bacteria (Figure S6, Supporting Information). used as purchased without further purification.
Furthermore, to evaluate the spread of bacteria in the whole Characterization: Ultraviolet–visible (UV–vis) absorption spectra were
body, the number of white blood cells (WBC) in blood, an indi- recorded at room temperature on a Lambad 800 spectrophotometer
using a quartz cell of 1  cm path length. Field-emission SEM images
cator of systemic inflammation, was also monitored (Figure were obtained with JEOL JSM-6700F SEM operated at an acceleration
S7, Supporting Information). The WBC counts in the hydrogel voltage of 3  kV. The dynamic mechanical properties of hydrogels were
group were within the normal range (5.0–25.0  ×  109 L−1). performed on a rheometer (TA Instruments-waters LLC) at room
However, the counts in the control group were significantly temperature. The optical density of cells was measured at 450 nm by a

Macromol. Rapid Commun. 2020, 2000441 2000441  (8 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

microplate reader (Multiskan EX, Thermo Fisher Scientific Inc., Shanghai, selected to establish diabetic wound models. After anesthesia with
P. R. China). Fluorescent photographs cell and histological staining intraperitoneal 1% pentobarbital sodium (100  mg kg−1), rat skin was
images were obtained from a fluorescence microscopy (IX51, Olympus, cleaned with 0.3% iodine. Then a 5 mm diameter full-thickness foot skin
Japan). The blood glucose level was measured by a glucose meter wound was made by punch biopsy instruments. The wound was washed
(Johnson, P. R. China). All histological staining images were analyzed by with saline, and implanted with Staphylococcus aureus and Escherichia
Image Pro-Plus (IPP) 6.0 (National Institutes of Health, Bethesda, MD). coli (1 × 108 CFUs in 10 × 10−6 m of PBS), respectively. For comparison,
All photographs were taken by a digital camera (Canon camera). the wound covered with 200 µL hydrogel was regarded as an experiment
Preparation of Hydrogels: The hydrogels were synthesized by group, while the wound just treated with PBS was regarded as a control
the following procedures. 1) Preparation of Ag NWs: Ag NPs were group. All the wounds were fixed with sterile gauze and fixed with an
fabricated according to literature procedures.[42–45] Briefly, anhydrous elastic adhesive bandage. The rats were individually housed in a cage for
ethylene glycol (5 mL) was heated to 151.5 °C under magnetic stirring observation. At the 0, 5, 10, 15, and 20 d postoperation, the wounds were
(260  rpm). At 1 h, the solution of CuCl2 dissolved in ethylene glycol recorded using a digital camera and quantitatively analyzed by Image
(4  mmol L−1, 0.9  mL) was injected into the heated ethylene glycol. Pro Plus 6.0. The residual wound area was calculated by the following
After 15  min, the solution of PVPON dissolved in ethylene glycol formula: Residual wound area (%) = Sn/S0  × 100% (S0: initial wound
(0.1 g, 2.5 mL) was added, followed by injection of the ethylene glycol area; Sn: wound area at different time points).[41]
solution of AgNO3 (0.05  g, 2.5  mL). The above solution was then Histopathological Evaluation: After 5 and 15 d of treatment, the
heated for another 30  min, and Ag NWs were synthesized. Products wound and surrounding tissues were collected. All the skin samples
were then washed with ethanol once and water three times. They were were fixed with 10% paraformaldehyde solution, and then embedded
stored in water before use. 2) Preparation of N-carboxyethyl chitosan: in paraffin for routine histological processing. On the basis of the
CEC was fabricated following previous reported procedures. Chitosan standard protocols, 5  mm thickness sections of the samples were
(2 g, 12.4 mmol) and acrylic acid (2.9 mL, 42.6 mmol) were dissolved prepared. In this experiment, H&E and MT staining were carried out to
in deionized water (100 mL), and then heated at 50 °C under magnetic assess the inflammation, morphology, tissue regeneration, and collagen
stirring for 72 h. Afterward, the above solution was cooled down, deposition. CD31 immunohistochemistry was applied to observe the
whose pH was adjusted to 10–12 by injecting 1 m NaOH. To remove neovascularization in regenerated granulation tissue, while Giemsa
by-products and other impurities, the solution was dialyzed (MWCO staining was used to investigate the bacterial colonization in wound
14  000) against deionized water for 3 d. After repeated change of tissues.[19,47,48]
water, the product was freeze-dried. 3) Preparation of hydrogels: In Statistical Analysis: The data were expressed as means ± standard
this experiment, PVA (0.5  g), agarose (0.025  g), CEC (0.025  g), and deviation (SD) from at least three independent experiments, and the
glycerin (0.2  mL) were dissolved in deionized water (10  mL), and statistical analysis was carried out using ANOVA with Tukey’s posthoc
this mixture was oil-bathed at 90  °C under magnetic stirring for 2  h. analysis (SPSS Inc., Chicago, IL). P < 0.05 was considered a statistically
Then different concentrations of Ag NWs (0, 0.05, 0.10, 0.15, and significant difference.
0.2  mg  mL−1) were added into the above solution, respectively. After
stirred for another 15 min, Na2B4O7 (0.04 mol L−1, 10 mL) was injected
into the above mixture, and further stirred at 90 °C for 10 min until a
hybrid hydrogel formed. Supporting Information
Biocompatibility of Hydrogels In Vitro: The hydrogel samples were cut
Supporting Information is available from the Wiley Online Library or
into thin slices placed on six-well plates. The prepared disc samples
from the author.
were added with DMEM containing 10% FBS, 1% penicillin and
streptomycin, and then incubated at 37  °C in a humidified atmosphere
containing CO2 (5%) for 3 d. The hydrogel extracts were subsequently
used for the following in vitro cell experiments. The proliferation of L929
cells on hydrogel extracts was measured by CCK-8. Briefly, L929 cells
Acknowledgements
at a density of 5 × 104 each well were cultured with hydrogel extracts. Y.Z. and Z.L. contributed equally to this work. This work was supported
For comparison, the control groups were cultured with DMEM. After by the National Natural Science Foundation of China (Grant Nos.
incubation for 1, 4, and 7 d, CCK-8 solution with a 10% volume of the 51861145311, 21174048, 81671804, and 81772456). J.W. and H.L. are thankful
medium was added into each well after replacing medium and then were to the Science and Technology Development Program of Jilin Province
incubated at 37 °C in a humidified atmosphere containing CO2 (5%) for (Grant Nos. 20190304123YY, 20180623050TC, 20180201041SF, and
2 h. After that, the above reaction solution (100 µL) was transferred into 20170204004GX), the Cultivation Program from the Second Hospital of Jilin
a new 96-well plate, and the optical density was measured at 450 nm by University for National Natural Science Foundation (KYPY2018-01), and the
a microplate reader.[10,13] Youth Talents Promotion Project of Jilin Province (Grant No. 192004). The
To evaluate the cell viability of L929 cultured in the hydrogel extracts, authors thank the cooperation with Prof. Hongdong Li by Open Project of
the Live/Dead assay was performed. 3 d after seeding, samples were State Key Laboratory of Super hard Materials (Jilin University).
incubated with 1  × 10−3 m calcein-AM for 1 h, then incubated with
1  μg  mL−1 propidium iodide (PI) for 5  min at 37  °C. The samples were
then imaged by fluorescence microscopy.[46]
Diabetic Rat Model Establishment and Wound Healing Examination: Conflict of Interest
Animal experiments were conducted according to the National Institutes
The authors declare no conflict of interest.
of Health’s Guide for the Care and Use of Laboratory Animals (NIH
Publications No. 8023, revised 1978). All experimental protocols were
approved and performed in accordance with the guidelines of the Animal
Ethics Committee of Jilin University. Adult male Sprague-Dawley (SD) Keywords
rats (200–250 g, 8–10 weeks) were obtained from the Animal Center of
Jilin University. The diabetes rats model was prepared by intraperitoneal dissolution, self-healing hydrogels, silver nanowires, stimuli-responsive
injection of STZ (55 mg kg−1). After one week of STZ administration, the hydrogels, wound dressing
blood glucose level was measured by a glucose meter from the tail vein.
Rats were diabetic when their blood glucose level exceeded 16.7  mmol Received: August 12, 2020
L−1. Blood glucose levels of rats were monitored throughout the study. Revised: September 3, 2020
Following by STZ-induced hyperglycemia for three weeks, 40 rats were Published online:

Macromol. Rapid Commun. 2020, 2000441 2000441  (9 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

[1] R. Barnett, Lancet 2019, 394, 557. [25] M. Li, W. Li, W. Cai, X. Zhang, Z. Wang, J. Street, W.-J. Ong, Z. Xia,
[2] R. Garg, Lancet 2019, 394, 1320. Q. Xu, Mater. Horiz. 2019, 6, 703.
[3] S. A.  Eming, B.  Brachvogel, T.  Odorisio, M.  Koch, J. Histochem. [26] M. Liao, P. Wan, J. Wen, M. Gong, X. Wu, Y. Wang, R. Shi, L. Zhang,
Cytochem. 2007, 42, 115. Adv. Funct. Mater. 2017, 27, 1703852.
[4] V. Falanga, Lancet 2005, 366, 1736. [27] A. K. M. B. Khoda, B. Koc, J. Med. Device 2012, 6, 031003.
[5] H. Wu, F. Li, W. Shao, J. Gao, D. Ling, ACS Cent. Sci. 2019, 5, 477. [28] X. Zhang, C. Huang, Y. Zhao, X. Jin, RSC Adv. 2017, 7, 39349.
[6] P. T.  Kumar, V. K.  Lakshmanan, T. V.  Anilkumar, C.  Ramya, [29] Y. N. Zhang, R. K. Avery, Q. Vallmajo-Martin, A. Assmann, A. Vegh,
P. Reshmi, A. G. Unnikrishnan, S. V. Nair, R. Jayakumar, ACS Appl. A. Memic, B. D. Olsen, N. Annabi, A. Khademhosseini, Adv. Funct.
Mater. Interfaces 2012, 4, 2618. Mater. 2015, 25, 4814.
[7] L. I. Moura, A. M. Dias, E. Carvalho, H. C. de Sousa, Acta Biomater. [30] J.  Shin, J. S.  Lee, C.  Lee, H.-J.  Park, K.  Yang, Y.  Jin, J. H.  Ryu,
2013, 9, 7093. K. S.  Hong, S.-H.  Moon, H.-M.  Chung, H. S.  Yang, S. H.  Um,
[8] H. Liu, C. Wang, C. Li, Y. Qin, Z. Wang, F. Yang, Z. Li, J. Wang, RSC J.-W. Oh, D.-I. Kim, H. Lee, S.-W. Cho, Adv. Funct. Mater. 2015, 25,
Adv. 2018, 8, 7533. 3814.
[9] P.  Mostafalu, A.  Tamayol, R.  Rahimi, M.  Ochoa, A.  Khalilpour, [31] J. Wang, J. Wei, Mater. Sci. Eng., C 2017, 80, 460.
G.  Kiaee, I. K.  Yazdi, S.  Bagherifard, M. R.  Dokmeci, B.  Ziaie, [32] W. Chen, P. Zhang, R. Zang, J. Fan, S. Wang, B. Wang, J. Meng, Adv.
S. R. Sonkusale, A. Khademhosseini, Small 2018, e1703509. Mater. 2020, 32, 1907413.
[10] G. Chen, Y. Yu, X. Wu, G. Wang, J. Ren, Y. Zhao, Adv. Funct. Mater. [33] Z.  Jia, Y.  Zeng, P.  Tang, D.  Gan, W.  Xing, Y.  Hou, K.  Wang, C.  Xie,
2018, 28, 1801386. X. Lu, Chem. Mater. 2019, 31, 5625.
[11] S. H. Kim, S. H. Lee, J. E. Lee, S. J. Park, K. Kim, I. S. Kim, Y. S. Lee, [34] K.  Parida, V.  Kumar, W.  Jiangxin, V.  Bhavanasi, R.  Bendi, P. S.  Lee,
N. S. Hwang, B. G. Kim, Biomaterials 2018, 178, 401. Adv. Mater. 2017, 29, 1702181.
[12] S. Li, S. Dong, W. Xu, S. Tu, L. Yan, C. Zhao, J. Ding, X. Chen, Adv. [35] H. Chen, X. Ma, S. Wu, H. Tian, Angew. Chem., Int. Ed. 2014, 53, 14149.
Sci. 2018, 5, 1700527. [36] S.  Lamping, T.  Otremba, B. J.  Ravoo, Angew. Chem., Int. Ed. 2018,
[13] Y.  Liang, X.  Zhao, T.  Hu, B.  Chen, Z.  Yin, P. X.  Ma, B.  Guo, Small 57, 2474.
2019, 15, 1900046. [37] W. Li, K. Dong, J. Ren, X. Qu, Angew. Chem., Int. Ed. 2016, 55, 8049.
[14] Y.  Liang, X.  Zhao, T.  Hu, Y.  Han, B.  Guo, J. Colloid Interface Sci. [38] M. E.  Roth-Konforti, M.  Comune, M.  Halperin-Sternfeld,
2019, 556, 514. I.  Grigoriants, D.  Shabat, L.  Adler-Abramovich, Macromol. Rapid
[15] J.  Qu, X.  Zhao, Y.  Liang, T.  Zhang, P. X.  Ma, B.  Guo, Biomaterials Commun. 2018, 39, e1800588.
2018, 183, 185. [39] S. Zhou, B. Wu, Q. Zhou, Y. Jian, X. Le, H. Lu, D. Zhang, J. Zhang,
[16] R. Wang, J. Li, W. Chen, T. Xu, S. Yun, Z. Xu, Z. Xu, T. Sato, B. Chi, Z. Zhang, T. Chen, Macromol. Rapid Commun. 2020, 41, 1900543.
H. Xu, Adv. Funct. Mater. 2017, 27, 1604894. [40] Y.  Chen, D.  Diaz-Dussan, D.  Wu, W.  Wang, Y.-Y.  Peng, A. B.  Asha,
[17] Y.  Hu, Z.  Zhang, Y.  Li, X.  Ding, D.  Li, C.  Shen, F. J.  Xu, Macromol. D. G. Hall, K. Ishihara, R. Narain, ACS Macro Lett. 2018, 7, 904.
Rapid Commun. 2018, 39, e1800069. [41] Y. Zhao, Z. Li, S. Song, K. Yang, H. Liu, Z. Yang, J. Wang, B. Yang,
[18] M. W.  Thielke, C.  Secker, H.  Schlaad, P.  Theato, Macromol. Rapid Q. Lin, Adv. Funct. Mater. 2019, 29, 1901474.
Commun. 2016, 37, 100. [42] P. C.  Hsu, X.  Liu, C.  Liu, X.  Xie, H. R.  Lee, A. J.  Welch, T.  Zhao,
[19] W.  Huang, Y.  Wang, Z.  Huang, X.  Wang, L.  Chen, Y.  Zhang, Y. Cui, Nano Lett. 2015, 15, 365.
L. Zhang, ACS Appl. Mater. Interfaces 2018, 10, 41076. [43] P.  Song, H.  Qin, H. L.  Gao, H. P.  Cong, S. H.  Yu, Nat. Commun.
[20] M.  Konieczynska, J.  Villa-Camacho, C.  Ghobril, M.  Perez-Viloria, 2018, 9, 2786.
K.  Tevis, W.  Blessing, A.  Nazarian, E.  Rodriguez, M.  Grinstaff, [44] J. Wang, F. Tang, Y. Wang, Q. Lu, S. Liu, L. Li, ACS Appl. Mater. Inter-
Angew. Chem., Int. Ed. 2016, 55, 9984. faces 2020, 12, 1558.
[21] M. D. Konieczynska, M. W. Grinstaff, Acc. Chem. Res. 2017, 50, 151. [45] F. Xu, Y. Zhu, Adv. Mater. 2012, 24, 5117.
[22] H. Lu, L. Yuan, X. Yu, C. Wu, D. He, J. Deng, Burns Trauma 2018, 6, 35. [46] D. Gan, T. Xu, W. Xing, X. Ge, L. Fang, K. Wang, F. Ren, X. Lu, Adv.
[23] J.  Valdez, C. D.  Cook, C. C.  Ahrens, A. J.  Wang, A.  Brown, Funct. Mater. 2019, 29, 1805964.
M.  Kumar, L.  Stockdale, D.  Rothenberg, K.  Renggli, E.  Gordon, [47] Y. Li, L. Yang, Y. Zeng, Y. Wu, Y. Wei, L. Tao, Chem. Mater. 2019, 31,
D. Lauffenburger, F. White, L. Griffith, Biomaterials 2017, 130, 90. 5576.
[24] X. Zhang, D. Wasserberg, C. Breukers, L. Terstappen, M. Beck, ACS [48] H.  Wang, H.  Zhu, W.  Fu, Y.  Zhang, B.  Xu, F.  Gao, Z.  Cao, W.  Liu,
Appl. Mater. Interfaces 2016, 8, 27539. Macromol. Rapid Commun. 2017, 38, 1600695.

Yue Zhao received her B.S. degree in polymeric materials and engineering at the College of
Chemistry, Jilin University in 2015. She is currently a Ph.D. student under the supervision of
Prof. Quan Lin at State Key Laboratory of Supramolecular Structure and Materials, Jilin University.
Her research is centered on design, preparation, and biological application of biomimetic
intelligent polymer hydrogel materials.

Macromol. Rapid Commun. 2020, 2000441 2000441  (10 of 11) © 2020 Wiley-VCH GmbH
www.advancedsciencenews.com www.mrc-journal.de

Zuhao Li’s, main research focus on two main directions: hydrogels as wound dressing to enhance
diabetic wound healing, as well as 3D-printed metal scaffolds to promote bone integration in
various orthopedic related diseases.

He Liu is an attending physician at the Orthopedic Medical Center of The Second Hospital of Jilin
University. His main research direction is the application of hydrogel in different disease models.

Jincheng Wang is a professor, doctoral supervisor, and the Director of Orthopedic Medical
Center of The Second Hospital of Jilin University. His research interest is focused on the clinical
application of 3D printing technology.

Quan Lin received his bachelor’s degree in polymer materials and engineering and master’s
degree in polymer chemistry and physics from JLU in 1991 and 1994, respectively. He started his
professional experience at the College of Chemistry, JLU from 1994, and he graduated with a Ph.D.
in polymer chemistry and physics from JLU in 2000. He joined Carleton University as a postdoc-
toral researcher during 2001–2003. His research interests focus on multifunctional nanomaterials
based on supramolecular chemistry for applications in biosensing, responsive, and biomimetic
polymeric hydrogels and applications in biological systems.

Macromol. Rapid Commun. 2020, 2000441 2000441  (11 of 11) © 2020 Wiley-VCH GmbH

You might also like