You are on page 1of 13

Chemical Engineering Journal 385 (2020) 123464

Contents lists available at ScienceDirect

Chemical Engineering Journal


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

Anti-oxidant electroactive and antibacterial nanofibrous wound dressings T


based on poly(ε-caprolactone)/quaternized chitosan-graft-polyaniline for
full-thickness skin wound healing

Jiahui Hea, Yongping Lianga, Mengting Shia, Baolin Guoa,b,
a
Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University, Xi’an 710049, China
b
Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an 710049, China

H I GH L IG H T S G R A P H I C A L A B S T R A C T

• Nanofibrous wound dressing with


comparable mechanical property to
soft tissue was designed.
• The dressing showed good electro-
activity and anti-oxidant ability.
• The dressing exhibited good bio-
compatibility and effective anti-
bacterial activity.
• the
The dressing significantly accelerated
healing process of full-thickness
skin wound.

A R T I C LE I N FO A B S T R A C T

Keywords: Developing a novel nanofibrous wound dressing with multi-functional properties, integrating suitable me-
Electroactive nanofiber chanical property, electroactivity, anti-oxidant and inherent antibacterial activity to promote wound healing
Electrospinning process is urgently desired, which could meet the increasing requirements of clinical needs. Herein, a series of
Anti-oxidant property antibacterial, anti-oxidant and electroactive nanofibrous membranes were fabricated by electrospinning poly(ε-
Antibacterial activity
caprolactone) (PCL) and quaternized chitosan-graft-polyaniline (QCSP) polymer solutions which combined the
Wound dressing
good mechanical property of PCL and multi-functionality of QCSP. The nanofibrous wound dressings exhibited
electroactivity, similar mechanical properties to soft tissue, free radical scavenging capacity, antibacterial
property, and biocompatibility. In particular, PCL/QCSP15 (15 wt% of QCSP in the sample) showed a good
balanced ability between antibacterial activity and cell proliferation, which significantly accelerated the healing
process of wound in a mouse full-thickness wounds defect model than commercial dressing (Tegaderm™ Film)
and pure PCL (PCL/QCSP0) nanofibrous membrane. Moreover, the histopathological examination and im-
munofluorescence staining showed that the wounds treated by PCL/QCSP15 nanofiber dressing exhibited higher
collagen deposition, granulation tissue thickness, and more angiogenesis. In one word, these antibacterial, anti-
oxidant, electroactivity nanofibrous membranes showed promising applications for full-thickness skin repair.


Corresponding author at: Frontier Institute of Science and Technology, and State Key Laboratory for Mechanical Behavior of Materials, Xi’an Jiaotong University,
Xi’an 710049, China.
E-mail address: baoling@mail.xjtu.edu.cn (B. Guo).

https://doi.org/10.1016/j.cej.2019.123464
Received 29 July 2019; Received in revised form 23 October 2019; Accepted 11 November 2019
Available online 12 November 2019
1385-8947/ © 2019 Elsevier B.V. All rights reserved.
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

1. Introduction electroactivity, and antioxidant ability were systematically character-


ized. Moreover, the antimicrobial activity of these nanofiber wound
The largest organ of human body is skin tissue, which acts as the dressings was assessed by performing an in vitro surface antibacterial
first barrier to the external environment against dehydration, chemical/ experiment. The blood compatibility and cytocompatibility of PCL/
radiological damages, and microorganisms invasion [1–3]. For those QCSP NFMs were evaluated by co-culturing with red blood cells (RBC)
damaged or destroyed skin tissue, the wound repair process appears and mouse fibroblasts (L929), respectively. Furthermore, quantitative
very complicated and needs highly orchestrated, which not only in- statistical analysis of wound contraction, histopathological examina-
volves the complex coordination between dermal and epidermal com- tions, collagen content, and immunofluorescence staining was em-
partments, but also between local microenvironment, resident cells, ployed to investigate the therapeutic efficacy of PCL/QCSP NFM wound
and immigrant cells [4–6]. Moreover, the lost tissue will be replaced by dressings. All the results demonstrated that these electroactive anti-
scar tissue when the wounds affect the dermis because the dermis lacks bacterial NFM wound dressings with suitable mechanical properties
the regenerative ability which is similar to the epidermis [7,8]. For and good free radical scavenging capacity have enormous potential as a
serious skin defects, the wound will have a severe impact on people's novel biomaterial for wound healing application.
health and even threaten their life [9]. Thus, a large number of different
kinds of biomaterials have been fabricated to accelerate wound healing
2. Materials and method
process and reduce the formation of scar tissue, such as rubbers,
sponges, films, hydrogels, and nanofibrous membranes (NFMs), etc
2.1. Materials
[1,10–14]. Among these dressings for wound healing application, NFMs
developed by electrospinning have become one of the current research
Chitosan (CS) with a molecular weight of 100–300 kDa was ob-
hotspots which are attributed to NFMs with high surface-volume ratio
tained from J&K Scientific Ltd. Poly(ε-caprolactone) (Mn = 80 kDa),
and microporous structure, and these are crucial properties for cell’s
acetic acid (AcOH), glycidyltrimethylammonium chloride (GTMAC),
migration, adhesion, growth, differentiation, and angiogenesis [15–17].
hydrochloric acid (HCl), ammonium persulfate, sodium hydroxide
Specifically, NFMs with the morphology similar to the extracellular
(NaOH), and trifluoroacetic acid (TFA) were obtained from Sigma. All
matrix (ECM) could attract fibroblasts to the dermis, which can secrete
other materials or chemical reagents were used without further pur-
growth factor, collagen, and other ECM components to accelerate the
ification unless otherwise stated.
repair of damaged tissue [18–20].
Synthetic polymers including poly(ε-caprolactone) (PCL), poly-
urethane, poly(vinyl acetate), polyacrylonitrile, poly(L-lactic acid-co- 2.2. Synthesis of quaternized chitosan grafted polyaniline (QCSP)
glycolic acid), and poly(vinyl alcohol) have been widely used to fab-
ricate wound dressings [21,22]. Particularly, PCL (a FDA approved QCSP was synthesized according to the references with modification
material) has been often selected as a main component to produce [32]. Materials and methods in Supporting Information (SI) shows
nanofibers due to its biodegradability, biocompatibility, spinnability, specific synthesis procedures of QCSP.
and high toughness [23,24]. Unfortunately, for the lack of functional
groups, PCL was often incorporated with other polymeric materials or
natural materials as wound dressing, such as antibacterial agents and so 2.3. Preparation of PCL/QCSP polymer solution
on [25–28]. The wound sites are often in a moist, warm and nutrient-
rich environment which increases the risk of bacterial infection of the PCL/QCSP NFMs were fabricated by electrospinning. Briefly, PCL
wound and further results in the prolonged and excessive inflammatory was dissolved in AcOH to form 15% (wt/vol) solution. QCSP was dis-
response [29,30]. Besides, the wound healing process can also be dis- solved in TFA at the concentration of 1.5–7.5 wt% overnight at room
turbed by other several factors, such as free radicals, necrosis, hypoxia, temperature. The electrospinning polymer solutions were obtained by
and so on [31]. To address this issue, developing a new type of NFM mixing PCL with QCSP polymer solution in a ratio of 2:1 (v/v) at 25 °C
wound dressing with multifunctional properties including inherent under vigorous stirring for 15 min. The samples with the QCSP contents
antibacterial activity, antioxidant ability, and electroactivity is highly (weight ratio) of 0%, 5%, 10%, 15% and 20% to PCL/QCSP NFMs were
desirable. named as PCL/QCSP0, PCL/QCSP5, PCL/QCSP10, PCL/QCSP15 and
Quaternized chitosan-graft-polyaniline (QCSP) successfully synthe- PCL/QCSP20, respectively.
sized in our previous work exhibited superior cytocompatibility and
antibacterial activity than pure quaternized chitosan because of the
2.4. Electrospinning parameters
combined antibacterial property of QCS and polyaniline [32]. It also
showed good electroactivity and can reduce excessive free radicals in
The specific parameters for electrospinning were set based on our
the wound sites which can lead to tissue damage, infection, and pro-
previous study with some modifications [33]. Briefly, the electrospin-
longed wound healing. We have further demonstrated that hydrogels
ning polymer solution was fed into a 5 mL glass syringe fitted with a
containing QCSP greatly accelerated the healing process of wound in a
gauge 19 stainless steel needle as the nozzle. The positive voltage and
mouse full-thickness wounds defect model, indicating that QCSP is an
negative voltage were set to 17 kV and −4 kV, respectively. The tip-to-
excellent component for wound dressing. However, designing of elec-
collector distance and flow rate of feed solutions were fixed at 15 cm
troactive nanofiber wound dressing based on quaternized chitosan-g-
and 0.04 mL h−1, respectively. Materials and methods in SI shows de-
polyaniline and PCL as a novel multifunctional wound dressing has not
tailed procedures.
been reported.
By combining the good mechanical property of PCL and multi-
functionality of QCSP, a series of antibacterial, electroactivity, anti- 2.5. Characterizations
oxidant nanofiber wound dressing were designed and produced in this
work, which showed biomimetic multifunctional features for wound The physical and chemical characterizations of the NFMs including
healing application. The healing efficiency of PCL/QCSP NFM wound Fourier transform infrared spectroscopy (FT-IR), scanning electron
dressings was further evaluated by performing a mouse full-thickness microscope (SEM), mechanical properties, and swelling ratio of PCL/
wounds defect model. The nanofiber wound dressings were fabricated QCSP NFMs was assessed according to our previous studies, and the
by electrospinning PCL/QCSP polymer solutions at a high DC voltage. detailed procedures were shown in Materials and methods in SI
The morphology, chemical structure, mechanical properties, [34–37].

2
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

2.6. Electroactivity of PCL/QCSP NFMs 2.12. Histological analysis

Cyclic voltammetry of PCL/QCSP was carried out on electro- Histomorphological determination during different phases of wound
chemical Workstation (CHI 660D Instruments) to assess electroactivity regeneration was carried out by Hematoxylin-Eosin (H&E) staining for
[38]. Briefly, the electrospinning polymer solution was dropped on the assessment of the inflammation and epidermal regeneration in wound
surface of an indium tin oxide (ITO) glass and then placed at 45 °C for area. Briefly, the regenerated wound tissue collected on 3rd, 7th, and
4 h to obtain the electroactive PCL/QCSP film. The electrochemical 14th day were fixed with 4% paraformaldehyde for 24 h. The sample
property of these PCL/QCSP NFMs was performed by using an Elec- was then embedded in paraffin and cross sectioned to 40-μm thick
trochemical Workstation (CHI 660D Instruments) with a scan rate of slices. The slices were stained with Hematoxylin-Eosin (Beyotime,
25 mV s−1. Materials and methods in SI shows detailed procedures. China) and analyzed and photo-captured by microscope (IX53,
Olympus, Japan). Materials and methods in SI shows specific proce-
2.7. Anti-oxidant ability of PCL/QCSP NFMs dures about animal surgery.

The anti-oxidant abilities of PCL/QCSP samples were tested by 2.13. Collagen deposition analysis
scavenging efficiency for the stable 1, 1-diphenyl-2-picrylhydrazyl
(DPPH) free radical according to our previous study [9]. Materials and Commercial kits (Jiancheng bioengineering, China) was used to
methods in SI shows specific procedures. estimate the content of hydroxyproline to evaluate the amount of col-
lagen in the wound. Briefly, the regenerated wound tissue collected on
the 3rd, 7th, and 14th day at the weight of 30 mg were made into disc-
2.8. Surface antibacterial activity shaped tissue (diameter = 1 cm), and collagen amount was evaluated
by estimating hydroxyproline content using a commercial kit. All op-
The antibacterial activity of all PCL/QCSP NFMs was measured by a erations were carried out strictly in accordance with manufacturer's
surface antibacterial activity test by against both Staphylococcus aureus instructions. Materials and methods in SI shows specific procedures.
(ATCC 29213) and Escherichia coli (ATCC 8379), which are re-
presentatives of gram-positive and gram-negative bacteria, respec- 2.14. Immunohistochemistry examination
tively. In brief, the sterilized PCL/QCSP NFMs were placed 24-well
culture plate. 10 μL of bacterial suspension in sterilized PBS was added The wound sections were immunohistochemistry stained with VEGF
onto the surface of PCL/QCSP NFMs. The inoculated PCL/QCSP NFMs and TNF-α to evaluate inflammation and blood vessels regeneration
were incubated at 37 °C in a relative humidified atmosphere (not less under a standard protocol [12]. Briefly, for immunofluorescence
than 90%) to evaluate the antibacterial property of these wound dres- staining, the fixed and frozen regenerated wound tissue sections col-
sings. The bacteria-killing ratios of PCL/QCSP NFMs were calculated by lected on the 3rd, 7th, and 14th day were stained with TNFA antibody
the following equation: (5 μg/mL) and Anti-VEGFA antibody (5 μg/mL). FITC-conjugated goat
bacteria count of control − survivor count on PCL/QCSPNFMs anti-mouse IgG (cwbiotech) and FITC-conjugated goat anti-rabbit IgG
kill% = (cwbiotech) were used as the secondary antibody to reveal TNF-α and
bacteria count of control
VEGF expression. The nuclei were stained with DAPI containing
× 100%
mounting solution. Slides were observed under an inverted fluorescence
The detail procedures for assessing the surface antibacterial activity microscope. Materials and methods in SI shows specific procedures.
of PCL/QCSP NFMs are available in Materials and methods in SI.
2.15. Statistical analysis
2.9. Hemolytic activity assay of PCL/QCSP NFMs
All the results were analyzed statistically and expressed as a
The blood compatibility of PCL/QCSP NFMs was evaluated ac- mean ± standard deviation (SD). One-way ANOVA was used to de-
cording to hemolysis activity assay by using the mouse blood [39,40]. termine statistical differences and followed by a Bonferroni post hoc
Materials and methods in SI shows specific procedures. test for multiple comparisons with SPSS, version 24 (IBM). Differences
were considered significant if P < 0.05.

2.10. Cytocompatibility evaluation of PCL/QCSP NFM wound dressings 3. Results and discussions

The cytotoxicity was evaluated to assess the cytocompatibility of all 3.1. Fabrication and characterization of PCL/QCSP NFM wound dressings
PCL/QCSP NFMs by a direct contact test between the NFM groups and
the L929 cells (mouse fibroblast cells) [41–43]. Materials and methods A series of PCL/QCSP NFMs with suitable mechanical properties,
in SI shows specific procedures. electroactivity, anti-oxidant ability, good biocompatibility, and ex-
cellent antibacterial activity were prepared for wound healing appli-
2.11. In vivo wound healing in a mouse full-thickness wounds defect model cation in this study (Fig. 1a). With PCL as the main component, con-
tinuous fibers with diameters ranging from 60 to 200 nm were prepared
A mouse full-thickness wounds defect model was conducted to as- with the increase of QCSP content in PCL/QCSP NFMs. Furthermore,
sess the in vivo wound healing capacity of PCL/QCSP NFMs according with the addition of QCSP, the NFM wound dressings were endowed
to the publications [9,44]. Female Kunming mice (30–40 g, 5–6-week with anti-bacterial, anti-oxidant, and electroactive property. It had been
age) were employed to conduct the in vivo experiments. All mice were demonstrated that electroactive scaffolds are conducive to the elec-
randomly divided into 3 groups including the group of Tegaderm™ film, trically excitable cell’s proliferation and differentiation, such as osteo-
PCL/QCSP0, and PCL/QCSP15. Each group contained 15 mice. The blasts, fibroblasts, neurons, myoblasts, and cardiomyocytes
process of wound regeneration was studied by wound area monitoring. [9,34,45–47]. QCSP polymer was firstly synthesized and then mixed
Materials and methods in SI shows specific procedures about animal with PCL under vigorous stirring for 15 min before electrospinning. The
surgery. All animal experiments were performed in accordance with detailed parameters of each PCL/QCSP NFMs are shown in Fig. 1b. All
current guidelines for the care of laboratory animals and were approved the NFMs exhibited good stability according to testing the mass loss by
by the animal research committee of Xi'an Jiaotong University. immersing each PCL/QCSP NFM in PBS at 37 °C (Fig. S1), which was

3
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 1. (a) Schematic diagram of production and characteristics of PCL/QCSP NFMs; (b) Parameters of each PCL/QCSP NFM samples; (c) FT-IR spectra of CS, QCSP,
PCL, and PCL/QCSP20 NFM; (d) SEM images of pure PCL and PCL/QCSP nanofibers at low magnification (5000×); (e) SEM images of pure PCL and PCL/QCSP
nanofibers at high magnification (20000×); (f) Diameter distribution of pure PCL and PCL/QCSP nanofibers.

mainly because PCL chains in PCL/QCSP polymer solution can make to quinine diimine ring and benzonoid diamine ring stretching of
strong entanglement with QCSP chains. The chemical structure of PCL/ polyaniline in QCSP. The FT-IR spectrum of PCL/QCSP20 shows all
QCSP NFMs was confirmed by FT-IR as shown in Fig. 1c. The char- these characteristic peaks. The morphologies of PCL/QCSP NFMs at
acteristic peak of PCL at about 1722 cm−1 is attributed to carbonyl different magnification were observed by SEM (Fig. 1d, e). A series of
stretching, and –COC– is seen at 1240 cm−1 and 1290 cm−1. The nanofiber wound dressings with a smooth surface and random or-
characteristic peak of chitosan (CS) and QCSP at about 1640 cm−1 is ientation were fabricated successfully. The nanofiber diameters of pure
attributed to carbonyl stretching vibration of acetylated amino group. PCL and PCL/QCSP nanofibers were 91 ± 24 nm (PCL/QCSP0),
The peak corresponding to the methyl band of GTMAC at 1475 cm−1 135 ± 33 nm (PCL/QCSP5), 169 ± 38 nm (PCL/QCSP10),
was present in the spectra of QCSP, indicating that GTMAC was suc- 174 ± 38 nm (PCL/QCSP15), and 175 ± 35 nm (PCL/QCSP20), re-
cessfully conjugated to the amine group on chitosan backbone. A pair of spectively (Fig. 1f).
characteristic peaks at about 1572 cm−1 and 1490 cm−1 are attributed

4
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 2. Mechanical properties and swelling ratio of PCL/QCSP NFMs. (a) Tensile strain; (b) Tensile stress; (c) Young's modulus; (d) Stress-strain profile of PCL/QCSP
NFMs; (e) Swelling ratio of PCL/QCSP NFMs in PBS at 37 °C.

3.2. Mechanical properties and swelling behavior of PCL/QCSP NFMs 3.3. Electrochemical property and antioxidant ability of PCL/QCSP NFMs

The essential requirements of the suitable multifunctional wound Electroactive polymers such as polyaniline, polypyrrole, and poly-
dressing materials should include the desirable mechanical properties thiophene with the ability to regulate cellular activities of electrically
and water uptake ability to provide protection on the wound site and excitable cells bring a novel focus on designing biomaterials for skin
absorb the effluence to promote the healing process of wound [12,48]. wound repair [51–53]. For example, fibroblast and myoblast cells,
For those joints that move and bend frequently like knee and ankle, which are one of the key players for orchestrating physiological tissue
wound dressing should match the extensibility of the native skin to repair, could adhere and proliferate on the electroactive polymers
maintain integrity of the dressing and prolong their lifespan during based wound dressings and then to accelerate the wound healing pro-
wound healing process. Fig. 2a and b shows the tensile strain, tensile cess [54–56]. It is known that electroactive materials can become
stress and Young's modulus of PCL/QCSP NFMs, respectively. These electrical conductors by reversible ion exchange between reduction
wound dressings have suitable stretchability between 81.7% and 48.1% state and oxidation state [57]. Cyclic voltammetry measurement was
(Fig. 2d), which is close to human skin (75–60%) except for PCL/QCSP0 conducted to assess the electroactivity of PCL/QCSP NFMs. As shown in
[49]. PCL/QCSP5 with the lowest QCSP content exhibited the highest Fig. 3a, two pairs of the oxidation/reduction peaks appeared at about
elongation-at-break (81.7%). Moreover, with the QCSP content in- 0.20 V, and 0.58 V, respectively. The first peak at 0.20 V was due to the
creasing, the elongation-at-break of PCL/QCSP NFMs were decreased leucoemeraldine to emeraldine state transition, while the second peak
gradually. However, compared with PCL/QCSP0 NFM, the Young’s at 0.58 V was attributed to the emeraldine oxidation state to the per-
modulus of PCL/QCSP samples obviously decreased with the addition nigraniline state. The cyclic voltammograms of PCL/QCSP NFMs are
of QCSP in PCL/QCSP nanofibrous matrix, indicating that QCSP de- similar to each other differing only by the broadness of the oxidation/
creased the PCL/QCSP nanofibrous membrane’s modulus. The Young’s reduction peaks, which shows QCSP dependence, and there is no oxi-
modulus of PCL/QCSP NFMs ranged from 2.4 MPa to 4.9 MPa, which dation/reduction peak in PCL/QCSP0 NFM (Fig. S2). All the results
are comparable to human soft tissue [50]. above demonstrated that these QCSP contained PCL/QCSP NFMs pos-
Swelling behavior of PCL/QCSP NFMs was determined in PBS at sess good electroactivity, indicating that they have great potential for
37 °C (Fig. 2e). All the groups reached a swelling equilibrium within the treatment of skin wounds [9,36,58].
3 h. The lowest water absorption about 70% was observed for the group In the inflammatory phase of wound healing, neutrophils, leuco-
of PCL/QCSP20 after 3 h, and 130%, 120% and 100% for PCL/QCSP5, cytes, and monocytes will be attracted to the wound sites by biologi-
PCL/QCSP10, and PCL/QCSP15, respectively, suggesting that the cally active mediators and then attack the microorganisms and foreign
swelling ratio decreased with increasing the content of QCSP in PCL/ debris via phagocytosis. This process will lead to the overproduction of
QCSP NFMs. The water uptake ability of PCL/QCSP NFMs indicates that free radicals including superoxide anion, hydrogen peroxide, and hy-
these wound dressings can absorb the exudate from the wound, sug- droxyl anion [59]. The excessive level of these free radicals will induce
gesting that these dressings have great potential as wound dressings. the disruption of the cellular oxidant/antioxidant balance and cause the
inactivation of enzyme, DNA breakage, and lipid peroxidation, which
further result in the extensive collateral damage to the surrounding skin

5
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 3. (a) Electrochemical property of PCL/QCSP20 NFM; (b) Antioxidant ability of PCL/QCSP15 NFM dispersion liquid. *p < 0.05, **p < 0.01.

tissue over the wound sites and causing a prolonged process of wound ammonium groups which is effective for killing bacteria by electrostatic
[60,61]. Therefore, an important strategy to enhance the healing pro- adherence [32,69]. What’s more, for those gram-negative bacteria, the
cess of wound is to reduce/scavenge the free radicals around the wound expression of genes that are vital to bacterial survival can be down-
sites by the introduction of antioxidant agents, thereby to protect the regulated with the existence of polyaniline, which will further affect the
cells or tissues from damage and then to accelerate the wound healing formation of cell wall and the metabolism of energy, and eventually
process [62,63]. Herein, we tested the scavenging efficiency for DPPH% lead to the death of those gram-negative bacteria [70–72]. Hence, the
to evaluate the antioxidant abilities of these PCL/QCSP NFMs and PCL/ synergetic antibacterial activity of PCL/QCSP NFMs was assigned to the
QCSP15 was taken as an example (Fig. 3b). The results suggested that complementary effect of positive-charged amino groups, quaternary
the antioxidant ability of PCL/QCSP15 NFM increased with the in- ammonium groups, and acidified polyaniline. Beyond this, PCL/
creasing concentration of QCSP and almost 70% of free radicals can be QCSP15 and PCL/QCSP20 groups with good antibacterial activity
cleared by 6 mg mL−1 of PCL/QCSP15 dispersion liquid. Besides, the suggested that they have great potential to effectively prevent bacterial
scavenging efficiency for DPPH% more than 80% was achieved when infection on the wound.
the content of PCL/QCSP15 dispersion liquid reached to 8 mg mL−1.
These data demonstrated the good antioxidant ability of these PCL/
QCSP NFMs. It has been confirmed that the dressings with antioxidant 3.5. Hemocompatibility and cytocompatibility of PCL/QCSP NFM
ability could significantly accelerate the in vivo wound healing process
[64–66]. Thus, these polyaniline-contained PCL/QCSP NFMs, as an Biomedical materials possessing good biocompatibility is the pre-
anti-oxidant wound dressing, have a great potential in wound healing requisite for novel wound dressing [73]. Thus, we tested hemo-
application. compatibility and cytocompatibility of the NFMs to estimate bio-
compatibility of these NFM wound dressings. The in vitro hemolysis test
was first carried out. The macroscopical color of all the five NFM
3.4. Antibacterial activity of PCL/QCSP NFMs groups, negative control group (PBS), and positive control group
(Triton X-100) were shown in Fig. 5a. All the NFM groups presented
It is well known that bacterial infections may result in increased light yellow that was similar to the negative control group, while the
exudate at the wound site and delay the healing process of wound positive control group was bright red. Furthermore, the quantitative
[67,68]. Hence, an optimum wound dressing material should possess data of hemolysis test was shown in Fig. 5b. Hemolysis ratio of all the
inherent antimicrobial properties to increase the wound healing rate by NFM wound dressings decreased with the QCSP content decreasing
casting down the number of pathogens and reducing the inflammatory (1.09%–0.44%). compared with other NFM groups, PCL/QCSP20 ex-
response of wounds [9,60]. A surface antibacterial activity test was hibited the highest hemolysis ratio (1.45%). All these data indicated
used to evaluate antibacterial activity of all PCL/QCSP NFMs by against that all the NFM groups showed excellent hemocompatibility for wound
both S. aureus and E. coli in this study. Firstly, the antibacterial test was healing application.
conducted with 105 CFU mL−1 of the bacterial suspensions and all PCL/ PCL/QCSP NFMs have great potential to promote the proliferation
QCSP NFM groups showed excellent killing ratio (> 90%) for both S. of electrically excitable cells due to the good electroactivity of QCSP
aureus and E. coli expect for PCL/QCSP0 NFM group (Fig. 4a, c). Thus, itself. Thus, the proliferation of L929 cells was assessed by a direct
to further detect the difference of antibacterial properties among these contact test, PCL/QCSP0 NFM was taken as control by seeding the same
NFM wound dressings, 106 CFU mL -1 of the bacterial suspension was initial density. The results of Alamar-Blue assay exhibited a significant
used in the following test and the results were shown in Fig. 4b and d. growth trend from the 1st to 5th day (Fig. 5c), indicating that the L929
The lowest bacteria-killing ratio for S. aureus about 63% and about cells had good growth throughout the whole experiment. There was no
71% for E. coli was observed for the group of PCL/QCSP5. Furthermore, obvious difference in cell viability between the QCSP contained groups
the killing ratio for S. aureus about 70%, 86%, and 92% and for E. coli and PCL/QCSP0 group on the first day. After 3 days of incubation, the
about 82%, 94%, and 95% were observed for PCL/QCSP10, PCL/ cell number with two or three times increase except for PCL/QCSP20,
QCSP15, and PCL/QCSP20, respectively, indicating that the anti- indicating that PCL/QCSP20 group has negative effect on L929 cells.
bacterial activity of the NFMs gradually increased with increasing the Besides, compared with PCL/QCSP0 group (P < 0.01) and other QCSP
content of QCSP in PCL/QCSP NFMs. contained groups, PCL/QCSP15 group showed the highest cell pro-
PCL/QCSP NFMs exhibited good antibacterial activity from QCSP. liferation on day 3. On the fifth day, PCL/QCSP5, PCL/QCSP10, and
QCS possessed positive-charged amino groups and quaternary PCL/QCSP15 groups exhibited higher cell number than PCL/QCSP0

6
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 4. Antibacterial activity for S. aureus of PCL/QCSP NFMs with the bacterial suspension at the concentration of 105 CFU mL−1 (a) and 106 CFU mL−1 (b);
Antibacterial activity for E. coli of PCL/QCSP NFMs with the bacterial suspension at the concentration of 105 CFU mL−1 (c) and 106 CFU mL−1 (d), *p < 0.05,
**p < 0.01.

and PCL/QCSP20 (P < 0.01), and there was no difference among the containing a higher content of QCSP. However, the cytotoxicity of PCL/
group of PCL/QCSP5, PCL/QCSP10, and PCL/QCSP15. All these results QCSP NFMs will also increase with QCSP content increasing.
confirmed that the PCL/QCSP5, PCL/QCSP10, and PCL/QCSP15 had Fortunately, PCL/QCSP15 NFM with a good balance between anti-
good biocompatibility and the ability to enhance the proliferation of bacterial properties and cytocompatibility have great potential in
L929 cells. treatment of skin wounds. Thus, the wound healing performance of the
A series of PCL and PCL/QCSP15 NFM wound dressings extracts PCL/QCSP NFMs was further evaluated in a mouse full-thickness
with concentrations ranging from 0 to 20 mg/mL were obtained after wounds defect model. PCL/QCSP15 with a suitable effect for cell pro-
soaking each NFMs with cell culture medium for 72 h to further eval- liferation and prominent antibacterial activity was selected as a re-
uate the biocompatibility of NFM wound dressings (Fig. 5). There was presentative. PCL/QCSP0 and Tegaderm™ film were taken as control
no difference in cell number between all the five different concentra- groups. On the 3rd, 7th, and 14th day, PCL/QCSP15 group showed the
tions of extracts, which confirmed that the extracts from PCL/QCSP15 advantageous wound contraction over Tegaderm™ film group and PCL/
had no cytotoxic leaching content. Besides, most of L929 cells were QCSP0 group (Fig. 6a-c). In detail, on the 3rd day, the wound areas of
green in all the NFM’s extract groups after 24 h incubation and showed all the groups reduced to some extent after the surgery, the largest
a spindle-like morphology, which was consistent with the quantitative wound contraction area (43%, P < 0.01) was presented in PCL/
data of alamarBlue® assay for wound dressing extracts of PCL/QCSP15 QCSP15 group, suggested that it had a higher effect on promoting
NFM. Overall, all these experiment results demonstrated that all these wound healing. On the 7th day, PCL/QCSP15 group and PCL/QCSP0
NFM wound dressings had good biocompatibility except for PCL/ group showed significant difference (P < 0.01) with Tegaderm™ film
QCSP20. PCL/QCSP15 group with balanced property of antibacterial group, while the wound contraction of PCL/QCSP15 group was higher
activity and cell proliferation was chosen for further evaluation of than the group of PCL/QCSP0 (P < 0.05), indicating that both two
wound healing performance in vivo. PCL/QCSP NFM groups had better therapeutic efficacy than Tegaderm™
film group. On the 14th day, the wounds treated with PCL/QCSP15
3.6. In vivo wound healing NFM or PCL/QCSP0 NFM were observed to have closure and had about
9% lead than Tegaderm™ film group in wound contraction (P < 0.05).
All the above results indicated that the physical properties will be Besides, even the regenerated skin of some mice had presented hair
significantly affected by the introduction of QCSP. The surface anti- after treated with PCL/QCSP15 for 14 days. These quantitative data
bacterial activities of PCL/QCSP NFMs also exhibited a QCSP depen- about the wound area demonstrate that PCL/QCSP15 NFM had better
dence and a more efficient antibacterial effect presented in the group wound healing effect than PCL/QCSP0 group and Tegaderm™ film

7
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 5. Hemocompatibility and cytocompatibility analysis of PCL/QCSP NFM wound dressings. (a) Images of RBC; (b) Hemolysis ratio (%) of the NFMs by direct
contact with the nanofiber wound dressings for 1 h; (c) Quantitative analysis of L929 cell proliferation by a direct contact with PCL/QCSP wound dressings and PCL/
QCSP0 was taken as a control, *p < 0.05,**p < 0.01; (d) Quantitative analysis of L929 cell viability in the extracts of PCL and PCL/QCSP15 for 3 days at different
concentrations, *p < 0.05,**p < 0.01; (e) LIVE/DEAD staining of L929 cells after incubated with the extracts for 24 h.

group. Our previous work had confirmed that all the healing stages can wound site in these three groups. What’s more, compared with Tega-
be accelerated by the wound dressing based on QCSP, which was due to derm™ and PCL/QCSP0 group, PCL/QCSP15 group exhibited less in-
the synergistic effect of QCS and polyaniline [9]. Beyond that, wound flammatory cells and more fibroblast cells around the impaired region,
healing process was further accelerated in the midanaphase due to which may be attributed to the antioxidant and electroactivity of PCL/
antioxidative wound dressing [74]. Compared with Tegaderm™ film QCSP15 NFM [9]. These results indicated that PCL/QCSP15 group with
group, PCL/QCSP0 group showed better therapeutic effect throughout the ability to partially suppress inflammatory cell infiltration and ac-
the whole healing stages because the NFMs with appropriate porosity celerate wound healing process among these three groups. With the
have the permeable ability to moisture and air, and maintain a partially further healing of the wounds, all three groups presented less in-
immobilized moist environment by absorbing the effluence from the flammatory cells on the 7th day, and a layer of epithelium was formed
wound area [75]. Thus, PCL/QCSP15 NFM with suitable physical in the group of PCL/QCSP15 and PCL/QCSP0. Compared with Tega-
structure and chemical properties could promote the wound healing derm™ film and PCL/QCSP0 group, PCL/QCSP15 group exhibited a
and showed better therapeutic efficacy than Tegaderm™ film and PCL/ higher regularity of both connective tissue and epithelium. On the 14th
QCSP0 NFM on wound healing. day, Tegaderm™ film group showed the basic structure of regenerated
wound tissue including epithelium and dermis, while PCL/QCSP0
group showed a few hair follicles formation. Importantly, PCL/QCSP15
3.7. Histological analysis of wound regeneration
group showed the maximum amount of well-proliferated fibroblast,
thickened epidermis, hair follicles, and mature blood vessels, suggested
Histological analysis was performed to further distinguish the
that PCL/QCSP15 had the best wound healing performance among
quality of the regenerated tissue in the wounds among commercial
these thress groups. In summary, PCL/QCSP15 group showed a higher
Tegaderm™ film, PCL/QCSP0, and PCL/QCSP15 groups by H&E
density of fibroblasts (on the 3rd day), completely formated epithelium
staining, the indicators mainly included inflammatory cells infiltration,
structure (on the 7th day), and high differentiated epithelial tissue
fibroblast immigration, connective tissue synthesis, and re-epitheliza-
which was closer to normal skin (on the 14th day), exhibiting the
tion [76]. The mild acute inflammatory responses were shown on the
prominent wound healing performance. PCL/QCSP0 group showed a
3rd day in Fig. 7a, fibroblasts and inflammatory cells migrated to the

8
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 6. (a) Photographs of the wound closure on the 3rd, 7th and 14th day for commercial Tegaderm™ film, PCL/QCSP0, and PCL/QCSP15 NFM; (b) The schematic
diagram of wounds closure on the 3rd, 7th and 14th day; (c) Quantitative statistical analysis of wounds closure for commercial Tegaderm™ film, PCL/QCSP0, and
PCL/QCSP15 NFM treatment. *P < 0.05, **P < 0.01.

better therapeutic effect than Tegaderm™ film group. cessation of the cell around wound bed [80]. Thus, we selected tumor
Granulation tissue consists of some growth factors, fibroblasts, ac- necrosis factor-α (TNF-α) as an indicator to verify the treatment effi-
cumulated ECM, and other cells [77], which can also work with par- cacy of PCL/QCSP NFMs in preventing infection by immuno-
enchymal cells in the repair process of some inflammatory injury [78]. fluorescence staining. Compared with the Tegaderm™ film and PCL/
Thus, an important indicator in wound healing process is the thickness QCSP0 NFM group, PCL/QCSP15 group showed the lowest expression
of granulation tissue, which can evaluate the therapeutic effect of the of TNF-α on day 7 and 14, PCL/QCSP0 group showed lower TNF-α
wound dressing. After 14 days of surgery, the granulation tissue in PCL/ expression than Tegaderm™ group during the whole wound healing
QCSP15 group exhibited approximately 225 mm thicker than that in stages (Fig. 8a), which was probably due to the introduction of QCS and
the group of PCL/QCSP0 (P < 0.05), while the granulation tissue of polyaniline that can endow the antibacterial ability to PCL/QCSP NFM
the Tegaderm™ film group was thinner than all the NFM groups (Fig. 7b wound dressings to reduce inflammation. In addition to preventing
and c). In one word, according to in vivo experimental studies, PCL/ infection, angiogenesis of the wound was further estimated. Vascular
QCSP15 showed the best wound healing performance among these endothelial growth factor (VEGF) was selected as an indicator to
three groups. evaluate wound repair by immunohistochemical staining of regenera-
tion wound sections on day 7 and 14. The vascular angiogenesis in PCL/
3.8. Collagen deposition analysis QCSP15 group appeared grossly more VEGF expression than Tega-
derm™ film and PCL/QCSP0 NFM group on day 7 (P < 0.05) (Fig. 8).
One of the important markers of wound healing is the total collagen Besides, VEGF expression was lowest in the Tegaderm™ film group and
level. Due to the metabolism of collagen, the therapeutic effects were there was no difference in the group of PCL/QCSP0 and PCL/QCSP15
evaluated by analyzing the hydroxyproline content, because the hy- on the 14th day. In conclusion, by synchronously reducing the pro-
droxyproline is a unique component in collagen fibers and possesses a duction of TNF-α and upregulating expression of VEGF, PCL/QCSP15
constant mass of about 13.4% [79]. Fig. 7d shows that the concentra- NFM group significantly accelerated the wound healing process than
tion of hydroxyproline in all groups increased continuously in the first Tegaderm™ films.
7 days and maintained at a stable level on the 14th day over the whole
healing stage. Compared with Tegaderm™ film group, PCL/QCSP NFM 4. Conclusion
groups exhibited higher collagen level during the 14 days (P < 0.01),
indicating that PCL/QCSP NFM groups had a better therapeutic effect A series of antibacterial anti-oxidant and electroactive nanofiber
than Tegaderm™ films, and PCL/QCSP15 group showed the best wound membranes were developed by electrospinning of PCL/QCSP polymer
healing performance through the whole experiment. In conclusion, by solutions, and in vivo experiments demonstrated that the wound
combining the results of H&E staining, the thickness of granulation healing process can be significantly accelerated by these multi-func-
tissue, and the deposition of collagen, PCL/QCSP15 group exhibited the tional NFM wound dressings. PCL/QCSP NFMs also exhibited tunable
superior therapeutic effects than Tegaderm™ film and PCL/QCSP0 water absorption and modulus by tuning the concentration of QCSP to
group, which illustrated that the PCL/QCSP15 wound dressing greatly PCL/QCSP NFMs. Hemocompatibility and cytocompatibility were
increased the healing process of wound. confirmed by a co-culture with erythrocytes or L929 cells, respectively.
For the in vivo wound healing test, PCL/QCSP15 NFM exhibited a faster
3.9. TNF-α and VEGF expression in regenerated wound tissue wound healing rate than Tegaderm™ Film and PCL/QCSP0 NFM.
Moreover, the wound treated with PCL/QCSP15 NFM showed fewer
During the wound healing process, many required cellular activities inflammatory infiltration, higher density of fibroblasts, thicker granu-
were adjusted by cytokines, which mainly regulated the initiation and lation tissue thickness and collagen deposition. The

9
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 7. (a) Histological analysis (H&E staining) of wound regeneration with different treatment of Tegaderm™ film dressing, PCL/QCSP0, and PCL/QCSP15 NFM
wound dressing on the 3rd, 7th, and 14th day, green lines show the boundary of epidermis and dermis; (b) Images of granulation tissue thickness on the 14th day; (c)
Quantitative statistical analysis of granulation tissue thickness for each treatment of Tegaderm™ film dressing, PCL/QCSP0 NFM, and PCL/QCSP15 NFM on the 14th,
*P < 0.05, **P < 0.01; (d) Collagen content in Tegaderm™ film, PCL/QCSP0 group, and PCL/QCSP15 group by testing hydroxyproline concentration. *P < 0.05,
**P < 0.01. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

immunofluorescence staining of TGF-α and VEGF indicated the lowest are potential candidates for treatment of cutaneous skin wound healing.
production of TGF-α and the highest expression of VEGF after treated
with PCL/QCSP15 NFM for 7 days, which meant that PCL/QCSP15
Declaration of Competing Interest
wound dressing promoted the regeneration of blood vessel and reduced
the production of pro-inflammatory factor around the wound site. In
The authors declare that they have no known competing financial
summary, these multifunctional PCL/QCSP nanofiber wound dressings
interests or personal relationships that could have appeared to

10
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

Fig. 8. Pictures of the regenerated wound tissue on 7th, and 14th day by immunofluorescence staining labeling with (a) TNF-α (green) and (b) VEGF (green), yellow
arrows show the expression of TNF-α and red arrows present VEGF; Quantitative statistical analysis of TNF-α-actinin (c) and VEGF (d) relative area percentage; For
all quantitative analysis, Tegaderm™ Film group on 7th day was taken as 100%, *P < 0.05, **P < 0.01. (For interpretation of the references to color in this figure
legend, the reader is referred to the web version of this article.)

influence the work reported in this paper. Medicine Research, College of Stomatology, Xi’an Jiaotong University
(No. 2019LHM-KFKT008).
Acknowledgment
Appendix A. Supplementary data
This work was jointly supported by the National Natural Science
Foundation of China (grant numbers: 51673155 and 51973172), State Supplementary data to this article can be found online at https://
Key Laboratory for Mechanical Behavior of Materials, the Fundamental doi.org/10.1016/j.cej.2019.123464.
Research Funds for the Central Universities, and the World-Class
Universities (Disciplines) and the Characteristic Development Guidance References
Funds for the Central Universities, Natural Science Foundation of
Shaanxi Province (No. 2018JM5026, and 2019TD-020), and Opening [1] X. Rui, G. Luo, H. Xia, W. He, Z. Jian, L. Bo, J. Tan, J. Zhou, D. Liu, Y. Wang, Novel
bilayer wound dressing composed of silicone rubber with particular micropores
Project of Key Laboratory of Shaanxi Province for Craniofacial Precision

11
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

enhanced wound re-epithelialization and contraction, Biomaterials 40 (2015) 1–11. Electrospun polycaprolactone membranes incorporated with ZnO nanoparticles as
[2] J. Mansbridge, Skin tissue engineering, J. Biomat. Sci.-Polym. E 19 (8) (2008) skin substitutes with enhanced fibroblast proliferation and wound healing, Rsc Adv.
955–968. 4 (47) (2014) 24777–24785.
[3] A.R. Unnithan, A. Ghavami Nejad, A.R.K. Sasikala, R.G. Thomas, Y.Y. Jeong, [30] A.R. Unnithan, N.A. Barakat, P.T. Pichiah, G. Gnanasekaran, R. Nirmala, Y.-S. Cha,
P. Murugesan, S. Nasseri, D. Wu, C.H. Park, C.S. Kim, Electrospun zwitterionic C.-H. Jung, M. El-Newehy, H.Y. Kim, Wound-dressing materials with antibacterial
nanofibers with in situ decelerated epithelialization property for non-adherent and activity from electrospun polyurethane–dextran nanofiber mats containing cipro-
easy removable wound dressing application, Chem. Eng. J. 287 (2016) 640–648. floxacin HCl, Carbohydr. Polym. 90 (4) (2012) 1786–1793.
[4] A. Wells, A. Nuschke, C.C. Yates, Skin tissue repair: matrix microenvironmental [31] J. Fang, T. Seki, H. Maeda, Therapeutic strategies by modulating oxygen stress in
influences, Matrix. Biol. 49 (2016) 25–36. cancer and inflammation, Adv. Drug Deliv. Rev. 61 (4) (2009) 290–302.
[5] L. Rittié, Cellular mechanisms of skin repair in humans and other mammals, J. Cell [32] X. Zhao, P. Li, B. Guo, P.X. Ma, Antibacterial and conductive injectable hydrogels
Commun. Signal. 10 (2) (2016) 103–120. based on quaternized chitosan-graft-polyaniline/oxidized dextran for tissue en-
[6] J. Boateng, O. Catanzano, Advanced therapeutic dressings for effective wound gineering, Acta Biomater. 26 (2015) 236–248.
healing–a review, J. Pharmacol. Sci. 104 (11) (2015) 3653–3680. [33] X. Ma, J. Ge, Y. Li, B. Guo, P.X. Ma, Nanofibrous electroactive scaffolds from a
[7] S.R. Gomes, G. Rodrigues, G.G. Martins, M.A. Roberto, M. Mafra, C.M.R. Henriques, chitosan-grafted-aniline tetramer by electrospinning for tissue engineering, Rsc
J.C. Silva, In vitro and in vivo evaluation of electrospun nanofibers of PCL, chitosan Adv. 4 (26) (2014) 13652–13661.
and gelatin: a comparative study, Mater. Sci. Eng. C 46 (2015) 348–358. [34] Y. Wu, L. Wang, B. Guo, P.X. Ma, Interwoven aligned conductive nanofiber yarn/
[8] B. Guo, J. Qu, X. Zhao, M. Zhang, Degradable conductive self-healing hydrogels hydrogel composite scaffolds for engineered 3D cardiac anisotropy, ACS Nano 11
based on dextran-graft-tetraaniline and N-carboxyethyl chitosan as injectable car- (6) (2017) 5646–5659.
riers for myoblast cell therapy and muscle regeneration, Acta Biomater. 84 (2019) [35] Z. Deng, T. Hu, Q. Lei, J. He, P.X. Ma, B. Guo, Stimuli-responsive conductive na-
180–193. nocomposite hydrogels with high stretchability, self-Healing, adhesiveness, and 3D
[9] X. Zhao, H. Wu, B. Guo, R. Dong, Y. Qiu, P.X. Ma, Antibacterial anti-oxidant printability for human motion sensing, ACS Appl. Mater. Inter. 11 (7) (2019)
electroactive injectable hydrogel as self-healing wound dressing with hemostasis 6796–6808.
and adhesiveness for cutaneous wound healing, Biomaterials 122 (2017) 34–47. [36] Y. Liang, X. Zhao, T. Hu, Y. Han, B. Guo, Mussel-inspired, antibacterial, conductive,
[10] Q. Li, Y. Niu, H. Diao, L. Wang, X. Chen, Y. Wang, L. Dong, C. Wang, In situ se- antioxidant, injectable composite hydrogel wound dressing to promote the re-
questration of endogenous PDGF-BB with an ECM-mimetic sponge for accelerated generation of infected skin, J. Colloids Interf. Sci 556 (2019) 514–528.
wound healing, Biomaterials 148 (2017) 54–68. [37] X. Zhao, B. Guo, H. Wu, Y. Liang, P.X. Ma, Injectable antibacterial conductive na-
[11] J. Li, D. Zhai, F. Lv, Q. Yu, H. Ma, J. Yin, Z. Yi, M. Liu, J. Chang, C. Wu, Preparation nocomposite cryogels with rapid shape recovery for noncompressible hemorrhage
of copper-containing bioactive glass/eggshell membrane nanocomposites for im- and wound healing, Nat. Commun. 9 (1) (2018) 2784.
proving angiogenesis, antibacterial activity and wound healing, Acta Biomater. 36 [38] L. Wang, Y. Wu, T. Hu, B. Guo, P.X. Ma, Electrospun conductive nanofibrous
(2016) 254–266. scaffolds for engineering cardiac tissue and 3D bioactuators, Acta Biomater. 59
[12] J. Qu, X. Zhao, Y. Liang, T. Zhang, P.X. Ma, B. Guo, Antibacterial adhesive in- (2017) 68–81.
jectable hydrogels with rapid self-healing, extensibility and compressibility as [39] A. Sasidharan, L.S. Panchakarla, A.R. Sadanandan, A. Ashokan, P. Chandran,
wound dressing for joints skin wound healing, Biomaterials 183 (2018) 185–199. C.M. Girish, D. Menon, S.V. Nair, C.N. Rao, M. Koyakutty, Hemocompatibility and
[13] I. Garcia-Orue, G. Gainza, F.B. Gutierrez, J.J. Aguirre, C. Evora, J.L. Pedraz, macrophage response of pristine and functionalized graphene, Small 8 (8) (2012)
R.M. Hernandez, A. Delgado, M. Igartua, Novel nanofibrous dressings containing 1251–1263.
rhEGF and Aloe vera for wound healing applications, Int. J. Pharmaceut. 523 (2) [40] T. Hu, Y. Wu, X. Zhao, L. Wang, L. Bi, P.X. Ma, B. Guo, Micropatterned, electro-
(2017) 556–566. active, and biodegradable poly(glycerol sebacate)-aniline trimer elastomer for
[14] W. Xu, Q. Song, J.F. Xu, M.J. Serpe, X. Zhang, Supramolecular hydrogels fabricated cardiac tissue engineering, Chem. Eng. J. 366 (2019) 208–222.
from supramonomers: a novel wound dressing material, ACS Appl. Mater. Inter. 9 [41] J. Qu, X. Zhao, P.X. Ma, B. Guo, Injectable antibacterial conductive hydrogels with
(13) (2017) 11368–11372. dual response to an electric field and pH for localized “smart” drug release, Acta
[15] N.G. Rim, C.S. Shin, H. Shin, Current approaches to electrospun nanofibers for Biomater. 72 (2018) 55–69.
tissue engineering, Biomed. Mater. 8 (1) (2013) 014102. [42] L. Wang, Y. Wu, B. Guo, P.X. Ma, Nanofiber yarn/hydrogel core–shell scaffolds
[16] Z.-M. Huang, Y.Z. Zhang, M. Kotaki, S. Ramakrishna, A review on polymer nano- mimicking native skeletal muscle tissue for guiding 3D myoblast alignment, elon-
fibers by electrospinning and their applications in nanocomposites, Compos. Sci. gation, and differentiation, ACS Nano 9 (9) (2015) 9167–9179.
Technol. 63 (15) (2003) 2223–2253. [43] J. Qu, X. Zhao, P.X. Ma, B. Guo, pH-responsive self-healing injectable hydrogel
[17] S. Miguel, M. Ribeiro, P. Coutinho, I. Correia, Electrospun polycaprolactone/aloe based on N-carboxyethyl chitosan for hepatocellular carcinoma therapy, Acta
vera_chitosan nanofibrous asymmetric membranes aimed for wound healing ap- Biomater. 58 (2017) 168–180.
plications, Polymers 9 (5) (2017) 183. [44] J. Qu, X. Zhao, Y. Liang, Y. Xu, P.X. Ma, B. Guo, Degradable conductive injectable
[18] M. Seon-Lutz, A.C. Couffin, S. Vignoud, G. Schlatter, A. Hebraud, Electrospinning in hydrogels as novel antibacterial, anti-oxidant wound dressings for wound healing,
water and in situ crosslinking of hyaluronic acid/cyclodextrin nanofibers: towards Chem. Eng. J. 362 (2019) 548–560.
wound dressing with controlled drug release, Carbohydr. polym. 207 (2019) [45] B. Guo, L. Glavas, A.-C. Albertsson, Biodegradable and electrically conducting
276–287. polymers for biomedical applications, Prog. Polym. Sci. 38 (9) (2013) 1263–1286.
[19] S. Ahn, C.O. Chantre, A.R. Gannon, J.U. Lind, P.H. Campbell, T. Grevesse, [46] Y. Wu, L. Wang, B. Guo, Y. Shao, P.X. Ma, Electroactive biodegradable polyurethane
B.B. O'Connor, K.K. Parker, Soy protein/cellulose nanofiber scaffolds mimicking significantly enhanced Schwann cells myelin gene expression and neurotrophin
skin extracellular matrix for enhanced wound healing, Adv. Healthc. Mater. 7 (9) secretion for peripheral nerve tissue engineering, Biomaterials 87 (2016) 18–31.
(2018) 1701175. [47] B. Guo, P.X. Ma, Synthetic biodegradable functional polymers for tissue en-
[20] S.-J. Liu, Y.-C. Kau, C.-Y. Chou, J.-K. Chen, R.-C. Wu, W.-L. Yeh, Electrospun PLGA/ gineering: a brief review, Sci. China Chem. 57 (4) (2014) 490–500.
collagen nanofibrous membrane as early-stage wound dressing, J. Membr. Sci. 355 [48] M.D. Konieczynska, J.C. Villa-Camacho, C. Ghobril, M. Perez-Viloria, K.M. Tevis,
(1–2) (2010) 53–59. W.A. Blessing, A. Nazarian, E.K. Rodriguez, M.W. Grinstaff, On-demand dissolution
[21] A. Memic, T. Abudula, H.S. Mohammed, K. Joshi Navare, T. Colombani, of a dendritic hydrogel-based dressing for second-degree burn wounds through
S.A. Bencherif, Latest progress in electrospun nanofibers for wound healing appli- thiol–thioester exchange reaction, Angew. Chem. Int. Ed. 55 (34) (2016)
cations, ACS Appl. Biol. Mater. 2 (3) (2019) 952–969. 9984–9987.
[22] B. Guo, P.X. Ma, Conducting polymers for tissue engineering, Biomacromolecules [49] N. Annabi, D. Rana, S.E. Shirzaei, R. Portillo-Lara, J.L. Gifford, M.M. Fares,
19 (6) (2018) 1764–1782. S.M. Mithieux, A.S. Weiss, Engineering a sprayable and elastic hydrogel adhesive
[23] A. Cipitria, A. Skelton, T.R. Dargaville, P.D. Dalton, D.W. Hutmacher, Design, with antimicrobial properties for wound healing, Biomaterials 139 (2017) 229–243.
fabrication and characterization of PCL electrospun scaffolds - a review, J. Mater. [50] X. Chen, Making electrodes stretchable, Small Methods 1 (4) (2017) 1600029.
Chem. 21 (26) (2011) 9419–9453. [51] G. Kaur, R. Adhikari, P. Cass, M. Bown, P. Gunatillake, Electrically conductive
[24] Y. Qian, X. Zhou, H. Sun, J. Yang, Y. Chen, C. Li, H. Wang, T. Xing, F. Zhang, N. Gu, polymers and composites for biomedical applications, Rsc Adv. 5 (47) (2015)
Biomimetic domain-active electrospun scaffolds facilitating bone regeneration sy- 37553–37567.
nergistically with antibacterial efficacy for bone defects, ACS Appl. Mater. Inter. 10 [52] Z. Deng, Y. Guo, X. Zhao, L. Li, R. Dong, B. Guo, P.X. Ma, Stretchable degradable
(4) (2018) 3248–3259. and electroactive shape memory copolymers with tunable recovery temperature
[25] S. Jiang, B.C. Ma, J. Reinholz, Q. Li, J. Wang, K.A. Zhang, K. Landfester, D. Crespy, enhance myogenic differentiation, Acta Biomater. 46 (2016) 234–244.
Efficient nanofibrous membranes for antibacterial wound dressing and UV protec- [53] M. Xie, L. Wang, J. Ge, B. Guo, P.X. Ma, Strong electroactive biodegradable shape
tion, ACS Appl. Mater. Inter. 8 (44) (2016) 29915–29922. memory polymer networks based on star-shaped polylactide and aniline trimer for
[26] A. Nasajpour, S. Ansari, C. Rinoldi, A.S. Rad, T. Aghaloo, S.R. Shin, Y.K. Mishra, bone tissue engineering, ACS Appl. Mater. Inter. 7 (12) (2015) 6772–6781.
R. Adelung, W. Swieszkowski, N. Annabi, A. Khademhosseini, A. Moshaverinia, [54] I.A. Darby, B. Laverdet, F. Bonté, A. Desmoulière, Fibroblasts and myofibroblasts in
A. Tamayol, A multifunctional polymeric periodontal membrane with osteogenic wound healing, Clin. Cosmet. Investig. Dermatol. 7 (2014) 301–311.
and antibacterial characteristics, Adv. Funct. Mater. 28 (3) (2018) 1703437. [55] L. Li, J. Ge, B. Guo, P.X. Ma, In situ forming biodegradable electroactive hydrogels,
[27] R. Nudelman, H. Alhmoud, B. Delalat, S. Fleicher, E. Fine, T. Guliakhmedova, Polym. Chem.-UK 5 (8) (2014) 2880–2890.
R. Elnathan, A. Nyska, N.H. Voelcker, M. Gozin, S. Richter, Jellyfish-based smart [56] D. Mawad, E. Stewart, D.L. Officer, T. Romeo, P. Wagner, K. Wagner, G.G. Wallace,
wound dressing devices containing in situ synthesized antibacterial nanoparticles, A single component conducting polymer hydrogel as a scaffold for tissue en-
Adv. Funct. Mater. 1902783 (2019). gineering, Adv. Funct. Mater. 22 (13) (2012) 2692–2699.
[28] F. Xu, B. Weng, R. Gilkerson, L.A. Materon, K. Lozano, Development of tannic acid/ [57] M. Zhang, B. Guo, Electroactive 3D scaffolds based on silk fibroin and water-borne
chitosan/pullulan composite nanofibers from aqueous solution for potential appli- polyaniline for skeletal muscle tissue engineering, Macromol. Biosci. 17 (9) (2017)
cations as wound dressing, Carbohydr. polym. 115 (2015) 16–24. 1700147.
[29] R. Augustine, E.A. Dominic, I. Reju, B. Kaimal, N. Kalarikkal, S. Thomas, [58] M. Yang, Y. Liang, Q. Gui, J. Chen, Y. Liu, Electroactive biocompatible materials for

12
J. He, et al. Chemical Engineering Journal 385 (2020) 123464

nerve cell stimulation, Mater. Res. Express 2 (4) (2015) 042001. healing by electroactive, antibacterial, and antioxidant polyurethane/siloxane
[59] C. Agyare, A.S. Dwobeng, N. Agyepong, Y.D. Boakye, K.B. Mensah, P.G. Ayande, dressing membranes: in vitro and in vivo evaluations, ACS Appl. Mater. Interfaces 7
M. Adarkwa-Yiadom, Antimicrobial, antioxidant, and wound healing properties of (43) (2015) 24296–24311.
kigelia africana (Lam.) Beneth. and Strophanthus hispidus DC, Adv. Pharmacol. Sci. [70] T.H. Qazi, R. Rai, A.R. Boccaccini, Tissue engineering of electrically responsive
2013 (2013) 10. tissues using polyaniline based polymers: a review, Biomaterials 35 (33) (2014)
[60] D. Gopinath, M.R. Ahmed, K. Gomathi, K. Chitra, P.K. Sehgal, R. Jayakumar, 9068–9086.
Dermal wound healing processes with curcumin incorporated collagen films, [71] H.-T. Chiu, R.-L. Chen, P.-Y. Wu, T.-Y. Chiang, S.-C. Chen, A study on the effects of
Biomaterials 25 (10) (2004) 1911–1917. the degree of deacetylation of chitosan films on physical and antibacterial prop-
[61] H. Cui, L. Cui, P. Zhang, Y. Huang, Y. Wei, X. Chen, In situ electroactive and an- erties, Poly-Plast. Technol. 46 (12) (2007) 1121–1127.
tioxidant supramolecular hydrogel based on cyclodextrin/copolymer inclusion for [72] M. Gizdavic-Nikolaidis, S. Ray, J.R. Bennett, A.J. Easteal, R.P. Cooney, Electrospun
tissue engineering repair, Macromol. Biosci. 14 (3) (2014) 440–450. functionalized polyaniline copolymer-based nanofibers with potential application
[62] V. Kant, A. Gopal, N.N. Pathak, P. Kumar, S.K. Tandan, D. Kumar, Antioxidant and in tissue engineering, Macromol. Biosci. 10 (12) (2010) 1424–1431.
anti-inflammatory potential of curcumin accelerated the cutaneous wound healing [73] M. Li, J. Chen, M. Shi, H. Zhang, P.X. Ma, B. Guo, Electroactive anti-oxidant
in streptozotocin-induced diabetic rats, Int. Immunopharmacol. 20 (2) (2014) polyurethane elastomers with shape memory property as non-adherent wound
322–330. dressing to enhance wound healing, Chem. Eng. J. 375 (2019) 121999.
[63] Y. Liang, X. Zhao, T. Hu, B. Chen, Z. Yin, P.X. Ma, B. Guo, Adhesive hemostatic [74] C. Gong, Q. Wu, Y. Wang, D. Zhang, F. Luo, X. Zhao, Y. Wei, Z. Qian, A biode-
conducting injectable composite hydrogels with sustained drug release and pho- gradable hydrogel system containing curcumin encapsulated in micelles for cuta-
tothermal antibacterial activity to promote full-thickness skin regeneration during neous wound healing, Biomaterials 34 (27) (2013) 6377–6387.
wound healing, Small 15 (12) (2019) 1900046. [75] X. Liu, T. Lin, J. Fang, G. Yao, H. Zhao, M. Dodson, X. Wang, In vivo wound healing
[64] İ. Tümen, E.K. Akkol, H. Taştan, I. Süntar, M. Kurtca, Research on the antioxidant, and antibacterial performances of electrospun nanofibre membranes, J. Biomed.
wound healing, and anti-inflammatory activities and the phytochemical composi- Mater. Res. A 94 (2) (2010) 499–508.
tion of maritime pine (Pinus pinaster Ait), J. Ethnopharmacol. 211 (2018) 235–246. [76] B. Balakrishnan, M. Mohanty, P.R. Umashankar, A. Jayakrishnan, Evaluation of an
[65] J.-G. Leu, S.-A. Chen, H.-M. Chen, W.-M. Wu, C.-F. Hung, Y.-D. Yao, C.-S. Tu, Y.- in situ forming hydrogel wound dressing based on oxidized alginate and gelatin,
J. Liang, The effects of gold nanoparticles in wound healing with antioxidant epi- Biomaterials 26 (32) (2005) 6335–6342.
gallocatechin gallate and α-lipoic acid, Nanomed.-Nanotechnol. 8 (5) (2012) [77] P. Mostafalu, G. Kiaee, G. Giatsidis, A. Khalilpour, M. Nabavinia, M.R. Dokmeci,
767–775. S. Sonkusale, D.P. Orgill, A. Tamayol, A. Khademhosseini, A textile dressing for
[66] K. Gouthamchandra, R. Mahmood, H. Manjunatha, Free radical scavenging, anti- temporal and dosage controlled drug delivery, Adv. Funct. Mater. 27 (41) (2017)
oxidant enzymes and wound healing activities of leaves extracts from 1702399.
Clerodendrum infortunatum L, Environ. Toxicol. Phap. 30 (1) (2010) 11–18. [78] C.G. Decker, Y. Wang, S.J. Paluck, L. Shen, J.A. Loo, A.J. Levine, L.S. Miller,
[67] P.T. Kumar, V.K. Lakshmanan, T.V. Anilkumar, C. Ramya, P. Reshmi, H.D. Maynard, Fibroblast growth factor 2 dimer with superagonist in vitro activity
A.G. Unnikrishnan, S.V. Nair, R. Jayakumar, Flexible and microporous chitosan improves granulation tissue formation during wound healing, Biomaterials 81
hydrogel/nano ZnO composite bandages for wound dressing: in vitro and in vivo (2016) 157–168.
evaluation, ACS Appl. Mater. Inter. 4 (5) (2012) 2618–2629. [79] D.D. Cissell, J.M. Link, J.C. Hu, K.A. Athanasiou, A modified hydroxyproline assay
[68] W.A. Sarhan, H.M. Azzazy, I.M. El-Sherbiny, Honey/chitosan nanofiber wound based on hydrochloric acid in ehrlich's solution accurately measures tissue collagen
dressing enriched with Allium sativum and Cleome droserifolia: enhanced anti- content, Tissue Eng. Part C Methods 23 (4) (2017) 243–250.
microbial and wound healing activity, ACS Appl. Mater. Inter. 8 (10) (2016) [80] T. Kondo, Y. Ishida, Molecular pathology of wound healing, Forensic Sci. Int. 203
6379–6390. (1) (2010) 93–98.
[69] R. Gharibi, H. Yeganeh, A. Rezapour-Lactoee, Z.M. Hassan, Stimulation of wound

13

You might also like