2020 - PVA Chitosan Espun Fibers

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Carbohydrate Polymers 232 (2020) 115786

Contents lists available at ScienceDirect

Carbohydrate Polymers
journal homepage: www.elsevier.com/locate/carbpol

Wound dressing from polyvinyl alcohol/chitosan electrospun fiber T


membrane loaded with OH-CATH30 nanoparticles
Pengfei Zoua, Wen-Hui Leeb,c, Zhiqin Gaoc, Di Qinc, Yuxia Wanga, Jiao Liua, Tongyi Sunc,**,
Yuanyuan Gaoa,*
a
School of Pharmacy, Weifang Medical University, Weifang, 261053, Shandong, China
b
Key Laboratory of Bioactive Peptide of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences,
Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, Yunnan, China
c
Key Laboratory of Biological Medicine in Universities of Shandong Province, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053,
Shandong, China

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

Keywords: Novel nanomaterials have been developed for antimicrobial and wound healing applications. Here, we report the
OH-CATH30 preparation of a polyvinyl alcohol/chitosan (PVA/CS) nanofiber with carboxymethyl chitosan nanoparticles
PVA (CMCS-OH30 NPs) encapsulating the antibacterial peptide OH-CATH30 (OH-30). The PVA/CS nanofibers con-
Wound healing taining OH-30 NPs (NP-30-NFs) obtained via electrospinning could achieve a secondary embedded OH-30. The
Nanofibers
effect of NP-30-NFs on the release of OH-30 was investigated through high-performance liquid chromatography.
Nanoparticles
The antibacterial activities of NP-30-NFs against Escherichia coli and Staphylococcus aureus were studied by
bacterial plate counting. NP-30-NFs containing different concentrations of NPs were applied to mouse skin
wounds to determine their effectiveness in promoting wound healing. Results showed that NP-30-NFs exhibited
antibacterial properties and promoted skin wound healing.

1. Introduction and cytokine production without eliciting harmful immune responses to


induce a regulatory effect on inflammatory responses and to treat
Surgery, burns, abrasions, and chronic ulcers are among the key various bacterial infections (Li et al., 2014; Zhang et al., 2010; Zhao
factors that cause skin trauma (Sun, Siprashvili, & Khavari, 2014). Skin et al., 2018). Notably, the dynamic balance of inflammatory response
damage is often accompanied with self-repair of the skin. These repair and reduced inflammation is an important guarantee for tissue repair
processes are essential for organismal homeostasis and survival (Rankin (Eming, Krieg, & Davidson, 2007; Rankin & Artis, 2018). AMPs, as
& Artis, 2018). Skin tissue repair is a dynamic and complex process that antibacterial agents, have great potential for wound dressing applica-
involves inflammation, angiogenesis, granulation tissue growth, foreign tions (Gomes, Mano, Queiroz, & Gouveia, 2015). However, limited
body reaction, and re-epithelialization (Gao et al., 2018; Zhou et al., stability and bioavailability restrict their applications because AMPs are
2016). Unfortunately, wound infections can easily occur during wound easily hydrolyzed by proteases (Silva et al., 2015). Therefore, effec-
healing, thereby delaying the healing process. Therefore, bacterial in- tively avoiding the enzyme degradation of AMPs and accurately con-
fection is a major concern in the treatment of open wounds and burns trolling their release are desired for active peptide nanomaterials (Silva
(Akia, Rodriguez, Materon, Gilkerson, & Lozano, 2019). et al., 2015). A drug delivery carrier should be developed to protect
Antibacterial peptides (AMPs) of natural origin may be clinical active peptides from proteases and release these peptides. We reported
candidates because of their broad-spectrum antibacterial activity, dif- that our prepared biodegradable drug nanoparticle (NP) CMCS-OH-30
ficulty in causing drug resistance, and regulatory effect on the body’s delivery system can promote OH-30 sustained release. Our results also
immune system. As a possible candidate drug against clinical drug-re- demonstrated that the prepared CMCS-OH-30 NPs accelerate the
sistant isolates (Li, Lee, & Zhang, 2012, 2013), cathelicidin OH-CATH30 healing of skin lesions and reduce scars on mice (Sun et al., 2018).
(OH-30) derived from king cobra selectively upregulates chemokine The application of occlusive wound dressings is a routine external


Corresponding author at: School of Pharmacy, Weifang Medical University, Baotong Road, Weifang, 261053, Shangdong, China.
⁎⁎
Corresponding author at: Key Laboratory of Biological Medicine in Universities of Shandong Province, School of Bioscience and Technology, Weifang Medical
University, Baotong Road, Weifang, 261053, Shangdong, China.
E-mail addresses: sd_sty@126.com (T. Sun), yyg20062006@126.com (Y. Gao).

https://doi.org/10.1016/j.carbpol.2019.115786
Received 23 March 2019; Received in revised form 12 December 2019; Accepted 26 December 2019
Available online 28 December 2019
0144-8617/ © 2020 Elsevier Ltd. All rights reserved.
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

intervention that helps accelerate re-epithelialization and alters the 2.2. Preparation of CMCS
inflammatory milieu to favor healing (Sun et al., 2014). Traditional
wound dressings, such as gauze, bandages, and sponges, are used to CMCS preparation was slightly modified in accordance with a pre-
prevent bacterial invasion, but limited swelling capacity restricts their viously described method (Jiang, Han, Li, Yang, & Liu, 2015). In brief,
applications (Fu et al., 2016). Modern wound dressings, such as hy- 2.5 g of CS was accurately weighed and dissolved in 45 mL of 40 % (w/
drogels and nanofibers produced by electrospinning active components w) NaOH solution and stirred overnight. CS was filtered and dissolved
derived from natural sources, have been widely explored to overcome in 80 % (v/v) isopropanol solution in a three-necked flask and a con-
this issue in regenerative medicine and trauma (Zhou, Sui, Mo, & Sun, densing device. Within 30 min, 15 mL of 50 % chloroacetic acid in
2017). However, biological dressing that creates a moist environment isopropanol solution was added dropwise, and the reaction was carried
to promote wound healing is also an ideal place for the colonization and out in a water bath at 50 °C for 4 h. A total of 40 % NaOH solution was
reproduction of pathogenic microbes (Pei, Sun, Sun, Wang, & Zhao, added to adjust pH to approximately 8.0. At the end of the reaction, the
2015). Therefore, effectively reducing bacterial infection and creating a sample was suction filtered, washed with 95 % ethanol solution six
relatively sterile microenvironment are important prerequisites for times, and dried prior to futher use.
developing an effective wound dressing. Stimulus-responsive CS and
poly (N-vinyl-2-pyrrolidone) (PVP) hydrogels for wound healing ap-
2.3. Preparation and evaluation of CMCS-OH-30 NPs
plication exhibit antibacterial activity (Rasool, Ata, & Islam, 2019), and
injectable hydrogels (Xan-CHO and NOCC) accelerate the reconstruc-
CMCS-OH-30 NPs were prepared via the electrostatic droplet
tion of the abdominal wall in rats (Huang et al., 2018). In addition,
method in accordance with previously described methods (Sun et al.,
wound dressings containing antibiotics (Garcia et al., 2017), non-
2018) with slight modifications. In brief, OH-30 was dissolved in
steroidal anti-inflammatory drugs, such as ibuprofen (Morgado, Miguel,
deionized water, and the previously prepared CMCS was dissolved in an
Correia, & Aguiar-Ricardo, 2017), titanium dioxide (El-Aassar, El
appropriate amount of deionized water (OH-30:CMCS = 1:2). OH-30
Fawal, El-Deeb, Hassan, & Mo, 2016), and silver nanoparticles (Ag-NPs)
solution was aspirated into a sterile syringe added with 0.65 kV positive
(Das, Kumar, Patil, Viswanathan, & Ghosh, 2015; Kohsari, Shariatinia,
voltage clamped on an electrospinning instrument (Beijing, China). OH-
& Pourmortazavi, 2016; Lee et al., 2014; Mokhena & Luyt, 2017), have
30 solution was pushed into the CMCS solution at a bolus rate of
shown good results against bacteria and are beneficial to wound
0.5 mm/min with magnetic stirring (250 rpm) until completion. After
healing.
the solution was stirred for 10 min, the sample was placed in an ul-
Electrospun nanofibers have been considered as applicable mate-
trasonic ice bath for 5 min at approximately 20 W for 3 s and stopped
rials for wound dressing applications because of their intrinsic prop-
for 2 s. The mixture was centrifuged at 12,000 rpm at 4 °C. After the
erties, such as high surface-area-to-volume ratio and swelling (Miguel
supernatant was removed, the precipitate was resuspended in deionized
et al., 2019). Nanofibers in these membranes can act as drug delivery
water, and ultrasonication was repeated. The prepared NPs were pre-
systems (Gao et al., 2016), which prompt the incorporation of biomo-
cooled for 24 h at −80 °C before they were freeze dried.
lecules within the fiber structure to prevent skin infections and improve
The particle size distribution of CMCS-OH-30 NPs was determined
healing (El-Aassar et al., 2016; Miguel et al., 2019). CS is a biode-
using a Malvern laser scattering particle size analyzer (Nano-ZS90,
gradable natural polysaccharide that promotes blood clotting and pain
Malvern, UK), and their morphological characteristics were observed
relief (Charensriwilaiwat et al., 2012). PVA is a water-soluble polymer
using a transmission electron microscope (JEM-2100; JEOL, Akishima-
widely used in wound dressings because of its excellent film-forming
shi, Japan). The encapsulation efficiency (EE) of CMCS-OH30 NPs was
and mechanical properties (Fu et al., 2016; Zhao, Chen, Yao, & Li,
identified. In brief, the free Cy5-labeled OH-30 released from CMCS-
2017). Besides, blended fiber membranes composed of PVA and CS are
OH30 NP in the supernatants was evaluated using a fluorescence ana-
extensively used in various fields, such as tissue engineering scaffolds,
lyzer (F-4600, Japan Corporation, Japan). EE was calculated with the
drug delivery (Miguel et al., 2019; Zarandi et al., 2015), new wound
following Eq. (1):
dressings (Zhao et al., 2017; Zhou et al., 2013), heavy metal adsorption
(Habiba, Afifi, Salleh, & Ang, 2017), and air filtration sterilization EE = (Wt − Wn) / Wt × 100, (1)
(Wang et al., 2018). Therefore, a PVA/CS system is expected to be a
where Wt and Wn refer to the weights of the total OH-30 used and the
good carrier of OH-30 and beneficial to the sustained release of re-
nonencapsulated OH-30, respectively (n = 3).
agents.
In this study, NP-30-NFs were used as dressing for mouse skin
wound to test the applicability of PVA/CS nanofibers as a drug delivery 2.4. Preparation of NP-30-NFs
carrier of OH-30 and wound dressing material via FTIR, SEM and LSCM.
We envisaged that NP-30-NFs could further enhance the bioavailability PVA/CS nanofibers with a CS concentration of 20 % or higher have
and stability of OH-30. The release behaviour of OH-30 in NP-30-NFs a bactericidal effect (Abdelgawad, Hudson, & Rojas, 2014). Nanofibers
was evaluated compared with CMCS−OH-30 NPs. In addition, anti- containing 10 % CS were prepared to eliminate the bactericidal action
bacterial tests and mouse wound healing experiments were also eval- of the nanofiber itself and highlight the antibacterial effect of AMPs.
uated. These characteristics showed the advantage of using the nano- The preparation method of the electrospun fiber membrane was slightly
fibers as wound dressing. modified on the basis of previously studied equations. Subsequently, 1
% (w/v) CS was dissolved in 15 % (v/v) acetic acid solution, and 9 %
2. Materials and methods (w/v) PVA was dissolved in deionized water and stirred at 95 °C for
1.5 h at a constant temperature. After the PVA solution was cooled to
2.1. Materials 25 °C, different final concentrations (i.e., 0.08 %, 0.12 %, and 0.27 %
w/v) of NPs were slowly added to CS, continuously stirred for 0.5 h and
OH-CATH30 (amino acid sequence, KFFKKLKNSVKKRAKKFFKKP- transferred to the PVA solution. The bubbles were then allowed to
RVIGVSIPF; purity > 96 %) was synthesized by GL Biochem (Shanghai, stand. The hydrogel solution was aspirated into a 5 mL sterile syringe
China). PVA (average molecular weight of 5.9―7.1 kDa) with a degree for electrospinning. The nanofiber filaments formed by the spinning
of polymerization of 1750 ± 50 was bought from Sinopharm Chemical solution jet were received by a foil wrapped receiver at a positive
Reagent Corporation (Shanghai, China). CS (average molecular weight voltage for 24 h. The distance between the nozzle (1 mm) and the re-
of 60―120 kDa) with a degree of deacethylation of > 95 % was pur- ceiver was 23 cm, the height of the nozzle was 39 ± 5 cm, the speed of
chased from Aladdin Industrial Corporation (Shanghai, China). the spinning solution was 0.03 ± 0.01 mm/min, the receiving voltage

2
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

was −1.44 ± 0.5 kV, the experimental temperature was room tem- 2.10. Antimicrobial assay
perature, and the relative humidity was 40 % ± 5 %. Pure nanofiber
(Pure) without NPs served as a control. The nanofibers were heated at Escherichia coli (ATCC 25922) and Staphylococcus aureus (ATCC
40 °C for 12 h and then stored in a vacuum-drying oven. 25923) were selected as test strains for the antibacterial activity test of
electrospun fibrin membranes (Pei et al., 2015). The two kinds of
2.5. Scanning electron microscopy (SEM) analysis bacteria were pre-activated for two generations, inoculated into Lur-
ia―Bertani (LB) liquid medium, and cultured at 37 °C for 4–6 h until
The samples were gold coated using a sputter coater. The structure the bacterial concentration reached 5 × 104 colony-forming units/mL.
inside the electrospun fiber membrane was observed through SEM After centrifugation (4000 rpm, 4 °C) was conducted for 10 min, the
(TESCAN, VEGA3, CZ). bacterial suspension was resuspended with a fresh medium. Different
weights of nanofiber membranes (1–10 mg) were transferred in an
2.6. Fourier transform infrared spectroscopy (FTIR) analysis Eppendorf tube containing 1 mL of bacterial suspension. The optical
density (OD) at a wavelength of 570 nm was measured using a micro-
The solid sample was prepared using a potassium bromide tableting plate reader (Bio Tek, ELX800, USA) after 24 h of incubation at 37 °C.
method. An appropriate amount of the sample was placed in an agate The assay was performed in triplicate.
mortar with potassium bromide and dried overnight in an oven at a
constant temperature of 60 °C. The mixture was added to a tableting 2.11. Cytotoxicity of the electrospun fiber membrane
mold, pressed to a transparent pressure under the pressure of a ta-
bleting machine, and detected by FTIR (Thermo, IS10, Shanghai, The cytotoxicity of nanofiber mats was evaluated following a pre-
China). All of the operations were performed in an infrared oven to viously described method (Moura et al., 2014). The nanofiber mat was
prevent moisture absorption. The background of potassium bromide sterilized in a UV environment for 30 min. The nanofiber membrane
was deducted as a blank. Each sample was tested three times. was then immersed in a serum-free medium (MEM, 1 % v/v penicillin
and streptomycin, 1 % v/v non-essential amino acids and 1 % v/v so-
2.7. Laser scanning confocal microscopy (LSCM) analysis dium pyruvate) and cultured at 25 °C for 24 h to produce different
concentrations of extraction media (0.25, 0.5, 1, 3, 5, 7, and 9 mg/mL).
The preparation method was the same as Sections 2.2 and 2.3, but Afterward, 100 μL of HaCaT cells was seeded in 96-well plates (Corning
the antimicrobial peptide was rhodamine B-labeled OH-30. A certain Incorporated, Corning, NY, USA) at a density of 5 × 104 cells/mL. After
area of the cut nanofiber membrane was placed on a slide and observed 48 h of culture, MEM supplemented with 10 % fetal bovine serum (FBS)
under LSCM (Leica TCS SP8, Wetzlar, DE, USA). The fluorescence on was aspirated, and the cells were incubated with different concentra-
the 50 μm × 30 μm × 20 μm nanofiber membrane with 0.01 % OH-30 tions of the extraction medium for 12 h. After the treatment was ad-
NPs was observed to determine the drug distribution on the fiber ministered, the cells were incubated with 10 μL of MTT (5 mg/mL) for
membrane. 4 h. Subsequently, 100 μL of dimethyl sulfoxide was added to each well.
After 20 min of incubation, absorbance was determined at a wavelength
2.8. Degree of swelling of 490 nm by using a microplate reader to calculate the relative cell
viability (%). The viability of untreated control cells was defined as 100
The swelling of the sample was quantified in accordance with pre- %.
viously described methods (Arthanari et al., 2016; Charernsriwilaiwat,
Rojanarata, Ngawhirunpat, & Opanasopit, 2012) with slight modifica- 2.12. Wound healing activity of electrospun fiber membranes
tions. The samples were swelled into PBS (pH 7.4) at 37 °C for 2, 4, 6, 8,
10, and 12 h. The swelling ratio was determined with Eq. (2) as follows: Five-week-old female specific pathogen-free (SPF)-grade KM mice
(average body weight of 20–25 g, 12 per group) were obtained from
Degree of swelling (%) = (W − Wd) / Wd × 100, (2)
Jinan Pengyue Animal Breeding Co., Ltd. The mice were first anesthe-
where W is the wet weight of the sample after it was immersed in PBS, tized by intraperitoneally injecting chloral hydrate (0.1 mL/10 g).
and Wd is the initial dry weight of the sample. Depilatory cream was used to clean the back skin of the mice. A Miltex®
Biopsy Punch (ID = 7 mm) with a plunger was placed perpendicular to
2.9. Drug release experiments the back skin and cut by rotation. Surgical scissors were used to remove
the intermediate skin to create a round wound with full thickness. The
In our controlled drug release study, the highest concentration of mice were randomized into six groups: untreated group, un-
the nanofiber membrane (0.27 %) was selected as the representative. A encapsulated drug group, 0.08 % NPs group, 0.12 % NPs group, 0.27 %
drug-loaded nanofiber mat (70 mg) was soaked in a centrifuge tube NPs group and positive control group (JINCHUANG® chitosan wound
containing 10 mL of PBS (pH 7.4) (Chen et al., 2015). The tube was care film). A 1 cm × 1 cm dressing was applied to the wound, replaced
incubated in a thermostat shaker at 37 °C and a rock speed of 100 rpm. every 24 h, and continuously changed for 5 days. Each mouse was in-
At an appropriate time, 1 mL of the release medium was removed from dividually placed in a squirrel cage of an SPF animal house to prevent
the centrifuge tube and replaced with an equal volume of fresh deio- mutual biting and the dressing from falling off. Each wound was ob-
nized water. The sample was analyzed through a high-performance li- served and photographed using a Nikon camera to record the wound
quid chromatograph (HPLC) to determine OH-30 by using an injection healing process. Three mice were then sacrificed on days 7 and 21
amount of 20 μL in a HPLC series system (Agilent 1260II, USA) with a postoperatively, and the samples were obtained for subsequent hema-
C18 column (120-EC, 4.6 mm × 15 cm, 4 μm, InfinityLab Poroshell). toxylin―eosin (HE) staining experiments. The wound healing rate was
The released OH-30 was detected at 280 nm. The mobile phase was calculated using Eq. (3):
acetonitrile (ACN): water (H2O). The gradient was first maintained for
Wound contraction (%) =(A0-At) / A0 × 100 %, (3)
5 min at 80 % H2O/0.1 % trifluoroacetic acid (TFA) and 20 % ACN/0.1
% TFA followed by 20 min at 60 % H2O/0.1 % TFA and 40 % ACN/0.1 where Ao is the original wound wound area, and At is the wound area of
% TFA, and the flow rate was set to 1 mL/min. The average was ob- tissue examination on day t. The excised skin tissue was fixed with 4 %
tained from three parallel samples. The amount of NPs released was paraformaldehyde tissue fixative for 24 h, rinsed, dehydrated, and
used as the control. embedded in paraffin. Sections were stained with HE stain and

3
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

Fig. 1. TEM micrographs showing the morphological features of CMCS-OH-30 NPs (A).
SEM images of nanofibers with different NP concentrations. Pure PVA/CS nanifibers (B), 0.08 % (w/v) NP-30-NFs (C), 0.12 % (w/v) NP-30-NFs (D), 0.27 % (w/v)
NP-30-NFs (E), and average diameter comparison (F) (n = 100).

horseshoe stain and photographed at 100× magnification. The animal p > 0.05 (ns) was considered not statistically significant.
protocol for this study was approved by the animal care and ethical
committee of the medical sector in Weifang Medical University. 3. Results and discussion

2.13. Statistical analysis 3.1. NP characterization

Data were collected and presented as the mean and standard error The cationic antibacterial peptide OH-30 could be ionically cross-
of the mean. The experimental data in different groups were compared linked with CMCS containing many carboxyl groups because of its
using one-way ANOVA in SPSS 15.0 (SPSS Inc., Chicago, IL, USA). chargeability. As such, it could self-assemble into NPs. The TEM pho-
*p < 0.05 and **p < 0.01 were considered statistically significant. tographs (Fig. 1A) and particle size distribution (Table 1) of CMCS-OH-

4
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

Table 1 nanofibers. As shown in the CS spectrum, the main characteristic peaks


Quality evaluation of CMCS-OH-30 NPs (n = 3). were located at 3441.91 (OeH stretching vibration and NeH stretching
Size (nm) PDI Zeta potential EE (%) vibration), 2873.41 (C–H stretching vibration), 1597.11 (amide CeO
(mV) stretching), and 1077.56 cm−1 (primary hydroxyl, CeO tensile vibra-
tion). By contrast, the CMCS spectrum showed the characteristic peak
−37.6 ± 1.5
Mean ± SD 164.6 ± 5.0 0.14 ± 0.02 92.14 ± 1.05
(1595.34 cm−1) of relevant carboxylate (−COO- not symmetrical
stretching) and the characteristic peak of carboxymethyl group
(1411.89 cm−1) (Mishra et al., 2011). The symmetrical stretching of the
30 NPs are shown. In the TEM images, NPs were relatively round, and
carboxylate CeO indicated the presence of carboxymethyl groups in
their particle size was 164.6 ± 5.0 nm, thereby facilitating re-coating
CMCS. The main characteristic peak of OH-30 at 3284.98 cm−1 (OeH
for the subsequent electrospinning. The polydispersity index indicated
stretching vibration and NeH stretching vibration) became a broad and
that the particle size distribution was uniform. The Z potential de-
blunt peak at 3296.38 cm−1 after OH-30 was synthesized into NPs and
monstrated that the particles did not easily settle, coagulate, or floc-
found in the fingerprint region. The multiple (−CH2)-in-plane bending
culate. The entire system was relatively stable and could meet the
vibration at 721.71 cm−1 was successfully masked. These findings in-
stability requirement (> 15 mV) possibly because of the following
dicated that CMCS and OH-30 were successfully crosslinked by inter-
factors. A decrease in the surface tension of the droplets was due to the
molecular interactions. After the NPs were recoated, the peak of
application of a certain unit of positive charges on the OH-30 solution,
3421.89 cm−1 was masked by the COO- of the PVA, showing a sharp
thereby causing the solution to drip into the low CMCS solution in a
peak (Habiba, Siddique et al., 2017; Zhou et al., 2017).
uniform stable state. The application of a positive charge might have
The 1H NMR spectra of CMCS are presented in Fig. S1. The spectra
distributed the originally disordered OH-30 molecules around the so-
of CMCS illustrate that the chemical shifts at 4.4 ppm were due to the
lution under the action of an electric field, thereby increasing the
protons of −CH2COO− at the O-position at C6 of CMCS. The results
contact area between OH-30 and CMCS molecules and ion crosslinking.
showed that the carboxylated carboxymethyl chitosan at the C6 posi-
Thus, the EE of the NPs prepared by the electrostatic droplet method
tion was successfully synthesized (Mi, Su, Fan, Zhu, & Zhang, 2013).
was improved compared with that of the extrusion method (Sun et al.,
2018).
3.2.4. Degree of swelling
Swelling is one of the important properties of nanofiber membranes
3.2. Nanofiber characterization for wound dressing applications (Arthanari et al., 2016). A wound
dressing with a good degree of swelling contributes to the absorption of
3.2.1. SEM analysis permeate from wound tissues, creating a moist microenvironment that
The SEM image shows electrospun nanofibers, suggesting that the promotes wound healing. The degree of swelling of nanofibers plays an
surface of the nanofiber filaments for each sample was smooth important role in the loading and release behavior of the drug. Fig. 4A
(Fig. 1B–E). Fig. 1F illustrates the average diameter of the electrospun shows the degree of swelling of various fiber membranes of different
fiber membranes with different NPs. A total of 100 fibers were analyzed NPs after they were soaked in PBS at 37 °C. The hydroxyl group on the
with Image Pro 8.0, and histograms were plotted. The average fiber PVA molecule can interact with the amino group and the hydroxyl
diameter was primarily determined in terms of solution viscosity and group on the CS molecule through intermolecular hydrogen bonding; at
conductivity (Haider, Haider, & Kang, 2018). The mean diameter of the the same time, CMCS can form a hydrogen bond with CS. In this study,
control group (mean ± SD) was 357.34 ± 110.45 nm, which de- the other components exhibited excellent swelling properties compared
creased from 371.80 ± 110.31 nm to 327.48 ± 114.28 nm after the with the control group, and the degree of swelling exceeded 100 %. The
drug was loaded. Given that the concentration of NPs in the solution degree of swelling of the nanofibers was related to the NP concentra-
was high, the uniformity of the fiber diameter was less maintained tion. This result might be attributed to the increased amount of CMCS in
possibly because the viscosity and conductivity of the original spinning the NPs, promoting the degree of crosslinking in the PVA/CS solution.
solution changed as the CMCS-OH30 NPs increased (Table S1). As the However, in all cases, the maximum swelling was attained at 4–6 h
concentration of the NPs increased, the viscosity of the PVA/CS system beyond which the degree of swelling was reduced. This behavior might
also increased, and the NPs had a certain unit of negative charge that be due to the release of OH-30 and the degradation or leaching of
increased their conductivity. A high concentration of the fiber mem- polymeric composites. The maximum swelling of gatifloxacin-loaded
brane exhibited a small amount of bead structures possibly because of alginate/PVA electrospun nanofibers was attained at 8 h (Arthanari
an increase in surface tension resulting from uneven jetting and diffi- et al., 2016). Thus, the faster the swelling, the more conducive the
culty in forming a Taylor cone. In terms of diameter, the particle size of creation of a relatively clean wound environment. This phenomenon
NPs was much smaller than the diameter of the electrospun fiber accelerated wound healing.
membrane. Thus, the NPs could be encapsulated by the fiber mem-
brane. Charernsriwilaiwat et al. (2012) also confirmed that viscosity 3.3. In vitro drug release studies
and conductivity are important factors affecting the diameter of nano-
fibers. Fig. 4B shows the in vitro release of OH-30 from NPs and nanofibers.
The prepared nanofibers exhibited good sustained release properties in
3.2.2. LSCM of NP-30-NFs vitro. The average cumulative release of OH-30 released from the na-
After a full-layer scan of the prepared NP-30-NFs, the results were nofibers was 14 % in the first 2 h and sustained release was observed for
intercepted (Fig. 2). Red fluorescence showed the NP distribution in any at least 24 h. The final cumulative release was 66 % ± 7.5 % in 24 h.
direction of the fiber membrane, indicating that NPs were successfully The release mechanism of the prepared nanofibers might be caused by
entrapped in the fiber membrane. The fiber membrane surface was the following scenarios. 1. The swelling degree of the nanofibers in-
smooth as observed under the optical microscope. The NPs were en- creased and the intermolecular gap increased with time, providing a
capsulated in the fiber membrane rather than irregularly adsorbed onto spatial basis for NP release. 2. The degree of ionic crosslinking between
the nanofiber surface. Nylon 6 nanofibers immersed in citrus juice so- OH-30 and nanocarriers weakened, and OH-30 was released.
lution for 5 min can eradicate bacteria (Akia et al., 2019).
3.4. Antimicrobial assay
3.2.3. FTIR analysis
Fig. 3 illustrates the FTIR spectra of CS, CMCS, OH-30 NPs, and The results of antibacterial experiments showed that the electrospun

5
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

Fig. 2. LSCM image of 0.27 % NP-30-NFs under different microscopes. Fluorescence microscope (A), o.ptical microscope (B), and combination of the two (C).

Fig. 4. Degree of swelling (%) of PVA/CS electrospun fiber membranes (A). OH-
30 conc. (0, 0.08 %, 0.12 %, 0.27 %) (n = 3).
In vitro release profile of OH-30 (plotted as a function of percentage cumulative
release vs. time) from nanofibers and CMCS-OH30 NPs at different time (B)
(n = 3).

Fig. 3. FTIR spectra of CMCS, CS, OH-30, NPs, PVA, and 0.27 % NP-30-NFs. Thus, an electrospun fiber membrane loaded with NPs could better
provide a relatively sterile microenvironment compared with a pure
electrospun fiber membrane. This phenomenon was beneficial to
fiber membrane loaded with NPs had superior antibacterial properties
wound healing.
compared with the simple electrospun fiber membranes (Fig. 5). This
finding was consistent with our previous research results (Sun et al.,
2018). When the concentration of the electrospun fiber membrane
carrying NPs reached 3 mg/mL, the inhibition rate was 80 % or more.

6
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

Fig. 5. The antibacterial activity of the nanofiber mats against (A) E. coli and (B) S. aureus after 24 h of incubation at 5 × 104 CFU/mL bacteria, 37℃ (n = 3).

could be released slowly from the nanofibers than CMCS-OH-30 NPs


during the healing process, which could against bacterias and accel-
erate wound healing. There is one advantage in using this nanofibers as
the wound dressing.
At day 12, 98.51 ± 0.37 % wound closure was observed in 0.27 %
NP-30-NFs while the chitosan/PVA/ZnO nanofibrous membranes
showed amuch lower percentage of contraction namely 90.5 ± 1.7 %
wound closure only (Ahmed et al., 2018). Consistent with our previous
findings, the present results indicated that the constructed OH-30 de-
livery system played an important role in the early wound healing
process.
Fig. 8A shows the histopathological analysis of the healing effects of
Fig. 6. Cytotoxicity assay of HaCaT cells. Cells and different concentrations of the samples stained with HE. The 7-day histopathological results
nanofiber membranes extracts were incubated for 12 h at 37℃ (n = 3). showed significant changes.
On day 21, the medicated group had a more uniform epithelial
3.5. Cytotoxicity evaluation surface thickness, a smoother surface layer, and a more consistent
connective tissue orientation compared with those of the control group.
The indirect cytotoxicity of various concentrations of medium ex- Fig. 8B shows the histopathological analysis of healing by using Mas-
tracts from nanofiber mats with different NPs is shown in Fig. 6. At son’s trichrome-stained samples. On day 7, the extent of collagen de-
1 mg/mL concentration, the cell viability of HaCaT decreased slightly position after wound induction significantly increased in all the wound-
possibly because of the excessive concentration of the released OH-30, inducing groups compared with that in the control group. The degree of
which exerted a certain degree of killing effect on cells. When the inflammatory response in the control group was significantly higher
concentration reached 0.5 mg/mL, the cell survival rate increased sig- than that in the dosing group possibly because of the im-
nificantly, and the cells proliferated. At 0.25 mg/mL, proliferation was munomodulatory effects of the previous OH-30. Epithelial regeneration
observed, but its degree was not obvious possibly because of the slightly is necessary to promote wound healing. At 24 h after the wound is in-
low OH-30 concentration. These data indicated that nanofiber mats flicted, basal cells isolated from the dermal layer of the wound interface
appeared to have a certain proliferative effect on cells and may be begin to divide and migrate to the defective area of the skin until the
developed as wound dressings. The findings of this study provided a thickness of the epidermis becomes normal (Jung et al., 2018). In this
basis for conducting subsequent animal wound experiments. The experiment, the epithelial regeneration ability of the medicated group
HaCaT cell morphology was studied by calcein-AM fluorescence was significantly better than that of the control group after 7 days.
staining (Fig. S2). The results indicated that the nanofibers might have Histopathological analysis confirmed that the acceleration of re-epi-
affected cell adhesion. This phenomenon might be advantageous for thelialization and collagen deposition was a wound healing effect in the
skin wound healing. Secondary damage to skin wounds was avoided dosing group, and these effects promoted wound healing.
during the change in dressing because the nanofibers acted as a drug
carrier for OH-30 rather than a tissue scaffold.
4. Conclusions

CMCS-OH-30 NPs were successfully encapsulated in nanofibers.


3.6. Evaluation of membrane performance during wound healing
Nanofibers loaded with different concentrations of NPs showed a strong
antibacterial activity. HE and Masson’s staining demonstrated that the
Wound healing was observed on days 0, 3, 6, 9, and 12 as shown in
wound healing ability of NP-loaded nanofibers was better than that of
Fig. 7A. The wound healing rate throughout the process is shown in
the blank control group. These results revealed that NP-loaded nano-
Fig. 7B. On the first 6 days after surgery, the wound healing rate of the
fibers had dual antibacterial and wound healing functions.
medicated group was clearer than that of the blank group, the control
group and the positive control group possibly of the continued slow
release of OH-30 (Table S2). After dosing was terminated on day 5, the Acknowledgments
wound healed slowly because the wound was not conducive to drug
penetration after scabbing. Six days after the operation, 0.27 % NP-30- This work was supported by the National Natural Science
NFs showed a faster wound closure rate of 83.74 ± 1.41 %, whereas Foundation of China (NSFC81673576 and 31600775), Shandong
the CMCS-OH30 NP group maintained a wound closure rate of Provincial Natural Science Foundation of China (ZR2016HM76),
69.33 ± 2.41 % (Sun et al., 2018). OH-30 contained in NF-30-NP Science and Technology Department of Yunnan Province (2019ZF003)

7
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

Fig. 7. Wound healing effects on mice models for different treated groups (A); Wound closure rate of all groups (B) (n = 3).

8
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

Fig. 8. Histopathology of the healing effect of samples on wound-induced skin damage with hematoxylin and eosin (HE) (A) stain ×20 (7, 21 days) or Masson’s
trichrome stain (B) ×20 (7, 21 days).

and Youth Innovative Team Development Plan of Universities in Gao, Z. H., Deng, C. J., Xie, Y. Y., Guo, X. L., Wang, Q. Q., Liu, L. Z., et al. (2018). Pore-
Shandong Province (2019KJM003). forming toxin-like protein complex expressed by frog promotes tissue repair. FASEB
Journal, 33(1), 782–795.
Garcia, M. C., Aldana, A. A., Tartara, L. I., Alovero, F., Strumia, M. C., Manzo, R. H., et al.
Appendix A. Supplementary data (2017). Bioadhesive and biocompatible films as wound dressing materials based on a
novel dendronized chitosan loaded with ciprofloxacin. Carbohydrate Polymers, 175,
75–86.
Supplementary material related to this article can be found, in the Gomes, A. P., Mano, J. F., Queiroz, J. A., & Gouveia, I. C. (2015). Incorporation of an-
online version, at doi:https://doi.org/10.1016/j.carbpol.2019.115786. timicrobial peptides on functionalized cotton gauzes for medical applications.
Carbohydrate Polymers, 127, 451–461.
Habiba, U., Afifi, A. M., Salleh, A., & Ang, B. C. (2017). Chitosan/(polyvinyl alcohol)/
References zeolite electrospun composite nanofibrous membrane for adsorption of Cr(6+), Fe
(3+) and Ni(2). Journal of Hazardous Materials, 322(Pt A), 182–194.
Abdelgawad, A. M., Hudson, S. M., & Rojas, O. J. (2014). Antimicrobial wound dressing Habiba, U., Siddique, T. A., Talebian, S., Lee, J. J. L., Salleh, A., Ang, B. C., et al. (2017).
nanofiber mats from multicomponent (chitosan/silver-NPs/polyvinyl alcohol) sys- Effect of deacetylation on property of electrospun chitosan/PVA nanofibrous mem-
tems. Carbohydrate Polymers, 100, 166–178. brane and removal of methyl orange, Fe(III) and Cr(VI) ions. Carbohydrate Polymers,
Ahmed, R., Tariq, M., Ali, I., Asghar, R., Noorunnisa Khanam, P., Augustine, R., et al. 177, 32–39.
(2018). Novel electrospun chitosan/polyvinyl alcohol/zinc oxide nanofibrous mats Haider, A., Haider, S., & Kang, I.-K. (2018). A comprehensive review summarizing the
with antibacterial and antioxidant properties for diabetic wound healing. effect of electrospinning parameters and potential applications of nanofibers in bio-
International Journal of Biological Macromolecules, 120(Pt A), 385–393. medical and biotechnology. Arabian Journal of Chemistry, 11(8), 1165–1188.
Akia, M., Rodriguez, C., Materon, L., Gilkerson, R., & Lozano, K. (2019). Antibacterial Huang, J., Deng, Y., Ren, J., Chen, G., Wang, G., Wang, F., et al. (2018). Novel in situ
activity of polymeric nanofiber membranes impregnated with Texas sour orange forming hydrogel based on xanthan and chitosan re-gelifying in liquids for local drug
juice. European Polymer Journal, 115, 1–5. delivery. Carbohydrate Polymers, 186, 54–63.
Arthanari, S., Mani, G., Jang, J. H., Choi, J. O., Cho, Y. H., Lee, J. H., et al. (2016). Jiang, Z., Han, B., Li, H., Yang, Y., & Liu, W. (2015). Carboxymethyl chitosan represses
Preparation and characterization of gatifloxacin-loaded alginate/poly (vinyl alcohol) tumor angiogenesis in vitro and in vivo. Carbohydrate Polymers, 129, 1–8.
electrospun nanofibers. Artificial Cells, Nanomedicine, and Biotechnology, 44(3), Jung, H. S., Kim, M. H., Shin, J. Y., Park, S. R., Jung, J. Y., & Park, W. H. (2018).
847–852. Electrospinning and wound healing activity of beta-chitin extracted from cuttlefish
Charernsriwilaiwat, N., Rojanarata, T., Ngawhirunpat, T., & Opanasopit, P. (2012). bone. Carbohydrate Polymers, 193, 205–211.
Electrospun chitosan/polyvinyl alcohol nanofibre mats for wound healing. Kohsari, I., Shariatinia, Z., & Pourmortazavi, S. M. (2016). Antibacterial electrospun
International Wound Journal, 11(2), 215–222. chitosan-polyethylene oxide nanocomposite mats containing bioactive silver nano-
Chen, W., Li, D., Ei-Shanshory, A., El-Newehy, M., Ei-Hamshary, H. A., Al-Deyab, S. S., particles. Carbohydrate Polymers, 140, 287–298.
et al. (2015). Dexamethasone loaded core-shell SF/PEO nanofibers via green elec- Lee, S. J., Heo, D. N., Moon, J. H., Ko, W. K., Lee, J. B., Bae, M. S., et al. (2014).
trospinning reduced endothelial cells inflammatory damage. Colloids and Surfaces B Electrospun chitosan nanofibers with controlled levels of silver nanoparticles.
Biointerfaces, 126, 561–568. Preparation, characterization and antibacterial activity. Carbohydrate Polymers, 111,
Das, A., Kumar, A., Patil, N. B., Viswanathan, C., & Ghosh, D. (2015). Preparation and 530–537.
characterization of silver nanoparticle loaded amorphous hydrogel of carbox- Li, S. A., Lee, W. H., & Zhang, Y. (2012). Efficacy of OH-CATH30 and its analogs against
ymethylcellulose for infected wounds. Carbohydrate Polymers, 130, 254–261. drug-resistant bacteria in vitro and in mouse models. Antimicrobial Agents and
El-Aassar, M. R., El Fawal, G. F., El-Deeb, N. M., Hassan, H. S., & Mo, X. (2016). Chemotherapy, 56(6), 3309–3317.
Electrospun polyvinyl alcohol/ pluronic F127 blended nanofibers containing tita- Li, S. A., Liu, J., Xiang, Y., Wang, Y. J., Lee, W. H., & Zhang, Y. (2014). Therapeutic
nium dioxide for antibacterial wound dressing. Applied Biochemistry and potential of the antimicrobial peptide OH-CATH30 for antibiotic-resistant
Biotechnology, 178(8), 1488–1502. Pseudomonas aeruginosa keratitis. Antimicrobial Agents and Chemotherapy, 58(6),
Eming, S. A., Krieg, T., & Davidson, J. M. (2007). Inflammation in wound repair: 3144–3150.
Molecular and cellular mechanisms. The Journal of Investigative Dermatology, 127(3), Li, S. A., Xiang, Y., Wang, Y. J., Liu, J., Lee, W. H., & Zhang, Y. (2013). Naturally oc-
514–525. curring antimicrobial peptide OH-CATH30 selectively regulates the innate immune
Fu, R., Li, C., Yu, C., Xie, H., Shi, S., Li, Z., et al. (2016). A novel electrospun membrane response to protect against sepsis. Journal of Medicinal Chemistry, 56(22), 9136–9145.
based on moxifloxacin hydrochloride/poly(vinyl alcohol)/sodium alginate for anti- Mi, Y., Su, R., Fan, D. D., Zhu, X. L., & Zhang, W. N. (2013). Preparation of N,O-car-
bacterial wound dressings in practical application. Drug Delivery, 23(3), 828–839. boxymethyl chitosan coated alginate microcapsules and their application to
Gao, Q., Huang, C., Sun, B., Aqeel, B. M., Wang, J., Chen, W., et al. (2016). Fabrication Bifidobacterium longum BIOMA 5920. Materials Science & Engineering C, Materials for
and characterization of metal stent coating with drug-loaded nanofiber film for Biological Applications, 33(5), 3047–3053.
gallstone dissolution. Journal of Biomaterials Applications, 31(5), 784–796. Miguel, S. P., Sequeira, R. S., Moreira, A. F., Cabral, C. S. D., Mendonca, A. G., Ferreira, P.,
et al. (2019). An overview of electrospun membranes loaded with bioactive

9
P. Zou, et al. Carbohydrate Polymers 232 (2020) 115786

molecules for improving the wound healing process. European Journal of treatment of cutaneous wounds. Science, 346(6212), 941–945.
Pharmaceutics and Biopharmaceutics, 139, 1–22. Sun, T., Zhan, B., Zhang, W., Qin, D., Xia, G., Zhang, H., et al. (2018). Carboxymethyl
Mishra, D., Bhunia, B., Banerjee, I., Datta, P., Dhara, S., & Maiti, T. K. (2011). chitosan nanoparticles loaded with bioactive peptide OH-CATH30 benefit nonscar
Enzymatically crosslinked carboxymethyl–chitosan/gelatin/nano-hydroxyapatite in- wound healing. International Journal of Nanomedicine, 13, 5771–5786.
jectable gels for in situ bone tissue engineering application. Materials Science and Wang, Z., Yan, F., Pei, H., Li, J., Cui, Z., & He, B. (2018). Antibacterial and en-
Engineering C, 31(7), 1295–1304. vironmentally friendly chitosan/polyvinyl alcohol blend membranes for air filtration.
Mokhena, T. C., & Luyt, A. S. (2017). Electrospun alginate nanofibres impregnated with Carbohydrate Polymers, 198, 241–248.
silver nanoparticles: Preparation, morphology and antibacterial properties. Zarandi, M. A., Zahedi, P., Rezaeian, I., Salehpour, A., Gholami, M., & Motealleh, B.
Carbohydrate Polymers, 165, 304–312. (2015). Drug release, cell adhesion and wound healing evaluations of electrospun
Morgado, P. I., Miguel, S. P., Correia, I. J., & Aguiar-Ricardo, A. (2017). Ibuprofen loaded carboxymethyl chitosan/polyethylene oxide nanofibres containing phenytoin sodium
PVA/chitosan membranes: A highly efficient strategy towards an improved skin and vitamin C. IET Nanobiotechnology, 9(4), 191–200.
wound healing. Carbohydrate Polymers, 159, 136–145. Zhang, Y., Zhao, H., Yu, G. Y., Liu, X. D., Shen, J. H., Lee, W. H., et al. (2010). Structure-
Moura, L. I., Dias, A. M., Leal, E. C., Carvalho, L., de Sousa, H. C., & Carvalho, E. (2014). function relationship of king cobra cathelicidin. Peptides, 31(8), 1488–1493.
Chitosan-based dressings loaded with neurotensin–an efficient strategy to improve Zhao, F., Lan, X. Q., Du, Y., Chen, P. Y., Zhao, J., Zhao, F., et al. (2018). King cobra
early diabetic wound healing. Acta Biomaterialia, 10(2), 843–857. peptide OH-CATH30 as a potential candidate drug through clinic drug-resistant
Pei, Z., Sun, Q., Sun, X., Wang, Y., & Zhao, P. (2015). Preparation and characterization of isolates. Zoological Research, 39(2), 87–96.
silver nanoparticles on silk fibroin/carboxymethylchitosan composite sponge as anti- Zhao, L., Chen, D., Yao, Q., & Li, M. (2017). Studies on the use of recombinant spider silk
bacterial wound dressing. Bio-medical Materials and Engineering, 26(Suppl 1), protein/polyvinyl alcohol electrospinning membrane as wound dressing. International
S111–118. Journal of Nanomedicine, 12, 8103–8114.
Rankin, L. C., & Artis, D. (2018). Beyond host defense: Emerging functions of the immune Zhou, T., Sui, B., Mo, X., & Sun, J. (2017). Multifunctional and biomimetic fish collagen/
system in regulating complex tissue physiology. Cell, 173(3), 554–567. bioactive glass nanofibers: Fabrication, antibacterial activity and inducing skin re-
Rasool, A., Ata, S., & Islam, A. (2019). Stimuli responsive biopolymer (chitosan) based generation in vitro and in vivo. International Journal of Nanomedicine, 12, 3495–3507.
blend hydrogels for wound healing application. Carbohydrate Polymers, 203, Zhou, T., Wang, N., Xue, Y., Ding, T., Liu, X., Mo, X., et al. (2016). Electrospun tilapia
423–429. collagen nanofibers accelerating wound healing via inducing keratinocytes pro-
Silva, J. P., Dhall, S., Garcia, M., Chan, A., Costa, C., Gama, M., et al. (2015). Improved liferation and differentiation. Colloids and Surfaces B Biointerfaces, 143, 415–422.
burn wound healing by the antimicrobial peptide LLKKK18 released from conjugates Zhou, Y., Yang, H., Liu, X., Mao, J., Gu, S., & Xu, W. (2013). Electrospinning of carbox-
with dextrin embedded in a carbopol gel. Acta Biomaterialia, 26, 249–262. yethyl chitosan/poly(vinyl alcohol)/silk fibroin nanoparticles for wound dressings.
Sun, B. K., Siprashvili, Z., & Khavari, P. A. (2014). Advances in skin grafting and International Journal of Biological Macromolecules, 53, 88–92.

10

You might also like