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

Carbohydrate Polymers 238 (2020) 116175

Contents lists available at ScienceDirect

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

Wound healing of nanofiber comprising Polygalacturonic/Hyaluronic acid T


embedded silver nanoparticles: In-vitro and in-vivo studies
M.R. El-Aassara,b,*,1, Omar M. Ibrahimb,c,1, Moustafa M.G. Foudad,**, Nagham G. El-Beherib,
Mona M. Agwae
a
Department of Chemistry, College of Science, Jouf University, Sakaka 2014, Saudi Arabia
b
Polymer Materials Research Department, Advanced Technology and New Material Institute, City of Scientific Research and Technological Applications (SRTA-City), New
Borg El-Arab City, Universities and Research Institutes District, Alexandria 21934, Egypt
c
Department of Medicine and Translational Research, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
d
Pre-Treatment and Finishing of Cellulosic-based Fibers Department, Textile Industries Research Division, National Research Center, 33 El- Behooth St, Dokki, Giza,
12311, Egypt
e
Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Division, National Research Centre, 33 El- Behooth St, Dokki,
Giza, 12311, Egypt

ARTICLE INFO ABSTRACT

Keywords: The current study is pertaining to develop a novel wound dressing, comprising natural biologically absorbable
Polygalacturonic acid materials for wound healing In-vivo. Wound dressing is composed of Polygalacturonic acid, Hyaluronic acid
Silver nanoparticles embedded silver nanoparticles, which is further fabricated to form nanofibrous mat, using electrospinning. Silver
Nanofiber nanoparticles was prepared using PGA. AgNPs in this formula will serve as an antioxidant and anti-inflammatory
Hyaluronic acid
that protect cells from destructive effect of elevated ROS and accelerate wound healing. The physical perfor-
Antibacterial
Wound dressing
mance and water contact angle for nanofiber was evaluated. The produced nanofiber was characterized by
Fourier-transform infrared (FTIR), scanning electron microscopy and thermal analysis. Also, the embedded
AgNPs was also characterized by UV–vis spectroscopy and TEM. The nanofiber mates embedded AgNPs was
applied to the wounded site of albino rats in-vivo. Histopathological assessment for the wound was fully per-
formed. Also, the antimicrobial activity for the fabricated wound dressing was evaluated against gram+ve and
gram −ve bacteria.

1. Introduction administration that lead to sever side effect due to higher serum level
(Musters, Burger, Buskens, Bemelman, & Tanis, 2015). So, a better
The skin of the human has a prominent protective role due to protection against microbial infection by fabrication of suitable anti-
stratum corneum layer that make skin acts like a permeation barrier microbial wound dressing is necessary for faster wound tissue re-
(Boer, Duchnik, Maleszka, & Marchlewicz, 2016). Cutaneous injury is generation (Li et al., 2016).
major problems for health care system all over the world as the skin The process of wound healing proceeds through a cascade of events
loses its protective function if any damage of this layer occurs, either by which include four different major phases; coagulation, inflammation,
wounds or burns (Ho, Walsh, Yue, Dardik, & Cheema, 2017). The proliferation and tissue remodeling (Shalaby et al., 2020). The in-
wound generally refers to a skin damage caused by surgery or trauma. flammatory response is a substantial phase of wound healing and it
Wound infection is one of the fundamental causes that hampered and begins within minutes after the injury and characterized by the man-
delay the healing process (Sarhan, Azzazy, & El-Sherbiny, 2016). For ufacturing of cytokines (pro-inflammatory) and stimulation of cellular
the antibiotics to be effective against and or prevent wound infection, a immunity (Wilkinson, White, & Chipman, 2011). Not only cytokines are
high concentration at the specified wound area is guaranteed that leads vital in initiating, sustaining and regulating the cellular response after
to higher local concentration and lower serum level instead of systemic injury but also, they have been involved in wound healing impairment.

Corresponding author at: Department of Chemistry, College of Science, Jouf University, Sakaka 2014, Saudi Arabia.

Corresponding author at: Pre-treatment and Finishing of Cellulosic-based Fibers Department, Textile Industries Research Division, National Research Centre,
⁎⁎

Dokki, Giza, 12622, Egypt.


E-mail addresses: mrelaassar@ju.edu.sa (M.R. El-Aassar), drmmfouda@gmail.com (M.M.G. Fouda).
1
M. R. El Aassar and Omar M. Ibrahim contributed equally to this work.

https://doi.org/10.1016/j.carbpol.2020.116175
Received 31 December 2019; Received in revised form 11 March 2020; Accepted 13 March 2020
Available online 09 April 2020
0144-8617/ © 2020 Elsevier Ltd. All rights reserved.
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Through the inflammatory stage of the wound healing macrophages conditions provided by silver nanoparticles that lead to reduced re-
and neutrophils invade the wound and produce great quantities of su- active cell infiltration, which further supported the fibrous connective
peroxide radical anions. This phenomenon is characterized as the “re- tissue proliferation and sequential keratin layer regeneration (Rath,
spiratory burst’. Moreover, a proinflammatory cytokines can stimulate Hussain, Chauhan, Garg, & Goyal, 2016).
other cells like fibroblast and chondrocytes to produce reactive oxygen Several methods in the literature, for the preparation of silver na-
species (ROS) (Meier et al., 1989). noparticles have been disclosed extensively. One of the common and
Reactive oxygen species (ROS) are considered chemically reactive cost-effective ways is the chemical reduction method (Abdel-Mohsen
species comprising oxygen. Examples include peroxides, superoxide, et al., 2014; Abdelsalam et al., 2019; Abu-Saied, Abdel-Halim, Fouda, &
hydroxyl radical, and singlet (Hayyan, Hashim, & AlNashef, 2016). It Al-Deyab, 2013; Dahlous, Abd-Elkader, Fouda, Al Othman, & El-Faham,
plays a fundamental role in the modulation of cell survival, cell death, 2019; El-Aassar, El Fawal, Kamoun, & Fouda, 2015; El-Aassar, Fouda, &
differentiation, cell signaling, and inflammation-related factor produc- Kenawy, 2013; El-Aassar, Hafez, El-Deeb, & Fouda, 2014; El-Hamshary
tion (Abdal Dayem et al., 2017). The production of these ROS in the et al., 2015; El-Naggar, Shaheen, Fouda, & Hebeish, 2016; El-Rafie
wound is needed for the defense against invading pathogenic bacteria et al., 2011; Fouda et al., 2016; Fouda, El-Aassar, & Al-Deyab, 2013;
as its part of the innate immune system (auf dem Keller, Kumin, Braun, Fouda et al., 2015; Fouda, EL Naggar, Shaheen, & Al Deyab, 2014;
& Werner, 2006). The substantial goal of the generation of higher ROS Hebeish, Abdel-Mohdy et al., 2011; Hebeish, El-Naggar et al., 2011; El-
levels during inflammatory stage of the wound healing is the destruc- Naggar et al., 2018; Přichystalová et al., 2014; Shaheen et al., 2016;
tion and elimination of the invading pathogenic microorganisms and/or Textor, Fouda, & Mahltig, 2010). Silver nitrate is frequently being used
degradation of the damaged tissue structures (Bonferoni et al., 2018). In as a precursor for silver ions., in addition, natural and/or synthetic
addition to the beneficial role of ROS in killing pathogenic bacteria, it polymer is being utilized this way in the reduction of silver ions, and in
also has a series of negative effects. At low concentration ROS are the the meantime, stabilize the formed silver nanoparticles as well.
critical mediators of various intracellular signaling (D’Autreaux & Polygalacturonic acid (PGA) in this current study is being used as
Toledano, 2007). It was reported that low ROS level that secreted in a natural candidate for reducing the silver ions into silver nanoparticles;
sustained manner at wound site participate in a variety of biological in the meantime, PGA is used as good stabilizer for the formed AgNPs
processes, comprising angiogenesis, epithelialization and granuloma- too. Polygalacturonic acid (PGA), which is namely sometimes, as Pectin
tous tissue formation (Nocchi, Björklund, Svensson, Engblom, & acid, is considered as natural occurring polymer, and it looks as
Ruzgas, 2017). On the other hand, higher ROS level can lead to sever transparent, gelatinous acid present in mature fruits and some other
cellular damage (Forman, Ursini, & Maiorino, 2014). So, the regulation vegetables (Peng et al., 2011). It is formed by the degradation of pectin,
of ROS levels in wound site is essential and required for efficient repair. and is produced through the interaction between pectinase and pectin.
Over production of ROS for an extended period is a substantial Hyaluronic acid (HA) is being used together with PGA in this current
cause for hindering wound healing process resulting in epithelial cell work also to improve the healing process (Fouda et al., 2016). As
deterioration, oxidative stress, and cytotoxicity that supports the ad- known, HA considered one of the best biomaterials based Poly-
vantage of the antioxidant agent for these symptoms (Bonferoni et al., saccharide utilized for wound care management particularly, due to its
2018; Pothireddy et al., 2016). biocompatibility, biodegradability, and atypical physicochemical fea-
Silver nanocarriers induce its antioxidant activity through reduc- tures. HA is naturally occurring, nonimmunogenic, anionic, non-sul-
tion, electron donating and radical scavenging abilities. So, silver na- fated Glycosaminogly can distributed broadly thru connective tissues,
noparticles have been recognized as scavenger of ROS and can decrease epithelia, neural tissues, and synovial fluids. HA is highly soluble water
the amount of free radicals emerging through inflammatory stage of and more hydrophilic than any other natural occurring polymers,
wound healing (Gopukumar, Sana-Fathima, Alexander, Alex, & therefore, it has a strong role in getting rid of lots of wound exudates,
Praseetha, 2016). They have the ability to cross the biological com- hence, enhance the chance for wound healing as well (El-Aassar et al.,
partment with a greater rate than bulk silver ions since that it gained a 2015).
great interest in the reduction of wound bioburden (Wilkinson et al., So, the main objective in this current study is to develop a wound
2011). The anti-inflammatory activity of silver nanoparticles has been dressing nanofiber mat, comprising natural biologically absorbable
reported by many researchers which helps in accelerating and adjust- materials; Hyaluronic, Polygalacturonic acid embedded silver nano-
ment of wound healing process (Tian et al., 2007; Wong et al., 2009). particles for wound healing application; in-vivo. Silver nanoparticles in
For the modulation of the healing process, diverse inflammatory med- this formula, will serve as an antioxidant and anti-inflammatory that
iators are produced within the wound area so that, the clarification of protect the cells from the destructive effect of elevated ROS and in the
both the pro- and anti-inflammatory cytokines is essential to protect the meantime, accelerate wound healing process (Scheme 1).
newly regenerative tissue from damage caused by cytokines imbalance,
oxidants and antioxidants around the wound area (Eming, Krieg, & 2. Materials and methods
Davidson, 2007). The successful tissue regeneration and wound repair
related with in vivo regulation of the inflammatory response as pro- 2.1. Materials
longed inflammatory response is among the causes of delaying wound
healing (Hafez, El-Aassar, Khalil, Al-Deyab, & Taha, 2011). Tian et al. Polygalacturonic acid (PGA) was purchased from TCI Chemicals
showed that the anti-inflammatory activity of silver nanoparticles is (Tokyo, Japan). Silver nitrate (AgNO3) (99.9%), Hyaluronic acid (HA)
mediated through cytokines modulation (Tian et al., 2007). and Poly (Vinyl) Alcohol (PVA) were obtained from Sigma-Aldrich
As previously mentioned in the above section that cytokines can (Schnelldorf, Germany). Microorganisms were collected from Protein
stimulate fibroblast and chondrocytes to produce ROS (Meier et al., Department, Genetic Engineering and Technological Application
1989). So, modulation of cytokines production by silver nanoparticles Institute (City of Scientific Research and Technological Application).
can participate in lowering ROS levels to avoid sever cellular damage They were completely purified on LB media (Luria Bertani medium (LB)
and delay wound healing (Yu et al., 2016). In previous study the rate of and incubated at 37 °C for 24 h.
wound healing of rat group treated with collagen nanofiber im-
pregnated silver nanoparticles were faster than plain collagen nanofiber 2.2. Preparation of silver nanoparticles-Polygalacturonic acid/Hyaluronic
with no silver nanoparticle. Histological examination showed an ac- acid/ poly (vinyl) alcohol (Ag-PGA/HA)-PVA antibacterial nanofiber
celerated collagen production, re-epithelization, and improved wound
contraction with silver nanoparticles composite collagen nanofibers. 2.2.1. Synthesis of silver nanoparticles (AgNPs)
These results were attributed to the anti-inflammatory and aseptic Polygalacturonic acid was dissolved in bi-distilled water to produce

2
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Scheme 1. Diagram illustrating the formation of (Ag-PGA/HA)-PVA.

1% (w/v) solutions. Complete dissolution was obtained by addition of concentration (range: 0.12–1% w/v).
1 M NaOH (pH = 11.7) and magnetic stirring at room temperature for
30 min. Then 400 μl of 0.45 mM AgNO3 was added under magnetic 2.3.2. Scanning (SEM) and transmission (TEM) electron microscopy
stirring for 1 h until yellow solution was obtained. Nanofibers and Silver nanoparticles morphology were captured by
scanning electron microscope “(JEOL-JSM-6360LA-Japan)”, while
2.2.2. Preparation of Hyaluronic- Polygalacturonic acid/silver powder by transmission electron microscope by a “JEM-1010 unit
nanoparticles (Ag-PGA/HA) blend solution (JEOL Ltd., Tokyo, Japan)”, provided with an AMT XR-80 digital
Blend solution was obtained by adding 0.25% (w/v) of HA to Ag- camera (Advanced Microscopy Techniques Corp., Woburn, MA, USA).
PGA solution under continuous magnetic stirring and raising the tem- Furthermore, Image J software was used to measure AgNPs mean
perature till 50 °C for 1 h, until homogenous solution was produced. particle size, and particle distribution was indicated by polydispersity
This was followed by 24 h freeze drying. index (PDI) according to the following formula (Clayton, Salameh,
Wereley, & Kinzer-Ursem, 2016):
2.2.3. Preparation of (Ag-PGA/HA)-PVA spinnable solution 2
Standard Deviation
The spinnable solution was prepared by dissolving PVA 8% (w/v) at Poly Dispersity Index (PDI) =
Mean Diameter (1)
30 °C, and then 1% of Ag-PGA/HA dry powder was added. This solution
was left under magnetic stirring overnight till the solution became
homogeneous. 2.3.3. Fourier transform infrared (FTIR) and Raman spectroscopy
The main changes in chemical composition of polymer blend were
2.2.4. Electrospinning process verified by FTIR (Shimadzu FT-IR8400 S, Japan), by measuring spectral
The prepared spinnable solution was inserted into syringe made of range of 4000–400 cm−1 of the fine powder. Raman spectra of the
glass having metal capillary needle (with inner diameter = 0.5 mm). copolymer fine powder were obtained by Bruker FT-Raman
“The electrospinning setup (nanoNC needle laboratory machine, Model: Spectrometer Multi RAM (Bruker Optics Inc. Billerica, MA USA) with
ESR100D, Seoul, Korea)” contained syringe pump and collector of FT-Raman microscope – Raman scope III was used.
grounded plate coated with aluminum foil, as well as high voltage
power source was applied to the polymer formulation; (Ag-PGA/HA)- 2.3.4. Thermogravimetric analysis (TGA)
PVA blend solution in a syringe thru an alligator clip adhered to the The thermal degeneration attribute of nanofibers was verified by
syringe needle. The electrospinning was accomplished at room tem- thermogravimetric analyzer “(Shimadzu Thermal Gravimetric Analysis
perature, and the working solutions were inserted from the syringe (TGA)—50, Japan)”, that was achieved under nitrogen at a heating rate
pump with a feed rate of 1.3 ml/h. The distance between the needle tip of 10 °C/min until 600 °C, with starting weight of6 mg sample.
and the grounded collector plate was 17 cm, and the high voltage was
applied at 11 kV. Dry nanofibers were gathered directly from the alu- 2.3.5. Tensile strength
minum foil and kept at room temperature. Nanofibers mechanical properties were assessed by tensile tester
(model AG-I, Shidmadzu, Japan). Nanofiber with 6 mm × 1 mm di-
2.3. Characterization mensions were analyzed at a crosshead speed of 30 mm/min. Digital
micrometer (Mituto, Tokyo, Japan), with a precision of 0.001 mm was
2.3.1. UV–vis spectroscopy used to measure nanofiber thickness. The average of five times was
Silver nanoparticles optical properties and synthesis was verified by obtained.
using UV- (LAMBDA 650) range 300–600 nm. 1 ml of pure PGA was
used as blank before sample measurement. Synthesis was optimized by 2.3.6. Water contact angle
observing AgNPs formation under several conditions; AgNO3 con- Water contact angle of each nanofiber was assessed by measuring
centration (range: 0.12-0.45 mM) and Polygalacturonic Acid their static contact angle (θ) (Ramé-Hart Instrument Goniometer, model

3
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Fig. 1. UV–vis absorption spectra of A. PGA concentration B. AgNO3 concentration effect on the preparation of Ag-PGA.

500-F1, France) by assigning a drop of ultra-pure water on it at three 2.5.2. Photo documentation and histological analysis
different locations. Starting immediately after surgery, images of the skin wounds at 3,
5, 8,10 and 14 days after surgery were captured with an optical digital
camera at a fixed focal distance (Abercrombie, Flint, & James, 1956).
2.4. In-vitro antibacterial study
Dressings were soaked with sterile saline until they could be removed
with minimal disturbance of the surface of the wound. From the healed
Nanofibers were cut into equal-sized pieces and sterilized by UV.
wound, a specimen sample of tissue is isolated from each group of rats
(Ag-PGA/HA)-PVA and (PGA/HA)-PVA were incubated with 100 μl of
for histopathological examination at day 10 and day 14 (Biswas, Maity,
Bacillus Subtilis, Staphylococcus Aureus and Escherichia Coli on agar
& Mukherjee, 2004). The animals were anesthetized using the anes-
plates at 37 °C for 24 h (Fouda, Knittel, Hipler, Elsner, & Schollmeyer,
thetic procedure described above, followed by intracardiac adminis-
2006; Selvaraj, Thangam, & Fathima, 2018).
tration of 0.5 ml of potassium chloride. Specimens of the wound area
were removed, fixed in 10% formalin, and sent for histopathological
2.5. In-vivo study examination and slide preparation according to routine procedures.
Slices of 5 μm from the skin pass through the center of the wound were
All carried experiments on rodents were following guidelines and stained with hematoxylin-eosin (H&E) and picrosirius, and analyzed
protocols, and approved by the Research Ethical Committee of under a polarized light microscope using the ACT-@U program for
Institutional Animal Care and Use Committee at Alexandria University histological examination and Masson’s trichrome staining (MTS) for
(ALEXU-IACUC) with approval number; AU14-190806-3-1. estimation of collagen deposition (Sarhan et al., 2016). The reduction
percentage in wound area was calculated from the following Eq. (2):
2.5.1. Preparation of full thickness wound model Healed Area
Wound Contraction %=
Twenty male rats 60 days old, with a mean weight of approximately Total Area (2)
180 g were used. The animals were accommodated in a separate
birdcage at an orderly temperature (25 ± 3 °C; 35–60% humidity)
below a 12 h light/dark cycle and received a standard diet including 3. Results and discussion
pelleted food “(Labina; Purina Nutrients Ltda, Sao Paulo-SP, Brazil)”
and mineral water ad libitum. This study was accepted by Local Ethical 3.1. UV–vis spectroscopy and TEM analysis of produced silver nanoparticles
Committee of Animal Research. The animals were counted, weighed
and then divided into four groups with five animals in each as follows; PGA was used as a green synthesis platform of AgNPs. Because it is
group I: applied PGA/HA/PVA nanofiber, group II: Ag-PGA/HA/PVA very rich in carboxylic and hydroxylic groups that transfer electrons to
nanofiber, group III: vehicle control and applied commercial ointment Ag+, resulting in Ag° nanoparticles. Therefore, PGA is acting as a re-
(Garamycin®) and group IV: vehicle control and applied saline. ducing agent for AgNPs synthesis. UV–vis spectroscopy was used to
As for surgical proceedings, the animals were subjecting to weight confirm synthesis and optical response of colloidal AgNPs. PGA con-
calculation, followed by anesthetized using intramuscular administra- centration is a limiting factor in the synthesis process of AgNPs. In
tion of 10% Ketamine Hydrochloride “(Dopalen®, 0.1 ml/100 g body Fig. 1A, maximum UV–vis spectra were observed upon using 0.5–1%
weight)” and 2%xylazine hydrochloride “(Calmium®, 0.1 ml/100 g PGA concentrations at wavelength of 415 nm. While lower spectra were
body weight)”. After shaving, Antisepsis of the area was achieved with observed at 0.12-0.25% PGA concentrations at wavelength of 418 nm.
4% alcohol-based iodine. In the center of the shaved area and with a In Fig. 1B, 0.45 mM of AgNO3showed the highest spectra at wavelength
fine scissor, a surgical skin lesion (Ø = 2.5 cm), 1 cm below the bone of 415 nm. While 0.25 mM and 0.12 mM AgNO3 concentrations showed
prominence on the back of the animals, was formed using a metallic intermediate and lowest spectra at wavelengths of 412 nm and 427 nm,
circular marker to delimit the area, and with the aid of a surgical respectively. The most predominant absorption band was 410–415 nm,
scalpel, the dorsal muscle fascia was exposed. The wound was cleaned which confirmed successful production of AgNPs and suggested that the
daily with alcohol, followed by topical application of treatments. produced particle size is ranging between 5–10 nm (Zahran, Ahmed, &
Finally, wounds were covered with sterile gauze and fixed with circular El-Rafie, 2014).
adhesive bands and the wound areas were calculated (Kodati, Burra, & TEM was used for further characterization of synthesized Ag-PGA
Kumar, 2011; Rath, Johal, & Goyal, 2011). powder. It displayed spherical silver particulates which were

4
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Fig. 2. (1) TEM image of A. Ag-PGA powder B. (Ag-PGA/HA)-PVA nanofiber, (2) SEM images of a. (Ag-PGA)-PVA b. (PGA/HA)-PVA c. (Ag-PGA/HA)-PVA na-
nofibers.

encapsulated within PGA with an average particle size of 8.6 nm con- electrospinning process of (PGA/HA)-PVA loaded with AgNPs.
firming what has been shown by UV–vis (Fig. 2.1.A). Polydispersity
index was used to estimate stability and distribution of produced
3.3. FTIR and Raman powder analysis
AgNPs. Synthesized AgNPs had PDI = 0.18 which indicated successful
encapsulation and homogenous distribution of produced nanoparticles.
FTIR and Raman spectra were used to identify characteristic peaks
Therefore, UV–vis and TEM confirmed that PGA is an efficient reducing
and major changes occurred within polymers blend and AgNPs loading.
and stabilizing agent for the synthesis of AgNPs.
In Fig. 3A, infrared absorption spectra of PGA/HA were compared
without and with AgNPs by observing shifting in O-H and NeH
3.2. Morphological analysis of nanofibers structure stretching peaks from 3446 cm−1 to 3566 cm-1, C–H stretching from
2935 cm-1 to 2941 cm-1, CeOeC from 1620 cm-1 to 1604 cm-1 and
Surface morphology and size of (Ag-PGA)-PVA, (PGA/HA)-PVA and CeOH bending from 1417 cm−1 to 1425 cm-1. Both PGA and HA have a
(Ag-PGA/HA)-PVA nanofibers were observed by using SEM. Fig. 2.2 is similar chemical structure and infrared spectra. However, PGA/HA and
showing that (Ag-PGA)-PVA nanofibers have an approximate diameter Ag-PGA/HA were showing a more prominent transmittance % ranging
of 244 nm, while (PGA/HA)-PVA and (Ag-PGA/HA)-PVA nanofibers between 3136 cm-1 and 3639 cm-1 due to NeH stretching of HA com-
diameters are 320 nm and 326 nm, respectively. Upon comparing ponent. The Raman spectra of Ag-PGA, PGA/HA and Ag-PGA/HA
average diameter of (Ag-PGA)-PVA nanofibers with (PGA/HA)-PVA and powders were also observed. In Fig. 3B, Ag-PGA and PGA/HA exhibited
(Ag-PGA/HA)-PVA nanofibers, 30% increase in diameter was observed. characteristic peaks at 1452 cm-1 and 1119 cm-1due to the CH2 group of
This increase could be due to the addition hyaluronan content in PGA PGA, and glycosidic bonds CeOeC present between the repeating units
solution, which led to the increased electrical charges on the surface of from HA, respectively. The intensity ratio of the peaks at 1458 cm-1
liquid droplet. No prominent changes were observed in nanofibers varied with the mixing combination of PGA, HA and AgNPs. The Ag-
morphology. While TEM was used to observe AgNPs component in- PGA/HA exhibited vibrational bands at 1458 cm-1 and 2912 cm-1which
corporated within (Ag-PGA/HA)-PVA nanofibers. Fig. 2.1.B is showing can be assigned to the NH2 group and CH group, respectively. There-
that AgNPs are homogenously distributed within (Ag-PGA/HA)-PVA fore, FTIR and Raman spectra confirmed PGA and HA successful
nanofiber. Therefore, SEM and TEM confirmed successful blending and AgNPs presence.

5
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Fig. 3. A. FTIR spectra B. Raman spectra of PGA/HA, Ag-PGA and Ag-PGA/HA powder.

3.4. Wettability, thermal and mechanical analysis of nanofibers with the fact that biopolymers with a higher content of amine-func-
tionalized hyaluronic acid induces higher values of contact angles were
TGA was used to characterize thermal stability property of nanofi- observed.
bers (Fig. 4A). AgNPs component did not alter thermal weight loss
pattern of (PGA/HA)-PVA. Both (PGA/HA)-PVA and (Ag-PGA/HA)- 3.5. Antibacterial effect
PVA nanofibers underwent through three major steps of weight loss.
They underwent 16% weight loss at 25–215 °C stage, 31% loss at Fig. 5 is showing the antibacterial effect of (PGA/HA)-PVA and (Ag-
216–310 °C stage and 14% final loss at 311–600 °C stage. On the other PGA/HA)-PVA nanofibers. It was obvious that (PGA/HA)-PVA nano-
hand, (Ag-PGA)-PVA underwent 6% weight loss at 25–215 °C 1st stage, fiber did not exhibit any zone inhibition/antibacterial effect. While
10% loss at 216–310 °C 2nd stage and 59% loss at 311–600 °C 3rd stage. upon adding AgNPs, (Ag-PGA/HA)-PVA nanofibers became very ef-
These variations in weight loss could be accounted to rapid rupture of fective against gram-positive bacterial strains; Bacillus Subtilis and Sta-
HA glycosidic bond content in both (PGA/HA)-PVA and (Ag-PGA/HA)- phylococcus Aureus, as well as gram-negative bacterial strain; Escherichia
PVA compared to (Ag-PGA)-PVA nanofiber. Coli. The antibacterial effect of AgNPs might be due to disruption of
Fig. 4B illustrates the mechanical properties of the (Ag-PGA)-PVA, bacterial cell membrane proteins and interaction with DNA.
(PGA/HA)-PVA, and (Ag-PGA/HA)-PVA nanofibers. The tensile stress
of the (Ag-PGA)-PVA nanofibers (∼4.1 MPa) was almost two time 3.6. Wound healing effect and histopathological assessment
stronger than the (PGA/HA)-PVA (∼ 1.9 MPa), and (Ag-PGA/HA)-PVA
(∼1.6 MPa). However, the tensile strength of the (Ag-PGA/HA)-PVA Fig. 6 displays the macroscopic appearance of the wounds during
nanofibers (∼30%) was increased to 22.5, and 10% for (PGA/HA)-PVA the time course of the experiment. No sign of abscess formation or
and (PGA/HA)-PVA, respectively. The (PGA/HA)-PVA nanofibers hypertrophic scars both in the early phase; 5th day or in the final phase;
showed a significant improvement in stress-strain curve, indicating that 14th day, respectively, were observed. The digital images of the wounds
their ability to resist network deformation was decreased with AgNPs allowed the estimation of the wound area progression in the groups
content increase. This improvement in stress-strain value of the nano- during the time course of the experiment.
fiber due to the HA structural that acts as additional crosslinking effect The trial shows that the contraction force is exerted by many factors
from the physical interaction between hyaluronan chains, PGA, and within the wounded area. The scab is responsible for a rapid and early
PVA chains. contraction in the initial stages which is probably due to simple dehy-
The wettability of produced nanofibers was observed by measuring dration. After five days there was a further abrupt onset of contraction,
contact angle (Fig. 4C). (Ag-PGA)-PVA, (PGA/HA)-PVA and (Ag-PGA predominant in the prepared (Ag-PGA/HA)-PVA nanofiber and blank
HA)-PVA nanofibers were exhibiting water contact angle of 67.9°, 59.2° (PGA/HA)-PVA nanofiber over others. This can be related to the de-
and 64.2°, respectively. (Ag-PGA HA)-PVA nanofibers are showing the creased capillary permeability and dehydration of the wound bed. With
lowest contact angle and best hydrophilicity. This may be associated the prepared nanofiber the contraction proceeds faster until epithelial

Fig. 4. A. TGA results of the weight loss B. tensile strength and C. Wettability test by using a contact angle tester of (Ag-PGA)-PVA, (PGA/HA)-PVA, and (Ag-PGA/
HA)-PVA nanofibers.

6
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Fig. 5. Antibacterial effect of (PGA/HA)-PVA and (Ag-PGA/HA)-PVA nanofibers on Bacillus Subtilis, Staphylococcus Aureus and Escherichia Coli.

closure is nearly complete. The wound contraction is depicted in Fig. 6, healing activity as compared to other groups that treated with Gar-
and is showing that the wound contracting ability of (Ag-PGA/HA)-PVA amycin® cream and control group (applied saline) in excision wound
nanofiber and blank (PGA/HA)-PVA nanofiber treated groups showed model (Fig. 6).
significant wound healing from day 8 onwards and the epithelization The inflammation is the self defense mechanism of the body to re-
period was on day 14. Whereas in the Garamycin® cream and control move critical stimuli, including irritants, pathogens or damaged cells
group (applied saline) it took more than 14 days (Fig. 6). during the healing process. Many cells are involved directly or in-
The results of excision wound model and % of wound contraction directly through production of inflammatory cytokine in the pathology
are illustrated in Table 1. Both the (Ag-PGA/HA)-PVA nanofiber and of chronic inflammation including macrophages, eosinophils and neu-
(PGA/HA)-PVA nanofiber treated groups exhibited significant wound trophils (Khansari, Shakiba, & Mahmoudi, 2009). The histopathological

Fig. 6. Macroscopic examination of the wounds treated with (PGA/HA)-PVA nanofiber (Group I), (Ag-PGA/HA)-PVA nanofibers (Group II), Garamycin® cream;
positive control (Group III) and saline; negative control (Group IV) for 14 days.

7
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Table 1
Evaluation of the wound area cm2 and wound contraction % of different groups over a period of 14 days.
Observation Days Wound Area (cm2) (Mean ± SEM) and Percentage of Wound Contraction

Group I Group II Group III Group IV

Area (cm2) % contraction Area (cm2) % contraction Area (cm2) % contraction Area (cm2) % contraction

0 5 ± 0.46 Nil 4.8 ± 0.5 Nil 5.15 ± 0.4 Nil 4.8 ± 0.3 Nil
3 2.9 ± 0.8 41.85 1.6 ± 0.2 66.95 3.40 ± 0.59 33.99 3.8 ± 0.3 19.47
5 2.2 ± 0.67 55.85 0.91 ± 0.16 81.14 2.35 ± 0.45 54.33 3.1 ± 0.2 34.94
8 1.26 ± 0.52 75.08 0.36 ± 0.10 92.49 1.89 ± 0.31 63.27 2.3 ± 0.25 51.55
10 0.59 ± 0.27 88.31 0.18 ± 0.07 96.23 0.84 ± 0.32 83.50 1.54 ± 0.17 68.08
14 0.13 ± 0.062 97.42 0.02 ± 0.01 99.51 0.38 ± 92.43 0.79 ± 0.15 83.65

Fig. 7. A. Histological analysis of treated wounds on day 10 and 14 using H and E stain. Epi: Epidermis; K: keratin layer; BV: blood vessels; HF: hair follicles; SG:
sebaceous glands; U: ulcer; M: muscle B. Histopathological evaluation of treated wounds using Masson’s trichome Stain (MTS) on day 10 and 14 (original magni-
fication = 100).

8
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Fig. 8. Histological analysis of normal skin using hematoxylin-eosin (H&E) staining (A) and Masson’s trichome (MTS) Stain (B).

examination to the injured skin area showed in Fig. 7 was used to il- inflammatory cells/neutrophils was high in control when compared
lustrate the possible mechanism of wound contraction in Fig. 6 com- with treated group as inflammatory cells were reduced due to the
pared to normal skin described in Fig. 8. strong antimicrobial activity of silver nanoparticles in the open wound
Fig. 7 A is showing all images of the histopathological examination site.
using H&E staining of the wounded skin sections from different groups For estimation and analysis of the intensity of the deposition of
at day 10 and 14 respectively compared to the normal skin section collagen fibers Masson’s trichrome (MTS) stain was used as collagen
Fig. 8A. The normal skin section showed the complete structural ele- synthesis is an essential step to ensure complete wound healing
ments of the healthy skin including intact epidermis (external epithe- (Fig. 7B). Collagen was observed as blue violet color. The stain was
lium formed of 2–3 cell layers), dermis (layer of connective tissue), successfully applied to estimate the collagen deposition progress during
sebaceous glands and hair follicles as illustrated in Fig. 8A. the formation of granulation tissue and matrix remolding. The intensity
These groups showed a varying response to the treatment at which of the blue stain equivalent to the relative amount of total collagen fiber
the best wound healing ability and complete epithelization was re- deposited. The normal skin showed uniform, mature, compact and
corded for group II; (Ag-PGA/HA)-PVA nanofiber followed by group I; normal collagen deposition as illustrated in Fig. 8B. Histopathological
applied blank (PGA/HA)-PVA nanofiber. The wound area showed no evaluation of the skin sections using Masson’s trichome stained tissues
ulcer with almost normal skin that filled with dense connective tissues of wound treated groups are illustrated in Fig. 7B. On both 10th and 14th
and was surrounded by a new dermal layer on both day 10 and 14 day, the skin group treated with saline (group IV) showed very much
(Fig. 7A). Well-formed epidermis (external epithelium formed of 2–3 less collagen deposition compared to other treated groups and normal
cell layers) and dermis (layer of connective tissue) were seen in the skin. The skin section from group treated with Garamycin® cream
tissues. The skin sections also showed the most basic structures of the (group III) showed a very less collagen deposition on day 10 whereas on
rat's skin like hair follicle (HF), sebaceous gland (SG) and blood vessels day 14 the deposited collagen was moderate in the granulation tissue.
(BV). Previous researches demonstrated the fundamental roles played Skin tissue section treated with blank (PGA/HA)-PVA nanofiber (group
by the angiogenesis in the regeneration of both epidermis (EPI) and I) showed a moderate collagen deposition in the granulation tissue on
dermis containing hair follicles (Gharibi, Yeganeh, Rezapour-Lactoee, & day 10 which increased to become more compact and mature on day
Hassan, 2015; Jhong et al., 2014). The faster healing and epithelization 14. While the skin tissue section treated with (Ag-PGA/HA)-PVA na-
could be attributed to the promising wound healing action of (HA) nofiber (group II) showed more compact, dense and matured collagen
(Sabra, Ragab, Agwa, & Rohani, 2020). fiber deposit parallel to the epidermis which mimic the normal collagen
On the other hand, the rat's skin section given group III; Garamycin® deposition on normal skin on both day 10 and 14. These results in-
cream showed a reduced size ulcer area on day 10 with early healing as dicated that among the all treated groups (Ag-PGA/HA)-PVA nanofiber
thin squamous epithelial layer and marked infiltration of both epi- (group II) exhibited a higher degree of collagen deposition followed by
dermis and dermis with polymorph neutrophils (PMNLs), inflammatory blank (PGA/HA)-PVA nanofiber (group I).
cells, more fibroblasts and blood vessels with absence of sebaceous
glands and hair follicles (Fig. 7A). On day 14 the skin showed no ulcer
characterized by re-epithelialization with highly distinct epidermis 4. Conclusion and perspectives
(Epi) (external epithelium formed of 2–3 cell layers), and dermis (layer
of connective tissue represented by red arrow). The complete skin re- This research is elucidating the biomedical activity of a novel
generation could be clarified by the presence of many blood vessels and electrospun Polygalacturonic/Hyaluronic acid nanofiber loaded with
the necessary cells like hair follicle and sebaceous gland. On the con- AgNPs. Successful AgNPs synthesis by PGA was verified by UV–vis
trary the skin rat group that applied saline (group IV) showed a greater spectra ranging between 410–415 nm and TEM image of 8.6 nm
ulcer width along with many invasive inflammatory cells, numerous average particulate diameter. SEM confirmed the electrospinning of
polymorph neutrophils (PMNLs), granulation tissues and absence of (Ag-PGA/HA)-PVA in nanoscale with an average diameter of 326 nm.
both sebaceous glands and intact hair follicles at both day 10 and 14. AgNPs component of (Ag-PGA/HA)-PVA nanofiber exhibited a robust
From the previous results it is clear that the histopathological ex- zone inhibition antibacterial activity against gram + ve and gram -ve
aminations for the rat skin are compatible with the results of photo bacteria. While HA component was responsible for the enhanced hy-
documentary analysis for wound healing assessment. Accumulation of drophilicity and strain activities. Additionally, the in-vivo study in al-
bino rat had shown maximum wound epithelization and collagen

9
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

deposition after 14 days of nanofiber administration. Therefore, we are scattering diffusometry. Biomicrofluidics, 10(5), 1–14.
offering an efficient nanofiber for a quick healing of wound infections. D’Autreaux, B., & Toledano, M. B. (2007). ROS as signalling molecules: Mechanisms that
generate specificity in ROS homeostasis. Nature Reviews Molecular Cell Biology, 8(10),
Both the cellular and molecular mechanisms perform a major role in 813–824.
the healing process. The healing quality is greatly governed by reg- Dahlous, K. A., Abd-Elkader, O. H., Fouda, M. M. G., Al Othman, Z., & El-Faham, A.
ulation of gene expression through healing. During different healing (2019). Eco-friendly method for silver nanoparticles immobilized decorated silica:
Synthesis & characterization and preliminary antibacterial activity. Journal of the
stages, a marked change in the level of the gene expression takes place. Taiwan Institute of Chemical Engineers, 95, 324–331.
The most important genes involved in wound healing process includes El-Aassar, M. R., El Fawal, G. F., Kamoun, E. A., & Fouda, M. M. G. (2015). Controlled
vascular endothelial growth factor (VEGF), transforming growth factor drug release from cross-linked κ-carrageenan/hyaluronic acid membranes.
International Journal of Biological Macromolecules, 77, 322–329.
beta (TGF-β) family, epidermal growth factor (EGF) family, interleukin El-Aassar, M. R., Fouda, M. M. G., & Kenawy, E. R. (2013). Electrospinning of functio-
(IL) family, and tumor necrosis factor-α family (TNF-α). At the late nalized copolymer nanofibers from poly(acrylonitrile-co-methyl methacrylate).
stage of healing the EGF family, TGF-β and VEGF are over expressed or Advances in Polymer Technology, 32(1), 1–11.
El-Aassar, M. R., Hafez, E. E., El-Deeb, N. M., & Fouda, M. M. G. (2014).
upregulated on contrary to TNF-α and IL family which is down regu-
Microencapsulation of lectin anti-cancer agent and controlled release by alginate
lated. In the future its expected to study the molecular mechanism of beads, biosafety approach. International Journal of Biological Macromolecules, 69,
the healing process by estimating the level gene expression of the above 88–94.
mentioned biomarkers quantitively by real time PCR in the skin of the El-Hamshary, H., Fouda, M. M. G., Moydeen, M., El-Newehy, M. H., Al-Deyab, S. S., &
Abdel-Megeed, A. (2015). Synthesis and antibacterial of carboxymethyl starch-
rat at the wound area at different healing stages during the healing grafted poly(vinyl imidazole) against some plant pathogens. International Journal of
process after treatment with our developed nanofiber. Biological Macromolecules, 72, 1466–1472.
El-Naggar, M. E., Shaheen, T. I., Fouda, M. M. G., & Hebeish, A. A. (2016). Eco-friendly
microwave-assisted green and rapid synthesis of well-stabilized gold and core–shell
CRediT authorship contribution statement silver–gold nanoparticles. Carbohydrate Polymers, 136, 1128–1136.
El-Rafie, M. H., El-Naggar, M. E., Ramadan, M. A., Fouda, M. M. G., Al-Deyab, S. S., &
M.R. El-Aassar: Conceptualization, Methodology, Funding acqui- Hebeish, A. (2011). Environmental synthesis of silver nanoparticles using hydro-
xypropyl starch and their characterization. Carbohydrate Polymers, 86(2), 630–635.
sition, Writing - original draft. Omar M. Ibrahim: Data curation, Eming, S. A., Krieg, T., & Davidson, J. M. (2007). Inflammation in wound repair:
Formal analysis, Investigation, Writing - review & editing. Moustafa Molecular and cellular mechanisms. The Journal of Investigative Dermatology, 127(3),
M.G. Fouda: Conceptualization, Methodology, Funding acquisition, 514–525.
Forman, H. J., Ursini, F., & Maiorino, M. (2014). An overview of mechanisms of redox
Writing - original draft. Nagham G. El-Beheri: Data curation, Formal signaling. Journal of Molecular and Cellular Cardiology, 73, 2–9.
analysis, Investigation, Writing - review & editing. Mona M. Agwa: Fouda, M. M. G., Abdel-Mohsen, A. M., Ebaid, H., Hassan, I., Al-Tamimi, J., Abdel-
Data curation, Formal analysis, Investigation, Writing - review & Rahman, R. M., et al. (2016). Wound healing of different molecular weight of hya-
luronan; in-vivo study. International Journal of Biological Macromolecules, 89,
editing.
582–591.
Fouda, M. M. G., El-Aassar, M. R., & Al-Deyab, S. S. (2013). Antimicrobial activity of
Declaration of Competing Interest carboxymethyl chitosan/polyethylene oxide nanofibers embedded silver nano-
particles. Carbohydrate Polymers, 92(2), 1012–1017.
Fouda, M. M. G., El-Aassar, M. R., El Fawal, G. F., Hafez, E. E., Masry, S. H. D., & Abdel-
Authors declare that no conflict of interest. Megeed, A. (2015). K-Carrageenan/poly vinyl pyrollidone/polyethylene glycol/silver
nanoparticles film for biomedical application. International Journal of Biological
Macromolecules, 74, 179–184.
Acknowledgements Fouda, M. M. G., EL Naggar, M. E., Shaheen, T. I., & Al Deyab, S. S. (2014). Composition
comprising nanoparticles and a method for the preparation thereof. EP 2604364 B1.
The authors would like to extend their sincere appreciation to the Fouda, M. M. G., Knittel, D., Hipler, U. C., Elsner, P., & Schollmeyer, E. (2006).
Antimycotic influence of β-cyclodextrin complexes - in vitro measurements using
Deanship of Scientific Research at Jouf University for funding the work laser nephelometry in microtiter plates. International Journal of Pharmaceutics, 311(1-
through the research group project No. 40/18. 2), 113–121.
Gharibi, R., Yeganeh, H., Rezapour-Lactoee, A., & Hassan, Z. M. (2015). Stimulation of
wound healing by electroactive, antibacterial, and antioxidant polyurethane/siloxane
References dressing membranes: In vitro and in vivo evaluations. ACS Applied Materials &
Interfaces, 7(43), 24296–24311.
Abdal Dayem, A., Hossain, M. K., Lee, S. B., Kim, K., Saha, S. K., Yang, G. M., et al. (2017). Gopukumar, S., Sana-Fathima, T., Alexander, P., Alex, V., & Praseetha, P. (2016).
The role of reactive oxygen species (ROS) in the biological activities of metallic na- Evaluation of antioxidant properties of silver nanoparticle embedded medicinal
noparticles. International Journal of Molecular Sciences, 18(1), 1–21. patch. Nanomedicine & Nanotechnology Open Access, 1, 1–3.
Abdel-Mohsen, A. M., Abdel-Rahman, R. M., Fouda, M. M. G., Vojtova, L., Uhrova, L., Hafez, E. E., El-Aassar, M., Khalil, K. A., Al-Deyab, S. S., & Taha, T. H. (2011). Poly
Hassan, A. F., et al. (2014). Preparation, characterization and cytotoxicity of schi- (acrylonitrile-co-methyl methacrylate) nanofibers grafted with bio-nanosilver parti-
zophyllan/silver nanoparticle composite. Carbohydrate Polymers, 102(1), 238–245. cles as antimicrobial against multidrug resistant bacteria. African Journal of
Abdelsalam, N. R., Fouda, M. M. G., Abdel-Megeed, A., Ajarem, J., Allam, A. A., & El- Biotechnology, 10(84), 19658–19669.
Naggar, M. E. (2019). Assessment of silver nanoparticles decorated starch and Hayyan, M., Hashim, M. A., & AlNashef, I. M. (2016). Superoxide ion: Generation and
commercial zinc nanoparticles with respect to their genotoxicity on onion. chemical implications. Chemical Reviews, 116(5), 3029–3085.
International Journal of Biological Macromolecules, 133, 1008–1018. Hebeish, A., Abdel-Mohdy, F. A., Fouda, M. M. G., Elsaid, Z., Essam, S., Tammam, G. H.,
Abercrombie, M., Flint, M. H., & James, D. W. (1956). Wound contraction in relation to et al. (2011). Green synthesis of easy care and antimicrobial cotton fabrics.
collagen formation in scorbutic guinea-pigs. Journal of Embryology and Experimental Carbohydrate Polymers, 86(4), 1684–1691.
Morphology, 4(2), 167–175. Hebeish, A., El-Naggar, M. E., Fouda, M. M. G., Ramadan, M. A., Al-Deyab, S. S., & El-
Abu-Saied, M. A., Abdel-Halim, E. S., Fouda, M. M. G., & Al-Deyab, S. S. (2013). Rafie, M. H. (2011). Highly effective antibacterial textiles containing green synthe-
Preparation and characterization of iminated polyacrylonitrile for the removal of sized silver nanoparticles. Carbohydrate Polymers, 86(2), 936–940.
methylene blue from aqueous solutions. International Journal of Electrochemical Ho, J., Walsh, C., Yue, D., Dardik, A., & Cheema, U. (2017). Current advancements and
Science, 8(4), 5121–5135. strategies in tissue engineering for wound healing: A comprehensive review. Advances
auf dem Keller, U., Kumin, A., Braun, S., & Werner, S. (2006). Reactive oxygen species in Wound Care (New Rochelle), 6(6), 191–209.
and their detoxification in healing skin wounds. The Journal of Investigative Jhong, J.-F., Venault, A., Liu, L., Zheng, J., Chen, S.-H., Higuchi, A., et al. (2014).
Dermatology Symposium Proceedings, 11(1), 106–111. Introducing mixed-charge copolymers as wound dressing biomaterials. ACS Applied
Biswas, T. K., Maity, L. N., & Mukherjee, B. (2004). Wound healing potential of Materials & Interfaces, 6(12), 9858–9870.
Pterocarpus santalinus linn: A pharmacological evaluation. The International Journal Khansari, N., Shakiba, Y., & Mahmoudi, M. (2009). Chronic inflammation and oxidative
of Lower Extremity Wounds, 3(3), 143–150. stress as a major cause of age-related diseases and cancer. Recent Patents on
Boer, M., Duchnik, E., Maleszka, R., & Marchlewicz, M. (2016). Structural and biophysical Inflammation & Allergy Drug Discovery, 3(1), 73–80.
characteristics of human skin in maintaining proper epidermal barrier function. Kodati, D. R., Burra, S., & Kumar, G. (2011). Evaluation of wound healing activity of
Postepy Dermatologii I Alergologii, 33(1), 1–5. methanolic root extract of Plumbago zeylanica L. in wistar albino rats. Asian Journal
Bonferoni, M. C., Sandri, G., Rossi, S., Dellera, E., Invernizzi, A., Boselli, C., et al. (2018). of Plant Science and Research, 1(2), 26–34.
Association of alpha tocopherol and Ag sulfadiazine chitosan oleate nanocarriers in Li, C. W., Wang, Q., Li, J., Hu, M., Shi, S. J., Li, Z. W., et al. (2016). Silver nanoparticles/
bioactive dressings supporting platelet lysate application to skin wounds. Marine chitosan oligosaccharide/poly(vinyl alcohol) nanofiber promotes wound healing by
Drugs, 16(2), 1–19. activating TGFbeta1/Smad signaling pathway. International Journal of Nanomedicine,
Clayton, K. N., Salameh, J. W., Wereley, S. T., & Kinzer-Ursem, T. L. (2016). Physical 11, 373–386.
characterization of nanoparticle size and surface modification using particle El-Naggar, M. A., Alrajhi, A. M., Fouda, M. M., Abdelkareem, E. M., Thabit, T. M., &

10
M.R. El-Aassar, et al. Carbohydrate Polymers 238 (2020) 116175

Bouqellah, N. A. (2018). Effect of silver nanoparticles on toxigenic fusarium spp. and Sarhan, W. A., Azzazy, H. M., & El-Sherbiny, I. M. (2016). Honey/chitosan nanofiber
deoxynivalenol secretion in some grains. Journal of AOAC International, 101(5), wound dressing enriched with Allium sativum and Cleome droserifolia: Enhanced
1534–1541. antimicrobial and wound healing activity. ACS Applied Materials & Interfaces, 8(10),
Meier, B., Radeke, H. H., Selle, S., Younes, M., Sies, H., Resch, K., et al. (1989). Human 6379–6390.
fibroblasts release reactive oxygen species in response to interleukin-1 or tumour Selvaraj, S., Thangam, R., & Fathima, N. N. (2018). Electrospinning of casein nanofibers
necrosis factor-alpha. The Biochemical Journal, 263(2), 539–545. with silver nanoparticles for potential biomedical applications. International Journal
Musters, G. D., Burger, J. W., Buskens, C. J., Bemelman, W. A., & Tanis, P. J. (2015). Local of Biological Macromolecules, 120, 1674–1681.
application of gentamicin in the prophylaxis of perineal wound infection after ab- Shaheen, T. I., El-Naggar, M. E., Hussein, J. S., El-Bana, M., Emara, E., El-Khayat, Z., et al.
dominoperineal resection: A systematic review. World Journal of Surgery, 39(11), (2016). Antidiabetic assessment; in vivo study of gold and core-shell silver-gold na-
2786–2794. noparticles on streptozotocin-induced diabetic rats. Biomedecine & Pharmacotherapy,
Nocchi, S., Björklund, S., Svensson, B., Engblom, J., & Ruzgas, T. (2017). Electrochemical 83, 865–875.
monitoring of native catalase activity in skin using skin covered oxygen electrode. Shalaby, M., Agwa, M., Saeed, H., Khedr, S. M., Morsy, O., & El-Demellawy, M. A. (2020).
Biosensors & Bioelectronics, 93, 9–13. Fish scale collagen preparation, characterization and its application in wound
Peng, H.-H., Chen, J.-W., Yang, T.-P., Kuo, C.-F., Wang, Y.-J., & Lee, M.-W. (2011). healing. Journal of Polymers and the Environment, 28(1), 166–178.
Polygalacturonic acid hydrogel with short-chain hyaluronate cross-linker to prevent Textor, T., Fouda, M. M. G., & Mahltig, B. (2010). Deposition of durable thin silver layers
postoperative adhesion. Journal of Bioactive and Compatible Polymers, 26(6), 552–564. onto polyamides employing a heterogeneous Tollens’ reaction. Applied Surface
Pothireddy, S., Kaliki, A., Mekapogu, A. R., Yegireddy, M., Pagadala, E. P., & Prasad, T. N. Science, 256(8), 2337–2342.
(2016). Evaluation of the wound healing efficacy of chemical and phytogenic silver Tian, J., Wong, K. K., Ho, C. M., Lok, C. N., Yu, W. Y., Che, C. M., et al. (2007). Topical
nanoparticles. IET Nanobiotechnology, 10(5), 340–348. delivery of silver nanoparticles promotes wound healing. ChemMedChem, 2(1),
Přichystalová, H., Almonasy, N., Abdel-Mohsen, A. M., Abdel-Rahman, R. M., Fouda, M. 129–136.
M. G., Vojtova, L., et al. (2014). Synthesis, characterization and antibacterial activity Wilkinson, L. J., White, R. J., & Chipman, J. K. (2011). Silver and nanoparticles of silver
of new fluorescent chitosan derivatives. International Journal of Biological in wound dressings: A review of efficacy and safety. Journal of Wound Care, 20(11),
Macromolecules, 65, 234–240. 543–549.
Rath, G., Hussain, T., Chauhan, G., Garg, T., & Goyal, A. K. (2016). Collagen nanofiber Wong, K. K., Cheung, S. O., Huang, L., Niu, J., Tao, C., Ho, C. M., et al. (2009). Further
containing silver nanoparticles for improved wound-healing applications. Journal of evidence of the anti-inflammatory effects of silver nanoparticles. ChemMedChem,
Drug Targeting, 24(6), 520–529. 4(7), 1129–1135.
Rath, G., Johal, E. S., & Goyal, A. K. (2011). Development of serratiopeptidase and me- Yu, K., Zhou, X., Zhu, T., Wu, T., Wang, J., Fang, J., et al. (2016). Fabrication of poly
tronidazole based alginate microspheres for wound healing. Artificial Cells, Blood (ester-urethane) urea elastomer/gelatin electrospun nanofibrous membranes for po-
Substitutes, and Immobilization Biotechnology, 39(1), 44–50. tential applications in skin tissue engineering. RSC Advances, 6(77), 73636–73644.
Sabra, S., Ragab, D. M., Agwa, M. M., & Rohani, S. (2020). Recent advances in electrospun Zahran, M., Ahmed, H. B., & El-Rafie, M. (2014). Alginate mediate for synthesis con-
nanofibers for some biomedical applications. European Journal of Pharmaceutical trollable sized AgNPs. Carbohydrate Polymers, 111, 10–17.
Sciences, 144, 1–22.

11

You might also like