You are on page 1of 15

Research Article

Cite This: ACS Appl. Mater. Interfaces 2018, 10, 16977−16991 www.acsami.org

Silk Sponges Ornamented with a Placenta-Derived Extracellular


Matrix Augment Full-Thickness Cutaneous Wound Healing by
Stimulating Neovascularization and Cellular Migration
Arun Prabhu Rameshbabu,† Kamakshi Bankoti,† Sayanti Datta,† Elavarasan Subramani,‡
Anupam Apoorva,§ Paulomi Ghosh,† Priti Prasanna Maity,† Padmavati Manchikanti,∥ Koel Chaudhury,‡
and Santanu Dhara*,†

Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, ‡School of Medical Science and
Technology, §School of Bio Science, and ∥School of Energy Science & Engineering, Indian Institute of Technology Kharagpur,
Downloaded via INDIAN INST OF TECH KHARAGPUR on November 17, 2019 at 06:18:04 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

Kharagpur 721302, India


*
S Supporting Information

ABSTRACT: Regeneration of full-thickness wounds without scar formation is a multifaceted


process, which depends on in situ dynamic interactions between the tissue-engineered skin
substitutes and a newly formed reparative tissue. However, the majority of the tissue-
engineered skin substitutes used so far in full-thickness wound healing cannot mimic the natural
extracellular matrix (ECM) complexity and thus are incapable of providing a suitable niche for
endogenous tissue repair. Herein, we demonstrated a simple approach to fabricate porous
hybrid ECM sponges (HEMS) using a placental ECM and silk fibroin for full-thickness wound
healing. HEMS with retained cytokines/growth factors provided a noncytotoxic environment in
vitro for human foreskin fibroblasts (HFFs), human epidermal keratinocytes (HEKs), and
human amniotic membrane-derived stem cells to adhere, infiltrate, and proliferate.
Interestingly, HEMS-conditioned media accelerated the migration of HFFs and HEKs owing
to the presence of cytokines/growth factors. Also, the ex vivo chick chorioallantoic membrane
assay of HEMS demonstrated its excellent vascularization potential by inducing and supporting
blood vessels. Additionally, HEMS when subcutaneously implanted demonstrated no severe
immune response to the host. Furthermore, HEMS implanted in full-thickness wounds in a rat model showed augmented healing
progression with well-organized epidermal−dermal junctions via pronounced angiogenesis, accelerated migration of HFFs/
HEKs, enhanced granulation tissue formation, and early re-epithelialization. Taken together, these findings show that porous
HEMS ornamented with cytokines/growth factors having superior physicomechanical properties may be an appropriate skin
substitute for full-thickness cutaneous wounds.
KEYWORDS: extracellular matrix, silk fibroin, cytokine/growth factor, neovascularization, cellular migration,
full-thickness wound healing

1. INTRODUCTION thickness skin injuries is the autologous skin grafting.5


Skin is the largest organ of the human body that serves the However, the limitations such as donor site morbidity and
onerous function of providing a physical shield against limited donor site availability have resulted in the need for the
microorganisms, thermal regulation for average hydration development of skin substitutes. In this context, bioengineered
retention, sensory information about the external environment, skin substitutes (hydrogels,6 sponges,7 and electrospun mats8)
and other vital functions.1 Skin injuries can be caused by from natural polymers and proteins can solve the problem of
genetic disorders, acute trauma, chronic wounds, or by complex autologous donor graft shortage, provide protection from fluid
surgeries.2 In complex full-thickness wounds, epidermal layer loss and contamination, as well as can deliver bioactive factors
along with dermal layer, sweat glands, hair follicles as well as the such as cytokines, dermal matrix components, and growth
underlying subcutaneous fat tissue is damaged. Specifically, factors to the wound bed for enhancing the host wound healing
wounds of critical size hamper the crucial functions of the skin responses.4,9
and can lead to complications such as microbial infection, a Silk fibroin (SF) from Bombyx mori, a versatile natural fibrous
water−electrolyte imbalance in the body, and severe cases that protein, has been extensively explored as a skin substitute
can be life-threatening.3 Therefore, skin injuries of critical size material in tissue engineering applications owing to its
(greater than 1 cm in diameter) require bioactive support with
clinical intervention to accelerate healing.4 Also, scarless wound Received: December 14, 2017
healing is desirable for regaining tissue functionality as well as Accepted: May 2, 2018
improved aesthetics. The gold standard treatment for full- Published: May 2, 2018

© 2018 American Chemical Society 16977 DOI: 10.1021/acsami.7b19007


ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

biocompatibility, slow degradability, low immunogenicity, Further, HEMS and CIMS were assessed for their potential in
remarkable mechanical properties, and hemostatic action.10−12 accelerating full-thickness wound healing using a rat model.
However, SF alone is inadequate for dermal tissue regeneration
because it lacks adequate cell-specific binding sites and limited 2. MATERIALS AND METHODS
growth factor adsorbing capacity.13 Thus, to enhance the 2.1. Decellularization of Human Placenta and Processing of
properties of SF, it has been blended with various natural Soluble ECM (pECM). All the experimental procedures were
macromolecules such as gelatin,14 hyaluronic acid,15 collagen,16 approved by the Institutional Ethical Committee of Indian Institute
of Technology, Kharagpur, India. Decellularization of human placenta
chitosan,17 and so forth. For instance, a scaffold fabricated from was performed according to the protocol reported in our previous
SF/elastin facilitated accelerated re-epithelialization in burn work.24 Briefly, the collected placentas were repeatedly washed with
wounds.18 In another study, the blend scaffold of SF/keratin Dulbecco’s phosphate-buffered saline (DPBS) until the blood was
significantly enhanced the cell adhesion/proliferation of L929 removed and then minced into small fragments by using a sterile
fibroblasts and increased the extracellular matrix (ECM) scalpel. The minced placenta was decellularized using 0.5% sodium
deposition of collagen type I.19 Though the scaffolds dodecyl sulfate (SDS; Sigma-Aldrich, USA) with 0.2% DNase (2000
mentioned above displayed superior cellular activities and U; Sigma-Aldrich, USA), 200 mg/mL RNase (Sigma-Aldrich, USA),
0.05% trypsin/ethylenediaminetetraacetic acid (EDTA; Gibco, USA),
ECM deposition, yet they lack a plethora of structural proteins, 100 U/mL penicillin, 100 mg/mL streptomycin (Gibco, USA), and 1
growth factors, and cytokines required for skin regeneration. mM phenyl methyl sulfonyl fluoride (Sigma-Aldrich, USA) in a sealed
In recent years, decellularized ECM has emerged as an rotating vessel. The procedure was conducted under strictly sterile
attractive biomaterial in regenerative medicine, which provides conditions, and the solution was changed every 24 h to prevent tissue
abundant biological cues for cell migration and proliferation degradation and contamination. The decellularized placentas were
and further directs/promotes differentiation.20 The human washed with PBS and stored at −80 °C for further processing.
placenta is a unique, complex organ that serves multiple Solubilization of ECM was performed as described elsewhere.25
functions to the developing fetus; also, it is a rich reservoir for a Briefly, the decellularized human placenta was pulverized to a powder
using a tissue homogenizer in the presence of liquid nitrogen. The
variety of growth factors and cytokines such as epidermal resulting powder was immersed and stirred in a 4 M urea buffer (240 g
growth factor (EGF), transforming growth factor-β (TGF-β), urea, 9 g NaCl, and 6 g Tris base in 1 L distilled water) containing a
fibroblast growth factor (FGF), platelet-derived growth factor protease inhibitor cocktail for 24 h. Subsequently, the samples were
(PDGF), and vascular endothelial growth factor (VEGF).21 subjected to ultrasonic homogenization (Branson Ultrasonics, USA) in
These growth factors/cytokines play an essential role in an ice bath. After centrifuging the samples at 14 000 rpm for 20 min at
migration/proliferation of fibroblasts/keratinocytes, homing of 4 °C, the supernatant was collected and then dialyzed using an 8000
mesenchymal stem cells, re-epithelialization, and neovasculari- molecular weight cutoff (MWCO) dialysis tubing against Tris-buffered
saline (TBS), that is, 9 g NaCl and 6 g Tris base in 1 L distilled water
zation that are considered essential for tissue repair and to remove urea. The contents of the dialysis tubes were centrifuged
regeneration in the early stage of wound healing. Direct again to remove protein aggregates, and then the supernatant was
administration of the human placental extract in the wound frozen at −80 °C, freeze-dried for 42 h, and pulverized to obtain
margin of full-thickness wounds was found to accelerate the pECM.
wound healing mechanism associated with an increase in TGF- 2.2. Quantification of Collagen and Glucosaminoglycans
β levels during the initial phase, resulting in increased (GAGs). The total collagen content was assessed by the hydroxypro-
inflammatory cell infiltration, and increased VEGF levels during line assay, and the sulfated GAG (sGAG) content was assessed by the
alcian blue assay26 in native placenta (NP) and pECM, as detailed in
the later stages, leading to increased new blood vessel the Supporting Information.
formation.22 Furthermore, the properties of the placenta, such 2.3. Cytokine Array for Detection of Growth Factors. Growth
as low immunogenicity, anti-inflammatory, and anti-scarring factors in NP and pECM were analyzed using a human cytokine
make it an ideal choice to treat full-thickness skin wounds.23 antibody array (RayBio C-Series C1000, USA) following the
In the present work, a hybrid ECM sponge (HEMS) was manufacturer’s protocol. Briefly, the samples were dissolved at 4 °C
fabricated for skin tissue engineering by incorporating a for 36 h in the buffer containing 0.1× protease inhibitor, 2 M urea, and
placenta-derived ECM (pECM) with SF, thus bringing together 50 mM Tris-HCl. The supernatant was collected by centrifugation at 4
°C. The obtained supernatant was incubated onto an array chip
the inherent advantages of both SF and pECM. Collagen (the
containing 120 different human cytokine antibodies after blocking.
major ECM component of native skin) is considered to be the The chip was then washed and incubated with biotinylated antibody
promising material of choice for wound healing applications cocktail. Subsequently, the signals were detected using a chemilumi-
owing to its biodegradability, superior biocompatibility, low nescence detection system after horseradish peroxidase (HRP)−
antigenicity, nontoxicity upon exogenous application, and high streptavidin incubation and washing.
tensile strength. Therefore, collagen was selected as a 2.4. SDS-Polyacrylamide Gel Electrophoresis (PAGE) and
representative control to demonstrate the superiority of Western Blotting. The finely powdered NP and pECM were
obtained by homogenizing in a buffer containing 65 mM dithiothreitol,
pECM containing intrinsic cytokines/growth factors for 4 M guanidine HCl, protease inhibitor cocktail, 10 mM EDTA, and 50
wound healing. Hence, a collagen-incorporated SF matrix mM sodium acetate (all Sigma-Aldrich, USA). The resulting mixture
sponge (CIMS) was also fabricated for comparative studies. was exposed to ultrasonic homogenization, and the supernatant was
The fabricated HEMS and CIMS were characterized for their collected after centrifugation (13 000 rpm for 20 min at 4 °C).
physicomechanical properties and evaluated for their bio- Bicinchoninic acid assay kit (Thermo Scientific, Rockford, USA) was
compatibility using the primary fibroblasts, keratinocytes, and used to quantify the total protein concentration of the homogenates
human amniotic membrane-derived stem cells (HAMSCs). In according to the manufacturer’s instruction. The extracted proteins
from NP and pECM were run in 10% PAGE and stained with
vitro wound healing/migration potential was evaluated by the
Coomassie blue after fixing.
scratch assay, and the ability to induce vascularization was The extracted proteins (40 μg) from NP and pECM after resolving
assessed using the chick chorioallantoic membrane (CAM) in SDS-PAGE were transferred onto the nitrocellulose membrane
model. Also, HEMS and CIMS were implanted subcutaneously (Millipore, USA) at 90 V for 2 h in the presence of a Tris-glycine
in rats to study the host immune reaction/toxicity in vivo. buffer. The nitrocellulose membranes containing the transferred

16978 DOI: 10.1021/acsami.7b19007


ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

proteins were treated with a primary antibody [hepatocyte growth where Td and Ts are the dry and wet weight of CIMS/HEMS,
factor (HGF; Santa Cruz, USA), TGF-β1 (Abcam, USA), VEGFA respectively. The experiments were conducted in triplicate and
(Abcam, USA), EGF (Santa Cruz, USA), insulin-like growth factor-1 averaged.
(IGF-1; Santa Cruz, USA), and PDGF-B (Santa Cruz, USA)] at 4 °C In vitro degradation of CIMS/HEMS (n = 5) was performed in PBS
overnight after blocking. The blots were subsequently incubated with containing 1 U/mL of lysozyme (hen egg-white, Sigma-Aldrich), pH
the HRP-conjugated secondary antibody for 1 h. Pierce ECL western 7.4 at 37 °C for a specified period according to the protocol described
blotting substrate kit (Thermo Scientific, USA) was used for in our previous publication.29
visualizing the immunoreactive proteins according to the manufac- 2.10. Fourier Transform Infrared (FTIR) Spectroscopy. The
turer’s instructions. Further, an automatic X-ray film processor was infrared spectra of CIMS and HEMS were obtained using a Thermo
used to develop the images, which were analyzed by ImageJ software Nicolet spectrophotometer (model NEXUS-870; Thermo Nicolet
(Rasband WS; NIH). Corporation, Madison, WI) in attenuated total reflection mode. The
2.5. Preparation of SF Solution. SF solution was prepared absorbance of spectra was recorded in the range of 4000−500 cm−1.
according to the procedure described elsewhere.27 Briefly, B. mori 2.11. Mechanical Testing. The mechanical properties of the
cocoons were chopped into small pieces and boiled (98 °C) for 30 samples (n = 10) were determined by tensile testing using the
min in an alkaline bath containing 0.02 M sodium carbonate (Sigma- universal testing machine (25 K, Hounsfield, UK) at room
Aldrich, USA) to obtain fibroin and to remove the sericin/ temperature with a 20 N load cell. The samples were cut into a
glycoproteins. The degummed silk was rinsed thoroughly with rectangular strip (∼5 mm width, ∼20 mm length, ∼1 mm thickness)
double-distilled water and allowed to dry at room temperature and soaked in PBS for 48 h. Uniaxial tensile testing was performed
overnight. The extracted fibroin was dissolved in 9.3 M LiBr (Sigma- under tension at a crosshead speed of 2 mm/min.
Aldrich, USA) solution at 60 °C for 4 h followed by dialysis for 72 h 2.12. Isolation of HAMSCs/HFFs/HEKs. HAMSCs were isolated
against double-distilled water using a dialysis membrane (3500 and characterized according to the protocol reported in our previous
MWCO) to remove LiBr. The obtained fibroin solution was publication.24 Briefly, the human placentas collected under stringent
centrifuged (9000 rpm at 4 °C for 20 min) to remove large sterile conditions were transported to the laboratory at 4 °C. The
aggregates, and the supernatant was concentrated to a final blood components were removed by rinsing with Hank’s balanced salt
concentration of 10% (w/v) by dialysis against poly(ethylene glycol) solution (Gibco, USA) containing 200 mg/mL streptomycin and 200
(10 000 MW, Sigma-Aldrich, USA). The concentrated fibroin solution U/mL penicillin. The amniotic membrane was detached from chorion
was passed through a 5 μm syringe filter (Millipore) to eliminate by a blunt dissection and was incubated with 0.05% trypsin−EDTA
minor impurities. solution (Gibco, USA) for two cycles of 30 min each, followed by
2.6. Fabrication of CIMS and HEMS. Collagen type I was discarding the supernatant. Subsequently, the tissue was digested with
extracted from fish scales according to the procedure described Earle’s balanced salt solution (Gibco, USA) containing 2 mg/mL
elsewhere28 and was grounded to powder in the presence of liquid collagenase Type IV (Gibco, USA) and 10 U/mL DNase I (Sigma-
nitrogen. Fibroin solution and pECM were obtained as described Aldrich, USA) for 60 min. After the end of the digestion, the
above. For fabrication of CIMS/HEMS, 0.25 g of pulverized collagen suspension was centrifuged at 1500 rpm to collect the cell pellet. The
type I/pECM, respectively, was dispersed in 5 mL of 10% (w/v) SF cell pellet was seeded in flasks (Nunc, USA) after suspending it in the
solution under constant stirring for 24 h at 4 °C. The homogeneously Dulbecco’s modified Eagle’s medium (DMEM; containing low
mixed viscous solution was subjected to sonication in an ice bath for 2 glucose, 10% fetal bovine serum (FBS), 1% antibiotic−antimycotic)
min, gently poured into sterile molds, frozen at −80 °C, and and transferred to an incubator. TrypLE Express Enzyme (Gibco,
subsequently lyophilized for 72 h to obtain CIMS/HEMS. The USA) was used to subpassage the cells, and cells before passage
lyophilized CIMS/HEMS was soaked in 70% (v/v) ethanol for 6 h to number five were used for experiments.
induce formation of β-sheets in the SF protein. Homogeneous Human foreskin fibroblasts (HFFs) and human epidermal
distribution of pECM or collagen in the SF matrix was assessed by keratinocytes (HEKs) were isolated as described elsewhere from
Masson’s trichrome (MT) staining as described in the Supporting circumcised human foreskin.30 Briefly, skin samples were collected in
Information. sterile containers and rinsed in PBS containing 200 mg/mL
2.7. Morphological Analysis by Scanning Electron Micros- streptomycin and 200 U/mL penicillin. Later, the samples were
copy. The microstructure of CIMS/HEMS was examined under a chopped into small pieces and incubated in Dispase II (Sigma-Aldrich,
scanning electron microscope (EVO ZEISS, Carl Zeiss SMT AG, USA) for overnight at 4 °C. Subsequently, epidermal and dermal
Oberkochen, Germany) at an accelerating voltage of 10−20 kV after layers were separated and incubated in TrypLE Express Enzyme/
fixing the samples with 4% paraformaldehyde. Briefly, the dried collagenase I (Gibco, USA), respectively, for further digestion. The
samples were placed on a sample holder using double-sided adhesive corresponding cell suspension was strained, and the cell pellet was
tapes and gold-coated for 90 s using a plasma coater under high obtained by centrifugation. Defined keratinocyte-serum-free media
vacuum to avoid the charging effect. (SFM) (Gibco, USA)/DMEM high glucose media (Gibco, USA) were
2.8. Porosity Measurement by Micro-Computed Tomog- used for culturing HEK/HFF, respectively. Cells after reaching 80%
raphy (Micro-CT). CIMS/HEMS was scanned (1000 scan slices/ confluency were passaged with TrypLE Express, and cells before
sample) using a micro-CT (GE Phoenix v|tom|ex s, Germany) at a passage five were used for further experiments.
voltage of 155 kV and a current of 45 mA, with a voxel of 7.2 μm. 2.13. Indirect Cytotoxicity Testing. 2.13.1. Preparation of
When analyzing the porosity of the samples, the threshold to be used HEMS-/CIMS-Conditioned Medium. HEMS/CIMS in the presence of
was obtained for each specimen by the threshold histogram offered by liquid nitrogen was grounded to a fine powder with a sterile porcelain
the VG Studio Max software (Volume Graphics Germany). mortar and pestle. The resulting HEMS/CIMS powder (25 mg/mL)
2.9. Swelling Behavior and Degradation Kinetics. The was added to a complete DMEM/defined keratinocyte-SFM medium
swelling behavior of CIMS and HEMS was examined for 64 h at 37 with 1% antibiotic−antimycotic in a sterile container and stirred inside
°C in sterile PBS (pH 7.4). At predetermined time intervals, the excess an incubator at 37 °C and 5% CO2 for 24 h. The conditioning material
PBS in samples was removed by gently wiping with a filter paper and was removed from the media by filtering the medium through a 0.22
then instantly weighed in an electronic balance (Mettler-Toledo μm syringe filter (Millipore, USA).
International Inc., USA) to calculate the PBS content in swollen 2.13.2. Morphological and Apoptosis Assessment. Primary HFFs
CIMS/HEMS. The swelling ratio was calculated using the formula and HEKs were used for investigating the effect of the CIMS-/HEMS-
conditioning medium on their morphological and functional attributes.
below
Briefly, HFFs/HEKs were cultured either with CIMS-/HEMS-
Ts − Td conditioned or normal medium on lysine-coated coverslips placed in
Swelling ratio (%) = × 100 a 12-well plate. The coverslips were removed after 96 h, stained with
Td rhodamine phalloidin (Invitrogen, USA) and DAPI (Invitrogen, USA)

16979 DOI: 10.1021/acsami.7b19007


ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

according to the manufacturer’s instructions. Apoptosis of HEKs in the g; males; n = 5). The experimental protocol followed was permitted by
presence of CIMS-/HEMS-conditioned media was also investigated the Institutional Ethical Committee of Indian Institute of Technology,
after 7 days of cultivation using the DeadEnd Fluorometric TUNEL Kharagpur, and all surgery was performed under anesthesia. Extreme
system (Promega, USA) according to the manufacturer’s guidelines. caution was taken to minimize the suffering of the animals. Cylindrical
2.13.3. Scratch Assay. The scratch assay for wound healing was CIMS/HEMS (diameter = ∼1 cm; height = ∼0.5 cm) was implanted
performed according to the protocol described elsewhere.31 HFFs/ on the dorsal side of rats after dissection and sutured using chromic
HEKs were cultured in 35 mm Petri dishes (Tarsons, India) in catgut under aseptic condition. After 28 days of implantation, the rats
DMEM/defined keratinocyte-SFM medium at 37 °C in a 5% CO2 in were euthanized, and the site of implantation along with the adjacent
an air atmosphere to produce confluent monolayers. To mimic tissue was retrieved for further analysis. The harvested samples were
wounds, a vertical scratch was made in the center of the monolayer fixed, dehydrated, and subsequently embedded in paraffin blocks. The
HFFs/HEKs using a sterile 200 μL pipette tip, and subsequently, the paraffin blocks containing the sample were microtomed, and the
Petri dishes were washed with sterile PBS to remove the detached sections were then stained with H&E, MT, toluidine blue (TB; Sigma-
HFFs/HEKs. Later, the Petri dishes were filled with either CIMS-/ Aldrich, USA), and anti-CD31 antibody (Abcam, USA) staining
HEMS-conditioned medium or control medium, and the Petri dishes according to the manufacturer’s instructions. All the sections were
were incubated at 37 °C in a 5% CO2 for 72 h. The migration of observed under a microscope to understand the cellular events. For
HFFs/HEKs in the denuded path was monitored by taking optical organ toxicity analyses, the major organs (heart, kidney, lung, and
images using an inverted phase contrast microscope (Carl Zeiss liver) were harvested (n = 3 per group) after 28 days of the treatment
AxioObserver) at the different period, and three independent period of different groups and processed for the histological
observers counted the number of cells that had migrated into the assessment of the tissue.
initially cell-free scratch area. 2.18. Full-Thickness Cutaneous Wound Healing Study. The
2.14. Analysis of HEMS-Conditioned Medium by ELISA. The in vivo experimental protocols were approved by the Institutional
quantitative direct enzyme immunoassay was performed to estimate Ethical Committee of Indian Institute of Technology, Kharagpur.
the levels of interleukin-6 (IL-6), interleukin-8 (IL-8), monocyte Albino Wistar rats (2 months old; 200 ± 10 g) were used to evaluate
chemotactic protein-1 (MCP-1), TGF-β1, and RANTES in HEMS- the full-thickness skin wound healing capacity of the different groups.
conditioned medium. The protein concentration of 20 μg/mL was Randomly, the rats were divided into three groups (n = 9 each group)
used for antigen binding in poly-L-lysine-coated 96-well plates. according to the treatments received on each rat. Three treatment
Following blocking with 3% bovine serum albumin (BSA), the wells groups were as follows: (1) SHAM: wounds that were left untreated
were incubated with antibodies against IL-6, IL-8, MCP-1, TGF-β1, and (2) CIMS and (3) HEMS were applied into the wound exclusively
and RANTES (Santa Cruz Biotechnology, Inc., USA). All wells were within the wound area. Before surgery, 2 cm diameter wounds were
washed with (Phosphate Buffered Saline with Tween 20) (PBST) and marked using a template, and standardized full-thickness skin wounds
incubated with a suitable secondary antibody conjugated with HRP were made on the dorsal side by excising to the level of the fascia, by
(Santa Cruz Biotechnology, Inc. CA, USA). After washing with PBST, using sterile forceps and scissors under general anesthesia. Petrolatum
100 μL of Femto-ELISA-HRP substrate (G-Biosciences, USA) was gauze pressure dressing was applied to all the groups, which prevented
added to each well and incubated for 10−15 min. The reaction was the samples from moving away from the wound bed. Care was taken
stopped with 1 N HCl, and the absorbance was measured at 450 nm. to ensure that the pressure dressing was not detached during the 21
2.15. Direct Cytotoxicity Testing. The cytotoxicity was tested by day treatment period. The rats were individually housed in
the direct cultivation of HAMSCs/HEKs/HFFs on TCP/CIMS/ temperature-controlled cabins and fed with a standard protein diet.
HEMS. Briefly, 5 × 104 HAMSCs/HFFs/HEKs were seeded on The cutaneous wounds were photographed after 0, 7, 14, and 21 days,
samples (n = 5) and cultivated in a 24-well tissue culture plate for 72 h. and the unhealed area was measured using ImageJ (Rasband WS;
The number of viable cells was calculated using Vybrants MTT Cell NIH) to assess the wound healing kinetics. The percentage of wound
Proliferation Assay Kit (Invitrogen, USA) according to the reduction was calculated according to the following formula
manufacturer’s instructions.
The viability of HEKs seeded on CIMS and HEMS was assessed A0 − At
Rate of wound closure = × 100
using Live/Dead Assay Kit (Life Technologies, NY) according to the A0
manufacturer’s protocol. Briefly, cell-seeded CIMS/HEMS after 7 days
was incubated for 60 min at 37 °C in a solution containing 2 μM where A0 and At are designated as the initial wound area and wound
calceinacetomethoxy (AM) and 4 μM ethidium homodimer. The area at the designated time, respectively.
samples were repeatedly washed with DPBS to avoid background The rats were sacrificed after 7, 14, and 21 days post-implantation,
staining and visualized under an inverted fluorescence microscope and the wound site with the surrounding muscle and skin was
(AxioVision, Zeiss, Germany). retrieved. They were then immediately fixed with 4% formaldehyde,
2.16. CAM Assay. The CAM assay was used as an ex vivo model to dehydrated, and embedded in a paraffin block. Sections of 3 μm
assess the potential of CIMS/HEMS to induce vascularization. In this thickness were cut and stained with H&E and MT. Further, the
study, fertilized white Leghorn chicken eggs were used for the CAM sections were also immune-stained with anti-CD31 and anti-
assay by following the procedure described elsewhere.32 Briefly, the cytokeratin 10 (CK-10; Abcam, USA) according to the manufacturer’s
eggs were incubated at 37 °C, 65% relative humidity egg incubator. On instruction. The photomicrograph of different stains was captured and
day 5, disks (5 mm diameter; n = 5) of CIMS and HEMS were examined to document cardinal features such an inflammatory
punched, sterilized, and hydrated with sterile PBS overnight. The response, necrosis, neovascularization, collagen deposition, granula-
sterile disk of CIMS and HEMS was placed over CAM, respectively; tion, and re-epithelialization to understand healing progression at
later, the eggshell was sealed. On day 8, the window was carefully different periods.
dissected, and CIMS/HEMS was photographed in situ. Three blind 2.19. Reverse Transcriptase-PCR (RT-PCR) Analyses of the
observers counted the number of blood vessels approaching toward Wound Area. RT-PCR was performed on a newly formed tissue in
the scaffolds. Further, the CIMS/HEMS along with the surrounding the wound area from different treatment groups, 21 days post-surgery.
membrane was retrieved carefully with forceps without getting tore off, The regenerated tissues (n = 5 per group) from different study groups
fixed in 4% paraformaldehyde, dehydrated, and subsequently were excised from the animal after sacrificing and ground to a powder
embedded in paraffin blocks. The paraffin blocks were sectioned and using liquid nitrogen. The total RNA was extracted using the TRIzol
stained with hematoxylin and eosin (H&E; Sigma-Aldrich, USA), MT reagent (Invitrogen, USA) according to the manufacturer’s instruc-
(Sigma-Aldrich, USA), and DAPI. tions. An equal quantity of isolated RNA was transcribed into cDNA
2.17. In Vivo CIMS/HEMS Cellular Response and Organ using a cDNA synthesis kit (Thermo Scientific, USA) according to the
Toxicity Analysis. The in vivo cellular response of CIMS/HEMS was manufacturer’s protocol. PCR amplification of the gene-specific
studied by subcutaneous implantation in albino Wistar rats (250 ± 10 primers (Supporting Information Table S1) was performed in a

16980 DOI: 10.1021/acsami.7b19007


ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 1. Profiling of cytokines/growth factors in NP and pECM. Y-error bars represent standard deviation.

Figure 2. (A) Total protein extracted from NP and pECM was separated by SDS-PAGE; (B) immunoblotting using different antibodies (anti-EGF,
anti-PDGF-B, anti-TGF-β1, anti-VEGFA, anti-IGF-1, and anti-HGF) and (C) their respective relative band intensities. Y-error bars represent
standard deviation, and single asterisks signify P < 0.05.

thermal cycler (Eppendorf Mastercycler, USA), and the PCR product 3. RESULTS
was imaged in UV gel doc (Bio-Rad, USA) after running it in 1%
agarose gels. The band intensity was analyzed using ImageJ (Rasband
3.1. Biochemical Characterization of Soluble Placenta
WS; NIH). ECM (pECM). The NP was subjected to decellularization using
2.20. Statistical Analysis. The data were analyzed using a combinatorial treatment involving homogenization, centrifu-
GraphPad Prism software (version 5.02, La Jolla, CA, USA) by one- gation, SDS, and nucleases. For the preparation of soluble
way analysis of variance and Tukey’s multiple comparison tests. The placenta ECM proteins (pECM), the ECM from the
level of significance was determined as P < 0.05 significance. decellularized placenta was solubilized using urea. To estimate
Experiments were repeated in triplicates, and data were represented the biochemical preservation in pECM after the decellulariza-
as mean ± standard deviation (SD) for n = 3 unless mentioned. tion and the solubilization process, pECM was quantified for
16981 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 3. SEM micrographs of (A) CIMS and (C) HEMS; micro-CT analysis of (B) CIMS and (D) HEMS.

Figure 4. (A) Swelling kinetics of CIMS and HEMS at 37 °C in PBS; (B) degradation profile of CIMS and HEMS incubated in PBS containing
lysozyme; (C) FTIR analysis of CIMS and HEMS; and (D) tensile testing of CIMS and HEMS incubated in PBS for 48 h (P > 0.05). Y-error bars
represent standard deviation.

major ECM components such as collagen and sGAG. As 3.2. Cytokine Array for Detection of Growth Factors.
observed in Supporting Information (Figure S1), retention of pECM and NP were arrayed on a glass chip containing 120
collagen (409.8 ± 45.44 μg mg−1) and sGAG (38.45 ± 7.42 μg different cytokine antibodies for detection of the retained
mg−1) in pECM after the decellularization and solubilization endogenous cytokines. As shown in Figure 1, 84 cytokines were
process was not significantly (P > 0.05) different from the readily detected in pECM, and the remaining 36 cytokines were
collagen (460.4 ± 18.622 μg mg−1) and sGAG (53.59 ± 14.41 not detected, which may be due to the sensitivity limitations of
μg mg−1) content of NP. These results demonstrated that the the array. Among the 84 detected bioactive molecules,
major ECM components were sufficiently retained in pECM cytokines [EGF, IGF-1, PDGF-B, HGF, VEGFA, TGF-β1,
after the decellularization and solubilization process, which will and FGF] which are considered critical for regulating the
make it an ideal matrix for tissue engineering applications. angiogenesis and skin wound healing were detected at high
16982 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

levels. Also, growth factors such as IL-6, IL-8, MCP-1, and HEKs were cultivated on lysine-coated coverslips with either
RANTES, which are reported to accelerate fibroblast and CIMS-/HEMS-conditioning medium or control medium for 96
keratinocyte migration and proliferation, were also detected in h. As shown in Figure 5A, HFFs/HEKs grown in HEMS-
pECM.
3.3. SDS-PAGE and Western Blotting of NP and pECM.
SDS-PAGE was used to investigate the proteins retained in
pECM. Figure 2A reveals some bands at lower molecular
weights signifying the presence of various proteins/peptides in
the pECM, which demonstrates the complexity of the pECM.
Western blotting was performed to confirm the presence of
endogenous bioactive molecules that are known to regulate
blood vessel formation and skin regeneration. Figure 2B reveals
the presence of retained growth factors such as EGF, IGF-1,
PDGF-B, HGF, VEGFA, and TGF-β1 after the decellulariza-
tion and solubilization process in pECM. From Figure 2C, no
significant difference (P > 0.05) was observed in the levels of
EGF, PDGF-B, TGF-β1, and VEGFA in pECM compared to
that in NP. However, a significant decrease (P < 0.05) was
observed in the levels of IGF-1 and HGF in pECM compared
to that in NP (Figure 2C).
3.4. Physicochemical Characterization of Scaffolds. In
this study, collagen I/pECM was blended with SF in aqueous
environment and successfully fabricated into CIMS/HEMS,
respectively, by molding and freeze-drying process. The
scanning electron microscopy (SEM) microstructures of
CIMS and HEMS are shown in Figure 3A,C, which revealed
a high degree of interconnected heterogeneous porous
structures within the matrices. The porosity of the samples
analyzed by micro-CT was approximately similar for CIMS
(85.27%) and HEMS (90.2%) as shown in Figure 3B,D. The
swelling behavior of the scaffolds is critical not only for
arbitrating the rate of nutrition and waste transport within the
scaffolds in situ, but it also helps in biological fixation to the Figure 5. (A) Rhodamine phalloidin−DAPI images of HFFs and
wound bed. As observed in Figure 4A, CIMS and HEMS HEKs cultivated in CIMS- and HEMS-conditioned medium (scale bar
demonstrated 55 and 64% swelling, respectively, when exposed represents 50 μm, red represents nucleus staining DAPI and green
to PBS for 0.5 h because of the rapid uptake of water. After this depicts cytoskeleton expression); (B) TUNEL assay images of HEKs
time, it attained a plateau, and no further increase in mass was cultivated in CIMS- and HEMS-conditioned medium (scale bar
observed until 64 h. Extended long-term swelling observations represents 50 μm).
are essential for more critical assessment. As shown in Figure
4B, CIMS (mass loss 97%) exhibited faster degradation as conditioned medium proliferated rapidly and were able to make
compared to HEMS (mass loss 90%) when incubated in intermittent contact via cellular protrusions and extensions with
lysozyme at 37 °C for 16 days. Also, CIMS and HEMS when well-spread cytoskeletons compared to those of HFFs/HEKs
incubated in PBS (pH 7.4) without lysozyme for 48 h showed a cultured in CIMS-conditioned/control medium. Also, there was
negligible mass loss (data not shown) and displayed a wet no difference in the morphology of HFFs/HEKs cultured in the
tensile strength of 0.065 ± 0.004 and 0.076 ± 0.003 MPa (P > CIMS-conditioned medium compared with the cells cultured in
0.05; Figure 4D). control medium. These observations demonstrated that CIMS/
Infrared absorption spectra (Figure 4C) of CIMS and HEMS HEMS did not release cytotoxic substances.
showed characteristic absorption peaks assigned to the peptide The TUNEL assay was performed to detect apoptosis after 7
bonds (−CONH−) that give rise to amide I (1600−1700 cm), days of HEKs’ cultivation either in control or in CIMS-/
amide II (1520−1540 cm), and amide III (1220−1300 cm) HEMS-conditioned medium. As shown in Figure 5B, no
signature peaks. Pure silk scaffolds showed amide I, amide II, significant apoptotic HEKs were observed in control or CIMS-/
and amide III bands at 1617, 1511, and 1226 cm−1; CIMS HEMS-conditioned medium treated groups, indicating that the
scaffolds showed amide I, amide II, and amide III bands at CIMS-/HEMS-conditioned medium had no detrimental effect
1636, 1528, and 1226 cm−1, whereas HEMS scaffolds showed on cells.
the characteristic peaks of amide I, amide II, and amide III 3.5.2. Scratch Assay. HFFs started to migrate from the
bands at 1623, 1533, and 1222 cm−1. Also, in CIMS and edges of the denuded/wounded areas within 6 h, whereas HEK
HEMS, the peak at 2869−1 corresponds to C−H symmetric migration was witnessed after 12 h in both the treatment
stretching. Notably, in CIMS, the 1636 cm−1 band is (CIMS-/HEMS-conditioned medium) and control groups
characteristic of the triple helix of native collagen and thus (Figure 6A,B). As determined by three independent observers,
suggests the existence of collagen in the native triple helical the numbers of HFFs that migrated into the scratch areas for
structures in the CIMS scaffold. HEMS-conditioned, CIMS-conditioned, and control group
3.5. Indirect Cytotoxicity Testing for CIMS and HEMS. were found to be (6 h): 134 ± 06, 86 ± 12, and 50 ± 05
3.5.1. Morphological and Apoptosis Assessment. HFFs and and (12 h): 295 ± 16, 160 ± 14, and 110 ± 06, respectively
16983 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 6. Scratch assay images of (A) HFFs and (B) HEKs cultivated in CIMS- and HEMS-conditioned medium at different time durations (scale
bar represents 100 μm); cell migration quantification of (C) HFFs and (D) HEKs. Y-error bars represent standard deviation, and triple asterisks
signify P < 0.001.

Figure 7. (A) Growth factors and cytokines detected in HEMS-conditioned medium by ELISA multiarray; (B) cellular metabolic activity of HFFs,
HEKs, and HAMSCs cultured in CIMS/HEMS according to the MTT assay; live−dead cell staining of HEKs cultivated in (C) CIMS and (D)
HEMS. Y-error bars represent standard deviation, and scale bar represents 50 μm.

(Figure 6C). Similarly, the numbers of HEKs that migrated into 3.6. Analysis of HEMS-Conditioned Medium by ELISA.
the scratch areas for HEMS-conditioned, CIMS-conditioned, The HEMS-conditioned media were assayed by ELISA to
and control group were (12 h): 313 ± 11, 208 ± 12, and 81 ± evaluate the presence of growth factors (IL-6, IL-8, MCP-1,
06 and (24 h): 654 ± 16, 355 ± 9, and 268 ± 08, respectively TGF-β1, and RANTES; known to accelerate migration and
(Figure 6D). It can be interpreted from Figure 6C,D that the proliferation of HFFs and HEKs) released from HEMS during
number of migrated HFFs/HEKs was more significant (p < the conditioning process. From Figure 7A, it can be observed
0.05) in the HEMS-conditioned medium treated group that detectable levels of MCP-1, TGF-β1, RANTES, IL-6, and
compared to that in other groups. IL-8 were present in the HEMS-conditioned media.
16984 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 8. Proangiogenic property analysis of CIMS and HEMS by the CAM assay; (A) macroscopic view, (B) quantification of vessels converging
toward the scaffolds, and (C) histological analysis of the retrieved CIMS and HEMS after 72 h of incubation (scale bar represents 100 μm). *
represents the implanted scaffold; Y-error bars represent standard deviation, and triple asterisks signify P < 0.001.

Figure 9. H&E, MT, TB, and anti-CD31 of the explanted CIMS/HEMS on day 21 after subcutaneous implantation (scale bar represents 50 μm;
blue represents nucleus staining DAPI, and green depicts antibody expression).

3.7. Direct Cytotoxicity Testing for CIMS and HEMS. HFFs, HEKs, and HAMSCs, which indicates the potential of
The 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bro- CIMS/HEMS in skin tissue engineering applications. Interest-
mide (MTT) cytotoxicity assay was performed to assess the ingly, the cells cultivated in HEMS displayed increased cellular
remnant toxic chemical residues that may be present in CIMS metabolic activity compared to that in CIMS and TCP,
and HEMS during the process of extraction/decellularization demonstrating its cytocompatibility. This increased cellular
and solubilization. As shown in Figure 7B, it was evident that metabolic activity in HEMS may be attributed to the presence
CIMS and HEMS supported the proliferation and viability of of cytokines/growth factors. Also, CIMS displayed higher cell
16985 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 10. Healing progression of full-thickness cutaneous wounds treated with SHAM, CIMS, and HEMS; (A) photographs of wounds and (B)
rate of wound closure on days 0, 7, 14, and 21; (C-D) RT-PCR analysis of skin wounds treated with SHAM, CIMS, and HEMS at 21 day
postwounding. Y-error bars represent standard deviation, single asterisks signify P < 0.05, and triple asterisks signify P < 0.001.

viability compared to TCP, owing to the 3D architecture of the wound healing process. From MT staining, no capsular layer/
scaffold. fibrosis was found, and dense collagen fiber deposition was
The Live/Dead assay was used to determine the adhesion visualized in the interfacial layer between the host and CIMS/
and viability of HEKs within CIMS/HEMS. On day 7, the HEMS, demonstrating stimulated cell infiltration. These results
majority of cells cultivated in CIMS and HEMS expressed green suggest that CIMS/HEMS does not trigger adverse host
fluorescence (live cells), and dead cells were rarely detected immune inflammatory response in vivo.
(Figure 7C,D). These results suggest that CIMS and HEMS The rat’s body mass of different groups was regularly
provided the necessary environment for adequate cell function monitored, and after 28 days of implantation, there was no
and did not induce significant cytotoxicity. significant difference in the weight of animals before and after
3.8. CAM Assay. The potential of CIMS/HEMS to induce treatment (CIMS-/HEMS-implanted group). Histological
angiogenesis was examined by implanting CIMS/HEMS on the analysis of the organs retrieved after 28 days of post-
CAM. After 3 days of implantation, HEMS was completely implantation showed no necrosis or significant pathological
enveloped by the CAM and facilitated superior integration with changes in the anatomical structure of the liver, heart, lung, and
the host, whereas this feature was not visualized in CIMS. Also, kidney of the treated group (Supporting Information, Figure
a number of allantoic vessels converged approaching toward the S2). Further, the cardiac muscle of both the CIMS and HEMS
HEMS, while the CIMS group showed the random progression groups showed no indication of fibrosis or inflammatory
of the allantoic vessels (Figure 8A,B). The quantitative analysis response. Also, while observing the liver, the distribution of
confirmed that the numbers of allantoic vessels converging hepatocytes in the treatment group was found to be similar to
toward the HEMS group (28 ± 3) were significantly (p < the typical anatomy. Likewise, no pulmonary fibrosis was
0.001) higher than the CIMS group (12 ± 2). Also, H&E, MT, detected in the lungs of the treatment groups. Besides, the
and DAPI-stained sections (Figure 8C) revealed that HEMS glomerulus structure in the kidney section was observed
promoted cell infiltration with homogeneous distribution, while without difficulty in both the groups. These results indicate that
CIMS supported moderate cell infiltration. CIMS and HEMS did not cause any organ toxicity.
3.9. In Vivo CIMS/HEMS Cellular Response and Organ 3.10. Full-Thickness Cutaneous Wound Healing
Toxicity Analysis. The CIMS/HEMS subcutaneously im- Study. The efficacy of CIMS/HEMS on accelerating the skin
planted groups appeared healthy, and no mortality was wound healing process was evaluated by creating a full-
recorded throughout the study period. After 28 days of thickness skin wound in a rat model. Full-thickness cutaneous
implantation, CIMS/HEMS was retrieved with the surrounding wounds treated with CIMS, HEMS, and SHAM groups were
tissues and histologically stained as shown in Figure 9. H&E photographed at different time durations (0, 7, 14, and 21 days)
staining revealed tissue integration/in-growth between the host to visualize the reduction of wound size and to understand the
tissue and the implanted CIMS/HEMS. Interestingly, CD31 post-operative healing process (Figure 10A). On day 0, CIMS
staining revealed considerably more number of blood vessels and HEMS were firmly attached to the wound bed and readily
(neo-capillary formation) in the HEMS-implanted group absorbed the wound exudates, thus leading to the biological
compared to that in the CIMS-implanted group. From TB fixation of dressing, and also prevented the exposure of wounds
staining, few mast cells were visualized in the host region to the external environment. Also, there was no indication of
surrounding the CIMS/HEMS, but no mast cells were found infection or inflammation in both the CIMS- and HEMS-
infiltrated into CIMS/HEMS. This minor inflammatory treated group throughout the study period. On day 7 post-
response after CIMS/HEMS implantation is attributed to the operation, wound sizes were reduced in all groups (Figure
early inflammatory response, which is similar to the regular 10B), but notably, the wound covered with HEMS-treated
16986 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 11. Histological micrographs of wound sections implanted with CIMS and HEMS at days 7, 14, and 21 after dermal excision by H&E staining
(black arrow indicates the initial wound boundary created; scale bar represents 250 μm; inset scale bar represents 50 μm).

Figure 12. Representative images of (A) MT and (B) anti-CD31 of wounds treated with SHAM, CIMS, and HEMS (scale bar for MT represents 50
μm; scale bar for anti-CD31 represents 100 μm; red represents nucleus staining DAPI, and green depicts antibody expression).

group (77 ± 1.8%) showed significantly (p < 0.05) higher HEMS group compared to that in the CIMS and SHAM
wound contraction compared to the CIMS (65 ± 3.4%) and groups.
SHAM (47 ± 2.09%) groups. Subsequently, after 14 days of MT staining of different groups was performed to
treatment, there was 83 ± 3.3% reduction (P < 0.05) in the demonstrate the collagen (major ECM component) deposition
wound area covered by HEMS, whereas wound size reductions in the wounds (Figure 12A). After 7 days post-treatment, low
were 71 ± 4.3 and 61 ± 5.3% for the wounds covered by the level of regenerative collagen deposition was observed in the
CIMS and SHAM groups, respectively. Interestingly, 21 days SHAM group, whereas comparatively more collagen fibers with
post-treatment, there were 81 ± 4.9 and 90 ± 5.7% reductions granulation tissue formation were visualized in the CIMS- and
in wound area covered by the SHAM and CIMS groups, HEMS-treated groups. Interestingly, on day 14, dermal collagen
respectively, while complete closure of wounds was observed in fibers of the SHAM group were disorganized and sparse,
the HEMS (99 ± 1%) group. whereas they were bundled and arranged in a parallel fashion
After 7 days post-treatment, H&E staining (Figure 11) for the CIMS and HEMS groups. On day 21, more pronounced
revealed eschar formation in all the groups. Considerably, a basket weave type arrangement revealing higher maturation
high number of fibroblast infiltration and pronounced neo- level of collagen was demonstrated particularly for the HEMS-
vascularization was witnessed in the wounds treated with treated group, while it was not distinctly visualized for CIMS
HEMS. On day 14 in wounds treated with HEMS, partial and SHAM.
development of the epidermis was observed, while CIMS and Immunofluorescence staining for CD31 for the CIMS/
SHAM groups exhibited poorly developed epidermis. Remark- HEMS group was further performed at 7 and 14 days to
ably, a small number of epidermal appendages such as demonstrate neovascularization (Figure 12B). After 7 days
sebaceous glands and hair follicles were also observed in the post-treatment, a large number of neo-microvessels were seen
regenerated skin treated with HEMS, while, in contrast, it was in the HEMS-treated group compared to that in the CIMS and
absent in the CIMS and SHAM groups. Further, 21 days post- SHAM groups. This increased vascularization in the HEMS
treatment, the SHAM group displayed incomplete healing group may provide more blood supply to the wound area and
associated with partially developed epithelium, whereas CIMS thus may accelerate the wound healing cascade. At day 14, neo-
displayed discontinuous epithelium. Interestingly, in the microvessels were still developing in the SHAM group,
wounds treated with HEMS, well-developed epidermis demonstrating the slow healing progression therein, while in
associated with differentiated epidermal cells, closely arranged contrast, the number of neo-microvessels reduced in CIMS and
basal cells, and layers of keratinocytes were evident, whereas HEMS revealing superior healing. Further, immunofluores-
these features were not prominent in CIMS and SHAM. cence staining for CK-10 was performed at 14 and 21 days
Moreover, the increased number of secondary structures such post-wounding to investigate the re-epithelialization and
as sebaceous glands and hair follicles was witnessed in the epidermal differentiation (Figure 13). After 14 days, no
16987 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Figure 13. Representative immunohistochemistry images of CK-10-stained histological sections on days 14 and 21 of SHAM, CIMS, and HEMS
(scale bar represents 50 μm; red represents nucleus staining DAPI, and green depicts antibody expression).

expression of CK-10 was observed in the SHAM group because cellular functions such as adhesion, migration, proliferation, and
of the lack of new epithelium formation. Similarly, the CIMS- differentiation. Also, it provides the structural support and
treated group also showed no significant expression of CK-10 at biochemical cues that are required for tissue development as
the suprabasal epithelium layer because the epithelialization and well as rejuvenation.35 It is of prime importance to retain these
its stratification were not distinct. Interestingly, HEMS biological entities in pECM during the decellularization and
supported the development of a new stratified thick epithelial solubilization process. The collagen and GAGs which are the
layer, which resulted in the enhanced expression of CK-10. On primary ECM components were well-preserved in pECM
day 21, the HEMS-treated group underwent further remodeling (Supporting Information Figure S1). Further, cytokine growth
and exhibited thinner epithelium revealing complete re- factor array (Figure 1) and western blot analysis (Figure 2B,C)
epithelialization, hence mimicking native epidermal tissue of pECM established the presence of cytokines/growth factors
architecture, whereas comparatively thicker epithelium was such as EGF, IGF-1, PDGF-B, HGF, VEGFA, TGF-β1, IL-6,
observed in the CIMS and SHAM groups. IL-8, MCP-1, and RANTES at high levels. Thus, the HEMS
3.11. Reverse Transcriptase-PCR (RT-PCR) Analyses of containing pECM with the relevant cytokines/growth factors
the Wound Area. In addition to histological and immuno- required for wound healing will be advantageous for accelerated
fluorescence staining to evaluate overall wound healing, RT- wound healing when compared to CIMS.36
PCR was further utilized to assess the expression profiles of Porosity and mechanical attributes of the scaffold are
genes associated with skin wound healing (COL I, COL III, essential criteria for skin repair and regeneration because it
KRT 10, KRT 14). As shown in Figure 10C,D, COL I provides a personalized environment for the cells to initiate the
expression is upregulated for the HEMS-treated group neotissue formation.37 Less porosity of the scaffold can lead to
compared to that for the CIMS and SHAM groups. This insufficient swelling affecting nutrient/gas transport, while
increased expression of COL I (predominant type of collagen ultrahigh-porosity of the scaffold can result in excess swelling
present in skin) may be attributed to the enhanced remodeling and structural deformation. The SEM (Figure 3A,C) and
in the HEMS group. However, on the other hand, the micro-CT (Figure 3B,D) images of CIMS and HEMS revealed
expression of COL III, KRT 10, and KRT 14 was down- interconnected fibrous morphology with an adequate degree of
regulated in the HEMS-treated group when compared to that porosity and swelling (Figure 4A), which will be beneficial for
in CIMS and SHAM (Figure 10C,D), which confirms that the cell adhesion, proliferation, and migration into the 3D
wound healing process is more complete in the HEMS-treated architecture of the scaffold. The tensile strength values (Figure
group compared to that in the CIMS and SHAM groups. 4D) of CIMS and HEMS were in the range of the values
reported for the naturally derived porous scaffold (heart
4. DISCUSSION muscles of humans/rats0.003−0.07 MPa; collagen gels of
To stimulate accelerated regeneration of full-thickness skin calfskin0.001−0.009 MPa) proposed for soft tissue engineer-
injuries, porous, non-immunogenic, bioactive matrix are ing applications.38,39 Further, HFFs and HEKs were cultivated
scaffolds of choice into which HFFs and HEKs can infiltrate, in the conditioning medium extracted from CIMS/HEMS to
proliferate, and form reparative tissues. In the present study, we study the effect of leachates from the scaffolds. Rhodamine
demonstrated a simple and efficient technique to fabricate phalloidin and DAPI staining (Figure 5A) of HFFs/HEKs
HEMS by blending pECM containing cytokines/growth factors grown in CIMS-/HEMS-conditioned media did not show any
with SF, thus bringing together the inherent advantages of both morphological aberrations, but interestingly HFFs/HEKs
SF and pECM for skin tissue engineering. Because collagen I is grown in HEMS-conditioned media demonstrated more well-
widely reported for its efficacy in skin tissue engineering,33,34 SF spread actin filaments compared to that in CIMS-conditioned
was also blended with collagen I to fabricate CIMS and thus media. The well-spread actin filaments are beneficial for
used for comparison studies with HEMS. pECM was establishing enhanced cell−cell contact and interaction with
successfully prepared by decellularizing human placenta using the environment.40 Also, the tunnel assay (Figure 5B) revealed
the protocol described in our previous study24 and solubilized that the CIMS-/HEMS-conditioned media did not cause any
using urea.25 ECM, a significant noncellular component of the apoptosis-related deleterious effect to the HEKs. The results of
cell, is considered as a direct modulator in regulating many the scratch assay (Figure 6) showed that the HEMS-
16988 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

conditioned medium significantly accelerated the migration of dynamic cell−cell or cell−matrix interactions and modulate
HFFs/HEKs in vitro compared to other groups. The presence various cellular responses.50 Also, different growth factors [IGF-
of cytokines/growth factors such as TGF-β1, IL-6, IL-8, MCP- 1, PDGF-B, TGF-β1, EGF, and VEGFA] stimulate angio-
1, and RANTES in the HEMS-conditioned medium was genesis, collagen deposition, and re-epithelialization by
detected by ELISA (Figure 7A). TGF-β1 and IL-6 are reported recruiting fibroblasts, keratinocytes, endothelial cells, stem
to stimulate increased cell migration of dermal fibroblasts and cells, and macrophages into the wound site.51 Histological
keratinocytes.41−45 Likewise, IL-8 is also reported to promote (Figure 11) and immunofluorescence (Figures 12B and 13)
skin re-epithelialization by increasing keratinocyte migration.45 investigation of the wounds covered with HEMS demonstrated
Also, MCP-1 and RANTES are known to promote dermal rapid healing with pronounced neovascularization and re-
wound healing by acting as a chemoattractant for the cells.46 epithelialization compared to the CIMS and SHAM groups.
Thus, it may be concluded that the presence of cytokines/ Further, MT staining (Figure 12A) of the wounds treated with
growth factors (TGF-β1, IL-6, IL-8, MCP-1, and RANTES) in HEMS revealed the higher turnover rate of bundled and
the HEMS-conditioned medium is responsible for the arranged collagen fibers, which lead to early wound stabilization
accelerated migration of HFFs/HEKs in vitro. Thus, in the suggesting its role in supporting skin remodeling. Predom-
course of wound healing, augmented fibroblast infiltration into inantly during the proliferation and remodeling phases, HGF is
the wound site during the initial days after injury has been a vital growth factor in scar formation by controlling the
reported to stimulate the wound healing process, and equilibrium between synthesis and degradation of ECM via
accelerated migration of basal keratinocytes from the wound stimulating the secretion of matrix metalloproteinase in
edges to the lesion leads to closure of the wound area, which in fibroblasts.52 Also, bFGF inhibits the phenotypic conversion
turn triggers the differentiation of basal keratinocyte to of fibroblasts to myofibroblasts, thus preventing scarring.53
keratinizing epidermal cells because of contact inhibition.47 Further, the complete wound closure without scar formation
The MTT assay (Figure 7B) demonstrated that HAMSCs/ was witnessed only in the HEMS group. Thus, our results
HEKs/HFFs cultivated directly in HEMS had a higher rate of indicate that the release of endogenous growth factors (IGF-1,
proliferation compared to that in CIMS and TCP. Also, live− PDGF-B, TGF-β1, EGF, VEGFA, HGF, and bFGF) from
dead staining (Figure 7C,D) revealed that HEKs successfully HEMS has contributed to superior neovascularization and
adhered, infiltrated, and remained viable in the HEMS accelerated re-epithelialization without scar formation. Also,
microenvironment. Thus, taken together, these findings expression profiles of genes (COL I, COL III, KRT 10, KRT
strongly suggest that HEMS containing cytokines/growth 14) associated with wound healing were studied in wound area
factors with desirable physical attributes supported the retrieved from different treatment groups after 21 days by RT-
adhesion, infiltration, migration, and proliferation of PCR (Figure 10C,D). The results demonstrated that SHAM
HAMSCs/HEKs/HFFs and therefore proved to be nontoxic expressed higher expression of KRT 10 and KRT 14 compared
and functionally competent for skin tissue engineering in vitro. to other groups because of the presence of more proliferating
Vascularization is considered to be a critical aspect for the or differentiating keratinocytes, which is indicative of
survival of engineered scaffolds; insufficient vasculature can lead incomplete healing and infers that the re-epithelialization
to necrosis or volume reduction after implantation and inhibits process is still ongoing.54 On the other hand, the HEMS
efficient regeneration of the skin defect.48 Herein, ex vivo CAM expressed lower expression of KRT 10 and KRT 14 as the
assay (Figure 8) using scaffolds revealed significantly more epithelialization process is more complete. Also, downregulated
number of allantoic vessels approaching toward the HEMS expression of COL III and upregulation of COL I in HEMS-
compared to the CIMS group. This enhanced angiogenic treated wounds relative to CIMS/SHAM are suggestive of early
response of the HEMS may be attributed to the preservation of remodeling because COL III is produced during early phases of
VEGFA (the most potent growth factor reported to promote wound healing and subsequently replaced by COL I in the final
the formation of new blood vessels49) in the HEMS. Further, it stages of wound healing.55 Therefore, HEMS is evidenced to be
is of crucial importance to assess the host immune response in a promising matrix that delivered the required cytokines/
vivo by subcutaneous grafting before investigating the healing growth factors and desirable microenvironment to augment
ability of CIMS/HEMS in full-thickness skin wounds. cutaneous wound healing.
Histological evaluation (Figure 9) of the subcutaneously
implanted CIMS/HEMS after 28 days displayed the absence
5. CONCLUSIONS
of tissue necrosis, granulomatous inflammation, or fibrous layer
development surrounding the CIMS/HEMS. Furthermore, the We successfully fabricated HEMS by a combinatorial approach
implanted HEMS exhibited its outstanding biocompatibility in for skin tissue engineering applications. The fabricated HEMS
vivo by supporting considerably more number of blood vessel retained the natural ECM composition and the cytokines/
formation compared to CIMS. Also, from H&E staining of growth factors which are known to accelerate the migration of
organs (Supporting Information, Figure S2) retrieved from the HFFs/HEKs, blood vessel induction/development, and re-
animals treated with CIMS/HEMS, no organ toxicity was epithelialization. Also, the 3D architecture of HEMS with
visualized in the major organs (heart, liver, lung, and kidney). adequate porosity and mechanical stability supported excellent
Overall, the absence of any adverse immune reaction in the viability and proliferation of HFFs/HEKs/HAMSCs in vitro.
host animals suggests that these engineered scaffolds are Further, HEMS demonstrated its superior vascularization
nonimmunogenic in vivo. potential by the CAM assay, and also no immune rejection
The efficacy of CIMS/HEMS in accelerating wound healing or organ toxicity was observed in the host after subcutaneous
was evaluated in the full-thickness cutaneous wound in a rat implantation. We also evidenced the ability of HEMS to
model. ECM components such as collagen, laminin, mediate early skin repair in full-thickness wound models.
fibronectin, elastin, and GAG are essential at every phase Therefore, this study provides an approach wherein human
(hemostasis to remodeling) of the wound healing cascade via pECM is incorporated with SF to produce a bioactive hybrid
16989 DOI: 10.1021/acsami.7b19007
ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

scaffold that has enormous potential for skin tissue engineering (10) Zhang, W.; Chen, L.; Chen, J.; Wang, L.; Gui, X.; Ran, J.; Xu,
applications. G.; Zhao, H.; Zeng, M.; Ji, J.; Qian, L.; Zhou, J.; Ouyang, H.; Zou, X.


Silk Fibroin Biomaterial Shows Safe and Effective Wound Healing in
ASSOCIATED CONTENT Animal Models and a Randomized Controlled Clinical Trial. Adv.
Healthcare Mater. 2017, 6, 1700121.
*
S Supporting Information
(11) Guan, G.; Bai, L.; Zuo, B.; Li, M.; Wu, Z.; Li, Y.; Wang, L.
The Supporting Information is available free of charge on the Promoted Dermis Healing From Full-Thickness Skin Defect by
ACS Publications website at DOI: 10.1021/acsami.7b19007. Porous Silk Fibroin Scaffolds (PSFSs). Biomed. Mater. Eng. 2010, 20,
Collagen and GAG quantification; H&E staining of 295−308.
heart, liver, lung, and kidney harvested from different (12) Gil, E. S.; Panilaitis, B.; Bellas, E.; Kaplan, D. L. Functionalized
treatment groups; MT staining of hybrid scaffolds; and Silk Biomaterials for Wound Healing. Adv. Healthcare Mater. 2013, 2,
gene-specific primers used for RT-PCR (PDF) 206−217.


(13) Yan, S.; Zhang, Q.; Wang, J.; Liu, Y.; Lu, S.; Li, M.; Kaplan, D. L.
Silk Fibroin/Chondroitin Sulfate/Hyaluronic Acid Ternary Scaffolds
AUTHOR INFORMATION for Dermal Tissue Reconstruction. Acta Biomater. 2013, 9, 6771−
Corresponding Author 6782.
*E-mail: sdhara@smst.iitkgp.ernet.in. (14) Xiong, S.; Zhang, X.; Lu, P.; Wu, Y.; Wang, Q.; Sun, H.; Heng,
B. C.; Bunpetch, V.; Zhang, S.; Ouyang, H. A Gelatin-Sulfonated Silk
ORCID
Composite Scaffold Based on 3D Printing Technology Enhances Skin
Koel Chaudhury: 0000-0002-9390-1179 Regeneration by Stimulating Epidermal Growth and Dermal Neo-
Santanu Dhara: 0000-0003-4443-7610 vascularisation. Sci. Rep. 2017, 7, 4288.
Notes (15) Fuentes, M. G.; Meinel, A. J.; Hilbe, M.; Meinel, L.; Merkle, H.
The authors declare no competing financial interest. P. Silk Fibroin/Hyaluronan Scaffolds for Human Mesenchymal Stem


Cell Culture in Tissue Engineering. Biomaterials 2009, 30, 5068−5076.
ACKNOWLEDGMENTS (16) Lv, Q.; Hu, K.; Feng, Q.; Cui, F. Fibroin/Collagen Hybrid
Hydrogels with Crosslinking Method: Preparation, Properties, and
The authors A.P.R. and S.D. acknowledge the fellowship Cytocompatibility. J. Biomed. Mater. Res., Part A 2008, 84A, 198−207.
support received from the Ministry of Human Resource (17) Bhardwaj, N.; Kundu, S. C. Silk Fibroin Protein and Chitosan
Development (MHRD, Govt. of India), K.B. from the Polyelectrolyte Complex Porous Scaffolds for Tissue Engineering
Department of Science & Technology (DST, Govt. of India) Applications. Carbohydr. Polym. 2011, 85, 325−333.
INSPIRE, and P.P.M. from the Indian Council of Medical (18) Vasconcelos, A.; Gomes, A. C.; Paulo, A. C. Novel Silk Fibroin/
Research (ICMR, Govt. of India). The funding of the project Elastin Wound Dressings. Acta Biomater. 2012, 8, 3049−3060.
(BT/PR7818/MED/32/279/2013) from the Department of (19) Bhardwaj, N.; Sow, W. T.; Devi, D.; Ng, K. W.; Mandal, B. B.;
Biotechnology (DBT, Govt. of India) is greatly acknowledged. Cho, N.-J. Silk Fibroin−Keratin Based 3D Scaffolds as a Dermal


Substitute for Skin Tissue Engineering. Integr. Biol. 2015, 7, 53−63.
REFERENCES (20) Aamodt, J. M.; Grainger, D. W. Extracellular Matrix-Based
Biomaterial Scaffolds and the Host Response. Biomaterials 2016, 86,
(1) Yildirimer, L.; Thanh, N. T. K.; Seifalian, A. M. Skin 68−82.
Regeneration Scaffolds: A Multimodal Bottom-up approach. Trends (21) Wildman, D. E. Review: Toward an Integrated Evolutionary
Biotechnol. 2012, 30, 638−648. Understanding of the Mammalian Placenta. Placenta 2011, 32, S142−
(2) Shevchenko, R. V.; James, S. L.; James, S. E. A Review of Tissue- S145.
Engineered Skin Bioconstructs Available for Skin Reconstruction. J. R. (22) Hong, J. W.; Lee, W. J.; Hahn, S. B.; Kim, B. J.; Lew, D. H. The
Soc., Interface 2010, 7, 229−258. Effect of Human Placenta Extract in a Wound Healing Model. Ann.
(3) Chen, X.; Zhang, M.; Wang, X.; Chen, Y.; Yan, Y.; Zhang, L.; Plast. Surg. 2010, 65, 96−100.
Zhang, L. Peptide-Modified Chitosan Hydrogels Promote Skin (23) De, D.; Chakraborty, P. D.; Bhattacharyya, D. Regulation of
Wound Healing by Enhancing Wound Angiogenesis and Inhibiting
Trypsin Activity by Peptide Fraction of an Aqueous Extract of Human
Inflammation. Am. J. Transl. Res. 2017, 9, 2352−2362.
Placenta Used as Wound Healer. J. Cell. Physiol. 2011, 226, 2033−
(4) Choi, J. S.; Kim, J. D.; Yoon, H. S.; Cho, Y. W. Full-Thickness
2040.
Skin Wound Healing Using Human Placenta-Derived Extracellular
(24) Rameshbabu, A. P.; Ghosh, P.; Subramani, E.; Bankoti, K.;
Matrix Containing Bioactive Molecules. Tissue Eng., Part A 2013, 19,
329−339. Kapat, K.; Datta, S.; Maity, P. P.; Subramanian, B.; Roy, S.; Chaudhury,
(5) Simman, R.; Phavixay, L. Split-Thickness Skin Grafts Remain the K.; Dhara, S. Investigating the Potential of Human Placenta-Derived
Gold Standard for the Closure of Large Acute and Chronic Wounds. J. Extracellular Matrix Sponges Coupled with Amniotic Membrane-
Am. Col. Certif. Wound Spec. 2011, 3, 55−59. Derived Stem Cells for Osteochondral Tissue Engineering. J. Mater.
(6) Lotz, C.; Schmid, F. F.; Oechsle, E.; Monaghan, M. G.; Walles, Chem. B 2016, 4, 613−625.
H.; Becker, F. G. Cross-Linked Collagen Hydrogel Matrix Resisting (25) Moore, M. C.; Pandolfi, V.; Mcfetridge, P. S. Novel Human-
Contraction to Facilitate Full-Thickness Skin Equivalents. ACS Appl. Derived Extracellular Matrix Induces In Vitro and In Vivo
Mater. Interfaces 2017, 9, 20417−20425. Vascularization and Inhibits Fibrosis. Biomaterials 2015, 49, 37−46.
(7) Li, Q.; Niu, Y.; Diao, H.; Wang, L.; Chen, X.; Wang, Y.; Dong, L.; (26) Frazier, S. B.; Roodhouse, K. A.; Hourcade, D. E.; Zhang, L. The
Wang, C. In Situ Sequestration of Endogenous PDGF-BB with an Quantification of Glycosaminoglycans: A Comparison of HPLC,
ECM-Mimetic Sponge for Accelerated Wound Healing. Biomaterials Carbazole, and Alcian Blue Methods. Open Glycosci. 2008, 1, 31−39.
2017, 148, 54−68. (27) Rockwood, D. N.; Preda, R. C.; Yücel, T.; Wang, X.; Lovett, M.
(8) Selvaraj, S.; Fathima, N. N. Fenugreek Incorporated Silk Fibroin L.; Kaplan, D. L. Materials Fabrication from Bombyx Mori Silk
NanofibersA Potential Antioxidant Scaffold for Enhanced Wound Fibroin. Nat. Protoc. 2011, 6, 1612−1631.
Healing. ACS Appl. Mater. Interfaces 2017, 9, 5916−5926. (28) Pati, F.; Adhikari, B.; Dhara, S. Isolation and Characterization of
(9) Devalliere, J.; Dooley, K.; Hu, Y.; Kelangi, S. S.; Uygun, B. E.; Fish Scale Collagen of Higher Thermal Stability. Bioresour. Technol.
Yarmush, M. L. Co-Delivery of a Growth Factor and a Tissue- 2010, 101, 3737−3742.
Protective Molecule Using Elastin Biopolymers Accelerates Wound (29) Ghosh, P.; Rameshbabu, A. P.; Dogra, N.; Dhara, S. 2,5-
Healing in Diabetic Mice. Biomaterials 2017, 141, 149−160. Dimethoxy 2,5-Dihydrofuran Crosslinked Chitosan Fibers Enhance

16990 DOI: 10.1021/acsami.7b19007


ACS Appl. Mater. Interfaces 2018, 10, 16977−16991
ACS Applied Materials & Interfaces Research Article

Bone Regeneration in Rabbit Femur Defects. RSC Adv. 2014, 4, (53) Desmouliere, A.; Darby, I. A.; Laverdet, B.; Bonté, F. Fibroblasts
19516−19524. and Myofibroblasts in Wound Healing. Clin., Cosmet. Invest. Dermatol.
(30) Aasen, T.; Izpisúa Belmonte, J. C. Isolation and Cultivation of 2014, 7, 301−311.
Human Keratinocytes from Skin or Plucked Hair for the Generation of (54) Levengood, S. L.; Erickson, A. E.; Chang, F.-c.; Zhang, M.
Induced Pluripotent Stem Cells. Nat. Protoc. 2010, 5, 371−382. Chitosan−Poly(Caprolactone) Nanofibers For Skin Repair. J. Mater.
(31) Liang, C.-C.; Park, A. Y.; Guan, J.-L. In Vitro Scratch Assay: A Chem. B 2017, 5, 1822−1833.
Convenient and Inexpensive Method for Analysis of Cell Migration in (55) Volk, S. W.; Wang, Y.; Mauldin, E. A.; Liechty, K. W.; Adams, S.
vitro. Nat. Protoc. 2007, 2, 329−333. L. Diminished Type III Collagen Promotes Myofibroblast Differ-
(32) Ribatti, D.; Nico, B.; Vacca, A.; Presta, M. The Gelatin Sponge- entiation and Increases Scar Deposition in Cutaneous Wound Healing.
Chorioallantoic Membrane Assay. Nat. Protoc. 2006, 1, 85−91. Cells Tissues Organs 2011, 194, 25−37.
(33) Brett, D. A Review of Collagen and Collagen-Based Wound
Dressings. Wounds 2008, 20, 347−356.
(34) Chattopadhyay, S.; Raines, R. T. Collagen-Based Biomaterials
for Wound Healing. Biopolymers 2014, 101, 821−833.
(35) Frantz, C.; Stewart, K. M.; Weaver, V. M. The Extracellular
Matrix at a Glance. J. Cell Sci. 2010, 123, 4195−4200.
(36) Kim, E. J.; Choi, J. S.; Kim, J. S.; Choi, Y. C.; Cho, Y. W.
Injectable and Thermosensitive Soluble Extracellular Matrix and
Methylcellulose Hydrogels for Stem Cell Delivery in Skin Wounds.
Biomacromolecules 2016, 17, 4−11.
(37) O’brien, F. J. Biomaterials & Scaffolds for Tissue Engineering.
Mater. Today 2011, 14, 88−95.
(38) Chen, Q.-Z.; Harding, S. E.; Ali, N. N.; Lyon, A. R.; Boccaccini,
A. R. Biomaterials in Cardiac Tissue Engineering: Ten Years of
Research Survey. Mater. Sci. Eng. 2008, 59, 1−37.
(39) Ö zyazgan, I.; Liman, N.; Dursun, N.; Güneş, I. The Effects of
Ovariectomy on the Mechanical Properties of Skin in Rats. Maturitas
2002, 43, 65−74.
(40) Revenu, C.; Athman, R.; Robine, S.; Louvard, D. The Co-
Workers of Actin Filaments: From Cell Structures to Signals. Nat. Rev.
Mol. Cell Biol. 2004, 5, 635−646.
(41) Postlethwaite, A. E.; Keski-Oja, J.; Moses, H. L.; Kang, A. H.
Stimulation of the Chemotactic Migration of Human Fibroblasts by
Transforming Growth Factor Beta. J. Exp. Med. 1987, 165, 251−256.
(42) Li, Y.; Fan, J.; Chen, M.; Li, W.; Woodley, D. T. Transforming
Growth Factor-Alpha: A Major Human Serum Factor that Promotes
Human Keratinocyte Migration. J. Invest. Dermatol. 2006, 126, 2096−
2105.
(43) Luckett, L. R.; Gallucci, R. M. Interleukin-6 (IL-6) Modulates
Migration and Matrix Metalloproteinase Function in Dermal
Fibroblasts from IL-6KO Mice. Br. J. Dermatol. 2007, 156, 1163−1171.
(44) Gallucci, R. M.; Sloan, D. K.; Heck, J. M.; Murray, A. R.; O’dell,
S. J. Interleukin 6 Indirectly Induces Keratinocyte Migration. J. Invest.
Dermatol. 2004, 122, 764−772.
(45) Michel, G.; Kemény, L.; Peter, R. U.; Beetz, A.; Ried, C.;
Arenberger, P.; Ruzicka, T. Interleukin-8 Receptor-Mediated Chemo-
taxis of Normal Human Epidermal Cells. FEBS Lett. 1992, 305, 241−
243.
(46) Dipietro, L. A.; Reintjes, M. G.; Low, Q. E. H.; Levi, B.; Gamelli,
R. L. Modulation of Macrophage Recruitment into Wounds by
Monocyte Chemoattractant Protein-1. Wound Repair Regen. 2001, 9,
28−33.
(47) Werner, S.; Grose, R. Regulation Healing by Growth of Wound
Factors and Cytokines. Physiol. Rev. 2003, 83, 835−870.
(48) Rouwkema, J.; Khademhosseini, A. Vascularization and
Angiogenesis in Tissue Engineering: Beyond Creating Static Net-
works. Trends Biotechnol. 2016, 34, 733−745.
(49) Yancopoulos, G. D.; Davis, S.; Gale, N. W.; Rudge, J. S.;
Wiegand, S. J.; Holash, J. Vascular-Specific Growth Factors and Blood
Vessel Formation. Nature 2000, 407, 242−248.
(50) Schultz, G. S.; Wysocki, A. Interactions between Extracellular
Matrix and Growth Factors in Wound Healing. Wound Repair Regen.
2009, 17, 153−162.
(51) Barrientos, S.; Brem, H.; Stojadinovic, O.; Tomic-Canic, M.
Clinical Application of Growth Factors and Cytokines in Wound
Healing. Wound Repair Regen. 2014, 22, 569−578.
(52) Jackson, W. M.; Nesti, L. J.; Tuan, R. S. Mesenchymal Stem Cell
Therapy for Attenuation of Scar Formation during Wound Healing.
Stem Cell Res. Ther. 2012, 3, 20.

16991 DOI: 10.1021/acsami.7b19007


ACS Appl. Mater. Interfaces 2018, 10, 16977−16991

You might also like