ArticlePDF Available

Human decellularized adipose matrix derived hydrogel assists mesenchymal stem cells delivery and accelerates chronic wound healing

Authors:

Abstract and Figures

Biological scaffolds based stem cell delivery methods have emerged as a promising approach for tissue repair and regeneration. Here we developed a hydrogel biological scaffold from human decellularized adipose matrix (hDAM) for human adipose‐derived stem cells (hASCs) delivery to accelerate chronic wound healing. The hDAM hydrogel was prepared by pepsin mediated digestion and pH controlled neutralization. The morphology, survival, proliferation, and angiogenic paracrine activity of hASCs cultured in the hydrogel were assessed. Moreover, the therapeutic efficacy of the hASCs‐hydrogel composite for impaired wound healing was evaluated by using a full‐thickness wound model on diabetic mouse. The developed hDAM hydrogel was a thermosensitive hydrogel, presented the biochemical complexity of native extracellular matrix and formed a porous nanofiber structure after gelation. The hydrogel can support hASCs adhesion, survival, and proliferation. Compared to standard culture condition, hASCs cultured in the hydrogel exhibited enhanced paracrine activity with increased secretion of hepatocyte growth factor. In the diabetic mice model with excisional full‐thickness skin wounds, mice treated with the hASCs‐hydrogel composite displayed accelerated wound closure and increased neovascularization. Our results suggested that the developed hDAM hydrogel can provide a favorable microenvironment for hASCs with augmented regeneration potential to accelerate chronic wound healing.
Content may be subject to copyright.
Human decellularized adipose matrix derived hydrogel assists mesenchymal stem cells
delivery and accelerates chronic wound healing
Zhaoyang Chen1, 2
Bowen Zhang4, 5
Jun Shu2
Haiyang Wang3, 4
Yudi Han2
Quan Zeng3,
4
Youbai Chen2
Jiafei Xi3, 4
Ran Tao2
Xuetao Pei3, 4
Wen Yue3, 4,*
Yan Han2,*
1Medical School of Chinese PLA, Beijing, China
2Department of Plastic and Reconstructive Surgery, the First Medical Centre,
Chinese PLA General Hospital, Beijing, China
3Stem Cell and Regenerative Medicine Lab, Institute of Health Service and
Transfusion Medicine, AMMS, Beijing, China
4South China Research Center for Stem Cell & Regenerative Medicine, SCIB,
Guangzhou, China
5Beijing Institute of Radiation Medicine, AMMS, Beijing, China
Correspondence
Wen Yue, Stem Cell and Regenerative Medicine Lab, Institute of Health Service and
Transfusion Medicine, AMMS, No. 27 Taiping Road, Haidian District, Beijing 100850, China.
Email: yuewen0206@126.com
Yan Han, Department of Plastic and Reconstructive Surgery, the First Medical Centre, Chinese
PLA General Hospital, No. 28 Fuxing Road, Haidian District,
Beijing 100853, China.
Email: 13720086335@163.com
This article has been accepted for publication and undergone full peer review but has not been
through the copyediting, typesetting, pagination and proofreading process which may lead to
differences between this version and the Version of Record. Please cite this article as doi:
10.1002/jbm.a.37133
This article is protected by copyright. All rights reserved.
Abstract
Biological scaffolds based stem cell delivery methods have emerged as a promising approach for
tissue repair and regeneration. Here we developed a hydrogel biological scaffold from human
decellularized adipose matrix (hDAM) for human adipose-derived stem cells (hASCs) delivery to
accelerate chronic wound healing. The hDAM hydrogel was prepared by pepsin mediated
digestion and pH controlled neutralization. The morphology, survival, proliferation and
angiogenic paracrine activity of hASCs cultured in the hydrogel were assessed. Moreover, the
therapeutic efficacy of the hASCs-hydrogel composite for impaired wound healing was evaluated
by using a full-thickness wound model on diabetic mouse. The developed hDAM hydrogel was a
thermosensitive hydrogel, presented the biochemical complexity of native extracellular matrix
(ECM) and formed a porous nanofiber structure after gelation. The hydrogel can support hASCs
adhesion, survival and proliferation. Compared to standard culture condition, hASCs cultured in
the hydrogel exhibited enhanced paracrine activity with increased secretion of hepatocyte growth
factor (HGF). In the diabetic mice model with excisional full-thickness skin wounds, mice treated
with the hASCs-hydrogel composite displayed accelerated wound closure and increased
neovascularization. Our results suggested that the developed hDAM hydrogel can provide a
favorable microenvironment for hASCs with augmented regeneration potential to accelerate
chronic wound healing.
KEYWORDS
chronic wound healing, hydrogel, human adipose-derived stem cells, extracellular matrix,
mesenchymal stem cells
1. INTRODUCTION
Chronic wounds are skin defects that fail to restore structural and functional integrity in an
orderly and timely manner [1]. This kind of wounds is refractory and recurrent, which lead to
difficulties in clinical treatment and tremendous suffering in patients [2]. Recent studies have
shown that mesenchymal stem cells (MSCs) are effective for chronic wound healing. MSCs,
mostly derived from bone marrow, adipose tissue, umbilical cord, can promote wound closure and
angiogenesis through its regenerative paracrine effects and multi-lineage differentiation potential
[3]. However, the short retention time and low survival rate of transplanted stem cells limited the
therapeutic effects of MSCs, especially when using the traditional cell direct injection approaches
[4]. Thus, researchers have been making efforts to develop more effective cell delivery methods.
Biological scaffolds with cell delivery potentials are showing promising therapeutic effects in
chronic wound treatment [5]. These scaffolds provide a favorable microenvironment for cell
delivery and contribute to cell adhension, proliferation and differentiation [6]. With regard to
MSCs, previous studies have used sheet-formed biological scaffolds such as collagen sponge and
acellular dermal matrix to deliver MSCs [7, 8]. However, this kind of scaffold asked for additional
cell inoculation procedure and could not be well conformed to irregular wound geometry. Thus,
hydrogel biological scaffolds with sol-gel transition property are better options, which are
characterized by easy cell encapsulation and filling irregular injury site [9].
Hydrogels are highly hydrated polymer materials composed of synthetic or natural polymer
chains [10]. In contrast to synthetic hydrogels, natural derived hydrogels perform better on
measures of bioactive and biocompatible capacities. In addition, natural-derived hydrogels can
mimic the cell microenvironment in vivo, which make them ideal vehicles for cell delivery [11].
However, the most studied natural-derived hydrogels, like collagen or hyaluronic acid, only
contain individual component of the extracellular matrix (ECM), which compromised their
potentials in providing the complex matrix microenvironment necessary for stem cell function and
tissue regeneration [12]. Developing intact ECM-derived hydrogels from living tissues is
becoming a research focus in recent years.
In this study, we aimed to develop an ECM-derived hydrogel from human decellularized
adipose tissue matrix (hDAM) for delivering human adipose-derive stem cells (hASCs) to chronic
wounds. The hydrogel was prepared by pepsin mediated digestion and pH controlled
neutralization. The hydrogel is characterized in terms of biochemical composition, rheological
property and gel ultrastructure. The morphology, survival, proliferation and angiogenic paracrine
activity of hASCs cultured in the hydrogel were assessed. Moreover, the therapeutic efficacy of
the hASCs-hydrogel composite for impaired wound healing was evaluated using a full-thickness
wound model on diabetic mouse.
2 MATERIALS AND METHODS
2.1 Human adipose tissue collection
Fresh and sterile human subcutaneous adipose tissues were collected from female donors
(aged 18 - 35 years; BMI, <30; hemoglobin, >110 g/l) who had undergone abdominal or thigh
liposuction surgery at the First Medical Central of Chinese PLA General Hospital with informed
consent. Donors with diabetes mellitus, hypertension, serious systemic metabolic diseases or lipid
disorders were excluded. Patients were treated with tumescent anesthesia (tumescent solution:
1,000 ml of 0.9% normal saline + 10 ml of 2% lidocaine + 1 ml of 0.1% adrenaline). Fat was
suctioned using liposuction needles (3 mm in diameter) and under negative pressure via a 20 ml
syringe (used for hASCs isolation) or a liposuction machine (used for preparation of hDAM
hydrogel). After delivered to the lab on ice in sterile and airtight containers, the samples were
thoroughly rinsed with 0.9% sodium chloride solution to remove tumescent fluid, red blood cells
and cell debris. Samples obtained with the liposuction machine if not used for hDAM preparation
immediately would been placed in a -80 refri gerator for short-term storage. The research was
approved by the Ethics Committee of Chinese PLA General Hospital (No. S2017-059-10) and
conducted according to the 2000 Helsinki Declaration.
2.2 Preparation of hDAM hydrogel
hDAM was prepared from human adipose tissue with an enzyme and solvent-based
decellularization method as described previously [13]. Following decellularization, the extracted
hDAM was freezed at -80 overnight and then dried using Gamma 2-16 LSC Freeze Dryer
(Christ, Osterode, German) for 24 hours. Decellularization efficiency of the hDAM was evaluated
by residual dsDNA and triglyceride content assay. For residual dsDNA content assay, total DNA
was extracted from the hDAM with the TIANamp Genomic DNA Kit (TIANGEN, Beijing, China)
and measured with the Quant-iT PicoGreen dsDNA Reagent and Kits (Invitrogen, Carlsbad, CA)
according to the manufacturer’s protocol (n=3) with the cutoff value of 50 ng/mg ECM (dry wt.).
The Cayman Triglyceride Colorimetric Assay kit (Chemical Inc, Ann Arbor, MI) was used to
measure the residual lipid content within the hDAM per the manufacturer’s instructions (n=3).
hDAM hydrogel was prepared by pepsin mediated digestion and pH controlled
neutralization. Briefly, 100 mg hDAM powder was digested with 10 mg 250 U/mg pepsin (Sigma,
St. Louis, MO) in 10 ml 0.01 M HCl at constant shaking for 72 hours at room temperature. The
hDAM pre-gel solution was formed by neutralizing the pH of hDAM digestion to 7.4 using 1 M
NaOH and 10 X alpha Minimum Essential Medium (α-MEM; Gibco, Waltham, MA), with
operating on ice. The hDAM hydrogel was prepared by incubating the pre-gel solution at 37 for
30 minutes. The hDAM pre-gel solution or hydrogel at 8 mg/ml was used for in vitro and in vivo
studies.
2.3 Biochemical composition analysis
The biochemical composition of the hDAM hydrogel was analyzed, including soluble
collagen content, sulfated Glycosaminoglycan (sGAG) content and protein profile.
hDAM digestion was diluted and assayed for soluble collagen content with the Sircol Soluble
Collagen Assay kit (Biocolor Ltd, Carrickfergus, United Kingdom) according to the
manufacturer’s instructions (n=3). Bovine collagen (500 μg/ml) was used as the reference standard
and 0.01 M HCl was used as the as the negative control. Absorbance at 555 nm of the reaction
solution was measured using spectramax M2 microplate reader (Molecular Devices, Sunnyvale,
CA).
hDAM pre-gel solution was assayed for sGAG content with the Blyscan sGAG Assay Kit
(Biocolor Ltd) according to the manufacturer’s instructions (n=3). Bovine tracheal chondroitin
4-sulfate (100 μg/ml) was used as the reference standard and ddH2O was used as the negative
control. Absorbance at 656 nm of the reaction solution was measured using the microplate reader.
The complex protein composition of the hDAM hydrogel was detected with Liquid
Chromatography-Tandem Mass Spectrometry (LC-MS/MS). Representative sample of hDAM
digestion was ultra-centrifuged with Millipore 10 KD centrifugal filter unite (Billerica, MA, USA)
to remove undigested particles. The filtrates were collected, followed by desiccating, desalting and
desiccating. Dried sample was re-dissolved in 1% formic acid, and 1 μg of sample was used for
subsequent analysis. For LS-MS/MS analyzing, the sample was firstly separated by the Easy-nLC
1200 high performance liquid chromatography system (Thermo Scientific, Waltham, MA) at 200
nl/min, and then analyzed on Orbitrap Fusion Lumos mass spectroscopy (Thermo Scientific) at
data dependent acquisition mode (60,000 resolution (FWHM), m/z 375-1600, Higher Energy
Collision Dissociation (HCD) mode using 30% collision energy). Generated MS/MS spectra data
were transferred to peptide sequences by comparing to the Uniprot database using the in-house
Mascot server 2.3.2 (Matrix Science, London, UK). GO analysis was used to characterize the
cellular component, molecular function and biological process of the identified high score (>20)
peptides by searching the DAVID bioinformatics resources.
2.4 Rheological property measurement
The rheological property of the hDAM hydrogel was measured with Anton Paar MCR302
rheometer (Graz, Austrian) operating with a 40 mm parallel plate system. The temperature was
controlled within 0.01 using a Peltier plate. 1 ml of hD A M pre-gel solution placed on ice was
loaded onto the rheometer plate pre-cooled to 0 . A f ter l oadi ng , the steady shear vi scosi ty w as
measured by applying a strain of 301% Pa at a frequency of 10 rad/s. The temperature was then
increased from 0 to 37 ℃ at 2 ℃ per 30 seconds to induce gelation, and a series of oscillatory
strain of 201% (0-29 ℃), 53% (31 ), 14% (33 ), 3% (35 ), and 1% (37 ) at a frequency of 10
rad/s were imposed to track the gelation kinetics.
2.5 Scanning electron microscopy
Scanning electron microscopy (SEM) was used to visualize the ultrastructure of the hDAM
hydrogel. For hydrogel samples preparation, gelation hDAM hydrogel were firstly fixed in cold
electron fixation solution (2.5% glutaraldehyde, Servicebio, Wuhan, China) for 24 hours at 4 ,
followed by secondary fixation in 1% osmic acid fix for 2 hours at room temperature. Fixed
samples were dehydrated in a graded series of alcohol (30, 50, 70, 90, 95, 100, 100% ethanol in
PBS) and isoamyl acetate for 15 minutes per wash. Samples were slowly dried in Quorum K850
critical point dryer (London, UK). After drying, the samples were sputter coated with gold for 30
seconds and imaged with Hitachi SU8100 SEM (Tokyo, Japan). A set of fiber network
characteristics such as fiber alignment, pore size, fiber diameter and node density (number of fiber
intersections per μm2) was measured from the acquired SEM images. The fiber architecture
features were automated extracted and quantified using the angiogenesis analyzer algorithm in
Image-J software (version 1.52a, NIH, USA).
2.6 Isolation and culture of hASCs
Primary hASCs were isolated from lipoaspirates according to the procedures described by Li
et al. [14]. Briefly, thoroughly rinsed adipose tissue was digested with equal volume 0.1%
collagenase I (Sigma) for 1 hour at 200 rpm and 37 . A fter d i g esti on, the stromal vascul ar
fraction (SVF) pellets were collected by centrifuging the samples at 270 x g for 10 minutes at room
temperature. Collected cell pellets were resuspended with MSC serum-free medium (SANLY,
Beijing, China), plated to a 10 cm culture disk and cultured at 37 with 5% CO
2. The culture
medium was firstly changed 24 hours after plating to remove non-adherent cells, and then changed
every 2 days until the cells achieved 80-90% confluence. hASCs were passaged with TrypLE
Express (Gibco) to detach them from culture surface and cells at culture passage 3-6 were used in
the follow studies.
2.7 Multipotent differentiation potential test of hASCs
The multipotent differentiation potential of the cultured hASCs to adipocytes, osteoblasts and
chondrocytes were tested with the hASCs Differentiation Medium Kit (Cyagen, Guangzhou,
China) according to the manufacture’s protocol. hASCs cultured in Dulbecco’s modified Eagle
medium (DMEM; Gibco) supplemented with 10% fetal bovine serum (FBS; Gibco) and 1%
penicillin-streptomycin (Gibco) were used as a control. After 3 weeks of adipogenic induction, the
lipid droplets formation capacity was evaluated by Oil-Red-O staining. After osteogenic
differentiation for 4 weeks, cells were fixed with 4% paraformaldehyde and stained with Alizarin
Red to visualize calcium deposition. Chondrogenic induction was conducted using a micromass
culture system. After 4 weeks of induction, cell aggregates were fixed with 10% formalin, paraffin
embedded, sectioned at 4 μm and stained with Alcian Blue to evaluate acid mucopolysaccharide
accumulation in ECM.
2.8 Flow Cytometry assay
The cultured hASCs were detached from plates, washed twice with PBS, and resuspended at
1.0×107 cells/ml. For fluorescent antibody labeling, 100 μl of sample was incubated with
monoclonal mouse anti-human antibodies for 30 minutes at room temperature. The anti-human
antibodies of PE-labeled CD29, PE-labeled CD34, PE-labeled CD105, PE-labeled CD117,
PE-labeled CD166, and PerCp-Cy5-labeled HLA-DR were purchased from BD Pharmingen (BD
Biosciences, San Diego, CA). The anti-human antibodies of APC-labeled CD31, APC-labeled
CD45 and FITC-labeled CD73 were purchased from eBioscience (San Diego, CA, USA).
Subsequently, the samples were washed twice with PBS, centrifuged for 5 minutes at 15294 x g,
and fixed with 4% paraformaldehyde. The labeled cells were detected on a Flow Cytometer (BD
FACSAria) and the data was analyzed using FlowJo software (version 10.0.7, BD Biosciences).
2.9 hASCs culture in the hDAM hydrogel
For assessing the biocompatibility of hASCs with the hDAM hydrogel, 2×106 hASCs were
suspended with 1 ml of hDAM pre-gel solution, and the MSC serum-free culture medium was
added after gelation.
hASCs morphology was observed with a microscopy and Hematoxylin and eosin (H&E)
staining after 7 days culture. For H&E staining, hASCs cultured in hDAM hydrogels were fixed
with 4% paraformaldehyde, paraffin embedded and sectioned at 4 μm.
hASCs viability was assessed with the LIVE/DEAD Viability/Cytotoxicity Kit (Molecular
Probes, Thermo Fisher Scientific) for 1, 3, 5 or 7 days with 150 μl/well of hASCs-hydrogel added
in triplicate to a 48-well plate. At each time point, the culture medium were aspirated, rinsed twice
with Dulbecco's Phosphate-Buffered Saline (DPBS) (Gibco). Then, 100 μl/well DPBS containing
2 μM Calcein-AM (excitation 494 nm, emission 517nm) and 5 μM Ethidium homodimer-1
(excitation 528 nm, emission 617nm) was added to each well, and incubated at 37 for 30
minutes. Stained cells were visualized with Nikon Eclipse Ti fluorescence microscopy (Tokyo,
Japan). The viability of hASCs was evaluated by quantification the ratio of viable cells to total
cells.
hASCs proliferative activity was evaluated with Ki-67 flow cytometry analysis after a 2 day
cultivate period α-MEM basic medium. hASCs cultured within the hydrogel were collected by
digesting with LiberraseTM TL (50 μg/ml, Roche, Merck) for 30 minutes at 37 and 5% CO
2. The
collected cells pellets were washed twice with PBS, and resuspended at 1.0×107 cells/ml. For
Ki-67 fluorescent antibody labeling, 100 μl of samples were fixed with 200 μl
Fixation/Permeabilization solution for 10 minutes at room temperature. After fixation, washed
twice with 1 ml 1X BD Perm/WashTM buffer, centrifuged for 5 minutes at 2000 g. Subsequently,
added PE-labeled mouse anti-Ki-67 (1:50 dilution,BD Biosciences) and incubated for 30 minutes
at room temperature, and then washed again. The labeled cells were detected on a Flow Cytometer
(BD FACSAria) and the data was analyzed using FlowJo software (version 10.0.7, BD
Biosciences).
2.10 Angiogenesis cytokine assay
The angiogenic paracrine of hASCs cultured in the hDAM hydrogel were assessed by
collecting the conditioned medium for antibody array assay (n=3). 1×106 hASCs/well suspended
in 1 ml of the MSC serum-free medium or 1 ml of the hDAM pre-gel solution were seeded on
6-well plates. After hASCs adherence or the hydrogel gelation, 3 ml of α-MEM medium was
added to replace the complete medium or 2 ml of α-MEM medium was added to cover the
hydrogel. After 48 hours cultivation, 1 ml/well of the supernatant was collected and centrifuged at
4 , 425 x g for 10 minutes. For angiogenic cytokines assay, the collected medium were
concentrated 10 X times with a 10 KD centrifugal filter unite (Millipore) and the concentration of
cytokines were analyzed using the Quantibody Human Angiogenesis Array 1 (QAH-ANG-1;
RayBiotech, Norcross, GA) according to the manufacturer’s protocol (n=3). Briefly, completely
air dried glass slide was firstly blocked with 100 μl/well of sample diluent for 1 hour at room
temperature. After blocking, the diluent were decanted and 100 μl of standard cytokines or
samples were added to each well and incubated overnight at 4 . The unconjugated proteins were
washed out using the wash buffer and deionized water. Subsequently, 80 μl of biotinylated
antibody cocktail was added to each well and incubated for 2 hour at room temperature, and then
washed again. To detect the conjugated antibody, 80 μl of Cy3 equivalent dye-conjugated
streptavidin was added to each well and incubated for 1 hour at room temperature, and then
washed again. The fluorescence was detected on InnoScan microarray scanner (Innopsys,
Carbonne, France), and the data was extracted and computed using the Q-Analyzer software. The
concentrations of factors in the conditioned media were reported after compensated for.
2.11 Animal study
All animal experiments were performed according to relevant institutional guidelines and
regulations of Beijing Medical Experimental Animal Care Commission and were approved by the
Animal Research Ethics Committee of Chinese PLA General Hospital (No. 2017-x13-25).
Twenty 9-week-old male KK/Upj-Ay/J mice (diabetic mice) were purchased from HFK
Biotechnology (Beijing, China) and randomized to four experiment groups: control, hydrogel
dressing, local hASCs injection, or hASCs-hydrogel composite treatment. After intraperitoneal
induction of anesthesia with 1% pentobarbital sodium, the hair of dorsum and abdomen were
shaved and depilated. Two 8 mm full-thickness wounds were created on either side of the dorsal
midline with a skin biopsy punch (Electron Microscopy Sciences, Hatfield, UK). For the hASCs
injection group, 2.4×105 hASCs suspended in 60 μl of α-MEM were injected subcutaneously
around the wound edge. For the hASCs-hydrogel composite or hydrogel dressing group, 60 μl of
hDAM hydrogel with/without 2.4×105 hASCs were transferred to the wounds after gelation. For
the control group, 60 μl of α-MEM were injected subcutaneously around the wound edge. All
wounds were covered with a transparent occlusive dressing (Smith&Nephew, Watford, UK).
Photographs were taken at days 0, 7, 10 and 14 after wound with a paper circle (inner
diameter 1.2 cm, outer diameter 1.6 cm) placed on the wound as a reference. Wound area was
measured using the Image-J software and the percentage of original wound area was calculated at
each time point. At the end of observation, wound samples were harvested for histological analysis
using paraffin sections. H&E staining was used to evaluate wound healing quality and
immunohistochemical (IHC) staining with antibody to CD31 (1:200 dilution; Servicebio) were
used to evaluate wound neovascularization. The dermal microvessel density (MVD) was counted
using four hpf at 200 X from each group by three independent blinded observers.
2.12 Statistical Analysis
All the experimental data are expressed as mean ± standard deviation. The student’s unpaired
t test was used for two groups comparison when the data meet normality and homogeneity.
ANOVA with an appropriate post-hoc test was used for multiple comparison. All the statistical
work were performed with SPSS 18.0 statistical software and p < .05 was considered as statistical
significance.
3 RESULTS
3.1 Preparation of hDAM hydrogel
The preparation process of hDAM hydrogel involves decellularization of adipose tissue,
freeze-drying, grinding, pepsin digestion and pH neutralization (Figure 1a). After 5 days of
decellularization, a small volume of white and hydrated hDAM was obtained. Feeze-dried hDAM
was a loose and porous material, and the average production per 100 ml of lipoaspirate was 142.4
± 43.4 mg (dry weight) (n=6). Fine powdered hDAM was obtained by cyomilling the lyophilized
hDAM with KZ-II grind mill (KangTao technology, Wuhan, China) and filtering through a 80
mesh sieve. Effectively decellularization and delipidization was confirmed, as compared to native
adipose tissue, the dsDNA content (49.59 ± 8.27 ng/mg vs. 4.74 ± 0.71 ng/mg, p < .001) and
triglyceride content (18.76 ± 1.02 μg/mg vs. 0.21 ± 0.16 μg/mg, p < .001) in lyophilized hDAM
were significantly reduced.
The hDAM hydrogel was successfully prepared by pepsin mediated solubilization and pH
controlled neutralization. After 72 hours pepsin digestion, the hDAM powder was solubilized to a
homogenous liquid. The pre-gel solution was found to be easily manipulated with a pipette.
Sol-gel transition property of the hDAM hydrogel was observed after incubating the pre-gel
solution at 37 ℃ for 30 mi nutes (Figure 1b). After gelation, the hD A M hydrogel can maintain a
stable form and stick to a well-plate cover (Figure 1c).
3.2 Characterization of hDAM hydrogel
To assess the potential of the hDAM hydrogel as a delivery vehicle for hASCs, we performed
the biochemical composition, rheological property, gel ultrastructure analysis.
The soluble collagen content in the hydrogel was 0.72 ± 0.04 mg/mg lyophilized hDAM, and
the sGAG content in the hydrogel was 2.34 ± 0.47 ug/mg lyophilized hDAM. There was total 142
proteins identified in the hDAM digestion by LS-MS/MS (Supplementary Table S1), and most of
them were various kinds of collagens and glycoproteins. 44 proteins with high score (>20)
(Supplementary Table S2) were included into the subsequently Gene ontology (GO) analysis
(Figure 2a). The cellular component ontology annotation results showed most of these proteins
were ECM origin. The molecular function ontology annotation results revealed these constitutes
mainly act as binding molecule. The biological process ontology annotation results indicated that
the top three processes in which these proteins participated were platelet degranulation, ECM
organization, and innate immune response.
The measurement results of rheological property displayed that the prepared hDAM hydrogel
was a thermosensitive hydrogel (Figure 2b). The temperature at which the storage modulus (G’)
equal to the loss modulus (G’’) is the sol-gel transition point and our experiments observed that
this transition point occurred at 31 ℃. W hen the temperature rose to 37 ℃, the G’ was roughl y 5
times the G’’. The viscosity of this hydrogel material was 91.05 mPa·s before gelation and showed
a substantial increase to 6207.7 mPa·s after gelation.
SEM images showed that the hDAM hydrogel possessed a loosely organized porous
nanofiber structure with interconnected pore and randomly oriented fibers (Figure 2c, left panel).
Visual inspection of the algorithm outputs showed that the fiber network features have been
accurately extracted (Figure 2c, right panel). Based on the measurement data, the average fiber
diameter of the hydrogel was 64.4 ± 10.7 nm, the average pore size was 0.112 ± 0.019 μm2, and the
average node density was 7.15 ± 0.53 nodes/μm2.
3.3 Isolation and identification of hASCs
The isolated and expanded hASCs were plastic-adherent in standard culture conditions and
exhibited typical fibroblast-like or spindle-shaped morphology (Figure 3a). Multipotent
differentiation test showed that the hASCs were able to differentiate into adipocytes, osteocytes,
and chondrocytes under standard in vitro differentiating conditions as demonstrated by Oil-Red-O,
Alizarin Red, and Alcian Blue staining (Figure 3b). Flow cytometry analysis results showed that
98% of the cultured hASCs expressed the classical MSCs markers of CD29, CD73, CD90, CD105
and CD166, while these cells lacked expression (≤ 2.3% positive) of the endothelial cell specific
marker of CD31, the hematopoietic cells specific marker of CD45 and CD117, and the major
histocompatibility complex II (MHC-II) molecular of HLA-DR (Figure 3c).
3.4 hASCs survival and function in the hDAM hydrogel
To assess hASCs survival and function in the hDAM hydrogel, we evaluated the
morphology, viability, proliferation and angiogenic paracrine activity of hASCs cultured in the
hydrogel.
Microscopical and H&E staining images showed that hASCs cultured in the hydrogel
exhibited a healthy fibroblast-like phenotype (Figure 4a), indicating they were attached to the
hydrogel bioscaffold. The high viability of hASCs embedded in the hydrogel was similar to that in
standard condition over a period of seven days according to the LIVE/DEAD fluorescent staining
results (Figure 4b). The Ki-67 flow cytometric assay results showed hASCs within the hydrogel
proliferated significantly faster compared to those cultured on plastic surface (Hydrogel 34.3 ±
1.3 %; vs. Plated 20.3 ± 1.1 %, p<0.001) (Figure 4c).
Angiogenic cytokines assay was used to evaluate the paracrine activity of hASCs cultured in
the hDAM hydrogel (Figure 4d, e). The results showed that hASCs secreted a large amount of
hepatocyte growth factor (HGF), vascular growth factor-A (VEGF-A), angiopoietin (ANG) and
ANG-2. Compared to those cultured on plastic plate, hASCs cultured in the hDAM hydrogel
secreted a significantly high level of HGF (404.57 ± 30.23 pg/mL vs. 1182.54 ± 90.26 pg/mL, p <
.001) .
3.5 Effect of hASCs-hydrogel composite on impaired wound healing
We utilized the diabetic mice model with impaired wound healing ability, and created
excisional full-thickness wounds to test the therapeutic effect of hASCs-hydrogel composite in
vivo (Figure 4a). Wounds healed significantly faster when treated with hASCs-hydrogel
composite or local hASCs injection compared to those of control group (Figure 4b, c). Moreover,
there was a significant acceleration of wound closure at day 7 in hASCs-hydrogel composite
treated group (28.6 ± 5.2% original wound area, p = .002) compared to local hASCs injection
wounds (44.8 ± 6.9%). Wounds treated with hydrogel dressing showed improved healing only at
day 10, but no difference at day 7 or day 14 relative to those of control group. In addition to
accelerate wound closure, hASCs-hydrogel composite treatment also displayed an improved effect
on skin architecture regeneration, which included a better restoration of cutaneous appendages and
increase of dermis thickness (Figure 5d).
To assess the wound neovascularization, we performed CD31 IHC staining and MVD
assessment (Figure 5e, f). The MVD of wounds on healing Day 14 was significantly increased
when treated with hASCs-hydrogel composite compared to other three groups (68.7 ± 11.1 vessels
per high power fields (hpf); vs. hASCs injection, p = 0.02; vs. hydrogel dressing, p = 2.6×10-4; vs.
control, p = 1.3×10-6). Moreover, compared to untreated wounds, neovascularization was also
significantly augmented in wounds treated with local hASCs injection or hydrogel dressing (28.3
± 4.8 vessels per hpf; vs. hASCs injection, p = 2.6×10-4; vs. hydrogel dressing, p = 3.1×10-4).
However, there was no significant difference in MVD between wounds treated with local hASCs
injection and hydrogel dressing (52.0 ± 10.1 vs. 42.5 ± 5.2 vessels per hpf, p = .07).
4 DISCUSSION
In this study, we developed a hydrogel biological scaffold from hDAM for hASCs delivery to
accelerate chronic wound healing. The developed hDAM hydrogel was a thermosensitive
hydrogel, retained the biochemical complexity of the native ECM and formed a porous nanofiber
structure after gelation. The hydrogel can support hASCs adhesion, survival and proliferationand
hASCs cultured in the hydrogel exhibited enhanced paracrine activity. In the diabetic mice model
with excisional full-thickness skin wounds, mice treated with the hASCs-hydrogel composite
displayed accelerated wound closure and increased neovascularization.
Previous studies have suggested that biological scaffolds based cell delivery methods were
beneficial for MSCs engraftment in chronic wound healing [15]. Our study demonstrated that
delivery of hASCs in hDAM hydrogel can accelerate impaired wound closure and promote
angiogenesis. Traditionally, biological scaffolds used by other studies were sheet formed like
collagen sponge or acellular dermal matrix, which usually requested additional inoculation and
incubation process for cell attachment before delivery to wound beds and could not adapt well to
the irregular defect geometry [7, 8]. The hDAM hydrogel we developed was a liquid solution
before gelation, making it easier to encapsulate cells. Moreover, its sol-gel transition property is
more conducive to cell delivery and site attachment. In contrast to synthetic polymeric hydrogels,
the hDAM hydrogel is derived from native ECM of adipose tissue, and therefore contains
structural and functional biomolecules secreted by resident cells, thus in favor of communicating
with stem cells in an interactive and dynamical manner. This kind of hydrogels can also act as an
inductive template for constructive remodeling in tissue repair and regeneration through
recruitment of endogenous stem and progenitor cells, antimicrobial activity, and modulation of
macrophage polarization [16].
The paracrine mechanism of MSCs in accelerating wound healing has been confirmed by
many studies [17]. Previous researchers have reported that tissue engineering approaches
enhanced the paracrine function of MSCs. However, these results were mainly obtained from
culturing of MSCs on electrospun fiber scaffolds [18, 19]. Our study demonstrated that the hDAM
hydrogel culture condition can also increase the angiogenic cytokines secretion of hASCs. The
enhanced secretome may account for augmented neovascularization in vivo, because increased
HGF secreted by hASCs is a trophic factor which can facilitate neovascularization during wound
healing [20]. The upregulated pro-angiogenic cytokines secretion of hASCs cultured in the hDAM
hydrogel may share a similar down-stream signal pathway with stimulated secretion of MSCs due
to hypoxia condition [21]. In-depth understanding of the interaction between materials and stem
cells can help guide the preparation of optimized materials to maximize the therapeutic efficacy of
stem cells. This finding suggests that the prepared hDAM hydrogel can provide a favorable
functional niche for MSCs and improve their regeneration potential.
The relatively long gelation time of the hDAM hydrogel after injection into wound area may
be one obstacle for its clinical application. But this could be improved by mixed with synthetic
materials or cross-linking agents [22]. Another thing need to be noted is that, since this material is
derived from human adipose tissue, attention should be paid to possible transmission of infectious
diseases and the difference in tissue activity between donors. However, this problem can be solved
by strict screening of infectious diseases and careful evaluation and selection of donors [23].
Despite the above mentioned shortcomings, the developed hDAM hydrogel is still a promising
bioscaffold material for chronic wounds. On one hand, human tissue-derived materials have
advantages over xenogeneic-derived materials in bioactivity, biocompatibility and
biodegradability [24]. On the other hand, if clinical application of stem cell transplantation in
wound treatment is restricted out of concern about ethical disputes, our hDAM hydrogel still has
potential as a carrier for exosomes or conditioned medium of stem cells [25, 26]. For example, van
Dongen et al have made a research on adipose tissue-derived ECM hydrogels as a release
platform for secreted paracrine factors, which provided a promising therapeutic modality to
promote several important wound healing-related processes by releasing factors in a controlled
way [27].
5 CONCLUSION
A ECM-derived hydrogel was developed from human decellularized adipose tissue by pepsin
mediated digestion and pH controlled neutralization. The developed hDAM hydrogel was a
thermosensitive hydrogel, presented the biochemical complexity of native ECM and formed a
porous nanofiber structure after gelation. The hydrogel can support hASCs adhesion, survival and
proliferation. Compared to standard culture condition, hASCs cultured in the hydrogel exhibited
enhanced paracrine activity with increased secretion of HGF. In the diabetic mice model with
excisional full-thickness skin wounds, mice treated with the hASCs-hydrogel composite displayed
accelerated wound closure and increased neovascularization. Our results suggested that the
developed hDAM hydrogel can provide a favorable microenvironment for hASCs with augmented
regeneration potential to accelerate chronic wound healing.
ACKNOWLEDGMENTS
This study was supported by the National Key R&D Program of China [2017YFA0103100,
2017YFA0103103, 2017YFA0103104]; and the Guangzhou Health Care and Cooperative
Innovation Major Project [201704020224, 201803040011, 201803040008, 201803040005].
DATA AVAILABILITY STATEMENT
All the data that support the findings of this study are availability in the supplement material of
this article.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
REFERENCES
1. Eming SA, Martin P, Tomic M. Wound repair and regeneration: mechanisms, signaling, and
translation, Sci Transl Med. 2014; 6: 265sr6.
2. Frykberg RG, Banks J. Challenges in the Treatment of Chronic Wounds, Adv Wound Care.
2015; 4: 560-582.
3. Hu MS, Borrelli MR, Lorenz HP, et al. Mesenchymal Stromal Cells and Cutaneous Wound
Healing: A Comprehensive Review of the Background, Role, and Therapeutic Potential, Stem
Cells Int. 2018; 2018: 6901983.
4. Lee DE, Ayoub N, Agrawl DK. Mesenchymal stem cells and cutaneous wound healing:
novel methods to increase cell delivery and therapeutic efficacy, Stem Cell Res Ther. 2016; 7.
5. Dash BC, Xu Z, Lin L, et al. Stem Cells and Engineered Scaffolds for Regenerative Wound
Healing, Bioengineering (Basel). 2018; 5: 23.
6. Turner NJ, Badylak SF. The Use of Biologic Scaffolds in the Treatment of Chronic
Nonhealing Wounds, Adv Wound Care. 2015; 4: 490-500.
7. O'Loughlin A, Kulkarni M, Creane M, et al. Topical administration of allogeneic
mesenchymal stromal cells seeded in a collagen scaffold augments wound healing and increases
angiogenesis in the diabetic rabbit ulcer, Diabetes. 2013; 62: 2588-2594.
8. Nie C, Zhang G, Yang D, et al. Targeted delivery of adipose-derived stem cells via acellular
dermal matrix enhances wound repair in diabetic rats, J Tissue Eng Regen Med. 2015; 9:
224-235.
9. Yu L, Ding J. Injectable hydrogels as unique biomedical materials, Chem Soc Rev. 2008; 37:
1473-1481.
10. Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design variables and
applications, Biomaterials. 2003; 24: 4337-4351.
11. Jabbar E. Hydrogels for Cell Delivery, Gels. 2018; 4.
12. Spang MT, Christman KL. Extracellular matrix hydrogel therapies: In vivo applications and
development, Acta Biomater. 2018; 68: 1-14.
13. Zhou ZQ, Chen Y, Chai M, et al. Adipose extracellular matrix promotes skin wound healing
by inducing the differentiation of adiposederived stem cells into fibroblasts, Int J Mol Med. 2019;
43: 890-900.
14. Li J, Curley JL, Floyd ZE, et al. Isolation of Human Adipose-Derived Stem Cells from
Lipoaspirates, Methods Mol Biol. 2018; 1773: 155-165.
15. Altman AM, Yan Y, Matthias N, et al. IFATS collection: Human adipose-derived stem cells
seeded on a silk fibroin-chitosan scaffold enhance wound repair in a murine soft tissue injury
model, Stem Cells (Dayton, Ohio). 2009; 27: 250-258.
16. Saldin LT, Cramer MC, Velankar SS, et al. Extracellular matrix hydrogels from
decellularized tissues: Structure and function, Acta Biomater. 2017; 49: 1-15.
17. Lee SH, Jin SY, Song JS, et al. Paracrine effects of adipose-derived stem cells on
keratinocytes and dermal fibroblasts, Ann dermatol. 2012; 24: 136-143.
18. Carter K, Lee HJ, Na KS, et al. Characterizing the impact of 2D and 3D culture conditions
on the therapeutic effects of human mesenchymal stem cell secretome on corneal wound healing
in vitro and ex vivo, Acta Biomater. 2019; 99: 247-257.
19. Su N, Gao PL, Wang K, et al. Fibrous scaffolds potentiate the paracrine function of
mesenchymal stem cells: A new dimension in cell-material interaction, Biomaterials. 2017; 141:
74-85.
20. Ding S, Merkulova T, Han ZC, Tobelem, G. HGF receptor up-regulation contributes to the
angiogenic phenotype of human endothelial cells and promotes angiogenesis in vitro, Blood.
2003; 101: 4816-4822.
21. Muscari C, Giordano E, Bonaf F, et al. Priming adult stem cells by hypoxic pretreatments
for applications in regenerative medicine, J Biomed Sci. 2013; 20: 63.
22. Zhu J, Marchant RE. Design properties of hydrogel tissue-engineering scaffolds, Expert
Rev Med Devices. 2011; 8: 607-626.
23. Chen FM, Liu X. Advancing biomaterials of human origin for tissue engineering, Prog
Polym Sci. 2016; 53: 86-168.
24. Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold material:
Structure and function, Acta Biomater. 2009; 5: 1-13.
25. Wu P, Zhang B, Shi H, et al. MSC-exosome: A novel cell-free therapy for cutaneous
regeneration, Cytotherapy. 2018; 20: 291-301.
26. Zhou P, Li X, Zhang B, et al. A human umbilical cord mesenchymal stem cell-conditioned
medium/chitosan/collagen-Glycerophosphate thermosensitive hydrogel promotes burn injury
healing in mice, Biomed Res Int. 2019; 2019: 5768285.
27. Van Dongen JA, Getova A, Brrouwer LA, et al. Adipose tissue-derived extracellular matrix
hydrogels as a release platform for secreted paracrine factors. J Tissue Eng Regen Med. 2019; 13:
973-985.
FIGURE LEGENDS
FIGURE 1 Preparation of hDAM hydrogel from human adipose tissue. Major steps of the
hDAM hydrogel preparation (a). Sol-gel transition property of the hDAM hydrogel (b).
Macroscopic appearance of the hDAM hydrogel (c).
FIGURE 2 Characterization of hDAM hydrogel. Gene ontology (GO) analysis results of the
high score proteins (>20) identified by LC-MS/MS (a). Rheological property of the hDAM
hydrogel (b). Scanning electron micrographs of the hDAM hydrogel (left panel) and the extracted
fiber network characteristics by algorithm (right panel) (c).
FIGURE 3 Identification of hASCs. Morphology of cultured hASCs (a). Multipotent
differentiation of hASCs (b), adipogenic differention was evaluated by Oil-Red-O staining of lipid
droplets (first column), osteogenic differentiation was evaluated by Alizarin Red staining of
deposited calcium (second column), chondrogenic differentiation was evaluated by Alcian Blue
staining of accumulated acid mucopolysaccharide in extracellular matrix (third column), scale bar
= 50 μm. Surface markers of hASCs as detected by flow cytometry (c).
FIGURE 4 hASCs survival and function in the hDAM hydrogel. Microscopy image and H&E
staining of hASCs cultured in the hydrogel for 7 days (a), scale bar = 100 μm. Live/Dead assay of
hASCs viability in the hydrogel (b), right panel is a representative fluorescence image at 7 days,
scale bar = 100 μm. Ki-67 flow cytometric analysis of hASCs proliferation in the hydrogel (c).
Heatmap of angiogenic cytokines concentration in conditioned medium from hASCs cultured in
the hydrogel and plastic plate (d), the scaled values represent the concentration of proteins
(pg/mL), and relative amounts of proteins in the conditioned medium from hASCs cultured in the
hydrogel compared to plastic plate (e; *p < .05, **p < .01, ***p < .001).
FIGURE 5 Effect of hASCs-hydrogel composite on impaired wound healing. Excisional
full-thickness wound model preparation and hASCs-hydrogel composite engraftment (a). Gross
photos (b) and Percentage of original wound area (c). H&E staining of wounds at day 14 (d), scale
bar = 100 μm. CD31 IHC staining of wounds at day14 (e), scale bar = 200 μm. and Quantification
of MVD (f; *p < .05, **p < .01, ***p < .001).
FIGURE 1
FIGURE 2
Accepted Article
This article is protected by copyright. All rights reserved.
FIGURE 3
Accepted Article
This article is protected by copyright. All rights reserved.
FIGURE 4
Accepted Article
This article is protected by copyright. All rights reserved.
FIGURE 5
Accepted Article
This article is protected by copyright. All rights reserved.
... During the proliferative phase of wound healing, ASCs also promote angiogenesis within the wounds through the secretion of pro-angiogenic factors, especially VEGF, Ang-1, and HGF [29,58,59]. We have shown that ASC-based dressings increase the vascular density (2.5-fold) in the granulation tissue and that the new capillary network is more organized. ...
... We have shown that ASC-based dressings increase the vascular density (2.5-fold) in the granulation tissue and that the new capillary network is more organized. Several studies have documented the beneficial effects of ASC-loaded scaffolds on neoangiogenesis of full-thickness wounds of various models of diabetic rodents; the vascular density of treated wounds increasing 1.4-to 6-fold when compared to wounds not receiving ASCs [53,55,56,58,[60][61][62][63]. We assessed the secretion levels of multiple molecules involved in the stimulation of the wound healing proliferative phase processes with an emphasis on pro-angiogenic factors. ...
Article
Full-text available
Long-term diabetes often leads to chronic wounds refractory to treatment. Cell-based therapies are actively investigated to enhance cutaneous healing. Various cell types are available to produce biological dressings, such as adipose-derived stem/stromal cells (ASCs), an attractive cell source considering their abundancy, accessibility, and therapeutic secretome. In this study, we produced human ASC-based dressings under a serum-free culture system using the self-assembly approach of tissue engineering. The dressings were applied every 4 days to full-thickness 8-mm splinted skin wounds created on the back of polygenic diabetic NONcNZO10/LtJ mice and streptozotocin-induced diabetic K14-H2B-GFP mice. Global wound closure kinetics evaluated macroscopically showed accelerated wound closure in both murine models, especially for NONcNZO10/LtJ; the treated group reaching 98.7% ± 2.3% global closure compared to 76.4% ± 11.8% for the untreated group on day 20 (p=0.0002). Histological analyses revealed that treated wounds exhibited healed skin of better quality with a well-differentiated epidermis and a more organized, homogeneous, and 1.6-fold thicker granulation tissue. Neovascularization, assessed by CD31 labeling, was 2.5-fold higher for the NONcNZO10/LtJ treated wounds. We thus describe the beneficial impact on wound healing of biologically active ASC-based dressings produced under an entirely serum-free production system facilitating clinical translation.
... In recent years, AAM has gradually become a promising approach for tissue repair and regeneration. In the full-thickness skin wound model, AAM-hydrogel accelerated wound closure and increased neovascularization (Chen et al., 2020). During the treatment of bone defects, AAM is defined as an alternative bone graft material with preserved ECM components (Ahn et al., 2022). ...
... In vivo study of nude mice with full-thickness wounds showed that AAM promotes wound healing and that AAM combined with adipose stem cells (ASCs) elicits the best effects (Xia et al., 2020). Similarly, delivery of ASCs using a hydrogel biological scaffold derived from human decellularized adipose matrix accelerates chronic wound healing and increases neovascularization (Chen et al., 2020). These studies revealed the therapeutic potential of AAM to promote wound healing. ...
Article
Full-text available
Dermal white adipocytes are closely associated with skin homeostasis and wound healing. However, it has not been fully investigated whether adipose-derived products improve wound healing. Here, we obtained adipose acellular matrix (AAM) and adipose-derived growth factors (ADGFs) from human adipose tissue and fabricated an ADGF-loaded AAM via surface modification with heparin. The product, HEP-ADGF-AAM, contained an adipose-derived scaffold and released ADGFs in a controlled fashion. To test its efficacy in promoting wound healing, mice with full thickness wound received three different treatments: HEP-ADGF-AAM, AAM and ADM. Control mice received no further treatments. Among these treatments, HEP-ADGF-AAM best improved wound healing. It induced adipogenesis in situ after in vivo implantation and provided an adipogenic microenvironment for wounds by releasing ADGFs. HEP-ADGF-AAM not only induced adipocyte regeneration, but also enhanced fibroblast migration, promoted vessel formation, accelerated wound closure, and enhanced wound epithelialization. Moreover, there was a close interaction between HEP-ADGF-AAM and the wound bed, and collagen was turned over in HEP-ADGF-AAM. These results show that HEP-ADGF-AAM might substantially improve re-epithelialization, angiogenesis, and skin appendage regeneration, and is thus a promising therapeutic biomaterial for skin wound healing.
... Furthermore, ECM contains cytokines that stimulate cell proliferation, increase blood perfusion, and promote vascularization, all of which aid in tissue regeneration. 23 All of these mechanisms are consistent with the therapeutic objectives of atrophic striae. The ECM in SVF-gel can accommodate ADSCs while also protecting them from phagocytosis by macrophages, allowing SVF-gel to play a multifaceted role in the treatment of SD. ...
Article
Full-text available
Background Striae Distensae (SD) is a common dermatological lesion. The mechanism of formation is unclear, the prevailing theory is mechanical pulling of the skin and hormonal changes. Traditional SD treatment methods include topical drugs, photoelectric therapy, stripping and others, but each has limitations. Stromal vascular fraction gel (SVF‐gel) is a filler physically prepared from granular fat, enriched with adipose‐derived stem cells (ADSCs) and extracellular matrix (ECM). A good effect in the treatment of neck lines, wounds, acne, and other aspects. SD formation and treatment goals are comparable to those of neck lines. In this study, SVF‐gel filling will be used to treat mature SD, and its effectiveness and safety will be discussed in detail. Methods From December 2019 to June 2022, recruit patients who want to treat SD caused by obesity or pregnancy among those who have “autologous fat aspiration” to change their body shape. Preoperatively, the area to be treated for SD was marked, autologous fat aspiration was performed, and the aspirated fat was prepared as SVF‐gel and filled into the preoperatively marked SD. All patients had preoperative and postoperative follow‐up with planar photographs and VISIA skin analyzer photographs to assess surgical results and safety from subjective and objective perspectives. Results A total of 36 patients were enrolled, with 31 of them successfully followed up on. The mean Global Aesthetic Improvement Scale (GAIS) score six months after surgery was 1.87 ± 0.03. At six months postoperatively, the overall patient satisfaction rate was 90%. The depth, area, and color of SD improved six months after surgery, and no serious complications occurred in any of the patients. Conclusions SVF‐gel is a safe and effective method of improving mature SD and can be used as a clinical treatment option.
Article
Full-text available
Tissue-engineered skin substitutes (TESS) emerged as a new therapeutic option to improve skin transplantation. However, establishing an adequate and rapid vascularization in TESS is a critical factor for their clinical application and successful engraftment in patients. Therefore, several methods have been applied to improve the vascularization of skin substitutes including (i) modifying the structural and physicochemical properties of dermal scaffolds; (ii) activating biological scaffolds with growth factor-releasing systems or gene vectors; and (iii) developing prevascularized skin substitutes by loading scaffolds with capillary-forming cells. This review provides a detailed overview of the most recent and important developments in the vascularization strategies for skin substitutes. On the one hand, we present cell-based approaches using stem cells, microvascular fragments, adipose tissue derived stromal vascular fraction, endothelial cells derived from blood and skin as well as other pro-angiogenic stimulation methods. On the other hand, we discuss how distinct 3D bioprinting techniques and microfluidics, miRNA manipulation, cell sheet engineering and photosynthetic scaffolds like GelMA, can enhance skin vascularization for clinical applications. Finally, we summarize and discuss the challenges and prospects of the currently available vascularization techniques that may serve as a steppingstone to a mainstream application of skin tissue engineering. Graphical abstract
Article
The nature of the wound typically dictates the available wound-healing therapies. On the other hand, inadequate care results in persistent wounds, fibrosis, tissue loss of function, and ultimately dismemberment. Suturing, unloading, irrigation, debridement, negative pressure therapies, growth factor supplementation, and grafting are examples of current medicines that all have limitations in terms of meeting the needs of full treatment. Natural or synthetic materials/polymers have been utilized to prepare smart thermosensitive hydrogels to facilitate quick and targeted action on wounds. These are intelligent hydrogel system that shows sol-gel transitions at physiological body temperature. Hydrogel provides a moist environment, scaffold-like structure, and localized delivery of drug/growth factor that speed up wound healing even if it eliminates side effects associated with systemic administration. In this article, we summarized the detailed mechanisms of wound healing, conventional strategies, and ongoing thorough research work in the field of thermosensitive hydrogels utilized for wound healing. Moreover, the clinical needs of this formulation, as evident from the commercially available systems are also described in the prior art.
Article
Full-text available
Matrix-bound nanovesicles (MBVs) are a recently discovered type of extracellular vesicles (EVs), and they are characterised by a strong adhesion to extracellular matrix structural proteins (ECM) and ECM-derived biomaterials. MBVs contain a highly bioactive and tissue-specific cargo that recapitulates the biological activity of the source ECM. The rich content of MBVs has shown to be capable of potent cell signalling and of modulating the immune system, thus the raising interest for their application in regenerative medicine. Given the tissue-specificity and the youthfulness of research on MBVs, until now they have only been isolated from a few ECM sources. Therefore, the objective of this research was to isolate and identify the presence of MBVs in decellularised bovine pericardium ECM and to characterise their protein content, which is expected to play a major role in their biological potential. The results showed that nanovesicles, corresponding to the definition of recently described MBVs, could be isolated from decellularised bovine pericardium ECM. Moreover, these MBVs were composed of numerous proteins and cytokines, thus preserving a highly potential biological effect. Overall, this research shows that bovine pericardium MBVs show a rich and tissue-specific biological potential.
Article
Full-text available
Wound healing is a dynamic and complex process in which the microenvironment at the wound site plays an important role. As a common material for wound healing, dressings accelerate wound healing and prevent external wound infections. Hydrogels have become a hot topic in wound-dressing research because of their high water content, good biocompatibility, and adjustable physical and chemical properties. Intelligent hydrogel dressings have attracted considerable attention because of their excellent environmental responsiveness. As smart polymer hydrogels, thermosensitive hydrogels can respond to small temperature changes in the environment, and their special properties make them superior to other hydrogels. This review mainly focuses on the research progress in thermosensitive intelligent hydrogel dressings for wound healing. Polymers suitable for hydrogel formation and the appropriate molecular design of the hydrogel network to achieve thermosensitive hydrogel properties are discussed, followed by the application of thermosensitive hydrogels as wound dressings. We also discuss the future perspectives of thermosensitive hydrogels as wound dressings and provide systematic theoretical support for wound healing.
Article
With the development of tissue engineering, the application of decellularized adipose matrix as scaffold material in tissue engineering has been intensively explored due to its wide source and excellent potential in tissue regeneration. Decellularized adipose matrix is a promising candidate for adipose tissue regeneration, while modification of decellularized adipose matrix scaffold can also allow it to transcend the limitations of adipose tissue source properties and applied to other tissue engineering fields, including cartilage and bone tissue engineering, neural tissue engineering, and skin tissue engineering. In this review, we summarized the development of the applications of decellularized adipose matrix in different tissue engineering and present future perspectives. Level of Evidence III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Article
Full-text available
We investigated the effects of a human umbilical cord mesenchymal stem cell-conditioned medium (MSC-CM)/chitosan/collagen/ β -glycerophosphate ( β -GP) thermosensitive hydrogel (MSC-CM/hydrogel) on mice with third-degree burns. MSC-CM was collected and mixed with chitosan, collagen, and β -GP to generate the thermosensitive MSC-CM/hydrogel, which was stored in the liquid phase at 4°C. The wounds of established third-degree burned mice were then externally covered with the MSC-CM/hydrogel, which formed a gel when placed on the wounds at physiological temperature. Injured mice in three additional groups were treated with unconditioned MSC medium (UM), MSC-CM, or UM/chitosan/collagen/ β -GP thermosensitive hydrogels. Skin wound samples were obtained 4, 14, and 28 days after burning for further analysis by hematoxylin and eosin and Ki-67 staining. Wound healing rates and times, in addition to immunohistochemical results, were then compared and analyzed among the four groups. Application of the MSC-CM/hydrogel shortened healing time, limited the area of inflammation, enhanced reepithelialization, promoted the formation of high-quality, well-vascularized granulation tissue, and attenuated the formation of fibrotic and hypertrophic scar tissue. In summary, MSC-CM/hydrogel effectively promotes wound healing in third-degree burned mice.
Article
Full-text available
Introduction Fat grafting is an established clinical intervention to promote tissue repair. The role of the fat's extracellular matrix (ECM) in regeneration is largely neglected. We investigated in vitro the use of human adipose tissue‐derived ECM hydrogels as release platform for factors secreted by adipose derived stromal cells (ASC) Material & methods Lipoaspirates from non‐diabetic and diabetic donors were decellularized. Finely powdered acellular ECM was evaluated for cell remainders and DNA content. Acellular ECM was digested and hydrogels were formed at 37 °C and their viscoelastic relaxation properties investigated. Release of ASC‐released factors from hydrogels was immune‐assessed and bio‐activity was determined by fibroblast proliferation and migration and endothelial angiogenesis. Results Acellular ECM contained no detectable cell remainders and negligible DNA contents. Viscoelastic relaxation measurements yielded no data for diabetic‐derived hydrogels due to gel instability. Hydrogels released several ASC‐released factors concurrently in a sustained fashion. Functionally, released factors stimulated fibroblast proliferation and migration as well as angiogenesis. No difference between non‐diabetic and diabetic hydrogels in release of factors was measured. Conclusion Adipose ECM hydrogels incubated with released factors by ASC are a promising new therapeutic modality to promote several important wound healing related processes by releasing factors in a controlled way.
Article
Full-text available
Fibroblasts are the major effector cells of skin wound healing. Adipose‑derived stem cells can differentiate into fibroblasts under certain conditions. In the present study, it was hypothesized that adipose‑derived stem cells (ADSCs) could be induced by the adipose extracellular matrix (ECM) to differentiate into fibroblasts in order to promote skin wound healing. First, flow cytometry was used to detect the ratio of fibroblasts and relative expression of the fibroblast markers cytokeratin 19 (CK19) and vimentin in ADSCs. Then, the effect of the adipose ECM during the differentiation of ADSCs into fibroblasts was investigated by detecting the total amount of collagen fibers and degree of fibrosis, and the proliferation and cell cycle of differentiated fibroblasts, using the MTT assay and flow cytometry analysis respectively. Finally, a mouse skin wound model was established and treated with PBS, ADSC suspension or ECM + ADSCs to compare wound healing rate and expression of collagen I and collagen III by immunohistochemistry. Following induction of ADSCs with the adipose ECM, more fibroblasts were found, expression of CK19 and vimentin increased, and a greater degree of fibrosis occurred, which revealed the positive effect of the adipose ECM on the differentiation of ADSCs into fibroblasts. In addition, the induced fibroblasts had enhanced proliferation activity, with more cells in the S phase and fewer in the G2/M phase. The in vivo experiment indicated that the ECM produced by the ADSCs had a faster wound healing rate and increased expression of collagen I and collagen III compared with mice injected with PBS or ADSCs alone, which verified that ADSCs induced by the adipose ECM had a positive effect on skin wound healing. The present study demonstrated that the adipose ECM in combination with ADSCs may be a novel therapeutic target for the repair of skin injury, due to the ability of the adipose ECM to induce the differentiation of ADSCs into fibroblasts and to facilitate the wound healing process.
Article
Full-text available
Article
Full-text available
Cutaneous wound repair is a highly coordinated cascade of cellular responses to injury which restores the epidermal integrity and its barrier functions. Even under optimal healing conditions, normal wound repair of adult human skin is imperfect and delayed healing and scarring are frequent occurrences. Dysregulated wound healing is a major concern for global healthcare, and, given the rise in diabetic and aging populations, this medicoeconomic disease burden will continue to rise. Therapies to reliably improve nonhealing wounds and reduce scarring are currently unavailable. Mesenchymal stromal cells (MSCs) have emerged as a powerful technique to improve skin wound healing. Their differentiation potential, ease of harvest, low immunogenicity, and integral role in native wound healing physiology make MSCs an attractive therapeutic remedy. MSCs promote cell migration, angiogenesis, epithelialization, and granulation tissue formation, which result in accelerated wound closure. MSCs encourage a regenerative, rather than fibrotic, wound healing microenvironment. Recent translational research efforts using modern bioengineering approaches have made progress in creating novel techniques for stromal cell delivery into healing wounds. This paper discusses experimental applications of various stromal cells to promote wound healing and discusses the novel methods used to increase MSC delivery and efficacy.
Article
Full-text available
The normal wound healing process involves a well-organized cascade of biological pathways and any failure in this process leads to wounds becoming chronic. Non-healing wounds are a burden on healthcare systems and set to increase with aging population and growing incidences of obesity and diabetes. Stem cell-based therapies have the potential to heal chronic wounds but have so far seen little success in the clinic. Current research has been focused on using polymeric biomaterial systems that can act as a niche for these stem cells to improve their survival and paracrine activity that would eventually promote wound healing. Furthermore, different modification strategies have been developed to improve stem cell survival and differentiation, ultimately promoting regenerative wound healing. This review focuses on advanced polymeric scaffolds that have been used to deliver stem cells and have been tested for their efficiency in preclinical animal models of wounds.
Article
The therapeutic effects of secreted factors (secretome) produced by bone marrow-derived human mesenchymal stem cells (MSCs) were evaluated as a function of their growth in 2D culture conditions and on 3D electrospun fiber scaffolds. Electrospun fiber scaffolds composed of polycaprolactone and gelatin were fabricated to provide a 3D microenvironment for MSCs, and their mechanical properties were optimized to be similar to corneal tissue. The secretome produced by the MSCs cultured on 3D fiber matrices versus 2D culture dishes were analyzed using a Luminex immunoassay, and the secretome of MSCs cultured on the 3D versus 2D substrates showed substantial compositional differences. Concentrations of factors such as HGF and ICAM-1 were increased over 5 times in 3D cultures compared to 2D cultures. In vitro proliferation and scratch-based wound healing assays were performed to compare the effects of the secretome on corneal fibroblast cells (CFCs) when delivered synchronously from co-cultured MSCs through a trans-well co-culture system versus asynchronously after harvesting the factors separately and adding them to the media. Cell viability of CFCs was sustained for 6 days when co-cultured with MSCs seeded on the fibers but decreased with time under other conditions. Scratch assays showed 95% closure at 48 h when CFCs were co-cultured with MSCs seeded on fibers, while the control group only exhibited 50% closure at 48 h. Electrospun fibers seeded with MSCs were then applied to a rabbit corneal organ culture system, and MSCs seeded on fibers promoted faster epithelialization and less scarring. Corneas were fixed and stained for alpha smooth muscle actin (α-SMA), and then analyzed by confocal microscopy. Immunostaining showed that expression of α-SMA was lower in corneas treated with MSCs seeded on fibers, suggesting suppression of myofibroblastic transformation. MSCs cultured on electrospun fibers facilitate wound healing in CFCs and on explanted corneas through differential secretome profiles compared to MSCs cultured on 2D substrates. Future work is merited to further understand the nature and basis of these differences and their effects in animal models. Statement of significance Previous studies have shown that the secretome of bone marrow-derived mesenchymal stem cells (MSC) is promotes corneal wound healing by facilitating improved wound closure rates and reduction of scarring and neovascularization. The present research is significant because it provides evidence for the modulation of the secretome as a function of the MSC culture environment. This leads to differential expression of therapeutic factors secreted, which can impact corneal epithelial and stromal healing after severe injury. In addition, this article shows that co-continuous delivery of the MSC secretome improves cell migration and proliferation over aliquoted delivery, and that MSCs grown on three-dimensional electrospun fiber constructs may provide a favorable microenvironment for cultured MSCs and as a carrier to deliver their secreted factors to the ocular surface.
Chapter
Adipose tissue is as an abundant and accessible source of stem cells with multipotent properties suitable for tissue engineering and regenerative medical applications. Here, we describe methods from our own laboratory and the literature for the isolation and expansion of adipose-derived stem cells (ASCs). We present a large scale procedure suitable for processing >100 mL volumes of lipoaspirate tissue specimens by collagenase digestion and a related procedure suitable for processing adipose tissue aspirates without digestion.
Article
Cutaneous regeneration is a dynamic and complex process that requires a series of coordinated interactions involving epidermal cells, dermal cells, growth factors, the extracellular matrix (ECM), nerves and blood vessels at a damaged site. Mesenchymal stromal cells (MSCs) have been reported to participate in all afore-mentioned stages. Exosomes are one of the key secretory products of MSCs, resembling the effect of parental MSCs. They can shuttle various proteins, messenger RNA (mRNA) and microRNAs (miRNAs) to modulate the activity of recipient cells, and play important roles in cutaneous wound healing. Compared with MSCs, exosomes are more convenient to store and transport. Moreover, they avoid many risks associated with cell transplantation. Therefore, MSC-exosome-mediated therapy may be more safe and efficient. In this review, we summarize the latest studies and observations on the role of MSC-exosome in the acute and chronic wound model and provide a comprehensive understanding of the role of exosomes in wound healing. This review can assist investigators in exploring new therapeutic strategies for enhancing the efficacy of MSC-exosome for cutaneous repair and regeneration.
Article
Decellularized extracellular matrix (ECM) has been widely used for tissue engineering applications and is becoming increasingly versatile as it can take many forms, including patches, powders, and hydrogels. Following additional processing, decellularized ECM can form an inducible hydrogel that can be injected, providing for new minimally-invasive procedure opportunities. ECM hydrogels have been derived from numerous tissue sources and applied to treat many disease models, such as ischemic injuries and organ regeneration or replacement. This review will focus on in vivo applications of ECM hydrogels and functional outcomes in disease models, as well as discuss considerations for clinical translation. Statement of Significance Extracellular matrix (ECM) hydrogel therapies are being developed to treat diseased or damaged tissues and organs throughout the body. Many ECM hydrogels are progressing from in vitro models to in vivo biocompatibility studies and functional models. There is significant potential for clinical translation of these therapies since one ECM hydrogel therapy is already in a Phase 1 clinical trial.