ArticlePDF Available

Activin B-activated Cdc42 signaling plays a key role in regulating adipose-derived mesenchymal stem cells-mediated skin wound healing

Authors:
  • 深圳大学

Abstract and Figures

Background In our previous study, activin B in combination with ADSCs enhances skin wound healing. However, the underlying molecular mechanisms are not well studied. Cdc42 is recognized to play a critical role in the regulation of stem cells. Methods Pull-down assay was performed to investigate the activity of Cdc42. The dominant-negative mutant of Cdc42 (Cdc42N17) was used to explore the role of Cdc42 in activin B-induced ADSCs migration, proliferation, and secretion in vitro. Cdc42N17-transfected ADSCs were injected into a full-thickness excisional wound model to explore their efficiency in wound healing in vivo. The wound healing efficacy was evaluated by the wound closure rates and histological examination. The neovascularization and wound contraction were detected by immunohistochemistry staining of CD31 and α-SMA. Finally, the underlying mechanisms were explored by RNA sequencing. Results Cdc42N17 inhibited ADSCs migration, proliferation, and secretion induced by activin B. Furthermore, Cdc42N17-transfected ADSCs inhibited the wound closure rate and suppressed the expression of CD31 and α-SMA induced by activin B in vivo. The RNA sequencing showed that the differentially expressed genes in Cdc42N17-transfected ADSCs versus ADSCs were associated with cell migration, proliferation, and adhesion. Further study revealed that the Cdc42-Erk-Srf pathway was required for activin B-induced proliferation in ADSCs. Conclusions Our study indicates that Cdc42 plays a crucial role in ADSCs-mediated skin wound healing induced by activin B. Graphical Abstract
This content is subject to copyright. Terms and conditions apply.
Huangetal. Stem Cell Research & Therapy (2022) 13:248
https://doi.org/10.1186/s13287-022-02918-9
RESEARCH
Activin B-activated Cdc42 signaling plays
akey role inregulating adipose-derived
mesenchymal stem cells-mediated skin wound
healing
Simin Huang1†, Xueer Wang1†, Min Zhang1, Mianbo Huang1, Yuan Yan1, Yinghua Chen1, Yijia Zhang1, Jinfu Xu1,
Lingwei Bu1, Ruyi Fan1, Huiyi Tang1, Canjun Zeng3, Lu Zhang2* and Lin Zhang1*
Abstract
Background: In our previous study, activin B in combination with ADSCs enhances skin wound healing. However,
the underlying molecular mechanisms are not well studied. Cdc42 is recognized to play a critical role in the regulation
of stem cells.
Methods: Pull-down assay was performed to investigate the activity of Cdc42. The dominant-negative mutant of
Cdc42 (Cdc42N17) was used to explore the role of Cdc42 in activin B-induced ADSCs migration, proliferation, and
secretion in vitro. Cdc42N17-transfected ADSCs were injected into a full-thickness excisional wound model to explore
their efficiency in wound healing in vivo. The wound healing efficacy was evaluated by the wound closure rates and
histological examination. The neovascularization and wound contraction were detected by immunohistochemistry
staining of CD31 and α-SMA. Finally, the underlying mechanisms were explored by RNA sequencing.
Results: Cdc42N17 inhibited ADSCs migration, proliferation, and secretion induced by activin B. Furthermore,
Cdc42N17-transfected ADSCs inhibited the wound closure rate and suppressed the expression of CD31 and α-SMA
induced by activin B in vivo. The RNA sequencing showed that the differentially expressed genes in Cdc42N17-trans-
fected ADSCs versus ADSCs were associated with cell migration, proliferation, and adhesion. Further study revealed
that the Cdc42-Erk-Srf pathway was required for activin B-induced proliferation in ADSCs.
Conclusions: Our study indicates that Cdc42 plays a crucial role in ADSCs-mediated skin wound healing induced by
activin B.
Keywords: ADSCs, Wound healing, Cdc42, RNA-seq
© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/. The Creative Commons Public Domain Dedication waiver (http:// creat iveco
mmons. org/ publi cdoma in/ zero/1. 0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
Open Access
Simin Huang and Xueer Wang have contributed equally to this work
*Correspondence: zlulu70@126.com; zlilyzh@126.com
1 Department of Histology and Embryology, NMPA Key Laboratory for Safety
Evaluation of Cosmetics, Key Laboratory of Construction and Detection
in Tissue Engineering of Guangdong Province, School of Basic Medical
Sciences, Southern Medical University, Guangzhou 510515, People’s Republic
of China
2 Key Laboratory of Functional Proteomics of Guangdong Province, Key
Laboratory of Mental Health of the Ministry of Education, School of Basic
Medical Sciences, Southern Medical University, Guangzhou 510515, China
Full list of author information is available at the end of the article
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Introduction
Stem-cell-based therapeutic strategies have shown con-
siderable potential to improve the rate and quality of skin
wound healing [1, 2]. Among a wide variety of stem cells
that show efficacies in wound healing, adipose-derived
mesenchymal stem cells (ADSCs) seem to possess the
least limitations for clinical applications [3, 4]. Trans-
plantation of ADSCs into full-thickness wounds is able
to improve wound healing by promoting angiogenesis,
modulating immune response, and inducing epitheliali-
zation in the wound [5, 6].
Many growth factors and cytokines were shown to play
a key role in the process of cutaneous wound healing
[710]. For example, activin B, a member of the TGF-β
superfamily, is involved in the process of skin wound
repair [11, 12]. In a previous study, we reported that
activin B was able to induce actin stress fiber formation
and migration of ADSCs to promote skin wound heal-
ing [13]. However, the mechanism in this process is still
poorly understood.
Cell division cycle 42 (Cdc42), a member of the Rho
GTPases family, contributes to cell migration by con-
trolling protrusions, adhesion, and contraction [1417].
Cdc42 is crucial for the regulation of stem cells’ behav-
iors. Loss of Cdc42 reduces α-SMA expression in mesen-
chymal stem cells [18]. e elevated activity of the Cdc42
in aged hematopoietic stem cells (HSCs) correlates with a
loss of polarity [19]. In a recent study, we have found that
Cdc42 influences the morphology and skeleton in bone
marrow-derived mesenchymal stem cells (BMSCs) [20].
However, the role of Cdc42 in ADSCs-mediated wound
healing remains unclear.
In this study, we used the dominant-negative mutant of
Cdc42 (Cdc42N17) to explore the role of Cdc42 in activin
B-induced ADSCs migration, proliferation, and secretion
in vitro. Cdc42N17-transfected ADSCs were injected
into a full-thickness excisional wound model to explore
their efficiency in wound healing invivo. Moreover, RNA
sequencing was used to explore the downstream mecha-
nism of Cdc42 in the regulation of wound healing pro-
cesses. Our study reveals an essential role of Cdc42 in
ADSCs-mediated wound healing.
Material andmethods
Animals
One hundred and eight specific pathogen-free (SPF)
class C57BL/6 male mice (26–28g) of 3months old were
purchased from Laboratory Animal Center of Southern
Medical University (Guangzhou, China; SCXK 2021-
0041). e mice were housed in a temperature-controlled
room (23 ± 2°C) under a 12-h light/dark cycle with avail-
able food and water adlibitum. e mice were allowed
1 week to adapt to the environment before experiments.
All the procedures related to the care and use of animals
in this study were in accordance with the guidelines of
National Institutes of Health (NIH) and was approved by
the Bioethics Committee of Southern Medical University
(Approval number: L2019084).
Isolation, identication, andlabeling ofADSCs
ADSCs’ isolation was performed as previously reported
[21, 22]. Mice were killed by cervical dislocation and
asepticized by soaking in 75% ethanol for 5 min. e
inguinal adipose tissues were withdrawn and washed
with PBS at 4 °C for three times. After removing the
blood vessels and lymph nodes carefully, the adipose
was cut into pieces and digested for 90 min at 37 °C
using 10% fetal bovine serum (FBS) and 125 U/mL col-
lagenase (Cat# 17018029, Collagenase Type I, Gibco)
[23]. By filtrating through 100-μm nylon filter mesh (Cat#
Graphical Abstract
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
352360, BD Falcon) and concentrated by centrifugation
at 300g for 5min, the stromal vascular fraction (SVF) was
attained [24]. en, ADSCs were resuspended in com-
plete medium (high-glucose Dulbecco’s modified Eagle’s
medium (H-DMEM) with 10% FBS and 1% penicillin-
streptomycin solution (Cat# C0222, Beyotime, China))
and cultured in a humid atmosphere with 5% CO2 at
37°C. e culture medium was updated every 48h. e
cells were monitored daily under an inverted phase-con-
trast microscope (Leica DMI4000 B) and passaged when
they reached 80–90% confluence.
Cells at passage three were used for 5,6-carboxyfluo-
rescein diacetate succinimidyl ester (CFSE) (Cat# C1157,
Invitrogen) labeling and identification according to the
standard listed in Additional file1.
Lentivirus infection
e dominant negative mutant Cdc42N17, the constitu-
tively active mutant Cdc42L61, and the EGFP lentiviral
vector were generated as described in our previous study
[20, 22]. e titers of Cdc42N17, Cdc42L61, and EGFP
vector were 1.63 × 109 TU/ml, 6.93 × 108 TU/ml, and
4.53 × 108TU/ml, respectively. e lentivirus was serum-
free incubated with ADSCs for 8 h. On the day 3 after
lentivirus infection, the infection efficiency was deter-
mined by examining green fluorescence in the cells using
fluorescent microscopy and pull-down assay.
The inhibitor treatment
Cells were serum-starved for 12h and then treated with
serum-free H-DMEM containing Srf inhibitor CCG-
100602 (15μM, Cat# HY-120855, Mce) for 24h [25, 26]
or Erk1/2 inhibitor SCH772984 (5μM, Cat# S7101, Sell-
eck) for 2h [27].
GST pull‑down assay
GTPase pull-down assay was performed according to
the manufacturer’s protocol (1:100, Cat # 14-325, Rac/
cdc42 Assay Reagent, Millipore) as described previously
[20, 28]. In short, after lysed with MLB buffer and deter-
mined the total protein concentration, the cell lysates
were divided into two equal parts: One was blotted for
total Cdc42, and the other was subjected to a PAK1 PBD
binding assay. e beads with Cdc42 GTP-bound were
captured, and the activation levels of Cdc42 were ana-
lyzed by western blot using an anti-Cdc42 antibody. e
detailed protocol for pull-down assay is described in
Additional file1.
Western blot analysis
Cells were lysed using radio-immunoprecipitation assay
(RIPA) buffer (Cat# P0013B, Beyotime, China), and
total protein was extracted and normalized according to
the concentrations determined by Enhanced BCA Pro-
tein Assay Kit (Cat# P0010, Beyotime, China). e total
protein was then re-suspended with 5 × loading buffer
(Ca# P0286, Beyotime, China) and boiled for 5 min.
Twenty-five mg protein from each sample was separated
by electrophoresis on 10% polyacrylamide gels (Cat#
PG112, Epizyme, China) and transferred onto polyvi-
nylidene difluoride membranes. e blots were blocked
with 5% skim milk (Cat# 232100, Genebase, China) and
incubated with a primary antibody Cdc42 (1:2000, Cat#
ab187643, Abcam), Srf (1:1000, Cat# 16821-1-AP, Pro-
teintech, China), or phosphorylation Erk (p-Erk) (1:1000,
Cat# 4370S, Cell Signaling) at 4°C overnight. After fur-
ther incubated with HRP-conjugated anti-rabbit IgG
(1:2000, Cat# ab6721, Abcam) secondary antibody for
1h at room temperature, the bands’ intensity was visual-
ized using the enhanced chemiluminescence (ECL) lumi-
nescence reagent (Cat# MA0186-1, Meilunbio, China)
and Gel Image System (Tanon-5200CE). e blots were
then washed with a commercial stripping buffer (Cat#
P0025N, Beyotime, China) for 10min at room temper-
ature and re-probed with antibodies against total Erk
(1:1000, Cat# 4695S, Cell Signaling). en, the intensity
of the band was quantified using ECL and Gel Image Sys-
tem (Tanon-5200CE).
Migration assay
ADSCs and cells transduced with Cdc42N17, Cdc42L61,
or EGFP vector were seeded into 24-well plastic plates
at the density of 1 × 105 cells per well and cultured until
they reached a confluence of 90%. After serum-starved
for 12h, the cells were divided into seven groups. Among
them, four groups (ADSCs, Cdc42N17, Cdc42L61, and
EGFP vector) were treated with serum-free H-DMEM,
while the other three groups (activin B+ADSCs, activin
B+Cdc42N17, and activin B+Cdc42L61) were treated
with H-DMEM containing 10ng/ml activin B. en, the
migration properties of cells were analyzed by the follow-
ing two methods.
For the scratch wound healing assay, the uniform
scratch wounds were scraped and photographed at 0, 24,
and 48h after being washed with PBS. e Image-Pro
Plus 6.0 software was used to calculate the scratching
area.
For the transwell assay, 500 µl preheated serum-free
H-DMEM was added to the upper and lower chamber,
respectively, and incubated for 1h at 37 °C. After dis-
carding the medium, a total of 200μl serum-free media
including 5 × 104 cells has been added into the upper
chamber, while the lower chamber was filled with 300µl
different culture media (H-DMEM or H-DMEM contain-
ing 10ng/ml activin B) as designed previously. After 24-h
incubation, the migrated cells under the chamber were
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
fixed with 4% paraformaldehyde solution (PFA) (Cat#
G0528, GBCBIO, China) for 15min, stained with crystal
violet staining solution (Cat# C0121, Beyotime, China)
and imagined by microscope.
Proliferation assay
For the cell proliferation experiment, ADSCs and cells
transduced with Cdc42N17, Cdc42L61, or the mock
vector were seeded on cover glass into 24-well plas-
tic plates at the density of 1 × 105cells per well. After
serum-starved for 12h, the cells were divided into eleven
groups. Among them, six groups (ADSCs, Cdc42N17,
Cdc42L61, CCG-100602, SCH772984, and EGFP vector)
were treated with serum-free H-DMEM, while the other
five groups (activin B+ADSCs, activin B+Cdc42N17,
activin B+Cdc42L61, activin B+CCG-100602, and
activin B+SCH772984) were treated with H-DMEM con-
taining 10ng/ml activin B. en, an addition of 12.5μM
EdU was added in each group. After incubating for 6h
at 37°C, cells were fixing, Apollo- and nuclear-stained
as manufacturer’s instructions (Cat# C10310-1, RiboBio,
China). e EdU-positive cells were observed under fluo-
rescence microscope.
Cytokine analysis
e cells of ADSCs and cells transduced with Cdc42N17
or Cdc42L61 have been selected for a 12-h serum-
free starve. After incubating for 24 h with H-DMEM
(for ADSCs, Cdc42N17, and Cdc42L61) or H-DMEM
with 10ng/ml activin B (for activin B+ADSCs, activin
B+Cdc42N17, and activin B+Cdc42L61), the cell cul-
ture supernatant was collected by centrifuging at 3000g
for 15min and filtering with a 0.22-μm filter to remove
apoptotic cells and debris. en, the secretion levels of
Col 1 (Cat# MU30364, Bisowamp, China) and VEGF
(Cat# MU30236, Bisowamp, China) were detected using
a commercially available enzyme-linked immunosorbent
assay (ELISA) system following the kit’s instructions.
Wound healing model
e wound healing model was established as we
described previously [13]. Mice were anesthetized by
injecting 2% pentobarbital sodium (1.5mL/kg) into their
intraperitoneal. After the dorsal hair was depilated using
a honey and wax mixture and cleaned with 70% ethanol,
skin punch was used to delineate an 8-mm round-shaped
image on the skin. en, sterile scissors were applied to
create full-thickness skin wounds on both the right and
left shaved dorsal skin after povidone iodine disinfection.
Each mouse had retained the capability to free ingest
food and water after the operation.
After wounding, mice were randomly divided into six
groups (n = 6 for each group) according to the treatment
on the site surrounding the wound (Table1) daily for 3
days. e cells and/or activin B were resuspended in PBS
and administered to the wounds by subcutaneous injec-
tion. Wound areas were measured photographically at
days 0, 3, 7, and 14 after surgery, and the rate of wound
closure was calculated with the following equation:
wound closure rate (%) = [(original wound area open
area on final day)/original wound area] × 100%.
Frozen sections
For analysis of the applied ADSCs within the wound site,
the wound and surrounding tissue were collected on day
3 and frozen with optimal cutting temperature (OCT)
compound. Serial sections(15μm) were taken from the
wound center to the edge by a freezing microtome at
20°C and adhered to the slides. After washing with
PBS and blocking in PBS containing 5% bovine serum
albumin (BSA) (Cat# ST023, Beyotime, China), the sec-
tions were stained with Hoechst 33258 (1:500, Cat#
H21491, Invitrogen) for 10min and examined by fluores-
cence microscopy.
Hematoxylin andeosin (H&E) staining andhistological
evaluation
Mice were killed at postoperative 3rd, 7th, and 14th day,
respectively. en, complete wound with 0.5-cm mar-
gin was carefully cut down and rinsed with PBS before
and after fixing by 4% paraformaldehyde solution (PFA)
(Cat# G0528, GBCBIO, China) for 3 days. en, the sam-
ples were dehydrated and embedded in paraffin. Serial
Table 1 Group on wound healing model
Group Name Treatment
1 Control 0.4 ml PBS
2Act B 0.4 ml 10 ng/ml activin B
3 ADSCs 0.4 ml 6 × 106/ml ADSCs
4Act B+ADSCs 0.4 ml 6 × 106/ml ADSCs induced by 10 ng/ml activin B for 12 h
5 ADSCs (Cdc42N17) 0.4 ml 6 × 106/ml ADSCs transduced with Cdc42N17
6Act B+ADSCs (Cdc42N17) 0.4 ml 6 × 106/ml ADSCs transduced with Cdc42N17 and
induced by 10 ng/ml activin B for 12 h
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
sections (5µm) were collected and H&E-stained accord-
ing to the detailed method described in Additional file1.
For histological evaluation, re-epithelialization and
granulation tissue formation was evaluating by using
H&E staining. e length of epithelial tongues of wound
skin sections on each group sample (n = 6) was meas-
ured between the wound edge and the leading edge of
migrating epithelial tongue with ImageJ 1.52a software
(National Institutes of Health, Bethesda, MD, USA).
For the granulation tissue formation, each group sam-
ple (n = 6) was given a granulation tissue score rang-
ing from 1 to 12. e criteria used for granulation
tissue scores of wound healing were referred to previ-
ous researches [2932] and our previous study [33] and
summarized in Additional file1: TableS1.
Masson’s trichrome staining
e slices adjacent to the wound center were used for
Masson’s trichrome staining using Masson’s trichrome
staining kit (Cat# MST-8004, Maixin-Bio, China) accord-
ing to the manufacturer’s instructions.
Immunohistochemical assay
e tissue slices were deparaffinized and rehydrated
(detailed procedure as previously described in H&E
staining). In order to retrieve antigen, samples were first
immersed in 0.1M citrate buffer at 96°C for 10min and
later incubated in 5% BSA for 2h. e slices were incu-
bated with antibody against CD31 (1:100, Cat# ab281583,
Abcam) and α-SMA (1:200, Cat# ab32575, Abcam) at
4 °C overnight. After rinsing with PBS and incubated
with biotinylated goat anti-rabbit secondary antibody
(Cat# PV-6001, ZSGB-BIO, China) for 2 h, the tissue
slices were colored with 3,3-diaminobenzidine (DAB)
(Cat# AR1022, Boster, China), stained with hematoxylin,
dehydrated with a gradient ethanol series, soaked with
xylene, and then sealed with resin. Finally, five random
locations of each tissue slice were selected to count the
new capillaries with endothelial cells positive for CD31
via microscope (400 ×) or to analyze the average opti-
cal density values for α-SMA expression at the indicated
time points using Image-Pro Plus 6.0 Software.
Total RNA preparation
ADSCs transduced with or without Cdc42N17
were seeded in 60-mm plastic culture plates. After
12-h serum-free starve, they were divided into four
groups according to different processing methods
with H-DMEM (ADSCs, Cdc42N17) or H-DMEM
with 10 ng/ml activin B (Act B+ADSCs and Act
B+Cdc42N17) and incubated for another 24h. Total
RNA of those cells was extracted, and genomic DNA
was simultaneously eliminated using EZ-10 DNAaway
RNA Mini-Preps Kit (Cat# B618133, Sangon Biotech,
China) according to the manufacturer’s instructions.
e quality and purity of the resulting RNA were
checked using the NanoDrop spectrophotometer;
only high-quality RNA sample (OD260/280 = 1.8–2.2,
OD260/230 2.0) could be used for sequencing and
qPCR analysis.
RNA sequencing
For the transcriptomic analysis, 1 μg total RNA was
used to prepare RNA-seq transcriptome library using
TruSeq RNA sample preparation kit from Illu-
mina (San Diego, CA). e raw paired-end reads were
trimmed, and the quality was controlled by SeqPrep
and Sickle with default parameters. e high-quality
clean data were aligned to the mouse genome using
HISAT2 software [34], and mapped data (reads) of each
sample were assembled by StringTie in a reference-
based approach [35].
To identify differential expression genes (DEGs)
between these samples, the expression levels of each
transcript and gene abundance were calculated according
to the transcripts per million reads (TPM) method and
RSEM, respectively [36]. Essentially, differential expres-
sion analysis was performed using the DESeq2 [37]/
EdgeR [38] with Q value 0.05; DEGs with |log2FC|> 1
and Q value 0.05 (DESeq2 or EdgeR)/Q value 0.001
(DEGseq) were considered to be significantly differen-
tially expressed genes. In addition, functional enrichment
analysis including GO and KEGG was performed to
identify which DEGs were significantly enriched in GO
terms and metabolic pathways at Bonferroni-corrected
P value 0.05 compared with the whole-transcriptome
(See figure on next page.)
Fig. 1 Cdc42 regulates activin B-mediated ADSCs migration in vitro. A Pull-down assays were performed to detect the amounts of GTP-bound
Cdc42 in ADSCs, ADSCs treated with activin B, respectively. B The ratios of GTP-Cdc42 versus total Cdc42 levels were analyzed. C Scratch wound
healing assays were performed to detect the migration of cells treated by seven methods, and photographs were taken at 0, 24, and 48 h after
scratch injury. Scale bar = 500 μm. D The healing rates were quantified by measuring the area of the injured region. E Transwell assays were
performed to detect the invasion of these cells, and photographs were taken at 24 h after seeds. Scale bar = 200 μm. F The migrated cells were
counted. All values are expressed as mean ± SD from three independent repeats. *P < 0.05
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Fig. 1 (See legend on previous page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
background. GO functional enrichment and KEGG path-
way analysis were carried out by Goatools and KOBAS
[39].
qPCR analysis
To further verify RNA-seq data, the genes described in
Table S2 with significant expression differences were
analyzed by qPCR using the Maxima SYBR Green/ROX
qPCR Master Mix kit (Cat# K0221, ermo Scientific)
according to the manufacturer’s instructions. To be spe-
cific, 1.0 μg total RNA was converted to cDNA using
MonScript RTIII All-in-One Mix with dsDNase kit
(Cat# MR05101S, Monad Biotech, China), and the qPCR
reactions were performed on the ABI 7900 system using
the following program: 95°C for 10min, and 40 cycles of
95°C for 10s, 60°C for 10s, and 72°C for 30s, followed
by dissociation steps. Reactions were run in triplicates.
e GAPDH housekeeping gene was used as an internal
control for data normalization across samples. e nor-
malization of Ct values for each gene and the determina-
tion of fold changes in gene expression (normalized to
control group) were calculated using the 2ΔΔCt method.
Statistical analysis
Numerical data were expressed as the means ± stand-
ard deviation (SD). All experiments were performed
at least three times. Statistical differences between the
groups were assessed using one-way analysis of vari-
ance (ANOVA) in SPSS20.0 software (IBM, Armonk, NY,
USA). A p value of less than 0.05 (*P < 0.05) was consid-
ered to be statistically significant.
Fig. 2 Cdc42 regulates activin B-mediated ADSCs’ proliferation and secretion in vitro. A Representative fluorescence imaging of EdU staining of
seven groups cells at 6 h. Scale bar = 50 μm. B The proliferation rates were quantified by calculating the percentage of EdU-positive cells. C Average
concentration of Col 1 and VEGF in the conditioned cell medium of six groups. All values are expressed as mean ± SD from three independent
repeats. *P < 0.05
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Results
Characterization ofisolated ADSCs
To validate ADSCs isolated from mice, we assessed cell
surface markers using immunocytochemistry. In support
of previous reports [13, 40], ADSCs were positive for
CD44 and CD90, while nearly negative for CD31 and
CD80 (Additional file1: Fig. S1A). e flow cytometry
was also assessed to characterize isolated ADSCs cell
Fig. 3 Cdc42-regulated activin B-induced ADSCs-mediated skin wound healing in vivo. A Representative macroscopic images of wounds treated
with PBS, activin B, ADSCs, activin B+ADSCs, ADSCs (Cdc42N17), or activin B+ADSCs (Cdc42N17) on days 0, 3, 7, and 14. B Quantitative analysis of
wound closure rate of six mice per group. All values are expressed as mean ± SD from six independent repeats. *P < 0.05
Fig. 4 Cdc42-regulated activin B-induced ADSCs-mediated re-epithelialization and granulation tissue formation in skin wound healing. A H&E
staining showed the re-epithelialization and granulation tissue formation of the six groups after wounding for 3, 7, and 14 days. Black and green
arrows represent the dermal border and epidermal margin (scale bar = 500 μm), whereas insets of main figures (1) and (2) represent wound
re-epithelialization and granulation tissue (scale bar = 100 μm), respectively. B The percentage of re-epithelialization ratio of six experiments. C
Histological scores of granulation tissue thickness. All values are expressed as mean ± SD from three independent repeats. *P < 0.05
(See figure on next page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Fig. 4 (See legend on previous page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
surface markers. ADSCs were positive for CD29, CD44,
CD90.2 and nearly negative for CD31, CD45, and CD117
(Additional file1: Fig. S1B). ADSCs were further char-
acterized by confirming their ability to undergo specific
osteogenic, adipogenic, and chondrogenic differentiation.
ese cells were positive for Alizarin Red staining (Addi-
tional file1: Fig. S1C), oil red O staining (Additional file1:
Fig. S1D), and Alcian Blue staining (Additional file1: Fig.
S1E), indicating osteogenic, adipogenic, and chondro-
genic respective cell-type differentiation. Only cells that
met these criteria were used in subsequent experiments.
Cdc42 regulates activin B‑induced ADSCs migration
We then investigated whether activin B regulated the
activity of Cdc42 in ADSCs by GST pull-down assay. We
found that the GTP-bound Cdc42 was increased from 30
to 120min after activin B treatment (Fig.1A, B).
To study the role of Cdc42 in activin B-induced
ADSCs migration and other biological functions, the
mock vector or the dominant-negative mutant of Cdc42
(Cdc42N17) and the constitutively active mutant of
Cdc42 (Cdc42L61) were transduced into ADSCs, respec-
tively. e efficient transduction of Cdc42N17 and
Cdc42L61 was confirmed by GFP imaging and pull-down
assay (Additional file1: Fig. S2).
We next examined whether Cdc42 was involved
in activin B-induced ADSCs migration. In a scratch-
ing assay, as reported previously [13], 10ng/ml activin
B effectively induced ADSCs migration (Fig. 1C, D).
However, Cdc42N17 inhibited ADSCs migration with
or without activin B after 24 and 48h (Fig.1C, D). e
wound healing rate of activin B-stimulated Cdc42N17-
transduced ADSCs was comparable to that of Cdc42N17-
transduced ADSCs without activin B stimulation
(Fig.1C, D). ese findings suggest that Cdc42N17 abol-
ished activin B-induced ADSCs migration. Consistent
with this finding, Cdc42N17 inhibited activin B-induced
ADSCs migration to the lower chamber in a transwell
assay (Fig.1E, F). However, Cdc42L61 did not promote
ADSCs migration with or without activin B (Fig.1C–F).
In summary, these results suggest that Cdc42 is nec-
essary but not sufficient for activin B-induced ADSCs
migration.
Cdc42 regulates activin B‑induced ADSCs proliferation
andsecretion
Next, we investigated the role of Cdc42 in the prolif-
eration and secretion of ADSCs treated with or without
activin B.
EdU-stained cell number was markedly increased in
the activin B group compared with the control group.
Cdc42N17 significantly decreased EdU-stained cell num-
ber in the ADSCs treated with or without activin B. Of
note, EdU-stained cell number in activin B-stimulated
Cdc42N17-transduced ADSCs was comparable to that in
Cdc42N17-transduced ADSCs without activin B stimula-
tion (Fig.2A, B). ese findings reveal that Cdc42 may
regulate ADSCs’ proliferation induced by activin B.
Moreover, the concentrations of Col 1 and VEGF were
markedly increased in the activin B group compared with
that in the control group, whereas Cdc42N17 signifi-
cantly inhibited the secretion of Col 1 and VEGF in the
ADSCs with or without activin B. us, Cdc42 contrib-
utes to ADSCs’ secretion induced by activin B (Fig.2C).
However, Cdc42L61 did not promote ADSCs’ prolifera-
tion and secretion with/without activin B (Fig. 2A-C).
Taken together, these findings suggest that Cdc42 is nec-
essary but not sufficient for ADSCs’ proliferation and
secretion induced by activin B.
Cdc42 promotes activin B‑induced ADSCs‑mediated skin
wound healing invivo
We then examined the role of Cdc42 on activin
B-induced ADSCs-mediated cutaneous wound healing
invivo.
To assess ADSCs administered, 5, 6-carboxyfluores-
cein diacetate succinimidyl ester (CFSE) was used to
label ADSCs before transplantation (Additional file 1:
Fig. S3A) and then we collected the frozen sections on
day 3 after ADSCs transplantation. Green fluorescence
(EGFP+) cells were detected in the ADSCs group, activin
B+ADSCs group, ADSCs (Cdc42N17) group, and activin
B+ADSCs (Cdc42N17) group but not in the activin B
group or control group (Additional file1: Fig. S3B). ese
data suggest that ADSCs were incorporated into wound
sites on day 3 after transplantation.
Wound closure rates were then evaluated. Consistently
with the reported study [13], the wound closure rates
(See figure on next page.)
Fig. 5 Cdc42 regulates activin B-induced ADSCs-mediated neovascularization and wound contraction in vivo. A Representative photomicrographs
of CD 31 immunohistochemical staining treated with PBS, activin B, ADSCs, activin B+ADSCs, ADSCs (Cdc42N17), and activin B+ADSCs (Cdc42N17),
respectively, at the specified time. The black arrow indicates positive staining of CD31. Scale bars = 200 μm. B Representative photomicrographs of
α-SMA immunohistochemical staining of wounds per group on days 3 and 7 after wounding. Scale bars = 200 μm. The areas stained with α-SMA
were determined by planimetric image analysis using Image-Pro Plus 6.0 software. All values are expressed as mean ± SD from three independent
repeats, *P < 0.05
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Fig. 5 (See legend on previous page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
in the activin B+ADSCs group were significantly accel-
erated compared with that of the control, activin B, or
ADSCs group (Fig.3A, B). However, the wound closure
rate of ADSCs (Cdc42N17) was significantly decreased
compared with that of the ADSCs group on days 3 and
7 after treatment (Fig. 3A, B). In addition, the wound
healing rate in the activin B+ADSCs (Cdc42N17) group
was also decreased compared with that of the activin
B+ADSCs group (Fig. 3A, B). As there was no differ-
ence in wound healing rate between ADSCs (Cdc42N17)
group and activin B+ADSCs (Cdc42N17) group, these
data suggest that Cdc42N17 inhibited wound healing rate
induced by activin B and ADSCs invivo.
Cdc42 promotes activin B‑induced re‑epithelialization,
granulation tissue formation, andcollagen deposition
inskin wound healing
e results of H&E staining revealed that re-epithe-
lialization in ADSCs (Cdc42N17) group was impaired
compared with that of ADSCs groups on day 3 and 7
with or without activin B (Fig.4A, B). Moreover, granu-
lation tissue scores of ADSCs (Cdc42N17) and activin
B+ADSCs (Cdc42N17) groups were much lower than
those of the ADSCs and activin B+ADSCs groups,
respectively. Re-epithelialization and granulation tissue
scores in ADSCs (Cdc42N17) group were similar to that
in activin B+ADSCs (Cdc42N17) group(Fig.4A–C).
e Masson trichrome staining experiment was used to
investigate the collagen deposition on day 7 and day 14
after treatment. After 7days of treatment, there was lit-
tle collagen deposition in the control group. In contrast,
more newly formed collagen appeared in the ADSCs and
activin B+ADSCs groups. However, there was still a large
area of tissue without collagen deposition in the ADSCs
(Cdc42N17) and activin B+ADSCs (Cdc42N17) groups.
After 14days of treatment, the collagen deposition pat-
tern in the activin B+ADSCs group was similar to that
in normal skin at the wound gap. However, the collagen
deposition pattern in the ADSCs (Cdc42N17) group was
similar to that in activin B+ADSCs (Cdc42N17) group,
which was significantly less mature than that in the
ADSCs group (Additional file1: Fig. S4).
ese data reveal that Cdc42 regulates activin
B-induced re-epithelialization, granulation tissue
formation, and collagen deposition or maturation after
wounding.
Cdc42 regulates theneovascularization andwound
contraction inactivin B‑induced ADSCs‑mediated skin
wound healing
Immunohistochemistry staining of CD31 and α-SMA
was used to evaluate the neovascularization and wound
contraction. We found that the expression of CD31 and
α-SMA in the wounds treated with ADSCs (Cdc42N17)
was decreased compared with that of ADSCs group on
days 7 and 14 (Fig.5A, B). Similarly, less expression of
CD31 and α-SMA in activin B+ADSCs (Cdc42N17)
group was observed compared with that of activin
B+ADSCs group (Fig.5A, B). Cdc42N17 inhibited CD31
and α-SMA expression in activin B+ADSCs group to the
levels comparable to that in ADSCs (Cdc42N17) group
(Fig. 5A, B). ese data suggest that Cdc42 promotes
the neovascularization and wound contraction in activin
B-induced ADSCs-mediated skin wound healing.
RNA sequencing identies possible mechanism ofCdc42
intheregulation ofADSCs biological function
RNA sequencing was further conducted in ADSCs group
and ADSCs (Cdc42N17) group to explore global gene
expression changes induced by Cdc42N17. By principal
component analysis (PCA), we found that there was a
strong correlation between biological duplicate samples
(Additional file1: Fig. S5). Compared with ADSCs group,
a total of 216 upregulated and 266 downregulated DEGs
were detected in ADSCs (Cdc42N17) (Fig.6A).
e KEGG pathway enrichment analysis found that
compared with ADSCs group, the upregulated DEGs
in ADSCs (Cdc42N17) group were enriched in micro-
RNAs in cancer, pathways in cancer, wnt, cytokine–
cytokine receptor interaction, cellular senescence, and
foxo (Fig. 6B). e downregulated DEGs in ADSCs
(Cdc42N17) were enriched in the pathways of cell cycle,
p53, regulating pluripotency of stem cells, focal adhesion,
TGF-β, and gap junction pathways (Fig.6C).
e GO enrichment analysis revealed that compared
with ADSCs group, the upregulated DEGs in ADSCs
(Cdc42N17) group were significantly associated with cell
differentiation, negative regulation of cell proliferation,
Fig. 6 Cdc42 deletion in ADSCs exhibits a distinct transcriptional signature. A Heatmap showing differentially expressed genes (DEGs) between
ADSCs (Con) and ADSCs transduced with Cdc42N17. There were 216 upregulated and 266 downregulated DEGs in ADSCs (Cdc42N17), respectively,
and were subjected to Kyoto Encyclopedia of Genes and Genomes pathways enriched (KEGG) and gene ontology (GO) analysis. B, C showing
the top 15 enrich pathways in upregulated and downregulated DEGs, while D showing the top 15 biological processes in upregulated and
downregulated DEGs. E Selected gene expression in some of the GO biological processes. F qPCR analysis of relative expression level of Cdkn2b,
Wnt11, Pdgfb, Iqgap3, Racgap1 and Cd248 in ADSCs (Cdc42N17). All values are expressed as mean ± SD from three independent repeats. *P < 0.05
(See figure on next page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Fig. 6 (See legend on previous page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 14 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
transcription and cell migration, positive regulation of
apoptotic process, and intracellular signal transduction
(Fig.6D). On the other hand, the downregulated DEGs
were associated with cell cycle, cell division, mitotic
nuclear division, cell adhesion, and cell proliferation
(Fig.6D).
We listed the upregulated and downregulated genes in
some of the GO enrichment terms (Fig.6E). e expres-
sion levels of Cdkn2b, Wnt11, Pdgfb, Iqgap3, Racgap1,
and Cd248 were verified by qPCR, and the results were
consistent with the RNA-seq (Fig.6F).
Taken together, these data suggest that Cdc42 plays a
role in cell proliferation, migration, adhesion, and cell
cycle of ADSCs. Furthermore, the signaling in wnt, foxo,
cell cycle, p53, and TGF-β pathway may contribute to
Cdc42-regulated biological function of ADSCs.
RNA sequencing identies thepossible mechanism
ofCdc42 intheregulation ofactivin B‑induced ADSCs
biological function
To further identify the mechanism of Cdc42 in the regu-
lation of activin B-induced ADSCs biological function,
RNA sequencing was performed in activin B+ADSCs
group and activin B+ADSCs (Cdc42N17) group. ere
was obvious difference between the two groups (Addi-
tional file 1: Fig. S6). Activin B+ADSCs (Cdc42N17)
group showed 355 upregulated DEGs and 615 downregu-
lated DEGs (Fig.7A).
KEGG pathway enrichment analysis revealed that
the upregulated DEGs were enriched in the pathway of
metabolism of xenobiotics by cytochrome P450, drug
metabolism—cytochrome P450, glutathione metabo-
lism, cytokine–cytokine receptor interaction, and pro-
teoglycans in cancer (Fig.7B). e downregulated DEGs
were enriched in the pathway of ECM–receptor interac-
tion, focal adhesion, Rap1, MAPK, TGF-β, and PI3K-Akt
(Fig.7C).
In the GO annotations analysis, the upregulated DEGs
were enriched in the biological processes of oxidation–
reduction process, transport, negative regulation of cell
proliferation, and aging (Fig. 7D). e downregulated
DEGs were enriched in the biological processes of cell
adhesion, signal transduction, positive regulation of cell
proliferation and migration, cell cycle, and angiogenesis
(Fig.7D). e detailed selected gene expression changes
in the biological processes of apoptosis, negative regula-
tion of proliferation, aging, migration, proliferation, and
differentiation are shown in Fig.7E.
Moreover, the changes in selected gene expression such
as Bmp6, Nkd1, Cd248, Id4, and Sox11 were confirmed
by qPCR (Fig.7F).
Taking together, these data imply that Cdc42 plays a
role in activin B-induced signal transduction, cell adhe-
sion, cell migration, and cell proliferation of ADSCs. Fur-
thermore, the signaling pathway of PI3K-Akt, pathway
in cancer, focal adhesion, Rap1, MAPK, and TGF-β may
contribute to Cdc42’s regulation of activin B-induced
biological function of ADSCs.
The Erk‑Srf pathway isinvolved inCdc42‑mediated activin
B‑induced ADSCs proliferation
Among the differentially expressed genes in the activin
B+ADSCs and activin B+ADSCs (Cdc42N17) group,
several members of the MAPK signaling pathways were
downregulated in activin B+ADSCs (Cdc42N17) group
(Fig. 8A). We confirmed the downregulation of Srf in
activin B+ADSCs (Cdc42N17) group by qPCR and west-
ern blot (Fig.8B, C). For Srf is a major downstream cyto-
solic transcription factor of Erk/MAPK signaling [41],
the effect of Cdc42 on Erk/MAPK activity and Srf expres-
sion was further explored. e result of qPCR and west-
ern blot confirmed that while Erk and its downstream Srf
were increased upon activin B treatment, this increase
was abolished by inhibition of Cdc42 (Fig.8B–F). Further
study of the mechanism revealed that the activation of
Erk signal in ADSCs by activin B was able to induce pro-
liferation. However, Erk inhibitor SCH772984 abolished
the growth of ADSCs with activin B (Fig. 8G, H). e
inhibition of Srf by CCG-100602 also suppressed ADSCs
proliferation induced by activin B (Fig.8G, H). ese data
suggest that Erk-Srf pathway is involved in Cdc42-medi-
ated activin B-induced ADSCs proliferation.
Discussion
We have previously found that activin B is able to induce
migration of ADSCs to promote skin wound healing
[13]. However, whether Cdc42 signaling is important in
ADSCs remained unknown. e current study found
that Cdc42 signaling is essential for ADSCs-mediated
(See figure on next page.)
Fig. 7 Cdc42 regulates activin B-induced ADSCs transcriptional signature. A Heatmap showing differentially expressed genes (DEGs) between
activin B+ADSCs and activin B+ADSCs (Cdc42N17). There were 355 upregulated (Red) and 615 (Blue) downregulated DEGs in activin B+ADSCs
(Cdc42N17), respectively, and were subjected to Kyoto Encyclopedia of Genes and Genomes pathways (KEGG) enriched and gene ontology (GO)
functional enrichment analysis. B and C showing the genes top 15 enrich pathway, while D showing the top 15 GO terms in upregulated and
downregulated DEGs. E Selected gene expression in some of the GO terms. F qPCR analysis of relative expression level of Bmp6, Nkd1, Cd248, Id4,
and Sox11 in Act B+ADSCs (Cdc42N17). All values are expressed as mean ± SD from three independent repeats. *P < 0.05
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 15 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Fig. 7 (See legend on previous page.)
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 16 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Fig. 8 Activin B-induced ADSCs proliferation by activating Cdc42-Erk-Srf pathway. A Genes expression related to MAPK signaling in Act B+ADSCs
(Cdc42N17) group. B qPCR analysis was performed to confirm the relative mRNA expression level of Srf in four groups. C, E Western blot assays
were performed to detect the level of Srf and phosphorylation Erk (p-Erk) in ADSCs, activin B+ADSCs, ADSCs (SCH772984) (Erk1/2 inhibitor) or
ADSCs (Cdc42N17) treated with or without activin B, respectively. D, F The band and the relative quantification levels of the expression of Srf
and phosphorylation Erk were analyzed. G Representative fluorescence imaging of EdU staining of ADSCs and ADSCs treated with Srf inhibitor
CCG-100602 or Erk1/2 inhibitor SCH772984 and with/without activin B at 6 h. Scale bar = 50 μm. H The proliferation rates were quantified by
calculating the percentage of EdU-positive cells. All values are expressed as mean ± SD from three independent repeats. *P < 0.05
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 17 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
cutaneous wound healing as well as proliferation and
secretion. While our previous study found that Cdc42
regulates BMSCs-mediated wound healing by affecting
Golgi reorientation [20], this study found that Cdc42
regulates ADSCs-mediated cutaneous wound healing by
affecting VEGF-mediated vascularization, myofibroblast
differentiation-mediated granulation tissue formation,
and Erk-Srf signaling pathway.
Cutaneous wound healing is a dynamic process that
involves overlapping phases, including inflammation,
granulation tissue formation, and matrix remodeling [42].
ADSCs have been shown to enhance wound healing via
their paracrine function [43]. e present study suggests
that Cdc42 signaling is partially responsible for activin
B-induced secretion of VEGF in ADSCs. VEGF stimulates
wound healing through angiogenesis [44]. Consistently,
we have found that the ADSCs-mediated vascularization
in cutaneous wound healing is suppressed upon Cdc42
suppression, as the expression of CD31 is decreased in
ADSCs (Cdc42N17) group induced by activin B. ese
data suggest that Cdc42 regulates activin B-induced
ADSCs-mediated vascularization in cutaneous wound
healing via regulation of VEGF secretion.
Myofibroblast, mostly characterized by high expression
of α-smooth muscle actin (α-SMA), is the major cell type
responsible for ECM production during wound healing
[45]. A recent study has shown that activin A promotes
myofibroblast differentiation of endometrial mesenchy-
mal stem cells [46]. In the present study, we found that
activin B may also promote myofibroblast differentiation
of ADSCs, as evidenced by an up-regulation of the expres-
sion of α-SMA in the wounds of activin B+ADSCs group.
In addition, we have found that the expressions of α-SMA
in the wounds treated with ADSCs (Cdc42N17) were
decreased compared with that of ADSCs group. Further-
more, our animal studies have revealed that the wound
granulation tissue formation is impaired upon Cdc42
suppression. ese data indicate that Cdc42 regulates the
ADSCs-mediated granulation tissue formation in cutane-
ous wound healing via regulation of myofibroblast differ-
entiation. In line with this, Ge’s study shows that Cdc42 is
required for TGFβ1-induced α-SMA expression in MSCs
[18]. ey have found that TGFβ1-induced expression
of α-SMA is significantly decreased in Cdc42 knockout
BMSCs after 24-h stimulation [18]. Hence, Cdc42 is nec-
essary for myofibroblast differentiation of MSCs (e.g.,
BMSCs and ADSCs).
Comprehensive understanding of the mechanism of
ADSCs in the wound healing process is of great signifi-
cance for further improving wound healing effect. Our
RNA sequencing begins to reveal potential mechanism
of Cdc42 in the regulation of activin B-induced ADSCs
biological function. e RNA sequencing results provide
evidence that Cdc42 globally controls gene expression pat-
tern of ADSCs treated with/without activin B and indicate
possible signaling pathways downstream of Cdc42 that is
associated with the regenerative ability of ADSCs. e Erk/
MAPK pathway is an essential intracellular signal trans-
duction pathway that controls cell proliferation [47, 48].
e previous study showed that exendin-4 treatment pro-
motes ADSCs growth via ERK signaling pathways [49]. In
the present study, we found that activin B contributes to
ADSCs proliferation via Cdc42-Erk-Srf pathway. Our find-
ings may help understand the comprehensive molecular
mechanisms of ADSCs in wound healing.
Conclusions
e present study shows that Cdc42 is a key regulator
of ADSCs-mediated cutaneous wound healing induced
by activin B. Firstly, Cdc42 regulates activin B-induced
migration and proliferation in ADSCs. Secondly, Cdc42
regulates ADSCs-mediated vascularization in cutaneous
wound healing via regulation of VEGF secretion. irdly,
Cdc42 regulates ADSCs-mediated granulation tissue for-
mation in cutaneous wound healing via regulation of
myofibroblast differentiation. In addition, RNA sequenc-
ing further identifies the potential mechanism of Cdc42
in the regulation of activin B-induced ADSCs biological
function. Last but not the least, activin B might activate
the Cdc42-Erk-Srf signaling pathway to promote the pro-
liferation of ADSCs in the skin wound healing process.
is study significantly advances the understanding of
the function of Cdc42 in the ADSCs-mediated cutaneous
wound healing.
Abbreviations
ADSCs: Adipose-derived mesenchymal stem cells; Act B: Activin B; Cdc42N17:
Cdc42 dominant-negative mutant; Cdc42L61: Cdc42 constitutively active
mutant; Srf: Serum response factor; Erk: Extracellular signal-regulated kinase;
MAPK: Mitogen-activated protein kinase; H-DMEM: High-glucose Dulbecco’s
modified Eagle’s medium; PBS: Phosphate-buffered saline; DAPI: 4,6-Diamid-
ino-2-phenylindole; EdU: 5-Ethynyl-2’-deoxyuridine; IHC: Immunohistochem-
istry; SDS-PAGE: Sodium dodecyl sulfate–polyacrylamide gel electrophoresis;
RNA-seq: RNA sequencing; DEGs: Differential expression genes; GO: Gene
ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; qPCR: Quantita-
tive real-time polymerase chain reaction; H&E: Hematoxylin and eosin.
Supplementary Information
The online version contains supplementary material available at https:// doi.
org/ 10. 1186/ s13287- 022- 02918-9.
Additional le1. Experimental details (characterization of ADSCs; the
detail methods for dehydration, paraffin embedding, and H&E; GST
pull-down assay and western blot); the criteria for histological evaluation
of cutaneous wound healing; the primers used in this study; characteriza-
tion of ADSCs; ADSCs transduced with lentivirus containing Cdc42N17,
Cdc42L61, and the EGFP vector; ADSCs participated in cutaneous wound
healing; Cdc42 regulates activin B-induced collagen deposition and matu-
ration after wounding; gene expression patterns analysis between ADSCs
and ADSCs (Cdc42N17); gene expression patterns analysis between
activin B+ADSCs and activin B+ADSCs (Cdc42N17).s
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 18 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
Acknowledgements
Not applicable.
Author contributions
SH and XW were responsible for the design of the study, performed the
experiments, collected and analyzed data, and wrote the manuscript. MZ,
MH, YY, YC, and CZ helped perform the main experiments and helped with
the surgical design of the animal model. YZ, JX, LB, RF, and HT participated in
cell experiments, animal surgeries, and animal care. Lu Z and Lin Z collectively
oversaw the collection of data and data interpretation and revised the manu-
script. All authors read and approved the final manuscript.
Funding
This work is supported by the National Natural Science Foundation of China
(81872514, 82073417, 81971297), Guangdong Basic and Applied Basic
Research Foundation (2020A1515011067, 2019A1515011213), Natural Science
Foundation of Guangdong Province (2018B030311062), and Program for
Changjiang Scholars and Innovative Research Team in University (IRT 16R37).
Availability of data and materials
The raw sequence data reported in this paper have been deposited in the
Genome Sequence Archive (Genomics, Proteomics & Bioinformatics 2021)
in National Genomics Data Center (Nucleic Acids Res 2022), China National
Center for Bioinformation/Beijing Institute of Genomics, and Chinese Acad-
emy of Sciences (GSA: CRA006666) that are publicly accessible at https:// ngdc.
cncb. ac. cn/ gsa. All data generated or analyzed during this study are included
in this published article and its Additional file 1.
Declarations
Ethics approval and consent to participate
All animal experiments were performed under an institutionally approved pro-
tocol for the use of animal research of Southern Medical University (Approval
number: L2019084).
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Author details
1 Depar tment of Histology and Embryology, NMPA Key Laboratory for Safety
Evaluation of Cosmetics, Key Laboratory of Construction and Detection in Tis-
sue Engineering of Guangdong Province, School of Basic Medical Sciences,
Southern Medical University, Guangzhou 510515, People’s Republic of China.
2 Key Laboratory of Functional Proteomics of Guangdong Province, Key Labo-
ratory of Mental Health of the Ministry of Education, School of Basic Medical
Sciences, Southern Medical University, Guangzhou 510515, China. 3 Depart-
ment of Orthopedics, Third Affiliated Hospital of Southern Medical Univer-
sity, Academy of Orthopedics Guangdong Province, Guangzhou 510630,
Guangdong, China.
Received: 8 October 2021 Accepted: 2 April 2022
References
1. Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi
DS. Skin tissue engineering: wound healing based on stem-cell-based
therapeutic strategies. Stem Cell Res Ther. 2019;10:111.
2. Kanji S, Das H. Advances of stem cell therapeutics in cutaneous wound
healing and regeneration. Mediators Inflamm. 2017;2017:5217967.
3. Mazini L, Rochette L, Amine M, Malka G. Regenerative capacity of adipose
derived stem cells (ADSCs), comparison with mesenchymal stem cells
(MSCs). Int J Mol Sci. 2019;20:2523.
4. Camara DAD, Shibli JA, Muller EA, De-Sa-Junior PL, Porcacchia AS, Blay
A, et al. Adipose tissue-derived stem cells: the biologic basis and future
directions for tissue engineering. Materials (Basel). 2020;13:3210.
5. Kocan B, Maziarz A, Tabarkiewicz J, Ochiya T, Banaś-Ząbczyk A. Trophic
activity and phenotype of adipose tissue-derived mesenchymal stem
cells as a background of their regenerative potential. Stem Cells Int.
2017;2017:1653254.
6. Strong AL, Neumeister MW, Levi B. Stem cells and tissue engineering:
regeneration of the skin and its contents. Clin Plast Surg. 2017;44:635–50.
7. Sheng L, Yang M, Liang Y, Li Q. Adipose tissue-derived stem cells (ADSCs)
transplantation promotes regeneration of expanded skin using a tissue
expansion model. Wound Repair Regen. 2013;21:746–54.
8. Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M.
Growth factors and cytokines in wound healing. Wound Repair Regen.
2008;16:585–601.
9. Werner S, Grose R. Regulation of wound healing by growth factors and
cytokines. Physiol Rev. 2003;83:835–70.
10. Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound healing: a cel-
lular perspective. Physiol Rev. 2019;99:665–706.
11. Werner S, Alzheimer C. Roles of activin in tissue repair, fibrosis, and inflam-
matory disease. Cytokine Growth Factor Rev. 2006;17:157–71.
12. Hübner G, Hu Q, Smola H, Werner S. Strong induction of activin expres-
sion after injury suggests an important role of activin in wound repair.
Dev Biol. 1996;173:490–8.
13. Zhang L, Xu P, Wang X, Zhang M, Yan Y, Chen Y, et al. Activin B regulates
adipose-derived mesenchymal stem cells to promote skin wound heal-
ing via activation of the MAPK signaling pathway. Int J Biochem Cell Biol.
2017;87:69–76.
14. Sadok A, Marshall CJ. Rho GTPases: masters of cell migration. Small
GTPases. 2014;5: e29710.
15. Etienne-Manneville S, Hall A. Integrin-mediated activation of Cdc42
controls cell polarity in migrating astrocytes through PKCzeta. Cell.
2001;106:489–98.
16. Srinivasan S, Wang F, Glavas S, Ott A, Hofmann F, Aktories K, et al. Rac and
Cdc42 play distinct roles in regulating PI(3,4,5)P3 and polarity during
neutrophil chemotaxis. J Cell Biol. 2003;160:375–85.
17. Zegers MM, Friedl P. Rho GTPases in collective cell migration. Small
GTPases. 2014;5: e28997.
18. Ge J, Burnier L, Adamopoulou M, Kwa MQ, Schaks M, Rottner K, et al.
RhoA, Rac1, and Cdc42 differentially regulate alphaSMA and collagen I
expression in mesenchymal stem cells. J Biol Chem. 2018;293:9358–69.
19. Florian MC, Dorr K, Niebel A, Daria D, Schrezenmeier H, Rojewski M, et al.
Cdc42 activity regulates hematopoietic stem cell aging and rejuvenation.
Cell Stem Cell. 2012;10:520–30.
20. Wang X, Tang P, Guo F, Zhang M, Yan Y, Huang M, et al. mDia1 and Cdc42
regulate Activin B-induced migration of bone marrow-derived mesen-
chymal stromal cells. Stem Cells. 2019;37:150–62.
21. Li J, Li H, Tian W. Isolation of murine adipose-derived stromal/stem cells
using an explant culture method. Methods Mol Biol. 2018;1773:167–71.
22. Tian Z, Lv X, Zhang M, Wang X, Chen Y, Tang P, et al. Deletion of epithelial
cell-specific Cdc42 leads to enamel hypermaturation in a condi-
tional knockout mouse model. Biochim Biophys Acta Mol Basis Dis.
2018;1864:2623–32.
23. Lim JS, Yoo G. Effects of adipose-derived stromal cells and of their extract
on wound healing in a mouse model. J Korean Med Sci. 2010;25:746–51.
24. Araña M, Mazo M, Aranda P, Pelacho B, Prosper F. Adipose tissue-derived
mesenchymal stem cells: isolation, expansion, and characterization.
Methods Mol Biol. 2013;1036:47–61.
25. Zhou N, Lee JJ, Stoll S, Ma B, Wiener R, Wang C, et al. Inhibition of SRF/
myocardin reduces aortic stiffness by targeting vascular smooth muscle
cell stiffening in hypertension. Cardiovasc Res. 2017;113:171–82.
26. Choi YJ, Koo JB, Kim HY, Seo JW, Lee EJ, Kim WR, et al. Umbilical cord/
placenta-derived mesenchymal stem cells inhibit fibrogenic activation
in human intestinal myofibroblasts via inhibition of myocardin-related
transcription factor A. Stem Cell Res Ther. 2019;10:291.
27. Tang P, Wang X, Zhang M, Huang S, Lin C, Yan F, et al. Activin B stimulates
mouse vibrissae growth and regulates cell proliferation and cell cycle
progression of hair matrix cells through ERK signaling. Int J Mol Sci.
2019;20:853.
28. Zhang M, Wang X, Guo F, Jia Q, Liu N, Chen Y, et al. Cdc42 deficiency leads
to epidermal barrier dysfunction by regulating intercellular junctions
and keratinization of epidermal cells during mouse skin development.
Theranostics. 2019;9:5065–84.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 19 of 19
Huangetal. Stem Cell Research & Therapy (2022) 13:248
fast, convenient online submission
thorough peer review by experienced researchers in your field
rapid publication on acceptance
support for research data, including large and complex data types
gold Open Access which fosters wider collaboration and increased citations
maximum visibility for your research: over 100M website views per year
At BMC, research is always in progress.
Learn more biomedcentral.com/submissions
Ready to submit your research
Ready to submit your research
? Choose BMC and benefit from:
? Choose BMC and benefit from:
29. Altavilla D, Saitta A, Cucinotta D, Galeano M, Deodato B, Colonna M, et al.
Inhibition of lipid peroxidation restores impaired vascular endothelial
growth factor expression and stimulates wound healing and angiogen-
esis in the genetically diabetic mouse. Diabetes. 2001;50:667–74.
30. Wu Y, Chen L, Scott PG, Tredget EE. Mesenchymal stem cells enhance
wound healing through differentiation and angiogenesis. Stem Cells.
2007;25:2648–59.
31. Jacobi J, Jang JJ, Sundram U, Dayoub H, Fajardo LF, Cooke JP. Nicotine
accelerates angiogenesis and wound healing in genetically diabetic
mice. Am J Pathol. 2002;161:97–104.
32. Galeano M, Altavilla D, Cucinotta D, Russo GT, Calo M, Bitto A, et al.
Recombinant human erythropoietin stimulates angiogenesis and wound
healing in the genetically diabetic mouse. Diabetes. 2004;53:2509–17.
33. Zhang M, Sun L, Wang X, Chen S, Kong Y, Liu N, et al. Activin B promotes
BMSC-mediated cutaneous wound healing by regulating cell migration
via the JNK-ERK signaling pathway. Cell Transplant. 2014;23:1061–73.
34. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low
memory requirements. Nat Methods. 2015;12:357–60.
35. Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL.
StringTie enables improved reconstruction of a transcriptome from RNA-
seq reads. Nat Biotechnol. 2015;33:290–5.
36. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-
Seq data with or without a reference genome. BMC Bioinformatics.
2011;12:323.
37. Love MI, Huber W, Anders S. Moderated estimation of fold change and
dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.
38. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package
for differential expression analysis of digital gene expression data. Bioin-
formatics. 2010;26:139–40.
39. Xie C, Mao X, Huang J, Ding Y, Wu J, Dong S, et al. KOBAS 2.0: A web
server for annotation and identification of enriched pathways and dis-
eases. Nucleic Acids Res. 2011;39:W316–22.
40. Bourin P, Bunnell BA, Casteilla L, Dominici M, Katz AJ, March KL, et al.
Stromal cells from the adipose tissue-derived stromal vascular fraction
and culture expanded adipose tissue-derived stromal/stem cells: a joint
statement of the International Federation for Adipose Therapeutics and
Science (IFATS) and the International Society for Cellular Therapy (ISCT).
Cytotherapy. 2013;15:641–8.
41. Gualdrini F, Esnault C, Horswell S, Stewart A, Matthews N, Treisman R. SRF
co-factors control the balance between cell proliferation and contractil-
ity. Mol Cell. 2016;64:1048–61.
42. Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and
regeneration. Nature. 2008;453:314–21.
43. Huayllani MT, Sarabia-Estrada R, Restrepo DJ, Boczar D, Sisti A, Nguyen JH,
et al. Adipose-derived stem cells in wound healing of full-thickness skin
defects: a review of the literature. J Plast Surg Hand Surg. 2020;54:263–79.
44. Bao P, Kodra A, Tomic-Canic M, Golinko MS, Ehrlich HP, Brem H. The role
of vascular endothelial growth factor in wound healing. J Surg Res.
2009;153:347–58.
45. Chen S, Shi J, Zhang M, Chen Y, Wang X, Zhang L, et al. Mesenchymal
stem cell-laden anti-inflammatory hydrogel enhances diabetic wound
healing. Sci Rep. 2015;5:18104.
46. Zhang Z, Wang J, Chen Y, Suo L, Chen H, Zhu L, et al. Activin a promotes
myofibroblast differentiation of endometrial mesenchymal stem cells via
STAT3-dependent Smad/CTGF pathway. Cell Commun Signal. 2019;17:45.
47. Hiratsuka T, Bordeu I, Pruessner G, Watt FM. Regulation of ERK basal
and pulsatile activity control proliferation and exit from the stem
cell compartment in mammalian epidermis. Proc Natl Acad Sci USA.
2020;117:17796–807.
48. Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways medi-
ated by ERK, JNK, and p38 protein kinases. Science. 2002;298:1911–2.
49. Zhang Y, Chen S, Liu B, Zhou H, Hu S, Zhou Y, et al. Exendin-4 promotes
proliferation of adipose-derived stem cells through ERK and JNK signaling
pathways. In Vitro Cell Dev Biol Anim. 2016;52:598–606.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... We then performed paired-end sequencing on an IlluminaHiseq4000 (LC Sciences, LLC, Houston, TX, USA), according to the manufacturer's instructions. DEGs were identi ed as previously described [31]. Brie y, DESeq2/EdgeR with Q value ≤ 0.05 was used for the differential expression analysis of RNA-seq data in terms of |log2FC| > 1 and Q value ≤ 0.05 (DESeq2 or EdgeR) or Q value ≤ 0.001 (DEGseq). ...
Preprint
Full-text available
Fibroblast growth factor 2 (FGF2) is a crucial factor in odontoblast differentiation and dentin matrix deposition, which facilitates pulpodentin repair and regeneration. Nevertheless, the specific biological function of FGF2 in odontoblastic differentiation remains unclear because it is controlled by complex signalling pathways. This study aimed to investigate the mechanism underlying the effect of FGF2 on osteo/odontogenic differentiation of stem cells from the apical papilla (SCAP). SCAP were pretreated with conditioned media containing FGF2 for one week, followed by culturing in induced differentiation medium for another week. RNA sequencing (RNA-seq) combined with quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to evaluate the pathways affected by FGF2 in SCAP. Osteo/odontogenic differentiation of SCAP was determined using Alizarin red S staining, alkaline phosphatase staining, RT-qPCR, and western blotting. Pretreatment with FGF2 for one week increased the osteo/odontogenic differentiation ability of SCAP. RNA-seq and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that phosphatidylinositol 3-kinase (PI3K)/AKT signalling is involved in the osteogenic function of FGF2. RT-qPCR results indicated that SCAP expressed FGF receptors, and western blotting showed that p-AKT was reduced in FGF2-pretreated SCAP. The activation of the PI3K/AKT pathway partially reversed the stimulatory effect of FGF2 on osteo/odontogenic differentiation of SCAP. Our findings suggest that pretreatment with FGF2 enhances the osteo/odontogenic differentiation ability of SCAP by inhibiting the PI3K/AKT pathway.
... Through series studies, Zhang et al. uncovered that activin B/Rho A/mDia1/Cdc42 axis plays a key function in bone marrow-derived mesenchymal stromal cells (BMSCs) migration by promoting membrane ruffling, microtubule morphology, and adhesion signaling dynamics. Inactivation of activin B-Cdc42 inhibits stimulation of Golgi polarization and adipose-derived mesenchymal stem cells-mediated skin wound healing [6][7][8]. Furthermore, overexpressing of activin B was identified in three different models of kidney fibrosis, as well as in human kidneys with fibrosis. Sox9-activin B provides a prospective entry point to surmount kidney fibrosis [9,10]. ...
Article
Full-text available
Background: Important roles of INHBB in various malignancies are increasingly identified. The underlying mechanisms in gastric cancer (GC) microenvironment are still greatly unexplored. Methods: The clinical significance of INHBB and the correlation between INHBB and p-p65 in GC were assessed through analyzing publicly available databases and human paraffin embedded GC tissues. The biological crosstalk of INHBB between GC cells and fibroblasts was explored both in vitro and in vivo. RNA-seq analyses were performed to determine the mechanisms which regulating fibroblasts reprogramming. Luciferase reporter assay and chromatin immunoprecipitation (CHIP) assay were used to verify the binding relationship of p65 and INHBB in GC cells. Results: Our study showed that INHBB level was significantly higher in GC, and that increased INHBB was associated with poor survival. INHBB positively regulates the proliferation, migration, and invasion of GC cells in vitro. Also, activin B promotes the occurrence of GC by reprogramming fibroblasts into cancer-associated fibroblasts (CAFs). The high expression of INHBB in GC cells activates the NF-κB pathway of normal gastric fibroblasts by secreting activin B, and promotes fibroblasts proliferation, migration, and invasion. In addition, activin B activates NF-κB pathway by controlling TRAF6 autoubiquitination to induce TAK1 phosphorylation in fibroblasts. Fibroblasts activated by activin B can induce the activation of p65 phosphorylation of GC cells by releasing pro-inflammatory factors IL-1β. p65 can directly bind to the INHBB promoter and increase the INHBB transcription of GC cells, thus establishing a positive regulatory feedback loop to promote the progression of GC. Conclusions: GC cells p65/INHBB/activin B and fibroblasts p65/IL-1β signal loop led to the formation of a whole tumor-promoting inflammatory microenvironment, which might be a promising therapeutic target for GC.
Article
Full-text available
Skin aging has been associated with the onset of various skin issues, and recent studies have identified an increase in Cdc42 activity in naturally aging mice. While previous literature has suggested that CASIN, a specific inhibitor of Cdc42 activity, may possess anti‐aging properties, its specific effects on the epidermis and dermis, as well as the underlying mechanisms in naturally aging mice, remain unclear. Our study revealed that CASIN demonstrated the ability to increase epidermal and dermal thickness, enhance dermal‐epidermal junction, and stimulate collagen and elastic fiber synthesis in 9‐, 15‐, and 24‐month‐old C57BL/6 mice in vivo. Moreover, CASIN was found to enhance the proliferation, differentiation, and colony formation and restore the cytoskeletal morphology of primary keratinocytes in naturally aging skin in vitro. Furthermore, the anti‐aging properties of CASIN on primary fibroblasts in aging mice were mediated by the ribosomal protein RPL4 using proteomic sequencing, influencing collagen synthesis and cytoskeletal morphology both in vitro and in vivo. Meanwhile, both subcutaneous injection and topical application exhibited anti‐aging effects for a duration of 21 days. Additionally, CASIN exhibited anti‐inflammatory properties, while reduced expression of RPL4 was associated with increased inflammation in the skin of naturally aging mice. Taken together, our results unveil a novel function of RPL4 in skin aging, providing a foundational basis for future investigations into ribosomal proteins. And CASIN shows promise as a potential anti‐aging agent for naturally aging mouse skin, suggesting potential applications in the field.
Article
Full-text available
Mesenchymal stem cells (MSCs) have been isolated from a variety of tissues using different methods. Active research have confirmed that the most accessible site to collect them is the adipose tissue; which has a significantly higher concentration of MSCs. Moreover; harvesting from adipose tissue is less invasive; there are no ethical limitations and a lower risk of severe complications. These adipose-derived stem cells (ASCs) are also able to increase at higher rates and showing telomerase activity, which acts by maintaining the DNA stability during cell divisions. Adipose-derived stem cells secret molecules that show important function in other cells vitality and mechanisms associated with the immune system, central nervous system, the heart and several muscles. They release cytokines involved in pro/anti-inflammatory, angiogenic and hematopoietic processes. Adipose-derived stem cells also have immunosuppressive properties and have been reported to be “immune privileged” since they show negative or low expression of human leukocyte antigens. Translational medicine and basic research projects can take advantage of bioprinting. This technology allows precise control for both scaffolds and cells. The properties of cell adhesion, migration, maturation, proliferation, mimicry of cell microenvironment, and differentiation should be promoted by the printed biomaterial used in tissue engineering. Self-renewal and potency are presented by MSCs, which implies in an open-source for 3D bioprinting and regenerative medicine. Considering these features and necessities, ASCs can be applied in the designing of tissue engineering products. Understanding the heterogeneity of ASCs and optimizing their properties can contribute to making the best therapeutic use of these cells and opening new paths to make tissue engineering even more useful.
Article
Full-text available
Significance Understanding how intracellular signaling cascades control cell fate is a key issue in stem cell biology. Here we show that exit from the stem cell compartment in mammalian epidermis is characterized by pulsatile ERK MAPK activity. Basal activity and pulses are differentially regulated by DUSP10 and DUSP6, two phosphatases that have been shown previously to regulate differentiation commitment in the epidermis. ERK activity is controlled both transcriptionally and posttranscriptionally. Spatial segregation of mean ERK activity and pulses is observed both in reconstituted human epidermis and in mouse epidermis. Our findings demonstrate the tight spatial and temporal regulation of ERK MAPK expression and activity in mammalian epidermis.
Article
Full-text available
Background: The lack of anti-fibrotic agents targeting intestinal fibrosis is a large unmet need in inflammatory bowel diseases, including Crohn's disease and ulcerative colitis. Previous studies have found that perinatal tissue (umbilical cord, UC; placenta, PL)-derived mesenchymal stem cells (MSCs) reduce fibrosis in several organs. However, their effects on human intestinal fibrosis are poorly understood. This study investigated the anti-fibrogenic properties and mechanisms of MSCs derived from UC and PL (UC/PL-MSCs) on human primary intestinal myofibroblasts (HIMFs). Methods: The HIMFs were treated with TGF-β1 and co-cultured with UC/PL-MSCs. We used a small molecular inhibitor CCG-100602 to examine whether serum response factor (SRF) and its transcriptional cofactor myocardin-related transcription factor A (MRTF-A) are involved in TGF-β1-induced fibrogenic activation in HIMFs. The anti-fibrogenic mechanism of UC/PL-MSCs on HIMFs was analyzed by detecting the expression of RhoA, MRTF-A, and SRF in HIMFs. Results: UC/PL-MSCs reduced TGF-β1-induced procollagen1A1, fibronectin, and α-smooth muscle actin expression in HIMFs. This anti-fibrogenic effect was more apparent in the UC-MSCs. TGF-β1 stimulation increased the expressions of RhoA, MRTF-A, and SRF in the HIMFs. TGF-β1 induced the synthesis of procollagen1A1, fibronectin, and α-smooth muscle actin through a MRTF-A/SRF-dependent mechanism. Co-culture with the UC/PL-MSCs downregulated fibrogenesis by inhibition of RhoA, MRTF-A, and SRF expression. Conclusions: UC/PL-MSCs suppress TGF-β1-induced fibrogenic activation in HIMFs by blocking the Rho/MRTF/SRF pathway and could be considered as a novel candidate for stem cell-based therapy of intestinal fibrosis.
Article
Full-text available
Rationale: Cdc42 is a Rho GTPase that regulates diverse cellular functions. Here, we used genetic techniques to investigate the role of Cdc42 in epidermal development and epidermal barrier formation. Methods: Keratinocyte-restricted Cdc42 knockout mice were generated with the Cre-LoxP system under the keratin 14 (K14) promoter. The skin and other tissues were collected from mutant and wild-type mice, and their cellular, molecular, morphological, and physiological features were analyzed. Results: Loss of Cdc42 in the epidermis in vivo resulted in neonatal lethality and impairment of epidermal barrier formation. Cdc42 deficiency led to the loss of epidermal stem cells. The absence of Cdc42 led to increased thickening of the epidermis, which was associated with increased proliferation and reduced apoptosis of keratinocytes. In addition, Cdc42 deficiency damaged tight junctions, adherens junctions and desmosomes. RNA sequencing results showed that the most significantly altered genes were enriched by the terms of “keratinization” and “cornified envelope” (CE). Among the differentially expressed genes in the CE term, several members of the small proline-rich protein (SPRR) family were upregulated. Further study revealed that there may be a Cdc42-SPRR pathway, which may correlate with epidermal barrier function. Conclusions: Our study indicates that Cdc42 is essential for epidermal development and epidermal barrier formation. Defects in Cdc42-SPRR signaling may be associated with skin barrier dysfunction and a variety of skin diseases.
Article
Full-text available
Adipose tissue is now on the top one of stem cell sources regarding its accessibility, abundance, and less painful collection procedure when compared to other sources. The adipose derived stem cells (ADSCs) that it contains can be maintained and expanded in culture for long periods of time without losing their differentiation capacity, leading to large cell quantities being increasingly used in cell therapy purposes. Many reports showed that ADSCs-based cell therapy products demonstrated optimal efficacy and efficiency in some clinical indications for both autologous and allogeneic purposes, hence becoming considered as potential tools for replacing, repairing, and regenerating dead or damaged cells. In this review, we analyzed the therapeutic advancement of ADSCs in comparison to bone marrow (BM) and umbilical cord (UC)-mesenchymal stem cells (MSCs) and designed the specific requirements to their best clinical practices and safety. Our analysis was focused on the ADSCs, rather than the whole stromal vascular fraction (SVF) cell populations, to facilitate characterization that is related to their source of origins. Clinical outcomes improvement suggested that these cells hold great promise in stem cell-based therapies in neurodegenerative, cardiovascular, and auto-immunes diseases.
Article
Full-text available
Background Endometriosis, characterized by the presence of functional endometrial tissues outside the uterus, is one of the most common gynecological disorders. Endometrial mesenchymal stem cells (MSCs) are crucial for the occurrence and development of endometriosis. Ectopic endometrial MSCs exist in the peritoneal cavity. Thus, the bioactive factors in endometriotic peritoneal fluid may regulate the biological behaviors of endometrial MSCs. Methods In this study, after assessing the concentration of Activin A in peritoneal fluid using ELISA, we isolated and cultured endometrial MSCs and investigated whether Activin A stimulated endometrial MSCs to differentiate into myofibroblasts and clarified the underlying mechanisms by quantitative real-time PCR, Western blot analysis, immunofluorescent staining, RNA interference and Chromatin immunoprecipitation. We also employed the inhibitors of Activin A to explore the possibility of suppressing the development of fibrosis in endometriosis using primary endometrial MSCs cultures and a mouse model of endometriosis. Results Here, we revealed that Activin A significantly elevated in endometriotic peritoneal fluid and activin receptor-like kinase (ALK4), the specific receptor for Activin A, obviously enhanced in ectopic endometrial MSCs compared with eutopic endometrial MSCs from women with or without endometriosis. Next, we found that Activin A drived myofibroblast differentiation of endometrial MSCs, with extremely enhanced expression of connective tissue growth factor (CTGF). CTGF was shown to be required for Activin A-induced expression of ACTA2, COL1A1 and FN1 in endometrial MSCs. CTGF induction by Activin A in endometrial MSCs involved the activation of Smad2/3, as evidenced by the phosphorylation and nuclear translocation of Smad2/3 as well as the binding of Smad2/3 to CTGF promoter. Furthermore, Smad/CTGF pathway in endometrial MSCs required activation of STAT3 while independent of PI3K, JNK and p-38 pathways. In addition, we also demonstrated that inhibition of Activin A pathway impeded myofibroblast differentiation of endometrial MSCs and ameliorated fibrosis in endometriosis mice. Conclusions Activin A promotes myofibroblast differentiation of endometrial mesenchymal stem cells via STAT3-dependent Smad/CTGF pathway. The results provided the first evidence that STAT3 acted as a crucial Activin A downstream mediator to regulate CTGF production. Our data may supplement the stem cell theory of endometriosis and provide the experimental basis to treat endometriosis-associated fibrosis by manipulating Activin A signaling. Electronic supplementary material The online version of this article (10.1186/s12964-019-0361-3) contains supplementary material, which is available to authorized users.
Article
Full-text available
Abstract Normal wound healing is a dynamic and complex multiple phase process involving coordinated interactions between growth factors, cytokines, chemokines, and various cells. Any failure in these phases may lead wounds to become chronic and have abnormal scar formation. Chronic wounds affect patients’ quality of life, since they require repetitive treatments and incur considerable medical costs. Thus, much effort has been focused on developing novel therapeutic approaches for wound treatment. Stem-cell-based therapeutic strategies have been proposed to treat these wounds. They have shown considerable potential for improving the rate and quality of wound healing and regenerating the skin. However, there are many challenges for using stem cells in skin regeneration. In this review, we present some sets of the data published on using embryonic stem cells, induced pluripotent stem cells, and adult stem cells in healing wounds. Additionally, we will discuss the different angles whereby these cells can contribute to their unique features and show the current drawbacks.
Article
Full-text available
Activins and their receptors play important roles in the control of hair follicle morphogenesis, but their role in vibrissae follicle growth remains unclear. To investigate the effect of Activin B on vibrissae follicles, the anagen induction assay and an in vitro vibrissae culture system were constructed. Hematoxylin and eosin staining were performed to determine the hair cycle stages. The 5-ethynyl-2′-deoxyuridine (EdU) and Cell Counting Kit-8 (CCK-8) assays were used to examine the cell proliferation. Flow cytometry was used to detect the cell cycle phase. Inhibitors and Western blot analysis were used to investigate the signaling pathway induced by Activin B. As a result, we found that the vibrissae follicle growth was accelerated by 10 ng/mL Activin B in the anagen induction assay and in an organ culture model. 10 ng/mL Activin B promoted hair matrix cell proliferation in vivo and in vitro. Moreover, Activin B modulates hair matrix cell growth through the ERK–Elk1 signaling pathway, and Activin B accelerates hair matrix cell transition from the G1/G0 phase to the S phase through the ERK–Cyclin D1 signaling pathway. Taken together, these results demonstrated that Activin B may promote mouse vibrissae growth by stimulating hair matrix cell proliferation and cell cycle progression through ERK signaling.
Article
The complex process of wound healing can be delayed in circumstances when the natural niche is extremely altered. Adipose-derived stem cells (ADSC) seem to be a promising therapy for these type of wounds. We aim to describe the studies that used ADSC for wound healing after a full-thickness skin defect, the ADSC mechanisms of action, and the outcomes of the different ADSC therapies applied to date. We performed a review by querying PubMed database for studies that evaluated the use of ADSC for wound healing. The Mesh terms, adipose stem cells AND (skin injury OR wound healing) and synonyms were used for the search. Our search recorded 312 articles. A total of 30 articles met the inclusion criteria. All were experimental in nature. ADSC was applied directly (5 [16.7%]), in sheets (2 [6.7%]), scaffolds (14 [46.7%]), skin grafts (3 [10%]), skin flaps (1 [3.3%]), as microvesicles or exosomes (4 [13.3%]), with adhesives for wound closure (1 [3.3%]), and in a concentrated conditioned hypoxia-preconditioned medium (1 [3.3%]). Most of the studies reported a benefit of ADSC and improvement of wound healing with all types of ADSC therapy. ADSC applied along with extracellular matrix, stromal cell-derived factor (SDF-1) or keratinocytes, or ADSC seeded in scaffolds showed better outcomes in wound healing than ADSC alone. ADSC have shown to promote angiogenesis, fibroblast migration, and up-regulation of macrophages chemotaxis to enhance the wound healing process. Further studies should be conducted to assure the efficacy and safety of the different ADSC therapies.
Article
Wound healing is one of the most complex processes in the human body. It involves the spatial and temporal synchronization of a variety of cell types with distinct roles in the phases of hemostasis, inflammation, growth, re-epithelialization, and remodeling. With the evolution of single cell technologies, it has been possible to uncover phenotypic and functional heterogeneity within several of these cell types. There have also been discoveries of rare, stem cell subsets within the skin, which are unipotent in the uninjured state, but become multipotent following skin injury. Unraveling the roles of each of these cell types and their interactions with each other is important in understanding the mechanisms of normal wound closure. Changes in the microenvironment including alterations in mechanical forces, oxygen levels, chemokines, extracellular matrix and growth factor synthesis directly impact cellular recruitment and activation, leading to impaired states of wound healing. Single cell technologies can be used to decipher these cellular alterations in diseased states such as in chronic wounds and hypertrophic scarring so that effective therapeutic solutions for healing wounds can be developed.