ArticlePDF Available

Apigenin Induces Dermal Collagen Synthesis Via smad2/3 Signaling Pathway

Authors:
  • Shanghai Tongji Hospital

Abstract and Figures

Decrease in fibroblast-produced collagen has been proven to be the pivotal cause of skin aging, but there is no satisfactory drug which directly increases dermal thickness and collage density. Here we found that a flavonoid natural product, apigenin, could significantly increase collagen synthesis. NIH/3T3 and primary human dermal fibroblasts (HDFs) were incubated with various concentrations of apigenin, with dimethyl sulfoxide (DMSO) serving as the negative control. Real-time reverse-transcription polymerase chain reaction (PCR), Western Blot, and Toluidine blue staining demonstrated that apigenin stimulated type-I and type-III collagen synthesis of fibroblasts on the mRNA and protein levels. Meanwhile, apigenin did not induce expression of alpha smooth muscle actin (α-SMA) in vitro and in vivo, a fibrotic marker in living tissues. Then the production of collagen was confirmed by Masson's trichrome stain, Picrosirius red stain and immunohistochemistry in mouse models. We also clarified that this compound induced collagen synthesis by activating smad2/3 signaling pathway. Taken together, without obvious influence on fibroblasts' apoptosis and viability, apigenin could promote the type-I and type-III collagen synthesis of dermal fibroblasts in vitro and in vivo, thus suggesting that apigenin may serve as a potential agent for esthetic and reconstructive skin rejuvenation.
Content may be subject to copyright.
European Journal of Histochemistry 2015; volume 59:2467
[page 98] [European Journal of Histochemistry 2015; 59:2467]
Apigenin induces dermal
collagen synthesis via smad2/3
signaling pathway
Y. Zhang,1J. Wang,1X. Cheng,2B. Yi,3
X. Zhang,4Q. Li1
1Department of Plastic and
Reconstructive Surgery, Shanghai 9th
People’s Hospital, School of Medicine,
Shanghai Jiao Tong University
2Department of Urology, Renji Hospital,
School of Medicine, Shanghai Jiao Tong
University
3Clinical College of General Hospital of
Beijing Military Region, Anhui Medical
University, Hefei
4The Key Laboratory of Stem Cell
Biology, Institute of Health Sciences,
Shanghai Institutes for Biological
Sciences, Chinese Academy of Sciences
and Shanghai Jiao Tong University School
of Medicine, Shanghai, China
Abstract
Decrease in fibroblast-produced collagen
has been proven to be the pivotal cause of skin
aging, but there is no satisfactory drug which
directly increases dermal thickness and col-
lage density. Here we found that a flavonoid
natural product, apigenin, could significantly
increase collagen synthesis. NIH/3T3 and pri-
mary human dermal fibroblasts (HDFs) were
incubated with various concentrations of api-
genin, with dimethyl sulfoxide (DMSO) serv-
ing as the negative control. Real-time reverse-
transcription polymerase chain reaction
(PCR), Western Blot, and Toluidine blue stain-
ing demonstrated that apigenin stimulated
type-I and type-III collagen synthesis of fibrob-
lasts on the mRNA and protein levels.
Meanwhile, apigenin did not induce expres-
sion of alpha smooth muscle actin (α-SMA) in
vitro and in vivo, a fibrotic marker in living tis-
sues. Then the production of collagen was con-
firmed by Masson’s trichrome stain,
Picrosirius red stain and immunohistochem-
istry in mouse models. We also clarified that
this compound induced collagen synthesis by
activating smad2/3 signaling pathway. Taken
together, without obvious influence on fibrob-
lasts’ apoptosis and viability, apigenin could
promote the type-I and type-III collagen synthe-
sis of dermal fibroblasts in vitro and in vivo,
thus suggesting that apigenin may serve as a
potential agent for esthetic and reconstructive
skin rejuvenation.
Introduction
Dermis consists of several structural compo-
nents, and collagen takes the major part. In
addition to glycosaminoglycans and elastin
fibers, dermal matrix in adult skin are com-
posed of type I (80-85%) and type III collagen
(10-15%).1It is of great importance that colla-
gen plays a main role in the texture and
appearance of skin. Skin aging is inevitably
associated with a disturbance in collagen
metabolism2(due to the decreased activity of
fibroblasts and their collagen synthesis), as
well as elastin. Increasing the collagen content
of dermis has been regarded as a well-effective
way for anti-aging in skin.
Collagen is a kind of biomacromolecule and
it cannot be absorbed through the stratum
corneum. Recently, some researches demon-
strated that collagen hydrolysate ingestion
might be beneficial to slow chronological skin
aging3and photoaging4in rats, and the density
of collagen fibrils increased compared with lac-
talbumin and water controls.5But the clinical
effect of oral collagen hydrolysate still lacks
convincing evidences. Up to now, injectable
collagen1or analogous composition6filling
implants are recognized as the well-accepted
treatment modality for cosmetic purposes.
However, maintaining skin appearance relies
on expansive and complex treatment repeated-
ly because of the short-term duration of exo-
genic injected collagen.
Apigenin (4,5,7-trihydroxyflavone), a
flavone subclass of flavonoid widely distributed
in many herbs, fruits, and vegetables, is a sub-
stantial component of the human diet and has
been shown to possess a variety of biological
characteristics, including anti-oxidative,7anti-
inflammatory effect,8tumor growth inhibition9
and promoting neurogenesis.10 It has been
shown that apigenin could enhance wound
healing and tissue repair in diabetic rat skin.11
In the process of wound healing, fibroblasts
secreted collagen and the formation of colla-
gen-rich granulation tissue are vital patho-
physiological mechanisms for wound closure.12
Given this, we wonder what effect would
apigenin have on fibroblasts and whether api-
genin could induce collagen synthesis in nor-
mal human dermal fibroblasts. Consequently,
we examined its effects on collagen synthesis
in normal human dermal fibroblasts in vitro
and tested its function in the skin aging mouse
model induced by D-Galactose. Furthermore,
we investigated the potential mechanism
involved in the positive effects of apigenin on
collagen expression in fibroblasts.
Materials and Methods
Cell culture
Primary human dermal fibroblasts were
obtained from adolescent foreskin tissue of ten
people (aged 8-12 years), and none of them
had a history with skin diseases. Skin tissue
was obtained after obtaining informed consent
from the patients, with the approval of the
ethics committee of Shanghai 9th People’s
Hospital and in conformity with the Helsinki
guidelines. NIH/3T3 and human dermal fibrob-
lasts (HDFs) were maintained in DMEM
(Hyclone, Thermo Fisher Scientific, Waltham,
MA, USA) supplemented with 10% fetal bovine
Correspondence: Xiaoling Zhang and Qingfeng
Li, Department of Plastic and Reconstructive
Surgery, Shanghai 9th People’s Hospital,
Shanghai Jiao Tong University School of
Medicine, 639 Zhizaoju Road, Shanghai 200011,
China.
Tel. +86.21.23271699-5615 - Fax: +86.21.63089567.
E-mails: (X. Zhang) xlzhang@sibs.ac.cn; (Q. Li)
dr.liqingfeng@yahoo.com
Key words: Apigenin, flavonoid, collagen I/III,
fibroblasts, smad2/3.
Contributions: YZ and JW contributed equally to
this work; YZ, JW, experimental work, data collec-
tion and interpretation; XC, participation in
experimental work design and coordination, data
acquisition; BY, participation in study design,
data collection, analysis of data and manuscript
preparation; QL, XZ, study design, data analysis
and interpretation, manuscript drafting.
Conflict of interest: the authors declare no con-
flict of interest.
Funding: this study was supported by grants from
the key project of the National Natural Science
Foundation (No. 81230042), the National Key
Project of Scientific and Technical Supporting
Programs Funded by Ministry of Science &
Technology of China (No. 2012BAI11B03) (Q. Li)
and the Chinese Academy of Sciences (No.
XDA01030102), Shanghai Municipal Commission
of Health and Family Planning (No.
2013ZYJB0501) (X. Zhang).
Received for publication: 28 November 2014.
Accepted for publication: 9 March 2015.
This work is licensed under a Creative Commons
Attribution NonCommercial 3.0 License (CC BY-
NC 3.0).
©Copyright Y. Zhang et al., 2015
Licensee PAGEPress, Italy
European Journal of Histochemistry 2015; 59:2467
doi:10.4081/ejh.2015.2467
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:54 Pagina 98
[European Journal of Histochemistry 2015; 59:2467] [page 99]
serum (Hyclone, Thermo Fisher Scientific),
100 U/mL penicillin, and 100 mg/L strepto-
mycin. Both HDFs and NIH/3T3 were incubated
at 37°C in a humidified atmosphere with 5%
CO2. Primary fibroblasts of passages 6-8 were
used. Toluidine blue (Sigma-Aldrich, St. Louis,
MO, USA) staining was used to assess extra-
cellular matrix synthesis.13
Real-time PCR analysis
The total RNA of cells was isolated using
TRIzol reagent (Invitrogen, Carlsbad, CA, USA)
and subjected to reverse transcription with
Oligo (dT) and M-MLV Reverse Transcriptase
(Thermo Fisher Scientific). Synthesized com-
plementary DNA (cDNA) was analysed with
quantitative real-time PCR using SYBR®
Premix (Takara, Dalian, China) and Roche480
system. Glyceraldehyde 3-phospate dehydroge-
nase (GAPDH) was used as a reference gene.
Primers sequences were as follows: collagen,
type I, alpha 2 (Col1a2; mouse), 5-GGTGAGC-
CTGGTCAAACGG-3 (forward) and 5- ACTGT-
GTCCTTTCACGCCTTT-3 (reverse); Col1a2
(human), 5-GGCCCTCAAGGTTTCCAAGG-3
(forward) and 5-CACCCTGTGGTCCAA-
CAACTC-3 (reverse); collagen, type III, alpha 1
(Col3a1; mouse), 5- CTGTAACATG-
GAAACTGGGGAAA-3 (forward) and 5-
CCATAGCTGAACTGAAAACCACC-3 (reverse);
Col3a1 (human), 5-TTGAAGGAGGATGTTCC-
CATCT-3 (forward) and 5- ACAGACA-
CATATTTGGCATGGTT-3 (reverse); matrix
metalloproteinases 1 (MMP1; human), 5-
AAAATTACACGCCAGATTTGCC-3 (forward)
and 5-GGTGTGACATTACTCCAGAGTTG-3
(reverse); matrix metalloproteinases 2
(MMP2; human), 5-TACAGGATCATTGGCTA-
CACACC-3 (forward) and 5-GGTCA-
CATCGCTCCAGACT-3 (reverse); matrix metal-
loproteinases 9 (MMP9; human), 5-TGTAC-
CGCTATGGTTACACTCG-3 (forward) and 5-
GGCAGGGACAGTTGCTTCT-3 (reverse); tissue
inhibitor of metalloproteinases1 (TIMP1;
human), 5-CTTCTGCAATTCCGACCTCGT-3
(forward) and 5-ACGCTGGTATAAGGTG-
GTCTG-3 (reverse); α-SMA (human), 5-
AAAAGACAGCTACGTGGGTGA-3 (forward) and
5-GCCATGTTCTATCGGGTACTTC-3 (reverse).
Cell viability assay
For the cell viability assay, HDFs were seed-
ed on 96-well plates (100 mL per well), fol-
lowed by apigenin (cat. no. 42251; Sigma-
Aldrich) or DMSO (Sigma-Aldrich) treatment.
After 3 or 5 days, cell culture medium was
replaced by Thiazolyl Blue Tetrazolium
Bromide (MTT) working solution, followed by
a 4-hour incubation at 37°C in a 5% CO2incu-
bator. After MTT working solution was
removed and DMSO added, the absorbances at
490 nm were detected.
Flow cytometric analysis
Cell apoptosis was assessed by flow cytome-
try using the Alexa Fluor®488 Annexin V/Dead
Cell Apoptosis Kit (Invitrogen, Carlsbad, CA,
USA). Following apigenin (5 μmol/L or 1
μmol/L) or dimethyl sulfoxide (DMSO) treat-
ment, harvested cells were suspended in 100
μL Annexin-binding buffer. Then, 5 μL Alexa
Fluor® 488 Annexin V and 1 μL PI working
solution were added and incubated with the
cells for 15 min in the dark. After the incuba-
tion period, 400 μL 1X Annexin-binding buffer
was added and mixed gently. The stained cells
were analysed directly by flow cytometry using
the Cell Quest program (Becton Dickinson, CA,
USA). Data were analysed using FlowJo soft-
ware.
Colony formation assay
Anchorage-dependent growth of HDFs were
investigated by monolayer colony formation
assay.14 Cells were cultured in a 6-well plate
(500 per well) and treated with 5 μmol/L or 1
μmol/L apigenin or DMSO. After cultured for
14 days, surviving colonies were stained 5 min
with Gentian Violet (Sigma-Aldrich) after 4%
paraformaldehyde fixation.
Immunofluorescence cell staining
Human dermal fibroblasts at a density of
2×103cells per well were seeded on cover
slides in 24-well plates and incubated
overnight. Cells were fixed with 4%
paraformaldehyde and blocked with 5% goat
serum in PBST (0.1% TritonX-100 in phos-
phate buffered saline) for 1 h. For F-actin
staining, cells were incubated with Alexa Fluor
488 Phalloidin (Cytoskeleton Inc., Denver, CO,
USA; 1:200) for 1 h at room temperature. For
α-SMA staining, cells were incubated with pri-
mary antibodies against α-SMA (Abcam,
Cambridge, UK, 1:200) for 2 h at room temper-
ature, followed by an Alexa Fluor 555-conjugat-
ed secondary antibody. For smad3 staining,
cells were incubated with primary antibodies
against smad3 (Cell Signaling Technology,
Beverly, MA, USA; 1:200) for 2 h at room tem-
perature, followed by an Alexa Fluor 488-conju-
gated secondary antibody. Immunofluore -
scence signals were captured using confocal
microscopy (LSM 510, META Laser Scanning
Microscope; Zeiss, Jena, Germany).
Smad2/3 knockdown by siRNA
RNA interference was performed using
smad2/3 siRNA (human) (sc-37238; Santa
Cruz Biotechnology, Dallas, TX, USA), target-
ing human smad2/3 and control siRNA (sc-
37007) as negative control. Transfection for
HDFs was conducted using Lipofectamine
RNAiMAX reagent (Invitrogen, Carlsbad, CA,
USA) according to the manufacturer’s protocol.
Western blot
Cultured cells were lysed using radioim-
munoprecipitation assay (RIPA) lysis buffer.
Protein concentrations were determined using
a micro bicinchoninic acid (BCA) assay
(Thermo Fisher Scientific). Twenty micro-
grams total protein extract was separated by
8% or 10% sodium dodecyl sulfate-polyacry-
lamide gel electrophoresis (SDS-PAGE) under
reducing conditions and electroblotted onto
polyvinylidene difluoride membranes
(Millipore, Bedford, MA, USA). The membrane
was then blocked and then incubated with pri-
mary antibodies overnight at 4°C.
The primary antibodies used included the
followings: anti-Col1a2, anti-Col3a1, anti-α-
SMA (Abcam; 1:1000), anti-cyclin-dependent
kinase (CDK) family, anti-cyclin D1, anti-
cyclin E1, anti-smads, anti-MAPK family (Cell
Signaling Technology, Beverly, MA, 1:1000),
anti-GAPDH (Sigma-Aldrich; 1:10,000).
Immunoreactive bands were quantitatively
analyzed with ImageJ software.
D-galactose-induced skin aging
mouse model and animal
experiments
Six-week-old female C57BL/6 mice were
purchased from the Shanghai Slac Laboratory
Animal (Slac, Shanghai, China). All animal
studies have been approved by the Animal
Care and Use Committee of Shanghai Jiao
Tong University. All efforts were made to mini-
mize animal suffering.
A total of 18 mice were randomly assigned to
three groups (n=6). Two groups of animals
received daily subcutaneous injection of D-
galactose (D-gal; 1000 mg/kg) for 8 weeks.15
The third group received phosphate buffered
saline (PBS) as a negative control. Two weeks
later, DMSO and apigenin (5 μmol/L) was
delivered by microneedles16 [MTS-Roller
Model: CR2 (0.2 mm)] to the dermis of D-
galactose treated mice once a day for 4 weeks,
respectively. Mice were sacrificed at the end of
treatment, and skin tissue was harvested for
further analyses.
Histology and immunohistochemistry
Paraformaldehyde-fixed paraffin-embedded
tissue sections (5 μm) were stained with
hematoxylin and eosin (H&E), Masson’s
trichrome (Trichrome stain LG solution,
HT10316; Sigma-Aldrich) and Picrosirius red
(Fluka, Buchs, Switzerland). For immunohis-
tochemical staining, the sections were detect-
ed with primary antibodies against collagen
I/III (Millipore; 1:1,000) and α-SMA (Abcam;
1:200) overnight at 4°C. After incubation with
the appropriate secondary antibodies, the sec-
tions were developed with diaminobenzidine
and counterstained with hematoxylin.
Original Paper
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:54 Pagina 99
[page 100] [European Journal of Histochemistry 2015; 59:2467]
Statistical analysis
Statistical differences were calculated using
Friedman’s analysis of variance (ANOVA), with
post-hoc least significant difference (LSD) test
as appropriate. A significant difference among
groups was set at P<0.05.
Results
Apigenin stimulated collagen
synthesis but had no effect on
matrix metalloproteinases
in vitro
Fibroblasts are the predominant mesenchy-
mal cells in the dermis, and their function is
strongly implicated in dermatology. To study
apigenin’s effect on fibroblasts (Figure 1A),
NIH/3T3 and HDFs were administered with
apigenin at the concentrations of 5 μmol/L. As
shown in Figure 1B, apigenin could potently
increase Toluidine blue staining after api-
genin treatment for 5 days, which mean the
Original Paper
Figure 1. Apigenin stimulated collagen synthesis of fibroblasts. A) The molecular structure of apigenin. B) Toluidine blue staining in
NIH/3T3 and HDFs for 5 days. C) Dose-dependent effects of apigenin on mRNA expression of Col1a2 and Col3a1 in NIH/3T3 and
HDFs for 3 days. D) The protein level of Col1a2 and Col3a1 was measured by Western Blot at 5 days after apigenin was applied at
concentrations of 0.1 μmol/L to 10 μmol/L. E) The expression of MMP1, MMP2, MMP9 and TIMP1 were also assessed by real-time
PCR. Data are presented as mean ± SD, n3; NS, not significant; *P<0.05; **P< 0.01; ***P<0.001.
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:54 Pagina 100
[European Journal of Histochemistry 2015; 59:2467] [page 101]
synthesis of extracellular matrix was
increased.13 Further real-time PCR analysis
showed that, in NIH/3T3 and HDFs, apigenin
(0.1 μmol/L - 10 μmol/L) dose-dependently
stimulated endogenous expression of Col1a2
and Col3a1. The most significant changes
were observed when NIH/3T3 and HDFs were
treated with apigenin at the concentration of 5
μmol/L, and the increase of Col3a1 was more
obvious than Col1a2 (Figure 1C). These upreg-
ulation effect of apigenin on collagen expres-
sion were then confirmed by Western blot
analysis. When HDFs were treated with api-
genin for 5 days, the protein level of Col1a2
and Col3a1 were higher than that of cells treat-
ed with DMSO (Figure 1D). In addition, we
next examined the effect of apigenin on
matrix metalloproteinases (MMPs) and
Original Paper
Figure 2. No obvious cytotoxicity exerted by apigenin on fibroblasts viability, apoptosis, proliferation, cell cycle and activation. A) Cell
viability was examined by MTT assays at 3 or 5 days after apigenin was applied in HDFs. B) Apoptosis was evaluated after treating
HDFs with 5 μmol/L or 1 μmol/L apigenin or DMSO; flow cytometry profile represents Alexa Fluor® 488 Annexin V staining in X
axis and PI in Y axis. C) The effect of apigenin on fibroblasts growth was investigated by monolayer colony formation assay. D) The
expression of cyclin E1, CDK4, cyclin D1, CDK2 and p-CDK2 proteins was analysed using Western blot in HDFs. E-F) The levels of
α-SMA mRNA and protein expression were measured by real-time PCR and Western blot. G) Immunofluorescence cell staining for α-
SMA and F-actin in cultured HDFs after incubation with apigenin or DMSO for 72 h; F-actin is shown by green fluorescence and α-
SMA is shown by red fluorescence; nucleus (blue) was stained with DAPI; scale bar: 50 μm. Data are presented as mean ± SD, n3;
NS, not significant; **P<0.01; ***P<0.001.
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:55 Pagina 101
TIMP1, well-known proteases that degrade col-
lagen proteins. The expression of MMP1,
MMP2, MMP9, and their inhibitor TIMP1 were
unchanged (Figure 1E).
Apigenin did not effect fibroblasts
viability and activity
Afterwards, we studied the effect of api-
genin on fibroblasts viability, apoptosis, prolif-
eration and activation. In vitro, MTT assays
showed that the viability of HDFs was similar
with those incubated with DMSO, when incu-
bated with apigenin (1 to 10 μmol/L) for 3 or 5
days, respectively (Figure 2A). In addition, no
significant differences in percentages of apop-
totic cells was observed after the exposure of
fibroblasts to apigenin (Figure 2B). To investi-
gate the effects of apigenin on proliferation
and cell cycle, colony formation assay and
Western Blot analysis of cell cycle related pro-
teins were performed. Colony forming ability
of HDFs was similar in apigenin-treated group
to DMSO-treated group (Figure 2C). The
expression levels of cell cycle associated pro-
teins remained unchanged between apigenin
and DMSO treated groups (Figure 2D). These
results suggested that apigenin had no obvi-
ous cytotoxicity on fibroblasts’ viability, apop-
tosis and proliferation.
Fibroblast overactivation leads to pathologi-
cal collagen deposition or scar formation.17
Myofibroblasts, known as activated fibroblasts,
are marked by α-SMA expression. To deter-
mine the effects of apigenin on fibroblasts’
activation, we evaluated the levels of α-SMA
mRNA and protein expression (Figure 2 E,F)
in cultured HDFs treated with apigenin. We
found that α-SMA mRNA expression had no
obvious change in apigenin-treated cells com-
pared with DMSO-treated cells. We also
showed that apigenin did not affect α-SMA
expression in vitro by immunofluorescence
staining (Figure 2G). These findings suggest-
ed that apigenin did not cause fibroblasts to
overactivate into myofibroblasts while colla-
gen synthesis was increasing.
Induction of collagen synthesis was
mediated by smad2/3 activation
To further explore the underlying mecha-
nism of how apigenin activated type-I and
type-III collagen gene expression, transform-
ing growth factor beta 1 (TGF-β1) and mito-
gen-activated protein kinase (MAPK) signal-
ing pathway were analysed. TGF-β1 is a proto-
typic fibrogenic cytokine, enhancing extracel-
lular matrix gene expression. Previous studies
proved that Col1a2 and Col3a1 were direct
TGF-β1/smad3 targets in human dermal
fibroblasts.18 As shown in Figure 3A, when
HDFs were treated for 12 h, apigenin (1
μmol/L or 5 μmol/L) markedly increased the
expression of phosphorylated smad2 and
smad3 in a dose-dependent manner, whereas
total smad2, smad3 and smad4 did not obvi-
ously alter. It also showed that apigenin had
sustained effect on promoting phosphorylation
of smad2 and smad3 after a 3-day treatment
(Figure 3B). Yoon et al.19 revealed that MAPK
pathway was involved with peptide-induced
collagen synthesis of fibroblasts. However,
when fibroblasts were treated with apigenin
for 12 h, the expression of total and phospho-
rylated JNK, ERK and p38 protein remained
unchanged, compared with DMSO (Figure
3C). Immunofluorescence experiments de -
monstrated that after treatment with apigenin
for 12 h, smad3 protein (labeled by green) was
significantly increased and mostly translocat-
ed into the nucleus (labeled by blue) (Figure
3D). By contrast, in the DMSO groups, smad3
were retained in the cytoplasm. Once targeted
knockdown smad2/3 by specific siRNA, the up-
regulation effect of apigenin on the expres-
sion of collagen type-I and type-III protein was
obviously reduced (Figure 3E), which con-
firmed that smad2/3 is required for the trans-
duction of apigenin effect on collagen expres-
sions.
Apigenin stimulated collagen syn-
thesis in the D-galactose-induced
skin aging mouse model
The in vivo effects of apigenin on collagen
synthesis was investigated in the D-galactose-
induced skin aging mouse model. The collagen
expression was showed by H&E, Masson’s
trichrome stain, Picrosirius red stain and
immunohistochemistry. Histology showed sig-
nificant changes in dermal thickness and den-
sity in samples obtained from D-gal-treated
mice compared with PBS control group
(Figure 4 A-D). After 1 month of apigenin
administration at the concentration of 5
μmol/L, the mice exhibited obviously
increased dermal thickness and collagen den-
sity compared with DMSO-treated mice
(Figure 4 A-D). Magnified images showed that
dermis in the apigenin-treated group exhibit-
ed compact and clearly evident staining,
whereas collagens were loosely distributed in
DMSO-treated dermis of the aging skin model
(Figure 4 A-D), in both Masson’s trichrome
stain, Picrosirius red stain and immunohisto-
chemistry examinations. Dermal collagen
could be subdivided into type I and type III col-
lagen after Picrosirius red staining under
polarized light. As shown in Figure 4F, api-
genin could significantly increase both colla-
gen type I and type III density in the dermis of
the skin aging mouse model. Quantitative data
of dermal thickness and collagen density
showed that mice subcutaneously injected
with D-gal showed thinner skin and less colla-
gen compared to control mice and apigenin-
treated mice demonstrated significantly thick-
er and compact dermis (Figure 4 E,F). The in
vivo study also demonstrates that apigenin
does not show any effect on activation of
fibroblasts (Figure 4G).
Discussion
Dermal atrophy is the major causes of aging
appearance.20 In vivo and in vitro studies show
that decline in the production of collagen in
aging fibroblasts is mainly responsible for
decreasing in dermal thickness seen in extrin-
sically aging skin, which reveals dermal atro-
phy, fragmentation, and irregular collagen
bundles.21 Since the ‘70s, animal and human
derived collagens have been studied for soft
tissue augmentation.22 Injectable filling
implants are now widely used for cosmetic pur-
poses. However, exogenic injectable collagen
often presented various complications such as
allergy, ecchymosis, local necrosis and infec-
tions of bacteria or virus. Scientists have tried
for decades to find other alternative to stimu-
late endogenous collagen synthesis. There are
several anti-oxidants, such as vitamins C and
E, co-enzyme Q10 and retinoids used for treat-
ing UV-induced skin aging and wrinkles.23, 24
However, only few compounds are able to
induce type I collagen synthesis25-27 and none
of them can stimulate both type I and type III
collagen synthesis according to the record in
literatures.
Apigenin, a plant flavone, has gained con-
siderable attention due to its health benefits,
chemopreventive properties and wide distribu-
tion in the plant kingdom.28 Many studies have
demonstrated that apigenin possesses a wide
range of biological activities to the skin. It has
been reported that apigenin can stimulate
nucleotide excision repair genes to protect
skin keratinocytes29 against UVB-induced skin
inflammation.30 Dietary apigenin attenuates
the development of atopic dermatitis-like skin
lesions in atopic dermatitis model.31 Apigenin
could also effectively reduce the incidence and
size of skin tumors caused by ultraviolet B
(UVB) exposure through the enhancement of
UVB-induced apoptosis.32 In this study, we
investigated the effect of apigenin on dermal
fibroblasts’ function.
At first, we found apigenin could increase
the mRNA expression of Col1a2 and Col3a1 in
NIH/3T3 and HDFs. With extracellular matrix
staining and Western Blot analysis, the stimu-
lative effect of collagen on protein level was
more significant. Although as reported in the
literature, basal levels of Col1a1 and α-SMA
mRNAs were reduced in fibroblasts treated
with high concentration of apigenin (20
μmol/L),33 our research confirmed that api-
genin of 25 μmol/L showed obvious cytotoxici-
[page 102] [European Journal of Histochemistry 2015; 59:2467]
Original Paper
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:55 Pagina 102
[European Journal of Histochemistry 2015; 59:2467] [page 103]
Original Paper
Figure 3. Apigenin-mediated collagen synthesis increase via smad2/3 signaling pathway. A,B) Western blot analysis and quantification
of phosphorylated and total smad2, smad3 and total smad4 in HDFs with or without apigenin stimulation for 12 h or 3 days. C)
Western Blot analysis of JNK, ERK and p38 in HDFs with or without apigenin stimulation for 12 h. D) Immunofluorescence experi-
ments: smad3 was labeled as green; nucleus (blue) was stained with DAPI; scale bar: 50 μm. E) Western blot showed the expression of
Col1a2 and Col3a1 in HDFs when cells were treated with DMSO, apigenin or apigenin with specific siRNA of smad2/3. Data are pre-
sented as mean ± SD, n3; NS, not significant; *P<0.05; **P<0.01; ***P<0.001.
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:55 Pagina 103
[page 104] [European Journal of Histochemistry 2015; 59:2467]
Original Paper
Figure 4. Apigenin increased dermal thickness and collagen density in the D-galactose-induced skin aging mouse model. A-D) H&E,
Masson’s trichrome, Picrosirius red and immunohistochemistry stained dermis of control mice and D-gal-treated mice respectively
received apigenin and DMSO; scale bars: 100 μm; zoom scale bars: 20 μm. E) Quantification of dermal thickness. F) Picrosirius red
staining under polarized light and quantification of type I and type III collagen density; collagen type I is shown as red fibers and col-
lagen type III is shown as green fibers; scale bars: 100 μm. G) Immunohistochemistry staining of α-SMA of control mice and D-gal-
treated mice respectively received apigenin and DMSO; scale bars: 100 μm; zoom scale bars: 20 μm. Data are presented as mean ± SD,
n=6/6/6; *P<0.05; **P<0.01; ***P<0.001.
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:55 Pagina 104
[European Journal of Histochemistry 2015; 59:2467] [page 105]
ty in fibroblasts. We believed that the attenuat-
ing effect of high concentration of apigenin on
phenotypic transitions in the analyzed cell pop-
ulations were not independent of its cytotoxic
activity. We detected the markers related to
extracellular matrix degradation and found
that apigenin had no effect on the balance of
MMPs/TIMPs.
We further observed that apigenin directly
activated smad2/3-dependent signaling path-
way. This is not surprising since this flavonoid
displays considerable muti-effect. It targets a
number of secondary messengers, including
those potentially involved in TGF-β1 signaling
pathway, such as NF-kB,34 MAPK/ERK,35
FAK,36,37 PKC38 and PI3K-Akt39 in a cell context-
dependent manner. We observed that apigenin
markedly increased the expression of phos-
phorylated smad2 and smad3 protein, while
total smad2, smad3 and smad4 protein all
remained unaltered. So, a more meticulous
network may connect TGF-β1 signaling path-
way and the abovementioned secondary mes-
sengers.
A previous study showed the accelerated
aging effect of D-gal injection on mouse skin,
as well as changes in dermal thickness and col-
lagen content.15 In order to confirm the effect
of apigenin on collagen synthesis in vivo, the
D-galactose-induced skin aging mouse model
was established. Our data indicated that skin
aging mice treated with apigenin showed
markedly increasing dermal thickness and col-
lagen expression, compared with DMSO-treat-
ed mice. Hou et al.40 reported that topical api-
genin improved epidermal permeability barrier
function by stimulating epidermal differentia-
tion, lipid synthesis and secretion, as well as
cutaneous antimicrobial peptide production,
and our result showed that dermal injection of
apigenin significantly increased dermal thick-
ness and density. So we could conclude that
apigenin caused different biological functions
with two forms of drug administration by act-
ing on epidermis or derma, which might indi-
cate the importance of choosing suitable
administration methods to different skin dis-
eases, even for the same drug.
Our study demonstrates that apigenin could
induce both type I and type III collagen synthe-
sis of fibroblasts in vitro and could increase
dermal thickness and collagen deposition in
the dermis of mice. This compound is a poten-
tial target for drug design and development for
esthetic and reconstructive purpose.
References
1. Baumann L, Kaufman J, Saghari S.
Collagen fillers. Dermatol Ther 2006;19:
134-40.
2. Fisher GJ, Wang ZQ, Datta SC, Varani J,
Kang S, Voorhees JJ. Pathophysiology of
premature skin aging induced by ultravio-
let light. N Engl J Med 1997;337:1419-28.
3. Liang JA, Pei XR, Zhang ZF, Wang N, Wang
JB, Li Y. The Protective Effects of Long-
Term Oral Administration of Marine
Collagen Hydrolysate from Chum Salmon
on Collagen Matrix Homeostasis in the
Chronological Aged Skin of Sprague-
Dawley Male Rats. J Food Sci 2010;75:
H230-8.
4. Hou H, Li BF, Zhang ZH, Xue CH, Yu GL,
Wang JF, et al. Moisture absorption and
retention properties, and activity in allevi-
ating skin photodamage of collagen
polypeptide from marine fish skin. Food
Chem 2012;135:1432-9.
5. Matsuda N, Koyama YI, Hosaka Y, Ueda H,
Watanabe T, Araya T, et al. Effects of inges-
tion of collagen peptide on collagen fibrils
and Glycosaminoglycans in the dermis. J
Nutr Sci Vitaminol (Tokyo) 2006;52:211-5.
6. Iannitti T, Morales-Medina JC, Coacci A,
Palmieri B. Experimental and Clinical
Efficacy of Two Hyaluronic Acid-based
Compounds of Different Cross-Linkage
and Composition in the Rejuvenation of
the Skin. Pharm Res Epub 2014 Jun 25.
7. Sharma H, Kanwal R, Bhaskaran N, Gupta
S. Plant flavone apigenin binds to nucleic
acid bases and reduces oxidative DNA
damage in prostate epithelial cells. PLoS
One 2014;9:e91588.
8. Wang J, Liu YT, Xiao L, Zhu L, Wang Q, Yan
T. Anti-Inflammatory Effects of Apigenin in
Lipopolysaccharide-Induced Inflammatory
in Acute Lung Injury by Suppressing COX-
2 and NF-kB Pathway. Inflammation
2014;37:2085-90.
9. Polier G, Giaisi M, Kohler R, Muller WW,
Lutz C, Buss EC, et al. Targeting CDK9 by
wogonin and related natural flavones
potentiates the anti-cancer efficacy of the
Bcl-2 family inhibitor ABT-263. Int J
Cancer 2015;136:688-98.
10. Taupin P. Apigenin and related compounds
stimulate adult neurogenesis. Mars, Inc.,
the Salk Institute for Biological Studies:
WO2008147483. Expert Opin Ther Pat
2009;19:523-7.
11. Lodhi S, Singhai AK. Wound healing effect
of flavonoid rich fraction and luteolin iso-
lated from Martynia annua Linn. on strep-
tozotocin induced diabetic rats. Asian Pac
J Trop Med 2013;6:253-9.
12. Singer AJ, Clark RA. Cutaneous wound
healing. N Engl J Med 1999;341:738-46.
13. Yano F, Hojo H, Ohba S, Fukai A, Hosaka Y,
Ikeda T, et al. A novel disease-modifying
osteoarthritis drug candidate targeting
Runx1. Ann Rheum Dis 2013;72:748-53.
14. Jin H, Wang X, Ying J, Wong AH, Cui Y,
Srivastava G, et al. Epigenetic silencing of
a Ca(2+)-regulated Ras GTPase-activating
protein RASAL defines a new mechanism
of Ras activation in human cancers. Proc
Natl Acad Sci U S A 2007;104:12353-8.
15. Zhang S, Dong Z, Peng Z, Lu F. Anti-aging
effect of adipose-derived stem cells in a
mouse model of skin aging induced by D-
galactose. PLoS One 2014;9:e97573.
16. Prausnitz MR. Microneedles for transder-
mal drug delivery. Adv Drug Deliv Rev
2004;56:581-7.
17. Wang J, Dodd C, Shankowsky HA, Scott PG,
Tredget EE, Wound Healing Research G.
Deep dermal fibroblasts contribute to
hypertrophic scarring. Lab Invest 2008;
88:1278-90.
18. Verrecchia F, Chu ML, Mauviel A.
Identification of novel TGF-beta/Smad
gene targets in dermal fibroblasts using a
combined cDNA microarray/promoter
transactivation approach. J Biol Chem
2001;276:17058-62.
19. Yoon JH, Kim J, Lee H, Kim SY, Jang HH,
Ryu SH, et al. Laminin peptide YIGSR
induces collagen synthesis in Hs27 human
dermal fibroblasts. Biochem Biophys Res
Commun 2012;428:416-21.
20. Fenske NA, Lober CW. Structural and func-
tional changes of normal aging skin. J Am
Acad Dermatol 1986;15:571-85.
21. Lavker RM. Structural alterations in
exposed and unexposed aged skin. J Invest
Dermatol 1979;73:59-66.
22. Klein AW, Elson ML. The history of sub-
stances for soft tissue augmentation.
Dermatol Surg 2000;26:1096-105.
23. Kwok HH, Yue PYK, Mak NK, Wong RNS.
Ginsenoside Rb-1 induces type I collagen
expression through peroxisome prolifera-
tor-activated receptor-delta. Biochem
Pharmacol 2012;84:532-9.
24. Winterfield L, Cather J, Cather J, Menter
A. Changing paradigms in dermatology:
Nuclear hormone receptors Clin Dermatol.
2003;21:447-54.
25. Lee J, Jung E, Yu H, Kim Y, Ha J, Kim YS,
et al. Mechanisms of carvacrol-induced
expression of type I collagen gene. J
Dermatol Sci 2008;52:160-9.
26. Choi MS, Yoo MS, Son DJ, Jung HY, Lee
SH, Jung JK, et al. Increase of collagen
synthesis by obovatol through stimulation
of the TGF-beta signaling and inhibition of
matrix metalloproteinase in UVB-irradiat-
ed human fibroblast. J Dermatol Sci
2007;46:127-37.
27. Wang J, Zhou J, Zhang N, Zhang X, Li Q. A
heterocyclic molecule kartogenin induces
collagen synthesis of human dermal
fibroblasts by activating the smad4/smad5
pathway. Biochem Biophys Res Commun
2014;450:568-74.
Original Paper
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:55 Pagina 105
[page 106] [European Journal of Histochemistry 2015; 59:2467]
28. Shukla S, Gupta S. Apigenin: a promising
molecule for cancer prevention. Pharm
Res 2010;27:962-78.
29. Das S, Das J, Paul A, Samadder A, Khuda-
Bukhsh AR. Apigenin, a bioactive
flavonoid from Lycopodium clavatum,
stimulates nucleotide excision repair
genes to protect skin keratinocytes from
ultraviolet B-induced reactive oxygen
species and DNA damage. J Acupunct
Meridian Stud 2013;6:252-62.
30. Byun S, Park J, Lee E, Lim S, Yu JG, Lee SJ,
et al. Src kinase is a direct target of api-
genin against UVB-induced skin inflam-
mation. Carcinogenesis 2013;34:397-405.
31. Yano S, Umeda D, Yamashita S, Yamada K,
Tachibana H. Dietary apigenin attenuates
the development of atopic dermatitis-like
skin lesions in NC/Nga mice. J Nutr
Biochem 2009;20:876-81.
32. Abu-Yousif AO, Smith KA, Getsios S, Green
KJ, Van Dross RT, Pelling JC.
Enhancement of UVB-induced apoptosis
by apigenin in human keratinocytes and
organotypic keratinocyte cultures. Cancer
Res 2008;68:3057-65.
33. Ricupero DA, Poliks CF, Rishikof DC,
Kuang PP, Goldstein RH. Apigenin
decreases expression of the myofibroblast
phenotype. FEBS Lett 2001;506:15-21.
34. Kang OH, Lee JH, Kwon DY. Apigenin
inhibits release of inflammatory media-
tors by blocking the NF-kappaB activation
pathways in the HMC-1 cells.
Immunopharmacol Immunotoxicol 2011;
33:473-9.
35. Hwang YP, Oh KN, Yun HJ, Jeong HG. The
flavonoids apigenin and luteolin suppress
ultraviolet A-induced matrix metallopro-
teinase-1 expression via MAPKs and AP-1-
dependent signaling in HaCaT cells. J
Dermatol Sci 2011;61:23-31.
36. Franzen CA, Amargo E, Todorovic V, Desai
BV, Huda S, Mirzoeva S, et al. The
Chemopreventive Bioflavonoid Apigenin
Inhibits Prostate Cancer Cell Motility
through the Focal Adhesion Kinase/Src
Signaling Mechanism. Cancer Prev Res
(Phila) 2009;2:830-41.
37. Hu XW, Meng D, Fang J. Apigenin inhibited
migration and invasion of human ovarian
cancer A2780 cells through focal adhesion
kinase. Carcinogenesis 2008;29:2369-76.
38. Balasubramanian S, Zhu L, Eckert RL.
Apigenin inhibition of involucrin gene
expression is associated with a specific
reduction in phosphorylation of protein
kinase C delta Tyr(311). J Biol Chem
2006;281:36162-72.
39. Shukla S, Gupta S. Apigenin-induced cell
cycle arrest is mediated by modulation of
MAPK, PI3K-Akt, and loss of cyclin D1
associated retinoblastoma dephosphoryla-
tion in human prostate cancer cells. Cell
Cycle 2007;6:1102-14.
40. Hou M, Sun R, Hupe M, Kim PL, Park K,
Crumrine D, et al. Topical apigenin
improves epidermal permeability barrier
homoeostasis in normal murine skin by
divergent mechanisms. Exp Dermatol
2013;22:210-5.
Original Paper
EJH_2015_02-article.qxp_Hrev_master 22/06/15 12:55 Pagina 106
... Here, we showed that apigenin and phloretin exert senomorphic effects on senescent fibroblasts by regulating extracellular matrix proteins ( Figure 6), which might have a beneficial effect on photoaged skin pigmentation. Interestingly, previous studies showed that apigenin induces collagen I synthesis in dermal fibroblasts while it does not affect MMP1 or TIMP1 expression [43]. According to our research, apigenin does not affect collagen I gene expression but downregulates and upregulates MMP1 and TIMP1 expression, respectively. ...
Article
Full-text available
Melasma is a pathology with multifactorial causes that results in hyperpigmentation of sun-exposed areas, particularly facial skin. New treatments targeting the different factors regulating this condition need to be effective with and have limited adverse effects. Here, we describe a novel combination of two natural compounds (apigenin and phloretin) that has synergistic effects regulating melanogenesis in vitro. Both compounds inhibit Wnt-stimulated melanogenesis and induce autophagy in melanocytes. Apigenin induces DKK1, a Wnt pathway inhibitor, and reduces VEGF, a melanogenesis and proangiogenic factor, in fibroblasts. Moreover, apigenin induces miR-675, a melanogenesis inhibitor miRNA that is reduced in melasma skin in melanocytes. Both compounds showed senomorphic effects by regulating extracellular-matrix-related genes in senescent fibroblasts. Topical application of the compounds also showed significant melanin reduction in a reconstructed human epidermis after 7 days. Thus, the combination of apigenin and phloretin shows promising results as an effective topical treatment of skin hyperpigmentation conditions.
... P. senegalensis is reported to be used in Benin, Egypt, Nigeria, Tanzania, and Ethiopia, for the treatment of skin conditions including swelling, wound infections, syphilitic sores, or other skin conditions (Andreozzi and Sardaky, 2022;Bothon et al., 2013;Getahun, 1976;Kio and Ola-Adams, 1990;Kokwaro, 1993;Tadele et al., 2023). These uses have not been directly validated but may be supported by the presence of several compounds such as quercetin, apigenin and oleic acid, which possess anti-inflammatory, wound healing, anti-collagenase and anti-elastase activity (Edwards et al., 2004;Sharma et al., 2018;Zhang et al., 2015). Similarly, Kokwaro (1993) reported that P. senegalensis has been traditionally used to manage diseases in cattle and remove ectoparasites from livestock. ...
... Treatment with apigenin in the current study is believed to prevent gastric ulcers by increasing the expression of TGF-β1 in the gastric mucosa of the treated animals. Likewise, apigenin was found to induce collagen synthesis in the skin through activating TGF-β1signaling (Zhang et al. 2015). ...
Article
Full-text available
Gastric ulcer disease is associated with significant morbidity and mortality rates. The most two common causes of the ulcer are Helicobacter pylori infection and non-steroidal anti-inflammatory drugs. In the past few decades, a significant decrease in the morbidity and mortality rate has been observed probably due to the discovery of proton pump inhibitors. However, the medications used to treat gastric ulcers impose several nauseous side effects. Therefore, recent studies focus on the use of natural products to treat gastric ulcers. In the current study, gastric ulcer was effectively induced using indomethacin, and the protective effect of apigenin, a potent antioxidant flavonoid, was assessed in comparison to omeprazole. The administration of a single oral indomethacin (50 mg/kg) induced gastric ulcer as manifested by hemorrhagic lesions in the gastric mucosa, increased ulcer index, and histopathological alterations. Indomethacin also increased lipid peroxidation, decreased the activities of the antioxidant enzymes superoxide dismutase (SOD) and catalase, increased the immunoreactivity of the inflammatory markers cyclo-oxygenase-2 (COX-2), tumor necrosis factor-alpha (TNF-α), and nuclear factor-kappa B (NF-κB), increased the transcription of the apoptotic marker, Bax, and decreased that of the antiapoptotic Bcl-2. Indomethacin also decreased the immunoreactivity of transforming growth factor-beta 1 (TGF-β1). On the other hand, pretreatment with apigenin (10 and 20 mg/kg) resulted in a dose-dependent improvement in the macroscopic and microscopic features of the gastric mucosa in a manner comparable to that of omeprazole. The gastroprotective effects of apigenin may be attributed to its anti-inflammatory, anti-antioxidant, and anti-apoptotic activities as well as enhancing the expression of TGF-β1. Further experimental and clinical research is required to confirm activity of apigenin as anti-ulcer agent. Graphical Abstract
... Flavonoids have different effects on skin extracellular matrix synthesis, with some of them inhibiting collagen synthesis and others promoting it [66]. It has been reported that apigenin glucosides promote collagen I synthesis in vitro [67,68]; however, in our study, M. recutita extract did not have an effect on collagen, while JC extract had a stimulating activity. β-thujaplicin is also known to have MMP-1-decreasing activity [69]. ...
Article
Full-text available
For skin health promotion and cosmetic applications, combinations of plant cell extracts are extensively utilized. As most natural ingredient suppliers offer crude extracts from individual plants or specific isolated compounds, the potential interactions between them are assessed in the development phase of cosmetic products. The industry seeks extract combinations that have undergone optimization and scrutiny for their bioactivities. This study presents a combination of two sustainably produced botanical ingredients and outlines their chemical composition, in vitro safety, and bioactivity for skin health enhancement. The amalgamation comprises the extract of Matricaria recutita processing waste and the extract from Juniperus communis callus culture. Chemical analysis revealed distinct compounds within the extracts, and their combination led to a broader array of potentially synergistic compounds. In vitro assessments on skin cells demonstrated that the combination possesses robust antioxidant properties and the ability to stimulate keratinocyte proliferation, along with regulating collagen type I and matrix metalloproteinase 1 (MMP-1) production by dermal fibroblasts. The identified traits of this combination render it an appealing cosmetic component. To the best of our knowledge, this represents the first case when the extracts derived from medicinal plant processing waste and biotechnological plant cell cultivation processes have been combined and evaluated for their bioactivity.
... This activity may be related to the inhibition of the activator protein-1 (AP-1), leading to the degradation of the collagen of the dermal matrix. This leads to greater resistance and elasticity of the vessel wall [68,69]. Naringenin is the aglycone of naringin [70]. ...
Article
Full-text available
Objective: Haemorrhoidal disease (HD) is a very diffuse anorectal condition that involves a large part of the population, both male and female of every age. Among the procedures proposed to treat HD, conventional excisional surgery remains one of the most performed. Milligan-Morgan (MM) technique is one of the most used haemorrhoidectomy techniques. In this technique, the wounds are left open and re-epithelialization requires almost 3-5 weeks, in which patients generally experience pain and intense discomfort improving over the weeks. Methods: The aim of this study was to evaluate the effect of topic administration of Benebeo Gel®, mainly composed by bergamot-derived flavonoids and hyaluronic acid, on post-operative wound healing after open MM haemorrhoidectomy. An observational prospective study was carried out, involving 205 patients aged between 18 and 75. Results and Conclusion: The results after 2 weeks of treatment seem to be promising with a very good clinical outcome and patient satisfaction within 1 month.
... Furthermore, it has protective effects on UV-induced aging [93,94] and in other models of cell senescence [84,[95][96][97]. The aglycone apigenin [98][99][100][101][102][103][104] also exerts anti-aging effects. With these results in mind, we hypothesize that rosmarinic acid and apigenin might be the major contributors for the reported antisenescent effects; however, synergistic effects and the contribution of other compounds cannot be discarded. ...
Article
Full-text available
Inflammation plays a pivotal role in the resolution of infection or tissue damage. In addition, inflammation is considered a hallmark of aging, which in turn compromises wound healing. Thymbra capitata is an aromatic plant, whose infusion is traditionally used as an anti-inflammatory and wound-healing agent. In this study, a T. capitata infusion was prepared and characterized by HPLC-PDA-ESI-MSn and its safety profile determined by the resazurin metabolic assay. The anti-inflammatory potential was revealed in lipopolysaccharide (LPS)-stimulated macrophages by assessing nitric oxide (NO) release and levels of inducible nitric oxide synthase (iNOS) and the interleukin-1β pro-form (pro-IL-1β). Wound-healing capacity was determined using the scratch assay. The activity of senescence-associated β-galactosidase was used to unveil the anti-senescent potential, along with the nuclear accumulation of yH2AX and p21 levels. The antiradical potential was assessed by DPPH and ABTS scavenging assays. The infusion contains predominantly rosmarinic acid and salvianolic acids. The extract decreased NO, iNOS, and pro-IL-1β levels. Interestingly, the extract promoted wound healing and decreased β-galactosidase activity, as well as yH2AX and p21 levels. The present work highlights strong antiradical, anti-inflammatory, and wound healing capacities, corroborating the traditional uses ascribed to this plant. We have described, for the first time for this extract, anti-senescent properties.
... Several studies have described the properties of specific flavonoids in preserving collagen stability [103,104]. For instance, anthocyanidins, natural plant pigments found in fruits, flowers, and certain vegetables, have been shown to stabilize collagens [105,106]. ...
Preprint
Full-text available
Collagen, the most abundant structural protein found in mammals, plays a vital role as a constituent of the extracellular matrix (ECM) that surrounds cells. Collagen fibrils are strengthened through the formation of covalent cross-links, which involve complex enzymatic and non-enzymatic reactions. Lysyl oxidase (LOX) is responsible for catalyzing the oxidative deamination of lysine and hydroxylysine residues, resulting in the production of aldehydes, allysine, and hydroxyallysine. These intermediates undergo spontaneous condensation reactions, leading to the formation of immature cross-links, which are the initial step in the development of mature covalent cross-links. Additionally, non-enzymatic glycation contributes to the formation of abnormal cross-linking in collagen fibrils. During glycation, specific lysine and arginine residues in collagen are modified by reducing sugars, leading to the creation of Advanced Glycation End-products (AGEs). These AGEs have been associated with changes in the mechanical properties of collagen fibers. Interestingly, various studies have reported that plant polyphenols possess amine oxidase-like activity and can act as potent inhibitors of protein glycation. This review article focuses on compiling literature describing polyphenols with amine oxidase-like activity and antiglycation properties. Specifically, we explore the molecular mechanisms by which specific flavonoids impact or protect the normal collagen cross-linking process. Furthermore, we discuss how these dual activities can be harnessed to generate properly cross-linked collagen molecules, thereby promoting the stabilization of highly organized collagen fibrils.
Article
Full-text available
New and improved skin care products and procedures have been established by technological advances and scientific investigation. By increasing the skin's moisture, firmness, and elasticity, anti-aging skin care can enhance the skin's overall condition. The objective of this study was to examine the potential of basil's chemical compounds as an in silicoanti-aging agent. Exploration of online databases, scholarly articles from national and international journals, and analysis using docking software are selected as examples of data collection methods. Matrix metalloproteinase 1 (MMP1), with PDB code 966C, is one of the targeted proteins related to skin anti-aging. Ladanein, acacetin, luteolin, 5-hydroxy-7,4'-dimethoxyflavone and genkwanin are five basil compounds that are predicted to exhibit anti-aging agents based on the presence of the binding affinity score indicator and the similarity of the appropriate attachment sites compared to the native ligand used. The scores for the binding affinity of luteolin, ladanein, acacetin, 5-hydroxy-7,4'-dimethoxyflavone, and genkwanin are -10, -9.9, -9.9, -9.8, and -9.6 kcal/mol, respectively. At the attachment positions of five basil compounds, the interactions with ASN 180, LEU 181 and ALA 182 key amino acids, which are the attachment sites for the native ligands, were also formed.
Article
Background: Natural components that can exert a wide range of anti-hair loss activity with fewer side effects are in high demand. The objective of this study was to investigate the anti-hair loss potential of Silybum marianum flower extract (SMFE) in vitro and in vivo. Methods: The effect of SMFE on dermal papilla cells was evaluated by measuring cell proliferation and VEGF production in hair follicle dermal papilla cells (HFDPCs). In addition, to confirm the effect of SMFE on dermal papilla senescence, SA-β-gal staining and senescence associated secretory phenotype (SASP) production such as IL-6 was observed in both replicative and hydrogen peroxide (H2 O2 )-induced senescence models. In a clinical study, hair growth was determined by reconstitution analysis after shaving the hair of the clinical subject's scalp and hair area. Results: SMFE increased the proliferation and VEGF production of HFDPCs. It also suppressed cellular senescence of HFDPCs and IL-6 production in replicative senescence and oxidative stress-induced senescence models. The hair density and total hair count at 16 and 24 weeks after using hair shampoo containing SMFE were significantly increased compared with those of the placebo group. Conclusion: SMFE has the potential to be used as a natural ingredient for alleviating hair loss.
Article
Full-text available
Glycation products accumulate during aging of slowly renewing tissue, including skin, and are suggested as an important mechanism underlying the skin aging process. Adipose-derived cells are widely used in the clinic to treat ischemic diseases and enhance wound healing. Interestingly, adipose-derived stem cells (ASCs) are also effective in anti-aging therapy, although the mechanism underlying their effects remains unknown. The purpose of the present study was to examine the anti-aging effect of ASCs in a D-galactose-induced aging animal model and to clarify the underlying mechanism. Six-week-old nude mice were subcutaneously injected with D-gal daily for 8 weeks. Two weeks after completion of treatment, mice were randomized to receive subcutaneous injections of 106 green fluorescent protein (GFP)-expressing ASCs, aminoguanidine (AG) or phosphate-buffered saline (PBS). Control mice received no treatment. We examined tissue histology and determined the activity of senescence-associated molecular markers such as superoxide dismutase (SOD) and malondialdehyde (MDA). Transplanted ASCs were detectable for 14 days and their GFP signal disappeared at day 28 after injection. ASCs inhibited advanced glycation end product (AGE) levels in our animal model as well as increased the SOD level and decreased the MDA level, all of which act to reverse the aging phenotype in a similar way to AG, an inhibitor of AGE formation. Furthermore, ASCs released angiogenic factors in vivo such as vascular endothelial growth factor, suggesting a skin trophic effect. These results demonstrate that ASCs may contribute to the regeneration of skin during aging. In addition, the data shows that ASCs provide a functional benefit by glycation suppression, antioxidation, and trophic effects in a mouse model of aging.
Article
Full-text available
Oxidative stress has been linked to prostate carcinogenesis as human prostate tissue is vulnerable to oxidative DNA damage. Apigenin, a dietary plant flavone, possesses anti-proliferative and anticancer effects; however, its antioxidant properties have not been fully elucidated. We investigated sub-cellular distribution of apigenin, it's binding to DNA and protective effects against H2O2-induced DNA damage using transformed human prostate epithelial RWPE-1 cells and prostate cancer LNCaP, PC-3 and DU145 cells. Exposure of cells to apigenin exhibited higher accumulation in RWPE-1 and LNCaP cells, compared to PC-3 and DU145 cells. The kinetics of apigenin uptake in LNCaP cells was estimated with a Km value of 5 µmole/L and Vmax of 190 pmoles/million cells/h. Sub-cellular fractionation demonstrated that nuclear matrix retains the highest concentration of apigenin (45.3%), followed by cytosol (23.9%), nuclear membranes (17.9%) and microsomes (12.9%), respectively. Spectroscopic analysis of apigenin with calf-thymus DNA exhibited intercalation as the dominant binding mode to DNA duplex. Apigenin exposure resulted in significant genoprotective effects in H2O2-stressed RWPE-1 cells by reduction in reactive oxygen species levels. In addition, apigenin exposure suppressed the formation of 8-hydroxy-2' deoxyguanosine and protected exposed cells from apoptosis. Our studies demonstrate that apigenin is readily taken up by normal prostatic epithelial cells and prostate cancer cells, and is incorporated into their nuclei, where its intercalation with nucleic acid bases may account for its antioxidant and chemopreventive activities.
Article
Full-text available
In this study, we examined the antioxidative and the DNA protective potentials of apigenin, a flavonoid polyphenol isolated from Lycopodium clavatum, in both in-vitro (HaCaT skin keratinocytes) and in-vivo (mice) models against UV-B radiation. We used DAPI staining in UV-B-irradiated HaCaT skin keratinocytes pre-treated with and without apigenin to assess DNA damage. We also used a flow-cytometric analysis in mice exposed to UV-B radiation with or without topical application of apigenin to assess, through a comet assay, chromosomal aberrations and quanta from reactive oxygen species (ROS) generation. Data from the stability curves for the Gibb's free energy determined from a melting-temperature profile study indicated that apigenin increased the stability of calf thymus DNA. Immunofluorescence studies revealed that apigenin caused a reduction in the number of cyclobutane pyrimidine dimers (CPDs) after 24 h, the time at which the nucleotide excision repair (NER) genes were activated. Thus, apigenin accelerated reversal of UV-B-induced CPDs through up-regulation of NER genes, removal of cyclobutane rings, inhibition of ROS generation, and down-regulation of NF-κB and MAPK, thereby revealing the precise mechanism of DNA repair.
Article
The mechanical and transport-enhancing properties of microneedles were examined. Microneedle arrays were inserted into epidermis and transdermal transport of calcein or fluorescein-labeled BSA was determined by spectrofluorimetry. Following this, microneedles were examined to determine if any breakage occurred during insertion and removal. It was observed that the microneedles are mechanically strong, able to increase transdermal transport by more than four orders of magnitude in vitro, and do not cause pain in human subjects.
Article
This study aims to evaluate the possible mechanisms responsible for the anti-inflammatory effects of apigenin lipopolysaccharide (LPS)-induced inflammatory in acute lung injury. In this study, the anti-inflammatory effects of apigenin on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice and the possible mechanisms involved in this protection were investigated. Pretreatment with apigenin prior to the administration of intratracheal LPS significantly induced a decrease in lung wet weight/dry weight ratio in total leukocyte number and neutrophil percent in the bronchoalveolar lavage fluid (BALF) and in IL-6 and IL-1β, the tumor neurosis factor-α (TNF-α) in the BALF. These results showed that anti-inflammatory effects of apigenin against the LPS-induced ALI may be due to its ability of primary inhibition of cyclooxygenase-2 (COX-2) gene expression and nuclear factor kB (NF-kB) gene expression of lung. The results presented here suggest that the protective mechanism of apigenin may be attributed partly to decreased production of proinflammatory cytokines through the inhibition of COX-2 and NF-kB activation. The results support that use of apigenin is beneficial in the treatment of ALI.
Article
Background: In the field of aesthetic medicine there is an increasing demand for safe and effective hyaluronic acid (HA) fillers to counteract the aging process. Methods and aims: We designed a study to evaluate the safety and histological biocompatibility of Aliaxin® Global Performance, a cross-linked HA filler and Viscoderm® Skinkò E, a product composed of non-cross-linked HA and a complex including vitamins, antioxidants, amino acids and minerals injected into the skin of guinea pigs. Then, we translated our findings into the clinical setting, administering a combination of these compounds to patients seeking a facial rejuvenation procedure targeting moderate-to-severe wrinkles affecting the nasolabial folds. Results: The animal study showed that the two compounds did not induce any significant inflammatory reactions and increased collagen and elastic fibers in the skin. In the clinical setting, injection of Aliaxin® Global Performance, followed by Viscoderm® Skinkò E, resulted in a higher improvement in nasolabial fold hydration, trans-epidermal water loss and wrinkle aesthetic appearance, if compared with a protocol based on Aliaxin® Global Performance alone. Conclusion: In summary, we show evidence on the safety and mechanism underlying two new HA-based compounds of different cross-linkage and composition, proposing that they can be safely used in combination in patients seeking facial rejuvenation procedures with long-lasting efficacy.
Article
Tumor initiation, progression and resistance to therapies are tightly associated with over-expression of anti-apoptotic proteins Bcl-2, Bcl-xL , Bcl-w and Mcl-1. ABT-263 (Navitoclax), an orally bio-available small-molecule mimetic of the Bcl-2 homology domain 3, inhibits Bcl-2, Bcl-xL , and Bcl-w and has shown anti-cancer effects mainly on lymphomas and lymphocytic leukemia. Despite promising results obtained from the clinical trials, the use of ABT-263 in patients is dose-limited due to causing thrombocytopenia via inhibition of Bcl-xL in platelets. ABT-199 specifically inhibits Bcl-2, however, its use is limited to tumors over-expressing only Bcl-2. Besides, many tumors resist treatment due to high levels of Mcl-1 expression or develop resistance via up-regulation of Mcl-1 during long-term exposure. These obstacles highlight the demand to improve the ABT-263-based therapy. In this study, we show that anti-cancer flavones, e.g. wogonin, baicalein, apigenin, chrysin and luteolin, enhance ABT-263-induced apoptosis in different cancer cell lines and in primary AML and ALL cells by down-regulation of Mcl-1 expression. Importantly, wogonin does not enhance the toxicity of ABT-263 to proliferating normal T cells and thrombocytes. Wogonin also potentiates the lethality of ABT-263 in cancer cells which have acquired resistance to ABT-263. Furthermore, we show that combination of wogonin with ABT-263 promotes in vivo tumor regression in a human T-cell leukemia xenograft mouse model. Our study demonstrates that wogonin (and related flavones) reduce the effective dose of ABT-263 thereby possibly decreasing the risk of adverse side effects. © 2014 Wiley Periodicals, Inc.