You are on page 1of 7

Carcinogenesis vol.25 no.8 pp.

1459--1465, 2004
doi:10.1093/carcin/bgh152

Silibinin prevents ultraviolet radiation-caused skin damages in SKH-1


hairless mice via a decrease in thymine dimer positive cells and an
up-regulation of p53-p21/Cip1 in epidermis

Sivanandhan Dhanalakshmi1,*, G.U.Mallikarjuna1,*, non-melanoma skin cancer (NMSC) (1). In addition to its
Rana P.Singh1 and Rajesh Agarwal1--3 carcinogenic activity, UV also causes photoaging, immuno-
1 suppression and sunburn (2--4). There is high morbidity with
Department of Pharmaceutical Sciences, School of Pharmacy
and 2University of Colorado Cancer Center, University of Colorado NMSC, and the associated medical cost is estimated to be over
Health Sciences Center, Denver, CO 80262, USA $500 million/year. In spite of skin cancer awareness in recent
years, the prediction is a further increase in NMSC cases

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


3
To whom correspondence should be addressed
Email: rajesh.agarwal@UCHSC.edu possibly because of several outdoor activities and depletion
in atmospheric ozone (5). This emphasizes the importance of
Non-melanoma skin cancer (NMSC) accounts for >1
developing additional strategies that could control and prevent
million new cases each year in the US alone suggesting
solar UV-caused NMSC. Although the usage of sunscreens
that more approaches are needed for its prevention and
has been an important protection strategy, further approaches
control. Earlier studies by us have shown that silymarin (a
are needed to more effectively protect human skin against
crude form of biologically active silibinin with some other
UV-caused damages as well as NMSC (6,7).
isomers), isolated from milk thistle, affords strong protec-
Chemoprevention of cancer is a novel and more effective
tion against ultraviolet (UV) radiation-induced NMSC in
means of cancer control where naturally occurring agents are
SKH-1 hairless mice; however, the molecular mechanisms
considered as a less toxic and more effective approach in
of its efficacy are not known. Here, we assessed the effect of
controlling various human malignancies including NMSC
silibinin on UV-induced DNA damage and p53-p21/Cip1
(8--12). For example, oral administration or topical application
accumulation, and their roles in UV-induced cell prolifera-
of green tea and black tea, with or without caffeine, has been
tion and apoptosis in SKH-1 hairless mouse epidermis.
shown to strongly prevent UV-induced skin cancer in mouse
Topical application of silibinin prior to, or immediately
skin (13), inhibit skin carcinogenesis in UV-exposed high-risk
after, UV irradiation resulted in a very strong protective
mice (12), and increase apoptotic sunburn cells by inducing
effect against UV-induced thymine dimer positive cells in
p53 and p21/Cip1 (14). Earlier studies by us have shown that
epidermis accounting for 76--85% (P 5 0.001) inhibition.
silymarin (a crude form of biologically active silibinin with
In other studies, silibinin treatment resulted in a further
some other isomers), isolated from milk thistle [Silybum
up-regulation of p53 by ~1.6-fold (P 5 0.001) together with
marianum (L.) Gaertn] plant, affords strong protection against:
an increase (~2-fold, P 5 0.001) in p21/Cip1 protein levels.
(i) UV-induced tumor initiation, promotion and complete
Proliferative cell nuclear antigen staining showed that sili-
carcinogenesis in SKH-1 hairless, and (ii) chemical carcino-
binin pre- or post-topical application significantly inhibits
genesis in SENCAR mouse skin (15--19). Completed studies
(40--52 and 20--40%, respectively, P 5 0.001) UV-induced
by us have also shown that silymarin inhibits UV- and chemi-
epidermal cell proliferation. In addition, silibinin strongly
cal tumor promoter-induced cellular and biochemical events
decreased UV-caused terminal deoxynucleotidyl transfer-
associated with skin tumor promotion (15--19); however, the
ase-mediated dUTP nick end labeling-positive apoptotic/
mechanism of its efficacy against UV-caused damages leading
sunburn cell formation (P 5 0.001). These findings suggest
to skin tumor initiation is not known, which was explored in
that silibinin affords strong protection against UV-induced
the present investigation.
damage in epidermis by a decrease in thymine dimer posi-
UV radiation is known to exert its tumor initiating effects
tive cells and an up-regulation of p53-p21/Cip1 possibly
primarily through the formation of cyclobutane pyrimidine
leading to an inhibition in both cell proliferation and apop-
dimers and 6-4 photoproducts (20) in DNA, as well as via
tosis. Comparable effects of silibinin following its pre- or
formation of reactive oxygen species that subsequently lead to
post-UV application suggest that mechanisms other than
the damage of DNA and other major cellular targets (21). The
sunscreen effect are operational in silibinin efficacy against
UV-damaged cell responds to these alterations either by
UV-caused skin damages.
enhanced DNA repair or by inducing apoptotic death, if the
damage is too severe; failure to do so, however, could lead to
the formation of an initiated cell (22). One important cellular
Introduction
response occurring after UV exposure and/or UV-caused DNA
Ultraviolet (UV) radiation, in the range of 290--320 and damage is phosphorylation of p53 at various serines that
320--400 nm for UVB and UVA wavelengths, respectively, increases its half-life causing its accumulation (23). This also
from the sunlight is the major etiologic factor for the increases the transcriptional activity of p53 leading to the
synthesis of its target genes such as p21/Cip1 that helps the
cells in: (i) arresting DNA synthesis and allowing more time
Abbreviations: H&E, hematoxylin and eosin; NMSC, non-melanoma skin for DNA repair, or (ii) modulating bax levels causing apoptosis
cancer; PCNA, proliferating cell nuclear antigen; UV, ultraviolet; TUNEL, (24,25). Consistent with above reports, in the present study, we
terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling. assessed the effect of topical application of silibinin on UV-

These authors contributed equally to this work and share first authorship. induced DNA damage in terms of thymine dimer positive
Carcinogenesis vol.25 no.8 # Oxford University Press 2004; all rights reserved. 1459
S.Dhanalakshmi et al.

cells, cell proliferation and apoptosis as well as the role of p53 anti-mouse secondary antibody (in case of p21 and PCNA) for 1 h at room
and p21/Cip1 molecules in these UV-caused early adaptive temperature followed by 30 min incubation with HRP-conjugated streptavidin.
changes, in female SKH-1 hairless mouse epidermis. Color development was achieved by incubation with DAB for 10 min at room
temperature. The sections were counterstained with Harris Hematoxylin, dehy-
drated and mounted.
Materials and methods TUNEL staining for apoptotic cells
Animals and UV light source Apoptotic cells were detected using the DeadEnd Colorimetric TUNEL system
(Promega, Madison, WI) following manufacturer's protocol with some mod-
Female SKH-1 hairless mice (5 weeks old) were obtained from Charles River
ifications. In brief, tissue sections after deparaffinization and re-hydration,
Laboratories (Wilmington, MA) and maintained in animal house facility at the
were permeabilized with Proteinase K (30 mg/ml) for 1 h at 37 C. Thereafter,
University of Colorado Health Sciences Center. Animals were fed with Purina
the sections were quenched of endogenous peroxidase activity using 3%
Chow and water ad libitum for a week before the start of the experiments. The
hydrogen peroxide for 10 min. After thorough washing with 1 PBS, sections
UV light source was a bank of four FS-40-T-12-UVB sunlamps equipped with
were incubated with equilibration buffer for 10 min, and then TdT reaction
a UVB Spectra 305 Dosimeter (Daavlin, Bryan, OH), which emitted ~80%
mixture was added to the sections, except for the negative control, and incu-
radiation in the range of 280--340 nm with a peak emission at 314 nm as
bated at 37 C for 1 h. The reaction was stopped by immersing the sections in
monitored with a SEL 240 photodetector, 103 filter and 1008 diffuser attached
2 saline-sodium citrate buffer for 15 min. Sections were then added with
to an IL1400A Research Radiometer (International Light, Newburyport, MA).
streptavidin-HRP (1:500) for 30 min at room temperature, and after repeated

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


The UVB irradiation doses were also calibrated using IL1400A radiometer.
washings, sections were incubated with substrate DAB until color develop-
Mice were exposed to UV irradiation as reported earlier (15) with a distance of
ment (~5--10 min). Sections were then mounted after re-hydration and
23 cm between the light source and the target skin. Silibinin used in the study
observed under 400 for TUNEL-positive cells.
was obtained commercially from Sigma Chemical Company (St Louis, MO),
and was analyzed by HPLC as a pure agent as reported earlier (17).
Measurement of apoptotic sunburn cells
Experimental design Skin sections were stained conventionally with H&E as mentioned earlier (15)
Animals were divided into five groups containing five animals in each group for the identification of sunburn cells. Apoptotic cells are morphologically
for each time-point of the study as follows: (i) unexposed and untreated control distinct due to cell shrinkage and nuclear condensation that stain darker by
mice, (ii) animals topically applied with 9 mg silibinin in 200 ml acetone/ H&E. Dark stained cells were scored in 5 random fields/sample and the
mouse, (iii) animals irradiated once with 180 mJ/cm2 UV dose, (iv) animals percentage/field was calculated.
topically applied with 9 mg silibinin in 200 ml acetone/mouse 30 min prior to
identical UV exposure as in group 3, and (v) animals topically applied with Immunohistochemical and statistical analyses
9 mg silibinin in 200 ml acetone/mouse immediately after identical UV All the microscopic immunohistochemical analyses were done using Zeiss
exposure as in group 3. Animals were killed after UV exposure at various Axioscop 2 microscope (Carl Zeiss, Jena, Germany). Pictures were taken by
time points, and dorsal skin was collected, fixed in 10% formalin for 8--10 h at Kodak DC290 camera under 400 magnification and processed by Kodak
4 C and processed for immunohistochemical analyses. Microscopy Documentation System 290 (Eastman Kodak Company, Roche-
Immunohistochemical and hematoxylin and eosin (H&E) staining analyses ster, NY). For statistical analysis, data were analyzed using the Jandel Scien-
tific SigmaStat 2.03 software (San Rafael, CA). The statistical significance of
Skin samples were processed conventionally before paraffin embedding. difference between control and UV-exposed, and UV-exposed versus silibinin
Briefly, skin samples were dehydrated in 70, 95 and 100% ethanol, cleared plus UV or UV plus silibinin-treated groups was determined by one way
in xylene and embedded in paraffin. Serial, 4 mm sections were cut and used analysis of variance (one-way ANOVA) followed by Bonferroni t-test for
for all immunohistochemical and H&E analyses. The sections were deparaffi- multiple comparisons. A P 5 0.05 was considered statistically significant.
nized, re-hydrated with water and used for measurement of thymine dimer,
p53, p21/Cip1, proliferating cell nuclear antigen (PCNA), terminal deoxy-
nucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) and apopto-
tic sunburn positive cells. All samples were coded and evaluated by two Results
investigators in a blinded manner and the mean  SE values were obtained
from the evaluation of multiple fields in each group. Five to ten representative Silibinin inhibits UV-induced DNA damage in SKH-1 hairless
fields were counted for each mouse at 400 magnification, and the data mouse epidermis
represent the results from at least five mice in each group. Formation of thymine dimers is one of the characteristics of
Measurement of thymine dimer positive cells UV-induced DNA damage, and occurs three times more often
Thymine dimer positive cells in epidermal layer were measured as described as compared with other photoproducts (27). Based on an ear-
earlier (26). Briefly, endogenous peroxidase activity was blocked by 10 min lier study showing that thymine dimer formation occurs as
incubation with 3% hydrogen peroxide, and slides were then incubated with
0.125% trypsin for 10 min at 37 C and then with 1 N HCl for 30 min at room
early as 5 min and peaks at 1 h after UV exposure (26), we
temperature. The sections were then blocked with 5% goat serum for 10 min assessed the effect of silibinin on thymine dimer positive cells
and incubated with peroxidase-conjugated monoclonal anti-thymine dimer in mouse epidermis 1 h after a single UV exposure at 180 mJ/
antibody (Kamiya Biomedical Company, Seattle, WA) for 90 min at room cm2 dose. As shown in Figure 1, compared with unexposed
temperature. Color was developed by incubating the sections in 3,30 -diamino- control mice, UV irradiation resulted in a very strong immu-
benzidine (DAB) for 10 min at room temperature. The sections were counter-
stained with Harris Hematoxylin, dehydrated and mounted. nostaining for thymine dimer in the mouse epidermis; how-
ever, topical application of silibinin prior to or immediately
Immunostaining for p53, p21/Cip1 and PCNA
after (immunostaining data not shown) UV exposure showed a
Skin sections were processed conventionally for immunostaining. Briefly,
tissue sections after deparaffinization and re-hydration were treated with remarkably reduced immunostaining for UV-caused thymine
0.01 M sodium citrate buffer (pH 6.0) in a microwave for 5 min at full power dimer positive cells in epidermis (Figure 1A--C). In quantita-
for antigen-retrieval. Sections were then quenched of endogenous peroxidase tive analysis, UV exposure resulted in 78  2% thymine dimer
activity by immersing in 3% hydrogen peroxide for 5 min at room temperature. positive cells in the epidermis while no such cells were
The sections were then blocked using 5% rabbit serum (only for p53 staining) observed in unexposed controls (Figure 1D). Pre- or post-
for 30 min at room temperature in a humid chamber, and then incubated at 4 C
overnight or at room temperature for 2 h (in case of PCNA) with anti-p53 (cat. topical application of silibinin, to UV exposure, resulted in
#NCL-p53-CM5p rabbit polyclonal antibody against recombinant mouse p53 only 12  1 and 19  1% thymine dimer positive cells,
protein, Novocastra Laboratories, Newcastle upon Tyne, UK), anti-p21/Cip1 accounting for 85 and 76% inhibition (P 5 0.001), respec-
(cat. #556430 purified mouse anti-p21 monoclonal antibody, BD Biosciences, tively (Figure 1D). These data clearly suggest that preventive
San Diego, CA) or anti-PCNA (cat. #M0879 mouse monoclonal anti-PCNA
antibody, Dako, Carpinteria, CA) antibody. In each case, negative controls
efficacy of silibinin against UV-caused skin tumor initiation
were treated only with PBS under identical conditions. The sections were then could be, in part, via an inhibition in UV-induced DNA
incubated with biotinylated anti-rabbit secondary antibody (in case of p53) or damage.
1460
Silibinin protects mouse skin against UV damage

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


Fig. 1. Silibinin decreases UV-caused thymine dimer positive cells in Fig. 2. Silibinin up-regulates UV-induced p53 and p21/Cip1 levels. Mice
epidermis. Mice were (A) unexposed, (B) exposed with UV (180 mJ/cm2 ) or were (A) unexposed, (B) exposed with UV (180 mJ/cm2 ) or (C) topically
(C) topically applied with 9 mg silibinin/200 ml acetone/mouse 30 min prior applied with 9 mg silibinin/200 ml acetone/mouse 30 min prior to UV
to UV exposure, and killed 1 h thereafter. (A--C) Skin was removed, fixed, exposure, and killed 8 h thereafter. Skin was processed conventionally for
paraffin-embedded and processed for immunohistochemical analysis of immunohistochemical analysis (A--C for p53) as described in the Materials
thymine dimer positive cells as described in the Materials and methods. and methods. Percent p53 (D) and p21/Cip1 (E) positive cells were calculated
(D) Percent thymine dimer positive cells were calculated as mean of five as mean of five randomly selected fields (400) from each skin sample. Data
randomly selected fields (400) from each skin sample. Data shown are shown are mean  SE of 25 fields/5 mice/group. Sb, Silibinin;  P 5 0.001.
mean  SE of 25 fields/5 mice/group. Sb, Silibinin;  P 5 0.001.

induces apoptosis when the damage is severe (28--30). Based


Silibinin up-regulates UV-induced activation of p53-p21/Cip1 on our findings showing that silibinin strongly reduces UV-
cascade caused thymine dimer positive cells, we next focused our
p53 is known to play a crucial role in DNA damage response efforts on assessing the effect of silibinin on UV-caused
where it arrests cell proliferation when the damage is mild or p53 accumulation. As shown in Figure 2, compared with a
1461
S.Dhanalakshmi et al.

negligible immunostaining for p53 in unexposed mouse epi-


dermis, a large number of p53-positive cells were observed in
the epidermis at 8 h after a single UV exposure of mice. These
results are consistent with an earlier report showing p53 accu-
mulation in mouse epidermis following 8--12 h of UV exposure
at an identical dose used in the present study (26). Silibinin
topical application prior to or immediately after UV exposure,
however, resulted in a further increase in p53-positive cells
(Figure 2A--C). Overall, in quantitative analysis, compared
with 0.81  0.32% p53-positive cells in unexposed control
epidermis, 8 h after UV exposure resulted in 23.8  1.8% p53-
positive cells, which were further increased by pre- or post-
topical application of silibinin to 36.4  2.1 and 36.9  1.2%
(P 5 0.001) p53-positive cells, respectively, accounting for
~1.6-fold increase (Figure 2D).

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


UV-induced increase in p53 protein accumulation is known to
enhance the synthesis of p21/Cip1 protein, which plays a crucial
role in the adaptive responses after UV exposure of the skin by
inhibiting cell proliferation (29). Based on our finding that
silibinin further enhances UV-induced p53-positive cells in epi-
dermis, same samples were also analyzed for p21/Cip1-positive
cells. Compared with almost no immunostaining in unexposed
controls, a strong immunoreactivity for p21/Cip1-positive cells
was observed in the epidermis at 8 h after a single UV irradiation
at 180 mJ/cm2 dose, which, similar to p53 results, was further
enhanced by pre- or post-application of silibinin (data not
shown). In quantitative estimates, UV exposure showed 9.9 
0.9% p21/Cip1-positive cells in epidermis compared with 0.34
 0.09% in unexposed controls; however, pre- or post-UV
silibinin treatment resulted in 21.0  1.8 and 18.2  1.8% p21/
Cip1-positive cells, respectively, accounting for ~2-fold increase
(P 5 0.001) over UV-alone group (Figure 2E). Together, the
data shown in Figures 1 and 2 clearly suggest that silibinin
reduces UV-caused thymine dimer positive cells and further
increases UV-activated p53-p21/Cip1 cascade in mouse epider-
mis, which might be involved in an overall protection against
UV-caused biological responses such as cell proliferation and
apoptosis induction. Accordingly, we next assessed the effect of
silibinin on these two biological endpoints.

Silibinin inhibits UV-induced cell proliferation in SKH-1


hairless mouse skin
As shown in Figure 3, compared with unexposed controls, a
single UV irradiation at the dose of 180 mJ/cm2 resulted in a
strong PCNA staining within 1 h of the irradiation; however,
topical application of silibinin prior to or immediately after UV
irradiation resulted in a strong inhibition in UV-induced PCNA-
positive cells in the epidermis (Figure 3A--C). A quantitative
Fig. 3. Silibinin inhibits UV-induced cell proliferation as observed by PCNA
analysis of the PCNA immunostaining showed that UV irradia- immunostaining. Mice were (A) unexposed, (B) exposed with UV (180 mJ/
tion caused 22.4, 18.4, 19.6 and 16.1% PCNA-positive cells at 1, cm2 ) or (C) topically applied with 9 mg silibinin/200 ml acetone/mouse 30 min
4, 8 and 12 h post-UV irradiation as compared with 3.8% prior to UV exposure and killed 1 h thereafter. Skin was removed, fixed,
PCNA-positive cells in un-irradiated control epidermis, respect- paraffin-embedded and processed for immunohistochemical analysis of
PCNA as described in the Materials and methods. (D) Under identical
ively (Figure 3D). However, topical application of silibinin at experimental conditions and treatments, mice were killed at 1, 4, 8 and 12 h
9 mg dose in 200 ml acetone/mouse, 30 min prior to or immedi- after UV exposure, and skin tissue samples were subjected to PCNA staining
ately after UV exposure resulted in a strong and statistically as described in the Materials and methods. Percent PCNA-positive cells were
significant (P 5 0.005 to P 5 0.001) inhibition in UV-induced calculated as mean of five randomly selected fields (400) from each skin
sample. Data shown are mean  SE of 25 fields/5 mice/group. Sb, Silibinin.
cell proliferation at all the time points studied, and accounted for
40--52 and 20--40% inhibition, respectively (Figure 3D).
Silibinin inhibits UV-induced apoptotic and sunburn cell p53-p21 accumulation, silibinin treatment prior to or immedi-
formation ately after UV exposure also significantly inhibited UV-
Consistent with its effect on a reduction in UV-caused thymine induced apoptosis (Figure 4A--D). As observed by TUNEL
dimer positive cells, and a further increase in UV-activated staining, compared with 2.9  0.73% (Figure 4A) apoptotic
1462
Silibinin protects mouse skin against UV damage

which exposure of mouse skin to UV resulted in 10.2 


0.94% (P 5 0.001) sunburn cells as compared with 0.88 
0.16% in un-irradiated control (Figure 4E). Consistent to
TUNEL staining results, the UV-caused sunburn cell forma-
tion was inhibited significantly to 5.0  0.37 and 6.8  0.62%
(P 5 0.001) sunburn cells, respectively, by pre- or post-topical
application of silibinin (Figure 4E).

Discussion
The central finding in the present study is that silibinin protects
SKH-1 hairless mouse skin from UV-induced DNA damage,
and that UV-caused cell proliferation and apoptotic sunburn
cell formation are prevented by silibinin possibly via further

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


induction in p53-p21/Cip1 cascade. It is also important to
emphasize here that we employed two different silibinin topi-
cal application protocols in which silibinin was applied 30 min
prior to UV exposure or immediately thereafter. This approach
was important to dissect out the silibinin efficacy against UV-
caused skin damage independent of its sunscreen effects. The
findings reported here clearly support our conclusion that
silibinin efficacy against UV-caused skin damage and photo-
carcinogenesis is not only due to its sunscreen effect.
One of the most important characteristics of UV-caused
carcinogenesis is DNA damage and mutagenesis, and thymine
dimers are known as `hot spots' of UV mutagenesis (20). In
our study, silibinin treatment resulted in a remarkable decrease
in UV-caused thymine dimer positive cells. Analysis of the
mismatch repair enzyme MSH2 showed that silibinin treat-
ment does not result in an increase in MSH2 levels (data not
shown). This suggests the possible involvement of other mis-
match repair enzymes. Further, there are some reports showing
that DNA mismatch repair system is inactivated by oxidative
stress (31). Strong anti-oxidant properties of silibinin reported
earlier by us in mouse skin studies (15,16,18) suggest that
suppression of oxidative stress by silibinin could be one of
the possible mechanisms that resulted in the activation of
repair enzymes much earlier as compared with UV alone;
thereby it is possible that thymine dimers were removed
much earlier in silibinin-treated animals than 1 h, the time
point used in our study. More studies are needed in future as
a function of time employing time points earlier than 1 h,
specifically between 5 and 60 min, to assess whether silibinin
causes a faster repair of UV-caused DNA damage ultimately
leading to a strong reduction in thymine dimer positive cells in
Fig. 4. Inhibitory effect of silibinin on UV-induced apoptosis. Mice were epidermis as observed in the present study at 1 h after UV
(A) unexposed, (B) exposed with UV (180 mJ/cm2 ) or (C) topically applied exposure of SKH-1 mouse skin. The other possibilities could
with 9 mg silibinin/200 ml acetone/mouse 30 min prior to UV exposure, and be that silibinin protects epidermal cells from UV-caused
killed 12 h thereafter. Skin was removed, fixed, paraffin-embedded and
processed for the analysis of apoptotic cells by TUNEL (A--C) as well as for thymine dimer positive cells by modulating DNA repair
the analysis of sunburn cells by H&E staining as described in the Materials enzymes other than MSH2 and/or by an alteration in ATM/
and methods. (D) Percent TUNEL-positive cells and (E) sunburn cells were ATR pathways. In this regard, recent studies have shown the
calculated as mean of five randomly selected fields (400) from each skin role and involvement of ATM/ATR in UV-induced DNA
sample. Data shown are mean  SE of 25 fields/5 mice/group. Sb, Silibinin;

P 5 0.001.
damage (32), and accordingly future studies are needed to
assess silibinin effect on these pathways as an upstream
response for its efficacy against UV-caused thymine dimer
cells in unexposed controls, a single UV irradiation resulted in positive cells in epidermis.
30.5  1.34% (Figure 4B) TUNEL-positive apoptotic cell p53 up-regulation following UV irradiation is known to
population. However, pre- or post-application of silibinin protect the cells from UV-caused cellular damage/s via several
showed only 9.0  0.81 (Figure 4C) and 20.6  1.08% apop- mechanisms including an induction in cell cycle arrest via
totic cells accounting for 70 and 32% decrease (P 5 0.001), transcriptional activation of p21/Cip1, or by inducing apopto-
respectively (Figure 4A--D). UV-caused apoptotic/sunburn sis through an activation of apoptotic proteins bax, Fas/Apo-1,
cell formation was further confirmed by H&E staining, in DR5 or by down-regulation of anti-apoptotic proteins such as
1463
S.Dhanalakshmi et al.

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


Fig. 5. Proposed mechanisms for the protective effect of silibinin on UV-induced photodamage in epidermal cells. Silibinin protects SKH-1 mouse epidermis
from DNA damaging effect of UV such as thymine dimer positive cells, thereby decreasing UV-caused apoptotic/sunburn cells. Further, silibinin inhibits
UV-induced epidermal cell proliferation via decreasing PCNA possibly through activation of p53-p21/Cip1.

bcl-2 and cellular inhibitor of apoptosis protein 2 (c-IAP2) study are consistent with an early report regarding apoptotic
(23,29). Some chemopreventive agents have been shown to nature of the sunburn cells (37).
selectively induce apoptosis of UV-damaged cells by up- Use of conventional chemical sunscreens has been an effec-
regulation of p53 (14,33). Earlier reports have shown that the tive measure in protecting skin against UV-caused damages and
level of p53 accumulation depends on the extent of DNA NMSC; several cancer chemopreventive agents, however, have
damage and that lower levels of p53 correspond to cell cycle also been shown to protect cells from UV by acting as a
arrest (34). In our study, silibinin treatment resulted in a further sunscreen (10,38,39). However, sunscreen usage as an only
increase in UV-induced p53 accumulation with a concomitant measure is not adequate because of infrequent use, incomplete
increase in p21/Cip1 protein levels, which is in accord with the protection and associated toxicity (7). In this regard, topical
inhibition of UV-induced cell proliferation and apoptosis by application of antioxidants has been shown to be beneficial
silibinin, suggesting their possible role in cell growth inhibi- against skin cancer as well as associated hazards such as
tion rather than apoptosis induction (Figure 5). photoaging. It is important to emphasize here that silibinin is
UV-induced increase in cell proliferation is an early neces- a flavonoid antioxidant with no known toxicity in both animal
sary event associated with UV-caused carcinogenesis that helps and human studies. Further, since the chemopreventive efficacy
the initiated cells to proceed further into cell cycle (24); this of silibinin has been well studied and established in several
response, however, could be prevented by arresting the cells at epithelial cancer models (16--18,40,41), it could be a potent
G1 or S phase of the cell cycle (35). We observed that UV- chemopreventive agent against NMSCs as well as UV-caused
induced PCNA-positive cells were strongly inhibited by silibi- skin damages and photoaging. In the present study, as pre-
nin treatment. It has been reported that UV-induced cell treatment of silibinin showed a moderately better protective
proliferation reaches the maximum level as early as 1 h after effect (than its post-treatment) against some of the UV-caused
exposure, and we observed that silibinin treatment showed a alterations in SKH-1 mouse epidermis, a sunscreen effect may
strong inhibitory effect even at that early time point. In addi- be an additional factor in silibinin-afforded protection.
tion, the inhibitory effect of silibinin on UV-caused cell pro- In conclusion, the results of the present study suggest that
liferation was sustained as observed up to 12 h after exposure. silibinin protects SKH-1 hairless mouse epidermis from UV-
These results suggest that inhibiting cell proliferation could be induced cell proliferation and apoptosis through a reduction in
one of the mechanisms by which silibinin protects damaged DNA damage and via an activation of p53-p21/Cip1 cascade.
cells from entering the cell cycle, thereby providing damaged These results warrant the development of silibinin as a phar-
cells additional time for repair and preventing their entry into macological non-toxic chemopreventive agent against human
apoptotic pathway in case the damage is severe. This sugges- NMSC and photoaging. In addition, more mechanistic studies
tion is in accord with the observation that silibinin treatment are needed in future to define the protective effect of silibinin
further up-regulates p21/Cip1 levels, which is known to inhibit on UV-induced DNA damage and apoptosis, and their biolo-
cell proliferation by a direct binding with PCNA (36). gical significance in an overall efficacy of silibinin against
An earlier study by us has shown that topical application of UV-caused skin damages and photocarcinogenesis.
silymarin strongly protects against UV-caused sunburn cell
formation in SKH-1 hairless mice (15). The results of the
present study showing the inhibitory effect of silibinin on Acknowledgement
UV-caused sunburn cell formation further support the previous
finding. In addition, the TUNEL staining results in the present This work was supported by USPHS grant CA64514.

1464
Silibinin protects mouse skin against UV damage

References 22. Li,G. and Ho,V.C. (1998) p53-dependent DNA repair and apoptosis
respond differently to high- and low-dose ultraviolet radiation. Br. J.
1. Gloster,H.M.,Jr and Brodland,D.G. (1996) The epidemiology of skin Dermatol., 139, 3--10.
cancer. Dermatol. Surg., 22, 217--226. 23. Ouhtit,A., Gorny,A., Muller,H.K., Hill,L.L., Owen-Schaub,L. and
2. Gilchrest,B.A. (1996) A review of skin ageing and its medical therapy. Ananthaswamy,H.N. (2000) Loss of Fas-ligand expression in mouse
Br. J. Dermatol., 135, 867--875. keratinocytes during UV carcinogenesis. Am. J. Pathol., 156, 201--207.
3. Cruz,J.R., Leverkus,P.D.M., Dougherty,I., Gleason,M.J., Eller,M., 24. Huang,L.C., Clarkin,K.C. and Wahl,G.M. (1996) Sensitivity and selectiv-
Yaar,M. and Gilchrest,B.A. (2000) Thymidine dinucleotides inhibit ity of the DNA damage sensor responsible for activating p53-dependent
contact hypersensitivity and activate the gene for tumor necrosis factor G1 arrest. Proc. Natl Acad. Sci. USA, 93, 4827--4832.
alpha1. J. Invest. Dermatol., 114, 253--258. 25. Mullauer,L., Gruber,P., Sebinger,D., Buch,J., Wohlfart,S. and Chott,A.
4. Kulms,D. and Schwarz,T. (2000) Molecular mechanisms of UV- (2001) Mutations in apoptosis genes: a pathogenetic factor for human
induced apoptosis. Photodermatol. Photoimmunol. Photomed., 16, disease. Mutat. Res., 488, 211--231.
195--201. 26. Lu,Y.-P., Lou,Y.-R., Yen,P., Mitchell,D., Huang,M.-T. and Conney,A.H.
5. McKenzie,R., Connor,B. and Bodeker,G. (1999) Increased summertime (1999) Time course for early adaptive responses to ultraviolet B light in
UV radiation in New Zealand in response to ozone loss. Science, 285, the epidermis of SKH-1 mice. Cancer Res., 59, 4591--4602.
1709--1711. 27. Tornaletti,S. and Pfeifer,G.P. (1996) UV damage and repair mechanisms
6. Wolf,P., Donawho,C.K. and Kripke,M.L. (1994) Effect of sunscreens in mammalian cells. Bioessays, 18, 221--228.
on UV radiation-induced enhancement of melanoma growth in mice. 28. Liebermann,D.A., Hoffman,B. and Steinman,R.A. (1995) Molecular

Downloaded from https://academic.oup.com/carcin/article/25/8/1459/2475943 by guest on 26 June 2022


J. Natl Cancer Inst., 86, 99--105. controls of growth arrest and apoptosis: p53-dependent and independent
7. Pinnell,S.R. (2003) Cutaneous photodamage, oxidative stress, and topical pathways. Oncogene, 11, 199--210.
antioxidant protection. J. Am. Acad. Dermatol., 48, 1--19. 29. Ponten,F., Berne,B., Ren,Z.P., Nister,M. and Ponten,J. (1995) Ultraviolet
8. Bickers,D.R. and Athar,M. (2000) Novel approaches to chemoprevention light induces expression of p53 and p21 in human skin: effect of sunscreen
of skin cancer. J. Dermatol., 27, 691--695. and constitutive p21 expression in skin appendages. J. Invest. Dermatol.,
9. Bode,A.M. and Dong,Z. (2000) Signal transduction pathways: targets for 105, 402--406.
chemoprevention of skin cancer. Lancet Oncol., 1, 181--188. 30. Mass,P., Hoffmann,K., Gambichler,T., Altmeyer,P. and Mannherz,H.G.
10. Conney,A.H., Lou,Y.R., Xie,J.G., Osawa,T., Newmark,H.L., Liu,Y., (2003) Premature keratinocyte death and expression of marker proteins of
Chang,R.L. and Huang,M.T. (1997) Some perspectives on dietary apoptosis in human skin after UVB exposure. Arch. Dermatol. Res., 295,
inhibition of carcinogenesis: studies with curcumin and tea. Proc. Soc. 71--79.
Exp. Biol. Med., 216, 234--245. 31. Chang,C.L., Marra,G., Chauhan,D.P., Ha,H.T., Chang,D.K.,
11. Hong,J.T., Kim,E.J., Ahn,K.S., Jung,M., Yun,Y.P., Park,Y.K. and Ricciardiello,L., Randolph,A., Carethers,J.M. and Boland,C.R. (2002)
Lee,S.H. (2001) Inhibitory effect of glycolic acid on ultraviolet-induced Oxidative stress inactivates the human DNA mismatch repair system.
skin tumorigenesis in SKH-1 hairless mice and its mechanism of action. Am. J. Physiol. Cell. Physiol., 283, C148--154.
Mol. Carcinog., 31, 152--160. 32. Goodarzi,A.A., Block,W.D. and Lees-Miller,S.P. (2003) The role of ATM
12. Lu,Y.P., Lou,Y.R., Lin,Y., Shih,W.J., Huang,M.T., Yang,C.S. and and ATR in DNA damage-induced cell cycle control. Prog. Cell Cycle
Conney,A.H. (2001) Inhibitory effects of orally administered green tea, Res., 5, 393--411.
black tea, and caffeine on skin carcinogenesis in mice previously treated 33. Liu,G.Y., Frank,N., Bartsch,H. and Lin,J.K. (1997) Induction of apoptosis
with ultraviolet B light (high-risk mice): relationship to decreased tissue by thiuramdisulfides, the reactive metabolites of dithiocarbamates, through
fat. Cancer Res., 61, 5002--5009. coordinative modulation of NFkappaB, c-fos/c-jun, and p53 proteins. Mol.
13. Huang,M.T., Xie,J.G., Wang,Z.Y., Ho,C.T., Lou,Y.R., Wang,C.X., Carcinog., 19, 74--82.
Hard,G.C. and Conney,A.H. (1997) Effects of tea, decaffeinated tea, and 34. Chen,X., Ko,L.J., Jayaraman,L. and Prives,C. (1996) p53 levels,
caffeine on UVB light-induced complete carcinogenesis in SKH-1 mice: functional domains, and DNA damage determine the extent of the
demonstration of caffeine as a biologically important constituent of tea. apoptotic response of tumor cells. Genes Dev., 10, 2438--2451.
Cancer Res., 57, 2623--2629. 35. Herzinger,T., Funk,J.O., Hillmer,K., Eick,D., Wolf,D.A. and Kind,P.
14. Lu,Y.P., Lou,Y.R., Li,X.H., Xie,J.G., Brash,D., Huang,M.T. and (1995) Ultraviolet B irradiation-induced G2 cell cycle arrest in human
Conney,A.H. (2000) Stimulatory effect of oral administration of green keratinocytes by inhibitory phosphorylation of the cdc2 cell cycle kinase.
tea or caffeine on ultraviolet light-induced increases in epidermal wild- Oncogene, 10, 2151--2156.
type p53, p21(WAF1/CIP1), and apoptotic sunburn cells in SKH-1 mice. 36. Ducoux,M., Urbach,S., Baldacci,G., Hubscher,U., Koundrioukoff,S.,
Cancer Res., 60, 4785--4791. Christensen,J. and Hughes,P. (2001) Mediation of proliferating cell
15. Katiyar,S.K., Korman,N.J., Mukhtar,H. and Agarwal,R. (1997) Protective nuclear antigen (PCNA)-dependent DNA replication through a conserved
effects of silymarin against photocarcinogenesis in a mouse skin model. p21(Cip1)-like PCNA-binding motif present in the third subunit of human
J. Natl Cancer Inst., 89, 556--566. DNA polymerase delta. J. Biol. Chem., 276, 49258--49266.
16. Zhao,J., Sharma,Y. and Agarwal,R. (1999) Significant inhibition by the 37. Gniadecki,R., Hansen,M. and Wulf,H.C. (2000) Resistance of senescent
flavonoid antioxidant silymarin against 12-O-tetradecanoylphorbol 13- keratinocytes to UV-induced apoptosis. Cell. Mol. Biol., 46, 121--127.
acetate-caused modulation of antioxidant and inflammatory enzymes, and 38. Record,I.R. and Dreosti,I.E. (1998) Protection by black tea and green tea
cyclooxygenase 2 and interleukin-1alpha expression in SENCAR mouse against UVB and UVA ‡ B induced skin cancer in hairless mice. Mutat.
epidermis: implications in the prevention of stage I tumor promotion. Res., 422, 191--199.
Mol. Carcinog., 26, 321--333. 39. Bair,W.B., Hart,N., Einspahr,J., Liu,G., Dong,Z., Alberts,D. and
17. Singh,R.P., Tyagi,A.K., Zhao,J. and Agarwal,R. (2002) Silymarin inhibits Bowden,T. (2002) Inhibitory effects of sodium salicylate and acetylsa-
growth and causes regression of established skin tumors in SENCAR mice licylic acid on UVB-induced mouse skin carcinogenesis. Cancer
via modulation of mitogen-activated protein kinases and induction of Epidemiol. Biomark. Prev., 11, 1645--1652.
apoptosis. Carcinogenesis, 23, 499--510. 40. Bhatia,N., Zhao,J., Wolf,D.M. and Agarwal,R. (1999) Inhibition of human
18. Singh,R.P. and Agarwal,R. (2002) Flavonoid antioxidant silymarin and carcinoma cell growth and DNA synthesis by silibinin, an active
skin cancer. Antioxid. Redox. Signal., 4, 655--663. constituent of milk thistle: comparison with silymarin. Cancer Lett.,
19. Lahiri-Chatterjee,M., Katiyar,S.K., Mohan,R.R. and Agarwal,R. (1999). A 147, 77--84.
flavonoid antioxidant, silymarin, affords exceptionally high protection 41. Singh,R.P., Dhanalakshmi,S., Tyagi,A.K., Chan,D.C., Agarwal,C. and
against tumor promotion in the SENCAR mouse skin tumorigenesis Agarwal,R. (2002) Dietary feeding of silibinin inhibits advance human
model. Cancer Res., 59, 622--632. prostate carcinoma growth in athymic nude mice and increases plasma
20. Kanjilal,S. and Ananthaswamy,H.N. (1996) In Weber,R., Miller,M. and insulin-like growth factor-binding protein-3 levels. Cancer Res., 62,
Goepfert,H. (eds) Basal and Squamous Cell Skin Cancers of the Head and 3063--3069.
Neck. Williams and Wilkins, Baltimore, pp. 25--26.
21. Pourzand,C. and Tyrrell,R.M. (1999) Apoptosis, the role of oxidative
stress and the example of solar UV radiation. Photochem. Photobiol., 70, Received January 22, 2004; revised February 25, 2004;
380--390. accepted March 9, 2004

1465

You might also like