You are on page 1of 9

European Journal of Pharmacology 559 (2007) 227 – 235

www.elsevier.com/locate/ejphar

Mechanisms underlying the inhibitory actions of the pentacyclic


triterpene α-amyrin in the mouse skin inflammation induced by
phorbol ester 12-O-tetradecanoylphorbol-13-acetate
Rodrigo Medeiros a,1 , Michel F. Otuki a,1,2 , Maria Christina W. Avellar b , João B. Calixto a,⁎
a
Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
b
Departamento de Farmacologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
Received 4 May 2006; received in revised form 5 December 2006; accepted 8 December 2006
Available online 29 December 2006

Abstract

The present study evaluated some of the mechanisms through which α-amyrin, a pentacyclic triterpene isolated from Protium Kleinii and other
plants, exerts its effects against 12-O-tetradecanoylphorbol-acetate (TPA)-induced skin inflammation in mice. Topical application of α-amyrin
(0.1–1 mg/ear) dose-dependently inhibited TPA-induced increase of prostaglandin E2 (PGE2) levels. In contrast with the selective cyclooxygenase
(COX)-1 SC560 [5-(4-chlorophenyl)-1-(4-methoxyphenyl)-3-trifluoromethylpyrazole] or COX-2 rofecoxib inhibitors, α-amyrin failed to alter
either COX-1 or COX-2 activities in vitro. Western blot analysis revealed that α-amyrin dose-dependently inhibited TPA-induced COX-2
expression in the mouse skin. The evaluation of nuclear factor-κB (NF-κB) pathway revealed that topical treatment with α-amyrin is able to
prevent IκBα degradation, p65/RelA phosphorylation and NF-κB activation. Moreover, α-amyrin given topically dose-dependently inhibited the
activation of upstream protein kinases, namely extracellular signal-regulated protein kinase (ERK), p38 mitogen-activated protein kinase (MAPK)
and protein kinase C (PKC)α, following topical TPA treatment. Collectively, present results suggest that topical skin application of α-amyrin
exerts a strong and rapid onset inhibition of TPA-induced inflammation. These effects seem to be associated with the suppression of skin PGE2
levels by mechanisms involving the suppression of COX-2 expression, via inhibition of upstream protein kinases – namely ERK, p38 MAPK and
PKCα – and blocking of NF-κB activation. These results indicate that α-amyrin-derivative could be potentially relevant for the development of a
topical agent for the management of inflammatory diseases.
© 2006 Elsevier B.V. All rights reserved.

Keywords: α-amyrin; Skin inflammation; PGE2; NF-κB; TPA; COX-2

1. Introduction and inflammatory systems play a pivotal role. Thus, a large


production of inflammatory mediators, such as cytokines
The skin is the largest organ in the body and one of its main (tumor necrosis factor-α, TNF-α; interleukin-(IL)1β) chemo-
functions is to protect the body from environmental and kines (monocyte chemotactic protein-1, MCP-1; IL-8), cell
endogenous noxious conditions, such as injury, infection and adhesion molecules (intercellular adhesion molecule-1, ICAM-
inflammation. In the pathogenesis of skin diseases, the immune 1; vascular cell adhesion molecule-1, VCAM-1), growth factors
and proteases, are produced and released at the site of skin
⁎ Corresponding author. Departamento de Farmacologia, Centro de Ciências injury (Homey, 2005; Okayama, 2005; Pivarcsi and Homey,
Biológicas, Universidade Federal de Santa Catarina, Campus Universitário 2005; Numerof et al., 2005).
Trindade, 88049-900, Florianópolis, SC, Brazil. Tel.: +55 48 33319491; fax: There is a substantial amount of experimental evidence
+55 48 32224164. indicating that the mitogen activated protein kinases (MAPKs)
E-mail addresses: calixto@farmaco.ufsc.br, calixto3@terra.com.br
(J.B. Calixto).
and nuclear factor-κB (NF-κB) have a critical role in both acute
1
The first two authors contributed equally to this work. and chronic inflammatory processes (Karin, 2006; Kyriakis and
2
Current address: Departamento de Farmacologia, Setor de Ciências Avruch, 2001). The stimulation of these cascades occurs in
Biológicas, Universidade Federal do Paraná, PR, Brazil. response to several stimuli, such as mechanical stress, inflam-
0014-2999/$ - see front matter © 2006 Elsevier B.V. All rights reserved.
doi:10.1016/j.ejphar.2006.12.005
228 R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235

matory cytokines, heat and chemical shock, or bacterial 2. Materials and methods
products, all of them also capable of stimulating cyclooxygen-
ase (COX)-2 inductions. It has been shown that binding of NF- 2.1. Reagents
κB to its specific cognate κB sites is functionally important for
COX-2 induction by the prototype tumor promoter 12-O- The following substances were used: TPA, α-amyrin,
tetradecanoylphorbol-13-acetate (TPA) (Chen et al., 2000). In dexamethasone acetate, phosphate-buffered saline (PBS), 4-
addition, the participation of activator protein-1 (AP-1) in cox-2 (2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES),
gene expression was also reported (Chun et al., 2004). indomethacin, Tween-20, Triton-X 100, glycerol, phenyl-
Accumulating evidence indicates that IκB kinase (IKK)- methylsulphonyl fluoride, leupeptin, pepstatin A, aprotinin,
dependent IκB degradation is a pre-requisite for NF-κB soybean trypsin inhibitor, benzamidine, sodium orthovanadate,
activation, and p38 mitogen-activated protein kinase (MAPK), β-glycerophosphate, sodium fluoride, dithiothreitol, β-mercap-
extracellular signal-regulated kinase (ERK) and protein kinase toethanol, dimethyl sulfoxide (DMSO), bromophenol blue
C (PKC) are also involved in NF-κB activation and COX-2 (Sigma Aldrich Co., São Paulo, Brazil), ethylenediamine
expression in response to TPA (Schulze-Osthoff et al., 1997; tetraacetic acid (EDTA), Tris–HCl, MgCl2, NaCl, KCl, sodium
Janssen-Heininger et al., 2001). acetate, methanol, acetone, absolute ethanol, Rofecoxib (Merck
α-Amyrin (Fig. 1) is a naturally-occurring pentacyclic and Co., Inc., Whitehouse Station, USA), NF-κB double-
triterpene found in various plants that are widespread in tropical stranded consensus oligonucleotide probe, T4 polynucleotide
regions (eg., Protium kleinii, Protium heptaphyllum and Mol- kinase (Promega, Madison, USA), poly(dI–dC), [γ32P]ATP,
denhawera nutans) (do Vale et al., 2005; Oliveira et al., 2005; protein A-sepharose (GE Healthcare, São Paulo, Brazil), 5-(4-
Otuki et al., 2005b). Some experimental evidence shows that α- chlorophenyl)-1-(4-methoxyphenyl)-3-trifluoromethylpyrazole
amyrin exhibits systemic antinociceptive, antiinflamatory, (SC560) (Boehringer-Ingelheim, Ingelheim, Germany), anti-
antipruritic, hepatoprotective and gastroprotective properties COX-2, anti-α-actin, anti-IκBα, anti-PKCα, anti-ERK, anti-
when assessed in vivo (Kweifio-Okai et al., 1994; Recio et al., phosphorylated-ERK, anti-p38 MAPK, anti-phosphorylated-
1995; Oliveira et al., 2004a,b). We have shown previously that p38 MAPK, anti-p65 NF-κB antibodies (Santa Cruz Biotech.
the mixture of triterpene α-amyrin and β-amyrin produces Inc., Santa Cruz, USA) and anti-phosphoserine antibody
consistent peripheral, spinal, and supraspinal antinociception in (Calbiochem–Novabiochem Co., San Diego, USA).
rodents, especially when assessed in inflammatory models of
pain. The mechanisms involved in the actions of this mixture 2.2. Animals
seem to involve the inhibition of both protein kinase A- and
PKC-dependent pathways (Otuki et al., 2005a). Furthermore, Experiments were conducted using male Swiss mice (20–30 g)
our previous study has also demonstrated that the topical kept in a controlled room temperature (22 ± 1 °C) and humidity
application of pentacyclic triterpene α-amyrin is capable of (60 to 80%) under a 12 h:12 h light–dark cycle (lights on 06:00 h).
reducing, in a dose-dependent manner, three important events All the procedures used in the present study complied with the
related to the TPA-induced skin inflammatory response: “Principles of laboratory animal care” from NIH publication No.
oedema formation, migration of polymorphonuclear leuko- 85-23 and were approved by the Institutional Ethics Committee of
cytes, and increase in tissue IL-1β levels (Otuki et al., 2005b). the Federal University of Santa Catarina (protocol numbers 262/
However, the mechanisms underlying these topical anti- CEUA and 23080.035334/2003-16/UFSC).
inflammatory properties of triterpene pentacyclic α-amyrin
are so far, only partially understood. 2.3. TPA induced skin inflammation
The aim of the present study was therefore to investigate
further, using functional, biochemical and molecular proce- The skin inflammation was induced in the right ear by the
dures, some of the mechanisms through which α-amyrin exerts topical application of 2.5 μg/ear of TPA dissolved in 20 μl of
its topical anti-inflammatory action on TPA-induced skin acetone. Control animals received the same volume of acetone
inflammation in mice. in the right ear. Both α-amyrin (0.1–1.0 mg/ear) and
dexamethasone (0.05 mg/ear, used as a positive control) were
dissolved in 20 μl of acetone, and were applied topically in
conjunction with TPA. Control animals received the same
volume of vehicle. Animals were sacrificed at 1 or 6 h after
TPA-treatment. Tissues were frozen in liquid N2, and stored at
− 70 °C until use.

2.4. Cyclooxygenase-1/cyclooxygenase-2 ovine enzyme assay

The activity COX-1/COX-2 in vitro was carried out by using


a colorimetric assay for detecting COX inhibitors (Cayman
Chemical Company, Ann Arbor, USA), according to the
Fig. 1. Structure of α-amyrin. manufacturer's protocol. The reaction was performed in the
R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235 229

presence of vehicle (DMSO, 0.5%) or α-amyrin (1–30 μM). for 2 h at room temperature with 1:1000 dilutions of primary
SC560 and rofecoxib were used as positive controls for the antibodies for COX-2, β-actin, IκBα, PKCα, ERK or phosphory-
inhibition COX-1 and COX-2 activity, respectively. Data is lated-ERK and for 16 h at 4 °C with 1:1000 dilutions of primary
expressed as the percentage of the activity in relation to the antibodies for p38 MAPK and phosphorylated-p38. Blots were
control group. washed three times with TBST at 5 min intervals followed by
incubation with 1:5000 dilution of appropriate horseradish
2.5. Measurement of prostaglandin E2 levels peroxidase conjugated secondary antibodies for 1 h and again
washed in TBST for three times. The transferred proteins were
The tissue PGE2 levels were determined 6 h after TPA visualized with an enhanced chemiluminescence (ECL) detection
application in 6 mm punch-biopsies obtained from the mouse kit according to the manufacturer's instructions (GE Healthcare).
ears. The tissues were homogenized in 100 mM phosphate
buffer (pH 7.4) containing 1 mM EDTA and 10 μM 2.8. Immunoprecipitation of p65 NF-κB
indomethacin. After addition of ethanol, the sample was
centrifuged at 1500 ×g for 10 min. Then 50 mM citrate buffer Equivalent amounts of cytosolic extracts (200 μg for each
(pH 3.5) was added to the supernatant, after which it was sample) were mixed with 40 μl of protein A-sepharose and 2 μl of
centrifuged again at 2500 ×g for 10 min. The resulting anti-p65 NF-κB antibody, and left overnight at 4 °C with
supernatant was applied to a 6 ml Sep-Pak C18 cartridge continuous shaking. Immunocomplexes were washed three times
(Waters Associate, Milford, USA) pre-activated with 5 ml with 500 μl of ice-cold buffer (10 mM Tris–HCl pH 7.5, 1 M
methanol and then with 5 ml distilled water. The cartridge NaCl, 0.2% Triton-X 100 and 2 mM EDTA), mixed with 40 μl of
was rinsed with 5 ml distilled water, followed by 5 ml SDS gel loading buffer and then boiled for 3 min. Samples were
hexane. The final eluate was obtained with 5 ml ethyl acetate then subjected to western blot analysis as described above. Proteins
containing 1% methanol and dried under N2 stream. After were incubated overnight with 0.1 μg/ml of anti-phosphoserine
dissolving the residue in a small amount of assay buffer, the antibody. Following the detection of the phosphoserine, the
prostaglandin E2 concentration was measured with an ELISA membranes were washed, stripped and then re-probed with anti-
kit (Cayman Chemical Company) according to the manufac- p65 NF-κB antibody (1:1000).
turer's instructions.
2.9. Electrophoretic Mobility Shift Assay (EMSA)
2.6. Preparation of cytosolic and membrane fractions
Nuclear protein extraction was performed as previously
Protein extraction was performed as previously described described (Medeiros et al., 2004). In brief, tissues were suspended
(Medeiros et al., 2004; Ferreira et al., 2005). In brief, tissues in 1 ml of ice-cold hypotonic lysis (10 mM HEPES, 1.50 mM
were homogenized in ice-cold buffer A (10 mM HEPES, pH MgCl2, 10 mM KCl, 0.5 mM PMSF, 1.5 μg/ml soybean trypsin
7.4; containing 1.5 mM MgCl2, 10 mM KCl, 1 mM inhibitor, 7 μg/ml pepstatin A, 5 μg/ml leupeptin, 0.1 mM
phenylmethylsulphonyl fluoride, 1 μg/ml leupeptin, 1 μg/ml benzamidine, and 0.5 mM DTT) and were then homogenized in a
pepstatin A, 1 μg/ml leupeptin, 1 μg/ml aprotinin, 1 mM Polytron (Brinkmann Instruments, Westbury, USA) for 1 min. The
sodium orthovanadate, 10 mM β-glycerophosphate, 50 mM homogenate was chilled on ice for 15 min, and then vigorously
sodium fluoride and 0.5 mM dithiothreitol). The homogenates shaken for 15 min in the presence of 10 μl of Nonidet P-40. The
were chilled on ice under vigorously shaken for 15 min. The nuclear fraction was precipitated by centrifugation at 1500 ×g for
membrane fraction was precipitated by centrifugation at 5 min. The nuclear pellet was resuspended in 100 μl of high salt
14,000 ×g for 60 min. The supernatant containing the cytosolic extraction buffer (20 mM HEPES (pH 7.9), 420 mM NaCl,
fraction was stored at − 70 °C until use. The membrane pellet 1.5 mM MgCl2, 0.2 mM EDTA, 25% (v/v) glycerol, 0.5 mM
was resuspended in 50 μl of buffer A containing 1% Triton X- PMSF, 1.5 μg/ml soybean trypsin inhibitor, 7 μg/ml pepstatin A,
100 and incubated under continuous shaking at 4 °C for 30 min. 5 μg/ml leupeptin, 0.1 mM benzamidine, and 0.5 mM DTT),
The membrane fraction was then centrifuged for 60 min at incubated under continuous shaking at 4 °C for 30 min, and then
14,000 ×g and the supernatant was aliquoted and stored at centrifuged for 15 min at 14,000 ×g. The supernatant was
− 70 °C. The protein concentration was determined using the aliquoted and stored at −70 °C. The protein concentration was
Bio-Rad protein assay kit (Bio-Rad, São Paulo, Brazil). determined using a protein assay kit (Bio-Rad).
For EMSAs, an NF-κB double-stranded consensus oligonu-
2.7. Western blot analysis cleotide probe (5′-AGTTGAGGGGACTTTCCCAGGC-3′) was
end-labeled with [γ-32P]ATP in the presence of T4 polynucleotide
Cell lysates (30 μg protein) were boiled in sodium dodecyl kinase (10 U) for 10 min at 37 °C. Unincorporated nucleotides
sulfate (SDS) sample buffer for 5 min before electrophoresis on were removed by passing the reaction mixture over a Sephadex G-
12% SDS-polyacrylamide gel. After transferring to a polyvinyli- 25 spin column (GE Healthcare). In a total volume of 20 μl,
dene fluoride (PVDF) membrane (GE Healthcare), the blots were nuclear extracts (10 μg) were incubated with gel shift binding
blocked with 5% fat-free dry milk-TBST buffer (tris-buffered buffer [10 mM Tris–HCl (pH 7.5), 1 mM MgCl2, 50 mM NaCl,
saline containing 0.5% Tween-20) for 1 h at room temperature and 0.5 mM DTT, 0.5 mM EDTA, 4% glycerol, and 1 μg of poly(dI–
then washed with TBST buffer. The membranes were incubated dC)] for 20 min at room temperature. Each sample was further
230 R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235

incubated for 30 min at room temperature with 25,000 cpm of


32
P-labeled NF-κB consensus oligonucleotide. Protein–DNA
complexes were resolved by nondenaturing 6% acrylamide/bis-
acrylamide (37.5/1) in 0.25 X Tris–borate/EDTA buffer at
150 V for 2 h. Finally, the gel was dried and exposed to an X-
ray film.

2.10. Software

All western blot and EMSA experiments were scanned to


acquire digital images using a Genius ColorPage Scanner (KYE
Systems Corp., Taipei, Taiwan). Digital images were processed
with the Photoshop software package (Adobe Systems, San
Jose, USA), complying with strict standards (Rossner and
Yamada, 2004). Band density measurements were made using
Scion Image software package (Scion Corporation, Maryland,
USA).

2.11. Statistical analysis


Fig. 3. Effect of α-amyrin on COX activity in vitro. The (A) COX-1 and (B)
COX-2 in vitro assay was carried out using a colorimetric assay for detection of
The results are presented as mean ± S.E.M. The statistical
COX inhibitors according to the manufacturer's protocol. α-Amyrin was tested
significance between the groups was assessed by means of one- at 10 or 30 μM concentration. Selective COX-1 SC560 (0.03 μM) and COX-2
way analysis of variance (ANOVA) followed by post-hoc rofecoxib (Rofe, 10 μM) inhibitors were used as a control. Figures are
Newman–Keuls test. The accepted level of significance for the representative of 3 similar experiments and each column represents the mean ±
tests was P < 0.05. All tests were carried out using the Statistica S.E.M. The asterisks denote the significance levels: ⁎⁎P < 0.01, compared with
vehicle (control) (one-way ANOVA followed by post-hoc Newman–Keuls test).
software package (StatSoft Inc., Tulsa, USA).

3. Results

3.1. α-Amyrin reduced TPA-induced PGE2 production in mouse


skin through inhibition of COX-2 expression, but not COX-1 or
COX-2 activity

High levels of prostaglandins are produced by COX enzymes


during the inflammatory response. As shown in Fig. 2, topical
application of TPA in the mouse ear induced a significant (about

Fig. 4. Effect of α-amyrin on TPA-induced COX-2 expression in mouse ear in


vivo. (A) Western blot analysis of COX-2 expression was performed on
Fig. 2. Effect of α-amyrin on TPA-induced PGE2 production in mouse skin in cytosolic extract 6 h after acetone (control, C) or TPA (2.5 μg/ear) treatment.
vivo. The PGE2 levels were measured 6 h after topical application of acetone Animals were topically treated, at same time of TPA administration, with vehicle
(control, C) or TPA (2.5 μg/ear) as described in the Material and methods (V), α-amyrin (0.1–1.0 mg/ear) or dexamethasone (0.05 mg/ear, Dexa).
section. Vehicle (V), α-amyrin (0.1–1.0 mg/ear) or dexamethasone (0.05 mg/ Immunoblot for β-actin was used as a loading control. (B) Quantification of
ear, Dexa) were administered topically in combination with TPA. Each column COX-2 protein expression normalized by β-actin. Each column represents the
represents the mean ± S.E.M. of 3 independent experiments. The asterisks denote mean ± S.E.M. of 3 independent experiments. The asterisks denote the
the significance levels: ⁎⁎P < 0.01, compared with control-treated mice, and significance levels: ⁎⁎P < 0.01, compared with control-treated mice, and
#P < 0.05 and ##P < 0.01, compared with vehicle-treated mice (one-way #P < 0.05 and ##P < 0.01, compared with vehicle-treated mice (one-way
ANOVA followed by post-hoc Newman–Keuls test). ANOVA followed by post-hoc Newman–Keuls test).
R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235 231

3.6-fold) increase of PGE2 levels. Local administration of α- 2003). Supporting this data, western blot analysis revealed that
amyrin (0.1–1.0 mg/ear), applied in conjunction with TPA, topical application of TPA in the ears of mice resulted in a
dose-dependently inhibited the increase of PGE2 formation, dramatic (4.5-fold) up-regulation of COX-2 protein expression
when evaluated 6 h after treatment (Fig. 2). The percentage of (Fig. 4). Topical application of the same doses of α-amyrin
inhibition obtained for α-amyrin at 1.0 mg/ear was 68 ± 4% which were effective in inhibiting the increase of PGE2 levels
(P < 0.01; n = 3). Additionally, when the animals were treated after application of TPA (0.1–1.0 mg/ear) caused a dose-
with the positive control drug dexamethasone (0.05 mg/ear), a dependent suppression of the COX-2 expression (Fig. 4). The
complete inhibition of TPA-induced PGE2 increase was maximal inhibition obtained at 1.0 mg/ear was 83 ± 3%
observed (Fig. 2). To gain further insights into the mechanisms (P < 0.01; n = 3). The treatment of mouse skin with α-amyrin
involved in the inhibitory effects of α-amyrin on PGE2 (1.0 mg/ear) alone did not influence the constitutive expression
production following TPA application in the mouse ear, we of COX-2 (data not shown).
firstly evaluated the possible influence of this compound on
COX-1 and COX-2 activity in vitro. In contrast to that reported 3.2. Inhibition of TPA-induced activation of NF-κB by
for the selective inhibitors of COX-1 (SC 560) or COX-2 α-amyrin in mouse skin
(rofecoxib), the α-amyrin triterpene (10–30 μM) failed to
significantly affect either COX-1 (Fig. 3A) or COX-2 (Fig. 3B) The 5′-flanking region of cox-2 gene promoter contains
activities in vitro. We next verified whether α-amyrin could binding sequences for various transcription factors including
inhibit PGE2 in the ear by interfering with COX-2 expression. It NF-κB (Kim and Fischer, 1998). Thus, we attempted to
has been previously shown that topical application of TPA evaluate the effects of α-amyrin on TPA-stimulated DNA
induces expression of COX-2 in mouse skin in vivo (Chun et al., binding of NF-κB in mouse skin. As depicted in Fig. 5, NF-κB

Fig. 5. Effect of α-amyrin on TPA-induced NF-κB pathway activation. (A) Nuclear factor-κB-DNA binding was measured 1 h after acetone (control, C) or TPA
(2.5 μg/ear) treatment in the nuclear extracts. (B) Densitometric analysis of nuclear factor-κB activation. (C) p65/RelA protein was immunoprecipitated in the ear
cytosol extracts 1 h after TPA treatment and western blot was performed for phosphoserine residue. Membranes were stripped and re-probed with anti-p65/RelA
antibody. (Bellow graph) Quantification of phosphoserine residue detection normalized by total p65/RelA. (D) Immunodetection of IκBα degradation after TPA
topical treatment in the cytosolic extract. β-actin was used as a loading control. (Bellow graph) Quantification of IκBα protein expression normalized by β-actin.
Vehicle (V), α-amyrin (0.1–1.0 mg/ear) or dexamethasone (0.05 mg/ear, Dexa) were administered topically in association with TPA. Each column represents the mean ±
S.E.M. of 3 independent experiments. The asterisks denote the significance levels: ⁎P < 0.05 and ⁎⁎P < 0.01, compared with control-treated mice, and #P < 0.05 and
##P < 0.01, compared with vehicle-treated mice (one-way ANOVA followed by post-hoc Newman–Keuls test).
232 R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235

activity was detectable in nuclear proteins obtained from inhibition of NF-κB obtained with the dose of 1.0 mg/ear was
acetone-treated mouse ears (control). However, DNA binding 98 ± 5% (P < 0.01; n = 3). Topical application of dexamethasone
activity was found to be significantly increased (P < 0.01; n = 3) (0.05 mg/ear) also largely (70 ± 6%) inhibited TPA-induced NF-
in nuclear extracts 1 h after TPA treatment. When mice were κB activation (Fig. 5A and B). Recent studies have suggested
treated with α-amyrin (0.1–1.0 mg/ear) the TPA-induced NF- that p65/RelA phosphorylation may be necessary for transcrip-
κB activation was markedly inhibited (Fig. 5A and B). The tional competence of NF-κB (Ghosh and Karin, 2002). To
assess the phosphorylation state of p65/RelA, we have used the
technique of immunoprecipitation for the subunit p65/RelA
followed by immunodetection of phosphoserine. As shown in
Fig. 5C, the topical application of TPA for 1 h in the mouse ears
resulted in a phosphorylation of the serine residues in the p65/
RelA subunit. In addition, the treatment with α-amyrin
significantly reduced the p65/RelA-serine residue phosphoryla-
tion (P < 0.01; n = 3). Our data also demonstrated that treatment
of mouse skin with TPA, in the presence or absence of α-amyrin
(1.0 mg/ear), did not interfere with the cytosolic levels of p65/
RelA (Fig. 5C). Because the nuclear translocation of NF-κB is
dependent on the phosphorylation and subsequent degradation
of IκBα (Ghosh and Karin, 2002), we further examined whether
α-amyrin could block TPA-induced degradation of IκBα. The
topical treatment with α-amyrin (0.1–1.0 mg/ear) dose-
dependently reduced the degradation of IκBα (Fig. 5D). At
the dose 1 mg/kg, α-amyrin completely blocked the degradation
of IκBα (P < 0.05; n = 3).

3.3. Effects of α-amyrin on TPA-induced activation of MAPKs

The MAPKs are proteins that mediate the signal transduction


from the cell surface to the nucleus, and NF-κB family proteins
are the important target molecules of these protein kinases
(Janssen-Heininger et al., 2001; Schulze-Osthoff et al., 1997;
Chun et al., 2003). Results of Fig. 6A and B indicate that very
low levels of phosphorylated ERK and p38 MAPK are detected
under basal conditions in the mouse ear. In contrast, in the TPA
treated group, a marked activation of ERK (4.2-fold; Fig. 6A)
and p38 MAPK (3.1-fold; Fig. 6B) is detected 1 h after
treatment. The topical treatment with α-amyrin (0.1–1.0 mg/
ear) dose-dependently reduced the activation of the both
MAPKs. At the dose of 1.0 mg/kg, α-amyrin almost completely
blocked the activation of ERK (P < 0.01; n = 3) and p38 MAPK
(P < 0.01; n = 3) induced by TPA (Fig. 6). Under these
experimental conditions, the level of the total form of each
kinase remained constant.

Fig. 6. Effect of α-amyrin on TPA-induced protein kinase activation. Activation


of (A) p38 MAPK, (B) ERK and (C) PKCα was measured 1 h after acetone
(control) or TPA (2.5 μg/ear) treatment. Animals were topically treated, at same
time of TPA administration, with vehicle (V), α-amyrin (0.1–1.0 mg/ear) or
dexamethasone (0.05 mg/ear, Dexa). (Bellow graphs) Quantification of
cytosolic phospho-p38 MAPK (p-p38) or phospho-ERK (p-ERK) normalized
by total p38 MAPK and ERK, respectively. PKCα activation was verified by
migration of protein from cytosol (cPKCα) to membrane (mPKCα). Results
represent a fraction between mPKCα and cPKCα. Each column of the graphs
represents the mean ± S.E.M. of 3 independent experiments. The asterisks denote
the significance levels: ⁎⁎P < 0.01, compared with control-treated mice, and
#P < 0.05 and ##P < 0.01, compared with vehicle-treated mice (one-way
ANOVA followed by post-hoc Newman–Keuls test).
R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235 233

3.4. Inhibitory effect of α-amyrin on TPA-induced activation of COX-1, COX-2 is generally, but not exclusively, induced in
PKC reply to stimulators such as growth factors, cytokines, tissue
injury and ultraviolet radiation (Fogh and Kragballe, 2000; Lee
PKC isoforms are major regulators of cutaneous homeostasis et al., 2003; Mitchell and Warner, 2006). Of note, the deletion of
and mediate inflammation in response to TPA (Mueller, 2006). one of the genes responsible for the production of COX-2 in
The results shown in Fig. 6C indicate that topical treatment with mice is capable of reducing the inflammatory process in animal
TPA for 1 h in the mouse ear was capable of activating PKCα models of cutaneous inflammation, indicating that arachidonic
isoform, as charged by its translocation from cytosol- to acid metabolites derived from COX-2 exert a critical role in the
membrane-rich homogenates achieved in treated tissues. The inflammatory skin diseases (Muller-Decker et al., 2002). The
application of α-amyrin (0.1–1.0 mg/ear) in the mouse ear results of the present study clearly show that the inhibition of
resulted in a dose-dependent inhibition of the TPA-induced the levels of PGE2 in the mouse ear after application of α-
PKCα activation (Fig. 6C). At the dose of 1.0 mg/kg, the amyrin is unrelated with its ability to inhibit the activity of either
calculated percentage of inhibition caused by α-amyrin was COX-1 or COX-2 enzymes. This conclusion derives from the
87 ± 6% (P < 0.01; n = 3). view that in contrast to that reported for the selective inhibitor of
COX-1 SC560 or COX-2 rofecoxib, α-amyrin, even at high
4. Discussion concentration in vitro, failed to interfere with their activity.
Next, we assessed whether the inhibition of ear levels of PGE2
Several reports of the literature have shown the effectiveness after topical application of α-amyrin could be associated with its
of α-amyrin or plants that contain this triterpene in rodent ability to interfere with the COX-2 expression. Our data show
models of inflammation, namely TPA-induced ear edema (Recio for the first time that the topical application of the triterpene α-
et al., 1995; Otuki et al., 2005b), carrageenan-induced paw amyrin, at the same dose range where it causes the skin's anti-
edema (Recio et al., 1995) and arthritis-induced by complete inflammatory property, dose-dependently inhibited the expres-
Freund's adjuvant (Kweifio-Okai et al., 1994). However, the sion of the COX enzyme. These observations could explain the
molecular mechanisms underlying these effects remain largely relatively strong and rapid onset topical anti-inflammatory
unidentified. A better understanding of the mode of action of α- property reported for this plant triterpene.
amyrin would enhance the potential therapeutic interest on this NF-κB has emerged as one of the most promising and
naturally-occurring pentacyclic triterpene. In the present study ubiquitous molecular targets in the prevention of cancer and
we have greatly extended our previous findings (Otuki et al., inflammation (Ghosh and Karin, 2002; Karin, 2006).
2005b), by demonstrating that the anti-inflammatory effects of Degradation of IκB allows the NF-κB to translocate to the
α-amyrin appear to rely on the down regulation of COX-2 nucleus, where it regulates the transcription of several genes,
expression and the inhibition of NF-κB and some protein including the COX-2 and several cytokine genes (Kim and
kinases. Fischer, 1998; Ghosh and Karin, 2002). Notably, several
The COX product of arachidonic acid, PGE2, has long been studies have indicated that the increase in the activation of
known to have proinflammatory properties. Intradermal injec- factor NF-κB has an important role in the development and
tions of PGE2 into human skin cause erythema, while higher maintenance of cutaneous inflammatory diseases, including
doses produce edema. Of note, PGE2 can amplify the vascular psoriasis, contact dermatitis and atopic dermatitis (Bell et al.,
permeability changes induced by other mediators (Fogh and 2003). Therefore, drugs that inhibit the activation of factor
Kragballe, 2000). Our present results show that at the same NF-κB are in principle, of potential interest for the treatment
range of dose that α-amyrin inhibited TPA-induced ear oedema cutaneous inflammatory diseases (Courtois, 2005). In the
and polymorphonuclear cell influx (Otuki et al., 2005b) it present study, we have shown that TPA produces an increase
caused a dose-dependent inhibition of ear levels of PGE2 in the in NF-κB DNA-binding activity, through a mechanism
mouse skin treated with TPA. Such results could explain, at involving the degradation of IκBα, phosphorylation of p65/
least in part, the topical and systemic anti-inflammatory and RelA and the subsequent translocation of NF-κB into the
antinociceptive properties of α-amyrin reported in previous nucleus. Of interest, we found that topical application of α-
works (Recio et al., 1995; Otuki et al., 2005a,b). A growing amyrin to the mouse skin at the same doses where it was
number of studies suggest that PGE2 is associated with effective in inhibiting COX-2 expression after TPA topical
pathogenesis of inflammatory skin disease, for instance atopic application (0.1–1.0 mg/ear), consistently and dose-depen-
dermatitis (Fogh and Kragballe, 2000; Neisius et al., 2002). dently inhibited TPA-induced NF-κB activation. This effect
Therefore, our data showing significant reduction of the tissue seems to be related to its ability of reducing IκBα degradation
levels of PGE2 after TPA application in the mouse ears caused and p65/RelA phosphorylation. Therefore, the inhibition of
by topical application of α-amyrin might be potentially relevant NF-κB activation by α-amyrin could explain its effects on the
in the research for new therapeutic agents which have expression of COX-2 enzyme, as well as the previously
prostanoids as a target. reported capability of α-amyrin to diminish IL-1β levels in
The well known enzymes cyclooxygenase COX-1 and the mouse skin following application of TPA (Otuki et al.,
COX-2 are responsible for the transformation of the arachidonic 2005b).
acid in prostaglandins, and have an important function in the MAPKs and PKC are critical components of cellular signal
inflammatory processes. In contrast to the constituent form transduction cascades. They are directly involved in many
234 R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235

diseases, including cancer and inflammation, and have become Bevilacqua, A., Ceriani, M.C., Capaccioli, S., Nicolin, A., 2003. Post-
one of the most important target classes for drug development transcriptional regulation of gene expression by degradation of messenger
RNAs. J. Cell. Physiol. 195, 356–372.
(Cohen, 2002; Dancey and Sausville, 2003). Thus, in the Chen, C.C., Sun, Y.T., Chen, J.J., Chiu, K.T., 2000. TNF-alpha-induced
present study we characterized the effect of topical administra- cyclooxygenase-2 expression in human lung epithelial cells: involvement of
tion of α-amyrin in TPA-induced PKC, as well as ERK and p38 the phospholipase C-gamma 2, protein kinase C-alpha, tyrosine kinase, NF-
MAPK activation. Our data demonstrated that topical treatment kappa B-inducing kinase, and I-kappa B kinase 1/2 pathway. J. Immunol.
165, 2719–2728.
with α-amyrin greatly inhibited the TPA-induced PKCα, ERK
Chun, K.S., Keum, Y.S., Han, S.S., Song, Y.S., Kim, S.H., Surh, Y.J., 2003.
and p38 MAPK activation at the same range of doses necessary Curcumin inhibits phorbol ester-induced expression of cyclooxygenase-2 in
to prevent the IκBα degradation and p65/RelA phosphorylation mouse skin through suppression of extracellular signal-regulated kinase
after TPA application. Several studies have indicated the activity and NF-kappaB activation. Carcinogenesis 24, 1515–1524.
participation of these protein kinases in the regulation of Chun, K.S., Kim, S.H., Song, Y.S., Surh, Y.J., 2004. Celecoxib inhibits phorbol
inflammatory gene expression, acting both at transcriptional ester-induced expression of COX-2 and activation of AP-1 and p38 MAP
kinase in mouse skin. Carcinogenesis 25, 713–722.
and posttranscriptional levels (Whitmarsh and Davis, 1996; Clark, A.R., Dean, J.L., Saklatvala, J., 2003. Post-transcriptional regulation of
Ono and Han, 2000; Yang et al., 2003; Clark et al., 2003). gene expression by mitogen-activated protein kinase p38. FEBS Lett. 546,
Recent reports have implicated both MAPKs and PKC in the 37–44.
up-regulation of COX-2 expression by TPA, through a Cohen, P., 2002. Protein kinases—the major drug targets of the twenty-first
mechanism dependent on NF-κB activation (Surh, 2003). century? Nat. Rev., Drug Discov. 1, 309–315.
Courtois, G., 2005. NF-kappaB in skin homeostasis. Exp. Dermatol. 14,
Likewise, the activation of these protein kinases has been 781–782.
implicated in the regulation of the half-lives of many Dancey, J., Sausville, E.A., 2003. Issues and progress with protein kinase
inflammatory mediator RNAs, including COX-2, TNF-α, and inhibitors for cancer treatment. Nat. Rev., Drug Discov. 2, 296–313.
IL-1β (Clark et al., 2003; Winzen et al., 1999; Bevilacqua et al., do Vale, A.E., David, J.M., Brandão, H.N., David, J.P., 2005. A new flavonol
2003). In this context, α-amyrin regulating the p38 MAPK, glycoside derivative from leaves of Moldenhawera nutans. Z. Naturforsch.
[C] 60, 45–49.
ERK and PKCα activation could decrease COX-2 expression Ferreira, J., Triches, K.M., Medeiros, R., Calixto, J.B., 2005. Mechanisms
by reducing the activity of transcriptional factors (eg, NF-κB), involved in the nociception produced by peripheral protein kinase c
by modulating the stability of some pro-inflammatory proteins activation in mice. Pain 117, 171–181.
mRNAs (eg, IL-1β and TNF-α), or even by diminishing the Fogh, K., Kragballe, K., 2000. Eicosanoids in inflammatory skin diseases.
stability of COX-2 mRNA. However, additional studies are Prostaglandins Other Lipid Mediat. 63, 43–54.
Ghosh, S., Karin, M., 2002. Missing pieces in the NF-kappaB puzzle. Cell 109,
necessary to confirm these hypotheses. S81–S96.
In summary, we have provided in vivo evidence that the Homey, B., 2005. Chemokines and inflammatory skin diseases. Adv. Dermatol.
plant-derived pentacyclic triterpene α-amyrin exhibits strong 21, 251–277.
and rapid onset topical anti-inflammatory actions in the mouse Janssen-Heininger, Y.M., Poynter, M.E., Baeuerle, P.A., 2001. Recent advances
towards understanding redox mechanisms in the activation of nuclear factor
ear edema induced by TPA through a mechanism that seems to
kappaB. Free Radic. Biol. Med. 28, 1317–1327.
involve its ability to inhibit the levels of PGE2, via inhibition of Karin, M., 2006. Nuclear factor-kappaB in cancer development and progression.
the COX-2 expression. Based on our results and in the previous Nature 441, 431–436.
investigations we might infer that α-amyrin exerts its anti- Kim, Y., Fischer, S.M., 1998. Transcriptional regulation of cyclooxygenase-2 in
inflammatory properties through regulation of PKCα and mouse skin carcinoma cells. Regulatory role of CCAAT/enhancer-binding
MAPKs activation probably by inhibiting the transcriptional proteins in the differential expression of cyclooxygenase-2 in normal and
neoplastic tissues. J. Biol. Chem. 273, 27686–27694.
activity of NF-κB. Collectively, the present findings suggest Kweifio-Okai, G., De Munk, F., Rumble, B.A., Macrides, T.A., Cropley, M.,
that α-amyrin might constitute a potential and relevant 1994. Antiarthritic mechanisms of amyrin triterpenes. Res. Commun. Mol.
alternative for the treatment of inflammatory diseases. Pathol. Pharmacol. 85, 45–55.
Kyriakis, J.M., Avruch, J., 2001. Mammalian mitogen-activated protein kinase
Acknowledgements signal transduction pathways activated by stress and inflammation. Physiol.
Rev. 81, 807–869.
Lee, J.L., Mukhtar, H., Bickers, D.R., Kopelovich, L., Athar, M., 2003.
We are grateful to the Merck and Co., Inc. for donating the Cyclooxygenases in the skin: pharmacological and toxicological implica-
Rofecoxib used in this work. This study was supported by the tions. Toxicol. Appl. Pharmacol. 192, 294–306.
Conselho Nacional de Desenvolvimento Científico (CNPq), by Medeiros, R., Cabrini, D.A., Ferreira, J., Fernandes, E.S., Mori, M.A., Pesquero,
J.B., Bader, M., Avellar, M.C., Campos, M.M., Calixto, J.B., 2004.
the Programa de Apoio aos Núcleos de Excelência (PRONEX)
Bradykinin B1 receptor expression induced by tissue damage in the rat
(Brazil) and by the Fundação de Apoio a Pesquisa de Santa portal vein: a critical role for mitogen-activated protein kinase and nuclear
Catarina (FAPESC). M.F.O and R.M. are PhD students in factor-kappaB signaling pathways. Circ. Res. 94, 1375–1382.
pharmacology, and they thank CNPq (Brazil) for fellowship Mitchell, J.A., Warner, T.D., 2006. COX isoforms in the cardiovascular system:
support. understanding the activities of non-steroidal antiinflammatory drugs. Nat.
Rev., Drug Discov. 5, 75–86.
Mueller, M.M., 2006. Inflammation in epithelial skin tumours: old stories and
References new ideas. Eur. J. Cancer 42, 735–744.
Muller-Decker, K., Neufang, G., Berger, I., Neumann, M., Marks, F.,
Bell, S., Degitz, K., Quirling, M., Jilg, N., Page, S., Brand, K., 2003. Furstenberger, G., 2002. Transgenic cyclooxygenase-2 overexpression
Involvement of NF-kappaB signalling in skin physiology and disease. Cell. sensitizes mouse skin for carcinogenesis. Proc. Natl. Acad. Sci. U. S. A.
Signal. 15, 1–7. 99, 12483–12488.
R. Medeiros et al. / European Journal of Pharmacology 559 (2007) 227–235 235

Neisius, U., Olsson, R., Rukwied, R., Lischetzki, G., Schmelz, M., 2002. triterpenes: evidence for participation of protein kinase C and protein
Prostaglandin E2 induces vasodilation and pruritus, but no protein kinase A pathways. J. Pharmacol. Exp. Ther. 313, 310–318.
extravasation in atopic dermatitis and controls. J. Am. Acad. Dermatol. Otuki, M.F., Vieira-Lima, F., Malheiros, A., Yunes, R.A., Calixto, J.B.,
47, 28–32. 2005b. Topical antiinflammatory effects of the ether extract from Protium
Numerof, R.P., Dinarello, C.A., Asadullah, K., 2005. Cytokines as potential kleinii and alpha-amyrin pentacyclic triterpene. Eur. J. Pharmacol. 507,
therapeutic targets for inflammatory skin diseases. Eur. Cytokine Netw. 16, 253–259.
101–103. Pivarcsi, A., Homey, B., 2005. Chemokine networks in atopic dermatitis: traffic
Okayama, Y., 2005. Oxidative stress in allergic and inflammatory skin diseases. signals of disease. Curr. Allergy Asthma Rep. 5, 284–290.
Curr. Drug Targets Inflamm. Allergy 4, 517–519. Recio, M.C., Giner, R.M., Manez, S., Rios, J.L., 1995. Structural requirements
Oliveira, F.A., Lima-Junior, R.C., Cordeiro, W.M., Vieira-Junior, G.M., Chaves, for the antiinflammatory activity of natural triterpenoids. Planta Med. 61,
M.H., Almeida, F.R., Silva, R.M., Santos, F.A., Rao, V.S., 2004a. Pentacyclic 182–185.
triterpenoids, alpha, beta-amyrins, suppress the scratching behavior in a mouse Rossner, M., Yamada, K.M., 2004. What's in a picture? The temptation of image
model of pruritus. Pharmacol. Biochem. Behav. 78, 719–725. manipulation. J. Cell Biol. 166, 11–15.
Oliveira, F.A., Vieira-Junior, G.M., Chaves, M.H., Almeida, F.R., Santos, K.A., Schulze-Osthoff, K., Ferrari, D., Riehemann, K., Wesselborg, S., 1997.
Martins, F.S., Silva, R.M., Santos, F.A., Rao, V.S., 2004b. Gastroprotective Regulation of NF-κB activation by MAP kinase cascades. Immunobiology
effect of the mixture of alpha- and beta-amyrin from Protium heptaphyllum: 198, 35–49.
role of capsaicin-sensitive primary afferent neurons. Planta Med. 70, Surh, Y.J., 2003. Cancer chemoprevention with dietary phytochemicals. Nat.
780–782. Rev., Cancer 3, 768–780.
Oliveira, F.A., Chaves, M.H., Almeida, F.R., Lima, R.C., Silva, R.M., Maia, Whitmarsh, A.J., Davis, R.J., 1996. Transcription factor AP-1 regulation by
J.L., Brito, G.A., Santos, F.A., Rao, V.S., 2005. Protective effect of alpha- mitogen-activated protein kinase signal transduction pathways. J. Mol. Med.
and beta-amyrin, a triterpene mixture from Protium heptaphyllum (Aubl.) 74, 589–607.
March. trunk wood resin, against acetaminophen-induced liver injury in Winzen, R., Kracht, M., Ritter, B., Wilhelm, A., Chen, C.Y., Shyu, A.B., Muller,
mice. J. Ethnopharmacol. 98, 103–108. M., Gaestel, M., Resch, K., Holtmann, H., 1999. The p38 MAP-kinase
Ono, K., Han, J., 2000. The p38 signal transduction pathway: activation and pathway signals for cytokine-induced mRNA stabilization via MAP-kinase-
function. Cell. Signal. 12, 1–13. activated protein kinase 2 and an AU-rich region-targeted mechanism.
Otuki, M.F., Ferreira, J., Lima, F.V., Meyre-Silva, C., Malheiros, A., Muller, EMBO J. 18, 4969–4980.
L.A., Cani, G.S., Santos, A.R., Yunes, R.A., Calixto, J.B., 2005a. Yang, S.H., Sharrocks, A.D., Whitmarsh, A.J., 2003. Transcriptional regulation
Antinociceptive properties of mixture of alpha-amyrin and beta-amyrin by the MAP-kinase signaling cascades. Gene 320, 3–21.

You might also like