You are on page 1of 9

Inflammation ( # 2013)

DOI: 10.1007/s10753-013-9732-x

Thymol Inhibits LPS-Stimulated Inflammatory Response via


Down-Regulation of NF-κB and MAPK Signaling Pathways
in Mouse Mammary Epithelial Cells

Dejie Liang,1 Fengyang Li,1 Yunhe Fu,1 Yongguo Cao,1 Xiaojing Song,1 Tiancheng Wang,1
Wei Wang,1 Mengyao Guo,1 Ershun Zhou,1 Depeng Li,1 Zhengtao Yang,1,2 and Naisheng Zhang1,2

Abstract—Thymol is a natural monoterpene phenol primarily found in thyme, oregano, and


tangerine peel. It has been shown to possess anti-inflammatory property both in vivo and in
vitro. In the present paper, we studied the anti-inflammatory effect of thymol in lipopolysac-
charide (LPS)-stimulated mouse mammary epithelial cells (mMECs). The mMECs were stim-
ulated with LPS in the presence or absence of thymol (10, 20, 40 μg/mL). The concentrations
of tumor necrosis factor α (TNF-α), interleukin (IL)-6, and IL-1β in the supernatants of culture
were determined using enzyme-linked immunosorbent assay. Cyclooxygenase-2 (COX-2), ind-
ucible nitric oxide synthase (iNOS), extracellular signal-regulated protein kinase (ERK), c-Jun
N-terminal kinase (JNK), nuclear factor-κB (NF-κB), and inhibitor protein of NF-κB (IκBα)
were measured using western blot. The results showed that thymol markedly inhibited the
production of TNF-α and IL-6 in LPS-stimulated mMECs. The expression of iNOS and COX-2
was also suppressed by thymol in a dose-dependent manner. Furthermore, thymol blocked the
phosphorylation of IκBα, NF-κB p65, ERK, JNK, and p38 mitogen-activated protein kinases
(MAPKs) in LPS-stimulated mMECs. These results indicate that thymol exerted anti-inflam-
matory property in LPS-stimulated mMECs by interfering the activation of NF-κB and MAPK
signaling pathways. Thereby, thymol may be a potential therapeutic agent against mastitis.

KEY WORDS: thymol; lipopolysaccharide (LPS); mammary epithelial cells (MECs); nuclear factor-kappaB
(NF-κB); mitogen-activated protein kinases (MAPKs).

INTRODUCTION than a physical barrier for pathogens and allergens but also
are capable of producing inflammatory mediators and
Inflammation is a complex pathophysiological pro- phagocytizing cellular or acellular debris in response to
cess mediated by a variety of signaling molecules, which these microbes [2, 3]. However, excessive inflammatory
response in the mammary gland may cause mastitis, which
were produced by leukocytes, macrophages, mast cells,
costs lots of expenses [4]. Lipopolysaccharide (LPS), a
and many other cell types [1]. In the mammary gland of structural component of the outer membrane of gram-
mammals, macrophages are the prime candidates for the negative bacteria, is a potent agent widely used to model
first line of defense against invaded bacteria and their bacterial challenges of mammalian cells and induces pro-
products. Mammary alveolar epithelial cells not only come inflammatory cytokine production. In recent years, there
in direct contact with the invading bacteria and act as more have been reports that LPS could effectively induce
mastitis and is recognized as a valuable tool to study the
1 effects of mastitis [2, 4, 5]. LPS-induced inflammation
Department of Clinical Veterinary Medicine, College of Veterinary
Medicine, Jilin University, Changchun, Jilin Province 130062,
is characterized by the release of pro-inflammatory
People’s Republic of China cytokines, such as tumor necrosis-α (TNF-α), interleukin
2
To whom correspondence should be addressed at Department of (IL)-6, IL-1β, and other mediators, those of which play
Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin important roles during the inflammation process [6, 7]. The
University, Changchun, Jilin Province 130062, People’s Republic of expression of inducible nitric oxide synthase (iNOS) and
China. E-mail: zhangns@jlu.edu.cn; yangzhengtao01@sina.com cyclooxygenase 2 (COX-2) was also reported to be

0360-3997/13/0000-0001/0 # 2013 Springer Science+Business Media New York


Liang, Li, Fu, Cao, Song, Wang, Wang, Guo, Zhou, Li, Yang, and Zhang

induced by the binding of LPS or TNF-α in mammary Chemicals and Reagents


epithelial cells [8–10]. MAPKs are a family of serine/
threonine protein kinases, which include members such as Thymol (purity >99.9 %) was purchased from the
extracellular signal receptor-activated kinase (ERK) 1/2, National Institute for the Control of Pharmaceutical and
p38, and c-Jun N-terminal kinase (JNK). It is widely Biological Products (Beijing, China). The thymol stock
accepted that mitogen-activated protein kinase (MAPK) solution was prepared with dimethyl sulfoxide (DMSO),
and the nuclear factor-κB (NF-κB) pathways play vital and the DMSO concentration in the solution did not exceed
roles in inflammation, both of which can induce the 0.1 %. LPS (Escherichia coli 055:B5) and 3-(4,5-dimeth-
expressions of various inflammatory mediators and pro- ylthiazol-2-y1)-2,5-diphenyltetrazolium bromide (MTT)
inflammatory cytokines [11–16].
were purchased from Sigma Chemical Co. (St. Louis,
Thymol (2-isopropyl-5-methylphenol) is a natural
MO, USA). Dulbecco's modified Eagle's medium (DMEM
monoterpene phenol primarily found in thyme, oregano,
F12/1:1), fetal calf serum (FCS), and trypsin/EDTA were
and tangerine peel (Fig. 1). It has been credited with a
purchased from Hyclone (Logan, UT, USA). Collagenase I
series of pharmacological properties, including antimicro-
and collagenase II were purchased from Invitrogen Corp.
bial, anti-inflammatory, and antioxidant effects [17–21].
(Carlsbad, CA, USA). Epidermal growth factor (EGF),
However, there are very limited literatures about the anti-
transferrin, and T3 were purchased from PeproTech;
inflammatory property of thymol on mastitis. The aim of
mouse TNF-α and IL-6 enzyme-linked immunosorbent
this study was to verify the potential anti-inflammatory
assay (ELISA) kits were purchased from Biolegend (San
effect of thymol on LPS-stimulated inflammatory response
Diego, CA, USA). Mouse Specific iNOS antibody, COX-2
in mouse mammary epithelial cells (mMECs).
antibody, mouse monoclonal phospho-specific p46–p54
JNK antibodies, mouse monoclonal phospho-specific p42–
p44 ERK antibodies, mouse monoclonal phospho-specific
MATERIALS AND METHODS p38 antibodies, mouse mAb phospho-NF-κB, mouse mAb
NF-κB, mouse mAb phospho-IκBα, and rabbit mAb IκBα
Animals were purchased from Cell Signaling Technology, Inc.
BALB/c mice (10–15-day gravid), weighing approx- (Beverly, MA, USA). β-actin and horseradish peroxidase-
imately 18–22 g, were purchased from Norman Bethune conjugated goat anti-rabbit and goat anti-mouse antibodies
University of Medical Science of Jilin University. The were provided by Tianjin Sungene Biotech Co., Ltd.
mice were housed in special room with 24±1 °C and 40– (Tianjin, China). All other chemicals were of reagent grade
80 % humidity. All the animals received food and water and were endotoxin free.
adequately. All the animal studies were conducted
according to the experimental practices and standards
approved by the Animal Welfare and Research Ethics Mouse Mammary Epithelial Cell Isolation
Committee at Jilin University (approval ID 20111106–2), and Culture
and all efforts were made to minimize suffering. Primary cultured mMECs were prepared as previously
described by Smalley [22]. Briefly, the mammary tissues
were removed aseptically from 15-day gravid BALB/c mice
and minced into pasty fine tissues. The minced tissues were
digested by collagenase I/II/trypsin mixture (Invitrogen) and
shaken at 37 °C. After filtration to remove undissociated
tissue and debris, the cells were collected by centrifugation
at 250×g for 5 min three times. The cell pellets were
resuspended in DMEM/F12 containing 10 % FCS and
incubated for 1 h at 37 °C, and then the supernatant was
collected. This step was repeated three times until the
fibroblasts were removed. After the last incubate, the cells
were resuspended in DMEM/F12 containing 10 % FCS,
0.5 % transferrin, 0.1 % T3, and 0.5 % EGF, and were
Fig. 1. Chemical structure of thymol. cultured at 37 °C with 5 % CO2. The medium was changed
Thymol Inhibits LPS-Stimulated Inflammatory Response

every day. In all the experiments, the cells were allowed to Substrate. The images were captured using a MicroChemi
acclimate for 24 h before any treatments. 4.2 system (DNR Bio Imaging Systems, Jerusalem, Israel).
The intensities of the bands were measured with ImageJ
MTT Assay for Cell Viability software.
The effect of thymol on cell viability was evaluated
Statistical Analysis
using the MTT assay. The mouse mammary epithelial
cells were exposed to thymol at a concentration of 200– All values were expressed as the mean ± SEM.
0 μg/mL), followed by stimulation with 1 μg/mL of LPS Differences between the mean values of normally
for 24 h in a 96-well microplate. Subsequently, 20 μL distributed data were assessed with a one-way ANOVA
MTT (5 mg/mL) was added to each well. The resulting (Dunnett's t test) and two-tailed Student's t test. P values
solution was incubated at 37 °C until blue deposits were of 0.05 or less were considered statistically significant.
visible. Cell-free supernatants were then removed and
resolved with 150 μL DMSO in each well. The optical
density was measured at 570 nm on a microplate reader. RESULTS

Cytokine Assay Effect of Thymol on mMEC Viability


5
The mMECs were plated onto 24-well plates (4×10 MTT is a monotetrazolium salt; the reduction of
cells/well) and pretreated with or without various which is one of the most frequently employed methods for
concentrations of thymol (10, 20, 40 μg/mL) for 1 h and the detection of cell cytotoxicity or viability following
then were stimulated with LPS (1 μg/mL) for 12 h. The exposure to toxic substances. This water-soluble salt is
supernatant was collected to measure the levels of TNF- converted to an insoluble purple formazan by succinate
α, IL-1β, and IL-6. All the operations were followed from dehydrogenase within the mitochondria of the living cells,
the instruction of the ELISA kits. and the purple formazan is in direct proportion to the
healthy cell numbers. In this study, the potential cytotox-
Western Blot Analysis icity of thymol was evaluated using the MTT assay after
incubating the cells for 24 h in the absence or presence of
The primary cultured mMECs were treated with LPS, and the result showed that cell viability was not
various concentrations of thymol for 1 h and stimulated
affected by thymol at the concentrations used (10, 20, and
with 1 μg/mL of LPS for 24 h (for iNOS and COX-2) or
40 μg/mL) (Fig. 2). Thus, the effects of thymol on mMECs
1 h (for NF-κB and MAPKs). Total cellular proteins were
were not attributable to cytotoxic effect.
extracted using mammalian protein extraction reagent
(Pierce, Rockford, IL, USA) supplemented with a 1:100
Effect of Thymol on Cytokine Release
dilution of phenylmethylsulfonyl fluoride (Sigma). The
in the Supernatants of LPS-stimulated mMECs
concentrations of the protein samples were determined
using BCA assay kits. For western blot analysis, 20 μg of LPS-induced inflammatory response is characterized
total protein per lane was resolved by SDS-PAGE and by the release of pro-inflammatory cytokines such as
blotted onto PVDF membrane. Mouse-specific iNOS TNF-α, IL-1β, and IL-6 [6, 7]. These cytokines play
antibody, COX-2 antibody, rabbit anti-phospho-p38, crucial roles during inflammation. TNF-α, along with
anti-phospho-JNK, anti-phospho-ERK, anti-phospho- IL-1β and IL-6, is recognized as an important early
IκB-3, and mouse anti-phospho-IκB-α monoclonal anti- inflammatory mediator, and the over-expression of this
body (Cell Signaling Technology) were used for protein cytokine can lead to severe pro-inflammatory reactions. To
detection, and the corresponding antibodies were used determine whether thymol affects the expression of
for the detection of unphosphorylated molecules. The inflammatory cytokines in LPS-stimulated mMECs, the
β-actin protein served as an internal control. The proteins expression of TNF-α, IL-1β, and IL-6 was detected using
were visualized using secondary HRP-conjugated goat ELISA. As shown in Fig. 3, the levels of TNF-α and IL-6
anti-rabbit and goat anti-mouse antibodies (Jackson significantly increased after LPS challenge compared with
ImmunoResearch Laboratories, West Grove, PA, USA) those of the control group (P<0.01), while the pretreatment
and Pierce Supersignal West Pico Chemiluminescent with thymol (20 and 40 μg/mL) reduced the production of
Liang, Li, Fu, Cao, Song, Wang, Wang, Guo, Zhou, Li, Yang, and Zhang

Fig. 2. Effect of thymol on the cell viability of mMECs. The cells were cultured with different concentrations of thymol (10, 20, and 40 μg/mL) in the
absence or presence of 1 μg/mL LPS for 24 h. The cell viability was determined using MTT assay. The values are presented as mean ± SEM (n=8).

TNF-α and IL-6 in a dose-dependent manner, and the not observed in our experiment, and the data were not
levels of cytokines were marked lower than those in the shown. The results showed that thymol attenuated the
LPS group (P<0.01). However, the level of IL-1β was inflammatory response via suppressing the pro-inflamma-

Fig. 3. Effects of thymol on the expression of TNF-α (a) and IL-6 (b) in LPS-stimulated mMECs. The cells were treated with 1 μg/mL LPS in the
absence or presence of thymol (10, 20, and 40 μg/mL) for 12 h. Levels of TNF-α (a) and IL-6 (b) in culture supernatants were measured using ELISA.
The data are representative of three independent experiments, and the values are presented as mean ± SEM. #P<0.01 significant difference from control;
*P<0.05 and **P<0.01 significant difference from LPS only.
Thymol Inhibits LPS-Stimulated Inflammatory Response

tory cytokine production in primary cultured LPS-stimu- the activation of NF-κB and MAPK signaling pathways in
lated mMECs. diverse mechanisms, it is of importance to investigate the
potential inhibitory activity of thymol on the activation of
Effect of Thymol on iNOS and COX-2 Protein NF-κB and MAPKs in mMECs. The levels of p-NF-κB
Expression in LPS-stimulated mMECs p65 and p-IκBα in the NF-κB signaling pathway and the
levels of p-p38, p-ERK, and p-JNK in the MAPK signaling
Both iNOS and COX-2 are important enzymes pathway were analyzed using western blot (Figs. 5 and 6).
involved in mediating the inflammation process. Up- The results showed that LPS treatment significantly
regulation of iNOS and COX-2 occurs in many patho- activated the NF-κB and MAPK signaling pathways. In
logical conditions, such as mastitis. Therefore, the anti- contrast, pretreatment with thymol (10, 20, and 40 μg/mL)
inflammatory activity of thymol can be demonstrated by inhibited the up-regulation of phosphorylation of NF-κB
its effect on iNOS and COX-2 expression in LPS-induced p65, p-IκBα, p38, ERK, and JNK as compared with the
mMECs. As shown in Fig. 4, western blot analysis LPS group. These results of our immunoblot analyses
showed that the expression of iNOS and COX-2 were suggest that the inhibition of TNF-α, IL-6, iNOS, and
strongly increased by LPS, while the application of COX-2 expression by thymol might block the LPS-
thymol (10, 20, and 40 μg/mL) 1 h prior to LPS treatment induced NF-κB and MAPK activation.
resulted in a dramatic reduction in the levels of iNOS and
COX-2 proteins in a dose-dependent manner in mMECs
(P<0.01 or P<0.05).
DISCUSSION
Effect of Thymol on the Activation of NF-κB
Mastitis is typically defined as the inflammation of
and MAPK Signaling Pathways
the mammary gland which is usually due to bacterial
NF-κB activation is critical in regulating gene infection in mammals [24]. Mammary epithelial cells
expression in LPS-induced inflammatory responses [23]. directly contact with the invading pathogens and involved
Since COX-2 and iNOS are frequently regulated through in initiating innate immune response during the process of

Fig. 4. Effects of thymol on inducible nitric oxide synthase and COX-2 protein expression in LPS-stimulated mMECs. The mMECs were treated with
different concentrations (10, 20, and 40 μg/mL) of thymol for 1 h and stimulated with 1 μg/mL of LPS for 24 h. Total cellular protein (20 μg) was
resolved by SDS-PAGE, transferred to a polyvinylidene difluoride membrane, and visualized by western blot analysis. Quantification of iNOS and COX-
2 protein expression was normalized to β-actin using a densitometer. Data are representative of three independent experiments, and the values are
presented as mean ± SEM. #P<0.01 significant difference from control; *P<0.05 and **P<0.01 significant difference from LPS only.
Liang, Li, Fu, Cao, Song, Wang, Wang, Guo, Zhou, Li, Yang, and Zhang

Fig. 5. Effects of thymol on phosphorylation of IκBα and NF-κB p65 in LPS-stimulated mMECs. The results illustrated that thymol could inhibit the
phosphorylation of IκBα and NF-κB p65. mMECs were treated with different concentrations (10, 20, and 40 μg/mL) of thymol for 1 h and stimulated
with 1 μg/mL of LPS for 1 h. Total cellular protein (20 μg) was resolved by SDS-PAGE, transferred to a polyvinylidene difluoride membrane, and
visualized by western blot analysis. Quantification of p-IκBα and p-p65 protein expression was normalized to β-actin using a densitometer. Data are
representative of three independent experiments, and the values are presented as mean ± SEM. #P<0.01 significant difference from control; *P<0.05 and
**P<0.01 significant difference from LPS only.

mammary inflammation [2]. Thymol is a p-cymene- therapeutic strategies may play a role in many inflamma-
derived compound primarily found in thyme, oregano, tory diseases, such as mastitis. In this experiment, we
and tangerine peel, which has been used in traditional found that the concentrations of TNF-α and IL-6 were
Chinese medicine for its multiple biological properties. In markedly increased in the culture 12 h after LPS
the present study, we demonstrated that thymol exerted stimulation, and pretreatment with thymol inhibited the
beneficial effects in LPS-induced mouse primary cultured production of TNF-α and IL-6 in a dose-dependent
epithelial cells, which was associated with the attenuation manner. Interestingly, the IL-1β level was not observed
of inflammatory responses. under our experimental conditions. A possible explana-
LPS, which is capable of eliciting cellular activation tion of this result may be the lower concentration of LPS
of alveolar macrophages and epithelial cells, and trigger- used and the difference in LPS challenging time in
ing the release of pro-inflammatory cytokines such as mMECs compared to those reported in an in vitro study
TNF-α, IL-1β, and IL-6 [2, 25], is known as the major [27]. Our results indicated that thymol attenuated the pro-
virulence factor of clinical mastitis. These pro-inflamma- inflammatory cytokine TNF-α and IL-1β release to exert
tory cytokines are typical examples of multifunctional positive effects against LPS-challenged mMECs.
mediators involved in the regulation of the immune COX-2 and iNOS have been implicated as important
response and inflammation. TNF-α and IL-1β are two enzymes that mediate inflammatory process [28]. COX-2
key components of the innate immune response to and iNOS are not usually expressed in quiescent cells; they
infection [7]. Likewise, IL-6 is a pleiotropic cytokine are transcriptionally induced in response to bacterial
with strong influences on inflammatory responses and a endotoxins and pro-inflammatory cytokines in macrophages
major effector of the acute phase reaction [2]. They are and various other cell types. A variety of studies have shown
produced by a variety of activated immune cells including that iNOS and COX-2 are up-regulated by pro-inflamma-
T cells, B cells, macrophages, and some of epithelial cells tory cytokines and further aggravate the inflammatory
[26]. The over-expression of these cytokines can lead to immune response during infection [29–31]. The pleiotropic
severe pro-inflammatory reactions. Thus, anti-TNF-α inflammatory mediators NO and PGE2, which are generated
Thymol Inhibits LPS-Stimulated Inflammatory Response

Fig. 6. Effects of thymol on the phosphorylation of extracellular p38, ERK, and JNK in LPS-stimulated mMECs. The results illustrated that thymol
could inhibit the phosphorylation of p38, ERK, and JNK. mMECs were treated with different concentrations (10, 20, and 40 μg/mL) of thymol for 1 h and
stimulated with 1 μg/mL of LPS for 1 h. Total cellular protein (20 μg) was resolved by SDS-PAGE, transferred to a polyvinylidene difluoride membrane,
and visualized by western blot analysis. Quantification of p-p38, p-ERK, and p-JNK protein expression was normalized to β-actin using a densitometer.
Data are representative of three independent experiments, and the values are presented as mean ± SEM. #P<0.01 significant difference from control;
*P<0.05 and **P<0.01 significant difference from LPS only.

by iNOS and COX-2, are involved in the pathogenesis of tory responses may be partially attributed to the inhibition of
inflammation and infection. Thymol has been shown to iNOS and COX-2 expression in LPS-challenged mMECs.
inhibit the LPS-stimulated NO generation in mouse The NF-κB signaling pathway has been considered
macrophage-like RAW 264.7 cells [32]. Marsik et al. to play a pivotal role in inflammatory phenotypic changes
concluded that thymol exhibited inhibitory effect on COX- in various pathophysiological conditions [34, 35]. NF-κB
2 in vitro [33]. In this study, we examined the effects of is one of the most ubiquitous and important transcription
thymol on iNOS and COX-2 expression in LPS-stimulated factors that regulate the expression of genes involved in
mMECs. In accordance with the previously described controlling inflammatory responses, such as the expres-
studies, we found that thymol (10, 20, and 40 μg/mL) sion of iNOS, COX-2, and other inflammatory-related
suppressed the LPS-induced iNOS and COX-2 protein cytokines, including TNF-α, IL-1β, and IL-6 [36, 37].
expression in mMECs in a dose-dependent manner, which Under normal conditions, NF-κB is present in its inactive
indicated that the beneficial effect of thymol on inflamma- cytoplasmic form bound to the repressor of NF-κB
Liang, Li, Fu, Cao, Song, Wang, Wang, Guo, Zhou, Li, Yang, and Zhang

(IκBs). However, in response to certain stimuli, such as REFERENCE


LPS, the degradation and phosphorylation of IκBα will
increase, which results in the release of free NF-κB p65 1. Asagiri, M., T. Hirai, T. Kunigami, S. Kamano, and H.J. Gober.
and translocation from the cytoplasm to the nucleus, 2008. Cathepsin K-dependent toll-like receptor 9 signaling revealed
in experimental arthritis. Science 319: 624–627.
leading to the transcription of specific target genes, such 2. Strandberg, Y., C. Gray, T. Vuocolo, L. Donaldson, M. Broadway,
as inflammatory-related cytokines, chemokines, adhesion and R. Tellam. 2005. Lipopolysaccharide and lipoteichoic acid
molecules, and other immune effector molecules [11, 12, induce different innate immune responses in bovine mammary
epithelial cells. Cytokine 31: 72–86.
38, 39]. The activation of JNK, p38, and ERK1/2 MAPK 3. Scull, C.M., W.D. Hays, and T.H. Fischer. 2010. Macrophage pro-
proteins is known to be involved in the regulation of inflammatory cytokine secretion is enhanced following interaction
iNOS and COX-2 expression [28]. MAPK signalling with autologous platelets. J Inflamm (Lond) 7: 53.
4. Vangroenweghe, F., P. Rainard, M. Paape, L. Duchateau, and C.
cascades have also been shown to be involved in the Burvenich. 2004. Increase of Escherichia coli inoculum doses
activation of NF-κB following LPS stimulation [23]. Our induces faster innate immune response in primiparous cows.
laboratory has recently reported that both the NF-κB and Journal of Dairy Science 87: 4132–4144.
5. Burvenich, C., V. Van Merris, J. Mehrzad, A. Diez-Fraile, and L.
MAPK pathways were activated in LPS-induced mastitis Duchateau. 2003. Severity of E. coli mastitis is mainly determined
[16, 40–42]. In the present study, we examined the effects by cow factors. Journal of Dairy Science 34: 521–564.
of thymol on the phosphorylation of IκBα and NF-κB 6. Rainard, P., and C. Riollet. 2006. Innate immunity of the bovine
mammary gland. Veterinary Research 37: 369–400.
p65 in the NF-κB signaling pathway, and the phosphor- 7. Lahouassa, H., E. Moussay, P. Rainard, and C. Riollet. 2007.
ylation of p38, ERK, and JNK in the MAPK signaling Differential cytokine and chemokine responses of bovine mammary
pathway. We found that pretreatment with thymol signif- epithelial cells to Staphylococcus aureus and Escherichia coli.
Cytokine 38: 12–21.
icantly suppressed the phosphorylation of NF-κB p65, 8. Schmitz, S., M.W. Pfaffl, H.H. Meyer, and R.M. Bruckmaier.
IκBα, p38, ERK, and JNK. The results demonstrated that 2004. Short-term changes of mRNA expression of various
thymol plays a central anti-inflammatory role in the inflammatory factors and milk proteins in mammary tissue
during LPS-induced mastitis. Domest Anim Endocrinology 26:
regulation of inflammatory responses, possibly through 111–126.
inhibiting the NF-κB and MAPK signaling pathways. 9. Chen, C.C., Y.T. Sun, J.J. Chen, and Y.J. Chang. 2001. Tumor
Therefore, the targeted inhibition of the induction and necrosis factor-alpha-induced cyclooxygenase-2 expression via
sequential activation of ceramide-dependent mitogen-activated
activity of NF-κB, COX-2, and/or iNOS has been protein kinases, and IkappaB kinase 1/2 in human alveolar epithelial
identified as a new therapeutic method in inflammatory cells. Molecular Pharmacology 59: 493–500.
diseases. The results in our study suggested that thymol 10. Boulanger, V., L. Bouchard, X. Zhao, and P. Lacasse. 2001.
Induction of nitric oxide production by bovine mammary epithelial
could promote the anti-inflammatory effect against LPS- cells and blood leukocytes. Journal of Dairy Science 84: 1430–
induced disfunction. Yet, these results are very primary, 1437.
and more work is needed to identify the exact target of 11. Karin, M., and Y. Ben-Neriah. 2000. Phosphorylation meets
ubiquitination: The control of NF-κB activity. Annual Review of
thymol in mammary epithelial cells. Immunology 18: 621–663.
In summary, our study demonstrated that the effect of 12. Wilson, S.J., B.A. Leone, D. Anderson, A. Manning, and S.T.
thymol on anti-inflammation not only comes from the Holgate. 1999. Immunohistochemical analysis of the activation of
NF-kappaB and expression of associated cytokines and adhesion
expression of inflammatory cytokines but also from the molecules in human models of allergic inflammation. The Journal of
iNOS and COX-2 pathway in LPS-stimulated mMECs. The Pathology 189: 265–272.
promising anti-inflammatory effect of thymol may involve in 13. Baeuerle, P.A. 1998. Pro-inflammatory signaling: Last pieces in the
NF-kappaB puzzle? Current Biology 8: R19–R22.
the inhibition of NF-κB and MAPK signaling pathways. Our 14. Han, J., J.D. Lee, L. Bibbs, and R.J. Ulevitch. 1994. A MAP kinase
results provide useful and additional mechanistic explana- targeted by endotoxin and hyperosmolarity in mammalian cells.
tions for the anti-inflammatory effects of thymol. Thereby, Science 265: 808–811.
15. Li, D., Y. Fu, W. Zhang, G. Su, and B. Liu. 2013. Salidroside
thymol may be a potential therapeutic agent against mastitis. attenuates inflammatory responses by suppressing nuclear factor-
kappaB and mitogen activated protein kinases activation in
lipopolysaccharide-induced mastitis in mice. Inflammation
ACKNOWLEDGMENTS Research 62: 9–15.
16. Fengyang, L., F. Yunhe, L. Bo, L. Zhicheng, L. Depeng, and
L. Dejie. 2012. Stevioside Suppressed inflammatory cytokine
This work was supported by grants from the National secretion by downregulation of NF-κB and MAPK signaling
Natural Science Foundation of China (no. 31272622 and no. pathways in LPS-stimulated RAW264. 7 cells. Inflammation
3120195) and the Research Fund for the Doctoral Program 35: 1669–1675.
17. Aeschbach, R., J. Loliger, B.C. Scott, A. Murcia, and J. Butler. 1994.
of Higher Education of China (no. 20110061130010 and Antioxidant actions of thymol, carvacrol, 6-gingerol, zingerone and
20120061120098). hydroxytyrosol. Food and Chemical Toxicology 32: 31–36.
Thymol Inhibits LPS-Stimulated Inflammatory Response

18. Robledo, S., E. Osorio, D. Muñoz, L.M. Jaramillo, and A. Restrepo. 31. Cao, X.Y., M. Dong, J.Z. Shen, B.B. Wu, C.M. Wu, and X.D. Du.
2005. In vitro and in vivo cytotoxicities and antileishmanial 2006. Tilmicosin and tylosin have anti-inflammatory properties via
activities of thymol and hemisynthetic derivatives. Antimicrobial modulation of COX-2 and iNOS gene expression and production of
Agents and Chemotherapy 49: 1652–1655. cytokines in LPS-induced macrophages and monocytes.
19. Ocana-Fuentes, A., E. Arranz-Gutierrez, F.J. Senorans, and G. International Journal of Antimicrobial Agents 27: 431–438.
Reglero. 2010. Supercritical fluid extraction of oregano (Origanum 32. Ogiwara, T. 2005. Radical scavenging activity of phenol, m-cresol
vulgare) essentials oils: Anti-inflammatory properties based on and thymol and their cytotoxicity. Journal of Meikai University
cytokine response on THP-1 macrophages. Food and Chemical School of Dentistry 33: 199–208.
Toxicology 48: 1568–1575. 33. Marsik, P., L. Kokoska, P. Landa, A. Nepovim, P. Soudek, and T.
20. Fachini-Queiroz FC, Kummer R, Estevao-Silva CF, Carvalho MD, Vanek. 2005. In vitro inhibitory effects of thymol and quinones of
and Cunha JM. 2012. Effects of thymol and carvacrol, constituents Nigella sativa seeds on cyclooxygenase-1-and-2-catalyzed prosta-
of Thymus vulgaris L. essential oil, on the inflammatory response. glandin E2 biosyntheses. Planta Medica 71: 739–742.
Evidence-Based Complementary and Alternative Medicine 2012: 34. Akira, S., and K. Takeda. 2004. Toll-like receptor signalling. Nature
657026. Reviews Immunology 4: 499–511.
21. Dhaneshwar, S., V. Patel, D. Patil, and G. Meena. 2013. Studies on 35. Gomez, P.F., M.H. Pillinger, M. Attur, N. Marjanovic, and M.
synthesis, stability, release and pharmacodynamic profile of a novel Dave. 2005. Resolution of inflammation: Prostaglandin E2
diacerein-thymol prodrug. Bioorganic & Medicinal Chemistry dissociates nuclear trafficking of individual NF-kappaB sub-
Letters 23: 55–61. units (p65, p50) in stimulated rheumatoid synovial fibroblasts.
22. Smalley, M.J. 2010. Isolation, culture and analysis of mouse The Journal of Immunology 175: 6924–6930.
mammary epithelial cells. Methods in Molecular Biology 633: 36. Yadav, P.N., Z. Liu, and M.M. Rafi. 2003. A diarylheptanoid from
139–170. lesser galangal (Alpinia officinarum) inhibits proinflammatory
23. Guha, M., and N. Mackman. 2001. LPS induction of gene mediators via inhibition of mitogen-activated protein kinase, p44/
expression in human monocytes. Cellular Signalling 13: 85–94. 42, and transcription factor nuclear factor-kappa B. Journal of
24. Nair, M.K., J. Joy, P. Vasudevan, L. Hinckley, T.A. Hoagland, and Pharmacology and Experimental Therapeutics 305: 925–931.
K.S. Venkitanarayanan. 2005. Antibacterial effect of caprylic acid 37. Chun, S.C., S.Y. Jee, S.G. Lee, S.J. Park, and J.R. Lee. 2007. Anti-
and monocaprylin on major bacterial mastitis pathogens. Journal of inflammatory activity of the methanol extract of Moutan cortex in
Dairy Science 88: 3488–3495. LPS-activated Raw264.7 cells. Evidence-Based Complementary
25. Stadnyk, A. 1994. Cytokine production by epithelial cells. The and Alternative Medicine 4: 327–333.
FASEB Journal 8: 1041–1047. 38. Bouwmeester, T., A. Bauch, H. Ruffner, P.O. Angrand, and G.
26. Aggarwal, B.B. 2003. Signalling pathways of the TNF super- Bergamini. 2004. A physical and functional map of the human TNF-
family: A double-edged sword. Nature Reviews Immunology 3: alpha/NF-kappa B signal transduction pathway. Nature Cell Biology
745–756. 6: 97–105.
27. Miao, J., Y. Fa, B. Gu, W. Zhu, and S. Zou. 2012. Taurine attenuates 39. Richmond, A. 2002. NF-κB, chemokine gene transcription and
lipopolysaccharide-induced disfunction in mouse mammary epithe- tumour growth. Nature Reviews Immunology 2: 664–674.
lial cells. Cytokine 59: 35–40. 40. Li, D., Y. Fu, W. Zhang, G. Su, B. Liu, and M. Guo. 2013.
28. Surh, Y.J., K.S. Chun, H.H. Cha, S.S. Han, Y.S. Keum, and K.K. Salidroside attenuates inflammatory responses by suppressing
Park. 2006. Molecular mechanisms underlying chemopreventive nuclear factor-κB and mitogen activated protein kinases activation
activities of anti-inflammatory phytochemicals: Down-regulation of in lipopolysaccharide-induced mastitis in mice. Inflammation
COX-2 and iNOS through suppression of NF-κB activation. Research 62: 9–15.
Mutation Research/Fundamental and Molecular Mechanisms of 41. Li, D., N. Zhang, Y. Cao, W. Zhang, and G. Su. 2013. Emodin
Mutagenesis 480: 243–268. ameliorates lipopolysaccharide-induced mastitis in mice by
29. Britten, N.J., Waldron, N.A., Watts, J.L., and Hallberg, J.W. 2003. inhibiting activation of NF-kappaB and MAPKs signal pathways.
Cyclooxygenase-2 inhibitor and antibacterial agent combination for European Journal of Pharmacology 705: 79–85.
intramammary treatment of mastitis, Google Patents, 2003. 42. Guo, M., N. Zhang, D. Li, D. Liang, and Z. Liu. 2013. Baicalin plays
30. Trakranrungsie, N., and J. Will. 1997. Expression of iNOS and an anti-inflammatory role through reducing nuclear factor-kappaB
COX-2 proteins during endotoxin-induced mastitis in guinea pigs. and p38 phosphorylation in S. aureus-induced mastitis.
FASEB Journal 11(3): 1829. International Immunopharmacology 16: 125–130.

You might also like