You are on page 1of 166

Thesis for the Degree of Doctor of Philosophy

Thymus vulgaris L. ameliorated atopic dermatitis


symptoms through controlling of histamine release

Seul A Seo

Graduate School of Biotechnology


Kyung Hee University
Seoul, Korea

February, 2021
Thymus vulgaris L. ameliorated atopic dermatitis
symptoms through controlling of histamine release

Seul A Seo

Graduate School of Biotechnology


Kyung Hee University
Seoul, Korea

February, 2021
Thymus vulgaris L. ameliorated atopic dermatitis
symptoms through controlling of histamine release

by
Seul A Seo

Advised by
Prof. Tae - Hoo Yi, O.M.D., Ph.D.

Submitted to the Graduate School of Biotechnology and the Faculty of the


Graduate School of Kyung Hee University in the partial fulfillment of
the requirements for degree of Doctor of Philosophy

Dissertation Committee :

Chairman Yeon Ju Kim

Tong Ho Kang

Se Chan Kang

Eunson Hwang

Tae-Hoo Yi
Contents

List of Table.........................................................................................................................

List of Figure.....................................................................................................................

Abbreviations...................................................................................................................vii

Abstracts.............................................................................................................................

Ⅰ. Introduction.........................................................................................................

1. Thymus vulgaris L.......................................................................................................1

2. Atopic dermatitis.........................................................................................................8

2.1. Definition of atopic dermatitis................................................................................9

2.2. Causes of atopic dermatitis.....................................................................................9

2.3. Symptoms of atopic dermatitis.............................................................................10

2.4. Pathogenesis of atopic dermatitis.........................................................................11

2.4.1. Imbalance of Th1/Th2 immune response.......................................................

2.4.2. Skin barrier dysfunction.................................................................................

2.5. Itching in atopic dermatitis...................................................................................15

2.5.1. Role of histamine in itching...........................................................................

2.5.2. Psychological stress and itching.....................................................................

3. Mechanism of atopic dermatitis...............................................................................24

3.1. Mast cells activation.............................................................................................24

i
3.2. Keratinocytes activation.......................................................................................28

4. Therapeutic agent of atopic dermatitis and allergic response................................31

4.1. Ketotifen fumarate...............................................................................................32

4.2. Tacrolimus...........................................................................................................33

4.3. Natural products-derived therapeurics..................................................................36

5. Purpose of this study.................................................................................................39

Ⅱ. Materials and Methods ............................................................................................41

1. Materials....................................................................................................................41

2. Sample preparation...................................................................................................42

3. HPLC analysis...........................................................................................................44

4. In vitro study..............................................................................................................45

4.1. Cell culture...........................................................................................................45

4.2. Sample treatment..................................................................................................45

4.3. Measurement of cell viability...............................................................................46

4.4. Measurement of NO production...........................................................................46

4.5. β-hexosaminidase release assay............................................................................47

4.6. Histamine release assay........................................................................................47

4.7. Measurement of intracellular calcium..................................................................48

4.8. Enzyme-linked immunosorbent assay (ELISA)....................................................48

4.9. Reverse transcription-polymerase chain reaction (RT-PCR).................................49

4.10. Western blot analysis.........................................................................................51


ii
5. In vivo study...............................................................................................................52

5.1. Experimental animals...........................................................................................52

5.2. Induction of AD-like skin lesion and topical application......................................52

5.3. Evaluation of AD-like skin symptoms..................................................................53

5.4. Measurement of physiological skin alteration......................................................55

5.4.1. Video scope...................................................................................................55

5.4.2. High resolution ultrasound............................................................................55

5.4.3. Hydration......................................................................................................56

5.4.4. TEWL............................................................................................................56

5.4.5. Erythema.......................................................................................................57

5.5. Evaluation of scratching behavior........................................................................57

5.6. Measurement of serum IgE level..........................................................................57

5.7. Histopathological analysis....................................................................................58

6. Statistical analysis.....................................................................................................59

Ⅲ. Results.......................................................................................................................60

1. The identification of active components from T. vulgaris.......................................60

2. The effects of T. vulgaris on atopic dermatitis and histamine related allergic

response: An in vitro study........................................................................................64

2.1. Effects of T. vulgaris on atopic dermatitis related inflammation in LPS-induced

Raw 264.7 cells.......................................................................................................64

iii
2.1.1. Cell viability..................................................................................................64

2.1.2. NO production...............................................................................................65

2.2. Effects of T. vulgaris on histamine related allergic response in DNP-IgE/BSA-

stimulated RBL-2H3 cells.....................................................................................67

2.2.1. Cell viability..................................................................................................67

2.2.2. β-hexosaminidase release..............................................................................68

2.2.3. Histamine release..........................................................................................69

2.2.4. Intracellular calcium levels............................................................................71

2.2.5. Signaling pathways in histamine release........................................................73

2.2.5.1. CRHR1 and CRHR2 expression.............................................................73

2.2.5.2. phosphorylation of PLCγ........................................................................75

2.2.5.3. phosphorylation of IP3R.........................................................................77

2.2.5.4. calcium channel protein expressions.......................................................79

2.3. Effect of T. vulgaris on atopic dermatitis related inflammation in TNF-α/IFN-γ-

stimulated HaCaT cells.........................................................................................81

2.3.1. Cell viability..................................................................................................81

2.3.2. Chemokine production..................................................................................82

2.3.3. mRNA expression of pro-inflammatory cytokine and

chemokines......................................................................................................84

2.3.4. Signaling pathways in pro-inflammatory cytokine and

iv
chemokines......................................................................................................86

2.3.4.1. MAPKs activation..................................................................................86

2.3.4.2. NFκB and STAT1 activation..................................................................88

3. The effects of T. vulgaris on atopic dermatitis and histamine related allergic

response: An in vivo study........................................................................................91

3.1. Morphological characteristics...............................................................................91

3.1.1. Body weight..................................................................................................91

3.1.2. Spleen weight................................................................................................93

3.1.3. Skin morphology...........................................................................................95

3.2. Physiological characteristics................................................................................98

3.2.1. Skin density...................................................................................................98

3.2.2. Skin hydration.............................................................................................100

3.2.3. TEWL..........................................................................................................101

3.2.4. Erythema index............................................................................................102

3.3. Itching related characteristics.............................................................................104

3.3.1. Scratching behavior.....................................................................................104

3.3.2. Serum IgE level...........................................................................................106

3.3.3. β-hexosaminidase and histamine release......................................................108

3.4. Signaling pathways in atopic dermatitis symptoms............................................110

3.4.1. CRHR1 and CRHR2 expression..................................................................110

v
3.4.2. MAPKs activation.......................................................................................112

3.4.3. IκBα and NFκB activation...........................................................................114

3.4.4. Filaggrin expression....................................................................................116

3.5. Histological characteristics.................................................................................118

3.5.1. Epidermal thickness.....................................................................................118

3.5.2. Accumulation of mast cells..........................................................................120

Ⅳ. Discussion................................................................................................................122

Ⅴ. Conclusion................................................................................................................135

Ⅵ. References...............................................................................................................136

국 문 초 록 ......................................................................................................................137

vi
List of Table

Table 1. Previous studies of T. vulgaris on biological effects...............................................4

Table 2. Previous studies of components of T. vulgaris on biological effects.......................7

Table 3. Indication and side effect of therapeutic agents.....................................................35

Table 4. Previous studies on the effect of natural products in AD.......................................37

Table 5. Previous studies on the effect of natural products in allergic skin

inflammation.................................................................................................................38

Table 6. List of primers for RT-PCR...................................................................................50

Table 7. Symptoms of AD...................................................................................................54

Table 8. Active components of T. vulgaris..........................................................................63

vii
List of Figure

Figure 1. Acute and chronic phases in AD..........................................................................15

Figure 2. Biosynthesis and metabolism of histamine...........................................................18

Figure 3. Effects of histamine through H1 and H4 receptors in skin allergic

reaction.........................................................................................................................19

Figure 4. Mechanism of mast cells activation.....................................................................27

Figure 5. Mechanism of keratinocytes activation................................................................30

Figure 6. Preparation of T. vulgaris extract.........................................................................43

Figure 7. HPLC analysis of T. vulgaris and caffeic acid and rosmarinic acid

standard.........................................................................................................................61

Figure 8. Effects of T. vulgaris on cell viability and NO production in LPS-induced Raw

264.7 cells...............................................................................................................66

Figure 9. Effects of T. vulgaris on cell viability, β-hexosaminidase, and histamine release in

DNP-IgE/BSA-stimulated RBL-2H3 cells...............................................................70

Figure 10. Effects of T. vulgaris on intracellular calcium levels in DNP-IgE/BSA-stimulated

RBL-2H3 cells........................................................................................................72

Figure 11. Effects of T. vulgaris on expression of CRHR1 and CRHR2 in DNP-IgE/BSA-

stimulated RBL-2H3 cells.......................................................................................74

Figure 12. Effects of T. vulgaris on phosphorylation of PLCγ in DNP-IgE/BSA-stimulated

viii
RBL-2H3 cells........................................................................................................76

Figure 13. Effects of T. vulgaris on phosphorylation of IP3R in DNP-IgE/BSA-stimulated

RBL-2H3 cells........................................................................................................78

Figure 14. Effects of T. vulgaris on expression of calcium channel proteins in

DNP-IgE/BSA-stimulated RBL-2H3 cells...............................................................80

Figure 15. Effects of T. vulgaris on cell viability and chemokine production in TNF-α/IFN-

γ-stimulated HaCaT cells..............................................................................................83

Figure 16. Effects of T. vulgaris on mRNA expression of pro-inflammatory cytokines and

chemokines in TNF-α/IFN-γ-stimulated HaCaT cells..............................................85

Figure 17. Effects of T. vulgaris on activation of MAPKs in TNF-α/IFN-γ-stimulated HaCaT

cells.........................................................................................................................87

Figure 18. Effects of T. vulgaris on NFκB signaling pathway in TNF-α/IFN-γ-stimulated

HaCaT cells..................................................................................................................89

Figure 19. Effects of T. vulgaris on STAT1 signaling pathway in TNF-α/IFN-γ-stimulated

HaCaT cells..................................................................................................................90

Figure 20. Effects of T. vulgaris on body weight in DFE-induced NC/Nga mice................92

Figure 21. Effects of T. vulgaris on spleen weight in DFE-induced NC/Nga

mice.............................................................................................................................. 94

Figure 22. Effects of T. vulgaris on skin morphology in DFE-induced NC/Nga

ix
mice.............................................................................................................................. 96

Figure 23. Effects of T. vulgaris on skin density in DFE-induced NC/Nga

mice.............................................................................................................................. 99

Figure 24. Effects of T. vulgaris on skin hydration, TEWL, and EI in DFE-induced NC/Nga

mice......................................................................................................................103

Figure 25. Effects of T. vulgaris on scratching behavior in DFE-induced NC/Nga

mice............................................................................................................................105

Figure 26. Effects of T. vulgaris on serum IgE level in DFE-induced NC/Nga

mice............................................................................................................................107

Figure 27. Effects of T. vulgaris on β-hexosaminidase and histamine release in DFE-induced

NC/Nga mice.........................................................................................................109

Figure 28. Effects of T. vulgaris on expression of CRHR1 and CRHR2 in DFE-induced

NC/Nga mice.........................................................................................................111

Figure 29. Effects of T. vulgaris on activation of MAPKs in DFE-induced NC/Nga

mice............................................................................................................................113

Figure 30. Effects of T. vulgaris on IκBα and NFκB in DFE-induced NC/Nga

mice............................................................................................................................115

Figure 31. Effects of T. vulgaris on filaggrin expression in DFE-induced NC/Nga

mice............................................................................................................................117

x
Figure 32. Effects of T. vulgaris on epidermal thickness in DFE-induced NC/Nga

mice............................................................................................................................119

Figure 33. Effects of T. vulgaris on accumulation of mast cells in DFE-induced NC/Nga

mice......................................................................................................................121

Figure 34. Inhibitory effects of T. vulgaris on mast cells activation..................................125

Figure 35. Inhibitory effects of T. vulgaris on keratinocytes activation............................128

Abbreviations

T. vulgaris Thymus vulgaris L.

AD Atopic dermatitis

TARC/CCL17 Thymus- and activation-regulated chemokine/C-C motif chemokine

ligand 17

MDC/CCL22 Macrophage-derived chemokine/C-C motif chemokine ligand 22

RANTES/CCL5 Regulated on activation, normal T cell expressed and secreted/ C-C

motif chemokine ligand 5

IL-6 Interleukin 6

IL-8 Interleukin 8

MAPK Mitogen-activated protein kinase

CRHR1 Corticotropin releasing hormone receptor 1

CRHR2 Corticotropin releasing hormone receptor 2

xi
DFE Dermatophagoides farinae

IgE Immunoglobulin E

SC Stratum corneum

TEWL Transepidermal water loss

Abstracts

Thymus vulgaris L. ameliorated atopic dermatitis


symptoms through controlling of histamine release

By Seul A Seo
Doctor of Philosophy in Science

Graduate School of Biotechnology, Kyung Hee University


Advised by Prof. Tae - Hoo Yi, O.M.D., Ph.D

Thymus vulgaris L., commonly known as thyme, is one of the aromatic herbs and has

been widely used worldwide. T. vulgaris has been reported to have antioxidant, anti-

inflammatory, and immunomodulatory activities, and particularly with regard to skin, anti-

photoaging and skin protective effects have been reported. However, there is no scientific

evidence about the inhibitory effects of T. vulgaris on AD. Thus, the present study was

performed to demonstrate the improving effects of T. vulgaris against AD symptoms.

The active components of T. vulgaris were detected by HPLC analysis. In the

in vitro study, the anti-atopic effects of T. vulgaris were examined in LPS-stimulated

xii
Raw264.7 cells, DNP-IgE/BSA-stimulated RBL-2H3 cells, and TNF-α/IFN-γ-stimulated

HaCaT cells using ELISA kits, RT-PCR, and western blotting. In the

in vivo study, the mitigating effects of T. vulgaris against AD were investigated by

measuring morphological and physiological changes, scratching behavior, AD symptoms

related protein expression, and histological changes in DFE-induced NC/Nga mice.

In the in vitro results, T. vulgaris significantly inhibited NO production in LPS-

stimulated Raw264.7 cells. It reduced the release of β-hexosaminidase and histamine via

regulation of calcium-related signaling pathway in DNP-IgE/BSA-stimulated RBL-2H3

cells. The production of TARC and MDC was remarkably suppressed by

T. vulgaris treatment in TNF-α/IFN-γ-stimulated HaCaT cells. Furthermore,

T. vulgaris influenced the decrease in mRNA expression of pro-inflammatory cytokine and

chemokines including IL-6, TARC, MDC, RANTES, and IL-8. It was shown to be due to the

modulation of MAPKs, STAT1, and NF-κB pathways. According to the in vivo results, it

was confirmed that topical application of T. vulgaris improves AD symptoms by reducing

the severity of dermatitis, TEWL, EI, scratching behavior, histamine release, epidermal

thickness, and infiltration of mast cells and elevating skin hydration and filaggrin

expression.

Taken together, this study revealed the ameliorating effects of T. vulgaris on AD-induced

in vitro and in vivo models. These findings suggest that T. vulgaris can be applied as a

xiii
natural product-derived therapeutic agent in the cosmetic and pharmaceutical industries, as

it attenuates itching by controlling histamine release, thereby improving symptoms of AD.

Keywords Thymus vulgaris L., atopic dermatitis, histamine, itching

xiv
Ⅰ. Introduction

1. Thymus vulgaris L.

Thymus vulgaris (T. vulgaris), commonly known as garden thyme, belong to the genus

Thymus (Lamiaceae family) and is native to Europe and Asia. It is highly aromatic, has

grey-green leaves, and blooms with purple or pink flowers in early summer.

T. vulgaris is considered to have high nutritional value because it is abundant in minerals

and vitamins that are beneficial to health. Vitamin A and vitamin C are abundant in

T. vulgaris, and vitamin B6 or pyridoxine, vitamin K, vitamin E, and folic acid are also

present. The health-promoting properties of T. vulgaris are known to be due to the

antioxidant and anti-inflammatory effects.

T. vulgaris is one of the most widely used spices in the world, characterized by having a

fragrant odor and a pungent taste. Because the flavor of T. vulgaris lasts for a long time, it

has an appetite-inducing effect and has been used in dishes such as soups, meat, pork

sausages, fish, and poultry dressings. T. vulgaris essential oil is used in aromatherapy, which

is known to be able to alleviate bites and stings, rheumatic aches, and pains. In addition, it

helps blood circulation to smooth the skin and is effective in acne treatment due to its

antibacterial activity.

T. vulgaris is one of the best known traditional medicines that has been applied in the

1
form of tea, ointment, tincture, syrup or steam inhalation. Since T. vulgaris is believed to be

effective in treating respiratory diseases such as dry coughs, asthma, and bronchitis, it has

been traditionally used for the treatment of these diseases. It is also known that vitamin C

present in T. vulgaris helps to improve immunity.

According to recent reports, it was verified that T. vulgaris essential oil and carvacrol, its

components, exert anti-inflammatory effects by allowing inhibition of inflammatory edema

and leukocyte migration. In oxLDL-Stimulated THP-1-Macrophages model,

T. vulgaris suppressed the production of proinflammatory mediators such as TNF-α, IL-1B,

and IL-6. It was revealed that T. vulgaris has a potential immunomodulatory role,

antioxidant activity, and a protective effect against plumbum (Pb) toxicity. In addtion,

antibacterial, antifungal, and neuroprotective effects of T. vulgaris have been reported.

Especially, in relation to the effect of T. vulgaris on skin, Sun et al. (2016) reported that

T. vulgaris is effective in improving skin wrinkles via inhibition of MAPK/AP-1 and

activation of NRF2-ARE antioxidant system. It was also demonstrated that

T. vulgaris and thymol, one of its active compounds, can reduce DNA damage induced by

UVA and UVB in NCTC 2544 cell line and protect skin against genotoxic damage caused by

UV radiation in an ex vivo human skin model. Moreover, study showed that T. vulgaris is

effective in the treatment of cutaneous leishmaniasis in balb/c mice. In relation to antifungal

2
activity, T. vulgaris essential oil and thymol has a therapeutic effect on experimentally

induced dermatomycoses in rats. However, few studies on skin inflammation have been

reported.

3
Table 1. Previous studies of T. vulgaris on biological effects

Reference
Biological effect Model
PMIDa

Antioxidant, UVB-irradiated NHDFs and UVB exposed hairless


27641753
Anti-aging mice
UVA and UVB-irradiated NCTC 2544 cell line 26338540
Skin protective UVA and UVB exposed human skin model (ex
27023828
vivo)
ear edema, carrageenan induced pleurisy, and
Anti-inflammatory 22919415
chemotaxis in vitro.
Anti-inflammatory oxLDL-Stimulated THP-1-Macrophages 22577523
Anti-leishmaniasis cutaneous leishmaniasis in balb/c mice. 19248657
Antimicrobial, Gram-negative bacteria (E. coli), gram-positive
antioxidant, bacteria (S. aureus and P. acnes), and fungi (C. 21979069
anti-inflammatory albicans and P. ovale), LPS-induced THP-1 cells
Immunomodulatory,
Lead (Pb)-intoxicated rats 31172438
antioxidant
Dermatomycoses induced rats 18651285
Antifungal
Fusarium oxysporum strains 28062283
Antibacterial Phytopathogen Allorhizobium vitis 29288304
Neuroprotective 5-fluorouracil (5-FLU) administered rats 31475590

PMIDa; PubMed identifier (PMID) is a unique number assigned to each PubMed record.

4
It was reported that T. vulgaris contains monoterpene phenols including thymol (10-

64%), p-cymene (9.1-22.2%), and carvacrol (0.4-20.6%). In addition, it also contains various

phenolic compound, such as rosmarinic acid, cymaroside, and caffeic acid.

Thymol is a major component of essential oils and is a naturally occurring phenol

monoterpene derivative of cymene and isomer of carvacrol. Many studies have been reported

on the biological effect of thymol, which has been shown to exert antioxidant effects through

the regulation of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione

peroxidase (GPx), and to inhibit the production of elastase. In addition, it was demonstrated

that thymol has anti-inflammatory effects which act by reduction of LPS-stimulated

inflammation and edema and alleviate atopic dermatitis induced by Staphylococcus aureus

membrane vesicles. p-cymene is a monocyclic monoterpene which regulates the expression

of pro-inflammatory cytokines through suppression of MAPK and NF-B, thereby showing

anti-inflammatory activity. Also, it has antioxidant activity via reduction of lipid

peroxidation and nitrite content and enhancement of SOD and catalase activity.

Carvacrol has a positive effect on the reduction of interleukin (IL)-1β, IL-4, IL-8 and

malondialdehyde (MDA) and immunomodulatory effects against ovalbumin-induced asthma

in rats.

Studies have revealed that rosmarinic acid alleviates atopic dermatitis and inhibits

5
reactive oxygen and nitrogen species and suppresses hydrogen

peroxide-induced oxidative stress and inflammatory response in normal human dermal

fibroblasts via down-regulation of NF-κB transcriptional activity. Cynaroside, also known as

luteolin 7-O-glucoside, enhances the antioxidant and antiapoptotic ability in ARPE-19 cells

by promoting the expression of p-Akt and attenuates inflammation through regulation of the

PPARγ/Nrf2/NF-κB signaling pathway. Jeon et al. demonstrated the anti-inflammatory

effect of caffeic acid through down-regulation of chemokines. Moreover, caffeic acid has

been found to induce cutaneous wound healing and restore the antioxidant defense system.

6
Table 2. Previous studies of components of T. vulgaris on biological effects

Reference
Component Structure Biological effects
PMIDa

Antioxidant 10703468
Thymol Anti-atopic 29679854
Anti-inflammatory 22919415

25856703
p-Cymene Anti-inflammatory
22486037

Antioxidant 32326410
Carvacrol
Immunomodulatory 31881223

Anti-atopic 19239556
Rosmarinic
Antioxidant 16087481
acid
Anti-inflammatory 29039587

Antioxidant 31157476
Cynaroside
Anti-inflammatory 31810125

Anti-allergic 26104582
Caffeic acid Wound healing 26929003
Anti-aging 22360712

PMIDa; PubMed identifier (PMID) is a unique number assigned to each PubMed record.

7
2. Atopic dermatitis

Atopic dermatitis (AD) is one of the allergic skin disorders that is associated with the

immune system. It is accompanied by itching, erythema, cracked skin, and swelling. The

activation of keratinocytes is known as a major characteristics of AD. It results in the

production of pro-inflammatory cytokines which promote chronic and self-amplifying

immune activation. For this reason, keratinocytes play a pivotal role in accelerating and

maintaining inflammation in AD.

AD is known as an immediate type hypersensitivity reaction mediated by the mast cells.

Mast cells are key effectors in IgE-mediated immediate allergic disease such as asthma and

AD. Mast cells are distributed in various tissues and cause the release of various mediators

such as histamine and pro-inflammatory cytokines through degranulation. In particular,

histamine is related to immediate phase of allergic inflammation which presents as

symptoms such as vasodilatation, increased vascular permeability and tissue edema. The

activation of mast cell-derived factors contributes to itching and inflammation in AD skin.

8
2.1. Definition of atopic dermatitis

Atopic dermatitis (AD) is a chronic and relapsing inflammatory skin disorder,

accompanied by eczema, itching, edema, and skin thickening. AD is increasing in adults as

well as in children and is considered a major problem not only in developed countries but

also in developing countries. Therefore, AD is one of the serious diseases of modern society

which causes mental stress and worsens the quality of life. Although many studies on AD

have been conducted, the pathological and genetic mechanisms of AD are not fully

understood.

2.2. Causes of atopic dermatitis

The exact causes of AD are not known, but is thought to be a complex disease triggered

by interaction of several factors. Genetic factors are known to include mutations in the

filaggrin gene, which is related to the skin barrier and environmental factors, including

stimuli such as air pollution, chemicals, irregular sleep, and stress. It is also associated with

immunomodulatory abnormalities caused by the destruction of the balance of the Th1 / Th2

immune response.

9
2.3. Symptoms of atopic dermatitis

Diagnosis of AD is based on clinical features, requiring itchy skin and at least three of

the following: history of involvement of the skin creases, history of asthma or hay fever,

history of generally dry skin in the past year, onset in a child under two years of age, and

visible flexural dermatitis. Acute skin lesions include erythematous macules and papules,

which are related to excoriations and erosions. In the chronic phase, lichenification occurs

after scratching the skin for a long time.

Clinical manifestations of atopic dermatitis are very diverse, and are classified into

infant, pediatric, and adult types according to age. In infancy, skin lesions occur on the scalp

or face, especially presenting as acute eczema such as erythema, edema, and swelling, and

often worsen suddenly. In childhood, skin lesions tend to occur on the flexures and dorsal

aspects of the limbs. In adolescents and adults, lichenification is observed as a result of skin

fibrosis and increased collagen erosion in the flexure, head and neck.

10
2.4. Pathogenesis of atopic dermatitis

In normal skin, the Th1 and Th2 type immune responses are balanced to maintain

immunological homeostasis, and the skin barrier function is normally performed to protect

against external invasion. On the other hand, in AD skin, immunological homeostasis is

collapsed, resulting in an imbalance of immunity, and destruction of the skin barrier

increases invasion of external irritants and causes an inflammatory reaction.

2.4.1. Imbalance of Th1/Th2 immune response

The infiltration of T helper cells (Th cells) is characteristic of AD skin lesions.

According to function, these cells can be divided into T helper type 1 (Th1) cells and T

helper type 2 (Th2) cells, which can cause an inflammatory reaction of the skin depending

on the activity and duration of skin lesions.

In the AD skin lesions, there is a difference in the expression of cytokine depending on

the phase. In the acute phase, Th2 cells secrete cytokines, including IL-4, IL-5, and IL-13,

and this activates mast cells to promote the secretion of soluble mediators such as histamine.

It stimulates B cells to promote the production of IgE. Eczematous skin lesions are known to

be characteristic of the acute phase. On the other hand, in the chronic phase, the production

of Th1 cells is increased compared Th2, resulting in increased expression of IFN-γ, IL-12,

IL-5, and granulocyte-macrophage colony-stimulating factor (GM-CSF). The characteristics

11
of the chronic phase include lichenification of skin and tissue remodeling. Since AD is

accompanied by severe itching, the Th2 type immune response is converted into a Th0 type

immune response due to the wound caused by scratching the lesion site as the disease

progresses, resulting in chronic inflammation. Therefore, the response by Th2 cell activation

is characteristic of the acute phase, and the response by Th1 cells and/or Th0 cells activation

is characteristic of the chronic phase. This indicates that Th2 as well as Th1 play an

important role in inducing and maintaining skin inflammation in AD skin.

Recently, the role of Th17 cells related cytokines in the pathogenesis of AD has been

reported. In the lesions of AD, Th17 cells are infiltrated, leading to the expression of Th17

related cytokines such as IL-17 and IL-22. They are recognized as a critical factors because

they serve to mediate the onset and progression of AD.

2.4.2. Skin barrier dysfunction

The skin is the most external organ of the human body; it prevents water loss in the body

and acts as a barrier to protect against external stimuli such as irritants, allergens, and

pathogens. However, in AD skin, the functioning of the skin barrier is impaired, which is

recognized as one of the most important factors in the pathogenesis of AD. Damage to the

skin barrier increases transepidermal water loss (TEWL), causing a decrease in the moisture

content of the stratum corneum, resulting in dry, scaly, rough, dull, slightly wrinkled skin.

12
This is due to the mutations of structural proteins such as filaggrin in keratinocytes, which

increases protease activity or decreases the activity of protease inhibitor and also causes

abnormal lipid composition.

Filaggrin is one of the most important elements in forming the skin barrier, and is a

protein produced by the degradation of profilaggrin, a high molecular weight protein

constituting keratohyalin granules present in epidermal granule cells. Filaggrin acts to

aggregate keratin in keratinocytes, and when degraded, it turns into a natural moisturizing

factor such as free amino acids, urocanic acid, and pyrrolidone carboxylic acid, which

maintain moisture in the stratum corneum. In this way, the mutation of filaggrin gene, which

plays an important role in skin barrier function, not only causes abnormal keratinocyte

structure and skin barrier dysfunction, but also reduces the moisture content of the stratum

corneum and increases TEWL. This is responsible for causing dry skin in AD, which may

increase external irritation and worsen AD.

13
Figure 1. Acute and chronic phases of AD

14
2.5. Itching in atopic dermatitis

2.5.1. Role of histamine in itching

Histamine (2-[4-imidazolyl]-ethylamine) is a biogenic amine and is well known as a

crucial mediator of numerous biological reactions. Histamine is usually secreted from mast

cells and basophils, which are the main cellular sources of histamine and can store it in

specific granules. It has also been found that histamine is produced in T cells and

keratinocytes in response to stimulation, which cannot store histamine.

Histamine synthesized from the amino acid histidine by the histidine decarboxylase

(HDC) is stored in the secretory granules of both mast cells and basophils. Stimuli activate

these cells, and then stored histamine is released. It results in diverse biological effects

through the activation of four distinct receptors (H1R, H2R, H3R, and H4R). Histamine

receptors are classified as G protein coupled receptor and various functions of histamine are

attributed to these four pharmacologically distinct receptors. The activation of H1R by

histamine is known to cause cellular migration, nociception, vasodilatation, and

bronchoconstriction, and also involved in Th1- and Th2-type immune responses. H2R plays

a role in regulating gastric acid secretion, airway mucus production, and vascular

permeability. H3R plays an important role in neuro-inflammatory diseases by regulating the

15
release of histamine, serotonin, acetylcholine, and other

neurotransmitters. H4R is mainly expressed in immune cells such as leukocytes and mast

cells. Inflammation and pruritus were mediated by receptors that play an important role in

chronic allergic contact dermatitis. Mast cell activation by H4R regulates the inflammatory

cascade through the release of the several inflammatory mediators. In addition, H4R was

reported to affect chemotaxis in mast cells and eosinophils. Especially with regard to skin,

the activation of H1R affects most immediate hypersensitivity reactions such as erythema,

pruritus, and edema. In addition, H1R promotes the proliferation of keratinocytes and causes

periostin release in fibroblasts. It was reported that histamine is also involved in itch

sensation of sensory neuron through H1R and H4R.

16
Figure 2. Biosynthesis and metabolism of histamine

17
Figure 3. Effects of histamine through H1 and H4 receptors in skin allergic reaction.

18
Histamine is known to be involved in various biological reactions. In the central nervous

system, histamine dilates blood vessels, causing dizziness, headache, vomiting and

insomnia. In addition, vasodilation causes arrhythmia, hypotension, and shock symptoms in

the cardiovascular system. In relation to the stomach and intestine, abdominal pain, diarrhea

and abdominal swelling due to gastric acid secretion appear, and is involved in smooth

muscle contraction, causing menstrual pain and urination. In the respiratory system,

histamine increases mucous secretion and vascular permeability, resulting in rhinitis and

coughing.

Especially in relation to the skin, symptoms such as itching, facial flushing and urticaria

are caused by histamine through increased vascular permeability and pain-sensitive

disorder, stimulation of nerve fibers. Histamine plays an important role in inflammatory skin

disease; initial reddening of skin, plasma extravasation and the development of a weal

(tissue oedema) and a flare (wider spread erythema) are considered features of skin

inflammation by histamine. These reactions are often accompanied by itching. Histamine

released from mast cell granules induces inflammation, which is mostly associated with

immediate hypersensitivity. In addition to causing skin inflammation and itching, histamine

has also been reported to affect skin barrier function. Histamine inhibits epidermal

differentiation and disturbs skin barrier function by reducing the formation of tight junctions

19
and the expression of filaggrin, and promotes proliferation of keratinocytes, leading to

hyperkeratosis.

Allergy symptoms further stimulate the release of histamine, which in turn leads to the

deterioration of the disease such as AD. Itching is hallmark symptom of AD, which causes

an unpleasant sensation and a desire to scratch, aggravating the cutaneous symptoms.

Patients with AD usually experience itching and scratching, which triggers psychological

problems such as sleep disorders and depression. This has a significant impact on the quality

of a patient’s life. Although there are various substances that trigger itching, such as

cytokines and substance P, histamine is still recognized as a major pruritogen. It is

responsible for at least half of the symptoms and signs of allergic reactions in the skin. Since

mast cells are a major source of stored histamine, the improvement of allergy symptoms and

immune dysfunction depends on the regulation of histamine release from mast cell. Upon

stimulation by an allergen, histamine is released from mast cells through degranulation,

which causes the excitation of a subset of unmyelinated C-fibers, leading to itching.

20
2.5.2. Psychological stress and itching

Psychological stress is general phenomenon reported to have a significant effect on

immune function. It is known as a risk factor for immune related diseases such as AD. The

regulation of immune and nerve systems by psychological stress can affect itching, which

results in worsening the condition of inflammatory diseases. Thus, stress is a main

contributor to aggravate itching in a variety of itching-associated diseases. There is a stress-

itching-scratching cycle that stress can causes or aggravates itching, in turn leading to

scratching and further itching. This can worsen stress; indeed, itching is associated with

anxiety and stress sensitivity in patients with AD, causing high levels of anxiety and stress in

patients with chronic and severe itching. Patients with chronic itching react sensitively to

itching-scratches cycles, which can lead to an increase in stress levels, eventually

aggravating itching. This vicarious cycle results in continuous scratching, deterioration of

diseases, and impaired quality of life. The central and peripheral activation of the

hypothalamic-pituitary-adrenal (HPA) axis and sympathetic nervous system are involved

with the induction or deterioration of itching caused by stress. Mast cells, keratinocytes, and

nerves are affected by these systems, which promote the secretion of neuropeptides including

histamine, substance P, and itchy cytokines.

21
In response to stress, CRH can be secreted from keratinocytes and mast cells. In

keratinocytes, the release of many mediators associated with the pruritogenic pathway can be

caused by CRH. Furthermore, CRH induces the secretion of the mediators that activate mast

cells. When mast cells are activated, histamine and cytokines, characterized by inflammatory

and vasodilatory mediators, are secreted. This eventually leads to itching. Inflammation

induces a reduction in the threshold for itching stimuli, which results in peripheral itching

sensitization.

Many skin disorders, such as AD, are correlated with an increase in the number of

activated mast cells, which tend to be further exacerbated by stress. Thus, mast cells are

recognized as a key immune effectors in the stress response. Mast cells manifest various

receptors, which rapidly sense and respond to stress signals from environmental and

immunological factors. Under stress, the release of mediators such as β-hexosaminidase and

histamine in mast cells is promoted through degranulation. In particular, histamine induces

an increase in vascular permeability and stimulates the cutaneous sensory nerves, which in

turn causes itching. Excessive activation of mast cells has a detrimental effect that is

associated with the onset and severity of diseases such as allergy.

Similar to the central axis, the peripheral HPA axis exists in the skin. This plays a key

role in the response to psychological stress. CRH is a major component of the HPA axis, and

22
is important in controlling the response to stress. CRH is known to regulate the stress

response through the two receptors, CRHR1 and CRHR2. According to a recent study, it

has been demonstrated that CRHR1 acts as a positive regulator in mast cell degranulation

induced by stress. CRHR1 is mainly involved in the anxiogenic effect of CRH. In contrast,

CRHR2 serves to inhibit mast cell degranulation via calcium related signaling pathways.

CRHR2 is involved in reduced anxiety-like behaviors and stress sensitivity. Since CRH and

related peptide are activated by mast cells, it is believed that mast cells serve an important

function as a sensor of psychological stress. Therefore, the regulation of these receptors can

be targeted in treating immune disorders that is associated with stress and mast cell

hyperactivity such as allergic inflammation.

23
3. Mechanism of atopic dermatitis

3.1. Mast cell activation

Mast cells play a pivotal role in allergic inflammatory reactions because the number of

mast cells and activation of mast cells are increased in AD lesions. The stimulation of

antigen presenting cells differentiate Th2 cells, which promotes the production of IgE in B

cells. Subsequently, IgE binds to high affinity IgE receptor (FcεRI) present on the surface of

mast cells, which activates these cells. It induces degranulation and promotes production of

inflammatory cytokines resulting in an allergic reaction. In an allergic reaction, mast cells

secret a variety of biological mediators including preformed secretory granules (containing

β-hexosaminidase and histamine), cytokines, and chemokines. When mast cells are

activated, β-hexosaminidase and histamine present in pre-stored vesicles are released by the

fusion of granule and plasma membrane during the acute phase, resulting in the delayed

production and secretion of inflammatory cytokines.

For mast cell activation, mobilization of Ca 2+ is a key process. Ca 2+ plays an important

role as a secondary messenger in the mast cell signaling pathways that modulates a variety

of cellular responses. According to the intracellular calcium level, the exocytosis of the mast

cell granules and secretion of the inflammatory cytokines are regulated by the

Ca2+ influx from the extracellular space. Upon allergen-mediated stimulation, CRHR1 is

24
activated, but CRHR2 is suppressed. This stimulates phosphorylation of phospholipase Cγ

(PLCγ), leading to the generation of inositol 1,4,5-trisphosphate(IP3), which binds to IP3

receptors present in the endoplasmic reticulum (ER) to induce intracellular calcium release

from ER. The release of ER Ca 2+ mediated by IP3 causes the emptying of intracellular

Ca2+ stores, subsequently activating a Ca 2+ influx to refill intracellular stores through plasma

membrane channels.

Store-operated Ca 2+ entry (SOCE) is known as a major mechanism of Ca 2+ influx. For

SOCE, stromal interaction molecule 1 (STIM1) present in the ER membrane plays a crucial

role as an endogenous Ca 2+ sensor. It senses the low concentration of Ca 2+ in ER, and

subsequently forms a complex with calcium release-activated calcium channel protein 1

(Orai1). Orai1 is one of the store-operated channels and exists in plasma membrane that is

regulated by the level of Ca 2+ in ER stores. As store-operated channels, there is also

transient receptor potential canonical channel 1 (TRPC1) which is involved in the

Ca2+ influx. As a result, the increase of cytosolic Ca 2+ level leads to the fusion of preformed

granules with the plasma membrane. This rapidly induces the degranulation of mast cells via

Ca2+ mediated exocytosis and promotes the secretion of inflammatory mediators such as

histamine, β-hexosaminidase, proteases, proteoglycans, and lipid mediators, thereby leading

25
to allergic inflammatory responses. It is believed that keratinocytes can be affected by

activation of mast cells.

26
Figure 4. Mechanism of mast cells activation

27
3.2. Keratinocytes activation

Keratinocytes present in the epidermis play an important role in the pathogenesis of

inflammatory skin diseases such as AD. When keratinocytes are exposed to stimuli such as

TNF-α and IFN-γ, cytokines and chemokines including thymus- and activation regulated

chemokine (TARC/CCL17), macrophage-derived chemokine (MDC/CCL22), regulated on

activation, normal T-cell expressed and secreted (RANTES/CCL5), and interleukin 8

(IL-8/CXCL8) are abnormally expressed. As a result, they increase the infiltration of Th

cells into local lesions. In the keratinocytes of AD skin, a number of cytokines and

chemokines involved in the response of immune cells is generated, and as a result,

inflammation of the skin lesions is exacerbated. Thus, these mediators are considered as

conclusive modulators in the pathogenesis of AD. It has been found that the regulation of

inflammatory responses related to AD is influenced by nuclear factor-kappaB (NF-κB),

janus kinase (JAK)/signal transducer and activator of transcription (STAT), and mitogen-

activated protein kinase (MAPK).

TNF-α is known to be involved in the activation of diverse signaling pathway such as

NF-κB, ERK, JNK, and p38 MAP kinases. In the resting state, NF-κB, a key transcription

factor of the inflammatory response, binds to IκB in the cytoplasm. Under the stimulation,

the IKK complex is activated, and IκB is subsequently phosphorylated and degraded. The

28
free NF-κB is released and translocated into the nucleus, which is responsible for activating

the expression of various genes associated with pro-inflammatory cytokines and

chemokines. In addition, when keratinocytes are stimulated, the MAPK signaling pathway is

activated, to subsequently upregulate NF-κB, resulting in acceleration of proinflammatory

cytokines and chemokines expression.

IFN-γ is important in the regulation of the immune responses because it is characterized

by possessing immunomodulatory activity. In the IFN-γ signaling pathway, IFN-γ binds to

its receptor and induces the activation of JAK, and then phosphorylates STAT1. STAT1 is a

member of the STAT protein family and this phosphorylation has been found to play a

crucial role in IFN-γ mediated inflammatory responses. When STAT1 is phosphorylated, it

translocates from the cytoplasm into the nucleus and then promotes the transcription of

genes associated with inflammation such as MDC. Thus, it is regarded that STAT1 is a

significant regulator of the IFN-γ signaling pathway.

29
Figure 5. Mechanism of keratinocytes activation

30
4. Therapeutic agents of atopic dermatitis and allergic response

Ketotifen is a relatively selective, non-competitive histamine antagonist (H1R) and mast

cell stabilizer. Ketotifen inhibits the release of mediators from cells involved in

hypersensitivity reactions. Decreased chemotaxis and activation of eosinophils has also been

demonstrated. Ketotifen has been shown to have little systemic exposure following topical

ocular administration. In human conjunctival allergen challenge studies, ketotifen fumarate

was significantly more effective than placebo in preventing ocular itching associated with

allergic conjunctivitis. The action of ketotifen occurs rapidly with an effect seen within

minutes after administration. Several studies have examined the efficacy of topical

tacrolimus in adult patients. In a double-blind, placebo-controlled study, 215 patients with

moderate to severe atopic dermatitis were randomized to receive 0.03, 0.1, or 0.3 percent

tacrolimus, or vehicle, applied twice daily. Patients were evaluated at three weeks using

atopic dermatitis severity scores. The decrease in scores for the three groups was 67, 83, and

75 percent for the 0.03, 0.1, and 0.3 percent ointments, respectively. The vehicle group

showed a 22 percent decrease in severity scores. The only statistically significant side effect

in treatment versus vehicle groups was a sensation of local burning, but this effect resulted in

no increase in the dropout rate.

31
4.1. Ketotifen fumarate

Ketotifen, well known as mast cell stabilizer, is a second-generation non-competitive H1

antihistamine. The pharmacodynamic properties of ketotifen are many, because it is an

inhibitor of the release and/or activity of mast cell and basophil mediators, including

histamine, neutrophil, and eosinophil chemotactic factors, arachidonic acid metabolites,

prostaglandins, and leukotrienes. The ocular, nasal, and dermal responses to applied allergen

in sensitized patients are attenuated with use of ketotifen. Oral ketotifen has been used in

patients with asthma, allergic rhinitis, allergic conjunctivitis, atopic dermatitis, chronic

urticaria, cold-induced urticaria, cholinergic urticaria, exercise-induced urticaria,

mastocytosis, and food allergy. For adults and older children with asthma or allergic

disease, the recommended dose of ketotifen is 1 mg twice daily. For young children 6

months to 3 years old, the recommended dose is 0.5 mg twice daily. Oral ketotifen is usually

well tolerated and relatively safe, with the most frequent side effect of sedation. This is seen

in approximately 10% to 20% of patients, especially at higher doses, but the effect decreases

within 1 to 2 weeks of use.3 Other rarer reactions include dizziness, dry mouth, nausea, and

headache, which have been reported in 1% to 2% of patients at initiation of therapy.

However, these side effects do not persist in patients on long-term treatment.

32
4.2. Tacrolimus

Tacrolimus is a calcineurin inhibitor and possess immunomodulatory and anti-

inflammatory properties. The mechanism of action of tacrolimus in allergic diseases

involves calcineurin inhibition, and downregulation of T-cell reactivity, IgE degranulation,

and its actions on mast cells, dendritic cells, basophils, eosinophils and inhibition of

transcription of proinflammatory cytokines.

Tacrolimus inhibits cytokine production following T cell activation and decreases Fc

epsilon RI expression on dendritic cells in the skin in atopic dermatitis. Tacrolimus targets

Ca2+-dependent protein phosphatase, calcineurin. The antigens bind to T-cell receptors which

result in increased production of Ca 2+. The increased intracellular Ca 2+ binds with calmodulin

and activate and phosphorylates nuclear factor of activated T Cells. The dephosphorylated

form of cytoplasmic NF-AT translocate from the cytosol into the nucleus and forms a

complex with the nuclear subunit of NF-AT, and can bind to the promoter region of several

cytokine genes and induce gene transcription. On the other hand, after penetrating the cell

membrane, tacrolimus binds to its intracellular receptor, the FK-binding protein complex and

blocks the function of the Ca 2+ and calmodulin-dependent phosphatase, calcineurin. This

interaction results in suppression of NF-AT-dependent cytokine gene transcription and

immunosuppression. Thus tacrolimus inhibits transcription of cytokines and suppresses T-

33
cell proliferation. Allergic diseases of the biological system comprise a spectrum of

diseases, with each condition being characterized by a complex immunopathology.

Tacrolimus is a potent immunomodulator that is effective in the treatment of various allergic

diseases by its pleiotropic immunosuppressive effects on the immune system.

34
Table 3. Indication and side effect of therapeutic agents

Therapeutic agent Indication Side effects

Allergic conjunctivitis
- Increased nosebleeds
Asthma
- Irritability
Ketotifen fumarate Atopic dermatitis
- Dry mouth
Chronic urticaria
- Drowsiness
Allergic rhinitis

Atopic dermatitis - Burning sensation


- Folliculitis
Contact Hypersensitivity
- Acne
Tacrolimus
Organ transplantation - Eczema herpeticum
- Herpes simplex infections
Ulcerative colitis
- Neurotoxicity

35
4.3. Natural products-derived therapeutics

Many studies have been performed on atopic dermatitis, but no complete treatment has

yet been found. Since atopic dermatitis is caused by various factors, different treatments may

be applied to each patient. The current therapeutics are known to have side effects if applied

chronically. However, natural products have fewer side effects and contain components with

various biological functions. For this reason, many studies have been conducted on the

effect of natural products for atopic dermatitis (Table 4 and 5).

36
Table 4. Previous studies on the effect of natural products in AD

Natural products Reference


Effect
(Korean name) PMIDa

Reduce histological manifestations of atopic skin


Saussurea lappa
lesions such as erosion, hyperplasia of the epidermis 24216625
(Mok-hyang)
and dermis, and inflammatory cell infiltration.
Suppress TNF-α/IFN-γ-induced production of TARC
and MDC in HaCaT cells by inhibiting MAPK
Hovenia dulcis
signaling and regulate immunoglobulin (Ig) E and
Thunb. 27696405
immunoglobulin G2a (IgG2a) levels in serum and
(Heot-gae-na-mu)
the expression of mRNA for Th1- and Th2-related
mediators in skin lesions.
Inhibited the expression of TNF-a, CCL17, IL-1b,
Moringa oleifera
and IL-6 mRNA, as well as that of MAPKs in
leaf 27744247
HaCaT keratinocytes in a concentration-dependent
(Moringa)
manner.
Patrinia scabiosifolia Attenuate the development of AD-like lesions by
Link increasing filaggrin expression and lowering IgE 28347830
(Ma-ta-ri) and inflammatory cytokine levels.
Regulate chemokine formation by inhibiting
Artemisia apiacea
activation of and ERK as well as the NF-κB
Hance 29932162
pathways and improve the skin p38 conditions in a
(Gae-sa-cheol-ssuk)
DNCB-induced AD-like mouse model

PMIDa; PubMed identifier (PMID) is a unique number assigned to each PubMed record.

37
Table 5. Previous studies on the effect of natural products in allergic skin inflammation

Natural products Reference


Effect
(Korean name) PMIDa

The release of histamine from mast cells was


Pogostemon cablin
reduced by AEPC, and this suppressive effect was 26531835
(Patchouli)
associated with the regulation of calcium influx.
F-AFE suppressed the production of TNF-α and
Arctium lappa PGE2 in a dose-dependent manner. F-AFE inhibited
26707911
(U-eong) the phosphorylation of Lyn, Fyn and Syk, which are
involved in the FcεRI signaling pathway.
Zanthoxylum Zanthoxylum coreanum Nakai inhibited both the
coreanum Nakai IgE-antigen complex or PMA/A23187-induced β-
30618741
(Wang-cho-pi hexosaminidase release and IL-4 production dose-
-na-mu) dependently in RBL-2H3 mast cells.
P. santalinus extract inhibited β-hexosaminidase and
Pterocarpus
histamine release and reduced tumor necrosis
santalinus 30609435
factor-α, interleukin-4, and prostaglandin E2
(Ja-dan-hyang)
secretion.
Schizonepeta degranulation of RBL-2H3 cells was assessed by
tenuifolia measuring the release of β-hexosaminidase, which 29849717
(Hyeong-gae) was suppressed by ST at 10 μg/mL.

PMIDa; PubMed identifier (PMID) is a unique number assigned to each PubMed record.

38
39
5. Purpose of this study

T. vulgaris is well known as one of the aromatic herbs that have been used worldwide

for the purpose of spices, cosmetics, and treatment due to its various effect. Antioxidant,

anti photoaging, and skin protection effects of T. vulgaris were reported, but no studies have

been reported on the inhibitory effect of T. vulgaris in atopic dermatitis models. Thus, this

study was conducted to investigate whether T. vulgaris is effective in improving atopic

dermatitis through the regulation of inflammatory response and histamine release.

In AD, steroids and immunosuppressive drug have been used for treatment. There are

many reports that long term use of steroids can cause serious side effects such as skin

atrophy, steroid-induced acne, and telangiectasia. Immunosuppressive drug has also been

reported to have adverse reactions including temporary skin irritation. As the number of

patients with AD continues to increase around the world, it is necessary to develop

alternative treatments that are safe with few side effects. Therefore, this study was conducted

to demonstrate the effect of T. vulgaris as a therapeutic agent of AD.

Anti-atopic and anti-allergic effects of T. vulgaris were studied in both in vitro and in

vivo AD models. In the in vitro study, the effects of T. vulgaris on mast cell degranulation

and expression of calcium related protein were investigated in DNP-IgE/BSA-stimulated

RBL-2H3 cells. Furthermore, the production of pro-inflammatory cytokines and

40
chemokines, as well as their signaling pathway including MAPKs, STAT1, and NF-κB, were

examined in TNF-α/IFN-γ-stimulated HaCaT cells. In the in vivo study, the effects of

T. vulgaris by topical application were examined following DFE treatment in NC/Nga mice

model. The clinical dermatitis severity scores, skin hydration, TEWL, scratching behavior,

serum IgE levels, histamine release, histological changes, and the expression of AD-related

protein markers were evaluated.

41
Ⅱ. Materials and Methods

1. Materials

The dried leaves of T. vulgaris was purchased from Mountain Rose Herbs Company

(Eugene, OR, USA) with USDA certificate. Dulbecco’s modified Eagle’s medium (DMEM),

Fetal bovine serum (FBS), antibiotics, and trypsin-EDTA were purchased from Gibco-BRL

(Grand Island, NY, USA). Enzyme-linked immunosorbent assay (ELISA) kits for

TARC/CCL17 and MDC/CCL22 were purchased from R&D systems (Human TARC/CCL17

Duoset ELISA kit, Human MDC/CCL22 Duoset ELISA kit; R&D systems, Inc.,

Minneapolis, MN, USA). Mouse IgE and histamine ELISA Kit were purchased from Abcam

(Cambridge, UK). Primary and secondary antibodies were obtained from Santa Cruz (Santa

Cruz, CA, USA), Cell signaling (Danvers, MA, USA), and Abcam (Cambridge, UK).

Rosmarinic acid, caffeic acid, lipopolysacaride (LPS), recombinant human TNF-α and IFN-

γ, Monoclonal Anti-dinitrophenyl antibody produced in mouse (Anti-DNP IgE), 3-(4,5-

Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and other reagents were

purchased from Sigma-Aldrich (St. Louis, MO, USA).

42
2. Sample preparation

Dried T. vulgaris (1.5 g) was extracted with ethanol (50%) in a ration of 1:10 (w/v) using

a digital orbital shaker, SHO-1D (Daihan, Korea) for 24 h at 24-25 ℃ three times. After 24 h

incubation, the extracts were filtered and evaporated under vacuum at 40 ℃; and yielded

17.8 % (w/v).

43
Figure 6. Preparation of T. vulgaris extract

44
3. HPLC analysis

The HPLC system (UltiMate 3000 LC system, Thermo Fisher and Voorhees Scientific

Inc., Sunnyvale, CA, USA) with a C18 column (4.6 x 250 mm, 5 μm; Hypersil GOLD,

Thermo Fisher Scientific Inc., Sunnyvale, CA, USA) was used to detect the polyphenolic

compounds of T. vulgaris. Other setting conditions were running samples in 50% methanol

at 1 mg/mL, a 10 μL sample was injected at a flow rate of 1 mL/min, and a detection

wavelength at 280 nm. The rosmarinic acid and caffeic acid standards were prepared in 50%

methanol with a range of concentrations (3.125 μg/mL to 100 μg/mL).

45
4. In vitro study

4.1. Cell culture

The murine macrophage RAW 264.7 cell line and rat basophilic leukemia RBL-2H3 cell

line were obtained from KCLB (Korea) and Human immortal keratinocyte HaCaT cell line

was obtained from ATCC (VA, USA). All cell lines were maintained in the incubator at 37℃

with humidified atmosphere containing with 5% of CO 2. Cells were cultivated in DMEM

media containing 10% heat-inactivated FBS and 1% antibiotics and antimycotic medication

using T-75 flask (Raw 264.7 cells) or 100 mm dishes (HaCaT cells and RBL-2H3 cells).

Then cells were seeded in 96 well (Raw 264.7, 1.0×10 5 cells/well; RBL-2H3,

1.0×104 cells/well) and 6 well (HaCaT, 1.0×10 6 cells/well; RBL-2H3, 5.0×10 5 cells/well) and

until the confluence reached to 80%.

4.2. Sample treatment

The cells were treated with various concentration (ranging from 1 to 100 µg/mL) of

T. vulgaris following sensitization. For Raw 264.7 cells, the cells were seeded at 96 well

then co-treated with T. vulgaris and stimulator after 24 h incubation. 1 µg/mL of LPS was

used as a stimulator and 1 µM of dexamethasone was used to a positive control. For HaCaT

cells, the cells were seeded at 6 well plate and were pretreated with

46
T. vulgaris for 30 min, and then stimulated with 10 ng/mL of TNF-α and IFN-γ for 30 min. 1

and 10 µg/mL of tacrolimus was used to a positive control. To analyze the section of

cytokines and chemokines, the cells were incubated 24 h after treatment. For RBL-2H3

cells, the cells were seeded at 6 well plate and were pretreated with

T. vulgaris for 30 min, and then stimulated with 100 μg/mL of compound48/80 for 30 min.

100 µM of cromolyn was used to a positive control.

4.3. Measurement of cell viability

MTT is a colorimetric assay used to measure cell viability. Specifically, viable cells

allow produce purple-colored formazan thereby it can be detected by spectrophotometer. At

the end of the incubation period, MTT reagent was added to adjust its concentration to 0.1

mg/mL. Next, the MTT treated cells were incubated for 3 h at 37°C in a

CO2 incubator, after which the medium was removed and 800 μL of DMSO was added to

dissolve the formazan crystals. Finally, the OD was read at 595nm using an ELISA reader

(Molecular Devices FilterMax F5; San Francisco. CA, USA).

4.4. Measurement of NO production

NO production of RAW 264.7 cells was measured using the Griess reagent system

(Promega, Fitchburg, WI, USA). The Cells were seeded in 96 well plate

(1.0×105 cells/well) and then incubated for 24h. Next, TV or LPS was treated and the

47
incubation was continued for an additional 24 h. 50 μL of a sulfanilamide solution and an 50

μL of N-(1-naphthyl)ethylenediamine dihydrochloride solution was then added to each well

and incubated for 10 min at room temperature in each process. Absorbance was recorded at

595 nm using a microplate reader.

4.5. β-hexosaminidase release assay

To investigate the suppressive effect of T. vulgaris on degranulation, a sign of type 1

immediate allergic response, secretion of β-hexosaminidase was analyzed. Following

suspension in 10% FBS-supplemented DMEM, RBL-2H3 cells were seeded in 96-well

plates at a density of 3×10 4 cells/well and incubated for 24 h. After washing twice with

siraganian buffer, cells were incubated with anti-DNP-IgE (50 ng/mL) for 24 h. IgE-

sensitized cells were treated various concentrations of T. vulgaris for 1 h at 37 ̊C. Then,

DNP-BSA was added to each well to reach a concentration of 100 ng/mL, and cells were

incubated for an additional 4 h under the same conditions. Afterwards, cells were placed in

an ice-bath for 10 min to terminate incubation. Supernatants were collected and 50 μL of

each sample was mixed with of substrate buffer (1 mM p-nitrophenyl-N-acetyl-β-D-

glucosamine in 0.1 M citrate buffer, pH 4.5). Following 1 h of incubation, reaction was

stopped with 200 μL/well stop solution (0.1 M Na 2CO3/NaHCO3, pH 10.0). Absorbance was

read at 405 nm using a microplate reader (Molecular Devices Co. Ltd., Sunnyvale, CA,

48
USA).

4.6. Histamine release assay

The release of histamine was quantified with commercial ELISA kit (Histamine ELISA

kit; Abcam, Cambridge, UK). After 2 h of sample treatment and stimulation, 1 mL of cell

culture supernatants were collected then the total concentration of histamine was measured

according to the manufacturer’s instruction.

4.7. Measurement of intracellular calcium

Fluo-3/AM (Invitrogen), a fluorescent indicator, was used to measure intracellular

calcium levels. RBL-2H3 cells were pre-incubated with Fluo-3/AM for 1 h, treated with

sample for 1 h, and then stimulated with DNP-HSA (100 ng/ml). The fluorescence intensity

was detected at an excitation wavelength of 485 nm and an emission wavelength of 535 nm

as previously described. Intracellular calcium levels were compared to those of non-

stimulated control cells, which were set at a value of one relative fluorescent unit.

4.8. Enzyme-linked immunosorbent assay (ELISA)

The Secretion of cytokines and chemokines were quantified with commercial ELISA kits

(Human CCL17/TARC Duoset ELISA Kit, Human CCL22/MDC Duoset ELISA Kit; R&D

systems, Inc., MN, USA). After 24 h of sample treatment and stimulation, 1 mL of cell

culture supernatants were collected then the total concentration of TARC/CCL17 and

49
MDC/CCL22 was measured according to the manufacturer’s instruction. The release of

histamine was quantified with commercial ELISA kit (Histamine ELISA kit; Abcam,

Cambridge, UK). After 30 min of sample treatment and stimulation, 1 mL of cell culture

supernatants were collected then the total concentration of histamine was measured

according to the manufacturer’s instruction.

4.9. Reverse transcription-polymerase chain reaction (RT-PCR)

Total cellular RNA was extracted from HaCaT cells using TRIzol (Invitrogen Co, Grnad

island, NT, US). RNA samples were quantified and a total of 4 μg of RNA was reverse

transcribed using 200 units of reverse-transcriptase and 0.5 μg/μL oligo-(dT)15 dimer

(Bioneer CO., Korea). The amplification was performed using a PCR premix (Bioneer CO.,

Korea). Primers for TARC/CCL17, MDC/CCL22, RANTES/CCL5, IL-6, and IL-8 were

described in Table 6. PCR was performed using a Veriti Thermal Cycler (Applied

Biosystems, Foster City, CA, USA, Table 6). PCR products were separated by 2.0% agarose

gel electrophoresis and visualized with ethidium bromide.

50
Table 6. List of primers for RT-PCR

Gene Sequence (5’-3’)

Forward 5΄- ACCACAGTCCATGCCATCAC -3΄,


GAPDH
Reverse 5΄-CCACCACCCTGTTGCTGTAC -3΄.
Forward 5΄ - CTCCTTCTCCACAAGCGCC -3΄
IL-6
Reverse 5΄ - GCCGAAGAGCCCTCAGGC -3΄
Forward 5΄ - TCAGTGCATAAAGACATACTCC -3΄
IL-8/CXCL8
Reverse 5΄ - TGGCATCTTCACTGATTCTTG -3΄
Forward 5΄ - ATGGCCCCACTGAAGATGCT -3΄
TARC/CCL17
Reverse 5΄ - TGAACACCAACGGTGGAGGT -3΄
Forward 5΄ - CCCCGTGCCGAGCACATCAAGGAGTATTT-3΄
RANTES/CCL5
Reverse 5΄ - CGTCCAGCCTGGGGAAGGTTTTTGTA -3΄
Forward 5΄ - AGGACAGAGCATGGCTCGCCTACAGA -3΄
MDC/CCL22
Reverse 5΄ - AATGGCAGGGAGGTAGGGCTCCTGA -3΄

51
4.10. Western blot analysis

The cells and skin tissues were extracted using RIPA buffer and the nucleoprotein and

cytoplasmic protein were separated using commercial kit (NE-PER nuclear and cytoplasmic

extraction reagents; Pierce). Next, cell and skin lysates were homogenized to yield

equivalent amounts of protein based on protein concentration measurements performed with

Bradford reagent (Bio-Rad, Hercules, CA, USA). Homogenized proteins were

electrophoresed by 10 to 15% sodium dodecyl sulfate-polyacrylamide gel (SDS-PAGE) and

thereafter transferred from SDS-PAGE to a nitrocellulose membrane (Amersham Pharmacia

Biotech, Buckinghamshire, UK). Non-specific binding was blocked with 5% Bovine serum

albumin in TBST (50 mmol / 1 Tris-HCL, pH 7.5, 150 mmol / 1 NaCl, and 0.1% Tween 20)

for 1 h at room temperature followed by overnight incubation of membranes with primary

antibodies at 4°C. The membranes were then washed with TBST three times and incubated

with secondary antibody (Santa Cruz Biotechnology Inc., CA, USA) for 1 h at room

temperature. Finally, proteins were visualized using a chemiluminescence detection ECL

reagent (GE Healthcare Life science, PA, USA) and quantified with UVI-1D software,

(UVITEC, Warwickshire, UK).

52
5. In vivo study

5.1. Experimental animals

Six-week-old male NC/Nga mice (21-26 g) were obtained from Central Lab Animals,

Inc. (Seoul, Korea). The animals were randomly divided into five groups of five mice per

cage and housed in conditions of 22 ± 1°C, 60 ± 5% humidity, and 12-h light/dark cycles.

Animals were acclimatized for 2 week before starting the study. The experimental protocol

[KHUASP(SE)-17-014] was approved by the Institutional Animal Care and Use Committee

of Kyung Hee University.

5.2. Induction of AD-like skin lesion and topical application

Eight week-old male NC/Nga mice were exposed with Biostir-AD (Biostir, Kobe,

Japan), a hydrophilic petrolatum-based ointment-containing extract of house dust mite

(Dermatophagoides farinae), according to the manufacturer’s instructions. After removing

mice hair, 150 μL of 4% (w/v) sodium dodecyl sulfate (SDS) was applied to that dorsal

skin. After drying, DFE (100 mg/mouse/time) was applied two times per week for 3 weeks.

Twenty five mice were divided into five groups of five mice per cage: (a) Normal (distilled

water only); (b) Control (DFE + distilled water); (c) Tacrolimus 0.1% (DFE + tacrolimus

0.1% in distilled water); (d) T. vulgaris 0.1% (DFE + T. vulgaris 0.1% in distilled water);

and (e) T. vulgaris 1% (DFE + T. vulgaris 1% in distilled water). Samples were applied three

53
times per week for 3 weeks. In this experiment, tacrolimus 0.1% was used as a positive

control.

5.3. Evaluation of AD-like skin symptoms

The relative severity of AD was evaluated macroscopically according to the following

five symptoms: erythema, edema, erosion, dryness, and lichenification. The total dermatitis

severity score was defined as the sum of component scores (0, no symptoms; 1, mild; 2,

moderate; 3, severe). The range of dermatitis score is 0 to 15. The dermatitis scoring was

performed by a blind test during the experimental period.

54
Table 7. Symptoms of AD

Score 0 Score 1 Score 2 Score 3


Intensity
None Mild Moderate Severe

Erythema

Edema

Erosion

Dryness

Lichenification

55
5.4. Measurement of physiological skin alteration

During the experimental period, physiological alteration of dorsal skin of experimental

animals were measured using skin measurement instrument (Dermalab ® combo, Cortex

Technology, Denmark). Skin parameters including video scope, high resolution ultrasound,

hydration, TEWL, and erythema were measured using appropriate probe then all data was

analyzed by Dermalab skinlab software (Cortex Technology, Denmark).

5.4.1. Video scope

The videoscope probe is able to magnify and visualize the surface of the skin using

polarized or non-polarized white light as the light source.

5.4.2. High resolution ultrasound

Ultrasound skin imaging is based on measuring the acoustic response from the skin,

when an acoustic pulse is sent into the skin. The energy of the acoustic pulse is very low and

will not affect the skin or other tissue in any way. When the emitted acoustic pulse hits the

different structures of the skin, part of the pulse will be reflected and part of the pulse will

be transmitted further into the skin. The reflected signal will travel back and be picked up by

the ultrasound transducer. After processing, the cross-sectional image as visualized on the

screen represents an intensity (ampli-tude) analysis of these returned signals.

The intensity of the received signal refers to a color scale, where dark colors represent

56
areas with low reflection (i.e. none or small changes in density between the structures in the

skin) and bright colors represent areas with strong reflections (i.e. significant changes in

density between structures).

5.4.3. Hydration

The hydration probe provides information about the hydration state by measuring the

conducting properties of the very upper layers of the skin, when subjected to an alternating

voltage. Accordingly, the method is referred to as a conductance measurement and the

output is presented in the unit of micro-Siemens (μS).

5.4.4. TEWL

Water loss as measured by the SkinLab Combo is based on Nilsson’s Vapor Pressure

Gradient method, an open chamber method with minimal impact on the skin being examined

and, accordingly, very low bias to the reading.

Two sets of temperature/humidity sensors are mounted in a measurement chamber at

different heights above the skin surface. The measurement chamber is open to allow the skin

to “breathe” freely, and the evaporation rate follows Fick’s Law of Diffusion:

Rate = P x (c1 - c2) / T

where P = permeability coefficient of membrane, (c1 - c2) = concentration gradient, T =

thick-ness of membrane.

57
To obtain comparable and reproducible results when measuring transepidermal water

loss stand-ardized measurement procedures are strongly recommended. Guidelines have

been published by The Standardization Group of the European Society of Contact

Dermatitis.

5.4.5. Erythema

The measurement of erythema is based on an active color detecting chip. Illumination is

provided by two high intensity white LED´s. Erythema is the redness index of the skin. This

measures is used for the estimation of the level of redness (hemoglobin) in the skin. The

higher the value the more redness in the skin.

5.5. Evaluation of scratching behavior

To evaluate the effect of T. vulgaris on the scratching behavior, it was measured once a

week for 3 weeks . Each mouse in all groups was videotaped for 15 min with digital camera

placed on the top of the cages. The one scratching bout was defined as a series of scratching

movement by the hind paw.

5.6. Measurement of serum IgE level

After the experimental period, blood samples were obtained from mice then were

separated by centrifugation at 14,000 × g for 20 min at 4 ℃. The supernatants were collected

thereafter the total serum IgE were measured using a mouse IgE enzyme-linked

58
immunosorbent assay kit (BD Bioscience, CA, USA) according to manufacturer’s

instruction.

5.7. Histopathological analysis

After sacrifice, the back skin of each mouse was fixed in 4% paraformaldehyde for 24 h.

Next, the dorsal skin specimens were embedded in paraffin and sectioned at 5 μm.

Hematoxylin and eosin (H&E) staining was used to observe epidermal thickness. Toluidine

blue (TB) staining was used to measure the degree of mast cell infiltration. After H&E and

TB staining, all stained skin specimens were observed by light microscope.

59
6. Statistical analysis

The results were determined using the Statistical Analysis System (GraphPad Prism 5).

All experiments were carried out in triplicate. The values were expressed as means ±

standard deviation (SD). Statistical comparison between different treatments was determined

using one-way analysis of variance (ANOVA) followed by Duncan’s test. For statistical

analysis, Student’s test was used to compare individual treatments to the controls. The level

of statistical significance was set as follows: # and * p < 0.05, ## and ** p < 0.01, and ###

and *** p < 0.001.

60
Ⅲ. Results

1. The identification of active components from T. vulgaris

The active components of T. vulgaris were detected by HPLC analysis. As shown in the

chromatogram, caffeic acid was confirmed as the active component at 17.5 min by 0.4%

(Figure 7A). Also, rosmarinic acid was found as the active component at 42.8 min by 5.8%

(Figure 7B).

61
(A)

62
(B)

Figure 7. HPLC analysis of T. vulgaris and caffeic acid and rosmarinic acid standard.
As an active component of T. vulgaris, (A) caffeic acid and (B) rosmarinic acid were
detected.

63
Table 8. Active components of T. vulgaris

T. vulgaris

Name Caffeic acid Name Rosmarinic acid

Formula C 9H8O 4 Formula C18H16O8

Molecular weight 180.16 Molecular weight 360.32

Content 0.4% Content 5.8%

64
2. The Effects of T. vulgaris on atopic dermatitis and histamine related allergic

response: An in vitro study

2.1. Effects of T. vulgaris on atopic dermatitis related inflammation in LPS-stimulated

Raw 264.7 cells

2.1.1. Cell viability

To investigate the effect of T. vulgaris on cell viability in Raw 264.7 cells, MTT assay

was conducted. As shown in Figure 8A, cell viability was reduced in LPS-stimulated

Raw264.7 cells compared with non-stimulated cells. The cells treated with

T. vulgaris showed no cytotoxicity.

65
2.1.2. NO production

Excessive NO production was observed following LPS-stimulation. Compared to non-

stimulated condition, LPS treatment triggered NO production by 552.6% (Figure 8B).

However, at a concentration of 100 µg/mL, T. vulgaris treatment significantly attenuated the

LPS-induced overproduction of NO by 57.1% of that in only LPS-stimulated

cells.

66
(A)

(B)

Figure 8. Effects of T. vulgaris on cell viability and NO production in LPS-stimulated


Raw 264.7 cells.
(A) Cell viability was measured by MTT assay. (B) Nitric oxide production was determined
by NO assay. Dexamethasone was used as a positive control. All data are shown as the mean
± standard deviation of at least three independent experiments. # and * indicate significant
differences between non-stimulated control and LPS-stimulated control, respectively.
###p < 0.001, versus non-stimulated control. **p < 0.01 and ***p < 0.001, versus LPS-

67
stimulated control.
2.2. Effects of T. vulgaris on histamine related allergic response in DNP-IgE/BSA-

stimulated RBL-2H3 cells

2.2.1. Cell viability

To determine cytotoxic influences of T. vulgaris in DNP-IgE/BSA-stimulated RBL-2H3

cells, MTT assay was performed. After sensitization with DNP-IgE, cells were pretreated

with ketotifen fumarate salt and T. vulgaris, and then stimulated with DNP-BSA. Non-

stimulated cells were regarded as 100% viability. Compared with non-stimulated cells,

DNP-IgE/BSA-stimulated cells showed a slight decrease in viability. Both ketotifen

fumarate salt and T. vulgaris treatment induced no noticeable cytotoxicity compared to non-

stimulated conditions (Figure 9A).

68
2.2.2. β-hexosaminidase release

To evaluate the effect of T. vulgaris on mast cell degranulation, β-hexosaminidase assay

was conducted in DNP-IgE/BSA-stimulated RBL-2H3 cells. β-hexosaminidase release

increased up to 787.0% in DNP-IgE/BSA-stimulated cells compared to non-stimulated cells.

However, it was reduced by treatment with ketotifen fumarate salt and

T. vulgaris. T. vulgaris significantly inhibited β-hexosaminidase release in a dose-dependent

manner at 50, 100, and 250 μg/mL by 24.5%, 36.5%, and 52.7%, respectively (Figure 9B).

69
2.2.3. Histamine release

The release of histamine, which plays an important role in allergic reactions, was

measured in DNP-IgE/BSA-stimulated RBL-2H3 cells. Histamine release increased up to

533.5% in DNP-IgE/BSA-stimulated cells compared to non-stimulated cells. In contrast, it

was suppressed in ketotifen fumarate salt and T. vulgaris treated cells. T. vulgaris markedly

decreased the release of histamine in a dose-dependent manner at 100 and 250 μg/mL by

32.5% and 38.0%, respectively (Figure 9C). These results showed that

T. vulgaris has anti-allergic effect by inhibiting mast cell degranulation.

70
(A)

(B)

(C)

Figure 9. Effects of T. vulgaris on cell viability, β-hexosaminidase, and histamine


release in DNP-IgE/BSA-stimulated RBL-2H3 cells.
(A) Cell viability, (B) β-hexosaminidase release, (C) Histamine release were determined.
Ketotifen fumarate and tacrolimus were used as a positive control. All data are shown as the
mean ± standard deviation of at least three independent experiments.
# and * indicate significant differences between non-stimulated control and DNP-IgE/BSA-
stimulated control, respectively. ###p < 0.001, versus non-stimulated control.
*p < 0.05, **p < 0.01, and ***p < 0.001, versus DNP-IgE/BSA-stimulated control.

71
72
2.2.4. Intracellular calcium levels

In mast cell degranulation, the change of intracellular calcium levels occur. The

increased calcium levels are essential in response to mast cell stimulation, which induces

granule-plasma membrane fusion, resulting in degranulation of mast cells. It leads to the

release of mediators such as β-hexosaminidase and histamine. Therefore, the inhibition of

intracellular calcium levels is important to suppress release of histamine. To determine the

mechanism by which T. vulgaris suppressed mast cell degranulation, intracellular calcium

levels were measured. The inhibitory effects of T. vulgaris on intracellular calcium release

were analyzed using the fluorescent indicator Fluo-3/AM. The intracellular calcium levels

were elevated by DNP-IgE/BSA-stimulation. However, treatment with T. vulgaris mitigated

the elevation of intracellular calcium levels at a concentration of 50, 100, and 250 μg/mL

(Figure 10).

73
Figure 10. Effects of T. vulgaris on intracellular calcium levels in DNP-IgE/BSA-
stimulated RBL-2H3 cells.
Intracellular calcium levels in DNP-IgE/BSA-stimulated RBL-2H3 cells. Ketotifen fumarate
and tacrolimus were used as a positive control. Intracellular calcium levels were compared to
those of untreated control cells, which were set at a value of one relative fluorescent unit. #
and * indicate significant differences between non-stimulated control and DNP-IgE/BSA-
stimulated control, respectively. ##p < 0.01 versus non-stimulated control. *p < 0.05 and
**p < 0.01, versus DNP-IgE/BSA-stimulated control.

74
2.2.5. Signaling pathways in histamine release

2.2.5.1. CRHR1 and CRHR2 expression

In mast cell degranulation, CRHR1 acts as a potentiator, while CRHR2 acts as an

inhibitor, resulting in histamine release. The effect of T. vulgaris on the expression of

CRHR1 and CRHR2 in DNP-IgE/BSA-stimulated RBL-2H3 cells was confirm by western

blot analysis. The stimulation of DNP-IgE/BSA augmented the expression of CRHR1, while

the expression of CRHR2 was suppressed in only DNP-IgE/BSA-stimulated cells. But, it

was altered by treatment with T. vulgaris. T. vulgaris decreased the expression of CRHR1 by

29.5% and induced the expression of CRHR2 up to 1246.5% at a concentration of 250

μg/mL, compared to only DNP-IgE/BSA-stimulated cells (Figure 11). These regulation

inhibits the mobilization of calcium, which in turn leads to a decrease in the release of

histamine.

75
(A)

(B)

Figure 11. Effects of T. vulgaris on expression of CRHR1 and CRHR2 in


DNP-IgE/BSA-stimulated RBL-2H3 cells.
(A) Protein expression of CRHR1 and CRHR2 in DNP-IgE/BSA-stimulated RBL-2H3 cells.
(B) Band intensities were quantified by densitometry, normalized to the level of β-actin and
calculated as a percentage of the basal response. All data are shown as the mean ± standard
deviation of at least three independent experiments. # and * indicate significant differences
between non-stimulated control and DNP-IgE/BSA-stimulated control, respectively.
#p < 0.05 and ##p < 0.01, versus non-stimulated control. *p < 0.05, **p < 0.01, and
***p < 0.001, versus DNP-IgE/BSA-stimulated control.

76
77
2.2.5.2. phosphorylation of PLCγ

Upon stimulation with DNP-IgE/BSA, PLCγ is activated through phosphorylation. The

phosphorylation of PLCγ generates IP3 which results in the release of intracellular calcium

from endoplasmic reticulum (ER). This causes mast cell degranulation, leading to an

increase in histamine release. To investigate the effect of T. vulgaris on phosphorylation of

PLCγ, RBL-2H3 cells were stimulated with DNP-IgE/BSA. The level of phosphorylated

PLCγ was up-regulated in cells that were only DNP-IgE/BSA-stimulated cells. However,

treatment with T. vulgaris inhibited the phosphorylation level of PLCγ by 81.2% at 50 μg/mL

(Figure 12). It was verified that the decrease in histamine release by

T. vulgaris is due to the reduction of calcium level through downregulation of PLCγ

phosphorylation.

78
(A)

(B)

Figure 12. Effects of T. vulgaris on phosphorylation of PLCγ in DNP-IgE/BSA-


stimulated RBL-2H3 cells.
(A) Protein expression of p-PLCγ and PLCγ in DNP-IgE/BSA-stimulated RBL-2H3 cells.
(B) Band intensities were quantified by densitometry, normalized to the level of β-actin and
calculated as a percentage of the basal response. All data are shown as the mean ± standard
deviation of at least three independent experiments. # and * indicate significant differences
between non-stimulated control and DNP-IgE/BSA-stimulated control, respectively.
###p < 0.001, versus non-stimulated control. *p < 0.05 and ***p < 0.001, versus
DNP-IgE/BSA-stimulated control.

79
2.2.5.3. phosphorylation of IP3R

After the phosphorylation of PLCγ, IP3 is generated and then binds to its receptor, IP3R,

existed in ER membrane. When IP3R is phosphorylated, calcium release from ER is

induced. Consequently, this causes the influx of extracellular calcium, which increases the

release of histamine through exocytosis of mast cells. To investigate the effect of

T. vulgaris on phosphorylation of IP3R, RBL-2H3 cells were stimulated with

DNP-IgE/BSA. As shown in Figure 13, the level of phosphorylated IP3R was up-regulated

in cells that were only DNP-IgE/BSA-stimulated cells. However, treatment with

T. vulgaris inhibited the phosphorylation level of IP3R by 98.3% at a concentration of 100

μg/mL. This results showed that T. vulgaris affects the suppression of calcium depletion in

ER.

80
(A)

(B)

Figure 13. Effects of T. vulgaris on phosphorylation of IP3R in DNP-IgE/BSA-


stimulated RBL-2H3 cells.
(A) Protein expression of p-PLCγ and PLCγ in DNP-IgE/BSA-stimulated RBL-2H3 cells.
(B) Band intensities were quantified by densitometry, normalized to the level of β-actin and
calculated as a percentage of the basal response. All data are shown as the mean ± standard
deviation of at least three independent experiments. # and * indicate significant differences
between non-stimulated control and DNP-IgE/BSA-stimulated control, respectively.
###p < 0.001, versus non-stimulated control. *p < 0.05 and ***p < 0.001, versus
DNP-IgE/BSA-stimulated control.

81
2.2.5.4. calcium channel protein expressions

The granulation of mast cell depends on the concentration of calcium released from ER.

After the release of calcium from ER, calcium channel proteins such as STIM1, Orai1, and

TRPC1 are activated. Due to this, the influx of extracellular calcium is elevated that

accelerates the release of histamine. Thus, the expression levels of STIM1, Orai1, and

TRPC1 were investigated. The stimulation with DNP-IgE/BSA increased the expression of

these proteins. In cells treated with T. vulgaris, the expressions of STIM1, Orai1, and

TRPC1 decreased by 20.8%, 69.0%, and 82.3% at 100 μg/mL, respectively (Figure 14).

These results indicated that T. vulgaris treatment blocks the extracellular calcium influx via

downregulation of calcium channel proteins, thereby reducing the release of histamine.

82
(A)

(B)

Figure 14. Effects of T. vulgaris on expression of calcium channel proteins in


DNP-IgE/BSA-stimulated RBL-2H3 cells.
(A) Protein expression of STIM1, Orai1, TRPC1 in DNP-IgE/BSA-stimulated RBL-2H3
cells. (B) Band intensities were quantified by densitometry, normalized to the level of β-
actin. All data are shown as the mean ± standard deviation of at least three independent
experiments. # and * indicate significant differences between non-stimulated control and
DNP-IgE/BSA-stimulated control, respectively. ##p < 0.01 and ###p < 0.001, versus non-
stimulated control. *p < 0.05, **p < 0.01, and ***p < 0.001, versus DNP-IgE/BSA-
stimulated control.

83
2.3. Effect of T. vulgaris on atopic dermatitis related inflammation in TNF-

α/IFN-γ-stimulated HaCaT cells

2.3.1. Cell viability

The cytotoxicity of T. vulgaris was confirmed by MTT assay in TNF-α/IFN-γ-stimulated

HaCaT cells. Cells were pretreated with T. vulgaris (10 and 100 μg/mL) and tacrolimus (1

and 10 μg/mL), and then stimulated with 10 ng/mL of TNF- α/IFN-γ. Non-stimulated cells

were regarded as 100% viability. Compared with non-stimulated cells, TNF-α/IFN-γ-

stimulated cells showed a slight decrease in viability. Both T. vulgaris and tacrolimus

treatment induced no noticeable cytotoxicity compared to only TNF-α/IFN-γ-stimulated

conditions (Figure 15A).

84
2.3.2. Chemokine production

The production of TARC/CCL17 and MDC/CCL22 was affected by T. vulgaris

treatment in TNF-α/IFN-γ-stimulated HaCaT cells. When cells were stimulated by

TNF-α/IFN-γ with no other pretreatment, the production of TARC/CCL17 and MDC/CCL22

was increased by 781.4% and 885.6%, respectively, compared to non-stimulated cells.

However, tacrolimus and T. vulgaris treated cell showed suppressed TARC/CCL17

production by 35.5% at 10 μg/mL of tacrolimus and by 89.0% at 100 μg/mL of

T. vulgaris, respectively, compared to those of only TNF-α/IFN-γ-stimulated cells. The

production of MDC/CCL22 also decreased by 41.5% at 1 μg/mL of tacrolimus and by 64.5%

at 100 μg/mL of T. vulgaris, respectively, compared with those of only TNF-α/IFN-γ-

stimulated cells. As a result, T. vulgaris treatment showed stronger inhibitory effect on

TARC/CCL17 and MDC/CCL22 production than tacrolimus treatment (Figure 15B and C).

85
(A)

(B) (C)

Figure 15. Effects of T. vulgaris on cell viability and chemokines production in


TNF-α/IFN-γ-stimulated HaCaT cells.
(A) Cell viability was measured using MTT assay. (B) TARC/CCL17 production, (C)
MDC/CCL22 production were determined by using ELISA kit. All data are shown as the
mean ± standard deviation of at least three independent experiments. # and * indicate
significant differences between non-stimulated control and TNF-α/IFN-γ-stimulated control,
respectively. ###p < 0.001, versus non-stimulated control. *p < 0.05, **p < 0.01, and
***p < 0.001, versus TNF-α/IFN-γ -stimulated control.

86
2.3.3. mRNA expression of pro-inflammatory cytokine and chemokines

As shown in Figure 16, pro-inflammatory cytokine (IL-6) and chemokines

(TARC/CCL17, MDC/CCL22, RANTES/CCL5, and IL-8/CXCL8) mRNA expressions were

increased after stimulation with TNF-α/IFN-γ. The mRNA expression levels of

IL-6, TARC/CCL17, MDC/CCL22, RANTES/CCL5, and IL-8/CXCL8 increased by

345.0%, 144.1%, 174.9%, 372.8%, and 201.2% in only TNF-α/IFN-γ-stimulated cells,

respectively, compared with the non-stimulated cells. Importantly,

T. vulgaris and tacrolimus markedly reduced those overexpression levels. Treatment with

T. vulgaris at 100 μg/mL decreased the mRNA expression levels of IL-6, TARC/CCL17,

MDC/CCL22, RANTES/CCL5, and IL-8/CXCL8 by 30.6%, 35.1%, 38.0%, 63.0%, and

30.9%, respectively, compared with only TNF-α/IFN-γ-stimulated cells.

87
(A)

(B)

Figure 16. Effects of T. vulgaris on mRNA expression of pro-inflammatory cytokines


and chemokines in TNF-α/IFN-γ-stimulated HaCaT cells.
(A) mRNA expression of IL-6, TARC, MDC, RANTES, and IL-8 in TNF-α/IFN-γ-
stimulated HaCaT cells. (B) An equimolar quantity of mRNA was quantified compared to
GAPDH. Tacrolimus was used as a positive control. All data are shown as the mean ±
standard deviation of at least three independent experiments. # and * indicate significant
differences between non-stimulated control and TNF-α/IFN-γ-stimulated control,
respectively. #p < 0.05, ##p < 0.01, and ###p < 0.001, versus non-stimulated control.

88
*p < 0.05, **p < 0.01, and ***p < 0.001, versus TNF-α/IFN-γ -stimulated control.

89
2.3.4. Signaling pathways in pro-inflammatory cytokine and chemokines

2.3.4.1. MAPKs activation

TNF-α/IFN-γ activated MAPK signaling pathways resulting in overexpression of pro-

inflammatory cytokines. The activation was examined by detecting the phosphorylation

level of MAPKs by western blot analysis. TNF-α/IFN-γ treatment triggered the

phosphorylation of p-38, JNK, and ERK by 125.5%, 171.4%, and 130.4%, respectively.

However, it was reversed by treatment of tacrolimus and T. vulgaris that they suppressed

this phosphorylation. T. vulgaris treatment at concentration of 100 μg/mL significantly

inhibited the phosphorylation of p-38 and JNK by 46.8% and 39.5%, respectively (Figure

17).

90
(A)

(B)

Figure 17. Effects of T. vulgaris on activation of MAPKs in TNF-α/IFN-γ-stimulated


HaCaT cells.
(A) Protein expression of p-p38, p38, p-JNK, JNK, p-ERK, and ERK in TNF-α/IFN-γ-
stimulated HaCaT cells. (B) Band intensities were quantified by densitometry, normalized to
the level of β-actin, and calculated as a percentage of the basal response. All data are shown
as the mean ± standard deviation of at least three independent experiments. # and * indicate
significant differences between non-stimulated control and TNF-α/IFN-γ-stimulated control,
respectively. #p < 0.05 and ##p < 0.01, versus non-stimulated control. *p < 0.05, **p <

91
0.01, and ***p < 0.001, versus TNF-α/IFN-γ -stimulated control.
2.3.4.2. NFκB and STAT1 activation

Inflammators increase the expression of IκB kinase (IKK) complex with three subunits

(IKKα, IKKβ, and IKKγ) leads to phosphorylation and degradation of the IκBα. The

phosphorylated IκBα binds to NF-κB. This complex moves into nucleus and activates

inflammatory genes. At the same time, STAT1 responses to stimuli and can lead to

overexpression of inflammatory and immune response genes.

As predicted, TNF-α/IFN-γ treatment triggered IKKα/β, IκB, NF-κB, and STAT1

phosphorylation as shown in Figure 18, 19. The phosphorylation of IKKα/β, IκB, NF-κB,

and STAT1 was overexpressed by 272.9%, 131.3%, 164.7%, and 129.0%, respectively, in

only TNF-α/IFN-γ stimulated cells, compared with non-stimulated cells. However, it was

reversed by treatment of tacrolimus and T. vulgaris. At a concentration of 100 μg/mL,

T. vulgaris significantly decreased the phosphorylation levels of IKKα/β, IκB, NF-κB, and

STAT1 by 39.0%, 71.3%, 39.3%, and 72.7%, respectively, compared with only TNF-α/IFN-

γ stimulated cells. Therefore, the results suggest that T. vulgaris can diminish the production

of inflammatory cytokines and chemokines via regulation these signaling pathways.

92
(A)

(B)

Figure 18. Effects of T. vulgaris on NFκB signaling pathway in TNF-α/IFN-γ-stimulated


HaCaT cells.
(A) Protein expression of p-IKKα/β, IKKα/β, p-IκBα, IκBα, p-NFκB, and NFκB in
TNF-α/IFN-γ-stimulated HaCaT cells. (B) Band intensities were quantified by densitometry,
normalized to the level of β-actin. All data are shown as the mean ± standard deviation. #
and * indicate significant differences between non-stimulated control and TNF-α/IFN-γ-
stimulated control, respectively. #p < 0.05 and ##p < 0.01, versus non-stimulated control.

93
*p < 0.05, **p < 0.01, and ***p < 0.001, versus TNF-α/IFN-γ -stimulated control.
(A)

(B)

Figure 19. Effects of T. vulgaris on STAT1 signaling pathway in TNF-α/IFN-γ-


stimulated HaCaT cells.
(A) Protein expression of p-STAT1 and STAT1 in TNF-α/IFN-γ-stimulated HaCaT cells.
(B) Band intensities were quantified by densitometry, normalized to the level of β-actin and
calculated as a percentage of the basal response. All data are shown as the mean ± standard
deviation of at least three independent experiments. # and * indicate significant differences
between non-stimulated control and TNF-α/IFN-γ-stimulated control, respectively. #p <
0.05, versus non-stimulated control. *p < 0.05 and **p < 0.01, versus TNF-α/IFN-γ -
stimulated control.

94
95
3. The Effects of T. vulgaris on atopic dermatitis and histamine related allergic

response: An in vivo study

3.1. Morphological characteristics

3.1.1. Body weight

AD skin lesions were treated with tacrolimus and T. vulgaris. Compared to normal

group, no noticeable changes in the body weight were observed in the other group. In

addition, no abnormal symptoms were recorded in all groups, indicating that the tested

samples ensure safety, without toxicity or adverse effects (Figure 20).

96
Figure 20. Effects of T. vulgaris on body weight in DFE-induced NC/Nga mice.
NC/Nga mice exposed to DFE and treated with tacrolimus and T. vulgaris during 3 weeks.
Body weight was measured during 3 weeks.

97
3.1.2. Spleen weight

AD induces a variety of responses in the immune system leading to increase the weight

of immune organs such as spleen. As expected, the weight of spleen was significantly

increased by 202.7% than that of the normal group. However, it was reduced by topical

application of tacrolimus and T. vulgaris. The average weight of tacrolimus-applied mice’s

spleen was decreased by 34.4% compared to that of the DFE-applied control group. In the

groups treated with T. vulgaris at 0.1% and 1%, the weight of spleen was significantly

decreased by 22.5% and 27.7%, respectively, compared to the control group (Figure 21).

98
Figure 21. Effects of T. vulgaris on spleen weight in DFE-induced NC/Nga mice.
Spleen weight in AD-induced mice. Values shown are the mean ± standard deviation. # and
* indicate significant differences between normal group and control group, respectively.
###p < 0.001, versus normal group. **p < 0.01 and ***p < 0.001, versus control group.

99
3.1.3. Skin morphology

The severity of dermatitis was determined according to the sums of the scores for each

symptoms once a week for 3 weeks. In DFE-induced mice, skin symptoms such as

erythema, edema, erosion, dryness, and lichenification were observed. After 3 weeks, the

DFE topical applied mice have all typical characteristics of atopic dermatitis disease as listed

above with maximum dermatitis score 9.0 ± 1.7. Topical application of tacrolimus and

T. vulgaris significantly improved the AD-like skin symptoms (Figure 22A). Among three

groups tacrolimus 0.1%, T. vulgaris 0.1%, and T. vulgaris 1%, the T. vulgaris 0.1% group

was the most effective sample in decreasing dermatitis symptoms in AD-induced mice skin

(Figure 22B).

100
(A)

101
102
(B)

Figure 22. Effects of T. vulgaris on skin morphology in DFE-induced NC/Nga mice.


(A) Clinical picture of DFE-induced AD-like lesions. (B) Dermatitis score. (G). The number
of mast cells. Five groups: Normal group, Control group (DFE treatment), PC group (DFE
treatment + Tacrolimus 0.1%), T. vulgaris 0.1% group (DFE treatment + T. vulgaris 0.1%),
T. vulgaris 1% group (DFE treatment + T. vulgaris 1%). Values shown are the mean ±
standard deviation of at least three independent experiments. # and * indicate significant
differences between normal group and control group, respectively.
##p < 0.01 and ###p < 0.001, versus normal group. *p < 0.05, **p < 0.01, and ***p <
0.001, versus control group.

103
3.2. Physiological characteristics

3.2.1. Skin density

To investigate the effects of T. vulgaris on skin density in DFE-induced NC/Nga mice,

living mice were applied to Dermalab combo skin measurement instrument using ultra sonic

flow probe. The skin density is visualized as green and yellow-colored graphics using

Dermalab combo skinlab software. As shown in Figure 23, DFE-induced control group

exhibited impaired skin density compared to non-treated normal group. However, treatment

of T. vulgaris recovered the abnormal loss of skin density.

104
(A)

(B)

Figure 23. Effects of T. vulgaris on skin density in DFE-induced NC/Nga mice.


(A) The levels of collagen and dermal thickness were determined using Dermalab Combo
(Cortex Technology, DK). (B) Relative density was measured.). Five groups: Normal group,
Control group (DFE treatment), PC group (DFE treatment + Tacrolimus 0.1%),
T. vulgaris 0.1% group (DFE treatment + T. vulgaris 0.1%), T. vulgaris 1% group (DFE
treatment + T. vulgaris 1%). Values shown are the mean ± standard deviation of at least
three independent experiments. # and * indicate significant differences between normal
group and control group, respectively. ###p < 0.001, versus normal group. **p < 0.01,
versus control group.

105
106
3.2.2. Skin hydration

AD causes skin dryness by reducing strantum corneum (SC) hydration. At the end of 3

weeks, SC hydration was diminished by 78.3% in the control group than that of the normal

group. These physiological changes were improved by topical application of tacrolimus and

T. vulgaris. SC hydration was significantly enhanced by over 300% for all topical

application including tacrolimus, T. vulgaris 0.1%, and T. vulgaris 1% (Figure 24A).

107
3.2.3. TEWL

AD causes skin dryness by increasing TEWL. At the end of 3 weeks, TEWL was

elevated by 371.9%. These physiological changes were improved by topical application of

tacrolimus and T. vulgaris. The TEWL was decreased from 56.7%-64.1% by treatment of all

topical solution (Figure 24B).

108
3.2.4. EI

It is known that EI is increased in AD. At the end of 3 weeks, EI was increased up to

1510.8% in the control group. These increased EI was improved by topical application of

tacrolimus and T. vulgaris. EI was reduced by treatment of all topical solution (Figure 24C).

109
(A) (B)

(C)

Figure 24. Effects of T. vulgaris on skin hydration, TEWL, and EI in DFE-induced


NC/Nga mice.
(A) SC hydration, (B) TEWL, and (C) EI were determined using Dermalab Combo (Cortex
Technology, DK). Five groups: Normal group, Control group (DFE treatment), PC group
(DFE treatment + Tacrolimus 0.1%), T. vulgaris 0.1% group (DFE treatment + T. vulgaris
0.1%), T. vulgaris 1% group (DFE treatment + T. vulgaris 1%). Values shown are the mean
± standard deviation. # and * indicate significant differences between normal group and
control group, respectively. ###p < 0.001, versus normal group. ***p < 0.001, versus

110
control group.

3.3. Itching related characteristics

3.3.1. Scratching behavior

During the experimental period, the number of scratches was assessed to verify

inhibitory effect of T. vulgaris on itching associated with AD. In the control group, topical

application of DFE led to a significant increase in the number of scratches 32 times higher

than that of the normal group. After topical application of tacrolimus and

T. vulgaris for 3 weeks, the number of scratches was remarkably reduced by 8.9 times for

tacrolimus group, by 6.5 times for the T. vulgaris 0.1% group, and by 3.4 times for the

T. vulgaris 1% group than that of the control group (Figure 25).

111
Figure 25. Effects of T. vulgaris on scratching behavior in DFE-induced NC/Nga mice.
Scratching behavior in DFE-induced NC/Nga mice. Five groups: Normal group, Control
group (DFE treatment), PC group (DFE treatment + Tacrolimus 0.1%),
T. vulgaris 0.1% group (DFE treatment + T. vulgaris 0.1%), T. vulgaris 1% group (DFE
treatment + T. vulgaris 1%). Values shown are the mean ± standard deviation. # and *
indicate significant differences between normal group and control group, respectively.
##p < 0.01 and ###p < 0.001, versus normal group. *p < 0.05, **p < 0.01, and ***p <
0.001, versus control group.

112
3.3.2. Serum IgE level

In the control group, serum IgE level was increased by 3832.9% compared with the

normal group. This serum IgE level was significantly reduced in the groups treated with

tacrolimus and T.vulgaris. Both T.vulgaris 0.1% and T.vulgaris 1% groups inhibited IgE by

75.6% and 91.4%, respectively, compared with the control group. These inhibitory effects of

T.vulgaris were more effective than that of the tacrolimus 0.1% group (Figure 26).

113
Figure 26. Effects of T. vulgaris on serum IgE level in DFE-induced NC/Nga mice.
Serum IgE level in DFE-induced NC/Nga mice. Five groups: Normal group, Control group
(DFE treatment), PC group (DFE treatment + Tacrolimus 0.1%), T. vulgaris 0.1% group
(DFE treatment + T. vulgaris 0.1%), T. vulgaris 1% group (DFE treatment + T. vulgaris
1%). Values shown are the mean ± standard deviation. # and * indicate significant
differences between normal group and control group, respectively.
###p < 0.001, versus normal group. **p < 0.01 and ***p < 0.001, versus control group.

114
3.3.3. β-hexosaminidase and histamine release

In the control group, serum β-hexosaminidase level was increased up to 134.9%

compared with the normal group. This serum β-hexosaminidase level was significantly

reduced in the groups treated with tacrolimus and T.vulgaris. Both T.vulgaris 0.1% and

T.vulgaris 1% groups inhibited β-hexosaminidase by 15.3% and 21.4%, respectively,

compared with the control group (Figure 27A).

In the control group, serum histamine level was increased up to 274.1% compared with

the normal group. In tacrolimus and T.vulgaris treated group, serum histamine levels were

markedly diminished. Both T.vulgaris 0.1% and T.vulgaris 1% groups inhibited histamine

release by 72.4% and 53.7%, respectively, compared with the control group (Figure 27B).

115
(A)

(B)

Figure 27. Effects of T. vulgaris on β-hexosaminidase and histamine release in DFE-


induced NC/Nga mice.
β-hexosaminidase and histamine release in DFE-induced NC/Nga mice. Values shown are
the mean ± standard deviation. # and * indicate significant differences between normal group
and control group, respectively. ###p < 0.001, versus normal group. *p < 0.05 and ***p <
0.001, versus control group.

116
117
3.4. Proteins expression

3.4.1. CRHR1 and CRHR2 expression

The effect of T. vulgaris on expression of CRHR1 and CRHR2 in DFE- induced NC/Nga

mice was confirm by western blot analysis. The expression of CRHR1 was increased by DFE

treatment, whereas the expression of CRHR2 was suppressed in only DFE-induced NC/Nga

mice. It was reversed by treatment with T. vulgaris. Compared to only DFE-induced NC/Nga

mice, treatment with 1% T. vulgaris reduced the expression of CRHR1 by 54.9% and

induced the expression of CRHR2 up to 249.3% (Figure 28).

118
(A)

(B)

Figure 28. Effects of T. vulgaris on expression of CRHR1 and CRHR2 in DFE-induced


NC/Nga mice.
(A) Protein expression of CRHR1 and CRHR2 in AD-induced mice skin. (B) Band
intensities were quantified by densitometry, normalized to the level of β-actin and calculated
as a percentage of the basal response. Values shown are the mean ± standard deviation. # and
* indicate significant differences between normal group and control group, respectively.
##p < 0.01 and ###p < 0.001, versus normal group. * p < 0.05, **p < 0.01, and ***p <
0.001, versus control group.

119
3.4.2. MAPKs activation

The expression levels of p-p38 and p-ERK were elevated in the control group by 269.0%

and 140.7%, respectively, than those of the normal group. Topical application of tacrolimus

and T. vulgaris induced downregulation of p-p38 and p-ERK expression. In the

T. vulgaris 1% group, the expression levels of p-p38 and p-ERK were reduced by 52.6% and

29.9%, respectively, than those of the control group (Figure 29).

120
(A)

(B)

Figure 29. Effects of T. vulgaris on activation of MAPKs in DFE-induced NC/Nga mice.


(A) Protein expression of MAPKs in AD-induced mice skin. (B) Band intensities were
quantified by densitometry, normalized to the level of β-actin and calculated as a percentage
of the basal response. Values shown are the mean ± standard deviation. # and * indicate
significant differences between normal group and control group, respectively.
##p < 0.01, versus normal group. *p < 0.05, **p < 0.01, and ***p < 0.001, versus control
group.

121
122
3.4.3. IκBα and NFκB activation

The expression levels of NF-κB and p-IκBα were elevated in the control group by 116.9%

and 1065.4%, respectively, than those of the normal group. On the other hand, the expression

of IκBα was significantly decreased by 60.9%. Topical application of tacrolimus and

T. vulgaris induced downregulation of p-IκBα expression and upregulation of IκBα

expression. In the T. vulgaris 1% group, the expression levels of p-IκBα were reduced by

77.7% and the expression level of IκBα was enhanced by 176.3% than those of the control

group (Figure 30).

123
(A)

(B) (C)

Figure 30. Effects of T. vulgaris on IκBα and NFκB in DFE-induced NC/Nga mice.
(A) Protein expression of IκBα and NFκB in AD-induced mice skin. (B) Band intensities
were quantified by densitometry, normalized to the level of β-actin and calculated as a
percentage of the basal response. Values shown are the mean ± standard deviation. # and *
indicate significant differences between normal group and control group, respectively.
#p < 0.05 and ###p < 0.001, versus normal group. * p < 0.05 and **p < 0.01, versus control
group.

124
3.4.4. Filaggrin expression

Filaggrin plays an important role in skin barrier function. In AD, it is characterized by a

decrease in filaggrin expression. Therefore, in this study, the effect of

T. vulgaris on the expression of filaggrin was investigated. Compared with the normal

group, the expression of filaggrin was diminished by 44.9% in the control group. Topical

application of tacrolimus and T. vulgaris upregulated filaggrin expression by 93.1-133.0%

compared with the control group (Figure 31).

125
(A)

(B)

Figure 31. Effects of T. vulgaris on filaggrin expression in DFE-induced NC/Nga mice.


(A) Protein expression of filaggrin in AD-induced mice skin. (B) Band intensities were
quantified by densitometry, normalized to the level of β-actin and calculated as a percentage
of the basal response. Values shown are the mean ± standard deviation. # and * indicate
significant differences between normal group and control group, respectively.
###p < 0.001, versus normal group. ***p < 0.001, versus control group.

126
3.5. Histological analysis

3.5.1. Epidermal thickness

The epidermal thickness in the control group was significantly increased by 712.7%

compared with the normal group. Treatment with tacrolimus, T. vulgaris 0.1%, and 1%

decreased the epidermal thickness by 44.5%, 70.0%, and 59.1%, respectively, compared with

that of the control group. Moreover, the epidermal thickness in the

T. vulgaris 0.1% group was thinner than that of the tacrolimus 0.1% group and superficial as

that of the normal group (Figure 32).

127
(A)

(B)

Figure 32. Effects of T. vulgaris on epidermal thickness in DFE-induced NC/Nga mice.


The dorsal skin’s histopathological features revealed by HE staining. Values shown are the
mean ± standard deviation. # and * indicate significant differences between normal group
and control group, respectively. ###p < 0.001, versus normal group. **p < 0.01 and

128
***p < 0.001, versus control group.

129
3.5.2. Accumulation of mast cells

Skin-infiltrating inflammatory cells, containing mast cells, eosinophils, Langerhans

cells, and CD4+ lymphocytes expressing skin colonization antigen, play a crucial role in the

initiation and exacerbation of inflammation in AD. To investigate the effect of

T. vulgaris on accumulation of mast cells, TB staining was performed. While the number of

mast cells in the control group increased compared to the normal group, this was reduced by

treatment with tacrolimus, T. vulgaris 0.1%, and 1% (Figure 33).

130
(A)

(B)

Figure 33. Effects of T. vulgaris on accumulation of mast cells in DFE-induced NC/Nga


mice.
The the number of mast cells revealed by TB staining. Values shown are the mean ±
standard deviation. # and * indicate significant differences between normal group and
control group, respectively. ##p < 0.01, versus normal group. *p < 0.05 and **p < 0.01,
versus control group.

131
Ⅳ. Discussion

AD is a chronic inflammatory skin disease that persists without complete recovery or

repeatedly recurs even in both childhood and adulthood. AD pathogenic mechanism can be

related with the exposure to environmental or chemical agents or abnormal genetic and

immunologic processes. However, the exact causes and treatment of AD have not fully

understood yet. For decades, many patients with AD have been using topical corticosteroids

or topical calcineurin inhibitors, but they are reported to have side effects. Long-term use of

topical corticosteroids can trigger skin atrophy, acne, and telangiectasia. Tacrolimus, a

calcineurin inhibitor, can cause side effects such as burning sensation and nephrotoxicity.

Thus, the development of natural product derived treatments that have few side effects to

substitute them is drawing attention.

T. vulgaris, commonly known as thyme, is an aromatic evergreen herb used as cosmetic,

spice and medicine. Several studies showed that T. vulgaris has a variety of beneficial

effects including antioxidant, antimicrobial, and anti-inflammatory activities. This

biological effects are known to be influenced by various components such as rosmarinic

acid, caffeic acid, and thymol. Although anti-inflammatory effects of

T. vulgaris are well known, the effects on AD have not been reported. Thus, this study was

132
conducted to reveal the anti-atopic effect of T. vulgaris in AD-induced in vitro and in vivo

models.

Macrophages are associated with immune response and control NO and pro-

inflammatory cytokines, which are regarded as pro-inflammatory mediators. Upon LPS

stimulation, excessive NO production was shown in Raw 264.7 cells. However, compared

with only LPS-stimulated cells, it was reversed by treatment of T. vulgaris at a concentration

of 100 μg/mL (Figure 8).

In allergic reactions, mast cells are considered as the main effector cells. They express

FcεRI, the high-affinity IgE receptor, on the surface and stimulated by allergen. When mast

cells are activated by cross-linking of membrane-bound IgE with FcεRI, the granules

present in them are degranulated, releasing vast amounts of stored mediators including β-

hexosaminidase, histamine, and proteases. Therefore, β-hexosaminidase and histamine are

recognized as markers for mast cell degranulation. These mediators are associated with the

regulation of inflammatory reactions in other immune-related cells including keratinocytes

and lymphocytes. In DNP-IgE/BSA-stimulated RBL-2H3 cells, the inhibitory effect of

T. vulgaris on mast cell degranulation was examined. It reduced mast cell degranulation by

suppressing the release of β-hexosaminidase and histamine (Figure 9).

133
The intracellular calcium levels play an important role in mast cell degranulation.

Ca2+ is a vital factor in degranulation of mast cells, which controls the fusion of preformed

secretory granules and plasma membranes and affects the release of mediators. The

activation of mast cells mediated by IgE results in an increase in intracellular calcium

levels. Under stimulation, PLCγ, major signaling enzyme, is activated by phosphorylation

which generates IP3 and diacylglycerol. IP3 bins to its receptor in ER that causes the

intracellular calcium release from ER stores. And then, the reduced calcium concentration in

ER is sensed by STIM1. It promotes the influx of calcium from extracellular space through

calcium channel proteins such as Orai1 and TRPC1. Consequently, the concentration of

cytosolic calcium is elevated, which leads to the fusion of preformed granules and plasma

membrane. It triggers the release of mediators such as β-hexosaminidase and histamine by

mast cell degranulation. In this study, it was verified that T. vulgaris significantly reduced

the intracellular calcium levels in a dose-dependent manner, compared with only

DNP-IgE/BSA-stimulated cells (Figure 10). This is due to the decrease in the

phosphorylation of PLCγ and IP3R by T. vulgaris treatment (Figure 12 and Figure 13).

Also, the lower expression of calcium channel proteins such as STIM1, Orai1, and TRPC1

was observed in T. vulgaris treated cells (Figure 14). These results suggest that

134
T. vulgaris suppresses exocytosis of mast cell granules by regulating the signaling pathway

associated with calcium, and consequently reduces the release of histamine.

135
Figure 34. Inhibitory effects of T. vulgaris on mast cells activation

136
In the present study, TNF-α/ IFN-γ mixture was used to stimulate keratinocytes. The

keratinocyte activation is a hallmark of the development of AD in acute and chronic phases.

AD is a skin disease in which Th2 immune response is activated. It is known that Th2

immune response is closely related to the increased expression of chemokines such as

TARC/CCL17 and MDC/CCL22. Th2 type cytokines are recruited by the secretion of these

chemokines. In patients with AD, serum levels of TARC/CCL17 and MDC/CCL22 are

elevated which are correlate with severity of AD. In the early stage of AD, increased

eosinophil and cytotoxic granules are found to associate with RANTES which is over

secreted by TNF-α and IFN-γ -induced keratinocytes. This suggests that these chemokines

can be used as markers to exhibit the severity of the disease. In the present study, data

showed that treatment of T. vulgaris remarkably reduced the production of TARC/CCL17

and MDC/CCL22 compared with those of only TNF-α/IFN-γ-stimulated HaCaT cells

(Figure 15). Moreover, it was verified that mRNA expression levels of inflammation-related

cytokine and chemokines, including IL-6, TARC/CCL17, MDC/CCL22, RANTES/CCL5,

and IL-8/CXCL8 were downregulated by treatment of T. vulgaris (Figure 16).

In AD processes, transcription factor NF-κB plays very important roles due to their

direct target genes coding for many pro-inflammatory cytokines. In AD-induced

inflammatory responses, due to activating the IκB kinase, the IκBs are degraded resulting in

137
the releasing of NF-κB from their inactive form in cytoplasm. NF-κB then moves to the

nucleus and binds affinity to target genes. In 2008, Sun et al. found that blockade of ERK

and p38 MAPK inhibited the nuclear translocation of phosphorylated NF-κB p65. Similar to

NF-κB, STAT1 is phosphorylated and moved into the nucleus under stimulation. It

facilitates gene transcription of factors such as MDC. Therefore, the activation of NF-κB and

STAT1 is critically involved in the regulation of inflammation. As shown in Figure 17,

T. vulgaris downregulated the phosphorylation of MAPKs induced by TNF-α/IFN-γ

stimulation. In addition, it was showed that the phosphorylation levels of IKKα/β, IκB, NF-

κB were suppressed by treatment of T. vulgaris (Figure 18). With regard to STAT1,

T. vulgaris reduced the phosphorylation of STAT1 (Figure 19). Thus, it was thought that the

reduction of pro-inflammatory cytokines and chemokines is due to regulation of these

signaling pathways. Based on our in vitro data, T. vulgaris can be considered as an effective

NF-κB inhibitor with inhibition of MAPKs activation, the phosphorylation of IκBα and

STAT1. Our findings suggest that T. vulgaris treatment has a significant influence on the

improvement of AD in in vitro model.

138
Figure 35. Inhibitory effects of T. vulgaris on keratinocytes activation

139
Based on the in vitro data, further in vivo studies were performed to determine the effect

of T. vulgaris on AD symptoms in DFE-induced NC/Nga mice. In this study, DFE, a major

species of house dust mite, was used to induce AD. Previous studies have shown that

repeated application of DFE causes severe AD skin lesions by elevating chemokine levels.

As expected, DFE induced the typical AD symptoms including scratching, thick skin,

erythema, dryness, lichenification, and rash in NC/Nga mice. However, it was alleviated by

topical application of T. vulgaris. In the 0.1% and 1% T. vulgaris group, AD symptoms

including erythema, edema, erosion, dryness, and lichenification were significantly improved

(Figure 22). Also, both impaired skin density and increased EI were recovered by

T. vulgaris treatment (Figure 23 and Figure 24C). In histological analysis, the epidermal

thickness in the T. vulgaris 0.1% and 1% group was reduced compared with that of the

control group (Figure 32). This is thought to be due to the regulation of protein expressions

associated with inflammatory responses such as MAPKs, IκBα, and NFκB (Figure 29 and

Figure 30).

Itching leads to the disruption of skin barrier by causing scratching which is one of the

main symptoms in AD. It tends to become a vicious cycle by itching-scratching cycle,

causing severe lesions. It results in unbearable pain for patients with AD. Furthermore,

itching converts a Th2-predominant acute phase to a Th1-dominant chronic phase, which is

140
main characteristic of AD symptoms. Mast cell activation and mast cell-derived mediators

play a crucial role in pathophysiology of itching. The scratching behavior is associated with

many mediators including neuropeptides, histamines, proteinases, and cytokines. In

particular, histamine released from mast cells is known to have a significant contribution to

the induction of itching in AD. Also, infiltration of mast cells is observed in the epidermis

and dermis of AD skin lesions. In this study, it was demonstrated that topical application of

T. vulgaris remarkably diminished the number of scratches via inhibition of serum IgE, β-

hexosaminidase, and histamine compared with the control group (Figure 26 and Figure 27).

TB staining of the dorsal skin of mice showed lower accumulation of mast cells in the

groups treated with 0.1% and 1% T. vulgaris at two concentration, compared to the control

group (Figure 33). These results suggest that T. vulgaris suppresses allergic inflammation

via inhibition of mast cell degranulation and infiltration.

AD is involved with impaired skin barrier which affects the reduction of skin hydration

and acidification. Dry and base skin is regarded as the primary characteristics in AD. The

more skin hydration and acidification is badly changed, the more symptoms of AD are

worse. One way to protect the skin barrier from AD is moisturizing. In patient with AD, the

skin TEWL increases and hydration decreases. In a previous study, appropriate moisturizing

improves the discomfort associated with dryness, repairs the skin barrier, and reduces the

141
quantity and potency of pharmacological intervention. Studies shown that impaired skin

barrier can be recovered by promotion of filaggrin expression. Filaggrin, key proteins of the

epidermal keratinocytes, has a crucial structural and functional role in the epidermis and

homeostasis of the skin. In the present data, SC hydration levels were elevated and TEWL

levels were decreased in the groups treated with T. vulgaris 0.1% and 1% (Figure 24A and

24B). It is thought that the stimulation of filaggrin expression by topical application of

T. vulgaris resulting in these effect (Figure 31).

As the active components of T. vulgaris, caffeic acid and rosmarinic acid were identified

(Figure 7). There are several studies on the pharmacological effects of these components

related to skin inflammation. Zhang et al. reported that caffeic acid has anti-inflammatory

activities in both acute and chronic contact dermatitis models through reduction of cutaneous

TNF-α, IL-6, and IL-8 levels. In addition, oral administration of caffeic acid significantly

inhibited scratching behavior. Regarding the effects of rosmarinic acid on the skin, it was

demonstrated that rosmarinic acid attenuates AD by reducing IFN-γ and IL-4 production and

total serum IgE levels in 2,4-dinitrofluorobenzene-treated NC/Nga mice. These previous

studies indicate that the anti-atopic effect of T. vulgaris may be due to these components.

Therefore, it is considered that it would be worthwhile to confirm the synergistic effect

through combination of caffeic acid and rosmarinic acid on AD-induced models.

142
The patient with AD experience extreme psychological stress due to the vicious cycle of

scratching-itching. It lowers the patient’s quality of life. Psychological stress is recognized

as a risk factor in immune-related diseases because it has a significant impact on immune

function. According to previous study, both CRHR1 and CRHR2 are critical regulator of

mast cell degranulation and stress-related disorders. CRHR1 is a positive modulator of

stress-induced mast cell degranulation. On the contrary, CRHR2 is a negative modulator of

mast cell degranulation in the acute response to immunological and psychological stress. In

this study, it was verified that T. vulgaris suppresses the expression of CRHR1 while

elevating the expression of CRHR2 in DNP-IgE/BSA-stimulated RBL-2H3 cells (Figure

11). This inhibited mast cell degranulation, resulting in reduced histamine release which is

responsible for itching. These findings provide a clue to the effect of

T. vulgaris on relieving psychological stress in patient with AD. Therefore, further studies

are needed to confirm the anti-stress effect of T. vulgaris, and it is considered that it can be

applied to various fields.

In the present study, ketotifen, known as mast cell stabilizer, and tacrolimus, known as a

calcineurin inhibitor, were used as a positive control in in vitro assay. Ketotifen is widely

applied in the treatment of allergic diseases including urticaria, asthma, and allergic

conjunctivitis. It not only exhibits anti-allergic effect by antagonizing H1R, but also has the

143
property of stabilizing the mast cells. Baba et al. reported that ketotifen inhibited the process

of exocytosis. It results in the reduction histamine release from mast cells. Tacrolimus has

immunomodulatory and anti-inflammatory effect. Tacrolimus binds to its receptor and

inhibits the action of calcium/calmodulin-dependent protein phosphatase, calcineurin. This

blocks the dephosphorylation of NFAT and consequently suppresses the transcription of

inflammatory cytokines. As shown in Figure 9B and 9C, ketotifen reduced the release of β-

hexosaminidase and histamine at a concentration of 100 μM. Also, it was showed that

decreased release of β-hexosaminidase and histamine in cells treated with 10 ng/mL (12.17

nM) of tacrolimus. These effects were contributed by regulation of calcium related signaling

pathway (Figure 10-14). The ability of tacrolimus to inhibit calcineurin is well known, but

there are few studies on the suppression effect of mast cell activation in

in vitro model. Therefore, the results of this study are considered to be valuable. Compared

to ketotifen, tacrolimus indicated its effect at about 8,000 times lower concentration.

Although tacrolimus has this advantages, it is very expensive. These single chemical-based

therapeutic agents have side effects and cost problems, which can be burdensome to

patients. Thus, this study is thought to be meaningful because it has confirmed that

T. vulgaris can substitute these therapeutic agents.

144
Overall, this study demonstrated the mitigating effect of T. vulgaris in AD-induced in

vitro and in vivo models. These results suggest that T. vulgaris is valuable as an ingredient

for alleviating AD symptoms. Traditionally, thyme oil has been used a lot, but several

studies reported that it is irritating to the skin. In order to overcome this limitation, 50%

ethanol extract was used in this study, and safety was proved through

in vitro and in vivo results. Since T. vulgaris is a natural-derived product, it can be a new

therapeutic agent with fewer side effects compared to steroids and immunosuppressive drug

which have been frequently used as a treatment for AD. Therefore, it will have a

considerable value in cosmetic and pharmaceutical industries.

145
Ⅴ. Conclusion

The present study was conducted to investigate the anti-atopic effects of

T. vulgaris on AD-induced in vitro and in vivo models.

The results could be concluded as follows:

First, T. vulgaris treatment significantly inhibited β-hexosaminidase and histamine

release by controlling the calcium related signaling pathway in DNP-IgE/BSA-stimulated

RBL-2H3 cells.

Second, T. vulgaris remarkably suppressed the production of pro-inflammatory cytokine

and chemokines via regulation of MAPKs, STAT1, and NFκB pathway in TNF-α/IFN-γ-

stimulated HaCaT cells.

Third, topical application of T. vulgaris improved physiological, histopathological

alterations, and scratching behavior in DFE-treated NC/Nga mice. It increased SC

hydration, filaggrin expression and reduced TEWL, EI, scratching behavior, total serum IgE

level, and histamine release.

Based on the present results, it was demonstrated that T. vulgaris is effective in

ameliorating AD symptoms. Therefore, it can be a new candidate for alleviating AD

symptoms in cosmetic and pharmaceutical industries.

146
Ⅵ. References

1. M. Sokovi1, J. Glamoclija, A. 2iri1, and D. Kataranovski. Antifungal Activity of the


Essential oil of Thymus vulgaris L. and Thymol on Experimentally Induced
Dermatomycoses. Drug Development and Industrial Pharmacy. 2008; 34:1388-
1393.

147
Abstracts in Korean

국문초록

히스타민 분비 조절을 통한 타임(Thymus vulgaris L.)의


아토피 피부염 증상 개선 효과

서슬아

경희대학교 대학원

생명공학원

지도교수 이 태 후

일반적으로 백리향으로 알려진 타임은 아로마 허브 중 하나로 전 세계적으로 사용되고

있다. 타임은 항산화, 항염 및 면역조절 활성이 있는 것으로 보고되었으며, 특히 피부와

관련하여 광노화 억제 및 피부 보호 효과에 대해 보고되었다. 그러나 타임의 아토피 피부염

억제 효과에 대해서는 과학적으로 검증된 바 없다. 따라서 본 연구는 타임의 아토피 피부염

증상 개선 효과를 검증하기 위하여 수행되었다.

타임의 활성 성분은 HPLC 분석법에 의해 확인되었다. In vitro 연구에서는 ELISA

kits, RT-PCR 및 western blot법을 통하여 LPS로 자극된 Raw264.7 세포주, DNP-IgE/BSA

로 자극된 RBL-2H3 세포주 및 TNF-α/IFN-γ로 자극된 HaCaT 세포주에서 타임의 아토피

피부염 억제 효과가 확인되었다. In vivo 연구에서는 집먼지 진드기로 아토피 피부염이

유도된 NC/Nga 동물에서 형태학적 및 생리학적 변화, 긁는 행동, AD 증상과 관련된

단백질 발현 및 조직학적 변화를 측정하여 타임의 아토피 피부염 개선 효과를 확인하였

다.

148
In vitro 실험 결과, 타임이 LPS로 자극된 Raw264.7 세포주에서 NO 생성량을

유의적으로 억제시켰다. DNP-IgE/BSA로 자극된 RBL-2H3 세포주에서는 타임이 칼슘

관련 경로의 조절을 통하여 베타헥소사미니다제 및 히스타민 분비를 감소시키는 것을

확인하였다. TNF-α/IFN-γ로 자극된 HaCaT 세포주에서 타임에 의해 TARC 및 MDC의

생성이 현저하게 억제되었다. 게다가, 타임이 IL-6, TARC, MDC, RANTES 및 IL-8과 같은

염증성 사이토카인 및 케모카인의 유전자 발현의 감소에 영향을 미치는 것을

확인하였으며 이는 MAPKs, STAT1 및 NF-κB 경로의 조절에 의한 것으로 나타났다. In

vivo 결과에 따르면, 타임의 국소 도포에 의해 피부염의 중증도, 경피수분손실량, 홍반량,

긁는 행동, 히스타민 분비, 표피 두께 및 비만세포 침윤이 감소된 반면 피부 수분도 및

filaggrin의 발현은 증가되어 아토피 피부염 증상이 개선되는 것을 확인하였다.

종합적으로 본 연구는 아토피 피부염이 유도된 in vitro 및 in vivo 모델에서 타임의

아토피 피부염 개선 효과를 검증하였다. 이러한 결과는 타임이 히스타민 분비를 조절하여

가려움증을 완화시킴으로써 아토피 피부염 증상 개선에 효과가 있음을 나타내므로 화장품

및 의약품 산업에서 천연물 유래의 소재로서 활용될 수 있음을 시사한다.

149

You might also like