88
Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung des akademischen Grades des Doktors der Naturwissenschaften an der Universität Konstanz (Fachbereich Biologie) vorgelegt von Marina I. Borisova Tag der mündlichen Prüfung: 09.07.2010 Referent: Prof. Dr. Dr. Thomas Hartung Referent: PD. Dr. Florian Gantner

Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

  • Upload
    others

  • View
    18

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

Role of lipopolysaccharide in pulmonary

inflammation and fibrosis – an in vitro study

Dissertation

zur Erlangung des akademischen Grades

des Doktors der Naturwissenschaften

an der Universität Konstanz (Fachbereich Biologie)

vorgelegt von

Marina I. Borisova

Tag der mündlichen Prüfung: 09.07.2010

Referent: Prof. Dr. Dr. Thomas Hartung

Referent: PD. Dr. Florian Gantner

Page 2: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

2

For Mom with all my love

На мама с любов

Page 3: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

3

ACKNOWLEDGEMENT

The work presented in this PhD thesis was carried out between August 2006 and

June 2010 at the Chair of Biochemical Pharmacology at the University of Konstanz

under the supervision of Prof. Dr. Dr. Thomas Hartung and PD Dr. Sonja von Aulock.

First of all, I want to thank Prof. Dr. Dr. Thomas Hartung for his valuable scientific

advice, inspiring discussions during my PhD work and for correcting my manuscripts.

I sincerely appreciate having such a warm and caring teacher. I wish you all the best

in your future career and private life.

I especially want to thank my supervisor PD Dr. Sonja von Aulock for her

unbelievable patience, outstanding support and stimulating ideas even in the time of

being a Mom. I am also grateful for the excellent working facilities and financial

support, for creating a friendly atmosphere and for reading my manuscripts.

Many thanks also go to Dr. Thomas Meergans for his never ending enthusiasm and

interest in my work, for his excellent experimental support and for his friendship.

I am indebted to all “Pulmo-net” members for the incredible time and for organization

of valuable seminars and courses and to the EU Framework 6 for financial support in

this work.

I thank Prof. Dr. Albrecht Wendel for welcoming me into his group and for the

unforgettable party time together.

Sincere thanks go to all current and former colleagues - Dr. Anna Góra, Dr. Christian

Draing, Dr. Christoph Rockel, PD Dr. Corinna Hermann, Dr. Mardas Daneshian, Dr.

Sebastian Bunk, Sonja Erath, Dr. Stefanie Sigel, Dr. Tobias Speicher, Dr. Oliver

Dehus and especially my wonderful students Jenny, Rebecca and Eva for sharing

good and bad times, for creating an excellent working atmosphere and for the

unforgettable time together inside and outside the lab.

Page 4: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

4

I also thank the team of current and former technicians Teodora Baris, Annette Haas,

Tamara Rupp, Margarete Kreuer-Ullmann and Anne Gnerlich for their technical

assistance.

I would like to thank my Mom and my sister for their never ending love and mental

support during the time I was far away from home.

Last but not at least I thank Christoph for his love and for creating a harmonic and

stress-free atmosphere in the last stage of my PhD work. I am so lucky to have you in

my life!

Page 5: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

5

TABLE OF CONTENTS

Abbreviations.................................................................................................. 8

I. General introduction ............................................................................... 11

1. INFLAMMATORY LUNG DISEASES ............................................................. 11

1.1. Physiology of the lung.................................................................................. 11

1.2. Inflammatory lung diseases ......................................................................... 12

1.3. Etiology of IPF ............................................................................................. 12

1.4. Treatment of IPF.......................................................................................... 14

1.4.1. Pharmacotherapy ..................................................................................... 14

1.4.2. Non-pharmacological treatment................................................................ 15

1.5. Animal models of IPF................................................................................... 16

2. MECHANISMS OF INFLAMMATION ............................................................. 17

2.1. Innate immunity ........................................................................................... 17

2.2. Lung innate immunity................................................................................... 18

2.3. Toll-like receptors and other pathogen recognition receptors ...................... 18

2.4. Pathogen associated molecular patterns..................................................... 20

2.4.1. Lipopolysaccharide ................................................................................... 20

2.4.2. Peptidoglycan ........................................................................................... 21

2.4.3. Staphyloccocal enterotoxin B.................................................................... 22

2.5. Toll-like receptor signaling ........................................................................... 22

2.6. Inflammatory cytokines ................................................................................ 23

2.6.1. Tumor necrosis factor-α............................................................................ 23

2.6.2. Interleukin-1 family of cytokines................................................................ 23

2.6.3. Interleukin-6 .............................................................................................. 25

2.6.4. Interleukin-8 .............................................................................................. 25

2.6.5. Transforming growth factor-β.................................................................... 25

3. IN VITRO MODELS OF PULMONARY INFLAMMATION AND DISEASE ..... 26

3.1. In vitro models of pulmonary inflammation .................................................. 26

3.1.1. Epithelial cells ........................................................................................... 27

3.1.2. Macrophages............................................................................................ 28

3.1.3. Co-culture models..................................................................................... 29

3.2. In vitro models of fibrosis ............................................................................. 29

3.2.1. Epithelial to mesenchymal transition......................................................... 29

Page 6: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

6

3.2.2. Initiators of EMT in vitro ............................................................................ 30

4. AIMS OF THE STUDY.................................................................................... 32

II. Lung epithelial cells constitutively produce an

immunomodulatory factor for cytokine release by mononuclear

cells.................................................................................................33

1. ABSTRACT..................................................................................................... 34

2. INTRODUCTION ............................................................................................ 35

3. RESULTS ....................................................................................................... 37

3.1. A549 cells modulate inflammatory response of PBMC upon LPS, SEB and

PGN stimulation.................................................................................................. 37

3.2. Modulation of cytokine secretion by epithelial cells does not require direct

cell-cell contact ................................................................................................... 38

3.3. αIL-1β, but not αTNF-α neutralizing antibody, inhibits epithelial cell-mediated

amplification of IL-6 and IL-8 release in co-culture ............................................. 38

3.4. LPS-induced inhibition of TNF-α in mixed co-culture is regulated on post-

transcriptional level ............................................................................................. 39

3.5. Conditioned supernatant from resting A549 cells induces down-regulation of

TNF-α protein and mRNA in LPS-stimulated PBMC........................................... 40

3.6. The TNF-α inhibitory compound appears to be a peptide............................ 43

4. DISCUSSION ................................................................................................. 44

III. Conditioned supernatant from lipopolysaccharide stimulated

blood mononuclear cells induces epithelial to mesenchymal

transition in A549 cells: role of IL-1β and TNF-α .............................. 48

1. ABSTRACT..................................................................................................... 49

2. INTRODUCTION ............................................................................................ 50

3. RESULTS ....................................................................................................... 52

3.1. TGF-β1 induces EMT in A549 cells ............................................................. 52

3.2. A549 cells stimulated with LPS do not undergo EMT .................................. 53

3.3. Conditioned supernatant from LPS-stimulated PBMC induces EMT in A549

cells .................................................................................................................... 54

3.4. LPS-induced EMT is not driven by TGF-β1 ................................................. 55

3.5. IL-1β and TNF-α induce EMT in A549 cells ................................................. 56

Page 7: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

7

3.6. LPS-stimulated PBMC produce IL-1β and TNF-α, which induce EMT in lung

epithelial cells ..................................................................................................... 58

4. DISCUSSION ................................................................................................. 60

5. MATERIAL AND METHODS .......................................................................... 63

5.1. Reagents and antibodies ............................................................................. 63

5.2. Cell line........................................................................................................ 63

5.3. Isolation of human peripheral blood mononuclear cells ............................... 63

5.4. Enzyme-linked immunosorbent assay ......................................................... 64

5.5. A549-PBMC co-cultures .............................................................................. 64

5.6. Neutralization of IL-1β and TNF-α activity.................................................... 65

5.7. Inhibition of TNF-α processing by Brefeldin A.............................................. 65

5.8. LPS stimulation of PBMC in medium or A549 conditioned supernatant ...... 66

5.9. Total RNA extraction and cDNA synthesis................................................... 66

5.10. Quantitative Real-Time PCR...................................................................... 66

5.11. Characterization of TNF-α anti-inflammatory compound ........................... 67

5.12. SDS-PAGE and Western blot .................................................................... 67

5.13. Statistics .................................................................................................... 68

IV. References............................................................................................... 69

Page 8: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

8

ABBREVIATIONS

AcP IL-1 receptor accessory protein

ALI acute lung injury

ANOVA analysis of variance

ARDS acute respiratory distress syndrome

BAL bronchoalveolar lavage

BCA bicinchronic acid

BSA bovine serum albumin

CD cluster of differentiation

COPD chronic obstructive pulmonary disease

DMEM Dulbecco’s Minimal Essential Medium

DNA deoxyribonucleic acid

ECL enhanced chemiluminescence

EGF epidermal growth factor

ELISA enzyme-linked immunosorbent assay

EMT epithelial to mesenchymal transition

FCS fetal calf serum

FGF fibroblast growth factor

GlcNAc N-acetyl-glucosamine

HGF hepatocyte growth factor

HRP horseradish peroxidise

HSA human serum albumin

HSP heat shock protein

hu human

ICE IL-1β converting enzyme

IgG immunoglobulin G

IL interleukin

IL-1ra interleukin-1 receptor antagonist

IPF idiopathic pulmonary fibrosis

IFN interferon

IRAK IL-1 receptor-associated kinase

IRF interferon regulatory factor

kDa kilo Dalton

Page 9: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

9

LPS lipopolysaccharide

LTA lipoteichoic acid

MCP macrophage chemotactic protein

MD-2 lymphocyte antigen 96

MIP macrophage inflammatory protein

MyD88 myeloid differentiation protein

min minute

mRNA messenger RNA

mu murine

MurNAc N-acetyl-muramic acid

NaCl sodium chloride

NFκB nuclear factor κB

NOD nucleotide-binding oligomerization domain

NSIP nonspecific interstitial pneumonia

PAGE polyacrylamide gel electrophoresis

PAMP pathogen-associated molecular pattern

PBMC peripheral blood mononuclear cells

PBS phosphate buffered saline

PCR polymerase chain reaction

PDGF platelet derived growth factor

PGN peptidoglycan

PRR pattern recognition receptor

RIPA radioimmunoprecipitation assay buffer

RPMI Roswell Park Memorial Institute medium

R receptor

RT room temperature

SDS sodium dodecyl sulphate

SEB staphylococcal enterotoxin B

SEM standard error of the mean

SP surfactant protein

SV40 simian virus 40

TBS-T Tris-buffered saline with Tween 20

TGF-β transforming growth factor beta

TIR toll-interleukin 1 receptor

Page 10: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

10

TLR toll-like receptor

TMB 3,3’,5,5’-tetramethylbenzidine

TNF tumor necrosis factor

TRAF TNF receptor-associated family of proteins

TRIF TIR-domain-containing adapter-inducing interferon-β

Tris tris(hydroxymethyl)aminomethane

Tween20 polyoxyethylensorbitan monolaurate

UIP usual interstitial pneumonia

Page 11: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

11

I. GENERAL INTRODUCTION

1. INFLAMMATORY LUNG DISEASES

1.1. Physiology of the lung

The main function of the respiratory system is gas exchange. The respiratory system

consists of two functionally and structurally distinct regions known as upper and lower

respiratory tract. The upper respiratory tract includes the nose, the nasal cavity,

larynx and trachea and serves to filter, warm and humidify the inhaled air.

Downstream the trachea divides into left and right main bronchi, which enter the lung

parenchymal tissue. Bifurcation of the trachea in 23 generations forms the conductive

zone (main bronchi, small bronchi, bronchioles, terminal bronchioles) and respiratory

zone (respiratory bronchioles, alveolar ducts and the alveoli) of the lower respiratory

tract. The alveoli, also called air sacs, are surrounded by many blood capillaries. This

arrangement ensures optimal gas diffusion on the thin air-blood interface composed

of alveolar epithelial cells and capillary endothelial cells (Ehrhardt et al., 2002).

The total alveolar epithelial surface of an average adult human lung is very large -

140 m2 (Gehr, Bachofen & Weibel, 1978), and is comprised of type I and type II

epithelial cells. Type I squamous epithelial cells, mainly involved in gas exchange,

represent 93% of the epithelial surface area and only 33% the alveolar epithelial cell

number. Alveolar cuboidal type II cells are much smaller and represent the remaining

7% of the surface area and 67% of the epithelial cell number (Crapo et al., 1982).

Type II cells are situated in the corners of the alveoli and their physiological functions

include to serve as progenitors of damaged type I cells (Bhaskaran et al., 2007) and

to produce and secrete pulmonary surfactant. Pulmonary surfactant is composed of

80-90% phospholipids and about 10% surfactant proteins (SP) A, B, C and D and

plays an important role in maintaining physiological conditions during breathing by

reducing the surface tension in the lungs (Fehrenbach, 2001), (Goldmann et al.,

2009), (Johnston et al.). Alveolar epithelial type II cells also participate in ion

transport across the alveolar epithelium (Berthiaume, Lesur & Dagenais, 1999).

The most abundant phagocytic cell population in the lung airspaces is presented by

the alveolar macrophages, also called dust cells. These cells, derived by

differentiation of blood monocytes, account for approximately 95% of the lung

leukocytes (Martin & Frevert, 2005) and have a density of 17 x 103 cells/µl alveolar

Page 12: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

12

fluid (Losa Garcia et al., 1999). The other 5% are distributed between lymphocytes,

neutrophils, dendritic cells and eosinophils.

1.2. Inflammatory lung diseases

Inflammation in general can be classified as acute or chronic. Acute lung

inflammation is initiated by resident alveolar macrophages and is characterized by

infiltration of blood monocytes and neutrophils into the lung tissue and in alveolar

spaces (Stockley, 1998), (Ulich et al., 1991). Acute inflammation goes through

phases of initiation, amplification and resolution coordinated by inflammatory

molecules called cytokines. During prolonged or chronic inflammation, resolution

cannot take place, progressive tissue destruction and development of pulmonary

illness occurs.

Inflammation is thought to play an important role in the pathogenesis of lung

diseases such as acute lung injury (ALI), acute respiratory distress syndrome

(ARDS), chronic obstructive pulmonary disease (COPD), and idiopathic pulmonary

fibrosis (IPF). IL-1β, TNF-α and TGF-β1 were shown as common inflammatory

mediators implicated in asthma, COPD and pulmonary fibrosis (Araya & Nishimura).

Anti-inflammatory treatment of patients with low doses of corticosteroids was found

effective for diseases like ALI, ARDS (Lamontagne et al., 2009), (Tang et al., 2009)

and asthma (Rodrigo, 2006), (Cates & Lasserson, 2009), but ineffective for COPD

(Barnes, 2005) and IPF (Mapel, Samet & Coultas, 1996), (Richeldi et al., 2003).

Interestingly, particularly in smokers, COPD and IPF were found to coexist (Gauldie

et al., 2006).

1.3. Etiology of IPF

IPF, also called cryptogenic fibrosing alveolitis, is a chronic inflammatory and fibrotic

lung disease with unknown etiology, characterized by progressive scar formation in

the lung parenchyma. It affects approximately 5 million people worldwide with slightly

greater prevalence in men then in women. IPF is the most lethal interstitial lung

disease with no current cure and a median survival of 5 years from the time of

diagnosis. Different factors have been proposed as etiological factors – infections,

Page 13: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

13

cigarette smoking, autoimmune disorders, chemotherapy, allergens and toxins (Kelly

et al., 2003), (Meltzer & Noble, 2008), (Taskar & Coultas, 2006).

The classical pathology of IPF, usual interstitial pneumonia (UIP), does not

demonstrate influx of inflammatory cells and is insensitive to conventional anti-

inflammatory treatment. In addition to UIP pathology, nonspecific interstitial

pneumonia (NSIP) is also diagnosed clinically as IPF. NSIP has more pathologic

inflammation than UIP. However, patients with IPF have been shown to have periods

of acute exacerbation characterized by inflammatory cell infiltration seen in

bronchoalveolar lavage samples and lung biopsies, and numerous inflammatory

mediators were also found in patients with active IPF (Kalluri, 2009), (Maher, Wells &

Laurent, 2007), (Meltzer & Noble, 2008), (Strieter & Mehrad, 2009), (Wilson & Wynn,

2009). However, hints exist that inflammation is a regulator of epithelial-

mesenchymal communication in the pathogenesis of this disease (Selman & Pardo,

2002), (Strieter & Mehrad, 2009), (Willis, duBois & Borok, 2006).

Fig. 1: Current hypothesis of IPF pathogenesis (Wynn, 2007).

Page 14: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

14

The current hypothesis shown in Figure 1 suggests that repeating episodes of

unknown stimulus induce epithelial injury and the subsequent activation of

inflammation, platelet activation and blood clot formation. The cytokines produced

further activate epithelial cells to produce chemokines and recruit leukocytes from the

blood circulation. Specific feature of the injured place is the accumulation of cells

called myofibroblasts, which represent the hallmark of the fibrotic lung.

Myofibroblasts are cells of mesenchymal origin that produce extracellular matrix

compounds like collagen and fibronectin. During the normal wound healing process

re-epithelialization and regeneration of damaged tissue occurs. In case of chronic

injury or uncontrolled inflammation persistent myofibroblast activation leads to

excessive matrix deposition and scar formation (Wynn, 2007).

Myofibroblasts, the key mediators of fibrosis in the lung, are believed to have 3

different cellular sources (Ramos et al., 2001), (Selman, King & Pardo, 2001). The

morphological similarity with resident pulmonary fibroblasts has led to the idea that

myofibroblasts derive from these cells. Transdifferentiation of fibroblasts into

myofibroblasts in vitro was induced by stimulation with pro-fibrotic growth factor TGF-

β1 through a smad-dependent mechanism (Phan, 2002). In addition a population of

circulating bone-marrow-derived progenitor cells (fibrocytes) producing extracellular

matrix compounds was found to be an important source of lung myofibroblasts during

IPF (Hashimoto et al., 2004), (Quan et al., 2004). The contribution of alveolar

epithelial cells to myofibroblasts through a process called epithelial to mesenchymal

transition (EMT) is also extensively discussed (Phan, 2002), (Willis et al., 2006).

Based on different models of lung injury the number of cells deriving from resident

fibroblasts is estimated to be 60-80%, from epithelial cells 10-25% and from

fibrocytes 5-25% (Kisseleva & Brenner, 2008a). However, the relative contribution of

each source of myofibroblasts to the progression of pulmonary fibrosis is still under

investigation.

1.4. Treatment of IPF

1.4.1. Pharmacotherapy

There is currently no effective treatment for IPF. Different drugs have been proposed

and different clinical trials have been ongoing for anti-inflammatory, anti-fibrotic, anti-

Page 15: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

15

antioxidant and anti-coagulant therapy. The traditional anti-inflammatory treatment

with corticosteroids was shown ineffective and even some data suggest rather

harmful than beneficial effects in treated patients (Flaherty et al., 2001). In addition,

immunosuppressive and cytotoxic agents such as cyclophosphamide (Zisman et al.,

2000), azathioprine (Raghu et al., 1991) and colchicin (Douglas et al., 1998) alone or

in combination with corticosteroids have shown disappointing results. Promising

results have been observed using an anti-fibrotic agent called pirfenidone, TGF-β

antagonist, shown to significantly decrease the number of acute exacerbations in IPF

(Azuma et al., 2005), (Raghu et al., 1999).

Increased amounts of TNF-α cytokine are observed in patients with IPF. Promising

results also point to etanercept, a TNF-α antagonist, which led to a decreased rate of

disease progression (Behr, 2007), (Raghu et al., 2008).

Confusing results were observed in phase II trials using a therapy with interferon-γ

(Meltzer & Noble, 2008). Moreover, clinical phase III trials testing were terminated in

2007 owing to observed side effects of treatment vs. placebo effects.

Another potential therapy for IPF is with N-acetylcysteine, which is a molecular

precursor of the naturally occurring antioxidant glutathione. Glutathione is lacking in

the epithelial lining fluid and intracellularly in BAL cells in the lungs of patients with

IPF (Meltzer & Noble, 2008). N-acetylcysteine, which was also shown to have anti-

inflammatory effects in the lung has more satisfactory results compared to other

current used substances to deteriorate IPF (Cui A., 2009).

1.4.2. Non-pharmacological treatment

Benefits of lung transplantation were demonstrated in patients with end-stage IPF.

The survival post transplantation was 73% at 1 year, 56% at 3 years, 44% at 5 years

and 24% at 10 years (Mason et al., 2007). One likely explanation for the low long

term survival could be the older age of patients undergoing surgery. However,

rejection represents a common problem and therefore transplantation is performed in

only carefully selected patients taking risks and benefits into consideration. In

addition, life-long treatment with immunosuppressants is necessary in order to

prevent lung rejection. Another challenge taking such a decision is the timing of

pulmonary transplantation (Meltzer & Noble, 2008), (Nathan, 2005).

Page 16: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

16

As supportive therapy supplemental oxygen was also considered. However, no

improvement of quality of life was of patients receiving oxygen was observed (De

Vries, Kessels & Drent, 2001).

1.5. Animal models of IPF

Involvement of different cells and inflammatory mediators are studied in rodent

models of lung fibrosis. Bleomycin, a cancer chemotherapeutic drug, was found to

induce as a side effect fibrosis with a pathology which closely resembles that of IPF.

Although important differences in the animal pulmonary fibrosis and human IPF

disease were observed, the bleomycin model is still considered a valuable tool in

studying molecular mechanisms of pulmonary fibrosis (Agostini & Gurrieri, 2006),

(Bringardner et al., 2008).

Pulmonary radiation is another known cause of lung injury and fibrosis in humans

(Coggle, Lambert & Moores, 1986), (Movsas et al., 1997). Generation of free radicals

induces DNA damage and apoptosis. In addition, the inflammatory cascade is

activated and increased infiltration of neutrophils and alveolar macrophages occurs.

Irradiation is also a well-characterized and used model of pulmonary fibrosis in mice.

Fibrosis can also be induced by inhaled irritants like silica and asbestos. The

crystalline form of silica is toxic. Inhalation in humans leads to accumulation of

alveolar macrophages, activation of inflammation and may induce fibrosis. Exposure

to larger silica doses induces a massive fibrosis. Inhalation of asbestos fibers leads

to their deposition in the lung epithelium and induces injury. Alveolar macrophages

engulf these fibers and initiate inflammatory cascade that finally results in fibrosis. An

important element in silica and asbestos models is the involvement of the

inflammatory cascade (Barboza et al., 2008), (Robledo & Mossman, 1999).

Another experimental model proposed by Kolb et al. (2001) shows that transient

expression of IL-1β induces ALI and pulmonary fibrosis in mice. This model was used

to study the inflammatory reaction in the lung. The high expression of IL-1β was

accompanied by increased expression of IL-6 and TNF-α and later by pro-fibrotic

cytokines like TGF-β1 and platelet derived growth factor (PDGF) (Kolb et al., 2001).

The role of environmental pollutants like lipopolysaccharide (LPS) in the

pathogenesis of pulmonary fibrosis was long under discussion. Single intratracheal

administration to LPS in mice was shown to induce ALI as pathological features (Liu

Page 17: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

17

& Luo, 2006). Phan et al. published that LPS-induced ALI could rather inhibit

bleomycin-induced pulmonary fibrosis (Phan & Fantone, 1984). Current

investigations indicate that chronic, but not acute, administration of LPS may induce

fibrotic lung disorders. Indeed, in 2008 Brass et al. have shown that long-term

exposure to aerosolized endotoxin and bleomycin-induced fibrosis share common

gene activation (Brass et al., 2008). Moreover, chronic LPS-induced ALI and

pulmonary fibrosis in mice was published by two independent research groups last

year, establishing a new pulmonary fibrosis model and indicating LPS as a link

between ALI and pulmonary fibrosis (He, Zhu & Jiang, 2009), (Li et al., 2009). This

model supports the previous postulations that IPF is in part an environmental disease

and indicates a correlation between LPS pollution and the manifestation of this

devastating disease (Taskar & Coultas, 2006).

2. MECHANISMS OF INFLAMMATION

2.1. Innate immunity

Higher vertebrates have developed two interactive protective systems: the innate and

adaptive immune system. The innate immune system is immediately responsible and

ready to recognize and inactivate infectious threats, i.e. bacteria, fungi, viruses, or

destroy cancer cells. It is older and consists of soluble proteins, which bind microbial

products, and phagocytic leukocytes, i.e. macrophages, dendritic cells and

neutrophils, which float through the bloodstream and migrate into tissues at sites of

infection, or reside in tissue waiting for foreign materials (Martin & Frevert, 2005),

(Strieter, Belperio & Keane, 2002). Innate immunity has relatively broad specificities.

It is based on the recognition of common microbial motifs called pathogen-associated

molecular patterns (PAMPs) by pattern recognition receptors (PRRs) expressed on

immune cells and as a result of their activation, cytokines and growth factors further

drive the specialized T- and B-cell responses of the adaptive immune system

(Takeda & Akira, 2005), (Toews, 2001). In contrast, the adaptive immune system,

also called acquired immunity, is characterized by memory and specificity. Together,

both immune systems enable the host to react to an array of microbial and other

products (Martin & Frevert, 2005).

Page 18: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

18

2.2. Lung innate immunity

The lung possesses different mechanisms to protect itself from pathogen invasion.

Particles that enter the lungs with sizes between 1 to 5 µm are quickly and easy

removed by the mechanism of cough reflex or the mucociliary escalator. During

mucociliary clearance, ciliated and mucus-producing cells of the conductive airways

trap foreign particles and physically remove them outside the respiratory system by

coordinated upward cilia beating (Diamond, Legarda & Ryan, 2000). Beside the

importance of lung immune cells, the respiratory epithelium has been also shown to

participate in lung innate defenses through expression of PAMPs (Greene &

McElvaney, 2005), and receptors for inflammatory mediators (Bader & Nettesheim,

1996). In addition, phagocytes and airway epithelial cells produce constitutively or

upon bacterial and cytokine activation different anti-microbial agents. Anti-microbial

peptides like α- and β-defensins and cathelicidins (LL-37) act not only as

endogenous antibiotics but were also found to act as chemokines for neutrophils and

to participate in regulation of inflammation in the lung (Diamond et al., 2000),

(Hiemstra, 2006), (Tecle, Tripathi & Hartshorn, in press). An important role of SP-A

and SP-D, produced by alveolar epithelial type II cells in local innate immunity was

also indicated (McCormack & Whitsett, 2002).

A concept for organ-specific regulation of immune responses was proposed by Raz

(2007), which postulated that key inflammatory lung reactions are driven also by non-

immune cells (Raz, 2007).

2.3. Toll-like receptors and other pathogen recognition receptors

The most important PRRs in mammals are Toll-like receptors (TLRs). The first Toll-

protein was identified in the fruit fly Drosophila melanogaster, acting as a key

receptor regulating antifungal defense (Lemaitre et al., 1996). Then, a homologue of

the Toll-protein was found in vertebrates and shown to activate the release of

cytokines in human monocytes (Medzhitov, Preston-Hurlburt & Janeway, 1997). To

date, 10 TLRs have been identified in humans and 13 in mice. They are

transmembrane receptors with highly conserved regions. The extracellular domain

consists of leucine-rich repeats, flanked by cystein-rich regions. The intracellular

Page 19: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

19

region contains a Toll-interleukin 1 receptor (TIR) domain, which shows similarities

with the cytoplasmic domain of the mammalian interleukin-1 type 1 receptor (IL1R1).

Most of the receptors are expressed predominantly on the surface of antigen-

presenting cells, except for TLR-3, -7, -8 and -9 which are intracellular (Mogensen,

2009), (Tabeta et al., 2004). TLR-2 has been shown to heterodimerize with TLR-1 or

TLR-6, enabling different PAMP recognition (Takeda & Akira, 2005). Illustration of the

TLR localization on immune cells is shown on Figure 2.

Fig. 2: Cellular localization of TLRs on antigen presenting cells (MacKichan, 2005)

Other PRRs also expressed on lung immune cells are scavenger receptors

(Palecanda et al., 1999), (Resnick et al., 1993), platelet-activating factor receptors

(Stengel et al., 1997), (Thivierge & Rola-Pleszczynski, 1995) or nucleotide

oligomerization domain (NOD) 1 and 2 receptors, found to be expressed

intracellularly (Chamaillard et al., 2003), (Girardin et al., 2003).

Epithelial cells represent a significant part of the lung tissue and therefore the

contribution of these cells to the innate immunity defense is an interesting area of

current research. Indeed, primary tracheal and bronchial epithelial cells were shown

Page 20: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

20

to express TLRs 1-6 and 9 (Greene & McElvaney, 2005); (Platz et al., 2004). In

contrast to immune cells, TLR-4 expression in airway epithelial cells and the A549

alveolar epithelial cell line was mainly observed in the intracellular compartment, as

also seen in interstitial epithelial cells (Guillot et al., 2004). Another study has shown

that TLR2 was predominantly expressed on monocytes with only little TLR-2 being

detected in airway epithelium (Armstrong et al., 2004), (Mayer & Dalpke, 2007). In

agreement, Uehara et al. found that various human epithelial cells, including from

lung, express functional TLR-4, NOD1 and NOD2 to produce rather anti-microbial

products like β-defensin 2 than inflammatory mediators (Uehara et al., 2007). All

these data indicate that epithelial cells possess a number of differences in PRRs

expression grade and function, compared to professional immune cells.

2.4. Pathogen associated molecular patterns

PRRs recognize a large array of bacterial, fungal and viral PAMPs (Takeda & Akira,

2005). In the current study one focus is on characterization of PAMPs of bacterial

origin – lipopolysaccharide (LPS), peptidoglycan (PGN) and Staphylococcal

enterotoxin B (SEB). Lower respiratory tract infections with these bacterial

compounds is known to cause sepsis and death in humans (Knapp, in press).

2.4.1. Lipopolysaccharide

Lipopolysaccharide (LPS), the major immunostimulatory compound of Gram-negative

bacteria, was shown to be recognized by TLR-4 (Akira, Takeda & Kaisho, 2001),

(Takeda & Akira, 2005). LPS, also termed endotoxin, is an amphiphilic molecule and

consists of an outer part made of the O-antigen, a core region and a lipid A anchor

(Figure 3) (Caroff et al., 2002), (Rietschel et al., 1996). The O-antigen is formed by

repeating oligosaccharide sequences, highly variable among the Gram-negative

bacterial strains. The lipid A anchor, also called glycolipid, is highly conserved and is

responsible for the immunostimulatory potency of LPS (Galanos et al., 1985),

(Tanamoto et al., 1984). The recognition of LPS is extensively studied. LPS initially

interacts with a serum protein, LPS-binding protein, which transfers LPS to CD14

receptor, expressed on macrophages. A cell surface protein MD-2 is required for

binding to TLR4. Accordingly, the LPS recognition complex consists of CD14, MD-2

Page 21: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

21

and TLR-4 (Toews, 2001). C3H/HeJ mice with non-functional TLR4 display impaired

LPS responses and are highly susceptible to Gram-negative infections (Poltorak et

al., 1998), (Wright et al., 1990).

Fig. 3: Structure of LPS (http://pathmicro.med.sc.edu/fox/lps.jpg).

2.4.2. Peptidoglycan

The bacterial cell wall of both Gram-negative and Gram-positive bacteria contains

peptidoglycan (PGN). PGN is a polymer of repeating units of N-acetyl-muramic acid

(MurNAc) and N-acetyl-glucosamine (GlcNAc). The carboxyl group of MurNAc is

linked to peptide subunits of four to five L- and D-amino acids that are cross-linked

with the peptide chain of the next MurNAc (D-alanine to mesodiaminopimelic acid).

The precise sequence of the peptide chain is species dependent, but it mainly

contains L-alanine, D-glutamine, lysine or diaminopimelic acid. Peptide bridges

between amino acids located in different glycan chains lead to the formation of a

complex three-dimensional network around the cytoplasmatic membrane (Weidel &

Pelzer, 1964). This arrangement of polymeric glycan, cross-linked by peptides, plays

a major role in the determination of cell shape and in maintenance of the physical

integrity of the bacterium. Although PGN has been demonstrated to play an important

role in bacterial recognition, a controversy remains whether PGN is recognized by

Page 22: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

22

extracellular TLR-2 (Asong et al., 2009), (Dziarski & Gupta, 2005) or has to be

internalized to activate intracellular signaling via cytosolic Nod1/2 receptors (Girardin

et al., 2003).

2.4.3. Staphyloccocal enterotoxin B

Staphyloccocal enterotoxin B (SEB) is an exotoxin produced by the Gram-positive

bacterium Staphylococcus aureus. It is also termed superantigen because of its

ability to bind to major histocompatibility complex class II proteins on target cells and

stimulate proliferation of specific receptors on T cells (Krakauer, 1999).

After inhalation, clinical features include fever, respiratory complaints (cough,

dyspnea), and gastrointestinal symptoms. Severe intoxication results in pulmonary

edema, shock, and death. SEB can be easily aerosolized and therefore, in some

countries it is produced as a biological weapon (Mattix et al., 1995).

2.5. Toll-like receptor signaling

Binding of specific ligands to their respective TLRs induces receptor dimerization and

subsequent conformational change allows recruitment of cytoplasmic TIR-domain

adaptor proteins. MyD88 was the first adapter molecule identified (Burns et al., 1998)

as playing a major role in the TLR signaling pathway. MyD88 recruits signaling

molecules including IRAK family kinases and TRAF6 allow downstream activation of

NFκB transcription factors and MAP kinases which lead to the expression of

adhesion molecules, chemokines, cytokines and colony-stimulatory factors and

interferon regulatory factors (IRFs), which in turn induce the expression of type I

interferon and interferon-induced antiviral genes (Mizgerd, 2006), (Mogensen, 2009;

Takeda & Akira, 2005). The best studied and related to observed pro-inflammatory

reactions is the NFκB signaling pathway.

MyD88 is recruited to all TLRs except for TLR-3 (O'Neill & Bowie, 2007). TLR-3

signaling is coordinated by TRIF adaptor protein and links to IRF downstream

pathways (Figure 2).

Page 23: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

23

2.6. Inflammatory cytokines

Cytokines are low molecular proteins or glycoproteins that mediate processes of

chemotaxis, growth, differentiation and cell death (Toews, 2001). Relevant cytokines

for the investigations in this thesis will be discussed here.

2.6.1. Tumor necrosis factor-α

Tumor necrosis factor-α (TNF-α) is one of the most important early pro-inflammatory

cytokines produced primarily by macrophages/monocytes upon microbial challenge

(Strieter et al., 2002). TNF-α is produced as a 26 kDa precursor protein which is first

displayed on the cell membrane (membrane TNF-α) and then cleaved by a protease

called TNF-α converting enzyme to 17 kDa non-glycosylated TNF-α. The mature

TNF-α protein is biologically active as a trimer and binds to two different TNF-α

membrane receptors - TNF-α receptor 1 (TNFR1, 55 kDa) and TNF-α receptor 2

(TNFR2, 75 kDa), which possess 1 to 6 cysteine-rich domains in the extracellular

part. TNF-R2 is mainly expressed on T-lymphocytes. TNFR1, also called Fas

receptor, is the universal receptor and therefore its signaling has been largely

studied. Upon ligand binding, TNFR1 trimerizes and activates downstream nuclear

factor NFκB, similar to TLR signaling and respectively enhancing transcription of

genes mediating innate immune responses (Idriss & Naismith, 2000), (Tang, Hung &

Klostergaard, 1996), (Toews, 2001), (Verstrepen et al., 2008).

2.6.2. Interleukin-1 family of cytokines

Interleukin-1 (IL-1) cytokine family includes IL-1α and IL-1β isoforms, encoded by 2

distinct genes, and their naturally occurring inhibitor IL-1 receptor antagonist (IL-1ra).

The members of this family are characterized by extracellular immunoglobulin (Ig)-

like structures and the presence of intracellular TIR domain. IL-1α and IL-1β are

synthesized as precursor molecules without leading sequence with a molecular

weight of 31 kDa. Processing of each precursor by a protease called calpain for IL-1α

and IL-1β converting enzyme (ICE) for IL-1β leads to the mature forms of 17 kDa and

16 kDa respectively. The mature IL-1α protein is mainly membrane-associated, while

IL-1β is a secreted protein. Both α and β isoforms bind to two different receptors IL-

1R1 and IL-1R2, but signal transduction is mediated only by IL-1R1. IL-1R2 contains

Page 24: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

24

a very short cytoplasmic tail and cannot convey intracellular signals. Upon binding of

IL-1, IL-1R1 interacts with IL-1 receptor accessory protein (AcP) and forms a

functional receptor complex. With the help of adapter molecules downstream IL-1

pathways overlap with TNF-α and TLR signaling (Figure 4). Inflammatory studies on

IL-1 cytokines concentrate on the role of IL-1β as a predominant secreted cytokine

upon bacterial challenge (Dinarello, 1996), (Sims, Giri & Dower, 1994), (Strieter et al.,

2002).

IL-1β and TNF-α cytokines share similar pro-inflammatory functions, but structural

analyses show only a very short amino acid sequence of homology between the two

proteins and no overlap in receptor binding sites. However, computer analysis of the

second structure shows that these proteins have 8 β-strands and no α-helix

structures in common (Larrick & Kunkel, 1988). All nucleated cells possess functional

IL-1 and TNF-α receptors and these cytokines play a key role in initiation and

augmentation of inflammatory responses (Kelley, 1990).

Fig. 4: Receptor recognition of TNF-α, IL-1, and LPS activates overlap in NFκB

signaling pathway (Strieter et al., 2002).

Page 25: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

25

2.6.3. Interleukin-6

Interleukin-6 (IL-6) is a cytokine produced by alveolar macrophages and blood

monocytes upon stimulation with LPS, IL-1β and TNF-α. Receptors for IL-6

recognition are expressed on different lung cells including epithelial cells, alveolar

macrophages and T-lymphocytes. Alveolar macrophages produce large amounts

upon endotoxin exposure and lower IL-6 amounts when exposed to IL-1β or TNF-α

(Kotloff, Little & Elias, 1990). IL-6 expression can be also induced by direct IL-1β and

TNF-α stimulation of the lung epithelial adenocarcinoma cell line A549 (Crestani et

al., 1994).

On systemic level IL-6 is the most important pro-inflammatory mediator of acute

phase response and fever. However, studies have also evaluated a possible anti-

inflammatory effect of this cytokine on systemic level and in the lung. Recombinant

IL-6 reduced TNF-α release in mice administrated with LPS intratracheally (Ulich et

al., 1991). Moreover, LPS administration in the lung in IL-6 gene deficient mice

induced increased neutrophils influx and significantly higher amounts of pro-

inflammatory cytokines TNF-α and macrophage inflammatory protein-1α (MIP-1α)

compared to cytokines in wild type mice (Xing et al., 1998).

2.6.4. Interleukin-8

Interleukin-8 (IL-8) belongs to the group of C-X-C chemokines characterized by

conserved cysteine residues (C), separated by another amino acid (X), and regulates

chemotaxis, adhesion and neutrophils activation in the lung (Strieter et al., 2002),

(Toews, 2001). Peripheral blood monocytes and alveolar macrophages are a

significant IL-8 source upon pathogen activation (Porreca et al., 1999), (Strieter et al.,

1990). In vitro studies also show that primary alveolar epithelial cells and cell lines,

including A549 are able to produce IL-8 upon TNF-α and IL-1β activation (Coulter et

al., 1999), (Henriquet et al., 2007), (Standiford et al., 1990).

Better understanding of the role of chemokines in the mechanisms of respiratory

diseases is still needed.

2.6.5. Transforming growth factor-β

Transforming growth factor-β (TGF-β) is growth factor family of cytokines which

exists in 3 isoforms (β1, β2 and β3) with 60 to 80% amino acid homology. In lavage

Page 26: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

26

fluid of healthy volunteers TGF-β was about 5 ng/ml (Kelley, 1990). It plays an

important role in different processes of growth arrest, apoptosis, transformation,

proliferation and differentiation (Agostini & Gurrieri, 2006), (Ahmed & Nawshad,

2007). Interestingly, TGF-β may induce completely opposite biological effects in

different effector cells - activation of proliferation in fibroblasts and inhibition of

proliferation in epithelial cells (Eickelberg, 2001). TGF-β is abundant in the lung and

is produced by a large number of cells in a latent form (Kelley, 1990). The mature

latent form is produced as an inactive homodimer together with latency associated

peptide. This latent form must be activated (transient acidification, alkanization or

proteases) before receptor binding. The active form of TGF-β binds to specific type II

receptors and a tetramer complex is formed through recruitment of type I receptor.

An additional type III receptor has been shown to augment TGF-β responses

(Massague & Gomis, 2006). Well studied is the smad signaling, although TGF-β

smad-independent pathways were also described (Derynck & Zhang, 2003),

(Eickelberg, 2001), (Zhang, 2009). Accumulating data suggest that adhesion

molecules integrins control also TGF-β activation, as well as regulate TGF-β

signaling pathway in health and in diseases like COPD, IPF and cancer (Margadant

& Sonnenberg, in press).

3. IN VITRO MODELS OF PULMONARY INFLAMMATION AND DISEASE

3.1. In vitro models of pulmonary inflammation

In vivo inflammatory studies allow observations within the whole lung. However, the

contribution of single lung cell populations cannot be determined in such a complex

system. Thus, in vitro studies are helpful in understanding details of mechanisms of

lung inflammation. In vitro inflammatory models usually consist of isolated pulmonary

cells - alveolar macrophages or type II epithelial cells. However, it should be kept in

mind that neutrophils, endothelial cells, fibroblasts and dendritic cells are also

discussed to play a role in lung inflammation.

Page 27: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

27

3.1.1. Epithelial cells

In vitro approaches to model the epithelium of the respiratory tract ranked in order of

diminishing complexity include isolated perfused organs, tissue explants and cell

culture models. The epithelial cell cultures include primary cultures with a limited life

span and immortalized tumor-derived or virus-transformed cell lines. A schematic

overview of the classification is given in figure 5.

Fig. 5: A) In vitro models of the lung epithelium. B) Subdivision of cell culture models.

For inflammatory studies cell culture models are largely used. Primary cells represent

the native epithelium better but their use faces certain limitations. These cells have to

be freshly isolated each time, isolation yield is very low and donor variations are

observed. Useful techniques and protocols for isolation of primary cells of the

respiratory tract were published by Rogers and Donnely (Rogers & Donnelly, 2001).

An advantage of using cell lines is that they are of known origin, and their immortal

nature allows prolonged studies. They give usually reproducible results, they are

easy to maintain and have low costs in comparison with other in vitro epithelial

models.

Transformed cells are usually manufactured by transformation of primary cells with

constructs containing the large T antigen of the SV40 virus or human papilloma virus

(Steimer, Haltner & Lehr, 2005).

Carcinoma cell lines are derived from various lung tumors. The most often used

cancer-derived model of the alveolar epithelium is the A549 cell line, isolated from

lung adenocarcinoma. This cell line shows similarities to the type II alveolar cells like

synthesis of phospholipids, presence of lamellar bodies and microvilli. However, they

isolated perfused organs

isolated tissues

cell culture models

cell culture models

cell lines primary cells

transformed cancer derived

A B

Page 28: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

28

are less suited for transport studies where other cell lines like Calu-3, BEAS-2B,

16HBE14o- have been preferably used (Kelley, 1990), (Steimer et al., 2005).

3.1.2. Macrophages

Isolation of alveolar macrophages is relatively simple procedure and only requires

bronchoalveolar lavage in mice or humans. Seeding of the isolated cells allows

selection of the alveolar macrophages, shown to strongly adhere on the cell culture

plate. The major disadvantage using mice is the low yield and a large number of

animals needed to obtain sufficient number of cells. Therefore, alternatives have

been proposed to replace the use of these cells (Knapp, in press).

Human blood monocytes can be isolated from a whole blood together with

lymphocytes in a mixed population called peripheral blood mononuclear cells (PBMC)

and further purified, e.g. by magnetic cell sorting (Radbruch et al., 1994). Both, blood

monocytes and peripheral blood mononuclear cells are commonly used primary cells

in studies of innate immunity (Krakauer, 2002), (Wewers & Herzyk, 1989).

Immunostimulatory effects of different pathogen molecules, especially LPS, were

tested and comparative cytokine/chemokine profiles of blood monocytes and alveolar

macrophages were evaluated in vitro. Alveolar macrophages were shown to have >

20 fold decreased ability to produce IL-1β cytokine compared to blood monocytes,

measured by enzyme linked immunosorbent assay (ELISA). Measurement of total IL-

1β amounts in lysates indicates decreased processing of IL-1β precursor protein in

alveolar macrophages and respectively accumulation intracellularly (Strieter et al.,

1989), (Wewers & Herzyk, 1989). In contrast, alveolar macrophages were shown to

have 6 to 8 fold increase in TNF-α production, in comparison with blood monocytes.

When both cell types were compared for IL-6 expression, it was appreciated that

alveolar macrophages produce significantly more of this cytokine than blood

monocytes in response to endotoxin (Kotloff et al., 1990). Moreover, LPS was shown

to induce similar IL-8 expression in alveolar macrophages and blood monocytes

(Kelley, 1990).

Another alternative to primary cells is the use of human monocytic (THP-1, U937) or

macrophage Mono-Mac-1 cell lines. SV-40 transformed alveolar macrophage cells

line (MH-S) or macrophage-like cell line (RAW 264.7) from mice were also applied in

studies of in vitro inflammation (Knapp, in press), (Mbawuike & Herscowitz, 1989).

Page 29: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

29

3.1.3. Co-culture models

An important role of both, macrophages and alveolar epithelial cells in innate immune

responses as mono-cultures has been suggested and discussed in details. However,

it is still unclear how is the expression of macrophage inflammatory cytokines upon

bacterial compounds modulated in the lung by the presence of alveolar epithelial

cells? Is this modulation stimulus-dependent or common for all PAMPs? Is it

dependent on cell-cell contact mechanisms? Even more intriguing to understand, is

which mechanisms modulate cytokine expression in a co-culture system of

macrophages and lung epithelial cells.

3.2. In vitro models of fibrosis

A better understanding the etiology of IPF may suggest new therapeutic targets.

Therefore, in vitro models of pulmonary fibroblasts, fibrocytes and lung epithelial

cells, are applied to characterize the contribution of each cell population in the

pathogenesis of IPF. Primary human lung fibroblasts, HFL-1 (embryonic) and IMR-90

human lung fibroblasts cell lines were used to assess the role of resident fibroblasts

and factors inducing the accumulation of myofibroblasts. Primary lung epithelial cells

and cell lines were used to study the process of EMT.

3.2.1. Epithelial to mesenchymal transition

The new hypothesis of EMT in the pathogenesis of IPF supports that fully

differentiated epithelial cells transform into mesenchymal cells (fibroblasts and

myofibroblasts). This EMT is a form of metaplasia and does not always require cell

division. During EMT epithelial cells lose their typical characteristics like cell-cell

contact, polarity, adhesion (E-cadherin), cytoskeleton proteins (cytokeratins) and

acquire spindle-like shape and mesenchymal markers like vimentin, α-smooth

muscle actin and collagen.

EMT was recently classified into 3 different types. Type 1 is crucial during

embryogenesis and organ development, where epithelial cells are able to give rise to

the mesenchyme. EMT can be also re-activated in adults during wound healing,

tissue reorganization and organ fibrosis (type 2 EMT); cancer progression and

metastasis (type 3 EMT). The opposite process, called mesenchymal to epithelial

Page 30: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

30

transition (MET), is also observed during embryonic development and in adults

(Kalluri & Weinberg, 2009), (Xu, Lamouille & Derynck, 2009).

So far, it is still unclear whether alveolar epithelial type I, type II or both of the cells

undergo EMT (Figure 6). The current knowledge suggests that lung injury activates

type II cells (ATII) to undergo EMT and type I cells (ATI) more likely undergo

apoptosis (Kisseleva & Brenner, 2008b), (Willis et al., 2006).

Another type of cell plasticity is the transdifferentiation of ATII cells into ATI as a

normal mechanism of cell regeneration in the alveolar spaces. Studies in vitro show

that ATI cells may also transdifferentiate into type II cells but the occurrence of this

process in vivo is still in doubt (Kim et al., 2006), (Willis et al., 2006).

Fig. 6: Mechanisms of epithelial cell plasticity. Adopted from Willis et al. (Willis et al.,

2006).

3.2.2. Initiators of EMT in vitro

Different growth factors, transforming growth factor family (TGF-β), epidermal growth

factor (EGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF)

(Margadant & Sonnenberg, in press), as well as wnt ligands (Konigshoff &

Eickelberg, in press) and matrix metalloproteinases (Selman & Pardo, 2006) have

been shown to induce EMT in vitro. The most potent recognized in vitro (Kasai et al.,

2005), (Liu, 2008), (Willis et al., 2005) as well as in vivo (Kim et al., 2006) EMT

inducer is the TGF-β1. Therefore, ongoing studies evaluate the precise signaling

EMT

MET

injury

injury ? Apoptosis/ necrosis

ATII

ATI

?

Myofibroblast

Page 31: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

31

pathways of TGF-β1 induced EMT in lung epithelial cells. Most commonly, smad-

signaling pathway has been activated upon ligand binding. Recruitment of smad 2

and 3 transcriptional factors to the TGF-β1 receptor complex is necessary for

initiation of the downstream cascade. Smad 2 and 3 molecules partner with smad 4

and the complex of smad 2, 3 and 4 transcriptional factors translocates to the

nucleus. As a result of smad-mediated transcriptional regulation, activation of target

genes results in repression of epithelial and activation of mesenchymal marker gene

expression (Willis et al., 2006), (Xu et al., 2009).

The findings that TGF-β is a “master switch” for EMT and fibrosis suggested that IPF

could be managed by anti-TGF-β therapy. Although promising results have been

shown still better characterization of other involved mediators is necessary to

improve IPF treatment.

Indeed, EMT was recognized as the possible link between lung inflammation and

fibrosis. Due to the late discovery of the chronic LPS–induced model of pulmonary

fibrosis, the hypothesis that IPF is in part environmental disease only entered the

discussion recently. Therefore, there are still no reports in vitro or even in vivo about

the role of environmental factors, particularly LPS and involved inflammatory

mediators in the EMT process.

Page 32: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

32

4. AIMS OF THE STUDY

Alveolar macrophages, the most abundant immune cells in the alveolar spaces, are

key players in maintaining homeostasis and regulating immune defenses in the lung.

However, the epithelial cells lining the airways are the first point of contact for inhaled

pathogens. In recent years the role of epithelial cells in lung innate immunity has

become a focus of research interest, as it was discovered that airway epithelial cells

possess not only passive barrier function, but also actively contribute to fighting

respiratory pathogens.

• The aim of the first part of the study was to establish an in vitro lung co-culture

model to study the contribution of epithelial cells to inflammation upon stimulation

with bacterial stimuli from Gram-positive and Gram-negative bacteria. In addition

mechanisms of LPS-induced inflammatory modulation were characterized. The co-

culture model was composed of the alveolar epithelial type II like A549 cell line and

peripheral blood mononuclear cells (PBMC).

Recent in vivo investigations suggest that chronic exposure to environmental factors

like LPS induces lung inflammation and subsequent chronic lung diseases, including

pulmonary fibrosis. Myofibroblasts, the main cell population in pulmonary fibrosis, are

central in the pathogenesis of this disease. It is thought that epithelial cells may

differentiate into myofibroblasts through a process called epithelial to mesenchymal

transition (EMT). Recent understandings in the molecular mechanisms of fibrosis

show that EMT is the missing link between inflammation and fibrosis and indicate an

important role of TGF-β1 as master switch for EMT induction. However, the role of

LPS in the process of EMT was not studied so far.

• The aim of the second study was to investigate whether LPS stimulation of PBMC

in vitro induces an inflammatory response that mediates EMT of lung epithelial A549

cells. A subsequent goal was to establish the identity of the cytokine(s) produced by

PBMC which mediated EMT of A549 cells.

Page 33: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

33

II.

Lung epithelial cells constitutively produce an immunomodulatory

factor for cytokine release by mononuclear cells

Page 34: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

34

1. ABSTRACT

The contribution of airway epithelial cells to the innate immune response in the lung

has been a focus of recent interest. A human mixed co-culture system of lung

epithelial cell line A549 and primary peripheral blood mononuclear cells (PBMC)

allowed us to study modulation of IL-1β, TNF-α, IL-6 and IL-8 expression triggered by

lipopolysaccharide (LPS), staphylococcal enterotoxin B (SEB) and peptidoglycan

(PGN) in comparison to respective monocultures. All immune stimuli induced

significantly less TNF-α and much higher IL-6 release in co-cultures compared to

PBMC alone. IL-1β release was stimulus-dependent: down-regulated upon LPS, up-

regulated upon PGN and not affected upon SEB stimulation. IL-8 amounts were

significantly increased in LPS and PGN-stimulated co-cultures and slightly increased

upon SEB stimulation. Transwell experiments showed that LPS-induced cytokine

modulation in co-culture is triggered by soluble factors. Using neutralizing IL-1β

antibody we demonstrated that PBMC-derived IL-1β mediates augmented IL-6 and

IL-8 production in LPS-stimulated co-cultures. A still unknown soluble factor in

conditioned supernatant of resting A549 cells was shown to down-regulate TNF-α

release by PBMC on mRNA level. Characterization of this anti-inflammatory

compound suggests a peptide nature and opens an interesting area of future

research.

In conclusion, lung epithelial cells modulate the inflammatory response by down-

regulating TNF-α and increasing IL-6 and IL-8 in co-cultures and must therefore be

considered actors in shaping lung inflammation.

Page 35: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

35

2. INTRODUCTION

The human lung is exposed to a large number of airborne pathogens as a result of

the daily inhalation of 10 000 liters of air. The observation that respiratory infections

are nevertheless rare is due to the presence of an efficient host defense system in

the lung. The airway epithelium represents a primary site of entrance and deposition

of potentially pathogenic microorganisms into the body, and therefore is equipped

with a variety of mechanisms to avoid infections (Bals & Hiemstra, 2004), (Diamond

et al., 2000). The epithelial lining fluid in the lower respiratory tract contains immune

cells: alveolar macrophages, T- and B-cells, neutrophils, eosinophils, mast cells and

dendritic cells. Alveolar macrophages, derived from blood monocyte differentiation,

account for up to 95% of the cells recovered by bronchoalveolar lavage (Bingisser &

Holt, 2001) and play a critical role in maintaining homeostasis, host defense,

response to foreign substances and tissue remodeling (Losa Garcia et al., 1999). To

combat infection, the phagocytic cells of the innate immune system express pattern

recognition receptors (PRRs), which recognize pathogen-associated molecular

patterns (PAMPs) on the surface of microorganisms. Toll-like receptors (TLRs)

function as major PRRs in mammals. TLRs are membrane-bound molecules

expressed on the surface or within intracellular compartments that participate in the

recognition of different microbial compounds. 10 members of this family (TLR 1-10)

have been identified in humans so far. The most prominent PAMP,

lipopolysaccharide (LPS or endotoxin) from the cell wall of gram-negative bacteria, is

recognized by TLR-4 (Akira et al., 2001), (Takeda & Akira, 2005). Intracellular

recognition of bacteria appears to also involve a TLR-independent system. Recent

studies indicate that nucleotide-binding oligomerization domain (NOD) 1 and 2

proteins recognize peptidoglycan (PGN) present in the cell wall of Gram-positive

bacteria (Mogensen, 2009). Staphyloccocal enterotoxin B (SEB, superantigen) acts

by directly binding to major histocompatibility complex class II molecules on antigen-

presenting cells and is recognized by αβ receptors on T-cells. Therefore, it is involved

in activation of the adaptive immune system response (Choi et al., 1989), (Krakauer,

2001).

A number of studies indicated that TLRs are also expressed in airway epithelial cells

(Gomez & Prince, 2008), (Muir et al., 2004). It was demonstrated that primary

tracheo-bronchial epithelial cells (Becker et al., 2000), alveolar epithelial type II A549

Page 36: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

36

and bronchial epithelial BEAS-2B cell lines (Schulz et al., 2002) express mRNA for

TLRs 1-6. Studies on TLR-4 localization in A549 and BEAS-2B cell lines have

described a perinuclear location in association with the Golgi apparatus, rather than

at the cell surface. Despite TLR-4 expression these cell lines were not directly

responsive to LPS in serum-free medium, rather in the presence of serum and

concentrations higher the 1µg/ml (Guillot et al., 2004). Furthermore, it was observed

that primary lung epithelial cells and cell lines, release IL-6 and IL-8 in response to IL-

1β and TNF-α stimulation (Coulter et al., 1999), (Henriquet et al., 2007), (Jiang,

Kunimoto & Patel, 1998). IL-1β and TNF-α, are the most important early responsive

pro-inflammatory cytokines by immune cells in innate immune response (Strieter et

al., 2002). Secretion of IL-8 chemokine during bacterial infections in the lung was a

prerequisite for recruitment of neutrophils into the alveolar space (Reutershan & Ley,

2004). Apart of acute phase reactions, IL-6 was found to participate in modulation of

lung immune responses exerting stimulus-dependent pro- and anti-inflammatory

activities (Xing et al., 1998).

An in vitro co-culture lung model was devised to study the contribution of epithelial

cells to inflammation in the lung. Here we worked with human pulmonary epithelial

type II cells (A549) and primary peripheral blood mononuclear cells (PBMC). Our

findings indicate a function of alveolar epithelial cells in modulating the inflammatory

reaction in the lung.

Page 37: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

37

3. RESULTS

3.1. A549 cells modulate inflammatory response of PBMC upon LPS, SEB

and PGN stimulation

A549 cells directly stimulated with 100 ng/ml LPS, 100 ng/ml SEB or 1 µg/ml PGN

did not release IL-1β, TNF-α, IL-6 or IL-8 (data not shown). However, the presence of

epithelial cells significantly down-regulated TNF-α and IL-1β expression of LPS-

stimulated PBMC (Figure 1A). In contrast, LPS exposure amplified IL-6 and IL-8

production in mixed co-cultures, compared to PBMC mono-cultures (Figure 1B).

A B

con LPS con LPS0

2

4

6TNF-αααα IL-1 ββββ

***

***

[ng

/ml]

con LPS con LPS0

50

100

150

200 IL-6 IL-8

******

[ng

/ml]

C D

con PGN SEB con PGN SEB0

5

10

15

20 TNF-αααα IL-1 ββββ

*** ***

***

ns

[ng

/ml]

con PGN SEB con PGN SEB0

50

100

150

200

250 IL-6 IL-8

***

***

***

ns

[ng

/ml]

Fig. 1: A549 cells modulate inflammatory response of PBMC alone upon LPS, SEB and

PGN stimulation. Mixed co-culture of A549 and PBMC (white bars) or PBMC alone (black

bars) were stimulated with 100 ng/ml LPS, 100 ng/ml SEB, 1 µg/ml PGN or PBS (con). After

24 h cell-free supernatants were assayed for TNF-α and IL-1β (A); IL-6 and IL-8 (B) by

ELISA. Data are means ± SEM from 11 PBMC donors in 3 independent experiments. TNF-α

and IL-1β (C); IL-6 and IL-8 (D) release upon stimulation with PGN and SEB were measured

by ELISA. Data are means ± SEM, 8 PBMC donors in 2 independent experiments (Repeated

measures ANOVA followed by Bonferroni’s multiple comparison test).

Page 38: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

38

We investigated whether cytokine modulation is similar upon stimulation with other

PAMPs. The presence of A549 cells inhibited TNF-α expression also dramatically in

PGN- and SEB-activated PBMC. PBMC produced very low IL-1β amounts upon SEB

stimulation and cytokine amounts were not affected in co-cultures (Figure 1C). In

contrast, PGN activated PBMC to produce IL-1β and its amounts were increased in

co-cultures. SEB-stimulated PBMC in mono-culture did not produce IL-6, but a

massive IL-6 amplification was present in PGN- and SEB-stimulated co-cultures. We

also observed that PGN significantly up-regulated and superantigen tended to

increase IL-8 expression in co-culture compared to PBMC alone (Figure 1D).

3.2. Modulation of cytokine secretion by epithelial cells does not require

direct cell-cell contact

To find out whether LPS-driven inflammatory effects in the co-culture system are

modulated by cell contact mechanisms cells in mono-culture and co-culture were

seeded on 24 well normal or transwell plates with 0.4 µm pore size of the membrane

inserts (Figure 2). Cytokine release of IL-1β, TNF-α, IL-6 and IL-8 in both settings

had similar patterns arguing that the modulation is mediated by soluble factors, not

cell-cell contact.

3.3. αIL-1β, but not αTNF-α neutralizing antibody, inhibits epithelial cell-

mediated amplification of IL-6 and IL-8 release in co-culture

It was previously observed that IL-1β and TNF-α induce IL-6 and IL-8 release in A549

cells (Coulter et al., 1999), (Henriquet et al., 2007), (Jiang et al., 1998). We confirmed

these results (data not shown) and hypothesized that IL-1β and TNF-α from PBMC

mediate the amplification of IL-6 and IL-8 release in LPS-activated co-cultures.

Antibodies against IL-1β and TNF-α were used to neutralize cytokine effects upon

LPS stimulation. The neutralizing activity of the αTNF-α and αIL-1β antibodies was

demonstrated by inhibited IL-6 and IL-8 production in A549 cells stimulated with TNF-

α and IL-1β in quantities released by LPS-stimulated PBMC (data not shown).

Antibodies alone did not induce IL-6 and IL-8 production in non-stimulated cells.

αTNF-α antibody did not affect IL-6 and IL-8 production in LPS-stimulated co-cultures

Page 39: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

39

and PBMC. Addition of αIL-1β to LPS-activated cells resulted in no significant

difference in IL-6 and IL-8 amounts between co-cultures and PBMC (Figure 3).

con LPS LPS0

1

2

3

4

Direct contact Transwells

***

***

TNF-αααα

TN

F- αα αα

[n

g/m

l]

con LPS LPS0.0

2.5

5.0

7.5

Direct contact Transwells

***

***IL-1ββββ

IL-1

ββ ββ [

ng

/ml]

con LPS LPS0

10

20

30

40

50

60

70

80

Direct contact Transwells

***

***

IL-6

IL-6

[n

g/m

l]

con LPS LPS0

10

20

30

40

****

Direct contact Transwells

IL-8

IL-8

[n

g/m

l]

Fig. 2: Comparison of cytokine induction in co-culture vs. PBMC upon LPS stimulation

in direct cell-cell contact and on transwells. Cells in mixed co-culture (white bars) or

PBMC (black bars) were seeded on 24 well plates in direct contact or were physically

separated by transwells (polycarbonate membrane, 0.4 µm pore size). After 3 h cells were

stimulated with LPS (100 ng/ml) or vehicle (PBS), supernatants were harvested after 24 h

and analyzed by ELISA. Data are means ± SEM from 6 PBMC donors in 2 independent

experiments (Repeated-measures ANOVA with Bonferroni’s multiple comparison test).

3.4. LPS-induced inhibition of TNF-α in mixed co-culture is regulated on

post-transcriptional level

In figure 4 we show that TNF-α down-regulation in LPS-stimulated co-cultures occurs

on post-transcriptional level. 3 h LPS stimulation of cells in co-culture or PBMC alone

in the presence of 5 µg/ml Brefeldin A blocked extracellular release of TNF-α, as

expected. This allowed the observation of a significant down-regulation of

intracellular pro-TNF-α in co-culture compared to amounts detected in PBMC.

Page 40: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

40

con

LPS αααα

TNF-

αααα

αααα

TNF-

αααα

LPS +

ββββ

IL-1

αααα

ββββ

IL-1

αααα

LPS +

0

50

100

150

200

250***

***IL-8

ns

IL-8

[n

g/m

l]

con

LPS αααα

TNF-

αααα

αααα

TNF-

αααα

LPS +

ββββ

IL-1

αααα

ββββ

IL-1

αααα

LPS +

0

25

50

75

100

125

150

******IL-6

nsIL-6

[n

g/m

l]

Fig. 3: Effects of αIL-1β and αTNF-α neutralizing antibody on IL-6 and IL-8 production

in co-culture vs. PBMC. Cells in co-culture (white bars) or PBMC mono-culture (black bars)

were seeded in direct cell-cell contact settings on 48 well plates. In addition, neutralizing

TNF-α and IL-1β antibodies (1 µg/ml) were added and after 1 h cells were stimulated with

vehicle (con) or 100 ng/ml LPS for 24 h. Supernatants were collected and analyzed for IL-8

and IL-6 expression by ELISA. Data are means ± SEM and represent 2 independent

experiments, 8 PBMC donors (Repeated-measures ANOVA with Bonferroni’s multiple

comparison test).

3.5. Conditioned supernatant from resting A549 cells induces down-

regulation of TNF-α protein and mRNA in LPS-stimulated PBMC

We next tested whether the inhibitory effect on TNF-α release is exerted also by

conditioned supernatant from non-stimulated A549 cells. A549 conditioned medium

from different time points (2 h, 4 h, 8 h and 24 h) was added to PBMC and after 3 h

cells were stimulated with 100 ng/ml LPS for additional 24 h. We observed down-

regulated TNF-α secretion by LPS-stimulated PBMC in a time dependent manner

Page 41: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

41

consistent with accumulation of an inhibitory factor in the supernatant of the resting

A549 cells (Figure 5).

con Brefeldin A LPS LPS + Brefeldin A0.0

0.5

1.0

1.5

2.0

2.5A - extracellular

***

TN

F-αα αα

[n

g/m

l]

con Brefeldin A LPS LPS + Brefeldin A0.0

0.2

0.4

0.6

0.8B - intracellular

***

TN

F-αα αα

[n

g/m

l]

Fig. 4: Regulation of TNF-α expression upon LPS stimulation occurs on post-

transcriptional level, not at release stage. Cells in mixed co-cultures (white bars) vs.

PBMC (black bars) were seeded on 48 well plates and after 3 h were stimulated with vehicle

(con) or 100 ng/ml LPS in the presence or absence of 5 µg/ml Brefeldin A. After 3 h

supernatants were collected for TNF-α ELISA measurement (A). Cell pellet was resuspended

in PBS and 3 freeze-thaw cycles were performed. Cell-free supernatants were harvested and

intracellular pro-TNF-α was analyzed by ELISA (B). Data are means ± SEM (8 PBMC donors

in 2 independent experiments). Statistical differences are analyzed by repeated-measures

ANOVA followed by Bonferroni’s multiple comparison test.

Page 42: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

42

2 4 8 24 con0

1

2

3

***

time [h]

****** ***

TN

F-αα αα

[n

g/m

l]

Fig. 5: Conditioned supernatant from resting A549 cells induces down-regulation of

TNF-α in LPS-stimulated PBMC. PBMC were seeded in conditioned supernatant (76%)

from A549 (white bars) or medium (black bars) collected after different conditioning times

(2h, 4h, 8h and 24h). After 3 h PBMC were stimulated with vehicle (con) or 100 ng/ml LPS

for 24 h. Cell-free supernatants were analyzed for TNF-α by ELISA. Data represent mean ±

SEM from 3 independent experiments, 9 PBMC donors (Repeated measures ANOVA with

Bonferonni’s multiple comparison test).

The fact that the inhibitory factor was produced by resting A549 cells simplified the

investigation of whether its effect is already found on mRNA level. Stimulation of

PBMC with LPS in the presence of 24 h epithelial-derived supernatant showed a

significantly decreased TNF-α mRNA levels (Figure 6).

A549 SN Medium0

25

50

75

100

125

150

**

TN

F- αα αα

mR

NA

no

rmali

zed

to

GA

PD

H

Fig. 6: Effect of conditioned supernatant from A549 cells on TNF-α mRNA regulation in

LPS-activated PBMC. PBMC were seeded in 76% 24 h conditioned supernatant from A549

cells (white bar) or medium (black bar). After 3 h, PBMC were incubated with PBS or

stimulated with 100 ng/ml LPS. Expression of TNF-α mRNA was examined by quantitative

RT-PCR. Transcript expression was normalized to the housekeeping gene GAPDH. Control

values were subtracted from LPS values. Results are means ± SEM from 7 PBMC donors in

duplicates in 3 independent experiments (Mann-Whitney Test).

Page 43: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

43

3.6. The TNF-α inhibitory compound appears to be a peptide

We aimed to characterize the TNF-α anti-inflammatory compound in the conditioned

supernatant of resting A549 cells. We worked with 76% and 30% supernatants. Our

results indicate that TNF-α inhibitory factor is heat and freeze (Figure 7A) stable.

However, 30% conditioned supernatant treatment with 0.5 mg/ml pronase reversed

TNF-α levels to those observed in the absence of conditioning. Experiments

identifying the size, using a filter with cut-off 10 kDa, have shown that eluted 76%

conditioned medium still possessed strong TNF-α suppressing activity (Figure 7B),

similar to observed without filtering and that filtrate of 30% conditioned medium

partially lost its activity (figure 7C). These data point towards a peptide as the anti-

inflammatory agent.

Fig. 7: TNF-α inhibitory compound

appears to be a peptide. A) 30%

conditioned supernatant from A549 cells

(A549 30%, white bars) or medium (black

bars) was not-treated; heated at 95°C for 15

min; digested with 0.5 mg/ml pronase and 2 x

freeze at -80°C. B) 76% (A549 76%) and C)

30% (A549 30%) conditioned supernatant

from A549 (white bars) or medium (black

bars) was filtrated through a filter with cut-off

10 kDa and eluted fractions were used for

experiment. 70µl PBMC were added to 380

µl supernatants and after 3 h were stimulated

with vehicle or 100 ng/ml LPS. After 24 h

cell-free supernatants were analyzed by

TNF-α ELISA. Data are means ± SEM (6

PBMC donors in 2 independent

experiments). Repeated measures ANOVA

with Bonferroni’s multiple comparison test

was applied for statistical differences.

con LPS con LPS con LPS con LPS0

1

2

3

4

5

95°C/15 min pronase freeze-thaw

A A549 30 %medium

******

***ns

TN

F- αα αα

[n

g/m

l]

con LPS con LPS0

1

2

3

4

5

eluted fraction

Bmedium

A549 76%

*** ***

TN

F- αα αα

[n

g/m

l]

con LPS con LPS0

1

2

3

4

5 C

eluted fraction

A549 30%

medium

******

TN

F- αα αα

[n

g/m

l]

Page 44: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

44

4. DISCUSSION

Until recently the focus of research into the innate immune response of the lung has

been on the role of resident alveolar macrophages. In 2007, a study has shown that

various human epithelial cells, including from lung, express functional TLRs, NOD1

and NOD2 to produce anti-microbial products like β-defensin 2 rather than

inflammatory mediators (Uehara et al., 2007). In agreement we found that stimulation

of A549 cells in mono-culture with LPS, SEB or PGN did not activate these cells to

produce IL-1β, TNF-α, IL-6 or IL-8. In addition, Schulz et al. have shown that LPS

activates A549 cells to produce IL-6 and IL-8 only in µg doses and in the presence of

serum (Schulz et al., 2002). These results indicate that during infection, direct

stimulation of airway epithelial cells with bacterial compounds is not a significant

source of inflammatory mediators.

To better mimic the in vivo situation in the lung we designed an in vitro co-culture

system of A549 cells and PBMC and studied regulation of inflammation upon immune

stimulation. Here we show that the presence of lung epithelial cells modulate the

immune response of stimulated PBMC. All of the 3 chosen stimuli (LPS, PGN and

SEB) induced significantly lower TNF-α, and much higher IL-6 expression in

stimulated co-cultures compared to cytokine release in respective stimulated PBMC

mono-cultures. Early produced TNF-α cytokine from alveolar macrophages was

found to play an important role during initiation and augmentation phases of

inflammation (Kelley, 1990), (Strieter et al., 2002). IL-6 was shown as pro-

inflammatory regulator of acute phase responses and fever. However, in an LPS-

induced acute lung inflammation model IL-6 was described rather as anti-

inflammatory cytokine (Xing et al., 1998). Leemans et al. postulated differential roles

of IL-6 in lung inflammation by LTA (anti-inflammatory) and PGN (pro-inflammatory

effect) both from Staphylococcus aureus (Leemans et al., 2002). These results

indicate that indeed epithelial cells may create an anti-inflammatory milieu in the lung

through modulation of TNF-α and IL-6 produced from alveolar macrophages.

IL-8 expression in LPS- and PGN-activated co-cultures was also significantly

enhanced and tended towards an increase in SEB-stimulated co-cultures. It is known

that IL-8 is a very potent chemoatractant for neutrophils in the alveolar space (Coulter

et al., 1999), (Reutershan & Ley, 2004), therefore chemokine amounts increase as a

prerequisite for resolution of infection.

Page 45: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

45

Modulation of IL-1β expression was stimulus-dependent: lower upon LPS, higher

upon PGN and unchanged upon SEB stimulation of co-cultures compared to

stimulated PBMC. Although this latter observation was very interesting, regulatory

mechanisms for IL-1β production were not further investigated in details.

To study whether LPS-induced inflammatory responses in co-culture require direct

cell-cell contact or are driven by soluble mediators, cells were seeded in parallel in

mixed and transwell co-cultures. Our results indicated no requirement for cell-cell

contact between lung epithelial cells and blood mononuclear cells for all modulated

co-culture inflammatory mediators. A critical point of using transwell plates was the

choice of the material for membrane inserts. Here polycarbonate material was used,

which produced reproducible and reliable results. Polyester membrane inserts from

the same company in contrast resulted in confusing and non-reproducible results in

comparative experiments. This indicates that the choice of membrane material is

crucial and has to be carefully selected according to the application and cell type.

We further investigated soluble mediators involved in LPS-induced mechanisms of

IL-6 and IL-8 modulation in co-culture. Interestingly, amplification of LPS-induced IL-6

and IL-8 release has also been reported in A549-neutrophil co-cultures (Grandel et

al., 2009). Indeed, stimulation of lung epithelial cells or lung adenocarcinoma cell

lines with IL-1β and TNF-α induces expression of IL-6 and IL-8 (Coulter et al., 1999),

(Henriquet et al., 2007), (Jiang et al., 1998). A549 cells were also found to express

IL-1 type I receptor (Coulter et al., 1999) and TNF-receptor I (Burvall, Palmberg &

Larsson, 2005). Here we show that only neutralizing antibody against IL-1β, but not

TNF-α, returns IL-6 and IL-8 expression in LPS-stimulated co-cultures to levels

comparable with those in LPS-stimulated PBMC mono-cultures. These results

indicate an important role of IL-1β in augmenting the IL-6 and IL-8 response to LPS

in the lung. The importance of IL-1β, but not TNF-α, in enhancing IL-8 expression in

silica-treated lung cell co-cultures of A549 and THP-1 monocyte-derived cell line was

also described (Herseth et al., 2008). One year later, the same group showed that IL-

1β is responsible also for increased IL-6 release in silica activated lung cell co-

cultures (Herseth et al., 2009).

We further characterized LPS-induced inhibition of TNF-α in co-culture. Brefeldin A

blocks protein transport from the endoplasmic reticulum to the Golgi apparatus,

allowing the determination of unprocessed cytokines. We demonstrated that pro-

TNF-α in LPS-stimulated PBMC was already significantly down-regulated by the

Page 46: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

46

presence of A549 cells. As we had observed that already conditioned supernatant

from resting A549 cells accumulated the anti-inflammatory activity over time, we

could use this to investigate whether the unknown factor acts on mRNA level.

Therefore, avoiding direct contact between the two cell populations and resulting

difficulties with normalization of the mRNA data, we studied transcriptional TNF-α

regulation of PBMC using 24h conditioned supernatant from A549 cells. TNF-α cDNA

data were also consistent with observed extracellular and intracellular TNF-α

inhibition in co-cultures. In agreement with our observations, it was published recently

that the bronchial epithelial BEAS-2B cell line inhibited secretion of TNF-α by human

monocytes and monocyte-derived macrophages on transcriptional level (Mayer et al.,

2008). However, this group tried to identify anti-inflammatory mediator(s) specifically

produced by lung epithelial cells. In our hands, substitution of A549 cells with

epithelial cervix HeLa cell line and embryonic kidney HEK293T cell lines in mixed co-

cultures with PBMC also resulted in down-regulated TNF-α production (unpublished

observations). These results indicate that inhibition of TNF-α release in co-culture is

not specific to the A549 cell line or epithelial cells of the lung.

Finally, initial characterization of the anti-inflammatory factor in A549-derived

conditioned medium showed that it is heat and freeze stable. This compound filters

through a 10 kDa cut-off filter membrane, but is inactivated by a protease called

pronase. These data suggest a peptide as a possible candidate but further

experiments are needed to prove its nature. Indeed, A549, HEK293T and HeLa cells

were shown to constitutively produce abundant amounts of peptide called beta

defensins 1 (Pazgier et al., 2006), (Valore et al., 1998), (Zucht et al., 1998), (Jang et

al., 2004), (Hegedus et al., 2008). Beta defensin 1 is a member of the defensins

family which was shown to be heat stable (Tunzi et al., 2000) and to have a size of

47 amino acids residues (5 kDa). In addition, defensins were recognized to act not

only as anti-microbial peptides but are also described as anti-inflammatory mediators

in innate immune responses (Semple et al.), (Tecle, Tripathi & Hartshorn).

Taken together, lung epithelial cells in LPS-stimulated co-cultures induce modulatory

effects shown by amplification of IL-6 and IL-8 in co-culture, induced by IL-1β and

down-regulation of TNF-α on protein and mRNA level, caused by a still unknown

soluble factor. The identification and characterization of a possibly novel anti-

inflammatory peptide would greatly improve our understanding of the regulation of

Page 47: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

47

inflammation and treatment of inflammatory diseases. Further co-culture studies are

necessary in better understanding the intimate inflammatory mechanisms in the lung.

Page 48: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

48

III.

Conditioned supernatant from lipopolysaccharide stimulated blood

mononuclear cells induces epithelial to mesenchymal transition in

A549 cells: role of IL-1β and TNF-α

Page 49: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

49

1. ABSTRACT

Recent investigations postulate that lung inflammation due to environmental

pollutants, including lipopolysaccharide (LPS) plays an important role in the

development and progression of different chronic lung diseases, including idiopathic

pulmonary fibrosis. Advances in understanding the molecular mechanisms of this

disease point to epithelial to mesenchymal transition (EMT) as a link between

inflammation and fibrosis, but so far the role of LPS in the process of EMT has not

been investigated. Therefore, we hypothesized that conditioned supernatant from

LPS stimulated peripheral blood mononuclear cells (PBMC) induces an inflammatory

cascade and subsequent EMT in lung epithelial type II A549 cell line. EMT in

epithelial cells was evidenced after 3 days by alteration in cell morphology assessed

by phase contrast microscopy and analysis of Western blot marker expression. A549

cells alone stimulated with LPS did not undergo EMT. Conditioned supernatant from

LPS stimulated PBMC activated A549 cells to undergo EMT, evidenced by

acquisition of mesenchymal morphology, down-regulation of epithelial cytokeratin

and up-regulation of mesenchymal vimentin expression. Direct stimulation with IL-1β

and TNF-α, but not IL-6, IL-8, MCP-1, MIP-1α and IL-10, shown to be produced from

PBMC upon LPS-stimulation, induced fibroblast-like morphology, decreased

cytokeratin and increased vimentin expression in A549 cells. Using neutralizing

antibodies for IL-1β, TNF-α and IL-1 receptor antagonist added to conditioned

supernatants from LPS-stimulated PBMC showed that IL-1β and TNF-α cytokines are

responsible for the observed EMT effect. Thus, our findings indicate a central role of

IL-1β and TNF-α in the in vitro LPS-driven EMT process in A549 cells.

Page 50: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

50

2. INTRODUCTION

Idiopathic pulmonary fibrosis (IPF) is a progressive and relentless inflammatory and

fibrotic lung disease. It has been recognized as the most lethal interstitial lung

disease and lung transplantation represents the only option for cure. IPF is

characterized by excessive matrix deposition and destruction of the lung architecture

(Agostini & Gurrieri, 2006), (Selman & Pardo, 2006).

The hallmark of pulmonary fibrosis is the abundance of myofibroblasts. It is

considered that these cells are derived from three different sources: resident lung

fibroblasts, blood-borne cells called fibrocytes, and from epithelial cells through

epithelial to mesenchymal transition (Scotton & Chambers, 2007). EMT is recognized

as a physiological mechanism occurring during embryonic development (Acloque et

al., 2009), and this process may be reactivated in adults during wound healing, tissue

regeneration, organ fibrosis and cancer progression (Kalluri, 2009). EMT is

characterized by loss of cell polarity and a change from cuboidal to fibroblastoid cell

shape. In addition, reorganization of the cytoskeleton system, down-regulation of

epithelial and acquisition of mesenchymal characteristics occurs (Kalluri & Neilson,

2003), (Xu et al., 2009), (Zavadil & Bottinger, 2005).

An experimental animal study has identified epithelial-derived mesenchymal cells in

TGF-β1 driven fibrosis (Kim et al., 2006). Wu et al. observed airway EMT in

bleomycin-induced peribronchial fibrosis mice (Wu et al., 2007). EMT can also be

observed in vitro in a variety of normal epithelial cells and epithelial-derived

carcinoma cell lines by stimulation with soluble growth factors, i.e. transforming

growth factor-β family (TGF-β), fibroblast growth factor (FGF), epidermal growth

factor (EGF) or hepatocyte growth factor (HGF) (Kasai et al., 2005), (Margadant &

Sonnenberg, in press), (Zhang, Dong & Yang, 2006).

In the inflammatory milieu of chronic lung diseases, the major cytokines present are

IL-1β and TNF-α (Araya & Nishimura, 2010). In an in vivo lung injury model, transient

over-expression of IL-1β in rat airway epithelial cells resulted in increased expression

of TNF-α and IL-6, followed by an increase in bronchoalveolar lavage of platelet

derived growth factor (PDGF) and TGF-β1 (Kolb et al., 2001). In a bleomycin-induced

animal model of pulmonary fibrosis soluble TNF-α was found as a prerequisite for the

development of this disease (Oikonomou et al., 2006), suggesting a relation between

IL-1β and TNF-α inflammatory cytokines and the conversion from inflammation into

Page 51: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

51

fibrosis. As a common link between inflammation and fibrosis recent findings point

EMT. This understanding was supported by a report that a cocktail of IL-1β, TNF-α

and IFN-γ augments TGF-β1 induced EMT in lung epithelial A549 cells (Liu, 2008).

The role of environmental factors such as pyrogenic bacterial or fungal components

in the pathogenesis of IPF is a subject of current investigations. LPS (endotoxin) is

the predominant pyrogen derived from Gram-negative bacteria. In 1984, Phan et al.

(Phan & Fantone, 1984) reported that a single intraperitoneal endotoxin injection in

mice inhibits bleomycin-caused pulmonary fibrosis. In contrast, Brass et al. (2008)

have shown that ongoing but not single LPS challenge and bleomycin-induced

fibrosis share common patterns of gene expression. It was also reported that LPS

three-hit regiment induces acute lung injury (ALI) and rapid pulmonary fibrosis in

mice (Li et al., 2009). In addition, He et al. found that 72 h LPS administration

induced ALI and caused pulmonary fibrosis (He et al., 2009). All these data taken

together indicate that chronic but not acute exposure to LPS induces pulmonary

fibrosis and represents a new paradigm in understanding the etiology of this disease.

Based on these results, we hypothesized that chronic exposure to environmental

pyrogens in the lung may cause EMT and subsequent fibrosis via the inflammatory

cytokine milieu. To test this hypothesis in vitro, we asked whether LPS stimulation of

peripheral blood mononuclear cells (PBMC) is able to induce an inflammatory

response that can mediate EMT of lung epithelial type II cells (A549). Here we show

that indeed exposure of PBMC to endotoxin activates secretion of soluble mediators

that induce EMT of A549 cells and that IL-1β and TNF-α play a central role in

mediating the EMT process. EMT in epithelial cells was evidenced by acquisition of

fibroblast-like morphology, significant decrease in cytokeratin and increase in

vimentin marker expression.

Page 52: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

52

3. RESULTS

3.1. TGF-β1 induces EMT in A549 cells

In agreement with other investigators (Kasai et al., 2005), (Liu, 2008) our study

demonstrated that TGF-β1 induces EMT in A549 cells. The lung epithelial cells

cultured in the presence of the growth factor adopted fibroblast-like morphology and

reduced their cell-cell contacts (Figure 1A). EMT was also evidenced by decrease in

E-cadherin and cytokeratin epithelial markers and increase in vimentin mesenchymal

marker expression (Figure 1B).

Fig. 1: TGF-β1 induces EMT in A549 cells. A549 cells (5 x 104 in 1ml) were plated on 24

well plate in DMEM + 10% FCS. After 16 h, culture medium was replaced by serum-free

RPMI-1640 and cells were stimulated with PBS (control) or with 5 ng/ml TGF-β1 for 72 h. A)

Phase contrast microphotographs of A549 cells show changes in cell morphology upon

treatment (magnification 100 x). B) Cell lysates were immunoblotted for E-cadherin, pan-

cytokeratin, vimentin and β-actin. 20 µg total proteins were loaded on each lane.

Representative blots are shown from 2 independent experiments.

A B

0 5

TGF-β1 [ng/ml]

E-cadherin

cytokeratin

vimentin

β-actin

Control

TGF-β1

Page 53: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

53

3.2. A549 cells stimulated with LPS do not undergo EMT

Although A549 cells have been shown to express TLR-4, predominantly in the

intracellular compartment (Guillot et al., 2004), even 10 µg/ml endotoxin exposure

was not able to induce IL-1β, TNF-α, IL-6 and IL-8 secretion in serum-free conditions

(data not shown). Stimulation of epithelial cells with 10 ng/ml LPS for 72 h did not

alter cell morphology and the expression of the molecular markers examined was not

affected (Figure 2).

Fig. 2: LPS does not induce EMT in lung epithelial cells. A549 cells (5 x 104 in 1ml) were

plated on 24 well plate in DMEM + 10% FCS. After 16 h, culture medium was replaced by

serum-free RPMI-1640 and cells were stimulated with PBS (control) or with 10 ng/ml LPS for

72 h. (A) Cell morphology was assessed under phase contrast light microscopy

(magnification 100 x). (B) Equal amount of total proteins (20 µg) were loaded in each lane

and E-cadherin, vimentin and hsp 90 α/β marker expressions were analyzed by Western blot.

Representative blots are shown from 2 different experiments.

0 10

LPS [ng/ml]

E-cadherin

vimentin

hsp 90 α/β

A B

Control

LPS

Page 54: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

54

3.3. Conditioned supernatant from LPS-stimulated PBMC induces EMT in

A549 cells

Transfer of supernatant from LPS-stimulated PBMC to A549 cells resulted in EMT.

This event has been evidenced by changes in cell morphology and marker

expression (Figure 3). The typical epithelial morphology was replaced by fibroblast-

like and the cells dramatically decreased their cytokeratin expression and acquisition

of vimentin mesenchymal marker expression was observed.

Control LPS

Control LPS0.0

0.5

1.0

***

cyto

kera

tin

/ h

sp

90

αα αα/ ββ ββ

Control LPS0.0

2.5

5.0

7.5

***

vim

en

tin

/ / / /h

sp

90

αα αα/ ββ ββ

Fig. 3: Effect of conditioned supernatant from LPS-stimulated PBMC on EMT in A549.

1 x 106 in 1ml PBMC were seeded on 24 well plates. After 2 h mononuclear cells were not

stimulated with PBS (control) or stimulated with 10 ng/ml LPS for 20 h at 37°C. A549 cells (5

x 104 in 1ml) were plated on 24 well plate in DMEM + 10% FCS. After 16 h epithelial culture

medium was replaced by cell-free supernatants from untreated or LPS-stimulated PBMC and

A549 cells were incubated for 72 h at 37°C. A and B panels represent the morphology of

A549 cells, incubated in supernatant from (A) untreated PBMC or (B) LPS-treated PBMC

(magnification 10 x). C and D panels show respectively the Western blot analysis for

A B

C D

Page 55: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

55

cytokeratin and vimentin. Results are based on densitometric analysis of the ratio of each

marker protein to hsp 90 α/β. For each donor control values were set to 1. Data are pooled

from 3 independent experiments (9 different PBMC donors) and represent a mean ± SEM

(Wilcoxon matched pairs test).

3.4. LPS-induced EMT is not driven by TGF-β1

We tested whether TGF-β1 plays a role in PBMC mediated LPS-induced EMT in

A549 cells. TGF-β1 production was analyzed in the supernatant of control and LPS-

stimulated PBMC after 20 h and in supernatants after incubation on epithelial cells for

3 days. Neither PBMC nor epithelial cells secreted TGF-β1 (data not shown),

although 50 ng/ml IFN-γ was shown to activate TGF-β1 secretion in PBMC (control

(94.7 ± 24.3) vs. IFN-γ (1323.3 ± 145.2) pg/ml production of 6 different PBMC

donors).

To investigate when factor(s) inducing EMT in A549 cells were produced PBMC were

stimulated for shorter times - 2 h, 4 h, 6 h, 8 h, 10 h and 20 h before transfer to

epithelial cells (Figure 4). Microscopy showed that 2 h LPS treatment of the PBMC

already induced weak but detectable mesenchymal-like morphology in A549 after

transfer of the supernatant. This alteration from epithelial into mesenchymal

morphology became statistically significant after 4 h conditioning, accumulated over

20 h time of PBMC stimulation with LPS. These results were consistent with a

decrease in cytokeratin and acquisition of vimentin marker expression and indicate

an important role of early pro-inflammatory mediator(s), accumulating with time in the

medium of LPS-stimulated immune cells.

A

Control 2 h LPS 4 h LPS

Page 56: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

56

B

con 2 4 6 8 10 200.0

0.5

1.0

1.5

time [h]

**

cyto

kera

tin

/ h

sp

90

αα αα/ ββ ββ

con 2 4 6 8 10 200.0

2.5

5.0

7.5

10.0

vim

en

tin

/ h

sp

90

αα αα/ ββ ββ

time [h]

**

* *

**

Fig. 4: Early produced inflammatory cytokine(s) mediate EMT in lung epithelial

cells. 1 x 106 (1 ml) PBMC were seeded and after 2 h were stimulated with PBS or with 10

ng/ml LPS for 2 h, 4 h, 6 h, 8 h, 10 h and 20 h at 37°C. A549 cells (5 x 104 in 1ml) were

plated on 24 well plate in DMEM + 10% FCS. After overnight culture epithelial cells were

incubated with supernatants from untreated or LPS-stimulated PBMC after the different

incubation times for 72 h. Panel A: Morphology of A549 (magnification 50 x) incubated with

conditioned medium from stimulated with vehicle PBMC (control); PBMC stimulated for 2h

with LPS (2 h LPS); PBMC stimulated for 4h with LPS (4 h LPS). Panel B: Western blot

analysis for cytokeratin and vimentin. Results are based on densitometric analysis of the

ratio of each marker protein to hsp 90 α/β. Control values were set to 1. Data are pooled from

2 independent experiments (4 different PBMC donors) and represent a mean ± SEM

(Repeated Measures ANOVA and Dunnett’s post test on paired row data).

3.5. IL-1β and TNF-α induce EMT in A549 cells

The major cytokines present in the inflammatory milieu of chronic lung diseases are

IL-1β and TNF-α (Lappalainen et al., 2005), which are also known to be the first

cytokines secreted upon stimulation of monocytes with LPS (Jansky, Reymanova &

Kopecky, 2003). Therefore, A549 cells were incubated with TNF-α (3 ng/ml) and IL-

1β (4 ng/ml) for 72 h and effects on cell morphology and marker expression were

investigated (Figure 5). Both pro-inflammatory cytokines induced a change to a

fibroblast-like shape in epithelial cells, reduced their cytokeratin and increased their

vimentin expression. We have also observed in agreement with other investigators

(Jansky et al., 2003) that LPS stimulation of PBMC induces production of IL-6, IL-8,

macrophage chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1α

Page 57: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

57

(MIP-1α) and IL-10, shown also to be elevated in IPF and experimental models of

fibrosis (Agostini & Gurrieri, 2006), (Gauldie, Jordana & Cox, 1993; Kelly et al.,

2003). Direct stimulation of A549 cells with these inflammatory mediators did not alter

cell morphology and investigated marker expression (data not shown).

A

B

Control IL-1β TNF-α IL-1β + TNF-α

Fig. 5: IL-1β and TNF-α induce EMT in A549. A549 (5 x 104 /ml) were stimulated with

vehicle (control), 4 ng/ml IL-1β, 3 ng/ml TNF-α and combination of both cytokines for 72 h in

serum-free medium. A) Phase contrast microphotographs are taken with 100 x magnification.

Control IL-1β (4 ng/ml)

TNF-α (3 ng/ml) IL-1β (4 ng/ml) + TNF-α (3 ng/ml)

cytokeratin

vimentin

hsp 90 α/β

Page 58: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

58

B). 20 µg of total proteins were loaded on each lane and Western blotting for vimentin and

cytokeratin expression was performed. Data represent one of 3 independent experiments.

3.6. LPS-stimulated PBMC produce IL-1β and TNF-α, which induce EMT

in lung epithelial cells

To investigate the role of these two pro-inflammatory cytokines in the observed EMT

effect of epithelial cells upon conditioning we used neutralizing antibodies against IL-

1β and TNF-α. PBMC-conditioned supernatants diluted to 75% were pre-incubated in

the absence or presence of 1 µg/ml of αIL-1β and αTNF-α, and transferred onto A549

cells for an additional 3 days. In the presence of the antibodies cytokeratin

expression was increased and vimentin expression was decreased, compared to the

marker expression observed in the absence of neutralizing antibodies (Figure 6).

Fig. 6: Neutralization of IL-1β and TNF-α in conditioned supernatant from PBMC with

αIL-1β and αTNF-α. 1 x 106 in 1ml PBMC were seeded on 24 well plates. After 2 h

mononuclear cells were not stimulated or stimulated with 10 ng/ml LPS for 20 h at 37°C.

A549 cells (5 x 104/ml) were plated on 24 well plate in DMEM + 10% FCS. Cell-free

supernatants from PBS- and LPS-stimulated PBMC (75%) were pre-incubated for 2 h with

and without 2 µg/ml of αIL-1β, αTNF-α or a combination of both antibodies at 37°C. Then

conditioned medium was transferred onto A549 cells for 72 h. Results are based on

densitometric analysis of the ratio of each marker protein to hsp 90 α/β. Control values are

set to 1. Western blot data are pooled from 3 independent experiments (9 different PBMC

donors) and represent a mean ± SEM. *p<0.05; **p<0.01, ***p<0.001 all versus LPS values

(Friedman test followed by Dunn’s post test on paired row data).

0

1

2

3

control

LPS

LPS + αααα IL-1ββββ

LPS + ααααTNF-αααα

LPS + α α α α IL-1β + αβ + αβ + αβ + α TNF-αααα

**

ns

5

10

**

vim

en

tin

/ h

sp

90

αα αα/ ββ ββ

0.0

0.5

1.0

control

LPS

LPS + αααα IL-1ββββ

LPS + ααααTNF-αααα

LPS + α α α α IL-1β + αβ + αβ + αβ + αTNF-αααα5

10

***

ns*

cyto

kera

tin

/ h

sp

90

αα αα/ ββ ββ

Page 59: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

59

These data were consistent with results using IL-1ra and αmuTNF-α (Figure 7). The

ability of αmuTNF-α to neutralize human TNF-α activity was shown by inhibition of IL-

8 and IL-6 production in TNF-α stimulated A549 cells (data not shown).

0.0

2.5

5.0

control

LPS

LPS + IL1ra + ααααmuTNF-αααα

5

10

**

ns

vim

en

tin

/ h

sp

90

αα αα/ββ ββ

0

1

control

LPS

LPS + IL1ra + ααααmuTNF-αααα

5

10

cyto

kera

tin

/ h

sp

90

αα αα/ββ ββ

**

ns

Fig. 7: Inhibition of IL-1β and TNF-α in conditioned SN from PBMC with IL-1ra and

αmuTNF-α. 1 x 106 in 1ml PBMC were seeded on 24 well plates. After 2 h mononuclear cells

were not stimulated (con) or stimulated with 10 ng/ml LPS for 20 h at 37°C. A549 cells (5 x

104/ml) were plated on 24 well plate in DMEM + 10% FCS. Cell-free supernatants from PBS-

and LPS-stimulated PBMC (75%) were pre-incubated for 2 h in the presence or absence of a

combination of 10 µg/ml and αmuTNF-α (500 µg/ml total protein) at 37°C. Then conditioned

medium was transferred onto A549 cells for 72 h. Results are based on densitometric

analysis of the ratio of each marker protein to hsp 90 α/β. Control values are set to 1.

Western blot data are pooled from 3 independent experiments (4 different PBMC donors)

and represent a mean ± SEM. *p<0.05; **p<0.01, ***p<0.001 all versus LPS values

(Friedman test followed by Dunn’s post test on paired row data).

Page 60: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

60

4. DISCUSSION

Inflammation is believed to play an important role in the pathogenesis of chronic lung

diseases such as asthma, chronic obstructive pulmonary disease (COPD) and

idiopathic pulmonary fibrosis (IPF) (Araya & Nishimura, 2010). IPF is a devastating

disease with unknown etiology and a prognosis worse than that of many cancers.

Increasing numbers of studies show that environmental pollutants and oxidative

stress may cause IPF (Maher et al., 2007), (Rahman et al., 1999), (Taskar & Coultas,

2006).

Myofibroblasts, the main cell population that constitutes fibroblastic foci in IPF, are

considered central in the pathogenesis of this disease. Emerging evidence suggests

not only interstitial fibroblasts and blood-borne fibrocytes but also epithelial cells can

differentiate into myofibroblasts through EMT as source (Scotton & Chambers, 2007).

In vitro EMT has been induced in various epithelial cell lines in response to TGF-β1

stimulation (Kasai et al., 2005), (Willis et al., 2005), (Zhang et al., 2006). Kasai et al.

for a first time reported that A549 cells undergo EMT upon stimulation with 5 ng/ml

TGF-β1 and observed up-regulation of fibronectin, down-regulation of E-cadherin,

slight decrease in cytokeratin 19 and no change in vimentin expression (Kasai et al.,

2005). In our studies we confirmed that these cells undergo EMT upon treatment with

TGF-β1 and that A549 cells adopted fibroblast-like morphology, lost expression of

epithelial phenotypic markers (E-cadherin and cytokeratin) and accumulated strongly

mesenchymal vimentin expression. As a difference Kasai et al. maintained A549 cells

in serum-free DMEM containing 0.1% bovine serum albumin (BSA) for 24 h prior

stimulation with cytokines. From our own observations 0.1% BSA in DMEM medium

induced time-dependent cell death in resting A549 cells and probably a loss of

surface cell marker expression.

So far the role of LPS, the principal endotoxin of Gram-negative bacteria, in the

process of EMT has not been investigated. Recent papers (Li et al., 2009), (He et al.,

2009) indicate that repeated LPS administration induces ALI and pulmonary fibrosis

in mice. Few studies also suggested that inflammation links to fibrosis through the

process of EMT (Liu, 2008), (Lopez-Novoa & Nieto, 2009). Therefore we

hypothesized that in vitro LPS stimulation of immune cells, i.e. PBMC, may induce an

inflammatory cascade and result in EMT in A549 cells. In the current study we

demonstrate that 20 h conditioned supernatant from LPS-stimulated PBMC induces

Page 61: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

61

EMT in A549 cells. This was evidenced by alteration in cell shape from epithelial into

elongated mesenchymal morphology, significant down-regulation of cytokeratin and

significant up-regulation in vimentin expression. By measuring TGF-β1 content in

supernatant from control and LPS stimulated PBMC and in supernatants incubated

on A549 cells for 3 days we determined that EMT was not driven by TGF-β1 in this

setting. However, it could be shown that recombinant IFN-γ induces significant TGF-

β1 expression in PBMC. In agreement, Olas et al. published that in vitro LPS-

stimulated blood monocytes and PBMC are not a significant source of TGF-β1 (Olas

et al., 2005). Moreover, it was observed here that endotoxin activated PBMC do not

release IFN-γ, as also previously seen by Krakauer (Krakauer, 2002).

The nature of the inflammatory mediator(s) involved in the LPS-driven EMT process

was studied. We tested the effect of conditioned supernatants from PBMC stimulated

with LPS for shorter times. We were surprised to see initiation of cell elongation of

epithelial cells incubated with supernatant after only 2 h conditioning. 4 h conditioned

supernatant induced a clear fibroblast-like morphology. Western blot results were

consistent with cell morphology data and indicated time-dependent down-regulation

of cytokeratin and up-regulation in vimentin expression. This directed us to

investigate the early inflammatory cytokines IL-1β and TNF-α as possible mediators.

These cytokines have been suggested to play a role in IPF (Allen & Spiteri, 2002).

Liu et al., 2008 (Liu, 2008) have shown that a mixture of inflammatory cytokines (IL-

1β, TNF-α, IFN-γ) augments EMT in A549 induced by TGF-β1. Vesey et al. reported

that IL-1β induces EMT in kidney epithelial cells (Vesey et al., 2002).

Here, we were able to show that IL-1β and TNF-α, but not IL-6, IL-8, MCP-1, MIP-1α,

IL-10, convert A549 cell morphology, decrease cytokeratin and increase vimentin

marker expression. Surprisingly, no synergistic effect on cell morphology and marker

expression was observed when A549 cells were incubated with both cytokines.

The role of IL-1β and TNF-α in LPS-activated mononuclear cell supernatants on the

EMT in A549 cells was further examined using neutralizing antibodies or IL-1ra. In

these experimental settings, 75% of conditioned supernatants from PBS-treated and

LPS-treated PBMC were incubated in the presence or absence of 2 µg/ml αIL-1β,

αTNF-α or a combination of both neutralizing antibodies. Inhibition of IL-1β and TNF-

α activity was also achieved using 10 µg/ml IL-1ra, 500 µg/ml total protein of

αmuTNF-α or a combination of both. Addition of antibodies or IL-1ra to control PBMC

supernatants did not alter cell morphology or marker expression. The effect of

Page 62: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

62

neutralizing antibodies or neutralizing antibody and IL-1ra showed greater results

when applied in combination both on cell morphology, cytokeratin and vimentin

expression. Thus, our results indicate a central role of IL-1β and TNF-α in the LPS-

driven EMT process in A549 cells. Interestingly, clinical trials to prevent IPF with

etanercept, a TNF-α antagonist, recently reported a decreased rate of IPF disease

progression and good tolerance (Raghu et al., 2008). However, there have been no

clinical trials with anakinra, IL-1 receptor antagonist effective in reducing the signs

and symptoms of rheumatoid arthritis but leading to increased risk of serious

infections for the treatment of IPF (Salliot, Dougados & Gossec, 2009), (Agostini &

Gurrieri, 2006).

Taking these results further, it would be highly interesting to test weather chronic LPS

administration in vivo causes EMT and weather this process can be attenuated or

inhibited in the presence of neutralizing αIL-1β and αTNF-α antibodies or IL-1ra.

Taken together, our findings indicate that in vitro LPS stimulation of PBMC induces

production of pro-inflammatory cytokines IL-1β and TNF-α able to drive the process

of EMT in the epithelial cell line A549 in the absence of TGF-β1.

Page 63: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

63

5. MATERIAL AND METHODS

5.1. Reagents and antibodies

LPS from Salmonella abortus equi, SEB from Staphylococcus aureus, PGN from

Staphylococcus aureus and Brefeldin A were purchased from Sigma (Deisenhofen,

Germany). Further substances were IL-1β and TNF-α from NIBSC (Hertfordshire,

UK) and IL-1 receptor antagonist (IL-1ra) (Kineret® 100 mg of Anakinra in a syringe,

Biovitrum, Sweden). Dulbecco’s PBS, DMEM medium with high glucose and fetal calf

serum (FCS) were from PAA Laboratories (Pasching, Austria) and RPMI-1640 cell

culture medium was purchased from Bio Whittaker (Verviers, Belgium). Recombinant

human TGF-β1 and polyclonal goat anti-human antibodies against TNF-α (αTNF-α)

and IL-1β (αIL-1β) were purchased from R&D Systems (Minneapolis, MN). Polyclonal

sheep anti-mouse TNF-α antibody (αmuTNF-α) was produced in-house (Sommer et

al., 2001). Mouse monoclonal anti-human antibodies against pan-cytokeratin and

vimentin were from Sigma; polyclonal rabbit anti-human hsp 90 α/β antibody was

from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA); polyclonal anti-human

β-actin antibody was from Cell Signaling Technology, Inc. (Danvers, USA). Goat anti-

rabbit immunoglobulin-HRP and goat anti-mouse immunoglobulin-HRP were supplied

by Jackson ImmunoResearch Laboratories, Inc. (West Grove, PA, USA).

5.2. Cell line

Human lung adenocarcinoma epithelial cell line A549 was supplied from ATCC/LGC

Promochem (Wesel, Deutschland). Cells were maintained in high glucose DMEM

medium supplemented with 10% heat-inactivated FCS at 37°C in a humidified 5%

CO2 atmosphere. A549 cells were harvested every 2 to 3 days after reaching 90 to

95% confluence up to passage 20.

5.3. Isolation of human peripheral blood mononuclear cells

PBMC from healthy volunteers were prepared with CPTTM Cell Preparation Tubes

(BD Biosciences, Heidelberg, Germany) according to the manufacturer instructions.

After centrifugation (1600 x g for 20 min), the white layer above the gel containing the

Page 64: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

64

PBMC was removed and the cells were washed 3 times (300 x g for 7 minutes) with

RPMI-1640 containing 2.5 IU/ml heparin (Ratiopharm, Ulm, Germany). Cell counts

were determined with a Pentra 60 apparatus (ABX Diagnostics, Montpellier, France).

5.4. Enzyme-linked immunosorbent assay

Cytokine release was determined by in-house sandwich enzyme-linked

immunosorbent assay (ELISA) based on commercial antibody pairs against human

TNF-α, IL-8 (both Thermo Scientific, Germany), IL-1β and IL-6 (both R&D Systems,

Minneapolis, USA). Recombinant standards for IL-1β, IL-6 and TNF-α were obtained

from NIBSC, Herts, UK, for IL-8 from PeproTech. For TGF-β1 measurements

commercial DuoSet kit was purchased from R&D Systems (Minneapolis, MN).

Assays were carried out in flat-bottom, ultrasorbent 96 well plates (MaxiSorp, NUNC,

Wiesbaden, Germany). Binding of biotinylated secondary antibody was quantified

using streptavidin-conjugated horseradish peroxidase (Jackson Immuno Research,

West Grove, PA, USA) and the substrate TMB (3,3’,5,5’-tetramethylbenzidine,

Sigma). The reaction was stopped by adding 50 µl of 1 M H2SO4 and absorption was

measured at λ = 450/690 nm in an ELISA reader (Rainbow, Tecan, Crailsheim,

Germany).

5.5. A549-PBMC co-cultures

Co-culture experiments were performed in direct contact between epithelial cells and

PBMC (mixed co-cultures) and on transwell plates (transwell co-cultures). In mixed

co-cultures 2 x 105 A549 cells in 500 µl growth medium were seeded on 48 well

plates or 24 well plates and incubated at 37°C overnight for 16 h. Next day PBMC

were prepared as described above. Supernatant of the A549 culture was discarded

and 450 µl RPMI-1640 with heparin was added. 450 µl 5 x 105 cells PBMC were

added to the epithelial cells or seeded on the same plate (PBMC mono-culture). After

3 h, cells were stimulated with vehicle (PBS) or 100 ng/ml LPS, 100 ng/ml SEB or 1

µg/ml PGN. Cell-free supernatants were collected after 24 h and stored at -80°C until

ELISA measurements.

Page 65: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

65

In transwell experiments 500 µl 2 x 105 A549 cells were seeded in growth medium in

the lower compartment of 24 well plates (Corning, NY, USA). The transwell inserts

material had a polycarbonate membrane with 0.4 µm pore size (# 3413). After

overnight incubation, medium was removed and 200 µl RPMI-1640 with heparin was

added into each well of the lower compartment. 250 µl RPMI-1640 with heparin was

added into the upper compartment of the transwells when A549 were incubated

alone. For transwell co-cultures and PBMC mono-cultures 250 µl 5 x 105 PBMC were

added to the upper compartment. After 3 h cells in transwell plates were stimulated

with vehicle (PBS) or 100 ng/ml LPS (into the upper compartment) in a final volume

of 500 µl. Cell-free supernatants were collected after 24 h and stored at -80°C until

ELISA measurements.

5.6. Neutralization of IL-1β and TNF-α activity

Cell-free supernatants from stimulated with vehicle or LPS-stimulated PBMC were

diluted to 75% (v/v) in RPMI-1640 with heparin and incubated for 2 h at 37°C in the

presence or absence of 1) 2 µg/ml αIL-1β, 2 µg/ml αTNF-α or a combination of both

neutralizing antibodies; 2) neutralizing αmuTNF-α (500 µg/ml total protein

concentration) antibody, 10 µg/ml IL-1 receptor antagonist (IL-1ra) or a combination

of αmuTNF-α and IL-1ra, before being transferred onto A549 cells, which were then

cultured for 3 additional days.

5.7. Inhibition of TNF-α processing by Brefeldin A

A549 and PBMC mono-cultures and mixed co-culture were seeded on 48 well plates

and after 3 h were left untreated (PBS) or stimulated with 100 ng/ml LPS, in the

presence or absence of 5 µg/ml Brefeldin A (500 µl final volume). After 3 h

stimulation, plates were centrifuged; supernatants were collected and stored at -80°C

until TNF-α ELISA measurement. The cell pellets were resuspended in 500 µl PBS

and 3 x freeze-thaw cycles were performed. Cell-free supernatants were harvested

and intracellular pro-TNF-α was assessed by ELISA.

Page 66: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

66

5.8. LPS stimulation of PBMC in medium or A549 conditioned

supernatant

A549 cells (2 x 105 in 500 µl) were seeded in growth medium. After 16 h culture

medium was replaced by 500 µl RPMI-1640 with heparin. In parallel, 500 µl RPMI-

1640 with heparin was also added on the 48 well plates. After 2 h, 4 h, 8 h and 24 h

supernatants from both conditions were harvested and stored at -80°C. PBMC were

prepared according to the protocol described above. 70 µl 5 x 105 cells were added

to 380 µl of collected supernatants (76% v/v) and after 3 h stimulated with vehicle

(PBS) or 100 ng/ml LPS in a final volume of 500 µl. After 24 h TNF-α cytokine was

assessed by ELISA.

5.9. Total RNA extraction and cDNA synthesis

70 µl 5 x 105 PBMC were added to 380 µl 24 h cell-free conditioned supernatant from

A549 cells or RPMI-1640 with heparin, and after 3 h PBMC were stimulated with

vehicle (PBS) or 100 ng/ml LPS. After 3 h plates were centrifuged and supernatants

were removed. Cell pellet was lysed and RNA was extracted using QIAamp RNA

Blood Mini Kit (Qiagen, Hilden, Germany), according to the manufacturer instructions.

RNA samples were diluted in RNAase free water and the DNA was digested. 150 ng

of total RNA was reverse transcribed using oligo (dT)16 primers (Thermo Hybaid,

Ulm, Germany) and M-MuLV Reverse Transcriptase (New England Biolabs, Ipswich,

USA).

5.10. Quantitative Real-Time PCR

cDNA was quantified by Real-Time PCR via the LightCycler system (Roche) with

LightCycler FastStart DNA Master SYBR Green (Roche) using specific primers from

ThermoScientific (ThermoScientific, Hamburg, Germany) for human TNF-α: forward

5´-AGG CCC CAG TTT GAA TTC TT -3´and reverse: 5´-TCC TTC AGA CAC CCT

CAA CC -3´; for human GAPDH: forward 5´-GAA GGT GAA GGT CGG AGT C-3´and

reverse: 5´-GAA GAT GGT GAT GGG ATT TC-3´. The specificity of the PCR

Page 67: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

67

products was confirmed by melting curve analysis. The expression levels were

normalized to the housekeeper GAPDH.

5.11. Characterization of TNF-α anti-inflammatory compound

RPMI-1640 with heparin medium, incubated at 37°C for 24 h in the presence or

absence of A549 cells, was collected and treated as described:

a) Boiling at 95°C for 15 min;

b) 2 x freeze-thaw cycles;

c) Protein digestion with 0.5 mg/ml pronase (Roche, Mannheim, Germany) (37°C, 1

h) and subsequent enzyme inactivation (95°C, 15 min). Cell pellet was discarded

by centrifugation (3000 x g, 3 min);

d) Centrifuge filtration (molecular weight cut-off 10 kDa; 2000 x g) in Amicon Ultra-15

centrifugal filter unit (Millipore, Schwalbach, Germany).

70 µl 5 x 105 cells were added to 380 µl of supernatants (76% or 30% v/v) and after 3

h stimulated with vehicle or 100 ng/ml LPS in a final volume of 500 µl. After 24 h

TNF-α cytokine was assessed by ELISA.

5.12. SDS-PAGE and Western blot

A549 cells were washed once with Dulbecco’s PBS and lysed on ice with RIPA buffer

(50 mM Tris-HCl, pH 8, 150 mM NaCl, 1% TritonX 100, 0.5% Sodium Deoxycholate,

0.1% SDS) containing protease inhibitor cocktail (Sigma Aldrich, St Louis, USA).

After scraping, cell suspensions were collected and cleared by centrifugation at

13,000 x g for 30 min at 4°C. Total protein concentration was measured using the

BCA protein assay kit (Pierce) with bovine serum albumin (BSA) as standard protein

according to the manufacturer. Equal amounts of protein (20 µg) were loaded for

each lane on 12% Novex® Tris-Glycine gels (Invitrogen, Carlsbad, CA), followed by

electrophoresis and protein transfers to BioTraceTM NT nitrocellulose membrane (Pall

Corporation, Pensacola, FL) by semi-dry blotting. After the transfer, membranes were

blocked for 1 h at RT with 5% non-fat dry milk in TBS-T buffer (50 mM Tris-base, 150

mM NaCl, 0.1% Tween20, pH 7.5) and then probed with appropriate primary

Page 68: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

68

antibody, diluted in TBST with 5% BSA overnight at 4°C. After washing, the

membranes were probed for 1 h at RT with appropriate peroxidase-conjugated

secondary antibodies. After further extensive washing the immunoblots were

visualized by ECL substrate (2.5 mM Luminol, 0.4 mM para- coumaric acid in 10 mM

Tris, pH 8.5) and using LAS-3000 imaging system (Fuji).

5.13. Statistics

Statistical analyses and graphs were performed using the Graph Pad Prism software

(Graph Pad Software, San Diego, USA). All data are given as means ± SEM. P

values less then 0.05 were considered significant (*p ≤ 0.05, **p ≤ 0.01 and ***p ≤

0.001).

Page 69: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

69

IV. REFERENCES

ACLOQUE, H., ADAMS, M. S., FISHWICK, K., BRONNER-FRASER, M. & NIETO, M. A. (2009).

Epithelial-mesenchymal transitions: the importance of changing cell state in

development and disease. J Clin Invest 119, 1438-49.

AGOSTINI, C. & GURRIERI, C. (2006). Chemokine/cytokine cocktail in idiopathic

pulmonary fibrosis. Proc Am Thorac Soc 3, 357-63.

AHMED, S. & NAWSHAD, A. (2007). Complexity in interpretation of embryonic epithelial-

mesenchymal transition in response to transforming growth factor-beta

signaling. Cells Tissues Organs 185, 131-45.

AKIRA, S., TAKEDA, K. & KAISHO, T. (2001). Toll-like receptors: critical proteins linking

innate and acquired immunity. Nat Immunol 2, 675-80.

ALLEN, J. T. & SPITERI, M. A. (2002). Growth factors in idiopathic pulmonary fibrosis:

relative roles. Respir Res 3, 13.

ARAYA, J. & NISHIMURA, S. L. Fibrogenic reactions in lung disease. Annu Rev Pathol

5, 77-98.

ARAYA, J. & NISHIMURA, S. L. (2010). Fibrogenic reactions in lung disease. Annu Rev

Pathol 5, 77-98.

ARMSTRONG, L., MEDFORD, A. R., UPPINGTON, K. M., ROBERTSON, J., WITHERDEN, I. R.,

TETLEY, T. D. & MILLAR, A. B. (2004). Expression of functional toll-like receptor-

2 and -4 on alveolar epithelial cells. Am J Respir Cell Mol Biol 31, 241-5.

ASONG, J., WOLFERT, M. A., MAITI, K. K., MILLER, D. & BOONS, G. J. (2009). Binding

and Cellular Activation Studies Reveal That Toll-like Receptor 2 Can

Differentially Recognize Peptidoglycan from Gram-positive and Gram-negative

Bacteria. J Biol Chem 284, 8643-53.

Page 70: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

70

AZUMA, A., NUKIWA, T., TSUBOI, E., SUGA, M., ABE, S., NAKATA, K., TAGUCHI, Y., NAGAI,

S., ITOH, H., OHI, M., SATO, A. & KUDOH, S. (2005). Double-blind, placebo-

controlled trial of pirfenidone in patients with idiopathic pulmonary fibrosis. Am

J Respir Crit Care Med 171, 1040-7.

BADER, T. & NETTESHEIM, P. (1996). Tumor necrosis factor-alpha modulates the

expression of its p60 receptor and several cytokines in rat tracheal epithelial

cells. J Immunol 157, 3089-96.

BALS, R. & HIEMSTRA, P. S. (2004). Innate immunity in the lung: how epithelial cells

fight against respiratory pathogens. Eur Respir J 23, 327-33.

BARBOZA, C. E., WINTER, D. H., SEISCENTO, M., SANTOS UDE, P. & TERRA FILHO, M.

(2008). Tuberculosis and silicosis: epidemiology, diagnosis and

chemoprophylaxis. J Bras Pneumol 34, 959-66.

BARNES, P. J. (2005). Emerging targets for COPD therapy. Curr Drug Targets

Inflamm Allergy 4, 675-83.

BECKER, M. N., DIAMOND, G., VERGHESE, M. W. & RANDELL, S. H. (2000). CD14-

dependent lipopolysaccharide-induced beta-defensin-2 expression in human

tracheobronchial epithelium. J Biol Chem 275, 29731-6.

BEHR, J. (2007). Novel aspects of treatment for interstitial lung diseases. Diffuse

Parenchymal Lung Disease 36, 117-126.

BERTHIAUME, Y., LESUR, O. & DAGENAIS, A. (1999). Treatment of adult respiratory

distress syndrome: plea for rescue therapy of the alveolar epithelium. Thorax

54, 150-60.

BHASKARAN, M., KOLLIPUTI, N., WANG, Y., GOU, D., CHINTAGARI, N. R. & LIU, L. (2007).

Trans-differentiation of alveolar epithelial type II cells to type I cells involves

autocrine signaling by transforming growth factor beta 1 through the Smad

pathway. J Biol Chem 282, 3968-76.

Page 71: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

71

BINGISSER, R. M. & HOLT, P. G. (2001). Immunomodulating mechanisms in the lower

respiratory tract: nitric oxide mediated interactions between alveolar

macrophages, epithelial cells, and T-cells. Swiss Med Wkly 131, 171-9.

BRASS, D. M., YANG, I. V., KENNEDY, M. P., WHITEHEAD, G. S., RUTLEDGE, H., BURCH, L.

H. & SCHWARTZ, D. A. (2008). Fibroproliferation in LPS-induced airway

remodeling and bleomycin-induced fibrosis share common patterns of gene

expression. Immunogenetics 60, 353-69.

BRINGARDNER, B. D., BARAN, C. P., EUBANK, T. D. & MARSH, C. B. (2008). The role of

inflammation in the pathogenesis of idiopathic pulmonary fibrosis. Antioxid

Redox Signal 10, 287-301.

BURNS, K., MARTINON, F., ESSLINGER, C., PAHL, H., SCHNEIDER, P., BODMER, J. L., DI

MARCO, F., FRENCH, L. & TSCHOPP, J. (1998). MyD88, an adapter protein

involved in interleukin-1 signaling. J Biol Chem 273, 12203-9.

BURVALL, K., PALMBERG, L. & LARSSON, K. (2005). Expression of TNFalpha and its

receptors R1 and R2 in human alveolar epithelial cells exposed to organic dust

and the effects of 8-bromo-cAMP and protein kinase A modulation. Inflamm

Res 54, 281-8.

CAROFF, M., KARIBIAN, D., CAVAILLON, J. M. & HAEFFNER-CAVAILLON, N. (2002).

Structural and functional analyses of bacterial lipopolysaccharides. Microbes

Infect 4, 915-26.

CATES, C. J. & LASSERSON, T. J. (2009). Regular treatment with formoterol versus

regular treatment with salmeterol for chronic asthma: serious adverse events.

Cochrane Database Syst Rev, CD007695.

CHAMAILLARD, M., HASHIMOTO, M., HORIE, Y., MASUMOTO, J., QIU, S., SAAB, L., OGURA,

Y., KAWASAKI, A., FUKASE, K., KUSUMOTO, S., VALVANO, M. A., FOSTER, S. J.,

MAK, T. W., NUNEZ, G. & INOHARA, N. (2003). An essential role for NOD1 in

host recognition of bacterial peptidoglycan containing diaminopimelic acid. Nat

Immunol 4, 702-7.

Page 72: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

72

CHOI, Y. W., KOTZIN, B., HERRON, L., CALLAHAN, J., MARRACK, P. & KAPPLER, J. (1989).

Interaction of Staphylococcus aureus toxin "superantigens" with human T

cells. Proc Natl Acad Sci U S A 86, 8941-5.

COGGLE, J. E., LAMBERT, B. E. & MOORES, S. R. (1986). Radiation effects in the lung.

Environ Health Perspect 70, 261-91.

COULTER, K. R., WEWERS, M. D., LOWE, M. P. & KNOELL, D. L. (1999). Extracellular

regulation of interleukin (IL)-1beta through lung epithelial cells and defective

IL-1 type II receptor expression. Am J Respir Cell Mol Biol 20, 964-75.

CRAPO, J. D., BARRY, B. E., GEHR, P., BACHOFEN, M. & WEIBEL, E. R. (1982). Cell

number and cell characteristics of the normal human lung. Am Rev Respir Dis

125, 740-5.

CRESTANI, B., CORNILLET, P., DEHOUX, M., ROLLAND, C., GUENOUNOU, M. & AUBIER, M.

(1994). Alveolar type II epithelial cells produce interleukin-6 in vitro and in vivo.

Regulation by alveolar macrophage secretory products. J Clin Invest 94, 731-

40.

CUI A., Y. Q., SARRIA R., NAKAMURA S., GUZMAN J., COSTABEL U. (2009). N-

acetylcysteine inhibits TNF-a, sTNFR, and TGF-b 1 release by alveolar

macrophages in idiopathic pulmonary fibrosis in vitro. SARCOIDOSIS

VASCULITIS AND DIFFUSE LUNG DISEASES 26, 147-154.

DE VRIES, J., KESSELS, B. L. & DRENT, M. (2001). Quality of life of idiopathic

pulmonary fibrosis patients. Eur Respir J 17, 954-61.

DERYNCK, R. & ZHANG, Y. E. (2003). Smad-dependent and Smad-independent

pathways in TGF-beta family signalling. Nature 425, 577-84.

DIAMOND, G., LEGARDA, D. & RYAN, L. K. (2000). The innate immune response of the

respiratory epithelium. Immunol Rev 173, 27-38.

DINARELLO, C. A. (1996). Biologic basis for interleukin-1 in disease. Blood 87, 2095-

147.

Page 73: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

73

DOUGLAS, W. W., RYU, J. H., SWENSEN, S. J., OFFORD, K. P., SCHROEDER, D. R.,

CARON, G. M. & DEREMEE, R. A. (1998). Colchicine versus prednisone in the

treatment of idiopathic pulmonary fibrosis. A randomized prospective study.

Members of the Lung Study Group. Am J Respir Crit Care Med 158, 220-5.

DZIARSKI, R. & GUPTA, D. (2005). Peptidoglycan recognition in innate immunity. J

Endotoxin Res 11, 304-10.

EHRHARDT, C., FIEGEL, J., FUCHS, S., ABU-DAHAB, R., SCHAEFER, U. F., HANES, J. &

LEHR, C. M. (2002). Drug absorption by the respiratory mucosa: cell culture

models and particulate drug carriers. J Aerosol Med 15, 131-9.

EICKELBERG, O. (2001). Endless healing: TGF-beta, SMADs, and fibrosis. FEBS Lett

506, 11-4.

FEHRENBACH, H. (2001). Alveolar epithelial type II cell: defender of the alveolus

revisited. Respir Res 2, 33-46.

FLAHERTY, K. R., TOEWS, G. B., LYNCH, J. P., 3RD, KAZEROONI, E. A., GROSS, B. H.,

STRAWDERMAN, R. L., HARIHARAN, K., FLINT, A. & MARTINEZ, F. J. (2001).

Steroids in idiopathic pulmonary fibrosis: a prospective assessment of adverse

reactions, response to therapy, and survival. Am J Med 110, 278-82.

GALANOS, C., LUDERITZ, O., RIETSCHEL, E. T., WESTPHAL, O., BRADE, H., BRADE, L.,

FREUDENBERG, M., SCHADE, U., IMOTO, M., YOSHIMURA, H. & ET AL. (1985).

Synthetic and natural Escherichia coli free lipid A express identical endotoxic

activities. Eur J Biochem 148, 1-5.

GAULDIE, J., JORDANA, M. & COX, G. (1993). Cytokines and pulmonary fibrosis. Thorax

48, 931-5.

GAULDIE, J., KOLB, M., ASK, K., MARTIN, G., BONNIAUD, P. & WARBURTON, D. (2006).

Smad3 signaling involved in pulmonary fibrosis and emphysema. Proc Am

Thorac Soc 3, 696-702.

Page 74: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

74

GEHR, P., BACHOFEN, M. & WEIBEL, E. R. (1978). The normal human lung:

ultrastructure and morphometric estimation of diffusion capacity. Respir

Physiol 32, 121-40.

GIRARDIN, S., BONECA, I., VIALA, J., CHAMAILLARD, M., LABIGNE, A., THOMAS, G.,

PHILPOTT, D. & SANSONETTI, P. (2003). Nod2 is a general sensor of

peptidoglycan through muramyl dipeptide (MDP) detection. Journal of

Biological Chemistry 278, 8869-8872.

GOLDMANN, T., KAHLER, D., SCHULTZ, H., ABDULLAH, M., LANG, D. S., STELLMACHER, F.

& VOLLMER, E. (2009). On the significance of Surfactant Protein-A within the

human lungs. Diagn Pathol 4, 8.

GOMEZ, M. I. & PRINCE, A. (2008). Airway epithelial cell signaling in response to

bacterial pathogens. Pediatr Pulmonol 43, 11-9.

GRANDEL, U., HEYGSTER, D., SIBELIUS, U., FINK, L., SIGEL, S., SEEGER, W.,

GRIMMINGER, F. & HATTAR, K. (2009). Amplification of lipopolysaccharide-

induced cytokine synthesis in non-small cell lung cancer/neutrophil cocultures.

Mol Cancer Res 7, 1729-35.

GREENE, C. M. & MCELVANEY, N. G. (2005). Toll-like receptor expression and function

in airway epithelial cells. Arch Immunol Ther Exp (Warsz) 53, 418-27.

GUILLOT, L., MEDJANE, S., LE-BARILLEC, K., BALLOY, V., DANEL, C., CHIGNARD, M. & SI-

TAHAR, M. (2004). Response of human pulmonary epithelial cells to

lipopolysaccharide involves Toll-like receptor 4 (TLR4)-dependent signaling

pathways: evidence for an intracellular compartmentalization of TLR4. J Biol

Chem 279, 2712-8.

HASHIMOTO, N., JIN, H., LIU, T., CHENSUE, S. W. & PHAN, S. H. (2004). Bone marrow-

derived progenitor cells in pulmonary fibrosis. J Clin Invest 113, 243-52.

Page 75: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

75

HE, Z., ZHU, Y. & JIANG, H. (2009). Inhibiting toll-like receptor 4 signaling ameliorates

pulmonary fibrosis during acute lung injury induced by lipopolysaccharide: an

experimental study. Respir Res 10, 126.

HEGEDUS, C. M., SKIBOLA, C. F., WARNER, M., SKIBOLA, D. R., ALEXANDER, D., LIM, S.,

DANGLEBEN, N. L., ZHANG, L., CLARK, M., PFEIFFER, R. M., STEINMAUS, C.,

SMITH, A. H., SMITH, M. T. & MOORE, L. E. (2008). Decreased urinary beta-

defensin-1 expression as a biomarker of response to arsenic. Toxicol Sci 106,

74-82.

HENRIQUET, C., GOUGAT, C., COMBES, A., LAZENNEC, G. & MATHIEU, M. (2007).

Differential regulation of RANTES and IL-8 expression in lung

adenocarcinoma cells. Lung Cancer 56, 167-74.

HERSETH, J. I., REFSNES, M., LAG, M. & SCHWARZE, P. E. (2009). Role of IL-1 beta and

COX2 in silica-induced IL-6 release and loss of pneumocytes in co-cultures.

Toxicol In Vitro 23, 1342-53.

HERSETH, J. I., VOLDEN, V., SCHWARZE, P. E., LAG, M. & REFSNES, M. (2008). IL-1beta

differently involved in IL-8 and FGF-2 release in crystalline silica-treated lung

cell co-cultures. Part Fibre Toxicol 5, 16.

HIEMSTRA, P. S. (2006). Defensins and cathelicidins in inflammatory lung disease:

beyond antimicrobial activity. Biochem Soc Trans 34, 276-8.

IDRISS, H. T. & NAISMITH, J. H. (2000). TNF alpha and the TNF receptor superfamily:

structure-function relationship(s). Microsc Res Tech 50, 184-95.

JANG, B. C., LIM, K. J., PAIK, J. H., KWON, Y. K., SHIN, S. W., KIM, S. C., JUNG, T. Y.,

KWON, T. K., CHO, J. W., BAEK, W. K., KIM, S. P., SUH, M. H. & SUH, S. I. (2004).

Up-regulation of human beta-defensin 2 by interleukin-1beta in A549 cells:

involvement of PI3K, PKC, p38 MAPK, JNK, and NF-kappaB. Biochem

Biophys Res Commun 320, 1026-33.

Page 76: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

76

JANSKY, L., REYMANOVA, P. & KOPECKY, J. (2003). Dynamics of cytokine production in

human peripheral blood mononuclear cells stimulated by LPS or infected by

Borrelia. Physiol Res 52, 593-8.

JIANG, Z., KUNIMOTO, M. & PATEL, J. A. (1998). Autocrine regulation and experimental

modulation of interleukin-6 expression by human pulmonary epithelial cells

infected with respiratory syncytial virus. J Virol 72, 2496-9.

JOHNSTON, L. C., GONZALES, L. W., LIGHTFOOT, R. T., GUTTENTAG, S. H. &

ISCHIROPOULOS, H. Opposing regulation of human alveolar type II cell

differentiation by nitric oxide and hyperoxia. Pediatr Res 67, 521-5.

KALLURI, R. (2009). EMT: when epithelial cells decide to become mesenchymal-like

cells. J Clin Invest 119, 1417-9.

KALLURI, R. & NEILSON, E. G. (2003). Epithelial-mesenchymal transition and its

implications for fibrosis. J Clin Invest 112, 1776-84.

KALLURI, R. & WEINBERG, R. A. (2009). The basics of epithelial-mesenchymal

transition. J Clin Invest 119, 1420-8.

KASAI, H., ALLEN, J. T., MASON, R. M., KAMIMURA, T. & ZHANG, Z. (2005). TGF-beta1

induces human alveolar epithelial to mesenchymal cell transition (EMT).

Respir Res 6, 56.

KELLEY, J. (1990). Cytokines of the lung. Am Rev Respir Dis 141, 765-88.

KELLY, M., KOLB, M., BONNIAUD, P. & GAULDIE, J. (2003). Re-evaluation of fibrogenic

cytokines in lung fibrosis. Curr Pharm Des 9, 39-49.

KIM, K. K., KUGLER, M. C., WOLTERS, P. J., ROBILLARD, L., GALVEZ, M. G., BRUMWELL,

A. N., SHEPPARD, D. & CHAPMAN, H. A. (2006). Alveolar epithelial cell

mesenchymal transition develops in vivo during pulmonary fibrosis and is

regulated by the extracellular matrix. Proc Natl Acad Sci U S A 103, 13180-5.

Page 77: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

77

KISSELEVA, T. & BRENNER, D. A. (2008a). Fibrogenesis of parenchymal organs. Proc

Am Thorac Soc 5, 338-42.

KISSELEVA, T. & BRENNER, D. A. (2008b). Mechanisms of fibrogenesis. Exp Biol Med

(Maywood) 233, 109-22.

KNAPP, S. (in press). LPS and bacterial lung inflammation models. Elsevier Ltd.

KOLB, M., MARGETTS, P. J., ANTHONY, D. C., PITOSSI, F. & GAULDIE, J. (2001).

Transient expression of IL-1beta induces acute lung injury and chronic repair

leading to pulmonary fibrosis. J Clin Invest 107, 1529-36.

KONIGSHOFF, M. & EICKELBERG, O. (in press). WNT signaling in lung disease: a failure

or a regeneration signal? Am J Respir Cell Mol Biol 42, 21-31.

KOTLOFF, R. M., LITTLE, J. & ELIAS, J. A. (1990). Human alveolar macrophage and

blood monocyte interleukin-6 production. Am J Respir Cell Mol Biol 3, 497-

505.

KRAKAUER, T. (1999). Immune response to staphylococcal superantigens. Immunol

Res 20, 163-73.

KRAKAUER, T. (2001). Suppression of endotoxin- and staphylococcal exotoxin-

induced cytokines and chemokines by a phospholipase C inhibitor in human

peripheral blood mononuclear cells. Clin Diagn Lab Immunol 8, 449-53.

KRAKAUER, T. (2002). Stimulant-dependent modulation of cytokines and chemokines

by airway epithelial cells: cross talk between pulmonary epithelial and

peripheral blood mononuclear cells. Clin Diagn Lab Immunol 9, 126-31.

LAMONTAGNE, F., BRIEL, M., GUYATT, G. H., COOK, D. J., BHATNAGAR, N. & MEADE, M.

(2009). Corticosteroid therapy for acute lung injury, acute respiratory distress

syndrome, and severe pneumonia: A meta-analysis of randomized controlled

trials. J Crit Care.

Page 78: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

78

LAPPALAINEN, U., WHITSETT, J. A., WERT, S. E., TICHELAAR, J. W. & BRY, K. (2005).

Interleukin-1beta causes pulmonary inflammation, emphysema, and airway

remodeling in the adult murine lung. Am J Respir Cell Mol Biol 32, 311-8.

LARRICK, J. W. & KUNKEL, S. L. (1988). The role of tumor necrosis factor and

interleukin 1 in the immunoinflammatory response. Pharm Res 5, 129-39.

LEEMANS, J. C., VERVOORDELDONK, M. J., FLORQUIN, S., VAN KESSEL, K. P. & VAN DER

POLL, T. (2002). Differential role of interleukin-6 in lung inflammation induced

by lipoteichoic acid and peptidoglycan from Staphylococcus aureus. Am J

Respir Crit Care Med 165, 1445-50.

LEMAITRE, B., NICOLAS, E., MICHAUT, L., REICHHART, J. M. & HOFFMANN, J. A. (1996).

The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the

potent antifungal response in Drosophila adults. Cell 86, 973-83.

LI, H., DU, S., YANG, L., CHEN, Y., HUANG, W., ZHANG, R., CUI, Y., YANG, J., CHEN, D.,

LI, Y., ZHANG, S., ZHOU, J., WEI, Z. & YAO, Z. (2009). Rapid pulmonary fibrosis

induced by acute lung injury via a lipopolysaccharide three-hit regimen. Innate

Immun 15, 143-54.

LIU, X. (2008). Inflammatory cytokines augments TGF-beta1-induced epithelial-

mesenchymal transition in A549 cells by up-regulating TbetaR-I. Cell Motil

Cytoskeleton 65, 935-44.

LIU, X. & LUO, F. (2006). Ghrelin attenuates LPS-induced acute lung injury via a

mechanism that involves the NO pathway. American College of Chest

Physicians.

LOPEZ-NOVOA, J. M. & NIETO, M. A. (2009). Inflammation and EMT: an alliance

towards organ fibrosis and cancer progression. EMBO Mol Med 1, 303-14.

LOSA GARCIA, J. E., RODRIGUEZ, F. M., MARTIN DE CABO, M. R., GARCIA SALGADO, M. J.,

LOSADA, J. P., VILLARON, L. G., LOPEZ, A. J. & ARELLANO, J. L. (1999).

Page 79: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

79

Evaluation of inflammatory cytokine secretion by human alveolar

macrophages. Mediators Inflamm 8, 43-51.

MACKICHAN, M. L. (2005). Toll bridge to immunity. Immune molecules hold promise

for vaccine adjuvant discovery. IAVI Rep 9, 1-5.

MAHER, T. M., WELLS, A. U. & LAURENT, G. J. (2007). Idiopathic pulmonary fibrosis:

multiple causes and multiple mechanisms? Eur Respir J 30, 835-9.

MAPEL, D. W., SAMET, J. M. & COULTAS, D. B. (1996). Corticosteroids and the

treatment of idiopathic pulmonary fibrosis. Past, present, and future. Chest

110, 1058-67.

MARGADANT, C. & SONNENBERG, A. (in press). Integrin-TGF-beta crosstalk in fibrosis,

cancer and wound healing. EMBO Rep 11, 97-105.

MARTIN, T. R. & FREVERT, C. W. (2005). Innate immunity in the lungs. Proc Am Thorac

Soc 2, 403-11.

MASON, D. P., BRIZZIO, M. E., ALSTER, J. M., MCNEILL, A. M., MURTHY, S. C., BUDEV, M.

M., MEHTA, A. C., MINAI, O. A., PETTERSSON, G. B. & BLACKSTONE, E. H. (2007).

Lung transplantation for idiopathic pulmonary fibrosis. Ann Thorac Surg 84,

1121-8.

MASSAGUE, J. & GOMIS, R. R. (2006). The logic of TGFbeta signaling. FEBS Lett 580,

2811-20.

MATTIX, M. E., HUNT, R. E., WILHELMSEN, C. L., JOHNSON, A. J. & BAZE, W. B. (1995).

Aerosolized staphylococcal enterotoxin B-induced pulmonary lesions in rhesus

monkeys (Macaca mulatta). Toxicol Pathol 23, 262-8.

MAYER, A. K., BARTZ, H., FEY, F., SCHMIDT, L. M. & DALPKE, A. H. (2008). Airway

epithelial cells modify immune responses by inducing an anti-inflammatory

microenvironment. Eur J Immunol 38, 1689-99.

Page 80: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

80

MAYER, A. K. & DALPKE, A. H. (2007). Regulation of local immunity by airway epithelial

cells. Arch Immunol Ther Exp (Warsz) 55, 353-62.

MBAWUIKE, I. N. & HERSCOWITZ, H. B. (1989). MH-S, a murine alveolar macrophage

cell line: morphological, cytochemical, and functional characteristics. J Leukoc

Biol 46, 119-27.

MCCORMACK, F. X. & WHITSETT, J. A. (2002). The pulmonary collectins, SP-A and SP-

D, orchestrate innate immunity in the lung. J Clin Invest 109, 707-12.

MEDZHITOV, R., PRESTON-HURLBURT, P. & JANEWAY, C. A., JR. (1997). A human

homologue of the Drosophila Toll protein signals activation of adaptive

immunity. Nature 388, 394-7.

MELTZER, E. B. & NOBLE, P. W. (2008). Idiopathic pulmonary fibrosis. Orphanet J Rare

Dis 3, 8.

MIZGERD, J. P. (2006). Lung infection--a public health priority. PLoS Med 3, e76.

MOGENSEN, T. H. (2009). Pathogen recognition and inflammatory signaling in innate

immune defenses. Clin Microbiol Rev 22, 240-73, Table of Contents.

MOVSAS, B., RAFFIN, T. A., EPSTEIN, A. H. & LINK, C. J., JR. (1997). Pulmonary

radiation injury. Chest 111, 1061-76.

MUIR, A., SOONG, G., SOKOL, S., REDDY, B., GOMEZ, M. I., VAN HEECKEREN, A. &

PRINCE, A. (2004). Toll-like receptors in normal and cystic fibrosis airway

epithelial cells. Am J Respir Cell Mol Biol 30, 777-83.

NATHAN, S. D. (2005). Lung transplantation: disease-specific considerations for

referral. Chest 127, 1006-16.

O'NEILL, L. A. & BOWIE, A. G. (2007). The family of five: TIR-domain-containing

adaptors in Toll-like receptor signalling. Nat Rev Immunol 7, 353-64.

Page 81: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

81

OIKONOMOU, N., HAROKOPOS, V., ZALEVSKY, J., VALAVANIS, C., KOTANIDOU, A.,

SZYMKOWSKI, D. E., KOLLIAS, G. & AIDINIS, V. (2006). Soluble TNF mediates the

transition from pulmonary inflammation to fibrosis. PLoS One 1, e108.

OLAS, K., BUTTERWECK, H., TESCHNER, W., SCHWARZ, H. P. & REIPERT, B. (2005).

Immunomodulatory properties of human serum immunoglobulin A: anti-

inflammatory and pro-inflammatory activities in human monocytes and

peripheral blood mononuclear cells. Clin Exp Immunol 140, 478-90.

PALECANDA, A., PAULAUSKIS, J., AL-MUTAIRI, E., IMRICH, A., QIN, G., SUZUKI, H.,

KODAMA, T., TRYGGVASON, K., KOZIEL, H. & KOBZIK, L. (1999). Role of the

scavenger receptor MARCO in alveolar macrophage binding of unopsonized

environmental particles. J Exp Med 189, 1497-506.

PAZGIER, M., HOOVER, D. M., YANG, D., LU, W. & LUBKOWSKI, J. (2006). Human beta-

defensins. Cell Mol Life Sci 63, 1294-313.

PHAN, S. H. (2002). The myofibroblast in pulmonary fibrosis. Chest 122, 286S-289S.

PHAN, S. H. & FANTONE, J. C. (1984). Inhibition of bleomycin-induced pulmonary

fibrosis by lipopolysaccharide. Lab Invest 50, 587-91.

PLATZ, J., BEISSWENGER, C., DALPKE, A., KOCZULLA, R., PINKENBURG, O., VOGELMEIER,

C. & BALS, R. (2004). Microbial DNA induces a host defense reaction of

human respiratory epithelial cells. J Immunol 173, 1219-23.

POLTORAK, A., HE, X., SMIRNOVA, I., LIU, M. Y., VAN HUFFEL, C., DU, X., BIRDWELL, D.,

ALEJOS, E., SILVA, M., GALANOS, C., FREUDENBERG, M., RICCIARDI-CASTAGNOLI,

P., LAYTON, B. & BEUTLER, B. (1998). Defective LPS signaling in C3H/HeJ and

C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282, 2085-8.

PORRECA, E., SERGI, R., BACCANTE, G., REALE, M., ORSINI, L., FEBBO, C. D., CASELLI,

G., CUCCURULLO, F. & BERTINI, R. (1999). Peripheral blood mononuclear cell

production of interleukin-8 and IL-8-dependent neutrophil function in

hypercholesterolemic patients. Atherosclerosis 146, 345-50.

Page 82: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

82

QUAN, T. E., COWPER, S., WU, S. P., BOCKENSTEDT, L. K. & BUCALA, R. (2004).

Circulating fibrocytes: collagen-secreting cells of the peripheral blood. Int J

Biochem Cell Biol 36, 598-606.

RADBRUCH, A., MECHTOLD, B., THIEL, A., MILTENYI, S. & PFLUGER, E. (1994). High-

gradient magnetic cell sorting. Methods Cell Biol 42 Pt B, 387-403.

RAGHU, G., BROWN, K. K., COSTABEL, U., COTTIN, V., DU BOIS, R. M., LASKY, J. A.,

THOMEER, M., UTZ, J. P., KHANDKER, R. K., MCDERMOTT, L. & FATENEJAD, S.

(2008). Treatment of idiopathic pulmonary fibrosis with etanercept: an

exploratory, placebo-controlled trial. Am J Respir Crit Care Med 178, 948-55.

RAGHU, G., DEPASO, W. J., CAIN, K., HAMMAR, S. P., WETZEL, C. E., DREIS, D. F.,

HUTCHINSON, J., PARDEE, N. E. & WINTERBAUER, R. H. (1991). Azathioprine

combined with prednisone in the treatment of idiopathic pulmonary fibrosis: a

prospective double-blind, randomized, placebo-controlled clinical trial. Am Rev

Respir Dis 144, 291-6.

RAGHU, G., JOHNSON, W. C., LOCKHART, D. & MAGETO, Y. (1999). Treatment of

idiopathic pulmonary fibrosis with a new antifibrotic agent, pirfenidone: results

of a prospective, open-label Phase II study. Am J Respir Crit Care Med 159,

1061-9.

RAHMAN, I., SKWARSKA, E., HENRY, M., DAVIS, M., O'CONNOR, C. M., FITZGERALD, M.

X., GREENING, A. & MACNEE, W. (1999). Systemic and pulmonary oxidative

stress in idiopathic pulmonary fibrosis. Free Radic Biol Med 27, 60-8.

RAMOS, C., MONTANO, M., GARCIA-ALVAREZ, J., RUIZ, V., UHAL, B. D., SELMAN, M. &

PARDO, A. (2001). Fibroblasts from idiopathic pulmonary fibrosis and normal

lungs differ in growth rate, apoptosis, and tissue inhibitor of metalloproteinases

expression. Am J Respir Cell Mol Biol 24, 591-8.

RAZ, E. (2007). Organ-specific regulation of innate immunity. Nat Immunol 8, 3-4.

Page 83: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

83

RESNICK, D., FREEDMAN, N. J., XU, S. & KRIEGER, M. (1993). Secreted extracellular

domains of macrophage scavenger receptors form elongated trimers which

specifically bind crocidolite asbestos. J Biol Chem 268, 3538-45.

REUTERSHAN, J. & LEY, K. (2004). Bench-to-bedside review: acute respiratory distress

syndrome - how neutrophils migrate into the lung. Crit Care 8, 453-61.

RICHELDI, L., DAVIES, H. R., FERRARA, G. & FRANCO, F. (2003). Corticosteroids for

idiopathic pulmonary fibrosis. Cochrane Database Syst Rev, CD002880.

RIETSCHEL, E. T., BRADE, H., HOLST, O., BRADE, L., MULLER-LOENNIES, S., MAMAT, U.,

ZAHRINGER, U., BECKMANN, F., SEYDEL, U., BRANDENBURG, K., ULMER, A. J.,

MATTERN, T., HEINE, H., SCHLETTER, J., LOPPNOW, H., SCHONBECK, U., FLAD, H.

D., HAUSCHILDT, S., SCHADE, U. F., DI PADOVA, F., KUSUMOTO, S. & SCHUMANN,

R. R. (1996). Bacterial endotoxin: Chemical constitution, biological recognition,

host response, and immunological detoxification. Curr Top Microbiol Immunol

216, 39-81.

ROBLEDO, R. & MOSSMAN, B. (1999). Cellular and molecular mechanisms of asbestos-

induced fibrosis. J Cell Physiol 180, 158-66.

RODRIGO, G. J. (2006). Rapid effects of inhaled corticosteroids in acute asthma: an

evidence-based evaluation. Chest 130, 1301-11.

ROGERS, D. F. & DONNELLY, L. E. (2001). Human airway inflammation: Sampling

techniques and analytical protocols. Humana.

SALLIOT, C., DOUGADOS, M. & GOSSEC, L. (2009). Risk of serious infections during

rituximab, abatacept and anakinra treatments for rheumatoid arthritis: meta-

analyses of randomised placebo-controlled trials. Ann Rheum Dis 68, 25-32.

SCHULZ, C., FARKAS, L., WOLF, K., KRATZEL, K., EISSNER, G. & PFEIFER, M. (2002).

Differences in LPS-induced activation of bronchial epithelial cells (BEAS-2B)

and type II-like pneumocytes (A-549). Scand J Immunol 56, 294-302.

Page 84: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

84

SCOTTON, C. J. & CHAMBERS, R. C. (2007). Molecular targets in pulmonary fibrosis:

the myofibroblast in focus. Chest 132, 1311-21.

SELMAN, M., KING, T. E. & PARDO, A. (2001). Idiopathic pulmonary fibrosis: prevailing

and evolving hypotheses about its pathogenesis and implications for therapy.

Ann Intern Med 134, 136-51.

SELMAN, M. & PARDO, A. (2002). Idiopathic pulmonary fibrosis: an

epithelial/fibroblastic cross-talk disorder. Respir Res 3, 3.

SELMAN, M. & PARDO, A. (2006). Role of epithelial cells in idiopathic pulmonary

fibrosis: from innocent targets to serial killers. Proc Am Thorac Soc 3, 364-72.

SEMPLE, F., WEBB, S., LI, H. N., PATEL, H. B., PERRETTI, M., JACKSON, I. J., GRAY, M.,

DAVIDSON, D. J. & DORIN, J. R. Human beta-defensin 3 has

immunosuppressive activity in vitro and in vivo. Eur J Immunol 40, 1073-8.

SIMS, J. E., GIRI, J. G. & DOWER, S. K. (1994). The two interleukin-1 receptors play

different roles in IL-1 actions. Clin Immunol Immunopathol 72, 9-14.

SOMMER, C., LINDENLAUB, T., TEUTEBERG, P., SCHAFERS, M., HARTUNG, T. & TOYKA, K.

V. (2001). Anti-TNF-neutralizing antibodies reduce pain-related behavior in

two different mouse models of painful mononeuropathy. Brain Res 913, 86-9.

STANDIFORD, T. J., KUNKEL, S. L., BASHA, M. A., CHENSUE, S. W., LYNCH, J. P., 3RD,

TOEWS, G. B., WESTWICK, J. & STRIETER, R. M. (1990). Interleukin-8 gene

expression by a pulmonary epithelial cell line. A model for cytokine networks in

the lung. J Clin Invest 86, 1945-53.

STEIMER, A., HALTNER, E. & LEHR, C. M. (2005). Cell culture models of the respiratory

tract relevant to pulmonary drug delivery. J Aerosol Med 18, 137-82.

STENGEL, D., ANTONUCCI, M., ARBORATI, M., HOURTON, D., GRIGLIO, S., CHAPMAN, M. J.

& NINIO, E. (1997). Expression of the PAF receptor in human monocyte-

derived macrophages is downregulated by oxidized LDL: relevance to the

Page 85: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

85

inflammatory phase of atherogenesis. Arterioscler Thromb Vasc Biol 17, 954-

62.

STOCKLEY, R. A. (1998). Lung infections. 1. Role of bacteria in the pathogenesis and

progression of acute and chronic lung infection. Thorax 53, 58-62.

STRIETER, R. M., BELPERIO, J. A. & KEANE, M. P. (2002). Cytokines in innate host

defense in the lung. J Clin Invest 109, 699-705.

STRIETER, R. M., CHENSUE, S. W., BASHA, M. A., STANDIFORD, T. J., LYNCH, J. P.,

BAGGIOLINI, M. & KUNKEL, S. L. (1990). Human alveolar macrophage gene

expression of interleukin-8 by tumor necrosis factor-alpha, lipopolysaccharide,

and interleukin-1 beta. Am J Respir Cell Mol Biol 2, 321-6.

STRIETER, R. M. & MEHRAD, B. (2009). New mechanisms of pulmonary fibrosis. Chest

136, 1364-70.

STRIETER, R. M., REMICK, D. G., LYNCH, J. P., 3RD, GENORD, M., RAIFORD, C.,

SPENGLER, R. & KUNKEL, S. L. (1989). Differential regulation of tumor necrosis

factor-alpha in human alveolar macrophages and peripheral blood monocytes:

a cellular and molecular analysis. Am J Respir Cell Mol Biol 1, 57-63.

TABETA, K., GEORGEL, P., JANSSEN, E., DU, X., HOEBE, K., CROZAT, K., MUDD, S.,

SHAMEL, L., SOVATH, S., GOODE, J., ALEXOPOULOU, L., FLAVELL, R. A. &

BEUTLER, B. (2004). Toll-like receptors 9 and 3 as essential components of

innate immune defense against mouse cytomegalovirus infection. Proc Natl

Acad Sci U S A 101, 3516-21.

TAKEDA, K. & AKIRA, S. (2005). Toll-like receptors in innate immunity. Int Immunol 17,

1-14.

TANAMOTO, K., ZAHRINGER, U., MCKENZIE, G. R., GALANOS, C., RIETSCHEL, E. T.,

LUDERITZ, O., KUSUMOTO, S. & SHIBA, T. (1984). Biological activities of

synthetic lipid A analogs: pyrogenicity, lethal toxicity, anticomplement activity,

Page 86: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

86

and induction of gelation of Limulus amoebocyte lysate. Infect Immun 44, 421-

6.

TANG, B. M., CRAIG, J. C., ESLICK, G. D., SEPPELT, I. & MCLEAN, A. S. (2009). Use of

corticosteroids in acute lung injury and acute respiratory distress syndrome: a

systematic review and meta-analysis. Crit Care Med 37, 1594-603.

TANG, P., HUNG, M. C. & KLOSTERGAARD, J. (1996). Human pro-tumor necrosis factor

is a homotrimer. Biochemistry 35, 8216-25.

TASKAR, V. S. & COULTAS, D. B. (2006). Is idiopathic pulmonary fibrosis an

environmental disease? Proc Am Thorac Soc 3, 293-8.

TECLE, T., TRIPATHI, S. & HARTSHORN, K. L. Defensins and cathelicidins in lung

immunity. Innate Immun.

TECLE, T., TRIPATHI, S. & HARTSHORN, K. L. (in press). Defensins and cathelicidins in

lung immunity. Innate Immun.

THIVIERGE, M. & ROLA-PLESZCZYNSKI, M. (1995). Up-regulation of inducible

cyclooxygenase gene expression by platelet-activating factor in activated rat

alveolar macrophages. J Immunol 154, 6593-9.

TOEWS, G. B. (2001). Cytokines and the lung. Eur Respir J Suppl 34, 3s-17s.

TUNZI, C. R., HARPER, P. A., BAR-OZ, B., VALORE, E. V., SEMPLE, J. L., WATSON-

MACDONELL, J., GANZ, T. & ITO, S. (2000). Beta-defensin expression in human

mammary gland epithelia. Pediatr Res 48, 30-5.

UEHARA, A., FUJIMOTO, Y., FUKASE, K. & TAKADA, H. (2007). Various human epithelial

cells express functional Toll-like receptors, NOD1 and NOD2 to produce anti-

microbial peptides, but not proinflammatory cytokines. Mol Immunol 44, 3100-

11.

ULICH, T. R., YIN, S., GUO, K., YI, E. S., REMICK, D. & DEL CASTILLO, J. (1991).

Intratracheal injection of endotoxin and cytokines. II. Interleukin-6 and

Page 87: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

87

transforming growth factor beta inhibit acute inflammation. Am J Pathol 138,

1097-101.

VALORE, E. V., PARK, C. H., QUAYLE, A. J., WILES, K. R., MCCRAY, P. B., JR. & GANZ, T.

(1998). Human beta-defensin-1: an antimicrobial peptide of urogenital tissues.

J Clin Invest 101, 1633-42.

VERSTREPEN, L., BEKAERT, T., CHAU, T. L., TAVERNIER, J., CHARIOT, A. & BEYAERT, R.

(2008). TLR-4, IL-1R and TNF-R signaling to NF-kappaB: variations on a

common theme. Cell Mol Life Sci 65, 2964-78.

VESEY, D., CHEUNG, C., CUTTLE, L., ENDRE, Z., GOBE, G. & JOHNSON, D. (2002).

Interleukin-1β induces human proximal tubule cell injury, α-smooth muscle

actin expression and fibronectin production. Kidney Int 2002 62, 31-40.

WEIDEL, W. & PELZER, H. (1964). Bagshaped Macromolecules--a New Outlook on

Bacterial Cell Walls. Adv Enzymol Relat Areas Mol Biol 26, 193-232.

WEWERS, M. D. & HERZYK, D. J. (1989). Alveolar macrophages differ from blood

monocytes in human IL-1 beta release. Quantitation by enzyme-linked

immunoassay. J Immunol 143, 1635-41.

WILLIS, B. C., DUBOIS, R. M. & BOROK, Z. (2006). Epithelial origin of myofibroblasts

during fibrosis in the lung. Proc Am Thorac Soc 3, 377-82.

WILLIS, B. C., LIEBLER, J. M., LUBY-PHELPS, K., NICHOLSON, A. G., CRANDALL, E. D., DU

BOIS, R. M. & BOROK, Z. (2005). Induction of epithelial-mesenchymal transition

in alveolar epithelial cells by transforming growth factor-beta1: potential role in

idiopathic pulmonary fibrosis. Am J Pathol 166, 1321-32.

WILSON, M. S. & WYNN, T. A. (2009). Pulmonary fibrosis: pathogenesis, etiology and

regulation. Mucosal Immunol 2, 103-21.

WRIGHT, S. D., RAMOS, R. A., TOBIAS, P. S., ULEVITCH, R. J. & MATHISON, J. C. (1990).

CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding

protein. Science 249, 1431-3.

Page 88: Role of lipopolysaccharide in pulmonary inflammation and ... · Role of lipopolysaccharide in pulmonary inflammation and fibrosis – an in vitro study Dissertation zur Erlangung

88

WU, Z., YANG, L., CAI, L., ZHANG, M., CHENG, X., YANG, X. & XU, J. (2007). Detection of

epithelial to mesenchymal transition in airways of a bleomycin induced

pulmonary fibrosis model derived from an alpha-smooth muscle actin-Cre

transgenic mouse. Respir Res 8, 1.

WYNN, T. A. (2007). Common and unique mechanisms regulate fibrosis in various

fibroproliferative diseases. J Clin Invest 117, 524-9.

XING, Z., GAULDIE, J., COX, G., BAUMANN, H., JORDANA, M., LEI, X. F. & ACHONG, M. K.

(1998). IL-6 is an antiinflammatory cytokine required for controlling local or

systemic acute inflammatory responses. J Clin Invest 101, 311-20.

XU, J., LAMOUILLE, S. & DERYNCK, R. (2009). TGF-beta-induced epithelial to

mesenchymal transition. Cell Res 19, 156-72.

ZAVADIL, J. & BOTTINGER, E. P. (2005). TGF-beta and epithelial-to-mesenchymal

transitions. Oncogene 24, 5764-74.

ZHANG, A., DONG, Z. & YANG, T. (2006). Prostaglandin D2 inhibits TGF-beta1-induced

epithelial-to-mesenchymal transition in MDCK cells. Am J Physiol Renal

Physiol 291, F1332-42.

ZHANG, Y. E. (2009). Non-Smad pathways in TGF-beta signaling. Cell Res 19, 128-

39.

ZISMAN, D. A., LYNCH, J. P., 3RD, TOEWS, G. B., KAZEROONI, E. A., FLINT, A. &

MARTINEZ, F. J. (2000). Cyclophosphamide in the treatment of idiopathic

pulmonary fibrosis: a prospective study in patients who failed to respond to

corticosteroids. Chest 117, 1619-26.

ZUCHT, H. D., GRABOWSKY, J., SCHRADER, M., LIEPKE, C., JURGENS, M., SCHULZ-

KNAPPE, P. & FORSSMANN, W. G. (1998). Human beta-defensin-1: A urinary

peptide present in variant molecular forms and its putative functional

implication. Eur J Med Res 3, 315-23.