Sunteți pe pagina 1din 108

Developmental and Pathological Roles

of BMP/Follistatin-like 1 in the Lung

 
PhD thesis

to obtain the degree of PhD at the


University of Groningen
on the authority of the
Rector Magnificus Prof. E. Sterken
and in accordance with
the decision by the College of Deans.

This thesis will be defended in public on 

Friday 6 October 2017 at 12.45 hours

by

Navessa Padma Tania


born on 12 April 1986
in Bandung, Indonesia
Supervisors
Prof. R. Gosens 
Prof. M. Schmidt 

Co-supervisor
Dr. H. Maarsingh

Assessment Committee
De rol van BMP/Follistatin-like 1 in
Prof. G. Folkerts  de ontwikkeling en de pathologie van de long
Prof. C. Bingle 
Prof. F.A.E. Kruyt

 
Proefschrift 

ter verkrijging van de graad van doctor aan de


Rijksuniversiteit Groningen
op gezag van de
rector magnificus prof. dr. E. Sterken
en volgensbesluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op 

vrijdag 6 oktober 2017 om 12.45 uur

door

Navessa Padma Tania


geboren op 12 april 1986
te Bandung, Indonesië
Paranymphs:
Christel Seegers
Thea van den Bosch

The research described in this thesis was carried out at the Department
of Molecular Pharmacology (Groningen Research Institute of Pharmacy,
University of Groningen, The Netherlands) according to the requirements
of the Graduate School of Science (Faculty of Science and Engineering,
University of Groningen, The Netherlands).

The research was financially supported by the Lung Foundation Netherlands


(grant 3.2.12.083).

Printing of this thesis was financially supported by the University Library of


the University of Groningen, the Graduate School of Science, and the Lung
Foundation Netherlands.

Cover story: COPD lung vs. developing lung– Putri Astri Wulandari
Printing: Ipskamp Printing B.V., Enschede, the Netherlands

ISBN (printed version): 978-94-034-0016-7


ISBN (electronic version): 978-94-034-0015-0

Copyright © 2017 by Navessa Padma Tania. All rights reserved. No parts of


Science knows no country, because knowledge belongs to humanity,
this thesis may be reproduced or transmitted in any form or by any means
and is the torch which illuminates the world – Louis Pasteur
without the prior permission from the author.
DEVELOPMENTAL AND PATHOLOGICAL ROLES OF BMP/FOLLISTATIN-LIKE 1

IN THE LUNG

Chapter 1. General Introduction.

Chapter 2. Endothelial follistatin-like-1 regulates the postnatal development of


the pulmonary vasculature by modulating BMP/Smad signaling.

Pulm Circ. 2017; 7(1):219-231

Chapter 3. The effect of cigarette smoking on pulmonary Follistatin-like 1 expres-


sion: potential implication in chronic obstructive pulmonary disease

Chapter 4. Activin-A: active in inflammation in COPD.


EurRespir J. 2014; 43: 954–955

Chapter 5. Pro-inflammatory roles of Follistatin-like 1 in coordinating epitheli-


al-mesenchymal communication in inflammatory cytokine release in
the lung


Submitted

Chapter 6. Regulation of pulmonary inflammation by mesenchymal cells.


Pulm.Pharmacol.Ther. 2014; 1-10

Chapter 7. Variant club cell differentiation is driven by bone morphogenetic pro-


tein 4 in adult human airway epithelium: Implications for goblet cell
metaplasia and basal cell hyperplasia in COPD.

Chapter 8. General discussion and summary.

Chapter 9. Dutch summary/Nederlandse samenvatting

Chapter 10. Indonesian summary/Rangkuman dalam bahasa Indonesia

Acknowledgement

Biography and list of publications

1
CHAPTER 1 1

GENERAL
INTRODUCTION

3
Preface symptoms of COPD include breathlessness, chronic and progressive dyspnea, chronic
cough, and excessive sputum production.4 The episodes of acute worsening of COPD
The primary aim of this thesis is to explore the functional roles of an endogenous symptoms, the-so-called exacerbations, aggravate primarily in patients with severe 1
bone morphogenetic protein (BMP) antagonist, Follistatin-like 1 (Fstl1), in lung disease, and approximately 78% is linked with respiratory infections.4 The molecular
development and in the pathological processes of chronic obstructive pulmonary and cellular mechanisms underlying the symptoms and pathobiology of COPD are
disease (COPD). BMP signaling plays active roles in lung development as evident in not completely understood. The current understanding from histopathology and
embryonic and perinatal lethality and lung developmental defect of BMP-deficient current available treatment of COPD is summarized below.
mice.1 It is now apparent that BMP signaling also plays crucial regulatory roles in
adult lung homeostasis by modulating inflammation and repair in the lung.2,3 1.1. The pathology of COPD
However, the involvement of BMP signaling has not been previously explored in the The pathology of COPD displays highly heterogeneous phenotypes involving
pathophysiology of COPD. We hypothesized that the imbalance between BMP and abnormal airway wall architectures, peribronchial fibrosis, destruction of alveolar
its antagonist Fstl1 contributes to dysregulation of early postnatal lung development, septa (emphysema), and altered airway epithelial composition, including goblet cell
airway inflammation, and adult airway epithelial cell differentiation similar to that metaplasia and loss of club cells.15,16 Major remodeling throughout the respiratory
observed in COPD. This thesis sheds light on Fstl1 as a novel mediator of airway tracts (including proximal, distal and peripheral airways, lung parenchyma and
epithelial-driven inflammation in the lung and potentially of goblet cell metaplasia pulmonary vasculature) and changes in the cellular differentiation profile of the
and the loss of club cells in COPD epithelium. Therefore, targeting Fstl1 is worth airway epithelium collectively manifest in a progressive decline of lung function.17 The
pursuing for the treatment of airway inflammation and epithelial remodeling in staging of COPD that reflects disease severity is typically determined by spirometry.4
COPD.The first part of this chapter provides an overview of COPD pathophysiology, A new hypothesis addresses the potential involvement of developmental pathways
including chronic airway inflammation, airway epithelial remodeling, vascular in COPD based on accumulating data from genome-wide association studies and
remodeling, and currently available treatment. The second part of this chapter microarray profiling, indicating dysregulation of developmental factors originally
covers the current understanding of BMP pathway in lung development and chronic known for their roles in embryonic development.18 BMP signaling is pivotal during
respiratory diseases. The third part of this chapter focuses on the latest discoveries lung development, nevertheless, it has been largely unexplored in adult lung as
on developmental and pathological roles of Fstl1. Finally, the scope of this thesis and well as the pathogenesis of COPD. BMP signaling is found decreased in the lung of
the specific aims of our studies are described in the last part of this chapter. pulmonary fibrosis patients and following bleomycin injury in mice.19,20 Accordingly,
accumulating evidence indicates a marked increase of the endogenous BMP
1. Chronic Obstructive Pulmonary Disease (COPD) antagonist Fstl1 in the lung of patients suffer from respiratory diseases.21–25 This
The Global Initiative for Chronic Obstructive Lung Diseases (GOLD) guidelines thus raises the possibility that an imbalance between BMP and its endogenous
stated COPD as “a common preventable and treatable disease, characterized by antagonists may lead to airway inflammation and airway epithelial remodeling in
persistent airflow limitation that is usually progressive and associated with an COPD.
enhanced chronic inflammatory response in the airways and the lung to noxious
particles or gases. Exacerbations and comorbidities contribute to the overall severity
1.2. Persistent inflammation of the airways in COPD
in individual patients’’.4 According to the World Health Organization, COPD will be
Chronic airway inflammation is the primary pathophysiological feature of COPD and
the third major cause of death worldwide by 2020, surpassing stroke.5 Cigarette
is believed to be the major contributor of structural changes in COPD airways.17 The
smoking accounts for 85-90% of COPD cases and is the best-known etiological
number of neutrophils and macrophages is positively correlated with COPD severity.26
factor for the development of COPD. However, 10-15% of COPD patients are never
Airway inflammation in COPD is associated with infiltration of various innate and
smokers and only about 15-20% of smokers manifest COPD,6 suggesting a role for
adaptive immune cells with prolonged retention time in the respiratory tracts,
environmental factors and susceptibility-associated (epi)genetic make-up. This
primarily in peripheral airways (bronchioles), lung parenchyma, and pulmonary
has led to the concept that impaired inflammatory responses to cigarettes smoke-
arteries.27,28 Previously, infiltrating leukocytes were considered to be the key player
induced injury manifest to COPD in susceptible persons.7 Accumulating evidence
of airway inflammation and the major factor in COPD development. However, latest
from genome-wide (epi)genetic association studies discovered several loci that are
discoveries show that the structural cells, such as bronchial epithelial cells, lung
strongly associated with COPD susceptibility.8–12 The major loci that associated with
fibroblasts, and airway smooth muscle cells also express and secrete molecules that
COPD susceptibility include HHIP (hedgehog interacting protein) and FAM13A (family
regulates innate airway inflammation and defense mechanisms.29–31
with sequence similarity 13 member A) on chromosome 4 as well as CHRNA3/5
(cholinergic nicotinic acetylcholine receptor) on chromosome 15.8,9,13,14 The common

4 5
As a part of the normal repair mechanism to injury, airway epithelium of Disruption of epithelial-driven innate immune functions is also strongly affected
healthy individuals mediate the activation of innate immune responses via pattern by impaired airway epithelial structure and composition following epithelial injury
recognition receptors (PRRs) to release “danger signals” which subsequently activate which will be described on the next section. Persistent airway inflammation leading 1
Toll-like receptor (TLRs).32 Activation of TLR receptor promotes release of cytokines to recruitment and activation of neutrophils and macrophages in COPD is illustrated
to recruit inflammatory cells to the site of injured epithelium in the lung. Resolution in Figure 1.
of inflammation is responsible for switching “off” the inflammatory response when
the toxic insults are successfully cleared out. However, chronic exposure to inhaled goblet cells ciliated cells club cells

toxic compounds, such as cigarette smoke, activates airway epithelial cells to secrete
a myriad of pro-inflammatory cytokines and chemokines, including interleukin-6 (IL-
6), CXC-chemokine ligand 1 (CXCL1), CXCL8, CC-chemokine ligand 2 (CCL2), C-X-C- Healthy epithelium
basal cells
Motif ligand 8, IL-1β, tumor necrosis factor (TNF)-α and granulocyte macrophage
colony-stimulating factor (GM-CSF).33–35 Airway inflammation in COPD is associated
with persistent and exaggerated innate and adaptive inflammatory responses
involving recruitment of CD8+ T-helper-1 (Th1) cells, neutrophils, monocytes, and
lymphocytes. Inflammatory mediators released by bronchial epithelial cells following
? Chapter 7
cigarette smoke-induced injury have a paracrine effect on neighboring mesenchymal BMP4

cells, such as lung fibroblasts and airway smooth muscle cells.30,36 Several studies goblet cell metaplasia loss of club cells

have shown that these mesenchymal cells are more than just structural cells as they
express and release inflammatory mediators, like IL-6, IL-8, TNF-α, which contribute
to the neutrophil and monocyte infiltration to the lung.30,31 COPD epithelium
The number of inflammatory cells, such as macrophages, neutrophils and ? Chapter 3
CD8+ T cells, is increased in sputum and bronchoalveoalar lavage (BAL) of COPD Fstl1
patients.37,38 Neutrophil chemotactic proteins including leukotriene B4 (LTB4), IL- Chapter 5 ? ? Chapter 5
8, CXCL1 and CXCL8, GRO-a (growth-related oncogene-a), and ENA-78 (epithelial
neutrophil-activating protein of 78 kDa) are increased in the sputum and BAL fluid
of COPD patients.39–41 Macrophages also induce release of LTB4, IL-8, TNF-α, MCP-1, lung fibroblasts airway smooth muscle cells

and reactive oxygen species upon cigarette smoke-induced injury. These secreted
factors activate and recruit neutrophils to the lung, leading to increase secretion IL-6
IL-8
of proteases (neutrophil elastase, proteinase-3, matrix metalloproteinases (MMP)-
8, MMP-9, cathepsin G) which is associated with emphysema.42,43 In addition, GM- Recruitment and ac�va�on of innate immune cells

CSF and granulocyte colony-stimulating factor (G-CSF) promote neutrophil survival,


proliferation, and differentiation.44,45 The transcription factor nuclear factor-κB
(NF-κB) is known to be the central regulator of inflammatory protein expression.46
In addition, E-selectin which mediates neutrophil adhesion to endothelial cells is neutrophils macrophages

increased on endothelial cells of bronchial mucosa of COPD patients.47 Macrophages


Figure 1. Persistent airway inflammation is a major hallmark of COPD pathogenesis. The
secrete chemokines interferon-c inducible protein (IP-10), interferon-inducible healthy epithelium lining of adult airways is composed of a mixture balance of goblet cells,
T-cell chemoattractant (I-TAC), and monokine-induced by interferon-c (Mig).48 The ciliated cells, basal cells and club cells. Cigarette smoke and inhaled toxic pollutants promotes
increased numbers of macrophages in COPD patients can be a result of increased airway epithelial remodeling, including goblet cell metaplasia, mucus hypersecretion,
and loss of secretory club cells. In addition, cigarette smoke and inhaled toxic pollutants
recruitment of circulatory monocytes and/or increased cell proliferation and survival. induce release of inflammatory mediators from epithelial cells with autocrine and paracrine
Collectively, accumulation of inflammatory cells in the lung aggravates a chronic effects on neighboring mesenchymal cells, such as fibroblasts and airway smooth muscle
inflammatory state and results in airway structural changes, airway obstruction, and cells. Pulmonary mesenchymal cells secrete IL-6 and IL-8 that contributes to an infiltration
of neutrophils and macrophages to the lungs. The abnormal accumulation of inflammatory
respiratory symptoms observed in COPD.49,50 cells aggravates airway inflammation in the lungs and leads to the development of COPD. The
expression of Fstl1 in COPD compared to non-COPD lungs is investigated in Chapter 3. The
role of Fstl1 in inflammatory cytokine release from pulmonary mesenchymal cells is shown
in Chapter 5.The effect of BMP4 on adult airway epithelial cells is examined in Chapter 7.

6 7
1.3. Remodeling of airway epithelium in COPD matrix, thickening of airway smooth muscle and narrowed airway lumen.64–66 In
Airway epithelium acts as the first protective barrier between the external COPD, infiltrating inflammatory cells release inflammatory mediators that promote
environment and the internal lung tissue and the rest of the body. The healthy production of extracellular matrix from pulmonary mesenchymal cells which 1
epithelium lining of adult large cartilaginous airways is made up of a layer of basal contribute to tissue remodeling and respiratory dysfunction.30 Multiple studies have
cells that are attached to the basal lamina underneath a layer of airway lumen reported increased extracellular matrix protein expression, such as fibronectin,
columnar cells with a balanced mixture of goblet cells, ciliated cells and club cells. collagen I, hyaluronan, and laminin in COPD airway and lung parenchyme.67–70 Airway
This epithelial structure is important for the mucociliary clearance function.16 In smooth muscle cell hyperplasia and hypertrophy cause thickening of smooth muscle
trachea, up to 60% of the total cell number are ciliated cells and approximately 20% bundles surrounding COPD airways, whereas airway fibroblasts contribute to sub-
are mucus secreting goblet cells.51,52 The number of ciliated cells falls to approximately epithelial fibrosis in the airway wall. The primary roles of pulmonary fibroblasts are
15% by the 5th airway generation. The distal airways predominantly consist of serous tissue repair and remodeling by producing diverse extracellular matrix proteins, such
and secretory club cells.29 In the proximal bronchioles, the epithelial cells become as fibronectin, collagen, and airway smooth muscle actin, whereas airway smooth
more cuboidal and, in addition to ciliated cells, contain secretory club cells. Finally, muscle cells are mainly responsible for airway contraction. Transforming growth
in the most distal bronchioles, only club cells are identified.53 factor (TGF)-β1 produced by epithelial cells in the small airways is a key player in
The cellular composition and function of COPD airway epithelium was found to tissue remodeling and is found to be markedly increased in COPD. It is associated
be altered as evident by the loss of tight junctions, basal and goblet cell hyperplasia, with increased deposition of extracellular matrix within and surrounding the smooth
and loss of secretory club cells and ciliated cells, resulting in loss of its barrier muscle bundles of COPD, which results in thickened airway walls and increased in
function and excessive mucus production.16,54 Cigarette smoke causes the goblet stiffness.71 An imbalance between the production and degradation of extracellular
cell hyperplasia and the loss of ciliated cells, resulting in a reduction of mucociliary matrix could possibly explain excessive matrix deposition and may lead to airway
clearance and mucus plug formation. In COPD, increased numbers of goblet cells fibrosis in COPD in addition to proliferating airway smooth muscle cells induced by
are also found in the small bronchi and bronchioles, where there are normally very cigarette smoking.72 In COPD, the number of myofibroblasts (airway fibroblasts with
few.55 The cellular events in epithelial repair after injury have been described.49,56,57 a more contractile phenotype) is increased and contributes to reduction of airway
The airway epithelium is more than just a physical barrier as evidenced by the elasticity.73 Fibroblasts from individuals with COPD have reduced capacity for tissue
fact that bronchial epithelial cell transcribe and secretes numerous soluble factors, repair due to increased prostaglandin E (PGE) and TGF-β1 expression.74 The thickness
such as cytokines, chemokines and growth factors. Club cells are important in of small airway wall is associated with COPD severity. The obstructed small airways
controlling lung inflammation by secreting a number of anti-inflammatory agents, are characterized by abnormal architecture of epithelium and smooth muscle layers
such as secretoglobin (SCGB3A) and uroplakin (UPK3A).58 Surfactant proteins and the presence of fibrosis.65,75
(SP-A, SP-B, SP-D), also called collectins, are produced by club cells and alveolar
epithelial cells and play an important role in airway epithelial defense mechanism.
1.5. Remodeling of the pulmonary vasculature in COPD
At the bronchiolar level, the club cell secretory protein (CCSP), also called CC10, is
Patients with COPD also frequently suffer from pulmonary arterial hypertension
able to modulate lung inflammatory and immune responses. However, club cells
are reduced in bronchial biopsies from COPD patients59,60 and in peripheral and (PAH), one of the common comorbidities of COPD. PAH is characterized by
central airways in lung tissue of COPD patients,61 which could contribute to the increased arterial pressure in the lungs as a result of narrowing of vessel lumen
aggravated inflammatory process. The air-liquid culture provides a valuable system due to vascular remodeling and increased vasoconstriction.76,77 Endothelial cells are
to mimic a well differentiated epithelial cells in vitro and permits the study of the re- important for vascular homeostasis by maintaining vascular tone and remodeling.78
epithelialization after injury.62,63 Therefore, this is an excellent tool to study epithelial Cigarette smoking disrupts endothelial functions of pulmonary arteries in COPD.79,80
cell differentiation under various condition. However, there are limited studies Endothelial cells are key players in regulation of cell and vessel growth, thus it is
addressing the functional roles of BMP signaling in the adult lung epithelial cells believed that endothelial dysfunction initiates vascular remodeling in COPD.
which may enlighten the consequence of aberrant BMP signaling in the context of Endothelial dysfunction leads to uncontrolled intimal endothelial cell proliferation
COPD. consequently resulting in thickened pulmonary muscular arteries observed in
smokers and patients with mild COPD.79,80 Inflamed lung tissue is hypoxic and
1.4. Remodeling of extracellular matrix in COPD promotes angiogenesis by increasing growth factors such as vascular endothelial
In healthy lung, non-cartilaginous small airways (<2 mm) contribute to only ~25% growth factor (VEGF), fibroblast growth factor (FGF), and epidermal growth factor
to the total airway resistance, whereas in COPD lungs, it becomes the major site (EGF).77,81 These growth factors are involved in tissue repair and vascular remodeling
of increased airway resistance as a result of increased deposition of extracellular which eventually contribute to microvascular remodeling observed in COPD. The

8 9
opposing effects of angiogenic and angiostatic factors such as chemokines (CXC 2.1. Players in BMP signaling
family) that induce or inhibit neovascularization, respectively, determines the 2.1.1. BMP ligands
angiogenesis process.77,82 VEGF and its receptor seem to be important for endothelial BMPs (which are also referred to as growth and differentiation factors (GDFs)) 1
and epithelial cell survival following injury. Numerous studies showed increased are secreted glycosylated proteins, with 20 different members identified and
expression of VEGF and VEGF receptors (Flk-1 and Flt-1) in pulmonary muscular characterized.98 BMP polypeptides consist of an N-terminal secretory signal peptide,
arteries of patients with mild and moderate COPD and in current smokers without a prodomain that ensures proper folding, and a C-terminal mature peptide. Mature
airway obstruction.81,83–85 An increase of angiogenic factors are also associated with BMPs are derived from cleavage of the BMP precursors by serine endoproteases in
thickening of the pulmonary arterial walls.76,81,83–85 However, VEGF expression is low the trans-Golgi network and subsequently secreted. Homo or heterodimerization
in total lung homogenates of severe emphysema patients.86 It has been proposed of BMP ligands forms an active BMP agonist. Secreted BMPs bind either to the
that low VEGF may be involved in the emphysema process through cell apoptotic extracellular matrix, extracellular antagonists, co-receptors or to transmembrane
mechanisms, as the lung microvascular endothelial cells and alveolar septal capillary receptors. Activation of BMP signal transduction leads to either transcriptional
cells needs VEGF for cell survival.86,87 Endothelial cell death leads to loss of capillaries responses (e.g. Smad-dependent signaling) or non-transcriptional responses (e.g.
may be a major mechanism of emphysema.86 Macrophages, CD8+ T lymphocytes, Smad-independent signaling, cytoskeletal rearrangements).98
neutrophils, fibroblasts that involves in immune response can promotes angiogenesis
in many ways by secreting TNF-α which also is an angiogenic factor. Additionally, 2.1.2. BMP receptors and co-receptors
the protein expression of endothelin-1 (ET-1) and ET-1 receptor ETA that mediate The BMP receptors are composed of two types of transmembrane serine/threonine
vascular smooth muscle conctration is increased in PAH.88,89 kinase receptors, type I and type II receptors, which are highly homologous but
not redundant, and are capable of activating both Smad-dependent and Smad-
1.6. Current treatments independent signaling. The receptors encompass an extracellular ligand binding
Currently, there is no effective cure for COPD. Inhaled corticosteroids (e.g. domain and a cytoplasmic serine/threonine kinase domain. The type I receptor
fluticasone, budesonide), phosphodiesterase 4 inhibitors (roflumilast), short- has two additional motifs in its kinase domain, a glycine/serine-rich region prior
acting (e.g. albuterol) or long-acting b2-adrenoreceptor agonists (e.g. salmeterol, the GS-box kinase domain and a L45 loop composed of eight amino acids.99 Type
formoterol), and long-acting anti-cholinergics (e.g. tiotropium, ipratropium, I receptors include activin receptor-like kinase 1 (ALK1), activin receptor type Ia
glycopyrronium bromide, umeclidinium) are used alone or in combination to relieve (ActRIa or ALK2), BMP receptor type Ia (BMPRIa or ALK3) and BMP receptor type
the symptoms and inflammation.90 However, these therapies do not change the Ib (BMPRIb or ALK6). These receptors form heteromeric complexes with the type
course of the disease.90 Therefore, understanding the molecular mechanism of COPD II receptors, including BMP receptor type II (BMPRII) and activin receptors type IIA
is undoubtedly important to identify a novel molecular target that can decelerate (ACVRIIA also known as ActRIIA) and activin receptors type IIB (ACVRIIB also known
the progression of COPD. A better understanding of impaired repair process which as ActRIIB).100 The specificity of BMP signaling is tightly controlled by selective
contributes to COPD may lead to the identification of new molecular targets and extracellular regulators, receptors, co-receptors, and cytosolic receptor-associated
more effective therapy options in COPD treatment. proteins to exert specific BMP functions which will be described in more detail
below. Activation of BMP signaling is preceded by oligomerization of BMP ligands
2. Bone morphogenetic protein (BMP) signaling in the lung and type I receptor kinase. Activated type I receptor undergo transphosphorylation
In 1965, Marshall R. Urist discovered osteogenic factors present in bone matrix of its GS-box via constitutively active type II BMP kinase receptors.98,99 After the
extracts capable of promoting de novo cartilage formation and bone formation formation of a heterotetrameric complex, the signal propagates to the nucleus either
in vivo, namely bone morphogenetic protein (BMP).91,92 BMP signaling belongs to in intracellular regulator Smad-dependent (canonical) or Smad-independent (non-
the TGF-β superfamily, which is phylogenetically conserved.91 Genetic studies in canonical) manner.98 Co-receptors that positively regulates BMP signaling are the
mice demonstrate the importance of BMP signaling in embryonic dorsal-ventral repulsive guidance molecules (RGM)-a and -c, Dragon (RGMb), the receptor tyrosine
paterning93–95 and embryonic lung development.3,96,97 It is now evident that kinase (RTK) c-Kit, and the TGF-β1/BMP type III receptors Endoglin and Betaglycan.
BMPs govern diverse cellular effects including stem cell maintenance, migration, In contrast, BMP signaling is negatively regulated by the pseudoreceptor BMP and
differentiation, proliferation, and programmed cell death.98 A growing body of activin membrane-bound protein (BAMBI) and the RTKs Ror2 and TrkC.98
evidence indicates the involvement of BMP signaling in normal lung physiology and
pathology conditions in adulthood, strongly suggesting that BMP signaling also plays
a pivotal role in adult lung homeostasis, including epithelial repair after injury.3

10 11
2.1.3. Downstream BMP/Smad-dependent signaling contains a nine-membered cysteine ring and the DAN/Cerberus family contains an
In the canonical pathway, the transduction of extracellular signals to regulation of eight-membered cysteine ring.110 Molecular regulation of BMP signaling pathway is
gene transcription inside the nucleus is mediated by Smad proteins.99 There are illustrated in Figure 2. 1
three types of Smad proteins, BMP receptor-regulated Smads (R-Smads), common
mediator Smad (co-Smad), and inhibitory Smads (I-Smads). BMP ligands bind to the
type I BMP receptors resulting in phosphorylation of R-Smads (Smad1/5/8), followed BMP Extracellular regulators
by trimeric complex formation with co-Smad (Smad4), and translocation of the (e.g.: Fstl1)
Gremlin

complex into the nucleus,98,100,101 which subsequently induces transcription of BMP


downstream target genes,102 presumably in collaboration with other co-repressors
and co–activators.103 I-Smads (Smad6 and Smad7) negatively regulate BMP/Smad
type III/ co-receptor ALK1
signaling by preventing phosphorylation of R-Smads, instead targeting R-Smads (e.g.: Endoglin) BMPRII
ALK2
ACVRIIA
and type I receptor for ubiquitin–proteasome degradation. Two conserved domains ACVRIIB
type II type I ALK3
ALK6 Extracellular
of R-Smads are the N-terminal DNA and protein binding MH1 (mad homology 1)
domain and the C-terminal MH2 domain which are connected by a variable proline- Cytoplasm

rich linker region.104,105 P


P

2.1.4. Downstream BMP/Smad-independent signaling


Smad-dependent Smad-independent
In addition to canonical BMP signaling, BMP ligands activate gene transcription
in a Smad-independent manner. BMP ligands can activate the MAPK pathway by TAK1
recruitment of TAK1, TAB1, and XIAP to type I BMP receptors, which subsequently P
Smad1/5
phosphorylate p38 MAPK or JNK.106 Additionally, ERK mediates transcription of
Smad4 P
BMP2-regulated genes.107 BMP regulates sets of genes which are involved in lung and Smad1/5 MAPK

vascular homeostasis, including epithelial and vascular remodeling. BMP regulated Erk JNK p38
genes, including jagged1, GATA2, and endoglin, have been identified in endothelial
cells.102 BMPs can also activate TGF-β activated kinase 1 (TAK1) leading to p38/ Nucleus
JNK/Erk MAP kinase activation.108,109 TAK1 mediates phosphorylation of Smad1 at
C-terminal serine residues, suggesting it functions as an upstream activating kinase Smad1/5
P

for Smad1/5/8 in BMP signaling.109 ON


Smad4 P ON
Smad1/5

2.1.5. Fine tuning of BMP signaling BRE BRE


BMP functions are tightly modulated by BMP antagonists, co-receptors, and Progenitor cell maintenance Proliferation
Differentiation
intracellular regulatory proteins. BMP antagonists function through direct binding
with BMP ligands and prevent BMPs from binding to their transmembrane receptors.
Pseudo-receptors sequester BMP ligands at the cell surface. Co-receptors and
intracellular regulatory proteins interact with the receptors to regulate downstream Figure 2. Molecular regulation of BMP signaling. Activation of BMP signaling is initiated by
binding of BMP ligands to the type I BMP receptors (ALK1, 2, 3, 6) followed by recruitment
BMP functioning. The spatiotemporal interplay between BMP and its regulators of constitutively active type II BMP receptor receptors (BMPRII, ACVRIIA, ACVRIIB) to form
govern developmental and cellular processes. BMP antagonists are cysteine knot a signal-activating complex. In cytoplasm, the signal propagates either via Smad-dependent
containing motif which form a functional homodimer. BMP antagonists bind or Smad-independent manner. In a Smad-dependent signaling, the transduction of BMP
signals is mediated by Smad1/5 proteins. Phosphorylated Smad1/5 proteins form a complex
selectively to distinct BMP ligands in a context- and concentration-dependent with Smad4 protein which subsequently translocate to nucleus and bind to a 52 base-pair
manner to maintain BMP gradients, which have been extensively studied during BMP response element (BRE) on the promoter region of the target genes. In a non-Smad
embryonic lung development. BMP antagonists include Noggin, gremlin, twisted signaling, BMP ligands can activate the MAPK pathway by recruitment of TAK1 to type I BMP
receptors, which subsequently phosphorylate Erk/JNK/p38. Thereby, phosphorylated Erk/
gastrulation (Tsg), and Fstl1. Antagonists are categorized based on their cysteine JNK/p38 can translocate into the nucleus. Among others, BMP functions are tightly regulated
knot motif: the Chordin/Noggin family contains a ten-membered cysteine ring, Tsg by co-receptors (e.g. Endoglin) and extracellular regulators (e.g. Fstl1).

12 13
2.2. BMP signaling in lung development and adult lung homeostasis 2.4. BMP signaling in pulmonary vascular diseases
Lung development is orchestrated by precise coordination of several molecular BMP signaling is important in embryonic vascular development and adult vascular
pathways, including BMP signaling. The canonical BMP signaling is important in lung homeostasis. Consequently, dysregulation of BMP signaling has been strongly 1
development.3 Fundamental roles of BMP signaling in lung development are evident associated with the pathogenesis of hereditary vascular diseases, including familial
from BMP mutations associated with congenital lung defects in human and genetic PAH.121 BMP signaling regulates vascular smooth muscle maturation, endothelial
studies in transgenic mice and familial mutation in PAH. Deletion of lung epithelial- cell proliferation and angiogenesis.122 Endothelial cells are a major player in vascular
specific Smad1 resulted in impaired airway branching morphogenesis, decreased homeostasis. Endothelial dysfunction, including abnormal proliferation, impaired
sacculation, disruption of distal lung epithelial cell proliferation and differentiation endothelial mesenchymal communication, decreased endothelial nitric-oxide
which contribute to severe postnatal respiratory distress. Wu et al. also revealed synthase (eNOS) activity, and loss of bioactive nitric oxide (NO), play a prominent
that BMP4/Smad1 signaling modulates other molecular pathways including Wnt/β- role in the development of PAH.113,123 Accumulating studies from genetic association
catenin by expressing Wnt inhibitory factor 1 (Wif1) in the developing fetal mouse study in hereditary vascular diseases and genetic studies in transgenic mice pinpoint
lung.111 Overexpression of BMP4 in the epithelium, on the other hand, leads to the mutation of BMP components to be strongly associated with PAH.124,125 Most
smaller lungs and to a marked decrease in epithelial cell proliferation.97 In adults, importantly, BMP2 and BMP4 failed to stimulate eNOS phosphorylation in PAECs
lung tissue homeostasis is required for continuous regeneration and subsequent derived from patients carry mutations in the BMPRII gene.113 This is supported by
differentiation of stem cells in order to maintain lung architecture and proper animal studies of lung endothelial-specific BMPR2 KO mice resulting in spontaneous
functions. BMP signaling maintains an undifferentiated state of the airway and PAH.126 Interestingly, impaired BMP signaling was also observed in common forms
alveolar epithelial progenitors for lung epithelial homeostasis and tissue injury of PAH, including hypoxic PAH.127 Mechanistically, the negative regulator of vascular
repair.3 smooth muscle cell proliferation and pro-apoptotic, BMP2 and its downstream non-
A tight and precise regulation of pulmonary vascular development is required canonical BMP signaling are upregulated after acute hypoxia exposure. In contrast,
for normal lung development.112 Canonical BMP signaling plays key roles during prolonged hypoxia exposure results in downregulation of BMPRII expression and
embryonic development of the pulmonary vasculature.3 Activation of BMPRII inactivation of its downstream signaling in pulmonary vasculature.127 In addition, the
promotes pulmonary artery endothelial cell (PAEC) survival, proliferation, and expression of BMPRIb, BMPRII, and Smad4, 5, 6, and 8 was decreased in accordance
migration. BMPRII is a novel mediator of endothelial nitric-oxide synthase with reduced levels of active Smad1 and Smad-regulated genes id1 and id3 in
activation.113 lungs of monocrotaline model of PAH.128 Furthermore, the expression of the BMP
antagonist gremlin 1 is increased in small pulmonary vessel walls of hypoxic PAH
2.3. BMP signaling in lung diseases mouse models.129 Taken together, these studies demonstrate a central role for BMP
Dysregulation of BMP signaling has been implicated in several lung diseases. Although and its inhibitors in pulmonary vascular remodeling and the pulmonary vascular
in adult lung tissue, active canonical BMP signaling is hardly detectable.3 Reactivation resistance in hypoxic PAH.129
of canonical BMP signaling is observed after lung injury with naphthalene and
bleomycin in the same pattern as early lung development in bronchial and alveolar 3. Follistatin-like 1
epithelial cells, respectively, presumably as part of repair processes.3 In addition, During lung development and adult homeostasis processes, the functions of
BMP ligands expression and BMP/Smad activity are upregulated inflamed bronchial BMPs are tightly controlled for proper morphogenesis. Although the expression
epithelium of allergic asthma.114,115 It has been demonstrated that activation of BMP of BMPs and their receptors is not strictly limited to certain areas, the expression
signaling induces expression of its own antagonist, implying a negative feedback of BMP antagonists is spatiotemporally tightly controlled, indicating that BMP
loop in the regulation of BMP signaling. The expression of the BMP4 antagonist antagonists fine tune the level of BMP functioning and act as molecular switches
gremlin was found to be upregulated in patients with idiopathic pulmonary fibrosis of BMP signaling.1 The endogenous antagonist of BMP, Fstl1 (also known as TGF-β-
(IPH).116 Additionally, the expression of BAMBI is increased in plasma and lung of stimulated clone 36 (TSC-36)), was originally identified as a TGF-β inducible gene in
COPD patients.117,118 The role of BMP in lung cancer is contradictive. In one hand, a mouse osteoblastic cell line with two isoforms.130,131Fstl1 is a secreted cysteine-rich
BMP2 is increased along with activation of Smad1/5 and its downstream target Id1 glycosylated protein of 38 kDa containing an follistatin (FS)-like domain containing
and promote tumor growth in human lung cancer.119 On the other hand, low levels 10 conserved cysteine residues and a strong similiarity with BM-40/osteonectin/
of BMP7 in lung cancer tissues are correlated with lymph node metastasis. In vitro, SPARC family.130 Human Fstl1 is highly conserved with >92% amino acid sequence
BMP7 negatively regulates cell adhesion and migration of a lung cancer cell line.120 identity with rat and mouse.132 The Fstl1 orthologues have been identified and
sequenced from human, rat, mouse, avian, xenopus, macaques, and fish.132–136 The

14 15
crystal structure of a truncated form of recombinant Fstl1 containing follistatin-like axis patterning.169 However, the role of Fstl1 in the development of pulmonary
domain has been resolved.137 Fstl1 is composed of three distinct domains, including vasculature has not been previously explored.
follistatin N-terminal domain-like (FOLN), extracellular calcium (EC)-binding domain 1
containing two EF-hand motifs, and a von Willebrand factor type C domain (VWC) 3.3. Physiological roles of Fstl1
with cell type specific glycosylation. However, unlike other family members, the Several studies reported that Fstl1 has diverse biological functions, including
calcium-binding domains of Fstl1 are non-functional.131 regulating cell proliferation,170–172 differentiation,171 migration , invasion,157 wound
healing,156 tissue repair,143,173,174 cell fate determination,139 and innate immune
3.1. Cellular expression of Fstl1 responses.172 The roles of Fstl1 in tissue repair in response to injury have been most
During embryonic development, Fstl1 mRNA is detected in different organs, but widely studied in the cardiovascular system. Fstl1 was found to be increased in
the highest expression is found in the lung, predominantly in mesenchymal cells, models of acute and chronic heart injury, including myocardial infarction, pressure
including vascular and airway smooth muscle cells,138,139 but also in endothelial cells overload-induced hyperthrophy, and ischemia/reperfusion injury.140,173,175,176
and goblet cells of airway epithelium. In adult tissue, Fstl1 has been proposed as Systemic administration of Fstl1 or overexpression of Fstl1 showed to be protective
a cardiokine,140–143 myokine,144,145 adipokine,146 and, osteogenic factor147 due to it in the cardiovascular system by reducing cell apoptosis and inflammatory responses,
is ubiquitous expression and functions in the heart, muscle, bone, skeletal muscle and promoting endothelial cell survival and trigger a revascularization process and
tissue,144,145,148 neurons,149 and trophoblasts.150 The effects of Fstl1 on its downstream restore tissue function.140,148,176,177 Recent studies demonstrated that Fstl1 expression
targets and the upstream factors known to regulate Fstl1 expression has been is also induced in response to lung injury.21,23,158 Further investigation is needed to
reviewed.151 In response to divers factors, such as TGF-β1, IL-1β, TNF-α and IL-6, unravel the impact of upregulation of Fstl1 in the lung. Collectively, these data suggest
the expression of Fstl1 gene was found to be highly expressed in mesenchymal that the injury-induced upregulation of Fstl1 play a clinically relevant role in the
lineages, including fibroblasts, cardiomyocytes, chondrocytes, adipocytes, and modulation of pathological processes. Furthermore, previous studies demonstrate
osteocytes.151–153However, recent studies showed that Fstl1 is also expressed by paracrine signaling of Fstl1 in cell and tissue communication.141,142,144,148,172 However,
hematopoietic lineages, such as bone marrow-derived osteoclasts147 and lung the roles of secreted Fstl1 in cell-cell communication in the lung have not been
alveolar macrophages.24IL-13 triggers lung alveolar macrophages to express a previously studied.
significant increase of Fstl1 mRNA, whereas TGF-β and Fstl1 increase Fstl1 expression
in lung epithelial cells.22,24 Furthermore, Fstl1 triggers inflammatory cells, ranging 3.4. The pathological roles of Fstl1 in inflammatory diseases
from monocytes, macrophages, and T-cells to express and release pro-inflammatory The function of Fstl1 has been most extensively studied in inflammatory diseases
cytokines and chemokines, including IL-1β, TNF-α, IL-6, IFN-γ, IL-8, MCP-1, and IP- and a dual role of Fstl1 in inflammatory responses has been reported. It is clear
10.151,154,155 Among others, expression of Fstl1 is negatively regulated by microRNAs that Fstl1 modulates inflammatory cytokine expression and release. However, the
(miRs), including miR-198,156 miR-27a,157 miR-21,158 and miR-206.159Although Fstl1 is precise effect of Fstl1 in inflammatory responses, whether as a pro-inflammatory
primarily known as a BMP antagonist,21,160,161 current findings suggest that Fstl1 may or an anti-inflammatory mediator remain controversial. A great body of evidence
also regulate various physiological processes by interacting with different receptors, identifies Fstl1 as a pro-inflammatory cytokine that is induced by inflammatory
including disco-interacting protein 2 homolog A (DIP2A),162–164 toll-like receptor 4 cytokines, in turn Fstl1 triggers inflammatory responses from target cells, particularly
(TLR4),165,166 and the sodium-potassium pump.149 inflammatory cells.178 Several studies have shown that Fstl1 is markedly upregulated
in inflammatory conditions.179–181 Furthermore, silencing of Fst1 decreases the
3.2. Fstl1 signaling in lung and pulmonary vascular development production of TNF-α, IL-6, and IL-8 from human arterial endothelial cells induced by
Fstl1 is expressed in the developing lung.25,138 Embryonic knockout of Fstl1 in mice oxidized low-density lipoprotein.166 Additionally, high Fstl1 is observed in response
results in an extensive distortion in lung morphology and neonatal lethality due to bacterial infection182 and low degree of inflammation in obese individuals.183
to respiratory failure, demonstrating the functional importance of Fstl1 in lung An anti-inflammatory role for Fstl1 is supported by other studies. Mice treated
development.160,161 Whole body knockout of Fstl1 exhibits smaller and abnormal with recombinant human Fstl1 show a decreased expression of IL-6, MMP3, and
lung morphology. Histological analysis demonstrated thickening of alveolar walls MMP9 in animal models of arthritis.184,185 Immuno suppressant roles of Fstl1 have
and reduction in airspaces and impaired alveolar epithelial differentiation as seen been also demonstrated in organ transplantation, and tissue injury.166,186,187 These
in reduction in mature surfactant protein level.160,161 In addition to embryonic discrepancies may be explained by the differences in the experimental conditions
lung morphogenesis,25,138,151,160,161 the crucial roles of Fstl1 in organogenesis have and different organs. However, the inflammatory roles of Fstl1 in COPD have not been
also been shown in other organs, including brain,167 heart,168 and dorsal/ventral previously explored. Furthermore, Fstl1 has been proposed as a biomarker in several
diseases, including systemic-onset juvenile rheumatoid arthritis,153 osteoarthritis,188

16 17
systemic juvenile idiopathic arthritis,181 chronic systolic heart failure,189 acute Scope of the thesis
coronary syndrome,190 inflammatory responses and oxidative stress.191
The notion that developmental signaling pathways are reactivated during chronic 1
3.5. The pathological roles of Fstl1 in airway remodeling in the lung lung disease attracted scientists to dissect the roles of these pathways related with
Recent studies showed upregulation of Fstl1 expression in the lungs patients the pathophysiology of COPD. However, little is known about the involvement
with severe asthma and idiopathic pulmonary fibrosis.22,23 Further in vivo studies of BMP signaling in COPD. BMP/Fstl1 signaling is a key player in embryonic
demonstrated that Fstl1 haploinsufficient mice and specific inactivation of Fstl1 lung morphogenesis,85,86 with diverse physiological roles including cellular
in myeloid cell lineages attenuate airway remodeling in animal models of asthma communication.139,142,172 Recent studies demonstrate that aberrant Fstl1 expression
and lung fibrosis, respectively.21,22 Studies from the same group demonstrated that is associated with several pathological lung conditions21–24,158 and inflammatory
recombinant Fstl1 induces lung macrophages to express pro-remodeling factor disorders,154,155,180 underscoring its great importance in adult tissue homeostasis
MMP9 mRNA and protein through TLR4 receptor activation.24 Conversely, the other and repair. However, the involvement of Fstl1 in COPD remains unexplored. This
study showed that Fstl1 attenuates MMP9 expression in animal model of arthritis.185 thesis will delineate the role of the endogenous BMP antagonist Fstl1 in lung
development and shed light on its potential contribution to the pathogenesis of
3.6. The pathological roles of Fstl1 in airway remodeling in cancer COPD. The specific aims of this thesis are to investigate the role of Fstl1 in pulmonary
Fstl1 has been implicated in several cancer, including lung cancer,192–194 pancreatic vasculature development and COPD, particularly in airway epithelial-mesenchymal
cancer,195 bone metastasis,196,197 ovarian cancer and endometrial carcinogenesis,198,199 cell communication and adult tracheal epithelial cell differentiation. To this aim, the
nasopharyngeal carcinoma,172 glioblastoma,200 and prostate cancer.201 However, the developmental roles of Fstl1 were studied in conditional endothelial-specific Fstl1
role of Fstl1 in cancer is complex and controversial. knockout mice and in vitro study using pulmonary cell lines, including bronchial
On one hand, Fstl1 has been reported as a pro-cancer metastasis. On the epithelial cells, lung fibroblasts, and airway smooth muscle cells.
other hand, Fstl1 shows an anti-cancer effect. High expression of Fstl1 is associated In Chapter 2, the role of endothelial Fstl1 in the postnatal development of the
with poor prognosis of glioblastoma200 and favors the progression of metastatic pulmonary vasculature was investigated. The changes in molecular signaling, lung
prostate cancer.201 Therefore, Fstl1 has been proposed as therapeutic target for physiology, and morphology of pulmonary vessels were dissected in endothelial-
prostate cancer.202 Targeting Fstl1 prevents bone metastasis by regulating the specific Fstl1 knockout mice compared to littermate controls.
immune function, suggesting that Fstl1 can mediate cancer cell invasion.197,203 Our investigation of Fstl1 expression and BMP/Smad activation in lung tissue
Fstl1 is significantly downregulated in metastatic kidney cancer. Fstl1 mRNA is of COPD patients is presented in Chapter 3, providing evidences of dysregulation of
significantly lower in cancerous tissue compared to adjacent normal tissue.204 A Fstl1 expression in different parts of COPD lung tissue.
recent study demonstrated an association between Fstl1 polymorphisms and renal Chapter 4 provides a perspective on the expression levels and contribution of
cell carcinoma (RCC) risk and prognosis, suggesting that the variant genotype CC of the TGF-β superfamily members, activin-A and its endogenous follistatin antagonist
rs1259293 is associated with RCC development by suppressing Fstl1 expression.205 in smoking-induced airway inflammation in COPD both in vitro and in vivo.
Fstl1 has been suggested as putative tumor suppressor gene as decreased Fstl1 The role of Fstl1 in inflammatory cytokine release from lung mesenchymal cells
has been reported,206 including in kidney cancer,204 lung cancer,192,194 ovarian and was explored in Chapter 5, which provides evidences on pro-inflammatory roles
endometrial cancer,198,199 nasopharyngeal carcinoma,172and bone metastasis of of Fstl1 1 in coordinating epithelial-mesenchymal communication in inflammatory
prostate cancer.196 Hypermethylation of the Fstl1 promoter is observed in cancer, cytokine release in the lung. We identified epithelial-derived factors in culture
leading to cell proliferation and invasion.172 Re-expression of Fstl1 and Fstl1 supernatants of Fstl1 overexpressing bronchial epithelial cells which mediate Fstl1-
treatment negatively regulates lung cancer cell invasion and metastasis193,207 and conditioned media-induced IL-6 and IL-8 release from lung fibroblasts.
result in growth inhibition of human lung cancer cell lines. Fstl1 transcripts are not
Chapter 6 describes the latest understanding of the contribution of pulmonary
detectable in highly aggressive and proliferative lung cancer cells and overexpression
mesenchymal cells in modulating airway inflammation in various lung diseases.
of Fstl1 shows an anti-proliferative effect.192 Tumor suppressant effects of Fstl1 in
Furthermore, the effect of BMP4 in differentiation of primary adult
ovarian and endometrial carcinogenesis has also been suggested.198 Knockdown of
Fstl1 induces apoptosis through a mitotic arrest and caspase-dependent cell death, tracheobronchial epithelialcells using air-liquid interface culture was studied in
suggesting that Fstl1 plays important roles in cellular proliferation and apoptosis in Chapter 7. The potential implications of aberrant BMP signaling related with airway
lung cancer cells.194 epithelial remodeling in COPD lungs was discussed.

18 19
References 23. Murphy N, Gaynor KU, Rowan SC, et al. Altered Expression of Bone Morphogenetic Protein
1. Yanagita M. BMP antagonists: their roles in development and involvement in pathophysiology. Accessory Proteins in Murine and Human Pulmonary Fibrosis. Am J Pathol. 2016;186(3):600-
Cytokine Growth Factor Rev. 2005;16(3):309-317. doi:10.1016/j.cytogfr.2005.02.007. 615. doi:10.1016/j.ajpath.2015.10.032.
2. Rock J, Königshoff M. Endogenous lung regeneration: potential and limitations. Am J Respir Crit 24. Miller M, Esnault S, Kurten RC, et al. Segmental allergen challenge increases levels of airway 1
Care Med. 2012;186(12):1213-1219. doi:10.1164/rccm.201207-1151PP. follistatin-like 1 in patients with asthma. J Allergy Clin Immunol. 2016;138(2):596-599.e4.
3. Sountoulidis A, Stavropoulos A, Giaglis S, et al. Activation of the canonical bone morphogenetic doi:10.1016/j.jaci.2016.01.019.
protein (BMP) pathway during lung morphogenesis and adult lung tissue repair. PloS One. 25. Li X, Fang Y, Li X, et al. Apical Secretion of FSTL1 in the Respiratory Epithelium for Normal Lung
2012;7(8):e41460. doi:10.1371/journal.pone.0041460. Development. PloS One. 2016;11(6):e0158385. doi:10.1371/journal.pone.0158385.
4. Vestbo J, Hurd SS, Agustí AG, et al. Global Strategy for the Diagnosis, Management, and Prevention 26. Di Stefano A, Capelli A, Lusuardi M, et al. Severity of airflow limitation is associated with
of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2013;187(4):347-365. severity of airway inflammation in smokers. Am J Respir Crit Care Med. 1998;158(4):1277-1285.
doi:10.1164/rccm.201204-0596PP. doi:10.1164/ajrccm.158.4.9802078.
5. Lopez AD, Murray CCJL. The global burden of disease, 1990–2020. Nat Med. 1998;4(11):1241- 27. Saetta M, Di Stefano A, Turato G, et al. CD8+ T-lymphocytes in peripheral airways of smokers
1243. doi:10.1038/3218. with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157(3 Pt 1):822-
6. Corhay J-L, Frusch N, Louis R. [COPD: genetics and environmental interactions]. Rev Médicale 826. doi:10.1164/ajrccm.157.3.9709027.
Liège. 2012;67(5-6):292-297. 28. Saetta M, Baraldo S, Corbino L, et al. CD8+ve cells in the lungs of smokers with chronic
7. Rovina N, Koutsoukou A, Koulouris NG. Inflammation and Immune Response in COPD: Where Do obstructive pulmonary disease. Am J Respir Crit Care Med. 1999;160(2):711-717. doi:10.1164/
We Stand? Mediators Inflamm. 2013;2013:e413735. doi:10.1155/2013/413735. ajrccm.160.2.9812020.
8. Pillai SG, Kong X, Edwards LD, et al. Loci identified by genome-wide association studies influence 29. Thorley AJ, Tetley TD. Pulmonary epithelium, cigarette smoke, and chronic obstructive pulmonary
different disease-related phenotypes in chronic obstructive pulmonary disease. Am J Respir Crit disease. Int J Chron Obstruct Pulmon Dis. 2007;2(4):409-428.
Care Med. 2010;182(12):1498-1505. doi:10.1164/rccm.201002-0151OC. 30. Alkhouri H, Poppinga WJ, Tania NP, Ammit A, Schuliga M. Regulation of pulmonary
9. van der Plaat DA, de Jong K, Lahousse L, et al. Genome-wide association study on the FEV1/FVC- inflammation by mesenchymal cells. Pulm Pharmacol Ther. 2014;29(2):156-165. doi:10.1016/j.
ratio in never-smokers identifies HHIP and FAM13A. J Allergy Clin Immunol. September 2016. pupt.2014.03.001.
doi:10.1016/j.jaci.2016.06.062. 31. Gosens R, Rieks D, Meurs H, et al. Muscarinic M3 receptor stimulation increases cigarette smoke-
10. Morrow JD, Cho MH, Hersh CP, et al. DNA methylation profiling in human lung tissue identifies induced IL-8 secretion by human airway smooth muscle cells. Eur Respir J. 2009;34(6):1436-
genes associated with COPD. Epigenetics. August 2016:0. doi:10.1080/15592294.2016.122645. 1443. doi:10.1183/09031936.00045209.
11. Hobbs BD, Parker MM, Chen H, et al. Exome Array Analysis Identifies a Common Variant in 32. Beg AA. Endogenous ligands of Toll-like receptors: implications for regulating inflammatory and
IL27 Associated with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. immune responses. Trends Immunol. 2002;23(11):509-512. doi:10.1016/S1471-4906(02)02317.
2016;194(1):48-57. doi:10.1164/rccm.201510-2053OC. 33. Mio T, Romberger DJ, Thompson AB, Robbins RA, Heires A, Rennard SI. Cigarette smoke
12. Bruse S, Moreau M, Bromberg Y, et al. Whole exome sequencing identifies novel candidate genes induces interleukin-8 release from human bronchial epithelial cells. Am J Respir Crit Care Med.
that modify chronic obstructive pulmonary disease susceptibility. Hum Genomics. 2016;10:1. 1997;155(5):1770-1776. doi:10.1164/ajrccm.155.5.9154890.
doi:10.1186/s40246-015-0058-7. 34. Mortaz E, Henricks P a. J, Kraneveld AD, Givi ME, Garssen J, Folkerts G. Cigarette smoke
13. Cho MH, Boutaoui N, Klanderman BJ, et al. Variants in FAM13A are associated with chronic induces the release of CXCL-8 from human bronchial epithelial cells via TLRs and induction
obstructive pulmonary disease. Nat Genet. 2010;42(3):200-202. doi:10.1038/ng.535. of the inflammasome. Biochim Biophys Acta. 2011;1812(9):1104-1110. doi:10.1016/j.
14. Pillai SG, Ge D, Zhu G, et al. A genome-wide association study in chronic obstructive pulmonary bbadis.2011.06.002.
disease (COPD): identification of two major susceptibility loci. PLoS Genet. 2009;5(3):e1000421. 35. Hellermann GR, Nagy SB, Kong X, Lockey RF, Mohapatra SS. Mechanism of cigarette smoke
15. Barnes PJ. Chronic Obstructive Pulmonary Disease. N Engl J Med. 2000;343(4):269- condensate-induced acute inflammatory response in human bronchial epithelial cells. Respir
280. doi:10.1056/NEJM200007273430407. Res. 2002;3:22.
16. Shaykhiev R, Crystal RG. Early events in the pathogenesis of chronic obstructive pulmonary 36. Osei ET, Brandsma C-A, Noordhoek JA, Timens W, Postma D, Heijink I. Crosstalk between
disease. Smoking-induced reprogramming of airway epithelial basal progenitor cells. Ann Am epithelium and fibroblasts; implications for COPD. Eur Respir J. 2014;44(Suppl 58):P3899.
Thorac Soc. 2014;11 Suppl 5:S252-258. doi:10.1513/AnnalsATS.201402-049AW. 37. Keatings VM, Barnes PJ. Granulocyte activation markers in induced sputum: comparison
17. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol. between chronic obstructive pulmonary disease, asthma, and normal subjects. Am J Respir Crit
2008;8(3):183-192. doi:10.1038/nri2254. Care Med. 1997;155(2):449-453. doi:10.1164/ajrccm.155.2.9032177.
18. Shi W, Chen F, Cardoso WV. Mechanisms of lung development: contribution to adult lung disease 38. Pesci A, Balbi B, Majori M, et al. Inflammatory cells and mediators in bronchial lavage of patients
and relevance to chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2009;6(7):558- with chronic obstructive pulmonary disease. Eur Respir J. 1998;12(2):380-386.
563. doi:10.1513/pats.200905-031RM. 39. Traves S, Culpitt S, Russell R, Barnes P, Donnelly L. Increased levels of the chemokines GROα and
19. Myllärniemi M, Lindholm P, Ryynänen MJ, et al. Gremlin-mediated decrease in bone MCP-1 in sputum samples from patients with COPD. Thorax. 2002;57(7):590-595. doi:10.1136/
morphogenetic protein signaling promotes pulmonary fibrosis. Am J Respir Crit Care Med. thorax.57.7.590.
2008;177(3):321-329. doi:10.1164/rccm.200706-945OC. 40. Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences in interleukin-8 and tumor necrosis
20. De Langhe E, Cailotto F, De Vooght V, et al. Enhanced endogenous bone morphogenetic protein factor-alpha in induced sputum from patients with chronic obstructive pulmonary disease or
signaling protects against bleomycin induced pulmonary fibrosis. Respir Res. 2015;16:38. asthma. Am J Respir Crit Care Med. 1996;153(2):530-534. doi:10.1164/ajrccm.153.2.8564092.
doi:10.1186/s12931-015-0202-x. 41. Nocker RE, Schoonbrood DF, van de Graaf EA, et al. Interleukin-8 in airway inflammation in
21. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary patients with asthma and chronic obstructive pulmonary disease. Int Arch Allergy Immunol.
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878. 1996;109(2):183-191.
22. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by Inducing 42. Beeh KM, Beier J. Handle with care: targeting neutrophils in chronic obstructive pulmonary
Oncostatin M. J Immunol Baltim Md 1950. September 2015. doi:10.4049/jimmunol.1501105. disease and severe asthma? Clin Exp Allergy J Br Soc Allergy Clin Immunol. 2006;36(2):142-157.

20 21
doi:10.1111/j.1365-2222.2006.02418.x. vitro. In Vitro Cell Dev Biol Anim. 1993;29A(6):481-492.
43. Roghanian A, Sallenave J-M. Neutrophil elastase (NE) and NE inhibitors: canonical and 64. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of airway obstruction in chronic obstructive
noncanonical functions in lung chronic inflammatory diseases (cystic fibrosis and chronic lung disease. N Engl J Med. 1968;278(25):1355-1360. doi:10.1056/NEJM196806202782501.
obstructive pulmonary disease). J Aerosol Med Pulm Drug Deliv. 2008;21(1):125-144. 65. Hogg JC, Chu F, Utokaparch S, et al. The Nature of Small-Airway Obstruction in Chronic Obstructive 1
doi:10.1089/jamp.2007.0653. Pulmonary Disease. N Engl J Med. 2004;350(26):2645-2653. doi:10.1056/NEJMoa032158.
44. Gomez-Cambronero J, Horn J, Paul CC, Baumann MA. Granulocyte-macrophage colony- 66. Hogg JC, Timens W. The Pathology of Chronic Obstructive Pulmonary Disease. Annu Rev Pathol
stimulating factor is a chemoattractant cytokine for human neutrophils: involvement of the Mech Dis. 2009;4(1):435-459. doi:10.1146/annurev.pathol.4.110807.092145.
ribosomal p70 S6 kinase signaling pathway. J Immunol Baltim Md 1950. 2003;171(12):6846- 67. Eurlings IMJ, Dentener MA, Cleutjens JPM, et al. Similar matrix alterations in alveolar and small
6855. airway walls of COPD patients. BMC Pulm Med. 2014;14:90. doi:10.1186/1471-2466-14-90.
45. Hamilton JA. GM-CSF in inflammation and autoimmunity. Trends Immunol. 2002;23(8):403-408. 68. Kranenburg AR, Willems-Widyastuti A, Moori WJ, et al. Enhanced bronchial expression of
doi:10.1016/S1471-4906(02)02260-3. extracellular matrix proteins in chronic obstructive pulmonary disease. Am J Clin Pathol.
46. Di Stefano A, Caramori G, Oates T, et al. Increased expression of nuclear factor-kappaB in 2006;126(5):725-735.
bronchial biopsies from smokers and patients with COPD. Eur Respir J. 2002;20(3):556-563. 69. Annoni R, Lanças T, Yukimatsu Tanigawa R, et al. Extracellular matrix composition in COPD. Eur
47. Di Stefano A, Maestrelli P, Roggeri A, et al. Upregulation of adhesion molecules in the bronchial Respir J. 2012;40(6):1362-1373. doi:10.1183/09031936.00192611.
mucosa of subjects with chronic obstructive bronchitis. Am J Respir Crit Care Med. 1994;149(3 70. Dentener MA, Vernooy JHJ, Hendriks S, Wouters EFM. Enhanced levels of hyaluronan in
Pt 1):803-810. doi:10.1164/ajrccm.149.3.7509705. lungs of patients with COPD: relationship with lung function and local inflammation. Thorax.
48. Saetta M, Mariani M, Panina-Bordignon P, et al. Increased expression of the chemokine 2005;60(2):114-119. doi:10.1136/thx.2003.020842.
receptor CXCR3 and its ligand CXCL10 in peripheral airways of smokers with chronic obstructive 71. Barnes PJ, Shapiro SD, Pauwels RA. Chronic obstructive pulmonary disease: molecular and
pulmonary disease. Am J Respir Crit Care Med. 2002;165(10):1404-1409. cellular mechanisms. Eur Respir J. 2003;22(4):672-688.
49. Puchelle E, Zahm J-M, Tournier J-M, Coraux C. Airway epithelial repair, regeneration, and 72. Pera T, Gosens R, Lesterhuis AH, et al. Cigarette smoke and lipopolysaccharide induce a
remodeling after injury in chronic obstructive pulmonary disease. Proc Am Thorac Soc. proliferative airway smooth muscle phenotype. Respir Res. 2010;11(1):48. doi:10.1186/1465-
2006;3(8):726-733. doi:10.1513/pats.200605-126SF. 9921-11-48.
50. Yoshida T, Tuder RM. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary 73. Evanko SP, Potter-Perigo S, Bollyky PL, Nepom GT, Wight TN. Hyaluronan and versican in the
disease. Physiol Rev. 2007;87(3):1047-1082. doi:10.1152/physrev.00048.2006. control of human T-lymphocyte adhesion and migration. Matrix Biol J Int Soc Matrix Biol.
51. Wanner A, Salathé M, O’Riordan TG. Mucociliary clearance in the airways. Am J Respir Crit Care 2012;31(2):90-100. doi:10.1016/j.matbio.2011.10.004.
Med. 1996;154(6 Pt 1):1868-1902. doi:10.1164/ajrccm.154.6.8970383. 74. Togo S, Holz O, Liu X, et al. Lung Fibroblast Repair Functions in Patients with Chronic Obstructive
52. Lozewicz S, Wells C, Gomez E, et al. Morphological integrity of the bronchial epithelium in mild Pulmonary Disease Are Altered by Multiple Mechanisms. Am J Respir Crit Care Med.
asthma. Thorax. 1990;45(1):12-15. 2008;178(3):248-260. doi:10.1164/rccm.200706-929OC.
53. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell 75. van den Berge M, ten Hacken NHT, Cohen J, Douma WR, Postma DS. Small airway disease in
populations. Nat Med. 2014;20(8):822-832. doi:10.1038/nm.3642. asthma and copd: Clinical implications. Chest. 2011;139(2):412-423. doi:10.1378/chest.10-1210.
54. Brune K, Frank J, Schwingshackl A, Finigan J, Sidhaye VK. Pulmonary epithelial barrier function: 76. Hashimoto M, Tanaka H, Abe S. Quantitative analysis of bronchial wall vascularity in the medium
some new players and mechanisms. Am J Physiol - Lung Cell Mol Physiol. 2015;308(8):L731-L745. and small airways of patients with asthma and COPD. Chest. 2005;127(3):965-972. doi:10.1378/
doi:10.1152/ajplung.00309.2014. chest.127.3.965.
55. Cosio MG, Hale KA, Niewoehner DE. Morphologic and morphometric effects of prolonged 77. Siafakas NM, Antoniou KM, Tzortzaki EG. Role of angiogenesis and vascular remodeling in
cigarette smoking on the small airways. Am Rev Respir Dis. 1980;122(2):265-221. doi:10.1164/ chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2007;2(4):453-462.
arrd.1980.122.2.265. 78. Green CE, Turner AM. The role of the endothelium in asthma and chronic obstructive pulmonary
56. Coraux C, Hajj R, Lesimple P, Puchelle E. In vivo models of human airway epithelium repair and disease (COPD). Respir Res. 2017;18:20. doi:10.1186/s12931-017-0505-1.
regeneration. Eur Respir Rev. 2005;14(97):131-136. doi:10.1183/09059180.05.00009702. 79. Santos S, Peinado VI, Ramírez J, et al. Characterization of pulmonary vascular remodelling in
57. Yahaya B. Understanding Cellular Mechanisms Underlying Airway Epithelial Repair: Selecting the smokers and patients with mild COPD. Eur Respir J. 2002;19(4):632-638.
Most Appropriate Animal Models. Sci World J. 2012;2012:e961684. doi:10.1100/2012/961684. 80. Ferrer E, Peinado VI, Díez M, et al. Effects of cigarette smoke on endothelial function of
58. Rokicki W, Rokicki M, Wojtacha J, Dżeljijli A. The role and importance of club cells (Clara cells) in pulmonary arteries in the guinea pig. Respir Res. 2009;10(1):76. doi:10.1186/1465-9921-10-76.
the pathogenesis of some respiratory diseases. Kardiochirurgia Torakochirurgia Pol Pol J Cardio- 81. Santos S, Peinado VI, Ramirez J, et al. Enhanced expression of vascular endothelial growth factor
Thorac Surg. 2016;13(1):26-30. doi:10.5114/kitp.2016.58961. in pulmonary arteries of smokers and patients with moderate chronic obstructive pulmonary
59. Gamez AS, Gras D, Petit A, et al. Supplementing Defect in Club Cell Secretory Protein Attenuates disease. Am J Respir Crit Care Med. 2003;167(9):1250-1256. doi:10.1164/rccm.200210-1233OC.
Airway Inflammation in COPD. Chest. 2015;147(6):1467-1476. doi:10.1378/chest.14-1174. 82. Nilsson I, Shibuya M, Wennström S. Differential activation of vascular genes by hypoxia in
60. Park HY, Churg A, Wright JL, et al. Club cell protein 16 and disease progression in chronic primary endothelial cells. Exp Cell Res. 2004;299(2):476-485. doi:10.1016/j.yexcr.2004.06.005.
obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(12):1413-1419. 83. Kranenburg AR, de Boer WI, Alagappan VKT, Sterk PJ, Sharma HS. Enhanced bronchial expression
doi:10.1164/rccm.201305-0892OC. of vascular endothelial growth factor and receptors (Flk-1 and Flt-1) in patients with chronic
61. Pilette C, Godding V, Kiss R, et al. Reduced epithelial expression of secretory component in small obstructive pulmonary disease. Thorax. 2005;60(2):106-113. doi:10.1136/thx.2004.023986.
airways correlates with airflow obstruction in chronic obstructive pulmonary disease. Am J 84. Calabrese C, Bocchino V, Vatrella A, et al. Evidence of angiogenesis in bronchial biopsies
Respir Crit Care Med. 2001;163(1):185-194. doi:10.1164/ajrccm.163.1.9912137. of smokers with and without airway obstruction. Respir Med. 2006;100(8):1415-1422.
62. Whitcutt MJ, Adler KB, Wu R. A biphasic chamber system for maintaining polarity of doi:10.1016/j.rmed.2005.11.009.
differentiation of cultured respiratory tract epithelial cells. Vitro Cell Dev Biol J Tissue Cult Assoc. 85. Peinado VI, Ramírez J, Roca J, Rodriguez-Roisin R, Barberà JA. Identification of vascular progenitor
1988;24(5):420-428. cells in pulmonary arteries of patients with chronic obstructive pulmonary disease. Am J Respir
63. Kaartinen L, Nettesheim P, Adler KB, Randell SH. Rat tracheal epithelial cell differentiation in Cell Mol Biol. 2006;34(3):257-263. doi:10.1165/rcmb.2005-0255OC.

22 23
86. Kasahara Y, Tuder RM, Taraseviciene-Stewart L, et al. Inhibition of VEGF receptors causes lung 110. Gazzerro E, Canalis E. Bone morphogenetic proteins and their antagonists. Rev Endocr Metab
cell apoptosis and emphysema. J Clin Invest. 2000;106(11):1311-1319. doi:10.1172/JCI10259. Disord. 2006;7(1-2):51-65. doi:10.1007/s11154-006-9000-6.
87. Voelkel NF, Gomez-Arroyo J, Mizuno S. COPD/emphysema: The vascular story. Pulm Circ. 111. Xu B, Chen C, Chen H, et al. Smad1 and its target gene Wif1 coordinate BMP and Wnt signaling
2011;1(3):320-326. doi:10.4103/2045-8932.87295. activities to regulate fetal lung development. Dev Camb Engl. 2011;138(5):925-935. doi:10.1242/ 1
88. White DG, Cannon TR, Garratt H, Mundin JW, Sumner MJ, Watts IS. Endothelin ETA and ETB dev.062687.
receptors mediate vascular smooth-muscle contraction. J Cardiovasc Pharmacol. 1993;22 Suppl 112. Kool H, Mous D, Tibboel D, de Klein A, Rottier RJ. Pulmonary vascular development goes awry in
8:S144-148. congenital lung abnormalities. Birth Defects Res Part C Embryo Today Rev. 2014;102(4):343-358.
89. Böhm F, Pernow J. The importance of endothelin-1 for vascular dysfunction in cardiovascular doi:10.1002/bdrc.21085.
disease. Cardiovasc Res. 2007;76(1):8-18. doi:10.1016/j.cardiores.2007.06.004. 113. Gangopahyay A, Oran M, Bauer EM, et al. Bone morphogenetic protein receptor II is a novel
90. Celli BR, MacNee W, ATS/ERS Task Force. Standards for the diagnosis and treatment of patients mediator of endothelial nitric-oxide synthase activation. J Biol Chem. 2011;286(38):33134-
with COPD: a summary of the ATS/ERS position paper. Eur Respir J. 2004;23(6):932-946. 33140. doi:10.1074/jbc.M111.274100.
91. Wozney JM, Rosen V, Celeste AJ, et al. Novel regulators of bone formation: molecular clones and 114. Rosendahl A, Pardali E, Speletas M, Ten Dijke P, Heldin C-H, Sideras P. Activation of bone
activities. Science. 1988;242(4885):1528-1534. morphogenetic protein/Smad signaling in bronchial epithelial cells during airway inflammation.
92. Urist MR. A morphogenetic matrix for differentiation of bone tissue. Calcif Tissue Res. Am J Respir Cell Mol Biol. 2002;27(2):160-169. doi:10.1165/ajrcmb.27.2.4779.
1970:Suppl:98-101. 115. Kariyawasam HH, Xanthou G, Barkans J, Aizen M, Kay AB, Robinson DS. Basal expression of
93. Wagner DS, Mullins MC. Modulation of BMP Activity in Dorsal-Ventral Pattern Formation by the bone morphogenetic protein receptor is reduced in mild asthma. Am J Respir Crit Care Med.
Chordin and Ogon Antagonists. Dev Biol. 2002;245(1):109-123. doi:10.1006/dbio.2002.0614. 2008;177(10):1074-1081. doi:10.1164/rccm.200709-1376OC.
94. Zuniga E, Rippen M, Alexander C, Schilling TF, Crump JG. Gremlin 2 regulates distinct roles of 116. Koli K, Myllärniemi M, Vuorinen K, et al. Bone morphogenetic protein-4 inhibitor gremlin is
BMP and Endothelin 1 signaling in dorsoventral patterning of the facial skeleton. Development. overexpressed in idiopathic pulmonary fibrosis. Am J Pathol. 2006;169(1):61-71. doi:10.2353/
2011;138(23):5147-5156. doi:10.1242/dev.067785. ajpath.2006.051263.
95. Hsu DR, Economides AN, Wang X, Eimon PM, Harland RM. The Xenopus dorsalizing factor 117. Zhang J-C, Chen G, Chen L, et al. TGF-β/BAMBI pathway dysfunction contributes to peripheral
Gremlin identifies a novel family of secreted proteins that antagonize BMP activities. Mol Cell. Th17/Treg imbalance in chronic obstructive pulmonary disease. Sci Rep. 2016;6:31911.
1998;1(5):673-683. doi:10.1038/srep31911.
96. Weaver M, Yingling JM, Dunn NR, Bellusci S, Hogan BL. Bmp signaling regulates proximal-distal 118. Drömann D, Rupp J, Rohmann K, et al. The TGF-beta-Pseudoreceptor BAMBI is strongly expressed
differentiation of endoderm in mouse lung development. Dev Camb Engl. 1999;126(18):4005- in COPD lungs and regulated by nontypeable Haemophilus influenzae. Respir Res. 2010;11:67.
4015. doi:10.1186/1465-9921-11-67.
97. Bellusci S, Henderson R, Winnier G, Oikawa T, Hogan BL. Evidence from normal expression 119. Langenfeld EM, Kong Y, Langenfeld J. Bone morphogenetic protein 2 stimulation of tumor
and targeted misexpression that bone morphogenetic protein (Bmp-4) plays a role in mouse growth involves the activation of Smad-1/5. Oncogene. 2006;25(5):685-692. doi:10.1038/
embryonic lung morphogenesis. Dev Camb Engl. 1996;122(6):1693-1702. sj.onc.1209110.
98. Sieber C, Kopf J, Hiepen C, Knaus P. Recent advances in BMP receptor signaling. Cytokine Growth 120. Chen J, Ye L, Xie F, Yang Y, Zhang L, Jiang WG. Expression of bone morphogenetic protein 7 in lung
Factor Rev. 2009;20(5-6):343-355. doi:10.1016/j.cytogfr.2009.10.007. cancer and its biological impact on lung cancer cells. Anticancer Res. 2010;30(4):1113-1120.
99. Shimasaki S, Moore RK, Otsuka F, Erickson GF. The bone morphogenetic protein system in 121. Atkinson C, Stewart S, Upton PD, et al. Primary Pulmonary Hypertension Is Associated With
mammalian reproduction. Endocr Rev. 2004;25(1):72-101. doi:10.1210/er.2003-0007. Reduced Pulmonary Vascular Expression of Type II Bone Morphogenetic Protein Receptor.
100. Miyazono K, Maeda S, Imamura T. BMP receptor signaling: transcriptional targets, regulation of Circulation. 2002;105(14):1672-1678. doi:10.1161/01.CIR.0000012754.72951.3D.
signals, and signaling cross-talk. Cytokine Growth Factor Rev. 2005;16(3):251-263. doi:10.1016/j. 122. Yao Y, Jumabay M, Wang A, Boström KI. Matrix Gla protein deficiency causes arteriovenous
cytogfr.2005.01.009. malformations in mice. J Clin Invest. 2011;121(8):2993-3004. doi:10.1172/JCI57567.
101. Massagué J, Chen YG. Controlling TGF-beta signaling. Genes Dev. 2000;14(6):627-644. 123. Frank DB, Lowery J, Anderson L, Brink M, Reese J, Caestecker M de. Increased susceptibility to
102. Morikawa M, Koinuma D, Tsutsumi S, et al. ChIP-seq reveals cell type-specific binding patterns hypoxic pulmonary hypertension in Bmpr2 mutant mice is associated with endothelial dysfunction
of BMP-specific Smads and a novel binding motif. Nucleic Acids Res. 2011;39(20):8712-8727. in the pulmonary vasculature. Am J Physiol - Lung Cell Mol Physiol. 2008;294(1):L98-L109.
doi:10.1093/nar/gkr572. doi:10.1152/ajplung.00034.2007.
103. Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell membrane to the nucleus. Cell. 124. International PPH Consortium, Lane KB, Machado RD, et al. Heterozygous germline mutations
2003;113(6):685-700. in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat
104. Wrana JL. Regulation of Smad activity. Cell. 2000;100(2):189-192. Genet. 2000;26(1):81-84. doi:10.1038/79226.
105. Xu L. Regulation of Smad Activities. Biochim Biophys Acta. 2006;1759(11-12):503-513. 125. Machado RD, Aldred MA, James V, et al. Mutations of the TGF-beta type II receptor BMPR2 in
doi:10.1016/j.bbaexp.2006.11.001. pulmonary arterial hypertension. Hum Mutat. 2006;27(2):121-132. doi:10.1002/humu.20285.
106. Yamaguchi K, Nagai S, Ninomiya-Tsuji J, et al. XIAP, a cellular member of the inhibitor of 126. Hong K-H, Lee YJ, Lee E, et al. Genetic ablation of the BMPR2 gene in pulmonary endothelium is
apoptosis protein family, links the receptors to TAB1-TAK1 in the BMP signaling pathway. EMBO sufficient to predispose to pulmonary arterial hypertension. Circulation. 2008;118(7):722-730.
J. 1999;18(1):179-187. doi:10.1093/emboj/18.1.179. doi:10.1161/CIRCULATIONAHA.107.736801.
107. Lai C-F, Cheng S-L. Signal transductions induced by bone morphogenetic protein-2 127. Takahashi H, Goto N, Kojima Y, et al. Downregulation of type II bone morphogenetic
and transforming growth factor-beta in normal human osteoblastic cells. J Biol Chem. protein receptor in hypoxic pulmonary hypertension. Am J Physiol - Lung Cell Mol Physiol.
2002;277(18):15514-15522. doi:10.1074/jbc.M200794200. 2006;290(3):L450-L458. doi:10.1152/ajplung.00206.2005.
108. Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family 128. Morty RE, Nejman B, Kwapiszewska G, et al. Dysregulated bone morphogenetic protein signaling
signalling. Nature. 2003;425(6958):577-584. doi:10.1038/nature02006. in monocrotaline-induced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol.
109. Shim J-H, Greenblatt MB, Xie M, et al. TAK1 is an essential regulator of BMP signalling in cartilage. 2007;27(5):1072-1078. doi:10.1161/ATVBAHA.107.141200.
EMBO J. 2009;28(14):2028-2041. doi:10.1038/emboj.2009.162. 129. Cahill E, Costello CM, Rowan SC, et al. Gremlin plays a key role in the pathogenesis of pulmonary

24 25
hypertension. Circulation. 2012;125(7):920-930. doi:10.1161/CIRCULATIONAHA.111.038125. dependent mechanism. J Biol Chem. 2008;283(47):32802-32811. doi:10.1074/jbc.M803440200.
130. Shibanuma M, Mashimo J, Mita A, Kuroki T, Nose K. Cloning from a mouse osteoblastic cell line 149. Li K-C, Zhang F-X, Li C-L, et al. Follistatin-like 1 suppresses sensory afferent transmission by
of a set of transforming-growth-factor-beta 1-regulated genes, one of which seems to encode a activating Na+,K+-ATPase. Neuron. 2011;69(5):974-987. doi:10.1016/j.neuron.2011.01.022.
follistatin-related polypeptide. Eur J Biochem FEBS. 1993;217(1):13-19. 150. Mouillet J-F, Mishima T, Paffaro AM do A, et al. The expression and post-transcriptional regulation 1
131. Hambrock HO, Kaufmann B, Müller S, et al. Structural characterization of TSC-36/Flik: analysis of of FSTL1 transcripts in placental trophoblasts. Placenta. 2015;36(11):1231-1238. doi:10.1016/j.
two charge isoforms. J Biol Chem. 2004;279(12):11727-11735. doi:10.1074/jbc.M309318200. placenta.2015.09.005.
132. Zwijsen A, Blockx H, Van Arnhem W, et al. Characterization of a rat C6 glioma-secreted follistatin- 151. Sylva M, Moorman AFM, van den Hoff MJB. Follistatin-like 1 in vertebrate development. Birth
related protein (FRP). Cloning and sequence of the human homologue. Eur J Biochem FEBS. Defects Res Part C Embryo Today Rev. 2013;99(1):61-69. doi:10.1002/bdrc.21030.
1994;225(3):937-946. 152. Chaly Y, Fu Y, Marinov A, et al. Follistatin-like protein 1 enhances NLRP3 inflammasome-
133. Patel K, Connolly DJ, Amthor H, Nose K, Cooke J. Cloning and early dorsal axial expression of Flik, mediated IL-1β secretion from monocytes and macrophages. Eur J Immunol. 2014;44(5):1467-
a chick follistatin-related gene: evidence for involvement in dorsalization/neural induction. Dev 1479. doi:10.1002/eji.201344063.
Biol. 1996;178(2):327-342. doi:10.1006/dbio.1996.0222. 153. Wilson DC, Marinov AD, Blair HC, et al. Follistatin-like protein 1 is a mesenchyme-derived
134. Okabayashi K, Shoji H, Onuma Y, et al. cDNA cloning and distribution of the Xenopus follistatin- inflammatory protein and may represent a biomarker for systemic-onset juvenile rheumatoid
related protein. Biochem Biophys Res Commun. 1999;254(1):42-48. doi:10.1006/bbrc.1998.9892. arthritis. Arthritis Rheum. 2010;62(8):2510-2516. doi:10.1002/art.27485.
135. Tochitani S, Liang F, Watakabe A, Hashikawa T, Yamamori T. The occ1 gene is preferentially 154. Miyamae T, Marinov AD, Sowders D, et al. Follistatin-Like Protein-1 Is a Novel Proinflammatory
expressed in the primary visual cortex in an activity-dependent manner: a pattern of gene Molecule. J Immunol. 2006;177(7):4758-4762. doi:10.4049/jimmunol.177.7.4758.
expression related to the cytoarchitectonic area in adult macaque neocortex. Eur J Neurosci. 155. Chaly Y, Marinov AD, Oxburgh L, Bushnell DS, Hirsch R. FSTL1 promotes arthritis in mice by
2001;13(2):297-307. enhancing inflammatory cytokine/chemokine expression. Arthritis Rheum. 2012;64(4):1082-
136. Sun Y-W, Li F-G, Chen J, Jiang X-Y, Zou S-M. Two follistatin-like 1 homologs are differentially 1088. doi:10.1002/art.33422.
expressed in adult tissues and during embryogenesis in grass carp (Ctenopharyngodon idellus). 156. Sundaram GM, Common JEA, Gopal FE, et al. /`See-saw/’ expression of microRNA-198 and
Gen Comp Endocrinol. 2015;223:1-8. doi:10.1016/j.ygcen.2015.09.017. FSTL1 from a single transcript in wound healing. Nature. 2013;495(7439):103-106. doi:10.1038/
137. Li L, Li X, Liu X, et al. Expression, characterization, and preliminary X-ray crystallographic nature11890.
analysis of recombinant murine Follistatin-like 1 expressed in Drosophila S2 cells. Biosci Trends. 157. Shi D-L, Shi G-R, Xie J, Du X-Z, Yang H. MicroRNA-27a Inhibits Cell Migration and Invasion of
2013;7(2):93-100. Fibroblast-Like Synoviocytes by Targeting Follistatin-Like Protein 1 in Rheumatoid Arthritis. Mol
138. Adams D, Larman B, Oxburgh L. Developmental expression of mouse Follistatin-like 1 (Fstl1): Cells. 2016;39(8):611-618. doi:10.14348/molcells.2016.0103.
Dynamic regulation during organogenesis of the kidney and lung. Gene Expr Patterns. 158. Liu M, Liu R, Wu H, et al. Radix puerariae extracts ameliorate paraquat-induced pulmonary
2007;7(4):491-500. doi:10.1016/j.modgep.2006.10.009. fibrosis by attenuating follistatin-like 1 and nuclear factor erythroid 2p45-related factor-2
139. Umezu T, Yamanouchi H, Iida Y, Miura M, Tomooka Y. Follistatin-like-1, a diffusible mesenchymal signalling pathways through downregulation of miRNA-21 expression. BMC Complement Altern
factor determines the fate of epithelium. Proc Natl Acad Sci U S A. 2010;107(10):4601-4606. Med. 2016;16:11. doi:10.1186/s12906-016-0991-6.
doi:10.1073/pnas.0909501107. 159. Rosenberg MI, Georges SA, Asawachaicharn A, Analau E, Tapscott SJ. MyoD inhibits Fstl1 and Utrn
140. Oshima Y, Ouchi N, Sato K, Izumiya Y, Pimentel DR, Walsh K. Follistatin-like 1 is an Akt-regulated expression by inducing transcription of miR-206. J Cell Biol. 2006;175(1):77-85. doi:10.1083/
cardioprotective factor that is secreted by the heart. Circulation. 2008;117(24):3099-3108. jcb.200603039.
doi:10.1161/CIRCULATIONAHA.108.767673. 160. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP)
141. Shimano M, Ouchi N, Nakamura K, et al. Cardiac myocyte follistatin-like 1 functions to attenuate 4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A.
hypertrophy following pressure overload. Proc Natl Acad Sci U S A. 2011;108(43):E899-906. 2011;108(17):7058-7063. doi:10.1073/pnas.1007293108.
doi:10.1073/pnas.1108559108. 161. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal
142. Hayakawa S, Ohashi K, Shibata R, et al. Cardiac Myocyte-Derived Follistatin-Like 1 Prevents and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616.
Renal Injury in a Subtotal Nephrectomy Model. J Am Soc Nephrol JASN. 2015;26(3):636-646. 162. Ouchi N, Asaumi Y, Ohashi K, et al. DIP2A functions as a FSTL1 receptor. J Biol Chem.
doi:10.1681/ASN.2014020210. 2010;285(10):7127-7134. doi:10.1074/jbc.M109.069468.
143. Wei K, Serpooshan V, Hurtado C, et al. Epicardial FSTL1 reconstitution regenerates the adult 163. Tanaka M, Murakami K, Ozaki S, et al. DIP2 disco-interacting protein 2 homolog A (Drosophila)
mammalian heart. Nature. 2015;525(7570):479-485. doi:10.1038/nature15372. is a candidate receptor for follistatin-related protein/follistatin-like 1--analysis of their binding
144. Miyabe M, Ohashi K, Shibata R, et al. Muscle-derived follistatin-like 1 functions to reduce with TGF-β superfamily proteins. FEBS J. 2010;277(20):4278-4289. doi:10.1111/j.1742-
neointimal formation after vascular injury. Cardiovasc Res. 2014;103(1):111-120. doi:10.1093/ 4658.2010.07816.x.
cvr/cvu105. 164. Liang X, Hu Q, Li B, et al. Follistatin-Like 1 Attenuates Apoptosis via Disco-Interacting
145. Görgens SW, Raschke S, Holven KB, Jensen J, Eckardt K, Eckel J. Regulation of follistatin-like Protein 2 Homolog A/Akt Pathway After Middle Cerebral Artery Occlusion in Rats. Stroke.
protein 1 expression and secretion in primary human skeletal muscle cells. Arch Physiol Biochem. 2014;45(10):3048-3054. doi:10.1161/STROKEAHA.114.006092.
2013;119(2):75-80. doi:10.3109/13813455.2013.768270. 165. Murakami K, Tanaka M, Usui T, et al. Follistatin-related protein/follistatin-like 1 evokes an
146. Wu Y, Zhou S, Smas CM. Downregulated expression of the secreted glycoprotein follistatin-like innate immune response via CD14 and toll-like receptor 4. FEBS Lett. 2012;586(4):319-324.
1 (Fstl1) is a robust hallmark of preadipocyte to adipocyte conversion. Mech Dev. 2010;127(3- doi:10.1016/j.febslet.2012.01.010.
4):183-202. doi:10.1016/j.mod.2009.12.003. 166. Guo J, Liang W, Li J, Long J. Knockdown of FSTL1 inhibits oxLDL-induced inflammation responses
147. Kim H-J, Kang WY, Seong SJ, Kim S-Y, Lim M-S, Yoon Y-R. Follistatin-like 1 promotes osteoclast through the TLR4/MyD88/NF-κB and MAPK pathway. Biochem Biophys Res Commun. August
formation via RANKL-mediated NF-κB activation and M-CSF-induced precursor proliferation. Cell 2016. doi:10.1016/j.bbrc.2016.08.138.
Signal. 2016;28(9):1137-1144. doi:10.1016/j.cellsig.2016.05.018. 167. Liu R, Yang Y, Shen J, et al. Fstl1 is involved in the regulation of radial glial scaffold development.
148. Ouchi N, Oshima Y, Ohashi K, et al. Follistatin-like 1, a secreted muscle protein, promotes Mol Brain. 2015;8:53. doi:10.1186/s13041-015-0144-8.
endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase- 168. van den Berg G, Somi S, Buffing AAM, Moorman AFM, van den Hoff MJB. Patterns of expression

26 27
of the Follistatin and Follistatin-like1 genes during chicken heart development: a potential IL-1β expression in cisplatin nephrotoxicity. Am J Physiol - Ren Physiol. 2010;299(6):F1320-F1327.
role in valvulogenesis and late heart muscle cell formation. Anat Rec Hoboken NJ 2007. doi:10.1152/ajprenal.00325.2010.
2007;290(7):783-787. doi:10.1002/ar.20559. 188. Wang Y, Li D, Xu N, et al. Follistatin-like protein 1: a serum biochemical marker reflecting the
169. Towers P, Patel K, Withington S, Isaac A, Cooke J. Flik, a chick follistatin-related gene, functions severity of joint damage in patients with osteoarthritis. Arthritis Res Ther. 2011;13(6):R193. 1
in gastrular dorsalisation/neural induction and in subsequent maintenance of midline Sonic doi:10.1186/ar3522.
hedgehog signalling. Dev Biol. 1999;214(2):298-317. doi:10.1006/dbio.1999.9421. 189. El-Armouche A, Ouchi N, Tanaka K, et al. Follistatin-like 1 in chronic systolic heart failure:
170. Ni S, Miao K, Zhou X, et al. The involvement of follistatin-like protein 1 in osteoarthritis by a marker of left ventricular remodeling. Circ Heart Fail. 2011;4(5):621-627. doi:10.1161/
elevating NF-κB-mediated inflammatory cytokines and enhancing fibroblast like synoviocyte CIRCHEARTFAILURE.110.960625.
proliferation. Arthritis Res Ther. 2015;17:91. doi:10.1186/s13075-015-0605-6. 190. Widera C, Horn-Wichmann R, Kempf T, et al. Circulating concentrations of follistatin-like
171. Chaly Y, Blair HC, Smith SM, et al. Follistatin-like protein 1 regulates chondrocyte proliferation 1 in healthy individuals and patients with acute coronary syndrome as assessed by an
and chondrogenic differentiation of mesenchymal stem cells. Ann Rheum Dis. 2015;74(7):1467- immunoluminometric sandwich assay. Clin Chem. 2009;55(10):1794-1800. doi:10.1373/
1473. doi:10.1136/annrheumdis-2013-204822. clinchem.2009.129411.
172. Zhou X, Xiao X, Huang T, et al. Epigenetic inactivation of follistatin-like 1 mediates tumor immune 191. Hayakawa S, Ohashi K, Shibata R, et al. Association of Circulating Follistatin-Like 1 Levels with
evasion in nasopharyngeal carcinoma. Oncotarget. 2016;7(13):16433-16444. doi:10.18632/ Inflammatory and Oxidative Stress Markers in Healthy Men. PloS One. 2016;11(5):e0153619.
oncotarget.7654. doi:10.1371/journal.pone.0153619.
173. Maruyama S, Nakamura K, Papanicolaou KN, et al. Follistatin-like 1 promotes cardiac 192. Sumitomo K, Kurisaki A, Yamakawa N, et al. Expression of a TGF-beta1 inducible gene, TSC-36,
fibroblast activation and protects the heart from rupture. EMBO Mol Med. 2016;8(8):949-966. causes growth inhibition in human lung cancer cell lines. Cancer Lett. 2000;155(1):37-46.
doi:10.15252/emmm.201506151. 193. Zhao W, Han H-B, Zhang Z-Q. Suppression of lung cancer cell invasion and metastasis by
174. Chen W, Xia J, Hu P, et al. Follistatin-like 1 protects cardiomyoblasts from injury induced by connexin43 involves the secretion of follistatin-like 1 mediated via histone acetylation. Int J
sodium nitroprusside through modulating Akt and Smad1/5/9 signaling. Biochem Biophys Res Biochem Cell Biol. 2011;43(10):1459-1468. doi:10.1016/j.biocel.2011.06.009.
Commun. 2016;469(3):418-423. doi:10.1016/j.bbrc.2015.12.026. 194. Bae K, Park KE, Han J, Kim J, Kim K, Yoon K-A. Mitotic cell death caused by follistatin-like 1
175. Lara-Pezzi E, Felkin LE, Birks EJ, et al. Expression of follistatin-related genes is altered in heart inhibition is associated with up-regulated Bim by inactivated Erk1/2 in human lung cancer cells.
failure. Endocrinology. 2008;149(11):5822-5827. doi:10.1210/en.2008-0151. Oncotarget. 2016;7(14):18076-18084. doi:10.18632/oncotarget.6729.
176. Ogura Y, Ouchi N, Ohashi K, et al. Therapeutic impact of follistatin-like 1 on myocardial 195. Hodgson G, Hager JH, Volik S, et al. Genome scanning with array CGH delineates regional
ischemic injury in preclinical models. Circulation. 2012;126(14):1728-1738. doi:10.1161/ alterations in mouse islet carcinomas. Nat Genet. 2001;29(4):459-464. doi:10.1038/ng771.
CIRCULATIONAHA.112.115089. 196. Ding T, He X-Z, Xu X-L, Xu H-Y, Zhou C-X, Wang Y-J. [Expression of serum FSTL-1 in bone
177. Alteköester A-K, Harvey RP. Bioengineered FSTL1 Patches Restore Cardiac Function metastasis of prostate cancer and its clinical implication]. Zhonghua Nan Ke Xue Natl J Androl.
Following Myocardial Infarction. Trends Mol Med. 2015;21(12):731-733. doi:10.1016/j. 2014;20(12):1090-1092.
molmed.2015.10.006. 197. Kudo-Saito C, Fuwa T, Murakami K, Kawakami Y. Targeting FSTL1 Prevents Tumor Bone Metastasis
178. Chaly Y, Hostager B, Smith S, Hirsch R. Follistatin-like protein 1 and its role in inflammation and and Consequent Immune Dysfunction. Cancer Res. 2013;73(20):6185-6193. doi:10.1158/0008-
inflammatory diseases. Immunol Res. 2014;59(1-3):266-272. doi:10.1007/s12026-014-8526-z. 5472.CAN-13-1364.
179. Ehara Y, Sakurai D, Tsuchiya N, et al. Follistatin-related protein gene (FRP) is expressed in the 198. Chan QKY, Ngan HYS, Ip PPC, Liu VWS, Xue WC, Cheung ANY. Tumor suppressor effect of
synovial tissues of rheumatoid arthritis, but its polymorphisms are not associated with genetic follistatin-like 1 in ovarian and endometrial carcinogenesis: a differential expression and
susceptibility. Clin Exp Rheumatol. 2004;22(6):707-712. functional analysis. Carcinogenesis. 2009;30(1):114-121. doi:10.1093/carcin/bgn215.
180. Clutter SD, Wilson DC, Marinov AD, Hirsch R. Follistatin-Like Protein 1 Promotes Arthritis by Up- 199. Ciarmela P, Florio P, Sigurdardottir M, et al. Follistatin-related gene expression, but not follistatin
Regulating IFN-γ. J Immunol. 2009;182(1):234-239. doi:10.4049/jimmunol.182.1.234. expression, is decreased in human endometrial adenocarcinoma. Eur J Endocrinol Eur Fed
181. Gorelik M, Fall N, Altaye M, et al. Follistatin-like protein 1 and the ferritin/erythrocyte Endocr Soc. 2004;151(2):251-257.
sedimentation rate ratio are potential biomarkers for dysregulated gene expression and 200. Reddy SP, Britto R, Vinnakota K, et al. Novel glioblastoma markers with diagnostic and prognostic
macrophage activation syndrome in systemic juvenile idiopathic arthritis. J Rheumatol. value identified through transcriptome analysis. Clin Cancer Res Off J Am Assoc Cancer Res.
2013;40(7):1191-1199. doi:10.3899/jrheum.121131. 2008;14(10):2978-2987. doi:10.1158/1078-0432.CCR-07-4821.
182. Campfield BT, Nolder CL, Marinov A, et al. Follistatin-like protein 1 is a critical mediator of 201. Trojan L, Schaaf A, Steidler A, et al. Identification of metastasis-associated genes in prostate
experimental Lyme arthritis and the humoral response to Borrelia burgdorferi infection. Microb cancer by genetic profiling of human prostate cancer cell lines. Anticancer Res. 2005;25(1A):183-
Pathog. 2014;73:70-79. doi:10.1016/j.micpath.2014.04.005. 191.
183. Fan N, Sun H, Wang Y, et al. Follistatin-Like 1: A Potential Mediator of Inflammation in Obesity. 202. Sepporta MV, Tumminello FM, Flandina C, et al. Follistatin as potential therapeutic target in
Mediators Inflamm. 2013;2013:e752519. doi:10.1155/2013/752519. prostate cancer. Target Oncol. 2013;8(4):215-223. doi:10.1007/s11523-013-0268-7.
184. Tanaka M, Ozaki S, Kawabata D, et al. Potential preventive effects of follistatin‐related protein/ 203. Kudo-Saito C. FSTL1 promotes bone metastasis by causing immune dysfunction. Oncoimmunology.
TSC‐36 on joint destruction and antagonistic modulation of its autoantibodies in rheumatoid 2013;2(11). doi:10.4161/onci.26528.
arthritis. Int Immunol. 2003;15(1):71-77. doi:10.1093/intimm/dxg005. 204. Tan X, Zhai Y, Chang W, et al. Global analysis of metastasis-associated gene expression in primary
185. Kawabata D, Tanaka M, Fujii T, et al. Ameliorative effects of follistatin-related protein/TSC-36/ cultures from clinical specimens of clear-cell renal-cell carcinoma. Int J Cancer. 2008;123(5):1080-
FSTL1 on joint inflammation in a mouse model of arthritis. Arthritis Rheum. 2004;50(2):660-668. 1088. doi:10.1002/ijc.23637.
doi:10.1002/art.20023. 205. Liu Y, Han X, Yu Y, et al. A genetic polymorphism affects the risk and prognosis of renal cell
186. Le Luduec JB, Condamine T, Louvet C, et al. An Immunomodulatory Role for Follistatin-Like 1 carcinoma: association with follistatin-like protein 1 expression. Sci Rep. 2016;6:26689.
in Heart Allograft Transplantation. Am J Transplant. 2008;8(11):2297-2306. doi:10.1111/j.1600- doi:10.1038/srep26689.
6143.2008.02398.x. 206. Mashimo J ’ichi, Maniwa R, Sugino H, Nose K. Decrease in the expression of a novel TGF β1-
187. Adams DC, Karolak MJ, Larman BW, Liaw L, Nolin JD, Oxburgh L. Follistatin-like 1 regulates renal inducible and ras-recision gene, TSC-36, in human cancer cells. Cancer Lett. 1997;113(1):213-

28 29
CHAPTER 2

Endothelial Follistatin-like 1 regulates


the postnatal development of
the pulmonary vasculature
by modulating BMP/Smad signaling

Navessa P. Tania, Harm Maarsingh, I. Sophie T. Bos, Andrea Mattiotti,


Stuti Prakash, Wim Timens, Quinn D. Gunst, Luis J Jimenez-Borreguero,
Martina Schmidt, Maurice J.B. van den Hoff, Reinoud Gosens
Endothelial follistatin-like-1 regulates the postnatal development of the pulmonary Abstract
vasculature by modulating BMP/Smad signaling.
BMP signaling regulates vascular smooth muscle maturation, endothelial cell
proliferation and tube formation. The endogenous BMP antagonist Follistatin-like
1 (Fstl1) is highly expressed in pulmonary vascular endothelium of the developing
mouse lung, suggesting a role in pulmonary vascular formation and vascular
homeostasis. The aim of this study was to investigate the role of Fstl1 in the pulmonary
Navessa P. Tania1, Harm Maarsingh2, I. Sophie T. Bos1, Andrea Mattiotti3, Stuti 2
vascular endothelium. To this aim, Fstl1 was conditionally deleted from endothelial
Prakash3, Wim Timens4, Quinn D. Gunst3, Luis J Jimenez-Borreguero5, Martina and endothelial-derived cells using Tie2-credrivenFstl1-KO mice (Fstl1-eKO mice).
Schmidt1, Maurice J.B. van den Hoff3, Reinoud Gosens1 Endothelial-specific Fstl1 deletion was postnatally lethal, as ~70% of Fstl1-eKO mice
died at 3 weeks after birth. Deletion of Fstl1 from endothelium resulted in a reduction
of right ventricular output at 3 weeks after birth compared to controls. This was
associated with pulmonary vascular remodeling, as the percentage of actin positive
small pulmonary vessels was increased at 3 weeks in Fstl1-eKO mice compared to
1
University of Groningen, Department of Molecular Pharmacology, Groningen
controls. Endothelial deletion of Fstl1 resulted in activation of Smad1/5/8 signaling
Research Institute for Asthma and COPD, Groningen, The Netherlands.
and increased BMP/Smad-regulated gene expression of Jagged1, Endoglin, and
2
Palm Beach Atlantic University, Department of Pharmaceutical Sciences, Lloyd L.
Gata2 at 1 week after birth compared to controls. In addition, potent vasoconstrictor
Gregory School of Pharmacy, West Palm Beach, Florida, USA.
Endothelin-1, the expression of which is driven by Gata2, was increased in
3
Academic Medical Center, Department of Anatomy, Embryology and Physiology,
expression,both on the mRNA and protein level,at 1 week after birth compared to
Amsterdam,The Netherlands.
controls. At 3 weeks, Jagged1 was reduced in the Fstl1-eKO mice whereas Endoglin
4
University of Groningen, University Medical Center Groningen, Department of
Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, and Endothelin-1 were unchanged.In conclusion, loss of endothelial Fstl1 in the
Groningen, The Netherlands. lung is associated with elevated BMP-regulated genes, impaired small pulmonary
5
Centro Nacional de Investigaciones Cardiovasculares & Hospital de La Princesa, vascular remodeling and decreased right ventricular output.
Madrid, Spain.

32 33
Introduction Materials and Methods

Pulmonary vascular development and maturation is a tightly controlledprocess Antibodies and reagents
that isorchestrated by multiple signaling pathways, including Bone Morphogenetic
Polyclonal rabbit anti-Fstl1 (#HPA035251), polyclonal rabbit anti-Jagged1
Protein (BMP),which control spatio-temporal gene expression patterns. BMP
(#HPA021555),and monoclonal mouse anti-β-actin (#A5441) were purchased
signaling is important in embryonicvascular developmentand adult vascular
from Sigma Aldrich (St. Louis, MO, USA). Polyclonal rabbit anti-α-smooth muscle
homeostasis.1,2Dysregulation of BMP signaling has been strongly associated with
actin(α-SMA,#AB5694) and polyclonal rabbit anti-CD31 (#AB28364)were obtained 2
the pathogenesis of hereditary vascular diseases, including familial pulmonary
from Abcam(Cambridge, UK). Polyclonal goat anti-mouse Endoglin (#AF1320) was
arterial hypertension (PAH), hereditary hemorrhagic telangiectasia, atherosclerosis
procured from R&D Systems (Oxford, UK). Polyclonal rabbit anti-phospho-Smad1/5/8
and cerebral cavernous malformation.3–11BMP signaling regulates vascular smooth
(#AB3848), monoclonal mouse anti-Troponin I (TnI) (#MAB16910)were obtained
muscle maturation, endothelial cell proliferation and tube formation.9,12 Tight spatio-
from Millipore (Molsheim, France) andpolyclonal rabbit anti-Smad1(#9743) from
temporal control of BMP signaling and biological availability is therefore crucial for
Cell Signaling (Danvers, MA, USA). Horseradish peroxidase (HRP)-conjugated donkey
the normal development of vascular structure and function, including in the lung.
anti-rabbit IgG (#711-035-152), donkey anti-goat IgG (#705-035-003), and donkey
BMP ligands elicittheir activity by binding to typeI BMP receptors (ALK1,
anti-mouse IgG (#715-035-150) were purchased from Jackson Immunoresearch
2,3,and 6) and by recruitingtype II BMP receptors (BMPRII, ACVRIIA, and ACVRIIB).
(West Grove, PA, USA). Alexa Fluor 647 donkey anti-mouse IgG (#A-31571) was
After forming a heterotetrameric complex, the signal propagates to the nucleus
obtained from Thermo Fisher Scientific (Waltham, MA, USA).All other chemicals
byspecifically phosphorylating the Smad proteinsSmad1/5/8. Phosphorylated
were of analytical grade.
Smad1/5/8 proteins form a complex with Smad4,which results in accumulation of
this complex in the nucleus. This directly and indirectly influences transcription of
Tie-2cre mediated endothelial-specificdeletion of Fstl1
target genes, among which Endoglin, Gata2 and Jagged1.13
Follistatin-like 1 (Fstl1) is anendogenous BMP antagonist that is highly expressed Conditional deletion of Fstl1 from vascular endothelial and endothelial-derived
in pulmonary vascular endothelium of thedeveloping mouse lung, suggesting a cells was achievedby crossing floxed homozygousFstl1fl/flmice with double
role in pulmonary vasculature development.14,15Fstl1 is important for normal lung heterozygousmice carrying one Fstl1 knockoutallele and the Tie2-cre cassette(Fstl1WT/
development.In fact, multiple organ defects were observed in whole-body knockout
KO Tie2-cre
)on a FVB background.15,17Within this cross endothelial specific knockout
of Fstl1 (Fstl1-KO).15,16Fstl1-KOmice showed increased thickness of alveolar walls and mice(Fstl1fl/KO Tie2-cre)and littermate controls(Fstl1fl/WT Tie2-cre, Fstl1fl/KO, and Fstl1fl/WT)
increased numbers of immature cuboidal alveolar epithelial cells in the lung.15,16 were generated.Fstl1fl/KO Tie2-cre mice are denoted as Fstl1-eKO and their littermate
In addition, Fstl1-KOmice exhibited impaired tracheal cartilage formation, alveolar controls are denoted as controls. The breeding lines were maintained in the animal
maturation and lung morphogenesis, resulting in respiratory failure and in postnatal facility of the University of Amsterdam. All experimental procedures complied with
lethality in mice.15,16Major defects in cardiac and skeletal development were institutional and national ethical guidelines regarding animal experimentation.
alsoreported in Fstl1-KOmice.15,16 Mice were sacrificed at 1 and 3 weeks after birth (1 week: n=12 Fstl1-eKO mice
Given the complex nature of BMP signaling and the predominant endothelial and n=16 controls; 3 weeks: n=11Fstl1-eKO miceand n=30 controls). The lung
expression of the BMP antagonist Fstl1 in the developing lung, this study aimed to tissue was collected. Lung lobes (right superior, middle, and inferior lobes) were
explore therole of endothelial Fstl1 signaling in pulmonary vascular development inflated with 50% Tissue-Tek (Sakura Finetek;Alphen-aan-den-Rijn, the Netherlands)
using Tie2-cremediated endothelial-specific deletion of Fstl1 in vivo. Specifically, in 0.9% NaCl (Braun; Kronberg, Germany) and fixed with 10% v/v formalin for
we aimed to investigate the impact of Fstl1 deletion on the morphology of the paraffin-embedded sections. The non-inflated lobes (left and post caval lobes)
pulmonary vasculature, on gene expression associated with BMP signaling, and its were snap-frozen in liquid nitrogen for mRNA and protein analysis. The pups were
implication on pulmonary function. genotyped using PCR with Fstl1 and cre-recombinase primer sets. Fstl1forward
(5’-GCCAGAATCCCACTCCATCG- 3’); Fstl1reverse (5’-TCGGAGCCTGGTGATAAGCG-3’);
cre-recombinase forward (5’-GGTTCGCAAGAACCTGATGGACAT-3’); cre-recombinase
reverse (5’-GCTAGAGCCTGTTTTGCACGTTCA-3’).

In situ hybridization
In situ hybridization was performed as previously described.18In short, lung sections
were deparaffinized and rehydrated in a graded series of alcohol, followed by 15

34 35
min incubation at 37°C in 10 mg/ml proteinase K dissolved in PBS. The sections were were expressed in arbitrary unitsas ratio ofthe starting concentration (N0) of each
post-fixed for 10 min in 4% paraformaldehyde (PFA) and 0.2% glutaraldehyde in geneofinterest corrected to the geometric mean of the N0value of two reference
PBS, followed by rinsing in PBS. Pre-hybridization was done for at least 1 hr at 70°C genes (B2m and Hprt). Comparing the expression levels of the genes of interest
in hybridization mix (50% formamide, 5xSSC (20xSSC; 3 M NaCl, 0.3 M tri-sodium between the three non-conditional knockout groups (Fstl1fl/WT Tie2-cre, Fstl1KO/fl, or
citrate, pH 4.5)), 1% blocking solution, 5 mM EDTA, 0.1% 3-[(3-Cholamidopropyl) Fstl1WT/fl mice), did not reveal significant differences, allowing pooling of the data
dimethylammonio]-1-propanesulfonate (Sigma Aldrich; St. Louis, MO, USA), 0.5 mg/ and using them as age-matched controls.
ml heparin (BD Biosciences; Erembodegem, Belgium), and 1 mg/ml yeast total RNA
Supplementary Table 1. Primers used for qRT-PCR analysis. 2
(Roche Applied Science; Penzberg, Germany). A digoxigenin (DIG)-labeled probe
(1 ng/ml) was added to the hybridization mix. Probes specific to Fstl1 were used. Gene Primer sequence (5’ 3’) NCBI accession number
After overnight hybridization, the sections were rinsed with 2xSSC, followed by Fstl1 Forward TCGCTGTGTCTGTTCCTGTGGC NM_008047.5
two washings(50% formamide, 2xSSC, pH 4.5) at 65°C, and rinsing with TNT (0.1 M Reverse TTCTGCTGTGCCCTGGTGCTTC
Tris-HCl, pH  7.5, 0.15 M NaCl, 0.05% Tween-20) at room temperature. The sections Eng Forward GCACCTTGTCCCAGGAAGTC NM_001146350.1
Reverse GGAGGCTTGGGATACTCACG
were incubated for 1 hr in MABT-block (100 mM maleic acid, 150 mM NaCl, pH 7.4,
Jag1 Forward CCAGTGTCAGAATGACGCCT NM_013822.5
0.05% Tween-20, 2% blocking solution), followed by 2 hr incubation in MABT-block
Reverse AGTGACCCCCATTCAAGCAG
containing 100 mU/ml alkaline phosphatase-conjugated anti-DIG Fab fragments Gata2 Forward CCCCTATCCCGTGAATCCG NM_008090.5
(Sigma Aldrich; St. Louis, MO, USA). After rinsing in TNT and subsequently in NTM Reverse GGTCCACTACTGTGTCTTGGG
(100 mM Tris, pH 9.0, 100 mM NaCl, 50 mM MgCl2), probe binding was visualized Edn1 Forward GGCCCAAAGTACCATGCAGA NM_010104.3
using nitro blue tetrazolium chloride and 5-bromo-4-chloro-3-indolyl-phosphate Reverse GATGGCCTCCAACCTTCGTA
(Roche Applied Science; Penzberg, Germany). The sections were dehydrated in B2m Forward ATGGGAAGCCGAACATACTG NM_009735.3
a graded alcohol series, rinsed in xylene, and embedded in Entellan (Millipore; Reverse CAGTCTCAGTGGGGGTGAAT
Molsheim, France). Images were captured using a Leica DFC320 camera mounted Hprt Forward AGGCCAGACTTTGTTGGATTTGA NM_013556.2
on an AxioPhot microscope (Zeiss; Oberkochen, Germany). Reverse ACTGGCAACATCAACAGGACTC
Abbreviations: Fstl1, Follistatin-like 1; Eng, Endoglin; Jag1, Jagged1; Gata2, GATA
Pulmonary physiological measurement binding protein 2; Edn1, Endothelin-1; B2m,b2-microglobulin; and Hprt, hypoxanthine
Pulmonary physiological functions were measured in miceat 1 week (n=4 Fstl1-eKO guanine phosphoribosyl transferase.
mice and n=14 controls)and 3 weeks (n=8 Fstl1-eKO mice and n=19 controls) after
birth using transthoracic echocardiography as previously described.19
Western blot
RNA isolation and real time quantitative PCR Total protein was extracted from lung tissue (left lobe) using RIPA lysis buffer
Total RNA was extracted from lung tissue (post caval lobe) using the NucleoSpin® RNA (RIPA lysis buffer (65 mM Tris, 155 mM NaCl, 1% Igepal CA-630, 0.25% sodium
isolation kit according to the manufacturer’s instruction (Macherey Nagel;Düren, deoxycholate, 1 mM EDTA, pH 7.4) supplemented with protease inhibitors (1 µg/ml
Germany). The total RNA concentration was determined using theNanoDrop® aprotinin, 1 µg/ml leupeptin, 1 µg/ml pepstatin A, 1 mM Na3VO4, 1 mM NaF, 1 mM
ND1000 spectrophotometer (Thermo Fisher Scientific;Waltham, MA, USA). Equal β-glycerophosphate). Equal amounts of protein lysate were subjected to SDS PAGE
amounts of total RNA were reverse transcribed using the Reverse Transcription electrophoresis and transferred to nitrocellulose or polyvinylidene difluoride (PVDF)
System (Promega; Madison, WI, USA) to generate cDNA. Diluted cDNA was mixed membranes to evaluate the expression level of non-phosphorylated (β-actin, Jagged1,
with FastStart Universal SYBR Green Master Mix (Roche Applied Science; Penzberg, Endoglin, total Smad1) , and phosphorylated (pSmad1/5/8)proteins, respectively.
Germany) and gene of interest primer sets (Biolegio;Nijmegen, the Netherlands). Proteins ofinterest were detected using primary antibodies for overnight at 4°C in in
Primer sequences are listed in Supplementary Table 1. Real-time quantitative PCR TBST (50 mM Tris-HCl, 150 mM NaCl, 0.05% (w/v) Tween-20, pH 7.4). The following
was performed using the Illumina Eco Personal qPCR System (Westburg; Leusden, day, membranes were incubated with HRP-conjugated secondary antibodies for 2 hr
The Netherlands). The qPCR reaction was started by denaturation at 95°C for 15 at room temperature. Protein bands were subsequently visualized using enhanced
minutes followed by 45 cycles of denaturation at 94°C for 30 s, annealing at 59°C chemiluminescence substrate(Perkin Elmer; Groningen, the Netherlands)using the
for 30 s and elongation at 72°C for 30 s. Final elongation was for 5 minutes at 72 °C. G-box gel documentation system (Syngene; Cambridge, UK). Protein band intensities
Real time PCR data was analyzed using LinRegPCR software version 2013.1.20 Data were quantified using Image Studio Lite version 5. Data are presented as ratio of non-

36 37
phosphorylated protein band intensity corrected to b-actinas a reference protein. in KP-mounting medium (Klinipath BV;Duiven, the Netherlands). Images were
For phosphorylated proteins, data are presented as ratio of phosphorylated protein captured using Cell^D imaging software using a light microscope (Olympus BX41;
corrected to total protein. Zoeterwoude, the Netherlands).

Data analysis
Immunostaining Data are presented as medians per genotype-group except otherwise stated.
Paraffin-embedded lung tissues were sectioned at 5 µm thickness and stained To determine the normality of data distribution, a Shapiro-Wilk normality test 2
for α-SMA (Abcam; Cambridge, UK), which was visualized by staining with HRP- was performed prior to further statistical analysis. The statistical significance of
conjugated secondary antibody and diaminobenzidine as a substrate (Sigma differences of normally distributed data was performed using an independent
Aldrich; St. Louis, MO, USA). To determine the actin content in large vessels (>50 samples 2-tailedt-test for comparing 2 groups or a two-way ANOVA followed by a
µm in diameter),21 the α-SMA staining surrounding muscular and elastic vessels post hocTukey multiple comparisons test for comparing more than 2 groups. For non-
was digitally captured in two lung tissue sections per animal and quantified using Gaussian distributed data, the statistical significance of differences was determined
Image J (National Institute of Health, USA) in a blinded fashion. Data are expressed usinga non-parametric Mann-Whitney U test for comparing 2 groups or non-
as ratio of the actin positive area over the square of the length of the tunica intima. parametric one-way ANOVA with apost hoc Kruskal-Wallis multiple comparisons test
To determine the total number of small vessels (<50 µm in diameter),21CD31 staining for comparing more than 2 groups. The Bonferroni correction was used to correct for
was performed in serial sections to stain for endothelial cells. Data are expressed multiple testing. Differences were considered to be statistically significant at p<0.05.
as the total number of CD31-positive small vessels corrected to lung surface area
(cm2). Furthermore, the number of actin positive small vessels as well as the total
number of CD31-positive small vessels was quantified in two lung tissue sections Results
per animal, in a blinded fashion. Data are expressed as the number of actin positive
small vessels corrected to the total number of CD31-positive small vessels. To Loss of Fstl1 from endothelial cells is postnatally lethal in mice.
evaluate muscularization of the right heart, paraffin-embedded heart tissues were We and others have previously demonstrated that the homozygote global Fstl1
sectioned at 10 µm and stained using the myocardial marker TnI which was visualized knockout mice die at birth due to respiratory distress. Here, we generated a mouse
by staining with Alexa Fluor 647-conjugated secondary antibody. The images were line in which Fstl1 is conditionally impaired in endothelial and endothelial-derived
digitally captured in a blinded manner using a fluorescent microscope (Leica Dm6000; cells using Tie2-cre targeted gene deletion, which will be referred to as Fstl1-eKO.
Wetzlar, Germany). The right ventricular free wall was outlined in each section and Fstl1-eKO knockout pups were born alive at the expected Mendelian ratio (25%).
the mean fluorescence intensity of the outlined structure was measured using 3D However, ~70% of Fstl1-eKO mice had died by 3 weeks after birth whereas all their
Amira software (Version 5.4.3). littermate controls, Fstl1fl/WT Tie2-cre, Fstl1KO/fl, and Fstl1WT/fl mice survived(Figure 1A). In
contrast to Fstl1-KO mice, which display abnormal tracheal cartilage formation and
Endothelin-1 enzyme-linked immunosorbent assays (ELISA) thickening of alveolar septa, Fstl1-eKO mice had macroscopically normal tracheal
The Endothelin-1 protein concentration in lung homogenates was determined using cartilage formation and normal alveolarization (Figure S1). In view of the postnatal
ELISA according to the manufacturer’s protocol (R&D system;Oxford,UK) in duplicate. lethality of Fstl1-eKO mice and the marked differences with the globalFstl1-KOmice,
The sample absorbance was determined at 450 nm and at 570 nm to correct for we quantified Fstl1 mRNA and protein in the lungs of these animals. Fstl1 was highly
optical imperfections using Gen5an software using a plate reader (BioTek; Winooski, expressed in the control mice at 1 week after birth and significantly declined 3 weeks
VT, USA). The lower and upper detection limits for Endothelin-1 were 0.39 pg/ml after birth (mRNA p<0.005; protein p<0.05; Figure 1B and 1C), suggesting Fstl1 is
and 25 pg/ml, respectively. crucial in the early stages of postnatal lung development. The expression of Fstl1
mRNA and protein in lung homogenates of Fstl1-eKO mice were significantly reduced
Hematoxylin and eosin staining compared to age-matched controls at 1 week after birth (mRNA p<0.005; protein
Paraffin-embedded lung tissue sections of 5 µm thick were deparaffinized and p<0.05; Figure 1B and 1C), whereas at 3 weeks after birth the expression of Fstl1
rehydrated in a graded series of alcohol, followed by hematoxylin staining. After mRNA and protein were not significantly different in Fstl1-eKO mice compared to
washing with flowing water, sections were counterstained with eosin. The sections age-matched controls (Figure 1B and 1C). In situ hybridization on sections of control
were dehydrated in a graded series of alcohol, rinsed in xylene, and embedded mice showed expression of Fstl1 mRNA both in blood vessels and lung parenchyma
at 1 week. At 3 weeks after birth the pattern of expression was not different, though

38 39
the staining intensity was markedly less, which is in line with the qPCR findings
(Figure 1D). In the Fstl1-eKO mice, the expression pattern of Fstl1 mRNA was similar Right ventricular output is reduced in Fstl1-eKO mice
to the control mice, except that Fstl1 mRNA was not or hardly detectable in the To shed light on the cause of neonatal lethality of Fstl1-eKO mice, we determined the
endothelium of the blood vesselsboth at 1 and 3 weeks after birth (Figure 1D). physiological function of the pulmonary vasculature using echocardiography. During
. normal postnatal development, a significant increase in right ventricular output was
observed at 3 weeks compared to 1 week as shown by the pulmonary valve (PV) and
velocity-time integral (VTI) parameters (p<0.05; Figure 2A). A significant reduction 2
in right ventricular output in Fstl1-eKO mice was observed at 3 weeks compared to
age-matched controls (p<0.05; Figure 2A). Other physiological parameters, including
PV peak gradient, PV mean gradient, PV peak velocity, and PV mean velocity, were
significantly increased at 3 weeks compared to 1 week after birth both in control
and in Fstl1-eKO mice (p<0.05; Figure 2B-2E). No differences between Fstl1-eKOmice
and age-matched controls were observed for these parameters (Figure 2B-2E). A
validated non-invasive method to assess pulmonary vascular resistance22,23 was
performed by measuring pulmonary acceleration time (PAT) corrected for the
pulmonary ejection time (ET). There was no significant difference in pulmonary
vascular resistance during normal postnatal lung development as reflected by the
ratio of PAT/ET in control mice at 3 weeks compared to 1 week after birth (Figure 2F).
A significant change was also not observed between Fstl1-eKO mice compared to
age-matched controls either at 1 week or 3 weeks after birth (Figure 2F). Moreover,
we evaluated the muscularization of the right ventricle using myocardial marker TnI,
as an indicative of right ventricle hypertrophy by measuring the volume of the right
ventricle. There were no changes in the right ventricle volume at 3 weeks compared
to 1 week after birth in the control mice (Figure 2G). At 3 weeks, there was a trend
towards increased RV volume in Fstl1-eKO mice compared to age-matched controls
(Figure 2G). A significant increase in RV volume was observed in Fstl1-eKO mice at 3
weeks compared to 1 week after birth (p=0.005; Figure 2G).

Figure 1. Endothelial deletion of Fstl1 in mice is postnatally lethal. (A) Kaplan Meijer survival
curve of Fstl1-eKO mice compared to controls. ~70% of Fstl1-eKO mice died at 3 weeks after
birth compared to controls. (B) Gene expression analysis of Fstl1 in lung homogenates using
quantitative real time PCR. Horizontal line represents the median of 16-30 mice per group.
(C) Immunoblot analysis of Fstl1 protein in lung homogenates. Data are expressed as means
± SEM of the ratio of Fstl1 protein corrected to β-actin as a reference protein from 4-9 mice
per group. (D) Representative images of Fstl1 in situ hybridization in lung tissue of Fstl1-
eKO mice compared to controls. Red arrowheads point to the absence or presence of Fstl1
probes. AW indicates an airway and V indicates a blood vessel. Each data point represents
an individual animal. *p<0.05; **p<0.01; ***p<0.005 compared to the indicated group; ns
= not significant.

40 41
Pulmonary vascular remodeling in Fstl1-eKO mice
In view of these results, we examined the effect of endothelial deletion of Fstl1 on
pulmonary vascular phenotypes and quantified the morphological changes and
actin content in large and small pulmonary vessels. During normal development, the
thickness of the large muscular arteries was unaltered in lung tissue of control mice
at 3 weeks compared to 1 week after birth(Figure 3A), whereas the actin content
was reduced in large elastic (p<0.005) and muscular arteries (p<0.005) in lung tissue 2
of control mice at 3 weeks compared to 1 week after birth (Figure 3B and 3C).In
Fstl1-eKO mice, the thickness and the actin content in large muscular arteries were
unaltered at 3 weeks compared to 1 week after birth (Figure 3A and 3C), whereas the
actin content in large elastic arteries was significantly reduced at 3 weeks compared
to 1 week after birth (p< 0.005; Figure 3B). No significant differences were observed
in the thickness of the large muscular arteries (Figure 3A) and in the actin content in
large elastic and muscular arteries in lung tissue of Fstl1-eKO mice compared to age-
matched controls at either 1 or 3 weeks after birth (Figure 3B and 3C). Reduction in
actin content at 3 weeks after birth seems to be limited to vascular smooth muscle
as there were no temporal changes in the actin content in the airway smooth muscle
bundlesand there was no observed difference between Fstl1-eKO mice compared to
age-matched controls(Figure 3D).
We also explored the number of small blood vessels in lung tissue as these
may contribute to an important extent to changes in pulmonary blood pressure.
In normal development, no significant temporal change in theCD31 positivetotal
number of vessels was observed, whereas the percentage of actin positive vessels
was significantly reduced at 3 weeks compared to 1 week after birth (p<0.01; Figure
3E and 3F). Although the CD31 positive total number of vessels was not significantly
different between Fstl1-eKO mice compared to age-matched controls (Figure 3E),
the percentage of actin positive small pulmonary vessels was significantly higher in
Fstl1-eKO mice at 3 weeks after birth compared to age-matched controls(p<0.05;
Figure 3F).

Figure 2. Right ventricular output is reduced in Fstl1-eKOmice. Pulmonary functions were


measured using echocardiograph. (A) Pulmonary valve (PV)-velocity time integral (VTI) was
significantly decreased in Fstl1-eKO mice compared to age-matched controls.(B) PV peak
gradient, (C) PV mean gradient, (D) PV peak velocity, (E) PV mean velocity, and (F) the ratio
of Pulmonary Acceleration Time (PAT) corrected by the pulmonary ejection time (ET) in
percentage were unaltered in Fstl1-eKO mice compared to age-matched controls. Data are
expressed as the mean ± SEM of 4-19 mice per group. (G) The right ventricle (RV) volume
of Fstl1-eKO mice was unaltered compared to age-matched controls. A significant increase
in RV volume was observed in Fstl1-eKO mice at 3 weeks compared to 1 week after birth.
Data are expressed as the mean ± SEM of 3 mice per group. *p<0.05; **p<0.01; ***p<0.005
compared to the indicated group; ns = not significant.

42 43
Increased BMP/Smad signaling in the lung of Fstl1-eKO mice
To unravel whether endothelial Fstl1 modulates BMP signaling during postnatal
development, we investigated the activity of BMP/Smad signaling in lung
homogenates using pSmad1/5/8 antibodies. During normal postnatal development,
Smad1/5/8 phosphorylation was higher at 3 weeks compared to 1 week after birth
(p<0.05; Figure 4A). In addition, we found an increased levels ofpSmad1/5/8in lung
homogenates of Fstl1-eKO compared to age-matched controls at 1 week after birth 2
(p<0.05; Figure 4A), whereas,mthe level of pSmad1/5/8at 3 weeks was unaltered in
Fstl1-eKO mice compared to age-matched controls (Figure 4A).
To study the role of BMP signaling further, we selected previously identified
pSmad1/5/8 target genes which relate to vascular remodeling24: Id3, Epas1, Zeb2,
Zfp423, Ephb4, Klf4, Flt1, Jag1, Eng, Gata2, Vegf. Of these, we found that the
expression of Jagged1, Endoglin, and Gata2 was altered in Fstl1-eKO mice compared
to age-matched controls with a nominal p value <0.05, whereas the expression level
of the other genes was not different. Therefore, Jagged1, Endoglin, and Gata2 were
included for further analysis. Consistent with the increased Smad1/5/8 activation,
Jagged1 and Endoglin mRNA and protein were increased in lung tissue of control
mice at 3 weeks compared to 1 week after birth (Jagged1 mRNA p<0.05; protein
p<0.05; Figure 4B and 4D; Endoglin protein p<0.01; Figure 4C and 4E). In line with
increased phosphorylation of Smad1/5/8, the expression of Jagged1 and Endoglin
mRNA and protein was also increased in lung homogenates of 1 week Fstl1-eKO
mice compared to age-matched controls (Jagged1 mRNA p<0.05; protein p<0.05;
Figure 4B and 4D; Endoglin mRNA p<0.05; protein p<0.01; Figure 4C and 4E). At 3
weeks, Jagged1 mRNA (p<0.01) and protein (p<0.05) were reduced in the Fstl1-eKO
mice compared to age-matched controls (Figure 4B and 4D), whereas Endoglin was
not different compared to age-matched controls (Figure 4C and 4E).
The mRNA expression of the transcription factor Gata2 was higher in the lung
homogenates of control mice at 3 weeks compared to 1 week after birth (p<0.01;
Figure 3. Number of actin-positive small pulmonary vessels is increased in the lung tissue Figure 5A). Accordingly, the level of mRNA and protein expression ofthe Gata2 target
of Fstl1-eKOmice at 3 weeks. (A) The thickness of large muscular arteries in lung tissue was gene Endothelin-1, a potent vasoconstrictor, was also increased in lung homogenates
quantified and expressed as ratio of total area of tunica media over the square of the length of control mice at 3 weeks compared to 1 week after birth (mRNA p<0.005; protein
of the tunica intima. Horizontal lines represent the median of n = 9-16 vessels from N= 6-8
mice per group. (B) The α-SMA content in large elastic arteries and (C) in large muscular p<0.05; Figure 5B and 5C). Gata2 mRNA and Endothelin-1 mRNA and protein levels
arteries in lung tissue was quantified and expressed as ratio of the actin positive area over were increased in lung homogenates of Fstl1-eKO mice compared to age-matched
the square of the length of tunica intima. Horizontal lines represent the median of n= 9-26 controls at 1 week after birth (Endothelin-1 mRNA p< 0.05; protein p<0.01; Figure
vessels per group from N= 5-12 mice per group. (D) The α-SMA content in airway smooth
muscle bundles surrounding the airway in lung tissue was quantified and expressed as ratio 5B and 5C). These levels did not further increase at 3 weeks after birth and were no
of the actin positive area over the square of the length of basement membrane. Horizontal longer different compared to age-matched controls (Figure 5A-C).
lines represent the median of n=9-34 vessels per group from N= 5-13 mice per group. (E)
Total number of vessels in lung tissue was quantified using CD31 staining and expressed as
ratio of total vessel number/lung surface area (cm2). Horizontal lines represent the median
of the vessel number/cm2 of N= 7-12 mice per group. (F) The percentage of actin positive
small pulmonary vessels in lung tissue was quantified and expressed as percentage of actin
positive small vessel number/total vessel number. Horizontal lines represent the median of
the percentage of actin positive vessels of N= 7-12 mice per group. Representative images
of tissue sections taken at 3 weeks after birth were shown. AW indicates an airway and
V indicates a blood vessel. Scale bars represent 100 µm. Magnification 200x. *p<0.05;
**p<0.01; ***p<0.005 compared to the indicated group.

44 45
2

Figure 4. Increased activation of Smad1/5/8 and Smad1/5/8-regulated genes in the lung Figure 5. Increased Endothelin-1 in the lung of Fstl1-eKO mice. (A) Pulmonary mRNA
of Fstl1-eKO mice. (A) Immunoblot analysis of pSmad1/5/8 in lung homogenates. Data are expression of Gata2 transcription factor and (B) Endothelin-1 in the lung homogenates. Data
expressed as means ± SEM of the ratios of pSmad1/5/8 corrected to total Smad1 of 4-9 are expressed as starting concentration N0 in arbitrary units corrected toB2m and Hprt as
mice per group. Pulmonary expression of the pSmad1/5/8-regulated genes, Jagged1 (B) reference genes. (C) Endothelin-1 protein concentration in lung homogenates is expressed
and Endoglin (C) in lung homogenates. Data are expressed as starting concentration N0 in as Endothelin-1 concentration in pg/ml per 1 µg protein lysates. Each data points represent
arbitrary units corrected to B2m and Hprt as reference genes. Horizontal lines represent an individual animal. Horizontal line represents the median of Endothelin-1 concentration
medians of 11-30 mice per group. (D) Immunoblot of Jagged1 protein in lung homogenates of 8-12 mice per group. *p<0.05; **p<0.01; ***p<0.005 compared to the indicated group.
and its quantification. Data are expressed as means ± SEM of the ratios of Jagged1 over
the reference protein β-actin of 4-9 mice per group. (D) Immunoblot of Endoglin in lung
homogenates and its quantification. Data are expressed as means ± SEM of the ratios of
Endoglin over the reference protein β-actin of 4-9 mice per group. *p<0.05; **p<0.01;
***p<0.005 compared to the indicated group.

46 47
Discussion of Fstl1 inhibit BMP signaling and its regulated genes, Gata2, Jagged1 and Endoglin,
indicating that modulation of BMP signaling by Fstl1 is important in early stages of
Proper formation and maturation of the pulmonary vasculature is essential to postnatal lung development. On the other hand, low levels of Fstl1 in later stages
support normal lung development and function.25 BMP signaling is crucial in dynamic of development lead to increased BMP-mediated Smad phosphorylation and its
processes of vessel growth and vessel maturation.9,12,26–29 Maturation of immature regulated genes Gata2, Jagged1, and Endoglin. Endoglin is a type III TGF-β receptor
vessels proceeds according to the developmental steps: (1) stabilization of the which is predominantly expressed in proliferating endothelial cells and triggers
endothelial cell proliferation and vascular remodeling.34–37 The importance of
immature vessels, (2) vessel branching, remodeling, and pruning and (3) vessel 2
specialization.30 Deregulation of molecules involved in vessel growth and maturation Endoglin for normal vascular formation and homeostasis is evident in Endoglin null
leads to vascular abnormalities and dysfunction. (Eng-/-) mice, which exhibit vascular deformities.37–40 Increased Endoglin expression
In this study, the role of endothelial Fstl1 in postnatal maturation of the in Fstl1-eKO mice at the early stages of postnatal pulmonary vascular development
pulmonary vasculature was investigated in vivo using Tie2-cre targeted endothelial- might shift the balance towards endothelial cell migration and proliferation instead
specific deletion. Conditional deletion of Fstl1 from endothelium was postnatally of maturation. This supports the contention that Fstl1-eKO mice have delayed
lethal, and resulted in small pulmonary vessel changes, pointing to a critical role of pulmonary vascular maturation.
endothelial Fstl1 in postnatal lung development. In line with the idea that Fstl1 is a The Notch ligand Jagged1 is an important regulator of cell fate in embryonic
BMP antagonist, phosphorylation of BMP/Smad signaling showed the inverse pattern development41 and vessel fate in pulmonary vascular maturation.10,33 In addition,
of Fstl1 expression level during normal postnatal development. In the early stages of it has a significant role in angiogenesis and vascular homeostasis.42,43 Ablation of
postnatal development, high Fstl1 expression is associated with low phosphorylated Jagged1 in mice is embryonically lethal and leads to vascular abnormalities and
Smad1/5/8 at 1 week after birth whereas at later stages of postnatal development, defects in vascular remodeling.33 The aberrant Jagged1 expression during early
decreased in Fstl1 expression is associated with increased phosphorylation of BMP/ stages of postnatal pulmonary vascular development in Fstl1-eKO mice might lead
Smad signaling in lung tissue of control mice. to deregulation of vascular remodeling and disruption of the vessel specialization
We also demonstrate that during early stages of postnatal development, high programs.
Fstl1 expression coincides with high actin content in the large muscular and elastic BMP/Smad increases expression of the transcription factor Gata2, subsequently
arteries and high percentage of actin positive small pulmonary vessels in lung tissue promoting the expression of the most potent vasoconstrictor Endothelin-1.44–46
of control mice. In later stages of postnatal development, low Fstl1 expression We demonstrate that Endothelin-1 levels are low at the early stages of normal
coexists with low actin content in the large muscular and elastic arteries and low development and increase at the later stages of normal development. We found
percentage of actin positive small pulmonary vessels in lung tissue of control mice. that endothelial deletion of Fstl1 is associated with increased Endothelin-1 mRNA
Recently, it was reported that Fstl1 mediates TGF-β-induced α-SMA expression by and protein expression at the early stages of development in the lung. Increased
antagonizing BMP signaling in lung fibroblasts.31 We speculate that Fstl1 antagonizes Endothelin-1 has been reported to promote proliferation and migration of
BMP signaling to facilitate TGF-β-induced α-SMA expression from vascular smooth endothelial cells and contribute to increased pulmonary vascular resistance and
muscle cells in the early stages of normal vascular development. Reduced Fstl1 levels subsequent right ventricle hypertrophy,47,48 suggesting pivotal roles of Endothelin-1
in later stages of development in turn result in reduction in actin content, suggesting in vessel formation, vascular remodeling, and vascular tone maintenance. Our data
that Fstl1 possibly mediates endothelial-mural cell communication by modulating suggest that Endothelin-1 expression is negatively regulated by Fstl1 via inhibiting
BMP/TGF-β signaling during normal pulmonary vascular development. Smad1/5/8 and the Gata2 transcription factor, presumably by antagonizing BMP
During normal postnatal vascular development, transient reduction of actin signaling. These findings are in line with previous studies demonstrating that
content in pulmonary vessels is normal and an indication of maturation of pulmonary Endothelin-1 expression is regulated by BMP via Smad1/5/8 in endothelial cells.44–46
vessels by reducing contractility of pulmonary arteries.32 Our data indicate that loss Lower right ventricular output and small vascular remodeling were visible
of Fstl1 in endothelium prevents this normal reduction of actin in small pulmonary at 3 weeks after birth, whereas elevation in BMP/Smad phosphorylation and of
vessels and as such delays pulmonary vascular maturation. its downstream targets Jagged1, Endoglin, Gata2, and Endothelin-1 were more
Mechanistically, Fstl1-eKO mice have increased Smad1/5/8 phosphorylation and prominent at 1 week after birth in Fstl1-eKO mice, indicating that molecular changes
expression of pSmad1/5/8-regulated genes in the lung, including Gata2, Endoglin, are more transient than their physiological and morphological consequences.
and Jagged1. Previous studies demonstrated the important role of Jagged1 and As a consequence the functional and morphological differences seen at 3 weeks
Endoglin in vascular specialization and vascular remodeling, respectively, in the later after birth were initiated earlier during postnatal development. Alternatively,
stages of vessel maturation.10,33,34 In early stages of normal development, high levels the activation of BMP/Smad signaling alone may not be sufficient to affect the
pulmonary vasculature structurally and functionally at early stages of development,

48 49
but instead becomes evident at a later time point. Despite of higher percentages
of actin positive small pulmonary vessels and notably decreased right ventricular
output, there were minimum morphological defects of large pulmonary vessels,
normal pulmonary vascular resistance as reflected by the ratio of PAT/ET in Fstl1-eKO
mice and a trend towards right ventricle hypertrophy. This may suggest very early
stages of pulmonary vascular dysfunction in Fstl1-eKO mice. This phenotype is also
commonly observed in patients with PAH, in which molecular changes/defects in
2
BMP signaling manifest early in the disease followed by physiological and structural
changes which are observed at later stages of the disease progression.49 In addition,
our data may imply that the molecular and morphological changes in the lung are
possibly a secondary consequence of the right heart dysfunction as reflected by
reduction in RV output. The morphological and physiological functions of the heart
of Fstl1-eKO mice are currently under investigation.
In conclusion, our findings demonstrate that loss of endothelial Fstl1 in the
lung is associated with increased BMP/Smad phosphorylation and elevations in its
downstream targets Jagged1, Endoglin, Gata2 and Endothelin-1. These changes are
associated with impaired small pulmonary vascular remodeling and decreased right
ventricular output. Taken together, our findings suggest a key role of Fstl1 in titrating
the level of BMP signaling for proper postnatal lung development.

Acknowledgements
The authors would like to thank Jan M. Ruijter for expert advice on real time PCR
data analysis and Frank Ensink for technical assistance. This study was financially
supported by a grant from the Netherlands Lung Foundation (grant 3.2.12.083).

Conflict of interest Supplementary Figure 1. Endothelial Fstl1 knockout mice showed normal alveolarization
as shown by hematoxylin and eosin staining. Alveoli surrounding the airways (A) and alveoli
The authors have no conflict of interest. in lung parenchyma (B) of Fstl1­-eKO mice and of age-matched controls show no differences
at 1 week after birth. Scale bars represent 100 µm. Magnification 200x.

50 51
References 21. Hong K-H, Lee YJ, Lee E, et al. Genetic ablation of the BMPR2 gene in pulmonary endothelium is
sufficient to predispose to pulmonary arterial hypertension. Circulation. 2008;118(7):722-730.
1. Lowery JW, de Caestecker MP. BMP signaling in vascular development and disease. Cytokine
doi:10.1161/CIRCULATIONAHA.107.736801.
Growth Factor Rev. 2010;21(4):287-298. doi:10.1016/j.cytogfr.2010.06.001.
22. Thibault HB, Kurtz B, Raher MJ, et al. Noninvasive assessment of murine pulmonary arterial
2. Liu D, Wang J, Kinzel B, et al. Dosage-dependent requirement of BMP type II receptor
pressure: validation and application to models of pulmonary hypertension. Circ Cardiovasc
for maintenance of vascular integrity. Blood. 2007;110(5):1502-1510. doi:10.1182/
Imaging. 2010;3(2):157-163. doi:10.1161/CIRCIMAGING.109.887109.
blood-2006-11-058594.
23. Weyman AE, Dillon JC, Feigenbaum H, Chang S. Echocardiographic Patterns of Pulmonic Valve
3. Machado RD, Aldred MA, James V, et al. Mutations of the TGF-beta type II receptor BMPR2 in
Motion with Pulmonary Hypertension. Circulation. 1974;50(5):905-910. doi:10.1161/01.
pulmonary arterial hypertension. Hum Mutat. 2006;27(2):121-132. doi:10.1002/humu.20285.
4. Harrison RE, Flanagan JA, Sankelo M, et al. Molecular and functional analysis identifies ALK-
CIR.50.5.905. 2
24. Morikawa M, Koinuma D, Tsutsumi S, et al. ChIP-seq reveals cell type-specific binding patterns
1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic
of BMP-specific Smads and a novel binding motif. Nucleic Acids Res. 2011;39(20):8712-8727.
telangiectasia. J Med Genet. 2003;40(12):865-871.
doi:10.1093/nar/gkr572.
5. Fujiwara M, Yagi H, Matsuoka R, et al. Implications of mutations of activin receptor-like kinase
25. Kool H, Mous D, Tibboel D, de Klein A, Rottier RJ. Pulmonary vascular development goes awry in
1 gene (ALK1) in addition to bone morphogenetic protein receptor II gene (BMPR2) in children
congenital lung abnormalities. Birth Defects Res Part C Embryo Today Rev. 2014;102(4):343-358.
with pulmonary arterial hypertension. Circ J Off J Jpn Circ Soc. 2008;72(1):127-133.
doi:10.1002/bdrc.21085.
6. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in
26. Gangopahyay A, Oran M, Bauer EM, et al. Bone morphogenetic protein receptor II is a novel
hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(2):189-195.
mediator of endothelial nitric-oxide synthase activation. J Biol Chem. 2011;286(38):33134-
7. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-beta binding protein of endothelial
33140. doi:10.1074/jbc.M111.274100.
cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8(4):345-
27. Xu J, Zhu D, Sonoda S, et al. Over-expression of BMP4 inhibits experimental choroidal
351. doi:10.1038/ng1294-345.
neovascularization by modulating VEGF and MMP-9. Angiogenesis. 2012;15(2):213-227.
8. Pi X, Lockyer P, Dyer LA, et al. Bmper inhibits endothelial expression of inflammatory adhesion
doi:10.1007/s10456-012-9254-4.
molecules and protects against atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32(9):2214-
28. Moreno-Miralles I, Ren R, Moser M, Hartnett ME, Patterson C. Bone morphogenetic protein
2222. doi:10.1161/ATVBAHA.112.252015.
endothelial cell precursor-derived regulator regulates retinal angiogenesis in vivo in a mouse
9. Yao Y, Jumabay M, Wang A, Boström KI. Matrix Gla protein deficiency causes arteriovenous
model of oxygen-induced retinopathy. Arterioscler Thromb Vasc Biol. 2011;31(10):2216-2222.
malformations in mice. J Clin Invest. 2011;121(8):2993-3004. doi:10.1172/JCI57567.
doi:10.1161/ATVBAHA.111.230235.
10. Yao Y, Yao J, Radparvar M, et al. Reducing Jagged 1 and 2 levels prevents cerebral arteriovenous
29. Helbing T, Rothweiler R, Ketterer E, et al. BMP activity controlled by BMPER regulates the
malformations in matrix Gla protein deficiency. Proc Natl Acad Sci U S A. 2013;110(47):19071-
proinflammatory phenotype of endothelium. Blood. 2011;118(18):5040-5049. doi:10.1182/
19076. doi:10.1073/pnas.1310905110.
blood-2011-03-339762.
11. International PPH Consortium, Lane KB, Machado RD, et al. Heterozygous germline mutations
30. Jain RK. Molecular regulation of vessel maturation. Nat Med. 2003;9(6):685-693. doi:10.1038/
in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat
nm0603-685.
Genet. 2000;26(1):81-84. doi:10.1038/79226.
31. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary
12. Suzuki Y, Ohga N, Morishita Y, Hida K, Miyazono K, Watabe T. BMP-9 induces proliferation of
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878.
multiple types of endothelial cells in vitro and in vivo. J Cell Sci. 2010;123(Pt 10):1684-1692.
32. Hall SM, Gorenflo M, Reader J, Lawson D, Haworth SG. Neonatal pulmonary hypertension
13. Wang RN, Green J, Wang Z, et al. Bone Morphogenetic Protein (BMP) signaling in development
prevents reorganisation of the pulmonary arterial smooth muscle cytoskeleton after birth. J
and human diseases. Genes Dis. 2014;1(1):87-105. doi:10.1016/j.gendis.2014.07.005.
Anat. 2000;196(Pt 3):391-403. doi:10.1046/j.1469-7580.2000.19630391.x.
14. Adams D, Larman B, Oxburgh L. Developmental expression of mouse Follistatin-like 1 (Fstl1):
33. Xue Y, Gao X, Lindsell CE, et al. Embryonic Lethality and Vascular Defects in Mice Lacking the
Dynamic regulation during organogenesis of the kidney and lung. Gene Expr Patterns.
Notch Ligand Jagged1. Hum Mol Genet. 1999;8(5):723-730. doi:10.1093/hmg/8.5.723.
2007;7(4):491-500. doi:10.1016/j.modgep.2006.10.009.
34. Chen Y, Hao Q, Kim H, et al. Soluble endoglin modulates aberrant cerebral vascular remodeling.
15. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal
Ann Neurol. 2009;66(1):19-27. doi:10.1002/ana.21710.
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616.
35. Lebrin F, Goumans M-J, Jonker L, et al. Endoglin promotes endothelial cell proliferation and TGF-
16. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP)
beta/ALK1 signal transduction. EMBO J. 2004;23(20):4018-4028. doi:10.1038/sj.emboj.7600386.
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A.
36. Pece-Barbara N, Vera S, Kathirkamathamby K, et al. Endoglin null endothelial cells proliferate
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108.
faster and are more responsive to transforming growth factor beta1 with higher affinity
17. Kisanuki YY, Hammer RE, Miyazaki J, Williams SC, Richardson JA, Yanagisawa M. Tie2-Cre transgenic
receptors and an activated Alk1 pathway. J Biol Chem. 2005;280(30):27800-27808. doi:10.1074/
mice: a new model for endothelial cell-lineage analysis in vivo. Dev Biol. 2001;230(2):230-242.
jbc.M503471200.
doi:10.1006/dbio.2000.0106.
37. Mahmoud M, Allinson KR, Zhai Z, et al. Pathogenesis of arteriovenous malformations in the
18. Somi S, Klein ATJ, Houweling AC, et al. Atrial and ventricular myosin heavy-chain expression in
absence of endoglin. Circ Res. 2010;106(8):1425-1433. doi:10.1161/CIRCRESAHA.109.211037.
the developing chicken heart: strengths and limitations of non-radioactive in situ hybridization.
38. Arthur HM, Ure J, Smith AJ, et al. Endoglin, an ancillary TGFbeta receptor, is required for
J Histochem Cytochem Off J Histochem Soc. 2006;54(6):649-664. doi:10.1369/jhc.5A6846.2006.
extraembryonic angiogenesis and plays a key role in heart development. Dev Biol. 2000;217(1):42-
19. Cruz-Adalia A, Jiménez-Borreguero LJ, Ramírez-Huesca M, et al. CD69 limits the severity
53. doi:10.1006/dbio.1999.9534.
of cardiomyopathy after autoimmune myocarditis. Circulation. 2010;122(14):1396-1404.
39. Li DY, Sorensen LK, Brooke BS, et al. Defective angiogenesis in mice lacking endoglin. Science.
doi:10.1161/CIRCULATIONAHA.110.952820.
1999;284(5419):1534-1537.
20. Ruijter JM, Ramakers C, Hoogaars WMH, et al. Amplification efficiency: linking baseline and bias
40. Bourdeau A, Dumont DJ, Letarte M. A murine model of hereditary hemorrhagic telangiectasia. J
in the analysis of quantitative PCR data. Nucleic Acids Res. 2009;37(6):e45. doi:10.1093/nar/
Clin Invest. 1999;104(10):1343-1351. doi:10.1172/JCI8088.
gkp045.
41. Le TT, Conley KW, Brown NL. Jagged 1 is necessary for normal mouse lens formation. Dev Biol.

52 53
CHAPTER 3 1

The effect of cigarette smoking


on pulmonary Follistatin-like 1
expression: potential implication in
chronic obstructive pulmonary disease

Navessa P. Tania, Reinoud Gosens, Wim Timens, Maurice J.B. van den
Hoff, Martina Schmidt, Harm Maarsingh

55
The effect of cigarette smoking on pulmonary Follistatin-like 1 expression: Abstract
potential implication in chronic obstructive pulmonary disease
Bone morphogenetic protein (BMP) signaling is pivotal for embryonic lung
morphogenesis and epithelial repair in adult. The endogenous BMP antagonist
Follistatin-like 1 (Fstl1) is increased in several inflammatory diseases, but has not
been explored in chronic obstructive pulmonary disease (COPD) yet. Here, we
Navessa P. Tania1*, Reinoud Gosens1, Wim Timens2, Maurice J.B. van den Hoff3,
report that Fstl1 protein expression is increased in airway epithelium of COPD ex-
Martina Schmidt1, Harm Maarsingh4
smokers compared to non-COPD ex-smokers. Accordingly, phosphorylated Smad1/5,
indicative of active BMP signaling, was found reduced in total lung homogenates.
Taken together, our study provides a novel link between Fstl1 and COPD, provoking
further studies to unravel the Fstl1 functions in the lung and its implication in COPD. 3
1
University of Groningen, Department of Molecular Pharmacology, Groningen
Research Institute for Asthma and COPD, Groningen, The Netherlands.
2
University of Groningen, University Medical Center Groningen, Department of
Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD,
Groningen, The Netherlands.
3
Academic Medical Center, Department of Anatomy, Embryology and Physiology,
Amsterdam, The Netherlands.
4
Palm Beach Atlantic University, Lloyd L. Gregory School of Pharmacy, Department of
Pharmaceutical Sciences, West Palm Beach, Florida, USA.

56 57
Letter to the editor

Chronic obstructive pulmonary disease (COPD) is characterized by persistent


inflammation of the airways and lung parenchyma primarily triggered by tobacco
smoke exposure.1 Bone morphogenetic proteins (BMPs) belong to the TGF-β
superfamily that regulate embryonic lung morphogenesis and adult airway epithelial
homeostasis.2 The endogenous BMP antagonist Follistatin-like 1 (Fstl1) is an
extracellular glycoprotein shown to be crucial during embryonic lung development3,4
and contributes to inflammatory processes, such as in arthritis.5 Despite its’
importance in lung development, little is known about the role of Fstl1 in the adult
lung in health and disease. Here, we determined the expression of Fstl1 in the lung 3
of COPD patients compared to individuals with normal lung function.
Lung tissue and homogenates were obtained from twenty-two ex-smoker COPD
patients of GOLD stage II (n=11) and IV (n=11) and from twenty-nine non-COPD
control individuals without airway obstruction and with different smoking status
(8 never-smokers, 10 current smokers and 11 ex-smokers) for immunostaining and
western blot analysis. COPD stages classification referred to the Global Initiative
for Chronic Obstructive Lung Disease (GOLD) criteria.6 Patient characteristics are
Figure 1. The expression of Fstl1 is increased in COPD lung. (A) Representative immunoblot
of Fstl1 and quantification of Fstl1 protein expression in lung homogenates of COPD stage II
and IV patients compared to non-COPD controls differentiated by smoking status: NS, never-
smoker; CS, current-smoker; ES, ex-smoker. (B) Representative image of Fstl1 staining in lung
tissue. Red arrowhead points to airway epithelium, green arrowhead points to endothelium,
blue arrowhead points to vascular smooth muscle bundles, and purple arrowhead points to
inflammatory cells. AW, airway; V, blood vessel. Scale bar represents 200 µm. Magnification
of 100x. (C) Representative images of Fstl1 staining in the airway epithelium within the
different groups. Semi quantitative analysis of Fstl1 immunostaining in airway epithelium
(D), endothelium of large muscular arteries (E), vascular smooth muscle bundles (F), and
inflammatory cells (G). Each dot represents an individual patient. The horizontal line
represents median of each group (n=5-7 for NS; n=8-10 for CS; n=8-12 for ES; n=11 for ES
COPD II n=11; n=10-11 for ES COPD IV). The number of analyzed compartments in each group
varied due to some patients did not have the cell type of interest in their tissue section.
Scoring of the staining intensity (absent, low, medium, high) was performed by two persons
independently in a blinded manner. Statistical significance of differences was determined
with either a one-way ANOVA followed by a post hoc Tukey multiple comparisons test or a
non-parametric one-way ANOVA with a post hoc Kruskal-Wallis multiple comparisons test
for Gaussian and non-Gaussian distributed data, respectively. The Bonferroni correction was
used to correct for multiple testing. *p<0.05; **p<0.01; ***p<0.005 compared to indicated
groups.

provided in supplementary Table S1 and S2.


We observed that the expression of Fstl1 protein was significantly increased
in total lung homogenates of non-COPD ex-smokers alone (p=0.033) or combined
with non-COPD current smokers (p=0.016) compared to non-COPD never-smokers
(Figure 1A), suggesting that smoking induces a long-lasting increase of pulmonary
Fstl1 protein. In support, in comparison with the non-COPD never smoker group,
the total expression of Fstl1 protein in lung homogenates was significantly increased
in patients with COPD stage II alone (p=0.004) or in combination with COPD stage
IV (p=0.007; Figure 1A), all of which are ex-smokers. Notably, Fstl1 was originally

58 59
identified as a TGF-β target gene and exerts immunomodulatory functions.7,8 The
pulmonary expression of another BMP antagonist Noggin was also studied but
was found unaffected by cigarette smoking or disease state (non-COPD controls
vs COPD patients; supplementary figure 1). This indicates that the increase in BMP
antagonists was selective for Fstl1.
To dissect which cell types express Fstl1 in the lung, immunostaining for Fstl1
was performed. Fstl1 protein was expressed in different cell types in the lung,
including airway epithelium, endothelium, vascular smooth muscle cells, and
inflammatory cells (Figure 1B). The intensity of Fstl1 staining was markedly stronger
in airway epithelium of COPD patients compared to non-COPD controls (Figure 1C).
Semi-quantitative analysis of Fstl1 staining demonstrated that Fstl1 was significantly
increased in the airway epithelium (p=0.002; Figure 1D), endothelium (p=0.002;
3
Figure 1E), vascular smooth muscle (p=0.003; Figure 1F) and inflammatory cells
(p=0.034; Figure 1G) of the combined COPD stage II and IV patients compared to
the combined non-COPD controls. However, we did not observe an ever-smoking
effect for any of the studied compartments in the non-COPD groups. Interestingly,
the expression of Fstl1 protein was significantly increased in airway epithelium of
COPD stage II and IV patients (all ex-smokers) compared to non-COPD ex-smokers
(p=0.004; Figure 1D). This might indicate that increased Fstl1 in airway epithelium is
associated with COPD and is not only associated with smoking. Figure 2. BMP-specific Smad1/5 activation is reduced in the lung homogenates of COPD
patients. Representative immunoblots and quantitative protein expression analysis of (A)
Given that Fstl1 is a BMP antagonist, we examined whether increased Fstl1 phosphorylated Smad1/5 and (B) BMP4 in lung homogenates of COPD stage II and IV patients
was associated with reduced BMP/Smad signaling in the lung by investigating the compared to non-COPD controls differentiated by smoking status. Each dot represents an
expression of phosphorylated, BMP-specific, Smad1/5 in total lung homogenates. individual patient. Horizontal line represents median of each group (n=5 for NS; n=8 for CS;
n=8 for ES; n=11 for ES COPD II; n=10 for ES COPD IV). Data are expressed as ratios of proteins
Indeed, we observed that, when compared to non-COPD never smokers, of interest over the reference protein. Unphosphorylted Smad1 was used as a reference
phosphorylated Smad1/5 was significantly reduced in the lung homogenates of protein for pSmad1/5, and GAPDH for BMP4. *p<0.05; **p<0.01; ***p<0.005 compared to
indicated groups.
non-COPD of current smokers alone (p=0.01), ex-smokers alone (p=0.006) and the
combination of both (p=0.001; Figure 2A). Similarly, the expression of phosphorylated
Smad1/5 was also decreased in both COPD stage II and IV ex-smoker groups In conclusion, cigarette smoking increases the expression of Fstl1 protein and
individually (p=0.002; p=0.003) and combined (p=0.00018; Figure 2A) compared to downregulates phosphorylated Smad1/5 in the lung which persists even after
non-COPD never smokers. These findings show that there is a clear contribution of smoking cessation. Airway epithelium of both COPD stage II and IV patients express
ever smoking to reduced phosphorylated Smad1/5. To exclude that the reduction Fstl1 to a higher degree compared to non-COPD ex-smokers. Increased expression
in phosphorylated Smad1/5 is caused by a reduced pulmonary expression of BMP4 of the BMP-antagonist Fstl1 was accompanied with decreased phosphorylation of
rather than the increased expression of Fstl1, BMP4 protein levels were examined BMP-specific Smad1/5 in the lung, suggesting inhibition of the BMP/Smad signaling
in the total lung homogenates. We did not observe any significant changes in BMP4 pathway by Fstl1. These findings provoke future investigations addressing the
protein among the different groups (Figure 2B). Taken together, our data suggest precise roles of Fstl1 in airway epithelial driven inflammation, epithelial remodeling,
increase expression of Fstl1 and a subsequent decreased phosphorylated Smad1/5 and epithelial repair in non-COPD smokers and its potential contribution to the
levels in the lung are associated with ever smoking effect. development and pathophysiology of COPD.
Previous studies demonstrated that in vitro and in vivo overexpression of
Fstl1 triggers increased production of inflammatory cytokines and chemokines.7,8
Furthermore, a growing body of evidence demonstrates that Fstl1 contributes to
fibrotic processes in pulmonary fibrosis and in the airway wall of severe asthma.9,10
Whether increased expression of Fstl1 is coupled to such functional roles in COPD
requires further investigation.

60 61
Table S2. Characteristics of COPD patients and non-COPD controls used for
immunoblot.
Characteristics Non-COPD Non-COPD Non-COPD COPD II COPD IV*

Smoking status 5 8 9 11 11
53.3 56.25 59.7 70.9 59.4
Number of subjects (45.0-65.0) (45.0-68.0) (46.0-68.0) (58.0-80.0) (50.0-73.0)
Age (years) 4/1 2/6 3/6 8/3 5/6
43.29 22.3 26.3 28.1
Male/female 0 (14.0-75.0) (1.5-65.0) (0.0 -45.0) (0.0-48.8)
101.8 95.52 100.2 61.4 22.0
Pack years (78.5-130.0) (74.0-107.0) (63.7-121.0) (41.7 -76.0) (13.0-36.0) 3
77.52 78.14 76.3 61.6 37.7
FEV1% predicted (73.0-83.0) (62.4-92.0) (64.0-85.3) (45.7-80.0) (25.0-66.0)
FEV1/FVC% 76.2 79.35 76.3 61.6 37.7
(73.0-83.0) (62.39-92.0) (64.0-86.2) (45.7-80.0) (25.0-66.0)

* FEV1% predicted and FEV1/FVC% were measured post bronchodilators.

Supplementary figure 1. The expression of the other endogenous BMP antagonist Noggin
was unaltered in lung homogenates of patients with COPD compared to non-COPD. (A) A
representative immunoblot and (B) quantitative protein expression analysis of Noggin in the
lung homogenates of COPD compared to non-COPD controls with different smoking status.
Each dot represents an individual patient. Horizontal line represents median of each group
(NS n=5; CS n=8; ES n=8; ES COPD II n=11; ES COPD IV n=10). Data are expressed as ratios
of Noggin over the GAPDH reference protein. The statistical significance of differences was
determined either with a one-way ANOVA followed by a post hoc Tukey multiple comparisons
test or a non-parametric one-way ANOVA with a post hoc Kruskal-Wallis multiple comparisons
test for Gaussian and non-Gaussian distributed data, respectively. The Bonferroni correction
was used to correct for multiple testing.

Table S1. Characteristics of COPD patients and non-COPD controls used for
immunostaining.
Characteristics Non-COPD Non-COPD Non-COPD COPD II COPD IV*

Smoking status NS CS ES ES ES

Number of subjects 8 10 11 11 11

Age (years) 55.1 55.6 62.11 70.9 59.4


(42.0-70.0) (45.0-68.0) (46.0-74.0) (58.0-80.0) (50.0-73.0)
Male/female 5/3 2/8 5/6 8/3 5/6
42.33 25.75 26.3 28.1
Pack years 0 (14.0-75.0) (1.5-65.0) (0.0 -45.0) (0.0-48.8)
FEV1% predicted 102.03 97.07 96.9 61.4 22.0
(78.5-130.0) (74.0-107.0) (63.7 -121.0) (41.7 -76.0) (13.0-36.0)
FEV1/FVC% 76.2 79.35 76.3 61.6 37.7
(73.0-83.0) (62.39-92.0) (64.0-86.2) (45.7-80.0) (25.0-66.0)

* FEV1% predicted and FEV1/FVC% were measured post bronchodilators. NS, never-smoker;
CS, current smoker; ES, ex-smoker.

62 63
Materials and Methods Footnotes
Contributors: NPT designed and performed the experiments, interpreted the data
Fstl1 immunostaining and drafted the first manuscript version. RG, HM, MS, WT designed the experiments,
Human lung tissue sections were stained with primary rabbit anti-human Fstl1 interpreted the data and contributed to the writing of the manuscript. All authors
antibody (1:200; ABS616, Merck Milipore, Billerica, MA, USA) overnight at 4°C. The approved of the final version of the manuscript.
following day, tissue sections were incubated with HRP-conjugated goat anti-rabbit
antibody for 2 h (1:100, 711-035-152, Jackson Immunoresearch, West Grove, PA, Funding: This study was financially supported by a grant from the Netherlands
USA). Lung Foundation (grant 3.2.12.083).

Immunoblot Competing interest: none declared.


Equal amounts of lung homogenates were loaded to SDS PAGE electrophoresis and 3
transferred to PVDF membranes. After incubation with 1x ROTI block (Carl Roth; Patient consent: not applicable, see below
Karlsruhe, Germany), membranes were incubated with antibody of interest: rabbit
anti-Fstl1 (1:2000; ABS616, Merck Millipore; Billerica, MA, USA), mouse anti-BMP4 Ethics approval: not applicable; Research was conducted according to the Research
(1:500; MAB757-100; R&D system, Oxford, UK), rabbit anti-pSmad1/5 (1:750; 9516; Code of the University Medical Center Groningen (http://www.umcg.nl/EN/
Cell Signaling; Danvers, MA, USA), rabbit anti-Smad1 (1:1000; 9743; Cell Signaling, Research/Researchers/General/ResearchCode/Paginas/default.aspx) and national
Danvers, MA, USA), mouse anti-GAPDH (1:2000; sc-47725; Santa Cruz Biotechnology, ethical, legal and professional guidelines (“Code of conduct; Dutch federation of
Heidelberg, Germany), goat anti-Noggin (1:1000; AF719; R&D system, Oxford, UK). biomedical scientific societies”; https://www.federa.org/).
The following day, horseradish peroxidase (HRP) conjugated antibodies were added,
either donkey anti-rabbit IgG (1:4,000; 711-035-152), anti-goat IgG (1:4,000; 705- Acknowledgement
035-003), or anti-mouse IgG (1:4000; 715-035-150 Jackson Immunoresearch, West The authors would like to thank Sophie Bos and Marjan Luinge for technical
Grove, PA, USA). Protein band intensities were quantified using Image Studio Lite assistance.
version 5.0.

Statistical analysis
Data are presented as individual data points with medians per patient-group unless
stated otherwise. To determine the normality of data distribution, a Shapiro-
Wilk normality test was done prior to further statistical analysis. The statistical
significance of differences of normally distributed data was performed using a
one-way ANOVA followed by a post hoc Tukey multiple comparisons test. For non-
Gaussian distributed data, the statistical significance of differences was determined
using a non-parametric one-way ANOVA with a post hoc Kruskal-Wallis multiple
comparisons test. The Bonferroni correction was used to correct for multiple testing.
Differences were considered to be statistically significant at p<0.05.

64 65
References
1. Garvey C. Recent updates in chronic obstructive pulmonary disease. Postgrad Med.
2016;128(2):231-238. doi:10.1080/00325481.2016.1118352.
2. Sountoulidis A, Stavropoulos A, Giaglis S, et al. Activation of the canonical bone morphogenetic
protein (BMP) pathway during lung morphogenesis and adult lung tissue repair. PloS One.
2012;7(8):e41460. doi:10.1371/journal.pone.0041460.
3. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP)
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A.
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108.
4. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616.
5. Chaly Y, Hostager B, Smith S, Hirsch R. Follistatin-like protein 1 and its role in inflammation and
inflammatory diseases. Immunol Res. 2014;59(1-3):266-272. doi:10.1007/s12026-014-8526-z.
6. Vestbo J, Hurd SS, Agustí AG, et al. Global Strategy for the Diagnosis, Management, and Prevention 3
of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2013;187(4):347-365.
doi:10.1164/rccm.201204-0596PP.
7. Miyamae T, Marinov AD, Sowders D, et al. Follistatin-Like Protein-1 Is a Novel Proinflammatory
Molecule. J Immunol. 2006;177(7):4758-4762. doi:10.4049/jimmunol.177.7.4758.
8. Chaly Y, Marinov AD, Oxburgh L, Bushnell DS, Hirsch R. FSTL1 promotes arthritis in mice by
enhancing inflammatory cytokine/chemokine expression. Arthritis Rheum. 2012;64(4):1082-
1088. doi:10.1002/art.33422.
9. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878.
10. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by Inducing
Oncostatin M. J Immunol Baltim Md 1950. September 2015. doi:10.4049/jimmunol.1501105.

66 67
CHAPTER 4

Activin-A: active in inflammation in COPD

Navessa P. Tania, Martina Schmidt, Reinoud Gosens

68
Activin-A: active in inflammation in COPD Chronic obstructive pulmonary disease (COPD) is characterised by irreversible
airflow limitation in which chronic inflammation of the airways plays a major
role. The persistent inflammation is triggered by inhaled toxic substances, such as
cigarette smoke. Accumulating evidence indicates the involvement of developmental
pathways in chronic lung diseases, including COPD.1 Signalling pathways such as
Navessa P. Tania, Martina Schmidt, Reinoud Gosens transforming growth factor (TGF)-β, WNT and Sonic hedgehog have all been linked
to COPD, either because of genetic associations or because of differential gene
and protein expression in lung tissue.2–4 Although these pathways were originally
primarily known for their role in development, it is now increasingly clear that
these pathways also play crucial regulatory roles in tissue inflammation and repair,
Department of Molecular Pharmacology and GRIAC Research Institute, University of providing a plausible explanation for the involvement of these pathways in COPD.3
Groningen, Groningen, The Netherlands. Activin-A, a member of the TGF-β superfamily, is an important regulator of
embryonic development, haematopoiesis and a broad range of tightly regulated
biological processes, including immunity and tissue repair.5 Dysregulation of activin-A
may contribute to the development of disease. Recently, increased expression of
activin-A has been demonstrated in pulmonary hypertension,6 acute lung injury7 4
and asthma.8 Oxidative stress, Toll-like receptor ligands and inflammatory cytokines
stimulate the expression and production of activin-A, which in turn regulates
inflammatory cytokine release, explaining its role in inflammatory responses.7
Accordingly, the endogenous activin-A inhibitor follistatin has attracted great
interest in view of its ability to counteract activin-A activity. Due to its involvement
in inflammatory responses, it is rational to connect activin-A to the pathogenesis of
inflammatory diseases. An imbalance between activin-A and follistatin potentially
leads to excess activin-A-mediated inflammatory responses. However, the roles of
activin-A and follistatin have not yet been established in COPD.
In this issue, Verhamme et al.9 demonstrate for the first time that activin-A is an
important regulator of cigarette smoke-induced inflammation in COPD. They found
that activin-A is elevated and activated in the airway epithelium, airway smooth
muscle and alveolar macrophages of COPD patients. These findings were further
validated in vivo, demonstrating that cigarette smoke induces marked expression
of activin-A in the lungs and bronchoalveolar lavage (BAL) fluid of mice. In vitro,
activin-A expression increased after cigarette smoke exposure of human bronchial
epithelial cells, whereas expression of follistatin was reduced. These data suggest
that cigarette smoke is a major factor involved in activin-A upregulation. Notably,
regardless of airflow limitation, lung tissues of current smokers had a significantly
higher level of activin-A mRNA expression compared with nonsmoker subjects,
whereas the increased activin-A protein expression in airway epithelium was specific
for COPD. Most intriguingly, this study revealed that inhibition of activin-A signalling
by administration of its endogenous inhibitor follistatin, attenuates cigarette smoke-
induced production of interleukin (IL)-6, monocyte chemoattractant protein-1,
tumour necrosis factor (TNF)-α, keratinocyte-derived chemokine and TGF-β1,
and induces the release of the anti-inflammatory cytokine IL-10. Furthermore,
administration of follistatin reduced the number of inflammatory cells, such as
monocytes, macrophages, neutrophils, and CD4+ and CD8+ T-cells, in the BAL fluid

70 71
of cigarette smoke-exposed mice. The authors conclude that activin-A is not an References
innocent bystander but plays an active role in cigarette smoke induced pulmonary 1. Shi W, Chen F, Cardoso WV. Mechanisms of lung development: contribution to adult lung disease
inflammation.9 and relevance to chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2009;6(7):558-
563. doi:10.1513/pats.200905-031RM.
The role of activin-A has been widely studied in inflammatory cells and a dual 2. Baarsma HA, Spanjer AIR, Haitsma G, et al. Activation of WNT / β-Catenin Signaling in Pulmonary
role of activin-A in inflammatory responses has been proposed. On one hand, Fibroblasts by TGF-β 1 Is Increased in Chronic Obstructive Pulmonary Disease. PLOS ONE.
activin-A promotes alveolar cell death and stimulates the production of pro- 2011;6(9):e25450. doi:10.1371/journal.pone.0025450.
inflammatory cytokines, including IL-6. Furthermore, activin-A is increased following 3. Rock J, Königshoff M. Endogenous lung regeneration: potential and limitations. Am J Respir Crit
Care Med. 2012;186(12):1213-1219. doi:10.1164/rccm.201207-1151PP.
pulmonary lipopolysaccharide (LPS) exposure and follistatin reduces LPS-induced 4. Van Durme YMTA, Eijgelsheim M, Joos GF, et al. Hedgehog-interacting protein is a COPD susceptibility
pulmonary inflammation in mice.7 On the other hand, follistatin was shown to gene: the Rotterdam Study. Eur Respir J. 2010;36(1):89-95. doi:10.1183/09031936.00129509.
augment the production of pro-inflammatory cytokines in concerted action with 5. Hedger MP, de Kretser DM. The activins and their binding protein, follistatin-Diagnostic
TNF-α and IL-13.8 This dual role was also noted in an LPS model of endotoxaemia, and therapeutic targets in inflammatory disease and fibrosis. Cytokine Growth Factor Rev.
2013;24(3):285-295. doi:10.1016/j.cytogfr.2013.03.003.
showing that low doses of follistatin augmented IL-6 expression, but reduced 6. Yndestad A, Larsen K-O, Oie E, et al. Elevated levels of activin A in clinical and experimental
TNF-α and IL-1β concentrations, whereas at higher follistatin concentrations IL-6 pulmonary hypertension. J Appl Physiol Bethesda Md 1985. 2009;106(4):1356-1364.
expression was normalised.10 Therefore, the role of activin-A in cigarette smoke- doi:10.1152/japplphysiol.90719.2008.
induced inflammation and COPD may be more complex, and studies are warranted 7. Apostolou E, Stavropoulos A, Sountoulidis A, et al. Activin-A overexpression in the murine lung
causes pathology that simulates acute respiratory distress syndrome. Am J Respir Crit Care Med. 4
to identify its role in more detail. In particular, its potential anti-inflammatory role 2012;185(4):382-391. doi:10.1164/rccm.201105-0784OC.
in the regulation of inflammation in COPD needs to be clarified and the therapeutic 8. Kariyawasam HH, Pegorier S, Barkans J, et al. Activin and transforming growth factor-beta
potential of activin-A inhibition needs to be explored in more detail. signaling pathways are activated after allergen challenge in mild asthma. J Allergy Clin Immunol.
Several intriguing questions arise from this study require further investigation. 2009;124(3):454-462. doi:10.1016/j.jaci.2009.06.022.
9. Verhamme FM, Bracke KR, Amatngalim GD, et al. Role of activin-A in cigarette smoke-induced
Activin-A regulates expression of its own inhibitor follistatin as a negative feedback inflammation and COPD. Eur Respir J. 2014;43(4):1028-1041. doi:10.1183/09031936.00082413.
mechanism to control the degree of inflammation.11 Increased expression of 10. Jones KL, Mansell A, Patella S, et al. Activin A is a critical component of the inflammatory
follistatin was not observed in COPD, suggesting that this negative feedback response, and its binding protein, follistatin, reduces mortality in endotoxemia. Proc Natl Acad
Sci U S A. 2007;104(41):16239-16244. doi:10.1073/pnas.0705971104.
mechanism might be altered, leading to uncontrolled inflammatory responses.
11. Bartholin L, Maguer-Satta V, Hayette S, et al. Transcription activation of FLRG and follistatin by
Therefore, it is important to examine the effect of cigarette smoke on activin-A activin A, through Smad proteins, participates in a negative feedback loop to modulate activin A
and follistatin expression in epithelial and inflammatory cells derived from COPD function. Oncogene. 2002;21(14):2227-2235. doi:10.1038/sj.onc.1205294.
patients. Moreover, it is rational to explore the role of activin-A in remodelling as 12. Aoki F, Kurabayashi M, Hasegawa Y, Kojima I. Attenuation of bleomycin-induced pulmonary
fibrosis by follistatin. Am J Respir Crit Care Med. 2005;172(6):713-720. doi:10.1164/rccm.200412-
activin-A shares homology and signalling characteristics with TGF-β1. In this respect,
1620OC.
follistatin inhibits bleomycin-induced pulmonary fibrosis12 and ovalbumin induced 13. Hardy CL, Nguyen H-A, Mohamud R, et al. The activin A antagonist follistatin inhibits asthmatic
airway remodelling.13 Interestingly, activin-A also promotes alveolar epithelial cell airway remodelling. Thorax. 2013;68(1):9-18. doi:10.1136/thoraxjnl-2011-201128.
death.7 Hence, it would be worthwhile to explore the role of activin-A in emphysema.
Furthermore, although administration of exogenous follistatin was able to inhibit
cigarette smoke-induced inflammation, additional study is needed to develop
strategies to restore the endogenous activin-A/follistatin imbalance.
In conclusion, the study of Verhamme et al.9 sheds light on the role of
activin-A and follistatin in COPD, and motivates further investigation. Persistent
and exaggerated production of activin-A might be a key factor provoking persistent
inflammation in COPD. Restoring the activin-A/follistatin imbalance is a therapeutic
strategy worth pursuing in COPD.

72 73
CHAPTER 5 1

Pro-inflammatory roles of Follistatin-like 1


in coordinating epithelial-mesenchymal
communication in inflammatory cytokine
release in the lung.

Navessa P. Tania, Harm Maarsingh, Maurice J.B. van den Hoff, Vanessa
Niburg, Andrew J. Halayko, Martina Schmidt, Reinoud Gosens

75
Pro-inflammatory roles of Follistatin-like 1 in coordinating epithelial-mesenchymal Abstract
communication in inflammatory cytokine release in the lung.
The pathogenesis of respiratory diseases such as asthma and chronic obstructive
pulmonary disease (COPD) has been linked to chronic inflammation of the lungs,
potentiated by inhaled toxic particles and infectious agents. The airway epithelium
releases inflammatory mediators with paracrine effects on neighboring cells,
Navessa P. Tania1,2, Harm Maarsingh3, Maurice J.B. van den Hoff4, Vanessa Niburg1, including lung fibroblasts and airway smooth muscle cells. Follistatin-like 1 (Fstl1) is a
secreted glycoprotein expressed by developing lung epithelium, and can contribute
Andrew J. Halayko5, Martina Schmidt1,2, Reinoud Gosens1,2
to inflammatory lung pathogenesis if it becomes dysregulated. Here, we aimed to
investigate the functional role of Fstl1 as a determinant of airway epithelial-driven local
inflammation. The human bronchial epithelial cell line (16HBE14o-) was transiently
University of Groningen, Department of Molecular Pharmacology, Groningen, The
1 transfected with FLAG-tagged Fstl1 or cyan fluorescent protein (control) constructs
to generate Fstl1 and CFP conditioned medium (Fstl1 CM and CFP CM, respectively).
Netherlands. The mRNA expression and protein release of IL-6 and IL-8 were determined in cell
lysates and culture supernatants from lung mesenchymal cells. An ARY005 human
2
University of Groningen, University Medical Center Groningen, Groningen Research
cytokine array kit was used to identify epithelial-derived secreted factors present
Institute for Asthma and COPD, Groningen, The Netherlands. in CM.Fstl1 CM increased IL-6 and IL-8 mRNA expression and protein release from
human lung mesenchymal cells. However, neither recombinant Fstl1 alone nor
3
Palm Beach Atlantic University, Lloyd L. Gregory School of Pharmacy, Department of direct overexpression or knockdown of Fstl1 in lung fibroblasts was sufficient to
Pharmaceutical Sciences, West Palm Beach, FL, USA. alter baseline or IL-1β-induced IL-6 and IL-8 protein release. Fstl1 overexpression 5
did induce inflammatory mediator release (GM-CSF, RANTES, IL-6, IL-8) from human
4
Academic Medical Center, Department of Anatomy, Embryology and Physiology, bronchial epithelial cells. In turn, we found that neutralizing antibodies for GM-
Amsterdam, The Netherlands. CSF, RANTES, IL-6 and IL-8 completely diminished Fstl1 CM-induced release of IL-6
and IL-8 from human lung fibroblasts. Our findings demonstrate that Fstl1 is a pro-
5
University of Manitoba, Department of Physiology and Pathophysiology, Winnipeg, inflammatory factor that can orchestrate the local microenvironment by promoting
Canada. airway epithelial cells to release a repertoire of mediators that induce a further
subset of inflammatory mediators from neighboring lung mesenchymal cells. These
data implicate a potential new role for Fstl1 in coordinating and potentiating local
inflammation in chronic lung disorders like asthma and COPD.

76 77
Introduction Material and Methods

The pulmonary epithelial lining serves as a boundary between the lung and the external Cell culture
environment. Hence, it acts as the first line of defense against inhaled toxic gases, The human bronchial epithelial cell line (16HBE14o-) was cultured on collagen-coated
particles and infectious agents. Epithelial cells signal changes in the environment to flasks in minimum essential medium (Lonza; Basel, Switzerland) supplemented
their neighboring cells, including inflammatory cells and mesenchymal cells such as with 10% (v/v) heat inactivated fetal bovine serum (FBS), 2 mM L-glutamine, 100
sub-epithelial fibroblasts and airway smooth muscle cells.1–3 In turn, a number of U/ml penicillin, 100 ug/ml streptomycin, 56 ug/ml gentamycin and 1.5 ug/ml
trophic and secretory responses are induced in mesenchymal cells, including the amphotericin B (Thermo Fisher Scientific; Waltham, MA, USA). Human fetal lung
biosynthesis of a myriad of cytokines and chemokines which eventually result in fibroblasts MRC-5 were maintained in Ham’s F12 medium (Lonza; Basel, Switzerland)
leukocyte influx to the lung.2,3 supplemented with 10% (v/v) FBS, 2 mM L-glutamine, 100 U/ml penicillin, 100 ug/
Follistatin-like 1 (Fstl1) is an extracellular glycoprotein belonging to a secreted ml streptomycin and 1.5 ug/ml amphotericin B (Thermo Fisher Scientific; Waltham,
protein acidic and rich in cysteine (SPARC) family, which is crucial for normal MA, USA). Human bronchial airway smooth muscle cells, immortalized by stable
lung development.4,5 High levels of Fstl1 have been reported in a number of ectopic expression of human telomerase reverse transcriptase (hTERT) as described
inflammatory diseases, such as rheumatoid arthritis, ulcerative colitis, systemic previously13 were maintained in Dulbecco’s modified Eagle’s medium (DMEM;
lupus erythematosus, systemic autoimmune disease, and obesity.6–8 Administration Thermo Fisher Scientific; Waltham, MA, USA) supplemented with 10% (v/v) FBS, 100
of Fstl1 aggravates arthritis in mice,9 whereas blocking Fstl1 using a neutralizing U/ml penicillin and 100 ug/ml streptomycin and 1.5 amphotericin B (Thermo Fisher
antibody ameliorated collagen-induced arthritis.10 Fstl1 regulates inflammatory Scientific; Waltham, MA, USA). Cells were maintained in a humidified incubator at
cytokine release from mesenchymal and inflammatory cells, for example the 37 °C in air/CO2 (95/5% v/v).
upregulation of IL-6 and IL-8 from stromal cells, synovial fibroblasts, adipocytes, and
macrophages in vitro7,9,11,12 and IFN-γ from T cells.10 These findings suggest that Fstl1 Generation of Fstl1 conditioned medium and stimulation of mesenchymal cells­ 5
may sub-serve a previously unrecognized role as a pro-inflammatory cytokine.9,11 16HBE14o- cells were grown to 80% confluence and transfected with either 1 µg/
Very little is known about the immunomodulatory function of Fstl1 in the lung. ml pcDNA3.1 containing FLAG-tagged mouse Fstl1 (Fstl1-FLAG) or cyan fluorescent
Thus, in the present study we tested the hypothesis that Fstl1 can mediate cytokine protein (CFP) as control for 24 h using transfection reagent SAINT-MIX (SM-1001-01,
release from pulmonary mesenchymal cells. Synvolux Therapeutics; Groningen, the Netherlands). The following day, cells were
washed and serum-free media was added. Cell culture supernatants were collected
48 h later. Culture supernatants obtained from 16HBE14o- cells overexpressing
Fstl1-FLAG or CFP were defined as Fstl1 conditioned medium (Fstl1 CM) or CFP CM,
respectively. Fstl1 CM and CFP CM were centrifuged at 4500g for 10 min and filtered
through a membrane of 0.2 µm pore size to remove cells and large cell debris,
respectively. Expression levels of Fstl1-FLAG were verified using Western blot. Fstl1
was detected using a rabbit anti-FLAG antibody (2368S, Cell Signaling; Danvers,
MA, USA) and horseradish peroxidase (HRP)-conjugated donkey anti-rabbit IgG as
secondary antibody (711-035-152, Jackson Immunoresearch; West Grove, PA, USA).
Protein bands were visualized using enhanced chemiluminescence substrate(ECL;
Perkin Elmer; Groningen, the Netherlands) in the G-box gel documentation system
(Syngene; Cambridge, UK).
To test the effects of Fstl1 CM and CFP CM on mesenchymal cells, lung fibroblasts
and airway smooth muscle cells were grown until 90% confluence. After serum
deprivation for 24 h, cultures were treated with different concentrations of Fstl1 CM
(20%, 40% and 50% v/v with 0.5% FBS in Ham’s F12 or in serum-free DMEM or CFP
CM (50% v/v) as controls for 24 h. Cells were lysed and culture supernatants were
collected.

78 79
Recombinant Fstl1 stimulation, gain- and loss-of function studies Table 1: Primers used for qRT-PCR analysis.
MRC-5 human lung fibroblasts were grown to 90% confluence in 24-wells cluster Gene Forward primer (5’ 3’) Reverse primers (5’ 3’’)
plates and serum-deprived for 24 h prior stimulation with or without 15, 50, 100
FSTL1 ACCACGATGTGGAAACGCTGGC TGCCATTACTGCCACACACAGGC
and 200 ng/ml recombinant human Fstl1 (1694-FN, R&D system; Oxford, UK)
for 24 h. To overexpress Fstl1, MRC-5 cells were transfected with either 1 µg/ml IL6 AGGAGACTTGCCTGGTGAAA TAAAGCTGCGCAGAATGAGA
mouse Fstl1-FLAG or CFP as controls using transfection reagent SAINT-MIX (SM- IL8 TAGCAAAATTGAGGCCAAGGC AAACCAAGGCACAGTGGAAC
1001-01, Synvolux Therapeutics; Groningen, the Netherlands). To silence Fstl1 gene
expression, MRC-5 cells were treated with either 50 pmol FSTL1 siRNA (sc-39815, RANTES TGCTGCTTTGCCTACATTGC TCGGGTGACAAAGACGACTG
Santa Cruz Biotechnology; Heidelberg, Germany) or scrambled siRNA as controls GM-CSF AGTTCTCTGGAGGATGTG GCT CTACTGTTTCATTCATCTCAGCAGC
(SI03650318, Qiagen; Venlo, the Netherlands) using transfection reagent SAINT-RED 18S CGCCGCTAGAGGTGAAATTC TTGGCAAATGCTTTCGCTC
siRNA/RNA (SR-1003-02, Synvolux Therapeutics; Groningen, the Netherlands). Cells
were serum deprived for 24 h prior to stimulation with or without 0.01, 0.1 and 1.0 SDHA GGGAAGACTACAAGGTGCGG CTCCAGTGCTCCTCAAAGGG
ng/ml IL-1β (11457756001, Sigma Aldrich; St. Louis, MO, USA). Culture supernatants HPRT1 AAGCCAGACTTTGTTGGATTTGA ACTGGCGATGTCAATAGGACTC
were collected and cells were lysed 24 h after stimulation.
Abbreviations used: FSTL1, Follistatin-like 1; IL6, interleukin 6; IL8, interleukin 8; RANTES,
regulated on activation, normal T cell expressed and secreted; GM-CSF, granulocyte
RNA isolation and Real time quantitative PCR macrophage-colony stimulating factor; 18S, 18S ribosomal RNA; SDHA, succinate
Total RNA was isolated using TRI Reagent® Solution (AM9738, Thermo Fisher dehydrogenase complex flavoprotein subunit A; and HPRT1, hypoxanthine guanine
Scientific; Waltham, MA, USA) according to the manufacturer’s instruction. The phosphoribosyl transferase 1.
total RNA concentration was determined using the Spectrophotometer NanoDrop®
ND1000 (Thermo Fisher Scientific; Waltham, MA, USA). Equal amounts of total RNA
5
Enzyme-linked immunosorbent assays (ELISA)
were reverse transcribed using Reverse Transcription System (Promega; Madison,
The concentration of IL-6 (M1916) and IL-8 (M1918) in culture supernatants
WI, USA). Diluted cDNA was mixed with FastStart Universal SYBR Green Master
was measured in triplicates using PeliKine compactTM ELISA kit according to the
Mix (Roche Applied Science; Penzberg, Germany) and gene of interest primers
manufacturer’s protocol (Sanquin; Amsterdam, the Netherlands). The concentration
sets (Biolegio; Nijmegen, the Netherlands). Primer sequences are listed in Table 1.
of IL-6 and IL-8 in the culture supernatants of lung fibroblasts and airway smooth
Real-time quantitative PCR was performed using the Illumina Eco Personal qPCR
muscle cells treated with conditioned medium obtained from 16HBE14o- cells was
System (Westburg; Leusden, The Netherlands). The qPCR reaction was started by
measured and corrected by subtracting the concentration of IL-6 and IL-8 already
denaturation at 95°C for 15 minutes followed by 45 cycles of denaturation at 94°C
present in Fstl1 CM and CFP CM controls. The sensitivities for IL-6 and IL-8 were
for 30 s, annealing at 59°C for 30 s and elongation at 72°C for 30 s. Final elongation
0.2 pg/ml and 1.0 pg/ml, respectively. The concentration of human GM-CSF and
was for 5 minutes at 72 °C. Real time PCR data were analyzed using LinRegPCR
RANTES was measured in duplicate in culture medium supernatants using human
software version 2013.1.14 Data were expressed in arbitrary units as ratio of the
GM-CSF (DGM00) and RANTES (DRN00B) quantikine ELISA kits according to the
starting concentration (N0) of each gene of interest corrected to the geometric mean
manufacturer’s protocol (R&D system; Oxford, UK). The optical density was measured
of the N0 value of 3 reference genes (18S, SDHA and HPRT1).
at 450 nm with 570 nm as a wavelength correction using Gen5an software using a
plate reader (BioTek; Winooski, VT, USA). The sensitivities for GM-CSF and RANTES
ELISA were 3 pg/ml and 2 pg/ml, respectively.

80 81
Human cytokine arrays Results
To identify potential inflammatory mediators in Fstl1 CM and CFP CM, a cytokine
array was performed using the Human Cytokine Array Kit (ARY005, R&D system; Fstl1 induces inflammatory cytokine release from mesenchymal cells
Oxford, UK) according to the manufacturer’s instruction. An antibody array that Bronchial epithelial cells express and release endogenous Fstl1 protein in vitro to
captures 29 cytokines was spotted in duplicate on nitrocellulose membranes. A a higher degree than lung fibroblasts and airway smooth muscle cells, which show
complete list of cytokines is listed in Table 2. Detection antibody/cytokine complexes only limited intracellular protein expression (Figure 1A and B) and release (Figure
bound to immobilized capture antibodies on the membrane were visualized using 1C and D). A previous study demonstrated crosstalk between lung epithelial and
ECL (Perkin Elmer, Groningen, the Netherlands) in the G-box gel documentation mesenchymal cells in driving inflammatory response in the lung.3 To investigate the
system (Syngene; Cambridge, UK).

Table 2. Human cytokines and chemokines studied in the antibody array.


C5a GM-CSF IL-13 Serpin E1/PAI-1
IL-4 IL-8 CCL4/MIP-1β IL-1ra/IL-1F3
IL-32α CCL2/MCP-1 IFN-γ TNF-α
CD40 ligand/TNFSF5 CXCL1/GRO α IL-16 IL-2
IL-5 IL-10 CCL5/RANTES IL-27
CXCL10/IP-10 MIF IL-1α/IL-1F1 TREM-1
G-CSF CCL1/I-309 IL-17 IL-23
IL-6 IL-12 p70 CXCL12/SDF-1 ICAM-1
CXCL11/I-TAC CCL3/MIP-1α IL-1β/IL-1F2 IL-17E
5
Neutralizing antibody treatment
MRC-5 cells were grown to 90% confluence and serum deprived for 24 h before
experiments. Fstl1 and CFP CM (50% v/v with 0.5% serum Ham’s F12) were
incubated with either neutralizing antibodies against IL-6 (MAB206), IL-8 (MAB208),
GM-CSF (MAB215-100), RANTES (AF-278-NA) or a combination of these neutralizing
antibodies (R&D system, Oxford, UK) 1 h prior to the addition of CM to the cells. IgG
isotype control was used as a control (MAB002, R&D system; Oxford, UK). Cells were
washed and maintained in Ham’s F12 medium containing 0.5% FBS before adding
the neutralizing antibody-treated CM. Culture supernatants were collected and cells
were lysed 24 h after treatment.

Statistical analysis
Data are presented as mean values ± standard error of the mean (SEM). Prior to
further statistical analysis, the normality of data distribution was examined using
a Shapiro-Wilk normality test. The statistical significance of differences of normal
Gaussian distributed data was determined using an independent samples 2-tailed
t-test for comparing 2 groups or two-way ANOVA followed by a post hoc Tukey Figure 1. Pulmonary bronchial epithelial cells express and release Fstl1. Representative
multiple comparisons test for comparing more than 2 groups. For non-Gaussian immunoblots and quantification of intracellular (A, B) and secreted (C, D) Fstl1 in bronchial
distributed data, the statistical significance of differences was done using a non- epithelial cells, MRC-5 lung fibroblasts and airway smooth muscle cells. Data represent
mean ± SEM of 7-9 independent cell lysates and culture supernatants. (E) The expression
parametric Mann-Whitney U test for comparing 2 groups or non-parametric one- of FSTL1 mRNA in Fstl1-FLAG overexpressing bronchial epithelial cells compared to CFP
way ANOVA with a post hoc Kruskal-Wallis multiple comparisons test for comparing controls. (F) A representative immunoblot and quantification of Fstl1 in supernatants of
more than 2 groups. All statistical analysis was performed using IBM SPSS Statistics Fstl1-FLAG overexpressing bronchial epithelial cells compared to CFP controls. Protein bands
were detected using anti-FLAG antibody. Data represent mean ± SEM of 6 independent
22 (Armonk, NY, USA). Differences were considered to be statistically significant at experiments. *p<0.05, **p<0.01, ***p<0.001 compared to bronchial epithelial cells or
p<0.05. controls.

82 83
effect of Fstl1 expressed by bronchial epithelial cells and its potential contribution by 2.0-fold (p<0.001) and 1.7-fold (p<0.001), respectively, in airway smooth muscle
to inflammatory cytokine release from mesenchymal cells, we overexpressed Fstl1 cells (Figure 2C and 2D)­.
in bronchial epithelial cells and treated lung fibroblasts and airway smooth muscle We further attempted to decipher whether Fstl1 CM also affected the release
cells with Fstl1 CM derived from bronchial epithelial cells. Overexpression of Fstl1- of IL-6 and IL-8 protein from mesenchymal cells, thus we determined IL-6 and IL-8
FLAG was verified in cell lysates and culture supernatants of bronchial epithelial cells. protein in culture supernatants from lung fibroblasts and airway smooth muscle cells,
Fstl1 mRNA was 4.3-fold increased (p<0.001; Figure 1E) in the Fstl1-overexpressing but corrected for the levels that were already present in the CM. In this manner we
bronchial epithelial cells, whereas the amount of secreted Fstl1 protein was 12.1- were able to demonstrate, similar to our measurement of mRNA abundance. Fstl1
fold increased (p<0.001; (Figure 1F). CM dose-dependently induced IL-6 and IL-8 protein release by 1.5-fold (57.6 ng/
Interestingly, treatment of lung fibroblasts and airway smooth muscle cells with ml; p<0.05) and 1.7-fold (106.0 ng/ml; p<0.001), respectively, from lung fibroblasts
Fstl1 CM dose-dependently increased mRNA expression and protein release of IL-6 (Figure 3A and 3B). Release of IL-6 and IL-8, respectively, from airway smooth muscle
and IL-8. At 50% concentration, in comparison to control CFP CM, Fstl1 CM induced cells was also increased by 3.1-fold (39.1 ng/ml; p<0.001) and 1.8-fold (104.1 ng/
IL-6 and IL-8 mRNA by 1.9-fold (p<0.05) and 1.6-fold (p<0.05), respectively, in lung ml; p<0.05) after Fstl1 CM exposure (Figure 3C and 3D). Collectively, these data
fibroblasts (Figure 2A and 2B). Similarly 50% Fstl1 CM increased IL-6 and IL-8 mRNA suggest that Fstl1 is involved in the inflammatory cytokine release from pulmonary
mesenchymal cell.

Figure 3. Fstl1 CM derived from bronchial epithelial cells induces IL-6 and IL-8 release from
mesenchymal cells. 16HBE14o- cells were grown to confluence and transfected with Fstl1-
Figure 2. Fstl1 CM derived from bronchial epithelial cells induces IL-6 and IL-8 mRNA FLAG or CFP (control) to generate Fstl1 CM and CFP CM, respectively. MRC-5 lung fibroblasts
expression in lung mesenchymal cells. 16HBE14o- cells were grown to confluence and and airway smooth muscle cells were stimulated with increasing percentages of Fstl1 CM
transfected with either Fstl1-FLAG or CFP (control) to generate Fstl1 CM and CFP CM, (20; 40; 50% v/v) or CFP CM (50% v/v). The concentration of IL-6 and IL-8 was measured
respectively. MRC-5 lung fibroblasts and airway smooth muscle cells were stimulated with using ELISA in culture supernatants of CM-treated lung fibroblasts (A, B) and airway smooth
increasing percentages of Fstl1 CM (20; 40; 50% v/v) or CFP CM (50% v/v). The expression of muscle cells (C, D) and in the Fstl1 CM and CFP CM. Data represent mean ± SEM of 4-8
IL-6 and IL-8 mRNA was measured in cell lysates of lung fibroblasts (A, B) and airway smooth independent experiments. The concentration of IL-6 and IL-8 in culture supernatants of
muscle cells (C,D) using qPCR. Data represent mean ± SEM of 4-8 independent experiments. mesenchymal cells was corrected for the amount of IL-6 and IL-8 present in the CM. *p<0.05,
*p<0.05, ***p<0.001 compared to CFP CM controls. **p<0.01, ***p<0.001 compared to CFP CM controls.

84 85
Recombinant Fstl1 and modulation of Fstl1 expression have no effect on
inflammatory cytokine expression and release from lung fibroblasts
Lung fibroblasts were treated with recombinant human Fstl1, this was, however,
not sufficient to alter mRNA abundance or protein release of IL-6 and IL-8 from
lung fibroblasts (Figure 4A-4D). Since recombinant Fstl1 may not fully represent the
native Fstl1 due to differences in post-translational modifications, we used another
approach to investigate the direct effect of Fstl1 on inflammatory cytokine release by
modulating the endogenous Fstl1 expression using gain- and loss-of-function studies
in lung fibroblasts. Overexpression using a pcDNA3.1 containing Fstl1-FLAG resulted
in a 7.8-fold increase in Fstl1 mRNA (p<0.001; Figure 5A), whereas silencing of Fstl1
using FSTL1 siRNA resulted in a 61% decrease in Fstl1 mRNA (p<0.001; Figure 6A).
Despite significant upregulation or silencing of Fstl1, we observed no effect on basal
or IL-1β-induced IL-6 and IL-8mRNA abundance (Figure 5B-C and 6B-C) or protein
release (Figure 5D-E and 6D-E) from lung fibroblasts.

Figure 6. Silencing of Fstl1 did not affect IL-1β-induced IL-6 and IL-8 mRNA expression
and protein release from lung fibroblasts. MRC-5 were transfected with either FSTL1
siRNA or scrambled siRNA (control) and treated with IL-1β (0.01, 0.1 and 1 ng/ml) for 24
h. (A) Verification of FSTL1 silencing (black bars) in lung fibroblasts compared to scrambled
siRNA controls (white bars). IL-6 and IL-8 mRNA expression in cell lysate (B,C) and protein 5
concentration in culture supernatants (D,E) were measured using qPCR and ELISA, respectively.
Data represent mean ± SEM of 3 independent experiments. *p<0.05, ***p<0.001 compared
to scrambled siRNA non-treated controls; n.s. = non-significant.

Levels of GM-CSF and IL6 are increased in Fstl1 CM derived from bronchial epithelial
cells
Our findings suggest that bronchial epithelium-derived mediators present in the
culture supernatants from Fstl1-overexpressing bronchial epithelial cells, but
not secreted Fstl1 itself, may promote inflammatory cytokine release from lung
mesenchymal cells. We tested whether Fstl1 induces inflammatory mediators from
bronchial epithelial cells which subsequently increase IL-6 and IL-8 release from lung
fibroblasts. To identify inflammatory mediators that are responsible for the Fstl1
CM-induced cytokine release from lung fibroblasts, we screened for factors that are
differentially present in Fstl1 CM compared to CFP CM using a human cytokine array
that screens for 29 cytokines (Table 2). Overexpression of Fstl1 in bronchial epithelial
cells increased levels of RANTES, IL-6, IL-8, and granulocyte-macrophage colony-
stimulating factor (GM-CSF), as evident from their higher intensity compared to CFP
Figure 5. Overexpression of Fstl1 did not affect IL-1β-induced IL-6 and IL-8 mRNA
CM array data (Figure 7A). Subsequent studies demonstrated that the mRNA levels
expression or protein release from lung fibroblasts. MRC-5 lung fibroblasts were for RANTES, and GM-CSF were indeed selectively increased in bronchial epithelial
transfected with either Fstl1-FLAG or CFP (control) and treated with IL-1β (0.01, 0.1 and 1 cellsthat overexpressFstl1, and released protein levels in culture media was also
ng/ml) for 24 h. (A) Verification of Fstl1-FLAG overexpression (black bars) in lung fibroblasts
compared to CFP controls (white bars). IL-6 and IL-8 mRNA expression in cell lysate (B,C) increased for IL-6 and GM-CSF (Figure 7B-I).
and protein concentration in culture supernatants (D,E) were measured using qPCR and
ELISA, respectively. Data represent mean ± SEM of 3 independent experiments. ***p<0.001
compared to CFP non-treated controls; n.s. = non-significant.

86 87
Neutralizing antibodies against GM-CSF, RANTES, IL-6 and IL-8 attenuate Fstl1 CM- either IL-6 (p<0.01) or IL-8 (p<0.05) (Figure 8A). The combination of GM-CSF, RANTES,
induced cytokine release from lung fibroblasts IL-6 and IL-8 neutralizing antibodies completely attenuated Fstl1 CM-induced IL6
To elucidate which bronchial epithelial-derived factors mediate Fstl1 CM-induced release from lung fibroblasts (p<0.001; Figure 8A). Conversely, Fstl1 CM retained
cytokine release from lung fibroblasts, we treated CM with neutralizing antibodies capacity to induce IL-8 release from lung fibroblasts despite pre-treatment with
alone or in combination prior to the addition of the CM to lung fibroblasts. IgG neutralizing antibodies against IL-6 (p=0.038), IL-8 (p=0.034), or RANTES (p=0.02)
isotype control-treated Fstl1 CM significantly increased IL-6 release (p<0.001) and alone (Figure 8B). Importantly, a combination of neutralizing antibodies for GM-CSF,
IL-8 release (p<0.01) from lung fibroblasts. However, Fstl1 CM-induced IL-6 release RANTES, IL-6 and IL-8 did completely dampened the Fstl1 CM-induced IL-8 release
from lung fibroblasts was significantly reduced by neutralizing antibodies against from lung fibroblasts (p<0.05, Figure 8B).

Figure 7. Inflammatory mediators are increased in culture supernatants of Fstl1


overexpressing bronchial epithelial cells. Fstl1 CM derived from Fstl1 overexpressing Figure 8.GM-CSF, RANTES, IL-6 and IL-8 neutralizing antibodies attenuate Fstl1 CM-induced
16HBE14o- cells was analyzed using human cytokine arrays compared to CFP CM controls. cytokine release from lung fibroblasts.Fstl1 CM (50% v/v, black bars) or CFP CM (50% v/v,
(A) Representative membranes show that RANTES, IL-6, IL-8 and GM-CSF were increased in white bars) were pre-incubated with antibodies targeting RANTES, IL-6, IL-8, GM-CSF or a
Fstl1 CM compared to CFP CM. The mRNA expression and protein concentration of RANTES combination of these neutralizing antibodies 1 h prior to addition to MRC-5 lung fibroblasts.
(B and C), IL-6 (D and E), IL-8 (F and G), GM-CSF (H and I) were in cell lysates and culture
supernatants, respectively, of Fstl1-FLAG overexpressing bronchial epithelial cells (black The protein concentration of IL-6 (A) andIL-8 (B) was measured in culture supernatants from
bars) compared to CFP controls (white bars). Data represent mean ± SEM of 6 independent lung fibroblasts after 24 h stimulation. Data represent mean ± SEM of 6 independent exper-
experiments. *p<0.05, ***p<0.001 compared to CFP overexpressing bronchial epithelial cell iments. **p<0.01, ***p<0.001 Fstl1 CM compared to CFP CM controls, #p<0.05, ##p<0.01,
controls; n.s. = non-significant. ###p<0.001 compared to IgG isotype control-treated Fstl1 CM.

88 89
Discussion alone did not completely inhibit this response. Intriguingly, neutralizing antibodies
selectively targeting IL-6 or IL-8 were able to diminish IL-6 release from lung
The pathogenesis of respiratory diseases has been linked to chronic inflammation of fibroblasts induced by Fstl1 CM, suggesting autocrine/paracrine regulation of these
the lungs in response to epithelial injury. The airway epithelium is the first protective cytokines.
barrier of the internal lung tissue from the external environment. Upon damage, We demonstrate here the first evidence on a direct link between Fstl1 and GM-
the epithelium releases inflammatory mediators to communicate with neighboring CSF or RANTES. Fstl1 overexpression increases GM-CSF gene expression and protein
cells as part of the defense and repair mechanism.3,15 Fstl1 has been implicated in release from bronchial epithelial cells. Though GM-CSF was originally identified
the pathogenesis of several inflammatory diseases and has been demonstrated to as a key regulator governing the proliferation and maturation of granulocyte and
modulate inflammatory responses in mouse models of inflammatory diseases.9,10,16 macrophage lineages,22 a growing body of evidence indicates that GM-CSF is also an
Recent in vitro, in vivo, and proteomic studies demonstrate the involvement of inflammatory mediator capable of modulating neutrophil migration and survival.22–25
Fstl1 in the pathological process of respiratory diseases, including lung fibrosis Plasma cytokine levels of GM-CSF is significantly upregulated in patients with COPD,
and asthma.17–20 Fstl1 has previously been identified as a mesenchymal-derived in particular during chronic bronchitis exacerbations.26–29 Overexpression of GM-CSF
inflammatory protein in joints.16,21 However, our data indicate that in the respiratory triggers lung inflammation and fibrosis in mice,30,31 whereas a neutralizing antibody
system, bronchial epithelial cells express and release Fstl1 in much greater amount against GM-CSF decreases neutrophil number in the bronchoalveolar lavage fluid
than mesenchymal cells. To test the functional relevance of Fstl1 in inflammatory (BALF) of a mouse model of cigarette smoke-induced inflammation.25,32 Moreover, as
cytokine release in the lung, we overexpressed Fstl1 in bronchial epithelial cells and overexpression of Fstl1 increases RANTES mRNA levels but does not induce RANTES
demonstrated that Fstl1 overexpression induces inflammatory cytokine release. release from bronchial epithelial cells, suggesting the effects of Fstl1 CM on RANTES
Moreover, Fstl1 CM induces significant cytokine release from lung mesenchymal biosynthesis is complex. RANTES, also known as CC chemokine ligand 5 (CCL5) is a
cells, likely mediated by GM-CSF, RANTES, IL-6 and IL-8 derived from bronchial chemotactic activator of eosinophils,33 that has been associated with asthma,34–36
epithelial cells. lung allergic inflammation, and bronchial hyperresponsiveness.33 5
Epithelial cells communicate with mesenchymal cells as part of their defense Fstl1 can signal via different signaling pathways, including bone morphogenetic
system and a role for Fstl1 in cell-cell communication has been previously protein (BMP) receptors,4,5 DIP2 disco-interacting protein 2 homolog A,6,37 toll-like
suggested.9,17 To investigate whether bronchial epithelial-derived Fstl1 affects the receptor 4 (TLR4),12 and Na+/K+-ATPase.8 As a collective, our findings and existing
inflammatory response in mesenchymal cells, we treated lung fibroblasts and airway evidence suggest that future studies are needed to delineate the mechanism
smooth muscle cells with medium-derived from Fstl1 overexpressing bronchial underlying differential Fstl1 effects on cytokine release from bronchial epithelial
epithelial cells (Fstl1 CM). We hypothesized that Fstl1-derived bronchial epithelial is cells and lung mesenchymal cells.
sufficient to mediate inflammatory cytokine release from mesenchymal cells. Fstl1 Although the exact mechanism of Fstl1 action in cytokine expression and release
CM increased IL-6 and IL-8 mRNA expression and protein release from mesenchymal remains to be elucidated, our study provides the first in vitro demonstration that
cells, suggesting that Fstl1 may regulate pro-inflammatory cytokine release in the Fstl1 induces the transcription and release of inflammatory cytokines, such as GM-
lung. These observations are in line with previous studies demonstrating that Fstl1 CSF and IL-6, in bronchial epithelial cells, which subsequently induce IL-6 and IL-8
promotes IL-6 and IL-8 release from mesenchymal cells.7,9,11,12 However, recombinant release from mesenchymal cells. We reveal epithelial Fstl1 to be a pro-inflammatory
Fstl1 treatment and direct modulation of Fstl1 gene expression in lung fibroblasts factor that mediates modulation of mesenchymal cell secretory function, specifically
showed no effect on baseline or IL-1β-induced IL-6 and IL-8 protein release, indicating inflammatory cytokine release. As such, Fstl1 might contribute to the recruitment of
that there are additional factors involved in the lung fibroblast response to Fstl1 CM. inflammatory cells to the lung. These findings provide a basis for future elucidation
Our data points to a paradigm in which Fstl1 induces the release of inflammatory of the role and mechanisms for Fstl1 control of airway biology and pathobiology in
mediators from bronchial epithelial cells that have a strong pro-secretory effect on health and disease.
nearby mesenchymal cells.
Using cytokine array we identified four potential factors that were increased
upon Fstl1 overexpression in bronchial epithelial cells, namely GM-CSF, RANTES,
IL-6, and IL-8. Our studies using neutralizing antibodies demonstrate that only the
combination of neutralizing antibodies targeting GM-CSF, RANTES, IL6, and IL8
completely blocked Fstl1 CM-induced IL-6 and IL-8 release from lung fibroblasts.
Pre-treatment of Fstl1 CM with neutralizing antibodies against GM-CSF and RANTES

90 91
Funding
This study was financially supported by a grant from the Netherlands Lung Foundation
(grant 3.2.12.083). NPT is supported by the Netherlands Lung Foundation. AJH is
supported by the Canada Research Chairs Program. The funding agency had no
influence on study design, data analysis and interpretation, or preparation of this
manuscript.

Authors’ contributions
NPT designed and carried out the experiments, analyzed and interpreted the data,
and drafted the manuscript. VN performed part of the experiments and analyzed
the data. MS, HM, RG, and MvdH designed the study and interpreted the data.
AJH kindly provided immortalized airway smooth muscle cells. MvdH generously
provided the Fstl1-FLAG and CFP plasmids. All authors revised the manuscript
critically for important intellectual content and approved the final version.

92 93
References 22. Hamilton JA. GM-CSF in inflammation and autoimmunity. Trends Immunol. 2002;23(8):403-408.
1. Rusznak C, Mills PR, Devalia JL, Sapsford RJ, Davies RJ, Lozewicz S. Effect of cigarette smoke on doi:10.1016/S1471-4906(02)02260-3.
the permeability and IL-1beta and sICAM-1 release from cultured human bronchial epithelial 23. Bozinovski S, Jones JE, Vlahos R, Hamilton JA, Anderson GP. Granulocyte/Macrophage-Colony-
cells of never-smokers, smokers, and patients with chronic obstructive pulmonary disease. Am J stimulating Factor (GM-CSF) Regulates Lung Innate Immunity to Lipopolysaccharide through Akt/
Respir Cell Mol Biol. 2000;23(4):530-536. doi:10.1165/ajrcmb.23.4.3959. Erk Activation of NFκB and AP-1 in Vivo. J Biol Chem. 2002;277(45):42808-42814. doi:10.1074/
2. Alkhouri H, Poppinga WJ, Tania NP, Ammit A, Schuliga M. Regulation of pulmonary jbc.M207840200.
inflammation by mesenchymal cells. Pulm Pharmacol Ther. 2014;29(2):156-165. doi:10.1016/j. 24. Gomez-Cambronero J, Horn J, Paul CC, Baumann MA. Granulocyte-macrophage colony-
pupt.2014.03.001. stimulating factor is a chemoattractant cytokine for human neutrophils: involvement of the
3. Osei ET, Brandsma C-A, Noordhoek JA, Timens W, Postma D, Heijink I. Crosstalk between ribosomal p70 S6 kinase signaling pathway. J Immunol Baltim Md 1950. 2003;171(12):6846-
epithelium and fibroblasts; implications for COPD. Eur Respir J. 2014;44(Suppl 58):P3899. 6855.
4. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP) 25. Vlahos R, Bozinovski S, Hamilton JA, Anderson GP. Therapeutic potential of
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A. treating chronic obstructive pulmonary disease (COPD) by neutralising granulocyte
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108. macrophage-colony stimulating factor (GM-CSF). Pharmacol Ther. 2006;112(1):106-
5. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal 115. doi:10.1016/j.pharmthera.2006.03.007.
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616. 26. D’Armiento JM, Scharf SM, Roth MD, et al. Eosinophil and T cell markers predict
6. Ouchi N, Asaumi Y, Ohashi K, et al. DIP2A functions as a FSTL1 receptor. J Biol Chem.
functional decline in COPD patients. Respir Res. 2009;10:113. doi:10.1186/1465-9921-
2010;285(10):7127-7134. doi:10.1074/jbc.M109.069468.
7. Fan N, Sun H, Wang Y, et al. Follistatin-Like 1: A Potential Mediator of Inflammation in Obesity. 10-113.
Mediators Inflamm. 2013;2013:e752519. doi:10.1155/2013/752519. 27. Balbi B, Bason C, Balleari E, et al. Increased bronchoalveolar granulocytes and
8. Li D, Wang Y, Xu N, et al. Follistatin-like protein 1 is elevated in systemic autoimmune diseases granulocyte/macrophage colony-stimulating factor during exacerbations of chronic
and correlated with disease activity in patients with rheumatoid arthritis. Arthritis Res Ther. bronchitis. Eur Respir J. 1997;10(4):846-850.
2011;13(1):R17. doi:10.1186/ar3241. 28. Stämpfli MR, Wiley RE, Neigh GS, et al. GM-CSF transgene expression in the airway
9. Miyamae T, Marinov AD, Sowders D, et al. Follistatin-Like Protein-1 Is a Novel Proinflammatory allows aerosolized ovalbumin to induce allergic sensitization in mice. J Clin Invest.
Molecule. J Immunol. 2006;177(7):4758-4762. doi:10.4049/jimmunol.177.7.4758. 1998;102(9):1704-1714.
10. Clutter SD, Wilson DC, Marinov AD, Hirsch R. Follistatin-Like Protein 1 Promotes Arthritis by Up- 5
29. Saha S, Doe C, Mistry V, et al. Granulocyte-macrophage colony-stimulating factor
Regulating IFN-γ. J Immunol. 2009;182(1):234-239. doi:10.4049/jimmunol.182.1.234.
expression in induced sputum and bronchial mucosa in asthma and COPD. Thorax.
11. Chaly Y, Marinov AD, Oxburgh L, Bushnell DS, Hirsch R. FSTL1 promotes arthritis in mice by
enhancing inflammatory cytokine/chemokine expression. Arthritis Rheum. 2012;64(4):1082- 2009;64(8):671-676. doi:10.1136/thx.2008.108290.
1088. doi:10.1002/art.33422. 30. Xing Z, Ohkawara Y, Jordana M, Graham F, Gauldie J. Transfer of granulocyte-macrophage
12. Murakami K, Tanaka M, Usui T, et al. Follistatin-related protein/follistatin-like 1 evokes an colony-stimulating factor gene to rat lung induces eosinophilia, monocytosis, and
innate immune response via CD14 and toll-like receptor 4. FEBS Lett. 2012;586(4):319-324. fibrotic reactions. J Clin Invest. 1996;97(4):1102-1110.
doi:10.1016/j.febslet.2012.01.010. 31. Lei XF, Ohkawara Y, Stämpfli MR, et al. Compartmentalized transgene expression of
13. Gosens R, Zaagsma J, Meurs H, Halayko AJ. Muscarinic receptor signaling in the pathophysiology granulocyte-macrophage colony-stimulating factor (GM-CSF) in mouse lung enhances
of asthma and COPD. Respir Res. 2006;7:73. doi:10.1186/1465-9921-7-73. allergic airways inflammation. Clin Exp Immunol. 1998;113(2):157-165.
14. Ruijter JM, Ramakers C, Hoogaars WMH, et al. Amplification efficiency: linking baseline and bias 32. Vlahos R, Bozinovski S, Chan SPJ, et al. Neutralizing granulocyte/macrophage colony-
in the analysis of quantitative PCR data. Nucleic Acids Res. 2009;37(6):e45. doi:10.1093/nar/
stimulating factor inhibits cigarette smoke-induced lung inflammation. Am J Respir Crit
gkp045.
15. Knight D. Epithelium–fibroblast interactions in response to airway inflammation. Immunol Cell Care Med. 2010;182(1):34-40. doi:10.1164/rccm.200912-1794OC.
Biol. 2001;79(2):160-164. doi:10.1046/j.1440-1711.2001.00988.x. 33. Conti P, DiGioacchino M. MCP-1 and RANTES are mediators of acute and chronic
16. Ouchi N, Oshima Y, Ohashi K, et al. Follistatin-like 1, a secreted muscle protein, promotes inflammation. Allergy Asthma Proc Off J Reg State Allergy Soc. 2001;22(3):133-137.
endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase- 34. Fryer AA, Spiteri MA, Bianco A, et al. The -403 G-->A promoter polymorphism in the
dependent mechanism. J Biol Chem. 2008;283(47):32802-32811. doi:10.1074/jbc.M803440200. RANTES gene is associated with atopy and asthma. Genes Immun. 2000;1(8):509-514.
17. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary doi:10.1038/sj.gene.6363717.
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878. 35. Al-Abdulhadi SA, Helms PJ, Main M, Smith O, Christie G. Preferential transmission and
18. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by Inducing association of the |[minus]|403 G → A promoter RANTES polymorphism with atopic
Oncostatin M. J Immunol Baltim Md 1950. September 2015. doi:10.4049/jimmunol.1501105.
asthma. Genes Immun. 2004;6(1):24-30. doi:10.1038/sj.gene.6364151.
19. Miller M, Esnault S, Kurten RC, et al. Segmental allergen challenge increases levels of airway
follistatin-like 1 in patients with asthma. J Allergy Clin Immunol. 2016;138(2):596-599.e4. 36. Hizawa N, Yamaguchi E, Konno S, Tanino Y, Jinushi E, Nishimura M. A Functional
doi:10.1016/j.jaci.2016.01.019. Polymorphism in the RANTES Gene Promoter Is Associated with the Development of
20. Murphy N, Gaynor KU, Rowan SC, et al. Altered Expression of Bone Morphogenetic Protein Late-Onset Asthma. Am J Respir Crit Care Med. 2002;166(5):686-690. doi:10.1164/
Accessory Proteins in Murine and Human Pulmonary Fibrosis. Am J Pathol. 2016;186(3):600- rccm.200202-090OC.
615. doi:10.1016/j.ajpath.2015.10.032. 37. Tanaka M, Murakami K, Ozaki S, et al. DIP2 disco-interacting protein 2 homolog A
21. Wilson DC, Marinov AD, Blair HC, et al. Follistatin-like protein 1 is a mesenchyme-derived (Drosophila) is a candidate receptor for follistatin-related protein/follistatin-like
inflammatory protein and may represent a biomarker for systemic-onset juvenile rheumatoid 1--analysis of their binding with TGF-β superfamily proteins. FEBS J. 2010;277(20):4278-
arthritis. Arthritis Rheum. 2010;62(8):2510-2516. doi:10.1002/art.27485. 4289. doi:10.1111/j.1742-4658.2010.07816.x.

94 95
CHAPTER 6 1

Regulation of pulmonary inflammation by


mesenchymal cells

Hatem Alkhouri, Wilfred J. Poppinga, Navessa P. Tania, Alaina Ammit,


Michael Schuliga

97
Regulation of pulmonary inflammation by mesenchymal cells Abstract

Pulmonary inflammation and tissue remodelling are common elements of chronic


respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD),
Hatem Alkhouri1, Wilfred J. Poppinga2,3, Navessa P. Tania2,3, Alaina Ammit1 andMi- idiopathic pulmonary fibrosis (IPF), and pulmonary hypertension (PH). In disease,
chael Schuliga4,5 pulmonary mesenchymal cells not only contribute to tissue remodelling, but also
have an important role in pulmonary inflammation. This review will describe the
immunomodulatory functions of pulmonary mesenchymal cells, such as airway
smooth muscle (ASM) cells and lung fibroblasts, in chronic respiratory disease.
An important theme of the review is that pulmonary mesenchymal cells not only
Respiratory Research Group, Faculty of Pharmacy, University of Sydney, Sydney,
1
respond to inflammatory mediators, but produce their own mediators, whether
New South Wales, Australia pro-inflammatory or pro-resolving, which influence the quantity and quality of the
lung immune response. The notion that defective pro-inflammatory or pro-resolving
Department of Molecular Pharmacology, University of Groningen, Groningen, The
2
signalling in these cells potentially contributes to disease progression is also
Netherlands
discussed. Finally, the concept of specifically targeting pulmonary mesenchymal cell
3
Groningen Research Institute of Asthma and COPD (GRIAC), University of Gronin- immunomodulatory function to improve therapeutic control of chronic respiratory
gen, Groningen, University Medical Centre Groningen, The Netherlands disease is considered.

4
Deptartment of Pharmacology and Therapeutics, University of Melbourne,
Parkville, Victoria, Australia
5
Lung Health Research Centre, University of Melbourne, Parkville, Victoria, Austra-
lia
6

98 99
1. Introduction by the synthesis and deposition of collagens I and III and other ECM components
(e.g. fibronectin), expand the volume of the ECM in the sub-epithelial layer of the
Worldwide, more than 250 million people suffer from a debilitating or lethal airway wall, within ASM bundles or in the lung interstitium.15 Aside from important
chronic respiratory disease,1 such as asthma, chronic obstructive pulmonary disease biomechanical contributions in tissue remodelling, pulmonary mesenchymal
(COPD), idiopathic pulmonary fibrosis (IPF) or pulmonary hypertension (PH). cells are also potent producers of an array of inflammatory mediators, including
Asthma, characterized by airway inflammation, remodelling and hyper-reactivity, is cytokines, chemokines and cell adhesion molecules (CAMs).16–19 These inflammatory
one of the most prevalent chronic respiratory diseases, causing ~1/4 of a million mediators, as well as the ECM produced by pulmonary mesenchymal cells, influence
deaths per year globally.1 COPD, comprised of irreversible breakdown of lung tissue the type and quantity of inflammatory cells that infiltrate airway and lung tissue in
(emphysema) and airway wall remodelling, contributes to ~3 million deaths per year, chronic respiratory disease. Furthermore, the potential importance of inflammatory
and is increasing in incidence.1,2 IPF, albeit less common than asthma or COPD, is a responses regulated by pulmonary mesenchymal cells in tissue remodelling is
lethal interstitial lung disease characterised by a relentlessly progressive and invasive becoming increasingly recognised. In this review, the immunomodulatory functions
form of lung parenchymal fibrosis.3 Secondary PH, a comorbidity caused primarily of pulmonary mesenchymal cells and their potential roles in the progression of
by hypoxia in lung disease, features increased pulmonary vascular resistance.4 There chronic respiratory disease will be described.
remains no effective treatment for severeasthma (5-10% of asthmatics), COPD and
IPF.5
The consistent presence of inflammatory cells in the lungs of patients
unequivocally establishes pulmonary inflammation as an important component
of chronic respiratory disease. The lung inflammatory profiles of patients vary
depending on the disease and severity, and change upon exacerbation.6–8 Airway
inflammation in asthma is associated with an increase in mast cells, eosinophils
and CD4+ T-helper-2 (Th2) lymphocytes. However, for asthmatics with fixed airway
obstruction, the inflammation is more neutrophilic with greater CD8+ T-helper-1
(Th1) cell involvement, akin to COPD, which is also characterised by fixed airway
obstruction.7 Whilst IPF has a predominant Th2 cell profile, the ratio of CD8+ to 6
CD4+lymphocytes increases with disease severity.8 Like COPD, neutrophils and
macrophages are also present in lung tissue of patients with IPF. In PH, perivascular
infiltration of dendritic cells, macrophages, mast cells, T-lymphocytes (CD4+ and CD8+)
and B-lymphocytes occurs.9 In chronic respiratory disease, infiltrating inflammatory
cells produce an array of inflammatory mediators which act by autocrine and
paracrine mechanisms to not only regulate inflammatory cell function, but also
pulmonary mesenchymal cells in tissue remodelling.
In chronic respiratory disease, there is an important relationship between
inflammation and tissue remodelling. The latter describes the structural changes in
lung tissue which may contribute to respiratory dysfunction. Pulmonary mesenchymal Figure 1. Immunomodulatory functions of pulmonary mesenchymal cells. The solid
black arrows designate the pro-inflammatory mediators produced directly or indirectly by
cells are structural cells with a well-recognised role in tissue remodelling processes pulmonary mesenchymal cells which contribute to pulmonary inflammation in disease.
in disease. In asthma and COPD, airway smooth muscle (ASM) cell hyperplasia and The black hatched arrows represent sources of pro-inflammatory mediators which regulate
hypertrophy cause ASM enlargement, whereas airway fibroblasts contribute to pulmonary mesenchymal cell function, including the production and expression of pro-
inflammatory mediators. The types and phenotype of the pulmonary mesenchymal and
sub-epithelial fibrosis in the airway wall.10,11 In IPF, lung fibroblasts lung fibroblasts1 inflammatory cells varies for disease and disease severity. Abbreviations are defined in the
have an integral role in the progressive fibrosis which begins in the lung interstitium text.
and invades alveoli spaces.12 In PH, pulmonary vascular smooth muscle cells have a
prominent role in the medial enlargement of blood vessels, which in effect reduces
lumen size, increasing vascular resistance.13 Abnormalities of the extracellular matrix
(ECM) are a key feature of tissue remodelling in lung disease.14 Mesenchymal cells,

100 101
2. Immunomodulatory function of pulmonary mesenchymal cells Table 1. Immunomodulatory proteins expressed by airway smooth muscle (ASM)
This section will provide an overview of the types of immunomodulatory functions cells and lung fibroblasts.
of pulmonary mesenchymal cells, as summarized in Figure 1. Type Protein Pulmonary mesenchymal cell
Cytokines IL-1 ASM23, lung fibroblasts19
2.1. Pro-inflammatory mediators
IL-4 Lung fibroblasts24
Pulmonary mesenchymal cells coordinate inflammatory responses by producing
IL-6 ASM,25 lung fibroblasts24
pro-inflammatory mediators which lead to inflammatory cell recruitment and IL-10 ASM23
activation. The production of pro-inflammatory mediators by ASM cells, particularly IL-11 ASM25
in the context of asthma, has been extensively studied and the subject of many IL-13 Lung fibroblasts26
reviews, including one recent review.20 Table 1 provides an overview of the broad GM-CSF ASM,27 lung fibroblasts19,28
range of cytokines, chemokines and CAMs, which have been shown to be expressed LIF ASM29
by pulmonary mesenchymal cells, primarily in in vitro cell culture studies. The ECM OX40 ligand ASM30
produced by these cells also influences inflammatory cell recruitment. Versican Chemokines CXCL1 (Gro-α) ASM,31 lung fibroblasts32
and hyaluronan for instance are ECM components produced by lung fibroblasts CXCL5 (ENA-78) Lung fibroblasts32
which regulate T-cell trafficking and functioning in inflamed lung tissue.21,22 Pro- CXCL6 (GCP-2) ASM23
inflammatory mediator expression in pulmonary mesenchymal cells is stimulated CXCL8 (IL-8) ASM,33 lung fibroblasts19,26
primarily by cytokines and growth factors produced by inflammatory cells and the CXCL9 (MIG) ASM34
CXCL10 (IP-10) ASM,35 lung fibroblasts36
epithelium.20 The regulation of immunomodulatory function of these cells also
CXCL11(ITAC) ASM34
involves pro-resolving mediators (section 2.2), the innate immune system (section
CXCL12 (SDF-1α) ASM,34 lung fibroblasts19
2.3), the plasminogen activation system (section 2.4) and the coagulation system
CCL2 (MCP-1) ASM,37 lung fibroblasts19
(section 2.5).
CCL3 (MIP-1α) ASM38
CCL5 (RANTES) ASM,39 lung fibroblasts19,26
2.2. Pro-resolving mediators CCL4 (MIP-1β) ASM23
Pulmonary mesenchymal cells may be targets for or produce mediators which CCL7 (MCP-3) ASM,38 lung fibroblasts40
have a role in resolving inflammation. Most pro-resolving mediators with anti- CCL8 (MCP-2) ASM37 6
inflammatory activity, including the resolvins, protectins and lipoxins, are derived CCL11 (Eotaxin) ASM,41 lung fibroblasts42
from dietary omega-3 polyunsaturated fatty acids.53 Administration of pro-resolving CCL16 (MTN-1) ASM23
lipid mediators, including protectin D1, resolvin D1 and resolvin E1, are protective CCL17 (TARC) ASM43
in models of lung injury and disease.54–57 Endogenous protectin D1 is increased CCL19 (MIP-3) ASM44
in the airways in response to allergen challenge, but less so for asthmatics than CCL20 (MIP-3α) ASM45
non-asthmatics.55,58 Such observations suggest that dys-regulated production of CX3CL1 (Fractalkine) ASM46
pro-resolving lipid mediators may contribute to chronic respiratory disease.59 SCF ASM47
CAMs ICAM-1 ASM,48 lung fibroblasts49
Whilst inflammatory cells are a major source of pro-resolving lipid mediators,55,58,59
VCAM-1 ASM,19,48 lung fibroblasts49
pulmonary mesenchymal cells are a target. In cultures of human lung fibroblasts,
CD40 ASM19,50
resolvin D1 inhibits cigarette smoke extract- and IL-1β-induced cytokine release60
CD44 ASM51
and endotoxin-induced COX-2 expression and prostaglandin E2 (PGE2) production.61
CD90 (Thy-1) Lung fibroblasts52
Current knowledge about the release of pro-resolving mediators by pulmonary
mesenchymal cells is limited, aside from the production of annexin A1, an anti- Abbreviations: CD, cluster of differentiation; ENA, epithelial-derived neutrophil activating;
inflammatory protein.24 Annexin A1, like resolvin D1, is a ligand for the lipoxin GM-CSF, granulocyte macrophage-colony stimulating factor; ICAM, intracellular adhesion
molecule; IL, interleukin; LIF, leukaemia inhibitory factor; SCF, Stem cell factor; VCAM,
A4 receptor, ALX/FPR2. Annexin A1 expression and release is increased in lung vascular cell adhesion molecule.
fibroblasts following treatment with glucocorticoids.24 Furthermore, the silencing of
annexin A1 augments TNFα-induced IL-6 release from lung fibroblasts,24 suggesting

102 103
that annexin A1 production may be an important immunomodulatory function of thrombin, the main activator of the coagulation system, TLR4-activating fibrinogen
pulmonary mesenchymal cells. cleavage products are generated. Interestingly, plasmin is also involved in the
formation of fibrinogen cleavage products,87 suggesting that the convergence of both
2.3. Toll like receptors (TLRs) the coagulation and plasminogen activation systems may play an important role in
Toll-like receptors (TLRs) activate the innate immune system in response to infection pulmonary inflammation in disease. Thrombin and factor Xa (FXa), another coagulant,
and tissue injury. TLR ligands are: (i) derived from pathogens, including bacterial also activate PAR receptors, including those on ASM cells and lung fibroblasts,88,89 to
cell-surface lipopolysaccharides (LPS) and the double stranded RNA of viruses; or elicit pro-inflammatory and remodelling activities.89–91 The targeting of thrombin or
(ii) formed endogenously, such as fibrinogen62 and annexin A2.63 The binding of FXa reduces pulmonary inflammation and tissue remodelling in murine models of
TLR ligands to their receptors leads to the activation of nuclear factor NF-κB and/or lung injury and disease.89,92,93
interferon regulatory transcription factor 3/7, which stimulates the gene expression
of inflammatory mediators. The dysregulation of TLR signalling may contribute to 3. Pulmonary mesenchymal cells in chronic respiratory disease
the development of chronic respiratory disease. The activation of TLR4 by fibrinogen 3.1. Asthma
cleavage products of coagulation proteases possibly contributes to asthma In asthma, allergen-induced airway inflammation contributes to airway hyper-
pathophysiology.62 Pulmonary mesenchymal cells express TLR2,64 TLR3,65 TLR4,66 and responsiveness (AHR), a process that involves spasmodic ASM contraction.
TLR967 and their activation stimulates IL-6, IL-8 and eotaxin production.68,69 ASM cells Inflammation has direct and indirect roles in AHR, causing vascular leakage, mucus
release the stress-response protein, annexin A2, which stimulates IL-6 production hyper-secretion, epithelial shedding, ASM thickening and sub-epithelial fibrosis.94
in both macrophages70 and ASM cells63 via TLR4. In lung fibroblasts, TLR3 activation Pro-inflammatory mediators produced by ASM cells and airway fibroblasts, including
stimulates the production of RANTES, IP-10, IL-8, type 1 IFN, TGF-β, IL-4 and IL-13,26,71 IL-8, IP-10, MIP-1α, RANTES and eotaxin, contribute to the recruitment of mast cells,
and TLR4 regulates proliferation.72 lymphocytes, eosinophils and neutrophils in asthma.34,95–97 ASM abnormalities in
asthmatics may contribute to an increased hyper-secretory phenotype. Cytokine-
2.4. The plasminogen activation system induced production of IL-8,98 IP-10,34 ITAC,34 eotaxin99 and MIP-3α100 is greater in
In interstitial lung tissue, the conversion of plasminogen to plasmin (“activation”), a cultures of ASM cells obtained from asthmatic donors than non-asthmatics donors.
pro-inflammatory serine protease,73 contributes to disease.74 Plasminogen, a plasma Furthermore, ASM cells of asthmatics produce relatively more collagen, fibronectin
protein, is relevant in lung pathology as vascular leak leads to its extravasation into and fibulin-1,101–103 ECM proteins which may facilitate inflammatory cell adhesion 6
inflamed lung tissue. Both lung fibroblasts and ASM cells activate extracellular and activation. Increased calcium handling, caused by abnormal sarco/endoplasmic
plasminogen with subsequent effects on IL-6 and IL-8 production and cell reticulum calcium ATPase (SERCA) pump function and expression, increases eotaxin
proliferation.63,75–77 These effects occur at low µg/mL concentrations of plasminogen, expression in ASM cells of asthmatics.104,105 Furthermore, JNK signalling and STAT-1
substantially lower than that detected in plasma. At higher concentrations of activation is diminished,106,107 whilst p65 NF-κB activation is higher98,107 in ASM cells
plasminogen, increased PGE2 synthesis and/or apoptosis are observed.63,78 For ASM of asthmatic than non-asthmatic donors. Additionally, TNF-α induced p38 mitogen-
cells, plasminogen activation is mediated by the urokinase plasminogen activator activated protein kinase (MAPK) signalling is greater in ASM cells of donors with
(uPA), in a manner accelerated by the annexin A2 hetero-tetramer (AIIt),63 an severe asthma, than other asthma or control groups.108 ASM cells of asthmatics lack
extracellular protein complex comprised of annexin A2 and S100A10 (p11). The expression of the full length transcription factor CCAAT/enhancer binding protein
AIIt also serves as a signal transducer for plasmin in mediating its pro-inflammatory (C/EBPα).109 As a consequence, C/EBPβ binding to chemokine promoters increases,
effects on ASM cells77 and macrophages.79 Whilst currently little is known about the causing cytokine hyper-secretion.98 This may be due to lack of an important anti-
role of annexin A2 in respiratory disease, it is becoming increasingly recognised for inflammatory protein, MAPK phosphatase 1 (MKP-1), a critical MAPK deactivator
its importance in cancer.80–84 Both uPA and annexin A2 may be novel drug targets in that is explored in greater depth in section 5.3. Reduced expression of MKP-1
the treatment of chronic respiratory disease74 (section 5.4). was responsible for over-activation of the p38 MAPK pathway and corticosteroid
insensitivity of alveolar macrophages in severe asthma compared with non-severe
2.5. The coagulation system asthma.110
The coagulation system also contributes to pulmonary inflammation in disease.62,85 Although airway fibroblast secretory function is an area of active research,28 asthma-
Like plasminogen, the inactive zymogens of coagulation proteases enter inflamed associated changes in airway fibroblast inflammatory mediator production is under-
lung tissue as a consequence of vascular leak, in a process accompanied by platelet explored. Whilst IL-1b-induced GM-CSF and IL-8 production is increased more in the
aggregation and activation of the coagulation cascade.86 Through the actions of airway fibroblasts of asthmatics than non-asthmatics,17 the mechanism behind this

104 105
differential cytokine production remains unknown. Interestingly, airway fibroblasts 3.4. Pulmonary hypertension
of asthmatics in culture express lower levels of IL-13 Rα2 (a decoy receptor for IL-13 PH, whether primary or secondary to an accompanying chronic respiratory disease
signalling) at baseline than airway fibroblasts of controls,111 possibly augmenting IL- is characterized by vasoconstriction, in situ thrombosis and pulmonary vascular
13-induced inflammation in asthma.112 remodelling. Hypoxia, chronic inflammation and shear stress contribute to PH
pathology.132,133 In proximal pulmonary vessels that were previously muscularized,
3.2. COPD medial thickening is caused by the hypertrophy, hyperplasia and ECM production of
COPD, characterized by shortness of breath, cough and mucus hyper-secretion, is resident pulmonary vascular smooth muscle cells. In previously non-muscular pre-
caused primarily by tobacco exposure. Genetics/epigenetics113,114 and the pulmonary capillary arterioles, the pulmonary vascular smooth muscle cells that contribute to
microbiome115 are also factors that may contribute to COPD. There is an increase in medial thickening are derived from intermediate cells in blood vessels or adventitial
ASM mass in COPD, albeit, primarily in the small airways.116 Studies investigating the fibroblasts, which differentiate into pulmonary vascular smooth muscle cells. In
role of ASM cells in COPD are under-represented when compared to asthma, most PH, the vascular adventitia has an important role in regulating and contributing
likely due to ASM-related pathology, such as AHR, being far more pronounced in to perivascular inflammation.134 Pulmonary adventitial fibroblasts, through the
asthma. In COPD, the number of airway fibroblasts with a more contractile phenotype production and release of pro-inflammatory mediators, induce the infiltration and
(myofibroblasts) is greater,21 likely caused by increases in the expression and activity activation of monocytes and macrophages. Epigenetic alterations in pulmonary
of rho-associated coiled-coil protein kinase 1 (ROCK1).117 Such increases will reduce adventitial fibroblasts from chronically hypoxic hypertensive calves are linked to a
airway elasticity, as will versican, the production of which is increased in airway heightened pro-inflammatory phenotype with the expression of IL-1β, IL-6, MCP-1,
fibroblasts of COPD patients.118,119 Airway fibroblasts of COPD patients also express CXCL12, RANTES, CCR7, CXCR4, GM-CSF and VCAM-1 being increased.19 In severe
higher levels of IL-6 and IL-8 than controls.119 Intriguingly, airway fibroblasts from PH, the epigenetic reprogramming of human pulmonary adventitial fibroblasts
COPD patients, and not from control subjects, produce prostacyclins in response to a pro-inflammatory phenotype is associated with the decreased expression of
to TGF-β.120 Prostacyclins have anti-inflammatory effects in pulmonary fibrosis and miR-124, which regulates Notch1/PTEN/FOXO3/p21Cip1 and p27Kip1 signalling.135
PH.121 Interestingly, aberrant PTEN phosphatase activity may also contribute to the pro-
inflammatory phenotype of pulmonary vascular smooth muscle cells in PH. PTEN,
3.3. IPF which inhibits Akt/PI3kinase signalling, regulates a number of cell processes including
Interstitial lung diseases (ILDs) are characterized by an abnormality in the inflammation.136 Selective deletion of the PTEN gene in pulmonary vascular smooth 6
interstitium, the area in the lung parenchyma between the capillaries and alveolar muscle cells increases macrophage infiltration and vascular remodelling in a murine
spaces. In IPF, a lethal form of ILD, the abnormality is a relentlessly progressive form model of PH.137
of fibrosis that causes irreversible damage of lung structure and function. Whilst an
increased number of inflammatory cells in the lungs of patients with IPF suggests a 4. Interactions between pulmonary mesenchymal cells and inflammatory cells
role of inflammation,122 an abnormal wound-repair response of epithelial/fibroblast 4.1. Mast cell-airway smooth muscle cell interactions
origin is thought to be an underlying cause.123 Lung fibroblasts have a pivotal Mast cell-ASM cell interactions have an important role in asthma pathophysiology.138
role in IPF, proliferating and differentiating into collagen producing, contractile The number of mast cells within the ASM layer of asthmatics is higher than non-
myofibroblasts to form fibroblastic foci. The expression of fibroblast growth factor asthmatics, correlating with disease severity.138–141 In asthma, the mast cells residing
(FGF9) is increased in IPF fibrotic foci in situ and lung fibroblasts of IPF patients in in the ASM layer are predominantly mast cellTC,35 being smaller and less granular30
vitro in response to TGF-β1.124 By contrast, IFN-inducible expression of STAT1 and compared to the mast cells found elsewhere in the airway wall, or within the ASM
IP-10 is repressed in lung fibroblasts of IPF patients.125 Human lung fibroblasts from layer of non-asthmatics. Mast cell recruitment to the ASM layer requires mast cell
IPF patients show constitutive activation of STAT3,52 which mediates oncostatin M expression of chemokine receptors and ASM cell production of chemokines. Lung
induced fibroblast chemotaxis.126 Oncostatin M is secreted by inflammatory cells, mast cells express a wide range of chemokine receptors, including CCR3, CXCR1,
such as macrophage and dendritic cells upon bacterial infection.127 Oncostatin M is 2, 3 and 4, with CXCR3 being the most highly expressed on mast cells within the
a potent mediator of pulmonary inflammation,128 and is involved in the induction of ASM layer in asthma.23,34 Important chemokines produced by ASM cells involved
pulmonary eosinophilia and goblet cell hyperplasia in mice,129,130 being up-regulated in mast cell recruitment include: IL-8 (binds CXCR1);142 IP-10 (binds CXCR3);23,34
in the lung of IPF patients. Furthermore, in lung fibroblasts, eotaxin expression is SDF-1α (binds CXCR4);143 RANTES (binds CCR1, 3 and 5);144 and eotaxin (binds
induced by oncostatin M, suggesting that lung fibroblasts play an important role in CCR3).142 These chemokines, in conjunction with SCF and TGF-β, are involved in
oncostatin M-induced inflammation in IPF.42,131 the movement of mast cells to the ASM layer.145 ASM cells of asthmatics produce

106 107
higher levels of IP-10 than ASM cells of non-asthmatics following treatment with Th1 5. Novel strategies to target pulmonary mesenchymal cell immunomodulatory
cytokines.34 Under Th2 inflammatory conditions, mast cell chemotaxis involves IL-8 function
and eotaxin.142 Interestingly, the ASM cells of non-asthmatics release factor(s) that
5.1. cAMP elevating agents
inhibit mast cell chemotaxis under either Th1 or Th2 inflammatory conditions.142
There is still a need to find new therapies for chronic respiratory diseases for which,
CXCL1 is an inhibitory factor of mast cell migration, and is produced less in ASM cells
anti-inflammatory glucocorticoids alone are ineffective.155 Roflumilast, an oral
of asthmatics than non-asthmatics.146 Upon ASM-mast cell contact, ASM cells induce
phosphodiesterase (PDE4) inhibitor, is an anti-inflammatory drug for COPD, but has
mast cell proliferation and maintain mast cell survival.147 This interaction is mediated
side effects including nausea. Interestingly, both PDE4 inhibitors and β2-adrenergic
by membrane-bound SCF expressed on ASM cells and soluble IL-6 and CADM1
receptor agonists cause a rise in intracellular second messenger cyclic AMP (cAMP),
produced by mast cells.147 In addition, numerous mast cell produced mediators
but are used pharmacologically for different targets, one inflammation, the other
directly affect ASM cell function, a topic that has been extensively reviewed.148–150
bronchoconstriction (in asthma and COPD). In cultures of normal human lung
These mediators cause exaggerated bronchoconstriction and also modulate ASM
fibroblasts, roflumilast, and the β2-agonist, indacaterol, act synergistically to attenuate
cell secretory function.146
inflammatory cytokine secretion and differentiation into a pro-fibrotic phenotype.156
The levels of PGE2, an endogenous lipid mediator that increases cAMP production,
4.2. Monocyte/macrophage-fibroblast interactions
are higher in lung157 and lung fibroblasts from COPD patients.158,159 However, in COPD,
In chronic respiratory disease, blood circulating monocytes infiltrate lung tissue
PGE2 effects on the cAMP pathway are reduced due to an increase in PDE4 activity.157
to differentiate into macrophages or dendritic cells. The phagocytic and antigen
As increased PDE4 activity also reduces β2-agonist effectiveness, these findings
presenting functions of these cells are important in innate and adaptive immunity
imply the potential benefit of combining PDE4 inhibitors with cyclic AMP elevating
respectively. Fibroblasts are ideally suited to regulate monocyte trafficking,
agonists. Interestingly, the addition of plasmin(ogen) to lung fibroblasts from
differentiation and functioning because of their synthetic capacity and sentinel-
IPF patients overcomes a similar PGE2 resistance, by rearranging the intracellular
like positioning in interstitial spaces. Monocytes stimulate GM-CSF production by
compartmentalization of the cAMP pathway.160 This rearrangement is mediated
lung fibroblasts in a manner involving physical contact between the two cell types.28
by an increased expression of the A-kinase anchoring protein AKAP9, a scaffolding
Lung fibroblast production of GM-CSF, which regulates monocyte/macrophage
protein for protein kinase A (PKA), which amplifies the downstream pathway of
function, is in turn increased by TNF-α and/or IL-1β, cytokines produced by activated
PGE2-cAMP-PKA.160 Thus the anti-inflammatory and anti-fibrotic properties of PGE2
macrophages. In mouse models of PH, pulmonary adventitial fibroblasts produce 6
on pulmonary mesenchymal cells may potentially be restored in chronic respiratory
soluble mediators, including GM-CSF, which influence monocyte/macrophage cell
diseases by approaches that rearrange cAMP compartmentalization.160
adhesion, infiltration and cytokine production.135 Airway fibroblasts from patients
with COPD express higher levels of the integrin αvβ8, which activates TGF-β, in
turn stimulating CCL2 and CCL20 production in airway fibroblasts by an autocrine 5.2. TGF- β1 pathways
manner.151 In a murine model of COPD, αvβ8-regulated CCL2 and CCL20 production Aberrant TGF-β1 signalling, important in regulating pulmonary mesenchymal cells
stimulates dendritic cell migration to boost an adaptive immune response.151 function, contributes to pulmonary inflammation and remodelling in disease.151,161,162
Alternatively activated (M2) macrophages, induced by Th2 cytokines (e.g. IL-4 and Inhibiting specific aspects of TGF-β1 signalling may be an effective strategy to treat
IL-13), are increasingly being recognised for their role in chronic respiratory disease. chronic respiratory disease.120 The TGF-β1 superfamily member, activin A, is linked
M2 activated macrophages are the pre-dominant macrophage phenotype present with the progression of PH, stimulating pulmonary vascular smooth muscle cell
in the lungs of IPF patients152 and are detected in higher numbers in the lung of proliferation.163 Administration of follistatin, an endogenous inhibitor of activin A
COPD patients who continue smoking than those who stop.153 The M2 macrophages attenuates inflammation and remodelling in a murine model of pulmonary fibrosis164
have an impaired role in innate immunity, but produce a myriad of pro-inflammatory and asthma.160 TGF-β1-inducible connective tissue growth factor (CTGF) is implicated
and pro-fibrogenic mediators such as TGF-β, IL-13, CCL2, CCL17, CCL18 and CCL22.152 in the pathogenesis of IPF. Inhibition of CTGF reduces collagen promoter activity
Alveolar macrophages from non-IPF donors produce more CCL18 when either and its expression in bleomycin-induced mice lung fibroblasts, suggesting CTGF
treated with Th2 cytokines, co-cultured with lung fibroblasts or exposed to native neutralization may be an option for the treatment of IPF.165 HS 6-O-sulfotransferases
collagen.154 The latter effect of collagen occurs in a manner mediated by the β2- 1 (HS6ST1) is up-regulated in lung fibroblasts of IPF patients,166 and its silencing
integrin.154 As CCL18 stimulates collagen production in lung fibroblasts, an axis reduces TGF-β1 activation and subsequent collagen I and α-smooth muscle actin
between M2 macrophages and lung fibroblasts, involving a CCL18-driven positive expression. Such data suggests that HS6ST1 inhibition could potentially reduce
feedback loop, may perpetuate fibrosis in IPF.154 TGF-β1 -mediated lung fibrosis. Interestingly, the IL-6 antagonist Sant7 attenuates
TGF-β1-induced proliferation of lung fibroblasts obtained from ILD patients,167

108 109
suggesting that targeting IL-6 may selectively block an important TGF-β1-mediated References
fibrotic response. Finally, inhibition of GSK-3, a mediator of TGF-β1-induced 1. Bousquet J, Khaltaev N. Global Surveillance, Prevention and Control of Chronic Respiratory
pulmonary mesenchymal cell differentiation, may also be a potential molecular Diseases - WHO - OMS -. WHO Press; 2007. http://apps.who.int/bookorders/anglais/detart1.
jsp?sesslan=1&codlan=1&codcol=15&codcch=708. Accessed March 29, 2017.
target for chronic lung diseases.168,169 2. Decramer M, Janssens W, Miravitlles M. Chronic obstructive pulmonary disease. Lancet Lond

Engl. 2012;379(9823):1341-1351. doi:10.1016/S0140-6736(11)60968-9.
5.3. MKP-1 3. Ding Q, Luckhardt T, Hecker L, et al. New insights into the pathogenesis and treatment of
In recent years, the important anti-inflammatory role played by the MAPK deactivator idiopathic pulmonary fibrosis. Drugs. 2011;71(8):981-1001. doi:10.2165/11591490-000000000-
00000.
MKP-1 in regulating inflammation in asthma has emerged. Upregulation of MKP-1 is 4. Shlobin OA, Nathan SD. Pulmonary hypertension secondary to interstitial lung disease. Expert
one of the ways in which common anti-asthma medicines, such as β2-agonists and Rev Respir Med. 2011;5(2):179-189. doi:10.1586/ers.11.11.
glucocorticoids, mediate their anti-inflammatory effects.31,170,171 MKP-1 is a critical 5. Durham A, Adcock IM, Tliba O. Steroid resistance in severe asthma: current mechanisms and
negative feedback controller, limiting the extent and duration of pro-inflammatory future treatment. Curr Pharm Des. 2011;17(7):674-684.
6. Tetley TD. Inflammatory cells and chronic obstructive pulmonary disease. Curr Drug Targets
MAPK-driven cellular signalling pathways in pulmonary mesenchymal cells such as Inflamm Allergy. 2005;4(6):607-618.
ASM.172,173 Without MKP-1, MAPK-mediated inflammation can continue unchecked 7. Tubby C, Harrison T, Todd I, Fairclough L. Immunological basis of reversible and fixed airways
with the clinical consequence in chronic respiratory disease underscored by recent disease. Clin Sci Lond Engl 1979. 2011;121(7):285-296. doi:10.1042/CS20110062.
demonstration of relative corticosteroid insensitivity in cells from people with 8. Papiris SA, Kollintza A, Karatza M, et al. CD8+ T lymphocytes in bronchoalveolar lavage in
idiopathic pulmonary fibrosis. J Inflamm Lond Engl. 2007;4:14. doi:10.1186/1476-9255-4-14.
severe asthma.108 The concept of enhancing MKP-1 expression and/or activity to 9. Savai R, Pullamsetti SS, Kolbe J, et al. Immune and inflammatory cell involvement in the pathology
control inflammation is currently under investigation,174 as is the potential use of p38 of idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med. 2012;186(9):897-908.
MAPK inhibitors to improve corticosteroid-mediated therapeutic control of chronic doi:10.1164/rccm.201202-0335OC.
respiratory disease, especially in severe asthma.108 Further studies are warranted. 10. Amin K, Lúdvíksdóttir D, Janson C, et al. Inflammation and structural changes in the airways
of patients with atopic and nonatopic asthma. BHR Group. Am J Respir Crit Care Med.
2000;162(6):2295-2301. doi:10.1164/ajrccm.162.6.9912001.
5.4. Urokinase & Annexin A2 11. Gosselink JV, Hayashi S, Elliott WM, et al. Differential Expression of Tissue Repair Genes in
the Pathogenesis of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med.
Urokinase and annexin A2 production by pulmonary mesenchymal cells is potentially
2010;181(12):1329-1335. doi:10.1164/rccm.200812-1902OC.
important in chronic respiratory disease (section 2.4). Both uPA and annexin A2 are 12. Noble PW, Barkauskas CE, Jiang D. Pulmonary fibrosis: patterns and perpetrators. J Clin Invest.
becoming increasingly recognised as important pathological mediators, particularly 2012;122(8):2756-2762. doi:10.1172/JCI60323. 6
in cancer, and their targeting by either pharmacological or antibody-based therapies 13. Hopkins N, McLoughlin P. The structural basis of pulmonary hypertension in chronic lung
disease: remodelling, rarefaction or angiogenesis? J Anat. 2002;201(4):335-348.
reduces tumour growth and/or metastasis in a number of pre-clinical cancer 14. Fixman ED, Stewart A, Martin JG. Basic mechanisms of development of airway structural changes
models.80–84 Furthermore, uPA inhibitors are well tolerated in humans and have in asthma. Eur Respir J. 2007;29(2):379-389. doi:10.1183/09031936.00053506.
provided promising results in recent phase I and II trials for cancer.81 Both uPA 15. Redington AE. Airway fibrosis in asthma: mechanisms, consequences, and potential for
and annexin A2 gene-deletion reduces pulmonary inflammation in various murine therapeutic intervention. Monaldi Arch Chest Dis Arch Monaldi Mal Torace. 2000;55(4):317-323.
16. Koziol-White CJ, Panettieri RA. Airway smooth muscle and immunomodulation in acute
models,77,175,176 and uPA antibodies reduce inflammation and oedema in a mouse exacerbations of airway disease. Immunol Rev. 2011;242(1):178-185. doi:10.1111/j.1600-
model of acute lung injury.177 However, further pre-clinical characterization of these 065X.2011.01022.x.
inhibitors as therapy for chronic respiratory disease is required. 17. Ward JE, Harris T, Bamford T, et al. Proliferation is not increased in airway myofibroblasts isolated
from asthmatics. Eur Respir J. 2008;32(2):362-371. doi:10.1183/09031936.00119307.
18. Flavell SJ, Hou TZ, Lax S, Filer AD, Salmon M, Buckley CD. Fibroblasts as novel therapeutic
6. Conclusion targets in chronic inflammation. Br J Pharmacol. 2008;153(Suppl 1):S241-S246. doi:10.1038/
In disease, pulmonary mesenchymal cells not only respond to inflammatory sj.bjp.0707487.
mediators, but also contribute to inflammation by producing chemokines, 19. Li M, Riddle SR, Frid MG, et al. Emergence of fibroblasts with a proinflammatory epigenetically
cytokines, CAMs and ECM matrix which recruit and activate inflammatory cells. The altered phenotype in severe hypoxic pulmonary hypertension. J Immunol Baltim Md 1950.
2011;187(5):2711-2722. doi:10.4049/jimmunol.1100479.
dysregulation of pulmonary mesenchymal cell immunomodulatory function is likely 20. Xia YC, Redhu NS, Moir LM, et al. Pro-inflammatory and immunomodulatory functions of airway
to contribute to the pathogenesis of lung disease. The specific targeting of aberrant smooth muscle: emerging concepts. Pulm Pharmacol Ther. 2013;26(1):64-74. doi:10.1016/j.
immunomodulatory functioning in pulmonary mesenchymal cells may be a strategy pupt.2012.05.006.
to treat lung disorders such as severe asthma, COPD, IPF and PH, for which there are 21. Evanko SP, Potter-Perigo S, Bollyky PL, Nepom GT, Wight TN. Hyaluronan and versican in the
control of human T-lymphocyte adhesion and migration. Matrix Biol J Int Soc Matrix Biol.
no current effective therapies. 2012;31(2):90-100. doi:10.1016/j.matbio.2011.10.004.
22. Li Y, Jiang D, Liang J, et al. Severe lung fibrosis requires an invasive fibroblast phenotype regulated
by hyaluronan and CD44. J Exp Med. 2011;208(7):1459-1471. doi:10.1084/jem.20102510.

110 111
23. Alkhouri H, Moir LM, Armour CL, Hughes JM. CXCL1 is a negative regulator of mast cell induced eotaxin responses in fibroblasts: regulation of the type-II IL-4 receptor chains IL-4Ralpha
chemotaxis to airway smooth muscle cell products in vitro. Clin Exp Allergy J Br Soc Allergy Clin and IL-13Ralpha1. Exp Cell Res. 2009;315(20):3486-3499. doi:10.1016/j.yexcr.2009.09.024.
Immunol. 2014;44(3):381-392. doi:10.1111/cea.12224. 43. Faffe DS, Whitehead T, Moore PE, et al. IL-13 and IL-4 promote TARC release in human airway
24. Jia Y, Morand EF, Song W, Cheng Q, Stewart A, Yang YH. Regulation of lung fibroblast activation smooth muscle cells: role of IL-4 receptor genotype. Am J Physiol Lung Cell Mol Physiol.
by annexin A1. J Cell Physiol. 2013;228(2):476-484. doi:10.1002/jcp.24156. 2003;285(4):L907-914. doi:10.1152/ajplung.00120.2003.
25. Elias JA, Wu Y, Zheng T, Panettieri R. Cytokine- and virus-stimulated airway smooth muscle cells 44. Kaur D, Saunders R, Berger P, et al. Airway smooth muscle and mast cell-derived CC chemokine
produce IL-11 and other IL-6-type cytokines. Am J Physiol. 1997;273(3 Pt 1):L648-655. ligand 19 mediate airway smooth muscle migration in asthma. Am J Respir Crit Care Med.
26. Rudd BD, Burstein E, Duckett CS, Li X, Lukacs NW. Differential role for TLR3 in respiratory syncytial 2006;174(11):1179-1188. doi:10.1164/rccm.200603-394OC.
virus-induced chemokine expression. J Virol. 2005;79(6):3350-3357. doi:10.1128/JVI.79.6.3350- 45. Jarai G, Sukkar M, Garrett S, et al. Effects of interleukin-1beta, interleukin-13 and transforming
3357.2005. growth factor-beta on gene expression in human airway smooth muscle using gene microarrays.
27. Saunders MA, Mitchell JA, Seldon PM, et al. Release of granulocyte-macrophage colony stimulating Eur J Pharmacol. 2004;497(3):255-265. doi:10.1016/j.ejphar.2004.06.055.
factor by human cultured airway smooth muscle cells: suppression by dexamethasone. Br J 46. Sukkar MB, Issa R, Xie S, Oltmanns U, Newton R, Chung KF. Fractalkine/CX3CL1 production by
Pharmacol. 1997;120(4):545-546. doi:10.1038/sj.bjp.0700998. human airway smooth muscle cells: induction by IFN-gamma and TNF-alpha and regulation
28. Fitzgerald SM, Chi DS, Hall HK, et al. GM-CSF induction in human lung fibroblasts by IL-1beta, by TGF-beta and corticosteroids. Am J Physiol Lung Cell Mol Physiol. 2004;287(6):L1230-1240.
TNF-alpha, and macrophage contact. J Interferon Cytokine Res Off J Int Soc Interferon Cytokine doi:10.1152/ajplung.00014.2004.
Res. 2003;23(2):57-65. doi:10.1089/107999003321455453. 47. Kassel O, Schmidlin F, Duvernelle C, Gasser B, Massard G, Frossard N. Human bronchial smooth
29. Knight DA, Lydell CP, Zhou D, Weir TD, Robert Schellenberg R, Bai TR. Leukemia inhibitory factor muscle cells in culture produce stem cell factor. Eur Respir J. 1999;13(5):951-954.
(LIF) and LIF receptor in human lung. Distribution and regulation of LIF release. Am J Respir Cell 48. Panettieri RA, Lazaar AL, Puré E, Albelda SM. Activation of cAMP-dependent pathways in human
Mol Biol. 1999;20(4):834-841. doi:10.1165/ajrcmb.20.4.3429. airway smooth muscle cells inhibits TNF-alpha-induced ICAM-1 and VCAM-1 expression and T
30. Burgess JK, Carlin S, Pack RA, et al. Detection and characterization of OX40 ligand expression lymphocyte adhesion. J Immunol Baltim Md 1950. 1995;154(5):2358-2365.
in human airway smooth muscle cells: a possible role in asthma? J Allergy Clin Immunol. 49. Spoelstra FM, Postma DS, Hovenga H, Noordhoek JA, Kauffman HF. Interferon-gamma and
2004;113(4):683-689. doi:10.1016/j.jaci.2003.12.311. interleukin-4 differentially regulate ICAM-1 and VCAM-1 expression on human lung fibroblasts.
31. Issa R, Xie S, Khorasani N, et al. Corticosteroid inhibition of growth-related oncogene protein- Eur Respir J. 1999;14(4):759-766.
alpha via mitogen-activated kinase phosphatase-1 in airway smooth muscle cells. J Immunol 50. Burgess JK, Blake AE, Boustany S, et al. CD40 and OX40 ligand are increased on stimulated
Baltim Md 1950. 2007;178(11):7366-7375. asthmatic airway smooth muscle. J Allergy Clin Immunol. 2005;115(2):302-308. doi:10.1016/j.
32. Brant KA, Fabisiak JP. Nickel and the Microbial Toxin, MALP-2, Stimulate Proangiogenic jaci.2004.11.004.
Mediators from Human Lung Fibroblasts via a HIF-1α and COX-2–Mediated Pathway. Toxicol Sci. 51. Lazaar AL, Albelda SM, Pilewski JM, Brennan B, Puré E, Panettieri RA. T lymphocytes adhere
2009;107(1):227-237. doi:10.1093/toxsci/kfn208. to airway smooth muscle cells via integrins and CD44 and induce smooth muscle cell DNA
33. Pang L, Knox AJ. Bradykinin stimulates IL-8 production in cultured human airway smooth muscle synthesis. J Exp Med. 1994;180(3):807-816.
cells: role of cyclooxygenase products. J Immunol Baltim Md 1950. 1998;161(5):2509-2515. 52. Pechkovsky DV, Prêle CM, Wong J, et al. STAT3-mediated signaling dysregulates lung fibroblast- 6
34. Brightling CE, Ammit AJ, Kaur D, et al. The CXCL10/CXCR3 axis mediates human lung mast cell myofibroblast activation and differentiation in UIP/IPF. Am J Pathol. 2012;180(4):1398-1412.
migration to asthmatic airway smooth muscle. Am J Respir Crit Care Med. 2005;171(10):1103- doi:10.1016/j.ajpath.2011.12.022.
1108. doi:10.1164/rccm.200409-1220OC. 53. Weylandt KH, Chiu C-Y, Gomolka B, Waechter SF, Wiedenmann B. Omega-3 fatty acids and their
35. Hardaker EL, Bacon AM, Carlson K, et al. Regulation of TNF-alpha- and IFN-gamma-induced lipid mediators: towards an understanding of resolvin and protectin formation. Prostaglandins
CXCL10 expression: participation of the airway smooth muscle in the pulmonary inflammatory Other Lipid Mediat. 2012;97(3-4):73-82. doi:10.1016/j.prostaglandins.2012.01.005.
response in chronic obstructive pulmonary disease. FASEB J Off Publ Fed Am Soc Exp Biol. 54. Aoki H, Hisada T, Ishizuka T, et al. Protective effect of resolvin E1 on the development of asthmatic
2004;18(1):191-193. doi:10.1096/fj.03-0170fje. airway inflammation. Biochem Biophys Res Commun. 2010;400(1):128-133. doi:10.1016/j.
36. Dong S, Zhang X, He Y, et al. Synergy of IL-27 and TNF-α in regulating CXCL10 expression in lung bbrc.2010.08.025.
fibroblasts. Am J Respir Cell Mol Biol. 2013;48(4):518-530. doi:10.1165/rcmb.2012-0340OC. 55. Levy BD, Kohli P, Gotlinger K, et al. Protectin D1 is generated in asthma and dampens airway
37. Wuyts WA, Vanaudenaerde BM, Dupont LJ, Demedts MG, Verleden GM. Modulation by cAMP inflammation and hyperresponsiveness. J Immunol Baltim Md 1950. 2007;178(1):496-502.
of IL-1beta-induced eotaxin and MCP-1 expression and release in human airway smooth muscle 56. Wang Y, Li D, Xu N, et al. Follistatin-like protein 1: a serum biochemical marker reflecting the
cells. Eur Respir J. 2003;22(2):220-226. severity of joint damage in patients with osteoarthritis. Arthritis Res Ther. 2011;13(6):R193.
38. Holgate ST, Bodey KS, Janezic A, Frew AJ, Kaplan AP, Teran LM. Release of RANTES, MIP-1 alpha, doi:10.1186/ar3522.
and MCP-1 into asthmatic airways following endobronchial allergen challenge. Am J Respir Crit 57. Rogerio AP, Haworth O, Croze R, et al. Resolvin D1 and aspirin-triggered resolvin D1 promote
Care Med. 1997;156(5):1377-1383. doi:10.1164/ajrccm.156.5.9610064. resolution of allergic airways responses. J Immunol Baltim Md 1950. 2012;189(4):1983-1991.
39. John M, Hirst SJ, Jose PJ, et al. Human airway smooth muscle cells express and release RANTES doi:10.4049/jimmunol.1101665.
in response to T helper 1 cytokines: regulation by T helper 2 cytokines and corticosteroids. J 58. Miyata J, Fukunaga K, Iwamoto R, et al. Dysregulated synthesis of protectin D1 in eosinophils
Immunol Baltim Md 1950. 1997;158(4):1841-1847. from patients with severe asthma. J Allergy Clin Immunol. 2013;131(2):353-360-2. doi:10.1016/j.
40. Choi ES, Jakubzick C, Carpenter KJ, et al. Enhanced monocyte chemoattractant protein-3/CC jaci.2012.07.048.
chemokine ligand-7 in usual interstitial pneumonia. Am J Respir Crit Care Med. 2004;170(5):508- 59. Levy BD. Resolvin D1 and Resolvin E1 Promote the Resolution of Allergic Airway Inflammation
515. doi:10.1164/rccm.200401-002OC. via Shared and Distinct Molecular Counter-Regulatory Pathways. Front Immunol. 2012;3:390.
41. Ghaffar O, Hamid Q, Renzi PM, et al. Constitutive and cytokine-stimulated expression of eotaxin doi:10.3389/fimmu.2012.00390.
by human airway smooth muscle cells. Am J Respir Crit Care Med. 1999;159(6):1933-1942. 60. Hsiao H-M, Sapinoro RE, Thatcher TH, et al. A novel anti-inflammatory and pro-resolving role for
doi:10.1164/ajrccm.159.6.9805039. resolvin D1 in acute cigarette smoke-induced lung inflammation. PloS One. 2013;8(3):e58258.
42. Fritz DK, Kerr C, Botelho F, Stampfli M, Richards CD. Oncostatin M (OSM) primes IL-13- and IL-4- doi:10.1371/journal.pone.0058258.

112 113
61. Wu D, Zheng S, Li W, et al. Novel biphasic role of resolvin D1 on expression of cyclooxygenase-2 80. Henneke I, Greschus S, Savai R, et al. Inhibition of urokinase activity reduces primary tumor
in lipopolysaccharide-stimulated lung fibroblasts is partly through PI3K/AKT and ERK2 pathways. growth and metastasis formation in a murine lung carcinoma model. Am J Respir Crit Care Med.
Mediators Inflamm. 2013;2013:964012. doi:10.1155/2013/964012. 2010;181(6):611-619. doi:10.1164/rccm.200903-0342OC.
62. Millien VO, Lu W, Shaw J, et al. Cleavage of fibrinogen by proteinases elicits allergic responses 81. Schmitt M, Harbeck N, Brünner N, et al. Cancer therapy trials employing level-of-evidence-1
through Toll-like receptor 4. Science. 2013;341(6147):792-796. doi:10.1126/science.1240342. disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn.
63. Stewart AG, Xia YC, Harris T, Royce S, Hamilton JA, Schuliga M. Plasminogen-stimulated airway 2011;11(6):617-634. doi:10.1586/erm.11.47.
smooth muscle cell proliferation is mediated by urokinase and annexin A2, involving plasmin- 82. Sharma M, Blackman MR, Sharma MC. Antibody-directed neutralization of annexin II (ANX II)
activated cell signalling. Br J Pharmacol. 2013;170(7):1421-1435. doi:10.1111/bph.12422. inhibits neoangiogenesis and human breast tumor growth in a xenograft model. Exp Mol Pathol.
64. Brant KA, Fabisiak JP. Nickel alterations of TLR2-dependent chemokine profiles in lung fibroblasts 2012;92(1):175-184. doi:10.1016/j.yexmp.2011.10.003.
are mediated by COX-2. Am J Respir Cell Mol Biol. 2008;38(5):591-599. doi:10.1165/rcmb.2007- 83. Sharma M, Ownbey RT, Sharma MC. Breast cancer cell surface annexin II induces cell migration
0314OC. and neoangiogenesis via tPA dependent plasmin generation. Exp Mol Pathol. 2010;88(2):278-
65. Sugiura H, Ichikawa T, Koarai A, et al. Activation of Toll-like receptor 3 augments myofibroblast 286. doi:10.1016/j.yexmp.2010.01.001.
differentiation. Am J Respir Cell Mol Biol. 2009;40(6):654-662. doi:10.1165/rcmb.2008-0371OC. 84. Zheng L, Foley K, Huang L, et al. Tyrosine 23 phosphorylation-dependent cell-surface localization
66. He Z, Zhu Y, Jiang H. Toll-like receptor 4 mediates lipopolysaccharide-induced collagen secretion of annexin A2 is required for invasion and metastases of pancreatic cancer. PloS One.
by phosphoinositide3-kinase-Akt pathway in fibroblasts during acute lung injury. J Recept Signal 2011;6(4):e19390. doi:10.1371/journal.pone.0019390.
Transduct Res. 2009;29(2):119-125. doi:10.1080/10799890902845690. 85. Chambers RC, Scotton CJ. Coagulation cascade proteinases in lung injury and fibrosis. Proc Am
67. Meneghin A, Choi ES, Evanoff HL, et al. TLR9 is expressed in idiopathic interstitial pneumonia Thorac Soc. 2012;9(3):96-101. doi:10.1513/pats.201201-006AW.
and its activation promotes in vitro myofibroblast differentiation. Histochem Cell Biol. 86. Lambrecht BN, Hammad H. Asthma and coagulation. N Engl J Med. 2013;369(20):1964-1966.
2008;130(5):979-992. doi:10.1007/s00418-008-0466-z. doi:10.1056/NEJMcibr1311045.
68. Sukkar MB, Xie S, Khorasani NM, et al. Toll-like receptor 2, 3, and 4 expression and function 87. Tchougounova E, Pejler G. Regulation of extravascular coagulation and fibrinolysis by heparin-
in human airway smooth muscle. J Allergy Clin Immunol. 2006;118(3):641-648. doi:10.1016/j. dependent mast cell chymase. FASEB J Off Publ Fed Am Soc Exp Biol. 2001;15(14):2763-2765.
jaci.2006.05.013. doi:10.1096/fj.01-0486fje.
69. Manetsch M, Seidel P, Heintz U, et al. TLR2 ligand engagement upregulates airway smooth muscle 88. Knight DA, Lim S, Scaffidi AK, et al. Protease-activated receptors in human airways: upregulation
TNFα-induced cytokine production. Am J Physiol Lung Cell Mol Physiol. 2012;302(9):L838-845. of PAR-2 in respiratory epithelium from patients with asthma. J Allergy Clin Immunol.
doi:10.1152/ajplung.00317.2011. 2001;108(5):797-803. doi:10.1067/mai.2001.119025.
70. Swisher JFA, Burton N, Bacot SM, Vogel SN, Feldman GM. Annexin A2 tetramer activates 89. Scotton CJ, Krupiczojc MA, Königshoff M, et al. Increased local expression of coagulation
human and murine macrophages through TLR4. Blood. 2010;115(3):549-558. doi:10.1182/ factor X contributes to the fibrotic response in human and murine lung injury. J Clin Invest.
blood-2009-06-226944. 2009;119(9):2550-2563. doi:10.1172/JCI33288.
71. O’Dwyer DN, Armstrong ME, Trujillo G, et al. The Toll-like receptor 3 L412F polymorphism 90. Ortiz-Stern A, Deng X, Smoktunowicz N, Mercer PF, Chambers RC. PAR-1-dependent and PAR-
and disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. independent pro-inflammatory signaling in human lung fibroblasts exposed to thrombin. J Cell 6
2013;188(12):1442-1450. doi:10.1164/rccm.201304-0760OC. Physiol. 2012;227(11):3575-3584. doi:10.1002/jcp.24061.
72. He Z, Gao Y, Deng Y, et al. Lipopolysaccharide induces lung fibroblast proliferation through 91. Tran T, Stewart AG. Protease-activated receptor (PAR)-independent growth and pro-
Toll-like receptor 4 signaling and the phosphoinositide3-kinase-Akt pathway. PloS One. inflammatory actions of thrombin on human cultured airway smooth muscle. Br J Pharmacol.
2012;7(4):e35926. doi:10.1371/journal.pone.0035926. 2003;138(5):865-875. doi:10.1038/sj.bjp.0705106.
73. Syrovets T, Lunov O, Simmet T. Plasmin as a proinflammatory cell activator. J Leukoc Biol. 92. Shinagawa K, Martin JA, Ploplis VA, Castellino FJ. Coagulation factor Xa modulates airway
2012;92(3):509-519. doi:10.1189/jlb.0212056. remodeling in a murine model of asthma. Am J Respir Crit Care Med. 2007;175(2):136-143.
74. Schuliga M, Westall G, Xia Y, Stewart AG. The plasminogen activation system: new targets in doi:10.1164/rccm.200608-1097OC.
lung inflammation and remodeling. Curr Opin Pharmacol. 2013;13(3):386-393. doi:10.1016/j. 93. Zhu W, Bi M, Liu Y, et al. Thrombin promotes airway remodeling via protease-activated receptor-1
coph.2013.05.014. and transforming growth factor-β1 in ovalbumin-allergic rats. Inhal Toxicol. 2013;25(10):577-
75. Pendurthi UR, Ngyuen M, Andrade-Gordon P, Petersen LC, Rao LVM. Plasmin induces Cyr61 586. doi:10.3109/08958378.2013.813995.
gene expression in fibroblasts via protease-activated receptor-1 and p44/42 mitogen-activated 94. Bousquet J, Jeffery PK, Busse WW, Johnson M, Vignola AM. Asthma. From bronchoconstriction
protein kinase-dependent signaling pathway. Arterioscler Thromb Vasc Biol. 2002;22(9):1421- to airways inflammation and remodeling. Am J Respir Crit Care Med. 2000;161(5):1720-1745.
1426. doi:10.1164/ajrccm.161.5.9903102.
76. Schuliga M, Harris T, Stewart AG. Plasminogen activation by airway smooth muscle is regulated 95. Pilette C, Francis JN, Till SJ, Durham SR. CCR4 ligands are up-regulated in the airways of atopic
by type I collagen. Am J Respir Cell Mol Biol. 2011;44(6):831-839. doi:10.1165/rcmb.2009- asthmatics after segmental allergen challenge. Eur Respir J. 2004;23(6):876-884.
0469OC. 96. Ordoñez CL, Shaughnessy TE, Matthay MA, Fahy JV. Increased neutrophil numbers and IL-8
77. Schuliga M, Langenbach S, Xia YC, et al. Plasminogen-stimulated inflammatory cytokine levels in airway secretions in acute severe asthma: Clinical and biologic significance. Am J Respir
production by airway smooth muscle cells is regulated by annexin A2. Am J Respir Cell Mol Biol. Crit Care Med. 2000;161(4 Pt 1):1185-1190. doi:10.1164/ajrccm.161.4.9812061.
2013;49(5):751-758. doi:10.1165/rcmb.2012-0404OC. 97. Taha RA, Minshall EM, Miotto D, et al. Eotaxin and monocyte chemotactic protein-4 mRNA
78. Bauman KA, Wettlaufer SH, Okunishi K, et al. The antifibrotic effects of plasminogen activation expression in small airways of asthmatic and nonasthmatic individuals. J Allergy Clin Immunol.
occur via prostaglandin E2 synthesis in humans and mice. J Clin Invest. 2010;120(6):1950-1960. 1999;103(3 Pt 1):476-483.
doi:10.1172/JCI38369. 98. John AE, Zhu YM, Brightling CE, Pang L, Knox AJ. Human airway smooth muscle cells from
79. Li Q, Laumonnier Y, Syrovets T, Simmet T. Plasmin triggers cytokine induction in human monocyte- asthmatic individuals have CXCL8 hypersecretion due to increased NF-kappa B p65, C/EBP
derived macrophages. Arterioscler Thromb Vasc Biol. 2007;27(6):1383-1389. doi:10.1161/ beta, and RNA polymerase II binding to the CXCL8 promoter. J Immunol Baltim Md 1950.
ATVBAHA.107.142901. 2009;183(7):4682-4692. doi:10.4049/jimmunol.0803832.

114 115
99. Chan V, Burgess JK, Ratoff JC, et al. Extracellular matrix regulates enhanced eotaxin expression 119. Zhang J, Wu L, Qu J, Bai C, Merrilees MJ, Black PN. Pro-inflammatory phenotype of COPD
in asthmatic airway smooth muscle cells. Am J Respir Crit Care Med. 2006;174(4):379-385. fibroblasts not compatible with repair in COPD lung. J Cell Mol Med. 2012;16(7):1522-1532.
doi:10.1164/rccm.200509-1420OC. doi:10.1111/j.1582-4934.2011.01492.x.
100. Faiz A, Oliver B, Black JL, Burgess JK. Ccl20 Production By Airway Smooth Muscle Cells Contributes 120. Larsson-Callerfelt A-K, Hallgren O, Andersson-Sjöland A, et al. Defective alterations in the collagen
To Mucus Hyperscretion And Increase Muscle Mass In Asthmatic Airways. Respirology. 2013;18. network to prostacyclin in COPD lung fibroblasts. Respir Res. 2013;14:21. doi:10.1186/1465-
https://insights.ovid.com/respirology/respy/2013/04/002/ccl20-production-airway-smooth- 9921-14-21.
muscle-cells/175/00075270. Accessed March 31, 2017. 121. Stitham J, Midgett C, Martin KA, Hwa J. Prostacyclin: an inflammatory paradox. Front Pharmacol.
101. Araujo BB, Dolhnikoff M, Silva LFF, et al. Extracellular matrix components and regulators in the airway 2011;2:24. doi:10.3389/fphar.2011.00024.
smooth muscle in asthma. Eur Respir J. 2008;32(1):61-69. doi:10.1183/09031936.00147807. 122. Nagai S, Handa T, Ito Y, Takeuchi M, Izumi T. Bronchoalveolar Lavage in Idiopathic Interstitial Lung
102. Johnson PRA, Burgess JK, Underwood PA, et al. Extracellular matrix proteins modulate asthmatic Diseases. Semin Respir Crit Care Med. 2007;28(05):496-503. doi:10.1055/s-2007-991522.
airway smooth muscle cell proliferation via an autocrine mechanism. J Allergy Clin Immunol. 123. Nathan N, Thouvenin G, Fauroux B, Corvol H, Clement A. Interstitial lung disease: physiopathology
2004;113(4):690-696. doi:10.1016/j.jaci.2003.12.312. in the context of lung growth. Paediatr Respir Rev. 2011;12(4):216-222. doi:10.1016/j.
103. Lau JY, Oliver BG, Baraket M, et al. Fibulin-1 is increased in asthma--a novel mediator of airway prrv.2011.04.003.
remodeling? PloS One. 2010;5(10):e13360. doi:10.1371/journal.pone.0013360. 124. Coffey E, Newman DR, Sannes PL. Expression of fibroblast growth factor 9 in normal human lung
104. Mahn K, Hirst SJ, Ying S, et al. Diminished sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) and idiopathic pulmonary fibrosis. J Histochem Cytochem Off J Histochem Soc. 2013;61(9):671-
expression contributes to airway remodelling in bronchial asthma. Proc Natl Acad Sci U S A. 679. doi:10.1369/0022155413497366.
2009;106(26):10775-10780. doi:10.1073/pnas.0902295106. 125. Lindahl GE, Stock CJ, Shi-Wen X, et al. Microarray profiling reveals suppressed interferon
105. Trian T, Benard G, Begueret H, et al. Bronchial smooth muscle remodeling involves calcium- stimulated gene program in fibroblasts from scleroderma-associated interstitial lung disease.
dependent enhanced mitochondrial biogenesis in asthma. J Exp Med. 2007;204(13):3173-3181. Respir Res. 2013;14:80. doi:10.1186/1465-9921-14-80.
doi:10.1084/jem.20070956. 126. Nagahama KY, Togo S, Holz O, et al. Oncostatin M modulates fibroblast function via signal
106. Alrashdan YA, Alkhouri H, Chen E, et al. Asthmatic airway smooth muscle CXCL10 production: transducers and activators of transcription proteins-3. Am J Respir Cell Mol Biol. 2013;49(4):582-
mitogen-activated protein kinase JNK involvement. Am J Physiol Lung Cell Mol Physiol. 591. doi:10.1165/rcmb.2012-0460OC.
2012;302(10):L1118-1127. doi:10.1152/ajplung.00232.2011. 127. Suda T, Chida K, Todate A, et al. Oncostatin M production by human dendritic cells in response
107. Tan X, Alrashdan YA, Alkhouri H, Oliver BGG, Armour CL, Hughes JM. Airway smooth muscle to bacterial products. Cytokine. 2002;17(6):335-340. doi:10.1006/cyto.2002.1023.
CXCR3 ligand production: regulation by JAK-STAT1 and intracellular Ca2+. Am J Physiol - Lung Cell 128. Botelho FM, Rangel-Moreno J, Fritz D, Randall TD, Xing Z, Richards CD. Pulmonary expression
Mol Physiol. 2013;304(11):L790-L802. doi:10.1152/ajplung.00356.2012. of oncostatin M (OSM) promotes inducible BALT formation independently of IL-6, despite a role
108. Chang P-J, Bhavsar PK, Michaeloudes C, Khorasani N, Chung KF. Corticosteroid insensitivity of for IL-6 in OSM-driven pulmonary inflammation. J Immunol Baltim Md 1950. 2013;191(3):1453-
chemokine expression in airway smooth muscle of patients with severe asthma. J Allergy Clin 1464. doi:10.4049/jimmunol.1203318.
Immunol. 2012;130(4):877-885.e5. doi:10.1016/j.jaci.2012.07.017. 129. Fritz DK, Kerr C, Fattouh R, et al. A mouse model of airway disease: oncostatin M-induced
109. Roth M, Johnson PRA, Borger P, et al. Dysfunctional interaction of C/EBPalpha and pulmonary eosinophilia, goblet cell hyperplasia, and airway hyperresponsiveness are STAT6 6
the glucocorticoid receptor in asthmatic bronchial smooth-muscle cells. N Engl J Med. dependent, and interstitial pulmonary fibrosis is STAT6 independent. J Immunol Baltim Md
2004;351(6):560-574. doi:10.1056/NEJMoa021660. 1950. 2011;186(2):1107-1118. doi:10.4049/jimmunol.0903476.
110. Hew M, Bhavsar P, Torrego A, et al. Relative corticosteroid insensitivity of peripheral blood 130. Mozaffarian A, Brewer AW, Trueblood ES, et al. Mechanisms of oncostatin M-induced pulmonary
mononuclear cells in severe asthma. Am J Respir Crit Care Med. 2006;174(2):134-141. inflammation and fibrosis. J Immunol Baltim Md 1950. 2008;181(10):7243-7253.
doi:10.1164/rccm.200512-1930OC. 131. Smyth DC, Kerr C, Li Y, Tang D, Richards CD. Oncostatin M induction of eotaxin-1 expression
111. Ingram JL, Huggins MJ, Church TD, et al. Airway fibroblasts in asthma manifest an invasive requires the convergence of PI3’K and ERK1/2 MAPK signal transduction pathways. Cell Signal.
phenotype. Am J Respir Crit Care Med. 2011;183(12):1625-1632. doi:10.1164/rccm.201009- 2008;20(6):1142-1150. doi:10.1016/j.cellsig.2008.02.001.
1452OC. 132. Pak O, Aldashev A, Welsh D, Peacock A. The effects of hypoxia on the cells of the pulmonary
112. Zheng T, Liu W, Oh S-Y, et al. IL-13 receptor alpha2 selectively inhibits IL-13-induced responses in vasculature. Eur Respir J. 2007;30(2):364-372. doi:10.1183/09031936.00128706.
the murine lung. J Immunol Baltim Md 1950. 2008;180(1):522-529. 133. Crosswhite P, Sun Z. Nitric oxide, oxidative stress and inflammation in pulmonary arterial
113. Han MK, Criner GJ. Update in chronic obstructive pulmonary disease 2012. Am J Respir Crit Care hypertension. J Hypertens. 2010;28(2):201-212. doi:10.1097/HJH.0b013e328332bcdb.
Med. 2013;188(1):29-34. doi:10.1164/rccm.201302-0319UP. 134. Stenmark KR, Frid MG, Yeager M, et al. Targeting the adventitial microenvironment in pulmonary
114. Qiu W, Baccarelli A, Carey VJ, et al. Variable DNA methylation is associated with chronic hypertension: A potential approach to therapy that considers epigenetic change. Pulm Circ.
obstructive pulmonary disease and lung function. Am J Respir Crit Care Med. 2012;185(4):373- 2012;2(1):3-14. doi:10.4103/2045-8932.94817.
381. doi:10.1164/rccm.201108-1382OC. 135. Wang D, Zhang H, Li M, et al. MicroRNA-124 controls the proliferative, migratory, and
115. Sze MA, Dimitriu PA, Hayashi S, et al. The lung tissue microbiome in chronic obstructive inflammatory phenotype of pulmonary vascular fibroblasts. Circ Res. 2014;114(1):67-78.
pulmonary disease. Am J Respir Crit Care Med. 2012;185(10):1073-1080. doi:10.1164/ doi:10.1161/CIRCRESAHA.114.301633.
rccm.201111-2075OC. 136. Schabbauer G, Matt U, Günzl P, et al. Myeloid PTEN promotes inflammation but impairs
116. Jeffery PK. Remodeling and inflammation of bronchi in asthma and chronic obstructive bactericidal activities during murine pneumococcal pneumonia. J Immunol Baltim Md 1950.
pulmonary disease. Proc Am Thorac Soc. 2004;1(3):176-183. doi:10.1513/pats.200402-009MS. 2010;185(1):468-476. doi:10.4049/jimmunol.0902221.
117. Hallgren O, Rolandsson S, Andersson-Sjöland A, et al. Enhanced ROCK1 dependent contractility 137. Horita H, Furgeson SB, Ostriker A, et al. Selective inactivation of PTEN in smooth muscle
in fibroblast from chronic obstructive pulmonary disease patients. J Transl Med. 2012;10:171. cells synergizes with hypoxia to induce severe pulmonary hypertension. J Am Heart Assoc.
doi:10.1186/1479-5876-10-171. 2013;2(3):e000188. doi:10.1161/JAHA.113.000188.
118. Hallgren O, Nihlberg K, Dahlbäck M, et al. Altered fibroblast proteoglycan production in COPD. 138. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, Pavord ID. Mast-cell infiltration
Respir Res. 2010;11:55. doi:10.1186/1465-9921-11-55. of airway smooth muscle in asthma. N Engl J Med. 2002;346(22):1699-1705. doi:10.1056/

116 117
NEJMoa012705. pulmonary disease fibroblasts. Am J Respir Crit Care Med. 2010;182(8):1020-1029. doi:10.1164/
139. Amin K, Janson C, Harvima I, Venge P, Nilsson G. CC chemokine receptors CCR1 and CCR4 are rccm.201001-0055OC.
expressed on airway mast cells in allergic asthma. J Allergy Clin Immunol. 2005;116(6):1383- 159. Togo S, Holz O, Liu X, et al. Lung Fibroblast Repair Functions in Patients with Chronic Obstructive
1386. doi:10.1016/j.jaci.2005.08.053. Pulmonary Disease Are Altered by Multiple Mechanisms. Am J Respir Crit Care Med.
140. Begueret H, Berger P, Vernejoux J, Dubuisson L, Marthan R, Tunon‐de‐Lara JM. Inflammation 2008;178(3):248-260. doi:10.1164/rccm.200706-929OC.
of bronchial smooth muscle in allergic asthma. Thorax. 2007;62(1):8-15. doi:10.1136/ 160. Okunishi K, Sisson TH, Huang SK, Hogaboam CM, Simon RH, Peters-Golden M. Plasmin
thx.2006.062141. overcomes resistance to prostaglandin E2 in fibrotic lung fibroblasts by reorganizing protein
141. Carroll NG, Mutavdzic S, James AL. Increased mast cells and neutrophils in submucosal mucous kinase A signaling. J Biol Chem. 2011;286(37):32231-32243. doi:10.1074/jbc.M111.235606.
glands and mucus plugging in patients with asthma. Thorax. 2002;57(8):677-682. 161. Xie S, Sukkar MB, Issa R, Oltmanns U, Nicholson AG, Chung KF. Regulation of TGF-beta 1-induced
142. Sutcliffe A, Kaur D, Page S, et al. Mast cell migration to Th2 stimulated airway smooth muscle connective tissue growth factor expression in airway smooth muscle cells. Am J Physiol Lung Cell
from asthmatics. Thorax. 2006;61(8):657-662. doi:10.1136/thx.2005.056770. Mol Physiol. 2005;288(1):L68-76. doi:10.1152/ajplung.00156.2004.
143. Juremalm M, Hjertson M, Olsson N, Harvima I, Nilsson K, Nilsson G. The chemokine receptor 162. Wynn TA. Integrating mechanisms of pulmonary fibrosis. J Exp Med. 2011;208(7):1339-1350.
CXCR4 is expressed within the mast cell lineage and its ligand stromal cell-derived factor-1alpha doi:10.1084/jem.20110551.
acts as a mast cell chemotaxin. Eur J Immunol. 2000;30(12):3614-3622. doi:10.1002/1521- 163. Yndestad A, Larsen K-O, Oie E, et al. Elevated levels of activin A in clinical and experimental
4141(200012)30:12&#60;3614::AID-IMMU3614&#62;3.0.CO;2-B. pulmonary hypertension. J Appl Physiol Bethesda Md 1985. 2009;106(4):1356-1364.
144. Juremalm M, Olsson N, Nilsson G. Selective CCL5/RANTES-induced mast cell migration through doi:10.1152/japplphysiol.90719.2008.
interactions with chemokine receptors CCR1 and CCR4. Biochem Biophys Res Commun. 164. Aoki F, Kurabayashi M, Hasegawa Y, Kojima I. Attenuation of bleomycin-induced pulmonary
2002;297(3):480-485. fibrosis by follistatin. Am J Respir Crit Care Med. 2005;172(6):713-720. doi:10.1164/rccm.200412-
145. Berger P, Girodet P-O, Begueret H, et al. Tryptase-stimulated human airway smooth muscle 1620OC.
cells induce cytokine synthesis and mast cell chemotaxis. FASEB J Off Publ Fed Am Soc Exp Biol. 165. Ponticos M, Holmes AM, Shi-wen X, et al. Pivotal role of connective tissue growth factor in
2003;17(14):2139-2141. doi:10.1096/fj.03-0041fje. lung fibrosis: MAPK-dependent transcriptional activation of type I collagen. Arthritis Rheum.
146. Alkhouri H, Hollins F, Moir LM, Brightling CE, Armour CL, Hughes JM. Human lung mast cells 2009;60(7):2142-2155. doi:10.1002/art.24620.
modulate the functions of airway smooth muscle cells in asthma. Allergy. 2011;66(9):1231- 166. Lu J, Auduong L, White ES, Yue X. Up-regulation of heparan sulfate 6-O-sulfation in idiopathic
1241. doi:10.1111/j.1398-9995.2011.02616.x. pulmonary fibrosis. Am J Respir Cell Mol Biol. 2014;50(1):106-114. doi:10.1165/rcmb.2013-
147. Hollins F, Kaur D, Yang W, et al. Human airway smooth muscle promotes human lung mast cell 0204OC.
survival, proliferation, and constitutive activation: cooperative roles for CADM1, stem cell factor, 167. Gallelli L, Falcone D, Pelaia G, et al. Interleukin-6 receptor superantagonist Sant7 inhibits
and IL-6. J Immunol Baltim Md 1950. 2008;181(4):2772-2780. TGF-beta-induced proliferation of human lung fibroblasts. Cell Prolif. 2008;41(3):393-407.
148. Bradding P. Mast cell regulation of airway smooth muscle function in asthma. Eur Respir J. doi:10.1111/j.1365-2184.2008.00538.x.
2007;29(5):827-830. doi:10.1183/09031936.00017707. 168. Baarsma HA, Engelbertink LHJM, van Hees LJ, et al. Glycogen synthase kinase-3 (GSK-3) regulates
149. John AE, Lukacs NW. Chemokines and Asthma. Sarcoidosis Vasc Diffuse Lung Dis. 2003;20(3):180- TGF-β₁-induced differentiation of pulmonary fibroblasts. Br J Pharmacol. 2013;169(3):590-603. 6
189. doi:10.1111/bph.12098.
150. Page S, Ammit AJ, Black JL, Armour CL. Human mast cell and airway smooth muscle cell 169. Baarsma HA, Meurs H, Halayko AJ, et al. Glycogen synthase kinase-3 regulates cigarette smoke
interactions: implications for asthma. Am J Physiol Lung Cell Mol Physiol. 2001;281(6):L1313- extract- and IL-1β-induced cytokine secretion by airway smooth muscle. Am J Physiol Lung Cell
1323. Mol Physiol. 2011;300(6):L910-919. doi:10.1152/ajplung.00232.2010.
151. Kitamura H, Cambier S, Somanath S, et al. Mouse and human lung fibroblasts regulate dendritic 170. Manetsch M, Rahman MM, Patel BS, et al. Long-acting β2-agonists increase fluticasone
cell trafficking, airway inflammation, and fibrosis through integrin αvβ8-mediated activation of propionate-induced mitogen-activated protein kinase phosphatase 1 (MKP-1) in airway smooth
TGF-β. J Clin Invest. 2011;121(7):2863-2875. doi:10.1172/JCI45589. muscle cells. PloS One. 2013;8(3):e59635. doi:10.1371/journal.pone.0059635.
152. Pechkovsky DV, Prasse A, Kollert F, et al. Alternatively activated alveolar macrophages in 171. Quante T, Ng YC, Ramsay EE, et al. Corticosteroids reduce IL-6 in ASM cells via up-regulation of
pulmonary fibrosis-mediator production and intracellular signal transduction. Clin Immunol MKP-1. Am J Respir Cell Mol Biol. 2008;39(2):208-217. doi:10.1165/rcmb.2007-0014OC.
Orlando Fla. 2010;137(1):89-101. doi:10.1016/j.clim.2010.06.017. 172. Che W, Manetsch M, Quante T, et al. Sphingosine 1-phosphate induces MKP-1 expression via
153. Kunz LIZ, Lapperre TS, Snoeck-Stroband JB, et al. Smoking status and anti-inflammatory p38 MAPK- and CREB-mediated pathways in airway smooth muscle cells. Biochim Biophys Acta.
macrophages in bronchoalveolar lavage and induced sputum in COPD. Respir Res. 2011;12:34. 2012;1823(10):1658-1665. doi:10.1016/j.bbamcr.2012.06.011.
doi:10.1186/1465-9921-12-34. 173. Manetsch M, Che W, Seidel P, Chen Y, Ammit AJ. MKP-1: a negative feedback effector that represses
154. Prasse A, Pechkovsky DV, Toews GB, et al. A vicious circle of alveolar macrophages and fibroblasts MAPK-mediated pro-inflammatory signaling pathways and cytokine secretion in human airway
perpetuates pulmonary fibrosis via CCL18. Am J Respir Crit Care Med. 2006;173(7):781-792. smooth muscle cells. Cell Signal. 2012;24(4):907-913. doi:10.1016/j.cellsig.2011.12.013.
doi:10.1164/rccm.200509-1518OC. 174. Doddareddy MR, Rawling T, Ammit AJ. Targeting mitogen-activated protein kinase phosphatase-1
155. Barnes PJ. New anti-inflammatory targets for chronic obstructive pulmonary disease. Nat Rev (MKP-1): structure-based design of MKP-1 inhibitors and upregulators. Curr Med Chem.
Drug Discov. 2013;12(7):543-559. doi:10.1038/nrd4025. 2012;19(2):163-173.
156. Tannheimer SL, Wright CD, Salmon M. Combination of roflumilast with a beta-2 adrenergic 175. Abraham E, Gyetko MR, Kuhn K, et al. Urokinase-type plasminogen activator potentiates
receptor agonist inhibits proinflammatory and profibrotic mediator release from human lung lipopolysaccharide-induced neutrophil activation. J Immunol Baltim Md 1950. 2003;170(11):5644-
fibroblasts. Respir Res. 2012;13:28. doi:10.1186/1465-9921-13-28. 5651.
157. Michalski J, Kanaji N, Liu X, et al. Attenuation of inhibitory prostaglandin E2 signaling in human 176. Swaisgood CM, French EL, Noga C, Simon RH, Ploplis VA. The development of bleomycin-induced
lung fibroblasts is mediated by phosphodiesterase 4. Am J Respir Cell Mol Biol. 2012;47(6):729- pulmonary fibrosis in mice deficient for components of the fibrinolytic system. Am J Pathol.
737. doi:10.1165/rcmb.2012-0057OC. 2000;157(1):177-187. doi:10.1016/S0002-9440(10)64529-4.
158. Sato T, Liu X, Nelson A, et al. Reduced miR-146a increases prostaglandin E₂in chronic obstructive 177. Wang X-Q, Bdeir K, Yarovoi S, Cines DB, Fang W, Abraham E. Involvement of the urokinase

118 119
CHAPTER 7 1

Variant club cell differentiation is


driven by bone morphogenetic
protein 4 in adult human airway
epithelium:
Implications for goblet cell metaplasia
and basal cell hyperplasia in COPD

Navessa P. Tania, Harm Maarsingh, Frank Ensink, John-Poul Ng-Blichfeldt,


Pieter S. Hiemstra, Maurice J.B. van den Hoff, Martina Schmidt,
Reinoud Gosens

121
Variant club cell differentiation is driven by bone morphogenetic protein 4 in adult Abstract
human airway epithelium: Implications for goblet cell metaplasia and basal cell
hyperplasia in COPD Activation of BMP signaling in adult airway epithelium is required for appropriate
regeneration after epithelial injury and plays key roles in proximal-to-distal
patterning of airway epithelial differentiation with highest expression in the distal
airways. COPD is associated with epithelial remodeling characterized by loss of
functional club cells, increased goblet cell metaplasia and basal cell hyperplasia.
Navessa P. Tania1,2, Harm Maarsingh3, Frank Ensink1, John-Poul Ng-Blichfeldt1,2, Pieter
Here, we studied the effects of BMP4 on adult airway epithelial differentiation,
S. Hiemstra4, Maurice J.B. van den Hoff5, Martina Schmidt1,2, Reinoud Gosens1,2
hypothesizing that BMP4 would regulate club cell differentiation, whilst suppressing
goblet cell differentiation. An in vitro air-liquid interface (ALI) culture system was
used to grow adult primary tracheobronchial epithelial cells (PTECs). After 14 days
of air-exposure in the presence or absence of BMP4, distinct morphological changes
were observed in BMP4-treated PTECs. BMP4 reduced gene expression for markers
University of Groningen, Department of Molecular Pharmacology, Groningen, The
1

Netherlands. of ciliated (Tektin-1), goblet (MUC5AC), and basal (TP63) cells. Interestingly, BMP4
increased marker gene expression for distal variant club cells (UPK3A, SCGB3A,
2
University of Groningen, University Medical Center Groningen, Groningen Research SFTPA), but not for common club cells (CYP2F1, SCGB1A1). Moreover, BMP4 induced
Institute for Asthma and COPD, Groningen, The Netherlands. gene expression of lung progenitor markers (NKX2.1, ITGA6). Mechanistically, BMP4
increased target genes downstream of Notch signaling (HEY1, HEY3) and TGF-β1
3
Palm Beach Atlantic University, Lloyd L. Gregory School of Pharmacy, Department of signaling (ID-1, SERPINE1). In conclusion, BMP4 promotes distal variant club cell
Pharmaceutical Sciences, West Palm Beach, Florida, USA. differentiation and suppresses proximal differentiation of goblet and ciliated cells.
Our findings provide a plausible connection between the loss of (variant) club cells
4
Dept. of Pulmonology, Leiden University Medical Centre, Leiden, The Netherlands.
and increased goblet cell metaplasia and basal cell hyperplasia on one hand and
5
Academic Medical Center, Department of Anatomy, Embryology and Physiology,
Amsterdam, The Netherlands. the observed reduction of BMP signaling in patients with COPD on the other hand.
Reactivation of BMP signaling in distal airway epithelium may be a strategy worth
pursuing to trigger differentiation and maintenance of distal variant club progenitor
cells capable of airway epithelial repair.

122 123
Introduction club cells and increased goblet cell metaplasia and basal cell hyperplasia on one
hand and the observed reduction of BMP signaling in patients with COPD on the
In fetal and neonatal lung development, BMP signaling is crucial in governing other. Restoring normal BMP signaling in distal airway epithelium may be a strategy
proximal-to-distal epithelial cell fate.1–3 Loss of BMP signaling in the developing worth pursuing to trigger differentiation and maintenance of distal variant club
mouse lung leads to severe loss of distal epithelial cell types, whilst promoting the progenitor cells capable of airway epithelial repair.
presence of proximal epithelial cell types - even in the most distal airway regions.3 In
adult lung, it has been proposed that BMP signaling is involved in tissue repair and Materials and methods
that inappropriate BMP activation might contribute to adult respiratory diseases,
including asthma and COPD.2–4 Activation of BMP signaling in adult airway epithelium Air-liquid interface cultured primary tracheobronchial epithelial cells (ALI-PTEC)-
after epithelial injury regenerates normal epithelial architecture in a similar pattern human PTECs were isolated from anonymous healthy lung transplant donors. Lung
as early lung development.5,6 transplant donors were selected according to Eurotransplant guidelines, including
The healthy epithelial lining of adult airways consists of a mixed population of absence of tumor and primary chronic lung diseases, such as asthma and COPD,
mucus-producing (MUC5AC+) goblet cells, (TEKT1+) ciliated cells, (TP63+) basal cells, and with <20 pack years of smoking history. PTECs were expanded in submerged
(SCGB1A1+/CYP2F+) club cells, and (SCGB3A2+/UPK3A+/SFTPA+) variant club cells.7,8 culture using keratinocyte serum free medium (17005-075, Invitrogen, CA, USA)
Goblet cells are normally expressed in the proximal airway epithelium, whereas supplemented with 25 µg/ml bovine pituitary extract and 0.2 ng/ml epidermal growth
(variant) club cells are predominantly expressed in the distal airway epithelium. In factor (37000-015, Invitrogen, CA, USA). Generation of mucociliary differentiated
COPD, airway epithelial cell differentiation programs are inappropriately activated PTEC cultures was conducted using air-liquid interface (ALI) system as previously
with loss of functional club cells, increased goblet cell metaplasia, and basal cell described.22,23 Prior to cell seeding in the ALI culture system, transwell semipermeable
hyperplasia as key pathological features of airway epithelium in the disease.9–12 Low inserts were coated for 2 h at 37 °C with a mixture of 30 µg/ml PureCol (Advanced
levels of secretoglobin family 1A member 1 (SCGB1A1, also known as a club (formerly BioMatrix, San Diego, CA, USA), 10 µg/ml human fibronectin, and 10 µg/ml BSA
clara) cell secretory protein CC10 or CC16) in serum and bronchoalveolar lavage have (Sigma Aldrich, St. Louis, MO, USA) dissolved in PBS. Passage 1-2 of PTECs were
been associated with the prevalence, severity and acceleration of a decline in forced seeded on coated Transwell 12 mm in diameter inserts at a density of 75,000 cells
expiratory volume in one second (FEV1) in COPD in several studies.10,13–17 Goblet cell (Corning, New York, USA) and cultured in submerged condition using B/D medium
metaplasia is characterized by increased numbers of goblet cells in the proximal composed of 1:1 DMEM (41966-029, Invitrogen, CA, USA) and BEBM supplemented
airways of COPD patients and by the appearance of goblet cells in more distal with BEGM BulletKit singlequots excluding gentamicin (CC-3170, Lonza, Verviers,
airway regions that normally do not express these cells.9 Importantly, our previous Belgium) plus freshly added retinoic acid (50 nM) (R-2625, Sigma Aldrich; St. Louis,
studies demonstrate that functional BMP4 signaling (Smad1/5/8 phosphorylation) MO, USA). For time course studies, fully confluent PTECs (~4 days) were air-exposed
is reduced in COPD (Chapter 3), which could explain the change in proximal-to-distal in the absence or presence of 100 ng/ml recombinant human BMP4 (314-BP, R&D 7
differentiation pattern. system, Oxford, UK) in basal medium. RNA was extracted for real time quantitative
The air-liquid interface (ALI) culture system provides a valuable tool to study PCR (RT-qPCR) and cells were fixed and sectioned for immunocytochemistry at day
airway epithelial differentiation in vitro mimicking a polarized pseudostratified 0, 4, 9, and 14. For concentration response studies, cells were air-exposed in the
epithelium.18,19 In ALI culture, the expression of the BMP4 gene is markedly increased absence or presence of 1, 10, 100 ng/ml recombinant human BMP4 (R&D system,
during mucociliary differentiation of adult airway epithelial cells.20 A recent study Oxford, UK) in basal medium for 14 days. For all experiments, the apical surface
demonstrates that BMP signaling is a negative regulator of basal progenitor cell was washed twice with PBS and freshly prepared BMP4-containing B/D medium was
proliferation in mouse tracheal epithelium6 and BMP4 drives specification of adult added every 48 h. At day 14, RNA was extracted for RT-qPCR and cells were fixed for
bronchoalveolar stem cells in mice,21 yet little is known about the role of BMP4 in immunocytochemistry (Figure 1A).
human airway epithelial cell differentiation. In addition, the underlying mechanisms
by which BMP exerts its effect in airway epithelial cell fate is still unclear. RNA isolation and RT-qPCR
In this study, we present evidence that BMP4 alters primary tracheobronchial Total RNA was extracted from ALI cultured cells using the NucleoSpin® RNA isolation
epithelial cells (PTECs) morphology and induces variant club cell marker gene kit according to manufacturer’s instruction (Macherey Nagel; Düren, Germany).
expression that is associated with high expression of distal epithelial and progenitor The total RNA concentration was determined using the NanoDrop® ND1000
markers. In parallel, BMP4 suppresses ciliated, goblet, and basal cell marker gene spectrophotometer (Thermo Scientific, Wilmington, MA). Equal amounts of total
expression. Our findings provide a plausible connection between the loss of (variant) RNA were reverse transcribed using the Reverse Transcription System (Promega;

124 125
Madison, WI, USA) to generate cDNA. Diluted cDNA was mixed with FastStart Immunocytochemistry
Universal SYBR Green Master Mix (Roche Applied Science; Penzberg, Germany) Cells were fixed on transwell inserts with 10% formalin and then paraffin-embedded
and gene-of-interest primer sets (Biolegio; Nijmegen, the Netherlands). Primer according to the ‘Costar® Transwell® Inserts for Histology preparation’ manufacturer’s
sequences are listed in Supplementary Table 1. Real-time quantitative PCR was procedure(Corning, New York, USA). Paraffin blocks were transversely cross-
performed using the Illumina Eco Personal qPCR System (Westburg; Leusden, The sectioned in 5 µm thick sections, which were fixed to glass slides for Hematoxylin
Netherlands). The qPCR reaction was started by denaturation at 95 °C for 15 minutes and eosinstaining (Sigma Aldrich; St. Louis, MO, USA). Sections were analysed using
followed by 45 cycles of denaturation at 94 °C for 30 s, annealing at 59 °C for 30 s a light microscope (Olympus BX41; Zoeterwoude, the Netherlands) using Cell^D
and elongation at 72 °C for 30 s. Final elongation was for 5 minutes at 72 °C. Real imaging software. Antibodies used were: SCGB3A (orb186085, Biorbyt, Cambridge,
time PCR data was analyzed using LinRegPCR software version 2013.1.24 Data were UK), UPK3A (HPA018415, Atlas antibody; Stockholm, Sweden), SFTPA (AB3420-I
expressed in arbitrary units as ratio of the starting concentration (N0) of each gene Millipore, Amsterdam-Zuidoost, the Netherlands).
of interest corrected to the geometric mean of the N0 value of 3 reference genes
(B2M, HPRT1, SDHA). Statistical analysis
Data are presented as mean ± standard error of the mean (SEM) of cultures
derived from 6 different donors. To determine the normality of data distribution, a
Supplementary Table 1: Primers used for RT-qPCR analysis Shapiro-Wilk normality test was performed prior to further statistical analysis. The
statistical significance of differences of normally distributed data was performed
Gene Forward primer (5’ 3’) Reverse primers (5’ 3’)
using an independent samples 2-tailed t-test for comparisons of 2 groups or a two-
TEKT1 GGCCAAGGTCATGGAAGAGAT ATCACGACACAGCTCCACG way ANOVA followed by a post hoc Tukey multiple comparisons test for comparing
MUC5AC CTGCCAGTCCTGCCTTTGTA GACCCTCCTCTCAATGGTGC more than 2 groups. For non-normally distributed data, statistical significance
TP63 CCTTACATCCAGCGTTTCGTAG TTTGTCTGTGTGCTCTGGGA
was determined using a non-parametric Mann-Whitney U test for comparing 2
groups or non-parametric one-way ANOVA with a post hoc Kruskal-Wallis multiple
UPK3A CGGTTCGGCTCGGCTG ATTCCTGGAAATGGCTGAGTCG
comparisons test for comparing more than 2 groups. Differences were considered to
SCGB3A2 TTACTCTGCTACTGCCTTCCTC CCTCCACAAGGTGCTCAACA be statistically significant at p<0.05.
SFTPA TGTGTGGGTCGCTGATTTCT GGGATACCAGGGCTTCCAAC
BMP4 AAGCGTAGCCCTAAGCATCA TGGTTGAGTTGAGGTGGTCA
Results
FSTL1 ACCACGATGTGGAAACGCTGGC TGCCATTACTGCCACACACAGGC
NOG TTCATGGCCACCTCGCCCCC ACTCTAGCCCTTTGATCTCG BMP4 negatively regulates pseudostratified epithelial differentiation in adult
SCGB1A1 CGTGTCATCGAAACCCTCCT GCTTTCTCTGGGCTTTTGGG human PTECs 7
CYP2F1 ACCCTCCTTAACACCGTCCA ATGGCGGTGAGGTACAGAAAG In comparison to untreated controls, BMP treated PTECs were characterized by
NKX2.1 CGCTCATTTGTTGGCGACTG CACTGAGAACGGAGTCGTGT
enlarged cell cytoplasm following 14-days of differentiation as evident by light mi-
croscopy (Figure 1B). Higher concentrations of BMP4 were associated with more
ITGA6 GCCAAAGATACTAGTGCCAAAGC CTTGAGGATCACCTACATAGAGCG
expanded cell cytoplasm compared to controls at day 14 (Figure 1C). A bilayer of
HEY1 GTAGTTAACTCCTCCCTGCCC GGGGACATGGAACCTAGAGC cells was observed in a cross-section of the submerged cultures of PTEC-controls at
HEY3 CTGCGTTCGCCATGAAGC GTTTCTCTATGATCCCTCTGCGT day 0. After 14 days of air exposure, cells formed polarized pseudostratified layers
HPRT1 AAGCCAGACTTTGTTGGATTTGA ACTGGCGATGTCAATAGGACTC with the luminal part covered with cells with a ciliated-like phenotype starting from
SDHA GGGAAGACTACAAGGTGCGG CTCCAGTGCTCCTCAAAGGG day 9 onwards in PTEC-controls. Strikingly, pseudostratified epithelium with ciliated
cells at the luminal side was not observed in BMP4 treated PTECs. Instead, a thin
B2M AAGCAGCATCATGGAGGTTTG AAGCAAGCAAGCAGAATTTGGA
single epithelial cell layer was observed at day 14 (Figure 1D). BMP4 treatment was
Abbreviations used: TEKT1, tektin-1; MUC5AC, mucin 5AC;TP63, tumor protein 63; UPK3A, associated with flattening of cuboidal epithelial cells (Figure 1E). These observations
uroplakin 3A; SCGB3A2, secretoglobin family 3A member 2; SFTPA, surfactant protein A; indicate that BMP4 inhibits normal pseudostratified epithelial differentiation and in-
BMP4, bone morphogenetic protein 4; FSTL1, follistatin-like 1; NOG, noggin; SCGB1A1, stead induces differentiation of a single layered epithelium with enlarged cytoplasm
secretoglobin family 1A member 1; CYP2F1, cytochrome P450 family 2 subfamily F member
1; NKX2.1,NK2 homeobox 1; ITGA6, integrin α6; HEY, hes-related family BHLH transcription in adult human PTECs.
factor with YRPW motif; HPRT1, hypoxanthine guanine phosphoribosyl transferase 1; SDHA,
succinate dehydrogenase complex flavoprotein subunit A; and B2M, β2-microglobulin.

126 127
BMP4 negatively regulates gene expression of ciliated, goblet, and basal cell
markers in adult human PTECs
To characterize the phenotype of the BMP4 induced cells in more detail, expression
of mRNA for markers of epithelial cell types, including ciliated, goblet, and basal
cells, was examined at days 0, 4, 9 and 14 of ALI culture. In PTEC-controls, the ciliated
cell marker Tektin-1 (p<0.05; Figure 2A,D) and the goblet cell marker MUC5AC
(p<0.05; Figure 2B,E) were significantly increased over time, whereas the basal cell

D. Cross sectioned view E.


Control BMP4 BMP4

Day 0 0 ng/ml
100 µm
100 µm
7
Day 4 1 ng/ml
100 µm 100 µm
100 µm

Day 9 10 ng/ml
100 µm
100 µm
100 µm

Day 14 100 ng/ml


100 µm 100 µm

100 µm

Figure 1. BMP4 inhibits pseudostratified epithelial differentiation in adult human PTECs.


(A) A schematic diagram of air-liquid-interface (ALI) culture model. (B) Representative top
view images of 100 ng/ml BMP4-treated PTECs morphology overtime and (C) in responses to Figure 2. BMP4 inhibits gene expression of ciliated, goblet, and basal cell markers in adult
different concentrations of BMP4 (1; 10; 100 ng/ml) at 14 days compared to PTEC-controls. human PTECs. Time-response effects of vehicle or BMP4 (100 ng/ml) on mRNA expression
Magnification 100x. Scale bars represent 500 µm. (D) Representative cross sectional images of theciliated cell marker Tektin-1 (A), the goblet cell marker MUC5AC (B), and the basal
of H&E staining of 100 ng/ml BMP4-treated PTECs morphology overtime and (E) in responses cell marker TP63 (C) in PTECs. Concentration response effects of BMP4 (1,10 and 100 ng/
to different concentrations of BMP4 (1, 10 and 100 ng/ml) at 14 days compared to PTEC- ml) on mRNA expression of Tektin-1 (D), MUC5AC (E), and TP63 (F) in PTECs at Day 14. Data
controls. Magnification 200x. Scale bars represent 100 µm. Images were taken using a light represent mean ± SEM in arbitrary units of 6 independent experiments from 6 different
microscope in each well of 6 independent experiments from 6 different donors. donors. *p<0.05 and **p<0.01 compared to time-matched PTEC-controls; #p<0.05 and
##
p<0.01 compared to Day 0 PTEC-controls; $p<0.05 and $$p<0.01 compared to day 14 PTEC-
controls; ns = not significant.

128 129
marker TP63 was significantly reduced (p<0.01; Figure 2C,F). Expression of mRNA
for markersof ciliated (Tektin-1), goblet (MUC5AC), and basal (TP63) cells was
reduced in BMP4-treated PTECs compared to controls at corresponding time points
(Figure 2A-C) and in response to different concentrations of BMP4 (Figure 2D-F).
These observations demonstrate that BMP4 represses goblet, ciliated, and basal cell
marker gene expression.

BMP4 induces variant club cell marker gene expression in adult human PTECs
Next, we studied the effects of BMP4 on club cells by determining the gene expression
of common (SCGB1A1, CYP2F1) and distal variant club cell markers (UPK3A, SCGB3A2,
SFTPA). In PTEC-controls, the mRNA expression of SCGB1A1, UPK3A (uroplakin
A), SCGB3A2 (secretoglobin 3A2), and SFTPA (surfactant protein A) was unaltered
(Figure 3A, C-E) whereas a time-dependent increase of CYP2F1 (p<0.01; Figure 3B)
was observed. In PTECs treated with BMP4, the mRNA expression of SCGB1A1 was
unaffected (Figure 3A), whereas CYP2F1 was significantly suppressed (p<0.01; Figure
3B). Intriguingly, the mRNA expression of variant club cell markers, including uroplakin
3A (p<0.05; Figure 3C), secretoglobin 3A2 (p<0.05; Figure 3D), and surfactant protein
A (p<0.01; Figure 3E) was markedly upregulatedby BMP4 during the 14 day period.
To confirm these findings, increasing concentrations of BMP4 were used to treat
PTECs for 14 days. As observed before, BMP4 did not alter SCGB1A1 (Figure 3F)
but significantly suppressed CYP2F1 (p<0.001; Figure 3G) mRNA expression in PTECs
treated with increasing concentrations of BMP4. Likewise, mRNA expression for the
distal variant club cell marker uroplakin 3A (p<0.001; Figure 3H), secretoglobin 3A2
(p<0.05; Figure 3I) and surfactant protein A (p<0.01; Figure 3J) were concentration-
dependently induced by BMP4 in PTECs. Taken together, these results demonstrate
that BMP4 suppresses marker gene expression for common club cells and promotes
markers for distal variant club cells in adult human PTECs.
7

Figure 3. BMP4 promotes variant club cell marker gene expression in adult human PTECs.
Time-response effects of vehicle or BMP4 (100 ng/ml) on mRNA expression of the common
club cell markers SCGB1A1 (A) and CYP2F1 (B), and the variant club cell markers uroplakin
3A (UPK3A; C), secretoglobin 3A2 (SGB3A2; D) and surfactant protein A (SFTPA; E) in PTECs.
Concentration response effects of BMP4 (1, 10 and 100 ng/ml) on mRNA expression of
SCGB1A1 (F), CYP2F1 (G), uroplakin 3A (H), secretoglobin 3A2 (I) and surfactant protein
A (J) in PTECs at Day 14. Data represent mean ± SEM in arbitrary units of 6 independent
experiments from 6 different donors. *p<0.05 and **p<0.01 compared to time-matched
PTEC-controls; ##p<0.01 compared to Day 0 PTEC-controls; $p<0.05 and $$p<0.01 compared
to Day 14 PTEC-controls; ns = not significant.

130 131
BMP4 induces time- and concentration-dependent increased of its own antagonist
Fstl1 during epithelial differentiation
To investigate the underlying mechanism of BMP4 in adult epithelial differentiation,
we explored the expression of BMP4 and its endogenous antagonists, FSTL1 and
Noggin. A previous study showed upregulation of BMP4 expression during epithelial
differentiation in ALI culture.20 In line with this, we observed that the mRNA
expression of BMP4 was upregulated in a time-dependent manner (p<0.01; Figure
4A) in PTEC-controls. Interestingly, BMP4 treatment significantly inhibited its own
gene transcription as evident by lower BMP4 mRNA levels (p<0.05; Figure 4A and
4D). The mRNA expression of FSTL1 was significantly reduced (P<0.05; Figure 4B)
whereas Noggin was unaltered (Figure 4C) after 14 days of air exposure. BMP4
concentration dependently increased mRNA expression of FSTL1 (P<0.01; Figure
4E), but not of Noggin (Figure 4F) at day 14.

BMP4 may regulate dedifferentiation of adult lung epithelial cells by modulating
expression of progenitor markers
At early stages of embryonic lung development, active BMP signaling is evident in
lung buds, which predominantly consist of lung-specific progenitor cells. Involvement
of BMP signaling in differentiation of lung progenitor cells from stem cells has been
reported.2 To address the question whether the expression of lung progenitor
markers in adult human PTECs is affected by BMP4, we determined the mRNA
expression of the lung progenitor markers NKX2.1 and ITGA6. In PTEC-controls,the
mRNA expression of NKX2.1 (p<0.01; Figure 5A) and ITGA6 (p<0.001; Figure 5B)
was significantly reduced over time, which was prevented by BMP4 (p<0.01 and
p<0.001; Figure 5A,B) in a concentration-dependent manner (Figure 5C,D). These
observations suggest that BMP4 modulates gene expression of lung progenitor
markers in adult human PTECs.
7
BMP4 induces Notch target gene expression during epithelial cell differentiation
To shed light on a possible mechanisms involved in BMP4-regulated distal variant
club cell differentiation, we examined the expression of Notch target genes (HEY1
and HEY3) and TGF-β1 target genes (ID-1 and SERPINE1) as previous studies sug-
gest that these genes are downstream target of active BMP signaling in pulmonary Figure 4. BMP4 induces gene expression of its antagonist Fstl1 in adult human PTECs
time and concentration dependently. Time-response effects of vehicle or BMP4 (100 ng/
cells.25,26 In PTEC-controls, a significant increase in mRNA expression of HEY1 (p<0.05; ml) on mRNA expression of BMP4 (A), FSTL1 (B), and Noggin (C) in PTECs. Concentration
Figure 6A) and ID-1 (p<0.05; Figure 6C), was observed, whereas HEY3 was unaltered response effects of BMP4 (1, 10 and 100 ng/ml) on mRNA expression of BMP4 (D), FSTL1
(Figure 6B) and SERPINE1 decreased (p<0.01; Figure 6D) over the course of 14 days (E), and Noggin (F) in PTECs at Day 14. Data represent mean ± SEM in arbitrary units of 6
independent experiments from 6 different donors. *p<0.05 and **p<0.01 compared to time-
of differentiation. BMP4 significantly induced mRNA expression of HEY1, HEY3, ID- matched PTEC-controls; #p<0.05 and ##p<0.01 compared to Day 0 PTEC-controls; $p<0.05
1, and SERPINE1 (p<0.01 all; Figure 6A-D) in a concentration-dependently manner and $$p<0.01 compared to Day 14 PTEC-controls.
(p<0.001; Figure 6G-H). These data suggest that both Notch and TGF-β signaling are
downstream of BMP4.

132 133
Figure 5. BMP4 induces gene expression of lung progenitor cell markers in adult human
PTECs. Time-response effects of vehicle or BMP4 (100 ng/ml) on mRNA expression ofNKX2.1
transcription factor(A) and integrin subunit α6 (B) in PTECs. Concentration response effects of
BMP4 (1; 10; 100 ng/ml) on mRNA expression of NKX2.1 transcription factor(C) and integrin
subunit α6 (D) in PTECs. Data represent mean ± SEM in arbitrary units of 6 independent 7
experiments from 6 different donors. *p<0.05, **p<0.01 and ***p<0.001 compared to time-
matched non-treated controls. #p<0.05, ##p<0.01 and ###p<0.001 compared to Day 0 PTEC-
controls; $p<0.05 and $$p<0.01 compared to Day 14 PTEC-controls.

Figure 6. BMP increases Notch and TGF target gene expression in adult human PTECs.
Time-response effects of vehicle or BMP4 (100 ng/ml) on mRNA expression of Notch targets
HEY1 (A) and HEY3 (B) and of TGF-β1 targets ID-1 (C) and SERPINE1 (D) in PTECs. Concentra-
tion response effects of BMP4 (1, 10 and 100 ng/ml) on mRNA expression of HEY1 (E), HEY3
(F), ID-1 (G) and SERPINE1 (H) in PTECs at Day 14. Data represent mean ± SEM in arbitrary
units of 6 independent experiments from 6 different donors. **p<0.01 and ***p<0.001 com-
pared to time-matched PTEC-controls; #p<0.05, ##p<0.01 compared to Day 0 PTEC-controls;
$p<0.05, $$p<0.01 and $$$p<0.001compared to Day 14 PTEC-controls.

134 135
Table 1. Summary
BMP4 induces distal variant club cell differentiation in adult human airway Characteristics Control BMP4
epithelium Pseudostratified Widened, flatten
To confirm that BMP4 inhibits goblet cell differentiation and promotes variant Morphology
columnar cells
club cell differentiation, periodic acid schiff (PAS) staining and immunostaining for TEKT1 + - Ciliated cell marker
distal variant club cell markers (SCGB3A/SFTPA) were performed, respectively. PAS MUC5AC + - Goblet cell marker
staining on cross-sections of PTEC-controls showed more intense staining intensity TP63 - - Basal cell marker
compared to BMP4 treated cells at day 14 (Figure 7A). In contrast, the expression of SCGB1A1 - -
distal variant club cell markers was more apparent in BMP4 treated cells compared CYP2F1 + -
Common club cell markers
Gene
to PTEC-controls at 14 days (Figure 7B). Taken together, these findings indicate that expression UPK3A - +
BMP4 may drive epithelial differentiation toward distal variant club cells in adult
SCGB3A - + Variant club cell markers
human PTECs. The observed mRNA expression profile of PTECs treated for 14 days
SFTPA - +
with or without BMP4 is summarized in Table 1.
NKX2.1 - +
Progenitor cell markers
ITGA6 - +
Abbreviations used: TEKT1, tektin-1; MUC5AC, mucin 5AC;TP63, tumor protein 63; SCGB1A1,
secretoglobin family 1A member 1; CYP2F1, cytochrome P450 family 2 subfamily F member
1; UPK3A, uroplakin 3A; SCGB3A2, secretoglobin family 3A member 2; SFTPA, surfactant
protein A; NKX2.1,NK2 homeobox 1; and ITGA6, integrin α6.

Acknowledgements
This study was financially supported by a grant from the Netherlands Lung Foundation
(grant 3.2.12.083).

Conflict of interests
The authors declare no financial conflict of interests.

Author contributions 7
NPT and RG conceived the study and designed the experiments. NPT carried out the
experiments, analyzed the data, and drafted the manuscript. FE performed some
experiments and analyzed the data. All co-authors approved the final version of the
manuscript.
Figure 7. BMP4 inhibits goblet cells and promotes distal variant club cell differentiation
in adult human PTECs. Representative images of PTEC-BMP4 compared to PTEC-controls
stained using periodic acid-schiff (PAS, A) or the variant club cell markers secretoglobin 3A2
or surfactant protein A (B) at Day 14. Magnification 200x. Scale bars represent 100 µm.

136 137
References cells via a BMP4-NFATc1-thrombospondin-1 axis. Cell. 2014;156(3):440-455. doi:10.1016/j.
1. Bellusci S, Henderson R, Winnier G, Oikawa T, Hogan BL. Evidence from normal expression cell.2013.12.039.
and targeted misexpression that bone morphogenetic protein (Bmp-4) plays a role in mouse 22. Mertens TCJ, Hiemstra PS, Taube C. Azithromycin differentially affects the IL-13-induced
embryonic lung morphogenesis. Dev Camb Engl. 1996;122(6):1693-1702. expression profile in human bronchial epithelial cells. Pulm Pharmacol Ther. 2016;39:14-20.
2. Sountoulidis A, Stavropoulos A, Giaglis S, et al. Activation of the canonical bone morphogenetic doi:10.1016/j.pupt.2016.05.005.
protein (BMP) pathway during lung morphogenesis and adult lung tissue repair. PloS One. 23. Amatngalim GD, van Wijck Y, de Mooij-Eijk Y, et al. Basal cells contribute to innate immunity
2012;7(8):e41460. doi:10.1371/journal.pone.0041460. of the airway epithelium through production of the antimicrobial protein RNase 7. J Immunol
3. Weaver M, Yingling JM, Dunn NR, Bellusci S, Hogan BL. Bmp signaling regulates proximal-distal Baltim Md 1950. 2015;194(7):3340-3350. doi:10.4049/jimmunol.1402169.
differentiation of endoderm in mouse lung development. Dev Camb Engl. 1999;126(18):4005- 24. Ruijter JM, Ramakers C, Hoogaars WMH, et al. Amplification efficiency: linking baseline and bias
4015. in the analysis of quantitative PCR data. Nucleic Acids Res. 2009;37(6):e45. doi:10.1093/nar/
4. Warburton D, Bellusci S. The molecular genetics of lung morphogenesis and injury repair. gkp045.
Paediatr Respir Rev. 2004;5 Suppl A:S283-287. 25. Rock JR, Gao X, Xue Y, Randell SH, Kong Y-Y, Hogan BLM. Notch-Dependent Differentiation of Adult
5. Shi W, Chen F, Cardoso WV. Mechanisms of lung development: contribution to adult lung disease Airway Basal Stem Cells. Cell Stem Cell. 2011;8(6):639-648. doi:10.1016/j.stem.2011.04.003.
and relevance to chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2009;6(7):558- 26. Cibois M, Luxardi G, Chevalier B, et al. BMP signalling controls the construction of vertebrate
563. doi:10.1513/pats.200905-031RM. mucociliary epithelia. Development. 2015;142(13):2352-2363. doi:10.1242/dev.118679.
6. Tadokoro T, Gao X, Hong CC, Hotten D, Hogan BLM. BMP signaling and cellular dynamics during 27. Rackley CR, Stripp BR. Building and maintaining the epithelium of the lung. J Clin Invest.
regeneration of airway epithelium from basal progenitors. Dev Camb Engl. 2016;143(5):764-773. 2012;122(8):2724-2730. doi:10.1172/JCI60519.
doi:10.1242/dev.126656. 28. Wells JM, Melton DA. Early mouse endoderm is patterned by soluble factors from adjacent germ
7. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell layers. Dev Camb Engl. 2000;127(8):1563-1572.
populations. Nat Med. 2014;20(8):822-832. doi:10.1038/nm.3642. 29. Mou H, Vinarsky V, Tata PR, et al. Dual SMAD Signaling Inhibition Enables Long-Term
8. Li F, He J, Wei J, Cho WC, Liu X. Diversity of Epithelial Stem Cell Types in Adult Lung. Stem Cells Expansion of Diverse Epithelial Basal Cells. Cell Stem Cell. 2016;19(2):217-231. doi:10.1016/j.
Int. 2015;2015:e728307. doi:10.1155/2015/728307. stem.2016.05.012.
9. Saetta M, Turato G, Baraldo S, et al. Goblet cell hyperplasia and epithelial inflammation 30. Reynolds SD, Giangreco A, Power JH, Stripp BR. Neuroepithelial bodies of pulmonary airways
in peripheral airways of smokers with both symptoms of chronic bronchitis and chronic serve as a reservoir of progenitor cells capable of epithelial regeneration. Am J Pathol.
airflow limitation. Am J Respir Crit Care Med. 2000;161(3 Pt 1):1016-1021. doi:10.1164/ 2000;156(1):269-278. doi:10.1016/S0002-9440(10)64727-X.
ajrccm.161.3.9907080. 31. Barnes PJ. CLub cells, their secretory protein, and copd. Chest. 2015;147(6):1447-1448.
10. Pilette C, Godding V, Kiss R, et al. Reduced epithelial expression of secretory component in small doi:10.1378/chest.14-3171.
airways correlates with airflow obstruction in chronic obstructive pulmonary disease. Am J 32. Shijubo N, Itoh Y, Yamaguchi T, et al. Clara cell protein-positive epithelial cells are reduced in
Respir Crit Care Med. 2001;163(1):185-194. doi:10.1164/ajrccm.163.1.9912137. small airways of asthmatics. Am J Respir Crit Care Med. 1999;160(3):930-933. doi:10.1164/
11. Gamez AS, Gras D, Petit A, et al. Supplementing defect in club cell secretory protein attenuates ajrccm.160.3.9803113.
airway inflammation in COPD. Chest. 2015;147(6):1467-1476. doi:10.1378/chest.14-1174. 33. Rokicki W, Rokicki M, Wojtacha J, Dżeljijli A. The role and importance of club cells (Clara cells) in
12. Shaykhiev R, Crystal RG. Early events in the pathogenesis of chronic obstructive pulmonary the pathogenesis of some respiratory diseases. Kardiochirurgia Torakochirurgia Pol Pol J Cardio-
disease. Smoking-induced reprogramming of airway epithelial basal progenitor cells. Ann Am Thorac Surg. 2016;13(1):26-30. doi:10.5114/kitp.2016.58961.
Thorac Soc. 2014;11 Suppl 5:S252-258. doi:10.1513/AnnalsATS.201402-049AW. 34. Myllärniemi M, Lindholm P, Ryynänen MJ, et al. Gremlin-mediated decrease in bone
morphogenetic protein signaling promotes pulmonary fibrosis. Am J Respir Crit Care Med.
13. Bernard A, Marchandise FX, Depelchin S, Lauwerys R, Sibille Y. Clara cell protein in serum and
2008;177(3):321-329. doi:10.1164/rccm.200706-945OC.
7
bronchoalveolar lavage. Eur Respir J. 1992;5(10):1231-1238.
14. Vestbo J, Edwards LD, Scanlon PD, et al. Changes in Forced Expiratory Volume in 1 Second over 35. De Langhe E, Cailotto F, De Vooght V, et al. Enhanced endogenous bone morphogenetic protein
Time in COPD. N Engl J Med. 2011;365(13):1184-1192. doi:10.1056/NEJMoa1105482. signaling protects against bleomycin induced pulmonary fibrosis. Respir Res. 2015;16:38.
15. Park HY, Churg A, Wright JL, et al. Club cell protein 16 and disease progression in chronic doi:10.1186/s12931-015-0202-x.
obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(12):1413-1419. 36. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary
doi:10.1164/rccm.201305-0892OC. fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878.
16. Lomas DA, Silverman EK, Edwards LD, et al. Evaluation of serum CC-16 as a biomarker for COPD 37. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by
in the ECLIPSE cohort. Thorax. 2008;63(12):1058-1063. doi:10.1136/thx.2008.102574. Inducing Oncostatin M. J Immunol Baltim Md 1950. 2015;195(8):3546-3556. doi:10.4049/
17. Guerra S, Halonen M, Vasquez MM, et al. The relation of circulating CC16 to lung function growth, jimmunol.1501105.
decline, and development of COPD across the lifespan. Lancet Respir Med. 2015;3(8):613-620. 38. Murphy N, Gaynor KU, Rowan SC, et al. Altered Expression of Bone Morphogenetic Protein
doi:10.1016/S2213-2600(15)00196-4. Accessory Proteins in Murine and Human Pulmonary Fibrosis. Am J Pathol. 2016;186(3):600-
18. Kaartinen L, Nettesheim P, Adler KB, Randell SH. Rat tracheal epithelial cell differentiation in 615. doi:10.1016/j.ajpath.2015.10.032.
vitro. In Vitro Cell Dev Biol Anim. 1993;29A(6):481-492. 39. Miller M, Esnault S, Kurten RC, et al. Segmental allergen challenge increases levels of airway
19. Whitcutt MJ, Adler KB, Wu R. A biphasic chamber system for maintaining polarity of follistatin-like 1 in patients with asthma. J Allergy Clin Immunol. 2016;138(2):596-599.e4.
differentiation of cultured respiratory tract epithelial cells. Vitro Cell Dev Biol J Tissue Cult Assoc. doi:10.1016/j.jaci.2016.01.019.
1988;24(5):420-428. 40. Liu M, Liu R, Wu H, et al. Radix puerariae extracts ameliorate paraquat-induced pulmonary
20. Ross AJ, Dailey LA, Brighton LE, Devlin RB. Transcriptional profiling of mucociliary differentiation fibrosis by attenuating follistatin-like 1 and nuclear factor erythroid 2p45-related factor-2
in human airway epithelial cells. Am J Respir Cell Mol Biol. 2007;37(2):169-185. doi:10.1165/ signalling pathways through downregulation of miRNA-21 expression. BMC Complement Altern
rcmb.2006-0466OC. Med. 2016;16:11. doi:10.1186/s12906-016-0991-6.
21. Lee J-H, Bhang DH, Beede A, et al. Lung stem cell differentiation in mice directed by endothelial 41. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal

138 139
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616.
42. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP)
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A.
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108.
43. Maeda Y, Chen G, Xu Y, et al. Airway epithelial transcription factor NK2 homeobox 1 inhibits
mucous cell metaplasia and Th2 inflammation. Am J Respir Crit Care Med. 2011;184(4):421-429.
doi:10.1164/rccm.201101-0106OC.
44. Chapman HA, Li X, Alexander JP, et al. Integrin α6β4 identifies an adult distal lung epithelial
population with regenerative potential in mice. J Clin Invest. 2011;121(7):2855-2862.
doi:10.1172/JCI57673.
45. Guha A, Vasconcelos M, Cai Y, et al. Neuroepithelial body microenvironment is a niche
for a distinct subset of Clara-like precursors in the developing airways. Proc Natl Acad Sci.
2012;109(31):12592-12597. doi:10.1073/pnas.1204710109.
46. Paul MK, Bisht B, Darmawan DO, et al. Dynamic changes in intracellular ROS levels regulate
airway basal stem cell homeostasis through Nrf2-dependent Notch signaling. Cell Stem Cell.
2014;15(2):199-214. doi:10.1016/j.stem.2014.05.009.
47. Lafkas D, Shelton A, Chiu C, et al. Therapeutic antibodies reveal Notch control of transdifferentiation
in the adult lung. Nature. 2015;528(7580):127-131. doi:10.1038/nature15715.

140 141
CHAPTER 8 1

GENERAL DISCUSSION AND


SUMMARY

143
Preface Although minimal pulmonary vascular remodeling was observed in large
muscular and elastic arteries in Fstl1-eKO mice, we found increased percentages
Chronic obstructive pulmonary disease (COPD) is characterized by progressive of actin positive small pulmonary vessels at the later stages of development at
irreversible airflow obstruction in which excessive and persistent inflammation postnatal day 21 (Chapter 2). In addition, we found that the right ventricular
of the airways plays an important role. As outlined in the general introduction output was decreased in Fstl1-eKO mice at later stages of pulmonary development,
(Chapter 1), the proper balance between bone morphogenetic protein-4 (BMP4) as determined by an echocardiography (Chapter 2). Furthermore, analysis of the
and its endogenous inhibitor Follistatin-like 1 (Fstl1) is crucial for normal embryonic underlying mechanisms leading to the lethality of Fstl1-eKO mice reveals that
lung morphogenesis1,2 and in adult steady-state conditions for proper biological phosphorylated Smad1/5/8 - an indicator of active BMP signaling - and BMP-
functions.3–5 The immunomodulatory functions of Fstl1 has been studied in various regulated proteins (Jagged1, endoglin, and endothelin-1) are increased at the
inflammatory diseases.6–8 However, the functional role of Fstl1 in postnatal lung early stages of development at postnatal day 7 (Chapter 2). A dynamic regulation
development and COPD lungs has not been previously explored. Recently, several of vasoconstriction is synergistically controlled by molecular changes, including an
studies demonstrated the involvement of Fstl1 in pulmonary fibrosis and severe increase in endothelin-1 release30,31 and/or morphological changes in smooth muscle
allergic asthma.9–13 Our studies described in this thesis reveal that Fstl1 is required for tissues, including an increase in α-smooth muscle actin and increased proliferation
pulmonary vascular development postnatally (Chapter 2). In addition, we show that of vascular smooth muscle cells.32,33
Fstl1 protein expression is increased in airway epithelium of COPD patients (Chapter In wildtype mice, a transition between a molecular to a morphological regulation
3). Addressing the potential implications of increased Fstl1 expression in airway of vasoconstriction was observed (Chapter 2). Increased protein expression of
epithelium, we found Fstl1 to be a pro-inflammatory mediator of airway epithelial- endothelin-1 simultaneously occurred with decreased percentages of actin positive
driven inflammation (Chapter 5). Furthermore, we found that BMP4 signaling small vessels at postnatal day 14 compared to day 7. This transition was normal
regulates variant club cell differentiation in adult airway epithelium (Chapter 7). and non-lethal (Chapter 2). However in the Fstl1-eKO mice, the protein expression
of endothelin-1 was already high at postnatal day 7 and remained high at day 21
whereas the percentages of actin positive vessels did not decrease by postnatal
1. The role of Fstl1 in lung development
day 21 (Chapter 2). The combination of a high endothelin-1 expression and a high
The development of the lung is orchestrated by tight temporal and spatial control
percentage of actin positive small pulmonary vessels may lead to an increase in
of multiple signaling pathways to form functional airway and vascular systems.
pulmonary vascular resistance due to vasoconstriction (Chapter 2). However,
BMP signaling is essential in dynamic processes of vessel growth and maturation,
no difference was observed in the pulmonary vascular resistance as reflected by
by governing vascular smooth muscle and endothelial cell proliferation and
the ratio of pulmonary acceleration time over ejection time (PAT/ET) in Fstl1-eKO
migration.14–19 Highly coordinated regulation of BMP signaling is essential for proper
mice compared to age-matched controls either at postnatal day 7 or 21 (Chapter
airway and vascular tree branching during lung development to enable proper lung
2). The pulmonary vascular resistance was measured in pulmonary valves of the
functioning.20 Several genetic vascular diseases have been linked with aberrant BMP
large pulmonary arteries where there was no pulmonary stenosis observed during
signaling, including familial pulmonary arterial hypertension (PAH).18,21–28 In line
echocardiography measurement in Fstl1-eKO mice. This may reflect very early stages
with such a central role for BMP signaling, whole body Fstl1 knockout (Fstl1-KO)
of pulmonary vascular dysfunction in Fstl1-eKO mice. Interestingly, such phenotype
mice display a smaller lung morphology and thickening of the alveolar septa. Fstl1-
is commonly observed in patients with PAH, where physiological and structural
KO mice suffer from respiratory distress and died shortly after birth,1,2 indicating 8
changes are initiated by molecular changes in BMP signaling followed by vascular
that Fstl1 is required for lung morphogenesis and alveolarization. Intriguingly, we
dysfunction and remodeling at later stages of the disease.34 It is likely that an increase
found that mice with endothelial-specific Fstl1 deletion (Fstl1-eKO) also suffered
of the vascular resistance occurs at the site of actin remodeling in small pulmonary
from respiratory distress and died 3 weeks after birth (Chapter 2). Postnatal lethality
vessels, due to resistance being a function of 1/r4 (according to Hagen-Poiseuille
of the Fstl1-eKO mice suggests a significant role of endothelial Fstl1 in postnatal
law), where “r” is the diameter of the vessel lumen.35
pulmonary vasculature development to support proper neonatal development,
In Fstl1-eKO mice, increased pulmonary levels of endothelin-1 at postnatal day
including the lung. Fstl1-eKO mice died at postnatal day 21 which is around the
7 may transiently increase pulmonary vascular resistance. Since pulmonary arteries
alveolar stage of lung development at postnatal days of 5-30, which is critical for
are connected to the heart, this initial increase in pulmonary vascular resistanceleads
alveolar and vascular network formation and maturation.20,29 Branching of the
to a decrease in right ventricular output to the lung at postnatal day 21 (Chapter
airway and vascular network must be coordinated to optimize exposure of blood
2). Consequently, morphological changes were observed in the heart and lungs of
to oxygen.20 The formation of the distal small pulmonary vascular network may be
Fstl1-eKO mice at postnatal day 21 such as right ventricle hypertrophy and small
disrupted in Fstl1-eKO mice and result in respiratory distress (Chapter 2).
pulmonary vascular remodeling, respectively (Chapter 2). The progressive and

144 145
irreversible defects in cardiopulmonary structures in Fstl1-eKO mice may contribute
to the respiratory distress, presumably because the remodeled small pulmonary
vessels are unable to adequately carry blood to the lungs. These may lead to a lack of
oxygen delivery to the brain and ultimately death. Therefore, it would be interesting
to examine the levels of oxygen and carbon dioxide in the blood in addition to
determine the radius and the actin content of small pulmonary vessels in the follow
up studies. Altogether, it is tempting to speculate that molecular changes in Fstl1-
eKO mice are transient and initiated earlier during postnatal development whereas
physiological functions and morphological consequences may manifest at later stages
of postnatal development. An alternative explanation could be that dysregulation of
BMP signaling by itself may not be adequate to affect structure and functions of
large pulmonary vasculature at early stages of development, but instead is more
pronounced at later stages of development. It is also possible that the molecular and
morphological changes in the lungs are a secondary consequence of the right heart
dysfunction due to thickening/muscularization of the right ventricle. The thickened
right ventricle could be associated with reduced right ventricular output that leads
to lung hypoxia. As consequences, the percentages of actin positive small vessels did
not decrease whilst the expression of endothelin-1 was high in the lungs at postnatal
day 21 to push the blood flow through the pulmonary capillaries to meet the oxygen
demand. It would be worthwhile in future studies to dissect cardiovascular structure
and functions in Fstl1-eKO mice to confirm the main cause of death of Fstl1-eKO mice
in addition to revealing what the function of Fstl1 is in cardiovascular system. The
developmental role of endothelial Fstl1 in modulating BMP/Smad signaling during
postnatal development of pulmonary vasculature is illustrated in Figure 1.

2. The role of Fstl1 in airway inflammation in COPD


One of the hallmarks of COPD pathogenesis is the exaggerated activation of the
innate immune system and abnormal tissue repair in response to long term
exposure to pneumotoxic inhaled pollutants, such as cigarette smoke. In turn, this Figure 1. The developmental role of endothelial Follistatin-like 1 in modulating BMP/
leads to airway epithelial remodeling and physiological changes which consequently Smad signaling during postnatal development of pulmonary vasculature. In the presence
contribute to the not fully reversible airflow limitation.36,37 As a first barrier to the of endothelial Fstl1 in the wildtype mice, BMP-induced phosphorylation of Smad1/5 and
subsequent downstream target genes are maintained for normal endothelial function
external milieu, airway epithelium secretes inflammatory mediators with paracrine and vascular homeostasis. Deletion of endothelial Fstl1 in Fstl1-eKO mice is associated 8
effects on neighboring mesenchymal cells, including lung fibroblasts and airway with overactivation of BMP-induced phosphorylated Smad1/5 and its downstream target
smooth muscle cells. Interestingly, a role for Fstl1 in cell-cell communication has genes, including Endoglin, Jag1, Gata2. In turn, increased Gata2 presumably increases the
expression of the most potent vasoconstrictor endothelin-1 which could contribute to
been previously reported.6,9 A growing body of evidence indicates pro-inflammatory vascular remodeling and vasoconstriction.
roles of Fstl1 in mediating secretion of inflammatory cytokine in the pathogenesis of
was expressed in different parts of the lung tissue, including airway epithelium,
several inflammatory diseases and in worsening the symptoms of these diseases.6,7,38
endothelium, vascular smooth muscle cells, and inflammatory cells (Chapter 3).
However, the link between Fstl1 and COPD pathogenesis has not been explored
The intensity of Fstl1 staining, used as an indicator of Fstl1 protein abundance, was
previously.
higher in airway epithelium, endothelium, vascular smooth muscle bundles, and
Our studies on the expression of Fstl1 in COPD demonstrated that Fstl1 expression
inflammatory cells of both COPD stage II and IV patients (all ex-smokers) compared to
was markedly increased in lung homogenates of non-COPD smokers and both stage II
individuals without airflow obstruction (Chapter 3). This might indicate that increased
and IV COPD patients compared to individuals without airflow obstruction (Chapter
Fstl1 expression associates with COPD. This thesis aims to address the potential
3). Further examination on lung tissue of COPD patients revealed that Fstl1 protein
implications of increased Fstl1 expression related to COPD pathogenesis (Chapter

146 147
5 and 7). Intriguingly, the expression of Fstl1 protein in lung homogenates of non- To mimic the observed upregulation of Fstl1 in airway epithelium of patients
COPD current smokers and ex-smokers was also higher compared to non-COPD non- with COPD and study its impact on inflammatory cytokine release from pulmonary
smokers, indicating an effect of ever smoking, presumably through smoke-induced mesenchymal cells, Fstl1 was overexpressed in epithelial cells. Overexpression of
inflammation (Chapter 3). It could be that acute induction of inflammatory mediator Fstl1 in bronchial epithelial cells increased the release of IL-6 and GM-CSF protein,
release and subsequent local inflammation involving recruitment of inflammatory indicating a pro-inflammatory role (Chapter 5). Fstl1 conditioned medium (Fstl1
cells to the lung promoted by cigarette smoke persist after smoking cessation. Given CM) derived from Fstl1 overexpressing bronchial epithelial cells was used to treat
that Fstl1 is a TGF-β1 inducible factor and TGF-β1 release is increased from bronchial pulmonary mesenchymal cells. Interestingly, we found that Fstl1 CM increased IL-6
epithelial cells after cigarette smoke treatment,39 this suggests that TGF-β1 could and IL-8 transcription and protein release from mesenchymal cells, suggesting that
mediate the observed upregulation of Fstl1 in response to cigarette smoke. Fstl1 may regulate pro-inflammatory cytokine release in the lung. Overexpression
We next explored the potential roles of Fstl1 in pulmonary cells in regulating of Fstl1 in bronchial epithelial cells triggers inflammatory cytokine release from
local inflammation in the lung (Chapter 5). We examined the expression of Fstl1 and these cells and CM derived from these Fstl1-overexpressing bronchial epithelial cells
BMP4 in pulmonary cells lines and found that in the respiratory system, bronchial induces cytokine release from pulmonary mesenchymal cells (Chapter 5). Although
epithelial cells express and secrete Fstl1 the most compared to lung mesenchymal previous studies reported that Fstl1 promotes direct release of IL-6 and IL-8 from
cells, such as lung fibroblasts and airway smooth muscle cells (Chapter 5). This is in mesenchymal cells.6,8,42,43, our data using recombinant Fstl1 treatment as well as gain-
line with our observation in lung tissue of individuals with normal lung function, in and-loss of Fstl1 functions in lung fibroblasts showed no effect at all on baseline or
which we demonstrated that Fstl1 is predominantly expressed by airway epithelial IL-1β-induced IL-6 and IL-8 protein release, suggesting that Fstl1 alone is insufficient
cells, particularly in the apical part of ciliated cells and to a lesser extent in airway to induce pro-inflammatory cytokine release from mesenchymal cells. Therefore,
smooth muscle cells (Chapter 3). Other studies have suggested that Fstl1 is a we speculate that Fstl1 may induce the release of inflammatory mediators from
mesenchymal-derived factor in the heart and joint.38,40 Given that Fstl1 is known bronchial epithelial cells which in turn regulate the inflammatory cytokine release
as an endogenous antagonist of BMP signaling in the lung,1,2 we examined the from mesenchymal cells. We identified several secreted factors as candidates for the
phosphorylation of BMP-specific Smad1/5 in lung homogenates of COPD patients effect of Fstl1 CM on IL-6 and IL-8 release from lung fibroblasts, namely IL-6, IL-8,
(all ex-smokers) compared to non-COPD with different smoking status. We observed GM-CSF and RANTES and confirmed their functional contribution using neutralizing
that phosphorylated Smad1/5 was significantly reduced in the lung homogenates antibody experiments. Collectively, our study provides the first in vitro demonstration
of non-COPD current smokers, non-COPD ex-smokers, and COPD stage II and stage that Fstl1 induces the transcription and release of inflammatory cytokines, such as
IV patients compared to non-COPD never-smokers (Chapter 3). Reduced levels of GM-CSF and IL-6, in bronchial epithelial cells, which subsequently are able to induce
phosphorylated Smad1/5 could be explained by increased expression of Fstl1 but the release of IL-6 and IL-8 from mesenchymal cells. In summary, we provide evidence
may also be explained by reduced pulmonary BMP4 expression. We found that there that Fstl1 is a pro-inflammatory factor that mediates epithelial-mesenchymal
was no significant difference in BMP4 protein levels in the lung homogenates among communication in regulating inflammatory cytokine release. Therefore, Fstl1 might
groups (Chapter 3). In pulmonary cell lines, BMP4 is predominantly expressed by contribute to the recruitment of inflammatory cells to the lung. In line with our
mesenchymal cells, such as lung fibroblasts and airway smooth muscle cells (data findings, recent in vitro, in vivo, and proteomic studies demonstrate the involvement
not shown) and human bronchial epithelial cells expressed and secreted the lowest of Fstl1 in the pathological process of respiratory diseases, including lung fibrosis
levels of BMP4. BMP4 expression is localized in the distal bud of the developing and asthma.9,11–13 Collectively, these findings may provide a basis for targeting
8
lung at the pseudoglandular stage (E11-E16.5), coordinating the proximal-distal Fstl1 as therapeutic approach for epithelial-driven airway inflammation. The pro-
axis of endoderm by controlling the differentiation of distal bud epithelial cells.41 inflammatory role of Fstl1 in coordinating epithelial-mesenchymal communication
Accordingly, phosphorylation of Smad1/5 in pulmonary cell lines followed the in inflammatory cytokine release in the lung is depicted in Figure 2.
pattern of BMP ligand levels, showing higher degree of canonical BMP signaling in
airway smooth muscle cells and lung fibroblasts compared to bronchial epithelial 3. Implications of Fstl1 in goblet cell metaplasia and basal cell hyperplasia in
cells. Differential cellular source of ligands and their inhibitors in the lung suggest COPD: The role of BMP4 in adult epithelial differentiation
potential crosstalk between epithelial and mesenchymal cells in governing biological BMP signaling is pivotal in governing proximal-to-distal axis patterning of airway
functions, including regulation of inflammatory cytokine release. These findings epithelium in fetal and neonatal lung development. In adult lungs, active BMP
provoked our study to understand the precise roles of Fstl1 in airway inflammation signaling airway is required for epithelial regeneration following injury.44–49 BMP4 is
in healthy subjects and to discuss the potential contribution to the development of a secreted mesenchymal derived factor that determines epithelial cell fate during
COPD (Chapter 5). lung development41. In addition, BMP4 is required for epithelial cell differentiation in

148 149
air-liquid interface (ALI) cultures, as its expression is time-dependently upregulated flattened morphology and are characterized by lack of secretoglobin 1A member
in an transcriptional profiling study.50 1 (SCGB1A1, also known as a club cell 10 kDa secretory protein CC10 or CC16) and
The airway epithelium is the first protective physical and functional barrier of the cytochrome p450 Cyp2f expression. In contrast, common club cells show typical
airways and lung against the external environment. The pseudostratified epithelium cuboidal epithelial phenotype with high expression of SCGB1A1 and cytochrome
lining of adult airways is composed of a mixed balance of goblet cells, ciliated cells, p450 Cyp2f making them sensitive to naphthalene injury.62,64,65 Loss of (variant) club
basal cells and club cells (formerly known as clara cells) that are properly distributed cells has been observed in COPD.60,61,66,67 Low serum levels of SCGB1A1 are not only
along the tracheobronchial tree.51,52 Mucus secreting goblet cells are predominantly associated with COPD prevalence and severity, but also with lung function decline
expressed in the proximal epithelium, along with basal cells, club cells, and ciliated in COPD as determined by an accelerated forced expiratory volume in one second
cells.53,54 In distal small bronchioles, secretory club cells, ciliated cells, and pulmonary (FEV1).68–72
neuroendocrine cells are abundant. Respiratory alveoli are covered with a thin During normal mucociliary differentiation, basal cells directly or indirectly
single layer of flat type I and cuboidal type II alveolar epithelial cells.55–57 (through intermediate club cells) give rise to goblet and ciliated cells. Intriguingly,
Cigarette smoke alters the composition of airway epithelium and contributes our study reveals that BMP4 drives basal cell differentiation towards variant club
to airway epithelial remodeling in COPD.58 Airway epithelial remodeling in COPD cells and suppresses further differentiation towards goblet and ciliated cells in adult
is characterized by goblet cell metaplasia, basal cell hyperplasia as well as loss of primary human bronchial epithelial cells (Chapter 7). Although a previous study
functional club cells in peripheral and central airways.59–61 Variant club cells are has demonstrated a role of BMP4 in proximal-distal epithelial differentiation in the
secretory cells that reside primarily in distal lungs with protective roles by secreting developing lung, 45 we are the first to show that BMP4 promotes variant club cell
uteroglobin, secretoglobin, and surfactant protein-like protein which have anti- differentiation in adult human airway epithelial cells in ALI cultures.
inflammatory functions and are important for epithelial regeneration.62 Variant In line with previous studies,73,74 we also found that functional BMP4 signaling
club cells possess stem cell-like properties and are capable of self-renewal and (Smad1/5/8 phosphorylation) is reduced whereas Fstl1 expression is increased in
differentiation into ciliated and goblet cells.57,63 Variant club cells have a distinct COPD lungs (Chapter 3). Increased Fstl1 in COPD airway epithelium might drive
epithelial differentiation towards goblet cell metaplasia, basal cell hyperplasia, and
loss of club cells which are typical features of remodeled distal COPD epithelium,
by inhibiting the negative regulation of the BMP pathway on goblet cell and basal
cell differentiation. Our study provides a plausible connection between the loss of
epithelial cells
club cells, and goblet cell and basal cell metaplasia on one hand and the observed
reduction in BMP/Smad signaling in COPD patients on the other hand.73,74
Fstl1
Moreover, we observed that BMP4 induces expression of its own endogenous
GM-CSF
antagonist Fstl1 during airway epithelial differentiation in a time- and concentration-
RANTES dependent manner (Chapter 7), implying that Fstl1 may be involved in BMP functions
IL-6
IL-8
during epithelial differentiation. Future studies using Fstl1 CM or Fstl1 neutralizing
antibody ββ and retraction of BMP4 treatment after 14 days of treatment in BMP4-
treated cells may answer whether the effect of BMP4 is reversible and whether
8
the switch towards the variant club cell differentiation program can be re-directed
towards a ciliated cell differentiation program. It has been shown previously that
mesenchymal cells BMP antagonists can function as a positive regulator of basal cell proliferation by
antagonizing BMP functions to block basal cell proliferation.75,76 Likewise, inhibition
BMP/TGF-β/Smad signaling promotes basal cell hyperplasia in primary human
IL-6
IL-8
epithelial cells.77 A persistent increase of the BMP antagonist Fstl1 in COPD could
thus trigger basal cell proliferation and basal cell hyperplasia which is commonly
Figure 2. The pro-inflammatory roles of Follistatin-like 1 in coordinating epithelial- observed in airway epithelial remodeling in COPD.
mesenchymal communication in inflammatory cytokine release in the lung. Overexpression
of Fstl1 in airway epithelium increases expression and release of inflammatory mediators,
including GM-CSF, RANTES, IL-6, and IL-8. In turn, these factors increases release of IL-6 and
IL-8 from pulmonary fibroblasts. For further details, see text.

150 151
In the developing lung, Fstl1 is expressed by goblet cells78 whereas in the healthy
adult lung, it are predominantly the ciliated cells that express Fstl1 (Chapter 3 and
5). In contrast, BMP4 is predominantly expressed by pulmonary mesenchymal cells,
such as lung fibroblasts and airway smooth muscle cells. Therefore it is tempting
to speculate that epithelial-mesenchymal communication via BMP signaling is an
important regulator of ciliated and goblet cell differentiation.79
Furthermore, we found that BMP-treated cells have increased expression of
notch-regulated genes, indicative of active notch signaling in adult human epithelial
differentiation (Chapter 3). Notch signaling determines the ultimate epithelial cell
fate in the airways. Therefore understanding the crosstalk between BMP and notch
in adult lung repair is important to explain the aberrant regeneration of remodeled
epithelium in COPD. In COPD, notch signaling components, e.g. Hey1 and Hey2,
are downregulated.70 Notch signaling promotes club cell fate at the expense of
ciliated cells as observed using conditional deletion of an enzyme responsible for
notch ligand-receptor binding, O-fucosyltransferase1 (Pofut1), or the transcriptional Figure 3. Variant club cell differentiation is driven by bone morphogenetic protein 4 in
effector of notch RBPjk or HES1 in mice.80–82 Guha and colleagues demonstrated that adult human airway epithelium: Implications for goblet cell metaplasia in COPD. Upon 14
the expression of the club cell marker secretoglobin 3A2 is driven by notch 1 activation days of air exposure, a cultured basal cell in air-liquid interface (ALI) system differentiates
into a pseudostratified epithelium composing of ciliated and mucus-producing goblet cells,
in vivo. They further proposed secretoglobin 3A2 expression and activation of notch which presumably derived from an intermediate club cell. Addition of BMP4 to the basal
signaling as early markers of the club cell differentiation program.62 In addition, it medium of ALI system every 48 hr suppresses the differentiation of goblet and ciliated cells,
was shown that Notch directly repressed MUC5AC transcription in lung epithelial whilst promotes the differentiation of variant club cells.
cells.81 Conversely, constitutive epithelial expression of notch 1 intracellular domain
(N1ICD) in developing mouse lung and notch activation in adult human airway
4. Future perspectives
epithelium triggers goblet cell metaplasia at the expense of ciliated cells.79
The data presented in this thesis shed light on previously unexplored roles for Fstl1/
Crosstalk between BMP and notch signaling is also observed during postnatal
BMP signaling in COPD and discuss the potential implications of increased expression
lung development. Endothelial deletion of Fstl1 in Fstl1-eKO mice was associated
of Fstl1 protein in airway inflammation and epithelial remodeling.
with increased BMP-regulated Jagged1 during postnatal lung development (Chapter
PAH is a common comorbidity in COPD.83,84 Our study provides in vivo evidence
2). In addition, notch downstream target genes, Hey1 and Hey3, were increased by
of a key role for endothelial Fstl1 in titrating the levels of BMP/Smad signaling during
BMP4 in the ALI cultured primary human bronchial epithelial cells. These observations
proper postnatal lung development (Chapter 2). However, the direct link and the
suggest that similar mechanisms of cell fate determination involving functional
precise functions of endothelial Fstl1 in pulmonary arterial smooth muscle cell
interactions between BMP and notch signaling is involved in adult lung cells and in
proliferation and in the regulation of actin remodeling in pulmonary arterial smooth
lung development. It would be interesting to characterize the composition of airway
muscle cells of lung neonates and COPD lungs remain to be elucidated. Uncontrolled
epithelium of Fstl1-eKO mice to examine whether variant club cell predominate Fstl1
proliferation of pulmonary arterial smooth muscle cells and impaired actin 8
deficient mouse epithelium. Additionally, future studies are necessary to address
remodeling contribute to vasoconstriction in PAH. Ex vivo studies using precision cut
whether inhibition of notch signaling using neutralizing antibodies can inhibit BMP-
lung slices may reveal the functional contribution of endothelial Fstl1 in pulmonary
induced variant club cell differentiation in the ALI culture system.
vascular contraction by comparing the vascular contractility of Fstl1-eKO with that of
To conclude, our studies demonstrate that BMP4 inhibits differentiation of
littermate controls. In addition, it is tempting to test the hypothesis that endothelial-
goblet cells whilst promoting differentiation of variant club cells in adult human
derived Fstl1 is a mediator of endothelial-mural cell communication in vascular tone
epithelium. Restoration of this imbalance in the lung of COPD patients may provide
regulation in the lung. An in vitro experiment using a co-culture system of human
a novel strategy to restore the observed decreased BMP-regulated Smad1/5 in the
lung microvascular endothelial cells and vascular smooth muscle cells could address
COPD lung and to reverse the loss of club cells whilst reducing goblet cell metaplasia
this question and could dissect the underlying mechanism by which Fstl1 regulates
and basal cell hyperplasia. Our study provokes future studies to address such a
actin expression in pulmonary vascular smooth muscle cells.
strategy in models of airway epithelial injury-repair in vitro and in vivo. The role of
BMP4 in adult human airway epithelial differentiation is illustrated in Figure 3.

152 153
In pulmonary vessels of COPD patients, we observed an increase in Fstl1 protein including proximal conducting airways and distal respiratory airways, to examine
expression in endothelial cells as well as vascular smooth muscle cells compared to whether there is an association between the relative expression (ratio) of BMP/Fstl1
individuals with normal lung function (Chapter 3). Although cell-specific expression and the epithelial cell composition in COPD airways.
of phosphorylated BMP-regulated Smad was not explored, we found that the To conclude, impaired activation of developmental Fstl1/BMP signaling following
BMP-regulated Smad activation was reduced in whole lung homogenates of COPD epithelial and vascular injury has been implicated in COPD thus it is important to
patients compared to non-COPD controls (Chapter 3). Activation of BMP signaling understand the functional role of Fstl1/BMP axis during lung development. This will
is reduced in idiopathic PAH patients,85–87 however, the contribution of Fstl1 to this lead to identify the abnormal tissue repair process in response to chronic lung injury
reduction was not studied. Previous studies demonstrated that another endogenous that can lead to the development of COPD.
BMP inhibitor, gremlin, was increased in endothelial cells of idiopathic and hypoxic-
induced PAH.88 Moreover, haploinsufficiency of gremlin 1 reduced pulmonary 5. Clinical and therapeutic relevance
vascular resistance by attenuating vascular remodeling,88 suggesting that BMP COPD is characterized by an irreversible obstruction of the airways and is associated
inhibition promotes vascular structural changes and leads to vasoconstriction.88 A with a progressive decline in lung function.90 Dysregulation of lung tissue repair as a
recent study shows that Fstl1 can antagonize BMP/Smad signaling whilst facilitating result of impaired activation of regenerative pathways following chronic lung injury
TGF-β-induced α-smooth muscle actin expression from lung fibroblasts thereby has been implicated in the development of COPD.91,92 A growing body of research
contributing to extracellular matrix remodeling and airway fibrosis.89 It could efforts has been focused on searching for a new drug target to improve current
probably be that Fstl1 increases α-smooth muscle actin production from vascular COPD treatment by targeting lung repair pathways. Thus, a better understanding
smooth muscle and promotes structural vascular changes and ultimately contributes of the developmental BMP/Fstl1 pathway in airway inflammation and epithelial
to increased vascular resistance and PAH. A future study evaluating the link between remodeling as parts of lung repair mechanisms will offer a novel molecular target
Fstl1 expression and pulmonary vascular function in patients with PAH secondary to for COPD therapy.
COPD would be valuable to perform. Persistent activation of innate immune responses through airway epithelium-
This thesis also explored the potential implications of increased Fstl1 in COPD, driven inflammation may contribute to chronic inflammation and contribute to
particularly in airway epithelium. A pro-inflammatory role of Fstl1, which mediates airway structural changes in COPD. Cigarette smoking increases the expression of
epithelial-mesenchymal communication in inflammatory cytokine release (Chapter Fstl1 protein even in the lung of non-COPD patients, which persists after smoking
4), may contribute to the innate immune response of the lung following environmental cessation (Chapter 3). Increased Fstl1 in airway epithelium of COPD stage II and IV
insults, e.g cigarette smoke. To provide insight into the potential pathways involved patients is associated with COPD and is not only associated with smoking (Chapter
in Fstl1-driven local airway inflammation in the lung, further studies are required to 3). Given the pro-inflammatory role of Fstl1 in respiratory innate immunity (Chapter
identify the receptor(s) by which Fstl1 exerts its function in airway epithelial cells as 5), targeting Fstl1 may offer a potential therapeutic value for COPD therapy. Blocking
well as in pulmonary mesenchymal cells. Fstl1 in the lung may repress GM-CSF and IL-6 release and limit airway-driven
Moreover, we demonstrate that BMP4 simultaneously inhibits differentiation of inflammation and subsequent influx of neutrophils and macrophages to the lung of
goblet cells while promoting differentiation of variant club cells (Chapter 5). Follow COPD patients.
up studies using mouse models of COPD are required to investigate the functional (Variant) club cells have anti-inflammatory and epithelial repair functions in the
effect of increased Fstl1 in airway epithelium to assess whether it antagonizes BMP lung.57,63 Loss of (variant) club cells has been linked to increased airway inflammation 8
signaling and contributes to goblet cell metaplasia, basal cell hyperplasia and loss of and COPD development.60,61,66,67 Restoration of club cell functions in COPD attenuates
variant club cells in in vitro and in vivo. Experiments using functional recombinant airway inflammation.60 The study presented in this thesis shows that BMP4 is able
Fstl1 and Fstl1 neutralizing antibodies would be valuable to examine the implication to promote variant club cell differentiation and suppress goblet cell and ciliated cell
of targeting Fstl1 in COPD. Given the postnatal lethality of Fstl1-eKO mice (Chapter differentiation (Chapter 7). Thereby, the loss of club cells in COPD epithelium60,61,66,67
2) that hinders further study in cigarette smoke-induced COPD mouse models, is likely to link with reduced BMP signaling in patients with COPD,73,74 presumably
follow up in vivo studies using doxycycline-induced epithelial-specific Fstl1-KO are by elevated expression of Fstl1 (Chapter 3). Reactivation of BMP signaling in
worth pursuing to evaluate the involvement of epithelial-derived Fstl1 in COPD airway epithelium may be a strategy worth pursuing to trigger differentiation and
development, including airway inflammation and epithelial cell differentiation. maintenance of variant club cells capable of airway epithelial repair as well as
Furthermore, it may be of interest to evaluate Fstl1 expression as well as BMP- suppressing the inflammatory response in the lung, whilst repressing goblet cell
regulated Smad phosphorylation in different parts of the tracheobronchial tree, metaplasia and basal cell hyperplasia in COPD.

154 155
6. Summary
The hypothetical model derived from this thesis is illustrated in Figure 4, depicting
BMP4
the potential implications of increased pulmonary expression of Fstl1 protein in
COPD airway epithelium. Collectively, our studies have revealed these following Chapter 7 goblet cell metaplasia loss of club cells
findings:
1. Endothelial Fstl1 is required for normal postnatal development of pulmonary
vasculature by modulating BMP/Smad signaling (Chapter 2).
2. Loss of endothelial Fstl1 is associated with an early expression of Endoglin, Chapter 3
Jagged1, Gata2 and the most potent vasoconstrictor endothelin-1, as well Fstl1

as with pulmonary small vessel remodeling, right ventricle hypertrophy and


right heart dysfunction. These abnormalities could cumulatively contribute Fstl1
to the observed respiratory distress in Fstl1-eKO mice (Chapter 2).
Chapter 5
3. The expression of Fstl1 protein is increased in the airway epithelium of COPD
GM-CSF
stage II and IV patients and is accompanied by decreased BMP-signaling RANTES
(Chapter 3). IL-6
IL-8
4. In comparison to the non-COPD never smoking group, ever smoking induces mesenchymal cells
a long lasting increase in the pulmonary expression of Fstl1 protein whilst
decreasing the pulmonary expression of BMP-regulated Smad1/5. This effect
IL-6
of ever smoking is present in non-COPD (ex-smokers and current smokers) IL-8
as well as in COPD stage II IV (all ex-smokers; Chapter 3).
5. Fstl1 promotes pro-inflammatory cytokine release (GM-CSF, RANTES, IL-6,
IL-8) from bronchial epithelial cells and coordinates epithelial-mesenchymal
communication, leading to subsequent pro-inflammatory cytokine release
(IL-6 and IL-8) from mesenchymal cells (Chapter 5).
6. BMP4 negatively regulates differentiation of goblet cells whilst promoting neutrophils macrophages
differentiation of variant club cells in adult bronchial epithelium. This
implies that the observed loss of club cells, goblet cell metaplasia and basal
cell hyperplasia in COPD could result from inhibition of BMP signaling due to
Airway inflammation
elevated levels of Fstl1 (Chapter 7).
Figure 4. The pathological implications of increased Fstl1 in airway epithelium of COPD
patients. The expression of Fstl1 protein is increased in the airway epithelium of both COPD
stage II and IV patients (Chapter 3). Given the importance role of Fstl1 in postnatal lung 8
development (Chapter 2) and increased Fstl1 protein expression in ever smoker non-COPD
and ex-smoker COPD lungs (Chapter 3), it could be that the Fstl1 protein is reactivated as part
of adult lung repair mechanism following epithelial injury, including cigarette smoking. Fstl1
is a pro-inflammatory cytokine mediating epithelial-mesenchymal communication in IL-6 and
IL-8 release (Chapter 5). Therefore, Fstl1 may contribute to the recruitment of neutrophils
and macrophages to the lungs. BMP4 inhibits differentiation of goblet cells whilst promoting
differentiation of variant club cells in adult human airway epithelium (Chapter 7). Increased
Fstl1 in COPD airway epithelium might drive epithelial differentiation towards goblet cell
hyperplasia and loss of club cells, which are the common hallmarks of remodeled COPD
airway epithelium. Fstl1 may inhibit the negative regulation of the BMP signaling in goblet
cells. Our findings provide a plausible connection between the loss of (variant) club cells and
goblet cell metaplasia (Chapter 7) with the observed reduction of BMP signaling in patients
with COPD stage II and IV (Chapter 3).

156 157
References congenital lung abnormalities. Birth Defects Res Part C Embryo Today Rev. 2014;102(4):343-358.
1. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal doi:10.1002/bdrc.21085.
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616. 21. International PPH Consortium, Lane KB, Machado RD, et al. Heterozygous germline mutations
2. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP) in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A. Genet. 2000;26(1):81-84. doi:10.1038/79226.
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108. 22. Machado RD, Aldred MA, James V, et al. Mutations of the TGF-beta type II receptor BMPR2 in
3. Umezu T, Yamanouchi H, Iida Y, Miura M, Tomooka Y. Follistatin-like-1, a diffusible mesenchymal pulmonary arterial hypertension. Hum Mutat. 2006;27(2):121-132. doi:10.1002/humu.20285.
factor determines the fate of epithelium. Proc Natl Acad Sci U S A. 2010;107(10):4601-4606. 23. Harrison RE, Flanagan JA, Sankelo M, et al. Molecular and functional analysis identifies ALK-
doi:10.1073/pnas.0909501107. 1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic
4. Hayakawa S, Ohashi K, Shibata R, et al. Cardiac Myocyte-Derived Follistatin-Like 1 Prevents telangiectasia. J Med Genet. 2003;40(12):865-871.
Renal Injury in a Subtotal Nephrectomy Model. J Am Soc Nephrol JASN. 2015;26(3):636-646. 24. Fujiwara M, Yagi H, Matsuoka R, et al. Implications of mutations of activin receptor-like kinase
doi:10.1681/ASN.2014020210. 1 gene (ALK1) in addition to bone morphogenetic protein receptor II gene (BMPR2) in children
5. Zhou X, Xiao X, Huang T, et al. Epigenetic inactivation of follistatin-like 1 mediates tumor immune with pulmonary arterial hypertension. Circ J Off J Jpn Circ Soc. 2008;72(1):127-133.
evasion in nasopharyngeal carcinoma. Oncotarget. 2016;7(13):16433-16444. doi:10.18632/ 25. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in
oncotarget.7654. hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(2):189-195. doi:10.1038/
6. Miyamae T, Marinov AD, Sowders D, et al. Follistatin-Like Protein-1 Is a Novel Proinflammatory ng0696-189.
Molecule. J Immunol. 2006;177(7):4758-4762. doi:10.4049/jimmunol.177.7.4758. 26. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-beta binding protein of endothelial
7. Clutter SD, Wilson DC, Marinov AD, Hirsch R. Follistatin-Like Protein 1 Promotes Arthritis by Up- cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8(4):345-
Regulating IFN-γ. J Immunol. 2009;182(1):234-239. doi:10.4049/jimmunol.182.1.234. 351. doi:10.1038/ng1294-345.
8. Chaly Y, Marinov AD, Oxburgh L, Bushnell DS, Hirsch R. FSTL1 promotes arthritis in mice by 27. Pi X, Lockyer P, Dyer LA, et al. Bmper inhibits endothelial expression of inflammatory adhesion
enhancing inflammatory cytokine/chemokine expression. Arthritis Rheum. 2012;64(4):1082- molecules and protects against atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32(9):2214-
1088. doi:10.1002/art.33422. 2222. doi:10.1161/ATVBAHA.112.252015.
9. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary 28. Yao Y, Yao J, Radparvar M, et al. Reducing Jagged 1 and 2 levels prevents cerebral arteriovenous
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878. malformations in matrix Gla protein deficiency. Proc Natl Acad Sci U S A. 2013;110(47):19071-
10. Liu M, Liu R, Wu H, et al. Radix puerariae extracts ameliorate paraquat-induced pulmonary 19076. doi:10.1073/pnas.1310905110.
fibrosis by attenuating follistatin-like 1 and nuclear factor erythroid 2p45-related factor-2 29. Jain RK. Molecular regulation of vessel maturation. Nat Med. 2003;9(6):685-693. doi:10.1038/
signalling pathways through downregulation of miRNA-21 expression. BMC Complement Altern nm0603-685.
Med. 2016;16:11. doi:10.1186/s12906-016-0991-6. 30. Schiffrin EL. Endothelin and endothelin antagonists in hypertension. J Hypertens. 1998;16(12 Pt
11. Murphy N, Gaynor KU, Rowan SC, et al. Altered Expression of Bone Morphogenetic Protein 2):1891-1895.
Accessory Proteins in Murine and Human Pulmonary Fibrosis. Am J Pathol. 2016;186(3):600- 31. Lee SH, Channick RN. Endothelin antagonism in pulmonary arterial hypertension. Semin Respir
615. doi:10.1016/j.ajpath.2015.10.032. Crit Care Med. 2005;26(4):402-408. doi:10.1055/s-2005-916155.
12. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by Inducing 32. Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular
Oncostatin M. J Immunol Baltim Md 1950. September 2015. doi:10.4049/jimmunol.1501105. Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle
13. Miller M, Esnault S, Kurten RC, et al. Segmental allergen challenge increases levels of airway Disorders. Pharmacol Rev. 2016;68(2):476-532. doi:10.1124/pr.115.010652.
follistatin-like 1 in patients with asthma. J Allergy Clin Immunol. 2016;138(2):596-599.e4. 33. Wilson JL, Yu J, Taylor L, Polgar P. Hyperplastic Growth of Pulmonary Artery Smooth Muscle Cells
doi:10.1016/j.jaci.2016.01.019. from Subjects with Pulmonary Arterial Hypertension Is Activated through JNK and p38 MAPK.
14. Gangopahyay A, Oran M, Bauer EM, et al. Bone morphogenetic protein receptor II is a novel PLoS ONE. 2015;10(4). doi:10.1371/journal.pone.0123662.
mediator of endothelial nitric-oxide synthase activation. J Biol Chem. 2011;286(38):33134- 34. Noordegraaf AV, Galiè N. The role of the right ventricle in pulmonary arterial hypertension. Eur
33140. doi:10.1074/jbc.M111.274100. Respir Rev. 2011;20(122):243-253. doi:10.1183/09059180.00006511.
15. Xu J, Zhu D, Sonoda S, et al. Over-expression of BMP4 inhibits experimental choroidal 35. NAEIJE R, CHESLER N. PULMONARY CIRCULATION AT EXERCISE. Compr Physiol. 2012;2(1):711-
neovascularization by modulating VEGF and MMP-9. Angiogenesis. 2012;15(2):213-227. 741. doi:10.1002/cphy.c100091. 8
doi:10.1007/s10456-012-9254-4. 36. Knight D. Epithelium–fibroblast interactions in response to airway inflammation. Immunol Cell
16. Moreno-Miralles I, Ren R, Moser M, Hartnett ME, Patterson C. Bone morphogenetic protein Biol. 2001;79(2):160-164. doi:10.1046/j.1440-1711.2001.00988.x.
endothelial cell precursor-derived regulator regulates retinal angiogenesis in vivo in a mouse 37. Osei ET, Brandsma C-A, Noordhoek JA, Timens W, Postma D, Heijink I. Crosstalk between
model of oxygen-induced retinopathy. Arterioscler Thromb Vasc Biol. 2011;31(10):2216-2222. epithelium and fibroblasts; implications for COPD. Eur Respir J. 2014;44(Suppl 58):P3899.
doi:10.1161/ATVBAHA.111.230235. 38. Ouchi N, Oshima Y, Ohashi K, et al. Follistatin-like 1, a secreted muscle protein, promotes
17. Helbing T, Rothweiler R, Ketterer E, et al. BMP activity controlled by BMPER regulates the endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase-
proinflammatory phenotype of endothelium. Blood. 2011;118(18):5040-5049. doi:10.1182/ dependent mechanism. J Biol Chem. 2008;283(47):32802-32811. doi:10.1074/jbc.M803440200.
blood-2011-03-339762. 39. Shibanuma M, Mashimo J, Mita A, Kuroki T, Nose K. Cloning from a mouse osteoblastic cell line
18. Yao Y, Jumabay M, Wang A, Boström KI. Matrix Gla protein deficiency causes arteriovenous of a set of transforming-growth-factor-beta 1-regulated genes, one of which seems to encode a
malformations in mice. J Clin Invest. 2011;121(8):2993-3004. doi:10.1172/JCI57567. follistatin-related polypeptide. Eur J Biochem FEBS. 1993;217(1):13-19.
19. Suzuki Y, Ohga N, Morishita Y, Hida K, Miyazono K, Watabe T. BMP-9 induces proliferation of 40. Wilson DC, Marinov AD, Blair HC, et al. Follistatin-like protein 1 is a mesenchyme-derived
multiple types of endothelial cells in vitro and in vivo. J Cell Sci. 2010;123(Pt 10):1684-1692. inflammatory protein and may represent a biomarker for systemic-onset juvenile rheumatoid
doi:10.1242/jcs.061556. arthritis. Arthritis Rheum. 2010;62(8):2510-2516. doi:10.1002/art.27485.
20. Kool H, Mous D, Tibboel D, de Klein A, Rottier RJ. Pulmonary vascular development goes awry in 41. Morrisey EE, Hogan BLM. Preparing for the first breath: genetic and cellular mechanisms in lung

158 159
development. Dev Cell. 2010;18(1):8-23. doi:10.1016/j.devcel.2009.12.010. for a distinct subset of Clara-like precursors in the developing airways. Proc Natl Acad Sci.
42. Murakami K, Tanaka M, Usui T, et al. Follistatin-related protein/follistatin-like 1 evokes an 2012;109(31):12592-12597. doi:10.1073/pnas.1204710109.
innate immune response via CD14 and toll-like receptor 4. FEBS Lett. 2012;586(4):319-324. 63. Barnes PJ. CLub cells, their secretory protein, and copd. Chest. 2015;147(6):1447-1448.
doi:10.1016/j.febslet.2012.01.010. doi:10.1378/chest.14-3171.
43. Fan N, Sun H, Wang Y, et al. Follistatin-Like 1: A Potential Mediator of Inflammation in Obesity. 64. Hung KS. Development of neuroepithelial bodies in pre- and postnatal mouse lungs: scanning
Mediators Inflamm. 2013;2013:e752519. doi:10.1155/2013/752519. electron microscopic study. Anat Rec. 1982;203(2):285-291. doi:10.1002/ar.1092030210.
44. Bellusci S, Henderson R, Winnier G, Oikawa T, Hogan BL. Evidence from normal expression 65. Reynolds SD, Giangreco A, Power JH, Stripp BR. Neuroepithelial bodies of pulmonary airways
and targeted misexpression that bone morphogenetic protein (Bmp-4) plays a role in mouse serve as a reservoir of progenitor cells capable of epithelial regeneration. Am J Pathol.
embryonic lung morphogenesis. Dev Camb Engl. 1996;122(6):1693-1702. 2000;156(1):269-278. doi:10.1016/S0002-9440(10)64727-X.
45. Weaver M, Yingling JM, Dunn NR, Bellusci S, Hogan BL. Bmp signaling regulates proximal-distal 66. Shijubo N, Itoh Y, Yamaguchi T, et al. Clara cell protein-positive epithelial cells are reduced in
differentiation of endoderm in mouse lung development. Dev Camb Engl. 1999;126(18):4005- small airways of asthmatics. Am J Respir Crit Care Med. 1999;160(3):930-933. doi:10.1164/
4015. ajrccm.160.3.9803113.
46. Sountoulidis A, Stavropoulos A, Giaglis S, et al. Activation of the canonical bone morphogenetic 67. Rokicki W, Rokicki M, Wojtacha J, Dżeljijli A. The role and importance of club cells (Clara cells) in
protein (BMP) pathway during lung morphogenesis and adult lung tissue repair. PloS One. the pathogenesis of some respiratory diseases. Kardiochirurgia Torakochirurgia Pol Pol J Cardio-
2012;7(8):e41460. doi:10.1371/journal.pone.0041460. Thorac Surg. 2016;13(1):26-30. doi:10.5114/kitp.2016.58961.
47. Ninomiya N, Michiue T, Asashima M, Kurisaki A. BMP signaling regulates the differentiation 68. Bernard A, Marchandise FX, Depelchin S, Lauwerys R, Sibille Y. Clara cell protein in serum and
of mouse embryonic stem cells into lung epithelial cell lineages. In Vitro Cell Dev Biol Anim. bronchoalveolar lavage. Eur Respir J. 1992;5(10):1231-1238.
2013;49(3):230-237. doi:10.1007/s11626-013-9589-1. 69. Vestbo J, Edwards LD, Scanlon PD, et al. Changes in Forced Expiratory Volume in 1 Second over
48. Huang SXL, Islam MN, O’Neill J, et al. Highly efficient generation of airway and lung epithelial cells Time in COPD. N Engl J Med. 2011;365(13):1184-1192. doi:10.1056/NEJMoa1105482.
from human pluripotent stem cells. Nat Biotechnol. 2014;32(1):84-91. doi:10.1038/nbt.2754. 70. Park HY, Churg A, Wright JL, et al. Club cell protein 16 and disease progression in chronic
49. Huang SXL, Green MD, de Carvalho AT, et al. The in vitro generation of lung and airway progenitor obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(12):1413-1419.
cells from human pluripotent stem cells. Nat Protoc. 2015;10(3):413-425. doi:10.1038/ doi:10.1164/rccm.201305-0892OC.
nprot.2015.023. 71. Lomas DA, Silverman EK, Edwards LD, et al. Evaluation of serum CC-16 as a biomarker for COPD
50. Ross AJ, Dailey LA, Brighton LE, Devlin RB. Transcriptional profiling of mucociliary differentiation in the ECLIPSE cohort. Thorax. 2008;63(12):1058-1063. doi:10.1136/thx.2008.102574.
in human airway epithelial cells. Am J Respir Cell Mol Biol. 2007;37(2):169-185. doi:10.1165/ 72. Guerra S, Halonen M, Vasquez MM, et al. The relation of circulating CC16 to lung function growth,
rcmb.2006-0466OC. decline, and development of COPD across the lifespan. Lancet Respir Med. 2015;3(8):613-620.
51. Fahy JV, Dickey BF. Airway Mucus Function and Dysfunction. N Engl J Med. 2010;363(23):2233- doi:10.1016/S2213-2600(15)00196-4.
2247. doi:10.1056/NEJMra0910061. 73. Myllärniemi M, Lindholm P, Ryynänen MJ, et al. Gremlin-mediated decrease in bone
52. Li F, He J, Wei J, Cho WC, Liu X. Diversity of Epithelial Stem Cell Types in Adult Lung. Stem Cells morphogenetic protein signaling promotes pulmonary fibrosis. Am J Respir Crit Care Med.
Int. 2015;2015:e728307. doi:10.1155/2015/728307. 2008;177(3):321-329. doi:10.1164/rccm.200706-945OC.
53. Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. Basal cells are a multipotent progenitor 74. De Langhe E, Cailotto F, De Vooght V, et al. Enhanced endogenous bone morphogenetic protein
capable of renewing the bronchial epithelium. Am J Pathol. 2004;164(2):577-588. doi:10.1016/ signaling protects against bleomycin induced pulmonary fibrosis. Respir Res. 2015;16:38.
S0002-9440(10)63147-1. doi:10.1186/s12931-015-0202-x.
54. Rock JR, Onaitis MW, Rawlins EL, et al. Basal cells as stem cells of the mouse trachea and 75. Cibois M, Luxardi G, Chevalier B, et al. BMP signalling controls the construction of vertebrate
human airway epithelium. Proc Natl Acad Sci U S A. 2009;106(31):12771-12775. doi:10.1073/ mucociliary epithelia. Development. 2015;142(13):2352-2363. doi:10.1242/dev.118679.
pnas.0906850106. 76. Tadokoro T, Gao X, Hong CC, Hotten D, Hogan BLM. BMP signaling and cellular dynamics during
55. Kimura J, Deutsch GH. Key mechanisms of early lung development. Pediatr Dev Pathol Off J Soc regeneration of airway epithelium from basal progenitors. Dev Camb Engl. 2016;143(5):764-
Pediatr Pathol Paediatr Pathol Soc. 2007;10(5):335-347. doi:10.2350/07-06-0290.1. 773. doi:10.1242/dev.126656.
56. Rawlins EL, Hogan BLM. Epithelial stem cells of the lung: privileged few or opportunities for 77. Mou H, Vinarsky V, Tata PR, et al. Dual SMAD Signaling Inhibition Enables Long-Term
many? Dev Camb Engl. 2006;133(13):2455-2465. doi:10.1242/dev.02407. Expansion of Diverse Epithelial Basal Cells. Cell Stem Cell. 2016;19(2):217-231. doi:10.1016/j.
57. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell stem.2016.05.012. 8
populations. Nat Med. 2014;20(8):822-832. doi:10.1038/nm.3642. 78. Adams D, Larman B, Oxburgh L. Developmental expression of mouse Follistatin-like 1 (Fstl1):
58. Schamberger AC, Staab-Weijnitz CA, Mise-Racek N, Eickelberg O. Cigarette smoke alters primary Dynamic regulation during organogenesis of the kidney and lung. Gene Expr Patterns.
human bronchial epithelial cell differentiation at the air-liquid interface. Sci Rep. 2015;5. 2007;7(4):491-500. doi:10.1016/j.modgep.2006.10.009.
doi:10.1038/srep08163. 79. Guseh JS, Bores SA, Stanger BZ, et al. Notch signaling promotes airway mucous metaplasia
59. Saetta M, Turato G, Baraldo S, et al. Goblet cell hyperplasia and epithelial inflammation and inhibits alveolar development. Development. 2009;136(10):1751-1759. doi:10.1242/
in peripheral airways of smokers with both symptoms of chronic bronchitis and chronic dev.029249.
airflow limitation. Am J Respir Crit Care Med. 2000;161(3 Pt 1):1016-1021. doi:10.1164/ 80. Lafkas D, Shelton A, Chiu C, et al. Therapeutic antibodies reveal Notch control of transdifferentiation
ajrccm.161.3.9907080. in the adult lung. Nature. 2015;528(7580):127-131. doi:10.1038/nature15715.
60. Gamez AS, Gras D, Petit A, et al. Supplementing defect in club cell secretory protein attenuates 81. Tsao P-N, Wei S-C, Wu M-F, et al. Notch signaling prevents mucous metaplasia in mouse
airway inflammation in COPD. Chest. 2015;147(6):1467-1476. doi:10.1378/chest.14-1174. conducting airways during postnatal development. Development. 2011;138(16):3533-3543.
61. Pilette C, Godding V, Kiss R, et al. Reduced epithelial expression of secretory component in small doi:10.1242/dev.063727.
airways correlates with airflow obstruction in chronic obstructive pulmonary disease. Am J 82. Ito T, Udaka N, Yazawa T, et al. Basic helix-loop-helix transcription factors regulate the
Respir Crit Care Med. 2001;163(1):185-194. doi:10.1164/ajrccm.163.1.9912137. neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development.
62. Guha A, Vasconcelos M, Cai Y, et al. Neuroepithelial body microenvironment is a niche 2000;127(18):3913-3921.

160 161
83. Siafakas NM, Antoniou KM, Tzortzaki EG. Role of angiogenesis and vascular remodeling in
chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2007;2(4):453-462.
84. Hashimoto M, Tanaka H, Abe S. Quantitative analysis of bronchial wall vascularity in the medium
and small airways of patients with asthma and COPD. Chest. 2005;127(3):965-972. doi:10.1378/
chest.127.3.965.
85. Morty RE, Nejman B, Kwapiszewska G, et al. Dysregulated bone morphogenetic protein signaling
in monocrotaline-induced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol.
2007;27(5):1072-1078. doi:10.1161/ATVBAHA.107.141200.
86. Takahashi K, Kogaki S, Matsushita T, Nasuno S, Kurotobi S, Ozono K. Hypoxia induces alteration
of bone morphogenetic protein receptor signaling in pulmonary artery endothelial cell. Pediatr
Res. 2007;61(4):392-397. doi:10.1203/pdr.0b013e3180332cba.
87. Long L, Crosby A, Yang X, et al. Altered Bone Morphogenetic Protein and Transforming Growth
Factor-β Signaling in Rat Models of Pulmonary Hypertension. Circulation. 2009;119(4):566-576.
doi:10.1161/CIRCULATIONAHA.108.821504.
88. Cahill E, Rowan SC, Sands M, et al. The pathophysiological basis of chronic hypoxic pulmonary
hypertension in the mouse: vasoconstrictor and structural mechanisms contribute equally. Exp
Physiol. 2012;97(6):796-806. doi:10.1113/expphysiol.2012.065474.
89. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878.
90. Vestbo J, Hurd SS, Agustí AG, et al. Global Strategy for the Diagnosis, Management, and Prevention
of Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2013;187(4):347-365.
doi:10.1164/rccm.201204-0596PP.
91. Shi W, Chen F, Cardoso WV. Mechanisms of lung development: contribution to adult lung disease
and relevance to chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2009;6(7):558-
563. doi:10.1513/pats.200905-031RM.
92. Boucherat O, Morissette MC, Provencher S, Bonnet S, Maltais F. Bridging Lung Development
with Chronic Obstructive Pulmonary Disease. Relevance of Developmental Pathways in Chronic
Obstructive Pulmonary Disease Pathogenesis. Am J Respir Crit Care Med. 2015;193(4):362-375.
doi:10.1164/rccm.201508-1518PP.

162 163
CHAPTER 9 1

DUTCH SUMMARY/
NEDERLANDSE
SAMENVATTING

165
Voorwoord

Chronisch obstructief longlijden (chronic obstructive pulmonary disease, COPD)


wordt gekenmerkt door een progressieve en irreversibele luchtwegobstructie waarin
buitensporige en hardnekkige ontsteking van de longen een belangrijke rol speelt.
Zoals aangegeven in de algemene inleiding (Hoofdstuk 1), is een juiste balans tussen
het zogenaamde Bone morphogenetic protein (BMP4) en zijn endogene remme
Follistatin-like 1 (Fstl1) van cruciaal belang voor goede biologische functies tijdens
normale embryonale long-morfogenese1,2 en in de volwassen long in rusttoestand.3–5
Daarnaast is een immuunmodulerende functie van Fstl1 aangetoond in verschillende
ontstekingsziekten.6–8 De functionele rol van Fstl1 in de postnatale ontwikkeling van
de longen en in de longen van COPD patiënten is echter nog niet eerder onderzocht.
Onlangs heeft een aantal studies de betrokkenheid van Fstl1 bij longfibrose en
ernstig allergisch astma aangetoond.9–13 De studies beschreven in dit proefschrift
laten zien dat Fstl1 vereist is voor de pulmonale vasculaire ontwikkeling na de
geboorte (Hoofdstuk 2). Bovendien tonen wij aan dat Fstl1 eiwitexpressie verhoogd
is in het epitheel van de luchtwegen van COPD patiënten (Hoofdstuk 3). We vonden
tevens dat Fstl1 een ontstekingsbevorderende factor in de luchtwegen is (Hoofdstuk
5). Verder vonden we dat BMP4 signalering de distale variant-clubceldifferentiatie
van het epitheel in de luchtwegen reguleert (Hoofdstuk 7). In zijn geheel laten
onze bevindingen aannemen dat Fstl1 een cruciale rol speelt in de ontwikkeling en
achteruitgang van COPD.

1. De rol van Fstl1 in luchtwegontsteking bij COPD patiënten


Figuur 1. De rol van endotheliaal Follistatin-like 1 bij het moduleren van BMP/Smad
De ontwikkeling van de long wordt nauwkeurig aangestuurd door meerdere signalering tijdens de postnatale ontwikkeling van de longvaten. In aanwezigheid van
signaaltransductieroutes, die de vorming van luchtwegen en vasculatuur reguleren. endotheliaal Fstl1 worden de BMP-geïnduceerde fosforylering van Smad1/5 en daarop
BMP signalering is essentieel in het dynamische proces van vaatgroei en rijping, volgende downstream targetgenen geremd. Deletie van endotheliaal Fstl1 in Fstl1-eKO
muizen is geassocieerd met over-activatie van door BMP gefosforyleerde Smad1/5 en de
door het reguleren van de proliferatie en migratie van vasculaire gladde spieren en targetgenen endogline, JAG1 en GATA2.
endotheelcellen.14–19 Sterk gecoördineerde regulatie van BMP signalering is essentieel
voor een goede longontwikkeling.20 Verscheidene genetische vaatziekten zijn in
verband gebracht met afwijkende BMP signalering, waaronder familiaire pulmonale netwerk moet worden gecoördineerd om de blootstelling van bloed aan zuurstof te
arteriële hypertensie (PAH).18,21–28 Deze centrale rol voor BMP signalering wordt optimaliseren.20 De vorming van het pulmonaire vasculaire netwerk bleek dan ook
ondersteund door de bevinding dat muizen waarbij het Fstl1 gen is uitgeschakeld verstoord in Fstl1-eKO muizen, leidend tot ademnood (Hoofdstuk 2).
(Fstl1-KO muizen) kleinere longen met verdikte alveolaire septa hebben. Deze muizen Hoewel minieme pulmonaire vasculaire remodeling in de grote en elastische
hebben last van ademnood en sterven vrijwel direct na de geboorte,1,2 wat aangeeft arteriën in Fstl1-eKO muizen werd waargenomen, vonden we met name verhoogde
dat Fstl1 nodig is voor longontwikkeling en alveolarisatie. Opvallend genoeg vonden percentages van actine bevattende kleine longvaten 21 dagen na de geboorte in 9
we dat muizen met endotheel-specifieke Fstl1 deletie (Fstl1-eKO) ook last hadden deze muizen (Hoofdstuk 2). Bovendien vonden we dat de cardiale output van het
van ademnood en 3 weken na de geboorte stierven (Hoofdstuk 2). De postnatale rechterventrikel was verlaagd in Fstl1-eKO muizen (Hoofdstuk 2). Verder onderzoek
sterfte van de Fstl1-eKO muizen suggereert een belangrijke rol voor endotheliaal naar de onderliggende mechanismen toonde aan dat gefosforyleerd Smad1/8/5
Fstl1 in postnatale ontwikkeling van de pulmonaire vasculatuur. Fstl1-eKO muizen - een aanwijzing voor actieve BMP signalering - en BMP-gereguleerde eiwitten
stierven gemiddeld 21 dagen na de geboorte, wat in het alveolaire stadium van (Jagged1, endogline, en endotheline-1) verhoogd waren tijdens het vroege stadium
de longontwikkeling is dat van cruciaal belang is voor de alveolaire en vasculaire van longontwikkeling (7 dagen na de geboorte) (Hoofdstuk 2). De remodeling van de
netwerkvorming en rijping.20,29 Vertakking van de luchtwegen en het vasculaire longvaatjes in de Fstl1-eKO muizen ging gepaard met een verhoogde eiwitexpressie

166 167
van endotheline-1, hetgeen een sterke vaatvernauwende werking heeft, na zowel 7 (Hoofdstuk 3). Nader onderzoek aan longweefsel van COPD-patiënten toonde
en 21 dagen na de geboorte. (Hoofdstuk 2). De combinatie van een hoge endotheline- aan dat het Fstl1 eiwit tot expressie werd gebracht in verschillende delen van het
1-expressie en een hoog percentage van actine bevattende kleine longvaten zou longweefsel, waaronder luchtwegepitheel, endotheel, vasculaire gladde spiercellen,
kunnen leiden tot een verhoging van de pulmonale vasculaire weerstand (Hoofdstuk en ontstekingscellen (Hoofdstuk 3). De intensiteit van Fstl1 kleuring, gebruikt
2). De progressieve en onomkeerbare defecten in longvatstructuur en-functie in als indicator voor de mate van aanwezigheid van het Fstl1 eiwit, was hoger in
Fstl1-eKO muizen zouden de ademnood kunnen verklaren, omdat de niet adequaat luchtwegepitheel, endotheel, vasculaire gladde spierbundels en ontstekingscellen
gevormde kleine longvaten niet in staat zijn om in de juiste mate bloed naar de van zowel patienten met stadium II en IV COPD (allen ex-rokers) in vergelijking
longen te brengen. Dit zou kunnen leiden tot een gebrek aan zuurstoftoevoer naar met personen zonder luchtwegobstructie (Hoofdstuk 3). Dit kan erop wijzen dat
de vitale organen en uiteindelijk tot de dood. Het zou dan ook interessant zijn om de verhoogde Fstl1 expressie geassocieerd is met COPD. Dit proefschrift is gericht op
zuurstof- en kooldioxideniveaus in het bloed te onderzoeken, en daarnaast ook de de mogelijke gevolgen van deze toegenomen Fstl1 expressie en de rol hiervan in de
diameter van de kleine longvaten en hoeveelheid spiermassa in de kleine longvaten pathogenese van COPD (Hoofdstuk 5 en 7). Intrigerend is dat de expressie van Fstl1
te bepalen in follow-up studies. in longhomogenaten van rokers zonder COPD en ex-rokers ook hoger is in vergelijking
Samengevat lijkt het dat Fstl1-eKO muizen gekenmerkt worden door tijdelijke met niet-rokers zonder COPD, hetgeen wijst op een effect van het ooit-hebben-
moleculaire veranderingen in BMP signalering, die vroeg beginnen tijdens de gerookt, waarschijnlijk door rook-geïnduceerde ontsteking (Hoofdstuk 3). Het zou
postnatale ontwikkeling (7 dagen na de geboorte), terwijl de fysiologische functies kunnen dat acute vrijzetting van ontstekingsmediatoren en de daaropvolgende
en morfologische gevolgen hiervan zich in latere stadia van postnatale ontwikkeling lokale ontsteking waarbij ontstekingscellen worden gerekruteerd in de longen
(21 dagen na de geboorte) manifesteren. Het is echter ook mogelijk dat de wordt bevorderd door sigarettenrook, wat blijft bestaan ​​na het stoppen met roken.
moleculaire en morfologische veranderingen in de longen een gevolg zijn van een Aangezien Fstl1 een TGF-β1 induceerbare factor is en TGF-β1 afgifte door bronchiale
slecht functionerend rechter ventrikel. Als gevolg van hypoxie die aldus ontstaat epitheelcellen wordt verhoogd na sigarettenrook behandeling,33 suggereert dit
kan remodeling van de longvaten en een toegenomen endotheline-1-expressie dat TGF-β1 de waargenomen up-regulatie van Fstl1 kan reguleren als reactie op
optreden. Toekomstige studies zouden de cardiovasculaire structuur en functies in sigarettenrook.
Fstl1-eKO muizen kunnen bestuderen om te onderzoeken wat de functie is van Fstl1 Vervolgens hebben we de mogelijke rol van Fstl1 in longcellen onderzocht in het
in het cardiovasculaire systeem. De rol van Fstl1 bij het moduleren van BMP/Smad reguleren van de lokale ontsteking (Hoofdstuk 5). Middels studies in verschillende
signalering tijdens de postnatale ontwikkeling van de longvaten wordt weergegeven cellijnen bleek dat bronchiale epitheelcellen het meeste Fstl1 tot expressie brengen
in Figuur 1. en uitscheiden in vergelijking met mesenchymale cellen, zoals longfibroblasten en
luchtweggladde spiercellen (Hoofdstuk 5). Dit komt overeen met onze waarneming
2. De rol van Fstl1 in ontsteking van de luchtwegen bij COPD patiënten in longweefsel van personen met een normale longfunctie dat Fstl1voornamelijk
Eén van de kenmerken van COPD is een inadequate activering van het aangeboren tot expressie wordt gebracht in epitheelcellen, met name in het apicale deel
immuunsysteem en abnormaal weefselherstel in reactie op langdurige blootstelling van trilhaarcellen en in mindere mate in gladde spiercellen in de luchtwegen
aan pneumotoxische, geïnhaleerde stoffen, zoals sigarettenrook. Op zijn beurt leidt (Hoofdstuk 3). Andere studies in het hart en de gewrichten suggereren dat Fstl1 een
dit tot epitheliale remodeling en fysiologische veranderingen die bijdragen aan mesenchymaal uitgescheiden factor is.32,34
de niet volledig reversibele luchtwegvernauwing.30,31 Als eerste barrière tegen het Gezien het feit dat Fstl1 bekend staat als een endogene antagonist van BMP
externe milieu scheidt luchtwegepitheel ontstekingsmediatoren uit met paracriene signalering in de longen,1,2 hebben wij de fosforylering van BMP-specifieke Smad1/5
effecten op naburige mesenchymale cellen, waaronder longfibroblasten en gladde onderzocht in longhomogenaten van COPD-patiënten (allen ex-rokers) in vergelijking
spiercellen van de luchtwegen. Interessant is dat een rol voor Fstl1 in cel-cel met personen zonder COPD met verschillend rookgedrag. Gefosforyleerd Smad1/5
communicatie al eerder is gerapporteerd.6,9 Een groeiende hoeveelheid bewijs geeft bleek aanzienlijk verminderd in longhomogenaten van rokers zonder COPD, ex-
rokers zonder COPD en COPD stadium II en stadium IV patiënte (allen ex-rokers) 9
aan dat Fstl1 een pro-inflammatoire rol heeft in het reguleren van de inflammatoire
cytokine secretie in de pathogenese van verschillende ontstekingsziekten en in de in vergelijking met mensen zonder COPD die nooit gerookt hebben (Hoofdstuk
verergering van de symptomen van deze ziekten.6,7,32 Het verband tussen Fstl1 en 3). Deze verminderde Smad1/5 fosforylering kan verklaard worden door de
COPD pathogenese is echter nog niet eerder onderzocht. verhoogde expressie van Fstl1, maar ook door verminderde pulmonaire BMP4
Onze studies naar de expressie van Fstl1 in COPD hebben aangetoond dat Fstl1 expressie. We vonden in de longhomogenaten echter geen significante verschillen
sterk verhoogd is in longhomogenaten van rokers zonder COPD en van stadium II in BMP4 eiwitgehalten tussen de groepen (Hoofdstuk 3). In cellijnen wordt BMP4
als IV COPD-patiënten in vergelijking met personen zonder luchtwegobstructie hoofdzakelijk tot expressie gebracht in mesenchymale cellen, zoals longfibroblasten
en luchtweggladde spiercellen, terwijl humane bronchiale epitheelcellen de laagste

168 169
expressie en uitscheiding van BMP4 hebben. De fosforylering van Smad1/5 in de tijdsafhankelijk was upgereguleerd in een transcriptieprofielbepaling.44
cellijnen volgde dit patroon van BMP ligand expressie. De verschillende cellulaire Het luchtwegepitheel is de eerste beschermende fysische en functionele
bronnen van liganden en hun remmers in de longen wijzen op een mogelijke barrière van de luchtwegen en longen tegen de externe omgeving. Het epitheel van
crosstalk tussen epitheliale en mesenchymale cellen in het aansturen van biologische luchtwegen in volwassen bestaat uit een gemengd evenwicht van slijmbekercellen,
functies, met inbegrip van de regulering van inflammatoire cytokine afgifte. Door trilhaarcellen, basale cellen en clubcellen die zijn verdeeld over de tracheobronchiale
deze bevindingen hebben we besloten om te onderzoeken wat de precieze rol boom.45 Slijmuitscheidende slijmbekercellen worden voornamelijk tot expressie
van Fstl1 in luchtwegontsteking in het algemeen, en de mogelijke bijdrage aan de gebracht in het proximale epithelium, samen met de basale cellen, club cellen
ontwikkeling van COPD in het bijzonder (Hoofdstuk 5). en trilhaarcellen.46,47 In de distale, kleine bronchioli zijn er secretoire club cellen,
De waargenomen toegenomen Fstl1 expressie in het luchtwegepitheel trilhaarcellen, en pulmonale neuro-endocriene cellen. Respiratoire longblaasjes zijn
patiënten met COPD werd nagebootst door Fstl1 tot overexpressie te brengen bedekt met een dunne, enkele laag van platte type I en type II kubische alveolaire
in epitheelcellen, en het effect hiervan op pro-inflammatoire cytokine afgifte epitheelcellen.48–50
van pulmonale mesenchymale cellen te bestuderen. Overexpressie van Fstl1 in Sigarettenrook verandert de samenstelling van luchtwegepitheel en draagt ​​bij
bronchiale epitheelcellen vergrootte de afgrifte van de ontstekingsfactoren IL-6 aan epitheliale remodeling bij COPD.51 Luchtwegepitheelremodeling bij COPD wordt
en GM-CSF wat duidt op een pro-inflammatoire functie (Hoofdstuk 5). Celmedium gekenmerkt door metaplasie van slijmbekercellen en basale cellen, en door verlies van
verkregen van bronchiale epitheelcellen waarin Fstl1 tot overexpressie was gebracht functionele clubcellen in de perifere en centrale luchtwegen.52–54 Variant clubcellen
(Fstl1-CM) werd gebruikt om mesenchymale cellen mee te behandelen. Interessant (voorheen claracellen genoemd) zijn secretoire cellen die zich voornamelijk bevinden
genoeg ontdekten we dat dit Fstl1-CM de IL-6 en IL-8-transcriptie en eiwit afgifte in de distale luchtwegen en een beschermende rol hebben door het afscheiden van
door mesenchymale cellen verhoogde, wat suggereert dat Fstl1 pro-inflammatoire uteroglobine, secretoglobine en surfactant, welke ontstekingsremmende functies
cytokine afgifte in de longen zou kunnen reguleren. (Hoofdstuk 5). Hoewel eerdere hebben en belangrijk zijn voor epitheliale regeneratie.55 Variant clubcellen hebben
studies meldden dat Fstl1 de directe afgifte van IL-6 en IL-8 uit mesenchymale stamcelachtige eigenschappen en zijn in staat tot zelfvernieuwing en differentiatie
cellen bevordert,6,8,35,36 vertonen onze gegevens met behulp van recombinante Fstl1 tot trilhaarcellen en slijmbekercellen.50,56 Variant clubcellen hebben een duidelijke
behandeling en overexpressie en knockdown van Fstl1 in longfibroblasten geen afgeplatte morfologie en worden gekenmerkt door een afwezigheid van CC10 en
enkel effect op het basale niveau van IL-1β geïnduceerde IL-6 en IL-8-eiwit afgifte, cytochroom p450 Cyp2f. Normale clubcellen vertonen een typisch kubusvormig
wat suggereert dat Fstl1 op zichzelf onvoldoende is om cytokine afgifte te induceren epitheliaal fenotype met hoge expressie van CC10 en cytochroom P450 Cyp2f,
uit mesenchymale cellen. Derhalve hypothetiseren wij dat Fstl1 de afgifte van waardoor ze gevoelig zijn voor naftaleen.55,57,58 Verlies van (variant) clubcellen is
ontstekingsmediatoren uit bronchiale epitheelcellen zou kunnen induceren, welke waargenomen bij COPD.53,54,59,60 Lage serum niveaus van club cell secretory protein
op hun beurt de cytokineafgifte uit mesenchymale cellen reguleren. We hebben (CC10, ook bekend als CC16 of SCGB1A1) worden niet alleen geassocieerd met de
verschillende uitgescheiden factoren geïdentificeerd als kandidaten voor het effect ernst en prevalentie van COPD, maar ook met een daling van de longfunctie bij
van Fstl1-CM op IL-6 en IL-8 afgifte uit de longfibroblasten, namelijk IL-6, IL-8, GM- COPD.61–65
CSF en RANTES, en hebben hun functionele bijdrage met behulp van experimenten Tijdens normale mucociliaire differentiatie ontstaan uit basale cellen direct of
met neutraliserende antilichamen bevestigd. Onze studie geeft het eerste in vitro indirect (via intermediaire clubcellen) slijmbekercellen en trilhaarcellen. Intrigerend
bewijs dat Fstl1 de transcriptie en afgifte van inflammatoire cytokinen, zoals GM-CSF genoeg blijkt uit onze studie dat BMP4 basale celdifferentiatie in de richting van
en IL-6, induceert in bronchiale epitheelcellen, die vervolgens de afgifte van IL-6 en variant clubcellen drijft en verdere differentiatie in de richting van slijmbekercellen
IL-8 uit mesenchymale cellen kunnen stimuleren. en trilharencellen onderdrukt (Hoofdstuk 7). Hoewel een eerdere studie een
rol voor BMP4 heeft aangetoond in proximale-distale epitheliale differentiatie
1. Fstl1 en metaplasie van slijmbekercellen in COPD: de rol van BMP4 in in de ontwikkelende longen,38 tonen wij als eerste aan dat BMP4 de variant
clubceldifferentiatie in volwassen humane epitheelcellen in ALI (air-liquid interface) 9
volwassen epitheliale differentiatie
BMP signalering is cruciaal in het aansturen van de proximaal-naar-distale kweken bevordert.
patroonvorming van luchtwegepitheel in foetale en neonatale longontwikkeling. Overeenkomstig met eerdere studies in COPD en longfibrose63,66,67 vonden
In volwassen longen is actieve BMP signalering vereist voor epitheliale regeneratie we dat de functionele BMP4 signalering (Smad1/5/8 fosforylering) is verminderd
na letsel.37–42 BMP4 is een uitgescheiden mesenchymale factor die het lot van in COPD, terwijl Fstl1 expressie is verhoogd (Hoofdstuk 3). Door het remmen van
epitheelcellen tijdens longontwikkeling bepaalt.43 Daarnaast is BMP4 vereist voor de negatieve regulatie van de BMP signaaltransductieroute op differentiatie van
epitheliale celdifferentiatie, omdat werd aangetoond dat de BMP4 expressie slijmbekercellen en basale cellen zou het Toegenomen Fstl1 in luchtwegepitheel
bij COPD kunnen bijdragen aan de epitheliale differentiatie in de richting van

170 171
slijmbekercelhyperplasie, basale celmetaplasie, en verlies van clubcellen. Dit zijn behandeling na 14 dagen in BMP4 behandelde cellen, zouden kunnen beantwoorden
typische kenmerken zijn van ge-remodeled distaal epitheel bij COPD. Onze studie of het effect van BMP4 omkeerbaar is, en of de schakeling naar het distale
toont een plausibel verband aan tussen het verlies van clubcellen en slijmbekercel- variant clubceldifferentiatieprogramma kan worden omgeleid naar een proximaal
en basale celmetaplasie enerzijds en de waargenomen vermindering van de BMP/ trilhaarceldifferentiatieprogramma. Eerder is aangetoond dat BMP-antagonisten
Smad signalering bij COPD-patiënten anderzijds.66,67 kunnen functioneren als een positieve regulator van basale celproliferatie.68,69 Een
Bovendien hebben we waargenomen dat BMP4 de expressie van zijn eigen aanhoudende toename van de BMP antagonist Fstl1 in COPD kan dus leiden tot
endogene antagonist Fstl1 induceert tijdens luchtwegepitheeldifferentiatie in een basale celproliferatie en basale celhyperplasie, wat gewoonlijk wordt waargenomen
tijds- en concentratieafhankelijke manier (Hoofdstuk 7). Dit impliceert dat Fstl1 in luchtwegepitheelremodeling bij COPD.
betrokken zou kunnen zijn bij BMP functies in epitheeldifferentiatie. Toekomstige Tijdens de longontwikkeling wordt Fstl1 tot expressie gebracht door
studies met Fstl1-CM of Fstl1 neutraliserend antilichaam, en het staken van BMP4 slijmbekercellen,70 terwijl het in de gezonde volwassen longen voornamelijk de
trilhaarcellen zijn die Fstl1 tot expressie brengen (Hoofdstuk 3 en 5). Daarentegen
wordt BMP4 voornamelijk tot expressie gebracht door mesenchymale cellen, zoals
BMP4 longfibroblasten en luchtweggladde spiercellen. Daarom vermoed ik dat epitheliale-
mesenchymale communicatie via BMP-signalering een belangrijke regulator van
Hoofdstuk 7 slijmbekercelmetaplasie verlies van clubcellen
trilhaarcel en slijmbekercel differentiatie is.77
Daarnaast vonden we dat BMP-behandelde cellen verhoogde expressie van
zognaamde Notch-gereguleerde genen hebben (Hoofdstuk 3). Notch bepaalt het
Hoofdstuk 3
uiteindelijke lot van epitheelcellen in de luchtwegen. Daarom is het belangrijk de
Fstl1
crosstalk tussen BMP en Notch bij longherstel van de volwassen longen te begrijpen
om de afwijkende regeneratie van geremodeled epitheel bij COPD te verklaren. In
Fstl1
COPD zijn Notch signaleringscomponenten, zoals Hey1 en Hey2, gedownreguleerd.63
Hoofdstuk 5 Crosstalk tussen BMP en Notch signalering wordt ook waargenomen tijdens de
GM-CSF postnatale ontwikkeling van de longen, waar endotheliale deletie van Fstl1 in Fstl1-
RANTES
IL-6
eKO muizen was geassocieerd met verhoogde BMP-gereguleerd Jagged1 (Hoofdstuk
IL-8 2). Daarnaast werden de Notch targetgenen Hey1 en Hey3 verhoogd tot expressie
mesenchymale cellen
gebracht in primaire humane bronchiale epitheelcellen na stimulatie met BMP4.
Deze waarnemingen suggereren dat soortgelijke mechanismen een rol spelen in
IL-6
IL-8 volwassen longcellen en in de ontwikkeling van de longen. Het zou interessant zijn
om de samenstelling van het epitheel van de luchtwegen van Fstl1-eKO muizen
te karakteriseren om te onderzoeken of variant clubcellen overheersen in het
epitheel van Fstl1 deficiënte muizen. Daarnaast zijn toekomstige studies nodig om
te onderzoeken of de remming van Notch signalering door middel van het gebruik
neutrofielen macrofagen
van neutraliserende antilichamen, BMP-geïnduceerde variant club cel differentiatie
in het ALI kweeksysteem kan remmen.
Samengevat tonen onze studies aan dat BMP4 de differentiatie van
slijmbekercellen remt, terwijl het de differentiatie van variant club-cellen bevordert.
Luchtwegontsteking
Herstel van deze onbalans in de longen van COPD patiënten zou een nieuwe strategie 9
Figuur 2. De pathologische gevolgen van het toegenomen Fstl1 niveau in het epitheel kunnen zijn om het herstel van de waargenomen verminderde BMP-gereguleerde
van de luchtwegen van COPD-patiënten. De expressie van het Fstl1 eiwit is toegenomen Smad1/5 in COPD longen te verbeteren, alsmede om het verlies van clubcellen tegen
in het luchtwegepitheel van COPD patiënten. Fstl1 is een pro-inflammatoir cytokine dat
de epitheliale-mesenchymale communicatie bij IL-6 en IL-8 afgifte medieert. Zo kan Fstl1 te gaan en de metaplasie van slijmbekercellen te verminderen. Onze studie spoort
bijdragen aan de rekrutering van neutrofielen en macrofagen naar de longen. BMP4 remt toekomstige studies aan om een ​​dergelijke strategie te onderzoeken in modellen
de differentiatie van slijmbekercellen, terwijl het de differentiatie van variant clubcellen in voor luchtwegepitheel schade en herstel in vitro en in vivo. De pathologische
het epitheel van de luchtwegen van de volwassen mens bevordert. Toegenomenase Fstl1 in
het epitheel van de luchtwegen bij COPD kan zo slijmbekercel hyperplasie bevorderen en het gevolgen van het toegenomen Fstl1 niveau in het epitheel van de luchtwegen van
verlies van clubcellen in de hand werken. COPD-patiënten is weergegeven in Figuur 2.

172 173
2. Toekomstperspectieven medieert bij de afgifte van cytokinen (Hoofdstuk 4), zou kunnen bijdragen aan de
De in dit proefschrift gepresenteerde gegevens belichten de tot nu toe onbekende aangeboren immuunrespons van de long die volgt op blootstelling aan bijvoorbeeld
rol van Fstl1/BMP signalering in COPD en bespreken de mogelijke gevolgen van sigarettenrook. Om inzicht te verkrijgen in de mogelijke signaaltransductieroutes
verhoogde expressie van het Fstl1 eiwit bij luchtwegontsteking en epitheliale die betrokken zijn bij Fstl1-aangedreven lokale luchtwegontsteking in de longen
remodeling. zijn verdere studies nodig om de receptor(en) waarmee Fstl1 zijn werking in
PAH is een veel voorkomende comorbiditeit bij COPD.71,72 Onze studie biedt in epitheelcellen en in pulmonaire mesenchymale cellen uitoefent te identificeren.
vivo bewijs voor een belangrijke rol voor endotheliaal Fstl1 in het titreren van de Verder tonen we aan dat BMP4 de differentiatie van slijmbekercellen remt,
niveaus van BMP/Smad signalering tijdens de postnatale ontwikkeling van de longen terwijl het de differentiatie van variant clubcellen bevordert (Hoofdstuk 5). Follow-up
(Hoofdstuk 2). Echter, de rechtstreekse link en de specifieke functies van endotheliaal studies met behulp van muismodellen van COPD zijn nodig om het functionele effect
Fstl1 in pulmonale arteriële celproliferatie van gladde spiercellen en bij de regulatie van de toegenomen Fstl1 in het luchtwegepitheel te onderzoeken, om te beoordelen
van actine remodeling in pulmonale arteriële gladde spiercellen in de longen van of dit de BMP signalering remt en aldus ​​bijdraagt aan de slijmbekercelmetaplasie en
pasgeborenen en in COPD longen moet nog worden opgehelderd. Ongecontroleerde het verlies van variant clubcellen. Experimenten met behulp van neutraliserende
proliferatie van pulmonale arteriële gladde spiercellen en verminderde actine antilichamen tegen Fstl1 zouden waardevol zijn om deze implicaties te onderzoeken.
remodeling dragen bij aan vaatvernauwing in PAH. Ex vivo studies met precision-cut Gezien de postnatale sterfte van Fstl1-eKO muizen (Hoofdstuk 2) wat verdere studies
lung slices zouden meer duidelijkheid kunnen scheppen over de functionele bijdrage in sigarettenrook-geïnduceerde COPD muismodellen belemmert, zijn follow-up in
van endotheliaal Fstl1 aan vernauwing van de longvaten. Een in vitro experiment vivo studies met behulp van doxycycline geïnduceerde epitheel-specifieke Fstl1-
met een co-kweeksysteem van humane long microvasculaire endotheelcellen en KO de moeite waard om de betrokkenheid van epitheliaal-uitgescheiden Fstl1 in
vasculaire gladde spiercellen zou de vraag kunnen beantwoorden hoe communicatie de ontwikkeling van COPD te bestuderen. Verder kan het van belang zijn om Fstl1
tussen deze celtypen de remodeling en vaatvernauwing reguleert. expressie en BMP-gereguleerde Smad-fosforylering in de proximale geleidende
In longvaten van COPD-patiënten zagen we een toename van Fstl1 eiwitexpressie luchtwegen en de distale luchtwegen te onderzoeken om te bepalen of er een verband
in endotheelcellen en vasculaire gladde spiercellen in vergelijking met personen is tussen de relatieve expressie (ratio) van BMP/Fstl1 en de epitheelcelsamenstelling
met een normale longfunctie (Hoofdstuk 3). Hoewel celspecifieke expressie van in de luchtwegen bij COPD.
door BMP gefosforyleerde Smad signalering niet werd onderzocht, bleek dat de Samengevat speelt een verstoorde activering van Fstl1/BMP
BMP-Smad gereguleerde activatie in longhomogenaten van COPD patiënten was signaaltransductie na epitheel- en vasculaire schade een rol bij COPD. Het is tevens
verminderd vergeleken met controles zonder COPD (Hoofdstuk 3). Activering van belangrijk om de functionele rol van de Fstl1/BMP as tijdens longontwikkeling te
BMP signalering is verminderd bij patiënten met idiopathische PAH.73–75 Echter, de begrijpen. Dit zal leiden tot beter begrip van het abnormale weefselherstelproces
bijdrage van Fstl1 aan deze verlaging werd niet onderzocht. Eerdere studies toonden bij COPD.
aan dat een endogene BMP-remmer, Gremlin, was verhoogd in endotheelcellen van
idiopathische PAH en van PAH veroorzaakt door zuurstofgebrek.76 Bovendien was bij
haploinsufficiëntie van Gremlin 1 de pulmonale vasculaire weerstand verminderd
door het verzwakken van vasculaire remodeling,76 wat suggereert dat remming
van BMP leidt tot vasculaire structurele veranderingen en tot vaatvernauwing.86
Uit recent onderzoek blijkt dat Fstl1 de BMP/Smad signalering kan remmen, terwijl
het de TGF-β-geïnduceerde α-smooth muscle actin expressie door longfibroblasten
vergemakkelijkt, waardoor wordt bijgedragen aan extracellulaire matrix remodeling
en luchtwegfibrose.9 Het zou zo kunnen zijn dat Fstl1 de α-smooth muscle actin-
productie door vasculaire gladde spieren verhoogt en structurele vasculaire 9
veranderingen bevordert, waarmee het uiteindelijk bijdraagt ​​aan toegenomen
vaatweerstand en PAH. Een toekomstige studie ter evaluatie van het verband tussen
Fstl1 expressie en pulmonale vaatfunctie bij patiënten met PAH secundair aan COPD
zou waardevol zijn om uit te voeren.
Dit proefschrift onderzocht ook de mogelijke gevolgen van de toegenomen
Fstl1 expressie bij COPD, met name in het epitheel van de luchtwegen. Een pro-
inflammatoire rol voor Fstl1, welke epitheliale-mesenchymale communicatie

174 175
References doi:10.1002/bdrc.21085.
1. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal 21. International PPH Consortium, Lane KB, Machado RD, et al. Heterozygous germline mutations
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616. in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat
2. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP) Genet. 2000;26(1):81-84. doi:10.1038/79226.
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A. 22. Machado RD, Aldred MA, James V, et al. Mutations of the TGF-beta type II receptor BMPR2 in
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108. pulmonary arterial hypertension. Hum Mutat. 2006;27(2):121-132. doi:10.1002/humu.20285.
3. Umezu T, Yamanouchi H, Iida Y, Miura M, Tomooka Y. Follistatin-like-1, a diffusible mesenchymal 23. Harrison RE, Flanagan JA, Sankelo M, et al. Molecular and functional analysis identifies ALK-
factor determines the fate of epithelium. Proc Natl Acad Sci U S A. 2010;107(10):4601-4606. 1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic
doi:10.1073/pnas.0909501107. telangiectasia. J Med Genet. 2003;40(12):865-871.
4. Hayakawa S, Ohashi K, Shibata R, et al. Cardiac Myocyte-Derived Follistatin-Like 1 Prevents 24. Fujiwara M, Yagi H, Matsuoka R, et al. Implications of mutations of activin receptor-like kinase
Renal Injury in a Subtotal Nephrectomy Model. J Am Soc Nephrol JASN. 2015;26(3):636-646. 1 gene (ALK1) in addition to bone morphogenetic protein receptor II gene (BMPR2) in children
doi:10.1681/ASN.2014020210. with pulmonary arterial hypertension. Circ J Off J Jpn Circ Soc. 2008;72(1):127-133.
5. Zhou X, Xiao X, Huang T, et al. Epigenetic inactivation of follistatin-like 1 mediates tumor immune 25. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in
evasion in nasopharyngeal carcinoma. Oncotarget. 2016;7(13):16433-16444. doi:10.18632/ hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(2):189-195. doi:10.1038/
oncotarget.7654. ng0696-189.
6. Miyamae T, Marinov AD, Sowders D, et al. Follistatin-Like Protein-1 Is a Novel Proinflammatory 26. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-beta binding protein of endothelial
Molecule. J Immunol. 2006;177(7):4758-4762. doi:10.4049/jimmunol.177.7.4758. cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8(4):345-
7. Clutter SD, Wilson DC, Marinov AD, Hirsch R. Follistatin-Like Protein 1 Promotes Arthritis by Up- 351. doi:10.1038/ng1294-345.
Regulating IFN-γ. J Immunol. 2009;182(1):234-239. doi:10.4049/jimmunol.182.1.234. 27. Pi X, Lockyer P, Dyer LA, et al. Bmper inhibits endothelial expression of inflammatory adhesion
8. Chaly Y, Marinov AD, Oxburgh L, Bushnell DS, Hirsch R. FSTL1 promotes arthritis in mice by molecules and protects against atherosclerosis. Arterioscler Thromb Vasc Biol. 2012;32(9):2214-
enhancing inflammatory cytokine/chemokine expression. Arthritis Rheum. 2012;64(4):1082- 2222. doi:10.1161/ATVBAHA.112.252015.
1088. doi:10.1002/art.33422. 28. Yao Y, Yao J, Radparvar M, et al. Reducing Jagged 1 and 2 levels prevents cerebral arteriovenous
9. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary malformations in matrix Gla protein deficiency. Proc Natl Acad Sci U S A. 2013;110(47):19071-
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878. 19076. doi:10.1073/pnas.1310905110.
10. Liu R, Yang Y, Shen J, et al. Fstl1 is involved in the regulation of radial glial scaffold development. 29. Jain RK. Molecular regulation of vessel maturation. Nat Med. 2003;9(6):685-693. doi:10.1038/
Mol Brain. 2015;8:53. doi:10.1186/s13041-015-0144-8. nm0603-685.
11. Murphy N, Gaynor KU, Rowan SC, et al. Altered Expression of Bone Morphogenetic Protein 30. Knight D. Epithelium–fibroblast interactions in response to airway inflammation. Immunol Cell
Accessory Proteins in Murine and Human Pulmonary Fibrosis. Am J Pathol. 2016;186(3):600- Biol. 2001;79(2):160-164. doi:10.1046/j.1440-1711.2001.00988.x.
615. doi:10.1016/j.ajpath.2015.10.032. 31. Osei ET, Brandsma C-A, Noordhoek JA, Timens W, Postma D, Heijink I. Crosstalk between
12. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by epithelium and fibroblasts; implications for COPD. Eur Respir J. 2014;44(Suppl 58):P3899.
Inducing Oncostatin M. J Immunol Baltim Md 1950. 2015;195(8):3546-3556. doi:10.4049/ 32. Ouchi N, Oshima Y, Ohashi K, et al. Follistatin-like 1, a secreted muscle protein, promotes
jimmunol.1501105. endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase-
13. Miller M, Esnault S, Kurten RC, et al. Segmental allergen challenge increases levels of airway dependent mechanism. J Biol Chem. 2008;283(47):32802-32811. doi:10.1074/jbc.M803440200.
follistatin-like 1 in patients with asthma. J Allergy Clin Immunol. 2016;138(2):596-599.e4. 33. Shibanuma M, Mashimo J, Mita A, Kuroki T, Nose K. Cloning from a mouse osteoblastic cell line
doi:10.1016/j.jaci.2016.01.019. of a set of transforming-growth-factor-beta 1-regulated genes, one of which seems to encode a
14. Gangopahyay A, Oran M, Bauer EM, et al. Bone morphogenetic protein receptor II is a novel follistatin-related polypeptide. Eur J Biochem FEBS. 1993;217(1):13-19.
mediator of endothelial nitric-oxide synthase activation. J Biol Chem. 2011;286(38):33134- 34. Wilson DC, Marinov AD, Blair HC, et al. Follistatin-like protein 1 is a mesenchyme-derived
33140. doi:10.1074/jbc.M111.274100. inflammatory protein and may represent a biomarker for systemic-onset juvenile rheumatoid
15. Xu J, Zhu D, Sonoda S, et al. Over-expression of BMP4 inhibits experimental choroidal arthritis. Arthritis Rheum. 2010;62(8):2510-2516. doi:10.1002/art.27485.
neovascularization by modulating VEGF and MMP-9. Angiogenesis. 2012;15(2):213-227. 35. Murakami K, Tanaka M, Usui T, et al. Follistatin-related protein/follistatin-like 1 evokes an
doi:10.1007/s10456-012-9254-4. innate immune response via CD14 and toll-like receptor 4. FEBS Lett. 2012;586(4):319-324.
16. Moreno-Miralles I, Ren R, Moser M, Hartnett ME, Patterson C. Bone morphogenetic protein doi:10.1016/j.febslet.2012.01.010.
endothelial cell precursor-derived regulator regulates retinal angiogenesis in vivo in a mouse 36. Fan N, Sun H, Wang Y, et al. Follistatin-Like 1: A Potential Mediator of Inflammation in Obesity.
model of oxygen-induced retinopathy. Arterioscler Thromb Vasc Biol. 2011;31(10):2216-2222. Mediators Inflamm. 2013;2013:e752519. doi:10.1155/2013/752519.
doi:10.1161/ATVBAHA.111.230235. 37. Bellusci S, Henderson R, Winnier G, Oikawa T, Hogan BL. Evidence from normal expression
17. Helbing T, Rothweiler R, Ketterer E, et al. BMP activity controlled by BMPER regulates the and targeted misexpression that bone morphogenetic protein (Bmp-4) plays a role in mouse 9
proinflammatory phenotype of endothelium. Blood. 2011;118(18):5040-5049. doi:10.1182/ embryonic lung morphogenesis. Dev Camb Engl. 1996;122(6):1693-1702.
blood-2011-03-339762. 38. Weaver M, Yingling JM, Dunn NR, Bellusci S, Hogan BL. Bmp signaling regulates proximal-distal
18. Yao Y, Jumabay M, Wang A, Boström KI. Matrix Gla protein deficiency causes arteriovenous differentiation of endoderm in mouse lung development. Dev Camb Engl. 1999;126(18):4005-
malformations in mice. J Clin Invest. 2011;121(8):2993-3004. doi:10.1172/JCI57567. 4015.
19. Suzuki Y, Ohga N, Morishita Y, Hida K, Miyazono K, Watabe T. BMP-9 induces proliferation of 39. Sountoulidis A, Stavropoulos A, Giaglis S, et al. Activation of the canonical bone morphogenetic
multiple types of endothelial cells in vitro and in vivo. J Cell Sci. 2010;123(Pt 10):1684-1692. protein (BMP) pathway during lung morphogenesis and adult lung tissue repair. PloS One.
doi:10.1242/jcs.061556. 2012;7(8):e41460. doi:10.1371/journal.pone.0041460.
20. Kool H, Mous D, Tibboel D, de Klein A, Rottier RJ. Pulmonary vascular development goes awry in 40. Ninomiya N, Michiue T, Asashima M, Kurisaki A. BMP signaling regulates the differentiation
congenital lung abnormalities. Birth Defects Res Part C Embryo Today Rev. 2014;102(4):343-358. of mouse embryonic stem cells into lung epithelial cell lineages. In Vitro Cell Dev Biol Anim.

176 177
2013;49(3):230-237. doi:10.1007/s11626-013-9589-1. 62. Vestbo J, Edwards LD, Scanlon PD, et al. Changes in Forced Expiratory Volume in 1 Second over
41. Huang SXL, Islam MN, O’Neill J, et al. Highly efficient generation of airway and lung epithelial cells Time in COPD. N Engl J Med. 2011;365(13):1184-1192. doi:10.1056/NEJMoa1105482.
from human pluripotent stem cells. Nat Biotechnol. 2014;32(1):84-91. doi:10.1038/nbt.2754. 63. Park HY, Churg A, Wright JL, et al. Club cell protein 16 and disease progression in chronic
42. Huang SXL, Green MD, de Carvalho AT, et al. The in vitro generation of lung and airway progenitor obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(12):1413-1419.
cells from human pluripotent stem cells. Nat Protoc. 2015;10(3):413-425. doi:10.1038/ doi:10.1164/rccm.201305-0892OC.
nprot.2015.023. 64. Lomas DA, Silverman EK, Edwards LD, et al. Evaluation of serum CC-16 as a biomarker for COPD
43. Morrisey EE, Hogan BLM. Preparing for the first breath: genetic and cellular mechanisms in lung in the ECLIPSE cohort. Thorax. 2008;63(12):1058-1063. doi:10.1136/thx.2008.102574.
development. Dev Cell. 2010;18(1):8-23. doi:10.1016/j.devcel.2009.12.010. 65. Guerra S, Halonen M, Vasquez MM, et al. The relation of circulating CC16 to lung function growth,
44. Ross AJ, Dailey LA, Brighton LE, Devlin RB. Transcriptional profiling of mucociliary differentiation decline, and development of COPD across the lifespan. Lancet Respir Med. 2015;3(8):613-620.
in human airway epithelial cells. Am J Respir Cell Mol Biol. 2007;37(2):169-185. doi:10.1165/ doi:10.1016/S2213-2600(15)00196-4.
rcmb.2006-0466OC. 66. Myllärniemi M, Lindholm P, Ryynänen MJ, et al. Gremlin-mediated decrease in bone
45. Fahy JV, Dickey BF. Airway Mucus Function and Dysfunction. N Engl J Med. 2010;363(23):2233- morphogenetic protein signaling promotes pulmonary fibrosis. Am J Respir Crit Care
2247. doi:10.1056/NEJMra0910061. Med. 2008;177(3):321-329. doi:10.1164/rccm.200706-945OC.
46. Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. Basal cells are a multipotent progenitor
67. De Langhe E, Cailotto F, De Vooght V, et al. Enhanced endogenous bone morphogenetic
capable of renewing the bronchial epithelium. Am J Pathol. 2004;164(2):577-588. doi:10.1016/
S0002-9440(10)63147-1.
protein signaling protects against bleomycin induced pulmonary fibrosis. Respir Res.
47. Rock JR, Onaitis MW, Rawlins EL, et al. Basal cells as stem cells of the mouse trachea and 2015;16:38. doi:10.1186/s12931-015-0202-x.
human airway epithelium. Proc Natl Acad Sci U S A. 2009;106(31):12771-12775. doi:10.1073/ 68. Cibois M, Luxardi G, Chevalier B, et al. BMP signalling controls the construction of
pnas.0906850106. vertebrate mucociliary epithelia. Development. 2015;142(13):2352-2363. doi:10.1242/
48. Kimura J, Deutsch GH. Key mechanisms of early lung development. Pediatr Dev Pathol Off J Soc dev.118679.
Pediatr Pathol Paediatr Pathol Soc. 2007;10(5):335-347. doi:10.2350/07-06-0290.1. 69. Tadokoro T, Gao X, Hong CC, Hotten D, Hogan BLM. BMP signaling and cellular
49. Rawlins EL, Hogan BLM. Epithelial stem cells of the lung: privileged few or opportunities for dynamics during regeneration of airway epithelium from basal progenitors. Dev Camb
many? Dev Camb Engl. 2006;133(13):2455-2465. doi:10.1242/dev.02407. Engl. 2016;143(5):764-773. doi:10.1242/dev.126656.
50. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell
70. Adams D, Larman B, Oxburgh L. Developmental expression of mouse Follistatin-like 1
populations. Nat Med. 2014;20(8):822-832. doi:10.1038/nm.3642.
51. Schamberger AC, Staab-Weijnitz CA, Mise-Racek N, Eickelberg O. Cigarette smoke alters primary
(Fstl1): Dynamic regulation during organogenesis of the kidney and lung. Gene Expr
human bronchial epithelial cell differentiation at the air-liquid interface. Sci Rep. 2015;5. Patterns. 2007;7(4):491-500. doi:10.1016/j.modgep.2006.10.009.
doi:10.1038/srep08163. 71. Siafakas NM, Antoniou KM, Tzortzaki EG. Role of angiogenesis and vascular
52. Saetta M, Turato G, Baraldo S, et al. Goblet cell hyperplasia and epithelial inflammation remodeling in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis.
in peripheral airways of smokers with both symptoms of chronic bronchitis and chronic 2007;2(4):453-462.
airflow limitation. Am J Respir Crit Care Med. 2000;161(3 Pt 1):1016-1021. doi:10.1164/ 72. Hashimoto M, Tanaka H, Abe S. Quantitative analysis of bronchial wall vascularity in the
ajrccm.161.3.9907080. medium and small airways of patients with asthma and COPD. Chest. 2005;127(3):965-
53. Gamez AS, Gras D, Petit A, et al. Supplementing defect in club cell secretory protein attenuates 972. doi:10.1378/chest.127.3.965.
airway inflammation in COPD. Chest. 2015;147(6):1467-1476. doi:10.1378/chest.14-1174.
73. Morty RE, Nejman B, Kwapiszewska G, et al. Dysregulated bone morphogenetic protein
54. Pilette C, Godding V, Kiss R, et al. Reduced epithelial expression of secretory component in small
airways correlates with airflow obstruction in chronic obstructive pulmonary disease. Am J
signaling in monocrotaline-induced pulmonary arterial hypertension. Arterioscler
Respir Crit Care Med. 2001;163(1):185-194. doi:10.1164/ajrccm.163.1.9912137. Thromb Vasc Biol. 2007;27(5):1072-1078. doi:10.1161/ATVBAHA.107.141200.
55. Guha A, Vasconcelos M, Cai Y, et al. Neuroepithelial body microenvironment is a niche 74. Takahashi K, Kogaki S, Matsushita T, Nasuno S, Kurotobi S, Ozono K. Hypoxia induces alteration
for a distinct subset of Clara-like precursors in the developing airways. Proc Natl Acad Sci. of bone morphogenetic protein receptor signaling in pulmonary artery endothelial cell. Pediatr
2012;109(31):12592-12597. doi:10.1073/pnas.1204710109. Res. 2007;61(4):392-397. doi:10.1203/pdr.0b013e3180332cba.
56. Barnes PJ. CLub cells, their secretory protein, and copd. Chest. 2015;147(6):1447-1448. 75. Long L, Crosby A, Yang X, et al. Altered Bone Morphogenetic Protein and Transforming
doi:10.1378/chest.14-3171. Growth Factor-β Signaling in Rat Models of Pulmonary Hypertension. Circulation.
57. Hung KS. Development of neuroepithelial bodies in pre- and postnatal mouse lungs: scanning 2009;119(4):566-576. doi:10.1161/CIRCULATIONAHA.108.821504.
electron microscopic study. Anat Rec. 1982;203(2):285-291. doi:10.1002/ar.1092030210. 76. Cahill E, Rowan SC, Sands M, et al. The pathophysiological basis of chronic
58. Reynolds SD, Giangreco A, Power JH, Stripp BR. Neuroepithelial bodies of pulmonary airways hypoxic pulmonary hypertension in the mouse: vasoconstrictor and structural
serve as a reservoir of progenitor cells capable of epithelial regeneration. Am J Pathol. mechanisms contribute equally. Exp Physiol. 2012;97(6):796-806. doi:10.1113/ 9
2000;156(1):269-278. doi:10.1016/S0002-9440(10)64727-X.
expphysiol.2012.065474.
59. Shijubo N, Itoh Y, Yamaguchi T, et al. Clara cell protein-positive epithelial cells are reduced in
small airways of asthmatics. Am J Respir Crit Care Med. 1999;160(3):930-933. doi:10.1164/
ajrccm.160.3.9803113.
60. Rokicki W, Rokicki M, Wojtacha J, Dżeljijli A. The role and importance of club cells (Clara cells) in
the pathogenesis of some respiratory diseases. Kardiochirurgia Torakochirurgia Pol Pol J Cardio-
Thorac Surg. 2016;13(1):26-30. doi:10.5114/kitp.2016.58961.
61. Bernard A, Marchandise FX, Depelchin S, Lauwerys R, Sibille Y. Clara cell protein in serum and
bronchoalveolar lavage. Eur Respir J. 1992;5(10):1231-1238.

178 179
CHAPTER 10 1

INDONESIAN SUMMARY/
RANGKUMAN DALAM
BAHASA INDONESIA

181
Pendahuluan Fstl1 pada sel-sel endotelium dalam perkembangan pembuluh paru-paru setelah
kelahiran. Pada mencit, hari ke-5 hingga ke-30 setelah kelahiran adalah tahap yang
Penyakit paru obstruktif kronik (PPOK) merupakan penyakit yang ditandai dengan penting untuk pembentukan, perkembangan, dan maturasi alveoli serta pembuluh
penyumbatan aliran udara yang bersifat irreversible dan memburuk seiring darah paru-paru.19,29 Pembentukan dan percabangan jaringan saluran pernapasan
waktu disertai dengan peradangan saluran pernapasan yang berlebihan. Seperti dan pembuluh darah harus terkoordinasi dengan baik sehingga paparan darah
yang dijelaskan pada bab pengantar (Bab 1), keseimbangan antara protein bone terhadap oksigen optimal.19 Pembentukan jaringan pembuluh kecil paru-paru pada
morfogenetik protein 4 (BMP4) dan inhibitor alaminya, Follistatin-like 1 (Fstl1) sangat mencit Fstl1-eKO mungkin terganggu sehingga menyebabkan gangguan pernapasan
penting dalam pertumbuhan paru-paru embrio normal1,2 dan dalam menjalankan (Bab 2).
fungsi biologis normal pada paru-paru dewasa.3–5 Beberapa penelitian sebelumnya Meskipun perubahan struktur pembuluh besar arteri otot dan elastis pada
menunjukkan bahwa Fstl1 berfungsi sebagai regulator sistem imun pada berbagai mencit Fstl1-eKO minimal, persentase pembuluh kecil paru-paru yang positif
penyakit inflamasi.6–8 Namun, fungsi dari Fstl1 dalam perkembangan paru-paru mengekspresikan protein aktin meningkat pada hari ke-21 setelah kelahiran (Bab
setelah kelahiran dan paru-paru penderita PPOK belum pernah diteliti sebelumnya. 2). Selain itu, output darah dari ventrikel kanan (right ventricular output) menurun
Baru-baru ini, beberapa hasil penelitian menunjukkan fungsi penting dari Fstl1 dalam pada mencit Fstl1-eKO pada hari ke-21 setelah kelahiran (Bab 2). Molekuler analisis
perkembangan fibrosis paru-paru dan alergi asma.9–13 Penelitian yang dipaparkan pada mencit Fstl1-eKO menunjukkan bahwa Smad1/5/8 terfosforilasi - indikator
dalam tesis ini menunjukkan bahwa Fstl1 diperlukan untuk perkembangan pembuluh aktifnya pensinyalan molekuler BMP - dan ekspresi protein-protein yang diatur oleh
paru-paru setelah kelahiran pada hewan model mencit (Bab 2). Selain itu, penelitian BMP (Jagged1, endoglin, dan endothelin-1) meningkat pada pada hari ke-7 setelah
ini juga menunjukkan peningkatan ekspresi protein Fstl1 pada sel epitel saluran kelahiran (Bab 2).
pernapasan pasien penderita PPOK (Bab 3). Implikasi dari meningkatnya ekspresi Regulasi vasokonstriksi yang dinamis diatur oleh perubahan molekuler,
protein Fstl1 pada sel epitel saluran pernapasan juga didiskusikan pada tesis ini. Hasil termasuk peningkatan endothelin-130,31 dan/atau perubahan morfologi pada
penelitian ini menunjukkan bahwa Fstl1 yang dihasilkan oleh sel-sel epitel saluran jaringan otot polos, termasuk meningkatnya proliferasi sel otot polos pembuluh
pernapasan berperan sebagai mediator peradangan pada paru-paru (Bab 5). Lebih darah dan jumlah ekspresi protein aktin pada pembuluh paru-paru.32,33 Pada mencit
lanjut, hasil penelitian ini juga menunjukkan bahwa BMP4 mengatur diferensiasi sel wildtype transisi regulasi vasokonstriksi dari molekuler ke perubahan morfologi
epitel saluran pernapasan menjadi sel klub varian (Bab 7). dapat diobservasi (Bab 2). Peningkatan ekspresi protein endothelin-1 terjadi
secara bersamaan dengan menurunnya persentase pembuluh kecil paru-paru
yang aktin pada hari ke-14 dibandingkan pada hari ke-7 setelah kelahiran. Transisi
1. Fungsi Fstl1 dalam perkembangan paru-paru
ini normal dan tidak mematikan (Bab 2).Namun pada mencit Fstl1-eKO, ekspresi
protein endothelin-1 sudah tinggi pada hari ke-7 dan tetap tinggi pada hari ke-21
Perkembangan saluran pernapasan dan sistem pembuluh paru-paru yang fungsional
setelah kelahiran, sedangkan persentase pembuluh kecil paru-paru yang positif
diatur secara ketat oleh berbagai jalur pensinyalan molekuler dalam sel. Pensinyalan
aktin tidak menurun pada hari ke-21 setelah kelahiran (Bab 2). Kombinasi ekspresi
molekuler oleh BMP sangat penting dalam proses pertumbuhan dan perkembangan
endothelin-1 yang tinggi dan persentase pembuluh kecil paru-paru yang positif
paru-paru, dengan mengatur proliferasi dan migrasi sel-sel otot polos and sel-
aktin yang tinggi menyebabkan peningkatan resistensi pembuluh darah paru-paru
sel endotel pembuluh,14–18 serta dalam proses percabangan saluran pernapasan
dan menyebabkan vasokonstriksi (Bab 2). Namun, berdasarkan rasio pulmonary
dan pembuluh paru-paru.19 Salah satu penyakit genetik pembuluh darah yang
acceleration time terhadap ejection time (PAT/ET), tidak ada perbedaan yang
disebabkan pensinyalan molekuler BMP yang tidak terkoordinasi baik adalah
signifikan pada resistensi pembuluh paru-paru pada mencit Fstl1-eKO dibandingkan
hipertensi arteri pulmonari (PAH).20–28 Sejalan dengan peran utama pensinyalan
dengan mencit kontrol baik pada hari ke-7 maupun ke-21 setelah kelahiran (Bab
molekuler BMP, mencit yang tidak mengekspresikan gen Fstl1 (Fstl1-knockout [KO])
2). Resistensi pembuluh paru-paru yang diukur pada katup arteri besar pulmonalis
mempunyai ukuran paru-paru yang lebih kecil dan memiliki alveolar septa yang
tidak menunjukkan adanya stenosis pulmonari. Observasi ini mencerminkan tahap
lebih tebal dibandingkan mencit wildtype. Mencit Fstl1-KO juga menderita sesak
awal dari disfungsi pembuluh paru-paru pada mencit Fstl1-eKO. Hal yang sama juga
napas dan meninggal sesaat setelah lahir.1,2 Penelitian ini menunjukkan bahwa Fstl1
biasa diamati pada pasien penderita PAH, di mana perubahan fisiologi dan struktur
berfungsi untuk morfogenesis dan alveolarisasi paru-paru selama perkembangan
diawali oleh perubahan pensinyalan molekuler BMP yang diikuti oleh disfungsi dan
embrio. Hasil penelitian yang dipaparkan pada tesis ini menunjukkan bahwa mencit 10
perubahan struktur pembuluh paru-paru pada stadium lanjut.34 Kemungkinan besar
dengan delesi gen Fstl1 spesifik pada sel-sel endotelium (Fstl1-eKO) juga menderita
peningkatan resistensi pembuluh paru-paru terjadi pada pembuluh kecil, karena
sesak napas dan hanya bertahan hidup selama 21 hari setelah dilahirkan (Bab 2).
resistansi menjadi fungsi 1/r4 (menurut hukum Hagen-Poiseuille), di mana “r” adalah
Kematian setelah kelahiran pada mencit Fstl1-eKO menunjukkan fungsi penting dari

182 183
diameter lumen.35
Pada mencit Fstl1-eKO, peningkatan kadar endothelin-1 secara transien pada
hari ke-7 setelah kelahiran dapat meningkatkan resistensi pembuluh paru-paru.
Karena arteri pulmonalis terhubung ke jantung, peningkatan resistansi pembuluh
pulmonalis ini menyebabkan penurunan output dari ventrikel kanan ke paru-paru
pada hari ke-21 setelah kelahiran (Bab 2). Akibatnya, perubahan morfologi terjadi
pada jantung dan paru-paru pada mencit Fstl1-eKO pada hari ke-21 setelah kelahiran,
seperti hipertrofi ventrikel kanan dan perubahan struktur pembuluh kecil paru-paru
(Bab 2). Perubahan yang progresif dan ireversibel pada struktur kardiopulmoner pada
mencit Fstl1-eKO dapat menyebabkan gangguan pernapasan di mana pembuluh
kecil paru-paru tidak mampu membawa darah ke paru-paru secara memadai.Hal ini
dapat menyebabkan kurangnya pengiriman oksigen ke otak dan akhirnya kematian.
Oleh karena itu, penelitian selanjutnya diperlukan untuk memeriksa kadar oksigen
dan karbon dioksida dalam darah selain menentukan radius dan kandungan aktin
pada pembuluh kecil paru-paru. Secara keseluruhan, perubahan molekuler pada
mencit Fstl1-eKO bersifat sementara dan dimulai sejak dini dalam perkembangan
paru-paru setelah kelahiran, sedangkan fungsi fisiologi dan konsekuensi morfologi
terjadi pada tahap selanjutnya. Disregulasi pensinyalan molekuler BMP saja mungkin
tidak memadai untuk menyebabkan perubahan struktur dan fungsi pembuluh paru-
paru besar pada tahap awal perkembangan, namun dapat diobservasi pada tahap
perkembangan paru-paru selanjutnya. Adapula kemungkinan bahwa perubahan
molekuler dan morfologi di paru-paru merupakan konsekuensi sekunder dari
disfungsi jantung kanan karena penebalan dinding ventrikel kanan. Ventrikel kanan
yang menebal dapat menyebabkan penurunan output dari ventrikel kanan yang Gambar 1. Fungsi Follistatin-like 1 dalam memodulasi pensinyalan molekuler BMP/Smad
kemudian menyebabkan hipoksia paru-paru. Sebagai konsekuensinya, persentase selama perkembangan pembuluh paru-paru setelah melahirkan. Pada mencit kontrol, BMP
menginduksi fosforilasi protein Smad1/5 dan gen targetnya untuk mempertahankan fungsi
pembuluh kecil paru-paru yang positif aktin tidak menurun sementara ekspresi normal dan homeostasis pembuluh paru-paru. Delesi Fstl1 dari endotelium pada mencit
endothelin-1 meningkat di paru-paru pada hari ke-21 setelah kelahiran untuk Fstl1-eKO menyebabkan aktivasi yang berlebihan dari protein Smad1/5 dan gen targetnya,
mendorong aliran darah melalui kapiler paru-paru. Penelitian selanjutnya diperlukan termasuk Endoglin, Jag1, Gata2. Peningkatan ekspresi Gata2 meningkatkan ekspresi
vasokonstriktor endotelin-1 yang dapat menyebabkan perubahan struktur pembuluh paru-
untuk mempelajari struktur dan fungsi kardiovaskular pada mencit Fstl1-eKO untuk paru dan vasokonstriksi.
mengkonfirmasi penyebab utama kematian mencit Fstl1-eKO. Fungsi Fstl1 pada
endotelium dalam mengatur sinyal BMP/Smad pada perkembangan pembuluh
paru-paru setelah kelahiran diilustrasikan pada Gambar 1.
2. Fungsi Fstl1 dalam inflamasi saluran pernapasan pada COPD
Salah satu patogenesis PPOK adalah aktivasi sistem kekebalan tubuh yang
berlebihan dan perbaikan jaringan yang abnormal sebagai respon terhadap polutan
udara, seperti asap rokok, dalam jangka waktu yang panjang. Hal ini menyebabkan
perubahan struktur epitelium dan perubahan fisiologis dari saluran pernapasan
yang berkontribusi terhadap keterbatasan aliran udara pada saluran pernapasan
PPOK.36,37 Sebagai barir pertama dari lingkungan eksternal, sel epitel saluran
pernapasan memproduksi mediator inflamasi untuk berkomunikasi dengan sel
10
mesenkim, termasuk fibroblas paru-paru dan sel otot polos saluran pernapasan.
Fungsi Fstl1 dalam komunikasi antar sel telah dilaporkan sebelumnya.6,9 Hasil
penelitian ini mengindikasikan fungsi Fstl1 dalam mengatur sekresi sitokin inflamasi

184 185
dalam beberapa penyakit inflamasi.6,7,38 Namun, keterlibatan Fstl1 dalam patogenesis produksi protein IL-6 dan GM-CSF, mengindikasikan fungsi Fstl1 sebagai regulator
PPOK belum pernah diteliti sebelumnya. sitokin inflamasi (Bab 5). Medium yang mengandung Fstl1 (Fstl1 CM) yang berasal
Penelitian ini menunjukkan bahwa ekspresi protein Fstl1 meningkat secara dari sel epitel yang mengekspresikan protein Fstl1 secara berlebihan menyebabkan
signifikan pada paru-paru perokok yang tidak menderita PPOK dan pada pasien PPOK meningkatnya produksi protein IL-6 dan IL-8 dari sel mesenkim paru-paru. Kandidat
stadium II dan IV dibandingkan dengan pasien yang tidak menderita PPOK (Bab 3). mediator yang disekresikan oleh sel epitel yang menyebabkan meningkatnya
Analisis lebih lanjut menunjukkan bahwa protein Fstl1 lebih tinggi ekspresinya pada produksi protein IL-6 and IL-8 dari fibroblas paru-paru adalah IL-6, IL-8, GM-CSF dan
sel epitel saluran pernapasan, endotelium, sel otot polos pembuluh paru-paru, dan RANTES. Penelitian ini secara keseluruhan menunjukkan bahwa Fstl1 menginduksi
sel imun pada jaringan paru-paru pasien PPOK stadium II dan IV (semua pernah transkripsi dan produksi sitokin inflamasi, seperti GM-CSF dan IL-6, dari sel epitel
menjadi perokok) dibandingkan jaringan paru-paru pasien yang tidak menderita PPOK bronkus, yang kemudian dapat menginduksi produksi IL-6 dan IL -8 dari sel mesenkim
(Bab 3). Observasi ini mengindikasikan bahwa peningkatan ekspresi protein Fstl1 paru-paru (Bab 5). Fstl1 adalah mediator inflamasi yang mengkoordinasi komunikasi
berhubungan dengan perkembangan PPOK. Tesis ini bertujuan untuk mengevaluasi sel epitel dan sel mesenkim dalam mengatur produksi sitokin inflamasi.Oleh karena
potensi implikasi dari meningkatnya ekspresi protein Fstl1 berkaitan dengan itu, Fstl1 dapat berperan dalam merekrut sel imun ke paru-paru. Sejalan dengan
patogenesis PPOK (Bab 5 dan 7). Ekspresi protein Fstl1 pada paru-paru perokok dan itu, beberapa penelitian sebelumnya menunjukkan keterlibatan Fstl1 dalam proses
mantan perokok yang tidak menderita PPOK juga lebih tinggi dibandingkan pasien patologis penyakit pernapasan, termasuk fibrosis paru-paru dan asma.9,11–13 Tesis
yang tidak pernah merokok dan tidak menderita PPOK. Hal ini mengindikasikan ini mengindikasikan bahwa protein Fstl1 dapat dijadikan target molekuler yang
bahwa adanya efek jangka panjang yang berkaitan dengan pernah merokok pada potensial untuk menghambat inflamasi pada saluran pernapasan pada paru-paru
ekspresi protein Fstl1 di paru-paru (Bab 3). Asap rokok menstimulasi produksi PPOK.
mediator inflamasi yang berkontribusi dalam perekrutan sel-sel imun ke paru-
paru, dimana efek asap rokok tersebut tetap ada setelah berhenti merokok. Fstl1 3. Implikasi dari protein Fstl1 pada sel goblet dan sel basal pada PPOK: Fungsi
adalah protein yang diregulasi oleh TGF-β1 dan asap rokok menyebabkan expresi dari BMP4 pada diferensiasi sel epitel saluran pernapasan dewasa
protein TGF-β1 meningkat pada sel epitel bronkus.39 Hal tersebut menunjukkan Pensinyalan molekuler BMP sangat penting dalam mengatur proksimal-distal axis
bahwa TGF-β1 dapat memediasi peningkatan ekspresi protein Fstl1 sebagai respons saluran pernapasan dalam perkembangan paru-paru embrio dan bayi. Pada paru-
terhadap asap rokok. paru dewasa, aktivasi pensinyalan molekuler BMP diperlukan untuk regenerasi
Sel epitel bronkus mengekspresikan dan mensekresikan protein Fstl1 yang jaringan epitel paru-paru.41–46 BMP4 adalah faktor yang disekresikan oleh jaringan
paling banyak dibandingkan dengan sel mesenkim paru-paru, seperti fibroblas mesenkim yang mampu menentukan nasib sel epitel selama perkembangan paru-
paru-paru dan sel otot polos saluran pernapasan (Bab 5). Sama halnya dengan paru.40 Selain itu, BMP4 diperlukan untuk diferensiasi sel epitel saluran pernapasan
sel-sel epitel saluran pernapasan pada jaringan paru-paru pasien non-PPOK juga dalam kultur in vitroair-liquid interface (ALI).47
mengekspresikan protein Fstl1 paling banyak (Bab 3). Mengingat bahwa Fstl1 adalah Sel epitel saluran pernapasan berfungsi sebagai pelindung pertama dari
inhibitor alami dari BMP4 di paru-paru,1,2 tingkat fosforilasi protein Smad1/5 lebih lingkungan luar. Lapisan sel epitel saluran pernapasan pada orang dewasa terdiri dari
rendah pada paru-paru perokok, mantan perokok non-PPOK, dan pada pasien PPOK sel goblet, sel bersilia, sel basal dan sel klub (sebelumnya dikenal sebagai sel clara).48
stadium II dan IV dibandingkan dengan pasien yang tidak pernah merokok dan tidak Sel goblet yang mensekresikan mukus sebagain besar berada pada bagian proksimal
menderita PPOK (Bab 3). Rendahnya tingkat fosforilasi protein Smad1/5 kemungkinan dari saluran pernapasan, begitu pula sel basal, sel klub, dan sel bersilia.49,50 Jumlah
disebabkan oleh peningkatan ekspresi protein Fstl1 di paru-paru (Bab 3).BMP4 pada sel klub sekretori, sel bersilia, dan sel neuroendokrin berlimpah pada bronkiolus.
umumnya diekspresikan oleh sel mesenkim, seperti fibroblas paru-paru dan sel Alveoli pernapasan dilapisi dengan lapisan tipis sel alveolar tipe I dan sel alveolar
otot polos saluran pernapasan sedangkan sel epitel bronkus mengekspresikan dan kuboidal tipe II.51–53
melepaskan protein BMP4 dengan konsentrasi yang rendah. Ekspresi protein BMP4 Asap rokok mengubah komposisi sel epitel saluran pernapasan dan berkontribusi
pada distal bud paru-paru yang sedang berkembang pada tahap pseudoglandular terhadap perubahan struktur sel epitel pada saluran pernapasan pada pasien
(E11-E16.5), mengkoordinasikan aksis proksimal-distal endoderm dengan mengatur PPOK.54 Perubahan struktur jaringan epitel pada saluran pernapasan pada pasien
diferensiasi sel epitel.40 Perbedaan sumber seluler ligan dan inhibitornya pada paru- PPOK ditandai dengan metaplasia sel goblet, hiperplasia sel basal, serta hilangnya
paru mengindikasikan adanya komunikasi antara sel epitel dan sel mesenkim dalam sel klub yang fungsional. Sel klub varian adalah sel sekretori yang berfungsi sebagai
mengatur fungsi biologis, termasuk regulasi sitokin inflamasi di paru-paru. Tesis ini 10
pelindung jaringan paru-paru dengan mengeluarkan uteroglobin, sekretoglobin, dan
menganalisis fungsi dari protein Fstl1 dalam inflamasi saluran pernapasan pada protein seperti surfaktan yang memiliki fungsi sebagai anti-inflamasi serta penting
individu yang sehat dan fungsinya dalam perkembangan PPOK (Bab 5). untuk regenerasi jaringan epitel saluran pernapasan.55 Sel klub varian memiliki
Ekspresi protein Fstl1 yang berlebihan pada sel epitel bronkus meningkatkan

186 187
karakteristik seperti sel punca dan mampu melakukan pembaharuan diri dan dibandingkan pasien non-PPOK.59
berdiferensiasi menjadi sel bersilia atau sel goblet.53,56 Tingkat serum SCGB1A1 yang Interaksi antara pensinyalan molekuler BMP dan Notch juga diamati selama
rendah tidak hanya berkaitan dengan prevalensi dan tingkat keparahan PPOK, tetapi perkembangan paru-paru setelah kelahiran pada hewan model mencit.Delesi Fstl1
juga dengan penurunan fungsi paru-paru pada pasien PPOK.57–61 pada sel endotel mencit Fstl1-eKO berkaitan dengan peningkatan Jagged1 pada
Pada diferensiasi jaringan epitel saluran pernapasan yang normal, sel basal perkembangan paru-paru setelah kelahiran kelahiran (Bab 2). Selain itu, Hey1 dan
secara langsung atau tidak langsung (melalui perantara sel klub) berdiferensiasi Hey3, meningkat di sel epitel bronkus manusia.
menjadi sel goblet atau sel bersilia. Tesis ini menunjukkan bahwa BMP4 menstimulasi Kesimpulannya, tesis ini menunjukkan bahwa BMP4 menghambat diferensiasi
diferensiasi sel basal ke sel klub varian namun menekan diferensiasi terhadap sel sel goblet sementara menstimulasi diferensiasi sel klub varian pada sel epitel saluran
goblet dan sel bersilia pada sel epitel bronkus dewasa (Bab 7). Meskipun penelitian pernapasan dewasa.Mengembalikan keseimbangan antara BMP4 dan inhibitornya
sebelumnya menunjukkan bahwa BMP4 berfungsi dalam diferensiasi jaringan epitel di paru-paru pasien PPOK merupakan strategi untuk memperbaiki hilangnya sel klub
pada paru-paru yang sedang berkembang,42 hasil penelitian yang dipaparkan dalam
tesis ini menunjukkan bahwa BMP4 menstimulasi diferensiasi sel klub varian pada
sel epitel saluran pernapasan.
Sejalan dengan penelitian sebelumnya,59,62,63 tesis ini juga menunjukkan bahwa BMP4

aktivasi pensinyalan molekuler BMP4 (fosforilasi Smad1/5/8) berkurang sedangkan Bab 7 jumlah sel goblet bertambah

ekspresi Fstl1 meningkat pada paru-paru pasien PPOK (Bab 3). Peningkatan Fstl1
pada sel epitel saluran pernapasan paru-paru PPOK dapat menstimulasi diferensiasi
sel epitel menjadi metaplasia sel goblet, hiperplasia sel basal, dan hilangnya sel klub jumlah sel klub berkurang
Bab 3
yang merupakan ciri khas dari jaringan epitel saluran pernapasan PPOK. Tesis ini Fstl1

menunjukkan adanya hubungan antara hilangnya sel klub, metaplasia sel goblet,
dan hiperplasia sel basal dengan rendahnya aktivasi pensinyalan molekular BMP/ Fstl1

Smad pada paru-paru pasien PPOK.62,63 Bab 5

Studi selanjutnya dengan menggunakan Fstl1 CM atau antibodi penetralisir GM-CSF


RANTES
Fstl1 dan retraksi BMP4 pada sel epitel dewasa setelah 14 hari pada kultur ALI dapat IL-6
IL-8
mengungkapkan apakah efek BMP4 reversible dan apakah proses diferensiasi sel klub mesenchymal cells

varian dapat dialihkan ke diferensiasi sel bersilia. Studi sebelumnya menunjukkan


bahwa inhibitor BMP dapat berfungsi sebagai regulator proliferasi sel basal dengan IL-6
IL-8
menghambat fungsi BMP.64,65 Tingginya protein Fstl1 pada saluran pernapasan pasien
PPOK dapat menstimulasi proliferasi sel basal yang berkontribusi pada hiperplasia
jaringan epitel saluran pernapasan paru-paru PPOK.
Pada paru-paru yang sedang berkembang, Fstl1 diekspresikan oleh sel goblet66
sedangkan pada paru-paru orang dewasa yang sehat, Fstl1 diekspresikan sebagian sel-sel neutrofil sel-sel makrofaga

besar oleh sel bersilia (Bab 3 dan 5). Sebaliknya, sebagian besar BMP4 diekspresikan
oleh sel mesenkim paru-paru, seperti fibroblas paru-paru dan sel otot polos saluran
pernapasan.Oleh karena itu, komunikasi sel epitel dan sel mesenkim melalui inflamasi saluran pernapasan

pensinyalan molekuler BMP penting dalam memodulasi diferensiasi sel bersilia dan
Gambar 2. Implikasi patologi peningkatan protein Fstl1 pada sel epitel saluran pernapasan
sel goblet. pada pasien PPOK. Ekspresi protein Fstl1 meningkat pada sel epitel saluran pernapasan pada
BMP4 menginduksi tingginya ekspresi gen-gen yang dikontrol oleh pensinyalan pasien PPOK stadium II dan IV. Protein Fstl1 kemungkinan diproduksi pada paru-paru dewasa
molekuler Notch selama diferensiasi sel epitel saluran pernapasan dewasa (Bab 3). sebagai mekanisme perbaikan jaringan paru-paru akibat paparan kronis terhadap asap rokok.
Fstl1 adalah sitokin inflamasi yang memediasi komunikasi antara sel epitel dan sel mesenkim
Pensinyalan molekuler Notch menentukan nasib sel epitel di saluran pernapasan. dalam produksi IL-6 dan IL-8. Oleh karena itu, Fstl1 dapat berkontribusi dalam perekrutan
Oleh karena itu, memahami interaksi antara pensinyalan molekuler BMP dan Notch neutrofil dan makrofag ke paru-paru dan menyebabkan inflamasi paru-paru kronis. BMP4 10
sangat penting untuk memperbaikidan regenerasi jaringan epitel saluran pernapasan menghambat diferensiasi sel goblet dan menstimulasi diferensiasi sel klub varian pada sel
epitel saluran pernapasan. Peningkatan Fstl1 pada sel epitel saluran pernapasan pada paru-
yang terganggu pada pasien PPOK. Jumlah komponen pensinyalan molekuler paru PPOK dapat menginduksi diferensiasi sel epitel menjadi sel goblet sementara jumlah
Notch, seperti Hey1 dan Hey2 lebih rendah tingkatnya pada pasien penderita PPOK sel klub berkurang.

188 189
sementara mengurangi metaplasia sel goblet dan hiperplasia sel basal. Gambar 2 BMP dan apakah Fstl1 berkontribusi langsung terhadap metaplasia sel goblet,
mengillustrasikan implikasi potensial dari meningkatnya protein Fstl1 pada sel-sel hiperplasia sel basal, dan hilangnya sel klub varian. Studi berikutnya menggunakan
epitel saluran pernapasan penderita COPD. antibodi penetralisir Fstl1 dan Fstl1 rekombinan protein akan bermanfaat untuk
mengevaluasi implikasi dari Fstl1 di paru-paru pasien penderita PPOK.
4. Perspektif masa depan Kesimpulannya, deregulasi pensinyalan molekuler Fstl1/BMP sebagai respon
Data yang dipaparkan dalam tesis ini menunjukkan fungsi dari Fstl1 pada paru- terhadap kerusakan kronis jaringan epitel saluran pernapasan dan pembuluh paru-
paru yang belum pernah diteliti sebelumnya.Tesis ini menganalisis fungsi dari paru dapat berkontribusi dalam perkembangan PPOK. Oleh karena itu, pemahaman
pensinyalan molekuler BMP/Fstl1 dan mengeksplorasi potensi implikasi dari yang baik mengenai mekanisme regulasi Fstl1/BMP dalam perkembangan paru-paru
meningkatnya ekspresi protein Fstl1 pada peradangan saluran pernapasan serta setelah kelahiran sangat penting untuk mengidentifikasi proses perbaikan jaringan
perubahan struktur lapisan epitel pada saluran pernapasan pasien PPOK. PAH paru-paru yang abnormal.
adalah komorbiditas yang umum pada pasien PPOK.67,68 Studi kami menunjukkan
salah satu fungsi dari Fstl1 pada sel endotel pembuluh paru-paru, yaitu mengatur
tingkat aktivasi BMP/Smad pada perkembangan paru-paru setelah kelahiran (Bab 2).
Proliferasi sel otot polos pembuluh paru-paru yang tidak terkontrol dan perubahan
struktur aktin menyebabkan vasokonstriksi pada pasien PAH. Studi ex vivo dengan
menggunakan precision cut lung slices dapat mengungkapkan kontribusi fungsional
dari Fstl1 endotel pada kontraksi pembuluh dengan membandingkan kontraktilitas
pembuluh paru-paru antara mencit Fstl1-eKO dengan kontrol. Eksperimen dengan
menggunakan sistem ko-kultur sel endotel mikrovaskuler dan sel otot polos
pembuluh paru-paru dapat mengungkapkan mekanisme Fstl1 dalam mengatur
ekspresi aktin pada sel otot polos pembuluh paru-paru.
Pada pembuluh paru-paru pasien PPOK, peningkatan ekspresi protein Fstl1 pada
sel endotel serta sel otot polos pembuluh paru-paru ditemukan pada pasien penderita
PPOK (Bab 3). Tesis ini juga menunjukkan bahwa tingkat aktivasi pensinyalan BMP/
Smad berkurang pada paru-paru pasien PPOK dibandingkan dengan pasien non-
PPOK (Bab 3). Aktivasi pensinyalan molekuler BMP juga berkurang pada pasien PAH
idiopatik,69–71 namun kontribusi Fstl1 terhadap berkurangnya aktivasi pensinyalan
molekuler BMP belum diteliti. Penelitian sebelumnya menunjukkan bahwa inhibitor
BMP lainnya, seperti gremlin, meningkat pada sel endotel dari pasien PAH idiopatik
maupun hipoksia.72
Studi selanjutnya untuk mengevaluasi hubungan antara ekspresi Fstl1 dan
fungsi pembuluh paru-paru pada pasien PPOK dengan komorbiditas PAH akan
sangat bermanfaat untuk dilakukan. Tesis ini juga mengeksplorasi implikasi potensial
peningkatan Fstl1 pada pasien PPOK, terutama pada sel epitel saluran pernapasan.
Fstl1 berperan sebagai pro-inflamasi karena dapat memediasi komunikasi antara
sel epitel dan sel mesenkim dalam mengatur produksi sitokin inflamasi (Bab 4).
Penelitian lebih lanjut diperlukan untuk mengidentifikasi reseptor Fstl1 yang terlibat
di sel epitel maupun sel mesenkim saluran pernapasan.
BMP4 menghambat diferensiasi sel goblet sementara menstimulasi diferensiasi
sel klub varian (Bab 5). Penelitian selanjutnya menggunakan mencit model PPOK
diperlukan untuk mengkonfirmasi efek fungsional dari meningkatnya ekspresi protein
10
Fstl1 pada jaringan epitel saluran pernapasan. Di samping itu, perlu dilakukan studi
selanjutnya untuk mengevaluasi apakah Fstl1 menghambat pensinyalan molekuler

190 191
References in BMPR2, encoding a TGF-beta receptor, cause familial primary pulmonary hypertension. Nat
1. Sylva M, Li VSW, Buffing AAA, et al. The BMP antagonist follistatin-like 1 is required for skeletal Genet. 2000;26(1):81-84. doi:10.1038/79226.
and lung organogenesis. PloS One. 2011;6(8):e22616. doi:10.1371/journal.pone.0022616. 21. Machado RD, Aldred MA, James V, et al. Mutations of the TGF-beta type II receptor BMPR2 in
2. Geng Y, Dong Y, Yu M, et al. Follistatin-like 1 (Fstl1) is a bone morphogenetic protein (BMP) pulmonary arterial hypertension. Hum Mutat. 2006;27(2):121-132. doi:10.1002/humu.20285.
4 signaling antagonist in controlling mouse lung development. Proc Natl Acad Sci U S A. 22. Harrison RE, Flanagan JA, Sankelo M, et al. Molecular and functional analysis identifies ALK-
2011;108(17):7058-7063. doi:10.1073/pnas.1007293108. 1 as the predominant cause of pulmonary hypertension related to hereditary haemorrhagic
3. Umezu T, Yamanouchi H, Iida Y, Miura M, Tomooka Y. Follistatin-like-1, a diffusible mesenchymal telangiectasia. J Med Genet. 2003;40(12):865-871.
factor determines the fate of epithelium. Proc Natl Acad Sci U S A. 2010;107(10):4601-4606. 23. Fujiwara M, Yagi H, Matsuoka R, et al. Implications of mutations of activin receptor-like kinase
doi:10.1073/pnas.0909501107. 1 gene (ALK1) in addition to bone morphogenetic protein receptor II gene (BMPR2) in children
4. Hayakawa S, Ohashi K, Shibata R, et al. Cardiac Myocyte-Derived Follistatin-Like 1 Prevents with pulmonary arterial hypertension. Circ J Off J Jpn Circ Soc. 2008;72(1):127-133.
Renal Injury in a Subtotal Nephrectomy Model. J Am Soc Nephrol JASN. 2015;26(3):636-646. 24. Johnson DW, Berg JN, Baldwin MA, et al. Mutations in the activin receptor-like kinase 1 gene in
doi:10.1681/ASN.2014020210. hereditary haemorrhagic telangiectasia type 2. Nat Genet. 1996;13(2):189-195. doi:10.1038/
5. Zhou X, Xiao X, Huang T, et al. Epigenetic inactivation of follistatin-like 1 mediates tumor immune ng0696-189.
evasion in nasopharyngeal carcinoma. Oncotarget. 2016;7(13):16433-16444. doi:10.18632/ 25. McAllister KA, Grogg KM, Johnson DW, et al. Endoglin, a TGF-beta binding protein of endothelial
oncotarget.7654. cells, is the gene for hereditary haemorrhagic telangiectasia type 1. Nat Genet. 1994;8(4):345-
6. Miyamae T, Marinov AD, Sowders D, et al. Follistatin-Like Protein-1 Is a Novel Proinflammatory 351. doi:10.1038/ng1294-345.
Molecule. J Immunol. 2006;177(7):4758-4762. doi:10.4049/jimmunol.177.7.4758. 26. Pi X, Schmitt CE, Xie L, et al. LRP1-dependent endocytic mechanism governs the signaling output
7. Clutter SD, Wilson DC, Marinov AD, Hirsch R. Follistatin-Like Protein 1 Promotes Arthritis by Up- of the bmp system in endothelial cells and in angiogenesis. Circ Res. 2012;111(5):564-574.
Regulating IFN-γ. J Immunol. 2009;182(1):234-239. doi:10.4049/jimmunol.182.1.234. doi:10.1161/CIRCRESAHA.112.274597.
8. Chaly Y, Marinov AD, Oxburgh L, Bushnell DS, Hirsch R. FSTL1 promotes arthritis in mice by 27. Yao Y, Jumabay M, Wang A, Boström KI. Matrix Gla protein deficiency causes arteriovenous
enhancing inflammatory cytokine/chemokine expression. Arthritis Rheum. 2012;64(4):1082- malformations in mice. J Clin Invest. 2011;121(8):2993-3004. doi:10.1172/JCI57567.
1088. doi:10.1002/art.33422. 28. Yao Y, Yao J, Radparvar M, et al. Reducing Jagged 1 and 2 levels prevents cerebral arteriovenous
9. Dong Y, Geng Y, Li L, et al. Blocking follistatin-like 1 attenuates bleomycin-induced pulmonary malformations in matrix Gla protein deficiency. Proc Natl Acad Sci U S A. 2013;110(47):19071-
fibrosis in mice. J Exp Med. 2015;212(2):235-252. doi:10.1084/jem.20121878. 19076. doi:10.1073/pnas.1310905110.
10. Liu R, Yang Y, Shen J, et al. Fstl1 is involved in the regulation of radial glial scaffold development. 29. Jain RK. Molecular regulation of vessel maturation. Nat Med. 2003;9(6):685-693. doi:10.1038/
Mol Brain. 2015;8:53. doi:10.1186/s13041-015-0144-8. nm0603-685.
11. Murphy N, Gaynor KU, Rowan SC, et al. Altered Expression of Bone Morphogenetic Protein 30. Schiffrin EL. Endothelin and endothelin antagonists in hypertension. J Hypertens. 1998;16(12 Pt
Accessory Proteins in Murine and Human Pulmonary Fibrosis. Am J Pathol. 2016;186(3):600- 2):1891-1895.
615. doi:10.1016/j.ajpath.2015.10.032. 31. Lee SH, Channick RN. Endothelin antagonism in pulmonary arterial hypertension. Semin Respir
12. Miller M, Beppu A, Rosenthal P, et al. Fstl1 Promotes Asthmatic Airway Remodeling by Crit Care Med. 2005;26(4):402-408. doi:10.1055/s-2005-916155.
Inducing Oncostatin M. J Immunol Baltim Md 1950. 2015;195(8):3546-3556. doi:10.4049/ 32. Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular
jimmunol.1501105. Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle
13. Miller M, Esnault S, Kurten RC, et al. Segmental allergen challenge increases levels of airway Disorders. Pharmacol Rev. 2016;68(2):476-532. doi:10.1124/pr.115.010652.
follistatin-like 1 in patients with asthma. J Allergy Clin Immunol. 2016;138(2):596-599.e4. 33. Wilson JL, Yu J, Taylor L, Polgar P. Hyperplastic Growth of Pulmonary Artery Smooth Muscle Cells
doi:10.1016/j.jaci.2016.01.019. from Subjects with Pulmonary Arterial Hypertension Is Activated through JNK and p38 MAPK.
14. Gangopahyay A, Oran M, Bauer EM, et al. Bone morphogenetic protein receptor II is a novel PLoS ONE. 2015;10(4). doi:10.1371/journal.pone.0123662.
mediator of endothelial nitric-oxide synthase activation. J Biol Chem. 2011;286(38):33134- 34. Noordegraaf AV, Galiè N. The role of the right ventricle in pulmonary arterial hypertension. Eur
33140. doi:10.1074/jbc.M111.274100. Respir Rev. 2011;20(122):243-253. doi:10.1183/09059180.00006511.
15. Xu J, Zhu D, Sonoda S, et al. Over-expression of BMP4 inhibits experimental choroidal 35. NAEIJE R, CHESLER N. PULMONARY CIRCULATION AT EXERCISE. Compr Physiol. 2012;2(1):711-
neovascularization by modulating VEGF and MMP-9. Angiogenesis. 2012;15(2):213-227. 741. doi:10.1002/cphy.c100091.
doi:10.1007/s10456-012-9254-4. 36. Knight D. Epithelium–fibroblast interactions in response to airway inflammation. Immunol Cell
16. Moreno-Miralles I, Ren R, Moser M, Hartnett ME, Patterson C. Bone morphogenetic protein Biol. 2001;79(2):160-164. doi:10.1046/j.1440-1711.2001.00988.x.
endothelial cell precursor-derived regulator regulates retinal angiogenesis in vivo in a mouse 37. Osei ET, Brandsma C-A, Noordhoek JA, Timens W, Postma D, Heijink I. Crosstalk between
model of oxygen-induced retinopathy. Arterioscler Thromb Vasc Biol. 2011;31(10):2216-2222. epithelium and fibroblasts; implications for COPD. Eur Respir J. 2014;44(Suppl 58):P3899.
doi:10.1161/ATVBAHA.111.230235. 38. Ouchi N, Oshima Y, Ohashi K, et al. Follistatin-like 1, a secreted muscle protein, promotes
17. Helbing T, Rothweiler R, Ketterer E, et al. BMP activity controlled by BMPER regulates the endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase-
proinflammatory phenotype of endothelium. Blood. 2011;118(18):5040-5049. doi:10.1182/ dependent mechanism. J Biol Chem. 2008;283(47):32802-32811. doi:10.1074/jbc.M803440200.
blood-2011-03-339762. 39. Shibanuma M, Mashimo J, Mita A, Kuroki T, Nose K. Cloning from a mouse osteoblastic cell line
18. Suzuki Y, Ohga N, Morishita Y, Hida K, Miyazono K, Watabe T. BMP-9 induces proliferation of of a set of transforming-growth-factor-beta 1-regulated genes, one of which seems to encode a
multiple types of endothelial cells in vitro and in vivo. J Cell Sci. 2010;123(Pt 10):1684-1692. follistatin-related polypeptide. Eur J Biochem FEBS. 1993;217(1):13-19.
doi:10.1242/jcs.061556. 40. Morrisey EE, Hogan BLM. Preparing for the first breath: genetic and cellular mechanisms in lung 10
19. Kool H, Mous D, Tibboel D, de Klein A, Rottier RJ. Pulmonary vascular development goes awry in development. Dev Cell. 2010;18(1):8-23. doi:10.1016/j.devcel.2009.12.010.
congenital lung abnormalities. Birth Defects Res Part C Embryo Today Rev. 2014;102(4):343-358. 41. Bellusci S, Henderson R, Winnier G, Oikawa T, Hogan BL. Evidence from normal expression
doi:10.1002/bdrc.21085. and targeted misexpression that bone morphogenetic protein (Bmp-4) plays a role in mouse
20. International PPH Consortium, Lane KB, Machado RD, et al. Heterozygous germline mutations embryonic lung morphogenesis. Dev Camb Engl. 1996;122(6):1693-1702.

192 193
42. Weaver M, Yingling JM, Dunn NR, Bellusci S, Hogan BL. Bmp signaling regulates proximal-distal signaling protects against bleomycin induced pulmonary fibrosis. Respir Res. 2015;16:38.
differentiation of endoderm in mouse lung development. Dev Camb Engl. 1999;126(18):4005- doi:10.1186/s12931-015-0202-x.
4015. 64. Cibois M, Luxardi G, Chevalier B, et al. BMP signalling controls the construction of vertebrate
43. Sountoulidis A, Stavropoulos A, Giaglis S, et al. Activation of the canonical bone morphogenetic mucociliary epithelia. Development. 2015;142(13):2352-2363. doi:10.1242/dev.118679.
protein (BMP) pathway during lung morphogenesis and adult lung tissue repair. PloS One. 65. Tadokoro T, Gao X, Hong CC, Hotten D, Hogan BLM. BMP signaling and cellular dynamics during
2012;7(8):e41460. doi:10.1371/journal.pone.0041460. regeneration of airway epithelium from basal progenitors. Dev Camb Engl. 2016;143(5):764-
44. Ninomiya N, Michiue T, Asashima M, Kurisaki A. BMP signaling regulates the differentiation 773. doi:10.1242/dev.126656.
of mouse embryonic stem cells into lung epithelial cell lineages. In Vitro Cell Dev Biol Anim. 66. Adams D, Larman B, Oxburgh L. Developmental expression of mouse Follistatin-like 1 (Fstl1):
2013;49(3):230-237. doi:10.1007/s11626-013-9589-1. Dynamic regulation during organogenesis of the kidney and lung. Gene Expr Patterns.
45. Huang SXL, Islam MN, O’Neill J, et al. Highly efficient generation of airway and lung epithelial cells 2007;7(4):491-500. doi:10.1016/j.modgep.2006.10.009.
from human pluripotent stem cells. Nat Biotechnol. 2014;32(1):84-91. doi:10.1038/nbt.2754. 67. Siafakas NM, Antoniou KM, Tzortzaki EG. Role of angiogenesis and vascular remodeling in
46. Huang SXL, Green MD, de Carvalho AT, et al. The in vitro generation of lung and airway progenitor chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2007;2(4):453-462.
cells from human pluripotent stem cells. Nat Protoc. 2015;10(3):413-425. doi:10.1038/ 68. Hashimoto M, Tanaka H, Abe S. Quantitative analysis of bronchial wall vascularity in the medium
nprot.2015.023. and small airways of patients with asthma and COPD. Chest. 2005;127(3):965-972. doi:10.1378/
47. Ross AJ, Dailey LA, Brighton LE, Devlin RB. Transcriptional profiling of mucociliary differentiation chest.127.3.965.
in human airway epithelial cells. Am J Respir Cell Mol Biol. 2007;37(2):169-185. doi:10.1165/ 69. Morty RE, Nejman B, Kwapiszewska G, et al. Dysregulated bone morphogenetic protein signaling
rcmb.2006-0466OC. in monocrotaline-induced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol.
48. Fahy JV, Dickey BF. Airway Mucus Function and Dysfunction. N Engl J Med. 2010;363(23):2233- 2007;27(5):1072-1078. doi:10.1161/ATVBAHA.107.141200.
2247. doi:10.1056/NEJMra0910061. 70. Takahashi K, Kogaki S, Matsushita T, Nasuno S, Kurotobi S, Ozono K. Hypoxia induces alteration
49. Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. Basal cells are a multipotent progenitor of bone morphogenetic protein receptor signaling in pulmonary artery endothelial cell. Pediatr
capable of renewing the bronchial epithelium. Am J Pathol. 2004;164(2):577-588. doi:10.1016/ Res. 2007;61(4):392-397. doi:10.1203/pdr.0b013e3180332cba.
S0002-9440(10)63147-1. 71. Long L, Crosby A, Yang X, et al. Altered Bone Morphogenetic Protein and Transforming Growth
50. Rock JR, Onaitis MW, Rawlins EL, et al. Basal cells as stem cells of the mouse trachea and Factor-β Signaling in Rat Models of Pulmonary Hypertension. Circulation. 2009;119(4):566-576.
human airway epithelium. Proc Natl Acad Sci U S A. 2009;106(31):12771-12775. doi:10.1073/ doi:10.1161/CIRCULATIONAHA.108.821504.
pnas.0906850106. 72. Cahill E, Rowan SC, Sands M, et al. The pathophysiological basis of chronic hypoxic pulmonary
51. Kimura J, Deutsch GH. Key mechanisms of early lung development. Pediatr Dev Pathol Off J Soc hypertension in the mouse: vasoconstrictor and structural mechanisms contribute equally. Exp
Pediatr Pathol Paediatr Pathol Soc. 2007;10(5):335-347. doi:10.2350/07-06-0290.1. Physiol. 2012;97(6):796-806. doi:10.1113/expphysiol.2012.065474.
52. Rawlins EL, Hogan BLM. Epithelial stem cells of the lung: privileged few or opportunities for
many? Dev Camb Engl. 2006;133(13):2455-2465. doi:10.1242/dev.02407.
53. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell
populations. Nat Med. 2014;20(8):822-832. doi:10.1038/nm.3642.
54. Schamberger AC, Staab-Weijnitz CA, Mise-Racek N, Eickelberg O. Cigarette smoke alters primary
human bronchial epithelial cell differentiation at the air-liquid interface. Sci Rep. 2015;5.
doi:10.1038/srep08163.
55. Guha A, Vasconcelos M, Cai Y, et al. Neuroepithelial body microenvironment is a niche
for a distinct subset of Clara-like precursors in the developing airways. Proc Natl Acad Sci.
2012;109(31):12592-12597. doi:10.1073/pnas.1204710109.
56. Barnes PJ. CLub cells, their secretory protein, and copd. Chest. 2015;147(6):1447-1448.
doi:10.1378/chest.14-3171.
57. Bernard A, Marchandise FX, Depelchin S, Lauwerys R, Sibille Y. Clara cell protein in serum and
bronchoalveolar lavage. Eur Respir J. 1992;5(10):1231-1238.
58. Vestbo J, Edwards LD, Scanlon PD, et al. Changes in Forced Expiratory Volume in 1 Second over
Time in COPD. N Engl J Med. 2011;365(13):1184-1192. doi:10.1056/NEJMoa1105482.
59. Park HY, Churg A, Wright JL, et al. Club cell protein 16 and disease progression in chronic
obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(12):1413-1419.
doi:10.1164/rccm.201305-0892OC.
60. Lomas DA, Silverman EK, Edwards LD, et al. Evaluation of serum CC-16 as a biomarker for COPD
in the ECLIPSE cohort. Thorax. 2008;63(12):1058-1063. doi:10.1136/thx.2008.102574.
61. Guerra S, Halonen M, Vasquez MM, et al. The relation of circulating CC16 to lung function growth,
decline, and development of COPD across the lifespan. Lancet Respir Med. 2015;3(8):613-620.
doi:10.1016/S2213-2600(15)00196-4. 10
62. Myllärniemi M, Lindholm P, Ryynänen MJ, et al. Gremlin-mediated decrease in bone
morphogenetic protein signaling promotes pulmonary fibrosis. Am J Respir Crit Care Med.
2008;177(3):321-329. doi:10.1164/rccm.200706-945OC.
63. De Langhe E, Cailotto F, De Vooght V, et al. Enhanced endogenous bone morphogenetic protein

194 195
ACKNOWLEDGEMENT
Acknowledgement for your insightful input in study design, critical feedback on the manuscripts,
and also kindly providing COPD patient materials that were very valuable to my
First of all, I would like to sincerely thank everyone who has made any contributions study. Andrew Halayko provided me with very valuable feedback on epithelial-
to the Fstl1 project and supported me through the process of starting up, during, mesenchymal communication manuscript. Pieter Hiemstra, thank you for critically
and finishing this long unforgettable journey. These collective supports have made reading and providing constructive feedback on the epithelial cell differentiation
the completion of my thesis possible. As it is rather impossible to mention everyone manuscript. Thank you Luis Jesus Jimenez-Borreguero for your insightful knowledge
individually, I apologize if I did not mention your name. For the following persons, I on cardiopulmonary physiological functions which was very valuable for the Fstl1
would like to specifically address my great appreciation. knockout manuscript. I am so grateful to have such experts to exchange ideas and
discuss the data with.
Most of all I am fully indebted to my primary promotor, Reinoud Gosens, for your
constructive feedback on the study design, experimental planning, manuscripts Thank you Sophie Bos for traveling with me all the way to Amsterdam to isolate the
and thesis finalization by giving me the necessary freedom to grow and develop lungs of newborn mouse pups and generating human COPD lung slices, in addition
scientifically and personally. You taught me by example on how to be a good future to teaching me how to paraffin-embed and slice the lung tissue. I wish to thank
leader. Thank you for your continuous supports and advice throughout my PhD. I am Marjan Luinge for your assistance in using the pressure cooker at the pathology lab.
glad and very grateful to work with you. Also, thank you for your quick response in
finalizing the manuscripts and this thesis. Thank you so much, Reinoud! Thank you Herman Meurs for your hospitality every time I joined your car to go to
conferences or a lab outing. You are extremely humble and your diverse interests
I am grateful for the full support of my promotor Martina Schmidt in the last four beyond science are so inspiring. You even traveled around Indonesia more than I have
years, especially in pursuing pharmacological education in England. Thank you for done. I enjoyed talking to you about your experience in different parts of Indonesia
giving me the opportunity to work on the Fstl1 project and for being very supportive and even successfully convinced me that Indonesia has a distinct smell. Thank you
in finalizing this thesis by providing me with feedback and suggestions. Amalia Dolga for several nice discussions we had about exciting experimental design
and some great advice on future career development.
I would also like to express my gratitude to my co-promoter in Florida, USA, Harm
Maarsingh for the opportunity to work on the Fstl1 project. Thank you for your My gratitude also extends to Marc Sylva for sharing your extensive knowledge and
insightful advice in study design and thorough feedback in data analysis, data experience in studying Fstl1, Andrea Mattiotti for your support in managing the Fstl1
interpretation, and critically reviewing this thesis as well as abstracts and posters knockout breeding line and fruitful discussions on how to analyze and interpret the
for conferences. Without your support, finalizing this thesis would not be feasible. data, to Stuti Prakash for collecting functional data in Madrid that are essential for
I really appreciate your time and efforts to maintain good communication through the publication and to Quinn Gunst for technical support in in situ hybridization
Skype meetings in the past four years. which were very valuable for the revision of the Fstl1 knockout manuscript. The
annual Groningen-Amsterdam Fstl1 meeting would not be successful without all of
To the Fstl1 expert in the heart, Maurice van den Hoff, I am extremely grateful your active participation and contributions. Thank you!
for your guidance and suggestions throughout my project, especially in the study
design and experimental plan of the Fstl1 knockout animal study. Thank you for Many thanks to the master students who have been working with me on this
your constructive feedback on the manuscripts and for giving me the opportunity Fstl1 project. Ken Ho Hua, you impressed me with your final report and great
to learn PCR data analysis directly from the expert, the LinReg software developer, understanding on Fstl1 pathways in different systems, Jorinke Koops, thank you for
Jan Ruijter. I thank Jan for the opportunity to participate in the intensive workshop all your hard work in optimizing the immunostaining before you went New castle,
on advanced real time PCR data analysis and for your guidance on selecting proper Australia. You were a quick learner and able to be independent quite quickly. This
reference genes. was great when I could not be physically present for a couple of weeks as I had to
recover from surgery. Vanessa Niburg, thank you for your continuous enthusiasm
Besides my promotors, I would like to thank the assessment committee, Frank Kruyt, and your persistence in finishing the ELISA experiments. You were a very well
Gert Folkerts, and Colin Bingle for critically reviewing and evaluating my thesis. organized person and fun to be around. Furthermore, thank you to Frank Ensink,
I wish to acknowledge the support provided by Wim Timens. I learned a lot from a humorous and motivated student, for your hard work in running lots of western
you about ‘’reading’’ the human lung pathology and vessel structure. Thank you blots. I always appreciate your honesty and enjoy exchanging new ideas and creative
experiments with you. Also, to the bachelor students Sefike Vardar, Rukiye Çiçek,

198 199
Barzo Sulaiman, Rosa Kortekaas, and Nuhoda Aldarij, thank you for all your hard Tijtske Oenema, Hana Cernecka, and Kuldeep Kumawat, thank you for your interest
work and dedication to the project. I hope all of you had a great time and learn a lot to get taste of Indonesian cultures by coming to watch my stage performance at the
during your internship. Indonesian day! And together with David Wright, we explored the Dutch island,
cycling around Terschelling during the very rare Dutch summer. That was my first
I would like to thank the following (ex)-colleagues for their kindly support, help, and time swimming with jellyfish in Dutch water! Thank you Mark Menzen for all the fun
for all the fun we have had in the last four years. A special thanks to my roommate, you brought every day, making lab life less serious. Since you left, the lab was never
Tim Koopmans, Timmy, thank you for being around, and for being you. You saw me the same.
laugh crazily or cry quietly. Thanks for all your support and being my ‘’living stress
ball’’. I will never forget when we had tapas with Pablo Munoz, encouraging me to Thanks to my friends for making my life worth living, more colorful, and never letting
keep going while I was down. And of course to our many, many fun times throughout me give up. Christel Seegers and Thea van den Bosch, I am glad to have you as
my PhD. Without you guys, I would not be writing this acknowledgment. Thanks a my friends and so grateful to have you as my paranymphs. Thank you for helping
bunch! I wish both of you good luck with your career and I look forward to reading me with the Dutch summary for this thesis. Together with Maureen Xu, we always
your publications in Science, Cell or Nature! Haoxiao Zuo, I secretly wished you would made time for girls night out, thank you for all the fun times we shared, for coming
not need to go to Germany every now and then, just stay in our office in Groningen to my house often and willing to try my experimental cooking without any doubt.
to add more woman power ;) Thanks to you and Bing Han for cooking local Chinese Maybe because I did not tell you they were experimental dishes ;) I could not expect
food every now and then, which really reminds me of my grandparents’ cooking. to have better friends than you all. It was so difficult to say good-bye when I moved
Also thanks Bing for helping me translate a Chinese abstract about Fstl1. I thank to Seattle but who knows, I might come back to the Netherlands one day. In the
John-Poul Ng-Blichfeldt for the inspiring discussions about epithelial differentiation. meantime, we will stay in touch.
If only you came back to Groningen earlier, I would probably have more time to do
some follow-up experiments. Thank you to my topmaster fellows, Vijay Anand and Neeraj Kumar for our many
great times together. We have known each other for as long as I moved to Groningen
Pavan Prabhala, thank you for many fun times, which included walking around and we have been through many ups and downs, as foreigners in Groningen,
Groningen to catch Pokémon. We caught more than 50 Pokémon in a day! Thanks experiencing all the new stuff for the first time. Tomas Kloosterman, you are very
to Anita Spanjer and Loes Kistemaker for teaching me how to isolate primary talented not only in science but also in music. Thank you for inviting me to your
human epithelial cells as well as perform ALI cultures, and to Eline van Dijk, thanks piano concerts, sharing your creative homemade salad recipes, and not to mention
for sharing primary human fibroblasts. Although it was rather short, I had shared for fixing my bike. I am so glad that you found the love of your life, Angelique
several nice memories with Mariska van den Berg, Inge Krabbendam and Xin Hui Kloosterman that also shares the same interest in science and music. Thanks Johana
Wu, especially during intensive preparation for Herman’s farewell party where the Isaza Correa for visiting me in Groningen, I hope everything goes well with your PhD
three of you practiced your music talents in my office. Thank you for all the sweet as well as personal life in Ireland. I missed our great time together with Karolina
moments we shared and I wish all of you to enjoy and get the most out of your PhD Szczrba during summer in Groningen. I hope we can meet again one day either in
experience. I thank Annet Zuidhof for the technical assistance in showing me how to Europe or in the US. Sunil Lee and Smitha Chandran, thank you for your hospitality
do immunohistochemistry for the first time. Thanks Carolina Elzinga for empowering and introducing me to authentic Indian foods. I would never forget the day that you
discussions that had given me a new perspective. Without it, I would not be able to had to fly to India in the early morning, when Katarzyna Butler and I had to catch
finish my thesis. Thanks Sepp Jansen for inspiring advice on writing and finishing a the train carrying all your big suitcases at the very last minute. It was crazy but fun!
thesis abroad, which I kept in mind while writing this thesis in Seattle.
Thank you Valentine Kheng for making the world seems so small by keeping in touch
Thank you Janneke Fekkes-Koolman, for your kindly help in sending the hard copy no matter where we are by visiting each other in different parts of the world. Thank
of my thesis to the reading committee member in England, in extending my RuG you for visiting me in the Netherlands and I was so glad to visit you in New York. I
email that enables me to submit my thesis, and in arranging the reimbursement of am so grateful to have such amazing friends like you and Rama “Radut” Dhenni!
travel expenses for the conferences and courses in the past four years. I would like Thanks for your continuous support. Dut, I really appreciate your time and effort for
to also thank you in advance for being very helpful in supporting my paranymphs critically reading and reviewing my Indonesian summary for this thesis.
to distribute the thesis. I would be lost without your help. Jacques Hille, you were
always caring about my wellbeing and progress in my thesis writing. I have greatly Robert Woudstra, thank you for kidnapping Wilfred for diving weekends, allowing
appreciated this, thank you so much for your concerns. me to have peaceful ME time. Also thank you for traveling to my hometown, Bandung

200 201
to be present at our wedding in Indonesia and to be the first person to visit us in PhD and about to move to Seattle. I am glad that you will come to my defense. Thank
Seattle! I hope you had great time in the US! you for countless empowering discussions we had and for designing my beautiful
artsy cover that turned out to be better than my imagination!
To the (former) members of Indonesian student association in Groningen (PPI
Groningen), thank you for organizing many events where I could speak Indonesian My parents, mama and papa, thank you for raising me and providing me with loving
and contribute to promote Indonesian cultures. Through this organization also I and caring environment, which allowed me to take a risky and difficult path that
met many PhD fellows from diverse backgrounds. I apologize I could not mention no one in our family had yet pursued, from a bachelor in science to a master study
everyone but I would like to thank all for all your support during my stay in abroad, until my PhD. You never doubted my choices, instead encouraging me to
Groningen. A special thanks to Awalia Febriana and Astri Ferdiana, for being such keep going, which eventually resulted in this thesis. Time difference and distance
inspiring role models, to Surahyo Sumarsono for reviewing and providing feedback have never been a problem to maintain our good communication, thank you for
for the Indonesian summary of this thesis. Thanks a bunch Daniel, words fall too your continuous support! Your support has helped me to persevere and go through
short to describe my gratitude for your kindly help and support in my first two years many difficult circumstances. Thanks my brother, Herren Calida for visiting me in
in Groningen. Glad to hear that you also finishing your PhD. I am convinced that Groningen, soon you will graduate and apply your business skills in to practice. Best
you will get a postdoctoral position soon. Rieza Aprianto, we have known each of luck! Thanks to my brother, Hanel Topada and his lovely wife, my sister-in-law
other since we were at ITB, you probably know me as ‘’mahasiswa nyasar’’ as I was Sylvia T. Gunawan for taking good care of our parents while I am so far, far away.
the only person from Microbiology that took Pharmacy courses. And later, to our Thanks Nel for introducing me to the world of investment and for being my personal
surprise, we ended up together again in Groningen. I wish you success to finalize financial advisor.
your thesis and good luck for your future career in the Netherlands! MacKenzie
Hadi, ‘who does not know Kenzi’, that was the first comment I heard from others My parents-in-law, my mem, Janke Huitema, thank you for teaching me how to be
before I met you. After I met you, I know why. Thank you for being very entertaining. creative in sewing and knitting. I am so grateful to have you because I know I can
Groningen has never been the same since you moved to Leiden. Amirah A. Alkaff, always be myself and come to you for support in the Netherlands. Thank you my
Putri D. Utari, and Doti Parameswari, thank you for all the fun we shared together heit, Marten Poppinga for accompanying me to get my surgery in Assen as well as
during the preparation of the Indonesian day every year as well as for your kindly for helping me moving in and out of 3 different houses, until we finally moved to
support in the past four years. Seattle. With heit and mem, I had lots of memorable time in Germany, Belgium and
all around the Netherlands. I would also like to extend a thank you to Alexander
Thank you the Marmalade gang, for all your mental support throughout my PhD and and Anje Poppinga for your hospitality and giving me my first adorable nephews in
being my friends in the last 13 years, no matter where you are in the world. Thank the Netherlands, Jurryt and Thymen. Thank you Avelien Huitema, for visiting me
you Mega Arfindari, Amalia Muntiyara, and Dessy Dwiyanti for always making time in Groningen multiple times. Congratulations on finishing your studies and taking
to meet up every time I visit Indonesia. To Ikha Mansoora and Maya Fitriyanti, we a bold move to look for your dream job! Marijke Huitema and Nicola Williams, it
finally live in the same country again. We definitely will catch up more often. I hope felt like yesterday that I came to your wedding in Belgium, instead it has been four
to visit you in South Korea soon, Anggita Karleshya. years. Time goes so fast! Thank you for visiting me whenever you came back to the
Netherlands. I am so glad to hear that both of you have settled down now in Ghent.
I would like to thank my cousin, Joe Wimpi Manikam, who helped me out since
my final bachelor project at the Pharmaceutical Biotechnology laboratory at ITB. Last but not least, thanks to Wilfred J. Poppinga for being such a sweet and loving
Without your help as a medical doctor, I would not be able to perform the functional husband, for your non-stop support and faith in me. Thank you for always taking
assay to test the enzyme activity I worked on in 2008. For my PhD, you were again good care of me, being an antidote of my stress, giving me all the freedom I wish
very helpful for inspiring discussions on the plausible cause of death of the Fstl1 for, and much more. No wonder I married you 3 times! We have been through many
knockout mice. You are very talented and knowledgeable and I know I can always challenges together in the past 5 years, including living on different continents twice.
come to you when I need clinical advice and medical doctor support. I hope we will never do it again. I am very happy to share my life with you. Words
could never be enough to express my gratitude to have you as my life partner.
A special thanks to Putri A. Wulandari, there are not enough words to thank you
enough. We have known each other for 18 years, you are like a sister to me. You Navessa P. Tania
have been my living witness of my long journey in pursuing my ambition. Thank you Seattle, WA
for visiting me when I just moved to Groningen and again when I was finishing up my June 2017

202 203
1

BIOGRAPHY
AND
LIST OF PUBLICATIONS

205
About the author
Navessa Padma Tania was born in Bandung, Indonesia on April 12nd in 1986. After
Published articles
finishing her final project at the Department of Pharmaceutical Biotechnology,
she obtained her bachelor degree in Microbiology at the Bandung Institute of N.P. Tania, H. Maarsingh, I. S. T. Bos, A. Mattiotti, S. Prakash, W. Timens, Q. D. Gunst, L.
Technology, Indonesia in 2008. Upon graduation, she worked as a research assistant J. Jimenez-Borreguero, M. Schmidt, M. J.B. van den Hoff, R. Gosens. Endothelial follistatin-
at the Mochtar Riady Institute for Nanotechnology. like-1 regulates the postnatal development of the pulmonary vasculature by modulating
In 2010, she was awarded with a talent grant from the Groningen Biomolecular BMP/Smad signaling. Pulm Circ. 2017;7(1) 219–231.
Sciences and Biotechnology Institute, to participate in a 2-year Topmaster program in
N.P. Tania, M. Schmidt, R. Gosens. Activin-A: active in inflammation in COPD. Eur Respir J.
Biomolecular Sciences at the University of Groningen, The Netherlands. During her
2014 Apr;43(4):954-5.
master studies, she developed her scientific skills at the Department of Molecular
Genetics, and she participated in the 2011 international Genetically Engineered H. Alkhouri, W.J. Poppinga, N.P. Tania, A. Ammit, M. Schuliga. Regulation of pulmonary
Machine (iGEM) competition in synthetic biology at the Massachusetts Institute of inflammation by mesenchymal cells.Pulm Pharmacol Ther. 2014;29(2):156-65.
Technology, USA. As a final master project, she performed a collaborative project
W.W. Wang, S.F. Ang, R. Kumar, C. Heah, A. Utama, N.P. Tania, H. Li, S.H. Tan, D. Poo, S.P.
at the University Medical Center Groningen between the Department of Medical
Choo, H.C. Toh. Identification of Serum Monocyte Chemoattractant Protein-1 and Prolactin
Oncology and the Department of Molecular Pharmacology. as Potential Tumor Markers in Hepatocellular Carcinoma. PLoS One. 2013, 8(7): e68904.
Upon completion of her master degree, she started her PhD studies at the
Department of Molecular Pharmacology, at the University of Groningen in close A. Utama, N.P. Tania, R. Dhenni, R.A. Gani, I. Hasan, A. Sanityoso, S.A. Lelosutan, R.
collaboration with the Academic Medical Center, Amsterdam. In 2015, she won an Martamala, L.A. Lesmana, A. Sulaiman, S. Tai. Genotype diversity of hepatitis C virus (HCV) in
abstract award from American Thoracic Society in Denver, USA. Her research focused HCV-associated liver disease patients in Indonesia. Liver Int. 2010, 30(8):1152-60.
on the roles of Follistatin-like 1 in the lung is presented in this thesis.

Published abstracts

N.P. Tania, R. Gosens, S. T. Bos, W. Timens, M. Schmidt, M. van den Hoff, H. Maarsingh.
Endothelial Follistatin-like 1 regulates the maturation of the pulmonary vasculature by
modulating BMP/Smad signaling. The FASEB Journal, 2016, 30 (1): 300.1.

N.P. Tania, R. Gosens, I.S.T. Bos, A. Mattiotti, S. Prakash, W. Timens, Q.D. Gunst, L.J. Jimenez-
Borreguero, M. Schmidt, M.J.B. van den Hoff, H. Maarsingh. Endothelial Follistatin-like-1 is
crucial for normal development of the pulmonary vasculature in mice. Am J Respir Crit Care
Med. 2015, 191: A5960.

N.P. Tania, R. Gosens, W. Timens, M. Schmidt, M. van den Hoff, H. Maarsingh. Follistatin-like
1 regulates inflammatory cytokine release from lung fibroblasts and airway smooth muscle
cells. Am J Respir Crit Care Med. 2014; 189:A5314.

206 207

S-ar putea să vă placă și