You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/225061505

Cellular Players in Lung Fibrosis

Article  in  Current Pharmaceutical Design · May 2012


DOI: 10.2174/138161212802430396 · Source: PubMed

CITATIONS READS
49 181

4 authors, including:

Annemarie Lekkerkerker Jamil Aarbiou


Galapagos NV Charles River Laboratories International, Inc
15 PUBLICATIONS   516 CITATIONS    40 PUBLICATIONS   1,515 CITATIONS   

SEE PROFILE SEE PROFILE

Richard Janssen
Galapagos NV
64 PUBLICATIONS   1,729 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Cytoskeletal biology View project

Drug discovery View project

All content following this page was uploaded by Richard Janssen on 07 March 2018.

The user has requested enhancement of the downloaded file.


Current Pharmaceutical Design, 2012, 18, 4093-4102 4093

Cellular Players in Lung Fibrosis

Annemarie N. Lekkerkerker, Jamil Aarbiou, Thomas van Es, Richard A.J. Janssen*

Galapagos BV, P.O. Box 127, 2300 AC Leiden, The Netherlands

Abstract: Pathogenic mechanisms involved in fibrosis of various organs share many common features. Myofibroblasts are thought to
play a major role in fibrosis through excessive deposition of extracellular matrix during wound healing processes. Myofibroblasts are ob-
served in fibrotic lesions, and whereas these derive from the hepatic stellate cells in liver, in lung they appear to originate from fibro-
blasts. The source of these fibroblasts has been the object of numerous studies over the recent years and points towards multiple sources.
First of all, resident fibroblasts are thought to differentiate into the more contractile myofibroblasts, secreting many extracellular matrix
proteins. Secondly, the epithelial to mesenchymal transition (EMT) of epithelial cells may also account for increased numbers of fibro-
blasts, though in vivo evidence in patient tissue.is still scarce. Thirdly, the enigmatic fibrocytes, stemming from the bone marrow, may
also account for increasing numbers of fibroblasts in fibrotic lesions. These pathogenic processes are further augmented by the generation
of so-called alternatively activated macrophages, which have direct and indirect effects on myofibroblast accumulation and collagen
deposition. TGF, which is produced predominantly by macrophages, plays a central role in all these processes by inducing EMT, driv-
ing differentiation of fibrocytes, and differentiation towards myofibroblasts.
This review describes the potential origins and roles of these fibrotic cells in the lung and discusses models to study these cells in vitro.
These models offer innovative approaches in target and drug discovery, aiming to uncover novel therapeutic targets that regulate the pro-
fibrotic phenotype of these cells.
Keywords: Fibrosis, IPF, EMT, epithelial cell, fibroblasts, fibrocytes, macrophages, TGF.

INTRODUCTION inflammatory and repair responses. Interestingly, hepatocytes and


There is considerable overlap between liver fibrosis and lung biliary epithelial cells in the liver [1, 2] and epithelial cells in the
fibrosis. Both diseases most likely originate from damage to the lung may contribute to fibrosis through the transition of epithelial
organ. While in the liver this may be due to viral infection, toxic cells into mesenchymal cells (epithelial mesenchymal transition,
compounds, or metabolic stress, in the lung the damage may be due EMT). These mesenchymal cells can then transdifferentiate into
to viral or bacterial infection, smoking, or exposure to allergens. In (myo)fibroblasts.
both tissues, the damage induces a cascade of cellular responses and There is strong evidence that activated fibroblasts and myofi-
molecular processes, trying to control the damage, to fight the in- broblasts play an important role in fibrosis. Myofibroblasts are
fection, and to repair the damaged tissue. In most instances, these contractile cells expressing -smooth muscle actin (SMA). These
processes are well controlled leading to proper healing without any cells contribute to excessive wound healing and to excessive depo-
pathological problems. In some cases, the repair process goes awry sition of extracellular matrix proteins (fibronectin, collagen,
and the immune and repair response overreact, leading to inflam- laminin). The source of these cells is the focus of intense research.
mation, uncontrolled apoptosis, improper tissue remodeling, and Several sources of fibroblasts have been put forward, including
excessive wound healing. proliferation of resident fibroblasts, generation of fibroblasts from
Fibrotic interstitial lung diseases are characterized by progres- epithelial cells through epithelial mesenchymal transition, and dif-
sive decline in lung function and premature death from respiratory ferentiation of circulating fibrocytes or mesenchymal progenitor
failure and include amongst others cystic fibrosis and familial pul- cells.
monary fibrosis. In this review we focus mostly on idiopathic pul- Furthermore, current studies indicate that macrophages as well
monary fibrosis (IPF), a rare disease with an estimated prevalence as fibrocytes are involved in disease progression by mediating fi-
of 14–43 per 100,000 people, depending on the case definition of broblast and myofibroblast activation. TGF, derived from macro-
IPF. It is a progressive disorder that culminates in premature death phages and epithelial cells, plays an important role in all these
from respiratory failure and in which no treatment intervention has processes. Below we will discuss the role of the various cell types
been effective to date. The only treatment of proven benefit is lung and the contribution of TGF in the various pro-fibrotic processes.
transplantation. IPF is characterized by progressive scarring of the The focus of this review is predominantly on the (de)differentiation
lung parenchyma, leading to areas of inflammation, excessive col- processes of the various cells and the profibrotic factors these cells
lagen deposition and fibroblast proliferation (fibroblastic foci), produce in IPF. (Fig. 1) gives an overview of the various cells dis-
adjacent to areas of normal, uninjured, and seemingly uninvolved cussed in this paper and how they contribute to the generation of
lung. While normal wound healing involves signals to terminate the (myo)fibroblasts.
accumulation of fibroblasts and the deposition of collagen, in IPF
this process fails. Epithelium of the Lung
In both liver and lung a similar cascade of events is induced The airway epithelium covers the inner walls of the airways and
leading to fibrosis. The damaged hepatocytes or lung epithelial cells serves as the first line of defense against potentially hazardous envi-
lose their protective function, including detoxification of the hepa- ronmental factors and microorganisms. We will first discuss the
tocytes and barrier function of the lung epithelium. As a result, physiological function of lung epithelium followed by its role in
toxic or microbial insults accumulate and exacerbate the fibrosis. The epithelium forms a pseudo stratified cell layer com-
posed of different epithelial cell types. These include basal, ciliated
and goblet cells that form the core of the epithelial layers in trachea,
*Address correspondence to this author at the Galapagos BV, P.O. Box 127, as well as large and small airway. Basal cells are predominantly
2300 AC Leiden, The Netherlands; Tel: +31-717506725;
E-mail: Richard.Janssen@glpg.com
located at the basolateral side and are considered as a pool of pro-

1873-4286/12 $58.00+.00 © 2012 Bentham Science Publishers


4094 Current Pharmaceutical Design, 2012, Vol. 18, No. 27 Lekkerkerker et al.

EMT Myofibroblast transition

A
Epithelial cells Fibroblasts Myofibroblasts

Fibrocyte to fibroblast differentiation Myofibroblast transition

B
Fibrocytes

Myofibroblast transition

Fig. (1). Overview of the various cell types in the lung that contribute to fibrosis through the generation of myofibroblasts. 1A: epithelial mesenchymal transi-
tion. 1B: fibrocyte differentiation. 1C: fibroblast to myofibroblast conversion.

genitor cells that proliferate and subsequently can differentiate into that protective measures of the airway epithelium are quite effec-
various epithelial cell types and thereby restore the composition and tive. Indeed the airway epithelium has a big arsenal of antimicrobial
function of the epithelial layer. Goblet cells contain large vacuoles defense mechanisms. Upon encountering potential invaders, the
filled with mucus. This mucus primarily consists of large glycopro- airway epithelium reacts by producing and releasing various factors
teins, called mucins that form a sticky substance. The mucus is that help neutralize the invader. The epithelial cells express various
secreted from the vacuoles into the lumen where it traps inhaled pattern recognition receptors, such as Toll-like receptors (TLRs),
noxious agents. that recognize and bind a variety of viruses and bacteria [6]. The
Ciliated cells are characterized by the presence of a brush of interaction of pathogens exerts a cascade of signaling pathways,
cilia that move in a coordinated fashion (ciliary beat). In this way, leading to the production of numerous antimicrobial peptides and
exogenous agents that are trapped in the mucus, are actively trans- proteins, such as defensins, SLPI, elafin, lysozyme and lactoferrin
ported out of the lungs. The more distally located small airways [7]. These predominantly cationic molecules show a broad spec-
(terminal bronchioli) also harbor so called Clara cells. These cells trum antibiotic capacity and are considered to act as the first line of
are characterized by the expression of Clara cell secretory protein defense, while innate, and at a later stage adaptive immune cells,
(CCSP, also designated CC10) and are thought to have diverse are recruited. The airway epithelium plays a prominent role in the
functions in lung homeostasis including roles in xenobiotic metabo- recruitment of innate immune cells such as neutrophils, mono-
lism, immune system regulation, and progenitor cell activity [3]. cytes/macrophages, and eosinophils. They produce CXCL8 (IL-8),
Like basal cells, Clara cells are believed to act as progenitors for CXCL1 (Gro-), and CXCL5 (ENA-78) to attract neutrophils [8-
regeneration of the airway epithelium upon injury [4, 5]. 10] to the site of inflammation, while monocyte and macrophage
recruitment is induced by the release of IL-1, CCL3 (MIP-1),
The airways form an immense structure of branching ducts, CCL2 (MCP-1), and TNF- [6, 9, 11]. Epithelial-derived IL-5,
ending up in the alveolar spaces separated by alveolar septa. The GM-CSF, CCL11 (eotaxin-1), CCL24 (eotaxin-2), and CCL5
alveolar septa consist of a thin single cell wall primarily formed by (RANTES) attract eosinophils [12].
two cell types, alveolar type I and type II cells. The branching and
compartmentalization of the alveolar spaces provides an enormous Airway epithelial cells also induce migration of adaptive im-
area enlargement where gas exchange takes place between the lu- mune cells (such as dendritic cells and T cells) into mucosae by
men and capillary vessels in the alveolar septa. producing chemokines. These include Th1 chemokines CXCL9
(Mig), CXCL10 (IP-10) and CXCL11 (I-TAC), Th2 chemokines
Another way the epithelium provides protection is by its barrier CCL1 (I-309) CCL17 (TARC) and CCL22 (MDC), as well as DC
function. The different cell types that comprise the epithelial layer chemokines CCL20 (MIP-3) [9, 13].
form so called tight junctions that restrict free entrance of exoge-
nous agents through the epithelial layer. These tight junctions con- Besides recruitment of inflammatory cells, the airway epithe-
sist of various intercellular proteins formed by adjacent cells, lium also helps regulate the inflammatory response by producing
thereby creating an epithelial layer that only allows active transport several pro- and anti-inflammatory cytokines, such as interferons
of molecules through the cells. Invasion of the subepithelial tissue (IFN), TNF, IL-1ß, and other interleukins. These cytokines help
by inhaled microorganisms may thereby only take place when the balance the inflammatory response in such a way that it is adequate
epithelial integrity is disturbed, e.g. by injury, or when microorgan- enough to eradicate the pathogens without any serious damage to
isms such as rhinoviruses and respiratory syncytial viruses have the surrounding tissue [6]. Although the inflammatory response is
adapted in such a way that they can enter epithelial cells. tightly regulated in healthy individuals, in certain disease states the
balance is disturbed, leading to an excess production of (pro)-
The fact that our lungs are not continuously invaded, while we inflammatory mediators and subsequently to tissue injury, accom-
are constantly exposed to these microorganisms, already indicates panied by a fibrotic process.
Cellular Players in Lung Fibrosis Current Pharmaceutical Design, 2012, Vol. 18, No. 27 4095

Epithelium and Fibrosis the fields of lung, kidney, and liver fibrosis. Using in vivo tracking
Lung fibrosis is a complex physiologic process, initiated upon studies, conflicting results have been reported for EMT in kidney
lung injury, and is the result of repair processes gone awry. It in- fibrosis. Early studies delivered evidence for a crucial role of EMT
volves massive deposition of matrix by an expanded pool of fibro- in renal fibrogenesis [25-27]. Later studies in kidney showed that
genic cells, disruption of the normal tissue architecture, and paren- there is no strong evidence for EMT contributing to the formation
chymal destruction. of interstitial myofibroblasts and that interstitial pericytes may have
a more important role [28, 29]. However, in an elegant approach,
It was long thought that the pool of fibrogenic cells, that are Kim et al. demonstrated that in mice alveolar epithelial cells, ex-
responsible for the fibrotic process, is solely derived from migrating pressing transgenic lung specific -galactosidase as marker, ac-
and subsequently expanding resident fibroblasts in the mesen- quired a mesenchymal phenotype when the animals were exposed
chyme. Recent findings however, prove that other cell types may to adenovirally expressed TGF. The cells acquired vimentin and
also contribute to this pool. Amongst these, epithelial cells have SMA and were localized in fibronectin rich areas [30]. This dem-
been shown to undergo profibrotic phenotypic changes, referred to onstrates that fibroblasts indeed can be derived from epithelial cells
as epithelial-mesenchymal transition. through EMT and may contribute to fibrosis. Interestingly, alveolar
Epithelial-mesenchymal Transition type II cells derived from IPF patients express higher levels of
EMT was already described in the early 1980s [14], constituting mRNA for profibrotic proteins and when cultured on fibronectin
part of embryonic development and organogenesis, and was later express SMA and vimentin in a TGF-dependent manner [31].
confirmed in studies showing that EMT contributes to the formation Other Pro-fibrotic Effects of Lung Epithelium
of the mesoderm and the neural crest [15, 16]. EMT is a process in The contribution of lung epithelial cells is not restricted to their
which epithelial cells lose their epithelial phenotype, acquire fibro- property to undergo EMT. Upon challenge, epithelial cells produce
blast-like properties, and display reduced cell adhesion and in- many factors that either have a direct or indirect effect on the fi-
creased motility. The reduction of adhesion molecules allows the brotic process. Warshamana et al. demonstrated that isolated mur-
cells to detach from the epithelial layer and migrate towards the site ine bronchiolar-alveolar epithelial cells produce factors including
of injury or inflammation where they exercise their profibrotic ef- platelet-derived growth factor (PDGF) isoforms A and B and TGFß
fects. EMT has gained quite some interest in the past few years as a [32]. Other studies have confirmed the expression of TGFß in lung-
potential contributor to fibrotic diseases and has been described in derived epithelial cells [33, 34]. Here, we will discuss how these
various organs. In vitro studies, performed in primary human bron- various factors, that are produced by epithelial cells, contribute to
chial epithelial cells (HBEC) and primary alveolar epithelial cells fibrosis.
(AEC), as well as in lung-derived epithelial carcinoma cells (A549
and BEAS2B) [17], demonstrated that these cells can be triggered TGFß is a pleiotropic cytokine that has many versatile effects
to undergo EMT [17-20]. These studies showed that epithelial cells on the fibrogenic process in the lung, including but not restricted to
undergoing EMT have lower levels of epithelial cell markers (E- the already discussed potentiation of EMT. TGFß isoforms have
cadherin, cytokeratins and ZO-1) and increased levels of mesen- also been shown to regulate deposition of many components of the
chymal cell markers (collagens I and III, N-cadherin, EDA- ECM components [35] and affect the migratory and differentiation
fibronectin, vimentin, S100A4). The collagens and fibronectin also potential of fibroblasts towards myofibroblasts [36]. TGFß may
contribute directly to buildup of excessive amounts of extracellular also affect the epithelial cell fate itself by promoting epithelial cell
matrix. Also the myofibroblast marker -smooth muscle actin apoptosis [37], as well as migration [30], possibly through a TGFß-
(SMA) was shown to be increased in cells that underwent transi- mediated autocrine loop.
tion. Besides these cell markers, other proteins that may play a role PDGF serves as a growth factor for mesenchymal cells and was
in the fibrotic process were shown to be enhanced upon triggering shown to be essential in myofibroblast development as PDGF-A
of EMT in lung epithelial cells. Various studies have demonstrated knock-out mice were devoid of alveolar myofibroblast progenitors
increased production of members of the matrix metalloprotease [38], while on the other hand, overexpression of PDGF-A, driven
(MMP) family [21, 22]. These proteases may affect the fibrotic by the surfactant protein C (SPC) promoter in lung epithelium of
process in many ways, due to their capacity to cleave and activate transgenic mice, is associated with mesenchymal hyperplasia [39].
various growth factors and ECM components. An imbalance of In addition, PGDF has the potency to affect fibrogenesis by induc-
these proteases and their natural inhibitors, called tissue inhibitors ing mesenchymal cell proliferation and migration, and by enhanc-
of metalloproteases (TIMP), may have profound effects on the gen- ing collagen and MMP production [40].
eration of injured tissue and subsequent fibrotic processes. The connective tissue growth factor (CTGF) is another factor
It is well established that TGFß is a key player in the process of that is produced by lung-derived epithelial cells and which has been
EMT. TGFß, elicited predominantly by activated inflammatory implicated in lung fibrogenesis. Kasai et al. have demonstrated that
cells, including macrophages that are attracted to the site of injury, TGFß-induced EMT in the lung carcinoma cell line A549 is ac-
binds the TGFß type 2 receptor (TGFBR2), which then forms a companied by increased production of CTGF in cells undergoing
complex with TGFß type 1 receptor (TGFBR1). This complex initi- EMT [19]. Also CTGF is considered a profibrotic marker. Various
ates a signaling cascade in which a complex of Smad2 and Smad3 studies have demonstrated its role in ECM production in a lung
and subsequently Smad4 is activated. The activated complex of fibrosis model [41], in human renal cells [42], and by gingival fi-
Smads translocates to the nucleus and induces gene transcription. broblasts [43]. Furthermore, CTGF has been shown to promote
Although TGFß appears to be essential for EMT, other factors may fibroblast cell migration, contraction, adhesion, and proliferation
influence this process. Inflammatory cytokines, such as IL-1ß and [44-48].
TNF [21, 22], and also bacteria [23] and viruses [24] were shown These studies are indicative for the regulatory role of lung
to enhance TGFß-induced markers of EMT, even though the cyto- epithelial cells in the fibrogenic process. It has to be noted that the
kines themselves do not have an EMT inducing capacity. The pre- production of the profibrotic molecules listed above is not solely
cise mechanisms of this enhancement need to be elucidated yet. produced by epithelial cells. As also discussed elsewhere in this
EMT in Disease review, mesenchymal and inflammatory cells may significantly
As already indicated, EMT is a normal physiologic process contribute to their production. The lung epithelium however may
during embryogenesis. Whether EMT really contributes to the gen- also affect the fibrogenic process indirectly through recruitment
eration of fibroblasts and fibrosis is currently intensely debated in and/or activation of these cells.
4096 Current Pharmaceutical Design, 2012, Vol. 18, No. 27 Lekkerkerker et al.

Fibroblasts gen I and III [74]. They most likely derive from a common myeloid
Unlike the tightly organized airway epithelium, the underlying precursor similar to dendritic cells and macrophages and can be
connective tissue (lamina propria) consists of mesenchymal cells, distinguished from monocytes, macrophages, and fibroblasts
bathing in a mesh of ECM. The two layers are separated by a dense through a combination of markers [75]. Expression of chemokine
cell free layer of ECM, called basement membrane. The fibroblasts receptors CCR2, CXCR4, and CCR7 enables them to migrate to
residing in this layer and the epithelium are in close contact. It is sites of injury, where especially CXCR4 is expressed on 90% of the
well established that the two cell types interact in a bidirectional circulating fibrocytes [76, 77].
fashion by producing many factors. Recently, it was described that fibrocytes, recruited from the
The fibroblast is considered as the classical cell type involved circulation into tissues, can differentiate into various cell types
in fibrosis. Fibroblasts isolated from IPF patients indeed show a depending on the tissue niche [78]. During the fibrocytes' journey
more profibrotic secretory phenotype and fail to invade the ECM, through the tissue, hematopoietic surface receptors CD34, CD45,
suggesting that intrinsic defects in the fibroblasts may underlie the and CXCR4 on the fibrocytes gradually decrease, whereas the ex-
pathogenesis of pulmonary fibrosis [49, 50]. In the presence of pression of the mesenchymal markers, such as collagen, fibronectin,
profibrotic stimuli, such as TGFß and ECM components (e.g. colla- and proteoglycans gradually increases. Within the lung, the re-
gen I and fibronectin), these cells differentiate into a myofibroblast cruited fibrocytes may further differentiate towards a myofibroblast
phenotype, characterized by expression of SMA, secretion of col- phenotype with the expression of SMA [79].
lagen type I and III, and increased migration and contractility [51]. Culture of Fibrocytes
Fibroblasts from IPF patients behave differently in various assays, To enable more in-depth research of fibrocytes, over the last
compared to cells from normal donors. For instance, IPF fibroblasts years multiple culture protocols have been set up using different
are relatively resistant to oxidative stress and Fas-induced apoptosis cellular sources. Early studies on culture of fibrocytes made use of
and this correlates with the expression of anti-apoptotic factors [52, a CD14+ enriched mononuclear cell population in contact with T
53]. The group of Riches demonstrated that TNF sensitizes pri- cells. It was also shown that especially TGF induced the differen-
mary human lung fibroblasts from IPF patients to FAS-induced tiation and functional activity of these cultured fibrocytes. These
apoptosis [54, 55]. IL-6 enhanced apoptosis in normal fibroblasts differentiated, cultured fibrocytes expressed SMA and contracted
but inhibited apoptosis in IPF cells [56]. Interestingly, PGE2 defi- collagen gels in vitro, two hallmarks of wound-healing myofibro-
ciency appears to protect human primary fibroblasts against apopto- blasts [76]. This was substantiated by a study demonstrating that
sis, while enhancing apoptosis in alveolar epithelial cells [57]. Cai increased TGF in serum stimulates the differentiation of the
et al. studied motility of human primary fibroblasts from IPF pa- CD14+ cells, isolated from peripheral blood of burn patients, into
tients and demonstrated that IPF fibroblasts have slightly enhanced collagen-producing cells [80].
migratory capacity compared to normal donors which may be due
to the lower levels of FRNK, an inhibitor of migratory capacity Current protocols mostly use either peripheral blood mononu-
[58]. Interestingly, FRNK is decreased in IPF patients and is also clear cells or purified CD14+ monocytes for fibrocyte differentia-
involved in controlling fibroblast to myofibroblast conversion [59]. tion in vitro [79]. However, other studies suggest that fibrocytes
Comparing human primary fibroblasts from IPF patients versus may also differentiate from a population of bone-marrow derived
those from normal donors, Xia et al. showed that IPF fibroblasts CD45+ CXCR4+ cells found in peripheral blood [74].
have enhanced proliferative capacity compared to normal cells. In the presence of serum, fibrocytes require up to 2 weeks to
Moreover, in patient cells proliferation is not inhibited by the inter- differentiate, whereas in the absence of serum this process is accel-
action of integrin with polymerized collagen in contrast to the pro- erated with cells appearing in culture after only a few days. A num-
liferative restraint in normal cells under these conditions [60]. ber of other molecules can influence the differentiation of fibro-
It is commonly believed that the fibroblast is the major con- cytes in vitro, and presumably in vivo. TGF, IL-4, and IL-13 all
tributor to the pool of myofibroblasts in fibrogenic foci. A prerequi- promote fibrocyte differentiation, where specifically TGF appears
site for this contribution is the migration of fibroblasts towards foci, to enhance kinetics of fibrocyte differentiation [81] [74]. It is cur-
and subsequent expansion and differentiation. Many factors have rently assumed that fibrocytes that differentiate in the presence or
been identified over the years that activate the fibroblast and absence of serum reflect the same cell type, differing only in their
thereby enhance their migratory and/or proliferative capacity. These kinetics of generation.
include the already discussed growth factors PDGF, CTGF, and In addition, TLR2 agonists indirectly inhibit fibrocyte differen-
TGFß, but also chemokines such as CCL11 [61], CCL21 [62], tiation and cause some other cell type in the PBMC population to
CCL24 [63], and CCL26 [63]. inhibit monocytes from differentiating into fibrocytes [82]. A very
recent study [83] shows opposing effects of two different forms of
Fibrocytes hyaluronic acid on fibrocyte differentiation from purified mono-
Fibrocytes are a unique cell population that have been identified cytes in serum free conditions. High molecular weight hyaluronic
and characterized to play a role in wound healing [64-66]. The acid potentiated fibrocyte differentiation, while low molecular
presence of the fibrocytes appears to be a characteristic of fibrotic weight hyaluronic acid inhibited fibrocyte differentiation.
diseases, from lung, heart, skin, kidney to liver [64] [67-69] [70]. The fibrocyte culture protocols have been explored in more
One of the unique features of these cells is their ability to home detail by Pilling et al. [84], identifying conditions that affect this
from the blood stream to sites of tissue damage. As a consequence, differentiation, including the choice of anti-coagulant, media and
fibrocytes might also contribute to the increasing number of fibro- media supplements, culture substrates, and cell density.
blasts in fibrotic lesions. Over the last couple of years, research on Role of Fibrocytes in Lung Fibrosis In vivo
fibrocytes has intensified by investigating the origin of these cells,
as well as its contribution to fibrotic lesions [71, 72]. In vitro studies provide evidence for differentiation of fibro-
cytes into fibroblasts and eventually myofibroblasts, indeed sug-
Origin and Characterization of Fibrocytes gesting that fibrocytes could play an important role in fibrosis.
Fibrocytes were first identified in 1994 as circulating cells that However, evidence for fibrocyte into myofibroblast differentiation
homed and extravasated into sites of tissue injury contributing to in vivo has been lacking until recently. A recent study in a mouse
wound healing [73]. Fibrocytes are bone marrow-derived circulat- model of bleomycin-induced pulmonary fibrosis signifies a mecha-
ing cells that express markers for both hematopoietic cells (CD34, nistic role of fibrocytes in IPF [85]. Fibrocytes expressing both
CD43, and CD45), as well as markers linked to stromal cells, colla- CD45 and collagen I traffic to the lungs and differentiate into myo-
Cellular Players in Lung Fibrosis Current Pharmaceutical Design, 2012, Vol. 18, No. 27 4097

fibroblasts [85] [77]. In addition, this study elegantly showed that characteristics facilitating their function specific for that anatomical
the chemokine receptor ligand pair CXCR4-CXCL12 plays a part in location. For example, the liver contains specialized macrophages
homing of the fibrocytes to the fibrotic lesions. Fibrosis was en- called Kupffer cells and the lungs contain alveolar macrophages.
hanced by intravenous injection of in vitro-generated human fibro- During an immune response, monocytes are recruited from the
cytes and the cells were recruited to the lungs via their expression circulation which differentiate into tissue macrophages. Following
of CXCR4. In an intratracheal FITC-induced pulmonary fibrosis tissue damage and/or infection macrophages exhibit primarily a
model, fibrocytes use the CCR2-CCL12 axis for their recruitment. pro-inflammatory phenotype and secrete pro-inflammatory media-
Adoptive transfer of CCR2-expressing fibrocytes augments FITC- tors, such as TNF and IL-1. These pro-inflammatory macrophages
induced fibrosis. Both CCL2 and CCL12 are chemotactic for fi- are referred to as classically activated macrophages or M1 macro-
brocytes, and neutralization of CCL12 significantly protected from phages. Various chronic inflammatory diseases and autoimmune
FITC-induced fibrosis. Therefore, CCL12 is probably the CCR2 diseases, such as rheumatoid arthritis, are associated with activation
ligand responsible for driving fibroblast proliferation in the mouse of M1 macrophages [93]. These M1 macrophages also produce Th1
[86]. and Th17 polarizing cytokines like IL-12 and IL-23 to further drive
Induction of lung fibrosis in IL-10 overexpressing mice is also and regulate the inflammatory response. Nitric oxide is another
associated with an increase of CD45 and collagen I expressing fi- mediator excreted by pro-inflammatory macrophages which is toxic
brocytes. In addition, CCR2 and its ligand, CCL2, are highly for microorganisms but also very damaging for healthy neighboring
upregulated in these mice, suggesting that IL-10-induced fibrocyte cells. The macrophages also produce MMPs facilitating tissue ho-
recruitment is CCR2-CCL2 specific [87]. In conclusion, various meostasis.
mouse models for pulmonary fibrosis all point toward a role for To prevent an exacerbated immune response and collateral
fibrocytes. damage to surrounding tissue, the M1 macrophage response needs
Several lines of evidence also imply a role for fibrocytes in to be tightly controlled. Macrophages that play a role in wound
human lung fibrosis ([65, 74, 88, 89]. Early evidence for a role of healing are characterized by an alternative activated phenotype and
fibrocytes in fibrosis stemmed from an asthma patient study, where are designated M2 macrophages. This subset secretes anti-
they detected fibrocyte-like cells in the bronchial mucosa of pa- inflammatory mediators and is strongly associated with Th2 medi-
tients with allergic asthma. The number of these cells increased ated inflammation (e.g. IL-4 production) and antagonizes M1
during an asthmatic reaction induced by allergen exposure, suggest- macrophages to regulate the immune response.
ing that circulating fibrocytes may function as precursors of bron- A tightly regulated balance between M1 and M2 macrophages
chial myofibroblasts and may contribute to the onset of fibrosis in is thus crucial for maintaining immunological homeostasis and
asthma [90]. A study of Mehrad et al. demonstrated that fibrotic tissue integrity. Recent studies have shown that M1 macrophages
interstitial lung diseases have enhanced expression of CXCL12 in can convert into M2 macrophages, indicating a dynamic balance
lungs and plasma of patients. The increased CXCL12 levels were between both macrophage subtypes [94]. A disturbed balance in the
associated with higher number of circulating fibrocytes in the pe- regulation of these macrophage subtypes may result in excessive
ripheral blood [88]. inflammation or wound healing and have detrimental effects on
This observation was confirmed in IPF patients where fibro- normal function of tissue and organs [94].
cytes were identified in the lung parenchyma. CXCL12, the specific Macrophages in Fibrotic Diseases
ligand for CXCR4, was elevated in bronchoalveolar lavage, indicat-
ing that also in IPF patients CXCR4-positive fibrocytes may mi- A major initiator of fibrosis is the persistence of exogenous and
grate into the IPF lungs contributing to myofibroblast expansion endogenous stimuli of pathogens or tissue injury [95]. Both M1 and
[65]. Recently the same group proposed the circulating fibrocyte as M2 macrophages are involved in the process of fibrosis. Figure 2
a novel biomarker for bronchiolitis obliterans syndrome (BOS) gives an overview of these two macrophage types and the factors
development in lung transplant patients, a fibrotic process resulting produced by macrophages involved in fibrosis. M1 macrophages
in progressive narrowing of bronchiolar lumens and airflow ob- produce MMPs, resulting in degradation of ECM and thereby facili-
struction. Also in these patients, increased circulating fibrocyte tating the influx of more inflammatory cells from the circulation
levels correlated with development of BOS after lung transplanta- [96]. They furthermore contribute to fibrosis through the production
tion [91]. of TNF and IL-1 [97, 98]. Nevertheless, M2 macrophages are
considered to be the predominant macrophage subtype contributing
Moreover, a study in IPF patients comparing the presence of to fibrosis. They play an important role in wound healing and pro-
fibrocytes in blood of IPF patients vs healthy controls clearly duce amongst others fibrogenic mediators such as PDGF and TGF
showed a higher number of fibrocytes characterized by CD34+ [99, 100]. A strong link between M2 macrophages and fibrosis was
CD45+ in peripheral blood for the IPF patients. A further increase made in human patients, including IPF patients, which exhibit a
of fibrocytes was observed during acute disease exacerbation highly progressive fibrotic disease in the absence of infection [101].
and these cells were also observed in fibrotic lesions of IPF pa- M2 macrophages are important in wound healing and produce
tients, corroborating the concept that fibrocytes contribute to the TIMPs and MMPs to remove apoptotic debris, thereby preventing
increased numbers of fibroblasts in fibrotic lesions [89]. Taken an immunological response of tissue damaging M1 macrophage.
together, these data suggest that fibrocytes are involved in the
pathogenesis of human lung fibrosis. Furthermore, tissue macrophages reside in close proximity of
myofibroblasts and are able to interact with these cells in various
Macrophages ways [102]. M2 macrophages can directly affect fibrosis by the
Macrophages are responsible for immune surveillance and tis- excretion of profibrotic mediators, such as TIMP, and thereby di-
sue homeostasis. They are able to engulf pathogens using a broad rectly inhibiting ECM turnover [94, 103, 104]. M2 macrophages
repertoire of pathogen recognition receptors (PRRs) and destroy also produce fibronectin, thus directly contributing to the buildup of
them via degradation within phagolysosomes. Within the process of excessive ECM [105].
tissue homeostasis, macrophages play an essential role in removing Besides the direct effect of M2 macrophages on fibrosis, M2
dead and dying cells and toxic materials. Furthermore, macro- macrophages also indirectly contribute to fibrosis through activa-
phages are crucial in the orchestration of the wound healing proc- tion of other cell types such as T cells, fibroblasts, and endothelial
ess. To perform these important functions, macrophages consist of cells and thereby aggravating fibrosis [104]. Macrophages engulf
different subpopulations which are strategically positioned through- pathogens and dead cells and subsequently present the antigens to T
out the body [92]. These specialized macrophages have specific cells. M2 macrophages are able to enhance Th2 responses [106] and
4098 Current Pharmaceutical Design, 2012, Vol. 18, No. 27 Lekkerkerker et al.

Secretion of factors
Inflammatory directly and indirectly
M1 macrophage promoting fibrosis

TNF
Monocyte Macrophage IL-1
MMPs

TGF
PDGF
CTGF
CCL18
Fibronectin
Profibrotic TSP1
M2 macrophage

Fig. (2). Overview of the macrophages that play a role in lung fibrosis and the factors that contribute to the direct or indirect induction of fibrosis.

Th2 cytokines in turn can enhance collagen synthesis in fibroblasts BEAS2B and 16HBE) as an in vitro model for molecular and cellu-
and promote myofibroblast formation [107-109]. Furthermore, by lar processes in lung epithelium, and contributed considerably to
engulfing dead cells, macrophages produce TGF which is a strong the insight we have nowadays on the signaling pathways epithelial
profibrotic factor [102, 110, 111]. cells utilize to exercise their effects. However, cell lines may not
Macrophages in IPF always provide the best model for studying molecular processes as
they often carry transforming mutations and have abnormal chro-
Based on experimental data with the bleomycin-induced fibro- mosome copy numbers. In addition, extensive passaging of cells
sis mouse model, alveolar macrophages are thought to produce the and varying culture conditions may introduce additional genetic and
majority of the active TGF that promotes pulmonary fibrosis post-transcriptional changes affecting molecular and cellular func-
[112]. Interestingly, the primary level of control is not in the regula- tion and causing inconsistencies between different reports. For ex-
tion of mRNA expression, but in the regulation of both the secre- ample, inconsistent characteristics have been reported for A549
tion and activation of latent TGF1 [113, 114]. Furthermore, acti- cells [130] and Swain et al., using Raman spectroscopy, demon-
vated alveolar macrophages from rat lungs after bleomycin admini- strated that A549 cells differ considerably from primary cells [131].
stration demonstrated elevated amounts of active TGF1, as well as In addition, the expression of various genes involved in cell com-
plasmin and thrombosponin-1 (TSP-1) [115]. Plasmin and TSP-1 munication and adhesion are differently regulated in cell lines ver-
are tightly involved with the activation of latent TGF1. TSP-1 is a sus primary cells [132]. Also at the functional level, lung cell lines
glycoprotein which is expressed by various cell types such as differ considerably from primary lung cells. For example, in con-
macrophages, fibroblasts and endothelial cells, and is often associ- trast to human primary lung epithelial cells, the commonly used cell
ated at sites of inflammation and wound healing [116]. Further- line BEAS-2B fails to differentiate and form tight junctions when
more, inhibition of TSP-1 with antisense technology inhibits TGF cultured on air-to-liquid interface [133]. Also, barrier function, cell
activation and subsequently fibrosis [117]. Plasmin dependent acti- spreading, and wound healing differ between primary cells and cell
vation of TGF may occur at the cell surface of the activated alveo- lines [134]. The use of cell lines may therefore introduce biases
lar macrophages and requires interaction between TSP-1 and CD36 towards certain molecular pathways or the risk that important cellu-
(a receptor for TSP-1) [118]. M2 macrophages are therefore in- lar processes are overlooked. Employment of primary cells and
volved in the activation and the production of latent TGF, thereby preferably those from fibrosis patients will minimize such risks and
adding another layer of complexity in the process of fibrosis. provide us with better insights in the molecular processes involved
PDGF, which stimulates the proliferation, survival, and migra- in fibrotic disease.
tion of myofibroblasts is also expressed by M2 macrophages [119- Novel studies are being developed to study the role of epithelial
121]. Furthermore, the percentage of PDGF-positive macro- cells in vitro in even greater detail and under more physiological
phages in IPF was 3-fold increased in comparison to normal lung, conditions. The use of cells of primary origin is thereby indispensa-
indicating a strong correlation between PDGF producing macro- ble. In lung biology many researchers groups have put considerable
phages and IPF [122]. effort in optimizing protocols for the isolation of primary human
CCL-18, also known as pulmonary activation-related chemo- lung-derived cells. Epithelial cell cultures from both bronchial
kine (PARC), is highly expressed in alveolar macrophages from (HBEC) and alveolar (AEC) origin are widely used for functional
IPF patients [123-125]. CCL-18 production is strongly associated studies. These cells are usually isolated from lung resection or
with activation of M2 macrophages, implicating that alveolar transplant tissue by enzymatic treatment, and are cultured in serum
macrophages have an M2 phenotypes in IPF [126]. CCL18 has free conditions, although culturing procedures differ from one insti-
some overlapping functions with TGF, such as stimulating colla- tution to the other. A major advantage of these cultures is that they
gen production by fibroblasts [127, 128]. Furthermore, Prasse et al. are also available from patients such as IPF, COPD, asthma and
showed that CCL-18 concentration in serum strongly correlates cystic fibrosis, allowing comparisons between patient groups, as
with severity of IPF and is a predictive value for mortality [129]. well as with normal individuals.
Towards New In Vitro Models A review by Van de Bovenkam et al. [135] gives a good over-
view of in vitro cell culture systems for studying the role of primary
In the past decades much effort was put in the development of hepatic stellate cells in liver fibrosis. In the fields of cystic fibrosis
in vitro and in vivo models to unravel the molecular mechanisms and asthma, the pulmonary research community frequently includes
regulating fibrotic processes in the lung. Initial studies focused on human primary cells from patients in their studies. Unfortunately,
various cell lines derived from the lung (e.g. A549, NCI-H292, the use of human primary lung cells from IPF patients in fibrosis-
Cellular Players in Lung Fibrosis Current Pharmaceutical Design, 2012, Vol. 18, No. 27 4099

relevant functional assays is limited. This is mainly due to limited HBEC = Human bronchial epithelial cells
availability and logistical, regulatory, and technical issues surround- IFN = Interferon
ing the access, isolation, and culture of such cells. Recently, the
ECIPF work group reported on those issues [136]. IL = Interleukin
It is, however, important to use these cells under physiological IPF = Idiopathic pulmonary fibrosis
conditions and in a disease-relevant context, e.g. by culturing M1 = Classicaly activated macrophage
epithelial cells on air-to-liquid interface or by setting up co-cultures M2 = Alternatively activated macrophage
of fibroblasts and macrophages. The study of cells, derived from
MMP = Matrix metalloproteinase
IPF patients, in functional assays relevant for fibrosis (such as pro-
liferation, apoptosis, (de)differentiation, EMT, motility, oxidative PARC = Pulmonary activation-related chemokine
stress, inflammatory responses), in combination with functional PBMC = Peripheral blood mononuclear cell
genomics, can give invaluable insight in the possible molecular PDGF = Platelet-derived growth factor
mechanisms contributing to fibrosis. Indeed, as described in this
review, cells from IPF patients behave differently in functional PRR = Pattern recognition receptor
assays, compared to cells from normal donors. Most studies using SMC = Smooth muscle cell
IPF patient material so far have focused on lung fibroblasts, as de- SPC = Surfactant protein C
scribed above, and demonstrate that fibroblasts from IPF patients,
in comparison with cells from normal donors, are more active in TGF = Transforming growth factor beta
various cellular assays, mimicking fibrosis disease processes. These TGFR = Transforming growth factor beta 1 receptor
studies demonstrate that fibroblasts from IPF patients behave dif- Th = T helper cell
ferently than cells from healthy donors. Limited experience has
TIMP = Tissue inhibitor of metalloproteinases
been reported on macrophages and epithelial cells from IPF pa-
tients, but also those cells appear to differ in their behavior from TLR = Toll-like receptor
normal cells. Therefore to understand molecular and cellular proc- TNF = Tumor necrosis factor
esses related to IPF, it is of importance to use IPF derived cells as TSP-1 = Thrombospondin-1
much as possible. If feasible, the assays should be set up in condi-
tions that mimic the pathophysiology as much as possible, includ- CONFLICT OF INTEREST
ing cultures of epithelial cells on air-to-liquid interface, co-culture
The authors confirm that this article content has no conflicts of
of epithelial cells, or fibroblasts with macrophages, etc.
interest.
CONCLUSIONS
ACKNOWLEDGEMENTS
Fibrosis and especially idiopathic pulmonary fibrosis is a dis-
The authors wish to thank Dr. R. Geels, Galapagos, for genera-
ease that is receiving increasing attention from scientists and the
tion of the figures.
pharmaceutical industry. Unfortunately, little is known about the
pathogenesis of fibrosis and only recently we are unraveling the REFERENCES
various cellular and molecular processes that contribute to this dis-
[1] Firrincieli D, Boissan M, Chignard N. Epithelial-mesenchymal
ease. The overall consensus is that fibrosis is the result of an imbal- transition in the liver. Gastroenterol Clin Biol 2010; 34: 523-8.
ance in the immune and repair response following infection and/or [2] Choi SS, Diehl AM. Epithelial-to-mesenchymal transitions in the
tissue damage. These responses are the result of an intricate inter- liver. Hepatology 2009; 50: 2007-13.
play between various cell types such as epithelial cells, fibroblasts, [3] Reynolds SD, Malkinson AM. Clara cell: progenitor for the bron-
macrophages, fibrocytes, smooth muscle cells and endothelial cells. chiolar epithelium. Int J Biochem Cell Biol 2009; 42: 1-4.
An imbalance in the activity in one or more of these cell types may [4] Rawlins EL, Okubo T, Xue Y, et al. The role of Scgb1a1+ Clara
contribute to fibrosis. One of the best studied factors in fibrosis is cells in the long-term maintenance and repair of lung airway, but
TGF. Although TGF appears to be a main player in various not alveolar, epithelium. Cell Stem Cell 2009; 4: 525-34.
forms of fibrosis, it is clear that other factors, produced by the vari- [5] Roomans GM. Tissue engineering and the use of stem/progenitor
cells for airway epithelium repair. Eur Cell Mater 2010; 19: 284-
ous cell types, have a critical role in onset and progress of the dis- 99.
ease. To further understand the cellular and molecular mechanisms [6] Vareille M, Kieninger E, Edwards MR, Regamey N. The airway
involved in fibrosis, better and more relevant in vitro models are epithelium: soldier in the fight against respiratory viruses. Clin Mi-
needed. A first step would be to set up more functional cellular crobiol Rev 2011; 24: 210-29.
assays employing patient-derived cells in physiological relevant [7] Hiemstra PS. Epithelial antimicrobial peptides and proteins: their
conditions. Such assays could then be used to perform genetics, role in host defence and inflammation. Paediatr Respir Rev 2001;
functional genomics, and drug discovery studies to identify bio- 2: 306-10.
markers, novel drug targets, and new drugs for the treatment of [8] Kishimoto TK, Jutila MA, Berg EL, Butcher EC. Neutrophil Mac-1
fibrosis. and MEL-14 adhesion proteins inversely regulated by chemotactic
factors. Science 1989; 245: 1238-41.
ABBREVIATIONS [9] Message SD, Johnston SL. Host defense function of the airway
epithelium in health and disease: clinical background. J Leukoc
AEC = Alveolar epithelial cells Biol 2004; 75: 5-17.
aSMA = Alpha-smooth muscle actin [10] Qiu Y, Zhu J, Bandi V, et al. Bronchial mucosal inflammation and
upregulation of CXC chemoattractants and receptors in severe ex-
BOS = Bronchiolitis obliterans syndrome acerbations of asthma. Thorax 2007; 62: 475-82.
CCSP = Clara cell secretory protein [11] See H, Wark P. Innate immune response to viral infection of the
lungs. Paediatr Respir Rev 2008; 9: 243-50.
CD = Cluster of differentiation [12] Gleich GJ. Mechanisms of eosinophil-associated inflammation. J
DC = Dendritic cell Allergy Clin Immunol 2000; 105: 651-63.
[13] Reibman J, Hsu Y, Chen LC, et al. Airway epithelial cells release
ECM = Extracellular matrix MIP-3alpha/CCL20 in response to cytokines and ambient particu-
EMT = Epithelial-mesenchymal transition late matter. Am J Respir Cell Mol Biol 2003; 28: 648-54.
FITC = Fluorescein isothiocyanate [14] Hay ED. Interaction of embryonic surface and cytoskeleton with
extracellular matrix. Am J Anat 1982; 165: 1-12.
4100 Current Pharmaceutical Design, 2012, Vol. 18, No. 27 Lekkerkerker et al.

[15] Nakaya Y, Sukowati EW, Wu Y, Sheng G. RhoA and microtubule [38] Bostrom H, Willetts K, Pekny M, et al. PDGF-A signaling is a
dynamics control cell-basement membrane interaction in EMT dur- critical event in lung alveolar myofibroblast development and al-
ing gastrulation. Nat Cell Biol 2008; 10: 765-75. veogenesis. Cell 1996; 85: 863-73.
[16] Taneyhill LA, Coles EG, Bronner-Fraser M. Snail2 directly re- [39] Li J, Hoyle GW. Overexpression of PDGF-A in the lung epithelium
presses cadherin6B during epithelial-to-mesenchymal transitions of of transgenic mice produces a lethal phenotype associated with hy-
the neural crest. Development 2007; 134: 1481-90. perplasia of mesenchymal cells. Dev Biol 2001; 239: 338-49.
[17] Doerner AM, Zuraw BL. TGF-beta1 induced epithelial to mesen- [40] Lepisto J, Peltonen J, Vaha-Kreula M, et al. Platelet-derived
chymal transition (EMT) in human bronchial epithelial cells is en- growth factor isoforms PDGF-AA, -AB and -BB exert specific ef-
hanced by IL-1beta but not abrogated by corticosteroids. Respir fects on collagen gene expression and mitotic activity of cultured
Res 2009; 10: 100. human wound fibroblasts. Biochem Biophys Res Commun 1995;
[18] Hackett TL, Warner SM, Stefanowicz D, et al. Induction of epithe- 209: 393-9.
lial-mesenchymal transition in primary airway epithelial cells from [41] Bonniaud P, Margetts PJ, Kolb M, et al. Adenoviral gene transfer
patients with asthma by transforming growth factor-beta1. Am J of connective tissue growth factor in the lung induces transient fi-
Respir Crit Care Med 2009; 180: 122-33. brosis. Am J Respir Crit Care Med 2003; 168: 770-8.
[19] Kasai H, Allen JT, Mason RM, et al. TGF-beta1 induces human [42] Gore-Hyer E, Shegogue D, Markiewicz M, et al. TGF-beta and
alveolar epithelial to mesenchymal cell transition (EMT). Respir CTGF have overlapping and distinct fibrogenic effects on human
Res 2005; 6: 56. renal cells. Am J Physiol Renal Physiol 2002; 283: F707-16.
[20] Yao HW, Xie QM, Chen JQ, et al. TGF-beta1 induces alveolar [43] Heng EC, Huang Y, Black SA, Jr., Trackman PC. CCN2, connec-
epithelial to mesenchymal transition in vitro. Life Sci 2004; 76: 29- tive tissue growth factor, stimulates collagen deposition by gingival
37. fibroblasts via module 3 and alpha6- and beta1 integrins. J Cell
[21] Borthwick LA, McIlroy EI, Gorowiec MR, et al. Inflammation and Biochem 2006; 98: 409-20.
epithelial to mesenchymal transition in lung transplant recipients: [44] Bogatkevich GS, Gustilo E, Oates JC, et al. Distinct PKC isoforms
role in dysregulated epithelial wound repair. Am J Transplant 2010; mediate cell survival and DNA synthesis in thrombin-induced myo-
10: 498-509. fibroblasts. Am J Physiol Lung Cell Mol Physiol 2005; 288: L190-
[22] Camara J, Jarai G. Epithelial-mesenchymal transition in primary 201.
human bronchial epithelial cells is Smad-dependent and enhanced [45] Crean JK, Finlay D, Murphy M, et al. The role of p42/44 MAPK
by fibronectin and TNF-alpha. Fibrogenesis Tissue Repair 2010; 3: and protein kinase B in connective tissue growth factor induced ex-
2. tracellular matrix protein production, cell migration, and actin cy-
[23] Borthwick LA, Sunny SS, Oliphant V, et al. Pseudomonas aerugi- toskeletal rearrangement in human mesangial cells. J Biol Chem
nosa accentuates epithelial-to-mesenchymal transition in the air- 2002; 277: 44187-94.
way. Eur Respir J 2011; 37: 1237-47. [46] Frazier K, Williams S, Kothapalli D, et al. Stimulation of fibroblast
[24] Shimamura M, Murphy-Ullrich JE, Britt WJ. Human cytomega- cell growth, matrix production, and granulation tissue formation by
lovirus induces TGF-beta1 activation in renal tubular epithelial connective tissue growth factor. J Invest Dermatol 1996; 107: 404-
cells after epithelial-to-mesenchymal transition. PLoS Pathog 2010; 11.
6: e1001170. [47] Grotendorst GR, Rahmanie H, Duncan MR. Combinatorial signal-
[25] Iwano M, Plieth D, Danoff TM, et al. Evidence that fibroblasts ing pathways determine fibroblast proliferation and myofibroblast
derive from epithelium during tissue fibrosis. J Clin Invest 2002; differentiation. Faseb J 2004; 18: 469-79.
110: 341-50. [48] Leask A, Holmes A, Abraham DJ. Connective tissue growth factor:
[26] Rastaldi MP. Epithelial-mesenchymal transition and its implica- a new and important player in the pathogenesis of fibrosis. Curr
tions for the development of renal tubulointerstitial fibrosis. J Rheumatol Rep 2002; 4: 136-42.
Nephrol 2006; 19: 407-12. [49] Ramos C, Montano M, Garcia-Alvarez J, et al. Fibroblasts from
[27] Yang J, Shultz RW, Mars WM, et al. Disruption of tissue-type idiopathic pulmonary fibrosis and normal lungs differ in growth
plasminogen activator gene in mice reduces renal interstitial fibro- rate, apoptosis, and tissue inhibitor of metalloproteinases expres-
sis in obstructive nephropathy. J Clin Invest 2002; 110: 1525-38. sion. Am J Respir Cell Mol Biol 2001; 24: 591-8.
[28] Grgic I, Duffield JS, Humphreys BD. The origin of interstitial [50] Lovgren AK, Kovacs JJ, Xie T, et al. beta-arrestin deficiency pro-
myofibroblasts in chronic kidney disease. Pediatr Nephrol 2012; tects against pulmonary fibrosis in mice and prevents fibroblast in-
27: 183-93. vasion of extracellular matrix. Sci Transl Med 2011; 3: 74ra23.
[29] Humphreys BD, Lin SL, Kobayashi A, et al. Fate tracing reveals [51] Wynn TA. Common and unique mechanisms regulate fibrosis in
the pericyte and not epithelial origin of myofibroblasts in kidney various fibroproliferative diseases. J Clin Invest 2007; 117: 524-9.
fibrosis. Am J Pathol 2010; 176: 85-97. [52] Moodley YP, Caterina P, Scaffidi AK, et al. Comparison of the
[30] Kim KK, Kugler MC, Wolters PJ, et al. Alveolar epithelial cell morphological and biochemical changes in normal human lung fi-
mesenchymal transition develops in vivo during pulmonary fibrosis broblasts and fibroblasts derived from lungs of patients with idio-
and is regulated by the extracellular matrix. Proc Natl Acad Sci pathic pulmonary fibrosis during FasL-induced apoptosis. J Pathol
USA 2006; 103: 13180-5. 2004; 202: 486-95.
[31] Marmai C, Sutherland RE, Kim KK, et al. Alveolar epithelial cells [53] Bocchino M, Agnese S, Fagone E, et al. Reactive oxygen species
express mesenchymal proteins in patients with idiopathic pulmo- are required for maintenance and differentiation of primary lung fi-
nary fibrosis. Am J Physiol Lung Cell Mol Physiol 2011; 301: L71- broblasts in idiopathic pulmonary fibrosis. PLoS One 2010; 5:
8. e14003.
[32] Warshamana GS, Corti M, Brody AR. TNF-alpha, PDGF, and [54] Frankel SK, Cosgrove GP, Cha SI, et al. TNF-alpha sensitizes
TGF-beta(1) expression by primary mouse bronchiolar-alveolar normal and fibrotic human lung fibroblasts to Fas-induced apopto-
epithelial and mesenchymal cells: tnf-alpha induces TGF-beta(1). sis. Am J Respir Cell Mol Biol 2006; 34: 293-304.
Exp Mol Pathol 2001; 71: 13-33. [55] Wynes MW, Edelman BL, Kostyk AG, et al. Increased cell surface
[33] Aubert JD, Dalal BI, Bai TR, et al. Transforming growth factor Fas expression is necessary and sufficient to sensitize lung fibro-
beta 1 gene expression in human airways. Thorax 1994; 49: 225- blasts to Fas ligation-induced apoptosis: implications for fibroblast
32. accumulation in idiopathic pulmonary fibrosis. J Immunol 2011;
[34] Coker RK, Laurent GJ, Shahzeidi S, et al. Diverse cellular TGF- 187: 527-37.
beta 1 and TGF-beta 3 gene expression in normal human and mur- [56] Moodley YP, Misso NL, Scaffidi AK, et al. Inverse effects of
ine lung. Eur Respir J 1996; 9: 2501-7. interleukin-6 on apoptosis of fibroblasts from pulmonary fibrosis
[35] Eickelberg O, Kohler E, Reichenberger F, et al. Extracellular ma- and normal lungs. Am J Respir Cell Mol Biol 2003; 29: 490-8.
trix deposition by primary human lung fibroblasts in response to [57] Maher TM, Evans IC, Bottoms SE, et al. Diminished prostaglandin
TGF-beta1 and TGF-beta3. Am J Physiol 1999; 276: L814-24. E2 contributes to the apoptosis paradox in idiopathic pulmonary fi-
[36] Scotton CJ, Chambers RC. Molecular targets in pulmonary fibrosis: brosis. Am J Respir Crit Care Med 2010; 182: 73-82.
the myofibroblast in focus. Chest 2007; 132: 1311-21. [58] Cai GQ, Zheng A, Tang Q, et al. Downregulation of FAK-related
[37] Hagimoto N, Kuwano K, Inoshima I, et al. TGF-beta 1 as an en- non-kinase mediates the migratory phenotype of human fibrotic
hancer of Fas-mediated apoptosis of lung epithelial cells. J Immu- lung fibroblasts. Exp Cell Res 2010; 316: 1600-9.
nol 2002; 168: 6470-8.
Cellular Players in Lung Fibrosis Current Pharmaceutical Design, 2012, Vol. 18, No. 27 4101

[59] Ding Q, Gladson CL, Wu H, et al. Focal adhesion kinase (FAK)- [85] Phillips RJ, Burdick MD, Hong K, et al. Circulating fibrocytes
related non-kinase inhibits myofibroblast differentiation through traffic to the lungs in response to CXCL12 and mediate fibrosis. J
differential MAPK activation in a FAK-dependent manner. J Biol Clin Invest 2004; 114: 438-46.
Chem 2008; 283: 26839-49. [86] Moore BB, Murray L, Das A, et al. The role of CCL12 in the re-
[60] Xia H, Diebold D, Nho R, et al. Pathological integrin signaling cruitment of fibrocytes and lung fibrosis. Am J Respir Cell Mol
enhances proliferation of primary lung fibroblasts from patients Biol 2006; 35: 175-81.
with idiopathic pulmonary fibrosis. J Exp Med 2008; 205: 1659-72. [87] Sun L, Louie MC, Vannella KM, et al. New concepts of IL-10-
[61] Puxeddu I, Bader R, Piliponsky AM, et al. The CC chemokine induced lung fibrosis: fibrocyte recruitment and M2 activation in a
eotaxin/CCL11 has a selective profibrogenic effect on human lung CCL2/CCR2 axis. Am J Physiol Lung Cell Mol Physiol 2011; 300:
fibroblasts. J Allergy Clin Immunol 2006; 117: 103-10. L341-53.
[62] Pierce EM, Carpenter K, Jakubzick C, et al. Idiopathic pulmonary [88] Mehrad B, Burdick MD, Zisman DA, et al. Circulating peripheral
fibrosis fibroblasts migrate and proliferate to CC chemokine ligand blood fibrocytes in human fibrotic interstitial lung disease. Bio-
21. Eur Respir J 2007; 29: 1082-93. chem Biophys Res Commun 2007; 353: 104-8.
[63] Kohan M, Puxeddu I, Reich R, et al. Eotaxin-2/CCL24 and eo- [89] Moeller A, Gilpin SE, Ask K, et al. Circulating fibrocytes are an
taxin-3/CCL26 exert differential profibrogenic effects on human indicator of poor prognosis in idiopathic pulmonary fibrosis. Am J
lung fibroblasts. Ann Allergy Asthma Immunol 2010; 104: 66-72. Respir Crit Care Med 2009; 179: 588-94.
[64] Gomperts BN, Strieter RM. Fibrocytes in lung disease. J Leukoc [90] Schmidt M, Sun G, Stacey MA, et al. Identification of circulating
Biol 2007; 82: 449-56. fibrocytes as precursors of bronchial myofibroblasts in asthma. J
[65] Andersson-Sjoland A, de Alba CG, Nihlberg K, et al. Fibrocytes Immunol 2003; 171: 380-9.
are a potential source of lung fibroblasts in idiopathic pulmonary [91] LaPar DJ, Burdick MD, Emaminia A, et al. Circulating fibrocytes
fibrosis. Int J Biochem Cell Biol 2008; 40: 2129-40. correlate with bronchiolitis obliterans syndrome development after
[66] Maharaj SS, Baroke E, Gauldie J, Kolb MR. Fibrocytes in chronic lung transplantation: a novel clinical biomarker. Ann Thorac Surg
lung disease - Facts and controversies. Pulm Pharmacol Ther 2011. 2011; 92: 470-7; discussion 477.
[67] Homer RJ, Herzog EL. Recent advances in pulmonary fibrosis: [92] Mantovani A, Sica A, Sozzani S, et al. The chemokine system in
implications for scleroderma. Curr Opin Rheumatol 2010; 22: 683- diverse forms of macrophage activation and polarization. Trends
9. Immunol 2004; 25: 677-86.
[68] Barnes JL, Glass WF, 2nd. Renal interstitial fibrosis: a critical [93] Murphy CA, Langrish CL, Chen Y, et al. Divergent pro- and anti-
evaluation of the origin of myofibroblasts. Contrib Nephrol 2011; inflammatory roles for IL-23 and IL-12 in joint autoimmune in-
169: 73-93. flammation. J Exp Med 2003; 198: 1951-7.
[69] Kisseleva T, Uchinami H, Feirt N, et al. Bone marrow-derived [94] Duffield JS, Forbes SJ, Constandinou CM, et al. Selective deple-
fibrocytes participate in pathogenesis of liver fibrosis. J Hepatol tion of macrophages reveals distinct, opposing roles during liver in-
2006; 45: 429-38. jury and repair. J Clin Invest 2005; 115: 56-65.
[70] Keeley EC, Mehrad B, Strieter RM. The role of fibrocytes in fi- [95] Meneghin A, Hogaboam CM. Infectious disease, the innate im-
brotic diseases of the lungs and heart. Fibrogenesis Tissue Repair mune response, and fibrosis. J Clin Invest 2007; 117: 530-8.
2011; 4: 2. [96] Jiang D, Liang J, Fan J, et al. Regulation of lung injury and repair
[71] Reilkoff RA, Bucala R, Herzog EL. Fibrocytes: emerging effector by Toll-like receptors and hyaluronan. Nat Med 2005; 11: 1173-9.
cells in chronic inflammation. Nat Rev Immunol 2011; 11: 427-35. [97] Raghu G, Brown KK, Costabel U, et al. Treatment of idiopathic
[72] du Bois RM. Strategies for treating idiopathic pulmonary fibrosis. pulmonary fibrosis with etanercept: an exploratory, placebo-
Nat Rev Drug Discov 2010; 9: 129-140. controlled trial. Am J Respir Crit Care Med 2008; 178: 948-55.
[73] Bucala R, Spiegel LA, Chesney J, et al. Circulating fibrocytes [98] Siwik DA, Chang DL, Colucci WS. Interleukin-1beta and tumor
define a new leukocyte subpopulation that mediates tissue repair. necrosis factor-alpha decrease collagen synthesis and increase ma-
Mol Med 1994; 1: 71-81. trix metalloproteinase activity in cardiac fibroblasts in vitro. Circ
[74] Strieter RM, Keeley EC, Hughes MA, et al. The role of circulating Res 2000; 86: 1259-65.
mesenchymal progenitor cells (fibrocytes) in the pathogenesis of [99] Song E, Ouyang N, Horbelt M, et al. Influence of alternatively and
pulmonary fibrosis. J Leukoc Biol 2009; 86: 1111-8. classically activated macrophages on fibrogenic activities of human
[75] Pilling D, Fan T, Huang D, et al. Identification of markers that fibroblasts. Cell Immunol 2000; 204: 19-28.
distinguish monocyte-derived fibrocytes from monocytes, macro- [100] Murray PJ, Wynn TA. Protective and pathogenic functions of
phages, and fibroblasts. PLoS One 2009; 4: e7475. macrophage subsets. Nat Rev Immunol 2011; 11: 723-37.
[76] Abe R, Donnelly SC, Peng T, et al. Peripheral blood fibrocytes: [101] Thannickal VJ, Toews GB, White ES, et al. Mechanisms of pul-
differentiation pathway and migration to wound sites. J Immunol monary fibrosis. Annu Rev Med 2004; 55: 395-417.
2001; 166: 7556-62. [102] Wynn TA, Barron L. Macrophages: master regulators of inflamma-
[77] Mehrad B, Burdick MD, Strieter RM. Fibrocyte CXCR4 regulation tion and fibrosis. Semin Liver Dis 2010; 30: 245-57.
as a therapeutic target in pulmonary fibrosis. Int J Biochem Cell [103] Selman M, Ruiz V, Cabrera S, et al. TIMP-1, -2, -3, and -4 in idio-
Biol 2009; 41: 1708-18. pathic pulmonary fibrosis. A prevailing nondegradative lung mi-
[78] Andersson-Sjoland A, Nihlberg K, Eriksson L, et al. Fibrocytes croenvironment? Am J Physiol Lung Cell Mol Physiol 2000; 279:
and the tissue niche in lung repair. Respir Res 2011; 12: 76. L562-74.
[79] Curnow SJ, Fairclough M, Schmutz C, et al. Distinct types of fi- [104] Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol
brocyte can differentiate from mononuclear cells in the presence 2008; 214: 199-210.
and absence of serum. PLoS One 2010; 5: e9730. [105] Gratchev A, Guillot P, Hakiy N, et al. Alternatively activated
[80] Yang L, Scott PG, Giuffre J, et al. Peripheral blood fibrocytes from macrophages differentially express fibronectin and its splice vari-
burn patients: identification and quantification of fibrocytes in ad- ants and the extracellular matrix protein betaIG-H3. Scand J Im-
herent cells cultured from peripheral blood mononuclear cells. Lab munol 2001; 53: 386-92.
Invest 2002; 82: 1183-92. [106] Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activa-
[81] Metz CN. Fibrocytes: a unique cell population implicated in wound tion and polarization. Front Biosci 2008; 13: 453-61.
healing. Cellular and Molecular Life Sciences 2003; 60: 1342- [107] Hashimoto S, Gon Y, Takeshita I, et al. IL-4 and IL-13 induce
1350. myofibroblastic phenotype of human lung fibroblasts through c-Jun
[82] Maharjan AS, Pilling D, Gomer RH. Toll-like receptor 2 agonists NH2-terminal kinase-dependent pathway. J Allergy Clin Immunol
inhibit human fibrocyte differentiation. Fibrogenesis Tissue Repair 2001; 107: 1001-8.
2010; 3: 23. [108] Murray LA, Argentieri RL, Farrell FX, et al. Hyper-responsiveness
[83] Maharjan AS, Pilling D, Gomer RH. High and low molecular of IPF/UIP fibroblasts: interplay between TGFbeta1, IL-13 and
weight hyaluronic acid differentially regulate human fibrocyte dif- CCL2. Int J Biochem Cell Biol 2008; 40: 2174-82.
ferentiation. PLoS One 2011; 6: e26078. [109] Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat
[84] Pilling D, Vakil V, Gomer RH. Improved serum-free culture condi- Rev Immunol 2004; 4: 583-94.
tions for the differentiation of human and murine fibrocytes. J Im- [110] Fadok VA, Bratton DL, Konowal A, et al. Macrophages that have
munol Methods 2009; 351: 62-70. ingested apoptotic cells in vitro inhibit proinflammatory cytokine
4102 Current Pharmaceutical Design, 2012, Vol. 18, No. 27 Lekkerkerker et al.

production through autocrine/paracrine mechanisms involving [123] Pechkovsky DV, Prasse A, Kollert F, et al. Alternatively activated
TGF-beta, PGE2, and PAF. J Clin Invest 1998; 101: 890-8. alveolar macrophages in pulmonary fibrosis-mediator production
[111] Rock KL, Kono H. The inflammatory response to cell death. Annu and intracellular signal transduction. Clin Immunol 2010; 137: 89-
Rev Pathol 2008; 3: 99-126. 101.
[112] Khalil N, Corne S, Whitman C, Yacyshyn H. Plasmin regulates the [124] Prasse A, Muller-Quernheim J. Non-invasive biomarkers in pul-
activation of cell-associated latent TGF-beta 1 secreted by rat al- monary fibrosis. Respirology 2009; 14: 788-95.
veolar macrophages after in vivo bleomycin injury. Am J Respir [125] Prasse A, Pechkovsky DV, Toews GB, et al. A vicious circle of
Cell Mol Biol 1996; 15: 252-9. alveolar macrophages and fibroblasts perpetuates pulmonary fibro-
[113] Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell sis via CCL18. Am J Respir Crit Care Med 2006; 173: 781-92.
biology. Nat Rev Immunol 2002; 2: 46-53. [126] Prasse A, Pechkovsky DV, Toews GB, et al. CCL18 as an indicator
[114] Munger JS, Huang X, Kawakatsu H, et al. The integrin alpha v beta of pulmonary fibrotic activity in idiopathic interstitial pneumonias
6 binds and activates latent TGF beta 1: a mechanism for regulating and systemic sclerosis. Arthritis Rheum 2007; 56: 1685-93.
pulmonary inflammation and fibrosis. Cell 1999; 96: 319-28. [127] Atamas SP, Luzina IG, Choi J, et al. Pulmonary and activation-
[115] Ide M, Ishii H, Mukae H, et al. High serum levels of throm- regulated chemokine stimulates collagen production in lung fibro-
bospondin-1 in patients with idiopathic interstitial pneumonia. blasts. Am J Respir Cell Mol Biol 2003; 29: 743-9.
Respir Med 2008; 102: 1625-30. [128] Luzina IG, Highsmith K, Pochetuhen K, et al. PKCalpha mediates
[116] Bornstein P. Diversity of function is inherent in matricellular pro- CCL18-stimulated collagen production in pulmonary fibroblasts.
teins: an appraisal of thrombospondin 1. J Cell Biol 1995; 130: Am J Respir Cell Mol Biol 2006; 35: 298-305.
503-6. [129] Prasse A, Probst C, Bargagli E, et al. Serum CC-chemokine ligand
[117] Daniel C, Takabatake Y, Mizui M, et al. Antisense oligonucleo- 18 concentration predicts outcome in idiopathic pulmonary fibrosis.
tides against thrombospondin-1 inhibit activation of tgf-beta in fi- Am J Respir Crit Care Med 2009; 179: 717-23.
brotic renal disease in the rat in vivo. Am J Pathol 2003; 163: 1185- [130] Witherden IR, Vanden Bon EJ, Goldstraw P, et al. Primary human
92. alveolar type II epithelial cell chemokine release: effects of ciga-
[118] Yehualaeshet T, O'Connor R, Green-Johnson J, et al. Activation of rette smoke and neutrophil elastase. Am J Respir Cell Mol Biol
rat alveolar macrophage-derived latent transforming growth factor 2004; 30: 500-9.
beta-1 by plasmin requires interaction with thrombospondin-1 and [131] Swain RJ, Kemp SJ, Goldstraw P, et al. Assessment of cell line
its cell surface receptor, CD36. Am J Pathol 1999; 155: 841-51. models of primary human cells by Raman spectral phenotyping.
[119] Bonner JC. Regulation of PDGF and its receptors in fibrotic dis- Biophys J 2010; 98: 1703-11.
eases. Cytokine Growth Factor Rev 2004; 15: 255-73. [132] Ertel A, Verghese A, Byers SW, et al. Pathway-specific differences
[120] Martinet Y, Rom WN, Grotendorst GR, et al. Exaggerated sponta- between tumor cell lines and normal and tumor tissue cells. Mol
neous release of platelet-derived growth factor by alveolar macro- Cancer 2006; 5: 55.
phages from patients with idiopathic pulmonary fibrosis. N Engl J [133] Stewart CE, Torr EE, Mohd Jamili NH, et al. Evaluation of differ-
Med 1987; 317: 202-9. entiated human bronchial epithelial cell culture systems for asthma
[121] Bonner JC, Osornio-Vargas AR, Badgett A, Brody AR. Differential research. J Allergy (Cairo) 2012; 2012: 943982.
proliferation of rat lung fibroblasts induced by the platelet-derived [134] Heijink IH, Brandenburg SM, Noordhoek JA, et al. Characterisa-
growth factor-AA, -AB, and -BB isoforms secreted by rat alveolar tion of cell adhesion in airway epithelial cell types using electric
macrophages. Am J Respir Cell Mol Biol 1991; 5: 539-47. cell-substrate impedance sensing. Eur Respir J 2010; 35: 894-903.
[122] Vignaud JM, Allam M, Martinet N, et al. Presence of platelet- [135] Van de Bovenkamp M, Groothuis GM, Meijer DK, Olinga P. Liver
derived growth factor in normal and fibrotic lung is specifically as- fibrosis in vitro: cell culture models and precision-cut liver slices.
sociated with interstitial macrophages, while both interstitial Toxicol In vitro 2007; 21: 545-57.
macrophages and alveolar epithelial cells express the c-sis proto- [136] Jenkins G, Blanchard A, Borok Z, et al. In search of the fibrotic
oncogene. Am J Respir Cell Mol Biol 1991; 5: 531-8. epithelial cell: opportunities for a collaborative network. Thorax
2012; 67: 179-182.

Received: March 6, 2012 Accepted: March 14, 2012

View publication stats

You might also like