ArticlePDF Available

TGF- 1 Induces Progressive Pleural Scarring and Subpleural Fibrosis

Authors:

Abstract and Figures

Pleural fibrosis is a misunderstood disorder which can cause severe restrictive lung disease with high morbidity and even mortality. The condition can develop in response to a large variety of diseases and tissue injury, among them infectious disease, asbestos, drugs, and radiation therapy. There is no efficient treatment to reverse established pleural fibrosis. TGF-beta1 is suspected, even if not proven, as a key cytokine in this process. In this study, we used adenoviral gene transfer of TGF-beta1 to the pleural mesothelium in rats. We show that local and transient TGF-beta1 overexpression induces homogenous, prolonged, and progressive pleural fibrosis without pleurodesis, associated with severe impairment of pulmonary function. We further demonstrate that pleural fibrosis can expand into the lung parenchyma from the visceral layer, but not into the muscle from the parietal layer. We provide evidence that matrix accumulation and fibrosis within the parenchyma evolved through a process involving "mesothelial-fibroblastoid transformation" and suggest that the pleural mesothelial cell may be an important player involved in the development of the subpleural distribution pattern known to be a hallmark of pulmonary fibrosis. This new model of pleural fibrosis will allow us to better understand the mechanisms of progressive fibrogenesis, and to explore novel antifibrotic therapies in the pleural cavity.
Content may be subject to copyright.
TGF-
1 Induces Progressive Pleural Scarring
and Subpleural Fibrosis
1
Nathalie Decologne,* Martin Kolb,
Peter J. Margetts,
Franck Menetrier,* Yves Artur,
Carmen Garrido,* Jack Gauldie,
Philippe Camus,*
§
and Philippe Bonniaud
2
*
§
Pleural fibrosis is a misunderstood disorder which can cause severe restrictive lung disease with high morbidity and even mor-
tality. The condition can develop in response to a large variety of diseases and tissue injury, among them infectious disease,
asbestos, drugs, and radiation therapy. There is no efficient treatment to reverse established pleural fibrosis. TGF-
1 is suspected,
even if not proven, as a key cytokine in this process. In this study, we used adenoviral gene transfer of TGF-
1 to the pleural
mesothelium in rats. We show that local and transient TGF-
1 overexpression induces homogenous, prolonged, and progressive
pleural fibrosis without pleurodesis, associated with severe impairment of pulmonary function. We further demonstrate that
pleural fibrosis can expand into the lung parenchyma from the visceral layer, but not into the muscle from the parietal layer. We
provide evidence that matrix accumulation and fibrosis within the parenchyma evolved through a process involving “mesothelial-
fibroblastoid transformation” and suggest that the pleural mesothelial cell may be an important player involved in the develop-
ment of the subpleural distribution pattern known to be a hallmark of pulmonary fibrosis. This new model of pleural fibrosis will
allow us to better understand the mechanisms of progressive fibrogenesis, and to explore novel antifibrotic therapies in the pleural
cavity. The Journal of Immunology, 2007, 179: 6043– 6051.
P
leural fibrosis can cause severe restrictive lung disease. It
is usually considered as a complication of other disorders
involving the chest cavity. Pneumonia, with parapneumo-
nic effusion and empyema, tuberculosis and asbestos are among
the most common causes for pleural fibrosis (1). Furthermore, nu-
merous drugs can contribute to the development of pleural fibrosis,
the best known being ergot drugs, cytostatic agents, and thoracic
irradiation (Refs. 2 and 3, and www.pneumotox.com (“The drug-
induced lung diseases”)). Other potential reasons are systemic con-
nective tissue disease, hemothorax, and progressive postthoracot-
omy scarring after coronary bypass (4). Depending on disease
severity, pleural fibrosis can compromise respiratory function,
markedly impair quality of life, and can be associated with high
morbidity or even mortality. There is no effective therapy to re-
verse established pleural fibrosis.
The pleura is a metabolically active membrane involved in
maintaining a dynamic homeostasis of fluid within the chest cav-
ity. The homeostasis is important for the mechanical properties of
chest wall and lungs, and a breakdown of the fluid balance can lead
to pleural effusion and ultimately fibrosis (5, 6). Pleural fibrosis
can be defined as excessive deposition of matrix components re-
sulting in the destruction of regular pleural tissue architecture. The
disorder can manifest itself as discrete localized lesions (pleural
plaques) or diffuse pleural thickening (5). Most research related to
pleural scarring concerns the induction of pleurodesis as an ap-
proach to treat chronic effusion associated with metastasized can-
cer (7). Animal studies have shown that TGF-
plays an active role
in pleurodesis as well as in pleural fluid formation (8 –12).
TGF-
is a multifunctional cytokine critically involved in the
pathogenesis of fibrosis through its potent effects on fibroblast dif-
ferentiation, extracellular matrix formation (13, 14), and epithelial-
to-mesenchymal transition (EMT)
3
(15). Peritoneal mesothelial
cells may undergo mesenchymal conversion (16, 17) and TGF-
1
gene transfer to the peritoneal mesothelium induces peritoneal fi-
brosis with evidence of mesothelial-to-mesenchymal transition or
“mesothelial-fibroblastoid transformation” (MFT) (18).
In this study, we used transient transfer of the active TGF-
1
gene by adenoviral vectors to the pleural cavity and mesothelium.
We demonstrate that this approach induces homogenous, pro-
longed, and progressive pleural fibrosis without pleurodesis, asso-
ciated with severe impairment of pulmonary function. This new
model of pleural fibrosis will allow us to better understand the
mechanisms of progressive fibrogenesis, and to explore novel an-
tifibrotic therapies in the pleural cavity. We further show that pleu-
ral fibrosis, through a process involving MFT, can expand into the
lung parenchyma from the visceral layer, but not into the muscle
from the parietal layer, suggesting that a distinct local environment
is required for progressive fibrotic responses in the tissue.
*Faculty of Medicine and Pharmacy, Institut National de la Sante´ et de la Recherche
Me´dicale (INSERM), Unite´ Mixte de Recherche (UMR) 866, Dijon, France;
UMR
1129, Flaveur, Vision et Comportement du Consommateur, Institut National de la
Recherche Agronomique, Etablissement National d’Enseignement Supe´rieur
Agronomique de Dijon, University of Burgundy, Dijon, France;
Department of Pa-
thology and Molecular Medicine, Centre for Gene Therapeutics, McMaster Univer-
sity, Hamilton, Ontario, Canada; and
§
Service de Pneumologie et Re´animation Res-
piratoire, Centre Hospitalier Universitaire du Bocage, Dijon, France
Received for publication June 12, 2007. Accepted for publication August 14, 2007.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
N.D. was supported by the Comite´deCoˆte d’Or de la Ligue Contre le Cancer and
by the Socie´te´ de Pneumologie de Langue Franc¸aise. P.B. was supported by Pneu-
mologie De´veloppement. M.K. is a Parker B. Francis Fellow and was supported by a
Career Development Award of Department of Medicine, McMaster University.
P.J.M. is a Canadian Institutes for Health Research Clinician Scientist.
2
Address correspondence and reprint requests to Dr. Philippe Bonniaud, Service de
Pneumologie et Re´animation Respiratoire, Centre Hospitalier Universitaire du Bo-
cage, 21079 Dijon, France. E-mail address: philippe.bonniaud@chu-dijon.fr
3
Abbreviations used in this paper: EMT, epithelial-to-mesenchymal transition; MFT,
mesothelial-fibroblastoid transformation; PLF, pleural lavage fluid; BAL, bronchoal-
veolar lavage; BALF, BAL fluid; HSP, heat shock protein; SMA, smooth muscle
actin; MMP, matrix metalloproteinase.
Copyright © 2007 by The American Association of Immunologists, Inc. 0022-1767/07/$2.00
The Journal of Immunology
www.jimmunol.org
Materials and Methods
Recombinant adenovirus
We used AdTGF-
1
223/225
, an adenovirus construct with a mutant TGF-
1
translated into spontaneously bioactive TGF-
1, AdLacZ (coding for
-ga-
lactosidase), and AdDL (control vectors) with no insert in the deleted E1
region for the experiments described. The construction of adenoviral vec-
tors is described in detail elsewhere (19, 20).
Animal treatment
Female Sprague-Dawley rats (Charles River Laboratories) weighing 200
225 g were housed in special pathogen-free conditions. Rodent laboratory
food and water were provided ad libitum. The animals were treated in
accordance to the guidelines of the Ministe`re de la Recherche et de la
Technologie (Paris, France). All animal procedures were performed with
inhalation anesthesia with isoflurane (TEM). A total of 1.3 10
9
PFU of
AdTGF-
1, AdLaCZ, or AdDL were administered in a volume of 800
l
of NaCl 0.9%, without any surgery, by intrapleural injection on the right
side (sixth space) with a 20-G needle, animals in a left lateral decubitus
position. For coadministration experiments, rats received, in 800
lof
0.9% NaCl, 1 10
9
PFU AdLacZ plus 1.3 10
9
PFU AdTGF-
1or1
10
9
PFU AdLacZ plus 1.3 10
9
PFU AdDL. Rats were euthanized by
abdominal aortic bleeding at days 4, 7, 14, 21, and 64 after adenoviral
administration. After slight incision through the diaphragm, 2.5 ml of 0.9%
NaCl were injected in the pleural space. The pleural lavage fluid (PLF) was
retrieved with a 1-ml needle and maintained on ice until further processing.
A canula was then placed into the trachea, the lungs were removed en bloc
and bronchoalveolar lavage (BAL) was performed as previously described
(21). Six milliliters of NaCl 0.9% was slowly injected intratracheally, re-
trieved, and maintained on ice. The lungs were connected through the
canula to a column filled with 4% formalin with a constant pressure of 20
cm H
2
O for 10 min. The total volume of formalin required to inflate the
lungs was considered as an estimate of total lung volume. Lungs, dia-
phragm, and chest wall were placed in 4% formalin for 24 h.
PLF and BAL fluid (BALF) were centrifuged at 2500 rpm for 15 min.
After removal of cell and debris pellets, supernatants were stored at 80°C
until further use. The pellets were resuspended in 1 ml of 0.9% NaCl.
Cytospins (200
l, 300 rpm, 2 min) were done in a cytocentrifuge (Cyto-
spin 4; Thermo Shandon) and stained with Giemsa (Sigma-Aldrich) for
differential cell count.
Determination of TGF-
1 levels
Active TGF-
1 was determined from BALF and PLF supernatants using
an ELISA kit for human TGF-
1 (R&D Systems), performed according to
the recommendations of the manufacturer. The sensitivity of this assay is
7 pg/ml.
-galactosidase staining
Cytochemical staining for
-galactosidase was performed on samples ob-
tained from four animals per time point after intrapleural injection of
AdLacZ, AdLacZ plus AdTGF-
1, or AdLacZ plus AdDL. After1hin
fixative (2% formaldehyde/0.2% glutaraldehyde), fresh tissue samples
were stained for6hinasolution containing potassium ferrous cyanide,
potassium ferric cyanide, magnesium chloride, Triton X-100 and 5-bromo-
4-chloro-3-indolyl-
-D-galactopyranoside (X-Gal; Sigma-Aldrich). The
samples were stored in 70% ethanol and then paraffin-processed and
-embedded. Five-micrometer sections were counterstained with nuclear
fast red as previously described (22).
Histology
Immunohistochemistry. Transverse sections of the lung were paraffin-
embedded, 5-
m sectioned, and stained with H&E, Masson-Trichrome and
Picrosirius Red or processed for collagen and heat shock protein (HSP) 47
immunohistochemistry. The primary Abs were a mouse monoclonal
(COL-1) to colligin-I (Abcam) and a mouse anti-HSP47 (colligin) mAb
(Stressgen; TebuBio); the secondary Abs were, respectively, a goat anti-
mouse IgG biotin conjugated (Chemicon International) and a biotinylated
goat anti-mouse/rabbit Ig (DakoCytomation). After peroxidase inhibition
(PBS plus H
2
O
2
, 20 min), tissue sections were incubated with the primary
Ab (1/50 dilution for collagen I and 1/500 dilution for HSP47, overnight at
4°C in humidity chamber). Tissue sections were then incubated with the
secondary Ab (1/500 dilution for collagen-I and 1/250 dilution for HSP47,
1 h). The streptavidin-HRP complex (DakoCytomation) was applied (1/300
dilution for collagen-I and 1/500 dilution for HSP47) during 45 min at
room temperature. 3-Amino-9-ethylcarbazole/hydrogen peroxide was used
as chromogen substrate. Slides were counterstained with hematoxylin.
For the purpose of being as distant from the intrapleural injection site
as possible, we always used the left lung for histology and morpho-
metric analysis.
Immunofluorescence. Dual staining with Abs to
-smooth muscle actin
(
-SMA) and cytokeratin was performed using fluorescently labeled Abs.
After Ag retrieval in boiling 10 mM citrate buffer (pH 6.0) for 45 min,
sections were incubated with
-SMA Ab (DakoCytomation) followed by a
secondary rabbit anti-mouse Ab labeled with Texas Red (Molecular
Probes). Sections were stained with a FITC-labeled Ab to pancytokeratin
(Sigma-Aldrich) and mounted with a 4,6-diamidino-2-phenylindole
(DAPI) nuclear stain (Vectashield; Vector Laboratories). Negative control
for immunofluorescent staining were conducted using IgG2a (DakoCyto-
mation) control Ab substituted for anti-
-SMA or normal goat serum sub-
stituted for anti-pancytokeratin. These sections were viewed with a Leica
DMR fluorescence microscope (Leica Microsystems).
FIGURE 1. Adenovirus-mediated gene transfer to the pleural me-
sothelium is transient and limited to the pleura. Lungs were fixed and
stained for
-galactosidase activity 4, 10, 14, and 21 days after AdLacZ
administration. A, Extensive uniform expression (blue staining) of ad-
enoviral gene product was seen until day 10 but had completely disap-
peared by day 21. B, Four days after AdLacZ intrapleural injection,
section of the lung demonstrated
-galactosidase staining limited to the
mesothelial cell monolayer. Gene transfer was also effective in me-
sothelial cells from the chest wall and the diaphragm (chest side only).
Sections were counterstained with nuclear fast red. No
-galactosidase
activity was observed in control vector AdDL-treated animals (A and
B); n 4 animals per day.
6044 TGF-
1-INDUCED PLEURAL FIBROSIS
Pleural thickness assessment
The pleural thickness was measured by histomorphometric measurement
on lung sections stained with Masson-Trichrome (200 times). Twenty-five
random measures per lung section were obtained for each animal using an
Eclipse E600 microscope (Nikon). Video images were captured with a 3
CCD color video camera (Sony/Nikon) and analyzed using an image an-
alyzing system (Archimed; Microvision Instruments).
Collagen quantitation
Collagen amount was analyzed on paraffin sections stained with Picrosirius
Red (20 times) as previously described (23, 24). Briefly, 25 random fields
for each animal were digitized under polarized transmission illumination.
The percentage of emission was quantified (morphometry software from
Histolab/Microvision Instruments) as a reflection of percentage collagen.
Collagen intensity in the pleura was measured within a rectangle deter-
mined by a constant length (200
m), the width depending on pleura thick-
ness. Collagen content was expressed as the percent of emission multiplied
by the surface of each rectangle. Collagen amount within the pulmonary
parenchyma was measured using circles randomly disposed at a constant
distance from the pleural surface (see Fig. 5A). Each circle had a constant
diameter of 640
m. Circle 1 was below the pleura. Circle 2 was deeper in
the parenchyma, directly underneath circle 1 in a perpendicular way to the
pleura. Large vessels and airways were excluded. The collagen content
within circles was expressed as the percent of emission.
Zymography
The hydrolytic activity of matrix metalloproteinase (MMP)-2 and MMP-9
in PLF and BAL was measured by gelatin zymography. Samples were
separated by 10% SDS-PAGE containing 0.1% gelatin (Sigma-Aldrich).
After electrophoresis, the gels were incubated in 2.5% Triton X-100 (Sigma-
Aldrich) for 30 min and were then placed in the activating buffer (50
mM Tris-HCl (pH 8), 10 mM CaCl
2
,5
M ZnSO
4
, 150 mM NaCl; Sigma-
Aldrich), overnight at 37°C. The gels were stained with 0.1% Coomassie
brilliant blue-250 solution (Sigma-Aldrich), during 30 min at 37°C. The
gels were then destained with several changes of 40% methanol and 7%
acetic acid. Zones of enzymatic activity were evident as clear bands against
blue background. Reference standards used were MMP-2 and MMP-9
(Chemicon International).
Statistical analysis
Comparisons between the AdTGF-
1-treated group and the control group
(AdDL) were performed by Mann-Whitney U test and comparisons be-
tween rats of the same group were performed by Wilcoxon test.
Results
Bilateral and transient gene transfer in rat pleura after right
intrapleural injection
Lungs were harvested en bloc at various time points after AdLacZ
administration into the right pleural space and stained for
-ga-
lactosidase activity (Fig. 1A). Adenovirus was highly efficient
in transfecting mesothelial cells, as demonstrated by wide-
spread expression of
-galactosidase in the visceral pleura by
days 4 and 10 (Fig. 1B). Mesothelial cells from the chest wall
and the diaphragm were transfected as well. The transgenic pro-
tein was strictly limited to pleural mesothelial cells and was not
observed within the pulmonary parenchyma (Fig. 1B)orinthe
peritoneal cavity. Gene transfer was transient and disappeared
over a period of 14 days. No
-galactosidase activity was found
in control rats (Fig. 1). Gene transfer was bilateral and uniform
due to anatomical connections between the right and left pleural
space in rodents.
FIGURE 2. TGF-
1 pleural fibrosis is associated with a severe lung
volume restriction: AdTGF-
1-treated rat lungs appeared much smaller
than their controls by day 64. For inflation of the lungs from AdTGF-
1-
treated rats, the volume of formalin needed at a constant pressure of 20 cm
H
2
O for 10 min, was significantly smaller than that of control rats by day
64; p 0.034; n 5 AdTGF-
1 and 3 AdDL.
FIGURE 3. TGF-
1 gene transfer to the
pleural mesothelium induces transient active
TGF-
1 production in PLF (ELISA): a
strong TGF-
1 increase in PLF was detected
by days 4 and 7 after AdTGF-
1 adminis-
tration but no longer by day 14. No TGF-
1
overexpression was detected in PLF from
control rats. In BALF from AdTGF-
1-
treated rats, only a slight TGF-
1 increase
was detected by day 7. , p 0.034 com-
pared with AdDL animals; n 5 AdTGF-
1
and 3–4 AdDL per day.
Table I. Cell count of BALF and PLF from rats treated with AdDL
or AdTGF-
1
a
Day
Total Cell Count (10
4
)
AdDL AdTGF-
1
PLF BALF PLF BALF
4 767 22 37 10 1953 1510 43 3
7 4900 1600 33 7 1133 402 44 9
14 2900 1888 58 9 1143 933 45 5
21 600 164 77 22 373 190 88 15
64 255 100 97 23 180 59 85 17
a
The cell pellets remaining after centrifugation of the BALF or PLF samples
were resuspended in 0.9% NaCl. The numbers of cells were counted by using a
hemocytometer. Cell differentials were determined with at least 300 cells. No
significant difference was observed between AdTGF-
1 and AdDL in total cell
count in PLF as well as in BALF. No difference was observed in differential count
in PLF with a majority of mononucleated cells.
6045The Journal of Immunology
TGF-
1 adenovector-mediated gene transfer to the pleura
Rats treated with the control virus (AdDL) were healthy and steadily
gained weight. In contrast, rats treated with AdTGF-
1 appeared sick
with ruffled fur and lost up to 15% of body weight in the first 2 wk
before recovering. Some animals had to be euthanized after
AdTGF-
1 administration due to poor condition and slow recovery.
These animals demonstrated fibrosis of pleura but were not included
in data analysis, because they did not reach the determined endpoints.
FIGURE 4. AdTGF-
1 transient gene transfer to the pleura induces a progressive pleural fibrosis. A, Four days after AdTGF-
1 administration, there
was already a slight collagen deposition with a thickened pleura. The collagen accumulation was increasing up to day 64 with a thick, dense pleura and
thickened alveolar wall underneath the pleura. In control rats, the pleura appeared normal with no collagen deposition at any time point (Masson-Trichrome,
200 times). HSP47 is strongly overexpressed from days 4 to 64 in AdTGF-
1-treated rats compared with control rats (HSP47 immunohistochemistry, 200
times). B, By histomorphometric measurement (collagen intensity, Picrosirius Red, 20 times, under polarized transmission illumination), the collagen
accumulation within the pleura was progressive up to day 64 (bar chart). The increased thickness (Masson-Trichrome, 200 times) was sustained up to day
64 (line). n 5 AdTGF-
1 and 3– 4 AdDL per day; p 0.01 for each day in both experiments when ADTGF-
1 was compared with AdDL rats from
the corresponding day. , p 0.05 compared with day 4 for collagen accumulation and thickness. C, Sixty-four days after intrapleural AdTGF-
1
administration, there was a collagen accumulation on the chest surface of the diaphragm. The chest wall surface demonstrated a moderate collagen
accumulation strictly limited to the surface.
6046 TGF-
1-INDUCED PLEURAL FIBROSIS
Macroscopically, the visceral pleural surface of AdTGF-
1-
treated lungs appeared at any examined time point homogenously
white. AdTGF-
1-treated lungs looked abnormal with shrinkage
when compared with control, an observation which was progres-
sive until day 64 (Fig. 2). Interestingly, the parietal pleura on the
chest wall did not look different between AdTGF-
1- and AdDL-
treated rats. Adhesions were rare and blunt dissection between
parietal and visceral pleura was easy. Lung volumes were assessed
by measuring the volume of formalin that drained into the lungs
after 10 min at a constant pressure of 20 cm H
2
O. Volumes were
reduced by 30% in AdTGF-
1-treated lungs compared with con-
trol by day 64 (7.45 0.76 ml and 10.17 0.17 ml, respectively,
p 0.034).
Intrapleural administration of AdTFG-
1 induced local and
transient overexpression of active TGF-
1
The concentration of transgenic protein was measured by ELISA
for active human TGF-
1 protein in the supernatants of PLF
and BALF. Analysis of PLF from AdTGF-
1-treated rats (Fig.
3) revealed significantly increased levels of active TGF-
1in
the pleural space by days 4 and 7 (4717 785 pg/ml and
2872 594 pg/ml, respectively) compared with AdDL control
rats (61 9 and 48 2 pg/ml, respectively). The transgenic
product was no longer detectable after day 14. In BALF, a small
increase in TGF-
1 was detected by day 7 in AdTGF-
1-
treated rats compared with control (332 103 pg/ml and 60
1 pg/ml, respectively, p 0.034).
AdTGF-
1-induced pleural effusion
By day 4, we found significant pleural effusion associated with
high concentration of TGF-
1 (3.3 1.1 ml, p 0.025 compared
with control). The volume of pleural effusion decreased over time
(1.1 0.6 ml by day 7, p 0.025 when compared with controls),
and was no longer present by day 14. No pleural effusion was
observed in control vector-treated animals at any time point. Total
cells in PLF were increased by days 4, 7, and 14 compared with
later time points, with no significant difference between AdDL-
and AdTGF-
1-treated rats (Table I). There was a predominance
of mononucleated cells in the differential cell count (98%) at
each time point in all AdDL- and AdTGF-
1-exposed animals. No
difference was observed in BALF cell count at any time point
between AdDL- and AdTGF-
1-treated animals.
TGF-
1 gene transfer induces bilateral and progressive fibrosis
Sections of left and right lung from the same animal were assessed
by histomorphometry to compare visceral pleura thickness be-
tween both sides after intrapleural AdTGF-
1 administration (into
FIGURE 5. AdTGF-
1 transient gene transfer to the pleural mesothe-
lium induces a progressive increase in collagen amount within the pulmo-
nary parenchyma. A, Sixty-four days after AdTGF-
1 administration, col-
lagen I (brown color) is strongly overexpressed within the pleura and
within the pulmonary parenchyma (collagen I immunohistochemistry, 200
times). B, Collagen levels were randomly measured at constant distance
from the pleura (Picrosirius Red staining, 20 times, under polarized trans-
mission illumination, as shown on the picture). Each circle has a 640-
m
diameter. Circle 1 is the one against the pleura. Circle 2 is deeper in the
parenchyma, directly underneath circle 1 in a perpendicular way to the
pleura. Large vessels and airways were excluded. C, Results are expressed
as mean quantity of collagen in AdTGF-
1-treated rats reported to these of
control rats (n 5 AdTGF-
1 and 3 AdDL); p 0.01 for each day when
ADTGF-
1 was compared with AdDL rats from the corresponding day.
, p 0.05 compared with AdTGF-
1 animals from days 4, 7, and 14.
FIGURE 6. TGF-
1 induces mesothelial cell migration within the fibrotic tissue. After AdLacZ/AdDL coadministration, transfected mesothelial cells
expressing
-galactosidase by day 6 are on a single monolayer as normally observed. After AdLacZ/AdTGF-
1 coadministration, mesothelial cells
transfected by day 0 with AdLacZ (blue staining) are still localized by day 2 on a single monolayer but clearly start to change the phenotype from a flat
to a more spindle like shape (please compare with Fig. 1B which shows cells exposed to AdLacZ but not AdTGF-
1 at the same 400 magnification).
These cells migrate into the fibrotic tissue by day 6 when TGF-
1 was overexpressed.
6047The Journal of Immunology
the right chest cavity). The findings demonstrate that pleural fi-
brosis after intrapleural administration of AdTGF-
1 is bilateral
and homogeneous (data not shown), due to anatomical connections
between the two cavities in rodent lung. Consequently, we decided
to perform all histomorphometric measurements on the left lung to
be distant from the injection side and potential local irritation of
the pleura by inflammatory cells or hemorrhage. We found that
pleural thickening began as early as at day 4 and progressively
increased through days 21 and 64 (Fig. 4A). Similarly, the amount
of collagen within the thickened pleura as assessed by picrosirius
red staining (a marker specific for collagen) was progressively
increased up to day 64 (Fig. 4B). The pleura appeared normal in
AdDL-treated rats at any time point without any indication for
collagen accumulation. The parietal pleura of the diaphragm
showed fibrotic changes with a uniform collagen deposition de-
spite normal aspect on visual examination (Fig. 4C). There was
no fibrosis on the peritoneal side of the diaphragm. The parietal
pleura of the chest wall showed even less collagen accumula-
tion (Fig. 4C).
HSP47, a collagen-specific chaperon and closely associated with
de novo synthesis of collagen, was present in the fibrotic tissue
from day 4 on through to day 64 indicating an active fibroprolif-
erative process (Fig. 4A).
Increase in the collagen level in the pulmonary parenchyma
Although fibrotic changes were strictly limited to the surface of
diaphragm and chest wall and exclusively pleural, they appeared
markedly different on the visceral pleura (Fig. 4A). Indeed, the
fibrotic changes invaded the lung parenchyma adjacent to the pleu-
ral surface (Fig. 5, A and B). We measured collagen density within
the parenchyma at a constant distance from the pleural surface and
demonstrated that intrapleural TGF-
1 gene transfer induces a
moderate but significant and progressive fibrotic response within
the pulmonary parenchyma. Collagen accumulation started right
underneath the pleura where it was most intense (Fig. 5C, circle 1)
and it increased progressively up to day 64. The amount of paren-
chymal collagen accumulation decreased with increasing distance
from the surface (Fig. 5C, circle 2) but was still progressive
over time.
Mesothelial-fibroblastoid transformation
In experiments with coadministration of AdLacZ and AdTGF-
1,
we observed that mesothelial cells transfected by day 0 with
AdLacZ (blue staining) progressively migrated into the fibrotic
tissue when TGF-
1 was overexpressed (Fig. 6). The mesothelial
cell layer contained abundant cytokeratin-positive cells (Fig. 7,
thin arrow) from day 4 to 64. By day 4 after AdTGF-
1, but not
AdDL, mesothelial cells became round and lost their intercellular
connection (Figs. 6 and 7). Seven and 14 days after AdTGF-
1,
immunofluorescence demonstrated the appearance of dual-labeled
cytokeratin and
-SMA-positive cells (Fig. 7, thick arrow) within
the fibrotic tissue. By day 64 numerous
-SMA-positive cells were
observed within the parenchyma underneath the strong pleural fi-
brotic tissue.
These phenotypical changes observed in mesothelial cells were
accompanied by a strong increase in gelatinolytic activity in the
FIGURE 7. TGF-
1 induces phenotypic transition. Immunofluores-
cence from pleural sections stained for cytokeratin (green),
-SMA (red),
with nuclear counterstain (blue, DAPI). The mesothelial cell layer demon-
strates cytokeratin-positive cells (thin arrows) from days 4 to 64. By days
7 and 14 after AdTGF-
1, there was dual-stained cytokeratin and
-SMA-
positive cells (thick arrows). By day 64, numerous
-SMA-positive cells
(dotted arrows) were observed within the parenchyma. Magnification,
400 for days 4, 7, and 14; 200 for day 64.
FIGURE 8. TGF-
1 induces a local MMP activity (gel-
atin zymography from PLF): AdTGF-
1 induces an in-
crease in MMP-2 activity by days 4, 7, and 14 and in
MMP-9 activity by day 4, compared with control rats (n
5 AdTGF-
1 and 3 AdDL per day).
6048 TGF-
1-INDUCED PLEURAL FIBROSIS
pleural fluid, which is a critical factor in the process of mesothe-
lial-to-mesenchymal transition. MMP-2 activity was markedly in-
creased as determined by gelatin zymography in PLF from
AdTGF-
1 compared with control-treated rats from days 4 to 14
and MMP-9 activity was increased by day 4 (Fig. 8).
Discussion
Pleural fibrosis is usually considered as complication of other dis-
orders involving the chest cavity, and it is unclear why only some
individuals develop progressive pleural scarring in response to in-
jury. In this study, we show that active TGF-
1 administered to the
rat pleural mesothelium by adenoviral gene transfer leads to pro-
gressive pleural fibrosis without pleurodesis, but with severe lung
volume restriction. We further report that pleural fibrotic changes
are associated with MFT and advance significantly into the lung
parenchyma, but not into adjacent chest wall or diaphragmatic
muscle.
Pleural fibrosis can develop in response to a large variety of
diseases and tissue injury. These are frequently but not exclusively
associated with inflammation. The majority of pleural inflamma-
tory processes resolves with treatment of the underlying cause or
improves spontaneously. However, sometimes chronic scarring
and fibrosis develops despite obvious resolution of the inflamma-
tory phase (1). Progressive scarring can also occur without major
inflammation, such as after surgical thoracotomy for coronary by-
pass (25). The trigger for switching a resolving to a progressively
fibrotic tissue response in the pleural cavity has not yet been iden-
tified. Several studies have demonstrated a potential role of the
growth factors basic fibroblast growth factor, platelet-derived
growth factor, and TGF-
1 (26). TGF-
1 has been widely studied
in the context of fibrotic diseases. This growth factor is known for
its anti-inflammatory effects, is chemotactic for fibroblasts and
promotes the accumulation of extracellular matrix (27). Transient
overexpression of active TGF-
1 in rat lungs by adenoviral gene
transfer causes progressive lung fibrosis without major inflamma-
tion, characterized by extensive deposition of extracellular matrix
proteins such as collagen and fibronectin and by accumulation of
myofibroblasts (20). In contrast, adenovector-mediated overex-
pression of IL-1
induces a severe inflammatory response in the
lung followed by progressive fibrosis, likely mediated through en-
dogenous up-regulation of TGF-
1 (21, 24). The profibrotic effects
of TGF-
do not only apply to bronchial and alveolar epithelial
cells but also to mesothelial cells (28). TGF-
stimulation causes
mesothelial cells to synthesize excessive collagen and other matrix
proteins in vitro (28). In the study reported here, we administered
the gene for spontaneously active TGF-
1 into the right pleural
cavity of rats and generated high levels of active protein in the
pleural fluid, with peak expression between days 4 and 7. This
approach resulted in severe and progressive pleural fibrosis. There
was a moderate mononuclear inflammatory response to the ade-
novirus with no difference between AdTGF-
1 and control vector-
treated rats. AdTGF-
1-induced fibrosis was progressive up to 64
days, long after the transgenic protein has disappeared from the
pleural fluid. We assume that this progressive fibrogenic response
is partly mediated by TGF-
autoinduction similar to what has
been discussed in lung fibrosis (20). It has recently been shown
that intrapleural injection of TGF-
2 stimulates mesothelial cells
to produce collagen and endogenous TGF-
, further increasing the
production of matrix proteins (10). Human mesothelial cells are
not only able to synthesize TGF-
but also have receptors for this
cytokine (11, 29), further supporting the hypothesis of autocrine
activation of TGF-
in pleural fibrosis.
The focus of most pleural fibrosis research is related to induc-
tion of adhesions and fibrotic changes with the clinical purpose of
pleurodesis, mostly for treatment of malignant or chronic and ther-
apy resistant pleural effusion (7). Talc and tetracycline instillation
into the chest cavity are the most common approaches to pleu-
rodesis (30). However, they are associated with significant clinical
problems such as high fever and pain. Even adult respiratory dis-
tress syndrome has been recognized as a complication in up to 9%
of patients receiving talc pleurodesis (31, 32). Hence, profibrotic
growth factors are considered as potential alternate therapies and
investigated in animal models. Lee et al. (8, 10) showed in a rabbit
or a sheep model that rTGF-
2 administration into the pleural
space induces initially marked fluid effusion, which is followed by
effective pleurodesis with multiple adhesions. In contrast to this
study, we noticed in our rat model only transient pleural effusion
at very early time points, followed by progressive fibrosis of the
pleura, but adhesions were almost absent (Fig. 2 and 4A). Both
these models support a major role for TGF-
in the development
and progression of pleural fibrosis, although the difference be-
tween TGF-
1 and 2 is somewhat unexpected. TGF-
1 and 2 are
thought to have similar effects on the formation of matrix, but the
route of administration may account for differences in the forma-
tion of adhesions (TGF-
1 through adenoviral gene transfer as
opposed to recombinant protein for the TGF-
2 studies) (8 –10).
We believe that the technique applied for intrapleural injection
may have had the most impact on the pleurodesis. Our experimen-
tal approach does not involve surgery and allows avoiding major
bleeding and additional inflammatory factors which can lead to
adhesions and pleurodesis (33). Finally, pleural adhesion and pleu-
ral thickening may be governed by different mechanisms.
Surprisingly, the value of anti-TGF-
strategies in the treatment
of pleural fibrosis and adhesions has not been investigated thor-
oughly. TGF-
is one of the major targets in the development of
antifibrotic therapies for pulmonary fibrosis, and a number of ex-
perimental studies in animals strongly support this approach (34,
35). In our model, we induced severe and homogenous bilateral
fibrosis of the visceral pleura, leading to significant restriction of
lung volumes. Pleural fibrosis in humans becomes clinically sig-
nificant with a restrictive pattern in lung function when it involves
major parts of the visceral pleura. This can cause substantial mor-
bidity and chronic respiratory failure, e.g., in asbestos lung, after
thoracic surgery, or hemothorax, in drug-induced pleural fibrosis
(ergots drug, previous chemotherapy for cancer, radiation therapy)
or in familial idiopathic pleural fibrosis. No effective medical treat-
ment is available for these conditions and surgical decortication is
frequently required, with limited therapeutic success (1). Anti-
TGF-
strategies are a promising therapeutic approach for preven-
tion or cure these complications. One study has shown that in-
trapleural injection of TGF-
Abs can reduce empyema-induced
pleural fibrosis in a rabbit model (11, 12). Although the role of
TGF-
in other pleural fibrotic disorders is not definitively proven,
we believe that our model could be very useful to further under-
stand how to halt the progression or even cure this chronic
condition.
The pattern and distribution of pleural fibrosis in our model was
different than expected, raising some interesting questions and hy-
potheses that need further exploration. Fibrotic changes were
strongest on the visceral pleura, characterized by increasing accu-
mulation of collagen by day 64. Less but still substantial fibrotic
thickening was seen on the parietal pleura of the diaphragm, and
only minor collagen was detectable on the parietal chest wall
pleura. Although no fibrosis was seen in the muscle of diaphragm
and chest wall, we interestingly found a significant increase of
collagen within the pulmonary parenchyma adjacent to the pleural
surface (Fig. 5) which diminished with farther distance from the
pleura. The presence of a strong positivity of subpleural HSP47 up
6049The Journal of Immunology
to day 64 (Fig. 4A) supports an ongoing fibroproliferative process
at this time point (36). It may be that diffusion of transgenic TGF-
from the pleural space into alveoli immediately underneath the
pleura contributed to this phenomenon. However, this would only
partly explain the progression of these changes over time as the
transgenic protein disappears within less than two weeks. We pro-
pose that the intrapulmonary collagen accumulation seen after in-
trapleural overexpression of TGF-
1 may also result from a pro-
gression of pleural fibrosis into the alveolar structure of the lungs.
Subpleural fibrosis may be result of a response characteristic for
the compartment lung, as Sime et al. (20) suggested earlier when
intrapulmonary overexpression of active TGF-
1 in rats resulted
in severe interstitial fibrosis starting centrally in the lung and even-
tually involved the visceral, but not parietal, pleura. Subpleural
fibrosis is one of the hallmarks of usual interstitial pneumonia (37).
Fibroblastic foci dominate in the earlier stages of disease, being a
sign for an ongoing fibrotic process, whereas honeycombing is
more indicative for advanced and “burned out” disease (38). It has
just been reported that fibroblastic foci may communicate much
closer with each other than anticipated. Cool et al. (39) suggest that
they may even form a network of fibroblasts, a “fibroblastic retic-
ulum” extending from the pleura to the underlying parenchyma.
The interconnecting fibroblasts described by this group were not
monoclonal, thus they likely result from a reactive rather than neo-
plastic response to tissue injury. The observation in our study fits
well into this concept, and we believe that our model will be ex-
tremely useful to investigate this hypothesis in detail. Further in-
dication for a potential modulating role of the pleura in fibrotic
lung disease is the ability of pleural mesothelial cells to transform
into myofibroblast-like cells. EMT is a central mechanism for di-
versifying the cells found in complex tissues (40). It is involved in
a variety of normal physiological as well as pathological processes
such as cancer progression and renal fibrosis (16, 17). Recent ev-
idence suggests a potential role for EMT in the pathogenesis of
pulmonary fibrosis (14, 15, 41– 43). MFT is a similar process and
has been described as a major factor in the development of peri-
toneal fibrosis (18). We demonstrate clear evidence that MFT oc-
curs in our model of AdTGF-
1-induced pleural fibrosis, and that
the mesenchymal cells derived through this process invade into the
lung parenchyma. We did not find obvious fibroblastic foci in our
model. Further research is needed to investigate whether the sub-
pleural distribution pattern of pulmonary fibrosis may be in part
related to a pleural process or involvement of mesothelial cells in
this disease.
In summary, we here present a novel model to examine the
pathogenesis of pleural fibrosis using transient adenoviral vector-
mediated overexpression of TGF-
1. The fibrotic process is pro-
gressive and interestingly invades into the subpleural zone of the
lung parenchyma. This is associated with MFT, and it can be spec-
ulated that the pleural mesothelium may be involved in the sub-
pleural distribution pattern of pulmonary fibrosis.
Acknowledgments
We thank V. San Giorgio and all the team from the animal quarter for their
invaluable and professional help.
Disclosures
The authors have no financial conflict of interest.
References
1. Jantz, M. A., and V. B. Antony. 2006. Pleural fibrosis. Clin. Chest Med. 27:
181–191.
2. Antony, V. B. 1998. Drug-induced pleural disease. Clin. Chest Med. 19:
331–340.
3. Camus, P., P. Bonniaud, A. Fanton, C. Camus, N. Baudaun, and P. Foucher.
2004. Drug-induced and iatrogenic infiltrative lung disease. Clin. Chest Med. 25:
479 –519, vi.
4. Huggins, J. T., and S. A. Sahn. 2004. Causes and management of pleural fibrosis.
Respirology 9: 441– 447.
5. Mutsaers, S. E., C. M. Prele, A. R. Brody, and S. Idell. 2004. Pathogenesis of
pleural fibrosis. Respirology 9: 428 440.
6. Antony, V. B. 2003. Immunological mechanisms in pleural disease. Eur. Respir.
J. 21: 539 –544.
7. Lee, Y. C., and R. W. Light. 2004. Management of malignant pleural effusions.
Respirology 9: 148 –156.
8. Lee, Y. C., K. B. Lane, R. E. Parker, D. S. Ayo, J. T. Rogers, R. W. Diters,
P. J. Thompson, and R. W. Light. 2000. Transforming growth factor
2
(TGF
2
)
produces effective pleurodesis in sheep with no systemic complications. Thorax
55: 1058 –1062.
9. Lee, Y. C., C. J. Devin, L. R. Teixeira, J. T. Rogers, P. J. Thompson, K. B. Lane,
and R. W. Light. 2001. Transforming growth factor
2 induced pleurodesis is not
inhibited by corticosteroids. Thorax 56: 643– 648.
10. Lee, Y. C., D. Malkerneker, C. J. Devin, P. J. Thompson, J. E. Johnson,
K. B. Lane, and R. W. Light. 2001. Comparing transforming growth factor
-2
and fibronectin as pleurodesing agents. Respirology 6: 281–286.
11. Sasse, S. A., M. R. Jadus, and G. D. Kukes. 2003. Pleural fluid transforming
growth factor-
1 correlates with pleural fibrosis in experimental empyema.
Am. J. Respir. Crit. Care Med. 168: 700 –705.
12. Kunz, C. R., M. R. Jadus, G. D. Kukes, F. Kramer, V. N. Nguyen, and
S. A. Sasse. 2004. Intrapleural injection of transforming growth factor-
antibody
inhibits pleural fibrosis in empyema. Chest 126: 1636 –1644.
13. Blobe, G. C., W. P. Schiemann, and H. F. Lodish. 2000. Role of transforming
growth factor
in human disease. N. Engl. J. Med. 342: 1350 –1358.
14. Yao, H. W., Q. M. Xie, J. Q. Chen, Y. M. Deng, and H. F. Tang. 2004. TGF-
1
induces alveolar epithelial to mesenchymal transition in vitro. Life Sci. 76:
29 –37.
15. Willis, B. C., R. M. duBois, and Z. Borok. 2006. Epithelial origin of myofibro-
blasts during fibrosis in the lung. Proc. Am. Thorac. Soc. 3: 377–382.
16. Yang, A. H., J. Y. Chen, and J. K. Lin. 2003. Myofibroblastic conversion of
mesothelial cells. Kidney Int. 63: 1530 –1539.
17. Yanez-Mo, M., E. Lara-Pezzi, R. Selgas, M. Ramirez-Huesca, C. Dominguez-
Jimenez, J. A. Jimenez-Heffernan, A. Aguilera, J. A. Sanchez-Tomero,
M. A. Bajo, V. Alvarez, et al. 2003. Peritoneal dialysis and epithelial-to-mesen-
chymal transition of mesothelial cells. N. Engl. J. Med. 348: 403– 413.
18. Margetts, P. J., P. Bonniaud, L. Liu, C. M. Hoff, C. J. Holmes, J. A. West-Mays,
and M. M. Kelly. 2005. Transient overexpression of TGF-
1 induces epithelial
mesenchymal transition in the rodent peritoneum. J. Am. Soc. Nephrol. 16:
425– 436.
19. Bett, A. J., W. Haddara, L. Prevec, and F. L. Graham. 1994. An efficient and
flexible system for construction of adenovirus vectors with insertions or deletions
in early regions 1 and 3. Proc. Natl. Acad. Sci. USA 91: 8802–8806.
20. Sime, P. J., Z. Xing, F. L. Graham, K. G. Csaky, and J. Gauldie. 1997. Ad-
enovector-mediated gene transfer of active transforming growth factor-
1 in-
duces prolonged severe fibrosis in rat lung. J. Clin. Invest. 100: 768 –776.
21. Kolb, M., P. J. Margetts, D. C. Anthony, F. Pitossi, and J. Gauldie. 2001. Tran-
sient expression of IL-1
induces acute lung injury and chronic repair leading to
pulmonary fibrosis. J. Clin. Invest. 107: 1529 –1536.
22. Margetts, P. J., M. Kolb, T. Galt, C. M. Hoff, T. R. Shockley, and J. Gauldie.
2001. Gene transfer of transforming growth factor-
1 to the rat peritoneum:
effects on membrane function. J. Am. Soc. Nephrol. 12: 2029 –2039.
23. Bonniaud, P., M. Kolb, T. Galt, J. Robertson, C. Robbins, M. Stampfli,
C. Lavery, P. J. Margetts, A. B. Roberts, and J. Gauldie. 2004. Smad3 null mice
develop airspace enlargement and are resistant to TGF-
-mediated pulmonary
fibrosis. J. Immunol. 173: 2099 –2108.
24. Bonniaud, P., P. J. Margetts, K. Ask, K. Flanders, J. Gauldie, and M. Kolb. 2005.
TGF-
and Smad3 signaling link inflammation to chronic fibrogenesis. J. Immu-
nol. 175: 5390 –5395.
25. Areno, J. P., J. P. McCartney, J. Eggerstedt, W. Grafton, and R. B. George. 1998.
Persistent pleural effusions following coronary bypass surgery. Chest 114:
311–314.
26. Mutsaers, S. E., I. Kalomenidis, N. A. Wilson, and Y. C. Lee. 2006. Growth
factors in pleural fibrosis. Curr. Opin. Pulm. Med. 12: 251–258.
27. Bonniaud, P., P. J. Margetts, M. Kolb, T. Haberberger, M. Kelly, J. Robertson,
and J. Gauldie. 2003. Adenoviral gene transfer of connective tissue growth factor
in the lung induces transient fibrosis. Am. J. Respir. Crit. Care Med. 168:
770 –778.
28. Lee, Y. C., and K. B. Lane. 2001. The many faces of transforming growth fac-
tor-
in pleural diseases. Curr. Opin. Pulm. Med. 7: 173–179.
29. Gerwin, B. I., J. F. Lechner, R. R. Reddel, A. B. Roberts, K. C. Robbins,
E. W. Gabrielson, and C. C. Harris. 1987. Comparison of production of trans-
forming growth factor-
and platelet-derived growth factor by normal human
mesothelial cells and mesothelioma cell lines. Cancer Res. 47: 6180 6184.
30. Antony, V. B., N. Nasreen, K. A. Mohammed, P. S. Sriram, W. Frank,
N. Schoenfeld, and R. Loddenkemper. 2004. Talc pleurodesis: basic fibroblast
growth factor mediates pleural fibrosis. Chest 126: 1522–1528.
31. Brant, A., and T. Eaton. 2001. Serious complications with talc slurry pleurodesis.
Respirology 6: 181–185.
32. Light, R. W. 2002. Talc for pleurodesis? Chest 122: 1506 –1508.
33. Mitchem, R. E., B. L. Herndon, R. M. Fiorella, A. Molteni, C. N. Battie, and
G. R. Reisz. 1999. Pleurodesis by autologous blood, doxycycline, and talc in a
rabbit model. Ann. Thorac. Surg. 67: 917–921.
6050 TGF-
1-INDUCED PLEURAL FIBROSIS
34. Ask, K., G. E. Martin, M. Kolb, and J. Gauldie. 2006. Targeting genes for treat-
ment in idiopathic pulmonary fibrosis: challenges and opportunities, promises
and pitfalls. Proc. Am. Thorac. Soc. 3: 389 –393.
35. Bonniaud, P., P. J. Margetts, M. Kolb, J. A. Schroeder, A. M. Kapoun, D. Damm,
A. Murphy, S. Chakravarty, S. Dugar, L. Higgins, et al. 2005. Progressive trans-
forming growth factor
1-induced lung fibrosis is blocked by an orally active
ALK5 kinase inhibitor. Am. J. Respir. Crit. Care Med. 171: 889 898.
36. Kakugawa, T., H. Mukae, T. Hayashi, H. Ishii, K. Abe, T. Fujii, H. Oku,
M. Miyazaki, J. Kadota, and S. Kohno. 2004. Pirfenidone attenuates expression
of HSP47 in murine bleomycin-induced pulmonary fibrosis. Eur. Respir. J. 24:
57– 65.
37. American Thoracic Society. 2000. Idiopathic pulmonary fibrosis: diagnosis and
treatment. International consensus statement. American Thoracic Society (ATS),
and the European Respiratory Society (ERS). Am. J. Respir. Crit. Care Med. 161:
646 664.
38. Noble, P. W. 2003. Idiopathic pulmonary fibrosis: new insights into classification
and pathogenesis usher in a new era therapeutic approaches. Am. J. Respir. Cell.
Mol. Biol. 29: S27–S31.
39. Cool, C. D., S. D. Groshong, P. R. Rai, P. M. Henson, J. S. Stewart, and
K. K. Brown. 2006. Fibroblast foci are not discrete sites of lung injury or repair:
the fibroblast reticulum. Am. J. Respir. Crit. Care Med. 174: 654 658.
40. Kalluri, R., and E. G. Neilson. 2003. Epithelial-mesenchymal transition and its
implications for fibrosis. J. Clin. Invest. 112: 1776 –1784.
41. Kim, K. K., M. C. Kugler, P. J. Wolters, L. Robillard, M. G. Galvez,
A. N. Brumwell, D. Sheppard, and H. A. Chapman. 2006. Alveolar epithelial cell
mesenchymal transition develops in vivo during pulmonary fibrosis and is reg-
ulated by the extracellular matrix. Proc. Natl. Acad. Sci. USA 103: 13180 –13185.
42. Kasai, H., J. T. Allen, R. M. Mason, T. Kamimura, and Z. Zhang. 2005. TGF-
1
induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir.
Res. 6: 56.
43. Willis, B. C., J. M. Liebler, K. Luby-Phelps, A. G. Nicholson, E. D. Crandall,
R. M. du Bois, and Z. Borok. 2005. Induction of epithelial-mesenchymal tran-
sition in alveolar epithelial cells by transforming growth factor-
1: potential role
in idiopathic pulmonary fibrosis. Am. J. Pathol. 166: 1321–1332.
6051The Journal of Immunology
... Fibrotic foci present within regions of otherwise healthy appearing tissue and fibrosis develops from the subpleural regions inward. [161][162][163] Fibrotic regions can have a broad range of mechanical and biochemical properties with the common feature of myofibroblast persistence. However, most in vitro cell culture systems model the fibrotic microenvironment as a homogenous system. ...
Article
Idiopathic pulmonary fibrosis (IPF) is a severe form of pulmonary fibrosis. IPF is a fatal disease with no cure and is challenging to diagnose. Unfortunately, due to the elusive etiology of IPF and a late diagnosis, there are no cures for IPF. Two FDA-approved drugs for IPF, nintedanib and pirfenidone, slow the progression of the disease, yet fail to cure or reverse it. Furthermore, most animal models have been unable to completely recapitulate the physiology of human IPF, resulting in the failure of many drug candidates in preclinical studies. In the last few decades, the development of new IPF drugs focused on changes at the cellular level, as it was believed that the cells were the main players in IPF development and progression. However, recent studies have shed light on the critical role of the extracellular matrix (ECM) in IPF development, where the ECM communicates with cells and initiates a positive feedback loop to promote fibrotic processes. Stemming from this shift in the understanding of fibrosis, there is a need to develop in vitro model systems that mimic the human lung microenvironment to better understand how biochemical and biomechanical cues drive fibrotic processes in IPF. However, current in vitro cell culture platforms, which may include substrates with different stiffness or natural hydrogels, have shortcomings in recapitulating the complexity of fibrosis. This review aims to draw a roadmap for developing advanced in vitro pulmonary fibrosis models, which can be leveraged to understand better different mechanisms involved in IPF and develop drug candidates with improved efficacy. We begin with a brief overview defining pulmonary fibrosis and highlight the importance of ECM components in the disease progression. We focus on fibroblasts and myofibroblasts in the context of ECM biology and fibrotic processes, as most conventional advanced in vitro models of pulmonary fibrosis use these cell types. We transition to discussing the parameters of the 3D microenvironment that are relevant in pulmonary fibrosis progression. Finally, the review ends by summarizing the state of the art in the field and future directions.
... For histomorphometric assay, the amount of collagen in paraffin-embedded tissue sections was quantified on lung slices from BLM-or NaCl-receiving mice treated or not with NCI-41356 after staining with Picro sirius Red as previously described [28]. ...
Article
Full-text available
Idiopathic pulmonary fibrosis is a chronic, progressive and lethal disease of unknown etiology that ranks among the most frequent interstitial lung diseases. Idiopathic pulmonary fibrosis is characterized by dysregulated healing mechanisms that lead to the accumulation of large amounts of collagen in the lung tissue that disrupts the alveolar architecture. The two currently available treatments, nintedanib and pirfenidone, are only able to slow down the disease without being curative. We demonstrated in the past that HSPB5, a low molecular weight heat shock protein, was involved in the development of fibrosis and therefore was a potential therapeutic target. Here, we have explored whether NCI-41356, a chemical inhibitor of HSPB5, can limit the development of pulmonary fibrosis. In vivo, we used a mouse model in which fibrosis was induced by intratracheal injection of bleomycin. Mice were treated with NaCl or NCI-41356 (six times intravenously or three times intratracheally). Fibrosis was evaluated by collagen quantification, immunofluorescence and TGF-β gene expression. In vitro, we studied the specific role of NCI-41356 on the chaperone function of HSPB5 and the inhibitory properties of NCI-41356 on HSPB5 interaction with its partner SMAD4 during fibrosis. TGF-β1 signaling was evaluated by immunofluorescence and Western Blot in epithelial cells treated with TGF-β1 with or without NCI-41356. In vivo, NCI-41356 reduced the accumulation of collagen, the expression of TGF-β1 and pro-fibrotic markers (PAI-1, α-SMA). In vitro, NCI-41356 decreased the interaction between HSPB5 and SMAD4 and thus modulated the SMAD4 canonical nuclear translocation involved in TGF-β1 signaling, which may explain NCI-41356 anti-fibrotic properties. In this study, we determined that inhibition of HSPB5 by NCI-41356 could limit pulmonary fibrosis in mice by limiting the synthesis of collagen and pro-fibrotic markers. At the molecular level, this outcome may be explained by the effect of NCI-41356 inhibiting HSPB5/SMAD4 interaction, thus modulating SMAD4 and TGF-β1 signaling. Further investigations are needed to determine whether these results can be transposed to humans.
... Possible inhibitory effects of CL40 on leukocyte-mesothelium interaction in lung pathology is of great interest to us. Mesothelial cells show epithelial phenotypes, including epithelial-mesenchymal transition (EMT), in response to transforming growth factor-ß [47,48]. Elucidating the potential role of CL40-positive glycans in the EMT of mesothelial cells may provide clues for understanding the physiological function of mesothelial CL40 glycans. ...
Article
Full-text available
Sialyl 6-sulfo Lewis X (6-sulfo sLeX) and its derivative sialyl 6-sulfo N-acetyllactosamine (LacNAc) are sialylated and sulfated glycans of sialomucins found in the high endothelial venules (HEVs) of secondary lymphoid organs. A component of 6-sulfo sLeX present in the core 1-extended O-linked glycans detected by the MECA-79 antibody was previously shown to exist in the lymphoid aggregate vasculature and bronchial mucosa of allergic and asthmatic lungs. The components of 6-sulfo sLeX in pulmonary tissues under physiological conditions remain to be analyzed. The CL40 antibody recognizes 6-sulfo sLeX and sialyl 6-sulfo LacNAc in O-linked and N-linked glycans, with absolute requirements for both GlcNAc-6-sulfation and sialylation. Immunostaining of normal mouse lungs with CL40 was performed and analyzed. The contribution of GlcNAc-6-O-sulfotransferases (GlcNAc6STs) to the synthesis of the CL40 epitope in the lungs was also elucidated. Here, we show that the expression of the CL40 epitope was specifically detected in the mesothelin-positive mesothelium of the pulmonary pleura. Moreover, GlcNAc6ST2 (encoded by Chst4) and GlcNAc6ST3 (encoded by Chst5), but not GlcNAc6ST1 (encoded by Chst2) or GlcNAc6ST4 (encoded by Chst7), are required for the synthesis of CL40-positive glycans in the lung mesothelium. Furthermore, neither GlcNAc6ST2 nor GlcNAc6ST3 is sufficient for in vivo expression of the CL40 epitope in the lung mesothelium, as demonstrated by GlcNAc6ST1/3/4 triple-knock-out and GlcNAc6ST1/2/4 triple-knock-out mice. These results indicate that CL40-positive sialylated and sulfated glycans are abundant in the pleural mesothelium and are synthesized complementarily by GlcNAc6ST2 and GlcNAc6ST3, under physiological conditions in mice.
... TGF-β1 is one of the strongest liver fibrosis--promoting factors [31,32]. During the development of fibrosis, TGF-β1 stimulates the positive feedback mechanism of TGF-β1 secreted by HSCs through an autocrine mechanism [33]. ...
Article
Introduction: Liver fibrosis is caused by continuous wound healing responses to various harmful stimuli, including viral infection, drugs, alcohol, and autoimmune liver disease. The purpose of this study was to examine the effects of extracts of Periplaneta americana (EPA) in rats with pig serum-induced liver fibrosis to preliminarily assess the antifibrotic effect of EPA. Material and methods: Seventy rats were randomly divided into 7 groups (10 rats in each group): HC, the healthy control group; FC, the fibrotic control group; TL, low-dose EPA treatment group group; TM, medium-dose EPA group; TH, high-dose EPA treatment group; TC1, Panax notoginseng/Salvia mitiorrhiza treatment control group 1; TC2, colchicine treatment control group 2. TC1 and TC2 were used as the positive control to demonstrate the difference between EPA and the effects of other compounds. The liver fibrosis model was induced by intraperitoneal injection of 0.5 mL pig serum twice a week for 13 weeks in all groups except for the HC group. The hepatic fibrosis model was established at the 7th week, and followingly, the corresponding compounds were administered once a day in all groups for 6 weeks. Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) activity was determined in rat blood serum. We also measured liver fibrosis-related serum markers, including hyaluronic acid (HA), mucin layer (LN), type III pre-collagen (PC-III) and type IV collagen (IV-C). Hematoxylin and eosin (H&E) and Masson stainings were used to assess liver morphology and determine the stage of fibrosis. Immunohistochemistry was used to detect the protein expression of NF-κB, α-smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1) and tissue inhibitor of metalloproteinase-1 (TIMP-1) in rat liver tissue. Results: Compared with that of the HC group, the liver tissue of the FC group presented obvious liver damage and collagen deposition. The serum levels of ALT, AST, HA, LN, PC-Ⅲ and Ⅳ-C and the expression of NF-κB, α-SMA, TGF-β1 and TIMP-1 in the FC group were significantly higher than those in the HC group, the EPA treatment groups, the TC1 group and the TC2 group (P < 0.01). The levels of serum ALT, AST, HA, LN, PC-Ⅲ and Ⅳ-C and the expression of α-SMA, NF-κB, TGF-β1 and TIMP-1 in the TL, TC1 and TC2 groups were significantly higher than those TM and TH groups (P < 0.05). EPA treatment significantly improved liver function, decreased collagen deposition and reversed the pathological changes related to liver fibrosis. Conclusions: We found that EPA could reduce liver inflammation, suppress liver cell degeneration and necrosis, and reduce the formation of liver fibrous tissue. Its mechanism might be associated with inhibiting the expression of TGF-β1, TIMP-1, NF-κB and α-SMA to block signal transduction pathways in the hepatic fibrosis process. Therefore, EPA, as a traditional Chinese medicine, might be potentially used to prevent and treat hepatic fibrosis in the future. However, further more experiments are necessary to verify its effectiveness and possible signaling pathways.
Article
During the development of pleural fibrosis, pleural mesothelial cells (PMCs) undergo phenotypic switching from differentiated mesothelial cells to mesenchymal cells (MesoMT). Here, we investigated how external stimuli such as TGF-β induce HPMC-derived myofibroblast differentiation to facilitate the development of pleural fibrosis. TGF- significantly increased di-phosphorylation but not mono-phosphorylation of myosin II regulatory light chain (RLC) in HPMCs. An increase in RLC di-phosphorylation was also found at the pleural layer of our carbon black bleomycin (CBB) pleural fibrosis mouse model, where it showed filamentous localization that coincided with αSMA in the cells in the pleura. Among the protein kinases that can phosphorylate myosin II RLC, ZIPK (zipper-interacting kinase) protein expression was significantly augmented after TGF-β stimulation. Further, ZIPK gene silencing attenuated RLC di-phosphorylation, suggesting that ZIPK is responsible for di-phosphorylation of myosin II in HPMCs. While TGF-β significantly increased expression of ZIP kinase protein, change in ZIP kinase mRNA was marginal, suggesting a post transcriptional mechanism for the regulation of post-transcriptional regulation of ZIP kinase expression by TGF-β. ZIPK gene knock-down (KD) also significantly reduced TGF-β induced up-regulation of αSMA expression. This finding suggests that ZIPK KD attenuates MesoMT. ZIPK KD diminished TGF-β induced contractility of HPMCs consistent with a ZIPK KD induced decrease in the di-phosphorylation of myosin II RLC. The present results implicate ZIPK in the regulation of the contractility of HPMC derived myofibroblasts, phenotype switching and myofibroblast differentiation of HPMCs via MesoMT.
Article
Progressive lung scarring due to persistent pleural organization often results in pleural fibrosis (PF). This process affects patients with complicated parapneumonic pleural effusions, empyema, and other pleural diseases prone to loculation. In PF, pleural mesothelial cells undergo mesomesenchymal transition (MesoMT) to become profibrotic, characterized by increased expression of α-smooth muscle actin (α-SMA) and matrix proteins, including collagen (Col)-1. In our previous study, we showed that blocking PI3K/Akt signaling inhibits MesoMT induction in human pleural mesothelial cells (HPMCs). However, the downstream signaling pathways leading to MesoMT induction remain obscure. Here, we investigated the role of mammalian target of rapamycin (mTOR) complexes (mTORC1/2) in MesoMT induction. Our studies show that activation of the downstream mediator mTORC1/2 complex is likewise a critical component of MesoMT. Specific targeting of mTORC1/2 complex using pharmacological inhibitors, such as INK128 and AZD8055, significantly inhibited TGF-β-induced MesoMT markers in HPMCs. We further identified mTORC2/Rictor complex as the principal contributor to MesoMT progression induced by TGF-β. Knockdown of Rictor, but not Raptor, attenuated TGF-β induced MesoMT in these cells. In these studies, we further show that concomitant activation of the SGK1/NDRG1 signaling cascade is essential for inducing MesoMT. Targeting SGK1 and NDRG1 with siRNA and small molecular inhibitors attenuated TGF-β-induced MesoMT in HPMCs. Additionally, preclinical studies in our Streptococcus pneumoniae mediated mouse model of PF showed that inhibition of mTORC1/2 with INK128 significantly attenuated the progression of PF in sub-acute and chronic injury. In conclusion, our studies demonstrate that mTORC2/Rictor-mediated activation of SGK1/NDRG1 are critical for MesoMT induction, and targeting this pathway could inhibit or even reverse the progression of MesoMT and PF.
Article
Idiopathic pulmonary fibrosis (IPF) is a rare interstitial lung disease with a poor prognosis. Chronic micro-injuries, mainly caused by environmental factors to an aging alveolar epithelium, would lead to the aberrant differentiation and accumulation of aberrant mesenchymal cells with a contractile phenotype, known as fibrosis-associated myofibroblasts, which trigger abnormal extracellular matrix accumulation and fibrosis. The origin of those pathological myofibroblasts in pulmonary fibrosis is not fully understood to date. Lineage tracing methods using mouse models have opened new avenues for studying cell fate in a pathological context. This review aims to present a non-exhaustive list of different potential sources of those harmful myofibroblasts during lung fibrosis, based on these in vivo approaches, and considering the normal and fibrotic lung cellular atlas recently established by single-cell RNA sequencing.
Article
The local microenvironment shapes macrophage differentiation in each tissue. We hypothesized that in the peritoneum, local factors in addition to retinoic acid can support GATA6-driven differentiation and function of peritoneal large cavity macrophages (LCMs). We found that soluble proteins produced by mesothelial cells lining the peritoneal cavity maintained GATA6 expression in cultured LCMs. Analysis of global gene expression of isolated mesothelial cells highlighted mesothelin (Msln) and its binding partner mucin 16 (Muc16) as candidate secreted ligands that potentially regulate GATA6 expression in peritoneal LCMs. Mice deficient for either of these molecules showed diminished GATA6 expression in peritoneal and pleural LCMs that was most prominent in aged mice. The more robust phenotype in older mice suggested that monocyte-derived macrophages were the target of Msln and Muc16. Cell transfer and bone marrow chimera experiments supported this hypothesis. We found that lethally irradiated Msln-/- and Muc16-/- mice reconstituted with wild-type bone marrow had lower levels of GATA6 expression in peritoneal and pleural LCMs. Similarly, during the resolution of zymosan-induced inflammation, repopulated peritoneal LCMs lacking expression of Msln or Muc16 expressed diminished GATA6. These data support a role for mesothelial cell-produced Msln and Muc16 in local macrophage differentiation within large cavity spaces such as the peritoneum. The effect appears to be most prominent on monocyte-derived macrophages that enter into this location as the host ages and also in response to infection.
Article
Full-text available
Transforming growth factor (TGF)-beta is a cytokine that has been demonstrated to be an important modulator of inflammation and angiogenesis, as well as a potent stimulator of pleural fluid production and fibrosis. We previously demonstrated that rising levels of pleural fluid TGF-beta(1) correlate with pleural fibrosis in experimental empyema in rabbits. In this study, our hypothesis is that neutralization of TGF-beta with an intrapleural injection of a monoclonal antibody to TGF-beta will decrease pleural fibrosis in empyema. Prospective, randomized, blinded study. Animal research laboratory. Nineteen rabbits. An empyema was induced in 19 rabbits by intrapleural injection of Pasteurella multocida. A panspecific monoclonal antibody to TGF-beta was injected into the pleural space on 2 subsequent concurrent days in nine rabbits. Ten rabbits received intrapleural injections of bacteria alone and served as controls. All animals were then killed on day 6. Immunohistochemistry, using the antibody to TGF-beta, was performed on pleural tissue specimens from the control rabbits. Immunohistochemistry revealed localization of TGF-beta to macrophages in the exudative material and the visceral pleura. After injection of the antibody to TGF-beta, the amount of purulent, exudative material in the pleural space of the nine experimental animals was markedly decreased at autopsy on day 6, relative to control animals. All markers of empyema and pleural fibrosis were also significantly decreased in the rabbits receiving intrapleural anti-TGF-beta. TGF-beta localizes to macrophages in experimental empyema. Early intrapleural injection of an antibody to TGF-beta inhibits empyema formation and significantly decreases pleural fibrosis in experimental empyema.
Article
BACKGROUND Talc and tetracyclines induce pleurodesis by directly injuring the pleura. The injury results in intense inflammation which subsequently leads to fibrosis. Corticosteroids can inhibit talc pleurodesis by reducing the inflammatory process. We hypothesised that transforming growth factor β 2 (TGFβ 2 ), a fibrogenic cytokine with immunomodulatory functions, could induce effective pleurodesis without generating significant pleural inflammation and therefore remain effective despite co-administration of corticosteroids. METHODS Thirty rabbits were divided into two groups. Rabbits in the steroid group received weekly intramuscular injections of triamcinolone diacetate (0.8 mg/kg). Ten rabbits in each group were given 5.0 μg TGFβ 2 intrapleurally via a chest tube while the remaining five received 1.7 μg TGFβ 2 . Pleurodesis was graded macroscopically after 14 days from 1 (none) to 8 (>50% symphysis). RESULTS TGFβ 2 produced excellent pleurodesis at both 5.0 μg and 1.7 μg doses. The pleural effusions produced after the injection were low in all inflammatory markers. No significant differences were seen between the steroid group and controls in macroscopic pleurodesis scores (7.2 (1.3) v 7.1 (1.2)), levels of inflammatory markers in the pleural fluids (leucocyte 1107 (387)/mm ³ v 1376 (581)/mm ³ ; protein 3.1 (0.3) mg/dl v 2.9 (0.3) mg/dl, and LDH 478 (232) IU/l v 502 (123) IU/l), and the degree of microscopic pleural fibrosis and pleural inflammation. CONCLUSIONS TGFβ 2 can induce effective pleurodesis and remains effective in the presence of high dose parenteral corticosteroids.
Article
The hallmark of idiopathic pulmonary fibrosis (IPF) is the myofibroblast, the cellular origin of which in the lung is unknown. We hypothesized that alveolar epithelial cells (AECs) may serve as a source of myofibroblasts through epithelial-mesenchymal transition (EMT). Effects of chronic exposure to transforming growth factor (TGF)-beta 1 on the phenotype of isolated rat AECs in primary culture and a rat type H cell line (RLE-6TN) were evaluated. Additionally, tissue samples from patients with IPF were evaluated for cells co-expressing epithelial (thyroid transcription factor (TTF)-1 and pro-surfactant protein-B (proSP-B), and mesenchymal (a-smooth muscle actin (alpha-SMA)) markers. RLE-6TN cells exposed to TGF-beta 1 for 6 days demonstrated increased expression of mesenchymal cell markers and a fibroblast-like morphology, an effect augmented by tumor necrosis factor-a (TNF-alpha). Exposure of rat AECs to TGF-beta 1 (100 pmol/L) resulted in increased expression of a-SMA, type I collagen, vimentin, and desmin, with concurrent transition to a fibroblast-like morphology and decreased expression of TTF-1, aquaporin-5 (AQP5), zonula occludens-1 (ZO-1), and cytokeratins. Cells coexpressing epithelial markers and a-SMA were abundant in lung tissue from IPF patients. These results suggest that AECs undergo EMT when chronically exposed to TGF-beta 1, raising the possibility that epithelial cells may serve as a novel source of myofibroblasts in IPF.
Article
BACKGROUND We have recently shown that transforming growth factor (TGF)β2induces effective pleurodesis in rabbits. However, rabbits have a thin pleura while humans have a thick visceral pleura. The effect of intrapleural administration of TGFβ2 in animals with a thick pleura and its associated systemic effects have not been investigated. This study was undertaken (1) to develop a new animal model for the study of pleurodesis using sheep which have a thick pleura resembling that of humans; (2) to study the efficacy of TGFβ2 as a pleurodesis agent in the sheep model; and (3) to assess whether histological changes occur in extrapulmonary organs after intrapleural administration of TGFβ2.METHODS Twelve sheep were divided into four groups and were given a single intrapleural injection of TGFβ2 in a concentration of 1.0 μg/kg, 0.5 μg/kg, 0.25 μg/kg or 0.125 μg/kg to the right pleural cavity via a chest tube. The left pleural cavity served as the control. Any pleural fluid that accumulated after the intrapleural TGFβ2 injection was collected and analysed. The degree of pleurodesis was graded from 1 (no adhesions) to 8 (complete symphysis >50% of chest wall) at day 14 when the sheep were killed. Biopsy specimens were taken from the lungs and extrapulmonary organs.RESULTSAll sheep that received ⩾0.25 μg/kg TGFβ2 developed excellent pleurodesis (score = 8) while those that received 0.125 μg/kg had a median score of 6. The pleurodesis score did not exceed 2 in the control (left) side of any sheep. Sheep receiving ⩾0.50 μg/kg TGFβ2 developed large exudative pleural effusions while those receiving a lower dose did not. The production of effusions neither hindered nor was necessary for inducing pleurodesis. There were no significant fibrotic changes in any of the extrapulmonary organs.CONCLUSION Intrapleural injection of 0.25–1.0 μg/kg TGFβ2 produces excellent pleurodesis in a new sheep model with no evidence of extrapulmonary fibrosis.
Article
In human tissues, normal homeostasis requires intricately balanced interactions between cells and the network of secreted proteins known as the extracellular matrix. These cooperative interactions involve numerous cytokines acting through specific cell-surface receptors. When the balance between the cells and the extracellular matrix is perturbed, disease can result. This is clearly evident in the interactions mediated by the cytokine transforming growth factor β (TGF-β). TGF-β is a member of a family of dimeric polypeptide growth factors that includes bone morphogenic proteins and activins. All of these growth factors share a cluster of conserved cysteine residues that form a common cysteine . . .
Article
Background: We have recently demonstrated that transforming growth factor beta-2 (TGF-β2) can produce effective pleurodesis. Whether this effect can be reproduced by the use of its downstream proteins is not known. This study compared the effectiveness of TGF-β2 and fibronectin in inducing pleurodesis in rabbits. Methodology: New Zealand white rabbits (1.5–2.0 kg) were given 1.7 μg of TGF-β2 (n = 5) or 2.0 mg of cellular fibronectin (n = 4) intrapleurally via a chest tube. The induced pleural fluid was collected and analyzed. The rabbits were sacrificed after 14 days. The pleurodesis was graded macroscopically from 1 (none) to 8 (symphysis > 50%). Results: All rabbits in the TGF-β2 group developed effective pleurodesis while none in the fibronectin group had scores > 2 (pleurodesis scores 7.0 ± 0.6 vs 1.3 ± 0.3, P < 0.001). Rabbits that received TGF-β2 produced large amounts of pleural fluid initially (< 4 days). Microscopically, the pleura of rabbits in the TGF-β2 group showed prominent spindle cell proliferation and collagen deposition, but no significant inflammation or mesothelial proliferation. Pleural tissues of rabbits in the fibronectin group had occasional thin collagen deposits only. The intrapleural administration of 2.0 mg of fibronectin, a downstream product of TGF-β, did not induce effective pleurodesis, as did the intrapleural administration of TGF-β2. Conclusions: The present study suggests that the mechanism by which TGF-β2 induces pleurodesis is not predominantly dependent on the production of fibronectin.
Article
Three-dimensional human head modeling is useful in video-conferencing or other virtual reality applications. However, manual construction of 3D models using CAD tools is often expensive and time-consuming. Here we present a robust and efficient method for the construction of a 3D human head model from perspective images viewing from different angles. In our system, a generic head model is first used, then three images of the head are required to adjust the deformable contours on the generic model to make it closer to the target head. Our contributions are as follows. Our system uses perspective images that are more realistic than orthographic projection approximations used in earlier works. Also, for shaping and positioning face organs, we present a method for estimating the camera focal length and the 3D coordinates of facial landmarks when the camera transformation is known. We also provide an alternative for the 3D coordinates estimation using epipolar geometry when the extrinsic parameters are absent. Our experiments demonstrate that our approach produces good and realistic results.