Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2008 Feb 4.
Published in final edited form as: Semin Respir Crit Care Med. 2006 Dec;27(6):600–612. doi: 10.1055/s-2006-957332

Epithelial–Mesenchymal Interactions in Pulmonary Fibrosis

Jeffrey C Horowitz 1, Victor J Thannickal 1
PMCID: PMC2225581  NIHMSID: NIHMS38182  PMID: 17195137

Abstract

Pulmonary fibrosis represents the sequelae of a variety of acute and chronic lung injuries of known and unknown etiologies. Tissue specimens obtained from patients with pulmonary fibrosis, regardless of the etiology, consistently show evidence of an ongoing wound-repair response. Epithelial–mesenchymal interactions have critical roles in normal lung development, tissue repair processes, and fibrosis. Current hypotheses propose that dysregulated function of, and impaired communication between, epithelial and mesenchymal cells prevent resolution of the wound-repair response and contribute to the pathobiology of pulmonary fibrosis. This hypothesis is supported by abundant evidence from patients, animal models, and cell-culture studies demonstrating abnormalities in epithelial cell and mesenchymal cell activities including proliferation, differentiation, and survival. This article reviews the aberrant epithelial and mesenchymal cellular phenotypes found in the context of pulmonary fibrosis and discusses the mechanisms that perpetuate these cellular phenotypes.

Keywords: Fibroblast, myofibroblast, apoptosis, wound repair, extracellular matrix

EPITHELIAL–MESENCHYMAL INTERACTIONS IN LUNG DEVELOPMENT, WOUND REPAIR, AND FIBROSIS

Epithelial and mesenchymal cell interactions have a critical role in lung development, and the precise temporal and spatial regulation of epithelial and mesenchymal cell functions is necessary for normal alveolarization.1 The importance of these interactions is exemplified by studies demonstrating that normal alveolarization requires lung epithelial cell secretion of soluble growth factors such as platelet derived growth factor A (PDGF-A), which provide survival signals to myofibroblasts during alveolar septation.2 Mice deficient in PDGF-A develop emphysema associated with excessive loss of myofibroblasts. During later stages of lung development, however, apoptosis of myofibroblasts is essential for alveolar wall thinning and remodeling to achieve the delicate alveolar structure seen in normal lung.3,4 Thus aberrant “cross-talk” between epithelial and mesenchymal cells can lead to profound abnormalities in lung development.

The repair response to injury in adult lung recapitulates several aspects of embryonic lung development.5,6 As in normal lung development, physiological wound-repair requires spatially and temporally regulated epithelial and mesenchymal cell responses to reestablish an intact epithelium. Epithelial cell proliferation, migration, and differentiation must be coordinated with mesenchymal cell recruitment, proliferation, differentiation with extracellular matrix (ECM) remodeling, and subsequent apoptosis of myofibroblasts.79 Dysregulation of this orchestrated wound-repair response can result in pathological scar formation due to myofibroblast accumulation and ECM remodeling.10

Pulmonary fibrosis can result from a variety of acute and chronic causes of lung injury.11 At the cellular level, pulmonary fibrosis—regardless of the etiology—represents an ongoing repair response to some form of persistent or recurrent injury, or perhaps a dysregulated or inappropriate repair response to a past stimulus of lung injury.11,12 This article reviews the aberrant epithelial and mesenchymal cellular phenotypes found in the context of pulmonary fibrosis, with a focus on idiopathic pulmonary fibrosis (IPF), and discusses the mechanisms that perpetuate these cellular phenotypes and contribute to disease pathobiology.

EPITHELIAL–MESENCHYMAL INTERACTIONS IN PULMONARY FIBROSIS

Current hypotheses propose that progressive lung fibrosis results from dysregulated function of, and/or communication between, epithelial and mesenchymal cells leading to a “vicious cycle” of epithelial cell injury and mesenchymal cell responses.11,1315 Myofibroblasts, key effector cells in fibrogenesis, aggregate within fibroblastic foci of usual interstitial pneumonia (UIP), the histopathologic correlate of IPF.16 These foci are thought to represent the organization of focal areas of acute lung injury, and the presence/extent of fibroblastic foci correlates with poor outcomes in IPF.1719 Overlying the fibroblastic foci are areas of damaged basement membrane and denuded epithelium.20,21 Additionally, alveolar epithelial cells (AECs) that are in close association with myofibroblasts have phenotypic changes that suggest the activation of a stereotypic wound-repair response.22 Supporting this concept, studies of IPF tissue have reported evidence of AEC proliferation and regenerative hyperplasia,21,23 bronchiolar and squamous metaplasia,2426 and apoptosis.2729

Our understanding of the direct interactions between epithelial and mesenchymal cells in the context of pulmonary fibrosis is quite limited. An ultrastructural examination of specimens from patients with pulmonary fibrosis identified epithelial cell extensions through “apertures” in the underlying basement membrane.20 Further studies suggested that, during lung repair responses, dynamic interactions between epithelial and mesenchymal cells allow for reciprocal regulation of proliferation and differentiation.30,31 A recent report demonstrated that the fibroblast “link” between the alveolar epithelium and endothelium is disrupted in emphysema specimens, providing further support for the important role of epithelial–mesenchymal interactions in a broader context of lung disease.32 The mechanism(s) underlying dysregulated epithelial and mesenchymal cell phenotypes in pulmonary fibrosis remain poorly understood. Evidence suggests that these complex cellular interactions are mediated by soluble factors acting through autocrine and paracrine mechanisms. Additionally, the ECM itself likely contributes both directly and indirectly to the aberrant cellular phenotypes seen in pulmonary fibrosis (Figure 1).

Figure 1.

Figure 1

Schematic representation of epithelial and mesenchymal cell phenotypes and their interactions in the context of pulmonary fibrosis. Epithelial cells overlying damaged basement membrane and fibroblastic foci demonstrate phenotypes suggestive of a dysregulated wound-repair response, including excessive apoptosis, dysregulated proliferation, and impaired migration and regeneration. In contrast, underlying mesenchymal cells demonstrate myofibroblast differentiation, resistance to apoptosis, proliferation and secretion of soluble factors, extracellular matrix (ECM) proteins, and oxidants. Both epithelial and mesenchymal cells secrete soluble mediators that function through paracrine and autocrine mechanisms to regulate cell phenotypes. Additionally, the ECM provides a substrate for epithelial–mesenchymal interactions and may also directly regulate epithelial and mesenchymal cell phenotypes.

EPITHELIAL CELL PHENOTYPES IN PULMONARY FIBROSIS

Regeneration of a damaged epithelium following injury is required for the reestablishment of normal tissue architecture and function. Failure to establish an intact epithelium may induce a persistent wound-repair response. In the context of pulmonary fibrosis, evidence from human disease, animal models, and cell-culture studies has identified abnormalities in epithelial cell survival/apoptosis, proliferation, and migration, which likely contribute to disease pathobiology.

Increased Epithelial Cell Apoptosis

Excessive AEC apoptosis is likely to play a key role in the pathobiology of IPF. Studies of IPF tissue have shown type II AEC apoptosis at the ultrastructural level and demonstrated that AEC apoptosis is increased in cells adjacent to fibroblastic foci.28,33 Moreover, Kuwano et al reported increased expression of the proapoptotic p53 and p21 tumor suppressor proteins associated with alveolar and bronchial epithelial cell apoptosis in a majority of IPF specimens but found no such apoptosis in emphysema specimens or normal lung tissue.27 In another study, p53 nuclear accumulation was identified in epithelial cells from IPF, but not other idiopathic interstitial pneumonia specimens.26 A recent study reported similar findings of excessive AEC apoptosis associated with increased expression of proapoptotic proteins (caspase-3 and Bax) and decreased expression of the anti-apoptotic Bcl-2 protein in IPF specimens.34

Further supporting the role of AEC apoptosis in the pathogenesis of pulmonary fibrosis are animal models showing that inhibition of AEC apoptosis attenuates pulmonary fibrosis, whereas the induction of AEC apoptosis promotes fibrotic responses. Studies have reported that inhibiting caspases, which are key components of intracellular apoptotic signaling cascades, lead to decreased fibrotic responses to intratracheal bleomycin.35,36 Moreover, several studies suggest that induction of AEC apoptosis may be sufficient for the development of pulmonary fibrosis. In one study, pulmonary fibrosis followed epithelial cell apoptosis induced by transient overexpression of the profibrotic mediator, transforming growth factor-β1 (TGF-β1).37

The Fas-Fas ligand pathway, a proapoptotic signaling cascade that is upregulated in patients with IPF, has also been investigated in the pathogenesis of pulmonary fibrosis.29,38 Inhaled Fas-activating antibodies induce pulmonary fibrosis and AEC apoptosis in mice.39,40 Additionally, Fas-mediated epithelial cell apoptosis is potentiated by TGF-β1.40 Studies in the bleomycin model of pulmonary fibrosis, however, have reported conflicting results. One study reported decreased fibrosis in transgenic mice deficient in Fas or Fas-ligand, whereas another study, using similar transgenic mice on a different background strain, found that deficiencies in the Fas-Fas ligand system did not reduce bleomycin-induced pulmonary fibrosis.41,42 A third study recently reported that bleomycin-induced pulmonary fibrosis was dependent on reactive oxygen species (ROS)-mediated epithelial cell apoptosis that is independent of the Fas pathway.43 Collectively, these animal models support a role for AEC apoptosis in the pathogenesis of pulmonary fibrosis but fail to define a specific mechanism for AEC apoptosis. Strain-dependent differences in the effects of bleomycin on lung epithelial cells and susceptibility to pulmonary fibrosis are well described in murine models.44,45 Thus it is likely that variable and overlapping mechanisms of AEC apoptosis may be induced in different contexts, depending on the stimulus for lung injury and genetic factors of the host.

Impaired Epithelial Cell Regeneration and Migration

Impaired epithelial cell regeneration and migration following injury may contribute to the development of pulmonary fibrosis.14 Several studies have reported abnormalities in epithelial cell proliferation in patients with IPF, although mechanisms remain unclear.18,22,26,46 In animal models, epithelial cell mitogens and motogens, such as hepatocyte growth factor (HGF) and keratinocyte growth factor (KGF) have been shown to attenuate pulmonary fibrosis.47,48 Similarly, granulocyte-monocyte colony stimulating factor (GM-CSF) induces epithelial cell migration, and mice deficient in GM-CSF develop increased fibrosis following lung injury.4951 Paracrine interactions between epithelial cells and fibroblasts may also contribute to impaired epithelial regeneration because TGF-β1 can block the mitogenic effects of KGF on AECs.52

The plasminogen activation system has been shown to have an important role in the pathogenesis of bleomycin-induced murine pulmonary fibrosis. Activation of plasminogen to plasmin is mediated by urokinase or tissue plasminogen activators (uPA or tPA), both of which are inhibited by PAI-1 (plasminogen activator-inhibitor 1). Plasmin, in turn, is a serine protease that participates in fibrinolysis, extracellular matrix regulation, angiogenesis, cell signaling, and cell migration.53 Activation of the plasminogen system protects mice from bleomycin-induced pulmonary fibrosis, whereas inhibition of this system promotes fibrosis.5456 Additional studies have established that these antifibrotic effects are not a function of fibrinolysis, suggesting that alternative mechanisms mediate antifibrotic effects of plasminogen activation.57,58

The plasminogen-activation system has been shown to modulate epithelial cell migration and wound closure.5962 For example, plasminogen activator inhibitor (PAI)-1 deficient mice exhibit accelerated wound healing60 and the presence of PAI-1 inhibits epithelial cell migration.61 Consistent with these findings, uPA and plasmin promote epithelial cell migration.59 Thus it is plausible that the antifibrotic effects of the plasminogen activation system are related, at least in part, to facilitation of epithelial cell migration.

MESENCHYMAL CELL PHENOTYPES

Fibroblasts are the most versatile of the mesenchymal cells and participate in the stereotypic repair and regenerative processes in virtually every human organ and tissue.7,8,10 Myofibroblasts are differentiated, contractile fibroblasts with characteristics intermediate between fibroblasts and smooth muscle cells.10,6365 In response to injury, fibroblasts infiltrate the wound, differentiate into myofibroblasts, and secrete ECM proteins that form a provisional matrix, which serves as a “scaffold” for tissue repair.64 Myofibroblast contraction of the provisional matrix facilitates repair by bringing healthy epithelial cells at the wound margin into close approximation. Collagen secretion stabilizes the contracted matrix surrounding myofibroblasts and provides tensile strength to granulation tissue.10 Recent studies have shown that the mesenchymal cells in the lung following injury may be derived from different sources, including the interstitial fibroblasts,63 circulating fibrocytes,6669 bone marrow–derived mesenchymal stem cells,70 and epithelial cells.7173 It is not known if specific mesenchymal cell phenotypes are determined by the site of origin, the tissue microenvironment, or both. Interestingly, mesenchymal cells derived from different sites may contribute to different aspects of wound repair.70 Moreover, mesenchymal cells isolated from different tissue microenvironments manifest phenotypic heterogeneity and “positional memory,” which may be maintained for extended periods in ex vivo cell culture.74,75

Mesenchymal Cell Survival and Apoptosis

Myofibroblast apoptosis is a normal event during the resolution of wound-repair responses.76 Failure of apoptosis leads to myofibroblast accumulation, persistent ECM contraction, secretion, and remodeling that results in pathological scar formation.10,77 Consistently, several studies have reported that both normal and fibrotic lung fibroblasts are resistant to apoptotic stimuli through several mechanisms.7884 Unlike epithelial cells, Fas activation is not sufficient to induce fibroblast apoptosis, although susceptibility to Fas-mediated apoptosis is increased in the presence of inflammatory cytokines.78,79,81,82,85,86 Additionally, the profibrotic mediator, TGF-β1, confers fibroblast resistance apoptosis induced by serum-deprivation and interleukin-1β.87,88 Contextual signals transmitted from the ECM may be important in the regulation of myofibroblast survival/apoptosis. A recent study, for example, showed that IPF-derived fibroblasts, compared with normal lung fibroblasts, had increased resistance to apoptosis imparted by expression the collagen binding receptor tyrosine kinase, discoidin domain receptor 1 (DDR1).84 In contractile collagen gels, however, myofibroblasts undergo apoptosis associated with biomechanical unloading.89,90 This stimulus for apoptosis is overcome by activation of antiapoptotic signaling pathways involving focal adhesion kinase (FAK) and protein kinase B (PKB/Akt), both of which are activated by TGF-β1 in fibroblasts.83,87,91,92 Paradoxically, a recent study reported that TGF-β1 may increase fibroblast apoptosis in a contractile collagen gel model.93 Thus apoptotic susceptibility of myofibroblasts may be dependent on integrated soluble and matrix-associated signals within the cellular microenvironment.

Few studies have assessed the role of myofibroblast survival in the pathogenesis of pulmonary fibrosis in vivo. Pharmacological inhibition of prosurvival signaling proteins, focal adhesion kinase (FAK), and protein kinase B (PKB/Akt), activated in lung myofibroblasts, attenuates the fibrogenic response to bleomycin-induced lung injury.94 Inhibition of p38 mitogen activated protein kinase (MAPK), which mediates TGF-β1-induced Akt activation and proliferative responses in myofibroblasts, similarly reduces bleomycin-induced pulmonary fibrosis.87,95,96 Studies in humans support deficient mesenchymal cell apoptosis in fibrotic disease states. One study showed decreased apoptosis of myofibroblasts in UIP/IPF tissue in comparison with cryptogenic organizing pneumonia, an idiopathic interstitial pneumonia (IIP) that is more responsive to therapy and has better outcomes than UIP/IPF.97 Furthermore, alveolar mesenchymal cells isolated from patients with nonresolving, or “fibroproliferative” acute respiratory distress syndrome (ARDS) are more resistant to apoptosis when compared with cells derived from patients with “resolving” ARDS.83 Together, these studies suggest that mesenchymal cell resistance to apoptosis may be associated with persistent and progressive fibrotic diseases that respond poorly to current interventions, and that promoting myofibroblast apoptosis may be an effective strategy to halt the progressive nature of these forms of fibrotic lung disease.

Myofibroblast Proliferation

The accumulation of fibroblasts and myofibroblasts may be due to decreased apoptosis, increased proliferation, or both. Investigations of fibroblast proliferation in IPF have produced conflicting results. An early study showed increased proliferation in fibroblasts from pulmonary fibrosis compared with normal controls.98 Among the fibrosis-derived clones, however, there was significant heterogeneity in proliferative rates, suggesting that a subpopulation of highly proliferative cells may account for the fibroblast accumulation.98 In another study, fibroblasts from specimens with “early fibrosis” had increased rates of proliferation, whereas fibroblasts from specimens with “dense” fibrosis had decreased proliferation compared with normal fibroblasts.99 Two other studies failed to find increased proliferation in IPF fibroblasts.100,101 Fibroblast and myofibroblast proliferation is likely influenced by soluble fibroblast mitogens within the alveolar microenvironment.95,102 IPF fibroblasts may possess enhanced, and sometimes divergent, mitogenic responses to certain soluble mediators103 while being resistant to other antiproliferative signals.100 Thus the variability in reported proliferative phenotypes of IPF fibroblasts/myofibroblasts may be explained, in part, by their derivation from heterogeneous fibrotic lesions of varying “maturity” and the presence of specific soluble factors within the cellular microenvironment.

SOLUBLE FACTORS MEDIATE EPITHELIAL AND MESENCHYMAL CELL PHENOTYPES

Transforming Growth Factor-β1

TGF-β1 is a dimeric polypeptide that is implicated in the pathogenesis of fibrotic disease in multiple organs and tissues, including IPF.104 Moreover, polymorphisms in the gene encoding TGF-β1 are associated with accelerated progression of IPF.105 Bronchoalveolar lavage (BAL) fluid from IPF patients contain increased concentrations of TGF-β1 compared with normal controls or patients with hypersensitivity pneumonitis (HP).40,106 Furthermore, BAL fluid from IPF patients, but not normal controls or HP patients, induces epithelial cell apoptosis, an effect that is blocked by the presence of TGF-β1 blocking antibodies.40 In animal models, lung-specific overexpression of TGF-β1 induces pulmonary fibrosis, whereas blockade of TGF-β1 or its downstream signaling pathways attenuates bleomycin-induced pulmonary fibrosis.107109 The cellular sources of TGF-β1 in the context of IPF have not been conclusively demonstrated, and there is evidence to suggesting that both myofibroblasts and damaged/aberrant epithelial cells may contribute.110117

The biological activities of TGF-β1 vary depending on cell type and context. As a general paradigm, TGF-β1 mediates divergent effects on epithelial and mesenchymal cell phenotypes. As a tumor suppressor, TGF-β1 induces cell-cycle arrest and promotes apoptosis in epithelial cells.40,118 TGF-β1 also antagonizes the mitogenic effects of KGF and enhances Fas-mediated apoptosis in lung epithelial cells.40,52,119,120 In contrast, TGF-β1 activates fibroblasts by inducing myofibroblast differentiation,92 survival,87,88 and proliferation95,101,102; moreover, TGF-β1 induces ECM production and remodeling101,121,122 and generation of extracellular ROS.123

Hepatocyte Growth Factor

HGF is an epithelial cell mitogen and motogen124,125 that is secreted by myofibroblasts and opposes certain TGF-β1 actions.126 HGF has antifibrotic activity in animal models of renal, hepatic, pulmonary, and myocardial fibrosis.48,127129 IPF fibroblasts secrete decreased levels of HGF compared with normal lung fibroblasts,130 and TGF-β1 decreases HGF expression by fibroblasts.131 The mechanisms by which HGF exerts its antifibrotic effects are likely related to its divergent effects on epithelial and mesenchymal cell phenotypes. HGF promotes proliferation and migration of epithelial cells, facilitating reepithelialization.61 In contrast, HGF induces apoptosis of myofibroblasts and mediates antifibrotic effects in vivo.128

Studies show that the antifibrotic effects of other mediators may be mediated through enhanced activation or responsiveness of cells to HGF. For example, the protective effects of plasminogen activation in animal models are mediated, at least in part, by HGF.132 Additionally, prostaglandin E2 (PGE2), an arachidonic acid metabolite that inhibits several aspects of myofibroblast function, increases HGF secretion by IPF-derived myofibroblasts to normal levels.130 Interferon gamma (IFN-γ), a cytokine with potential antifibrotic properties,133 upregulates HGF receptor expression on AECs.134

Keratinocyte Growth Factor

KGF is another fibroblast-secreted epithelial cell mitogen/motogen that attenuates pulmonary fibrosis in animal models.47,135137 KGF confers resistance to Fas-mediated apoptosis in lung epithelial cells.138 Furthermore, KGF induces the secretion of PGE2 by fibroblasts in coculture with AECs.139 The proliferative effects of KGF on AECs, however, are antagonized by TGF-β1.52 Moreover, a recent study shows that IPF-fibroblasts secrete decreased amounts of KGF following stimulation with IL-1β than normal fibroblasts.140 Collectively, these studies suggest that impaired regulation of KGF, through decreased fibroblast secretion, decreased epithelial cell responsiveness, or a disruption in homeostatic epithelial–mesenchymal interactions may contribute to the pathogenesis of pulmonary fibrosis.

Angiotensin II

Angiotensin II (AII) is a fibroblast-derived soluble mediator implicated in the pathogenesis of pulmonary fibrosis. In animal models, pharmacologic inhibition of angiotensin converting enzyme (ACE) and inhibition of the type 1 angiotensin receptor (by pharmacological and genetic approaches) have been shown to abrogate bleomycin-induced pulmonary fibrosis.35,141,142 IPF-derived fibroblasts secrete AII, and AII induces epithelial cell apoptosis.33 Moreover, Fas-mediated epithelial cell apoptosis requires both AII and the angiotensin receptor.143,144 Inhibition of ACE blocks Fas-mediated epithelial cell apoptosis.145 In fibroblasts, however, AII promotes proliferation.102,146 These autocrine effects may be indirect, however, because AII can induce the synthesis of TGF-β1 in several cell systems.147,148

Reactive Oxygen Species

Oxidant-mediated injury to epithelial cells has been implicated in the pathobiology of IPF.149 Epithelial lining fluid from IPF patients contains increased levels of myeloperoxidase, and inflammatory cells from IPF patients generate increased levels of ROS.149 Levels of the antioxidant, glutathione (GSH), are decreased in the epithelial lining fluid of IPF patients, and this increases following treatment with oral N-acetylcysteine (NAC).150152 Recent clinical trials have supported a role for antioxidant therapy as an adjunct to standard therapy in IPF; the effects of NAC monotherapy have yet to be determined.153,154

Although generally thought of in the context of leukocyte biology and inflammation, reactive oxygen species are also important mediators of intra- and intercellular signaling.155 In IPF, extracellular oxidants are generated not only by inflammatory cells but by myofibroblasts as well.123,149,156 Recent studies demonstrate that paracrine secretion of hydrogen peroxide (H2O2) by IPF myofibroblasts induces epithelial cell death in ex vivo coculture, demonstrating another potential mechanism of epithelial cell injury/apoptosis in IPF.157

Arachidonic Acid Metabolites

The balance between arachidonic acid metabolites, pros-taglandins and leukotrienes has been shown to have a role in the pathobiology of pulmonary fibrosis.158 IPF patients have an imbalance in these mediators characterized by decreased PGE2 in BAL fluid,159 decreased epithelial expression of cyclooxygenase-1 and -2 (COX-1 and COX-2),160 and increased levels of leukotrienes in lung tissue.161 In animal models, COX-2 deficient mice develop increased fibrosis following intratracheal bleomycin, whereas leukotriene-deficient mice are protected from fibrosis.162,163

PGE2 suppresses several fibroblast activities, including proliferation,164166 chemotaxis and migration,167,168 differentiation,169 and collagen production.170 PGE2 is secreted by lung epithelial cells and lung fibroblasts, suggesting that homeostatic suppression of myofibroblast function may be mediated through both autocrine and paracrine mechanisms.165,168171 Moreover, recent studies show that KGF can enhance alveolar epithelial cell secretion of PGE2.139 Thus the decreased expression of COX in IPF epithelial cells suggests that a decreased capacity for PGE2 synthesis/secretion may contribute to pulmonary fibrosis through impaired suppression of myofibroblast activation.

Endothelin-1

Secreted by alveolar epithelial cells, vascular endothelial cells, and macrophages,172 endothelin-1 promotes fibroblast proliferation, contraction, differentiation, and ECM production.173176 AECs from IPF specimens express high levels of endothelin-1.177,178 In animal models, overexpression of endothelin-1 leads to progressive pulmonary fibrosis,179 and an endothelin-1 receptor antagonist attenuates bleomycin-induced pulmonary fibrosis.180

THE EXTRACELLULAR MATRIX MEDIATES EPITHELIAL–MESENCHYMAL INTERACTIONS AND AFFECTS CELLULAR PHENOTYPES

The ECM provides a dynamic and biologically active substrate for epithelial–mesenchymal interactions; the composition of the ECM regulates cellular phenotypes in the context of lung injury and repair.181,182 As an example, adhesion to an intact basement membrane (epithelial cells) or the ECM (mesenchymal cells) provides survival signals for these cells; the loss of adhesion-mediated signaling leads to a form of apoptosis termed anoikis.183,184 Moreover, the ECM itself may promote or perpetuate aberrant cellular phenotypes. Adhesion-mediated signaling pathways activated by cell–matrix interactions are required for myofibroblast differentiation induced by TGF-β1 and promote myofibroblast survival.91,92 Additionally, extra domain-A (ED-A) containing fibronectin has been shown to be necessary for myofibroblast differentiation, and myofibroblasts in fibroblastic foci of UIP/IPF colocalize with ED-A containing fibronectin.16,185 Finally, the biomechanical properties of the ECM are also likely to regulate fibroblast/myofibroblast phenotypes.10,186188

The composition and biomechanics of the ECM, in turn, are affected by mesenchymal and epithelial cells. Myofibroblast and epithelial cell–derived mediators interact with proteolytic pathways to regulate ECM remodeling.189192 Additionally, myofibroblast secretion of ROS induces posttranslational modifications in ECM proteins.193 The dynamic composition and interactions between ECM components may lead to further changes in biomechanics and adhesion-mediated signaling, thereby promoting and perpetuating epithelial and mesenchymal cell phenotypes.

Epithelial–Mesenchymal Transition

Epithelial–mesenchymal transition (EMT) is a well-conserved cellular process by which epithelial cells acquire the morphological and molecular characteristics of mesenchymal cells; this process plays an important role in embryonic morphogenesis and cancer metastasis.194,195 Recent studies report that alveolar epithelial cells are capable of TGF-β1-mediated EMT in vitro,72,73,196 and one report found that epithelial and mesenchymal markers colocalize in UIP/IPF tissue.71 Although there is evidence to suggest that EMT contributes to the development of renal fibrosis in murine models, the role of EMT in lung fibrosis remains undefined and requires further study.194,197

CONCLUSION

The regulation of epithelial–mesenchymal interactions is a key control point in normal and pathological wound-repair responses. In the context of tissue injury, coordinated and regulated interactions between epithelial cells, mesenchymal cells, and the ECM are required for the reestablishment of normal tissue architecture and function. In fibrotic disease, this delicate balance becomes dysregulated, as evidenced by inappropriate loss of epithelial architecture, persistent myofibroblast activation, and accumulation within fibroblastic foci. The causes of this dysregulation remain undefined but are likely due to a combination of genetic and environmental factors within the context of persistent or recurrent lung injury leading to aberrant autocrine and paracrine signaling between epithelial and mesenchymal cells.

Acknowledgments

This work was supported, in part, by National Institutes of Health grants K08 HL081059 (to J.C.H.); R01 HL67967 and P50 HL74024 (to V.J.T).

References

  • 1.Hogan BL, Yingling JM. Epithelial/mesenchymal interactions and branching morphogenesis of the lung. Curr Opin Genet Dev. 1998;8:481–486. doi: 10.1016/s0959-437x(98)80121-4. [DOI] [PubMed] [Google Scholar]
  • 2.Bostrom H, Willetts K, Pekny M, et al. PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell. 1996;85:863–873. doi: 10.1016/s0092-8674(00)81270-2. [DOI] [PubMed] [Google Scholar]
  • 3.Bruce MC, Honaker CE, Cross RJ. Lung fibroblasts undergo apoptosis following alveolarization. Am J Respir Cell Mol Biol. 1999;20:228–236. doi: 10.1165/ajrcmb.20.2.3150. [DOI] [PubMed] [Google Scholar]
  • 4.Schittny JC, Djonov V, Fine A, Burri PH. Programmed cell death contributes to postnatal lung development. Am J Respir Cell Mol Biol. 1998;18:786–793. doi: 10.1165/ajrcmb.18.6.3031. [DOI] [PubMed] [Google Scholar]
  • 5.Warburton D, Tefft D, Mailleux A, et al. Do lung remodeling, repair, and regeneration recapitulate respiratory ontogeny? Am J Respir Crit Care Med. 2001;164(10 Pt 2):S59–S62. doi: 10.1164/ajrccm.164.supplement_2.2106064. [DOI] [PubMed] [Google Scholar]
  • 6.Demayo F, Minoo P, Plopper CG, Schuger L, Shannon J, Torday JS. Mesenchymal–epithelial interactions in lung development and repair: are modeling and remodeling the same process? Am J Physiol Lung Cell Mol Physiol. 2002;283:L510–L517. doi: 10.1152/ajplung.00144.2002. [DOI] [PubMed] [Google Scholar]
  • 7.Singer AJ, Clark RA. Cutaneous wound healing. N Engl J Med. 1999;341:738–746. doi: 10.1056/NEJM199909023411006. [DOI] [PubMed] [Google Scholar]
  • 8.Martin P. Wound healing: aiming for perfect skin regeneration. Science. 1997;276:75–81. doi: 10.1126/science.276.5309.75. [DOI] [PubMed] [Google Scholar]
  • 9.Desmouliere A, Chaponnier C, Gabbiani G. Tissue repair, contraction, and the myofibroblast. Wound Repair Regen. 2005;13:7–12. doi: 10.1111/j.1067-1927.2005.130102.x. [DOI] [PubMed] [Google Scholar]
  • 10.Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002;3:349–363. doi: 10.1038/nrm809. [DOI] [PubMed] [Google Scholar]
  • 11.Thannickal VJ, Toews GB, White ES, Lynch JP, III, Martinez FJ. Mechanisms of pulmonary fibrosis. Annu Rev Med. 2004;55:395–417. doi: 10.1146/annurev.med.55.091902.103810. [DOI] [PubMed] [Google Scholar]
  • 12.Rennard SI, Bitterman PB, Crystal RG. Response of the lower respiratory tract to injury. Mechanisms of repair of the parenchymal cells of the alveolar wall. Chest. 1983;84:735–739. doi: 10.1378/chest.84.6.735. [DOI] [PubMed] [Google Scholar]
  • 13.Selman M, Pardo A. Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-talk disorder. Respir Res. 2002;3:3. doi: 10.1186/rr175. www.respiratory-research.com. [DOI] [PMC free article] [PubMed]
  • 14.Selman M, King TE, Pardo A. Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med. 2001;134:136–151. doi: 10.7326/0003-4819-134-2-200101160-00015. [DOI] [PubMed] [Google Scholar]
  • 15.Gross TJ, Hunninghake GW. Idiopathic pulmonary fibrosis. N Engl J Med. 2001;345:517–525. doi: 10.1056/NEJMra003200. [DOI] [PubMed] [Google Scholar]
  • 16.Kuhn C, McDonald JA. The roles of the myofibroblast in idiopathic pulmonary fibrosis: ultrastructural and immunohistochemical features of sites of active extracellular matrix synthesis. Am J Pathol. 1991;138:1257–1265. [PMC free article] [PubMed] [Google Scholar]
  • 17.Katzenstein AL, Myers JL. Idiopathic pulmonary fibrosis: clinical relevance of pathologic classification. Am J Respir Crit Care Med. 1998;157(4 Pt 1):1301–1315. doi: 10.1164/ajrccm.157.4.9707039. [DOI] [PubMed] [Google Scholar]
  • 18.Kasper M, Haroske G. Alterations in the alveolar epithelium after injury leading to pulmonary fibrosis. Histol Histopathol. 1996;11:463–483. [PubMed] [Google Scholar]
  • 19.King TE, Jr, Schwarz MI, Brown K, et al. Idiopathic pulmonary fibrosis: relationship between histopathologic features and mortality. Am J Respir Crit Care Med. 2001;164:1025–1032. doi: 10.1164/ajrccm.164.6.2001056. [DOI] [PubMed] [Google Scholar]
  • 20.Brody AR, Craighead JE. Interstitial associations of cells lining air spaces in human pulmonary fibrosis. Virchows Arch A Pathol Anat Histol. 1976;372:39–49. doi: 10.1007/BF00429715. [DOI] [PubMed] [Google Scholar]
  • 21.Katzenstein AL. Pathogenesis of “fibrosis” in interstitial pneumonia: an electron microscopic study. Hum Pathol. 1985;16:1015–1024. doi: 10.1016/s0046-8177(85)80279-3. [DOI] [PubMed] [Google Scholar]
  • 22.Coalson JJ. The ultrastructure of human fibrosing alveolitis. Virchows Arch A Pathol Anat Histol. 1982;395:181–199. doi: 10.1007/BF00429611. [DOI] [PubMed] [Google Scholar]
  • 23.Corrin B, Dewar A, Rodriguez-Roisin R, Turner-Warwick M. Fine structural changes in cryptogenic fibrosing alveolitis and asbestosis. J Pathol. 1985;147:107–119. doi: 10.1002/path.1711470206. [DOI] [PubMed] [Google Scholar]
  • 24.Sutinen S, Rainio P, Huhti E, Pokela R. Ultrastructure of terminal respiratory epithelium and prognosis in chronic interstitial pneumonia. Eur J Respir Dis. 1980;61:325–336. [PubMed] [Google Scholar]
  • 25.Kawanami O, Ferrans VJ, Crystal RG. Structure of alveolar epithelial cells in patients with fibrotic lung disorders. Lab Invest. 1982;46:39–53. [PubMed] [Google Scholar]
  • 26.Chilosi M, Poletti V, Murer B, et al. Abnormal reepithelialization and lung remodeling in idiopathic pulmonary fibrosis: the role of deltaN-p63. Lab Invest. 2002;82:1335–1345. doi: 10.1097/01.lab.0000032380.82232.67. [DOI] [PubMed] [Google Scholar]
  • 27.Kuwano K, Kunitake R, Kawasaki M, et al. P21Waf1/Cip1/Sdi1 and p53 expression in association with DNA strand breaks in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 1996;154(2 Pt 1):477–483. doi: 10.1164/ajrccm.154.2.8756825. [DOI] [PubMed] [Google Scholar]
  • 28.Barbas-Filho JV, Ferreira MA, Sesso A, Kairalla RA, Carvalho CR, Capelozzi VL. Evidence of type II pneumocyte apoptosis in the pathogenesis of idiopathic pulmonary fibrosis (IFP)/usual interstitial pneumonia (UIP) J Clin Pathol. 2001;54:132–138. doi: 10.1136/jcp.54.2.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Maeyama T, Kuwano K, Kawasaki M, et al. Upregulation of Fas-signalling molecules in lung epithelial cells from patients with idiopathic pulmonary fibrosis. Eur Respir J. 2001;17:180–189. doi: 10.1183/09031936.01.17201800. [DOI] [PubMed] [Google Scholar]
  • 30.Brody AR, Soler P, Basset F, Haschek WM, Witschi H. Epithelial-mesenchymal associations of cells in human pulmonary fibrosis and in BHT-oxygen-induced fibrosis in mice. Exp Lung Res. 1981;2:207–220. doi: 10.3109/01902148109052316. [DOI] [PubMed] [Google Scholar]
  • 31.Adamson IY, Hedgecock C, Bowden DH. Epithelial cell–fibroblast interactions in lung injury and repair. Am J Pathol. 1990;137:385–392. [PMC free article] [PubMed] [Google Scholar]
  • 32.Sirianni FE, Milaninezhad A, Chu FS, Walker DC. Alteration of fibroblast architecture and loss of basal lamina apertures in human emphysematous lung. Am J Respir Crit Care Med. 2006;173:632–638. doi: 10.1164/rccm.200509-1434OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Uhal BD, Joshi I, Hughes WF, Ramos C, Pardo A, Selman M. Alveolar epithelial cell death adjacent to underlying myofibroblasts in advanced fibrotic human lung. Am J Physiol. 1998;275(6 Pt 1):L1192–L1199. doi: 10.1152/ajplung.1998.275.6.L1192. [DOI] [PubMed] [Google Scholar]
  • 34.Plataki M, Koutsopoulos AV, Darivianaki K, Delides G, Siafakas NM, Bouros D. Expression of apoptotic and antiapoptotic markers in epithelial cells in idiopathic pulmonary fibrosis. Chest. 2005;127:266–274. doi: 10.1378/chest.127.1.266. [DOI] [PubMed] [Google Scholar]
  • 35.Wang R, Ibarra-Sunga O, Verlinski L, Pick R, Uhal BD. Abrogation of bleomycin-induced epithelial apoptosis and lung fibrosis by captopril or by a caspase inhibitor. Am J Physiol Lung Cell Mol Physiol. 2000;279:L143–L151. doi: 10.1152/ajplung.2000.279.1.L143. [DOI] [PubMed] [Google Scholar]
  • 36.Kuwano K, Kunitake R, Maeyama T, et al. Attenuation of bleomycin-induced pneumopathy in mice by a caspase inhibitor. Am J Physiol Lung Cell Mol Physiol. 2001;280:L316–L325. doi: 10.1152/ajplung.2001.280.2.L316. [DOI] [PubMed] [Google Scholar]
  • 37.Lee CG, Cho SJ, Kang MJ, et al. Early growth response gene 1-mediated apoptosis is essential for transforming growth factor beta1-induced pulmonary fibrosis. J Exp Med. 2004;200:377–389. doi: 10.1084/jem.20040104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Strasser A, O’Connor L, Dixit VM. Apoptosis signaling. Annu Rev Biochem. 2000;69:217–245. doi: 10.1146/annurev.biochem.69.1.217. [DOI] [PubMed] [Google Scholar]
  • 39.Hagimoto N, Kuwano K, Miyazaki H, et al. Induction of apoptosis and pulmonary fibrosis in mice in response to ligation of Fas antigen. Am J Respir Cell Mol Biol. 1997;17:272–278. doi: 10.1165/ajrcmb.17.3.2893. [DOI] [PubMed] [Google Scholar]
  • 40.Hagimoto N, Kuwano K, Inoshima I, et al. TGF-beta 1 as an enhancer of Fas-mediated apoptosis of lung epithelial cells. J Immunol. 2002;168:6470–6478. doi: 10.4049/jimmunol.168.12.6470. [DOI] [PubMed] [Google Scholar]
  • 41.Kuwano K, Hagimoto N, Kawasaki M, et al. Essential roles of the Fas-Fas ligand pathway in the development of pulmonary fibrosis. J Clin Invest. 1999;104:13–19. doi: 10.1172/JCI5628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Aoshiba K, Yasui S, Tamaoki J, Nagai A. The Fas/Fas-ligand system is not required for bleomycin-induced pulmonary fibrosis in mice. Am J Respir Crit Care Med. 2000;162(2 Pt 1):695–700. doi: 10.1164/ajrccm.162.2.9907012. [DOI] [PubMed] [Google Scholar]
  • 43.Wallach-Dayan SB, Izbicki G, Cohen PY, Gerstl-Golan R, Fine A, Breuer R. Bleomycin initiates apoptosis of lung epithelial cells by ROS but not by Fas/FasL pathway. Am J Physiol Lung Cell Mol Physiol. 2006;290:L790–L796. doi: 10.1152/ajplung.00300.2004. [DOI] [PubMed] [Google Scholar]
  • 44.Schrier DJ, Kunkel RG, Phan SH. The role of strain variation in murine bleomycin-induced pulmonary fibrosis. Am Rev Respir Dis. 1983;127:63–66. doi: 10.1164/arrd.1983.127.1.63. [DOI] [PubMed] [Google Scholar]
  • 45.Daly HE, Baecher-Allan CM, Barth RK, D’Angio CT, Finkelstein JN. Bleomycin induces strain-dependent alterations in the pattern of epithelial cell-specific marker expression in mouse lung. Toxicol Appl Pharmacol. 1997;142:303–310. doi: 10.1006/taap.1996.8056. [DOI] [PubMed] [Google Scholar]
  • 46.Qunn L, Takemura T, Ikushima S, et al. Hyperplastic epithelial foci in honeycomb lesions in idiopathic pulmonary fibrosis. Virchows Arch. 2002;441:271–278. doi: 10.1007/s00428-002-0618-9. [DOI] [PubMed] [Google Scholar]
  • 47.Deterding RR, Havill AM, Yano T, et al. Prevention of bleomycin-induced lung injury in rats by keratinocyte growth factor. Proc Assoc Am Physicians. 1997;109:254–268. [PubMed] [Google Scholar]
  • 48.Dohi M, Hasegawa T, Yamamoto K, Marshall BC. Hepatocyte growth factor attenuates collagen accumulation in a murine model of pulmonary fibrosis. Am J Respir Crit Care Med. 2000;162:2302–2307. doi: 10.1164/ajrccm.162.6.9908097. [DOI] [PubMed] [Google Scholar]
  • 49.Groves RW, Schmidt-Lucke JA. Recombinant human GM-CSF in the treatment of poorly healing wounds. Adv Skin Wound Care. 2000;13(3 Pt 1):107–112. [PubMed] [Google Scholar]
  • 50.Christensen PJ, Bailie MB, Goodman RE, O’Brien AD, Toews GB, Paine R., III Role of diminished epithelial GM-CSF in the pathogenesis of bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2000;279:L487–L495. doi: 10.1152/ajplung.2000.279.3.L487. [DOI] [PubMed] [Google Scholar]
  • 51.Moore BB, Coffey MJ, Christensen P, et al. GM-CSF regulates bleomycin-induced pulmonary fibrosis via a prostaglandin-dependent mechanism. J Immunol. 2000;165:4032–4039. doi: 10.4049/jimmunol.165.7.4032. [DOI] [PubMed] [Google Scholar]
  • 52.Zhang F, Nielsen LD, Lucas JJ, Mason RJ. Transforming growth factor-beta antagonizes alveolar type II cell proliferation induced by keratinocyte growth factor. Am J Respir Cell Mol Biol. 2004;31:679–686. doi: 10.1165/rcmb.2004-0182OC. [DOI] [PubMed] [Google Scholar]
  • 53.Irigoyen JP, Munoz-Canoves P, Montero L, Koziczak M, Nagamine Y. The plasminogen activator system: biology and regulation. Cell Mol Life Sci. 1999;56:104–132. doi: 10.1007/PL00000615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Eitzman DT, McCoy RD, Zheng X, et al. Bleomycin-induced pulmonary fibrosis in transgenic mice that either lack or overexpress the murine plasminogen activator inhibitor-1 gene. J Clin Invest. 1996;97:232–237. doi: 10.1172/JCI118396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sisson TH, Hattori N, Xu Y, Simon RH. Treatment of bleomycin-induced pulmonary fibrosis by transfer of urokinase-type plasminogen activator genes. Hum Gene Ther. 1999;10:2315–2323. doi: 10.1089/10430349950016960. [DOI] [PubMed] [Google Scholar]
  • 56.Sisson TH, Hanson KE, Subbotina N, Patwardhan A, Hattori N, Simon RH. Inducible lung-specific urokinase expression reduces fibrosis and mortality after lung injury in mice. Am J Physiol Lung Cell Mol Physiol. 2002;283:L1023–L1032. doi: 10.1152/ajplung.00049.2002. [DOI] [PubMed] [Google Scholar]
  • 57.Hattori N, Degen JL, Sisson TH, et al. Bleomycin-induced pulmonary fibrosis in fibrinogen-null mice. J Clin Invest. 2000;106:1341–1350. doi: 10.1172/JCI10531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Swaisgood CM, French EL, Noga C, Simon RH, Ploplis VA. The development of bleomycin-induced pulmonary fibrosis in mice deficient for components of the fibrinolytic system. Am J Pathol. 2000;157:177–187. doi: 10.1016/S0002-9440(10)64529-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Legrand C, Polette M, Tournier JM, et al. uPA/plasmin system-mediated MMP-9 activation is implicated in bronchial epithelial cell migration. Exp Cell Res. 2001;264:326–336. doi: 10.1006/excr.2000.5125. [DOI] [PubMed] [Google Scholar]
  • 60.Chan JC, Duszczyszyn DA, Castellino FJ, Ploplis VA. Accelerated skin wound healing in plasminogen activator inhibitor-1-deficient mice. Am J Pathol. 2001;159:1681–1688. doi: 10.1016/S0002-9440(10)63015-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lazar MH, Christensen PJ, Du M, et al. Plasminogen activator inhibitor-1 impairs alveolar epithelial repair by binding to vitronectin. Am J Respir Cell Mol Biol. 2004;31:672–678. doi: 10.1165/rcmb.2004-0025OC. [DOI] [PubMed] [Google Scholar]
  • 62.Van Leer C, Stutz M, Haeberli A, Geiser T. Urokinase plasminogen activator released by alveolar epithelial cells modulates alveolar epithelial repair in vitro. Thromb Haemost. 2005;94:1257–1264. doi: 10.1160/TH05-03-0162. [DOI] [PubMed] [Google Scholar]
  • 63.Phan SH. The myofibroblast in pulmonary fibrosis. Chest. 2002;122(Suppl 6):286S–289S. doi: 10.1378/chest.122.6_suppl.286s. [DOI] [PubMed] [Google Scholar]
  • 64.Gabbiani G. The myofibroblast in wound healing and fibrocontractive diseases. J Pathol. 2003;200:500–503. doi: 10.1002/path.1427. [DOI] [PubMed] [Google Scholar]
  • 65.Chambers RC, Leoni P, Kaminski N, Laurent GJ, Heller RA. Global expression profiling of fibroblast responses to transforming growth factor-beta1 reveals the induction of inhibitor of differentiation-1 and provides evidence of smooth muscle cell phenotypic switching. Am J Pathol. 2003;162:533–546. doi: 10.1016/s0002-9440(10)63847-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Schmidt M, Sun G, Stacey MA, Mori L, Mattoli S. Identification of circulating fibrocytes as precursors of bronchial myofibroblasts in asthma. J Immunol. 2003;171:380–389. doi: 10.4049/jimmunol.171.1.380. [DOI] [PubMed] [Google Scholar]
  • 67.Quan TE, Cowper S, Wu SP, Bockenstedt LK, Bucala R. Circulating fibrocytes: collagen-secreting cells of the peripheral blood. Int J Biochem Cell Biol. 2004;36:598–606. doi: 10.1016/j.biocel.2003.10.005. [DOI] [PubMed] [Google Scholar]
  • 68.Phillips RJ, Burdick MD, Hong K, et al. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest. 2004;114:438–446. doi: 10.1172/JCI20997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Moore BB, Kolodsick JE, Thannickal VJ, et al. CCR2-mediated recruitment of fibrocytes to the alveolar space after fibrotic injury. Am J Pathol. 2005;166:675–684. doi: 10.1016/S0002-9440(10)62289-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hashimoto N, Jin H, Liu T, Chensue SW, Phan SH. Bone marrow–derived progenitor cells in pulmonary fibrosis. J Clin Invest. 2004;113:243–252. doi: 10.1172/JCI18847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Willis BC, Liebler JM, Luby-Phelps K, et al. Induction of epithelial–mesenchymal transition in alveolar epithelial cells by transforming growth factor-beta1: potential role in idiopathic pulmonary fibrosis. Am J Pathol. 2005;166:1321–1332. doi: 10.1016/s0002-9440(10)62351-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yao HW, Xie QM, Chen JQ, Deng YM, Tang HF. TGF-beta1 induces alveolar epithelial to mesenchymal transition in vitro. Life Sci. 2004;76:29–37. doi: 10.1016/j.lfs.2004.06.019. [DOI] [PubMed] [Google Scholar]
  • 73.Kasai H, Allen JT, Mason RM, Kamimura T, Zhang Z. TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT) Respir Res. 2005;6:56. doi: 10.1186/1465-9921-6-56. www.respiratory-research.com. [DOI] [PMC free article] [PubMed]
  • 74.Kotaru C, Schoonover KJ, Trudeau JB, et al. Regional fibroblast heterogeneity in the lung: implications for remodeling. Am J Respir Crit Care Med. 2006;173(11):1208–1215. doi: 10.1164/rccm.200508-1218OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Chang HY, Chi JT, Dudoit S, et al. Diversity, topographic differentiation, and positional memory in human fibroblasts. Proc Natl Acad Sci USA. 2002;99:12877–12882. doi: 10.1073/pnas.162488599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Desmouliere A, Redard M, Darby I, Gabbiani G. Apoptosis mediates the decrease in cellularity during the transition between granulation tissue and scar. Am J Pathol. 1995;146:56–66. [PMC free article] [PubMed] [Google Scholar]
  • 77.Kissin EY, Lemaire R, Korn JH, Lafyatis R. Transforming growth factor beta induces fibroblast fibrillin-1 matrix formation. Arthritis Rheum. 2002;46:3000–3009. doi: 10.1002/art.10621. [DOI] [PubMed] [Google Scholar]
  • 78.Tanaka T, Yoshimi M, Maeyama T, Hagimoto N, Kuwano K, Hara N. Resistance to Fas-mediated apoptosis in human lung fibroblast. Eur Respir J. 2002;20:359–368. doi: 10.1183/09031936.02.00252602. [DOI] [PubMed] [Google Scholar]
  • 79.Buhling F, Wille A, Rocken C, et al. Altered expression of membrane-bound and soluble CD95/Fas contributes to the resistance of fibrotic lung fibroblasts to FasL induced apoptosis. Respir Res. 2005;6:37. doi: 10.1186/1465-9921-6-37. www.respiratory-research.com. [DOI] [PMC free article] [PubMed]
  • 80.Moodley YP, Misso NL, Scaffidi AK, et al. Inverse effects of interleukin-6 on apoptosis of fibroblasts from pulmonary fibrosis and normal lungs. Am J Respir Cell Mol Biol. 2003;29:490–498. doi: 10.1165/rcmb.2002-0262OC. [DOI] [PubMed] [Google Scholar]
  • 81.Frankel SK, Cosgrove GP, Cha SI, et al. TNF-alpha sensitizes normal and fibrotic human lung fibroblasts to Fas-induced apoptosis. Am J Respir Cell Mol Biol. 2006;34:293–304. doi: 10.1165/rcmb.2005-0155OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Moodley YP, Caterina P, Scaffidi AK, et al. Comparison of the morphological and biochemical changes in normal human lung fibroblasts and fibroblasts derived from lungs of patients with idiopathic pulmonary fibrosis during FasL-induced apoptosis. J Pathol. 2004;202:486–495. doi: 10.1002/path.1531. [DOI] [PubMed] [Google Scholar]
  • 83.Horowitz JC, Cui Z, Moore TA, et al. Constitutive activation of prosurvival signaling in alveolar mesenchymal cells isolated from patients with nonresolving acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol. 2006;290:L415–L425. doi: 10.1152/ajplung.00276.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Matsuyama W, Watanabe M, Shirahama Y, et al. Discoidin domain receptor 1 contributes to the survival of lung fibroblast in idiopathic pulmonary fibrosis. Am J Pathol. 2006;168:866–877. doi: 10.2353/ajpath.2006.050801. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 85.Chodon T, Sugihara T, Igawa HH, Funayama E, Furukawa H. Keloid-derived fibroblasts are refractory to Fas-mediated apoptosis and neutralization of autocrine transforming growth factor-beta1 can abrogate this resistance. Am J Pathol. 2000;157:1661–1669. doi: 10.1016/s0002-9440(10)64803-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Jelaska A, Korn JH. Role of apoptosis and transforming growth factor beta1 in fibroblast selection and activation in systemic sclerosis. Arthritis Rheum. 2000;43:2230–2239. doi: 10.1002/1529-0131(200010)43:10<2230::AID-ANR10>3.0.CO;2-8. [DOI] [PubMed] [Google Scholar]
  • 87.Horowitz JC, Lee DY, Waghray M, et al. Activation of the pro-survival phosphatidylinositol 3-kinase/AKT pathway by transforming growth factor-beta1 in mesenchymal cells is mediated by p38 MAPK-dependent induction of an autocrine growth factor. J Biol Chem. 2004;279:1359–1367. doi: 10.1074/jbc.M306248200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhang HY, Phan SH. Inhibition of myofibroblast apoptosis by transforming growth factor beta(1) Am J Respir Cell Mol Biol. 1999;21:658–665. doi: 10.1165/ajrcmb.21.6.3720. [DOI] [PubMed] [Google Scholar]
  • 89.Grinnell F, Zhu M, Carlson MA, Abrams JM. Release of mechanical tension triggers apoptosis of human fibroblasts in a model of regressing granulation tissue. Exp Cell Res. 1999;248:608–619. doi: 10.1006/excr.1999.4440. [DOI] [PubMed] [Google Scholar]
  • 90.Zhu YK, Umino T, Liu XD, et al. Contraction of fibroblast-containing collagen gels: initial collagen concentration regulates the degree of contraction and cell survival. In Vitro Cell Dev Biol Anim. 2001;37:10–16. doi: 10.1290/1071-2690(2001)037<0010:COFCCG>2.0.CO;2. [DOI] [PubMed] [Google Scholar]
  • 91.Xia H, Nho RS, Kahm J, Kleidon J, Henke CA. Focal adhesion kinase is upstream of phosphatidylinositol 3-kinase/Akt in regulating fibroblast survival in response to contraction of type I collagen matrices via a beta 1 integrin viability signaling pathway. J Biol Chem. 2004;279:33024–33034. doi: 10.1074/jbc.M313265200. [DOI] [PubMed] [Google Scholar]
  • 92.Thannickal VJ, Lee DY, White ES, et al. Myofibroblast differentiation by transforming growth factor-beta1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J Biol Chem. 2003;278:12384–12389. doi: 10.1074/jbc.M208544200. [DOI] [PubMed] [Google Scholar]
  • 93.Kobayashi T, Liu X, Wen FQ, et al. Smad3 mediates TGF-beta1 induction of VEGF production in lung fibroblasts. Biochem Biophys Res Commun. 2005;327:393–398. doi: 10.1016/j.bbrc.2004.12.032. [DOI] [PubMed] [Google Scholar]
  • 94.Vittal R, Horowitz JC, Moore BB, et al. Modulation of prosurvival signaling in fibroblasts by a protein kinase inhibitor protects against fibrotic tissue injury. Am J Pathol. 2005;166:367–375. doi: 10.1016/S0002-9440(10)62260-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Khalil N, Xu YD, O’Connor R, Duronio V. Proliferation of pulmonary interstitial fibroblasts is mediated by transforming growth factor-beta1-induced release of extracellular fibroblast growth factor-2 and phosphorylation of p38 MAPK and JNK. J Biol Chem. 2005;280:43000–43009. doi: 10.1074/jbc.M510441200. [DOI] [PubMed] [Google Scholar]
  • 96.Matsuoka H, Arai T, Mori M, et al. A p38 MAPK inhibitor, FR-167653, ameliorates murine bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2002;283:L103–L112. doi: 10.1152/ajplung.00187.2001. [DOI] [PubMed] [Google Scholar]
  • 97.Lappi-Blanco E, Soini Y, Paakko P. Apoptotic activity is increased in the newly formed fibromyxoid connective tissue in bronchiolitis obliterans organizing pneumonia. Lung. 1999;177:367–376. doi: 10.1007/pl00007654. [DOI] [PubMed] [Google Scholar]
  • 98.Jordana M, Schulman J, McSharry C, et al. Heterogeneous proliferative characteristics of human adult lung fibroblast lines and clonally derived fibroblasts from control and fibrotic tissue. Am Rev Respir Dis. 1988;137:579–584. doi: 10.1164/ajrccm/137.3.579. [DOI] [PubMed] [Google Scholar]
  • 99.Raghu G, Chen YY, Rusch V, Rabinovitch PS. Differential proliferation of fibroblasts cultured from normal and fibrotic human lungs. Am Rev Respir Dis. 1988;138:703–708. doi: 10.1164/ajrccm/138.3.703. [DOI] [PubMed] [Google Scholar]
  • 100.Mio T, Nagai S, Kitaichi M, Kawatani A, Izumi T. Proliferative characteristics of fibroblast lines derived from open lung biopsy specimens of patients with IPF (UIP) Chest. 1992;102:832–837. doi: 10.1378/chest.102.3.832. [DOI] [PubMed] [Google Scholar]
  • 101.Ramos C, Montano M, Garcia-Alvarez J, et al. Fibroblasts from idiopathic pulmonary fibrosis and normal lungs differ in growth rate, apoptosis, and tissue inhibitor of metalloproteinases expression. Am J Respir Cell Mol Biol. 2001;24:591–598. doi: 10.1165/ajrcmb.24.5.4333. [DOI] [PubMed] [Google Scholar]
  • 102.Marshall RP, McAnulty RJ, Laurent GJ. Angiotensin II is mitogenic for human lung fibroblasts via activation of the type 1 receptor. Am J Respir Crit Care Med. 2000;161:1999–2004. doi: 10.1164/ajrccm.161.6.9907004. [DOI] [PubMed] [Google Scholar]
  • 103.Moodley YP, Scaffidi AK, Misso NL, et al. Fibroblasts isolated from normal lungs and those with idiopathic pulmonary fibrosis differ in interleukin-6/gp130-mediated cell signaling and proliferation. Am J Pathol. 2003;163:345–354. doi: 10.1016/S0002-9440(10)63658-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Border WA, Noble NA. Transforming growth factor beta in tissue fibrosis. N Engl J Med. 1994;331:1286–1292. doi: 10.1056/NEJM199411103311907. [DOI] [PubMed] [Google Scholar]
  • 105.Xaubet A, Marin-Arguedas A, Lario S, et al. Transforming growth factor-beta1 gene polymorphisms are associated with disease progression in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2003;168:431–435. doi: 10.1164/rccm.200210-1165OC. [DOI] [PubMed] [Google Scholar]
  • 106.Khalil N, Parekh TV, O’Connor R, et al. Regulation of the effects of TGF-beta 1 by activation of latent TGF-beta 1 and differential expression of TGF-beta receptors (T beta R-I and T beta R-II) in idiopathic pulmonary fibrosis. Thorax. 2001;56:907–915. doi: 10.1136/thorax.56.12.907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector-mediated gene transfer of active transforming growth factor-beta1 induces prolonged severe fibrosis in rat lung. J Clin Invest. 1997;100:768–776. doi: 10.1172/JCI119590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bonniaud P, Margetts PJ, Kolb M, et al. Progressive transforming growth factor {beta}1-induced lung fibrosis is blocked by an orally active ALK5 kinase inhibitor. Am J Respir Crit Care Med. 2005;171:889–898. doi: 10.1164/rccm.200405-612OC. [DOI] [PubMed] [Google Scholar]
  • 109.Bonniaud P, Kolb M, Galt T, et al. Smad3 null mice develop airspace enlargement and are resistant to TGF-beta-mediated pulmonary fibrosis. J Immunol. 2004;173:2099–2108. doi: 10.4049/jimmunol.173.3.2099. [DOI] [PubMed] [Google Scholar]
  • 110.Kelley J, Shull S, Walsh JJ, Cutroneo KR, Absher M. Auto-induction of transforming growth factor-beta in human lung fibroblasts. Am J Respir Cell Mol Biol. 1993;8:417–424. doi: 10.1165/ajrcmb/8.4.417. [DOI] [PubMed] [Google Scholar]
  • 111.Kelley J, Fabisiak JP, Hawes K, Absher M. Cytokine signaling in lung: transforming growth factor-beta secretion by lung fibroblasts. Am J Physiol. 1991;260(2 Pt 1):L123–L128. doi: 10.1152/ajplung.1991.260.2.L123. [DOI] [PubMed] [Google Scholar]
  • 112.Breen E, Shull S, Burne S, et al. Bleomycin regulation of transforming growth factor-beta mRNA in rat lung fibroblasts. Am J Respir Cell Mol Biol. 1992;6:146–152. doi: 10.1165/ajrcmb/6.2.146. [DOI] [PubMed] [Google Scholar]
  • 113.Bergeron A, Soler P, Kambouchner M, et al. Cytokine profiles in idiopathic pulmonary fibrosis suggest an important role for TGF-beta and IL-10. Eur Respir J. 2003;22:69–76. doi: 10.1183/09031936.03.00014703. [DOI] [PubMed] [Google Scholar]
  • 114.Coker RK, Laurent GJ, Jeffery PK, du Bois RM, Black CM, McAnulty RJ. Localisation of transforming growth factor beta1 and beta3 mRNA transcripts in normal and fibrotic human lung. Thorax. 2001;56:549–556. doi: 10.1136/thorax.56.7.549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Kapanci Y, Desmouliere A, Pache JC, Redard M, Gabbiani G. Cytoskeletal protein modulation in pulmonary alveolar myofibroblasts during idiopathic pulmonary fibrosis. Possible role of transforming growth factor beta and tumor necrosis factor alpha. Am J Respir Crit Care Med. 1995;152(6 Pt 1):2163–2169. doi: 10.1164/ajrccm.152.6.8520791. [DOI] [PubMed] [Google Scholar]
  • 116.Khalil N, O’Connor RN, Flanders KC, Unruh H. TGF-beta 1, but not TGF-beta 2 or TGF-beta 3, is differentially present in epithelial cells of advanced pulmonary fibrosis: an immunohistochemical study. Am J Respir Cell Mol Biol. 1996;14:131–138. doi: 10.1165/ajrcmb.14.2.8630262. [DOI] [PubMed] [Google Scholar]
  • 117.Xu YD, Hua J, Mui A, O’Connor R, Grotendorst G, Khalil N. Release of biologically active TGF-beta1 by alveolar epithelial cells results in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2003;285:L527–L539. doi: 10.1152/ajplung.00298.2002. [DOI] [PubMed] [Google Scholar]
  • 118.Yoo J, Ghiassi M, Jirmanova L, et al. Transforming growth factor-beta-induced apoptosis is mediated by Smad-dependent expression of GADD45b through p38 activation. J Biol Chem. 2003;278:43001–43007. doi: 10.1074/jbc.M307869200. [DOI] [PubMed] [Google Scholar]
  • 119.Yanagisawa K, Osada H, Masuda A, et al. Induction of apoptosis by Smad3 and down-regulation of Smad3 expression in response to TGF-beta in human normal lung epithelial cells. Oncogene. 1998;17:1743–1747. doi: 10.1038/sj.onc.1202052. [DOI] [PubMed] [Google Scholar]
  • 120.Solovyan VT, Keski-Oja J. Proteolytic activation of latent TGF-beta precedes caspase-3 activation and enhances apoptotic death of lung epithelial cells. J Cell Physiol. 2006;207:445–453. doi: 10.1002/jcp.20607. [DOI] [PubMed] [Google Scholar]
  • 121.Varga J, Rosenbloom J, Jimenez SA. Transforming growth factor beta (TGF beta) causes a persistent increase in steady-state amounts of type I and type III collagen and fibronectin mRNAs in normal human dermal fibroblasts. Biochem J. 1987;247:597–604. doi: 10.1042/bj2470597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Eickelberg O, Kohler E, Reichenberger F, et al. Extracellular matrix deposition by primary human lung fibroblasts in response to TGF-beta1 and TGF-beta3. Am J Physiol. 1999;276(5 Pt 1):L814–L824. doi: 10.1152/ajplung.1999.276.5.L814. [DOI] [PubMed] [Google Scholar]
  • 123.Thannickal VJ, Fanburg BL. Activation of an H2O2-generating NADH oxidase in human lung fibroblasts by transforming growth factor beta 1. J Biol Chem. 1995;270:30334–30338. doi: 10.1074/jbc.270.51.30334. [DOI] [PubMed] [Google Scholar]
  • 124.Shiratori M, Michalopoulos G, Shinozuka H, Singh G, Ogasawara H, Katyal SL. Hepatocyte growth factor stimulates DNA synthesis in alveolar epithelial type II cells in vitro. Am J Respir Cell Mol Biol. 1995;12:171–180. doi: 10.1165/ajrcmb.12.2.7532419. [DOI] [PubMed] [Google Scholar]
  • 125.Nishimura S, Takahashi M, Ota S, Hirano M, Hiraishi H. Hepatocyte growth factor accelerates restitution of intestinal epithelial cells. J Gastroenterol. 1998;33:172–178. doi: 10.1007/s005350050066. [DOI] [PubMed] [Google Scholar]
  • 126.Inoue T, Okada H, Kobayashi T, et al. Hepatocyte growth factor counteracts transforming growth factor-beta1, through attenuation of connective tissue growth factor induction, and prevents renal fibrogenesis in 5/6 nephrectomized mice. FASEB J. 2003;17:268–270. doi: 10.1096/fj.02-0442fje. [DOI] [PubMed] [Google Scholar]
  • 127.Dworkin LD, Gong R, Tolbert E, et al. Hepatocyte growth factor ameliorates progression of interstitial fibrosis in rats with established renal injury. Kidney Int. 2004;65:409–419. doi: 10.1111/j.1523-1755.2004.00417.x. [DOI] [PubMed] [Google Scholar]
  • 128.Mizuno S, Matsumoto K, Li MY, Nakamura T. HGF reduces advancing lung fibrosis in mice: a potential role for MMP-dependent myofibroblast apoptosis. FASEB J. 2005;19:580–582. doi: 10.1096/fj.04-1535fje. [DOI] [PubMed] [Google Scholar]
  • 129.Taniyama Y, Morishita R, Nakagami H, et al. Potential contribution of a novel antifibrotic factor, hepatocyte growth factor, to prevention of myocardial fibrosis by angiotensin II blockade in cardiomyopathic hamsters. Circulation. 2000;102:246–252. doi: 10.1161/01.cir.102.2.246. [DOI] [PubMed] [Google Scholar]
  • 130.Marchand-Adam S, Marchal J, Cohen M, et al. Defect of hepatocyte growth factor secretion by fibroblasts in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2003;168:1156–1161. doi: 10.1164/rccm.200212-1514OC. [DOI] [PubMed] [Google Scholar]
  • 131.Matsumoto K, Tajima H, Okazaki H, Nakamura T. Negative regulation of hepatocyte growth factor gene expression in human lung fibroblasts and leukemic cells by transforming growth factor-beta 1 and glucocorticoids. J Biol Chem. 1992;267:24917–24920. [PubMed] [Google Scholar]
  • 132.Hattori N, Mizuno S, Yoshida Y, et al. The plasminogen activation system reduces fibrosis in the lung by a hepatocyte growth factor-dependent mechanism. Am J Pathol. 2004;164:1091–1098. doi: 10.1016/S0002-9440(10)63196-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Eickelberg O, Pansky A, Koehler E, et al. Molecular mechanisms of TGF-(beta) antagonism by interferon (gamma) and cyclosporine A in lung fibroblasts. FASEB J. 2001;15:797–806. doi: 10.1096/fj.00-0233com. [DOI] [PubMed] [Google Scholar]
  • 134.Nagahori T, Dohi M, Matsumoto K, et al. Interferon-gamma upregulates the c-Met/hepatocyte growth factor receptor expression in alveolar epithelial cells. Am J Respir Cell Mol Biol. 1999;21:490–497. doi: 10.1165/ajrcmb.21.4.3614. [DOI] [PubMed] [Google Scholar]
  • 135.Rubin JS, Osada H, Finch PW, Taylor WG, Rudikoff S, Aaronson SA. Purification and characterization of a newly identified growth factor specific for epithelial cells. Proc Natl Acad Sci USA. 1989;86:802–806. doi: 10.1073/pnas.86.3.802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Finch PW, Rubin JS, Miki T, Ron D, Aaronson SA. Human KGF is FGF-related with properties of a paracrine effector of epithelial cell growth. Science. 1989;245(4919):752–755. doi: 10.1126/science.2475908. [DOI] [PubMed] [Google Scholar]
  • 137.Deterding RR, Jacoby CR, Shannon JM. Acidic fibroblast growth factor and keratinocyte growth factor stimulate fetal rat pulmonary epithelial growth. Am J Physiol. 1996;271(4 Pt 1):L495–L505. doi: 10.1152/ajplung.1996.271.4.L495. [DOI] [PubMed] [Google Scholar]
  • 138.Bao S, Wang Y, Sweeney P, et al. Keratinocyte growth factor induces Akt kinase activity and inhibits Fas-mediated apoptosis in A549 lung epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2005;288:L36–L42. doi: 10.1152/ajplung.00309.2003. [DOI] [PubMed] [Google Scholar]
  • 139.Portnoy J, Pan T, Dinarello CA, et al. Alveolar type II cells inhibit fibroblast proliferation: role of IL-1alpha. Am J Physiol Lung Cell Mol Physiol. 2006;290:L307–L316. doi: 10.1152/ajplung.00102.2005. [DOI] [PubMed] [Google Scholar]
  • 140.Marchand-Adam S, Plantier L, Bernuau D, et al. Keratinocyte growth factor expression by fibroblasts in pulmonary fibrosis: poor response to interleukin-1beta. Am J Respir Cell Mol Biol. 2005;32:470–477. doi: 10.1165/rcmb.2004-0205OC. [DOI] [PubMed] [Google Scholar]
  • 141.Li X, Rayford H, Uhal BD. Essential roles for angiotensin receptor AT1a in bleomycin-induced apoptosis and lung fibrosis in mice. Am J Pathol. 2003;163:2523–2530. doi: 10.1016/S0002-9440(10)63607-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Otsuka M, Takahashi H, Shiratori M, Chiba H, Abe S. Reduction of bleomycin induced lung fibrosis by candesartan cilexetil, an angiotensin II type 1 receptor antagonist. Thorax. 2004;59:31–38. doi: 10.1136/thx.2003.000893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wang R, Zagariya A, Ang E, Ibarra-Sunga O, Uhal BD. Fas-induced apoptosis of alveolar epithelial cells requires ANG II generation and receptor interaction. Am J Physiol. 1999;277(6 Pt 1):L1245–L1250. doi: 10.1152/ajplung.1999.277.6.L1245. [DOI] [PubMed] [Google Scholar]
  • 144.Papp M, Li X, Zhuang J, Wang R, Uhal BD. Angiotensin receptor subtype AT(1) mediates alveolar epithelial cell apoptosis in response to ANG II. Am J Physiol Lung Cell Mol Physiol. 2002;282:L713–L718. doi: 10.1152/ajplung.00103.2001. [DOI] [PubMed] [Google Scholar]
  • 145.Uhal BD, Gidea C, Bargout R, et al. Captopril inhibits apoptosis in human lung epithelial cells: a potential antifibrotic mechanism. Am J Physiol. 1998;275(5 Pt 1):L1013–L1017. doi: 10.1152/ajplung.1998.275.5.L1013. [DOI] [PubMed] [Google Scholar]
  • 146.Nguyen L, Ward WF, Ts’ao CH, Molteni A. Captopril inhibits proliferation of human lung fibroblasts in culture: a potential antifibrotic mechanism. Proc Soc Exp Biol Med. 1994;205:80–84. doi: 10.3181/00379727-205-43681. [DOI] [PubMed] [Google Scholar]
  • 147.Ketteler M, Noble NA, Border WA. Transforming growth factor-beta and angiotensin II: the missing link from glomerular hyperfiltration to glomerulosclerosis? Annu Rev Physiol. 1995;57:279–295. doi: 10.1146/annurev.ph.57.030195.001431. [DOI] [PubMed] [Google Scholar]
  • 148.Campbell SE, Katwa LC. Angiotensin II stimulated expression of transforming growth factor-beta1 in cardiac fibroblasts and myofibroblasts. J Mol Cell Cardiol. 1997;29:1947–1958. doi: 10.1006/jmcc.1997.0435. [DOI] [PubMed] [Google Scholar]
  • 149.Cantin AM, North SL, Fells GA, Hubbard RC, Crystal RG. Oxidant-mediated epithelial cell injury in idiopathic pulmonary fibrosis. J Clin Invest. 1987;79:1665–1673. doi: 10.1172/JCI113005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Cantin AM, Hubbard RC, Crystal RG. Glutathione deficiency in the epithelial lining fluid of the lower respiratory tract in idiopathic pulmonary fibrosis. Am Rev Respir Dis. 1989;139:370–372. doi: 10.1164/ajrccm/139.2.370. [DOI] [PubMed] [Google Scholar]
  • 151.Meyer A, Buhl R, Magnussen H. The effect of oral N-acetylcysteine on lung glutathione levels in idiopathic pulmonary fibrosis. Eur Respir J. 1994;7:431–436. doi: 10.1183/09031936.94.07030431. [DOI] [PubMed] [Google Scholar]
  • 152.Behr J, Maier K, Degenkolb B, Krombach F, Vogelmeier C. Antioxidative and clinical effects of high-dose N-acetylcysteine in fibrosing alveolitis: adjunctive therapy to maintenance immunosuppression. Am J Respir Crit Care Med. 1997;156:1897–1901. doi: 10.1164/ajrccm.156.6.9706065. [DOI] [PubMed] [Google Scholar]
  • 153.Demedts M, Behr J, Buhl R, et al. High-dose acetylcysteine in idiopathic pulmonary fibrosis. N Engl J Med. 2005;353:2229–2242. doi: 10.1056/NEJMoa042976. [DOI] [PubMed] [Google Scholar]
  • 154.Hunninghake GW. Antioxidant therapy for idiopathic pulmonary fibrosis. N Engl J Med. 2005;353:2285–2287. doi: 10.1056/NEJMe058210. [DOI] [PubMed] [Google Scholar]
  • 155.Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1005–L1028. doi: 10.1152/ajplung.2000.279.6.L1005. [DOI] [PubMed] [Google Scholar]
  • 156.Thannickal VJ, Aldweib KD, Fanburg BL. Tyrosine phosphorylation regulates H2O2 production in lung fibroblasts stimulated by transforming growth factor beta1. J Biol Chem. 1998;273:23611–23615. doi: 10.1074/jbc.273.36.23611. [DOI] [PubMed] [Google Scholar]
  • 157.Waghray M, Cui Z, Horowitz JC, et al. Hydrogen peroxide is a diffusible paracrine signal for the induction of epithelial cell death by activated myofibroblasts. FASEB J. 2005;19:854–856. doi: 10.1096/fj.04-2882fje. [DOI] [PubMed] [Google Scholar]
  • 158.Charbeneau RP, Peters-Golden M. Eicosanoids: mediators and therapeutic targets in fibrotic lung disease. Clin Sci (Lond) 2005;108:479–491. doi: 10.1042/CS20050012. [DOI] [PubMed] [Google Scholar]
  • 159.Borok Z, Gillissen A, Buhl R, et al. Augmentation of functional prostaglandin E levels on the respiratory epithelial surface by aerosol administration of prostaglandin E. Am Rev Respir Dis. 1991;144:1080–1084. doi: 10.1164/ajrccm/144.5.1080. [DOI] [PubMed] [Google Scholar]
  • 160.Petkova DK, Clelland CA, Ronan JE, Lewis S, Knox AJ. Reduced expression of cyclooxygenase (COX) in idiopathic pulmonary fibrosis and sarcoidosis. Histopathology. 2003;43:381–386. doi: 10.1046/j.1365-2559.2003.01718.x. [DOI] [PubMed] [Google Scholar]
  • 161.Wilborn J, Bailie M, Coffey M, Burdick M, Strieter R, Peters-Golden M. Constitutive activation of 5-lipoxygenase in the lungs of patients with idiopathic pulmonary fibrosis. J Clin Invest. 1996;97:1827–1836. doi: 10.1172/JCI118612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Keerthisingam CB, Jenkins RG, Harrison NK, et al. Cyclooxygenase-2 deficiency results in a loss of the anti-proliferative response to transforming growth factor-beta in human fibrotic lung fibroblasts and promotes bleomycin-induced pulmonary fibrosis in mice. Am J Pathol. 2001;158:1411–1422. doi: 10.1016/s0002-9440(10)64092-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Peters-Golden M, Bailie M, Marshall T, et al. Protection from pulmonary fibrosis in leukotriene-deficient mice. Am J Respir Crit Care Med. 2002;165:229–235. doi: 10.1164/ajrccm.165.2.2104050. [DOI] [PubMed] [Google Scholar]
  • 164.Bitterman PB, Wewers MD, Rennard SI, Adelberg S, Crystal RG. Modulation of alveolar macrophage-driven fibroblast proliferation by alternative macrophage mediators. J Clin Invest. 1986;77:700–708. doi: 10.1172/JCI112364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Lama V, Moore BB, Christensen P, Toews GB, Peters-Golden M. Prostaglandin E2 synthesis and suppression of fibroblast proliferation by alveolar epithelial cells is cyclo-oxygenase-2-dependent. Am J Respir Cell Mol Biol. 2002;27:752–758. doi: 10.1165/rcmb.4857. [DOI] [PubMed] [Google Scholar]
  • 166.Pan T, Mason RJ, Westcott JY, Shannon JM. Rat alveolar type II cells inhibit lung fibroblast proliferation in vitro. Am J Respir Cell Mol Biol. 2001;25:353–361. doi: 10.1165/ajrcmb.25.3.4004. [DOI] [PubMed] [Google Scholar]
  • 167.Kohyama T, Ertl RF, Valenti V, et al. Prostaglandin E(2) inhibits fibroblast chemotaxis. Am J Physiol Lung Cell Mol Physiol. 2001;281:L1257–L1263. doi: 10.1152/ajplung.2001.281.5.L1257. [DOI] [PubMed] [Google Scholar]
  • 168.White ES, Atrasz RG, Dickie EG, et al. Prostaglandin E(2) inhibits fibroblast migration by E-prostanoid 2 receptor-mediated increase in PTEN activity. Am J Respir Cell Mol Biol. 2005;32:135–141. doi: 10.1165/rcmb.2004-0126OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Kolodsick JE, Peters-Golden M, Larios J, Toews GB, Thannickal VJ, Moore BB. Prostaglandin E2 inhibits fibroblast to myofibroblast transition via E. prostanoid receptor 2 signaling and cyclic adenosine monophosphate elevation. Am J Respir Cell Mol Biol. 2003;29:537–544. doi: 10.1165/rcmb.2002-0243OC. [DOI] [PubMed] [Google Scholar]
  • 170.Goldstein RH, Polgar P. The effect and interaction of bradykinin and prostaglandins on protein and collagen production by lung fibroblasts. J Biol Chem. 1982;257:8630–8633. [PubMed] [Google Scholar]
  • 171.Ghosh M, Stewart A, Tucker DE, Bonventre JV, Murphy RC, Leslie CC. Role of cytosolic phospholipase A(2) in prostaglandin E(2) production by lung fibroblasts. Am J Respir Cell Mol Biol. 2004;30:91–100. doi: 10.1165/rcmb.2003-0005OC. [DOI] [PubMed] [Google Scholar]
  • 172.Ehrenreich H, Anderson RW, Fox CH, et al. Endothelins, peptides with potent vasoactive properties, are produced by human macrophages. J Exp Med. 1990;172:1741–1748. doi: 10.1084/jem.172.6.1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Shahar I, Fireman E, Topilsky M, et al. Effect of endothelin-1 on alpha-smooth muscle actin expression and on alveolar fibroblasts proliferation in interstitial lung diseases. Int J Immunopharmacol. 1999;21:759–775. doi: 10.1016/s0192-0561(99)00056-9. [DOI] [PubMed] [Google Scholar]
  • 174.Shi-Wen X, Chen Y, Denton CP, et al. Endothelin-1 promotes myofibroblast induction through the ETA receptor via a rac/phosphoinositide 3-kinase/Akt-dependent pathway and is essential for the enhanced contractile phenotype of fibrotic fibroblasts. Mol Biol Cell. 2004;15:2707–2719. doi: 10.1091/mbc.E03-12-0902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Goto T, Yanaga F, Ohtsuki I. Studies on the endothelin-1-induced contraction of rat granulation tissue pouch mediated by myofibroblasts. Biochim Biophys Acta. 1998;1405(1–3):55–66. doi: 10.1016/s0167-4889(98)00105-0. [DOI] [PubMed] [Google Scholar]
  • 176.Xu SW, Howat SL, Renzoni EA, et al. Endothelin-1 induces expression of matrix-associated genes in lung fibroblasts through MEK/ERK. J Biol Chem. 2004;279:23098–23103. doi: 10.1074/jbc.M311430200. [DOI] [PubMed] [Google Scholar]
  • 177.Giaid A, Michel RP, Stewart DJ, Sheppard M, Corrin B, Hamid Q. Expression of endothelin-1 in lungs of patients with cryptogenic fibrosing alveolitis. Lancet. 1993;341:1550–1554. doi: 10.1016/0140-6736(93)90694-c. [DOI] [PubMed] [Google Scholar]
  • 178.Saleh D, Furukawa K, Tsao MS, et al. Elevated expression of endothelin-1 and endothelin-converting enzyme-1 in idiopathic pulmonary fibrosis: possible involvement of proinflammatory cytokines. Am J Respir Cell Mol Biol. 1997;16:187–193. doi: 10.1165/ajrcmb.16.2.9032126. [DOI] [PubMed] [Google Scholar]
  • 179.Hocher B, Schwarz A, Fagan KA, et al. Pulmonary fibrosis and chronic lung inflammation in ET-1 transgenic mice. Am J Respir Cell Mol Biol. 2000;23:19–26. doi: 10.1165/ajrcmb.23.1.4030. [DOI] [PubMed] [Google Scholar]
  • 180.Park SH, Saleh D, Giaid A, Michel RP. Increased endothelin-1 in bleomycin-induced pulmonary fibrosis and the effect of an endothelin receptor antagonist. Am J Respir Crit Care Med. 1997;156(2 Pt 1):600–608. doi: 10.1164/ajrccm.156.2.9607123. [DOI] [PubMed] [Google Scholar]
  • 181.McGowan SE. Extracellular matrix and the regulation of lung development and repair. FASEB J. 1992;6:2895–2904. [PubMed] [Google Scholar]
  • 182.Bienkowski RS, Gotkin MG. Control of collagen deposition in mammalian lung. Proc Soc Exp Biol Med. 1995;209:118–140. doi: 10.3181/00379727-209-43886a. [DOI] [PubMed] [Google Scholar]
  • 183.Frisch SM, Francis H. Disruption of epithelial cell–matrix interactions induces apoptosis. J Cell Biol. 1994;124:619–626. doi: 10.1083/jcb.124.4.619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Hadden HL, Henke CA. Induction of lung fibroblast apoptosis by soluble fibronectin peptides. Am J Respir Crit Care Med. 2000;162(4 Pt 1):1553–1560. doi: 10.1164/ajrccm.162.4.2001015. [DOI] [PubMed] [Google Scholar]
  • 185.Serini G, Bochaton-Piallat ML, Ropraz P, et al. The fibronectin domain ED-A is crucial for myofibroblastic phenotype induction by transforming growth factor-beta1. J Cell Biol. 1998;142:873–881. doi: 10.1083/jcb.142.3.873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Swartz MA, Tschumperlin DJ, Kamm RD, Drazen JM. Mechanical stress is communicated between different cell types to elicit matrix remodeling. Proc Natl Acad Sci USA. 2001;98:6180–6185. doi: 10.1073/pnas.111133298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Eckes B, Krieg T. Regulation of connective tissue homeostasis in the skin by mechanical forces. Clin Exp Rheumatol. 2004;22(Suppl 33):S73–S76. [PubMed] [Google Scholar]
  • 188.Hinz B, Gabbiani G. Mechanisms of force generation and transmission by myofibroblasts. Curr Opin Biotechnol. 2003;14:538–546. doi: 10.1016/j.copbio.2003.08.006. [DOI] [PubMed] [Google Scholar]
  • 189.Selman M, Ruiz V, Cabrera S, et al. TIMP-1, -2, -3, and -4 in idiopathic pulmonary fibrosis: a prevailing nondegradative lung microenvironment? Am J Physiol Lung Cell Mol Physiol. 2000;279:L562–L574. doi: 10.1152/ajplung.2000.279.3.L562. [DOI] [PubMed] [Google Scholar]
  • 190.Legrand C, Gilles C, Zahm JM, et al. Airway epithelial cell migration dynamics. MMP-9 role in cell-extracellular matrix remodeling. J Cell Biol. 1999;146:517–529. doi: 10.1083/jcb.146.2.517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Lee SL, Dickson RB, Lin CY. Activation of hepatocyte growth factor and urokinase/plasminogen activator by matriptase, an epithelial membrane serine protease. J Biol Chem. 2000;275:36720–36725. doi: 10.1074/jbc.M007802200. [DOI] [PubMed] [Google Scholar]
  • 192.Hardie WD, Le Cras TD, Jiang K, Tichelaar JW, Azhar M, Korfhagen TR. Conditional expression of transforming growth factor-alpha in adult mouse lung causes pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2004;286:L741–L749. doi: 10.1152/ajplung.00208.2003. [DOI] [PubMed] [Google Scholar]
  • 193.Larios JM, Budhiraja R, Fanburg BL, Thannickal VJ. Oxidative protein cross-linking reactions involving L-tyrosine in transforming growth factor-beta1-stimulated fibroblasts. J Biol Chem. 2001;276:17437–17441. doi: 10.1074/jbc.M100426200. [DOI] [PubMed] [Google Scholar]
  • 194.Siegel PM, Massague J. Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer. Nat Rev Cancer. 2003;3:807–821. doi: 10.1038/nrc1208. [DOI] [PubMed] [Google Scholar]
  • 195.Kalluri R, Neilson EG. Epithelial–mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003;112:1776–1784. doi: 10.1172/JCI20530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Willis BC, Liebler JM, Luby-Phelps K, et al. Induction of epithelial-mesenchymal transition in alveolar epithelial cells by transforming growth factor-{beta}1: potential role in idiopathic pulmonary fibrosis. Am J Pathol. 2005;166:1321–1332. doi: 10.1016/s0002-9440(10)62351-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Zeisberg M, Bottiglio C, Kumar N, et al. Bone morphogenic protein-7 inhibits progression of chronic renal fibrosis associated with two genetic mouse models. Am J Physiol Renal Physiol. 2003;285:F1060–F1067. doi: 10.1152/ajprenal.00191.2002. [DOI] [PubMed] [Google Scholar]

RESOURCES