American Journal of Respiratory Cell and Molecular Biology

Prostaglandin (PG)E2 is a bioactive eicosanoid that regulates many biologically important processes in part due to its ability to signal through four distinct G-protein–coupled receptors with differential signaling activity and unique expression patterns in different cell types. Although PGE2 has been linked to malignancy in many organs, it is believed to play a beneficial role in the setting of fibrotic lung disease. This is in part due to the ability of PGE2 to limit many of the pathobiologic features of lung fibroblasts and myofibroblasts, including the ability of PGE2 to limit fibroblast proliferation, migration, collagen secretion, and, as originally reported in the Journal by us in 2003, the ability to limit transforming growth factor (TGF)-β–induced myofibroblast differentiation. In the setting of lung fibrosis, PGE2 production and signaling is often diminished. In the last 8 years, significant advances have been made to better understand the dysregulation of PGE2 production and signaling in the setting of lung fibrosis. We also have a clearer picture of how PGE2 inhibits myofibroblast differentiation and the receptor signaling pathways that can influence fibroblast proliferation. This review highlights these recent advances and offers new insights into the potential ways that PGE2 and its downstream signals can be regulated for therapeutic benefit in a disease that has no validated treatment options.

This review highlights the current understanding of how prostaglandin E2 can limit lung fibrosis and reviews research findings to explain why production and signaling of this eicosanoid is impaired in lung fibrosis. It also reviews how E prostanoid receptor use can influence the response of prostaglandin E2 on fibroblasts.

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive interstitial lung disease characterized by alveolar epithelial cell injury, fibroblast accumulation, and differentiation to myofibroblasts (1). The end result is contraction of alveolar architecture and the relentless deposition of extracellular matrices including collagen 1, collagen 3, and fibronectin (2, 3). The pathogenesis of the disease is poorly understood, but studies in patients with IPF have indicated that this disease is characterized by a loss in production of prostaglandins, including prostaglandin (PG)E2 (46). PGE2 signaling has many inhibitory actions on lung cells that could potentially suppress fibrogenesis, including the ability of PGE2 to limit lung myofibroblast differentiation (7). This observation, which is being highlighted in this issue, was first reported by us in 2003.

More recent work in animal models and human tissues has expanded our understanding of the regulation and role of prostaglandins in fibrotic lung disease. These advances include the identification of epigenetic changes that explain the inhibition of prostaglandin production in fibrotic lung tissue (8, 9) and studies that have elucidated PGE2 signaling defects in fibrotic fibroblasts (1012). We also have a better understanding of how epithelial–mesenchymal crosstalk is regulated by PGE2 (1315) and the signaling pathways that allow PGE2 to limit myofibroblast differentiation and migration (16, 17) and the circumstances in which PGE2 can promote fibroblast proliferation to serve a profibrotic role (1820). This review summarizes the current state of our understanding regarding the role(s) that PGE2 signaling plays in modulating lung fibrosis.

The term “pulmonary fibrosis” is used clinically to describe several forms of diffuse interstitial lung diseases classified as idiopathic interstitial pneumonias (IIP) with a common hallmark of fibrosis. Histopathological assessment can further differentiate usual interstitial pneumonia (UIP) from other forms of IIP (21). Idiopathic pulmonary fibrosis (IPF), the clinical correlate to histological UIP in the absence of an identifiable cause, is the most common and most progressive form of IIP (22), while being least responsive to therapy. Although IPF is considered rare, precise epidemiological data are lacking primarily due to methodologies that lack histological confirmation. However, one study examining narrow criteria by diagnostic coding estimated prevalence at approximately 14 cases per 100,000, with increasing prevalence associated with aging (23).

Typical symptoms include progressive breathlessness and nonproductive cough, with pulmonary function testing classically demonstrating reduced lung volumes and impaired gas exchange (21). High-resolution computer tomography typically shows lower lobe–predominant disease with septal thickening, traction bronchiectasis, and often the presence of honeycombing, which represents cystic fibrotic airspaces (24). Such findings in the proper clinical context are strongly related to the likely finding of UIP by surgical lung biopsy (24). Histologically, there is the presence of patchwork fibrosis containing significant collagen deposition and patchy inflammation in a nonuniform distribution (25). Securing a histological diagnosis of UIP requires the identification of “fibroblastic foci,” which are focal clusters of fibroblasts and myofibroblasts within young connective tissues thought to represent sites of ongoing lung injury. The presence of these fibroblastic foci is inversely correlated with disease survival and response to therapy (26). Shortened survival is associated with extent of fibroblastic foci rather than degree of cellularity or alveolar wall fibrosis, suggesting that mortality in IPF arises from abnormal fibrobroblast and myofibroblast accumulation rather than a predominantly inflammatory process (26).

IPF carries an overall poor prognosis. A histological diagnosis of UIP is the most important factor determining survival in patients with IIP (27). Median survival from time of diagnosis has been reported as 2.5 to 3.5 years (28), with most patients dying from disease progression to respiratory insufficiency. Subsequent studies have revealed that there can be a variable clinical course, ranging from subclinical and slowly progressive disease to rapid acute decompensation and death (29, 30). The search for biomarkers to predict disease progression is ongoing. Treatment options generally center around immunosuppressive regimens including prednisone and azathioprine, but there is no clear clinical benefit for these therapies in most patients and use of these drugs is being reevaluated in an ongoing NIH-sponsored clinical trial.

Fibroblasts are largely responsible for synthesizing the extracellular matrix components seen in fibrotic lesions. The matrix proteins in pulmonary fibrosis are mostly composed of collagens type 1 and 3 and fibronectin (3). Many growth and differentiation factors can affect the ability of fibroblasts to proliferate and produce matrix proteins. The best studied molecule in this regard is transforming growth factor (TGF)-β. TGF-β added to purified fibroblasts can have proliferative or differentiative effects (7, 31, 32). TGF-β induces expression of α-smooth muscle actin (α-SMA) within fibroblasts. α-SMA expression is associated with the formation of stress fibers in nonmuscle cells. This fibroblast with contractile properties is designated as a myofibroblast (33). Myofibroblasts are thought to be instrumental for the lung contraction, alveolar collapse, and matrix deposition (3, 33, 34) seen in pulmonary fibrosis. As such, strategies that limit myofibroblast differentiation may be beneficial for the treatment of lung fibrosis. As discussed below, PGE2 is a potent inhibitor of myofibroblast differentiation (7).

All cells are capable of arachidonic acid (AA) release and its metabolism to bioactive eicosanoids (35). Free AA is liberated from the sn2 position of membrane phospholipids via the actions of phospholipase A2. Once liberated, the free AA can be metabolized via either of two major pathways (36). The 5-lipoxygenase pathway gives rise to leukotrienes and is found primarily in leukocytes (37), although alveolar epithelial cells can produce low levels of cysteinyl leukotrienes (38). In contrast, the cyclooxygenase pathway yields prostanoid products, including prostaglandins, thromboxane, and prostacyclin (PGI2). This pathway is active in bone-marrow–derived cells and in structural cells (36, 39, 40). The initial step in this pathway involves the conversion of AA to PGH2 via cyclooxygenase (COX)-1 or COX-2 enzymes. Conventional dogma suggests that the COX-1 enzyme is responsible for constitutive production of PGH2, whereas the expression of the COX-2 enzyme is inducible and transient via a number of inflammatory stimuli. However, bronchial and alveolar epithelial cells express COX-2 constitutively (39, 41, 42). Formation of specific prostanoid end-products from PGH2 is mediated by cell-specific distal prostaglandin synthase enzymes (e.g., PGE synthases), which are present in constitutive and inducible isoforms (43). The COX enzymes are the targets for nonsteroidal antiinflammatory drugs, including aspirin and indomethacin (44). Biological changes that result in the shunting of AA preferentially to the lipoxygenase or the cyclooxygenase pathway can have profound effects on homeostasis and disease.

COX-2 is considered the rate-limiting enzyme for the production of prostaglandins, and numerous inflammatory and injury signals are known to up-regulate COX-2 expression, including lipopolysaccharide, IL-1β, TGF-β, hepatocyte growth factor (HGF), and plasmin (8, 13, 45). Transcriptional up-regulation of COX-2 is mediated via activation of transcription factor binding to the 5′ UTR of the COX-2 gene, which contains DNA binding sites for NFkB, AP-1, and cAMP response elements (CREs) (46). In addition, inflammatory and injury signals regulate COX-2 via regulation of histone acetylation and chromosome accessibility (8). In contrast, transcriptional silencing of the COX-2 gene is associated with hypermethylation of the CpG islands in the 5′UTR (47). Because COX-2 expression must be tightly controlled, additional posttranscriptional and posttranslational mechanisms have been described that limit COX-2 activity. One such mechanism is mRNA stability, which is influenced by the complex array of AU-rich domains within the 3′UTR of COX-2. Various transacting factors can bind to these elements to regulate stability (reviewed in Ref. 48). Additional regulatory mechanisms for COX-2 expression involve miRNA inhibition of translation via interactions with the 3′UTR of COX-2 (48), differential utilization of polyadenylation sites (48), proteosomal protein degradation pathways, and suicide inactivation of COX catalytic activity (reviewed in Ref. (49). Expression of the distal synthetic enzyme microsomal PGE2 synthase-1 has also been shown to be regulated via mRNA stability (50). Many diseases, including chronic inflammation, fever, arthritis, and certain cancers, have been linked to aberrant expression of COX-2 and enhanced prostanoid synthesis (49, 51, 52). In some cases, these pathologies are linked to the inhibitory actions of prostaglandins on innate and adaptive immune function (5355). In contrast, mounting evidence (discussed below) suggests that decreased expression of prostaglandins relative to leukotrienes is predictive of, and contributes to, fibrotic lung disease (4, 6, 8, 14, 56, 57).

There are four E prostanoid (EP) receptors, designated EP1, EP2, EP3, and EP4. In many cases, the differential effects of prostaglandins in different cell types and tissues are mediated through differential activation of 7-transmembrane–spanning EP receptors (58). The functions of the EP receptors are dictated by the intracellular signaling machinery coupled to each receptor (reviewed in Ref. 58). Stimulation of EP2 and EP4 increases cyclic adenosine monophosphate (cAMP) levels within the receptor-bearing cell and can signal smooth muscle cell relaxation. Signaling via EP1 increases intracellular Ca2+ and induces smooth muscle cell contraction. The EP3 receptor decreases cAMP and inhibits smooth muscle relaxation (5861). Regarding lung fibroblasts, the antifibrotic actions of PGE2 have been shown to be mediated via stimulation of EP2 and EP4, resulting in the activation of cAMP, and via downstream effects on protein kinase (PK)A or exchange protein activated by cAMP (7, 11, 62). Inhibition of fibroblast collagen expression involves activation of PKA (62, 63), whereas inhibition of proliferation requires exchange protein activated by cAMP (62).

Similar to COX-2, expression of the EP receptors, especially EP2, is also tightly controlled. EP2 is frequently silenced in neuroblastoma cell lines, and the DNA methylation pattern in a portion of the CpG islands is correlated inversely with EP2 expression (64). Additionally, aberrant methylation of EP2 is noted more frequently in advanced neuroblastoma cancers (64). Similarly, a loss of EP2 expression is noted in many non–small cell lung carcinomas, and expression can be restored after treatment with a demethylating agent (65). Because PGE2 can promote tumorigenesis in lung cancer, aberrant methylation, which limited EP2 expression, was correlated with better patient outcome. As discussed below, a similar epigenetic reduction of EP2 expression in patients with lung fibrosis (where PGE2 signaling is beneficial) can have the opposite effect and worsen patient outcomes.

Homeostasis within the healthy lung microenvironment is dependent on alveolar epithelial cell (AEC)–mesenchymal cell crosstalk (2, 66). The evidence for this concept is: (1) in normal lung sections, foot processes from AECs can be seen extending through the basement membrane and making contact with the mesenchymal cells; (2) in vitro, AECs from normal animals inhibit fibroblast proliferation; and (3) fibrotic injury results in loss or damage to AECs. The ability of epithelial cells to limit fibroblast proliferation is critically dependent on the ability of the AECs to produce prostaglandins (41). The actions of prostaglandins (specifically PGE2 and PGI2) inhibit fibroblast function. Specifically, prostaglandins decrease fibroblast chemotaxis (67), decrease fibroblast proliferation (11, 62, 63, 6870), decrease fibroblast growth factor receptor expression (71), decrease fibroblast collagen synthesis (63, 72, 73), inhibit myofibroblast differentiation (7, 16), and increase collagen degradation (74). The pathways by which PGE2 limits myofibroblast differentiation are discussed in more detail below.

Prostaglandins also play important roles in the regulation of apoptosis within the lung, and the fact that patients with IPF exhibit increased apoptosis of AECs but diminished apoptosis of fibroblasts has been termed the “apoptosis paradox.” Recent studies have demonstrated that reduced expression of COX-2 and PGE2 in fibroblasts from patients with IPF is one factor that promotes fibroblast survival in the fibrotic lung (14). In normal human lung fibroblasts, PGE2 increases apoptosis and potentiates apoptotic signals delivered by Fas ligand. The ability of PGE2 to promote normal fibroblast apoptosis requires signaling via EP2/EP4 and a reduction in activity of the prosurvival molecule protein kinase B (Akt) (75). In contrast, fibroblasts from patients with IPF are resistant to the proapoptotic effects of PGE2 (75). Figure 1 diagrams the homeostatic and antifibrotic actions of PGE2 signaling in lung epithelial cells and fibroblasts.

In response to TGF-β, fibroblasts undergo Smad-dependent signaling and develop characteristic changes known as stress fibers caused by the reorganization of α-SMA into filamentous bundles (7, 16, 76, 77). In addition, TGF-β signaling induces the reorganization of the actin cytoskeleton and the formation of adhesive signaling complexes known as focal adhesions. Focal adhesions are populated by focal adhesion kinase, F-actin, paxillin, vinculin, and αvβ3 integrins (78, 79). The formation of these adhesive contacts is critical for myofibroblast differentiation. When cells are treated with TGF-β in suspension culture, no myofibroblast differentiation occurs (77). The formation of focal adhesions involves activation of one or more members of the small Ras GTPase family (Rho A, Rac, or CDC42) (80, 81).

Our original study published in 2003 demonstrated that PGE2 signaling led to increased cAMP via the EP2 receptor, which, in turn, could inhibit TGF-β–induced myofibroblast differentiation and limit collagen secretion (7). Our study showed that PGE2 did not function by interfering with TGF-β–induced Smad phosphorylation or translocation to the nucleus. Rather, PGE2 altered cytoskeletal architecture and disrupted the formation of focal adhesions (16) (Figure 2). Additionally, PGE2 signaling through EP2 activates phosphatase and tensin homolog deleted on chromosome 10 (PTEN), and this results in diminished fibroblast proliferation (82). TGF-β is only able to induce myofibroblast differentiation in the absence of PTEN (76); thus, PGE2 activation of PTEN is another mechanism by which this prostanoid limits myofibroblast differentiation. Strategies that amplify the inhibitory cAMP signals generated by PGE2 are also beneficial in limiting myofibroblast differentiation. Phosphodiesterase 4 (PDE4) inhibitors can prevent the breakdown of cAMP, and, as such, potentiate the effects of PGE2 in limiting myofibroblast differentiation (83, 84) and can limit collagen gel contraction and chemotaxis (85). More recently, knockdown of PDE4B and PDE4D subtypes were shown to limit TGF-β–induced myofibroblast differentiation on their own (86). In summary, these data suggest that PGE2 signaling should be beneficial in limiting fibrosis; thus, investigators were eager to understand why production of PGE2 was diminished in IPF lungs (46). Furthermore, the observation that fibroblasts from fibrotic murine and human lungs were often refractory to the inhibitory effects of PGE2 (1012) stimulated additional studies to understand this disease-related heterogeneity.

Derangement of eicosanoid synthesis can be seen in human and animal lung fibrosis studies. Leukotriene levels have been reported to be greater in bronchoalveolar lavage fluid and lung homogenates from patients with IPF than from healthy volunteers (87, 88). Alveolar macrophages are the main source of leukotriene synthesis and are responsible for the increased production of leukotriene C4 and leukotriene B4 noted in IPF lung homogenates (57). Animal models are also characterized by increased leukotriene production after lung injury. Mice genetically deficient in leukotriene production (5-lipoxygenase knock-out mice) are protected from bleomycin-induced pulmonary fibrosis (89). Because cysteinyl leukotrienes are known to induce proliferation and collagen synthesis in mesenchymal cells (68, 90, 91), increases in metabolism of AA via the 5-lipoxygenase pathway can enhance fibrogenesis.

Conversely, reduced PGE2 levels have been reported in bronchoalveolar lavage fluid and alveolar macrophage–conditioned media from patients with IPF (4, 56). These observations are consistent with findings of reduced COX-2 expression in patients with IPF (8, 14, 92). Fibroblasts from patients with IPF are unable to up-regulate the COX-2 enzyme in response to stimuli and as such are deficient in PGE2 production (5, 6, 93). Reduced PGE2 synthesis has also been reported for fibroblasts isolated from rat lungs after bleomycin-induced pulmonary fibrosis (94). Animal models characterized by reduced PGE2 synthesis in the lung via administration of indomethacin (95) or the gene-deletion of COX-2 (96) manifest worse bleomycin-induced fibrosis. Thus, in the injured lung, the functional loss of prostaglandins has severe consequences for fibroproliferation.

The mechanisms responsible for PGE2 loss in the fibrotic lung are varied and include effects of soluble mediators and epigenetics. Profibrotic injury to epithelial cells often results in the release of chemokines, including CCL2 (97). In the presence of CCL2, PGE2 production by lung AECs is diminished, and fibroproliferation increases (15). This is one mechanism that explains why mice defective in CCR2 (the receptor for CCL2) are protected from fibrotic injury (97, 98). Another well known consequence of fibrotic injury is the leakage of plasma from damaged vasculature and the activation of the coagulation cascade. However, in the fibrotic lung, the extravascular fibrin is not effectively cleared, and epithelial repair is blocked due to a marked increase in the expression of plasminogen activation inhibitor (PAI)-1 relative to urokinase-type plasminogen activator (uPA) (99101). PAI-1 blocks uPA and thus prevents the generation of plasmin and inhibits the proteolytic activation and release of the epithelial repair molecule HGF (102, 103). We recently showed that plasmin up-regulates COX-2 and stimulates PGE2 production in AECs, fibroblasts, and fibrocytes via HGF (13). Thus, one consequence of the increased PAI-1 levels noted in fibrotic lungs is diminished production of PGE2. Because PGE2 has also been shown to promote HGF activation (104), this positive antifibrotic feedback loop is likely missing in IPF.

More recently, epigenetic changes have been identified that contribute to diminished expression of COX-2 and thus PGE2 in fibrotic lungs. Using fibroblasts isolated from normal or fibrotic lungs, Coward and colleagues demonstrated that COX-2 mRNA levels were reduced in fibrotic fibroblasts, but mRNA degradation rates were similar (8). Further investigation demonstrated that although some fibroblasts from IPF lungs contained appropriate levels of transcription factors that drive COX-2 expression (including NFkB p65, CEBPb, and CREB-1), these factors were prevented from binding to the COX-2 promoter in the native chromatin configuration. This inhibition of transcription factor binding was correlated with defective histone H3 and H4 acetylation due to reduced recruitment of histone acetyltransferases and increased recruitment of histone deactylease containing co-repressor complexes (8). In other fibrotic fibroblasts, the phosphorylation of CREB (a downstream effector of cAMP activation of PKA) was shown to be defective (105). Taken together, these epigenetic changes noted in fibroblasts from patients with IPF prevent COX-2 gene transcription in IPF and contribute to diminished production of PGE2. Figure 3 highlights alterations in the PGE2 production and signaling pathways that have been noted during lung fibrosis.

Not only is PGE2 production altered in the fibrotic lung, but PGE2 signaling can also be impaired. Transcriptional or epigenetic decreases in EP2 or EP4 expression can limit the inhibitory signaling of PGE2 in fibroblasts. We have previously shown that EP2 levels are diminished in fibroblasts isolated from mice on Day 14 after bleomycin or fluorescein isothiocyanate treatment (11). This loss of EP2 expression resulted in blunted cAMP responses and a reduced ability of PGE2 to inhibit proliferation and collagen secretion in these cells. Fibroblasts from patients with IPF have also been shown to be refractory to PGE2 signaling and identified mechanisms include decreased expression of EP2 as well as diminished expression of PKA (a downstream target of cAMP and EP2 signaling) (10). More recently, hypermethylation of the PGE receptor 2 gene (PTGER2) promoter has been identified as a mechanism for reduced EP2 expression in fibroblasts from patients with IPF and fibrotic mice (9). An increase in Akt signal transduction is believed to be one mechanism that drives the hypermethylation status of the PTGER2 promoter (9). Additionally, action of the enzyme PTEN is known to up-regulate EP2 expression (12) and IPF fibroblasts; particularly those in the fibrotic foci are known to be PTEN deficient (76). Each of these mechanisms would result in diminished signaling via the inhibitory EP2 receptor, and, as such, these findings explain in part why fibroblasts from patients with IPF are largely refractory to PGE2 inhibitory signaling. Understanding the differences that exist in the cell types that comprise the normal and fibrotic lung is important for the design of future therapeutics.

Although we have detailed the antifibrotic actions of PGE2 signaling via EP2 and EP4 and noted that lung fibrosis is often associated with reduced production of PGE2 or defective EP2 signaling, in some instances, PGE2 can promote fibroblast proliferation. When this occurs, it is via EP1 or EP3 signaling. Studies looking at the fibroproliferative response after acute lung injury (ALI) identified a dose-dependent effect of PGE2 on fibroblast proliferation. At midrange concentrations (10−9 to 10−7 M), PGE2 enhanced proliferation of lung fibroblasts via EP3 stimulation, whereas at extremely low (< 10−10 M) or high concentrations (> 10−6 M), PGE2 suppressed lung fibroblast proliferation via EP2 stimulation (20). This study found that the range of PGE2 concentrations noted in edema fluid from patients with ALI were in the concentration range to stimulate fibroblast proliferation, thus implicating EP3 stimulation in the fibroproliferative consequences of ALI (20). EP1 stimulation has also been reported to promote fibroblast calcium mobilization and increased proliferation of NIH 3T3 fibroblasts (19). Additionally, in recent studies using neonatal rat ventricular fibroblasts that expressed all four EP receptor subtypes, PGE2 stimulation increased the number of cells in S phase and increased expression of cyclin D3. These same effects were mimicked with the EP1/EP3 agonist sulprostone (18) and implicate EP1/EP3 stimulation in cardiac fibrosis. Thus, PGE2 is capable of inhibiting fibroproliferation via EP2/EP4 or promoting proliferation via EP1/EP3 stimulation depending on the particular context of the fibroblasts.

Homeostatic balance within the lung requires appropriate crosstalk between alveolar epithelial cells, fibroblasts, and inflammatory cells (see Figure 1). Production of PGE2 by alveolar epithelial cells is believed to be an important factor for limiting fibroproliferation and promoting appropriate alveolar epithelial repair. In the normal lung, PGE2 signaling via EP2 receptor–mediated elevations in cAMP can induce fibroblast apoptosis and, as we showed, limit myofibroblast transformation, proliferation, and collagen secretion. However, in the fibrotic lung, various perturbations alter the homeostatic balance. PGE2 production is limited via inflammatory mediators and epigenetic silencing of the COX-2 promoter. Furthermore, fibrotic fibroblasts lose EP2 receptor expression and may lose expression of the downstream effectors PKA and phospho-CREB. It is also interesting that fibrosis is a male-predominant disease, and at least one study has suggested that male gender is associated with reduced EP2 and EP4 levels and reduced PGE2 production in splenic macrophages after trauma (106). If these gender differences are true in response to lung injury as well, it may in part explain the gender differences that are noted in IPF.

Future Directions and Clinical Implications

It is clear that PGE2 production plays an important role in determining homeostasis in the normal lung. As such, it is tempting to suggest that strategies aimed at delivering PGE2 to the lung might be beneficial in treating patients with IPF. However, there are several caveats to this type of therapy. First, the therapy would only be effective in patients in whom EP2 signaling was intact. Determining this may require surgical or transbronchial biopsies, and PGE2 responsiveness in isolated fibroblasts must be determined on an individual patient basis. Second, the half-life of PGE2 is quite short, so new innovations would be needed to deliver this lipid as a therapeutic. One possibility includes creating derivatives with similar EP2 binding and signaling capacities. Alternatively, EP2 agonists may be more stable. Another important aspect to consider is that PGE2 has differential effects depending on the cell type and the EP receptor profile it encounters. Thus, intravenous administration may not be effective due to deleterious effects on the vascular system. It is also possible that systemic administration of PGE2 could promote diseases that are often associated with pathological overexpression of PGE2, such as colon cancer, persistent inflammation, and arthritis (49, 51, 52). This means that inhalation therapy may be the best option for patients with lung fibrosis, but at present nebulization of PGE2 or EP2 agonists is not practical. Alternative therapeutic strategies could be to prevent the breakdown of cAMP to maximize its inhibitory signaling capacity in fibrotic lungs. This may be achieved by administration of PDE4 inhibitors. Again, this therapy would only be effective in patients in whom modest PGE2 production and cAMP generation was intact in the lung, and this type of therapy may predispose to other diseases and malignancy. Methylation of the promoters for COX-2 and EP2 has been demonstrated to limit PGE2 expression in some patients. Thus, it is possible that inhibitors of methyl transferases may be effective in reversing the methylation status of these genes and increasing PGE2 production and signaling. The potential down sides to this therapy involve the off-target effects that these agents could have on other genes. Global demethylation may not be beneficial.

It may be possible to target signals downstream of EP2, such as cAMP elevation or PKA activation. Particularly if these therapies could be delivered in a cell-specific manner, they may be effective in inhibiting myofibroblast differentiation and activation even in patients with EP2 receptor defects. This targeted therapy may also avoid the potential proliferative effects of PGE2 binding to EP1 or EP3 receptors in some organs. Finally, it is possible that cell-directed therapies could be evolved to treat patients with IPF. Fibrotic regions of the lung may be quite difficult to treat by inhalational therapies due to limited airflow in these regions. It is possible that cells such as mesenchymal stem cells or potentially fibrocytes could be engineered to deliver abundant PGE2, EP2 agonists, or adenyl cyclase stimulators. In this way, the therapeutic cell type may be able to use chemokine receptors and inflammatory signals to colocalize to areas of fibrosis and as such may offer a more targeted delivery of an antifibrotic therapy.

The authors thank Peedikayil Thomas for help with the confocal microscopy in Figure 2.

1. Kuhn C. Pathology. In: , Phan S, Thrall R, editors. Pulmonary fibrosis. New York: Marcel Dekker, Inc.; 1995. pp. 5983.
2. Selman M, King TE, Pardo A. Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med 2001;134:136151.
3. Kuhn C, Boldt J, King TE, Crouch E, Vartio T, McDonald J. An immunohistochemical study of architectural remodeling and connective tissue synthesis in pulmonary fibrosis. Am Rev Respir Dis 1989;140:16931703.
4. Borok Z, Gillissen A, Buhl R, Hoyt RF, Hubbard RC, Ozaki T, Rennard SI, Crystal RG. Augmentation of functional prostaglandin E levels on the respiratory epithelial surface by aerosol administration of prostaglandin E. Am Rev Respir Dis 1991;144:10801084.
5. Vancheri C, Sortino MA, Tomaselli V, Mastruzzo C, Condorelli F, Bellistri G, Pistorio MP, Canonico PL, Crimi N. Different expression of TNF-alpha receptors and prostaglandin E(2) production in normal and fibrotic lung fibroblasts: potential implications for the evolution of the inflammatory process. Am J Respir Cell Mol Biol 2000;22:628634.
6. Wilborn J, Crofford LJ, Burdick MD, Kunkel SL, Strieter RM, Peters-Golden M. Cultured lung fibroblasts isolated from patients with idiopathic pulmonary fibrosis have a diminished capacity to synthesize prostaglandin E2 and to express cyclooxygenase-2. J Clin Invest 1995;95:18611868.
7. 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;29:537544.
8. Coward WR, Watts K, Feghali-Bostwick CA, Knox A, Pang L. Defective histone acetylation is responsible for the diminished expression of cyclooxygenase 2 in idiopathic pulmonary fibrosis. Mol Cell Biol 2009;29:43254339.
9. Huang SK, Fisher AS, Scruggs AM, White ES, Hogaboam CM, Richardson BC, Peters-Golden M. Hypermethylation of PTGER2 confers prostaglandin E2 resistance in fibrotic fibroblasts from humans and mice. Am J Pathol 2010;177:22452255.
10. Huang SK, Wettlaufer SH, Hogaboam CM, Flaherty KR, Martinez FJ, Myers JL, Colby TV, Travis WD, Toews GB, Peters-Golden M. Variable prostaglandin E2 resistance in fibroblasts from patients with usual interstitial pneumonia. Am J Respir Crit Care Med 2008;177:6674.
11. Moore BB, Ballinger MN, White ES, Green ME, Herrygers AB, Wilke CA, Toews GB, Peters-Golden M. Bleomycin-induced E prostanoid receptor changes alter fibroblast responses to prostaglandin E2. J Immunol 2005;174:56445649.
12. Sagana RL, Yan M, Cornett AM, Tsui JL, Stephenson DA, Huang SK, Moore BB, Ballinger MN, Melonakos J, Kontos CD, et al.. Phosphatase and tensin homologue on chromosome 10 (PTEN) directs prostaglandin E2-mediated fibroblast responses via regulation of E prostanoid 2 receptor expression. J Biol Chem 2009;284:3226432271.
13. Bauman KA, Wettlaufer SH, Okunishi K, Vannella KM, Stoolman JS, Huang SK, Courey AJ, White ES, Hogaboam CM, Simon RH, et al.. The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice. J Clin Invest 2010;120:19501960.
14. Maher TM, Evans IC, Bottoms SE, Mercer PF, Thorley AJ, Nicholson AG, Laurent GJ, Tetley TD, Chambers RC, McAnulty RJ. Diminished prostaglandin E2 contributes to the apoptosis paradox in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2010;182:7382.
15. Moore BB, Peters-Golden M, Christensen PJ, Lama V, Kuziel WA, Paine R. 3rd, Toews GB. Alveolar epithelial cell inhibition of fibroblast proliferation is regulated by MCP-1/CCR2 and mediated by PGE2. Am J Physiol Lung Cell Mol Physiol 2003;284:L342349.
16. Thomas PE, Peters-Golden M, White ES, Thannickal VJ, Moore BB. PGE(2) inhibition of TGF-beta1-induced myofibroblast differentiation is Smad-independent but involves cell shape and adhesion-dependent signaling. Am J Physiol Lung Cell Mol Physiol 2007;293:L417L428.
17. White ES, Atrasz RG, Dickie EG, Aronoff DM, Stambolic V, Mak TW, Moore BB, Peters-Golden M. Prostaglandin E(2) inhibits fibroblast migration by E-prostanoid 2 receptor-mediated increase in PTEN activity. Am J Respir Cell Mol Biol 2005;32:135141.
18. Harding P, Lapointe MC. Prostaglandin E2 increases cardiac fibroblast proliferation and increases cyclin D expression via EP1 receptor. Prostaglandins Leukot Essent Fatty Acids 2011;84:147152.
19. Watanabe T, Satoh H, Togoh M, Taniguchi S, Hashimoto Y, Kurokawa K. Positive and negative regulation of cell proliferation through prostaglandin receptors in NIH-3T3 cells. J Cell Physiol 1996;169:401409.
20. White KE, Ding Q, Moore BB, Peters-Golden M, Ware LB, Matthay MA, Olman MA. Prostaglandin E2 mediates IL-1beta-related fibroblast mitogenic effects in acute lung injury through differential utilization of prostanoid receptors. J Immunol 2008;180:637646.
21. American Thoracic Society/European Respiratory Society International Multidisciplinary Consensus Classification of the Idiopathic Interstitial Pneumonias. Am J Respir Crit Care Med 2002;165:277304.
22. Chapman J, Farver C. Idiopathic interstitial lung disease: a review of recent classifications. Clin Pulm Med 2004;11:1724.
23. Raghu G, Weycker D, Edelsberg J, Bradford WZ, Oster G. Incidence and prevalence of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2006;174:810816.
24. Schmidt SL, Sundaram B, Flaherty KR. Diagnosing fibrotic lung disease: when is high-resolution computed tomography sufficient to make a diagnosis of idiopathic pulmonary fibrosis?. Respirology 2009;14:934939.
25. Katzenstein AL, Myers JL. Idiopathic pulmonary fibrosis: clinical relevance of pathologic classification. Am J Respir Crit Care Med 1998;157:13011315.
26. King TE, Schwarz MI, Brown K, Tooze JA, Colby TV, Waldron JA, Flint A, Thurlbeck W, Cherniack RM. Idiopathic pulmonary fibrosis: relationship between histopathologic features and mortality. Am J Respir Crit Care Med 2001;164:10251032.
27. Flaherty KR, Toews GB, Travis WD, Colby TV, Kazerooni EA, Gross BH, Jain A, Strawderman RL, Paine R, Flint A, et al.. Clinical significance of histological classification of idiopathic interstitial pneumonia. Eur Respir J 2002;19:275283.
28. American Thoracic Society. 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 2000;161:646664.
29. Collard HR, Moore BB, Flaherty KR, Brown KK, Kaner RJ, King TE, Lasky JA, Loyd JE, Noth I, Olman MA, et al.. Acute exacerbations of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2007;176:636643.
30. Fernandez Perez ER, Daniels CE, Schroeder DR, St Sauver J, Hartman TE, Bartholmai BJ, Yi ES, Ryu JH. Incidence, prevalence, and clinical course of idiopathic pulmonary fibrosis: a population-based study. Chest 2009;137:129137.
31. Shukla A, Meisler N, Cutroneo KR. Perspective article: transforming growth factor-beta: crossroad of glucocorticoid and bleomycin regulation of collagen synthesis in lung fibroblasts. Wound Repair Regen 1999;7:133140.
32. Zhang K, Phan SH. Cytokines and pulmonary fibrosis. Biol Signals 1996;5:232239.
33. Gabbiani G. The myofibroblast: a key cell for wound healing and fibrocontractive diseases. Prog Clin Biol Res 1981;54:183194.
34. Kuhn C, McDonald J. The roles of the myofibroblast in idiopathic pulmonary fibrosis: ultrastructural and immunohistochemical features of sites of active extracellular matrix synthesis. Am J Pathol 1991;138:12571265.
35. Seeds M, Bass D. Regulation and metabolism of arachidonic acid. Clin Rev Allergy Immunol 1999;17:526.
36. Needleman P, Turk J, Jakschik B, Morrison A, Lefkowith J. Arachidonic acid metabolism. Annu Rev Biochem 1986;55:69102.
37. Ford-Hutchinson A, Gresser M, Young R. 5-Lipoxygenase. Annu Rev Biochem 1994;63:383417.
38. Vannella KM, Luckhardt TR, Wilke CA, van Dyk LF, Toews GB, Moore BB. Latent herpesvirus infection augments experimental pulmonary fibrosis. Am J Respir Crit Care Med 2009;181:465477.
39. Ermert L, Ermert M, Goppelt-Struebe M, Walmrath D, Grimminger F, Steudel W, Ghofrani HA, Homberger C, Duncker H, Seeger W. Cyclooxygenase isoenzyme localization and mRNA expression in rat lungs. Am J Respir Cell Mol Biol 1998;18:479488.
40. Simon LS. Role and regulation of cyclooxygenase-2 during inflammation. Am J Med 1999;106:37S42S.
41. Lama V, Moore B, Christensen P, Toews G, Peters-Golden M. Prostaglandin E2 synthesis and suppression of fibroblast proliferation by alveolar epithelial cells is cyclooxygenase-2 dependent. Am J Respir Cell Mol Biol 2002;27:752758.
42. Walenga R, Kester M, Coroneos E, Butcher S, Dwivdi R, Statt C. Constitutive expression of prostaglandin endoperoxide G/H synthetase (PGHS)-2 but not PGHS-1 in human tracheal epithelial cells in vitro. Prostaglandins 1996;52:341359.
43. Hara S, Kamei D, Sasaki Y, Tanemoto A, Nakatani Y, Murakami M. Prostaglandin E synthases: understanding their pathophysiological roles through mouse genetic models. Biochimie 2010;92:651659.
44. Rainsford KD. Anti-inflammatory drugs in the 21st century. Subcell Biochem 2007;42:327.
45. Brock TG, McNish RW, Mancuso P, Coffey MJ, Peters-Golden M. Prolonged lipopolysaccharide inhibits leukotriene synthesis in peritoneal macrophages: mediation by nitric oxide and prostaglandins. Prostaglandins Other Lipid Mediat 2003;71:131145.
46. Appleby SB, Ristimaki A, Neilson K, Narko K, Hla T. Structure of the human cyclo-oxygenase-2 gene. Biochem J 1994;302:723727.
47. Song SH, Jong HS, Choi HH, Inoue H, Tanabe T, Kim NK, Bang YJ. Transcriptional silencing of cyclooxygenase-2 by hyper-methylation of the 5′ CpG island in human gastric carcinoma cells. Cancer Res 2001;61:46284635.
48. Harper KA, Tyson-Capper AJ. Complexity of COX-2 gene regulation. Biochem Soc Trans 2008;36:543545.
49. Mbonye UR, Song I. Posttranscriptional and posttranslational determinants of cyclooxygenase expression. BMB Rep 2009;42:552560.
50. Degousee N, Angoulvant D, Fazel S, Stefanski E, Saha S, Iliescu K, Lindsay TF, Fish JE, Marsden PA, Li RK, et al.. c-Jun N-terminal kinase-mediated stabilization of microsomal prostaglandin E2 synthase-1 mRNA regulates delayed microsomal prostaglandin E2 synthase-1 expression and prostaglandin E2 biosynthesis by cardiomyocytes. J Biol Chem 2006;281:1644316452.
51. Ghosh N, Chaki R, Mandal V, Mandal SC. COX-2 as a target for cancer chemotherapy. Pharmacol Rep 2010;62:233244.
52. Hinz B, Brune K. Pain and osteoarthritis: new drugs and mechanisms. Curr Opin Rheumatol 2004;16:628633.
53. Sakata D, Yao C, Narumiya S. Emerging roles of prostanoids in T cell-mediated immunity. IUBMB Life 2010;62:591596.
54. Rocca B, FitzGerald GA. Cyclooxygenases and prostaglandins: shaping up the immune response. Int Immunopharmacol 2002;2:603630.
55. Serezani CH, Ballinger MN, Aronoff DM, Peters-Golden M. Cyclic AMP: master regulator of innate immune cell function. Am J Respir Cell Mol Biol 2008;39:127132.
56. Ozaki T, Moriguchi H, Nakamura Y, Kamei T, Yasuoka S, Ogura T. Regulatory effects of prostaglandin E2 on fibronectin release from human macrophages. Am Rev Respir Dis 1990;141:965969.
57. 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;97:18271836.
58. Narumiya S, Sugimoto Y, Ushikubi F. Prostanoid receptors: structures, properties, and functions. Physiol Rev 1999;79:11931226.
59. Honda A, Sugimoto Y, Namba T, Watabe A, Irie A, Negishi M, Narumiya S, Ichikawa A. Cloning and expression of a cDNA for mouse prostaglandin E receptor EP2 subtype. J Biol Chem 1993;268:77597762.
60. Sugimoto Y, Namba T, Honda A, Hayashi Y, Negishi M, Ichikawa A, Narumiya S. Cloning and expression of a cDNA for mouse prostaglandin E receptor EP3 subtype. J Biol Chem 1992;267:64636466.
61. Watabe A, Sugimoto Y, Honda A, Irie A, Namba T, Negishi M, Ito S, Narumiya S, Ichikawa A. Cloning and expression of cDNA for a mouse EP1 subtype of prostaglandin E receptor. J Biol Chem 1993;268:2017520178.
62. Huang SK, Wettlaufer SH, Chung J. Peters-Golden M. Prostaglandin E2 inhibits specific lung fibroblast functions via selective actions of PKA and Epac-1. Am J Respir Cell Mol Biol 2008;39:482489.
63. Huang SK, Wettlaufer SH, Hogaboam CM, Aronoff DM. Peters-Golden M. Prostaglandin E2 inhibits collagen expression and proliferation in patient-derived normal lung fibroblasts via E prostanoid 2 receptor and cAMP signaling. Am J Physiol Lung Cell Mol Physiol 2007;292:L405L413.
64. Sugino Y, Misawa A, Inoue J, Kitagawa M, Hosoi H, Sugimoto T, Imoto I, Inazawa J. Epigenetic silencing of prostaglandin E receptor 2 (PTGER2) is associated with progression of neuroblastomas. Oncogene 2007;26:74017413.
65. Tian L, Suzuki M, Nakajima T, Kubo R, Sekine Y, Shibuya K, Hiroshima K, Nakatani Y, Fujisawa T, Yoshino I. Clinical significance of aberrant methylation of prostaglandin E receptor 2 (PTGER2) in nonsmall cell lung cancer: association with prognosis, PTGER2 expression, and epidermal growth factor receptor mutation. Cancer 2008;113:13961403.
66. Selman M, Pardo A. Role of epithelial cells in idiopathic pulmonary fibrosis: from innocent targets to serial killers. Proc Am Thorac Soc 2006;3:364372.
67. Kohyama T, Ertl RF, Valenti V, Spurzem J, Kawamoto M, Nakamura Y, Veys T, Allegra L, Romberger D, Rennard SI. Prostaglandin E(2) inhibits fibroblast chemotaxis. Am J Physiol Lung Cell Mol Physiol 2001;281:L1257L1263.
68. Baud L, Perez J, Denis M, Ardaillou R. Modulation of fibroblast proliferation by sulfidopeptide leukotrienes: effect of indomethacin. J Immunol 1987;138:11901195.
69. Bitterman P, Wewers N, Rennard S, Adelberg S, Cyrstal R. Modulation of alveolar macrophage-driven fibroblast proliferation by alternative macrophage mediators. J Clin Invest 1986;77:700708.
70. Elias J, Rossman M, Zurier R, Daniele R. Human alveolar macrophage inhibition of lung fibroblast growth: a prostaglandin-dependent process. Am Rev Respir Dis 1985;131:9499.
71. Boyle JE, Lindroos PM, Rice AB, Zhang L, Zeldin DC, Bonner JC. Prostaglandin-E2 counteracts interleukin-1beta-stimulated upregulation of platelet-derived growth factor alpha-receptor on rat pulmonary myofibroblasts. Am J Respir Cell Mol Biol 1999;20:433440.
72. Goldstein R, Polgar P. The effect and interaction of bradykinan and prostaglandins on protein and collagen production by lung fibroblasts. J Biol Chem 1982;257:86308633.
73. Korn J, Halushka P, Leroy E. Mononuclear cell modulation of connective tissue function: suppression of fibroblast growth by stimulation of endogenous prostaglandin production. J Clin Invest 1980;65:543554.
74. Baum BJ, Moss J, Breul SD, Berg RA, Crystal RG. Effect of cyclic AMP on the intracellular degradation of newly synthesized collagen. J Biol Chem 1980;255:28432847.
75. Huang SK, White ES, Wettlaufer SH, Grifka H, Hogaboam CM, Thannickal VJ, Horowitz JC, Peters-Golden M. Prostaglandin E(2) induces fibroblast apoptosis by modulating multiple survival pathways. FASEB J 2009;23:43174326.
76. White ES, Atrasz RG, Hu B, Phan SH, Stambolic V, Mak TW, Hogaboam CM, Flaherty KR, Martinez FJ, Kontos CD, et al.. Negative regulation of myofibroblast differentiation by PTEN (Phosphatase and Tensin Homolog Deleted on chromosome 10). Am J Respir Crit Care Med 2006;173:112121.
77. Thannickal VJ, Lee DY, White ES, Cui Z, Larios JM, Chacon R, Horowitz JC, Day RM, Thomas PE. Myofibroblast differentiation by transforming growth factor-beta1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J Biol Chem 2003;278:1238412389.
78. Katz BZ, Zamir E, Bershadsky A, Kam Z, Yamada KM, Geiger B. Physical state of the extracellular matrix regulates the structure and molecular composition of cell-matrix adhesions. Mol Biol Cell 2000;11:10471060.
79. Zamir E, Katz M, Posen Y, Erez N, Yamada KM, Katz BZ, Lin S, Lin DC, Bershadsky A, Kam Z, et al.. Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts. Nat Cell Biol 2000;2:191196.
80. Hall A. Rho GTPases and the actin cytoskeleton. Science 1998;279:509514.
81. Ridley AJ, Hall A. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell 1992;70:389399.
82. White ES, Atrasz RG, Dickie EG, Aronoff DM, Stambolic V, Mak TW, Moore BB, Peters-Golden M. Prostaglandin E(2) inhibits fibroblast migration by E-prostanoid 2 receptor-mediated increase in PTEN activity. Am J Respir Cell Mol Biol 2005;32:135141.
83. Dunkern TR, Feurstein D, Rossi GA, Sabatini F, Hatzelmann A. Inhibition of TGF-beta induced lung fibroblast to myofibroblast conversion by phosphodiesterase inhibiting drugs and activators of soluble guanylyl cyclase. Eur J Pharmacol 2007;572:1222.
84. Togo S, Liu X, Wang X, Sugiura H, Kamio K, Kawasaki S, Kobayashi T, Ertl RF, Ahn Y, Holz O, et al.. PDE4 inhibitors roflumilast and rolipram augment PGE2 inhibition of TGF-{beta}1-stimulated fibroblasts. Am J Physiol Lung Cell Mol Physiol 2009;296:L959L969.
85. Kohyama T, Liu X, Wen FQ, Zhu YK, Wang H, Kim HJ, Takizawa H, Cieslinski LB, Barnette MS, Rennard SI. PDE4 inhibitors attenuate fibroblast chemotaxis and contraction of native collagen gels. Am J Respir Cell Mol Biol 2002;26:694701.
86. Selige J, Hatzelmann A, Dunkern T. The differential impact of PDE4 subtypes in human lung fibroblasts on cytokine-induced proliferation and myofibroblast conversion. J Cell Physiol ( In press )
87. Ozaki O, Hayashi H, Tani K, Ogushi F, Yasuoka U, Ogura T. Neutrophil chemotactic factor in the respiratory tract of patients with chronic airway diseases or idiopathic pulmonary fibrosis. Am Rev Respir Dis 1992;145:8591.
88. Wardlaw A, Hay H, Cromwell O, Collins J, Kay A. Leukotrienes, LTC4 and LTB4, in bronchoalveolar lavage in bronchial asthma and other respiratory diseases. J Allergy Clin Immunol 1989;84:1926.
89. Peters-Golden M, Bailie M, Marshall T, Wilke C, Phan SH, Toews GB, Moore BB. Protection from pulmonary fibrosis in leukotriene-deficient mice. Am J Respir Crit Care Med 2002;165:229235.
90. Phan SH, McGarry B, Loeffler K, Kunkel S. Binding of leukotriene C4 to rat lung fibroblasts and stimulation of collagen synthesis in vitro. Biochemistry 1988;27:28462853.
91. Vannella KM, McMillan TR, Charbeneau RP, Wilke CA, Thomas PE, Toews GB, Peters-Golden M, Moore BB. Cysteinyl leukotrienes are autocrine and paracrine regulators of fibrocyte function. J Immunol 2007;179:78837890.
92. Petkova DK, Clelland CA, Ronan JE, Lewis S, Knox AJ. Reduced expression of cyclooxygenase (COX) in idiopathic pulmonary fibrosis and sarcoidosis. Histopathology 2003;43:381386.
93. McAnulty RJ, Hernandez-Rodriguez NA, Mutsaers S, Coker R, Laurent G. Indomethacin suppresses the anti-proliferative effects of transforming growth factor-beta isoforms on fibroblast cell cultures. Biochem J 1997;321:639643.
94. Ogushi F, Endo T, Tani K, Asada K, Kawano T, Maniwa HTK, Sone S. Decreased prostaglandin E2 synthesis by lung fibroblasts isolated from rats with bleomycin-induced lung fibrosis. Int J Exp Pathol 1999;80:4149.
95. Moore B, Coffey M, Christensen P, Sitterding S, Ngan R, McDonald R, Phare S, Peters-Golden M, Paine R. III, Toews G. GM-CSF regulates bleomycin-induced pulmonary fibrosis via a prostaglandin E2 mechanism. J Immunol 2000;165:40324039.
96. Keerthisingam CB, Jenkins RG, Harrison NK, Hernandez-Rodriguez NA, Booth H, Laurent GJ, Hart SL, Foster ML, McAnulty RJ. 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;158:14111422.
97. Moore B, Paine R, Christensen P, Moore T, Sitterding S, Ngan R, Wilke C, Kuziel W, Toews G. Protection from pulmonary fibrosis in the absence of CCR2 signaling. J Immunol 2001;167:43684377.
98. Gharaee-Kermani M, McCullumsmith RE, Charo IF, Kunkel SL, Phan SH. CC-chemokine receptor 2 required for bleomycin-induced pulmonary fibrosis. Cytokine 2003;24:266276.
99. Bertozzi P, Astedt B, Zenzius L, Lynch K, LeMaire F, Zapol W, Chapman HA. Depressed bronchoalveolar urokinase activity in patients with adult respiratory distress syndrome. N Engl J Med 1990;322:890897.
100. Chapman HA, Allen CL, Stone OL. Abnormalities in pathways of alveolar fibrin turnover among patients with interstitial lung disease. Am Rev Respir Dis 1986;133:437443.
101. Idell S, James KK, Levin EG, Schwartz BS, Manchanda N, Maunder RJ, Martin TR, McLarty J, Fair DS. Local abnormalities in coagulation and fibrinolytic pathways predispose to alveolar fibrin deposition in the adult respiratory distress syndrome. J Clin Invest 1989;84:695705.
102. Hattori N, Degen JL, Sisson TH, Liu H, Moore BB, Pandrangi RG, Simon RH, Drew AF. Bleomycin-induced pulmonary fibrosis in fibrinogen-null mice. J Clin Invest 2000;106:13411350.
103. Matsuoka H, Sisson TH, Nishiuma T, Simon RH. Plasminogen-mediated activation and release of hepatocyte growth factor from extracellular matrix. Am J Respir Cell Mol Biol 2006;35:705713.
104. Marchand-Adam S, Fabre A, Mailleux AA, Marchal J, Quesnel C, Kataoka H, Aubier M, Dehoux M, Soler P, Crestani B. Defect of pro-hepatocyte growth factor activation by fibroblasts in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2006;174:5866.
105. Liu X, Sun SQ, Ostrom RS. Fibrotic lung fibroblasts show blunted inhibition by cAMP due to deficient cAMP response element-binding protein phosphorylation. J Pharmacol Exp Ther 2005;315:678687.
106. Stapleton PP, Strong VE, Freeman TA, Winter J, Yan Z, Daly JM. Gender affects macrophage cytokine and prostaglandin E2 production and PGE2 receptor expression after trauma. J Surg Res 2004;122:17.
Correspondence and requests for reprints should be addressed to Bethany B. Moore, Ph.D., 4053 BSRB, 109 Zina Pitcher Pl., Ann Arbor, MI 48109-2200. E-mail:

This work was supported by NIH grants HL087846, AI065543, and HL091745.

Originally Published in Press as DOI: 10.1165/rcmb.2011-0025RT on March 18, 2011

Author disclosures

Related

No related items
American Journal of Respiratory Cell and Molecular Biology
45
3

Click to see any corrections or updates and to confirm this is the authentic version of record