Abstract
Purpose of review
The lung in systemic sclerosis (scleroderma) is susceptible to fibrosis and the ensuing respiratory insufficiency contributes to significant morbidity and mortality in this disease. The lack of effective therapies for pulmonary fibrosis has spurred a re-evaluation of pathobiological paradigms and therapeutic strategies in scleroderma-associated interstitial lung disease and in idiopathic pulmonary fibrosis. The purpose of this review is to examine emerging new therapeutic targets that modulate pro-fibrotic phenotypes of tissue-resident cells and the associated aberrant tissue remodeling responses in fibrotic disorders.
Recent findings
Progressive forms of tissue fibrosis, including scleroderma, are characterized by an accumulation of activated mesenchymal cells and their secreted extracellular matrix proteins in association with dysrepair of epithelial and endothelial cells. Recent studies suggest that emergence of cellular phenotypes that perpetuate loss of cellular homeostasis is characteristic of many fibrosis-related clinical syndromes.
Summary
Therapeutic strategies that modulate the fate/phenotype of reparative structural cells, including epithelial, endothelial, and mesenchymal cells, offer new opportunities for the development of more effective drugs for the treatment of fibrosis.
Keywords: Epithelial Cells, Mesenchymal Stem Cells, Fibroblasts, Apoptosis, Protein Kinase Inhibitors, PPAR gamma, Losartan, Bosentan, Hydroxymethylglutaryl CoA Reductase Inhibitors
Introduction
The focus on developing new treatments for fibrosis has shifted from anti-inflammatory and immune-modulating agents to therapeutic strategies that more directly target the fibrogenic process. The paradigm that inflammation leads to fibrosis (in a “serial” pathway) has been supplanted by the concept that inflammation and fibrosis may be independent of each other (by “parallel”, albeit interacting, pathways) [1,2]. This paradigm shift has been spurred, in large part, by clinical trials that have demonstrated minimal clinical benefit with agents that target immune/inflammatory pathways, both in scleroderma [3] and idiopathic pulmonary fibrosis (IPF) [4]. For example, a large multicenter randomized placebo-controlled study of cyclophosphamide versus placebo in patients with relatively early interstitial lung disease secondary to scleroderma yielded modest beneficial effects on lung function over a 12 month follow-up period [3].
In this review, we will focus our discussion on pathophysiologic mechanisms underlying fibrogenesis, and novel anti-fibrotic therapies that are currently in various stages of preclinical/clinical development. We will also focus this discussion on pulmonary fibrosis, in particular scleroderma and IPF, which share several key pathophysiological mechanisms.
Fibrosis: Aberrant Cellular Phenotypes and Loss of Homeostasis
The pathogenesis of tissue fibrosis is complex. Deciphering mechanisms underlying this chronic disorder requires integration of information from molecular/cellular biology, animal models of injury-provoked fibrosis, studies with disease-specific fibrotic cells/tissues, and clinical behavior/responses of the disease in human subjects. A logical starting point is to carefully examine the histopathology of tissue fibrosis in humans. In electron microscopy ultrastructural studies, there is evidence for an apparent defect in epithelial regeneration in association with expanded populations of activated mesenchymal cells [5]. Typically, there is increased rate of apoptosis in alveolar epithelial cells, while mesenchymal cells themselves, in particular myofibroblasts, acquire an apoptosis-resistant phenotype [6].
A number of different mechanisms for alveolar epithelial cell apoptosis have been proposed [6]. Recent reports suggest an important role for endoplasmic reticulum (ER) stress in apoptosis of type 2 alveolar epithelial cells (AT2 cells) [7,8]. ER stress and activation of the unfolded protein response in AT2 cells lining areas of fibrotic remodeling in IPF lungs are reported, both in patents with mutations in surfactant protein-C (SP-C) and in sporadic cases in which such mutations were absent [7]. ER stress pathway proteins and markers of apoptosis are expressed in AT2 cells near dense zones of fibrosis in IPF, but not in the apoptosis of alveolar epithelium associated with chronic obstructive pulmonary disease (COPD) [8]. Another mechanism relevant to apoptosis-susceptibility, which may also contribute to impaired epithelial regeneration, is the role of telomere maintenance and senescence of adult stem cells [9-11].
In contrast to epithelial/endothelial cells which appear to be injure-prone and apoptosis-susceptible, (myo)fibroblasts become resistant to apoptosis, both in IPF [6,12] and in scleroderma [13-15]. We believe studies aimed at understanding mechanisms for the apparent differential susceptibilities to apoptosis of epithelial/endothelial cells versus mesenchymal cells will provide important insights into the progressive nature of this fatal lung disease. Importantly, such studies have the potential to uncover novel therapeutic targets that regulate the pro-fibrotic phenotype/fates of tissue-resident cells.
Therapeutic Strategies to Promote Epithelial/Endothelial Regeneration and Repair
Epithelial lining cells and vascular endothelial cells (to varying degrees) of organ systems that develop fibrosis are typically in a state of dysrepair. In classic studies with explanted murine lungs, it has been demonstrated that injury to the alveolar epithelium, when associated with ineffective repair, is sufficient to disturb normal epithelial-mesenchymal interactions and promote fibrogenesis [16]; that this occurs in a blood-free environment supports a direct role for tissue-resident cells in the fibrogenic process. A diverse number of phenotypes and fates of alveolar epithelial cells (AECs) have been described in fibrotic tissues; this includes hyperplastic epithelium [17], apoptosis [18], and epithelial-mesenchymal transition (EMT) [19,20]. Together, this suggests that the process of alveolar regeneration that involves differentiation of tissue-resident facultative stem cells of the alveolar epithelium, the AT2 cells, to type 1 alveolar epithelial (AT1) cells fails to proceed normally in the fibrotic lung. Thus, therapeutic strategies which promote normal alveolar regeneration may be expected to be of benefit in pulmonary fibrosis. A few such strategies are discussed here.
Hepatocyte growth factor (HGF)
HGF signaling via its receptor, c-met, modulates pro-fibrotic phenotypes of potentially several cell types, including epithelial cells. In addition to functioning as a mitogen, motogen, and survival factor for epithelial cells, a number of recent studies indicate that HGF prevents EMT by interfering with transforming growth factor-β (TGF-β) signaling [21]. The ability of HGF to alter the fate of other cell types such as to promote myofibroblast apoptosis may also contribute to its in vivo anti-fibrotic effects [22]. In animal models of bleomycin injury-induced lung fibrosis, administration of HGF even when administered in a delayed manner has been shown to protect against fibrosis [23-26]. The optimal strategies for delivery of HGF to target tissues need to be determined, although recombinant forms of HGF are currently undergoing Phase I/II trials for acute liver failure (www.clinicaltrials.gov; NCT00225901) and chronic venous leg ulcers (www.clinicaltrials.gov; NCT00797706).
Keratinocyte growth factor (KGF)
KGF is produced primarily by mesenchymal cells and is known to be a potent paracrine mediator of proliferation, migration and differentiation of AT2 epithelial cells. Interestingly fibroblasts isolated from lungs of IPF patients have diminished capacity to induce KGF secretion [27]. Similar to studies with HGF, administration of KGF protects against fibrosis in animal models of bleomycin lung injury [28-30]. Human recombinant KGF (palifermin) has been approved for the treatment of severe oral mucositis complicating myelotoxic therapy and hematopoietic stem cell support in patients with hematological malignancies. To our knowledge, no clinical trials of KGF for fibrotic disorders have been undertaken.
Nitric Oxide (NO)
In addition to its well recognized vasodilatory and vasculoprotective actions, NO may mediate anti-fibrotic effects by actions on epithelial cells and/or mesenchymal cells. Studies in rat AECs suggest that endogenous generation of NO may prevent AECs from undergoing an EMT-like phenotype and exogenous NO suppresses TGF-β1-induced EMT [31]. NO has been shown to mediate anti-fibrotic effects in vivo, as demonstrated in a baboon model of chronic lung disease associated with premature birth [32]. We are unaware of any clinical trials of NO for fibrotic disorders (excluding cystic fibrosis); however, the NIH-sponsored IPF Clinical Research Network (IPFnet) is conducting a Phase III trial of sildenafil, a phosphodiesterase type 5 (PDE5) inhibitor that augments NO-cGMP signaling, to test exercise performance in patients with advanced IPF (www.clinicaltrials.gov; NCT00517933).
eIF4E inhibitor-1 (4Ei-1)
eIF4E is the rate-controlling component of the translation initiation complex e1F4F that associates with the cap structure of mRNA to initiate protein synthesis in eukaryotic cells, essential for growth and survival of neoplastic and fibrogenic cells. Ghosh and colleagues [33] sought to explore the effect of a recently synthesized inihibitor of the association of elF4E with the mRNA cap (4Ei-1) in a zebrafish EMT model. 4Ei-1 inhibited cap-dependent translation in a dose-dependent manner in zebrafish embryos without evidence of developmental abnormalities. Furthermore, embryo explants injected with elF4E underwent EMT whereas embryos co-injected with 4Ei-1 had complete abrogation of EMT without evidence of toxicity defined by morphological/pathological parameters [33]; compounds designed to inhibit cap-dependent translation deserve further exploration in pre-clinical disease models of cancer and fibrosis.
Cell-based therapies
There has been recent excitement over the prospects of cell-based therapies for the treatment of chronic degenerative diseases. If such therapies can be designed to enhance normal epithelial regeneration in fibrotic organs, this enthusiasm may be justifiable. Two cell-based therapeutic approaches that have garnered significant interest in this regard include mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs). Murine bone marrow-derived MSCs have been shown to home to the lung, adopt an epithelium-like phenotype, and reduce inflammation and collagen deposition in lungs of bleomycin-injured mice [34]. Subsequently, the protective effect of these exogenously administered MSCs appears to be mediated by the release of ant-inflammatory paracrine mediators [35]. It appears that the ability of bone marrow cells to reconstitute and engraft lung epithelium is more limited than initially estimated [36]. There is evidence for tissue-resident MSCs in adult organ systems, including the lung [37-39]. The behavior and phenotype/fate of tissue-resident MSCs deserve further study, perhaps even before embarking on clinical trials of exogenous MSCs for fibrotic diseases.
The potential for generating organ-specific epithelial cells from embryonic stem cells (ESCs) or iPSCs affords another opportunity to regenerate epithelium which is invariably anarchic in fibrotic disorders. Strategies for generating pure populations of AT2 cells from human ESCs have been described [40]. Here again, one would like to know why the endogenous hyperplastic AT2 cells of the fibrotic IPF lung fail to differentiate normally into AT1 cells; if it were related to an altered (differentiation-prohibitive) cellular microenvironment, such cell-based therapies would be predicted to fail. On the other hand, if relative insufficiency or inherent defect of endogenous AT2 cells (e.g. a genetic defect that results in a protein trafficking abnormality and ER stress) prevent their ability to differentiate into AT1 cells, exogenous delivery of “normal” AT2 cells, when successfully engrafted, may prove successful.
Therapeutic Strategies that Target Mesenchymal Cell Activation and Survival
Activated mesenchymal cells are effectors of the exuberant extracellular matrix (ECM) production and architectural tissue remodeling, characteristic features of progressive fibrotic disorders. Myofibroblasts represent a differentiated/specialized cell type capable of mediating enhanced ECM synthesis and tissue contracture [41]. Myofibroblast differentiation is critically dependent on TGF-β1 [42,43], in addition to biomechanical tension signaling [44]. While a number of pharmaceutical companies are developing strategies to interfere with TGF-β1 signaling in cancer and fibrosis, more specific strategies downstream of TGF-β ligand(s)/receptor(s) to inhibit myofibroblast differentiation are also being developed; we will discuss a few of these here along with selected agents designed to inhibit myofibroblast contractility and/or survival.
Peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists
PPAR-γ is one of the three subtypes of PPARs and is a member of the nuclear hormone receptor superfamily. PPARs were originally found to be important regulators of lipid and glucose homeostasis; however, more recently, PPAR-γ has been implicated in the regulation of inflammation, fibrosis, and cancer. The thiazolidinediones (TZDs) such as rosiglitazone, pioglitazone and troglitazone are synthetic PPAR-γ agonists used successfully for the treatment of diabetes type II since the 1990s [45]. Both natural and synthetic PPAR-γ agonists have been shown to decrease lung injury/fibrosis induced by intratracheal administration of bleomycin in mice [46]. More recently, fibroblasts isolated from patients with different types of fibrotic lung disease when stimulated with PPAR-γ agonists demonstrate reduced proliferative responses to mitogens and attenuated TGF-β1-induced myofibroblast differentiation [47]; additionally, troglitazone administered 10 days after the intratracheal instillation of bleomycin in mice was effective in inhibiting lung fibrosis. In a murine model of subcutaneous bleomycin-induced scleroderma, the concomitant intraperitoneal injection of rosiglitazone inhibited early inflammation responses and abrogated skin fibrosis, local collagen accumulation, lipoatrophy and reduced tissue accumulation of myofibroblasts [48]. A subset of mice in this study did not receive rosiglitazone until 7 days after bleomycin injury and, despite this, there was evidence of reduced skin fibrosis [48], supporting the notion that PPAR-γ agonists may abrogate fibrosis through more direct “anti-fibrotic” mechanisms rather than through its recognized anti-inflammatory effects.
Protein kinase inhibitors
Protein kinases have become an important target for drug development [49]. This enthusiasm has been largely driven by the success of imatinib mesylate (Gleevec™), the first orally administered protein kinase inhibitor (PKI) approved for the treatment of chronic myelogenous leukemia (CML) [50]. Imatinib inhibits the Bcr-Abl tyrosine kinase which is constitutively activated in CML [51]. However, although imatinib is a relatively specific inhibitor of Bcr-Abl kinase, it also inhibits the c-Kit and PDGF receptor tyrosine kinases [52]. Based on its ability to inhibit non-SMAD TGF-β signaling via c-Abl tyrosine kinase [53] and PDGF receptor signaling [52], imatinib appears to mediate in vivo anti-fibrotic effects in animal models of pulmonary fibrosis [53] and dermal fibrosis [54,55]. Some studies indicate that imatinib may arrest and even reverse established fibrosis [55,56], while others indicate that delayed administration of imatinib during the post-inflammatory phase may not be as effective [57,58]. A potential concern with imatinib is its detrimental effects on epithelial cells, which may interfere with regenerative capacity of epithelium, and counterbalance putative beneficial effects on activated mesenchymal cells [58]. A Phase II multicenter double-blind, randomized and placebo controlled trial of imatinib in IPF was completed in 2007; however, results have not been published (www.clinicaltrials.gov; NCT00131274). Another Phase II study of the efficacy and tolerability of imatinib in dermal fibrosis associated with scleroderma has completed enrollment and results will be available in early 2010 (www.clinicaltrials.gov; NCT00613171). PKIs that target pro-survival signaling pathways in mesenchymal cells may also have utility in the treatment of fibrotic disorders [59].
Modulation of the contractile myofibroblast phenotype
Pharmacologic agents that modulate the contractile phenotype of myofibroblasts may be particularly effective due to a central role of this cellular phenotype in chronic fibro-contractive disorders. Such agents might include inhibitors of endothelin-1 (ET-1) and Rho kinase (ROCK), a more downstream mediator of ligand-receptor binding/activation. ET-1 is a potent endogenous vasoconstrictor that is implicated in the pathogenesis of pulmonary arterial hypertension (PAH), and the dual specificity ET receptor antagonist, bosentan, is FDA-approved for treatment of PAH [60]. Pre-clinical studies support a potential role for bosentan as an anti-fibrotic agent [61,62]. A randomized placebo-controlled clinical trial enrolled 158 patients with relatively early IPF to receive either placebo or bosentan for 12 months [63]. This study failed to demonstrate efficacy based on the primary endpoint of six-minute walk distance at 12 months; however, a post hoc analysis of patients that were diagnosed via a surgical lung biopsy demonstrated both a survival and a quality of life advantage in favor of bosentan. A larger multicenter, placebo-controlled, randomized study with bosentan in IPF patients diagnosed via a surgical lung biopsy and with minimal fibrotic changes on HRCT of the chest is underway [64].
The RhoA/ROCK pathway is a critical regulator of cellular contractility [65]. A small molecule inhibitor of ROCK, fasudil, is approved for the treatment of cerebral vasospasm in Japan. Fasudil has also been also recently been studied in U.S. populations for other disease indications (www.clinicaltrials.gov). One of these, a Phase III study of fasudil in the treatment of Raynaud's phenomenon is currently ongoing enrolling (www.clinicaltrials.gov; NCT00498615). To our knowledge, fasudil has not been studied for the treatment of fibrotic diseases. The 3-hydroxy-3-methylglutaryl CoenzymeA (HMG-CoA) reductase inhibitors (statins), in addition to their cholesterol-lowering properties, inhibit RhoA/ROCK activation [66]. Simvastatin inhibits myofibroblast differentiation and contractility in human lung fibroblasts derived from IPF patients [67]. Mice treated with simvastatin, prior to bleomycin-induced lung fibrosis and for the duration of the study, demonstrate a reduction in inflammatory cell influx and lung collagen accumulation in association with downregulation of CTGF and TGF-β1 expression [68]. Testing a potential role of statins in subgroups of patients with fibrosis may be warranted.
Modulators of oxidative stress and redox signaling
Oxidative stress has been implicated in the pathogenesis of tissue fibrosis [69,70]. Additionally, there is now ample evidence that reactive oxygen species (ROS) function as signaling molecules when they are generated in a compartmentalized and regulated manner [71,72]. The NADPH oxidase (NOX) family of enzymes, which catalyze the reduction of O2 to form ROS, are likely to have roles in normal cellular physiology; indeed, the number of NOX enzymes have increased during eukaryotic evolution with seven NOX isoforms identified in mammals [73-75]. However, the roles of individual NOX family members in normal physiology and mechanisms by which they contribute to disease pathogenesis are only beginning to be elucidated.
Myofibroblasts generate ROS in response to TGF-β1 and the NOX4 isoform has been identified as a source of TGF-β1-induced ROS generation in cardiac and lung myofibroblasts; NOX4 is implicated in the induction of myofibroblast differentiation [76,77]. A recent study reported that NOX4 is upregulated in lungs of patients with IPF and in mice subjected to non-infectious lung injury. In lung mesenchymal cells, NOX4-dependent generation of H2O2 is required for TGF-β1-induced myofibroblast differentiation, extracellular matrix (ECM) production, and contractility. Furthermore, genetic or pharmacologic targeting of NOX4 inhibited fibrogenesis in two different murine models of lung injury [77]. NOX4-dependent extracellular generation of H2O2 by lung myofibroblasts may mediate additional fibrogenic effects in tissues by inducing epithelial cell apoptosis [78] and/or epithelial-mesenchymal transition [79], or by inducing ECM crosslinking reactions in the presence of extracellular heme peroxidases [80]. These studies support a role for NOX4 in tissue repair functions of myofibroblasts and fibrogenesis as well as provide proof-of-concept for therapeutic targeting of NOX4 in abrogating lung fibrosis. The development of small molecule inhibitors and/or other strategies targeting NOX4 offers substantial promise for the treatment of recalcitrant fibrotic disorders, such as IPF and scleroderma.
Conclusion
A number of different mechanisms have been implicated in the pathogenesis of progressive fibrosing disorders. Contemporary concepts regarding fibrogenic mechanisms have converged on a paradigm of epithelial-mesenchymal disrepair and failure of tissue regeneration. Novel therapeutic agents based on this paradigm are beginning to produce promising results in pre-clinical disease-specific cell culture models and animal models of fibrosis. However, promising pre-clinical results, and even preliminary (Phase I/II) clinical studies, do not always translate into clear benefit for patients with the disease. Future well-conducted and controlled clinical trials are necessary before these novel agents can be recommended for the treatment of progressive fibrosing disorders.
Acknowledgments
This work is supported in part by National Institutes of Health grants, U10 HL080510 (Gulf South IPF Clinical Research Network), and R01 HL067967.
References
- 1.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]
- 2.Thannickal VJ, Toews GB, White ES, et al. Mechanisms of pulmonary fibrosis. Annu Rev Med. 2004;55:395–417. doi: 10.1146/annurev.med.55.091902.103810. [DOI] [PubMed] [Google Scholar]; *This review advances the concept of inflammation and fibrosis as “parallel” (in contrast to “serial”) responses to tissue injury. Based on this paradigm, anti-inflammatory and immune-modulatory drug therapies may be ineffective as anti-fibrotic agents.
- 3.Tashkin DP, Elashoff R, Clements PJ, et al. Cyclophosphamide versus placebo in scleroderma lung disease. N Engl J Med. 2006;354:2655–2666. doi: 10.1056/NEJMoa055120. [DOI] [PubMed] [Google Scholar]; **This study reported a modest, although statistically significant, benefit in lung function (forced vital capacity) at one year of treatment with cyclophosphamide versus placebo; based on the toxicity profile of the drug and marginal clinical benefit, routine use of cyclophosphamide for scleroderma lung disease is not recommended
- 4.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;161:646–664. doi: 10.1164/ajrccm.161.2.ats3-00. [DOI] [PubMed] [Google Scholar]
- 5.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]
- 6.Thannickal VJ, Horowitz JC. Evolving concepts of apoptosis in idiopathic pulmonary fibrosis. Proc Am Thorac Soc. 2006;3:350–356. doi: 10.1513/pats.200601-001TK. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lawson WE, Crossno PF, Polosukhin VV, et al. Endoplasmic reticulum stress in alveolar epithelial cells is prominent in IPF: association with altered surfactant protein processing and herpesvirus infection. Am J Physiol Lung Cell Mol Physiol. 2008;294:L1119–1126. doi: 10.1152/ajplung.00382.2007. [DOI] [PubMed] [Google Scholar]; *This was the first study to indicate ER stress and activation of the unfolded protein response in AT2 cells lining areas of fibrotic remodeling in IPF lungs, both in patents with mutations in surfactant protein-C (SP-C) and in sporadic cases in which such mutations were absent. It also suggests herpesvirus-induced ER stress and apoptosis of AT2 cells as a potential mechanism for disease exacerbation/progression.
- 8.Korfei M, Ruppert C, Mahavadi P, et al. Epithelial endoplasmic reticulum stress and apoptosis in sporadic idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2008;178:838–846. doi: 10.1164/rccm.200802-313OC. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This study demonstrated ER stress pathway proteins in association with apoptosis of AT2 cells in IPF lung tissue, supporting a potential role for ER-mediated apoptosis of regenerating alveolar epithelial cells in the initiation/progression of this fibrotic lung disease.
- 9.Alder JK, Chen JJ, Lancaster L, et al. Short telomeres are a risk factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci U S A. 2008;105:13051–13056. doi: 10.1073/pnas.0804280105. [DOI] [PMC free article] [PubMed] [Google Scholar]; **This study demonstrates that a short telomere phenotype in leukocytes and organ-specific cells may underlie the pathogenesis of sporadic cases of IIP and provides a plausible explanation for the age-related onset of IPF.
- 10.Cronkhite JT, Xing C, Raghu G, et al. Telomere shortening in familial and sporadic pulmonary fibrosis. Am J Respir Crit Care Med. 2008;178:729–737. doi: 10.1164/rccm.200804-550OC. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This study indicates that at least a subset of patients with pulmonary fibrosis have shortened telomeres that cannot be explained by coding mutations in telomerase and supports the notion of impaired regenerative potential in adult stem cells as a possible mechanism for susceptibility to fibrosis.
- 11.Thannickal VJ, Loyd JE. Idiopathic pulmonary fibrosis: a disorder of lung regeneration? Am J Respir Crit Care Med. 2008;178:663–665. doi: 10.1164/rccm.200807-1127ED. [DOI] [PubMed] [Google Scholar]
- 12.Horowitz JC, Rogers DS, Sharma V, et al. Combinatorial activation of FAK and AKT by transforming growth factor-beta1 confers an anoikis-resistant phenotype to myofibroblasts. Cell Signal. 2007;19:761–771. doi: 10.1016/j.cellsig.2006.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jun JB, Kuechle M, Harlan JM, et al. Fibroblast and endothelial apoptosis in systemic sclerosis. Curr Opin Rheumatol. 2003;15:756–760. doi: 10.1097/00002281-200311000-00012. [DOI] [PubMed] [Google Scholar]
- 14.Mimura Y, Ihn H, Jinnin M, et al. Constitutive phosphorylation of focal adhesion kinase is involved in the myofibroblast differentiation of scleroderma fibroblasts. J Invest Dermatol. 2005;124:886–892. doi: 10.1111/j.0022-202X.2005.23701.x. [DOI] [PubMed] [Google Scholar]
- 15.Jun JB, Kuechle M, Min J, et al. Scleroderma fibroblasts demonstrate enhanced activation of Akt (protein kinase B) in situ. J Invest Dermatol. 2005;124:298–303. doi: 10.1111/j.0022-202X.2004.23559.x. [DOI] [PubMed] [Google Scholar]
- 16.Adamson IY, Young L, Bowden DH. Relationship of alveolar epithelial injury and repair to the induction of pulmonary fibrosis. Am J Pathol. 1988;130:377–383. [PMC free article] [PubMed] [Google Scholar]
- 17.Chilosi M, Poletti V, Zamo A, et al. Aberrant Wnt/beta-catenin pathway activation in idiopathic pulmonary fibrosis. Am J Pathol. 2003;162:1495–1502. doi: 10.1016/s0002-9440(10)64282-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Uhal BD. Apoptosis in lung fibrosis and repair. Chest. 2002;122:293S–298S. doi: 10.1378/chest.122.6_suppl.293s. [DOI] [PubMed] [Google Scholar]
- 19.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]; **This was the first study to support a role for EMT in the pathogenesis of fibrotic lung disease
- 20.Kim KK, Kugler MC, Wolters PJ, et al. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci U S A. 2006;103:13180–13185. doi: 10.1073/pnas.0605669103. [DOI] [PMC free article] [PubMed] [Google Scholar]; **This study provides proof for a significant contribution from EMT-derived myofibroblasts in fibrogenesis following experimental lung injury in mice
- 21.Shukla MN, Rose JL, Ray R, et al. Hepatocyte growth factor inhibits epithelial to myofibroblast transition in lung cells via Smad7. Am J Respir Cell Mol Biol. 2009;40:643–653. doi: 10.1165/rcmb.2008-0217OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Mizuno S, Matsumoto K, Li MY, et al. 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]
- 23.Yaekashiwa M, Nakayama S, Ohnuma K, et al. Simultaneous or delayed administration of hepatocyte growth factor equally represses the fibrotic changes in murine lung injury induced by bleomycin. A morphologic study. Am J Respir Crit Care Med. 1997;156:1937–1944. doi: 10.1164/ajrccm.156.6.9611057. [DOI] [PubMed] [Google Scholar]
- 24.Dohi M, Hasegawa T, Yamamoto K, et al. 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]
- 25.Watanabe M, Ebina M, Orson FM, et al. Hepatocyte growth factor gene transfer to alveolar septa for effective suppression of lung fibrosis. Mol Ther. 2005;12:58–67. doi: 10.1016/j.ymthe.2005.02.019. [DOI] [PubMed] [Google Scholar]
- 26.Gazdhar A, Fachinger P, van Leer C, et al. Gene transfer of hepatocyte growth factor by electroporation reduces bleomycin-induced lung fibrosis. Am J Physiol Lung Cell Mol Physiol. 2007;292:L529–536. doi: 10.1152/ajplung.00082.2006. [DOI] [PubMed] [Google Scholar]
- 27.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]
- 28.Yi ES, Williams ST, Lee H, et al. Keratinocyte growth factor ameliorates radiation- and bleomycin-induced lung injury and mortality. Am J Pathol. 1996;149:1963–1970. [PMC free article] [PubMed] [Google Scholar]
- 29.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]
- 30.Sugahara K, Iyama K, Kuroda MJ, et al. Double intratracheal instillation of keratinocyte growth factor prevents bleomycin-induced lung fibrosis in rats. J Pathol. 1998;186:90–98. doi: 10.1002/(SICI)1096-9896(199809)186:1<90::AID-PATH137>3.0.CO;2-X. [DOI] [PubMed] [Google Scholar]
- 31.Vyas-Read S, Shaul PW, Yuhanna IS, et al. Nitric oxide attenuates epithelial-mesenchymal transition in alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2007;293:L212–221. doi: 10.1152/ajplung.00475.2006. [DOI] [PubMed] [Google Scholar]
- 32.McCurnin DC, Pierce RA, Chang LY, et al. Inhaled NO improves early pulmonary function and modifies lung growth and elastin deposition in a baboon model of neonatal chronic lung disease. Am J Physiol Lung Cell Mol Physiol. 2005;288:L450–459. doi: 10.1152/ajplung.00347.2004. [DOI] [PubMed] [Google Scholar]
- 33.Ghosh B, Benyumov AO, Ghosh P, et al. Nontoxic chemical interdiction of the epithelial-to-mesenchymal transition by targeting cap-dependent translation. ACS Chem Biol. 2009;4:367–377. doi: 10.1021/cb9000475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ortiz LA, Gambelli F, McBride C, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A. 2003;100:8407–8411. doi: 10.1073/pnas.1432929100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ortiz LA, Dutreil M, Fattman C, et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci U S A. 2007;104:11002–11007. doi: 10.1073/pnas.0704421104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kotton DN, Fabian AJ, Mulligan RC. Failure of bone marrow to reconstitute lung epithelium. Am J Respir Cell Mol Biol. 2005;33:328–334. doi: 10.1165/rcmb.2005-0175RC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119:2204–2213. doi: 10.1242/jcs.02932. [DOI] [PubMed] [Google Scholar]
- 38.Lama VN, Smith L, Badri L, et al. Evidence for tissue-resident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J Clin Invest. 2007;117:989–996. doi: 10.1172/JCI29713. [DOI] [PMC free article] [PubMed] [Google Scholar]; **This study provides the first evidence for the existence of a population of mesenchymal progenitor/stem cells that reside locally in the human adult lung
- 39.Summer R, Fitzsimmons K, Dwyer D, et al. Isolation of an adult mouse lung mesenchymal progenitor cell population. Am J Respir Cell Mol Biol. 2007;37:152–159. doi: 10.1165/rcmb.2006-0386OC. [DOI] [PMC free article] [PubMed] [Google Scholar]; *This study provides additional proof for resident mesenchymal progenitor cells in the postnatal mammalian lung
- 40.Wang D, Haviland DL, Burns AR, et al. A pure population of lung alveolar epithelial type II cells derived from human embryonic stem cells. Proc Natl Acad Sci U S A. 2007;104:4449–4454. doi: 10.1073/pnas.0700052104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hinz B, Phan SH, Thannickal VJ, et al. The myofibroblast: one function, multiple origins. Am J Pathol. 2007;170:1807–1816. doi: 10.2353/ajpath.2007.070112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Desmouliere A, Geinoz A, Gabbiani F, et al. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J Cell Biol. 1993;122:103–111. doi: 10.1083/jcb.122.1.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.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]
- 44.Hinz B, Mastrangelo D, Iselin CE, et al. Mechanical tension controls granulation tissue contractile activity and myofibroblast differentiation. Am J Pathol. 2001;159:1009–1020. doi: 10.1016/S0002-9440(10)61776-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Becker J, Delayre-Orthez C, Frossard N, et al. Regulation of inflammation by PPARs: a future approach to treat lung inflammatory diseases? Fundam Clin Pharmacol. 2006;20:429–447. doi: 10.1111/j.1472-8206.2006.00425.x. [DOI] [PubMed] [Google Scholar]
- 46.Genovese T, Cuzzocrea S, Di Paola R, et al. Effect of rosiglitazone and 15-deoxy-Delta12,14-prostaglandin J2 on bleomycin-induced lung injury. Eur Respir J. 2005;25:225–234. doi: 10.1183/09031936.05.00049704. [DOI] [PubMed] [Google Scholar]
- 47.Milam JE, Keshamouni VG, Phan SH, et al. PPAR-gamma agonists inhibit profibrotic phenotypes in human lung fibroblasts and bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2008;294:L891–901. doi: 10.1152/ajplung.00333.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wu M, Melichian DS, Chang E, et al. Rosiglitazone abrogates bleomycin-induced scleroderma and blocks profibrotic responses through peroxisome proliferator-activated receptor-gamma. Am J Pathol. 2009;174:519–533. doi: 10.2353/ajpath.2009.080574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cohen P. Protein kinases--the major drug targets of the twenty-first century? Nat Rev Drug Discov. 2002;1:309–315. doi: 10.1038/nrd773. [DOI] [PubMed] [Google Scholar]
- 50.Savage DG, Antman KH. Imatinib mesylate--a new oral targeted therapy. N Engl J Med. 2002;346:683–693. doi: 10.1056/NEJMra013339. [DOI] [PubMed] [Google Scholar]
- 51.Druker BJ. STI571 (Gleevec) as a paradigm for cancer therapy. Trends Mol Med. 2002;8:S14–18. doi: 10.1016/s1471-4914(02)02305-5. [DOI] [PubMed] [Google Scholar]
- 52.Buchdunger E, Cioffi CL, Law N, et al. Abl protein-tyrosine kinase inhibitor STI571 inhibits in vitro signal transduction mediated by c-kit and platelet-derived growth factor receptors. J Pharmacol Exp Ther. 2000;295:139–145. [PubMed] [Google Scholar]
- 53.Daniels CE, Wilkes MC, Edens M, et al. Imatinib mesylate inhibits the profibrogenic activity of TGF-beta and prevents bleomycin-mediated lung fibrosis. J Clin Invest. 2004;114:1308–1316. doi: 10.1172/JCI19603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Distler JH, Jungel A, Huber LC, et al. Imatinib mesylate reduces production of extracellular matrix and prevents development of experimental dermal fibrosis. Arthritis Rheum. 2007;56:311–322. doi: 10.1002/art.22314. [DOI] [PubMed] [Google Scholar]
- 55.Akhmetshina A, Venalis P, Dees C, et al. Treatment with imatinib prevents fibrosis in different preclinical models of systemic sclerosis and induces regression of established fibrosis. Arthritis Rheum. 2009;60:219–224. doi: 10.1002/art.24186. [DOI] [PubMed] [Google Scholar]
- 56.Chaudhary NI, Schnapp A, Park JE. Pharmacologic differentiation of inflammation and fibrosis in the rat bleomycin model. Am J Respir Crit Care Med. 2006;173:769–776. doi: 10.1164/rccm.200505-717OC. [DOI] [PubMed] [Google Scholar]
- 57.Aono Y, Nishioka Y, Inayama M, et al. Imatinib as a novel antifibrotic agent in bleomycin-induced pulmonary fibrosis in mice. Am J Respir Crit Care Med. 2005;171:1279–1285. doi: 10.1164/rccm.200404-531OC. [DOI] [PubMed] [Google Scholar]
- 58.Vittal R, Zhang H, Han MK, et al. Effects of the protein kinase inhibitor, imatinib mesylate, on epithelial/mesenchymal phenotypes: implications for treatment of fibrotic diseases. J Pharmacol Exp Ther. 2007;321:35–44. doi: 10.1124/jpet.106.113407. [DOI] [PubMed] [Google Scholar]
- 59.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]
- 60.Rubin LJ, Badesch DB, Barst RJ, et al. Bosentan therapy for pulmonary arterial hypertension. N Engl J Med. 2002;346:896–903. doi: 10.1056/NEJMoa012212. [DOI] [PubMed] [Google Scholar]
- 61.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]
- 62.Park SH, Saleh D, Giaid A, et al. Increased endothelin-1 in bleomycin-induced pulmonary fibrosis and the effect of an endothelin receptor antagonist. Am J Respir Crit Care Med. 1997;156:600–608. doi: 10.1164/ajrccm.156.2.9607123. [DOI] [PubMed] [Google Scholar]
- 63.King TE, Jr, Behr J, Brown KK, et al. BUILD-1: a randomized placebo-controlled trial of bosentan in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2008;177:75–81. doi: 10.1164/rccm.200705-732OC. [DOI] [PubMed] [Google Scholar]
- 64.King TE., Jr Bosentan for idiopathic pulmonary fibrosis. Curr Opin Investig Drugs. 2008;9:1171–1179. [PubMed] [Google Scholar]
- 65.Burridge K, Chrzanowska-Wodnicka M. Focal adhesions, contractility, and signaling. Annu Rev Cell Dev Biol. 1996;12:463–518. doi: 10.1146/annurev.cellbio.12.1.463. [DOI] [PubMed] [Google Scholar]
- 66.Liao JK, Seto M, Noma K. Rho kinase (ROCK) inhibitors. J Cardiovasc Pharmacol. 2007;50:17–24. doi: 10.1097/FJC.0b013e318070d1bd. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Watts KL, Sampson EM, Schultz GS, et al. Simvastatin inhibits growth factor expression and modulates profibrogenic markers in lung fibroblasts. Am J Respir Cell Mol Biol. 2005;32:290–300. doi: 10.1165/rcmb.2004-0127OC. [DOI] [PubMed] [Google Scholar]
- 68.Ou XM, Feng YL, Wen FQ, et al. Simvastatin attenuates bleomycin-induced pulmonary fibrosis in mice. Chin Med J (Engl) 2008;121:1821–1829. [PubMed] [Google Scholar]
- 69.Kinnula VL, Fattman CL, Tan RJ, et al. Oxidative stress in pulmonary fibrosis: a possible role for redox modulatory therapy. Am J Respir Crit Care Med. 2005;172:417–422. doi: 10.1164/rccm.200501-017PP. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gabrielli A, Svegliati S, Moroncini G, et al. Oxidative stress and the pathogenesis of scleroderma: the Murrell's hypothesis revisited. Semin Immunopathol. 2008;30:329–337. doi: 10.1007/s00281-008-0125-4. [DOI] [PubMed] [Google Scholar]
- 71.Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1005–1028. doi: 10.1152/ajplung.2000.279.6.L1005. [DOI] [PubMed] [Google Scholar]
- 72.Thannickal VJ. Oxygen in the evolution of complex life and the price we pay. Am J Respir Cell Mol Biol. 2009;40:507–510. doi: 10.1165/rcmb.2008-0360PS. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Kawahara T, Quinn MT, Lambeth JD. Molecular evolution of the reactive oxygen-generating NADPH oxidase (Nox/Duox) family of enzymes. BMC Evol Biol. 2007;7:109. doi: 10.1186/1471-2148-7-109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bedard K, Lardy B, Krause KH. NOX family NADPH oxidases: not just in mammals. Biochimie. 2007;89:1107–1112. doi: 10.1016/j.biochi.2007.01.012. [DOI] [PubMed] [Google Scholar]
- 75.Sumimoto H. Structure, regulation and evolution of Nox-family NADPH oxidases that produce reactive oxygen species. FEBS J. 2008;275:3249–3277. doi: 10.1111/j.1742-4658.2008.06488.x. [DOI] [PubMed] [Google Scholar]
- 76.Cucoranu I, Clempus R, Dikalova A, et al. NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ Res. 2005;97:900–907. doi: 10.1161/01.RES.0000187457.24338.3D. [DOI] [PubMed] [Google Scholar]
- 77.Hecker L, Vittal R, Jones T, et al. NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med. 2009 doi: 10.1038/nm.2005. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]; **This study indicates a critical role for NOX4 in myofibroblast activation and that therapeutic targeting of NOX4 may represent an effective anti-fibrotic strategy
- 78.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]
- 79.Rhyu DY, Yang Y, Ha H, et al. Role of reactive oxygen species in TGF-beta1-induced mitogen-activated protein kinase activation and epithelial-mesenchymal transition in renal tubular epithelial cells. J Am Soc Nephrol. 2005;16:667–675. doi: 10.1681/ASN.2004050425. [DOI] [PubMed] [Google Scholar]
- 80.Larios JM, Budhiraja R, Fanburg BL, et al. 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]