Pulmonary Hypertension and Idiopathic Pulmonary Fibrosis: A Tale of ...

46 downloads 0 Views 936KB Size Report
Virginia Commonwealth University, Richmond, Virginia. Idiopathic pulmonary fibrosis (IPF) is ...... Strange C, Highland KB. Pulmonary hypertension in interstitial ...
Translational Review Pulmonary Hypertension and Idiopathic Pulmonary Fibrosis A Tale of Angiogenesis, Apoptosis, and Growth Factors Laszlo Farkas1,2,3, Jack Gauldie1, Norbert F. Voelkel3, and Martin Kolb1,2 1 Departments of Medicine, Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada; 2Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada; and 3Victoria Johnson Center for Obstructive Lung Diseases, Virginia Commonwealth University, Richmond, Virginia

Idiopathic pulmonary fibrosis (IPF) is a disabling disease of the lung parenchyma, characterized by progressive accumulation of scar tissue and myofibroblast activation after repetitive epithelial microinjury. The therapeutic options are limited, and patients usually die within a few years after diagnosis. Pulmonary hypertension (PH) in IPF has been increasingly recognized as a condition with relevance for the overall prognosis. Treatment trials are being designed, but to be effective, it is crucial to better understand the pathobiology of PH in IPF: the traditional concept, that hypoxic vasoconstriction and accumulation of scar tissue are mainly responsible for the development of PH in IPF, has been challenged. Recent studies, including our own in vivo research, suggest that the underlying pathobiology is much more complex, and includes a complicated interaction of epithelial cells, fibroblasts, and vascular cells. This interaction seems to be regulated by a large variety of angiogenesis promoters and inhibitors, as well as growth factors. Central components seem to be endothelial apoptosis and growth factor–induced remodeling of the pulmonary artery wall. The present review gives a conceptual overview about known and putative mechanisms that are involved in the development of PH in IPF. This report summarizes currently available therapeutic options, and also translates experimental research to discuss potential novel biomarkers and therapeutic strategies derived from new concepts in pathogenesis. Keywords: idiopathic pulmonary fibrosis; pulmonary hypertension; angiogenesis; apoptosis; growth factors

Idiopathic pulmonary fibrosis (IPF) is a crippling disease because of progressive scarring of the lung tissue and the lung function of patients with IPF is characterized by a restrictive volume pattern and reduced gas exchange capacity (1–3). The typical clinical course is determined by increasing shortness of breath, decreasing exercise capacity, and, ultimately, death. Usual interstitial pneumonia (UIP) represents the histopathology representative of IPF. Features of UIP include a heterogeneous, patchy distribution of fibrosis with subpleural and basal predominance, accumulation of activated myofibroblasts in subepithelial fibroblastic foci, and excessive deposition of extracellular matrix

(Received in original form October 3, 2010 and in final form October 3, 2010) This work was supported by grants from GlaxoSmithKline, the Canadian Institutes for Health Research, and a research fellowship from the German Research Foundation (L.F.). Correspondence and requests for reprints should be addressed to Martin Kolb, M.D., Ph.D., Departments of Medicine, Pathology and Molecular Medicine, McMaster University, Firestone Institute for Respiratory Health, 50 Charlton Avenue East, Room T2121, Hamilton, ON L8N 4A6, Canada. E-mail: kolbm@ mcmaster.ca Am J Respir Cell Mol Biol Vol 45. pp 1–15, 2011 Originally Published in Press as DOI: 10.1165/rcmb.2010-0365TR on November 5, 2010 Internet address: www.atsjournals.org

(ECM) components, such as collagen and fibronectin (2, 4, 5). Transforming growth factor (TGF)–b1 is a key factor which promotes fibrosis and is, therefore, a main focus for therapy. Inflammation is not crucial for the progression of fibrosis, but a perpetual activation of TGF-b signaling through the Smad3 pathway is (6–9). IPF is not sufficiently treatable, and the prognosis after diagnosis is limited to an average survival of 3–5 years (2, 4). The mortality is predicted by a progressive decrease of the forced vital capacity, reduced distance in the 6 minute walk test with elevated O2 desaturation, and presence of pulmonary hypertension (PH) (10–18). Pulmonary arterial hypertension (PAH) has been defined as an elevation of the mean pulmonary artery (PA) pressure (PAP) over 25 mm Hg at rest, and, at the same time, a normal pulmonary capillary wedge pressure (19). PH in IPF has been increasingly recognized, and treatment trials are being designed (3, 20). The most recent clinical classification of PH categorizes PH in IPF into category 3.2 (PH associated with lung diseases and/or hypoxia; subclass, interstitial lung disease [ILD]) (21). The prevalence of PH in patients with IPF is between 32 and 85%, and PH seems to develop over time in most patients with IPF (17, 20, 22–24). Although most of the PH is moderate, pressures of systemic levels can be found in patients with IPF. Increased levels of brain natriuretic peptide may predict PH in patients with IPF (25). Although early diagnosis of PH is important, the overlapping main symptoms—shortness of breath and exercise limitation—make it difficult to detect PH in these patients (3). Typical physical findings associated with PH are usually detected when the pulmonary vascular disease is advanced. To be effective in the treatment of IPF-associated PH, we need to understand how PH develops and progresses in IPF. The pathobiology of PH in IPF is incompletely understood and research groups have only recently started to focus on the vascular aspects of chronic lung fibrosis. The results of recent clinical studies do not support the hypothesis that the predominant mechanisms for the development of PH in IPF are hypoxic vasoconstriction and pulmonary capillary loss after scar tissue accumulation: the presence of PH cannot be explained in all patients with IPF by hypoxemia or degree of lung function reduction (23, 26–28). Considering the complex pathophysiology of IPF, it seems likely that the biological processes underlying fibrosis progression are also involved in the vascular remodeling and PH (20). Our group has recently shown, in a model of experimental pulmonary fibrosis (PF), that the development of fibrosis and PH are closely connected (29). Here, we review the reported data in this field and discuss findings in other lung diseases associated with interstitial fibrosis in the context of IPF to develop an overall concept that integrates the factors and the processes involved in the development of PH in PF.

2

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011

PULMONARY VASCULAR DEVELOPMENT

VASCULAR REMODELING IN IPF

Wound repair or fibrogenesis in the lung is associated with (or caused by) the reactivation of pathways and programs that have been involved in lung development (30). To understand the interactions between vascular and interstitial processes in PF, it is necessary to revisit how the pulmonary vasculature and the parenchyma relate to each other during lung embryology. The pulmonary vascular system develops in close proximity to and synchronous with airway and alveolus formation (31). During the embryonic phase of lung development, lung buds appear and branch into the surrounding mesenchyme. These buds are supplied by capillaries that connect to the aortic sac via PAs, and to the prospective left atrium of the heart via pulmonary veins (31–33). Cartilage, glands, and smooth muscle cells (SMCs) develop and epithelium differentiates in the airway walls during the pseudoglandular stage. In addition, this phase is also responsible for establishing preacinary airways, arteries, and veins (31). During the canalicular and saccular stages, further branching occurs into respiratory airways, which are accompanied by arteries and veins. Endothelial and epithelial basement membranes fuse, thereby facilitating gas exchange. A necessary requirement for the formation of an effective gas exchange is the close proximity of capillaries and epithelium after the differentiation of cuboid type 2 alveolar epithelial cells (AECs) into thin type 1 cells, and a decrease of the surrounding mesenchyme. AEC2 starts surfactant production around 24–25 weeks of gestation, alveoli appear, and cup-shaped alveoli with double capillary loops develop, thus increasing the area of the alveolar capillary bed. Airways, arteries, and veins increase in size (31). Some studies suggest that blood vessels in the lung originate from existing vessels through angiogenesis (proliferation and migration of existing endothelial cells [ECs]). Other studies have produced evidence that angiogenesis may only play a role for proximal arteries, whereas vasculogenesis (new development and proliferation of EC) may be important for the development of peripheral arteries and capillaries, which later connect to the proximal arteries (32–36). Several angiogenic growth factors have been implicated in lung vascular development. The most widely investigated is vascular endothelial growth factor (VEGF), which is involved in both angiogenesis and vasculogenesis. Epithelial cells seem to be a predominant source of VEGF during lung development, which suggests that airway and alveolar epithelium are crucial for the generation of the entire pulmonary vasculature (37–40). Impaired VEGF expression and signaling do not only have detrimental effects on vascular development and maintenance, but also on the structure and integrity of the whole lung (40–42). Other angiogenic factors, such as angiopoietin (Ang)-1, are also involved in blood vessel formation (43). In addition, tightly regulated levels and activity of growth factors, such as TGF-b, are important for coordinated vascular development, as illustrated by the findings that mice overexpressing TGF-b1 during lung development show impaired epithelial differentiation and decreased capillarization (44). The three main sources of vascular SMCs (VSMCs) in the developing lung may explain some mechanisms of VSMC generation found later in PH pathophysiology: (1) bronchial SMCs form the inner layer of VSMCs in the arteries accompanying the penultimate airways (31, 33); (2) fibroblasts surrounding the arterial wall differentiate into VSMCs after the release of chemoattractant growth factors, such as platelet-derived growth factor (PDGF) or epidermal growth factor from the endothelium in response to Ang1 secreted by mesenchymal cells (45); (3) VSMCs can also arise from the inside of the vessel through endothelial-to-mesenchymal transition (46). Similar transition mechanisms might be involved in the pulmonary arterial remodeling in the adult, fibrotic lung.

It is not surprising that vascular remodeling in a patchy, heterogeneous disease such as IPF is not homogeneous, and that the vascular density varies in IPF lungs. Fibrotic areas have less blood vessels, but adjacent, nonfibrotic tissue seems highly vascularized (Figures 1B and 1C) (47–49). Fibrotic regions contain apoptotic, but also proliferating, ECs, eventually resulting in anastomoses between alveolar capillaries and pulmonary veins with an aberrant vascular architecture (Figures 1C–1H) (47, 50, 51). These changes result in a reduction in the cross-sectional vascular area throughout the lung. It appears that, in human IPF, vascular rarefaction develops not only after expansion of scar tissue, but also due to an imbalance between angiogenic and angiostatic factors: the reduced expression of angiogenic factors, such as vascular endothelial growth factor (VEGF), is paralleled by elevation of angiostatic molecules, such as pigment epithelium– derived factor (PEDF) (47, 48). Although there is some controversy regarding up-regulated proangiogenic and down-regulated angiostatic chemokines in IPF, we were able to directly link, in our animal model of PF, decreased VEGF levels to EC apoptosis and PH (29, 52–55). EC apoptosis could be also induced by elevated oxidative stress (3, 56). It is interesting in the context of EC apoptosis in IPF that reduced numbers of circulating endothelial progenitor cells (EPCs) were found in IPF. This suggests that either reduced availability or increased pulmonary homing to sites of injury, possibly associated with dysfunction of EPCs, could be involved in endothelial injury in IPF (57, 58). Figure 2 shows a synopsis of mechanisms that are putatively involved in EC apoptosis in IPF. The histopathologic changes in PAs of UIP lungs show a broad range of structural alterations, from isolated thickening of the smooth muscle layer and proliferative intima lesions, to complete occlusion of the vessel by scar tissue and plexiform lesions (Figure 1D). The extent of these changes increases with advancing fibrosis of the surrounding tissue (59). We were able to correlate fibrosis, TGF-b activity, angiostatic environment, EC apoptosis, and PA muscularization in our animal model of PF. Based on these findings, we suggest that VSMC growth factors may be released from apoptotic EC in fibrotic regions, and that mediators from the surrounding fibrotic tissue also contribute to augmented PA muscularization (29, 60). Such molecules, likely involved in both processes, are TGF-b, angiotensin (AT) II, endothelin (ET)-1, and PDGF (20, 60–62). Again, oxidative stress could also play a role for VSMC and fibroblast proliferation, and thereby contribute to pathological changes in all three vascular layers of the PA (3, 56). A number of other important mechanisms will probably contribute to vascular remodeling and PH in IPF, such as endothelial dysfunction, with reduced release of vasodilators and inhibitors of VSMC proliferation, notably nitric oxide (NO) and prostacyclin (29, 60, 61, 63–65). Reduced vasodilator release and enhanced secretion of vasoconstrictors, such as ET-1, thromboxane A2, and AT II, points toward a possible component of vasoconstriction, thus contributing to increased PAP and remodeling (60, 61). Conceptually, these mechanisms are summarized in Figure 3. The same mediators may also promote reduction in cross-sectional vascular area through thrombotic obstruction of vessels (3). The complex biochemistry in the fibrotic tissue with release of growth factors, such as TGF-b, may also result in endothelial-to-mesenchymal transition, which might contribute to the myofibroblast accumulation in intima fibrosis or VSMC growth during media thickening (66–69). Inhibition of bonemorphogenic protein–induced mesenchymal cell apoptosis through up-regulation of one of their antagonists, gremlin, might also add to vascular remodeling, similarly to findings in idiopathic PAH (70–72). In addition, mast cells occur at sites of epithelial

Translational Review

3

Figure 2. Synopsis of potential mechanisms involved in EC apoptosis in idiopathic pulmonary fibrosis (IPF). Changes in expression or activity of various factors may finally culminate in EC apoptosis in IPF. Straight lines: evidence, dotted lines: likely or controversial. EPC, endothelial progenitor cell.

Figure 1. Vascular remodeling in usual interstitial pneumonia (UIP) (A) shows UIP lung with fibroblastic focus (FF), hyperplastic epithelial cells (arrows), and matrix deposition (hematoxylin and eosin). (B and C ) Normal vascular density in nonfibrotic area (B) and reduced vascular density in the fibrotic region of UIP lung (C ). CD31 immunohistochemistry. (D) Intimalesion (I) and media (M) thickening in a bifurcating pulmonary artery (PA) in UIP lung. Closed arrow demonstrates inner elastic lamina; open arrow demonstrates outer elastic lamina. The ‘‘A’’ demonstrates adventitia fibrosis and communication with the surrounding fibrotic tissue (image of Elastica van Gieson-stained section). (E ) Double immunofluorescence staining demonstrates apoptotic endothelial cell (ECs) (arrows) inside a PA. Red, terminal deoxynucleotidyl transferase dUTP nick end labeling ( TUNEL); green, CD31; blue, nuclear counterstaining with 49,6-diamidino-2-phenylindole (DAPI). (F ) Intimafibrosis with bud-like partial lumen occlusion (arrows), likely developed through angioproliferation (image of Elastica van Gieson-stained section). Scale bars, 100 mm (A–C ), 50 mm (D and F ), and 20 mm (E ).

injury, and they might play a role in either repair or disease progression via tryptase, histamine, or ET (73). Mast cells have been found around PAs in both human and experimental PH, but experimental studies did not result in a definitive concept about mast cells contributing to vascular remodeling or mediating a protective mechanism (74–76). Future investigation will need to uncover additional potential mechanisms, such as serotonin levels, inflammation, and alterations of Ca21 and K1 currents in SMC dysfunction, as well as activity of Rho guanosin triphosphatases, due to their relevance in other PH classes (77). Figure 4 summarizes possible mechanisms that are likely to contribute to PA wall remodeling.

VASCULAR REMODELING IN OTHER ILDS Systemic Sclerosis

Systemic sclerosis (SSc or scleroderma), an unusual autoimmune disease without known cause, is defined by skin fibrosis, Raynaud’s phenomenon, and possible involvement of other organ

systems, such as lung, kidney, and the musculoskeletal, cardiovascular, and gastrointestinal system (78). Pulmonary pathology is the predominant cause of death in SSc, and the main findings are ILD and PH (79). Nonspecific interstitial pneumonia is the preponderant histologic phenotype, but some patients with SSc also have UIP (80, 81). PH can be found with or without association to ILD, and has a prevalence of 45% in SSc-ILD (28). Lower estimates around 10% have been shown by other groups (82, 83). Endothelial apoptosis has been identified as the initiating event of the pathological processes leading to SSc (84). Circulating autoantibodies, including anti-fibrillarin and anti-EC antibodies, represent the autoimmune component of SSc, and these autoantibodies are likely responsible for EC injury and apoptosis (85–87). The damage leads to recruitment of inflammatory cells with predominantly CD81 T cells and a T helper (Th) type 2 cytokine pattern (e.g., monocyte chemoattractant protein-1, CXC ligand (CXCL)8 and regulated upon activation, normal T cell expressed and secreted) (88–91). Elevated levels of profibrotic growth factors, such as TGF-b, PDGF, CTGF (connective tissue growth factor), or ET-1, result in mesenchymal cell proliferation and ECM accumulation (92–96). The initial stage of SSc is characterized by inflammation and an increased angiogenic response, with elevated levels of VEGF in the beginning. Later on, reversible vasospasm and reduction in capillary density are found, eventually culminating in obliterative vasculopathy with intima proliferation and adventitia fibrosis (77, 97). There is an overall antiangiogenic environment in advanced SSc, characterized by reduced capillary density, which could contribute to PH as in IPF (51, 97). Proliferative intima and media lesions might be a consequence of EC apoptosis, secondary mediators, and local inflammation (77). It is interesting that vascular and fibrotic processes can influence each other via production of profibrotic growth factors (98). Anti-fibroblast antibodies could promote vascular remodeling even without EC injury (99). There is a variety of additional potential mechanisms that might be involved in the development of PAH, which are more or less related to the autoimmune component of the disease. Potentially involved mechanisms include various other circulating autoantibodies, such as anti-PDGF receptor, anti-centromere, anti–topoisomerase 1, or anti–matrix metalloproteinase (MMP) 1–3 antibodies (96). The investigation of PH in SSc-associated ILD will generate new concepts regarding the close relationship and interaction between inflammation and autoimmunity, and interstitial and pulmonary vascular remodeling. In addition, we are inclined to believe that these processes argue in favor of similar mechanisms underlying PH in different forms of ILD. Nevertheless, the complex pathobiology of SSc with potential involvement of multiple extrapulmonary organs will make it

4

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011

difficult to specifically treat the pulmonary vascular disease, and makes it understandable that therapy of PAH and ILD in SSc will have to be also a therapy of the systemic autoimmune disease. This is clearly illustrated by the potential involvement of the myocardium in SSc, which can be found in 15–35% of patients with SSc (96, 100). Inflammation, vascular lesions, and cardiac fibrosis may lead to impaired cardiac function, and cardiac dysfunction has been shown to occur in the right ventricle, but also in the left ventricle, in SSc (100–102). Sarcoidosis

Sarcoidosis is a systemic inflammatory disease characterized by noncaseating granulomas. The pathology is almost always found in mediastinal lymph nodes and the lungs (103). An unknown antigenic stimulus activates a sequence of immune responses, followed by T cell and macrophage activation, and a Th1-type immune response (103). Typical findings are accumulation of inflammatory cells (T cells) in the lung due to locally increased cytokines and chemokines, such as monocyte chemoattractant protein–1, CXCL8, or regulated upon activation, normal T cell expressed and secreted (103, 104). The resulting granulomatous inflammation can improve without persistent damage to the lung, but can also progress to PF (Figure 5A). When PH is detected in patients with sarcoidosis, then PH is typically found together with already advanced PF. Similar to SSc, some patients may have PH without underlying ILD. Nevertheless, PH is a predictor of increased mortality in all of these patients (27). The prevalence of PH in sarcoidosis varies between 5.7 and 73.8% in advanced disease (28). Several mechanisms seem to be involved in the development of PH in sarcoidosis: the granuloma tissue itself is rarely capillarized (Figure 5B). Due to their anatomical localization, PAs are frequently affected by the granulomatous inflammation with occlusion, perivascular fibrosis, or vasculitis (Figures 5C and 5D) (105–107). Compression or invasion of pulmonary veins by inflammatory cells may also result in hemodynamic changes similar to pulmonary veno-occlusive disease (105–109). Additional potential mechanisms are direct mechanical compression of PAs by enlarged lymphatic tissue, luminal obstruction by plexiform lesions, hypoxic vasoconstriction, and diastolic or systolic dysfunction due to myocardial sarcoidosis (105). Bronchoalveolar lavage fluid (BALF) of patients with sarcoidosis shows enhanced levels of ET-1. Therefore, ET-1–induced mesenchymal cell proliferation is regarded as important for the development of vascular remodeling and PH in sarcoidosis (110). Although reduced BALF VEGF levels and changes in gene activity of apoptosis regulators have been shown, there is currently no clear evidence regarding a role of angiogenesis or EC injury in the development of sarcoidosis-associated PH (111–113). Therapeutic considerations in sarcoidosis do not only include immunosuppression and specific PH therapy, but sarcoidosis treatment also has to target various extrapulmonary manifestations of sarcoidosis, including myocardial sarcoidosis, if existent (114).

CONTRIBUTION OF SPECIFIC FACTORS This paragraph summarizes the potential contribution of selected factors to vascular remodeling in IPF, under consideration of a possible relevance for disease monitoring and therapy. Nevertheless, it has to be kept in mind that blood or BALF levels do not necessarily reproduce the expression or activity of the specified molecules within the pulmonary vascular system, especially in the heterogeneously altered lung of patients with IPF. VEGF

VEGF is a major angiogenic growth factor, and is essential for sprouting and migration of EC during angiogenesis, but it also has

important functions for the maintenance of the vascular system: it is important for EC survival and proliferation, and inhibition of VEGF signaling in the adult healthy lung results in EC apoptosis and severe damage to the alveolar structure with the development of emphysema (115, 116). VEGF transcription is mainly induced by hypoxia-inducible factor–1a, which accumulates in the cells under hypoxia, but is also increased by TGF-b1 (117). Different receptors for VEGF have been characterized, but VEGF receptor 2 mediates vascular homeostasis and survival. Pulmonary VEGF expression can be detected in macrophages, epithelial cells, and ECs, but also in mesenchymal cells (118). In addition, a significant percentage of lung VEGF is matrix bound (118). Production of NO via endothelial NO synthase is required for many of the endothelial effects of VEGF, but NO and prostacyclin synthesis are also required for vasodilation (115). In human IPF, VEGF has been shown to be reduced in BALF and fibrotic regions of the lung, especially within fibroblastic foci (47, 48, 119). In these areas, EC apoptosis is correlated with reduced vascular density and decreased capillary branching (47, 48, 51). We have shown that the local VEGF deficit in fibrotic tissue is directly involved in vascular rarefaction, PA remodeling, and PH through apoptosis of ECs (29). Interestingly, VEGF can also promote fibrosis through angiogenesis and collaboration with TGF-b, resulting in enhanced ECM production of fibroblasts (29). In addition, VEGF inhibition can reduce fibrosis in the bleomycin model of PF (120). The bleomycin model is widely used to investigate disease mechanisms in PF, and to evaluate new treatment options (121). Bleomycin induces epithelial injury in the rodent lung, with subsequent inflammation and transition to fibrosis after roughly 8–10 days (122). Studies investigating VEGF and angiogenesis almost exclusively used the bleomycin model, which differs from IPF, especially in terms of microvascular changes: as opposed to capillary loss found in IPF, bleomycin-induced fibrosis shows neovascularization (120). VEGF is significantly up-regulated during the inflammatory phase in this model, and inhibition of VEGF results in reduced fibrosis, most likely due to reduced leukocyte trafficking and protein leakage (120). In contrast to our recent work, most publications investigating angiogenesis inhibition as a therapeutic option for PF have not taken into consideration that this approach can also negatively affect vascular maintenance and promote the development of PH. The vasoprotective effects of VEGF have also been documented by several PH studies (123–128). The current literature suggests two possible roles for VEGF in the clinic: (1) VEGF has potential as a therapeutic target due to its high angiogenic potential, but systemic therapy with VEGF also bears significant potential side effects—VEGF increases vascular permeability with the results of edema and hemorrhage, activates mesenchymal cells, and can promote the growth of occult tumors (29, 129). (2) VEGF inhibition can be useful to limit local angioproliferation, but it could also have detrimental effects on vascular integrity by promoting EC apoptosis. To find a satisfactory way out of this dilemma, compartment- or cell-specific treatment strategies need to be evaluated (e.g., by using genetically modified EPCs, as already investigated in other forms of PH [57, 58]). In addition to therapeutic considerations, VEGF levels have been frequently investigated in BALF and blood, but the results have so far been conflicting, and demonstrate that a factor might well be very important in pathogenesis, but not necessarily suitable as a potential biomarker (119, 130). PEDF

Decreased VEGF expression in the fibrotic tissue is accompanied by elevated levels of PEDF, predominantly in the epithelium overlying fibroblastic foci and the foci themselves. In the normal

Translational Review

Figure 3. Concept for the development of pulmonary hypertension (PH) in IPF/UIP. Epithelial injury with subsequent production of different mediators is the hallmark of fibrosis induction. These mediators induce fibroblast activation with extracellular matrix (ECM) deposition, which leads to fibrosis. Some of these mediators (e.g., TGF-b) also activate ECs and, as a result of a shift in favor of increased angiostatic (e.g., pigment epithelium–derived factor [PEDF]) and reduced angiogenic factors (e.g., vascular endothelial growth factor [VEGF]), EC apoptosis results. Apoptotic ECs produce less vasodilators, but more vasoconstrictors, which leads to augmented vasoconstriction of smooth muscle cells (SMCs). At the same time, EC apoptosis gives rise to a reduction in vascular density, but also to enhanced production of vascular SMC (VSMC) growth factors, which is important for remodeling of mesenchymal cells in the PA wall. However, EC apoptosis also results in proliferation of apoptosisresistant ECs or endothelial progenitors, with the consequence of angioproliferative lesions, including plexiform lesions. Another component of PA wall remodeling is the release of additional factors generated in the fibrotic tissue, which contribute to PA wall remodeling from the outside of the vessel.

lung, PEDF is found in airway and AECs, ECs, and mesenchymal cells (48). PEDF shares structural homology with members of the serpin family of protease inhibitors, but does not seem to have inhibitory activity on its own (131). Among the various actions of PEDF, we find a very strong angiostatic component and inhibition of VEGF-induced angiogenesis, as well as induction of EC apoptosis and inhibition of fibroblast growth (132–136). Recent studies have identified a putatitive 60-kD PEDF receptor

Figure 4. Synopsis of potential mechanisms involved in PA wall remodeling in IPF. Increased expression or activity of different mediators and other mechanisms may contribute to different aspects of PA wall remodeling in IPF. Straight lines, evidence; dotted lines, possible mechanism.

5

Figure 5. Vascular remodeling in pulmonary sarcoidosis. (A) A noncaseating granuloma in the lung of a patient with sarcoidosis. Note the density of the tissue with cellular infiltration, hyperplastic epithelium, and fibrosis (hematoxylin and eosin). (B) Vascular density in a granuloma of a sarcoidosis lung. Note the massively reduced vascular density inside the dense granuloma tissue (CD31 immunohistochemistry). (C ) PA wall remodeling inside a sarcoidosis granuloma, demonstrating extensive intima (I) hypertrophy, moderate media thickening (M), and possible infiltration of the vessel wall by the granulomatous process from the outside at the bottom of the image. Closed arrow, internal elastic lamina; open arrow, external elastic lamina. Note that the vessel adventitia is not distinguishable from the surrounding granuloma tissue (image from Elastica van Gieson-stained section). (D) Intima and media proliferation in a PA inside a sarcoidosis granuloma. Closed arrows, proliferating ECs; open arrow, proliferating VSMC (proliferating cell nuclear antigen immunohistochemistry). Scale bar, 100 mm (A and B), 50 mm (C and D).

on EC, the nonintegrin laminin receptor, and also an 80-kD receptor on neurons, PLA2/nutrin/patatin-like phospholipase domain–containing 2 (137–142). The PEDF molecule contains two corresponding functional epitopes: a 34-mer peptide, and a 44-mer peptide (143). The 44-mer peptide binds to the 80-kD receptor on neurons and mediates neuroprotection (144). The 34-mer peptide is responsible for the anti-angiogenic effects of PEDF, and it seems to bind to the 60-kD receptor on ECs (141, 143, 144). The various functions of PEDF are highly regulated at the post-translational level, through phosphorylation at different amino acid residues and several binding sites for ECM components (e.g., collagen 1, heparin, or hyaluronan [145–150]). These binding sites induce conformational changes (heparin), or are necessary for the antiangiogenic activity of PEDF (collagen 1) (148, 149). However, association with the ECM might be not only a mechanism to regulate the activity of PEDF, but could also

Figure 6. Sequence of CXCL8. Amino acid sequence of CXCL8, a ELR1 CXC chemokine, demonstrating the ELR motif (red ) directly in front of the first of four highly conserved cysteine residues (blue), demonstrating disulfide bonds (green) between the first and third and the second and fourth conserved cysteine residues. The sequence is shown in two rows for the largest mature CXCL8 form (77 amino acids), according to Ref. 280.

6

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011

explain the localization of PEDF to the ECM-rich fibrotic tissue (151). Other explanations for local PEDF accumulation in fibrosis are increased production, either from damaged epithelial cells or from activated myofibroblasts, as well as decreased degradation of PEDF, due to reduced MMP-2 and MMP-9 activity in fibrotic tissue paralleled with enhanced expression of tissue inhibitor of metalloproteinases (29, 48, 152, 153). Our data in the AdTGF-b1 model indicate that PEDF is early up-regulated in fibrotic areas (29). PEDF is likely involved in EC apoptosis through inhibition of VEGF, but the overall function of PEDF in fibrotic lung disease is unclear. Production in injured epithelial cells may be an attempt to reduce fibroblast activity and local inflammation (134, 154–156). This makes sense when PEDF is regarded as a protective factor, as shown previously in kidney and brain (157–160). However, the strong antiangiogenic effect of PEDF with subsequent EC apoptosis is likely detrimental in a highly vascularized organ such as the lung. PEDF inhibition (e.g., by antibodies or soluble receptor fragments), seems to be a promising therapeutic approach, once it has been clarified which role PEDF has in the development or progression of PH. Careful studies need to evaluate whether PEDF inhibition might also cause negative effects, such as increased fibrogenic activity, similar to increased VEGF activity (29). Further research is also needed to evaluate whether PEDF levels in blood or BALF can be used as a potential predictor of disease activity and, therefore, prognosis. Such studies have yielded interesting results in the field of oncology (with reduced PEDF levels as a negative outcome predictor), and recently also in patients with heart failure (161– 164). TGF-b

The TGF-b family in mammals consists of three isoforms. All TGF-bs have a homologous sequence and similar functions during wound repair and tissue remodeling (165). They are major profibrotic growth factors involved in ECM turnover and mesenchymal cell activity. Most lung cells express TGF-b, and there is evidence of signaling. TGF-b also has important immunesuppressive functions during inflammation. TGF-b is mainly produced by macrophages, epithelial cells, and fibroblasts. Secreted TGF-b requires activation by cleavage of its latency-associated peptide. Two receptors, types I and II, bind TGF-b, and distinct intracellular pathways, such as the Smad2/3 pathway, mitogenactivated protein kinases, phosphoinositide 3-kinases, or Rho GTPases, mediate its intracellular actions. Mainly, the Smad pathway is relevant in tissue fibrosis (166). Importantly, signaling through the Smad2/3 pathway results in myofibroblast activation, characterized by elevated gene expression of ECM components and profibrotic mediators (166). TGF-b inhibits proliferation and/or function of various immune cells, such as T lymphocytes, natural killer cells, B lymphocytes, macrophages, dendritic cells, and granulocytes (167). In addition, TGF-b is also responsible for the induction of Forkhead box P3 (Foxp3) in CD41 CD252 Foxp32 precursors of regulatory T cells outside of the thymus, but also for the maintenance of Foxp3 expression and the regulatory function of regulatory T cells (167–171). However, TGF-b can also support inflammation (e.g., through differentiation of Th17 cells [172]). TGF-b1 is the most commonly investigated isoform, and has concentration-dependent effects on angiogenesis. At lower doses, TGF-b1 activates ECs through enhanced expression of angiogenic growth factors and proteinases (173). At high doses, it decreases EC growth or induces apoptosis, promotes basement membrane formation, and induces differentiation and recruitment of VSMCs (174, 175). Elevated TGF-b1 levels in fibrotic areas of IPF lungs may contribute to local EC apoptosis, and thereby vascular rarefaction, but also to locally increased mus-

cularization of PAs (48, 176–178). In summary, TGF-b has effects on a large number of different cell types, suggesting that a close coordination of TGF-b activation is of crucial importance to maintain tissue homeostasis (167). Therapeutic inhibition of TGF-b and its pathway appears to be very useful to target the fibrotic process, but also the pulmonary vascular disease, as demonstrated by in vivo studies (178, 179). The central importance of TGF-b would suggest that TGF-b could also be a valuable biomarker in IPF, but TGF-b measurements in BALF are highly variable, which can limit the value of this marker, and indicates the need for larger studies to establish more reliable fluid levels (180, 181). Fibroblast Growth Factor–2

Fibroblast growth factor (FGF)–2 is a heparin-binding FGF with important functions in angiogenesis (182, 183). It signals through the high-affinity tyrosine kinase FGF receptors, FGFR1 and FGFR2, on ECs, and induces EC proliferation and migration (184). The main intracellular pathways that are activated are mitogen-activated protein kinase and protein kinase C (185). In addition to the effects on EC, FGF-2 also contributes to degradation of ECM through induction of MMPs and plasminogen activator expression as a preliminary step in angiogenesis (182). FGF-2 is reduced in fibroblastic foci of IPF lungs, but is expressed in the surrounding epithelium, similarly to VEGF (186). Although reduced levels of FGF-2 in fibrotic tissue are consistent with decreased capillary density, and suggest a potential contribution to EC apoptosis and vascular rarefaction, FGF-2 might also be involved in VSMC growth, similarly to human PH and experimental animal PH models, suggesting that FGF-2 might have a role in vascular remodeling in PF (187, 188). The relevance of FGF-2 in IPF is currently unclear: FGF-2 could help to reduce EC apoptosis, but the overall effects of FGF-2 on fibrotic process and PA remodeling warrant further investigation, as does the potential of FGF2 as a biomarker. CXC Chemokines

Chemokines of the CXC family have four highly conserved cysteine residues, with the first two separated by a nonconserved amino acid, and a second, three-amino-acid motif determines the angiogenic activity. CXC chemokines with the three-amino-acid sequence, Glu-Leu-Arg (ELR), right ahead of the first cysteine amino acid, are proangiogenic ELR1 CXC chemokines, whereas ELR2 CXC chemokines have antiangiogenic properties (55, 189, 190). A representative sequence is shown in Figure 6. Relevant ELR1 CXC chemokines are CXC ligand (CXCL) 1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, and CXCL8 (55, 189, 190). Interactions with various other factors are involved to promote angiogenesis (e.g., VEGF induces CXCL8 secretion from ECs, and thereby perpetuates angiogenesis [55, 191]). ELR1 CXC chemokines bind to CXC receptor (CXCR) 1 and CXCR2 (192). These receptors are expressed by ECs, although CXCR2 is involved in the major angiogenic effects in microvascular lung ECs in vitro (55, 192–195). A CXC chemokine with profibrotic effects via stromal cell or mesenchymal progenitor recruitment is CXCL12, which also has angiogenic effects through its receptor, CXCR4 (55, 196). The most frequently found ELR2 CXC chemokines are CXCL4, CXCL9, CXCL10, and CXCL11 (55). These chemokines represent a link between angiostasis and immune function, because they are induced by Th1-produced cytokines (55). Their receptor, CXCR3, is important for recruitment of leukocytes and inhibition of angiogenesis (55, 197). In human IPF, the balance between ELR1 CXC chemokines and ELR2 CXC chemokines is shifted toward ELR1 CXC

Translational Review

chemokines, which indicates a proangiogenic chemokine environment (53, 54, 198, 199). Depletion of ELR1 CXC chemokines or administration of ELR2 CXC chemokines in the bleomycin model of lung fibrosis results in a reduction of both angiogenesis and fibrosis (52, 200, 201). In apparent contrast, Ebina and colleagues (47) demonstrated decreased CXCL8 in fibrotic areas of IPF lungs, similar to VEGF. These contradictory results likely reflect the extensive heterogeneity of human UIP in terms of angiogenesis with evidence of EC proliferation and augmented capillary density in ‘‘interface’’ regions directly adjacent to the fibrotic tissue, where EC apoptosis and vascular rarefaction dominate (47, 51). The exact function of CXC chemokines in IPF and PH pathophysiology (counterregulatory process, contribution to PA remodeling, response to epithelial injury or inflammation) needs to be elucidated. This information is critical in deciding whether and which of the CXC chemokines might be useful as therapeutic targets or biomarkers in IPF-related PH. PDGF

PDGF-A and PDGF-B are best characterized and mainly investigated, whereas PDGF-C and PDGF-D have only been recently described (202). PDGF expression is found in many different cell types, including fibroblasts, epithelial cells, and platelets (182). PDGF is biologically active in the form of heteroor homodimers of the A and B chains, and binds to complexes of a and b subtypes of PDGF receptors (202). PDGF is responsible for chemoattraction and proliferation of mesenchymal cells of the vessel wall, such as pericytes and VSMCs. Pericytes are a–smooth muscle actin1 mesenchymal cells surrounding capillary structures, which are required for angiogenesis and microvascular stability (182, 203, 204). In early IPF, PDGF is not only found in macrophages, fibroblasts, and AEC2, but also in EC and VSMC, suggesting a contribution of PDGF to vascular remodeling (205). In contrast, vessels in advanced IPF do not exhibit PDGF expression. In addition to fibroblast proliferation and migration, PDGF seems to be involved in PA wall remodeling through activation of VSMCs and fibroblasts, which results in enhanced muscularization, as well as intima and adventitia fibrosis (206). This hypothesis is supported by experiments in models of PH (207). Therefore, PDGF inhibition could evolve as a useful target to reduce both, fibrogenesis and vascular remodeling. In addition, serum or BALF levels of PDGF could be predictors of outcome, but future studies are needed to establish the relevance of PDGF. Endostatin

The angiostatic endostatins are generated via cleavage of collagen XVIII at its protease-sensitive hinge region, and have molecular weights of 20–30 kD. Various pathways are involved in the angiostatic activity of endostatin, such as inhibition of VEGF receptor 2 and MMP-2 (208). The precursor molecule, collagen XVIII, is found in alveolar capillary and epithelial basement membranes (209, 210). IPF lungs contain elevated mRNA levels of collagen XVIII. Increased proteolysis of alveolar collagen XVIII by different enzymes, such as elastase, cathepsin, and different MMPs, seems to be responsible for the elevated levels of endostatin in IPF serum and lung (211, 212). It has been suggested that endostatin is produced by injured epithelial cells, which again, similarly to PEDF, may link epithelial injury and vascular rarefaction in IPF. Experimental data from hypoxic PH have implicated endostatin in PA vascular remodeling (213). Two main conclusions can be drawn: (1) inhibition of endostatin could reduce the consequences of epithelial injury on the pulmonary vasculature, and be, therefore, a useful target for future therapy; and (2) serum endostatin levels may represent a valuable param-

7

eter to evaluate the extent of pulmonary epithelial and vascular injury, but also disease progression. ET-1

The most important vascular functions of ET-1 are vasoconstriction and growth of VSMCs (61). This protein signals through interaction with ETA and ETB receptors, both G-protein coupled receptors (214). Expression of ET-1 is widely found throughout the lung, including in ECs, airway, and alveolar epithelium (215, 216). The levels of ET-1 seem to be increased in the lungs of patients with IPF, with the main expression localized to ECs and epithelial cells within fibrotic areas (216, 217). ET-1 has important profibrotic actions, and is involved in PA wall remodeling in different forms of human and experimental PH, as well as of lung fibrosis models (218–220). The extent of PH correlates directly with ET-1 levels in the peripheral blood of patients with IPF (221). It is likely that ET-1 is involved in remodeling of all three artery layers in IPF, because ET-1 could be released by injured ECs and epithelial cells within the fibrotic areas, and thereby contribute to intima lesions and VSMC growth from the inside (release from ECs) and from the outside (secretion from epithelial cells) (29, 222). Although experimental research suggests that ET-1 would be a central target to treat IPF and associated PH, the current clinical data regarding ET receptor inhibition in IPF show only very limited success. There are only limited data available regarding the potential of ET-1 as a biomarker, but the results so far indicate that urine rather than plasma levels could be used (223). AT II

The octapeptide, AT II, mediates the main effects of the renin– AT system. Renin and the AT-converting enzyme, which is prominently expressed in the lung capillary EC, cleaves angiotensinogen into its active form, AT II. AT II is not only a very potent vasoconstrictor, but is also involved in VSMC growth and proliferation (224). AT II is likely involved in the pathophysiology of PH, taking into account elevated expression of AT-converting enzyme in ECs of patients with PH; however, successful reduction of experimental PH through AT II inhibition has not been found in patients with PAH (224, 225). In addition, a putative, important role has been shown for AT II in epithelial cell apoptosis and fibrosis in both human IPF and experimental lung fibrosis (226, 227). Enhanced AT II in the fibrotic tissue may contribute to the development of PH through vasoconstriction and PA wall remodeling (60, 228). AT inhibition might be a valid therapeutic approach in IPF-associated PH, but the overall relevance needs to be established through ongoing research. AT II or AT-converting enzyme levels could be used as biomarkers, but the prognostic relevance needs to be established (229). Ang-1/Ang-2/Tie-system

The Ang–Tie ligand–receptor system has important functions in regulating vascular integrity and maintenance of a quiescent EC state (230). The system consists of two tyrosine kinase receptors, Tie-1 and Tie-2, and four ligands, Ang-1, Ang-2, Ang-3, and Ang4 (230, 231). Research has mainly focused on Ang-1 and Ang-2. The receptors have almost exclusively been detected on EC and hematopoietic stem cells (232–235). Ang-1 is constitutively expressed by cells of mesenchymal lineage, such as pericytes, VSMC, fibroblasts, and some tumor cells (236–238). The main role of Ang-1 is to keep ECs in a quiescent state, a function that requires signaling through the endothelial receptor, Tie-2 (230). Ang-1/Tie-2 interactions mainly activate the antiapoptotic Akt pathway (239). In contrast, Ang-2 expression is almost exclusively

8

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011

found in ECs, and is very low in quiescent ECs, but it dramatically rises once ECs are activated (230, 240, 241). The functions of ECderived Ang-2 are highly context dependent: Ang-2 promotes angiogenesis in concert with VEGF, but results in EC apoptosis and subsequent vessel regression without VEGF (230, 242). Current knowledge about the Ang–Tie system in IPF is very limited, but studies from kidney fibrosis show that Ang-1 therapy induces increased capillarization, most probably via stabilization of vessels (243, 244). There is some controversy about how Ang-1 effects experimental PH in rodents: although cell-based gene transfer of Ang-1 improved survival and pulmonary hemodynamics in monocrotaline-induced PH, Ang-1 overexpression induced PH in healthy rats (245, 246). Nevertheless, the Ang-1/ Ang-2/Tie-2 system may have an important impact on understanding the complex processes leading to vascular rarefaction in lung fibrosis, and to discovering promising new treatment options. In IPF, activation of EC with induction of Ang-2 expression might contribute to EC apoptosis in the context of local angiogenesis inhibition in the fibrotic tissue. On the other side, reduced Ang-1 release from dysfunctional VSMC could also destabilize ECs locally, thereby contributing to EC damage. Therefore, supplementation of Ang-1, or inhibition of Ang-2 could help to restore endothelial integrity in IPF, possibly without the severe side effects of VEGF. Although Angs might be valuable biomarkers for the pulmonary vascular disease in IPF, there are currently no data available that would allow a statement. Angiostatin

Angiostatin is produced via proteolysis of plasminogen by a macrophage-derived metalloelastase and other MMPs, or by reduction of plasmin (247–249). Angiostatin consists of the first four kringle domains of plasmin. The kringle is a triple loop structure linked by three pairs of disulfide bonds (250). Angiostatin inhibits proliferation, induces apoptosis, and prevents EC migration in vitro (251, 252). The molecular mechanisms include activation of focal adhesion kinase, ceramide generation, RhoA activation, and inhibition of an a/b ATP synthase on the surface of ECs (252–256). Elevated levels of platelet angiostatin have been reported in some patients with idiopathic PAH (257). Pulmonary overexpression of angiostatin aggravated PH in a model of chronic hypoxia, believed to be because of decreased vascularization (258). Angiostatin may be linked to the pathophysiology of IPF via altered plasminogen activation. In normal tissue, plasminogen is cleaved by tissue or urokinase plasminogen activator to form the active enzyme, plasmin, which contributes directly and via increased MMP activity to ECM degradation (259). In IPF, plasminogen activator inhibitor–1 is induced by TGF-b and inhibits plasminogen activation (259). This results in reduced decomposition of ECM and increased recruitment of inflammatory cells (259). Noncleaved plasminogen may then be cleaved increasingly to generate angiostatin, possibly by macrophage-derived metalloelastase, or by a direct effect of plasminogen activator inhibitor–1 on plasminogen. Locally elevated angiostatin levels in the fibrotic tissue may contribute to EC apoptosis and vascular rarefaction (260). However, the involvement of angiostatin in fibrosis and IPF-related PH needs to be established before angiostatin can be considered as a new therapeutic target, or even as a predictor of clinical outcome.

THERAPY The current therapeutic options are quite limited for IPF, and even more so for IPF-associated PH. The major problem in treating both fibrosis and pulmonary vascular disease is the amount of organized scar tissue inside the fibrotic lung, which no longer

takes part in active fibrogenesis. These areas are already fully organized, and active myofibroblasts are rarely found. This suggests that the IPF lung has areas with active fibrogenesis, which are typically characterized by the presence of fibroblastic foci, histologically normal appearing areas, and regions with the aforementioned organized scar tissue. The latter areas represent regions of final, nonreversible damage, not only to the interstitium, but very likely also to the pulmonary vasculature. This implies that these regions will likely not be accessible for therapy, and that therapeutic interventions will have to concentrate on the more active areas of the fibrotic lung. Therapy of PH in IPF will have to be a multimodal therapeutic approach that has to be closely connected with IPF therapy itself. There are several aspects that will need attention when designing therapeutic interventions for IPF and associated PH: (1) the progression of the fibrotic process needs to be decelerated or inhibited, because this would also help to slow down PH development or progression due to the close relationship between fibrosis and pulmonary vascular pathology, as discussed previously; (2) the integrity of the lung vasculature needs to be stabilized, especially endothelial injury, and apoptosis warrants reduction, due to its importance for PA wall remodeling, vascular rarefaction, and angioproliferation; and (3) right ventricular function needs to be maintained. Current IPF therapy has mainly focused on lung fibrosis itself, and has met rather limited success. Anti-inflammatory treatment using drugs, such as azathioprine, corticosteroids, cyclophosphamide, or methotrexate, has been the major strategy (3, 261). Overall, anti-inflammatory or immunosuppressive therapy has been useless in IPF, suggesting that inflammation might not be a central component of IPF (262). Recent clinical trials have shown small benefits (e.g., for the antifibrotic molecule, pirfenidone, or the antioxidant, N-acetylcysteine). Pirfenidone is a newly developed drug with combined anti-inflammatory, antioxidant, and antifibrotic effects, and clinical trial have shown small benefits (263, 264). Antioxidant strategies have been investigated before, after evidence that epithelial injury in IPF might be related to oxidative stress. A primarily antioxidant strategy using N-acetylcysteine seems to be beneficial for patients with IPF (265, 266). These therapeutic agents will be used to inhibit fibrosis progression. Other therapeutic agents targeting fibrogenesis are currently studied for IPF and IPF-PH, including IFNs, antagonists to cytokines, growth factors or their receptors, and antiangiogenic agents (details are available at www. clinicaltrials.gov) (266). Especially antiangiogenic therapy will require careful evalulation to find out whether angiogenesis inhibition will result in antifibrotic and antiproliferative effects instead of a proapoptotic outcome with accelerated progression of PH. There are currently no specific therapies for PH in IPF, and treatment guidelines for PH therapy have been primarily evaluated in other forms of PH. Although therapeutic trials for IPFassociated PH are currently underway, these studies mainly investigate drugs that are already used for other types of PH with more or less success (267). Prostacyclin-based therapies could reduce possible vasoconstriction and platelet aggregation (268, 269). Inhaled NO could be used as short-term therapy in advanced IPF or during acute deterioration (270). However, these vasodilators have to be tested carefully to rule out increased right–left shunting within the fibrotic lung. Sildenafil, a phosphodiesterase-5 inhibitor, is also a likely successful candidate in IPF, is frequently used in PAH, and has positive effects not only in the pulmonary vasculature (vasodilatation and inhibition of VSMC growth), but also selectively in the right ventricle (reduction of hypertrophy and elevated contractility) (270, 271). Whether or not Sildenafil can fulfill the promises from preliminary studies is currently being investigated in the STEP-IPF (Sildenafil Trial on

Translational Review

Exercise Performance in IPF, and Pulmonary Arterial Hypertension Secondary to IPF and Treatment with Sildenafil Trial [details available at www.clinicaltrials.gov]) (271, 272). ET receptor antagonists could have some benefit by inhibiting vasoconstriction and VSMC growth, and bosentan, dual ET receptor antagonist, is being tested for IPF-associated PH, despite its limited previous success in IPF or other PAH forms (270, 273) in the Pulmonary Arterial Hypertension Secondary to Idiopathic Pulmonary Fibrosis and Treatment with Bosentan Trial (www. clinicaltrials.gov). Anticoagulation strategies with coumadin or warfarin, similar to what is recommended for PH, may also be helpful in IPF (274). Future research is needed to select specific angiogenic or angiostatic growth factors as targets for future therapeutic concepts (e.g., to reduce endothelial apoptosis or angioproliferation). An additional concept, which could gain relevance in IPF and is currently tested in PAH, is the use of genetically modified EPCs (e.g., endothelial NO synthase transfected) to support endothelial repair, where animal and preliminary human data are very promising (128, 275, 276). There are limited therapeutic strategies currently aiming at right ventricular hypertrophy or failure. As mentioned, phoshodiesterase-5 inhibition can selectively improve right ventricular function. Further research is warranted to understand how the sick fibrotic lung affects the heart (e.g., whether there are systemic effects on angiogenesis in IPF), as could be suggested after Turner-Warwick’s study (277), which might also affect capillarization, cardiomyocyte growth, or matrix production in the myocardium. Understanding these complex interactions will be important in designing the aforementioned multimodal therapeutic approach for IPF and assocated PH. Future therapeutic strategies could also include the use of mesenchymal stem cells for myocardial regeneration, as currently being investigated in left ventricular failure of various origins (278). Lung transplantation (LTX) can be regarded as the ultimate of all therapeutic options, can be performed as single or double LTX, and results in decreased PAP after surgery (279). Due to the fast progression of IPF, integration with an LTX center should be considered early after diagnosis.

CONCLUSIONS AND TRANSLATIONAL OUTLOOK The pathobiology of IPF-associated pulmonary vascular disease is complicated, and the structural changes range from capillary loss and compression by fibrotic tissue to complex vascular lesions with intima and adventitia fibrosis and media thickening. Derived from studies in patient biopsies and our own experimental investigations, we believe that the main pathogenetic processes are a localized angiostatic shift, with reduction in angiogenic factors, such as VEGF, and an elevation of angiostatic factors, such as PEDF, EC injury, and apoptosis, vasoconstriction, and VSMC and fibroblast growth after growth factor release and activation. Several important translational aspects arise from our concept: 1. The interaction of angiogenesis and growth factors in vascular pathology in IPF is a field that will require extensive attention to find successful therapies, and future studies need to include investigations of the pulmonary vascular integrity. This is particularly so when it comes to trials that interfere with angiogenesis and growth factors, as indicated by our own animal research demonstrating that a proangiogenic factor (VEGF) reduces EC apoptosis and thereby PH. 2. Different pro- and anti-angiogenic and growth factors can be used as potential biomarkers to monitor disease pro-

9

gression by measuring their blood or BALF concentrations. Due to the variability in the measurements of these molecules, larger scale studies are warranted to find the best suitable marker or marker combination. 3. After our own experimental data, indicating a close spatiotemporal relationship between fibrotic process, EC injury, and vascular remodeling (including angioproliferation, muscularization, adventitiafibrosis, and vascular rarefaction), therapy of PH in IPF will likely have to interfere with several of these changes to obtain best results. This could be accomplished by targeting various pathways at the same time. Future research should provide additional clarification of the complex interactions to help to design more successful therapies for IPF-related PH. 4. Based on evidence in other forms of PH, it is likely that another important component of PH treatment in IPF will be to preserve right ventricular function. 5. Depending on the results of future clinical trials, currently used PH therapies might or might not be a part of this concept, but the current clinical studies have shown a significant benefit of some PH treatments in IPF-associated PH. Author Disclosure: None of the authors has a financial relationship with a commercial entity that has an interest in the subject of this manuscript.

References 1. American Thoracic Society/European Respiratory Society international multidisciplinary consensus classification of the idiopathic interstitial pneumonias. This joint statement of the American Thoracic Society (ATS), and the European Respiratory Society (ERS) was adopted by the ATS Board of Directors, June 2001 and by the ERS Executive Committee, June 2001. Am J Respir Crit Care Med 2002;165:277–304. 2. 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. 3. Behr J, Ryu JH. Pulmonary hypertension in interstitial lung disease. Eur Respir J 2008;31:1357–1367. 4. Ryu JH, Colby TV, Hartman TE. Idiopathic pulmonary fibrosis: current concepts. Mayo Clin Proc 1998;73:1085–1101. 5. Gross TJ, Hunninghake GW. Idiopathic pulmonary fibrosis. N Engl J Med 2001;345:517–525. 6. Kolb M, Margetts PJ, Anthony DC, Pitossi F, Gauldie J. Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. J Clin Invest 2001;107:1529–1536. 7. Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovectormediated gene transfer of active transforming growth factor–beta 1 induces prolonged severe fibrosis in rat lung. J Clin Invest 1997;100: 768–776. 8. Sime PJ, Marr RA, Gauldie D, Xing Z, Hewlett BR, Graham FL, Gauldie J. Transfer of tumor necrosis factor–alpha to rat lung induces severe pulmonary inflammation and patchy interstitial fibrogenesis with induction of transforming growth factor–beta1 and myofibroblasts. Am J Pathol 1998;153:825–832. 9. Bonniaud P, Margetts PJ, Ask K, Flanders K, Gauldie J, Kolb M. TGFbeta and Smad3 signaling link inflammation to chronic fibrogenesis. J Immunol 2005;175:5390–5395. 10. Flaherty KR, Mumford JA, Murray S, Kazerooni EA, Gross BH, Colby TV, Travis WD, Flint A, Toews GB, Lynch JP III, et al. Prognostic implications of physiologic and radiographic changes in idiopathic interstitial pneumonia. Am J Respir Crit Care Med 2003;168:543–548. 11. Collard HR, King TE Jr, Bartelson BB, Vourlekis JS, Schwarz MI, Brown KK. Changes in clinical and physiologic variables predict survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2003;168:538–542. 12. Kawut SM, O’Shea MK, Bartels MN, Wilt JS, Sonett JR, Arcasoy SM. Exercise testing determines survival in patients with diffuse paren-

10

13.

14.

15.

16.

17.

18. 19.

20.

21.

22.

23.

24.

25.

26. 27. 28.

29.

30. 31. 32.

33.

34.

35. 36. 37.

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011 chymal lung disease evaluated for lung transplantation. Respir Med 2005;99:1431–1439. Lettieri CJ, Nathan SD, Browning RF, Barnett SD, Ahmad S, Shorr AF. The distance–saturation product predicts mortality in idiopathic pulmonary fibrosis. Respir Med 2006;100:1734–1741. Lama VN, Flaherty KR, Toews GB, Colby TV, Travis WD, Long Q, Murray S, Kazerooni EA, Gross BH, Lynch JP III, et al. Prognostic value of desaturation during a 6-minute walk test in idiopathic interstitial pneumonia. Am J Respir Crit Care Med 2003;168:1084–1090. Hallstrand TS, Boitano LJ, Johnson WC, Spada CA, Hayes JG, Raghu G. The timed walk test as a measure of severity and survival in idiopathic pulmonary fibrosis. Eur Respir J 2005;25:96–103. Lederer DJ, Arcasoy SM, Wilt JS, D’Ovidio F, Sonett JR, Kawut SM. Six-minute-walk distance predicts waiting list survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2006;174:659–664. Lettieri CJ, Nathan SD, Barnett SD, Ahmad S, Shorr AF. Prevalence and outcomes of pulmonary arterial hypertension in advanced idiopathic pulmonary fibrosis. Chest 2006;129:746–752. Patel NM, Lederer DJ, Borczuk AC, Kawut SM. Pulmonary hypertension in idiopathic pulmonary fibrosis. Chest 2007;132:998–1006. D’Alonzo GE, Barst RJ, Ayres SM, Bergofsky EH, Brundage BH, Detre KM, Fishman AP, Goldring RM, Groves BM, Kernis JT, et al. Survival in patients with primary pulmonary hypertension. Results from a national prospective registry. Ann Intern Med 1991;115:343–349. Nathan SD, Noble PW, Tuder RM. Idiopathic pulmonary fibrosis and pulmonary hypertension: connecting the dots. Am J Respir Crit Care Med 2007;175:875–880. Simonneau G, Robbins IM, Beghetti M, Channick RN, Delcroix M, Denton CP, Elliott CG, Gaine SP, Gladwin MT, Jing Z-C, et al. Updated clinical classification of pulmonary hypertension. J Am Coll Cardiol 2009;54(1 Suppl 1):S43–S54. Nadrous HF, Pellikka PA, Krowka MJ, Swanson KL, Chaowalit N, Decker PA, Ryu JH. The impact of pulmonary hypertension on survival in patients with idiopathic pulmonary fibrosis. Chest 2005; 128(6 Suppl):616S–617S. Nadrous HF, Pellikka PA, Krowka MJ, Swanson KL, Chaowalit N, Decker PA, Ryu JH. Pulmonary hypertension in patients with idiopathic pulmonary fibrosis. Chest 2005;128:2393–2399. Nathan SD, Shlobin OA, Ahmad S, Koch J, Barnett SD, Ad N, Burton N, Leslie K. Serial development of pulmonary hypertension in patients with idiopathic pulmonary fibrosis. Respiration 2008;76: 288–294. Leuchte HH, Neurohr C, Baumgartner R, Holzapfel M, Giehrl W, Vogeser M, Behr J. Brain natriuretic peptide and exercise capacity in lung fibrosis and pulmonary hypertension. Am J Respir Crit Care Med 2004;170:360–365. Strange C, Highland KB. Pulmonary hypertension in interstitial lung disease. Curr Opin Pulm Med 2005;11:452–455. Shorr AF, Davies DB, Nathan SD. Outcomes for patients with sarcoidosis awaiting lung transplantation. Chest 2002;122:233–238. Ryu JH, Krowka MJ, Pellikka PA, Swanson KL, McGoon MD. Pulmonary hypertension in patients with interstitial lung diseases. Mayo Clin Proc 2007;82:342–350. Farkas L, Farkas D, Ask K, Mo¨ller A, Gauldie J, Margetts P, Inman M, Kolb M. VEGF ameliorates pulmonary hypertension through inhibition of endothelial apoptosis in experimental lung fibrosis in rats. J Clin Invest 2009;119:1298–1311. Ms S, Pardo A, Kaminski N. Idiopathic pulmonary fibrosis: aberrant recapitulation of developmental programs? PLoS Med 2008;5:e62. Hislop AA. Airway and blood vessel interaction during lung development. J Anat 2002;201:325–334. Hall SM, Hislop AA, Haworth SG. Origin, differentiation, and maturation of human pulmonary veins. Am J Respir Cell Mol Biol 2002;26:333–340. Hall SM, Hislop AA, Pierce CM, Haworth SG. Prenatal origins of human intrapulmonary arteries: formation and smooth muscle maturation. Am J Respir Cell Mol Biol 2000;23:194–203. Hislop A, Reid L. Fetal and childhood development of the intrapulmonary veins in man—branching pattern and structure. Thorax 1973;28:313–319. Hislop A, Reid L. Intra-pulmonary arterial development during fetal life-branching pattern and structure. J Anat 1972;113:35–48. deMello DE, Sawyer D, Galvin N, Reid LM. Early fetal development of lung vasculature. Am J Respir Cell Mol Biol 1997;16:568–581. Shifren JL, Doldi N, Ferrara N, Mesiano S, Jaffe RB. In the human fetus, vascular endothelial growth factor is expressed in epithelial

38.

39.

40.

41.

42.

43.

44.

45. 46.

47.

48.

49.

50.

51.

52.

53.

54.

55. 56.

57.

58.

59.

cells and myocytes, but not vascular endothelium: implications for mode of action. J Clin Endocrinol Metab 1994;79:316–322. Acarregui MJ, Penisten ST, Goss KL, Ramirez K, Snyder JM. Vascular endothelial growth factor gene expression in human fetal lung in vitro. Am J Respir Cell Mol Biol 1999;20:14–23. Healy AM, Morgenthau L, Zhu X, Farber HW, Cardoso WV. VEGF is deposited in the subepithelial matrix at the leading edge of branching airways and stimulates neovascularization in the murine embryonic lung. Dev Dyn 2000;219:341–352. Ng YS, Rohan R, Sunday ME, Demello DE, D’Amore PA. Differential expression of VEGF isoforms in mouse during development and in the adult. Dev Dyn 2001;220:112–121. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC. Failure of blood-island formation and vasculogenesis in Flk-1–deficient mice. Nature 1995;376:62–66. Jakkula M, Le Cras TD, Gebb S, Hirth KP, Tuder RM, Voelkel NF, Abman SH. Inhibition of angiogenesis decreases alveolarization in the developing rat lung. Am J Physiol Lung Cell Mol Physiol 2000; 279:L600–L607. Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Fujiwara Y, Gendron-Maguire M, Gridley T, Wolburg H, Risau W, Qin Y. Distinct roles of the receptor tyrosine kinases TIE-1 and TIE-2 in blood vessel formation. Nature 1995;376:70–74. Zeng X, Gray M, Stahlman MT, Whitsett JA. TGF-beta1 perturbs vascular development and inhibits epithelial differentiation in fetal lung in vivo. Dev Dyn 2001;221:289–301. Folkman J, D’Amore PA. Blood vessel formation: what is its molecular basis? Cell 1996;87:1153–1155. Arciniegas E, Neves CY, Carrillo LM, Zambrano EA, Ramı´rez R. Endothelial–mesenchymal transition occurs during embryonic pulmonary artery development. Endothelium 2005;12:193–200. Ebina M, Shimizukawa M, Shibata N, Kimura Y, Suzuki T, Endo M, Sasano H, Kondo T, Nukiwa T. Heterogeneous increase in CD34positive alveolar capillaries in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2004;169:1203–1208. Cosgrove GP, Brown KK, Schiemann WP, Serls AE, Parr JE, Geraci MW, Schwarz MI, Cool CD, Worthen GS. Pigment epithelium– derived factor in idiopathic pulmonary fibrosis: a role in aberrant angiogenesis. Am J Respir Crit Care Med 2004;170:242–251. Simler NR, Brenchley PE, Horrocks AW, Greaves SM, Hasleton PS, Egan JJ. Angiogenic cytokines in patients with idiopathic interstitial pneumonia. Thorax 2004;59:581–585. Tachihara A, Jin E, Matsuoka T, Ghazizadeh M, Yoshino S, Takemura T, Travis WD, Kawanami O. Critical roles of capillary endothelial cells for alveolar remodeling in nonspecific and usual interstitial pneumonias. J Nippon Med Sch 2006;73:203–213. Renzoni EA, Walsh DA, Salmon M, Wells AU, Sestini P, Nicholson AG, Veeraraghavan S, Bishop AE, Romanska HM, Pantelidis P, et al. Interstitial vascularity in fibrosing alveolitis. Am J Respir Crit Care Med 2003;167:438–443. Burdick MD, Murray LA, Keane MP, Xue YY, Zisman DA, Belperio JA, Strieter RM. CXCL11 attenuates bleomycin-induced pulmonary fibrosis via inhibition of vascular remodeling. Am J Respir Crit Care Med 2005;171:261–268. Keane MP, Arenberg DA, Lynch JP III, Whyte RI, Iannettoni MD, Burdick MD, Wilke CA, Morris SB, Glass MC, DiGiovine B, et al. The CXC chemokines, IL-8 and IP-10, regulate angiogenic activity in idiopathic pulmonary fibrosis. J Immunol 1997;159:1437–1443. Keane MP, Belperio JA, Burdick MD, Lynch JP, Fishbein MC, Strieter RM. ENA-78 is an important angiogenic factor in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2001;164:2239– 2242. Strieter RM, Gomperts BN, Keane MP. The role of CXC chemokines in pulmonary fibrosis. J Clin Invest 2007;117:549–556. Teng R-J, Eis A, Bakhutashvili I, Arul N, Konduri GG. Increased superoxide production contributes to the impaired angiogenesis of fetal pulmonary arteries with in utero pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2009;297:L184–L195. Fadini GP, Avogaro A, Ferraccioli G, Agostini C. Endothelial progenitors in pulmonary hypertension: new pathophysiology and therapeutic implications. Eur Respir J 2010;35:418–425. Fadini GP, Schiavon M, Rea F, Avogaro A, Agostini C. Depletion of endothelial progenitor cells may link pulmonary fibrosis and pulmonary hypertension. Am J Respir Crit Care Med 2007;176:724–725. Parra ER, David YR, da Costa LR, Ab’Saber A, Sousa R, Kairalla RA, de Carvalho CR, Filho MT, Capelozzi VL. Heterogeneous remod-

Translational Review

60.

61. 62.

63.

64. 65.

66.

67.

68.

69.

70.

71.

72.

73. 74.

75.

76.

77.

78.

79.

80.

81.

eling of lung vessels in idiopathic pulmonary fibrosis. Lung 2005;183: 291–300. Sakao S, Taraseviciene-Stewart L, Wood K, Cool CD, Voelkel NF. Apoptosis of pulmonary microvascular endothelial cells stimulates vascular smooth muscle cell growth. Am J Physiol Lung Cell Mol Physiol 2006;291:L362–L368. Budhiraja R, Tuder RM, Hassoun PM. Endothelial dysfunction in pulmonary hypertension. Circulation 2004;109:159–165. Ask K, Martin GE, Kolb M, Gauldie J. Targeting genes for treatment in idiopathic pulmonary fibrosis: challenges and opportunities, promises and pitfalls. Proc Am Thorac Soc 2006;3:389–393. Perrella MA, Edell ES, Krowka MJ, Cortese DA, Burnett JC Jr. Endothelium-derived relaxing factor in pulmonary and renal circulations during hypoxia. Am J Physiol 1992;263:R45–R50. Dinh-Xuan AT. Endothelial modulation of pulmonary vascular tone. Eur Respir J 1992;5:757–762. Smith AP, Demoncheaux EA, Higenbottam TW. Nitric oxide gas decreases endothelin-1 mRNA in cultured pulmonary artery endothelial cells. Nitric Oxide 2002;6:153–159. Hashimoto N, Phan SH, Imaizumi K, Matsuo M, Nakashima H, Kawabe T, Shimokata K, Hasegawa Y. Endothelial–mesenchymal transition in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 2010;43:161–172. Arciniegas E, Frid MG, Douglas IS, Stenmark KR. Perspectives on endothelial-to-mesenchymal transition: potential contribution to vascular remodeling in chronic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2007;293:L1–L8. Frid MG, Kale VA, Stenmark KR. Mature vascular endothelium can give rise to smooth muscle cells via endothelial–mesenchymal transdifferentiation: in vitro analysis. Circ Res 2002;90:1189–1196. Sakao S, Taraseviciene-Stewart L, Cool CD, Tada Y, Kasahara Y, Kurosu K, Tanabe N, Takiguchi Y, Tatsumi K, Kuriyama T, et al. VEGF-R blockade causes endothelial cell apoptosis, expansion of surviving CD341 precursor cells and transdifferentiation to smooth muscle–like and neuronal-like cells. FASEB J 2007;21:3640–3652. Koli K, Myllarniemi M, Vuorinen K, Salmenkivi K, Ryynanen MJ, Kinnula VL, Keski-Oja J. Bone morphogenetic protein–4 inhibitor gremlin is overexpressed in idiopathic pulmonary fibrosis. Am J Pathol 2006;169:61–71. Atkinson C, Stewart S, Upton PD, Machado R, Thomson JR, Trembath RC, Morrell NW. Primary pulmonary hypertension is associated with reduced pulmonary vascular expression of type II bone morphogenetic protein receptor. Circulation 2002;105:1672–1678. Costello CM, Howell K, Cahill E, McBryan J, Konigshoff M, Eickelberg O, Gaine S, Martin F, McLoughlin P. Lung-selective gene responses to alveolar hypoxia: potential role for the bone morphogenetic antagonist gremlin in pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2008;295:L272–L284. Warner JA, Kroegel C. Pulmonary immune cells in health and disease: mast cells and basophils. Eur Respir J 1994;7:1326–1341. Mitani Y, Ueda M, Maruyama K, Shimpo H, Kojima A, Matsumura M, Aoki K, Sakurai M. Mast cell chymase in pulmonary hypertension. Thorax 1999;54:88–90. Zhu YJ, Kradin R, Brandstetter RD, Staton G, Moss J, Hales CA. Hypoxic pulmonary hypertension in the mast cell–deficient mouse. J Appl Physiol 1983;54:680–686. Williams A, Heath D, Kay JM, Smith P. Lung mast cells in rats exposed to acute hypoxia, and chronic hypoxia with recovery. Thorax 1977; 32:287–295. Hassoun PM, Mouthon L, Barbera JA, Eddahibi S, Flores SC, Grimminger F, Jones PL, Maitland ML, Michelakis ED, Morrell NW, et al. Inflammation, growth factors, and pulmonary vascular remodeling. J Am Coll Cardiol 2009; 54(1, Suppl)S10–S19. Bournia V, Vlachoyiannopoulos P, Selmi C, Moutsopoulos H, Gershwin M. Recent advances in the treatment of systemic sclerosis. Clin Rev Allergy Immunol 2009;36:176–200. Antoniou KM, Wells AU. Scleroderma lung disease: evolving understanding in light of newer studies. Curr Opin Rheumatol 2008;20: 686–691. Bouros D, Wells AU, Nicholson AG, Colby TV, Polychronopoulos V, Pantelidis P, Haslam PL, Vassilakis DA, Black CM, du Bois RM. Histopathologic subsets of fibrosing alveolitis in patients with systemic sclerosis and their relationship to outcome. Am J Respir Crit Care Med 2002;165:1581–1586. du Bois RM. Mechanisms of scleroderma-induced lung disease. Proc Am Thorac Soc 2007;4:434–438.

11 82. Hachulla E, Gressin V, Guillevin L, Carpentier P, Diot E, Sibilia J, Kahan A, Cabane J, Frances C, Launay D, et al. Early detection of pulmonary arterial hypertension in systemic sclerosis: a French nationwide prospective multicenter study. Arthritis Rheum 2005;52: 3792–3800. 83. Mukerjee D, St George D, Coleiro B, Knight C, Denton CP, Davar J, Black CM, Coghlan JG. Prevalence and outcome in systemic sclerosis associated pulmonary arterial hypertension: application of a registry approach. Ann Rheum Dis 2003;62:1088–1093. 84. Sgonc R, Gruschwitz MS, Dietrich H, Recheis H, Gershwin ME, Wick G. Endothelial cell apoptosis is a primary pathogenetic event underlying skin lesions in avian and human scleroderma. J Clin Invest 1996;98:785–792. 85. Nicolls MR, Taraseviciene-Stewart L, Rai PR, Badesch DB, Voelkel NF. Autoimmunity and pulmonary hypertension: a perspective. Eur Respir J 2005;26:1110–1118. 86. Okano Y, Medsger TA Jr. Autoantibody to Th ribonucleoprotein (nucleolar 7–2 RNA protein particle) in patients with systemic sclerosis. Arthritis Rheum 1990;33:1822–1828. 87. Negi VS, Tripathy NK, Misra R, Nityanand S. Antiendothelial cell antibodies in scleroderma correlate with severe digital ischemia and pulmonary arterial hypertension. J Rheumatol 1998;25:462–466. 88. Harrison NK, Myers AR, Corrin B, Soosay G, Dewar A, Black CM, Du Bois RM, Turner-Warwick M. Structural features of interstitial lung disease in systemic sclerosis. Am Rev Respir Dis 1991;144:706–713. 89. Yurovsky VV, Wigley FM, Wise RA, White B. Skewing of the CD81 T-cell repertoire in the lungs of patients with systemic sclerosis. Hum Immunol 1996;48:84–97. 90. Majumdar S, Li D, Ansari T, Pantelidis P, Black CM, Gizycki M, du Bois RM, Jeffery PK. Different cytokine profiles in cryptogenic fibrosing alveolitis and fibrosing alveolitis associated with systemic sclerosis: a quantitative study of open lung biopsies. Eur Respir J 1999;14:251–257. 91. Cerinic MM, Valentini G, Sorano GG, D’Angelo S, Cuomo G, Fenu L, Generini S, Cinotti S, Morfini M, Pignone A, et al. Blood coagulation, fibrinolysis, and markers of endothelial dysfunction in systemic sclerosis. Semin Arthritis Rheum 2003;32:285–295. 92. Luzina IG, Atamas SP, Wise R, Wigley FM, Xiao HQ, White B. Gene expression in bronchoalveolar lavage cells from scleroderma patients. Am J Respir Cell Mol Biol 2002;26:549–557. 93. Luzina IG, Atamas SP, Wise R, Wigley FM, Choi J, Xiao HQ, White B. Occurrence of an activated, profibrotic pattern of gene expression in lung CD81 T cells from scleroderma patients. Arthritis Rheum 2003; 48:2262–2274. 94. Scala E, Pallotta S, Frezzolini A, Abeni D, Barbieri C, Sampogna F, De Pita O, Puddu P, Paganelli R, Russo G. Cytokine and chemokine levels in systemic sclerosis: relationship with cutaneous and internal organ involvement. Clin Exp Immunol 2004;138:540–546. 95. Meloni F, Caporali R, Marone Bianco A, Paschetto E, Morosini M, Fietta AM, Bobbio-Pallavicini F, Pozzi E, Montecucco C. Cytokine profile of bronchoalveolar lavage in systemic sclerosis with interstitial lung disease: comparison with usual interstitial pneumonia. Ann Rheum Dis 2004;63:892–894. 96. Le Pavec J, Humbert M, Mouthon L, Hassoun PM. Systemic sclerosis– associated pulmonary arterial hypertension. Am J Respir Crit Care Med 2010;181:1285–1293. 97. Guiducci S, Distler O, Distler JHW, Matucci-Cerinic M. Mechanisms of vascular damage in ssc—implications for vascular treatment strategies. Rheumatology 2008;47(Suppl 5):v18–v20. 98. Laplante P, Raymond MA, Gagnon G, Vigneault N, Sasseville AM, Langelier Y, Bernard M, Raymond Y, Hebert MJ. Novel fibrogenic pathways are activated in response to endothelial apoptosis: implications in the pathophysiology of systemic sclerosis. J Immunol 2005; 174:5740–5749. 99. Tamby MC, Humbert M, Guilpain P, Servettaz A, Dupin N, Christner JJ, Simonneau G, Fermanian J, Weill B, Guillevin L, et al. Antibodies to fibroblasts in idiopathic and scleroderma-associated pulmonary hypertension. Eur Respir J 2006;28:799–807. 100. Kahan A, Coghlan G, McLaughlin V. Cardiac complications of systemic sclerosis. Rheumatology (Oxford) 2009;48:iii45–iii48. 101. Steen VD, Follansbee WP, Conte CG, Medsger TA Jr. Thallium perfusion defects predict subsequent cardiac dysfunction in patients with systemic sclerosis. Arthritis Rheum 1996;39:677–681. 102. de Groote P, Gressin V, Hachulla E, Carpentier P, Guillevin L, Kahan A, Cabane J, Frances C, Lamblin N, Diot E, et al. Evaluation of cardiac abnormalities by Doppler echocardiography in a large

12

103. 104.

105. 106.

107.

108. 109.

110.

111.

112.

113.

114. 115.

116.

117.

118. 119.

120.

121.

122.

123.

124.

125.

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011 nationwide multicentric cohort of patients with systemic sclerosis. Ann Rheum Dis 2008;67:31–36. Gerke AK, Hunninghake G. The immunology of sarcoidosis. Clin Chest Med 2008;29:379–390. Agostini C, Facco M, Chilosi M, Semenzato G. Alveolar macrophage-T cell interactions during Th1-type sarcoid inflammation. Microsc Res Tech 2001;53:278–287. Diaz-Guzman E, Farver C, Parambil J, Culver DA. Pulmonary hypertension caused by sarcoidosis. Clin Chest Med 2008;29:549–563. Rosen Y, Moon S, Huang CT, Gourin A, Lyons HA. Granulomatous pulmonary angiitis in sarcoidosis. Arch Pathol Lab Med 1977;101:170– 174. Takemura T, Matsui Y, Oritsu M, Akiyama O, Hiraga Y, Omichi M, Hirasawa M, Saiki S, Tamura S, Mochizuki I, et al. Pulmonary vascular involvement in sarcoidosis: granulomatous angiitis and microangiopathy in transbronchial lung biopsies. Virchows Arch A Pathol Anat Histopathol 1991;418:361–368. Hoffstein V, Ranganathan N, Mullen JB. Sarcoidosis simulating pulmonary veno-occlusive disease. Am Rev Respir Dis 1986;134:809–811. Schachter EN, Smith GJ, Cohen GS, Lee SH, Laser A, Gee JB. Pulmonary granulomas in a patient with pulmonary veno-occlusive disease. Chest 1975;67:487–489. Reichenberger F, Schauer J, Kellner K, Sack U, Stiehl P, Winkler J. Different expression of endothelin in the bronchoalveolar lavage in patients with pulmonary diseases. Lung 2001;179:163–174. Koyama S, Sato E, Haniuda M, Numanami H, Nagai S, Izumi T. Decreased level of vascular endothelial growth factor in bronchoalveolar lavage fluid of normal smokers and patients with pulmonary fibrosis. Am J Respir Crit Care Med 2002;166:382–385. Hofmann S, Franke A, Fischer A, Jacobs G, Nothnagel M, Gaede KI, Schurmann M, Muller-Quernheim J, Krawczak M, Rosenstiel P, et al. Genome-wide association study identifies ANXA11 as a new susceptibility locus for sarcoidosis. Nat Genet 2008;40:1103–1106. Wasfi YS, Silveira LJ, Jonth A, Hokanson JE, Fingerlin T, Sato H, Parsons CE, Lympany P, Welsh K, du Bois RM, et al. Fas promoter polymorphisms: genetic predisposition to sarcoidosis in AfricanAmericans. Tissue Antigens 2008;72:39–48. Pierre-Louis B, Prasad A, Frishman WH. Cardiac manifestations of sarcoidosis and therapeutic options. Cardiol Rev 2009;17:153–158. Voelkel NF, Vandivier RW, Tuder RM. Vascular endothelial growth factor in the lung. Am J Physiol Lung Cell Mol Physiol 2006;290: L209–L221. Kasahara Y, Tuder RM, Taraseviciene-Stewart L, Le Cras TD, Abman S, Hirth PK, Waltenberger J, Voelkel NF. Inhibition of VEGF receptors causes lung cell apoptosis and emphysema. J Clin Invest 2000;106:1311–1319. Ramirez-Bergeron DL, Runge A, Adelman DM, Gohil M, Simon MC. HIF-dependent hematopoietic factors regulate the development of the embryonic vasculature. Dev Cell 2006;11:81–92. Tuder RM, Yun JH. Vascular endothelial growth factor of the lung: friend or foe. Curr Opin Pharmacol 2008;8:255–260. Meyer KC, Cardoni A, Xiang ZZ. Vascular endothelial growth factor in bronchoalveolar lavage from normal subjects and patients with diffuse parenchymal lung disease. J Lab Clin Med 2000;135:332–338. Hamada N, Kuwano K, Yamada M, Hagimoto N, Hiasa K, Egashira K, Nakashima N, Maeyama T, Yoshimi M, Nakanishi Y. Anti-vascular endothelial growth factor gene therapy attenuates lung injury and fibrosis in mice. J Immunol 2005;175:1224–1231. Moeller A, Ask K, Warburton D, Gauldie J, Kolb M. The bleomycin animal model: a useful tool to investigate treatment options for idiopathic pulmonary fibrosis? Int J Biochem Cell Biol 2008;40:362–382. Borzone G, Moreno R, Urrea R, Meneses M, Oyarzun M, Lisboa C. Bleomycin-induced chronic lung damage does not resemble human idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2001;163: 1648–1653. Campbell AI, Zhao Y, Sandhu R, Stewart DJ. Cell-based gene transfer of vascular endothelial growth factor attenuates monocrotalineinduced pulmonary hypertension. Circulation 2001;104:2242–2248. Fujita M, Mason RJ, Cool C, Shannon JM, Hara N, Fagan KA. Pulmonary hypertension in TNF-alpha–overexpressing mice is associated with decreased VEGF gene expression. J Appl Physiol 2002; 93:2162–2170. Grover TR, Parker TA, Markham NE, Abman SH. RhVEGF treatment preserves pulmonary vascular reactivity and structure in an experimental model of pulmonary hypertension in fetal sheep. Am J Physiol Lung Cell Mol Physiol 2005;289:L315–L321.

126. Le Cras TD, Markham NE, Tuder RM, Voelkel NF, Abman SH. Treatment of newborn rats with a VEGF receptor inhibitor causes pulmonary hypertension and abnormal lung structure. Am J Physiol Lung Cell Mol Physiol 2002;283:L555–L562. 127. Partovian C, Adnot S, Raffestin B, Louzier V, Levame M, Mavier IM, Lemarchand P, Eddahibi S. Adenovirus-mediated lung vascular endothelial growth factor overexpression protects against hypoxic pulmonary hypertension in rats. Am J Respir Cell Mol Biol 2000;23: 762–771. 128. Zhao YD, Courtman DW, Ng DS, Robb MJ, Deng YP, Trogadis J, Han RN, Stewart DJ. Microvascular regeneration in established pulmonary hypertension by angiogenic gene transfer. Am J Respir Cell Mol Biol 2006;35:182–189. 129. Jiang B, Liu L. Chapter 2: PI3K/PTEN signaling in angiogenesis and tumorigenesis. In: George FVW, George K, editors. Advances in cancer research, Volume 102. San Diego, CA: Academic Press; 2009. Pp. 19–65. 130. Vasakova M, Sterclova M, Kolesar L, Slavcev A, Pohunek P, Sulc J, Skibova J, Striz I. Bronchoalveolar lavage fluid cellular characteristics, functional parameters and cytokine and chemokine levels in interstitial lung diseases. Scand J Immunol 2009;69:268–274. 131. Tombran-Tink J, Barnstable CJ. PEDF: a multifaceted neurotrophic factor. Nat Rev Neurosci 2003;4:628–636. 132. Chen L, Zhang SS, Barnstable CJ, Tombran-Tink J. PEDF induces apoptosis in human endothelial cells by activating p38 MAP kinase dependent cleavage of multiple caspases. Biochem Biophys Res Commun 2006;348:1288–1295. 133. Ho TC, Chen SL, Yang YC, Liao CL, Cheng HC, Tsao YP. PEDF induces p53-mediated apoptosis through PPAR gamma signaling in human umbilical vein endothelial cells. Cardiovasc Res 2007;76:213–223. 134. Pignolo RJ, Francis MK, Rotenberg MO, Cristofalo VJ. Putative role for EPC-1/PEDF in the G0 growth arrest of human diploid fibroblasts. J Cell Physiol 2003;195:12–20. 135. Dawson DW, Volpert OV, Gillis P, Crawford SE, Xu H, Benedict W, Bouck NP. Pigment epithelium–derived factor: a potent inhibitor of angiogenesis. Science 1999;285:245–248. 136. Zhang SX, Wang JJ, Gao G, Parke K, Ma JX. Pigment epithelium– derived factor downregulates vascular endothelial growth factor (VEGF) expression and inhibits VEGF–VEGF receptor 2 binding in diabetic retinopathy. J Mol Endocrinol 2006;37:1–12. 137. Yamagishi S, Inagaki Y, Nakamura K, Abe R, Shimizu T, Yoshimura A, Imaizumi T. Pigment epithelium–derived factor inhibits TNFalpha–induced interleukin-6 expression in endothelial cells by suppressing NADPH oxidase–mediated reactive oxygen species generation. J Mol Cell Cardiol 2004;37:497–506. 138. Alberdi E, Aymerich MS, Becerra SP. Binding of pigment epithelium– derived factor (PEDF) to retinoblastoma cells and cerebellar granule neurons: evidence for a PEDF receptor. J Biol Chem 1999;274:31605–31612. 139. Bilak MM, Becerra SP, Vincent AM, Moss BH, Aymerich MS, Kuncl RW. Identification of the neuroprotective molecular region of pigment epithelium–derived factor and its binding sites on motor neurons. J Neurosci 2002;22:9378–9386. 140. Aymerich MS, Alberdi EM, Martinez A, Becerra SP. Evidence for pigment epithelium–derived factor receptors in the neural retina. Invest Ophthalmol Vis Sci 2001;42:3287–3293. 141. Bernard A, Gao-Li J, Franco CA, Bouceba T, Huet A, Li Z. Laminin receptor involvement in the anti-angiogenic activity of pigment epithelium–derived factor. J Biol Chem 2009;284:10480–10490. 142. Notari L, Baladron V, Aroca-Aguilar JD, Balko N, Heredia R, Meyer C, Notario PM, Saravanamuthu S, Nueda ML, Sanchez-Sanchez F, et al. Identification of a lipase-linked cell membrane receptor for pigment epithelium–derived factor. J Biol Chem 2006;281:38022–38037. 143. Filleur S, Volz K, Nelius T, Mirochnik Y, Huang H, Zaichuk TA, Aymerich MS, Becerra SP, Yap R, Veliceasa D, et al. Two functional epitopes of pigment epithelial–derived factor block angiogenesis and induce differentiation in prostate cancer. Cancer Res 2005;65:5144–5152. 144. Filleur S, Nelius T, de Riese W, Kennedy RC. Characterization of PEDF: a multi-functional serpin family protein. J Cell Biochem 2009;106:769–775. 145. Maik-Rachline G, Seger R. Variable phosphorylation states of pigmentepithelium–derived factor differentially regulate its function. Blood 2006;107:2745–2752. 146. Maik-Rachline G, Shaltiel S, Seger R. Extracellular phosphorylation converts pigment epithelium–derived factor from a neurotrophic to an antiangiogenic factor. Blood 2005;105:670–678.

Translational Review 147. Hosomichi J, Yasui N, Koide T, Soma K, Morita I. Involvement of the collagen I–binding motif in the anti-angiogenic activity of pigment epithelium–derived factor. Biochem Biophys Res Commun 2005;335: 756–761. 148. Meyer C, Notari L, Becerra SP. Mapping the type I collagen–binding site on pigment epithelium–derived factor: implications for its antiangiogenic activity. J Biol Chem 2002;277:45400–45407. 149. Valnickova Z, Petersen SV, Nielsen SB, Otzen DE, Enghild JJ. Heparin binding induces a conformational change in pigment epithelium–derived factor. J Biol Chem 2007;282:6661–6667. 150. Becerra SP, Perez-Mediavilla LA, Weldon JE, Locatelli-Hoops S, Senanayake P, Notari L, Notario V, Hollyfield JG. Pigment epithelium– derived factor binds to hyaluronan: mapping of a hyaluronan binding site. J Biol Chem 2008;283:33310–33320. 151. Bensadoun ES, Burke AK, Hogg JC, Roberts CR. Proteoglycan deposition in pulmonary fibrosis. Am J Respir Crit Care Med 1996;154:1819–1828. 152. Notari L, Miller A, Martinez A, Amaral J, Ju M, Robinson G, Smith LE, Becerra SP. Pigment epithelium–derived factor is a substrate for matrix metalloproteinase type 2 and type 9: implications for downregulation in hypoxia. Invest Ophthalmol Vis Sci 2005;46:2736–2747. 153. Selman M, Ruiz V, Cabrera S, Segura L, Ramirez R, Barrios R, Pardo A. TIMP-1, -2, -3, and -4 in idiopathic pulmonary fibrosis: a prevailing nondegradative lung microenvironment? Am J Physiol Lung Cell Mol Physiol 2000;279:L562–L574. 154. Zamiri P, Masli S, Streilein JW, Taylor AW. Pigment epithelial growth factor suppresses inflammation by modulating macrophage activation. Invest Ophthalmol Vis Sci 2006;47:3912–3918. 155. Zamiri P, Sugita S, Streilein JW. Immunosuppressive properties of the pigmented epithelial cells and the subretinal space. Chem Immunol Allergy 2007;92:86–93. 156. Zhang SX, Wang JJ, Gao G, Shao C, Mott R, Ma JX. Pigment epithelium–derived factor (PEDF) is an endogenous antiinflammatory factor. FASEB J 2006;20:323–325. 157. Wang JJ, Zhang SX, Lu K, Chen Y, Mott R, Sato S, Ma JX. Decreased expression of pigment epithelium–derived factor is involved in the pathogenesis of diabetic nephropathy. Diabetes 2005;54:243–250. 158. Wang JJ, Zhang SX, Mott R, Knapp RR, Cao W, Lau K, Ma JX. Salutary effect of pigment epithelium–derived factor in diabetic nephropathy: evidence for antifibrogenic activities. Diabetes 2006;55: 1678–1685. 159. Cao W, Tombran-Tink J, Chen W, Mrazek D, Elias R, McGinnis JF. Pigment epithelium–derived factor protects cultured retinal neurons against hydrogen peroxide–induced cell death. J Neurosci Res 1999; 57:789–800. 160. Pang IH, Zeng H, Fleenor DL, Clark AF. Pigment epithelium–derived factor protects retinal ganglion cells. BMC Neurosci 2007;8:11. 161. Cai J, Parr C, Watkins G, Jiang WG, Boulton M. Decreased pigment epithelium–derived factor expression in human breast cancer progression. Clin Cancer Res 2006;12:3510–3517. 162. Uehara H, Miyamoto M, Kato K, Ebihara Y, Kaneko H, Hashimoto H, Murakami Y, Hase R, Takahashi R, Mega S, et al. Expression of pigment epithelium–derived factor decreases liver metastasis and correlates with favorable prognosis for patients with ductal pancreatic adenocarcinoma. Cancer Res 2004;64:3533–3537. 163. Zhang L, Chen J, Ke Y, Mansel RE, Jiang WG. Expression of pigment epithelial derived factor is reduced in non–small cell lung cancer and is linked to clinical outcome. Int J Mol Med 2006;17:937–944. 164. Rychli K, Niessner A, Hohensinner PJ, Mahdy Ali K, Kaun C, Neuhold S, Zorn G, Richter B, Hulsmann M, Berger R, et al. Prognostic value of pigment epithelium–derived factor in advanced heart failure patients. Chest 2010;138:656–664. 165. O’Kane S, Ferguson MW. Transforming growth factor beta S and wound healing. Int J Biochem Cell Biol 1997;29:63–78. 166. Gauldie J, Kolb M, Ask K, Martin G, Bonniaud P, Warburton D. Smad3 signaling involved in pulmonary fibrosis and emphysema. Proc Am Thorac Soc 2006;3:696–702. 167. Prud’homme GJ. Pathobiology of transforming growth factor [beta] in cancer, fibrosis and immunologic disease, and therapeutic considerations. Lab Invest 2007;87:1077–1091. 168. Zheng SG, Wang J, Wang P, Gray JD, Horwitz DA. IL-2 is essential for TGF-beta to convert naive CD41CD252 cells to CD251FOXP31 regulatory T cells and for expansion of these cells. J Immunol 2007; 178:2018–2027. 169. Marie JC, Letterio JJ, Gavin M, Rudensky AY. TGF-beta1 maintains suppressor function and FOXP3 expression in CD41CD251 regulatory T cells. J Exp Med 2005;201:1061–1067.

13 170. Pyzik M, Piccirillo CA. TGF-beta1 modulates FOXP3 expression and regulatory activity in distinct CD41 T cell subsets. J Leukoc Biol 2007;82:335–346. 171. Selvaraj RK, Geiger TL. A kinetic and dynamic analysis of foxp3 induced in t cells by TGF-beta. J Immunol 2007;179:11 p following 1390. 172. Wahl SM, Hunt DA, Wakefield LM, McCartney-Francis N, Wahl LM, Roberts AB, Sporn MB. Transforming growth factor type beta induces monocyte chemotaxis and growth factor production. Proc Natl Acad Sci USA 1987;84:5788–5792. 173. Pertovaara L, Kaipainen A, Mustonen T, Orpana A, Ferrara N, Saksela O, Alitalo K. Vascular endothelial growth factor is induced in response to transforming growth factor–beta in fibroblastic and epithelial cells. J Biol Chem 1994;269:6271–6274. 174. Carmeliet P. Angiogenesis in health and disease. Nat Med 2003;9:653–660. 175. Pollman MJ, Naumovski L, Gibbons GH. Vascular cell apoptosis: cell type–specific modulation by transforming growth factor–b1 in endothelial cells versus smooth muscle cells. Circulation 1999;99: 2019–2026. 176. Khalil N, O’Connor R, Unruh H, Warren P, Kemp A, Greenberg A. Enhanced expression and immunohistochemical distribution of transforming growth factor–beta in idiopathic pulmonary fibrosis. Chest 1991;99(3 Suppl):65S–66S. 177. Bergeron A, Soler P, Kambouchner M, Loiseau P, Milleron B, Valeyre D, Hance AJ, Tazi A. Cytokine profiles in idiopathic pulmonary fibrosis suggest an important role for TGF-beta and IL-10. Eur Respir J 2003;22:69–76. 178. Zaiman AL, Podowski M, Medicherla S, Gordy K, Xu F, Zhen L, Shimoda LA, Neptune E, Higgins L, Murphy A, et al. Role of the TGF-fbetag/ALK5 signaling pathway in monocrotaline-induced pulmonary hypertension. Am J Respir Crit Care Med 2008;177: 896–905. 179. Bonniaud P, Margetts PJ, Kolb M, Schroeder JA, Kapoun AM, Damm D, Murphy A, Chakravarty S, Dugar S, Higgins L, et al. Progressive transforming growth factor beta1–induced lung fibrosis is blocked by an orally active ALK5 kinase inhibitor. Am J Respir Crit Care Med 2005;171:889–898. 180. Hiwatari N, Shimura S, Yamauchi K, Nara M, Hida W, Shirato K. Significance of elevated procollagen-III–peptide and transforming growth factor–beta levels of bronchoalveolar lavage fluids from idiopathic pulmonary fibrosis patients. Tohoku J Exp Med 1997; 181:285–295. 181. Meloni F, Caporali R, Marone Bianco A, Paschetto E, Morosini M, Fietta AM, Patrizio V, Bobbio-Pallavicini F, Pozzi E, Montecucco C. BAL cytokine profile in different interstitial lung diseases: a focus on systemic sclerosis. Sarcoidosis Vasc Diffuse Lung Dis 2004;21:111–118. 182. Otrock ZK, Mahfouz RA, Makarem JA, Shamseddine AI. Understanding the biology of angiogenesis: review of the most important molecular mechanisms. Blood Cells Mol Dis 2007;39:212–220. 183. Thomas KA. Fibroblast growth factors. FASEB J 1987;1:434–440. 184. Cross MJ, Claesson-Welsh L. FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends Pharmacol Sci 2001;22:201–207. 185. Presta M, Tiberio L, Rusnati M, Dell’Era P, Ragnotti G. Basic fibroblast growth factor requires a long-lasting activation of protein kinase C to induce cell proliferation in transformed fetal bovine aortic endothelial cells. Cell Regul 1991;2:719–726. 186. Lappi-Blanco E, Soini Y, Kinnula V, Paakko P. VEGF and BFGF are highly expressed in intraluminal fibromyxoid lesions in bronchiolitis obliterans organizing pneumonia. J Pathol 2002;196:220–227. 187. Benisty JI, McLaughlin VV, Landzberg MJ, Rich JD, Newburger JW, Rich S, Folkman J. Elevated basic fibroblast growth factor levels in patients with pulmonary arterial hypertension. Chest 2004;126:1255–1261. 188. Izikki M. Endothelial-derived FGF2 contributes to the progression of pulmonary hypertension in humans and rodents. J Clin Invest 2009; 119:512–523. 189. Belperio JA, Keane MP, Arenberg DA, Addison CL, Ehlert JE, Burdick MD, Strieter RM. CXC chemokines in angiogenesis. J Leukoc Biol 2000;68:1–8. 190. Strieter RM, Polverini PJ, Kunkel SL, Arenberg DA, Burdick MD, Kasper J, Dzuiba J, Van Damme J, Walz A, Marriott D, et al. The functional role of the ELR motif in CXC chemokine–mediated angiogenesis. J Biol Chem 1995;270:27348–27357. 191. Nor JE, Christensen J, Liu J, Peters M, Mooney DJ, Strieter RM, Polverini PJ. Up-regulation of BCL-2 in microvascular endothelial cells enhances intratumoral angiogenesis and accelerates tumor growth. Cancer Res 2001;61:2183–2188.

14

AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY VOL 45 2011

192. Addison CL, Daniel TO, Burdick MD, Liu H, Ehlert JE, Xue YY, Buechi L, Walz A, Richmond A, Strieter RM. The CXC chemokine receptor 2, CXCR2, is the putative receptor for ELR1 CXC chemokine– induced angiogenic activity. J Immunol 2000;165:5269–5277. 193. Murdoch C, Monk PN, Finn A. CXC chemokine receptor expression on human endothelial cells. Cytokine 1999;11:704–712. 194. Salcedo R, Resau JH, Halverson D, Hudson EA, Dambach M, Powell D, Wasserman KEN, Oppenheim JJ. Differential expression and responsiveness of chemokine receptors (CXCR1–3) by human microvascular endothelial cells and umbilical vein endothelial cells. FASEB J 2000;14:2055–2064. 195. Schraufstatter IU, Trieu K, Zhao M, Rose DM, Terkeltaub RA, Burger M. IL-8–mediated cell migration in endothelial cells depends on cathepsin B activity and transactivation of the epidermal growth factor receptor. J Immunol 2003;171:6714–6722. 196. Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY, Belperio JA, Keane MP, Strieter RM. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest 2004;114:438–446. 197. Marcel L, Pius L, Nicole B, Eckart M, Bernhard M. Lymphocytespecific chemokine receptor CXCR3: regulation, chemokine binding and gene localization. Eur J Immunol 1998;28:3696–3705. 198. Antoniou KM, Tzouvelekis A, Alexandrakis MG, Sfiridaki K, Tsiligianni I, Rachiotis G, Tzanakis N, Bouros D, Milic-Emili J, Siafakas NM. Different angiogenic activity in pulmonary sarcoidosis and idiopathic pulmonary fibrosis. Chest 2006;130:982–988. 199. Belperio JA, Dy M, Murray L, Burdick MD, Xue YY, Strieter RM, Keane MP. The role of the Th2 CC chemokine ligand CCL17 in pulmonary fibrosis. J Immunol 2004;173:4692–4698. 200. Keane MP, Belperio JA, Moore TA, Moore BB, Arenberg DA, Smith RE, Burdick MD, Kunkel SL, Strieter RM. Neutralization of the CXC chemokine, macrophage inflammatory protein-2, attenuates bleomycin-induced pulmonary fibrosis. J Immunol 1999;162:5511–5518. 201. Keane MP, Belperio JA, Arenberg DA, Burdick MD, Xu ZJ, Xue YY, Strieter RM. IFN-fgammag–inducible protein–10 attenuates bleomycininduced pulmonary fibrosis via inhibition of angiogenesis. J Immunol 1999;163:5686–5692. 202. Fredriksson L, Li H, Eriksson U. The PDGF family: four gene products form five dimeric isoforms. Cytokine Growth Factor Rev 2004;15: 197–204. 203. Lindahl P, Johansson BR, Leveen P, Betsholtz C. Pericyte loss and microaneurysm formation in PDGF-B–deficient mice. Science 1997; 277:242–245. 204. Hellstrom M, Gerhardt H, Kalen M, Li X, Eriksson U, Wolburg H, Betsholtz C. Lack of pericytes leads to endothelial hyperplasia and abnormal vascular morphogenesis. J Cell Biol 2001;153:543–553. 205. Homma S, Nagaoka I, Abe H, Takahashi K, Seyama K, Nukiwa T, Kira S. Localization of platelet-derived growth factor and insulin-like growth factor I in the fibrotic lung. Am J Respir Crit Care Med 1995; 152:2084–2089. 206. Antoniades HN, Bravo MA, Avila RE, Galanopoulos T, NevilleGolden J, Maxwell M, Selman M. Platelet-derived growth factor in idiopathic pulmonary fibrosis. J Clin Invest 1990;86:1055–1064. 207. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R, Roth M, Sydykov A, Lai YJ, Weissmann N, Seeger W, et al. Reversal of experimental pulmonary hypertension by PDGF inhibition. J Clin Invest 2005;115:2811–2821. 208. Clamp AR, Jayson GC. The clinical potential of antiangiogenic fragments of extracellular matrix proteins. Br J Cancer 2005;93: 967–972. 209. Felbor U, Dreier L, Bryant RA, Ploegh HL, Olsen BR, Mothes W. Secreted cathepsin l generates endostatin from collagen XVIII. EMBO J 2000;19:1187–1194. 210. O’Reilly MS, Boehm T, Shing Y, Fukai N, Vasios G, Lane WS, Flynn E, Birkhead JR, Olsen BR, Folkman J. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 1997;88:277–285. 211. Yang IV, Burch LH, Steele MP, Savov JD, Hollingsworth JW, McElvania-Tekippe E, Berman KG, Speer MC, Sporn TA, Brown KK, et al. Gene expression profiling of familial and sporadic interstitial pneumonia. Am J Respir Crit Care Med 2007;175:45–54. 212. Thickett DR, Poole AR, Millar AB. The balance between collagen synthesis and degradation in diffuse lung disease. Sarcoidosis Vasc Diffuse Lung Dis 2001;18:27–33. 213. Paddenberg R, Faulhammer P, Goldenberg A, Kummer W. Hypoxiainduced increase of endostatin in murine aorta and lung. Histochem Cell Biol 2006;125:497–508.

214. Masaki T, Yanagisawa M, Goto K. Physiology and pharmacology of endothelins. Med Res Rev 1992;12:391–421. 215. Giaid A, Polak JM, Gaitonde V, Hamid QA, Moscoso G, Legon S, Uwanogho D, Roncalli M, Shinmi O, Sawamura T, et al. Distribution of endothelin-like immunoreactivity and mRNA in the developing and adult human lung. Am J Respir Cell Mol Biol 1991;4:50–58. 216. Giaid A, Michel RP, Stewart DJ, Sheppard M, Corrin B, Hamid Q. Expression of endothelin-1 in lungs of patients with cryptogenic fibrosing alveolitis. Lancet 1993;341:1550–1554. 217. Saleh D, Furukawa K, Tsao MS, Maghazachi A, Corrin B, Yanagisawa M, Barnes PJ, Giaid A. Elevated expression of endothelin-1 and endothelin-converting enzyme–1 in idiopathic pulmonary fibrosis: possible involvement of proinflammatory cytokines. Am J Respir Cell Mol Biol 1997;16:187–193. 218. Giaid A, Yanagisawa M, Langleben D, Michel RP, Levy R, Shennib H, Kimura S, Masaki T, Duguid WP, Stewart DJ. Expression of endothelin-1 in the lungs of patients with pulmonary hypertension. N Engl J Med 1993;328:1732–1739. 219. Dupuis J, Prie S. The ET(A)-receptor antagonist LU 135252 prevents the progression of established pulmonary hypertension induced by monocrotaline in rats. J Cardiovasc Pharmacol Ther 1999;4:33–39. 220. Park SH, Saleh D, Giaid A, Michel RP. Increased endothelin-1 in bleomycin-induced pulmonary fibrosis and the effect of an endothelin receptor antagonist. Am J Respir Crit Care Med 1997;156:600–608. 221. Trakada G, Spiropoulos K. Arterial endothelin-1 in interstitial lung disease patients with pulmonary hypertension. Monaldi Arch Chest Dis 2001;56:379–383. 222. Comellas AP, Briva A. Role of endothelin-1 in acute lung injury. Transl Res 2009;153:263–271. 223. Sofia M, Mormile M, Faraone S, Alifano M, Carratu P, Carratu L. Endothelin-1 excretion in urine in active pulmonary sarcoidosis and in other interstitial lung diseases. Sarcoidosis 1995;12:118–123. 224. Jeffery TK, Morrell NW. Molecular and cellular basis of pulmonary vascular remodeling in pulmonary hypertension. Prog Cardiovasc Dis 2002;45:173–202. 225. Morrell N, Higham M, Phillips P, Shakur BH, Robinson P, Beddoes R. Pilot study of losartan for pulmonary hypertension in chronic obstructive pulmonary disease. Respir Res 2005;6:88. 226. Wang R, Zagariya A, Ibarra-Sunga O, Gidea C, Ang E, Deshmukh S, Chaudhary G, Baraboutis J, Filippatos G, Uhal BD. Angiotensin II induces apoptosis in human and rat alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 1999;276:L885–L889. 227. Wang R, Ibarra-Sunga O, Verlinski L, Pick R, Uhal BD. Abrogation of bleomycin-induced epithelial apoptosis and lung fibrosis by captopril or by a caspase inhibitor. Am J Physiol Lung Cell Mol Physiol 2000; 279:L143–L151. 228. Maniatis NA, Kotanidou A, Catravas JD, Orfanos SE. Endothelial pathomechanisms in acute lung injury. Vascul Pharmacol 2008;49: 119–133. 229. Specks U, Martin WJ II, Rohrbach MS. Bronchoalveolar lavage fluid angiotensin-converting enzyme in interstitial lung diseases. Am Rev Respir Dis 1990;141:117–123. 230. Fiedler U, Augustin HG. Angiopoietins: a link between angiogenesis and inflammation. Trends Immunol 2006;27:552–558. 231. Gale NW, Yancopoulos GD. Growth factors acting via endothelial cell–specific receptor tyrosine kinases: VEGFs, angiopoietins, and ephrins in vascular development. Genes Dev 1999;13:1055–1066. 232. Partanen J, Armstrong E, Makela TP, Korhonen J, Sandberg M, Renkonen R, Knuutila S, Huebner K, Alitalo K. A novel endothelial cell surface receptor tyrosine kinase with extracellular epidermal growth factor homology domains. Mol Cell Biol 1992;12:1698–1707. 233. Iwama A, Hamaguchi I, Hashiyama M, Murayama Y, Yasunaga K, Suda T. Molecular cloning and characterization of mouse TIE and TEK receptor tyrosine kinase genes and their expression in hematopoietic stem cells. Biochem Biophys Res Commun 1993;195:301–309. 234. Sato TN, Qin Y, Kozak CA, Audus KL. TIE-1 and TIE-2 define another class of putative receptor tyrosine kinase genes expressed in early embryonic vascular system. Proc Natl Acad Sci USA 1993;90: 9355–9358. 235. Schnurch H, Risau W. Expression of TIE-2, a member of a novel family of receptor tyrosine kinases, in the endothelial cell lineage. Development 1993;119:957–968. 236. Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre PC, et al. Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by secretion-trap expression cloning. Cell 1996;87:1161–1169.

Translational Review 237. Stratmann A, Risau W, Plate KH. Cell type–specific expression of angiopoietin-1 and angiopoietin-2 suggests a role in glioblastoma angiogenesis. Am J Pathol 1998;153:1459–1466. 238. Sugimachi K, Tanaka S, Taguchi K, Aishima S, Shimada M, Tsuneyoshi M. Angiopoietin switching regulates angiogenesis and progression of human hepatocellular carcinoma. J Clin Pathol 2003;56:854–860. 239. Papapetropoulos A, Fulton D, Mahboubi K, Kalb RG, O’Connor DS, Li F, Altieri DC, Sessa WC. Angiopoietin-1 inhibits endothelial cell apoptosis via the Akt/survivin pathway. J Biol Chem 2000;275:9102– 9105. 240. Hegen A, Koidl S, Weindel K, Marme D, Augustin HG, Fiedler U. Expression of angiopoietin-2 in endothelial cells is controlled by positive and negative regulatory promoter elements. Arterioscler Thromb Vasc Biol 2004;24:1803–1809. 241. Oh H, Takagi H, Suzuma K, Otani A, Matsumura M, Honda Y. Hypoxia and vascular endothelial growth factor selectively upregulate angiopoietin-2 in bovine microvascular endothelial cells. J Biol Chem 1999;274:15732–15739. 242. Lobov IB, Brooks PC, Lang RA. Angiopoietin-2 displays VEGFdependent modulation of capillary structure and endothelial cell survival in vivo. Proc Natl Acad Sci USA 2002;99:11205–11210. 243. Long DA, Price KL, Ioffe E, Gannon CM, Gnudi L, White KE, Yancopoulos GD, Rudge JS, Woolf AS. Angiopoietin-1 therapy enhances fibrosis and inflammation following folic acid–induced acute renal injury. Kidney Int 2008;74:300–309. 244. Kim W, Moon SO, Lee SY, Jang KY, Cho CH, Koh GY, Choi KS, Yoon KH, Sung MJ, Kim DH, et al. Comp–angiopoietin-1 ameliorates renal fibrosis in a unilateral ureteral obstruction model. J Am Soc Nephrol 2006;17:2474–2483. 245. Zhao YD, Campbell AI, Robb M, Ng D, Stewart DJ. Protective role of angiopoietin-1 in experimental pulmonary hypertension. Circ Res 2003;92:984–991. 246. Sullivan CC, Du L, Chu D, Cho AJ, Kido M, Wolf PL, Jamieson SW, Thistlethwaite PA. Induction of pulmonary hypertension by an angiopoietin 1/TIE2/serotonin pathway. Proc Natl Acad Sci USA 2003;100:12331–12336. 247. Dong Z, Kumar R, Yang X, Fidler IJ. Macrophage-derived metalloelastase is responsible for the generation of angiostatin in Lewis lung carcinoma. Cell 1997;88:801–810. 248. Patterson BC, Sang QA. Angiostatin-converting enzyme activities of human matrilysin (MMP-7) and gelatinase B/type IV collagenase (MMP-9). J Biol Chem 1997;272:28823–28825. 249. Stathakis P, Fitzgerald M, Matthias LJ, Chesterman CN, Hogg PJ. Generation of angiostatin by reduction and proteolysis of plasmin: catalysis by a plasmin reductase secreted by cultured cells. J Biol Chem 1997;272:20641–20645. 250. Cao Y, Xue L. Angiostatin. Semin Thromb Hemost 2004;30:83–93. 251. Cao Y, Ji RW, Davidson D, Schaller J, Marti D, Sohndel S, McCance SG, O’Reilly MS, Llinas M, Folkman J. Kringle domains of human angiostatin: characterization of the anti-proliferative activity on endothelial cells. J Biol Chem 1996;271:29461–29467. 252. Claesson-Welsh L, Welsh M, Ito N, Anand-Apte B, Soker S, Zetter B, O’Reilly M, Folkman J. Angiostatin induces endothelial cell apoptosis and activation of focal adhesion kinase independently of the integrinbinding motif RGD. Proc Natl Acad Sci USA 1998;95:5579–5583. 253. Gupta N, Nodzenski E, Khodarev NN, Yu J, Khorasani L, Beckett MA, Kufe DW, Weichselbaum RR. Angiostatin effects on endothelial cells mediated by ceramide and RhoA. EMBO Rep 2001;2:536– 540. 254. Moser TL, Stack MS, Asplin I, Enghild JJ, Hojrup P, Everitt L, Hubchak S, Schnaper HW, Pizzo SV. Angiostatin binds ATP synthase on the surface of human endothelial cells. Proc Natl Acad Sci USA 1999;96:2811–2816. 255. Moser TL, Kenan DJ, Ashley TA, Roy JA, Goodman MD, Misra UK, Cheek DJ, Pizzo SV. Endothelial cell surface F1-F0 ATP synthase is active in ATP synthesis and is inhibited by angiostatin. Proc Natl Acad Sci USA 2001;98:6656–6661. 256. Chavakis E, Dimmeler S. Regulation of endothelial cell survival and apoptosis during angiogenesis. Arterioscler Thromb Vasc Biol 2002; 22:887–893. 257. Jurasz P, Ng D, Granton JT, Courtman DW, Stewart DJ. Elevated platelet angiostatin and circulating endothelial microfragments in idiopathic pulmonary arterial hypertension: a preliminary study. Thromb Res 2010;125:53–60.

15 258. Pascaud M-A, Griscelli F, Raoul W, Marcos E, Opolon P, Raffestin B, Perricaudet M, Adnot S, Eddahibi S. Lung overexpression of angiostatin aggravates pulmonary hypertension in chronically hypoxic mice. Am J Respir Cell Mol Biol 2003;29:449–457. 259. Liu RM. Oxidative stress, plasminogen activator inhibitor 1, and lung fibrosis. Antioxid Redox Signal 2008;10:303–319. 260. Mulligan-Kehoe MJ, Wagner R, Wieland C, Powell R. A truncated plasminogen activator inhibitor-1 protein induces and inhibits angiostatin (kringles 1–3), a plasminogen cleavage product. J Biol Chem 2001;276:8588–8596. 261. Behr J, Kolb M, Cox G. Treating IPF—all or nothing? A pro–con debate. Respirology 2009;14:1072–1081. 262. Gauldie J. Pro: inflammatory mechanisms are a minor component of the pathogenesis of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2002;165:1205–1206. 263. Azuma A, Nukiwa T, Tsuboi E, Suga M, Abe S, Nakata K, Taguchi Y, Nagai S, Itoh H, Ohi M, et al. Double-blind, placebo-controlled trial of pirfenidone in patients with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2005;171:1040–1047. 264. Taniguchi H, Ebina M, Kondoh Y, Ogura T, Azuma A, Suga M, Taguchi Y, Takahashi H, Nakata K, Sato A, et al. Pirfenidone in idiopathic pulmonary fibrosis. Eur Respir J 2010;35:821–829. 265. Demedts M, Behr J, Buhl R, Costabel U, Dekhuijzen R, Jansen HM, MacNee W, Thomeer M, Wallaert B, Laurent F, et al. High-dose acetylcysteine in idiopathic pulmonary fibrosis. N Engl J Med 2005; 353:2229–2242. 266. Kim R, Meyer KC. Review: therapies for interstitial lung disease: past, present and future. Ther Adv Respir Dis 2008;2:319–338. 267. Barst RJ, Gibbs JSR, Ghofrani HA, Hoeper MM, McLaughlin VV, Rubin LJ, Sitbon O, Tapson VF, Galie` N. Updated evidence-based treatment algorithm in pulmonary arterial hypertension. J Am Coll Cardiol 2009;54(1 Suppl 1)S78–S84. 268. Fisher KA, Serlin DM, Wilson KC, Walter RE, Berman JS, Farber HW. Sarcoidosis-associated pulmonary hypertension: outcome with long-term epoprostenol treatment. Chest 2006;130:1481–1488. 269. Strange C, Bolster M, Mazur J, Taylor M, Gossage JR, Silver R. Hemodynamic effects of epoprostenol in patients with systemic sclerosis and pulmonary hypertension. Chest 2000;118:1077–1082. 270. Archer SL, Weir EK, Wilkins MR. Basic science of pulmonary arterial hypertension for clinicians: new concepts and experimental therapies. Circulation 2010;121:2045–2066. 271. Ghofrani HA, Wiedemann R, Rose F, Schermuly RT, Olschewski H, Weissmann N, Gunther A, Walmrath D, Seeger W, Grimminger F. Sildenafil for treatment of lung fibrosis and pulmonary hypertension: a randomised controlled trial. Lancet 2002;360:895–900. 272. Olschewski H, Ghofrani HA, Walmrath D, Schermuly R, TemmesfeldWollbruck B, Grimminger F, Seeger W. Inhaled prostacyclin and iloprost in severe pulmonary hypertension secondary to lung fibrosis. Am J Respir Crit Care Med 1999;160:600–607. 273. King TE Jr, Behr J, Brown KK, du Bois RM, Raghu G. Bosentan use in idiopathic pulmonary fibrosis (ipf): Results of the placebo-controlled build-1 study [abstract]. Proc Am Thorac Soc 2006;3:A524. 274. Kubo H, Nakayama K, Yanai M, Suzuki T, Yamaya M, Watanabe M, Sasaki H. Anticoagulant therapy for idiopathic pulmonary fibrosis. Chest 2005;128:1475–1482. 275. Chen MW, Ni L, Zhao XG, Niu XY. The inhibition of 20(R)-ginsenoside Rg3 on the expressions of angiogenesis factors proteins in human lung adenocarcinoma cell line A549 and HUVEC304 cell. Zhongguo Zhong Yao Za Zhi 2005;30:357–360. 276. Zhao YD, Courtman DW, Deng Y, Kugathasan L, Zhang Q, Stewart DJ. Rescue of monocrotaline-induced pulmonary arterial hypertension using bone marrow–derived endothelial-like progenitor cells: efficacy of combined cell and eNOS gene therapy in established disease. Circ Res 2005;96:442–450. 277. Turner-Warwick M. Systemic arterial patterns in the lung and clubbing of the fingers. Thorax 1963;18:238–250. 278. Paul D, Samuel SM, Maulik N. Mesenchymal stem cell: present challenges and prospective cellular cardiomyoplasty approaches for myocardial regeneration. Antioxid Redox Signal 2009;11:1841–1855. 279. Polomis D, Runo JR, Meyer KC. Pulmonary hypertension in interstitial lung disease. Curr Opin Pulm Med 2008;14:462–469. 280. Baggiolini M, Walz A, Kunkel SL. Neutrophil-activating peptide-1/ interleukin 8, a novel cytokine that activates neutrophils. J Clin Invest 1989;84:1045–1049.