The WNT family of signaling proteins is essential to organ development in general and lung morphogenesis in particular. Originally identified as a developmentally active signaling pathway, the WNT pathway has recently been linked to the pathogenesis of important lung diseases, in particular lung cancer and pulmonary fibrosis. This review summarizes our current understanding about WNT signaling in lung development and disease, and is structured into three chapters. The first chapter presents an introduction to WNT signaling, outlining WNT proteins, their receptors and signaling intermediates, as well as the regulation of this complex pathway. The second chapter focuses on the role of WNT signaling in the normal embryonic and adult lung, and highlights recent findings of altered WNT signaling in lung diseases, such as lung cancer, pulmonary fibrosis, or pulmonary arterial hypertension. In the last chapter, we will discuss novel data and ideas about the biological effects of WNT signaling on the cellular level, highlighting pleiotropic effects induced by WNT ligands on distinct cell types, and how these cellular effects may be relevant to the pathogenesis of the aforementioned diseases.
This review summarizes our current understanding about WNT signaling in lung development and disease. We discuss novel data and ideas about the biological effects of WNT signaling on the cellular level, highlighting pleiotropic effects induced by WNT ligands on distinct cell types, and how these cellular effects may be relevant to the pathogenesis of the aforementioned diseases.
The WNT family constitutes a large family of secreted glycoproteins with highly conserved cysteine residues (1–4). The name “WNT” is derived from a synthesis of the two genes wingless and int-1. The int-1 gene was first described by Nusse and Varmus as a proto-oncogene that caused mammary tumors, when the mouse mammary tumor virus (MMTV) was integrated in the int-1 locus (5, 6). At about the same time, Nüsslein-Volhard and colleagues (7) described that fruit flies (Drosophila melanogaster) lacking the gene wingless were unable to develop wings. In the following years, wingless and int-1 were found to encode identical proteins, and the new name, WNT, was formed (8, 9). To date, 19 different WNT proteins have been identified in humans, most of which have been shown to be expressed in a tightly regulated spatiotemporal manner. Many WNT proteins share a high amino acid sequence homology—in particular, those that are grouped within similar subfamilies (e.g., WNT7a and WNT7b). The array of WNT genes identified and characterized thus far suggests that different WNT ligands can functionally compensate a loss of function of a distinct WNT ligand. Secreted WNT proteins are usually quite insoluble and hydrophobic due to extensive palmitoylation of the mature protein (10). This post-translational modification is critical for proper WNT signaling, but, at the same time, has also delayed the purification, production, and subsequent analysis of recombinant WNT proteins for a long time (11). New insights into the production, secretion, or transportation of WNT ligands have only recently been presented, which are covered in detail by excellent reviews (1, 12).
WNT signaling cascades can be divided into at least the three following distinct pathways. First, the “canonical” WNT/β-catenin pathway involves WNT ligand binding to cell surface receptors and cytosolic stabilization and nuclear translocation of β-catenin for target gene expression. Second, the WNT/Ca2+ pathway signals via calmodulin kinase II and protein kinase C. Third, the WNT/JNK or planar cell polarity pathway signals through small GTPases, and is implicated in cytoskeletal organization and epithelial cell polarity. Some WNT ligands are known to activate both the canonical and the noncanonical pathway, whereas others, such as WNT5a, appear to be specific for noncanonical signaling (1, 3, 10).
The best-characterized WNT signaling pathway is the β-catenin–dependent WNT signaling pathway (Figure 1). Here, in the absence of active WNT ligands, β-catenin is bound to the scaffold proteins Axin and adenomatosis polyposis coli (APC), and constitutively phosphorylated at four N-terminal residues via interaction with casein kinase I and glycogen synthase kinase (GSK)-3β. Phosphorylated β-catenin is a target for subsequent ubiquitination and proteosomal degradation via the β-transducin repeat containing protein.

In the presence of WNT ligands, the membrane receptors, frizzled (FZD) and low-density lipoprotein receptor–related protein (LRP) 5 or 6, are activated upon ligand binding. The FZDs are seven transmembrane spanning receptors, and represent the primary receptor for WNT ligand binding, whereas LRP5/6 are single-pass transmembrane receptors, and represent WNT coreceptors. Up to now, 10 FZDs have been described and investigated (4, 13). In addition, alternative WNT receptors, such as Ryk, have also been identified lately (14). The number and diversity of different WNT ligands and receptors is remarkable, and clearly emphasizes the system's complexity, as well as the ongoing challenge to identify specific ligand–receptor interactions that activate distinct intracellular signaling pathways. This multiplicity of WNT ligand–receptor interactions have recently been covered in great detail (4, 12, 13, 15), and is not within the scope of this review.
In the canonical WNT/β-catenin pathway, WNT ligand binding to FZD and LRP leads to the phosphorylation of LRP6 by GSK-3β and casein kinase γ in its cytoplasmic region, which leads to the recruitment of the cytosolic proteins, Dishevelled (DVL) 1–3 and Axin (Figure 1). Subsequently, β-catenin phosphorylation is inhibited and its degradation attenuated. Accumulated β-catenin then undergoes nuclear translocation, and regulates target gene expression via interaction with members of the T cell–specific transcription factor (TCF)/lymphoid enhancer–binding factor (LEF) family (1, 3). In the absence of β-catenin, TCF and Groucho form a constitutive repressor complex. After its nuclear translocation, β-catenin disrupts this interaction of TCF and Groucho, and regulates gene transcription by binding to its target sequence. The transcriptional profile of the WNT/β-catenin pathway is diverse, and the number of cell-specific, as well as general WNT target genes, is steadily increasing (16–18). WNT target genes also include WNT signaling components themselves, such as Dickkopf (DKK) 1 (19) or Axin2 (20), indicating feedback regulation of this system, as well as several oncogenes (i.e., Cyclin D or c-myc [21, 22]).
The WNT signaling pathway is highly regulated, and different WNT inhibitors have been described thus far (23–25). It has not yet been fully determined, in most cases, whether these proteins consistently inhibit or facilitate WNT signaling, which depends on their expression levels and the cellular context. We therefore prefer to use the term “WNT regulators” rather than “WNT inhibitors.” In general, WNT regulators can be divided into two different groups. The first include secreted FZD receptor proteins (sFRPs) 1, 2, 4, and 5, as well as WNT inhibitory factor (WIF). The sFRPs represent extracellular WNT regulators that directly bind WNT ligands, thereby preventing ligand binding to the signaling receptors, FZD and LRP. The sFRPs exhibit a high degree of homology with the ligand-binding domains of FZD, which explains their binding capacity for WNT ligands (23, 26). Similar mechanisms have been proposed for WIF (27).
The second group of WNT regulators include DKK proteins and Wise. These extracellular proteins use a fundamentally different mechanism to modulate WNT signaling, as DKK and Wise bind to the WNT-binding domain of LRP6 (25, 28). Four different DKK proteins have thus far been discovered, which all share conserved cysteine-rich domains. DKK1 is the founding member of the family, which was originally identified as an embryonic head inducer and WNT inhibitor in Xenopus (29). Whereas DKK1, -2, and -4 share a high degree of sequence homology, DKK3 is less related to the other three DKKs. DKK1 is classically known as a WNT inhibitor, whereas DKK2 can function as either an activator or an inhibitor of WNT signaling, depending on its cellular context (30, 31). Recently, it has been shown that DKK1 and -2 also exhibit a high binding affinity for a second class of receptors, termed Kremen (KRM) (32–34). The formation of a ternary complex of DKK, KRM, and LRP6 is thought to lead to endocytosis of the whole receptor complex, thereby blocking WNT signaling (35–37). Although KRM1 and -2 have been shown to potentiate the ability of DKK to modulate WNT signaling, it is likely that DKK can even modulate WNT signaling in the absence of KRM.
Another WNT regulator (Wise) has recently been identified, which has been shown to interact with LRP6, with the ability to activate or inhibit WNT signaling in a context-dependent manner (28). Most WNT regulators, such as sFRP or DKK, are highly regulated on the transcriptional level, and present direct feedback control of WNT stimulation (19, 20). Most recently, novel WNT regulators have been identified, such as the R-Spondin family (38, 39). R-Spondins are secreted proteins capable of promoting WNT signaling, probably due to binding to FZD and LRP receptors; their precise mechanism of action, however, remains elusive (14).
In addition to the aforementioned regulatory mechanisms, the interactions of WNT proteins with other signaling pathways potentially control WNT action. Several studies have demonstrated that the WNT pathway interacts with other pathways, such as the transforming growth factor (TGF)-β, connective tissue growth factor (CTGF), sonic hedgehog (SHH), or Notch pathway (40–45). These cross-pathway communications play an essential role for the spatiotemporal activity of WNT signaling, and direct evidence of single-pathway interactions will be discussed in the following chapters.
The number of studies investigating WNT signaling in the lung has increased exponentially in recent years. The first evidence of WNT expression in the lung was published in 1990 (46). Subsequent to this finding, the first years of WNT research in the lung have largely focused on lung cancer and lung development. More recently, WNT signaling in lung fibrosis and lung stem cells developed as a promising research area. In the following chapters, we will highlight these studies and summarize the current understanding of WNT signaling in lung development and disease.
The lung arises from a small diverticulum in the anterior foregut endoderm at the laryngotracheal groove. The respiratory epithelium then invades the surrounding mesenchyme, followed by the process of branching morphogenesis (43, 47). This process depends on the precise coordination of epithelial–mesenchymal interactions involving cell–cell and cell–matrix interactions, controlled by an orchestra of secreted growth factors (48). Multiple signaling pathways have been implicated in the control of branching morphogenesis, these include, among others, the fibroblast growth factor, TGF-β, bone-morphogenic proteins (BMPs), SHH, or WNT (47, 49).
WNT signaling is known to regulate epithelial and mesenchymal cell biology in an autocrine and paracrine fashion (48, 50). Several WNT ligands, receptors, and components of the canonical pathway are expressed in a highly cell-specific fashion in the developing lung. For instance, WNT2 is highly expressed in the distal mesenchyme (51), whereas WNT7b is expressed predominantly in the epithelium (52). In addition, WNT5a is expressed in both cell types (53). Despite early and strong expression of WNT2 in the developing lung, transgenic deletion of WNT2 did not result in any detectable defects of lung development and function (54), probably due to functional redundancy of WNT2 proteins. Shortly after this initial study, WNT2b, another WNT2 family member, was identified and characterized; this may have compensated for the loss of WNT2 seen in the initial study (55).
Transgenic deletion of WNT7b, by replacement of the first exon with a lacZ-coding region (WNT7blacZ), causes perinatal death due to respiratory failure. WNT7blacZ mice are characterized by impaired mesenchymal growth and vascular development due to defective autocrine and paracrine WNT signaling in airway epithelium (52). The observed defects in vascular smooth muscle development suggest a role of WNT7b signaling in vascular lung diseases, such as pulmonary arterial hypertension (PAH) (52, 56). In another study, a conditional null allele of the WNT7b gene was generated by removal of exon 3 and, in contrast to the previous report, these mice demonstrated a reduction in the proliferation of both epithelium and mesenchyme, as well as normal vascular smooth muscle development (57). It has been shown that WNT7b consists of two different isoforms generated by two initiating exons. The later study proposed a hypomorphic allele in the WNT7blacZ mice due to the presence of an alternative exon 1, which may lead to partial loss of function, and thus may explain the aforementioned effects (57). On the cellular level, it has been shown that WNT7b is expressed by lung epithelium, and activates the canonical, but not the noncanonical, WNT pathway in vascular smooth muscle cells through FZD1 in a paracrine fashion. Furthermore, WNT7b exhibits autocrine signaling activity via binding to FZD10, which is expressed on the surface of epithelial cells (56).
Deletion of WNT5a by homologous recombination in mice leads to overexpansion of distal airways and expanded interstitium, accompanied by increased SHH expression (53). In addition, transgenic mice with epithelium-specific overexpression of WNT5a exhibited reduced epithelial branching morphogenesis and distal air space enlargement. WNT5a overexpression led to increased mesenchymal fibroblast growth factor signaling and concomitant decreased epithelial SHH signaling (58). This strongly suggests that WNT proteins interact with other signaling pathways in lung development.
Active WNT signaling in lung development has also been demonstrated using transgenic WNT reporter mice and nuclear β-catenin staining. TOPGAL or BATGAL mice, both of which harbor a β-galactosidase gene under the control of a LEF/TCF–inducible promoter fragment (TOP for Tcf-optimal-promoter and BAT for beta-catenin-activated transgene), revealed active canonical WNT signaling early throughout the epithelium and the mesenchyme adjacent to proximal airways at Embryonic Days (E) 10.5–12.5. This signaling disappeared first in the mesenchyme, and was subsequently reduced in the epithelium at E13.5–E18.5 (50, 59, 60).
Epithelial cell–specific expression of constitutively active β-catenin leads to epithelial cell dysplasia and ectopic differentiation of alveolar epithelial type II cells in the conducting airways during embryonic development. Postnatally, these mice exhibit air space enlargement and develop pulmonary tumors (61). Lung epithelial cell–specific deletion of β-catenin, in contrast, results in disruption of branching morphogenesis, with distorted differentiation of the peripheral lung. The mice died neonatally due to respiratory failure (62). Because β-catenin functions as a key regulator of WNT signaling, as well as cell adhesion processes, disruption of β-catenin may not only result in distorted WNT signaling, but also dysregulated cell adhesion (63).
Further evidence for this key role of canonical WNT signaling in epithelial function was provided by targeted overexpression of the WNT regulator, DKK1, in lung epithelium, which resulted in disruption of proximal–distal patterning via inhibition of active WNT signaling (60). DKK1 treatment of mouse lung organ cultures also led to impaired branching morphogenesis and defects in the formation of the pulmonary vascular network (64). In this study, inhibition of fibronectin using a neutralizing antibody led to similar branching defects as observed by DKK1 induction. In contrast, addition of exogenous fibronectin rescued the DKK1-mediated phenotype, emphasizing the importance of extracellular matrix formation in these processes (64). It has to be pointed out, however, that, in an above-mentioned study (58), epithelium-specific overexpression of WNT5a led to an inhibition of epithelial branching morphogenesis.
The fact that some of the above-mentioned studies have resulted in apparent opposite results further underlines the complexity of the WNT signaling system, which is subjected to a tightly regulated spatiotemporal expression pattern in the lung. Further studies—most importantly, in human lung development—are required to clarify these issues. Overall, however, these studies have demonstrated a critical role of properly regulated WNT signaling for normal epithelial–mesenchymal interaction during lung morphogenesis, and emphasized the deleterious impact of distorted WNT signaling on proper lung development.
In the adult lung, most WNT components, including canonical and noncanonical WNT ligands, receptors, regulators, and intracellular signal transducers are expressed at detectable levels. Recently, we have demonstrated that WNT1 and -3a mainly localized to bronchial and alveolar epithelium, along with expression of WNT1 in pulmonary endothelial and smooth muscle cells (65). Quantification of the mRNA levels of canonical WNT/β-catenin signaling components in lung tissue samples of transplant donors revealed that the WNT ligands, WNT1, -2, -3a, and -7b, were expressed at similar levels in normal adult lung tissue, whereas WNT10b exhibited low expression. The most abundant receptors in the human lung were FZD1/4 and LRP5/6. The main canonical WNT signal transducers, GSK-3β and β-catenin, as well as members of the TCF/LEF family, were expressed, with the exception of TCF1 (65). In support of this, Winn and colleagues (66) demonstrated the expression of several WNT proteins in lung epithelial cell lines. Thus, it is evident that the adult lung expresses all required components for WNT signaling, but functional studies thereof are missing thus far.
The role of WNT signaling in various types of cancer is well established, and excellent reviews have been published on this topic (67, 68). This chapter will focus on studies of lung cancer, which is the leading cause of death among cancer patients worldwide. Lung cancer can be separated in to two major forms: non–small cell lung cancer (NSCLC) and small cell lung cancer, which account for 80 and 20% of all lung carcinomas, respectively (69). A multistep oncogenic process, involving tumor suppressors as well as oncogenes, seems to trigger the transition of normal epithelial cells to metaplastic cells, and, subsequently, carcinoma cells (70). Dysregulated WNT signaling in cancer has been primarily found in colon cancer, and more recently, studies focused on lung cancer—in particular, NSCLC (69, 71). It has been shown that mutations in key WNT signaling genes, such as APC or β-catenin, are frequently associated with colon cancer, whereas such mutations seem to be rare in lung cancer (72–74).
Several WNT proteins are differentially expressed in NSCLC specimens, including WNT1, -2, and -7a. WNT1 is overexpressed in NSCLC samples, and cancer cells expressing WNT1 are resistant to apoptotic therapies (75, 76). Conversely, inhibition of WNT1 led to apoptosis in human cancer cells, and reduced tumor growth in vivo and in vitro (75). Likewise, WNT2 was overexpressed in NSCLC, and blockade of WNT2 induced apoptosis in cancer cells (77). Huang and colleagues (78) reported that WNT5a gene expression was higher in squamous cell carcinoma compared with adenocarcinoma, suggesting that WNT5a expression is responsible for more aggressive forms of NSCLC. Interestingly, WNT7a is decreased in NSCLC. Forced overexpression of WNT7a and FZD9 reversed cellular transformation by inhibiting cell growth and inducing epithelial cell differentiation in NSCLC (66). These antitumorigenic effects were mediated by activation of peroxisome proliferator–activated receptor γ, suggesting a noncanonical WNT signaling pathway involved in tumorigenesis (79).
Uematsu and colleagues (80) provided evidence that active WNT signaling in NSCLC is mediated by overexpression of the intracellular signal transducer, DVL. Specifically, DVL3 was overexpressed in microdissected NSCLC samples, and inhibition of DVL decreased β-catenin expression and cell growth (80). The WNT regulator, WIF, as well as sFRP1 and DKK3, are down-regulated in several cancer types, including NSCLC, due to transcriptional silencing via hypermethylation of their promoters (81–85). It has also been reported that promoter hypermethylation of WIF and sFRP1 was more frequent in colorectal metastases compared with the corresponding primary lung adenocarcinomas, whereas APC promoter methylation was significantly more common in primary lung adenocarcinomas (86). This differential methylation pattern may thus allow easier discrimination of primary lung tumors and lung metastases of different primary tumors.
In contrast to the above-mentioned results, the role of β-catenin in lung cancer remains to be fully elucidated. Mutations in the β-catenin gene are rare in lung cancer cell lines or primary lung carcinomas. Several groups have observed reduced expression of β-catenin in lung carcinoma, which was associated with poorer prognosis (87, 88), possibly due to proliferative cellular activity and undifferentiation in NSCLC (89). Conversely, Hommura and colleagues (90) reported that increased β-catenin expression is associated with enhanced cell proliferation and a beneficial prognosis. These results are most probably due to multiple roles of β-catenin in transducing canonical WNT pathways, as well as in regulating cellular adhesion. In sum, increased expression of WNT proteins, along with decreased expression of WNT regulators, is likely involved in the initial phase of tumorigenesis, as well as in the ongoing, multistep process of lung cancerogenesis (Figure 2, left panel).

Aberrant activation of a developmental pathway is a common and well investigated feature of cancer, but this mechanism is largely underappreciated in nonmalignant diseases. New technologies, such as high-throughput whole-genome expression or proteomic profiling, have recently contributed to our understanding of pathways that are involved in disease progression.
In pulmonary fibrosis, studies in animal models, as well as human disease, have provided evidence for the reactivation of developmental programs, including the WNT signaling pathway, which was recently summarized in an excellent review (91). Idiopathic pulmonary fibrosis (IPF), the most common form of idiopathic interstitial pneumonias, represents a progressive and lethal disorder with limited responsiveness to currently available therapies (92–94). It is well accepted that repetitive injury and subsequent repair of alveolar epithelial type II (ATII) cells, in the presence or absence of local inflammation, represent a key pathogenic mechanism in IPF (95–98). This, in turn, leads to aberrant growth factor activation, and perpetuation of fibrotic transformation and aggregation of activated myofibroblasts (fibroblast foci), which promote excessive extracellular matrix (ECM) deposition (99). Fibroblast foci occur in subepithelial layers, close to areas of alveolar epithelial cell injury and repair, suggesting that impaired epithelial–mesenchymal crosstalk contributes to the pathobiology of IPF (95, 100). Unbiased microarray screens have revealed the overexpression of WNT genes, including WNT2 and -5a, the receptors FZD7 and -10, and WNT regulators, such as sFRP1 and -2, in IPF lungs compared with normal lungs or those with other interstitial lung diseases (91, 101–103). In addition, several WNT target genes, such as matrix metalloproteinase 7 (104), osteopontin (105), or WNT1-inducible signaling protein (WISP) 1, were recently identified in experimental and idiopathic lung fibrosis (102, 106, 107). Chilosi and colleagues (108) reported nuclear localization of β-catenin in ATII cells and interstitial fibroblasts in IPF lungs, indicative of activated WNT signaling (109). In addition, we have recently reported that several components of the canonical WNT signaling pathway are overexpressed in IPF (65), as well as in experimental lung fibrosis (107). Canonical WNT signaling components (including WNT ligands, β-catenin, or GSK-3β) localized mainly to the bronchial and alveolar epithelium, as observed by immunohistochemistry and gene expression analysis of ATII cells (65). Importantly, increased activity of the WNT pathway in IPF was documented by increased phosphorylation of LRP6 and GSK-3β (65), which has recently been demonstrated to be a sensitive indicator of WNT activity in tissue sections (110, 111). In addition, we recently demonstrated the activation of canonical WNT signaling in experimental lung fibrosis in vivo, along with increased expression of the WNT target gene, WISP1, in hyperplastic ATII cells. Most importantly, inhibition of WISP1 led to the attenuation of experimental lung fibrosis in vivo (107).
In contrast, the role of WNT signaling in lung inflammation is largely unexplored. Recently, Lewis and colleagues (106) provided a comparison of the gene expression profile of 12 different mouse models of infection, allergy, or lung injury. The authors reported regulation of the WNT signaling pathway only in the mouse model of bleomycin-induced lung fibrosis, but not in any other inflammatory lung disease models. In another study, Douglas and colleagues (112) investigated the resolution phase after lung injury, which is characterized by a decrease of inflammation, re-epithelialization, and matrix remodeling. The authors used a mouse model of oxidant-induced injury with subsequent hyperoxic exposure, and demonstrated increased nuclear β-catenin during the fibroproliferative phase after acute lung injury. The authors showed a concomitant increase in E-cadherin expression and epithelial cell proliferation, supporting a noncanonical cadherin–catenin axis involved in the resolution of inflammation. Both studies strongly suggest a role for WNT signaling in the resolution and regeneration phase after lung injury. It is therefore likely that this resolution process is distorted in IPF (Figure 2, right panel). This is further supported by studies in rheumatoid arthritis, in which WNT proteins induced the expression of cytokines, and appeared to be responsible for an activated phenotype of fibroblast-like synovial cells that exhibit characteristics of immature mesenchymal cells. These cells are involved in long-standing disease, with joint destruction and attempted regeneration (113).
Of interest, WNT signaling crosstalk with other profibrotic growth factors, such as TGF-β, has recently been reported. TGF-β is a key mediator of IPF disease progression, inducing epithelial-to-mesenchymal transition (EMT), fibroblast-to-myofibroblast activation, and extracellular matrix production. With respect to fibrosis, activation of β-catenin by TGF-β in hypertrophic scars has been reported (41). Thus, crosstalk between WNT and TGF-β in lung fibrosis is of major interest that needs to be explored in much more detail. Another growth factor known to be involved in lung fibrosis, CTGF, binds to the WNT coreceptor, LRP6, revealing additional ways of action for CTGF in the fibrotic process (40). Furthermore, expression of SHH is increased in IPF, but not in other interstitial lung diseases (114). In a mouse model of fibrotic disease, but not in nonfibrotic inflammation, SHH and TGF-β were up-regulated in epithelial cells (115).
In sum, WNT signaling seems to be reactivated in response to an as-yet unknown injurious stimulus in IPF, most likely as “attempted regeneration” of lung epithelium. This may explain the findings of hyperplastic epithelium and EMT in IPF lungs (Figure 2, right panel). Unfortunately, this “attempted regeneration” is not only insufficient to restore normal lung architecture in IPF, but, moreover, further drives fibrogenesis and IPF progression by paracrine actions on fibroblasts (65, 107).
PAH is a devastating disorder, characterized by cellular and structural changes in pulmonary arteries (116, 117). Recently, a gene expression analysis of pulmonary arterial resistance vessels revealed differentially regulated canonical and noncanonical WNT genes in PAH (118). One distinct feature of PAH is the increased abundance of α-smooth muscle actin–positive cells in the vessel wall. The possible origin of these cells include: (1) proliferation of resident vascular smooth muscle cells or adventitial fibroblasts; (2) recruitment of vascular progenitor cells; or (3) the transition of endothelial cells into a mesenchymal phenotype, a process that has been suggested to play an important role during cardiovascular development (116–119). The last concept is similar to the process of EMT, which is known to be regulated by WNT signaling, and will be discussed in a separate chapter.
A pathological hallmark of PAH is the “plexiform lesion,” which consists of conglomerates of cells obliterating the vascular lumen. These conglomerates are composed of endothelial cells, smooth muscle cells, mast cells, or lymphocytes (116, 117). In a recent article, Rai and colleagues (120) hypothesized that plexiform lesions reflect complex vascular lesions exhibiting neoplastic features. The authors reported nuclear β-catenin staining and loss of WNT7a in endothelial cells of plexiform lesions in PAH. A recent study demonstrated that BMP2, via its receptor, BMPRII, led to pulmonary arterial endothelial cell proliferation and survival via activation of the WNT signaling pathway (121). Mutations in BMPRII are associated with PAH, suggesting that impaired BMP–WNT signaling is involved in PAH pathogenesis (121).
Although studies investigating WNT signaling in the lung have increased over the past years, our knowledge about the mechanisms and functions of WNT signaling on the cellular level in the lung remains limited. In the following section, we highlight current biological effects of WNT signaling in distinct lung cell types, underlining novel ideas on how this may contribute to the development of chronic lung disease.
Most of the internal surface area of the lung is lined by ATII and type I (ATI) cells, which play a multifunctional role in lung homeostasis, and exhibit a high degree of cell plasticity (122–124). These cells are involved in cell–cell adhesion, ATII-to-ATI cell transition, proliferation, and EMT. WNT signaling has been shown to regulate many of these cellular functions, and, if dysregulated, may contribute to the initiation and progression of several lung diseases—particularly, lung cancer and fibrosis.
EMT is defined as a reversible phenotypic switching of epithelial-to-fibroblast-like cells. EMT has been described in the process of embryonic development, as well as in oncogenic progression and metastasis (125, 126). The orchestrated series of events during EMT involve changes in cell polarity, loss of epithelial cell markers, such as E-cadherin, remodeling of epithelial cell–cell and cell–matrix adhesion contacts, reorganization of the actin cytoskeleton, induction of mesenchymal gene expression, or enhanced cell migration (126, 127).
In lung cancer, EMT is thought to promote cancer invasion and metastasis, and it has been proposed to be a survival strategy for cells to avoid induction of apoptosis by drug treatment (128, 129). Barr and colleagues (130) reported increased resistance to therapies, such as epidermal growth factor receptor inhibition by erlotinib treatment, in cancer cells exhibiting an EMT phenotype. The impact of WNT signaling as a regulator of EMT has been demonstrated by the direct participation of β-catenin in these processes (131, 132), as well as by induction of EMT by WNT ligands in vitro (133). Evidence for the involvement of WNT signaling in the process of EMT in lung cancer in vivo, however, is missing.
In organ fibrosis, accumulating evidence suggests EMT as a possible mechanism that increases the (myo-)fibroblast pool, thereby influencing the process of repair and remodeling after epithelial injury in a number of tissues (134). In pulmonary fibrosis, it was recently demonstrated that TGF-β induces EMT in alveolar epithelial cells in vitro and in vivo (135–137), and that β-catenin is involved in the process of EMT in alveolar epithelial cells (138). Furthermore, epithelial as well as mesenchymal markers colocalize to hyperplastic ATII cells in lung tissue of patients with IPF, suggesting that EMT represents a significant mechanism contributing to IPF pathogenesis (136). Recent studies revealed that the WNT target gene, WISP1, induces EMT in vitro, further highlighting the role of WNT signaling in the process of EMT in lung fibrosis (107).
A key feature of all epithelial cells is the generation of tight cell–cell adhesions. In several lung diseases, the normal alveolar epithelial cell layer is disrupted, and epithelial cells exhibit a proliferative or migratory phenotype, facilitating processes, such as epithelial cell transformation, EMT, or metastasis (125, 127, 139). Cadherins are cell adhesion receptors, such as E-cadherin in epithelial cells, which interact with β-catenin (140, 141). This is of particular interest, as β-catenin is one of the key signaling molecules in WNT signaling. This dual role of β-catenin emphasizes the close connection between cell adhesion and WNT signaling (142–144). This was highlighted in two recent studies reporting different conformational changes of β-catenin that allow its discrimination of WNT signaling and cell adhesion participation (145, 146). It is thought that only a minority of the cellular β-catenin content in epithelial cells contributes to WNT signaling, whereas the majority of β-catenin molecules is present within the cytosol and at cell–cell contacts (144, 147). Future studies are needed to understand how β-catenin is able to differentiate between these two pathways—in particular, in lung epithelium.
In NSCLC, enhanced expression of WNT1 and -2 drives epithelial cell proliferation, and inhibition thereof induces epithelial cell apoptosis (75, 77). In contrast, WNT7a expression is decreased in NSCLC, and gain-of-function studies of WNT7a in NSCLC cell lines revealed a reversal of cell transformation and growth (66, 79). Furthermore, WNT7a induces the expression of E-cadherin, also suggesting an involvement of WNT7a in the prevention/reversal of EMT (148).
In IPF, initial alveolar epithelial cell damage and subsequent impaired repair are thought to represent key pathogenic features (95, 97, 149). Hyperplastic, proliferating alveolar epithelial cells are frequently found in pulmonary fibrosis, and alveolar epithelial cell proliferation may be involved in this impaired repair (107, 150–152). Recently, we reported increased proliferation of alveolar epithelial cells, in concert with up-regulation of WNT target genes, in response to canonical WNT activation, suggesting a role of active WNT signaling in impaired epithelial cell repair mechanism (65, 107). It has to be highlighted that an increased incidence of lung cancer in IPF suggests a link between epithelial cell hyperplasia, impaired repair, and carcinogenesis (153–155). While further research is clearly required to elucidate these associations, studies thus far indicate active WNT signaling as a common molecular mechanism linking alveolar epithelial cell transformation and hyperplasia with fibrosis or cancer.
Proliferation and activation of (myo-)fibroblasts is a key feature in both lung cancer and lung fibrosis (156, 157). The (myo-)fibroblasts contribute to tumor stroma and fibroblast accumulation in IPF by enhanced production of ECM and secretion of growth factors. Recent studies have demonstrated the capability of lung fibroblasts to respond to WNT stimulation (65, 107, 158–160). Gene expression profiling of human lung fibroblasts treated with WNT3a revealed several new WNT-induced genes in fibroblasts, among these genes that play a role in fibroblast-to-myofibroblast differentiation (158). In addition, Chen and colleagues (160) reported differences in the expression of genes involved in cell motility/adhesion, cell cycle, or cytoskeletal formation, in WNT3a-stimulated NIH-3T3 cells. Furthermore, functional studies on NIH-3T3 and lung fibroblasts revealed activation and increased collagen deposition in response to canonical WNT signaling (65) or WISP1 (107). Taken together, these data support a role of WNT signaling on profibrotic phenotypes of lung fibroblasts—in particular, via paracrine effects of epithelium-derived WNT ligands.
An emerging area of research is dedicated to the role of WNT signaling in somatic or adult stem cell maintenance and differentiation. Stem cells were first identified as human bone marrow cells that exhibit the potential for proliferation, self-renewal, and differentiation into an array of different cell types. These properties favored stem cells for a range of therapeutic applications and, in the last years, have attracted huge scientific interest. Several studies have characterized stem cell populations and their ability to repair damaged organs. WNT proteins, for the most part, have been proposed to regulate stem cell maintenance and self-renewal (11, 69, 161, 162).
In the lung, local stem cell niches in the airway epithelium have been described (163–165). A number of cells at the bronchoalveolar duct junction exhibit features of stem cells. These “bronchoalveolar stem cells” (BASCs) coexpress Clara and alveolar epithelial cell marker proteins, and appear to be refractory to injury due to naphthalene treatment (166). BASCs divide after induction of damage, which results in repair of epithelial tissue. Different from other tissue stem cells, only rare BASCs may be activated to participate in epithelial repair after depletion of so-called “transit-amplifying cells,” cells characterized as progenies of tissue stem cells with proliferative capacity (69, 165, 167). Recently, the involvement of WNT signaling in lung stem cell maintenance and activation has been pointed out (168–170). Zhang and colleagues (168) demonstrated that the transcription factor, GATA6, influences the temporal appearance and number of BASC in the lung by regulating WNT signaling. Loss of GATA6 led to increased WNT signaling and BASC expansion, with decreased differentiation. Furthermore, Reynolds and colleagues (169) provide evidence that β-catenin affects stem cell maintenance within the slowly renewing epithelium of the lung. Conditional potentiation of β-catenin signaling in the embryonic lung results in amplification of rather undifferentiated airway stem cells. In contrast, a recent study from the same group suggests that β-catenin signaling is not essential for stem cell maintenance in the adult lung, suggesting important differences in the regulation of β-catenin in the developing and adult lung (171).
Along these lines, it has been suggested that BASCs may give rise to cancer stem cells in lung cancer and be involved in epithelial cell transformation, thus fostering cancer development (166). Lately, in addition to BASC, the role of mesenchymal stem cells (MSCs) in the lung has become of interest (172, 173). Importantly, it has been reported that MSCs exhibit a distinct WNT profile, which takes part in lineage specification (174). In lung cancer, a contribution of MSCs was shown by in vitro migration studies that demonstrated an enhanced migration of MSC toward tumor cells. Furthermore, MSCs that expressed the TNF-related apoptosis-inducing ligand inhibited tumor growth when applied to an experimental lung cancer model (175), underlining the potential of MSCs as a novel therapeutic strategy. In other cancers, however, the application of MSCs has been shown to promote tumor progression and invasion (176).
In lung fibrosis, fibroblasts were classically thought to be of resident tissue origin; however, recent experiments in mouse models, as well as human fibrosis, revealed increased recruitment of bone marrow–derived cells at sites of lung fibrosis (177–181). Ortiz and colleagues (178) reported that systemically administered MSCs home to the lung in response to injury, adopt an epithelial-like phenotype, and reduce the amount of fibrosis. On the other hand, circulating fibrocytes, derived from hematopoietic stem cells in the blood, have been described to contribute to the fibroblast pool in lung fibrosis, thus promoting fibrogenesis (180).
In addition to bone marrow–derived MSCs, a recent study of cells derived from human lung allografts revealed the existence of a multipotent mesenchymal cell population, which is locally resident in the adult human lung and exhibits extended life span in vivo (182). Future studies are clearly needed to improve our knowledge about the origin and contribution of different stem cells in chronic lung diseases—in particular, with respect to WNT signaling in these processes.
It is safe to state at this point in time that the WNT signaling pathway is involved in chronic lung disease. Modulation of the WNT pathway may, therefore, present as a suitable and promising therapeutic strategy in the future. Successful implementation of therapeutic modulation of WNT signaling, however, clearly requires further elucidation of the following disease-specific aspects. In lung carcinoma, increased expression of WNT proteins, along with decreased expression of WNT regulators, is likely involved in the initial phase of tumorigenesis, as well as in the ongoing multistep process of lung cancerogenesis, thus representing an initial “failure signal” in epithelial cells. In lung fibrosis, studies thus far suggest that WNT signaling is activated after an initial injury, thus, most likely representing a regeneration signal of the damaged epithelium. Unfortunately, this “attempted regeneration” is not only insufficient to restore normal lung architecture, but, moreover, drives fibrogenesis and IPF progression by, for example, epithelial cell transformation or EMT, fibroblast proliferation and activation, or interaction with other profibrotic growth factors, altogether perpetuating fibrogenesis.
1. | Logan CY, Nusse R. The WNT signaling pathway in development and disease. Annu Rev Cell Dev Biol 2004;20:781–810. |
2. | Dale TC. Signal transduction by the WNT family of ligands. Biochem J 1998;329:209–223. |
3. | Moon RT, Kohn AD, De Ferrari GV, Kaykas A. WNT and beta-catenin signalling: Diseases and therapies. Nat Rev Genet 2004;5:691–701. |
4. | Cadigan KM, Liu YI. WNT signaling: complexity at the surface. J Cell Sci 2006;119:395–402. |
5. | Nusse R, van Ooyen A, Cox D, Fung YK, Varmus H. Mode of proviral activation of a putative mammary oncogene (INT-1) on mouse chromosome 15. Nature 1984;307:131–136. |
6. | Nusse R, Varmus HE. Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell 1982;31:99–109. |
7. | Nüsslein-Volhard C, Wieschaus E. Mutations affecting segment number and polarity in Drosophila. Nature 1980;287:795–801. |
8. | Rijsewijk F, Schuermann M, Wagenaar E, Parren P, Weigel D, Nusse R. The Drosophila homolog of the mouse mammary oncogene INT-1 is identical to the segment polarity gene wingless. Cell 1987;50:649–657. |
9. | Nusse R, Brown A, Papkoff J, Scambler P, Shackleford G, McMahon A, Moon R, Varmus H. A new nomenclature for INT -1 and related genes: the WNT gene family. Cell 1991;64:231. |
10. | Mikels AJ, Nusse R. WNTs as ligands: processing, secretion and reception. Oncogene 2006;25:7461–7468. |
11. | Willert K, Brown JD, Danenberg E, Duncan AW, Weissman IL, Reya T, Yates JR III, Nusse R. WNT proteins are lipid-modified and can act as stem cell growth factors. Nature 2003;423:448–452. |
12. | Kikuchi A, Yamamoto H, Kishida S. Multiplicity of the interactions of WNT proteins and their receptors. Cell Signal 2007;19:659–671. |
13. | Gordon MD, Nusse R. WNT signaling: multiple pathways, multiple receptors, and multiple transcription factors. J Biol Chem 2006;281:22429–22433. |
14. | Hendrickx M, Leyns L. Non-conventional frizzled ligands and WNT receptors. Dev Growth Differ 2008;50:229–243. |
15. | MacDonald BT, Yokota C, Tamai K, Zeng X, He X. WNT signal amplification via activity, cooperativity, and regulation of multiple intracellular pppsp motifs in the WNT co-receptor LRP6. J Biol Chem 2008;283:16115–16123. |
16. | Nusse R. WNT targets: repression and activation. Trends Genet 1999;15:1–3. |
17. | Moon RT, Bowerman B, Boutros M, Perrimon N. The promise and perils of WNT signaling through beta-catenin. Science 2002;296:1644–1646. |
18. | Vlad A, Rohrs S, Klein-Hitpass L, Muller O. The first five years of the WNT targetome. Cell Signal 2008;20:795–802. |
19. | Niida A, Hiroko T, Kasai M, Furukawa Y, Nakamura Y, Suzuki Y, Sugano S, Akiyama T. DKK 1, a negative regulator of WNT signaling, is a target of the beta-catenin/TCF pathway. Oncogene 2004;23:8520–8526. |
20. | Yan D, Wiesmann M, Rohan M, Chan V, Jefferson AB, Guo L, Sakamoto D, Caothien RH, Fuller JH, Reinhard C, et al. Elevated expression of axin2 and hNKd mRNA provides evidence that WNT/beta-catenin signaling is activated in human colon tumors. Proc Natl Acad Sci USA 2001;98:14973–14978. |
21. | He TC, Sparks AB, Rago C, Hermeking H, Zawel L, da Costa LT, Morin PJ, Vogelstein B, Kinzler KW. Identification of c-myc as a target of the APC pathway. Science 1998;281:1509–1512. |
22. | Tetsu O, McCormick F. Beta-catenin regulates expression of cyclin d1 in colon carcinoma cells. Nature 1999;398:422–426. |
23. | Kawano Y, Kypta R. Secreted antagonists of the WNT signalling pathway. J Cell Sci 2003;116:2627–2634. |
24. | Shi Y, He B, You L, Jablons DM. Roles of secreted frizzled-related proteins in cancer. Acta Pharmacol Sin 2007;28:1499–1504. |
25. | Niehrs C. Function and biological roles of the Dickkopf family of WNT modulators. Oncogene 2006;25:7469–7481. |
26. | Bovolenta P, Esteve P, Ruiz JM, Cisneros E, Lopez-Rios J. Beyond WNT inhibition: new functions of secreted frizzled-related proteins in development and disease. J Cell Sci 2008;121:737–746. |
27. | Hsieh JC, Kodjabachian L, Rebbert ML, Rattner A, Smallwood PM, Samos CH, Nusse R, Dawid IB, Nathans J. A new secreted protein that binds to WNT proteins and inhibits their activities. Nature 1999;398:431–436. |
28. | Itasaki N, Jones CM, Mercurio S, Rowe A, Domingos PM, Smith JC, Krumlauf R. Wise, a context-dependent activator and inhibitor of WNT signalling. Development 2003;130:4295–4305. |
29. | Glinka A, Wu W, Delius H, Monaghan AP, Blumenstock C, Niehrs C. Dickkopf-1 is a member of a new family of secreted proteins and functions in head induction. Nature 1998;391:357–362. |
30. | Wu W, Glinka A, Delius H, Niehrs C. Mutual antagonism between Dickkopf 1 and Dickkopf 2 regulates WNT/beta-catenin signalling. Curr Biol 2000;10:1611–1614. |
31. | Mao B, Niehrs C. Kremen2 modulates Dickkopf 2 activity during WNT/LRP6 signaling. Gene 2003;302:179–183. |
32. | Mao B, Wu W, Davidson G, Marhold J, Li M, Mechler BM, Delius H, Hoppe D, Stannek P, Walter C, et al. Kremen proteins are Dickkopf receptors that regulate WNT/beta-catenin signalling. Nature 2002;417:664–667. |
33. | Nakamura T, Nakamura T, Matsumoto K. The functions and possible significance of Kremen as the gatekeeper of WNT signalling in development and pathology. J Cell Mol Med 2008;12:391–408. |
34. | Wang K, Zhang Y, Li X, Chen L, Wang H, Wu J, Zheng J, Wu D. Characterization of the Kremen-binding site on DKK1 and elucidation of the role of Kremen in DKK-mediated WNT antagonism. J Biol Chem 2008;283:23371–23375. |
35. | Bafico A, Liu G, Yaniv A, Gazit A, Aaronson SA. Novel mechanism of WNT signalling inhibition mediated by Dickkopf-1 interaction with LRP6/arrow. Nat Cell Biol 2001;3:683–686. |
36. | Brott BK, Sokol SY. Regulation of WNT/LRP signaling by distinct domains of Dickkopf proteins. Mol Cell Biol 2002;22:6100–6110. |
37. | Mao B, Wu W, Li Y, Hoppe D, Stannek P, Glinka A, Niehrs C. LDL-receptor–related protein 6 is a receptor for Dickkopf proteins. Nature 2001;411:321–325. |
38. | Kim KA, Wagle M, Tran K, Zhan X, Dixon MA, Liu S, Gros D, Korver W, Yonkovich S, Tomasevic N, et al. R-spondin family members regulate the WNT pathway by a common mechanism. Mol Biol Cell 2008;19:2588–2596. |
39. | Nam JS, Turcotte TJ, Yoon JK. Dynamic expression of R-spondin family genes in mouse development. Gene Expr Patterns 2007;7:306–312. |
40. | Mercurio S, Latinkic B, Itasaki N, Krumlauf R, Smith JC. Connective-tissue growth factor modulates WNT signalling and interacts with the WNT receptor complex. Development 2004;131:2137–2147. |
41. | Sato M. Upregulation of the WNT/beta-catenin pathway induced by transforming growth factor-beta in hypertrophic scars and keloids. Acta Derm Venereol 2006;86:300–307. |
42. | Hing HK, Sun X, Artavanis-Tsakonas S. Modulation of wingless signaling by notch in Drosophila. Mech Dev 1994;47:261–268. |
43. | Warburton D, Bellusci S, De Langhe S, Del Moral PM, Fleury V, Mailleux A, Tefft D, Unbekandt M, Wang K, Shi W. Molecular mechanisms of early lung specification and branching morphogenesis. Pediatr Res 2005;57:26R–37R. |
44. | Reddy S, Andl T, Bagasra A, Lu MM, Epstein DJ, Morrisey EE, Millar SE. Characterization of WNT gene expression in developing and postnatal hair follicles and identification of WNT5a as a target of sonic hedgehog in hair follicle morphogenesis. Mech Dev 2001;107:69–82. |
45. | Watt FM. Unexpected hedgehog–WNT interactions in epithelial differentiation. Trends Mol Med 2004;10:577–580. |
46. | Gavin BJ, McMahon JA, McMahon AP. Expression of multiple novel WNT-1/INT-1–related genes during fetal and adult mouse development. Genes Dev 1990;4:2319–2332. |
47. | Cardoso WV, Lu J. Regulation of early lung morphogenesis: questions, facts and controversies. Development 2006;133:1611–1624. |
48. | Shannon JM, Hyatt BA. Epithelial–mesenchymal interactions in the developing lung. Annu Rev Physiol 2004;66:625–645. |
49. | Hogan BL, Yingling JM. Epithelial/mesenchymal interactions and branching morphogenesis of the lung. Curr Opin Genet Dev 1998;8:481–486. |
50. | Okubo T, Hogan BL. Hyperactive WNT signaling changes the developmental potential of embryonic lung endoderm. J Biol 2004;3:11. |
51. | Levay-Young BK, Navre M. Growth and developmental regulation of WNT-2 (IRP) gene in mesenchymal cells of fetal lung. Am J Physiol 1992;262:L672–L683. |
52. | Shu W, Jiang YQ, Lu MM, Morrisey EE. WNT7b regulates mesenchymal proliferation and vascular development in the lung. Development 2002;129:4831–4842. |
53. | Li C, Xiao J, Hormi K, Borok Z, Minoo P. WNT5a participates in distal lung morphogenesis. Dev Biol 2002;248:68–81. |
54. | Monkley SJ, Delaney SJ, Pennisi DJ, Christiansen JH, Wainwright BJ. Targeted disruption of the WNT2 gene results in placentation defects. Development 1996;122:3343–3353. |
55. | Zakin LD, Mazan S, Maury M, Martin N, Guenet JL, Brulet P. Structure and expression of WNT13, a novel mouse WNT2 related gene. Mech Dev 1998;73:107–116. |
56. | Wang Z, Shu W, Lu MM, Morrisey EE. WNT7b activates canonical signaling in epithelial and vascular smooth muscle cells through interactions with FZD1, FZD10, and LRP5. Mol Cell Biol 2005;25:5022–5030. |
57. | Rajagopal J, Carroll TJ, Guseh JS, Bores SA, Blank LJ, Anderson WJ, Yu J, Zhou Q, McMahon AP, Melton DA. WNT7b stimulates embryonic lung growth by coordinately increasing the replication of epithelium and mesenchyme. Development 2008;135:1625–1634. |
58. | Li C, Hu L, Xiao J, Chen H, Li JT, Bellusci S, Delanghe S, Minoo P. WNT5a regulates SHH and FGF10 signaling during lung development. Dev Biol 2005;287:86–97. |
59. | Maretto S, Cordenonsi M, Dupont S, Braghetta P, Broccoli V, Hassan AB, Volpin D, Bressan GM, Piccolo S. Mapping WNT/beta-catenin signaling during mouse development and in colorectal tumors. Proc Natl Acad Sci USA 2003;100:3299–3304. |
60. | Shu W, Guttentag S, Wang Z, Andl T, Ballard P, Lu MM, Piccolo S, Birchmeier W, Whitsett JA, Millar SE, et al. WNT/beta-catenin signaling acts upstream of n-myc, BMP4, and FGF signaling to regulate proximal–distal patterning in the lung. Dev Biol 2005;283:226–239. |
61. | Mucenski ML, Nation JM, Thitoff AR, Besnard V, Xu Y, Wert SE, Harada N, Taketo MM, Stahlman MT, Whitsett JA. Beta-catenin regulates differentiation of respiratory epithelial cells in vivo. Am J Physiol 2005;289:L971–L979. |
62. | Mucenski ML, Wert SE, Nation JM, Loudy DE, Huelsken J, Birchmeier W, Morrisey EE, Whitsett JA. Beta-catenin is required for specification of proximal/distal cell fate during lung morphogenesis. J Biol Chem 2003;278:40231–40238. |
63. | Dean CH, Miller LA, Smith AN, Dufort D, Lang RA, Niswander LA. Canonical WNT signaling negatively regulates branching morphogenesis of the lung and lacrimal gland. Dev Biol 2005;286:270–286. |
64. | De Langhe SP, Sala FG, Del Moral PM, Fairbanks TJ, Yamada KM, Warburton D, Burns RC, Bellusci S. Dickkopf-1 (DKK1) reveals that fibronectin is a major target of WNT signaling in branching morphogenesis of the mouse embryonic lung. Dev Biol 2005;277:316–331. |
65. | Königshoff M, Balsara N, Pfaff EM, Kramer M, Chrobak I, Seeger W, Eickelberg O. Functional WNT signaling is increased in idiopathic pulmonary fibrosis. PLoS One 2008;3:e2142. |
66. | Winn RA, Marek L, Han SY, Rodriguez K, Rodriguez N, Hammond M, Van Scoyk M, Acosta H, Mirus J, Barry N, et al. Restoration of WNT-7a expression reverses non-small cell lung cancer cellular transformation through frizzled-9–mediated growth inhibition and promotion of cell differentiation. J Biol Chem 2005;280:19625–19634. |
67. | Paul S, Dey A. WNT signaling and cancer development: therapeutic implication. Neoplasma 2008;55:165–176. |
68. | Nusse R. WNT signaling in disease and in development. Cell Res 2005;15:28–32. |
69. | He B, Jablons DM. WNT signaling in stem cells and lung cancer. Ernst Schering Found Symp Proc 2006;(5):27–58. |
70. | Sato M, Shames DS, Gazdar AF, Minna JD. A translational view of the molecular pathogenesis of lung cancer. J Thorac Oncol 2007;2:327–343. |
71. | Van Scoyk M, Randall J, Sergew A, Williams LM, Tennis M, Winn RA. WNT signaling pathway and lung disease. Transl Res 2008;151:175–180. |
72. | Ueda M, Gemmill RM, West J, Winn R, Sugita M, Tanaka N, Ueki M, Drabkin HA. Mutations of the beta- and gamma-catenin genes are uncommon in human lung, breast, kidney, cervical and ovarian carcinomas. Br J Cancer 2001;85:64–68. |
73. | Ohgaki H, Kros JM, Okamoto Y, Gaspert A, Huang H, Kurrer MO. APC mutations are infrequent but present in human lung cancer. Cancer Lett 2004;207:197–203. |
74. | Mazieres J, He B, You L, Xu Z, Jablons DM. WNT signaling in lung cancer. Cancer Lett 2005;222:1–10. |
75. | He B, You L, Uematsu K, Xu Z, Lee AY, Matsangou M, McCormick F, Jablons DM. A monoclonal antibody against WNT-1 induces apoptosis in human cancer cells. Neoplasia 2004;6:7–14. |
76. | Chen S, Guttridge DC, You Z, Zhang Z, Fribley A, Mayo MW, Kitajewski J, Wang CY. WNT-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor–mediated transcription. J Cell Biol 2001;152:87–96. |
77. | You L, He B, Xu Z, Uematsu K, Mazieres J, Mikami I, Reguart N, Moody TW, Kitajewski J, McCormick F, et al. Inhibition of WNT-2–mediated signaling induces programmed cell death in non–small-cell lung cancer cells. Oncogene 2004;23:6170–6174. |
78. | Huang CL, Liu D, Nakano J, Ishikawa S, Kontani K, Yokomise H, Ueno M. WNT5a expression is associated with the tumor proliferation and the stromal vascular endothelial growth factor: an expression in non–small-cell lung cancer. J Clin Oncol 2005;23:8765–8773. |
79. | Winn RA, Van Scoyk M, Hammond M, Rodriguez K, Crossno JT Jr, Heasley LE, Nemenoff RA. Antitumorigenic effect of WNT 7a and FZD 9 in non–small cell lung cancer cells is mediated through ERK-5–dependent activation of peroxisome proliferator–activated receptor gamma. J Biol Chem 2006;281:26943–26950. |
80. | Uematsu K, He B, You L, Xu Z, McCormick F, Jablons DM. Activation of the WNT pathway in non small cell lung cancer: evidence of Dishevelled overexpression. Oncogene 2003;22:7218–7221. |
81. | Kim J, You L, Xu Z, Kuchenbecker K, Raz D, He B, Jablons D. WNT inhibitory factor inhibits lung cancer cell growth. J Thorac Cardiovasc Surg 2007;133:733–737. |
82. | Mazieres J, He B, You L, Xu Z, Lee AY, Mikami I, Reguart N, Rosell R, McCormick F, Jablons DM. WNT inhibitory factor-1 is silenced by promoter hypermethylation in human lung cancer. Cancer Res 2004;64:4717–4720. |
83. | Wissmann C, Wild PJ, Kaiser S, Roepcke S, Stoehr R, Woenckhaus M, Kristiansen G, Hsieh JC, Hofstaedter F, Hartmann A, et al. Wif1, a component of the WNT pathway, is down-regulated in prostate, breast, lung, and bladder cancer. J Pathol 2003;201:204–212. |
84. | Yue W, Sun Q, Dacic S, Landreneau RJ, Siegfried JM, Yu J, Zhang L. Downregulation of DKK3 activates beta-catenin/TCF-4 signaling in lung cancer. Carcinogenesis 2008;29:84–92. |
85. | Lee AY, He B, You L, Dadfarmay S, Xu Z, Mazieres J, Mikami I, McCormick F, Jablons DM. Expression of the secreted Frizzled–related protein gene family is downregulated in human mesothelioma. Oncogene 2004;23:6672–6676. |
86. | Tang M, Torres-Lanzas J, Lopez-Rios F, Esteller M, Sanchez-Cespedes M. WNT signaling promoter hypermethylation distinguishes lung primary adenocarcinomas from colorectal metastasis to the lung. Int J Cancer 2006;119:2603–2606. |
87. | Retera JM, Leers MP, Sulzer MA, Theunissen PH. The expression of beta-catenin in non–small-cell lung cancer: A clinicopathological study. J Clin Pathol 1998;51:891–894. |
88. | Kase S, Sugio K, Yamazaki K, Okamoto T, Yano T, Sugimachi K. Expression of E-cadherin and beta-catenin in human non–small cell lung cancer and the clinical significance. Clin Cancer Res 2000;6:4789–4796. |
89. | Pirinen RT, Hirvikoski P, Johansson RT, Hollmen S, Kosma VM. Reduced expression of alpha-catenin, beta-catenin, and gamma-catenin is associated with high cell proliferative activity and poor differentiation in non–small cell lung cancer. J Clin Pathol 2001;54:391–395. |
90. | Hommura F, Furuuchi K, Yamazaki K, Ogura S, Kinoshita I, Shimizu M, Moriuchi T, Katoh H, Nishimura M, Dosaka-Akita H. Increased expression of beta-catenin predicts better prognosis in nonsmall cell lung carcinomas. Cancer 2002;94:752–758. |
91. | Selman M, Pardo A, Kaminski N. Idiopathic pulmonary fibrosis: aberrant recapitulation of developmental programs? PLoS Med 2008;5:e62. |
92. | Martinez FJ, Safrin S, Weycker D, Starko KM, Bradford WZ, King TE Jr, Flaherty KR, Schwartz DA, Noble PW, Raghu G, et al. The clinical course of patients with idiopathic pulmonary fibrosis. Ann Intern Med 2005;142:963–967. |
93. | Walter N, Collard HR, King TE Jr. Current perspectives on the treatment of idiopathic pulmonary fibrosis. Proc Am Thorac Soc 2006;3:330–338. |
94. | American Thoracic Society/European Respiratory Society international multidisciplinary consensus classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med 2002;165:277–304. |
95. | Selman M, King TE, Pardo A. Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med 2001;134:136–151. |
96. | Chapman HA. Disorders of lung matrix remodeling. J Clin Invest 2004;113:148–157. |
97. | Horowitz JC, Thannickal VJ. Epithelial–mesenchymal interactions in pulmonary fibrosis. Semin Respir Crit Care Med 2006;27:600–612. |
98. | Strieter RM. Pathogenesis and natural history of usual interstitial pneumonia: the whole story or the last chapter of a long novel. Chest 2005;128(5 Suppl 1)526S–532S. |
99. | Visscher DW, Myers JL. Histologic spectrum of idiopathic interstitial pneumonias. Proc Am Thorac Soc 2006;3:322–329. |
100. | Noble PW. Idiopathic pulmonary fibrosis: natural history and prognosis. Clin Chest Med 2006;27(1 Suppl 1)S11–S16. |
101. | Kaminski N, Rosas IO. Gene expression profiling as a window into idiopathic pulmonary fibrosis pathogenesis: can we identify the right target genes? Proc Am Thorac Soc 2006;3:339–344. |
102. | Selman M, Pardo A, Barrera L, Estrada A, Watson SR, Wilson K, Aziz N, Kaminski N, Zlotnik A. Gene expression profiles distinguish idiopathic pulmonary fibrosis from hypersensitivity pneumonitis. Am J Respir Crit Care Med 2006;173:188–198. |
103. | 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. |
104. | Zuo F, Kaminski N, Eugui E, Allard J, Yakhini Z, Ben-Dor A, Lollini L, Morris D, Kim Y, DeLustro B, et al. Gene expression analysis reveals matrilysin as a key regulator of pulmonary fibrosis in mice and humans. Proc Natl Acad Sci USA 2002;99:6292–6297. |
105. | Pardo A, Gibson K, Cisneros J, Richards TJ, Yang Y, Becerril C, Yousem S, Herrera I, Ruiz V, Selman M, et al. Up-regulation and profibrotic role of osteopontin in human idiopathic pulmonary fibrosis. PLoS Med 2005;2:e251. |
106. | Lewis CC, Yang JY, Huang X, Banerjee SK, Blackburn MR, Baluk P, McDonald DM, Blackwell TS, Nagabhushanam V, Peters W, et al. Disease-specific gene expression profiling in multiple models of lung disease. Am J Respir Crit Care Med 2008;177:376–387. |
107. | Königshoff M, Kramer M, Balsara N, Wilhelm J, Amarie OV, Jahn A, Rose F, Fink L, Seeger W, Schaefer L, et al. WNT1-inducible signaling protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. J Clin Invest 2009;119:772–787. |
108. | Chilosi M, Poletti V, Zamo A, Lestani M, Montagna L, Piccoli P, Pedron S, Bertaso M, Scarpa A, Murer B, et al. Aberrant WNT/beta-catenin pathway activation in idiopathic pulmonary fibrosis. Am J Pathol 2003;162:1495–1502. |
109. | Morrisey EE. WNT signaling and pulmonary fibrosis. Am J Pathol 2003;162:1393–1397. |
110. | Bilic J, Huang YL, Davidson G, Zimmermann T, Cruciat CM, Bienz M, Niehrs C. WNT induces injury and promotes Dishevelled-dependent LRP6 phosphorylation. Science 2007;316:1619–1622. |
111. | Forde JE, Dale TC. Glycogen synthase kinase 3: a key regulator of cellular fate. Cell Mol Life Sci 2007;64:1930–1944. |
112. | Douglas IS, Diaz del Valle F, Winn RA, Voelkel NF. Beta-catenin in the fibroproliferative response to acute lung injury. Am J Respir Cell Mol Biol 2006;34:274–285. |
113. | Sen M, Lauterbach K, El-Gabalawy H, Firestein GS, Corr M, Carson DA. Expression and function of Wingless and Frizzled homologs in rheumatoid arthritis. Proc Natl Acad Sci USA 2000;97:2791–2796. |
114. | Coon DR, Roberts DJ, Loscertales M, Kradin R. Differential epithelial expression of SHH and FOXf1 in usual and nonspecific interstitial pneumonia. Exp Mol Pathol 2006;80:119–123. |
115. | Stewart GA, Hoyne GF, Ahmad SA, Jarman E, Wallace WA, Harrison DJ, Haslett C, Lamb JR, Howie SE. Expression of the developmental sonic hedgehog (SHH) signalling pathway is up-regulated in chronic lung fibrosis and the SHH receptor Patched 1 is present in circulating T lymphocytes. J Pathol 2003;199:488–495. |
116. | Humbert M, Morrell NW, Archer SL, Stenmark KR, MacLean MR, Lang IM, Christman BW, Weir EK, Eickelberg O, Voelkel NF, et al. Cellular and molecular pathobiology of pulmonary arterial hypertension. J Am Coll Cardiol 2004;43(12 Suppl S)13S–24S. |
117. | Jeffery TK, Morrell NW. Molecular and cellular basis of pulmonary vascular remodeling in pulmonary hypertension. Prog Cardiovasc Dis 2002;45:173–202. |
118. | Laumanns IP, Fink L, Wilhelm J, Wolff JC, Mitnacht-Kraus R, Graef-Hoechst S, Stein MM, Bohle RM, Klepetko W, Hoda MA, et al. The noncanonical WNT pathway is operative in idiopathic pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2009;40:683–691. |
119. | 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 2007;293:L1–L8. |
120. | Rai PR, Cool CD, King JA, Stevens T, Burns N, Winn RA, Kasper M, Voelkel NF. The cancer paradigm of severe pulmonary arterial hypertension. Am J Respir Crit Care Med 2008;178:558–564. |
121. | de Jesus Perez VA, Alastalo TP, Wu JC, Axelrod JD, Cooke JP, Amieva M, Rabinovitch M. Bone morphogenetic protein 2 induces pulmonary angiogenesis via Wnt–beta-catenin and Wnt-RhoA-Rac1 pathways. J Cell Biol 2009;184:83–99. |
122. | Mason RJ. Biology of alveolar type II cells. Respirology 2006;11:S12–S15. |
123. | Matthay MA, Robriquet L, Fang X. Alveolar epithelium: role in lung fluid balance and acute lung injury. Proc Am Thorac Soc 2005;2:206–213. |
124. | Dobbs LG, Johnson MD. Alveolar epithelial transport in the adult lung. Respir Physiol Neurobiol 2007;159:283–300. |
125. | Lee JM, Dedhar S, Kalluri R, Thompson EW. The epithelial–mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol 2006;172:973–981. |
126. | Thiery JP. Epithelial–mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 2003;15:740–746. |
127. | Zavadil J, Bottinger EP. TGF-beta and epithelial-to-mesenchymal transitions. Oncogene 2005;24:5764–5774. |
128. | Huber MA, Kraut N, Beug H. Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol 2005;17:548–558. |
129. | Dasari V, Gallup M, Lemjabbar H, Maltseva I, McNamara N. Epithelial–mesenchymal transition in lung cancer: is tobacco the “smoking gun”? Am J Respir Cell Mol Biol 2006;35:3–9. |
130. | Barr S, Thomson S, Buck E, Russo S, Petti F, Sujka-Kwok I, Eyzaguirre A, Rosenfeld-Franklin M, Gibson NW, Miglarese M, et al. Bypassing cellular EGF receptor dependence through epithelial-to-mesenchymal-like transitions. Clin Exp Med 2008;25:685–693. |
131. | Kim K, Lu Z, Hay ED. Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int 2002;26:463–476. |
132. | Kato N, Shimmura S, Kawakita T, Miyashita H, Ogawa Y, Yoshida S, Higa K, Okano H, Tsubota K. Beta-catenin activation and epithelial–mesenchymal transition in the pathogenesis of pterygium. Invest Ophthalmol Vis Sci 2007;48:1511–1517. |
133. | Dissanayake SK, Wade MS, Johnson CE, O'Connell MP, Leotlela PD, French AD, Shah KV, Hewitt KJ, Rosenthal DT, Indig FE, et al. The WNT5a/PKC pathway mediates motility in melanoma cells via the inhibition of metastasis suppressors, and initiation of an epithelial to mesenchymal transition. J Biol Chem 2007;282:17259–17271. |
134. | Willis BC, Borok Z. TGF-beta–induced EMT: mechanisms and implications for fibrotic lung disease. Am J Physiol 2007;293:L525–L534. |
135. | Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci USA 2006;103:13180–13185. |
136. | Willis BC, Liebler JM, Luby-Phelps K, Nicholson AG, Crandall ED, du Bois RM, Borok Z. Induction of epithelial–mesenchymal transition in alveolar epithelial cells by transforming growth factor-beta1: potential role in idiopathic pulmonary fibrosis. Am J Pathol 2005;166:1321–1332. |
137. | Kasai H, Allen JT, Mason RM, Kamimura T, Zhang Z. TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir Res 2005;6:56. |
138. | Kim KK, Wei Y, Szekeres C, Kugler MC, Wolters PJ, Hill ML, Frank JA, Brumwell AN, Wheeler SE, Kreidberg JA, et al. Epithelial cell alpha3beta1 integrin links beta-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J Clin Invest 2009;119:213–224. |
139. | Benjamin JM, Nelson WJ. Bench to bedside and back again: molecular mechanisms of alpha-catenin function and roles in tumorigenesis. Semin Cancer Biol 2008;18:53–64. |
140. | Aberle H, Schwartz H, Kemler R. Cadherin-catenin complex: protein interactions and their implications for cadherin function. J Cell Biochem 1996;61:514–523. |
141. | Gottardi CJ, Gumbiner BM. Adhesion signaling: how beta-catenin interacts with its partners. Curr Biol 2001;11:R792–R794. |
142. | Nelson WJ, Nusse R. Convergence of WNT, beta-catenin, and cadherin pathways. Science 2004;303:1483–1487. |
143. | Harris TJ, Peifer M. Decisions, decisions: beta-catenin chooses between adhesion and transcription. Trends Cell Biol 2005;15:234–237. |
144. | Brembeck FH, Rosario M, Birchmeier W. Balancing cell adhesion and WNT signaling, the key role of beta-catenin. Curr Opin Genet Dev 2006;16:51–59. |
145. | Gottardi CJ, Gumbiner BM. Distinct molecular forms of beta-catenin are targeted to adhesive or transcriptional complexes. J Cell Biol 2004;167:339–349. |
146. | Brembeck FH, Schwarz-Romond T, Bakkers J, Wilhelm S, Hammerschmidt M, Birchmeier W. Essential role of bcl9–2 in the switch between beta-catenin's adhesive and transcriptional functions. Genes Dev 2004;18:2225–2230. |
147. | Perez-Moreno M, Fuchs E. Catenins: keeping cells from getting their signals crossed. Dev Cell 2006;11:601–612. |
148. | Ohira T, Gemmill RM, Ferguson K, Kusy S, Roche J, Brambilla E, Zeng C, Baron A, Bemis L, Erickson P, et al. WNT7a induces E-cadherin in lung cancer cells. Proc Natl Acad Sci USA 2003;100:10429–10434. |
149. | Thannickal VJ, Toews GB, White ES, Lynch JP III, Martinez FJ. Mechanisms of pulmonary fibrosis. Annu Rev Med 2004;55:395–417. |
150. | Chilosi M, Poletti V, Murer B, Lestani M, Cancellieri A, Montagna L, Piccoli P, Cangi G, Semenzato G, Doglioni C. Abnormal re-epithelialization and lung remodeling in idiopathic pulmonary fibrosis: the role of deltan-p63. Lab Invest 2002;82:1335–1345. |
151. | Kasper M, Haroske G. Alterations in the alveolar epithelium after injury leading to pulmonary fibrosis. Histol Histopathol 1996;11:463–483. |
152. | Qunn L, Takemura T, Ikushima S, Ando T, Yanagawa T, Akiyama O, Oritsu M, Tanaka N, Kuroki T. Hyperplastic epithelial foci in honeycomb lesions in idiopathic pulmonary fibrosis. Virchows Arch 2002;441:271–278. |
153. | Ma Y, Seneviratne CK, Koss M. Idiopathic pulmonary fibrosis and malignancy. Curr Opin Pulm Med 2001;7:278–282. |
154. | Sharma OP, Lamb C. Cancer in interstitial pulmonary fibrosis and sarcoidosis. Curr Opin Pulm Med 2003;9:398–401. |
155. | Samet JM. Does idiopathic pulmonary fibrosis increase lung cancer risk? Am J Respir Crit Care Med 2000;161:1–2. |
156. | Micke P, Ostman A. Exploring the tumour environment: cancer-associated fibroblasts as targets in cancer therapy. Expert Opin Ther Targets 2005;9:1217–1233. |
157. | Phan SH. Fibroblast phenotypes in pulmonary fibrosis. Am J Respir Cell Mol Biol 2003;29(3 Suppl)S87–S92. |
158. | Klapholz-Brown Z, Walmsley GG, Nusse YM, Nusse R, Brown PO. Transcriptional program induced by WNT protein in human fibroblasts suggests mechanisms for cell cooperativity in defining tissue microenvironments. PLoS One 2007;2:e945. |
159. | Boucherat O, Franco-Montoya ML, Thibault C, Incitti R, Chailley-Heu B, Delacourt C, Bourbon JR. Gene expression profiling in lung fibroblasts reveals new players in alveolarization. Physiol Genomics 2007;32:128–141. |
160. | Chen S, McLean S, Carter DE, Leask A. The gene expression profile induced by WNT 3a in NIH 3T3 fibroblasts. J Cell Commun Signal 2007;1:175–183. |
161. | Reya T, Clevers H. WNT signalling in stem cells and cancer. Nature 2005;434:843–850. |
162. | Kleber M, Sommer L. WNT signaling and the regulation of stem cell function. Curr Opin Cell Biol 2004;16:681–687. |
163. | Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR. Basal cells are a multipotent progenitor capable of renewing the bronchial epithelium. Am J Pathol 2004;164:577–588. |
164. | Giangreco A, Reynolds SD, Stripp BR. Terminal bronchioles harbor a unique airway stem cell population that localizes to the bronchoalveolar duct junction. Am J Pathol 2002;161:173–182. |
165. | Stripp BR, Shapiro SD. Stem cells in lung disease, repair, and the potential for therapeutic interventions: State-of-the-art and future challenges. Am J Respir Cell Mol Biol 2006;34:517–518. |
166. | Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S, Crowley D, Bronson RT, Jacks T. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 2005;121:823–835. |
167. | Stripp BR, Reynolds SD. Maintenance and repair of the bronchiolar epithelium. Proc Am Thorac Soc 2008;5:328–333. |
168. | Zhang Y, Goss AM, Cohen ED, Kadzik R, Lepore JJ, Muthukumaraswamy K, Yang J, DeMayo FJ, Whitsett JA, Parmacek MS, et al. A GATA6-WNT pathway required for epithelial stem cell development and airway regeneration. Nat Genet 2008;40:862–870. |
169. | Reynolds SD, Zemke AC, Giangreco A, Brockway BL, Teisanu RM, Drake JA, Mariani T, Di PY, Taketo MM, Stripp BR. Conditional stabilization of beta-catenin expands the pool of lung stem cells. Stem Cells 2008;26:1337–1346. |
170. | Borok Z, Li C, Liebler J, Aghamohammadi N, Londhe VA, Minoo P. Developmental pathways and specification of intrapulmonary stem cells. Pediatr Res 2006;59:84R–93R. |
171. | Zemke AC, Teisanu RM, Giangreco A, Drake JA, Brockway BL, Reynolds SD, Stripp BR. β-catenin is not necessary for maintenance or repair of the bronchiolar epithelium. Am J Respir Cell Mol Biol (In press). |
172. | Warburton D, Perin L, Defilippo R, Bellusci S, Shi W, Driscoll B. Stem/progenitor cells in lung development, injury repair, and regeneration. Proc Am Thorac Soc 2008;5:703–706. |
173. | Loebinger MR, Sage EK, Janes SM. Mesenchymal stem cells as vectors for lung disease. Proc Am Thorac Soc 2008;5:711–716. |
174. | Etheridge SL, Spencer GJ, Heath DJ, Genever PG. Expression profiling and functional analysis of WNT signaling mechanisms in mesenchymal stem cells. Stem Cells 2004;22:849–860. |
175. | Mohr A, Lyons M, Deedigan L, Harte T, Shaw G, Howard L, Barry F, O'Brien T, Zwacka R. Mesenchymal stem cells expressing TRAIL lead to tumour growth inhibition in an experimental lung cancer model. J Cell Mol Med 2008;12:2628–2643. |
176. | Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, Richardson AL, Polyak K, Tubo R, Weinberg RA. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007;449:557–563. |
177. | Rojas M, Xu J, Woods CR, Mora AL, Spears W, Roman J, Brigham KL. Bone marrow–derived mesenchymal stem cells in repair of the injured lung. Am J Respir Cell Mol Biol 2005;33:145–152. |
178. | Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, Phinney DG. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci USA 2003;100:8407–8411. |
179. | Hashimoto N, Jin H, Liu T, Chensue SW, Phan SH. Bone marrow–derived progenitor cells in pulmonary fibrosis. J Clin Invest 2004;113:243–252. |
180. | 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. |
181. | Mehrad B, Burdick MD, Zisman DA, Keane MP, Belperio JA, Strieter RM. Circulating peripheral blood fibrocytes in human fibrotic interstitial lung disease. Biochem Biophys Res Commun 2007;353:104–108. |
182. | Lama VN, Smith L, Badri L, Flint A, Andrei AC, Murray S, Wang Z, Liao H, Toews GB, Krebsbach PH, et al. Evidence for tissue-resident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J Clin Invest 2007;117:989–996. |