Next Article in Journal
The Effect of Agomelatine Treatment on Diabetes-Induced Cognitive Impairments in Rats: Concomitant Alterations in the Hippocampal Neuron Numbers
Next Article in Special Issue
Roles of the TGF-β–VEGF-C Pathway in Fibrosis-Related Lymphangiogenesis
Previous Article in Journal
Maternal Resveratrol Therapy Protects Male Rat Offspring against Programmed Hypertension Induced by TCDD and Dexamethasone Exposures: Is It Relevant to Aryl Hydrocarbon Receptor?
Previous Article in Special Issue
Effects of Er:YAG and Diode Laser Irradiation on Dental Pulp Cells and Tissues
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TGF-β Signaling in Lung Health and Disease

1
Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
2
Division for Health Service Promotion, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
3
Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(8), 2460; https://doi.org/10.3390/ijms19082460
Submission received: 10 July 2018 / Revised: 18 August 2018 / Accepted: 18 August 2018 / Published: 20 August 2018
(This article belongs to the Special Issue TGF-Beta Super Family Signaling)

Abstract

:
Transforming growth factor (TGF)-β is an evolutionarily conserved pleiotropic factor that regulates a myriad of biological processes including development, tissue regeneration, immune responses, and tumorigenesis. TGF-β is necessary for lung organogenesis and homeostasis as evidenced by genetically engineered mouse models. TGF-β is crucial for epithelial-mesenchymal interactions during lung branching morphogenesis and alveolarization. Expression and activation of the three TGF-β ligand isoforms in the lungs are temporally and spatially regulated by multiple mechanisms. The lungs are structurally exposed to extrinsic stimuli and pathogens, and are susceptible to inflammation, allergic reactions, and carcinogenesis. Upregulation of TGF-β ligands is observed in major pulmonary diseases, including pulmonary fibrosis, emphysema, bronchial asthma, and lung cancer. TGF-β regulates multiple cellular processes such as growth suppression of epithelial cells, alveolar epithelial cell differentiation, fibroblast activation, and extracellular matrix organization. These effects are closely associated with tissue remodeling in pulmonary fibrosis and emphysema. TGF-β is also central to T cell homeostasis and is deeply involved in asthmatic airway inflammation. TGF-β is the most potent inducer of epithelial-mesenchymal transition in non-small cell lung cancer cells and is pivotal to the development of tumor-promoting microenvironment in the lung cancer tissue. This review summarizes and integrates the current knowledge of TGF-β signaling relevant to lung health and disease.

Graphical Abstract

1. Introduction

The transforming growth factor (TGF)-β superfamily consists of three isoforms of TGF-β, Activin, Nodal, bone morphogenetic proteins (BMPs), growth and differentiation factors (GDFs), and others. In mammals, 33 proteins are known to be members of the TGF-β superfamily [1]. TGF-β is the prototypic and most-studied factor, and exhibits versatile functions in almost all cell types. Molecular cloning of human TGF-β was reported in 1985 [2], and its receptors were subsequently identified [3]. Numerous studies over the past decades have reported that TGF-β regulates a wide range of biological processes including cell proliferation, differentiation, apoptosis, extracellular matrix (ECM) synthesis, and stem/progenitor cell fates, thereby affecting embryogenesis, morphogenesis, wound healing, and immune responses of multiple organs. TGF-β family members diversified and adapted to regulate biological systems that evolved over time. TGF-β family members are evolutionarily conserved from arthropods, although TGF-β ligands originated at a later stage of evolution.
Development of the respiratory system is an essential event for vertebrate land adaptation. The fetal lungs are first inflated at birth; thereafter, repeated cycles of respiration continue throughout the entire lifespan. The lungs are structurally exposed to extrinsic stimuli and pathogens, and are susceptible to inflammation, allergic reactions, and carcinogenesis. TGF-β is necessary for lung organogenesis and homeostasis, and is involved in many respiratory diseases, including pulmonary fibrosis, emphysema, bronchial asthma, and lung cancer [4]. In this review, we focus on TGF-β signaling and integrate the current literature to address its role in lung health and disease.

2. TGF-β Activation in the Extracellular Milieu

Secretion of TGF-β alone is not sufficient for its bioavailability. TGF-β needs to be activated by several mechanisms, including proteolysis, low pH, reactive oxygen species (ROS), and thrombospondin-1 [5,6]. Specific integrins such as integrin αVβ6 also activate TGF-β by sensing traction forces or stiffness of the ECM [7].
TGF-β peptides are synthesized as latent precursors and are cleaved to form a mature TGF-β dimer noncovalently associated with latency-associated peptide (LAP). LAP of TGF-β1 or TGF-β3 has an integrin recognition motif (RGD sequence) and binds to integrins. The secreted complex of TGF-β and LAP is bound by latent TGF-β binding protein (LTBP), which forms a large latent complex (LLC) [8]. LTBP is a matrix protein incorporated in the ECM, and latent TGF-β (TGF-β-LAP complex) can be stored in the extracellular milieu.
Animal studies have demonstrated functional importance of LTBP isoforms in lung morphogenesis. LTBP-3-null mice display defective lung alveolarization [9]. LTBP-4 is highly expressed in the murine developing lung [10], and is necessary for elastic fiber assembly on microfibrils [11]. LTBP-4-deficient mice show severe pulmonary emphysema [12] due to impaired elastogenesis and structural defects of lung alveoli. These phenotypes are presumably associated with dysregulated TGF-β signaling.
Upon activation, TGF-β is released from the LAP and rapidly mobilized to exert its activity through binding to cell surface receptors. Temporospatial distribution of latent TGF-β and its receptors in the tissue and its dynamic activation contribute to the diversity of TGF-β-mediated biological processes. Intriguingly, a mechanical stress-mediated mechanism of TGF-β activation has been reported [13], and integrins bound to the TGF-β-LAP complex mediate cell contractile forces that lead to TGF-β activation. LTBP incorporated in the ECM is also bound to the LAP that provides mechanical resistance. Such pulling forces result in conformational changes of latent TGF-β and concomitant TGF-β release. Latent TGF-β therefore serves as a sensor for mechanical stress, and released TGF-β acts as an effector. TGF-β itself further regulates expression of integrins and ECM proteins [14,15], which in turn participate in the fine-tuning of TGF-β activation by binding to LAP and LTBP. Because the lung is a highly mechanical organ with a unique alveolar structure, mechanical stress-mediated regulation of TGF-β activity and ECM organization are likely to be important parameters for understanding pulmonary physiology and pathology.

3. Context-Dependency of TGF-β Signaling

Activated TGF-β in a dimeric form binds to type I (TGFβR-I, also known as ALK-5) and type II (TGFβR-II) receptors. TGFβR-I is phosphorylated by TGFβR-II upon ligand binding, and in the canonical Smad-dependent pathway, TGFβR-I phosphorylates the intracellular signal transducers, Smad2 and Smad3. Phosphorylated Smad2 and Smad3 interact with Smad4, and translocate into the nucleus [16]. Smad complexes regulate target gene transcription in association with various transcription factors, transcriptional coactivators, and corepressors. In addition to this canonical pathway, TGF-β also activates mitogen-associated protein kinase cascades and RhoA GTPase [17].
TGF-β acts on a wide variety of cell types and induces various cellular responses. For example, TGF-β is involved in stem cell self-renewal and also controls differentiation of multiple cell lineages [18]. Context-dependent responses are regulated by multiple aspects of the TGF-β signaling cascade, including crosstalk with other signals and transcriptional regulation [19]. The three TGF-β ligand isoforms show differential expression patterns and biological effects. Intracellular Smad7 is known to inhibit TGF-β signaling through various mechanisms, and is important as a negative feedback regulator [20,21]. TGF-β receptors and Smad molecules are post-translationally controlled by ubiquitination-mediated degradation [22], and their activity and signaling magnitude are also regulated by protein modifications [23,24].
Cell type specification is regulated by a few master transcription factors, and forced overexpression of such master genes is sufficient for inducing many cell lineages [25]. Recent genome-wide chromatin occupancy and gene expression analyses have revealed that cell type-specific effects of TGF-β are achieved by cooperation between Smad complexes and master transcription factors. For example, induction of Myod1, a master transcription factor for myogenesis, directs Smad3-mediated transactivation of muscle-specific genes [18].
NKX2-1 (also known as thyroid transcription factor-1) is a homeodomain transcription factor expressed in pulmonary epithelial cells. NKX2-1 is a master regulator of lung morphogenesis and is indispensable for tracheoesophageal separation, branching morphogenesis, and alveolar maturation [26]. NKX2-1 is essential for transactivation of lung-specific genes, such as the alveolar epithelial cell-specific surfactant genes (SPA, SPB, and SPC) as well as the club cell-specific gene, CC10 [27]. Convergence of NKX2-1 and TGF-β signaling has been shown to be important for transcriptional regulations in lung epithelial cells. TGF-β represses transcription of SPB, and Smad3 interacts with NKX2-1 and decreases its activity [28]. Moreover, genome-wide chromatin occupancy analysis using lung adenocarcinoma cells has revealed that NKX2-1 colocalizes with Smad3 to regulate a subset of genes while competing with Smad3-Smad4 complex formation resulting in altered genomic binding profiles of Smad3 [29]. Both pro-tumorigenic and tumor-suppressive roles of NKX2-1 have been reported in lung adenocarcinoma [30]. NKX2-1 and TGF-β signaling therefore appear to be a key component for regulating lung adenocarcinoma cell features.

4. TGF-β Signaling in Lung Organogenesis

Lung development takes place through coordinated growth and differentiation of the endoderm-derived epithelium and the mesoderm-derived mesenchyme. During the embryonic stage, lung buds arise from the anterior foregut, and branching morphogenesis follows during the pseudoglandular stage [31].
Lung morphogenesis is controlled by multiple signals such as fibroblast growth factor, sonic hedgehog (SHH), Wnt/β-catenin, and BMP. In cooperation with these signals, TGF-β plays key roles in epithelial-mesenchymal interactions. During the pseudoglandular stage, the three isoforms of TGF-β show different expression patterns [32]. TGF-β1 is expressed throughout the mesenchyme and is highly localized in the area underlying the epithelial branching point. TGF-β2 is localized in the distal epithelium, and TGF-β3 is detected in the proximal mesenchyme and distal epithelium.
Different pulmonary phenotypes have been reported in the knockout mice of TGF-β isoforms, indicating distinct functions for the different isoforms (Table 1). TGF-β1-deficient mice show systemic inflammation, and in the lungs, generalized perivasculitis or interstitial pneumonia is observed [33,34]. TGF-β2-null mice have postnatal lung defects with collapsed distal airways [35]. TGF-β3 deficiency leads to defective lungs with alveolar hypoplasia and mesenchymal thickening [36].
Conditional abrogation of TGFβR-II and TGF-β signaling inhibition in SPC-expressing lung epithelial cells result in retarded postnatal alveologenesis, but without an apparent prenatal phenotype [37]. Conditional TGFβR-II knockout mice generated using Nkx2-1-Cre display abnormal alveolarization and emphysema [38]. Smad3-deficient mice show impaired alveolarization and centrilobular emphysema [39,40], similar to the effect of TGFβR-II abrogation in lung epithelial cells. Notably, deletion of TGFβR-I in epithelial cells using Gata5-Cre leads to immature alveoli and a disorganized epithelium with reduced club cell population [41]. All of these indicate that TGF-β signaling is necessary for lung epithelial cell differentiation and maturation.
Mesenchymal abrogation of TGFβR-II disrupts lung branching morphogenesis, resulting in cystic malformation of the bronchi. This phenotype was shown to be associated with dysregulated SHH signaling in the mesenchyme [42]. Mesodermal inactivation of TGFβR-I results in pulmonary hypoplasia due to impaired differentiation of mesodermal progenitor cells [43].
Ectopic expression of TGF-β1 in the lung epithelium disrupts lung morphogenesis and perturbs epithelial differentiation [44]. Moreover, exogenous TGF-β exerts an inhibitory effect on lung branching morphogenesis as demonstrated in explant cultures [45].
Taken together, TGF-β signaling appears to play distinct and critical roles in the lung epithelium and mesenchyme, and is required for epithelial-mesenchymal interactions to achieve lung branching morphogenesis and alveologenesis (Figure 1).

5. TGF-β Signaling in Lung Alveolar Epithelial Growth and Differentiation

Differentiated airway epithelial cells include basal, secretory, ciliated, and neuroendocrine cells, and the alveoli are lined by alveolar epithelial type I and type II cells (Figure 1). Alveolar epithelial type I cells cover the majority of the alveolar surface, allowing for gas exchange, while type II cells are involved in pulmonary surfactant production [46]. As previously mentioned, analyses of genetically engineered mouse models have revealed crucial roles for TGF-β signaling in lung epithelial growth and differentiation (Table 1).
It is widely accepted that TGF-β shows cytostatic effects in most epithelial cells, and TGF-β has been shown to inhibit proliferation of alveolar epithelial type II cells [47]. TGF-β is also known as the most powerful inducer of epithelial-mesenchymal transition (EMT) [48]. EMT is a biological process where polarized epithelial cells acquire mesenchymal phenotypes with enhanced cell motility. Mechanistically, TGF-β induces transcriptional repressors, SNAI1, SNAI2, ZEB1, and ZEB2, which subsequently repress adherens junction and tight junction proteins such as E-cadherin and ZO-1, thereby disrupting epithelial cell junction and apical-basal polarity [49] (Figure 2A). ZEB1 and ZEB2 also repress the miRNA-200 family and miR-205, which target ZEB1 and ZEB2, and thereby derepress the EMT regulatory axis of TGF-β and ZEB proteins [50]. TGF-β induces EMT in rat alveolar epithelial cells, which may contribute to disease conditions by endowing epithelial cells with migratory and anti-apoptotic phenotypes [51,52].
A series of pioneering studies established primary cultures of alveolar epithelial type II cells that form polarized monolayers and spontaneously differentiate into type I cells [53]. TGF-β inhibits expression of alveolar epithelial type II cell markers and enhances differentiation of cultured type II cells into type I cells [54] (Figure 2B). Thus, the effects of TGF-β in alveolar epithelial type II cells include EMT induction and type I cell differentiation, which may depend on cell culture conditions.
It is thought that alveolar epithelial type II cells contribute to alveolar homeostasis by self-renewing and differentiating into type I cells [55]. Conversely, a subset of alveolar epithelial type I cells generate type II cells, illustrating the plasticity of alveolar epithelial cells [56]. Importantly, conversion of type I to type II cells is facilitated by TGF-β signaling inhibition in organoid culture [56].
Recently, cell culture methods to induce alveolar epithelial cells from pluripotent stem cells have been developed [57,58]. Inhibition of TGF-β signaling has been shown to augment in vitro differentiation of anterior foregut endoderm cells that give rise to NKX2-1-positive alveolar epithelial progenitors [59].
Taken together, TGF-β plays multifaceted roles in alveolar epithelial cells, including cell growth suppression, EMT, and regulating reciprocal differentiation between type I and type II cells.

6. TGF-β Signaling in Pulmonary Diseases

6.1. TGF-β Signaling in Pulmonary Fibrosis and Emphysema

Pulmonary fibrosis is a chronic and progressive lung disease, in which repeated wound and repair processes lead to irreversible structural alterations and tissue stiffening [60]. Pathophysiological steps include alveolar epithelial damage by extrinsic irritants, fibroblast activation, and persistent fibrotic reaction. Differentiation of lung fibroblasts into myofibroblasts is a key step in the development of tissue fibrosis. Myofibroblasts express α-smooth muscle actin (α-SMA) as a marker of activated fibroblasts, and are capable of ECM production including collagen, laminin, and fibronectin. TGF-β is the most potent factor for the induction of myofibroblast differentiation (Figure 2C), and increased expression of TGF-β has been reported in fibrotic lungs. The major cellular sources of TGF-β in pulmonary fibrosis have been shown to be alveolar macrophages and metaplastic type II alveolar epithelial cells [61,62]. In addition to ECM components, TGF-β induces integrins, matrix metalloproteinases, protease inhibitors, and regulators of small GTPases [14,15]. These molecules participate in tissue remodeling and influence cell-ECM interactions. In addition, TGF-β is thought to promote lung fibrosis by suppressing production of anti-fibrotic molecules such as hepatocyte growth factor and prostaglandin E2 [63,64]. Furthermore, TGF-β inhibits alveolar epithelial cell growth and repair. Thus, TGF-β is a key player in fibrotic processes, acting on both fibroblasts and alveolar epithelial cells.
Smoking-induced emphysema is the main cause of chronic obstructive pulmonary disease (COPD), and is characterized by lung hyperinflation and enlargement of air spaces distal to the terminal bronchioles. Recently, an association between TGFB2 polymorphisms and COPD has been reported [65]. TGF-β1 expression has been shown in small airway epithelial cells among smokers and patients with COPD [66], suggesting that pathologically activated TGF-β signaling is involved in the pathogenesis of emphysema.
In murine models, fibrosis and emphysema are often discussed as contrasting disorders. As previously mentioned, conditional deletion of TGFβR-II in lung epithelial cells results in emphysema-like phenotypes whereas these mice are protected from bleomycin-induced pulmonary fibrosis [37,38]. Smad3-deficient mice show progressive alveolar destruction resembling emphysema while they are protected from bleomycin-induced lung fibrosis [67]. Itgb6-null mice develop age-related emphysema [68], similar to the effects of Smad3 deficiency [39,40] or TGFβR-II abrogation in lung epithelial cells [37,38]. Supporting the notion that the phenotype of Itgb6 deletion is caused by disruption of integrin αvβ6-mediated TGF-β activation, ectopic expression of active TGF-β1 overcomes the effect of Itgb6 deletion [68]. On the other hand, Itgb6-null mice are resistant to bleomycin-induced lung injury and fibrosis [69]. Another study consistently showed that αvβ6-blocking antibody attenuated bleomycin-induced pulmonary fibrosis [70]. These models suggest that lack of TGF-β signaling predisposes the lungs to develop emphysema while conferring resistance to fibrosis.
In contrast to this simplified dichotomy in mouse models, the coexistence of fibrosis and emphysema in the same patient is common. Of clinical importance, pulmonary fibrosis and COPD are independent risk factors for lung cancer (Figure 3). Notably, combined pulmonary fibrosis and emphysema (CPFE) is increasingly recognized as an entity with severely impaired gas exchange and higher frequencies of pulmonary hypertension and lung cancer [71] (Figure 3). Pathologically activated TGF-β signaling in relation to smoking and ageing are involved in emphysema and fibrosis, and common molecular mechanisms have therefore been postulated in both diseases. Because dynamic organization and turnover of the ECM in the lung tissue is largely regulated by TGF-β, these processes might represent different forms of tissue remodeling as a consequence of pathologically activated TGF-β signaling. However, it remains largely unknown how TGF-β signaling differentially contributes to these two contrasting phenotypes.

6.2. Dual Roles of TGF-β in Asthma Involving Immune Response and Airway Remodeling

Bronchial asthma is a global health problem that affects more than 300 million people worldwide [72]. Asthma is characterized by chronic airway inflammation and hyperresponsiveness mediated by T helper type 2 (Th2) cells and the related cytokines, interleukin (IL)-4, IL-5, IL-9, and IL-13. These cytokines cause chronic inflammation, pulmonary eosinophilia, mucus cell hyperplasia, smooth muscle contraction, and airway remodeling. In addition to Th2 cells, Th17 cells that secrete IL-17A and IL-17F also participate in the development of allergic airway inflammation [72]. Inhaled corticosteroids have greatly improved the management of asthma in recent decades. However, some asthmatic patients are refractory to this therapy because of irreversible narrowing of the airway [73]. Thickening of the airway smooth muscle layer, a hallmark of asthmatic airway remodeling, is related to the clinical severity of asthma [74]. Recently, increasing attention has been focused on asthma and COPD overlap as a clinical entity (Figure 3).
The importance of TGF-β signaling in the pathogenesis of asthma has been illustrated by genome-wide association studies. It has been reported that SMAD3 polymorphism is associated with asthma [75]. A meta-analysis further suggested that C509T or T869C polymorphism of TGFB1 gene may predispose to asthma [76]. Moreover, a recent study reported that patients with Loeys-Dietz syndrome who harbor loss-of-function mutations in TGFβR-I or TGFβR-II frequently develop allergic diseases including asthma [77].
TGF-β1 concentration in bronchoalveolar lavage fluid is elevated in atopic asthma [78], and TGF-β expression is increased in bronchial specimens of asthmatic patients [79]. Bronchial epithelial cells and eosinophils are the major source of TGF-β in the asthmatic airway [80]. TGF-β enhances airway smooth muscle proliferation and ECM deposition by activated fibroblasts, eventually leading to structural alterations of the airway. Moreover, it has been reported that TGF-β1 modulates airway smooth muscle shortening and hyperresponsiveness by enhancing excitation-contraction coupling [81].
The pathological role of TGF-β in asthma is not restricted to airway remodeling, and its effect on the immune response is thought to be more important than previously recognized. TGF-β induces Foxp3 gene expression in CD4+ CD25- naïve T cells in the presence of T cell receptor stimulation, which in turn mediates differentiation of regulatory T (Treg) cells with immunosuppressive functions [82] (Figure 2D). Notably, TGF-β is further produced by Treg cells. TGF-β1-null mice consistently fail to maintain peripheral Treg cells, resembling the phenotype of Foxp3-null mice [83]. Of therapeutic importance, it has been reported that adoptive transfer of Treg cells ameliorates allergic airway inflammation in a murine model [84]. T cell-specific deletion of TGFβR-II results in a reduction of peripheral Treg cells, and also inhibits thymic CD8+ T cell maturation and natural killer T (NKT) cell development in the thymus [85]. CD4+ T helper cells differentiate into Th1, Th2, and Th17 cells characterized by the signature cytokines, IFN-γ, IL-4, and IL-17, respectively [86]. TGF-β is known to induce Th17 cells in combination with IL-6, which contribute to neutrophilic recruitment into the airway and Th2 cell-mediated eosinophilic inflammation [87,88]. In contrast, TGF-β has inhibitory effects on Th1 and Th2 cell differentiation through suppression of T-bet and GATA3 transcription factors, respectively [89] (Figure 2D). Thus, TGF-β plays multimodal roles in Th2 type immune response. TGF-β suppresses Th2 cells directly or through induction of Treg cells. On the other hand, TGF-β also enhances Th2 cell-mediated action by inducing Th17 cells.
Collectively, TGF-β positively regulates Treg, Th17, NKT, and CD8+ T cells while inhibiting Th1 and Th2 differentiation. Thus, TGF-β plays multifunctional roles in T cell differentiation and homeostasis, thereby affecting the immune system in the asthmatic airway.

6.3. TGF-β Mediates EMT in Non-Small Cell Lung Cancer (NSCLC)

Lung cancer is the leading cause of cancer-related mortality worldwide [90]. NSCLC comprises the majority of lung cancers, which include the histological subtypes of adenocarcinoma and squamous cell carcinoma. Higher TGF-β expression levels are associated with lymph node metastasis and tumor angiogenesis in NSCLC [91], and tumor cells established from NSCLC express TGF-β ligands.
It is widely believed that TGF-β plays dual roles during tumor progression [92]. TGF-β suppresses epithelial cell proliferation and acts as a tumor suppressor in the early stage of tumorigenesis, and loss-of-function mutations in TGF-β signaling components have been identified in several cancer types. Inactivating mutations of TGFβR-II are frequent in colorectal carcinomas with microsatellite instability [93], and loss of chromosomal region 18q21, encoding SMAD4 gene, is found in pancreatic and colorectal cancers [94]. Mutations of TGFβR-II and SMAD4 are rare in NSCLC [95], whereas epidermal growth factor receptor (EGFR) and KRAS mutations are frequently found in lung adenocarcinomas [96].
In addition to its cytostatic effects, TGF-β potently induces EMT in cancer cells, which represents a tumor-promoting aspect of TGF-β activity in the later stages of cancer progression. Cultured cancer cells usually lack cell polarity or epithelial integrity, and remain in an intermediary state of partial or incomplete EMT [97]. NSCLC cell lines could therefore be classified into epithelial-like and mesenchymal-like subgroups based on gene expression profiling [98,99]. Recent studies using NSCLC cell lines have focused on the TGF-β-mediated completion of EMT programs.
TGF-β-mediated EMT is modified by crosstalk with other signaling pathways. In A549 lung adenocarcinoma cells, TNF-α enhances EMT and induces cancer cells with a secretory phenotype capable of producing various cytokines and chemokines [14]. Of particular importance is the convergence between oncogenic KRAS and TGF-β signaling in EMT processes [100]. Transduction of oncogenic KRAS dramatically enhances TGF-β-elicited SNAI1 expression, and TGF-β induces SNAI1 more strongly in cancer cells harboring constitutively active KRAS mutations.
TGF-β-mediated EMT in cancer cells is associated with aggressive features such as resistance to apoptosis, acquisition of stem cell traits, and chemoresistance [101]. On the other hand, recent studies have suggested that EMT is likely dispensable for metastasis [102].

6.4. TGF-β Orchestrates the Tumor Microenvironment

Besides its direct effect on cancer cells, TGF-β facilitates invasion and metastatic spread through reciprocal interactions between cancer cells and the tumor stromal microenvironment [103]. TGF-β orchestrates the development of tumor stroma and promotes angiogenesis, immune evasion, and remodeling of the ECM [104,105]. The stromal reaction presumably mediated by TGF-β is associated with poor prognosis in resected lung adenocarcinomas [106]. Tumor stroma comprises cancer-associated fibroblasts (CAFs), immune cells, blood vessels, and the ECM [107]. CAFs are activated by TGF-β and promote tumor progression by secretion of soluble factors and ECM remodeling [108,109]. The gene signatures related to TGF-β signaling are enriched in CAFs isolated from NSCLC tissues compared to their normal counterparts [110]. Tumor stroma shows dysregulated ECM organization and tissue stiffness, which are similar to the features of pulmonary fibrosis. Of clinical importance, cancer tissue fibrosis is associated with increased interstitial fluid pressure, which impedes delivery of cancer therapeutics [111].
Tumor angiogenesis is crucial for the delivery of oxygen and nutrients to cancer cells. TGF-β is known to stimulate angiogenic factors such as vascular endothelial growth factor and connective tissue growth factor produced by cancer cells or stromal fibroblasts. On the other hand, it has been suggested that TGF-β negatively regulates lymphangiogenesis [112].
Cancer progression is thought to be dependent on immunosurveillance evasion, and this notion is supported by the recent success of immune checkpoint inhibitors as therapeutic options for NSCLC. As previously mentioned, TGF-β participates in the maintenance of T cell homeostasis and induces Treg cells that limit tumor immune responses [82]. Moreover, TGF-β in the cancer tissue polarizes tumor-associated macrophages, myeloid-derived suppressor cells, and tumor-associated neutrophils to tumor-promoting phenotypes [113,114]. It has also been reported that TGF-β signaling blockade by TGF-β blocking antibody or TGFβR-I kinase inhibitor enhances anti-tumor immunity and shows therapeutic benefits [115,116].
Taken together, TGF-β facilitates cancer progression through diverse host-tumor interactions, including fibroblast activation, ECM remodeling, angiogenesis, and immune evasion.

6.5. TGF-β Signaling in Small Cell Lung Cancer (SCLC)

SCLC is a highly aggressive lung cancer subtype that accounts for 10–15% of lung cancers [117]. SCLC originates from lung neuroendocrine cell precursors, and achaete-scute complex homolog 1 (ASCL1) is a master transcription factor for neuroendocrine differentiation [118]. SCLC cell lines and tissue samples contain heterogeneous gene expression patterns. A variant subtype of SCLC with poor neuroendocrine differentiation and low ASCL1 expression has been recently reported [119,120].
Most classic SCLC cell lines lack TGFβR-II expression and are unresponsive to TGF-β [121], whereas variant SCLC cell lines tend to express relatively high levels of TGFβR-II. A recent report described molecular mechanisms responsible for this SCLC heterogeneity [122]. In lung epithelial/cancer cells with TGFβR-II expression, endogenous TGF-β suppresses ASCL1 expression. However, TGF-β signaling is inactivated in classic SCLC cells and ASCL1 expression is derepressed. Because TGF-β signaling inactivation and ASCL1 induction can promote SCLC survival, loss of TGF-β responsiveness is probably an important aspect of SCLC pathogenesis. Detailed analyses of genomic profiles in heterogeneous SCLC tumor samples may be helpful to dissect the exact role of TGF-β signaling in SCLC and its relation to neuroendocrine differentiation.

7. Gene Regulatory Networks Responsible for the TGF-β-Induced Cellular Response

The genome, epigenome, and transcriptome in a broad range of cell types and disease conditions have been characterized using recent advances in next-generation sequencing technology. Histone modification, DNA methylation, and gene expression profiling data across the whole genome have been included in public databases [123]. Genome-wide chromatin-binding profiling data have further provided mechanistic insights into transcriptional regulations [124]. It is becoming clear that a subset of transcription factors control cell lineage determination through activation of cell type-specific super-enhancers, which are enriched with active histone marks and defined as large genomic regions occupied by high densities of master transcription factors [125,126]. In parallel with these advances in genome biology, context-dependent transcriptional regulations by TGF-β-Smad pathway is being elucidated in more detail.
It has been reported that T-box family genes such as TBX4 are associated with super-enhancers and are involved in defining cellular identity of lung fibroblasts. TGF-β potently suppresses T-box family genes while inducing myofibroblast differentiation, and super-enhancers unique to lung fibroblasts are globally suppressed in lung CAFs. Thus, it has been proposed that TGF-β induces redistribution of super-enhancers at the expense of cell type-specific basal super-enhancers [127].
In the process of TGF-β-mediated EMT in A549 lung adenocarcinoma cells, a few transcription factors associated with super-enhancers (HNF4A, JUNB, and ETS2) have been shown to constitute a gene regulatory network that synergistically induces EMT [128]. As such, TGF-β-mediated cell differentiation or phenotypic conversion is coupled with dynamic reorganization of core transcriptional networks that involve the Smad complex, master transcription factors, and super-enhancers.

8. Future Perspectives

TGF-β is essential for lung organogenesis, homeostasis, and pathological conditions. Because TGF-β is involved in the pathogenesis of pulmonary fibrosis and NSCLC, there have been clinical trials testing the efficacy of inhibitors, antisense oligonucleotides, and neutralizing antibodies targeting TGF-β [129]. Moreover, TGF-β inhibition is emerging as a promising strategy for augmenting cancer immunotherapy [115,116].
Recently, single cell transcriptome analysis has become a powerful tool for investigating heterogeneous cell populations and differentiation processes in various conditions [130,131]. In-depth analyses using single cell-based technologies could further elucidate molecular mechanisms underlying TGF-β-mediated cell type-specific responses in physiological and pathological processes.

Funding

This work was supported by the Takeda Science Foundation (A.S.), the Uehara Memorial Foundation Research Fellowship (M.H.), and JSPS KAKENHI #18K08170 (A.S.) and #16H02653 (T.N.).

Acknowledgments

We express our sincere appreciation to Zea Borok for her constructive suggestions and critical reading of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Morikawa, M.; Derynck, R.; Miyazono, K. TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology. Cold Spring Harb. Perspect. Biol. 2016, 8, a021873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Derynck, R.; Jarrett, J.A.; Chen, E.Y.; Eaton, D.H.; Bell, J.R.; Assoian, R.K.; Roberts, A.B.; Sporn, M.B.; Goeddel, D.V. Human transforming growth factor-beta complementary DNA sequence and expression in normal and transformed cells. Nature 1985, 316, 701–705. [Google Scholar] [CrossRef] [PubMed]
  3. Franzén, P.; ten Dijke, P.; Ichijo, H.; Yamashita, H.; Schulz, P.; Heldin, C.H.; Miyazono, K. Cloning of a TGF beta type I receptor that forms a heteromeric complex with the TGF beta type II receptor. Cell 1993, 75, 681–692. [Google Scholar] [CrossRef]
  4. Aschner, Y.; Downey, G.P. Transforming Growth Factor-β: Master Regulator of the Respiratory System in Health and Disease. Am. J. Respir. Cell Mol. Biol. 2016, 54, 647–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Crawford, S.E.; Stellmach, V.; Murphy-Ullrich, J.E.; Ribeiro, S.M.; Lawler, J.; Hynes, R.O.; Boivin, G.P.; Bouck, N. Thrombospondin-1 is a major activator of TGF-beta1 in vivo. Cell 1998, 93, 1159–1170. [Google Scholar] [CrossRef]
  6. Miyazono, K.; Heldin, C.H. Role for carbohydrate structures in TGF-beta 1 latency. Nature 1989, 338, 158–160. [Google Scholar] [CrossRef] [PubMed]
  7. Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-β structure and activation. Nature 2011, 474, 343–349. [Google Scholar] [CrossRef] [PubMed]
  8. Kanzaki, T.; Olofsson, A.; Morén, A.; Wernstedt, C.; Hellman, U.; Miyazono, K.; Claesson-Welsh, L.; Heldin, C.H. TGF-beta 1 binding protein: A component of the large latent complex of TGF-beta 1 with multiple repeat sequences. Cell 1990, 61, 1051–1061. [Google Scholar] [CrossRef]
  9. Colarossi, C.; Chen, Y.; Obata, H.; Jurukovski, V.; Fontana, L.; Dabovic, B.; Rifkin, D.B. Lung alveolar septation defects in Ltbp-3-null mice. Am. J. Pathol. 2005, 167, 419–428. [Google Scholar] [CrossRef]
  10. Dabovic, B.; Chen, Y.; Choi, J.; Davis, E.C.; Sakai, L.Y.; Todorovic, V.; Vassallo, M.; Zilberberg, L.; Singh, A.; Rifkin, D.B. Control of lung development by latent TGF-β binding proteins. J. Cell. Physiol. 2011, 226, 1499–1509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Noda, K.; Dabovic, B.; Takagi, K.; Inoue, T.; Horiguchi, M.; Hirai, M.; Fujikawa, Y.; Akama, T.O.; Kusumoto, K.; Zilberberg, L.; et al. Latent TGF-β binding protein 4 promotes elastic fiber assembly by interacting with fibulin-5. Proc. Natl. Acad. Sci. USA 2013, 110, 2852–2857. [Google Scholar] [CrossRef] [PubMed]
  12. Sterner-Kock, A.; Thorey, I.S.; Koli, K.; Wempe, F.; Otte, J.; Bangsow, T.; Kuhlmeier, K.; Kirchner, T.; Jin, S.; Keski-Oja, J.; et al. Disruption of the gene encoding the latent transforming growth factor-beta binding protein 4 (LTBP-4) causes abnormal lung development, cardiomyopathy, and colorectal cancer. Genes Dev. 2002, 16, 2264–2273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Buscemi, L.; Ramonet, D.; Klingberg, F.; Formey, A.; Smith-Clerc, J.; Meister, J.J.; Hinz, B. The single-molecule mechanics of the latent TGF-β1 complex. Curr. Biol. 2011, 21, 2046–2054. [Google Scholar] [CrossRef] [PubMed]
  14. Saito, A.; Suzuki, H.I.; Horie, M.; Ohshima, M.; Morishita, Y.; Abiko, Y.; Nagase, T. An integrated expression profiling reveals target genes of TGF-β and TNF-α possibly mediated by microRNAs in lung cancer cells. PLoS ONE 2013, 8, e56587. [Google Scholar] [CrossRef] [PubMed]
  15. Horie, M.; Saito, A.; Noguchi, S.; Yamaguchi, Y.; Ohshima, M.; Morishita, Y.; Suzuki, H.I.; Kohyama, T.; Nagase, T. Differential knockdown of TGF-β ligands in a three-dimensional co-culture tumor-stromal interaction model of lung cancer. BMC Cancer 2014, 14, 580. [Google Scholar] [CrossRef] [PubMed]
  16. Heldin, C.H.; Miyazono, K.; ten Dijke, P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature 1997, 390, 465–471. [Google Scholar] [CrossRef] [PubMed]
  17. Derynck, R.; Zhang, Y.E. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature 2003, 425, 577–584. [Google Scholar] [CrossRef] [PubMed]
  18. Mullen, A.C.; Orlando, D.A.; Newman, J.J.; Lovén, J.; Kumar, R.M.; Bilodeau, S.; Reddy, J.; Guenther, M.G.; DeKoter, R.P.; Young, R.A. Master transcription factors determine cell-type-specific responses to TGF-β signaling. Cell 2011, 147, 565–576. [Google Scholar] [CrossRef] [PubMed]
  19. Massague, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef] [PubMed]
  20. Miyazawa, K.; Miyazono, K. Regulation of TGF-β Family Signaling by Inhibitory Smads. Cold Spring Harb. Perspect. Biol. 2017, 9, a022095. [Google Scholar] [CrossRef] [PubMed]
  21. Nakao, A.; Afrakhte, M.; Morén, A.; Nakayama, T.; Christian, J.L.; Heuchel, R.; Itoh, S.; Kawabata, M.; Heldin, N.E.; Heldin, C.H.; et al. Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling. Nature 1997, 389, 631–635. [Google Scholar] [PubMed]
  22. Ebisawa, T.; Fukuchi, M.; Murakami, G.; Chiba, T.; Tanaka, K.; Imamura, T.; Miyazono, K. Smurf1 interacts with transforming growth factor-beta type I receptor through Smad7 and induces receptor degradation. J. Biol. Chem. 2001, 276, 12477–12480. [Google Scholar] [CrossRef] [PubMed]
  23. Massague, J. Integration of Smad and MAPK pathways: A link and a linker revisited. Genes Dev. 2003, 17, 2993–2997. [Google Scholar] [CrossRef] [PubMed]
  24. Kang, J.S.; Saunier, E.F.; Akhurst, R.J.; Derynck, R. The type I TGF-beta receptor is covalently modified and regulated by sumoylation. Nat. Cell Biol. 2008, 10, 654–664. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, J.; Du, Y.; Deng, H. Direct lineage reprogramming: Strategies, mechanisms, and applications. Cell Stem Cell 2015, 16, 119–134. [Google Scholar] [CrossRef] [PubMed]
  26. Minoo, P.; Su, G.; Drum, H.; Bringas, P.; Kimura, S. Defects in tracheoesophageal and lung morphogenesis in Nkx2.1(−/−) mouse embryos. Dev. Biol. 1999, 209, 60–71. [Google Scholar] [CrossRef] [PubMed]
  27. Bohinski, R.J.; Di Lauro, R.; Whitsett, J.A. The lung-specific surfactant protein B gene promoter is a target for thyroid transcription factor 1 and hepatocyte nuclear factor 3, indicating common factors for organ-specific gene expression along the foregut axis. Mol. Cell. Biol. 1994, 14, 5671–5681. [Google Scholar] [CrossRef] [PubMed]
  28. Minoo, P.; Hu, L.; Zhu, N.; Borok, Z.; Bellusci, S.; Groffen, J.; Kardassis, D.; Li, C. SMAD3 prevents binding of NKX2.1 and FOXA1 to the SpB promoter through its MH1 and MH2 domains. Nucleic Acids Res. 2008, 36, 179–188. [Google Scholar] [CrossRef] [PubMed]
  29. Isogaya, K.; Koinuma, D.; Tsutsumi, S.; Saito, R.A.; Miyazawa, K.; Aburatani, H.; Miyazono, K. A Smad3 and TTF-1/NKX2-1 complex regulates Smad4-independent gene expression. Cell Res. 2014, 24, 994–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Saito, R.A.; Watabe, T.; Horiguchi, K.; Kohyama, T.; Saitoh, M.; Nagase, T.; Miyazono, K. Thyroid transcription factor-1 inhibits transforming growth factor-beta-mediated epithelial-to-mesenchymal transition in lung adenocarcinoma cells. Cancer Res. 2009, 69, 2783–2791. [Google Scholar] [CrossRef] [PubMed]
  31. Herriges, M.; Morrisey, E.E. Lung development: Orchestrating the generation and regeneration of a complex organ. Development 2014, 141, 502–513. [Google Scholar] [CrossRef] [PubMed]
  32. Bragg, A.D.; Moses, H.L.; Serra, R. Signaling to the epithelium is not sufficient to mediate all of the effects of transforming growth factor beta and bone morphogenetic protein 4 on murine embryonic lung development. Mech. Dev. 2001, 109, 13–26. [Google Scholar] [CrossRef]
  33. Shull, M.M.; Ormsby, I.; Kier, A.B.; Pawlowski, S.; Diebold, R.J.; Yin, M.; Allen, R.; Sidman, C.; Proetzel, G.; Calvin, D.; et al. Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature 1992, 359, 693–699. [Google Scholar] [CrossRef] [PubMed]
  34. Kulkarni, A.B.; Huh, C.G.; Becker, D.; Geiser, A.; Lyght, M.; Flanders, K.C.; Roberts, A.B.; Sporn, M.B.; Ward, J.M.; Karlsson, S. Transforming growth factor beta 1 null mutation in mice causes excessive inflammatory response and early death. Proc. Natl. Acad. Sci. USA 1993, 90, 770–774. [Google Scholar] [CrossRef] [PubMed]
  35. Sanford, L.P.; Ormsby, I.; Gittenberger-de Groot, A.C.; Sariola, H.; Friedman, R.; Boivin, G.P.; Cardell, E.L.; Doetschman, T. TGFbeta2 knockout mice have multiple developmental defects that are non-overlapping with other TGFbeta knockout phenotypes. Development 1997, 124, 2659–2670. [Google Scholar] [PubMed]
  36. Kaartinen, V.; Voncken, J.W.; Shuler, C.; Warburton, D.; Bu, D.; Heisterkamp, N.; Groffen, J. Abnormal lung development and cleft palate in mice lacking TGF-beta 3 indicates defects of epithelial-mesenchymal interaction. Nat. Genet. 1995, 11, 415–421. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, H.; Zhuang, F.; Liu, Y.H.; Xu, B.; Del Moral, P.; Deng, W.; Chai, Y.; Kolb, M.; Gauldie, J.; Warburton, D.; et al. TGF-beta receptor II in epithelia versus mesenchyme plays distinct roles in the developing lung. Eur. Respir. J. 2008, 32, 285–295. [Google Scholar] [CrossRef] [PubMed]
  38. Li, M.; Krishnaveni, M.S.; Li, C.; Zhou, B.; Xing, Y.; Banfalvi, A.; Li, A.; Lombardi, V.; Akbari, O.; Borok, Z.; et al. Epithelium-specific deletion of TGF-β receptor type II protects mice from bleomycin-induced pulmonary fibrosis. J. Clin. Investig. 2011, 121, 277–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Bonniaud, P.; Kolb, M.; Galt, T.; Robertson, J.; Robbins, C.; Stampfli, M.; Lavery, C.; Margetts, P.J.; Roberts, A.B.; Gauldie, J. Smad3 null mice develop airspace enlargement and are resistant to TGF-beta-mediated pulmonary fibrosis. J. Immunol. 2004, 173, 2099–2108. [Google Scholar] [CrossRef] [PubMed]
  40. Chen, H.; Sun, J.; Buckley, S.; Chen, C.; Warburton, D.; Wang, X.F.; Shi, W. Abnormal mouse lung alveolarization caused by Smad3 deficiency is a developmental antecedent of centrilobular emphysema. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005, 288, L683–L691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Xing, Y.; Li, C.; Li, A.; Sridurongrit, S.; Tiozzo, C.; Bellusci, S.; Borok, Z.; Kaartinen, V.; Minoo, P. Signaling via Alk5 controls the ontogeny of lung Clara cells. Development 2010, 137, 825–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Li, M.; Li, C.; Liu, Y.H.; Xing, Y.; Hu, L.; Borok, Z.; Kwong, K.Y.; Minoo, P. Mesodermal deletion of transforming growth factor-beta receptor II disrupts lung epithelial morphogenesis: Cross-talk between TGF-beta and Sonic hedgehog pathways. J. Biol. Chem. 2008, 283, 36257–36264. [Google Scholar] [CrossRef] [PubMed]
  43. Li, A.; Ma, S.; Smith, S.M.; Lee, M.K.; Fischer, A.; Borok, Z.; Bellusci, S.; Li, C.; Minoo, P. Mesodermal ALK5 controls lung myofibroblast versus lipofibroblast cell fate. BMC Biol. 2016, 14, 19. [Google Scholar] [CrossRef] [PubMed]
  44. Zhou, L.; Dey, C.R.; Wert, S.E.; Whitsett, J.A. Arrested lung morphogenesis in transgenic mice bearing an SP-C-TGF-beta 1 chimeric gene. Dev. Biol. 1996, 175, 227–238. [Google Scholar] [CrossRef] [PubMed]
  45. Xing, Y.; Li, C.; Hu, L.; Tiozzo, C.; Li, M.; Chai, Y.; Bellusci, S.; Anderson, S.; Minoo, P. Mechanisms of TGFbeta inhibition of LUNG endodermal morphogenesis: The role of TbetaRII, Smads, Nkx2.1 and Pten. Dev. Biol. 2008, 320, 340–350. [Google Scholar] [CrossRef] [PubMed]
  46. Hogan, B.L.; Barkauskas, C.E.; Chapman, H.A.; Epstein, J.A.; Jain, R.; Hsia, C.C.; Niklason, L.; Calle, E.; Le, A.; Randell, S.H.; et al. Repair and regeneration of the respiratory system: Complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014, 15, 123–138. [Google Scholar] [CrossRef] [PubMed]
  47. Ryan, R.M.; Mineo-Kuhn, M.M.; Kramer, C.M.; Finkelstein, J.N. Growth factors alter neonatal type II alveolar epithelial cell proliferation. Am. J. Physiol. 1994, 266, L17–L22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Saitoh, M. Epithelial-mesenchymal transition is regulated at post-transcriptional levels by transforming growth factor-β signaling during tumor progression. Cancer Sci. 2015, 106, 481–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Shirakihara, T.; Saitoh, M.; Miyazono, K. Differential regulation of epithelial and mesenchymal markers by deltaEF1 proteins in epithelial mesenchymal transition induced by TGF-beta. Mol. Biol. Cell 2007, 18, 3533–3544. [Google Scholar] [CrossRef] [PubMed]
  50. Gregory, P.A.; Bert, A.G.; Paterson, E.L.; Barry, S.C.; Tsykin, A.; Farshid, G.; Vadas, M.A.; Khew-Goodall, Y.; Goodall, G.J. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat. Cell Biol. 2008, 10, 593–601. [Google Scholar] [CrossRef] [PubMed]
  51. Willis, B.C.; Liebler, J.M.; Luby-Phelps, K.; Nicholson, A.G.; Crandall, E.D.; du Bois, R.M.; 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. [Google Scholar] [CrossRef]
  52. Kage, H.; Borok, Z. EMT and interstitial lung disease: A mysterious relationship. Curr. Opin. Pulm. Med. 2012, 18, 517–523. [Google Scholar] [CrossRef] [PubMed]
  53. Borok, Z.; Hami, A.; Danto, S.I.; Zabski, S.M.; Crandall, E.D. Rat serum inhibits progression of alveolar epithelial cells toward the type I cell phenotype in vitro. Am. J. Respir. Cell Mol. Biol. 1995, 12, 50–55. [Google Scholar] [CrossRef] [PubMed]
  54. Zhao, L.; Yee, M.; O’Reilly, M.A. Transdifferentiation of alveolar epithelial type II to type I cells is controlled by opposing TGF-β and BMP signaling. Am. J. Physiol. Lung Cell. Mol. Physiol. 2013, 305, L409–L418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Barkauskas, C.E.; Cronce, M.J.; Rackley, C.R.; Bowie, E.J.; Keene, D.R.; Stripp, B.R.; Randell, S.H.; Noble, P.W.; Hogan, B.L. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Investig. 2013, 123, 3025–3036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Jain, R.; Barkauskas, C.E.; Takeda, N.; Bowie, E.J.; Aghajanian, H.; Wang, Q.; Padmanabhan, A.; Manderfield, L.J.; Gupta, M.; Li, D.; et al. Plasticity of Hopx(+) type I alveolar cells to regenerate type II cells in the lung. Nat. Commun. 2015, 6, 6727. [Google Scholar] [CrossRef] [PubMed]
  57. Jacob, A.; Morley, M.; Hawkins, F.; McCauley, K.B.; Jean, J.C.; Heins, H.; Na, C.L.; Weaver, T.E.; Vedaie, M.; Hurley, K.; et al. Differentiation of Human Pluripotent Stem Cells into Functional Lung Alveolar Epithelial Cells. Cell Stem Cell 2017, 21, 472–488. [Google Scholar] [CrossRef] [PubMed]
  58. Yamamoto, Y.; Gotoh, S.; Korogi, Y.; Seki, M.; Konishi, S.; Ikeo, S.; Sone, N.; Nagasaki, T.; Matsumoto, H.; Muro, S.; et al. Long-term expansion of alveolar stem cells derived from human iPS cells in organoids. Nat. Methods 2017, 14, 1097–1106. [Google Scholar] [CrossRef] [PubMed]
  59. Green, M.D.; Chen, A.; Nostro, M.C.; d’Souza, S.L.; Schaniel, C.; Lemischka, I.R.; Gouon-Evans, V.; Keller, G.; Snoeck, H.W. Generation of anterior foregut endoderm from human embryonic and induced pluripotent stem cells. Nat. Biotechnol. 2011, 29, 267–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. King, T.E., Jr.; Pardo, A.; Selman, M. Idiopathic pulmonary fibrosis. Lancet 2011, 378, 1949–1961. [Google Scholar] [CrossRef]
  61. Coker, R.K.; Laurent, G.J.; Jeffery, P.K.; du Bois, R.M.; Black, C.M.; McAnulty, R.J. Localisation of transforming growth factor beta1 and beta3 mRNA transcripts in normal and fibrotic human lung. Thorax 2001, 56, 549–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Broekelmann, T.J.; Limper, A.H.; Colby, T.V.; McDonald, J.A. Transforming growth factor beta 1 is present at sites of extracellular matrix gene expression in human pulmonary fibrosis. Proc. Natl. Acad. Sci. USA 1991, 88, 6642–6646. [Google Scholar] [CrossRef] [PubMed]
  63. Garrison, G.; Huang, S.K.; Okunishi, K.; Scott, J.P.; Kumar Penke, L.R.; Scruggs, A.M.; Peters-Golden, M. Reversal of myofibroblast differentiation by prostaglandin E(2). Am. J. Respir. Cell Mol. Biol. 2013, 48, 550–558. [Google Scholar] [CrossRef] [PubMed]
  64. Bauman, K.A.; Wettlaufer, S.H.; Okunishi, K.; Vannella, K.M.; Stoolman, J.S.; Huang, S.K.; Courey, A.J.; White, E.S.; Hogaboam, C.M.; Simon, R.H.; et al. The antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice. J. Clin. Investig. 2010, 120, 1950–1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Cho, M.H.; McDonald, M.L.; Zhou, X.; Mattheisen, M.; Castaldi, P.J.; Hersh, C.P.; Demeo, D.L.; Sylvia, J.S.; Ziniti, J.; Laird, N.M.; et al. Risk loci for chronic obstructive pulmonary disease: A genome-wide association study and meta-analysis. Lancet Respir. Med. 2014, 2, 214–225. [Google Scholar] [CrossRef]
  66. Takizawa, H.; Tanaka, M.; Takami, K.; Ohtoshi, T.; Ito, K.; Satoh, M.; Okada, Y.; Yamasawa, F.; Nakahara, K.; Umeda, A. Increased expression of transforming growth factor-beta1 in small airway epithelium from tobacco smokers and patients with chronic obstructive pulmonary disease (COPD). Am. J. Respir. Crit. Care Med. 2001, 163, 1476–1483. [Google Scholar] [CrossRef] [PubMed]
  67. Zhao, J.; Shi, W.; Wang, Y.L.; Chen, H.; Bringas, P., Jr.; Datto, M.B.; Frederick, J.P.; Wang, X.F.; Warburton, D. Smad3 deficiency attenuates bleomycin-induced pulmonary fibrosis in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 2002, 282, L585–L593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Morris, D.G.; Huang, X.; Kaminski, N.; Wang, Y.; Shapiro, S.D.; Dolganov, G.; Glick, A.; Sheppard, D. Loss of integrin alpha(v)beta6-mediated TGF-beta activation causes Mmp12-dependent emphysema. Nature 2003, 422, 169–173. [Google Scholar] [CrossRef] [PubMed]
  69. Munger, J.S.; Huang, X.; Kawakatsu, H.; Griffiths, M.J.; Dalton, S.L.; Wu, J.; Pittet, J.F.; Kaminski, N.; Garat, C.; Matthay, M.A.; et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: A mechanism for regulating pulmonary inflammation and fibrosis. Cell 1999, 96, 319–328. [Google Scholar] [CrossRef]
  70. Horan, G.S.; Wood, S.; Ona, V.; Li, D.J.; Lukashev, M.E.; Weinreb, P.H.; Simon, K.J.; Hahm, K.; Allaire, N.E.; Rinaldi, N.J.; et al. Partial inhibition of integrin alpha(v)beta6 prevents pulmonary fibrosis without exacerbating inflammation. Am. J. Respir. Crit. Care Med. 2008, 177, 56–65. [Google Scholar] [CrossRef] [PubMed]
  71. Jankowich, M.D.; Roundsm, S.I.S. Combined pulmonary fibrosis and emphysema syndrome: A review. Chest 2012, 141, 222–231. [Google Scholar] [CrossRef] [PubMed]
  72. Papi, A.; Brightling, C.; Pedersen, S.E.; Reddel, H.K. Asthma. Lancet 2018, 391, 783–800. [Google Scholar] [CrossRef]
  73. Hirota, N.; Martin, J.G. Mechanisms of airway remodeling. Chest 2013, 144, 1026–1032. [Google Scholar] [CrossRef] [PubMed]
  74. James, A.L.; Bai, T.R.; Mauad, T.; Abramson, M.J.; Dolhnikoff, M.; McKay, K.O.; Maxwell, P.S.; Elliot, J.G.; Green, F.H. Airway smooth muscle thickness in asthma is related to severity but not duration of asthma. Eur. Respir. J. 2009, 34, 1040–1045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Moffatt, M.F.; Gut, I.G.; Demenais, F.; Strachan, D.P.; Bouzigon, E.; Heath, S.; von Mutius, E.; Farrall, M.; Lathrop, M.; Cookson, W.O. A large-scale, consortium-based genomewide association study of asthma. N. Engl. J. Med. 2010, 363, 1211–1221. [Google Scholar] [CrossRef] [PubMed]
  76. Yao, Y.S.; Chang, W.W.; He, L.P.; Jin, Y.L.; Li, C.P. An updated meta-analysis of transforming growth factor-β1 gene: Three well-characterized polymorphisms with asthma. Hum. Immunol. 2016, 77, 1291–1299. [Google Scholar] [CrossRef] [PubMed]
  77. Frischmeyer-Guerrerio, P.A.; Guerrerio, A.L.; Oswald, G.; Chichester, K.; Myers, L.; Halushka, M.K.; Oliva-Hemker, M.; Wood, R.A.; Dietz, H.C. TGFβ receptor mutations impose a strong predisposition for human allergic disease. Sci. Transl. Med. 2013, 5, 195ra94. [Google Scholar] [CrossRef] [PubMed]
  78. Redington, A.E.; Madden, J.; Frew, A.J.; Djukanovic, R.; Roche, W.R.; Holgate, S.T.; Howarth, P.H. Transforming growth factor-beta 1 in asthma. Measurement in bronchoalveolar lavage fluid. Am. J. Respir. Crit. Care Med. 1997, 156, 642–647. [Google Scholar] [CrossRef] [PubMed]
  79. Vignola, A.M.; Chanez, P.; Chiappara, G.; Merendino, A.; Pace, E.; Rizzo, A.; la Rocca, A.M.; Bellia, V.; Bonsignore, G.; Bousquet, J. Transforming growth factor-beta expression in mucosal biopsies in asthma and chronic bronchitis. Am. J. Respir. Crit. Care Med. 1997, 156, 591–599. [Google Scholar] [CrossRef] [PubMed]
  80. Ohno, I.; Nitta, Y.; Yamauchi, K.; Hoshi, H.; Honma, M.; Woolley, K.; O’Byrne, P.; Tamura, G.; Jordana, M.; Shirato, K. Transforming growth factor beta 1 (TGF beta 1) gene expression by eosinophils in asthmatic airway inflammation. Am. J. Respir. Cell Mol. Biol. 1996, 15, 404–409. [Google Scholar] [CrossRef] [PubMed]
  81. Ojiaku, C.A.; Cao, G.; Zhu, W.; Yoo, E.J.; Shumyatcher, M.; Himes, B.E.; An, S.S.; Panettieri, R.A., Jr. TGF-β1 Evokes Human Airway Smooth Muscle Cell Shortening and Hyperresponsiveness via Smad3. Am. J. Respir. Cell Mol. Biol. 2018, 58, 575–584. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, W.; Jin, W.; Hardegen, N.; Lei, K.J.; Li, L.; Marinos, N.; McGrady, G.; Wahl, S.M. Conversion of peripheral CD4 + CD25 − naive T cells to CD4 + CD25 + regulatory T cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 2003, 198, 1875–1886. [Google Scholar] [CrossRef] [PubMed]
  83. Marie, J.C.; Letterio, J.J.; Gavin, M.; Rudensky, A.Y. TGF-beta1 maintains suppressor function and Foxp3 expression in CD4 + CD25 + regulatory T cells. J. Exp. Med. 2005, 201, 1061–1067. [Google Scholar] [CrossRef] [PubMed]
  84. Kearley, J.; Robinson, D.S.; Lloyd, C.M. CD4 + CD25 + regulatory T cells reverse established allergic airway inflammation and prevent airway remodeling. J. Allergy Clin. Immunol. 2008, 122, 617–624. [Google Scholar] [CrossRef] [PubMed]
  85. Li, M.O.; Sanjabi, S.; Flavell, R.A. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 2006, 25, 455–471. [Google Scholar] [CrossRef] [PubMed]
  86. Zhu, J.; Yamane, H.; Paul, W.E. Differentiation of effector CD4 T cell populations. Annu. Rev. Immunol. 2010, 28, 445–489. [Google Scholar] [CrossRef] [PubMed]
  87. Bettelli, E.; Carrier, Y.; Gao, W.; Korn, T.; Strom, T.B.; Oukka, M.; Weiner, H.L.; Kuchroo, V.K. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006, 441, 235–238. [Google Scholar] [CrossRef] [PubMed]
  88. Mangan, P.R.; Harrington, L.E.; O’Quinn, D.B.; Helms, W.S.; Bullard, D.C.; Elson, C.O.; Hatton, R.D.; Wahl, S.M.; Schoeb, T.R.; Weaver, C.T. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature 2006, 441, 231–234. [Google Scholar] [CrossRef] [PubMed]
  89. Chen, W.; ten Dijke, P. Immunoregulation by members of the TGFβ superfamily. Nat. Rev. Immunol. 2016, 16, 723–740. [Google Scholar] [CrossRef] [PubMed]
  90. Herbst, R.S.; Heymach, J.V.; Lippman, S.M. Lung cancer. N. Engl. J. Med. 2008, 359, 1367–1380. [Google Scholar] [CrossRef] [PubMed]
  91. Hasegawa, Y.; Takanashi, S.; Kanehira, Y.; Tsushima, T.; Imai, T.; Okumura, K. Transforming growth factor β1 level correlates with angiogenesis, tumor progression, and prognosis in patients with nonsmall cell lung carcinoma. Cancer 2001, 91, 964–971. [Google Scholar] [CrossRef]
  92. Massague, J. TGFbeta in Cancer. Cell 2008, 134, 215–230. [Google Scholar] [CrossRef] [PubMed]
  93. Markowitz, S.; Wang, J.; Myeroff, L.; Parsons, R.; Sun, L.; Lutterbaugh, J.; Fan, R.S.; Zborowska, E.; Kinzler, K.W.; Vogelstein, B.; et al. Inactivation of the type II TGF-beta receptor in colon cancer cells with microsatellite instability. Science 1995, 268, 1336–1338. [Google Scholar] [CrossRef] [PubMed]
  94. Hahn, S.A.; Schutte, M.; Hoque, A.T.; Moskaluk, C.A.; da Costa, L.T.; Rozenblum, E.; Weinstein, C.L.; Fischer, A.; Yeo, C.J.; Hruban, R.H.; et al. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science 1996, 271, 350–353. [Google Scholar] [CrossRef] [PubMed]
  95. Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature 2014, 511, 543–550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kohno, T.; Nakaoku, T.; Tsuta, K.; Tsuchihara, K.; Matsumoto, S.; Yoh, K.; Goto, K. Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer. Transl. Lung Cancer Res. 2015, 4, 156–164. [Google Scholar] [PubMed]
  97. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Investig. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Yauch, R.L.; Januario, T.; Eberhard, D.A.; Cavet, G.; Zhu, W.; Fu, L.; Pham, T.Q.; Soriano, R.; Stinson, J.; Seshagiri, S.; et al. Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin. Cancer Res. 2005, 11, 8686–8698. [Google Scholar] [CrossRef] [PubMed]
  99. Schliekelman, M.J.; Taguchi, A.; Zhu, J.; Dai, X.; Rodriguez, J.; Celiktas, M.; Zhang, Q.; Chin, A.; Wong, C.H.; Wang, H.; et al. Molecular portraits of epithelial, mesenchymal, and hybrid States in lung adenocarcinoma and their relevance to survival. Cancer Res. 2015, 75, 1789–1800. [Google Scholar] [CrossRef] [PubMed]
  100. Horiguchi, K.; Shirakihara, T.; Nakano, A.; Imamura, T.; Miyazono, K.; Saitoh, M. Role of Ras signaling in the induction of snail by transforming growth factor-beta. J. Biol. Chem. 2009, 284, 245–253. [Google Scholar] [CrossRef] [PubMed]
  101. Mani, S.A.; Guo, W.; Liao, M.J.; Eaton, E.N.; Ayyanan, A.; Zhou, A.Y.; Brooks, M.; Reinhard, F.; Zhang, C.C.; Shipitsin, M.; et al. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 2008, 133, 704–715. [Google Scholar] [CrossRef] [PubMed]
  102. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Bierie, B.; Moses, H.L. Tumour microenvironment: TGFbeta: The molecular Jekyll and Hyde of cancer. Nat. Rev. Cancer 2006, 6, 506–520. [Google Scholar] [CrossRef] [PubMed]
  104. Ikushima, H.; Miyazono, K. TGFbeta signalling: A complex web in cancer progression. Nat. Rev. Cancer 2010, 10, 415–424. [Google Scholar] [CrossRef] [PubMed]
  105. Pickup, M.; Novitskiy, S.; Moses, H.L. The roles of TGFβ in the tumour microenvironment. Nat. Rev. Cancer 2013, 13, 788–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Noguchi, M.; Morikawa, A.; Kawasaki, M.; Matsuno, Y.; Yamada, T.; Hirohashi, S.; Kondo, H.; Shimosato, Y. Small adenocarcinoma of the lung. Histologic characteristics and prognosis. Cancer 1995, 75, 2844–2852. [Google Scholar] [CrossRef]
  107. Micke, P.; Ostman, A. Tumour-stroma interaction: Cancer-associated fibroblasts as novel targets in anti-cancer therapy? Lung Cancer 2004, 45 (Suppl. 2), S163–S175. [Google Scholar] [CrossRef] [PubMed]
  108. Saito, R.A.; Micke, P.; Paulsson, J.; Augsten, M.; Peña, C.; Jönsson, P.; Botling, J.; Edlund, K.; Johansson, L.; Carlsson, P.; et al. Forkhead box F1 regulates tumor-promoting properties of cancer-associated fibroblasts in lung cancer. Cancer Res. 2010, 70, 2644–2654. [Google Scholar] [CrossRef] [PubMed]
  109. Horie, M.; Saito, A.; Mikami, Y.; Ohshima, M.; Morishita, Y.; Nakajima, J.; Kohyama, T.; Nagase, T. Characterization of human lung cancer-associated fibroblasts in three-dimensional in vitro co-culture model. Biochem. Biophys. Res. Commun. 2012, 423, 158–163. [Google Scholar] [CrossRef] [PubMed]
  110. Navab, R.; Strumpf, D.; Bandarchi, B.; Zhu, C.Q.; Pintilie, M.; Ramnarine, V.R.; Ibrahimov, E.; Radulovich, N.; Leung, L.; Barczyk, M.; et al. Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer. Proc. Natl. Acad. Sci. USA 2011, 108, 7160–7165. [Google Scholar] [CrossRef] [PubMed]
  111. Heldin, C.H.; Rubin, K.; Pietras, K.; Ostman, A. High interstitial fluid pressure—An obstacle in cancer therapy. Nat. Rev. Cancer 2004, 4, 806–813. [Google Scholar] [CrossRef] [PubMed]
  112. Yoshimatsu, Y.; Miyazaki, H.; Watabe, T. Roles of signaling and transcriptional networks in pathological lymphangiogenesis. Adv. Drug Deliv. Rev. 2016, 99 Pt B, 161–171. [Google Scholar] [CrossRef]
  113. Flavell, R.A.; Sanjabi, S.; Wrzesinski, S.H.; Licona-Limón, P. The polarization of immune cells in the tumour environment by TGFbeta. Nat. Rev. Immunol. 2010, 10, 554–567. [Google Scholar] [CrossRef] [PubMed]
  114. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [PubMed]
  115. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed]
  116. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel Iii, E.E.; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef] [PubMed]
  117. Micke, P.; Faldum, A.; Metz, T.; Beeh, K.M.; Bittinger, F.; Hengstler, J.G.; Buhl, R. Staging small cell lung cancer: Veterans Administration Lung Study Group versus International Association for the Study of Lung Cancer—What limits limited disease? Lung Cancer 2002, 37, 271–276. [Google Scholar] [CrossRef]
  118. Borges, M.; Linnoila, R.I.; van de Velde, H.J.; Chen, H.; Nelkin, B.D.; Mabry, M.; Baylin, S.B.; Ball, D.W. An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature 1997, 386, 852–855. [Google Scholar] [CrossRef] [PubMed]
  119. Horie, M.; Saito, A.; Ohshima, M.; Suzuki, H.I.; Nagase, T. YAP and TAZ modulate cell phenotype in a subset of small cell lung cancer. Cancer Sci. 2016, 107, 1755–1766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Mollaoglu, G.; Guthrie, M.R.; Böhm, S.; Brägelmann, J.; Can, I.; Ballieu, P.M.; Marx, A.; George, J.; Heinen, C.; Chalishazar, M.D.; et al. MYC Drives Progression of Small Cell Lung Cancer to a Variant Neuroendocrine Subtype with Vulnerability to Aurora Kinase Inhibition. Cancer Cell 2017, 31, 270–285. [Google Scholar] [CrossRef] [PubMed]
  121. Hougaard, S.; Norgaard, P.; Abrahamsen, N.; Moses, H.L.; Spang-Thomsen, M.; Poulsen, H.S. Inactivation of the transforming growth factor β type II receptor in human small cell lung cancer cell lines. Br. J. Cancer 1999, 79, 1005–1011. [Google Scholar] [CrossRef] [PubMed]
  122. Murai, F.; Koinuma, D.; Shinozaki-Ushiku, A.; Fukayama, M.; Miyaozono, K.; Ehata, S. EZH2 promotes progression of small cell lung cancer by suppressing the TGF-β-Smad-ASCL1 pathway. Cell Discov. 2015, 1, 15026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Horie, M.; Kaczkowski, B.; Ohshima, M.; Matsuzaki, H.; Noguchi, S.; Mikami, Y.; Lizio, M.; Itoh, M.; Kawaji, H.; Lassmann, T.; et al. Integrative CAGE and DNA Methylation Profiling Identify Epigenetically Regulated Genes in NSCLC. Mol. Cancer Res. 2017, 15, 1354–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Morikawa, M.; Koinuma, D.; Miyazono, K.; Heldin, C.H. Genome-wide mechanisms of Smad binding. Oncogene 2013, 32, 1609–1615. [Google Scholar] [CrossRef] [PubMed]
  125. Whyte, W.A.; Orlando, D.A.; Hnisz, D.; Abraham, B.J.; Lin, C.Y.; Kagey, M.H.; Rahl, P.B.; Lee, T.I.; Young, R.A. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 2013, 153, 307–319. [Google Scholar] [CrossRef] [PubMed]
  126. Suzuki, H.I.; Young, R.A.; Sharp, P.A. Super-Enhancer-Mediated RNA Processing Revealed by Integrative MicroRNA Network Analysis. Cell 2017, 168, 1000–1014. [Google Scholar] [CrossRef] [PubMed]
  127. Horie, M.; Miyashita, N.; Mikami, Y.; Noguchi, S.; Yamauchi, Y.; Suzukawa, M.; Fukami, T.; Ohta, K.; Asano, Y.; Sato, S.; et al. TBX4 is involved in the super-enhancer-driven transcriptional programs underlying features specific to lung fibroblasts. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 314, L177–L191. [Google Scholar] [CrossRef] [PubMed]
  128. Chang, H.; Liu, Y.; Xue, M.; Liu, H.; Du, S.; Zhang, L.; Wang, P. Synergistic action of master transcription factors controls epithelial-to-mesenchymal transition. Nucleic Acids Res. 2016, 44, 2514–2527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Akhurst, R.J.; Hata, A. Targeting the TGFb signalling pathway in disease. Nat. Rev. Drug Discov. 2012, 11, 790–811. [Google Scholar] [CrossRef] [PubMed]
  130. Nabhan, A.N.; Brownfield, D.G.; Harbury, P.B.; Krasnow, M.A.; Desai, T.J. Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 2018, 359, 1118–1123. [Google Scholar] [CrossRef] [PubMed]
  131. Treutlein, B.; Brownfield, D.G.; Wu, A.R.; Neff, N.F.; Mantalas, G.L.; Espinoza, F.H.; Desai, T.J.; Krasnow, M.A.; Quake, S.R. Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature 2014, 509, 371–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Structure of the airway and alveolus. TGF-β regulates epithelial-mesenchymal interactions and is crucial for branching morphogenesis and alveologenesis during development.
Figure 1. Structure of the airway and alveolus. TGF-β regulates epithelial-mesenchymal interactions and is crucial for branching morphogenesis and alveologenesis during development.
Ijms 19 02460 g001
Figure 2. Context-dependent action of TGF-β. (A) TGF-β promotes epithelial-mesenchymal transition (EMT) in alveolar epithelial cells to confer a mesenchymal phenotype, or in lung cancer cells to enhance migratory and invasive capacities. TGF-β induces EMT-related transcriptional repressors (SNAI1/SNAI2 and ZEB1/ZEB2) and inhibits the action of NKX2-1, a homeodomain transcription factor important for lung epithelial cell differentiation; (B) TGF-β promotes transdifferentiation of surfactant protein C (SPC)-positive type II alveolar epithelial cells to type I alveolar epithelial cells that express podoplanin (T1α); (C) TGF-β promotes transdifferentiation of lung fibroblasts to myofibroblasts positive for α-smooth muscle actin (α-SMA), and downregulates TBX4, a T-box family transcription factor unique to lung fibroblasts; (D) TGF-β suppresses induction of T helper type 1 (Th1) and type 2 (Th2) cells while positively regulating cell lineage specification from naïve CD4+ T cells to Th17 and regulatory T (Treg) cells. T-bet, GATA3, RORγt, and Foxp3 are master transcription factors for Th1, Th2, Th17, and Treg cells, respectively.
Figure 2. Context-dependent action of TGF-β. (A) TGF-β promotes epithelial-mesenchymal transition (EMT) in alveolar epithelial cells to confer a mesenchymal phenotype, or in lung cancer cells to enhance migratory and invasive capacities. TGF-β induces EMT-related transcriptional repressors (SNAI1/SNAI2 and ZEB1/ZEB2) and inhibits the action of NKX2-1, a homeodomain transcription factor important for lung epithelial cell differentiation; (B) TGF-β promotes transdifferentiation of surfactant protein C (SPC)-positive type II alveolar epithelial cells to type I alveolar epithelial cells that express podoplanin (T1α); (C) TGF-β promotes transdifferentiation of lung fibroblasts to myofibroblasts positive for α-smooth muscle actin (α-SMA), and downregulates TBX4, a T-box family transcription factor unique to lung fibroblasts; (D) TGF-β suppresses induction of T helper type 1 (Th1) and type 2 (Th2) cells while positively regulating cell lineage specification from naïve CD4+ T cells to Th17 and regulatory T (Treg) cells. T-bet, GATA3, RORγt, and Foxp3 are master transcription factors for Th1, Th2, Th17, and Treg cells, respectively.
Ijms 19 02460 g002
Figure 3. Upregulation of TGF-β ligands is observed in bronchial asthma, pulmonary fibrosis, emphysema, and lung cancer. Combined pulmonary fibrosis and emphysema (CPFE) and asthma-COPD overlap are increasingly recognized as distinct entities. Pulmonary fibrosis and emphysema are known as independent risk factors for lung cancer.
Figure 3. Upregulation of TGF-β ligands is observed in bronchial asthma, pulmonary fibrosis, emphysema, and lung cancer. Combined pulmonary fibrosis and emphysema (CPFE) and asthma-COPD overlap are increasingly recognized as distinct entities. Pulmonary fibrosis and emphysema are known as independent risk factors for lung cancer.
Ijms 19 02460 g003
Table 1. Genetically engineered mouse models that demonstrate the roles of TGF-β signaling in lung organogenesis.
Table 1. Genetically engineered mouse models that demonstrate the roles of TGF-β signaling in lung organogenesis.
Mouse ModelInductionTarget CellPhenotype in the LungsPhenotype in Other OrgansReference
Tgfb1 (−/−) perivasculitis with lymphocytic and plasmacytic infiltrationsystemic inflammation[33]
Tgfb1 (−/−) perivasculitis with lymphocytic and plasmacytic infiltration; interstitial pneumoniasystemic inflammation[34]
Tgfb2 (−/−) collapsed distal airways with dilated conducting airwaysCardiac, craniofacial, limb, spinal column, eye, inner ear, and urogenital defects[35]
Tgfb3 (−/−) atelectatic, pseudograndular histology with alveolar hypoplasia; mesenchymal thickening; extensive intrapulmonary and pleural hemorrhage; dilated conducting airwayscleft palate[36]
Smad3 (−/−) progressive lung airspace enlargement and emphysematous changesdefects in immune function with inflammatory lesions (originally reported by Yang X et al. in 1999)[39]
Smad3 (−/−) reduced pulmonary alveolarization and subsequent centrilobular emphysemadecreased growth rate (originally reported by Datto MB et al. in 1999)[40]
Tgfbr2flox/floxcrossed with SPC-rtTA/TetO-Cre micelung epithelial cellsretardation of postnatal lung alveolarization with markedly decreased type I alveolar epithelial cells [37]
Tgfbr2flox/floxcrossed with Dermo1-Cre micemesoderm-derived tissue including lung mesenchymeabnormal lung branching and reduced cell proliferationdefective secondary ventral body wall formation, congenital diaphragmatic hernia, and abnormal cardiac development[37]
Tgfbr2flox/floxcrossed with Dermo1-Cre micemesoderm-derived tissue including lung mesenchymefailure in branching morphogenesis and cystic airway malformationsabnormalities in multiple organs[42]
Tgfbr2flox/floxcrossed with Nkx2-1-Cre micelung epithelial cellsalveolar enlargement and non-progressive emphysema; resistance to TGF-β-mediated, bleomycin-induced lung injury [38]
Tgfbr1flox/floxcrossed with GATA5-Cre miceembryonic lung epitheliumimmature alveoli and formation of a disorganized and multi-layered epithelium in the proximal airways; marked reduction in the number of club cells [41]
Tgfbr1flox/floxcrossed with Dermo1-Cre micemesoderm-derived tissue including lung mesenchymereduced submesothelial mesenchyme; restricted α-SMA-positive cell fate and promoted lipofibroblast differentiation; defective epithelial differentiation; disrupted pulmonary vasculogenesis [43]

Share and Cite

MDPI and ACS Style

Saito, A.; Horie, M.; Nagase, T. TGF-β Signaling in Lung Health and Disease. Int. J. Mol. Sci. 2018, 19, 2460. https://doi.org/10.3390/ijms19082460

AMA Style

Saito A, Horie M, Nagase T. TGF-β Signaling in Lung Health and Disease. International Journal of Molecular Sciences. 2018; 19(8):2460. https://doi.org/10.3390/ijms19082460

Chicago/Turabian Style

Saito, Akira, Masafumi Horie, and Takahide Nagase. 2018. "TGF-β Signaling in Lung Health and Disease" International Journal of Molecular Sciences 19, no. 8: 2460. https://doi.org/10.3390/ijms19082460

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop