Respiratory Medicine (2014) 108, 1409e1423
Available online at www.sciencedirect.com
ScienceDirect journal homepage: www.elsevier.com/locate/rmed
REVIEW
Transforming growth factor b and severe asthma: A perfect storm Mazen Al-Alawi a, Tidi Hassan a, Sanjay H. Chotirmall b,* a b
Department of Respiratory Medicine, Mater Misericordiae Hospital, Eccles Street, Dublin 7, Ireland Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
Received 8 May 2014; accepted 21 August 2014
Available online 6 September 2014
KEYWORDS TGF-b; Asthma; Eosinophil; Therapy; Target
Summary Asthma is a chronic inflammatory airway disease involving complex interplay between resident and infiltrative cells, which in turn are regulated by a wide range of host mediators. Identifying useful biomarkers correlating with clinical symptoms and degree of airway obstruction remain important to effective future asthma treatments. Transforming growth factor b (TGF-b) is a major mediator involved in pro-inflammatory responses and fibrotic tissue remodeling within the asthmatic lung. Its role however, as a therapeutic target remains controversial. The aim of this review is to highlight its role in severe asthma including interactions with adaptive Thelper cells, cytokines and differentiation through regulatory T-cells. Associations between TGF-b and eosinophils will be addressed and the effects of genetic polymorphisms of the TGF-b1 gene explored in the context of asthma. We highlight TGF-b1 as a potential future therapeutic target in severe asthma including its importance in identifying emerging clinical phenotypes in asthmatic subjects who may be suitable for individualized therapy through TGF-b modulation. ª 2014 Elsevier Ltd. All rights reserved.
Contents Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1410 Transforming growth factor beta (TGF-ß) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1411 TGF-ß airway remodeling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1412 TGF-ß1 immunosupressive effects and regulatory inflammatory tolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . 1413 Pro-inflammatory role of TGF-ß1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1413 TGF-ß and eosinophil crosstalk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1414
* Corresponding author. E-mail address:
[email protected] (S.H. Chotirmall). http://dx.doi.org/10.1016/j.rmed.2014.08.008 0954-6111/ª 2014 Elsevier Ltd. All rights reserved.
1410
M. Al-Alawi et al. Genetic TGF-ß modifiers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1414 TGF-ß targeted therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1415 Conclusion and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1416 Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1416 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1417
Introduction Asthma is a complex chronic inflammatory lung disease where eosinophils, T-lymphocytes, macrophages, mast cells, neutrophils and epithelial cells play key roles in releasing cellular mediators including cytokines, chemokines and growth factors that all influence disease. Inhaled allergens sensitize dendritic cells to stimulate the proliferation of T helper type 2 (Th2) cells and subsequent release of cytokines that include interleukin IL-4, IL-5 and IL-13 [1]. Sensitized epithelial cells also release profibrotic mediators that include transforming growth factor-b (TGFb), fibroblast growth factor (Fgf) and endothelin, stimulating fibroblasts and myofibroblasts to release collagen, proteoglycan, and glycoproteins that induce airway thickening [2]. These mediators control the chronic inflammation observed that induces bronchoconstriction and airway remodeling with subsequent structural airway changes. Allergic asthma, representing a subset of asthmatics is characterized by chronic mucosal Th2 dominated inflammation. TGF-b plays a central role in the complex relationship between the propagation of the inflammatory cascade within the airway and association with suppressor T cell immune function. This review aims to highlight the evidence for the role of TGF-b in severe asthma which while contributing to disease pathogenesis remains less wellstudied. Its role in the asthmatic airway has been controversial likely because of its complex signaling pathways and immune interactions that influence several differing asthma phenotypes. As both airway remodeling
and poor treatment response are significant in severe asthma, it would appear logical that TGF-b would be relevant in this context and warrants review [3]. Severe asthma is a spectrum of disease with significant morbidity and mortality [4]. The National Heart, Lung, and Blood Institute (NHLBI) Severe Asthma Research Program identifies five clusters of phenotypically distinct individuals with severe asthma by performing cluster analysis. The groups range from early onset atopic asthma with normal lung function to subjects with severe airflow obstruction (Table 1) [5]. Airway remodeling in asthma constitutes subepithelial fibrosis, deposition of extracellular matrix protein, goblet cell hyperplasia, mucus gland and smooth muscle hypertrophy, and epithelial damage [6e8]. Inflammatory pathways regulate all such features [8]. Several key mediators including TGF-b, vascular endothelial growth factor (VEGF), ADAM metallopeptidase domain 33 (ADAM-33), matrix metalloproteinase-9 (MMP-9) and the Th2 cytokine family including IL-5, 13 and 14 have been identified. TGF-b has been a focal point of considerable investigation as both a mediator and effector molecule in the Th2 driven immune cascade. Produced by numerous cell types including epithelial cells, eosinophils, macrophages, and fibroblasts, it is involved in epithelial transformation, subepithelial fibrosis, airway smooth muscle (ASM) remodeling, microvascular changes, and mucus production. Insight into mechanisms of TGF-b signaling provides key information into the crosstalk between activation pathways, T-regulatory (Treg) signaling, and immune cell interactions hence its importance in the context of asthma.
Table 1 Demographics, clinical characteristics and biomarker subsets classified by cluster analysis. Five clinical phenotypes of asthma identified using unsupervised hierarchical cluster analysis [5]. Clusters contain subjects who meet the American Thoracic Society definition of severe asthma and correlate with phenotypes that can be based on the outlined clinical variables. Definition of abbreviation: FEV1 e Forced expiratory volume in 1 s, FVC e Forced vital capacity, BMI e Body mass index. Cluster group
1
2
3
4
5
FEV1 (%) FEV1/FVC (%) Age (mean yrs) Gender (% female) Asthma onset (%12 yrs) BMI > 30 (%) Atopy (%) Sputum neutrophils (%) Representative patient profile
108 78 27 80 39 24 85 23 Younger female patient with childhood onset asthma
68e108 74 33 67 36 31 78 33 Older female patient with childhood onset asthma
68e108 74 50 71 100 58 64 38 Older female patient with high BMI and late onset asthma
<68 64 38 53 28 44 83 35 Adult patient with childhood onset asthma and atopy
<68 57 49 63 69 51 66 48 Older female patient with late onset asthma and minimal atopy
Transforming growth factor beta in severe asthma Key pathological features of severe asthma share processes occurring in idiopathic pulmonary fibrosis and pulmonary arterial hypertension [9]. These include barrier dysfunction, smooth muscle proliferation and accumulation of lamina reticularis, myofibroblasts and smooth muscle cells that lead to fibrotic airway remodeling [10,11]. Characterization of pathophysiologic sub-phenotypes of severe refractory asthma is beneficial in expanding our understanding of the underlying biologic factors that are potentially ‘druggable’ therapeutic targets. For instance, sputum eosinophilia identified by cluster analysis is characteristic of later onset asthma (>12 years), severe eosinophilic inflammation and association with nasal polyposis and rhinosinusitis [12e15]. Targeting eosinophilia with the IL-4-blocker dupilumab has demonstrated fewer asthma exacerbations and a reduced level of Th2-associated inflammatory markers [16]. Treatment of severe refractory asthma remains a key clinical challenge and is associated with a high and disproportionate consumption of healthcare resources. Therefore, emerging avenues to improve understanding and therapy is critical for progress in the field [12,17].
Transforming growth factor beta (TGF-ß) The TGF-ß superfamily of ligands are multifunctional regulators implicated in various biological processes within the airways including alveolarization, epithelial and endothelial barrier function, immune cell recruitment, platelet aggregation, apoptosis, cell differentiation and proliferation [18]. These regulators consist of more than 33 members including TGF-ß, bone morphogenetic proteins (BMP),
1411 growth and differentiation factors (GDFs), activins and inhibins. This super-family in humans demonstrates three mammalian isoforms (TGF-ß1, 2 and 3) that share 60e80% homology. TGF-b isoforms have a number of specific and shared roles in the regulation of airway inflammation and the remodeling process [19]. The three isoforms described each have important roles in the regulation of inflammation, cell growth and differentiation [20]. TGF-b bound to latencyassociated peptide (LAP) is inactive and therefore understanding its regulation is pivotal to appreciating functional consequences [21,22]. Although each isoform is encoded by differing genes, important properties are shared such as common cell surface receptors and cellular targets [23]. The expression of TGFb-1 is altered in the asthmatic airway and current evidence suggests this is the case for both human and animal settings. Known as the ubiquitous prototype of this family, TGF-b1 is most prevalent in mammalian tissues [24]. Produced as an inactive latent complex made up of LAP and latent TGF-ß-binding protein (LTBP), it is targeted to the extracellular matrix (ECM) [25]. Maturation of inactive complexes depends on dissociation of covalent bonds that in turn rely on pH changes enabling binding to cell surface receptors. TGF-ß1 activation however requires further binding of a(v) integrin to a sequence in its pro-domain [23]. Its ability to recruit single transmembrane subunits into a hetero-oligomer enables TGF-ß1 to form a functional receptor (Fig. 1). TGF-ß1 signal transduction occurs through Smad-dependent or independent pathways [26]. TGF-ß1 type I and II receptors both transmembrane serine/threonine kinases (e.g. activin receptor-like kinase 5 (ALK5)) stimulate a cascade of intra-
Figure 1 TGF-b1 activation. Activation of TGF-ß1 receptors leads to dimerization [1] and phosphorylation of receptor-activated Smad proteins [2] that act as transcription factors to regulate gene expression. Smad 2 and 3 form complexes with Smad 4 [3] that translocate into the nucleus [4]. In a negative feedback loop, inhibitory Smad 7 induced by Smad 3 blocks TGF-ß signaling by binding to the type I receptor.[5].
1412 cytoplasmic intermediates termed Smads which facilitate signaling, and are surrogates to assess levels of pathway activity. Activated TGF-ß1 receptors lead to phosphorylation of Smad proteins that in turn are transcription factors regulating gene expression. Smads 2 and 3 form complexes with Co-Smad (Smad 4) and translocate into the nucleus. In a negative feedback loop, inhibitory Smad (Smad 7) induced by Smad 3 blocks TGF-ß signaling by type I receptor binding (Fig. 1). Increased TGF-ß1 signaling as evidenced by increased phospho-Smad 2 levels, have been reported in the bronchial biopsies of asthmatic subjects [27]. Smad 3 signaling however is required for the recognized allergeninduced airway remodeling and myofibroblast accumulation [28]. TGF-ß1 also activates Smad independent pathways such as the mitogen-activated protein kinase (MAPK) pathways: extra cellular signal-regulated kinase (ERK), p38 MAPK and c-Jun-N-terminal kinase (JNK) [29e31]. TGF-b2 isoforms are expressed by eosinophils and are the predominant form in severe allergic asthma where it promotes profibrotic responses affecting airway remodeling in addition to regulating mucin production and impacting on non-asthma related atopy in children [32e35]. Interestingly, TGF-b3 is implicated in the development of a severe phenotype of chronic airway remodeling induced by house dust mite (HDM) [35]. In this setting, increased mucus production, collagen deposition, and a dysregulated cytokine environment are observed. Our understanding of isoform specific effects of TGF-b in the context of asthma is only in its infancy and will need to be considered in future treatment studies. While TGF-ß is secreted by almost all immune cells including fibroblasts, endothelial cells and vascular/ASM cells [36e39], airway epithelia remain the major site of TGF-ß1 expression. In asthmatic airways however, TGF-ß expression is markedly increased and further augmented by the infiltrative inflammatory groups of cells that serve as an additional reservoir. An example of this is active eosinophil recruitment by chemokine ligand 5 (CCL-5) and CCL-11 that account for 80% of TGF-b expression in asthma [40e42]. A reduced deposition of the TGF-ß binding proteoglycan decorin within the asthmatic airway wall further increases its bioavailability in this context [16]. Owing to its abundance in the asthmatic airway and its complex but well understood signaling mechanisms, it appears logical that therapies directed at TGF-ß1 be considered in the context of severe refractory asthma.
TGF-ß airway remodeling Airway remodeling is the pathophysiologic modification of normal airway wall structure comprising a complex reorganization of the wall’s molecular and cellular constituents [43]. Including a repair process in response to airway wall injury, the dysregulated ensuing inflammation leads to structural change that results in loss of epithelial integrity, basement membrane thickening, subepithelial fibrosis, mucus gland and goblet-cell hyperplasia, smooth muscle hypertrophy and increased airway vascularity [44e46]. Fibrosis is the end-point of such a process within the ECM that also affects other organs including the liver and kidney. During its induction, TGF-ß1 promotes target genes
M. Al-Alawi et al. including connective tissue growth factor (CTGF), a-smooth muscle actin, collagen and plasminogen activator inhibitor [43,47]. In mild-to-moderate asthma, such modifications are partially reversible, however in severe chronic asthma, this becomes irreversible. Studies show that airway remodeling is primarily responsible for symptoms associated with decreased pulmonary function [48]. In the context of asthma, TGF-ß induce both anti- and pro-apoptotic effects in airway epithelial cells [47]. Antiapoptotic effects are mediated through the SMAD 2/3 pathway and occur in the absence of chemical or physical stress. Chronic allergenic exposure allows TGF-ß1 to induce apoptosis through activation of MAPK-signaling [49]. Injury to surface epithelia follow aggravated by a dysregulated repair process causing detachment of these cells and epithelialemesenchymal transition (EMT) [50]. TGF-ß1 also affects sub-epithelial fibrosis by increasing deposition of ECM such as type I and III collagen, fibronectin and proteoglycans. By inducing fibroblastic differentiation to myofibroblasts, increased proliferation follows [51,52]. Fibroblasts maintain crucial roles during the pulmonary fibrotic response which are dependent on activation of TGF-ß1 via ECM receptor integrins [53,54]. Additionally, TGF-ß1 induces myofibroblast differentiation and collagen production in nasal polyposis re-iterating its important role in airway disease [55]. Matrix metalloproteinases (MMPs) are key enzymes responsible for ECM remodeling, cell migration, and airway epithelial repair [56]. Cytokines including TGF-b1 play pivotal roles in MMP-9 production and secretion [57]. The quantity of submucosal neutrophils and macrophages, but not eosinophils, is significantly higher in asthmatics and in addition MMP-9 staining of the subepithelial basement membrane detected [58]. Neutrophils from allergic asthmatics produce and release MMP-9 upon direct allergen exposure [59]. In tandem with up-regulation of TGF-b1 gene expression by neutrophil elastase (NE) [39], activated neutrophils release reactive oxygen species (ROS) that exacerbate the airway remodeling process by promoting goblet cell hyperplasia [60]. As described previously, TGF-ß is rendered latent, by binding to LAP, which is in turn is covalently bound to a fibrillin protein LTBP. Inflammatory ECM degradation releases latent TGF-beLAP complexes that dissociate and increase active TGF-b bioavailability within the airway [61]. LAP complexes also act as substrates for MMP-2, 9, 13 and 14 [61]. MMPs alter transcription of ECM proteins responsible for collagen fibril laydown [62,63], and function as proteases that lyse ECM components with release of bioactive byproducts. In turn pro-inflammatory IL-13 stimulates collagen type I production by airway fibroblasts in an MMP and TGF-b1 dependent manner in asthma [64]. TGF-b also stimulates IL-6 which acts on ASM cells to induce release of eotaxin and VEGF concurrently mediating smooth muscle driven mast cell activation [65e67]. Despite our depth of knowledge of epithelial repair mechanisms, key questions relating to how activated cells organize into a matrix that functions as a regulator of the repair process still remain unanswered [56]. By far, the most crucial advance of our understanding of TGF-ß1 function within the airway is its regulatory role in EMT. Pulmonary alveolar epithelium, primary human
Transforming growth factor beta in severe asthma bronchial epithelia and airway epithelial cells isolated from asthmatic lungs have all demonstrated the ability to undergo EMT upon TGF-ß1 stimulation. This process can be inhibited with use of TGF-ß1 inhibitors such as Smad 7 [68,69]. In addition, TGF-ß1 influences EMT degradation by delicately balancing MMP and tissue-inhibitor of metalloproteinase (TIMP) activity during airway remodeling [57,70]. Sub-epithelial basement membrane (BM) thickness correlates with asthma severity [71,72]. Severe asthma is characterized by an increased ASM mass extending from the trachea to the smallest bronchioles and alveolar ducts [73e76]. Increases in ASM mass are a key feature of the airway remodeling process that includes MAPK mediated thickening of the BM. It is predominantly influenced by migration of MMPs toward the epithelium to form new matrix bundles [77]. TGF-ß mediated proliferation of collagen, fibronectin, serum and platelet-derived growth factor all play a role and require signaling through RGD (Arg-Gly-Asp) binding integrins [78]. This leads to stiffer airways and greater fixed airflow obstruction characteristically seen in severe asthma [79].
TGF-ß1 immunosupressive effects and regulatory inflammatory tolerance In its role as a pleiotropic and multifunctional growth factor, TGF-ß1 is a master regulator of immune responses resulting in fibrosis. As a potent anti-inflammatory, its chemo-attractive properties cause accumulation of macrophages and granulocytes in local inflammatory sites [80]. Additionally, it promotes development and differentiation of Th17 and forkheard box P3 (FoxP3) regulatory T-cells while inducing IL-9-producing T-helper cells [24,81,82]. Regulatory T cells (Treg) are a subpopulation of CD4þ T cells that maintain immunological self-tolerance and control the development of autoimmune disease [83]. Treg cells express the high affinity interleukin-2 (IL-2) receptor (CD25) and the transcription factor, Foxp3 that play critical roles in the function and development of Tregs [84]. Two subtypes of Foxp3þ CD25þ CD4þ Tregs are described: i) natural Treg (nTreg) from the thymus and ii) acquired/ induced Treg (aTreg/iTreg) that develop peripherally in response to TGF-b stimulation [84]. Foxp3þ CD25þ CD4þ Tregs regulate allergy, auto-immunity, transplant rejection, tumor immunity, and microbial responses [83]. Immunosuppressive Treg effects are mediated by IL-2R (CD25) and indirectly through antigen presenting cells (APCs) which in turn are mediated by cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) [85,86]. Although Foxp3þ CD4þ Tregs have suppressive capabilities, CD4 Foxp3 expression alone does not always indicate suppressive consequences. Transient Foxp3 expression in subsets of TGF-b-induced iTregs do not functionally equate to nTreg [87,88]. Emerging evidence provide insight into the regulatory behavior of Tregs in the context of immune function, stimulators in certain conditions whilst suppressive in others largely dependent on the context, location, conditions and target cells in question [89,90]. TGF-b suppresses immune responses through inhibition of inflammatory cell function and promotion of Treg
1413 production. TGF-b functions as an inducer for Treg generation [91]. It converts naive CD25 T cells to regulatory CD25 þ cells, and additionally regulates Treg suppressive function [92e94]. Antigens may induce TGF-beproducing Treg (Th3), which in turn mediates immunosuppression [95]. Blocking TGF-b in vivo abolishes tolerance and impairs CD25þFoxp3þ Treg induction; however the precise role of TGF-b in inducing mucosal Th3 cells is unclear. Evidence underpinning molecular mechanisms that drive Treg proliferation, activation, survival and targeting will in the future likely provide potential therapeutic targets in severe asthma. By regulation of the homeostasis of lymphocytes and Tregs, which in turn inhibit Th1 and Th2 responses, inflammation can be restricted. TGF-ß1 prevents housedust mite-induced allergic reactions in murine lungs by generating CD4þ and CD25þ Treg cells [96]. Administration of CD4þ and CD25þ Treg cells in an experimental setting reduces established lung eosinophilia, Th2 infiltration and promotes expression of IL-5, IL-13 and TGF-ß1 in airway specimens [97]. TGF-ß1 directly delivered into the trachea suppresses allergen-induced inflammation suggestive of a potential therapeutic role [98]. The evidence underlying the molecular mechanisms driving immunosuppressive Treg cell proliferation, activation, survival and subsequent targeting however remain to be elucidated.
Pro-inflammatory role of TGF-ß1 TGF-ß1 also plays a key role in inflammation acting as a potent chemotactic factor and activator for a variety of inflammatory cells. Both inflammatory and structural cells produce TGF-ß1 contributing to the increased levels observed in BAL fluid obtained from asthmatic airways [20,99,100]. Increased TGF-ß1 is associated with higher macrophage concentrations following allergen challenge and significantly amplified inflammation through induction of Th17 cells [42,80,101]. TGF-ß1 airway concentrations have been associated with airflow limitation in a cross-sectional sample of children with severe asthma [102]. Increased expression of airway TGF-ß1 is coupled with an increase in BAL macrophages and concentrations of lipid peroxidation biomarkers 8isoprostanes and malondialdehyde. Oxidative stress may thus mediate TGF-b1 effects and promote airway remodeling in children with severe asthma. Nuclear factor-like 2 (Nrf2) is another factor responsible for regulating glutathione, antioxidant responses and reducing ASM proliferation in severe asthma [103]. This pathway is also disrupted in severe asthma due to chronic oxidative stress [104]. TGFß also plays an important role here, suppressing Nrf2 activity in ASM cells and consequently promoting a dysregulated antioxidant response [103]. What remains lacking is a definitive link between elevated airway TGF-ß1 and progressive loss of pulmonary function from an oxidative perspective. Increased expression and activation of airway TGF-ß1 coupled with an increased airway oxidant burden underlies the physiological alterations accompanying severe asthma in children. Traffic-related air pollutants and cigarette smoke cause oxidant mediated airway inflammation and
1414 are also associated with asthma in children [105,106]. Children with the TGF-b 509 TT genotype are at higher risk of asthma occurrence if they live near a freeway or were exposed to tobacco smoke in utero [107]. In an experimental model of rat tracheal explants, two-to threefold increases in TGF-b1 expression was observed when the explants were exposed to urban air particles [108] and cigarette smoke [109]. Cigarette smoke exposure is known to enhance antigen-induced mast cell activation via TGF-b/ Smad signaling pathways in a mouse model of allergic asthma [110]. Kang et al. (2007) demonstrated that cigarette smoking is a potent stimulator of IL-18 release from pulmonary macrophages [111]. In turn, activated macrophages and airway epithelial cells release IL-18 that contributes to the proinflammatory cascade in asthma [112]. For instance, it enhances remodeling through production of IFN-g, IL-13 and TGF-b1 in an asthma mouse model [113]. In further animal work, chronic exposure to house dust mite demonstrated elevated IL-4, IL-13 and TGF-ß1 accompanied by goblet cell hyperplasia, subepithelial fibrosis and increased ASM surface area [114]. Taken together this current evidence suggests that inflammatory and allergic mechanisms both influence the TGF-ß pathway, and insight into the effect of cigarette smoking on the induction of TGF-b1 release from other cells remains to be explored. Current guidelines for randomized controlled trial recruitment include recruitment of patients “free” of comorbid disease that demonstrate good inhaler technique, and an asthma diagnosis. Cigarette smoking for example typically results in patients being excluded from consideration. Consequently, there is exclusion of current and exsmokers in addition to patients with at least a ten-pack year smoking history. Given the foundation of knowledge on the effects of cigarette smoking on murine models of asthma [109,110], there is an inherent bias in current trial recruitment preventing the extrapolation of understanding in vivo TGF-b mediated effects in human asthmatic smokers [115]. Th2 cytokines IL-5 and IL-13 have been intimately linked to airway TGF-ß1. IL-5 receptor deficient mice have undetectable airway eosinophils and reduced TGF-ß1 concentrations concomitant with decreased fibrotic change in a chronic exposure model [116]. Conversely, IL-5 overexpression leads to increased eosinophils, TGF-ß, higher collagen content and more fibrotic consequences. Inhibiting TGF-ß1 signaling specifically in T-cell subsets has led to enhanced airway inflammation, hypersensitivity and increased Th2 cytokine production hence its potential therapeutic role has been controversial [117]. IL-13 induces TGF-ß1 expression through synergistic mechanisms on airway processes including remodeling and fibrosis [64,118]. IL-13 deficient mice have decreased signal transduction, activation of transcription-6 (STAT6) phosphorylation and limited TGF-ß1 activity following allergic stimulation resulting in attenuated bronchial hyperresponsiveness, ASM hyperplasia, and immunoglobulin-E (IgE) synthesis [34,119]. An increased quantity and size of goblet cells and mucosal glands are characteristic of asthma-related airway obstruction. TGF-ß2 importantly regulates the transcription and translation of mucin in bronchial epithelial cells [34] and anti-TGF-ß treatment in animal models of asthma illustrate a decreased goblet cell burden [120].
M. Al-Alawi et al. Expression of VEGF promotes angiogenesis and permeability. Changes to airway microvasculature accompany airway remodeling and result in VEGF release. TGF-ß1 promotes further microvascular congestion through upregulation of pro-angiogenic factors such as VEGF in the severe asthmatic airway [121]. Interestingly, VEGF inhibition has been shown to attenuate peribronchial fibrosis through effects on the phosphoinositide 3-kinase(PI3K)/Akt pathway [122].
TGF-ß and eosinophil crosstalk Asthma severity is related to a relationship between TGF-ß expression and the presence of submucosal eosinophils [40e42,123]. Minshall et al. examined bronchial biopsies of severe asthmatics and demonstrated that 65% of TGF-ß1 mRNA-positive cells were eosinophils and that 75% of eosinophils were positive for TGF-ß1 mRNA, findings reflective of earlier work performed by others [40,42,124]. The study interestingly also reported strong correlations between mRNA-positive TGF-ß1 cells beneath the basement membrane and degree of eosinophilia within the same compartment. The extent of TGF-ß1 expression is significantly higher in those with severe compared to mild asthma. TGF-ß is therefore upregulated in severe asthma and a strong correlation exists between TGF-ß1 expression, eosinophil burden and asthma severity. In the upper airway, eosinophils remain a major source of TGF-ß in patients with allergic disease. Linear relationships are evident between the degree of eosinophilia and TGF-ß1 expression in patients with rhinosinusitis [47]. Similar associations are observed with atopic dermatitis where Th2-predominant infiltration similar to that in asthma is the major driver of the inflammatory state [125]. Persistent eosinophilia relates to asthma disease severity and correlates with fixed airflow obstruction, frequent exacerbations, hospitalizations and increased intubation rates [15,126,127]. Targeting the eosinophil itself has been effective and studies of the anti-IL-5 agent, mepolizumab illustrates significant reduction in exacerbation rates in corticosteroid dependent patients. In addition, improved symptom control and quality of life is noted [128e130]. Flood-Page et al. attempted treatment of patients with mild atopic asthma with mepolizumab resulting in decreased tissue eosinophilia, lung TGF-ß1 expression, reduced remodeling and ECM deposition [41]. Such observations were further reinforced when IL-5 knockout mice chronically exposed to allergens showed reductions in the pulmonary expression of TGF-ß1 [131].
Genetic TGF-ß modifiers Gene polymorphisms in the TGF-ß1 promoter region are associated with the development of asthma [9,132]. Several single nucleotide polymorphisms (SNPs) are associated with peripheral blood eosinophilia in asthmatic patients [133]. Lerodiakonou et al. (2013) identified asthmatic carriers of the TGF-ß1 SNP rs6957-G to have significantly higher submucosal eosinophils and macrophages. In the same study, the SNPs rs1800469-T and rs1800470-C were associated with less FEV1 decline and rs4803455-A with accelerated FEV1
Transforming growth factor beta in severe asthma decline. Also of interest, three other SNPs that associated with increased airflow obstruction (rs6957, rs1800469, and rs1800470) were also linked to less rapid decline in nonsmokers. Such data provide evidence of critical interactions between TGF-ß1 SNPs and effects of smoking on asthma severity. Other disease associations link the polymorphisms with survival in non-small cell lung cancer [134], breast cancer [135], and risk of radiation pneumonitis [136]. A recent meta-analysis suggested that the 509/T polymorphism in TGF-ß1 gene as a risk factor for asthma susceptibility and phenotype [137,138]. Polymorphism T869C has also been directly associated to asthma severity [139e142]. Furthermore, Freimuth et al. importantly concluded that differential inheritance of genetic loci, TGFbm, alters biological responses to reduced TGF-ß1 signaling in an experimental asthma model [143]. TGF-ß antagonists for treatment of lung diseases might therefore give diverse outcomes among patient cohorts, dependent on genetic variation. Recent work has also shown that patients with TGF-ß receptor II mutations were strongly predisposed to the development of multiple immunologic phenotypes, including asthma, food allergies, eczema, allergic rhinitis, and eosinophilic gastrointestinal disease [144]. In LoeyseDeitz syndrome (LDS), a condition with naturally occurring mutations in this gene, almost half of all patients had been diagnosed with asthma and increased eosinophil counts, elevated IgE and increased Th2cytokines IL-5 and IL-13. T-cells sequestered from such patients showed increased Smad 2 and 3 phosphorylation in response to TGF-ß1 that in turn enhanced its signaling. Angiotensin-II receptor blockade (e.g. losartan) interestingly mitigates TGF-ß signaling in lymphocytes of LDS patients suggesting that this or other related treatment approaches hold promise in the prevention of major cardiovascular complications and perhaps the development of asthma if administered at an early stage [145,146]. Future work should re-visit the previously demonstrated interaction between blocking the renin-angiotensin system and effects on TGF-ß1 signaling in asthma.
TGF-ß targeted therapy Current therapeutic guidelines for the treatment of asthma are ineffective at targeting any reversal of the airway structural remodeling process. Corticosteroids however have been shown to exhibit an inhibitory effect on TGF-ß1 expression in a murine model of asthma hence illustrating some albeit minimal potential in affecting this irreversible process [147]. In humans however, a two-week course of oral corticosteroids did not inhibit TGF-ß production and was deemed ineffective at modulating the fibrotic process [101]. Although these findings validate other studies demonstrating minimal effects of corticosteroids on the airway remodeling process, recent evidence identifies novel mechanistic effects of glucocorticoids on TGF-b signaling cross talk [148]. Glucocorticoids recruited the accessory TGF-b receptor, Tgfbr3, and Smad-1 to shift TGFb signaling from the Tgfbr1/Smad2/3 axis to an Acvrl1/ Smad1 axis in lung fibroblasts. Such findings illustrate the importance of glucocorticoids on the array of TGF-b mediated responses in inflammatory airway diseases.
1415 In-vitro and -vivo evidence suggests a role for activin A, a promising target in regulating the inflammatory fibrotic remodeling process within the airway [149,150]. It is a member of the TGF-b superfamily through which inhibition, by follistatin, decreases mucus hypersecretion, subepithelial collagen deposition and thickening of the subepithelial smooth muscle, indicating a potential therapeutic role in the prevention of airway remodeling [151]. Tiotropium bromide reduced levels of TGF-ß1 in BAL of OVA-sensitized mice. In addition, it abrogated airway hyper-responsiveness to serotonin and inhibited Th2 cytokine production [152]. Such inhibitory effects on acute versus chronic asthma are dependent on differential expression of muscarinic receptor subtypes [153]. Evidence by Schaafsma et al. highlights that simvastatin may inhibit TGF-b1-induced fibronectin expression in airway fibroblasts [154] which in turn attenuates fibroblast-to-myofibroblast transition in asthmatic settings [155]. Targeting TGF-ß represents a major challenge as a target for severe asthma therapy. The complexity of its signaling network has thus far hindered the pursuit of specific drug candidates targeting TGF-ß. Neutralizing antibodies that block TGF-ß activity through phosho-Smad 2 appear at least in preliminary animal work to prevent lung fibrosis in asthma [156,157]. SD-208 has been trialed as a TGF-bRI kinase inhibitor in a chronic rat allergic asthma model [158]. Effects include inhibiting all TGF-b isoforms, bronchial and ASM cell proliferation and goblet cell hyperplasia all induced by exposure to allergens. The effects of TGF-ß1 on airway remodeling are variable and largely depend on the local environment and model used for investigation [47]. The therapeutic treatment of mice with anti-TGF-ß antibodies significantly reduced pulmonary peri-bronchiolar ECM deposition, ASM proliferation, and mucus production without affecting established airway inflammation and Th2 cytokine production. These data suggest therefore that inflammation and remodeling may be uncoupled during a prolonged allergic challenge [120]. Chronic exposure to HDM demonstrated elevated IL-4, IL-13 and TGF-ß1 levels accompanied by goblet cell hyperplasia, subepithelial fibrosis and increased ASM surface area [114]. Difficulties however arise as contrasting evidence has shown that anti-TGF-b treatment in the context of continuous or intermittent HDM exposure had no effect on the development of HDM-induced airway remodeling. TGF-ß neutralization in fact exacerbated eosinophilic infiltrates and led to increased airway hyper-responsiveness in some cases [159]. These findings may in part be explained by the differential expression of TGF-b isoforms in response to duration of HDM exposure [35]. Taken together, this work suggests that we remain unclear as to the true benefits of targeting the TGF-b pathway and need to be fairly specific during the process to ensure beneficial as opposed to adverse effects. Anti-TGF-ß1 antibodies have been demonstrated to inhibit monocyte/macrophage recruitment and reduce eosinophil and lymphocyte numbers within the airway [19]. Treatment with mepacrine, a synthetic anti-malarial drug reduces TGF-ß1 and subsequently sub-epithelial airway fibrosis [160]. The cytokine IFN-g has been shown to downregulate TGF-ß2 through Smad-7, however their therapeutic consequences in the asthmatic airway remain undetermined [161].
1416
M. Al-Alawi et al.
Figure 2 Multifunctional role of TGF-b in severe asthma. TGF, produced mainly by eosinophils in the asthmatic airway exerts multiple effects that influence inflammation and airway remodeling. (EMT: Epithelial-mesenchymal transition, ECM: Extracellular matrix, MAPK:p38-mitogen-activated protein kinase, Nrf2: Nuclear factor-like2, Th1/2: T helper cells 1/2, Treg: Regulatory T cells, IL: Interleukin). (Blue arrow indicates activation, red line indicates inhibition). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
An integrated approach to target factors regulating inflammation, cell migration, and tissue remodeling in asthmatic airways has important pathophysiological and clinical consequences that must be recognized in the context of emerging asthma therapeutics that target Th2driven inflammation [162]. Use of antisense oligonucleotide therapy against TGF-ß has been applied in animal models in an attempt to reduce postoperative scarring in ocular glaucoma [163]. Similarly, adenoviral delivery to overexpress Smad-2 within the lung results surprisingly in an increased sub-epithelial collagen deposition and ASM hyperplasia. These effects lead to thickening of the ASM layer following HDM challenge [164] (Fig. 2). In view of contrasting outcomes to anti-TGF-b approaches particularly in the context of allergy and asthma, future work focused on the complex signaling network involved will be necessary before we can progress to valid clinical study.
Conclusion and future directions Suboptimal patient selection for certain asthmatic clinical studies may at least partially account for the contrasting pool of evidence with regards differences in TGF-ß1 immunohistochemical staining between asthmatics and healthy controls [32,165,166]. This is further complicated by our emerging understanding of the various clinical phenotypes within the asthma spectrum. Airway TGF-ß1 release is inducible and transient however does require an inflammatory or antigenic challenge [47]. Effective
modulation of TGF-ß is therefore dependent on proliferative factors that induce its transient release in asthma subsequently regulating ASM thickness [167e169]. Contrasting effects of TGF-b may be due to differences in cell origin (airway versus alveolar, transformed versus primary and human versus rodent), differences in matrix or integrin expression, or concentration-dependent responses [56]. Latent TGF-ß activation and isoform differentiation further complicates the asthmatic setting and adds to the complexity of its role in the diseased airway. Stored in large quantities within the airway ECM, it plays major roles in regulating inflammation and its consequences. While down regulating the inflammatory process through the induction of immunosuppressive Tregs, it also initiates structural remodeling. A delicate balance between such phenomena defines the precise role for TGF-ß in the clinicopathophysiologic phenotypes observed in severe asthma. TGF-ß modulation is potentially useful in immunosuppression while pure global inhibition may have anti-fibrotic effects. Development of targeted TGF-ß therapy will likely need to be individualized specifically to the phenotypic spectrum of patients with severe asthma where immunosuppression may be desirable if it is to become a successful component in future asthma care.
Conflicts of interest None of the authors have any conflicts of interest to disclose with respect to this manuscript.
Transforming growth factor beta in severe asthma
References [1] Kudo M, Ishigatsubo Y, Aoki I. Pathology of asthma. Front Microbiol 2013;4(263) [PubMed PMID: 24032029. Pubmed Central PMCID: 3768124. Epub 2013/09/14]. [2] Holgate ST, Davies DE, Puddicombe S, Richter A, Lackie P, Lordan J, et al. Mechanisms of airway epithelial damage: epithelial-mesenchymal interactions in the pathogenesis of asthma. Eur Respir J Suppl 2003 Sep;44:24se9s [PubMed PMID: 14582897. Epub 2003/10/30]. [3] Bonfield TL, Ross KR. Asthma heterogeneity and therapeutic options from the clinic to the bench. Curr Opin Allergy Clin Immunol 2012 Feb;12(1):60e7 [PubMed PMID: 22193053. Epub 2011/12/24. eng]. [4] Wener RR, Bel EH. Severe refractory asthma: an update. Eur Respir Rev 2013 Sep 1;22(129):227e35 [PubMed PMID: 23997049. Epub 2013/09/03. eng]. [5] Moore WC, Meyers DA, Wenzel SE, Teague WG, Li H, Li X, et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med 2010 Feb 15;181(4):315e23 [PubMed PMID: 19892860. Epub 2009/11/07. eng]. [6] Mauad T, Bel EH, Sterk PJ. Asthma therapy and airway remodeling. J Allergy Clin Immunol 2007 Nov;120(5): 997e1009 [quiz 10-1. PubMed PMID: 17681364. Epub 2007/08/08]. [7] Pascual RM, Peters SP. Airway remodeling contributes to the progressive loss of lung function in asthma: an overview. J Allergy Clin Immunol 2005 Sep;116(3):477e86 [quiz 87. PubMed PMID: 16159612. Epub 2005/09/15]. [8] Broide DH. Immunologic and inflammatory mechanisms that drive asthma progression to remodeling. J Allergy Clin Immunol 2008 Mar;121(3):560e70 [quiz 71-2. PubMed PMID: 18328887. Pubmed Central PMCID: 2386668. Epub 2008/03/11]. [9] Budd DC, Holmes AM. Targeting TGFbeta superfamily ligand accessory proteins as novel therapeutics for chronic lung disorders. Pharmacol Ther 2012 Sep;135(3):279e91 [PubMed PMID: 22722064. Epub 2012/06/23. eng]. [10] Brightling CE, Gupta S, Gonem S, Siddiqui S. Lung damage and airway remodelling in severe asthma. Clin Exp Allergy 2012 May;42(5):638e49 [PubMed PMID: 22192725. Epub 2011/12/24. eng]. [11] Hoshino M, Nakamura Y, Sim JJ. Expression of growth factors and remodelling of the airway wall in bronchial asthma. Thorax 1998 Jan;53(1):21e7 [PubMed PMID: 9577517. Epub 1998/05/13. eng]. [12] Haldar P, Pavord ID, Shaw DE, Berry MA, Thomas M, Brightling CE, et al. Cluster analysis and clinical asthma phenotypes. Am J Respir Crit Care Med 2008 Aug 1;178(3): 218e24 [PubMed PMID: 18480428. Epub 2008/05/16. eng]. [13] Mascia K, Haselkorn T, Deniz YM, Miller DP, Bleecker ER, Borish L, et al. Aspirin sensitivity and severity of asthma: evidence for irreversible airway obstruction in patients with severe or difficult-to-treat asthma. J Allergy Clin Immunol 2005 Nov;116(5):970e5 [PubMed PMID: 16275362. Epub 2005/11/09. eng]. [14] ten Brinke A, Grootendorst DC, Schmidt JT, De Bruine FT, van Buchem MA, Sterk PJ, et al. Chronic sinusitis in severe asthma is related to sputum eosinophilia. J Allergy Clin Immunol 2002 Apr;109(4):621e6 [PubMed PMID: 11941310. Epub 2002/04/10. eng]. [15] Sakagami T, Hasegawa T, Koya T, Furukawa T, Kawakami H, Kimura Y, et al. Cluster analysis identifies characteristic phenotypes of asthma with accelerated lung function decline. J Asthma 2014;51(2):113e8 [PubMed PMID: 24102534. Epub 2013/10/10. Eng].
1417 [16] Wenzel S, Ford L, Pearlman D, Spector S, Sher L, Skobieranda F, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med 2013 Jun 27;368(26): 2455e66 [PubMed PMID: 23688323. Epub 2013/05/22. eng]. [17] Ivanova JI, Bergman R, Birnbaum HG, Colice GL, Silverman RA, McLaurin K. Effect of asthma exacerbations on health care costs among asthmatic patients with moderate and severe persistent asthma. J Allergy Clin Immunol 2012 May;129(5):1229e35 [PubMed PMID: 22326484. Epub 2012/02/14. eng]. [18] Shi M, Zhu J, Wang R, Chen X, Mi L, Walz T, et al. Latent TGFbeta structure and activation. Nature 2011 Jun 16;474(7351): 343e9 [PubMed PMID: 21677751. Epub 2011/06/17. eng]. [19] Bottoms SE, Howell JE, Reinhardt AK, Evans IC, McAnulty RJ. Tgf-Beta isoform specific regulation of airway inflammation and remodelling in a murine model of asthma. PLoS One 2010;5(3):e9674 [PubMed PMID: 20300191. Epub 2010/03/20. eng]. [20] Howell JE, McAnulty RJ. TGF-beta: its role in asthma and therapeutic potential. Curr Drug Targets 2006 May;7(5): 547e65 [PubMed PMID: 16719766. Epub 2006/05/25. eng]. [21] Fjellbirkeland L, Cambier S, Broaddus VC, Hill A, Brunetta P, Dolganov G, et al. Integrin alphavbeta8-mediated activation of transforming growth factor-beta inhibits human airway epithelial proliferation in intact bronchial tissue. Am J Pathol 2003 Aug;163(2):533e42 [PubMed PMID: 12875973. Pubmed Central PMCID: 1868219. Epub 2003/07/24]. [22] Nishimura SL. Integrin-mediated transforming growth factorbeta activation, a potential therapeutic target in fibrogenic disorders. Am J Pathol 2009 Oct;175(4):1362e70 [PubMed PMID: 19729474. Pubmed Central PMCID: 2751532. Epub 2009/09/05]. [23] Giacomini MM, Travis MA, Kudo M, Sheppard D. Epithelial cells utilize cortical actin/myosin to activate latent TGFbeta through integrin alpha(v)beta(6)-dependent physical force. Exp Cell Res 2012 Apr 1;318(6):716e22 [PubMed PMID: 22309779. Epub 2012/02/09. eng]. [24] Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-beta regulation of immune responses. Annu Rev Immunol 2006;24:99e146 [PubMed PMID: 16551245. Epub 2006/03/23. eng]. [25] Hyytiainen M, Penttinen C, Keski-Oja J. Latent TGF-beta binding proteins: extracellular matrix association and roles in TGF-beta activation. Crit Rev Clin Lab Sci 2004; 41(3):233e64 [PubMed PMID: 15307633. Epub 2004/08/17. eng]. [26] Malhotra N, Kang J. SMAD regulatory networks construct a balanced immune system. Immunology 2013 May;139(1): 1e10 [PubMed PMID: 23347175. Epub 2013/01/26. eng]. [27] Sagara H, Okada T, Okumura K, Ogawa H, Ra C, Fukuda T, et al. Activation of TGF-beta/Smad2 signaling is associated with airway remodeling in asthma. J Allergy Clin Immunol 2002 Aug;110(2):249e54 [PubMed PMID: 12170265. Epub 2002/08/10. eng]. [28] Le AV, Cho JY, Miller M, McElwain S, Golgotiu K, Broide DH. Inhibition of allergen-induced airway remodeling in Smad 3deficient mice. J Immunol 2007 Jun 1;178(11):7310e6 [PubMed PMID: 17513781. Epub 2007/05/22. eng]. [29] Yeganeh B, Mukherjee S, Moir LM, Kumawat K, Kashani HH, Bagchi RA, et al. Novel non-canonical TGF-beta signaling networks: emerging roles in airway smooth muscle phenotype and function. Pulm Pharmacol Ther 2013 Feb;26(1): 50e63 [PubMed PMID: 22874922. Epub 2012/08/10. eng]. [30] Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res 2009 Jan;19(1):128e39 [PubMed PMID: 19114990. Epub 2008/12/31. eng]. [31] Wiehler S, Cuvelier SL, Chakrabarti S, Patel KD. p38 MAP kinase regulates rapid matrix metalloproteinase-9 release
1418
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
M. Al-Alawi et al. from eosinophils. Biochem Biophys Res Commun 2004 Mar 5; 315(2):463e70 [PubMed PMID: 14766231. Epub 2004/02/10]. Balzar S, Chu HW, Silkoff P, Cundall M, Trudeau JB, Strand M, et al. Increased TGF-beta2 in severe asthma with eosinophilia. J Allergy Clin Immunol 2005 Jan;115(1):110e7 [PubMed PMID: 15637555. Epub 2005/01/08. eng]. Torrego A, Hew M, Oates T, Sukkar M, Fan Chung K. Expression and activation of TGF-beta isoforms in acute allergeninduced remodelling in asthma. Thorax 2007 Apr;62(4): 307e13 [PubMed PMID: 17251317. Pubmed Central PMCID: 1892798. Epub 2007/01/26]. Harrop CA, Gore RB, Evans CM, Thornton DJ, Herrick SE. TGFbeta(2) decreases baseline and IL-13-stimulated mucin production by primary human bronchial epithelial cells. Exp Lung Res 2013 Feb;39(1):39e47 [PubMed PMID: 23249391. Epub 2012/12/20]. Li G, Fox 3rd J, Liu Z, Liu J, Gao GF, Jin Y, et al. Lyn mitigates mouse airway remodeling by downregulating the TGF-beta3 isoform in house dust mite models. J Immunol 2013 Dec 1; 191(11):5359e70 [PubMed PMID: 24127553. Pubmed Central PMCID: 3863691. Epub 2013/10/16]. Kelley J, Kovacs EJ, Nicholson K, Fabisiak JP. Transforming growth factor-beta production by lung macrophages and fibroblasts. Chest 1991 Mar;99(Suppl. 3):85Se6S [PubMed PMID: 1705204. Epub 1991/03/01. eng]. Coker RK, Laurent GJ, Shahzeidi S, Hernandez-Rodriguez NA, Pantelidis P, du Bois RM, et al. Diverse cellular TGF-beta 1 and TGF-beta 3 gene expression in normal human and murine lung. Eur Respir J 1996 Dec;9(12):2501e7 [PubMed PMID: 8980960. Epub 1996/12/01. eng]. de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J, Hiemstra PS, et al. Transforming growth factor beta1 and recruitment of macrophages and mast cells in airways in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 1998 Dec;158(6):1951e7 [PubMed PMID: 9847291. Epub 1998/12/16. eng]. Lee KY, Ho SC, Lin HC, Lin SM, Liu CY, Huang CD, et al. Neutrophil-derived elastase induces TGF-beta1 secretion in human airway smooth muscle via NF-kappaB pathway. Am J Respir Cell Mol Biol 2006 Oct;35(4):407e14 [PubMed PMID: 16690986. Epub 2006/05/13. eng]. Minshall EM, Leung DY, Martin RJ, Song YL, Cameron L, Ernst P, et al. Eosinophil-associated TGF-beta1 mRNA expression and airways fibrosis in bronchial asthma. Am J Respir Cell Mol Biol 1997 Sep;17(3):326e33 [PubMed PMID: 9308919. Epub 1997/10/06. eng]. Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig MS, et al. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest 2003 Oct;112(7):1029e36 [PubMed PMID: 14523040. Epub 2003/10/03. eng]. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo A, et al. Transforming growth factor-beta expression in mucosal biopsies in asthma and chronic bronchitis. Am J Respir Crit Care Med 1997 Aug;156(2 Pt 1):591e9 [PubMed PMID: 9279245. Epub 1997/08/01. eng]. Postma DS, Timens W. Remodeling in asthma and chronic obstructive pulmonary disease. Proc Am Thorac Soc 2006 Jul; 3(5):434e9 [PubMed PMID: 16799088. Epub 2006/06/27. eng]. Naylor B. The shedding of the mucosa of the bronchial tree in asthma. Thorax 1962 Mar;17:69e72 [PubMed PMID: 14478653. Epub 1962/03/01. eng]. Roche WR, Beasley R, Williams JH, Holgate ST. Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1989 Mar 11; 1(8637):520e4 [PubMed PMID: 2466184. Epub 1989/03/11. eng].
[46] Aikawa T, Shimura S, Sasaki H, Ebina M, Takishima T. Marked goblet cell hyperplasia with mucus accumulation in the airways of patients who died of severe acute asthma attack. Chest 1992 Apr;101(4):916e21 [PubMed PMID: 1555462. Epub 1992/04/01. eng]. [47] Halwani R, Al-Muhsen S, Al-Jahdali H, Hamid Q. Role of transforming growth factor-beta in airway remodeling in asthma. Am J Respir Cell Mol Biol 2011 Feb;44(2):127e33 [PubMed PMID: 20525803. Epub 2010/06/08. eng]. [48] Lazaar AL, Panettieri Jr RA. Is airway remodeling clinically relevant in asthma? Am J Med 2003 Dec 1;115(8):652e9 [PubMed PMID: 14656618. Epub 2003/12/06. eng]. [49] Undevia NS, Dorscheid DR, Marroquin BA, Gugliotta WL, Tse R, White SR. Smad and p38-MAPK signaling mediates apoptotic effects of transforming growth factor-beta1 in human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2004 Sep;287(3):L515e24 [PubMed PMID: 15132952. Epub 2004/05/11. eng]. [50] Davies DE. The role of the epithelium in airway remodeling in asthma. Proc Am Thorac Soc 2009 Dec;6(8):678e82 [PubMed PMID: 20008875. Epub 2009/12/17. eng]. [51] Michalik M, Pierzchalska M, Legutko A, Ura M, Ostaszewska A, Soja J, et al. Asthmatic bronchial fibroblasts demonstrate enhanced potential to differentiate into myofibroblasts in culture. Med Sci Monit 2009 Jul;15(7):BR194e201 [PubMed PMID: 19564819. Epub 2009/07/01. eng]. [52] Khalil N, Xu YD, O’Connor R, Duronio V. Proliferation of pulmonary interstitial fibroblasts is mediated by transforming growth factor-beta1-induced release of extracellular fibroblast growth factor-2 and phosphorylation of p38 MAPK and JNK. J Biol Chem 2005 Dec 30; 280(52):43000e9 [PubMed PMID: 16246848. Epub 2005/10/26. eng]. [53] Kitamura H, Cambier S, Somanath S, Barker T, Minagawa S, Markovics J, et al. Mouse and human lung fibroblasts regulate dendritic cell trafficking, airway inflammation, and fibrosis through integrin alphavbeta8-mediated activation of TGFbeta. J Clin Invest 2011 Jul;121(7):2863e75 [PubMed PMID: 21646718. Epub 2011/06/08. eng]. [54] Tatler AL, John AE, Jolly L, Habgood A, Porte J, Brightling C, et al. Integrin alphavbeta5-mediated TGF-beta activation by airway smooth muscle cells in asthma. J Immunol 2011 Dec 1; 187(11):6094e107 [PubMed PMID: 22025551. Pubmed Central PMCID: 3242066. Epub 2011/10/26]. [55] Semlali A, Jacques E, Rouabhia M, Milot J, Laviolette M, Chakir J. Regulation of epithelial cell proliferation by bronchial fibroblasts obtained from mild asthmatic subjects. Allergy 2010 Nov;65(11):1438e45 [PubMed PMID: 20456314. Epub 2010/05/12. eng]. [56] Crosby LM, Waters CM. Epithelial repair mechanisms in the lung. Am J Physiol Lung Cell Mol Physiol 2010 Jun;298(6): L715e31 [PubMed PMID: 20363851. Pubmed Central PMCID: 2886606. Epub 2010/04/07]. [57] Ohbayashi H, Shimokata K. Matrix metalloproteinase-9 and airway remodeling in asthma. Curr Drug Targets Inflamm Allergy 2005 Apr;4(2):177e81 [PubMed PMID: 15853739. Epub 2005/04/28. eng]. [58] Wenzel SE, Balzar S, Cundall M, Chu HW. Subepithelial basement membrane immunoreactivity for matrix metalloproteinase 9: association with asthma severity, neutrophilic inflammation, and wound repair. J Allergy Clin Immunol 2003 Jun;111(6):1345e52 [PubMed PMID: 12789238. Epub 2003/06/06. eng]. [59] Ventura I, Vega A, Chacon P, Chamorro C, Aroca R, Gomez E, et al. Neutrophils from allergic asthmatic patients produce and release metalloproteinase-9 upon direct exposure to allergens. Allergy 2014 Jul;69(7):898e905 [PubMed PMID: 24773508. Epub 2014/04/30].
Transforming growth factor beta in severe asthma [60] Mazaki Y, Hashimoto S, Tsujimura T, Morishige M, Hashimoto A, Aritake K, et al. Neutrophil direction sensing and superoxide production linked by the GTPase-activating protein GIT2. Nat Immunol 2006 Jul;7(7):724e31 [PubMed PMID: 16715100. Epub 2006/05/23]. [61] Mott JD, Werb Z. Regulation of matrix biology by matrix metalloproteinases. Curr Opin Cell Biol 2004 Oct;16(5): 558e64 [PubMed PMID: 15363807. Pubmed Central PMCID: 2775446. Epub 2004/09/15]. [62] Vincenti MP, Brinckerhoff CE. Transcriptional regulation of collagenase (MMP-1, MMP-13) genes in arthritis: integration of complex signaling pathways for the recruitment of genespecific transcription factors. Arthritis Res 2002;4(3): 157e64 [PubMed PMID: 12010565. Pubmed Central PMCID: 128926. Epub 2002/05/16]. [63] Kassim SY, Gharib SA, Mecham BH, Birkland TP, Parks WC, McGuire JK. Individual matrix metalloproteinases control distinct transcriptional responses in airway epithelial cells infected with Pseudomonas aeruginosa. Infect Immun 2007 Dec;75(12):5640e50 [PubMed PMID: 17923522. Pubmed Central PMCID: 2168342. Epub 2007/10/10]. [64] Firszt R, Francisco D, Church TD, Thomas JM, Ingram JL, Kraft M. Interleukin-13 induces collagen type-1 expression through matrix metalloproteinase-2 and transforming growth factor-beta1 in airway fibroblasts in asthma. Eur Respir J 2014 Feb;43(2):464e73 [PubMed PMID: 23682108. Epub 2013/05/18]. [65] Ammit AJ, Moir LM, Oliver BG, Hughes JM, Alkhouri H, Ge Q, et al. Effect of IL-6 trans-signaling on the pro-remodeling phenotype of airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2007 Jan;292(1):L199e206 [PubMed PMID: 16936245. Epub 2006/08/29]. [66] Hollins F, Kaur D, Yang W, Cruse G, Saunders R, Sutcliffe A, et al. Human airway smooth muscle promotes human lung mast cell survival, proliferation, and constitutive activation: cooperative roles for CADM1, stem cell factor, and IL-6. J Immunol 2008 Aug 15;181(4):2772e80 [PubMed PMID: 18684968. Pubmed Central PMCID: 3992374. Epub 2008/08/08]. [67] Michaeloudes C, Sukkar MB, Khorasani NM, Bhavsar PK, Chung KF. TGF-beta regulates Nox4, MnSOD and catalase expression, and IL-6 release in airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2011 Feb;300(2):L295e304 [PubMed PMID: 21131394. Pubmed Central PMCID: 3043811. Epub 2010/12/07]. [68] Camara J, Jarai G. Epithelial-mesenchymal transition in primary human bronchial epithelial cells is Smad-dependent and enhanced by fibronectin and TNF-alpha. Fibrogenes Tissue Repair 2010;3(1):2 [PubMed PMID: 20051102. Epub 2010/01/07. eng]. [69] Hackett TL, Warner SM, Stefanowicz D, Shaheen F, Pechkovsky DV, Murray LA, et al. Induction of epithelialmesenchymal transition in primary airway epithelial cells from patients with asthma by transforming growth factorbeta1. Am J Respir Crit Care Med 2009 Jul 15;180(2):122e33 [PubMed PMID: 19406982. Epub 2009/05/02. eng]. [70] Watelet JB, Bachert C, Claeys C, Van Cauwenberge P. Matrix metalloproteinases MMP-7, MMP-9 and their tissue inhibitor TIMP-1: expression in chronic sinusitis vs nasal polyposis. Allergy 2004 Jan;59(1):54e60 [PubMed PMID: 14674934. Epub 2003/12/17. eng]. [71] Bourdin A, Neveu D, Vachier I, Paganin F, Godard P, Chanez P. Specificity of basement membrane thickening in severe asthma. J Allergy Clin Immunol 2007 Jun;119(6):1367e74 [PubMed PMID: 17481707. Epub 2007/05/08]. [72] James AL, Bai TR, Mauad T, Abramson MJ, Dolhnikoff M, McKay KO, et al. Airway smooth muscle thickness in asthma is related to severity but not duration of asthma. Eur Respir J
1419
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
[87]
2009 Nov;34(5):1040e5 [PubMed PMID: 19282340. Epub 2009/03/14]. James AL, Maxwell PS, Pearce-Pinto G, Elliot JG, Carroll NG. The relationship of reticular basement membrane thickness to airway wall remodeling in asthma. Am J Respir Crit Care Med 2002 Dec 15;166(12 Pt 1):1590e5 [PubMed PMID: 12471074. Epub 2002/12/10]. Hyde DM, Miller LA, Schelegle ES, Fanucchi MV, Van Winkle LS, Tyler NK, et al. Asthma: a comparison of animal models using stereological methods. Eur Respir Rev December 1, 2006;15(101):122e35. Hamid Q. Pathogenesis of small airways in asthma. Respiration. Int Rev Thorac Dis 2012;84(1):4e11 [PubMed PMID: 22759947. Epub 2012/07/05]. James AL, Elliot JG, Jones RL, Carroll ML, Mauad T, Bai TR, et al. Airway smooth muscle hypertrophy and hyperplasia in asthma. Am J Respir Crit Care Med 2012 May 15;185(10): 1058e64 [PubMed PMID: 22403800. Epub 2012/03/10]. Ito I, Fixman ED, Asai K, Yoshida M, Gounni AS, Martin JG, et al. Platelet-derived growth factor and transforming growth factor-beta modulate the expression of matrix metalloproteinases and migratory function of human airway smooth muscle cells. Clin Exp Allergy 2009 Sep;39(9): 1370e80 [PubMed PMID: 19522858. Epub 2009/06/16. eng]. Dekkers BG, Bos IS, Gosens R, Halayko AJ, Zaagsma J, Meurs H. The integrin-blocking peptide RGDS inhibits airway smooth muscle remodeling in a guinea pig model of allergic asthma. Am J Respir Crit Care Med 2010 Mar 15;181(6): 556e65 [PubMed PMID: 20019343. Epub 2009/12/19]. Holgate ST, Polosa R. The mechanisms, diagnosis, and management of severe asthma in adults. Lancet 2006 Aug 26; 368(9537):780e93 [PubMed PMID: 16935689. Epub 2006/08/29. eng]. Yang YC, Zhang N, Van Crombruggen K, Hu GH, Hong SL, Bachert C. Transforming growth factor-beta1 in inflammatory airway disease: a key for understanding inflammation and remodeling. Allergy 2012 Oct;67(10):1193e202 [PubMed PMID: 22913656. Epub 2012/08/24. eng]. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 2006 May 11;441(7090):235e8 [PubMed PMID: 16648838. Epub 2006/05/02. eng]. Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, et al. IL-4 inhibits TGF-beta-induced Foxp3þ T cells and, together with TGF-beta, generates IL-9þ IL-10þ Foxp3(-) effector T cells. Nat Immunol 2008 Dec;9(12): 1347e55 [PubMed PMID: 18997793. Epub 2008/11/11. eng]. Sakaguchi S. Naturally arising CD4þ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol 2004;22:531e62 [PubMed PMID: 15032588. Epub 2004/03/23]. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008 May 30;133(5):775e87 [PubMed PMID: 18510923. Epub 2008/05/31]. Sakaguchi S, Wing K. Immunology. Damping by depletion. Science 2011 Apr 29;332(6029):542e3 [PubMed PMID: 21527700. Epub 2011/04/30]. Wing K, Yamaguchi T, Sakaguchi S. Cell-autonomous and -non-autonomous roles of CTLA-4 in immune regulation. Trends Immunol 2011 Sep;32(9):428e33 [PubMed PMID: 21723783. Epub 2011/07/05]. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al. Expression of helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3þ T regulatory cells. J Immunol 2010 Apr 1;184(7):3433e41 [PubMed PMID: 20181882. Pubmed Central PMCID: 3725574. Epub 2010/02/26].
1420 [88] Lahl K, Mayer CT, Bopp T, Huehn J, Loddenkemper C, Eberl G, et al. Nonfunctional regulatory T cells and defective control of Th2 cytokine production in natural scurfy mutant mice. J Immunol 2009 Nov 1;183(9):5662e72 [PubMed PMID: 19812199. Epub 2009/10/09]. [89] Pandiyan P, Zheng L, Lenardo MJ. The molecular mechanisms of regulatory T cell immunosuppression. Front Immunol 2011; 2(60) [PubMed PMID: 22566849. Pubmed Central PMCID: 3342245. Epub 2011/01/01]. [90] Joetham A, Okamoto M, Takeda K, Schedel M, Ohnishi H, Dakhama A, et al. CD8 regulates T regulatory cell production of IL-6 and maintains their suppressive phenotype in allergic lung disease. J Immunol 2011 Jan 1;186(1):113e20 [PubMed PMID: 21115736. Pubmed Central PMCID: 3127584. Epub 2010/12/01]. [91] Wan YY, Flavell RA. Regulatory T cells, transforming growth factor-beta, and immune suppression. Proc Am Thorac Soc 2007 Jul;4(3):271e6 [PubMed PMID: 17607012. Pubmed Central PMCID: 2647629. Epub 2007/07/04]. [92] Andersson J, Tran DQ, Pesu M, Davidson TS, Ramsey H, O’Shea JJ, et al. CD4þ FoxP3þ regulatory T cells confer infectious tolerance in a TGF-beta-dependent manner. J Exp Med 2008 Sep 1;205(9):1975e81 [PubMed PMID: 18710931. Pubmed Central PMCID: 2526184. Epub 2008/08/20]. [93] Pandiyan P, Lenardo MJ. The control of CD4þCD25þFoxp3þ regulatory T cell survival. Biol direct 2008;3(6) [PubMed PMID: 18304352. Pubmed Central PMCID: 2270257. Epub 2008/02/29]. [94] Shevach EM, Tran DQ, Davidson TS, Andersson J. The critical contribution of TGF-beta to the induction of Foxp3 expression and regulatory T cell function. Eur J Immunol 2008 Apr; 38(4):915e7 [PubMed PMID: 18395859. Pubmed Central PMCID: 2662375. Epub 2008/04/09]. [95] Chen Y, Kuchroo VK, Inobe J, Hafler DA, Weiner HL. Regulatory T cell clones induced by oral tolerance: suppression of autoimmune encephalomyelitis. Science 1994 Aug 26; 265(5176):1237e40 [PubMed PMID: 7520605. Epub 1994/08/26]. [96] Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4þCD25- naive T cells to CD4þCD25þ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med 2003 Dec 15; 198(12):1875e86 [PubMed PMID: 14676299. Epub 2003/12/17. eng]. [97] Kearley J, Robinson DS, Lloyd CM. CD4þCD25þ regulatory T cells reverse established allergic airway inflammation and prevent airway remodeling. J Allergy Clin Immunol 2008 Sep; 122(3). 617e624 e6. [PubMed PMID: 18672278. Epub 2008/08/02. eng]. [98] Joetham A, Takeda K, Taube C, Miyahara N, Matsubara S, Koya T, et al. Naturally occurring lung CD4(þ)CD25(þ) T cell regulation of airway allergic responses depends on IL-10 induction of TGF-beta. J Immunol 2007 Feb 1;178(3):1433e42 [PubMed PMID: 17237391. Epub 2007/01/24. eng]. [99] Duvernelle C, Freund V, Frossard N. Transforming growth factor-beta and its role in asthma. Pulm Pharmacol Ther 2003;16(4):181e96 [PubMed PMID: 12850120. Epub 2003/07/10. eng]. [100] Van Hove CL, Maes T, Joos GF, Tournoy KG. Chronic inflammation in asthma: a contest of persistence vs resolution. Allergy 2008 Sep;63(9):1095e109 [PubMed PMID: 18616676. Epub 2008/07/12. eng]. [101] Chakir J, Shannon J, Molet S, Fukakusa M, Elias J, Laviolette M, et al. Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGFbeta, IL-11, IL-17, and type I and type III collagen expression. J Allergy Clin Immunol 2003 Jun;111(6):1293e8 [PubMed PMID: 12789232. Epub 2003/06/06. eng].
M. Al-Alawi et al. [102] Brown SD, Baxter KM, Stephenson ST, Esper AM, Brown LA, Fitzpatrick AM. Airway TGF-beta1 and oxidant stress in children with severe asthma: association with airflow limitation. J Allergy Clin Immunol 2012 Feb;129(2):388e96. 96 e1-8. [PubMed PMID: 22206775. Pubmed Central PMCID: 3268912. Epub 2011/12/31]. [103] Michaeloudes C, Chang PJ, Petrou M, Chung KF. Transforming growth factor-beta and nuclear factor E2-related factor 2 regulate antioxidant responses in airway smooth muscle cells: role in asthma. Am J Respir Crit Care Med 2011 Oct 15; 184(8):894e903 [PubMed PMID: 21799075. Pubmed Central PMCID: 3402549. Epub 2011/07/30]. [104] Fitzpatrick AM, Stephenson ST, Hadley GR, Burwell L, Penugonda M, Simon DM, et al. Thiol redox disturbances in children with severe asthma are associated with posttranslational modification of the transcription factor nuclear factor (erythroid-derived 2)-like 2. J Allergy Clin Immunol 2011 Jun;127(6):1604e11 [PubMed PMID: 21514635. Pubmed Central PMCID: 3105207. Epub 2011/04/26]. [105] Boulet LP, Lemiere C, Archambault F, Carrier G, Descary MC, Deschesnes F. Smoking and asthma: clinical and radiologic features, lung function, and airway inflammation. Chest 2006 Mar;129(3):661e8 [PubMed PMID: 16537865. Epub 2006/03/16]. [106] McConnell R, Berhane K, Yao L, Jerrett M, Lurmann F, Gilliland F, et al. Traffic, susceptibility, and childhood asthma. Environ Health Perspect 2006 May;114(5):766e72 [PubMed PMID: 16675435. Pubmed Central PMCID: 1459934. Epub 2006/05/06]. [107] Salam MT, Gauderman WJ, McConnell R, Lin PC, Gilliland FD. Transforming growth factor- 1 C-509T polymorphism, oxidant stress, and early-onset childhood asthma. Am J Respir Crit Care Med 2007 Dec 15;176(12):1192e9 [PubMed PMID: 17673695. Pubmed Central PMCID: 2176104. Epub 2007/08/04]. [108] Dai J, Xie C, Vincent R, Churg A. Air pollution particles produce airway wall remodeling in rat tracheal explants. Am J Respir Cell Mol Biol 2003 Sep;29(3 Pt 1):352e8 [PubMed PMID: 12649123. Epub 2003/03/22]. [109] Wang RD, Wright JL, Churg A. Transforming growth factorbeta1 drives airway remodeling in cigarette smoke-exposed tracheal explants. Am J Respir Cell Mol Biol 2005 Oct;33(4): 387e93 [PubMed PMID: 15994428. Epub 2005/07/05]. [110] Kim DY, Kwon EY, Hong GU, Lee YS, Lee SH, Ro JY. Cigarette smoke exacerbates mouse allergic asthma through Smad proteins expressed in mast cells. Respir Res 2011;12(49) [PubMed PMID: 21496353. Pubmed Central PMCID: 3098800. Epub 2011/04/19]. [111] Kang MJ, Homer RJ, Gallo A, Lee CG, Crothers KA, Cho SJ, et al. IL-18 is induced and IL-18 receptor alpha plays a critical role in the pathogenesis of cigarette smoke-induced pulmonary emphysema and inflammation. J Immunol 2007 Feb 1;178(3):1948e59 [PubMed PMID: 17237446. Epub 2007/01/24]. [112] Lee KS, Kim SR, Park SJ, Min KH, Lee KY, Jin SM, et al. Antioxidant down-regulates interleukin-18 expression in asthma. Mol Pharmacol 2006 Oct;70(4):1184e93 [PubMed PMID: 16822930. Epub 2006/07/11]. [113] Yamagata S, Tomita K, Sato R, Niwa A, Higashino H, Tohda Y. Interleukin-18-deficient mice exhibit diminished chronic inflammation and airway remodelling in ovalbumin-induced asthma model. Clin Exp Immunol 2008 Dec;154(3):295e304 [PubMed PMID: 18826499. Epub 2008/10/02. eng]. [114] Chen ZG, Zhang TT, Li HT, Chen FH, Zou XL, Ji JZ, et al. Neutralization of TSLP inhibits airway remodeling in a murine model of allergic asthma induced by chronic exposure to house dust mite. PLoS One 2013;8(1):e51268 [PubMed PMID: 23300949. Epub 2013/01/10. eng].
Transforming growth factor beta in severe asthma [115] Price D, Bjermer L, Popov TA, Chisholm A. Integrating evidence for managing asthma in patients who smoke. Allergy Asthma Immunol Res 2014 Mar;6(2):114e20 [PubMed PMID: 24587946. Pubmed Central PMCID: 3936038. Epub 2014/03/04]. [116] Tanaka H, Komai M, Nagao K, Ishizaki M, Kajiwara D, Takatsu K, et al. Role of interleukin-5 and eosinophils in allergen-induced airway remodeling in mice. Am J Respir Cell Mol Biol 2004 Jul;31(1):62e8 [PubMed PMID: 14975941. Epub 2004/02/21. eng]. [117] Nakao A, Miike S, Hatano M, Okumura K, Tokuhisa T, Ra C, et al. Blockade of transforming growth factor beta/Smad signaling in T cells by overexpression of Smad7 enhances antigen-induced airway inflammation and airway reactivity. J Exp Med 2000 Jul 17;192(2):151e8 [PubMed PMID: 10899902. Epub 2000/07/19. eng]. [118] Ingram JL, Huggins MJ, Church TD, Li Y, Francisco DC, Degan S, et al. Airway fibroblasts in asthma manifest an invasive phenotype. Am J Respir Crit Care Med 2011 Jun 15; 183(12):1625e32 [PubMed PMID: 21471104. Epub 2011/04/08. eng]. [119] Hirota JA, Ask K, Fritz D, Ellis R, Wattie J, Richards CD, et al. Role of STAT6 and SMAD2 in a model of chronic allergen exposure: a mouse strain comparison study. Clin Exp Allergy 2009 Jan;39(1):147e58 [PubMed PMID: 19032363. Epub 2008/11/27. eng]. [120] McMillan SJ, Xanthou G, Lloyd CM. Manipulation of allergeninduced airway remodeling by treatment with anti-TGF-beta antibody: effect on the Smad signaling pathway. J Immunol 2005 May 1;174(9):5774e80 [PubMed PMID: 15843580. Epub 2005/04/22. eng]. [121] Kobayashi T, Liu X, Wen FQ, Fang Q, Abe S, Wang XQ, et al. Smad3 mediates TGF-beta1 induction of VEGF production in lung fibroblasts. Biochem Biophys Res Commun 2005 Feb 11; 327(2):393e8 [PubMed PMID: 15629128. Epub 2005/01/05. eng]. [122] Lee KS, Park SJ, Kim SR, Min KH, Lee KY, Choe YH, et al. Inhibition of VEGF blocks TGF-beta1 production through a PI3K/Akt signalling pathway. Eur Respir J 2008 Mar;31(3): 523e31 [PubMed PMID: 18057050. Epub 2007/12/07]. [123] Redington AE, Madden J, Frew AJ, Djukanovic R, Roche WR, Holgate ST, et al. Transforming growth factor-beta 1 in asthma. Measurement in bronchoalveolar lavage fluid. Am J Respir Crit Care Med 1997 Aug;156(2 Pt 1):642e7 [PubMed 0PMID: 9279252. Epub 1997/08/01. eng]. [124] Ohno I, Nitta Y, Yamauchi K, Hoshi H, Honma M, Woolley K, et al. Transforming growth factor beta 1 (TGF beta 1) gene expression by eosinophils in asthmatic airway inflammation. Am J Respir Cell Mol Biol 1996 Sep;15(3):404e9 [PubMed PMID: 8810646. Epub 1996/09/01. eng]. [125] Elias PM, Hatano Y, Williams ML. Basis for the barrier abnormality in atopic dermatitis: outside-inside-outside pathogenic mechanisms. J Allergy Clin Immunol 2008 Jun;121(6): 1337e43 [PubMed PMID: 18329087. Pubmed Central PMCID: 2706021. Epub 2008/03/11]. [126] Green RH, Brightling CE, McKenna S, Hargadon B, Parker D, Bradding P, et al. Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial. Lancet 2002 Nov 30;360(9347):1715e21 [PubMed PMID: 12480423. Epub 2002/12/14. eng]. [127] Wenzel SE, Schwartz LB, Langmack EL, Halliday JL, Trudeau JB, Gibbs RL, et al. Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med 1999 Sep;160(3):1001e8 [PubMed PMID: 10471631. Epub 1999/09/03. eng]. [128] Nair P, Pizzichini MM, Kjarsgaard M, Inman MD, Efthimiadis A, Pizzichini E, et al. Mepolizumab for prednisone-dependent
1421
[129]
[130]
[131]
[132]
[133]
[134]
[135]
[136]
[137]
[138]
[139]
[140]
[141]
asthma with sputum eosinophilia. N Engl J Med 2009 Mar 5; 360(10):985e93 [PubMed PMID: 19264687. Epub 2009/03/07. eng]. Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med 2009 Mar 5;360(10): 973e84 [PubMed PMID: 19264686. Epub 2009/03/07. eng]. Pavord ID, Korn S, Howarth P, Bleecker ER, Buhl R, Keene ON, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 2012 Aug 18;380(9842):651e9 [PubMed PMID: 22901886. Epub 2012/08/21. eng]. Cho JY, Miller M, Baek KJ, Han JW, Nayar J, Lee SY, et al. Inhibition of airway remodeling in IL-5-deficient mice. J Clin Invest 2004 Feb;113(4):551e60 [PubMed PMID: 14966564. Epub 2004/02/18. eng]. Chiang CH, Chuang CH, Liu SL, Shen HD. Genetic polymorphism of transforming growth factor beta1 and tumor necrosis factor alpha is associated with asthma and modulates the severity of asthma. Respir Care 2013 Aug;58(8): 1343e50 [PubMed PMID: 23466425. Epub 2013/03/08]. Meng J, Thongngarm T, Nakajima M, Yamashita N, Ohta K, Bates CA, et al. Association of transforming growth factorbeta1 single nucleotide polymorphism C-509T with allergy and immunological activities. Int Arch Allergy Immunol 2005 Oct;138(2):151e60 [PubMed PMID: 16179826. Epub 2005/09/24. eng]. Yuan X, Wei Q, Komaki R, Liu Z, Yang J, Tucker SL, et al. Polymorphisms predict distant metastasis-free survival in patients with inoperable non-small-cell lung Cancer after definitive radiotherapy. PloS One 2013;8(6):e65659 [PubMed PMID: 23840350. Pubmed Central PMCID: 3686751]. Pooja S, Francis A, Rajender S, Tamang R, Rajkumar R, Saini KS, et al. Strong impact of TGF-beta1 gene polymorphisms on breast cancer risk in Indian women: a casecontrol and population-based study. PloS One 2013;8(10): e75979 [PubMed PMID: 24146803. Pubmed Central PMCID: 3798290]. Niu X, Li H, Chen Z, Liu Y, Kan M, Zhou D, et al. A study of ethnic differences in TGFbeta1 gene polymorphisms and effects on the risk of radiation pneumonitis in non-small-cell lung cancer. J Thorac Oncol 2012 Nov;7(11):1668e75 [PubMed PMID: 23059779]. Zhang Y, Zhang J, Huang J, Li X, He C, Tian C, et al. Polymorphisms in the transforming growth factor-beta1 gene and the risk of asthma: a meta-analysis. Respirology 2010 May; 15(4):643e50 [PubMed PMID: 20409029. Epub 2010/04/23. eng]. Ierodiakonou D, Postma DS, Koppelman GH, Gerritsen J, ten Hacken NH, Timens W, et al. TGF-beta1 polymorphisms and asthma severity, airway inflammation, and remodeling. J Allergy Clin Immunol 2013 Feb;131(2):582e5 [PubMed PMID: 23111237. Epub 2012/11/01]. Liebhart J, Polak M, Dabrowski A, Dobek R, Liebhart E, DorWojnarowska A, et al. The G/G genotype of transforming growth factor beta 1 (TGF-beta1) single nucleotide (þ915G/C) polymorphism coincident with other host and environmental factors is associated with irreversible bronchoconstriction in asthmatics. Int J Immunogenet 2008 Dec; 35(6):417e22 [PubMed PMID: 19046298. Epub 2008/12/03. eng]. Pulleyn LJ, Newton R, Adcock IM, Barnes PJ. TGFbeta1 allele association with asthma severity. Hum Genet 2001 Dec; 109(6):623e7 [PubMed PMID: 11810274. Epub 2002/01/26. eng]. de Faria IC, de Faria EJ, Toro AA, Ribeiro JD, Bertuzzo CS. Association of TGF-beta1, CD14, IL-4, IL-4R and ADAM33 gene polymorphisms with asthma severity in children and
1422
[142]
[143]
[144]
[145]
[146]
[147]
[148]
[149]
[150]
[151]
[152]
[153]
[154]
M. Al-Alawi et al. adolescents. J Pediatr (Rio J) 2008 May-Jun;84(3):203e10 [PubMed PMID: 18425216. Epub 2008/04/22. eng]. Che Z, Zhu X, Yao C, Liu Y, Chen Y, Cao J, et al. The association between the C-509T and T869C polymorphisms of TGF-beta1 gene and the risk of asthma: a meta-analysis. Hum Immunol 2014 Feb;75(2):141e50 [PubMed PMID: 24269701. Epub 2013/11/26]. Freimuth J, Clermont FF, Huang X, DeSapio A, Tokuyasu TA, Sheppard D, et al. Epistatic interactions between Tgfb1 and genetic loci, Tgfbm2 and Tgfbm3, determine susceptibility to an asthmatic stimulus. Proc Natl Acad Sci U S A 2012 Oct 30; 109(44):18042e7 [PubMed PMID: 23064636. Pubmed Central PMCID: 3497801. Epub 2012/10/16]. Frischmeyer-Guerrerio PA, Guerrerio AL, Oswald G, Chichester K, Myers L, Halushka MK, et al. TGFbeta receptor mutations impose a strong predisposition for human allergic disease. Sci Transl Med 2013 Jul 24;5(195):195ra94 [PubMed PMID: 23884466. Epub 2013/07/26. eng]. Habashi JP, Judge DP, Holm TM, Cohn RD, Loeys BL, Cooper TK, et al. Losartan, an AT1 antagonist, prevents aortic aneurysm in a mouse model of Marfan syndrome. Science 2006 Apr 7;312(5770):117e21 [PubMed PMID: 16601194. Pubmed Central PMCID: 1482474. Epub 2006/04/08]. Lim DS, Lutucuta S, Bachireddy P, Youker K, Evans A, Entman M, et al. Angiotensin II blockade reverses myocardial fibrosis in a transgenic mouse model of human hypertrophic cardiomyopathy. Circulation 2001 Feb 13;103(6):789e91 [PubMed PMID: 11171784. Pubmed Central PMCID: 2779524. Epub 2001/02/15]. Miller M, Cho JY, McElwain K, McElwain S, Shim JY, Manni M, et al. Corticosteroids prevent myofibroblast accumulation and airway remodeling in mice. Am J Physiol Lung Cell Mol Physiol 2006 Jan;290(1):L162e9 [PubMed PMID: 16344333. Epub 2005/12/14. eng]. Schwartze JT, Becker S, Sakkas E, Wujak LA, Niess G, Usemann J, et al. Glucocorticoids recruit Tgfbr3 and Smad1 to shift transforming growth factor-beta signaling from the Tgfbr1/Smad2/3 axis to the Acvrl1/Smad1 axis in lung fibroblasts. J Biol Chem 2014 Feb 7;289(6):3262e75 [PubMed PMID: 24347165. Pubmed Central PMCID: 3916529. Epub 2013/12/19]. Karagiannidis C, Hense G, Martin C, Epstein M, Ruckert B, Mantel PY, et al. Activin A is an acute allergen-responsive cytokine and provides a link to TGF-beta-mediated airway remodeling in asthma. J Allergy Clin Immunol 2006 Jan; 117(1):111e8 [PubMed PMID: 16387593. Epub 2006/01/03]. Kariyawasam HH, Pegorier S, Barkans J, Xanthou G, Aizen M, Ying S, et al. Activin and transforming growth factor-beta signaling pathways are activated after allergen challenge in mild asthma. J Allergy Clin Immunol 2009 Sep;124(3):454e62 [PubMed PMID: 19733294. Epub 2009/09/08]. Hardy CL, Nguyen HA, Mohamud R, Yao J, Oh DY, Plebanski M, et al. The activin A antagonist follistatin inhibits asthmatic airway remodelling. Thorax 2013 Jan;68(1):9e18 [PubMed PMID: 23051972. Epub 2012/10/12]. Ohta S, Oda N, Yokoe T, Tanaka A, Yamamoto Y, Watanabe Y, et al. Effect of tiotropium bromide on airway inflammation and remodelling in a mouse model of asthma. Clin Exp Allergy 2010 Aug;40(8):1266e75 [PubMed PMID: 20337647. Epub 2010/03/27]. Kang JY, Rhee CK, Kim JS, Park CK, Kim SJ, Lee SH, et al. Effect of tiotropium bromide on airway remodeling in a chronic asthma model. Ann allergy Asthma Immunol 2012 Jul;109(1):29e35 [PubMed PMID: 22727154. Epub 2012/06/26]. Schaafsma D, McNeill KD, Mutawe MM, Ghavami S, Unruh H, Jacques E, et al. Simvastatin inhibits TGFbeta1-induced
[155]
[156]
[157]
[158]
[159]
[160]
[161]
[162]
[163]
[164]
[165]
[166]
fibronectin in human airway fibroblasts. Respir Res 2011; 12(113) [PubMed PMID: 21864337. Pubmed Central PMCID: 3173339. Epub 2011/08/26]. Michalik M, Soczek E, Kosinska M, Rak M, Wojcik KA, Lasota S, et al. Lovastatin-induced decrease of intracellular cholesterol level attenuates fibroblast-to-myofibroblast transition in bronchial fibroblasts derived from asthmatic patients. Eur J Pharmacol 2013 Mar 15;704(1e3):23e32 [PubMed PMID: 23485731. Epub 2013/03/15]. McCormick LL, Zhang Y, Tootell E, Gilliam AC. Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J Immunol 1999 Nov 15;163(10):5693e9 [PubMed PMID: 10553100. Epub 1999/11/24. eng]. Fukasawa H, Yamamoto T, Suzuki H, Togawa A, Ohashi N, Fujigaki Y, et al. Treatment with anti-TGF-beta antibody ameliorates chronic progressive nephritis by inhibiting Smad/TGF-beta signaling. Kidney Int 2004 Jan;65(1):63e74 [PubMed PMID: 14675037. Epub 2003/12/17. eng]. Leung SY, Niimi A, Noble A, Oates T, Williams AS, Medicherla S, et al. Effect of transforming growth factorbeta receptor I kinase inhibitor 2,4-disubstituted pteridine (SD-208) in chronic allergic airway inflammation and remodeling. J Pharmacol Exp Ther 2006 Nov;319(2):586e94 [PubMed PMID: 16888081. Epub 2006/08/05]. Fattouh R, Midence NG, Arias K, Johnson JR, Walker TD, Goncharova S, et al. Transforming growth factor-beta regulates house dust mite-induced allergic airway inflammation but not airway remodeling. Am J Respir Crit Care Med 2008 Mar 15;177(6):593e603 [PubMed PMID: 18174546. Epub 2008/01/05. eng]. Mabalirajan U, Aich J, Agrawal A, Ghosh B. Mepacrine inhibits subepithelial fibrosis by reducing the expression of arginase and TGF-beta1 in an extended subacute mouse model of allergic asthma. Am J Physiol Lung Cell Mol Physiol 2009 Sep;297(3):L411e9 [PubMed PMID: 19542246. Epub 2009/06/23. eng]. Wen FQ, Liu X, Kobayashi T, Abe S, Fang Q, Kohyama T, et al. Interferon-gamma inhibits transforming growth factor-beta production in human airway epithelial cells by targeting Smads. Am J Respir Cell Mol Biol 2004 Jun;30(6):816e22 [PubMed PMID: 14722222. Epub 2004/01/15. eng]. Choy DF, Modrek B, Abbas AR, Kummerfeld S, Clark HF, Wu LC, et al. Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways. J Immunol 2011 Feb 1;186(3):1861e9 [PubMed PMID: 21187436. Pubmed Central PMCID: 3981556. Epub 2010/12/29]. Cordeiro MF, Mead A, Ali RR, Alexander RA, Murray S, Chen C, et al. Novel antisense oligonucleotides targeting TGF-beta inhibit in vivo scarring and improve surgical outcome. Gene Ther 2003 Jan;10(1):59e71 [PubMed PMID: 12525838. Epub 2003/01/15. eng]. Gregory LG, Mathie SA, Walker SA, Pegorier S, Jones CP, Lloyd CM. Overexpression of Smad2 drives house dust mitemediated airway remodeling and airway hyperresponsiveness via activin and IL-25. Am J Respir Crit Care Med 2010 Jul 15;182(2):143e54 [PubMed PMID: 20339149. Epub 2010/03/27. eng]. Redington AE, Roche WR, Holgate ST, Howarth PH. Colocalization of immunoreactive transforming growth factorbeta 1 and decorin in bronchial biopsies from asthmatic and normal subjects. J Pathol 1998 Dec;186(4):410e5 [PubMed PMID: 10209491. Epub 1999/04/21. eng]. Aubert JD, Dalal BI, Bai TR, Roberts CR, Hayashi S, Hogg JC. Transforming growth factor beta 1 gene expression in human airways. Thorax 1994 Mar;49(3):225e32 [PubMed PMID: 8202878. Epub 1994/03/01. eng].
Transforming growth factor beta in severe asthma [167] Bosse Y, Thompson C, Stankova J, Rola-Pleszczynski M. Fibroblast growth factor 2 and transforming growth factor beta1 synergism in human bronchial smooth muscle cell proliferation. Am J Respir Cell Mol Biol 2006 Jun;34(6):746e53 [PubMed PMID: 16439802. Epub 2006/01/28. eng]. [168] Bosse Y, Rola-Pleszczynski M. FGF2 in asthmatic airwaysmooth-muscle-cell hyperplasia. Trends Mol Med 2008 Jan;
1423 14(1):3e11 [PubMed PMID: 18055262. Epub 2007/12/07. eng]. [169] Redington AE, Roche WR, Madden J, Frew AJ, Djukanovic R, Holgate ST, et al. Basic fibroblast growth factor in asthma: measurement in bronchoalveolar lavage fluid basally and following allergen challenge. J Allergy Clin Immunol 2001 Feb;107(2):384e7 [PubMed PMID: 11174209. Epub 2001/02/15. eng].