Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jan 1.
Published in final edited form as: Biochim Biophys Acta. 2012 Oct 6;1832(7):1049–1060. doi: 10.1016/j.bbadis.2012.09.014

Cytokine mediated tissue fibrosis

Lee A Borthwick a,b,*, Thomas A Wynn b, Andrew J Fisher a,c
PMCID: PMC3787896  NIHMSID: NIHMS516837  PMID: 23046809

Abstract

Acute inflammation is a recognised part of normal wound healing. However, when inflammation fails to resolve and a chronic inflammatory response is established this process can become dysregulated resulting in pathological wound repair, accumulation of permanent fibrotic scar tissue at the site of injury and the failure to return the tissue to normal function. Fibrosis can affect any organ including the lung, skin, heart, kidney and liver and it is estimated that 45% of deaths in the western world can now be attributed to diseases where fibrosis plays a major aetiological role. In this review we examine the evidence that cytokines play a vital role in the acute and chronic inflammatory responses that drive fibrosis in injured tissues. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.

Keywords: Fibrosis, Cytokine, Macrophage, Fibroblast, Myofibroblast, Inflammation

1. Introduction

Physiological wound repair is a complex, highly orchestrated process that allows for the replacement of dead or damaged cells and is critically important in restoring homeostasis to a tissue after injury. Wound repair can be loosely defined by three overlapping stages; an initial response, a recovery of integrity, followed finally by resolution of the wound back to a functional epithelium. It requires a tightly regulated spatial and temporal response from key structural cells in the organ such as epithelial cells, endothelial cells and fibroblasts but also from immune and progenitor cells drawn from the circulatory system [1]. Acute inflammation is a recognised part of normal wound healing by serving as an innate immune response to the disrupted epithelial surface until it is reinstated. However, when inflammation fails to resolve and a chronic inflammatory response is established this process can become dysregulated resulting in pathological wound repair and the accumulation of permanent fibrotic scar tissue at the site of injury (Fig. 1). This fibrosis is characterised by the excessive accumulation of extra cellular matrix (ECM) components including collagens, fibronectin and hyaluronic acid at the site of tissue injury, leading to a decrease in organ function and, in some cases, organ failure and death [2]. It is estimated that 45% of deaths in the western world can now be attributed to diseases where fibrosis plays a major aetiological role [3]. Fibrosis can affect any organ including the lung, skin, heart, kidney and liver and may represent an aberrant response to a single major injury but more commonly is a response to a persistent or repetitive injury. In this review we examine the evidence for cytokines released as part of an acute or chronic inflammatory response in driving fibrosis in injured tissues.

Fig. 1.

Fig. 1

Pathological vs. physiological wound healing. Infection or exposure to harmful molecules can lead to epithelial damage and loss of epithelial integrity. Following injury fibroblasts, endothelial cells, and neighbouring epithelial cells release a range of soluble factors that trigger clotting and initiate the development of a provisional ECM. The aggregation and subsequent degranulation of platelets triggers increased blood flow, vasculature dilation and vasculature permeability allowing the effective recruitment of inflammatory cells to the site of tissue injury. The first responders are the neutrophils, eosinophils and basophils that are responsible for neutralising any invading pathogens via an oxidative burst response and eliminating cell debris/dying cells by phagocytosis. The granulocyte number in the site of epithelial injury peaks rapidly, within minutes, but is followed by a rapid decline. Once in the wound micro-environment the recruited monocytes mature to increase the number of macrophages in the wound and perform similar functions to those described for granulocytes. In addition they produce cytokines and chemokines that amplify the wound response by promoting the formation and stabilisation of a provisional ECM and promoting angiogenesis. Myofibroblast numbers are increased at the wound site from several sources (see Fig. 2). Once recruited to the wound area the myofibroblasts become activated and traverse the provisional ECM until they reach the edge of the wound and initiate contraction of the wound. Finally epithelial cells at the edge of the wound loosen adherence junctions and migrate over the ECM to restore a continuous epithelium and tissue homeostasis. At this point the myofibroblasts in the wound area undergo apoptosis and the macrophage numbers are significantly reduced via egress into the lymphatic system. Fibrosis occurs when the initial wound is severe, the wound repair process becomes dysregulated or the source of epithelial damage persists resulting in repeated injury and chronic inflammation.

2. Pathogen and damage associated molecular patterns in fibrosis

A functional epithelium provides an efficient barrier against microorganisms and other potentially harmful molecules via a wide range of mechanisms including mucociliary clearance, maintenance of epithelial adherence and tight junctions, homeostasis of ion and water transport and secretion of antibacterial, antimicrobial and antiprotease molecules [4]. However the epithelium is often located on vulnerable surfaces that receive significant challenges to their integrity such as the gut, skin and lungs. These tissues are routinely exposed to the external environment and a range of harmful molecules including bacteria and viruses, tobacco smoke, asbestos, silica and diesel exhaust that can lead to epithelial activation and, in cases of chronic exposure, epithelial damage, shedding and denudation.

Numerous fibrotic diseases are believed to have an infectious aetiology with bacteria (Pseudomonas aeruginosa, Mycobacterium tuberculosis), viruses (HCV, Respiratory syncytial virus), fungi (Aspergillus fumigatus, Cryptococcus neoformans) and multi-cellular parasites (Schistosoma mansoni, Toxoplasma gondii) driving wounding, chronic inflammation and subsequent fibrosis in multiple organs [513]. Pathogen by-products including bacterial DNA and double stranded RNA, peptidoglycan, lipopolysaccharide and flagellin, collectively referred to as pathogen-associated molecular patterns (PAMPs), are recognised by pattern recognition receptors (PRR) on a wide range of cell types including immune cells (macrophages, neutrophils, T-cells, B-cells, dendritic cell, eosinophils) and structural cells (epithelial cells, fibroblasts, adipocytes) [14,15]. The interaction between PAMPs and PRR provides an evolutionarily conserved mechanism that provides the first line of defence against invading pathogens and activates numerous proinflammatory cytokine and chemokine pathways, leading to the eradication of the pathogen. The failure to clear the pathogen or its PAMPs provides a persistent source of tissue injury, chronic inflammation and creates an environment that might favour fibrosis. For example persistent colonisation of the allograft with Pseudomonas aeruginosa following lung transplantation is strongly associated with the subsequent development of bronchiolitis obliterans syndrome (BOS) [16] and prolonged infection with hepatitis C virus (HCV) or hepatitis B virus (HBV) leads to loss of liver architecture and function and ultimately cirrhosis [17,18].

It is also increasingly apparent that PRRs provide mechanisms for mounting inflammatory and wound-healing responses to sterile tissue trauma [19]. When epithelial cells are damaged or dying their membranes lose integrity and intracellular proteins leak into the external environment. These damage associated molecular pattern molecules (DAMPs) or alarmins include high-mobility box group 1 (HMGB-1), heat-shock proteins (HSP60, HSP70), interleukin (IL)-33 and IL-1α among others [20]. DAMPs can trigger innate immune responses in a wide variety of cell types via engagement of PRR and provides an important homeostatic mechanism by which the immune system can sense and mount wound-repair responses in damaged tissues [21]. However, there is also evidence that DAMPs can contribute to the pathogenesis of many inflammatory and fibrotic diseases. For example IL-33 is strongly associated with fibrosis in chronic liver injury [22] and is increased in systemic sclerosis patients, correlating with the extent of skin sclerosis and the severity of pulmonary fibrosis [23]. In addition HMGB-1 levels are elevated in the bronchoalveolar lavage (BAL) of patients with idiopathic pulmonary fibrosis (IPF) and hypersensitivity pneumonitis [24].

Fibroblasts express a number of PRR including toll-like receptors (TLR) and IL-1R therefore PAMPs and DAMPs can directly activate them and drive their differentiation to myofibroblasts [7,25,26]. It has also recently been suggested that there are differences in TLR expression and activation between normal fibroblasts and those isolated from patients with severe idiopathic pulmonary pneumonia [7,27]. Taken together these data suggest that myofibroblasts in a fibrotic environment may be maintained in a heightened state of readiness, primed to respond to small quantities of DAMPs and PAMPs. Therefore inhibiting TLR signalling may represent a novel approach to limit activation of the innate immune response, decrease inflammation and limit or reverse fibrosis.

3. Origin of myofibroblasts in fibrotic tissues

The origin of the myofibroblasts during fibrosis, and the relative contribution of myofibroblasts from each source to fibrosis, is a matter of on-going debate (Fig. 2). It was originally thought that the activation or proliferation of local resident stromal cells and their differentiation into myofibroblasts was the only source of myofibroblasts during fibrosis [2833]. In healthy tissue fibroblasts are quiescent and are primarily involved in routine maintenance of the ECM during homeostasis. During both physiological and pathological wound repair the fibroblast is activated and differentiates into a myofibroblast [31,34,35]. Transforming growth factor-β1 (TGF-β1) continues to be regarded as the key growth factor involved in driving fibrosis [36] and can drive fibroblast to myofibroblast differentiation both in vitro and in vivo [37,38]. In addition to TGF-β1, a range of cytokines and growth factors have been shown to drive myofibroblast differentiation including IL-4, IL-13, and platelet derived growth factor (PDGF) among others [3947].

Fig. 2.

Fig. 2

Proposed origins of myofibroblasts in fibrosis. It was originally thought that the activation or proliferation of local resident stromal cells and their differentiation in to myofibroblasts was the only source of myofibroblasts during fibrosis. However it is also now widely believed that myofibroblasts are derived from at least four other sources; through the recruitment and differentiation of fibrocytes, through the activation and proliferation of pericytes, via epithelial to mesenchymal transition (EMT) or via endothelial to mesenchymal transition (EnMT).

It is also now widely believed that myofibroblasts could be derived from at least four other sources [4850]. Fibrocytes were originally described as fibroblast like, peripheral cells that migrate into regions of tissue injury [51]. They express fibroblast specific proteins as well as the hematopoietic stem cell marker (CD34) and the leukocyte common antigen (CD45). Fibrocytes migrate to wound sites in response to different chemokine signals including secondary lymphoid chemokine (CCL21) and stromal cell-derived factor (CXCL12) [5254].

Over time the expression of CD34 and CD45 is reduced and the cells differentiate into myofibroblasts [55,56]. Fibrocyte differentiation is regulated by multiple soluble mediators such as IL-4, IL-13 and PDGF [52,55]. There is significant literature indicating that fibrocytes are associated with organ fibrosis in pulmonary fibrosis, bronchial asthma, skin wounds, intimal hyperplasia and kidney fibrosis [49,52,54,5760].

Epithelial to mesenchymal transition (EMT) describes the transdifferentiation of an epithelial cell to a cell with myofibroblast-like features. During EMT epithelial cells downregulate epithelial marker expression, upregulate mesenchymal markers expression and gain functional characteristics of mesenchymal cells [50,61,62]. TGF-β1 continues to be regarded as the masterswitch regulating fibrosis [6365] and EMT driven by TGF-β1 has been suggested to play a role in fibrosis in multiple organs [6668]. The ability of other cytokines to drive EMT directly remains more controvertial. For example there is conflicting data reqarding the ability of TNFα to drive EMT in the absence of TGF-β1 [62,6973]. However, there is compelling evidence that cytokines including TNFα and Il-1β are able to accentuate TGF-β1 driven EMT in a range of cell types [71,7378]. However, recently the contribution of myofibroblasts derived via EMT to fibrosis has been questioned due to contradicting reports using lineage tracing models in mice [68,7991] (for a more comprehensive assessment of the role of EMT in fibrosis please see Epithelial injury and lung repair – Harold Chapman, Renal epithelial injury – Wilhelm Kriz, Epithelium ER stress as a fibrotic stimulus – Timothy Blackwell also in this special issue).

More recently endothelial to mesenchymal transition (EnMT) has been suggested as a potential source of myofibroblasts during wound healing and fibrosis. EnMT was originally thought to be a phenomenon confined to embryonic development but in 2007, Zeisberg et al. published evidence suggesting that up to 35% of fibroblasts present in the fibrotic myocardium of mice with aortic banding originated from endothelial cells [92]. Consequently several other studies have suggested an important role for EnMT in cardiac, renal and pulmonary fibrosis [9398]. Like EMT, EnMT can be driven by TGF-β1 and TGF-β2 [92] and accentuated by cytokines such as TNFα and IL-1β [99101].

Pericytes are cells of mesenchymal origin that are intimately involved in the development, maturation, stabilisation and remodelling of the vasculature during homeostasis and angiogenesis [102]. Recently Lin and colleagues identified that pericytes are closely associated with the vasculature in normal kidney cortex and medulla. However in response to injury these pericytes detach from the vasculature, rapidly up regulate collagen production and α-smooth muscle actin (α-SMA) expression, migrate into the interstitial space and increase myofibroblast numbers [103]. A subsequent study adopting a genetic fate mapping approach to label pericytes (as well as mesangial cells and smooth muscle cells) identified that in a range of kidney injury models the number of labelled cells increased approximately 15 fold in 2–3 weeks strongly suggesting an important role for pericytes in fibrosis in the kidney [87]. Consequently a role for pericytes as a precursor for myofibroblasts has been proposed in other tissues including the spinal cord, lung, skin and skeletal muscle [91,104107].

The challenge remains to effectively identify the quantity of myofibroblasts derived from each of the above sources in a fibrotic organ and determining the relative contribution of each to disease pathology to improve our understanding of the mechanisms driving fibrosis and allow the development of novel therapeutic targets.

4. TGF-β dependent and independent fibrosis

TGF-β is the most extensively studied molecule in fibrosis. There are three TGF-β isoforms (TGF-β1–3); all have similar biological activity, although each isoform is expressed in a distinct pattern under control of a unique promoter [108,109]. Although a wide variety of cell types produce and respond to TGF-β it is TGF-β1 that has been primarily linked to tissue fibrosis [110]. TGF-β1 is released from cells in a latent complex formed by binding to latency-associated protein (LAP), which holds TGF-β1 in an inactive state. To achieve an active state, TGF-β1 must dissociate from LAP, a process that can be catalysed by a range of agents including cathepsins, plasmin, calpain, thrombospondin, matrix metalloproteinases (MMPs) and integrins [109,111114] (for a more comprehensive assessment of the role of integrin biology please see Integrin biology and ECM interactions – Dean Sheppard also in this special issue). Once activated TGF-β1 has been shown to signal primarily via heteromeric complexes of type II and type I serine/threonine kinase receptors which activate the SMAD signalling pathway, a homolog of the mothers against decapentaplegic drosophila proteins, and modulates the transcription of important target genes including pro-collagen I (COL1A1) and pro-collagen III (COL3A1) [115118].

Numerous animal models have demonstrated an important role for TGF-β in the pathogenesis of fibrotic conditions [38,119122]. For example TGF-β inhibition attenuated hepatic, renal and cardiac fibrosis in various animal models [123125]. Extensive evidence suggests that the canonical TGF-β type I receptor (ALK5)/Smad3 pathway is critically involved in the pathogenesis of fibrosis driven by TGF-β in many tissues. For example, the oral administration of an inhibitor of the kinase activity of ALK5 inhibited fibrosis in a rat model of TGF-β1-induced pulmonary fibrosis [126] and Smad3 null mice show attenuated fibrosis in bleomycin induced pulmonary fibrosis, renal interstitial fibrosis and cardiac fibrosis [127130].

However, not all fibrosis is dependent on TGF-β1 and several Smad3/TGF-β1 independent mechanisms of fibrosis have been described in the lung and other tissues [131133] suggesting that other mediators can act separately from the Smad3/TGF-β1 pathway.

5. Th2 cytokines in fibrosis

The T-helper 2 (Th2) cytokines IL-4 and IL-13 share many biological functions as both exploit the same IL-4Rα/Stat6 signalling pathway [134]; for example IL-4 and IL-13 can drive the differentiation of resident fibroblast and recruited fibrocytes to myofibroblast in a range of tissues [39,41,4345]. However the development of IL-13 transgenic mice and knockout mice, as well as IL-13 specific antagonists, has revealed unique and non-redundant roles for IL-4 and IL-13 in vivo [135138].

Although the contribution of IL-4 to fibrosis varies in different diseases it is considered a potent fibrotic mediator with one study suggesting that IL-4 is nearly twice as effective as TGF-β in inducing collagen synthesis from human skin derived fibroblasts [139]. One of the first in vivo reports to investigate the contribution of IL-4 in fibrosis was a study of Schistosomiasis in mice, in which neutralising antibodies to IL-4 were shown to significantly reduce the development of hepatic fibrosis [12]. Subsequently inhibitors of IL-4 were also found to reduce dermal fibrosis in a chronic skin graft rejection model and in a mouse model of scleroderma [140,141]. In addition IL-4 is found at increased levels in BAL fluids of patients with IPF, in the pulmonary interstitium of individuals with cryptogenic fibrosing alveolitis and in peripheral blood mononuclear cells (PBMCs) of patients suffering from periportal fibrosis of the liver [142144].

When IL-4 and IL-13 were inhibited independently, IL-13 was identified as the dominant effector cytokine of fibrosis in several experimental models [135,145149]. For example the overexpression of IL-13 in the lung triggered significant subepithelial airway fibrosis in mice in the absence of any other inflammatory stimulus [137]. In contrast, despite developing an intense inflammatory phenotype, the overexpression of IL-4 in the lung was not associated with evidence of subepithelial fibrosis [150]. Anti-IL-13 treatment has been shown to markedly reduce collagen deposition in the lungs of animals challenged with Aspergillus fumigatus conidia [146]. In schistosomiasis, collagen deposition was decreased by more than 85% following IL-13 blockade although the egg-induced inflammatory response was maintained, including no attenuation of IL-4 production [135,151,152]. IL-13 binds to two primary receptor chains, IL-13Rα1, which also binds IL-4 and thus accounts for the functional overlap of the cytokines, and IL13Rα2 [138,153]. IL-13Rα2 is generally considered to be a decoy receptor for IL-13 since it has a short cytoplasmic tail which is devoid of signalling activity [154]. IL-13Rα2 binds IL-13 with four orders of magnitude higher affinity and specificity than IL-13Rα1 [155] and is believed to exert an inhibitory function by blocking the formation of functional IL-13-IL13Rα1 complexes [138]. In agreement with this, mice lacking the IL-13Rα2 decoy receptor have enhanced IL-13 activity [156]. When infected with Schistosoma mansoni the IL-13Rα2-deficient mice had significantly increased liver fibrosis despite no change in the inflammatory response [157] suggesting that IL-13Rα2 directly inhibits the ECM-remodelling activity of IL-13. Soluble IL-13Rα2-Fc is a highly effective inhibitor of IL-13 that has been shown to ameliorate the progression of established fibrotic disease [135,138,151,158] suggesting that modulation of the IL-13 signalling pathway may be a viable therapeutic target in fibrosis.

Macrophages demonstrate remarkable plasticity and change their physiology in response to the microenvironment [159]. Interestingly, selective depletion of macrophages in a model of liver fibrosis revealed distinct populations of macrophages associated with both injury and recovery phases of inflammatory scarring [160]. IL-4 and IL-13 promote the transition of resident macrophages into M2 or alternatively activated macrophages (AAMϕ). In vitro and in vivo studies in mice have shown that this phenotype is characterised by elevated expression of the mannose receptor (CD206), Ym1, Relm-α, (also known as FIZZ-1), major histocompatibility complex class II antigens and arginase-1 [161]. Expression of arginase-1 by AAMϕ is of particular interest because this enzyme controls L-proline production, which is required for collagen synthesis by activated myofibroblasts [162]. AAMϕ have also been implicated in the development of Th2 effector responses, production of fibrogenic cytokines and recruitment of fibrocytes [163,164] leading to suggestions that AAMϕ are important inducers of wound healing and fibrosis. However a recently published study employing LysMCreIL-4Rα−/flox mice, in which Cre-mediated recombination results in deletion of the IL-4Rα chain in the myeloid cell lineage and therefore macrophages cannot recognise IL-4 or IL-13, demonstrated that egg-induced granulomas and liver fibrosis developed normally in the absence of AAMϕ following infection with Schistosoma mansoni [165]. Surprisingly depleting arginase-1 activity specifically in AAMϕ exacerbated the development of liver fibrosis and increased the Th2 immune response suggested that AAMϕ are required for the suppression and resolution of fibrosis [166]. The data suggest that AAMϕ may compete with myofibroblasts for L-arginine which is required for collagen synthesis and could be exploited to ameliorate fibrotic disease. However first it will be important to investigate if AAMϕ have similar inhibitory roles in other models of fibrosis.

IL-5 may also play an important role in fibrosis through the recruitment, differentiation and activation of eosinophils. IL-5 and eosinophils have been observed in a variety of diseases including skin allograft rejection and pulmonary fibrosis and eosinophils are an important source of pro-fibrotic cytokines and growth factors such as TGF-β1 and IL-13 [141,167,168]. Anti-IL-5 and IL-5 gene deletion have been shown to suppress eosinophilia and remodelling in murine models of allergic asthma [169171] and decreased granuloma size in chronic Schistosoma mansoni infection [172]. However several other studies have failed to show a reduction in fibrosis in liver, skin and lung raising doubts about the importance of IL-5 and eosinophils in disease [173175]. A recent study by Huaux et al. paradoxically demonstrated that IL-5−/− mice are not protected from bleomycin induced pulmonary fibrosis but that excessive amounts of IL-5 can exacerbate bleomycin induced pulmonary fibrosis [175] suggesting that IL-5 may be accentuating rather than driving fibrosis.

IL-10 is a multifunctional cytokine with diverse effects on most hemopoietic cell types that was first identified for its ability to inhibit the activation and effector function of T cells, monocytes, and macrophages. The primary function of IL-10 appears to be to limit and ultimately terminate inflammatory responses [176]. In agreement with a role as a suppressive cytokine, IL-10 deficient animals show significantly more severe hepatic and pancreatic fibrosis in response to challenge with carbon tetrachloride (CCL4) and cerulein respectively and mice treated with endogenous IL-10 develop significantly less liver, lung and pancreatic fibrosis [177180]. This provides supportive evidence for the important role that pro-inflammatory inflammation can play in fibrogenesis. One potential mechanism of action for IL-10 may be via the direct inhibition of collagen synthesis and secretion from fibroblasts. For example culturing human scar derived fibroblasts with IL-10 induced a decrease in type1 pro-collagen protein and mRNA and the addition of anti-IL-10 to cultured hepatic stellate cells caused enhanced collagen production under basal or stimulated condition [181,182]. Consequently some success in clinical studies have been reported including a reduction in serum alanine aminotransferase (ALT), hepatic inflammation and a reduction in fibrosis score in chronic hepatitis C patients treated with IL-10 [183].

6. IL-17A and its role in fibrosis

Th17 cells are a subset of CD4+ T-helper cells that differ from Th1 and Th2 cells in development and function and are characterised by the production of their signature cytokine IL-17. Differentiation of Th17 cells requires the combined actions of TGF-β, IL-6, and IL-21 in mice, whereas IL-6 and IL-21 can be replaced by IL-23 or IL-1β in humans [184186]. These cytokines induce the expression of the orphan nuclear receptor RORγt that is the key transcription factor that orchestrates the differentiation of this effector cell lineage [187]. Once established the expression of IL-23 is required for stabilisation and expansion of these cells in vivo [188,189]. The development of Th17 cells can be suppressed by IFNγ, IL-2, IL-27 and IL-4 [190194].

Il-17 is recognised as an inflammatory cytokine that exerts its function mainly on myeloid cells, epithelial cells and mesenchymal cells to induce the expression of a range of cytokines and chemokines, which in turn increase granulopoiesis and recruitment of leukocytes, mainly neutrophils, to the site of inflammation [195,196]. IL-17A antibody neutralisation reduced neutrophil influx during the early lung response to silica particles and endotoxin exposure [197,198] and intratracheal instillation of human recombinant IL-17 selectively recruited neutrophils into rat airways [199,200]. IL-17A expression is associated with the persistent neutrophilia observed in a variety of diseases including bacterial pneumonia and cystic fibrosis in the lung, acute lesions in atopic dermatitis and in renal allografts during acute rejection where the number of IL-17 positive cells are independent predictors of worse graft outcome [201205]. IL-17 has also been shown to be elevated in the BAL of patients with IPF, with the recruitment of neutrophils to the BAL an important predictor of early mortality in IPF patients [206,207].

Several studies have suggested a possible contribution for IL-17A in the development of chronic fibroproliferative diseases [187]. For example Th17 cytokines are increased during the development of bleomycin induced skin fibrosis and IL-17A promotes the development of dilated cardiomyopathy, with blockade of IL-17A attenuating myocarditis-induced cardiac fibrosis and ameliorating ventricular function [208,209]. Several studies have also revealed that the development of hepatic granulomas in mice infected with Schistosoma mansoni is in part dependent on Th17 responses and that treatment with IL-17 neutralising antibodies significantly reduces granuloma formation in some strains of mice [210212]. IL-17A has also been shown to be important for the development of pulmonary fibrosis after exposure to bleomycin. Detailed mechanistic studies in mice with bleomycin-induced fibrosis suggested that bleomycin-induced IL-17A production is also highly dependent on TGF-β1 signalling, and recombinant IL-17A-driven fibrosis is dependent on the downstream profibrotic activity of TGF-β1, suggesting co-dependent roles for IL-17A and TGF-β1 in the development of pulmonary fibrosis [207].

The aforementioned data suggest that targeting components of the IL-17A signaling pathway is a potential strategy for the development of novel therapeutic agents against fibroproliferative diseases.

7. Th1 cytokines in fibrosis

Numerous experimental models of fibrosis have documented potent anti-fibrotic functions of the archetypical Th-1 cytokine IFNγ. For example IFNγ inhibits the activation and proliferation of hepatic stellate cells (HSC) and subsequent ECM deposition in a rat model of liver fibrosis induced by dimethylnitrosamine [213]. In addition IFNγ treatment ameliorates bleomycin induced lung fibrosis and reduced glomerulosclerosis and tubulointerstitial fibrosis in the rat subtotal nephrectomy model [213,214]. Mechanistically, IFNγ is believed to inhibit fibrosis by antagonising the pro-fibrotic activity of TGF-β1. TGF-β1 induced phosphorylation of Smad3 and its subsequent translocation to the nucleus is inhibited by IFNγ resulting in the decreased activation of TGF-β1 responsive genes. In addition, acting through Janus-associated kinase (Jak1) and Stat1, IFNγ induces the expression of Smad7, an antagonistic SMAD, which prevents the interaction of Smad3 with the TGF-β receptor, further attenuating TGF-β-induced signalling [215]. IFNγ can also directly inhibit fibroblast proliferation, TGF-β1 induced expression of the genes encoding procollagen I and procollagen III, and collagen synthesis in activated myofibroblasts as well as inhibiting the Th2 cytokine induced differentiation of fibrocytes into myofibroblasts [216,217]. Similar antifibrotic activity has been reported for IL-12, primarily via its ability to stimulate IFNγ production in Th1 cells. In schistosomiasis, treatment with recombinant IL-12 significantly reduced collagen deposition associated with chronic granuloma formation, while having no effect on the establishment of infection [218] and IL-12 treatment caused a significant reduction in the hydroxyproline content of the lung in the bleomycin mouse model of lung fibrosis [219].

However despite compelling evidence supporting an antifibrotic role for IFNγ both in vitro and in vivo, clinical studies employing the use of IFNγ have generated conflicting results. Positively, IFNγ treatment reduces liver fibrosis progression in people chronically infected with HCV [220]. In contrast, large randomised placebo controlled clinical trials in patients with IPF have revealed that treatment with IFNγ did not significantly improve survival, lung function, gas exchange, or the quality of life. In addition more patients in the IFNγ group had constitutional signs and symptoms (influenza-like illness, fatigue, fever, and chills) than those on placebo [221,222].

The M1 or classically activated macrophages (CAMϕ) are produced during cell mediated responses and are a vital component of host defence. Such macrophage activation depends on IFNγ, a cytokine network involving IL-12 and exposure to microbial products. These macrophages secrete high levels of pro-inflammatory cytokines including TNFα and IL-1β and their activation must be tightly controlled because the cytokines and mediators they produce can lead to host-tissue damage [223]. TNFα and IL-1β have been identified as important targets in a variety of fibrotic conditions including IPF and asbestosis [224,225] and the overexpression of either TNFα or IL-1β in the lungs of mice leads to spontaneous pulmonary fibrosis [226,227]. Studies have subsequently identified that TNFα is essential for the development of bleomycin and silica induced pulmonary fibrosis, CCL4 induced hepatic fibrosis and non-alcoholic steatohepatitis in mice [228231]. Recently clinical trials employing inhibitors of the TNFα pathway such as etanercept and infliximab have been initiated to evaluate the potential clinical benefit for the treatment of IPF and other fibrotic diseases. One study in IPF reported that etanercept was well tolerated and showed a non-significant reduction in disease progression in several physiologic, functional, and quality-of-life endpoints [232].

Additionally, several studies have documented profibrotic activity for IL-1β in pulmonary fibrosis induced by bleomycin and silica, liver fibrosis in hypercholesterolemic mice, renal interstitial fibrosis resulting from unilateral ureteric obstruction and cardiovascular fibrosis after myocardial infarction [233235]. IL-1β was found to be increased in the BAL of patients with IPF and acute respiratory distress syndrome (ARDS), with persistent elevation predicting poor outcome [207,236]. Recent studies have shown IL-1β driven pulmonary fibrosis to be dependent on IL-17A [207,237]. IL-1β has also been shown to drive EMT and myofibroblast differentiation via a TGF-β1 dependent mechanism, confirming that it functions as a potent upstream driver of fibrosis [238]. In addition, IL-1β and TNFα have been demonstrated to accentuate TGF-β1 driven EMT and EnMT [71,73,74,76,99101] highlighting another potential mechanism by which IL-1β and TNFα drive fibrosis.

8. Fibrosis – lessons from lung transplantation

The ability to investigate the role of the immune system in early fibrotic disease is limited as patients often present with established fibrosis and significant loss of organ function already. However there is a condition where it is possible to study the fibrotic remodelling process very early in disease and even before it has begun. Obliterative bronchiolitis (OB) affects 50% of lung transplant recipients limiting survival to a median of approximately 5 years and provides a valid human model of chronic inflammatory airway disease leading to dramatic fibrotic remodelling and loss of lung function over a short time course. The rate of disease progression, as measured by reduction in forced expiratory volume in 1 second (FEV1), can be ten times that seen in other chronic progressive inflammatory airways diseases such as chronic obstructive pulmonary disease [239]. Two decades of research of this condition have identified important mechanisms linking inflammation, the immune response and the development of fibrosis post lung transplant.

Several studies highlight an important role for innate immunity, PAMPs and PRR in the fibrotic remodeling seen in OB. For example, the acquisition of Pseudomonas aeruginosa in the transplanted airway is associated with an increased risk of developing in OB [16,240,241]. In addition, lung transplant patients with loss of function polymorphisms in TLR4 demonstrate significantly less acute rejection and a trend towards reduced severity of OB [242]. Similarly, patients with gain of function polymorphisms in CD14, which binds LPS and promotes signaling through TLR4, develop OB earlier after transplant and demonstrate increased OB related deaths. Interestingly, patients with a gain of function polymorphism in CD14 also have significantly greater TNFα and IFNγ in the peripheral blood implying a heightened state of innate immune activation drives the development of increased post-transplant rejection [243].

There is growing interest in the role of the macrophage as an effector cell in allograft injury and fibrosis. In the murine heterotopic tracheal transplant model depletion of recipient macrophages significantly abrogates obliteration of the transplanted airway [244]. Furthermore airway macrophages isolated from post-transplant patients secrete increased levels of pro-inflammatory cytokines compared to control patients [245,246] leading to an elevated expression of a variety of acute inflammatory cytokines in the BAL of patients with OB including TNFα, IL-1β and IL-8 [247,248]. It has been demonstrated that there is an early elevation in Th1-cytokines in lung transplant patients who developed OB compared to stable recipients and normal control subjects [249]. Furthermore, CD4+ T cells in patients who developed OB are of a Th1-phenotype suggesting that the microenvironment within the lung allograft may skew the immune response towards a Th1 phenotype that predisposes to OB [250].

Several studies have shown that the neo-macrolide azithromycin given at sub-minimum inhibitory concentration for respiratory pathogens can reverse the decline in lung function in some patients with OB [251,252]. Furthermore a randomised placebo controlled study demonstrating that patients receiving azithromycin after lung transplantation had a lower incidence of OB compared with those receiving placebo in their first two years post transplantation [253]. Neo-macrolides are a group of antibiotics that are bacteriostatic and only bactericidal at high concentrations. Independently of their antimicrobial activity, macrolides possess immunomodulatory properties that may contribute to clinical benefits observed in patients with OB. The mechanism of action was initially believed to be through a reduction in airway neutrophilia and IL-8 in the lung [254]. However, azithromycin has recently been shown to modulate inflammation by shifting macrophage polarisation towards an AAMϕ phenotype identifying another possible mechanism of action [255].

The development of OB in a mouse model is associated with a significant elevation in TNFα levels at the onset of fibrosis [256] and neutralising antibodies to TNFα prevents the development of OB in this model [257]. Similar effects have also been reported in OB in rat tracheal allografts [258] and in a heterotopic porcine bronchial transplantation model [259]. However, to date there have been no reported trials of using biological agents targeting TNFα in OB post lung transplantation, although there is a single case report indicating improvement in wellbeing and FEV1 after Infliximab therapy in a child who developed OB following bone marrow transplantation [260]. Lung transplant recipients who go on to develop OB also showed an elevation in IL-17 compared to stable lung transplant recipients [247] and neutralising IL-17 prevented OB in the heterotopic tracheal transplant model in mice [261]. To date no studies have reported investigated the efficacy of anti-IL-17 therapies in OB.

TGF-β1 is present in elevated levels in the BAL of patients with OB [262] and blocking TGF-β1 or its downstream signalling inhibits OB in the heterotopic tracheal transplant model [263,264]. Mechanistically, bronchial epithelial cells isolated from stable lung transplant recipients undergo EMT when exposed to TGF-β1 [62] and this can be accentuated by the addition of TNFα or IL-1β or by co-culturing the cells with a Pseudomonas aeruginosa activated macrophage cell line [78,265]. In addition co-localisation of both epithelial (E-cadherin) and mesenchymal (α-SMA) markers in epithelial cells of post-transplant patients has been reported [62] highlighting EMT as one potential source of myofibroblasts in the development of OB. As well as an elevated level of TGF-β1, it has also been reported that IL-13 is elevated and biologically active in BAL during the development of BOS. Furthermore translational studies using a mouse model of OB showed that neutralisation of IL-13 reduced airway allograft matrix deposition and OB [148]. Both TGF-β1 and IL-13 can drive the differentiation of resident fibroblasts to myofibroblasts identifying a likely second source of myofibroblasts in OB. Finally, a higher proportion of circulating fibrocytes was measured in patients with OB Grade ≥ 1 than in those with OB Grade 0 (p) [266] highlighting a possible role for fibrocytes in allograft rejection. In agreement with this, inhibiting CXCL12 blocks fibrocyte migration and differentiation and attenuates OB in the murine heterotopic tracheal transplant model [267].

The aforementioned data highlight the complex nature of fibrosis in the transplant lung and the large number of potential therapeutic targets to limit disease progression. However, due to the ability to accurately identify patients that are likely to develop the disease before they present with clinical symptoms and the rapid decline in FEV1 as an experimental indication of disease progression, the development of OB after lung transplant could also be a valuable and cost effective tool for testing novel therapeutic strategies in the development of fibrosis.

9. Conclusion

A review of the literature pertaining to the role of cytokines in fibrosis highlights the wide range of functions a single cytokine can perform on numerous cell types and provides the possibility that targeting a single cytokine may provide a way of blocking/reversing at least some of the fibrotic process in disease. However the literature also tells us that a wide number of cytokines can perform several very similar functions and therefore compensate for the loss of another. How do we choose which cytokine/cytokines to target as potential therapeutics? And are these targets going to work universally or will there be organ specific and disease specific roles identified? Given the diverse importance for Th1, Th17 and Th2 cytokines described in the literature in driving fibrosis in different organs, and indeed different diseases in the same organ, it seem unlikely that the treatment of fibrosis will be universal or organ specific and instead will likely be disease specific. Depending on the stage of disease when a patient is diagnosed, the treatment approach may also be tailored. For example if a patient presents early, i.e. with progressive fibrosis, the inhibition of ECM production is an obvious target to limit the development of fibrosis. However if a patient presents late in disease, i.e. with established fibrosis, the resolution of already deposited ECM is critical. Given the vast number of potential therapeutic targets and strategies it is important that a well-defined and considered approach to translating the wealth of experimental knowledge into clinically beneficial therapies is applied. The slow progression of many fibrotic diseases makes clinical trials expensive and prohibitive. Therefore quantitative clinical endpoints such as serum bio-markers and imaging techniques to accurately measure the rate of disease progression are desperately needed. The burden of diseases where inflammation and fibrosis plays an important role continues to grow and therefore the need for safe and effective anti-fibrotic therapies is great and is also likely to increase.

Acknowledgments

We would like to express our sincere appreciation and thanks to all of our colleagues, both past and present, for their guidance and support. We also thank Aaron Gardner for assistance with figure preparation. LAB is supported by a Marie Curie international outgoing fellowship from the European Union Framework Programme 7. TAW is supported by the Intramural Research Program of the NIH/NIAID.

Footnotes

This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.

References

  • 1.Gardner A, Borthwick LA, Fisher AJ. Lung epithelial wound healing in health and disease. Expert Rev Respir Med. 2010;4:647–660. doi: 10.1586/ers.10.62. [DOI] [PubMed] [Google Scholar]
  • 2.Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028–1040. doi: 10.1038/nm.2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Wynn TA. Fibrotic disease and the T (H)1/T (H)2 paradigm. Nat Rev Immunol. 2004;4:583–594. doi: 10.1038/nri1412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Marchiando AM, Graham WV, Turner JR. Epithelial barriers in homeostasis and disease. Annu Rev Pathol. 2010;5:119–144. doi: 10.1146/annurev.pathol.4.110807.092135. [DOI] [PubMed] [Google Scholar]
  • 5.Hogaboam CM, Blease K, Mehrad B, Steinhauser ML, Standiford TJ, Kunkel SL, Lukacs NW. Chronic airway hyperreactivity, goblet cell hyperplasia, and peribronchial fibrosis during allergic airway disease induced by Aspergillus fumigatus. Am J Pathol. 2000;156:723–732. doi: 10.1016/S0002-9440(10)64775-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cosma CL, Sherman DR, Ramakrishnan L. The secret lives of the pathogenic mycobacteria. Annu Rev Microbiol. 2003;57:641–676. doi: 10.1146/annurev.micro.57.030502.091033. [DOI] [PubMed] [Google Scholar]
  • 7.Meneghin A, Hogaboam CM. Infectious disease, the innate immune response, and fibrosis. J Clin Invest. 2007;117:530–538. doi: 10.1172/JCI30595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schaaf B, Wieghorst A, Aries SP, Dalhoff K, Braun J. Neutrophil inflammation and activation in bronchiectasis: comparison with pneumonia and idiopathic pulmonary fibrosis. Respiration. 2000;67:52–59. doi: 10.1159/000029463. [DOI] [PubMed] [Google Scholar]
  • 9.Kleiner DE. The liver biopsy in chronic hepatitis C: a view from the other side of the microscope. Semin Liver Dis. 2005;25:52–64. doi: 10.1055/s-2005-864781. [DOI] [PubMed] [Google Scholar]
  • 10.Glanville AR, Scott AI, Morton JM, Aboyoun CL, Plit ML, Carter IW, Malouf MA. Intravenous ribavirin is a safe and cost-effective treatment for respiratory syncytial virus infection after lung transplantation. J Heart Lung Transplant. 2005;24:2114–2119. doi: 10.1016/j.healun.2005.06.027. [DOI] [PubMed] [Google Scholar]
  • 11.Arora S, Hernandez Y, Erb-Downward JR, McDonald RA, Toews GB, Huffnagle GB. Role of IFN-gamma in regulating T2 immunity and the development of alternatively activated macrophages during allergic bronchopulmonary mycosis. J Immunol. 2005;174:6346–6356. doi: 10.4049/jimmunol.174.10.6346. [DOI] [PubMed] [Google Scholar]
  • 12.Cheever AW, Williams ME, Wynn TA, Finkelman FD, Seder RA, Cox TM, Hieny S, Caspar P, Sher A. Anti-IL-4 treatment of Schistosoma mansoni-infected mice inhibits development of T cells and non-B, non-T cells expressing Th2 cytokines while decreasing egg-induced hepatic fibrosis. J Immunol. 1994;153:753–759. [PubMed] [Google Scholar]
  • 13.Noel W, Raes G, Hassanzadeh Ghassabeh G, De Baetselier P, Beschin A. Alternatively activated macrophages during parasite infections. Trends Parasitol. 2004;20:126–133. doi: 10.1016/j.pt.2004.01.004. [DOI] [PubMed] [Google Scholar]
  • 14.Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499–511. doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
  • 15.Chtarbanova S, Imler JL. Microbial sensing by Toll receptors: a historical perspective. Arterioscler Thromb Vasc Biol. 2011;31:1734–1738. doi: 10.1161/ATVBAHA.108.179523. [DOI] [PubMed] [Google Scholar]
  • 16.Botha P, Archer L, Anderson RL, Lordan J, Dark JH, Corris PA, Gould K, Fisher AJ. Pseudomonas aeruginosa colonization of the allograft after lung transplantation and the risk of bronchiolitis obliterans syndrome. Transplantation. 2008;85:771–774. doi: 10.1097/TP.0b013e31816651de. [DOI] [PubMed] [Google Scholar]
  • 17.Thung SN. Histologic findings in recurrent HBV. Liver Transpl. 2006;12:S50–S53. doi: 10.1002/lt.20946. [DOI] [PubMed] [Google Scholar]
  • 18.Martinez-Hernandez A, Amenta PS. The extracellular matrix in hepatic regeneration. FASEB J. 1995;9:1401–1410. doi: 10.1096/fasebj.9.14.7589981. [DOI] [PubMed] [Google Scholar]
  • 19.Holt PG, Sly PD. Interaction between adaptive and innate immune pathways in the pathogenesis of atopic asthma: operation of a lung/bone marrow axis. Chest. 2011;139:1165–1171. doi: 10.1378/chest.10-2397. [DOI] [PubMed] [Google Scholar]
  • 20.Piccinini AM, Midwood KS. DAMPening inflammation by modulating TLR signalling. Mediat Inflamm. 2010;2010:21. doi: 10.1155/2010/672395. http://dx.doi.org/10.1155/2010/672395 (Article ID 672395) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Stavitsky AB. The innate immune response to infection, toxins and trauma evolved into networks of interactive, defensive, reparative, regulatory, injurious and pathogenic pathways. Mol Immunol. 2007;44:2787–2799. doi: 10.1016/j.molimm.2007.01.011. [DOI] [PubMed] [Google Scholar]
  • 22.Marvie P, Lisbonne M, L’Helgoualc’h A, Rauch M, Turlin B, Preisser L, Bourd-Boittin K, Theret N, Gascan H, Piquet-Pellorce C, Samson M. Interleukin-33 overexpression is associated with liver fibrosis in mice and humans. J Cell Mol Med. 2010;14:1726–1739. doi: 10.1111/j.1582-4934.2009.00801.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yanaba K, Yoshizaki A, Asano Y, Kadono T, Sato S. Serum IL-33 levels are raised in patients with systemic sclerosis: association with extent of skin sclerosis and severity of pulmonary fibrosis. Clin Rheumatol. 2011;30:825–830. doi: 10.1007/s10067-011-1686-5. [DOI] [PubMed] [Google Scholar]
  • 24.Hamada N, Maeyama T, Kawaguchi T, Yoshimi M, Fukumoto J, Yamada M, Yamada S, Kuwano K, Nakanishi Y. The role of high mobility group box1 in pulmonary fibrosis. Am J Respir Cell Mol Biol. 2008;39:440–447. doi: 10.1165/rcmb.2007-0330OC. [DOI] [PubMed] [Google Scholar]
  • 25.Otte JM, Rosenberg IM, Podolsky DK. Intestinal myofibroblasts in innate immune responses of the intestine. Gastroenterology. 2003;124:1866–1878. doi: 10.1016/s0016-5085(03)00403-7. [DOI] [PubMed] [Google Scholar]
  • 26.Coelho AL, Hogaboam CM, Kunkel SL. Chemokines provide the sustained inflammatory bridge between innate and acquired immunity. Cytokine Growth Factor Rev. 2005;16:553–560. doi: 10.1016/j.cytogfr.2005.03.004. [DOI] [PubMed] [Google Scholar]
  • 27.Meneghin A, Choi ES, Evanoff HL, Kunkel SL, Martinez FJ, Flaherty KR, Toews GB, Hogaboam CM. TLR9 is expressed in idiopathic interstitial pneumonia and its activation promotes in vitro myofibroblast differentiation. Histochem Cell Biol. 2008;130:979–992. doi: 10.1007/s00418-008-0466-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Furness PN. Extracellular matrix and the kidney. J Clin Pathol. 1996;49:355–359. doi: 10.1136/jcp.49.5.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol. 2008;214:199–210. doi: 10.1002/path.2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wynn TA. Common and unique mechanisms regulate fibrosis in various fibroproliferative diseases. J Clin Invest. 2007;117:524–529. doi: 10.1172/JCI31487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Moreira RK. Hepatic stellate cells and liver fibrosis. Arch Pathol Lab Med. 2007;131:1728–1734. doi: 10.5858/2007-131-1728-HSCALF. [DOI] [PubMed] [Google Scholar]
  • 32.Gressner OA, Weiskirchen R, Gressner AM. Evolving concepts of liver fibrogenesis provide new diagnostic and therapeutic options. Comp Hepatol. 2007;6:7. doi: 10.1186/1476-5926-6-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G. The myofibroblast: one function, multiple origins. Am J Pathol. 2007;170:1807–1816. doi: 10.2353/ajpath.2007.070112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Desmouliere A, Darby IA, Gabbiani G. Normal and pathologic soft tissue remodeling: role of the myofibroblast, with special emphasis on liver and kidney fibrosis. Lab Invest. 2003;83:1689–1707. doi: 10.1097/01.lab.0000101911.53973.90. [DOI] [PubMed] [Google Scholar]
  • 35.Hinz B. Formation and function of the myofibroblast during tissue repair. J Invest Dermatol. 2007;127:526–537. doi: 10.1038/sj.jid.5700613. [DOI] [PubMed] [Google Scholar]
  • 36.Border WA, Noble NA. Transforming growth factor beta in tissue fibrosis. N Engl J Med. 1994;331:1286–1292. doi: 10.1056/NEJM199411103311907. [DOI] [PubMed] [Google Scholar]
  • 37.Desmouliere A, Geinoz A, Gabbiani F, Gabbiani G. Transforming growth factor-beta 1 induces alpha-smooth muscle actin expression in granulation tissue myofibroblasts and in quiescent and growing cultured fibroblasts. J Cell Biol. 1993;122:103–111. doi: 10.1083/jcb.122.1.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J. Adenovector-mediated gene transfer of active transforming growth factor-beta1 induces prolonged severe fibrosis in rat lung. J Clin Invest. 1997;100:768–776. doi: 10.1172/JCI119590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Mattey DL, Dawes PT, Nixon NB, Slater H. Transforming growth factor beta 1 and interleukin 4 induced alpha smooth muscle actin expression and myofibroblast-like differentiation in human synovial fibroblasts in vitro: modulation by basic fibroblast growth factor. Ann Rheum Dis. 1997;56:426–431. doi: 10.1136/ard.56.7.426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tang WW, Ulich TR, Lacey DL, Hill DC, Qi M, Kaufman SA, Van GY, Tarpley JE, Yee JS. Platelet-derived growth factor-BB induces renal tubulointerstitial myofibroblast formation and tubulointerstitial fibrosis. Am J Pathol. 1996;148:1169–1180. [PMC free article] [PubMed] [Google Scholar]
  • 41.Salmon-Ehr V, Serpier H, Nawrocki B, Gillery P, Clavel C, Kalis B, Birembaut P, Maquart FX. Expression of interleukin-4 in scleroderma skin specimens and scleroderma fibroblast cultures. Potential role in fibrosis. Arch Dermatol. 1996;132:802–806. [PubMed] [Google Scholar]
  • 42.Lund PK, Zimmermann EM. Insulin-like growth factors and inflammatory bowel disease. Baillieres Clin Gastroenterol. 1996;10:83–96. doi: 10.1016/s0950-3528(96)90041-x. [DOI] [PubMed] [Google Scholar]
  • 43.Doucet C, Brouty-Boye D, Pottin-Clemenceau C, Canonica GW, Jasmin C, Azzarone B. Interleukin (IL) 4 and IL-13 act on human lung fibroblasts. Implication in asthma. J Clin Invest. 1998;101:2129–2139. doi: 10.1172/JCI741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hashimoto S, Gon Y, Takeshita I, Maruoka S, Horie T. IL-4 and IL-13 induce myofibroblastic phenotype of human lung fibroblasts through c-Jun NH2-terminal kinase-dependent pathway. J Allergy Clin Immunol. 2001;107:1001–1008. doi: 10.1067/mai.2001.114702. [DOI] [PubMed] [Google Scholar]
  • 45.Saito A, Okazaki H, Sugawara I, Yamamoto K, Takizawa H. Potential action of IL-4 and IL-13 as fibrogenic factors on lung fibroblasts in vitro. Int Arch Allergy Immunol. 2003;132:168–176. doi: 10.1159/000073718. [DOI] [PubMed] [Google Scholar]
  • 46.Brenzel A, Gressner AM. Characterization of insulin-like growth factor (IGF)-I-receptor binding sites during in vitro transformation of rat hepatic stellate cells to myofibroblasts. Eur J Clin Chem Clin Biochem. 1996;34:401–409. doi: 10.1515/cclm.1996.34.5.401. [DOI] [PubMed] [Google Scholar]
  • 47.Oh SJ, Kurz H, Christ B, Wilting J. Platelet-derived growth factor-B induces transformation of fibrocytes into spindle-shaped myofibroblasts in vivo. Histochem Cell Biol. 1998;109:349–357. doi: 10.1007/s004180050235. [DOI] [PubMed] [Google Scholar]
  • 48.Piera-Velazquez S, Li Z, Jimenez SA. Role of endothelial–mesenchymal transition (EndoMT) in the pathogenesis of fibrotic disorders. Am J Pathol. 2011;179:1074–1080. doi: 10.1016/j.ajpath.2011.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Quan TE, Cowper SE, Bucala R. The role of circulating fibrocytes in fibrosis. Curr Rheumatol Rep. 2006;8:145–150. doi: 10.1007/s11926-006-0055-x. [DOI] [PubMed] [Google Scholar]
  • 50.Kalluri R, Neilson EG. Epithelial–mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003;112:1776–1784. doi: 10.1172/JCI20530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bucala R, Spiegel LA, Chesney J, Hogan M, Cerami A. Circulating fibrocytes define a new leukocyte subpopulation that mediates tissue repair. Mol Med. 1994;1:71–81. [PMC free article] [PubMed] [Google Scholar]
  • 52.Abe R, Donnelly SC, Peng T, Bucala R, Metz CN. Peripheral blood fibrocytes: differentiation pathway and migration to wound sites. J Immunol. 2001;166:7556–7562. doi: 10.4049/jimmunol.166.12.7556. [DOI] [PubMed] [Google Scholar]
  • 53.Chesney J, Bucala R. Peripheral blood fibrocytes: novel fibroblast-like cells that present antigen and mediate tissue repair. Biochem Soc Trans. 1997;25:520–524. doi: 10.1042/bst0250520. [DOI] [PubMed] [Google Scholar]
  • 54.Phillips RJ, Burdick MD, Hong K, Lutz MA, Murray LA, Xue YY, Belperio JA, Keane MP, Strieter RM. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J Clin Invest. 2004;114:438–446. doi: 10.1172/JCI20997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Aiba S, Tagami H. Inverse correlation between CD34 expression and proline-4-hydroxylase immunoreactivity on spindle cells noted in hypertrophic scars and keloids. J Cutan Pathol. 1997;24:65–69. doi: 10.1111/j.1600-0560.1997.tb01098.x. [DOI] [PubMed] [Google Scholar]
  • 56.Mori L, Bellini A, Stacey MA, Schmidt M, Mattoli S. Fibrocytes contribute to the myofibroblast population in wounded skin and originate from the bone marrow. Exp Cell Res. 2005;304:81–90. doi: 10.1016/j.yexcr.2004.11.011. [DOI] [PubMed] [Google Scholar]
  • 57.Wada T, Sakai N, Matsushima K, Kaneko S. Fibrocytes: a new insight into kidney fibrosis. Kidney Int. 2007;72:269–273. doi: 10.1038/sj.ki.5002325. [DOI] [PubMed] [Google Scholar]
  • 58.Schmidt M, Sun G, Stacey MA, Mori L, Mattoli S. Identification of circulating fibrocytes as precursors of bronchial myofibroblasts in asthma. J Immunol. 2003;171:380–389. doi: 10.4049/jimmunol.171.1.380. [DOI] [PubMed] [Google Scholar]
  • 59.Yang L, Scott PG, Giuffre J, Shankowsky HA, Ghahary A, Tredget EE. Peripheral blood fibrocytes from burn patients: identification and quantification of fibrocytes in adherent cells cultured from peripheral blood mononuclear cells. Lab Invest. 2002;82:1183–1192. doi: 10.1097/01.lab.0000027841.50269.61. [DOI] [PubMed] [Google Scholar]
  • 60.Varcoe RL, Mikhail M, Guiffre AK, Pennings G, Vicaretti M, Hawthorne WJ, Fletcher JP, Medbury HJ. The role of the fibrocyte in intimal hyperplasia. J Thromb Haemost. 2006;4:1125–1133. doi: 10.1111/j.1538-7836.2006.01924.x. [DOI] [PubMed] [Google Scholar]
  • 61.Jain R, Shaul PW, Borok Z, Willis BC. Endothelin-1 induces alveolar epithelial–mesenchymal transition through endothelin type A receptor-mediated production of TGF-beta1. Am J Respir Cell Mol Biol. 2007;37:38–47. doi: 10.1165/rcmb.2006-0353OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Borthwick LA, Parker SM, Brougham KA, Johnson GE, Gorowiec MR, Ward C, Lordan JL, Corris PA, Kirby JA, Fisher AJ. Epithelial to mesenchymal transition (EMT) and airway remodelling after human lung transplantation. Thorax. 2009;64:770–777. doi: 10.1136/thx.2008.104133. [DOI] [PubMed] [Google Scholar]
  • 63.Yamamoto T, Nakamura T, Noble NA, Ruoslahti E, Border WA. Expression of transforming growth factor beta is elevated in human and experimental diabetic nephropathy. Proc Natl Acad Sci U S A. 1993;90:1814–1818. doi: 10.1073/pnas.90.5.1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Czaja MJ, Weiner FR, Flanders KC, Giambrone MA, Wind R, Biempica L, Zern MA. In vitro and in vivo association of transforming growth factor-beta 1 with hepatic fibrosis. J Cell Biol. 1989;108:2477–2482. doi: 10.1083/jcb.108.6.2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Broekelmann TJ, Limper AH, Colby TV, McDonald JA. Transforming growth factor beta 1 is present at sites of extracellular matrix gene expression in human pulmonary fibrosis. Proc Natl Acad Sci U S A. 1991;88:6642–6646. doi: 10.1073/pnas.88.15.6642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Guarino M, Tosoni A, Nebuloni M. Direct contribution of epithelium to organ fibrosis: epithelial–mesenchymal transition. Hum Pathol. 2009;40:1365–1376. doi: 10.1016/j.humpath.2009.02.020. [DOI] [PubMed] [Google Scholar]
  • 67.Liu Y. New insights into epithelial–mesenchymal transition in kidney fibrosis. J Am Soc Nephrol. 2010;21:212–222. doi: 10.1681/ASN.2008121226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Zeisberg M, Yang C, Martino M, Duncan MB, Rieder F, Tanjore H, Kalluri R. Fibroblasts derive from hepatocytes in liver fibrosis via epithelial to mesenchymal transition. J Biol Chem. 2007;282:23337–23347. doi: 10.1074/jbc.M700194200. [DOI] [PubMed] [Google Scholar]
  • 69.Chuang MJ, Sun KH, Tang SJ, Deng MW, Wu YH, Sung JS, Cha TL, Sun GH. Tumor-derived tumor necrosis factor-alpha promotes progression and epithelial–mesenchymal transition in renal cell carcinoma cells. Cancer Sci. 2008;99:905–913. doi: 10.1111/j.1349-7006.2008.00756.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Grund EM, Kagan D, Tran CA, Zeitvogel A, Starzinski-Powitz A, Nataraja S, Palmer SS. Tumor necrosis factor-alpha regulates inflammatory and mesenchymal responses via mitogen-activated protein kinase kinase, p38, and nuclear factor kappaB in human endometriotic epithelial cells. Mol Pharmacol. 2008;73:1394–1404. doi: 10.1124/mol.107.042176. [DOI] [PubMed] [Google Scholar]
  • 71.Camara J, Jarai G. Epithelial–mesenchymal transition in primary human bronchial epithelial cells is Smad-dependent and enhanced by fibronectin and TNF-alpha. Fibrogenesis Tissue Repair. 2010 Jan 5;3(1):2. doi: 10.1186/1755-1536-3-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kasai H, Allen JT, Mason RM, Kamimura T, Zhang Z. TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT) Respir Res. 2005;6:56. doi: 10.1186/1465-9921-6-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Yamauchi Y, Kohyama T, Takizawa H, Kamitani S, Desaki M, Takami K, Kawasaki S, Kato J, Nagase T. Tumor necrosis factor-alpha enhances both epithelial–mesenchymal transition and cell contraction induced in A549 human alveolar epithelial cells by transforming growth factor-beta1. Exp Lung Res. 2010 Feb;36(1):12–24. doi: 10.3109/01902140903042589. [DOI] [PubMed] [Google Scholar]
  • 74.Liu X. Inflammatory cytokines augments TGF-beta1-induced epithelial–mesenchymal transition in A549 cells by up-regulating TbetaR-I. Cell Motil Cytoskeleton. 2008;65:935–944. doi: 10.1002/cm.20315. [DOI] [PubMed] [Google Scholar]
  • 75.Sullivan NJ, Sasser AK, Axel AE, Vesuna F, Raman V, Ramirez N, Oberyszyn TM, Hall BM. Interleukin-6 induces an epithelial–mesenchymal transition phenotype in human breast cancer cells. Oncogene. 2009;28:2940–2947. doi: 10.1038/onc.2009.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Doerner AM, Zuraw BL. TGF-beta1 induced epithelial to mesenchymal transition (EMT) in human bronchial epithelial cells is enhanced by IL-1beta but not abrogated by corticosteroids. Respir Res. 2009;10:100. doi: 10.1186/1465-9921-10-100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fernando RI, Castillo MD, Litzinger M, Hamilton DH, Palena C. IL-8 signaling plays a critical role in the epithelial–mesenchymal transition of human carcinoma cells. Cancer Res. 2011;71:5296–5306. doi: 10.1158/0008-5472.CAN-11-0156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Borthwick LA, McIlroy EI, Gorowiec MR, Brodlie M, Johnson GE, Ward C, Lordan JL, Corris PA, Kirby JA, Fisher AJ. Inflammation and epithelial to mesenchymal transition in lung transplant recipients: role in dysregulated epithelial wound repair. Am J Transplant. 2010;10:498–509. doi: 10.1111/j.1600-6143.2009.02953.x. [DOI] [PubMed] [Google Scholar]
  • 79.Zeisberg M, Duffield JS. Resolved: EMT produces fibroblasts in the kidney. J Am Soc Nephrol. 2010;21:1247–1253. doi: 10.1681/ASN.2010060616. [DOI] [PubMed] [Google Scholar]
  • 80.Higgins DF, Kimura K, Bernhardt WM, Shrimanker N, Akai Y, Hohenstein B, Saito Y, Johnson RS, Kretzler M, Cohen CD, Eckardt KU, Iwano M, Haase VH. Hypoxia promotes fibrogenesis in vivo via HIF-1 stimulation of epithelial-to-mesenchymal transition. J Clin Invest. 2007;117:3810–3820. doi: 10.1172/JCI30487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Flier SN, Tanjore H, Kokkotou EG, Sugimoto H, Zeisberg M, Kalluri R. Identification of epithelial to mesenchymal transition as a novel source of fibroblasts in intestinal fibrosis. J Biol Chem. 2010;285:20202–20212. doi: 10.1074/jbc.M110.102012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest. 2002;110:341–350. doi: 10.1172/JCI15518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA. Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci U S A. 2006;103:13180–13185. doi: 10.1073/pnas.0605669103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Kim KK, Wei Y, Szekeres C, Kugler MC, Wolters PJ, Hill ML, Frank JA, Brumwell AN, Wheeler SE, Kreidberg JA, Chapman HA. Epithelial cell alpha3beta1 integrin links beta-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J Clin Invest. 2009;119:213–224. doi: 10.1172/JCI36940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tanjore H, Xu XC, Polosukhin VV, Degryse AL, Li B, Han W, Sherrill TP, Plieth D, Neilson EG, Blackwell TS, Lawson WE. Contribution of epithelial-derived fibroblasts to bleomycin-induced lung fibrosis. Am J Respir Crit Care Med. 2009;180:657–665. doi: 10.1164/rccm.200903-0322OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zeisberg M, Kalluri R. Fibroblasts emerge via epithelial–mesenchymal transition in chronic kidney fibrosis. Front Biosci. 2008;13:6991–6998. doi: 10.2741/3204. [DOI] [PubMed] [Google Scholar]
  • 87.Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, Valerius MT, McMahon AP, Duffield JS. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol. 2010;176:85–97. doi: 10.2353/ajpath.2010.090517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chase LG, Ulloa-Montoya F, Kidder BL, Verfaillie CM. Islet-derived fibroblast-like cells are not derived via epithelial–mesenchymal transition from Pdx-1 or insulin-positive cells. Diabetes. 2007;56:3–7. doi: 10.2337/db06-1165. [DOI] [PubMed] [Google Scholar]
  • 89.Taura K, Miura K, Iwaisako K, Osterreicher CH, Kodama Y, Penz-Osterreicher M, Brenner DA. Hepatocytes do not undergo epithelial–mesenchymal transition in liver fibrosis in mice. Hepatology. 2010;51:1027–1036. doi: 10.1002/hep.23368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Le Hir M, Hegyi I, Cueni-Loffing D, Loffing J, Kaissling B. Characterization of renal interstitial fibroblast-specific protein 1/S100A4-positive cells in healthy and inflamed rodent kidneys. Histochem Cell Biol. 2005;123:335–346. doi: 10.1007/s00418-005-0788-z. [DOI] [PubMed] [Google Scholar]
  • 91.Rock JR, Barkauskas CE, Cronce MJ, Xue Y, Harris JR, Liang J, Noble PW, Hogan BL. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc Natl Acad Sci U S A. 2011;108:E1475–E1483. doi: 10.1073/pnas.1117988108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Sayegh MH, Izumo S, Kalluri R. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007;13:952–961. doi: 10.1038/nm1613. [DOI] [PubMed] [Google Scholar]
  • 93.Hashimoto N, Phan SH, Imaizumi K, Matsuo M, Nakashima H, Kawabe T, Shimokata K, Hasegawa Y. Endothelial–mesenchymal transition in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 2010;43:161–172. doi: 10.1165/rcmb.2009-0031OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol. 2008;19:2282–2287. doi: 10.1681/ASN.2008050513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Widyantoro B, Emoto N, Nakayama K, Anggrahini DW, Adiarto S, Iwasa N, Yagi K, Miyagawa K, Rikitake Y, Suzuki T, Kisanuki YY, Yanagisawa M, Hirata K. Endothelial cell-derived endothelin-1 promotes cardiac fibrosis in diabetic hearts through stimulation of endothelial-to-mesenchymal transition. Circulation. 2010;121:2407–2418. doi: 10.1161/CIRCULATIONAHA.110.938217. [DOI] [PubMed] [Google Scholar]
  • 96.Li J, Qu X, Yao J, Caruana G, Ricardo SD, Yamamoto Y, Yamamoto H, Bertram JF. Blockade of endothelial–mesenchymal transition by a Smad3 inhibitor delays the early development of streptozotocin-induced diabetic nephropathy. Diabetes. 2010;59:2612–2624. doi: 10.2337/db09-1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Li J, Qu X, Bertram JF. Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol. 2009;175:1380–1388. doi: 10.2353/ajpath.2009.090096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zeisberg EM, Potenta S, Xie L, Zeisberg M, Kalluri R. Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts. Cancer Res. 2007;67:10123–10128. doi: 10.1158/0008-5472.CAN-07-3127. [DOI] [PubMed] [Google Scholar]
  • 99.Maleszewska M, Moonen JR, Huijkman N, van de Sluis B, Krenning G, Harmsen MC. IL-1beta and TGFbeta2 synergistically induce endothelial to mesenchymal transition in an NFkappaB-dependent manner. Immunobiology. 2012 doi: 10.1016/j.imbio.2012.05.026. [DOI] [PubMed] [Google Scholar]
  • 100.Rieder F, Kessler SP, West GA, Bhilocha S, de la Motte C, Sadler TM, Gopalan B, Stylianou E, Fiocchi C. Inflammation-induced endothelial-to-mesenchymal transition: a novel mechanism of intestinal fibrosis. Am J Pathol. 2011;179:2660–2673. doi: 10.1016/j.ajpath.2011.07.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Dai H, Huang H, Wang SL, Xu X, Jian Y, Cui WH, Zhang M, Zhang B, Jiang JX. Role of tumor necrosis factor alpha in endothelial–mesenchymal transition in vitro. Zhonghua Shao Shang Za Zhi. 2012;28:19–24. [PubMed] [Google Scholar]
  • 102.Schrimpf C, Duffield JS. Mechanisms of fibrosis: the role of the pericyte. Curr Opin Nephrol Hypertens. 2011;20:297–305. doi: 10.1097/MNH.0b013e328344c3d4. [DOI] [PubMed] [Google Scholar]
  • 103.Lin SL, Kisseleva T, Brenner DA, Duffield JS. Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol. 2008;173:1617–1627. doi: 10.2353/ajpath.2008.080433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mellgren AM, Smith CL, Olsen GS, Eskiocak B, Zhou B, Kazi MN, Ruiz FR, Pu WT, Tallquist MD. Platelet-derived growth factor receptor beta signaling is required for efficient epicardial cell migration and development of two distinct coronary vascular smooth muscle cell populations. Circ Res. 2008;103:1393–1401. doi: 10.1161/CIRCRESAHA.108.176768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Murphy MM, Lawson JA, Mathew SJ, Hutcheson DA, Kardon G. Satellite cells, connective tissue fibroblasts and their interactions are crucial for muscle regeneration. Development. 2011;138:3625–3637. doi: 10.1242/dev.064162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Liu S, Taghavi R, Leask A. Connective tissue growth factor is induced in bleomycin-induced skin scleroderma. J Cell Commun Signal. 2010;4:25–30. doi: 10.1007/s12079-009-0081-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Goritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisen J. A pericyte origin of spinal cord scar tissue. Science. 2011;333:238–242. doi: 10.1126/science.1203165. [DOI] [PubMed] [Google Scholar]
  • 108.Attisano L, Wrana JL. Signal transduction by members of the transforming growth factor-beta superfamily. Cytokine Growth Factor Rev. 1996;7:327–339. doi: 10.1016/s1359-6101(96)00042-1. [DOI] [PubMed] [Google Scholar]
  • 109.Letterio JJ, Roberts AB. Regulation of immune responses by TGF-beta. Annu Rev Immunol. 1998;16:137–161. doi: 10.1146/annurev.immunol.16.1.137. [DOI] [PubMed] [Google Scholar]
  • 110.Ask K, Bonniaud P, Maass K, Eickelberg O, Margetts PJ, Warburton D, Groffen J, Gauldie J, Kolb M. Progressive pulmonary fibrosis is mediated by TGF-beta isoform 1 but not TGF-beta3. Int J Biochem Cell Biol. 2008;40:484–495. doi: 10.1016/j.biocel.2007.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell biology. Nat Rev Immunol. 2002;2:46–53. doi: 10.1038/nri704. [DOI] [PubMed] [Google Scholar]
  • 112.Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J, Pittet JF, Kaminski N, Garat C, Matthay MA, Rifkin DB, Sheppard D. 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. doi: 10.1016/s0092-8674(00)80545-0. [DOI] [PubMed] [Google Scholar]
  • 113.Jenkins RG, Su X, Su G, Scotton CJ, Camerer E, Laurent GJ, Davis GE, Chambers RC, Matthay MA, Sheppard D. Ligation of protease-activated receptor 1 enhances alpha (v) beta6 integrin-dependent TGF-beta activation and promotes acute lung injury. J Clin Invest. 2006;116:1606–1614. doi: 10.1172/JCI27183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H, Sheppard D, Broaddus VC, Nishimura SL. The integrin alpha (v) beta8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-beta1. J Cell Biol. 2002;157:493–507. doi: 10.1083/jcb.200109100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Roberts AB, Russo A, Felici A, Flanders KC. Smad3: a key player in pathogenetic mechanisms dependent on TGF-beta. Ann N Y Acad Sci. 2003;995:1–10. doi: 10.1111/j.1749-6632.2003.tb03205.x. [DOI] [PubMed] [Google Scholar]
  • 116.Flanders KC. Smad3 as a mediator of the fibrotic response. Int J Exp Pathol. 2004;85:47–64. doi: 10.1111/j.0959-9673.2004.00377.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 2003;425:577–584. doi: 10.1038/nature02006. [DOI] [PubMed] [Google Scholar]
  • 118.Valcourt U, Kowanetz M, Niimi H, Heldin CH, Moustakas A. TGF-beta and the Smad signaling pathway support transcriptomic reprogramming during epithelial–mesenchymal cell transition. Mol Biol Cell. 2005;16:1987–2002. doi: 10.1091/mbc.E04-08-0658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Leask A, Abraham DJ. TGF-beta signaling and the fibrotic response. FASEB J. 2004;18:816–827. doi: 10.1096/fj.03-1273rev. [DOI] [PubMed] [Google Scholar]
  • 120.Pohlers D, Brenmoehl J, Loffler I, Muller CK, Leipner C, Schultze-Mosgau S, Stallmach A, Kinne RW, Wolf G. TGF-beta and fibrosis in different organs —molecular pathway imprints. Biochim Biophys Acta. 2009;1792:746–756. doi: 10.1016/j.bbadis.2009.06.004. [DOI] [PubMed] [Google Scholar]
  • 121.Biernacka A, Dobaczewski M, Frangogiannis NG. TGF-beta signaling in fibrosis. Growth Factors. 2011;29:196–202. doi: 10.3109/08977194.2011.595714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sanderson N, Factor V, Nagy P, Kopp J, Kondaiah P, Wakefield L, Roberts AB, Sporn MB, Thorgeirsson SS. Hepatic expression of mature transforming growth factor beta 1 in transgenic mice results in multiple tissue lesions. Proc Natl Acad Sci U S A. 1995;92:2572–2576. doi: 10.1073/pnas.92.7.2572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Teekakirikul P, Eminaga S, Toka O, Alcalai R, Wang L, Wakimoto H, Nayor M, Konno T, Gorham JM, Wolf CM, Kim JB, Schmitt JP, Molkentin JD, Norris RA, Tager AM, Hoffman SR, Markwald RR, Seidman CE, Seidman JG. Cardiac fibrosis in mice with hypertrophic cardiomyopathy is mediated by non-myocyte proliferation and requires Tgf-beta. J Clin Invest. 2010;120:3520–3529. doi: 10.1172/JCI42028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Fukasawa H, Yamamoto T, Suzuki H, Togawa A, Ohashi N, Fujigaki Y, Uchida C, Aoki M, Hosono M, Kitagawa M, Hishida A. Treatment with anti-TGF-beta antibody ameliorates chronic progressive nephritis by inhibiting Smad/TGF-beta signaling. Kidney Int. 2004;65:63–74. doi: 10.1111/j.1523-1755.2004.00393.x. [DOI] [PubMed] [Google Scholar]
  • 125.Nakamura T, Sakata R, Ueno T, Sata M, Ueno H. Inhibition of transforming growth factor beta prevents progression of liver fibrosis and enhances hepatocyte regeneration in dimethylnitrosamine-treated rats. Hepatology. 2000;32:247–255. doi: 10.1053/jhep.2000.9109. [DOI] [PubMed] [Google Scholar]
  • 126.Bonniaud P, Margetts PJ, Kolb M, Schroeder JA, Kapoun AM, Damm D, Murphy A, Chakravarty S, Dugar S, Higgins L, Protter AA, Gauldie J. Progressive transforming growth factor beta1-induced lung fibrosis is blocked by an orally active ALK5 kinase inhibitor. Am J Respir Crit Care Med. 2005;171:889–898. doi: 10.1164/rccm.200405-612OC. [DOI] [PubMed] [Google Scholar]
  • 127.Zhao J, Shi W, Wang YL, Chen H, Bringas P, Jr, Datto MB, Frederick JP, Wang XF, Warburton D. Smad3 deficiency attenuates bleomycin-induced pulmonary fibrosis in mice. Am J Physiol Lung Cell Mol Physiol. 2002;282:L585–L593. doi: 10.1152/ajplung.00151.2001. [DOI] [PubMed] [Google Scholar]
  • 128.Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A. Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J Clin Invest. 2003;112:1486–1494. doi: 10.1172/JCI19270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bujak M, Ren G, Kweon HJ, Dobaczewski M, Reddy A, Taffet G, Wang XF, Frangogiannis NG. Essential role of Smad3 in infarct healing and in the pathogenesis of cardiac remodeling. Circulation. 2007;116:2127–2138. doi: 10.1161/CIRCULATIONAHA.107.704197. [DOI] [PubMed] [Google Scholar]
  • 130.Dobaczewski M, Chen W, Frangogiannis NG. Transforming growth factor (TGF)-beta signaling in cardiac remodeling. J Mol Cell Cardiol. 2011;51:600–606. doi: 10.1016/j.yjmcc.2010.10.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Ma LJ, Yang H, Gaspert A, Carlesso G, Barty MM, Davidson JM, Sheppard D, Fogo AB. Transforming growth factor-beta-dependent and -independent pathways of induction of tubulointerstitial fibrosis in beta6(−/−) mice. Am J Pathol. 2003;163:1261–1273. doi: 10.1016/s0002-9440(10)63486-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Kaviratne M, Hesse M, Leusink M, Cheever AW, Davies SJ, McKerrow JH, Wakefield LM, Letterio JJ, Wynn TA. IL-13 activates a mechanism of tissue fibrosis that is completely TGF-beta independent. J Immunol. 2004;173:4020–4029. doi: 10.4049/jimmunol.173.6.4020. [DOI] [PubMed] [Google Scholar]
  • 133.Ashcroft GS, Yang X, Glick AB, Weinstein M, Letterio JL, Mizel DE, Anzano M, Greenwell-Wild T, Wahl SM, Deng C, Roberts AB. Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response. Nat Cell Biol. 1999;1:260–266. doi: 10.1038/12971. [DOI] [PubMed] [Google Scholar]
  • 134.Zurawski SM, Vega F, Jr, Huyghe B, Zurawski G. Receptors for interleukin-13 and interleukin-4 are complex and share a novel component that functions in signal transduction. EMBO J. 1993;12:2663–2670. doi: 10.1002/j.1460-2075.1993.tb05927.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Chiaramonte MG, Donaldson DD, Cheever AW, Wynn TA. An IL-13 inhibitor blocks the development of hepatic fibrosis during a T-helper type 2-dominated inflammatory response. J Clin Invest. 1999;104:777–785. doi: 10.1172/JCI7325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.McKenzie GJ, Emson CL, Bell SE, Anderson S, Fallon P, Zurawski G, Murray R, Grencis R, McKenzie AN. Impaired development of Th2 cells in IL-13-deficient mice. Immunity. 1998;9:423–432. doi: 10.1016/s1074-7613(00)80625-1. [DOI] [PubMed] [Google Scholar]
  • 137.Zhu Z, Homer RJ, Wang Z, Chen Q, Geba GP, Wang J, Zhang Y, Elias JA. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Invest. 1999;103:779–788. doi: 10.1172/JCI5909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Donaldson DD, Whitters MJ, Fitz LJ, Neben TY, Finnerty H, Henderson SL, O’Hara RM, Jr, Beier DR, Turner KJ, Wood CR, Collins M. The murine IL-13 receptor alpha 2: molecular cloning, characterization, and comparison with murine IL-13 receptor alpha 1. J Immunol. 1998;161:2317–2324. [PubMed] [Google Scholar]
  • 139.Fertin C, Nicolas JF, Gillery P, Kalis B, Banchereau J, Maquart FX. Interleukin-4 stimulates collagen synthesis by normal and scleroderma fibroblasts in dermal equivalents. Cell Mol Biol. 1991;37:823–829. [PubMed] [Google Scholar]
  • 140.Ong C, Wong C, Roberts CR, Teh HS, Jirik FR. Anti-IL-4 treatment prevents dermal collagen deposition in the tight-skin mouse model of scleroderma. Eur J Immunol. 1998;28:2619–2629. doi: 10.1002/(SICI)1521-4141(199809)28:09<2619::AID-IMMU2619>3.0.CO;2-M. [DOI] [PubMed] [Google Scholar]
  • 141.Le Moine A, Flamand V, Demoor FX, Noel JC, Surquin M, Kiss R, Nahori MA, Pretolani M, Goldman M, Abramowicz D. Critical roles for IL-4, IL-5, and eosinophils in chronic skin allograft rejection. J Clin Invest. 1999;103:1659–1667. doi: 10.1172/JCI5504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Emura M, Nagai S, Takeuchi M, Kitaichi M, Izumi T. In vitro production of B cell growth factor and B cell differentiation factor by peripheral blood mononu-clear cells and bronchoalveolar lavage T lymphocytes from patients with idiopathic pulmonary fibrosis. Clin Exp Immunol. 1990;82:133–139. doi: 10.1111/j.1365-2249.1990.tb05416.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wallace WA, Ramage EA, Lamb D, Howie SE. A type 2 (Th2-like) pattern of immune response predominates in the pulmonary interstitium of patients with cryptogenic fibrosing alveolitis (CFA) Clin Exp Immunol. 1995;101:436–441. doi: 10.1111/j.1365-2249.1995.tb03131.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Booth M, Mwatha JK, Joseph S, Jones FM, Kadzo H, Ireri E, Kazibwe F, Kemijumbi J, Kariuki C, Kimani G, Ouma JH, Kabatereine NB, Vennervald BJ, Dunne DW. Periportal fibrosis in human Schistosoma mansoni infection is associated with low IL-10, low IFN-gamma, high TNF-alpha, or low RANTES, depending on age and gender. J Immunol. 2004;172:1295–1303. doi: 10.4049/jimmunol.172.2.1295. [DOI] [PubMed] [Google Scholar]
  • 145.Aliprantis AO, Wang J, Fathman JW, Lemaire R, Dorfman DM, Lafyatis R, Glimcher LH. Transcription factor T-bet regulates skin sclerosis through its function in innate immunity and via IL-13. Proc Natl Acad Sci U S A. 2007;104:2827–2830. doi: 10.1073/pnas.0700021104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Blease K, Jakubzick C, Westwick J, Lukacs N, Kunkel SL, Hogaboam CM. Therapeutic effect of IL-13 immunoneutralization during chronic experimental fungal asthma. J Immunol. 2001;166:5219–5224. doi: 10.4049/jimmunol.166.8.5219. [DOI] [PubMed] [Google Scholar]
  • 147.Kumar RK, Herbert C, Yang M, Koskinen AM, McKenzie AN, Foster PS. Role of interleukin-13 in eosinophil accumulation and airway remodelling in a mouse model of chronic asthma. Clin Exp Allergy. 2002;32:1104–1111. doi: 10.1046/j.1365-2222.2002.01420.x. [DOI] [PubMed] [Google Scholar]
  • 148.Keane MP, Gomperts BN, Weigt S, Xue YY, Burdick MD, Nakamura H, Zisman DA, Ardehali A, Saggar R, Lynch JP, III, Hogaboam C, Kunkel SL, Lukacs NW, Ross DJ, Grusby MJ, Strieter RM, Belperio JA. IL-13 is pivotal in the fibro-obliterative process of bronchiolitis obliterans syndrome. J Immunol. 2007;178:511–519. doi: 10.4049/jimmunol.178.1.511. [DOI] [PubMed] [Google Scholar]
  • 149.Kolodsick JE, Toews GB, Jakubzick C, Hogaboam C, Moore TA, McKenzie A, Wilke CA, Chrisman CJ, Moore BB. Protection from fluorescein isothiocyanate-induced fibrosis in IL-13-deficient, but not IL-4-deficient, mice results from impaired collagen synthesis by fibroblasts. J Immunol. 2004;172:4068–4076. doi: 10.4049/jimmunol.172.7.4068. [DOI] [PubMed] [Google Scholar]
  • 150.Rankin JA, Picarella DE, Geba GP, Temann UA, Prasad B, DiCosmo B, Tarallo A, Stripp B, Whitsett J, Flavell RA. Phenotypic and physiologic characterization of transgenic mice expressing interleukin 4 in the lung: lymphocytic and eosinophilic inflammation without airway hyperreactivity. Proc Natl Acad Sci U S A. 1996;93:7821–7825. doi: 10.1073/pnas.93.15.7821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Chiaramonte MG, Cheever AW, Malley JD, Donaldson DD, Wynn TA. Studies of murine schistosomiasis reveal interleukin-13 blockade as a treatment for established and progressive liver fibrosis. Hepatology. 2001;34:273–282. doi: 10.1053/jhep.2001.26376. [DOI] [PubMed] [Google Scholar]
  • 152.Fallon PG, Richardson EJ, McKenzie GJ, McKenzie AN. Schistosome infection of transgenic mice defines distinct and contrasting pathogenic roles for IL-4 and IL-13: IL-13 is a profibrotic agent. J Immunol. 2000;164:2585–2591. doi: 10.4049/jimmunol.164.5.2585. [DOI] [PubMed] [Google Scholar]
  • 153.Hilton DJ, Zhang JG, Metcalf D, Alexander WS, Nicola NA, Willson TA. Cloning and characterization of a binding subunit of the interleukin 13 receptor that is also a component of the interleukin 4 receptor. Proc Natl Acad Sci U S A. 1996;93:497–501. doi: 10.1073/pnas.93.1.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kelly-Welch AE, Hanson EM, Boothby MR, Keegan AD. Interleukin-4 and interleukin-13 signaling connections maps. Science. 2003;300:1527–1528. doi: 10.1126/science.1085458. [DOI] [PubMed] [Google Scholar]
  • 155.Lupardus PJ, Birnbaum ME, Garcia KC. Molecular basis for shared cytokine recognition revealed in the structure of an unusually high affinity complex between IL-13 and IL-13Ralpha2. Structure. 2010;18:332–342. doi: 10.1016/j.str.2010.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wood N, Whitters MJ, Jacobson BA, Witek J, Sypek JP, Kasaian M, Eppihimer MJ, Unger M, Tanaka T, Goldman SJ, Collins M, Donaldson DD, Grusby MJ. Enhanced interleukin (IL)-13 responses in mice lacking IL-13 receptor alpha 2. J Exp Med. 2003;197:703–709. doi: 10.1084/jem.20020906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Chiaramonte MG, Mentink-Kane M, Jacobson BA, Cheever AW, Whitters MJ, Goad ME, Wong A, Collins M, Donaldson DD, Grusby MJ, Wynn TA. Regulation and function of the interleukin 13 receptor alpha 2 during a T helper cell type 2-dominant immune response. J Exp Med. 2003;197:687–701. doi: 10.1084/jem.20020903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Taube C, Duez C, Cui ZH, Takeda K, Rha YH, Park JW, Balhorn A, Donaldson DD, Dakhama A, Gelfand EW. The role of IL-13 in established allergic airway disease. J Immunol. 2002;169:6482–6489. doi: 10.4049/jimmunol.169.11.6482. [DOI] [PubMed] [Google Scholar]
  • 159.Gordon S. Alternative activation of macrophages. Nat Rev. 2003;3:23–35. doi: 10.1038/nri978. [DOI] [PubMed] [Google Scholar]
  • 160.Duffield JS, Forbes SJ, Constandinou CM, Clay S, Partolina M, Vuthoori S, Wu S, Lang R, Iredale JP. Selective depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J Clin Invest. 2005;115:56–65. doi: 10.1172/JCI22675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010;30:245–257. doi: 10.1055/s-0030-1255354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Hesse M, Modolell M, La Flamme AC, Schito M, Fuentes JM, Cheever AW, Pearce EJ, Wynn TA. Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol. 2001;167:6533–6544. doi: 10.4049/jimmunol.167.11.6533. [DOI] [PubMed] [Google Scholar]
  • 163.Song E, Ouyang N, Horbelt M, Antus B, Wang M, Exton MS. Influence of alternatively and classically activated macrophages on fibrogenic activities of human fibroblasts. Cell Immunol. 2000;204:19–28. doi: 10.1006/cimm.2000.1687. [DOI] [PubMed] [Google Scholar]
  • 164.Sun L, Louie MC, Vannella KM, Wilke CA, LeVine AM, Moore BB, Shanley TP. New concepts of IL-10-induced lung fibrosis: fibrocyte recruitment and M2 activation in a CCL2/CCR2 axis. Am J Physiol Lung Cell Mol Physiol. 2011;300:L341–L353. doi: 10.1152/ajplung.00122.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Herbert DR, Holscher C, Mohrs M, Arendse B, Schwegmann A, Radwanska M, Leeto M, Kirsch R, Hall P, Mossmann H, Claussen B, Forster I, Brombacher F. Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology. Immunity. 2004;20:623–635. doi: 10.1016/s1074-7613(04)00107-4. [DOI] [PubMed] [Google Scholar]
  • 166.Pesce JT, Ramalingam TR, Mentink-Kane MM, Wilson MS, El Kasmi KC, Smith AM, Thompson RW, Cheever AW, Murray PJ, Wynn TA. Arginase-1-expressing macrophages suppress Th2 cytokine-driven inflammation and fibrosis. PLoS Pathog. 2009;5:e1000371. doi: 10.1371/journal.ppat.1000371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Gharaee-Kermani M, Phan SH. Lung interleukin-5 expression in murine bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 1997;16:438–447. doi: 10.1165/ajrcmb.16.4.9115755. [DOI] [PubMed] [Google Scholar]
  • 168.Ochkur SI, Jacobsen EA, Protheroe CA, Biechele TL, Pero RS, McGarry MP, Wang H, O’Neill KR, Colbert DC, Colby TV, Shen H, Blackburn MR, Irvin CC, Lee JJ, Lee NA. Coexpression of IL-5 and eotaxin-2 in mice creates an eosinophil-dependent model of respiratory inflammation with characteristics of severe asthma. J Immunol. 2007;178:7879–7889. doi: 10.4049/jimmunol.178.12.7879. [DOI] [PubMed] [Google Scholar]
  • 169.Trifilieff A, Fujitani Y, Coyle AJ, Kopf M, Bertrand C. IL-5 deficiency abolishes aspects of airway remodelling in a murine model of lung inflammation. Clin Exp Allergy. 2001;31:934–942. doi: 10.1046/j.1365-2222.2001.01084.x. [DOI] [PubMed] [Google Scholar]
  • 170.Blyth DI, Wharton TF, Pedrick MS, Savage TJ, Sanjar S. Airway subepithelial fibrosis in a murine model of atopic asthma: suppression by dexamethasone or anti-interleukin-5 antibody. Am J Respir Cell Mol Biol. 2000;23:241–246. doi: 10.1165/ajrcmb.23.2.3999. [DOI] [PubMed] [Google Scholar]
  • 171.Cho JY, Miller M, Baek KJ, Han JW, Nayar J, Lee SY, McElwain K, McElwain S, Friedman S, Broide DH. Inhibition of airway remodeling in IL-5-deficient mice. J Clin Invest. 2004;113:551–560. doi: 10.1172/JCI19133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Reiman RM, Thompson RW, Feng CG, Hari D, Knight R, Cheever AW, Rosenberg HF, Wynn TA. Interleukin-5 (IL-5) augments the progression of liver fibrosis by regulating IL-13 activity. Infect Immun. 2006;74:1471–1479. doi: 10.1128/IAI.74.3.1471-1479.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Sher A, Coffman RL, Hieny S, Scott P, Cheever AW. Interleukin 5 is required for the blood and tissue eosinophilia but not granuloma formation induced by infection with Schistosoma mansoni. Proc Natl Acad Sci U S A. 1990;87:61–65. doi: 10.1073/pnas.87.1.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Hao H, Cohen DA, Jennings CD, Bryson JS, Kaplan AM. Bleomycin-induced pulmonary fibrosis is independent of eosinophils. J Leukoc Biol. 2000;68:515–521. [PubMed] [Google Scholar]
  • 175.Huaux F, Liu T, McGarry B, Ullenbruch M, Xing Z, Phan SH. Eosinophils and T lymphocytes possess distinct roles in bleomycin-induced lung injury and fibrosis. J Immunol. 2003;171:5470–5481. doi: 10.4049/jimmunol.171.10.5470. [DOI] [PubMed] [Google Scholar]
  • 176.Moore KW, de Waal Malefyt R, Coffman RL, O’Garra A. Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19:683–765. doi: 10.1146/annurev.immunol.19.1.683. [DOI] [PubMed] [Google Scholar]
  • 177.Demols A, Van Laethem JL, Quertinmont E, Degraef C, Delhaye M, Geerts A, Deviere J. Endogenous interleukin-10 modulates fibrosis and regeneration in experimental chronic pancreatitis. Am J Physiol Gastrointest Liver Physiol. 2002;282:G1105–G1112. doi: 10.1152/ajpgi.00431.2001. [DOI] [PubMed] [Google Scholar]
  • 178.Louis H, Van Laethem JL, Wu W, Quertinmont E, Degraef C, Van den Berg K, Demols A, Goldman M, Le Moine O, Geerts A, Deviere J. Interleukin-10 controls neutrophilic infiltration, hepatocyte proliferation, and liver fibrosis induced by carbon tetrachloride in mice. Hepatology. 1998;28:1607–1615. doi: 10.1002/hep.510280621. [DOI] [PubMed] [Google Scholar]
  • 179.Thompson K, Maltby J, Fallowfield J, McAulay M, Millward-Sadler H, Sheron N. Interleukin-10 expression and function in experimental murine liver inflammation and fibrosis. Hepatology. 1998;28:1597–1606. doi: 10.1002/hep.510280620. [DOI] [PubMed] [Google Scholar]
  • 180.Arai T, Abe K, Matsuoka H, Yoshida M, Mori M, Goya S, Kida H, Nishino K, Osaki T, Tachibana I, Kaneda Y, Hayashi S. Introduction of the interleukin-10 gene into mice inhibited bleomycin-induced lung injury in vivo. Am J Physiol Lung Cell Mol Physiol. 2000;278:L914–L922. doi: 10.1152/ajplung.2000.278.5.L914. [DOI] [PubMed] [Google Scholar]
  • 181.Wang SC, Ohata M, Schrum L, Rippe RA, Tsukamoto H. Expression of interleukin-10 by in vitro and in vivo activated hepatic stellate cells. J Biol Chem. 1998;273:302–308. doi: 10.1074/jbc.273.1.302. [DOI] [PubMed] [Google Scholar]
  • 182.Wangoo A, Laban C, Cook HT, Glenville B, Shaw RJ. Interleukin-10- and corticosteroid-induced reduction in type I procollagen in a human ex vivo scar culture. Int J Exp Pathol. 1997;78:33–41. doi: 10.1046/j.1365-2613.1997.d01-241.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Nelson DR, Tu Z, Soldevila-Pico C, Abdelmalek M, Zhu H, Xu YL, Cabrera R, Liu C, Davis GL. Long-term interleukin 10 therapy in chronic hepatitis C patients has a proviral and anti-inflammatory effect. Hepatology. 2003;38:859–868. doi: 10.1053/jhep.2003.50427. [DOI] [PubMed] [Google Scholar]
  • 184.Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24:179–189. doi: 10.1016/j.immuni.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 185.Veldhoen M, Stockinger B. TGFbeta1, a “Jack of all trades”: the link with pro-inflammatory IL-17-producing T cells. Trends Immunol. 2006;27:358–361. doi: 10.1016/j.it.2006.06.001. [DOI] [PubMed] [Google Scholar]
  • 186.Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, Mattson JD, Basham B, Smith K, Chen T, Morel F, Lecron JC, Kastelein RA, Cua DJ, McClanahan TK, Bowman EP, de Waal Malefyt R. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol. 2007;8:950–957. doi: 10.1038/ni1497. [DOI] [PubMed] [Google Scholar]
  • 187.Ivanov BS, II, McKenzie L, Zhou CE, Tadokoro A, Lepelley JJ, Lafaille DJ, Cua DJ, Littman DR. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–1133. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
  • 188.Harrington LE, Mangan PR, Weaver CT. Expanding the effector CD4 T-cell repertoire: the Th17 lineage. Curr Opin Immunol. 2006;18:349–356. doi: 10.1016/j.coi.2006.03.017. [DOI] [PubMed] [Google Scholar]
  • 189.Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233–240. doi: 10.1084/jem.20041257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. Transforming growth factor-beta induces development of the T (H) 17 lineage. Nature. 2006;441:231–234. doi: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
  • 191.Batten M, Li J, Yi S, Kljavin NM, Danilenko DM, Lucas S, Lee J, de Sauvage FJ, Ghilardi N. Interleukin 27 limits autoimmune encephalomyelitis by suppressing the development of interleukin 17-producing T cells. Nat Immunol. 2006;7:929–936. doi: 10.1038/ni1375. [DOI] [PubMed] [Google Scholar]
  • 192.Stumhofer JS, Laurence A, Wilson EH, Huang E, Tato CM, Johnson LM, Villarino AV, Huang Q, Yoshimura A, Sehy D, Saris CJ, O’Shea JJ, Hennighausen L, Ernst M, Hunter CA. Interleukin 27 negatively regulates the development of interleukin 17-producing T helper cells during chronic inflammation of the central nervous system. Nat Immunol. 2006;7:937–945. doi: 10.1038/ni1376. [DOI] [PubMed] [Google Scholar]
  • 193.Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB, Meylan F, Siegel R, Hennighausen L, Shevach EM, O’Shea J. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. 2007;26:371–381. doi: 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 194.Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, Weaver CT. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:1123–1132. doi: 10.1038/ni1254. [DOI] [PubMed] [Google Scholar]
  • 195.Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity. 2008;28:454–467. doi: 10.1016/j.immuni.2008.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Dong C. Regulation and pro-inflammatory function of interleukin-17 family cytokines. Immunol Rev. 2008;226:80–86. doi: 10.1111/j.1600-065X.2008.00709.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Miyamoto M, Prause O, Sjostrand M, Laan M, Lotvall J, Linden A. Endogenous IL-17 as a mediator of neutrophil recruitment caused by endotoxin exposure in mouse airways. J Immunol. 2003;170:4665–4672. doi: 10.4049/jimmunol.170.9.4665. [DOI] [PubMed] [Google Scholar]
  • 198.Lo Re S, Dumoutier L, Couillin I, Van Vyve C, Yakoub Y, Uwambayinema F, Marien B, van den Brule S, Van Snick J, Uyttenhove C, Ryffel B, Renauld JC, Lison D, Huaux F. IL-17A-producing gammadelta T and Th17 lymphocytes mediate lung inflammation but not fibrosis in experimental silicosis. J Immunol. 2010;184:6367–6377. doi: 10.4049/jimmunol.0900459. [DOI] [PubMed] [Google Scholar]
  • 199.Laan M, Cui ZH, Hoshino H, Lotvall J, Sjostrand M, Gruenert DC, Skoogh BE, Linden A. Neutrophil recruitment by human IL-17 via C-X-C chemokine release in the airways. J Immunol. 1999;162:2347–2352. [PubMed] [Google Scholar]
  • 200.Hoshino H, Lotvall J, Skoogh BE, Linden A. Neutrophil recruitment by interleukin-17 into rat airways in vivo. Role of tachykinins. Am J Respir Crit Care Med. 1999;159:1423–1428. doi: 10.1164/ajrccm.159.5.9806008. [DOI] [PubMed] [Google Scholar]
  • 201.Ye P, Rodriguez FH, Kanaly S, Stocking KL, Schurr J, Schwarzenberger P, Oliver P, Huang W, Zhang P, Zhang J, Shellito JE, Bagby GJ, Nelson S, Charrier K, Peschon JJ, Kolls JK. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med. 2001;194:519–527. doi: 10.1084/jem.194.4.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Toda M, Leung DY, Molet S, Boguniewicz M, Taha R, Christodoulopoulos P, Fukuda T, Elias JA, Hamid QA. Polarized in vivo expression of IL-11 and IL-17 between acute and chronic skin lesions. J Allergy Clin Immunol. 2003;111:875–881. doi: 10.1067/mai.2003.1414. [DOI] [PubMed] [Google Scholar]
  • 203.Yapici U, Kers J, Bemelman FJ, Roelofs JJ, Groothoff JW, van der Loos CM, van Donselaarvan KA, der Pant MM, Idu N, ten Claessen IJ, Florquin Berge S. Interleukin-17 positive cells accumulate in renal allografts during acute rejection and are independent predictors of worse graft outcome. Transpl Int. 2011;24:1008–1017. doi: 10.1111/j.1432-2277.2011.01302.x. [DOI] [PubMed] [Google Scholar]
  • 204.Hsu E, Shi H, Jordan RM, Lyons-Weiler J, Pilewski JM, Feghali-Bostwick CA. Lung tissues in patients with systemic sclerosis have gene expression patterns unique to pulmonary fibrosis and pulmonary hypertension. Arthritis Rheum. 2011;63:783–794. doi: 10.1002/art.30159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Brodlie M, McKean MC, Johnson GE, Anderson AE, Hilkens CM, Fisher AJ, Corris PA, Lordan JL, Ward C. Raised interleukin-17 is immunolocalised to neutrophils in cystic fibrosis lung disease. Eur Respir J. 2011;37:1378–1385. doi: 10.1183/09031936.00067110. [DOI] [PubMed] [Google Scholar]
  • 206.Kinder BW, Brown KK, Schwarz MI, Ix JH, Kervitsky A, King TE., Jr Baseline BAL neutrophilia predicts early mortality in idiopathic pulmonary fibrosis. Chest. 2008;133:226–232. doi: 10.1378/chest.07-1948. [DOI] [PubMed] [Google Scholar]
  • 207.Wilson MS, Madala SK, Ramalingam TR, Gochuico BR, Rosas IO, Cheever AW, Wynn TA. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med. 2010;207:535–552. doi: 10.1084/jem.20092121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Yoshizaki A, Yanaba K, Iwata Y, Komura K, Ogawa A, Akiyama Y, Muroi E, Hara T, Ogawa F, Takenaka M, Shimizu K, Hasegawa M, Fujimoto M, Tedder TF, Sato S. Cell adhesion molecules regulate fibrotic process via Th1/Th2/Th17 cell balance in a bleomycin-induced scleroderma model. J Immunol. 2010;185:2502–2515. doi: 10.4049/jimmunol.0901778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Baldeviano GC, Barin JG, Talor MV, Srinivasan S, Bedja D, Zheng D, Gabrielson K, Iwakura Y, Rose NR, Cihakova D. Interleukin-17A is dispensable for myocarditis but essential for the progression to dilated cardiomyopathy. Circ Res. 2010;106:1646–1655. doi: 10.1161/CIRCRESAHA.109.213157. [DOI] [PubMed] [Google Scholar]
  • 210.Rutitzky LI, Bazzone L, Shainheit MG, Joyce-Shaikh B, Cua DJ, Stadecker MJ. IL-23 is required for the development of severe egg-induced immunopathology in schistosomiasis and for lesional expression of IL-17. J Immunol. 2008;180:2486–2495. doi: 10.4049/jimmunol.180.4.2486. [DOI] [PubMed] [Google Scholar]
  • 211.Rutitzky LI, Lopes da Rosa JR, Stadecker MJ. Severe CD4 T cell-mediated immunopathology in murine schistosomiasis is dependent on IL-12p40 and correlates with high levels of IL-17. J Immunol. 2005;175:3920–3926. doi: 10.4049/jimmunol.175.6.3920. [DOI] [PubMed] [Google Scholar]
  • 212.Rutitzky LI, Smith PM, Stadecker MJ. T-bet protects against exacerbation of schistosome egg-induced immunopathology by regulating Th17-mediated inflammation. Eur J Immunol. 2009;39:2470–2481. doi: 10.1002/eji.200939325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Baroni GS, D’Ambrosio L, Curto P, Casini A, Mancini R, Jezequel AM, Benedetti A. Interferon gamma decreases hepatic stellate cell activation and extracellular matrix deposition in rat liver fibrosis. Hepatology. 1996;23:1189–1199. doi: 10.1002/hep.510230538. [DOI] [PubMed] [Google Scholar]
  • 214.Oldroyd SD, Thomas GL, Gabbiani G, El Nahas AM. Interferon-gamma inhibits experimental renal fibrosis. Kidney Int. 1999;56:2116–2127. doi: 10.1046/j.1523-1755.1999.00775.x. [DOI] [PubMed] [Google Scholar]
  • 215.Ulloa L, Doody J, Massague J. Inhibition of transforming growth factor-beta/SMAD signalling by the interferon-gamma/STAT pathway. Nature. 1999;397:710–713. doi: 10.1038/17826. [DOI] [PubMed] [Google Scholar]
  • 216.Shao DD, Suresh R, Vakil V, Gomer RH, Pilling D. Pivotal advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J Leukoc Biol. 2008;83:1323–1333. doi: 10.1189/jlb.1107782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Gurujeyalakshmi G, Giri SN. Molecular mechanisms of antifibrotic effect of interferon gamma in bleomycin-mouse model of lung fibrosis: downregulation of TGF-beta and procollagen I and III gene expression. Exp Lung Res. 1995;21:791–808. doi: 10.3109/01902149509050842. [DOI] [PubMed] [Google Scholar]
  • 218.Wynn TA, Cheever AW, Jankovic D, Poindexter RW, Caspar P, Lewis FA, Sher A. An IL-12-based vaccination method for preventing fibrosis induced by schistosome infection. Nature. 1995;376:594–596. doi: 10.1038/376594a0. [DOI] [PubMed] [Google Scholar]
  • 219.Keane MP, Belperio JA, Burdick MD, Strieter RM. IL-12 attenuates bleomycin-induced pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2001;281:L92–L97. doi: 10.1152/ajplung.2001.281.1.L92. [DOI] [PubMed] [Google Scholar]
  • 220.Poynard T, Yuen MF, Ratziu V, Lai CL. Viral hepatitis C. Lancet. 2003;362:2095–2100. doi: 10.1016/s0140-6736(03)15109-4. [DOI] [PubMed] [Google Scholar]
  • 221.Raghu G, Brown KK, Bradford WZ, Starko K, Noble PW, Schwartz DA, King TE., Jr A placebo-controlled trial of interferon gamma-1b in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2004;350:125–133. doi: 10.1056/NEJMoa030511. [DOI] [PubMed] [Google Scholar]
  • 222.King TE, Jr, Albera C, Bradford WZ, Costabel U, Hormel P, Lancaster L, Noble PW, Sahn SA, Szwarcberg J, Thomeer M, Valeyre D, du Bois RM. Effect of interferon gamma-1b on survival in patients with idiopathic pulmonary fibrosis (INSPIRE): a multicentre, randomised, placebo-controlled trial. Lancet. 2009;374:222–228. doi: 10.1016/S0140-6736(09)60551-1. [DOI] [PubMed] [Google Scholar]
  • 223.Szekanecz Z, Koch AE. Macrophages and their products in rheumatoid arthritis. Curr Opin Rheumatol. 2007;19:289–295. doi: 10.1097/BOR.0b013e32805e87ae. [DOI] [PubMed] [Google Scholar]
  • 224.Hasegawa M, Fujimoto M, Kikuchi K, Takehara K. Elevated serum tumor necrosis factor-alpha levels in patients with systemic sclerosis: association with pulmonary fibrosis. J Rheumatol. 1997;24:663–665. [PubMed] [Google Scholar]
  • 225.Zhang Y, Lee TC, Guillemin B, Yu MC, Rom WN. Enhanced IL-1 beta and tumor necrosis factor-alpha release and messenger RNA expression in macrophages from idiopathic pulmonary fibrosis or after asbestos exposure. J Immunol. 1993;150:4188–4196. [PubMed] [Google Scholar]
  • 226.Miyazaki Y, Araki K, Vesin C, Garcia I, Kapanci Y, Whitsett JA, Piguet PF, Vassalli P. Expression of a tumor necrosis factor-alpha transgene in murine lung causes lymphocytic and fibrosing alveolitis. A mouse model of progressive pulmonary fibrosis. J Clin Invest. 1995;96:250–259. doi: 10.1172/JCI118029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Kolb M, Margetts PJ, Anthony DC, Pitossi F, Gauldie J. Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. J Clin Invest. 2001;107:1529–1536. doi: 10.1172/JCI12568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Bahcecioglu IH, Koca SS, Poyrazoglu OK, Yalniz M, Ozercan IH, Ustundag B, Sahin K, Dagli AF, Isik A. Hepatoprotective effect of infliximab, an anti-TNF-alpha agent, on carbon tetrachloride-induced hepatic fibrosis. Inflammation. 2008;31:215–221. doi: 10.1007/s10753-008-9067-1. [DOI] [PubMed] [Google Scholar]
  • 229.Piguet PF, Collart MA, Grau GE, Sappino AP, Vassalli P. Requirement of tumour necrosis factor for development of silica-induced pulmonary fibrosis. Nature. 1990;344:245–247. doi: 10.1038/344245a0. [DOI] [PubMed] [Google Scholar]
  • 230.Piguet PF, Collart MA, Grau GE, Kapanci Y, Vassalli P. Tumor necrosis factor/cachectin plays a key role in bleomycin-induced pneumopathy and fibrosis. J Exp Med. 1989;170:655–663. doi: 10.1084/jem.170.3.655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Tomita K, Tamiya G, Ando S, Ohsumi K, Chiyo T, Mizutani A, Kitamura N, Toda K, Kaneko T, Horie Y, Han JY, Kato S, Shimoda M, Oike Y, Tomizawa M, Makino S, Ohkura T, Saito H, Kumagai N, Nagata H, Ishii H, Hibi T. Tumour necrosis factor alpha signalling through activation of Kupffer cells plays an essential role in liver fibrosis of non-alcoholic steatohepatitis in mice. Gut. 2006;55:415–424. doi: 10.1136/gut.2005.071118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Raghu G, Brown KK, Costabel U, Cottin V, du Bois RM, Lasky JA, Thomeer M, Utz JP, Khandker RK, McDermott L, Fatenejad S. Treatment of idiopathic pulmonary fibrosis with etanercept: an exploratory, placebo-controlled trial. Am J Respir Crit Care Med. 2008;178:948–955. doi: 10.1164/rccm.200709-1446OC. [DOI] [PubMed] [Google Scholar]
  • 233.Bujak M, Frangogiannis NG. The role of IL-1 in the pathogenesis of heart disease. Arch Immunol Ther Exp (Warsz) 2009;57:165–176. doi: 10.1007/s00005-009-0024-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Jones LK, O’Sullivan KM, Semple T, Kuligowski MP, Fukami K, Ma FY, Nikolic-Paterson DJ, Holdsworth SR, Kitching AR. IL-1RI deficiency ameliorates early experimental renal interstitial fibrosis. Nephrol Dial Transplant. 2009;24:3024–3032. doi: 10.1093/ndt/gfp214. [DOI] [PubMed] [Google Scholar]
  • 235.Kamari Y, Shaish A, Vax E, Shemesh S, Kandel-Kfir M, Arbel Y, Olteanu S, Barshack I, Dotan S, Voronov E, Dinarello CA, Apte RN, Harats D. Lack of interleukin-1alpha or interleukin-1beta inhibits transformation of steatosis to steatohepatitis and liver fibrosis in hypercholesterolemic mice. J Hepatol. 2011;55:1086–1094. doi: 10.1016/j.jhep.2011.01.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Meduri GU, Kohler G, Headley S, Tolley E, Stentz F, Postlethwaite A. Inflammatory cytokines in the BAL of patients with ARDS. Persistent elevation over time predicts poor outcome. Chest. 1995;108:1303–1314. doi: 10.1378/chest.108.5.1303. [DOI] [PubMed] [Google Scholar]
  • 237.Gasse P, Riteau N, Vacher R, Michel ML, Fautrel A, di Padova F, Fick L, Charron S, Lagente V, Eberl G, Le Bert M, Quesniaux VF, Huaux F, Leite-de-Moraes M, Ryffel B, Couillin I. IL-1 and IL-23 mediate early IL-17A production in pulmonary inflammation leading to late fibrosis. PLoS One. 2011;6:e23185. doi: 10.1371/journal.pone.0023185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Fan JM, Huang XR, Ng YY, Nikolic-Paterson DJ, Mu W, Atkins RC, Lan HY. Interleukin-1 induces tubular epithelial-myofibroblast transdifferentiation through a transforming growth factor-beta1-dependent mechanism in vitro. Am J Kidney Dis. 2001;37:820–831. doi: 10.1016/s0272-6386(01)80132-3. [DOI] [PubMed] [Google Scholar]
  • 239.Christie JD, Edwards LB, Kucheryavaya AY, Benden C, Dobbels F, Kirk R, Rahmel AO, Stehlik J, Hertz MI. The Registry of the International Society for Heart and Lung Transplantation: twenty-eighth adult lung and heart-lung transplant report—2011. J Heart Lung Transplant. 2011;30:1104–1122. doi: 10.1016/j.healun.2011.08.004. [DOI] [PubMed] [Google Scholar]
  • 240.Valentine VG, Gupta MR, Walker JE, Jr, Seoane L, Bonvillain RW, Lombard GA, Weill D, Dhillon GS. Effect of etiology and timing of respiratory tract infections on development of bronchiolitis obliterans syndrome. J Heart Lung Transplant. 2009;28:163–169. doi: 10.1016/j.healun.2008.11.907. [DOI] [PubMed] [Google Scholar]
  • 241.Vos R, Vanaudenaerde BM, Geudens N, Dupont LJ, Van Raemdonck DE, Verleden GM. Pseudomonal airway colonisation: risk factor for bronchiolitis obliterans syndrome after lung transplantation? Eur Respir J. 2008;31:1037–1045. doi: 10.1183/09031936.00128607. [DOI] [PubMed] [Google Scholar]
  • 242.Palmer SM, Burch LH, Trindade AJ, Davis RD, Herczyk WF, Reinsmoen NL, Schwartz DA. Innate immunity influences long-term outcomes after human lung transplant. Am J Respir Crit Care Med. 2005;171:780–785. doi: 10.1164/rccm.200408-1129OC. [DOI] [PubMed] [Google Scholar]
  • 243.Palmer SM, Klimecki W, Yu L, Reinsmoen NL, Snyder LD, Ganous TM, Burch L, Schwartz DA. Genetic regulation of rejection and survival following human lung transplantation by the innate immune receptor CD14. Am J Transplant. 2007;7:693–699. doi: 10.1111/j.1600-6143.2007.01669.x. [DOI] [PubMed] [Google Scholar]
  • 244.Oyaizu T, Okada Y, Shoji W, Matsumura Y, Shimada K, Sado T, Sato M, Kondo T. Reduction of recipient macrophages by gadolinium chloride prevents development of obliterative airway disease in a rat model of heterotopic tracheal transplantation. Transplantation. 2003;76:1214–1220. doi: 10.1097/01.TP.0000088672.48259.F1. [DOI] [PubMed] [Google Scholar]
  • 245.Rizzo M, SivaSai KS, Smith MA, Trulock EP, Lynch JP, Patterson GA, Mohanakumar T. Increased expression of inflammatory cytokines and adhesion molecules by alveolar macrophages of human lung allograft recipients with acute rejection: decline with resolution of rejection. J Heart Lung Transplant. 2000;19:858–865. doi: 10.1016/s1053-2498(00)00165-0. [DOI] [PubMed] [Google Scholar]
  • 246.Bharat A, Narayanan K, Street T, Fields RC, Steward N, Aloush A, Meyers B, Schuessler R, Trulock EP, Patterson GA, Mohanakumar T. Early posttransplant inflammation promotes the development of alloimmunity and chronic human lung allograft rejection. Transplantation. 2007;83:150–158. doi: 10.1097/01.tp.0000250579.08042.b6. [DOI] [PubMed] [Google Scholar]
  • 247.Vanaudenaerde BM, De Vleeschauwer SI, Vos R, Meyts I, Bullens DM, Reynders V, Wuyts WA, Van Raemdonck DE, Dupont LJ, Verleden GM. The role of the IL23/IL17 axis in bronchiolitis obliterans syndrome after lung transplantation. Am J Transplant. 2008;8:1911–1920. doi: 10.1111/j.1600-6143.2008.02321.x. [DOI] [PubMed] [Google Scholar]
  • 248.Riise GC, Williams A, Kjellstrom C, Schersten H, Andersson BA, Kelly FJ. Bronchiolitis obliterans syndrome in lung transplant recipients is associated with increased neutrophil activity and decreased antioxidant status in the lung. Eur Respir J. 1998;12:82–88. doi: 10.1183/09031936.98.12010082. [DOI] [PubMed] [Google Scholar]
  • 249.Boehler A, Bai XH, Liu M, Cassivi S, Chamberlain D, Slutsky AS, Keshavjee S. Upregulation of T-helper 1 cytokines and chemokine expression in post-transplant airway obliteration. Am J Respir Crit Care Med. 1999;159:1910–1917. doi: 10.1164/ajrccm.159.6.9806035. [DOI] [PubMed] [Google Scholar]
  • 250.Hodge G, Hodge S, Chambers D, Reynolds PN, Holmes M. Bronchiolitis obliterans syndrome is associated with absence of suppression of peripheral blood Th1 proinflammatory cytokines. Transplantation. 2009;88:211–218. doi: 10.1097/TP.0b013e3181ac170f. [DOI] [PubMed] [Google Scholar]
  • 251.Jain R, Hachem RR, Morrell MR, Trulock EP, Chakinala MM, Yusen RD, Huang HJ, Mohanakumar T, Patterson GA, Walter MJ. Azithromycin is associated with increased survival in lung transplant recipients with bronchiolitis obliterans syndrome. J Heart Lung Transplant. 2010;29:531–537. doi: 10.1016/j.healun.2009.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Yates B, Murphy DM, Forrest IA, Ward C, Rutherford RM, Fisher AJ, Lordan JL, Dark JH, Corris PA. Azithromycin reverses airflow obstruction in established bronchiolitis obliterans syndrome. Am J Respir Crit Care Med. 2005;172:772–775. doi: 10.1164/rccm.200411-1537OC. [DOI] [PubMed] [Google Scholar]
  • 253.Vos R, Vanaudenaerde BM, Verleden SE, De Vleeschauwer SI, Willems-Widyastuti A, Van Raemdonck DE, Schoonis A, Nawrot TS, Dupont LJ, Verleden GM. A randomized placebo-controlled trial of azithromycin to prevent bronchiolitis obliterans syndrome after lung transplantation. Eur Respir J. 2011 doi: 10.1183/09031936.00068310. [DOI] [PubMed] [Google Scholar]
  • 254.Kanoh S, Rubin BK. Mechanisms of action and clinical application of macrolides as immunomodulatory medications. Clin Microbiol Rev. 2010;23:590–615. doi: 10.1128/CMR.00078-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Feola DJ, Garvy BA, Cory TJ, Birket SE, Hoy H, Hayes D, Jr, Murphy BS. Azithromycin alters macrophage phenotype and pulmonary compartmentalization during lung infection with Pseudomonas. Antimicrob Agents Chemother. 2010;54:2437–2447. doi: 10.1128/AAC.01424-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 256.Aris RM, Walsh S, Chalermskulrat W, Hathwar V, Neuringer IP. Growth factor upregulation during obliterative bronchiolitis in the mouse model. Am J Respir Crit Care Med. 2002;166:417–422. doi: 10.1164/rccm.2102106. [DOI] [PubMed] [Google Scholar]
  • 257.Smith CR, Jaramillo A, Lu KC, Higuchi T, Kaleem Z, Mohanakumar T. Prevention of obliterative airway disease in HLA-A2-transgenic tracheal allografts by neutralization of tumor necrosis factor. Transplantation. 2001;72:1512–1518. doi: 10.1097/00007890-200111150-00007. [DOI] [PubMed] [Google Scholar]
  • 258.Farivar AS, Mackinnon-Patterson B, McCourtie AS, Namkung J, Ward PA, Mulligan MS. Obliterative airway disease in rat tracheal allografts requires tumor necrosis factor alpha. Exp Mol Pathol. 2005;78:190–197. doi: 10.1016/j.yexmp.2004.10.008. [DOI] [PubMed] [Google Scholar]
  • 259.Alho HS, Maasilta PK, Harjula AL, Hammainen P, Salminen J, Salminen US. Tumor necrosis factor-alpha in a porcine bronchial model of obliterative bronchiolitis. Transplantation. 2003;76:516–523. doi: 10.1097/01.TP.0000074700.30536.76. [DOI] [PubMed] [Google Scholar]
  • 260.Fullmer JJ, Fan LL, Dishop MK, Rodgers C, Krance R. Successful treatment of bronchiolitis obliterans in a bone marrow transplant patient with tumor necrosis factor-alpha blockade. Pediatrics. 2005;116:767–770. doi: 10.1542/peds.2005-0806. [DOI] [PubMed] [Google Scholar]
  • 261.Fan L, Benson HL, Vittal R, Mickler EA, Presson R, Fisher AJ, Cummings OW, Heidler KM, Keller MR, Burlingham WJ, Wilkes DS. Neutralizing IL-17 prevents obliterative bronchiolitis in murine orthotopic lung transplantation. Am J Transplant. 2011;11:911–922. doi: 10.1111/j.1600-6143.2011.03482.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Elssner A, Jaumann F, Dobmann S, Behr J, Schwaiblmair M, Reichenspurner H, Furst H, Briegel J, Vogelmeier C. Elevated levels of interleukin-8 and transforming growth factor-beta in bronchoalveolar lavage fluid from patients with bronchiolitis obliterans syndrome: proinflammatory role of bronchial epithelial cells. Munich Lung Transplant Group. Transplantation. 2000;70:362–367. doi: 10.1097/00007890-200007270-00022. [DOI] [PubMed] [Google Scholar]
  • 263.Ramirez AM, Takagawa S, Sekosan M, Jaffe HA, Varga J, Roman J. Smad3 deficiency ameliorates experimental obliterative bronchiolitis in a heterotopic tracheal transplantation model. Am J Pathol. 2004;165:1223–1232. doi: 10.1016/S0002-9440(10)63382-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Liu M, Suga M, Maclean AA, St George JA, Souza DW, Keshavjee S. Soluble transforming growth factor-beta type III receptor gene transfection inhibits fibrous airway obliteration in a rat model of Bronchiolitis obliterans. Am J Respir Crit Care Med. 2002;165:419–423. doi: 10.1164/ajrccm.165.3.2102108. [DOI] [PubMed] [Google Scholar]
  • 265.Borthwick LA, Sunny SS, Oliphant V, Perry J, Brodlie M, Johnson GE, Ward C, Gould K, Corris PA, De Soyza A, Fisher AJ. Pseudomonas aeruginosa accentuates epithelial-to-mesenchymal transition in the airway. Eur Respir J. 2011;37:1237–1247. doi: 10.1183/09031936.00088410. [DOI] [PubMed] [Google Scholar]
  • 266.Gilpin SE, Lung KC, Sato M, Singer LG, Keshavjee S, Waddell TK. Altered progenitor cell and cytokine profiles in bronchiolitis obliterans syndrome. J Heart Lung Transplant. 2012;31:222–228. doi: 10.1016/j.healun.2011.11.012. [DOI] [PubMed] [Google Scholar]
  • 267.Harris DA, Zhao Y, Lapar DJ, Emaminia A, Steidle JF, Stoler M, Linden J, Kron IL, Lau CL. Inhibiting CXCL12 blocks fibrocyte migration and differentiation and attenuates bronchiolitis obliterans in a murine heterotopic tracheal transplant model. J Thorac Cardiovasc Surg. 2012 doi: 10.1016/j.jtcvs.2012.03.079. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES