Abstract
The cardinal features of asthma include pulmonary inflammation and airway hyperresponsiveness (AHR). Classically, asthma, specifically allergic asthma, has been attributed to a hyperactive Th2 cell immune response. However, the Th2 cell-mediated inflammation model has failed to adequately explain many of the clinical and molecular aspects of asthma. In addition, the outcomes of Th2-targeted therapeutic trials have been disappointing. Thus, asthma is now believed to be a complex and heterogeneous disorder, with several molecular mechanisms underlying the airway inflammation and AHR that is associated with asthma. The original classification of Th1 and Th2 pathways has recently been expanded to include additional effector Th cell subsets. These include Th17, Th9 and Treg cells. Emerging data highlight the involvement of these new Th cell subsets in the initiation and augmentation of airway inflammation and asthmatic responses. We now review the roles of these recently classified effector Th cell subsets in asthmatic inflammation and the insights they may provide in addition to the traditional Th2 paradigm. The hope is that a clearer understanding of the inflammatory pathways involved and the mediators of inflammation will yield better targeted therapeutics.
Keywords: Asthma, Th17, Th2, Th1, Th9, Animal modes
INTRODUCTION
Bronchial asthma is a respiratory disease that affects nearly one in ten individuals in the developed world (Fanta, 2009; Umetsu et al., 2002). Indeed, due to the persistent rise in the incidence and prevalence of this disease throughout the past 2–3 decades, it is now estimated that asthma has reached epidemic proportions. In fact, in industrialized nations, the most common chronic disease of childhood is asthma (van de Kant et al., 2009). Although current therapies are effective in suppressing disease symptoms, to date, there are no preventive treatments or cures for asthma. A better understanding of general pulmonary immunity and specifically, the initiators of inflammation associated with asthma, is therefore needed for development of new therapeutics.
Inflammation in Asthma
The cardinal features of asthma include airway inflammation and airway hyperreactivity (AHR) which results in loss of pulmonary function. Symptoms of the disease comprise wheezing, breathlessness, chest tightness, and coughing due to reversible airway obstruction. Initially, studies indicated that asthma is an atopic disorder of the airways which involves activated T helper lymphocytes, specifically CD4+ Th2 cells. Analysis of bronchial biopsies and bronchoalveolar lavage (BAL) fluids from asthmatic patients showed an infiltration of Th2 lymphocytes, eosinophils and degranulated mast cells (Azzawi et al., 1990; Robinson et al., 1993; Robinson et al., 1992). These results framed research efforts for many years and suggested that, following exposure to allergen, activated Th2 cells and their cytokines, namely interleukin-4 (IL-4), IL-5, IL-9 and IL-13, orchestrate eosinophilic airway inflammation as well as stimulate B-cells to generate allergen-specific IgE antibody, and that these events ultimately lead to the release of preformed or newly synthesized inflammatory mediators from mast cells (Cohn et al., 2004; Wills-Karp, 1999).
However, recent studies have suggested that eosinophilic airway inflammation actually occurs in less than 50% of asthmatic cases (Douwes et al., 2002; Simpson et al., 2006). The most common form of asthma is allergic asthma (Barrios et al., 2006; Masoli et al., 2004), yet multiple and distinct forms of asthma exist. These other forms can be induced by exercise, air pollution, aspirin, or viral infection (Matangkasombut et al., 2009). Moreover, the type and intensity of inflammatory cellular infiltrates as well as the site of infiltration can vary depending upon the degree of disease severity and the form of asthma (Vignola et al., 1998). In addition, as many as 12 different types of inflammatory cells and more than 100 inflammatory mediators have been associated with the pulmonary inflammation that occurs in asthma (Anderson, 2008; Kiley et al., 2007). Thus, asthma is clearly a complex and heterogeneous disease. In order to fully understand the process of disease pathogenesis, it will be essential to identify the factors that initiate, intensify, and mediate the immune response that results in airway inflammation and hyperreactivity. Although the presence of Th2 cells and eosinophils can explain many features of asthma, it is becoming increasingly obvious that this paradigm may be too simplistic.
New T Helper Cell Subsets
In recent years, as the complexity of asthma has become better appreciated, newly emerging CD4+ Th cell subsets have been linked to general disease pathogenesis. It is evident that Th cell functions are considerably more complex and heterogeneous than originally thought. The original characterization of the Th1 and Th2 pathways has now been expanded to include additional Th cell subsets, each with their own cytokine repertoire and transcription factors.
These include regulatory T cells (Treg) (Robinson, 2009), Th17 cells (Louten et al., 2009) and most recently, Th cells that produce IL-9 (Th9 cells) (Soroosh and Doherty, 2009). The discovery of new Th cell subsets has motivated investigators to revisit and reinterpret existing models of autoimmunity and allergy, including asthma. Furthermore, T cell lineage commitment has recently been shown to be less rigid than previously appreciated (Zhou et al., 2009). Epigenetic modification of cytokine genes and key transcriptional regulator genes direct T cell differentiation and appear to allow T cells to adapt to specific effector profiles. In this review, we discuss the classic Th2 inflammation paradigm, its role in lung mucosal inflammation and the apparent limitations of this model, which underlie discrepancies observed in human asthma. We also discuss the role of Th1, Th9, Treg and Th17 cells in lung mucosal inflammation and asthma, and their potential role in resolving these discrepancies. Ultimately, insights into the role of Th cell subsets in asthma pathogenesis will allow design of new targets for therapy.
The Th2 Cell Paradigm
It is widely believed that Th2 cells initiate and perpetuate asthma. At the onset of detailed studies, CD4+ Th2 cells were found to be present in the lungs of asthmatic patients, particularly those with allergic asthma, as indicated by the fact that cells obtained from BAL fluids of asthmatic patients contain more IL-4 and IL-5 mRNA than do cells from BAL fluids of non-asthmatic patients or patients who have other lung diseases such as pneumonia or sarcoidosis (Robinson et al., 1992; Ying et al., 1997). Additionally, IL-4 and IL-5 expression in asthmatic patients is predominantly T cell-derived (Walker et al., 1994).
The specific contribution of Th2 lymphocytes has also been documented in classical allergen challenge studies. In these studies, sensitized asthmatics were exposed to aerosolized allergen and found to subsequently exhibit airflow obstruction, an influx of eosinophils and T lymphocytes, and an increase in Th2 cytokines in BAL and bronchial mucosa (Barrett and Austen, 2009). Furthermore, the level of T cell infiltrate observed in the BAL correlates with the severity of asthma (Kay, 1997). These results indicate an undoubtedly vital role for Th2 cells in the development for some forms of human asthma.
Mouse models of antigen-induced airway inflammation have also reinforced a central role for Th2 lymphocytes and their cytokines in asthma. Using an experimental ovalbumin (OVA) asthma model, mice receiving OVA-specific, DO11.10 TCR-transgenic Th2 cells, but not Th1 cells, developed hallmark traits of asthma, such as eosinophilic airway inflammation and AHR, following OVA challenge (Cohn et al., 1997; Cohn et al., 1998). Moreover, development of allergic lung inflammation could be prevented in OVA-sensitized wild-type (WT) mice by depletion of CD4+ T cells prior to allergen challenge (Gavett et al., 1994).
The specific roles of individual Th2 cytokines were elucidated through sensitization and airway challenge studies in mice using either transgenic cytokine overexpression, targeted gene deletion, or blocking anti-cytokine antibodies. The results from these studies demonstrated a central role for IL-4 in the generation of Th2 cells and B-cell IgE production; for IL-5 in the promotion of eosinophilic airway inflammation and damage; for IL-9 in the recruitment, proliferation, and activation of mast cells; and for IL-13 in the induction of AHR, goblet cell hyperplasia, mucus secretion, and fibrosis (Fig. 1) (Burrows et al., 1989; Holgate, 2008; Kay, 2006; Larche et al., 2003). As a result of activation of allergen-specific Th2 cells, allergen-specific IgE is produced and binds to IgE receptors on the surface of mast cells, which, after crosslinking by specific allergen, induces the activation of these cells. Mast cell activation then results in release of preformed mediators, such as histamine and leukotrienes, which directly affect airway smooth muscle and mucous glands, ultimately causing AHR (Galli, 1997; Lane and Lee, 1996; Rossi and Olivieri, 1997). Overall, these studies have led to the dominant concept that activated Th2 cells orchestrate pulmonary immune responses and mediate the lung inflammation and AHR that is seen in asthmatic patients.
Figure 1.
T helper cell subsets in asthma. As allergens enter the airway, professional antigen-presenting cells take up the allergen and present it to naïve CD4+ T helper cells. This leads to the activation of allergen-specific Th2 cells. Th2 cells produce IL-4, which induces B-cell activation and IgE antibody production; IL-5, which induces eosinophil recruitment; and IL-13, which promotes mucus production and AHR. In addition to Th2 cells, Th17 cells can also be activated. Th17 cells produce IL-17 and IL-22. These cytokines induce epithelial cells to produce the proteins MUC5B and MUC5AC and granulopoietic factors such as IL-8 and G-CSF, which cause neutrophil recruitment and expansion. There is evidence that Th1-cell responses might also be responsible for some of the pathogenic features observed in patients suffering from chronic forms of allergy, e.g., IFN-γ has been shown to exacerbate an established asthmatic response. IL-9 produced by Th9 cells leads to the recruitment, development, and activation of mast cells. Upon degranulation, IgE-sensitized mast cells release both preformed and newly synthesized mediators in sensitized individuals. These include histamine and leukotrienes, which promote airway inflammation and mucus production. IL-10-producing Treg cells have an immunosuppressive effect on Th2 cell activation that occurs during asthmatic reactions, especially in decreasing production of IgE antibody.
LIMITATIONS OF THE TH2 PARADIGM
Despite clarifying many features of asthma, some clinical and molecular features that have been documented in human asthma are not adequately explained by the Th2 paradigm. Even as the Th2 inflammation model was being advanced, concerns were raised about whether it would actually lead to improved treatments for asthma (Anderson and Coyle, 1994; Coyle et al., 1995). First, the Th2 paradigm does not explain non-allergic asthma. Non-allergic asthma can be induced by exercise, air pollution, aspirin, or viral infection. In fact, viral infections are known to exacerbate asthma (Barrett and Austen, 2009). In addition, in the absence of an allergen, granulocytic cellular infiltration, including infiltration of degranulated mast cells, can still occur and result in airway hyperreactivity, emphasizing the fact that a specific allergen is dispensable for non-allergic pulmonary inflammation.
Second, the Th2 paradigm predicts that eosinophilic infiltration and subsequent inflammation should provoke AHR, but no clear cause and effect exists (Alvarez et al., 2000a; Alvarez et al., 2000b). Studies have shown that atopy associated with eosinophilic activation, and AHR are not complementary (Woolcock and Peat, 2000). Th2 immunity is fundamental to atopy, and atopy is a risk factor for asthma, but atopy alone is a poor predictor of disease. In fact, most patients with allergic rhinitis and allergen sensitization do not exhibit AHR and therefore are not asthmatic (Corren, 1997). These results suggest that the presence of Th2 cells and allergen-specific IgE is not sufficient for development of asthma. Third, bronchial biopsies from asthmatic patients commonly have evidence of neutrophilic inflammation.
Neutrophils are frequently present in the lungs of asthmatics, particularly in patients with severe disease, or with corticosteroid resistant asthma (Green et al., 2002; Simpson et al., 2007; Wenzel et al., 1999). Fourth, therapeutics targeted to Th2 cells and their cytokines, which are often effective in Th2 animal disease models, have been found to have minimal effectiveness in the clinical setting. For instance, administration of antibodies that prevent binding of IgE to FcεRI has had limited therapeutic efficacy, as has inoculation of neutralizing anti-IL-5 antibodies. These treatments typically reduce eosinophilic inflammation but often do not affect AHR or the late asthmatic response.
Additionally, interventions such as anti-IL-4 antibodies or IL-4/IL-13 antagonists have weak effects and have also not reduced AHR in clinical asthma trials (Bryan et al., 2000; Holgate and Polosa, 2008; Leckie et al., 2000; Wenzel et al., 2007). These results suggest that immunological factors in addition to Th2 cells regulate asthma. Finally, asthmatic patients exhibit elevated levels of non-Th2 cytokines and factors, such as interferon (IFN)-γ (Cho et al., 2005; Nakao et al., 2001) and IL-17 (Bullens et al., 2006; Oboki et al., 2008). Taken together, these observations suggest that regulatory pathways in addition to Th2 cells and eosinophils may contribute to the development of the asthma phenotype. In spite of these contradictions, Th2 immunity has been shown to be clinically relevant specifically for childhood asthma with atopy as well as mild allergic adult asthma. Indeed, in these forms of asthma, Th2 directed therapies have shown to be effective (Wenzel et al., 2007). However, it is clear that asthma is complex and that Th2 immunity does not explain all forms of the disorder.
Activity of Th1 Cells
From its inception, the Th1/Th2 model has postulated that Th1 cells could have a beneficial effect on asthma by dampening the activity of Th2 cells. Indeed, Th1 cells have been shown to inhibit development and proliferation of Th2 cells (Abbas et al., 1996). IFN-γ, the hallmark cytokine produced by Th1 cells, has also been shown to abrogate IgE production and eosinophilia (Coffman et al., 1988; Iwamoto et al., 1996), and in animal models, exogenous administration of IFN-γ can result in suppression of allergic airway inflammation (Holgate and Polosa, 2008). Nevertheless, studies involving subcutaneous administration of recombinant human IFN-γ showed no improvement in asthmatic patients compared with controls (Boguniewicz et al., 1995). In fact, in human asthma, IFN-γ production is actually upregulated and appears to contribute to disease pathogenesis (Cho et al., 2005; Nakao et al., 2001).
For instance, in severe asthmatic patients, serum levels of IFN-γ increase during an asthmatic attack (Corrigan and Kay, 1990). Increases in IFN-γ have also been found in BAL cell cultures obtained from asthmatic patients, whether incubated alone or in the presence of allergen (Cembrzynska-Nowak et al., 1993). In an attempt to counterbalance Th2 cell-induced AHR, allergen-specific Th1 cells were adoptively transferred into naïve mice and although these cells demonstrated an ability to migrate to the lungs, they were found to intensify severe airway inflammation and production of IFN-γ, which surprisingly, appeared to contribute to the activation of eosinophils (Hansen et al., 1999). It has recently been shown that the production of IFN-γ together with an established Th2-cell response, results in increased inflammation, possibly by damaging the epithelial cell barrier (Reisinger et al., 2005). Together these observations suggest that the role of Th1 cells in asthma is more complex than predicted. Indeed, Th1 and Th2 cells may not simply serve to counterbalance each other in a dichotomous manner, but rather may act together in a harmful manner in asthmatic individuals.
Th9, a New Subset of T Helper Cells
IL-9 has generally been categorized as a Th2 cytokine (Kay, 2006; Larche et al., 2003). Recently, a new Th subset that preferentially produces IL-9 and that appears to be distinct from Th2 cells, has been reported to provide a unique contribution to immune responses (Dardalhon et al., 2008; Veldhoen et al., 2008). The IL-9-producing T cell lineage was discovered when it was found that, under certain circumstances, naturally arising CD4+CD25+ regulatory T cells (nTreg) and inducible regulatory T cells (iTreg) generated in the presence of TGF-β produced more IL-9 upon activation than Th2 cells (Liu et al., 2006; Lu et al., 2006). However, IL-9 production was found to be absent from Foxp3+ nTregs isolated from the thymus or Foxp3+ iTregs generated from naïve T cells (Veldhoen et al., 2008). However, committed Th2 cells cultured in the presence of TGF-β and IL-4, discontinue expressing the Th2 transcription factor, GATA3, as well as the Th2 cytokines, IL-4, IL-5 and IL-13, while initiating transcription of IL-9 (Veldhoen et al., 2008). These results highlight the differential role of TGF-β in T-cell differentiation such that, in the presence of IL-6, Th17 cell differentiation occurs (see below), but in the presence of IL-4, Th9 cells develop. These experiments, which are relevant to chronic diseases, identified a distinct population of IL-9-producing helper T cells which appear to have a role in allergic diseases as well as asthma.
Th9 Cells in Asthma
The role of IL-9 in asthma, as a Th2 cytokine, has long been recognized. Since the elucidation of Th9 cells and their potential inflammatory function, studies have begun revisiting the role of these cells in asthma. As was documented with other Th2 cytokines, IL-9 mRNA-positive cells have been shown to be elevated in bronchial biopsies from asthmatic patients compared to normal patients or patients with chronic bronchitis or sarcoidosis (Shimbara et al., 2000; Ying et al., 2002). IL-9 levels also increase following allergen challenge of asthmatics compared to normal controls (Erpenbeck et al., 2003a). In the airways, the CD3+ lymphocyte population was found to be the primary source of IL-9 (Erpenbeck et al., 2003b; Tsicopoulos et al., 2004).
The specific role for IL-9 in asthma pathogenesis has primarily been investigated in murine models. Upon systemic overexpression in IL-9 transgenic mice, there was increased AHR, heightened eosinophilic infiltration and increased levels of IgE production following antigen challenge (McLane et al., 1998). However, overexpression of IL-9 was able to enhance asthmatic inflammation only in the presence of specific antigen since the transgenic mice had no evidence of eosinophilic inflammation in the absence of antigen challenge. In contrast, when IL-9 was selectively overexpressed in the lung, using a clara cell (CC10) promoter, eosinophilic airway inflammation, mast cell accumulation as well as AHR occurred without antigen challenge (Temann et al., 1998). In addition, these transgenic mice demonstrated increased mucus production and fibrosis, features of airway remodeling associated with chronic asthma. The role of IL-9 in asthma has also been evaluated using IL-9 deficient mice. Using the OVA-induced asthma model, one group found no changes in AHR, cellular infiltration, mucus production or peribronchial inflammation in IL-9 gene deficient mice compared to WT mice (McMillan et al., 2002).
However, another group reported a significant decrease in AHR and eosinophilic infiltration after administration of an IL-9 blocking antibody at the antigen challenge phase (Cheng et al., 2002). Thus, IL-9 may not be required for the initiation of asthma, but it might play a role in sustaining and modulating subsequent airway responses to antigen. Indeed, the presence of IL-9 is strongly associated in both mice and humans with mast cell accumulation in the airways, as well as AHR and goblet cell metaplasia (Longphre et al., 1999). Also, IL-9 can significantly contribute to the recruitment of mast cells, as immature cells, from the bone marrow and their subsequent differentiation in vivo (Lu et al., 2006).
Undoubtedly, IL-9 maintains the ability to significantly contribute to asthma, specifically chronic asthma. Analysis of specimens from allergic patients has revealed that T cells can be a major source of IL-9 and mouse models have indicated that IL-9 has multiple effects in the development and maintenance of allergic inflammation and airway remodeling. However, it is unknown whether IL-9-secreting T cells are distinct from Th2 cells or whether Th2 cells can be reprogrammed to become Th9 cells. Overall, it appears that T cells are a significant source of IL-9, which can have multiple effects in the development and, likely more importantly, in the maintenance of airway inflammation.
Treg Cells
Another pivotal subset of CD4+ T cells, namely regulatory T cells (Treg cells), has recently been identified as having a distinct role in asthma pathogenesis. Regulatory T cells are characterized by the expression of the transcription factor Foxp3 (Forkhead box p3) and the IL-2 receptor (CD25), and are known to produce the inhibitory cytokines IL-10 and TGF-β (Wing et al., 2006). Treg cells have been strongly associated with suppression of allergic responses in murine models of asthma. For example, adoptive transfer of Treg cells following the onset of allergic airway inflammation, has been shown to downregulate established inflammation and prevent airway remodeling (Kearley et al., 2008).
Further studies demonstrated that the suppressive ability of the adoptively transferred, antigen-specific Treg cells was dependent on IL-10. Not only did the adoptively transferred Treg cells produce IL-10, but these cells also induced IL-10 production from bystander recipient CD4+ T cells (Kearley et al., 2005). In agreement with these findings, depletion of Treg cells before allergen sensitization was found to enhance the severity of airway inflammation and AHR (Lewkowich et al., 2005). There is also strong evidence in humans that Treg cells inhibit Th2 cell responses, which suggests that atopy can result from an imbalance between Th2 cells and Treg cells (Fig. 1) (Larche, 2007). Interestingly, there are substantial decreases in the frequencies of allergen-specific IL-10-producing Treg cells, and increases in IL-4-producing T cells, in allergic individuals compared to healthy, nonatopic individuals (Akdis et al., 2004), which further highlights the close interplay between Treg and T effector cells in asthma.
The Role of IL-10
One significant feature of Treg cell function is the role of IL-10. IL-10 has been shown to be an effective immunosuppressive mediator of allergic responses in the lung, as well as other mucosal sites. Targeted deletion of IL-10, specifically in Treg cells, results in development of spontaneous colitis as well as increased AHR and inflammation in mice exposed to inhaled allergen (Grunig et al., 1997; Rubtsov et al., 2008). IL-10 dampens Th2 effector cell activation and inhibits both early- and late-phase asthmatic responses, including both mast cell activation and eosinophilic inflammation (O’Garra et al., 2008). Furthermore, the administration of exogenous IL-10 can inhibit AHR and alleviate asthma pathology.
It can also inhibit IgE production while promoting IgG4 production, an immunoglobulin isotype generally believed to be protective against allergic responses (Till et al., 2004). To further test the ability of IL-10 to inhibit inflammation, the gene encoding IL-10 was instilled intratracheally into IL-10 deficient mice following allergen challenge and was found to significantly suppress airway inflammation and AHR (Fu et al., 2006). The role of IL-10 in asthma has also been investigated in human allergic and asthmatic patients. There are substantial reductions in IL-10 transcript and protein levels, and increased amounts of proinflammatory cytokines, in the BAL fluids and in alveolar macrophages of asthmatic patients compared to healthy control subjects (John et al., 1998).
Additionally, a polymorphism in the human IL-10 gene promoter, which results in reduced IL-10 expression, has been correlated with severe asthma (Lim et al., 1998). Interestingly, recent evidence suggests that depending on the cytokine milieu, Th2 cells may convert into Th cells that express IL-10 (Dardalhon et al., 2008). Indeed, IL-10-producing Treg cells are key players in the complex immunological mechanisms designed to maintain homeostasis in the lung. Overall, this evidence suggests that IL-10-producing Treg cells are able to suppress inflammatory immune responses while lack of these cells heightens the inflammation seen in asthma.
Th17 cells
The Th1/Th2 paradigm has recently been expanded to include Th17 cells (Steinman, 2007). IL-17-producing T cells were first isolated from human rheumatoid synovial tissue (Aarvak et al., 1999) and found to be induced by microbial lipopeptides (Infante-Duarte et al., 2000). These results ultimately led to the hypothesis that IL-17A production actually designates a distinct subset of CD4+ T helper cells that principally functions in inflammatory reactions. Soon afterwards, use of IL-17-deficient mice (Nakae et al., 2002; Nakae et al., 2003) and antibody-mediated IL-17A neutralization (Bush et al., 2002) demonstrated that IL-17-producing T cells mediate pathology in autoimmune models. IL-17 production was found to be promoted in vitro by IL-23, which stimulated CD4+ Th cells distinct from either the Th1 nor Th2 pathway (Aggarwal et al., 2003).
For instance, mice that lack the IL-12 p40 subunit, which is shared by IL-23, or the p19 subunit that is specific to IL-23, were found to develop resistance to autoimmune diseases due to the absence of IL-17-producing T cells (Cua et al., 2003; Harrington et al., 2005; Murphy et al., 2003; Park et al., 2005). Although IL-23 can enhance IL-17 expression, actual Th17 cell differentiation from uncommitted cell precursors in mice requires IL-6 and TGF-β, both in vitro (Veldhoen et al., 2006) and in vivo (Bettelli et al., 2006; Mangan et al., 2006). On the other hand, IL-6 plus IL-1β or IL-21 have been found to be critical for Th17 development in humans (Manel et al., 2008). These cytokines, acting via STAT3, induce increased expression of the key transcription factors for Th17 cell differentiation namely, retinoic acid-related orphan receptors (ROR)γt and RORα (Ivanov et al., 2006). The identification of these specific transcription factors further supports the concept that IL-17-producing cells represent a distinct T helper cell lineage.
The production of IL-17A, IL-17F, IL-22, and, to a lesser extent, tumor necrosis factor (TNF) and IL-6, currently defines the Th17 pathway (Bettelli et al., 2008). Th17 cells are now accepted to represent a third CD4+ Th subset, which has led to the resolution of some inconsistencies in the Th1/Th2 paradigm. Additionally, IL-17 has been implicated in asthma development (Hellings et al., 2003). Thus, a full understanding of the effector functions of Th17 cells during pulmonary inflammation may be key to the ultimate control of asthma pathogenesis.
Th17 Cells and Asthma Pathogenesis
Increased expression of IL-17A has been detected in the lungs, sputum, BAL fluids, and sera of asthmatic patients (Barczyk et al., 2003; Bullens et al., 2006; Chakir et al., 2003; Laan et al., 2002; Wong et al., 2001), and IL-17A expression is correlated with the extent of AHR following administration of the lung irritant, methacholine (Barczyk et al., 2003; Kolls et al., 2003).
Additionally, IL-17 production was found to have a significant role in collagen deposition and airway remodeling, traits of chronic asthma (Molet et al., 2003). IL-17A and/or IL-17F can orchestrate local inflammation by inducing human bronchial fibroblasts and airway smooth muscle cells to release proinflammatory cytokines such as TNF-α, IL-1β, granulocyte colony-stimulating factor (G-CSF), and IL-6, as well as the neutrophil chemotactic proteins, IL-8 and CXCL1/Gro-α (Fig. 1) (Molet et al., 2001). IL-17A can also act in concert with IL-6 to induce human bronchial epithelial cells to produce the mucus proteins MUC5B and MUC5AC (Chen et al., 2003; Kawaguchi et al., 2001a; Laan et al., 1999). Previously, goblet cell hyperplasia (a hallmark asthmatic trait) was linked, albeit weakly, to Th2 cytokine production. The more recent results, in addition to other studies (Oda et al., 2005; Wakashin et al., 2008), now convincingly show that IL-17 maintains increased mucin gene expression in the airways and may underscore a significant role of IL-17 in airway inflammation.
Animal studies have further implicated Th17 cells and their cytokines in provocation of lung inflammation. Over-expression of IL-17A in the lung has been found to result in neutrophilic infiltration and elevated levels of granulopoeitic factors such as CXCL1/Gro-α, IL-8, and G-CSF (Laan et al., 1999; Schwarzenberger et al., 1998). Conversely, mice deficient for IL-17A or IL-17A receptor (IL-17RA) expression exhibited diminished pulmonary neutrophil recruitment in response to allergen challenge (Nakae et al., 2002). Taken together, these results suggest that IL-17A plays a pivotal role in lung inflammation, especially in those cases of severe asthma in humans that are characterized by a neutrophil-dominant cellular infiltrate. However, the role of IL-17 in murine models of allergic asthma, which are typically Th2-cell-mediated, has been more obscure. On the one hand, following sensitization and allergen challenge in an OVA asthma model, IL-17A- or IL-17RA-deficient mice showed impaired induction of OVA-specific T cells, reduced eosinophilic inflammation, and lower levels of serum IgE production (Durrant et al., 2009; Nakae et al., 2002; Schnyder-Candrian et al., 2006), suggesting that IL-17 is required for the initiation of asthma.
Similarly, blocking the effects of IL-17 following OVA challenge in mice that were sensitized epicutaneously, resulted in reduced neutrophil influx into the lung and reversal of bronchial hyperreactivity (He et al., 2007). However, IL-17 neutralization with anti-IL-17A mAb in an OVA-challenge asthma model also caused elevated eosinophilic infiltration and IL-5 production in BAL (Schnyder-Candrian et al., 2006), suggesting an inhibitory role of IL-17A on established Th2-driven allergic immune responses. Furthermore, when IL-6-deficient mice, which are defective in Th17 cell development, were epicutaneously sensitized to allergen, Th2 cells were activated and the animals developed exacerbated pulmonary eosinophilic inflammation (Wang et al., 2000). These results suggested that treatment with exogenous IL-17 would alleviate established Th2-mediated asthma. Indeed, recombinant IL-17 administered i.n. during the challenge phase of allergen appeared to inhibit asthma responses such as AHR and eosinophilic inflammation (Schnyder-Candrian et al., 2006). These findings underscore a crucial role of this cytokine in pulmonary inflammation but also emphasize a need for further clarification of this pathway in the disease process.
In addition to Th2 cell-driven murine models of allergic asthma, the role of IL-17A has been studied in Th17-dominant models of asthma. Mice deficient in T-bet, the transcription factor necessary for Th1 cell development, have been shown to develop an allergic immune response that is dominated by neutrophilic influx that is associated with increased expression of IL-17A, but not IL-4 or IL-5 (Durrant et al., 2009; Fujiwara et al., 2007). Furthermore, OVA challenge of DO11.10 OVA-specific TCR transgenic mice has been reported to result in neutrophilic inflammation without an increase in serum IgE (Wilder et al., 2001).
In this model, Th1 and Th17 cells/cytokines, but not Th2 cells/cytokines, have been found to be increased in BAL from DO11.10 and OTII mice exposed to OVA inhalation (Nakae et al., 2007). Following neutralization of IL-17A during the OVA challenge phase in T-bet deficient mice, neutrophil-mediated lung inflammation was alleviated as well as AHR (Durrant et al., 2009). Similarly, IL-17-deficient DO11.10 mice (Nakae et al., 2002) and IL-17-deficient OTII mice (Nakae et al., 2007) demonstrated decreased neutrophilic airway inflammation and AHR following OVA challenge. Taken together, these observations indicate that IL-17/Th17 cells are responsible for airway neutrophilia in asthma models that use T-bet deficient or OVA-specific TCR transgenic mice.
Several studies have also described the potential role of IL-17 in human lung disease and it has been shown that IL-17 can induce pulmonary inflammation (Sharkhuu et al., 2006). Specifically, accumulating evidence implicates IL-17 in the development and progression of human asthma. IL-17 expression is upregulated after allergen challenge, and in vitro allergen stimulation of T cells from atopic asthmatic patients leads to enhanced IL-17 production compared to the levels seen in nonallergic control subjects (Hashimoto et al., 2005; Kawaguchi et al., 2001b). As has been seen with other cytokines, the level of IL-17A expression correlates with the severity of AHR in patients. Interestingly, IL-17A has also been shown to induce the release of the eosinophil-recruiting chemokine eotaxin from airway smooth muscle cells, thus likely aiding in eosinophilic inflammation, and both IL-17A and IL-17F have been shown to be able to induce the release of inflammatory mediators, such as CXCL1/Gro-α and IL-8, from human eosinophils in vitro (Cheung et al., 2008; Rahman et al., 2006).
Although eosinophilic airway inflammation is recognized as a hallmark feature of disease in some asthmatic patients, recent studies indicate an significant role for neutrophils in asthma (Monteseirin, 2009). Although approximately 50% of asthma cases demonstrate eosinophilic inflammation, most other cases of asthma exhibit an increase in airway neutrophils and interleukin 8 (IL-8) (Douwes et al., 2002). Indeed, the severity of asthma may be determined by the balance between IL-4 and IL-17 levels as well as the amount of eosinophilic versus neutrophilic infiltration in the lung. It is possible that IL-17 has a subdominant role in allergic asthma, but a central role in nonallergic asthma in which neutrophils play a critical role. Overall, Th17 cells have an undeniable role in asthma disease pathogenesis and as their role is clarified, this cell pathway will become an attractive target for asthma.
Targeting Th17 cells with a Th1 Cytokine
Many therapeutic approaches for regulating asthmatic responses have relied upon the Th1/Th2 paradigm and numerous studies have compellingly demonstrated that Th1 cells can inhibit the development of Th2-mediated airway inflammation (Coyle et al., 1996). However, therapeutic strategies based on the Th1/Th2 model have thus far led to disappointing outcomes in some asthmatic patients (see above). For instance, IL-12, a key cytokine in modulating the balance between Th1 and Th2 cells, inhibits AHR and airway eosinophilia after antigen challenge in animal models (Kips et al., 1996; Schwarze et al., 1998).
However, when asthmatic patients were treated with recombinant IL-12, there was no significant effect on AHR or on the asthmatic reaction (Bryan et al., 2000). These results suggest that either IL-12 is ineffective in its ability to regulate Th1/Th2 imbalances in humans, which from animal studies seems to be unlikely, or there is an incomplete understanding of the mechanisms mediating the asthmatic response. In any case, the experience in clinical trials has reiterated the need for a better understanding of the molecular mechanisms that drive immune responses in the lung.
Of particular interest are recent studies showing that T-bet is significantly decreased in the airways of asthmatic patients (Finotto et al., 2002) and that polymorphisms of this gene in humans are correlated with airway inflammation and AHR (Munthe-Kaas et al., 2008; Raby et al., 2006). T-bet is a T-box transcription factor and is crucial for Th1 cell differentiation and IFN-γ production. In the absence of T-bet, Th cell responses following allergen challenge have been shown to be predominantly Th17-mediated (Durrant et al., 2009; Fujiwara et al., 2007). Recently, IL-12 was used to target Th17 cell-mediated lung inflammation in a T-bet deficient OVA asthma model. In the absence of T-bet, there was little to no IFN-γ expression; however, antigen-induced airway inflammation as well as AHR was almost completely suppressed following treatment with IL-12 during the OVA challenge phase.
Surprisingly, this suppression was found to be dependant on IL-10, which acted through inhibiting IL-6 and TGF-β (Fig. 2) (Durrant and Metzger, submitted for publication). These results suggest that in a Th17 cell-mediated model, IL-12 has a strong immunosuppressive, IFN-γ-independent effect and could represent a novel mechanistic advance in which the Th1-inducing cytokine, IL-12, inhibits inflammatory Th17 cytokine production through induction of IL-10.
Figure 2.
A novel cytokine-based therapy in asthma. Allergic inflammation associated with asthma has been characterized as primarily an eosinophilic/Th2-mediated disease. However, in the absence of T-bet, the master transcription factor necessary for Th1 cell development and IFN-γ production, allergic inflammation is largely mediated by a neutrophilic/Th17 cell response. The Th1 cell promoting cytokine, IL-12, can inhibit this Th17-mediated inflammation and AHR in the absence of IFN-γ production, through induction of pulmonary IL-10 expression.
CONCLUSION
Asthma is now regarded as a complex and heterogeneous disorder. The historical view of this disease as solely a Th2-mediated condition has been challenged by inconsistencies observed in clinical presentation, by responses to targeted treatments (or lack thereof), and by variations in pathogenic phenotypes as well as inflammatory cellular infiltrates. Indeed, the degree of disease variability suggests that other Th immune responses, beyond the canonical Th2 pathway, have important and possibly complementary roles in pulmonary inflammation and AHR.
Despite significant attempts into understanding the basic immune mechanisms involved in asthma, the prevalence of this disorder has continued to rise and the lack of efficacy of anti-inflammatory treatments has been disappointing. The emerging body of evidence from human and mouse models has now demonstrated that in addition to Th2 cells, Th1, Th17, Treg and Th9 cells, each have unique abilities to influence airway inflammation and hyperresponsiveness. We predict that continued identification of different lymphoid cell subsets and delineation of their precise roles in asthma initiation and exacerbation will lead to new insights that ultimately will allow development of novel and better targeted therapeutic approaches for human asthma.
Footnotes
Declaration of interest: The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper
References
- Aarvak T, Chabaud M, Miossec P, Natvig JB. IL-17 is produced by some proinflammatory Th1/Th0 cells but not by Th2 cells. J Immunol. 1999;162:1246–1251. [PubMed] [Google Scholar]
- Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes. Nature. 1996;383:787–793. doi: 10.1038/383787a0. [DOI] [PubMed] [Google Scholar]
- Aggarwal S, Ghilardi N, Xie MH, de Sauvage FJ, Gurney AL. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J Biol Chem. 2003;278:1910–1914. doi: 10.1074/jbc.M207577200. [DOI] [PubMed] [Google Scholar]
- Akdis M, Verhagen J, Taylor A, Karamloo F, Karagiannidis C, Crameri R, Thunberg S, Deniz G, Valenta R, Fiebig H, et al. Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med. 2004;199:1567–1575. doi: 10.1084/jem.20032058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alvarez MJ, Olaguibel JM, Garcia BE, Rodriquez A, Tabar AI, Urbiola E. Airway inflammation in asthma and perennial allergic rhinitis. Relationship with nonspecific bronchial responsiveness and maximal airway narrowing. Allergy. 2000a;55:355–362. doi: 10.1034/j.1398-9995.2000.00312.x. [DOI] [PubMed] [Google Scholar]
- Alvarez MJ, Olaguibel JM, Garcia BE, Tabar AI, Urbiola E. Comparison of allergen-induced changes in bronchial hyperresponsiveness and airway inflammation between mildly allergic asthma patients and allergic rhinitis patients. Allergy. 2000b;55:531–539. doi: 10.1034/j.1398-9995.2000.00534.x. [DOI] [PubMed] [Google Scholar]
- Anderson GP. Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet. 2008;372:1107–1119. doi: 10.1016/S0140-6736(08)61452-X. [DOI] [PubMed] [Google Scholar]
- Anderson GP, Coyle AJ. TH2 and ‘TH2-like’ cells in allergy and asthma: pharmacological perspectives. Trends Pharmacol Sci. 1994;15:324–332. doi: 10.1016/0165-6147(94)90027-2. [DOI] [PubMed] [Google Scholar]
- Azzawi M, Bradley B, Jeffery PK, Frew AJ, Wardlaw AJ, Knowles G, Assoufi B, Collins JV, Durham S, Kay AB. Identification of activated T lymphocytes and eosinophils in bronchial biopsies in stable atopic asthma. Am Rev Respir Dis. 1990;142:1407–1413. doi: 10.1164/ajrccm/142.6_Pt_1.1407. [DOI] [PubMed] [Google Scholar]
- Barczyk A, Pierzchala W, Sozanska E. Interleukin-17 in sputum correlates with airway hyperresponsiveness to methacholine. Respir Med. 2003;97:726–733. doi: 10.1053/rmed.2003.1507. [DOI] [PubMed] [Google Scholar]
- Barrett NA, Austen KF. Innate cells and T helper 2 cell immunity in airway inflammation. Immunity. 2009;31:425–437. doi: 10.1016/j.immuni.2009.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrios RJ, Kheradmand F, Batts L, Corry DB. Asthma: pathology and pathophysiology. Arch Pathol Lab Med. 2006;130:447–451. doi: 10.5858/2006-130-447-APAP. [DOI] [PubMed] [Google Scholar]
- Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- Bettelli E, Korn T, Oukka M, Kuchroo VK. Induction and effector functions of T(H)17 cells. Nature. 2008;453:1051–1057. doi: 10.1038/nature07036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boguniewicz M, Martin RJ, Martin D, Gibson U, Celniker A, Williams M, Leung DY. The effects of nebulized recombinant interferon-gamma in asthmatic airways. J Allergy Clin Immunol. 1995;95:133–135. doi: 10.1016/s0091-6749(95)70162-1. [DOI] [PubMed] [Google Scholar]
- Bryan SA, O’Connor BJ, Matti S, Leckie MJ, Kanabar V, Khan J, Warrington SJ, Renzetti L, Rames A, Bock JA, et al. Effects of recombinant human interleukin-12 on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356:2149–2153. doi: 10.1016/S0140-6736(00)03497-8. [DOI] [PubMed] [Google Scholar]
- Bullens DM, Truyen E, Coteur L, Dilissen E, Hellings PW, Dupont LJ, Ceuppens JL. IL-17 mRNA in sputum of asthmatic patients: linking T cell driven inflammation and granulocytic influx? Respir Res. 2006;7:135. doi: 10.1186/1465-9921-7-135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burrows B, Martinez FD, Halonen M, Barbee RA, Cline MG. Association of asthma with serum IgE levels and skin-test reactivity to allergens. N Engl J Med. 1989;32:271–277. doi: 10.1056/NEJM198902023200502. [DOI] [PubMed] [Google Scholar]
- Bush KA, Farmer KM, Walker JS, Kirkham BW. Reduction of joint inflammation and bone erosion in rat adjuvant arthritis by treatment with interleukin-17 receptor IgG1 Fc fusion protein. Arthritis Rheum. 2002;46:802–805. doi: 10.1002/art.10173. [DOI] [PubMed] [Google Scholar]
- Cembrzynska-Nowak M, Szklarz E, Inglot AD, Teodorczyk-Injeyan JA. Elevated release of tumor necrosis factor-alpha and interferon-gamma by bronchoalveolar leukocytes from patients with bronchial asthma. Am Rev Respir Dis. 1993;147:291–295. doi: 10.1164/ajrccm/147.2.291. [DOI] [PubMed] [Google Scholar]
- Chakir J, Shannon J, Molet S, Fukakusa M, Elias J, Laviolette M, Boulet LP, Hamid Q. Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. J Allergy Clin Immunol. 2003;111:1293–1298. doi: 10.1067/mai.2003.1557. [DOI] [PubMed] [Google Scholar]
- Chen Y, Thai P, Zhao YH, Ho YS, DeSouza MM, Wu R. Stimulation of airway mucin gene expression by interleukin (IL)-17 through IL-6 paracrine/autocrine loop. J Biol Chem. 2003;278:17036–17043. doi: 10.1074/jbc.M210429200. [DOI] [PubMed] [Google Scholar]
- Cheng G, Arima M, Honda K, Hirata H, Eda F, Yoshida N, Fukushima F, Ishii Y, Fukuda T. Anti-interleukin-9 antibody treatment inhibits airway inflammation and hyperreactivity in mouse asthma model. Am J Respir Crit Care Med. 2002;166:409–416. doi: 10.1164/rccm.2105079. [DOI] [PubMed] [Google Scholar]
- Cheung PF, Wong CK, Lam CW. Molecular mechanisms of cytokine and chemokine release from eosinophils activated by IL-17A, IL-17F, and IL-23: implication for Th17 lymphocytes-mediated allergic inflammation. J Immunol. 2008;180:5625–5635. doi: 10.4049/jimmunol.180.8.5625. [DOI] [PubMed] [Google Scholar]
- Cho SH, Stanciu LA, Holgate ST, Johnston SL. Increased interleukin-4, interleukin-5, and interferon-gamma in airway CD4+ and CD8+ T cells in atopic asthma. Am J Respir Crit Care Med. 2005;171:224–230. doi: 10.1164/rccm.200310-1416OC. [DOI] [PubMed] [Google Scholar]
- Coffman RL, Seymour BW, Lebman DA, Hiraki DD, Christiansen JA, Shrader B, Cherwinski HM, Savelkoul HF, Finkelman FD, Bond MW, et al. The role of helper T cell products in mouse B cell differentiation and isotype regulation. Immunol Rev. 1988;102:5–28. doi: 10.1111/j.1600-065x.1988.tb00739.x. [DOI] [PubMed] [Google Scholar]
- Cohn L, Elias JA, Chupp GL. Asthma: mechanisms of disease persistence and progression. Annu Rev Immunol. 2004;22:789–815. doi: 10.1146/annurev.immunol.22.012703.104716. [DOI] [PubMed] [Google Scholar]
- Cohn L, Homer RJ, Marinov A, Rankin J, Bottomly K. Induction of airway mucus production By T helper 2 (Th2) cells: a critical role for interleukin 4 in cell recruitment but not mucus production. J Exp Med. 1997;186:1737–1747. doi: 10.1084/jem.186.10.1737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohn L, Tepper JS, Bottomly K. IL-4-independent induction of airway hyperresponsiveness by Th2, but not Th1, cells. J Immunol. 1998;161:3813–3816. [PubMed] [Google Scholar]
- Corren J. Allergic rhinitis and asthma: how important is the link? J Allergy Clin Immunol. 1997;99:S781–786. doi: 10.1016/s0091-6749(97)70127-1. [DOI] [PubMed] [Google Scholar]
- Corrigan CJ, Kay AB. CD4 T-lymphocyte activation in acute severe asthma. Relationship to disease severity and atopic status. Am Rev Respir Dis. 1990;141:970–977. doi: 10.1164/ajrccm/141.4_Pt_1.. [DOI] [PubMed] [Google Scholar]
- Coyle AJ, Le Gros G, Bertrand C, Tsuyuki S, Heusser CH, Kopf M, Anderson GP. Interleukin-4 is required for the induction of lung Th2 mucosal immunity. Am J Respir Cell Mol Biol. 1995;13:54–59. doi: 10.1165/ajrcmb.13.1.7598937. [DOI] [PubMed] [Google Scholar]
- Coyle AJ, Tsuyuki S, Bertrand C, Huang S, Aguet M, Alkan SS, Anderson GP. Mice lacking the IFN-gamma receptor have impaired ability to resolve a lung eosinophilic inflammatory response associated with a prolonged capacity of T cells to exhibit a Th2 cytokine profile. J Immunol. 1996;156:2680–2685. [PubMed] [Google Scholar]
- Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour B, Lucian L, To W, Kwan S, Churakova T, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421:744–748. doi: 10.1038/nature01355. [DOI] [PubMed] [Google Scholar]
- Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, Mitsdoerffer M, Strom TB, Elyaman W, Ho IC, et al. IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+ IL-10+ Foxp3(−) effector T cells. Nat Immunol. 2008;9:1347–1355. doi: 10.1038/ni.1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Douwes J, Gibson P, Pekkanen J, Pearce N. Non-eosinophilic asthma: importance and possible mechanisms. Thorax. 2002;57:643–648. doi: 10.1136/thorax.57.7.643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Durrant DM, Gaffen SL, Riesenfeld EP, Irvin CG, Metzger DW. Development of allergen-induced airway inflammation in the absence of T-bet regulation is dependent on IL-17. J Immunol. 2009;183:5293–5300. doi: 10.4049/jimmunol.0803109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erpenbeck VJ, Hohlfeld JM, Discher M, Krentel H, Hagenberg A, Braun A, Krug N. Increased expression of interleukin-9 messenger RNA after segmental allergen challenge in allergic asthmatics. Chest. 2003a;123:370S. [PubMed] [Google Scholar]
- Erpenbeck VJ, Hohlfeld JM, Volkmann B, Hagenberg A, Geldmacher H, Braun A, Krug N. Segmental allergen challenge in patients with atopic asthma leads to increased IL-9 expression in bronchoalveolar lavage fluid lymphocytes. J Allergy Clin Immunol. 2003b;111:1319–1327. doi: 10.1067/mai.2003.1485. [DOI] [PubMed] [Google Scholar]
- Fanta CH. Asthma. N Engl J Med. 2009;360:1002–1014. doi: 10.1056/NEJMra0804579. [DOI] [PubMed] [Google Scholar]
- Finotto S, Neurath MF, Glickman JN, Qin S, Lehr HA, Green FH, Ackerman K, Haley K, Galle PR, Szabo SJ, et al. Development of spontaneous airway changes consistent with human asthma in mice lacking T-bet. Science. 2002;295:336–338. doi: 10.1126/science.1065544. [DOI] [PubMed] [Google Scholar]
- Fu CL, Chuang YH, Chau LY, Chiang BL. Effects of adenovirus-expressing IL-10 in alleviating airway inflammation in asthma. J Gene Med. 2006;8:1393–1399. doi: 10.1002/jgm.974. [DOI] [PubMed] [Google Scholar]
- Fujiwara M, Hirose K, Kagami S, Takatori H, Wakashin H, Tamachi T, Watanabe N, Saito Y, Iwamoto I, Nakajima H. T-bet inhibits both TH2 cell-mediated eosinophil recruitment and TH17 cell-mediated neutrophil recruitment into the airways. J Allergy Clin Immunol. 2007;119:662–670. doi: 10.1016/j.jaci.2006.12.643. [DOI] [PubMed] [Google Scholar]
- Galli SJ. Complexity and redundancy in the pathogenesis of asthma: reassessing the roles of mast cells and T cells. J Exp Med. 1997;186:343–347. doi: 10.1084/jem.186.3.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gavett SH, Chen X, Finkelman F, Wills-Karp M. Depletion of murine CD4+ T lymphocytes prevents antigen-induced airway hyperreactivity and pulmonary eosinophilia. Am J Respir Cell Mol Biol. 1994;10:587–593. doi: 10.1165/ajrcmb.10.6.8003337. [DOI] [PubMed] [Google Scholar]
- Green RH, Brightling CE, Woltmann G, Parker D, Wardlaw AJ, Pavord ID. Analysis of induced sputum in adults with asthma: identification of subgroup with isolated sputum neutrophilia and poor response to inhaled corticosteroids. Thorax. 2002;57:875–879. doi: 10.1136/thorax.57.10.875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grunig G, Corry DB, Leach MW, Seymour BW, Kurup VP, Rennick DM. Interleukin-10 is a natural suppressor of cytokine production and inflammation in a murine model of allergic bronchopulmonary aspergillosis. J Exp Med. 1997;185:1089–1099. doi: 10.1084/jem.185.6.1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hansen G, Berry G, DeKruyff RH, Umetsu DT. Allergen-specific Th1 cells fail to counterbalance Th2 cell-induced airway hyperreactivity but cause severe airway inflammation. J Clin Invest. 1999;103:175–183. doi: 10.1172/JCI5155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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]
- Hashimoto T, Akiyama K, Kobayashi N, Mori A. Comparison of IL-17 production by helper T cells among atopic and nonatopic asthmatics and control subjects. Int Arch Allergy Immunol. 2005;137(Suppl 1):51–54. doi: 10.1159/000085432. [DOI] [PubMed] [Google Scholar]
- He R, Oyoshi MK, Jin H, Geha RS. Epicutaneous antigen exposure induces a Th17 response that drives airway inflammation after inhalation challenge. Proc Natl Acad Sci USA. 2007;104:15817–15822. doi: 10.1073/pnas.0706942104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hellings PW, Kasran A, Liu Z, Vandekerckhove P, Wuyts A, Overbergh L, Mathieu C, Ceuppens JL. Interleukin-17 orchestrates the granulocyte influx into airways after allergen inhalation in a mouse model of allergic asthma. Am J Respir Cell Mol Biol. 2003;28:42–50. doi: 10.1165/rcmb.4832. [DOI] [PubMed] [Google Scholar]
- Holgate ST. Pathogenesis of asthma. Clin Exp Allergy. 2008;38:872–897. doi: 10.1111/j.1365-2222.2008.02971.x. [DOI] [PubMed] [Google Scholar]
- Holgate ST, Polosa R. Treatment strategies for allergy and asthma. Nat Rev Immunol. 2008;8:218–230. doi: 10.1038/nri2262. [DOI] [PubMed] [Google Scholar]
- Infante-Duarte C, Horton HF, Byrne MC, Kamradt T. Microbial lipopeptides induce the production of IL-17 in Th cells. J Immunol. 2000;165:6107–6115. doi: 10.4049/jimmunol.165.11.6107. [DOI] [PubMed] [Google Scholar]
- Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, 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]
- Iwamoto I, Kumano K, Kasai M, Kurasawa K, Nakao A. Interleukin-12 prevents antigen-induced eosinophil recruitment into mouse airways. Am J Respir Crit Care Med. 1996;154:1257–1260. doi: 10.1164/ajrccm.154.5.8912732. [DOI] [PubMed] [Google Scholar]
- John M, Lim S, Seybold J, Jose P, Robichaud A, O’Connor B, Barnes PJ, Chung KF. Inhaled corticosteroids increase interleukin-10 but reduce macrophage inflammatory protein-1alpha, granulocyte-macrophage colony-stimulating factor, and interferon-gamma release from alveolar macrophages in asthma. Am J Respir Crit Care Med. 1998;157:256–262. doi: 10.1164/ajrccm.157.1.9703079. [DOI] [PubMed] [Google Scholar]
- Kawaguchi M, Kokubu F, Kuga H, Matsukura S, Hoshino H, Ieki K, Imai T, Adachi M, Huang SK. Modulation of bronchial epithelial cells by IL-17. J Allergy Clin Immunol. 2001a;108:804–809. doi: 10.1067/mai.2001.119027. [DOI] [PubMed] [Google Scholar]
- Kawaguchi M, Onuchic LF, Li XD, Essayan DM, Schroeder J, Xiao HQ, Liu MC, Krishnaswamy G, Germino G, Huang SK. Identification of a novel cytokine, ML-1, and its expression in subjects with asthma. J Immunol. 2001b;167:4430–4435. doi: 10.4049/jimmunol.167.8.4430. [DOI] [PubMed] [Google Scholar]
- Kay AB. T cells as orchestrators of the asthmatic response. Ciba Found Symp. 1997;206:56–67. discussion 67–70:106–110. [PubMed] [Google Scholar]
- Kay AB. The role of T lymphocytes in asthma. Chem Immunol Allergy. 2006;91:59–75. doi: 10.1159/000090230. [DOI] [PubMed] [Google Scholar]
- Kearley J, Barker JE, Robinson DS, Lloyd CM. Resolution of airway inflammation and hyperreactivity after in vivo transfer of CD4+CD25+ regulatory T cells is interleukin 10 dependent. J Exp Med. 2005;202:1539–1547. doi: 10.1084/jem.20051166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kearley J, Robinson DS, Lloyd CM. CD4+CD25+ regulatory T cells reverse established allergic airway inflammation and prevent airway remodeling. J Allergy Clin Immunol. 2008;122:617–624. e616. doi: 10.1016/j.jaci.2008.05.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiley J, Smith R, Noel P. Asthma phenotypes. Curr Opin Pulm Med. 2007;13:19–23. doi: 10.1097/MCP.0b013e328011b84b. [DOI] [PubMed] [Google Scholar]
- Kips JC, Brusselle GJ, Joos GF, Peleman RA, Tavernier JH, Devos RR, Pauwels RA. Interleukin-12 inhibits antigen-induced airway hyperresponsiveness in mice. Am J Respir Crit Care Med. 1996;153:535–539. doi: 10.1164/ajrccm.153.2.8564093. [DOI] [PubMed] [Google Scholar]
- Kolls JK, Kanaly ST, Ramsay AJ. Interleukin-17: an emerging role in lung inflammation. Am J Respir Cell Mol Biol. 2003;28:9–11. doi: 10.1165/rcmb.2002-0255PS. [DOI] [PubMed] [Google Scholar]
- 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]
- Laan M, Palmberg L, Larsson K, Linden A. Free, soluble interleukin-17 protein during severe inflammation in human airways. Eur Respir J. 2002;19:534–537. doi: 10.1183/09031936.02.00280902. [DOI] [PubMed] [Google Scholar]
- Lane SJ, Lee TH. Mast cell effector mechanisms. J Allergy Clin Immunol. 1996;98:S67–71. doi: 10.1016/s0091-6749(96)80131-x. discussion S71–72. [DOI] [PubMed] [Google Scholar]
- Larche M. Regulatory T cells in allergy and asthma. Chest. 2007;132:1007–1014. doi: 10.1378/chest.06-2434. [DOI] [PubMed] [Google Scholar]
- Larche M, Robinson DS, Kay AB. The role of T lymphocytes in the pathogenesis of asthma. J Allergy Clin Immunol. 2003;111:450–463. doi: 10.1067/mai.2003.169. quiz 464. [DOI] [PubMed] [Google Scholar]
- Leckie MJ, ten Brinke A, Khan J, Diamant Z, O’Connor BJ, Walls CM, Mathur AK, Cowley HC, Chung KF, Djukanovic R, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyperresponsiveness, and the late asthmatic response. Lancet. 2000;356:2144–2148. doi: 10.1016/s0140-6736(00)03496-6. [DOI] [PubMed] [Google Scholar]
- Lewkowich IP, Herman NS, Schleifer KW, Dance MP, Chen BL, Dienger KM, Sproles AA, Shah JS, Kohl J, Belkaid Y, Wills-Karp M. CD4+CD25+ T cells protect against experimentally induced asthma and alter pulmonary dendritic cell phenotype and function. J Exp Med. 2005;202:1549–1561. doi: 10.1084/jem.20051506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim S, Crawley E, Woo P, Barnes PJ. Haplotype associated with low interleukin-10 production in patients with severe asthma. Lancet. 1998;352:113. doi: 10.1016/S0140-6736(98)85018-6. [DOI] [PubMed] [Google Scholar]
- Liu Y, Teige I, Birnir B, Issazadeh-Navikas S. Neuron-mediated generation of regulatory T cells from encephalitogenic T cells suppresses EAE. Nat Med. 2006;12:518–525. doi: 10.1038/nm1402. [DOI] [PubMed] [Google Scholar]
- Longphre M, Li D, Gallup M, Drori E, Ordonez CL, Redman T, Wenzel S, Bice DE, Fahy JV, Basbaum C. Allergen-induced IL-9 directly stimulates mucin transcription in respiratory epithelial cells. J Clin Invest. 1999;104:1375–1382. doi: 10.1172/JCI6097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Louten J, Boniface K, de Waal Malefyt R. Development and function of TH17 cells in health and disease. J Allergy Clin Immunol. 2009;123:1004–1011. doi: 10.1016/j.jaci.2009.04.003. [DOI] [PubMed] [Google Scholar]
- Lu LF, Lind EF, Gondek DC, Bennett KA, Gleeson MW, Pino-Lagos K, Scott ZA, Coyle AJ, Reed JL, Van Snick J, et al. Mast cells are essential intermediaries in regulatory T-cell tolerance. Nature. 2006;442:997–1002. doi: 10.1038/nature05010. [DOI] [PubMed] [Google Scholar]
- Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming growth factor-beta and induction of the nuclear receptor RORgammat. Nat Immunol. 2008;9:641–649. doi: 10.1038/ni.1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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]
- Masoli M, Fabian D, Holt S, Beasley R. The global burden of asthma: executive summary of the GINA Dissemination Committee report. Allergy. 2004;59:469–478. doi: 10.1111/j.1398-9995.2004.00526.x. [DOI] [PubMed] [Google Scholar]
- Matangkasombut P, Pichavant M, Dekruyff RH, Umetsu DT. Natural killer T cells and the regulation of asthma. Mucosal Immunol. 2009;2:383–392. doi: 10.1038/mi.2009.96. [DOI] [PubMed] [Google Scholar]
- McLane MP, Haczku A, van de Rijn M, Weiss C, Ferrante V, MacDonald D, Renauld JC, Nicolaides NC, Holroyd KJ, Levitt RC. Interleukin-9 promotes allergen-induced eosinophilic inflammation and airway hyperresponsiveness in transgenic mice. Am J Respir Cell Mol Biol. 1998;19:713–720. doi: 10.1165/ajrcmb.19.5.3457. [DOI] [PubMed] [Google Scholar]
- McMillan SJ, Bishop B, Townsend MJ, McKenzie AN, Lloyd CM. The absence of interleukin 9 does not affect the development of allergen-induced pulmonary inflammation nor airway hyperreactivity. J Exp Med. 2002;195:51–57. doi: 10.1084/jem.20011732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molet S, Hamid Q, Davoine F, Nutku E, Taha R, Page N, Olivenstein R, Elias J, Chakir J. IL-17 is increased in asthmatic airways and induces human bronchial fibroblasts to produce cytokines. J Allergy Clin Immunol. 2001;108:430–438. doi: 10.1067/mai.2001.117929. [DOI] [PubMed] [Google Scholar]
- Molet SM, Hamid QA, Hamilos DL. IL-11 and IL-17 expression in nasal polyps: relationship to collagen deposition and suppression by intranasal fluticasone propionate. Laryngoscope. 2003;113:1803–1812. doi: 10.1097/00005537-200310000-00027. [DOI] [PubMed] [Google Scholar]
- Munthe-Kaas MC, Carlsen KH, Haland G, Devulapalli CS, Gervin K, Egeland T, Carlsen KL, Undlien D. T cell-specific T-box transcription factor haplotype is associated with allergic asthma in children. J Allergy Clin Immunol. 2008;121:51–56. doi: 10.1016/j.jaci.2007.07.068. [DOI] [PubMed] [Google Scholar]
- Murphy CA, Langrish CL, Chen Y, Blumenschein W, McClanahan T, Kastelein RA, Sedgwick JD, Cua DJ. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med. 2003;198:1951–1957. doi: 10.1084/jem.20030896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakae S, Ho LH, Yu M, Monteforte R, Iikura M, Suto H, Galli SJ. Mast cell-derived TNF contributes to airway hyperreactivity, inflammation, and TH2 cytokine production in an asthma model in mice. J Allergy Clin Immunol. 2007;120:48–55. doi: 10.1016/j.jaci.2007.02.046. [DOI] [PubMed] [Google Scholar]
- Nakae S, Komiyama Y, Nambu A, Sudo K, Iwase M, Homma I, Sekikawa K, Asano M, Iwakura Y. Antigen-specific T cell sensitization is impaired in IL-17-deficient mice, causing suppression of allergic cellular and humoral responses. Immunity. 2002;17:375–387. doi: 10.1016/s1074-7613(02)00391-6. [DOI] [PubMed] [Google Scholar]
- Nakae S, Nambu A, Sudo K, Iwakura Y. Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice. J Immunol. 2003;171:6173–6177. doi: 10.4049/jimmunol.171.11.6173. [DOI] [PubMed] [Google Scholar]
- Nakao F, Ihara K, Kusuhara K, Sasaki Y, Kinukawa N, Takabayashi A, Nishima S, Hara T. Association of IFN-gamma and IFN regulatory factor 1 polymorphisms with childhood atopic asthma. J Allergy Clin Immunol. 2001;107:499–504. doi: 10.1067/mai.2001.113051. [DOI] [PubMed] [Google Scholar]
- O’Garra A, Barrat FJ, Castro AG, Vicari A, Hawrylowicz C. Strategies for use of IL-10 or its antagonists in human disease. Immunol Rev. 2008;223:114–131. doi: 10.1111/j.1600-065X.2008.00635.x. [DOI] [PubMed] [Google Scholar]
- Oboki K, Ohno T, Saito H, Nakae S. Th17 and allergy. Allergol Int. 2008;57:121–134. doi: 10.2332/allergolint.R-07-160. [DOI] [PubMed] [Google Scholar]
- Oda N, Canelos PB, Essayan DM, Plunkett BA, Myers AC, Huang SK. Interleukin-17F induces pulmonary neutrophilia and amplifies antigen-induced allergic response. Am J Respir Crit Care Med. 2005;171:12–18. doi: 10.1164/rccm.200406-778OC. [DOI] [PubMed] [Google Scholar]
- Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, Wang Y, Hood L, Zhu Z, Tian Q, Dong C. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005;6:1133–1141. doi: 10.1038/ni1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raby BA, Hwang ES, Van Steen K, Tantisira K, Peng S, Litonjua A, Lazarus R, Giallourakis C, Rioux JD, Sparrow D, et al. T-bet polymorphisms are associated with asthma and airway hyperresponsiveness. Am J Respir Crit Care Med. 2006;173:64–70. doi: 10.1164/rccm.200503-505OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rahman MS, Yamasaki A, Yang J, Shan L, Halayko AJ, Gounni AS. IL-17A induces eotaxin-1/CC chemokine ligand 11 expression in human airway smooth muscle cells: role of MAPK (Erk1/2, JNK, and p38) pathways. J Immunol. 2006;177:4064–4071. doi: 10.4049/jimmunol.177.6.4064. [DOI] [PubMed] [Google Scholar]
- Reisinger J, Triendl A, Kuchler E, Bohle B, Krauth MT, Rauter I, Valent P, Koenig F, Valenta R, Niederberger V. IFN-gamma-enhanced allergen penetration across respiratory epithelium augments allergic inflammation. J Allergy Clin Immunol. 2005;115:973–981. doi: 10.1016/j.jaci.2005.01.021. [DOI] [PubMed] [Google Scholar]
- Robinson DS. Regulatory T cells and asthma. Clin Exp Allergy. 2009;39:1314–1323. doi: 10.1111/j.1365-2222.2009.03301.x. [DOI] [PubMed] [Google Scholar]
- Robinson DS, Bentley AM, Hartnell A, Kay AB, Durham SR. Activated memory T helper cells in bronchoalveolar lavage fluid from patients with atopic asthma: relation to asthma symptoms, lung function, and bronchial responsiveness. Thorax. 1993;48:26–32. doi: 10.1136/thx.48.1.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson DS, Hamid Q, Ying S, Tsicopoulos A, Barkans J, Bentley AM, Corrigan C, Durham SR, Kay AB. Predominant TH2-like bronchoalveolar T-lymphocyte population in atopic asthma. N Engl J Med. 1992;326:298–304. doi: 10.1056/NEJM199201303260504. [DOI] [PubMed] [Google Scholar]
- Rossi GL, Olivieri D. Does the mast cell still have a key role in asthma? Chest. 1997;112:523–529. doi: 10.1378/chest.112.2.523. [DOI] [PubMed] [Google Scholar]
- Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, Treuting P, Siewe L, Roers A, Henderson WR, Jr, et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity. 2008;28:546–558. doi: 10.1016/j.immuni.2008.02.017. [DOI] [PubMed] [Google Scholar]
- Schnyder-Candrian S, Togbe D, Couillin I, Mercier I, Brombacher F, Quesniaux V, Fossiez F, Ryffel B, Schnyder B. Interleukin-17 is a negative regulator of established allergic asthma. J Exp Med. 2006;203:2715–2725. doi: 10.1084/jem.20061401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schwarze J, Hamelmann E, Cieslewicz G, Tomkinson A, Joetham A, Bradley K, Gelfand EW. Local treatment with IL-12 is an effective inhibitor of airway hyperresponsiveness and lung eosinophilia after airway challenge in sensitized mice. J Allergy Clin Immunol. 1998;102:86–93. doi: 10.1016/s0091-6749(98)70058-2. [DOI] [PubMed] [Google Scholar]
- Schwarzenberger P, La Russa V, Miller A, Ye P, Huang W, Zieske A, Nelson S, Bagby GJ, Stoltz D, Mynatt RL, et al. IL-17 stimulates granulopoiesis in mice: use of an alternate, novel gene therapy-derived method for in vivo evaluation of cytokines. J Immunol. 1998;161:6383–6389. [PubMed] [Google Scholar]
- Sharkhuu T, Matthaei KI, Forbes E, Mahalingam S, Hogan SP, Hansbro PM, Foster PS. Mechanism of interleukin-25 (IL-17E)-induced pulmonary inflammation and airways hyper-reactivity. Clin Exp Allergy. 2006;36:1575–1583. doi: 10.1111/j.1365-2222.2006.02595.x. [DOI] [PubMed] [Google Scholar]
- Shimbara A, Christodoulopoulos P, Soussi-Gounni A, Olivenstein R, Nakamura Y, Levitt RC, Nicolaides NC, Holroyd KJ, Tsicopoulos A, Lafitte JJ, et al. IL-9 and its receptor in allergic and nonallergic lung disease: increased expression in asthma. J Allergy Clin Immunol. 2000;105:108–115. doi: 10.1016/s0091-6749(00)90185-4. [DOI] [PubMed] [Google Scholar]
- Simpson JL, Grissell TV, Douwes J, Scott RJ, Boyle MJ, Gibson PG. Innate immune activation in neutrophilic asthma and bronchiectasis. Thorax. 2007;62:211–218. doi: 10.1136/thx.2006.061358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simpson JL, Scott R, Boyle MJ, Gibson PG. Inflammatory subtypes in asthma: assessment and identification using induced sputum. Respirology. 2006;11:54–61. doi: 10.1111/j.1440-1843.2006.00784.x. [DOI] [PubMed] [Google Scholar]
- Soroosh P, Doherty TA. Th9 and allergic disease. Immunology. 2009;127:450–458. doi: 10.1111/j.1365-2567.2009.03114.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steinman L. A brief history of T(H)17, the first major revision in the T(H)1/ T(H)2 hypothesis of T cell-mediated tissue damage. Nat Med. 2007;13:139–145. doi: 10.1038/nm1551. [DOI] [PubMed] [Google Scholar]
- Temann UA, Geba GP, Rankin JA, Flavell RA. Expression of interleukin 9 in the lungs of transgenic mice causes airway inflammation, mast cell hyperplasia, and bronchial hyperresponsiveness. J Exp Med. 1998;188:1307–1320. doi: 10.1084/jem.188.7.1307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Till SJ, Francis JN, Nouri-Aria K, Durham SR. Mechanisms of immunotherapy. J, Allergy Clin Immunol. 2004;113:1025–1034. doi: 10.1016/j.jaci.2004.03.024. quiz 1035. [DOI] [PubMed] [Google Scholar]
- Tsicopoulos A, Shimbara A, de Nadai P, Aldewachi O, Lamblin C, Lassalle P, Walls AF, Senechal S, Levitt RC, Darras J, et al. Involvement of IL-9 in the bronchial phenotype of patients with nasal polyposis. J Allergy Clin Immunol. 2004;113:462–469. doi: 10.1016/j.jaci.2003.12.009. [DOI] [PubMed] [Google Scholar]
- Umetsu DT, McIntire JJ, Akbari O, Macaubas C, DeKruyff RH. Asthma: an epidemic of dysregulated immunity. Nat Immunol. 2002;3:715–720. doi: 10.1038/ni0802-715. [DOI] [PubMed] [Google Scholar]
- van de Kant KD, Klaassen EM, Jobsis Q, Nijhuis AJ, van Schayck OC, Dompeling E. Early diagnosis of asthma in young children by using non-invasive biomarkers of airway inflammation and early lung function measurements: study protocol of a case-control study. BMC Publ Health. 2009;9:210. doi: 10.1186/1471-2458-9-210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGF-beta 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]
- Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, Buer J, Martin B, Wilhelm C, Stockinger B. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat Immunol. 2008;9:1341–1346. doi: 10.1038/ni.1659. [DOI] [PubMed] [Google Scholar]
- Vignola AM, Chanez P, Campbell AM, Souques F, Lebel B, Enander I, Bousquet J. Airway inflammation in mild intermittent and in persistent asthma. Am J Respir Crit Care Med. 1998;157:403–409. doi: 10.1164/ajrccm.157.2.96-08040. [DOI] [PubMed] [Google Scholar]
- Wakashin H, Hirose K, Maezawa Y, Kagami S, Suto A, Watanabe N, Saito Y, Hatano M, Tokuhisa T, Iwakura Y, et al. IL-23 and Th17 cells enhance Th2-cell-mediated eosinophilic airway inflammation in mice. Am J Respir Crit Care Med. 2008;178:1023–1032. doi: 10.1164/rccm.200801-086OC. [DOI] [PubMed] [Google Scholar]
- Walker C, Bauer W, Braun RK, Menz G, Braun P, Schwarz F, Hansel TT, Villiger B. Activated T cells and cytokines in bronchoalveolar lavages from patients with various lung diseases associated with eosinophilia. Am J Respir Crit Care Med. 1994;150:1038–1048. doi: 10.1164/ajrccm.150.4.7921434. [DOI] [PubMed] [Google Scholar]
- Wang J, Homer RJ, Chen Q, Elias JA. Endogenous and exogenous IL-6 inhibit aeroallergen-induced Th2 inflammation. J Immunol. 2000;165:4051–4061. doi: 10.4049/jimmunol.165.7.4051. [DOI] [PubMed] [Google Scholar]
- Wenzel S, Wilbraham D, Fuller R, Getz EB, Longphre M. Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet. 2007;370:1422–1431. doi: 10.1016/S0140-6736(07)61600-6. [DOI] [PubMed] [Google Scholar]
- Wenzel SE, Schwartz LB, Langmack EL, Halliday JL, Trudeau JB, Gibbs RL, Chu HW. Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respir Crit Care Med. 1999;160:1001–1008. doi: 10.1164/ajrccm.160.3.9812110. [DOI] [PubMed] [Google Scholar]
- Wilder JA, Collie DD, Bice DE, Tesfaigzi Y, Lyons CR, Lipscomb MF. Ovalbumin aerosols induce airway hyperreactivity in naive DO11.10 T cell receptor transgenic mice without pulmonary eosinophilia or OVA-specific antibody. J Leukoc Biol. 2001;69:538–547. [PubMed] [Google Scholar]
- Wills-Karp M. Immunologic basis of antigen-induced airway hyperresponsiveness. Annu Rev Immunol. 1999;17:255–281. doi: 10.1146/annurev.immunol.17.1.255. [DOI] [PubMed] [Google Scholar]
- Wing K, Fehervari Z, Sakaguchi S. Emerging possibilities in the development and function of regulatory T cells. Int Immunol. 2006;18:991–1000. doi: 10.1093/intimm/dxl044. [DOI] [PubMed] [Google Scholar]
- Wong CK, Ho CY, Ko FW, Chan CH, Ho AS, Hui DS, Lam CW. Proinflammatory cytokines (IL-17, IL-6, IL-18 and IL-12) and Th cytokines (IFN-gamma, IL-4, IL-10 and IL-13) in patients with allergic asthma. Clin Exp Immunol. 2001;125:177–183. doi: 10.1046/j.1365-2249.2001.01602.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Woolcock AJ, Peat J. What is the relationship between airway hyperresponsiveness and atopy? Am J Respir Crit Care Med. 2000;161:S215–S217. doi: 10.1164/ajrccm.161.supplement_2.a1q4-14. [DOI] [PubMed] [Google Scholar]
- Ying S, Humbert M, Barkans J, Corrigan CJ, Pfister R, Menz G, Larche M, Robinson DS, Durham SR, Kay AB. Expression of IL-4 and IL-5 mRNA and protein product by CD4+ and CD8+ T cells, eosinophils, and mast cells in bronchial biopsies obtained from atopic and nonatopic (intrinsic) asthmatics. J Immunol. 1997;158:3539–3544. [PubMed] [Google Scholar]
- Ying S, Meng Q, Kay AB, Robinson DS. Elevated expression of interleukin-9 mRNA in the bronchial mucosa of atopic asthmatics and allergen-induced cutaneous late-phase reaction: relationships to eosinophils, mast cells and T lymphocytes. Clin Exp Allergy. 2002;32:866–871. doi: 10.1046/j.1365-2222.2002.01376.x. [DOI] [PubMed] [Google Scholar]
- Zhou L, Chong MM, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity. 2009;30:646–655. doi: 10.1016/j.immuni.2009.05.001. [DOI] [PubMed] [Google Scholar]


