Abstract
Asthma is a common, disabling inflammatory respiratory disease that has increased in frequency and severity in developed nations. We review studies of murine allergic airway disease (MAAD2) and human asthma that evaluate the importance of Th2 cytokines, Th2 response-promoting cytokines, IL-17 and pro- and anti-inflammatory cytokines in MAAD and human asthma. We discuss murine studies that directly stimulate airways with specific cytokines or delete, inactivate, neutralize or block specific cytokines or their receptors, as well as controversial issues, including the roles of IL-5, IL-17 and IL-13Rα2 in MAAD and IL-4Rα expression by specific cell types. Studies of human asthmatic cytokine gene and protein expression, linkage of cytokine polymorphisms to asthma, cytokine responses to allergen stimulation and clinical responses to cytokine antagonists are discussed as well. Results of these analyses establish the importance of specific cytokines in MAAD and human asthma and have therapeutic implications.
Introduction
Atopic asthma is an inflammatory respiratory disorder that, along with other allergic conditions, has more than doubled in prevalence and severity in developed countries during the past 60 years. Atopic asthma is common; approximately 34.1 million Americans develop asthma during their lifetime and approximately 70% of individuals with this diagnosis have allergies (1, 2). A great deal has been learned about the pathogenesis of asthma during the past 30 years and much of this new knowledge relates to the roles of cytokines in asthma pathogenesis. Inhalation of allergens stimulates both bone marrow- and non-bone marrow-derived cells of the innate immune system to secrete cytokines that promote antigen presentation to CD4+ T cells and influence both antigen-presenting cells and the T cells themselves in a way the promotes a Th2 response (3). Th2 cytokines — IL-4, IL-5, IL-9 and IL-13 (4) — then induce the changes in the airways and lung parenchyma that are associated with asthma: airway eosinophilia, pulmonary lymphocytosis and mastocytosis, alternative macrophage activation, epithelial cell proliferation with goblet cell hyperplasia (GCH) and increased mucus secretion, smooth muscle hyperplasia, hypertrophy and hypercontractility, subepithelial fibrosis, IgE secretion, increased production of chemokines that attract T cells, eosinophils, neutrophils and mast cells or their precursors to the lungs, and airway hyperresponsiveness (AHR, defined as increased sensitivity to agents, such as cholinergic agents and other stimuli that cause smooth contraction that increases airway resistance by narrowing airways) (4, 5). Together, these changes in airway structure and function result in the clinical picture of asthma: episodic difficulty in breathing with wheezing and/or coughing that is caused by reversible airway obstruction and is ameliorated by inhalation of β-adrenergic agonists.
Cytokine roles in murine allergic airway disease
The importance of Th2 cytokines
Experiments performed largely in mice have provided a consensus view of cytokine roles in asthma pathophysiology that stresses the importance of the Th2 cytokines. IL-4 and IL-13 stimulate multiple features of asthma (Table I) by binding and signaling through specific receptors; IL-4 binds to both the type I and type II IL-4Rs while IL-13 binds selectively to the type II IL-4R. Both IL-4Rs signal through IL-4Rα, which activates the transcription factor, Stat6 (6). Each IL-4R additional contains a second polypeptide that is required to activate IL-4Rα chain: the cytokine receptor common γ chain (γc) for the type I IL-4R and IL-13Rα1 for the type II IL-4R. Because both IL-4 and IL-13 bind to the type II IL-4R, there are probably no unique IL-4R-mediated effects of IL-13, while selective binding of IL-4 by the type I IL-4R and the expression of γc but not IL-13Rα1 by some bone marrow-derived cells, including T cells, most B cells (in the mouse) and mast cells, accounts for stimulation of these cell types by IL-4 but not IL-13 (6). Studies with mice deficient in IL-13Rα1 demonstrate that signaling through the type II IL-4R is required to induce GCH and AHR, but may be less important than signaling through the type I IL-4R for induction of airway eosinophilia (7, 8). IL-13 is more important than IL-4 for induction of GCH, AHR and chronic remodeling changes, including smooth muscle hyperplasia and subepithelial fibrosis (9, 10), even though either cytokine can stimulate all of these features (11-13). The considerably higher lung levels of IL-13 than IL-4 in murine allergic airway disease (MAAD) (8) probably account to a large extent for the predominant role of IL-13, although type I IL-4R-mediated IL-4 induction of IL-10 and IFN-γ (14), which can inhibit AHR and GCH (15, 16), may also contribute. Differences in the binding of IL-4 and IL-13 to the type II IL-4R most likely also contribute to the dominant role of IL-13 in AHR and GCH induction: IL-4 initially interacts with the type II IL-4R by binding with relatively high affinity to IL-4Rα; the IL-4/IL-4Rα complex then recruits IL-13Rα1 to form the signaling complex. In contrast, IL-13 initially binds to IL-13Rα1 with relatively low affinity; the resulting complex then binds IL-4Rα to form the signaling complex. The higher affinity of the initial binding step for IL-4 than IL-13 appears to allow low concentrations of IL-4 to signal more effectively than low concentrations of IL-13, while the higher cell membrane concentration of IL-13Rα1 than IL-4Rα appears to allow high concentrations of IL-13 to signal more strongly than IL-4 through the type II IL-4R receptor (17, 18).
Table I. Cytokines that directly promote features of asthma.
Feature | Cytokines Implicated |
---|---|
Eosinophilia | IL-4, IL-5, IL-13 |
Goblet Cell Metaplasia | IL-4, IL-13 |
Airway Hyperresponsiveness | IL-4, IL-13, IL-17A |
IgE Production | IL-4, IL-13 |
Mastocytosis | IL-3, IL-9 |
Alternative Macrophage Activation | IL-4, IL-13 |
Smooth Muscle Remodeling | IL-4, IL-13 |
Th2 Induction/Maintenance | IL-4, IL-9, IL-17E (IL-25), IL-33, TSLP |
Subepithelial fibrosis | IL-4, IL-13 |
In contrast to the greater role of IL-13 than IL-4 in inducing and maintaining AHR, GCH and airway remodeling, both IL-4 and IL-13 appear to contribute importantly to alternative macrophage and dendritic cell activation (macrophages and dendritic cells express both γc and IL-13Rα1 and thus, both IL-4Rs) (19-21) and IL-4 has a greater role than IL-13 in the induction of airway eosinophilia (8, 10, 12). The reason for IL-4’s stronger stimulation of airway eosinophilia is presently unclear, because both IL-4 and IL-13 signal through the type II IL-4R to induce production of chemokines, including eotaxin-1 (CCL11) and eotaxin-2 (CCL24), which attract eosinophils to the lungs (8). IL-4 effects on other cell types, such as vascular endothelial cells, or on eosinophils themselves that may respond primarily through the type I IL-4R, may account for the difference. Expression of γc, but not IL-13Rα1 by most murine B cells also accounts for the unique requirement for IL-4, in mice, for induction of IgE responses to T cell-dependent Ags (8, 22, 23). The most critical role of IL-4 in allergic airway disease, however, is in induction of the Th2 cytokine response. This role is not shared by IL-13, because T cells express γc but not IL-13Rα1 (24, 25). Although IL-4 is not an absolute requirement for naïve T cell differentiation into Th2 cells (26), IL-4-induced Stat6 stimulation of GATA3, the transcription factor required for Th2 differentiation (27), amplifies Th2 differentiation sufficiently to be required in many instances for the development of a predominant Th2 response (28); this requirement has been observed in most, but not all mouse models of asthma (29-35). Consistent with this, the presence of supernormal concentrations of IL-4 allows the generation of Th2 responses to Ags administered via the airways in an otherwise tolerogenic manner, suggesting that airway IL-4 responses to one Ag probably increase the risk of developing allergic responses to other inhaled Ags (36).
The importance of IL-4 and IL-13 in the induction and maintenance of murine allergic airway disease have been demonstrated in several ways: 1) induction of key features of allergic airway disease by intranasal (i.n.) or intratracheal (i.t.) administration of either cytokine (8, 9, 12, 37); 2) transgenic overexpression of either cytokine in airway epithelial cells (11, 13); 3) suppression of allergen-induced allergic airway disease by administration of neutralizing mAbs to IL-4 or IL-13 (33, 38), soluble IL-4Rα (31) or soluble IL-13Rα2 (9, 10), a blocking anti-IL-4Rα mAb (39), or a mutant IL-4 that acts as an IL-4R antagonist (40, 41); 4) genetic deletion of IL-4 (42, 43), IL-13 (44), IL-4Rα (8) or IL-13Rα1 (7, 8); or 5) suppression of one of these molecules with anti-sense DNA (45). Results differ in the above-cited papers, depending on the precise allergen, immunization schedule, mouse strain and inhibitory procedure employed. However, with a few exceptions, inhibition of IL-4 suppresses the development of a pulmonary Th2 response and allergic airway disease as well as already established airway eosinophilia and IgE production, but does not affect established AHR or GCH (33); inhibition of IL-13 can suppress even established AHR and GCH but has less effect on eosinophilia or IgE production (10); absence of IL-13Rα1 prevents AHR and GCH but has little effect on the development of an IgE response and only modestly prevents development of eosinophilia (8) and inhibition of IL-4Rα suppresses all features of even established allergic airway disease with the exception of mastocytosis (45). However, currently available anti-IL-4Rα mAbs have been less effective than direct IL-13 antagonists at suppressing GCH and AHR, most likely because these anti-IL-4Rα mAbs do not remove cell membrane IL-4Rα and are less effective at blocking IL-4Rα signaling by IL-13 than by IL-4 (F. Finkelman, unpublished data).
Although IL-4 and IL-13 are generally the cytokines accorded primacy in the induction and maintenance of allergic airway disease, the consensus view also confers importance on IL-5 and, to a lesser extent, IL-9. There is general agreement that IL-5 is critical for stimulating eosinophil development, survival, activation and response to other cytokines, including the eotaxins (46, 47). The importance of airway eosinophils in the induction and maintenance of the airway Th2 response, AHR and GCH is debated and may be mouse strain- and allergen-dependent (see section on controversies, below); however, IL-5-dependent eosinophils are generally agreed to make an important contribution to asthmatic airway remodeling, especially sub-epithelial fibrosis (48, 49).
Airway overexpression of IL-9 can induce the full allergic airway disease phenotype, including eosinophilia, GCH and AHR (50); however, these effects result primarily from IL-9 induction of increased IL-5 and IL-13 secretion by non-T cells (51) and IL-9 has not been found to be important in most studies of MAAD (52), although there is one exception (53). IL-9 does, however, directly promote airway mastocytosis (54) and migration of mast cell precursors to the lungs; an effect not shared by IL-3, IL-4, IL-5, or IL-13 (55).
Cytokines that stimulate allergic airway disease by promoting a Th2 response
Airway over-expression of other cytokines, including IL-25 (also called IL-17E), IL-33 and thymic stromal lymphopoietin (TSLP) induces and promotes allergic airway disease indirectly by stimulating production of IL-4, IL-5 and/or IL-13 (29, 56, 57). Unlike IL-9, which is produced by T cells, mast cells and basophils, allergen stimulation of epithelial cells appears to be important for the initial production of IL-25 (58), IL-33 (3) and TSLP (59). Each of the 4 cytokines that promote allergic airway disease indirectly by promoting IL-4, IL-5 and/or IL-13 secretion has been shown to be important in at least some mouse models of asthma in studies in which the cytokine or its receptor was blocked by inhibitory proteins or deleted genetically (53, 60-62). In addition to direct effects on T cells that can promote Th2 cytokine production, IL-9 and IL-33 stimulate non-T cells to secrete Th2-associated cytokines (51, 57, 63) and TSLP stimulates dendritic cells to express the Th2 differentiation-promoting co-receptor, OX40 ligand (64). IL-25 has also been reported to promote AHR independently of its stimulatory effects of Th2 cytokine secretion; neutralization of IL-25 when already sensitized mice were exposed to aerosolized Ag suppressed AHR without influencing airway inflammation or Th2 cytokine production and inhaled IL-25 induced AHR in mice genetically deficient in IL-4, IL-5, IL-9 and IL-13 (60).
IL-25, IL-33 and TSLP are not the only cytokines implicated in Th2 response induction. AHR, Th2 cytokine production and IgE levels are significantly reduced in allergen-immunized IL-1α/IL-1β-deficient mice and increased in IL-1R antagonist-deficient mice (65). Similarly, GM-CSF promotes development of a Th2 response, even in the absence of IL-4 (66). The GM-CSF contribution to AAD is mediated, at least in part, by stimulating activation, airway migration and proliferation of myeloid dendritic cells that can present Ag in a way that promotes Th2 differentiation (67). However, although GM-CSF deficiency or neutralization can inhibit diesel exhaust particle-induced AHR (68) and allergen-induced development of GCH, airway eosinophilia and IL-5 production (69), it has not always inhibited allergen-induced IL-4 or IL-13 production or AHR (69).
The effects of the IL-1 family cytokine, IL-18, on Th2 cytokine production and MAAD are more complex. Genetic deficiency of IL-18 has been reported to exacerbate (70) or inhibit (71) different MAAD models and the effects of airway IL-18 inoculation on MAAD vary and depend on time and route of IL-18 relative to allergen administration. This variability most likely reflects the different effects of IL-18 when it interacts with different cytokines; specifically, IL-18 enhances IL-12 induction of a Th1 response, IL-2 induction of a Th2 response and IL-3 induction of Th2 cytokine production by mast cells and basophils (72).
Cytokine suppression of allergic airway disease
Not all cytokines promote allergic airway disease; IL-12, type I and type II IFNs and the “anti-inflammatory” cytokines, IL-10 and TGF-β, suppress this disorder (15, 16, 73-80), although there are caveats to this generalization that probably reflect the pleomorphic effects of each of these cytokines. Endogenously produced IL-12p40, a component of IL-12 and IL-23, is required to suppress AHR and peribronchial fibrosis, but not airway eosinophilia, in a chronic allergen administration model of allergic disease (81) and treatment with IL-12 can suppress even established allergic airway disease (76). The suppressive effects of IL-12 depend partially on IL-12 induction of IFN-γ (76) and treatment with large doses of IFN-γ or type I IFN can also suppress at least some features of allergic airway inflammation, especially eosinophilia, in worm infection models (80). Consistent with this, IFN-γ deficiency has been reported to increase the duration of eosinophilia in allergen-immunized mice (82). Mice deficient in T-bet, the transcription factor required for Th1 responses, spontaneously develop allergic airway disease (83) that is IL-13-dependent (84). In contrast to these observations, AHR was induced to the same extent when naïve mice were inoculated with both Th1 and Th2 cells plus the appropriate Ag as when they were inoculated with only Th2 cells plus Ag (85). Even more surprisingly, adoptive transfer of eosinophils into the lungs of SCID mice has been reported to induce IFN-γ-dependent AHR (86) and transgenic IFN-γ expression in mouse lung has been reported to induce increased IL-5 and IL-13 production and airway eosinophilia (87). Although one may speculate that low levels of IFN-γ may promote allergic airway disease by increasing Ag presentation and inflammatory cell recruitment while higher levels of IFN-γ suppress Th2 responses and Th2 cytokine effects, the specific conditions that determine the net effect of IFN-γ on allergic responses in the lung and other organs remain to be determined.
The importance of IL-10 as an allergy-limiting cytokine is supported by observations that IL-10 treatment suppresses AHR, GCH and airway eosinophilia, that anti-IL-10 or anti-IL-10R mAb treatment has the opposite effect in most studies (16, 73, 88, 89), and that suppression of AHR by regulatory T cells is IL-10-dependent (90). Surprisingly, however, AHR has been difficult to induce in IL-10-deficient mice (91). This apparent discrepancy has recently been shown to reflect IL-10 suppression of IL-13Rα2, a soluble and cell membrane protein that can act as an IL-13 antagonist by binding IL-13 without inducing pro-allergic signaling (92). Consequently, mice deficient in both IL-10 and IL-13Rα2 develop more severe allergic airway disease than mice deficient in either (92). MAAD is also suppressed by another cytokine associated with regulatory T cells, TGF-β (73, 79). Chronic TGF-β treatment, however, may have the risk of enhancing development of airway smooth muscle hyperplasia and pulmonary fibrosis (93, 94).
Controversies in cytokine involvement in MAAD
The role of IL-5 in murine AAD
The importance of IL-5 and the eosinophil, which is predominantly IL-5-dependent, for induction and maintenance of the main features of MAAD, other than airway remodeling, very likely depends on the mouse strain studied, the allergen and precise immunization protocol used and possibly the local bacterial flora and animal husbandry practices. Without considering these variables, it is impossible to draw straightforward conclusions from the copious literature. One group that used a GATA1 mutation to prevent production of mature eosinophils found that these cells contribute importantly to the generation of a Th2 response in C57BL/6 mice by producing or promoting production of chemokines that recruit T cells that subsequently produce a Th2 response; however, eosinophils were not required for this purpose in similarly immunized BALB/c mice (95, 96). Consistent with this strain dependency, several reports have described an IL-5 requirement for GCH and AHR in C57BL/6, but not BALB/c mice (30, 32, 33, 44, 97, 98). Also consistent with the concept that IL-5 promotes allergic responses indirectly by promoting a Th2 response, the induction of GCH by transgenic pulmonary IL-5 overproduction has been reported to be CD4+ T cell and IL-4-dependent (99). However, intravenous IL-5 reconstituted AHR and airway eosinophilia in allergen immunized mixed background C57BL/6 – 129 mice that were Stat6-deficient, suggesting that IL-4 and IL-13 were not required (100) (Stat6 is required for IL-4/IL-13-induced AHR), and two groups have reported anti-IL-5 mAb suppression of AHR in BALB/c mice (101, 102). Endogenously produced IL-5 has also been reported to allow the development of AHR in allergen-immunized IL-4-deficient mice (103) and even to allow the development of IL-13-dependent AHR in IL-4Rα-deficient mice (through an unknown mechanism) (104). The safest conclusions to draw from these observations are that: 1) eosinophils and IL-5 contribute more to AHR and GCH in C57BL/6 mice than in BALB/c mice; and 2) very robust allergen immunization procedures and/or environmental features may induce redundant mechanisms that bypass Stat6 and IL-4Rα requirements that are seen under more typical circumstances.
The role of IL-17 in murine AAD
Because IL-17A and IL-17F potently induce CXC chemokines and neutrophil responses, among their multiple effects (105, 106), it is reasonable to expect IL-17A and IL-17F to be involved in the airway neutrophilia that characterizes many mouse models of asthma. Indeed, IL-17A and IL-17F have been reported to stimulate airway neutrophilia and have additional important effects in MAAD; however, these additional effects have varied considerably in different studies. IL-17RA-sufficient, but not IL-17RA-deficient mice (which fail to respond to IL-17A, IL-17F and IL-17E/IL-25 (107, 108)) develop MAAD when sensitized with ovalbumin and subsequently challenged i.n. with the same Ag (109). Failure of the IL-17RA-deficient mice to develop MAAD may have reflected a lack of responsiveness to IL-17E rather than IL-17A or blocking of both IL-17A and IL-17F, which have similar effects. This possibility is supported by a study in which ovalbumin immunization induced the development of airway inflammation and AHR to the same extent in IL-17A-deficient and sufficient mice that had a mixed genetic background (110). IL-17A neutralization that was restricted to the time of i.n. ovalbumin challenge exacerbated airway eosinophilia and AHR in C57BL/6 mice in one study, while IL-17A administration had the opposite effects (109). However, in a different study, i.t. administration of IL-17A immediately after challenge of ovalbumin-sensitized C57BL/6 mice with aerosolized ovalbumin exacerbated MAAD, including AHR (111). Induction of AHR by inoculation with IL-17A was not replicated by inoculation of IL-17F in this study and required both neutrophils and the presence of Th2 cytokines. In contrast, pulmonary transduction of BALB/c mice with the gene for IL-17F was reported to increase AHR and to exacerbate AHR induced by ovalbumin sensitization and challenge (112). In yet another study performed with a BALB/c - ovalbumin variant of MAAD, neutralization of IL-17 at the time of ovalbumin challenge inhibited airway neutrophilia and exacerbated airway eosinophilia but had little effect on AHR (113). In contrast, neutralization of IL-17 at the time of antigen challenge in another BALB/c - ovalbumin MAAD study suppressed airway neutrophilia, eosinophilia and AHR (114). More studies are required to determine the reasons for the differences reported in these papers and to evaluate the implications of these results for human asthma.
Our own observations, however, suggest that some of the variability in the effects of IL-17 in MAAD is mouse strain-related. We find that considerable pulmonary IL-17A is produced when MAAD is induced in A/J mice, which are particularly susceptible to allergen inoculation, while little or no pulmonary IL-17A if produced when MAAD is induced in C3H mice, which are resistant to this disease. Neutralization experiments demonstrate that IL-17A synergistically enhances IL-13-dependent AHR in A/J mice, while, not surprisingly, neutralization of IL-17A has little or no effect on MAAD in C3H mice (M. Wills-Karp, submitted). This variability may be relevant to humans, inasmuch as particularly high IL-17A concentrations are found in the lungs of people who have severe asthma with large numbers of BAL neutrophils (115, 116).
The role of IL-13Rα2 in MAAD
In addition to binding to IL-13Rα1 with low affinity and subsequently forming a high affinity association with an IL-13Rα1/IL-4Rα signaling complex, IL-13 binds with high affinity to IL-13Rα2 (117). In the mouse, IL-13Rα2 exists in two forms, a soluble form present normally in serum in low ng/ml concentrations and a cell membrane form that is expressed by smooth muscle and possibly other cell types (118, 119 and G. K. Khurana Hershey, unpublished data). These two forms are generated predominantly by alternative mRNA splicing, although proteolytic cleavage of the membrane form can generate small amounts of the soluble form (111). IL-13Rα2 has been found in several experimental systems to be a potent IL-13 antagonist; mice that lack a functional IL-13Rα2 gene generally develop more severe IL-13-mediated pathology (120-122). One group, however, has reported a signaling pathway by which an IL-13 interaction with membrane IL-13Rα2 can promote TGF-β production and pulmonary fibrosis (94) and another has reported that complexes of IL-13 and soluble IL-13Rα2 have a long in vivo half-life and promote expression of two genes; one associated with exacerbation of oxidative inflammation and the other associated with increased activation of inflammatory and antigen presenting cells (118). Neither mechanism, however, activates Stat6, which is required for IL-13-induction of GCH and AHR (123), and IL-13Rα2-deficient mice are reported to develop more severe MAAD than wild-type mice following allergen inoculation (124). Surprisingly, we find that this is not always the case; some allergen administration protocols at least initially induce disease that is more severe in wild-type than in IL-13Rα2-deficient mice (G. K. Khurana Hershey and M. Wills-Karp, unpublished data). It is not known if this reflects one of the two mechanisms noted above or, possibly, generation of an IL-13Rα2-bound IL-13 pool at the cell membrane that can then be transferred to the type II IL-4R. Additional studies are required to evaluate these possibilities and determine how they influence asthma in humans, who express only the membrane isoform of IL-13Rα2 (125, 126). Indeed, the difference in IL-13Rα2 expression between mouse and man suggests that this molecule may function differently in these two species and raises the possibility that these species may also have differences in IL-13 function.
Cells directly involved in IL-4Rα-mediated AHR
Although multiple cytokines act on multiple cell types to induce the full features of MAAD, hypotheses about how cytokines induce AHR have focused predominantly on IL-13, epithelial cells and smooth muscle cells. The focus on IL-13 is explainable by its ability to directly induce AHR and on its requirement in most studies of allergen-induced AHR (9, 10). Smooth muscle and epithelial cells have been implicated by in vitro studies demonstrating that IL-13 can act directly on smooth muscle to increase its contractility (127-136) and in vitro and in vivo demonstrations that IL-13 directly induces epithelial cell hyperplasia and GCH (124, 137, 138), which narrow airways, so that a given contraction by airway smooth muscle will induce a greater than normal increase in airway resistance (139). The importance of the IL-13 – epithelial cell axis was illustrated by demonstration that AHR develops in mice that overproduce IL-13 in their lungs and express Stat6 only in airway epithelium (124); however, AHR was also induced by IL-13 in mice that selectively lack IL-4Rα in airway epithelial cells (137). This indication that at least one additional cell type must contribute to IL-13-induced AHR recently led to studies with mice that express IL-4Rα only on smooth muscle or on all cell types other than smooth muscle (F. Finkelman, unpublished data). Studies with these mice demonstrate that direct IL-4 and IL-13 effects on smooth muscle, like airway epithelium, are sufficient but not necessary to induce AHR and support the use of the mouse as an appropriate model for studying smooth muscle contributions to asthma pathology. Additional studies are required to determine if IL-13-induced AHR can be totally accounted for by its effects on epithelial and smooth muscle cells.
Applicability of murine studies to human asthma
MAAD is an imperfect model of human asthma and allergy-related effector and regulatory immune mechanisms in mouse and man are usually similar in general but can differ in important details (140). Consequently, while cytokine studies with MAAD can be used to make predictions about cytokine roles in human asthma and to investigate mechanisms by which cytokines may promote or inhibit asthma pathogenesis, human studies must be performed to confirm or refute hypotheses generated in the mouse. When comparing the results of mouse and human studies, however, it is important to consider differences in the questions being addressed; for example, most murine cytokine neutralization studies investigate whether a specific cytokine is important in MAAD induction, while human cytokine neutralization studies always evaluate individuals who already have asthma and, thus, study importance of disease maintenance rather than induction. It is likely that some cytokines are more crucial for asthma induction than for maintaining established disease.
Cytokine roles in human asthma
Specific cytokines have been associated with human asthma through 4 types of studies: 1) comparison of levels of cytokine gene expression or protein in blood, exhaled breath condensates, or BAL or sputum cells between asthmatics and non-asthmatics; 2) comparison of the allele frequency of polymorphic cytokine genes between asthmatics and non-asthmatics or severe vs. less severe asthmatics; 3) determination of cytokine production following installation of allergens into bronchi; and 4) determination of the clinical effects of cytokine neutralization or cytokine receptor blocking on established asthma.
Comparison of cytokine expression by asthmatic and non-asthmatic individuals
Although variable results have been reported in studies that correlate cytokine production and cytokine gene expression in blood or BAL or sputum cells with asthma, most human studies associate asthma with increased gene expression and secretion of IL-4 (141-149) and IL-5 (141, 143-145, 150) and some provide similar data for IL-3 (151, 152), IL-9 (153), IL-13 (146, 152), GM-CSF (151, 152), or Stat6 (154). In contrast, as was true for mice, some human studies suggest a positive, and some a negative association between asthma and airway production of IFN-γ (147-149, 152, 155, 156). In addition, two studies suggest that IL-18 levels are lower in airway secretions of asthmatic individuals than in non-asthmatics (157, 158).
Genetic polymorphisms linked to asthma
Approximately 100 studies have been published that link increased frequency or severity of asthma to cytokine gene or cytokine signaling gene polymorphisms. More than 10 studies have linked asthma frequency or severity to polymorphisms in genes that encode IL-4 (159-168), IL-13 (162, 169, 170), TNF (159, 171-177) and IL-4Rα (18, 162, 165, 168, 178-181). Several studies have also linked asthma to genes that encode the IL-1 receptor antagonist (IL-1RA) (182-184), IL-10 (185-189), IL-18 (190-194), IFN-γ (195-197), TGF-β1 (178, 185, 186, 198-202) and Stat6 (162, 203), while polymorphisms in genes that encode IL-1β (182), IL-2 (186), IL-6 (183), IL-9 (204), IL-12/27p40 (205, 206), IL-15 (207), IL-17F (208), IL-21 (209), IL-27p28 (210), LT-α (211), TSLP (212), TGF-β2 (213) and Stat4 (196) have been linked to asthma frequency or severity by fewer studies (214). Some of these studies link asthma risk to a single nucleotide polymorphism (SNP) in a cytokine gene exon (18), suggesting that polymorphic variants influence cytokine function or longevity, while other studies link asthma risk to a SNP in a cytokine promoter (164), suggesting that polymorphic variants influence cytokine production. Several polymorphism studies have identified combinations of allelic variants of different genes that together appear to additively or synergistically influence the risk of asthma; these include combinations of allelic variants of the genes that encode TNF and IL-13 (172), IL-10 and TGF-β (185), IL-4Rα and IL-9R (215), Stat4, Stat6 and IFN-γ (196), IL-13 and IL-4Rα (169, 201) and IL-4, IL-13, IL-4Rα and Stat6 (162). Studies that fail to demonstrate linkage to asthma have also been published for cytokine or cytokine gene polymorphisms, including IL-1β (189), IL-4 (179), IL-12/IL-23p40 (216), IL-16 (160, 217), IL-17F (218), TGF-β1 (219, 220), IL-18 (221), TNF (222), IL-4Rα (222), Stat4 (223) and Stat6 (223).
Positive linkage associations do not necessarily indicate that a specific cytokine or cytokine signaling molecule is involved in asthma pathogenesis; the gene studied may instead be associated by linkage disequilibrium with a polymorphic variant of a different gene that is more directly involved. Negative studies also are not conclusive; they may have investigated the “wrong” polymorphic variant of a gene that really is involved in asthma pathogenesis, a specific racial or ethnic group in which polymorphic variants of the gene studied are not risk factors for asthma, or too few patients to identify a genuine association. Additionally, some SNP studies that link specific cytokines to asthma report linkage only for individuals with exposure to specific environmental factors, such as dust mite allergen (224, 225), freeway traffic (202) or tobacco smoke (222).
Relatively few studies demonstrate a mechanism that explains how a cytokine or cytokine-related polymorphism can influence asthma frequency or severity; exceptions include demonstration of an IL-4 promoter polymorphism that appears to increase asthma frequency by increasing IL-4 production (226), an IL-13 polymorphism that appears to exacerbate asthma by enhancing IL-13 activation of Stat6 and decreasing IL-13 affinity for IL-13Rα2 (169, 227); an IL-17F polymorphism that protects against asthma by antagonizing wild-type IL-17F activity (208); an IL-18 polymorphism that appears to increase asthma severity by increasing IL-18 expression (228) and an IL-4Rα polymorphism that increases asthma severity by promoting signaling pathways that synergize with Stat6 activation to induce genes that contribute to allergic inflammation (18).
Effects of allergen and cytokine challenge
Although bronchoscopic studies in which in vivo cytokine production by a lung segment is evaluated after allergen challenge of that lung segment have provided variable results, increased gene expression or secretion of IL-4, IL-5, IL-13 and GM-CSF have been reported by most studies (229-233). Consistent with this and with mouse studies, inhalation of recombinant human IL-4 induced AHR and sputum eosinophilic within 24 hours (234). Inhalation of recombinant human IL-5 by Chinese mild asthmatic subjects was also reported to induce AHR and airway eosinophilia in one study (235); however, these observations were not reproduced in a two studies in which British mild asthmatic subjects inhaled the same or a larger dose of recombinant human IL-5 from the same source (236, 237).
Clinical trials
Because individuals come to clinical attention only after they develop symptoms of disease, the most practical clinical question relating cytokines and asthma is whether a particular cytokine or cytokine receptor is involved in maintaining, rather than inducing asthma. Although in vivo neutralization studies with cytokine or cytokine receptor antagonists provide the most definitive way to evaluate the importance of a cytokine in maintaining established asthma, only a relatively small number of these studies have been reported and some of these have limitations that make them difficult to interpret.
Studies that administered soluble IL-4Rα as an IL-4 antagonist initially showed some promise of increasing asthma control (238), but this failed to be replicated by a later, larger study (239). Initial clinical trials of anti-IL-5 mAb in asthma patients showed considerable decreases of blood eosinophilia, a partial decrease in lung eosinophilia and no beneficial effects on lung function or symptoms (240, 241), but appeared to have a beneficial effect on remodeling by decreasing deposition of extracellular matrix proteins (242). More recent studies that have evaluated anti-IL-5 mAb effects in more defined patient populations have provided more impressive results: intravenous injection of anti-IL-5 mAb increased the amount of air expelled during the initial second of forced expiration (FEV1) and decreased asthma exacerbations, steroid requirement and blood and sputum eosinophilia in patients with steroid-resistant asthma with sputum eosinophilia in one study (243), and decreased severe exacerbations and blood and sputum eosinophilia and increased quality of life without increasing FEV1 or decreasing symptoms or AHR in a second study (244).
Treatment with either of 2 TNF antagonists was reported to induce a statistically significant beneficial effect: an increase in asthma control in one study (245), a decrease in asthma exacerbations in a second study (246) and decreased AHR with increased FEV1 and asthma-related quality of life in the third study (247). However, the beneficial effects were different for the three studies and were relatively small.
Perhaps the most impressive observations have been made in a phase 2a trial of pitrakinra, a mutated recombinant human IL-4 that blocks IL-4 and IL-13 effects by binding to IL-4Rα without signaling. Pitrakinra inhalation substantially decreased the allergen-induced decrease in FEV1 and accelerated the recovery from this effect of allergen inhalation (248). Phase 2b studies of inhaled pitrakinra in asthmatics are reported to be in progress.
Undoubtedly, the small number of reports of clinical trials of cytokine or cytokine receptor antagonists reflects the high cost of such studies and other economic considerations; however, it is also likely that additional studies have been performed, but not reported because of negative results. In this regard, it needs to be appreciated that the results of human trials, like animal studies, can be misleading. It is understandable, but unfortunate that maximum doses of cytokine or cytokine receptor antagonists in many human trials may be determined by economic rather than biologic considerations and that most trials are not set up to determine the extent to which a targeted cytokine or cytokine receptor is blocked in the lungs. Consequently, it can be impossible to determine whether negative results of a clinical trial reflect lack of importance of the targeted molecule in the maintenance of human asthma or inadequate blocking of the target. Similarly, clinical trials with cytokine antagonists do not always investigate whether the antagonist eliminates the cytokine or forms a soluble complex with the cytokine that may prolong its in vivo half-life and allow agonist effects when the complex dissociates (249, 250). In the same vein, clinical trials with cytokine receptor antagonists don’t always investigate the effect of the agent on cell membrane expression of the targeted receptor, although removal of the receptor would likely have a better inhibitory effect than simple receptor occupancy and increased expression of the receptor might be deleterious. It would seem advisable for future clinical trials to address these issues when possible.
Conclusions
Data from both murine and human studies support the importance of Th2 cytokines and, to a lesser extent, inflammatory cytokines produced by the innate immune system, in MAAD/asthma development and maintenance. These same data, however, demonstrate that murine strain differences and differences in the allergens used to induce MAAD and the protocols used to administer these allergens all influence the importance of a particular cytokine in MAAD induction and maintenance. Other environmental influences, such as animal husbandry practices and variations in “normal” bacterial flora, may also affect the role of a particular cytokine in murine AAD. Similar factors are likely to influence the importance of a particular cytokine in human asthma, as shown by differences in the association of cytokine gene polymorphisms with asthma in different ethnic and racial groups and by the different outcomes of anti-IL-5 mAb therapy in patients whose asthma had different clinical characteristics. Taken together, mouse and human studies presently suggest that therapies that effectively suppress IL-13 or IL-13 along with IL-4 and, possibly, other Th2 cytokines may offer the best cytokine-based approach for suppressing established asthma, with the caveats that no single therapy is likely to be effective for all individuals with this disease and that different cytokine-based therapies are likely to prove most effective for different groups of asthmatic individuals. We believe that identification and validation of cytokine, cytokine receptor, and cytokine-associated signaling molecules that are appropriate therapeutic targets will require additional mouse studies, particularly studies of molecules involved in maintenance of established MAAD, as well as clinical trials that evaluate mechanisms in addition to toxicity and efficacy.
Footnotes
We thank the U. S. Department for Veterans Affairs for Merit Grant support and the National Institutes of Health for research support (P01 HL076383 and R01 HL097360).
Abbreviations: AHR, airway hyperresponsiveness; FEV1, volume of air expelled during the initial second of forced expiration; GCH, goblet cell hyperplasia; i.n., intranasal; i.t., intratracheal; MAAD, murine allergic airway disease.
References
- 1.Akinbami LJ. Advance data from vital and health statistics. National Center for Health Statistics; Hyattsville, MD: 2006. The state of childhood asthma, United States, 1980-2005; pp. 1–24. [PubMed] [Google Scholar]
- 2.American Academy of Allergy, Asthma and Immunology Asthma Statistics. 2009 http://www.aaaai.org/media/statistics/asthma-statistics.asp.
- 3.Saenz SA, Taylor BC, Artis D. Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol Rev. 2008;226:172–190. doi: 10.1111/j.1600-065X.2008.00713.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Finkelman F. Cytokine regulation of type 2 immunity. In: Austen KF, Frank MM, Atkinson JP, Cantor H, editors. Samter’s Immunologic Diseases. 6th ed. Lippincott Williams & Wilkins; Philadelphia: 2001. pp. 111–126. [Google Scholar]
- 5.Steel MD, Holgate ST. Asthma. In: Austen KF, Frank MM, Atkinson JP, Cantor H, editors. Samter’s Immunologic Diseases. Sixth ed. Lippincott Williams & Wilkins; Philadelphia: 2001. pp. 836–864. [Google Scholar]
- 6.Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE. The IL-4 receptor: signaling mechanisms and biologic functions. Annu Rev Immunol. 1999;17:701–738. doi: 10.1146/annurev.immunol.17.1.701. [DOI] [PubMed] [Google Scholar]
- 7.Ramalingam TR, Pesce JT, Sheikh F, Cheever AW, Mentink-Kane MM, Wilson MS, Stevens S, Valenzuela DM, Murphy AJ, Yancopoulos GD, Urban JF, Jr., Donnelly RP, Wynn TA. Unique functions of the type II interleukin 4 receptor identified in mice lacking the interleukin 13 receptor α1 chain. Nat Immunol. 2008;9:25–33. doi: 10.1038/ni1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Munitz A, Brandt EB, Mingler M, Finkelman FD, Rothenberg ME. Distinct roles for IL-13 and IL-4 via IL-13 receptor α1 and the type II IL-4 receptor in asthma pathogenesis. Proc Natl Acad Sci U S A. 2008;105:7240–7245. doi: 10.1073/pnas.0802465105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Grunig G, Warnock M, Wakil AE, Venkayya R, Brombacher F, Rennick DM, Sheppard D, Mohrs M, Donaldson DD, Locksley RM, Corry DB. Requirement for IL-13 independently of IL-4 in experimental asthma. Science. 1998;282:2261–2263. doi: 10.1126/science.282.5397.2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wills-Karp M, Luyimbazi J, Xu X, Schofield B, Neben TY, Karp CL, Donaldson DD. Interleukin-13: central mediator of allergic asthma. Science. 1998;282:2258–2261. doi: 10.1126/science.282.5397.2258. [DOI] [PubMed] [Google Scholar]
- 11.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]
- 12.Perkins C, Wills-Karp M, Finkelman FD. IL-4 induces IL-13-independent allergic airway inflammation. J Allergy Clin Immunol. 2006;118:410–419. doi: 10.1016/j.jaci.2006.06.004. [DOI] [PubMed] [Google Scholar]
- 13.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]
- 14.Morris SC, Orekhova T, Meadows MJ, Heidorn SM, Yang J, Finkelman FD. IL-4 induces in vivo production of IFN-γ by NK and NKT cells. J Immunol. 2006;176:5299–5305. doi: 10.4049/jimmunol.176.9.5299. [DOI] [PubMed] [Google Scholar]
- 15.Huang TJ, MacAry PA, Eynott P, Moussavi A, Daniel KC, Askenase PW, Kemeny DM, Chung KF. Allergen-specific Th1 cells counteract efferent Th2 cell-dependent bronchial hyperresponsiveness and eosinophilic inflammation partly via IFN-γ. J Immunol. 2001;166:207–217. doi: 10.4049/jimmunol.166.1.207. [DOI] [PubMed] [Google Scholar]
- 16.Oh JW, Seroogy CM, Meyer EH, Akbari O, Berry G, Fathman CG, Dekruyff RH, Umetsu DT. CD4 T-helper cells engineered to produce IL-10 prevent allergen-induced airway hyperreactivity and inflammation. J Allergy Clin Immunol. 2002;110:460–468. doi: 10.1067/mai.2002.127512. [DOI] [PubMed] [Google Scholar]
- 17.LaPorte SL, Juo ZS, Vaclavikova J, Colf LA, Qi X, Heller NM, Keegan AD, Garcia KC. Molecular and structural basis of cytokine receptor pleiotropy in the interleukin-4/13 system. Cell. 2008;132:259–272. doi: 10.1016/j.cell.2007.12.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Tachdjian R, Mathias C, Al Khatib S, Bryce PJ, Kim HS, Blaeser F, O’Connor BD, Rzymkiewicz D, Chen A, Holtzman MJ, Hershey GK, Garn H, Harb H, Renz H, Oettgen HC, Chatila TA. Pathogenicity of a disease-associated human IL-4 receptor allele in experimental asthma. J Exp Med. 2009;206:2191–2204. doi: 10.1084/jem.20091480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lutz MB, Schnare M, Menges M, Rossner S, Rollinghoff M, Schuler G, Gessner A. Differential functions of IL-4 receptor types I and II for dendritic cell maturation and IL-12 production and their dependency on GM-CSF. J Immunol. 2002;169:3574–3580. doi: 10.4049/jimmunol.169.7.3574. [DOI] [PubMed] [Google Scholar]
- 20.Webb DC, Cai Y, Matthaei KI, Foster PS. Comparative roles of IL-4, IL-13, and IL-4Rα in dendritic cell maturation and CD4+ Th2 cell function. J Immunol. 2007;178:219–227. doi: 10.4049/jimmunol.178.1.219. [DOI] [PubMed] [Google Scholar]
- 21.Heller NM, Qi X, Junttila IS, Shirey KA, Vogel SN, Paul WE, Keegan AD. Type I IL-4Rs selectively activate IRS-2 to induce target gene expression in macrophages. Sci Signal. 2008;1:ra17. doi: 10.1126/scisignal.1164795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Emson CL, Bell SE, Jones A, Wisden W, McKenzie AN. Interleukin (IL)-4-independent induction of immunoglobulin (Ig)E, and perturbation of T cell development in transgenic mice expressing IL-13. J Exp Med. 1998;188:399–404. doi: 10.1084/jem.188.2.399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Finkelman FD, Katona IM, Urban JF, Jr., Holmes J, Ohara J, Tung AS, Sample JV, Paul WE. IL-4 is required to generate and sustain in vivo IgE responses. J Immunol. 1988;141:2335–2341. [PubMed] [Google Scholar]
- 24.Le Gros G, Ben-Sasson SZ, Seder R, Finkelman FD, Paul WE. Generation of interleukin 4 (IL-4)-producing cells in vivo and in vitro: IL-2 and IL-4 are required for in vitro generation of IL-4- producing cells. J Exp Med. 1990;172:921–929. doi: 10.1084/jem.172.3.921. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chomarat P, Banchereau J. Interleukin-4 and interleukin-13: their similarities and discrepancies. Int Rev Immunol. 1998;17:1–52. doi: 10.3109/08830189809084486. [DOI] [PubMed] [Google Scholar]
- 26.Finkelman FD, Morris SC, Orekhova T, Mori M, Donaldson D, Reiner SL, Reilly NL, Schopf L, Urban JF., Jr. Stat6 regulation of in vivo IL-4 responses. J Immunol. 2000;164:2303–2310. doi: 10.4049/jimmunol.164.5.2303. [DOI] [PubMed] [Google Scholar]
- 27.Zheng W, Flavell RA. The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell. 1997;89:587–596. doi: 10.1016/s0092-8674(00)80240-8. [DOI] [PubMed] [Google Scholar]
- 28.Ouyang W, Lohning M, Gao Z, Assenmacher M, Ranganath S, Radbruch A, Murphy KM. Stat6-independent GATA-3 autoactivation directs IL-4-independent Th2 development and commitment. Immunity. 2000;12:27–37. doi: 10.1016/s1074-7613(00)80156-9. [DOI] [PubMed] [Google Scholar]
- 29.Zhou B, Headley MB, Aye T, Tocker J, Comeau MR, Ziegler SF. Reversal of thymic stromal lymphopoietin-induced airway inflammation through inhibition of Th2 responses. J Immunol. 2008;181:6557–6562. doi: 10.4049/jimmunol.181.9.6557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Webb DC, McKenzie AN, Koskinen AM, Yang M, Mattes J, Foster PS. Integrated signals between IL-13, IL-4, and IL-5 regulate airways hyperreactivity. J Immunol. 2000;165:108–113. doi: 10.4049/jimmunol.165.1.108. [DOI] [PubMed] [Google Scholar]
- 31.Henderson WR, Jr., Chi EY, Maliszewski CR. Soluble IL-4 receptor inhibits airway inflammation following allergen challenge in a mouse model of asthma. J Immunol. 2000;164:1086–1095. doi: 10.4049/jimmunol.164.2.1086. [DOI] [PubMed] [Google Scholar]
- 32.Hamelmann E, Wahn U, Gelfand EW. Role of the Th2 cytokines in the development of allergen-induced airway inflammation and hyperresponsiveness. Int Arch Allergy Immunol. 1999;118:90–94. doi: 10.1159/000024037. [DOI] [PubMed] [Google Scholar]
- 33.Corry DB, Folkesson HG, Warnock ML, Erle DJ, Matthay MA, Wiener-Kronish JP, Locksley RM. Interleukin 4, but not interleukin 5 or eosinophils, is required in a murine model of acute airway hyperreactivity. J Exp Med. 1996;183:109–117. doi: 10.1084/jem.183.1.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.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]
- 35.Kips JC, Brusselle GG, Joos GF, Peleman RA, Devos RR, Tavernier JH, Pauwels RA. Importance of interleukin-4 and interleukin-12 in allergen-induced airway changes in mice. Int Arch Allergy Immunol. 1995;107:115–118. doi: 10.1159/000236947. [DOI] [PubMed] [Google Scholar]
- 36.Dittrich AM, Chen HC, Xu L, Ranney P, Connolly S, Yarovinsky TO, Bottomly HK. A new mechanism for inhalational priming: IL-4 bypasses innate immune signals. J Immunol. 2008;181:7307–7315. doi: 10.4049/jimmunol.181.10.7307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Venkayya R, Lam M, Willkom M, Grunig G, Corry DB, Erle DJ. The Th2 lymphocyte products IL-4 and IL-13 rapidly induce airway hyperresponsiveness through direct effects on resident airway cells. Am J Respir Cell Mol Biol. 2002;26:202–208. doi: 10.1165/ajrcmb.26.2.4600. [DOI] [PubMed] [Google Scholar]
- 38.Yang G, Volk A, Petley T, Emmell E, Giles-Komar J, Shang X, Li J, Das AM, Shealy D, Griswold DE, Li L. Anti-IL-13 monoclonal antibody inhibits airway hyperresponsiveness, inflammation and airway remodeling. Cytokine. 2004;28:224–232. doi: 10.1016/j.cyto.2004.08.007. [DOI] [PubMed] [Google Scholar]
- 39.Gavett SH, O’Hearn DJ, Karp CL, Patel EA, Schofield BH, Finkelman FD, Wills-Karp M. Interleukin-4 receptor blockade prevents airway responses induced by antigen challenge in mice. Am J Physiol. 1997;272:L253–261. doi: 10.1152/ajplung.1997.272.2.L253. [DOI] [PubMed] [Google Scholar]
- 40.Nishikubo K, Murata Y, Tamaki S, Sugama K, Imanaka-Yoshida K, Yuda N, Kai M, Takamura S, Sebald W, Adachi Y, Yasutomi Y. A single administration of interleukin-4 antagonistic mutant DNA inhibits allergic airway inflammation in a mouse model of asthma. Gene Ther. 2003;10:2119–2125. doi: 10.1038/sj.gt.3302131. [DOI] [PubMed] [Google Scholar]
- 41.Tomkinson A, Duez C, Cieslewicz G, Pratt JC, Joetham A, Shanafelt MC, Gundel R, Gelfand EW. A murine IL-4 receptor antagonist that inhibits IL-4- and IL-13-induced responses prevents antigen-induced airway eosinophilia and airway hyperresponsiveness. J Immunol. 2001;166:5792–5800. doi: 10.4049/jimmunol.166.9.5792. [DOI] [PubMed] [Google Scholar]
- 42.Corry DB, Grunig G, Hadeiba H, Kurup VP, Warnock ML, Sheppard D, Rennick DM, Locksley RM. Requirements for allergen-induced airway hyperreactivity in T and B cell-deficient mice. Mol Med. 1998;4:344–355. [PMC free article] [PubMed] [Google Scholar]
- 43.Hogan SP, Mould A, Kikutani H, Ramsay AJ, Foster PS. Aeroallergen-induced eosinophilic inflammation, lung damage, and airways hyperreactivity in mice can occur independently of IL-4 and allergen-specific immunoglobulins. J Clin Invest. 1997;99:1329–1339. doi: 10.1172/JCI119292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Leigh R, Ellis R, Wattie JN, Hirota JA, Matthaei KI, Foster PS, O’Byrne PM, Inman MD. Type 2 cytokines in the pathogenesis of sustained airway dysfunction and airway remodeling in mice. Am J Respir Crit Care Med. 2004;169:860–867. doi: 10.1164/rccm.200305-706OC. [DOI] [PubMed] [Google Scholar]
- 45.Karras JG, Crosby JR, Guha M, Tung D, Miller DA, Gaarde WA, Geary RS, Monia BP, Gregory SA. Anti-inflammatory activity of inhaled IL-4 receptor-α antisense oligonucleotide in mice. Am J Respir Cell Mol Biol. 2007;36:276–285. doi: 10.1165/rcmb.2005-0456OC. [DOI] [PubMed] [Google Scholar]
- 46.Rothenberg ME, Hogan SP. The eosinophil. Annu Rev Immunol. 2006;24:147–174. doi: 10.1146/annurev.immunol.24.021605.090720. [DOI] [PubMed] [Google Scholar]
- 47.Coffman RL, Seymour BW, Hudak S, Jackson J, Rennick D. Antibody to interleukin-5 inhibits helminth-induced eosinophilia in mice. Science. 1989;245:308–310. doi: 10.1126/science.2787531. [DOI] [PubMed] [Google Scholar]
- 48.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]
- 49.Tanaka H, Komai M, Nagao K, Ishizaki M, Kajiwara D, Takatsu K, Delespesse G, Nagai H. Role of IL-5 and eosinophils in allergen-induced airway remodeling in mice. Am J Respir Cell Mol Biol. 2004 doi: 10.1165/rcmb.2003-0305OC. [DOI] [PubMed] [Google Scholar]
- 50.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]
- 51.Temann UA, Ray P, Flavell RA. Pulmonary overexpression of IL-9 induces Th2 cytokine expression, leading to immune pathology. J Clin Invest. 2002;109:29–39. doi: 10.1172/JCI13696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.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]
- 53.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]
- 54.Steenwinckel V, Louahed J, Orabona C, Huaux F, Warnier G, McKenzie A, Lison D, Levitt R, Renauld JC. IL-13 mediates in vivo IL-9 activities on lung epithelial cells but not on hematopoietic cells. J Immunol. 2007;178:3244–3251. doi: 10.4049/jimmunol.178.5.3244. [DOI] [PubMed] [Google Scholar]
- 55.Jones TG, Hallgren J, Humbles A, Burwell T, Finkelman FD, Alcaide P, Austen KF, Gurish MF. Antigen-induced increases in pulmonary mast cell progenitor numbers depend on IL-9 and CD1d-restricted NKT cells. J Immunol. 2009;183:5251–5260. doi: 10.4049/jimmunol.0901471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.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]
- 57.Kondo Y, Yoshimoto T, Yasuda K, Futatsugi-Yumikura S, Morimoto M, Hayashi N, Hoshino T, Fujimoto J, Nakanishi K. Administration of IL-33 induces airway hyperresponsiveness and goblet cell hyperplasia in the lungs in the absence of adaptive immune system. Int Immunol. 2008;20:791–800. doi: 10.1093/intimm/dxn037. [DOI] [PubMed] [Google Scholar]
- 58.Wakashin H, Hirose K, Maezawa Y, Kagami S, Suto A, Watanabe N, Saito Y, Hatano M, Tokuhisa T, Iwakura Y, Puccetti P, Iwamoto I, Nakajima H. 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]
- 59.Kouzaki H, O’Grady SM, Lawrence CB, Kita H. Proteases induce production of thymic stromal lymphopoietin by airway epithelial cells through protease-activated receptor-2. J Immunol. 2009;183:1427–1434. doi: 10.4049/jimmunol.0900904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ballantyne SJ, Barlow JL, Jolin HE, Nath P, Williams AS, Chung KF, Sturton G, Wong SH, McKenzie AN. Blocking IL-25 prevents airway hyperresponsiveness in allergic asthma. J Allergy Clin Immunol. 2007;120:1324–1331. doi: 10.1016/j.jaci.2007.07.051. [DOI] [PubMed] [Google Scholar]
- 61.Liu X, Li M, Wu Y, Zhou Y, Zeng L, Huang T. Anti-IL-33 antibody treatment inhibits airway inflammation in a murine model of allergic asthma. Biochem Biophys Res Commun. 2009;386:181–185. doi: 10.1016/j.bbrc.2009.06.008. [DOI] [PubMed] [Google Scholar]
- 62.Shi L, Leu SW, Xu F, Zhou X, Yin H, Cai L, Zhang L. Local blockade of TSLP receptor alleviated allergic disease by regulating airway dendritic cells. Clin Immunol. 2008;129:202–210. doi: 10.1016/j.clim.2008.07.015. [DOI] [PubMed] [Google Scholar]
- 63.Smithgall MD, Comeau MR, Yoon BR, Kaufman D, Armitage R, Smith DE. IL-33 amplifies both Th1- and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells. Int Immunol. 2008;20:1019–1030. doi: 10.1093/intimm/dxn060. [DOI] [PubMed] [Google Scholar]
- 64.Liu YJ. Thymic stromal lymphopoietin and OX40 ligand pathway in the initiation of dendritic cell-mediated allergic inflammation. J Allergy Clin Immunol. 2007;120:238–244. doi: 10.1016/j.jaci.2007.06.004. [DOI] [PubMed] [Google Scholar]
- 65.Nakae S, Komiyama Y, Yokoyama H, Nambu A, Umeda M, Iwase M, Homma I, Sudo K, Horai R, Asano M, Iwakura Y. IL-1 is required for allergen-specific Th2 cell activation and the development of airway hypersensitivity response. Int Immunol. 2003;15:483–490. doi: 10.1093/intimm/dxg054. [DOI] [PubMed] [Google Scholar]
- 66.Ritz SA, Cundall MJ, Gajewska BU, Alvarez D, Gutierrez-Ramos JC, Coyle AJ, McKenzie AN, Stampfli MR, Jordana M. Granulocyte macrophage colony-stimulating factor-driven respiratory mucosal sensitization induces Th2 differentiation and function independently of interleukin-4. Am J Respir Cell Mol Biol. 2002;27:428–435. doi: 10.1165/rcmb.4824. [DOI] [PubMed] [Google Scholar]
- 67.Cates EC, Gajewska BU, Goncharova S, Alvarez D, Fattouh R, Coyle AJ, Gutierrez-Ramos JC, Jordana M. Effect of GM-CSF on immune, inflammatory, and clinical responses to ragweed in a novel mouse model of mucosal sensitization. J Allergy Clin Immunol. 2003;111:1076–1086. doi: 10.1067/mai.2003.1460. [DOI] [PubMed] [Google Scholar]
- 68.Ohta K, Yamashita N, Tajima M, Miyasaka T, Nakano J, Nakajima M, Ishii A, Horiuchi T, Mano K, Miyamoto T. Diesel exhaust particulate induces airway hyperresponsiveness in a murine model: essential role of GM-CSF. J Allergy Clin Immunol. 1999;104:1024–1030. doi: 10.1016/s0091-6749(99)70084-9. [DOI] [PubMed] [Google Scholar]
- 69.Su YC, Rolph MS, Hansbro NG, Mackay CR, Sewell WA. Granulocyte-macrophage colony-stimulating factor is required for bronchial eosinophilia in a murine model of allergic airway inflammation. J Immunol. 2008;180:2600–2607. doi: 10.4049/jimmunol.180.4.2600. [DOI] [PubMed] [Google Scholar]
- 70.Kodama T, Matsuyama T, Kuribayashi K, Nishioka Y, Sugita M, Akira S, Nakanishi K, Okamura H. IL-18 deficiency selectively enhances allergen-induced eosinophilia in mice. J Allergy Clin Immunol. 2000;105:45–53. doi: 10.1016/s0091-6749(00)90176-3. [DOI] [PubMed] [Google Scholar]
- 71.Yamagata S, Tomita K, Sato R, Niwa A, Higashino H, Tohda Y. Interleukin-18-deficient mice exhibit diminished chronic inflammation and airway remodelling in ovalbumin-induced asthma model. Clin Exp Immunol. 2008;154:295–304. doi: 10.1111/j.1365-2249.2008.03772.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ishikawa Y, Yoshimoto T, Nakanishi K. Contribution of IL-18-induced innate T cell activation to airway inflammation with mucus hypersecretion and airway hyperresponsiveness. Int Immunol. 2006;18:847–855. doi: 10.1093/intimm/dxl021. [DOI] [PubMed] [Google Scholar]
- 73.Fu CL, Ye YL, Lee YL, Chiang BL. Effects of overexpression of IL-10, IL-12, TGF-β and IL-4 on allergen induced change in bronchial responsiveness. Respir Res. 2006;7:72. doi: 10.1186/1465-9921-7-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.van Scott MR, Justice JP, Bradfield JF, Enright E, Sigounas A, Sur S. IL-10 reduces Th2 cytokine production and eosinophilia but augments airway reactivity in allergic mice. Am J Physiol Lung Cell Mol Physiol. 2000;278:L667–674. doi: 10.1152/ajplung.2000.278.4.L667. [DOI] [PubMed] [Google Scholar]
- 75.Hogan SP, Foster PS, Tan X, Ramsay AJ. Mucosal IL-12 gene delivery inhibits allergic airways disease and restores local antiviral immunity. Eur J Immunol. 1998;28:413–423. doi: 10.1002/(SICI)1521-4141(199802)28:02<413::AID-IMMU413>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
- 76.Gavett SH, O’Hearn DJ, Li X, Huang SK, Finkelman FD, Wills-Karp M. Interleukin 12 inhibits antigen-induced airway hyperresponsiveness, inflammation, and Th2 cytokine expression in mice. J Exp Med. 1995;182:1527–1536. doi: 10.1084/jem.182.5.1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kuribayashi K, Kodama T, Okamura H, Sugita M, Matsuyama T. Effects of post-inhalation treatment with interleukin-12 on airway hyper-reactivity, eosinophilia and interleukin-18 receptor expression in a mouse model of asthma. Clin Exp Allergy. 2002;32:641–649. doi: 10.1046/j.0954-7894.2002.01346.x. [DOI] [PubMed] [Google Scholar]
- 78.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]
- 79.Hansen G, McIntire JJ, Yeung VP, Berry G, Thorbecke GJ, Chen L, DeKruyff RH, Umetsu DT. CD4+ T helper cells engineered to produce latent TGF-β1 reverse allergen-induced airway hyperreactivity and inflammation. J Clin Invest. 2000;105:61–70. doi: 10.1172/JCI7589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Urban JF, Jr., Madden KB, Cheever AW, Trotta PP, Katona IM, Finkelman FD. IFN inhibits inflammatory responses and protective immunity in mice infected with the nematode parasite. Nippostrongylus brasiliensis. J Immunol. 1993;151:7086–7094. [PubMed] [Google Scholar]
- 81.Onari Y, Yokoyama A, Haruta Y, Nakashima T, Iwamoto H, Hattori N, Kohno N. IL-12p40 is essential for the down-regulation of airway hyperresponsiveness in a mouse model of bronchial asthma with prolonged antigen exposure. Clin Exp Allergy. 2009;39:290–298. doi: 10.1111/j.1365-2222.2008.03131.x. [DOI] [PubMed] [Google Scholar]
- 82.Coyle AJ, Tsuyuki S, Bertrand C, Huang S, Aguet M, Alkan SS, Anderson GP. Mice lacking the IFN-γ 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]
- 83.Finotto S, Neurath MF, Glickman JN, Qin S, Lehr HA, Green FH, Ackerman K, Haley K, Galle PR, Szabo SJ, Drazen JM, De Sanctis GT, Glimcher LH. 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]
- 84.Finotto S, Hausding M, Doganci A, Maxeiner JH, Lehr HA, Luft C, Galle PR, Glimcher LH. Asthmatic changes in mice lacking T-bet are mediated by IL-13. Int Immunol. 2005;17:993–1007. doi: 10.1093/intimm/dxh281. [DOI] [PubMed] [Google Scholar]
- 85.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]
- 86.Kanda A, Driss V, Hornez N, Abdallah M, Roumier T, Abboud G, Legrand F, Staumont-Salle D, Queant S, Bertout J, Fleury S, Remy P, Papin JP, Julia V, Capron M, Dombrowicz D. Eosinophil-derived IFN-γ induces airway hyperresponsiveness and lung inflammation in the absence of lymphocytes. J Allergy Clin Immunol. 2009 doi: 10.1016/j.jaci.2009.04.031. [DOI] [PubMed] [Google Scholar]
- 87.Koch M, Witzenrath M, Reuter C, Herma M, Schutte H, Suttorp N, Collins H, Kaufmann SH. Role of local pulmonary IFN-γ expression in murine allergic airway inflammation. Am J Respir Cell Mol Biol. 2006;35:211–219. doi: 10.1165/rcmb.2005-0293OC. [DOI] [PubMed] [Google Scholar]
- 88.Tournoy KG, Kips JC, Pauwels RA. Endogenous interleukin-10 suppresses allergen-induced airway inflammation and nonspecific airway responsiveness. Clin Exp Allergy. 2000;30:775–783. doi: 10.1046/j.1365-2222.2000.00838.x. [DOI] [PubMed] [Google Scholar]
- 89.Akbari O, DeKruyff RH, Umetsu DT. Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nat Immunol. 2001;2:725–731. doi: 10.1038/90667. [DOI] [PubMed] [Google Scholar]
- 90.Presser K, Schwinge D, Wegmann M, Huber S, Schmitt S, Quaas A, Maxeiner JH, Finotto S, Lohse AW, Blessing M, Schramm C. Coexpression of TGF-β1 and IL-10 enables regulatory T cells to completely suppress airway hyperreactivity. J Immunol. 2008;181:7751–7758. doi: 10.4049/jimmunol.181.11.7751. [DOI] [PubMed] [Google Scholar]
- 91.Makela MJ, Kanehiro A, Borish L, Dakhama A, Loader J, Joetham A, Xing Z, Jordana M, Larsen GL, Gelfand EW. IL-10 is necessary for the expression of airway hyperresponsiveness but not pulmonary inflammation after allergic sensitization. Proc Natl Acad Sci U S A. 2000;97:6007–6012. doi: 10.1073/pnas.100118997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wilson MS, Elnekave E, Mentink-Kane MM, Hodges MG, Pesce JT, Ramalingam TR, Thompson RW, Kamanaka M, Flavell RA, Keane-Myers A, Cheever AW, Wynn TA. IL-13Rα2 and IL-10 coordinately suppress airway inflammation, airway-hyperreactivity, and fibrosis in mice. J Clin Invest. 2007;117:2941–2951. doi: 10.1172/JCI31546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Chung KF. Cytokines in chronic obstructive pulmonary disease. Eur Respir J Suppl. 2001;34:50s–59s. [PubMed] [Google Scholar]
- 94.Fichtner-Feigl S, Strober W, Kawakami K, Puri RK, Kitani A. IL-13 signaling through the IL-13α2 receptor is involved in induction of TGF-β1 production and fibrosis. Nat Med. 2006;12:99–106. doi: 10.1038/nm1332. [DOI] [PubMed] [Google Scholar]
- 95.Walsh ER, Sahu N, Kearley J, Benjamin E, Kang BH, Humbles A, August A. Strain-specific requirement for eosinophils in the recruitment of T cells to the lung during the development of allergic asthma. J Exp Med. 2008;205:1285–1292. doi: 10.1084/jem.20071836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Humbles AA, Lloyd CM, McMillan SJ, Friend DS, Xanthou G, McKenna EE, Ghiran S, Gerard NP, Yu C, Orkin SH, Gerard C. A critical role for eosinophils in allergic airways remodeling. Science. 2004;305:1776–1779. doi: 10.1126/science.1100283. [DOI] [PubMed] [Google Scholar]
- 97.Foster PS, Hogan SP, Ramsay AJ, Matthaei KI, Young IG. Interleukin 5 deficiency abolishes eosinophilia, airways hyperreactivity, and lung damage in a mouse asthma model. J Exp Med. 1996;183:195–201. doi: 10.1084/jem.183.1.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Nagai H, Yamaguchi S, Tanaka H. The role of interleukin-5 (IL-5) in allergic airway hyperresponsiveness in mice. Ann N Y Acad Sci. 1996;796:91–96. doi: 10.1111/j.1749-6632.1996.tb32570.x. [DOI] [PubMed] [Google Scholar]
- 99.Justice JP, Crosby J, Borchers MT, Tomkinson A, Lee JJ, Lee NA. CD4+ T cell-dependent airway mucus production occurs in response to IL-5 expression in lung. Am J Physiol Lung Cell Mol Physiol. 2002;282:L1066–1074. doi: 10.1152/ajplung.00195.2001. [DOI] [PubMed] [Google Scholar]
- 100.Tomkinson A, Kanehiro A, Rabinovitch N, Joetham A, Cieslewicz G, Gelfand EW. The failure of STAT6-deficient mice to develop airway eosinophilia and airway hyperresponsiveness is overcome by interleukin-5. Am J Respir Crit Care Med. 1999;160:1283–1291. doi: 10.1164/ajrccm.160.4.9809065. [DOI] [PubMed] [Google Scholar]
- 101.Hogan SP, Koskinen A, Foster PS. Interleukin-5 and eosinophils induce airway damage and bronchial hyperreactivity during allergic airway inflammation in BALB/c mice. Immunol Cell Biol. 1997;75:284–288. doi: 10.1038/icb.1997.43. [DOI] [PubMed] [Google Scholar]
- 102.Hamelmann E, Oshiba A, Loader J, Larsen GL, Gleich G, Lee J, Gelfand EW. Antiinterleukin-5 antibody prevents airway hyperresponsiveness in a murine model of airway sensitization. Am J Respir Crit Care Med. 1997;155:819–825. doi: 10.1164/ajrccm.155.3.9117011. [DOI] [PubMed] [Google Scholar]
- 103.Hogan SP, Matthaei KI, Young JM, Koskinen A, Young IG, Foster PS. A novel T cell-regulated mechanism modulating allergen-induced airways hyperreactivity in BALB/c mice independently of IL-4 and IL-5. J Immunol. 1998;161:1501–1509. [PubMed] [Google Scholar]
- 104.Webb DC, Mahalingam S, Cai Y, Matthaei KI, Donaldson DD, Foster PS. Antigen-specific production of interleukin (IL)-13 and IL-5 cooperate to mediate IL-4Rα-independent airway hyperreactivity. Eur. J. Immunol. 2003;33:3377–3385. doi: 10.1002/eji.200324178. [DOI] [PubMed] [Google Scholar]
- 105.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]
- 106.Nembrini C, Marsland BJ, Kopf M. IL-17-producing T cells in lung immunity and inflammation. J Allergy Clin Immunol. 2009;123:986–994. doi: 10.1016/j.jaci.2009.03.033. [DOI] [PubMed] [Google Scholar]
- 107.Toy D, Kugler D, Wolfson M, Vanden Bos T, Gurgel J, Derry J, Tocker J, Peschon J. Cutting edge: interleukin 17 signals through a heteromeric receptor complex. J Immunol. 2006;177:36–39. doi: 10.4049/jimmunol.177.1.36. [DOI] [PubMed] [Google Scholar]
- 108.Rickel EA, Siegel LA, Yoon BR, Rottman JB, Kugler DG, Swart DA, Anders PM, Tocker JE, Comeau MR, Budelsky AL. Identification of functional roles for both IL-17RB and IL-17RA in mediating IL-25-induced activities. J Immunol. 2008;181:4299–4310. doi: 10.4049/jimmunol.181.6.4299. [DOI] [PubMed] [Google Scholar]
- 109.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]
- 110.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]
- 111.Wilson RH, Whitehead GS, Nakano H, Free ME, Kolls JK, Cook DN. Allergic sensitization through the airway primes Th17-dependent neutrophilia and airway hyperresponsiveness. Am J Respir Crit Care Med. 2009;180:720–730. doi: 10.1164/rccm.200904-0573OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.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]
- 113.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]
- 114.Song C, Luo L, Lei Z, Li B, Liang Z, Liu G, Li D, Zhang G, Huang B, Feng ZH. IL-17-producing alveolar macrophages mediate allergic lung inflammation related to asthma. J Immunol. 2008;181:6117–6124. doi: 10.4049/jimmunol.181.9.6117. [DOI] [PubMed] [Google Scholar]
- 115.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]
- 116.Linden A, Hoshino H, Laan M. Airway neutrophils and interleukin-1. Eur Respir J. 2000;15:973–977. doi: 10.1034/j.1399-3003.2000.15e28.x. [DOI] [PubMed] [Google Scholar]
- 117.Finkelman FD, Wynn TA, Donaldson DD, Urban JF. The role of IL-13 in helminth-induced inflammation and protective immunity against nematode infections. Curr Opin Immunol. 1999;11:420–426. doi: 10.1016/S0952-7915(99)80070-3. [DOI] [PubMed] [Google Scholar]
- 118.Khodoun M, Lewis CC, Yang JQ, Orekov T, Potter C, Wynn T, Mentink-Kane M, Hershey GK, Wills-Karp M, Finkelman FD. Differences in expression, affinity, and function of soluble (s)IL-4Rα and sIL-13Rα2 suggest opposite effects on allergic responses. J Immunol. 2007;179:6429–6438. doi: 10.4049/jimmunol.179.10.6429. [DOI] [PubMed] [Google Scholar]
- 119.Tabata Y, Chen W, Warrier MR, Gibson AM, Daines MO, Hershey GK. Allergy-driven alternative splicing of IL-13 receptor α2 yields distinct membrane and soluble forms. J Immunol. 2006;177:7905–7912. doi: 10.4049/jimmunol.177.11.7905. [DOI] [PubMed] [Google Scholar]
- 120.Kellner J, Gamarra F, Welsch U, Jorres RA, Huber RM, Bergner A. IL-13Rα2 reverses the effects of IL-13 and IL-4 on bronchial reactivity and acetylcholine-induced Ca+ signaling. Int Arch Allergy Immunol. 2007;142:199–210. doi: 10.1159/000097022. [DOI] [PubMed] [Google Scholar]
- 121.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]
- 122.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 α2 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]
- 123.Kuperman D, Schofield B, Wills-Karp M, Grusby MJ. Signal transducer and activator of transcription factor 6 (Stat6)-deficient mice are protected from antigen-induced airway hyperresponsiveness and mucus production. J Exp Med. 1998;187:939–948. doi: 10.1084/jem.187.6.939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kuperman DA, Huang X, Koth LL, Chang GH, Dolganov GM, Zhu Z, Elias JA, Sheppard D, Erle DJ. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat Med. 2002;8:885–889. doi: 10.1038/nm734. [DOI] [PubMed] [Google Scholar]
- 125.O’Toole M, Legault H, Ramsey R, Wynn TA, Kasaian MT. A novel and sensitive ELISA reveals that the soluble form of IL-13R-α2 is not expressed in plasma of healthy or asthmatic subjects. Clin Exp Allergy. 2008;38:594–601. doi: 10.1111/j.1365-2222.2007.02921.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Chen W, Sivaprasad U, Tabata Y, Gibson AM, Stier MT, Finkelman FD, Khurana Hershey GK. IL-13 receptor α2 membrane and soluble isoforms differ in human and mouse. J Immunol. 2009 doi: 10.4049/jimmunol.0901028. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.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]
- 128.Laporte JC, Moore PE, Baraldo S, Jouvin MH, Church TL, Schwartzman IN, Panettieri RA, Jr., Kinet JP, Shore SA. Direct effects of interleukin-13 on signaling pathways for physiological responses in cultured human airway smooth muscle cells. Am J Respir Crit Care Med. 2001;164:141–148. doi: 10.1164/ajrccm.164.1.2008060. [DOI] [PubMed] [Google Scholar]
- 129.Akiho H, Blennerhassett P, Deng Y, Collins SM. Role of IL-4, IL-13, and STAT6 in inflammation-induced hypercontractility of murine smooth muscle cells. Am J Physiol Gastrointest Liver Physiol. 2002;282:G226–232. doi: 10.1152/ajpgi.2002.282.2.G226. [DOI] [PubMed] [Google Scholar]
- 130.Shore SA, Moore PE. Effects of cytokines on contractile and dilator responses of airway smooth muscle. Clin Exp Pharmacol Physiol. 2002;29:859–866. doi: 10.1046/j.1440-1681.2002.03756.x. [DOI] [PubMed] [Google Scholar]
- 131.Tliba O, Deshpande D, Chen H, Van Besien C, Kannan M, Panettieri RA, Jr., Amrani Y. IL-13 enhances agonist-evoked calcium signals and contractile responses in airway smooth muscle. Br J Pharmacol. 2003;140:1159–1162. doi: 10.1038/sj.bjp.0705558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Akiho H, Deng Y, Blennerhassett P, Kanbayashi H, Collins SM. Mechanisms underlying the maintenance of muscle hypercontractility in a model of postinfective gut dysfunction. Gastroenterology. 2005;129:131–141. doi: 10.1053/j.gastro.2005.03.049. [DOI] [PubMed] [Google Scholar]
- 133.Akiho H, Lovato P, Deng Y, Ceponis PJ, Blennerhassett P, Collins SM. Interleukin-4- and -13-induced hypercontractility of human intestinal muscle cells-implication for motility changes in Crohn’s disease. Am J Physiol Gastrointest Liver Physiol. 2005;288:G609–615. doi: 10.1152/ajpgi.00273.2004. [DOI] [PubMed] [Google Scholar]
- 134.Eum SY, Maghni K, Tolloczko B, Eidelman DH, Martin JG. IL-13 may mediate allergen-induced hyperresponsiveness independently of IL-5 or eotaxin by effects on airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 2005;288:L576–584. doi: 10.1152/ajplung.00380.2003. [DOI] [PubMed] [Google Scholar]
- 135.Farghaly HS, Blagbrough IS, Medina-Tato DA, Watson ML. Interleukin 13 increases contractility of murine tracheal smooth muscle by a phosphoinositide 3-kinase p110δ-dependent mechanism. Mol Pharmacol. 2008;73:1530–1537. doi: 10.1124/mol.108.045419. [DOI] [PubMed] [Google Scholar]
- 136.Ohta Y, Hayashi M, Kanemaru T, Abe K, Ito Y, Oike M. Dual modulation of airway smooth muscle contraction by Th2 cytokines via matrix metalloproteinase-1 production. J Immunol. 2008;180:4191–4199. doi: 10.4049/jimmunol.180.6.4191. [DOI] [PubMed] [Google Scholar]
- 137.Kuperman DA, Huang X, Nguyenvu L, Holscher C, Brombacher F, Erle DJ. IL-4 receptor signaling in Clara cells is required for allergen-induced mucus production. J Immunol. 2005;175:3746–3752. doi: 10.4049/jimmunol.175.6.3746. [DOI] [PubMed] [Google Scholar]
- 138.Lewis CC, Aronow B, Hutton J, Santeliz J, Dienger K, Herman N, Finkelman FD, Wills-Karp M. Unique and overlapping gene expression patterns driven by IL-4 and IL-13 in the mouse lung. J Allergy Clin Immunol. 2009;123:795–804 e798. doi: 10.1016/j.jaci.2009.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Bates JH, Rincon M, Irvin CG. Animal models of asthma. Am J Physiol Lung Cell Mol Physiol. 2009;297:L401–410. doi: 10.1152/ajplung.00027.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Finkelman FD, Wills-Karp M. Usefulness and optimization of mouse models of allergic airway disease. J Allergy Clin Immunol. 2008;121:603–606. doi: 10.1016/j.jaci.2008.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Brasier AR, Victor S, Boetticher G, Ju H, Lee C, Bleecker ER, Castro M, Busse WW, Calhoun WJ. Molecular phenotyping of severe asthma using pattern recognition of bronchoalveolar lavage-derived cytokines. J Allergy Clin Immunol. 2008;121:30–37 e36. doi: 10.1016/j.jaci.2007.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Bertorelli G, Bocchino V, Zhou X, Zanini A, Bernini MV, Damia R, Di Comite V, Grima P, Olivieri D. Dendritic cell number is related to IL-4 expression in the airways of atopic asthmatic subjects. Allergy. 2000;55:449–454. doi: 10.1034/j.1398-9995.2000.055005449.x. [DOI] [PubMed] [Google Scholar]
- 143.Borger P, Ten Hacken NH, Vellenga E, Kauffman HF, Postma DS. Peripheral blood T lymphocytes from asthmatic patients are primed for enhanced expression of interleukin (IL)-4 and IL-5 mRNA: associations with lung function and serum IgE. Clin Exp Allergy. 1999;29:772–779. doi: 10.1046/j.1365-2222.1999.00478.x. [DOI] [PubMed] [Google Scholar]
- 144.Krug N, Jung T, Napp U, Wagner K, Schultze-Werninghaus G, Heusser C, Rieger CH, Schauer U, Fabel H. Frequencies of T cells expressing interleukin-4 and interleukin-5 in atopic asthmatic children. Comparison with atopic asthmatic adults. Am J Respir Crit Care Med. 1998;158:754–759. doi: 10.1164/ajrccm.158.3.9507038. [DOI] [PubMed] [Google Scholar]
- 145.Humbert M, Durham SR, Ying S, Kimmitt P, Barkans J, Assoufi B, Pfister R, Menz G, Robinson DS, Kay AB, Corrigan CJ. IL-4 and IL-5 mRNA and protein in bronchial biopsies from patients with atopic and nonatopic asthma: evidence against “intrinsic” asthma being a distinct immunopathologic entity. Am J Respir Crit Care Med. 1996;154:1497–1504. doi: 10.1164/ajrccm.154.5.8912771. [DOI] [PubMed] [Google Scholar]
- 146.Kotsimbos TC, Ernst P, Hamid QA. Interleukin-13 and interleukin-4 are coexpressed in atopic asthma. Proc Assoc Am Physicians. 1996;108:368–373. [PubMed] [Google Scholar]
- 147.Pala P, Message SD, Johnston SL, Openshaw PJ. Increased aeroallergen-specific interleukin-4-producing T cells in asthmatic adults. Clin Exp Allergy. 2002;32:1739–1744. doi: 10.1046/j.1365-2222.2002.01548.x. [DOI] [PubMed] [Google Scholar]
- 148.Olivenstein R, Taha R, Minshall EM, Hamid QA. IL-4 and IL-5 mRNA expression in induced sputum of asthmatic subjects: comparison with bronchial wash. J Allergy Clin Immunol. 1999;103:238–245. doi: 10.1016/s0091-6749(99)70497-5. [DOI] [PubMed] [Google Scholar]
- 149.Akpinarli A, Guc D, Kalayci O, Yigitbas E, Ozon A. Increased interleukin-4 and decreased interferon gamma production in children with asthma: function of atopy or asthma. J Asthma. 2002;39:159–165. doi: 10.1081/jas-120002197. [DOI] [PubMed] [Google Scholar]
- 150.Humbert M, Corrigan CJ, Kimmitt P, Till SJ, Kay AB, Durham SR. Relationship between IL-4 and IL-5 mRNA expression and disease severity in atopic asthma. Am J Respir Crit Care Med. 1997;156:704–708. doi: 10.1164/ajrccm.156.3.9610033. [DOI] [PubMed] [Google Scholar]
- 151.Robinson DS, Ying S, Bentley AM, Meng Q, North J, Durham SR, Kay AB, Hamid Q. Relationships among numbers of bronchoalveolar lavage cells expressing messenger ribonucleic acid for cytokines, asthma symptoms, and airway methacholine responsiveness in atopic asthma. J Allergy Clin Immunol. 1993;92:397–403. doi: 10.1016/0091-6749(93)90118-y. [DOI] [PubMed] [Google Scholar]
- 152.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]
- 153.Devos S, Cormont F, Vrtala S, Hooghe-Peters E, Pirson F, Snick J. Allergen-induced interleukin-9 production in vitro: correlation with atopy in human adults and comparison with interleukin-5 and interleukin-13. Clin Exp Allergy. 2006;36:174–182. doi: 10.1111/j.1365-2222.2006.02422.x. [DOI] [PubMed] [Google Scholar]
- 154.Mullings RE, Wilson SJ, Puddicombe SM, Lordan JL, Bucchieri F, Djukanovic R, Howarth PH, Harper S, Holgate ST, Davies DE. Signal transducer and activator of transcription 6 (STAT-6) expression and function in asthmatic bronchial epithelium. J Allergy Clin Immunol. 2001;108:832–838. doi: 10.1067/mai.2001.119554. [DOI] [PubMed] [Google Scholar]
- 155.Shannon J, Ernst P, Yamauchi Y, Olivenstein R, Lemiere C, Foley S, Cicora L, Ludwig M, Hamid Q, Martin JG. Differences in airway cytokine profile in severe asthma compared to moderate asthma. Chest. 2008;133:420–426. doi: 10.1378/chest.07-1881. [DOI] [PubMed] [Google Scholar]
- 156.Cho SH, Stanciu LA, Holgate ST, Johnston SL. Increased interleukin-4, interleukin-5, and interferon-γ 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]
- 157.Ho LP, Davis M, Denison A, Wood FT, Greening AP. Reduced interleukin-18 levels in BAL specimens from patients with asthma compared to patients with sarcoidosis and healthy control subjects. Chest. 2002;121:1421–1426. doi: 10.1378/chest.121.5.1421. [DOI] [PubMed] [Google Scholar]
- 158.Rovina N, Dima E, Gerassimou C, Kollintza A, Gratziou C, Roussos C. IL-18 in induced sputum and airway hyperresponsiveness in mild asthmatics: effect of smoking. Respir Med. 2009;103:1919–1925. doi: 10.1016/j.rmed.2009.05.023. [DOI] [PubMed] [Google Scholar]
- 159.Kamali-Sarvestani E, Ghayomi MA, Nekoee A. Association of TNF-α-308 G/A and IL-4 -589 C/T gene promoter polymorphisms with asthma susceptibility in the south of Iran. J Investig Allergol Clin Immunol. 2007;17:361–366. [PubMed] [Google Scholar]
- 160.Hosseini-Farahabadi S, Tavakkol-Afshari J, Rafatpanah H, Farid Hosseini R, Khaje Daluei M. Association between the polymorphisms of IL-4 gene promoter (−590C>T), IL-13 coding region (R130Q) and IL-16 gene promoter (−295T>C) and allergic asthma. Iran J Allergy Asthma Immunol. 2007;6:9–14. [PubMed] [Google Scholar]
- 161.Gervaziev YV, Kaznacheev VA, Gervazieva VB. Allelic polymorphisms in the interleukin-4 promoter regions and their association with bronchial asthma among the Russian population. Int Arch Allergy Immunol. 2006;141:257–264. doi: 10.1159/000095295. [DOI] [PubMed] [Google Scholar]
- 162.Kabesch M, Schedel M, Carr D, Woitsch B, Fritzsch C, Weiland SK, von Mutius E. IL-4/IL-13 pathway genetics strongly influence serum IgE levels and childhood asthma. J Allergy Clin Immunol. 2006;117:269–274. doi: 10.1016/j.jaci.2005.10.024. [DOI] [PubMed] [Google Scholar]
- 163.Basehore MJ, Howard TD, Lange LA, Moore WC, Hawkins GA, Marshik PL, Harkins MS, Meyers DA, Bleecker ER. A comprehensive evaluation of IL4 variants in ethnically diverse populations: association of total serum IgE levels and asthma in white subjects. J Allergy Clin Immunol. 2004;114:80–87. doi: 10.1016/j.jaci.2004.05.035. [DOI] [PubMed] [Google Scholar]
- 164.Kabesch M, Tzotcheva I, Carr D, Hofler C, Weiland SK, Fritzsch C, von Mutius E, Martinez FD. A complete screening of the IL4 gene: novel polymorphisms and their association with asthma and IgE in childhood. J Allergy Clin Immunol. 2003;112:893–898. doi: 10.1016/j.jaci.2003.08.033. [DOI] [PubMed] [Google Scholar]
- 165.Beghe B, Barton S, Rorke S, Peng Q, Sayers I, Gaunt T, Keith TP, Clough JB, Holgate ST, Holloway JW. Polymorphisms in the interleukin-4 and interleukin-4 receptor α chain genes confer susceptibility to asthma and atopy in a Caucasian population. Clin Exp Allergy. 2003;33:1111–1117. doi: 10.1046/j.1365-2222.2003.01731.x. [DOI] [PubMed] [Google Scholar]
- 166.Zhou Y, Fu J, Wu J, Li C. Cloning and polymorphism analysis of IL-4 proximal promoter in asthmatic children. Chin Med J (Engl) 2002;115:1624–1627. [PubMed] [Google Scholar]
- 167.Noguchi E, Nukaga-Nishio Y, Jian Z, Yokouchi Y, Kamioka M, Yamakawa-Kobayashi K, Hamaguchi H, Matsui A, Shibasaki M, Arinami T. Haplotypes of the 5′ region of the IL-4 gene and SNPs in the intergene sequence between the IL-4 and IL-13 genes are associated with atopic asthma. Hum Immunol. 2001;62:1251–1257. doi: 10.1016/s0198-8859(01)00338-x. [DOI] [PubMed] [Google Scholar]
- 168.Sandford AJ, Chagani T, Zhu S, Weir TD, Bai TR, Spinelli JJ, Fitzgerald JM, Behbehani NA, Tan WC, Pare PD. Polymorphisms in the IL4, IL4RA, and FCERIB genes and asthma severity. J Allergy Clin Immunol. 2000;106:135–140. doi: 10.1067/mai.2000.107926. [DOI] [PubMed] [Google Scholar]
- 169.Chen W, Ericksen MB, Levin LS, Khurana Hershey GK. Functional effect of the R110Q IL13 genetic variant alone and in combination with IL4RA genetic variants. J Allergy Clin Immunol. 2004;114:553–560. doi: 10.1016/j.jaci.2004.04.044. [DOI] [PubMed] [Google Scholar]
- 170.Howard TD, Whittaker PA, Zaiman AL, Koppelman GH, Xu J, Hanley MT, Meyers DA, Postma DS, Bleecker ER. Identification and association of polymorphisms in the interleukin-13 gene with asthma and atopy in a Dutch population. Am J Respir Cell Mol Biol. 2001;25:377–384. doi: 10.1165/ajrcmb.25.3.4483. [DOI] [PubMed] [Google Scholar]
- 171.Mahdaviani SA, Rezaei N, Moradi B, Dorkhosh S, Amirzargar AA, Movahedi M. Proinflammatory cytokine gene polymorphisms among Iranian patients with asthma. J Clin Immunol. 2009;29:57–62. doi: 10.1007/s10875-008-9232-1. [DOI] [PubMed] [Google Scholar]
- 172.Kim HB, Kang MJ, Lee SY, Jin HS, Kim JH, Kim BS, Jang SO, Lee YC, Sohn MH, Kim KE, Hong SJ. Combined effect of tumour necrosis factor-α and interleukin-13 polymorphisms on bronchial hyperresponsiveness in Korean children with asthma. Clin Exp Allergy. 2008;38:774–780. doi: 10.1111/j.1365-2222.2008.02965.x. [DOI] [PubMed] [Google Scholar]
- 173.Noguchi E, Yokouchi Y, Shibasaki M, Inudou M, Nakahara S, Nogami T, Kamioka M, Yamakawa-Kobayashi K, Ichikawa K, Matsui A, Arinami T. Association between TNFA polymorphism and the development of asthma in the Japanese population. Am J Respir Crit Care Med. 2002;166:43–46. doi: 10.1164/rccm.2110052. [DOI] [PubMed] [Google Scholar]
- 174.Witte JS, Palmer LJ, O’Connor RD, Hopkins PJ, Hall JM. Relation between tumour necrosis factor polymorphism TNFα-308 and risk of asthma. Eur J Hum Genet. 2002;10:82–85. doi: 10.1038/sj.ejhg.5200746. [DOI] [PubMed] [Google Scholar]
- 175.Li Kam Wa TC, Mansur AH, Britton J, Williams G, Pavord I, Richards K, Campbell DA, Morton N, Holgate ST, Morrison JF. Association between −308 tumour necrosis factor promoter polymorphism and bronchial hyperreactivity in asthma. Clin Exp Allergy. 1999;29:1204–1208. doi: 10.1046/j.1365-2222.1999.00638.x. [DOI] [PubMed] [Google Scholar]
- 176.Chagani T, Pare PD, Zhu S, Weir TD, Bai TR, Behbehani NA, Fitzgerald JM, Sandford AJ. Prevalence of tumor necrosis factor-α and angiotensin converting enzyme polymorphisms in mild/moderate and fatal/near-fatal asthma. Am J Respir Crit Care Med. 1999;160:278–282. doi: 10.1164/ajrccm.160.1.9808032. [DOI] [PubMed] [Google Scholar]
- 177.Albuquerque RV, Hayden CM, Palmer LJ, Laing IA, Rye PJ, Gibson NA, Burton PR, Goldblatt J, Lesouef PN. Association of polymorphisms within the tumour necrosis factor (TNF) genes and childhood asthma. Clin Exp Allergy. 1998;28:578–584. doi: 10.1046/j.1365-2222.1998.00273.x. [DOI] [PubMed] [Google Scholar]
- 178.de Faria IC, de Faria EJ, Toro AA, Ribeiro JD, Bertuzzo CS. Association of TGF-β1, CD14, IL-4, IL-4R and ADAM33 gene polymorphisms with asthma severity in children and adolescents. J Pediatr (Rio J) 2008;84:203–210. doi: 10.2223/JPED.1783. [DOI] [PubMed] [Google Scholar]
- 179.Cui T, Wang L, Wu J, Hu L, Xie J. Polymorphisms of IL-4, IL-4 Rα, and AICDA genes in adult allergic asthma. J Huazhong Univ Sci Technolog Med Sci. 2003;23:134–137. doi: 10.1007/BF02859936. [DOI] [PubMed] [Google Scholar]
- 180.Risma KA, Wang N, Andrews RP, Cunningham CM, Ericksen MB, Bernstein JA, Chakraborty R, Hershey GK. V75R576 IL-4 receptor α is associated with allergic asthma and enhanced IL-4 receptor function. J Immunol. 2002;169:1604–1610. doi: 10.4049/jimmunol.169.3.1604. [DOI] [PubMed] [Google Scholar]
- 181.Hershey GK, Friedrich MF, Esswein LA, Thomas ML, Chatila TA. The association of atopy with a gain-of-function mutation in the α subunit of the interleukin-4 receptor. N Engl J Med. 1997;337:1720–1725. doi: 10.1056/NEJM199712113372403. [DOI] [PubMed] [Google Scholar]
- 182.Zeyrek D, Demir E, Alpman A, Ozkinay F, Gulen F, Tanac R. Association of interleukin-1β and interleukin-1 receptor antagonist gene polymorphisms in Turkish children with atopic asthma. Allergy Asthma Proc. 2008;29:468–474. doi: 10.2500/aap.2008.29.3154. [DOI] [PubMed] [Google Scholar]
- 183.Settin A, Zedan M, Farag M, Ezz El Regal M, Osman E. Gene polymorphisms of IL-6(-174) G/C and IL-1Ra VNTR in asthmatic children. Indian J Pediatr. 2008;75:1019–1023. doi: 10.1007/s12098-008-0161-z. [DOI] [PubMed] [Google Scholar]
- 184.Gohlke H, Illig T, Bahnweg M, Klopp N, Andre E, Altmuller J, Herbon N, Werner M, Knapp M, Pescollderungg L, Boner A, Malerba G, Pignatti PF, Wjst M. Association of the interleukin-1 receptor antagonist gene with asthma. Am J Respir Crit Care Med. 2004;169:1217–1223. doi: 10.1164/rccm.200302-281OC. [DOI] [PubMed] [Google Scholar]
- 185.Kim SH, Yang EM, Lee HN, Cho BY, Ye YM, Park HS. Combined effect of IL-10 and TGF-β1 promoter polymorphisms as a risk factor for aspirin-intolerant asthma and rhinosinusitis. Allergy. 2009;64:1221–1225. doi: 10.1111/j.1398-9995.2009.01989.x. [DOI] [PubMed] [Google Scholar]
- 186.Movahedi M, Mahdaviani SA, Rezaei N, Moradi B, Dorkhosh S, Amirzargar AA. IL-10, TGF-β, IL-2, IL-12, and IFN-γ cytokine gene polymorphisms in asthma. J Asthma. 2008;45:790–794. doi: 10.1080/02770900802207261. [DOI] [PubMed] [Google Scholar]
- 187.Chatterjee R, Batra J, Kumar A, Mabalirajan U, Nahid S, Niphadkar PV, Ghosh B. Interleukin-10 promoter polymorphisms and atopic asthma in North Indians. Clin Exp Allergy. 2005;35:914–919. doi: 10.1111/j.1365-2222.2005.02273.x. [DOI] [PubMed] [Google Scholar]
- 188.Lyon H, Lange C, Lake S, Silverman EK, Randolph AG, Kwiatkowski D, Raby BA, Lazarus R, Weiland KM, Laird N, Weiss ST. IL10 gene polymorphisms are associated with asthma phenotypes in children. Genet Epidemiol. 2004;26:155–165. doi: 10.1002/gepi.10298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hang LW, Hsia TC, Chen WC, Chen HY, Tsai JJ, Tsai FJ. Interleukin-10 gene -627 allele variants, not interleukin-1β gene and receptor antagonist gene polymorphisms, are associated with atopic bronchial asthma. J Clin Lab Anal. 2003;17:168–173. doi: 10.1002/jcla.10088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Harada M, Obara K, Hirota T, Yoshimoto T, Hitomi Y, Sakashita M, Doi S, Miyatake A, Fujita K, Enomoto T, Taniguchi M, Higashi N, Fukutomi Y, Nakanishi K, Nakamura Y, Tamari M. A functional polymorphism in IL-18 is associated with severity of bronchial asthma. Am J Respir Crit Care Med. 2009 doi: 10.1164/rccm.200905-0652OC. [DOI] [PubMed] [Google Scholar]
- 191.Lee CC, Lin WY, Wan L, Tsai Y, Tsai CH, Huang CM, Chen CP, Tsai FJ. Association of interleukin-18 gene polymorphism with asthma in Chinese patients. J Clin Lab Anal. 2008;22:39–44. doi: 10.1002/jcla.20218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Higa S, Hirano T, Mayumi M, Hiraoka M, Ohshima Y, Nambu M, Yamaguchi E, Hizawa N, Kondo N, Matsui E, Katada Y, Miyatake A, Kawase I, Tanaka T. Association between interleukin-18 gene polymorphism 105A/C and asthma. Clin Exp Allergy. 2003;33:1097–1102. doi: 10.1046/j.1365-2222.2003.01739.x. [DOI] [PubMed] [Google Scholar]
- 193.Lachheb J, Chelbi H, Ammar J, Hamzaoui K, Hamzaoui A. Promoter polymorphism of the IL-18 gene is associated with atopic asthma in Tunisian children. Int J Immunogenet. 2008;35:63–68. doi: 10.1111/j.1744-313X.2007.00738.x. [DOI] [PubMed] [Google Scholar]
- 194.Imboden M, Nicod L, Nieters A, Glaus E, Matyas G, Bircher AJ, Ackermann-Liebrich U, Berger W, Probst-Hensch NM. The common G-allele of interleukin-18 single-nucleotide polymorphism is a genetic risk factor for atopic asthma. The SAPALDIA Cohort Study. Clin Exp Allergy. 2006;36:211–218. doi: 10.1111/j.1365-2222.2006.02424.x. [DOI] [PubMed] [Google Scholar]
- 195.Kumar A, Ghosh B. A single nucleotide polymorphism (A --> G) in intron 3 of IFNγ gene is associated with asthma. Genes Immun. 2008;9:294–301. doi: 10.1038/gene.2008.17. [DOI] [PubMed] [Google Scholar]
- 196.Li Y, Wu B, Xiong H, Zhu C, Zhang L. Polymorphisms of STAT-6, STAT-4 and IFN-γ genes and the risk of asthma in Chinese population. Respir Med. 2007;101:1977–1981. doi: 10.1016/j.rmed.2007.04.006. [DOI] [PubMed] [Google Scholar]
- 197.Wang TN, Chu YT, Chen WY, Feng WW, Shih NH, Hsiang CH, Ko YC. Association of interferon-γ and interferon regulatory factor 1 polymorphisms with asthma in a family-based association study in Taiwan. Clin Exp Allergy. 2006;36:1147–1152. doi: 10.1111/j.1365-2222.2006.02551.x. [DOI] [PubMed] [Google Scholar]
- 198.Mak JC, Leung HC, Ho SP, Law BK, Ho AS, Lam WK, Ip MS, Chan-Yeung MM. Analysis of TGF-β1 gene polymorphisms in Hong Kong Chinese patients with asthma. J Allergy Clin Immunol. 2006;117:92–96. doi: 10.1016/j.jaci.2005.08.049. [DOI] [PubMed] [Google Scholar]
- 199.Nagpal K, Sharma S, C BR, Nahid S, Niphadkar PV, Sharma SK, Ghosh B. TGFβ1 haplotypes and asthma in Indian populations. J Allergy Clin Immunol. 2005;115:527–533. doi: 10.1016/j.jaci.2004.11.048. [DOI] [PubMed] [Google Scholar]
- 200.Silverman ES, Palmer LJ, Subramaniam V, Hallock A, Mathew S, Vallone J, Faffe DS, Shikanai T, Raby BA, Weiss ST, Shore SA. Transforming growth factor-β1 promoter polymorphism C-509T is associated with asthma. Am J Respir Crit Care Med. 2004;169:214–219. doi: 10.1164/rccm.200307-973OC. [DOI] [PubMed] [Google Scholar]
- 201.Pulleyn LJ, Newton R, Adcock IM, Barnes PJ. TGFβ1 allele association with asthma severity. Hum Genet. 2001;109:623–627. doi: 10.1007/s00439-001-0617-y. [DOI] [PubMed] [Google Scholar]
- 202.Liebhart J, Polak M, Dabrowski A, Dobek R, Liebhart E, Dor-Wojnarowska A, Barg W, Kulczak A, Medrala W, Gladysz U, Lange A. The G/G genotype of transforming growth factor beta 1 (TGF-β1) single nucleotide (+915G/C) polymorphism coincident with other host and environmental factors is associated with irreversible bronchoconstriction in asthmatics. Int J Immunogenet. 2008;35:417–422. doi: 10.1111/j.1744-313X.2008.00771.x. [DOI] [PubMed] [Google Scholar]
- 203.Tamura K, Arakawa H, Suzuki M, Kobayashi Y, Mochizuki H, Kato M, Tokuyama K, Morikawa A. Novel dinucleotide repeat polymorphism in the first exon of the STAT-6 gene is associated with allergic diseases. Clin Exp Allergy. 2001;31:1509–1514. doi: 10.1046/j.1365-2222.2001.01191.x. [DOI] [PubMed] [Google Scholar]
- 204.Aschard H, Bouzigon E, Corda E, Ulgen A, Dizier MH, Gormand F, Lathrop M, Kauffmann F, Demenais F. Sex-specific effect of IL9 polymorphisms on lung function and polysensitization. Genes Immun. 2009;10:559–565. doi: 10.1038/gene.2009.46. [DOI] [PubMed] [Google Scholar]
- 205.Hirota T, Suzuki Y, Hasegawa K, Obara K, Matsuda A, Akahoshi M, Nakashima K, Cheng L, Takahashi N, Shimizu M, Doi S, Fujita K, Enomoto T, Ebisawa M, Yoshihara S, Nakamura Y, Kishi F, Shirakawa T, Tamari M. Functional haplotypes of IL-12B are associated with childhood atopic asthma. J Allergy Clin Immunol. 2005;116:789–795. doi: 10.1016/j.jaci.2005.06.010. [DOI] [PubMed] [Google Scholar]
- 206.Randolph AG, Lange C, Silverman EK, Lazarus R, Silverman ES, Raby B, Brown A, Ozonoff A, Richter B, Weiss ST. The IL12B gene is associated with asthma. Am J Hum Genet. 2004;75:709–715. doi: 10.1086/424886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Bierbaum S, Nickel R, Zitnik S, Ahlert I, Lau S, Deichmann KA, Wahn U, Heinzmann A. Confirmation of association of IL-15 with pediatric asthma and comparison of different controls. Allergy. 2006;61:576–580. doi: 10.1111/j.1398-9995.2006.01059.x. [DOI] [PubMed] [Google Scholar]
- 208.Kawaguchi M, Takahashi D, Hizawa N, Suzuki S, Matsukura S, Kokubu F, Maeda Y, Fukui Y, Konno S, Huang SK, Nishimura M, Adachi M. IL-17F sequence variant (His161Arg) is associated with protection against asthma and antagonizes wild-type IL-17F activity. J Allergy Clin Immunol. 2006;117:795–801. doi: 10.1016/j.jaci.2005.12.1346. [DOI] [PubMed] [Google Scholar]
- 209.Chatterjee R, Batra J, Ghosh B. A common exonic variant of interleukin21 confers susceptibility to atopic asthma. Int Arch Allergy Immunol. 2009;148:137–146. doi: 10.1159/000155744. [DOI] [PubMed] [Google Scholar]
- 210.Chae SC, Li CS, Kim KM, Yang JY, Zhang Q, Lee YC, Yang YS, Chung HT. Identification of polymorphisms in human interleukin-27 and their association with asthma in a Korean population. J Hum Genet. 2007;52:355–361. doi: 10.1007/s10038-007-0123-8. [DOI] [PubMed] [Google Scholar]
- 211.Huang SC, Wu WJ, Sun HL, Lue KH, Hsu CH, Liao PF, Ku MS. Association of a lymphotoxin-α gene polymorphism and atopic asthma in Taiwanese children. Pediatr Neonatol. 2008;49:30–34. doi: 10.1016/S1875-9572(08)60008-X. [DOI] [PubMed] [Google Scholar]
- 212.He JQ, Hallstrand TS, Knight D, Chan-Yeung M, Sandford A, Tripp B, Zamar D, Bosse Y, Kozyrskyj AL, James A, Laprise C, Daley D. A thymic stromal lymphopoietin gene variant is associated with asthma and airway hyperresponsiveness. J Allergy Clin Immunol. 2009;124:222–229. doi: 10.1016/j.jaci.2009.04.018. [DOI] [PubMed] [Google Scholar]
- 213.Hatsushika K, Hirota T, Harada M, Sakashita M, Kanzaki M, Takano S, Doi S, Fujita K, Enomoto T, Ebisawa M, Yoshihara S, Sagara H, Fukuda T, Masuyama K, Katoh R, Matsumoto K, Saito H, Ogawa H, Tamari M, Nakao A. Transforming growth factor-β2 polymorphisms are associated with childhood atopic asthma. Clin Exp Allergy. 2007;37:1165–1174. doi: 10.1111/j.1365-2222.2007.02768.x. [DOI] [PubMed] [Google Scholar]
- 214.Ober C, Hoffjan S. Asthma genetics 2006: the long and winding road to gene discovery. Genes Immun. 2006;7:95–100. doi: 10.1038/sj.gene.6364284. [DOI] [PubMed] [Google Scholar]
- 215.Melen E, Umerkajeff S, Nyberg F, Zucchelli M, Lindstedt A, Gullsten H, Wickman M, Pershagen G, Kere J. Interaction between variants in the interleukin-4 receptor α and interleukin-9 receptor genes in childhood wheezing: evidence from a birth cohort study. Clin Exp Allergy. 2006;36:1391–1398. doi: 10.1111/j.1365-2222.2006.02577.x. [DOI] [PubMed] [Google Scholar]
- 216.Noguchi E, Yokouchi Y, Shibasaki M, Kamioka M, Yamakawa-Kobayashi K, Matsui A, Arinami T. Identification of missense mutation in the IL12B gene: lack of association between IL12B polymorphisms and asthma and allergic rhinitis in the Japanese population. Genes Immun. 2001;2:401–403. doi: 10.1038/sj.gene.6363790. [DOI] [PubMed] [Google Scholar]
- 217.Chiang CH, Tang YC, Lin MW, Chung MY. Association between the IL-4 promoter polymorphisms and asthma or severity of hyperresponsiveness in Taiwanese. Respirology. 2007;12:42–48. doi: 10.1111/j.1440-1843.2006.00960.x. [DOI] [PubMed] [Google Scholar]
- 218.Ramsey CD, Lazarus R, Camargo CA, Jr., Weiss ST, Celedon JC. Polymorphisms in the interleukin 17F gene (IL17F) and asthma. Genes Immun. 2005;6:236–241. doi: 10.1038/sj.gene.6364170. [DOI] [PubMed] [Google Scholar]
- 219.Aytekin C, Dogu F, Ikinciogullari A, Egin Y, Yuksek M, Bozdogan G, Akar N, Babacan E. TGF-β1-915G/C and TNF-α-308G/A polymorphisms in children with asthma. Tuberk Toraks. 2009;57:62–67. [PubMed] [Google Scholar]
- 220.Heinzmann A, Bauer E, Ganter K, Kurz T, Deichmann KA. Polymorphisms of the TGF-β1 gene are not associated with bronchial asthma in Caucasian children. Pediatr Allergy Immunol. 2005;16:310–314. doi: 10.1111/j.1399-3038.2005.00287.x. [DOI] [PubMed] [Google Scholar]
- 221.Heinzmann A, Gerhold K, Ganter K, Kurz T, Schuchmann L, Keitzer R, Berner R, Deichmann KA. Association study of polymorphisms within interleukin-18 in juvenile idiopathic arthritis and bronchial asthma. Allergy. 2004;59:845–849. doi: 10.1111/j.1398-9995.2004.00538.x. [DOI] [PubMed] [Google Scholar]
- 222.Mak JC, Ko FW, Chu CM, Leung HC, Chan HW, Cheung AH, Ip MS, Chan-Yeung M. Polymorphisms in the IL-4, IL-4 receptor α chain, TNF-α, and lymphotoxin-α genes and risk of asthma in Hong Kong Chinese adults. Int Arch Allergy Immunol. 2007;144:114–122. doi: 10.1159/000103222. [DOI] [PubMed] [Google Scholar]
- 223.Pykalainen M, Kinos R, Valkonen S, Rydman P, Kilpelainen M, Laitinen LA, Karjalainen J, Nieminen M, Hurme M, Kere J, Laitinen T, Lahesmaa R. Association analysis of common variants of STAT6, GATA3, and STAT4 to asthma and high serum IgE phenotypes. J Allergy Clin Immunol. 2005;115:80–87. doi: 10.1016/j.jaci.2004.10.006. [DOI] [PubMed] [Google Scholar]
- 224.Sharma S, Raby BA, Hunninghake GM, Soto-Quiros M, Avila L, Murphy AJ, Lasky-Su J, Klanderman BJ, Sylvia JS, Weiss ST, Celedon JC. Variants in TGFB1, dust mite exposure, and disease severity in children with asthma. Am J Respir Crit Care Med. 2009;179:356–362. doi: 10.1164/rccm.200808-1268OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Hunninghake GM, Soto-Quiros ME, Lasky-Su J, Avila L, Ly NP, Liang C, Klanderman BJ, Raby BA, Gold DR, Weiss ST, Celedon JC. Dust mite exposure modifies the effect of functional IL10 polymorphisms on allergy and asthma exacerbations. J Allergy Clin Immunol. 2008;122:93–98. 98 e91–95. doi: 10.1016/j.jaci.2008.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Li Y, Guo B, Zhang L, Han J, Wu B, Xiong H. Association between C-589T polymorphisms of interleukin-4 gene promoter and asthma: a meta-analysis. Respir Med. 2008;102:984–992. doi: 10.1016/j.rmed.2008.02.008. [DOI] [PubMed] [Google Scholar]
- 227.Vladich FD, Brazille SM, Stern D, Peck ML, Ghittoni R, Vercelli D. IL-13 R130Q, a common variant associated with allergy and asthma, enhances effector mechanisms essential for human allergic inflammation. J Clin Invest. 2005;115:747–754. doi: 10.1172/JCI22818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Pawlik A, Kaminski M, Kusnierczyk P, Kurzawski M, Dziedziejko V, Adamska M, Safranow K, Gawronska-Szklarz B. Interleukin-18 promoter polymorphism in patients with atopic asthma. Tissue Antigens. 2007;70:314–318. doi: 10.1111/j.1399-0039.2007.00908.x. [DOI] [PubMed] [Google Scholar]
- 229.Virchow JC, Jr., Walker C, Hafner D, Kortsik C, Werner P, Matthys H, Kroegel C. T cells and cytokines in bronchoalveolar lavage fluid after segmental allergen provocation in atopic asthma. Am J Respir Crit Care Med. 1995;151:960–968. doi: 10.1164/ajrccm/151.4.960. [DOI] [PubMed] [Google Scholar]
- 230.Huang SK, Xiao HQ, Kleine-Tebbe J, Paciotti G, Marsh DG, Lichtenstein LM, Liu MC. IL-13 expression at the sites of allergen challenge in patients with asthma. J Immunol. 1995;155:2688–2694. [PubMed] [Google Scholar]
- 231.Kroegel C, Julius P, Matthys H, Virchow JC, Jr., Luttmann W. Endobronchial secretion of interleukin-13 following local allergen challenge in atopic asthma: relationship to interleukin-4 and eosinophil counts. Eur Respir J. 1996;9:899–904. doi: 10.1183/09031936.96.09050899. [DOI] [PubMed] [Google Scholar]
- 232.Schroeder JT, Lichtenstein LM, Roche EM, Xiao H, Liu MC. IL-4 production by human basophils found in the lung following segmental allergen challenge. J Allergy Clin Immunol. 2001;107:265–271. doi: 10.1067/mai.2001.112846. [DOI] [PubMed] [Google Scholar]
- 233.Robinson D, Hamid Q, Bentley A, Ying S, Kay AB, Durham SR. Activation of CD4+ T cells, increased TH2-type cytokine mRNA expression, and eosinophil recruitment in bronchoalveolar lavage after allergen inhalation challenge in patients with atopic asthma. J Allergy Clin Immunol. 1993;92:313–324. doi: 10.1016/0091-6749(93)90175-f. [DOI] [PubMed] [Google Scholar]
- 234.Shi HZ, Deng JM, Xu H, Nong ZX, Xiao CQ, Liu ZM, Qin SM, Jiang HX, Liu GN, Chen YQ. Effect of inhaled interleukin-4 on airway hyperreactivity in asthmatics. Am J Respir Crit Care Med. 1998;157:1818–1821. doi: 10.1164/ajrccm.157.6.9710023. [DOI] [PubMed] [Google Scholar]
- 235.Shi HZ, Xiao CQ, Zhong D, Qin SM, Liu Y, Liang GR, Xu H, Chen YQ, Long XM, Xie ZF. Effect of inhaled interleukin-5 on airway hyperreactivity and eosinophilia in asthmatics. Am J Respir Crit Care Med. 1998;157:204–209. doi: 10.1164/ajrccm.157.1.9703027. [DOI] [PubMed] [Google Scholar]
- 236.van Rensen EL, Stirling RG, Scheerens J, Staples K, Sterk PJ, Barnes PJ, Chung KF. Evidence for systemic rather than pulmonary effects of interleukin-5 administration in asthma. Thorax. 2001;56:935–940. doi: 10.1136/thorax.56.12.935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Menzies-Gow AN, Flood-Page PT, Robinson DS, Kay AB. Effect of inhaled interleukin-5 on eosinophil progenitors in the bronchi and bone marrow of asthmatic and non-asthmatic volunteers. Clin Exp Allergy. 2007;37:1023–1032. doi: 10.1111/j.1365-2222.2007.02735.x. [DOI] [PubMed] [Google Scholar]
- 238.Borish LC, Nelson HS, Corren J, Bensch G, Busse WW, Whitmore JB, Agosti JM. Efficacy of soluble IL-4 receptor for the treatment of adults with asthma. J Allergy Clin Immunol. 2001;107:963–970. doi: 10.1067/mai.2001.115624. [DOI] [PubMed] [Google Scholar]
- 239.Riffo-Vasquez Y, Spina D. Role of cytokines and chemokines in bronchial hyperresponsiveness and airway inflammation. Pharmacol Ther. 2002;94:185–211. doi: 10.1016/s0163-7258(02)00217-6. [DOI] [PubMed] [Google Scholar]
- 240.Leckie MJ, ten Brinke A, Khan J, Diamant Z, O’Connor BJ, Walls CM, Mathur AK, Cowley HC, Chung KF, Djukanovic R, Hansel TT, Holgate ST, Sterk PJ, Barnes PJ. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356:2144–2148. doi: 10.1016/s0140-6736(00)03496-6. [DOI] [PubMed] [Google Scholar]
- 241.Flood-Page PT, Menzies-Gow AN, Kay AB, Robinson DS. Eosinophil’s role remains uncertain as anti-interleukin-5 only partially depletes numbers in asthmatic airway. Am J Respir Crit Care Med. 2003;167:199–204. doi: 10.1164/rccm.200208-789OC. [DOI] [PubMed] [Google Scholar]
- 242.Flood-Page P, Menzies-Gow A, Phipps S, Ying S, Wangoo A, Ludwig MS, Barnes N, Robinson D, Kay AB. Anti-IL-5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003;112:1029–1036. doi: 10.1172/JCI17974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Nair P, Pizzichini MM, Kjarsgaard M, Inman MD, Efthimiadis A, Pizzichini E, Hargreave FE, O’Byrne PM. Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N Engl J Med. 2009;360:985–993. doi: 10.1056/NEJMoa0805435. [DOI] [PubMed] [Google Scholar]
- 244.Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, Marshall RP, Bradding P, Green RH, Wardlaw AJ, Pavord ID. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360:973–984. doi: 10.1056/NEJMoa0808991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Morjaria JB, Chauhan AJ, Babu KS, Polosa R, Davies DE, Holgate ST. The role of a soluble TNFα receptor fusion protein (etanercept) in corticosteroid refractory asthma: a double blind, randomised, placebo controlled trial. Thorax. 2008;63:584–591. doi: 10.1136/thx.2007.086314. [DOI] [PubMed] [Google Scholar]
- 246.Erin EM, Leaker BR, Nicholson GC, Tan AJ, Green LM, Neighbour H, Zacharasiewicz AS, Turner J, Barnathan ES, Kon OM, Barnes PJ, Hansel TT. The effects of a monoclonal antibody directed against tumor necrosis factor-α in asthma. Am J Respir Crit Care Med. 2006;174:753–762. doi: 10.1164/rccm.200601-072OC. [DOI] [PubMed] [Google Scholar]
- 247.Berry MA, Hargadon B, Shelley M, Parker D, Shaw DE, Green RH, Bradding P, Brightling CE, Wardlaw AJ, Pavord ID. Evidence of a role of tumor necrosis factor α in refractory asthma. N Engl J Med. 2006;354:697–708. doi: 10.1056/NEJMoa050580. [DOI] [PubMed] [Google Scholar]
- 248.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]
- 249.Finkelman FD, Madden KB, Morris SC, Holmes JM, Boiani N, Katona IM, Maliszewski CR. Anti-cytokine antibodies as carrier proteins. Prolongation of in vivo effects of exogenous cytokines by injection of cytokine-anti-cytokine antibody complexes. J Immunol. 1993;151:1235–1244. [PubMed] [Google Scholar]
- 250.Phelan JD, Orekov T, Finkelman FD. Cutting edge: mechanism of enhancement of in vivo cytokine effects by anti-cytokine monoclonal antibodies. J Immunol. 2008;180:44–48. doi: 10.4049/jimmunol.180.1.44. [DOI] [PubMed] [Google Scholar]