Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Jun 1.
Published in final edited form as: J Allergy Clin Immunol. 2013 Mar 16;131(6):1504–1512. doi: 10.1016/j.jaci.2013.01.035

Prostaglandin D2 pathway upregulation: Relation to asthma severity, control, and TH2 inflammation

Merritt L Fajt a, Stacy L Gelhaus b, Bruce Freeman b, Crystal E Uvalle a, John B Trudeau a, Fernando Holguin a, Sally E Wenzel a
PMCID: PMC3889167  NIHMSID: NIHMS459466  PMID: 23506843

Abstract

Background

Bronchoalveolar lavage (BAL) fluid prostaglandin D2 (PGD2) levels are increased in patients with severe, poorly controlled asthma in association with epithelial mast cells (MCs). PGD2, which is generated by hematopoietic prostaglandin D synthase (HPGDS), acts on 3 G protein–coupled receptors, including chemoattractant receptor–homologous molecule expressed on TH2 lymphocytes (CRTH2) and PGD2 receptor 1 (DP1). However, much remains to be understood regarding the presence and activation of these pathway elements in asthmatic patients.

Objective

We sought to compare the expression and activation of PGD2 pathway elements in bronchoscopically obtained samples from healthy control subjects and asthmatic patients across a range of disease severity and control, as well as in relation to TH2 pathway elements.

Methods

Epithelial cells and BAL fluid were evaluated for HPGDS (quantitative real-time PCR/immunohistochemistry [IHC]) and PGD2 (ELISA/liquid chromatography mass spectrometry) in relation to levels of MC proteases. Expression of the 2 inflammatory cell receptors DP1 and CRTH2 was evaluated on luminal cells. These PGD2 pathway markers were then compared with asthma severity, level of control, and markers of TH2 inflammation (blood eosinophils and fraction of exhaled nitric oxide).

Results

Confirming previous results, BAL fluid PGD2 levels were highest in patients with severe asthma (overall P = .0001). Epithelial cell compartment HPGDS mRNA and IHC values differed among groups (P = .008 and P < .0001, respectively) and correlated with MC protease mRNA. CRTH2 mRNA and IHC values were highest in patients with severe asthma (P = .001 and P = .0001, respectively). Asthma exacerbations, poor asthma control, and TH2 inflammatory markers were associated with higher PGD2, HPGDS, and CRTH2 levels.

Conclusion

The current study identifies coordinated upregulation of the PGD2 pathway in patients with severe, poorly controlled, TH2-high asthma despite corticosteroid use.

Keywords: Asthma control, chemoattractant receptor–homologous molecule expressed on TH2 lymphocytes, prostaglandin D2, severe asthma, TH2 inflammation


Although severe asthma remains poorly understood,1-3 many studies support mast cell (MC) involvement.4-6 Activated MCs generate lipid mediators, particularly cysteinyl leukotrienes and prostaglandin D2 (PGD2),7 with increased PGD2 levels recently reported in bronchoalveolar lavage (BAL) fluid from patients with severe asthma (SA), particularly in relation to frequent/severe exacerbations.6 PGD2 is generated by prostaglandin D synthases after conversion of arachidonic acid to PGG2 and PGH2 by COXs.8 The lipocalin type is expressed in brain, cardiac, and adipose tissue, whereas hematopoietic prostaglandin D synthase (HPGDS) is mainly expressed in MCs, macrophages, dendritic cells, and TH2 cells.8-10 Studies in patients with nasal polyps, rhinosinusitis, and eosinophilic esophagitis suggest that MCs are the predominant source of HPGDS.11-13

PGD2 acts through the 3 G protein–coupled receptors thromboxane receptor (TP), PGD2 receptor 1 (DP1), and chemoattractant receptor–homologous molecule expressed on TH2 lymphocytes (CRTH2/DP2), only 2 of which are found on inflammatory cells.9,10 Binding PGD2 (and thromboxane) with high affinity, TP promotes smooth muscle constriction and platelet aggregation and likely contributes to allergen-induced bronchoconstriction.9,10 In contrast, DP1 activation causes vasodilation and bronchodilation in smooth muscle, inhibits platelet aggregation, and can promote polarization and recruitment of TH2 lymphocytes through inhibition of TH1 cytokines.14,15 CRTH2 activation on TH2 lymphocytes, eosinophils, and basophils enhances chemotaxis and activation and potentially contributes to the development and maintenance of a TH2 immune process.9,10 Additionally, CRTH2 activation of TH2 cells induces cytokine production, which could promote IgE pathway activation on MCs, further enhancing PGD2 generation.

On the basis of our previous findings of increased epithelial MC numbers and PGD2 levels in SAs6 and recent positive CRTH2 antagonist studies in asthmatic patients,16-18 we hypothesized that increased PGD2 levels would be accompanied by expression of its synthesizing enzyme, HPGDS, as well as upregulation of at least 1 of its receptors, CRTH2, on eosinophils and CD3 lymphocytes. This coordinated pathway upregulation would associate with poor asthma control, increased exacerbations, and a TH2 immune process. To address this, epithelial and BAL cells and fluid from asthmatic patients and healthy control subjects (HCs) were evaluated for PGD2 and HPGDS in relation to MC proteases, as well as DP1 and CRTH2 on luminal inflammatory cells. These PGD2 pathway markers were then compared with asthma severity, level of control, and TH2 inflammatory markers. Some results have previously been reported in abstracts.19-21

RESULTS

Demographics

Bronchoscopic samples were obtained from 112 subjects, and groups did not differ by race or sex (Table I). SAs were older (overall P < .0001, all intergroup P < .0002) and had a higher body mass index compared with HCs (intergroup P < .0001). Less atopy and lower serum IgE levels and blood eosinophil numbers were found in HCs compared with asthmatic patients. SAs had the lowest FEV1 percent predicted values, which were lower than in all other groups (overall P < .0001, all intergroup P < .0001). Leukotriene modifier use was more common in SAs compared with that seen in patients with milder asthma. Long-acting β-agonist use did not differ between SAs and the Mild-Mod/ICS group. Forty (87%) SAs were using systemic corticosteroids.

TABLE I.

Baseline demographic characteristics (n = 112)

Subject group
HCs (n = 33) Mild/no ICS
group (n = 11)
Mild-Mod/ICS
group (n = 22)
Severe asthma (n = 46) Overall difference
(P value)
Age (y)* 27 (24-39) 26 (20-31) 30 (23-39) 47 (37-54) <.0001
Sex, male/female 16/17 2/9 5/17 14/32 .12
Race, AA/white/other 23/3/7 6/3/2 14/5/3 37/6/3 .29
BMI (kg/m2)* 24 (22-29) 27 (24-31) 26 (24-28) 31 (26-35) .0007
Atopy (%) 58 100 91 76 .007§
Serum IgE (kU/L)* 28 (14-65) 98 (55-252) 109 (42-405) 166 (44-501) <.0001§
Blood eosinophils (/mL)* 100 (100-200) 200 (100-300) 200 (100-300) 300 (100-400) .012
Baseline FEV1 (% predicted)* 96 (93-105) 89 (86-97) 90 (71-105) 56 (41-71) <.0001
Exhaled NO (ppb)* 20 (13-34) 37 (25-66) 25 (17-35) 39 (23-81) .0003
Hx exacerbation (%) NA 9 27 96 <.0001
Use of LTM (no.) NA 0 4 32 <.0001
Use of LABAs (no.) NA NA 15 38 NA

If not indicated otherwise, analyses were done with Pearson χ2 tests.

AA, African American; BMI, body mass index; LABA, long-acting β-agonist; LTM, leukotriene modifier; NA, not applicable.

*

Data were analyzed by using Kruskal-Wallis tests.

Significant intergroup comparisons were noted as follows: HCs versus SAs

HCs versus Mild-Mod/ICS group

§

HCs versus Mild/no ICS group

Mild/no ICS group versus SAs

Mild-Mod/ICS group versus SAs.

All 112 subjects had BAL fluid PGD2 measurements. Because of the availability or quality of the samples, 89 (80%) had epithelial cell brushing and 98 (87%) had BAL cell mRNA data (see Table E1 in this article’s Online Repository at www.jacionline.org). Immunohistochemistry (IHC) data were available in a subset (n = 52 [epithelial] and n = 47 [BAL cell]). The subgroup with additional IHC data did not differ from those with mRNA and PGD2 data in any basic demographic characteristic, lung function, or medication use (see Table E2 in this article’s Online Repository at www.jacionline.org). In total, 88 (79%) had data from each of the 3 sample types (BAL fluid, BAL cells, and epithelial brushings; see Fig E1 and Table E3 in this article’s Online Repository at www.jacionline.org for subanalysis based on the completeness of the parameters).

Severe asthma is associated with evidence of PGD2 pathway activation

We previously reported increased PGD2 levels in BAL fluid of SAs compared with that of HCs and patients with milder asthma, 33 of whom are included in the current analysis.6 The addition of 79 new subjects expanded and confirmed these prior findings because BAL fluid PGD2 concentrations differed among the groups (overall P = .0001) and differentiated SAs from HCs and the Mild-Mod/ICS group (Fig 1). Analysis of the 79 nonoverlapping subjects confirmed the findings of the larger cohort and validated the findings from the previous report by Balzar et al6 (overall P = .0016, see Fig E2 in this article’s Online Repository at www.jacionline.org). Because BAL fluid PGD2 levels were generally low, LCMS confirmed PGD2 levels in 10 subjects and correlated with ELISA-determined levels (rs = 0.80, P = .006, see Fig E3 in this article’s Online Repository at www.jacionline.org).

FIG 1.

FIG 1

PGD2 levels measured by using ELISA in BAL fluid samples.

The PGD2 synthesizing enzyme HPGDS is increased in the asthmatic epithelium

mRNA

Epithelial cell brushing HPGDS mRNA levels differed among the groups (overall P = .008), were higher in SAs compared with HCs, and tended to be higher in the Mild-Mod/ICS group compared with those seen in HCs (Fig 2, A). In all subjects epithelial cell brushing HPGDS and MC protease mRNA levels correlated (rs = 0.75, P < .001 [tryptase]; rs = 0.67, P < .001 [CPA3]; Fig 2, B; see Table E4 in this article’s Online Repository at www.jacionline.org for MC mRNA by subject groups). In SAs (n = 35) the correlation of HPGDS with MC proteases was even stronger (rs = 0.88, P < .0001 [tryptase]; rs = 0.86, P < .0001 [CPA3]), suggesting a common epithelial MC source, whereas the correlation in HCs (n = 26) was less strong (rs = 0.48, P = .01 [tryptase]; rs = 0.32, P = .11 [CPA3]). HPGDS mRNA levels in the epithelium did not correlate with BAL fluid PGD2 levels, even when evaluated based on severity.

FIG 2.

FIG 2

A, qRT-PCR of HPGDS mRNA transcripts in epithelial cell brushings relative to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). *The difference between HCs and the Mild-Mod/ICS group tended to be different (P = .01) but did not reach significance. B, Correlation between HPGDS and tryptase epithelial cell brushing mRNA levels.

IHC/protein

Epithelial brushing HPGDS staining differed across groups (total n = 52, overall P < .0001). The percentage of HPGDS+ cells was highest in SAs and differentiated SAs from both the milder asthma group and the HCs (all intergroup P ≤ .0004; Fig 3, A). Staining in SAs suggested that positive cells included MCs (Fig 3, B). Epithelial cells themselves did not stain for HPGDS. HPGDS+ cell numbers and mRNA levels modestly correlated (n = 48, rs = 0.38, P = .008). Similar to HPGDS mRNA levels, HPGDS+ cell numbers correlated with MC protease mRNA levels (rs = 0.40, P = .004 [tryptase]; rs = 0.45, P = .001 [CPA3]) and also correlated with BAL fluid PGD2 levels (rs = 0.44, P = .001). Adjustment for severity did not substantially improve these relationships.

FIG 3.

FIG 3

Epithelial brushing cell cytospin preparations. A, Comparison of the percentage of total HPGDS+ cells, as determined by means of IHC. B, Cytospin preparations (fixed in paraformaldehyde) stained with HPGDS antibody. Red arrows, HPGDS+ cells from an SA. Panels are shown at ×40 magnification.

Severe asthma is associated with increased CRTH2 receptor expression

mRNA (Fig 4)

FIG 4.

FIG 4

qRT-PCR of DP1 and CRTH2 mRNA transcripts in BAL cells relative to glyceraldehyde-3-phosphate dehydrogenase (GAPDH). *The difference between the Mild-Mod/ICS group versus SAs tended to be different (P = .01) but did not reach significance.

Although DP1 mRNA levels did not differ among groups (overall P = .88), CRTH2 mRNA levels did differ (overall P = .001), being highest in SAs, significantly higher in SAs than in the Mild/no ICS group and HCs (each intergroup P ≤ .005), and tending to be higher in SAs than in the Mild-Mod/ICS group (intergroup P = .01). Despite the lack of difference in DP1 expression, DP1 and CRTH2 mRNA levels correlated (rs = 0.62, P < .0001 for all subjects).

IHC/protein

The percentage of DP1+ cells did not differ among subject groups (overall P = .27; Fig 5, A and B; see Table E4). Double immunostaining of BAL cells identified CRTH2+ cells (single positive, likely eosinophils), CD3+ lymphocytes, and CRTH2+CD3+ lymphocytes (double positive; Fig 5, D). The total percentage of CRTH2+ inflammatory cells (sum of CRTH2+ [single-positive] and CRTH2+CD3+ [double-positive] cells per total cells counted) was increased in SAs (overall P = .0001; Fig 5, C). Total CRTH2+ cell percentages differentiated HCs from all asthmatic groups and SAs from the Mild-Mod/ICS group (each intergroup P ≤ .007). The percentages of single- and double-positive CRTH2 cells were also higher in SAs (see Table E5 in this article’s Online Repository at www.jacionline.org). The percentage of CRTH2+ cells correlated with PGD2 levels (rs = 0.46, P = .001). There was no correlation between CRTH2+ cell numbers and mRNA levels.

FIG 5.

FIG 5

BAL cell cytospin preparations. A and C, Comparison of the percentage of total BAL cells positive for DP1 (Fig 5, A) and CRTH2 (Fig 5, C), as determined by means of IHC, is shown. B, Cytospin preparations stained with DP1 antibody. Red arrows, DP1+ cells. Upper left panel, HC; lower left panel, Mild-Mod/ICS subject; right panel, SA. Panels are shown at ×40 magnification. D, Cytospin preparations from an SA double stained with CRTH2 and CD3 antibodies. The upper panel is shown at ×40 magnification. The lower panel is shown at ×100 magnification. Red arrow, CRTH2+/CD3+ lymphocyte; blue arrow, CRTH2+ eosinophil; black arrow, CD3+ lymphocyte.

Clinical implications of PGD2 pathway markers in asthmatic patients

Asthma exacerbations

In the previous 12 months, 96% of SAs, 27% of the Mild-Mod/ICS group, and 9% of the Mild/no ICS group experienced an asthma exacerbation (overall P < .0001, Table I). A historical exacerbation was associated with higher BAL fluid PGD2 levels, HPGDS+ cell numbers, and CRTH2+ cell numbers and mRNA levels compared with those seen in asthmatic patients without such a history (all P < .05). Epithelial HPGDS mRNA and BAL cell DP1 levels did not differ between asthmatic patients with and without exacerbations (Table II).

TABLE II.

History of recent exacerbation and PGD2 pathway markers in all asthmatic patients

Recent asthma exacerbation:
No
Yes
p
PGD2 pathway marker No. Median (IQR) No. Median (IQR) value
BAL fluid PGD2 (pg/mL) 28 6.0 (4.6-8.8) 51 8.5 (6.0-19) .001
Epithelial HPGDS mRNA 24 5.0 (1.2-8.7) 39 3.3 (1.8-9.5) .99
HPGDS+ cells in
 epithelium (%)
21 1.0 (0.6-2.3) 19 4.0 (2.7-6.0) .0002
BAL cell CRTH2 mRNA 26 0.1 (0.04-0.2) 43 0.2 (0.1-1.7) .015
CRTH2+ BAL cells (%) 19 3.9 (2.1-6.3) 16 5.9 (4.4-9.0) .048
BAL cell DP1 mRNA 26 2.1 (1.1-6.3) 43 1.4 (0.5-9.1) .60
DP1+ BAL cells (%) 19 4.4 (3.3-5.9) 16 4.0 (3.0-4.5) .21

IQR, Interquartile range.

Poor asthma control

Eleven asthmatic patients met the NAEPP criteria2 for well-controlled, 25 for not well-controlled, and 43 for very poorly controlled asthma. Although the majority of patients with very poorly controlled asthma had severe asthma, 6 patients with milder asthma were also in this category. Each control group had asthmatic patients from each severity group (see Table E6 in this article’s Online Repository at www.jacionline.org). The majority of patients with very poorly controlled asthma met several criteria for poor control (70% met ≥2 criteria). BAL fluid PGD2 levels, HPGDS+ cell numbers, and CRTH2+ cell numbers and mRNA levels varied by level of control, with higher values in patients with very poorly controlled asthma (all P < .04, Table III). PGD2 and CRTH2 mRNA levels differentiated patients with well-controlled from those with very poorly controlled asthma (intergroup P = .01 and P = .001, respectively). HPGDS mRNA and BAL DP1 levels did not differ by level of control.

TABLE III.

Level of NAEPP asthma control and PGD2 pathway markers in all asthmatic patients

Level of asthma control
Well controlled
Not well controlled
Very poorly controlled
PGD2 pathway marker No. Median (IQR) No. Median (IQR) No. Median (IQR Overall P value
BAL fluid PGD2 (pg/mL) 11 6.5 (4.1-7.0) 25 7.0 (5.0- 10.5) 43 9.0 (6.0-19.0) .014*
Epithelial HPGDS mRNA 10 6.2 (0.9-17.6) 21 4.4 (2.5-8.0) 32 3.0 (1.7-8.1) .84
HPGDS+ cells in
 epithelium (%)
7 1.4 (0.6-2.2) 16 2.0 (0.1-3.1) 17 3.8 (1.8-4.8) .038
BAL cell CRTH2 mRNA 10 0.03 (0.008-0.1) 25 0.15 (0.06-0.8) 34 0.2 (0.1-1.8) .004*
CRTH2+ BAL cells (%) 6 4.7 (3.0-5.9) 14 2.5 (1.6-6.5) 15 5.9 (4.3-9.5) .041
BAL cell DP1 mRNA 10 0.7 (0.2-2.2) 25 3.6 (1.0-5.7) 34 1.8 (0.7-9.2) .11
DP1+ BAL cells (%) 6 3.6 (2.7-5.4) 14 4.6 (3.3-5.9) 15 3.9 (3.3-4.6) .35

IQR, Interquartile range.

*

Significant intergroup comparisons were found between patients with well-controlled asthma and very those with poorly controlled asthma. The asthma control proxy was based on 2007 NAEPP guidelines.

Association with TH2 inflammation

Peripheral blood eosinophilia (≥300/mL) and high Feno levels (≥33 ppb, the median in these asthmatic patients), which were used as putative TH2 inflammation markers,24-26 were evaluated in relation to PGD2 pathway markers. Asthmatic patients with high blood eosinophil numbers had increased PGD2 levels, HPGDS+ cell numbers and mRNA levels, and CRTH2 mRNA levels (all P ≤ .035). Those with high Feno levels had increased PGD2 levels (P = .02, see Table E7 in this article’s Online Repository at www.jacionline.org). Asthmatic patients with both high Feno levels and high peripheral eosinophil numbers compared with those with low levels of both had significantly increased PGD2 and CRTH2 mRNA levels (P = .008 and P = .006, respectively). These data support the association of PGD2 pathway activation with TH2 inflammatory markers, despite the use of ICSs and even OCSs.

DISCUSSION

This study identified a coordinated upregulation of the PGD2 pathway, from enzyme to product to receptor, particularly in patients with severe, poorly controlled, TH2-associated asthma. Expanding on our previous study, BAL fluid PGD2 levels, measured by means of ELISA and confirmed by using the highly sensitive and specific LCMS, were highest in SAs.6 Levels of the likely enzymatic source of this PGD2, HPGDS, were also higher in the SA epithelium, correlated with its product (PGD2), and strongly correlated with MC proteases, supporting an MC source. Although protein and mRNA levels for both PGD2 receptors were detectable in luminal inflammatory cells from all groups, neither DP1+ cells nor mRNA levels differed across groups. In contrast, BAL cell CRTH2+ cell numbers and mRNA levels were highest in SAs and markedly different from those seen in patients with milder asthma and HCs, with increases in both single-positive (likely eosinophils and perhaps basophils) and double-positive CRTH2+/CD3+ cell numbers. Finally, upregulation of several elements of this pathway tracked with markers of TH2 inflammation, including both Feno and blood eosinophils.

In addition to a differentiation of this pathway by using traditional severity definitions, this study was a novel evaluation of its relationship to an index of asthma control, which closely mirrors that suggested in asthma guidelines and which was able to distinguish patients with well-controlled, not well-controlled, and very poorly controlled asthma on the basis of symptoms, β-agonist use, and FEV1. When asthmatic patients were characterized into these groups, those with the worst control again had the greatest expression and activation of this pathway, with increases in epithelial HPGDS, BAL fluid PGD2, and luminal cell CRTH2 levels. Similarly, assessing future risk, asthmatic patients with an exacerbation in the prior 12 months had increased pathway activation compared with those without an exacerbation.

PGD2 generation requires phospholipases to generate arachidonic acid, COXs to convert arachidonic acid to PGH2, and prostaglandin D synthases to catalyze the isomerization of PGH2 to PGD2.8-10 The involvement of multiple enzymes in PGD2 generation suggests that differences in any of these enzymes could contribute to differences in BAL fluid PGD2 levels. However, the correlation between PGD2 and HPGDS suggests that upregulation and activation of this enzyme plays a critical role. Unfortunately, human data on the regulation and activation of HPGDS, a glutathione-S-transferase, are limited.8,9 HPGDS expression and activity have been reported to be regulated by octomer-binding protein 1, although the transcription factors regulating human and murine HPGDS expression differ quite dramatically, suggesting species-specific regulation.9,27,28 Additionally, little is known about expressional control in MCs, and in fact, HPGDS might be constitutively expressed, suggesting a mere increase in MC numbers could increase HPGDS presence/activation.9,10 Data from this study strongly suggest MCs are the source of HPGDS and ultimately PGD2 in asthmatic patients. Although the presence of MC HPGDS is well described in the nose and esophagus,11-13 this is the first study linking HPGDS to the MC in asthmatic patients. Interestingly, the weaker correlations of HPGDS and MCs in the HCs suggest that PGD2 might arise from other sources in these subjects, including macrophages.9,10 Finally, although a recent study reported HPGDS expression in eosinophils, very few immunostained HPGDS+ cells appeared to be eosinophils.29

No matter the cell mix, the HPGDS product PGD2 was confirmed to be increased in BAL fluid in patients with severe and poorly controlled asthma. However, similar to the previous report,6 PGD2 levels were low, raising questions about the specificity of these ELISA-based values. The ELISA levels were then validated in small numbers by using LCMS, correlating extremely well (rs = 0.80, P = .006). Thus we are confident that they reflect actual values. Because PGD2 is rapidly converted to various metabolites,9,10 the strong relationship of the ELISA and LCMS PGD2 values, despite low levels, support the ongoing generation of the unstable parent compound, which is possibly indicative of chronic MC activation.10

The effects of PGD2 depend on which of the 3 receptors is activated and its location.9,10 TP, which is likely responsible for PGD2’s acute bronchoconstricting effects, is found primarily on smooth muscle and platelets and thus not studied here. DP1, which is found on a variety of cells, including smooth muscle cells, goblet cells, dendritic cells, platelets, TH2 cells, and eosinophils, can be both anti-inflammatory and proinflammatory.9,10 DP1 activation causes vasodilation and bronchodilation, likely through cyclic AMP increases, whereas reports are conflicting on inflammation.9,10,14,15 Results of DP1 antagonist studies have been negative to date,30 which is consistent with our data showing that DP1 did not differentiate asthma severity, control, exacerbation, or TH2 inflammation.

Despite binding PGD2 with similar affinity to DP1, CRTH2 is not structurally related and signals through a different mechanism, inhibiting adenyl cyclase, reducing cyclic AMP, and increasing intracellular calcium levels, thereby activating chemotaxis, cell shape change, and TH2 cytokine expression.9,10 In human TH2 cells PGD2 induced TH2 cytokine production in a dose-dependent manner.31 A selective CRTH2, but not DP1, agonist mimicked PGD2’s effects, suggesting that TH2 activation is mediated by CRTH2 and not DP1.31 This is supported by our findings of positive relationships between both CRTH2 mRNA and IHC/protein with BAL fluid PGD2 levels. Furthermore, BAL cell CRTH2 levels were strongly related to asthma severity, exacerbation, and control. Supporting these pathologic studies, a selective oral CRTH2 antagonist, OC000459, improved FEV1 and symptoms and decreased eosinophil numbers in patients with steroid-naive asthma.17 OC000459 also inhibited human TH2 cell chemotaxis, cytokine production, and eosinophil activation and blunted the late asthmatic response and sputum eosinophil increase after allergen challenge.18,32 Recently, the CRTH2 antagonist AMG 853 did not improve symptoms or lung function in patients with moderate-to-severe asthma receiving ICSs,33 suggesting that targeting a specific asthmatic phenotype is important for drug efficacy.

These in vitro and pharmacologic studies all support a strong relationship of PGD2 and CRTH2 to TH2 immune processes. Additionally, expression profiling studies related MCs to a TH2 immune process in patients with steroid-naive asthma.34 To determine whether a relationship persisted in patients with more severe asthma, we compared expression of our PGD2 pathway markers with both Feno levels and blood eosinophil numbers, which were reported to be surrogates of TH2 inflammation.24-26 Asthmatic patients with high blood eosinophil numbers had significantly higher PGD2, HPGDS, and CRTH2 expression than those with low eosinophil numbers. Furthermore, those with high Feno levels also had increased PGD2 levels. These relationships persisted despite corticosteroid use. Thus our data link the PGD2 pathway, in particular PGD2 and CRTH2, to a TH2 immune process, perhaps through direct effects of PGD2/CRTH2 to activate and maintain TH2 cells, as well as eosinophils.

This study is not without some limitations. Although SARP did not directly assess corticosteroid adherence, SAs had numerically higher levels of HPGDS, CRTH2, and PGD2 than any other group. Thus if subjects were simply not taking their corticosteroids, these levels should have been similar to those seen in the groups with milder asthma. The asthma control proxy used here obtained symptoms, activity, medication frequency, and even FEV1 data days if not weeks before the actual bronchoscopy, with the symptom and β-agonist scale used relying on a 3-month recall. Despite this, increases in PGD2 pathway elements were associated with historical worsening of asthma control and historical exacerbations, which is consistent with an association with an exacerbation-prone phenotype. Lastly, not every measurement was performed on every subject because of limited human samples, but the overall sample size for such a human-based study is still quite large.

In summary, this study presents evidence for an active PGD2 pathway, with increases in levels of the enzyme HPGDS in association with its product, PGD2, and one of its receptors, CRTH2, in a TH2 asthma phenotype, specifically in association with severe poorly controlled disease. The relationship of the PGD2-generating enzyme to the epithelial MC signature suggests that the source for this PGD2 is the MC, whereas the increased symptoms could be due to activation, migration, or both of both eosinophils and TH2 lymphocytes. However, much remains to be understood regarding the factors regulating and activating this pathway in asthmatic patients. Studies targeting CRTH2 antagonists in patients with severe, poorly controlled asthma with evidence for TH2 pathway activation are needed to determine whether this pathway is critical to the maintenance or even augmentation of TH2 inflammation and the accompanying asthma.

METHODS

Subjects, allergy testing, and spirometry

Male and female subjects 18 to 65 years of age were recruited through SARP and the Electrophilic Fatty Acid Derivatives in Asthma study. Subjects were nonsmokers in the last year and had a 5 or less pack-year smoking history. Subjects completed clinical questionnaires, allergy testing, blood draw (including complete blood count with differential and IgE measurement), and Feno measurement, as previously described.E1,E2 Baseline and postbronchodilator spirometry was performed according to the ATS guidelines and the SARP protocol.E1 Atopy was assessed by means of skin prick testing to 14 common aeroallergens. Subjects were considered atopic if there was 1 or more positive skin reaction (wheal larger than that elicited by the saline control). Serum IgE levels and complete blood counts with differentials were measured in the clinical laboratory of the University of Pittsburgh Medical Center.

Questionnaires and classification of asthma control

Information collected through questionnaires included general demographics, medical history, medication use, smoking history, and frequency of asthma symptoms and exacerbations. An asthma disease control measure was constructed to proxy for the 2007 NAEPP guidelines for those 12 years of age or older.E3 Five domains were analyzed by using variables captured within the clinical and medication questionnaires and baseline spirometry. Asthma control was classified into 3 groups: well controlled, not well controlled, and very poorly controlled based on daytime symptom frequency (shortness of breath, wheezing, and chest tightness), nighttime awakenings, interference with normal activity, SABA use, and FEV1 percent predicted. Subjects rated symptoms, awakening, and SABA use frequency over the past 3 months on a 1- to 6-point scale: 1, never; 2, monthly; 3, weekly but 2 or fewer times per week; 4, more than 2 times per week but less than daily; 5, daily; and 6, 2 or more times per day. Interference with normal activity over the past 2 weeks was determined by using the Juniper AQLQ activity limitation domain.E4 Consistent with NAEPP guidelines, subjects were assigned to the lowest asthma control group when 1 or more of the 5 criteria for that group was met. Well-controlled asthma was classified by using the following: symptoms 2 or fewer days per week (1, 2, or 3 on frequency scale), nighttime awakenings 2 or fewer times per month (1 or 2 on frequency scale), no interference with normal activity (activity score on AQLQ of 7-5), use of SABAs 2 or fewer days per week (1, 2, or 3 on frequency scale), and FEV1 of greater than 80% of predicted value. Patients with not well-controlled asthma had symptoms more than 2 days per week (4 or 5 on the frequency scale), nighttime awakenings 1 to 3 times per week (3 or 4 on the frequency scale), some limitation of normal activity (activity score on AQLQ of <5-3), SABA use for symptom control on more than 2 days per week (4 or 5 on the frequency scale), and FEV1 of 60% to 80% of predicted value. Patients with very poorly controlled asthma reported symptoms throughout the day (6 on the frequency scale), nighttime awakenings 4 or more times per week (5 or 6 on the frequency scale), extreme limitation in normal activity (activity score on AQLQ of <3-1), SABA use several times per day (6 on the frequency scale), and FEV1 of less than 60% of predicted value.

Bronchoscopy and sample processing

During bronchoscopy, epithelial brushings from the proximal airways and BAL cells and fluid were obtained and processed according to previously published protocols and the SARP manual of procedures.E2,E5 BAL cells and fluid were obtained and separated by means of centrifugation at 600g.E1 BAL fluid from the first 100 mL was placed in aliquots and stored at −80°C for measurement of PGD2 levels. Cells were placed in Qiazol (Qiagen) for extraction of mRNA.

BAL fluid PGD2 measurement

ELISA

PGD2 levels were measured by using an ELISA with commercially available components (Cayman Chemical Company) at ElisaTech, as previously described.E2 Samples were first purified and concentrated with C18 Sep-Pak cartridges (Waters Corp, Milford, Mass) and then derivatized per the ELISA manufacturer’s instructions. The assay’s limit of detection was 2 to 5 pg/mL.

LCMS

A subset of matched BAL fluid samples (n = 10) was analyzed by using LCMS to validate the PGD2 measurements. PGD2 was purchased from Cayman Chemical Company. Water and acetonitrile were purchased from Honeywell Burdick & Jackson (Morristown, NJ). Four milliliters of BAL fluid was used for the liquid-liquid extraction of PGD2. Internal standard, 10 μL of 50 ng/mL [2H9]-PGD2, was added to each sample, followed by an equilibration time of 10 minutes. Next, 4 mL of ethyl acetatewas added, and the samples were shaken for 30 minutes and then centrifuged for 10 minutes at 2900g. The organic layer was removed, dried under nitrogen, and reconstituted in 100 μL of methanol. An injection volume of 10 μL was used for analysis with LCMS.

Analyses of PGD2 levels were conducted with a Shimadzu LC-20AD HPLC system (Columbia, Md) coupled to an AB-Sciex API-5000 triple-quadrupole mass spectrometer (Framingham, Mass). The liquid chromatography (LC) method used a Phenomenex Luna C18(2) column (2.0 mm ID × 150 mm, 3 μm) by using water with 0.1% acetic acid as mobile phase A and acetonitrile with 0.1% acetic acid as mobile phase B at a flow rate of 200 μL/min. The gradient increased from 10% B to 55% B in 45 minutes, followed by a 3-minute wash step at 100% B and back to equilibration at 10% B. The following mass spectrometer parameters were set: collision gas, 4 units; curtain gas, 40 units; ion source gas 1, 40 units; ion source gas 2, 40 units; ion spray voltage, −4.2 kV; ionization temperature, 350°C; decluster potential, −55 V; entrance potential, −5 V; collision energy, −22 eV; and collision cell exit potential, −18.4 V. Single reaction monitoring (SRM) analyses were conducted in negative electro-spray ionization (ESI) mode. The following transitions were monitored for PGD2: m/z 351.2 (M-H+) → m/z 271.2 (M-H+-2H2O-CO2) and m/z 360.2 (M-H+) → m/z 280.2 (M-H+-2H2O-CO2) for [2H9]-PGD2.

PGD2 standard was used to prepare calibration solutions at different concentrations (1, 10, 50, 100, 500, 1000, and 5000 pg of each). The corresponding [2H9]-PGD2 internal standard was then added to the calibration solutions and experimental samples (500 pg). Calibration solutions underwent the same sample preparation and analytic procedures as the experimental samples, as described above. Calibration curves were calculated with a linear regression analysis of the peak area ratios of standard versus the internal standard. Prostaglandin levels were calculated by means of interpolation from the calibration curve and reported in picograms per milliliter. The assay’s limit of detection for PGD2 was 0.5 pg/mL.

qRT-PCR

Total RNA was extracted from epithelial cell brushings and BAL cells in Qiazol (Qiagen). Reverse transcription was performed with 1 μg of total RNA and random hexamers in a 50-μL reaction, according to the manufacturer’s protocol (PE Applied Biosystems, Foster City, Calif). Epithelial brushing mRNA expression of HPGDS and the MC proteases tryptase and CPA3 was determined by using qRT-PCR, as previously described.E2 BAL cell mRNA expression for the PGD2 receptors DP1 and CRTH2/DP2 was also evaluated by means of qRT-PCR. The primers and probes were all purchased from Applied Biosystems (Assays on Demand: HPGDS, Hs00183950_m1; tryptase, Hs02576518_gH; CPA3, Hs00157019_m1; CRTH2, Hs01867513_s1; and DP1, Hs00830594_s1). The probes were labeled with the 5′-reporter dye 6-carboxy fluorescein and the 3′-quencher dye 6-carboxy N, N, N’,N’tetramethylrhodamine. VIC-labeled ready-for-use human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) probe and primers were also obtained from Applied Biosystems (GenBank accession no. NM-002046; part no. 4310884E). Real-time PCR was performed on the ABIPrism 7900 sequence detection system (Applied Biosystems) in the core facilities of the University of Pittsburgh. The mRNA levels for each of the markers were determined by indexing to GAPDH with the formula 1/2ΔCt × 1000. Analysis was performed on epithelial brushing samples if the GAPDH threshold was less than 27 cycles and BAL cell samples if the GAPDH threshold was less than 30.5 cycles.

IHC

All IHC was performed on cytospin preparations fixed in 2% to 4% paraformaldehyde.

Epithelial brushing cytospin preparations for PGD2 enzyme (HPGDS)

After the appropriate blocking steps, cytospin preparations were incubated with rabbit polyclonal antihuman HPGDS antibody (Cayman Chemical Company) at 1:100 dilution overnight at 4°C. Sections were rinsed, incubated with biotinylated secondary goat anti-rabbit antibody for 1 hour at room temperature, rinsed, and incubated in ABC reagent for 45 minutes (both from Vector Laboratories, Burlingame, Calif). Sections were developed with the chromogen 3-amino-9-ethylcarbazole (AEC) and then counterstained with hematoxylin. Slides were overlaid with Crystal Mount (Electron Microscopy Sciences, Hatfield, Pa). Slides were blinded, and numbers of HPGDS+ cells were counted by 2 independent observers from a total of at least 500 cells in random fields at ×40 magnification with the Nikon Eclipse TS100 (Mellville, NY). The percentage of HPGDS+ cells was calculated by dividing the number of HPGDS+ cells by the total number of cells counted. Control specimens were processed in the same manner, but the primary antibody was omitted.

BAL cell cytospin preparations for PGD2 receptors

After the appropriate blocking steps, cytospin preparations were incubated with rabbit polyclonal antibody against the DP1 receptor (Cayman Chemical Company) at 1:200 dilution overnight at 4°C. Sections were rinsed, incubated with biotinylated secondary goat anti-rabbit antibody for 1 hour at room temperature, rinsed, and incubated in ABC reagent for 45 minutes (both from Vector Laboratories). Sections were rinsed again and developed with the chromogen AEC and then counterstained with hematoxylin. Slides were overlaid with Crystal Mount (Electron Microscopy Sciences). Slides were blinded, and the number of DP1+ cells was counted by 2 independent observers from a total of at least 500 cells in random fields at ×40 magnification with the Nikon Eclipse TS100. The percentage of DP1+ cells was determined by dividing the number of DP1+ cells by the total number of inflammatory cells counted. Control specimens were processed in the same manner, but the primary antibody was omitted.

BAL cell cytospin preparations were double-labeled for CRTH2 and CD3 by using sequential immunostaining with anti-CRTH2 rat mAb (1:20, BM16, Santa Cruz Biotechnology) and anti-CD3 mouse mAb (1:200, Becton Dickinson, San Jose, Calif). After blocking, cytospin preparations were incubated with CRTH2 antibody, rinsed, incubated with biotinylated secondary rabbit anti-rat antibody followed by ABC reagent (both from Vector laboratories), and developed with AEC. After blocking with normal serum, rinsing, and treatment with the Avidin/Biotin blocking kit (Vector Laboratories), cytospin preparations were incubated with CD3 antibody and then biotinylated secondary horse anti-mouse antibody (Vector Laboratories) for 1 hour. Slides were treated with ABC–alkaline phosphatase complex and developed with alkaline phosphatase substrate solution as the chromogen (both from Vector Laboratories). Slides were blinded, and the number of cells positive for CRTH2, CD3, or both were counted by 2 independent observers from a total of at least 600 cells, as previously described. Cells were classified as CD3+ only (blue staining), CRTH2+ only (red), CRTH2+/CD3+ double positive, or negative for either marker. Controls were performed, as previously described.

Supplementary Material

01

FIG E1. Final selection of the study population. In total, 88 (79%) had data from each of the 3 sample types (BAL fluid, BAL cells, and epithelial brushings).

FIG E2. PGD2 levels measured by means of ELISA in BAL fluid samples in nonoverlapping subjects (n = 79).

FIG E3. Correlation between PGD2 levels measured by using ELISA and LCMS. Relative levels for BAL fluid PGD2 (in picograms per milliliter) by using ELISA and LCMS are shown on a log10 scale. In this subset (n = 10) the values were compared by using Spearman correlation and strongly correlated (rs = 0.80, P = .006), confirming the ELISA results.

TABLE E1. Reason for missing mRNA data by subject severity

TABLE E2. Baseline demographic comparison for all subjects versus subjects with additional IHC data

TABLE E3. PGD2 pathway markers analyzed by completeness of parameters

TABLE E4. Relative epithelial brushing MC protease mRNA by subject group indexed to GAPDH

TABLE E5. PGD2 receptor IHC data of BAL cell cytospin preparations (n = 47)

TABLE E6. Distribution of asthmatic patients based on asthma severity and NAEPP asthma control proxy (n = 79)

TABLE E7. Relationship between TH2 inflammatory markers and PGD2 pathway markers in asthmatic patients

Key messages.

  • Levels of several elements of the PGD2 pathway, including the enzyme HPGDS, the CRTH2 receptor, and the end product PGD2 are increased in patients with severe, poorly controlled asthma.

  • These pathway elements associate with markers of TH2 inflammation and exacerbations.

  • The PGD2 pathway could be a potential new therapeutic target in patients with severe, poorly controlled, TH2-high asthma unresponsive to standard therapies.

Acknowledgments

Supported by National Institute of Health/National Heart, Lung, and Blood Institute grants HL-69174, HL-109152-01, HL064937-10, NIH-F32 AI085633, and CTSI UL1 RR024153.

Abbreviations used

AEC

3-Amino-9-ethylcarbazole

AQLQ

Asthma Quality of Life Questionnaire

ATS

American Thoracic Society

BAL

Bronchoalveolar lavage

CPA3

Carboxypeptidase A3

CRTH2/DP2

Chemoattractant receptor–homologous molecule expressed on TH2 lymphocytes

DP1

PGD2 receptor 1

Feno

fraction of exhaled nitric oxide

GAPDH

Glyceraldehyde-3-phosphate dehydrogenase

HC

Healthy control subject

HPGDS

Hematopoietic prostaglandin D synthase

ICS

Inhaled corticosteroid

IHC

Immunohistochemistry

LCMS

Liquid chromatography mass spectrometry

MC

Mast cell

NAEPP

National Asthma Education and Prevention Program

OCS

Oral corticosteroid

PGD2

Prostaglandin D2

qRT-PCR

Quantitative real-time PCR

SA

Patient with severe asthma

SABA

Short-acting β-agonist

SARP

Severe Asthma Research Program

TP

Thromboxane receptor

Footnotes

Disclosure of potential conflict of interest: M. L. Fajt has received grants from the National Institutes of Health (NIH). S. L. Gelhaus has received grants/has grants pending from the NIH. B. Freeman has received grants from the NIH; has received travel expenses from the NIH; has patents planned, pending, or issued by Complexa; has stock/stock options in Complexa and Nitromega. S. E. Wenzel has received consulting fees or honoraria from Actelion and Merck; has received payment for a Multicenter Study conducted for Array; has consultant arrangements with Amgen, Regeneron, and Novartis; and has grants/grants pending from Amgen, Genentech MedImmune, Sanofi Aventis, GlaxoSmithKline, and Merck. The rest of the authors declare that they have no relevant conflicts of interest.

REFERENCES

  • 1.American Thoracic Society Proceedings of the ATS workshop on refractory asthma: current understanding, recommendations, and unanswered questions. Am J Respir Crit Care Med. 2000;162:2341–51. doi: 10.1164/ajrccm.162.6.ats9-00. [DOI] [PubMed] [Google Scholar]
  • 2.National Asthma Education and Prevention Program . Guidelines for the diagnosis and management of asthma: expert panel report 3 (EPR3) National Institutes of Health/National Heart, Lung, and Blood Institute; Bethesda (MD): [Accessed January 16, 2012]. 2007. Publication no. 08-4051. Available at: http://www.nhlbi.nih.gov/guidelines/asthma/asthgdln.pdf. [Google Scholar]
  • 3.Moore WC, Bleecker ER, Curran-Everett D, Erzurum SC, Ameredes BT, Bacharier L, et al. National Heart, Lung, Blood Institute’s Severe Asthma Research Program. Characterization of the severe asthma phenotype by the National Heart, Lung, and Blood Institute’s Severe Asthma Research Program. J Allergy Clin Immunol. 2007;119:405–13. doi: 10.1016/j.jaci.2006.11.639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Balzar S, Chu HW, Strand M, Wenzel S. Relationship of small airway chymase-positive mast cells and lung function in severe asthma. Am J Respir Crit Care Med. 2005;171:431–9. doi: 10.1164/rccm.200407-949OC. [DOI] [PubMed] [Google Scholar]
  • 5.Wenzel SE, Balzar S, Ampleford E, Hawkins GA, Busse WW, Calhoun WJ, et al. IL4R alpha mutations are associated with asthma exacerbations and mast cell/IgE expression. Am J Respir Crit Care Med. 2007;175:570–6. doi: 10.1164/rccm.200607-909OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Balzar S, Fajt ML, Comhair SA, Erzurum SC, Bleecker E, Busse WW, et al. Mast cell phenotype, location, and activation in severe asthma: data from the severe asthma research program. Am J Respir Crit Care Med. 2011;183:299–309. doi: 10.1164/rccm.201002-0295OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Galli SJ, Nakae S, Tsai M. Mast cells in the development of adaptive immune responses. Nat Immunol. 2005;6:135–42. doi: 10.1038/ni1158. [DOI] [PubMed] [Google Scholar]
  • 8.Kanaoka Y, Fujimori K, Kikuno R, Sakaguchi Y, Urade Y, Hayaishi O. Structure and chromosomal localization of human and mouse genes for hematopoietic prostaglandin D synthase. Conservation of the ancestral genomic structure of sigma-class glutathione S-transferase. Eur J Biochem. 2000;267:3315–22. doi: 10.1046/j.1432-1327.2000.01362.x. [DOI] [PubMed] [Google Scholar]
  • 9.Herlong JL, Scott TR. Positioning prostanoids of the D and J series in the immunopathogenic scheme. Immunol Lett. 2006;102:121–31. doi: 10.1016/j.imlet.2005.10.004. [DOI] [PubMed] [Google Scholar]
  • 10.Pettipher R, Hansel TT, Armer R. Antagonism of the prostaglandin D2 receptors DP1 and CRTH2 as an approach to treat allergic diseases. Nat Rev Drug Discov. 2007;6:313–25. doi: 10.1038/nrd2266. [DOI] [PubMed] [Google Scholar]
  • 11.Nantel F, Fong C, Lamontagne S, Wright DH, Giaid A, Desrosiers M, et al. Expression of prostaglandin D synthase and the prostaglandin D2 receptors DP and CRTH2 in human nasal mucosa. Prostaglandins Other Lipid Mediat. 2004;73:87–101. doi: 10.1016/j.prostaglandins.2003.12.002. [DOI] [PubMed] [Google Scholar]
  • 12.Okano M, Fujiwara T, Sugata Y, Gotoh D, Masaoka Y, Sogo M, et al. Presence and characterization of prostaglandin D2-related molecules in nasal mucosa of patients with allergic rhinitis. Am J Rhinol. 2006;20:342–8. doi: 10.2500/ajr.2006.20.2865. [DOI] [PubMed] [Google Scholar]
  • 13.Abonia JP, Blanchard C, Butz BB, Rainey HF, Collins MH, Stringer K, et al. Involvement of mast cells in eosinophilic esophagitis. JAllergy Clin Immunol. 2010;126:140–9. doi: 10.1016/j.jaci.2010.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tanaka K, Hirai H, Takano S, Nakamura M, Nagata K. Effects of prostaglandin D2 on helper T cell functions. Biochem Biophys Res Commun. 2004;316:1009–14. doi: 10.1016/j.bbrc.2004.02.151. [DOI] [PubMed] [Google Scholar]
  • 15.Gosset P, Pichavant M, Faveeuw C, Bureau F, Tonnel AB, Trottein F. Prostaglandin D2 affects the differentiation and functions of human dendritic cells: impact on the T cell response. Eur J Immunol. 2005;35:1491–500. doi: 10.1002/eji.200425319. [DOI] [PubMed] [Google Scholar]
  • 16.Stebbins KJ, Broadhead AR, Correa LD, Scott JM, Truong YP, Stearns BA, et al. Therapeutic efficacy of AM156, a novel prostanoid DP2 receptor antagonist, in murine models of allergic rhinitis and house dust mite-induced pulmonary inflammation. Eur J Pharmacol. 2010;638:142–9. doi: 10.1016/j.ejphar.2010.04.031. [DOI] [PubMed] [Google Scholar]
  • 17.Barnes N, Pavord I, Chuchalin A, Bell J, Hunter M, Lewis T, et al. A randomized, double-blind, placebo-controlled study of the CRTH2 antagonist OC000459 in moderate persistent asthma. Clin Exp Allergy. 2012;42:38–48. doi: 10.1111/j.1365-2222.2011.03813.x. [DOI] [PubMed] [Google Scholar]
  • 18.Singh D, Cadden P, Hunter M, Collins LP, Perkins M, Pettipher R, et al. Inhibition of the asthmatic allergen challenge response by the CRTH2 antagonist OC000459. Eur Respir J. 2012;41:46–52. doi: 10.1183/09031936.00092111. [DOI] [PubMed] [Google Scholar]
  • 19.Fajt ML, Balzar S, Trudeau JB, Westcott JY, Wenzel SE. Elevated prostaglandin D2 (PGD2) levels in bronchoalveolar lavage fluid (BALF) in severe asthma. J Allergy Clin Immunol. 2010;125:AB43. [Google Scholar]
  • 20.Fajt ML, Balzar S, Trudeau JB, Hu H, Wenzel SE. Epithelial hematopoietic prostaglandin (PG) D synthase is increased in asthma. Am J Respir Crit Care Med. 2010;181:A1440. [Google Scholar]
  • 21.Fajt ML, Balzar S, Trudeau JB, Wenzel SE. CRTH2 receptor and its ligand, PGD2, are increased in severe asthma. Am J Respir Crit Care Med. 2011;183:A4446. [Google Scholar]
  • 22.Juniper EF, Guyatt GH, Epstein RS, Ferrie PJ, Jaeschke R, Hiller TK. Evaluation of impairment of health related quality of life in asthma: development of a questionnaire for use in clinical trials. Thorax. 1992;47:76–83. doi: 10.1136/thx.47.2.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moore WC, Evans MD, Bleecker ER, Busse WW, Calhoun WJ, Castro M, et al. National Heart, Lung, and Blood Institute’s Severe Asthma Research Program. Safety of investigative bronchoscopy in the Severe Asthma Research Program. J Allergy Clin Immunol. 2011;128:328–36.e3. doi: 10.1016/j.jaci.2011.02.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chibana K, Trudeau JB, Mustovich AT, Hu H, Zhao J, Balzar S, et al. IL-13 induced increases in nitrite levels are primarily driven by increases in inducible nitric oxide synthase as compared with effects on arginases in human primary bronchial epithelial cells. Clin Exp Allergy. 2008;38:936–46. doi: 10.1111/j.1365-2222.2008.02969.x. [DOI] [PubMed] [Google Scholar]
  • 25.Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, Arron JR, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med. 2011;365:1088–98. doi: 10.1056/NEJMoa1106469. [DOI] [PubMed] [Google Scholar]
  • 26.Pavord ID, Korn S, Howarth P, Bleecker ER, Buhl R, Keene ON, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet. 2012;380:651–9. doi: 10.1016/S0140-6736(12)60988-X. [DOI] [PubMed] [Google Scholar]
  • 27.Mahmud I, Ueda N, Yamaguchi H, Yamashita R, Yamamoto S, Kanaoka Y, et al. Prostaglandin D synthase in human megakaryoblastic cells. J Biol Chem. 1997;272:28263–6. doi: 10.1074/jbc.272.45.28263. [DOI] [PubMed] [Google Scholar]
  • 28.Fujimori K, Kanaoka Y, Sakaguchi Y, Urade Y. Transcriptional activation of the human hematopoietic prostaglandin D synthase gene in megakaryoblastic cells. Roles of the oct-1 element in the 5′-flanking region and the AP-2 element in the untranslated exon 1. J Biol Chem. 2000;275:40511–6. doi: 10.1074/jbc.M007688200. [DOI] [PubMed] [Google Scholar]
  • 29.Luna-Gomes T, Magalhães KG, Mesquita-Santos FP, Bakker-Abreu I, Samico RF, Molinaro R, et al. Eosinophils as a novel cell source of prostaglandin D2: autocrine role in allergic inflammation. J Immunol. 2011;187:6518–26. doi: 10.4049/jimmunol.1101806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Philip G, van Adelsberg J, Loeys T, Liu N, Wong P, Lai E, et al. Clinical studies of the DP1 antagonist laropiprant in asthma and allergic rhinitis. J Allergy Clin Immunol. 2009;124:942–8. doi: 10.1016/j.jaci.2009.07.006. [DOI] [PubMed] [Google Scholar]
  • 31.Xue L, Gyles SL, Wettey FR, Gazi L, Townsend E, Hunter MG, et al. Prostaglandin D2 causes preferential induction of proinflammatory Th2 cytokine production through an action on chemoattractant receptor-like molecule expressed on Th2 cells. J Immunol. 2005;175:6531–6. doi: 10.4049/jimmunol.175.10.6531. [DOI] [PubMed] [Google Scholar]
  • 32.Pettipher R, Vinall SL, Xue L, Speight G, Townsend ER, Gazi L, et al. Pharmacologic profile of OC000459, a potent, selective, and orally active D prostanoid receptor 2 antagonist that inhibits mast cell-dependent activation of T helper 2 lymphocytes and eosinophils. J Pharmacol Exp Ther. 2012;340:473–82. doi: 10.1124/jpet.111.187203. [DOI] [PubMed] [Google Scholar]
  • 33.Busse WW, Wenzel SE, Meltzer EO, Kerwin EM, Liu MC, Zhang N, et al. Safety and efficacy of the prostaglandin D(2) receptor antagonist AMG 853 in asthmatic patients. J Allergy Clin Immunol. 2013;131:339–45. doi: 10.1016/j.jaci.2012.10.013. [DOI] [PubMed] [Google Scholar]
  • 34.Dougherty RH, Sidhu SS, Raman K, Solon M, Solberg OD, Caughey GH, et al. Accumulation of intraepithelial mast cells with a unique protease phenotype in T(H)2-high asthma. J Allergy Clin Immunol. 2010;125:1046–53. doi: 10.1016/j.jaci.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • E1.Moore WC, Bleecker ER, Curran-Everett D, Erzurum SC, Ameredes BT, Bacharier L, et al. National Heart, Lung, Blood Institute’s Severe Asthma Research Program. Characterization of the severe asthma phenotype by the National Heart, Lung, and Blood Institute’s Severe Asthma Research Program. J Allergy Clin Immunol. 2007;119:405–13. doi: 10.1016/j.jaci.2006.11.639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • E2.Balzar S, Fajt ML, Comhair SA, Erzurum SC, Bleecker E, Busse WW, et al. Mast cell phenotype, location, and activation in severe asthma: data from the severe asthma research program. Am J Respir Crit Care Med. 2011;183:299–309. doi: 10.1164/rccm.201002-0295OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • E3.National Asthma Education and Prevention Program . Guidelines for the diagnosis and management of asthma: expert panel report 3 (EPR3) National Institutes of Health/National Heart, Lung, and Blood Institute; Bethesda (MD): [Accessed January 16, 2012]. 2007. Publication no. 08-4051. Available at: http://www.nhlbi.nih.gov/guidelines/asthma/asthgdln.pdf. [Google Scholar]
  • E4.Juniper EF, Guyatt GH, Epstein RS, Ferrie PJ, Jaeschke R, Hiller TK. Evaluation of impairment of health related quality of life in asthma: development of a questionnaire for use in clinical trials. Thorax. 1992;47:76–83. doi: 10.1136/thx.47.2.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • E5.Moore WC, Evans MD, Bleecker ER, Busse WW, Calhoun WJ, Castro M, et al. National Heart, Lung, and Blood Institute’s Severe Asthma Research Program. Safety of investigative bronchoscopy in the Severe Asthma Research Program. J Allergy Clin Immunol. 2011;128:328–36.e3. doi: 10.1016/j.jaci.2011.02.042. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

01

FIG E1. Final selection of the study population. In total, 88 (79%) had data from each of the 3 sample types (BAL fluid, BAL cells, and epithelial brushings).

FIG E2. PGD2 levels measured by means of ELISA in BAL fluid samples in nonoverlapping subjects (n = 79).

FIG E3. Correlation between PGD2 levels measured by using ELISA and LCMS. Relative levels for BAL fluid PGD2 (in picograms per milliliter) by using ELISA and LCMS are shown on a log10 scale. In this subset (n = 10) the values were compared by using Spearman correlation and strongly correlated (rs = 0.80, P = .006), confirming the ELISA results.

TABLE E1. Reason for missing mRNA data by subject severity

TABLE E2. Baseline demographic comparison for all subjects versus subjects with additional IHC data

TABLE E3. PGD2 pathway markers analyzed by completeness of parameters

TABLE E4. Relative epithelial brushing MC protease mRNA by subject group indexed to GAPDH

TABLE E5. PGD2 receptor IHC data of BAL cell cytospin preparations (n = 47)

TABLE E6. Distribution of asthmatic patients based on asthma severity and NAEPP asthma control proxy (n = 79)

TABLE E7. Relationship between TH2 inflammatory markers and PGD2 pathway markers in asthmatic patients

RESOURCES