Gene-metabolite expression in blood can discriminate allergen-induced isolated early from dual asthmatic responses

Amrit Singh, Masatsugu Yamamoto, Sarah H Y Kam, Jian Ruan, Gail M Gauvreau, Paul M O'Byrne, J Mark FitzGerald, Robert Schellenberg, Louis-Philippe Boulet, Gabriella Wojewodka, Cynthia Kanagaratham, Juan B De Sanctis, Danuta Radzioch, Scott J Tebbutt, Amrit Singh, Masatsugu Yamamoto, Sarah H Y Kam, Jian Ruan, Gail M Gauvreau, Paul M O'Byrne, J Mark FitzGerald, Robert Schellenberg, Louis-Philippe Boulet, Gabriella Wojewodka, Cynthia Kanagaratham, Juan B De Sanctis, Danuta Radzioch, Scott J Tebbutt

Abstract

Some asthmatic individuals undergoing allergen inhalation challenge develop an isolated early response whereas others develop a dual response (early plus late response). In the present study we have used transcriptomics (microarrays) and metabolomics (mass spectrometry) of peripheral blood to identify molecular patterns that can discriminate allergen-induced isolated early from dual asthmatic responses. Peripheral blood was obtained prior to (pre-) and 2 hours post allergen inhalation challenge from 33 study participants. In an initial cohort of 14 participants, complete blood counts indicated significant differences in neutrophil and lymphocyte counts at pre-challenge between early and dual responders. At post-challenge, significant genes (ALOX15, FADS2 and LPCAT2) and metabolites (lysolipids) were enriched in lipid metabolism pathways. Enzymes encoding for these genes are involved in membrane biogenesis and metabolism of fatty acids into pro-inflammatory and anti-inflammatory mediators. Correlation analysis indicated a strong negative correlation between ALOX15, FADS2, and IL5RA expression with 2-arachidonoylglycerophosphocholine levels in dual responders. However, measuring arachidonic acid and docosahexaenoic acid levels in a validation cohort of 19 participants indicated that the free form of DHA (nmoles/µg of protein) was significantly (p = 0.03) different between early and dual responders after allergen challenge. Collectively these results may suggest an imbalance in lipid metabolism which dictates pro- (anti-) inflammatory and pro-resolving mechanisms. Future studies with larger sample sizes may reveal novel mechanisms and therapeutic targets of the late phase asthmatic response.

Conflict of interest statement

Competing Interests: The authors have the following interests. This study was funded by AllerGen NCE Inc. Metabolon Incorporated (Durham, North Carolina, United States) performed the metabolite profiling and data processing for this study. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Lung function during allergen inhalation…
Figure 1. Lung function during allergen inhalation challenge.
Forced expiratory volume in 1 second (FEV1) measurements at 0.2 h, 0.3 h, 0.5 h, 0.8 h, 1 h, 1.5 h, 2 h, 3 h, 4 h, 5 h, 6 h and 7 h for eight early and six dual responders. FEV1 measurements are statistically different (*p<0.05) between early and dual responders at each of the time points between three and seven hours inclusive. The p-value for each time point comparing ERs and DRs was computed using a robust linear model (see Methods).
Figure 2. Complete blood counts.
Figure 2. Complete blood counts.
Relative frequencies of various leukocytes obtained at pre and post-challenge. The relative neutrophil counts were significantly (p = 0.01) elevated in ERs compared to DRs, whereas the relative lymphocyte counts were significantly reduced (p = 0.02) in ERs compared to DRs at pre-challenge. The p-value was computed using a robust linear model (see Methods); * denotes p

Figure 3. Gene network.

Ingenuity Pathway Analysis…

Figure 3. Gene network.

Ingenuity Pathway Analysis network of differentially expressed genes at post-challenge comparing…

Figure 3. Gene network.
Ingenuity Pathway Analysis network of differentially expressed genes at post-challenge comparing ERs and DRs. Dash lines indicate indirect relationships, whereas solid lines indicate direct relationships. The red colour indicates significant genes at an FDR of 10%. The grey colour indicates non-significant genes at an FDR of 10%.

Figure 4. Network plots highlighting the correlation…

Figure 4. Network plots highlighting the correlation between ΔG and ΔM for early and dual…

Figure 4. Network plots highlighting the correlation between ΔG and ΔM for early and dual responders.
A. Gene-metabolite clusters for early responders (ΔG25,8 and ΔM11,8). B. Gene-metabolite clusters for dual responders (ΔG25,6 and ΔM11,6). A correlation coefficient cut-off of 0.5 is applied to both networks.

Figure 5. Levels of free docosahexaenoic acid…

Figure 5. Levels of free docosahexaenoic acid in the plasma of early and dual responders…

Figure 5. Levels of free docosahexaenoic acid in the plasma of early and dual responders undergoing allergen inhalation challenge.
Levels of DHA (left) decrease in early responders from pre to post-challenge whereas no consistent change is observed in dual responders following allergen challenge. At post-challenge (scaled to pre-challenge levels), DHA was differentially expressed (p = 0.033) between ERs and DRs (right). The p-value was computed using a robust linear model (see Methods).
Figure 3. Gene network.
Figure 3. Gene network.
Ingenuity Pathway Analysis network of differentially expressed genes at post-challenge comparing ERs and DRs. Dash lines indicate indirect relationships, whereas solid lines indicate direct relationships. The red colour indicates significant genes at an FDR of 10%. The grey colour indicates non-significant genes at an FDR of 10%.
Figure 4. Network plots highlighting the correlation…
Figure 4. Network plots highlighting the correlation between ΔG and ΔM for early and dual responders.
A. Gene-metabolite clusters for early responders (ΔG25,8 and ΔM11,8). B. Gene-metabolite clusters for dual responders (ΔG25,6 and ΔM11,6). A correlation coefficient cut-off of 0.5 is applied to both networks.
Figure 5. Levels of free docosahexaenoic acid…
Figure 5. Levels of free docosahexaenoic acid in the plasma of early and dual responders undergoing allergen inhalation challenge.
Levels of DHA (left) decrease in early responders from pre to post-challenge whereas no consistent change is observed in dual responders following allergen challenge. At post-challenge (scaled to pre-challenge levels), DHA was differentially expressed (p = 0.033) between ERs and DRs (right). The p-value was computed using a robust linear model (see Methods).

References

    1. Anderson GP (2008) Endotyping asthma: New insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet 372: 1107–1119.
    1. Tulic MK, Christodoulopoulos P, Hamid Q (2001) Small airway inflammation in asthma. Respir Res 2: 333–339.
    1. Naylor B (1962) The shedding of the mucosa of the bronchial tree in asthma. Thorax 17: 69–72.
    1. Tesfaigzi Y (2008) Regulation of mucous cell metaplasia in bronchial asthma. Curr Mol Med 8: 408–415.
    1. Ebina M, Yaegashi H, Chiba R, Takahashi T, Motomiya M, et al. (1990) Hyperreactive site in the airway tree of asthmatic patients revealed by thickening of bronchial muscles. A morphometric study. Am Rev Respir Dis 141: 1327–1332.
    1. Luskin AT (2005) What the asthma end points we know and love do and do not tell us. J Allergy Clin Immunol 115: S539–45.
    1. Gauvreau GM, Evans MY (2007) Allergen inhalation challenge: A human model of asthma exacerbation. Contrib Microbiol 14: 21–32.
    1. Murray JJ, Tonnel AB, Brash AR, Roberts LJ,2nd, Gosset P, et al (1985) Prostaglandin D2 is released during acute allergic bronchospasm in man. Trans Assoc Am Physicians 98: 275–280.
    1. Liu MC, Hubbard WC, Proud D, Stealey BA, Galli SJ, et al. (1991) Immediate and late inflammatory responses to ragweed antigen challenge of the peripheral airways in allergic asthmatics. cellular, mediator, and permeability changes. Am Rev Respir Dis 144: 51–58.
    1. Gounni AS, Lamkhioued B, Ochiai K, Tanaka Y, Delaporte E, et al. (1994) High-affinity IgE receptor on eosinophils is involved in defence against parasites. Nature 367: 183–186.
    1. Jarjour NN, Calhoun WJ, Kelly EA, Gleich GJ, Schwartz LB, et al. (1997) The immediate and late allergic response to segmental bronchopulmonary provocation in asthma. Am J Respir Crit Care Med 155: 1515–1521.
    1. Wenzel SE, Westcott JY, Smith HR, Larsen GL (1989) Spectrum of prostanoid release after bronchoalveolar allergen challenge in atopic asthmatics and in control groups. an alteration in the ratio of bronchoconstrictive to bronchoprotective mediators. Am Rev Respir Dis 139: 450–457.
    1. Verstraelen S, Bloemen K, Nelissen I, Witters H, Schoeters G, et al. (2008) Cell types involved in allergic asthma and their use in in vitro models to assess respiratory sensitization. Toxicol In Vitro 22: 1419–1431.
    1. Bousquet J, Jeffery PK, Busse WW, Johnson M, Vignola AM (2000) Asthma. From bronchoconstriction to airways inflammation and remodeling. Am J Respir Crit Care Med 161: 1720–1745.
    1. Hansel NN, Diette GB (2007) Gene expression profiling in human asthma. Proc Am Thorac Soc 4: 32–36.
    1. Adamko DJ, Sykes BD, Rowe BH (2012) The metabolomics of asthma: Novel diagnostic potential. Chest 141: 1295–1302.
    1. Yao PL, Tsai MF, Lin YC, Wang CH, Liao WY, et al. (2005) Global expression profiling of theophylline response genes in macrophages: Evidence of airway anti-inflammatory regulation. Respir Res 6: 89.
    1. Hakonarson H, Halapi E, Whelan R, Gulcher J, Stefansson K, et al. (2001) Association between IL-1beta/TNF-alpha-induced glucocorticoid-sensitive changes in multiple gene expression and altered responsiveness in airway smooth muscle. Am J Respir Cell Mol Biol 25: 761–771.
    1. Kam SH, Singh A, He JQ, Ruan J, Gauvreau GM, et al. (2012) Peripheral blood gene expression changes during allergen inhalation challenge in atopic asthmatic individuals. J Asthma 49: 219–226.
    1. Yamamoto M, Singh A, Ruan J, Gauvreau GM, O'Byrne PM, et al. (2012) Decreased miR-192 expression in peripheral blood of asthmatic individuals undergoing an allergen inhalation challenge. BMC Genomics 13: 655–2164-13-655.
    1. Wood LJ, Inman MD, Watson RM, Foley R, Denburg JA, et al. (1998) Changes in bone marrow inflammatory cell progenitors after inhaled allergen in asthmatic subjects. Am J Respir Crit Care Med 157: 99–105.
    1. Gauvreau GM, Lee JM, Watson RM, Irani AM, Schwartz LB, et al. (2000) Increased numbers of both airway basophils and mast cells in sputum after allergen inhalation challenge of atopic asthmatics. Am J Respir Crit Care Med 161: 1473–1478.
    1. Matsumoto K, Gauvreau GM, Rerecich T, Watson RM, Wood LJ, et al. (2002) IL-10 production in circulating T cells differs between allergen-induced isolated early and dual asthmatic responders. J Allergy Clin Immunol 109: 281–286.
    1. Singh A, Freue GV, Oosthuizen JL, Kam SH, Ruan J, et al... (2012) Plasma proteomics can discriminate isolated early from dual responses in asthmatic individuals undergoing an allergen inhalation challenge. Proteomics Clin Appl.
    1. Hankinson JL, Odencrantz JR, Fedan KB (1999) Spirometric reference values from a sample of the general U.S. population. Am J Respir Crit Care Med 159: 179–187.
    1. Cockcroft D, Murdock K, Mink J (1983) Determination of histamine PC20: Comparison of linear and logarithmic interpolation. Chest 84: 505–2.
    1. Gauvreau GM, Boulet LP, Cockcroft DW, Fitzgerald JM, Carlsten C, et al. (2011) Effects of interleukin-13 blockade on allergen-induced airway responses in mild atopic asthma. Am J Respir Crit Care Med 183: 1007–1014.
    1. Gautier L, Cope L, Bolstad BM, Irizarry RA (2004) Affy–analysis of affymetrix GeneChip data at the probe level. Bioinformatics 20: 307–315.
    1. Hochreiter S, Clevert DA, Obermayer K (2006) A new summarization method for affymetrix probe level data. Bioinformatics 22: 943–949.
    1. Talloen W, Clevert DA, Hochreiter S, Amaratunga D, Bijnens L, et al. (2007) I/NI-calls for the exclusion of non-informative genes: A highly effective filtering tool for microarray data. Bioinformatics 23: 2897–2902.
    1. Bourgon R, Gentleman R, Huber W (2010) Independent filtering increases detection power for high-throughput experiments. PNAS 107: 9546–6.
    1. Team RDC (2009) R: A language and environment for statistical computing.
    1. Folch J, Lees M, Sloane Stanley GH (1957) A simple method for the isolation and purification of total lipides from animal tissues. J Biol Chem 226: 497–509.
    1. Schlenk H, Gellerman JL (1960) Esterification of fatty acids with diazomethane on a small scale. Anal. Chem. 32: 1412–3.
    1. Symth GK (2004) Linear models and empirical bayes methods for assessing differential expression in microarrays experiments. Statistical Applications in Genetics and Molecular Biology Article 3.
    1. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: A practical and powerful approach to multiple testing. 57: 289–12.
    1. Huber PJ (1981) Robust statistics. Wiley.
    1. Hotelling H (1936) Relations between two sets of variates. Biometrika 28: 321–57.
    1. Gonzalez I, Dejean S, Martin PGP, Goncalves O, Besse P, et al. (2009) Highlight relationships between heterogeneous biological data through graphical displays based on regularized canonical correlation analysis. Journal of Biological Systems 17: 173–27.
    1. Lattka E, Illig T, Koletzko B, Heinrich J (2010) Genetic variants of the FADS1 FADS2 gene cluster as related to essential fatty acid metabolism. Curr Opin Lipidol 21: 64–69.
    1. Serhan CN, Chiang N, Van Dyke TE (2008) Resolving inflammation: Dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol 8: 349–361.
    1. Feltenmark S, Gautam N, Brunnstrom A, Griffiths W, Backman L, et al. (2008) Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells. Proc Natl Acad Sci U S A 105: 680–685.
    1. Levy BD (2010) Resolvins and protectins: Natural pharmacophores for resolution biology. Prostaglandins Leukot Essent Fatty Acids 82: 327–332.
    1. Shindou H, Hishikawa D, Nakanishi H, Harayama T, Ishii S, et al. (2007) A single enzyme catalyzes both platelet-activating factor production and membrane biogenesis of inflammatory cells. cloning and characterization of acetyl-CoA:LYSO-PAF acetyltransferase. J Biol Chem 282: 6532–6539.
    1. Wenzel SE (2012) Asthma phenotypes: The evolution from clinical to molecular approaches. Nat Med 18: 716–725.
    1. Hamann J, Koning N, Pouwels W, Ulfman LH, van Eijk M, et al. (2007) EMR1, the human homolog of F4/80, is an eosinophil-specific receptor. Eur J Immunol 37: 2797–2802.
    1. Seeds MC, Peachman KK, Bowton DL, Sivertson KL, Chilton FH (2009) Regulation of arachidonate remodeling enzymes impacts eosinophil survival during allergic asthma. Am J Respir Cell Mol Biol 41: 358–366.
    1. Rzehak P, Thijs C, Standl M, Mommers M, Glaser C, et al. (2010) Variants of the FADS1 FADS2 gene cluster, blood levels of polyunsaturated fatty acids and eczema in children within the first 2 years of life. PLoS One 5: e13261.
    1. Chiang N, Arita M, Serhan CN (2005) Anti-inflammatory circuitry: Lipoxin, aspirin-triggered lipoxins and their receptor ALX. Prostaglandins Leukot Essent Fatty Acids 73: 163–177.
    1. Levy BD, Kohli P, Gotlinger K, Haworth O, Hong S, et al. (2007) Protectin D1 is generated in asthma and dampens airway inflammation and hyperresponsiveness. J Immunol 178: 496–502.
    1. Perogamvros I, Ray DW, Trainer PJ (2012) Regulation of cortisol bioavailability–effects on hormone measurement and action. Nat Rev Endocrinol 8: 717–727.
    1. Barnes PJ (1992) Bradykinin and asthma. Thorax 47: 979–983.
    1. Levy BD, Vachier I, Serhan CN (2012) Resolution of inflammation in asthma. Clin Chest Med 33: 559–570.
    1. Hammad H, Lambrecht BN (2008) Dendritic cells and epithelial cells: Linking innate and adaptive immunity in asthma. Nat Rev Immunol 8: 193–204.
    1. Hatzivlassiou M, Grainge C, Kehagia V, Lau L, Howarth PH (2010) The allergen specificity of the late asthmatic reaction. Allergy 65: 355–358.

Source: PubMed

3
Subscribe