Network analysis of quantitative proteomics on asthmatic bronchi: effects of inhaled glucocorticoid treatment

Serena E O'Neil, Brigita Sitkauskiene, Agne Babusyte, Algirda Krisiukeniene, Kristina Stravinskaite-Bieksiene, Raimundas Sakalauskas, Carina Sihlbom, Linda Ekerljung, Elisabet Carlsohn, Jan Lötvall, Serena E O'Neil, Brigita Sitkauskiene, Agne Babusyte, Algirda Krisiukeniene, Kristina Stravinskaite-Bieksiene, Raimundas Sakalauskas, Carina Sihlbom, Linda Ekerljung, Elisabet Carlsohn, Jan Lötvall

Abstract

Background: Proteomic studies of respiratory disorders have the potential to identify protein biomarkers for diagnosis and disease monitoring. Utilisation of sensitive quantitative proteomic methods creates opportunities to determine individual patient proteomes. The aim of the current study was to determine if quantitative proteomics of bronchial biopsies from asthmatics can distinguish relevant biological functions and whether inhaled glucocorticoid treatment affects these functions.

Methods: Endobronchial biopsies were taken from untreated asthmatic patients (n = 12) and healthy controls (n = 3). Asthmatic patients were randomised to double blind treatment with either placebo or budesonide (800 μg daily for 3 months) and new biopsies were obtained. Proteins extracted from the biopsies were digested and analysed using isobaric tags for relative and absolute quantitation combined with a nanoLC-LTQ Orbitrap mass spectrometer. Spectra obtained were used to identify and quantify proteins. Pathways analysis was performed using Ingenuity Pathway Analysis to identify significant biological pathways in asthma and determine how the expression of these pathways was changed by treatment.

Results: More than 1800 proteins were identified and quantified in the bronchial biopsies of subjects. The pathway analysis revealed acute phase response signalling, cell-to-cell signalling and tissue development associations with proteins expressed in asthmatics compared to controls. The functions and pathways associated with placebo and budesonide treatment showed distinct differences, including the decreased association with acute phase proteins as a result of budesonide treatment compared to placebo.

Conclusions: Proteomic analysis of bronchial biopsy material can be used to identify and quantify proteins using highly sensitive technologies, without the need for pooling of samples from several patients. Distinct pathophysiological features of asthma can be identified using this approach and the expression of these features is changed by inhaled glucocorticoid treatment. Quantitative proteomics may be applied to identify mechanisms of disease that may assist in the accurate and timely diagnosis of asthma.

Trial registration: ClinicalTrials.gov registration NCT01378039.

Figures

Figure 1
Figure 1
Schematic overview of the experimental workflow. The workflow used in the proteomics analysis of human endobronchial biopsies. Proteins were extracted and digested, before the peptides were labelled with iTRAQ® Reagents in a four-plex set. Each four-plex, containing one reference pool and three samples, were fractionated using ion exchange chromatography. Each fraction was then subjected to nano LC-MS/MS. The resulting spectra were searched for identification and quantification. The identified and quantified proteins were then analysed using Ingenuity Pathways Analysis.
Figure 2
Figure 2
FENO and FEV1% predicted of healthy controls and asthmatics. The scatter plots show the airway inflammation and lung function of healthy controls (n = 3) and untreated asthmatics (n = 12) as assessed by FEV1% predicted (A) and fractional exhaled nitric oxide (FENO) (B). Asthmatics had significantly increased airway inflammation and significantly decreased lung function, compared to healthy controls. The bars indicate the mean ± SEM. * = indicates p value < 0.05.
Figure 3
Figure 3
Network of differentially expressed proteins in untreated asthmatics compared to healthy controls. The top network generated by Ingenuity Pathways Analysis from quantified proteins (1.5 fold change threshold) of untreated asthmatics compared to healthy controls, contains the functions of haematological system development and function, lipid metabolism and molecular transport. Molecules with bold outline are significantly different between the two groups using a Student's t-test. Lines indicate relationships between molecules. Arrows at the end of these lines indicated the direction of the interaction. Molecules that are up-regulated and down-regulated in the dataset are coloured red and green respectively. Grey molecules do not meet the cut-off threshold. Uncoloured molecules have been added from the Ingenuity Knowledge Base. Entrez gene names for molecules are: ACAT1 = acetyl-CoA acetyltransferase 1, APOA1 = apolipoprotein A-I, APOA2 = apolipoprotein A-II, CA2 = carbonic anhydrase II, CAP1 = CAP, adenylate cyclase-associated protein 1 (yeast), CS = citrate synthase, GLO1 = glyoxalase I, GOLGA3 = golgin A3, HBB = hemoglobin (beta), HBD = hemoglobin (delta), HBG1 = hemoglobin (gamma A), HLA-B = major histocompatibility complex, class I, B, HNRNPH1 = heterogeneous nuclear ribonucleoprotein H1 (H), MVP = major vault protein, MYLK = myosin light chain kinase, NONO = non-POU domain containing, octamer-binding, PAK2 = p21 protein (Cdc42/Rac)-activated kinase 2, RAB14 = RAB14, member RAS oncogene family, RPL8 = ribosomal protein L8, SYNCRIP = synaptotagmin binding, cytoplasmic RNA interacting protein, TAGLN2 = transgelin 2, TARDBP = TAR DNA binding protein, TNS1 = tensin 1, TPM3 = tropomyosin 3, UQCRB = ubiquinol-cytochrome c reductase binding protein, UQCRC2 = ubiquinol-cytochrome c reductase core protein II, VASP = vasodilator-stimulated phosphoprotein.
Figure 4
Figure 4
Network of differentially expressed proteins of post placebo compared to paired pre placebo biopsy. The top network generated by Ingenuity Pathways Analysis from quantified proteins of post placebo asthmatics compared to their pre placebo biopsy. The network contains the functions of hair and skin development and function, organ development, dermatological diseases and conditions. Molecules with bold outline are significantly different between the two groups using a Student's t-test. Lines indicate relationships between molecules. Arrows at the end of these lines indicated the direction of the interaction. Molecules that are up-regulated and down-regulated in the dataset are coloured red and green respectively. Grey molecules do not meet the cut-off threshold. Uncoloured molecules have been added from the IPA KB. Entrez gene names for molecules are: CFL1 = cofilin 1 (non-muscle) DES = desmin, E2F7 = E2F transcription factor 7, HDGF = hepatoma-derived growth factor, HRG = histidine-rich glycoprotein, IGHG3 = immunoglobulin heavy constant gamma 3 (G3 m marker), IGHG4 = immunoglobulin heavy constant gamma 4 (G4 m marker), IGKC = immunoglobulin kappa constant, KRT2 = keratin 2, KRT4 = keratin 4, KRT5 = keratin 5, KRT8 = keratin 8, KRT9 = keratin 9, KRT13 = keratin 13, KRT14 = keratin 14, KRT15 = keratin 15, KRT17 = keratin 17, KRT19 = keratin 19, KRT6A = keratin 6A, PPIF = peptidylprolyl isomerase F, PPL = periplakin, S100P = S100 calcium binding protein P, SERPINA4 = serpin peptidase inhibitor, clade A (alpha-1 antiproteinase, antitrypsin), member 4, TACSTD2 = tumor-associated calcium signal transducer 2, TMOD2 = tropomodulin 2 (neuronal), TNFSF13B = tumor necrosis factor (ligand) superfamily, member 13b, TPI1 = triosephosphate isomerase 1, TPM1 = tropomyosin 1 (alpha).
Figure 5
Figure 5
Network of differentially expressed proteins of post budesonide compared to paired pre treatment biopsy. The top network generated by Ingenuity Pathways Analysis from quantified proteins of post budesonide asthmatics compared to their pre placebo biopsy. The network contains the functions of cancer, genetic disorder and respiratory disease. Molecules with bold outline are significantly different between the two groups using a Student's t-test. Lines indicate relationships between molecules. Arrows at the end of these lines indicated the direction of the interaction. Molecules that are up-regulated and down-regulated in the dataset are coloured red and green respectively. Grey molecules do not meet the cut-off threshold. Uncoloured molecules have been added from the IPA KB. Entrez gene names for molecules are: CAPNS1 = calpain, small subunit 1, CAST = calpastatin, CFL1 = cofilin 1 (non-muscle), CKB = creatine kinase, brain, DPYSL2 = dihydropyrimidinase-like 2, DPYSL3 = dihydropyrimidinase-like 3, DPYSL5 = dihydropyrimidinase-like 5, DYNLL2 = dynein, light chain, LC8-type 2, ENO1 = enolase 1, (alpha), GPI = glucose-6-phosphate isomerase, H2AFX = H2A histone family, member X, HNRNPD = heterogeneous nuclear ribonucleoprotein D (AU-rich element RNA binding protein 1, 37 kDa), HYOU1 = hypoxia up-regulated 1, ILF3 = interleukin enhancer binding factor 3, 90 kDa, KRT4 = keratin 4, KRT9 = keratin 9, KRT6A = keratin 6A, LDHA = lactate dehydrogenase A, PPP1CC = protein phosphatase 1, catalytic subunit, gamma isozyme, RPL31 = ribosomal protein L31, SERBP1 = SERPINE1 mRNA binding protein 1, SYNCRIP = synaptotagmin binding, cytoplasmic RNA interacting protein, TTR = transthyretin, VIM = vimentin, XRCC5 = X-ray repair complementing defective repair in Chinese hamster cells 5 (double-strand-break rejoining), YWHAE = tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, epsilon polypeptide, YWHAZ = tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide.

References

    1. Lemanske RF Jr, Busse WW. Asthma: clinical expression and molecular mechanisms. Journal of Allergy and Clinical Immunology. 2010;125:S95–102. doi: 10.1016/j.jaci.2009.10.047.
    1. Masoli M, Fabian D, Holt S, Beasley R. The global burden of asthma: executive summary of the GINA Dissemination Committee report. Allergy. 2004;59:469–478. doi: 10.1111/j.1398-9995.2004.00526.x.
    1. Rolph MS, Sisavanh M, Liu SM, Mackay CR. Clues to asthma pathogenesis from microarray expression studies. Pharmacology and Therapeutics. 2006;109:284–294. doi: 10.1016/j.pharmthera.2005.08.009.
    1. Greenlee KJ, Corry DB, Engler DA, Matsunami RK, Tessier P, Cook RG, Werb Z, Kheradmand F. Proteomic identification of in vivo substrates for matrix metalloproteinases 2 and 9 reveals a mechanism for resolution of inflammation. Journal of Immunology. 2006;177:7312–7321.
    1. Jeong H, Rhim T, Ahn MH, Yoon PO, Kim SH, Chung IY, Uh S, Kim SI, Park CS. Proteomic analysis of differently expressed proteins in a mouse model for allergic asthma. J Korean Med Sci. 2005;20:579–585. doi: 10.3346/jkms.2005.20.4.579.
    1. Wong WS, Zhao J. Proteome analysis of chronically inflamed lungs in a mouse chronic asthma model. International Archives of Allergy and Immunology. 2008;147:179–189. doi: 10.1159/000142040.
    1. Zhang L, Wang M, Kang X, Boontheung P, Li N, Nel AE, Loo JA. Oxidative stress and asthma: proteome analysis of chitinase-like proteins and FIZZ1 in lung tissue and bronchoalveolar lavage fluid. Journal of Proteome Research. 2009;8:1631–1638. doi: 10.1021/pr800685h.
    1. Larsen K, Malmstrom J, Wildt M, Dahlqvist C, Hansson L, Marko-Varga G, Bjermer L, Scheja A, Westergren-Thorsson G. Functional and phenotypical comparison of myofibroblasts derived from biopsies and bronchoalveolar lavage in mild asthma and scleroderma. Respir Res. 2006;7:11. doi: 10.1186/1465-9921-7-11.
    1. Gray RD, MacGregor G, Noble D, Imrie M, Dewar M, Boyd AC, Innes JA, Porteous DJ, Greening AP. Sputum proteomics in inflammatory and suppurative respiratory diseases. American Journal of Respiratory and Critical Care Medicine. 2008;178:444–452. doi: 10.1164/rccm.200703-409OC.
    1. Wu J, Kobayashi M, Sousa EA, Liu W, Cai J, Goldman SJ, Dorner AJ, Projan SJ, Kavuru MS, Qiu Y, Thomassen MJ. Differential proteomic analysis of bronchoalveolar lavage fluid in asthmatics following segmental antigen challenge. Molecular and Cellular Proteomics. 2005;4:1251–1264. doi: 10.1074/mcp.M500041-MCP200.
    1. Jeong HC, Lee SY, Lee EJ, Jung KH, Kang EH, Kim JH, Park EK, Lee SH, Uhm CS, Cho Y. et al.Proteomic analysis of peripheral T-lymphocytes in patients with asthma. Chest. 2007;132:489–496. doi: 10.1378/chest.06-2980.
    1. Bloemen K, Van Den Heuvel R, Govarts E, Hooyberghs J, Nelen V, Witters E, Desager K, Schoeters G. A new approach to study exhaled proteins as potential biomarkers for asthma. Clin Exp Allergy. 2011;41:346–356. doi: 10.1111/j.1365-2222.2010.03638.x.
    1. Cho WC. Proteomics technologies and challenges. Genomics Proteomics Bioinformatics. 2007;5:77–85. doi: 10.1016/S1672-0229(07)60018-7.
    1. Romero R, Kusanovic JP, Gotsch F, Erez O, Vaisbuch E, Mazaki-Tovi S, Moser A, Tam S, Leszyk J, Master SR. et al.Isobaric labeling and tandem mass spectrometry: a novel approach for profiling and quantifying proteins differentially expressed in amniotic fluid in preterm labor with and without intra-amniotic infection/inflammation. Journal of Maternal-Fetal and Neonatal Medicine. 2010;23:261–280. doi: 10.3109/14767050903067386.
    1. Wang H, Chavali S, Mobini R, Muraro A, Barbon F, Boldrin D, Aberg N, Benson M. A pathway-based approach to find novel markers of local glucocorticoid treatment in intermittent allergic rhinitis. Allergy. 2011;66:132–140. doi: 10.1111/j.1398-9995.2010.02444.x.
    1. Chaerkady R, Letzen B, Renuse S, Sahasrabuddhe NA, Kumar P, All AH, Thakor NV, Delanghe B, Gearhart JD, Pandey A, Kerr CL. Quantitative temporal proteomic analysis of human embryonic stem cell differentiation into oligodendrocyte progenitor cells. Proteomics. in press .
    1. Dean RA, Overall CM. Proteomics discovery of metalloproteinase substrates in the cellular context by iTRAQ labeling reveals a diverse MMP-2 substrate degradome. Mol Cell Proteomics. 2007;6:611–623. doi: 10.1074/mcp.M600341-MCP200.
    1. Neilson KA, Mariani M, Haynes PA. Quantitative proteomic analysis of cold-responsive proteins in rice. Proteomics. 2011;11:1696–1706.
    1. Global Strategy for Asthma Management and Prevention. NIH publication No 02-3659 Global Initiative for Asthma National Institutes of Health National Heart, Lung and Blood Institute. 2002.
    1. Carlsohn E, Nystrom J, Karlsson H, Svennerholm AM, Nilsson CL. Characterization of the outer membrane protein profile from disease-related Helicobacter pylori isolates by subcellular fractionation and nano-LC FT-ICR MS analysis. Journal of Proteome Research. 2006;5:3197–3204. doi: 10.1021/pr060181p.
    1. Thomas PD, Campbell MJ, Kejariwal A, Mi H, Karlak B, Daverman R, Diemer K, Muruganujan A, Narechania A. PANTHER: a library of protein families and subfamilies indexed by function. Genome Research. 2003;13:2129–2141. doi: 10.1101/gr.772403.
    1. Thomas PD, Kejariwal A, Guo N, Mi H, Campbell MJ, Muruganujan A, Lazareva-Ulitsky B. Applications for protein sequence-function evolution data: mRNA/protein expression analysis and coding SNP scoring tools. Nucleic Acids Research. 2006;34:W645–650. doi: 10.1093/nar/gkl229.
    1. Saeed AI, Bhagabati NK, Braisted JC, Liang W, Sharov V, Howe EA, Li J, Thiagarajan M, White JA, Quackenbush J. TM4 microarray software suite. Methods Enzymol. 2006;411:134–193.
    1. Saeed AI, Sharov V, White J, Li J, Liang W, Bhagabati N, Braisted J, Klapa M, Currier T, Thiagarajan M. et al.TM4: a free, open-source system for microarray data management and analysis. Biotechniques. 2003;34:374–378.
    1. Watanabe Y, Hashimoto Y, Shiratsuchi A, Takizawa T, Nakanishi Y. Augmentation of fatality of influenza in mice by inhibition of phagocytosis. Biochem Biophys Res Commun. 2005;337:881–886. doi: 10.1016/j.bbrc.2005.09.133.
    1. Altraja S, Jaama J, Valk E, Altraja A. Changes in the proteome of human bronchial epithelial cells following stimulation with leucotriene E4 and transforming growth factor-beta1. Respirology. 2009;14:39–45. doi: 10.1111/j.1440-1843.2008.01414.x.
    1. Waldburg N, Kahne T, Reisenauer A, Rocken C, Welte T, Buhling F. Clinical proteomics in lung diseases. Pathol Res Pract. 2004;200:147–154. doi: 10.1016/j.prp.2004.02.006.
    1. Sherry B, Yarlett N, Strupp A, Cerami A. Identification of cyclophilin as a proinflammatory secretory product of lipopolysaccharide-activated macrophages. Proc Natl Acad Sci USA. 1992;89:3511–3515. doi: 10.1073/pnas.89.8.3511.
    1. van Diepen A, Brand HK, Sama I, Lambooy LH, van den Heuvel LP, van der Well L, Huynen M, Osterhaus AD, Andeweg AC, Hermans PW. Quantitative proteome profiling of respiratory virus-infected lung epithelial cells. J Proteomics. 2010;73:1680–1693. doi: 10.1016/j.jprot.2010.04.008.
    1. Lakind JS, Holgate ST, Ownby DR, Mansur AH, Helms PJ, Pyatt D, Hays SM. A critical review of the use of Clara cell secretory protein (CC16) as a biomarker of acute or chronic pulmonary effects. Biomarkers. 2007;12:445–467. doi: 10.1080/13547500701359327.
    1. Kang JS, Yoon YD, Ahn JH, Kim SC, Kim KH, Kim HM, Moon EY. B cell-activating factor is a novel diagnosis parameter for asthma. International Archives of Allergy and Immunology. 2006;141:181–188. doi: 10.1159/000094897.
    1. Sutherland AP, Ng LG, Fletcher CA, Shum B, Newton RA, Grey ST, Rolph MS, Mackay F, Mackay CR. BAFF augments certain Th1-associated inflammatory responses. Journal of Immunology. 2005;174:5537–5544.
    1. Voisin SN, Krakovska O, Matta A, Desouza LV, Romaschin AD, Colgan TJ, Siu KW. Identification of Novel Molecular Targets for Endometrial Cancer Using a Drill-Down LC-MS/MS Approach with iTRAQ. PLoS ONE. 2011;6:e16352. doi: 10.1371/journal.pone.0016352.
    1. Nishimura T, Nomura M, Tojo H, Hamasaki H, Fukuda T, Fujii K, Mikami S, Bando Y, Kato H. Proteomic analysis of laser-microdissected paraffin-embedded tissues: (2) MRM assay for stage-related proteins upon non-metastatic lung adenocarcinoma. J Proteomics. 2010;73:1100–1110. doi: 10.1016/j.jprot.2009.11.010.
    1. Kiesler P, Haynes PA, Shi L, Kao PN, Wysocki VH, Vercelli D. NF45 and NF90 regulate HS4-dependent interleukin-13 transcription in T cells. Journal of Biological Chemistry. 2010;285:8256–8267. doi: 10.1074/jbc.M109.041004.
    1. Hackett TL, Warner SM, Stefanowicz D, Shaheen F, Pechkovsky DV, Murray LA, Argentieri R, Kicic A, Stick SM, Bai TR, Knight DA. Induction of epithelial-mesenchymal transition in primary airway epithelial cells from patients with asthma by transforming growth factor-beta1. American Journal of Respiratory and Critical Care Medicine. 2009;180:122–133. doi: 10.1164/rccm.200811-1730OC.
    1. Johnson JR, Roos A, Berg T, Nord M, Fuxe J. Chronic respiratory aeroallergen exposure in mice induces epithelial-mesenchymal transition in the large airways. PLoS ONE. 2011;6:e16175. doi: 10.1371/journal.pone.0016175.
    1. Hoshino M, Nakagawa T, Sano Y, Hirai K. Effect of inhaled corticosteroid on an immunoreactive thymus and activation-regulated chemokine expression in the bronchial biopsies from asthmatics. Allergy. 2005;60:317–322. doi: 10.1111/j.1398-9995.2005.00694.x.
    1. Laprise C, Sladek R, Ponton A, Bernier MC, Hudson TJ, Laviolette M. Functional classes of bronchial mucosa genes that are differentially expressed in asthma. BMC Genomics. 2004;5:21. doi: 10.1186/1471-2164-5-21.
    1. Lotvall J, Akdis CA, Bacharier LB, Bjermer L, Casale TB, Custovic A, Lemanske RF Jr, Wardlaw AJ, Wenzel SE, Greenberger PA. Asthma endotypes: a new approach to classification of disease entities within the asthma syndrome. Journal of Allergy and Clinical Immunology. 2011;127:355–360. doi: 10.1016/j.jaci.2010.11.037.
    1. Haldar P, Pavord ID, Shaw DE, Berry MA, Thomas M, Brightling CE, Wardlaw AJ, Green RH. Cluster analysis and clinical asthma phenotypes. Am J Respir Crit Care Med. 2008;178:218–224. doi: 10.1164/rccm.200711-1754OC.
    1. Berry M, Morgan A, Shaw DE, Parker D, Green R, Brightling C, Bradding P, Wardlaw AJ, Pavord ID. Pathological features and inhaled corticosteroid response of eosinophilic and non-eosinophilic asthma. Thorax. 2007;62:1043–1049. doi: 10.1136/thx.2006.073429.
    1. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nature Reviews Immunology. 2008;8:183–192. doi: 10.1038/nri2254.

Source: PubMed

3
購読する