1,311
Views
21
CrossRef citations to date
0
Altmetric
Review

Transcriptomics, proteomics and metabolomics driven biomarker discovery in COPD: an update

, , , , &
Pages 897-913 | Received 06 Feb 2016, Accepted 02 Jun 2016, Published online: 20 Jun 2016

References

  • Global Initiative for Chronic Obstructive Lung Disease. Global strategy for the diagnosis, management, and prevention of COPD. [updated 2010; cited 2016 Jan 10]. Available from: www.goldcopd.org/uploads/users/files/GOLDReport_April112011.pdf
  • World Health Organization. Chronic respiratory diseases. Burden of COPD. [cited 2016 Jan 10]. Available from: www.who.int/respiratory/copd/burden/en/
  • Csikesz NG, Gartman EJ. New developments in the assessment of COPD: early diagnosis is key. Int J Chron Obstruct Pulmon Dis. 2014;9:277–286.
  • Chen H, Wang X. Significance of bioinformatics in research of chronic obstructive pulmonary disease. J Clin Bioinforma. 2011;1:35.
  • Caudle WM, Bammler TK, Lin Y, et al. Using ‘omics’ to define pathogenesis and biomarkers of Parkinson’s disease. Expert Rev Neurother. 2010;10:925–942.
  • Janvilisri T. Omics-based identification of biomarkers for nasopharyngeal carcinoma. Dis Markers. 2015;2015:762128.
  • Baggerman G, Vierstraete E, De Loof A, et al. Gel-based versus gel-free proteomics: a review. Comb Chem High Throughput Screen. 2005;8:669–677.
  • Griffiths WJ. Metabolomics, metabonomics and metabolite profiling. Cambridge: RSC Publishing; 2008.
  • Pan Z, Raftery D. Comparing and combining NMR spectroscopy and mass spectrometry in metabolomics. Anal Bioanal Chem. 2007;387:525–527.
  • Soininen P, Kangas AJ, Würtz P, et al. High-throughput serum NMR metabonomics for cost-effective holistic studies on systemic metabolism. Analyst. 2009;134:1781–1785.
  • Wang Z, Gerstein M, Snyder M. RNA-seq: a revolutionary tool for transcriptomics. Nat Rev Genet. 2009;10:57–63.
  • Comandini A, Rogliani P, Nunziata A, et al. Biomarkers of lung damage associated with tobacco smoke in induced sputum. Respir Med. 2009;103:1592–1613.
  • Pinto-Plata V, Toso J, Lee K, et al. Use of proteomic patterns of serum biomarkers in patients with chronic obstructive pulmonary disease: correlation with clinical parameters. Proc Am Thorac Soc. 2006;3:465–466.
  • Wheelock CE, Goss VM, Balgoma D, et al. Application of ’omics technologies to biomarker discovery in inflammatory lung diseases. Eur Respir J. 2013;42:802–825.
  • Scherp P, Ku G, Coleman L, et al. Gel-based and gel-free proteomic technologies. Methods Mol Biol. 2011;702:163–190.
  • Frantzi M, Bhat A, Latosinska A. Clinical proteomic biomarkers: relevant issues on study design & technical considerations in biomarker development. Clin Transl Med. 2014;3:7.
  • Chandramouli K, Qian P-Y., et al. Proteomics: challenges, techniques and possibilities to overcome biological sample complexity. Hum Genomics Proteomics. 2009;2009:1–22.
  • Verrills NM, Irwin JA, He XY, et al. Identification of novel diagnostic biomarkers for asthma and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2011;183:1633–1643.
  • Rana GSJB, York TP, Edmiston JS, et al. Proteomic biomarkers in plasma that differentiate rapid and slow decline in lung function in adult cigarette smokers with chronic obstructive pulmonary disease (COPD). Anal Bioanal Chem. 2010;397:1809–1819.
  • Merali S, Barrero CA, Bowler RP, et al. Analysis of the plasma proteome in COPD: novel low abundance proteins reflect the severity of lung remodeling. Copd. 2014;11:177–189.
  • Nicholas B, Djukanović R. Induced sputum: a window to lung pathology. Biochem Soc Trans. 2009;37:868–872.
  • Pelaia G, Terracciano R, Vatrella A, et al. Application of proteomics and peptidomics to COPD. Biomed Res Int. 2014;2014:1–8.
  • Pastor MD, Nogal A, Molina-Pinelo S, et al. Identification of proteomic signatures associated with lung cancer and COPD. J Proteomics. 2013;89:227–237.
  • Nicholas BL, Skipp P, Barton S, et al. Identification of lipocalin and apolipoprotein A1 as biomarkers of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181:1049–1060.
  • Ohlmeier S, Mazur W, Linja-Aho A, et al. Sputum proteomics identifies elevated PIGR levels in smokers and mild-to-moderate COPD. J Proteome Res. 2012;11:599–608.
  • Gao J, Ohlmeier S, Nieminen P, et al. Elevated sputum BPIFB1 levels in smokers with chronic obstructive pulmonary disease: a longitudinal study. Am J Physiol Lung Cell Mol Physiol. 2015;309:L17–L26.
  • Terracciano R, Preianò M, Palladino GP, et al. Peptidome profiling of induced sputum by mesoporous silica beads and MALDI-TOF MS for non-invasive biomarker discovery of chronic inflammatory lung diseases. Proteomics. 2011;11:3402–3414.
  • Titz B, Sewer A, Schneider T, et al. Alterations in the sputum proteome and transcriptome in smokers and early-stage COPD subjects. J Proteomics. 2015;128:306–320.
  • Baraniuk JN, Casado B, Pannell LK, et al. Protein networks in induced sputum from smokers and COPD patients. Int J Chron Obstruct Pulmon Dis. 2015;10:1957–1975.
  • Mutlu GM, Garey KW, Robbins RA, et al. Collection and analysis of exhaled breath condensate in humans. Am J Respir Crit Care Med. 2001;164:731–737.
  • Griese M, Noss J, von Bredow C. Protein pattern of exhaled breath condensate and saliva. Proteomics. 2002;2:690–696.
  • Casado B, Luisetti M, Iadarola P, et al. Advances in proteomic techniques for biomarker discovery in COPD. Expert Rev Clin Immunol. 2011;7:111–123.
  • Bowler RP, Ellison MC, Reisdorph N. Proteomics in pulmonary medicine. Chest. 2006;130:567–574.
  • Fumagalli M, Ferrari F, Luisetti M, et al. Profiling the proteome of exhaled breath condensate in healthy smokers and COPD patients by LC-MS/MS. Int J Mol Sci. 2012;13:13894–13910.
  • Bartlett JA, Albertolle ME, Wohlford-Lenane C, et al. Protein composition of bronchoalveolar lavage fluid and airway surface liquid from newborn pigs. Am J Physiol Lung Cell Mol Physiol. 2013;305:L256–L266.
  • Kipnis E, Hansen K, Sawa T, et al. Proteomic analysis of undiluted lung epithelial lining fluid. Chest. 2008;134:338–345.
  • Merkel D, Rist W, Seither P, et al. Proteomic study of human bronchoalveolar lavage fluids from smokers with chronic obstructive pulmonary disease by combining surface-enhanced laser desorption/ionization-mass spectrometry profiling with mass spectrometric protein identification. Proteomics. 2005;5:2972–2980.
  • Terracciano R, Pelaia G, Preianò M, et al. Asthma and COPD proteomics: current approaches and future directions. Proteomics Clin Appl. 2015;9:203–220.
  • Franciosi L, Govorukhina N, Fusetti F, et al. Proteomic analysis of human epithelial lining fluid by microfluidics-based nanoLC-MS/MS: a feasibility study. Electrophoresis. 2013;34:2683–2694.
  • Franciosi L, Postma DS, van den Berge M, et al. Susceptibility to COPD: differential proteomic profiling after acute smoking. PLoS One. 2014;9:e102037.
  • Magi B, Bargagli E, Bini L, et al. Proteome analysis of bronchoalveolar lavage in lung diseases. Proteomics. 2006;6:6354–6369.
  • Savino R, Paduano S, Preianò M, et al. The proteomics big challenge for biomarkers and new drug-targets discovery. Int J Mol Sci. 2012;13:13926–13948.
  • Tu C, Mammen MJ, Li J, et al. Large-scale, ion-current-based proteomics investigation of bronchoalveolar lavage fluid in chronic obstructive pulmonary disease patients. J Proteome Res. 2014;13:627−629.
  • Chen H, Wang D, Bai C, et al. Proteomics-based biomarkers in chronic obstructive pulmonary disease. J Proteome Res. 2010;9:2798–2808.
  • Kelsen SG, Duan X, Ji R, et al. Cigarette smoke induces an unfolded protein response in the human lung: a proteomic approach. Am J Respir Cell Mol Biol. 2008;38:541–550.
  • Ohlmeier S, Vuolanto M, Toljamo T, et al. Proteomics of human lung tissue identifies surfactant protein A as a marker of chronic obstructive pulmonary disease. J Proteome Res. 2008;7:5125–5132.
  • Lee EJ, In KH, Kim JH, et al. Proteomic analysis in lung tissue of smokers and COPD patients. Chest. 2009;135:344–352.
  • Alexandre BM, Charro N, Blonder J, et al. Profiling the erythrocyte membrane proteome isolated from patients diagnosed with chronic obstructive pulmonary disease. J Proteomics. 2012;76:259–269.
  • Ishikawa N, Ohlmeier S, Salmenkivi K, et al. Hemoglobin α and β are ubiquitous in the human lung, decline in idiopathic pulmonary fibrosis but not in COPD. Respir Res. 2010;11:123.
  • Ohlmeier S, Nieminen P, Gao J, et al. Lung tissue proteomics identifies elevated transglutaminase 2 levels in stable chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol. 2016;310:L1155-L1165. doi:10.1152/ajplung.00021.2016.
  • Nicholson JK, Lindon JC, Holmes E. ‘Metabonomics’: understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica. 1999;29:1181–1189.
  • Lindon JC, Nicholson JK. Spectroscopic and statistical techniques for information recovery in metabonomics and metabolomics. Annu Rev Anal Chem (Palo Alto Calif). 2008;1:45–69.
  • Nicholson JK, Lindon JC. Systems biology: metabonomics. Nature. 2008;455:1054–1056.
  • Patti GJ, Yanes O, Siuzdak G. Metabolomics: the apogee of the omictriology. Nat Rev Mol Cell Biol. 2012;13:263–269.
  • Laurentiis GD, Paris D, Melck D, et al. Metabonomic analysis of exhaled breath condensate in adults by nuclear magnetic resonance spectroscopy. Eur Respir J. 2008;32:1175–1183.
  • Laurentiis GD, Paris D, Melck D, et al. Separating smoking-related diseases using NMR-based metabolomics of exhaled breath condensate. J Proteome Res. 2013;12:1502–1511.
  • Izquierdo-García JL, Peces-Barba G, Heili S, et al. Is NMR-based metabolomic analysis of exhaled breath condensate accurate? Eur Respir J. 2011;37:468–470.
  • Basanta M, Jarvis RM, Xu Y, et al. Non-invasive metabolomic analysis of breath using differential mobility spectrometry in patients with chronic obstructive pulmonary disease and healthy smokers. Analyst. 2010;135:315–320.
  • Basanta M, Ibrahim B, Dockry R, et al. Exhaled volatile organic compounds for phenotyping chronic obstructive pulmonary disease: a cross-sectional study. Respir Res. 2012;13:72.
  • Fens N, de Nijs SB, Peters S, et al. Exhaled air molecular profiling in relation to inflammatory subtype and activity in COPD. Eur Respir J. 2011;38:1301–1309.
  • McClay JL, Adkins DE, Isern NG, et al. (1)H nuclear magnetic resonance metabolomics analysis identifies novel urinary biomarkers for lung function. J Proteome Res. 2010;9:3083–3090.
  • Wang L, Tang Y, Liu S, et al. Metabonomic profiling of serum and urine by 1H NMR based spectroscopy discriminates patients with chronic obstructive pulmonary disease and healthy individuals. PLoS One. 2013;8:e65675.
  • Adamko DJ, Nair P, Mayers I, et al. Metabolomic profiling of asthma and chronic obstructive pulmonary disease: a pilot study differentiating diseases. J Allergy Clin Immunol. 2015;136:571–580.
  • Ząbek A, Stanimirova I, Deja S, et al. Fusion of the (1)H NMR data of serum, urine and exhaled breath condensate in order to discriminate chronic obstructive pulmonary disease and obstructive sleep apnea syndrome. Metabolomics. 2015;11:1563–1574.
  • Ubhi BK, Riley JH, Shaw PA, et al. Metabolic profiling detects biomarkers of protein degradation in COPD patients. Eur Respir J. 2012;40:345–355.
  • Ubhi BK, Cheng KK, Dong J, et al. Targeted metabolomics identifies perturbations in amino acid metabolism that sub-classify patients with COPD. Mol Biosyst. 2012;8:3125–3133.
  • Deja S, Porebska I, Kowal A, et al. Metabolomics provide new insights on lung cancer staging and discrimination from chronic obstructive pulmonary disease. J Pharm Biomed Anal. 2014;100:369–380.
  • Paige M, Burdick MD, Kim S, et al. Pilot analysis of the plasma metabolite profiles associated with emphysematous chronic obstructive pulmonary disease phenotype. Biochem Biophys Res Commun. 2011;413:588–593.
  • Chen Q, Deeb RS, Ma Y, et al. Serum metabolite biomarkers discriminate healthy smokers from COPD smokers. PLoS One. 2015;10:e0143937.
  • Telenga ED, Hoffmann RF, t’Kindt R, et al. Untargeted lipidomic analysis in chronic obstructive pulmonary disease. Uncovering sphingolipids. Am J Respir Crit Care Med. 2014;190:155–164.
  • Bowler RP, Jacobson S, Cruickshank C, et al. Plasma sphingolipids associated with chronic obstructive pulmonary disease phenotypes. Am J Respir Crit Care Med. 2015;191:275–284.
  • Malone JH, Oliver B. Microarrays, deep sequencing and the true measure of the transcriptome. BMC Biol. 2011;9:34.
  • Chaussabel D, Pascual V, Banchereau J. Assessing the human immune system through blood transcriptomics. BMC Biol. 2010;8:84.
  • Almansa R, Socias L, Sanchez-Garcia M, et al. Critical COPD respiratory illness is linked to increased transcriptomic activity of neutrophil proteases genes. BMC Res Notes. 2012;5:401.
  • Bahr TM, Hughes GJ, Armstrong M, et al. Peripheral blood mononuclear cell gene expression in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2013;49:316–323.
  • Grundy S, Plumb J, Lea S, et al. Down regulation of T cell receptor expression in COPD pulmonary CD8 cells. PLoS One. 2013;8:e71629.
  • Roberts MEP, Higgs BW, Brohawn P, et al. CD4+ T-cell profiles and peripheral blood ex-vivo responses to T-cell directed stimulation delineate COPD phenotypes. Chronic Obstr Pulm Dis (Miami). 2015;2:268–280.
  • Agler AH, Crystal RG, Mezey JG, et al. Differential expression of vitamin E and selenium-responsive genes by disease severity in chronic obstructive pulmonary disease. Copd. 2013;10:450–458.
  • Kim WJ, Lim JH, Lee JS, et al. Comprehensive analysis of transcriptome sequencing data in the lung tissues of COPD subjects. Int J Genomics. 2015;2015:1–9.
  • Faner R, Cruz T, Casserras T, et al. Network analysis of lung transcriptomics reveals a distinct B cell signature in emphysema. Am J Respir Crit Care Med. 2016;193:1242–1253.
  • Singh D, Fox SM, Tal-Singer R, et al. Induced sputum genes associated with spirometric and radiological disease severity in COPD ex-smokers. Thorax. 2011;66:489–495.
  • Kim DK, Cho MH, Hersh CP, et al. Genome-wide association analysis of blood biomarkers in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;186:1238–1247.
  • Skates SJ, Gillette MA, LaBaer J, et al. Statistical design for biospecimen cohort size in proteomics based biomarker discovery and verification studies. J Proteome Res. 2013;12:5383–5394.
  • Diz AP, Truebano M, Skibinski DOF. The consequences of sample pooling in proteomics: an empirical study. Electrophoresis. 2009;30:2967–2975.
  • Kendziorski C, Irizarry RA, Chen K-S, et al. On the utility of pooling biological samples in microarray experiments. Proc Natl Acad Sci USA. 2005;102:4252–4257.
  • Müller P, Parmigiani G, Robert C, et al. Optimal sample size for multiple testing. J Am Stat Assoc. 2004;99:990–100.
  • Tibshirani R. A simple method for assessing sample sizes in microarray experiments. BMC Bioinformatics. 2006;7:106.
  • Liu P, Hwanga JTG. Quick calculation for sample size while controlling false discovery rate with application to microarray analysis. Bioinformatics. 2007;23:739–746.
  • Lin W-J, Hsueh H-M, Chen JJ. Power and sample size estimation in microarray studies. BMC Bioinformatics. 2010;11:48.
  • Nyamundanda G, Gormley IC, Fan Y, et al. MetSizeR: selecting the optimal sample size for metabolomic studies using an analysis based approach. BMC Bioinformatics. 2013;14:338.
  • Xia J, Sinelnikov IV, Han B, et al. MetaboAnalyst 3.0––making metabolomics more meaningful. Nucleic Acids Res. 2015;43:W251–W257.
  • Pan W, Lin J, Le CT. How many replicates of arrays are required to detect gene expression changes in microarray experiments? A mixture model approach. Genome Biol. 2002;3:research0022.
  • Dobbin K, Simon R. Sample size determination in microarray experiments for class comparison and prognostic classification. Biostatistics. 2005;6:27–38.
  • Pawitan Y, Michiels S, Koscielny S, et al. False discovery rate, sensitivity and sample size for microarray studies. Bioinformatics. 2005;21:3017–3024.
  • Yang MC, Yang JJ, McIndoe RA, et al. Microarray experimental design: power and sample size considerations. Physiol Genomics. 2003;16:24–28.
  • Qiu W, Lee MT, Whitmore GA Sizepower: sample size and power calculation in micorarray studies. R package version 1.42.0. [cited 2016 May 15]. Available from: www.bioconductor.org/packages/release/bioc/html/sizepower.html
  • Dionisi HM, Harms G, Layton AC, et al. Power analysis for real-time PCR quantification of genes in activated sludge and analysis of the variability introduced by DNA extraction. Appl Environ Microbiol. 2003;69:6597–6604.
  • Kitchen RR, Kubista M, Tichopad A. Statistical aspects of quantitative real-time PCR experiment design. Methods. 2010;50:231–236.
  • Hashimoto S, Qu W, Ahsan B, et al. High resolution analysis of the 5ʹ-end transcriptome using a next generation DNA sequencer. PLoS One. 2009;4:e4108.
  • Li C-I, Su P-F, Guo Y, et al. Sample size calculation for differential expression analysis of RNA-seq data under poisson distribution. Int J Comput Biol Drug Des. 2013;6:358–375.
  • Marioni JC, Mason CE, Mane SM, et al. Rnaseq: an assessment of technical reproducibility and comparison with gene expression arrays. Genome Res. 2008;18:1509–1517.
  • Wang L, Feng Z, Wang X, et al. DEGseq: an R package for identifying differentially expressed genes from RNA-seq data. Bioinformatics. 2010;26:136–138.
  • Li J, Witten DM, Johnstone IM, et al. Normalization, testing, and false discovery rate estimation for rna-sequencing data. Biostatistics. 2011;14:1–16.
  • Hart SN, Therneau TM, Zhang Y, et al. Calculating sample size estimates for RNA sequencing data. J Comput Biol. 2013;20:970–978.
  • Haznadar M, Maruvada P, Mette E, et al. Navigating the road ahead: addressing challenges for use of metabolomics in epidemiology studies. Metabolomics. 2014;10:176–178.
  • Stringer KA, McKay RT, Karnovsky A, et al. metabolomics and its application to acute lung diseases. Front Immunol. 2016;7:44.
  • Cavailles A, Brinchault-Rabin G, Dixmier A, et al. Comorbidities of COPD. Eur Respir Rev. 2013;22:454–475.
  • Hardin M, Silverman EK. Chronic obstructive pulmonary disease genetics: a review of the past and a look into the future. Copd. 2014;1:33–46.
  • Kjeldgaard P, Dahl R, Løkke A, et al. Detection of COPD in a high-risk population: should the diagnostic work-up include bronchodilator reversibility testing? Int J Chron Obstruct Pulmon Dis. 2015;10:407–414.
  • Ning MM, Lo EH. Opportunities and challenges in omics. Transl Stroke Res. 2010;1:233–237.
  • Drucker E, Krapfenbauer K. Pitfalls and limitations in translation from biomarker discovery to clinical utility in predictive and personalised medicine. Epma J. 2013;4:7.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.