Microarray analysis of long non-coding RNAs in COPD lung tissue Hui Bi1§, Ji Zhou1§, Dandan Wu1, Wei Gao1, Lingling Li1, Like Yu2, Feng Liu2, Mao Huang1, Ian M Adcock3, Peter J Barnes3, Xin Yao1* 1Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; 2 Department of Respiratory Medicine, Nanjing Chest Hospital; 3Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK §These authors contributed equally to this work. *Corresponding author Dr Xin Yao Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, China, 210029 Tel: (86) 18651608881; Fax: (86) 2583724440 Email: yaoxin@njmu.edu.cn Keywords: long noncoding RNA, COPD, Microarray analysis Abstract Background: Long noncoding RNAs (lncRNAs) play an important regulatory role in a variety of biological and pathological processes and are implicated in the pathogenesis of many human diseases. In this study, we describe the expression of lncRNAs and mRNAs in lung tissue form three non-smokers, five smokers without chronic obstructive pulmonary disease (COPD) and five smokers with COPD using microarray analysis. Methodology: RNA was extracted from human lung tissue and analysed using an Agilent Human lncRNA + mRNA Array v2.0 system. Gene Ontology (GO) and pathway analysis was then performed and mapped genes to the Kyoto Encyclopedia of Genes and Genomes (KEGG) database. Results: 39,253 distinct lncRNA transcripts were detected in the lung tissues of all subjects. In smokers without COPD 87 lncRNAs were significantly up-regulated and 244 down-regulated compared to non-smokers with RNA50010︱UCSC-9199-1005 (fold-change: 13.02297) and RNA58351|CombinedLit_316_550 (fold-change: 12.609704) the most overand under-regulated respectively. In contrast, in COPD patients 120 lncRNAs were overexpressed and 43 underexpressed compared with smokers without COPD with RNA44121 ︱ UCSC-2000-3182 (fold-change: 8.721127) and RNA43510 ︱ UCSC-1260-3754 (fold-change: 5.527549) being the most over- and under-regulated respectively. GO and pathway analysis indicated that cigarette smoking was associated with activation of metabolic pathways and whereas COPD transcripts were associated with ‘hematopoietic cell lineage', intermediary metabolism and immune system processes. Conclusions/Significance: Our study is the first to determine the genome-wide expression of lncRNAs in lung tissue by microarray analysis. The results show that cigarette smoking and the presence of COPD alters the expression of lncRNAs in lung tissues and that these lncRNAs might play a roles in modulating pathways implicated in disease onset and progression such as intermediary metabolism and the immune system. Introduction Chronic obstructive pulmonary disease (COPD), a progressive disease characterised by airway limitation, is considered the third leading cause of mortality today and estimated by researchers to become the sixth most common cause of disability world-wide by 2020.1 COPD seriously threatens the quality of life of humans, and treatment of the disease often results in high medical costs.2,3 To the best of our knowledge, inflammation, protease-antiprotease imbalance and oxidative stress play crucial roles in the progression of COPD but they are not fully accountable for the disease process. Although cigarette smoking is the most common risk factor that contributes to the pathogenesis of COPD, only about 20% of smokers develop COPD,4 which suggests that genetic predisposition or epigenetic factors or some other cellular susceptibility mechanisms must play an important role in determining which smokers develop airway obstruction. With the advancement of high-resolution microarray analysis methods and genome-wide sequencing technology, long noncoding RNA (lncRNA) has generated considerable interest over the last decade. LncRNA is generally defined as transcripts of greater than 200 nucleotides without protein coding functions. LncRNAs play important roles in the regulation of gene expression5-7 in diverse biological and pathological processes8-14 and are abnormally expressed in many human diseases.14 Several studies show that lncRNAs participate in the process of oxidative stress, inflammation, apoptosis, cell growth and viability.13,15,16 Thai et al.,16 for example, recently found that lncRNA-SCAL1 (smoke and cancer-associated lncRNA1) could mediate protection against oxidative stress-induced by cigarette smoke extract by acting downstream of NRF2 in airway epithelial cells. Over the last decade, although there has been considerable progress in understanding lncRNAs;, the exact functions of most lncRNAs remain unknown. The expression and biological functions of lncRNAs in COPD are currently unknown. In our study, we profiled the expression of lncRNAs and mRNAs in the lung tissue of three non-smokers, five smokers without COPD and five smokers with COPD using microarray analysis. Our results showed that lncRNA and mRNA expression profiles differ significantly between the lung tissues of non-smokers, smokers and smokers with COPD. This finding suggests that altered expression levels of lncRNAs play important roles in the occurrence and development of COPD. An investigation into the relations between various signal pathways and lncRNAs may supply novel insights into the molecular regulation of COPD and provide new methods for diagnosing and treating COPD. Materials and Methods Patient samples and RNA extraction The study was approved by the Medical Ethical Committee of the First Affiliated Hospital of Nanjing Medical University. Samples were obtained from lung resection tissue taken from three non-smokers, five normal smokers and five smokers with COPD for microarray analysis of lncRNAs. Smokers and non-smokers were on no medication, whilst 3 COPD patients were on inhaled corticosteroids andβ-adrenergic agonists and three only used theophylline. All patients were confirmed as suffering from lung cancer following pathological analysis. Clinical information on these subjects is summarised in Table 1. Lung tissue extracted from each subject was snap-frozen in liquid nitrogen immediately after resection. Total RNA was extracted using Trizol reagent (Invitrogen) according to the manufacturer’s instructions. Total RNA from each sample was quantified using a NanoDrop ND-1000, and RNA integrity was assessed using standard denaturing agarose gel electrophoresis. DNA microarray An Agilent Human lncRNA + mRNA Array v2.0 system was designed with four identical arrays per slide (4 x 180K format); each array contained a probe set comprising 39,000 human lncRNA transcripts and 32,000 human mRNAs. These lncRNA and mRNA target sequences were merged from multiple databases: 4,765 were from RefSeq; 12,754 were from ENSEMBL; 8,195 lincRNA were from the John Rinn Lab;17 1,289 were from NRED (ncRNA Expression Database); 17,203 were from H-InvDB; 2,975 were from ENCODE; 529 were from Combined Lit; 1,053 were from Antisense ncRNA pipeline; 407 were from Hox ncRNAs; 481 were from UCRs and 848 were from the Chen Ruisheng Lab (Institute of Biophysics, Chinese Academy of Science). Each RNA transcript was detected by 2 probes. The array also contained 4,974 Agilent control probes. RNA amplification, labelling and hybridisation cDNA labelled with fluorescent dyes (Cy5 and Cy3-dCTP) was produced using Eberwine’s linear RNA amplification method with subsequent enzymatic reaction.18 The yield of labelled cDNA was improved by using a CapitalBio cRNA amplification and labelling kit (CapitalBio). Double-stranded cDNAs (containing the T7 RNA polymerase promoter sequence) were synthesised from 1g of total RNA using CbcScript reverse transcriptase with a cDNA synthesis system according to the manufacturer’s protocol (Capitalbio) with T7 Oligo (dT) and T7 Oligo (dN) primers. After completion of double-stranded cDNA (dsDNA) synthesis using DNA polymerase and RNase H, the dsDNA products were purified using a PCR NucleoSpin Extract II Kit (MN) and eluted with 30L of elution buffer. The eluted double-stranded cDNA products were vacuum-evaporated to 16L and then subjected to 40L in vitro transcription reactions at 37°C for 14h using T7 Enzyme Mix. The amplified cRNA was purified using an RNA clean-up kit (MN). Klenow enzyme labelling was adopted after reverse transcription using CbcScript II reverse transcriptase. Briefly, 2g of amplified RNA was mixed with 4g of random nanomer, denatured at 65°C for 5 min, and chilled on ice. Then, 5L of 4×first-strand buffer, 2L of 0.1M DTT, and 1.5L of CbcScript II reverse transcriptase were added to the RNA. The mixtures were incubated first at 25°C for 10min and then at 37°C for 90min. The cDNA products were purified using the PCR NucleoSpin Extract II Kit (MN) and then vacuum-evaporated to 14L. The cDNA was mixed with 4g of random nanomer, heated to 95 C for 3min, and then snap-cooled on ice for 5min. Five L of Klenow buffer, dNTP and Cy5-dCTP or Cy3-dCTP (GE Healthcare) were added to yield final concentrations of 240M dATP, 240M dGTP, 240M dTTP, 120M dCTP, and 40M Cy-dCTP. 1.2 L of Klenow enzyme was then added to this mixture, and the reaction was performed at 37 C for 90min. Labelled cDNA was purified with the PCR NucleoSpin Extract II Kit (MN), and re-suspended in elution buffer. Labelled controls and test samples labelled with Cy5-dCTP and Cy3-dCTP were dissolved in 80L of hybridisation solution containing 3×SSC, 0.2% SDS, 5× Denhardt’s solution and 25% formamide. The DNA in the hybridisation solution was denatured at 95°C for 3min prior to loading onto a microarray. Arrays were hybridised and pre-formed in a CapitalBio BioMixerTM II Hybridisation Station overnight at a rotation speed of 8rpm and 42 C and then washed with 0.2% SDS, 2× SSC at 42°C for 5min and 0.2× SSC at room temperature for 5min. Data analysis The lncRNA + mRNA array data were analysed for data summarisation, normalisation and quality control using GeneSpring V11.5 software (Agilent). To select differentially expressed genes, we used threshold values of ≥2- and ≤2-fold change and a Benjamini-Hochberg corrected p-value of 0.05. The data was Log2 transformed and median centred by genes using the Adjust Data function of CLUSTER 3.0 software. Further analysis was performed by hierarchical clustering with average linkages. Finally, we performed tree visualisation using Java Treeview (Stanford University School of Medicine, Stanford, CA, USA). Functional group analysis We used the http://bioinfo.capitalbio.com/mas3/ system to conduct Gene Ontology (GO) and pathway analysis. The GO project provides a controlled vocabulary for describing genes and gene product attributed to any organism. The ontology covers three domains: biological process, cellular component and molecular function. Pathway analysis is a functional analysis method that maps genes to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Statistical analysis was performed to determine the degree of enrichment of the genes in each pathway. Construction of the co-expression network The coding-non-coding gene co-expression network (CNC network) was constructed based on the correlation analysis between the differentially expressed lncRNA and mRNA. For each pair of genes, a Pearson correlation was calculated and the significant correlation pairs were chosen to construct the network. LncRNAs and mRNAs with Pearson correlation coefficients not less than 0.99 were selected to draw the network though the open source bioinformatics software Cytoscape. In a network analysis, the degree of centrality is defined as the number of linkages one node has to the other. A degree is the simplest and most important measures of a gene centrality within a network that determining the relative importance.19 Quantitative real-time PCR Total RNA was extracted using TRIzol reagent (Invitrogen, CA, USA) and then reverse-transcribed using the PrimeScript RT Reagent Kit with gDNA Eraser (Perfect Real Time) (TaKaRa, Dalian, China). Six lncRNA expressions in tissues were measured by quantitative PCR (qPCR) using SYBRGreen assays (TaKaRa, Dalian, China). used as an internal control. GAPDH was The primers used in this study were listed in Table 2. To obtain quantitative results, the expression of each lncRNA was represented as fold-changes using the △△Ct method.20 Differences in lncRNA expression were analysed using Student’s t-test with SPSS (version 16.0, SPSS Inc.). A value of p<0.05 was considered significant. Results Overview of the lncRNA profiles 39,253 distinct lncRNA transcripts were detected in the lung tissues of non-smokers (NS), smokers without COPD (Smokers No COPD, SNC) and smokers with COPD using microarrays (Supplemental Table 1). SNC patients expressed 5,782 more lncRNAs than NS and this number was further increased in COPD patients. Of these, 87 were significantly (≥2-fold) up-regulated and 244 down-regulated in SNC versus NS (Supplemental Table 2). Compared with NS patients, RNA50010︱UCSC-9199-1005 (fold-change: 13.02297) and RNA58351|CombinedLit_316_550 (fold-change: 12.609704) were the most over- and under-regulated lncRNAs in SNC patients. In contrast, 120 lncRNAs were overexpressed and 43 underexpressed in COPD patients compared with SNC (Supplemental Table 3). RNA44121 ︱ UCSC-2000-3182 (fold-change: 8.721127) and RNA43510 ︱ UCSC-1260-3754 (fold-change: 5.527549) were the most over- and under-regulated lncRNAs in COPD patients compared with SNC subjects. (RNA42913|UCSC_554_4998, RNA43477|UCSC_1222_3795 Only 4 lncRNAs RNA53319|H-InvDB_585_590, and RNA49307|UCSC_8363_1189) were differently expressed with statistical significance (Supplemental Table 4) in both SNC versus NS and between COPD and SNC. LncRNAs may be associated with the occurrence and development of COPD. We used hierarchical clustering analysis to arrange the samples into groups based on their expression levels, thereby allowing us to hypothesise the relationships among samples. The dendrogram in figure 1 shows the relationships between the lncRNA expression patterns between SNC and NS (Fig. 1A) and between COPD patients and SNC (Fig. 1B). Overview of the mRNA profiles 32,207 coding transcripts could be detected in the lung tissues of NS, SNC and COPD patients (Supplemental Table 5). Among these transcripts, the differential (≥2-fold) expression of 97 up-regulated and 110 down-regulated mRNAs reached statistical significance between SNC and NS (Supplemental Table 6). In addition, 97 mRNA transcripts were significantly over-expressed and 37 significantly under-expressed in COPD patients but not in SNC patients (Supplemental Table 7). Clustering analysis shows the relationships among the mRNA expression patterns between SNC and NS (Fig. 2A) and between COPD patients and SNC (Fig. 2B). Overview of the co-expression network A coding-non-coding gene co-expression network (CNC network) was constructed based on the correlation analysis between the differentially expressed lncRNAs and mRNAs in COPD patients compared with healthy smokers. In this co-expression network, the CNC network node included 76 lncRNAs and 55 mRNAs (Fig. 3) with lncRNAs and mRNAs interactions presented as showing positive or negative correlations. The CNC network indicates that one mRNA could correlate with one or two lncRNAs and vice versa. The CNC network indicates potential internal adjustment mechanisms between different lncRNAs and mRNAs in COPD. GO and pathway analyses GO analysis was performed to determine the genes and gene product enrichment involved in biological processes, cellular components and molecular functions. Fisher’s exact test was used to determine whether or not the overlap between the differentially expressed gene list and the GO annotation list is greater than that expected. In the group ‘SNC versus NS’, the highest enriched GOs targeted genes by the significantly different transcripts were cellular process (ontology: biological process), protein binding (ontology: molecular function) and cell part (ontology: cellular component) (Figs. 4, 5 and 6). Pathway analysis indicated that 60 pathways corresponded to significantly different transcripts and that the most enriched network was ‘neuroactive ligand-receptor interaction,’ composed of five targeted genes (Table 3 & Supplementary Table 8). GO and pathway analyses showed that some differently expressed mRNAs caused by cigarette smoking are involved in primary metabolism and key signalling pathways (e.g., calcium signalling pathway, cytokine-cytokine receptor interaction). In the group ‘COPD patients versus SNC’, we found that the highest enriched GOs targeted genes by significantly different transcripts were cellular process (ontology: biological process), protein binding (ontology: molecular function) and cell part (ontology: cellular component) (Figs. 4, 5 and 6). Pathway analysis indicated that 38 pathways corresponded to significantly different transcripts and that the most enriched network was ‘hematopoietic cell lineage', which was composed of five targeted genes (Table 3 & Supplementary Table 9). GO and pathway analyses also showed that different mRNAs may be involved in primary metabolism and immunological signalling pathways. Real time quantitative PCR validation We selected seven over-regulated lncRNAs (RNA37093 |ENCODE-1963-473, RNA175499|ENST00000544591 RNA48255|UCSC-7110-1509, RNA174930|ENST00000508732, RNA47218|UCSC_5826_1803, RNA53748|H-InvDB_1025_432, RNA44480|UCSC_2447_2885, RNA43329|UCSC_1041_4013, RNA44021|UCSC_1880_3276 RNA39398|RefSeq_1374_2314, and RNA39240|RefSeq_1208_2498) and three under-regulated lncRNAs (RNA165538|XLOC-007769, RNA147089|nc-HOXC11-109 and RNA35262|ENCODE-120-2690) for confirmation of expression in two sets of lung tissue samples using qPCR (Table 4). Gene array expression data are expressed as mean values. The results obtained indicate 71.4% consistency between the qPCR findings and microarray data. Discussion Previous evidence has shown that lncRNAs are important functional molecules engaged in diverse gene regulatory functions, including many biological and pathological processes.8-14 LncRNAs are closely correlated with a variety of human diseases,14 including cancer and asthma.21 However, no reports on the expression of lncRNAs in COPD have yet been published. In the present study, we investigated lncRNA expression profiles in the lung tissues of NS, SNC and smokers with COPD using microarray analysis and found that lncRNA expression levels were statistically significantly altered among these groups. We found that hundreds of lncRNAs were differentially expressed (≥2-fold change) in lung tissue from smokers without COPD compared with non-smokers and in lung tissue from smokers with COPD and smokers without COPD. Also, hundreds of differentially expressed lncRNAs (≥2-fold change) were found in smokers without COPD but not in non-smokers. results indicate that smoking alters the expression of lncRNAs. These GO and pathway analyses revealed that these lncRNAs are associated with changes in key pathogenic processes of COPD caused by cigarette smoking. GO and pathway analyses showed that different lncRNAs and mRNAs are involved in the metabolism of several cellular mediators and in signalling pathways linked to the altered immune system in COPD patients. Recent studies have uncovered the potential roles of adaptive immunity22-24 and autoimmune responses25 in patients with COPD. A specific pattern of inflammation appears to occur in the airways and parenchyma of COPD patients who smoke, with the predominance of CD8+ and CD4+ cells and, in more severe diseases, the presence of lymphoid follicles containing B lymphocytes and T cells.26 The strong agreement between our results and the previous findings suggests that further research on the defence mechanism of the immune system against COPD will be useful in revealing other mechanisms related to the disease. We found that the expression of many lncRNAs is significantly correlated with the expression of dozens of protein-coding genes through the CNC co-expression network. These lncRNAs may be involved in the regulatory expression of coding genes in COPD patients. Transcription of lncRNAs is now known to regulate the expression of genes in close genomic proximity (cis-acting regulation) and to target distant transcriptional activators or repressors (trans-acting) via a variety of mechanisms.7 Several studies have shown that knockdown or low expression of lncRNAs could lead to the decreased expression of their neighbouring protein-coding genes. An over-regulated lncRNA, UCSC_3382_2454, was found to be positively associated with cytochrome P450 1B1 (CYP1B1) enzymes. CYP enzymes play a key role in catalysing specific enzymes associated with breakdown and removal of deleterious xenobiotics and products of oxidative stress resulting from tobacco smoking in COPD patients.27 Interestingly, the over-regulated lncRNAs UCSC_2447_2885 and UCSC_2_26019 were found to be negatively correlated with haptoglobin and raised plasma haptoglobin is associated with the increased incidence of COPD exacerbations resulting in hospitalisation.28 The function of lncRNAs has been most extensively studied in tumorigenesis where they play important roles in the regulatory mechanisms of oxidative stress, inflammation, apoptosis, cell growth and viability.13,15,16 All these processes have also been implicated in the etiology of COPD.29 In the management of COPD, interleukin-6 (IL-6) is a useful clinical biomarker predicting worsening exercise tolerance and greater mortality,30 whilst Perry et al. reported that IL-6 release could be regulated by miR-221, a type of ncRNA, in airway smooth muscle cells, through modulation of the cyclin-dependent kinase inhibitors, p21(WAF1) and p27(kip1).31 In our study, induction of RNA175876|ENST00000554946 could affect the progress of inflammation by regulating the neighbouring protein-coding gene NF-B activating protein 1 (also known as TAB3 or MAP3K7 Binding Protein 3). Induction of RNA147089|nc-HOXC11-109, which probably appears to regulate the neighbouring protein-coding gene keratin 7, affects cell growth and viability. Keratin 7 is a Type II member of the keratin superfamily, and loss of keratin 7 leads to increased proliferation of the bladder urothelium.32 under-regulated lnRNAs, such RNA45078|UCSC_3181_2530, A large number of significantly over- and as RNA43329|UCSC_1041_4013, RNA43297|UCSC_1005_4076 RNA53055|H-InvDB_314_668, are located near the IG gene. and These findings are in line with the results from GO and pathway analyses and suggest the regulation of immune mechanisms in the progress of COPD. Although indicating several potentially important pathways in COPD this study has some limitations. Firstly the low number of subjects used in each group and the RT-qPCR analysis of only 10/14 lncRNAs means that validation of these changes needs to be performed in larger numbers of subjects. Furthermore, since the studies were performed in lung tissues and not individual cell types it is possible that the differences seen reflect altered recruitment and/or activation of infiltrating inflammatory cells in response to disease or cigarette smoking. Future experiments will be needed to determine the cellular source of these lncRNAs using in situ hybridisation and/or laser capture microdissection of single cell types as there is no validated bioinformatics package to deconvolute the lncRNA data to assign to individual cell types. Knock down or overexpression of specific lncRNAs leading to altered expression of mRNAs could then be associated with individual cell types. Third, the presence of lung cancer in response to smoking could affect lncRNA expression as previously described.33 Our experimental data showed the changes of LncRNAs in COPD patients compared with control group but ideal control group would be samples from non-smoking lung cancer patients. In conclusion, our study is the first to determine genome-wide lncRNA expression in COPD lung in comparison with NS and smokers without COPD by microarray analysis. Cigarette smoking altered the expression of hundreds of lncNRAs, which might play a role, at least in part, in the pathological changes generated by cigarette smoking. We also found large numbers of lncRNAs differentially expressed in COPD patients independent of smoking status. GO and pathway analyses also suggested that COPD may have components of metabolic and systemic immune diseases. Investigations into the relations between various signalling pathways and lncRNAs may supply novel insights into the molecular regulation of COPD and provide new methods for diagnosing and treating COPD. Further research is necessary to understand the molecular mechanisms and biological functions of lncRNAs in COPD. Acknowledgements: This study was helped by Dr. Liang Chen and Dr. Quan Zhu for the clinical information support. Contributors: XY conceived and designed the experiments, HB, JZ, FL and DW performed the experiments, WG and LL analyzed the data, HB and LY wrote the initial draft of the manuscript and MH, IMA, PJB and XY revised the manuscript and indicated where additional experimental data was required. All authors read and approved the final manuscript. Competing interests: The authors have declared that no competing interests exist. Funding: This study was supported by the National Natural Science Foundation of China (81070025), Jiangsu Health Promotion Project, and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD, JX10231801). References 1. Global strategy for the diagnosis,management,and prevention of chronic obstructive pulmonary disease. 2011. 2. Carr SJ, Hill K, Brooks D, Goldstein RS. Pulmonary rehabilitation after acute exacerbation of chronic obstructive pulmonary disease in patients who previously completed a pulmonary rehabilitation program. Journal of cardiopulmonary rehabilitation and prevention 2009;29(5):318-24. 3. Sullivan SD, Ramsey SD, Lee TA. The economic burden of COPD. Chest 2000;117(2 Suppl):5S-9S. 4. Lokke A, Lange P, Scharling H, Fabricius P, Vestbo J. Developing COPD: a 25 year follow up study of the general population. Thorax 2006;61(11):935-9. 5. Costa FF. Non-coding RNAs, epigenetics and complexity. Gene 2008;410(1):9-17. 6. Faghihi MA, Wahlestedt C. Regulatory roles of natural antisense transcripts. Nature reviews. Molecular cell biology 2009;10(9):637-43. 7. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell 2009;136(4):629-41. 8. Wang KC, Chang HY. Molecular mechanisms of long noncoding RNAs. Molecular cell 2011;43(6):904-14. 9. Brockdorff N, Ashworth A, Kay GF, McCabe VM, Norris DP, Cooper PJ, et al. The product of the mouse Xist gene is a 15 kb inactive X-specific transcript containing no conserved ORF and located in the nucleus. Cell 1992;71(3):515-26. 10. Brown CJ, Hendrich BD, Rupert JL, Lafreniere RG, Xing Y, Lawrence J, et al. The human XIST gene: analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell 1992;71(3):527-42. 11. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010;464(7291):1071-6. 12. Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK, Munson G, et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 2011;477(7364):295-300. 13. Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proceedings of the National Academy of Sciences of the United States of America 2009;106(28):11667-72. 14. Wapinski O, Chang HY. Long noncoding RNAs and human disease. Trends in cell biology 2011;21(6):354-61. 15. Massone S, Vassallo I, Fiorino G, Castelnuovo M, Barbieri F, Borghi R, et al. 17A, a novel non-coding RNA, regulates GABA B alternative splicing and signaling in response to inflammatory stimuli and in Alzheimer disease. Neurobiology of disease 2011;41(2):308-17. 16. Thai P, Statt S, Chen CH, Liang E, Campbell C, Wu R. Characterization of a Novel Long Non-Coding RNA, SCAL1, Induced by Cigarette Smoke and Elevated in Lung Cancer Cell Lines. American journal of respiratory cell and molecular biology 2013. 17. Orom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, et al. Long noncoding RNAs with enhancer-like function in human cells. Cell 2010;143(1):46-58. 18. Patterson TA, Lobenhofer EK, Fulmer-Smentek SB, Collins PJ, Chu TM, Bao W, et al. Performance comparison of one-color and two-color platforms within the MicroArray Quality Control (MAQC) project. Nature biotechnology 2006;24(9):1140-50. 19. Barabasi AL, Oltvai ZN. Network biology: understanding the cell's functional organization. Nature reviews. Genetics 2004;5(2):101-13. 20. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic acids research 2001;29(9):e45. 21. Tsitsiou E, Williams AE, Moschos SA, Patel K, Rossios C, Jiang X, et al. Transcriptome analysis shows activation of circulating CD8+ T cells in patients with severe asthma. The Journal of allergy and clinical immunology 2012;129(1):95-103. 22. Motz GT, Eppert BL, Sun G, Wesselkamper SC, Linke MJ, Deka R, et al. Persistence of lung CD8 T cell oligoclonal expansions upon smoking cessation in a mouse model of cigarette smoke-induced emphysema. Journal of immunology (Baltimore, Md. : 1950) 2008;181(11):8036-43. 23. Motz GT, Eppert BL, Wesselkamper SC, Flury JL, Borchers MT. Chronic cigarette smoke exposure generates pathogenic T cells capable of driving COPD-like disease in Rag2-/- mice. American journal of respiratory and critical care medicine 2010;181(11):1223-33. 24. Sullivan AK, Simonian PL, Falta MT, Mitchell JD, Cosgrove GP, Brown KK, et al. Oligoclonal CD4+ T cells in the lungs of patients with severe emphysema. American journal of respiratory and critical care medicine 2005;172(5):590-6. 25. Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S, et al. Antielastin autoimmunity in tobacco smoking-induced emphysema. Nature medicine 2007;13(5):567-9. 26. Rovina N, Koutsoukou A, Koulouris NG. Inflammation and Immune Response in COPD: Where Do We Stand? Mediators of inflammation 2013;2013:413735. 27. Hukkanen J, Pelkonen O, Hakkola J, Raunio H. Expression and regulation of xenobiotic-metabolizing cytochrome P450 (CYP) enzymes in human lung. Critical reviews in toxicology 2002;32(5):391-411. 28. Engstrom G, Segelstorm N, Ekberg-Aronsson M, Nilsson PM, Lindgarde F, Lofdahl CG. Plasma markers of inflammation and incidence of hospitalisations for COPD: results from a population-based cohort study. Thorax 2009;64(3):211-5. 29. Barnes PJ, Shapiro SD, Pauwels RA. Chronic obstructive pulmonary disease: molecular and cellular mechanisms. The European respiratory journal 2003;22(4):672-88. 30. Ferrari R, Tanni SE, Caram LM, Correa C, Correa CR, Godoy I. Three-year follow-up of Interleukin 6 and C-reactive protein in chronic obstructive pulmonary disease. Respiratory research 2013;14:24. 31. Perry MM, Baker JE, Gibeon DS, Adcock IM, Chung KF. Airway smooth muscle hyperproliferation is regulated by microRNA-221 in severe asthma. American journal of respiratory cell and molecular biology 2014;50(1):7-17. 32. Sandilands A, Smith FJ, Lunny DP, Campbell LE, Davidson KM, Maccallum SF, et al. Generation and characterisation of keratin 7 (k7) knockout mice. PloS one 2013;8(5):e64404. 33. Schmidt LH, Spieker T, Koschmieder S, Schaffers S, Humberg J, Jungen D, et al. The long noncoding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer 2011;6(12):1984-92. Table 1. Clinical characteristics of volunteers Parameters Healthy non-smokers Healthy smokers COPD 3/0 5/0 5/0 58±8.5 65.6±7.9 72.4±9.2 0 23.7±1.4 29.8±2.3 Non-smoker Current smoker Current smoker FEV1% predicated NA 108.7±7.3 68.4±1.2 FEV1/FVC NA 98.3±1.4 67.2±0.8 24.2±1.6 24.4±2.2 25.2±3.4 Sex(M/F) Age (years) Smoking status Pack year Current smoker/ former/ non-smoke BMI All data are presented as meanSD NA = not available FEV1 = forced expiratory volume in 1 second FVC = forced vital capacity BMI = body mass index Table 2. PCR Primers used Primer RNA147089|nc-HOXC11 sequence(5'to 3') Forward- CAGGAACTCATGAGCGTGAA Reverse- TCATGTAAAGCAGCCACAGC RNA165538|XLOC_007769 Forward- ACAGGAGGTGGTGTCTCTGG Reverse- CTGGTGCTGAGTGAGCCATA RNA175499|ENST00000544591 Forward- ATGCCAAGAATCTGGACACC Reverse- TTCTGTCCCATCGTGTTCAA RNA37093|ENCODE_1963_473 Forward- GATGGGGTTCCACCATGTAG Reverse- GGGTGGCACATGTTATGGTT RNA48255|UCSC_7110_1509 Forward- CGAGAGTAGGAGTGAGGCAG Reverse- ATCTGGTTTCCTGGTAGCCC RNA35262|ENCODE_120_2690 RNA174930|ENST00000508732 RNA47218|UCSC_5826_1803 RNA53748|H-InvDB_1025_432 RNA44480|UCSC_2447_2885 RNA43329|UCSC_1041_4013 RNA39398|RefSeq_1374_2314 RNA44021|UCSC_1880_3276 RNA39240|RefSeq_1208_2498 Forward- CTGGGATTACAGGCGTGAGT Reverse- GCTTCTACGCCACCTCAGTC Forward-ATCCTCGGTGCTGTGACTCT Reverse-ATTTCCCCACAAACACCAAA Forward-GGTGTTGGGAAAACTGGCTA Reverse-GCCATTGCTTTTGGTGTTTT Forward-CCGTCACTCCATATGTCACG Reverse-GCTGCTTTTCCTTTCAGGTG Forward-CCCAGTTCATGGCTACTGGT Reverse-GTTCCAAGGAGGGTCTGTCA Forward-AAGAGCCTCTCCCTGTCTCC Reverse-GGCACTGTAGCACACGCTTA Forward-ATCTCCGTGCTGAAGCTGTT Reverse-AGCTGTATCCCACCAACCAG Forward-CCAGCGACACAGAAGTTTCA Reverse-AGGCTGCAGGCTGAGTGTAT Forward-ACGTGTTCCCTCATCTGTCC Reverse-TTACAGACGTGAGCCACTGC Table 3. Pathway analysis between Smokers without COPD (SNC), non-smokers (NS) and COPD patients. SNC versus NS Pathway COPD versus SNC Count p-Value q-Value 5 1.74E-04 4.96E-05 Neuroactive ligand-receptor Hematopoietic interaction Count p-Value q-Value 5 1.02E-07 8.53E-06 3 8.38E-05 0.001005105 3 1.28E-04 0.00134719 3 7.06E-04 0.003953134 2 9.44E-04 0.004957273 2 0.003723262 0.011583481 2 0.0165396 0.029560137 2 0.023637951 0.036770146 2 0.043219276 0.050422489 cell lineage Calcium signaling pathway Leukocyte Pathway 4 5.34E-04 1.07E-04 3 0.0019025 1.46E-04 transendothelial Retinol metabolism B cell receptor signaling migration pathway Cell adhesion molecules Folate biosynthesis 2 0.0020231 1.50E-04 (CAMs) Cell adhesion molecules Primary 3 0.002542 1.75E-04 (CAMs) immunodeficiency Metabolism of xenobiotics by Natural killer cell mediated 3 0.0027624 1.78E-04 cytotoxicity cytochrome P450 ABC transporters - General 2 0.0035731 2.04E-04 2 0.0055292 2.91E-04 Pathogenic Escherichia coli Purine metabolism Calcium infection - EHEC signaling pathway Neuroactive Pathogenic Escherichia coli 2 0.0055292 2.91E-04 ligand-receptor infection - EPEC interaction Inositol phosphate 2 0.0055292 2.91E-04 2 0.0063377 3.07E-04 2 0.0081051 3.52E-04 Renal cell carcinoma 2 0.0090623 3.86E-04 Chronic myeloid leukemia 2 0.0100675 3.92E-04 2 0.0103262 3.92E-04 ECM-receptor interaction 2 0.0125004 4.03E-04 Small cell lung cancer 2 0.0130725 4.12E-04 Hematopoietic cell lineage 2 0.0133628 4.12E-04 3 0.0159888 4.38E-04 GnRH signaling pathway 2 0.0197399 4.76E-04 Wnt signaling pathway 2 0.0377008 6.01E-04 metabolism Acute myeloid leukemia Adipocytokine signaling pathway Phosphatidylinositol signaling system Cytokine-cytokine receptor interaction Table 4. Validation of gene array expression data by RT-qPCR ProbeName RNA37093|ENCODE_1963_473 RNA175499|ENST00000544591 RNA48255|UCSC_7110_1509 RNA174930|ENST00000508732 RNA47218|UCSC_5826_1803 RNA53748|H-InvDB_1025_432 RNA44480|UCSC_2447_2885 RNA43329|UCSC_1041_4013 RNA39398|RefSeq_1374_2314 RNA44021|UCSC_1880_3276 RNA39240|RefSeq_1208_2498 RNA165538|XLOC_007769 RNA147089|nc-HOXC11-109 RNA35262|ENCODE_120_2690 P Value gene array 0.0038056 0.0268387 0.004614 5.0455465 3.632206 2.7927952 P Value (RT-qPCR) 0.0293 0.8639 0.108 0.018148698 2.77533 0.0203 up 0.002129219 0.004474363 0.014477932 0.00621317 0.02302896 0.01373219 0.0010184 0.0067393 0.0081903 0.0371716 1.98455 2.24561 2.40442 2.4931402 5.3074193 2.1264117 2.0241427 3.0975757 3.0646358 2.9544559 0.0103 0.0037 0.027 0.0209 0.124 0.0076 0.0261 0.0211 0.0055 0.642 up up up up up up up down down down Fold change Regulation up up up Figure legends: Figure 1. Heat maps showing the differential expression and hierarchical clustering of lncRNAs in (A) smokers with no COPD (SNC) compared with non-smokers (NS) and in (B) COPD patients compared with SNC. Figure 2. Heat maps showing the differential expression and hierarchical clustering of mRNAs in (A) smokers with no COPD (SNC) compared with non-smokers (NS) and in (B) COPD patients compared with SNC. Figure 3. Coding-non-coding gene co-expression network (CNC network) between the differentially expressed lncRNAs and mRNAs in COPD patients compared with smokers without COPD. The green spots represent mRNAs and the yellow spots represent lncRNAs. Red lines represent a positive interaction between transcripts and blue lines a negative interaction. Figure 4. The biological process of enriched GOs targeted genes by the significantly different transcripts in (A) smokers with no COPD (SNC) compared with non-smokers (NS) and (B) COPD compared with SNC. Figure 5. The molecular function of enriched GOs targeted genes by the significantly different transcripts in (A) smokers with no COPD (SNC) compared with non-smokers (NS) and (B) COPD compared with SNC. Figure 6. The cellular component of enriched GOs targeted genes by the significantly different transcripts in (A) smokers with no COPD (SNC) compared with non-smokers (NS) and (B) COPD compared with SNC.