Genome-wide MicroRNA Expression Profiles in COPD: Early Predictors for Cancer Development

Andreas Keller, Tobias Fehlmann, Nicole Ludwig, Mustafa Kahraman, Thomas Laufer, Christina Backes, Claus Vogelmeier, Caroline Diener, Frank Biertz, Christian Herr, Rudolf A Jörres, Hans-Peter Lenhof, Eckart Meese, Robert Bals, COSYCONET Study Group, Andreas Keller, Tobias Fehlmann, Nicole Ludwig, Mustafa Kahraman, Thomas Laufer, Christina Backes, Claus Vogelmeier, Caroline Diener, Frank Biertz, Christian Herr, Rudolf A Jörres, Hans-Peter Lenhof, Eckart Meese, Robert Bals, COSYCONET Study Group

Abstract

Chronic obstructive pulmonary disease (COPD) significantly increases the risk of developing cancer. Biomarker studies frequently follow a case-control set-up in which patients diagnosed with a disease are compared to controls. Longitudinal cohort studies such as the COPD-centered German COPD and SYstemic consequences-COmorbidities NETwork (COSYCONET) study provide the patient and biomaterial base for discovering predictive molecular markers. We asked whether microRNA (miRNA) profiles in blood collected from COPD patients prior to a tumor diagnosis could support an early diagnosis of tumor development independent of the tumor type. From 2741 participants of COSYCONET diagnosed with COPD, we selected 534 individuals including 33 patients who developed cancer during the follow-up period of 54 months and 501 patients who did not develop cancer, but had similar age, gender and smoking history. Genome-wide miRNA profiles were generated and evaluated using machine learning techniques. For patients developing cancer we identified nine miRNAs with significantly decreased abundance (two-tailed unpaired t-test adjusted for multiple testing P < 0.05), including members of the miR-320 family. The identified miRNAs regulate different cancer-related pathways including the MAPK pathway (P = 2.3 × 10-5). We also observed the impact of confounding factors on the generated miRNA profiles, underlining the value of our matched analysis. For selected miRNAs, qRT-PCR analysis was applied to validate the results. In conclusion, we identified several miRNAs in blood of COPD patients, which could serve as candidates for biomarkers to help identify COPD patients at risk of developing cancer.

Keywords: Biomarker; COPD; COSYCONET; Cancer; Lung; microRNA.

Copyright © 2018. Production and hosting by Elsevier B.V.

Figures

Figure 1
Figure 1
Experimental design We profiled individuals with COPD (blue) who either did not develop cancer (Cohort 1) or developed cancer (Cohort 2) during the 54-month follow-up. Genome-wide blood-borne miRNA profiles were generated for all 534 patients. These expression data were correlated with clinical data from the COSYCONET data repository using statistical approaches implemented in the programming environment R. COPD, chronic obstructive pulmonary disease; COSYCONET, COPD and SYstemic consequences-COmorbidities NETwork.
Figure 2
Figure 2
Volcano plots for the pair-wise comparison of patients developing cancer and patients not developing cancer Volcano plots show on the Y-axis the negative log10P value of the t-test and on the Y-axis the log2 fold change. Green dots indicate miRNAs with higher expression in blood samples from Cohort 1, while red dots represent miRNAs with higher expression in blood samples from Cohort 2 according to the raw P < 0.05. All dots with raw P ≥ 0.05 are colored in gray. The horizontal blue line separates miRNAs that are significant following Benjamini–Hochberg adjustment (above the line; adjusted P < 0.05) and miRNAs that are not significant anymore following the adjustment (below the line).
Figure 3
Figure 3
miRNA-target interaction network for the four core miRNAs The graphic shows for the four core miRNAs miR-320e, miR-517a-3p, miR-519d-3p, and miR-320c the joint target genes from the miRTarbase. miRNA nodes are colored in brown, genes targeted by two miRNAs are colored in blue and genes targeted by three miRNAs are colored in green.
Figure 4
Figure 4
Box-Whisker and Volcano plots for miRNAs including the information of a cancer history A. Box whisker plot for miR-150-5p for the four patient groups shown in Figure 1. Patients in Group 3 showed significantly decreased expression of miR-150-5p. B. Volcano plot for the two extreme groups: Group 1 and Group 4. Green dots indicate miRNAs with higher expression in blood samples from Group 1, while red dots represent miRNAs with higher expression in blood samples from Group 4 according to the unadjusted P < 0.05. All dots with unadjusted P ≥ 0.05 are colored in gray. The horizontal blue line separates miRNAs that are significant following Benjamini–Hochberg adjustment (above the line; adjusted P < 0.05) and miRNAs that are not significant anymore following the adjustment (below the line).
Figure 5
Figure 5
qRT-PCR validation of the expression of three miRNAs The graphic shows the CT values of expression in blood from patients developing cancer (n = 31) and those not developing cancer (n = 56) for the three miRNAs that were evaluated by qRT-PCR. * indicates significant difference in miRNA expression (P < 0.05; two-tailed t-test) between the two patient groups. n.s., non significant.

References

    1. Decramer M., Janssens W. Chronic obstructive pulmonary disease and comorbidities. Lancet Respir Med. 2013;1:73–83.
    1. Ho C.H., Chen Y.C., Wang J.J., Liao K.M. Incidence and relative risk for developing cancer among patients with COPD: a nationwide cohort study in Taiwan. BMJ Open. 2017;7
    1. Durham A.L., Adcock I.M. The relationship between COPD and lung cancer. Lung Cancer. 2015;90:121–127.
    1. Zakharkina T., Heinzel E., Koczulla R.A., Greulich T., Rentz K., Pauling J.K. Analysis of the airway microbiota of healthy individuals and patients with chronic obstructive pulmonary disease by T-RFLP and clone sequencing. PLoS One. 2013;8
    1. Sekine Y., Katsura H., Koh E., Hiroshima K., Fujisawa T. Early detection of COPD is important for lung cancer surveillance. Eur Respir J. 2012;39:1230–1240.
    1. Raviv S., Hawkins K.A., DeCamp M.M., Jr., Kalhan R. Lung cancer in chronic obstructive pulmonary disease: enhancing surgical options and outcomes. Am J Respir Crit Care Med. 2011;183:1138–1146.
    1. Biswas A., Mehta H.J., Folch E.E. Chronic obstructive pulmonary disease and lung cancer: inter-relationships. Curr Opin Pulm Med. 2018;24:152–160.
    1. Cho W.C., Kwan C.K., Yau S., So P.P., Poon P.C., Au J.S. The role of inflammation in the pathogenesis of lung cancer. Expert Opin Ther Targets. 2011;15:1127–1137.
    1. Kornum J.B., Svaerke C., Thomsen R.W., Lange P., Sorensen H.T. Chronic obstructive pulmonary disease and cancer risk: a Danish nationwide cohort study. Respir Med. 2012;106:845–852.
    1. Buist A.S., Vollmer W.M., McBurnie M.A. Worldwide burden of COPD in high- and low-income countries. Part I. The burden of obstructive lung disease (BOLD) initiative. Int J Tuberc Lung Dis. 2008;12:703–708.
    1. Chen W., Brehm J.M., Manichaikul A., Cho M.H., Boutaoui N., Yan Q. A genome-wide association study of chronic obstructive pulmonary disease in Hispanics. Ann Am Thorac Soc. 2015;12:340–348.
    1. Cho M.H., Castaldi P.J., Wan E.S., Siedlinski M., Hersh C.P., Demeo D.L. A genome-wide association study of COPD identifies a susceptibility locus on chromosome 19q13. Hum Mol Genet. 2012;21:947–957.
    1. Wain L.V., Shrine N., Artigas M.S., Erzurumluoglu A.M., Noyvert B., Bossini-Castillo L. Genome-wide association analyses for lung function and chronic obstructive pulmonary disease identify new loci and potential druggable targets. Nat Genet. 2017;49:416–425.
    1. Kowalczyk M.S., Higgs D.R., Gingeras T.R. Molecular biology: RNA discrimination. Nature. 2012;482:310–311.
    1. Jankowsky E., Harris M.E. Specificity and nonspecificity in RNA–protein interactions. Nat Rev Mol Cell Biol. 2015;16:533–544.
    1. Dang X., Qu X., Wang W., Liao C., Li Y., Zhang X. Bioinformatic analysis of microRNA and mRNA Regulation in peripheral blood mononuclear cells of patients with chronic obstructive pulmonary disease. Respir Res. 2017;18:4.
    1. Kara M., Kirkil G., Kalemci S. Differential expression of microRNAs in chronic obstructive pulmonary disease. Adv Clin Exp Med. 2016;25:21–26.
    1. Inamura K., Ishikawa Y. MicroRNA in lung cancer: novel biomarkers and potential tools for treatment. J Clin Med. 2016;5:36.
    1. Song Y., Yu X., Zang Z., Zhao G. Circulating or tissue microRNAs and extracellular vesicles as potential lung cancer biomarkers: a systematic review. Int J Biol Markers. 2018;33:3–9.
    1. Leidinger P., Brefort T., Backes C., Krapp M., Galata V., Beier M. High-throughput qRT-PCR validation of blood microRNAs in non-small cell lung cancer. Oncotarget. 2016;7:4611–4623.
    1. Keller A., Leidinger P., Bauer A., Elsharawy A., Haas J., Backes C. Toward the blood-borne miRNome of human diseases. Nat Methods. 2011;8:841–843.
    1. Wei J., Li F., Yang J., Liu X., Cho W.C. MicroRNAs as regulators of airborne pollution-induced lung inflammation and carcinogenesis. Arch Toxicol. 2015;89:677–685.
    1. Keller A., Leidinger P., Vogel B., Backes C., ElSharawy A., Galata V. miRNAs can be generally associated with human pathologies as exemplified for miR-144. BMC Med. 2014;12:224.
    1. Leidinger P., Keller A., Borries A., Huwer H., Rohling M., Huebers J. Specific peripheral miRNA profiles for distinguishing lung cancer from COPD. Lung Cancer. 2011;74:41–47.
    1. Backes C., Meese E., Keller A. Specific miRNA disease biomarkers in blood, serum and plasma: challenges and prospects. Mol Diagn Ther. 2016;20:509–518.
    1. Cho W.C. Promises and challenges in developing miRNA as a molecular diagnostic tool for lung cancer. Expert Rev Mol Diagn. 2011;11:763–766.
    1. Karch A., Vogelmeier C., Welte T., Bals R., Kauczor H.U., Biederer J. The German COPD cohort COSYCONET: aims, methods and descriptive analysis of the study population at baseline. Respir Med. 2016;114:27–37.
    1. Mannino D.M., Buist A.S. Global burden of COPD: risk factors, prevalence, and future trends. Lancet. 2007;370:765–773.
    1. Molina J.R., Yang P., Cassivi S.D., Schild S.E., Adjei A.A. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83:584–594.
    1. Fan L., Qi H., Teng J., Su B., Chen H., Wang C. Identification of serum miRNAs by nano-quantum dots microarray as diagnostic biomarkers for early detection of non-small cell lung cancer. Tumour Biol. 2016;37:7777–7784.
    1. Jin X., Chen Y., Chen H., Fei S., Chen D., Cai X. Evaluation of tumor-derived exosomal miRNA as potential diagnostic biomarkers for early-stage non-small cell lung cancer using next-generation sequencing. Clin Cancer Res. 2017;23:5311–5319.
    1. Keller A., Leidinger P., Borries A., Wendschlag A., Wucherpfennig F., Scheffler M. miRNAs in lung cancer – studying complex fingerprints in patient's blood cells by microarray experiments. BMC Cancer. 2009;9:353.
    1. Allavena G., Boyd C., Oo K.S., Maellaro E., Zhivotovsky B., Kaminskyy V.O. Suppressed translation and ULK1 degradation as potential mechanisms of autophagy limitation under prolonged starvation. Autophagy. 2016;12:2085–2097.
    1. Pal J.K., Ray S.S., Pal S.K. Identifying relevant group of miRNAs in cancer using fuzzy mutual information. Med Biol Eng Comput. 2016;54:701–710.
    1. Saha S., Mitra S., Yadav R.K. A stack-based ensemble framework for detecting cancer microRNA biomarkers. Genomics Proteomics Bioinformatics. 2017;15:381–388.
    1. Ludwig N., Leidinger P., Becker K., Backes C., Fehlmann T., Pallasch C. Distribution of miRNA expression across human tissues. Nucleic Acids Res. 2016;44:3865–3877.
    1. Fehlmann T., Ludwig N., Backes C., Meese E., Keller A. Distribution of microRNA biomarker candidates in solid tissues and body fluids. RNA Biol. 2016;13:1084–1088.
    1. Keller A., Leidinger P., Gislefoss R., Haugen A., Langseth H., Staehler P. Stable serum miRNA profiles as potential tool for non-invasive lung cancer diagnosis. RNA Biol. 2011;8:506–516.
    1. Keller A., Backes C., Haas J., Leidinger P., Maetzler W., Deuschle C. Validating Alzheimer's disease micro RNAs using next-generation sequencing. Alzheimers Dement. 2016;12:565–576.
    1. Backes C., Sedaghat-Hamedani F., Frese K., Hart M., Ludwig N., Meder B. Bias in high-throughput analysis of miRNAs and implications for biomarker studies. Anal Chem. 2016;88:2088–2095.
    1. Fehlmann T., Reinheimer S., Geng C., Su X., Drmanac S., Alexeev A. cPAS-based sequencing on the BGISEQ-500 to explore small non-coding RNAs. Clin Epigenetics. 2016;8:123.
    1. Ludwig N., Becker M., Schumann T., Speer T., Fehlmann T., Keller A. Bias in recent miRBase annotations potentially associated with RNA quality issues. Sci Rep. 2017;7:5162.
    1. Marx A., Backes C., Meese E., Lenhof H.P., Keller A. EDISON-WMW: exact dynamic programing solution of the Wilcoxon–Mann–Whitney test. Genomics Proteomics Bioinformatics. 2016;14:55–61.
    1. Backes C., Khaleeq Q.T., Meese E., Keller A. miEAA: microRNA enrichment analysis and annotation. Nucleic Acids Res. 2016;44:W110–W116.
    1. Backes C., Keller A., Kuentzer J., Kneissl B., Comtesse N., Elnakady Y.A. GeneTrail–advanced gene set enrichment analysis. Nucleic Acids Res. 2007;35:W186–W192.
    1. Keller A., Backes C., Lenhof H.P. Computation of significance scores of unweighted Gene Set Enrichment Analyses. BMC Bioinformatics. 2007;8:290.
    1. Zafari S., Backes C., Leidinger P., Meese E., Keller A. Regulatory microRNA networks: complex patterns of target pathways for disease-related and housekeeping microRNAs. Genomics Proteomics Bioinformatics. 2015;13:159–168.

Source: PubMed

Подписаться