The sputum microbiome is distinct between COPD and health, independent of smoking history

Koirobi Haldar, Leena George, Zhang Wang, Vijay Mistry, Mohammadali Yavari Ramsheh, Robert C Free, Catherine John, Nicola F Reeve, Bruce E Miller, Ruth Tal-Singer, Adam J Webb, Anthony J Brookes, Martin D Tobin, Dave Singh, Gavin C Donaldson, Jadwiga A Wedzicha, James R Brown, Michael R Barer, Christopher E Brightling, Koirobi Haldar, Leena George, Zhang Wang, Vijay Mistry, Mohammadali Yavari Ramsheh, Robert C Free, Catherine John, Nicola F Reeve, Bruce E Miller, Ruth Tal-Singer, Adam J Webb, Anthony J Brookes, Martin D Tobin, Dave Singh, Gavin C Donaldson, Jadwiga A Wedzicha, James R Brown, Michael R Barer, Christopher E Brightling

Abstract

Background: Airway bacterial dysbiosis is a feature of chronic obstructive pulmonary disease (COPD). However, there is limited comparative data of the lung microbiome between healthy smokers, non-smokers and COPD.

Methods: We compared the 16S rRNA gene-based sputum microbiome generated from pair-ended Illumina sequencing of 124 healthy subjects (28 smokers and 96 non-smokers with normal lung function), with single stable samples from 218 COPD subjects collected from three UK clinical centres as part of the COPDMAP consortium.

Results: In healthy subjects Firmicutes, Bacteroidetes and Actinobacteria were the major phyla constituting 88% of the total reads, and Streptococcus, Veillonella, Prevotella, Actinomyces and Rothia were the dominant genera. Haemophilus formed only 3% of the healthy microbiome. In contrast, Proteobacteria was the most dominant phylum accounting for 50% of the microbiome in COPD subjects, with Haemophilus and Moraxella at genus level contributing 25 and 3% respectively. There were no differences in the microbiome profile within healthy and COPD subgroups when stratified based on smoking history. Principal coordinate analysis on operational taxonomic units showed two distinct clusters, representative of healthy and COPD subjects (PERMANOVA, p = 0·001).

Conclusion: The healthy and COPD sputum microbiomes are distinct and independent of smoking history. Our results underline the important role for Gammaproteobacteria in COPD.

Keywords: COPD; Haemophilus; Healthy airway; Microbiome; Proteobacteria.

Conflict of interest statement

KH, LG, ZW, VM, MYR, RCF, NFR, AJW, AJB, MRB have nothing to declare. JRB, BEM and RTS are employees and shareholders of GSK; GCD reports grants and personal fees from Astrazeneca and grants from Micom ltd and American Thoracic Society; MDT reports grants from GSK and other from Orion; JAW reports grants from GSK, grants from Johnson and Johnson, other from Novartis, other from Boehringer Ingelheim, other from Astra Zeneca, other from GSK, grants from GSK, grants from Astra Zeneca, grants from Boehringer Ingelheim, grants from Novartis; DS reports grants and personal fees from GlaxoSmithKline, grants and personal fees from AstraZeneca, grants and personal fees from Boehringer Ingleheim, grants and personal fees from Chiesi, personal fees from Cipla, personal fees from Genentech, grants and personal fees from Glenmark, grants and personal fees from Menarini, grants and personal fees from Mundipharma, grants and personal fees from Novartis, personal fees from Peptinnovate, grants and personal fees from Pfizer, grants and personal fees from Pulmatrix, grants and personal fees from Therevance, grants and personal fees from Verona; CEB reports grants and personal fees from GSK, grants and personal fees from Novartis, grants and personal fees from Genentech, grants and personal fees from Chiesi, personal fees from Sanofi/Regeneron, grants and personal fees from 4DPharma, grants and personal fees from BI, grants and personal fees from Mologics, grants and personal fees from Gossamer, grants and personal fees from AZ/MedImmune, grants and personal fees from TEVA, outside the submitted work. CEB reports grants from MRC COPDMAP, grants from AirPROM (FP7–270194), during the conduct of the study.

Figures

Fig. 1
Fig. 1
Microbiome profile of Healthy volunteers based on smoking pack year history. a Relative abundance of major phyla between all healthy (n = 124) represented in the outer ring followed by healthy < 10 PY smoking history subgroup (n = 96) in the middle ring and innermost ring representing healthy ≥10 PY history subgroup (n = 28). b Relative abundance of major genera between all healthy (n = 124) represented in the outer ring followed by healthy < 10 PY (n = 96) in the middle ring and innermost ring representing healthy ≥10 PY history (n = 28). c Principal coordinate analysis (PCoA) analysis of weighted unifrac distance measures relative to pack year history. d Alpha diversity indices comparison between. < 10 PY and ≥ 10 PY smoking sub-groups. Chao1 and observed_otus are represented as bar chart as mean and standard deviation; Shannon index is represented by box whisker plot showing median, interquartile range and minimum and maximum. **. P < 0.01
Fig. 2
Fig. 2
Microbiome profile of COPD subjects based on smoking pack year history. a Relative abundance of major phyla between all COPD (n = 218) represented in the outer ring followed by ex-smokers (n = 148) in the middle ring and innermost ring representing current smokers history (n = 70). b Relative abundance of major genera between all COPD (n = 218) represented in the outer ring followed by ex- smokers (n = 148) in the middle ring and innermost ring representing current smoker (n = 70). c PCoA analysis of weighted unifrac distance measures relative to pack year history. d Alpha diversity indices comparison between the two smoking groups
Fig. 3
Fig. 3
Comparison of Microbiome profile between Healthy and COPD. a Relative abundance of major phyla between COPD (n = 218) represented in the outer and inner ring representing healthy (n = 124). b Relative abundance of major genera between COPD (n = 218) represented in the outer ring and inner ring representing healthy volunteers (n = 28). c PCoA analysis of weighted unifrac distance measures between healthy and COPD subjects. d Alpha diversity indices comparison between Healthy and COPD subjects. ****, P < 0.00001
Fig. 4
Fig. 4
Bacterial groups distinguishing health and COPD microbiome. Each of the circles in the cladogram represent a bacterial taxa and each ring a taxonomy level starting with Kingdom (Archaea and Bacteria) in the innermost circle. Green coloured circles and zones represent bacterial taxa dominant in health and red in COPD. Circle sizes are correlated to bacterial abundance. Taxa level phylum (p_) and class (c_) are mentioned in the figure. Order (o_), Family (f_) and genus (g_) are abbreviated in the figure
Fig. 5
Fig. 5
Predictive functional profiling shows distinct clustering of COPD and Healthy sputum microbiome. a PCA analysis of functional groups inferred from 16S rDNA microbial community. b lists the top 19 functional groups which were significantly different (p < 0.05, multiple comparison corrected) and had > 0.2% difference between COPD and healthy subjects

References

    1. Sethi S, Murphy TF. Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N Engl J Med. 2008;359(22):2355–2365. doi: 10.1056/NEJMra0800353.
    1. Dickson RP, Martinez FJ, Huffnagle GB. The role of the microbiome in exacerbations of chronic lung diseases. Lancet. 2014;384(9944):691–702. doi: 10.1016/S0140-6736(14)61136-3.
    1. Erb-Downward JR, Thompson DL, Han MK, Freeman CM, McCloskey L, Schmidt LA, et al. Analysis of the lung microbiome in the "healthy" smoker and in COPD. PLoS One. 2011;6(2):e16384. doi: 10.1371/journal.pone.0016384.
    1. Pragman AA, Kim HB, Reilly CS, Wendt C, Isaacson RE. The lung microbiome in moderate and severe chronic obstructive pulmonary disease. PLoS One. 2012;7(10):e47305. doi: 10.1371/journal.pone.0047305.
    1. Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, et al. Disordered microbial communities in asthmatic airways. PLoS One. 2010;5(1):e8578. doi: 10.1371/journal.pone.0008578.
    1. Einarsson GG, Comer DM, McIlreavey L, Parkhill J, Ennis M, Tunney MM, et al. Community dynamics and the lower airway microbiota in stable chronic obstructive pulmonary disease, smokers and healthy non-smokers. Thorax. 2016;71(9):795–803. doi: 10.1136/thoraxjnl-2015-207235.
    1. Cabrera-Rubio R, Garcia-Núñez M, Setó L, Antó JM, Moya A, Monsó E, et al. Microbiome diversity in the bronchial tracts of patients with chronic obstructive pulmonary disease. J Clin Microbiol. 2012;50(11):3562–3568. doi: 10.1128/JCM.00767-12.
    1. Huang YJ, Sethi S, Murphy T, Nariya S, Boushey HA, Lynch SV. Airway microbiome dynamics in exacerbations of chronic obstructive pulmonary disease. J Clin Microbiol. 2014;52:2813–2823. doi: 10.1128/JCM.00035-14.
    1. Wang Z, Bafadhel M, Haldar K, Spivak A, Mayhew D, Miller BE, et al. Lung microbiome dynamics in COPD exacerbations. Eur Respir J. 2016;47(4):1082–1092. doi: 10.1183/13993003.01406-2015.
    1. Haldar K, Bafadhel M, Lau K, Berg A, Kwambana B, Kebadze T, et al. Microbiome balance in sputum determined by PCR stratifies COPD exacerbations and shows potential for selective use of antibiotics. PLoS One. 2017;12(8):e0182833. doi: 10.1371/journal.pone.0182833.
    1. Charlson ES, Chen J, Custers-Allen R, Bittinger K, Li H, Sinha R, et al. Disordered microbial communities in the upper respiratory tract of cigarette smokers. PLoS One. 2010;5(12):e15216. doi: 10.1371/journal.pone.0015216.
    1. Johannessen A, Omenaas ER, Bakke PS, Gulsvik A. Implications of reversibility testing on prevalence and risk factors for chronic obstructive pulmonary disease: a community study. Thorax. 2005;60(10):842–847. doi: 10.1136/thx.2005.043943.
    1. Wang Z, Singh R, Miller BE, Tal-Singer R, Van Horn S, Tomsho L, et al. Sputum microbiome temporal variability and dysbiosis in chronic obstructive pulmonary disease exacerbations: an analysis of the COPDMAP study. Thorax. 2018;73(4):331–338. doi: 10.1136/thoraxjnl-2017-210741.
    1. Kuczynski J, Stombaugh J, Walters WA, González A, Caporaso JG, Knight R. Using QIIME to analyze 16S rRNA gene sequences from microbial communities. Curr Protoc Microbiol. 2012;27:1E–15.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Aronesty E. Command-line tools for processing biological sequencing data. 2011.
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011;27(16):2194–2200. doi: 10.1093/bioinformatics/btr381.
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. 2007;73(16):5261–5267. doi: 10.1128/AEM.00062-07.
    1. Lozupone C, Knight R. UniFrac: a new phylogenetic method for comparing microbial communities. Appl Environ Microbiol. 2005;71(12):8228–8235. doi: 10.1128/AEM.71.12.8228-8235.2005.
    1. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotech. 2013;31(9):814–821. doi: 10.1038/nbt.2676.
    1. Parks DH, Tyson GW, Hugenholtz P, Beiko RG. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics. 2014;30(21):3123–3124. doi: 10.1093/bioinformatics/btu494.
    1. Anderson MJ. A new method for non-parametric multivariate analysis of variance. Aust Ecol. 2001;26(1):32–46.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60. doi: 10.1186/gb-2011-12-6-r60.
    1. Bergen B, Endres S, Engel A, Zark M, Dittmar T, Sommer U, et al. Acidification and warming affect prominent bacteria in two seasonal phytoplankton bloom mesocosms. Environ Microbiol. 2016;18(12):4579–4595. doi: 10.1111/1462-2920.13549.
    1. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13(9):R79. doi: 10.1186/gb-2012-13-9-r79.
    1. Diao W, Shen N, Du Y, Erb-Downward JR, Sun X, Guo C, et al. Symptom-related sputum microbiota in stable chronic obstructive pulmonary disease. Int J Chronic Obstruct Pulmon Dis. 2018;13:2289–2299. doi: 10.2147/COPD.S167618.
    1. Sze MA, Dimitriu PA, Hayashi S, Elliott WM, McDonough JE, Gosselink JV, et al. The lung tissue microbiome in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;185(10):1073–1080. doi: 10.1164/rccm.201111-2075OC.
    1. Pragman AA, Lyu T, Baller JA, Gould TJ, Kelly RF, Reilly CS, et al. The lung tissue microbiota of mild and moderate chronic obstructive pulmonary disease. Microbiome. 2018;6:7. doi: 10.1186/s40168-017-0381-4.
    1. Pragman AA, Kim HB, Reilly CS, Wendt C, Isaacson RE. Chronic obstructive pulmonary disease lung microbiota diversity may be mediated by age or inhaled corticosteroid use. J Clin Microbiol. 2015;53:1050. doi: 10.1128/JCM.03320-14.
    1. Morris A, Beck JM, Schloss PD, Campbell TB, Crothers K, Curtis JL, et al. Comparison of the respiratory microbiome in healthy nonsmokers and smokers. Am J Respir Crit Care Med. 2013;187(10):1067–1075. doi: 10.1164/rccm.201210-1913OC.
    1. Shukla SD, Walters EH, Simpson JL, Keely S, Wark PAB, O’Toole RF, et al. Hypoxia-inducible factor and bacterial infections in chronic obstructive pulmonary disease. Respirology. 2020;25(1):53–63. doi: 10.1111/resp.13722.
    1. Bafadhel M, Haldar K, Barker B, Patel H, Mistry V, Barer MR, et al. Airway bacteria measured by quantitative polymerase chain reaction and culture in patients with stable COPD: relationship with neutrophilic airway inflammation, exacerbation frequency, and lung function. Int J Chron Obstruct Pulmon Dis. 2015;10:1075–1083. doi: 10.2147/COPD.S80091.

Source: PubMed

3
Subskrybuj