Dysbiosis of lower respiratory tract microbiome are associated with inflammation and microbial function variety

Kang-Jie Li, Zi-Long Chen, Yao Huang, Rui Zhang, Xiao-Qian Luan, Ting-Ting Lei, Ling Chen, Kang-Jie Li, Zi-Long Chen, Yao Huang, Rui Zhang, Xiao-Qian Luan, Ting-Ting Lei, Ling Chen

Abstract

Background: Lower respiratory tract (LRT) microbiome has been reported to associate with pulmonary diseases. Unregulated inflammation is an underlying cause of variable lung diseases. The lung microbiome may play an important role in the smoking-induced inflammatory lung diseases. What's more, the function of microbiome may be more important for understanding how microbes interact with host. Our study aims to explore the effects of smoking on the lower respiratory tract microbiome, the association between variation of lower respiratory tract microbiome and inflammation and whether smoking exposure changes the function of lower respiratory tract microbime.

Methods: Forty male mice were randomly divided into smoking group and non-smoking group, and the smoking group was exposed to cigarette smoke for 2 h per day for 90 days. After experiment, the blood samples were collected to measure the concentration of interleukin-6 (IL-6) and C reactive protein (CRP) by ELISA. Lung tissue samples were used to detect the community and diversity of lower respiratory tract microbiome through 16S rRNA gene quantification and sequencing technology. ANOSIM and STAMP were performed to analyze the differences of the microbial community structure between smoking group and non-smoking group. SPSS 24.0 software was used to analyze the correlations between microbiome and inflammation mediators through scatter plots and Spearman correlation coefficient. Microbial metabolic function was predicted by PICRUSt based on the 16 s rRNA gene quantification and sequencing results. PATRIC database was searched for the potential pathogenic bacteria in lower respiratory tract.

Results: Our results suggested that smoking had markedly effects on the microbiota structure of lower respiratory tract based on Bray-Curtis distance (R2 = 0.084, p = 0.005) and on unweighted uniFrac distance (R2 = 0.131, p = 0.002). Smoking mainly affected the abundance of microbiome which belong to Proteobacteria phyla and Firmicutes phyla. Moreover, our results also found that smoking increased the abundance of Acinetobacter, Bacillus and Staphylococcus, which were defined as pathogenic bacteria. Inflammatory mediators were observed to associate with certain microbiome at every level. Most of microbiome which were associated with inflammation belonged to Proteobacteria phyla or Firmicutes phyla. Moreover, we found that the decreased microbiome in smoking group, including Oceanospirillales, Desulfuromonadales, Nesterenkonia, and Lactobacillaceae, all were negatively correlated with IL-6 or CRP. Based on the level of inflammation, the abundance of microbiome differs. At genus level, Lactobacillus, Pelagibacterium, Geobacter and Zoogloea were significantly higher in smoking group with lower IL-6 concentration. The abundance of microbiome was not observed any statistical difference in subgroups with different weight. Three dominant genus, defined as pathogen, were found higher in the smoking group. The microbial functional prediction analysis revealed that ABC-type transport systems, transcription factors, amino acide transport and metabolism, arginine and proline metabolism et al. were distinctively decreased in smoking group, while the proportions of replication, recombination and repair, ribosome, DNA repair and recombination proteins were increased in smoking group (q < 0.05).

Conclusions: Members of Proteobacteria phyla and Firmicutes phyla played an important role in the microbial community composition and keeping a relatively balanced homeostasis. Microbiome dysbiosis might break the balance of immune system to drive lung inflammation. There might exist potential probiotics in lower respiratory tract, such as Lactobacillaceae. The altered function of Lower respiratory tract microbiome under smoking exposure may affect the physiological homeostasis of host.

Keywords: Function of microbiome; Inflammatory mediators; Lower respiratory tract; Microbiome; Smoking.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Heat map of correlation coefficient value. Column A represents the correlation between IL-6 and microbiome in smoking group. Column B represents the correlation between CRP and microbiome in smoking group. Column C represents the correlation between IL-6 and microbiome in non-smoking group. Column D represents the correlation between CRP and microbiome in non-smoking group. Twenty microbiome which significantly associate with any inflammatory mediator in either smoking group or non-smoking group are stated
Fig. 2
Fig. 2
Relative abundance of function at KEGG Level1. Samples B2 to B20 belong to the smoking group and samples c3 to c16 belong to the non-smoking group
Fig. 3
Fig. 3
Difference of metabolism, genetic information processing and environmental information processing in two groups (level 1). Group B colored with blue represents the smoking group and group C colored with yellow represents the non-smoking group
Fig. 4
Fig. 4
41 level 2 KEGG Orthology groups in smoking and non-smoking groups. Samples A2 to B20 belong to the smoking group and samples C1 to C18 belong to the non-smoking group
Fig. 5
Fig. 5
Differences among the 41 KEGG2 pathway between two groups. Group B colored with blue represents the smoking group and group C colored with yellow represents the non-smoking group
Fig. 6
Fig. 6
The differences of metabolic pathways at KEGG level 3 between two groups. Group B colored with blue represents the smoking group and group C colored with yellow represents the non-smoking group
Fig. 7
Fig. 7
The category of General function prediction at COG level2. Samples A2 to B20 belong to the smoking group and samples C1 to C18 belong to the non-smoking group
Fig. 8
Fig. 8
The difference of specific functions between two groups at COG level2. Group B colored with blue represents the smoking group and group C colored with yellow represents the non-smoking group
Fig. 9
Fig. 9
Difference of the specific function of proteins in two groups. Group B colored with blue represents the smoking group and group C colored with yellow represents the non-smoking group

References

    1. Wilson M. Bacteriology of humans: an ecological perspective. Malden, MA: Blackwell Publishing; 2008.
    1. Dethlefsen L, McFall-Ngai M, Relman DA. An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature. 2007;449:811. doi: 10.1038/nature06245.
    1. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell. 2015;163:367–380. doi: 10.1016/j.cell.2015.08.058.
    1. Dalal SR, Chang EB. The microbial basis of inflammatory bowel diseases. J Clin Invest. 2014;124:4190–4196. doi: 10.1172/JCI72330.
    1. Crowe SE. Helicobacter infection, chronic inflammation, and the development of malignancy. Curr Opin Gastroenterol. 2005;21(1):32–38.
    1. Beck JM, Young VB, Huffnagle GB. The microbiome of the lung. Transl Res. 2012;160:258–266. doi: 10.1016/j.trsl.2012.02.005.
    1. Remot A, Descamps D, Noordine ML, Boukadiri A, Mathieu E, Robert V, et al. Bacteria isolated from lungmodulate asthma susceptibility in mice. ISME J. 2017;11(5):1061–1074. doi: 10.1038/ismej.2016.181.
    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(8):2813–2823. doi: 10.1128/JCM.00035-14.
    1. Mogayzel PJ, Naureckas ET, Robinson KA, et al. Cystic fibrosis pulmonary guidelines chronic medications for maintenance of lung health. Am J Resp Crit Care. 2013;187(7):680–689. doi: 10.1164/rccm.201207-1160OE.
    1. Perez-Warnisher MT, De Miguel MDPC, Seijo LM. Tobacco use worldwide: legislative efforts to curb consumption. Ann Glob Health. 2018;84(4):571–579. doi: 10.29024/aogh.2362.
    1. Walser T, Cui X, Yanagawa J, et al. Smoking and lung cancer: the role of inflammation. Proc Am Thorac Soc. 2008;5(8):811–815. doi: 10.1513/pats.200809-100TH.
    1. Cho WC, Kwan CK, Yau S, et al. The role of inflammation in the pathogenesis of lung cancer. Expert Opin Ther Targets. 2011;15(9):1127–1137. doi: 10.1517/14728222.2011.599801.
    1. Chen J, Pitmon E, Wang K. Microbiome, inflammation and colorectal cancer. Semin Immunol. 2017;32:43–53. doi: 10.1016/j.smim.2017.09.006.
    1. Zhang R, Chen L, Cao L, Li KJ, Huang Y, Luan XQ, Li G. Effects of smoking on the lower respiratory tract microbiome in mice. Respir Res. 2018;19(1):253. doi: 10.1186/s12931-018-0959-9.
    1. Dickson RP, Erb-Downward JR, Martinez FJ, et al. The microbiome and the respiratory tract. Annu Rev Physiol. 2016;78:481–504. doi: 10.1146/annurev-physiol-021115-105238.
    1. Gibbons SM. Microbial community ecology: function over phylogeny [J] Nat Ecol Evol. 2017;1:0032. doi: 10.1038/s41559-016-0032.
    1. Louca S, Parfrey LW, Doebeli M. Decoupling function and taxonomy in the global oceanmicrobiome [J] Science. 2016;353(6305):1272–1277. doi: 10.1126/science.aaf4507.
    1. Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. doi: 10.1038/nature11234.
    1. Dang RY, Zhao CY, Fan X, Zhao ZL, Jiang XG, Li G. Effect of cigarette smoke on bacteria distribution of lower respiratory tract in mice. Shandong Med J. 2018;58(20):9–12.
    1. Mason KL, Erb Downward JR, Mason KD, Falkowski NR, Eaton KA, Kao JY, et al. Candida albicans and bacterial microbiota interactions in the cecum during recolonization following broad-spectrum antibiotic therapy. Infect Immun. 2012;80:3371–3380. doi: 10.1128/IAI.00449-12.
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci U S A. 2011;108(Suppl1):4516–4522. doi: 10.1073/pnas.1000080107.
    1. Kozich JJ, Westcott SL, Baxter NT, Highlander SK, Schloss PD. Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microbiol. 2013;79:5112–5120. doi: 10.1128/AEM.01043-13.
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing mothur: open-source, platform-independent, communitysupported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75:7537–7541. doi: 10.1128/AEM.01541-09.
    1. Schloss PD. MiSeq SOP-mothur. 2015.
    1. Teo SM, Mok D, Pham K, et al. The infant nasopharyngeal microbiome impacts severity of lower respiratory infection and risk of asthma development. Cell Host Microbe. 2015;17(5):704–715. doi: 10.1016/j.chom.2015.03.008.
    1. Amato KR, Yeoman CJ, Kent A, Righini N, Carbonero F, Estrada A, et al. Habitat degradation impacts black howler monkey (Alouatta pigra) gastrointestinal microbiomes. ISME J. 2013;7:1344–1353. doi: 10.1038/ismej.2013.16.
    1. Douglas GM, Beiko RG, Langille MGI. Predicting the functional potential of the microbiome from marker genes using PICRUSt. Methods Mol Biol. 1849;2018:169–177.
    1. Laurenzi GA, Potter RT, Kass EH. Bacteriologic flora of the lower respiratory tract. N Engl J Med. 1961;265:1273–1278. doi: 10.1056/NEJM196112282652601.
    1. Charlson ES, Bittinger K, Haas AR, Fitzgerald AS, Frank I, Yadav A, et al. Topographical continuity of bacterial populations in the healthy human respiratory tract. Am J Respir Crit Care Med. 2011;184:957–963. doi: 10.1164/rccm.201104-0655OC.
    1. Muegge BD, Kuczynski J, Knights D, Clemente JC, González A, Fontana L, et al. Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science. 2011;332:970–974. doi: 10.1126/science.1198719.
    1. Hansen AK, Hansen CH, Krych L, Nielsen DS. Impact of the gut microbiota on rodent models of human disease. World J Gastroenterol. 2014;20:17727–17736. doi: 10.3748/wjg.v20.i47.17727.
    1. Magalhaes AP, Azevedo NF, Pereira MO, et al. The cystic fibrosis microbiome in an ecological perspective and its impact in antibiotic therapy. Appl Microbiol Biotechnol. 2016;100(3):1163–1181. doi: 10.1007/s00253-015-7177-x.
    1. U.S. Department of Health and Human Services . The Health Consequences of Involuntary Exposure to Tobacco Smoke – A Report of the Surgeon General. 2006. The Health Consequences of Involuntary Exposure to Tobacco Smoke – A Report of the Surgeon General.
    1. U.S. Department of Health and Human Services . The Health Consequences of Smoking. 2004. The Health Consequences of Smoking: What it means to you – A Report of the Surgeon General.
    1. Singh N, Vats A, Sharma A, Arora A, Kumar A. The development of lower respiratory tract microbiome in mice. Microbiome. 2017;5(1):61. doi: 10.1186/s40168-017-0277-3.
    1. Barfod KK, Roggenbuck M, Hansen LH, Schjørring S, Larsen ST, Sørensen SJ, et al. The murine lung microbiome in relation to the intestinal and vaginal bacterial communities. BMC Microbiol. 2013;13:303. doi: 10.1186/1471-2180-13-303.
    1. Huang YJ, Kim E, Cox MJ, et al. A persistent and diverse airway microbiota present during chronic obstructive pulmonary disease exacerbations Omics. OMICS A J Integr Biol. 2010;14(1):9–59. doi: 10.1089/omi.2009.0100.
    1. Mejri I, Ourari B, Cherif H, et al. Pulmonary tuberculosis and lung cancer: a complex interaction. Eur Respir J. 2016;48:1. doi: 10.1183/13993003.00884-2016.
    1. Liu HX, Tao LL, Zhang J, Zhu YG, Zheng Y, Liu D, et al. Difference of lower airway microbiome in bilateral protected specimen brush between lung cancer patients with unilateral lobar masses and control subjects. Int J Cancer. 2018;142(4):769–778. doi: 10.1002/ijc.31098.
    1. Koh TJ, DiPietro LA. Inflammation and wound healing: the role of the macrophage. Expert Rev Mol Med. 2011;13:e23. doi: 10.1017/S1462399411001943.
    1. Barfod KK, Vrankx K, Mirsepasi-Lauridsen HC, Hansen JS, Hougaard KS, Larsen ST, et al. The murine lung microbiome changes during lung inflammation and intranasal Vancomycin treatment. Open Microbiol J. 2015;9:167–179. doi: 10.2174/1874285801509010167.
    1. Huffnagle GB, Dickson RP, Lukacs NW. The respiratory tract microbiome and lung inflammation: a two-way street. Mucosal Immunol. 2017;10(2):299–306. doi: 10.1038/mi.2016.108.
    1. Dickson RP, Erb-Downward JR, Martinez FJ, Huffnagle GB. The microbiome and the respiratory tract. Annu Rev Physiol. 2016;78:481–504. doi: 10.1146/annurev-physiol-021115-105238.
    1. Scales BS, Dickson RP, Huffnagle GB. A tale of two sites: how inflammation can reshape the microbiomes of the gut and lungs. J Leukoc Biol. 2016;100(5):943–950. doi: 10.1189/jlb.3MR0316-106R.
    1. Bernasconi E, Pattaroni C, Koutsokera A, Pison C, Kessler R, Benden C, et al. Airway microbiota determines innate cell inflammatory or tissue remodeling profiles in lung transplantation. Am J Respir Crit Care Med. 2016;194(10):1252–1263. doi: 10.1164/rccm.201512-2424OC.
    1. Winter SE, Winter MG, Xavier MN, Thiennimitr P, Poon V, Keestra AM, et al. Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science. 2013;339(6120):708–711. doi: 10.1126/science.1232467.
    1. Winter SE, Baumler AJ. Why related bacterial species bloom simultaneously in the gut: principles underlying the ‘Like will to like’ concept. Cell Microbiol. 2014;16(2):179–184. doi: 10.1111/cmi.12245.
    1. Winter SE, Baumler AJ. Dysbiosis in the inflamed intestine: chance favors the prepared microbe. Gut Microbes. 2014;5(1):71–73. doi: 10.4161/gmic.27129.
    1. O’Sullivan BP, Freedman SD. Cystic fibrosis. Lancet. 2009;373(9678):1891–1904. doi: 10.1016/S0140-6736(09)60327-5.
    1. Nimgampalle M, Kuna Y. Anti-Alzheimer properties of probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer's disease induced albino rats. J Clin Diagn Res. 2017;11:KC01–KC05.
    1. Dubinkina VB, Tyakht AV, Odintsova VY, Yarygin KS, Kovarsky BA. Links of gut microbiota composition with alcohol dependence syndrome and alcoholic liver disease. Microbiome. 2017;5:141. doi: 10.1186/s40168-017-0359-2.
    1. Xk W, Long Y, Zhang ZY, Sl W, Zhang MY, Wang PC. Airway Dysbacteriosis Exacerbated Murine Airway Allergic Inflammation. J SUN Yat-sen Univ (Med Sci) 2017;38(3):350–358.
    1. Gollwitzer E, Saglani O, Trompette A, Yadava K, Sherburn R, Mccoy K, et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med. 2014;20:642–647. doi: 10.1038/nm.3568.
    1. Wang J, Lesko M, Badri MH, Kapoor BC, Wu BG, Li Y, et al. Lung microbiome and host immune tone in subjects with idiopathic pulmonary fibrosis treated with inhaled interferon-gamma. ERJ Open Res. 2017;3(3):00008. doi: 10.1183/23120541.00008-2017.
    1. Wang K, Wang J, Wei F, Zhao N, Yang F, Ren X. Expression of TLR4 in Non-Small Cell Lung Cancer Is Associated with PD-L1 and Poor Prognosis in Patients Receiving Pulmonectomy. Front Immunol. 2017;8:456. doi: 10.3389/fimmu.2017.00456.
    1. Greten FR, Eckmann L, Greten TF, Park JM, Li Z-W, Egan LJ, et al. IKKβ Links Inflammation and Tumorigenesis in a Mouse Model of Colitis-Associated Cancer. Cell. 2004;118:285–296. doi: 10.1016/j.cell.2004.07.013.
    1. Jin C, Lagoudas GK, Zhao C, Bullman S, Bhutkar A, Hu B, et al. Commensal Microbiota Promote Lung Cancer Development via gammadelta T Cells. CELL. 2019;176(5):998–1013. doi: 10.1016/j.cell.2018.12.040.
    1. Costello EK, Stagaman K, Dethlefsen L, Bohannan BJM, Relman DA. The application of ecological theory toward an understanding of the human microbiome. Science. 2012;336:1255–1262. doi: 10.1126/science.1224203.
    1. Li D, Chen H, Zhao J, Zhang H, Chen W. Potential functions of the gastrointestinal microbiome inhabiting the length of the rat digest tract. Int J Mol Sci. 2019;20(5):1232. doi: 10.3390/ijms20051232.
    1. Ionescu AA, Nixon LS, Luzio S, Lewis-Jenkins V, Evans WD, Stone MD, et al. Pulmonary function, body composition, and protein catabolism in adults with cystic fibrosis. Am J Respir Crit Care Med. 2002;165:495–500. doi: 10.1164/ajrccm.165.4.2104065.

Source: PubMed

3
Abonneren