Altered fecal microbiome and metabolome in adult patients with non-cystic fibrosis bronchiectasis

Wen-Wen Wang, Bei Mao, Yang Liu, Shu-Yi Gu, Hai-Wen Lu, Jiu-Wu Bai, Shuo Liang, Jia-Wei Yang, Jian-Xiong Li, Xiao Su, Hai-Yang Hu, Chen Wang, Jin-Fu Xu, Wen-Wen Wang, Bei Mao, Yang Liu, Shu-Yi Gu, Hai-Wen Lu, Jiu-Wu Bai, Shuo Liang, Jia-Wei Yang, Jian-Xiong Li, Xiao Su, Hai-Yang Hu, Chen Wang, Jin-Fu Xu

Abstract

Background: Emerging experimental and epidemiological evidence highlights a crucial cross-talk between the intestinal flora and the lungs, termed the "gut-lung axis". However, the function of the gut microbiota in bronchiectasis remains undefined. In this study, we aimed to perform a multi-omics-based approach to identify the gut microbiome and metabolic profiles in patients with bronchiectasis.

Methods: Fecal samples collected from non-CF bronchiectasis patients (BE group, n = 61) and healthy volunteers (HC group, n = 37) were analyzed by 16 S ribosomal RNA (rRNA) sequencing. The BE group was divided into two groups based on their clinical status: acute exacerbation (AE group, n = 31) and stable phase (SP group, n = 30). Further, metabolome (lipid chromatography-mass spectrometry, LC-MS) analyses were conducted in randomly selected patients (n = 29) and healthy volunteers (n = 31).

Results: Decreased fecal microbial diversity and differential microbial and metabolic compositions were observed in bronchiectasis patients. Correlation analyses indicated associations between the differential genera and clinical parameters such as bronchiectasis severity index (BSI). Disease-associated gut microbiota was screened out, with eight genera exhibited high accuracy in distinguishing SP patients from HCs in the discovery cohort and validation cohort using a random forest model. Further correlation networks were applied to illustrate the relations connecting disease-associated genera and metabolites.

Conclusion: The study uncovered the relationships among the decreased fecal microbial diversity, differential microbial and metabolic compositions in bronchiectasis patients by performing a multi-omics-based approach. It is the first study to characterize the gut microbiome and metabolome in bronchiectasis, and to uncover the gut microbiota's potentiality as biomarkers for bronchiectasis.

Trial registration: This study is registered with ClinicalTrials.gov, number NCT04490447.

Keywords: Biomarkers; Bronchiectasis; Gut microbiome; Metabolomics.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Flowchart. One hundred forty-five fecal samples were collected from Shanghai Pulmonary Hospital. Samples were randomly allocated to the discovery cohort and validation cohort. In the discovery cohort, 30, 31, and 37 fecal samples collected from the SP, AE, and HC groups, respectively, were assigned to 16 S rRNA gene sequencing. Thirty-one and 29 samples from HC and SP groups were assigned to LC-MS analysis
Fig. 2
Fig. 2
Decreased fecal microbial diversity in patients with bronchiectasis. a Venn diagram displaying the identified OTUs in three groups. α diversity estimated by Chao1 index (b) and observed species index (c). d β diversity calculated among the three groups using the unweighted UniFrac by PCoA. *P < 0.05, **P < 0.01, ***P < 0.001
Fig. 3
Fig. 3
Distinct phylogenetic profiling of gut microbiota among the three groups. The relative proportions of dominant taxa at the phylum level (a) and genus level (b). The top 20 variant genera between HC and SP (c), HC and AE (d). e Correlations between variant genera and clinical parameters were evaluated by Spearman’s correlation analyses. +P < 0.05, *P < 0.01
Fig. 4
Fig. 4
Identification and validation of bronchiectasis patients based on gut microbial genera. a LDA analyses identify differential taxa between HC and SP with LDA scores (log10) > 2. b Taxonomic cladogram indicating the phylogenetic distribution of gut microbiota in HC and SP. c MDA values of candidate genera are listed in descending order. The ROC curve was generated by the RF model using eight genera in the discovery cohort (d) and validation cohort (e)
Fig. 5
Fig. 5
Differential fecal metabolomic features in HC and SP groups. a OPLS-DA score plot. b OPLS-DA validation plot intercepts: R2Y = (0.0, 0.7233), Q2 = (0.0, – 0.1979). c Bubble plot displaying the significantly enriched pathways. d Correlation network of the 8 genera and metabolites with P < 0.01. The size of each node is proportional to the quantity of related metabolites and the thickness of the line is proportional to the correlation coefficient. Lines between nodes and triangles indicate positive correlations (red) or negative correlations (blue)

References

    1. Martinez-Garcia MA, Polverino E, Aksamit T. Bronchiectasis and Chronic Airway Disease: It Is Not Just About Asthma and COPD. Chest. 2018;154(4):737–9.
    1. McShane PJ, Naureckas ET, Tino G, Strek ME. Non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2013;188(6):647–56.
    1. Swenson C, Schraufnagel D, Sadikot R. What is Bronchiectasis? Am J Respir Crit Care Med. 2017;195(8):P15-P6.
    1. Diel R, Chalmers JD, Rabe KF, Nienhaus A, Loddenkemper R, Ringshausen FC. Economic burden of bronchiectasis in Germany. Eur Respir J. 2019;53(2).
    1. Lin JL, Xu JF, Qu JM. Bronchiectasis in China. Ann Am Thorac Soc. 2016;13(5):609–16.
    1. Flume PA, Chalmers JD, Olivier KN. Advances in bronchiectasis: endotyping, genetics, microbiome, and disease heterogeneity. Lancet. 2018;392(10150):880–90.
    1. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50.
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16(6):341–52.
    1. Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71.
    1. McCarville JL, Chen GY, Cuevas VD, Troha K, Ayres JS. Microbiota Metabolites in Health and Disease. Annu Rev Immunol. 2020;38:147–70.
    1. Schuijt TJ, Lankelma JM, Scicluna BP, de Sousa e Melo F, Roelofs JJ, de Boer JD, et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut. 2016;65(4):575–83.
    1. Barcik W, Boutin RCT, Sokolowska M, Finlay BB. The Role of Lung and Gut Microbiota in the Pathology of Asthma. Immunity. 2020;52(2):241–55.
    1. Budden KF, Gellatly SL, Wood DL, Cooper MA, Morrison M, Hugenholtz P, et al. Emerging pathogenic links between microbiota and the gut-lung axis. Nat Rev Microbiol. 2017;15(1):55–63.
    1. Trompette A, Gollwitzer ES, Yadava K, Sichelstiel AK, Sprenger N, Ngom-Bru C, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med. 2014;20(2):159–66.
    1. Stokholm J, Blaser MJ, Thorsen J, Rasmussen MA, Waage J, Vinding RK, et al. Maturation of the gut microbiome and risk of asthma in childhood. Nat Commun. 2018;9(1):141.
    1. Fujimura KE, Lynch SV. Microbiota in allergy and asthma and the emerging relationship with the gut microbiome. Cell Host Microbe. 2015;17(5):592–602.
    1. Hoen AG, Li J, Moulton LA, O’Toole GA, Housman ML, Koestler DC, et al. Associations between Gut Microbial Colonization in Early Life and Respiratory Outcomes in Cystic Fibrosis. J Pediatr. 2015;167(1):138–47.
    1. Varraso R, Chiuve SE, Fung TT, Barr RG, Hu FB, Willett WC, et al. Alternate Healthy Eating Index 2010 and risk of chronic obstructive pulmonary disease among US women and men: prospective study. BMJ. 2015;350:h286.
    1. Bowerman KL, Rehman SF, Vaughan A, Lachner N, Budden KF, Kim RY, et al. Disease-associated gut microbiome and metabolome changes in patients with chronic obstructive pulmonary disease. Nat Commun. 2020;11(1):5886.
    1. Pasteur MC, Bilton D, Hill AT. British Thoracic Society Bronchiectasis non CFGG. British Thoracic Society guideline for non-CF bronchiectasis. Thorax. 2010;65(Suppl 1):i1–58.
    1. Scheperjans F, Aho V, Pereira PA, Koskinen K, Paulin L, Pekkonen E, et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov Disord. 2015;30(3):350–8.
    1. Hill AT, Haworth CS, Aliberti S, Barker A, Blasi F, Boersma W, et al. Pulmonary exacerbation in adults with bronchiectasis: a consensus definition for clinical research. Eur Respir J. 2017;49(6).
    1. Ren Z, Li A, Jiang J, Zhou L, Yu Z, Lu H, et al. Gut microbiome analysis as a tool towards targeted non-invasive biomarkers for early hepatocellular carcinoma. Gut. 2019;68(6):1014–23.
    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.
    1. Brown DG, Rao S, Weir TL, O’Malia J, Bazan M, Brown RJ, et al. Metabolomics and metabolic pathway networks from human colorectal cancers, adjacent mucosa, and stool. Cancer Metab. 2016;4:11.
    1. Bolnick DI, Snowberg LK, Hirsch PE, Lauber CL, Org E, Parks B, et al. Individual diet has sex-dependent effects on vertebrate gut microbiota. Nat Commun. 2014;5:4500.
    1. Yang B, Jang HJ, Chung SJ, Yoo SJ, Kim T, Kim SH, et al. Factors associated with bronchiectasis in Korea: a national database study. Ann Transl Med. 2020;8(21):1350.
    1. Yang B, Han K, Kim SH, Lee DH, Park SH, Yoo JE, et al. Being underweight increases the risk of non-cystic fibrosis bronchiectasis in the young population: a nationwide population-based study. Nutrients. 2021;13(9).
    1. Russell SL, Gold MJ, Reynolds LA, Willing BP, Dimitriu P, Thorson L, et al. Perinatal antibiotic-induced shifts in gut microbiota have differential effects on inflammatory lung diseases. J Allergy Clin Immunol. 2015;135(1):100–9.
    1. Wypych TP, Wickramasinghe LC, Marsland BJ. The influence of the microbiome on respiratory health. Nat Immunol. 2019;20(10):1279–90.
    1. Kaluza J, Harris H, Wallin A, Linden A, Wolk A. Dietary Fiber Intake and Risk of Chronic Obstructive Pulmonary Disease: A Prospective Cohort Study of Men. Epidemiology. 2018;29(2):254–60.
    1. Gauguet S, D’Ortona S, Ahnger-Pier K, Duan B, Surana NK, Lu R, et al. Intestinal Microbiota of Mice Influences Resistance to Staphylococcus aureus Pneumonia. Infect Immun. 2015;83(10):4003–14.
    1. Dumas A, Bernard L, Poquet Y, Lugo-Villarino G, Neyrolles O. The role of the lung microbiota and the gut-lung axis in respiratory infectious diseases. Cell Microbiol. 2018;20(12):e12966.
    1. Butland RJ, Cole P, Citron KM, Turner-Warwick M. Chronic bronchial suppuration and inflammatory bowel disease. Q J Med. 1981;50(197):63–75.
    1. Black H, Mendoza M, Murin S. Thoracic manifestations of inflammatory bowel disease. Chest. 2007;131(2):524–32.
    1. Shenoy MK, Fadrosh DW, Lin DL, Worodria W, Byanyima P, Musisi E, et al. Gut microbiota in HIV-pneumonia patients is related to peripheral CD4 counts, lung microbiota, and in vitro macrophage dysfunction. Microbiome. 2019;7(1):37.
    1. Chang CJ, Lin CS, Lu CC, Martel J, Ko YF, Ojcius DM, et al. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat Commun. 2015;6:7489.
    1. Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015;65(6):1331–40.
    1. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200.
    1. Akimova T, Beier UH, Liu Y, Wang L, Hancock WW. Histone/protein deacetylases and T-cell immune responses. Blood. 2012;119(11):2443–51.
    1. Chang PV, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc Natl Acad Sci U S A. 2014;111(6):2247–52.
    1. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun. 2013;4:1829.
    1. Eltzschig HK, Sitkovsky MV, Robson SC. Purinergic signaling during inflammation. N Engl J Med. 2012;367(24):2322–33.
    1. Esther CR, Jr, Turkovic L, Rosenow T, Muhlebach MS, Boucher RC, Ranganathan S, et al. Metabolomic biomarkers predictive of early structural lung disease in cystic fibrosis. Eur Respir J. 2016;48(6):1612–21.
    1. Hannun YA, Obeid LM. Sphingolipids and their metabolism in physiology and disease. Nat Rev Mol Cell Biol. 2018;19(3):175–91.
    1. Schirmer M, Garner A, Vlamakis H, Xavier RJ. Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol. 2019;17(8):497–511.
    1. Haus JM, Kashyap SR, Kasumov T, Zhang R, Kelly KR, Defronzo RA, et al. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes. 2009;58(2):337–43.
    1. Edsfeldt A, Duner P, Stahlman M, Mollet IG, Asciutto G, Grufman H, et al. Sphingolipids Contribute to Human Atherosclerotic Plaque Inflammation. Arterioscler Thromb Vasc Biol. 2016;36(6):1132–40.
    1. Pewzner-Jung Y, Tavakoli Tabazavareh S, Grassme H, Becker KA, Japtok L, Steinmann J, et al. Sphingoid long chain bases prevent lung infection by Pseudomonas aeruginosa. EMBO Mol Med. 2014;6(9):1205–14.
    1. Quinn RA, Phelan VV, Whiteson KL, Garg N, Bailey BA, Lim YW, et al. Microbial, host and xenobiotic diversity in the cystic fibrosis sputum metabolome. ISME J. 2016;10(6):1483–98.
    1. Sharman DF. Metabolism of Tryptamine and Related Compounds in the Central Nervous System. Br Med Bull. 1965;21:62–5.
    1. Williams BB, Van Benschoten AH, Cimermancic P, Donia MS, Zimmermann M, Taketani M, et al. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine. Cell Host Microbe. 2014;16(4):495–503.
    1. Mawe GM, Hoffman JM. Serotonin signalling in the gut–functions, dysfunctions and therapeutic targets. Nat Rev Gastroenterol Hepatol. 2013;10(8):473–86.
    1. Bhattarai Y, Jie S, Linden DR, Ghatak S, Mars RAT, Williams BB, et al. Bacterially Derived Tryptamine Increases Mucus Release by Activating a Host Receptor in a Mouse Model of Inflammatory Bowel Disease. iScience. 2020;23(12):101798.
    1. Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR, Till L, Grover M, et al. Gut Microbiota-Produced Tryptamine Activates an Epithelial G-Protein-Coupled Receptor to Increase Colonic Secretion. Cell Host Microbe. 2018;23(6):775–85.

Source: PubMed

3
Abonnere