Disease-associated gut microbiome and metabolome changes in patients with chronic obstructive pulmonary disease

Kate L Bowerman, Saima Firdous Rehman, Annalicia Vaughan, Nancy Lachner, Kurtis F Budden, Richard Y Kim, David L A Wood, Shaan L Gellatly, Shakti D Shukla, Lisa G Wood, Ian A Yang, Peter A Wark, Philip Hugenholtz, Philip M Hansbro, Kate L Bowerman, Saima Firdous Rehman, Annalicia Vaughan, Nancy Lachner, Kurtis F Budden, Richard Y Kim, David L A Wood, Shaan L Gellatly, Shakti D Shukla, Lisa G Wood, Ian A Yang, Peter A Wark, Philip Hugenholtz, Philip M Hansbro

Abstract

Chronic obstructive pulmonary disease (COPD) is the third commonest cause of death globally, and manifests as a progressive inflammatory lung disease with no curative treatment. The lung microbiome contributes to COPD progression, but the function of the gut microbiome remains unclear. Here we examine the faecal microbiome and metabolome of COPD patients and healthy controls, finding 146 bacterial species differing between the two groups. Several species, including Streptococcus sp000187445, Streptococcus vestibularis and multiple members of the family Lachnospiraceae, also correlate with reduced lung function. Untargeted metabolomics identifies a COPD signature comprising 46% lipid, 20% xenobiotic and 20% amino acid related metabolites. Furthermore, we describe a disease-associated network connecting Streptococcus parasanguinis_B with COPD-associated metabolites, including N-acetylglutamate and its analogue N-carbamoylglutamate. While correlative, our results suggest that the faecal microbiome and metabolome of COPD patients are distinct from those of healthy individuals, and may thus aid in the search for biomarkers for COPD.

Conflict of interest statement

P.H. is a co-founder of Microba Life Sciences Limited, and D.L.A.W. is currently an employee of Microba. The remaining authors declare no competing interests.

Figures

Fig. 1. Faecal microbiota of COPD patients…
Fig. 1. Faecal microbiota of COPD patients (n = 28) can be distinguished from that of healthy individuals (n = 29) using 16S rRNA gene amplicon sequencing.
a Principal component (PC) analysis undertaken at the sequence variant level using read counts transformed using log-cumulative-sum scaling. b Multivariate sparse partial least-squares discriminant analysis (sPLS-DA) of read counts transformed using log-cumulative-sum scaling at the sequence variant level. c Sequence variants contributing to separation along with component 1 of sPLS-DA from b. Bar length indicates loading coefficient weight of selected genomes, ranked by importance, bottom to top; bar colour indicates the group in which the sequence variant has the highest median abundance, red = COPD, blue = healthy. d Heatmap of read counts transformed using log-cumulative-sum scaling of discriminatory sequence variants identified along with component 1 of sPLS-DA from (b).
Fig. 2. Metagenomic sequencing-based exploration of COPD-associated…
Fig. 2. Metagenomic sequencing-based exploration of COPD-associated (n = 28) faecal microbiomes supports distinction from those of healthy individuals (n = 29).
a Multivariate sparse partial least-squares discriminant analysis (sPLS-DA) of read-mapping-based relative abundance at the genome level of the faecal microbiome, filtered for genomes with minimum 0.05% relative abundance in at least one sample. b Genomes contributing to separation along with component 1 of sPLS-DA from (a). Bar length indicates loading coefficient weight of selected genomes, ranked by importance, bottom to top; bar colour indicates the group in which the genome has the highest median abundance, red = COPD, blue = healthy. c Heatmap of discriminatory genomes along component 1 of sPLS-DA from (a). Data are centred with log-ratio-transformed relative abundance.
Fig. 3. Correlation of members of the…
Fig. 3. Correlation of members of the faecal microbiome with lung function.
Spearman’s rho calculated between mapping-based read counts per genome and phenotypic scores. Genomes included are those from Supplementary Data 11, with enrichment in either COPD or healthy samples indicated by the coloured bar along the top of the heatmap. White stars within heatmap boxes indicate significant results (*p < 0.05; **p < 0.01, Student’s t test (two-sided), Benjamini–Hochberg adjustment for multiple comparisons. Exact p values are provided in Supplementary Data 34). Genome abundances were centred with log-ratio transformation prior to analysis. FVC forced vital capacity, FEV forced expiratory volume, WBC white blood cell, RBC red blood cell, MCV mean corpuscular volume, MCH mean corpuscular haemoglobin, RDW red cell distribution width. COPD: n = 28; healthy: n = 29.
Fig. 4. Faecal metabolome of COPD patients…
Fig. 4. Faecal metabolome of COPD patients (n = 28) is distinguished from that of healthy individuals (n = 29) using a multi-omic analysis.
a DIABLO sample plot demonstrating discrimination between COPD and healthy samples based on microbiome data. b Genomes contributing to separation along with component 1 of (a). Bar length indicates loading coefficient weight of selected genomes, ranked by importance, bottom to top; bar colour indicates the group in which the sequence variant has the highest median abundance, red = COPD, blue = healthy. Microbiome data are centred log-ratio-transformed relative abundance, filtered for genomes with minimum 0.05% relative abundance in at least ten samples. c DIABLO sample plot demonstrating discrimination between groups based on metabolomics data. d Metabolites contributing to separation along with component 1 of (c). Metabolome data are log-transformed median-scaled values with missing values imputed using the minimum value for each compound, filtered for metabolites returning measurements in at least ten samples.
Fig. 5. COPD-associated species correlate with metabolites…
Fig. 5. COPD-associated species correlate with metabolites differentiating COPD (n = 28) and healthy (n = 29) individuals.
Species and metabolites included are those identified as significantly differential between COPD and healthy samples, including age, sex and BMI within the relevant models (Supplementary Data 7 and 21). Enrichment in either group indicated by coloured bars to the left and top of the plot. Significant correlations denoted by white stars (*p < 0.05; **p < 0.01, Student’s t test (two-sided), Benjamini–Hochberg adjustment for multiple comparisons. Exact p values are provided in Supplementary Data 35). Higher taxonomy of species (order, family) and super pathway of metabolites are indicated by coloured bars.
Fig. 6. Integration of faecal microbiomes and…
Fig. 6. Integration of faecal microbiomes and metabolomes identifies a COPD-associated network.
ac Integration of microbiome and metabolome datasets using the software DIABLO produced association networks showing correlations between bacterial species and metabolites. A positive correlation between nodes is indicated by red connecting lines, negative correlation by blue. Species and metabolites enriched in COPD or healthy samples are denoted by solid or dashed borders, respectively. Black borders indicate significance in the linear model adjusted for age, sex and BMI (p < 0.05, Wald test (two-sided) with Benjamini–Hochberg adjustment for multiple comparisons, Supplementary Data 7 and 21) and grey borders indicate selection by MixOmics as discriminatory along with component 1 of Fig. 4a, b. All metabolites significant within the linear model were also selected by MixOmics. The abundance of metabolites (log-transformed median scaled) significant in the linear model provided as boxplots adjacent to the relevant nodes. Each box centres on the median, with lower and upper bounds representing the first and third quartile (25th and 75th percentile), respectively. Whiskers extend 1.5 times the interquartile range from the outer bounds. Microbiome data filtered for genomes with minimum of 0.05% relative abundance in ≥10 samples. Metabolome data filtered for metabolites returning measurements in ≥10 samples. Microbiome data are centred log-ratio- transformed relative abundance. Metabolomics data are log-transformed median-scaled values with missing values imputed using a minimum value for each compound. COPD: n = 28; healthy: n = 29.
Fig. 7. Association of gut microbiome members…
Fig. 7. Association of gut microbiome members with COPD replicate in an independent cohort.
a Multivariate sparse partial least-squares discriminant analysis (sPLS-DA) of read-mapping-based relative abundance at the genome level of the faecal microbiome, filtered for genomes with minimum 0.05% relative abundance in at least one sample. b Genomes contributing to separation along with component 1 of sPLS-DA from (a). Bar length indicates loading coefficient weight of selected genomes, ranked by importance, bottom to top; bar colour indicates the group in which the genome has the highest median abundance, red = COPD, blue = healthy. Genomes marked with * are those within the discriminatory signature defined for the study cohort (Fig. 2), # indicates genomes associated with clinical phenotypes (Fig. 3) and ^ indicates genomes within the disease-associated network (Fig. 6). COPD: n = 16; healthy: n = 22.

References

    1. Rabe KF, Watz H. Chronic obstructive pulmonary disease. Lancet. 2017;389:1931–1940. doi: 10.1016/S0140-6736(17)31222-9.
    1. Keely S, Talley NJ, Hansbro PM. Pulmonary-intestinal cross-talk in mucosal inflammatory disease. Mucosal Immunol. 2012;5:7–18. doi: 10.1038/mi.2011.55.
    1. Naghavi M, et al. Global, regional, and national age-sex specific mortality for 264 causes of death, 1980-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017;390:1151–1210. doi: 10.1016/S0140-6736(17)32152-9.
    1. Mannino DM, Buist AS. Global burden of COPD: risk factors, prevalence, and future trends. Lancet. 2007;370:765–773. doi: 10.1016/S0140-6736(07)61380-4.
    1. Fricker M, et al. Chronic cigarette smoke exposure induces systemic hypoxia that drives intestinal dysfunction. JCI Insight. 2018;3:e94040. doi: 10.1172/jci.insight.94040.
    1. Løkke A, Lange P, Scharling H, Fabricius P, Vestbo J. Developing COPD: a 25 year follow up study of the general population. Thorax. 2006;61:935–939. doi: 10.1136/thx.2006.062802.
    1. Jones B, et al. Animal models of COPD: what do they tell us? Respirology. 2017;22:21–32. doi: 10.1111/resp.12908.
    1. Yang I. A., Clarke M. S., Sim E. H. A., Fong K. M. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst. Rev. (2012).
    1. Calverley PMA, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N. Engl. J. Med. 2007;356:775–789. doi: 10.1056/NEJMoa063070.
    1. Wedzicha JA, Seemungal TAR. COPD exacerbations: defining their cause and prevention. Lancet. 2007;370:786–796. doi: 10.1016/S0140-6736(07)61382-8.
    1. Leung JM, et al. The role of acute and chronic respiratory colonization and infections in the pathogenesis of COPD. Respirology. 2017;22:634–650. doi: 10.1111/resp.13032.
    1. Wilkinson TMA, et al. A prospective, observational cohort study of the seasonal dynamics of airway pathogens in the aetiology of exacerbations in COPD. Thorax. 2017;72:919–927. doi: 10.1136/thoraxjnl-2016-209023.
    1. Wang Z, et al. Airway host-microbiome interactions in chronic obstructive pulmonary disease. Respir. Res. 2019;20:113. doi: 10.1186/s12931-019-1085-z.
    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:e47305. doi: 10.1371/journal.pone.0047305.
    1. Ren L, et al. Transcriptionally active lung microbiome and its association with bacterial biomass and host inflammatory status. mSystems. 2018;3:e00199–00118. doi: 10.1128/mSystems.00199-18.
    1. Sze MA, et al. Host response to the lung microbiome in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2015;192:438–445. doi: 10.1164/rccm.201502-0223OC.
    1. Huffnagle GB, Dickson RP, Lukacs NW. The respiratory tract microbiome and lung inflammation: a two-way street. Mucosal Immunol. 2016;10:299. doi: 10.1038/mi.2016.108.
    1. Budden KF, et al. Functional effects of the microbiota in chronic respiratory disease. Lancet Respir. Med. 2019;7:907–920. doi: 10.1016/S2213-2600(18)30510-1.
    1. Budden KF, et al. Emerging pathogenic links between microbiota and the gut–lung axis. Nat. Rev. Microbiol. 2016;15:55. doi: 10.1038/nrmicro.2016.142.
    1. Arrieta MC, et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 2015;7:307ra152. doi: 10.1126/scitranslmed.aab2271.
    1. Trompette A, et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014;20:159. doi: 10.1038/nm.3444.
    1. Thorburn AN, et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat. Commun. 2015;6:7320. doi: 10.1038/ncomms8320.
    1. Trompette A, et al. Dietary fiber confers protection against flu by shaping Ly6c− patrolling monocyte hematopoiesis and CD8+ T cell metabolism. Immunity. 2018;48:992–1005.e1008. doi: 10.1016/j.immuni.2018.04.022.
    1. Ekbom A, Brandt L, Granath F, Löfdahl C-G, Egesten A. Increased risk of both ulcerative colitis and Crohn’s disease in a population suffering from COPD. Lung. 2008;186:167–172. doi: 10.1007/s00408-008-9080-z.
    1. Mateer SW, et al. Potential mechanisms regulating pulmonary pathology in inflammatory bowel disease. J. Leukoc. Biol. 2015;98:727–737. doi: 10.1189/jlb.3RU1114-563R.
    1. Vaughan A, Frazer ZA, Hansbro PM, Yang IA. COPD and the gut-lung axis: the therapeutic potential of fibre. J. Thorac. Dis. 2019;11:S2173–S2180. doi: 10.21037/jtd.2019.10.40.
    1. Chunxi L, Haiyue L, Yanxia L, Jianbing P, Jin S. The gut microbiota and respiratory diseases: new evidence. J. Immunol. Res. 2020;2020:2340670–2340670. doi: 10.1155/2020/2340670.
    1. Ferretti P, et al. Mother-to-infant microbial transmission from different body sites shapes the developing infant gut microbiome. Cell Host Microbe. 2018;24:133–145.e135. doi: 10.1016/j.chom.2018.06.005.
    1. Franzosa EA, et al. Identifying personal microbiomes using metagenomic codes. Proc. Natl Acad. Sci. USA. 2015;112:E2930–E2938. doi: 10.1073/pnas.1423854112.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Rohart F, Gautier B, Singh A, Lê, Cao K-A. mixOmics: an R package for ‘omics feature selection and multiple data integration. PLoS Comput. Biol. 2017;13:e1005752. doi: 10.1371/journal.pcbi.1005752.
    1. McCabe RM, Donkersloot JA. Adherence of Veillonella species mediated by extracellular glucosyltransferase from Streptococcus salivarius. Infect. Immun. 1977;18:726–734. doi: 10.1128/IAI.18.3.726-734.1977.
    1. McNab R, et al. Cell wall-anchored CshA polypeptide (259 kilodaltons) in Streptococcus gordonii forms surface fibrils that confer hydrophobic and adhesive properties. J. Bacteriol. 1999;181:3087–3095. doi: 10.1128/JB.181.10.3087-3095.1999.
    1. Kolachala VL, et al. Epithelial-derived fibronectin expression, signaling, and function in intestinal inflammation. J. Biol. Chem. 2007;282:32965–32973. doi: 10.1074/jbc.M704388200.
    1. Dammeier J, Brauchle M, Falk W, Grotendorst GR, Werner S. Connective tissue growth factor: a novel regulator of mucosal repair and fibrosis in inflammatory bowel disease? Int J. Biochem. Cell Biol. 1998;30:909–922. doi: 10.1016/S1357-2725(98)00046-6.
    1. Annoni R, et al. Extracellular matrix composition in COPD. Eur. Respir. J. 2012;40:1362–1373. doi: 10.1183/09031936.00192611.
    1. Liu G, et al. Fibulin-1 regulates the pathogenesis of tissue remodeling in respiratory diseases. JCI Insight. 2016;1:e86380.
    1. Bensing BA, Seepersaud R, Yen YT, Sullam PM. Selective transport by SecA2: an expanding family of customized motor proteins. Biochim. Biophys. Acta. 2014;1843:1674–1686. doi: 10.1016/j.bbamcr.2013.10.019.
    1. Eijkelkamp BA, McDevitt CA, Kitten T. Manganese uptake and streptococcal virulence. Biometals. 2015;28:491–508. doi: 10.1007/s10534-015-9826-z.
    1. Nayfach S, Shi ZJ, Seshadri R, Pollard KS, Kyrpides NC. New insights from uncultivated genomes of the global human gut microbiome. Nature. 2019;568:505–510. doi: 10.1038/s41586-019-1058-x.
    1. Mingrone G, Castagneto-Gissey L, Macé K. Use of dicarboxylic acids in type 2 diabetes. Br. J. Clin. Pharm. 2013;75:671–676. doi: 10.1111/j.1365-2125.2012.04177.x.
    1. Miura Y. The biological significance of ω-oxidation of fatty acids. Proc. Jpn Acad. Ser. B Phys. Biol. Sci. 2013;89:370–382. doi: 10.2183/pjab.89.370.
    1. Parke D, Garcia MA, Ornston LN. Cloning and genetic characterization of dca genes required for β-oxidation of straight-chain dicarboxylic acids in Acinetobacter sp. strain ADP1. Appl. Environ. Microbiol. 2001;67:4817–4827. doi: 10.1128/AEM.67.10.4817-4827.2001.
    1. Park HS, et al. Statins increase mitochondrial and peroxisomal fatty acid oxidation in the liver and prevent non-alcoholic steatohepatitis in mice. Diabetes Metab. J. 2016;40:376–385. doi: 10.4093/dmj.2016.40.5.376.
    1. Jacobs JP, et al. A disease-associated microbial and metabolomics state in relatives of pediatric inflammatory bowel disease patients. Cell. Mol. Gastroenterol. Hepatol. 2016;2:750–766. doi: 10.1016/j.jcmgh.2016.06.004.
    1. Ma W, et al. Advances in cadaverine bacterial production and its applications. Engineering. 2017;3:308–317. doi: 10.1016/J.ENG.2017.03.012.
    1. Shi X, Yao D, Chen C. Identification of N-acetyltaurine as a novel metabolite of ethanol through metabolomics-guided biochemical analysis. J. Biol. Chem. 2012;287:6336–6349. doi: 10.1074/jbc.M111.312199.
    1. Landa M, Burns AS, Roth SJ, Moran MA. Bacterial transcriptome remodeling during sequential co-culture with a marine dinoflagellate and diatom. ISME J. 2017;11:2677–2690. doi: 10.1038/ismej.2017.117.
    1. Hession AO, Esrey EG, Croes RA, Maxwell CA. N-Acetylglutamate and N-Acetylaspartate in soybeans (Glycine max L.), maize (Zea maize L.), and other foodstuffs. J. Agric. Food Chem. 2008;56:9121–9126. doi: 10.1021/jf801523c.
    1. Caldovic L, Tuchman M. N-acetylglutamate and its changing role through evolution. Biochem. J. 2003;372:279–290. doi: 10.1042/bj20030002.
    1. Sankaranarayanan K, et al. Gut microbiome diversity among Cheyenne and Arapaho individuals from western Oklahoma. Curr. Biol. 2015;25:3161–3169. doi: 10.1016/j.cub.2015.10.060.
    1. Henriksen CM, Nielsen J, Villadsen J. Cyclization of alpha-aminoadipic acid into the delta-lactam 6-oxo-piperidine-2-carboxylic acid by Penicillium chrysogenum. J. Antibiot. 1998;51:99–106. doi: 10.7164/antibiotics.51.99.
    1. Wu G, Knabe DA, Kim SW. Arginine nutrition in neonatal pigs. J. Nutr. 2004;134:2783S–2790S. doi: 10.1093/jn/134.10.2783S.
    1. Cao W, et al. Dietary arginine and N-carbamylglutamate supplementation enhances the antioxidant statuses of the liver and plasma against oxidative stress in rats. Food Funct. 2016;7:2303–2311. doi: 10.1039/C5FO01194A.
    1. Wu X, Zhang Y, Liu Z, Li TJ, Yin YL. Effects of oral supplementation with glutamate or combination of glutamate and N-carbamylglutamate on intestinal mucosa morphology and epithelium cell proliferation in weanling piglets. J. Anim. Sci. 2012;90:337–339. doi: 10.2527/jas.53752.
    1. Kodani S, Imoto A, Mitsutani A, Murakami M. Isolation and identification of the antialgal compound, harmane (1-methyl-β-carboline), produced by the algicidal bacterium, Pseudomonas sp. K44-1. J. Appl. Phycol. 2002;14:109–114. doi: 10.1023/A:1019533414018.
    1. Arshad N, Zitterl-Eglseer K, Hasnain S, Hess M. Effect of Peganum harmala or its β-carboline alkaloids on certain antibiotic resistant strains of bacteria and protozoa from poultry. Phytother. Res. 2008;22:1533–1538. doi: 10.1002/ptr.2528.
    1. Jakobsen H, et al. The alkaloid compound harmane increases the lifespan of Caenorhabditis elegans during bacterial infection, by modulating the nematode’s innate immune response. PLoS ONE. 2013;8:e60519. doi: 10.1371/journal.pone.0060519.
    1. Biedermann L, et al. Smoking cessation induces profound changes in the composition of the intestinal microbiota in humans. PLoS ONE. 2013;8:e59260. doi: 10.1371/journal.pone.0059260.
    1. Shanahan ER, et al. Influence of cigarette smoking on the human duodenal mucosa-associated microbiota. Microbiome. 2018;6:150. doi: 10.1186/s40168-018-0531-3.
    1. Chan K-G, et al. Genome anatomy of Streptococcus parasanguinis strain C1A, isolated from a patient with acute exacerbation of chronic obstructive pulmonary disease, reveals unusual genomic features. Genome Announc. 2015;3:e00541–00515.
    1. Schmidt TS, et al. Extensive transmission of microbes along the gastrointestinal tract. eLife. 2019;8:e42693. doi: 10.7554/eLife.42693.
    1. Cvejic L, et al. Laryngeal penetration and aspiration in individuals with stable COPD. Respirology. 2011;16:269–275. doi: 10.1111/j.1440-1843.2010.01875.x.
    1. Igartua C, et al. Host genetic variation in mucosal immunity pathways influences the upper airway microbiome. Microbiome. 2017;5:16. doi: 10.1186/s40168-016-0227-5.
    1. Wacklin P, et al. Secretor genotype (FUT2 gene) is strongly associated with the composition of bifidobacteria in the human intestine. PLoS ONE. 2011;6:e20113. doi: 10.1371/journal.pone.0020113.
    1. Gomez A, et al. Host genetic control of the oral microbiome in health and disease. Cell Host Microbe. 2017;22:269–278.e263. doi: 10.1016/j.chom.2017.08.013.
    1. Zhang J, et al. A phylo-functional core of gut microbiota in healthy young Chinese cohorts across lifestyles, geography and ethnicities. ISME J. 2015;9:1979–1990. doi: 10.1038/ismej.2015.11.
    1. Sekelja M, Berget I, Næs T, Rudi K. Unveiling an abundant core microbiota in the human adult colon by a phylogroup-independent searching approach. ISME J. 2011;5:519–531. doi: 10.1038/ismej.2010.129.
    1. Zuo K, et al. Disordered gut microbiota and alterations in metabolic patterns are associated with atrial fibrillation. Gigascience. 2019;8:giz058. doi: 10.1093/gigascience/giz058.
    1. Gomes AC, Hoffmann C, Mota JF. The human gut microbiota: metabolism and perspective in obesity. Gut Microbes. 2018;9:308–325.
    1. Schirmer M, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167:1125–1136.e1128. doi: 10.1016/j.cell.2016.10.020.
    1. Hoenderdos K, Condliffe A. The neutrophil in chronic obstructive pulmonary disease. Am. J. Respir. Cell Mol. Biol. 2013;48:531–539. doi: 10.1165/rcmb.2012-0492TR.
    1. Fritz JH. Arginine cools the inflamed gut. Infect. Immun. 2013;81:3500–3502. doi: 10.1128/IAI.00789-13.
    1. Maarsingh H, Zaagsma J, Meurs H. Arginine homeostasis in allergic asthma. Eur. J. Pharm. 2008;585:375–384. doi: 10.1016/j.ejphar.2008.02.096.
    1. Halper-Stromberg E, et al. Bronchoalveolar lavage fluid from COPD patients reveals more compounds associated with disease than matched plasma. Metabolites. 2019;9:157. doi: 10.3390/metabo9080157.
    1. Menni C, et al. Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women. Sci. Rep. 2017;7:11079. doi: 10.1038/s41598-017-10382-2.
    1. Wanders RJA, Komen J, Kemp S. Fatty acid omega-oxidation as a rescue pathway for fatty acid oxidation disorders in humans. FEBS J. 2011;278:182–194. doi: 10.1111/j.1742-4658.2010.07947.x.
    1. Yu B, et al. Metabolomics identifies novel blood biomarkers of pulmonary function and COPD in the general population. Metabolites. 2019;9:61. doi: 10.3390/metabo9040061.
    1. Agarwal AR, et al. Systemic immuno-metabolic alterations in chronic obstructive pulmonary disease (COPD) Respir. Res. 2019;20:171. doi: 10.1186/s12931-019-1139-2.
    1. Naz S, et al. Metabolomics analysis identifies sex-associated metabotypes of oxidative stress and the autotaxin–lysoPA axis in COPD. Eur. Respir. J. 2017;49:1602322. doi: 10.1183/13993003.02322-2016.
    1. Lee T, et al. Oral versus intravenous iron replacement therapy distinctly alters the gut microbiota and metabolome in patients with IBD. Gut. 2017;66:863–871. doi: 10.1136/gutjnl-2015-309940.
    1. Franzosa EA, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019;4:293–305. doi: 10.1038/s41564-018-0306-4.
    1. Maslowski KM, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–1286. doi: 10.1038/nature08530.
    1. McLoughlin R, et al. Soluble fibre supplementation with and without a probiotic in adults with asthma: a 7-day randomised, double blind, three way cross-over trial. EBioMedicine. 2019;46:473–485. doi: 10.1016/j.ebiom.2019.07.048.
    1. Halnes, I. et al. Soluble fibre meal challenge reduces airway inflammation and expression of GPR43 and GPR41 in asthma. Nutrients9, 57 (2017).
    1. Beckett EL, et al. A new short-term mouse model of chronic obstructive pulmonary disease identifies a role for mast cell tryptase in pathogenesis. J. Allergy Clin. Immunol. 2013;131:752–762. doi: 10.1016/j.jaci.2012.11.053.
    1. Hansbro PM, et al. Importance of mast cell Prss31/transmembrane tryptase/tryptase-gamma in lung function and experimental chronic obstructive pulmonary disease and colitis. J. Biol. Chem. 2014;289:18214–18227. doi: 10.1074/jbc.M114.548594.
    1. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnetjournal17, 10–12 (2011).
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120. doi: 10.1093/bioinformatics/btu170.
    1. Caporaso JG, et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods. 2010;7:335–336. doi: 10.1038/nmeth.f.303.
    1. Callahan BJ, et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods. 2016;13:581. doi: 10.1038/nmeth.3869.
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J. Mol. Biol. 1990;215:403–410. doi: 10.1016/S0022-2836(05)80360-2.
    1. Quast C, et al. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2013;41:D590–D596. doi: 10.1093/nar/gks1219.
    1. Paulson JN, Stine OC, Bravo HC, Pop M. Differential abundance analysis for microbial marker-gene surveys. Nat. Methods. 2013;10:1200–1202. doi: 10.1038/nmeth.2658.
    1. Oksanen J. et al. vegan: Community Ecology Package. R Package Version 23-1. (2015).
    1. Kolde R. pheatmap: Pretty Heatmaps. R Package Version 107. (2015).
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Nurk S, Meleshko D, Korobeynikov A, Pevzner PA. metaSPAdes: a new versatile metagenomic assembler. Genome Res. 2017;27:824–834. doi: 10.1101/gr.213959.116.
    1. Kang DD, Froula J, Egan R, Wang Z. MetaBAT, an efficient tool for accurately reconstructing single genomes from complex microbial communities. PeerJ. 2015;3:e1165. doi: 10.7717/peerj.1165.
    1. Parks DH, Imelfort M, Skennerton CT, Hugenholtz P, Tyson GW. CheckM: assessing the quality of microbial genomes recovered from isolates, single cells, and metagenomes. Genome Res. 2015;25:1043–1055. doi: 10.1101/gr.186072.114.
    1. Olm MR, Brown CT, Brooks B, Banfield JF. dRep: a tool for fast and accurate genomic comparisons that enables improved genome recovery from metagenomes through de-replication. ISME J. 2017;11:2864–2868. doi: 10.1038/ismej.2017.126.
    1. Parks DH, et al. A standardized bacterial taxonomy based on genome phylogeny substantially revises the tree of life. Nat. Biotechnol. 2018;36:996. doi: 10.1038/nbt.4229.
    1. Chaumeil P-A, Mussig AJ, Hugenholtz P, Parks DH. GTDB-Tk: a toolkit to classify genomes with the Genome Taxonomy Database. Bioinformatics. 2019;36:1925–1927.
    1. Pedersen BS, Quinlan AR. Mosdepth: quick coverage calculation for genomes and exomes. Bioinformatics. 2018;34:867–868. doi: 10.1093/bioinformatics/btx699.
    1. Revelle W. psych: Procedures for Personality and Psychological Research. R Package Version 1812. (2018).
    1. Wei T., Simko V. corrplot: Visualization of a Correlation Matrix. R Package Version 084. (2018).
    1. Hyatt D, et al. Prodigal: prokaryotic gene recognition and translation initiation site identification. BMC Bioinform. 2010;11:119. doi: 10.1186/1471-2105-11-119.
    1. Mistry J, Finn RD, Eddy SR, Bateman A, Punta M. Challenges in homology search: HMMER3 and convergent evolution of coiled-coil regions. Nucleic Acids Res. 2013;41:e121. doi: 10.1093/nar/gkt263.
    1. Yin Y, et al. dbCAN: a web resource for automated carbohydrate-active enzyme annotation. Nucleic Acids Res. 2012;40:W445–W451. doi: 10.1093/nar/gks479.
    1. Finn RD, et al. Pfam: the protein families database. Nucleic Acids Res. 2014;42:D222–D230. doi: 10.1093/nar/gkt1223.
    1. Haft DH, Selengut JD, White O. The TIGRFAMs database of protein families. Nucleic Acids Res. 2003;31:371–373. doi: 10.1093/nar/gkg128.
    1. Suzek BE, Huang HZ, McGarvey P, Mazumder R, Wu CH. UniRef: comprehensive and non-redundant UniProt reference clusters. Bioinformatics. 2007;23:1282–1288. doi: 10.1093/bioinformatics/btm098.
    1. Evans, A. M. et al. High resolution mass spectrometry improves data quantity and quality as compared to unit mass resolution mass spectrometry in high-throughput profiling metabolomics. Metabolomics4, 1000132 (2014).
    1. DeHaven CD, Evans AM, Dai H, Lawton KA. Organization of GC/MS and LC/MS metabolomics data into chemical libraries. J. Cheminform. 2010;2:9. doi: 10.1186/1758-2946-2-9.
    1. De Livera AM, Olshansky G, Simpson JA, Creek DJ. NormalizeMets: assessing, selecting and implementing statistical methods for normalizing metabolomics data. Metabolomics. 2018;14:54. doi: 10.1007/s11306-018-1347-7.

Source: PubMed

3
Tilaa