Taxonomic signatures of cause-specific mortality risk in human gut microbiome

Aaro Salosensaari, Ville Laitinen, Aki S Havulinna, Guillaume Meric, Susan Cheng, Markus Perola, Liisa Valsta, Georg Alfthan, Michael Inouye, Jeramie D Watrous, Tao Long, Rodolfo A Salido, Karenina Sanders, Caitriona Brennan, Gregory C Humphrey, Jon G Sanders, Mohit Jain, Pekka Jousilahti, Veikko Salomaa, Rob Knight, Leo Lahti, Teemu Niiranen, Aaro Salosensaari, Ville Laitinen, Aki S Havulinna, Guillaume Meric, Susan Cheng, Markus Perola, Liisa Valsta, Georg Alfthan, Michael Inouye, Jeramie D Watrous, Tao Long, Rodolfo A Salido, Karenina Sanders, Caitriona Brennan, Gregory C Humphrey, Jon G Sanders, Mohit Jain, Pekka Jousilahti, Veikko Salomaa, Rob Knight, Leo Lahti, Teemu Niiranen

Abstract

The collection of fecal material and developments in sequencing technologies have enabled standardised and non-invasive gut microbiome profiling. Microbiome composition from several large cohorts have been cross-sectionally linked to various lifestyle factors and diseases. In spite of these advances, prospective associations between microbiome composition and health have remained uncharacterised due to the lack of sufficiently large and representative population cohorts with comprehensive follow-up data. Here, we analyse the long-term association between gut microbiome variation and mortality in a well-phenotyped and representative population cohort from Finland (n = 7211). We report robust taxonomic and functional microbiome signatures related to the Enterobacteriaceae family that are associated with mortality risk during a 15-year follow-up. Our results extend previous cross-sectional studies, and help to establish the basis for examining long-term associations between human gut microbiome composition, incident outcomes, and general health status.

Conflict of interest statement

The authors declare the following competing interests: V.S. has consulted for Novo Nordisk and Sanofi and received honoraria from these companies. He also has ongoing research collaboration with Bayer AG, all unrelated to this study. The other authors declare no competing interests.

Figures

Fig. 1. Study sample and gut microbiome…
Fig. 1. Study sample and gut microbiome characteristics.
a At baseline, the study sample (n = 7211) had a balanced sex ratio (55% women in red:men in blue), a mean age of 49 years (range 24–74; left panel) and a mean body mass index (BMI) of 27 kg/m2 (range 16–57; middle panel). During a median follow-up time of 14.8 years, 721 of 7055 (10.2%) participants with complete data who were included in the prospective analysis died (right panel). b A total of 7211 out of 13,500 randomly sampled individuals (53.4% participation rate) from six catchment areas in Finland underwent stool sampling, a physical examination and filled in a questionnaire on health behaviour, history of diseases and current health. c Principal coordinate analysis (PCoA) indicates sample similarity based on species-level taxonomic composition. The colour indicates the dominant (most abundant) genus in each sample. Altogether, 96% of the samples are dominated by one of the six genera that are indicated in the figure.
Fig. 2. Principal components and mortality risk.
Fig. 2. Principal components and mortality risk.
Association between mortality risk and the first three principal components of beta diversity (PC). Black line indicates the estimated hazard ratio compared to median PC value and blue area the 95% confidence interval (CI). Unit variance increase in the PCs were related to hazard ratios of 0.92 (95% CI, 0.85–0.99; FDR-adjusted P = 0.065; two-tailed Wald test), 0.95 (95% CI, 0.87–1.02; FDR-adjusted P = 0.17; two-tailed Wald test) and 1.14 (95% CI, 1.07–1.23; FDR-adjusted P = 0.001; two-tailed Wald test) for PC1–PC3, respectively. Analyses are adjusted for age, body mass index, sex, smoking, diabetes, use of antineoplastic and immunomodulating agents, systolic blood pressure and self-reported antihypertensive medication. The dashed line represents a hazard ratio of 1 set at median PC value. HR hazard ratio.
Fig. 3. The association between Enterobacteriaceae abundance…
Fig. 3. The association between Enterobacteriaceae abundance and cause-specific mortality.
Cox hazard ratios and 95% confidence intervals are reported per unit variance increase in Enterobacteriaceae abundance. Box sizes are inversely proportional to P values. The entire study sample (n = 7211) was examined independently with each cause of death as end point. Analyses are adjusted for age, body mass index, sex, smoking, diabetes, use of antineoplastic and immunomodulating agents, systolic blood pressure and self-reported antihypertensive medication. HR hazard ratio, FDR false discovery rate.
Fig. 4. Taxonomic subnetwork associated with increased…
Fig. 4. Taxonomic subnetwork associated with increased mortality risk.
a Abundance variation across the study population for the subnetwork that exhibits the strongest mortality associations (CLR-transformed abundances centred at zero and scaled to unit variance). The samples are ordered by the total relative abundance of the subnetwork. b The observed subnetwork structure and mortality risk. The total subnetwork abundance was associated with elevated mortality with a hazard ratio of 1.155 (95% confidence interval [CI], 1.08–1.24; P = 0.0002, Wald two-tailed test statistic for Cox regression, 4.07) The respective hazard ratios were 1.17 (95% CI, 1.07–1.27; P = 0.001, Wald statistic 3.66) in the Eastern and 1.14 (95% CI, 1.001–1.31; P = 0.15, Wald statistic 2.02) in the Western Finnish populations. The analyses are conducted after excluding rare taxa and adjusted for age, body mass index, sex, smoking, diabetes, use of antineoplastic and immunomodulating agents, systolic blood pressure and self-reported antihypertensive medication; P values are FDR-adjusted.

References

    1. Jackson, M. A. et al. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat. Commun. 9, 2655 (2018).
    1. Falony G, et al. Population-level analysis of gut microbiome variation. Science. 2016;352:560–564. doi: 10.1126/science.aad3503.
    1. McDonald, D. et al. American Gut: an Open Platform for Citizen Science Microbiome Research. mSystems3, e00031-18 (2018).
    1. Zhernakova A, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–569. doi: 10.1126/science.aad3369.
    1. Gilbert JA, et al. Current understanding of the human microbiome. Nat. Med. 2018;24:392–400. doi: 10.1038/nm.4517.
    1. Rajilić-Stojanović, M., Heilig, H. G. H. J., Tims, S., Zoetendal, E. G. & de Vos, W. M. Long-term monitoring of the human intestinal microbiota composition. Environ. Microbiol 15, 1146–1159 (2013).
    1. Stewart CJ, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562:583–588. doi: 10.1038/s41586-018-0617-x.
    1. Vatanen T, et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature. 2018;562:589–594. doi: 10.1038/s41586-018-0620-2.
    1. Borodulin K, et al. Forty-year trends in cardiovascular risk factors in Finland. Eur. J. Public Health. 2015;25:539–546. doi: 10.1093/eurpub/cku174.
    1. Haukka J. Finnish health and social welfare registers in epidemiological research. Nor. Epidemiol. 2009;14:113–120.
    1. Sund R. Quality of the Finnish Hospital Discharge Register: a systematic review. Scand. J. Public Health. 2012;40:505–515. doi: 10.1177/1403494812456637.
    1. Lim ET, et al. Distribution and medical impact of loss-of-function variants in the Finnish founder population. PLoS Genet. 2014;10:e1004494. doi: 10.1371/journal.pgen.1004494.
    1. GBD 2017 Risk Factor Collaborators. Global, regional, and national comparative risk assessment of 84 behavioural, environmental and occupational, and metabolic risks or clusters of risks for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet. 2018;392:1923–1994. doi: 10.1016/S0140-6736(18)32225-6.
    1. Kerminen S, et al. Fine-scale genetic structure in Finland. G3. 2017;7:3459–3468. doi: 10.1534/g3.117.300217.
    1. Pekkanen J, Manton KG, Stallard E, Nissinen A, Karvonen MJ. Risk factor dynamics, mortality and life expectancy differences between eastern and western Finland: the Finnish Cohorts of the Seven Countries Study. Int. J. Epidemiol. 1992;21:406–419. doi: 10.1093/ije/21.2.406.
    1. Riddle MS, DuPont HL, Connor BA. ACG Clinical Guideline: diagnosis, treatment, and prevention of acute diarrheal infections in adults. Am. J. Gastroenterol. 2016;111:602–622. doi: 10.1038/ajg.2016.126.
    1. Ghazalpour A, Cespedes I, Bennett BJ, Allayee H. Expanding role of gut microbiota in lipid metabolism. Curr. Opin. Lipidol. 2016;27:141–147. doi: 10.1097/MOL.0000000000000278.
    1. Utzschneider KM, Kratz M, Damman CJ, Hullar M. Mechanisms linking the gut microbiome and glucose metabolism. J. Clin. Endocrinol. Metab. 2016;101:1445–1454. doi: 10.1210/jc.2015-4251.
    1. Wilson ID, Nicholson JK. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl. Res. 2017;179:204–222. doi: 10.1016/j.trsl.2016.08.002.
    1. Vich Vila A, et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat. Commun. 2020;11:362. doi: 10.1038/s41467-019-14177-z.
    1. Maier L, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555:623–628. doi: 10.1038/nature25979.
    1. Fung TC, Olson CA, Hsiao EY. Interactions between the microbiota, immune and nervous systems in health and disease. Nat. Neurosci. 2017;20:145–155. doi: 10.1038/nn.4476.
    1. FinnGen Project. et al. Exome sequencing of Finnish isolates enhances rare-variant association power. Nature. 2019;572:323–328. doi: 10.1038/s41586-019-1457-z.
    1. Pencina MJ, D’Agostino RB, Vasan RS. Statistical methods for assessment of added usefulness of new biomarkers. Clin. Chem. Lab. Med. 2010;48:1703–1711. doi: 10.1515/CCLM.2010.340.
    1. Cook NR. Use and misuse of the receiver operating characteristic curve in risk prediction. Circulation. 2007;115:928–935. doi: 10.1161/CIRCULATIONAHA.106.672402.
    1. Taylor J, Tibshirani RJ. Statistical learning and selective inference. Proc. Natl Acad. Sci. USA. 2015;112:7629–7634. doi: 10.1073/pnas.1507583112.
    1. Bair E, Hastie T, Paul D, Tibshirani R. Prediction by supervised principal components. J. Am. Stat. Assoc. 2006;101:119–137. doi: 10.1198/016214505000000628.
    1. Duvallet C, Gibbons SM, Gurry T, Irizarry RA, Alm EJ. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat. Commun. 2017;8:1784. doi: 10.1038/s41467-017-01973-8.
    1. Donaldson GP, Lee SM, Mazmanian SK. Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 2016;14:20–32. doi: 10.1038/nrmicro3552.
    1. Lupp C, et al. Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe. 2007;2:204. doi: 10.1016/j.chom.2007.08.002.
    1. Moreno E, et al. Relationship between Escherichia coli strains causing acute cystitis in women and the fecal E. coli population of the host. J. Clin. Microbiol. 2008;46:2529–2534. doi: 10.1128/JCM.00813-08.
    1. Kaper JB, Nataro JP, Mobley HLT. Pathogenic Escherichia coli. Nat. Rev. Microbiol. 2004;2:123–140. doi: 10.1038/nrmicro818.
    1. Wiles TJ, Kulesus RR, Mulvey MA. Origins and virulence mechanisms of uropathogenic Escherichia coli. Exp. Mol. Pathol. 2008;85:11–19. doi: 10.1016/j.yexmp.2008.03.007.
    1. Dautzenberg MJD, et al. The association between colonization with carbapenemase-producing enterobacteriaceae and overall ICU mortality: an observational cohort study. Crit. Care Med. 2015;43:1170–1177. doi: 10.1097/CCM.0000000000001028.
    1. Hyle EP, et al. Impact of inadequate initial antimicrobial therapy on mortality in infections due to extended-spectrum beta-lactamase-producing enterobacteriaceae: variability by site of infection. Arch. Intern. Med. 2005;165:1375–1380. doi: 10.1001/archinte.165.12.1375.
    1. Gutiérrez-Gutiérrez B, et al. Effect of appropriate combination therapy on mortality of patients with bloodstream infections due to carbapenemase-producing Enterobacteriaceae (INCREMENT): a retrospective cohort study. Lancet Infect. Dis. 2017;17:726–734. doi: 10.1016/S1473-3099(17)30228-1.
    1. He Y, et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 2018;24:1532–1535. doi: 10.1038/s41591-018-0164-x.
    1. Tigchelaar EF, et al. Cohort profile: LifeLines DEEP, a prospective, general population cohort study in the northern Netherlands: study design and baseline characteristics. BMJ Open. 2015;5:e006772. doi: 10.1136/bmjopen-2014-006772.
    1. Hillmann, B. et al. Evaluating the information content of shallow shotgun metagenomics. mSystems3, e00069-18 (2018).
    1. Ye SH, Siddle KJ, Park DJ, Sabeti PC. Benchmarking metagenomics tools for taxonomic classification. Cell. 2019;178:779–794. doi: 10.1016/j.cell.2019.07.010.
    1. Sanders JG, et al. Optimizing sequencing protocols for leaderboard metagenomics by combining long and short reads. Genome Biol. 2019;20:1–14. doi: 10.1186/s13059-019-1834-9.
    1. Glenn, T. C. et al. Adapterama I: universal stubs and primers for thousands of dual-indexed Illumina libraries (iTru & iNext). 10.1101/049114 (2016).
    1. Liu B, Zheng D, Jin Q, Chen L, Yang J. VFDB 2019: a comparative pathogenomic platform with an interactive web interface. Nucleic Acids Res. 2019;47:D687–D692. doi: 10.1093/nar/gky1080.
    1. Eren AM, et al. Anvi’o: an advanced analysis and visualization platform for ‘omics data. PeerJ. 2015;3:e1319. doi: 10.7717/peerj.1319.
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. World Health Organization. International Statistical Classification of Diseases and Related Health Problems (World Health Organization, 2004).
    1. Anatomical Therapeutic Chemical Classification System (WHO). The SAGE Encyclopedia of Pharmacology and Society (SAGE Publications, 2016).
    1. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2018).
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B. 1995;57:289–300.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE. 2013;8:e61217. doi: 10.1371/journal.pone.0061217.
    1. Kurtz ZD, et al. Sparse and compositionally robust inference of microbial ecological networks. PLoS Comput. Biol. 2015;11:e1004226. doi: 10.1371/journal.pcbi.1004226.
    1. Liu H, Roeder K, Wasserman L. Stability approach to regularization selection (StARS) for high dimensional graphical models. Adv. Neural Inf. Process. Syst. 2010;24:1432–1440.
    1. Cox DR. Regression models and life‐tables. J. R. Stat. Soc. Ser. B. 1972;32:187–220.
    1. Therneau, T. M. Package ‘survival’. (2015).
    1. Ishwaran H, Kogalur UB, Blackstone EH, Lauer MS. Random survival forests. Ann. Appl. Stat. 2008;2:841–860. doi: 10.1214/08-AOAS169.
    1. Ishwaran, H., Kogalur, U. B. & Kogalur, M. U. B. Package ‘randomForestSRC’. (2018).
    1. Harrell FE, Jr., Califf RM, Pryor DB, Lee KL, Rosati RA. Evaluating the yield of medical tests. JAMA. 1982;247:2543–2546. doi: 10.1001/jama.1982.03320430047030.
    1. Uchiyama T, Irie M, Mori H, Kurokawa K, Yamada T. FuncTree: Functional Analysis and Visualization for Large-Scale Omics Data. PLoS ONE. 2015;10:e0126967. doi: 10.1371/journal.pone.0126967.

Source: PubMed

3
Suscribir