A comprehensive assessment of demographic, environmental, and host genetic associations with gut microbiome diversity in healthy individuals

Petar Scepanovic, Flavia Hodel, Stanislas Mondot, Valentin Partula, Allyson Byrd, Christian Hammer, Cécile Alanio, Jacob Bergstedt, Etienne Patin, Mathilde Touvier, Olivier Lantz, Matthew L Albert, Darragh Duffy, Lluis Quintana-Murci, Jacques Fellay, Milieu Intérieur Consortium, Laurent Abel, Andres Alcover, Hugues Aschard, Kalla Astrom, Philippe Bousso, Pierre Bruhns, Ana Cumano, Caroline Demangel, Ludovic Deriano, James Di Santo, Françoise Dromer, Darragh Duffy, Gérard Eberl, Jost Enninga, Jacques Fellay, Odile Gelpi, Ivo Gomperts-Boneca, Milena Hasan, Serge Hercberg, Olivier Lantz, Claude Leclerc, Hugo Mouquet, Sandra Pellegrini, Stanislas Pol, Antonio Rausell, Lars Rogge, Anavaj Sakuntabhai, Olivier Schwartz, Benno Schwikowski, Spencer Shorte, Vassili Soumelis, Frédéric Tangy, Eric Tartour, Antoine Toubert, Mathilde Touvier, Marie-Noëlle Ungeheuer, Matthew L Albert, Lluis Quintana-Murci, Petar Scepanovic, Flavia Hodel, Stanislas Mondot, Valentin Partula, Allyson Byrd, Christian Hammer, Cécile Alanio, Jacob Bergstedt, Etienne Patin, Mathilde Touvier, Olivier Lantz, Matthew L Albert, Darragh Duffy, Lluis Quintana-Murci, Jacques Fellay, Milieu Intérieur Consortium, Laurent Abel, Andres Alcover, Hugues Aschard, Kalla Astrom, Philippe Bousso, Pierre Bruhns, Ana Cumano, Caroline Demangel, Ludovic Deriano, James Di Santo, Françoise Dromer, Darragh Duffy, Gérard Eberl, Jost Enninga, Jacques Fellay, Odile Gelpi, Ivo Gomperts-Boneca, Milena Hasan, Serge Hercberg, Olivier Lantz, Claude Leclerc, Hugo Mouquet, Sandra Pellegrini, Stanislas Pol, Antonio Rausell, Lars Rogge, Anavaj Sakuntabhai, Olivier Schwartz, Benno Schwikowski, Spencer Shorte, Vassili Soumelis, Frédéric Tangy, Eric Tartour, Antoine Toubert, Mathilde Touvier, Marie-Noëlle Ungeheuer, Matthew L Albert, Lluis Quintana-Murci

Abstract

Background: The gut microbiome is an important determinant of human health. Its composition has been shown to be influenced by multiple environmental factors and likely by host genetic variation. In the framework of the Milieu Intérieur Consortium, a total of 1000 healthy individuals of western European ancestry, with a 1:1 sex ratio and evenly stratified across five decades of life (age 20-69), were recruited. We generated 16S ribosomal RNA profiles from stool samples for 858 participants. We investigated genetic and non-genetic factors that contribute to individual differences in fecal microbiome composition.

Results: Among 110 demographic, clinical, and environmental factors, 11 were identified as significantly correlated with α-diversity, ß-diversity, or abundance of specific microbial communities in multivariable models. Age and blood alanine aminotransferase levels showed the strongest associations with microbiome diversity. In total, all non-genetic factors explained 16.4% of the variance. We then searched for associations between > 5 million single nucleotide polymorphisms and the same indicators of fecal microbiome diversity, including the significant non-genetic factors as covariates. No genome-wide significant associations were identified after correction for multiple testing. A small fraction of previously reported associations between human genetic variants and specific taxa could be replicated in our cohort, while no replication was observed for any of the diversity metrics.

Conclusion: In a well-characterized cohort of healthy individuals, we identified several non-genetic variables associated with fecal microbiome diversity. In contrast, host genetics only had a negligible influence. Demographic and environmental factors are thus the main contributors to fecal microbiome composition in healthy individuals.

Trial registration: ClinicalTrials.gov identifier NCT01699893.

Keywords: 16S rRNA gene sequencing; Demographics; Environment; GWAS; Genomics; Gut; Healthy; Human; Microbiome.

Conflict of interest statement

C.H., A.B., and M.L.A. are employees of Genentech Inc., a member of The Roche Group. The other authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Non-genetic variables. Six categories of non-genetic variables investigated in this study. In the parenthesis are the number of variables per each category and for each two representative examples. Full description of the variables is available in Additional file 2: Table S1
Fig. 2
Fig. 2
Gut microbiome diversity. a Box-plots of relative abundances of 8 phyla that were observed in more than 10% of the donors. Outliers are also represented. b Violin plot of Simpson’s diversity index values observed among MI study participants. c Multidimensional scaling plot of Bray-Curtis dissimilarity matrix with study participants colored according to relative abundance of Firmicutes
Fig. 3
Fig. 3
Association of non-genetic variables with Simpson’s index. Significant variables from the univariate test and their Spearman ρ values (right-hand side). Heatmap represents the ANOVA’s p values from the multivariable test, and the asterisks denote the statistical significance (***p < 0.001, **p < 0.01, *p < 0.05). The results for other α-diversity metrics are available in Additional file 2: Table S3
Fig. 4
Fig. 4
Association of non-genetic variables with Bray-Curtis index. Significant variables from the univariate test and their R2 values (right-hand side). Heatmap represents the PERMANOVA’s p values from the multivariable test, and the asterisks denote the statistical significance (***p < 0.001, **p < 0.01, *p < 0.05). The results for other β-diversity metrics are available in Additional file 2: Table S5
Fig. 5
Fig. 5
Results of genome-wide association study between host genetic variants and microbiome diversity metrics. a Manhattan plot for Simpson’s diversity metric (representative α-diversity metric). The dashed horizontal line denotes the genome-wide significance threshold (Pα-threshold < 1.25 × 10−8). b Manhattan plot for Bray-Curtis dissimilarity matrix (representative ß-diversity index). The dashed horizontal line denotes the genome-wide significance threshold (Pβ-threshold < 1.67 × 10−8)

References

    1. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol Rev. 2010;90:859–904. doi: 10.1152/physrev.00045.2009.
    1. Honda K, Littman DR. The microbiome in infectious disease and inflammation. Annu Rev Immunol. 2012;30:759–795. doi: 10.1146/annurev-immunol-020711-074937.
    1. Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu H, Yu C, Li S, Jian M, Zhou Y, Li Y, Zhang X, Li S, Qin N, Yang H, Wang J, Brunak S, Doré J, Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J, Consortium MHIT, Bork P, Ehrlich SD, Wang J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi: 10.1038/nature08821.
    1. Knight R, Callewaert C, Marotz C, Hyde ER, Debelius JW, McDonald D, Sogin ML. The microbiome and human biology. Annu Rev Genomics Hum Genet. 2017;18:65–86. doi: 10.1146/annurev-genom-083115-022438.
    1. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med. 2016;8:51. doi: 10.1186/s13073-016-0307-y.
    1. Human Microbiome Project Consortium Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. doi: 10.1038/nature11234.
    1. Gill SR, Pop M, Deboy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, Gordon JI, Relman DA, Fraser-Liggett CM, Nelson KE. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312:1355–1359. doi: 10.1126/science.1124234.
    1. Wexler AG, Goodman AL. An insider's perspective: bacteroides as a window into the microbiome. Nat Microbiol. 2017;2:17026. doi: 10.1038/nmicrobiol.2017.26.
    1. Lloyd-Price J, Mahurkar A, Rahnavard G, Crabtree J, Orvis J, Hall AB, Brady A, Creasy HH, McCracken C, Giglio MG, McDonald D, Franzosa EA, Knight R, White O, Huttenhower C. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature. 2017;550:61–66. doi: 10.1038/nature23889.
    1. Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, Abe F, Osawa R. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90. doi: 10.1186/s12866-016-0708-5.
    1. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, Li Y, Xia Y, Xie H, Zhong H, Khan MT, Zhang J, Li J, Xiao L, Al-Aama J, Zhang D, Lee YS, Kotowska D, Colding C, Tremaroli V, Yin Y, Bergman S, Xu X, Madsen L, Kristiansen K, Dahlgren J, Wang J. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703. doi: 10.1016/j.chom.2015.04.004.
    1. Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, Clemente JC, Knight R, Heath AC, Leibel RL, Rosenbaum M, Gordon JI. The long-term stability of the human gut microbiota. Science. 2013;341:1237439. doi: 10.1126/science.1237439.
    1. Schroeder BO, Bäckhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nat Med. 2016;22:1079–1089. doi: 10.1038/nm.4185.
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, Heath AC, Warner B, Reeder J, Kuczynski J, Caporaso JG, Lozupone CA, Lauber C, Clemente JC, Knights D, Knight R, Gordon JI. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–227. doi: 10.1038/nature11053.
    1. Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, Ben-Yacov O, Lador D, Avnit-Sagi T, Lotan-Pompan M, Suez J, Mahdi JA, Matot E, Malka G, Kosower N, Rein M, Zilberman-Schapira G, Dohnalová L, Pevsner-Fischer M, Bikovsky R, Halpern Z, Elinav E, Segal E. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163:1079–1094. doi: 10.1016/j.cell.2015.11.001.
    1. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, Kurilshikov A, Bonder MJ, Valles-Colomer M, Vandeputte D, Tito RY, Chaffron S, Rymenans L, Verspecht C, De SL, Lima-Mendez G, D'hoe K, Jonckheere K, Homola D, Garcia R, Tigchelaar EF, Eeckhaudt L, Fu J, Henckaerts L, Zhernakova A, Wijmenga C, Raes J. Population-level analysis of gut microbiome variation. Science. 2016;352:560–564. doi: 10.1126/science.aad3503.
    1. Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S, Wang J, Imhann F, Brandsma E, Jankipersadsing SA, Joossens M, Cenit MC, Deelen P, Swertz MA, Weersma RK, Feskens EJ, Netea MG, Gevers D, Jonkers D, Franke L, Aulchenko YS, Huttenhower C, Raes J, Hofker MH, Xavier RJ, Wijmenga C, Fu J. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–569. doi: 10.1126/science.aad3369.
    1. Jackson MA, Verdi S, Maxan ME, Shin CM, Zierer J, Bowyer RCE, Martin T, Williams FMK, Menni C, Bell JT, Spector TD, Steves CJ. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat Commun. 2018;9:2655. doi: 10.1038/s41467-018-05184-7.
    1. Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, Costea PI, Godneva A, Kalka IN, Bar N, Shilo S, Lador D, Vila AV, Zmora N, Pevsner-Fischer M, Israeli D, Kosower N, Malka G, Wolf BC, Avnit-Sagi T, Lotan-Pompan M, Weinberger A, Halpern Z, Carmi S, Fu J, Wijmenga C, Zhernakova A, Elinav E, Segal E. Environment dominates over host genetics in shaping human gut microbiota. Nature. 2018;555:210–215. doi: 10.1038/nature25973.
    1. Goodrich JK, Davenport ER, Beaumont M, Jackson MA, Knight R, Ober C, Spector TD, Bell JT, Clark AG, Ley RE. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe. 2016;19:731–743. doi: 10.1016/j.chom.2016.04.017.
    1. Wang J, Thingholm LB, Skiecevičienė J, Rausch P, Kummen M, Hov JR, Degenhardt F, Heinsen FA, Rühlemann MC, Szymczak S, Holm K, Esko T, Sun J, Pricop-Jeckstadt M, Al-Dury S, Bohov P, Bethune J, Sommer F, Ellinghaus D, Berge RK, Hübenthal M, Koch M, Schwarz K, Rimbach G, Hübbe P, Pan WH, Sheibani-Tezerji R, Häsler R, Rosenstiel P, D’ Amato M, Cloppenborg-Schmidt K, Künzel S, Laudes M, Marschall HU, Lieb W, Nöthlings U, Karlsen TH, Baines JF, Franke A. Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat Genet. 2016;48:1396–1406. doi: 10.1038/ng.3695.
    1. Turpin Williams, Espin-Garcia Osvaldo, Xu Wei, Silverberg Mark S, Kevans David, Smith Michelle I, Guttman David S, Griffiths Anne, Panaccione Remo, Otley Anthony, Xu Lizhen, Shestopaloff Konstantin, Moreno-Hagelsieb Gabriel, Paterson Andrew D, Croitoru Kenneth. Association of host genome with intestinal microbial composition in a large healthy cohort. Nature Genetics. 2016;48(11):1413–1417. doi: 10.1038/ng.3693.
    1. Khachatryan ZA, Ktsoyan ZA, Manukyan GP, Kelly D, Ghazaryan KA, Aminov RI. Predominant role of host genetics in controlling the composition of gut microbiota. PLoS One. 2008;3:e3064. doi: 10.1371/journal.pone.0003064.
    1. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, Spector TD, Clark AG, Ley RE. Human genetics shape the gut microbiome. Cell. 2014;159:789–799. doi: 10.1016/j.cell.2014.09.053.
    1. Blekhman R, Goodrich JK, Huang K, Sun Q, Bukowski R, Bell JT, Spector TD, Keinan A, Ley RE, Gevers D, Clark AG. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015;16:191. doi: 10.1186/s13059-015-0759-1.
    1. Davenport ER, Cusanovich DA, Michelini K, Barreiro LB, Ober C, Gilad Y. Genome-wide association studies of the human gut microbiota. PLoS One. 2015;10:e0140301. doi: 10.1371/journal.pone.0140301.
    1. Bonder MJ, Kurilshikov A, Tigchelaar EF, Mujagic Z, Imhann F, Vila AV, Deelen P, Vatanen T, Schirmer M, Smeekens SP, Zhernakova DV, Jankipersadsing SA, Jaeger M, Oosting M, Cenit MC, Masclee AA, Swertz MA, Li Y, Kumar V, Joosten L, Harmsen H, Weersma RK, Franke L, Hofker MH, Xavier RJ, Jonkers D, Netea MG, Wijmenga C, Fu J, Zhernakova A. The effect of host genetics on the gut microbiome. Nat Genet. 2016;48:1407–1412. doi: 10.1038/ng.3663.
    1. Kolde R, Franzosa EA, Rahnavard G, Hall AB, Vlamakis H, Stevens C, Daly MJ, Xavier RJ, Huttenhower C. Host genetic variation and its microbiome interactions within the Human Microbiome Project. Genome Med. 2018;10:6. doi: 10.1186/s13073-018-0515-8.
    1. Partula Valentin, Mondot Stanislas, Torres Marion J, Kesse-Guyot Emmanuelle, Deschasaux Mélanie, Assmann Karen, Latino-Martel Paule, Buscail Camille, Julia Chantal, Galan Pilar, Hercberg Serge, Rouilly Vincent, Thomas Stéphanie, Quintana-Murci Lluis, Albert Matthew L, Duffy Darragh, Lantz Olivier, Touvier Mathilde. Associations between usual diet and gut microbiota composition: results from the Milieu Intérieur cross-sectional study. The American Journal of Clinical Nutrition. 2019;109(5):1472–1483. doi: 10.1093/ajcn/nqz029.
    1. Thomas S, Rouilly V, Patin E, Alanio C, Dubois A, Delval C, Marquier LG, Fauchoux N, Sayegrih S, Vray M, Duffy D, Quintana-Murci L, Albert ML. Milieu Intérieur Consortium. The Milieu Intérieur study—an integrative approach for study of human immunological variance. Clin Immunol. 2015;157:277–293. doi: 10.1016/j.clim.2014.12.004.
    1. Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol. 2018;15:397–411. doi: 10.1038/s41575-018-0011-z.
    1. Schmidt TSB, Raes J, Bork P. The human gut microbiome: from association to modulation. Cell. 2018;172:1198–1215. doi: 10.1016/j.cell.2018.02.044.
    1. Finucane MM, Sharpton TJ, Laurent TJ, Pollard KS. A taxonomic signature of obesity in the microbiome? Getting to the guts of the matter. PLoS One. 2014;9:e84689. doi: 10.1371/journal.pone.0084689.
    1. Willems A. The family Comamonadaceae. In: Rosenberg E, DeLong EF, Lory S, Stackebrandt E, Thompson F, editors. The prokaryotes: Alphaproteobacteria and Betaproteobacteria. Berlin: Springer; 2014. pp. 777–851.
    1. Madden RH, Bryder MJ, Poole NJ. Isolation and characterization of an anaerobic, cellulolytic bacterium, Clostridium papyrosolvens sp-nov. Int J Syst Bacteriol. 1982;32:87–91. doi: 10.1099/00207713-32-1-87.
    1. Truong DT, Tett A, Pasolli E, Huttenhower C, Segata N. Microbial strain-level population structure and genetic diversity from metagenomes. Genome Res. 2017;27:626–638. doi: 10.1101/gr.216242.116.
    1. Forster SC, Kumar N, Anonye BO, Almeida A, Viciani E, Stares MD, Dunn M, Mkandawire TT, Zhu A, Shao Y, Pike LJ, Louie T, Browne HP, Mitchell AL, Neville BA, Finn RD, Lawley TD. A human gut bacterial genome and culture collection for improved metagenomic analyses. Nat Biotechnol. 2019;37:186–192. doi: 10.1038/s41587-018-0009-7.
    1. Vatanen T, Plichta DR, Somani J, Münch PC, Arthur TD, Hall AB, Rudolf S, Oakeley EJ, Ke X, Young RA, Haiser HJ, Kolde R, Yassour M, Luopajärvi K, Siljander H, Virtanen SM, Ilonen J, Uibo R, Tillmann V, Mokurov S, Dorshakova N, Porter JA, McHardy AC, Lähdesmäki H, Vlamakis H, Huttenhower C, Knip M, Xavier RJ. Genomic variation and strain-specific functional adaptation in the human gut microbiome during early life. Nat Microbiol. 2019;4:470–479. doi: 10.1038/s41564-018-0321-5.
    1. Wang J, Kurilshikov A, Radjabzadeh D, Turpin W, Croitoru K, Bonder MJ, Jackson MA, Medina-Gomez C, Frost F, Homuth G, Rühlemann M, Hughes D, Kim HN, MiBioGen Consortium Initiative. Spector TD, Bell JT, Steves CJ, Timpson N, Franke A, Wijmenga C, Meyer K, Kacprowski T, Franke L, Paterson AD, Raes J, Kraaij R, Zhernakova A. Meta-analysis of human genome-microbiome association studies: the MiBioGen consortium initiative. Microbiome. 2018;6:101. doi: 10.1186/s40168-018-0479-3.
    1. Ó Cuív P, Aguirre de Cárcer D, Jones M, Klaassens ES, Worthley DL, Whitehall VL, Kang S, CS MS, Leggett BA, Morrison M. The effects from DNA extraction methods on the evaluation of microbial diversity associated with human colonic tissue. Microb Ecol. 2011;61:353–362. doi: 10.1007/s00248-010-9771-x.
    1. Yu Z, Morrison M. Improved extraction of PCR-quality community DNA from digesta and fecal samples. Biotechniques. 2004;36:808–812. doi: 10.2144/04365ST04.
    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. Joshi NA, Fass JN. Sickle: a sliding-window, adaptive, quality-based trimming tool for FastQ files. 2011.
    1. Bankevich A, Nurk S, Antipov D, Gurevich AA, Dvorkin M, Kulikov AS, Lesin VM, Nikolenko SI, Pham S, Prjibelski AD, Pyshkin AV, Sirotkin AV, Vyahhi N, Tesler G, Alekseyev MA, Pevzner PA. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J Comput Biol. 2012;19:455–477. doi: 10.1089/cmb.2012.0021.
    1. Zhang J, Kobert K, Flouri T, Stamatakis A. PEAR: a fast and accurate Illumina Paired-End reAd mergeR. Bioinformatics. 2014;30:614–620. doi: 10.1093/bioinformatics/btt593.
    1. Rognes T, Flouri T, Nichols B, Quince C, Mahé F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584. doi: 10.7717/peerj.2584.
    1. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011;27:2194–2200. doi: 10.1093/bioinformatics/btr381.
    1. Lagkouvardos I, Fischer S, Kumar N, Clavel T. Rhea: a transparent and modular R pipeline for microbial profiling based on 16S rRNA gene amplicons. PeerJ. 2017;5:e2836. doi: 10.7717/peerj.2836.
    1. Cole JR, Wang Q, Cardenas E, Fish J, Chai B, Farris RJ, Kulam-Syed-Mohideen AS, McGarrell DM, Marsh T, Garrity GM, Tiedje JM. The Ribosomal Database Project: improved alignments and new tools for rRNA analysis. Nucleic Acids Res. 2009;37:D141–D145. doi: 10.1093/nar/gkn879.
    1. Nawrocki EP, Kolbe DL, Eddy SR. Infernal 1.0: inference of RNA alignments. Bioinformatics. 2009;25:1335–1337. doi: 10.1093/bioinformatics/btp157.
    1. Price MN, Dehal PS, Arkin AP. FastTree 2--approximately maximum-likelihood trees for large alignments. PLoS One. 2010;5:e9490. doi: 10.1371/journal.pone.0009490.
    1. Yee TW. The VGAM package for categorical data analysis. J Stat Softw. 2010;32:1–34. doi: 10.18637/jss.v032.i10.
    1. Oksanen J, Blanchet FG, Friendly M, Kindt R, Legendre P, McGlinn D, Minchin PR, O'Hara RB, Simpson GL, Peter Solymos P, Stevens MHH, Szoecs E, Wagner H. Vegan: community ecology package. R package version 2.5–2. 2018.
    1. van Buuren S, Groothuis-Oudshoorn K. MICE: Multivariate Imputation by Chained Equations in R. J Stat Softw. 2011;45:1–67. doi: 10.18637/jss.v045.i03.
    1. Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, Reyes JA, Shah SA, LeLeiko N, Snapper SB, Bousvaros A, Korzenik J, Sands BE, Xavier RJ, Huttenhower C. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79. doi: 10.1186/gb-2012-13-9-r79.
    1. Patin E, Hasan M, Bergstedt J, Rouilly V, Libri V, Urrutia A, Alanio C, Scepanovic P, Hammer C, Jönsson F, Beitz B, Quach H, Lim YW, Hunkapiller J, Zepeda M, Green C, Piasecka B, Leloup L, Rogge L, Huetz F, Peguillet I, Lantz O, Fontes M, Di Santo JP, Thomas S, Fellay J, Duffy D, Quintana-Murci L, Albert ML, for The Milieu Intérieur Consortium Natural variation in innate immune cell parameters is preferentially driven by genetic factors. Nat Immunol. 2018;19:302–314. doi: 10.1038/s41590-018-0049-7.
    1. Scepanovic P, Alanio C, Hammer C, Hodel F, Bergstedt J, Patin E, Thorball CW, Chaturvedi N, Charbit B, Abel L, Quintana-Murci L, Duffy D, Albert ML, Fellay J, Milieu Intérieur Consortium Human genetic variants and age are the strongest predictors of humoral immune responses to common pathogens and vaccines. Genome Med. 2018;10:59. doi: 10.1186/s13073-018-0568-8.
    1. Manichaikul A, Mychaleckyj JC, Rich SS, Daly K, Sale M, Chen WM. Robust relationship inference in genome-wide association studies. Bioinformatics. 2010;26:2867–2873. doi: 10.1093/bioinformatics/btq559.
    1. Patterson N, Price AL, Reich D. Population structure and eigenanalysis. PLoS Genet. 2006;2:e190. doi: 10.1371/journal.pgen.0020190.
    1. Loh PR, Danecek P, Palamara PF, Fuchsberger C, A Reshef Y, K Finucane H, Schoenherr S, Forer L, McCarthy S, Abecasis GR, Durbin R, Price A L. Reference-based phasing using the Haplotype Reference Consortium panel. Nat Genet. 2016;48:1443–1448. doi: 10.1038/ng.3679.
    1. McCarthy S, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet. 2016;48:1279–1283. doi: 10.1038/ng.3643.
    1. Jia X, Han B, Onengut-Gumuscu S, Chen WM, Concannon PJ, Rich SS, Raychaudhuri S, de Bakker PI. Imputing amino acid polymorphisms in human leukocyte antigens. PLoS One. 2013;8:e64683. doi: 10.1371/journal.pone.0064683.
    1. Vukcevic D, Traherne JA, Næss S, Ellinghaus E, Kamatani Y, Dilthey A, Lathrop M, Karlsen TH, Franke A, Moffatt M, Cookson W, Trowsdale J, McVean G, Sawcer S, Leslie S. Imputation of KIR types from SNP variation data. Am J Hum Genet. 2015;97:593–607. doi: 10.1016/j.ajhg.2015.09.005.
    1. O'Connell J, Gurdasani D, Delaneau O, Pirastu N, Ulivi S, Cocca M, Traglia M, Huang J, Huffman JE, Rudan I, McQuillan R, Fraser RM, Campbell H, Polasek O, Asiki G, Ekoru K, Hayward C, Wright AF, Vitart V, Navarro P, Zagury JF, Wilson JF, Toniolo D, Gasparini P, Soranzo N, Sandhu MS, Marchini J. A general approach for haplotype phasing across the full spectrum of relatedness. PLoS Genet. 2014;10:e1004234. doi: 10.1371/journal.pgen.1004234.
    1. Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Secondgeneration PLINK: rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7. doi: 10.1186/s13742-015-0047-8.
    1. Hua X, Song L, Yu G, Goedert JJ, Abnet CC, Landi MT, Shi J. MicrobiomeGWAS: a tool for identifying host genetic variants associated with microbiome composition. bioRxiv. 2015. 10.1101/031187.

Source: PubMed

3
Předplatit