Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Environment dominates over host genetics in shaping human gut microbiota

Abstract

Human gut microbiome composition is shaped by multiple factors but the relative contribution of host genetics remains elusive. Here we examine genotype and microbiome data from 1,046 healthy individuals with several distinct ancestral origins who share a relatively common environment, and demonstrate that the gut microbiome is not significantly associated with genetic ancestry, and that host genetics have a minor role in determining microbiome composition. We show that, by contrast, there are significant similarities in the compositions of the microbiomes of genetically unrelated individuals who share a household, and that over 20% of the inter-person microbiome variability is associated with factors related to diet, drugs and anthropometric measurements. We further demonstrate that microbiome data significantly improve the prediction accuracy for many human traits, such as glucose and obesity measures, compared to models that use only host genetic and environmental data. These results suggest that microbiome alterations aimed at improving clinical outcomes may be carried out across diverse genetic backgrounds.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Genetic ancestry is not significantly associated with microbiome composition.
Figure 2: Genetic kinship is weakly associated with microbiome composition.
Figure 3: Limited evidence for microbiome associations with specific SNPs.
Figure 4: The gut microbiome can be used to infer a significant fraction of the variance of several human phenotypes.

Similar content being viewed by others

References

  1. Le Chatelier, E . et al. Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546 (2013)

    Article  CAS  PubMed  Google Scholar 

  2. Clemente, J. C ., Ursell, L. K ., Parfrey, L. W. & Knight, R. The impact of the gut microbiota on human health: an integrative view. Cell 148, 1258–1270 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kurilshikov, A ., Wijmenga, C ., Fu, J . & Zhernakova, A. Host genetics and gut microbiome: challenges and perspectives. Trends Immunol. 38, 633–647 (2017)

    Article  CAS  PubMed  Google Scholar 

  4. Goodrich, J. K. K . et al. Human genetics shape the gut microbiome. Cell 159, 789–799 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Turpin, W . et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat. Genet. 48, 1413–1417 (2016)

    Article  CAS  PubMed  Google Scholar 

  6. Goodrich, J. K . et al. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe 19, 731–743 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Goodrich, J. K ., Davenport, E. R ., Clark, A. G. & Ley, R. E. The relationship between the human genome and microbiome comes into view. Annu. Rev. Genet. 51, 413–433 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bonder, M. J . et al. The effect of host genetics on the gut microbiome. Nat. Genet. 48, 1407–1412 (2016)

    Article  CAS  PubMed  Google Scholar 

  9. Wang, J . et al. Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat. Genet. 48, 1396–1406 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Blekhman, R . et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 16, 191 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Xie, H . et al. Shotgun metagenomics of 250 adult twins reveals genetic and environmental impacts on the gut microbiome. Cell Syst. 3, 572–584 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zeevi, D . et al. Personalized nutrition by prediction of glycemic responses. Cell 163, 1079–1094 (2015)

    Article  CAS  PubMed  Google Scholar 

  13. Zhernakova, A . et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352, 565–569 (2016)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  14. Falony, G . et al. Population-level analysis of gut microbiome variation. Science 352, 560–564 (2016)

    Article  ADS  CAS  PubMed  Google Scholar 

  15. Fu, J . et al. The gut microbiome contributes to a substantial proportion of the variation in blood lipids. Circ. Res. 117, 817–824 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Behar, D. M. et al. The genome-wide structure of the Jewish people. Nature 466, 238–242 (2010)

    Article  ADS  CAS  PubMed  Google Scholar 

  17. Legendre P. & Legendre. L. Numerical Ecology Vol. 24, 3rd edn (Elsevier, 2012)

  18. Visscher, P. M . & Goddard, M. E. A general unified framework to assess the sampling variance of heritability estimates using pedigree or marker-based relationships. Genetics 199, 223–232 (2015)

    Article  PubMed  Google Scholar 

  19. Rühlemann, M. C . et al. Application of the distance-based F test in an mGWAS investigating β diversity of intestinal microbiota identifies variants in SLC9A8 (NHE8) and 3 other loci. Gut Microbes https://doi.org/10.1080/19490976.2017.1356979 (2017)

  20. Song, S. J . et al. Cohabiting family members share microbiota with one another and with their dogs. eLife 2, e00458 (2013)

    Article  PubMed  PubMed Central  Google Scholar 

  21. McArdle, B. H . & Anderson, M. J. Fitting multivariate models to community data: a comment on distance-based redundancy analysis. Ecology 82, 290–297 (2001)

    Article  Google Scholar 

  22. Visscher, P. M ., Hill, W. G. & Wray, N. R. Heritability in the genomics era--concepts and misconceptions. Nat. Rev. Genet. 9, 255–266 (2008)

    Article  CAS  PubMed  Google Scholar 

  23. Yang, J . et al. Common SNPs explain a large proportion of the heritability for human height. Nat. Genet. 42, 565–569 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, J ., Lee, S. H ., Goddard, M. E. & Visscher, P. M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Schweiger, R . et al. in RECOMB 2017: Research in Computational Molecular Biology (ed. Sahinalp, S. ) 241–256 (Springer, 2017)

  26. Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case Control Consortium 2. A genome-wide association study identifies new psoriasis susceptibility loci and an interaction between HLA-C and ERAP1. Nat. Genet. 42, 985–990 (2010)

  27. Shi, H ., Kichaev, G. & Pasaniuc, B. Contrasting the genetic architecture of 30 complex traits from summary association data. Am. J. Hum. Genet. 99, 139–153 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Speed, D ., Cai, N ., Johnson, M. R ., Nejentsev, S . & Balding, D. J. Reevaluation of SNP heritability in complex human traits. Nat. Genet. 49, 986–992 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ge, T ., Chen, C. Y ., Neale, B. M ., Sabuncu, M. R . & Smoller, J. W. Phenome-wide heritability analysis of the UK Biobank. PLoS Genet. 13, e1006711 (2017)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zaitlen, N . et al. Using extended genealogy to estimate components of heritability for 23 quantitative and dichotomous traits. PLoS Genet. 9, e1003520 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Vattikuti, S ., Guo, J. & Chow, C. C. Heritability and genetic correlations explained by common SNPs for metabolic syndrome traits. PLoS Genet. 8, e1002637 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Arpegård, J . et al. Comparison of heritability of cystatin C- and creatinine-based estimates of kidney function and their relation to heritability of cardiovascular disease. J. Am. Heart Assoc. 4, e001467 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xia, C . et al. Pedigree- and SNP-associated genetics and recent environment are the major contributors to anthropometric and cardiometabolic trait variation. PLoS Genet. 12, e1005804 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Heckerman, D . et al. Linear mixed model for heritability estimation that explicitly addresses environmental variation. Proc. Natl Acad. Sci. USA 113, 7377–7382 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Antonopoulos, D. A . et al. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect. Immun. 77, 2367–2375 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Caporaso, J. G . et al. Moving pictures of the human microbiome. Genome Biol. 12, R50 (2011)

    Article  PubMed  PubMed Central  Google Scholar 

  37. Lozupone, C. A ., Stombaugh, J. I ., Gordon, J. I ., Jansson, J. K. & Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 489, 220–230 (2012)

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Widmer, C . et al. Further improvements to linear mixed models for genome-wide association studies. Sci. Rep. 4, 6874 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  39. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Segata, N. et al. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat. Methods 9, 811–814 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Loh, P.-R. et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat. Genet. 48, 1443–1448 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Howie, B. N., Donnelly, P. & Marchini, J. A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet. 5, e1000529 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Auton, A. et al. A global reference for human genetic variation. Nature 526, 68–74 (2015)

    Article  ADS  CAS  PubMed  Google Scholar 

  45. Carmi, S. et al. Sequencing an Ashkenazi reference panel supports population-targeted personal genomics and illuminates Jewish and European origins. Nat. Commun. 5, 4835 (2014)

    Article  ADS  CAS  PubMed  Google Scholar 

  46. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Marco-Sola, S., Sammeth, M., Guigó, R. & Ribeca, P. The GEM mapper: fast, accurate and versatile alignment by filtration. Nat. Methods 9, 1185–1188 (2012)

    Article  CAS  PubMed  Google Scholar 

  48. Li, J. et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 32, 834–841 (2014)

    Article  CAS  PubMed  Google Scholar 

  49. Hong, C. et al. PathoScope 2.0: a complete computational framework for strain identification in environmental or clinical sequencing samples. Microbiome 2, 33 (2014)

    Article  PubMed  PubMed Central  Google Scholar 

  50. Conomos, M. P., Reiner, A. P., Weir, B. S. & Thornton, T. A. Model-free estimation of recent genetic relatedness. Am. J. Hum. Genet. 98, 127–148 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Conomos, M. P., Miller, M. B. & Thornton, T. A. Robust inference of population structure for ancestry prediction and correction of stratification in the presence of relatedness. Genet. Epidemiol. 39, 276–293 (2015)

    Article  PubMed  PubMed Central  Google Scholar 

  52. Manichaikul, A. et al. Robust relationship inference in genome-wide association studies. Bioinformatics 26, 2867–2873 (2010)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Conomos, M. P. et al. Genetic diversity and association studies in US Hispanic/Latino populations: applications in the Hispanic Community Health Study/Study of Latinos. Am. J. Hum. Genet. 98, 165–184 (2016)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Oksanen, J. et al. vegan: community ecology package. https://cran.r-project.org/web/packages/vegan/index.html (2017)

  55. Hastie, T ., Tibshirani, R. & Friedman, J. The Elements of Statistical Learning: Data Mining, Inference, and Prediction (Springer, 2009)

  56. Chen, T. & Guestrin, C. Xgboost: A scalable tree boosting system. In Proc. 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining 785–794 (ACM, 2016)

  57. Listgarten, J. et al. Improved linear mixed models for genome-wide association studies. Nat. Methods 9, 525–526 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Yang, J., Zaitlen, N. A., Goddard, M. E., Visscher, P. M. & Price, A. L. Advantages and pitfalls in the application of mixed-model association methods. Nat. Genet. 46, 100–106 (2014)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ingram, C. J. E., Mulcare, C. A., Itan, Y., Thomas, M. G. & Swallow, D. M. Lactose digestion and the evolutionary genetics of lactase persistence. Hum. Genet. 124, 579–591 (2009)

    Article  CAS  PubMed  Google Scholar 

  60. Schweiger, R. et al. RL-SKAT: an exact and efficient score test for heritability and set tests. Genetics 207, 1275–1283 (2017)

    PubMed  PubMed Central  Google Scholar 

  61. de los Campos, G., Vazquez, A. I., Fernando, R., Klimentidis, Y. C. & Sorensen, D. Prediction of complex human traits using the genomic best linear unbiased predictor. PLoS Genet. 9, e1003608 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhao, N. et al. Testing in microbiome-profiling studies with MiRKAT, the microbiome regression-based kernel association test. Am. J. Hum. Genet. 96, 797–807 (2015)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Segal and Elinav group members for discussions; J. Goodrich for sharing the processed twins microbiome data with us; and participants and staff of the LifeLines DEEP cohort for their collaboration. S.C. thanks the Abisch–Frenkel Foundation. This study makes use of data generated by the Wellcome Trust Case Control Consortium. A full list of the investigators who contributed to the generation of the data is available from www.wtccc.org.uk. Funding for the project was provided by the Wellcome Trust under awards 076113 and 085475. E.S. is supported by the Crown Human Genome Center; the Else Kroener Fresenius Foundation; D. L. Schwarz; J. N. Halpern; L. Steinberg; and grants funded by the European Research Council and the Israel Science Foundation. E.E. is supported by Y. and R. Ungar, the Gurwin Family Fund for Scientific Research, the Leona M. and Harry B. Helmsley Charitable Trust, the Israel Science Foundation and the Helmholtz Foundation. E.E. holds the Sir Marc and Lady Tania Feldmann Professorial Chair in Immunology, is a senior fellow of the Canadian Institute for Advanced Research, and is an international scholar at the Bill and Melinda Gates Foundation and Howard Hughes Medical Institute. D.R. received a Levi Eshkol PhD Scholarship for Personalized Medicine by the Israeli Ministry of Science. LLD was made possible by grants from the Top Institute Food and Nutrition (GH001) to C.W. C.W. is funded by a European Research Council (ERC) advanced grant (FP/2007-2013/ERC grant 2012-322698), a Netherlands Organization for Scientific Research (NWO) Spinoza prize (NWO SPI 92-266) and the Stiftelsen Kristian Gerhard Jebsen foundation (Norway). A.Z. holds a Rosalind Franklin Fellowship (University of Groningen), ERC starting grant (715772) and NWO Vidi grant (178.056). J.F. is funded by an NWO Vidi grant (NWO-VIDI 864.13.013). A.Z. and J.F. are also funded by CardioVasculair Onderzoek Nederland (CVON 2012-03).

Author information

Authors and Affiliations

Authors

Contributions

D.R., O.W. and E.B. conceived the project, designed and conducted all analyses, interpreted the results, wrote the manuscript and are listed in random order. A.K., A.V.V., J.F., C.W. and A.Z. performed the analyses of the Dutch cohort and interpreted the results. T.K., D.Z. and A.W. designed protocols and supervised data collection. T.K., D.Z., P.I.C., A.G., I.N.K. and N.B. conducted microbiome analyses. S.S. and D.L. designed nutritional and drug databases. N.Z., M.P.-F, D.I. and Z.H. coordinated and supervised clinical aspects of data collection. N.K., G.M. and B.C.W. coordinated and designed data collection. T.A.-S., M.L.-P. and A.W. developed protocols and performed genotyping and microbiome sequencing. S.C. designed the genetic analyses. E.E. and E.S. conceived and directed the project and analyses, designed the analyses, interpreted the results and wrote the manuscript.

Corresponding authors

Correspondence to Eran Elinav or Eran Segal.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Reviewer Information Nature thanks M. Georges and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Figure 1 Limited evidence for microbiome associations with genetic ancestry or kinship across multiple functional and taxonomic levels.

ap, Each row is similar to Figs 1b, d–e, 2b, but is based on the abundance of bacterial genes (ad), genera (eh), genera based on 16S rRNA gene sequencing data (il) or phyla (mp). a, d, e, h, m, p, n = 715 genotyped individuals; i, l, n = 481 individuals with 16S rRNA gene sequencing data; b, f, n, n = 737 individuals for whom the ancestries of all grandparents are known; j, n = 509 individuals with 16S rRNA gene sequencing data for whom the ancestries of all grandparents are known; c, g, o, n = 946 individuals; and k, n = 650 individuals with 16S rRNA gene sequencing data.

Source data

Extended Data Figure 2 Limited evidence for associations between microbiome β-diversity and specific SNPs.

The quantile–quantile plot shows that only two SNPs are significantly associated with microbiome β-diversity at P < 5 × 10−8, computed using a distance-based F test with n = 715 unrelated genotyped individuals. λGC, genomic inflation factor.

Extended Data Figure 3 Individuals who share a household at present or have shared one in the past have significantly similar microbiomes.

First-degree relatives and individuals with present household sharing have significantly similar species and bacterial gene abundances (P < 0.01; permutation testing). a–c, Box plots depict the distribution of Bray–Curtis dissimilarities across pairs of individuals at the phylum (a), species (b) and bacterial genes (c) level. Each panel shows the Bray–Curtis dissimilaries among all pairs of (i) first-degree relatives, who are likely to have experienced present or past household sharing (n = 55 pairs); (ii) second-to-fifth-degree relatives, who are unlikely to have experienced present or past household sharing (n = 24 pairs); (iii) unrelated individuals self-reported to currently share a household (n = 32 pairs); and (iv) all other individuals (n = 255,891 pairs). The lower and upper limits of the boxes represent the 25% and 75% percentiles, respectively, and the top and bottom whiskers represent the 5% and 95% percentiles, respectively. The P value ranges for all panels are: **P < 0.01 and ***P < 0.005.

Source data

Extended Data Figure 4 The gut microbiome is significantly associated with multiple environmental factors.

The fraction of variance of the microbiome β-diversity matrix that can be inferred from different categories of environmental factors is shown. n = 715 individuals (Supplementary Table 17); numbers in parentheses indicate the number of features in each category. The fraction of inferred variance can reflect both the information that the category conveys on the microbiome as well as the number of factors in the category, which depends on the questionnaire used in the study.

Source data

Extended Data Figure 5 b2 estimates and phenotype prediction results when using various data sources.

Each row is similar to Fig. 4c–e, but is based on a different data source. ac, Relative abundance of genera, obtained from 16S rRNA gene sequencing (using n = 464 individuals). df, Relative abundance of genera, obtained from metagenomic sequencing (using n = 715 individuals). gi, Relative abundance of phyla (using n = 715 individuals). jl, Relative abundance of species (using n = 715 individuals). mo, Relative abundance of bacterial genes in the LLD cohort (using n = 836 individuals). Note that two phenotypes that were analysed in the Israeli cohort (lactose consumption and glycaemic status) were not available for the LLD cohort, and two phenotypes available for the LLD cohort and shown here (LDL cholesterol and triglycerides) were not available for the Israeli cohort. The P value ranges for all panels are: *FDR < 0.05, **FDR < 0.01 and ***FDR < 0.001.

Source data

Extended Data Table 1 Baseline characteristics of the cohort
Extended Data Table 2 No significant association between ancestral or genetic similarity and the gut microbiome

Supplementary information

Life Sciences Reporting Summary (PDF 74 kb)

Supplementary Information

This file contains Power Simulations: A detailed description of our power simulations procedure and Statistical Aspects of the Microbiome-Association Index: A clarification regarding the assumptions behind the derivation of the microbiome-association index, and their statistical implications. (PDF 1083 kb)

Supplementary Tables

This file contains Supplementary Tables 1-28. (XLSX 510 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rothschild, D., Weissbrod, O., Barkan, E. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215 (2018). https://doi.org/10.1038/nature25973

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature25973

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing