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.

Effect of host genetics on the gut microbiome in 7,738 participants of the Dutch Microbiome Project

An Author Correction to this article was published on 25 July 2022

This article has been updated

Abstract

Host genetics are known to influence the gut microbiome, yet their role remains poorly understood. To robustly characterize these effects, we performed a genome-wide association study of 207 taxa and 205 pathways representing microbial composition and function in 7,738 participants of the Dutch Microbiome Project. Two robust, study-wide significant (P < 1.89 × 10−10) signals near the LCT and ABO genes were found to be associated with multiple microbial taxa and pathways and were replicated in two independent cohorts. The LCT locus associations seemed modulated by lactose intake, whereas those at ABO could be explained by participant secretor status determined by their FUT2 genotype. Twenty-two other loci showed suggestive evidence (P < 5 × 10−8) of association with microbial taxa and pathways. At a more lenient threshold, the number of loci we identified strongly correlated with trait heritability, suggesting that much larger sample sizes are needed to elucidate the remaining effects of host genetics on the gut microbiome.

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

Fig. 1: Genome-wide association scan results.
Fig. 2: Association at the LCT locus and interaction with lactose intake.
Fig. 3: Association with blood types and interaction with FUT2.
Fig. 4: Weighted Spearman correlation between estimated heritability and number of suggestive loci.
Fig. 5: Power analysis taking into account bacterial presence levels.

Data availability

Raw sequencing microbiome data are available at European Genome-Phenome archive (accession number EGAS00001005027). Genotyping data and participant metadata are not publicly available to protect participants’ privacy, and neither can be deposited in public repositories to respect the research agreements in the informed consent. The data can be accessed by all bona-fide researchers with a scientific proposal by contacting the LifeLines Biobank (instructions at https://www.lifelines.nl/researcher/how-to-apply). Researchers will need to fill in an application form that will be reviewed within 2 weeks. If the proposed research complies with LifeLines regulations, such as noncommercial use and warranty of participants’ privacy, then researchers will receive a financial offer and a data and material transfer agreement to sign. In general, data will be released within 2 weeks after signing the offer and data and material transfer agreement. The data will be released in a remote system (the LifeLines workspace) running on a high-performance computer cluster to ensure data quality and security. The full GWAS summary statistical data for all 207 taxa and 205 pathways are instead available for direct download at NHGRI-EBI GWAS Catalog (https://www.ebi.ac.uk/gwas/) under the study accession numbers GCST90027446-GCST90027857 (accession numbers for each specific taxa and pathways can be found in Supplementary Table 13) or at https://dutchmicrobiomeproject.molgeniscloud.org. The processed microbiome data (taxonomy and pathway abundance per individual) can also be downloaded after filling in a request form available at the same website and after signing a data access agreement. This study also used the following databases: UniRef90 v.0.1.1 protein database and the ChocoPhlAn pangenome databases available within the Humann2 pipeline (https://huttenhower.sph.harvard.edu/humann2/), the Genome Taxonomy Database (https://gtdb.ecogenomic.org/) and the IEU GWAS database (https://gwas.mrcieu.ac.uk/). All other data supporting the findings of this study are available within the paper and Supplementary Note.

Change history

References

  1. Valdes, A. M., Walter, J., Segal, E. & Spector, T. D. Role of the gut microbiota in nutrition and health. BMJ 361, k2179 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hall, A. B., Tolonen, A. C. & Xavier, R. J. Human genetic variation and the gut microbiome in disease. Nat. Rev. Genet. 18, 690–699 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Fan, Y. & Pederson, O. Gut microbiota in human metabolic health and disease.Nat. Rev. Microbiol. 19, 55–71 (2020).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Falony, G. et al. Population-level analysis of gut microbiome variation. Science 352, 560–564 (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. Rothschild, D. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 555, 210–215 (2018).

    Article  CAS  PubMed  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. 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 

  11. Hughes, D. A. et al. Genome-wide associations of human gut microbiome variation and implications for causal inference analyses. Nat. Microbiol. 5, 1079–1087 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Kurilshikov, A. et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat. Genet. 53, 156–165 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  15. Vieira-Silva, S. et al. Species–function relationships shape ecological properties of the human gut microbiome. Nat. Microbiol. 1, 1–8 (2016).

    Article  Google Scholar 

  16. Gacesa, R. et al. The Dutch Microbiome Project defines factors that shape the healthy gut microbiome. Preprint at bioRxiv https://doi.org/10.1101/2020.11.27.401125 (2020).

  17. Sanna, S. et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 51, 600–605 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Qin, Y. et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Preprint at medRxiv https://doi.org/10.1101/2020.09.12.20193045 (2020).

  19. Zhou, W. et al. Efficiently controlling for case-control imbalance and sample relatedness in large-scale genetic association studies. Nat. Genet. 50, 1335–1341 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Liu, X. et al. A genome-wide association study for gut metagenome in Chinese adults illuminates complex diseases. Cell Discov. 7, 9 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Van Der Heide, H. M., Magnee, W. & Van Loghem, J. J. Blood group frequencies in the Netherlands. Am. J. Hum. Genet. 3, 344–347 (1951).

    Google Scholar 

  22. Rühlemann, M. C. et al. Genome-wide association study in 8,956 German individuals identifies influence of ABO histo-blood groups on gut microbiome. Nat. Genet. 53, 147–155 (2021).

    Article  PubMed  Google Scholar 

  23. Turroni, F. et al. Bifidobacterium bifidum as an example of a specialized human gut commensal. Front. Microbiol. 5, 437 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Turroni, F., Milani, C., van Sinderen, D. & Ventura, M. Genetic strategies for mucin metabolism in Bifidobacterium bifidum PRL2010: an example of possible human-microbe co-evolution. Gut Microbes 2, 183–189 (2011).

    Article  PubMed  Google Scholar 

  25. Bonder, M. J. et al. Genetic and epigenetic regulation of gene expression in fetal and adult human livers. BMC Genomics 15, 860 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Moon, Jee-Young et al. Milk intake, host LCT genotype and gut Bifidobacteria in relation to obesity: results from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). Circulation 141, AP459 (2020).

    Article  Google Scholar 

  27. Arnolds, K. L., Martin, C. G. & Lozupone, C. A. Blood type and the microbiome: untangling a complex relationship with lessons from pathogens. Curr. Opin. Microbiol. 56, 59–66 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Mäkivuokko, H. et al. Association between the ABO blood group and the human intestinal microbiota composition. BMC Microbiol. 12, 94 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Liu, X. et al. Inter-determination of blood metabolite levels and gut microbiome supported by Mendelian randomization. Preprint at bioRxiv https://doi.org/10.1101/2020.06.30.181438 (2020).

  30. Motta, V., Luise, D., Bosi, P. & Trevisi, P. Faecal microbiota shift during weaning transition in piglets and evaluation of AO blood types as shaping factor for the bacterial community profile. PLoS One 14, e0217001 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yang, H., et al. An ancient deletion in the ABO gene affects the composition of the porcine microbiome by altering intestinal N-acetyl-galactosamine concentrations. Preprint at bioRxiv https://www.biorxiv.org/content/10.1101/2020.07.16.206219v1 (2020).

  32. Chen, L. et al. The long-term genetic stability and individual specificity of the human gut microbiome. Cell 184, 2302–2315 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Bersaglieri, T. et al. Genetic signatures of strong recent positive selection at the lactase gene. Am. J. Hum. Genet. 74, 1111–1120 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Auer, P. L. et al. Imputation of exome sequence variants into population- based samples and blood-cell-trait-associated loci in African Americans: NHLBI GO Exome Sequencing Project. Am. J. Hum. Genet. 91, 794–808 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Ségurel, L. et al. The ABO blood group is a trans-species polymorphism in primates. Proc. Natl Acad. Sci. USA 109, 18493–18498 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Band, G. et al. Insights into malaria susceptibility using genome-wide data on 17,000 individuals from Africa, Asia and Oceania. Nat. Commun. 10, 5732 (2019).

    Article  Google Scholar 

  37. Barua, D. & Paguio, A. S. ABO blood groups and cholera. Ann. Hum. Biol. 4, 489–492 (1977).

    Article  CAS  PubMed  Google Scholar 

  38. Watanabe, K. et al. A global overview of pleiotropy and genetic architecture in complex traits. Nat. Genet. 51, 1339–1348 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. Naitza, S. et al. A genome-wide association scan on the levels of markers of inflammation in sardinians reveals associations that underpin its complex regulation. PLoS Genet. 8, e1002480 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Suzuki, T. A. et al. The role of the microbiota in human genetic adaptation. Science 370, eaaz6827 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Parks, D. H. et al. A complete domain-to-species taxonomy for Bacteria and Archaea. Nat. Biotechnol. 38, 1079–1086 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Wade, K. H. & Hall, L. J. Improving causality in microbiome research: can human genetic epidemiology help? Wellcome Open Res. 4, 199 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Pirastu, N. et al. Using genetics to disentangle the complex relationship between food choices and health status. Preprint at bioRxiv https://doi.org/10.1101/829952 (2019).

  44. Wang, C. et al. High-salt diet has a certain impact on protein digestion and gut microbiota: a sequencing and proteome combined study. Front. Microbiol. 8, 1838 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Culligan, E. P. et al. Combined metagenomic and phenomic approaches identify a novel salt tolerance gene from the human gut microbiome. Front. Microbiol. 5, 189 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  47. Zhong, V. W. et al. A genome-wide association study of bitter and sweet beverage consumption. Hum. Mol. Genet. 28, 2449–2457 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bjørkhaug, S. T. et al. Characterization of gut microbiota composition and functions in patients with chronic alcohol overconsumption. Gut Microbes 10, 663–675 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Sanna, S. et al. Common variants in the GDF5-UQCC region are associated with variation in human height. Nat. Genet. 40, 198–203 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Weedon, M. N. et al. A common variant of HMGA2 is associated with adult and childhood height in the general population. Nat. Genet. 39, 1245–1250 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Loos, R. J. F. et al. Common variants near MC4R are associated with fat mass, weight and risk of obesity. Nat. Genet. 40, 768–775 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Tigchelaar, E. F. et al. Cohort profile: LifeLines DEEP, a prospective, general population cohort study in the northern Netherlands: study design and baseline characteristics. BMJ Open 5, e006772 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Scholtens, S. et al. Cohort Profile: LifeLines, a three-generation cohort study and biobank. Int. J. Epidemiol. 44, 1172–1180 (2015).

    Article  PubMed  Google Scholar 

  54. the Haplotype Reference Consortium. et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 48, 1279–1283 (2016).

    Article  Google Scholar 

  55. Lopera Maya, E. A. et al. Lack of association between genetic variants at ACE2 and TMPRSS2 genes involved in SARS-CoV-2 infection and human quantitative phenotypes. Front. Genet. 11, 613 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  56. The 1000 Genomes Project Consortium. A global reference for human genetic variation. Nature 526, 68–74 (2015).

    Article  Google Scholar 

  57. Francioli, L. C. et al. Whole-genome sequence variation, population structure and demographic history of the Dutch population. Nat. Genet. 46, 818–825 (2014).

    Article  CAS  Google Scholar 

  58. Truong, D. T. et al. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat. Methods 12, 902–903 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Franzosa, E. A. et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15, 962–968 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Suzek, B. E. et al. UniRef clusters: a comprehensive and scalable alternative for improving sequence similarity searches. Bioinforma. Oxf. Engl. 31, 926–932 (2015).

    Article  CAS  Google Scholar 

  61. Buchfink, B., Xie, C. & Huson, D. H. Fast and sensitive protein alignment using DIAMOND. Nat. Methods 12, 59–60 (2015).

    Article  CAS  PubMed  Google Scholar 

  62. Siebelink, E., Geelen, A. & de Vries, J. H. M. Self-reported energy intake by FFQ compared with actual energy intake to maintain body weight in 516 adults. Br. J. Nutr. 106, 274–281 (2011).

    Article  CAS  PubMed  Google Scholar 

  63. Willett, W. C. Nutritional Epidemiology (Oxford Univ. Press, 2012).

  64. Zheng, X. et al. SAIGEgds: an efficient statistical tool for large-scale PheWAS with mixed models. Bioinformatics 37, 728–730 (2021).

    Article  CAS  PubMed  Google Scholar 

  65. The Severe Covid-19 GWAS Group Genome-wide association study of severe COVID-19 with respiratory failure. N. Engl. J. Med. 383, 1522–1534 (2020).

    Article  Google Scholar 

  66. Chang, C. C. et al. Second-generation PLINK: rising to the challenge of larger and richer datasets. GigaScience 4, 7 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Elsworth, B. et al. The MRC IEU OpenGWAS data infrastructure. Preprint at bioRxiv https://doi.org/10.1101/2020.08.10.244293 (2020).

  68. Hemani, G. et al. The MR-Base platform supports systematic causal inference across the human phenome. eLife 7, e34408 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Burgess, S. & Thompson, S. G. Mendelian Randomization Methods for Using Genetic Variants in Causal Estimation (CRC Press, 2015).

  70. Bowden, J., Davey Smith, G. & Burgess, S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int. J. Epidemiol. 44, 512–525 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Teslovich, T. M. et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466, 707–713 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge the services of the LifeLines Cohort Study, the contributing research centers delivering data to LifeLines and all the study participants. The LifeLines initiative was made possible by subsidy from the Dutch Ministry of Health, Welfare and Sport; the Dutch Ministry of Economic Affairs; the University Medical Center Groningen (UMCG); the University of Groningen (UG) and the Provinces of the North of the Netherlands (Drenthe, Friesland and Groningen). This project was carried out under LifeLines project number OV18_0464. We thank Mathieu Plateel and Jody Geelderloos-Arends for their contribution in genotyping the LifeLines samples, Kate McIntyre for help developing the manuscript, Marije van der Geest for setting up the website for sharing summary statistics and Patrick Deelen for discussion of results. We also thank the UMCG Genomics Coordination Center, the UG Center for Information Technology and their sponsors (BBMRI-NL and TarGet) for storage and computational infrastructure and Novogene for providing gut metagenome sequencing of all DMP samples. Finally, we thank the UK Biobank for making their resource available. Analyses of UK Biobank data described in this work were carried out under project number 48548 to C.W. The generation and management of genotype data for the LifeLines Cohort Study was supported by the UMCG Genetics LifeLines Initiative. Genotyping quality control was supported by UMCG (HAP grant CD017.0031/ronde 2017-2/nr 324). Metagenomics sequencing of the cohort was mainly funded by the CardioVasculair Onderzoek Nederland (CVON) (grant CVON 2012-03) to M. Hofken (who died in 2016), J.F. and A.Z., as well as other grants to R.K.W. and C.W. (listed below). This work was further supported by the collaborative TIMID project LSHM18057-SGF financed by the allowance made available by Top Sector Life Sciences & Health to Samenwerkende Gezondheidsfondsen to stimulate public/private partnerships and cofinancing by health foundations that are part of the Samenwerkende Gezondheidsfondsen (R.K.W.); the the Seerave Foundation (R.K.W.); European Research Council (ERC) starting grant 715772 (S.Z.), consolidator grant 101001678 (J.F.) and advanced grant ERC-671274 (C.W.); Netherlands Organization for Scientific Research VIDI grant 016.178.056 (A.Z.), gravitation grant ExposomeNL 024.004.017 (A.Z.), VICI grant VI.C.202.022 (J.F.), gravitation grant The Netherlands Organ-on-Chip Initiative 024.003.001 (C.W.) and Spinoza award NWOSPI 92-266 (C.W.); CVON grant 2018-27 (A.Z. and J.F.); the EurHealth-1Health INTERREG V A 202085 project (H.J.M.H.); Foundation De Cock-Hadders grant 20:20-13 (L.C.); a joint fellowship from the UMCG and China Scholarship Council (CSC201708320268 to L.C.); and Colciencias fellowship ed.783 (E.A.L.-M.).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

E.A.L.-M., A.K. and S.H. performed genetic analyses. A.v.d.G. performed MR and power analyses. A.K., S.H., L.C., A.V.V. and R.G. processed microbiome data. S.A.-S., T.S., V.C., M.A.Y.K., L.A.B. and M.F.B.G. processed samples meta-data. P.B.T.N. and M.A.S. provided resources on the HP computing cluster and assisted with data management. H.J.M.H., C.W., J.F., R.K.W. and A.Z. provided funding resources and designed the DMP. A.Z. and S.S. supervised the study. E.A.L.-M., A.K., A.v.d.G., S.H., S.A.S., A.Z. and S.S. drafted the manuscript. All authors were involved in data interpretation and provided critical input to the manuscript draft.

Corresponding authors

Correspondence to Alexandra Zhernakova or Serena Sanna.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks A. Franke and T. Zhang for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Cladogram plot tree of taxonomic relations between bacteria of the class Actinobacteria and their associations with host genetics.

Each node shows a taxonomic level (from outside to inside: phylogenetic group, phylum, class, order, family, genus and species). Note that branch lengths do not represent phylogenetic distance. Inner labels represent genetic locus. External labels represent the clade. Nodes with dotted lines indicate that the GWAS was not performed for that taxa. Node color corresponds to different levels of significance as described in the legend. a, Depicts associations detected at the MCM6/LCT locus with each taxa, using the most significant p-value observed between rs4988235 and rs182549. b, Depicts associations at the ABO locus with each taxa, using the most significant p-value observed between rs8176645 and rs550057.

Extended Data Fig. 2 Association at the LCT locus and interaction with lactose intake in other members of family Bifidobacteriaceae.

Relative abundances of taxa, natural log–transformed and adjusted by age and sex, compared between LP (rs182549 C/T or T/T) and LI (rs182549 C/C) participants and among individuals with low or high daily lactose intake levels. The y axis represents the relative abundance of the microbial feature, natural log–transformed and adjusted by age and sex. Density distribution is displayed with violin plots, while boxplots represent summary statistics: the center line represents the median, the box hinges represent the lower and upper quartiles (percentiles 25 and 75) of the distribution, the upper whisker extends to the maximum value no further than 1.5*IQR (where IQR is the interquartile range) from the upper hinge, the lower whisker extends to the minimum value no further than 1.5*IQR from the lower hinge, and data beyond the end of the whiskers are outliers plotted as individual points. a and c, Relative abundances for the taxa between LP and LI participants. b and d, Comparisons of abundance between lactose intake levels, low (<first quartile) and high (≥ first quartile), stratified by lactose persistence status. The distributions are shown for s. Bifidobacterium adolescentis (top) and s. Bifidobacterium longum (bottom). P-values were obtained with a two-sided Wilcoxon rank test. n: number of participants.

Extended Data Fig. 3 Graphical representation of MR results with a Benjamini–Hochberg FDR q value < 0.1.

a, Effect size in standard deviation units of 3 variants associated with Alistipes abundance changes that were used as instrumental variables (effects estimated on 7,728 independent samples) (x-axis) versus effect size in standard deviation units of the same variants for salt intake (estimated effects estimated on 462,630 independent samples) (y-axis). Error bars represent standard errors (SE) of each effect size (beta + SE and beta-SE). The orange and blue lines represent lines whose slope is the causal estimate from MR methods IVW and Egger, respectively. b, A plot similar to a, but the x axis is the effect size in standard deviation units for instrumental variants selected for Collinsella (effects estimated on 7,210 independent samples) abundance and on the y-axis for Triglyceride levels (effects estimated on 343,992 independent individuals).

Supplementary information

Supplementary Note

The file contains supplementary text, discussion and references.

Reporting Summary.

Peer Review Information.

Supplementary Tables

Supplementary Tables 1–13.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lopera-Maya, E.A., Kurilshikov, A., van der Graaf, A. et al. Effect of host genetics on the gut microbiome in 7,738 participants of the Dutch Microbiome Project. Nat Genet 54, 143–151 (2022). https://doi.org/10.1038/s41588-021-00992-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00992-y

This article is cited by

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