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.

  • Article
  • Published:

Integration of epidemiologic, pharmacologic, genetic and gut microbiome data in a drug–metabolite atlas

Abstract

Progress in high-throughput metabolic profiling provides unprecedented opportunities to obtain insights into the effects of drugs on human metabolism. The Biobanking BioMolecular Research Infrastructure of the Netherlands has constructed an atlas of drug–metabolite associations for 87 commonly prescribed drugs and 150 clinically relevant plasma-based metabolites assessed by proton nuclear magnetic resonance. The atlas includes a meta-analysis of ten cohorts (18,873 persons) and uncovers 1,071 drug–metabolite associations after evaluation of confounders including co-treatment. We show that the effect estimates of statins on metabolites from the cross-sectional study are comparable to those from intervention and genetic observational studies. Further data integration links proton pump inhibitors to circulating metabolites, liver function, hepatic steatosis and the gut microbiome. Our atlas provides a tool for targeted experimental pharmaceutical research and clinical trials to improve drug efficacy, safety and repurposing. We provide a web-based resource for visualization of the atlas (http://bbmri.researchlumc.nl/atlas/).

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

Access options

Buy this article

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

Fig. 1: Drug–metabolite associations in model 1 versus model 2.
Fig. 2: Drug–metabolite associations in model 2 versus model 3.
Fig. 3: Drug–metabolite associations in model 3 versus significance after disentangling the indicated disease/endophenotype effect.
Fig. 4: Comparison of statin–metabolite associations between cross-sectional, longitudinal and genetic studies.
Fig. 5: Integrated data of PPIs, metabolites, liver function measurements and gut microbiome.

Similar content being viewed by others

Data availability

All summary statistics of the meta-analysis, and those utilized in compilation of the figures, are made available through the Supplementary tables. In regard to the availability of the raw data, the analyses are based on a meta-analysis of multiple Dutch studies. The raw metabolomics data of the studies are pooled in a single database. The quantified metabolic biomarker datasets included in this study are available through the BBMRI-NL website http://www.bbmri.nl/omics-metabolomics/, where details of how to access the data through centralized computational facilities are described. To request data, researchers are required to fill out and sign the data access request and code-of-conduct forms. Applications compliant with ethical and legal legislations will be reviewed by the BBMRI-NL board in regard to overlap with other ongoing projects before access is granted. Data on medication used in the current study are available through the individual studies on reasonable request. To obtain these, the principal investigator of the cohorts can be contacted through http://www.bbmri.nl/omics-metabolomics/. No custom code or mathematical algorithm was used in the current study.

References

  1. Patti, G. J., Yanes, O. & Siuzdak, G. Innovation: metabolomics: the apogee of the omics trilogy. Nat. Rev. Mol. Cell Biol. 13, 263–269 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Park, J. E., Lim, H. R., Kim, J. W. & Shin, K. H. Metabolite changes in risk of type 2 diabetes mellitus in cohort studies: a systematic review and meta-analysis. Diabetes Res. Clin. Pract. 140, 216–227 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. McGarrah, R. W., Crown, S. B., Zhang, G. F., Shah, S. H. & Newgard, C. B. Cardiovascular metabolomics. Circ. Res. 122, 1238–1258 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Liu, J. et al. A Mendelian randomization study of metabolite profiles, fasting glucose, and type 2 diabetes. Diabetes 66, 2915–2926 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Wang, T. J. et al. Metabolite profiles and the risk of developing diabetes. Nat. Med. 17, 448–453 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. van der Lee, S. J. et al. Circulating metabolites and general cognitive ability and dementia: evidence from 11 cohort studies. Alzheimers Dement. 14, 707–722 (2018).

    Article  PubMed  Google Scholar 

  7. Mapstone, M. et al. Plasma phospholipids identify antecedent memory impairment in older adults. Nat. Med. 20, 415–418 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Thorburn, A. N. et al. Evidence that asthma is a developmental origin disease influenced by maternal diet and bacterial metabolites. Nat. Commun. 6, 7320 (2015).

    Article  CAS  PubMed  Google Scholar 

  9. Mabalirajan, U. et al. Linoleic acid metabolite drives severe asthma by causing airway epithelial injury. Sci. Rep. 3, 1349 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Illig, T. et al. A genome-wide perspective of genetic variation in human metabolism. Nat. Genet. 42, 137–141 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Kettunen, J. et al. Genome-wide study for circulating metabolites identifies 62 loci and reveals novel systemic effects of LPA. Nat. Commun. 7, 11122 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Draisma, H. H. et al. Genome-wide association study identifies novel genetic variants contributing to variation in blood metabolite levels. Nat. Commun. 6, 7208 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Suhre, K. et al. Human metabolic individuality in biomedical and pharmaceutical research. Nature 477, 54–60 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Beger, R. D. et al. Metabolomics enables precision medicine: ‘A White Paper, Community Perspective’. Metabolomics 12, 149 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Rappaport, S. M., Barupal, D. K., Wishart, D., Vineis, P. & Scalbert, A. The blood exposome and its role in discovering causes of disease. Environ. Health Perspect. 122, 769–774 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Schutte, B. A. et al. The effect of standardized food intake on the association between BMI and (1)H-NMR metabolites. Sci. Rep. 6, 38980 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wurtz, P. et al. Metabolomic profiling of statin use and genetic inhibition of HMG-CoA reductase. J. Am. Coll. Cardiol. 67, 1200–1210 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Altmaier, E. et al. Metabolomics approach reveals effects of antihypertensives and lipid-lowering drugs on the human metabolism. Eur. J. Epidemiol. 29, 325–336 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Elbadawi-Sidhu, M. et al. Pharmacometabolomic signature links simvastatin therapy and insulin resistance. Metabolomics 13, 11 (2017).

    Article  PubMed  CAS  Google Scholar 

  20. Kaddurah-Daouk, R. et al. Lipidomic analysis of variation in response to simvastatin in the Cholesterol and Pharmacogenetics Study. Metabolomics 6, 191–201 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xu, T. et al. Effects of metformin on metabolite profiles and LDL cholesterol in patients with type 2 diabetes. Diabetes Care 38, 1858–1867 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. t Hart, L. M. et al. Blood metabolomic measures associate with present and future glycemic control in type 2 diabetes. J. Clin. Endocrinol. Metab. 103, 4569–4579 (2018).

    Article  Google Scholar 

  23. Moosavinasab, S. et al. ‘RE:fine drugs’: an interactive dashboard to access drug repurposing opportunities. Database https://doi.org/10.1093/database/baw083 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Voora, D. & Shah, S. H. Pharmacometabolomics meets genetics: a ‘natural’ clinical trial of statin effects. J. Am. Coll. Cardiol. 67, 1211–1213 (2016).

    Article  PubMed  Google Scholar 

  25. Wishart, D. S. Emerging applications of metabolomics in drug discovery and precision medicine. Nat. Rev. Drug Discov. 15, 473–484 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Van Norman, G. A. Drugs, devices, and the FDA: Part 1: an overview of approval processes for drugs. JACC Basic Transl. Sci. 1, 170–179 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  27. US Food and Drug Administration. 22 Case studies where phase 2 and phase 3 trials had divergent results https://www.fda.gov/about-fda/reports/22-case-studies-where-phase-2-and-phase-3-trials-had-divergent-results (2017).

  28. Brahma, D. K., Wahlang, J. B., Marak, M. D. & Ch Sangma, M. Adverse drug reactions in the elderly. J. Pharmacol. Pharmacother. 4, 91–94 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Wurtz, P. et al. Quantitative serum nuclear magnetic resonance metabolomics in large-scale epidemiology: a primer on -omic technologies. Am. J. Epidemiol. 186, 1084–1096 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Ahola-Olli, A. V. et al. Circulating metabolites and the risk of type 2 diabetes: a prospective study of 11,896 young adults from four Finnish cohorts. Diabetologia 62, 2298–2309 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ference, B. A. et al. Association of triglyceride-lowering LPL variants and LDL-C-lowering LDLR variants with risk of coronary heart disease. JAMA 321, 364–373 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Holmes, M. V. et al. Lipids, lipoproteins, and metabolites and risk of myocardial infarction and stroke. J. Am. Coll. Cardiol. 71, 620–632 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Onderwater, G. L. J. et al. Large-scale plasma metabolome analysis reveals alterations in HDL metabolism in migraine. Neurology 92, e1899–e1911 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Struja, T. et al. Metabolomics for prediction of relapse in Graves’ disease: observational pilot study. Front. Endocrinol. (Lausanne) 9, 623 (2018).

    Article  Google Scholar 

  35. Deelen, J. et al. A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals. Nat. Commun. 10, 3346 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Fischer, K. et al. Biomarker profiling by nuclear magnetic resonance spectroscopy for the prediction of all-cause mortality: an observational study of 17,345 persons. PLoS Med. 11, e1001606 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  37. 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 

  38. Bajaj, J. S. et al. Proton pump inhibitor initiation and withdrawal affects gut microbiota and readmission risk in cirrhosis. Am. J. Gastroenterol. 113, 1177–1186 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Imhann, F. et al. Proton pump inhibitors affect the gut microbiome. Gut 65, 740–748 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Llorente, C. et al. Gastric acid suppression promotes alcoholic liver disease by inducing overgrowth of intestinal Enterococcus. Nat. Commun. 8, 837 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Jackson, M. A. et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut 65, 749–756 (2016).

    Article  PubMed  CAS  Google Scholar 

  42. Liu, R. et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat. Med. 23, 859–868 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Pedersen, H. K. et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535, 376–381 (2016).

    Article  CAS  PubMed  Google Scholar 

  44. Kontush, A. HDL particle number and size as predictors of cardiovascular disease. Front. Pharmacol. 6, 218 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Ahola-Olli, A. V. et al. Circulating metabolites and the risk of type 2 diabetes: a prospective study of 11,896 young adults from four Finnish cohorts. Diabetologia 62, 2298–2309 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Mitchell, A. J., Vaze, A. & Rao, S. Clinical diagnosis of depression in primary care: a meta-analysis. Lancet 374, 609–619 (2009).

    Article  PubMed  Google Scholar 

  47. Bajaj, J. S. et al. Systems biology analysis of omeprazole therapy in cirrhosis demonstrates significant shifts in gut microbiota composition and function. Am. J. Physiol. Gastrointest. Liver Physiol. 307, G951–957 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Hoyles, L. et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat. Med. 24, 1070–1080 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Qin, N. et al. Alterations of the human gut microbiome in liver cirrhosis. Nature 513, 59–64 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Bates, C., Adams, W. & Handschumacher, R. Control of the formation of uridine diphospho-N-acetyl-hexosamine and glycoprotein synthesis in rat liver. J. Biol. Chemi. 241, 1705–1712 (1966).

    Article  CAS  Google Scholar 

  51. Kettunen, J. et al. Biomarker glycoprotein acetyls is associated with the risk of a wide spectrum of incident diseases and stratifies mortality risk in angiography patients. Circ. Genom. Precis. Med. 11, e002234 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Akinkuolie, A. O., Buring, J. E., Ridker, P. M. & Mora, S. A novel protein glycan biomarker and future cardiovascular disease events. J. Am. Heart Assoc. 3, e001221 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Akinkuolie, A. O., Pradhan, A. D., Buring, J. E., Ridker, P. M. & Mora, S. Novel protein glycan side-chain biomarker and risk of incident type 2 diabetes mellitus. Arterioscler. Thromb. Vasc. Biol. 35, 1544–1550 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ikram, M. A. et al. The Rotterdam Study: 2018 update on objectives, design and main results. Eur. J. Epidemiol. 32, 807–850 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Boomsma, D. I. et al. Netherlands Twin Register: from twins to twin families. Twin Res. Hum. Genet. 9, 849–857 (2006).

    Article  PubMed  Google Scholar 

  56. Penninx, B. W. et al. The Netherlands Study of Depression and Anxiety (NESDA): rationale, objectives and methods. Int. J. Methods Psychiatr. Res. 17, 121–140 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Schoenmaker, M. et al. Evidence of genetic enrichment for exceptional survival using a family approach: the Leiden Longevity Study. Eur. J. Hum. Genet. 14, 79–84 (2006).

    Article  PubMed  Google Scholar 

  58. 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 

  59. van der Heijden, A. A. et al. The Hoorn Diabetes Care System (DCS) cohort. A prospective cohort of persons with type 2 diabetes treated in primary care in the Netherlands. BMJ Open 7, e015599 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Geleijnse, J. M. et al. Effect of low doses of n-3 fatty acids on cardiovascular diseases in 4,837 post-myocardial infarction patients: design and baseline characteristics of the Alpha Omega Trial. Am. Heart J. 159, 539–546 e532 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. Schram, M. T. et al. The Maastricht Study: an extensive phenotyping study on determinants of type 2 diabetes, its complications and its comorbidities. Eur. J. Epidemiol. 29, 439–451 (2014).

    Article  PubMed  Google Scholar 

  62. Sayed-Tabatabaei, F. A. et al. Heritability of the function and structure of the arterial wall: findings of the Erasmus Rucphen Family (ERF) study. Stroke 36, 2351–2356 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. van Oosterhout, W. P. et al. Validation of the web-based LUMINA questionnaire for recruiting large cohorts of migraineurs. Cephalalgia 31, 1359–1367 (2011).

    Article  PubMed  Google Scholar 

  64. de Mutsert, R. et al. The Netherlands Epidemiology of Obesity (NEO) study: study design and data collection. Eur. J. Epidemiol. 28, 513–523 (2013).

    Article  PubMed  Google Scholar 

  65. Soininen, P., Kangas, A. J., Wurtz, P., Suna, T. & Ala-Korpela, M. Quantitative serum nuclear magnetic resonance metabolomics in cardiovascular epidemiology and genetics. Circ. Cardiovasc. Genet. 8, 192–206 (2015).

    Article  CAS  PubMed  Google Scholar 

  66. Inouye, M. et al. Metabonomic, transcriptomic, and genomic variation of a population cohort. Mol. Syst. Biol. 6, 441 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  67. van den Akker, E. et al. Predicting biological age based on the BBMRI-NL 1H-NMR metabolomics repository. Preprint at bioRxiv, 632919 (2019).

  68. Sturm, R. The effects of obesity, smoking, and drinking on medical problems and costs. Health Aff. (Millwood) 21, 245–253 (2002).

    Article  Google Scholar 

  69. Van Gaal, L. F., Mertens, I. L. & De Block, C. E. Mechanisms linking obesity with cardiovascular disease. Nature 444, 875–880 (2006).

    Article  PubMed  CAS  Google Scholar 

  70. Li, J. & Ji, L. Adjusting multiple testing in multilocus analyses using the eigenvalues of a correlation matrix. Heredity (Edinb.) 95, 221–227 (2005).

    Article  CAS  Google Scholar 

  71. Viechtbauer, W. Conducting meta-analyses in R with the metafor package. J. Stat. Softw. 36, 1–48 (2010).

    Article  Google Scholar 

  72. Lawlor, D. A., Harbord, R. M., Sterne, J. A., Timpson, N. & Davey Smith, G. Mendelian randomization: using genes as instruments for making causal inferences in epidemiology. Stat. Med. 27, 1133–1163 (2008).

    Article  PubMed  Google Scholar 

  73. Details and Considerations of the UK Biobank GWAS (Neale Lab, accessed 10 December 2018); http://www.nealelab.is/blog/2017/9/11/details-and-considerations-of-the-uk-biobank-gwas

  74. Wray, N. R. et al. Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat. Genet. 50, 668–681 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. 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 

  76. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 106, 3143–3421 (2002).

  77. Bot, M. et al. Metabolomics profile in depression: a pooled analysis of 230 metabolic markers in 5,283 cases with depression and 10,145 controls. Biol. Psychiatr. (2019).

  78. Koehler, E. M. et al. Presence of diabetes mellitus and steatosis is associated with liver stiffness in a general population: the Rotterdam Study. Hepatology 63, 138–147 (2016).

    Article  CAS  PubMed  Google Scholar 

  79. Hamaguchi, M. et al. The severity of ultrasonographic findings in nonalcoholic fatty liver disease reflects the metabolic syndrome and visceral fat accumulation. Am. J. Gastroenterol. 102, 2708–2715 (2007).

    Article  PubMed  Google Scholar 

  80. Vojinovic, D. et al. Relationship between gut microbiota and circulating metabolites in population-based cohorts. Nat. Commun. 10, 5813 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge all participants included in the cohorts. We also acknowledge the BBMRI Metabolomics Consortium (see Supplementary Information, http://www.bbmri.nl/omics-metabolomics/) funded by BBMRI-NL, a research infrastructure financed by the Dutch government through the Netherlands Organisation for Scientific Research (NWO) (grant nos. 184.021.007 and 184033111). This work is part of the CardioVasculair Onderzoek Nederland (CVON 2012-03), the Common mechanisms and pathways in Stroke and Alzheimer’s disease (CoSTREAM) project (www.costream.eu, grant agreement no. 667375), the Memorabel program (project no. 733050814), Netherlands X-omics Research Infrastructure and U01-AG061359 NIA. The full list of funding information for each cohort can be found in the cohort acknowledgements below. J.L., C.M.v.D. and A.D. benefitted from exchange grants from the Personalized pREvention of Chronic DIseases consortium (no. H2020-MSCA-RISE-2014). A.D. is supported by the Dutch Science Organization (ZonMW-VENI, grant no. 2015). L.C. is supported by a joint PhD fellowship from China Scholarship Council (no. 201708320268) and University of Groningen. M.G.N. is supported by Royal Netherlands Academy of Science Professor Award (no. PAH/6635) to D.I.B. D.O.M.-K. is supported by the ZonMW-VENI (grant no. 916.14.023). B.H.C.S. received a grant from TransQST (no. 116030-2; IMI2). D.R. is funded by an Erasmus MC mRACE grant (Profiling of the human gut microbiome). Cohort acknowledgements, ERF: we are grateful to all study participants and their relatives, general practitioners and neurologists for their contributions and to P. Veraart for her help in genealogy, J. Vergeer for supervision of the laboratory work and P. Snijders for his help in data collection. ERF was supported by the Consortium for Systems Biology (NCSB), both within the framework of the Netherlands Genomics Initiative (NGI)/NWO). The ERF study as a part of European Special Populations Research Network (EUROSPAN) was supported by European Commission FP6 STRP, grant no. 018947 (LSHG-CT-2006-01947) and also received funding from the European Community’s Seventh Framework Programme (FP7/2007-2013)/grant agreement no. HEALTH-F4-2007-201413 by the European Commission under the program ‘Quality of Life and Management of the Living Resources’ of the 5th Framework Programme (no. QLG2-CT-2002-01254), as well as the FP7 project EUROHEADPAIN (no. 602633). High-throughput analysis of ERF data was supported by a joint grant from NWO and the Russian Foundation for Basic Research (NWO-RFBR no. 047.017.043). High-throughput metabolomics measurements in the ERF study were supported by BBMRI-NL. Rotterdam Study: we thank the study subjects, the staff from Rotterdam Study and the participating pharmacists and general practitioners. The Rotterdam Study is supported by Erasmus MC and Erasmus University Rotterdam; by NWO, the Netherlands Organisation for Health Research and Development (ZonMw), the Research Institute for Diseases in the Elderly (RIDE), the Netherlands Genomics Initiative (NGI), the Ministry of Education, Culture and Science, the Ministry of HealthWelfare and Sports, the European Commission (DG XII) and the Municipality of Rotterdam. L.L. reports expert consultation from Boehringer Ingelheim and Novartis, and unrestricted grants from AstraZeneca and Chiesi. We are also grateful to nurse ultrasonographist, Mrs. van Wijngaarden for performing abdominal ultrasonography and liver stiffness measurements. The generation and management of stool microbiome data for Rotterdam Study (Rotterdam Study III-2) were executed by the Human Genotyping Facility of the Genetic Laboratory of the Department of Internal Medicine, Erasmus MC, Rotterdam. We thank N. El Faquir and J. Verkroost-Van Heemst for their help in sample collection and registration, and P. van der Wal, K. Arabe, H. Razawy and K. Singh Asra for their help in DNA isolation and sequencing. Furthermore, we thank J. Raes and J. Wang (KU Leuven, Belgium) for their guidance in 16 S rRNA profiling and dataset generation. NTR: we are grateful to all twins and their relatives for their continued participation. Funding was obtained from NWO and MagW/ZonMW (grant nos. 904-61-090, 985-10-002, 904-61-193,480-04-004, 400-05-717, Addiction-31160008, Middelgroot-911-09-032 and Spinozapremie 56-464-14192); BBMRI-NL (no. 184.021.007); VU University’s Institute for Health and Care Research (no. EMGO1); Neuroscience Campus Amsterdam (NCA); the European Community’s Seventh Framework Program (no. FP7/2007-2013); ENGAGE (no. HEALTH-F4-2007-201413); and the European Science Council (ERCAdvanced, no. 230374). M.G.N. is supported by the ZonMw grant, ‘Genetics as a research tool: a natural experiment to elucidate the causal effects of social mobility on health’ (pnr:531003014), ZonMw project: ‘Can sex- and gender-specific gene expression and epigenetics explain sex-differences in disease prevalence and etiology?’ (pnr:849200011) and grant no. R01AG054628 02 S. NESDA: the infrastructure for the NESDA study (www.nesda.nl) was funded through the Geestkracht program of ZonMw (grant no. 10-000-1002) and through financial contributions of participating universities and mental health care organizations (VU University Medical Center, GGZ inGeest, Leiden University Medical Center, Leiden University, GGZ Rivierduinen, University Medical Center Groningen, University of Groningen, Lentis, GGZ Friesland, GGZ Drenthe and Rob Giel Onderzoekscentrum). LLS: we thank all participants. This study was supported by a grant from the Innovation-Oriented Research Program on Genomics (SenterNovem, no. IGE05007), the Centre for Medical Systems Biology and the Netherlands Consortium for Healthy Ageing (grant no. 050-060-810), all within the framework of NWO by the BBMRI Metabolomics Consortium funded by BBMRI-NL (NWO, grant nos. 184.021.007 and 184033111). LifeLines DEEP: we thank participants and staff of the LifeLines DEEP cohort for their collaboration. We thank J. Dekens, M. Platteel, A. Maatman and J. Arends for management and technical support. This project was funded by the Netherlands Heart Foundation (IN-CONTROL CVON, grant no. 2012-03 to A.Z. and J.F.; by NWO (nos. NWO-VIDI 864.13.013 to J.F. and NWO-VIDI 016.178.056 to A.Z.; and by the European Research Council Starting Grant no. 715772 to A.Z., who also holds a Rosalind Franklin Fellowship from the University of Groningen. Hoorn DCS: we thank participants of this study and research staff of the Diabetes Care System West-Friesland. High-throughput metabolomics measurements in the DCS study were supported by BBMRI-NL and the Parelsnoer Initiative which is part of, and is funded by, the Dutch Federation of University Medical Centres and, from 2007 to 2011, received initial funding from the Dutch Government. To perform additional research (in subsamples of the DCS cohort), funding was received from several sources including the Dutch Federation of University Medical Centres, health insurers, NWO, ZonMw, the Dutch Diabetes Foundation, the European Foundation for the Study of Diabetes, International Diabetes Federation, the European Innovative Medicine Initiative and the European Union. Alpha Omega Cohort: the Alpha Omega Cohort is registered with ClinicalTrials.gov. (identifier: NCT03192410). It was funded by the Netherlands Heart Foundation (grant no. 200T401) and the National Institutes of Health (NIH, grant no. R01HL076200). J.M.G. received funding from Unilever for analyses of dietary and circulating fatty acids in the Alpha Omega Cohort. High-throughput metabolomics measurements for the Alpha Omega Cohort were supported by BBMRI-NL. TMS: this study was supported by the European Regional Development Fund via OP-Zuid, the Province of Limburg, the Dutch Ministry of Economic Affairs (grant no. 31 O.041), Stichting De Weijerhorst (Maastricht, the Netherlands), the Pearl String Initiative Diabetes (Amsterdam, the Netherlands), CARIM School for Cardiovascular Diseases (Maastricht, the Netherlands), Stichting Annadal (Maastricht, the Netherlands), Health Foundation Limburg (Maastricht, the Netherlands) and by unrestricted grants from Janssen-Cilag B.V. (Tilburg, the Netherlands), Novo Nordisk Farma B.V. (Alphen aan den Rijn, the Netherlands) and Sanofi-Aventis Netherlands B.V. (Gouda, the Netherlands). LUMINA: the LUMINA study is funded by grants obtained from ZonMw (no. 90700217) and VIDI (ZonMw, no. 91711319) (to G.M.T.); by NCSB and the Centre for Medical System Biology (CMSB), both within the framework of the Netherlands Genomics Initiative (NGI)/NWO (to A.M.J.M.v.d.M.) and by the FP7 EU project EUROHEADPAIN (grant no. 602633) (to A.M.J.M.v.d.M. and G.M.T.). NEO: the authors of the NEO study thank all individuals who participated, all participating general practitioners for inviting eligible participants and all research nurses for collection of the data. We thank the NEO study group—P. van Beelen, P. Noordijk and I. de Jonge—for coordination, laboratory and data management of the study. Genotyping in the NEO study was supported by Centre National de Génotypage (Paris, France), headed by J.-F. Deleuze. This study was supported by the participating departments, the Division and the Board of Directors of Leiden University Medical Center and by Leiden University, Research Profile Area Vascular and Regenerative Medicine. The study was also supported by the Netherlands Cardiovascular Research Initiative: an initiative with the support of the Dutch Heart Foundation (no. CVON2014-02 ENERGISE).

Author information

Authors and Affiliations

Authors

Contributions

A.D., J.F., J.M.G., L.L., J.L., M.N., L.M.t’H, C.M.v.D. and A.Z. contributed to study design. M. Beekman, J.W.J.B., D.I.B., I.d.B., P.J.M.E., J.F., J.M.G., D.O.M.-K., G.L.J.O., B.W.J.H.P., F.R., P.E.S., C.D.A.S., B.H.C.S., L.M.t’H., G.M.T., A.M.J.M.v.d.M., A.A.W.A.v.d.H., G.J.H.v.d.K., K.W.v.D., C.M.v.D., M.M.J.v.G., G.W., R.K., S.D.M., A.G.U. and A.Z. contributed to cohort design and management. I.C.W.A., M. Beekman, D.I.B., I.d.B., J.F., J.M.G., T.H., G.L.J.O., R.P., P.E.S., C.D.A.S., B.H.S., H.E.D.S., L.M.t’H, G.M.T., A.M.J.M.v.d.M., A.A.W.A.v.d.H., G.J.H.v.d.K., C.M.v.D., M.M.J.v.G., D.v.H., G.W., D.V., N.A., D.R., R.K., S.D.M., A.G.U. and A.Z. contributed to cohort data collection. M. Beekman, M. Bot, L.C., I.d.B., J.M.G., L.L., R.L.-G., J.L., M.G.N., R.C.S., L.M.H., C.S.T., E.B.v.d.A., D.V., D.R., L.J.M.A. and E.W. contributed to data analysis. J.B.v.K. contributed to web development. A.D., J.M.G., L.L., J.L. and C.M.v.D. contributed to writing of the manuscript. I.C.W.A., M. Beekman, J.W.J.B., D.I.B., M. Bot, L.C., I.d.B., A.D., P.J.M.E., J.F., J.M.G., L.L., R.L.-G., J.L., Y.M., D.O.M.K., M.N., G.L.J.O., B.P., R.P., F.R., R.C.S., B.H.S., L.M.t’H, G.M.T., C.T., E.B.v.d.A., A.M.J.M.v.d.M., A.A.W.A.v.d.H., G.J.H.v.d.K., K.W.v.D., C.M.v.D., M.M.J.v.G., G.W., D.V., N.A., R.K., L.J.M.A., S.D.M. and A.Z. contributed to critical review of manuscript.

Corresponding authors

Correspondence to Jun Liu or Cornelia M. van Duijn.

Ethics declarations

Competing interests

D.O.M.-K. is a part-time clinical research consultant for Metabolon, Inc. All other authors have nothing to disclose. The funders had no role in the study design, data collection and analysis, decision to publish or preparation of the manuscript.

Additional information

Peer review information Jennifer Sargent was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Correlation between metabolites in Rotterdam Study.

The correlation matrix of metabolites were performed by Pearson’s correlation (n = 5,191). The hierarchical cluster analysis was used in the clustering. Color in the boxes, correlation coefficient.

Extended Data Fig. 2 Drug–metabolite associations in model 1 versus model 2.

The drugs with at least one significant metabolite association in baseline model (model 1) by linear regression are shown. The first letter of the ATC code precedes the drug name, to identify different categories. Sample sizes of the drug users and non-users in model 1 (age and sex adjusted) and model 2 (age, sex, BMI and smoking adjusted) are shown following drug names, respectively. Dark red, positive significant associations in model 1 (P-value < 1.9 × 10−5); light red, positive nonsignificant associations in model 1 (P-value ≥ 1.9 × 10−5); dark blue, negatively significant associations in model 1 (P-value < 1.9 × 10−5); light blue, negatively nonsignificant associations in model 1 (P-value ≥ 1.9 × 10−5). Asterisks in boxes denote that neither direction nor significance status were different between model 1 and model 2 (P-value < 1.9 × 10−5). Two-tailed tests were used.

Extended Data Fig. 3 Drug–metabolite associations in model 2 versus model 3.

The drugs with at least one significant metabolite association in model 2 (age, sex, BMI and smoking adjusted) by linear regression are shown. The first letter of the ATC code is shown preceding the drug name, to identify different categories. Sample sizes of the drug users and non-users in model 2 and model 3 (age, sex, BMI, smoking and co-treatment adjusted) are shown following drug names, respectively. Dark red, positive significant associations in model 2 (P-value < 1.9 × 10−5); light red, positive nonsignificant associations in model 2 (P-value ≥ 1.9 × 10−5) dark blue, negatively significant associations in in model 2 (P-value < 1.9 × 10−5); light blue, negatively nonsignificant associations in in model 2 (P-value ≥ 1.9 × 10−5). Asterisks in boxes denote that neither direction nor significance status was different between model 2 and model 3 (P-value threshold is multiple testing-corrected per drug; See Supplementary Table 4). Two-tailed tests were used.

Extended Data Fig. 4 Correlation between drugs.

The correlation matrix of metabolites were performed by Spearman’s correlation (n = 6,631). The first letter of the ATC code is shown preceding the drug name, to identify different categories. Sample size of the drug users and non-users is shown following drug names. The depth of the color refers to the correlation coefficients. Asterisks in boxes denote the positively significant correlations (P-value < 5.9 × 10−4). Two-tailed tests were used.

Extended Data Fig. 5 Drug–metabolite Associations in model 3 versus single drug test.

The first letter of the ATC code is shown preceding the drug name, to identify different categories. Single drug test: Association analysis (linear regression) in the sub-samples of patients who use one drug only (one-drug-users) and all-treatment-naive controls. Sample size of the drug users and non-users in model 3 (age, sex, BMI, smoking and co-treatment adjusted) and the single drug test are shown following drug names, respectively. Dark red, positive significant associations in model 3 which are available for the single drug test; light red, positive non-significant associations in model 3 or not available for the single drug test; dark blue, negatively significant associations in model 3 which are available for the single drug test; light blue, negatively non-significant associations in model 3 or not available for the single drug test. Asterisks in boxes denote that the significant associations confirmed in the single drug test (P threshold is multiple testing-corrected per drug; see Supplementary Table 4). Two-tailed tests were used.

Extended Data Fig. 6 Association of PPI/dosage and the PPI-related metabolites.

The association of dosage of PPI and metabolites were tested by linear regression in Rotterdam Study (n = 700). The PPI-related metabolites were selected in model 3. DDD, defined daily dose of PPI. (/), sample size of user/non-user. Red, positive association, blue, negative association. The depth of the color refers to the association estimates. Asterisks in boxes denote significance after correcting for multiple test (P-value < 0.004). Two-tailed tests were used.

Extended Data Fig. 7 Association of specific PPI drugs and the PPI-related metabolites.

The association of PPI drugs (A02BC) and metabolites were tested by linear regression in Rotterdam Study. The PPI-related metabolites were selected in model 3. A02BC01, omeprazole; A02BC02, pantoprazole; A02BC03, lansoprazole; A02BC04, rabeprazole; A02BC05, esomeprazole. (/), sample size of user/non-user. Red, positive association; blue: negative association. The depth of the color refers to the association estimates. Asterisks in boxes denote significance after correcting for multiple test (P-value < 0.004). Two-tailed tests were used.

Extended Data Fig. 8 The effect of population structure on metabolite clustering across datasets.

Principal component (PC) analysis was performed using joint metabolite data from the cohorts (AlphaOmega, n = 877; ERF, n = 778; RS1, RS Dataset 1, n = 2,975; RS2, RS Dataset 2, n = 729; RS3, RS Dataset 3, n = 1,487; TMS, n = 854). Two-tailed tests were used.

Supplementary information

Supplementary Information

Supplementary Fig. 1 and information.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–15.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, J., Lahousse, L., Nivard, M.G. et al. Integration of epidemiologic, pharmacologic, genetic and gut microbiome data in a drug–metabolite atlas. Nat Med 26, 110–117 (2020). https://doi.org/10.1038/s41591-019-0722-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0722-x

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research