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.

Human postprandial responses to food and potential for precision nutrition

A Publisher Correction to this article was published on 20 October 2020

This article has been updated

Abstract

Metabolic responses to food influence risk of cardiometabolic disease, but large-scale high-resolution studies are lacking. We recruited n = 1,002 twins and unrelated healthy adults in the United Kingdom to the PREDICT 1 study and assessed postprandial metabolic responses in a clinical setting and at home. We observed large inter-individual variability (as measured by the population coefficient of variation (s.d./mean, %)) in postprandial responses of blood triglyceride (103%), glucose (68%) and insulin (59%) following identical meals. Person-specific factors, such as gut microbiome, had a greater influence (7.1% of variance) than did meal macronutrients (3.6%) for postprandial lipemia, but not for postprandial glycemia (6.0% and 15.4%, respectively); genetic variants had a modest impact on predictions (9.5% for glucose, 0.8% for triglyceride, 0.2% for C-peptide). Findings were independently validated in a US cohort (n = 100 people). We developed a machine-learning model that predicted both triglyceride (r = 0.47) and glycemic (r = 0.77) responses to food intake. These findings may be informative for developing personalized diet strategies. The ClinicalTrials.gov registration identifier is NCT03479866.

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: Experimental design.
Fig. 2: Variation in postprandial responses.
Fig. 3: Relationship of baseline values, genetic and microbiome factors with postprandial responses.
Fig. 4: Machine-learning models fitted to postprandial measures.
Fig. 5: Associations between fasting and postprandial values for triglyceride, C-peptide and glucose concentrations with clinical measures in the UK cohort.
Fig. 6: Person-specific diversity in postprandial response.

Similar content being viewed by others

Data availability

The data used for analysis in this study are held by the department of Twin Research at Kings College London. The data can be released to bona fide researchers using our normal procedures overseen by the Wellcome Trust and its guidelines as part of our core funding. We receive around 100 requests per year for our datasets and have a meeting 3 times per month with independent members to assess proposals. The application is at https://twinsuk.ac.uk/resources-for-researchers/access-our-data/. This means that the data need to be anonymized and conform to GDPR standards. Specifically for this paper, all the variables used in the models can be requested as well as the summary outcome measures for each person. The 16S microbiome data will be uploaded onto the EBI website (https://www.ebi.ac.uk/) with unlimited access. Source data for Figs. 26 and Extended Data Figs. 24 are presented with the paper

Code availability

The scripts for statistical analysis are freely available upon request to the Department of Twins Research and Genetic Epidemiology at King’s College London. Application is via https://twinsuk.ac.uk/resources-for-researchers/access-our-data/. The scripts used for machine-learning analyses can be found in the Supplementary Software.

Change history

References

  1. GBDD 2017 Diet Collaborators. Health effects of dietary risks in 195 countries, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 393, 1958–1972 (2019).

    Article  Google Scholar 

  2. US Department of Health and Human Services & US Department of Agriculture. 2015–2020 Dietary Guidelines for Americans 8th edn (2015).

  3. Karpyn, A. Food and Public Health: A Practical Introduction (Oxford University Press, 2018).

  4. European Food Safety Authority. Dietary reference values for nutrients summary report. EFSA Supporting Publications 14, e15121E (2017).

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

    Article  CAS  PubMed  Google Scholar 

  6. Tebani, A. & Bekri, S. Paving the way to precision nutrition through metabolomics. Front. Nutr. 6, 41 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Kolovou, G. D. et al. Assessment and clinical relevance of non-fasting and postprandial triglycerides: an expert panel statement. Curr. Vasc. Pharmacol. 9, 258–270 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Astley, C. M. et al. Genetic evidence that carbohydrate-stimulated insulin secretion leads to obesity. Clin. Chem. 64, 192–200 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Blaak, E. E. et al. Impact of postprandial glycaemia on health and prevention of disease. Obes. Rev. 13, 923–984 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. DECODE Study Group, European Diabetes Epidemiology Group. Glucose tolerance and cardiovascular mortality: comparison of fasting and 2-hour diagnostic criteria. Arch. Intern. Med. 161, 397–405 (2001).

  11. Ning, F. et al. Cardiovascular disease mortality in Europeans in relation to fasting and 2-h plasma glucose levels within a normoglycemic range. Diabetes Care 33, 2211–2216 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Bansal, S. et al. Fasting compared with nonfasting triglycerides and risk of cardiovascular events in women. JAMA 298, 309–316 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Lindman, A. S., Veierod, M. B., Tverdal, A., Pedersen, J. I. & Selmer, R. Nonfasting triglycerides and risk of cardiovascular death in men and women from the Norwegian Counties Study. Eur. J. Epidemiol. 25, 789–798 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jackson, K. G., Poppitt, S. D. & Minihane, A. M. Postprandial lipemia and cardiovascular disease risk: Interrelationships between dietary, physiological and genetic determinants. Atherosclerosis 220, 22–33 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. Stumvoll, M., Goldstein, B. J. & van Haeften, T. W. Type 2 diabetes: principles of pathogenesis and therapy. Lancet 365, 1333–1346 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Moayyeri, A., Hammond, C. J., Hart, D. J. & Spector, T. D. The UK Adult Twin Registry (TwinsUK Resource). Twin Res. Hum. Genet. 16, 144–149 (2013).

    Article  PubMed  Google Scholar 

  17. Prokopenko, I. et al. A central role for GRB10 in regulation of islet function in man. PLoS Genet. 10, e1004235 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Saxena, R. et al. Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat. Genet. 42, 142–148 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Wojczynski, M. K. et al. Genome-wide association study of triglyceride response to a high-fat meal among participants of the NHLBI Genetics of Lipid Lowering Drugs and Diet Network (GOLDN). Metabolism 64, 1359–1371 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. Almgren, P. et al. Genetic determinants of circulating GIP and GLP-1 concentrations. JCI Insight 2, e93306 (2017).

  22. Wojcikowski, M., Ballester, P. J. & Siedlecki, P. Performance of machine-learning scoring functions in structure-based virtual screening. Sci. Rep. 7, 46710 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Hastie, T., Tibshirani, R. & Friedman, J. The Elements of Statistical Learning: Data Mining, Inference, and Prediction (Springer New York, 2013).

  24. Russell, W. R. et al. Impact of diet composition on blood glucose regulation. Crit. Rev. Food Sci. Nutr. 56, 541–590 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Jeong, Y. et al. A review of recent evidence from meal-based diet interventions and clinical biomarkers for improvement of glucose regulation. Prev. Nutr. Food Sci. 25, 9–24 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Delahanty, L. M. et al. Effects of weight loss, weight cycling, and weight loss maintenance on diabetes incidence and change in cardiometabolic traits in the Diabetes Prevention Program. Diabetes Care 37, 2738–2745 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Diabetes Prevention Program Research Group. et al. 10-year follow-up of diabetes incidence and weight loss in the diabetes prevention program outcomes study. Lancet 374, 1677–1686 (2009).

    Article  PubMed Central  Google Scholar 

  28. Knowler, W. C. et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N. Engl. J. Med. 346, 393–403 (2002).

    Article  CAS  PubMed  Google Scholar 

  29. Papandonatos, G. D. et al. Genetic predisposition to weight loss and regain with lifestyle intervention: analyses from the diabetes prevention program and the look AHEAD randomized controlled trials. Diabetes 64, 4312–4321 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Taylor, R. Banting Memorial Lecture 2012: reversing the twin cycles of type 2 diabetes. Diabet. Med. 30, 267–275 (2013).

  31. Bell, K. J. et al. Impact of fat, protein, and glycemic index on postprandial glucose control in type 1 diabetes: implications for intensive diabetes management in the continuous glucose monitoring era. Diabetes Care 38, 1008–1015 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Hansson, P. et al. Meals with similar fat content from different dairy products induce different postprandial triglyceride responses in healthy adults: a randomized controlled cross-over trial. J. Nutr. 149, 422–431 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Atkinson, F. S., Hancock, D., Petocz, P. & Brand-Miller, J. C. The physiologic and phenotypic significance of variation in human amylase gene copy number. Am. J. Clin. Nutr. 108, 737–748 (2018).

    Article  PubMed  Google Scholar 

  34. Gardner, C. D. et al. Effect of low-fat vs low-carbohydrate diet on 12-month weight loss in overweight adults and the association with genotype pattern or insulin secretion: The DIETFITS randomized clinical trial. JAMA 319, 667–679 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Viitasalo, A. et al. Genetic predisposition to higher body fat yet lower cardiometabolic risk in children and adolescents. Int. J. Obes. (Lond.) 43, 2007–2016 (2019).

    Article  CAS  Google Scholar 

  36. Hughes, R. L., Kable, M. E., Marco, M. & Keim, N. L. The role of the gut microbiome in predicting response to diet and the development of precision nutrition models. Part II: results. Adv. Nutr. 10, 979–998 (2019).

  37. Lee, J. G. et al. Deep learning in medical imaging: general overview. Korean J. Radiol. 18, 570–584 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Dubois, C. et al. Effects of graded amounts (0–50 g) of dietary fat on postprandial lipemia and lipoproteins in normolipidemic adults. Am. J. Clin. Nutr. 67, 31–38 (1998).

    Article  CAS  PubMed  Google Scholar 

  39. Garcia-Perez, I. et al. Objective assessment of dietary patterns by use of metabolic phenotyping: a randomised, controlled, crossover trial. Lancet Diabetes Endocrinol. 5, 184–195 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Zierer, J. et al. The fecal metabolome as a functional readout of the gut microbiome. Nat. Genet. 50, 790–795 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Berry, S. D. D. et al. Personalised REsponses to DIetary Composition Trial (PREDICT): an intervention study to determine inter-individual differences in postprandial response to foods. Protoc. Exch. https://doi.org/10.21203/rs.2.20798/v1 (2020).

  42. Verdi, S. et al. TwinsUK: the UK adult twin registry update. Twin Res. Hum. Genet. 22, 523–529 (2019).

  43. Mulligan, A. A. et al. A new tool for converting food frequency questionnaire data into nutrient and food group values: FETA research methods and availability. BMJ Open. 4, e004503 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Flint, A., Raben, A., Blundell, J. E. & Astrup, A. Reproducibility, power and validity of visual analogue scales in assessment of appetite sensations in single test meal studies. Int. J. Obes. Relat. Metab Disord. 24, 38–48 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Callahan, B. J. et al. DADA2: High-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gloor, G. B., Wu, J. R., Pawlowsky-Glahn, V. & Egozcue, J. J. It’s all relative: analyzing microbiome data as compositions. Ann Epidemiol. 26, 322–329 (2016).

    Article  PubMed  Google Scholar 

  47. Thomas, A. M. et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat. Med. 25, 667–678 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. 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 

  49. Johansson, J., Becker, C., Persson, N. G., Fex, M. & Torn, C. C-peptide in dried blood spots. Scand. J. Clin. Lab. Invest. 70, 404–409 (2010).

    Article  CAS  PubMed  Google Scholar 

  50. Samson, C. E., Galia, A. L., Llave, K. I., Zacarias, M. B. & Mercado-Asis, L. B. Postprandial peaking and plateauing of triglycerides and VLDL in patients with underlying cardiovascular diseases despite treatment. Int. J. Endocrinol. Metab. 10, 587–593 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Madhu, S., Sinha, B., Aslam, M., Mehrotra, G. & Dwivedi, S. Postprandial triglyceride responses and endothelial function in prediabetic first-degree relatives of patients with diabetes. J .Clin. Lipidol. 11, 1415–1420 (2017).

    Article  PubMed  Google Scholar 

  52. Nakamura, A. et al. Different postprandial lipid metabolism and insulin resistance between non-diabetic patients with and without coronary artery disease. J. Cardiol. 66, 435–444 (2015).

    Article  PubMed  Google Scholar 

  53. van Hees, V. T. et al. Estimating sleep parameters using an accelerometer without sleep diary. Sci. Rep. 8, 12975 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Eitel, I. et al. Genome-wide association study in takotsubo syndrome—preliminary results and future directions. Int. J. Cardiol. 236, 335–339 (2017).

    Article  PubMed  Google Scholar 

  55. Slinker, B. K. & Glantz, S. A. Multiple regression for physiological data analysis: the problem of multicollinearity. Am. J. Physiol. 249, R1–R12 (1985).

    Article  CAS  PubMed  Google Scholar 

  56. Purcell, S. Variance components models for gene-environment interaction in twin analysis. Twin Res. 5, 554–571 (2002).

    Article  PubMed  Google Scholar 

  57. Gelman, A. & Hill, J. Data Analysis Using Regression and Multilevel/Hierarchical Models (Cambridge University Press, 2007).

  58. Salvatier, J., Wiecki, T. V. & Fonnesbeck, C. Probabilistic programming in Python using PyMC3. PeerJ Comput. Sci. 2, e55 (2016).

  59. Goff, D. C. Jr. et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 129, S49–S73 (2014).

    Article  PubMed  Google Scholar 

  60. American Diabetes Association Classification and Diagnosis of Diabetes. Diabetes Care 39, S13–S22 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We express our sincere thanks to the participants of the PREDICT 1 study. We thank M. McCarthy (University of Oxford) and L. Hodson (University of Oxford) for valuable feedback on the manuscript. We thank the staff of Z. Global, the Department of Twin Research and Massachusetts General Hospital, for their tireless work in contributing to the running of the study and data collection. We thank Abbott for their support with their CGMs. This work was supported by Zoe Global and also received support from grants from the Wellcome Trust (212904/Z/18/Z) and the Medical Research Council (MRC)/British Heart Foundation Ancestry and Biological Informative Markers for Stratification of Hypertension (AIMHY; MR/M016560/1). S.E.B. was supported in part by a grant funded by the BBSRC (BB/NO12739/1). P.W.F. was supported in part by grants from the European Research Council (CoG-2015_681742_NASCENT), Swedish Research Council, Novo Nordisk Foundation and the Swedish Foundation for Strategic Research (IRC award). A.M.V. was supported by the National Institute for Health Research Nottingham Biomedical Research Centre. TwinsUK is funded by the Wellcome Trust, Medical Research Council, European Union, Chronic Disease Research Foundation (CDRF), Zoe Global and the National Institute for Health Research (NIHR)-funded BioResource, Clinical Research Facility and Biomedical Research Centre based at Guy’s and St Thomas’ NHS Foundation Trust in partnership with King’s College London. J.M. was supported by a fellowship funded by the European Commission Horizon 2020 program (H2020-MSCA-IF-2015-703787) and by National Institutes of Health grant P30 DK40561. N.S. received support from the European Research Council (ERC-STG project MetaPG), the European H2020 program (ONCOBIOME-825410 project and MASTER-818368 project), and the National Cancer Institute of the National Institutes of Health (1U01CA230551).

Author information

Authors and Affiliations

Authors

Contributions

Obtained funding: J.W., G.H., T.D.S. Study design and developed concept: S.E.B., A.M.V., J.W., G.H., R.D., A.T.C., N.S., P.W., P.W.F., T.D.S. Data collection: S.E.B., D.A.D., H.A.K., M. Mangino, J.M., I.L., D.H. Data analysis: A.M.V., M. Mangino., F.A., R.D., J.C., C.B., S.G., E.B., M. Mazidi, P.W., N.S. Study coordination: S.E.B., H.A.K., D.A.D., G.H., J.W. Writing the manuscript: S.E.B., A.M.V., D.A.D., M. Mazidi, J.W., J.C., I.L., J.M.O., C.D.G., L.M.D., A.T.C., N.S., P.W.F., T.D.S. All authors reviewed and revised the final manuscript.

Corresponding authors

Correspondence to Ana M. Valdes or Tim D. Spector.

Ethics declarations

Competing interests

T.D.S., S.E.B., A.M.V., F.A., P.W.F., L.M.D. and N.S. are consultants to Zoe Global Ltd (‘Zoe’). J.W., G.H., R.D., H.A.K., J.C., C.B., S.G., E.B., P.W. and I.L. are or have been employees of Zoe. Other authors have no conflict of interest to declare.

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 Consort Diagrams for UK and US populations in the PREDICT 1 study.

Consort Diagrams for a, UK and b, US populations in the PREDICT 1 study.

Extended Data Fig. 2 Repeatability in the PREDICT 1 study.

Repeatability in the PREDICT 1 study. a, Intraclass correlations. b, Pearson’s correlation and CV of 2h-iAUCs measured with two monitors worn by the same participant (n = 377). P-value from two-sided t-test c, Mean and standard error of fasting and postprandial serum insulin and C-peptide concentrations during the clinic visit in the PREDICT 1 study, n = 1,036.

Source data

Extended Data Fig. 3 Frequency distribution of in-person ranking for 6 of meals shown in Fig. 6a.

Frequency distribution of in-person ranking for 6 of meals shown in Fig. 6a. (High fat 40 g = meal 7, High protein = meal 8, UK average = meal 2, High carb = meal 4, OGTT = meal 5, Uk average at lunch = meal 2). n = 1102 participants.

Source data

Extended Data Fig. 4 Machine learning comparisons, cross validation and repeatability.

Machine learning comparisons, cross validation and repeatability. a, Bland-Altman plots comparing predicted and measured postprandial responses in TG, glucose and C-peptide using UK and US data. Sample sizes used were (n = number of meals) triglyceride UK: n = 958 US: n = 91; C-peptide UK: n = 957 US: n = 93; Glucose UK: n = 11550 US: n = 1200. b, Leave-one-out cross-validated Pearson R scores in PREDICT UK. 5-fold cross validation for triglyceride6h-rise on n = 958 meals, for lgucoseiAUC0-2h on n = 11,550 meals, p-values shown for two-sided t-test c, comparison of models using repeat meals vs not using them.

Source data

Supplementary information

Reporting Summary

Supplementary Software

Scripts used for data analysis and machine-learning model.

Supplementary Tables 1–5

Supplementary Tables 1–5.

Source data

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Berry, S.E., Valdes, A.M., Drew, D.A. et al. Human postprandial responses to food and potential for precision nutrition. Nat Med 26, 964–973 (2020). https://doi.org/10.1038/s41591-020-0934-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0934-0

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