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.

  • Resource
  • Published:

Integration of multi-omics data and deep phenotyping enables prediction of cytokine responses

This article has been updated

Abstract

The immune response to pathogens varies substantially among people. Whereas both genetic and nongenetic factors contribute to interperson variation, their relative contributions and potential predictive power have remained largely unknown. By systematically correlating host factors in 534 healthy volunteers, including baseline immunological parameters and molecular profiles (genome, metabolome and gut microbiome), with cytokine production after stimulation with 20 pathogens, we identified distinct patterns of co-regulation. Among the 91 different cytokine–stimulus pairs, 11 categories of host factors together explained up to 67% of interindividual variation in cytokine production induced by stimulation. A computational model based on genetic data predicted the genetic component of stimulus-induced cytokine production (correlation 0.28–0.89), and nongenetic factors influenced cytokine production as well.

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: Analysis of baseline immunological parameters and molecular profiling shows that baseline parameters are intercorrelated.
Fig. 2: Contribution of baseline immunological parameters and multi-omics to cytokine variation.
Fig. 3: Examples of baseline molecules differentially associated with cytokine responses.
Fig. 4: Cumulative contribution of multiple baseline traits to the variation in stimulated cytokine production.
Fig. 5: Integrating gene expression profiles and cytokine production in response to C. albicans.
Fig. 6: Stimulated cytokine production correlates with genetic risk score for autoimmune diseases.
Fig. 7: Cytokine production in response to pathogens can be predicted on the basis of genetics and baseline immune profiles.
Fig. 8: Prediction using the genetic model in an independent dataset shows that some cytokine–stimulus pairs can be predicted successfully.

Similar content being viewed by others

Change history

  • 18 June 2018

    In the version of this article initially published, it was categorized as an Article. The correct category is Resource. The error has been corrected in the HTML and PDF version of the article.

References

  1. Fairfax, B. P. et al. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Kumar, V., Wijmenga, C. & Xavier, R. J. Genetics of immune-mediated disorders: from genome-wide association to molecular mechanism. Curr. Opin. Immunol. 31, 51–57 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Lee, M. N. et al. Common genetic variants modulate pathogen-sensing responses in human dendritic cells. Science 343, 1246980 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Netea, M. G., Wijmenga, C. & O’Neill, L. A. J. Genetic variation in Toll-like receptors and disease susceptibility. Nat. Immunol. 13, 535–542 (2012).

    Article  PubMed  CAS  Google Scholar 

  5. Ye, C. J. et al. Intersection of population variation and autoimmunity genetics in human T cell activation. Science 345, 1254665 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Brodin, P. & Davis, M. M. Human immune system variation. Nat. Rev. Immunol. 17, 21–29 (2017).

    Article  PubMed  CAS  Google Scholar 

  7. Li, Y. et al. A functional genomics approach to understand variation in cytokine production in humans. Cell 167, 1099–1110.e14 (2016).

    Article  PubMed  CAS  Google Scholar 

  8. Schirmer, M. et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell 167, 1125–1136.e8 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Ter Horst, R. et al. Host and environmental factors influencing individual human cytokine responses. Cell 167, 1111–1124.e13 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Brannian, J. D., Zhao, Y. & McElroy, M. Leptin inhibits gonadotrophin-stimulated granulosa cell progesterone production by antagonizing insulin action. Hum. Reprod. 14, 1445–1448 (1999).

    Article  PubMed  CAS  Google Scholar 

  11. Härle, P. et al. Possible role of leptin in hypoandrogenicity in patients with systemic lupus erythematosus and rheumatoid arthritis. Ann. Rheum. Dis. 63, 809–816 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Blum, W. F. et al. Plasma leptin levels in healthy children and adolescents: dependence on body mass index, body fat mass, gender, pubertal stage, and testosterone. J. Clin. Endocrinol. Metab. 82, 2904–2910 (1997).

    PubMed  CAS  Google Scholar 

  13. Behre, H. M., Simoni, M. & Nieschlag, E. Strong association between serum levels of leptin and testosterone in men. Clin. Endocrinol. (Oxf.) 47, 237–240 (1997).

    Article  CAS  Google Scholar 

  14. Xu, T. et al. P-selectin cross-links PSGL-1 and enhances neutrophil adhesion to fibrinogen and ICAM-1 in a Src kinase-dependent, but GPCR-independent mechanism. Cell Adhes. Migr. 1, 115–123 (2007).

    Article  Google Scholar 

  15. Gawaz, M., Langer, H. & May, A. E. Platelets in inflammation and atherogenesis. J. Clin. Invest. 115, 3378–3384 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Furman, D. et al. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol. Syst. Biol. 9, 659 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Furman, D. et al. Cytomegalovirus infection enhances the immune response to influenza. Sci. Transl. Med. 7, 281ra43 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Furman, D. et al. Systems analysis of sex differences reveals an immunosuppressive role for testosterone in the response to influenza vaccination. Proc. Natl. Acad. Sci. USA 111, 869–874 (2014).

    Article  PubMed  CAS  Google Scholar 

  19. Davis, M. M., Tato, C. M. & Furman, D. Systems immunology: just getting started. Nat. Immunol. 18, 725–732 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Ashburner, M. et al. Gene ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Shah, S. et al. Improving phenotypic prediction by combining genetic and epigenetic associations. Am. J. Hum. Genet. 97, 75–85 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Novick, D. et al. Interleukin-18 binding protein: a novel modulator of the Th1 cytokine response. Immunity 10, 127–136 (1999).

    Article  PubMed  CAS  Google Scholar 

  23. Okamura, H. et al. Cloning of a new cytokine that induces IFN-γ production by T cells. Nature 378, 88–91 (1995).

    Article  PubMed  CAS  Google Scholar 

  24. Vinolo, M. A. R., Rodrigues, H. G., Nachbar, R. T. & Curi, R. Regulation of inflammation by short chain fatty acids. Nutrients 3, 858–876 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Tedelind, S., Westberg, F., Kjerrulf, M. & Vidal, A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J. Gastroenterol. 13, 2826–2832 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Cavaglieri, C. R. et al. Differential effects of short-chain fatty acids on proliferation and production of pro- and anti-inflammatory cytokines by cultured lymphocytes. Life Sci. 73, 1683–1690 (2003).

    Article  PubMed  CAS  Google Scholar 

  27. Coëffier, M., Marion, R., Ducrotté, P. & Déchelotte, P. Modulating effect of glutamine on IL-1β-induced cytokine production by human gut. Clin. Nutr. 22, 407–413 (2003).

    Article  PubMed  CAS  Google Scholar 

  28. Lecleire, S. et al. Combined glutamine and arginine decrease proinflammatory cytokine production by biopsies from Crohn’s patients in association with changes in nuclear factor-kappaB and p38 mitogen-activated protein kinase pathways. J. Nutr. 138, 2481–2486 (2008).

    Article  PubMed  CAS  Google Scholar 

  29. Genome of the Netherlands Consortium. Whole-genome sequence variation, population structure and demographic history of the Dutch population. Nat. Genet. 46, 818–825 (2014).

    Article  CAS  Google Scholar 

  30. Lvovs, D., Favorova, O. O. & Favorov, A. V. A Polygenic approach to the study of polygenic diseases. Acta Naturae 4, 59–71 (2012).

    PubMed  PubMed Central  CAS  Google Scholar 

  31. Li, Y. et al. Inter-individual variability and genetic influences on cytokine responses to bacteria and fungi. Nat. Med. 22, 952–960 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Brodin, P. et al. Variation in the human immune system is largely driven by non-heritable influences. Cell 160, 37–47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Barreiro, L. B. & Quintana-Murci, L. From evolutionary genetics to human immunology: how selection shapes host defence genes. Nat. Rev. Genet. 11, 17–30 (2010).

    Article  PubMed  CAS  Google Scholar 

  34. Casals, F. et al. Genetic adaptation of the antibacterial human innate immunity network. BMC Evol. Biol. 11, 202 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Andrés, A. M. et al. Targets of balancing selection in the human genome. Mol. Biol. Evol. 26, 2755–2764 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Pickrell, J. K. et al. Signals of recent positive selection in a worldwide sample of human populations. Genome Res. 19, 826–837 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Tang, K., Thornton, K. R. & Stoneking, M. A new approach for using genome scans to detect recent positive selection in the human genome. PLoS Biol. 5, e171 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Voight, B. F., Kudaravalli, S., Wen, X. & Pritchard, J. K. A map of recent positive selection in the human genome. PLoS Biol. 4, e72 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wang, E. T., Kodama, G., Baldi, P. & Moyzis, R. K. Global landscape of recent inferred Darwinian selection for Homo sapiens. Proc. Natl. Acad. Sci. USA 103, 135–140 (2006).

    Article  PubMed  CAS  Google Scholar 

  40. Williamson, S. H. et al. Localizing recent adaptive evolution in the human genome. PLoS Genet. 3, e90 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Wang, F., Beck-García, K., Zorzin, C., Schamel, W. W. A. & Davis, M. M. Inhibition of T cell receptor signaling by cholesterol sulfate, a naturally occurring derivative of membrane cholesterol. Nat. Immunol. 17, 844–850 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Aguirre-Gamboa, R. et al. Differential effects of environmental and genetic factors on t and b cell immune traits. Cell Rep. 17, 2474–2487 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  PubMed  CAS  Google Scholar 

  44. Anders, S., Pyl, P. T. & Huber, W. HTSeq: a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  PubMed  CAS  Google Scholar 

  45. Zou, H. & Hastie, T. Regularization and variable selection via the elastic net. J. R. Stat. Soc. Series B Stat. Methodol. 67, 301–320 (2005).

    Article  Google Scholar 

  46. Endelman, J. B. Ridge regression and other kernels for genomic selection with r package rrBLUP. Plant Genome 4, 250–255 (2011).

    Article  Google Scholar 

  47. Riedelsheimer, C. et al. Genomic and metabolic prediction of complex heterotic traits in hybrid maize. Nat. Genet. 44, 217–220 (2012).

    Article  PubMed  CAS  Google Scholar 

  48. Morris, A. P. et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat. Genet. 44, 981–990 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Paternoster, L. et al. Multi-ancestry genome-wide association study of 21,000 cases and 95,000 controls identifies new risk loci for atopic dermatitis. Nat. Genet. 47, 1449–1456 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Putov, N. V., Bulatov, P. K., Gorovenko, G. G., Fedoseev, G. B. & Brusilovskiĭ, B. M. Classification of unspecific diseases of the bronchopulmonary system. Vrach. Delo 52–56 (1977).

  51. Köttgen, A. et al. Genome-wide association analyses identify 18 new loci associated with serum urate concentrations. Nat. Genet. 45, 145–154 (2013).

    Article  PubMed  CAS  Google Scholar 

  52. Liu, J. Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Trynka, G. et al. Dense genotyping identifies and localizes multiple common and rare variant association signals in celiac disease. Nat. Genet. 43, 1193–1201 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Hinks, A. et al. Dense genotyping of immune-related disease regions identifies 14 new susceptibility loci for juvenile idiopathic arthritis. Nat. Genet. 45, 664–669 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. International Multiple Sclerosis Genetics Consortium. et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 476, 214–219 (2011).

    Article  CAS  Google Scholar 

  56. Cordell, H. J. et al. International genome-wide meta-analysis identifies new primary biliary cirrhosis risk loci and targetable pathogenic pathways. Nat. Commun. 6, 8019 (2015).

    Article  PubMed  CAS  Google Scholar 

  57. Tsoi, L. C. et al. Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat. Genet. 44, 1341–1348 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Okada, Y. et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376–381 (2014).

    Article  PubMed  CAS  Google Scholar 

  59. Bentham, J. et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat. Genet. 47, 1457–1464 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Onengut-Gumuscu, S. et al. Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat. Genet. 47, 381–386 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank all of the volunteers in the 500FG and GoNL cohorts for their participation. We thank T. A. Wassenaar and L. Steenhuis of the Hanze University of Applied Science for providing input on the project and helping with the analyses. We thank K. Mc Intyre for editing the text. We thank the EAGLE eczema consortium for making their summary statistics publicly available. The HFGP is supported by a European Research Council (ERC) Consolidator grant (ERC 310372). This study was further supported by an IN-CONTROL CVON grant (CVON2012-03) and a Netherlands Organization for Scientific Research (NWO) Spinoza prize (NWO SPI 94-212) to M.G.N.; an ERC advanced grant (FP/2007-2013/ERC grant 2012-322698) and an NWO Spinoza prize (NWO SPI 92-266) to C.W.; a European Union Seventh Framework Programme grant (EU FP7) TANDEM project (HEALTH-F3-2012-305279) to C.W. and V.K.; and an NWO VENI grant (NWO 863.13.011) and an ZonMw-OffRoad-91215206 grant to Y.L. M.O. was supported by an NWO VENI grant (016.176.006). R.J.X. was supported by National Institutes of Health (NIH) grants DK43351, AT009708 and AI137325.

Author information

Authors and Affiliations

Authors

Contributions

Y.L., C.W. and M.G.N. designed the study. M.O., S.P.S., M.J., R.T.N.-M., H.J.P.M.K., I.J., R.J.X. and L.A.B.J. performed the experiments and processed the data. U.V. collected and preprocessed public summary statistics. O.B.B. performed statistical analysis with assistance from R.A.-G., S.S., U.V. and L.F.; O.B.B., M.Z., Y.L., S.W., V.K., M.G.N., and C.W. interpreted the data. Y.L., C.W., M.G.N. and O.B.B. wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Cisca Wijmenga, Mihai G. Netea or Yang Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated Supplementary Information

Supplementary Figure 1 Overview of variation in data in the 500FG cohort.

(a) Distributions of age in 500FG shows that there is a large proportion of young people. (b) Distribution in BMI in 500FG shows most people are within a healthy BMI range. (c) Sample variance after age, gender and season correction of gene expression quantified by RNAseq (n=89). The top 75 most varying genes are shown (d) Sample variance of cell counts after correction for age, gender and season effects shows effector T-cell populations are among the most variable between individuals (n=472). (e) Sample variance of baseline molecular components and immune parameters (n=489).

Supplementary Figure 2 Validation and replication experiments.

(a) HDL cholesterol decreases cytokine production in response to A. fumigatus. Log2 cytokine expression relative to RPMI in PBMCs cultured using LPDP and LPDP+HDL shows a decrease in expression upon stimulation with A. fumigatus. Experiments were performed in 6 healthy individuals. (b) Spearman correlations after age and gender correction between cytokine production and circulating IL-18BP concentration in 500FG (left, n=489) and a subset of individuals from 300-OB filtered on BMI < 28 (right, n=51). (c) Cytokine production of TNF-α and IL-6 by PBMC derived macrophages in response to E. coli and S. aureus is lowered in cells cultured on a medium containing acetate but not for C. albicans. Y-axis shows log2 fold changes in cytokine level compared to the non-acetate condition in macrophages cultured on a medium with acetate and stimulated with C. albicans, S. Aureus and E. coli in 6 healthy Dutch donors. Blue indicates TNFA. Red indicates IL-6.

Supplementary Figure 3 Baseline molecular components are differentially associated with cytokine responses to pathogens.

Spearman correlations between stimulated cytokine production (after correction for age- and gender-effects) and the top 5 (when taking effect size into account) most-selected metabolites (total cholesterol level in HDL3, glutamine, free cholesterol and α-1 acid glycoprotein) and mediators (α-1 antitrepsin, IL-18BP, adiponecting, resistin and leptin) show differential effects between monocyte- and lymphocyte-derived cytokine measurements as well as across multiple stimulations. Sample size was n = 377 for the metabolites and n = 489 for the modulators, with each sample representing an individual.

Supplementary Figure 4 Estimation of the unique contributions of multiple baseline immunological parameters to the variation in stimulated cytokine production.

Adjusted R2 between a full and multiple-reduced multivariate linear models (MVLM) giving an estimation unique of the effect. Each colored bar represents how much additional variation (on top of the preceding colors) the MVLM for that category explains when conditioned on the other 9 MVLMs. The combined dataset consisted of 266 samples. Each sample represents an individual. Gene expression was not included in this analysis because of the relatively small sample size of the RNA-seq experiment after overlapping with the other datasets (n = 69). X-axis denotes adjusted R2 values. Y-axis denotes different cytokine-stimulation pairs.

Supplementary Figure 5 Correlations between stimulated cytokine production and polygenic risk scores for immune disease.

(a) Spearman correlations between PRS and stimulated cytokine production remain largely stable across multiple GWAS thresholds. Spearman correlations between PRS and cytokine measurements (n = 430) show similar direction and effect size across the multiple GWAS thresholds (indicated by the color of the boxplot) used for calculating PRS for most immune diseases tested. (b) Distribution of permuted mean correlations between disease risk in monocyte derived cytokines (left panel) and lymphocyte derived cytokines (right panel) shows the measured mean estimate (red arrow) differs from the permuted distribution for all diseases showing significant changes.

Supplementary Figure 6 Correlations between predicted and measured stimulated cytokine level for multiple tissue types.

(a) Correlations between predicted and measured cytokine level for each of the 10 cross validation folds. Predicted using multivariate linear models (MVLM) trained by Elastic Net (n = 392). (b) Correlations between predicted and measured cytokine level for each of the 10 cross validation folds. Predicted using multivariate linear models MVLMs trained by RR-blup (n = 392). (C) Correlations between predicted and measured cytokine level for each of the 10 cross validation folds. Predicted using MVLMs trained by PLS (n = 392). (e) Correlations of predicted and measured cytokine level of MVLMs trained with all categories and gene expression shows model large variation (n=69).

Supplementary information

Supplementary Figures and Supplementary Tables

Supplementary Figures 1–6 and Supplementary Tables 1–7

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bakker, O.B., Aguirre-Gamboa, R., Sanna, S. et al. Integration of multi-omics data and deep phenotyping enables prediction of cytokine responses. Nat Immunol 19, 776–786 (2018). https://doi.org/10.1038/s41590-018-0121-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0121-3

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