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.

A trans-acting locus regulates an anti-viral expression network and type 1 diabetes risk

Abstract

Combined analyses of gene networks and DNA sequence variation can provide new insights into the aetiology of common diseases that may not be apparent from genome-wide association studies alone. Recent advances in rat genomics are facilitating systems-genetics approaches1,2. Here we report the use of integrated genome-wide approaches across seven rat tissues to identify gene networks and the loci underlying their regulation. We defined an interferon regulatory factor 7 (IRF73)-driven inflammatory network (IDIN) enriched for viral response genes, which represents a molecular biomarker for macrophages and which was regulated in multiple tissues by a locus on rat chromosome 15q25. We show that Epstein–Barr virus induced gene 2 (Ebi2, also known as Gpr183), which lies at this locus and controls B lymphocyte migration4,5, is expressed in macrophages and regulates the IDIN. The human orthologous locus on chromosome 13q32 controlled the human equivalent of the IDIN, which was conserved in monocytes. IDIN genes were more likely to associate with susceptibility to type 1 diabetes (T1D)—a macrophage-associated autoimmune disease—than randomly selected immune response genes (P = 8.85 × 10−6). The human locus controlling the IDIN was associated with the risk of T1D at single nucleotide polymorphism rs9585056 (P = 7.0 × 10−10; odds ratio, 1.15), which was one of five single nucleotide polymorphisms in this region associated with EBI2 (GPR183) expression. These data implicate IRF7 network genes and their regulatory locus in the pathogenesis of T1D.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: The rat Irf7 -driven inflammatory gene network.
Figure 2: Genetic mapping of regulatory hot spots for the IDIN.
Figure 3: A gene network and locus for T1D risk.

Similar content being viewed by others

Accession codes

Primary accessions

ArrayExpress

References

  1. Hubner, N. et al. Integrated transcriptional profiling and linkage analysis for identification of genes underlying disease. Nature Genet. 37, 243–253 (2005)

    Article  CAS  Google Scholar 

  2. Petretto, E. et al. Integrated genomic approaches implicate osteoglycin (Ogn) in the regulation of left ventricular mass. Nature Genet. 40, 546–552 (2008)

    Article  CAS  Google Scholar 

  3. Honda, K. et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 434, 772–777 (2005)

    Article  ADS  CAS  Google Scholar 

  4. Pereira, J. P., Kelly, L. M., Xu, Y. & Cyster, J. G. EBI2 mediates B cell segregation between the outer and centre follicle. Nature 460, 1122–1126 (2009)

    Article  ADS  CAS  Google Scholar 

  5. Gatto, D., Paus, D., Basten, A., Mackay, C. R. & Brink, R. Guidance of B cells by the orphan G protein-coupled receptor EBI2 shapes humoral immune responses. Immunity 31, 259–269 (2009)

    Article  CAS  Google Scholar 

  6. Altshuler, D., Daly, M. J. & Lander, E. S. Genetic mapping in human disease. Science 322, 881–888 (2008)

    Article  ADS  CAS  Google Scholar 

  7. Schadt, E. E. Molecular networks as sensors and drivers of common human diseases. Nature 461, 218–223 (2009)

    Article  ADS  CAS  Google Scholar 

  8. Chen, Y. et al. Variations in DNA elucidate molecular networks that cause disease. Nature 452, 429–435 (2008)

    Article  ADS  CAS  Google Scholar 

  9. Dimas, A. S. et al. Common regulatory variation impacts gene expression in a cell type-dependent manner. Science 325, 1246–1250 (2009)

    Article  ADS  CAS  Google Scholar 

  10. Brem, R. B., Yvert, G., Clinton, R. & Kruglyak, L. Genetic dissection of transcriptional regulation in budding yeast. Science 296, 752–755 (2002)

    Article  ADS  CAS  Google Scholar 

  11. Yvert, G. et al. Trans-acting regulatory variation in Saccharomyces cerevisiae and the role of transcription factors. Nature Genet. 35, 57–64 (2003)

    Article  CAS  Google Scholar 

  12. Roider, H. G., Manke, T., O’Keeffe, S., Vingron, M. & Haas, S. A. PASTAA: identifying transcription factors associated with sets of co-regulated genes. Bioinformatics 25, 435–442 (2009)

    Article  CAS  Google Scholar 

  13. Petretto, E. et al. New insights into the genetic control of gene expression using a Bayesian multi-tissue approach. PLoS Comput. Biol. 6, e1000737 (2010)

    Article  Google Scholar 

  14. Breitling, R. et al. Genetical genomics: spotlight on QTL hotspots. PLoS Genet. 4, e1000232 (2008)

    Article  Google Scholar 

  15. Nathan, C. & Ding, A. Nonresolving inflammation. Cell 140, 871–882 (2010)

    Article  CAS  Google Scholar 

  16. Eizirik, D. L., Colli, M. L. & Ortis, F. The role of inflammation in insulitis and β-cell loss in type 1 diabetes. Nature Rev. Endocrinol. 5, 219–226 (2009)

    Article  CAS  Google Scholar 

  17. Holness, C. L. & Simmons, D. L. Molecular cloning of CD68, a human macrophage marker related to lysosomal glycoproteins. Blood 81, 1607–1613 (1993)

    CAS  PubMed  Google Scholar 

  18. Saar, K. et al. SNP and haplotype mapping for genetic analysis in the rat. Nature Genet. 40, 560–566 (2008)

    Article  CAS  Google Scholar 

  19. Atanur, S. S. et al. The genome sequence of the spontaneously hypertensive rat: analysis and functional significance. Genome Res. 20, 791–803 (2010)

    Article  CAS  Google Scholar 

  20. Zeller, T. et al. Genetics and beyond–the transcriptome of human monocytes and disease susceptibility. PLoS ONE 5, e10693 (2010)

    Article  ADS  Google Scholar 

  21. Plagnol, V., Smyth, D. J., Todd, J. A. & Clayton, D. G. Statistical independence of the colocalized association signals for type 1 diabetes and RPS26 gene expression on chromosome 12q13. Biostatistics 10, 327–334 (2009)

    Article  Google Scholar 

  22. von Herrath, M. Diabetes: a virus–gene collaboration. Nature 459, 518–519 (2009)

    Article  ADS  CAS  Google Scholar 

  23. Nejentsev, S., Walker, N., Riches, D., Egholm, M. & Todd, J. A. Rare variants of IFIH1, a gene implicated in antiviral responses, protect against type 1 diabetes. Science 324, 387–389 (2009)

    Article  ADS  CAS  Google Scholar 

  24. Smyth, D. J. et al. A genome-wide association study of nonsynonymous SNPs identifies a type 1 diabetes locus in the interferon-induced helicase (IFIH1) region. Nature Genet. 38, 617–619 (2006)

    Article  CAS  Google Scholar 

  25. Barrett, J. C. et al. Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nature Genet. 41, 703–707 (2009)

    Article  CAS  Google Scholar 

  26. Li, Q. et al. Interferon-α initiates type 1 diabetes in nonobese diabetic mice. Proc. Natl Acad. Sci. USA 105, 12439–12444 (2008)

    Article  ADS  CAS  Google Scholar 

  27. Cooper, J. D. et al. Follow-up of 1715 SNPs from the Wellcome Trust Case Control Consortium genome-wide association study in type I diabetes families. Genes Immun. 10 (suppl. 1). S85–S94 (2009)

    Article  Google Scholar 

  28. Kawai, T. et al. Interferon-α induction through Toll-like receptors involves a direct interaction of IRF7 with MyD88 and TRAF6. Nature Immunol. 5, 1061–1068 (2004)

    Article  CAS  Google Scholar 

  29. The Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007)

Download references

Acknowledgements

We acknowledge funding from the German National Genome Research Network (NGFN-Plus ‘Genetics of Heart Failure’), the Helmholtz Association Alliance on Systems Biology (MSBN), EURATools (LSHG-CT-2005-019015), European Union FP6 (LSHM-CT-2006-037593), PHC ALLIANCE 2009 (19419PH), UK National Institute for Health Research Biomedical Research Unit (Royal Brompton and Harefield NHS Trusts, University Hospitals of Leicester NHS Trusts) and Biomedical Research Centre (Imperial College NHS Trust) awards, the British Heart Foundation, grant P301/10/0290 from the Grant Agency of the Czech Republic, grant 1M6837805002 from the Ministry of Education of the Czech Republic, the Fondation Leducq, the Medical Research Council UK, Research Councils UK, the Juvenile Diabetes Research Foundation International, National Institute for Health Research (UK), National Institute of Diabetes and Digestive and Kidney Diseases (USA), and the Wellcome Trust. The research leading to these results has received funding from the European Community’s Seventh Framework Programme (FP7/2007-2013) under grant agreement no. HEALTH-F4-2010-241504 (EURATRANS). O. Burren performed T1DBase analyses.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

S.A.C., N.H. and E.P. initiated the study. M.H., E.P., N.H. and S.A.C. participated in the conception, design and coordination of the study. H.L., Y.L., R.S., Y.A.L., S.P., C.R., K.S. and R.B. performed genetic, biochemical and functional analyses in rats. E.E.G. and J.G.C. provided Ebi2GFP/+ mouse data. M.P. and T.J.A. contributed materials and discussion of the manuscript. M.H., E.P., C.W., D.J.S., D.C., A.B., S.R.L., L.B., M.R. and L.T. designed and applied the modelling methodology and statistical analyses. M.H., E.P. and H. Schulz performed eQTL analysis in the rat. L.B. designed and performed the Bayesian analysis. C.W., D.J.S. and D.C. performed association analyses in humans. M.H., O.H., H.R. and M.V. designed and performed bioinformatics analyses in rats. J.E., C.H., S.M., W.H.O., C.M.R., N.J.S., H. Schunkert, A.H.G., S.B., T.M., T.Z., S.S., A.Z., M.R., L.T. and F.C. provided the human monocyte expression data and contributed to the transcriptomic analyses in the Cardiogenics Study and Gutenberg Heart Study cohorts. M.H., E.P., N.H. and S.A.C. wrote the paper with significant contributions from C.W. and J.A.T. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to Norbert Hubner or Stuart A. Cook.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Additional information

Microarray expression data in the rat have been deposited at ArrayExpress with the following identity codes: skeletal muscle, E-TABM-458; aorta, E-MTAB-322; liver, E-MTAB-323.

A list of participants and their affiliations appears at the end of the paper.

Supplementary information

Supplementary Information

This file contains Supplementary Information and Data, additional references and a list of contributors to the Cardiogenics Transcriptomic Study. (PDF 403 kb)

Supplementary Figures

This file contains Supplementary Figures 1-9 with legends. (PDF 2461 kb)

Supplementary Tables

This file contains Supplementary Tables 1-10. (PDF 1077 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Heinig, M., Petretto, E., Wallace, C. et al. A trans-acting locus regulates an anti-viral expression network and type 1 diabetes risk. Nature 467, 460–464 (2010). https://doi.org/10.1038/nature09386

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature09386

This article is cited by

Comments

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

Search

Quick links

Nature Briefing

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

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