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.

  • Letter
  • Published:

Gain-of-function mutations in IFIH1 cause a spectrum of human disease phenotypes associated with upregulated type I interferon signaling

Abstract

The type I interferon system is integral to human antiviral immunity. However, inappropriate stimulation or defective negative regulation of this system can lead to inflammatory disease. We sought to determine the molecular basis of genetically uncharacterized cases of the type I interferonopathy Aicardi-Goutières syndrome and of other undefined neurological and immunological phenotypes also demonstrating an upregulated type I interferon response. We found that heterozygous mutations in the cytosolic double-stranded RNA receptor gene IFIH1 (also called MDA5) cause a spectrum of neuroimmunological features consistently associated with an enhanced interferon state. Cellular and biochemical assays indicate that these mutations confer gain of function such that mutant IFIH1 binds RNA more avidly, leading to increased baseline and ligand-induced interferon signaling. Our results demonstrate that aberrant sensing of nucleic acids can cause immune upregulation.

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: Schematic of the human IFIH1 gene.
Figure 2: Quantitative RT-PCR (qPCR) of a panel of six ISGs in whole blood measured in IFIH1 mutation–positive probands and mutation-negative relatives and interferon scores in mutation-positive individuals, mutation-negative relatives and controls.
Figure 3: IFIH1 mutants activate the interferon signaling pathway more efficiently than wild-type IFIH1.
Figure 4: IFIH1 mutants form filaments.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

Protein Data Bank

References

  1. Gresser, I., Tovey, M.G., Maury, C. & Chouroulinkov, I. Lethality of interferon preparations for newborn mice. Nature 258, 76–78 (1975).

    Article  CAS  Google Scholar 

  2. Gresser, I. et al. Progressive glomerulonephritis in mice treated with interferon preparations at birth. Nature 263, 420–422 (1976).

    Article  CAS  Google Scholar 

  3. Gresser, I. et al. Interferon-induced disease in mice and rats. Ann. NY Acad. Sci. 350, 12–20 (1980).

    Article  CAS  Google Scholar 

  4. Crow, Y.J. Type I interferonopathies: a novel set of inborn errors of immunity. Ann. NY Acad. Sci. 1238, 91–98 (2011).

    Article  CAS  Google Scholar 

  5. Crow, Y.J. et al. Mutations in the gene encoding the 3′-5′ DNA exonuclease TREX1 cause Aicardi-Goutières syndrome at the AGS1 locus. Nat. Genet. 38, 917–920 (2006).

    Article  CAS  Google Scholar 

  6. Crow, Y.J. et al. Mutations in genes encoding ribonuclease H2 subunits cause Aicardi-Goutières syndrome and mimic congenital viral brain infection. Nat. Genet. 38, 910–916 (2006).

    Article  CAS  Google Scholar 

  7. Rice, G.I. et al. Mutations involved in Aicardi-Goutières syndrome implicate SAMHD1 as regulator of the innate immune response. Nat. Genet. 41, 829–832 (2009).

    Article  CAS  Google Scholar 

  8. Rice, G.I. et al. Mutations in ADAR1 cause Aicardi-Goutières syndrome associated with a type I interferon signature. Nat. Genet. 44, 1243–1248 (2012).

    Article  CAS  Google Scholar 

  9. Rice, G.I. et al. Assessment of interferon-related biomarkers in Aicardi-Goutières syndrome associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR: a case-control study. Lancet Neurol. 12, 1159–1169 (2013).

    Article  CAS  Google Scholar 

  10. Briggs, T.A. et al. Tartrate-resistant acid phosphatase deficiency causes a bone dysplasia with autoimmunity and a type I interferon expression signature. Nat. Genet. 43, 127–131 (2011).

    Article  CAS  Google Scholar 

  11. Goubau, D., Deddouche, S. & Reis, E.S.C. Cytosolic sensing of viruses. Immunity 38, 855–869 (2013).

    Article  CAS  Google Scholar 

  12. Peisley, A. et al. Cooperative assembly and dynamic disassembly of MDA5 filaments for viral dsRNA recognition. Proc. Natl. Acad. Sci. USA 108, 21010–21015 (2011).

    Article  CAS  Google Scholar 

  13. Peisley, A. et al. Kinetic mechanism for viral dsRNA length discrimination by MDA5 filaments. Proc. Natl. Acad. Sci. USA 109, E3340–E3349 (2012).

    Article  CAS  Google Scholar 

  14. Wu, B. et al. Structural basis for dsRNA recognition, filament formation, and antiviral signal activation by MDA5. Cell 152, 276–289 (2013).

    Article  CAS  Google Scholar 

  15. Rice, G. et al. Heterozygous mutations in TREX1 cause familial chilblain lupus and dominant Aicardi-Goutières syndrome. Am. J. Hum. Genet. 80, 811–815 (2007).

    Article  CAS  Google Scholar 

  16. Tojo, K. et al. Dystonia, mental deterioration, and dyschromatosis symmetrica hereditaria in a family with ADAR1 mutation. Mov. Disord. 21, 1510–1513 (2006).

    Article  Google Scholar 

  17. Livingston, J.H. et al. A type I interferon signature identifies bilateral striatal necrosis due to mutations in ADAR1. J. Med. Genet. 51, 76–82 (2014).

    Article  CAS  Google Scholar 

  18. Kondo, T. et al. Dyschromatosis symmetrica hereditaria associated with neurological disorders. J. Dermatol. 35, 662–666 (2008).

    Article  Google Scholar 

  19. Bennett, L. et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J. Exp. Med. 197, 711–723 (2003).

    Article  CAS  Google Scholar 

  20. Baechler, E.C. et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc. Natl. Acad. Sci. USA 100, 2610–2615 (2003).

    Article  CAS  Google Scholar 

  21. Cen, H. et al. Association of IFIH1 rs1990760 polymorphism with susceptibility to autoimmune diseases: a meta-analysis. Autoimmunity 46, 455–462 (2013).

    Article  CAS  Google Scholar 

  22. Crampton, S.P., Deane, J.A., Feigenbaum, L. & Bolland, S. Ifih1 gene dose effect reveals MDA5-mediated chronic type I IFN gene signature, viral resistance, and accelerated autoimmunity. J. Immunol. 188, 1451–1459 (2012).

    Article  CAS  Google Scholar 

  23. Nejentsev, S. et al. Rare variants of IFIH1, a gene implicated in antiviral responses, protect against type 1 diabetes. Science 324, 387–389 (2009).

    Article  CAS  Google Scholar 

  24. Crow, Y.J. & Rehwinkel, J. Aicardi-Goutières syndrome and related phenotypes: linking nucleic acid metabolism with autoimmunity. Hum. Mol. Genet. 18, R130–R136 (2009).

    Article  CAS  Google Scholar 

  25. Stetson, D.B. Endogenous retroelements and autoimmune disease. Curr. Opin. Immunol. 24, 692–697 (2012).

    Article  CAS  Google Scholar 

  26. Deddouche, S. et al. Identification of an LGP2-associated MDA5 agonist in picornavirus-infected cells. eLife 3, e01535 (2014).

    Article  Google Scholar 

  27. Funabiki, M. et al. Autoimmune disorders associated with gain of function of the intracellular sensor MDA5. Immunity 40, 199–212 (2014).

    Article  CAS  Google Scholar 

  28. Crow, Y.J. et al. Therapies in Aicardi-Goutières syndrome. Clin. Exp. Immunol. 175, 1–8 (2014).

    Article  CAS  Google Scholar 

  29. Herries, D.G. Enzyme Structure and Mechanism 2nd edn. (W H Freeman, New York, 1985).

Download references

Acknowledgements

We sincerely thank the participating families for the use of genetic samples and clinical information and all clinicians who contributed samples and data not included in this manuscript. We thank D. Chase for proofreading the manuscript. We thank G. Notarangelo for helpful discussion. Y.d.T.D. holds a Novartis Foundation postdoctoral fellowship. S.H. holds a Pew scholarship and Career Development award from Boston Children's Hospital. Y.J.C. acknowledges the Manchester Biomedical Research Centre, Manchester Academic Health Sciences Centre, the Greater Manchester Comprehensive Local Research Network, the Great Ormond Street Hospital Children's Charity, the European Union's Seventh Framework Programme (FP7/2007-2013) under grant agreement 241779 and the European Research Council (GA 309449). The authors would like to thank the NHLBI GO ESP and its ongoing studies, which produced and provided exome variant calls for comparison: the Lung GO Sequencing Project (HL-102923), the Women's Health Initiative (WHI) Sequencing Project (HL-102924), the Broad GO Sequencing Project (HL-102925), the Seattle GO Sequencing Project (HL-102926) and the Heart GO Sequencing Project (HL-103010).

Author information

Authors and Affiliations

Authors

Contributions

B.H.A., J.O. and S.G.W. performed exome sequencing. E.M.J., G.I.R. and Y.J.C. performed exome data analysis. G.I.R. performed quantitative PCR analysis and Sanger sequencing with assistance from E.M.J. and G.M.A.F. G.M.A.F. and B.H.A. performed genotyping analysis with assistance from G.I.R. Y.d.T.D. performed IFIH1 protein studies. S.H. performed modeling studies. Y.J.C. and S.H. designed and supervised the project and wrote the manuscript, with support from G.I.R. G.A., B.B.-M., E.M.B., R.B., M.W.B., M. Casarano, M. Chouchane, R.C., A.E.C., N.J.V.C., R.C.D., J.E.D., L.D.W., I.D., L.F., E.F., B.I., L.L., A.R.L., P.L., C.L., J.H.L., C.M.L., M.M.M., A.M.-P., I.B.M., M.P.M., C.M., S.O., P.P.P., E.R., R.A.R., D.R., E.S., C.S., M.S., J.L.T., A.V., C.V., J.P.V., K.W., R.N.W., L.A.W. and S.M.Z. identified affected individuals or assisted with related clinical and laboratory studies.

Corresponding authors

Correspondence to Sun Hur or Yanick J Crow.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Families with mutations in IFIH1.

Family number is shown above each pedigree. Sequence chromatograms are given where available. IFIH1 protein status is stated if known, with WT denoting wild type. De novo mutations are annotated in parentheses. Uncolored shapes represent unaffected individuals. Black-filled shapes represent clinically affected individuals. Gray-filled shapes represent clinically asymptomatic mutation-positive individuals. A diagonal line indicates deceased status. IS denotes interferon score (for participants with repeat samples, the mean combined measurement is given).

Supplementary Figure 2 CLUSTAL Omega alignment of IFIH1 homologs.

IFIH1 homologs were identified on Ensembl and aligned using CLUSTAL Omega. Amino acids altered by IFIH1 mutations are highlighted by a red box. H.Sapiens, Homo sapiens (ENSP00000263642); P.troglodytes, Pan troglodytes (ENSPTRG00000012582); M.mulatta, Macaca mulatta (ENSMMUG00000003202); R.Norvegicus, Rattus norvegicus (ENSRNOG00000006227); M.Musculus, Mus musculus (ENSMUSG00000026896); C.Familiaris, Canis familiaris (ENSCAFG00000010438); B.Taurus, Bos Taurus (ENSBTAG00000008142); G.gallus, Gallus gallus (ENSGALG00000011089); X.Tropicalis, Xenopus tropicalis (ENSXETG00000013176); T.nigroviridis, Tetraodon nigroviridis (ENSTNIG00000016500); D.Rerio, Danio rerio (ENSDARG00000018553). Homology to the human IFIH1 reference sequence (ENSP00000263642): human–P. troglodytes, 100%; human–M. mulatta, 98%; human–B. torus, 84%; human–C. familiaris, 83%; human–M. musculus, 80%; human–R. norvegicus, 80%; human–G. gallus, 56%; human–X. tropicalis, 55%; human–D. rerio, 49%; human–T. nigroviridis, 44%.

Supplementary Figure 3 CLUSTAL Omega alignment of RNA helicases.

RNA helicases were identified on Ensembl and aligned using CLUSTAL Omega. Amino acids altered by IFIH1 mutations are highlighted by a red box. IFIH1 (ENSP00000263642), DDX58 (RIG-I) (ENSP00000369213), DHX58 (LGP2) (ENSP00000251642). Homology of human IFIH1 (reference sequence ENSP00000263642) to DDX58 (RIG-I) and DHX58 (LGP2) is 31% and 41%, respectively.

Supplementary Figure 4 Individual ISG transcript levels in IFIH1 mutation–positive individuals, their IFIH1 mutation–negative relatives and a cohort of 29 controls.

qPCR of a panel of six ISGs in whole blood measured in 5 families with mutations in IFIH1 and 29 healthy controls. RQ is equal to 2−ΔΔCt, with –ΔΔCt ± s.d. (i.e., the normalized fold change relative to a calibrator). Each value is derived from three technical replicates. For ease of presentation, mutation-negative relatives (n = 13) have been combined with the control cohort (M-NR+C) in this figure. Horizontal red bars show the median RQ value for each probe in each group. For individuals with repeat samples (biological replicates), all measurements are shown. Data were analyzed by one-way ANOVA using Dunnett's multiple-comparison test. ***P < 0.0001.

Source data

Supplementary information

Supplementary Text and Figures

Supplementary Note, Supplementary Figures 1–4 and Supplementary Tables 1–7 (PDF 1979 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Rice, G., del Toro Duany, Y., Jenkinson, E. et al. Gain-of-function mutations in IFIH1 cause a spectrum of human disease phenotypes associated with upregulated type I interferon signaling. Nat Genet 46, 503–509 (2014). https://doi.org/10.1038/ng.2933

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.2933

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