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:

Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease

This article has been updated

Abstract

Systemic autoinflammatory diseases are driven by abnormal activation of innate immunity1. Herein we describe a new disease caused by high-penetrance heterozygous germline mutations in TNFAIP3, which encodes the NF-κB regulatory protein A20, in six unrelated families with early-onset systemic inflammation. The disorder resembles Behçet's disease, which is typically considered a polygenic disorder with onset in early adulthood2. A20 is a potent inhibitor of the NF-κB signaling pathway3. Mutant, truncated A20 proteins are likely to act through haploinsufficiency because they do not exert a dominant-negative effect in overexpression experiments. Patient-derived cells show increased degradation of IκBα and nuclear translocation of the NF-κB p65 subunit together with increased expression of NF-κB–mediated proinflammatory cytokines. A20 restricts NF-κB signals via its deubiquitinase activity. In cells expressing mutant A20 protein, there is defective removal of Lys63-linked ubiquitin from TRAF6, NEMO and RIP1 after stimulation with tumor necrosis factor (TNF). NF-κB–dependent proinflammatory cytokines are potential therapeutic targets for the patients with this disease.

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: TNFAIP3 mutations cause a dominantly inherited systemic inflammatory disease.
Figure 2: Enhanced NF-κB signaling in transiently transfected 293T cells and patient-derived cells.
Figure 3: Impaired TNFR signaling and deubiquitinase function of mutant A20.
Figure 4: Patient-derived immune cells have a strong inflammatory signature.
Figure 5: Spontaneous NLRP3 inflammasome activity in PBMCs from patients with TNFAIP3 mutations.

Similar content being viewed by others

Accession codes

Primary accessions

Sequence Read Archive

Referenced accessions

NCBI Reference Sequence

Change history

  • 16 December 2015

    In the version of this article initially published online, the name of author Jonathan J. Lyons was misspelled, and the p.Thr604Argfs*93 variant identified in family 4 was listed incorrectly in Figure 1a. These errors have been corrected for the print, PDF and HTML versions of this article

References

  1. van Kempen, T.S., Wenink, M.H., Leijten, E.F., Radstake, T.R. & Boes, M. Perception of self: distinguishing autoimmunity from autoinflammation. Nat. Rev. Rheumatol. 11, 483–492 (2015).

    Article  CAS  Google Scholar 

  2. Sakane, T., Takeno, M., Suzuki, N. & Inaba, G. Behçet’s disease. N. Engl. J. Med. 341, 1284–1291 (1999).

    Article  CAS  Google Scholar 

  3. Catrysse, L., Vereecke, L., Beyaert, R. & van Loo, G. A20 in inflammation and autoimmunity. Trends Immunol. 35, 22–31 (2014).

    Article  CAS  Google Scholar 

  4. Russo, R.A. & Brogan, P.A. Monogenic autoinflammatory diseases. Rheumatology 53, 1927–1939 (2014).

    Article  CAS  Google Scholar 

  5. Martinon, F. & Aksentijevich, I. New players driving inflammation in monogenic autoinflammatory diseases. Nat. Rev. Rheumatol. 11, 11–20 (2015).

    Article  CAS  Google Scholar 

  6. Moghaddas, F. & Masters, S.L. Monogenic autoinflammatory diseases: cytokinopathies. Cytokine 74, 237–246 (2015).

    Article  CAS  Google Scholar 

  7. Jesus, A.A. & Goldbach-Mansky, R. IL-1 blockade in autoinflammatory syndromes. Annu. Rev. Med. 65, 223–244 (2014).

    Article  CAS  Google Scholar 

  8. Kuehn, H.S. et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 345, 1623–1627 (2014).

    Article  CAS  Google Scholar 

  9. Flanagan, S.E. et al. Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat. Genet. 46, 812–814 (2014).

    Article  CAS  Google Scholar 

  10. Malynn, B.A. & Ma, A. A20 takes on tumors: tumor suppression by an ubiquitin-editing enzyme. J. Exp. Med. 206, 977–980 (2009).

    Article  CAS  Google Scholar 

  11. Hymowitz, S.G. & Wertz, I.E. A20: from ubiquitin editing to tumour suppression. Nat. Rev. Cancer 10, 332–341 (2010).

    Article  CAS  Google Scholar 

  12. Wertz, I.E. et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-κB signalling. Nature 430, 694–699 (2004).

    Article  CAS  Google Scholar 

  13. Bosanac, I. et al. Ubiquitin binding to A20 ZnF4 is required for modulation of NF-κB signaling. Mol. Cell 40, 548–557 (2010).

    Article  CAS  Google Scholar 

  14. Lu, T.T. et al. Dimerization and ubiquitin mediated recruitment of A20, a complex deubiquitinating enzyme. Immunity 38, 896–905 (2013).

    Article  CAS  Google Scholar 

  15. Ma, A. & Malynn, B.A. A20: linking a complex regulator of ubiquitylation to immunity and human disease. Nat. Rev. Immunol. 12, 774–785 (2012).

    Article  CAS  Google Scholar 

  16. Coornaert, B., Carpentier, I. & Beyaert, R. A20: central gatekeeper in inflammation and immunity. J. Biol. Chem. 284, 8217–8221 (2009).

    Article  CAS  Google Scholar 

  17. Vande Walle, L. et al. Negative regulation of the NLRP3 inflammasome by A20 protects against arthritis. Nature 512, 69–73 (2014).

    Article  Google Scholar 

  18. Duong, B.H. et al. A20 restricts ubiquitination of pro-interleukin-1β protein complexes and suppresses NLRP3 inflammasome activity. Immunity 42, 55–67 (2015).

    Article  CAS  Google Scholar 

  19. Lee, G.S. et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 492, 123–127 (2012).

    Article  CAS  Google Scholar 

  20. Coll, R.C. et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat. Med. 21, 248–255 (2015).

    Article  CAS  Google Scholar 

  21. Lee, E.G. et al. Failure to regulate TNF-induced NF-κB and cell death responses in A20-deficient mice. Science 289, 2350–2354 (2000).

    Article  CAS  Google Scholar 

  22. Martin, F. & Dixit, V.M. A20 edits ubiquitin and autoimmune paradigms. Nat. Genet. 43, 822–823 (2011).

    Article  CAS  Google Scholar 

  23. Matmati, M. et al. A20 (TNFAIP3) deficiency in myeloid cells triggers erosive polyarthritis resembling rheumatoid arthritis. Nat. Genet. 43, 908–912 (2011).

    Article  CAS  Google Scholar 

  24. Tavares, R.M. et al. The ubiquitin modifying enzyme A20 restricts B cell survival and prevents autoimmunity. Immunity 33, 181–191 (2010).

    Article  CAS  Google Scholar 

  25. Kato, M. et al. Frequent inactivation of A20 in B-cell lymphomas. Nature 459, 712–716 (2009).

    Article  CAS  Google Scholar 

  26. Graham, R.R. et al. Genetic variants near TNFAIP3 on 6q23 are associated with systemic lupus erythematosus. Nat. Genet. 40, 1059–1061 (2008).

    Article  CAS  Google Scholar 

  27. Plenge, R.M. et al. Two independent alleles at 6q23 associated with risk of rheumatoid arthritis. Nat. Genet. 39, 1477–1482 (2007).

    Article  CAS  Google Scholar 

  28. Thomson, W. et al. Rheumatoid arthritis association at 6q23. Nat. Genet. 39, 1431–1433 (2007).

    Article  CAS  Google Scholar 

  29. Lodolce, J.P. et al. African-derived genetic polymorphisms in TNFAIP3 mediate risk for autoimmunity. J. Immunol. 184, 7001–7009 (2010).

    Article  CAS  Google Scholar 

  30. Musone, S.L. et al. Multiple polymorphisms in the TNFAIP3 region are independently associated with systemic lupus erythematosus. Nat. Genet. 40, 1062–1064 (2008).

    Article  CAS  Google Scholar 

  31. Adrianto, I. et al. Association of a functional variant downstream of TNFAIP3 with systemic lupus erythematosus. Nat. Genet. 43, 253–258 (2011).

    Article  CAS  Google Scholar 

  32. Nair, R.P. et al. Genome-wide scan reveals association of psoriasis with IL-23 and NF-κB pathways. Nat. Genet. 41, 199–204 (2009).

    Article  CAS  Google Scholar 

  33. Fung, E.Y. et al. Analysis of 17 autoimmune disease–associated variants in type 1 diabetes identifies 6q23/TNFAIP3 as a susceptibility locus. Genes Immun. 10, 188–191 (2009).

    Article  CAS  Google Scholar 

  34. Trynka, G. et al. Coeliac disease–associated risk variants in TNFAIP3 and REL implicate altered NF-κB signalling. Gut 58, 1078–1083 (2009).

    Article  CAS  Google Scholar 

  35. Musone, S.L. et al. Sequencing of TNFAIP3 and association of variants with multiple autoimmune diseases. Genes Immun. 12, 176–182 (2011).

    Article  CAS  Google Scholar 

  36. Nititham, J. et al. Meta-analysis of the TNFAIP3 region in psoriasis reveals a risk haplotype that is distinct from other autoimmune diseases. Genes Immun. 16, 120–126 (2015).

    Article  CAS  Google Scholar 

  37. Schuijs, M.J. et al. Farm dust and endotoxin protect against allergy through A20 induction in lung epithelial cells. Science 349, 1106–1110 (2015).

    Article  CAS  Google Scholar 

  38. Rieux-Laucat, F. & Casanova, J.L. Autoimmunity by haploinsufficiency. Science 345, 1560–1561 (2014).

    Article  CAS  Google Scholar 

  39. Boisson, B. et al. Immunodeficiency, autoinflammation and amylopectinosis in humans with inherited HOIL-1 and LUBAC deficiency. Nat. Immunol. 13, 1178–1186 (2012).

    Article  CAS  Google Scholar 

  40. Boisson, B. et al. Human HOIP and LUBAC deficiency underlies autoinflammation, immunodeficiency, amylopectinosis, and lymphangiectasia. J. Exp. Med. 212, 939–951 (2015).

    Article  CAS  Google Scholar 

  41. Zhou, Q. et al. Early-onset stroke and vasculopathy associated with mutations in ADA2. N. Engl. J. Med. 370, 911–920 (2014).

    Article  CAS  Google Scholar 

  42. Kirino, Y. et al. Targeted resequencing implicates the familial Mediterranean fever gene MEFV and the Toll-like receptor 4 gene TLR4 in Behçet disease. Proc. Natl. Acad. Sci. USA 110, 8134–8139 (2013).

    Article  CAS  Google Scholar 

  43. Lim, K.L. et al. Parkin mediates nonclassical, proteasomal-independent ubiquitination of synphilin-1: implications for Lewy body formation. J. Neurosci. 25, 2002–2009 (2005).

    Article  CAS  Google Scholar 

  44. Li, L. et al. Localization of A20 to a lysosome-associated compartment and its role in NFκB signaling. Biochim. Biophys. Acta 1783, 1140–1149 (2008).

    Article  CAS  Google Scholar 

  45. Ma, C.A. et al. Dendritic cells from humans with hypomorphic mutations in IKBKG/NEMO have impaired mitogen-activated protein kinase activity. Hum. Mutat. 32, 318–324 (2011).

    Article  CAS  Google Scholar 

  46. Wang, H.Y. et al. Cbl promotes ubiquitination of the T cell receptor ζ through an adaptor function of Zap-70. J. Biol. Chem. 276, 26004–26011 (2001).

    Article  CAS  Google Scholar 

  47. Wang, H.Y. et al. Antibody deficiency associated with an inherited autosomal dominant mutation in TWEAK. Proc. Natl. Acad. Sci. USA 110, 5127–5132 (2013).

    Article  CAS  Google Scholar 

  48. Zhang, Y. et al. Autosomal recessive phosphoglucomutase 3 (PGM3) mutations link glycosylation defects to atopy, immune deficiency, autoimmunity, and neurocognitive impairment. J. Allergy Clin. Immunol. 133, 1400–1409 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank V. Dixit for helpful discussions. We also thank all the patients and their families, and the healthy children and adult controls, for their enthusiastic support during this research study. This research was supported by the Intramural Research Programs of the National Human Genome Research Institute, the National Institute of Arthritis and Musculoskeletal and Skin Diseases, the National Heart, Lung, and Blood Institute, the National Institute of Allergy and Infectious Diseases, and the US National Institutes of Health Clinical Center. S.O. received royalties for consulting and speaking from Novartis and SOBI. H.L.L. received royalties for consulting from Baxter. E.D. was recipient of the Research Fellowship Program for International Researchers, which is supported by the Scientific and Technological Research Council of Turkey (TUBITAK; B.14.2.TBT.0.06.01-219-84).

Author information

Authors and Affiliations

Authors

Contributions

Q.Z., H.W., D.L.K. and I.A. designed the study. Q.Z., H.W., M.S., Y.H.P., Y.Z., D.Y., M.T., W.L.T., J.J.L., X.Y., C.O., C.C., D.T.C., S.C.C., E.P.H. and Z.Y. performed experiments. Q.Z., H.W., K.Z., R.M.S., M.B., J.D.M., M.G. and J.C. analyzed and interpreted the data. D.M.S., E.D., A.K.O., D.L.S., P.H., S.A.H., A.J., B.K.B., H.L.L., A.v.R.-K., C.S., E.D.B., A.G., S.O., R.M.L. and D.L.K. enrolled the patients and collected and interpreted clinical information. K.Z., J.C.M. and I.E.W. provided technical support and comments. Q.Z., H.W., R.M.S., D.L.K. and I.A. wrote the manuscript. D.L.K. and I.A. directed and supervised the research. All authors contributed to the review and approval of the manuscript.

Corresponding author

Correspondence to Ivona Aksentijevich.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 TNFAIP3-associated haplotypes in family 1.

The p.Leu227* mutation (shown as a star) arose de novo in the first affected member of the family (P1). Red haplotypes carry the TNFAIP3 p.Leu227* mutation. Haplotypes of unavailable family members were inferred from reconstructed haplotypes and are shown in a purple font.

Supplementary Figure 2 Identification of TNFAIP3 mutations using exome sequencing, Sanger sequencing and targeted sequencing.

(a) Whole-exome sequencing identified a common gene mutated in two families. Schematic representation of the exome data-filtering approach used to select for novel and dominantly inherited variants segregating with disease in family 1 and family 2. TNFAIP3 is the only gene in common for these two families. SNV, single-nucleotide variants, including missense variants, splice-site variants and stop codon variants; INDEL, frameshift and non-frameshift insertions and deletions. (b) Electropherograms for the five TNFAIP3 mutations identified in five families. M1, M2, M3, M4 and M5 indicate TNFAIP3 mutant alleles. (c) A p.Pro268Leufs*19 mutation was identified by targeted gene sequencing. Sanger sequencing validated the p.Pro268Leufs*19 mutation in the proband and confirmed its presence in the other two affected family members. Age of onset of the proband was 29 years; her older daughter was 15 years old, and her younger daughter was 13 years old. The early symptoms included oral and genital ulcers, mild fever and skin rash.

Supplementary Figure 3 NF-κB reporter assay in a human T cell lymphoblast-like cell line (Jurkat cells).

NF-κB activity was assayed in cells transiently transfected with a mock control or with wild-type (WT) or mutant TNFAIP3 plasmid. NF-κB activity was determined on the basis of gated GFP+ cells (bottom). Mutant plasmids were less efficient in suppressing NF-κB activity than the wild-type plasmid. The mean fluorescence intensity (MFI) of Thy1 is the indicator of NF-κB activity.

Supplementary Figure 4 TNFAIP3 haploinsufficiency causes upregulation of the NF-κB signaling pathway.

(a) Patient 2 (P2) showed increased IκBα degradation and increased phosphorylation of p38 and JNK following stimulation with TNF. (b) Increased ratio of phosphorylated IκBα to total IκBα in PBMCs and fibroblasts in patients (P2 and P6) by ImageJ analysis.

Supplementary Figure 5 TNFAIP3 haploinsufficiency causes upregulation of the NF-κB signaling pathway.

(a) Patient 6 (P6) showed evidence for increased nuclear translocation of the p65 NF-κB subunit in TNF-stimulated PBMCs. (b) Immunofluorescence staining of NF-κB p65 in control (top) and patient-derived (bottom) fibroblasts under no stimulation (left) and with stimulation by 0.2 ng/ml TNF for 30 min (right). p65 translocation is indicated by red arrowheads. (c) The accompanying frequency plot for the different levels of activation under no stimulation (left) and stimulation by 0.2 ng/ml TNF for 30 min (right). Patient-derived fibroblasts are significantly more activated than control fibroblasts (Mann-Whitney test, P < 0.0001) under basal resting conditions and after TNF stimulation, as shown by the shifted distribution of nuclear p65 intensities.

Supplementary Figure 6 Impaired deubiquitinase function of mutant A20 in PBMCs and fibroblasts.

(a) In PBMCs, A20-deficient patients accumulated high-molecular-weight ubiquitin aggregates and K63-ubiquitinated RIP1. Top, ImageJ analysis of high-molecular-weight K63-linked ubiquitin aggregates for Figure 3e, top. Middle, increased K63-ubiquitinated RIP1 in patients. Bottom, RIP1 expression in lysate as control. (b) In fibroblasts, A20-deficient patients accumulated high-molecular-weight ubiquitin aggregates and K63-ubiquitinated RIP1. First panel, ImageJ analysis of high-molecular-weight K63-linked ubiquitin aggregates for Figure 3f, top. Second panel, increased K63-ubiquitinated RIP1 in patients. Third panel, increased high-molecular-weight ubiquitin aggregates of TNFR1 in patients. Fourth and fifth panels, RIP1 and TNFR1 expression in lysate as control, respectively.

Supplementary Figure 7 Gene expression of proinflammatory cytokines in differentiated M1 macrophages.

Human monocytes were isolated and differentiated into M1 macrophages using human GM-CSF (20 ng/ml) as described in the Online Methods. Cells were stimulated with or without LPS (100 ng/ml) for 6 h. Relative mRNA expression of IL-1β and TNF was analyzed in four patients and four healthy controls.

Supplementary Figure 8 Intracellular staining of TNF in T cells.

Increased staining for TNF in CD3+ T cells from patients 1, 4 and 6 is observed in SEB (staphylococcal enterotoxin B)-stimulated cells compared to three non-carrier controls.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1 and 2. (PDF 1350 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, Q., Wang, H., Schwartz, D. et al. Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat Genet 48, 67–73 (2016). https://doi.org/10.1038/ng.3459

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3459

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