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:

Two distinct ubiquitin-binding motifs in A20 mediate its anti-inflammatory and cell-protective activities

Abstract

Protein ubiquitination regulates protein stability and modulates the composition of signaling complexes. A20 is a negative regulator of inflammatory signaling, but the molecular mechanisms involved are ill understood. Here, we generated Tnfaip3 gene-targeted A20 mutant mice bearing inactivating mutations in the zinc finger 7 (ZnF7) and ZnF4 ubiquitin-binding domains, revealing that binding to polyubiquitin is essential for A20 to suppress inflammatory disease. We demonstrate that a functional ZnF7 domain was required for recruiting A20 to the tumor necrosis factor receptor 1 (TNFR1) signaling complex and to suppress inflammatory signaling and cell death. The combined inactivation of ZnF4 and ZnF7 phenocopied the postnatal lethality and severe multiorgan inflammation of A20-deficient mice. Conditional tissue-specific expression of mutant A20 further revealed the key role of ubiquitin-binding in myeloid and intestinal epithelial cells. Collectively, these results demonstrate that the anti-inflammatory and cytoprotective functions of A20 are largely dependent on its ubiquitin-binding properties.

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: A20ZnF7/ZnF7 knock-in mice develop spontaneous inflammatory pathology and are sensitized to TNF-induced toxicity.
Fig. 2: ZnF7 is critical for A20-mediated suppression of inflammatory signaling and cell death.
Fig. 3: A20ZnF4ZnF7/ZnF4ZnF7 knock-in mice phenocopy A20 knockout mice.
Fig. 4: Tissue-specific A20ZnF4ZnF7 expression phenocopies tissue-specific A20 deficiency.

Similar content being viewed by others

Data availability

Source data for Fig. 2 and Extended Data Fig. 4 are included with this paper. All other data supporting the findings of this study are available from the corresponding author on reasonable request.

References

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

    Article  CAS  Google Scholar 

  2. Martens, A. & van Loo, G. A20 at the crossroads of cell death, inflammation, and autoimmunity. Cold Spring Harb. Perspect. Biol. 12, a036418 (2020).

    Article  Google Scholar 

  3. 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 

  4. 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 

  5. De, A., Dainichi, T., Rathinam, C. V. & Ghosh, S. The deubiquitinase activity of A20 is dispensable for NF-κB signaling. EMBO Rep. 15, 775–783 (2014).

    Article  CAS  Google Scholar 

  6. Wertz, I. E. et al. Phosphorylation and linear ubiquitin direct A20 inhibition of inflammation. Nature 528, 370–375 (2015).

    Article  CAS  Google Scholar 

  7. 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 

  8. Tokunaga, F. et al. Specific recognition of linear polyubiquitin by A20 zinc finger 7 is involved in NF-κB regulation. EMBO J. 31, 3856–3870 (2012).

    Article  CAS  Google Scholar 

  9. Draber, P. et al. LUBAC-recruited CYLD and A20 regulate gene activation and cell death by exerting opposing effects on linear ubiquitin in signaling complexes. Cell Rep. 13, 2258–2272 (2015).

    Article  CAS  Google Scholar 

  10. Verhelst, K. et al. A20 inhibits LUBAC-mediated NF-κB activation by binding linear polyubiquitin chains via its zinc finger 7. EMBO J. 31, 3845–3855 (2012).

    Article  CAS  Google Scholar 

  11. Polykratis, A. et al. A20 prevents inflammasome-dependent arthritis by inhibiting macrophage necroptosis through its ZnF7 ubiquitin-binding domain. Nat. Cell Biol. 21, 731–742 (2019).

    Article  CAS  Google Scholar 

  12. Turer, E. E. et al. Homeostatic MyD88-dependent signals cause lethal inflammation in the absence of A20. J. Exp. Med. 205, 451–464 (2008).

    Article  CAS  Google Scholar 

  13. Vereecke, L. et al. Enterocyte-specific A20 deficiency sensitizes to tumor necrosis factor-induced toxicity and experimental colitis. J. Exp. Med. 207, 1513–1523 (2010).

    Article  CAS  Google Scholar 

  14. Priem, D. et al. A20 protects cells from TNF-induced apoptosis through linear ubiquitin-dependent and -independent mechanisms. Cell Death Dis. 10, 692 (2019).

    Article  Google Scholar 

  15. 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 

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

    Article  Google Scholar 

  17. 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 

  18. 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 

  19. 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 

  20. Sokhi, U. K. et al. Dissection and function of autoimmunity-associated TNFAIP3 (A20) gene enhancers in humanized mouse models. Nat. Commun. 9, 658 (2018).

    Article  Google Scholar 

  21. Wang, S., Wen, F., Wiley, G. B., Kinter, M. T. & Gaffney, P. M. An enhancer element harboring variants associated with systemic lupus erythematosus engages the TNFAIP3 promoter to influence A20 expression. PLoS Genet. 9, e1003750 (2013).

    Article  CAS  Google Scholar 

  22. Compagno, M. et al. Mutations of multiple genes cause deregulation of NF-κB in diffuse large B-cell lymphoma. Nature 459, 717–721 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  24. Schmitz, R. et al. TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. J. Exp. Med. 206, 981–989 (2009).

    Article  CAS  Google Scholar 

  25. Zhou, Q. et al. Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease. Nat. Genet. 48, 67–73 (2016).

    Article  CAS  Google Scholar 

  26. Clausen, B. E., Burkhardt, C., Reith, W., Renkawitz, R. & Forster, I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8, 265–277 (1999).

    Article  CAS  Google Scholar 

  27. Madison, B. B. et al. cis Elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J. Biol. Chem. 277, 33275–33283 (2002).

    Article  CAS  Google Scholar 

  28. Adachi, O. et al. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity 9, 143–150 (1998).

    Article  CAS  Google Scholar 

  29. Armaka, M., Ospelt, C., Pasparakis, M. & Kollias, G. The p55TNFR–IKK2–Ripk3 axis orchestrates arthritis by regulating death and inflammatory pathways in synovial fibroblasts. Nat. Commun. 9, 618 (2018).

    Article  Google Scholar 

  30. Baird, D., Murray, D., Payne, R. & Soutar, D. An Introduction to GenStat for Windows v.19 (Genstat, 2017).

Download references

Acknowledgements

We thank D. Huyghebaert, L. Bellen and D. Vanhede for animal care and A. Fossoul and M. Gennadi for excellent technical assistance. We also thank the InfrafrontierGR infrastructure (ERDF and NSRF 2007–2013 and 2014–2020) for providing histology and mCT facilities. A.M. is supported by a grant from the ‘Concerted Research Actions’ (GOA) of the Ghent University. Research in the G.v.L. laboratory is supported by research grants from The Research Foundation—Flanders (FWO), the ‘Geneeskundige Stichting Koningin Elisabeth’ (GSKE), the CBC Banque Prize, the Charcot Foundation, the ‘Belgian Foundation against Cancer’, ‘Kom op tegen Kanker’ and the GOA of Ghent University. The M.A. laboratory is supported by a start-up grant from the Stavros Niarchos Foundation to BSRC ‘Alexander Fleming’.

Author information

Authors and Affiliations

Authors

Contributions

A.M., D.P., E.H., J.V., S.R., L.C., S.V., L.D., M.S., H.V., K.S., T.H. and K.I. performed the experiments. A.M., D.P., E.H., J.V., L.C., S.V., A.W., S.J., M.L., R.B., M.A., M.J.M.B. and G.v.L. analyzed the data. G.v.L. provided ideas and coordinated the project. A.M. and G.v.L. wrote the manuscript.

Corresponding author

Correspondence to Geert van Loo.

Ethics declarations

Competing interests

M.L. is an employee of Janssen Pharmaceutica. All other authors declare no competing interests.

Additional information

Editor recognition statement: L. A. Dempsey 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 A20ZnF7 knock-in mice.

a, Schematic depiction of the A20/Tnfaip3 locus indicating the sequence of the single-stranded oligonucleotide used for mutating the ZnF7 domain that was introduced by pronuclear injection into mouse zygotes, and sequencing result of the wild-type (WT) and of the targeted ZnF7 knock-in allele. Boxes indicate exons 3 to 9 (E3–E9). b, Birth and survival rates of control (A20+/+), A20ZnF7/+ and A20ZnF7/ZnF7 offspring from A20ZnF7/+ x A20ZnF7/+ breeding couples. c, Gross appearance of A20ZnF7/+ and A20ZnF7/ZnF7 mice. d, Representative pictures of spleen and inguinal lymph nodes from 28-week-old control (A20+/+) and A20ZnF7/ZnF7 littermate mice. e, Representative pictures of hindpaws of 15-week-old control (A20+/+) and A20ZnF7/ZnF7 littermates, showing extensive swelling of the toes of the A20ZnF7/ZnF7 mice. f, Representative micro-CT pictures of hindpaws (left) and knees (right) of 28-week-old control (A20+/+) and A20ZnF7/ZnF7 littermates. g, Representative hematoxylin-eosin-stained histological images of ankle joints (left) and toes (right) from 28-week-old control (A20+/+) and A20ZnF7/ZnF7 littermates. Scale bar, 500 µm.

Extended Data Fig. 2 FACS immunophenotyping of spleen of control and A20ZnF7 mice.

ac, General gating strategy as applied for immune cell populations described in Fig. 1g. (a) Lymphocytes, singlets, live, CD3CD19 + (B cells), CD3 + CD19 (T cells) and CD3CD19NK1.1+ (NK cells); (b) non-debris, singlets, live, lineage (CD3CD19NK1.1-), F4/80+, CD64 + and autofluorescent; (c) non-debris, singlets, live, lineage, Ly6G+CD11b + (neutrophils) and Ly6GSiglecFLy6ChiCD11b + (monocytes). FSC: forward scatter, SSC: side scatter, A: Area, H: height, W: width, L/D: live/dead. df, Bar graphs representing absolute numbers of total (left) and naive (right) CD4 T cells (d), total (left) and naive (right) CD8 T cells (e) and yd T cells (f) as measured by flow cytometry in the spleens of A20+/+, A20ZnF/+ and A20ZnF7/ZnF7 animals. Data are expressed as mean ± SEM. *, ** represent p < 0.05 and p < 0.01 (Two-sided non-parametric Mann-Whitney test between indicated genotypes).

Extended Data Fig. 3 MyD88-dependent mechanisms contribute to the local inflammatory pathology in A20ZnF7 mice.

a, b, Gross appearance (a) and body weight (b) of of 10 week-old A20ZnF7/+MyD88+/-, A20ZnF7/ZnF7MyD88+/- and A20ZnF7/ZnF7 MyD88-/- mice. Each dot represents a biologically independent mouse (A20ZnF7/+MyD88+/-, n = 9; A20ZnF7/ZnF7MyD88+/-, n = 13 and A20ZnF7/ZnF7 MyD88-/-, n = 6). Data are expressed as mean ± SEM. * and **** represent p < 0.05 and p < 0.0001, respectively (parametric two-way ANOVA between indicated genotypes). c, Representative hematoxylin-eosin-stained sections of liver from 18-week-old A20ZnF7/ZnF7MyD88+/+ and A20ZnF7/ZnF7MyD88-/- littermates. Scale bar, 50 μm. Picture representative for 3 biologically independent mice. d, Representative pictures of hindpaws of 10-week-old A20ZnF7/ZnF7MyD88+/+ and A20ZnF7/ZnF7MyD88-/- littermates. Pictures representative for 3 biologically independent mice e, Levels of IL-6 and TNF in serum of A20+/+ MyD88+/+, A20+/+ MyD88-/-, A20ZnF7/ZnF7MyD88+/+ and A20ZnF7/ZnF7MyD88-/- mice. Each dot represents a biologically independent mouse (A20+/+ MyD88+/+, n = 9; A20+/+ MyD88-/-, n = 3; A20ZnF7/ZnF7MyD88+/+, n = 13 and A20ZnF7/ZnF7MyD88-/-, n = 6). Data are expressed as mean ± SEM. *, ** represent p < 0.05 and p = 0.0033 respectively (parametric one-way ANOVA between indicated genotypes).

Extended Data Fig. 4 ZnF7 is critical for A20-mediated suppression of inflammatory signaling.

Western blot analysis of whole cell lysates from A20+/+, A20ZnF7/ZnF7 and A20myel-KO BMDMs stimulated with TNF as indicated. β-tubulin is shown as a loading control. Figure representative for 3 independent experiments.

Source data

Extended Data Fig. 5 A20ZnF4ZnF7 knock-in mice.

a, Schematic depiction of the A20/Tnfaip3 locus indicating the position of ZnF4 and ZnF7 mutations. Boxes indicate exons 3 to 9 (E3–E9). Sequences of the donor vector, containing ~1 kb 5′ and 3′ homologous arms around the Cys-to-Ala mutations used for mutating the ZnF4 and ZnF7 domains, that were introduced by pronuclear injection into mouse zygotes. Sequencing result of the wild-type (WT) allele and of the targeted ZnF4 and ZnF7 knock-in alleles. b, Gross appearance of 2-week old control (A20+/+), A20ZnF4ZnF7/+ and A20ZnF4ZnF7/ZnF4ZnF7 mice. c, Gross appearance of 2-week old A20ZnF4ZnF7/ZnF4ZnF7MyD88-/- mice compared to A20ZnF4ZnF7/ZnF4ZnF7MyD88+/-mice.

Extended Data Fig. 6 Conditional ‘floxed’ A20ZnF4/ZnF7 knock-in mice.

a, Targeting scheme showing the LoxP-flanked (Floxed) and knock-in A20 alleles. Boxes indicate exons 3 to 9 (E3-E9). LoxP sites are indicated by arrowheads. b, Gross appearance of 2 week-old control (Tnfaip3ZnF4ZnF7/ZnF4ZnF7CreDel+/+) and Tnfaip3ZnF4ZnF7/ZnF4ZnF7CreDelTg/+ littermate mice. c, Representative histological images of ankle joints from 30-week-old littermate mice with the indicated genotypes. Bone erosion was detected by tartrate-resistant acid phosphatase (TRAP) staining of osteoclast activity, and cartilage destruction was assessed by proteoglycan staining with toluidine blue. H/E, haematoxylin and eosin. Scale bar: 500 μm. Pictures representative for 5 biologically independent mice. d, Clinical score, based on loss in body weight, stool consistency, and presence of fecal blood, of 30 week-old Tnfaip3ZnF4ZnF7/ZnF4ZnF7vilCreTg/+ (n = 4) and control (Tnfaip3ZnF4ZnF7/ZnF4ZnF7vilCre+/+, n = 4) littermate mice treated with 1.5 % DSS. The experiment was stopped at day 5 since Tnfaip3ZnF4ZnF7/ZnF4ZnF7vilCreTg/+ started dying. Data are expressed as mean ± SEM. * represents p = 0.0204 (2-way ANOVA with Sidak’s multiple comparison) e, Intestinal permeability assay using FITC-labelled dextran in 30-week-old Tnfaip3ZnF4ZnF7/ZnF4ZnF7vilCreTg/+ (n = 4) and control (Tnfaip3ZnF4ZnF7/ZnF4ZnF7vilCre+/+, n = 4) mice before and after 5 days of DSS treatment. Data are expressed as mean ± SEM. * represents p = 0.0143 (2-way ANOVA with Sidak’s multiple comparison).

Supplementary information

Source data

Source Data Fig. 2

Unprocessed western blots

Source Data Extended Data Fig. 4

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Martens, A., Priem, D., Hoste, E. et al. Two distinct ubiquitin-binding motifs in A20 mediate its anti-inflammatory and cell-protective activities. Nat Immunol 21, 381–387 (2020). https://doi.org/10.1038/s41590-020-0621-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0621-9

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