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.

  • Article
  • Published:

Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease

Abstract

RIPK1 is a key regulator of innate immune signalling pathways. To ensure an optimal inflammatory response, RIPK1 is regulated post-translationally by well-characterized ubiquitylation and phosphorylation events, as well as by caspase-8-mediated cleavage1,2,3,4,5,6,7. The physiological relevance of this cleavage event remains unclear, although it is thought to inhibit activation of RIPK3 and necroptosis8. Here we show that the heterozygous missense mutations D324N, D324H and D324Y prevent caspase cleavage of RIPK1 in humans and result in an early-onset periodic fever syndrome and severe intermittent lymphadenopathy—a condition we term ‘cleavage-resistant RIPK1-induced autoinflammatory syndrome’. To define the mechanism for this disease, we generated a cleavage-resistant Ripk1D325A mutant mouse strain. Whereas Ripk1−/− mice died postnatally from systemic inflammation, Ripk1D325A/D325A mice died during embryogenesis. Embryonic lethality was completely prevented by the combined loss of Casp8 and Ripk3, but not by loss of Ripk3 or Mlkl alone. Loss of RIPK1 kinase activity also prevented Ripk1D325A/D325A embryonic lethality, although the mice died before weaning from multi-organ inflammation in a RIPK3-dependent manner. Consistently, Ripk1D325A/D325A and Ripk1D325A/+ cells were hypersensitive to RIPK3-dependent TNF-induced apoptosis and necroptosis. Heterozygous Ripk1D325A/+ mice were viable and grossly normal, but were hyper-responsive to inflammatory stimuli in vivo. Our results demonstrate the importance of caspase-mediated RIPK1 cleavage during embryonic development and show that caspase cleavage of RIPK1 not only inhibits necroptosis but also maintains inflammatory homeostasis throughout life.

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: Heterozygous mutations of the RIPK1 caspase-8 cleavage site cause autoinflammatory disease.
Fig. 2: Homozygous mutation of the RIPK1 caspase-8 cleavage site in mice causes early embryonic lethality.
Fig. 3: Ripk1D325A/D325A and Ripk1D325A/+ cells are hypersensitive to TNF-induced death.
Fig. 4: RIPK1 cleavage limits inflammation in vivo.

Similar content being viewed by others

Data availability

The original RNA sequencing data are uploaded and available at the Gene Expression Omnibus (GEO) under accession GSE127572. All other data are available from the corresponding authors upon reasonable request.

References

  1. Bertrand, M. J. M. et al. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol. Cell 30, 689–700 (2008).

    Article  MathSciNet  PubMed  CAS  Google Scholar 

  2. Dondelinger, Y. et al. MK2 phosphorylation of RIPK1 regulates TNF-mediated cell death. Nat. Cell Biol. 19, 1237–1247 (2017).

    Article  PubMed  CAS  Google Scholar 

  3. Dondelinger, Y. et al. NF-κB-independent role of IKKα/IKKβ in preventing RIPK1 kinase-dependent apoptotic and necroptotic cell death during TNF signaling. Mol. Cell 60, 63–76 (2015).

    Article  PubMed  CAS  Google Scholar 

  4. Jaco, I. et al. MK2 phosphorylates RIPK1 to prevent TNF-induced cell death. Mol. Cell 66, 698–710.e5 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Feltham, R. et al. Mind bomb regulates cell death during TNF signaling by suppressing RIPK1’s cytotoxic potential. Cell Reports 23, 470–484 (2018).

    Article  PubMed  CAS  Google Scholar 

  6. Menon, M. B. et al. p38MAPK/MK2-dependent phosphorylation controls cytotoxic RIPK1 signalling in inflammation and infection. Nat. Cell Biol. 19, 1248–1259 (2017).

    Article  PubMed  CAS  Google Scholar 

  7. Lafont, E. et al. TBK1 and IKKε prevent TNF-induced cell death by RIPK1 phosphorylation. Nat. Cell Biol. 20, 1389–1399 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Oberst, A. et al. Catalytic activity of the caspase-8–FLIPL complex inhibits RIPK3-dependent necrosis. Nature 471, 363–367 (2011).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  9. Kim, J. W., Choi, E. J. & Joe, C. O. Activation of death-inducing signaling complex (DISC) by pro-apoptotic C-terminal fragment of RIP. Oncogene 19, 4491–4499 (2000).

    Article  PubMed  CAS  Google Scholar 

  10. Lin, Y., Devin, A., Rodriguez, Y. & Liu, Z. G. Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Genes Dev. 13, 2514–2526 (1999).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. van Raam, B. J., Ehrnhoefer, D. E., Hayden, M. R. & Salvesen, G. S. Intrinsic cleavage of receptor-interacting protein kinase-1 by caspase-6. Cell Death Differ. 20, 86–96 (2013).

    Article  PubMed  CAS  Google Scholar 

  12. Dillon, C. P. et al. RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3. Cell 157, 1189–1202 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Kaiser, W. J. et al. RIP1 suppresses innate immune necrotic as well as apoptotic cell death during mammalian parturition. Proc. Natl Acad. Sci. USA 111, 7753–7758 (2014).

    Article  PubMed  ADS  CAS  PubMed Central  Google Scholar 

  14. Kelliher, M. A. et al. The death domain kinase RIP mediates the TNF-induced NF-κB signal. Immunity 8, 297–303 (1998).

    Article  PubMed  CAS  Google Scholar 

  15. Rickard, J. A. et al. RIPK1 regulates RIPK3-MLKL-driven systemic inflammation and emergency hematopoiesis. Cell 157, 1175–1188 (2014).

    Article  PubMed  CAS  Google Scholar 

  16. Moulin, M. et al. IAPs limit activation of RIP kinases by TNF receptor 1 during development. EMBO J. 31, 1679–1691 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Peltzer, N. et al. LUBAC is essential for embryogenesis by preventing cell death and enabling haematopoiesis. Nature 557, 112–117 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  18. Peltzer, N. et al. HOIP deficiency causes embryonic lethality by aberrant TNFR1-mediated endothelial cell death. Cell Reports 9, 153–165 (2014).

    Article  PubMed  CAS  Google Scholar 

  19. Varfolomeev, E. E. et al. Targeted disruption of the mouse Caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally. Immunity 9, 267–276 (1998).

    Article  PubMed  CAS  Google Scholar 

  20. Yeh, W. C. et al. FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis. Science 279, 1954–1958 (1998).

    Article  PubMed  ADS  CAS  Google Scholar 

  21. Yeh, W. C. et al. Requirement for Casper (c-FLIP) in regulation of death receptor-induced apoptosis and embryonic development. Immunity 12, 633–642 (2000).

    Article  PubMed  CAS  Google Scholar 

  22. Kaiser, W. J. et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 471, 368–372 (2011).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  23. Alvarez-Diaz, S. et al. The pseudokinase MLKL and the kinase RIPK3 have distinct roles in autoimmune disease caused by loss of death-receptor-induced apoptosis. Immunity 45, 513–526 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Zhang, X., Dowling, J. P. & Zhang, J. RIPK1 can mediate apoptosis in addition to necroptosis during embryonic development. Cell Death Dis. 10, 245 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Dondelinger, Y. et al. Serine 25 phosphorylation inhibits RIPK1 kinase-dependent cell death in models of infection and inflammation. Nat. Commun. 10, 1729 (2019).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  26. Geng, J. et al. Regulation of RIPK1 activation by TAK1-mediated phosphorylation dictates apoptosis and necroptosis. Nat. Commun. 8, 359 (2017).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  27. Stennicke, H. R. & Salvesen, G. S. Catalytic properties of the caspases. Cell Death Differ. 6, 1054–1059 (1999).

    Article  PubMed  CAS  Google Scholar 

  28. Wong, W. W. et al. RIPK1 is not essential for TNFR1-induced activation of NF-κB. Cell Death Differ. 17, 482–487 (2010).

    Article  PubMed  CAS  Google Scholar 

  29. Newton, K. et al. RIPK1 inhibits ZBP1-driven necroptosis during development. Nature 540, 129–133 (2016).

    Article  PubMed  ADS  CAS  Google Scholar 

  30. Cuchet-Lourenço, D. et al. Biallelic RIPK1 mutations in humans cause severe immunodeficiency, arthritis, and intestinal inflammation. Science 361, 810–813 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  31. Micheau, O., Lens, S., Gaide, O., Alevizopoulos, K. & Tschopp, J. NF-kappaB signals induce the expression of c-FLIP. Mol. Cell. Biol. 21, 5299–5305 (2001).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Croft, S. N., Walker, E. J. & Ghildyal, R. Human Rhinovirus 3C protease cleaves RIPK1, concurrent with caspase 8 activation. Sci. Rep. 8, 1569 (2018).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  33. Pearson, J. S. et al. EspL is a bacterial cysteine protease effector that cleaves RHIM proteins to block necroptosis and inflammation. Nat. Microbiol. 2, 16258 (2017).

    Article  PubMed  CAS  Google Scholar 

  34. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  36. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protocols 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  37. Sun, J., Nishiyama, T., Shimizu, K. & Kadota, K. TCC: an R package for comparing tag count data with robust normalization strategies. BMC Bioinformatics 14, 219 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  PubMed  CAS  Google Scholar 

  39. Newton, K., Sun, X. & Dixit, V. M. Kinase RIP3 is dispensable for normal NF-κBs, signaling by the B-cell and T-cell receptors, tumor necrosis factor receptor 1, and Toll-like receptors 2 and 4. Mol. Cell. Biol. 24, 1464–1469 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Murphy, J. M. et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39, 443–453 (2013).

    Article  PubMed  CAS  Google Scholar 

  41. Conos, S. A., Lawlor, K. E., Vaux, D. L., Vince, J. E. & Lindqvist, L. M. Cell death is not essential for caspase-1-mediated interleukin-1β activation and secretion. Cell Death Differ. 23, 1827–1838 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Mandal, P. et al. RIP3 induces apoptosis independent of pronecrotic kinase activity. Mol. Cell 56, 481–495 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Newton, K. et al. Activity of protein kinase RIPK3 determines whether cells die by necroptosis or apoptosis. Science 343, 1357–1360 (2014).

    Article  PubMed  ADS  CAS  Google Scholar 

Download references

Acknowledgements

This study was funded by the Intramural Research Programs of the National Human Genome Research Institute, the Intramural Research Program of NIH, NIH Clinical Center, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institute of Allergy and Infectious Diseases, and National Heart, Lung, and Blood Institute, by European Research Council Advanced Grant 787826, by NHMRC grants 1025594, 1046984, 1145788, 1162765 and 1163581, NHMRC fellowships 1081421 and 1107149, by the Stafford Fox Foundation and was made possible through Victorian State Government Operational Infrastructure Support, Australian Government NHMRC IRIISS (9000433) and Australian Cancer Research Fund. N.L. is supported by project grant 1145588 from the Cancer Australia and Cure Cancer Australia Foundation and a Victorian Cancer Agency Mid-career Fellowship 17030. This work used the sequencing resources at the NIH Intramural Sequencing Center and the computational resources of the Biowulf Linux cluster at NIH (http://biowulf.nih.gov). We thank the families for their participation, D. Follmann for statistical advice, T. Uldrick and D. Fajgenbaum for assistance procuring samples, and D. Adams, A. Negro, A. Walts and Y. Yang for clinical and technical assistance, C. Liegeois for IT assistance and the staff of the WEHI Bioservices facility for mouse husbandry. The generation of Ripk1D325A and Ripk1D138N,D325A mice used in this study was supported by the Australian Phenomics Network (APN) and the Australian Government through the National Collaborative Research Infrastructure Strategy (NCRIS) program.

Author information

Authors and Affiliations

Authors

Contributions

N.L., S.E.B. and H.O. designed and performed experiments and interpreted data. G.M.W., D.C., L.L., M.S., T.K., K.E.L., K.J.M.Z., N.E., K.S.-A., C.B., W.L.T., M.D.B., H.S.K., D.Y., H.A., N.S., L.W., L.Z., N.S.M., D.B.B., G.G.-C., C.H., H.W., J.J.C., N.I.D, M.M., A.L., Q.Z., I.A., J.C.M., A.K.V. and J.S. performed experiments. A.J.K., M.J.H., L.W. and M.P. generated the CRISPR mice. D.L.S., P.M.H., A.K.O., G.P.P.-P., B.K.B., A.J., T.M.R., A.J.G. and A.K.S. provided the clinical data. E.D.H., S.L.M., M.J.L., M.B., S.D.R. and M.G. contributed reagents, analysis and interpretation. N.L., S.E.B., D.L.K. and J.S. conceived the project and wrote the paper with input from all authors.

Corresponding authors

Correspondence to Najoua Lalaoui, Steven E. Boyden, Daniel L. Kastner or John Silke.

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.

Extended data figures and tables

Extended Data Fig. 1 Inflammatory gene signature in P7 whole-blood RNA.

a, MA plot between two P7 samples and two unrelated adolescent healthy controls, both sequenced with technical duplicates. TCC-edgeR package of R followed by adjustment for multiple comparisons detected 1,394 differentially expressed genes (false discovery rate < 0.05), with 903 genes upregulated in P7, and 491 genes downregulated in P7. b, Representative Gene Ontology terms associated with immune signalling.

Extended Data Fig. 2 Exome reads in family 1.

Excerpts of coverage histograms and aligned exome sequence reads for the proband and her parents in family 1, displayed using the integrative genomics viewer, demonstrate de novo occurrence of the c.970G>A (p.D324N) missense mutation in the LXXD caspase-6/8 cleavage motif preceding the cleavage site (arrow). Paternity and maternity were confirmed using Mendelian inheritance error rates from the same exome data.

Extended Data Fig. 3 ‘Kinase-dead’ RIPK1 or combined loss of Ripk3 and Casp8 rescue Ripk1D325A/D325A lethality.

a, b, Observed numbers of offspring from Ripk1D325A/+ intercrosses and numbers expected from Mendelian ratios at the indicated stage of development. Ripk1D325A/D325A mice are E10.5. All observed E11.5 Ripk1D325A/D325A embryos were dead and most of the E10.5 Ripk1D325A/D325A embryos were abnormal, as described in Fig. 2a, b. Loss of Ripk3 rescued to E12.5; however, 50% of the embryos were abnormal. None of the Ripk1D325A/D325ARipk3−/− mice were born. All observed E11.5 Ripk1D325A/D325AMlkl−/− embryos were dead, showing that loss of Mlkl did not provide any protection. All Ripk1D325A/D325ARipk3−/−Casp8−/− mice were born and developed ALPS owing to loss of Casp8. c, Kaplan–Meyer survival curves of the indicated genotypes. d, Cervical lymph nodes (LN), spleen and thymus of 17-week-old mice of the indicated genotypes. Pictures are representative of five mice per genotype. e, Tissue sections of 18-day-old Ripk1D138N,D325A/+, Ripk1D138N,D325A/D138N,D325A and control mice stained with H&E (left) and anti-CC3 (brown; right). Pictures are representative of two mice per genotype.

Extended Data Fig. 4 Ripk1D325A/+ cells are hypersensitive to TNF-induced death.

ac, MDFs (a), BMDMs (b) and MEFs (c) of the indicated genotypes were treated with either a high dose of TNF (T100; 100 ng ml−1) or a low dose of TNF (T; 10 ng ml−1) combined with SMAC mimetic (S; 100 nM), caspase inhibitor (I; 5 µM), RIPK3 inhibitor (R; 1 µM), necrostatin (N; 10 µM), TAK1 inhibitor (TAKi; 100 nM), IKK inhibitor (IKKi; 100 nM), MK2 inhibitor (MK2i; 2 µM) or cycloheximide (1 µg ml−1) for 16 h. Cell death was quantified by propidium iodide uptake and time-lapse imaging every 30–45 min using IncuCyte. Duplicates are shown for each genotype. Graphs are representative of three (MEFs and MDFs) and two (BMDMs) biologically independent cell lines per genotype repeated independently. d, MEFs were treated as in Fig. 3d for 2 h. e, f, MDFs (e) and MEFs (f) were treated as in Fig. 3d for the indicated times. Results in df are representative of two independent experiments. β-Actin was used as a loading control. g, BMDMs were treated with TNF (100 ng ml−1) combined with SMAC mimetic (500 nM) with or without caspase inhibitor (5 µM) for 90 min, and lysates were immunoprecipitated with anti-FADD. Results are representative of two independent experiments. For gel source data, see Supplementary Fig. 2.

Extended Data Fig. 5 Alternative cleavage of RIPK1.

a, MEFs were treated with 10 ng ml−1 TNF combined with 500 nM SMAC mimetic for 2 h. b, Doxycycline-inducible caspase-8-gyrase41, wild-type and mutant mouse RIPK1 constructs or GFP were co-expressed in 293T cells. Cells were treated for 2 h with 1 μg ml−1 doxycycline to induce caspase-8-gyrase expression and then for 2 h with 700 nM coumermycin to dimerize caspase-8-gyrase. Antibody recognizing the N-terminal end of RIPK1 was used. Results are representative of four (a) and two (b) independent experiments. For gel source data, see Supplementary Fig. 2.

Extended Data Fig. 6 RIPK1 cleavage limits inflammation in an NF-κB-independent manner.

a, Serum cytokine levels in wild-type and Ripk1D325A/+ mice treated for 3 h with 50 μg of poly(I:C). Each dot represents a mouse. Data are mean ± s.e.m., n = 3 mice. b, TNF levels in the supernatant (S/N) of two unrelated adolescent controls (Ctl RIPK1+/+) and P7 RIPK1D324Y/+ PBMCs treated for 3 h with 5 µg ml−1 poly(I:C). Data are mean of triplicates. c, Body temperature of mice of the indicated genotypes after injection of 2 mg kg−1 LPS. Each line represent a mouse; n = 5 mice per genotype. d, BMDMs of the indicated genotypes were treated for 24 h with 25 ng ml−1 LPS or with 2.5 μg ml−1 poly(I:C). Cell death was quantified by propidium iodide staining and flow cytometry. Each dot represents a biological repeat. Graph shows mean; n = 1 for Ripk1+/+ and n = 2 for Ripk1D325A/+. e, f, BMDMs (e) and MDFs (f) were treated with 100 ng ml−1 of TNF for the indicated times. Results are representative of two independent experiments. β-Actin was used as a loading control. For gel source data, see Supplementary Fig. 2. g, NF-κB activation in fibroblasts derived from patient skin biopsies was assessed by measuring nuclear translocation of subunit p65. Each dot represents the median of more than 1,000 single-cell measurements of nuclear mean p65 fluorescent intensities for one individual subject. Data are mean ± s.d., n = 4 patients and 4 controls. P values determined by unpaired one-tailed (a) or unpaired two-tailed (g) t-tests.

Extended Data Fig. 7 Proposed model for RIPK1(D325A)-induced cell death.

Left, TNF binding to TNFR1 triggers the formation of complex I, and subsequent ubiquitylation and phosphorylation of RIPK1. These post-translational modifications (PTMs) inhibit the cytotoxic activity of RIPK1. Complex I formation activates NF-κB- and MAPK-dependent survival genes such as CFLAR, which encodes cFLIP. Subsequently, a cytosolic complex II containing FADD, caspase-8, RIPK1 and cFLIP is formed. In this complex, cFLIP inhibits caspase-8 activity so that a restricted number of substrates (such as RIPK1) are cleaved, but others (such as pro-caspase-3) are not. Cleavage of RIPK1 dismantles complex II. Activation of the NF-κB and MAPK signalling pathways PTM of RIPK1 prevent TNF from inducing cell death, resulting in cell survival (top left). Inhibition of the NF-κB or MAPK signalling pathways reduces levels of cFLIP and accelerates formation of complex II, resulting in cell death via apoptosis (middle left). When NF-κB or MAPK signalling is disrupted in caspase-8-deficient conditions, RIPK1 is not cleaved and autophosphorylates, which triggers the recruitment of RIPK3 and its autophosphorylation. RIPK3 phosphorylates MLKL and necroptosis occurs (bottom left). Right, according to this model, lack of RIPK1 cleavage could result in several distinct outcomes, as follows. (1) RIPK1 accumulation could stabilize complex II, and the presence of cFLIP and inhibitory PTMs to RIPK1 may prevent caspase-8 from killing, resulting in cell survival. (2) The accumulation of ‘uncleavable’ RIPK1 to complex II could override the inhibitory RIPK1 PTMs, resulting in autophosphorylation of RIPK1 and recruitment of RIPK3, leading to necroptosis. (3) RIPK1 accumulation could result in activated caspase-8 that cleaves RIPK3, resulting in cell survival. (4) Stabilization of complex II could result in recruitment and activation of caspase-8 that induces apoptosis and possibly prevents necroptosis by cleaving RIPK3. (5) Finally, the accumulation of RIPK1 could result in activation of both RIPK3 and caspase-8 and therefore induce both apoptotic and necroptotic cell death. In terms of how these potential outcomes match with our data, in homozygote Ripk1D325A cells, both caspase-8 and RIPK3 are activated after TNF signalling, which suggests that apoptosis and necroptosis occur at the same time (Figs. 2d, 3a, b). However, according to these models, loss of RIPK3 limits caspase-8 activation (Fig. 3a, b). This suggests that the recruitment of RIPK3 to complex II increases the recruitment and activation of caspase-8. A precedent for this observation comes from experiments in which RIPK3 inhibitors promoted RIPK1-dependent caspase-8 activation42,43, in a manner we term ‘reverse activation’. In our experiments, however, RIPK3 activation occurs downstream of TNF signalling, which suggests that reverse activation might represent a physiological amplification loop that increases caspase-8 activation. Yet, this requirement for RIPK3 is not present in all cells, as the embryonic lethality of the RIPK1-cleavage mutant is only partially rescued by loss of Ripk3. In the heterozygote Ripk1D325A cells, caspase-8 cleaves wild-type RIPK1, thus limiting TNF-induced cell death as compared to homozygote cells. However, reduction of cFLIP and/or RIPK1 PTMs by treatment with IAP, TAK1, IKK or translational inhibitors decreases the threshold of TNF sensitivity (Extended Data Fig. 4). This may cause the hyper-inflammatory response observed in patients with CRIA syndrome (Fig. 1).

Extended Data Table 1 Leukocyte surface markers in patients with CRIA syndrome
Extended Data Table 2 Effect of tocilizumab treatment and RIPK1 caspase cleavage site mutations is absent in known autoinflammatory diseases
Extended Data Table 3 Conservation of RIPK1 caspase-8 cleavage site

Supplementary information

Supplementary Figures

This file contains Supplementary Figures 1-2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lalaoui, N., Boyden, S.E., Oda, H. et al. Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease. Nature 577, 103–108 (2020). https://doi.org/10.1038/s41586-019-1828-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1828-5

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