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:

Biallelic human SHARPIN loss of function induces autoinflammation and immunodeficiency

Abstract

The linear ubiquitin assembly complex (LUBAC) consists of HOIP, HOIL-1 and SHARPIN and is essential for proper immune responses. Individuals with HOIP and HOIL-1 deficiencies present with severe immunodeficiency, autoinflammation and glycogen storage disease. In mice, the loss of Sharpin leads to severe dermatitis due to excessive keratinocyte cell death. Here, we report two individuals with SHARPIN deficiency who manifest autoinflammatory symptoms but unexpectedly no dermatological problems. Fibroblasts and B cells from these individuals showed attenuated canonical NF-κB responses and a propensity for cell death mediated by TNF superfamily members. Both SHARPIN-deficient and HOIP-deficient individuals showed a substantial reduction of secondary lymphoid germinal center B cell development. Treatment of one SHARPIN-deficient individual with anti-TNF therapies led to complete clinical and transcriptomic resolution of autoinflammation. These findings underscore the critical function of the LUBAC as a gatekeeper for cell death-mediated immune dysregulation in humans.

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: Human SHARPIN deficiency causes autoinflammation and hepatic glycogenosis.
Fig. 2: Human SHARPIN deficiency impairs canonical NF-κB-mediated signaling.
Fig. 3: Human LUBAC deficiencies trigger excessive TNF-induced cell death.
Fig. 4: Loss of SHARPIN causes joint inflammation in humans and mice.
Fig. 5: Human LUBAC deficiencies cause defective GC formation.
Fig. 6: Human LUBAC deficiencies cause dysregulation in B cell activation and death.
Fig. 7: TNF inhibitors resolve systemic inflammation associated with SHARPIN deficiency.

Similar content being viewed by others

Data availability

RNA sequence data have been deposited in the Gene Expression Omnibus under the accession code GSE261031. Exome sequencing data will not be made publicly available as they contain information that might compromise research participant privacy/consent. Source data are provided with this paper.

References

  1. Iwai, K., Fujita, H. & Sasaki, Y. Linear ubiquitin chains: NF-κB signalling, cell death and beyond. Nat. Rev. Mol. Cell Biol. 15, 503–508 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Fuseya, Y. et al. The HOIL-1L ligase modulates immune signalling and cell death via monoubiquitination of LUBAC. Nat. Cell Biol. 22, 663–673 (2020).

    Article  CAS  PubMed  Google Scholar 

  3. Kelsall, I. R., Zhang, J., Knebel, A., Arthur, J. S. C. & Cohen, P. The E3 ligase HOIL-1 catalyses ester bond formation between ubiquitin and components of the Myddosome in mammalian cells. Proc. Natl Acad. Sci. USA 116, 13293–13298 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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  PubMed  PubMed Central  Google Scholar 

  6. Oda, H. et al. Second case of HOIP deficiency expands clinical features and defines inflammatory transcriptome regulated by LUBAC. Front. Immunol. 10, 479 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. HogenEsch, H. et al. A spontaneous mutation characterized by chronic proliferative dermatitis in C57BL mice. Am. J. Pathol. 143, 972–982 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Gerlach, B. et al. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature 471, 591–596 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Ikeda, F. et al. SHARPIN forms a linear ubiquitin ligase complex regulating NF-κB activity and apoptosis. Nature 471, 637–641 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kumari, S. et al. SHARPIN prevents skin inflammation by inhibiting TNFR1-induced keratinocyte apoptosis. eLife 3, e03422 (2014).

  13. Rickard, J. A. et al. TNFR1-dependent cell death drives inflammation in SHARPIN-deficient mice. eLife 3, e03464 (2014).

  14. Tokunaga, F. et al. SHARPIN is a component of the NF-κB-activating linear ubiquitin chain assembly complex. Nature 471, 633–636 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sasaki, Y. et al. Defective immune responses in mice lacking LUBAC-mediated linear ubiquitination in B cells. EMBO J. 32, 2463–2476 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. 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  PubMed  PubMed Central  Google Scholar 

  18. Vince, J. E. et al. IAP antagonists target cIAP1 to induce TNFα-dependent apoptosis. Cell 131, 682–693 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. de Almagro, M. C., Goncharov, T., Newton, K. & Vucic, D. Cellular IAP proteins and LUBAC differentially regulate necrosome-associated RIP1 ubiquitination. Cell Death Dis. 6, e1800 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Laurien, L. et al. Autophosphorylation at serine 166 regulates RIP kinase 1-mediated cell death and inflammation. Nat. Commun. 11, 1747 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kay, J. & Calabrese, L. The role of interleukin-1 in the pathogenesis of rheumatoid arthritis. Rheumatology 43, iii2–iii9 (2004).

    Article  PubMed  Google Scholar 

  22. Wijbrandts, C. A. et al. The clinical response to infliximab in rheumatoid arthritis is in part dependent on pretreatment tumour necrosis factor alpha expression in the synovium. Ann. Rheum. Dis. 67, 1139–1144 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Sundberg, J. P. et al. Keratinocyte-specific deletion of SHARPIN induces atopic dermatitis-like inflammation in mice. PLoS ONE 15, e0235295 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang, J. et al. LUBAC suppresses IL-21-induced apoptosis in CD40-activated murine B cells and promotes germinal center B cell survival and the T-dependent antibody response. Front. Immunol. 12, 658048 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Roco, J. A. et al. Class-switch recombination occurs infrequently in germinal centers. Immunity 51, 337–350 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Elsner, R. A. & Shlomchik, M. J. Germinal center and extrafollicular B cell responses in vaccination, immunity, and autoimmunity. Immunity 53, 1136–1150 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Teh, C. E. et al. Linear ubiquitin chain assembly complex coordinates late thymic T-cell differentiation and regulatory T-cell homeostasis. Nat. Commun. 7, 13353 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Park, Y. et al. SHARPIN controls regulatory T cells by negatively modulating the T cell antigen receptor complex. Nat. Immunol. 17, 286–296 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. HogenEsch, H. et al. Increased expression of type 2 cytokines in chronic proliferative dermatitis (cpdm) mutant mice and resolution of inflammation following treatment with IL-12. Eur. J. Immunol. 31, 734–742 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. van Zelm, M. C. et al. Human CD19 and CD40L deficiencies impair antibody selection and differentially affect somatic hypermutation. J. Allergy Clin. Immunol. 134, 135–144 (2014).

    Article  PubMed  Google Scholar 

  31. Meyers, G. et al. Activation-induced cytidine deaminase (AID) is required for B-cell tolerance in humans. Proc. Natl Acad. Sci. USA 108, 11554–11559 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. McGowan, H. W. et al. Sharpin is a key regulator of skeletal homeostasis in a TNF-dependent manner. J. Musculoskelet. Neuronal Interact. 14, 454–463 (2014).

    CAS  PubMed  Google Scholar 

  33. Kim, H. et al. Development of a validated interferon score using NanoString technology. J. Interferon Cytokine Res. 38, 171–185 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Panayotova-Dimitrova, D. et al. cFLIP regulates skin homeostasis and protects against TNF-induced keratinocyte apoptosis. Cell Rep. 5, 397–408 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Weinlich, R. et al. Protective roles for caspase-8 and cFLIP in adult homeostasis. Cell Rep. 5, 340–348 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Orning, P. et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science 362, 1064–1069 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sarhan, J. et al. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc. Natl Acad. Sci. USA 115, E10888–E10897 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Gurung, P., Lamkanfi, M. & Kanneganti, T. D. Cutting edge: SHARPIN is required for optimal NLRP3 inflammasome activation. J. Immunol. 194, 2064–2067 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Douglas, T., Champagne, C., Morizot, A., Lapointe, J. M. & Saleh, M. The inflammatory caspases-1 and -11 mediate the pathogenesis of dermatitis in SHARPIN-deficient mice. J. Immunol. 195, 2365–2373 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Gurung, P., Sharma, B. R. & Kanneganti, T. D. Distinct role of IL-1β in instigating disease in Sharpin(cpdm) mice. Sci. Rep. 6, 36634 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Anderton, H. et al. Langerhans cells are an essential cellular intermediary in chronic dermatitis. Cell Rep. 39, 110922 (2022).

    Article  CAS  PubMed  Google Scholar 

  42. Anderton, H., Wicks, I. P. & Silke, J. Cell death in chronic inflammation: breaking the cycle to treat rheumatic disease. Nat. Rev. Rheumatol. 16, 496–513 (2020).

    Article  PubMed  Google Scholar 

  43. van Loo, G. & Bertrand, M. J. M. Death by TNF: a road to inflammation. Nat. Rev. Immunol. 23, 289–303 (2023).

    Article  PubMed  Google Scholar 

  44. Pasparakis, M. & Vandenabeele, P. Necroptosis and its role in inflammation. Nature 517, 311–320 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Mifflin, L., Ofengeim, D. & Yuan, J. Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target. Nat. Rev. Drug Discov. 19, 553–571 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Weisel, K. et al. A randomized, placebo-controlled experimental medicine study of RIPK1 inhibitor GSK2982772 in patients with moderate to severe rheumatoid arthritis. Arthritis Res. Ther. 23, 85 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Weisel, K. et al. A randomised, placebo-controlled study of RIPK1 inhibitor GSK2982772 in patients with active ulcerative colitis. BMJ Open Gastroenterol. 8, e000680 (2021).

  48. Lalaoui, N. et al. Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease. Nature 577, 103–108 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Tao, P. et al. A dominant autoinflammatory disease caused by non-cleavable variants of RIPK1. Nature 577, 109–114 (2020).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Li, Y. et al. Human RIPK1 deficiency causes combined immunodeficiency and inflammatory bowel diseases. Proc. Natl Acad. Sci. USA 116, 970–975 (2019).

    Article  CAS  PubMed  Google Scholar 

  52. Taft, J. et al. Human TBK1 deficiency leads to autoinflammation driven by TNF-induced cell death. Cell 184, 4447–4463 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Badran, Y. R. et al. Human RELA haploinsufficiency results in autosomal-dominant chronic mucocutaneous ulceration. J. Exp. Med. 214, 1937–1947 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Damgaard, R. B. et al. OTULIN deficiency in ORAS causes cell type-specific LUBAC degradation, dysregulated TNF signalling and cell death. EMBO Mol. Med. 11, e9324 (2019).

  55. Zinngrebe, J. et al. LUBAC deficiency perturbs TLR3 signaling to cause immunodeficiency and autoinflammation. J. Exp. Med. 213, 2671–2689 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kelsall, I. R. et al. HOIL-1 ubiquitin ligase activity targets unbranched glucosaccharides and is required to prevent polyglucosan accumulation. EMBO J. 41, e109700 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Otten, E. G. et al. Ubiquitylation of lipopolysaccharide by RNF213 during bacterial infection. Nature 594, 111–116 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Matsumoto, M. L. et al. Engineering and structural characterization of a linear polyubiquitin-specific antibody. J. Mol. Biol. 418, 134–144 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Zinngrebe, J. et al. Compound heterozygous variants in OTULIN are associated with fulminant atypical late-onset ORAS. EMBO Mol. Med. 14, e14901 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Samson, A. L. et al. A toolbox for imaging RIPK1, RIPK3, and MLKL in mouse and human cells. Cell Death Differ. 28, 2126–2144 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wang, K. et al. Structural mechanism for GSDMD targeting by autoprocessed caspases in pyroptosis. Cell 180, 941–955 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the participants and their family members and the healthy donors for their enthusiastic support during this research study and M. Pasparakis, H. Kashkar and A. Annibaldi (Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases and Center for Molecular Medicine Cologne, University of Cologne, Germany) for logistic support and valuable suggestions. We thank the The Walter and Eliza Hall Institute Histology Core Facility for their support and assistance in this work. This study was supported, in part, by the Intramural Research Programs of the National Human Genome Research Institute, the National Institute of Arthritis and Musculoskeletal and Skin Diseases, the National Institute of Allergy and Infectious Diseases, the National Cancer Institute, the National Heart, Lung, and Blood Institute, the Clinical Center of the NIH and the Peter MacCallum Cancer Foundation. H.O. was supported by the Deutsche Forschungsgemeinschaft (DFG; German Research Foundation), including CRC1403 (414786233) and Germany’s Excellence Strategy (EXC 2030 (390661388)) and by Fritz-Thyssen Stiftung (10.23.1.013MN). K.E.L. is supported by an Australian Research Council Future Fellowship (FT190100266). N.L., J. Silke and H.A. are supported by National Health and Medical Research Council Leadership Investigator Grants (2017929, 1195038 and 1194144). This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government National Health and Medical Research Council IRIISS (GNT9000719). S.B. is supported by France’s National Research Agency, the Investment for the Future Program (ANR-11-LABX-0070_TRANSPLANTEX) as well as Strasbourg’s Interdisciplinary Thematic Institute for Precision Medicine, CNRS and INSERM, funded by IdEx Unistra (ANR-10-IDEX-0002) and SFRI-STRAT’US (ANR-20-SFRI-0012). H.W. is supported by Cancer Research UK (A27323), a Wellcome Trust Investigator Award (214342/Z/18/Z), the Medical Research Council (MR/S00811X/1), the DFG (CRC1399, 413326622; CRC1530, 455784452; CRC1403, 414786233), an Alexander von Humboldt Foundation Professorship Award and CANTAR. The Laboratory of Human Genetics of Infectious Diseases is supported by the Howard Hughes Medical Institute, the Rockefeller University, the St. Giles Foundation, the NIH (R21AI159728 to B.B. & P01AI061093 to J.L.C.), the National Center for Advancing Translational Sciences (NCATS), NIH Clinical and Translational Science Award (CTSA) program (UL1TR001866), the French National Research Agency (ANR) under the ‘Investments for the Future’ program (ANR-10-IAHU-01), the Integrative Biology of Emerging Infectious Diseases Laboratory of Excellence (ANR-10-LABX-62-IBEID), the French Foundation for Medical Research (FRM) (EQU201903007798), the Square Foundation, Grandir – Fonds de solidarité pour l’enfance, William E. Ford, General Atlantic’s Chairman and Chief Executive Officer, Gabriel Caillaux, General Atlantic’s Co-President, Managing Director and Head of Business in EMEA, and the General Atlantic Foundation, Institut National de la Santé et de la Recherche Médicale (INSERM) and the University of Paris Cité.

Author information

Authors and Affiliations

Authors

Contributions

H.O., N.L., I.A. and D.L.K. conceived and designed the study, analyzed the data and wrote the manuscript. K.M., P.P.C., E.R., O.V., O.K., C.R., S.N., H.S.K., M. Swart, Y.W., N.I.Ç., A.M., R.C., Q.X., S.P., D.B.B., J.J.C., K.D., C.L.S., H.A., K.E.L., H.Y., D.Y., M.B., D.R., W.L.T., M.G. and J.T. performed experiments. B.M., J.M., J. Stoddard and J.N. analyzed and interpreted the results. K.M., M.N., A.K.O., P.H., T.R., N.T.D., H.K., V.Z., N.M., M. Shahrooei, N. Parvaneh, N.A.-O., R.C., J.D., P.M., M.J.K., B.B., J.-L.C., S.B. and A.P.R. developed the human materials and/or recruited participants. C.T.M., K.I., S.D.R., L.D.N., J. Silke, P.L.S., N. Peltzer and H.W. provided critical scientific input and/or reagents. H.O. and N.L. wrote the initial draft of the paper. All authors contributed to the final review and editing of the paper.

Corresponding authors

Correspondence to Hirotsugu Oda, Najoua Lalaoui, Ivona Aksentijevich or Daniel L. Kastner.

Ethics declarations

Competing interests

S.P. is currently an employee of AstraZeneca and may own stock or stock options. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Geert van Loo and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: N. Bernard, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Genetic and molecular investigations of sharpenia.

(a) Sanger sequence electropherograms demonstrating homozygous frameshift variants in the patients. (b) Population allele frequency and CADD score for SHARPIN variants homozygous in public databases. The two SHARPIN variants appear in red. CADDMutation Significance Score (MSC) cutoff for SHARPIN (90% confidence interval) was indicated by dashed line. NR: not reported. (c, d) Normalized mRNA levels of SHARPIN in (c) PBMCs and (d) fibroblasts from LUBAC-deficient patients and healthy controls. RNA was extracted from each sample and was measured with technical quadruplicates. Mean value is displayed as a bar. (e, f) Supporting data for Fig. 2e. TNFR1-signaling complex (TNFR1-SC) formation in fibroblasts from P1 and two unrelated healthy controls. Fibroblasts were stimulated with modified tandem affinity purification (moTAP)-tagged TNF (1 μg/ml) for the indicated times. TNFR1-SC was purified with anti-FLAG immunoprecipitation, and analyzed by western blotting. (g) Normal induction of non-canonical NF-κB in P1. Total PBMCs were stimulated with anti-CD3 (aCD3) for the indicated durations, and the expression of NFKB2 p100 (full length) and p52 (active form) was detected by western blot. Sis: P1’s sister carrying a heterozygous p.Leu74ProfsX86 variant. Representative result of two independent experiments.

Source data

Extended Data Fig. 2 Cytokine expression studies ex vivo in LUBAC-deficient patients.

(a) Cytokine expression in LUBAC-deficient monocytes. PBMCs from SHARPIN (P1)- and HOIP-deficient patients and two healthy controls were stimulated with IL-1β (10 ng/ml) for 6 h, and the intracellular accumulation of cytokines in CD14+ monocytes was quantified by flow cytometry. (b, c) Cytokine secretion from LUBAC-deficient PBMCs. PBMCs from SHARPIN and HOIP-deficient patients and two healthy controls were stimulated with (b) LPS (1 μg/ml) or (c) IL-1β (10 ng/ml) for 6 h, and secreted cytokines were measured by ELISA. (d, e) Cytokine secretion from LUBAC-deficient fibroblasts. Fibroblasts from SHARPIN (P1)- and HOIP-deficient patients and two healthy controls were stimulated with (d) LPS (1 μg/ml) or (e) IL-1β (10 ng/ml) for 24 h, and secreted cytokines were measured by ELISA. (a-e) The experiments were performed with biological triplicates (a,d,e) or duplicates (b,c), and shown are the representatives of two independent experiments. Mean values ± s.d are displayed.

Extended Data Fig. 3 Cell death induction assays.

(a) Cell death assay using immortalized mouse embryonic fibroblasts from Sharpin-deficient mice stably reconstituted with wild-type SHARPIN or patient-derived SHARPIN mutants (P1 and P2). (b, c) Cell death assay using fibroblasts from a SHARPIN-deficient patient (P1), patients with HOIL1 deficiency, otulipenia and cleavage resistant RIPK1-induced autoinflammation (CRIA), and two unrelated healthy controls. The cells were stimulated with TNF (100 ng/ml) combined with (b) smac mimetic (SM: compound A: 100 nM) or (c) human recombinant TWEAK (50 ng/ml), in the presence or absence of zVAD (pan-caspase inhibitor: 20 μM) or Nec1 (RIPK1 inhibitor: 50 μM). The dead cell percentages after 16 h of treatment are shown. (a-c) The experiments were performed with biological triplicates (a, b) or duplicates (c), and shown are the representatives of two (a, c) or five (b) independent experiments. Mean values ± s.d are displayed. Quantitative data were analyzed using one-way ANOVA followed by Tukey-Kramer test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001, N.S., not significant. (d) Supporting data for Fig. 3c. Western blot analysis of caspase-3 cleavage in cell lysates from fibroblasts from a SHARPIN-deficient patient and two unrelated healthy controls. The cells were stimulated with TNF (100 ng/ml) and cycloheximide (CHX 50 μg/ml) for the indicated times. (e, f) Necroptosis induction assay using EBV-immortalized lymphoblastoid cells (e) and fibroblasts (f) from SHARPIN-deficient patient. The fibroblasts were stimulated with TSZ (TNF, smac mimetic (compound A) and zVAD) for the indicated time. HT29 cells were used as a positive control for the phospho-antibodies. Representative result of two independent experiments. (g) Supporting data for Fig. 3d. Complex II immunoprecipitation in fibroblasts from SHARPIN-deficienct P1 compared with an unrelated healthy control. Fibroblasts were stimulated with TNF + zVAD for the indicated times, and the lysates were subjected to immunoprecipitation.

Source data

Extended Data Fig. 4 In vivo and in vitro characterization of cell death in human LUBAC deficiency.

(a) Hematoxylin and eosin (H&E) staining of colon biopsy samples from LUBAC-deficient patients. Bars: 0.4 mm. These images are representative of three biopsy specimens per donor. (b, c) Supporting data for Fig. 3g to validate the specificity of p-RIPK1 antibody to detect RIPK1 Ser166 phosphorylation with (b) western blot and (c) immunocytochemistry. HT29 cells were stimulated with TSZ (TNF + smac mimetic (BV6) + zVAD) for 4 h. Note that TSZ-stimulated cells show positive staining of pRIPK1, which was removed by λ-phosphatase treatment. (d, e) Supporting data for Fig. 3h to validate the antibody specificity for cleaved GSDMD (Asp275) with (d) western blot and (e) immunocytochemistry. THP1 cells were pre-incubated with LPS for 3 h and were further stimulated with nigericin for another 1 h. (b-e) These experiments were aimed to confirm the specificity of the antibodies and were not repeated. (f) Cleavage of GSDME in dermal fibroblasts stimulated with TNF (100 ng/ml) + CHX (50 μg/ml) for the indicated times. HeLa cells were used as a positive control. Representative result of two independent experiments.

Source data

Extended Data Fig. 5 Characterization of joint inflammation in SHARPIN deficiency.

(a) Multiplex ELISA measurement of chemokines in the sterile synovial fluid from P1 before the initiation of anti-TNF treatment, compared with osteoarthritis (OA) control donors (N = 7). The samples were measured in technical triplicate (P1) or duplicate (OA), respectively. Mean values ± s.d are displayed. (b) Quantification of CD45 positive cells in tendons of shoulder joints of control and Sharpin-deficient mice (n = 4 for each group). Data are represented as mean values + SEM. Significance calculated with a two-tailed Mann-Whitney test. (c) Representative hematoxylin and eosin (H&E) staining sections of elbow joints from Sharpin-deficient mice (N = 2) and wild-type littermate controls (N = 2). The arrowhead indicates an inflamed ligament.

Extended Data Fig. 6 Characterization of secondary lymphoid organs in LUBAC deficiencies.

(a, b) Aberrant formation of lymphoid follicles and paracortex in secondary lymphoid organs from LUBAC deficient patients. (a) Lymph node histology of a HOIP-deficient patient compared with a control specimen from an unrelated donor. (b) Adenoid histology of SHARPIN-deficient P1 compared with a control specimen from an unrelated donor. The immunohistochemistry staining was not repeated due to the limited clinical specimens. (c) Gating strategy for the adenoid spectral flow cytometry analysis.

Extended Data Fig. 7 Spectral flow cytometry analysis of human adenoids.

(ae) Adenoid single-cell suspensions from SHARPIN-deficient P1 and 10 unrelated pediatric control donors were analyzed. (a) Quantification of CD3+, CD4+, CD8+ and CD20+ populations in adenoid samples. (b, c) Surface immunoglobulin expression in the (b) germinal center B and (c) memory B populations. (d, e) Quantification of T cell subpopulations in the adenoid samples. Mean values ± s.d are displayed.

Extended Data Fig. 8 Normal T cell phenotyping results in the SHARPIN-deficient patient ex vivo.

(a) T cell proliferation assay. PMBCs were incubated with Cell Trace Violet, stimulated with anti-CD3/28 or PHA for 72 h and analyzed by flow cytometer. (b) Intracellular cytokine staining for Th1, Th2 and Th17 populations. PBMCs were stimulated with PMA (100 ng/ml) and ionomycin (1 μM) for 5 h with Brefeldin A. Stimulated cells were surface stained, fixed and permeabilized with BD Cytofix/Cytoperm kit. Cells were further stained for intracellular cytokines and analyzed by flow cytometry. Ctrl: unrelated healthy control, Sister: sister carrying the heterozygous frameshift SHARPIN variant p.Leu74ProfsX86. Representative result of two independent experiments.

Extended Data Fig. 9 Whole blood RNA sequencing.

(a, b) mRNA expression of selected cytokines (a) and chemokines (b) in the pre- and post-anti-TNF treatment P1 whole blood RNA samples as well as four age-matched healthy controls. (c) A heatmap demonstrating the changes of genes representative for type I interferon-stimulated gene signature in pre- and post-treatment samples from the SHARPIN-deficient P1.

Supplementary information

Source data

Source Data

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Oda, H., Manthiram, K., Chavan, P.P. et al. Biallelic human SHARPIN loss of function induces autoinflammation and immunodeficiency. Nat Immunol (2024). https://doi.org/10.1038/s41590-024-01817-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41590-024-01817-w

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