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:

FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation

Abstract

Cytosolic sensing of pathogens and damage by myeloid and barrier epithelial cells assembles large complexes called inflammasomes, which activate inflammatory caspases to process cytokines (IL-1β) and gasdermin D (GSDMD). Cleaved GSDMD forms membrane pores, leading to cytokine release and inflammatory cell death (pyroptosis). Inhibiting GSDMD is an attractive strategy to curb inflammation. Here we identify disulfiram, a drug for treating alcohol addiction, as an inhibitor of pore formation by GSDMD but not other members of the GSDM family. Disulfiram blocks pyroptosis and cytokine release in cells and lipopolysaccharide-induced septic death in mice. At nanomolar concentration, disulfiram covalently modifies human/mouse Cys191/Cys192 in GSDMD to block pore formation. Disulfiram still allows IL-1β and GSDMD processing, but abrogates pore formation, thereby preventing IL-1β release and pyroptosis. The role of disulfiram in inhibiting GSDMD provides new therapeutic indications for repurposing this safe drug to counteract inflammation, which contributes to many human diseases.

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: High-throughput screen identifies disulfiram as an inhibitor of GSDMD pore formation.
Fig. 2: Disulfiram inhibits pyroptosis and IL-1β secretion.
Fig. 3: Disulfiram inhibition of liposome leakage is mediated primarily by direct inhibition of GSDMD pore formation.
Fig. 4: Disulfiram covalently modifies GSDMD Cys191.
Fig. 5: GSDMD pore formation is the main target of disulfiram.
Fig. 6: Disulfiram protects against LPS-induced sepsis.

Similar content being viewed by others

Data availability

All relevant data are available in the Source Data or Extended Data of the manuscript.

References

  1. Rathinam, V. A., Vanaja, S. K. & Fitzgerald, K. A. Regulation of inflammasome signaling. Nat. Immunol. 13, 332–333 (2012).

    Article  CAS  Google Scholar 

  2. Lamkanfi, M. & Dixit, V. M. Inflammasomes and their roles in health and disease. Annu. Rev. Cell Dev. Biol. 28, 137–161 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Broz, P. & Dixit, V. M. Inflammasomes: mechanism of assembly, regulation and signalling. Nat. Rev. Immunol. 16, 407–420 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Kayagaki, N. et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 (2015).

    Article  CAS  PubMed  Google Scholar 

  5. Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. He, W. T. et al. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res. 25, 1285–1298 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Aglietti, R. A. et al. GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes. Proc. Natl Acad. Sci. USA 113, 7858–7863 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sborgi, L. et al. GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. EMBO J. 35, 1766–1778 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ding, J. et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature 535, 111–116 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Liu, X. et al. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 535, 153–158 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chen, X. et al. Pyroptosis is driven by non-selective gasdermin-D pore and its morphology is different from MLKL channel-mediated necroptosis. Cell Res. 26, 1007–1020 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Russo, H. M. et al. Active caspase-1 induces plasma membrane pores that precede pyroptotic lysis and are blocked by lanthanides. J. Immunol. 197, 1353–1367 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Ruan, J., Xia, S., Liu, X., Lieberman, J. & Wu, H. Cryo-EM structure of the gasdermin A3 membrane pore. Nature 557, 62–67 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Jo, E.-K., Kim, J. K., Shin, D.-M. & Sasakawa, C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell. Mol. Immunol. 13, 148–159 (2016).

    Article  CAS  PubMed  Google Scholar 

  15. Frangogiannis, N. G., Smith, C. W. & Entman, M. L. The inflammatory response in myocardial infarction. Cardiovasc. Res. 53, 31–47 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Hoffman, H. M., Wanderer, A. A. & Broide, D. H. Familial cold autoinflammatory syndrome: phenotype and genotype of an autosomal dominant periodic fever. J. Allergy Clin. Immunol. 108, 615–620 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Muckle, T. J. & Wells, M. Urticaria, deafness, and amyloidosis: a new heredo-familial syndrome. QJM 31, 235–248 (1962).

    CAS  PubMed  Google Scholar 

  18. Prieur, A. M. et al. A chronic, infantile, neurological, cutaneous and articular (CINCA) syndrome. A specific entity analysed in 30 patients. Scand. J. Rheumatol. Suppl. 66, 57–68 (1987).

    Article  CAS  PubMed  Google Scholar 

  19. Hoffman, H. M., Mueller, J. L., Broide, D. H., Wanderer, A. A. & Kolodner, R. D. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle–Wells syndrome. Nat. Genet. 29, 301–305 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Aksentijevich, I. et al. De novo CIAS1 mutations, cytokine activation, and evidence for genetic heterogeneity in patients with neonatal-onset multisystem inflammatory disease (NOMID): a new member of the expanding family of pyrin-associated autoinflammatory diseases. Arthritis Rheum. 46, 3340–3348 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Masters, S. L. et al. Familial autoinflammation with neutrophilic dermatosis reveals a regulatory mechanism of pyrin activation. Sci. Translat. Med. 8, 332ra345 (2016).

    Article  CAS  Google Scholar 

  22. Rello, J., Valenzuela-Sánchez, F., Ruiz-Rodriguez, M. & Moyano, S. Sepsis: a review of advances in management. Adv. Ther. 34, 2393–2411 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Dellinger, R. P. et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock, 2012. Intensive Care Med. 39, 165–228 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wright, C. & Moore, R. D. Disulfiram treatment of alcoholism. Am. J. Med. 88, 647–655 (1990).

    Article  CAS  PubMed  Google Scholar 

  25. Skrott, Z. et al. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature 552, 194–199 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sborgi, L. et al. Structure and assembly of the mouse ASC inflammasome by combined NMR spectroscopy and cryo-electron microscopy. Proc. Natl Acad. Sci. USA 112, 13237–13242 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, J. H., Chung, T. D. & Oldenburg, K. R. A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J. Biomol. Screen 4, 67–73 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Degterev, A. et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 4, 313–321 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Shen, M. L., Johnson, K. L., Mays, D. C., Lipsky, J. J. & Naylor, S. Determination of in vivo adducts of disulfiram with mitochondrial aldehyde dehydrogenase. Biochem. Pharmacol. 61, 537–545 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Petersen, E. N. The pharmacology and toxicology of disulfiram and its metabolites. Acta Psychiatr. Scand. Suppl. 369, 7–13 (1992).

    Article  CAS  PubMed  Google Scholar 

  32. Nobel, C. S., Kimland, M., Nicholson, D. W., Orrenius, S. & Slater, A. F. Disulfiram is a potent inhibitor of proteases of the caspase family. Chem. Res. Toxicol. 10, 1319–1324 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Hagn, F., Nasr, M. L. & Wagner, G. Assembly of phospholipid nanodiscs of controlled size for structural studies of membrane proteins by NMR. Nat. Protoc. 13, 79–98 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Castillo-Villanueva, A. et al. Disulfiram as a novel inactivator of Giardia lamblia triosephosphate isomerase with antigiardial potential. Int. J. Parasitol. Drugs Drug. Resist. 7, 425–432 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Sanchez, R., Riddle, M., Woo, J. & Momand, J. Prediction of reversibly oxidized protein cysteine thiols using protein structure properties. Protein Sci. 17, 473–481 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Rathkey, J. K. et al. Chemical disruption of the pyroptotic pore-forming protein gasdermin D inhibits inflammatory cell death and sepsis. Sci. Immunol. 3, eaat2738 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Juliana, C. et al. Anti-inflammatory compounds parthenolide and Bay 11-7082 are direct inhibitors of the inflammasome. J. Biol. Chem. 285, 9792–9802 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Nair, A. B. & Jacob, S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 7, 27–31 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Kayagaki, N. et al. Non-canonical inflammasome activation targets caspase-11. Nature 479, 117–121 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Sollberger, G. et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci. Immunol. 3, eaar6689 (2018).

    Article  PubMed  Google Scholar 

  41. Cvek, B. Nonprofit drugs as the salvation of the world’s healthcare systems: the case of Antabuse (disulfiram). Drug Discov. Today 17, 409–412 (2012).

    Article  PubMed  Google Scholar 

  42. Nasr, M. L. et al. Covalently circularized nanodiscs for studying membrane proteins and viral entry. Nat. Methods 14, 49–52 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the US National Institutes of Health (grant nos. DP1HD087988 to H.W.; R01Al139914 to H.W. and J.L.; R01AI123265 to J.L.; and R01 AI142642, R01 AI145274, R01 AI141386, R01HL092020 and P01HL095489 to H.R.L.), National Natural Science Foundation of China (grant no. 31972897), Key Research Program of the Chinese Academy of Sciences (grant no. ZDBS-LY-SM008), Shanghai Municipal Science and Technology Major Project (grant no. 2019SHZDZX02), Rising-Star Program of Shanghai Science and Technology Committee (grant no. 19QA1409800 to X.L.), a grant from FAMRI (no. CIA 123008 to H.R.L.), Cancer Research Institute Irvington Postdoctoral Fellowship Program (to J.J.H.), Charles A. King Trust Postdoctoral Fellowship Program (to J.R., X.L., Z.Z.) and a US DOD Breast Cancer Research Program Breakthrough Fellowship Award (Y.Z.). We thank J. Smith, G. Frey, J. Nale, D. Wrobel and the entire staff of the ICCB-L for their outstanding technical support.

Author information

Authors and Affiliations

Authors

Contributions

H.W. and J.J.H. conceived the study. J.J.H., S.X. and J.R. optimized the liposome leakage assay. J.J.H performed the high-throughput screen and the validation experiments in vitro. S.X. performed negative staining electron microscopy. X. Liu, Z.Z., J.Z., X. Lou, Y.B., J.W., L.R.H. and V.G.M. performed cellular experiments. X. Liu, Y.Z., L.Z. and H.R.L. carried out studies in mice. X. Luo ran mass spectrometry. J.K. advised on chemistry. H.W. and J.L. supervised the project. H.W., J.J.H., J.L. and X. Liu wrote the manuscript with input from all authors.

Corresponding authors

Correspondence to Xing Liu, Judy Lieberman or Hao Wu.

Ethics declarations

Competing interests

J.L. and H.W. are cofounders of Ventus Therapeutics. The other 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

Extended Data Fig. 1 Optimization and hits from the liposome leakage assay screen.

(a–c) Optimization of the Tb3+/DPA assay. a, GSDMD (2.5 μM) and caspase-11 (2.5 μM) were incubated in liposome solutions at various concentrations in 20 mM HEPES buffer (150 mM NaCl) for 1 h. The concentration of liposome lipids for the screen was set at 50 μM. n = 3 independent experiments. The mean ± s.e.m. is shown. b, Different concentrations of GSDMD and caspase-11 (1:1 ratio) were incubated in liposome (50 μM) solutions for 1 h. The concentration of GSDMD used in the screen was set at 0.3 μM. n = 3 independent experiments. The mean ± s.e.m. is shown. c, Different concentrations of caspase-11 and GSDMD (0.3 μM) were incubated in liposome (50 μM) solutions for 1 h. The concentration of caspase-11 used in the screen was set at 0.15 μM. n = 3 independent experiments. The mean ± s.e.m. is shown. The fluorescence intensity at 545 nm was measured after excitation at 276 nm. d, Hit compounds evaluated in binding and/or cell-based assays. (e) Mouse iBMDMs were pretreated or not with disulfiram (C-23) ranging from 5-40 μM for 1 h before transfection with PBS or poly(dA:dT) and analyzed for cell viability by CellTiter-Glo assay 4 hrs later. Graphs show mean ± s.d; data are representative of three independent experiments with replicates (n = 3) and similar results. Data were analyzed using two-tailed Student’s t-test. **P < 0.01.

Source data

Extended Data Fig. 2 The activity of disulfiram in cells is greatly increased by Cu(II).

a, DTC–copper complex formation of disulfiram metabolite diethyldithiocarbamate (DTC) with Cu(II). b, Dose response curves of inhibition of liposome leakage by disulfiram (C-23) or DTC in the presence or absence of Cu(II). n = 3 independent experiments. The mean ± s.e.m. is shown. c, LPS-primed THP-1 were pretreated with C-23 or DTC in the presence or absence of Cu(II) for 1 h before adding nigericin or medium for 2 hrs. Cell death was determined by CytoTox96 assay. n = 3 independent experiments. The mean ± s.e.m. is shown.

Source data

Extended Data Fig. 3 Effect of disulfiram on caspase-1 and caspase-11.

(a, b) Time course of caspase-1 (a) and caspase-11 (b) activity in the presence of indicated concentrations of disulfiram. Caspases (0.5 U) were incubated with disulfiram (at indicated concentrations for 1 h before adding Ac-YVAD-AMC (40 μM)). (c,d) Dose response curve of disulfiram in the caspase-1 (a) and caspase-11 (b) activity assay. (e,f) Time course of caspase-1 (e) and caspase-11 (f) activity in the presence of indicated concentrations of disulfiram + Cu(II). Caspases (0.5 U) were incubated with disulfiram + Cu(II) (at indicated concentrations for 1 h before adding Ac-YVAD-AMC (40 μM)). (g,h) Dose response curve of disulfiram + Cu(II) in the caspase-1 (e) and caspase-11 (f) activity assay. (a-h) n = 3 independent experiments. The mean ± s.e.m. is shown. Fluorescence intensity at 460 nm was measured after excitation at 350 nm.

Source data

Extended Data Fig. 4 Disulfiram covalently modifies human GSDMD on Cys 191.

a, Disulfiram was preincubated for 1 h with N-acetylcysteine (NAC, 500 μM) or medium before evaluating whether it inhibited pyroptosis of LPS + nigericin treated THP-1 cells. Disulfiram 2-fold dilutions ranged from 5-40 μM. Graphs show mean ± s.d; data are representative of three independent experiments with replicates (n = 3) and similar results. Data were analyzed using two-tailed Student’s t-test. Graphs show the mean ± s.d. and data shown are representative of three independent experiments. **P < 0.01. (b, c) nano-LC-MS/MS spectrum for the peptide containing C191 in human GSDMD. Data are representative of three independent experiments. b, MS/MS spectrum for peptide FSLPGATCLQGEGQGHLSQK modified on cysteine (red) by carbamidomethyl. Protein coverage was 73%. c, MS/MS spectrum for peptide FSLPGATCLQGEGQGHLSQK modified on cysteine (red) by disulfiram. Protein coverage was 72%.

Source data

Extended Data Fig. 5 Disulfiram covalently modifies GSDMD Cys191.

a, Sequence alignment of GSDMA3, hGSDMA, mGSDMD and hGSDMD showing Cys residues (highlighted in red). b, GSDMD (0.3 μM) was preincubated with the indicated concentrations of disulfiram (0–5.6 μM) for indicated times (2–90 min) before caspase-11 (0.15 μM) and liposomes (50 μM) were added. n = 3 independent experiments. The mean ± s.e.m. is shown. c, FL mouse GSDMD or wildtype, C192S or C39A GSDMD-NT were transiently expressed in HEK293T cells. Cell death was determined by CytoTox96 cytotoxicity assay 20 hrs after transfection. c, shows the mean ± s.d. of 1 representative experiment of three independent experiments performed. Comparison in (c) was calculated by two-tailed Student’s t-test. *P < 0.05.

Source data

Extended Data Fig. 6 Mouse monoclonal antibody recognizes full-length human GSDMD and the GSDMD-NT pore form on immunoblots and by immunofluorescence microscopy.

The monoclonal antibody against GSDMD was generated by immunizing mice with recombinant human GSDMD and boosting with recombinant human GSDMD-NT as described in Methods. a, HEK293T cells were transfected with the indicated plasmids and cell lysates were analysed by immunoblot of reducing gels probed with the indicated antibodies. b, Cell lysates of HCT116, 293 T and THP-1 cells, treated or not with nigericin, were immunoblotted with the indicated antibodies. 293 T cells do not express endogenous GSDMD. c, 293T and THP-1 cells were stained with the anti-GSDMD monoclonal antibody and co-stained with DAPI (blue). 293T cells show no background staining. Data are representative of at least three independent experiments.

Source data

Extended Data Fig. 7 Disulfiram protects against LPS-induced sepsis.

(a–c) Mice were pretreated with disulfiram (50 mg/kg) or vehicle (Ctrl) by intraperitoneal injection 24 and 4 hrs before intraperitoneal challenge with 15 mg/kg LPS and followed for survival. Serum IL-1β (a), TNF (b) and IL-6 (c) were measured by Luminex Multiplex Assay (n = 5/group) 6 hrs post LPS challenge. Shown are mean ± s.e.m. Statistical differences between the groups were calculated by multiple t-test. Type I error was corrected by the Holm-Sidak method.

Source data

Extended Data Fig. 8 Dose response curve of other compounds in GSDMD-mediated liposome leakage assay.

Dose response curve of necrosulfonamide (a), Bay 11-7082 (b), dimethyl fumarate (DMF) (c), afatinib (d), ibrutinib (e), and LDC7559 (f) in liposome leakage induced by 0.3 μM GSDMD plus 0.15 μM caspase-11. (a-f) n = 3 independent experiments. The mean ± s.e.m. is shown.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical Source Data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data

Source Data Extended Data Fig. 2

Statistical Source Data

Source Data Extended Data Fig. 3

Statistical Source Data

Source Data Extended Data Fig. 4

Statistical Source Data

Source Data Extended Data Fig. 5

Statistical Source Data

Source Data Extended Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 8

Statistical Source Data

Source Data Fig. 3

Unprocessed gels

Source Data Fig. 5

Unprocessed western blots

Source Data Fig. 7

Unprocessed western blots

Source Data Extended Data Fig. 6

Unprocessed western blots

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, J.J., Liu, X., Xia, S. et al. FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat Immunol 21, 736–745 (2020). https://doi.org/10.1038/s41590-020-0669-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0669-6

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