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:

M24B aminopeptidase inhibitors selectively activate the CARD8 inflammasome

Abstract

Inflammasomes are multiprotein complexes that sense intracellular danger signals and induce pyroptosis. CARD8 and NLRP1 are related inflammasomes that are repressed by the enzymatic activities and protein structures of the dipeptidyl peptidases 8 and 9 (DPP8/9). Potent DPP8/9 inhibitors such as Val-boroPro (VbP) activate both NLRP1 and CARD8, but chemical probes that selectively activate only one have not been identified. Here we report a small molecule called CQ31 that selectively activates CARD8. CQ31 inhibits the M24B aminopeptidases prolidase (PEPD) and Xaa-Pro aminopeptidase 1 (XPNPEP1), leading to the accumulation of proline-containing peptides that inhibit DPP8/9 and thereby activate CARD8. NLRP1 is distinct from CARD8 in that it directly contacts DPP8/9’s active site; these proline-containing peptides, unlike VbP, do not disrupt this repressive interaction and thus do not activate NLRP1. We expect that CQ31 will now become a valuable tool to study CARD8 biology.

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: Esterified Xaa-Pro dipeptides selectively activate the CARD8 inflammasome.
Fig. 2: CQ04 and CQ31 inhibit PEPD.
Fig. 3: CQ31 selectively activates the CARD8 inflammasome.
Fig. 4: CQ31 indirectly inhibits DPP8/9 activity.
Fig. 5: CQ31 engages a target in addition to PEPD.
Fig. 6: Dual inhibition of XPNPEP1 and PEPD activates CARD8.

Similar content being viewed by others

Data availability

The proteomics dataset is available in the PRIDE database: accession code, PXD027707; project name, ‘Target identification of CQ31, a selective activator of the CARD8 inflammasome’ (https://www.ebi.ac.uk/pride/archive/projects/PXD027707). All other data in this study are available within the paper, the Supplementary Information and the Extended Data, and/or from the corresponding author upon reasonable request. Source data are provided with this paper.

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Lamkanfi, M. & Dixit, V. M. Mechanisms and functions of inflammasomes. Cell 157, 1013–1022 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Rathinam, V. A. & Fitzgerald, K. A. Inflammasome complexes: emerging mechanisms and effector functions. Cell 165, 792–800 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. D’Osualdo, A. et al. CARD8 and NLRP1 undergo autoproteolytic processing through a ZU5-like domain. PLoS ONE 6, e27396 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Finger, J. N. et al. Autolytic proteolysis within the function to find domain (FIIND) is required for NLRP1 inflammasome activity. J. Biol. Chem. 287, 25030–25037 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Frew, B. C., Joag, V. R. & Mogridge, J. Proteolytic processing of Nlrp1b is required for inflammasome activity. PLoS Pathog. 8, e1002659 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chui, A. J. et al. N-terminal degradation activates the NLRP1B inflammasome. Science 364, 82–85 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sandstrom, A. et al. Functional degradation: a mechanism of NLRP1 inflammasome activation by diverse pathogen enzymes. Science 364, eaau1330 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Taabazuing, C. Y., Griswold, A. R. & Bachovchin, D. A. The NLRP1 and CARD8 inflammasomes. Immunol. Rev. 297, 13–25 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bachovchin, D. A. NLRP1: a jack of all trades, or a master of one? Mol. Cell 81, 423–425 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Okondo, M. C. et al. DPP8 and DPP9 inhibition induces pro-caspase-1-dependent monocyte and macrophage pyroptosis. Nat. Chem. Biol. 13, 46–53 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Okondo, M. C. et al. Inhibition of Dpp8/9 activates the Nlrp1b inflammasome. Cell Chem. Biol. 25, 262–267 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Johnson, D. C. et al. DPP8/DPP9 inhibitor-induced pyroptosis for treatment of acute myeloid leukemia. Nat. Med. 24, 1151–1156 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhong, F. L. et al. Human DPP9 represses NLRP1 inflammasome and protects against autoinflammatory diseases via both peptidase activity and FIIND domain binding. J. Biol. Chem. 293, 18864–18878 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gai, K. et al. DPP8/9 inhibitors are universal activators of functional NLRP1 alleles. Cell Death Dis. 10, 587 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Griswold, A. R. et al. DPP9’s enzymatic activity and not its binding to CARD8 inhibits inflammasome activation. ACS Chem. Biol. 14, 2424–2429 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chui, A. J. et al. Activation of the CARD8 inflammasome requires a disordered region. Cell Rep. 33, 108264 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Hollingsworth, L. R. et al. DPP9 sequesters the C terminus of NLRP1 to repress inflammasome activation. Nature 592, 778–783 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sharif, H. et al. Dipeptidyl peptidase 9 sets a threshold for CARD8 inflammasome formation by sequestering its active C-terminal fragment. Immunity 54, 1392–1404 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Huang, M. et al. Structural and biochemical mechanisms of NLRP1 inhibition by DPP9. Nature 592, 773–777 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Van Goethem, S. et al. Inhibitors of dipeptidyl peptidase 8 and dipeptidyl peptidase 9. Part 2: isoindoline containing inhibitors. Bioorg. Med. Chem. Lett. 18, 4159–4162 (2008).

    Article  PubMed  CAS  Google Scholar 

  22. Linder, A. et al. CARD8 inflammasome activation triggers pyroptosis in human T cells. EMBO J. 39, e105071 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Johnson, D. C. et al. DPP8/9 inhibitors activate the CARD8 inflammasome in resting lymphocytes. Cell Death Dis. 11, 628 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhong, F. L. et al. Germline NLRP1 mutations cause skin inflammatory and cancer susceptibility syndromes via inflammasome activation. Cell 167, 187–202 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Robinson, K. S. et al. Enteroviral 3C protease activates the human NLRP1 inflammasome in airway epithelia. Science 370, eaay2002 (2020).

    Article  CAS  PubMed  Google Scholar 

  26. Drutman, S. B. et al. Homozygous NLRP1 gain-of-function mutation in siblings with a syndromic form of recurrent respiratory papillomatosis. Proc. Natl Acad. Sci. USA 116, 19055–19063 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Grandemange, S. et al. A new autoinflammatory and autoimmune syndrome associated with NLRP1 mutations: NAIAD (NLRP1-associated autoinflammation with arthritis and dyskeratosis). Ann. Rheum. Dis. 76, 1191–1198 (2017).

    Article  CAS  PubMed  Google Scholar 

  28. Ball, D. P. et al. Caspase-1 interdomain linker cleavage is required for pyroptosis. Life Sci. Alliance 3, e202000664 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Hollingsworth, L. R. et al. Mechanism of filament formation in UPA-promoted CARD8 and NLRP1 inflammasomes. Nat. Commun. 12, 189 (2021).

    Article  CAS  Google Scholar 

  30. Gong, Q. et al. Structural basis for distinct inflammasome complex assembly by human NLRP1 and CARD8. Nat. Commun. 12, 188 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tang, H. K. et al. Biochemical properties and expression profile of human prolyl dipeptidase DPP9. Arch. Biochem. Biophys. 485, 120–127 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Lee, H. J. et al. Investigation of the dimer interface and substrate specificity of prolyl dipeptidase DPP8. J. Biol. Chem. 281, 38653–38662 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Geiss-Friedlander, R. et al. The cytoplasmic peptidase DPP9 is rate-limiting for degradation of proline-containing peptides. J. Biol. Chem. 284, 27211–27219 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Griswold, A. R. et al. A chemical strategy for protease substrate profiling. Cell Chem. Biol. 26, 901–907 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brandt, W. et al. A model of the active site of dipeptidyl peptidase IV predicted by comparative molecular field analysis and molecular modelling simulations. Int. J. Pept. Protein Res. 46, 494–507 (1995).

    Article  CAS  PubMed  Google Scholar 

  36. Hikida, A., Ito, K., Motoyama, T., Kato, R. & Kawarasaki, Y. Systematic analysis of a dipeptide library for inhibitor development using human dipeptidyl peptidase IV produced by a Saccharomyces cerevisiae expression system. Biochem. Biophys. Res. Commun. 430, 1217–1222 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Lan, V. T. et al. Analyzing a dipeptide library to identify human dipeptidyl peptidase IV inhibitor. Food Chem. 175, 66–73 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Sekine, K., Fujii, H., Abe, F. & Nishikawa, K. Augmentation of death ligand-induced apoptosis by aminopeptidase inhibitors in human solid tumor cell lines. Int. J. Cancer 94, 485–491 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Krige, D. et al. CHR-2797: an antiproliferative aminopeptidase inhibitor that leads to amino acid deprivation in human leukemic cells. Cancer Res. 68, 6669–6679 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Lupi, A., Tenni, R., Rossi, A., Cetta, G. & Forlino, A. Human prolidase and prolidase deficiency: an overview on the characterization of the enzyme involved in proline recycling and on the effects of its mutations. Amino Acids 35, 739–752 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Wilk, P. et al. Substrate specificity and reaction mechanism of human prolidase. FEBS J. 284, 2870–2885 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Maggiora, L. L., Orawski, A. T. & Simmons, W. H. Apstatin analogue inhibitors of aminopeptidase P, a bradykinin-degrading enzyme. J. Med. Chem. 42, 2394–2402 (1999).

    Article  CAS  PubMed  Google Scholar 

  43. Singh, R. et al. Structure of the human aminopeptidase XPNPEP3 and comparison of its in vitro activity with Icp55 orthologs: insights into diverse cellular processes. J. Biol. Chem. 292, 10035–10047 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bissonnette, R. et al. Prolidase deficiency: a multisystemic hereditary disorder. J. Am. Acad. Dermatol. 29, 818–821 (1993).

    Article  CAS  PubMed  Google Scholar 

  45. Dickson, M. A. et al. Human keratinocytes that express hTERT and also bypass a p16INK4a-enforced mechanism that limits life span become immortal yet retain normal growth and differentiation characteristics. Mol. Cell Biol. 20, 1436–1447 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lupi, A. et al. Characterization of a new PEPD allele causing prolidase deficiency in two unrelated patients: natural-occurrent mutations as a tool to investigate structure–function relationship. J. Hum. Genet. 49, 500–506 (2004).

    Article  CAS  PubMed  Google Scholar 

  49. Duhrkop, K. et al. SIRIUS 4: a rapid tool for turning tandem mass spectra into metabolite structure information. Nat. Methods 16, 299–302 (2019).

    Article  PubMed  CAS  Google Scholar 

  50. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Pew Charitable Trusts (D.A.B. is a Pew-Stewart Scholar in Cancer Research), the NIH (R01 AI137168 and R01 AI163170 to D.A.B.; T32 GM007739-Andersen to A.R.G.; NIH T32 GM115327-Tan to E.L.O.-H.; F30 CA008748 to A.R.G.; the MSKCC Core Grant P30 CA008748; R25AI140472 to J.R.C.), Gabrielle’s Angel Foundation (D.A.B.), Mr William H. and Mrs Alice Goodwin, the Commonwealth Foundation for Cancer Research, and The Center for Experimental Therapeutics of Memorial Sloan Kettering Cancer Center (D.A.B.), the Emerson Collective (D.A.B.) and a Marie-Josée Kravis Women in Science Endeavor (WISE) fellowship (S.D.R.).

Author information

Authors and Affiliations

Authors

Contributions

D.A.B. conceived and directed the project. S.D.R., E.L.O.-H., Q.C., Q.W., A.R.G., D.P.B., H.-C.H., A.J.C., S.Y., and D.J.C. performed cloning, gene editing, biochemistry and cell biology experiments. A.B. synthesized peptides. Q.C. synthesized all other small molecules and performed chemoproteomics experiments. E.L.O.-H., J.R.C. and M.S. performed and analyzed metabolomics experiments. D.A.B. and S.D.R. wrote the manuscript.

Corresponding author

Correspondence to Daniel A. Bachovchin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Rebecca Coll and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Activation of the NLRP1 and CARD8 inflammasomes.

The proteasome-mediated degradation of the NT fragments of NLRP1 and CARD8 release the CT fragments from autoinhibition. DPP8/9 inhibitors and several other danger signals (for example, pathogen proteases) accelerate the rate of NT degradation. If the rate of degradation is slow (top), CT fragments are restrained in ternary complexes consisting of the CT fragment, a full-length (FL) PRR, and DPP9. Disruption of the ternary complex (for example, by VbP) can release the CT fragments to form an inflammasome. If the rate of degradation is fast (bottom), sufficient CT fragments are released to overwhelm the DPP9 ternary complex checkpoint and to form an inflammasome. NLRP1 is activated by a similar overall mechanism, but only CARD8 is shown for clarity. Key differences between NLRP1 and CARD8 are discussed in the text.

Extended Data Fig. 2 Certain Xaa-Pro dipeptides inhibit DPP9 and activate CARD8.

(a) Inhibition of recombinant DPP9 by the twenty XP dipeptides in an AP-AMC cleavage assay. (b) Inhibition of AP-AMC cleavage activity in HEK 293 T cell lysates (pretreated with 10 μM sitagliptin to inhibit any DPP4 activity) by indicated compounds. (c) The indicated cell types were immunoblotted for proteins involved in VbP-induced pyroptosis. MV4;11 and THP-1 cells express CARD8, whereas N/TERT-1 and HEKa cells express NLRP1. Asterisks indicate background bands. (d,e) The indicated MV4;11 cells were treated with compounds (1 mM) and monitored for PI uptake over 12 h (d) or analyzed by CTG and CTF after 24 h (e). (f) MV4;11 cells were treated with XP-OMes (1 mM) for 14 h before CTG and CTF analyses. (g) The indicated AML cell lines were treated with VP-OMe (dose range = 5mM-19.5 μM, 2-fold dilution) for 24 h before CTG analysis. (h) Primary resting CD3+ T-cells were treated with VP-OMe (1 mM), IP-OMe (1 mM) or VbP (10 μM) for 18 h before immunoblot analysis. Data is representative of two independent experiments. (i) J774.1 macrophages were treated with VP-OMe (1 mM) or VbP (2 μM) for 24 h before assaying for LDH release. (j) The indicated RAW264.7 cells were treated with VP-OMe or IP-Ome (5 mM, 24 h) before CTF and CTG analyses. Data in d, e, and g (n = 4) and a,b, f, i, and j (n = 3) are means ± SEM of replicates. All data except where indicated, including immunoblots, are representative of three or more independent experiments.

Source data

Extended Data Fig. 3 CQ31 releases CQ04 in cells and causes pyroptosis.

(a) The indicated MV4;11 cells were treated with CQ04 or CQ31 for 24 h before assessing cell viability by CTG. Data are means ± SEM of 3 biological replicates. (b) The indicated MV4;11 cells were treated with CQ31 (16 μM) or VbP (16 μM), incubated for 4 h, and stained with PI. PI uptake was recorded for 12 h. Data are means ± SEM of 10 replicates. a and b are representative of three or more independent experiments. (c) HEK 293 T cells were treated with vehicle control (DMSO) or CQ31(10 μM) for 24 h before intracellular metabolites were extracted. Methyl ester and free acid of CQ31 were measured by LC-MS and confirmed against pure standards.

Source data

Extended Data Fig. 4 CQ31 selectively activates the CARD8 inflammasome.

(a) Viability of cell lines after treatment with CQ31 for 24 h as assessed by CTF. (b) Human resting T-cells were treated with CQ31 (10 μM) or VbP (10 μM) for 24 h before assessing cell viability by CTG and pyroptotic and apoptotic markers by immunoblotting. Data is representative of two independent experiments. (c) N/TERT-1 immortalized keratinocyte cells were treated with CQ31 (10 μM) or VbP (10 μM), incubated for 1 h, and stained with PI. PI uptake was assessed over 14 h. (d) HEKa immortalized keratinocyte cells were treated with CQ31 (10 μM) or VbP (10 μM), before assaying for LDH release. (e) HEK 293 T cells stably expressing CASP1 and GSDMD were transfected with plasmids expressing NLRP1 and ASC and treated with CQ31 (10 μM) or VbP (10 μM) for 24 h. GSDMD cleavage was assessed by immunoblotting. (f) RAW264.7 cells were treated with CQ31 (100 μM) or VbP (2 μM) for 24 h, before assaying for LDH release. (n = 3) (g) J774.1 cells were treated with various doses of CQ31 before cell viability was measured by CTG and CTF. Data in a (n = 4), b,d,f (n = 3), and c,g (n = 6) are means ± SEM of the indicated replicates. All data except where indicated, including immunoblots, are representative of three or more independent experiments.

Source data

Extended Data Fig. 5 CQ31-induced pyroptosis is DPP8/9 dependent.

(a,b) Indicated THP-1 cells were treated with VP-OMe (1 mM, 14 h) (a) or varying doses of CQ31 or VbP (b) before assaying cell viability by CTG and CTF assays. (c) HEK 293T cells were treated with vehicle control (DMSO), CQ31 (10 μM), or VbP (10 μM) for 6 h before intracellular metabolites were extracted, and dipeptide concentrations were measured by LC-MS. Data in a (n = 4) and b,c (n = 3) are means ± SEM of the indicated replicates. Data in a and b are representative of three or more independent experiments.

Source data

Extended Data Fig. 6 PEPD knockout cells are sensitive to CQ31.

(a) The indicated MV4;11 cells were treated with compounds (16 μM) for 4 h before monitoring for PI uptake. Data is mean ± SEM of 10 biological replicates. (b) The indicated MV4;11 cells were treated with varying doses of CQ31 before assessing cell viability by CTG and CTF assays. (c) The indicated MV4;11 cells were all treated with VX-765 (50 μM) to prevent pyroptosis. Cells were then co-treated with DMSO (control) or CQ31 (10 μM). Intracellular metabolites were extracted and dipeptide concentrations were measured by LC-MS. Data are means ± SEM of the 3 biological replicates, unless indicated otherwise. Data in a and b are representative of three or more independent experiments.

Source data

Extended Data Fig. 7 CQ31 also targets XPNPEP1.

(a) Representative TMT-labeled peptides used to quantify the enrichment of the indicated proteins by CQ73. (b) CETSA analysis of CQ04 (0.5, 5 μM) and CQ31 (0.5, 5 μM) in THP-1Cas9 lysates is representative of three or more independent experiments.

Source data

Extended Data Fig. 8 Dual PEPD and XPNPEP1 inhibition induces pyroptosis.

(a) XPNPEP1/PEPD knockout THP-1 cells were treated with the indicated concentrations of CQ31 for 48 h prior to CTG analysis. (b) The indicated THP-1 cells were treated with VbP (10 μM) or CQ31 (20 μM) for 48 h prior to evaluating supernatants for IL-1β levels by ELISA and evaluating lysates and supernatants for IL-1β cleavage by immunoblotting. (c) Evaluation of XPNPEP1 and PEPD levels in MV4;11 cells stably expressing Cas9 and treated with sgRNAs targeting XPNPEP1. (d) The indicated cells were treated with CQ31 for 24 h before LDH release was evaluated. (e) The indicated MV4;11 cells were treated with VbP (10 μM) or CQ31 (20 μM) for 48 h before LDH release and immunoblot analyses. PEPD/XPNPEP1 knockout MV4;11 cells were generated with sgPEPD_2 and sgXPNPEP1. Data are means ± SEM of biological replicates. *** p < 0.001, ** p < 0.01 by two-sided Students t-test. (f) The indicated MV4;11 cells were treated with CQ31 for 48 h before assessing cell viability by CTF. (g) Inhibition of AP-AMC cleavage in HEK 293 T lysates by methyl esters of IPI or VPI tripeptides. (h) The indicated MV4;11 cells were treated with methyl esters of IPI or VPI for 24 h before CTG analysis. Data in b-h are means ± SEM of 3 replicates. All data, including immunoblots, are representative of three or more independent experiments.

Source data

Extended Data Fig. 9 Schematic comparing CQ31-induced and VbP-induced activation inflammasome activation.

(a) Weak DPP8/9 inhibition selectively activates CARD8, whereas strong DPP8/9 inhibition activates both NLRP1 and CARD8. (b) Strong DPP8/9 inhibitors (for example, VbP) are required to compete with the neo-N-terminus of the NLRP1CT fragment for the DPP8/9 active site and destabilize the ternary complex. (c) CARD8 does not directly interact with the DPP8/9 active site, and thus strong inhibitors are not needed to disrupt that interaction. It has not yet been established precisely how DPP8/9 inhibitors impact the CARD8-DPP9 ternary complex in cells. Red dots in b and c represent the DPP9 active site serine.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2 and note.

Reporting Summary

41589_2021_964_MOESM3_ESM.xlsx

Supplementary Data 1 Supplementary dataset. List of proteins enriched by CQ73 analyzed by TMT-mass spectrometry. Rank-ordered list of proteins enriched by CQ73 and whose binding was competed off by CQ31. No probe treatment corresponds to TMT labels 126 and 127, CQ73 treatment corresponds to TMT labels 128 and 129, and CQ73 treatment in competition with CQ31 corresponds to TMT labels 130 and 131.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Figure 7

Unprocessed western blots.

Source Data Extended Data Figure 8

Unprocessed western blots.

Source Data Extended Data Figure 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rao, S.D., Chen, Q., Wang, Q. et al. M24B aminopeptidase inhibitors selectively activate the CARD8 inflammasome. Nat Chem Biol 18, 565–574 (2022). https://doi.org/10.1038/s41589-021-00964-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-021-00964-7

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