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:

Chemical inhibition of DPP9 sensitizes the CARD8 inflammasome in HIV-1-infected cells

Abstract

Non-nucleoside reverse transcriptase inhibitors (NNRTIs) induce pyroptosis of HIV-1-infected CD4+ T cells through induction of intracellular HIV-1 protease activity, which activates the CARD8 inflammasome. Because high concentrations of NNRTIs are required for efficient elimination of HIV-1-infected cells, it is important to elucidate ways to sensitize the CARD8 inflammasome to NNRTI-induced activation. We show that this sensitization can be achieved through chemical inhibition of the CARD8 negative regulator DPP9. The DPP9 inhibitor Val-boroPro (VbP) can kill HIV-1-infected cells without the presence of NNRTIs and act synergistically with NNRTIs to promote clearance of HIV-1-infected cells in vitro and in humanized mice. More importantly, VbP is able to enhance clearance of residual HIV-1 in CD4+ T cells isolated from people living with HIV (PLWH). We also show that VbP can partially overcome NNRTI resistance. This offers a promising strategy for enhancing NNRTI efficacy in the elimination of HIV-1 reservoirs in PLWH.

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: NNRTIs induce dose-dependent death of HIV-1-infected cells.
Fig. 2: DPP9 inhibition sensitizes the CARD8 inflammasome to HIV-1.
Fig. 3: VbP enhancement is CARD8- and HIV-1 protease dependent.
Fig. 4: CARD8 inflammasome sensitization overcomes NNRTI resistance.
Fig. 5: VbP enhances clearance of HIV-1-infected cells in mice.
Fig. 6: VbP enhances clearance of latent HIV-1.

Similar content being viewed by others

Data availability

Data supporting the findings of this study are available within the paper and its Supplementary Information files. Raw data generated with flow cytometry are available upon request from the corresponding author. Unprocessed western blots from the Supplementary figures are provided at the end of the Supplementary Information file. Source data are provided with this paper.

References

  1. Gupta, R. K. et al. HIV-1 remission following CCR5Δ32/Δ32 haematopoietic stem-cell transplantation. Nature 568, 244–248 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Hütter, G. et al. Long-term control of HIV by CCR5Δ32/Δ32 stem-cell transplantation. N. Engl. J. Med. 360, 692–698 (2009).

    Article  PubMed  Google Scholar 

  3. Castro-Gonzalez, S., Colomer-Lluch, M. & Serra-Moreno, R. Barriers for HIV cure: the latent reservoir. AIDS Res. Hum. Retroviruses 34, 739–759 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Finzi, D. et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278, 1295–1300 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Ganor, Y. et al. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat. Microbiol. 4, 633–644 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Eisele, E. & Siliciano, R. F. Redefining the viral reservoirs that prevent HIV-1 eradication. Immunity 37, 377–388 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kim, Y., Anderson, J. L. & Lewin, S. R. Getting the ‘kill’ into ‘shock and kill’: strategies to eliminate latent HIV. Cell Host Microbe 23, 14–26 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wang, Q. et al. CARD8 is an inflammasome sensor for HIV-1 protease activity. Science 371, eabe1707 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  10. Gross, O., Thomas, C. J., Guarda, G. & Tschopp, J. The inflammasome: an integrated view. Immunol. Rev. 243, 136–151 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. 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 

  12. 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 

  13. 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 

  14. 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  Google Scholar 

  15. Hsiao, J. C. et al. A ubiquitin-independent proteasome pathway controls activation of the CARD8 inflammasome. J. Biol. Chem. 298, 102032 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. 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  PubMed Central  Google Scholar 

  17. Figueiredo, A. et al. Potent nonnucleoside reverse transcriptase inhibitors target HIV-1 Gag-Pol. PLoS Pathog. 2, e119 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Phillips, R. E. et al. Human immunodeficiency virus genetic variation that can escape cytotoxic T-cell recognition. Nature 354, 453–459 (1991).

    Article  CAS  PubMed  Google Scholar 

  19. Kwong, P. D. et al. HIV-1 evades antibody-mediated neutralization through conformational masking of receptor-binding sites. Nature 420, 678–682 (2002).

    Article  CAS  PubMed  Google Scholar 

  20. Caskey, M. et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 522, 487–491 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rhee, S. Y. et al. HIV-1 protease, reverse transcriptase and integrase variation. J. Virol. 90, 6058–6070 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jochmans, D. et al. Selective killing of human immunodeficiency virus infected cells by non-nucleoside reverse transcriptase inhibitor-induced activation of HIV protease. Retrovirology 7, 89 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Zerbato, J. M., Tachedjian, G. & Sluis-Cremer, N. Nonnucleoside reverse transcriptase inhibitors reduce HIV-1 production from latently infected resting CD4. Antimicrob. Agents Chemother. 61, e01736–16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Boffito, M. et al. Protein binding in antiretroviral therapies. AIDS Res. Hum. Retroviruses 19, 825–835 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Almond, L. M., Hoggard, P. G., Edirisinghe, D., Khoo, S. H. & Back, D. J. Intracellular and plasma pharmacokinetics of efavirenz in HIV-infected individuals. J. Antimicrob. Chemother. 56, 738–744 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Rotger, M. et al. Influence of CYP2B6 polymorphism on plasma and intracellular concentrations and toxicity of efavirenz and nevirapine in HIV-infected patients. Pharmacogenet. Genomics 15, 1–5 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Tanaka, R. et al. Intracellular efavirenz levels in peripheral blood mononuclear cells from human immunodeficiency virus-infected individuals. Antimicrob. Agents Chemother. 52, 782–785 (2008).

    Article  CAS  PubMed  Google Scholar 

  28. Griswold, A. 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 

  29. Wu, J. J. et al. Biochemistry, pharmacokinetics and toxicology of a potent and selective DPP8/9 inhibitor. Biochem. Pharmacol. 78, 203–210 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Lankas, G. R. et al. Dipeptidyl peptidase IV inhibition for the treatment of type 2 diabetes: potential importance of selectivity over dipeptidyl peptidases 8 and 9. Diabetes 54, 2988–2994 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Ianevski, A., Giri, A. & Aittokallio, T. SynergyFinder 2.0: visual analytics of multi-drug combination strategies. Nucleic Acids Res. 48, W488–W493 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Loewe, S. The problem of synergism and antagonism of combined drugs. Arzneimiettelforschung 3, 286–290 (1953).

    Google Scholar 

  33. Nie, Z. et al. HIV-1 protease processes procaspase 8 to cause mitochondrial release of cytochrome c, caspase cleavage and nuclear fragmentation. Cell Death Differ. 9, 1172–1184 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Preston, B. D. & Dougherty, J. P. Mechanisms of retroviral mutation. Trends Microbiol. 4, 16–21 (1996).

    Article  CAS  PubMed  Google Scholar 

  35. Azijn, H. et al. TMC278, a next-generation nonnucleoside reverse transcriptase inhibitor (NNRTI), active against wild-type and NNRTI-resistant HIV-1. Antimicrob. Agents Chemother. 54, 718–727 (2010).

    Article  CAS  PubMed  Google Scholar 

  36. Waters, J. M. et al. Mutations in the thumb-connection and RNase H domain of HIV type-1 reverse transcriptase of antiretroviral treatment-experienced patients. Antivir. Ther. 14, 231–239 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. King, R. W., Klabe, R. M., Reid, C. D. & Erickson-Viitanen, S. K. Potency of nonnucleoside reverse transcriptase inhibitors (NNRTIs) used in combination with other human immunodeficiency virus NNRTIs, NRTIs or protease inhibitors. Antimicrob. Agents Chemother. 46, 1640–1646 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Basson, A. E. et al. Impact of drug resistance-associated amino acid changes in HIV-1 subtype C on susceptibility to newer nonnucleoside reverse transcriptase inhibitors. Antimicrob. Agents Chemother. 59, 960–971 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Rongvaux, A. et al. Development and function of human innate immune cells in a humanized mouse model. Nat. Biotechnol. 32, 364–372 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Herndler-Brandstetter, D. et al. Humanized mouse model supports development, function and tissue residency of human natural killer cells. Proc. Natl Acad. Sci. USA 114, E9626–E9634 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Siliciano, J. D. et al. Enhanced culture assay for detection and quantitation of latently infected, resting CD4+ T cells carrying replication-competent virus in HIV-1 infected individuals. Methods Mol. Biol. 304, 3–15 (2005).

    PubMed  Google Scholar 

  42. Rao, S. D. et al. M24B aminopeptidase inhibitors selectively activate the CARD8 inflammasome. Nat. Chem. Biol. 18, 565–574 (2022).

    Article  CAS  PubMed  Google Scholar 

  43. Laird, G. M. et al. Measuring the frequency of latent HIV-1 in resting CD4+ T cells using a limiting dilution coculture assay. Methods Mol. Biol. 1354, 239–253 (2016).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the volunteers who participated in this study, and L. Kessels, M. Klebert, A. Haile, T. Spitz and T. Minor for study subject recruitment. We thank Regeneron Pharmaceuticals and the Richard Flavell Laboratory at Yale University for generating the human cytokine knock-in mice. The following reagents were obtained through the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH: rilpivirine, efavirenz, lopinavir, etravirine, nevirapine, maraviroc, T-20, tenofovir, raltegravir, pNL4-3-GFP, international HIV-1 isolates and HIV-1 p24 antibodies. Funding in support of this work was provided by NIH grants nos. R01AI162203 and R01AI155162 (L.S.) and F31AI165251 (K.M.C.). The organizations that supplied these funds had no part in the planning or execution of the work presented in this study.

Author information

Authors and Affiliations

Authors

Contributions

L.S., K.M.C. and Q.W. designed the study. L.S. and K.M.C. analyzed the data and wrote the manuscript. J.G.K. edited the manuscript, performed experiments using dox-inducible CARD8 constructs and assisted in mouse and IUPM experiments. Q.W. conducted DPP9 knockdown experiments, provided CARD8- and Caspase 1-KO cell lines and generated dox-inducible CARD8 cell lines. H.G. conducted experiments of live/dead staining of GFP-sorted THP-1 cells. K.M.C. conducted all other experiments. R.M.P. supervised the studies using clinical samples.

Corresponding author

Correspondence to Liang Shan.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Andrew Badley, Bernhard Ellinger, Franklin Zhong 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 Cell killing measurement.

a, Scheme of the killing assays for single-round HIV-1 reporter viruses. Primary CD4+ T cells were infected with HIV-1 reporter viruses for 3 days before NNRTI treatment for 2 days. b, c, Representative flow cytometry plots are shown for one replicate of one donor. Percent killing is calculated as the percent infection of the treatment condition divided by the percent infection of DMSO control. Percent infection was determined by GFP (b) or intracellular p24 (c). d, Percent killing calculated from c. Unpaired two-sided t test. **** p < 0.0001. Error bars show mean values with SEM (n = 3). e, The log fold increase in EC50 calculated from the treatment of three donors of CD4+ T cells with EFV, RPV and ETR with or without the presence of 50% human serum (HS) in the culture media. Data from Fig. 1f–h were used.

Extended Data Fig. 2 DPP9 inhibition enhances NNRTI-mediated killing in THP1 cells.

a, Two shRNA constructs delivered by lentiviral vectors for knockdown of DPP9 are confirmed by immunoblotting (experiment repeated three times with similar results). b, Killing of HIV-1-infected THP1 cells by RPV. THP-1 cells were transduced with lentiviruses carrying DPP9-specific or scramble shRNA. Two-way ANOVA with Dunnett’s multiple comparison test (n = 3). c, Killing of HIV infected THP-1 cells treated for two days with DMSO, EFV, 1g244, or combination (n = 3). One-way ANOVA with Tukey’s multiple comparison test. * p < 0.05 and **** p < 0.0001. Error bars show mean values with SEM. d, Log fold changes in EC50 due to VbP. e, Combination treatment denotes a synergistic relationship as evidenced by Loewe’s additivity model calculated with SynergyFinder2.0. Areas in red denote increased synergy. In (d, e), data from Fig. 2b were used for the analysis.

Source data

Extended Data Fig. 3 CARD8 inflammasome activation is dependent on Gag-Pol expression.

a, Representative flow cytometry plots of CARD8-KO or Cas9 control THP-1 cells treated with DMSO, EFV (3 µM), VbP (1 µM), or combination. Heatmap plots are colored according to mean fluorescent intensity (MFI) of GFP with higher MFI colored in red and lower MFI in green. b, Mean fluorescent intensity of three replicates from (a). Two-way ANOVA with Dunnett’s multiple comparison test. c, Frequency of HIV-1 DNA+ cells measured by qPCR. CARD8-KO or Cas9 control THP-1 cells were infected with HIV-1 for 3 days and treated with DMSO, EFV (3 µM), VbP (1 µM), or combination for 2 days. HIV-1 gag levels were normalized to POLR2A to determine the frequency of HIV-1 DNA+ cells. The frequency in the treatment groups was measured as the percent of the DMSO control (n = 3). Two-way ANOVA with Tukey’s multiple comparison test. * p < 0.05, ** p < 0.01 and **** p < 0.0001. Error bars show mean values with SEM.

Extended Data Fig. 4 VbP cytotoxicity is CARD8 and HIV-1 specific.

a, VbP cytotoxicity. THP-1 cells were treated for two days with EFV with or without VbP. Cell viability was determined by the MTS assay denoted by the heatmap. b, VbP toxicity is CARD8 dependent. CARD8-KO and Cas9 control THP-1 cells were treated with VbP for 4 days. Cell viability was determined by the MTS assay. Two-way ANOVA with Dunnett’s multiple comparison test. c, d, VbP killing of THP-1 cells is HIV-1 and CARD8 dependent. CARD8-KO or Cas9 control THP-1 cells were infected with HIV-1 reporter viruses. On day 3 post infection, GFP+ and GFP- cells were purified by sorting before treatment with DMSO, EFV (3 µM), VbP (1 µM), or combination for 2 days. In c, representative flow cytometry plots of live dead staining. In (d), percent live cells normalized to DMSO control for Cas9 control (top) and CARD8-KO (bottom). One-way ANOVA with Tukey’s multiple comparison test. e, f, Time course of live/dead staining of CD4+ T cells treated with DMSO, EFV (3 µM), VbP, or combinations. Primary CD4+ T cells were co-stimulated with CD3 and CD28 antibodies for 3 days before EFV and VbP treatment. Percent live were normalized to DMSO control. In (e), representative FACS plots for the 48 hr time point. Error bars show mean values with SEM (n = 3). * p < 0.05, *** p < 0.001 and **** p < 0.0001.

Extended Data Fig. 5 VbP sensitizes the CARD8 inflammasome to HIV-1.

a, VbP overcomes reduced RPV killing efficacy by human serum. Dose response curves for killing of HIV-1-infected CD4+ T cells treated with RPV or combo (VbP 1 µM) with or without the presence of 50% human serum. Zero values of RPV were plotted at 0.1 nM to allow for log transformation. Error bars show mean values with SEM (n = 3). be, Time course treatment of three donors of HIV-1 infected primary CD4+ T cells (b, c) or THP-1 cells (d, e). Cells were treated with DMSO, EFV (0.5 µM), VbP (1 µM), or combo. Fold change enhancement of combination treatment in comparison to EFV alone treatment was shown in (c) and (e).

Extended Data Fig. 6 Characterization of VbP enhancement of CARD8 activation.

a, VbP enhancement and killing is HIV-1 protease dependent. Percent killing of HIV-1-infected THP-1 cells treated for two days with DMSO, EFV, VbP, or combination with or without protease inhibitor LPV (1 µM). One-way ANOVA with Tukey’s multiple comparison test. Error bars show mean values with SEM (n = 3). b, CARD8 cleavage in transfected HEK293T cells. HEK293T cells were co-transfected with a CARD8-expressing plasmid and the HIV-1 plasmid pNL4-3-GFP with the presence of indicated drugs. EFV: 3 µM. VbP: 1 µM. LPV: 1 µM. Cell lysates were collected 24hrs post transfection for western blot analysis. This experiment was repeated an additional two times and provided similar results. c, CARD8 cleavage in HIV-1-infected MT4 cells. MT4 cells were stably transduced with lentiviral vectors expressing WT or FAFA CARD8. Cells were infected for three days prior to treatment with DMSO, EFV (3 µM), VbP (1 µM), or EFV and VbP combination in the presence of a proteasome inhibitor MG132 (5 µM) to block degradation of the neo-C-fragment. Cell lysates were collected six hours post treatment for western blot analysis. This experiment was repeated an additional two times and provided similar results. d, Percent killing of CARD8-KO THP-1 cells replete with Dox-inducible CARD8 constructs for WT, S297A, or FAFA. Conditions shown were treated as in Fig. 3E but were not dox induced. One-way ANOVA with Dunnett’s multiple comparison test. **** p < 0.0001.

Source data

Extended Data Fig. 7 VbP enhancement and killing is Caspase 8 independent.

a, Two sgRNA constructs for knockout of Caspase 8 are confirmed by immunoblotting (experiment repeated three times with similar results). b, Percent killing of Cas9 Control, Casp8-KO, or CARD8-KO THP-1 cells infected with HIV-1 reporter viruses. Infected cells were treated with EFV (3 µM) with or without VbP for two days. Two-way ANOVA with Dunnett’s multiple comparison test. Error bars show mean values with SEM (n = 3). **** p < 0.0001.

Source data

Extended Data Fig. 8 Killing of cells infected with NNRTI RAMs by VbP.

Primary CD4+ T cells were infected with HIV-1 reporter viruses carrying various NNRTI RAMs for 3 days before treatment with 1 µM VbP for two days. Error bars show mean values with SEM (n = 3). One-way ANOVA with Dunnett’s multiple comparison test. ** p < 0.01 and **** p < 0.0001.

Extended Data Fig. 9 Combination treatment effects in vivo.

Primary CD4+ T cells were infected with the HIV-1 reporter virus NL4-3-Pol. Three days post infection, these cells were transfused into mice (5–10 million cells per mouse). EFV and VbP were provided by IV injection immediately after cell infusion. Remaining infected cells were measured by flow cytometry. a, Representative flow cytometry plots measuring remaining infected CD4+ T cells in blood 6hrs post treatment. b, c, blood samples were collected 6hrs and 24 hr post EFV and VbP treatment (n = 4) or control (n = 3). d, Lung tissues were collected 24 hrs post EFV and VbP treatment. Two-way ANOVA with Sidak’s multiple hypothesis test. e, f, Comparison of killing between IV and IP injection of EFV and VbP. Blood samples were collected 6hrs and 24 hr post EFV and VbP treatment. Two-way ANOVA with Sidak’s multiple hypothesis test. (IV sample sizes are the same as b-d, IP samples sizes are as follows: DMSO = 17, EFV = 12, combination = 20. g, CD4+ T cell counts from lung tissues of mice treated with control (n = 5), EFV (n = 4), VbP (n = 5), or combination (n = 5) after 24 hours. Two-way ANOVA with Dunnett’s multiple hypothesis test. h, CD4+ T cell counts from lung tissues from mice with control (n = 5), single-dose (n = 5), or multi-dose (n = 7) combination treatment regimens. Two-way ANOVA with Dunnett’s multiple hypothesis test. Error bars show mean values with SEM. ** p < 0.01, **** p < 0.0001.

Extended Data Fig. 10 DPP9 inhibition enhances clearance of HIV-1 clinical isolates.

a, b, CD4+ T cells were infected with HIV-LAI for four days and treated for one day with DMSO, EFV (1 µM), VbP (1 µM), or combination. In (a), representative flow cytometry plots. Heatmap plots are colored according to mean fluorescent intensity (MFI) of intracellular HIV-p24 (PE) with higher MFI colored in red and lower MFI in green. In (b), Mean fluorescent intensity of three replicates from (a). One-way ANOVA with Dunnett’s multiple hypothesis test. ch, 1G244 enhances killing of primary CD4+ T cells infected with various HIV-1 clinical isolates. Cells were infected for 4–6 days before treated with DMSO, EFV, 1g244, or combination for one day prior to intracellular HIV-p24 staining. One-way ANOVA with Tukey’s multiple comparisons test. Error bars show mean values with SEM (n = 3). * p < 0.05, ** p < 0.01, ** p < 0.001 and **** p < 0.0001.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Source data

Source Data Extended Data Fig. 2

Uncropped, unprocessed scans of blots.

Source Data Extended Data Fig. 6

Uncropped, unprocessed scans of blots.

Source Data Extended Data Fig. 7

Uncropped, unprocessed scans of blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Clark, K.M., Kim, J.G., Wang, Q. et al. Chemical inhibition of DPP9 sensitizes the CARD8 inflammasome in HIV-1-infected cells. Nat Chem Biol 19, 431–439 (2023). https://doi.org/10.1038/s41589-022-01182-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-022-01182-5

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