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:

Streptococcal pyrogenic exotoxin B cleaves GSDMA and triggers pyroptosis

An Author Correction to this article was published on 27 July 2022

This article has been updated

Abstract

Gasdermins, a family of five pore-forming proteins (GSDMA–GSDME) in humans expressed predominantly in the skin, mucosa and immune sentinel cells, are key executioners of inflammatory cell death (pyroptosis), which recruits immune cells to infection sites and promotes protective immunity1,2. Pore formation is triggered by gasdermin cleavage1,2. Although the proteases that activate GSDMB, C, D and E have been identified, how GSDMA—the dominant gasdermin in the skin—is activated, remains unknown. Streptococcus pyogenes, also known as group A Streptococcus (GAS), is a major skin pathogen that causes substantial morbidity and mortality worldwide3. Here we show that the GAS cysteine protease SpeB virulence factor triggers keratinocyte pyroptosis by cleaving GSDMA after Gln246, unleashing an active N-terminal fragment that triggers pyroptosis. Gsdma1 genetic deficiency blunts mouse immune responses to GAS, resulting in uncontrolled bacterial dissemination and death. GSDMA acts as both a sensor and substrate of GAS SpeB and as an effector to trigger pyroptosis, adding a simple one-molecule mechanism for host recognition and control of virulence of a dangerous microbial pathogen.

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: The GAS virulence factor SpeB triggers lytic death of skin epithelial cells.
Fig. 2: SpeB triggers pyroptosis in a GSDMA-dependent manner.
Fig. 3: SpeB directly cleaves GSDMA after Gln246.
Fig. 4: Gsdma1 deficiency blunts host anti-GAS immunity.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are included in this manuscript and its supplementary information. Source data are provided with this paper.

Change history

References

  1. Broz, P., Pelegrin, P. & Shao, F. The gasdermins, a protein family executing cell death and inflammation. Nat. Rev. Immunol. 20, 143–157 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Liu, X., Xia, S., Zhang, Z., Wu, H. & Lieberman, J. Channelling inflammation: gasdermins in physiology and disease. Nat. Rev. Drug Discov. 20, 384–405 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Cole, J. N., Barnett, T. C., Nizet, V. & Walker, M. J. Molecular insight into invasive group A streptococcal disease. Nat. Rev. Microbiol. 9, 724–736 (2011).

    Article  CAS  PubMed  Google Scholar 

  4. Musser, J. M., Stockbauer, K., Kapur, V. & Rudgers, G. W. Substitution of cysteine 192 in a highly conserved Streptococcus pyogenes extracellular cysteine protease (interleukin 1beta convertase) alters proteolytic activity and ablates zymogen processing. Infect. Immun. 64, 1913–1917 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Svensson, M. D. et al. Role for a secreted cysteine proteinase in the establishment of host tissue tropism by group A streptococci. Mol. Microbiol. 38, 242–253 (2000).

    Article  CAS  PubMed  Google Scholar 

  6. Cole, J. N. et al. Trigger for group A streptococcal M1T1 invasive disease. FASEB J. 20, 1745–1747 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Shelburne, S. A., 3rd et al. Growth characteristics of and virulence factor production by group A Streptococcus during cultivation in human saliva. Infect. Immun. 73, 4723–4731 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Aziz, R. K. et al. Invasive M1T1 group A Streptococcus undergoes a phase-shift in vivo to prevent proteolytic degradation of multiple virulence factors by SpeB. Mol. Microbiol. 51, 123–134 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  Google Scholar 

  11. 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 

  12. 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 

  13. Sumitomo, T. et al. Streptococcal cysteine protease-mediated cleavage of desmogleins is involved in the pathogenesis of cutaneous infection. Front. Cell Infect. Microbiol. 8, 10 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Saouda, M., Wu, W., Conran, P. & Boyle, M. D. Streptococcal pyrogenic exotoxin B enhances tissue damage initiated by other Streptococcus pyogenes products. J. Infect. Dis. 184, 723–731 (2001).

    Article  CAS  PubMed  Google Scholar 

  15. Paz, I. et al. Galectin-3, a marker for vacuole lysis by invasive pathogens. Cell Microbiol. 12, 530–544 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Doran, J. D. et al. Autocatalytic processing of the streptococcal cysteine protease zymogen: processing mechanism and characterization of the autoproteolytic cleavage sites. Eur. J. Biochem. 263, 145–151 (1999).

    Article  CAS  PubMed  Google Scholar 

  17. Gerwin, B. I., Stein, W. H. & Moore, S. On the specificity of streptococcal proteinase. J. Biol. Chem. 241, 3331–3339 (1966).

    Article  CAS  PubMed  Google Scholar 

  18. Nomizu, M. et al. Substrate specificity of the streptococcal cysteine protease. J. Biol. Chem. 276, 44551–44556 (2001).

    Article  CAS  PubMed  Google Scholar 

  19. Carroll, R. K. & Musser, J. M. From transcription to activation: how group A Streptococcus, the flesh-eating pathogen, regulates SpeB cysteine protease production. Mol. Microbiol. 81, 588–601 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Lichtenberger, B. M. et al. Epidermal EGFR controls cutaneous host defense and prevents inflammation. Sci. Transl. Med. 5, 199ra111 (2013).

    Article  PubMed  Google Scholar 

  21. Sawada, Y. et al. Cutaneous innate immune tolerance is mediated by epigenetic control of MAP2K3 by HDAC8/9. Sci. Immunol. 6, eabe1935 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kolaczkowska, E. & Kubes, P. Neutrophil recruitment and function in health and inflammation. Nat. Rev. Immunol. 13, 159–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Barnett, T. C. et al. The globally disseminated M1T1 clone of group A Streptococcus evades autophagy for intracellular replication. Cell Host Microbe 14, 675–682 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. LaRock, C. N. et al. IL-1β is an innate immune sensor of microbial proteolysis. Sci. Immunol. 1, eaah3539 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Nitsche-Schmitz, D. P., Rohde, M. & Chhatwal, G. S. Invasion mechanisms of Gram-positive pathogenic cocci. Thromb. Haemost. 98, 488–496 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Rohde, M. & Chhatwal, G. S. Adherence and invasion of streptococci to eukaryotic cells and their role in disease pathogenesis. Curr. Top. Microbiol. Immunol. 368, 83–110 (2013).

    PubMed  Google Scholar 

  27. Hynes, W. & Sloan, M. in Streptococcus pyogenes: Basic Biology to Clinical Manifestations (eds Ferretti, J. J. et al.) (2016).

  28. Zhang, W., Rong, C., Chen, C. & Gao, G. F. Type-IVC secretion system: a novel subclass of type IV secretion system (T4SS) common existing in gram-positive genus Streptococcus. PLoS ONE 7, e46390 (2012).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  29. Richter, J. et al. Streptolysins are the primary inflammasome activators in macrophages during Streptococcus pyogenes infection. Immunol. Cell Biol. 99, 1040–1052(2021).

    Article  CAS  PubMed  Google Scholar 

  30. Richter, J., Brouwer, S., Schroder, K. & Walker, M. J. Inflammasome activation and IL-1β signalling in group A Streptococcus disease. Cell Microbiol. 23, e13373 (2021).

    Article  CAS  PubMed  Google Scholar 

  31. Brouwer, S., Barnett, T. C., Rivera-Hernandez, T., Rohde, M. & Walker, M. J. Streptococcus pyogenes adhesion and colonization. FEBS Lett. 590, 3739–3757 (2016).

    Article  CAS  PubMed  Google Scholar 

  32. Carothers, K. E. et al. The streptococcal protease SpeB antagonizes the biofilms of the human pathogen Staphylococcus aureus USA300 through cleavage of the staphylococcal SdrC protein. J. Bacteriol. 202, e00008-e00020 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Zhou, W. et al. Structure of the human cGAS–DNA complex reveals enhanced control of immune surveillance. Cell 174, 300–311.e311, (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kranzusch, P. J. et al. Ancient origin of cGAS–STING reveals mechanism of universal 2′,3′ cGAMP signaling. Mol. Cell 59, 891–903 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Orzalli, M. H. & Kagan, J. C. A one-protein signaling pathway in the innate immune system. Sci. Immunol. 1, eaah6184 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Mitchell, P. S., Sandstrom, A. & Vance, R. E. The NLRP1 inflammasome: new mechanistic insights and unresolved mysteries. Curr. Opin. Immunol. 60, 37–45 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Whatmore, A. M. & Kehoe, M. A. Horizontal gene transfer in the evolution of group A streptococcal emm-like genes: gene mosaics and variation in Vir regulons. Mol. Microbiol. 11, 363–374 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Zheng, Z. Z. et al. The lysosomal Rag–Ragulator complex licenses RIPK1-and caspase-8-mediated pyroptosis by Yersinia. Science 372, eabg0269(2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, W. et al. Quality control, modeling, and visualization of CRISPR screens with MAGeCK-VISPR. Genome Biol. 16, 281, (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Shepard, L. A., Shatursky, O., Johnson, A. E. & Tweten, R. K. The mechanism of pore assembly for a cholesterol-dependent cytolysin: formation of a large prepore complex precedes the insertion of the transmembrane β-hairpins. Biochemistry 39, 10284–10293 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank V. Nizet for sharing S. pyogenes isolate M1T1 strain 5448 and its isogenic mutant strains (ΔspeB, covR/S, ΔcepA and Δmac variants) as well as SpeB constructs, Z. Zhang for providing S. pyogenes M9, M12 amd M73 strains, C. Wang for providing Phage-Flag vector, and Y. Chen for providing a modified pET vector with an N-terminal 6×His-SUMO tag. This work was supported by National Key R&D Program of China (2020YFA0509600), National Natural Science Foundation of China (32122034, 31972897), Key Research Program of the Chinese Academy of Sciences (ZDBS-LY-SM008), Shanghai Pilot Program for Basic Research–Chinese Academy of Sciences, Shanghai Branch (JCYJ-SHFY-2021-009), Strategic Priority Research Program of Chinese Academy of Sciences (XDB29030300), Shanghai Municipal Science and Technology Major Project (2019SHZDZX02) (X.L.), NIH R01CA240955 and R01AI39914 (J.L.), NIH R01AI127654 (T.S.K.) and China Postdoctoral Science Foundation (2019M650193), Guangzhou Science and Technology Project (202102020093) (W.D.).

Author information

Authors and Affiliations

Authors

Contributions

W.D., Y.B., F.D., Y.P., J.L. and X.L. conceived the study. W.D., Y.B., F.D. and Y.P. designed and performed most experiments with assistance from R. Min, Z.W. and W.L. Z. Zheng and S.M. performed the CRISPR screen and sequencing data analysis, respectively. R. Miao and Z. Zhang provided technical support. W.D., Y.B., F.D., Y.P. and X.L. analysed the data. All authors discussed the results and commented on the manuscript. W.D., Y.B., F.D., Y.P., T.S.K., J.L. and X.L. wrote the manuscript. X.L. supervised the study.

Corresponding author

Correspondence to Xing Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review information

Nature thanks Dario Zamboni 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 figures and tables

Extended Data Fig. 1 SpeB-deficient GAS triggers systemic infection.

a, DNA sequence comparison of GAS isolate M1T1 strain 5448 and its isogenic mutant strains (ΔcepA, Δmac, covR/S and ΔspeB variants). b, c, RT-PCR (b) and immunoblot analysis (c) of the expression of SpeB in the indicated GAS strains. df, mice were infected or not with the indicated GAS strains. d, IHC analysis of neutrophil infiltration at infection site on day 1. Scale bar: 100 μm. e, Bacteria load measured from skin lesions, spleens and livers of mice infected or not with GAS. f, Survival rate of mice challenged or not with the indicated GAS (n = 18 mice per group). e, box plots show all points, min to max (n = 5 mice per group). The center line, upper limit and lower limit of the box denote median, 25th and 75th percentiles and the whiskers denote the minimum and maximum values of data. e, Two-tailed Student’s t-test; f, Mantel-Cox log-rank test. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 2 SpeB contributes to local tissue destruction.

a, b, The re-expression of SpeB in ΔspeB GAS was confirmed by both RT-PCR (a) and immunoblot analysis (b). ch, mice were infected or not with the indicated GAS strains. c, Representative image of skin lesions of mice challenged with GAS or not for 1 day. d, Quantification of skin lesion size. e, Histopathology of skin biopsies analysed by H&E staining. f, g, IHC analysis and quantification of neutrophil infiltration at infection site. h, Bacteria load measured from skin lesions, spleens and livers of mice infected or not with GAS. Scale bar: 100 μm. d, g, show mean ± s.d. (n = 5 mice per group); h, box plots show all points, min to max (n = 5 mice per group). The center line, upper limit and lower limit of the box denote median, 25th and 75th percentiles and the whiskers denote the minimum and maximum values of data. d, g, h, Two-tailed Student’s t-test. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 3 SpeB triggers lytic death of primary mouse keratinocytes and A431 cells.

a, Primary mouse keratinocytes were infected with GAS isolate M1T1 strain 5448 or its isogenic mutant strains for the indicated times. Percentage of internalized GAS was shown by counting intracellular CFUs relative to the inoculum (left panel). Cell cytotoxicity of all the cells in a well was measured by LDH release assay (right panel). bd, Primary keratinocytes infected with FITC-labeled GAS strains were washed with PBS before cells were stained and analyzed by confocal fluorescence imaging. Cell borders are outlined with dashed lines. Cells with intracellular GAS were quantified from 700 cells (n = 5, mean ± s.d.) (b). e, Levels of hyaluronic acid capsule at logarithmic and stationary growth phases. f, RT-PCR (left panel) and immunoblot analysis (right panel) of the expression of SpeB in the indicated GAS strains. g, h Primary keratinocytes infected or not with the indicated GAS strains for 2.5 h were analysed by phase-contrast microscopy (g), LDH release (h). i, j, A431 cells infected or not with GAS isolate M1T1 strain 5448 or its isogenic mutant strains for 2.5 h were analysed by phase-contrast microscopy (i) and LDH release (j). k, l, Equal amounts of recombinant of WT SpeB or protease activity-deficient mutant mSpeB were respectively electroporated into A431 cells for 1 h or directly added into cell culture medium for 2.5 h, followed by cell morphology observation by phase-contrast microscopy (k), cell viability assessment by CellTiter-Glo luminescent assay (l). g, i, Arrowheads indicate pyroptotic cells. c, d, g, i, k, scale bar: 10 μm. Graphs show mean ± s.d. of triplicate wells. h, j, One-way ANOVA; l, Two-tailed Student’s t-test. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 4 Validation of additional hits identified from CRISPR screen of SpeB-triggered lytic cell death.

a, List of top hits from CRISPR screen of SpeB-triggered lytic cell death. b, GSDMA-knockout cells used in this study. Gene coding sequences were present as black boxes. Top sequence track is the gene wild-type allele. Location of gRNAs is indicated with blue bars and PAM sequences were underscored. cl, WT and the indicated gene knockout A431 cells were transfected or not with recombinant SpeB by electroporation, followed by cell morphology observation by phase-contrast microscopy (c, e, g, i, k), cell viability assessment by CellTiter-Glo luminescent assay (d, f, h, j, l). Scale bar: 10 μm. Graphs show mean ± s.d. of triplicate wells. Data are representative of at least three independent experiments.

Source data

Extended Data Fig. 5 SpeB specifically targets and cleaves GSDMA.

a, b, In vitro cleavage assay of recombinant GSDMA, GSDMD (a) or GSDME (b) by incubation with recombinant WT SpeB (100 nM) or mSpeB (250 nM) for 0.5 h. c, Whole cell lysates of A431 cells infected or not with the indicated GAS strains were subjected to immunoblot analysis for the indicated proteins. df, 293T cells were transfected with the indicated plasmids (Flag-tagged GSDMA, Myc-tagged bacterial proteases) before analysed by phase-contrast microscopy (d), LDH release (e) and immunoblot of whole cell lysates with the indicated antibodies (f). g, h, Flag-tagged GSDMA was treated or not with SpeB, staphopains (ScpA, SspB), or cathepsins (Cathepsin B, L, D) before subjected to immunoblot analysis with the indicated antibodies. i, GSDMA N-terminal and C-terminal cleavage products (p27 and p23) were analysed by mass spectrometry (MS) and Edman sequencing, respectively. Individual peptides identified by MS and shown in black bars were mapped against N-terminal GSDMA (upper right panel). Middle right panel shows the N-terminal sequence of p23 determined by Edman sequencing, the diagram of GSDMA two-domain architecture, and SpeB cleavage site Gln246 highlighted in green. Bottom panel shows the positions (P) on the substrate of SpeB (GSDMA) which are counted and numbered (P3-P2-P1-P1’-P2’-P3’) from the point of cleavage. j, Whole cell lysates of 293T cells transfected with the indicated plasmids were collected and subjected to immunoblot analysis. Scale bar: 10 μm. Data are representative of at least three independent experiments. Graphs show mean ± s.d. of triplicate wells. e, One-way ANOVA. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 6 GSDMA-NT produced by SpeB cleavage is sufficient to initiate pyroptosis, but does not injure bystander cells.

a, Coomassie Blue-stained SDS-PAGE gel showing purified recombinant GSDMA, GSDMA-NT and GSDMA-CT. bd, Equal amounts of recombinant full-length GSDMA, GSDMA-NT (1–246aa), GSDMA-CT, full-length GSDMD, Caspase-11 or full-length GSDMD plus Caspase-11 were electroporated into 293T cells respectively, followed by cell morphology observation by phase-contrast microscopy (b), cell viability analysis by CellTiter-Glo luminescent assay (c), and cell death assessment by PI uptake (d). eg, Equal amounts of recombinant of full-length GSDMA, GSDMA-NT (1–246aa), GSDMA-CT, full-length GSDMD, Caspase-11 or full-length GSDMD plus Caspase-11 were respectively added directly into cell culture medium, followed by cell morphology observation by phase-contrast microscopy (e), cell viability analysis by CellTiter-Glo luminescent assay (f), and cell death assessment by PI uptake (g). Pyroptotic cells form large ballooning bubbles; Scale bar: 20 μm. Graphs show mean ± s.d. of triplicate wells (c, f) or mean ± s.e.m. of quadruplicate wells (d, g). c, Two-tailed Student’s t-test. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 7 GSDMA 1-246aa, but not 1-214aa possesses pyroptosis-inducing activity.

a, Coomassie Blue-stained SDS-PAGE gel showing recombinant engineered GSDMA with Flag tag-3C protease cleavage sequence inserted immediately after residue G214, Q246, treated with 3C protease. bd, 293T cells were transfected with the indicated plasmids (Empty vector or Flag-tagged Gasdermin) before cell death was observed by phase-contrast microscopy (b) and determined by LDH release assay (c), whole cell lysates were collected for immunoblot analysis (d). Arrowheads indicate pyroptotic cells; Scale bar: 10 μm. Graphs show mean ± s.d. of triplicate wells. c, Two-tailed Student’s t-test. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 8 Phospholipid binding property and liposome-disrupting activity of GSDMA-NT.

a, Lipid strips dotted with indicated phospholipids (left panel) were incubated with noncovalent complex of cleaved GSDMA with a Flag-tag inserted right before the cleavage site, unprocessed full-length GSDMA, or 3C protease, followed by immunoblot analysis with an anti-Flag antibody (right panel). b, Indicated liposomes incubated with noncovalent complex of cleaved GSDMA (NT + CT) and full-length GSDMA (FL) were subjected to sedimentation by ultracentrifugation. Proteins in both liposome-free supernatant (S) and liposome-containing pellet (P) were analysed by SDS-PAGE. c, Indicated recombinant proteins incubated or not with CL liposomes and glutaraldehyde were analyzed by SDS-agarose gel electrophoresis and subsequent Coomassie Blue staining. dh, Leakage of PC-PE liposomes containing additional PS or CL was monitored in real-time by terbium (Tb3+) fluorescence after incubation with recombinant gasdermins proteins in the presence or absence of recombinant SpeB or enzymatically inactive SpeB C192S (mSpeB) as indicated, or cysteine protease inhibitor E64 if necessary. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 9 GSDMA proteins in different species.

Multiple sequences alignment of human (h), chimpanzee (cp), monkey (mk), rat (r), dog (dg) GSDMA and mouse Gsdma1, Gsdma2 and Gsdma3 was performed using the ClustalW2 algorithm and plotted by ESPript program. Identical residues are highlighted by red background, and similar residues are indicated in red. Ile 245 and Gln246 (*) in human GSDMA are highly conserved in chimpanzee, monkey, rat, dog GSDMA as well as mouse Gsdma1.

Extended Data Fig. 10 SpeB cleaves mouse Gsdma1 and releases its N-terminal pyroptosis-inducing activity.

a, Whole cell lysates of 293T cells transfected with the indicated plasmids were collected and subjected to immunoblot analysis with the indicated antibodies. b, Coomassie Blue-stained SDS-PAGE gel showing in vitro cleavage of recombinant WT Gsdma1 or mutant Gsdma1 I246N/Q247E (1 μM) incubated with or without recombinant WT SpeB (100 nM) for 0.5 h. c, Whole cell lysates of 293T cells transfected with the indicated plasmids were collected and subjected to immunoblot analysis. d, Leakage of PC-PE liposomes containing additional PS or CL was monitored in real-time by terbium (Tb3+) fluorescence after incubation with recombinant gasdermins proteins in the presence or absence of recombinant SpeB as indicated. e, f, 293T cells transfected with the indicated plasmids were analysed by phase-contrast microscopy (e), LDH release (f). Arrowheads indicate pyroptotic cells; Scale bar: 10 μm. Graphs show mean ± s.d. of triplicate wells. f, One-way ANOVA. Data are representative of at least three independent experiments. For gel source data, see Supplementary Fig. 1.

Source data

Extended Data Fig. 11 Gsdma1 deficiency affects host immune responses against subcutaneous but not intraperitoneal infection of GAS WT.

a, WT and Gsdma1−/− mice were subcutaneously infected or not with GAS isolate M1T1 strain 5448 or its isogenic mutant strain (ΔspeB variant). IHC analysis of neutrophil infiltration at infection site on day 1. Scale bar: 100 μm. b, Quantification of neutrophil infiltration at infection site. c, Cutaneous sections from Gsdma1-/- mice infected or not for 18 h with FITC-labelled GAS WT were subjected to immunofluorescence staining with anti-keratin 14. Nucleus was stained with DAPI. Dashed lines delineate the boundaries of epidermis. Scale bar: 10 μm. d, Survival rate of mice intraperitoneally administrated with the indicated GAS (n = 12 mice per group). e, Model of SpeB-triggered GSDMA activation and subsequent pyroptosis of skin epithelial cells during GAS infection. b, show mean ± s.d. (n = 5 mice per group); Two-tailed Student’s t-test. Data are representative of at least three independent experiments.

Source data

Supplementary information

Supplementary Figure 1

Uncropped blots used to prepare main and extended data figures.

Reporting Summary

Supplementary Table 1

Oligonucleotide primers used in this study for cloning DNA fragments containing cepA, mac, covR/S, speB, emm, Rgg and RocA genes from the GAS genome.

Source data

Rights and permissions

Springer Nature or its licensor 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

Deng, W., Bai, Y., Deng, F. et al. Streptococcal pyrogenic exotoxin B cleaves GSDMA and triggers pyroptosis. Nature 602, 496–502 (2022). https://doi.org/10.1038/s41586-021-04384-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-021-04384-4

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology