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:

Host extracellular vesicles confer cytosolic access to systemic LPS licensing non-canonical inflammasome sensing and pyroptosis

Abstract

Intracellular surveillance for systemic microbial components during homeostasis and infections governs host physiology and immunity. However, a long-standing question is how circulating microbial ligands become accessible to intracellular receptors. Here we show a role for host-derived extracellular vesicles (EVs) in this process; human and murine plasma-derived and cell culture-derived EVs have an intrinsic capacity to bind bacterial lipopolysaccharide (LPS). Remarkably, circulating host EVs capture blood-borne LPS in vivo, and the LPS-laden EVs confer cytosolic access for LPS, triggering non-canonical inflammasome activation of gasdermin D and pyroptosis. Mechanistically, the interaction between the lipid bilayer of EVs and the lipid A of LPS underlies EV capture of LPS, and the intracellular transfer of LPS by EVs is mediated by CD14. Overall, this study demonstrates that EVs capture and escort systemic LPS to the cytosol licensing inflammasome responses, uncovering EVs as a previously unrecognized link between systemic microbial ligands and intracellular surveillance.

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: Circulating host-derived EVs capture LPS.
Fig. 2: Host-derived EVs bind LPS in vivo.
Fig. 3: Cytosolic delivery of LPS by host-derived EVs.
Fig. 4: EV-mediated cytosolic LPS delivery activates the non-canonical inflammasome.
Fig. 5: The effect of EV depletion on intracellular LPS sensing.
Fig. 6: The EV lipid bilayer–lipid A interaction underlies EV capture of LPS.
Fig. 7: EV-mediated cytosolic delivery of LPS is CD14 dependent.

Similar content being viewed by others

Data availability

Uncropped immunoblot images for this article are included in the Supplementary Information. Source data are provided with this paper. All other data supporting the findings of the paper are available from the corresponding author upon reasonable request.

References

  1. Gabanyi, I. et al. Bacterial sensing via neuronal Nod2 regulates appetite and body temperature. Science 376, eabj3986 (2022).

    CAS  PubMed  Google Scholar 

  2. Clarke, T. B. et al. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat. Med. 16, 228–231 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Farrokhi, V. et al. Bacterial lipodipeptide, Lipid 654, is a microbiome-associated biomarker for multiple sclerosis. Clin. Transl. Immunol. 2, e8 (2013).

    CAS  Google Scholar 

  4. Huang, Z. et al. Antibody neutralization of microbiota-derived circulating peptidoglycan dampens inflammation and ameliorates autoimmunity. Nat. Microbiol. 4, 766–773 (2019).

    CAS  PubMed  Google Scholar 

  5. Moltke von, J., Ayres, J. S., Kofoed, E. M., Chavarría-Smith, J. & Vance, R. E. Recognition of bacteria by inflammasomes. Annu Rev. Immunol. 31, 73–106 (2013).

    Google Scholar 

  6. Kawai, T. & Akira, S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat. Immunol. 11, 373–384 (2010).

    CAS  PubMed  Google Scholar 

  7. Poltorak, A. et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: mutations in Tlr4 gene. Science 282, 2085–2088 (1998).

    CAS  PubMed  Google Scholar 

  8. Rathinam, V. A. K., Zhao, Y. & Shao, F. Innate immunity to intracellular LPS. Nat. Immunol. 20, 527–533 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Kayagaki, N. et al. Non-canonical inflammasome activation by intracellular LPS independent of TLR4. Science 341, 1246–1249 (2013).

    CAS  PubMed  Google Scholar 

  10. Hagar, J. A., Powell, D. A., Aachoui, Y., Ernst, R. K. & Miao, E. A. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science 341, 1250–1253 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Shi, J. et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514, 187 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  14. Vanaja, S. K. et al. Bacterial outer membrane vesicles mediate cytosolic localization of LPS and caspase-11 activation. Cell 165, 1106–1119 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Luo, Z. et al. Variation in blood microbial lipopolysaccharide (LPS) contributes to immune reconstitution in response to suppressive antiretroviral therapy in HIV. EBioMedicine 80, 104037 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Mohammad, S. & Thiemermann, C. Role of metabolic endotoxemia in systemic inflammation and potential interventions. Front. Immunol. 11, 594150 (2021).

    PubMed  PubMed Central  Google Scholar 

  17. Kell, D. B. & Pretorius, E. On the translocation of bacteria and their lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integr. Biol. 7, 1339–1377 (2015).

    CAS  Google Scholar 

  18. Prins, J. M., Deventer van, S. J., Kuijper, E. J. & Speelman, P. Clinical relevance of antibiotic-induced endotoxin release. Antimicrob. Agents Chemother. 38, 1211–1218 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Matsuura, M. Structural modifications of bacterial lipopolysaccharide that facilitate Gram-negative bacteria evasion of host innate immunity. Front Immunol. 4, 109 (2013).

    PubMed  PubMed Central  Google Scholar 

  20. Fukui, H. Endotoxin and other microbial translocation markers in the blood: a clue to understand leaky gut syndrome. Cell. Mol. Med. Open Access https://doi.org/10.21767/2573-5365.100023 (2016).

  21. Vasudevan, S. O., Russo, A. J., Kumari, P., Vanaja, S. K. & Rathinam, V. A. A TLR4-independent critical role for CD14 in intracellular LPS sensing. Cell Rep. 39, 110755 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Colombo, M., Raposo, G. & Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 30, 1–35 (2014).

    Google Scholar 

  23. Jeppesen, D. K. et al. Reassessment of exosome composition. Cell 177, 428–445.e18 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kalluri, R. & LeBleu, V. S. The biology, function, and biomedical applications of exosomes. Science 367, eaau6977 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. O’Brien, K., Breyne, K., Ughetto, S., Laurent, L. C. & Breakefield, X. O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 21, 585–606 (2020).

    PubMed  PubMed Central  Google Scholar 

  26. Kim, D.-K. et al. EVpedia: a community web portal for extracellular vesicles research. Bioinformatics 31, 933–939 (2015).

    CAS  PubMed  Google Scholar 

  27. Wang, N. et al. Circulating exosomes contain protein biomarkers of metastatic non‐small‐cell lung cancer. Cancer Sci. 109, 1701–1709 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Li, W. et al. LPS induces active HMGB1 release from hepatocytes into exosomes through the coordinated activities of TLR4 and caspase-11/GSDMD signaling. Front. Immunol. 11, 229 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Cosme, J., Guo, H., Hadipour-Lakmehsari, S., Emili, A. & Gramolini, A. O. Hypoxia-induced changes in the fibroblast secretome, exosome, and whole-cell proteome using cultured, cardiac-derived cells isolated from neonatal mice. J. Proteome Res. 16, 2836–2847 (2017).

    CAS  PubMed  Google Scholar 

  30. Jin, M., Drwal, G., Bourgeois, T., Saltz, J. & Wu, H. M. Distinct proteome features of plasma microparticles. Proteomics 5, 1940–1952 (2005).

    CAS  PubMed  Google Scholar 

  31. Kugeratski, F. G. et al. Quantitative proteomics identifies the core proteome of exosomes with syntenin-1 as the highest abundant protein and a putative universal biomarker. Nat. Cell Biol. 23, 631–641 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Budden, C. F. et al. Inflammasome‐induced extracellular vesicles harbour distinct RNA signatures and alter bystander macrophage responses. J. Extracell. Vesicles 10, e12127 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Lobb, R. J. et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 4, 27031 (2015).

    PubMed  Google Scholar 

  34. Mathieu, M., Martin-Jaular, L., Lavieu, G. & Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 21, 9–17 (2019).

    CAS  PubMed  Google Scholar 

  35. Jeppesen, D. K., Zhang, Q., Franklin, J. L. & Coffey, R. J. Extracellular vesicles and nanoparticles: emerging complexities. Trends Cell Biol. https://doi.org/10.1016/j.tcb.2023.01.002 (2023).

    Article  PubMed  Google Scholar 

  36. Cheng, K. T. et al. Caspase-11–mediated endothelial pyroptosis underlies endotoxemia-induced lung injury. J. Clin. Invest. 127, 4124–4135 (2017).

    PubMed  PubMed Central  Google Scholar 

  37. Kumari, P., Russo, A. J., Wright, S. S., Muthupalani, S. & Rathinam, V. A. Hierarchical cell-type-specific functions of caspase-11 in LPS shock and antibacterial host defense. Cell Rep. 35, 109012 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Russo, A. J. et al. Intracellular immune sensing promotes inflammation via gasdermin D-driven release of a lectin alarmin. Nat. Immunol. 22, 154–165 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Gong, T., Liu, L., Jiang, W. & Zhou, R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat. Rev. Immunol. 20, 95–112 (2020).

    CAS  PubMed  Google Scholar 

  40. Catalano, M. & O’Driscoll, L. Inhibiting extracellular vesicles formation and release: a review of EV inhibitors. J. Extracell. Vesicles 9, 1703244 (2020).

    CAS  PubMed  Google Scholar 

  41. Zhang, H., Lu, J., Liu, J., Zhang, G. & Lu, A. Advances in the discovery of exosome inhibitors in cancer. J. Enzyme Inhib. Med. Chem. 35, 1322–1330 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Lu, M. et al. Comparison of exosome-mimicking liposomes with conventional liposomes for intracellular delivery of siRNA. Int. J. Pharm. 550, 100–113 (2018).

    CAS  PubMed  Google Scholar 

  43. Raetz, C. R. H., Reynolds, C. M., Trent, M. S. & Bishop, R. E. Lipid A modification systems in Gram-negative bacteria. Annu. Rev. Biochem. 76, 295–329 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

    CAS  PubMed  Google Scholar 

  45. Zanoni, I. et al. CD14 controls the LPS-induced endocytosis of Toll-like receptor 4. Cell 147, 868–880 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Wright, S. D., Ramos, R. A., Tobias, P. S., Ulevitch, R. J. & Mathison, J. C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 249, 1431 1433 (1990).

    PubMed  Google Scholar 

  47. Kitchens, R. L. & Munford, R. S. CD14-dependent internalization of bacterial lipopolysaccharide (LPS) is strongly influenced by LPS aggregation but not by cellular responses to LPS. J. Immunol. 160, 1920–1928 (1998).

    CAS  PubMed  Google Scholar 

  48. Tan, Y., Zanoni, I., Cullen, T. W., Goodman, A. L. & Kagan, J. C. Mechanisms of Toll-like receptor 4 endocytosis reveal a common immune-evasion strategy used by pathogenic and commensal bacteria. Immunity 43, 909–922 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Keller, M. D. et al. Decoy exosomes provide protection against bacterial toxins. Nature 579, 260–264 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Robbins, P. D. & Morelli, A. E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 14, 195–208 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Théry, C., Ostrowski, M. & Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 9, 581–593 (2009).

    PubMed  Google Scholar 

  52. Vreugdenhil, A. C. E. et al. Lipopolysaccharide (LPS)-binding protein mediates LPS detoxification by chylomicrons. J. Immunol. 170, 1399–1405 (2003).

    CAS  PubMed  Google Scholar 

  53. Vreugdenhil, A. C., Snoek, A. M., van ′t Veer, C., Greve, J. W. & Buurman, W. A. LPS-binding protein circulates in association with apoB-containing lipoproteins and enhances endotoxin-LDL/VLDL interaction. J. Clin. Invest. 107, 225–234 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Berbée, J. F. P., Havekes, L. M. & Rensen, P. C. N. Apolipoproteins modulate the inflammatory response to lipopolysaccharide. J. Endotoxin Res. 11, 97–103 (2005).

    PubMed  Google Scholar 

  55. Yao, Z. et al. Blood-borne lipopolysaccharide is rapidly eliminated by liver sinusoidal endothelial cells via high-density lipoprotein. J. Immunol. 197, 2390–2399 (2016).

    CAS  PubMed  Google Scholar 

  56. Topchiy, E. et al. Lipopolysaccharide is cleared from the circulation by hepatocytes via the low-density lipoprotein receptor. PLoS ONE 11, e0155030 (2016).

    PubMed  PubMed Central  Google Scholar 

  57. Deng, M. et al. The endotoxin delivery protein HMGB1 mediates caspase-11-dependent lethality in sepsis. Immunity 49, 740–753.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  59. Lamkanfi, M. et al. Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J. Immunol. 185, 4385–4392 (2010).

    CAS  PubMed  Google Scholar 

  60. Kayagaki, N. et al. NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 591, 131–136 (2021).

    CAS  PubMed  Google Scholar 

  61. Minihane, A. M. et al. Low-grade inflammation, diet composition and health: current research evidence and its translation. Br. J. Nutr. 114, 999–1012 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Glaros, T. G. et al. Causes and consequences of low-grade endotoxemia and inflammatory diseases. Front. Biosci. 5, 754–765 (2013).

    Google Scholar 

  63. Geng, S. et al. The persistence of low-grade inflammatory monocytes contributes to aggravated atherosclerosis. Nat. Commun. 7, 13436 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Pasternak, B. A. et al. Lipopolysaccharide exposure is linked to activation of the acute phase response and growth failure in pediatric Crohn’s disease and murine colitis. Inflamm. Bowel Dis. 16, 856–869 (2010).

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank V. Dixit and K. Fitzgerald for Casp11−/− mice and N. Frank for the HEK-Blue TLR4 reporter cell line. This work was supported by the National Institutes of Health grant nos. R01AI119015 and R01AI148491 (V.A.R.) and R01AI132850 (S.K.V.). V.F.-Á., I.R. and M.B. were supported by the Deutsche Forschungsgemeinschaft (German Research Foundation) under Germany’s Excellence Strategy (EXC 2051: Balance of the Microverse, project number 390713860). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

V.A.R., P.K. and S.K.V. conceived the study. V.A.R., P.K., S.O.V. and S.K.V. designed the experiments and wrote the paper. P.K., S.O.V., A.J.R., S.S.W., V.F.-Á., D.K., E.R.J., I.R., M.B. and J.S.P. performed experiments, analysed data or provided technical or conceptual help. K.F. and A.S. provided biotin–lipid A, and Y.Z. provided EV mimic liposomes.

Corresponding author

Correspondence to Vijay A. Rathinam.

Ethics declarations

Competing interests

Y.Z.’s company FormuMax is interested in the sales of exosome-mimicking liposomes. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Xuetao Cao, Raghu Kalluri and Rongbin Zhou 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 Host-derived EVs bind LPS in vivo.

a, Plasma EV levels in wild-type (WT) mice injected with PBS or LPS (10 mg/kg) for indicated times as assessed by nanoparticle tracking analysis (NTA). b, Size distribution of EVs isolated from the plasma of WT mice injected with PBS or LPS (25 mg/kg) 1.5 h post-injection via OptiPrep density gradient method as assessed by NTA. c, Immunoblotting analysis of EVs isolated from the plasma of PBS- or LPS-injected WT mice as in (b). d, Negative staining TEM of EVs isolated from the plasma of PBS or LPS-injected WT mice as in (b). e, LPS content of the EVs isolated from WT mice injected with PBS or LPS as assessed by the LAL assay (n = 4). f,g, Percentage of FITC-LPS+ve EVs (f) and FITC histogram and mean fluorescence intensity (MFI) of EVs (g) isolated from mice injected with PBS or FITC-LPS (25 mg/kg) 1.5 h post-injection as assessed by ImageStream flow cytometry (n = 3). h, Immunoblotting analysis of EVs isolated by the DIC method using CD9, CD63, and CD81 antibodies-coated magnetic beads from the plasma of PBS- or FITC-LPS (25 mg/kg)-injected WT mice. i,j, LPS content of the DIC-isolated EVs from PBS- or FITC-LPS-injected mice 1.5 h post-injection as assessed by the LAL (n = 3) (i) and HEK-Blue TLR4 reporter cell (n = 3) (j) assays. k,l, Percentage of FITC-LPS+ve EVs (k) and FITC histogram and mean fluorescence intensity (MFI) of EVs (l) isolated by the DIC method from mice injected with PBS or FITC-LPS as assessed by flow cytometry (n = 3). Combined data from two (a) or three (e,f,g,i,j,k,l) or representative data of two (b,c,d,h) independent experiments are shown. Data are shown as mean±s.e.m (a). Each circle represents a mouse, and the horizontal lines represent mean (a, eg and il). P values were determined by one-way ANOVA with Dunnett’s post- test (a) or unpaired two-tailed t-test (eg and il). Scale bar, 50 nm (d).

Source data

Extended Data Fig. 2 Microvesicles bind LPS in vivo.

(a–b) Percentage of FITC-LPS+ve microvesicles (MVs; annexin A1+ve) and LPS content of MVs isolated from Casp11−/− mice injected with PBS or FITC-LPS via ultracentrifugation as assessed by ImageStream flow cytometry (a) and the LAL assay (b). Each circle represents pooled sample from two mice. Combined data from two independent experiments are shown.

Source data

Extended Data Fig. 3 Plasma EVs bind LPS independent of blood components.

a, LPS content of the EVs isolated by the SEC method from the plasma incubated with PBS or FITC-LPS (500 μg) at 37 °C for 45 min as assessed by the LAL assay (n = 3). b,c, Percentage of FITC-LPS+ve EVs (b) and FITC histogram and mean fluorescence intensity (MFI) of EVs (c) isolated by the SEC method from the plasma incubated with PBS or FITC-LPS in vitro as assessed by ImageStream flow cytometry (n = 3). df and hj, EVs isolated from the plasma by the SEC method were incubated with PBS, FITC-LPS, or biotin-LPS as indicated at 37 °C for 45 min in vitro, and the LPS binding was assessed by the LAL assay (n = 3 or 6 as indicated) (d,h) and ImageStream flow cytometry (n = 3 or 6 as indicated) (e,f,i,j). g, TEM of EVs isolated and treated as described above and stained with anti-FITC-gold particles. Arrows indicate LPS. Combined data from three (a–c and h–j) or six (d–f) experiments or one representative of two experiments (g) are shown. Each circle represents a mouse, and the horizontal lines represent the mean in a–f and h–j. P values were determined by unpaired two-tailed t-test. Scale bar, 50 nm (g).

Source data

Extended Data Fig. 4 Human plasma EVs and endothelial and epithelial cell EVs bind LPS.

a–h, EVs isolated from the human plasma by the SEC method were incubated with PBS, FITC-LPS or biotin-LPS as indicated at 37 °C for 45 min, and the LPS binding was assessed by the LAL (a,e) and HEK-Blue TLR4 reporter cell (b,f) assays and ImageStream flow cytometry (c,d,g,h) (n = 3). i–p, EVs isolated from bEnd.3 endothelial (i–l) and HeLa (m–p) cells cultured in serum-free conditions by the UC method were incubated with PBS or FITC-LPS (500 μg) as indicated at 37 °C for 45 min followed by re-purification via SEC, and the LPS binding was assessed by the LAL (i,m) and HEK-Blue TLR4 reporter cell (j,n) assays and ImageStream flow cytometry (k,l,o,p) (n = 3). Combined data from three experiments (a–p) are shown. Each circle represents an independent experiment, and the horizontal lines represent the mean. P values were determined by unpaired two-tailed t-test (a–p).

Source data

Extended Data Fig. 5 Intracellular localization of LPS is EV-dependent.

(a–d) Confocal microscopy of Casp11−/− iBMDMs stimulated for 4 h with unlabeled EVs (a), FITC-LPS (b), FITC-LPS-EVs (c), or CellBrite Steady Membrane 488-labeled-EVs (d) and stained with anti-CD45, anti-EEA1 and anti-FITC antibodies to visualize plasma membrane, early endosomes and LPS, respectively. Arrows indicate intracellular localization of EV-bound FITC-LPS (c) or CellBrite 488-labeled-EVs (d). Images representative of two experiments are shown. Scale bar: 5 μm.

Extended Data Fig. 6 LPS-laden host EVs induce endolysosomal membrane disruption.

Confocal images of WT BMDMs stimulated with LPS or LPS-EVs for 6 h and stained for galectin-3 (red), lamp1 (yellow; a), and Rab5 (blue; b). Scale bar: 5 μM. White arrows indicate the colocalization of galectin-3 and lamp1 or Rab5. Images representative of two experiments are shown.

Extended Data Fig. 7 EV-bound LPS activates the noncanonical inflammasome.

a,b, GSDMD and caspase-4 in lysates (a) and cell death (b) of IFN-γ-primed (10 ng/ml) THP1 monocytes stimulated as indicated for 16 h. c,d, Cleaved IL-1β (p17) and caspase-1 (p20) in the supernatants and indicated proteins in lysates of (c) and IL-1β secretion (d) by IFN-γ-primed WT and Casp11−/− BMDMs stimulated as indicated for 16 h. e–h, GSDMD, caspase-11 and β-actin in lysates (e,g) and cell death (f,h) of IFN-γ-primed indicated BMDMs stimulated as indicated for 16 h. i,j, Indicated proteins in lysates (i) and cell death (j) of IFN-γ-primed RAW macrophages stimulated as indicated for 16 h. k, IL-6 and TNF secretion by indicated BMDMs stimulated with PBS-EVs, LPS-EVs (2.5 μg LPS/ml) or LPS (2.5 μg/ml) for 16 h. l, Plasma IL-6 and TNF in indicated mice administered with LPS-EVs for 6 h (n = 6). m, Survival of poly(I:C)-primed WT and Tlr4−/− mice injected with LPS-EVs (n = 6). n, Baseline IL-1β and IL-18 in the plasma of WT mice injected with DMSO (vehicle) or GW4869 (2.5 μg/g) twice 24 h apart. DMSO-treated LPS-injected mice are shown as controls (n = 6). o, Survival of Tlr4−/− mice injected with DMSO or GW4869 (2.5 μg/g) on days 1–3, primed with poly(I:C) on day 3 and injected with LPS (25 μg/g) 6 h later (n = 5). p, Survival of indicated mice injected with DMSO or GW4869 (2.5 μg/g) on days 1–3 and injected with LPS (25 μg/g) on day 3 (n = 6). q–s, GSDMD, caspase-11 and β-actin in the liver and spleen (q) and IL-18 and IL-1β in the plasma (r,s; n = 6) of WT mice pretreated with DMSO (2.5 μg/g) or Nexinhib20 (25 μg/g) on days 1 and 2 and injected with LPS (25 μg/g) on day 3 for 8 h. Each circle represents a mouse, and the horizontal lines represent mean (l,n,r,s). Each lane represents a mouse (q). Data are presented as mean±s.e.m (b,d,f,h,j,k). Combined data from two (k–p,r,s) or three (b,d,f,h,j) experiments or one experiment representative of two (a,c,e,g,i,q) are shown. P values were determined by one-way ANOVA with Dunnett’s post-test (b,j,n), two-way ANOVA with Sidak’s post-test (d,f,h,k), unpaired two-tailed t-test (l,r,s) or Mantel-Cox test (m,o,p).

Source data

Extended Data Fig. 8 EV binding of LPS is not mediated by common LPS binding proteins.

a–d, LPS binding by EVs preincubated with isotype control, anti-HMGB1 or anti-LBP antibodies (Ab) and incubated with FITC-LPS (500 μg) at 37 °C for 45 min as assessed by ImageStream flow cytometry (a,c) and the HEK-Blue TLR4 reporter cell assay (b,d). Combined data from three experiments are shown, and the horizontal lines represent mean. P values were determined by unpaired two-tailed t-test.

Source data

Extended Data Fig. 9 EV binding of LPS is not mediated by EV surface proteins.

a,b, Immunoblotting (a) and negative staining TEM (b) of unshaved EVs and EVs subjected to surface protein shaving with trypsin (shaved EVs). c,d, Unshaved and shaved EVs incubated with FITC-LPS at 37 °C for 45 min were subjected to NTA (c), and the HEK-Blue TLR4 reporter cell assay (d). e,f, Percentage of FITC-LPS+ve EVs (e) and FITC MFI (f) of EVs isolated from FITC-LPS-injected mice and subjected or not to surface-protein shaving. Combined data from three (c,d) or four (e,f) experiments or one experiment representative of two (a,b) are shown, and horizontal lines represent mean (c–f). P values were determined by unpaired two-tailed t-test.

Source data

Extended Data Fig. 10 LPS-bound EVs colocalize with CD14.

a,b, Casp11−/− iBMDMs were stimulated with FITC-LPS-EVs unlabeled (a) or labeled with CellBrite Steady 550 dye (b). After 4 h, cells were stained with anti-CD14, anti-EEA1, and anti-FITC antibodies and subjected to confocal imaging. In (b), light blue arrows indicate colocalization of FITC-LPS-EVs (CellBrite-labeled) with CD14 (top two rows) and their internalization (bottom two rows). Images are representative of two experiments. Scale bar, 5 µm (a,b).

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 Fig. 7

Statistical source data.

Source Data Fig. 1

Uncropped western blots.

Source Data Fig. 2

Uncropped western blots.

Source Data Fig. 4

Uncropped western blots.

Source Data Fig. 5

Uncropped western blots.

Source Data Fig. 7

Uncropped western blots.

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. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 1

Uncropped western blots.

Source Data Extended Data Fig. 7

Uncropped western blots.

Source Data Extended Data Fig. 9

Uncropped western 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

Kumari, P., Vasudevan, S.O., Russo, A.J. et al. Host extracellular vesicles confer cytosolic access to systemic LPS licensing non-canonical inflammasome sensing and pyroptosis. Nat Cell Biol 25, 1860–1872 (2023). https://doi.org/10.1038/s41556-023-01269-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-023-01269-8

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