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:

Intracellular immune sensing promotes inflammation via gasdermin D–driven release of a lectin alarmin

Abstract

Inflammatory caspase sensing of cytosolic lipopolysaccharide (LPS) triggers pyroptosis and the concurrent release of damage-associated molecular patterns (DAMPs). Collectively, DAMPs are key determinants that shape the aftermath of inflammatory cell death. However, the identity and function of the individual DAMPs released are poorly defined. Our proteomics study revealed that cytosolic LPS sensing triggered the release of galectin-1, a β-galactoside-binding lectin. Galectin-1 release is a common feature of inflammatory cell death, including necroptosis. In vivo studies using galectin-1-deficient mice, recombinant galectin-1 and galectin-1-neutralizing antibody showed that galectin-1 promotes inflammation and plays a detrimental role in LPS-induced lethality. Mechanistically, galectin-1 inhibition of CD45 (Ptprc) underlies its unfavorable role in endotoxin shock. Finally, we found increased galectin-1 in sera from human patients with sepsis. Overall, we uncovered galectin-1 as a bona fide DAMP released as a consequence of cytosolic LPS sensing, identifying a new outcome of inflammatory cell death.

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: Proteomic identification of intracellular LPS-elicited release of galectin-1.
Fig. 2: Cytosolic LPS-induced galectin-1 release in vitro is dependent on caspase-11/4 and GSDMD.
Fig. 3: Cytosolic LPS-induced release of galectin-1 is dependent on caspase-11 and GSDMD in vivo.
Fig. 4: Necroptosis triggers galectin-1 release.
Fig. 5: Galectin-1 plays a detrimental role during LPS septic shock.
Fig. 6: Galectin-1 amplifies systemic inflammatory responses during LPS shock.
Fig. 7: Galectin-1 inhibition of CD45 underlies its detrimental role in endotoxin shock.

Similar content being viewed by others

Data availability

The RNA-seq data have been deposited with the Gene Expression Omnibus (accession number GSE140892). Source data and uncropped immunoblot images are included in the paper as supplementary information. All other data supporting the findings of the paper are available from the corresponding author upon reasonable request.

References

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  4. Shi, J. et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514, 187–192 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Kayagaki, N. et al. Noncanonical inflammasome activation by intracellular LPS independent of TLR4. Science 341, 1246–1249 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  10. Kang, R. et al. Lipid peroxidation drives gasdermin D-mediated pyroptosis in lethal polymicrobial sepsis. Cell Host Microbe 24, 97–108.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  12. Song, F. et al. Sphingosine-1-phosphate receptor 2 signaling promotes caspase-11–dependent macrophage pyroptosis and worsens Escherichia coli sepsis outcome. Anesthesiology 129, 311–320 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  14. Wang, H. et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 285, 248–251 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Keller, M., Rüegg, A., Werner, S. & Beer, H.-D. Active caspase-1 is a regulator of unconventional protein secretion. Cell 132, 818–831 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Sundblad, V., Morosi, L. G., Geffner, J. R. & Rabinovich, G. A. Galectin-1: a jack-of-all-trades in the resolution of acute and chronic inflammation. J. Immunol. 199, 3721–3730 (2017).

    Article  CAS  PubMed  Google Scholar 

  17. Camby, I., Le Mercier, M., Lefranc, F. & Kiss, R. Galectin-1: a small protein with major functions. Glycobiology 16, 137R–157R (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Lee, H.-J. et al. Application of a peptide‐based PF2D platform for quantitative proteomics in disease biomarker discovery. Proteomics 8, 3371–3381 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Banerjee, I. et al. Gasdermin D restrains type I interferon response to cytosolic DNA by disrupting ionic homeostasis. Immunity 49, 413–426.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Carta, S., Lavieri, R. & Rubartelli, A. Different members of the IL-1 family come out in different ways: DAMPs vs. cytokines? Front. Immunol. 4, 123 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Piccioli, P. & Rubartelli, A. The secretion of IL-1β and options for release. Semin. Immunol. 25, 425–429 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Evavold, C. L. et al. The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages. Immunity 48, 35–44.e6 (2018).

    Article  CAS  PubMed  Google Scholar 

  23. Chen, Y. et al. Gasdermin D drives the nonexosomal secretion of galectin-3, an insulin signal antagonist. J. Immunol. 203, 2712–2723 (2019).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Frank, D. & Vince, J. E. Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 26, 99–114 (2019).

    Article  PubMed  Google Scholar 

  26. Galluzzi, L., Kepp, O., Chan, F. K.-M. & Kroemer, G. Necroptosis: mechanisms and relevance to disease. Annu. Rev. Pathol. 12, 103–130 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Delano, M. J. & Ward, P. A. Sepsis-induced immune dysfunction: can immune therapies reduce mortality? J. Clin. Invest. 126, 23–31 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Zhang, W. & Coopersmith, C. M. Dying as a pathway to death in sepsis. Anesthesiology 129, 238–240 (2018).

    Article  PubMed  Google Scholar 

  29. Mandal, P. et al. Caspase-8 collaborates with caspase-11 to drive tissue damage and execution of endotoxic shock. Immunity 49, 42–55.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Schenck, E. J. et al. Circulating cell death biomarker TRAIL is associated with increased organ dysfunction in sepsis. JCI Insight 4, e127143 (2019).

    Article  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  32. Rabinovich, G. A. & Toscano, M. A. Turning ‘sweet’ on immunity: galectin–glycan interactions in immune tolerance and inflammation. Nat. Rev. Immunol. 9, 338–352 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Earl, L. A., Bi, S. & Baum, L. G. N- and O-glycans modulate galectin-1 binding, CD45 signaling, and T cell death. J. Biol. Chem. 285, 2232–2244 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Walzel, H., Schulz, U., Neels, P. & Brock, J. Galectin-1, a natural ligand for the receptor-type protein tyrosine phosphatase CD45. Immunol. Lett. 67, 193–202 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Irie-Sasaki, J. et al. CD45 is a JAK phosphatase and negatively regulates cytokine receptor signalling. Nature 409, 349–354 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Piercy, J., Petrova, S., Tchilian, E. Z. & Beverley, P. C. L. CD45 negatively regulates tumour necrosis factor and interleukin-6 production in dendritic cells. Immunology 118, 250–256 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zanoni, I. et al. An endogenous caspase-11 ligand elicits interleukin-1 release from living dendritic cells. Science 352, 1232–1236 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wolf, A. J. et al. Hexokinase is an innate immune receptor for the detection of bacterial peptidoglycan. Cell 166, 624–636 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lei, T. et al. Galectin-1 enhances TNFα-induced inflammatory responses in Sertoli cells through activation of MAPK signalling. Sci. Rep. 8, 3741 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Pérez, C. V. et al. Dual roles of endogenous and exogenous galectin-1 in the control of testicular immunopathology. Sci. Rep. 5, 12259 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Toegel, S. et al. Galectin-1 couples glycobiology to inflammation in osteoarthritis through the activation of an NF-κB-regulated gene network. J. Immunol. 196, 1910–1921 (2016).

    Article  CAS  PubMed  Google Scholar 

  42. Starossom, S. C. et al. Galectin-1 deactivates classically activated microglia and protects from inflammation-induced neurodegeneration. Immunity 37, 249–263 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Huang, X.-T. et al. Galectin-1 ameliorates lipopolysaccharide-induced acute lung injury via AMPK-Nrf2 pathway in mice. Free Radic. Biol. Med. 146, 222–233 (2020).

    Article  CAS  PubMed  Google Scholar 

  44. Ge, X. N. et al. Regulation of eosinophilia and allergic airway inflammation by the glycan-binding protein galectin-1. Proc. Natl Acad. Sci. USA 113, E4837–E4846 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Rodrigues, L. C. et al. Galectin-1 modulation of neutrophil reactive oxygen species production depends on the cell activation state. Mol. Immunol. 116, 80–89 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Croci, D. O. et al. Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors. Cell 156, 744–758 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Di Lella, S. et al. When galectins recognize glycans: from biochemistry to physiology and back again. Biochemistry 50, 7842–7857 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Poirier, F. & Robertson, E. J. Normal development of mice carrying a null mutation in the gene encoding the L14 S-type lectin. Development 119, 1229–1236 (1993).

    Article  CAS  PubMed  Google Scholar 

  49. Martínez Allo, V. C. et al. Suppression of age-related salivary gland autoimmunity by glycosylation-dependent galectin-1-driven immune inhibitory circuits. Proc. Natl Acad. Sci. USA 117, 6630–6639 (2020).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  50. Rathinam, V. A. K. et al. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by Gram-negative bacteria. Cell 150, 606–619 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ménoret, A. et al. The oxazolidinone derivative locostatin induces cytokine appeasement. J. Immunol. 183, 7489–7496 (2009).

    Article  PubMed  CAS  Google Scholar 

  52. Fettis, M. M. & Hudalla, G. A. Engineering reactive oxygen species-resistant galectin-1 dimers with enhanced lectin activity. Bioconjug. Chem. 29, 2489–2496 (2018).

    Article  CAS  PubMed  Google Scholar 

  53. Pérez Sáez, J. M. et al. Characterization of a neutralizing anti-human galectin-1 monoclonal antibody with angioregulatory and immunomodulatory activities. Angiogenesis https://doi.org/10.1007/s10456-020-09749-3 (2020).

  54. Bone, R. C. et al. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Chest 101, 1644–1655 (1992).

    Article  CAS  PubMed  Google Scholar 

  55. Singer, M. et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 315, 801–810 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Conejo-Garcia for the Lgals1−/− mice and V. Dixit (Genentech) and K. Fitzgerald (University of Massachusetts Medical School) for the Casp11−/− and Gsdmd−/− mice, S. Yeung and K. Khanna for help with the multiplex analysis and G. Fong for the MS1 cell line (University of Connecticut Health). This work was supported by the National Institutes of Health (grant nos. AI119015 and AI135528 to V.A.R.); Agencia Nacional de Promoción Científica y Tecnológica, Fundación Sales and Fundación Bunge y Born to G.A.R.; the Federal Ministry of Education and Research, Germany (grant no. 01EO1502) and Deutsche Forschungsgemeinschaft (German Research Foundation) under Germany’s Excellence Strategy-EXC 2051, project ID 390713860 (to M.B.).

Author information

Authors and Affiliations

Authors

Contributions

V.A.R. and A.J.R. conceived the study. V.A.R., G.A.R. and A.J.R. designed the experiments and wrote the manuscript. A.J.R., S.O.V., A.M., S.P.M.H., P.K., J.M.P.S., C.W., K.C., S.K.V., J.R., G.A.R. and A.T.V. performed the experiments, analyzed the data or provided technical or conceptual help. S.D., S.D.D., M.B. and C.S. performed the human sample analysis. M.M.F., R.L., A.W. and G.A.H. provided the recombinant galectin-1 and GFP. B.Z. and C.L. performed the RNA-seq analysis.

Corresponding author

Correspondence to Vijay A. Rathinam.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Derek Abbott and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Galectin-1 is released as a consequence of inflammasome activation.

a, Experimental design for profiling caspase-11-mediated release of DAMPs by PF2D-based proteomics. WT and Casp11–/– BMDMs were infected with EHEC (MOI=50) for 16 h and supernatants were harvested and concentrated and processed on the Beckman Coulter ProteomeLab PF2D platform. b, Galectin-1 secretion by Pam3CSK4-primed WT and Lgals1–/– BMDMs stimulated with EHEC (MOI=50) or poly(dA:dT) transfection for 16 h or 10 µM nigericin for 1 h. c, Immunoblot of galectin-1 (14.5 kDa) in the supernatant and lysates of BMDMs stimulated as in (b). d, IL-1α secretion by Pam3CSK4-primed WT, Casp11–/–, and Casp1–/– BMDMs stimulated with 10 μM nigericin for 1 h. e, Immunoblot of caspase-11 and β-actin in lysates of MS1 endothelial cells transduced with LentiCRISPR V2 plasmid expressing control (GFP) sgRNA or caspase-11 sgRNA. f, Immunoblot of caspase-4 and β-actin in lysates of HeLa cells transduced with LentiCRISPR V2 plasmid expressing control (GFP) sgRNA or caspase-4 sgRNA. g, Immunoblot of GSDMD in the lysates of Pam3CSK4-primed WT BMDMs infected with EHEC or S. flexneri (MOI=50) for 16 h or 10 µM nigericin for 1 h in the presence or absence of 50 mM glycine. h, Galectin-1 release from liposomes packaged with galectin-1 and incubated with either 1 µg recombinant active caspase-11, 2 µg recombinant gasdermin D, both caspase-11 and gasdermin D, or 0.1% Triton X-100. Data are presented as mean ± SEM of one experiment representative of two (b,h; h is the replicate for the Fig. 2i). Combined data from two independent experiments (d) are shown as mean ± SEM. Immunoblots (c,eg) are representative of two independent experiments.

Source data

Extended Data Fig. 2 Galectin-1 contributes to lethality during sepsis.

a, Plasma amounts of ALT and LDH in WT, Lgals1–/–, Casp11–/–, and Gsdmd–/– mice injected i.p. with 5 mg/kg LPS for 18 h. b, Survival of WT (n=9), Lgals1–/– (n=9), and Casp11–/– mice (n=4) injected i.p. with E. coli (5x108 CFU). c, Survival of Lgals1–/– mice injected i.p. with LPS (3 mg/kg) followed by i.p. injection of PBS (n=4), 100 µg of recombinant GFP (n=4), or 100 µg rGal-1 (n=4) 1 h later. d, Survival of Lgals1–/– mice injected i.p. with LPS (3 mg/kg) followed by i.p. injection of 100 µg of rGal-1 (n=6) or heat inactivated rGal-1 (HI rGal-1) (n=6) 1 h later. e, TNF and IL-6 secretion by WT BMDMs stimulated with 5 µM rGal-1 or 1 µg LPS for 16 h. f, Survival of WT and Casp11–/– mice injected i.p. with PBS (n=3) or 100 µg of rGal-1 (n=3). Combined data from two independent experiments are shown (b,d, and e). In (e) data are presented as mean ± SEM. In (a), each circle represents a mouse and the horizontal lines represent mean. ns, not significant (one-way ANOVA). *p < 0.05, one-way ANOVA (a); Mantel-Cox test (d).

Source data

Extended Data Fig. 3 Galectin-1 amplifies systemic inflammatory responses during LPS shock.

a,b, Cytokine and chemokine levels in the lung (a) and spleen (b) homogenates of WT and Lgals1–/– mice injected i.p. with 5 mg/kg LPS for 20 h. Combined data from two independent experiments are shown. Each circle represents a mouse and the horizontal lines represent mean. *p < 0.05; unpaired two-tailed t test (ab); ns, not significant.

Source data

Extended Data Fig. 4 Galectin-1 amplifies systemic inflammatory responses during endotoxemia.

a, Indicated cytokine and chemokine levels in the plasma of WT (n=5), Lgals1–/– (n=5), Casp11–/– (n=5), and Gsdmd–/– (n=5) mice injected i.p. with 5 mg/kg LPS for 20 h. Each circle represents a mouse and the horizontal lines represent mean. *p < 0.05; one-way ANOVA followed by Sidak’s post-test.

Source data

Extended Data Fig. 5 Galectin-1 functions in a glycan-dependent manner during endotoxemia.

a, Survival of WT, Mgat5–/–, and C2gnt1–/– mice injected i.p. with 5 mg/kg LPS followed by i.p. injection of PBS or 100 µg of rGal-1 1 h later (WT LPS PBS n=6, WT LPS rGal-1 n=7, Mgat5–/– LPS PBS n=5, Mgat5–/– LPS rGal-1 n=5, C2gnt1–/– LPS PBS n=9 and C2gnt1–/– LPS rGal-1 n=11). b, Plasma amounts of ALT and LDH in WT (n=6), Mgat5–/– (n=5), and C2gnt1–/– (n=9) mice injected i.p. with 5 mg/kg LPS for 18 h. c, Galectin-1 levels in the plasma of WT (n=6), Mgat5–/– (n=5), and C2gnt1–/– (n=9) mice injected i.p. with 5 mg/kg LPS for 18 h. d, Immunoblot of CD45 that was immunoprecipitated with anti-CD45 antibody or IgG control antibody from the spleen homogenates of WT mice. Each circle represents a mouse and the horizontal lines represent mean (b,c). ns, not significant; one-way ANOVA followed by the Sidak’s post-test. Immunoblots (d) are representative of two independent experiments (d).

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 2

Uncropped immunoblot image.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Uncropped immunoblot image.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Uncropped immunoblot image.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Uncropped immunoblot image.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Uncropped immunoblot image.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Uncropped immunoblot image.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Russo, A.J., Vasudevan, S.O., Méndez-Huergo, S.P. et al. Intracellular immune sensing promotes inflammation via gasdermin D–driven release of a lectin alarmin. Nat Immunol 22, 154–165 (2021). https://doi.org/10.1038/s41590-020-00844-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-00844-7

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