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:

Extracellular SQSTM1 mediates bacterial septic death in mice through insulin receptor signalling

Abstract

Sepsis is the most common cause of death for patients in intensive care worldwide due to a dysregulated host response to infection. Here, we investigate the role of sequestosome-1 (SQSTM1/p62), an autophagy receptor that functions as a regulator of innate immunity, in sepsis. We find that lipopolysaccharide elicits gasdermin D-dependent pyroptosis to enable passive SQSTM1 release from macrophages and monocytes, whereas transmembrane protein 173-dependent TANK-binding kinase 1 activation results in the phosphorylation of SQSTM1 at Ser403 and subsequent SQSTM1 secretion from macrophages and monocytes. Moreover, extracellular SQSTM1 binds to insulin receptor, which in turn activates a nuclear factor kappa B-dependent metabolic pathway, leading to aerobic glycolysis and polarization of macrophages. Intraperitoneal injection of anti-SQSTM1-neutralizing monoclonal antibodies or conditional depletion of Insr in myeloid cells using the Cre–loxP system protects mice from lethal sepsis (caecal ligation and puncture or infection by Escherichia coli or Streptococcus pneumoniae) and endotoxaemia. We also report that circulating SQSTM1 and the messenger RNA expression levels of SQSTM1 and INSR in peripheral blood mononuclear cells are related to the severity of sepsis in 40 patients. Thus, extracellular SQSTM1 has a pathological role in sepsis and could be targeted to develop therapies for sepsis.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Activation of MYD88-dependent TLR4 signalling induces the expression and secretion of SQSTM1.
Fig. 2: TMEM173-dependent TBK1 activation mediates SQSTM1 phosphorylation, expression and secretion.
Fig. 3: Activation of the GSDMD-dependent pyroptosis pathway induces passive release of SQSTM1.
Fig. 4: Exogenous SQSTM1 promotes metabolic programming and macrophage polarization.
Fig. 5: INSR is the major receptor for the immunometabolic activity of SQSTM1.
Fig. 6: The SQSTM1–INSR pathway mediates CLP-induced polymicrobial sepsis.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available within the article and its supplementary information or from the corresponding author upon reasonable request. Source data are provided with this paper.

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Hotchkiss, R. S. et al. Sepsis and septic shock. Nat. Rev. Dis. Primers 2, 16045 (2016).

    PubMed  PubMed Central  Google Scholar 

  3. Deretic, V., Saitoh, T. & Akira, S. Autophagy in infection, inflammation and immunity. Nat. Rev. Immunol. 13, 722–737 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Levine, B., Mizushima, N. & Virgin, H. W. Autophagy in immunity and inflammation. Nature 469, 323–335 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Klionsky, D. J. & Emr, S. D. Autophagy as a regulated pathway of cellular degradation. Science 290, 1717–1721 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Levine, B. & Kroemer, G. Biological functions of autophagy genes: a disease perspective. Cell 176, 11–42 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Xie, Y. et al. Posttranslational modification of autophagy-related proteins in macroautophagy. Autophagy 11, 28–45 (2015).

    PubMed  Google Scholar 

  8. Dikic, I. & Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 19, 349–364 (2018).

    CAS  PubMed  Google Scholar 

  9. Cho, S. J. et al. Plasma ATG5 is increased in Alzheimer’s disease. Sci. Rep. 9, 4741 (2019).

    PubMed  PubMed Central  Google Scholar 

  10. Castellazzi, M. et al. Correlation between auto/mitophagic processes and magnetic resonance imaging activity in multiple sclerosis patients. J. Neuroinflammation 16, 131 (2019).

    PubMed  PubMed Central  Google Scholar 

  11. Naguib, M. & Rashed, L. A. Serum level of the autophagy biomarker Beclin-1 in patients with diabetic kidney disease. Diabetes Res. Clin. Pract. 143, 56–61 (2018).

    CAS  PubMed  Google Scholar 

  12. Wang, X. et al. Defective lysosomal clearance of autophagosomes and its clinical implications in nonalcoholic steatohepatitis. FASEB J. 32, 37–51 (2018).

    CAS  PubMed  Google Scholar 

  13. Kim, J. Y. & Ozato, K. The sequestosome 1/p62 attenuates cytokine gene expression in activated macrophages by inhibiting IFN regulatory factor 8 and TNF receptor-associated factor 6/NF-κB activity. J. Immunol. 182, 2131–2140 (2009).

    CAS  PubMed  Google Scholar 

  14. Ponpuak, M. et al. Delivery of cytosolic components by autophagic adaptor protein p62 endows autophagosomes with unique antimicrobial properties. Immunity 32, 329–341 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Zheng, Y. T. et al. The adaptor protein p62/SQSTM1 targets invading bacteria to the autophagy pathway. J. Immunol. 183, 5909–5916 (2009).

    CAS  PubMed  Google Scholar 

  16. Fujita, K., Maeda, D., Xiao, Q. & Srinivasula, S. M. Nrf2-mediated induction of p62 controls Toll-like receptor-4-driven aggresome-like induced structure formation and autophagic degradation. Proc. Natl Acad. Sci. USA 108, 1427–1432 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lee, H.-M. et al. Autophagy negatively regulates keratinocyte inflammatory responses via scaffolding protein p62/SQSTM1. J. Immunol. 186, 1248–1258 (2011).

    CAS  PubMed  Google Scholar 

  18. Duran, A. et al. The signaling adaptor p62 is an important NF-κB mediator in tumorigenesis. Cancer Cell 13, 343–354 (2008).

    CAS  PubMed  Google Scholar 

  19. Wooten, M. W. et al. The p62 scaffold regulates nerve growth factor-induced NF-κB activation by influencing TRAF6 polyubiquitination. J. Biol. Chem. 280, 35625–35629 (2005).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  21. Kang, R. et al. HMGB1 in health and disease. Mol. Aspects Med. 40, 1–116 (2014).

    CAS  PubMed  Google Scholar 

  22. Hesse, D. G. et al. Cytokine appearance in human endotoxemia and primate bacteremia. Surg. Gynecol. Obstet. 166, 147–153 (1988).

    CAS  PubMed  Google Scholar 

  23. 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 

  24. Kawai, T. et al. Lipopolysaccharide stimulates the MyD88-independent pathway and results in activation of IFN-regulatory factor 3 and the expression of a subset of lipopolysaccharide-inducible genes. J. Immunol. 167, 5887–5894 (2001).

    CAS  PubMed  Google Scholar 

  25. Vanlandingham, P. A. & Ceresa, B. P. Rab7 regulates late endocytic trafficking downstream of multivesicular body biogenesis and cargo sequestration. J. Biol. Chem. 284, 12110–12124 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Dong, X.-P. et al. The type IV mucolipidosis-associated protein TRPML1 is an endolysosomal iron release channel. Nature 455, 992–996 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ubersax, J. A. & Ferrell, J. E. Jr. Mechanisms of specificity in protein phosphorylation. Nat. Rev. Mol. Cell Biol. 8, 530–541 (2007).

    CAS  PubMed  Google Scholar 

  28. Matsumoto, G., Wada, K., Okuno, M., Kurosawa, M. & Nukina, N. Serine 403 phosphorylation of p62/SQSTM1 regulates selective autophagic clearance of ubiquitinated proteins. Mol. Cell 44, 279–289 (2011).

    CAS  PubMed  Google Scholar 

  29. Pilli, M. et al. TBK-1 promotes autophagy-mediated antimicrobial defense by controlling autophagosome maturation. Immunity 37, 223–234 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Perry, A. K., Chow, E. K., Goodnough, J. B., Yeh, W.-C. & Cheng, G. Differential requirement for TANK-binding kinase-1 in type I interferon responses to Toll-like receptor activation and viral infection. J. Exp. Med. 199, 1651–1658 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Heipertz, E. L., Harper, J. & Walker, W. E. STING and TRIF contribute to mouse sepsis, depending on severity of the disease model. Shock 47, 621–631 (2017).

    CAS  PubMed  Google Scholar 

  32. Hu, Q. et al. STING-mediated intestinal barrier dysfunction contributes to lethal sepsis. EBioMedicine 41, 497–508 (2019).

    PubMed  PubMed Central  Google Scholar 

  33. Zeng, L. ALK is a therapeutic target for lethal sepsis. Sci. Transl. Med. 9, eaan5689 (2017).

    PubMed  PubMed Central  Google Scholar 

  34. Zhang, H. et al. TMEM173 drives lethal coagulation in sepsis. Cell Host Microbe 27, 556–570 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Xia, P. et al. Sox2 functions as a sequence-specific DNA sensor in neutrophils to initiate innate immunity against microbial infection. Nat. Immunol. 16, 366–375 (2015).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  37. Orning, P. et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science 362, 1064–1069 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Sarhan, J. et al. Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc. Natl Acad. Sci. USA 115, E10888–E10897 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Freemerman, A. J. et al. Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J. Biol. Chem. 289, 7884–7896 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Lu, Q. & Lemke, G. Homeostatic regulation of the immune system by receptor tyrosine kinases of the Tyro 3 family. Science 293, 306–311 (2001).

    CAS  PubMed  Google Scholar 

  41. Chaudhuri, A. Regulation of macrophage polarization by RON receptor tyrosine kinase signaling. Front. Immunol. 5, 546 (2014).

    PubMed  PubMed Central  Google Scholar 

  42. Liu, C.-P. et al. NF-κB pathways are involved in M1 polarization of RAW 264.7 macrophage by polyporus polysaccharide in the tumor microenvironment. PLoS ONE 12, e0188317 (2017).

    PubMed  PubMed Central  Google Scholar 

  43. Mauro, C. et al. NF-κB controls energy homeostasis and metabolic adaptation by upregulating mitochondrial respiration. Nat. Cell Biol. 13, 1272–1279 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Spec, A. et al. T cells from patients with Candida sepsis display a suppressive immunophenotype. Crit. Care 20, 15 (2016).

    PubMed  PubMed Central  Google Scholar 

  45. Moscat, J., Karin, M. & Diaz-Meco, M. T. p62 in cancer: signaling adaptor beyond autophagy. Cell 167, 606–609 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Sánchez-Martín, P., Saito, T. & Komatsu, M. p62/SQSTM1: ‘jack of all trades’ in health and cancer. FEBS J. 286, 8–23 (2019).

    PubMed  Google Scholar 

  47. Barber, G. N. STING: infection, inflammation and cancer. Nat. Rev. Immunol. 15, 760–770 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Gaidt, M. M. et al. The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3. Cell 171, 1110–1124 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Li, N. et al. STING-IRF3 contributes to lipopolysaccharide-induced cardiac dysfunction, inflammation, apoptosis and pyroptosis by activating NLRP3. Redox Biol. 24, 101215 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Han, S. et al. Macrophage insulin receptor deficiency increases ER stress-induced apoptosis and necrotic core formation in advanced atherosclerotic lesions. Cell Metab. 3, 257–266 (2006).

    CAS  PubMed  Google Scholar 

  51. Mauer, J. et al. Myeloid cell-restricted insulin receptor deficiency protects against obesity-induced inflammation and systemic insulin resistance. PLoS Genet. 6, e1000938 (2010).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Peng, T. et al. Disruption of phospholipase Cγ1 signalling attenuates cardiac tumor necrosis factor-α expression and improves myocardial function during endotoxemia. Cardiovasc. Res. 78, 90–97 (2008).

    CAS  PubMed  Google Scholar 

  54. Weischenfeldt, J. & Porse, B. Bone marrow-derived macrophages (BMM): isolation and applications. Cold Spring Harb. Protoc. https://doi.org/10.1101/pdb.prot5080 (2008).

  55. Kang, R. et al. A novel PINK1- and PARK2-dependent protective neuroimmune pathway in lethal sepsis. Autophagy 12, 2374–2385 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Deng, W. et al. The circadian clock controls immune checkpoint pathway in sepsis. Cell Rep. 24, 366–378 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Yang, L. et al. PKM2 regulates the Warburg effect and promotes HMGB1 release in sepsis. Nat. Commun. 5, 4436 (2014).

    CAS  PubMed  Google Scholar 

  58. Chen, R. et al. cAMP metabolism controls caspase-11 inflammasome activation and pyroptosis in sepsis. Sci. Adv. 5, eaav5562 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. 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 

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

    CAS  PubMed  Google Scholar 

  61. Taylor, F. B. Jr. et al. Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb. Haemost. 86, 1327–1330 (2001).

    CAS  PubMed  Google Scholar 

  62. Zhu, S. et al. HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Res. 77, 2064–2077 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Song, X. et al. AMPK-mediated BECN1 phosphorylation promotes ferroptosis by directly blocking system Xc activity. Curr. Biol. 28, 2388–2399 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Tang, D. et al. Endogenous HMGB1 regulates autophagy. J. Cell Biol. 190, 881–892 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Tang, D. et al. High-mobility group box 1 is essential for mitochondrial quality control. Cell Metab. 13, 701–711 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Tan, J. M. J. et al. An ATG16L1-dependent pathway promotes plasma membrane repair and limits Listeria monocytogenes cell-to-cell spread. Nat. Microbiol. 3, 1472–1485 (2018).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Primm for his critical reading of the manuscript. G.K. is supported by the Ligue Contre le Cancer (équipe labellisée), Agence National de la Recherche (ANR)—Projets blancs, ANR under the frame of E-Rare-2, ERA-Net for Research Programmes on Rare Diseases, Association pour la Recherche sur le Cancer, Cancéropôle Ile-de-France, Chancelerie des Universités de Paris (Legs Poix), Fondation pour la Recherche Médicale, a donation by Elior, the European Research Area Network on Cardiovascular Diseases (MINOTAUR), Gustave Roussy Odyssea, the European Union Horizon 2020 Project Oncobiome, Fondation Carrefour, High-end Foreign Expert Program in China (nos. GDW20171100085 and GDW20181100051), Institut National du Cancer, Inserm (HTE), Institut Universitaire de France, LeDucq Foundation, LabEx Immuno-Oncology, RHU Torino Lumière, the Seerave Foundation, the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination and the SIRIC Cancer Research and Personalized Medicine. J.L. is supported by grants from the National Natural Science Foundation of China (nos. 31671435, 81400132 and 81772508). J.J. is supported by a grant from the National Natural Science Foundation of China (no. 81530063). L.Z. is supported by an Excellent Youth Grant of the State Key Laboratory of Trauma, Burns and Combined Injury of China (no. SKLYQ201901).

Author information

Authors and Affiliations

Authors

Contributions

B.Z., D.T., R.K. and J.J. conceived and planned the experiments. B.Z., J.L., L.Z., S.Z., D.T. and R.K. carried out the simulations, sample preparation and analysed the data. T.R.B. provided the mouse strains. H.W., T.R.B., G.K., D.J.K. and H.J.Z. edited the manuscript and contributed to the interpretation of the results. D.T. and R.K. wrote the manuscript. All authors provided critical feedback and helped shape the research, analysis and manuscript.

Corresponding authors

Correspondence to Jianxin Jiang, Daolin Tang or Rui Kang.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 LPS induces SQSTM1 secretion in monocytes and macrophages.

a, ELISA analysis of SQSTM1 release in THP1 (a human monocytic cell line), primary human blood monocyte-derived macrophages (HPBMs), primary mouse peritoneal macrophages (MPMs), primary mouse lung macrophages (MLMs), and primary mouse hepatic macrophages (MHMs) following treatment with LPS (200 ng/ml) for 3-48 h (n = 5 well /group; two-tailed t test, versus untreated group). b, LDH analysis of cytotoxicity in the indicated cells following treatment with LPS (200 ng/ml) for 3-48 h (n = 5 well /group). c, The indicated BMDMs were treated with LPS (200 ng/ml) for 24 h and the level of Sqstm1 mRNA was assayed by qPCR (n = 3 well /group; two-tailed t test, versus control LPS group). Data in (ac) are presented as mean ± SD. Data in (ab) are from two independent experiments. Data in (c) are from three independent experiments.

Extended Data Fig. 2 The activation of the GSDMD-dependent pyroptosis pathway induces SQSTM1 release.

a, Analysis of cytotoxicity, HMGB1 release, and SQSTM1 release in the indicated poly (I:C)-primed BMDMs following LPS electroporation (LPSe) or E. coli (MOI = 25) infection for 24 h (n = 5 well/group; two-tailed t test, versus wild type group). b, Analysis of cytotoxicity, HMGB1 release, and SQSTM1 release in the indicated LPS-primed BMDMs following ATP (5 mM, 3 h) or nigericin (10 µM, 3 h) treatment (n = 5 well/group; two-tailed t test, versus wild type group). c, Analysis of cytotoxicity, HMGB1 release, and SQSTM1 release in the indicated BMDMs following Y. pestis (MOI = 25) infection or LPS5Z7 (LPS [50 ng/ml] + 5z7 [400 nM]) (n = 5 well/group; two-tailed t test, versus wild type group). Data in (ac) are presented as mean ± SD from two independent experiments.

Extended Data Fig. 3 Exogenous SQSTM1 promotes glucose uptake and lactate production.

a,b, Indicated primary macrophages were stimulated with rSQSTM1 (10, 100, and 1000 ng/ml) or boiled rSQSTM1 (BrSQSTM1) for 24 h. The glucose uptake (a) and lactate production (b) were assayed (n = 3 well/group; one-way ANOVA test, versus control group). c, TMs and HPBMs were treated with rSQSTM1 (100 ng/ml) and IL4 (50 ng/ml) for 48 h and the mRNA expression of Il10 and Arg1 were assayed (n = 3 well/group; two-tailed t test, versus control group). HPBMs, primary human blood monocyte-derived macrophages; MPMs, primary mouse peritoneal macrophages; MLMs, primary mouse lung macrophages; MHMs, primary mouse hepatic macrophages. Data in (ac) are presented as mean ± SD from two independent experiments.

Extended Data Fig. 4 Effects of TLRs on rSQSTM1-induced NFKB activity.

The indicated TLR-knockdown THP1 cells were stimulated with (a) pam3CSK4 (1 ng/ml), (b) HKLM (107 cells/ml), (c) poly(I:C) (10 µg/ml), (d) LPS (200 ng/ml), (e) FLA-ST (100 ng/ml), (f) FSL1 (0.1 ng/ml), (g) imiquimod (5 µg/ml), (h) ssRNA40 (5 µg/ml), (i) ODN2006 (10 µg/ml), or (ai) rSQSTM1 (100 ng/ml) for 24 h and the levels of NFKB-induced Lucia luciferase were assessed (n = 3 well/group; two-tailed t test, versus control group). Data in (ai) are presented as mean ± SD from two independent experiments.

Extended Data Fig. 5 PLCG1 and PTK2B mediate the activity of SQSTM1.

a, Heatmap of kinase phosphorylation in the indicated TMs after rSQSTM1 (100 ng/ml) stimulation for 24 h. b, Western blot analysis of protein expression in the indicated TMs following treatment with rSQSTM1 (100 ng/ml) for 24 h. c, Indicated TMs (control, SLC2A1KD, or SLC2A4KD) were stimulated with insulin (100 nM) for 24 h. The glucose uptake was assayed (n = 3 well/group; two-tailed t test, versus control group). AU, arbitrary units. d,e, Analysis of protein expression and NFKB activity in TMs following treatment with insulin (100 nM) for 24 h (n = 3 well/group). f, Analysis of interaction between PTK2B and INSR in membrane protein extraction from TMs following treatment with insulin (100 nM) or rSQSTM1 (100 ng/ml) for 24 h. g, Analysis of interaction between PLCG1 and RELA in whole protein extraction from indicated TMs following treatment with rSQSTM1 (100 ng/ml) for 24 h. h, Indicated cells were treated with rSQSTM1 (100 ng/ml) for 24 h, and the levels of lactate and IL6 mRNA were assayed (n = 3 well/group). Data in (c), (e), and (h) are presented as mean ± SD. Data in (a) is from one independent experiment. Data in (b), (d), (f), and (g) are from two independent experiments. Data in (c), (e), and (h) are from three independent experiments.

Extended Data Fig. 6 The SQSTM1-INSR pathway mediates CLP-induced polymicrobial sepsis.

af, The serum level of IL6 (a), IL1B (b), TNF (c), HMGB1 (d), LDH (e), and lactate (f) were assayed in indicated CLP-induced mice with or without anti-SQSTM1 monoclonal antibodies (20 mg/kg) treatment or depletion of INSR in myeloid cells (n = 5 mice/group; two-tailed t test, versus control group). g, Administration of anti-SQSTM1 monoclonal antibodies (20 mg/kg) and/or depletion of INSR in myeloid cells in mice prevented CLP-induced animal death (n = 10 mice/group; Log-rank test). hj, Survival of the indicated mice after E. coli (h) or S. pneumoniae (i) infection or (j) CLP-induced sepsis (n = 10 mice/group; Log-rank test). Data in (af) are presented as mean ± SD from three independent experiments. Data in (gj) are from two independent experiments.

Extended Data Fig. 7 The inhibition of the SQSTM1-INSR pathway protects mice against LPS-induced endotoxemia.

a, Administration of anti-SQSTM1 monoclonal antibodies (20 mg/kg) or depletion of INSR in myeloid cells in mice prevented LPS (10 mg/kg)-induced animal death (n = 10 mice/group; Log-rank test). bk, In parallel, the serum levels of CK (b), AMY (c), BUN (d), GPT/ALT I (e), IL6 (f), IL1B (g), TNF (h), HMGB1 (i), LDH (j), and lactate (k) were assayed (n = 5 mice/group; two-tailed t test, versus control group). Data in (bk) are presented as mean ± SD. Data in (ak) are from two independent experiments.

Extended Data Fig. 8 The inhibition of the SQSTM1-INSR pathway protects mice against bacterial sepsis.

ac, Survival of the indicated mice in E. coli (a) or S. pneumoniae (b) infection or (c) clinically relevant moderate CLP-induced sepsis (n = 10 mice/group; Log-rank test). d,e, In parallel, the levels of apoptosis (d) and Pdcd1 mRNA (e) were assayed at 72 h (n = 5 mice/group; two-tailed t test, versus control group). Data in (d, e) are presented as mean ± SD. Data in (ae) are from two independent experiments.

Extended Data Fig. 9 The SQSTM1-INSR pathway promotes systemic coagulation in bacterial sepsis.

The levels of blood markers of DIC (ae) were assayed at 72 h in the indicated mice in E. coli or S. pneumoniae infection or clinically relevant moderate CLP-induced sepsis (n = 5 mice/group; two-tailed t test, versus control group). Data in (ae) are presented as mean ± SD from two independent experiments.

Extended Data Fig. 10 Association of the SQSTM1-INSR axis with the severity of sepsis in patients.

af, The correlation assay between SQSTM1, INSR, SOFA, and DIC score in patients with sepsis and septic shock (n=40 cases). g, Pearson correlation heatmap of immune mediators.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 2

Numerical data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, B., Liu, J., Zeng, L. et al. Extracellular SQSTM1 mediates bacterial septic death in mice through insulin receptor signalling. Nat Microbiol 5, 1576–1587 (2020). https://doi.org/10.1038/s41564-020-00795-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-00795-7

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